Skip main navigation

Mice Lacking the Vascular Endothelial Growth Factor-B Gene (Vegfb) Have Smaller Hearts, Dysfunctional Coronary Vasculature, and Impaired Recovery From Cardiac Ischemia

Originally publishedhttps://doi.org/10.1161/01.RES.86.2.e29Circulation Research. 2000;86:e29–e35

    Abstract

    Abstract—Vascular endothelial growth factor-B (VEGF-B) is closely related to VEGF-A, an effector of blood vessel growth during development and disease and a strong candidate for angiogenic therapies. To further study the in vivo function of VEGF-B, we have generated Vegfb knockout mice (Vegfb−/−). Unlike Vegfa knockout mice, which die during embryogenesis, Vegfb−/− mice are healthy and fertile. Despite appearing overtly normal, Vegfb−/− hearts are reduced in size and display vascular dysfunction after coronary occlusion and impaired recovery from experimentally induced myocardial ischemia. These findings reveal a role for VEGF-B in the development or function of coronary vasculature and suggest potential clinical use in therapeutic angiogenesis. The full text of this article is available at http://www.circresaha.org.

    Vascular endothelial growth factor-B (VEGF-B)123 is a secreted growth factor that has strong sequence homology with VEGF-A, a primary regulator of angiogenesis in development, corpus luteum formation, wound healing, and cancer.4 VEGF-B can form stable heterodimers with VEGF-A3 and is generally coexpressed with VEGF-A.56 VEGF-B can bind to two of the VEGF-A receptors, VEGFR-17 and neuropilin-1,8 suggesting that it may regulate the bioavailability and/or action of VEGF-A.5 Although VEGF-B has been reported to behave as an endothelial cell mitogen,2 part of the mitogenic activity reported may be due to VEGF-B/VEGF-A heterodimers.5

    Several mouse models have been generated by gene knockout technology where the genes encoding Vegf-A or its receptors have been mutated. Both Vegfa−/− and Vegfa+/− mice are unable to survive to term due to a general impairment of blood vessel formation in the early embryo.910Vegfa120/120 mice, where only two of the three major Vegf-A isoforms have been knocked out, die postnatally after cardiac failure due to widespread myocardial ischemia.11Vegfr1−/− mice die as embryos due to defects in angiogenesis,12 but partial knockout mice, where only the tyrosine kinase–encoding portion of the Vegfr1 gene is deleted, develop normal vasculature.13

    To study the in vivo role of Vegf-B, we have generated a knockout mouse line and found that, unlike the Vegf-A–related knockouts, Vegfb−/− mice appear outwardly normal and fertile. Because Vegfb transcripts are expressed predominantly in the heart during murine embryogenesis and adult life,1141516 suggesting a specific role for Vegf-B during cardiac development, we have concentrated on studying the cardiac phenotype in these mice. Vegfb−/− hearts are reduced in size compared with hearts of Vegfb+/+ littermates and display clinical symptoms of impaired recovery from experimentally induced ischemia. The results suggest an essential role for Vegf-B in establishment of a fully functional coronary vasculature and highlight the potential of this cytokine for application in the emerging field of therapeutic angiogenesis.

    Materials and Methods

    Generation of Vegfb+/− Mice

    All mice used for the present study were supplied by the Animal Resources Centre (Western Australia), and their treatment was in accordance with the National Health and Medical Research Council (NH&MRC) guidelines for the care of experimental animals.

    Targeted inactivation of the Vegfb gene was achieved by replacing exons 3 to 7 (Figure 1a) with a promoter-less β-geo cassette. The β-geo gene was preceded by an internal ribosomal entry site to give cap-independent translation of β-geo.17 Targeted 129/SvJ ES cells (C1368) were injected into C57BL/6J blastocysts to produce chimeras. Progeny of germline-transmitting chimeras were genotyped by polymerase chain reaction (PCR) amplification of tail-tip DNA using PCR1, 5′-TTT GAT GGC CCC AGC CAC-3′; PCR2, 5′-CCC CCA GCT GAC TGC TCG-3′; and PCR3, 5′-CTA GTG GAT CCC CCG GGC-3′ (Figure 1b).

    β-Gal Staining and Immunohistochemistry

    Frozen sections and whole embryos were stained for β-Gal as described.18 For quantitation of capillary density, transverse sections of the left ventricle (LV) were cut at comparable levels in Vegfb+/+ and Vegfb−/− P30 hearts (30 days postpartum) (4 hearts each), immunostained with anti–PECAM-1 (clone M13, Pharmingen), and the capillaries counted using ImagePlus software on 7 randomly chosen fields (×40 magnification, ≈0.06 mm2 per field) in the epicardial, endocardial, and midmyocardial portion of the LV. Because no difference between genotypes was found within each portion, capillary density data were averaged for each heart. Coronary vessels were counted as anti–smooth muscle α-actin (FITC conjugated, clone 1A4, Sigma)–stained vessels in whole sections.

    Heart Weight and LV Thickness

    Vegfb+/+, Vegfb+/−, and Vegfb−/− mice of either 129/SvJ or C57BL/6J×129/SvJ background were weighed. After dissection, the hearts were trimmed of surrounding tissue and weighed. A subset of the P25 hearts was fixed in formalin and microdissected to obtain a similar angle of section. LV thickness was measured on sections with a stage micrometer (n=10 Vegfb+/+ hearts; n=16 Vegfb+/− hearts; n=14 Vegfb−/− hearts).

    Langendorff Perfusion

    Hearts were isolated from mice anesthetized with 60 mg/kg sodium pentobarbital. Vegfb+/+ (161±7 mg wet heart weight [WHW], n=15), Vegfb+/− (152±6 mg WHW, n=14), and Vegfb−/− mice (155±7 mg WHW, n=16) hearts were perfused in the Langendorff mode as described.19

    For ischemia, baseline measurements were recorded from Vegfb+/+ (n=8), Vegfb+/− (n=8), and Vegfb−/− hearts (n=8) after 30 minutes of stabilization. Global normothermic ischemia was initiated for 20 minutes before 30 minutes of aerobic reperfusion. To examine reactive hyperemia, a subset of hearts (n=7 for Vegfb+/+, n=6 for Vegfb+/−, and n=8 for Vegfb−/−) was perfused as described above and after stabilization was subjected to a single 20-second period of zero flow followed by reperfusion at 90 mm Hg perfusion pressure. The coronary flow response was recorded, peak hyperemic flows were measured in individual experiments, and percentage of flow-debt repayment over the initial 60 seconds of reperfusion was calculated as follows:

    where total coronary flows were measured in mL/g and were calculated by digital integration of coronary flow for the 60 seconds before and 60 seconds after occlusion using the Chart V3.5.6 program (AD Instruments, Castle Hill, Australia), and flow-debt was calculated as basal coronary flow (mL/60 seconds/g)×20 seconds of occlusion.

    Statistical Analyses

    Body/heart weight, LV thickness, and capillary density data were analyzed using unpaired Student’s t tests. Body/heart weight data were also analyzed using the generalized estimation equation.20 Hyperemia data were analyzed via one-way ANOVA and functional parameters by two-way ANOVA for repeated measures. Where significant effects were detected, the Tukey’s HSD post hoc test was used. In all tests, significance was accepted at P<0.05.

    An expanded Materials and Methods section is available online at http://www.circresaha.org.

    Results

    Generation of the Vegfb−/− Mouse

    The Vegfb knockout mice generated with the modified Vegfb locus shown in Figure 1a were produced in normal mendelian ratios, were healthy and fertile, and did not display any overt phenotype. The genotype of mice was determined by PCR amplification of tail-tip DNA from P10 pups (Figure 1b). Rather than producing Vegf-B, this modified locus results in β-Gal expression under the control of the endogenous Vegfb promoter (herein referred to as Vegf-B/β-Gal). Because the Vegfb+/− mice generated in this manner had no obvious developmental defects, we assumed that Vegf-B/β-Gal expression in these mice accurately reflects the endogenous Vegfb expression pattern.

    Cardiac Vegf-B/β-Gal Expression Pattern in the Vegfb+/− Mouse

    Using β-Gal staining, Vegf-B/β-Gal expression was first detected in the heart at E10.5 (embryonic day 10.5), it became prominent at E12.5 (Figure 2a) and further increased thereafter (Figure 2b). Throughout development, Vegf-B/β-Gal expression appeared to be restricted to the myocardium (Figures 2c through 2g) and subepicardium (Figures 2g and 2h) and remained undetectable in endothelial cells, including those of the endocardium and coronary endothelium. Endocardial derivatives, such as the valve leaflets were always devoid of Vegf-B/β-Gal expression. During development, the highest concentration of Vegf-B/β-Gal–expressing cells was seen in the right ventricular myocardium and right aspect of the interventricular septum (Figures 2a through 2c). Lower Vegf-B/β-Gal expression was detectable in the LV (Figures 2a and 2c) and the right atrial appendage (Figure 2d). The lowest expression was found in the atrial wall (Figures 2c, 2d, and 2f), where coronary angiogenesis is less conspicuous (Figures 2e and 2f). Within the right ventricle (RV), Vegf-B/β-Gal staining was prevalent in the trabeculations (Figure 2d). The intensity of Vegf-B/β-Gal expression increased further in the neonate heart (Figures 2i and 2j) in correlation with the massive early postnatal coronary capillary and vessel growth.11 The prevalence of Vegf-B/β-Gal expression switches from the RV to the LV in the early neonatal period (Figures 2i and 2j) reflects the predominant early postnatal capillarization of this chamber.21 In the juvenile heart, the ventricular prevalence of expression is lost, and the density of Vegf-B/β-Gal–expressing cells is similar in the ventricles and the atria (data not shown).

    The great arteries in the heart expressed low levels of Vegf-B/β-Gal at all stages of development (eg, Figure 2b) and in juvenile mice (data not shown). Vegf-B/β-Gal was undetectable in the tunica intima and media of coronary vessels (Figures 2j, arrowheads, and 2k and 2l), although we found Vegf-B/β-Gal expression in other vessels in the body (eg, the intralobar component of the pulmonary arteries) (data not shown).

    Postnatal Heart Growth in Vegfb−/− Mice

    Although histological examination of all organs revealed no differences between genotypes, Vegfb−/− hearts frequently appeared marginally smaller than their Vegfb+/+ and Vegfb+/− littermates (Figure 3a). We recorded the total body and heart weight of 122 animals including male and female Vegfb+/+, Vegfb+/−, and Vegfb−/− mice at several ages between P3 and P91. These mice were grouped as P3 to P9 mice (Vegfb+/+, n=15; Vegfb+/−, n=20; and Vegfb−/−, n=13) and P25 or older (Vegfb+/+, n=20; Vegfb+/−, n=28; and Vegfb−/−, n=27). We found no consistent genotype-dependent decrease in body weigh; however, heart weight was always reduced in Vegfb−/− mice. To account for the inherent interlitter and intralitter variability in body weight, due to sex, age, and genetic background, we used heart/body ratios to display the results. Although we found no significant difference in heart/body ratio in relation to sex or genetic background, statistical analysis revealed a dramatic increase in heart/body ratio from P3-9 to P25 (or older) in all animals regardless of genotype (Figure 3b). There were no differences in percentage of heart/body weight ratios among genotypes in P3-9 mice (Vegfb+/+, 0.64±0.02; Vegfb+/−, 0.59±0.03; and Vegfb−/−, 0.66±0.03), but we found a significant (P<0.05) decrease in percentage of heart/body weight ratio in P25 (or older) Vegfb−/− (0.78±0.02) mice compared with Vegfb+/+ (0.87±0.04) and Vegfb+/− (0.89±0.02) mice (Figure 3b). When familial (litter/parents) correlation among mice was taken into account, this significant difference remained (data not shown). No significant difference was found between Vegfb+/+ and Vegfb+/− mice at any stage.

    The dramatic increase in heart weight during the first few weeks after birth is well documented and appears to be mainly due to massive growth of the coronary capillaries and vessels, but cardiomyocyte proliferation and hypertrophy also contribute.22 No difference in the size of myocardial cells was found in histological sections in Vegfb+/+, Vegfb+/−, or Vegfb−/− hearts (data not shown). LV thickness was significantly decreased in P25 Vegfb−/− (0.80±0.03 mm,* n=14) compared with Vegfb+/+ (0.89±0.03 mm, n=10) and Vegfb+/− (0.91±0.02 mm, n=16) hearts (*P=0.059 versus Vegfb+/+ and P<0.05 versus Vegfb+/−). Analysis of capillary density using standard morphometric measures found no significant differences between P30 Vegfb+/+ (2321±255 capillaries/mm2) and Vegfb−/− (2334±253 capillaries/mm2) hearts. Vessel density measures in adjacent heart sections also showed no differences between Vegfb+/+ (270±10 vessels/section) and Vegfb−/− (275±14 vessels/section) hearts.

    Reactive Hyperemic Responses in Vegfb+/+, Vegfb+/−, and Vegfb−/− Hearts

    Baseline contractile function and coronary flow were equivalent in Langendorff-perfused hearts from all three groups under normoxic conditions (see Table online, http://www.circresaha.org). To test whether alterations in vascular function would be more evident during active responses to modified myocardial O2 delivery, we exposed hearts to transient (20 seconds) coronary occlusion and studied the hyperemic response on reperfusion. The reactive hyperemic responses differed subtly between groups (Figure 4). Although peak hyperemic flow was comparable in all three groups of hearts (32 to 36 mL · min−1 · g−1) (Figure 4a), overall flow-debt repayment during the initial 60 seconds of reperfusion (during which flow recovered to preocclusion levels) was significantly lower in Vegfb−/− mice (≈60%) versus the other two groups (≈100%) (Figure 4b). There were no differences in repayment between Vegfb+/+ and Vegfb+/− hearts. These findings indicate that the functional status of the coronary vasculature is impaired in Vegfb−/− mice.

    Responses to Ischemia-Reperfusion in Vegfb+/+, Vegfb+/−, and Vegfb−/− Hearts

    As noted, baseline functional parameters were comparable in hearts from Vegfb+/+, Vegfb+/−, and Vegfb−/− mice (see Table online, http://www.circresaha.org). Global normothermic ischemia completely abolished contractile function in all hearts within 2 to 3 minutes and caused a rapid rise in diastolic pressure. Time to onset of contracture and peak-developed contracture are indicators of the severity of ischemic injury. Although no difference was found in the rate of contracture development, peak contracture during ischemia was greater in Vegfb−/− compared with Vegfb+/+ and Vegfb+/− hearts (Figure 5a). Diastolic pressure was significantly elevated in Vegfb−/− hearts compared with Vegfb+/+ and Vegfb+/− hearts during reperfusion and recovered minimally (≈73 mm Hg) relative to the other two groups (≈35 mm Hg) (Figure 5a). Recovery of contractile function was slightly depressed throughout reperfusion in Vegfb−/− hearts, with the rate-pressure product being significantly lower at 30 minutes compared with Vegfb+/+ and Vegfb+/− hearts (Figure 5b). Coronary flow responses did not differ between the three groups at any time point (Figure 5c).

    Discussion

    In the present study, we have used promoter trap LacZ expression in the Vegfb+/− mouse to correlate Vegf-B/β-Gal expression to processes of vascularization in the heart, the major site of Vegf-B expression. The general developmental pattern of Vegf-B expression has previously been interpreted to reflect a paracrine action of Vegf-B on the developing vasculature, although no attempts were made to correlate expression patterns with developmental processes of vascularization in any individual organ.16 Although Vegfb transcripts are produced in the heart from E8.5, Vegf-B protein expression is not detected until E10.5.16 We can first detect Vegf-B/β-Gal at E10.5 in the interventricular septum and propose that Vegf-B/β-Gal (and therefore Vegf-B) production increases substantially at this time point, in spatial and temporal correlation with the commencement of coronary endothelial growth in the heart.23 Indeed, Vegf-B/β-Gal levels increase both throughout development and after birth, closely correlating with the progression of cardiac angiogenesis. Vegf-B/β-Gal is conspicuous in the subepicardium where heart angiogenesis has been shown to commence.24 It is more densely expressed in the ventricles than the atria and correlates with the degree of coronary angiogenesis that is more advanced in the ventricles than in the atria at fetal stages. After birth, Vegf-B/β-Gal expression increases further in the LV, at a time when substantial capillary growth occurs predominantly in this chamber.2122 The disparity between LV and RV capillary growth rates decreases several days after birth.21 Accordingly, by P25, Vegf-B/β-Gal expression becomes even throughout the heart with levels of expression similar in RV, LV, and the atria. Despite the correlation between capillarization and Vegf-B/β-Gal expression, such expression was not found in the smooth muscle cells of the differentiated coronary vessels. It is therefore likely that Vegf-B may exert its paracrine action on the microvasculature surrounding the expressing myocardium, rather than on the endothelium of the coronary vessels.

    We find that Vegfb−/− hearts appear morphologically and functionally normal in the unstressed animal but do not undergo the same extent of postnatal growth as those of Vegfb+/+ and Vegfb+/− animals. Postnatal heart growth appears to be mainly due to the substantial increase in the coronary microvasculature and vessels.1122 This increase has been attributed to the action of Vegf-A164 and Vegf-A188,11 because mice lacking these Vegf-A isoforms die as a consequence of severe heart ischemia due to an almost total absence of postnatal capillary and coronary vessel growth. Postnatal ablation of Vegf-A (and possibly Vegf-B) function by administering a soluble Flt-1 receptor (mFlt(1–3)-IgG)25 is also lethal. In the heart, this treatment leads to cardiomyocyte necrosis and massive capillary and vessel density reduction.25 Because Vegf-B is coexpressed with Vegf-A in the myocardium of the ventricles,6 can form biologically active heterodimers with Vegf-A, and also binds Flt-1,7 it is likely that the abnormal coronary angiogenesis described above is a result of interference with the normal function of both Vegf-A and Vegf-B. We tested whether the observed Vegfb−/− reduction in heart weight was a consequence of impaired growth of the vascular network by measures of coronary capillary and vessel density. We found no significant differences between Vegfb−/− and Vegfb+/+ hearts, although additional studies measuring lumen size, patency, and permeability of capillaries and vessels in the heart will reveal whether any structural abnormalities in the vascular network of Vegfb−/− hearts may be responsible for reduced volume of this organ. Alternatively, the observed microcardia could be attributed to an effect of Vegf-B on cardiomyocyte growth. Vegf-B effect on heart muscle could be mediated by the Vegf-B167 (and Vegf-A165) receptor, neuropilin-1, which is expressed in the developing cardiac muscle. However, this is unlikely, because cardiomyocytes do not appear affected in size or function in the Vegfb−/− heart, and we cannot rule out a direct effect of Vegf-B ablation on myocytes. It is worth noting, nevertheless, that cardiomyocytes are normal in the Vegfa120/120 mouse, where neuropilin-1 ligand Vegf-A165 has been ablated.11 A slight decrease in left ventricular thickness in the Vegfb−/− heart may indicate that some developmental hypoplasia, resulting from suboptimal vascularization, could be responsible for the observed microcardia.

    Ablating Vegfb expression reduced the ability to repay coronary flow after a transient coronary occlusion. This occurred despite baseline coronary flow in the Vegfb−/− heart appearing normal, which was not unexpected given that moderate impairment of vascularization or vascular function that might result from deletion of the Vegfb gene could be compensated by enhanced intrinsic vasodilatation. Impairment of flow-debt repayment, despite similar peak flows, suggests inhibition of flow-mediated dilatation, which occurs subsequent to the immediate hyperemic response, indicating that the functional status of the coronary vasculature is impaired in some way in Vegfb−/− hearts. Reactive hyperemia is thought to be mediated by the combined actions of nitric oxide (NO) and adenosine,26 with potential involvement of KATP channels.27 The prolongation of the hyperemic response is thought to be at least partially NO dependent.28 Thus, one possible mechanism contributing to this change is an impaired NO production. However, deletion of the endothelial NO synthase gene fails to alter peak hyperemic flow, flow repayment, and adenosine responses in murine hearts.29

    Heart rate was almost identical in hearts of all genotypes before and after ischemia, and no significant differences existed for heart rate between any groups at any time. Interestingly, deletion of Vegfb reduced functional recovery from ischemia-reperfusion and appeared to worsen contracture during ischemia. The mechanism of contracture is not well understood but may involve rigor-bond formation as a result of impaired glycolytic ATP formation.30 During reperfusion, diastolic dysfunction was significantly greater in knockout mice; the difference was wholly due to a change in contractile force and not rate. Recovery of the rate-pressure product was slightly reduced whereas coronary flow was similar in all three groups. Although a reduced reflow or perfusion could have explained the dysfunction, this was not supported by the measures of total myocardial perfusion. However, this does not exclude a more subtle change in flow distribution that is not reflected in the total flow response. The postischemic elevation in diastolic pressure is likely to reflect altered Ca2+ handling in reperfused tissue, resulting in enhanced diastolic Ca2+ levels.31 Ca2+ handling is energy dependent, particularly at the level of the sarcoplasmic reticulum. Knockout of the Vegfb gene could conceivably lead to impaired postischemic recovery of energy metabolism, owing to maldistribution of coronary flow, such that myocardial handling of Ca2+ is impaired. Further experiments addressing patency, permeability, and responses to vasodilatory stimuli in the ventricular microvasculature of Vegfb−/− hearts will reveal whether this is indeed the case. The increased diastolic dysfunction during ischemia is largely independent of the coronary vasculature and may reflect a developmental effect of Vegfb deletion on heart growth or function, as suggested by the smaller hearts and reduced left ventricular thickness in Vegfb−/− mice.

    In the present study, we have shown that, despite heart morphology and function being normal in Vegfb−/− mice, the response to coronary occlusion and myocardial recovery from ischemia are compromised. Thus, although Vegf-B may play a redundant role in establishing the coronary vasculature, our results define a unique role in the development and maintenance of function in response to ischemic insult.

    
          Figure 1.

    Figure 1. a, Diagram of the murine Vegfb gene (top), the targeting construct used to generate a Vegfb+/− mouse (middle), and the final targeted locus (bottom). The exons of the Vegfb gene are shown as numbered boxes with the open reading frame as open boxes. The location and orientation of the PCR primers used to genotype mice are shown as PCR1, PCR2, and PCR3. b, Genotyping PCR on Vegfb+/+, Vegfb+/−, and Vegfb−/− DNA, using the primers shown in panel a.

    
          Figure 2.

    Figure 2. Vegf-B/β-Gal localization in the heart during development and after birth. a, Whole-mount E12.5 Vegf-B/β-Gal–stained Vegfb+/− (right) and Vegfb+/− (left) hearts showing higher expression in the RV. b through d, Whole mount and sections of Vegf-B/β-Gal–stained E15.5 hearts. b, Vegfb+/− whole mount. Vegf-B/β-Gal levels are very low in the aorta and pulmonary trunk. c, Longitudinal section of the Vegfb+/− heart shown in panel b. Note higher levels of Vegf-B/β-Gal expression (staining blue) in the RV and right side of the interventricular septum (*) than the LV. d, Sagittal section through the RV. Note expression in the trabeculations of the ventricle and absence in the atria. e through h, E17.5 hearts. e, Longitudinal heart section stained for PECAM-1 (staining brown). The rectangles represent the regions in panels f and g. f and g, Vegf-B/β-Gal and anti–PECAM-1 double labeling. f, Atrial region. The endocardium (arrows) is stained with anti–PECAM-1, but no capillaries have formed in this region. g, RV. Anti–PECAM-1 stains the endocardium (arrow) and the endothelium of numerous capillaries (arrowhead) surrounded by Vegf-B/β-Gal–expressing cardiomyocytes in the myocardium and subepicardium. h, Higher magnification of the subepicardial region stained for Vegf-B/β-Gal expression showing several Vegf-B/β-Gal–positive cells. i through k, P3 Vegfb+/− mouse hearts. i and j, Vegf-B/β-Gal staining of transverse heart sections. j, Section through the atrioventricular transition. Note prominent staining in the LV. j, Section through the ventricular region. Note the absence of staining around the coronary vessels (arrowheads). k and l, Contiguous sections through a coronary arteriole stained for PECAM-1 (j) and Vegf-B/β-Gal (k). Vegf-B/β-Gal–expressing cardiomyocytes surround the capillaries, but Vegf-B/β-Gal is undetectable in the endothelium and smooth muscle layers surrounding the artery. rv indicates right ventricle; lv, left ventricle; *, interventricular septum; ra, right atrium; aa, atrial appendage; m, myocardium; se, subepicardium; and p, pulmonary trunk. Bar=500 μm (a, b, d, e, i, and j) and 100 μm (c, f, g, h, k, and l).

    
          Figure 3.

    Figure 3. Reduced postnatal heart size in Vegfb−/− hearts. a, Appearance of P25 Vegfb−/−, Vegfb+/−, and Vegfb+/+ hearts from same-sex littermates, illustrating a slightly reduced Vegfb−/− heart size. b, Percentage of body/weight ratio is significantly reduced in the juvenile (>P25) Vegfb−/− mice but not in early postnatal mice (P3-9), indicating the impaired growth of the Vegfb−/− hearts in the first few weeks after birth. Values are mean±SEM. *P<0.05 vs Vegfb+/+ mice; P<0.01 vs Vegfb+/− mice.

    
          Figure 4.

    Figure 4. Reactive hyperemic responses to 20-second coronary occlusion in Langendorff-perfused Vegfb+/+, Vegfb+/−, and Vegfb−/− hearts. a, Baseline and peak hyperemic flows. b, Flow-debt repayments over the initial 60 seconds of reperfusion. Values are mean±SEM. *P<0.05 vs Vegfb+/+ hearts.

    
          Figure 5.

    Figure 5. Functional responses of Langendorff-perfused Vegfb+/+, Vegfb+/−, and Vegfb−/− hearts to 20 minutes of global normothermic ischemia and 30 minutes of reperfusion. a, Responses for left ventricular diastolic pressure. Rate-pressure product (heart rate×left ventricular developed pressure) (b) and coronary flow (c) are shown. Values are mean±SEM. *P<0.05 vs Vegfb+/+ hearts.

    This work was supported by the Queensland Cancer Fund and AMRAD Corporation Ltd. Nicholas K. Hayward is a recipient of a NH&MRC Senior Research Fellowship, and Sean M. Grimmond holds a NH&MRC C.J. Martin Traveling Fellowship. Graham F. Kay and Melissa Little are Fellows of the Sylvia and Charles Viertel Charitable Foundation. We would like to thank Michael Walsh for histological services and Dr Patrick Ward for advanced statistical analysis.

    Footnotes

    Correspondence to Graham Kay, Queensland Institute of Medical Research, Post Office Royal Brisbane Hospital, Queensland 4029, Australia. E-mail

    References

    • 1 Grimmond S, Lagercrantz J, Drinkwater C, Silins G, Townson S, Pollock P, Gotley D, Carson E, Rakar S, Nordenskjold M, Ward L, Hayward N, Weber G. Cloning and characterization of a novel human gene related to vascular endothelial growth factor. Genome Res.1996; 6:124–131.CrossrefMedlineGoogle Scholar
    • 2 Olofsson B, Pajusola K, Kaipainen A, von Euler G, Joukov V, Saksela O, Orpana A, Pettersson RF, Alitalo K, Eriksson U. Vascular endothelial growth factor B, a novel growth factor for endothelial cells. Proc Natl Acad Sci U S A.1996; 93:2576–2581.CrossrefMedlineGoogle Scholar
    • 3 Olofsson B, Pajusola K, von Euler G, Chilov D, Alitalo K, Eriksson U. Genomic organization of the mouse and human genes for vascular endothelial growth factor B (VEGF-B) and characterization of a second splice isoform. J Biol Chem.1996; 271:19310–19317.CrossrefMedlineGoogle Scholar
    • 4 Ferrara N. Vascular endothelial growth factor: molecular and biological aspects. Curr Top Microbiol Immunol.1999; 237:1–30.MedlineGoogle Scholar
    • 5 Eriksson U, Alitalo K. Structure, expression and receptor-binding properties of novel vascular endothelial growth factors. Curr Top Microbiol Immunol.1999; 237:41–57.MedlineGoogle Scholar
    • 6 Miquerol L, Gertsenstein M, Harpal K, Rossant J, Nagy A. Multiple developmental roles of VEGF suggested by a LacZ-tagged allele. Dev Biol.1999; 212:307–322.CrossrefMedlineGoogle Scholar
    • 7 Olofsson B, Korpelainen E, Pepper MS, Mandriota SJ, Aase K, Kumar V, Gunji Y, Jeltsch MM, Shibuya M, Alitalo K, Eriksson U. Vascular endothelial growth factor B (VEGF-B) binds to VEGF receptor-1 and regulates plasminogen activator activity in endothelial cells. Proc Natl Acad Sci U S A.1998; 95:11709–11714.CrossrefMedlineGoogle Scholar
    • 8 Makinen T, Olofsson B, Karpanen T, Hellman U, Soker S, Klagsbrun M, Eriksson U, Alitalo K. Differential binding of vascular endothelial growth factor B splice and proteolytic isoforms to neuropilin-1. J Biol Chem.1999; 274:21217–21222.CrossrefMedlineGoogle Scholar
    • 9 Ferrara N, Carver-Moore K, Chen H, Dowd M, Lu L, O’Shea KS, Powell-Braxton L, Hillan KJ, Moore MW. Heterozygous embryonic lethality induced by targeted inactivation of the VEGF gene. Nature.1996; 380:439–442.CrossrefMedlineGoogle Scholar
    • 10 Carmeliet P, Ferreira V, Breier G, Pollefeyt S, Kieckens L, Gertsenstein M, Fahrig M, Vandenhoeck A, Harpal K, Eberhardt C, Declercq C, Pawling J, Moons L, Collen D, Risau W, Nagy A. Abnormal blood vessel development and lethality in embryos lacking a single VEGF allele. Nature.1996; 380:435–439.CrossrefMedlineGoogle Scholar
    • 11 Carmeliet P, Ng YS, Nuyens D, Theilmeier G, Brusselmans K, Cornelissen I, Ehler E, Kakkar VV, Stalmans I, Mattot V, Perriard JC, Dewerchin M, Flameng W, Nagy A, Lupu F, Moons L, Collen D, D’Amore PA, Shima DT. Impaired myocardial angiogenesis and ischemic cardiomyopathy in mice lacking the vascular endothelial growth factor isoforms VEGF164 and VEGF188. Nat Med.1999; 5:495–502.CrossrefMedlineGoogle Scholar
    • 12 Fong GH, Rossant J, Gertsenstein M, Breitman ML. Role of the Flt-1 receptor tyrosine kinase in regulating the assembly of vascular endothelium. Nature.1995; 376:66–70.CrossrefMedlineGoogle Scholar
    • 13 Hiratsuka S, Minowa O, Kuno J, Noda T, Shibuya M. Flt-1 lacking the tyrosine kinase domain is sufficient for normal development and angiogenesis in mice. Proc Natl Acad Sci U S A.1998; 95:9349–9354.CrossrefMedlineGoogle Scholar
    • 14 Lagercrantz J, Larsson C, Grimmond S, Fredriksson M, Weber G, Piehl F. Expression of the VEGF-related factor gene in pre- and postnatal mouse. Biochem Biophys Res Commun.1996; 220:147–152.CrossrefMedlineGoogle Scholar
    • 15 Lagercrantz J, Farnebo F, Larsson C, Tvrdik T, Weber G, Piehl F. A comparative study of the expression patterns for vegf, vegf-b/vrf and vegf-c in the developing and adult mouse. Biochim Biophys Acta.1998; 1398:157–163.CrossrefMedlineGoogle Scholar
    • 16 Aase K, Lymboussaki A, Kaipainen A, Olofsson B, Alitalo K, Eriksson U. Localization of VEGF-B in the mouse embryo suggests a paracrine role of the growth factor in the developing vasculature. Dev Dyn.1999; 215:12–25.CrossrefMedlineGoogle Scholar
    • 17 Mountford P, Zevnik B, Duwel A, Nichols J, Li M, Dani C, Robertson M, Chambers I, Smith A. Dicistronic targeting constructs: reporters and modifiers of mammalian gene expression. Proc Natl Acad Sci U S A.1994; 91:4303–4307.CrossrefMedlineGoogle Scholar
    • 18 Oberdick J, Wallace JD, Lewin A, Smeyne RJ. Transgenic expression to monitor dynamic organization of neuronal development: use of the Escherichia coli lacZ gene product, B-galactosidase. Neuroprotocols.1994; 5:54–62.Google Scholar
    • 19 Headrick JP, McKirdy JC, Willis RJ. Functional and metabolic effects of extracellular magnesium in normoxic and ischemic myocardium. Am J Physiol.1998; 275:H917–H929.MedlineGoogle Scholar
    • 20 Fitzmaurice GM, Liard NM, Rotnitzky AG. Regression models for discrete and longitudinal responses. Stat Sci.1993; 8:284–309.CrossrefGoogle Scholar
    • 21 Olivetti G, Anversa P, Loud AV. Morphometric study of early postnatal development in the left and right ventricular myocardium of the rat, II: tissue composition, capillary growth, and sarcoplasmic alterations. Circ Res.1980; 46:503–512.CrossrefMedlineGoogle Scholar
    • 22 Hudlicka O, Brown MD. Postnatal growth of the heart and its blood vessels. J Vasc Res.1996; 33:266–287.CrossrefMedlineGoogle Scholar
    • 23 Viragh S, Challice CE. The origin of the epicardium and the embryonic myocardial circulation in the mouse. Anat Rec.1981; 201:157–168.CrossrefMedlineGoogle Scholar
    • 24 Mikawa T, Gourdie RG. Pericardial mesoderm generates a population of coronary smooth muscle cells migrating into the heart along with ingrowth of the epicardial organ. Dev Biol.1996; 174:221–232.CrossrefMedlineGoogle Scholar
    • 25 Gerber HP, Hillan KJ, Ryan AM, Kowalski J, Keller GA, Rangell L, Wright BD, Radtke F, Aguet M, Ferrara N. VEGF is required for growth and survival in neonatal mice. Development.1999; 126:1149–1159.CrossrefMedlineGoogle Scholar
    • 26 Gryglewski RJ, Chlopicki S, Niezabitowski P. Endothelial control of coronary flow in perfused guinea pig heart. Basic Res Cardiol.1995; 90:119–124.CrossrefMedlineGoogle Scholar
    • 27 Kanatsuka H, Sekiguchi N, Sato K, Akai K, Wang Y, Komaru T, Ashikawa K, Takishima T. Microvascular sites and mechanisms responsible for reactive hyperemia in the coronary circulation of the beating canine heart. Circ Res.1992; 71:912–922.CrossrefMedlineGoogle Scholar
    • 28 Gattullo D, Pagliaro P, Linden RJ, Merletti A, Losano G. The role of nitric oxide in the initiation and in the duration of some vasodilator responses in the coronary circulation. Pflugers Arch.1995; 430:96–104.CrossrefMedlineGoogle Scholar
    • 29 Godecke A, Decking UK, Ding Z, Hirchenhain J, Bidmon HJ, Godecke S, Schrader J. Coronary hemodynamics in endothelial NO synthase knockout mice. Circ Res.1998; 82:186–194.CrossrefMedlineGoogle Scholar
    • 30 Kingsley PB, Sako EY, Yang MQ, Zimmer SD, Ugurbil K, Foker JE, From AH. Ischemic contracture begins when anaerobic glycolysis stops: a 31P-NMR study of isolated rat hearts. Am J Physiol.1991; 261:H469–H478.CrossrefMedlineGoogle Scholar
    • 31 Meissner A, Morgan JP. Contractile dysfunction and abnormal Ca2+ modulation during postischemic reperfusion in rat heart. Am J Physiol.1995; 268:H100–H111.MedlineGoogle Scholar

    eLetters(0)

    eLetters should relate to an article recently published in the journal and are not a forum for providing unpublished data. Comments are reviewed for appropriate use of tone and language. Comments are not peer-reviewed. Acceptable comments are posted to the journal website only. Comments are not published in an issue and are not indexed in PubMed. Comments should be no longer than 500 words and will only be posted online. References are limited to 10. Authors of the article cited in the comment will be invited to reply, as appropriate.

    Comments and feedback on AHA/ASA Scientific Statements and Guidelines should be directed to the AHA/ASA Manuscript Oversight Committee via its Correspondence page.