Abstract 249: Plasma Exosomes Impair Angiogenesis in Ischemic Hind Limb of Diabetic Mice- Role of Histone Methylation

2020 ◽  
Vol 127 (Suppl_1) ◽  
Author(s):  
Zhongjian Cheng ◽  
May M Truongcao ◽  
Chunlin Wang ◽  
Venkata NS Garikipati ◽  
Yan Tang ◽  
...  

Background: Critical limb ischemia (CLI) is one of most prevenient cardiovascular disease in diabetic patients. Recent evidence suggests that altered cargo and function of plasma exosomes (plasma-Exo) may play an important role in diabetes-induced cardiovascular complications. Here, we tested the hypotheses that inhibition of exosome biosynthesis/release improves ischemic hind limb (IHL) repair in db/db mice. Methods: Plasma-Exo from db/+ and db/db mice were isolated by density-gradient ultracentrifugation. Unilateral IHL in mice was conducted by ligation of left femoral artery. Blood perfusion in IHL was measured by Laser Doppler Imager. Results: Diabetic plasma-Exo impaired tube formation/migration of human microvascular endothelial cells (HMVECs) and blood perfusion in IHL of C57BL/6J mice. Exosome inhibitor GW4869 improved blood flow, capillary density, cell survival, and rescued necrosis of toe/toenail and fibrosis in IHL muscle of db/db mice. Mechanistically, diabetic plasma-Exo decreased secretion of pro-angiogenic factor Ang I&II, artemin, FGF2 and IGFBP1&2, and increased repressive transcriptional mark H3K27me3 and its methylase enhancer of zest homolog-2 (EZH2) in HMVECs. EZH2 inhibitor GSK343 rescued diabetic plasma-Exo-impaired tube formation and secretion of FGF2/artemin from HMVECs. Moreover, GW4869 reduced EZH2 and H3K27me3 protein expression in lung microvascular ECs of IHL db/db mice. Finally, diabetic plasma-Exo increased H3K27me3 level at promoter of artemin and FGF2. Conclusions: Diabetic plasma-Exo impair angiogenesis and IHL injury repair. Diabetic plasma-Exo impair reparative property of ECs via, at least in part, enhancement of EZH2/H3K27me3/artemin and FGF2 cascade. Inhibition of plasma-Exo biosynthesis/secretion improve IHL repair in db/db mice. Plasma-Exo may be a novel target for prevention/treatment of CLI in diabetic patients.

2017 ◽  
Vol 121 (suppl_1) ◽  
Author(s):  
Zhongjian Cheng ◽  
Venkata Naga Srikanth Garikipati ◽  
Maria Cimini ◽  
Chunlin Wang ◽  
May Trungcao ◽  
...  

Background: Critical limb ischemia (CLI), a life-threatening condition characterized by pain at rest and tissue loss with ulcer and gangrene, imposes a major public healthy burden, resulting in high mortality and disability. The occurrence of CLI in patients with diabetes mellitus is very frequent. However, the effective therapy for CLI in diabetic patients is absent. Recent studies demonstrated that exosome from diabetic animals/cells has detrimental effects on the post-injury cardiovascular repair. Here, we tested the hypotheses that exosome inhibition in vivo improves blood flow recovery and protects skeletal muscle in ischemic hindlimbs of diabetic db/db mice following surgical ischemia. Methods and Results: Exosomes were isolated from bone-marrow derived progenitor cells or plasma in non-diabetic db/+ and diabetic db/db mice by ultracentrifugation. Diabetic exosome (5 ug/ml) inhibited tube formation of human cardiac microvascular endothelial cells. Unilateral hindlimb ischemia surgery was conducted by ligation of left femoral artery in 12-week old, male db/+ and db/db mice. Exosome inhibitor GW4869 (2 μg/g body weight) was given by intraperitoneal injection every other day for 4 weeks starting from one week before the HLI surgery. HLI mice injected with vehicle served as controls. Mice were divided into four groups: 1) db/+ + vehicle; 2) db/db+ vehicle; 3) db/+ GW4869; 4) db/db + GW4869. GW4869 decreased necrosis and loss of toe/toenail, improved blood flow, enhanced capillary/arterial density, skeletal muscle architecture and cell survival in ischemic hindlimb of diabetic db/db mice 21 days post-ligation. Conclusions: Although preliminary, our experiments suggest that therapeutic targeting of dysfunctional exosome secretion could represent a new avenue for the prevention and treatment of ischemic injury in diabetic patients.


2020 ◽  
Vol 10 (23) ◽  
pp. 8562
Author(s):  
Hyun Yang ◽  
Dong Ho Jung ◽  
Hye Won Lee ◽  
Dongoh Lee ◽  
Byoung Seob Ko

Tetragonia tetragonoides (Pall.) Kuntze (TTK) is grown for the edible leaves, and can be used as food. And which commonly called Beonhaengcho in Republic of Korea. TTK is found along the seaside of the Jeju-Island and it has long been consumed as a food for women’s health. We investigated the effects of TTK on peripheral circulation disorder during menopausal transition and/or menopause in a hind-limb ischemic (HLI) mouse model. Chemotactic motility and tube formation of vascular epithelial cells were evaluated in human umbilical vein endothelial cells (HUVECs). Female C57BL/6 mice were fed a TTK (150 or 450 mg/kg/day) for four weeks and the rate of blood flow was assessed using a laser Doppler after HLI. TTK treatment significantly increased cell migration and the branch interval value of tubular structure in a dose-dependently. In the TTK treatment group, blood flow rate was significant induced at 7, 14, and 28 days after HLI, compared with the vehicle. TTK treatment also an increase in capillary density, and the highest levels of pERK(1/2), pAkt, pPLCγ1 and pFAK proteins compared to the vehicle control. These results suggest that extract of TTK may ameliorate the blood flow via improvement of peripheral angiogenesis under hind-limb ischemic stress in a menopausal mouse model.


2021 ◽  
Author(s):  
Hongjuan Liu ◽  
Yang Dai ◽  
Yawei Xu ◽  
Lin Lu

Abstract Background Apo A-Ⅳ played an key role in Cardiovascular diseases, but the effect and mechanism of glycated apo A-Ⅳ on angiogenesis remains unclear. Methods In this study, we demonstrated that oral administration of glycated apoA-Ⅳ impaired blood perfusion recovery in a mouse hind-limb ischemia model. A reduction in blood perfusion recovery at day 21 was observed in the ischemic tissue of apoA-Ⅳ and glycated apoA-Ⅳ-treated mice. Results In this study, we demonstrated that glycated apo A-Ⅳ impaired blood perfusion recovery in a mouse hind-limb ischemia model. And in vitro study also showed that glycated apo A-Ⅳ inhibited the migration, proliferation, and tube-formation abilities of endothelial cells. Further research revealed that glycated apo A-Ⅳ regulated angiogenesis partly by interrupting Nur77. In addition, CML levels were increased in patients with Lower limb circulation (n = 30) compared with those with no limb circulation(n = 50). Conclusions We found impaired angiogenesis induced by glycated apo A-Ⅳ might contribute to poor coronary collateral growth by inhibit the expression of Nur77.


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.


Author(s):  
Junya Suzuki ◽  
Yuuki Shimizu ◽  
Kazuhito Tsuzuki ◽  
Zhongyue Pu ◽  
Shingo Narita ◽  
...  

Therapeutic angiogenesis with autologous stem/progenitor cells is a promising novel strategy for treatment of severe ischemic diseases. Human clinical trials utilizing autologous adipose-derived regenerative cells (ADRCs) have not reported treatment-related critical adverse effects thus far. However, there is still a large knowledge gap whether treatment of ischemic diseases with angiogenic therapy using ADRCs would promote unfavorable angiogenesis associated with tumors in vivo. Herein, we addressed this clinical question using a mouse hind limb ischemia (HLI) and simultaneous remote tumor implantation model. C57BL/6J background wild-type mice were injected with murine B16F10 melanoma cells on their back, one day before ischemic surgery. These mice were subjected to surgical unilateral hindlimb ischemia, followed by ADRCs implantation or PBS injection into the hindlimb ischemic muscles on the next day. Intramuscular implantation of ADRCs enhanced tissue capillary density and blood flow examined by a laser Doppler blood perfusion analysis in hind limb. However, this therapeutic regimen for ischemic limb using ADRCs did not affect remote melanoma growth nor the density of its feeder artery, angiogenesis and lymphatic vessels compared to the PBS group. In addition, no distant metastases were detected in any of the mice regardless the group. In conclusion, local implantation of ADRCs promotes angiogenesis in response to tissue ischemia in the hind limb without promoting remote tumor growth and related angio/lymphangiogenesis. Therapeutic angiogenesis to the ischemic hind limb using ADRCs seems to be safe regarding remote tumor growth.


2005 ◽  
Vol 12 (4) ◽  
pp. 469-478 ◽  
Author(s):  
Rong Fang Lan ◽  
Zheng Xiang Liu ◽  
Xiao Chun Liu ◽  
Yu E. Song ◽  
Dao Wen Wang

Circulation ◽  
2014 ◽  
Vol 130 (suppl_2) ◽  
Author(s):  
Mahesh Thirunavukkarasu ◽  
Inam A Shaikh ◽  
Vaithinathan Selvaraju ◽  
J.Alexandar Palesty ◽  
Nilanjana Maulik

Introduction: Peripheral artery disease affects 12-20% Americans over the age of 60. Thioredoxin-1 (Trx-1) is a class of small redox proteins. We have demonstrated earlier that Trx-1 reduces oxidative stress resulting in less inflammation and increased angiogenesis in cardiac muscle via heme oxygenase-1 (HO-1) and VEGF after myocardial infarction. In the current study, we evaluate the effect of Trx-1 on post-ischemic hindlimb recovery. Methods: Peripheral artery disease was mimicked using a hindlimb ischemia (HLI) model. Wild type (WT) and Trx-1 transgenic (Trx-1Tg/+) mice (8-12 weeks old) were subjected to femoral artery ligation. Following surgery, mice were observed for 5 weeks. Serial laser doppler images were obtained, and perfusion ratios between the ischemic and non-ischemic limbs were calculated at set time intervals. The perfusion ratios were compared between WT and Trx-1Tg/+ groups. Immunohistochemical analysis of the skeletal muscle was performed to quantify the extent of fibrosis, capillary and arteriolar density 35 days after surgery. In addition, another set of experiments was designed with Ad.Trx-1 gene therapy after femoral artery ligation to study the molecular mechanism of neovascularization with Trx-1. Results: The recovery of hind limb perfusion was significantly increased in Trx-1Tg/+ mice at day 7 (0.19 ± 0.03 vs. 0.36 ± 0.07 (n=12-9), day-21 (0.37 ± 0.05 vs. 0.62 ± 0.03 (n=12-9), and day 28 (0.40 ± 0.04 vs. 0.79 ± 0.04 (n=10-9); p<0.05). Capillary density [1265 ± 87.8 vs. 762.4 ± 86.6 counts/mm2 ; (n=5); p<0.05] and arteriolar density [36.2 ± 2.96 vs. 22± 1.33 counts/mm2 ; (n=5); p<0.05] staining showed significant increase in Trx-1Tg/+ mice as compared to WT mice. Picrosirrus Red and immunofluorescence staining showed decreased fibrosis [8.3 ± 0.46 vs. 22.2 ± 1.04 (n=5); p<0.0001] and increased HO-1 expression respectively in Trx-1Tg/+ mice group as compared to WT mice, respectively. Trx-1 gene therapy study also revealed by Western blot analysis, increased Trx-1 (4.2 fold) and HO-1 (8.2 fold) expression in Ad.Trx-1-HLI as compared to Ad.LacZ-HLI. Conclusions: Our results suggest that Trx-1 is a potential therapeutic agent to increase blood perfusion and angiogenesis for the treatment of critical limb ischemia patients.


2021 ◽  
Author(s):  
Si-Jia Sun ◽  
Ming Dong ◽  
Wing-Hon Lai ◽  
Wai-In Ho ◽  
Rui Wei ◽  
...  

Abstract We sought to demonstrate whether therapeutic efficacy can be improved by combination of repeated intravenous administration and local transplantation of human induced pluripotential stem cell derived MSCs (hiPSC-MSCs). In this study, mice model of hind-limb ischemia was established by ligation of left femoral artery. hiPSC-MSCs (5x105) was intravenously administrated immediately after induction of hind limb ischemia with or without following intravenous administration of hiPSC-MSCs every week or every 3 days. Intramuscular transplantation of hiPSC-MSCs (3x106) was performed one week after induction of hind-limb ischemia. We compared the therapeutic efficacy and cell survival of intramuscular transplantation of hiPSC-MSCs with or without a single or repeated intravenous administration of hiPSC-MSCs. Repeated intravenous administration of hiPSC-MSCs could increase splenic regulatory T cells (Tregs) activation, decrease splenic natural killer (NK) cells expression, promote the polarization of M2 macrophages in the ischemic area and improved blood perfusion in the ischemic limbs. The improved therapeutic efficacy of MSC-based therapy was due to both increased engraftment of intramuscular transplanted hiPSC-MSCs and intravenous infused hiPSC-MSCs. In conclusion, our study supported a combination of repeated systemic infusion and local transplantation of hiPSC-MSCs for cardiovascular disease.


2013 ◽  
Vol 217 (3) ◽  
pp. S28
Author(s):  
Salim Abunnaja ◽  
Vaithinathan Selvara ◽  
Mahesh Thirunavukka ◽  
David Mcfadden ◽  
J. Alexander Palesty ◽  
...  

Blood ◽  
2011 ◽  
Vol 118 (20) ◽  
pp. 5701-5712 ◽  
Author(s):  
Nikhlesh K. Singh ◽  
Venkatesh Kundumani-Sridharan ◽  
Gadiparthi N. Rao

Abstract To understand the mechanisms by which 15(S)-hydroxyeicosatetraenoic acid (15(S)-HETE) activates Rac1 in the induction of angiogenesis, we studied the role of 3-hydroxy-3-methylglutaryl–coenzyme A (HMG-CoA) reductase and αPix. 15(S)-HETE stimulated Rac1 in a sustained manner in human dermal microvascular endothelial cells (HDMVECs). Simvastatin, a potent inhibitor of HMG-CoA reductase, suppressed 15(S)-HETE–induced Rac1 activation in HDMVECs affecting their migration and tube formation. 15(S)-HETE by inducing HMG-CoA reductase expression caused increased farnesylation and membrane translocation of Rac1 where it became activated by Src-dependent αPix stimulation. Mevalonate rescued 15(S)-HETE–induced Rac1 farnesylation and membrane translocation in HDMVECs and the migration and tube formation of these cells from inhibition by simvastatin. Down-regulation of αPix inhibited 15(S)-HETE–induced HDMVEC migration and tube formation. Hind-limb ischemia induced Rac1 farnesylation and activation leading to increased angiogenesis and these effects were blocked by simvastatin and rescued by mevalonate in WT mice. In contrast, hind-limb ischemia failed to induce Rac1 farnesylation and activation as well as angiogenic response in 12/15-Lox−/− mice. Activation of Src and αPix were also compromised at least to some extent in 12/15-Lox−/− mice compared with WT mice in response to hind-limb ischemia. Together, these findings demonstrate for the first time that HMG-CoA reductase plays a determinant role in 12/15-Lox–induced angiogenesis.


Sign in / Sign up

Export Citation Format

Share Document