Abstract 533: Shear-Induced Collateral Artery Growth Modulated by Endoglin but Not by ALK1

2012 ◽  
Vol 32 (suppl_1) ◽  
Author(s):  
Margreet R de Vries ◽  
Leonard Seghers ◽  
Imo E Hoefer ◽  
Evangelia Pardali ◽  
Beerend P Hierck ◽  
...  

Introduction: TGF- β stimulates both shear stress induced arteriogenesis and ischemia induced angiogenesis, by signaling through different receptors. In this study the role of TGF-β receptors ALK1 and endoglin is assessed in a murine model for hind limb ischemia induced neovascularization. Methods and Results: Femoral artery ligation was performed in mice heterozygous for either endoglin, or ALK1 and littermate controls. Blood flow recovery, monitored by laser doppler perfusion imaging, was significantly hampered in both endoglin and ALK1 heterozygous mice compared to controls by maximal 40% and 49%, respectively. Collateral artery size was significantly reduced in endoglin heterozygous mice compared to controls, but not in ALK1 heterozygous mice. Capillary density in ischemic calf muscles was unaffected, but capillaries from endoglin and ALK1 heterozygous mice were significantly larger when compared to controls. Furthermore, both endoglin and ALK1 heterozygous mice showed significant more erythrocyte extravasation near the capillaries than their littermate controls, pointing towards less mature capillaries. Murine endothelial cells were exposed to shear stress in vitro to study the differential role of endoglin and ALK1 in shear induced neovascularization. This resulted in high levels of endoglin messengerRNA, but not of ALK1. Conclusion: Here we demonstrated that both endoglin and ALK1 play a crucial role in blood flow recovery. Importantly, endoglin is essential in both shear induced collateral artery growth and in ischemia induced angiogenesis, whereas ALK1 is only involved in ischemia induced angiogenesis.

2021 ◽  
Vol 18 (2) ◽  
pp. 147916412199903
Author(s):  
Laura Croteau ◽  
Clément Mercier ◽  
Étienne Fafard-Couture ◽  
Alexandre Nadeau ◽  
Stéphanie Robillard ◽  
...  

Aims: Peripheral artery disease is a complication of diabetes leading to critical hindlimb ischemia. Diabetes-induced inhibition of VEGF actions is associated with the activation of protein kinase Cδ (PKCδ). We aim to specifically investigate the role of PKCδ in endothelial cell (EC) function and VEGF signaling. Methods: Nondiabetic and diabetic mice, with ( ec-Prkcd−/−) or without ( ec-Prkcdf/f) endothelial deletion of PKCδ, underwent femoral artery ligation. Blood flow reperfusion was assessed up to 4 weeks post-surgery. Capillary density, EC apoptosis and VEGF signaling were evaluated in the ischemic muscle. Src homology region 2 domain-containing phosphatase-1 (SHP-1) phosphatase activity was assessed in vitro using primary ECs. Results: Ischemic muscle of diabetic ec-Prkcdf/f mice exhibited reduced blood flow reperfusion and capillary density while apoptosis increased as compared to nondiabetic ec-Prkcdf/f mice. In contrast, blood flow reperfusion and capillary density were significantly improved in diabetic ec-Prkcd−/− mice. VEGF signaling pathway was restored in diabetic ec-Prkcd−/− mice. The deletion of PKCδ in ECs prevented diabetes-induced VEGF unresponsiveness through a reduction of SHP-1 phosphatase activity. Conclusions: Our data provide new highlights in mechanisms by which PKCδ activation in EC contributed to poor collateral vessel formation, thus, offering novel therapeutic targets to improve angiogenesis in the diabetic limb.


Circulation ◽  
2014 ◽  
Vol 130 (suppl_2) ◽  
Author(s):  
Sebastian Cremer ◽  
Anne Klotzsche-von Ameln ◽  
Alessia Orlandi ◽  
Irina Korovina ◽  
Bettina Gercken ◽  
...  

Developmental endothelial locus-1 (Del-1) is an endothelial cell-derived secreted protein circulating in blood and associated with the cell surface and the extracellular matrix. As we previously demonstrated, Del-1 restricts leukocyte recruitment by inhibiting the β2-integrin, LFA-1. Leukocytes and progenitor cells (PC) may contribute to angiogenesis. The role of endogenous Del-1 in angiogenesis is elusive. We found, that physiological angiogenesis of the developing retina was not affected in the Del-1-/- mice compared to the wildtype (WT) mice. Surprisingly, Del-1-/- mice displayed a significantly increased angiogenic response compared to WT mice after induction of hind limb ischemia (144 ± 6 % increase of capillary density) and retinal ischemia (retinopathy of prematurity model) suggesting that endogenous Del-1 is an inhibitor of ischemia-induced neovascularization. Silencing of Del-1 with siRNA did not affect the angiogenic sprouting of endothelial cell (EC) spheroids, indicating that Del-1 blocks angiogenesis in a non-endothelial cell autonomous pathway. Soluble Del-1 blocked the adhesion of inflammatory cells on EC monolayers. In line with these results, ischemic muscles and ischemic retinae from Del-1-/- mice displayed an enhanced infiltration with inflammatory cells compared to the WT mice. Since Del-1 blocks inflammatory cell homing by inhibiting the leukocytic LFA-1-integrin, we addressed the role of the Del-1/LFA-1-integrin interaction on the inhibitory function of endogenous Del-1 on angiogenesis. Indeed, Del-1/LFA-1-double deficiency reversed the pro-angiogenic phenotype of the Del-1-/- mice to the level of WT mice in the model of hind limb ischemia. Thus, the inhibitory role of Del-1 on neovascularization is mediated by the interaction of Del-1 with the LFA-1-integrin. Moreover, Del-1-deficiency led to an increased homing of intravenously injected murine fluorescence-labeled WT Lin- BM PC in ischemic muscles in comparison to WT mice after the induction of hind limb ischemia. Taken together, Del-1 acts as a negative regulator of ischemia-induced angiogenesis by interacting with the LFA-1-integrin expressed in hematopoietic cells, thereby inhibiting the homing of hematopoietic cells to ischemic tissues.


Circulation ◽  
2008 ◽  
Vol 118 (suppl_18) ◽  
Author(s):  
Marie-Ange Renault ◽  
Jerome Roncalli ◽  
Joern Tongers ◽  
Sol Misener ◽  
Tina Thorne ◽  
...  

Gli transcription factors are mediators of hedgehog signaling and have been shown to be critical in several steps during development. We have shown that the Hedgehog pathway is reactivated in the adult cardiovascular system under ischemic conditions, however the specific role of Gli3 has not been elucidated. Adenoviral mediated overexpression of Gli3 promotes HUVEC migration (250±58% of control, p<0.001) while down regulation of Gli3 via siRNA delayed tube formation on Matrigel (total tube length after 8 hours 6.86 vs. 70.76 control), suggesting a possible role of Gli3 in angiogenesis. We next investigated the role of Gli3 in angiogenesis using Gli3 +/− (Gli3 +/XtJ ) mice, a well established model of reduced Gli3 expression. VEGF-induced corneal angiogenesis was impaired in Gli3 +/− mice compared to WT. The role of Gli3 in angiogenesis was then confirmed in two ischemia models. Hind-limb ischemia (HLI) was induced by resection of the left femoral artery. Capillary density was reduced by a mean of 48.40±12.08% in Gli3 +/− mice vs. WT 7, 14 and 28 days. Myocardial infarction (MI) was induced by ligation of the LAD. 28 days after MI, left ventricular function assessed by echo and histological analysis revealed that Gli3 +/− mice exhibit reduced ejection fraction (27.92±4.49% versus 37.56±7.02% for the WT, p=0.004), increased fibrosis area (33.65±9.73% versus 19.81±5.40% for the WT, p=0.007) and a decrease capillary density in the ischemic and border zones. These data indicate that Gli3 deficiency leads to impaired angiogenesis in both ischemic and non ischemic conditions. Moreover, the impairment in ischemia induced neovascularization is associated with more severe impairment of cardiac function after MI. The mechanism of Gli3’s effects was then investigated in vitro . Promoter reporter assays revealed that Gli3 overexpression inhibits Gli-dependent transcription, while Western analysis show increased Akt phosphorylation, activation of the ERK1/2 and increased c-Fos expression. Using a dominant negative Akt expressing virus and a MEK1/2 inhibitor, we show that Gli3 induced-EC migration is dependent on Akt and ERK1/2. These studies provide the first evidence that the Gli3 transcription factor regulates angiogenesis and EC phenotype.


Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 3697-3697
Author(s):  
Eun-Sun Yoo ◽  
KiHwan Kwon ◽  
Jee-Young Ahn ◽  
Soo-Ah Oh ◽  
Hye-Jung Chang ◽  
...  

Abstract Backgroud: Human umbilical cord blood (UCB) contains a high number of endothelial progenitor cells (EPCs) and may be useful for the treatment of ischemic disease. Recently, we have isolated EPCs from UCB having different biologic properties for angiogenic capabilities in vitro. In this present study, the aim is to examine the usefulness of OECs in hind-limb ischemia. Methods: Mononuclear cells from UCB cultured using EGM-2 medium with VEGF, IGF-1 and FGF for 21 days. Early spindle-shaped cells (early OECs), which were grown during the first week of culture and late cobblestone shaped cells (late OECs), which were in peak growth during the third week of culture were found. The hind-limb ischemia was established as follows: Athymic nude mice (BALB/C-nu) 18–22 g in weight were anesthetized with pentobarbital (60 mg/kg) and their left femoral arteries and main extension arteries were operatively resected. To examine the effect of the vasculogenesis of the two types of OECs, the mice were divided into three groups (PBS, early and late OECs). Twenty-four hours after operative excision 5 × 105 OECs in 200 μl and an equal volume of PBS were administered by intramuscular injection into the mice on hind-limb ischemia. To compare the effect of OECs on neovascularization in vivo, the analysis of blood flow of ischemic and healthy hind limbs was performed on days 1 and 21 after surgery using near-infrared (NIR) imaging with incocyanne Green (ICG). Results: Late OECs expressed a high level of mRNA on endothelial marker genes and formed capillary tubes in Matrigel plates. The early spindle cells excreted more angiogenic cytokines and had more migratory ability. We divided the mice into two groups according to the degree of perfusion; good (22.5–50%/min) and poor (0–22.5%) perfusion. OECs improved the blood flow of the ischemic hind-limb in the ’good’ perfusion group but not in the ’poor’ perfusion group. Early OECs led to a more significant improvement in blood flow than that of the late OECs. Conclusion: The different types of OECs from UCB have different biologic properties in vitro and different vasculogenic potential in vivo as well. The results might have potential application for the treatment of hind-limb ischemia.


Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 1790-1790
Author(s):  
M.R. Finney ◽  
L.R. Fanning ◽  
P.J. Vincent ◽  
D.G. Winter ◽  
M.A. Hoffman ◽  
...  

Abstract Recent reports have utilized a variety of cell types for cellular therapy in mediating therapeutic angiogenesis in response to ischemia. We sought to assess the vasculogeneic potential of selected CD133+ hematopoietic stem cells (HSC) from umbilical cord blood (UCB) utilizing in vitro functional assays and an in vivo murine hind-limb ischemia model. Methods & Results: Mononuclear cells (MNC) from UCB or bone marrow (BM) were incubated with CD133+ conjugated magnetic beads, followed by automated sorting through magnetic columns (Miltenyi). Routine yield of CD133+ cells was 0.5±0.2% of UCB MNC and 0.7±0.3% of BM MNC, with a purity of 79±2% (UCB, n=30) and 84±5% (BM, n=12). Surface expression in the UCB CD133+ population was 3.6±1.5% KDR(VEGFR2), 8.7± 3.8% CXCR4 and 22.7±2.8% CD105 compared to 9.2±1.8% KDR, 14.4±1.3% CXCR4 and 23.7±2.3% CD105 in the BM CD133+ population. We measured chemotactic migration of cells towards SDF-1 (100ng/mL) compared to control wells containing media alone. The fold increase over control was 4.9±2.9 UCB MNC, 1.8±0.7 UCB CD133+ and 8.3±1.7 BM CD133+ (n=3). Angiogenic protein assays of CD133+ cells demonstrated elevated levels of IL-8 production as compared to MNC (103+/−380 pg/mL greater in CD133+ than MNC from the same UCB unit) when cultured for 24h in basal media. NOD/SCID mice underwent ligation of the right femoral artery and were given cells or vehicle control via intracardiac injection immediately following injury. Mice were given 1 x 106 MNC or 0.5 x 106 CD133+ cells. Laser Doppler flow measurements were obtained from both limbs each week for 6 weeks and the ratio of perfusion in the ischemic/healthy limb was calculated. At 28 days, perfusion ratios were statistically higher in study groups receiving UCB CD133+ cells, 0.55±0.06 (n=9), BM CD133+ cells 0.47±0.07 (n=8), BM MNC 0.48±0.8 (n=6) compared to cytokine controls 0.37±0.03 (n=12, p<0.05). Mice receiving UCB MNC did not show statistically significant improvement in measured blood flow over control animals 0.42±0.05 (n=8, p=0.34). At sacrifice, bone marrow was harvested to assess engraftment of human cells by flow cytometric analysis. Mice injected with UCB CD133+ cells showed 19±4.9% positive huCD45 cells compared to 2.5±0.6% for UCB MNC, 1.6±0.4% for BM CD133+ cells and 2.3±0.3% for BM MNC (n=3). Histological studies from day 42 tissue samples of muscle distal to arterial ligation were evaluated for capillary density. Control animals had capillary density of 131±6.9 cells/mm2. Capillary density was statistically higher that controls in animals receiving UCB CD133+ (320±18; p<0.0001), BM CD133++ (183±9.3; p<0.0001), and UCB MNC (164±10.5; p=0.011). Mice treated with BM MNC (135±9.4) did not have a statistically significant increase in capillary density from controls (p=0.73). In addition, animals treated with either UCB or BM-derived CD133+ cells had statistically higher capillary density than unselected MNC (p=<0.0001 and p=0.0004, respectively). Conclusions: In vitro functional assays showed that UCB-derived CD133+ HSC demonstrate enhanced homing capability (migration) as well as the potential for cellular recruitment (via IL-8 production) for angiogenesis in response to ischemia. Furthermore, UCB derived CD133+ HSC mediate significantly improved blood flow in an in vivo murine hind-limb injury model of ischemia, indicating the greater vasculogenic potential of selected CD133+ cells from of this stem cell source.


2012 ◽  
Vol 2012 ◽  
pp. 1-10
Author(s):  
Kurt Belisle ◽  
Martin Andrassy ◽  
Jochen Schneider ◽  
Stephan Schiekofer

Objectives. Type 1 and 2 diabetes carry risk factors for the development of microvascular diseases with associated impairment of angiogenic repair. Here, we investigated whether adiponectin, an adipocyte-specific adipocytokine with antiatherosclerotic and antidiabetic properties, regulates angiogenic repair in response to tissue ischemia in Leprdb/db and streptozocin-treated diabetic mouse models. Methods. Adenoviral vectors containing the gene for β-galactosidase, full-length mouse adiponectin, and dominant-negative AMPKα2 were used in streptozocin-treated male Leprdb/db mice, after which hind limb blood flow was measured using a laser doppler blood flow analyzer. Results. The angiogenic repair of ischemic hind limbs was impaired in both streptozocin-treated and Leprdb/db mice compared to wild-type mice as evaluated by laser doppler flow and capillary density analyses. Adenovirus-mediated administration of adiponectin accelerated angiogenic repair after hind limb ischemia in WT mice, but not in Leprdb/db mice or mice treated with streptozocin. In vitro experiments using HUVECs highlighted the antiapoptotic and proangiogenic properties of adiponectin but could not demonstrate accelerated differentiation of endothelial cells into tube-like structures at elevated glucose levels. Conclusions. External administration of adiponectin at elevated glucose levels may not be useful in the treatment of diabetes mellitus-related vascular deficiency diseases.


Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 3922-3922
Author(s):  
Marcie R. Finney ◽  
Matthew Joseph ◽  
Daniel G. Winter ◽  
Omar Masari ◽  
Margaret Kozik ◽  
...  

Abstract Recent reports have studied the use of various cell populations from bone marrow, peripheral blood and umbilical cord blood (UCB) in mediating therapeutic angiogenesis. We sought to investigate the surface phenotype of UCB derived-mononuclear cells (MNC), CD133+ cells and endothelial generating cells (EGC) using flow cytometry and in vitro functional migration studies. A murine hind-limb injury ischemia model was used to assess in vivo efficacy of the different cell populations. METHODS: Mononuclear cells were isolated by density centrifugation. CD133+ cells were isolated from UCB MNC by magnetic separation (Miltenyi). EGC were derived by adherence of the CD133- cells overnight on fibronectin-coated tissue culture plates in EGM2 media (Clonetics). Surface phenotype was determined by flow cytometry for stem cell markers, CD133 and CD34, stromal markers CD73 and CD105, KDR (VEGFR2), and the receptor for SDF-1, CXCR4. Modified Boyden chambers (Neuroprobe) were used to observe chemotactic migration of MNC, CD133, EGC or the combination of CD133 and EGC towards SDF-1 (100ng/mL) compared to control wells containing media alone. In the in vivo studies, 40 NOD.SCID mice underwent right femoral artery ligation. Mice were randomized into five study groups: Cytokines (EGM2 media, n=5), MNC (n=5), CD133 (n=5), EGC (n=7), or CD133 and EGC (1:2 ratio, n=4). Laser Doppler blood flow measurements were recorded weekly for four weeks and the ratio of ischemic/non-ischemic leg was calculated. At day 28, tissue samples were harvested for histological studies. RESULTS: Surface phenotype by flow cytometry showed an average purity of 78.67% +/− 2.41% for the selection of CD133. In addition, an enhancement of the expression of CXCR4 was seen in the EGC population after overnight exposure to fibronectin and EGM2 media. Surface Phenotype of UCB MNC, CD133 and EGC MNC CD133 EGC CD34 4.03 87.42 3.40 CD133 3.19 78.67 5.06 CD73 5.07 N/A 7.15 CD105 6.74 22.74 33.78 KDR 7.14 3.59 37.65 CXCR4 28.54 8.66 64.57 The in vitro functional migration assays showed increased migration of MNC, EGC, and CD133 with EGC to SDF-1, where the CD133 cells alone showed no increased migration compared to control media. In the in vivo murine model of hind-limb ischemia, the blood flow ratio of ischemic/non-ischemic limb was used to estimate the rate of blood flow recovery. The rates of blood flow recovery were 0.0198 (CD133), 0.02 (CD133 and EGC), 0.0163 (MNC), 0.016 (EGC) and 0.0123 (cytokines). The rates were significantly different between CD133 and cytokines (p=0.011) and between CD133 and EGC compared to cytokines (p=0.011). The difference between MNC and cytokines (p=0.156) and between EGC and cytokines (p=0.176) was not significant. Histological studies are ongoing. CONCLUSION: Surface phenotype of UCB-derived MNC, CD133, and EGC were compared by flow cytometry. The in vitro functional chemotactic capacity toward SDF-1 of these cell types was determined by migration assays. The infusion of CD133+ or the combination of CD133 and EGC cells augmented the rate of blood flow recovery in the in vivo murine hind-limb model of ischemia compared to the crude MNC prep or the ECG alone.


Blood ◽  
2020 ◽  
Vol 136 (Supplement 1) ◽  
pp. 8-8
Author(s):  
Qian Ye ◽  
Shuyang He ◽  
Joseph Gleason ◽  
Bhavani Stout ◽  
Navjot Shah ◽  
...  

Background Ischemic cardiovascular diseases are the major causes of death in the world. Recently, mesenchymal or tissue progenitor cell derived exosomes are being investigated as an emerging therapeutic agent for treating cardiovascular diseases due to their potentials in restoring the damaged vasculature via promoting angiogenesis. Exosome therapy has the advantages in low immunogenicity and stability features comparing to cells 1. Tissue and organ derived exosomes could have a broader functionality than those derived from a homogeneous cell population as they contain multiple cell types. Placenta is a large and highly vascularized organ and placenta exosomes are known to play essential roles during pregnancy in supporting fetus development 2. Placenta is abundant with endothelial cells, epithelial cells, cytotrophoblast, syncytiotrophoblasts and stem cells including both hematopoietic and non-hematopoietic stem cells. It is perceivable that exosomes isolated from a placenta possess the functions of the cells in a placenta. Materials and Methods Full-term human placentas were obtained under the full consent of healthy donors. Placenta tissues were cultured in serum-free DMEM medium supplemented with antibiotics. After culturing 8 to 16 hours, supernatant was harvested, medium was changed every 8 to 12 hours for up to two days. Placenta derived exosomes (pExo) were isolated through sequential centrifugation of the supernatant of the placenta culture and final preparation were suspended in PBS and stored at -80oC. Quantification of pExo was performed with Bicinchoninic Acid protein assay kit. pExo size was determined using NanoSight. MILLIPLEX-MAP human cytokine/chemokine-PX41 was used to analyze the cytokine composition of pExo. In vitro cell migration assays were performed with seeding 1x10e5/mL human umbilical vessel endothelial cells (HUVEC) in basal media (BM) on the top chamber of an 8-um transwell on a 24-well plate with 500uL BM with or without pExo in the bottom chamber. After 6-24 hours of culture, transwells were observed under an inverted microscope after staining. In vitro cell proliferation assay was performed by seeding HUVEC at 2-4x10e3 cells/96-well with a water-soluble tetrazolium salt-based cell viability assay. To evaluate the pro-angiogenic activity of pExo in vivo, a hind limb ischemia (HLI) model in db/db diabetic mice was used. After surgical induction of HLI on Day 0, pExo from two different donors or vehicle (n=15 each group) was injected via i.v. at 100ug/mice or 100uL on Days 1, 6 and 11. The blood flow through hind paws was measured by Speckle Doppler. Results Utilizing the established cultivation and isolation methods, we achieved the average yield of pExo at 327±91 mg per placenta (n=10). pExo has an average size of 118±15 nm (n=10) as determined with nano-particle tracking analysis, consistent with consensus exosome size ranging 50nm to 200nm 1. Flow cytometry and Western blot analyses confirmed that pExo exhibits characteristic markers known for exosomes including CD9, CD63 and CD81. pExo contained abundant IL-8, HGF, FGF2, PDGF-BB, RANTES, MCP-1 and GM-CSF which are known to play roles in supporting angiogenesis and chemotaxis. In co-culture assays, pExo promoted the proliferation of HUVECs by 149±13% (n=5) comparing to basal medium controls. pExo also demonstated chemotactic activities comparable to that of complete HUVEC growth medium in stimulating the migration of HUVECs across membrane of transwell. Chlorpromazine and Pitstop2 abolished pExo-induced transwell-migration of HUVECs, suggesting that HUVECs may uptake pExo through endocytosis. In the HLI study, pExo treated mice showed significantly higher (50% to 250%) blood flow on Days 14, 28 and 35 comparing to the vehicle group. Histology analysis showed pExo treated mice had 40% to 140% higher density of CD34+ capillary on Day 35, suggesting a significant increase of angiogenesis in the pExo treated groups comparing to the vehicle group. Conclusion: In summary, pExo contains proangiogenic cytokines and chemokines and demonstrates the promising pro-angiogenic activities in vitro and in HLI mice model. These results support further development of pExo as a potential therapeutic agent for the treatment of cardiosvascular diseases. References: Phinney and Pittenger (2017). Stem Cells. 35:851 Sarker et al., (2014). Journal of Translational Medicine. 12:204 Disclosures Ye: Celularity Inc.: Current Employment, Patents & Royalties. He:Celularity Inc.: Current Employment, Current equity holder in private company. Gleason:Celularity Inc.: Current Employment, Current equity holder in private company. Stout:Celularity: Current Employment, Current equity holder in private company. Shah:Celularity: Current Employment. Somanchi:Celularity Inc.: Current Employment, Current equity holder in private company. Zhang:Celularity Inc.: Current Employment, Current equity holder in private company. Hariri:Celularity Inc.: Current Employment, Current equity holder in private company.


2008 ◽  
Vol 389 (6) ◽  
Author(s):  
Robert S. Smith ◽  
Lin Gao ◽  
Lee Chao ◽  
Julie Chao

Abstract Adenovirus-mediated kallikrein delivery has been shown to promote blood vessel growth in the limb under both ischemic and normoperfused conditions. Here we investigated whether a continuous supply of kallikrein and kinin peptide can induce neovascularization in a rat model of hindlimb ischemia. Rats underwent femoral artery ligation and localized injection of tissue kallikrein, bradykinin or B1 receptor agonist, followed by infusion of proteins by osmotic minipump. Regional blood flow was monitored weekly by laser Doppler perfusion imaging. Three weeks after surgery, rats receiving kallikrein and kinins showed a significant increase in the perfusion ratio of ischemic vs. normoperfused limb compared to control rats. Similarly, a microsphere assay showed that kallikrein and kinins significantly increased regional blood flow without altering blood pressure. Moreover, kallikrein and kinins significantly augmented capillary and arteriole densities, as quantified by immunostaining with CD-31 and smooth muscle α-actin. Both tissue kallikrein and bradykinin increased hemoglobin content in Matrigel implants in mice, providing further evidence of the angiogenic properties. Kinins, when delivered subcutaneously via Matrigel in rats, also increased regional perfusion. This is the first demonstration that local application of tissue kallikrein protein or kinin peptide has therapeutic value in the treatment of ischemic disease by promoting neovascularization.


2021 ◽  
Vol 11 (1) ◽  
Author(s):  
Sonia Selbonne ◽  
Celina Madjene ◽  
Benjamin Salmon ◽  
Yacine Boulaftali ◽  
Marie-Christine Bouton ◽  
...  

AbstractWe previously identified the inhibitory serpin protease nexin-1 (PN-1) as an important player of the angiogenic balance with anti-angiogenic activity in physiological conditions. In the present study, we aimed to determine the role of PN-1 on pathological angiogenesis and particularly in response to ischemia, in the mouse model induced by femoral artery ligation. In wild-type (WT) muscle, we observed an upregulation of PN-1 mRNA and protein after ischemia. Angiography analysis showed that femoral artery perfusion was more rapidly restored in PN-1−/− mice than in WT mice. Moreover, immunohistochemistry showed that capillary density increased following ischemia to a greater extent in PN-1−/− than in WT muscles. Moreover, leukocyte recruitment and IL-6 and MCP-1 levels were also increased in PN-1−/− mice compared to WT after ischemia. This increase was accompanied by a higher overexpression of the growth factor midkine, known to promote leukocyte trafficking and to modulate expression of proinflammatory cytokines. Our results thus suggest that the higher expression of midkine observed in PN-1- deficient mice can increase leukocyte recruitment in response to higher levels of MCP-1, finally driving neoangiogenesis. Thus, PN-1 can limit neovascularisation in pathological conditions, including post-ischemic reperfusion of the lower limbs.


Sign in / Sign up

Export Citation Format

Share Document