Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Biotechnol Bioeng ; 103(4): 796-807, 2009 Jul 01.
Article in English | MEDLINE | ID: mdl-19326468

ABSTRACT

We are creating synthetic pharmaceuticals with angiogenic activity and potential to promote vascular invasion. We previously demonstrated that one of these molecules, phthalimide neovascular factor 1 (PNF1), significantly expands microvascular networks in vivo following sustained release from poly(lactic-co-glycolic acid) (PLAGA) films. In addition, to probe PNF1 mode of action, we recently applied a novel pathway-based compendium analysis to a multi-timepoint, controlled microarray data set of PNF1-treated (vs. control) human microvascular endothelial cells (HMVECs), and we identified induction of tumor necrosis factor-alpha (TNF-alpha) and, subsequently, transforming growth factor-beta (TGF-beta) signaling networks by PNF1. Here we validate this microarray data set with quantitative real-time polymerase chain reaction (RT-PCR) analysis. Subsequently, we probe this data set and identify three specific TGF-beta-induced genes with regulation by PNF1 conserved over multiple timepoints-amyloid beta (A4) precursor protein (APP), early growth response 1 (EGR-1), and matrix metalloproteinase 14 (MMP14 or MT1-MMP)-that are also implicated in angiogenesis. We further focus on MMP14 given its unique role in angiogenesis, and we validate MT1-MMP modulation by PNF1 with an in vitro fluorescence assay that demonstrates the direct effects that PNF1 exerts on functional metalloproteinase activity. We also utilize endothelial cord formation in collagen gels to show that PNF1-induced stimulation of endothelial cord network formation in vitro is in some way MT1-MMP-dependent. Ultimately, this new network analysis of our transcriptional footprint characterizing PNF1 activity 1-48 h post-supplementation in HMVECs coupled with corresponding validating experiments suggests a key set of a few specific targets that are involved in PNF1 mode of action and important for successful promotion of the neovascularization that we have observed by the drug in vivo.


Subject(s)
Endothelial Cells/drug effects , Enzyme Inhibitors/pharmacology , Indoles/pharmacology , Matrix Metalloproteinase 14/metabolism , Cells, Cultured , Gene Expression Profiling , Humans , Oligonucleotide Array Sequence Analysis , Organ Culture Techniques
2.
J Biomed Mater Res A ; 91(2): 317-23, 2009 Nov.
Article in English | MEDLINE | ID: mdl-18980190

ABSTRACT

Poly(D,L-lactic-co-glycolic acid) (PLGA) is a biodegradable polymer that is widely used for drug delivery. However, the degradation of PLGA alters the local microenvironment and may influence tissue structure and/or function. Here, we studied whether PLGA degradation affects the structure of the arteriolar microcirculation through arteriogenic expansion of maximum lumenal diameters and/or the formation of new smooth muscle-coated vessels. Single microspheres comprised of 50:50 PLGA (521 +/- 52.7 microm diameter), 50:50 PLGA with bovine serum albumin (BSA) (547 +/- 62.2 microm), 85:15 PLGA (474 +/- 52.6 microm), or 85:15 PLGA with BSA (469 +/- 57.2 microm) were implanted into mouse dorsal skinfold window chambers, and longitudinal arteriolar diameter measurements were made in the presence of a vasodilator (10(-4)M adenosine) over 7 days. At the end of the 7-day period, the length density of all smooth muscle-coated microvessels was also determined. Implantation of the window chamber alone elicited a 22% increase in maximum arteriolar diameter. However, the addition of 85:15 and 50:50 PLGA microspheres, bearing either BSA or no protein, elicited a significant enhancement of this arteriogenic response, with final maximum arteriolar diameters ranging from 36 to 46% more than their original size. Interestingly, the influence of PLGA degradation on microvascular structure was limited to lumenal arteriolar expansion, as we observed no significant differences in length density of smooth muscle-coated microvessels. We conclude that the degradation of PLGA microspheres may elicit an arteriogenic response in subcutaneous tissue in the dorsal skinfold window chamber; however, it has no apparent effect on the total length of smooth muscle-coated microvasculature.


Subject(s)
Biocompatible Materials/metabolism , Lactic Acid/metabolism , Polyglycolic Acid/metabolism , Skin/blood supply , Absorbable Implants , Actins/analysis , Animals , Arterioles/metabolism , Arterioles/ultrastructure , Cattle , Male , Mice , Mice, Inbred C57BL , Microspheres , Muscle, Smooth/ultrastructure , Polylactic Acid-Polyglycolic Acid Copolymer , Serum Albumin, Bovine/administration & dosage , Serum Albumin, Bovine/metabolism , Skin/ultrastructure
3.
Biomaterials ; 29(35): 4698-708, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18804278

ABSTRACT

Phthalimide neovascular factor (PNF1, formerly SC-3-149) is a potent stimulator of proangiogenic signaling pathways in endothelial cells. In this study, we evaluated the in vivo effects of sustained PNF1 release to promote ingrowth and expansion of microvascular networks surrounding biomaterial implants. The dorsal skinfold window chamber was used to evaluate the structural remodeling response of the local microvasculature. PNF1 was released from poly(lactic-co-glycolic acid) (PLAGA) films, and a transport model was utilized to predict PNF1 penetration into the surrounding tissue. PNF1 significantly expanded microvascular networks within a 2mm radius from implants after 3 and 7 days by increasing microvessel length density and lumenal diameter of local arterioles and venules. Staining of histological sections with CD11b showed enhanced recruitment of circulating white blood cells, including monocytes, which are critical for the process of vessel enlargement through arteriogenesis. As PNF1 has been shown to modulate MT1-MMP, a facilitator of CCL2 dependent leukocyte transmigration, aspects of window chamber experiments were repeated in CCR2(-/-) (CCL2 receptor) mouse chimeras to more fully explore the critical nature of monocyte recruitment on the therapeutic benefits of PNF1 function in vivo.


Subject(s)
Indoles/pharmacology , Microvessels/drug effects , Neovascularization, Physiologic/drug effects , Animals , Cell Line , Cell Movement/drug effects , Cell Proliferation/drug effects , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Monocytes/drug effects , Receptors, CCR2/deficiency , Receptors, CCR2/genetics
4.
Bioinformatics ; 24(20): 2384-90, 2008 Oct 15.
Article in English | MEDLINE | ID: mdl-18718940

ABSTRACT

MOTIVATION: Computational techniques have been applied to experimental datasets to identify drug mode-of-action. A shortcoming of existing approaches is the requirement of large reference databases of compound expression profiles. Here, we developed a new pathway-based compendium analysis that couples multi-timepoint, controlled microarray data for a single compound with systems-based network analysis to elucidate drug mechanism more efficiently. RESULTS: We applied this approach to a transcriptional regulatory footprint of phthalimide neovascular factor 1 (PNF1)-a novel synthetic small molecule that exhibits significant in vitro endothelial potency-spanning 1-48 h post-supplementation in human micro-vascular endothelial cells (HMVEC) to comprehensively interrogate PNF1 effects. We concluded that PNF1 first induces tumor necrosis factor-alpha (TNF-alpha) signaling pathway function which in turn affects transforming growth factor-beta (TGF-beta) signaling. These results are consistent with our previous observations of PNF1-directed TGF-beta signaling at 24 h, including differential regulation of TGF-beta-induced matrix metalloproteinase 14 (MMP14/MT1-MMP) which is implicated in angiogenesis. Ultimately, we illustrate how our pathway-based compendium analysis more efficiently generates hypotheses for compound mechanism than existing techniques.


Subject(s)
Angiogenesis Inducing Agents/pharmacology , Gene Expression Profiling , Indoles/pharmacology , Signal Transduction , Cells, Cultured , Endothelial Cells/metabolism , Humans , Matrix Metalloproteinase 14/metabolism , Models, Biological , Oligonucleotide Array Sequence Analysis , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism , Tumor Necrosis Factor-alpha/metabolism
5.
Biomaterials ; 29(19): 2869-77, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18405965

ABSTRACT

Sphingosine 1-phosphate (S1P) is a bioactive phospholipid that impacts migration, proliferation, and survival in diverse cell types, including endothelial cells, smooth muscle cells, and osteoblast-like cells. In this study, we investigated the effects of sustained release of S1P on microvascular remodeling and associated bone defect healing in vivo. The murine dorsal skinfold window chamber model was used to evaluate the structural remodeling response of the microvasculature. Our results demonstrated that 1:400 (w/w) loading and subsequent sustained release of S1P from poly(lactic-co-glycolic acid) (PLAGA) significantly enhanced lumenal diameter expansion of arterioles and venules after 3 and 7 days. Incorporation of 5-bromo-2-deoxyuridine (BrdU) at day 7 revealed significant increases in mural cell proliferation in response to S1P delivery. Additionally, three-dimensional (3D) scaffolds loaded with S1P (1:400) were implanted into critical-size rat calvarial defects, and healing of bony defects was assessed by radiograph X-ray, microcomputed tomography (muCT), and histology. Sustained release of S1P significantly increased the formation of new bone after 2 and 6 weeks of healing and histological results suggest increased numbers of blood vessels in the defect site. Taken together, these experiments support the use of S1P delivery for promoting microvessel diameter expansion and improving the healing outcomes of tissue-engineered therapies.


Subject(s)
Arteries/growth & development , Bone Development , Lysophospholipids/administration & dosage , Sphingosine/analogs & derivatives , Tissue Engineering , Animals , Bromodeoxyuridine , Fluorescent Antibody Technique , Male , Mice , Mice, Inbred C57BL , Rats , Sphingosine/administration & dosage
6.
Tissue Eng ; 13(10): 2561-75, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17723106

ABSTRACT

Neovascularization is essential for the survival and successful integration of most engineering tissues after implantation in vivo. The objective of this study was to elucidate possible mechanisms of phthalimide neovascular factor 1 (PNF1), a new synthetic small molecule proposed for therapeutic induction of angiogenesis. Complementary deoxyribonucleic acid microarray analysis was used to identify 568 transcripts in human microvascular endothelial cells (HMVECs) that were significantly regulated after 24-h stimulation with 30 muM of PNF1, previously known as SC-3-149. Network analysis tools were used to identify genetic networks of the global biological processes involved in PNF1 stimulation and to describe known molecular and cellular functions that the drug regulated most highly. Examination of the most significantly perturbed networks identified gene products associated with transforming growth factor-beta (TGF-beta), which has many known effects on angiogenesis, and related signal transduction pathways. These include molecules integral to the thrombospondin, plasminogen, fibroblast growth factor, epidermal growth factor, ephrin, Rho, and Ras signaling pathways that are essential to endothelial function. Moreover, real-time reverse-transcriptase polymerase chain reaction (RT-PCR) of select genes showed significant increases in TGF-beta-associated receptors endoglin and beta glycan. These experiments provide important insight into the pro-angiogenic mechanism of PNF1, namely, TGF-beta-associated signaling pathways, and may ultimately offer new molecular targets for directed drug discovery.


Subject(s)
Angiogenic Proteins/metabolism , Endothelial Cells/physiology , Gene Expression Profiling/methods , Neovascularization, Physiologic/drug effects , Neovascularization, Physiologic/genetics , Signal Transduction/drug effects , Signal Transduction/genetics , Cells, Cultured , Drug Delivery Systems/methods , Endothelial Cells/drug effects , Humans , Software
7.
Tissue Eng ; 12(7): 1903-13, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16889520

ABSTRACT

Engineering of implantable tissues requires rapid induction of angiogenesis to meet the significant oxygen and nutrient demands of cells during tissue repair. To this end, our laboratories have utilized medicinal chemistry to synthesize non-peptide-based inducers of angiogenesis to aid tissue engineering. In this study, we describe the evaluation of SC-3-149, a small molecule compound with proliferative effects on vascular endothelial cells. Specifically, exogenous exposure of SC-3-149 induced an 18-fold increase in proliferation of human microvascular endothelial cells in vitro at low micromolar potency by day 14 in culture. Moreover, SC-3-149 significantly increased the formation of endothelial cord and tubelike structures in vitro, and improved endothelial scratch wound healing within 24 h. SC-3-149 also significantly inhibited vascular endothelial cell death owing to serum deprivation and high acidity (pH 6). Concurrent incubation of SC-3-149 with vascular endothelial growth factor increased cell survivability under serum-deprived conditions by an additional 7%. In addition, in vivo injection of SC-3-149 into the rat mesentery produced qualitative increases in microvessel length density. Taken together, our studies suggest that SC-3-149 and its analogs may serve as promising new angiogenic agents for targeted drug delivery and therapeutic angiogenesis in tissue engineering.


Subject(s)
Angiogenesis Inducing Agents/pharmacology , Cell Proliferation/drug effects , Endothelial Cells/physiology , Heterocyclic Compounds, 3-Ring/pharmacology , Indoles/chemical synthesis , Neovascularization, Physiologic/drug effects , Angiogenesis Inducing Agents/chemical synthesis , Animals , Bioprosthesis , Cells, Cultured , Endothelial Cells/cytology , Female , Heterocyclic Compounds, 3-Ring/chemical synthesis , Humans , Indoles/pharmacology , Rats , Rats, Sprague-Dawley , Time Factors , Tissue Engineering , Wound Healing/drug effects
8.
Langmuir ; 21(1): 225-8, 2005 Jan 04.
Article in English | MEDLINE | ID: mdl-15620307

ABSTRACT

A direct comparison of surface loading, interface shear strength, and interface hydrolytic stability was made between a phosphonate and two siloxane monolayers formed on the native oxide surface of Ti-6Al-4V. Surface loading for the phosphonate was ca. four times greater (on a nanomole/area basis) than for the siloxanes; mechanical strengths per surface-bound molecule were comparable, but the hydrolytic stability (pH 7.5) of the siloxanes was poor. These results suggest that phosphonate monolayer interfaces are more desirable than comparable siloxane ones for applications where such interfaces contact even slightly alkaline water.

SELECTION OF CITATIONS
SEARCH DETAIL