Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Sci Rep ; 14(1): 218, 2024 01 02.
Article in English | MEDLINE | ID: mdl-38168761

ABSTRACT

Notch signaling is universally conserved in metazoans where it is important for a wide variety of both normal and abnormal physiology. All four mammalian Notch receptors are activated by a conserved mechanism that releases Notch intracellular domains (NICDs) from the plasma membrane to translocate to the nucleus. Once there, NICDs interact through highly conserved ankyrin domains to form head-to-head homodimers on Notch sensitive promoters and stimulate transcription. Due to the highly conserved nature of these Notch ankyrin domains in all four mammalian Notch proteins, we hypothesized that NICDs may also engage in heterodimerization. Our results reveal the presence of two NICD dimerization states that can both engage in homo and heterodimerization. Using a Co-IP approach, we show that all NICD's can form non-transcriptionally active dimers and that the N4ICD appears to perform this function better than the other NICDs. Using a combination of ChIP analysis and transcriptional reporter assays, we also demonstrate the formation of transcriptionally active heterodimers that form on DNA. In particular, we demonstrate heterodimerization between the N2ICD and N4ICD and show that this heterodimer pair appears to exhibit differential activity on various Notch sensitive promoters. These results illustrate a new diversification of Notch signaling mechanisms which will help us better understand basic Notch function.


Subject(s)
Ankyrins , Receptors, Notch , Animals , Ankyrins/genetics , Receptors, Notch/genetics , Receptors, Notch/metabolism , Promoter Regions, Genetic , Mammals/metabolism
2.
Res Sq ; 2023 Jul 26.
Article in English | MEDLINE | ID: mdl-37546896

ABSTRACT

Notch signaling is universally conserved in metazoans where it is important for a wide variety of both normal and abnormal physiology. All four mammalian Notch receptors are activated by a conserved mechanism that releases Notch intracellular domains (NICDs) from the plasma membrane to translocate to the nucleus. Once there, NICDs interact through highly conserved ankyrin domains to form head-to-head homodimers on Notch sensitive promoters and stimulate transcription. Due to the highly conserved nature of these Notch ankyrin domains in all four mammalian Notch proteins, we hypothesized that NICDs may also engage in heterodimerization. Our results reveal the presence of two NICD dimerization states that can both engage in homo and heterodimerization. Using a Co-IP approach, we show that all NICD's can form non-transcriptionally active dimers and that the N4ICD appears to perform this function better than the other NICDs. Using a combination of ChIP analysis and transcriptional reporter assays, we also demonstrate the formation of transcriptionally active heterodimers that form on DNA. In particular, we demonstrate heterodimerization between the N2ICD and N4ICD and show that this heterodimer pair appears to exhibit differential activity on various Notch sensitive promoters. These results illustrate a new diversification of Notch signaling mechanisms which will help us better understand basic Notch function.

3.
PLoS One ; 15(11): e0234101, 2020.
Article in English | MEDLINE | ID: mdl-33232323

ABSTRACT

Notch signaling is essential for multicellular life, regulating core functions such as cellular identity, differentiation, and fate. These processes require highly sensitive systems to avoid going awry, and one such regulatory mechanism is through Notch intracellular domain dimerization. Select Notch target genes contain sequence-paired sites (SPS); motifs in which two Notch transcriptional activation complexes can bind and interact through Notch's ankyrin domain, resulting in enhanced transcriptional activation. This mechanism has been mostly studied through Notch1, and to date, the abilities of the other Notch family members have been left unexplored. Through the utilization of minimalized, SPS-driven luciferase assays, we were able to test the functional capacity of Notch dimers. Here we show that the Notch 2 and 3 NICDs also exhibit dimerization-induced signaling, following the same stringent requirements as seen with Notch1. Furthermore, our data suggested that Notch4 may also exhibit dimerization-induced signaling, although the amino acids required for Notch4 NICD dimerization appear to be different than those required for Notch 1, 2, and 3 NICD dimerization. Interestingly, we identified a mechanical difference between canonical and cryptic SPSs, leading to differences in their dimerization-induced regulation. Finally, we profiled the Notch family members' SPS gap distance preferences and found that they all prefer a 16-nucleotide gap, with little room for variation. In summary, this work highlights the potent and highly specific nature of Notch dimerization and refines the scope of this regulatory function.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Mutagenesis , Receptors, Notch/chemistry , Receptors, Notch/metabolism , Animals , Base Sequence , HEK293 Cells , Humans , Mice , Promoter Regions, Genetic , Protein Binding , Protein Domains , Protein Multimerization , Receptor, Notch2/chemistry , Receptor, Notch2/genetics , Receptor, Notch2/metabolism , Receptor, Notch3/chemistry , Receptor, Notch3/genetics , Receptor, Notch3/metabolism , Receptor, Notch4/chemistry , Receptor, Notch4/genetics , Receptor, Notch4/metabolism , Receptors, Notch/genetics , Signal Transduction , Transcriptional Activation
4.
Int J Mol Sci ; 21(6)2020 Mar 20.
Article in English | MEDLINE | ID: mdl-32244989

ABSTRACT

The Center of Biomedical Research Excellence in Matrix Biology strives to improve our understanding of extracellular matrix at molecular, cellular, tissue, and organismal levels to generate new knowledge about pathophysiology, normal development, and regenerative medicine. The primary goals of the Center are to i) support junior investigators, ii) enhance the productivity of established scientists, iii) facilitate collaboration between both junior and established researchers, and iv) build biomedical research infrastructure that will support research relevant to cell-matrix interactions in disease progression, tissue repair and regeneration, and v) provide access to instrumentation and technical support. A Pilot Project program provides funding to investigators who propose applying their expertise to matrix biology questions. Support from the National Institute of General Medical Sciences at the National Institutes of Health that established the Center of Biomedical Research Excellence in Matrix Biology has significantly enhanced the infrastructure and the capabilities of researchers at Boise State University, leading to new approaches that address disease diagnosis, prevention, and treatment. New multidisciplinary collaborations have been formed with investigators who may not have previously considered how their biomedical research programs addressed fundamental and applied questions involving the extracellular matrix. Collaborations with the broader matrix biology community are encouraged.


Subject(s)
Biomedical Research , Cooperative Behavior , Extracellular Matrix/metabolism , Research Personnel , Advisory Committees , Career Choice , Humans , Students
5.
PLoS One ; 14(1): e0210607, 2019.
Article in English | MEDLINE | ID: mdl-30653610

ABSTRACT

Resveratrol is a polyphenolic compound produced by plants which makes its way into the human diet through plant-based foods. It has been shown to provide many health benefits, helping to ward of age-related diseases and promoting cardiovascular health. Additionally, resveratrol is a potent activator of the Notch signaling pathway. While resveratrol receives the most attention as a polyphenolic nutraceutical, other compounds with similar structures may be more potent regulators of specific cellular processes. Here, we compare resveratrol, apigenin, chrysin, genistein, luteolin, myricetin, piceatannol, pterostilbene, and quercetin for their ability to regulate Notch signaling. In addition, we compare the ability of these polyphenolic compounds to regulate endothelial cell viability, proliferation, and migration. Out of these compounds we found that resveratrol is the best activator of Notch signaling, however, other similar compounds are also capable of stimulating Notch. We also discovered that several of these polyphenols were able to inhibit endothelial cell proliferation. Finally, we found that many of these polyphenols are potent inhibitors of endothelial migration during wound healing assays. These findings provide the first side-by-side comparison of the regulation of Notch signaling, and endothelial cell proliferation and migration, by nine polyphenolic compounds.


Subject(s)
Cell Proliferation/drug effects , Endothelial Cells/drug effects , Polyphenols/pharmacology , Receptors, Notch/metabolism , Resveratrol/pharmacology , Signal Transduction/drug effects , Antioxidants/pharmacology , Apigenin/pharmacology , Cell Movement/drug effects , Cell Survival/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Endothelial Cells/cytology , Endothelial Cells/metabolism , HEK293 Cells , Humans , Quercetin/pharmacology
6.
Sci Rep ; 8(1): 15515, 2018 10 19.
Article in English | MEDLINE | ID: mdl-30341382

ABSTRACT

Notch signaling is a form of intercellular communication which plays pivotal roles at various stages in development and disease. Previous findings have hinted that integrins and extracellular matrix may regulate Notch signaling, although a mechanistic basis for this interaction had not been identified. Here, we reveal that the regulation of Notch by integrins and extracellular matrix is carried out by Src family kinases (SFKs) working downstream of integrins. We identify a physical interaction between the SFK member, c-Src, and the Notch intracellular domain (NICD) that is enhanced by ß3 integrin and the integrin binding ECM protein, MAGP2. Our results demonstrate that c-Src directly phosphorylates the NICD at specific tyrosine residues and that mutation of these phosphorylation sites increases Notch responsive transcriptional activity. Furthermore, we also find that phosphorylation of the NICD by SFKs attenuates Notch mediated transcription by decreasing recruitment of MAML to the Notch co-transcriptional complex. Finally, we also find that SFK activity decreases NICD half-life. Collectively, our results provide important mechanistic data that underlie the emerging role of Notch as a general sensor and responder to extracellular signals.


Subject(s)
CSK Tyrosine-Protein Kinase/metabolism , DNA-Binding Proteins/metabolism , Endothelium, Vascular/physiology , Extracellular Matrix/metabolism , Receptor, Notch1/metabolism , Transcription Factors/metabolism , Cell Line , Contractile Proteins/metabolism , Endothelium, Vascular/pathology , Half-Life , Humans , Integrin beta3/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Phosphorylation , Protein Binding , Protein Stability , Signal Transduction
7.
Int J Mol Sci ; 19(2)2018 Feb 02.
Article in English | MEDLINE | ID: mdl-29393909

ABSTRACT

The traditional view of integrins portrays these highly conserved cell surface receptors as mediators of cellular attachment to the extracellular matrix (ECM), and to a lesser degree, as coordinators of leukocyte adhesion to the endothelium. These canonical activities are indispensable; however, there is also a wide variety of integrin functions mediated by non-ECM ligands that transcend the traditional roles of integrins. Some of these unorthodox roles involve cell-cell interactions and are engaged to support immune functions such as leukocyte transmigration, recognition of opsonization factors, and stimulation of neutrophil extracellular traps. Other cell-cell interactions mediated by integrins include hematopoietic stem cell and tumor cell homing to target tissues. Integrins also serve as cell-surface receptors for various growth factors, hormones, and small molecules. Interestingly, integrins have also been exploited by a wide variety of organisms including viruses and bacteria to support infectious activities such as cellular adhesion and/or cellular internalization. Additionally, the disruption of integrin function through the use of soluble integrin ligands is a common strategy adopted by several parasites in order to inhibit blood clotting during hematophagy, or by venomous snakes to kill prey. In this review, we strive to go beyond the matrix and summarize non-ECM ligands that interact with integrins in order to highlight these non-traditional functions of integrins.


Subject(s)
Extracellular Matrix/metabolism , Hormones/metabolism , Integrins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Snake Venoms/metabolism , Viral Proteins/metabolism , Animals , Cell Communication , Cell Movement/drug effects , Cell Movement/immunology , Endothelium/cytology , Endothelium/immunology , Extracellular Matrix/immunology , Extracellular Traps/immunology , Extracellular Traps/metabolism , Gene Expression Regulation , Hormones/pharmacology , Humans , Integrins/immunology , Intercellular Signaling Peptides and Proteins/pharmacology , Leukocytes/cytology , Leukocytes/immunology , Ligands , Opsonin Proteins/immunology , Opsonin Proteins/metabolism , Protein Binding , Signal Transduction , Snake Venoms/toxicity
8.
Dev Biol ; 418(2): 227-41, 2016 10 15.
Article in English | MEDLINE | ID: mdl-27565024

ABSTRACT

The Notch signaling cascade is an evolutionarily ancient system that allows cells to interact with their microenvironmental neighbors through direct cell-cell interactions, thereby directing a variety of developmental processes. Recent research is discovering that Notch signaling is also responsive to a broad variety of stimuli beyond cell-cell interactions, including: ECM composition, crosstalk with other signaling systems, shear stress, hypoxia, and hyperglycemia. Given this emerging understanding of Notch responsiveness to microenvironmental conditions, it appears that the classical view of Notch as a mechanism enabling cell-cell interactions, is only a part of a broader function to integrate microenvironmental cues. In this review, we summarize and discuss published data supporting the idea that the full function of Notch signaling is to serve as an integrator of microenvironmental signals thus allowing cells to sense and respond to a multitude of conditions around them.


Subject(s)
Receptors, Notch/physiology , Animals , Cellular Microenvironment/physiology , Extracellular Matrix/physiology , Humans , Hyperglycemia/physiopathology , Hypoxia/physiopathology , Integrins/physiology , Models, Biological , Receptor Cross-Talk/physiology , Signal Transduction/physiology , Stress, Physiological/physiology , Transforming Growth Factor beta/physiology , Vascular Endothelial Growth Factor A/physiology , Wnt Signaling Pathway/physiology
9.
Exp Cell Res ; 341(1): 84-91, 2016 Feb 01.
Article in English | MEDLINE | ID: mdl-26808411

ABSTRACT

Canonical Notch signaling involves Notch receptor activation via interaction with cell surface bound Notch ligand. Recent findings also indicate that Notch signaling may be modulated by cross-talk with other signaling mechanisms. The ECM protein MAGP2 was previously shown to regulate Notch in a cell type dependent manner, although the molecular details of this interaction have not been dissected. Here, we report that MAGP2 cell type specific control of Notch is independent of individual Notch receptor-ligand combinations but dependent on interaction with RGD binding integrins. Overexpressed MAGP2 was found to suppress transcriptional activity from the Notch responsive Hes1 promoter activity in endothelial cells, while overexpression of a RGD→RGE MAGP2 mutant increased Notch signaling in the same cell type. This effect was not unique to MAGP2 since the RGD domain of the ECM protein EGFL7 was also found to be an important modulator of Hes1 promoter activity. Independently of MAGP2 or EGFL7, inhibition of RGD-binding integrins with soluble RGD peptides also increased accumulation of active N1ICD fragments and Notch responsive promoter activity independently of changes in Notch1, Jag1, or Dll4 expression. Finally, ß1 or ß3 integrin blocking antibodies also enhanced Notch signaling. Collectively, these results answer the question of how MAGP2 controls cell type dependent Notch signaling, but more importantly uncover a new mechanism to understand how extracellular matrices and cellular environments impact Notch signaling.


Subject(s)
Contractile Proteins/metabolism , Glycoproteins/metabolism , Integrins/metabolism , Oligopeptides/metabolism , Receptors, Notch/metabolism , Signal Transduction , Humans , Intercellular Signaling Peptides and Proteins
10.
PLoS One ; 10(3): e0119279, 2015.
Article in English | MEDLINE | ID: mdl-25775018

ABSTRACT

Cyclosporin A (CSA) suppresses immune function by blocking the cyclophilin A and calcineurin/NFAT signaling pathways. In addition to immunosuppression, CSA has also been shown to have a wide range of effects in the cardiovascular system including disruption of heart valve development, smooth muscle cell proliferation, and angiogenesis inhibition. Circumstantial evidence has suggested that CSA might control Notch signaling which is also a potent regulator of cardiovascular function. Therefore, the goal of this project was to determine if CSA controls Notch and to dissect the molecular mechanism(s) by which CSA impacts cardiovascular homeostasis. We found that CSA blocked JAG1, but not Dll4 mediated Notch1 NICD cleavage in transfected 293T cells and decreased Notch signaling in zebrafish embryos. CSA suppression of Notch was linked to cyclophilin A but not calcineurin/NFAT inhibition since N-MeVal-4-CsA but not FK506 decreased Notch1 NICD cleavage. To examine the effect of CSA on vascular development and function, double transgenic Fli1-GFP/Gata1-RFP zebrafish embryos were treated with CSA and monitored for vasculogenesis, angiogenesis, and overall cardiovascular function. Vascular patterning was not obviously impacted by CSA treatment and contrary to the anti-angiogenic activity ascribed to CSA, angiogenic sprouting of ISV vessels was normal in CSA treated embryos. Most strikingly, CSA treated embryos exhibited a progressive decline in blood flow that was associated with eventual collapse of vascular luminal structures. Vascular collapse in zebrafish embryos was partially rescued by global Notch inhibition with DAPT suggesting that disruption of normal Notch signaling by CSA may be linked to vascular collapse. However, multiple signaling pathways likely cause the vascular collapse phenotype since both cyclophilin A and calcineurin/NFAT were required for normal vascular function. Collectively, these results show that CSA is a novel inhibitor of Notch signaling and vascular function in zebrafish embryos.


Subject(s)
Cyclosporine/pharmacology , Neovascularization, Physiologic/drug effects , Receptors, Notch/metabolism , Signal Transduction/drug effects , Zebrafish/embryology , Animals , Calcineurin/metabolism , Calcium-Binding Proteins/metabolism , Cyclophilin A/metabolism , Embryo, Nonmammalian/blood supply , Embryo, Nonmammalian/drug effects , Gene Expression Regulation/drug effects , HEK293 Cells , Humans , Intercellular Signaling Peptides and Proteins/metabolism , Jagged-1 Protein , Membrane Proteins/metabolism , Serrate-Jagged Proteins , Zebrafish Proteins
11.
Cancer Microenviron ; 6(3): 263-71, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23775523

ABSTRACT

A series of overexpression studies have shown that lumican suppresses angiogenesis in tumors produced from pancreatic adenocarcinoma, fibrosarcoma, and melanoma tumor cells. Despite lumican's anti-angiogenic activity, a clear correlation of differential expression of lumican in various cancers and cancer malignancy has failed to emerge. Therefore, we hypothesized that either 1.) endogenously expressed lumican is not anti-angiogenic or alternatively that 2.) lumican exhibits angiostatic activity only in limited microenvironments. Previously, lumican was shown to suppress tumor growth and angiogenesis in subcutaneously injected PanO2 pancreatic adenocarcinoma cells. Therefore, to determine if endogenously expressed lumican is anti-angiogenic we subcutaneously injected PanO2 cells into wild-type and lumican knockout mice and compared tumor growth and vascular densities of the resulting tumors. We found that tumors grown in lumican knockout animals were larger and contained significantly elevated vascular densities compared to those grown in wild-type mice. Interestingly however lumican knockout animals did not exhibit enhanced angiogenesis in aortic ring assays, matrigel plugs, or healing wound biopsies raising the possibility that lumican suppresses angiogenesis only in tumor microenvironments. To test this possibility, we sought a tumor model wherein lumican did not exhibit anti-angiogenic activity. Utilizing the 4T1 breast cancer model, we found that lumican suppressed 4T1 tumor growth and lung metastasis, but not angiogenesis. In conclusion, these results show that the angiostatic activity of lumican is dependent on currently undefined microenvironmental cues and therefore helps to understand why differential expression of lumican does not consistently correlate with human tumor malignancy.

12.
Microvasc Res ; 85: 24-33, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23110920

ABSTRACT

Matrix Gla Protein (MGP) is an ECM molecule commonly associated with dysfunctions of large blood vessels such as arteriosclerosis and atherosclerosis. However, the exact role of MGP in the microvasculature is not clear. Utilizing a mouse MGP knockout model we found that MGP suppresses angiogenic sprouting from mouse aorta restricts microvascular density in cardiac and skeletal muscle, and is an endogenous inhibitor of tumor angiogenesis. Similarly, morpholino based knockdown of MGP in zebrafish embryos caused a progressive loss of luminal structures in intersegmental vessels, a phenotype reminiscent of Dll4/Notch inhibition. Accordingly, MGP suppressed Notch-dependent Hes-1 promoter activity and expression of Jagged1 mRNA relative to Dll4 mRNA. However, inhibition of BMP but not Notch or VEGF signaling reversed the excessive angiogenic sprouting phenotype of MGP knockout aortic rings suggesting that MGP may normally suppress angiogenic sprouting by blocking BMP signaling. Collectively, these results suggest that MGP is a multi-functional inhibitor of normal and abnormal angiogenesis that may function by coordinating with both Notch and BMP signaling pathways.


Subject(s)
Calcium-Binding Proteins/physiology , Extracellular Matrix Proteins/physiology , Neoplasms/blood supply , Neovascularization, Pathologic , Animals , Aorta/pathology , Calcium-Binding Proteins/metabolism , Extracellular Matrix Proteins/metabolism , Female , Gene Expression Regulation, Developmental , Immunohistochemistry/methods , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microcirculation , Muscle, Skeletal/metabolism , Myocardium/metabolism , Neovascularization, Physiologic , Phenotype , Polymerase Chain Reaction/methods , Receptors, Notch/metabolism , Zebrafish , Matrix Gla Protein
13.
PLoS One ; 5(11): e13905, 2010 Nov 09.
Article in English | MEDLINE | ID: mdl-21085487

ABSTRACT

BACKGROUND: Matricellular proteins are extracellular regulators of cellular adhesion, signaling and performing a variety of physiological behaviors such as proliferation, migration and differentiation. Within vascular microenvironments, matricellular proteins exert both positive and negative regulatory cues to vascular endothelium. The relative balance of these matricellular cues is believed to be critical for vascular homeostasis, angiogenesis activation or angiogenesis resolution. However, our knowledge of matricellular proteins within vascular microenvironments and the mechanisms by which these proteins impact vascular function remain largely undefined. The matricellular protein lipocalin-7 (LCN7) is found throughout vascular microenvironments, and circumstantial evidence suggests that LCN7 may be an important regulator of angiogenesis. Therefore, we hypothesized that LCN7 may be an important regulator of vascular function. METHODOLOGY AND PRINCIPAL FINDINGS: To test this hypothesis, we examined the effect of LCN7 overexpression, recombinant protein and gene knockdown in a series of in vitro and in vivo models of angiogenesis. We found that overexpression of LCN7 in MB114 and SVEC murine endothelial cell lines or administration of highly purified recombinant LCN7 protein increased endothelial cell invasion. Similarly, LCN7 increased angiogenic sprouting from quiescent endothelial cell monolayers and ex vivo aortic rings. Moreover, LCN7 increased endothelial cell sensitivity to TGF-ß but did not affect sensitivity to other pro-angiogenic growth factors including bFGF and VEGF. Finally, morpholino based knockdown of LCN7 in zebrafish embryos specifically inhibited angiogenic sprouting but did not affect vasculogenesis within injected embryos. CONCLUSIONS AND SIGNIFICANCE: No functional analysis has previously been performed to elucidate the function of LCN7 in vascular or other cellular processes. Collectively, our results show for the first time that LCN7 is an important pro-angiogenic matricellular protein of vascular microenvironments.


Subject(s)
Cell Movement/drug effects , Endothelial Cells/drug effects , Lipocalins/pharmacology , Neoplasm Proteins/pharmacology , Neovascularization, Physiologic/drug effects , Amino Acid Sequence , Animals , Aorta/drug effects , Aorta/physiology , Blotting, Western , Cell Line , Cell Proliferation/drug effects , Embryo, Nonmammalian/embryology , Embryo, Nonmammalian/metabolism , Endothelial Cells/metabolism , Endothelial Cells/physiology , Gene Expression , Gene Knockdown Techniques , In Vitro Techniques , Lipocalins/genetics , Lipocalins/metabolism , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology , Reverse Transcriptase Polymerase Chain Reaction , Sequence Homology, Amino Acid , Transforming Growth Factor beta/pharmacology , Zebrafish/embryology , Zebrafish/genetics
14.
Cancer Microenviron ; 4(1): 115-26, 2010 Nov 18.
Article in English | MEDLINE | ID: mdl-21505566

ABSTRACT

Matrikines are important components of tumor microenvironments that integrate communication between extracellular matricies and membrane-bound receptors thereby regulating cellular behaviors. One such matrikine that is differentially expressed in cancer microenvironments is the extracellular matrix protein lumican; however its precise role in cancer remains ambiguous. To study the effects of lumican on cancer cells, we created lumican-overexpressing cell lines from murine fibrosarcoma (MCA102) and pancreatic adenocarcinoma (Pan02) cells. Lumican overexpression in Pan02 cells increased invasiveness, decreased soft agar colony size, and increased proliferation. Conversely in MCA102 cells, lumican decreased invasiveness, increased soft agar colony size, but did not influence proliferation. In contrast to these pleiotropic in vitro results, lumican overexpression within the in vivo tumor microenvironment produced uniformly smaller tumors. Importantly, reduced tumor size was correlated with reduced vascular density. Consistent with lumican's proposed anti-angiogenic activity, lumican increased endothelial cell apoptosis. Importantly, lumican was previously shown to influence Fas expression and our results show that lumican enhanced Fas mediated endothelial cell apoptosis although we were unable to detect any difference in Fas or Fas ligand expression between lumican-overexpressing and control cells. Interestingly, lumican had no effect on MCA102 apoptosis, suggesting that the observed reduction in tumor size is specifically due to endothelial cell apoptosis rather than a direct effect on the cancerous cells themselves. Therefore, this study is the first to demonstrate a causal relationship between tumor reduction and lumican's effect on angiogenesis as opposed to an effect on the cancerous cells themselves.

15.
Carcinogenesis ; 29(12): 2243-51, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18713838

ABSTRACT

Epithelial-mesenchymal transition (EMT) is a normal physiological process that regulates tissue development, remodeling and repair; however, aberrant EMT also elicits disease development in humans, including lung fibrosis, rheumatoid arthritis and cancer cell metastasis. Transforming growth factor-beta (TGF-beta) is a master regulator of EMT in normal mammary epithelial cells (MECs), wherein this pleiotropic cytokine also functions as a potent suppressor of mammary tumorigenesis. In contrast, malignant MECs typically evolve resistance to TGF-beta-mediated cytostasis and develop the ability to proliferate, invade and metastasize when stimulated by TGF-beta. It therefore stands to reason that establishing how TGF-beta promotes EMT may offer new insights into targeting the oncogenic activities of TGF-beta in human breast cancers. By monitoring alterations in the actin cytoskeleton and various markers of EMT, we show here that the TGF-beta gene target, fibulin-5 (FBLN5), initiates EMT and enhances that induced by TGF-beta. Whereas normal MECs contain few FBLN5 transcripts, those induced to undergo EMT by TGF-beta show significant upregulation of FBLN5 messenger RNA, suggesting that EMT and the dedifferentiation of MECs override the repression of FBLN5 expression in polarized MECs. We also show that FBLN5 stimulated matrix metalloproteinase expression and activity, leading to MEC invasion and EMT, to elevated Twist expression and to reduced E-cadherin expression. Finally, FBLN5 promoted anchorage-independent growth in normal and malignant MECs, as well as enhanced the growth of 4T1 tumors in mice. Taken together, these findings identify a novel EMT and tumor-promoting function for FBLN5 in developing and progressing breast cancers.


Subject(s)
Breast Neoplasms/pathology , Cell Transformation, Neoplastic/metabolism , Extracellular Matrix Proteins/metabolism , Metalloproteases/metabolism , Recombinant Proteins/metabolism , Transforming Growth Factor beta/metabolism , Animals , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Differentiation/physiology , Cell Line, Tumor , Epithelial Cells/metabolism , Epithelial Cells/pathology , Extracellular Matrix Proteins/genetics , Female , Immunohistochemistry , Mammary Glands, Animal/pathology , Mice , RNA, Messenger/analysis , Recombinant Proteins/genetics , Reverse Transcriptase Polymerase Chain Reaction
16.
Microvasc Res ; 76(1): 7-14, 2008 May.
Article in English | MEDLINE | ID: mdl-18417156

ABSTRACT

Angiogenesis is highly sensitive to the composition of the vascular microenvironment, however, our understanding of the structural and matricellular components of the vascular microenvironment that regulate angiogenesis and the molecular mechanisms by which these molecules function remains incomplete. Our previous results described a novel pro-angiogenic activity for Microfibril-Associated Glycoprotein-2 (MAGP-2), but did not address the molecular mechanism(s) by which this is accomplished. We now demonstrate that MAGP-2 promotes angiogenic cell sprouting by antagonizing Notch signaling pathways in endothelial cells. MAGP-2 decreased basal and Jagged1 induced expression from the Notch sensitive Hes-1 promoter in ECs, and blocked Jagged1 stimulated Notch1 receptor processing in transiently transfected 293T cells. Interestingly, inhibition of Notch signaling by MAGP-2 seems to be restricted to ECs since MAGP-2 increased Hes-1 promoter activity and Notch1 receptor processing in heterologous cell types. Importantly, constitutive activation of the Notch signaling pathway blocked the ability of MAGP-2 to promote angiogenic cell sprouting, as well as morphological changes associated with angiogenesis. Collectively, these observations indicate that MAGP-2 promotes angiogenic cell spouting in vitro by antagonizing Notch signaling pathways in ECs.


Subject(s)
Contractile Proteins/physiology , Endothelial Cells/metabolism , Glycoproteins/physiology , Neovascularization, Physiologic/physiology , Receptors, Notch/physiology , Signal Transduction/physiology , Actins/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Cell Line , Cell Line, Tumor , Cells, Cultured , Contractile Proteins/genetics , Contractile Proteins/pharmacology , Dipeptides/pharmacology , Endothelial Cells/cytology , Endothelial Cells/drug effects , Enzyme Inhibitors/pharmacology , Gene Expression/drug effects , Glycoproteins/genetics , Glycoproteins/pharmacology , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Intercellular Junctions/metabolism , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Jagged-1 Protein , Luciferases/genetics , Luciferases/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Microscopy, Fluorescence , Neovascularization, Physiologic/drug effects , Receptor, Notch1/genetics , Receptor, Notch1/metabolism , Receptors, Notch/genetics , Recombinant Proteins/pharmacology , Serrate-Jagged Proteins , Signal Transduction/drug effects , Transcription Factor HES-1 , Transfection , Transforming Growth Factor beta/pharmacology , beta Catenin/metabolism
17.
Angiogenesis ; 10(3): 197-216, 2007.
Article in English | MEDLINE | ID: mdl-17632767

ABSTRACT

Remodeling of vascular microenvironments during normal and tumor-induced angiogenesis is an important, yet poorly understood mechanism by which endothelial cells (ECs) contribute to the activation or resolution of angiogenesis. We used microarray analyses to monitor changes in the transcriptome of ECs undergoing angiogenesis when cultured onto Matrigel matrices. This strategy identified 308 genes whose expression in ECs was altered at least 3-fold by angiogenesis, of which 63 genes were found to encode for secretory proteins. In vitro assays that modeled key steps in the angiogenic process showed that several identified genes possessed pro- or anti-angiogenic activities (e.g., SMOC-2, secreted modular calcium-binding protein-2; CRELD-2, cysteine-rich with EGF-like domains-1; MAGP-2, microfibril-associated glycoprotein-2; lumican; and ECM-1, extracellular matrix protein-1). In particular, MAGP-2 expression potentiated EC proliferation and p38 MAPK activation stimulated by the pro-angiogenic factors, basic fibroblast growth factor (bFGF), epidermal growth factor (EGF), and vascular endothelial growth factor (VEGF); it also stimulated EC invasion and angiogenic sprouting, and more importantly, promoted the development and infiltration of vessels into Matrigel plugs implanted into genetically normal mice. Conversely, lumican inhibited EC activation of p38 MAPK, as well as their invasion, angiogenic sprouting, and vessel formation in mice. Collectively, our findings provide new insights into how EC stromal remodeling regulates angiogenesis activation and resolution, as well as identify two novel EC-secreted stromal proteins that modulate angiogenesis both in vitro and in vivo.


Subject(s)
Chondroitin Sulfate Proteoglycans/biosynthesis , Collagen/metabolism , Contractile Proteins/biosynthesis , Extracellular Matrix Proteins/biosynthesis , Gene Expression Regulation , Keratan Sulfate/biosynthesis , Laminin/metabolism , Neovascularization, Physiologic , Proteoglycans/metabolism , Transcription, Genetic , Animals , Brain/blood supply , Capillaries/cytology , Cells, Cultured , Chondroitin Sulfate Proteoglycans/genetics , Contractile Proteins/genetics , Drug Combinations , Endothelial Cells/cytology , Endothelial Cells/metabolism , Endothelium, Vascular/cytology , Extracellular Matrix/metabolism , Extracellular Matrix Proteins/genetics , Keratan Sulfate/genetics , Lumican , Mice , Oligonucleotide Array Sequence Analysis , RNA Splicing Factors , Recombinant Proteins/biosynthesis , Retroviridae/genetics
18.
Cancer Res ; 66(5): 2621-9, 2006 Mar 01.
Article in English | MEDLINE | ID: mdl-16510581

ABSTRACT

Lethal tumor growth and progression cannot occur without angiogenesis, which facilitates cancer cell proliferation, survival, and dissemination. Fibulins (FBLN) 5 and 3 are widely expressed extracellular matrix proteins that regulate cell proliferation in a context-specific manner. Reduced FBLN-5 expression has been associated with cancer formation and progression in humans, whereas its constitutive expression antagonizes endothelial cell angiogenic sprouting in vitro. Thus, FBLN-5 may suppress tumorigenesis by preventing tumor angiogenesis. FBLN-3 is homologous to FBLN-5 and expressed in endothelial cells, yet its role in tumorigenesis and angiogenesis is unknown. We find FBLN-3 expression to be altered in some human tumors and that its constitutive expression in endothelial cells inhibited their proliferation, invasion, and angiogenic sprouting, as well as their response to vascular endothelial growth factor as measured by p38 mitogen-activated protein kinase activation. In endothelial cells, both FBLNs (a) reduced angiogenic sprouting stimulated by basic fibroblast growth factor (bFGF); (b) inhibited matrix metalloproteinase expression and activity; and (c) stimulated tissue inhibitor of metalloproteinase expression. More importantly, both FBLNs prevented angiogenesis and vessel infiltration into bFGF-supplemented Matrigel plugs implanted in genetically normal mice, as well as decreased the growth and blood vessel density in tumors produced by MCA102 fibrosarcoma cells implanted s.c. into syngeneic mice. Our findings establish FBLN-3 and FBLN-5 as novel angiostatic agents capable of reducing tumor angiogenesis and, consequently, tumor growth in vivo and suggest that these angiostatic activities may one day be exploited to combat tumor angiogenesis and metastasis in cancer patients.


Subject(s)
Extracellular Matrix Proteins/physiology , Fibrosarcoma/blood supply , Amino Acid Sequence , Animals , Brain/blood supply , Cells, Cultured , Endothelial Cells/cytology , Endothelial Cells/metabolism , Extracellular Matrix Proteins/biosynthesis , Extracellular Matrix Proteins/genetics , Female , Fibrosarcoma/genetics , Humans , Matrix Metalloproteinase Inhibitors , Matrix Metalloproteinases/biosynthesis , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics , Sequence Homology, Amino Acid , Thrombospondin 1/antagonists & inhibitors , Thrombospondin 1/biosynthesis , Tissue Inhibitor of Metalloproteinases/antagonists & inhibitors , Tissue Inhibitor of Metalloproteinases/biosynthesis
19.
Future Oncol ; 1(1): 23-35, 2005 Feb.
Article in English | MEDLINE | ID: mdl-16555973

ABSTRACT

Tumorigenesis is the process by which normal cells evolve the capacity to evade and overcome the constraints normally placed upon their growth and survival. During cancer progression, indolent tumors experience an array of genetic and epigenetic events that ultimately coordinate the development of tumor metastasis, which is the most lethal facet of cancer and the leading cause of cancer-related death. The therapeutic necessity to combat tumor metastasis continues to drive investigations aimed at identifying novel regulators of this deadly process. Fibulin-5 is a newly described extracellular matrix protein that is important for normal embryonic development and organogenesis. Fibulin-5 expression may also be associated with the suppression of tumor formation through its control of cell proliferation, motility and angiogenic sprouting. Here, the tumor suppressing activities of fibulin-5 are reviewed, and the potential use and targeting of fibulin-5 to combat growth and metastasis of human malignancies is discussed.


Subject(s)
Extracellular Matrix Proteins/physiology , Neoplasms/etiology , Amino Acid Sequence , Animals , Cell Differentiation , Epithelial Cells/cytology , Extracellular Matrix Proteins/chemistry , Humans , Mesoderm/cytology , Mitogen-Activated Protein Kinase 1/physiology , Mitogen-Activated Protein Kinase 3/physiology , Molecular Sequence Data , Neovascularization, Physiologic , Recombinant Proteins/chemistry
20.
Mol Biol Cell ; 16(2): 609-25, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15548600

ABSTRACT

Tubulogenesis by epithelial cells regulates kidney, lung, and mammary development, whereas that by endothelial cells regulates vascular development. Although functionally dissimilar, the processes necessary for tubulation by epithelial and endothelial cells are very similar. We performed microarray analysis to further our understanding of tubulogenesis and observed a robust induction of regulator of G protein signaling 4 (RGS4) mRNA expression solely in tubulating cells, thereby implicating RGS4 as a potential regulator of tubulogenesis. Accordingly, RGS4 overexpression delayed and altered lung epithelial cell tubulation by selectively inhibiting G protein-mediated p38 MAPK activation, and, consequently, by reducing epithelial cell proliferation, migration, and expression of vascular endothelial growth factor (VEGF). The tubulogenic defects imparted by RGS4 in epithelial cells, including its reduction in VEGF expression, were rescued by overexpression of constitutively active MKK6, an activator of p38 MAPK. Similarly, RGS4 overexpression abrogated endothelial cell angiogenic sprouting by inhibiting their synthesis of DNA and invasion through synthetic basement membranes. We further show that RGS4 expression antagonized VEGF stimulation of DNA synthesis and extracellular signal-regulated kinase (ERK)1/ERK2 and p38 MAPK activation as well as ERK1/ERK2 activation stimulated by endothelin-1 and angiotensin II. RGS4 had no effect on the phosphorylation of Smad1 and Smad2 by bone morphogenic protein-7 and transforming growth factor-beta, respectively, indicating that RGS4 selectively inhibits G protein and VEGF signaling in endothelial cells. Finally, we found that RGS4 reduced endothelial cell response to VEGF by decreasing VEGF receptor-2 (KDR) expression. We therefore propose RGS4 as a novel antagonist of epithelial and endothelial cell tubulogenesis that selectively antagonizes intracellular signaling by G proteins and VEGF, thereby inhibiting cell proliferation, migration, and invasion, and VEGF and KDR expression.


Subject(s)
Epithelial Cells/drug effects , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Neovascularization, Physiologic/physiology , RGS Proteins/metabolism , Vascular Endothelial Growth Factors/drug effects , Animals , Brain/blood supply , Calcium-Calmodulin-Dependent Protein Kinases/antagonists & inhibitors , Cell Line , Cell Movement/drug effects , Cell Proliferation/drug effects , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Enzyme Activation , Epithelial Cells/metabolism , Lung/cytology , Lung/growth & development , Mice , Microarray Analysis , Mink , Models, Biological , Morphogenesis , RGS Proteins/chemistry , RNA, Messenger/metabolism , Signal Transduction , Vascular Endothelial Growth Factors/genetics , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors
SELECTION OF CITATIONS
SEARCH DETAIL
...