VEGF is essential for hypoxia-inducible factor-mediated neovascularization but dispensable for endothelial sprouting.
Journal: 2011/October - Proceedings of the National Academy of Sciences of the United States of America
ISSN: 1091-6490
Abstract:
Although our understanding of the molecular regulation of adult neovascularization has advanced tremendously, vascular-targeted therapies for tissue ischemia remain suboptimal. The master regulatory transcription factors of the hypoxia-inducible factor (HIF) family are attractive therapeutic targets because they coordinately up-regulate multiple genes controlling neovascularization. Here, we used an inducible model of epithelial HIF-1 activation, the TetON-HIF-1 mouse, to test the requirement for VEGF in HIF-1 mediated neovascularization. TetON-HIF-1, K14-Cre, and VEGF(flox/flox) alleles were combined to create TetON-HIF-1:VEGF(Δ) mice to activate HIF-1 and its target genes in adult basal keratinocytes in the absence of concomitant VEGF. HIF-1 induction failed to produce neovascularization in TetON-HIF-1:VEGF(Δ) mice despite robust up-regulation of multiple proangiogenic HIF targets, including PlGF, adrenomedullin, angiogenin, and PAI-1. In contrast, endothelial sprouting was preserved, enhanced, and more persistent, consistent with marked reduction in Dll4-Notch-1 signaling. Optical-resolution photoacoustic microscopy, which provides noninvasive, label-free, high resolution, and wide-field vascular imaging, revealed the absence of both capillary expansion and arteriovenous remodeling in serially imaged individual TetON-HIF-1:VEGF(Δ) mice. Impaired TetON-HIF-1:VEGF(Δ) neovascularization could be partially rescued by 12-O-tetradecanoylphorbol-13-acetate skin treatment. These data suggest that therapeutic angiogenesis for ischemic cardiovascular disease may require treatment with both HIF-1 and VEGF.
Relations:
Content
Citations
(57)
References
(58)
Grants
(63)
Conditions
(1)
Drugs
(1)
Chemicals
(4)
Genes
(2)
Organisms
(2)
Processes
(2)
Anatomy
(5)
Similar articles
Articles by the same authors
Discussion board
Proc Natl Acad Sci U S A 108(32): 13264-13269

VEGF is essential for hypoxia-inducible factor-mediated neovascularization but dispensable for endothelial sprouting

TetON-HIF-1:VEGF Mice Lack Angiogenesis and Vessel Remodeling.

To test the role of VEGF during epithelial HIF-1–induced neovascularization, we combined TetON-HIF-1 mice (11) with K14-Cre transgenic and VEGF floxed (VEGF) knock-in mice (1215). The final genotype of these composite mice was K14-rtTA:TRE-HIF-1α:K14-Cre:VEGF. This genotype, designated as TetON-HIF-1:VEGF, was n = 12 in the FVB/n strain. Doxycycline (DOX) treatment of 8- to 12-wk-old TetON-HIF-1:VEGF mice induced a robust neovascularization that peaked and plateaued 14 d after initiation (Fig. 1 A and B). In contrast, neovascularization failed to occur in TetON-HIF-1:VEGF composite mice (Fig. 1 A and B). Moreover, contrary to prior reports (16), there was no reduction of microvascular density in TetON-HIF-1:VEGF composite mice, at baseline before DOX induction compared with nontransgenic controls, likely due to differences in tissue fixation/epitope preservation techniques and endothelial marker antibody detection reagents. Induction of HIF-1α protein expression was detectable in both interfollicular epidermal and hair follicle outer root sheath basal keratinocytes in both TetON-HIF-1:VEGF and TetON-HIF-1:VEGF mice after 14 days of DOX induction (Fig. 1C).

An external file that holds a picture, illustration, etc.
Object name is pnas.1101321108fig01.jpg

Cre-mediated basal keratinocyte VEGF deletion abrogated HIF-1–induced neovascularization. (A) CD31 immunofluorescence (red) revealed robust neovascularization in TetON-HIF-1:VEGF (VEGF) compared with TetON-HIF-1:VEGF (VEGF) mice, which remained similar to baseline controls. DAPI was used as a nuclear counterstain. (B) Quantitative analysis of CD31+ vascular area revealed a threefold elevation in TetON-HIF-1:VEGF compared with TetON-HIF-1:VEGF mice induced with doxycycline for similar time intervals (n = 3–6 mice per time point). (C) Representative HIF-1α transgene immunofluorescence indicating localization to the epidermal and hair follicle basal cell compartment after 14 d of DOX induction. (B) VEGF or VEGF data at each DOX day were compared with VEGF day 0 data by using the unpaired Student's t test (**P < 0.01 and ***P < 0.001). (Scale bars: A, 200 μm: C, 50 μm.)

Temporal control of HIF-1 allowed us to deploy an emerging technology for noninvasive and label-free functional determination of microvascular dynamics, optical-resolution photoacoustic microscopy (OR-PAM) (17, 18). The resolving power and image segmentation capabilities of OR-PAM enabled multiparameter determination of vascular morphology including capillary volume as an indicator of angiogenesis, and arteriovenous (arteriolar/venular) volume, vessel length, diameter, and tortuosity as measures of vascular remodeling. Because OR-PAM uses hemoglobin for contrast (17), both the imaging and quantitative segmentation analysis were based on perfused vessels, which, similar to ante mortem lectin perfusion (10), is distinct from data derived from endothelial marker analysis of postmortem tissue sections. Serial OR-PAM monitoring of individual mice from days 0 to 60 of continuous DOX provision revealed that HIF-1 activation in TetON-HIF-1:VEGF mice produced an eightfold increase in capillary volume and a 3.5-fold elevation of arteriovenous and overall vascular volume compared with DOX-induced TetON-HIF-1:VEGF or day 0 TetON-HIF-1:VEGF controls (Fig. 2A and Fig. S1B). OR-PAM segmentation analysis suggested that capillary volume fell between days 14 and 30 in the TetON-HIF-1:VEGF mice (Fig. S1). The difference between these data and the microvessel density analysis based on CD31 decoration of all vessels could be due to the lack of perfusion of CD31-positive vessels or that OR-PAM acquires wide field volumetric data and immunofluorescence analyzes microvascular area in 2D tissue sections. OR-PAM also demonstrated abrogation of HIF-1–mediated vasodilatation and tortuosity induction in the absence of epithelial VEGF (Fig. S1B).

An external file that holds a picture, illustration, etc.
Object name is pnas.1101321108fig02.jpg

TetON-HIF-1:VEGF mice lacked vessel remodeling. (A) Angiogenesis determination and accompanying vascular remodeling by OR-PAM. OR-PAM was performed in the same VEGF or VEGF transgenic mouse serially for 60 d (days 0–30 are represented). Increased capillary density (red asterisk) is evident particularly by day 14, whereas arteriovenous remodeling (increase in vessel diameter and tortuosity) is detectable at day 14 and prominent by day 30 (blue arrow) (see also Fig. S1). (Scale bar: 500 μm.)

Loss of HIF-1–Induced VEGF Produces Sprouting Without Endothelial Proliferation.

An outstanding feature of the TetON-HIF-1 model is the ability to dissect discrete stages of neovascular network development and its maintenance due to temporal control of HIF-1 gain of function (11). Here, we tested the necessity of VEGF for HIF-1 induction of the earliest stages of neovascularization, endothelial sprouting, and proliferation. DOX-treated NTG and day 0 TetON-HIF-1:VEGF mice were used interchangeably in all studies, because they were phenotypically and functionally similar. Endothelial proliferation remained at barely detectable levels after HIF-1 activation in TetON-HIF-1:VEGF mice, in contrast to a robust induction in TetON-HIF-1:VEGF counterparts (Fig. S2A). Surprisingly, TetON-HIF-1:VEGF mice evidenced a prolonged interval of endothelial sprouting up to day 14 during HIF-1 induction (Fig. 3A, Left and Fig. S2B). In contrast, sprout elaboration in TetON-HIF-1:VEGF was restricted to the immediate postinduction interval, markedly evident at DOX day 3, but nearly undetectable by DOX day 14 (Fig. 3A, Left and Fig. S2B). Sprouts were not detectable in NTG mice at any interval during DOX treatment (Fig. 3A, Left and Fig. S2B). Because Dll4-Notch1 activation regulates vascular sprouting, and is downstream of VEGF signaling via VEGFR2 and VEGFR3 (1921), we interrogated mouse ear cross-sections for dual Dll4/CD31 coimmunofluorescence (Fig. 3A, Right). Endothelial Dll4 expression levels in TetON-HIF-1:VEGF mice were low and similar to those of their NTG counterparts up to 30 d of continuous HIF-1 induction (Fig. 3A, Right). In contrast, TetON-HIF-1:VEGF mice evidenced marked endothelial Dll4 up-regulation after 14 d of HIF-1 induction (Fig. 3, Right Middle). We further explored Notch-1 signaling by determining the expression kinetics and localization of the Notch intracellular domain (NICD), the product of the gamma secretase-mediated receptor cleavage and the active transcriptional Notch component (22). The NICD was detectable in both the day 14 TetON-HIF-1:VEGF endothelium and in basal keratinocytes (Fig. 3B, arrowheads). In contrast, the NICD was detectable only in a few basal keratinocytes of TetON-HIF-1:VEGF mice. The levels of Dll4 and the downstream Notch target Hey 1 mRNA were elevated 7- to 10-fold in TetON-HIF-1:VEGF mice compared with 14-d DOX-treated NTG controls (Fig. 3C). Dll4 and Hey-1 were induced at low, but statistically significant, levels in TetON-HIF-1:VEGF mice compared with NTG controls (Fig. 3C). Collectively, these data suggest that HIF-1 and its downstream targets (see below) are able to induce endothelial sprouting, yet these sprouts are nonproductive in forming neovessels in the absence of VEGF. The paucity of epidermal NICD in the TetON-HIF-1:VEGF mice demonstrates that VEGF is required for robust Notch activation in this cellular tissue compartment after HIF-1 induction.

An external file that holds a picture, illustration, etc.
Object name is pnas.1101321108fig03.jpg

Vessel sprouting increase with low-level increase of Notch activity in VEGF mice. (A) Representative confocal CD31 immunofluorescence showing vessel sprouting in TetON-HIF-1:VEGF and TetON-HIF-1VEGF mice (arrows; Left). Note multiple sprouts in VEGF mice at DOX day 14 compared with their absence in VEGF mice. Representative dual Dll4/CD31 immunofluorescence (Right). VEGF mice showed marked endothelial Dll4 upregulation from 3 d of HIF-1 induction compared to VEGF δ mice and NTG counterparts. Sprouts were not detectable in DOX-treated NTG mice (Left). (B) Representative immunofluorescence of cleaved Notch1 intracellular domain (NICD, Val1744) in TetON-HIF-1:VEGF and TetON-HIF-1VEGF mice (arrows; Middle and Bottom). (C) RT-PCR analysis (arbitrary units normalized to histone 3.3A) of the Notch1 ligand Dll4 and downstream target Hey1 showing marked 7- to 10-fold elevation in day 14 VEGF mice (n = 3–4 mice per time point). In contrast, Dll4 and Hey-1 were induced at low, albeit statistically significant levels in VEGF mice. Data are mean ± SD. Statistical analysis was the unpaired Student t test comparing each VEGF or VEGF DOX treatment day to comparably treated NTG controls: *P < 0.05, **P < 0.01 and ***P < 0.001. (Scale bars: A, Left, 20 μm; A, Right, 200 μm; B, 50 μm.)

Induction of Multiple HIF-1 Angiogenic Target Genes Is Insufficient for Angiogenesis in the Absence of VEGF.

Previous work has highlighted the angiogenic potential of several direct and indirect HIF-1 transcriptional targets (2329). To determine whether these targets were induced by HIF-1 in the absence of VEGF, whole skin extracts were analyzed by using RT-PCR, ELISA, or antibody microarrays (Fig. 4 A and E). First, we determined the functional efficiency of keratin-14 regulated Cre-mediated VEGF deletion. Both mRNA and whole tissue ELISA revealed baseline or even lower VEGF levels, compared with DOX-treated NTG controls, despite HIF-1 induction.

An external file that holds a picture, illustration, etc.
Object name is pnas.1101321108fig04.jpg

Induction of multiple HIF-1 angiogenic targets in TetON-HIF-1:VEGF mice. (A–D) RT-PCR determination (arbitrary unit normalized against histone 3.3A) of differential patterns of HIF-1 target gene mRNA expression. Adrenomedullin (ADM) and iNOS increased 2.4-fold by day 3 in VEGF mice, but further increased by sevenfold during continuous HIF-1 induction for 14 and 30 d, yet did not reach the induction levels of VEGF positive controls. Similarly, carbonic anhydrase IX (CAIX) progressively increased in VEGF mice to levels comparable to TetON-HIF-1:VEGF positive controls by day 30 (n = 3–4 mice per time point). (E) Whole tissue ELISA revealed baseline or lower VEGF levels in VEGF mice, compared with DOX-treated NTG controls, during HIF-1 induction (n = 3 mice per time point). (F) PIGF was up-regulated in VEGF mice at similar levels as VEGF controls (n = 3 mice per time point). Data analyzed as in Fig. 1: *P < 0.05, **P < 0.01, and ***P < 0.001.

Other targets exhibited four expression patterns. PIGF, angiogenin, MMP-9, PAI-1, and MMP-3 were up-regulated in TetON-HIF-1:VEGF mice at levels comparable with TetON-HIF-1:VEGF controls (Fig. 4F and Fig. S3 A and B). Adrenomedullin (ADM) and iNOS expression were lower in TetON-HIF-1:VEGF mice on DOX day 3, increased during continuous HIF-1 activation for 14 and 30 d, yet did not reach the levels evident in TetON-HIF-1:VEGF positive controls (Fig. 4 B and C). Carbonic anhydrase IX (CAIX) progressively increased in TetON-HIF-1:VEGF mice to levels comparable with TetON-HIF-1:VEGF positive controls by 30 d of continuous HIF-1 induction (Fig. 4D). The fourth expression pattern tracked with VEGF expression, lack or markedly reduced induction in TetON-HIF-1:VEGF mice compared with marked up-regulation in TetON-HIF-1:VEGF counterparts. These molecules, osteopontin (OPN), pentraxin-3 (PTX-3), cysteine-rich 61/connective tissue growth factor/nephroblastoma overexpressed (CCN3/NOV), and macrophage chemoattractant protein-1 (MCP-1), are expressed in activated inflammatory and stromal fibroblasts (3032). Their expression pattern was consistent with the marked diminution in stromal myeloid cell recruitment and retention in TetON-HIF-1:VEGF compared with TetON-HIF-1:VEGF mice (see below).

Epithelial VEGF Loss Markedly Impairs HIF-1–Mediated Myeloid Cell Recruitment.

HIF-1 is known to induce myeloid cell mobilization, recruitment, and retention via VEGF, PlGF, and SDF1 (11, 3336). As such, we determined myeloid cell recruitment to HIF-1 stimulated skin in the absence of concomitant VEGF. Surprisingly, there was no significant recruitment of CD45+, CD11b+, or F4/80+ myeloid, or mast cells in TetON-HIF-1:VEGF mice after 14 d of HIF-1 induction despite significant up-regulation of the VEGFR1 ligand, PlGF (Figs. 4F and 5 AC and Fig. S4). By day 30, a significant increase in both CD45 and CD11b myeloid cells was achieved in TetON-HIF-1:VEGF mice; however, these myeloid cells were insufficient to facilitate neovascularization in the absence of epithelial VEGF (Fig. 5 AC and Fig. S4). Myeloid cell infiltration after chronic HIF-1 gain of function in DOX day 30 TetON-HIF-1:VEGF mice was similar to our previous work with germ-line skin-targeted constitutive HIF-1 mutants in transgenic mice (37). There, stromal myeloid cell trafficking was regulated by chemokines not VEGF.

An external file that holds a picture, illustration, etc.
Object name is pnas.1101321108fig05.jpg

HIF-1–mediated myeloid cell recruitment is abrogated in the absence of VEGF. (A) Representative CD31/CD11b coimmunofluorescence showing marked temporal delay and diminution of stromal myeloid cell infiltration in VEGF mice compared with early and robust CD11b+ myeloid cell recruitment inVEGF counterparts. (B and C) Quantification of stromal area occupied by CD11b+ (B) (n = 3–4 mice per time point) and CD45+ cells (C; see Fig. S5 for representative immunofluorescent images) (n = 3–6 mice per time point). Data analyzed as in Fig. 1: *P < 0.05, **P < 0.01, and ***P < 0.001. (Scale bar: 200 μm.)

12-O-Tetradecanoylphorbol-13-Acetate (TPA)-Induced Inflammation Produces Extensive Vascular Remodeling with Minimal Microvascular Density Increase.

Topical phorbol ester induces angiogenesis via enhanced VEGF secretion from the activated epithelium (38, 39). Single-dose TPA treatment produces transient skin changes resolving after 3–4 d in wild type mice. Previously, we discovered that germ-line K14-HIF-1 transgenic mice responded to single-dose TPA challenge with a marked and prolonged stromal and intraepithelial neutrophil infiltrate persisting for 3 wk (37). Neutrophil recruitment was secondary to HIF-1–enhanced NFκB signaling and NFκB chemokine target gene up-regulation by transgenic keratinocytes. We capitalized on this strategy to determine whether the lack of HIF-1–mediated neovascularization in TetON-HIF-1:VEGF vasculature could be rescued. TetON-HIF-1:VEGF and TetON-HIF-1:VEGF were DOX-induced for 14 d to prime the keratinocytes and stroma and emulate germ-line constitutive activation. One dose of topical TPA was followed by ear harvest 10 d later. As reported, TPA treatment produced a marked recruitment and retention of CD45 stromal myeloid cells in TetON-HIF-1:VEGF mice (37), without affecting the frequency of CD11b macrophages (Fig. S5 A and B). Both TPA-treated DOX day 14 TetON-HIF-1:VEGF and TetON-HIF-1:VEGF mice ear epidermis was similarly punctuated by microabscesses filled with neutrophils (Fig. S6, Right), an observation consistent with our previous findings (37). Moreover, TPA did not further augment VEGF expression or neovascularization in TetON-HIF-1:VEGF mice (Fig. 6 A and B). In contrast, single-dose TPA in TetON-HIF-1:VEGF mice produced a twofold increase in VEGF expression associated with a similar fold increase in neovessel area. TPA also produced a similar influx of neutrophils in TetON-HIF-1:VEGF mice (Fig. S5A, Left and B, Upper). Partial rescue of HIF-1–mediated neovascularization in TetON-HIF-1:VEGF mice was associated with expansion of an epidermal population of unrecombined VEGF alleles and abscess-derived neutrophils (Fig. S5B). However, VEGF expression was undetectable in the majority of stromal neutrophils not associated with intraepidermal abcesses. TPA treatment also produced a statistically significant increase in capillary and total vessel volume, and a trend towards an increase in vessel tortuosity, as determined by OR-PAM (Fig. S7 A and B).

An external file that holds a picture, illustration, etc.
Object name is pnas.1101321108fig06.jpg

12-O-Tetradecanoylphorbol-13-acetate (TPA) treatment partially rescues neovascularization and results in moderate VEGF induction. (A) Quantification of CD31(+) area in (n = 3–6 mice per time point) revealed a twofold increase in neovascularization in TPA-treated VEGF mice. (B) Whole ear tissue ELISA for VEGF. A single dose of TPA in VEGF mice produced a twofold increase in VEGF. In contrast, VEGF levels were not further increased in TPA-treated VEGF mice. (n = 3–6 mice per time point). Data analyzed as in Fig. 1: *P < 0.05, **P < 0.01, and ***P < 0.001.

Mouse Development, Target Expression Analysis, Photoacoustic Microscopy, and Inflammation Challenge.

Detailed description of the mouse intercrosses to generate target and control genotypes, tissue molecular and expression analysis, photoacoustic microscopy, and inflammation challenge experiments are available in SI Materials and Methods. The Animal Studies Committee of Washington University in St. Louis approved all animal care and experimental procedures.

Statistical Analysis.

The data are reported as the mean ± SD. Data from DOX-treated TetON-HIF-1:VEGF or TetON-HIF-1VEGF mice were compared either with day 0 or NTG mice treated with DOX for the same interval by using the unpaired Student t test (GraphPad Prism 5). There were 3–6 mice per group unless otherwise indicated.

Supplementary Material

Supporting Information:
Urology Division, Department of Surgery, and
Department of Medicine, University of Hawaii, 96813; and
Department of Biomedical Engineering and
Siteman Cancer Center, Washington University in St. Louis, 63110
Corresponding author.
To whom correspondence may be addressed. E-mail: ude.ltsuw.sisoduw@jtiebra or ude.ltsuw.demoib@gnawhl.
Edited* by Gregg L. Semenza, The Johns Hopkins University School of Medicine, Baltimore, MD, and approved June 24, 2011 (received for review January 26, 2011)

Author contributions: S.O., S.H., K.M., L.V.W., and J.M.A. designed research; S.O., S.H., J.K., A.S., and R.E.S. performed research; R.S. and K.M. contributed new reagents/analytic tools; S.O., S.H., J.K., J.Y., L.V.W., and J.M.A. analyzed data; and S.O., S.H., J.Y., L.V.W., and J.M.A. wrote the paper.

S.O. and S.H. contributed equally to this work.
Edited* by Gregg L. Semenza, The Johns Hopkins University School of Medicine, Baltimore, MD, and approved June 24, 2011 (received for review January 26, 2011)

Abstract

Although our understanding of the molecular regulation of adult neovascularization has advanced tremendously, vascular-targeted therapies for tissue ischemia remain suboptimal. The master regulatory transcription factors of the hypoxia-inducible factor (HIF) family are attractive therapeutic targets because they coordinately up-regulate multiple genes controlling neovascularization. Here, we used an inducible model of epithelial HIF-1 activation, the TetON-HIF-1 mouse, to test the requirement for VEGF in HIF-1 mediated neovascularization. TetON-HIF-1, K14-Cre, and VEGF alleles were combined to create TetON-HIF-1:VEGF mice to activate HIF-1 and its target genes in adult basal keratinocytes in the absence of concomitant VEGF. HIF-1 induction failed to produce neovascularization in TetON-HIF-1:VEGF mice despite robust up-regulation of multiple proangiogenic HIF targets, including PlGF, adrenomedullin, angiogenin, and PAI-1. In contrast, endothelial sprouting was preserved, enhanced, and more persistent, consistent with marked reduction in Dll4-Notch-1 signaling. Optical-resolution photoacoustic microscopy, which provides noninvasive, label-free, high resolution, and wide-field vascular imaging, revealed the absence of both capillary expansion and arteriovenous remodeling in serially imaged individual TetON-HIF-1:VEGF mice. Impaired TetON-HIF-1:VEGF neovascularization could be partially rescued by 12-O-tetradecanoylphorbol-13-acetate skin treatment. These data suggest that therapeutic angiogenesis for ischemic cardiovascular disease may require treatment with both HIF-1 and VEGF.

Keywords: vascular biology, conditional expression, photoacoustic tomography, capillary imaging
Abstract

Neovascularization is crucial for solid tumor growth and metastatic spread, for wound healing, and for tissue preservation and recovery after ischemia. Although tremendous progress has been made in understanding the molecular circuits regulating neovascularization in both benign and malignant disease, our knowledge remains incomplete. Solid tumors evade angiogenesis inhibitors, and efficacious therapeutic angiogenesis for tissue ischemia remains a tantalizing prospect.

The hypoxia-inducible factors-1 and -2 (HIF-1 and HIF-2) are αβ heterodimeric proteins that make crucial contributions to neovascularization in wound healing, and benign or malignant diseases (1, 2). HIFs are master regulatory transcription factors with >100 known, and potentially hundreds more, target genes containing the core RCGTG enhancer sequence (36). A collection of HIF target genes coordinates neovascularization at several levels including endothelial cell proliferation, microvessel tone, vascular remodeling, and proangiogenic myeloid cell recruitment (7).

Numerous reports suggested that HIF produced a more robust neovasculature, with more normal structure and function, compared with single angiogenic factor overexpression (2, 810). However, the contribution of individual target genes encoding vascular functions to the collective HIF-1 neovascular phenotype is unknown. Here, we tested the hypothesis that VEGF was dispensable for HIF-mediated neovascularization. We used a unique conditional model of adult neovascularization in the skin, the TetON-HIF-1 transgenic mouse (11) that displays multistage neovascularization, and myeloid cell recruitment and retention in the absence of disease. TetON-HIF-1 mice were engineered for germ-line Cre-mediated VEGF deletion in the same basal keratinocytes targeted for conditional adult HIF-1α induction. VEGF deletion abrogated neovascularization despite coordinate up-regulation of multiple angiogenic growth factors. Surprisingly, endothelial sprouting was activated after HIF-1 induction; however, these sprouts were nonproductive for neovessel development. Despite robust PlGF, adrenomedullin, and PAI-1 induction, HIF-1 activation in the absence of VEGF failed to affect vascular remodeling, and myeloid cell recruitment was markedly impaired. Thus, we provide evidence that VEGF is required for HIF-1–mediated adult neovascularization, but that certain elements of the angiogenic response proceed without this factor.

Click here to view.

Acknowledgments

We thank Hans Peter Gerber and Napoleon Ferrara for the gift of the VEGF mice, Rick Bruick for the gift of the TRE-HIF-1α plasmid for construction of the TRE-HIF-1α transgenic mice, and Adam Glick for the gift of the K5-rtTA transgenic mice. This work was supported by National Institutes of Health Grants R01-CA90722, R01 EB000712, R01 NS46214, R01 EB008085, and U54 CA136398, and the Beatrice Roe Urologic Cancer Fund.

Acknowledgments

Footnotes

Conflict of interest statement: L.V.W. has financial interest in Microphotoacoustics, Inc., and Endra, Inc., which, however, did not support this work. Other authors declare no competing financial interest.

*This Direct Submission article had a prearranged editor.

This article contains supporting information online at www.pnas.org/lookup/suppl/doi:10.1073/pnas.1101321108/-/DCSupplemental.

Footnotes

References

  • 1. Hickey MM, Simon MCRegulation of angiogenesis by hypoxia and hypoxia-inducible factors. Curr Top Dev Biol. 2006;76:217–257.[PubMed][Google Scholar]
  • 2. Rey S, Semenza GLHypoxia-inducible factor-1-dependent mechanisms of vascularization and vascular remodelling. Cardiovasc Res. 2010;86:236–242.[Google Scholar]
  • 3. Wenger RH, Stiehl DP, Camenisch GIntegration of oxygen signaling at the consensus HRE. Sci STKE. 2005;2005:re12.[PubMed][Google Scholar]
  • 4. Ortiz-Barahona A, Villar D, Pescador N, Amigo J, del Peso LGenome-wide identification of hypoxia-inducible factor binding sites and target genes by a probabilistic model integrating transcription-profiling data and in silico binding site prediction. Nucleic Acids Res. 2010;38:2332–2345.[Google Scholar]
  • 5. Mole DR, et al Genome-wide association of hypoxia-inducible factor (HIF)-1alpha and HIF-2alpha DNA binding with expression profiling of hypoxia-inducible transcripts. J Biol Chem. 2009;284:16767–16775.[Google Scholar]
  • 6. Benita Y, et al An integrative genomics approach identifies Hypoxia Inducible Factor-1 (HIF-1)-target genes that form the core response to hypoxia. Nucleic Acids Res. 2009;37:4587–4602.[Google Scholar]
  • 7. Semenza GLVascular responses to hypoxia and ischemia. Arterioscler Thromb Vasc Biol. 2010;30:648.[Google Scholar]
  • 8. Kelly BD, et al Cell type-specific regulation of angiogenic growth factor gene expression and induction of angiogenesis in nonischemic tissue by a constitutively active form of hypoxia-inducible factor 1. Circ Res. 2003;93:1074–1081.[PubMed][Google Scholar]
  • 9. Hirota K, Semenza GLRegulation of angiogenesis by hypoxia-inducible factor 1. Crit Rev Oncol Hematol. 2006;59:15–26.[PubMed][Google Scholar]
  • 10. Elson DA, et al Induction of hypervascularity without leakage or inflammation in transgenic mice overexpressing hypoxia-inducible factor-1alpha. Genes Dev. 2001;15:2520–2532.[Google Scholar]
  • 11. Oladipupo SS, et al Conditional HIF-1 induction produces multistage neovascularization with stage-specific sensitivity to VEGFR inhibitors and myeloid cell independence. Blood. 2011;117:4142–4153.[Google Scholar]
  • 12. Bekeredjian R, et al Conditional HIF-1alpha expression produces a reversible cardiomyopathy. PLoS ONE. 2010;5:e11693.[Google Scholar]
  • 13. Diamond I, Owolabi T, Marco M, Lam C, Glick AConditional gene expression in the epidermis of transgenic mice using the tetracycline-regulated transactivators tTA and rTA linked to the keratin 5 promoter. J Invest Dermatol. 2000;115:788–794.[PubMed][Google Scholar]
  • 14. Jonkers J, et al Synergistic tumor suppressor activity of BRCA2 and p53 in a conditional mouse model for breast cancer. Nat Genet. 2001;29:418–425.[PubMed][Google Scholar]
  • 15. Gerber HP, et al VEGF is required for growth and survival in neonatal mice. Development. 1999;126:1149–1159.[PubMed][Google Scholar]
  • 16. Rossiter H, et al Loss of vascular endothelial growth factor a activity in murine epidermal keratinocytes delays wound healing and inhibits tumor formation. Cancer Res. 2004;64:3508–3516.[PubMed][Google Scholar]
  • 17. Wang LVMultiscale photoacoustic microscopy and computed tomography. Nat Photonics. 2009;3:503–509.[Google Scholar]
  • 18. Maslov K, Zhang HF, Hu S, Wang LVOptical-resolution photoacoustic microscopy for in vivo imaging of single capillaries. Opt Lett. 2008;33:929–931.[PubMed][Google Scholar]
  • 19. Tammela T, et al Blocking VEGFR-3 suppresses angiogenic sprouting and vascular network formation. Nature. 2008;454:656–660.[PubMed][Google Scholar]
  • 20. Thurston G, Noguera-Troise I, Yancopoulos GDThe Delta paradox: DLL4 blockade leads to more tumour vessels but less tumour growth. Nat Rev Cancer. 2007;7:327–331.[PubMed][Google Scholar]
  • 21. Hellström M, et al Dll4 signalling through Notch1 regulates formation of tip cells during angiogenesis. Nature. 2007;445:776–780.[PubMed][Google Scholar]
  • 22. Kopan R, Ilagan MXGThe canonical Notch signaling pathway: Unfolding the activation mechanism. Cell. 2009;137:216–233.[Google Scholar]
  • 23. Odorisio T, et al Mice overexpressing placenta growth factor exhibit increased vascularization and vessel permeability. J Cell Sci. 2002;115:2559–2567.[PubMed][Google Scholar]
  • 24. Luttun A, et al Revascularization of ischemic tissues by PlGF treatment, and inhibition of tumor angiogenesis, arthritis and atherosclerosis by anti-Flt1. Nat Med. 2002;8:831–840.[PubMed][Google Scholar]
  • 25. Roy H, et al Adenovirus-mediated gene transfer of placental growth factor to perivascular tissue induces angiogenesis via upregulation of the expression of endogenous vascular endothelial growth factor-A. Hum Gene Ther. 2005;16:1422–1428.[PubMed][Google Scholar]
  • 26. Abe M, et al Adrenomedullin augments collateral development in response to acute ischemia. Biochem Biophys Res Commun. 2003;306:10–15.[PubMed][Google Scholar]
  • 27. Oehler MK, Hague S, Rees MC, Bicknell RAdrenomedullin promotes formation of xenografted endometrial tumors by stimulation of autocrine growth and angiogenesis. Oncogene. 2002;21:2815–2821.[PubMed][Google Scholar]
  • 28. Raghu H, et al Suppression of uPA and uPAR attenuates angiogenin mediated angiogenesis in endothelial and glioblastoma cell lines. PLoS ONE. 2010;5:e12458.[Google Scholar]
  • 29. Yoshioka N, Wang L, Kishimoto K, Tsuji T, Hu GFA therapeutic target for prostate cancer based on angiogenin-stimulated angiogenesis and cancer cell proliferation. Proc Natl Acad Sci USA. 2006;103:14519–14524.[Google Scholar]
  • 30. Mantovani A, Garlanda C, Doni A, Bottazzi BPentraxins in innate immunity: From C-reactive protein to the long pentraxin PTX3. J Clin Immunol. 2008;28:1–13.[PubMed][Google Scholar]
  • 31. Keeley EC, Mehrad B, Strieter RMChemokines as mediators of neovascularization. Arterioscler Thromb Vasc Biol. 2008;28:1928–1936.[Google Scholar]
  • 32. Scatena M, Liaw L, Giachelli CMOsteopontin: A multifunctional molecule regulating chronic inflammation and vascular disease. Arterioscler Thromb Vasc Biol. 2007;27:2302–2309.[PubMed][Google Scholar]
  • 33. Ceradini DJ, et al Progenitor cell trafficking is regulated by hypoxic gradients through HIF-1 induction of SDF-1. Nat Med. 2004;10:858–864.[PubMed][Google Scholar]
  • 34. Grunewald M, et al VEGF-induced adult neovascularization: Recruitment, retention, and role of accessory cells. Cell. 2006;124:175–189.[PubMed][Google Scholar]
  • 35. Hattori K, et al Placental growth factor reconstitutes hematopoiesis by recruiting VEGFR1(+) stem cells from bone-marrow microenvironment. Nat Med. 2002;8:841–849.[Google Scholar]
  • 36. Fischer C, et al Anti-PlGF inhibits growth of VEGF(R)-inhibitor-resistant tumors without affecting healthy vessels. Cell. 2007;131:463–475.[PubMed][Google Scholar]
  • 37. Scortegagna M, et al HIF-1alpha regulates epithelial inflammation by cell autonomous NFkappaB activation and paracrine stromal remodeling. Blood. 2008;111:3343–3354.[Google Scholar]
  • 38. Kishimoto J, et al In vivo detection of human vascular endothelial growth factor promoter activity in transgenic mouse skin. Am J Pathol. 2000;157:103–110.[Google Scholar]
  • 39. Morris PB, Hida T, Blackshear PJ, Klintworth GK, Swain JLTumor-promoting phorbol esters induce angiogenesis in vivo. Am J Physiol. 1988;254:C318–C322.[PubMed][Google Scholar]
  • 40. Nagy JA, Shih SC, Wong WH, Dvorak AM, Dvorak HF. Chapter 3. The adenoviral vector angiogenesis/lymphangiogenesis assay. Methods Enzymol. 2008;444:43–64.[PubMed]
  • 41. Hanahan D, Folkman JPatterns and emerging mechanisms of the angiogenic switch during tumorigenesis. Cell. 1996;86:353–364.[PubMed][Google Scholar]
  • 42. Park JE, Chen HH, Winer J, Houck KA, Ferrara N. Placenta growth factor. Potentiation of vascular endothelial growth factor bioactivity, in vitro and in vivo, and high affinity binding to Flt-1 but not to Flk-1/KDR. J Biol Chem. 1994;269:25646–25654.[PubMed]
  • 43. Landgren E, Schiller P, Cao Y, Claesson-Welsh LPlacenta growth factor stimulates MAP kinase and mitogenicity but not phospholipase C-gamma and migration of endothelial cells expressing Flt 1. Oncogene. 1998;16:359–367.[PubMed][Google Scholar]
  • 44. Eriksson A, et al Placenta growth factor-1 antagonizes VEGF-induced angiogenesis and tumor growth by the formation of functionally inactive PlGF-1/VEGF heterodimers. Cancer Cell. 2002;1:99–108.[PubMed][Google Scholar]
  • 45. Autiero M, et al Role of PlGF in the intra- and intermolecular cross talk between the VEGF receptors Flt1 and Flk1. Nat Med. 2003;9:936–943.[PubMed][Google Scholar]
  • 46. Pipp F, et al VEGFR-1-selective VEGF homologue PlGF is arteriogenic: Evidence for a monocyte-mediated mechanism. Circ Res. 2003;92:378–385.[PubMed][Google Scholar]
  • 47. Van de Veire S, et al Further pharmacological and genetic evidence for the efficacy of PlGF inhibition in cancer and eye disease. Cell. 2010;141:178–190.[PubMed][Google Scholar]
  • 48. Bais C, et al PlGF blockade does not inhibit angiogenesis during primary tumor growth. Cell. 2010;141:166–177.[PubMed][Google Scholar]
  • 49. Nikitenko LL, et al Adrenomedullin and CGRP interact with endogenous calcitonin-receptor-like receptor in endothelial cells and induce its desensitisation by different mechanisms. J Cell Sci. 2006;119:910–922.[PubMed][Google Scholar]
  • 50. Ishikawa T, et al Adrenomedullin antagonist suppresses in vivo growth of human pancreatic cancer cells in SCID mice by suppressing angiogenesis. Oncogene. 2003;22:1238–1242.[PubMed][Google Scholar]
  • 51. Shindo T, et al Vascular abnormalities and elevated blood pressure in mice lacking adrenomedullin gene. Circulation. 2001;104:1964–1971.[PubMed][Google Scholar]
  • 52. Nikitenko LL, Smith DM, Bicknell R, Rees MCTranscriptional regulation of the CRLR gene in human microvascular endothelial cells by hypoxia. FASEB J. 2003;17:1499–1501.[PubMed][Google Scholar]
  • 53. Tsuzuki Y, et al Vascular endothelial growth factor (VEGF) modulation by targeting hypoxia-inducible factor-1alpha—> hypoxia response element—> VEGF cascade differentially regulates vascular response and growth rate in tumors. Cancer Res. 2000;60:6248–6252.[PubMed][Google Scholar]
  • 54. Casanovas O, Hicklin DJ, Bergers G, Hanahan DDrug resistance by evasion of antiangiogenic targeting of VEGF signaling in late-stage pancreatic islet tumors. Cancer Cell. 2005;8:299–309.[PubMed][Google Scholar]
  • 55. Bosch-Marce M, et al Effects of aging and hypoxia-inducible factor-1 activity on angiogenic cell mobilization and recovery of perfusion after limb ischemia. Circ Res. 2007;101:1310–1318.[PubMed][Google Scholar]
  • 56. Rey S, et al Synergistic effect of HIF-1alpha gene therapy and HIF-1-activated bone marrow-derived angiogenic cells in a mouse model of limb ischemia. Proc Natl Acad Sci USA. 2009;106:20399–20404.[Google Scholar]
  • 57. Sadoun E, Reed MJImpaired angiogenesis in aging is associated with alterations in vessel density, matrix composition, inflammatory response, and growth factor expression. J Histochem Cytochem. 2003;51:1119–1130.[PubMed][Google Scholar]
  • 58. Iemitsu M, Maeda S, Jesmin S, Otsuki T, Miyauchi TExercise training improves aging-induced downregulation of VEGF angiogenic signaling cascade in hearts. Am J Physiol Heart Circ Physiol. 2006;291:H1290–H1298.[PubMed][Google Scholar]
Collaboration tool especially designed for Life Science professionals.Drag-and-drop any entity to your messages.