Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
1.
J Biol Chem ; 286(49): 42017-42026, 2011 Dec 09.
Article in English | MEDLINE | ID: mdl-22002053

ABSTRACT

The vascular endothelium continually senses and responds to biochemical and mechanical stimuli to appropriately initiate angiogenesis. We have shown previously that fluid wall shear stress (WSS) and sphingosine 1-phosphate (S1P) cooperatively initiate the invasion of human umbilical vein endothelial cells into collagen matrices (Kang, H., Bayless, K. J., and Kaunas, R. (2008) Am. J. Physiol. Heart Circ. Physiol. 295, H2087-2097). Here, we investigated the role of calpains in the regulation of endothelial cell invasion in response to WSS and S1P. Calpain inhibition significantly decreased S1P- and WSS-induced invasion. Short hairpin RNA-mediated gene silencing demonstrated that calpain 1 and 2 were required for WSS and S1P-induced invasion. Also, S1P synergized with WSS to induce invasion and to activate calpains and promote calpain membrane localization. Calpain inhibition results in a cell morphology consistent with reduced matrix proteolysis. Membrane type 1-matrix metalloproteinase (MT1-MMP) has been shown by others to regulate endothelial cell invasion, prompting us to test whether calpain acted upstream of MT1-MMP. S1P and WSS synergistically activated MT1-MMP and induced cell membrane localization of MT1-MMP in a calpain-dependent manner. Calpain activation, MT1-MMP activation and MT1-MMP membrane localization were all maximal with 5.3 dynes/cm(2) WSS and S1P treatment, which correlated with maximal invasion responses. Our data show for the first time that 5.3 dynes/cm(2) WSS in the presence of S1P combine to activate calpains, which direct MT1-MMP membrane localization to initiate endothelial sprouting into three-dimensional collagen matrices.


Subject(s)
Calpain/metabolism , Lysophospholipids/metabolism , Matrix Metalloproteinase 14/metabolism , Sphingosine/analogs & derivatives , Cell Membrane/metabolism , Endothelial Cells/cytology , Endothelium, Vascular/pathology , Extracellular Matrix/metabolism , Gene Silencing , Humans , Microscopy, Fluorescence/methods , Neovascularization, Pathologic , Shear Strength , Sphingosine/metabolism , Stress, Mechanical , Subcellular Fractions/metabolism , Umbilical Veins/cytology
2.
Angiogenesis ; 15(2): 287-303, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22407449

ABSTRACT

Endothelial cells normally line the vasculature and remain quiescent. However, these cells can be rapidly stimulated to undergo morphogenesis and initiate new blood vessel formation given the proper cues. This study reports a new mechanism for initiating angiogenic sprout formation that involves vimentin, the major intermediate filament protein in endothelial cells. Initial studies confirmed vimentin was required for sphingosine 1-phosphate (S1P)- and growth factor (GF)-induced endothelial cell invasion, and vimentin was cleaved by calpains during invasion. Calpains were predominantly activated by GF and were required for sprout initiation. Because others have reported membrane type 1-matrix metalloproteinase (MT1-MMP) is required for endothelial sprouting responses, we tested whether vimentin and calpain acted upstream of MT1-MMP. Both calpain and vimentin were required for successful MT1-MMP membrane translocation, which was stimulated by S1P. In addition, vimentin complexed with MT1-MMP in a manner that required both the cytoplasmic domain of MT1-MMP and calpain activation, which increased the soluble pool of vimentin in endothelial cells. Altogether, these data indicate that pro-angiogenic signals converge to activate calpain-dependent vimentin cleavage and increase vimentin solubility, which act upstream to facilitate MT1-MMP membrane translocation, resulting in successful endothelial sprout formation in three-dimensional collagen matrices. These findings help explain why S1P and GF synergize to stimulate robust sprouting in 3D collagen matrices.


Subject(s)
Calpain/metabolism , Cell Membrane/metabolism , Endothelial Cells/metabolism , Matrix Metalloproteinase 14/metabolism , Proteolysis , Vimentin/metabolism , Calpain/genetics , Cell Membrane/genetics , Cells, Cultured , Endothelial Cells/cytology , Humans , Lysophospholipids/metabolism , Matrix Metalloproteinase 14/genetics , Protein Transport/physiology , Solubility , Sphingosine/analogs & derivatives , Sphingosine/metabolism , Vimentin/genetics
3.
EMBO Mol Med ; 13(7): e14089, 2021 07 07.
Article in English | MEDLINE | ID: mdl-34102002

ABSTRACT

The tyrosine phosphatase SHP2 is oncogenic in cancers driven by receptor-tyrosine-kinases, and SHP2 inhibition reduces tumor growth. Here, we report that SHP2 is an essential promoter of endothelial cell survival and growth in the remodeling tumor vasculature. Using genetic and chemical approaches to inhibit SHP2 activity in endothelial cells, we show that SHP2 inhibits pro-apoptotic STAT3 and stimulates proliferative ERK1/2 signaling. Systemic SHP2 inhibition in mice bearing tumor types selected for SHP2-independent tumor cell growth promotes degeneration of the tumor vasculature and blood extravasation; reduces tumor vascularity and blood perfusion; and increases tumor necrosis. Reduction of tumor growth ensues, independent of SHP2 targeting in the tumor cells, blocking immune checkpoints, or recruiting macrophages. We also show that inhibiting the Angiopoietin/TIE2/AKT cascade magnifies the vascular and anti-tumor effects of SHP2 inhibition by blocking tumor endothelial AKT signaling, not a target of SHP2. Since the SHP2 and Ang2/TIE2 pathways are active in vascular endothelial cells of human melanoma and colon carcinoma, SHP2 inhibitors alone or with Ang2/TIE2 inhibitors hold promise to effectively target the tumor endothelium.


Subject(s)
Neoplasms , Protein Tyrosine Phosphatase, Non-Receptor Type 11 , Animals , Endothelial Cells/metabolism , Mice , Neoplasms/drug therapy , Protein Tyrosine Phosphatase, Non-Receptor Type 11/metabolism , Receptor Protein-Tyrosine Kinases , Signal Transduction
4.
Biol Reprod ; 82(5): 876-87, 2010 May.
Article in English | MEDLINE | ID: mdl-20107206

ABSTRACT

Because sphingosine 1-phosphate (S1P) is a potent stimulator of angiogenesis, we hypothesized that the S1P pathway is activated to stimulate endometrial/placental angiogenesis during pregnancy. We initially localized S1P signaling pathway members in the gravid and nongravid uterine horns of unilaterally pregnant ewes. Sphingosine kinase-1 expression was greater in gravid compared to nongravid horns. In situ hybridization revealed elevated expression of sphingosine 1-phosphate phosphatase (SGPP1) in gravid interplacentomal endometrial stroma on Days 20 and 40 compared to the nongravid uterine horn, but expression increased in endometrium of the nongravid uterine horn between Days 40 and 120. SGPP1 expression increased in placentomes late in gestation. Sphingosine 1-phosphate lyase mRNA was modestly expressed at Day 20 and then decreased. In contrast, sphingosine 1-phosphate receptor 1 (S1PR1) mRNA increased in endometrium and caruncular stroma of the gravid uterine horn. Treatment with FTY720 and VPC23019, S1P receptor antagonists, blocked human and ovine endothelial cell invasion using an in vitro model of sprouting angiogenesis. Knockdown of S1PR1 with siRNA reduced invasion responses as well. We previously reported that delta-like 4 (DLL4) and A disintegrin and metalloproteinase with thrombospondin-like repeats 1 (ADAMTS1) participate in endothelial cell invasion stimulated by S1P and growth factors in vitro, and thus investigated whether their expression correlated with areas undergoing angiogenesis in vivo. DLL4 expression was similar to S1PR1, while ADAMTS1 mRNA was expressed by endometria of both nongravid and gravid horns, as well as conceptus and placentomes. These results establish that S1P signaling pathway members and S1P- and growth factor-regulated genes are prominent in uterine and placental tissue and in some cases are correlated with areas undergoing angiogenesis. Thus, S1P signaling may be crucial for proper fetal-placental development.


Subject(s)
Endometrium/metabolism , Lysophospholipids/metabolism , Neovascularization, Physiologic/physiology , Placentation/physiology , Pregnancy, Animal , Sphingosine/analogs & derivatives , Animals , Cell Line , Endometrium/blood supply , Female , Humans , Lyases/genetics , Lyases/metabolism , Lysophospholipids/genetics , Membrane Proteins/genetics , Membrane Proteins/metabolism , Phosphoric Monoester Hydrolases/genetics , Phosphoric Monoester Hydrolases/metabolism , Placenta/blood supply , Placenta/metabolism , Pregnancy , RNA, Messenger/analysis , Receptors, Lysosphingolipid/genetics , Receptors, Lysosphingolipid/metabolism , Second Messenger Systems/physiology , Sheep , Signal Transduction/physiology , Sphingosine/genetics , Sphingosine/metabolism , Tissue Distribution
5.
Mol Oncol ; 13(11): 2441-2459, 2019 11.
Article in English | MEDLINE | ID: mdl-31545551

ABSTRACT

Advanced colorectal carcinoma is currently incurable, and new therapies are urgently needed. We report that phosphotyrosine-dependent Eph receptor signaling sustains colorectal carcinoma cell survival, thereby uncovering a survival pathway active in colorectal carcinoma cells. We find that genetic and biochemical inhibition of Eph tyrosine kinase activity or depletion of the Eph ligand EphrinB2 reproducibly induces colorectal carcinoma cell death by autophagy. Spautin and 3-methyladenine, inhibitors of early steps in the autophagic pathway, significantly reduce autophagy-mediated cell death that follows inhibition of phosphotyrosine-dependent Eph signaling in colorectal cancer cells. A small-molecule inhibitor of the Eph kinase, NVP-BHG712 or its regioisomer NVP-Iso, reduces human colorectal cancer cell growth in vitro and tumor growth in mice. Colorectal cancers express the EphrinB ligand and its Eph receptors at significantly higher levels than numerous other cancer types, supporting Eph signaling inhibition as a potential new strategy for the broad treatment of colorectal carcinoma.


Subject(s)
Autophagy , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/pathology , Molecular Targeted Therapy , Receptors, Eph Family/metabolism , Signal Transduction , Animals , Autophagy/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Ephrin-B2/metabolism , Female , Gene Silencing/drug effects , Mice , Mice, Nude , Protein Kinase Inhibitors/pharmacology , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Signal Transduction/drug effects , Survival Analysis
6.
J Natl Cancer Inst ; 109(9)2017 09 01.
Article in English | MEDLINE | ID: mdl-28376153

ABSTRACT

Background: Primary effusion lymphoma (PEL) is a Kaposi's sarcoma herpes virus (KSHV)-induced lymphoma that typically arises in body cavities of HIV-infected patients. PEL cells are often co-infected with Epstein-Barr virus (EBV). "PEL-like" lymphoma is a KSHV-unrelated lymphoma that arises in body cavities of HIV-negative patients. "PEL-like" lymphoma is sometimes EBV positive. The derivation of PEL/"PEL-like" cells is unclear. Methods: Mesothelial cells were cultured from body cavity effusions of 23 patients. Cell proliferation, cytokine secretion, marker phenotypes, KSHV/EBV infection, and clonality were evaluated by standard methods. Gene expression was measured by quantitative polymerase chain reaction and immunoblotting. A mouse model of PEL (3 mice/group) was used to evaluate tumorigenicity. Results: We found that the mesothelia derived from six effusions of HIV-infected patients with PEL or other KSHV-associated diseases contained rare KSHV + or EBV + mesothelial cells. After extended culture (16-17 weeks), some mesothelial cells underwent a trans-differentiation process, generating lymphoid-type CD45 + /B220 + , CD5 + , CD27 + , CD43 + , CD11c + , and CD3 - cells resembling "B1-cells," most commonly found in mouse body cavities. These "B1-like" cells were short lived. However, long-term KSHV + EBV - and EBV + KSHV - clonal cell lines emerged from mesothelial cultures from two patients that were clonally distinct from the monoclonal or polyclonal B-cell populations found in the patients' original effusions. Conclusions: Mesothelial-to-lymphoid transformation is a newly identified in vitro process that generates "B1-like" cells and is associated with the emergence of long-lived KSHV or EBV-infected cell lines in KSHV-infected patients. These results identify mesothelial cultures as a source of PEL cells and lymphoid cells in humans.


Subject(s)
Epithelium/pathology , Lymphoma, Primary Effusion/pathology , Adult , Aged , Animals , Disease Models, Animal , Epithelial-Mesenchymal Transition , Epstein-Barr Virus Infections/pathology , Epstein-Barr Virus Infections/virology , Herpesviridae Infections/pathology , Herpesviridae Infections/virology , Herpesvirus 4, Human/isolation & purification , Herpesvirus 8, Human/isolation & purification , Humans , Lymphoma, Primary Effusion/virology , Male , Mice , Mice, Inbred NOD , Mice, SCID , Middle Aged , Young Adult
7.
Sci Rep ; 6: 27195, 2016 06 02.
Article in English | MEDLINE | ID: mdl-27250641

ABSTRACT

Adult-type intraembryonic hematopoiesis arises from specialized endothelial cells of the dorsal aorta (DA). Despite the critical importance of this specialized endothelium for establishment of hematopoietic stem cells and adult hematopoietic lineages, the mechanisms regulating its emergence are incompletely understood. We show that EphrinB2, a principal regulator of endothelial cell function, controls the development of endothelium producing adult-type hematopoiesis. The absence of EphrinB2 impairs DA-derived hematopoiesis. Transmembrane EphrinB2 and its EphB4 receptor interact in the emerging DA, which transiently harbors EphrinB2(+) and EphB4(+) endothelial cells, thereby providing an opportunity for bi-directional cell-to-cell signaling to control the emergence of the hemogenic endothelium. Embryonic Stem (ES) cell-derived EphrinB2(+) cells are enriched with hemogenic endothelial precursors. EphrinB2 silencing impairs ES generation of hematopoietic cells but not generation of endothelial cells. The identification of EphrinB2 as an essential regulator of adult hematopoiesis provides important insight in the regulation of early hematopoietic commitment.


Subject(s)
Aorta/cytology , Ephrin-B2/metabolism , Hemangioblasts/cytology , Mouse Embryonic Stem Cells/cytology , Animals , Aorta/metabolism , Cell Differentiation , Cell Line , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Hemangioblasts/metabolism , Hematopoiesis , Mice , Mouse Embryonic Stem Cells/metabolism , Tissue Culture Techniques
8.
Sci Signal ; 9(419): ra28, 2016 Mar 15.
Article in English | MEDLINE | ID: mdl-26980441

ABSTRACT

Interleukin-23 (IL-23), a heterodimeric cytokine composed of the unique p19 peptide (IL-23p19) and a peptide called IL-12p40, which is shared with IL-12, is implicated in Crohn's disease, rheumatoid arthritis, psoriasis, and other immune-mediated inflammatory diseases. Endothelial cells produce the IL-23p19 peptide in the absence of the IL-12p40 chain and thus do not make heterodimeric IL-23. We found that intercellular IL-23p19 increased the cell surface abundances of intercellular adhesion molecule-1 (ICAM-1) and vascular cell adhesion molecule-1 (VCAM-1) on endothelial cells, which enhanced the attachment of leukocytes and increased their transendothelial migration. Intracellular p19 associated with the cytokine receptor subunit gp130 and stimulated the gp130-dependent activation of signal transducer and activator of transcription 3 (STAT3) signaling. Proinflammatory factors promoted the generation of IL-23p19 in endothelial cells. The adventitial capillaries of inflamed temporal arteries in patients with giant-cell arteritis (GCA) had endothelial p19 protein associated with gp130, but did not contain the IL-12p40 chain. Because adventitial capillaries are essential for the entry of inflammatory cells into arterial walls, these data suggest that p19 may contribute to GCA disease and could represent a therapeutic target. Our results provide evidence that IL-23p19 is a previously unrecognized endothelial proinflammatory peptide that promotes leukocyte transendothelial migration, advancing our current understanding of the complexities of inflammatory responses.


Subject(s)
Cytokine Receptor gp130/metabolism , Human Umbilical Vein Endothelial Cells/metabolism , Inflammation Mediators/metabolism , Interleukin-23 Subunit p19/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction , Cytokine Receptor gp130/genetics , Humans , Intercellular Adhesion Molecule-1/genetics , Intercellular Adhesion Molecule-1/metabolism , Interleukin-23 Subunit p19/genetics , STAT3 Transcription Factor/genetics , Vascular Cell Adhesion Molecule-1/genetics , Vascular Cell Adhesion Molecule-1/metabolism
9.
J Clin Invest ; 126(12): 4554-4568, 2016 12 01.
Article in English | MEDLINE | ID: mdl-27820703

ABSTRACT

Hematopoietic stem and progenitor cells (HSPCs) reside in the bone marrow. Stress signals from cancer and other conditions promote HSPC mobilization into circulation and subsequent homing to tissue microenvironments. HSPC infiltration into tissue microenvironments can influence disease progression; notably, in cancer, HSPCs encourage tumor growth. Here we have uncovered a mutually exclusive distribution of EPHB4 receptors in bone marrow sinusoids and ephrin B2 ligands in hematopoietic cells. We determined that signaling interactions between EPHB4 and ephrin B2 control HSPC mobilization from the bone marrow. In mice, blockade of the EPHB4/ephrin B2 signaling pathway reduced mobilization of HSPCs and other myeloid cells to the circulation. EPHB4/ephrin B2 blockade also reduced HSPC infiltration into tumors as well as tumor progression in murine models of melanoma and mammary cancer. These results identify EPHB4/ephrin B2 signaling as critical to HSPC mobilization from bone marrow and provide a potential strategy for reducing cancer progression by targeting the bone marrow.


Subject(s)
Bone Marrow/metabolism , Hematopoietic Stem Cell Mobilization , Hematopoietic Stem Cells/metabolism , Receptor, EphB4/metabolism , Signal Transduction/physiology , Stem Cell Niche/physiology , Animals , Cell Line , Ephrin-B2/genetics , Ephrin-B2/metabolism , Mice , Receptor, EphB4/genetics
10.
Cancer Res ; 74(7): 2038-49, 2014 Apr 01.
Article in English | MEDLINE | ID: mdl-24520074

ABSTRACT

Myeloid cells that orchestrate malignant progression in the tumor microenvironment offer targets for a generalized strategy to attack solid tumors. Through an analysis of tumor microenvironments, we explored an experimental model of lung cancer that uncovered a network of Dll4/Notch/TGF-ß1 signals that links myeloid cells to cancer progression. Myeloid cells attracted to the tumor microenvironment by the tumor-derived cytokines CCL2 and M-CSF expressed increased levels of the Notch ligand Dll4, thereby activating Notch signaling in the tumor cells and amplifying tumor-intrinsic Notch activation. Heightened Dll4/Notch signaling in tumor cells magnified TGF-ß-induced pSMAD2/3 signaling and was required to sustain TGF-ß-induced tumor cell growth. Conversely, Notch blockade reduced TGF-ß signaling and limited lung carcinoma tumor progression. Corroborating these findings, by interrogating RNAseq results from tumor and adjacent normal tissue in clinical specimens of human head and neck squamous carcinoma, we found evidence that TGF-ß/Notch crosstalk contributed to progression. In summary, the myeloid cell-carcinoma signaling network we describe uncovers novel mechanistic links between the tumor microenvironment and tumor growth, highlighting new opportunities to target tumors where this network is active.


Subject(s)
Intracellular Signaling Peptides and Proteins/physiology , Membrane Proteins/physiology , Myeloid Cells/pathology , Neoplasms, Experimental/pathology , Receptors, Notch/physiology , Signal Transduction/physiology , Transforming Growth Factor beta/physiology , Tumor Microenvironment , Adaptor Proteins, Signal Transducing , Animals , Calcium-Binding Proteins , Cell Proliferation , Disease Progression , Female , Male , Mice
11.
Cureus ; 5(1): 82, 2013 Jan 18.
Article in English | MEDLINE | ID: mdl-25493215

ABSTRACT

Although Vascular Endothelial Growth Factor (VEGF)-targeted therapies have shown efficacy in the treatment of certain advanced cancers, benefits to patients have been modest, which is attributed to tumor resistance to VEGF neutralization. Recent efforts to identify new targets to inhibit tumor angiogenesis have identified Bv8 (prokineticin 2), a myeloid cell-derived protein that promotes endothelial cell growth and tumor angiogenesis, but many mechanistic aspects of the pro-tumorigenic function of Bv8 are unclear. Here we demonstrate that CD11b+, Ly6C+, Ly6G+ granulocytes are the predominant cell source of Bv8 expression in bone marrow, spleen and in tumor tissues. Using granulocyte-deficient Growth factor independence-1 (Gfi1)-null mutant mice and normal littermates, we found that EL4 lymphoma tumors grow significantly larger in the granulocyte and Bv8-deficient mutant mice in comparison to the normal mice that display abundant tumor-associated granulocytes and Bv8 expression. Conversely, Lewis lung carcinoma (LLC-1) tumors grew to a significantly greater size in the normal mice in comparison to the Gfi1-null mice, but normal granulocyte tumor infiltration was modest. Quantitative analysis of tissue vascularization showed that EL4 and LLC-1 tumors from normal and Gfi1-mutant mice are similarly vascularized. These results confirm the critical contribution of the tumor microenvironment in determining the rate of tumor progression independently of tumor angiogenesis, and reveal some of the complexities of granulocyte and Bv8 functions in modulating tumor growth.

12.
Matrix Biol ; 28(8): 470-9, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19666115

ABSTRACT

In this study, we investigated potential mechanisms through which the known anti-angiogenic factor, tissue inhibitor of metalloproteinase-3 (TIMP-3) blocks angiogenesis. As a strategy to identify TIMP-3 binding proteins, we used tandem affinity purification, employing recombinant adenoviruses constructed to deliver TIMP-3 fused to C-terminal S and His tags (TIMP-3-S-His) or TIMP-1-S-His control to endothelial cells prior to extraction. Western blotting of final eluates revealed robust binding of A Disintegrin and Metalloproteinase (ADAM) 17 and a slight association of ADAM15 to TIMP-3, but not TIMP-1 control. To confirm a functional requirement for ADAM15 and 17 in mediating angiogenic events, a model of endothelial cell invasion was utilized. Silencing of ADAM17, but not ADAM15, expression using small interfering RNA (siRNA) interfered with invasion, resulting in decreased density of invading cells and decreased invasion distance. Stable EC lines expressing short hairpin RNA directed to ADAM17 were similarly inhibited. To confirm these results, dominant negative mutants (DeltaMPs) of ADAM10, ADAM15 or ADAM17 were delivered using recombinant lentiviruses. Expression of ADAM17 DeltaMP, but not ADAM10 or ADAM15 DeltaMP, decreased invasion density and distance. Further, time-lapse analyses revealed ADAM17 DeltaMP cells exhibited far greater numbers of protruding sprouts compared to control, suggesting an inability of extended processes to retract properly. Immunofluorescence analyses revealed ADAM17 localized to bifurcations in invading sprouts. These data jointly indicate a role for ADAM17 in modulating endothelial sprouting events during angiogenesis.


Subject(s)
ADAM Proteins/metabolism , Collagen , Endothelial Cells/metabolism , Neovascularization, Physiologic/physiology , Tissue Inhibitor of Metalloproteinase-3/metabolism , Tissue Scaffolds , ADAM Proteins/genetics , ADAM10 Protein , ADAM17 Protein , Amyloid Precursor Protein Secretases/genetics , Amyloid Precursor Protein Secretases/metabolism , Cell Surface Extensions/enzymology , Cells, Cultured , Endothelial Cells/cytology , Endothelial Cells/enzymology , Gene Expression/genetics , Humans , Membrane Proteins/genetics , Membrane Proteins/metabolism , Protein Interaction Mapping , RNA, Small Interfering/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Sequence Deletion/physiology , Tissue Inhibitor of Metalloproteinase-1/genetics , Tissue Inhibitor of Metalloproteinase-1/metabolism , Tissue Inhibitor of Metalloproteinase-3/genetics , Transduction, Genetic
13.
Nat Protoc ; 4(12): 1888-98, 2009.
Article in English | MEDLINE | ID: mdl-20010936

ABSTRACT

Seeding a monolayer of primary human endothelial cells on the surface of a polymerized three-dimensional collagen matrix in the presence of pro-angiogenic stimuli allows manipulation and analysis of rapid sprouting responses. This protocol is useful for elucidating incompletely defined intracellular mechanisms downstream of pro-angiogenic factors that regulate sprout formation and initiation, and can also be used to test the efficacy of pro-and anti-angiogenic compounds. We present protocols to culture endothelial cells, prepare three-dimensional collagen matrices and quantify and image rapid endothelial sprouting responses (24 h). This protocol can be carried out using either type I or type II collagen matrices with primary endothelial cells isolated from macrovascular and microvascular sources of varying species.


Subject(s)
Cell Culture Techniques , Collagen , Endothelial Cells/physiology , Angiogenesis Inducing Agents/pharmacology , Animals , Endothelium, Vascular/physiology , Green Fluorescent Proteins/analysis , Humans , Neovascularization, Physiologic , Rats
14.
Am J Physiol Cell Physiol ; 295(5): C1215-29, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18787072

ABSTRACT

Sprouting angiogenesis is a multistep process consisting of basement membrane degradation, endothelial cell (EC) activation, proliferation, invasion, lumen formation, and sprout stabilization. Such complexity is consistent with a requirement for orchestration of individual gene expression alongside multiple signaling pathways. To better understand the mechanisms that direct the transformation of adherent ECs on the surface of collagen matrices to develop multicellular invading sprouts, we analyzed differential gene expression with time using a defined in vitro model of EC invasion driven by the combination of sphingosine-1-phosphate, basic FGF, and VEGF. Gene expression changes were confirmed by real-time PCR and Western blot analyses. A cohort of cell adhesion molecule genes involved in adherens junction and cell-extracellular matrix (ECM) interactions were upregulated, whereas a set of genes associated with tight junctions were downregulated. Numerous genes encoding ECM proteins and proteases were induced, indicating that biosynthesis and remodeling of ECM is indispensable for sprouting angiogenesis. Knockdown of a highly upregulated gene, a disintegrin and metalloproteinase with thrombospondin-type repeats-1 (ADAMTS1), decreased invasion responses, confirming a role for ADAMTS1 in mediating EC invasion. Furthermore, differential expression of multiple members of the Wnt and Notch pathways was observed. Functional experiments indicated that inhibition and activation of the Notch signaling pathway stimulated and inhibited EC invasion responses, respectively. This study has enhanced the molecular road map of gene expression changes that occur during endothelial invasion and highlighted the utility of three-dimensional models to study EC morphogenesis.


Subject(s)
Cell Adhesion/genetics , Cell Movement/genetics , Collagen/metabolism , Endothelial Cells/metabolism , Gene Expression Profiling , Neovascularization, Physiologic/genetics , Wound Healing/genetics , ADAM Proteins/genetics , ADAM Proteins/metabolism , ADAMTS1 Protein , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/metabolism , Cells, Cultured , Endothelial Cells/enzymology , Extracellular Matrix Proteins/genetics , Extracellular Matrix Proteins/metabolism , Gene Expression Profiling/methods , Gene Expression Regulation , Humans , Oligonucleotide Array Sequence Analysis , Peptide Hydrolases/genetics , Peptide Hydrolases/metabolism , RNA, Messenger/metabolism , Receptors, Notch/genetics , Receptors, Notch/metabolism , Signal Transduction/genetics , Time Factors , Wnt Proteins/genetics , Wnt Proteins/metabolism
15.
Mol Cell Biol ; 28(6): 1936-46, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18160708

ABSTRACT

The short splice variant of the basic helix-loop-helix Per-Arnt-Sim transcription factor Singleminded-2, SIM2s, has been implicated in development and is frequently lost or reduced in primary breast tumors. Here, we show that loss of Sim2s causes aberrant mouse mammary gland ductal development with features suggestive of malignant transformation, including increased proliferation, loss of polarity, down-regulation of E-cadherin, and invasion of the surrounding stroma. Additionally, knockdown of SIM2s in MCF-7 breast cancer cells contributed to an epithelial-mesenchymal transition (EMT) and increased tumorigenesis. In both Sim2(-/-) mammary glands and SIM2s-depleted MCF7 cells, these changes were associated with increased SLUG and MMP2 levels. SIM2s protein was detectable on the SLUG promoter, and overexpression of SIM2s repressed expression from a SLUG-controlled reporter in a dose-dependent manner. To our knowledge, SIM2s is the first protein shown to bind and repress the SLUG promoter, providing a plausible explanation for the development role and breast tumor-suppressive activity of SIM2s. Together, our results suggest that SIM2s is a key regulator of mammary-ductal development and that loss of SIM2s expression is associated with an invasive, EMT-like phenotype.


Subject(s)
Adenocarcinoma/pathology , Basic Helix-Loop-Helix Transcription Factors/physiology , Breast Neoplasms/pathology , Cell Transdifferentiation/physiology , Cell Transformation, Neoplastic/genetics , Epithelial Cells/cytology , Mammary Glands, Animal/metabolism , Matrix Metalloproteinase 2/biosynthesis , Mesoderm/cytology , Transcription Factors/biosynthesis , Animals , Basic Helix-Loop-Helix Transcription Factors/deficiency , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Line, Tumor/cytology , Cell Line, Tumor/metabolism , Female , Gene Expression Regulation , Humans , Mammary Glands, Animal/cytology , Matrix Metalloproteinase 2/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Nude , Neoplasm Proteins/deficiency , Neoplasm Proteins/genetics , Neoplasm Proteins/physiology , Protein Isoforms/deficiency , Protein Isoforms/physiology , Snail Family Transcription Factors , Transcription Factors/genetics , Up-Regulation
16.
Carcinogenesis ; 28(2): 259-66, 2007 Feb.
Article in English | MEDLINE | ID: mdl-16840439

ABSTRACT

Single-minded 2 (SIM2) is a member of the bHLH-PAS family of transcription factors. SIM2 was initially identified by positional cloning on chromosome 21 and is thought to contribute to the etiology of trisomy-21 [Down syndrome (DS)]. In addition to the physical and mental deficiencies associated with this genetic disease, it has become apparent that women with DS are 10-25 times less likely to die from breast cancer in comparison with age-matched normal populations. This is thought to be a result of gene dosage effect of tumor suppressor genes on chromosome 21. Here, we report that a splice variant of SIM2, SIM2 short (SIM2s), is differentially expressed in normal breast and breast cancer-derived cell lines and is downregulated in human breast cancer samples. Re-establishment of SIM2s in MDA-MB-435 breast cancer cells significantly reduced proliferation, anchorage-independent growth and invasive potential. Consistent with its role as a transcriptional repressor, SIM2s directly decreased expression of matrix metalloprotease-3, a known mediator of breast cancer metastasis. These results suggest that SIM2s has breast tumor suppressive activity.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/physiology , Breast Neoplasms/pathology , Cell Division/physiology , Neoplasm Invasiveness , Base Sequence , Cell Line, Tumor , Chromosomes, Human, Pair 21 , DNA Primers , Gene Dosage , Humans , Matrix Metalloproteinase 3/genetics , Promoter Regions, Genetic
17.
J Biol Chem ; 281(16): 10839-48, 2006 Apr 21.
Article in English | MEDLINE | ID: mdl-16484282

ABSTRACT

Single-minded 1 and 2 are unique members of the basic helix-loop-helix Per-Arnt-Sim family as they are transcriptional repressors. Here we report the identification and transcriptional characterization of mouse Sim2s, a splice variant of Sim2, which is missing the carboxyl Pro/Ala-rich repressive domain. Sim2s is expressed at high levels in kidney and skeletal muscle; however, the ratio of Sim2 to Sim2s mRNA differs between these tissues. Similar to full-length Sim2, Sim2s interacts with Arnt and to a lesser extent, Arnt2. The effects of Sim2s on transcriptional regulation through hypoxia, dioxin, and central midline response elements are different than that of full-length Sim2. Specifically, Sim2s exerts a less repressive effect on hypoxia-induced gene expression than full-length Sim2, but is just as effective as Sim2 at repressing TCDD-induced gene expression from a dioxin response element. Interestingly, Sim2s bind to and activates expression from a central midline response element-controlled reporter through an Arnt transactivation domain-dependent mechanism. The differences in expression pattern, protein interactions, and transcriptional activities between Sim2 and Sim2s may reflect differential roles each isoform plays during development or in tissue-specific effects on other protein-mediated pathways.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/biosynthesis , Basic Helix-Loop-Helix Transcription Factors/genetics , Gene Expression Regulation , Alternative Splicing , Animals , Base Sequence , Cell Line , DNA Primers/chemistry , DNA, Complementary/metabolism , Dioxins/chemistry , Exons , Female , Genes, Reporter , Humans , Hypoxia , Immunoprecipitation , Kidney/metabolism , Luciferases/metabolism , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Muscle, Skeletal/metabolism , Plasmids/metabolism , Protein Biosynthesis , Protein Isoforms , Protein Structure, Tertiary , RNA/metabolism , Response Elements , Reverse Transcriptase Polymerase Chain Reaction , Sequence Homology, Nucleic Acid , Tissue Distribution , Transcription, Genetic , Transcriptional Activation , Transfection , Two-Hybrid System Techniques
SELECTION OF CITATIONS
SEARCH DETAIL