Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 6 de 6
1.
Am J Nephrol ; 51(11): 891-902, 2020.
Article En | MEDLINE | ID: mdl-33130676

BACKGROUND: Chronic renovascular disease (RVD) can lead to a progressive loss of renal function, and current treatments are inefficient. We designed a fusion of vascular endothelial growth factor (VEGF) conjugated to an elastin-like polypeptide (ELP) carrier protein with an N-terminal kidney-targeting peptide (KTP). We tested the hypothesis that KTP-ELP-VEGF therapy will effectively recover renal function with an improved targeting profile. Further, we aimed to elucidate potential mechanisms driving renal recovery. METHODS: Unilateral RVD was induced in 14 pigs. Six weeks later, renal blood flow (RBF) and glomerular filtration rate (GFR) were quantified by multidetector CT imaging. Pigs then received a single intrarenal injection of KTP-ELP-VEGF or vehicle. CT quantification of renal hemodynamics was repeated 4 weeks later, and then pigs were euthanized. Ex vivo renal microvascular (MV) density and media-to-lumen ratio, macrophage infiltration, and fibrosis were quantified. In parallel, THP-1 human monocytes were differentiated into naïve macrophages (M0) or inflammatory macrophages (M1) and incubated with VEGF, KTP-ELP, KTP-ELP-VEGF, or control media. The mRNA expression of macrophage polarization and angiogenic markers was quantified (qPCR). RESULTS: Intrarenal KTP-ELP-VEGF improved RBF, GFR, and MV density and attenuated MV media-to-lumen ratio and renal fibrosis compared to placebo, accompanied by augmented renal M2 macrophages. In vitro, exposure to VEGF/KTP-ELP-VEGF shifted M0 macrophages to a proangiogenic M2 phenotype while M1s were nonresponsive to VEGF treatment. CONCLUSIONS: Our results support the efficacy of a new renal-specific biologic construct in recovering renal function and suggest that VEGF may directly influence macrophage phenotype as a possible mechanism to improve MV integrity and function in the stenotic kidney.


Recombinant Fusion Proteins/administration & dosage , Recovery of Function/drug effects , Renal Artery Obstruction/drug therapy , Vascular Endothelial Growth Factor A/administration & dosage , Animals , Atherosclerosis/complications , Atherosclerosis/drug therapy , Disease Models, Animal , Elastin/administration & dosage , Elastin/genetics , Female , Glomerular Filtration Rate/drug effects , Glomerular Filtration Rate/physiology , Humans , Kidney/blood supply , Kidney/drug effects , Kidney/physiology , Male , Microvessels/drug effects , Microvessels/physiology , Peptides/administration & dosage , Peptides/genetics , Recombinant Fusion Proteins/genetics , Renal Artery Obstruction/etiology , Renal Circulation/drug effects , Sus scrofa , Vascular Endothelial Growth Factor A/genetics
2.
Am J Physiol Renal Physiol ; 319(1): F139-F148, 2020 07 01.
Article En | MEDLINE | ID: mdl-32538151

Inflammation is a major determinant for the progression of chronic kidney disease (CKD). NF-κB is a master transcription factor upregulated in CKD that promotes inflammation and regulates apoptosis and vascular remodeling. We aimed to modulate this pathway for CKD therapy in a swine model of CKD using a peptide inhibitor of the NF-κB p50 subunit (p50i) fused to a protein carrier [elastin-like polypeptide (ELP)] and equipped with a cell-penetrating peptide (SynB1). We hypothesized that intrarenal SynB1-ELP-p50i therapy would inhibit NF-κB-driven inflammation and induce renal recovery. CKD was induced in 14 pigs. After 6 wk, pigs received single intrarenal SynB1-ELP-p50i therapy (10 mg/kg) or placebo (n = 7 each). Renal hemodynamics were quantified in vivo using multidetector computed tomography before and 8 wk after treatment. Pigs were then euthanized. Ex vivo experiments were performed to quantify renal activation of NF-κB, expression of downstream mediators of NF-κB signaling, renal microvascular density, inflammation, and fibrosis. Fourteen weeks of CKD stimulated NF-κB signaling and downstream mediators (e.g., TNF-α, monocyte chemoattractant protein-1, and IL-6) accompanying loss of renal function, inflammation, fibrosis, and microvascular rarefaction versus controls. All of these were improved after SynB1-ELP-p50i therapy, accompanied by reduced circulating inflammatory cytokines as well, which were evident up to 8 wk after treatment. Current treatments for CKD are largely ineffective. Our study shows the feasibility of a new treatment to induce renal recovery by offsetting inflammation at a molecular level. It also supports the therapeutic potential of targeted inhibition of the NF-κB pathway using novel drug delivery technology in a translational model of CKD.


Kidney/drug effects , NF-kappa B/antagonists & inhibitors , Renal Insufficiency, Chronic/drug therapy , Animals , Disease Models, Animal , Drug Delivery Systems , Fibrosis/drug therapy , Fibrosis/metabolism , Fibrosis/pathology , Kidney/metabolism , Kidney/pathology , Renal Circulation/drug effects , Renal Insufficiency, Chronic/metabolism , Renal Insufficiency, Chronic/pathology , Signal Transduction/drug effects , Swine
3.
Hypertension ; 74(5): 1113-1123, 2019 11.
Article En | MEDLINE | ID: mdl-31542966

Chronic kidney disease (CKD) universally associates with renal microvascular rarefaction and inflammation, but whether a link exists between these 2 processes is unclear. We designed a therapeutic construct of VEGF (vascular endothelial growth factor) fused to an ELP (elastin-like polypeptide) carrier and show that it improves renal function in experimental renovascular disease. We test the hypothesis that ELP-VEGF therapy will improve CKD, and that recovery will be driven by decreasing microvascular rarefaction partly via modulation of macrophage phenotype and inflammation. CKD was induced in 14 pigs, which were observed for 14 weeks. At 6 weeks, renal blood flow and filtration were quantified using multidetector computed tomography, and then pigs received single intrarenal ELP-VEGF or placebo (n=7 each). Renal function was quantified again 4 and 8 weeks later. Pigs were euthanized and renal microvascular density, angiogenic and inflammatory markers, fibrosis, macrophage infiltration, and phenotype were quantified. Loss of renal hemodynamics in CKD was progressively recovered by ELP-VEGF therapy, accompanied by improved renal microvascular density, fibrosis, and expression of inflammatory mediators. Although renal macrophage infiltration was similar in both CKD groups, ELP-VEGF therapy distinctly shifted their phenotype from proinflammatory M1 to VEGF-expressing M2. Our study unravels potential mechanisms and feasibility of a new strategy to offset progression of CKD using drug-delivery technologies. The results indicate that renal recovery after ELP-VEGF therapy was largely driven by modulation of renal macrophages toward VEGF-expressing M2 phenotype, restoring VEGF signaling and sustaining improvement of renal function and microvascular integrity in CKD.


Elastin/pharmacology , Macrophages/drug effects , Renal Circulation/drug effects , Renal Insufficiency, Chronic/drug therapy , Vascular Endothelial Growth Factor A/pharmacology , Animals , Biopsy, Needle , Cells, Cultured , Disease Models, Animal , Drug Carriers , Immunohistochemistry , Injections, Intralesional , Kidney Function Tests , Macrophages/cytology , Microcirculation/drug effects , Multidetector Computed Tomography/methods , Random Allocation , Recovery of Function , Renal Insufficiency, Chronic/diagnostic imaging , Renal Insufficiency, Chronic/pathology , Sensitivity and Specificity , Sus scrofa , Tissue and Organ Harvesting , Treatment Outcome
4.
Am J Physiol Renal Physiol ; 316(5): F1016-F1025, 2019 05 01.
Article En | MEDLINE | ID: mdl-30892933

Renal angioplasty and stenting (PTRAs) resolves renal artery stenosis, but inconsistently improves renal function, possibly due to persistent parenchymal damage. We developed a bioengineered fusion of a drug delivery vector (elastin-like polypeptide, ELP) with vascular endothelial growth factor (VEGF), and showed its therapeutic efficacy. We tested the hypothesis that combined ELP-VEGF therapy with PTRAs improves renal recovery more efficiently than PTRAs alone, by protecting the stenotic renal parenchyma. Unilateral renovascular disease (RVD) was induced by renal artery stenosis in 14 pigs. Six weeks later, stenotic kidney blood flow (RBF) and glomerular filtration rate (GFR) were quantified in vivo using multidetector CT. Blood and urine were collected during in vivo studies. All pigs underwent PTRAs and then were randomized into single intrarenal ELP-VEGF administration or placebo (n = 7 each) groups. Pigs were observed for four additional weeks, in vivo CT studies were repeated, and then pigs were euthanized for ex vivo studies to quantify renal microvascular (MV) density, angiogenic factor expression, and morphometric analysis. Renal hemodynamics were similarly blunted in all RVD pigs. PTRAs resolved stenosis but modestly improved RBF and GFR. However, combined PTRAs+ ELP-VEGF improved RBF, GFR, regional perfusion, plasma creatinine, asymmetric dimethlyarginine (ADMA), and albuminuria compared with PTRAs alone, accompanied by improved angiogenic signaling, MV density, and renal fibrosis. Greater improvement of renal function via coadjuvant ELP-VEGF therapy may be driven by enhanced MV proliferation and repair, which ameliorates MV rarefaction and fibrogenic activity that PTRAs alone cannot offset. Thus, our study supports a novel strategy to boost renal recovery in RVD after PTRAs.


Angiogenesis Inducing Agents/administration & dosage , Angioplasty, Balloon , Hemodynamics/drug effects , Kidney/blood supply , Peptides/administration & dosage , Renal Artery Obstruction/therapy , Renal Circulation/drug effects , Vascular Endothelial Growth Factor A/administration & dosage , Vascular Remodeling/drug effects , Angioplasty, Balloon/instrumentation , Animals , Disease Models, Animal , Fibrosis , Glomerular Filtration Rate/drug effects , Humans , Kidney/pathology , Recombinant Fusion Proteins/administration & dosage , Renal Artery Obstruction/diagnostic imaging , Renal Artery Obstruction/pathology , Renal Artery Obstruction/physiopathology , Stents , Sus scrofa
5.
Am J Physiol Renal Physiol ; 315(2): F364-F373, 2018 08 01.
Article En | MEDLINE | ID: mdl-29693449

Animal models of chronic kidney disease (CKD) are critical for understanding its pathophysiology and for therapeutic development. The cardiovascular and renal anatomy and physiology of the pig are virtually identical to humans. This study aimed to develop a novel translational model of CKD that mimics the pathological features of CKD in humans. CKD was induced in seven domestic pigs by bilateral renal artery stenosis and diet-induced dyslipidemia. Animals were observed for a total of 14 wk. Renal hemodynamics and function were quantified in vivo using multi-detector CT after 6, 10, and 14 wk of CKD. Urine and blood were collected at each time-point, and blood pressure was continuously measured (telemetry). After completion of in vivo studies, pigs were euthanized, kidneys were removed, and microvascular (MV) architecture (µCT), markers of renal injury, inflammation, and fibrosis were evaluated ex vivo. Additional pigs were used as controls ( n = 7). Renal blood flow and glomerular filtration were reduced by 50% in CKD, accompanied by hypertension and elevated plasma creatinine, albumin-to-creatinine ratio and increased urinary KIM-1 and NGAL, suggesting renal injury. Furthermore, 14 wk of CKD resulted in cortical and medullary MV remodeling and loss, inflammation, glomerulosclerosis, tubular atrophy, and tubule-interstitial fibrosis compared with controls. The current study characterizes a novel model of CKD that mimics several of the pathological features observed in human CKD, irrespective of the etiology. Current approaches only slow rather than halt CKD progression, and this novel model may offer a suitable platform for the development of new treatments in a translational fashion.


Kidney , Renal Insufficiency, Chronic , Translational Research, Biomedical/methods , Animals , Biomarkers/blood , Biomarkers/urine , Diet, High-Fat , Disease Models, Animal , Disease Progression , Dyslipidemias/complications , Female , Glomerular Filtration Rate , Hemodynamics , Humans , Kidney/blood supply , Kidney/metabolism , Kidney/pathology , Renal Artery Obstruction/complications , Renal Circulation , Renal Insufficiency, Chronic/etiology , Renal Insufficiency, Chronic/metabolism , Renal Insufficiency, Chronic/pathology , Renal Insufficiency, Chronic/physiopathology , Species Specificity , Sus scrofa , Time Factors
6.
Kidney Int ; 93(4): 842-854, 2018 04.
Article En | MEDLINE | ID: mdl-29273331

We recently developed a therapeutic biopolymer composed of an elastin-like polypeptide (ELP) fused to vascular endothelial growth factor (VEGF) and showed long-term renoprotective effects in experimental renovascular disease after a single intra-renal administration. Here, we sought to determine the specificity, safety, efficacy, and mechanisms of renoprotection of ELP-VEGF after systemic therapy in renovascular disease. We tested whether kidney selectivity of the ELP carrier would reduce off-target binding of VEGF in other organs. In vivo bio-distribution after systemic administration of ELP-VEGF in swine was determined in kidneys, liver, spleen, and heart. Stenotic-kidney renal blood flow and glomerular filtration rate were quantified in vivo using multi-detector computed tomography (CT) after six weeks of renovascular disease, then treated with a single intravenous dose of ELP-VEGF or placebo and observed for four weeks. CT studies were then repeated and the pigs euthanized. Ex vivo studies quantified renal microvascular density (micro-CT) and fibrosis. Kidneys, liver, spleen, and heart were excised to quantify the expression of angiogenic mediators and markers of progenitor cells. ELP-VEGF accumulated predominantly in the kidney and stimulated renal blood flow, glomerular filtration rate, improved cortical microvascular density, and renal fibrosis, and was accompanied by enhanced renal expression of VEGF, downstream mediators of VEGF signaling, and markers of progenitor cells compared to placebo. Expression of angiogenic factors in liver, spleen, and heart were not different compared to placebo-control. Thus, ELP efficiently directs VEGF to the kidney after systemic administration and induces long-term renoprotection without off-target effects, supporting the feasibility and safety of renal therapeutic angiogenesis via systemic administration of a novel kidney-specific bioengineered compound.


Angiogenesis Inducing Agents/pharmacology , Kidney/blood supply , Kidney/drug effects , Neovascularization, Physiologic/drug effects , Peptides/pharmacology , Renal Artery Obstruction/drug therapy , Vascular Endothelial Growth Factor A/pharmacology , Angiogenesis Inducing Agents/administration & dosage , Angiogenesis Inducing Agents/pharmacokinetics , Angiogenesis Inducing Agents/toxicity , Animals , Apoptosis/drug effects , Biomarkers/metabolism , Disease Models, Animal , Drug Carriers , Fibrosis , Glomerular Filtration Rate/drug effects , Injections, Intravenous , Kidney/metabolism , Kidney/pathology , Peptides/administration & dosage , Peptides/pharmacokinetics , Peptides/toxicity , Recombinant Fusion Proteins/pharmacology , Renal Artery Obstruction/metabolism , Renal Artery Obstruction/pathology , Renal Artery Obstruction/physiopathology , Renal Circulation/drug effects , Stem Cells/drug effects , Stem Cells/metabolism , Sus scrofa , Tissue Distribution , Vascular Endothelial Growth Factor A/administration & dosage , Vascular Endothelial Growth Factor A/pharmacokinetics , Vascular Endothelial Growth Factor A/toxicity
...