Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
1.
Br J Cancer ; 130(3): 476-482, 2024 02.
Article in English | MEDLINE | ID: mdl-38135713

ABSTRACT

BACKGROUND: Our preclinical work revealed tumour hypoxia induces homologous recombination deficiency (HRD), increasing sensitivity to Poly (ADP-ribose) polymerase inhibitors. We aimed to induce tumour hypoxia with ramucirumab thereby sensitising tumours to olaparib. PATIENTS AND METHODS: This multi-institution single-arm Phase 1/2 trial enrolled patients with metastatic gastroesophageal adenocarcinoma refractory to ≥1 systemic treatment. In dose escalation, olaparib was evaluated at escalating dose levels with ramucirumab 8 mg/kg day 1 in 14-day cycles. The primary endpoint of Phase 1 was the recommended Phase 2 dose (RP2D), and in Phase 2 the primary endpoint was the overall response rate (ORR). RESULTS: Fifty-one patients received ramucirumab and olaparib. The RP2D was olaparib 300 mg twice daily with ramucirumab 8 mg/kg. In evaluable patients at the RP2D the ORR was 6/43 (14%) (95% CI 4.7-25.6). The median progression-free survival (PFS) was 2.8 months (95% CI 2.3-4.2) and median overall survival (OS) was 7.3 months (95% CI 5.7-13.0). Non-statistically significant improvements in PFS and OS were observed for patients with tumours with mutations in HRD genes. CONCLUSIONS: Olaparib and ramucirumab is well-tolerated with efficacy that exceeds historical controls with ramucirumab single agent for gastric cancer in a heavily pre-treated patient population.


Subject(s)
Adenocarcinoma , Esophageal Neoplasms , Piperazines , Stomach Neoplasms , Humans , Ramucirumab , Stomach Neoplasms/drug therapy , Stomach Neoplasms/genetics , Phthalazines , Adenocarcinoma/drug therapy , Adenocarcinoma/genetics , Esophagogastric Junction , Antineoplastic Combined Chemotherapy Protocols/adverse effects
2.
J Med Case Rep ; 13(1): 389, 2019 Dec 25.
Article in English | MEDLINE | ID: mdl-31874650

ABSTRACT

BACKGROUND: Atrial myxomas are generally considered benign neoplasms. The majority of tumors are sporadic and less than 10% are associated with an autosomal dominant condition known as the Carney complex, which is most often caused by germline mutation in the gene PRKAR1A. Whether this gene plays a role in the development of sporadic myxomas has been an area of debate, although recent studies have suggested that some fraction of sporadic tumors also carry mutations in PRKARIA. Extra-cardiac complications of atrial myxoma include dissemination of tumor to the brain; however, the dissemination of viable invasive tumor cells is exceedingly rare. CASE PRESENTATION: We present here a 48-year-old white woman who developed multiple intracranial hemorrhagic lesions secondary to tumor embolism that progressed to 'false' aneurysm formation and invasion through the vascular wall into brain parenchyma 7 months after resection of an atrial myxoma. Whole exome sequencing of her tumor revealed multiple mutations in PRKAR1A not found in her germline deoxyribonucleic acid (DNA), suggesting that the myxoma in this patient was sporadic. CONCLUSIONS: Our patient illustrates that mutations in PRKAR1A may be found in sporadic lesions. Whether the presence of this mutation affects the clinical behavior of sporadic tumors and increases risk for metastasis is not clear. Regardless, the protein kinase A pathway which is regulated by PRKAR1A represents a possible target for treatment in patients with metastatic cardiac myxomas harboring mutations in the PRKARIA gene.


Subject(s)
Brain Neoplasms/secondary , Carney Complex/diagnosis , Cyclic AMP-Dependent Protein Kinase RIalpha Subunit/genetics , Dopamine Agents/therapeutic use , Heart Neoplasms/diagnosis , Memantine/therapeutic use , Myxoma/diagnosis , Brain Neoplasms/physiopathology , Brain Neoplasms/therapy , Carney Complex/genetics , Chemoradiotherapy , Female , Gene Expression Regulation, Neoplastic , Genes, Tumor Suppressor , Germ-Line Mutation , Heart Neoplasms/physiopathology , Heart Neoplasms/therapy , Humans , Intracranial Hemorrhages , Middle Aged , Myxoma/physiopathology , Myxoma/therapy , Treatment Outcome , Exome Sequencing
3.
Behav Brain Res ; 333: 74-82, 2017 08 30.
Article in English | MEDLINE | ID: mdl-28666838

ABSTRACT

Matrix Metalloproteinase2, (MMP2, gelatinase A) is a zinc-containing enzyme with a broad substrate specificity including components of the extracellular matrix, cell surface molecules and a wide range bioactive molecules. MMP2 is known to play important roles in a variety of signaling pathways and processes in a wide range of cell types and tissues. In this report we elucidate the effects of the absence of MMP2 in Neural Precursor Cells (NPC) derived from C57BL/6 MMP2 KO mice and in primary and secondary neurosphere formation. We observed smaller neurosphere numbers and sizes, decreased NPC numbers, PCNA expression, DNA and Akt activation in MMP2 KO NPC compared to WT NPC. We also found decreased neurosphere formation and NPC migration outward from adherent neurospheres, decreased CXCR4 and nestin expression and increased GFAP and neuro-filament expression in MMP2 KO NPC compared to Wt NPC. MMP2 KO mice were found to exhibit increased anxiety manifested in open field activity assays compared to Wt mice. MMP2 KO mice also exhibited differences in motor activities manifested by decreased balance and endurance during Rota-rod testing. These studies illustrate an important role of MMP2 in cognitive and motor behaviors and confirm its importance in NPC activities crucial to brain development, growth and response to and recovery from injury.


Subject(s)
Cognition/physiology , Matrix Metalloproteinase 2/deficiency , Motor Activity/genetics , Neural Stem Cells/physiology , Animals , Animals, Newborn , Cell Movement/genetics , Cell Proliferation/genetics , Cells, Cultured , Exploratory Behavior/physiology , Gene Expression Regulation/genetics , Matrix Metalloproteinase 2/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Motor Activity/physiology , Nerve Tissue Proteins/metabolism , Neurogenesis/genetics , Oncogene Protein v-akt/metabolism , Proliferating Cell Nuclear Antigen/metabolism , Receptors, CXCR4/metabolism , Spatial Learning/physiology
4.
J Mol Diagn ; 19(2): 313-327, 2017 03.
Article in English | MEDLINE | ID: mdl-28188106

ABSTRACT

The National Cancer Institute-Molecular Analysis for Therapy Choice (NCI-MATCH) trial is a national signal-finding precision medicine study that relies on genomic assays to screen and enroll patients with relapsed or refractory cancer after standard treatments. We report the analytical validation processes for the next-generation sequencing (NGS) assay that was tailored for regulatory compliant use in the trial. The Oncomine Cancer Panel assay and the Personal Genome Machine were used in four networked laboratories accredited for the Clinical Laboratory Improvement Amendments. Using formalin-fixed paraffin-embedded clinical specimens and cell lines, we found that the assay achieved overall sensitivity of 96.98% for 265 known mutations and 99.99% specificity. High reproducibility in detecting all reportable variants was observed, with a 99.99% mean interoperator pairwise concordance across the four laboratories. The limit of detection for each variant type was 2.8% for single-nucleotide variants, 10.5% for insertion/deletions, 6.8% for large insertion/deletions (gap ≥4 bp), and four copies for gene amplification. The assay system from biopsy collection through reporting was tested and found to be fully fit for purpose. Our results indicate that the NCI-MATCH NGS assay met the criteria for the intended clinical use and that high reproducibility of a complex NGS assay is achievable across multiple clinical laboratories. Our validation approaches can serve as a template for development and validation of other NGS assays for precision medicine.


Subject(s)
High-Throughput Nucleotide Sequencing , Neoplasms/diagnosis , Neoplasms/genetics , Clinical Trials as Topic , Computational Biology/methods , Genetic Variation , Genomics/methods , Genomics/standards , High-Throughput Nucleotide Sequencing/methods , High-Throughput Nucleotide Sequencing/standards , Humans , Quality Assurance, Health Care , Quality Control , Reproducibility of Results , Sensitivity and Specificity , Workflow
5.
PLoS One ; 8(10): e76265, 2013.
Article in English | MEDLINE | ID: mdl-24146847

ABSTRACT

Premature infants often experience chronic hypoxia, resulting in cognitive & motor neurodevelopmental handicaps. These sometimes devastating handicaps are thought to be caused by compromised neural precursor cell (NPC) repair/recovery resulting in variable central nervous system (CNS) repair/recovery. We have identified differential responses of two mouse strains (C57BL/6 & CD1) to chronic hypoxia that span the range of responsiveness noted in the premature human population. We previously correlated several CNS tissue and cellular behaviors with the different behavioral parameters manifested by these two strains. In this report, we use unbiased array technology to interrogate the transcriptome of the subventricular zone (SVZ) in these strains. Our results illustrate differences in mRNA expression in the SVZ of both C57BL/6 and CD1 mice following hypoxia as well as differences between C57BL/6 and CD1 SVZ under both normoxic and hypoxic conditions. Differences in expression were found in gene sets associated with Sox10-mediated neural functions that explain, in part, the differential cognitive and motor responsiveness to hypoxic insult. This may shed additional light on our understanding of the variable responses noted in the human premature infant population and facilitate early intervention approaches. Further interrogation of the differentially expressed gene sets will provide a more complete understanding of the differential responses to, and recovery from, hypoxic insult allowing for more informed modeling of the ranges of disease severity observed in the very premature human population.


Subject(s)
Cerebral Ventricles/blood supply , Cerebral Ventricles/metabolism , Gene Expression Profiling , Hypoxia/genetics , Hypoxia/pathology , Models, Neurological , Animals , Cerebral Ventricles/pathology , Databases, Genetic , Down-Regulation/genetics , Gene Ontology , Humans , Mice , Mice, Inbred C57BL , Oligonucleotide Array Sequence Analysis , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reproducibility of Results , SOXE Transcription Factors/metabolism , Up-Regulation/genetics
6.
Angiogenesis ; 16(3): 689-705, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23504212

ABSTRACT

Vascular integrity is a critical parameter in normal growth and development. Loss of appropriate vascular barrier function is present in various immune- and injury-mediated pathological conditions. CD44 is an adhesion molecule expressed by multiple cell types, including endothelial cells (EC). The goal of the present study was to examine how loss of CD44 affected vascular permeability. Using C57BL/6 WT and CD44-KO mice, we found no significant permeability to Evan's Blue in either strain at baseline. However, there was significantly increased histamine-induced permeability in CD44-deficient mice compared to WT counterparts. Similar results were observed in vitro, where CD44-deficient endothelial monolayers were also impermeable to 40kD-FITC dextran in the absence of vasoactive challenge, but exhibited enhanced and prolonged permeability following histamine. However, CD44-KO monolayers have reduced baseline barrier strength by electrical resistance, which correlated with increased permeability, at baseline, to smaller molecular weight 4-kD FITC-dextran, suggesting weakly formed endothelial junctions. The CD44-KO EC displayed several characteristics consistent with impaired barrier function/dysfunctional EC junctions, including differential expression, phosphorylation, and localization of endothelial junction proteins, increased matrix metalloprotease expression, and altered cellular morphology. Reduced platelet endothelial cell adhesion molecule-1 (PECAM-1) expression by CD44-KO EC in vivo and in vitro was also observed. Reconstitution of murine CD44 or PECAM-1 restored these defects to near WT status, suggesting CD44 regulates vascular permeability and integrity through a PECAM-1 dependent mechanism.


Subject(s)
Capillary Permeability/physiology , Endothelium, Vascular/physiology , Hyaluronan Receptors/metabolism , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Animals , Blotting, Western , Capillary Permeability/genetics , Dextrans , Endothelium, Vascular/metabolism , Evans Blue , Fluorescein-5-isothiocyanate/analogs & derivatives , Hyaluronan Receptors/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Time Factors
7.
Angiogenesis ; 14(2): 173-85, 2011 May.
Article in English | MEDLINE | ID: mdl-21253820

ABSTRACT

The neurogenic areas of the brain are highly organized structures in which there is dynamic reciprocal modulation of neural stem cells (NSC) and microvascular endothelial cells (BEC) resulting in control of neural stem cell and vascular proliferation, survival and differentiation throughout the life of the individual. Select molecules such as GSK-3ß, functioning as signaling nodes, and their downstream signaling components including HIF-1α, HIF-2α and ß-catenin participate in regulating and orchestrating the diverse responses involved in this complex process. In this report we demonstrate GSK-3ß's role as a signaling node in two mouse strains (C57BL/6, which have been found to respond to and recover from a hypoxic insult from P3 to P11 poorly and CD-1, which have been found to respond to and recover from a hypoxic insult from P3 to P11 well both in vivo and in vitro) which mimic the wide range of responsiveness to hypoxic insult observed in the very low birth weight premature infant population. Differences in levels of neural stem cell and microvascular endothelial cell GSK-3ß activation, ß-catenin serine phosphorylation, HIF-1α and 2α, BDNF, SDF-1 and VEGF, ß-III-tubulin and cleaved notch-1 expression in C57BL/6 and CD-1 subventricular zone tissues, and cultured NSC and BEC were noted. Specifically, CD1 pups, SVZ tissues and isolated NSC and BEC exhibit less GSK-3ß and ß-catenin serine phoslphorylation and greater HIF-1α and 2α, BDNF, SDF-1 and VEGF, ß-III-tubulin and cleaved notch-1 expression compared to C57BL/6. Correlating with these changes were differences of several neural stem cell and microvascular endothelial cell behaviors including proliferation, apoptosis, migration and differentiation with CD1 NSC exhibiting greater proliferation and migration and decreased apoptosis and differentiation and CD1 BEC exhibiting greater angiogenesis. Further, upon treatment with nanomolar concentrations of a GSK-3ß inhibitor (SB412682), C57 NSC and BEC behaviors could be brought to CD1 levels, consistent with the concept of GSK-3ß functioning as a multifunctional signaling pathway node, modulating several behaviors in these cells. Lastly, the therapeutic potential of targeting GSK-3ß is discussed.


Subject(s)
Cell Communication , Endothelial Cells/cytology , Endothelial Cells/enzymology , Glycogen Synthase Kinase 3/metabolism , Neural Stem Cells/cytology , Neural Stem Cells/enzymology , Signal Transduction , Aminophenols/pharmacology , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Brain/cytology , Cell Communication/drug effects , Cell Differentiation/drug effects , Cell Movement/drug effects , Cell Proliferation/drug effects , Endothelial Cells/drug effects , Enzyme Activation/drug effects , Glycogen Synthase Kinase 3/antagonists & inhibitors , Glycogen Synthase Kinase 3 beta , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Male , Maleimides/pharmacology , Mice , Mice, Inbred C57BL , Neovascularization, Physiologic/drug effects , Neural Stem Cells/drug effects , Neurogenesis/drug effects , Phosphorylation/drug effects , Phosphoserine/metabolism , Receptor Cross-Talk/drug effects , Signal Transduction/drug effects , Solubility/drug effects , Species Specificity , beta Catenin/metabolism
8.
Am J Pathol ; 169(1): 314-24, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16816383

ABSTRACT

Platelet endothelial cell adhesion molecule-1 (PECAM-1/CD31) regulates a variety of endothelial and immune cell biological responses. PECAM-1-null mice exhibit prolonged and increased permeability after inflammatory insults. We observed that in PECAM-1-null endothelial cells (ECs), beta-catenin remained tyrosine phosphorylated, coinciding with a sustained increase in permeability. Src homology 2 domain containing phosphatase 2 (SHP-2) association with beta-catenin was diminished in PECAM-1-null ECs, suggesting that lack of PECAM-1 inhibits the ability of this adherens junction component to become dephosphorylated, promoting a sustained increase in permeability. beta-Catenin/Glycogen synthase kinase 3 (GSK-3beta) association and beta-catenin serine phosphorylation levels were increased and beta-catenin expression levels were reduced in PECAM-1-null ECs. Glycogen synthase kinase 3 (GSK-3beta) serine phosphorylation (inactivation) was blunted in PECAM-1-null ECs after histamine treatment or shear stress. Our data suggest that PECAM-1 serves as a critical dynamic regulator of endothelial barrier permeability. On stimulation by a vasoactive substance or shear stress, PECAM-1 became tyrosine phosphorylated, enabling recruitment of SHP-2 and tyrosine-phosphorylated beta-catenin to its cytoplasmic domain, facilitating dephosphorylation of beta-catenin, and allowing reconstitution of adherens junctions. In addition, PECAM-1 modulated the levels of beta-catenin by regulating the activity of GSK-3beta, which in turn affected the serine phosphorylation of beta-catenin and its proteosomal degradation, affecting the ability of the cell to reform adherens junctions in a timely fashion.


Subject(s)
Capillary Permeability/physiology , Endothelial Cells/metabolism , Glycogen Synthase Kinase 3/metabolism , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Signal Transduction/physiology , beta Catenin/metabolism , Animals , Blotting, Western , Cells, Cultured , Fluorescent Antibody Technique , Glycogen Synthase Kinase 3 beta , Histamine/pharmacology , Histamine Agents/pharmacology , Humans , Mice , Models, Biological , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Platelet Endothelial Cell Adhesion Molecule-1/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Histamine/drug effects , Receptors, Histamine/metabolism
9.
Biochem Biophys Res Commun ; 329(4): 1225-33, 2005 Apr 22.
Article in English | MEDLINE | ID: mdl-15766557

ABSTRACT

Platelet endothelial cell adhesion molecule-1 (PECAM-1) binds tyrosine-phosphorylated beta-catenin and modulates beta-catenin localization and sequestration. The biological significance of this interaction, while still unclear, it has been postulated to be involved in modulating adherens junction dynamics in response to perturbants [J. Clin. Invest. 109 (2002) 383]. Here we demonstrate that tyrosine-phosphorylated beta-catenin, and to a lesser extent unphosphorylated beta-catenin, interact with a portion of the cytoplasmic domain of PECAM-1 encoded by exon 15. Using RT-PCR, we obtained products representing alternatively spliced PECAM-1 isoforms from mouse kidney total mRNA and generated PECAM-1-GST constructs expressing full length and naturally occurring alternatively spliced PECAM-1 variants. Co-precipitation assays revealed that the protein sequence encoded by exon 15 is necessary for beta-catenin binding. Transfections using deletion mutants confirmed the importance of the exon 15 sequence in this interaction. In contrast, gamma-catenin-PECAM-1 interactions are thought to be modulated by an as yet undefined PECAM-1 serine phosphorylation and appear to mediate dynamic PECAM-1 intermediate filament cytoskeletal interactions [J. Biol. Chem. 275 (2000) 21435]. Here we demonstrate that the PECAM-1-gamma-catenin interaction occurs via an exon 13-mediated process. GST-pull-down assays illustrated the importance of the exon 13 sequence in this interaction. Further, using site-directed mutagenesis of S(673) to C and D and S(669 and 670) to C, we confirmed the importance of S(673) and its phosphorylation state as a mediator of gamma-catenin-PECAM-1 binding. Our studies define the exons of the PECAM-1 cytoplasmic domain that is involved in mediating these PECAM-1-catenin family member interactions and will allow investigators to better define the biological functions resulting from these interactions.


Subject(s)
Cytoskeletal Proteins/metabolism , Platelet Endothelial Cell Adhesion Molecule-1/chemistry , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Trans-Activators/metabolism , Cell Line , Cytoskeletal Proteins/genetics , Desmoplakins , Exons/genetics , Humans , Phosphoserine/metabolism , Platelet Endothelial Cell Adhesion Molecule-1/genetics , Protein Binding , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Deletion/genetics , Trans-Activators/genetics , beta Catenin , gamma Catenin
10.
Am J Pathol ; 166(1): 185-96, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15632011

ABSTRACT

Platelet endothelial cell adhesion molecule-1 (PECAM-1, CD31), an adhesion molecule expressed on hematopoietic and endothelial cells, mediates apoptosis, cell proliferation, and migration and maintains endothelial integrity in addition to its roles as a modulator of lymphocyte and platelet signaling and facilitator of neutrophil transmigration. Recent data suggest that CD31 functions as a scaffolding protein to regulate phosphorylation of the signal transducers and activators of transcription (STAT) family of signaling molecules, particularly STAT3 and STAT5. STAT3 regulates the acute phase response to innate immune stimuli such as lipopolysaccharide (LPS) and promotes recovery from LPS-induced septic shock. Here we demonstrate that CD31-deficient mice have reduced survival during endotoxic LPS-induced shock. As compared to wild-type controls, CD31-deficient mice showed enhanced vascular permeability; increased apoptotic cell death in liver, kidney, and spleen; and elevated levels of serum tumor necrosis factor alpha (TNF-alpha), interferon gamma (IFNgamma), MCP-1, MCP-5, sTNRF, and IL-6. In response to LPS in vivo and in vitro, splenocytes and endothelial cells from knockout mice had reduced levels of phosphorylated STAT3. These results suggest that CD31 is necessary for maintenance of endothelial integrity and prevention of apoptosis during septic shock and for STAT3-mediated acute phase responses that promote survival during septic shock.


Subject(s)
DNA-Binding Proteins/physiology , Platelet Endothelial Cell Adhesion Molecule-1/genetics , Platelet Endothelial Cell Adhesion Molecule-1/immunology , Shock, Septic/pathology , Trans-Activators/physiology , Animals , Cells, Cultured , DNA-Binding Proteins/genetics , Disease Susceptibility , Endothelium, Vascular/pathology , Female , Flow Cytometry , Gene Expression Regulation , Lipopolysaccharides/toxicity , Mice , Mice, Inbred C57BL , Mice, Knockout , Pulmonary Circulation , STAT3 Transcription Factor , Spleen/drug effects , Spleen/pathology , Trans-Activators/genetics , Tumor Necrosis Factor-alpha/genetics , Vanadates/pharmacology
11.
FASEB J ; 17(11): 1458-69, 2003 Aug.
Article in English | MEDLINE | ID: mdl-12890700

ABSTRACT

Platelet endothelial cell adhesion molecule-1 (PECAM-1), an immunoglobulin family vascular adhesion molecule, is involved in endothelial cell migration and angiogenesis (1, 2). We found that endothelial cells lacking PECAM-1 exhibit increased single cell motility and extension formation but poor wound healing migration, reminiscent of cells in which Rho activity has been suppressed by overexpressing a GTPase-activating protein (3). The ability of PECAM-1 to restore wound healing migration to PECAM-1-deficient cells was independent of its extracellular domain or signaling via its immunoreceptor tyrosine-based inhibitory motif. PECAM-1-deficient endothelial cells had a selective defect in RhoGTP loading, and inhibition of Rho activity mimicked the PECAM-1-deficient phenotype of increased chemokinetic single cell motility at the expense of coordinated wound healing migration. The wound healing advantage of PECAM-1-positive endothelial cells was not only Rho mediated but pertussis toxin inhibitable, characteristic of migration mediated by heterotrimeric G-protein-linked seven-transmembrane receptor signaling such as signaling in response to the serum sphingolipid sphingosine-1-phosphate (S1P) (4, 5). Indeed, we found that the wound healing defect of PECAM-1 null endothelial cells is minimized in sphingolipid-depleted media; moreover, PECAM-1 null endothelial cells fail to increase their migration in response to S1P. We have also found that PECAM-1 localizes to rafts and that in its absence heterotrimeric G-protein components are differentially recruited to rafts, providing a potential mechanism for PECAM-1-mediated coordination of S1P signaling. PECAM-1 may thus support the effective S1P/RhoGTP signaling required for wound healing endothelial migration by allowing for the spatially directed, coordinated activation of Galpha signaling pathways.


Subject(s)
Chemotaxis , Endothelium/physiology , Lysophospholipids , Platelet Endothelial Cell Adhesion Molecule-1/physiology , Sphingosine/analogs & derivatives , Animals , Endothelium/cytology , Gene Deletion , Membrane Microdomains/chemistry , Mice , Platelet Endothelial Cell Adhesion Molecule-1/analysis , Platelet Endothelial Cell Adhesion Molecule-1/genetics , Signal Transduction , Sphingosine/pharmacology , Tumor Cells, Cultured , Wound Healing , rho GTP-Binding Proteins/metabolism
12.
Biochem Biophys Res Commun ; 303(1): 212-8, 2003 Mar 28.
Article in English | MEDLINE | ID: mdl-12646189

ABSTRACT

Platelet endothelial cell adhesion molecule-1 (PECAM-1) binds tyrosine phosphorylated beta-catenin and modulates beta-catenin localization [J. Immunol. 158 (7) (1997) 3408; J. Cell Sci. 112 (Pt 18) (1999) 3005]. To elucidate functional consequences of this interaction, we studied endothelial cells from PECAM-1 knockout animals and compared them to PECAM-1 expressing endothelial cells [Mol. Biol. Cell 11 (9) (2000) 3109]. We noted an increase in the expression of beta-catenin protein in PECAM-1 expressing endothelial cells. Further, by immunofluorescence, beta-catenin localized to the cell membrane as well as to the nucleus in PECAM-1 positive endothelial cells, whereas cells not expressing PECAM-1 stained for beta-catenin only at the membrane. Additionally, we demonstrate that PECAM-1 lacking the majority of the cytoplasmic domain promotes significantly less accumulation of transcriptionally active beta-catenin than full-length PECAM-1. Finally, we note an increased proliferative rate in the PECAM-1 reconstituted cells compared to the endothelial cells lacking PECAM-1. Taken together, our data suggest that PECAM-1, an adhesion molecule, affects cell proliferation via accumulation of transcriptionally active beta-catenin.


Subject(s)
Cytoskeletal Proteins/metabolism , Endothelium/cytology , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Platelet Endothelial Cell Adhesion Molecule-1/physiology , Trans-Activators/metabolism , Actins/metabolism , Animals , Blotting, Western , Cell Adhesion , Cell Division , Cytoplasm/metabolism , Flow Cytometry , Humans , Lung/cytology , Mice , Mice, Knockout , Microscopy, Fluorescence , Precipitin Tests , Signal Transduction , Transcription, Genetic , Transfection , beta Catenin
13.
J Cell Biol ; 160(4): 605-15, 2003 Feb 17.
Article in English | MEDLINE | ID: mdl-12591918

ABSTRACT

Atrioventricular (AV) septal defects resulting from aberrant endocardial cushion (EC) formation are observed at increased rates in infants of diabetic mothers. EC formation occurs via an epithelial-mesenchymal transformation (EMT), involving transformation of endocardial cells into mesenchymal cells, migration, and invasion into extracellular matrix. Here, we report that elevated glucose inhibits EMT by reducing myocardial vascular endothelial growth factor A (VEGF-A). This effect is reversed with exogenous recombinant mouse VEGF-A165, whereas addition of soluble VEGF receptor-1 blocks EMT. We show that disruption of EMT is associated with persistence of platelet endothelial cell adhesion molecule-1 (PECAM-1) and decreased matrix metalloproteinase-2 (MMP-2) expression. These findings correlate with retention of a nontransformed endocardial sheet and lack of invasion. The MMP inhibitor GM6001 blocks invasion, whereas explants from PECAM-1 deficient mice exhibit MMP-2 induction and normal EMT in high glucose. PECAM-1-negative endothelial cells are highly motile and express more MMP-2 than do PECAM-1-positive endothelial cells. During EMT, loss of PECAM-1 similarly promotes single cell motility and MMP-2 expression. Our findings suggest that high glucose-induced inhibition of AV cushion morphogenesis results from decreased myocardial VEGF-A expression and is, in part, mediated by persistent endocardial cell PECAM-1 expression and failure to up-regulate MMP-2 expression.


Subject(s)
Angiogenesis Inducing Agents/metabolism , Glucose/metabolism , Heart/embryology , Matrix Metalloproteinase 2/metabolism , Morphogenesis , Myocardium/metabolism , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Vascular Endothelial Growth Factor A , Angiogenesis Inducing Agents/genetics , Animals , Cell Movement/physiology , Cell Size , Cells, Cultured , Culture Techniques , Dipeptides/metabolism , Embryo, Mammalian/anatomy & histology , Embryo, Mammalian/physiology , Endocardial Cushion Defects , Female , Male , Matrix Metalloproteinase 2/genetics , Mice , Myocardium/cytology , Platelet Endothelial Cell Adhesion Molecule-1/genetics , Protease Inhibitors/metabolism , Recombinant Fusion Proteins/metabolism , Vascular Endothelial Growth Factor Receptor-1/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...