Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 4.538
Filter
Add more filters

Publication year range
1.
Proc Natl Acad Sci U S A ; 120(46): e2307480120, 2023 Nov 14.
Article in English | MEDLINE | ID: mdl-37943835

ABSTRACT

Ischemic diseases lead to considerable morbidity and mortality, yet conventional clinical treatment strategies for therapeutic angiogenesis fall short of being impactful. Despite the potential of biomaterials to deliver pro-angiogenic molecules at the infarct site to induce angiogenesis, their efficacy has been impeded by aberrant vascular activation and off-target circulation. Here, we present a semisynthetic low-molecular sulfated chitosan oligosaccharide (SCOS) that efficiently induces therapeutic arteriogenesis with a spontaneous generation of collateral circulation and blood reperfusion in rodent models of hind limb ischemia and myocardial infarction. SCOS elicits anti-inflammatory macrophages' (Mφs') differentiation into perivascular Mφs, which in turn directs artery formation via a cell-to-cell communication rather than secretory factor regulation. SCOS-mediated arteriogenesis requires a canonical Notch signaling pathway in Mφs via the glycosylation of protein O-glucosyltransferases 2, which results in promoting arterial differentiation and tissue repair in ischemia. Thus, this highly bioactive oligosaccharide can be harnessed to direct efficiently therapeutic arteriogenesis and perfusion for the treatment of ischemic diseases.


Subject(s)
Neovascularization, Physiologic , Sulfates , Mice , Animals , Neovascularization, Physiologic/physiology , Sulfates/metabolism , Mice, Knockout , Muscle, Skeletal/metabolism , Ischemia/metabolism , Macrophages/metabolism , Hindlimb/blood supply , Disease Models, Animal
2.
Circ Res ; 132(1): 34-48, 2023 01 06.
Article in English | MEDLINE | ID: mdl-36448444

ABSTRACT

BACKGROUND: Lower-limb peripheral artery disease is one of the major complications of diabetes. Peripheral artery disease is associated with poor limb and cardiovascular prognoses, along with a dramatic decrease in life expectancy. Despite major medical advances in the treatment of diabetes, a substantial therapeutic gap remains in the peripheral artery disease population. Praliciguat is an orally available sGC (soluble guanylate cyclase) stimulator that has been reported both preclinically and in early stage clinical trials to have favorable effects in metabolic and hemodynamic outcomes, suggesting that it may have a potential beneficial effect in peripheral artery disease. METHODS: We evaluated the effect of praliciguat on hind limb ischemia recovery in a mouse model of type 2 diabetes. Hind limb ischemia was induced in leptin receptor-deficient (Leprdb/db) mice by ligation and excision of the left femoral artery. Praliciguat (10 mg/kg/day) was administered in the diet starting 3 days before surgery. RESULTS: Twenty-eight days after surgery, ischemic foot perfusion and function parameters were better in praliciguat-treated mice than in vehicle controls. Improved ischemic foot perfusion was not associated with either improved traditional cardiovascular risk factors (ie, weight, glycemia) or increased angiogenesis. However, treatment with praliciguat significantly increased arteriole diameter, decreased ICAM1 (intercellular adhesion molecule 1) expression, and prevented the accumulation of oxidative proangiogenic and proinflammatory muscle fibers. While investigating the mechanism underlying the beneficial effects of praliciguat therapy, we found that praliciguat significantly downregulated Myh2 and Cxcl12 mRNA expression in cultured myoblasts and that conditioned medium form praliciguat-treated myoblast decreased ICAM1 mRNA expression in endothelial cells. These results suggest that praliciguat therapy may decrease ICAM1 expression in endothelial cells by downregulating Cxcl12 in myocytes. CONCLUSIONS: Our results demonstrated that praliciguat promotes blood flow recovery in the ischemic muscle of mice with type 2 diabetes, at least in part by increasing arteriole diameter and by downregulating ICAM1 expression.


Subject(s)
Diabetes Mellitus, Type 2 , Peripheral Arterial Disease , Mice , Animals , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/drug therapy , Receptors, Leptin/genetics , Endothelial Cells/metabolism , Ischemia/metabolism , Disease Models, Animal , Reperfusion , Peripheral Arterial Disease/complications , Hindlimb/blood supply , Neovascularization, Physiologic , Muscle, Skeletal/metabolism , Mice, Inbred C57BL
3.
Arterioscler Thromb Vasc Biol ; 44(6): 1225-1245, 2024 06.
Article in English | MEDLINE | ID: mdl-38511325

ABSTRACT

BACKGROUND: Restoring the capacity of endothelial progenitor cells (EPCs) to promote angiogenesis is the major therapeutic strategy of diabetic peripheral artery disease. The aim of this study was to investigate the effects of GLP-1 (glucagon-like peptide 1; 32-36)-an end product of GLP-1-on angiogenesis of EPCs and T1DM (type 1 diabetes) mice, as well as its interaction with the classical GLP-1R (GLP-1 receptor) pathway and its effect on mitochondrial metabolism. METHODS: In in vivo experiments, we conducted streptozocin-induced type 1 diabetic mice as a murine model of unilateral hind limb ischemia to examine the therapeutic potential of GLP-1(32-36) on angiogenesis. We also generated Glp1r-/- mice to detect whether GLP-1R is required for angiogenic function of GLP-1(32-36). In in vitro experiments, EPCs isolated from the mouse bone marrow and human umbilical cord blood samples were used to detect GLP-1(32-36)-mediated angiogenic capability under high glucose treatment. RESULTS: We demonstrated that GLP-1(32-36) did not affect insulin secretion but could significantly rescue angiogenic function and blood perfusion in ischemic limb of streptozocin-induced T1DM mice, a function similar to its parental GLP-1. We also found that GLP-1(32-36) promotes angiogenesis in EPCs exposed to high glucose. Specifically, GLP-1(32-36) has a causal role in improving fragile mitochondrial function and metabolism via the GLP-1R-mediated pathway. We further demonstrated that GLP-1(32-36) rescued diabetic ischemic lower limbs by activating the GLP-1R-dependent eNOS (endothelial NO synthase)/cGMP/PKG (protein kinase G) pathway. CONCLUSIONS: Our study provides a novel mechanism with which GLP-1(32-36) acts in modulating metabolic reprogramming toward glycolytic flux in partnership with GLP-1R for improved angiogenesis in high glucose-exposed EPCs and T1DM murine models. We propose that GLP-1(32-36) could be used as a monotherapy or add-on therapy with existing treatments for peripheral artery disease. REGISTRATION: URL: www.ebi.ac.uk/metabolights/; Unique identifier: MTBLS9543.


Subject(s)
Diabetes Mellitus, Experimental , Endothelial Progenitor Cells , Glucagon-Like Peptide 1 , Glucagon-Like Peptide-1 Receptor , Glycolysis , Hindlimb , Ischemia , Mice, Inbred C57BL , Mice, Knockout , Neovascularization, Physiologic , Signal Transduction , Animals , Ischemia/drug therapy , Ischemia/physiopathology , Ischemia/metabolism , Glucagon-Like Peptide-1 Receptor/metabolism , Glucagon-Like Peptide-1 Receptor/agonists , Neovascularization, Physiologic/drug effects , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/metabolism , Glycolysis/drug effects , Glucagon-Like Peptide 1/analogs & derivatives , Glucagon-Like Peptide 1/pharmacology , Humans , Hindlimb/blood supply , Male , Endothelial Progenitor Cells/metabolism , Endothelial Progenitor Cells/drug effects , Diabetic Angiopathies/metabolism , Diabetic Angiopathies/physiopathology , Diabetic Angiopathies/drug therapy , Diabetic Angiopathies/etiology , Nitric Oxide Synthase Type III/metabolism , Diabetes Mellitus, Type 1/drug therapy , Diabetes Mellitus, Type 1/metabolism , Cells, Cultured , Angiogenesis Inducing Agents/pharmacology , Peptide Fragments/pharmacology , Mice , Muscle, Skeletal/blood supply , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Disease Models, Animal , Incretins/pharmacology , Angiogenesis
4.
Ann Surg ; 280(4): 570-583, 2024 Oct 01.
Article in English | MEDLINE | ID: mdl-38975668

ABSTRACT

OBJECTIVE: This study focuses on dose-response investigation using a codon-optimized and de novo-synthesized E-Selectin/AAV2 (E-Sel/AAV2) vector in preparation for Investigational New Drug enabling of subsequent clinical studies. BACKGROUND: Gene therapy is a potential solution for patients suffering from chronic limb-threatening ischemia. Understanding the dose for effective gene delivery is crucial for future Investigational New Drug-enabling studies. METHODS: Expression of the codon-optimized E-Selectin gene was assessed by flow cytometry following in vitro cell transfection assay and RT-qPCR for murine limbs injected in vivo with AAV-m-E-Selectin (E-Sel/AAV2). Dose-response studies involved 3 cohorts of FVB/NJ mice (n=6/group) with escalating log doses of E-Selectin/AAV2 injected intramuscularly in divided aliquots, ranging from 2 × 10 9 VG to 2 × 10 11 VG, into ischemic limbs created by left femoral artery/vein ligation/excision and administration of nitric oxide synthase inhibitor, L-NAME. Limb perfusion, extent of gangrene free limb, functional limb recovery, and therapeutic angiogenesis were assessed. RESULTS: Codon-optimized E-Sel/AAV2 gene therapy exhibits a superior expression level than WT E-Sel/AAV2 gene therapy both in vitro and in vivo. Mice treated with a high dose (2 × 10 11 VG) of E-Sel/AAV2 showed significantly improved perfusion indices, lower Faber scores, increased running stamina, and neovascularization compared with lower doses tested with control groups, indicating a distinct dose-dependent response. No toxicity was detected in any of the animal groups studied. CONCLUSIONS: E-Sel/AAV2 Vascular Regeneration Gene Therapy holds promise for enhancing the recovery of ischemic hindlimb perfusion and function, with the effective dose identified in this study as 2 × 10 11 VG aliquots injected intramuscularly.


Subject(s)
Codon , E-Selectin , Genetic Therapy , Hindlimb , Ischemia , Animals , Genetic Therapy/methods , Mice , Ischemia/therapy , Hindlimb/blood supply , Dependovirus/genetics , Genetic Vectors , Disease Models, Animal , Neovascularization, Physiologic , Male , Regeneration
5.
Exp Physiol ; 109(9): 1604-1613, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38888281

ABSTRACT

Skeletal muscle atrophy and dysfunction commonly accompany cardiovascular diseases such as peripheral arterial disease and may be partially attributable to systemic inflammation. We sought to determine whether acute systemic inflammation in a model of hindlimb ischaemia (HLI) could affect skeletal muscle macrophage infiltration, fibre size, or capillarization, independent of the ischaemia. Eight-week-old C57BL/6 male mice underwent either Sham or HLI surgery, and were killed 1, 3, or 7 days post-surgery. Circulating inflammatory cytokine concentrations were measured, as well as immune cell infiltration and morphology of skeletal muscle from both limbs of HLI and Sham mice. In HLI compared with Sham mice at day 1, plasma interleukin-1ß levels were 216% higher (0.48 ± 0.10 vs. 0.15 ± 0.01 pg/µL, P = 0.005) and decreased by day 3. This was followed by increased macrophage presence in muscle from both ischaemic and non-ischaemic limbs of HLI mice by day 7 (7.3- and 2.3-fold greater than Sham, respectively, P < 0.0001). In HLI mice, muscle from the ischaemic limb had 21% lower fibre cross-sectional area than the non-ischaemic limb (724 ± 28 vs. 916 ± 46 µm2, P = 0.01), but the non-ischaemic limb of HLI mice was no different from Sham. This shows that HLI induces acute systemic inflammation accompanied by immune infiltration in both ischaemic and remote skeletal muscle; however, this did not induce skeletal muscle atrophy in remote muscle within the 7-day time course of this study. This effect of local skeletal muscle ischaemia on the inflammatory status of remote skeletal muscle may signal a priming of muscle for subsequent atrophy over a longer time course. HIGHLIGHTS: What is the central question of this study? Does hindlimb ischaemia-induced inflammation cause acute immune, inflammatory and morphological alterations in remote non-ischaemic skeletal muscle? What is the main finding and its importance? Hindlimb ischaemia induced systemic inflammation with subsequent neutrophil and macrophage infiltration in both ischaemic and non-ischaemic skeletal muscle; however, morphological changes did not occur in non-ischaemic muscle within 7 days. These immune alterations may have functional implications that take longer than 7 days to manifest, and subsequent or prolonged systemic inflammation and immune infiltration of muscle could lead to morphological changes and functional decline.


Subject(s)
Hindlimb , Inflammation , Ischemia , Macrophages , Mice, Inbred C57BL , Muscle, Skeletal , Animals , Hindlimb/blood supply , Muscle, Skeletal/pathology , Male , Ischemia/pathology , Ischemia/immunology , Inflammation/pathology , Mice , Macrophages/pathology , Macrophages/immunology , Muscular Atrophy/pathology , Interleukin-1beta/metabolism
6.
Biomacromolecules ; 25(10): 6656-6665, 2024 Oct 14.
Article in English | MEDLINE | ID: mdl-39264109

ABSTRACT

Critical limb ischemia (CLI) is a peripheral arterial disease resulting from chronic inflammation of vascular systems. Recent studies have shown that inhibiting macrophage inflammation has the potential to treat CLI, and extracellular vesicles (EVs) from endothelial cells can inhibit macrophage activation. However, the limited cell-targeting capabilities and rapid clearance of EVs from the injection site limit the in vivo application of the EVs. Here, we modified endothelial EVs with platelet membranes (pM/EVs) to boost the inhibition effects on macrophage inflammation and developed an injectable alginate-based collagen composite (ACC) hydrogel for localized delivery of pM/EVs (pM/EVs@ACC) for CLI treatment. We found that pM/EVs can effectively inhibit macrophage inflammation in vitro. Furthermore, pM/EVs@ACC treatment significantly promotes the recovery of limb functions, restoring the feet' blood supply and relieving inflammation. Our findings provide compelling evidence that the pM/EVs@ACC injectable system mediating delivery of pM/EVs is a promising strategy for CLI treatment.


Subject(s)
Alginates , Extracellular Vesicles , Hydrogels , Ischemia , Animals , Alginates/chemistry , Hydrogels/chemistry , Hydrogels/pharmacology , Extracellular Vesicles/chemistry , Extracellular Vesicles/metabolism , Ischemia/therapy , Ischemia/pathology , Mice , Humans , Human Umbilical Vein Endothelial Cells , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Macrophages/metabolism , Macrophages/drug effects , Male , Inflammation , RAW 264.7 Cells , Hindlimb/blood supply , Mice, Inbred C57BL
7.
Arterioscler Thromb Vasc Biol ; 43(7): 1262-1277, 2023 07.
Article in English | MEDLINE | ID: mdl-37051932

ABSTRACT

BACKGROUND: Peripheral vascular disease remains a leading cause of vascular morbidity and mortality worldwide despite advances in medical and surgical therapy. Besides traditional approaches, which can only restore blood flow to native arteries, an alternative approach is to enhance the growth of new vessels, thereby facilitating the physiological response to ischemia. METHODS: The ActinCreER/R26VT2/GK3 Rainbow reporter mouse was used for unbiased in vivo survey of injury-responsive vasculogenic clonal formation. Prospective isolation and transplantation were used to determine vessel-forming capacity of different populations. Single-cell RNA-sequencing was used to characterize distinct vessel-forming populations and their interactions. RESULTS: Two populations of distinct vascular stem/progenitor cells (VSPCs) were identified from adipose-derived mesenchymal stromal cells: VSPC1 is CD45-Ter119-Tie2+PDGFRa-CD31+CD105highSca1low, which gives rise to stunted vessels (incomplete tubular structures) in a transplant setting, and VSPC2 which is CD45-Ter119-Tie2+PDGFRa+CD31-CD105lowSca1high and forms stunted vessels and fat. Interestingly, cotransplantation of VSPC1 and VSPC2 is required to form functional vessels that improve perfusion in the mouse hindlimb ischemia model. Similarly, VSPC1 and VSPC2 populations isolated from human adipose tissue could rescue the ischemic condition in mice. CONCLUSIONS: These findings suggest that autologous cotransplantation of synergistic VSPCs from nonessential adipose tissue can promote neovascularization and represents a promising treatment for ischemic disease.


Subject(s)
Mesenchymal Stem Cells , Neovascularization, Physiologic , Mice , Humans , Animals , Neovascularization, Physiologic/physiology , Adipose Tissue , Neovascularization, Pathologic , Ischemia/therapy , Disease Models, Animal , Hindlimb/blood supply
8.
Arterioscler Thromb Vasc Biol ; 43(4): 504-518, 2023 04.
Article in English | MEDLINE | ID: mdl-36756881

ABSTRACT

BACKGROUND: Angiogenesis is a promising strategy for those with peripheral artery disease. Macrophage-centered inflammation is intended to govern the deficiency of the angiogenic response after hindlimb ischemia. However, little is known about the mechanism of macrophage activation beyond signals from cytokines and chemokines. We sought to identify a novel mechanical signal from the ischemic microenvironment that provokes macrophages and the subsequent inflammatory cascade and to investigate the potential role of Piezo-type mechanosensitive ion channels (Piezo) on macrophages during this process. METHODS: Myeloid cell-specific Piezo1 (Piezo-type mechanosensitive ion channel component 1) knockout (Piezo1ΔMΦ) mice were generated by crossing Piezo1fl/fl (LysM-Cre-/-; Piezo1 flox/flox) mice with LysM-Cre transgenic mice to assess the roles of Piezo1 in macrophages after hindlimb ischemia. Furthermore, in vitro studies were carried out in bone marrow-derived macrophages to decipher the underlying mechanism. RESULTS: We found that tissue stiffness gradually increased after hindlimb ischemia, as indicated by Young's modulus. Compared to Piezo2, Piezo1 expression and activation were markedly upregulated in macrophages from ischemic tissues in concurrence with increased tissue stiffness. Piezo1ΔMΦ mice exhibited improved perfusion recovery by enhancing angiogenesis. Matrigel tube formation assays revealed that Piezo1 deletion promoted angiogenesis by enhancing FGF2 (fibroblast growth factor-2) paracrine signaling in macrophages. Conversely, activation of Piezo1 by increased stiffness or the agonist Yoda1 led to reduced FGF2 production in bone marrow-derived macrophages, which could be blocked by Piezo1 silencing. Mechanistically, Piezo1 mediated extracellular Ca2+ influx and activated Ca2+-dependent CaMKII (calcium/calmodulin-dependent protein kinase II)/ETS1 (ETS proto-oncogene 1) signaling, leading to transcriptional inactivation of FGF2. CONCLUSIONS: This study uncovers a crucial role of microenvironmental stiffness in exacerbating the macrophage-dependent deficient angiogenic response. Deletion of macrophage Piezo1 promotes perfusion recovery after hindlimb ischemia through CaMKII/ETS1-mediated transcriptional activation of FGF2. This provides a promising therapeutic strategy to enhance angiogenesis in ischemic diseases.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Type 2 , Fibroblast Growth Factor 2 , Animals , Mice , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Fibroblast Growth Factor 2/metabolism , Ion Channels , Mice, Transgenic , Macrophages/metabolism , Ischemia , Perfusion , Hindlimb/blood supply
9.
Arterioscler Thromb Vasc Biol ; 43(1): e46-e61, 2023 01.
Article in English | MEDLINE | ID: mdl-36384268

ABSTRACT

BACKGROUND: Diabetes is a major risk factor for peripheral arterial disease. Clinical and preclinical studies suggest an impaired collateral remodeling and angiogenesis in response to atherosclerotic arterial occlusion in diabetic conditions, although the underlying mechanisms are poorly understood. OBJECTIVE: To clarify the cellular and molecular mechanisms underlying impaired postischemic adaptive vascular responses and to evaluate rHDL (reconstituted HDL)-ApoA-I nanotherapy to rescue the defect in type 2 diabetic mouse model of hindlimb ischemia. METHODS AND RESULTS: Hindlimb ischemia was induced by unilateral femoral artery ligation. Collateral and capillary parameters together with blood flow recovery were analyzed from normoxic adductor and ischemic gastrocnemius muscles, respectively, at day 3 and 7 post-ligation. In response to femoral artery ligation, collateral lumen area was significantly reduced in normoxic adductor muscles. Distally, ischemic gastrocnemius muscles displayed impaired perfusion recovery and angiogenesis paralleled with persistent inflammation. Muscle-specific mRNA sequencing revealed differential expression of genes critical for smooth muscle proliferation and sprouting angiogenesis in normoxic adductor and ischemic gastrocnemius, respectively, at day 7 post-ligation. Genes typical for macrophage (Mϕ) subsets were differentially expressed across both muscle types. Cell-specific gene expression, flow cytometry, and immunohistochemistry revealed persistent IFN-I response gene upregulation in arterial endothelial cells, ECs and Mϕs from T2DM mice associated with impaired collateral remodeling, angiogenesis and perfusion recovery. Furthermore, rHDL nanotherapy rescued impaired collateral remodeling and angiogenesis through dampening EC and Mϕ inflammation in T2DM mice. CONCLUSIONS: Our results suggest that an impaired collateral remodeling and sprouting angiogenesis in T2DM mice is associated with persistent IFN-I response in ECs and Mϕs. Dampening persistent inflammation and skewing ECs and Mϕ phenotype toward less inflammatory ones using rHDL nanotherapy may serve as a potential therapeutic target for T2DM peripheral arterial disease.


Subject(s)
Diabetes Mellitus, Experimental , Diabetes Mellitus, Type 2 , Peripheral Arterial Disease , Mice , Animals , Neovascularization, Physiologic , Endothelial Cells/metabolism , Apolipoprotein A-I/metabolism , Macrophages/metabolism , Ischemia , Muscle, Skeletal/blood supply , Femoral Artery/metabolism , Diabetes Mellitus, Type 2/genetics , Inflammation/metabolism , Peripheral Arterial Disease/metabolism , Phenotype , Hindlimb/blood supply , Mice, Inbred C57BL , Collateral Circulation
10.
Biol Pharm Bull ; 47(4): 868-871, 2024.
Article in English | MEDLINE | ID: mdl-38644197

ABSTRACT

Restoration of blood flow in skeletal muscle after a prolonged period of ischemia induces muscular ischemia-reperfusion injury, leading to local injury/dysfunction in muscles followed by systemic inflammatory responses. However, preventive/curative agents for skeletal muscle ischemia injury are unavailable in clinics to date. Increasing evidence has validated that carbon monoxide (CO) prevents the progression of ischemia-reperfusion injury in various organs owing to its versatile bioactivity. Previously, we developed a bioinspired CO donor, CO-bound red blood cells (CO-RBC), which mimics the dynamics of RBC-associated CO in the body. In the present study, we have tested the therapeutic potential of CO-RBC in muscular injury/dysfunction and secondary systemic inflammation induced by skeletal muscle ischemia-reperfusion. The results indicate that CO-RBC rather than RBC alone suppressed elevation of plasma creatine phosphokinase, a marker of muscular injury, in rats subjected to both hind limbs ischemia-reperfusion. In addition, the results of the treadmill walking test revealed a significantly decreased muscular motor function in RBC-treated rats subjected to both hind limbs ischemia-reperfusion than that in healthy rats, however, CO-RBC treatment facilitated sustained muscular motor functions after hind limbs ischemia-reperfusion. Furthermore, CO-RBC rather than RBC suppressed the production of tumour necrosis factor (TNF)-α and interleukin (IL)-6, which were upregulated by muscular ischemia-reperfusion. Interestingly, CO-RBC treatment induced higher levels of IL-10 compared to saline or RBC treatments. Based on these findings, we suggest that CO-RBC exhibits a suppressive effect against skeletal muscle injury/dysfunction and systemic inflammatory responses after skeletal muscle ischemia-reperfusion.


Subject(s)
Carbon Monoxide , Inflammation , Muscle, Skeletal , Rats, Sprague-Dawley , Reperfusion Injury , Animals , Reperfusion Injury/drug therapy , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Male , Inflammation/drug therapy , Erythrocytes/drug effects , Erythrocytes/metabolism , Rats , Creatine Kinase/blood , Hindlimb/blood supply , Tumor Necrosis Factor-alpha/metabolism , Tumor Necrosis Factor-alpha/blood , Interleukin-6/metabolism , Interleukin-6/blood
11.
Cryobiology ; 116: 104950, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39134131

ABSTRACT

Ischemia is a major limiting factor in Vascularized Composite Allotransplantation (VCA) as irreversible muscular injury can occur after as early as 4-6 h of static cold storage (SCS). Organ preservation technologies have led to the development of storage protocols extending rat liver ex vivo preservation up to 4 days. Development of such a protocol for VCAs has the added challenge of inherent ice nucleating factors of the graft, therefore, this study focused on developing a robust protocol for VCA supercooling. Rodent partial hindlimbs underwent subnormothermic machine perfusion (SNMP) with several loading solutions, followed by a storage solution with cryoprotective agents (CPA) developed for VCAs. Storage occurred in suspended animation for 24h and VCAs were recovered using SNMP with modified Steen. This study shows a robust VCA supercooling preservation protocol in a rodent model. Further optimization is expected to allow for its application in a transplantation model, which would be a breakthrough in the field of VCA preservation.


Subject(s)
Cryopreservation , Cryoprotective Agents , Hindlimb , Organ Preservation , Vascularized Composite Allotransplantation , Animals , Rats , Hindlimb/blood supply , Male , Vascularized Composite Allotransplantation/methods , Cryopreservation/methods , Organ Preservation/methods , Cryoprotective Agents/pharmacology , Organ Preservation Solutions/pharmacology , Perfusion/methods , Composite Tissue Allografts
12.
Proc Natl Acad Sci U S A ; 118(28)2021 07 13.
Article in English | MEDLINE | ID: mdl-34244428

ABSTRACT

The emerging field of regenerative cell therapy is still limited by the few cell types that can reliably be differentiated from pluripotent stem cells and by the immune hurdle of commercially scalable allogeneic cell therapeutics. Here, we show that gene-edited, immune-evasive cell grafts can survive and successfully treat diseases in immunocompetent, fully allogeneic recipients. Transplanted endothelial cells improved perfusion and increased the likelihood of limb preservation in mice with critical limb ischemia. Endothelial cell grafts transduced to express a transgene for alpha1-antitrypsin (A1AT) successfully restored physiologic A1AT serum levels in mice with genetic A1AT deficiency. This cell therapy prevented both structural and functional changes of emphysematous lung disease. A mixture of endothelial cells and cardiomyocytes was injected into infarcted mouse hearts, and both cell types orthotopically engrafted in the ischemic areas. Cell therapy led to an improvement in invasive hemodynamic heart failure parameters. Our study supports the development of hypoimmune, universal regenerative cell therapeutics for cost-effective treatments of major diseases.


Subject(s)
Cardiovascular Diseases/immunology , Cardiovascular Diseases/therapy , Immunocompetence , Induced Pluripotent Stem Cells/immunology , Lung Diseases/immunology , Lung Diseases/therapy , Stem Cell Transplantation , Animals , Endothelial Cells/transplantation , Heart Failure/therapy , Hindlimb/blood supply , Hindlimb/pathology , Ischemia/pathology , Mice, Inbred BALB C , Mice, Inbred C57BL , Myocytes, Cardiac/transplantation , Transplantation, Homologous , alpha 1-Antitrypsin/metabolism
13.
Proc Natl Acad Sci U S A ; 118(51)2021 12 21.
Article in English | MEDLINE | ID: mdl-34911761

ABSTRACT

Arterial remodeling is an important adaptive mechanism that maintains normal fluid shear stress in a variety of physiologic and pathologic conditions. Inward remodeling, a process that leads to reduction in arterial diameter, plays a critical role in progression of such common diseases as hypertension and atherosclerosis. Yet, despite its pathogenic importance, molecular mechanisms controlling inward remodeling remain undefined. Mitogen-activated protein kinases (MAPKs) perform a number of functions ranging from control of proliferation to migration and cell-fate transitions. While the MAPK ERK1/2 signaling pathway has been extensively examined in the endothelium, less is known about the role of the MEKK3/ERK5 pathway in vascular remodeling. To better define the role played by this signaling cascade, we studied the effect of endothelial-specific deletion of its key upstream MAP3K, MEKK3, in adult mice. The gene's deletion resulted in a gradual inward remodeling of both pulmonary and systematic arteries, leading to spontaneous hypertension in both vascular circuits and accelerated progression of atherosclerosis in hyperlipidemic mice. Molecular analysis revealed activation of TGFß-signaling both in vitro and in vivo. Endothelial-specific TGFßR1 knockout prevented inward arterial remodeling in MEKK3 endothelial knockout mice. These data point to the unexpected participation of endothelial MEKK3 in regulation of TGFßR1-Smad2/3 signaling and inward arterial remodeling in artery diseases.


Subject(s)
Hypertension, Pulmonary/pathology , MAP Kinase Kinase Kinase 1/metabolism , MAP Kinase Kinase Kinase 3/metabolism , Transforming Growth Factor beta/metabolism , Vascular Remodeling/physiology , Animals , Gene Deletion , Gene Expression Regulation/drug effects , Genotype , Hindlimb/blood supply , Human Umbilical Vein Endothelial Cells , Humans , Hypertension, Pulmonary/metabolism , Ischemia , MAP Kinase Kinase Kinase 1/genetics , MAP Kinase Kinase Kinase 3/genetics , Mice , Receptors, Transforming Growth Factor beta/genetics , Receptors, Transforming Growth Factor beta/metabolism , Selective Estrogen Receptor Modulators/toxicity , Signal Transduction , Tamoxifen/toxicity , Transforming Growth Factor beta/genetics
14.
Int J Mol Sci ; 25(6)2024 Mar 15.
Article in English | MEDLINE | ID: mdl-38542330

ABSTRACT

Angiogenesis is a critical physiological response to ischemia but becomes pathological when dysregulated and driven excessively by inflammation. We recently identified a novel angiogenic role for tripartite-motif-containing protein 2 (TRIM2) whereby lentiviral shRNA-mediated TRIM2 knockdown impaired endothelial angiogenic functions in vitro. This study sought to determine whether these effects could be translated in vivo and to determine the molecular mechanisms involved. CRISPR/Cas9-generated Trim2-/- mice that underwent a periarterial collar model of inflammation-induced angiogenesis exhibited significantly less adventitial macrophage infiltration relative to wildtype (WT) littermates, concomitant with decreased mRNA expression of macrophage marker Cd68 and reduced adventitial proliferating neovessels. Mechanistically, TRIM2 knockdown in endothelial cells in vitro attenuated inflammation-driven induction of critical angiogenic mediators, including nuclear HIF-1α, and curbed the phosphorylation of downstream effector eNOS. Conversely, in a hindlimb ischemia model of hypoxia-mediated angiogenesis, there were no differences in blood flow reperfusion to the ischemic hindlimbs of Trim2-/- and WT mice despite a decrease in proliferating neovessels and arterioles. TRIM2 knockdown in vitro attenuated hypoxia-driven induction of nuclear HIF-1α but had no further downstream effects on other angiogenic proteins. Our study has implications for understanding the role of TRIM2 in the regulation of angiogenesis in both pathophysiological contexts.


Subject(s)
Angiogenesis , Endothelial Cells , Animals , Mice , Endothelial Cells/metabolism , Hindlimb/blood supply , Hypoxia/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Inflammation/metabolism , Ischemia/metabolism , Neovascularization, Pathologic/metabolism , Neovascularization, Physiologic/genetics
15.
J Avian Med Surg ; 38(2): 91-97, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38980818

ABSTRACT

Intravenous regional limb perfusion (IVRLP) has been used in the treatment of pododermatitis and distal limb infections, which are significant causes of morbidity in avian species. This intravenous drug administration technique is designed to achieve high drug tissue concentrations while minimizing systemic toxic effects. Amikacin is commonly used for IVRLP in veterinary medicine, but dosing guidelines have not been established for its use in birds. The current study aimed to determine the tissue concentration of amikacin after a single IVRLP administration in healthy, euhydrated leghorn hen chickens (Gallus gallus domesticus). Chickens received a single IVRLP dose of 10 mg/kg amikacin and were euthanatized posttreatment at 1 hour (n = 6), 12 hours (n = 6), and 24 hours (n = 6) to assess tissue and synovial fluid concentrations of amikacin in the injected leg. Mean tissue concentrations were highest 1 hour post-IVRLP (synovial fluid = 153.0 µg/mL, metatarsal pad tissue = 26.05 µg/mL) before declining at the 12- and 24-hour time points. This indicates that administration of amikacin via IVRLP can reach minimum inhibitory concentrations of common bacterial isolates in tissues after a single treatment with 10 mg/kg amikacin. Regional limb perfusion every 24 hours is recommended, although the minimum days of treatment may be case dependent and vary based on response to therapy.


Subject(s)
Amikacin , Anti-Bacterial Agents , Chickens , Animals , Amikacin/pharmacokinetics , Amikacin/administration & dosage , Anti-Bacterial Agents/pharmacokinetics , Anti-Bacterial Agents/administration & dosage , Synovial Fluid/chemistry , Perfusion/veterinary , Female , Hindlimb/blood supply
16.
Bull Exp Biol Med ; 177(1): 26-29, 2024 May.
Article in English | MEDLINE | ID: mdl-38954303

ABSTRACT

We present a two-stage model for the study of chronic hind limb ischemia in rats. In the area of ischemia, sclerotic changes with atrophic rhabdomyocytes and reduced vascularization were revealed. CD31 expression in the endothelium increased proportionally to the number of vessels in the ischemic zone, and at the same time, focal expression of ßIII-tubulin was detected in the newly formed nerve fibers. These histological features are equivalent to the development of peripheral arterial disease in humans, which allows using our model in the search for new therapeutic strategies.


Subject(s)
Disease Models, Animal , Hindlimb , Ischemia , Muscle, Skeletal , Animals , Rats , Muscle, Skeletal/pathology , Muscle, Skeletal/metabolism , Muscle, Skeletal/blood supply , Hindlimb/blood supply , Hindlimb/pathology , Ischemia/pathology , Ischemia/metabolism , Ischemia/physiopathology , Male , Rats, Wistar , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Tubulin/metabolism , Peripheral Arterial Disease/pathology , Peripheral Arterial Disease/metabolism , Peripheral Arterial Disease/physiopathology
17.
Angiogenesis ; 26(3): 385-407, 2023 08.
Article in English | MEDLINE | ID: mdl-36933174

ABSTRACT

The molecular mechanisms of angiogenesis have been intensely studied, but many genes that control endothelial behavior and fate still need to be described. Here, we characterize the role of Apold1 (Apolipoprotein L domain containing 1) in angiogenesis in vivo and in vitro. Single-cell analyses reveal that - across tissues - the expression of Apold1 is restricted to the vasculature and that Apold1 expression in endothelial cells (ECs) is highly sensitive to environmental factors. Using Apold1-/- mice, we find that Apold1 is dispensable for development and does not affect postnatal retinal angiogenesis nor alters the vascular network in adult brain and muscle. However, when exposed to ischemic conditions following photothrombotic stroke as well as femoral artery ligation, Apold1-/- mice display dramatic impairments in recovery and revascularization. We also find that human tumor endothelial cells express strikingly higher levels of Apold1 and that Apold1 deletion in mice stunts the growth of subcutaneous B16 melanoma tumors, which have smaller and poorly perfused vessels. Mechanistically, Apold1 is activated in ECs upon growth factor stimulation as well as in hypoxia, and Apold1 intrinsically controls EC proliferation but not migration. Our data demonstrate that Apold1 is a key regulator of angiogenesis in pathological settings, whereas it does not affect developmental angiogenesis, thus making it a promising candidate for clinical investigation.


Subject(s)
Endothelial Cells , Neovascularization, Physiologic , Animals , Humans , Mice , Endothelial Cells/metabolism , Hindlimb/blood supply , Hypoxia/metabolism , Ischemia/pathology , Mice, Inbred C57BL , Mice, Knockout , Neovascularization, Physiologic/genetics , Immediate-Early Proteins/metabolism
18.
J Vasc Res ; 60(3): 148-159, 2023.
Article in English | MEDLINE | ID: mdl-37336198

ABSTRACT

INTRODUCTION: Peripheral arterial disease (PAD) occurs from atherosclerotic obstruction of arteries in the lower extremities. Restoration of perfusion requires angiogenesis and arteriogenesis through migration and differentiation of endothelial progenitor cells (EPCs) and macrophages at the site of injury. The time of recruitment has not been fully investigated. In this study, we investigated the infiltration of these cells in murine hind limb ischemia (HLI) model of PAD. METHODS: EPCs and M1-like and M2-like macrophages from ischemic skeletal muscles were quantified by flow cytometry at day-0, 1, 3, 7, and 14 post-HLI. RESULTS: The abundance of EPCs increased from day 1 and was highest on day 7 until day 14. M1-like population similarly increased and was highest on day 14 during the experiment. M2-like population was significantly greater than M1-like at baseline but surpassed the highest value of M1-like by day 7 during the experiment. Muscle regeneration and capillary density also increased and were highest at days 3 and 7, respectively, during the experiment. All mice achieved near full perfusion recovery by day 14. CONCLUSION: Thus, we observed a gradual increase in the percentage of EPC's and this was temporally paralleled with initial increase in M1-like followed by sustained increased in M2-like macrophages and perfusion recovered post-HLI.


Subject(s)
Endothelial Progenitor Cells , Peripheral Arterial Disease , Mice , Animals , Ischemia , Arteries , Hindlimb/blood supply , Macrophages , Muscle, Skeletal/blood supply , Neovascularization, Physiologic , Disease Models, Animal , Mice, Inbred C57BL
19.
Microvasc Res ; 145: 104425, 2023 01.
Article in English | MEDLINE | ID: mdl-36089076

ABSTRACT

BACKGROUND: The simulation of limb ischemia in large laboratory animals is a complex and currently topical task in experimental medicine. Meanwhile, there is a demand for a reliable and effective model of limb ischemia for further testing of medicines to stimulate circulation and induce angiogenesis, gene medicines in particular. Aim of this study was to develop and experimentally test an effective method of simulation of hind limb ischemia. METHODS: Female Vietnamese pot-bellied pigs were chosen as biological models. The reproduction of the pathology was evaluated using the following methods: laser doppler flowmetry, laboratory test of venous blood, immunohistochemical reaction with antibodies against CD31, a specific marker of endothelial cells, Van Gieson's staining of muscles for presence of connective tissue and clinical observation to detect the presence of lameness in pigs. RESULTS: Laser doppler flowmetry recorded a significant decrease in the intensity of the blood circulation and a marked decrease in temperate in the operated limb. Increased lactate and creatine kinase were registered immediately after the surgery and were absent 3 or more days later. Clinical observation demonstrated presence of walking lameness. Histological and immunohistochemical methods revealed a credible increase in connective tissue area and a reduction in the number of blood vessels in the muscles, confirming the presence of ischemia. CONCLUSIONS: An effective approach to modeling limb ischemia has been developed and experimentally tested. The proposed model may be used in cardiovascular surgery and will allow further testing of new medications designed to treat ischemia of hind limbs.


Subject(s)
Endothelial Cells , Peripheral Vascular Diseases , Female , Swine , Animals , Endothelial Cells/pathology , Lameness, Animal , Disease Models, Animal , Ischemia , Hindlimb/blood supply , Neovascularization, Physiologic
SELECTION OF CITATIONS
SEARCH DETAIL