Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 260
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Mol Pharm ; 21(7): 3310-3320, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38856116

ABSTRACT

Siponimod is a promising agent for the inhibition of ocular neovascularization in diabetic retinopathy and age-related macular degeneration. Siponimod's development for ophthalmological application is hindered by the limited information available on the drug's solubility, stability, ocular pharmacokinetics (PK), and toxicity in vivo. In this study, we investigated the aqueous stability of siponimod under stress conditions (up to 60 °C) and its degradation behavior in solution. Additionally, siponimod's ocular PK and toxicity were investigated using intravitreal injection of two different doses (either 1300 or 6500 ng) in an albino rabbit model. Siponimod concentration was quantified in the extracted vitreous, and the PK parameters were calculated. The drug half-life after administration of the low and high doses was 2.8 and 3.9 h, respectively. The data obtained in vivo was used to test the ability of published in silico models to predict siponimod's PK accurately. Two models that correlated siponimod's molecular descriptors with its elimination from the vitreous closely predicted the half-life. Furthermore, 24 h and 7 days after intravitreal injections, the retinas showed no signs of toxicity. This study provides important information necessary for the formulation and development of siponimod for ophthalmologic applications. The short half-life of siponimod necessitates the development of a sustained drug delivery system to maintain therapeutic concentrations over an extended period, while the lack of short-term ocular toxicity observed in the retinas of siponimod-treated rabbits supports possible clinical use.


Subject(s)
Azetidines , Intravitreal Injections , Animals , Rabbits , Azetidines/pharmacokinetics , Azetidines/administration & dosage , Half-Life , Vitreous Body/drug effects , Vitreous Body/metabolism , Male , Retina/drug effects , Retina/metabolism , Eye/drug effects , Eye/metabolism , Diabetic Retinopathy/drug therapy , Angiogenesis Inhibitors/pharmacokinetics , Angiogenesis Inhibitors/administration & dosage , Angiogenesis Inhibitors/toxicity , Solubility , Macular Degeneration/drug therapy , Benzyl Compounds
2.
Arch Toxicol ; 98(3): 943-956, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38285066

ABSTRACT

Angiogenesis is a key process in embryonic development, a disruption of this process can lead to severe developmental defects, such as limb malformations. The identification of molecular perturbations representative of antiangiogenesis in zebrafish embryo (ZFE) may guide the assessment of developmental toxicity from an endpoint- to a mechanism-based approach, thereby improving the extrapolation of findings to humans. Thus, the aim of the study was to discover molecular changes characteristic of antiangiogenesis and developmental toxicity. We exposed ZFEs to two antiangiogenic drugs (SU4312, sorafenib) and two developmental toxicants (methotrexate, rotenone) with putative antiangiogenic action. Molecular changes were measured by performing untargeted metabolomics in single embryos. The metabolome response was accompanied by the occurrence of morphological alterations. Two distinct metabolic effect patterns were observed. The first pattern comprised common effects of two specific angiogenesis inhibitors and the known teratogen methotrexate, strongly suggesting a shared mode of action of antiangiogenesis and developmental toxicity. The second pattern involved joint effects of methotrexate and rotenone, likely related to disturbances in energy metabolism. The metabolites of the first pattern, such as phosphatidylserines, pterines, retinol, or coenzyme Q precursors, represented potential links to antiangiogenesis and related developmental toxicity. The metabolic effect pattern can contribute to biomarker identification for a mechanism-based toxicological testing.


Subject(s)
Angiogenesis Inhibitors , Zebrafish , Animals , Humans , Angiogenesis Inhibitors/toxicity , Angiogenesis Inhibitors/metabolism , Angiogenesis , Methotrexate/toxicity , Rotenone/pharmacology , Embryo, Nonmammalian , Metabolomics
3.
Toxicol Appl Pharmacol ; 450: 116162, 2022 09 01.
Article in English | MEDLINE | ID: mdl-35830948

ABSTRACT

Pathological angiogenesis is fundamental to progression of cancerous tumors and blinding eye diseases. Anti-angiogenic receptor tyrosine kinase inhibitors (TKIs) are in broad use for the treatment of these diseases. With more and more TKIs available, it is a challenge to make an optimal choice. It remains unclear whether TKIs demonstrate similar anti-angiogenesis activities in different tissues. Many TKIs have shown varying degrees of toxic effects that should also be considered in clinical use. This study investigates the anti-angiogenic effects of 13 FDA-approved TKIs on the intersegmental vessels (ISVs), subintestinal vessels (SIVs) and retinal vasculature in zebrafish embryos. The results show that vascular endothelial growth factor receptor TKIs (VEGFR-TKIs) exhibit anti-angiogenic abilities similarly on ISVs and SIVs, and their efficacy is consistent with their IC50 values against VEGFR2. In addition, VEGFR-TKIs selectively induces the apoptosis of endothelial cells in immature vessels. Among all TKIs tested, axitinib demonstrates a strong inhibition on retinal neovascularization at a low dose that do not strongly affect ISVs and SIVs, supporting its potential application for retinal diseases. Zebrafish embryos demonstrate cardiotoxicity after VEGFR-TKIs treatment, and ponatinib and sorafenib show a narrow therapeutic window, suggesting that these two drugs may need to be dosed more carefully in patients. We propose that zebrafish is an ideal model for studying in vivo antiangiogenic efficacy and cardiotoxicity of TKIs.


Subject(s)
Neoplasms , Zebrafish , Angiogenesis Inhibitors/therapeutic use , Angiogenesis Inhibitors/toxicity , Animals , Cardiotoxicity/drug therapy , Endothelial Cells/metabolism , Neoplasms/drug therapy , Protein Kinase Inhibitors/therapeutic use , Protein Kinase Inhibitors/toxicity , Vascular Endothelial Growth Factor A/metabolism , Zebrafish/metabolism
4.
Toxicol Appl Pharmacol ; 414: 115420, 2021 03 01.
Article in English | MEDLINE | ID: mdl-33503445

ABSTRACT

As a VEGF-targeting agent, sorafenib has been used to treat a number of solid tumors but can easily lead to adverse vascular effects. To elucidate the underlying mechanism, rat mesenteric arteries were subjected to organ cultured in the presence of different concentrations of sorafenib (0, 3, 6 and 9 mg/L) with or without inhibitors (U0126, 10-5 M; SB203580, 10-5 M; SP200126, 10-5 M) of MAPK kinases, and then acetylcholine- or sodium nitroprusside-induced vasodilation and sarafotoxin 6c-induced vasoconstriction were monitored by a sensitive myograph. The NO synthetases, the nitrite levels, the endothelial marker CD31,the ETB and ETA receptors and the phosphorylation of MAPK kinases were studied. Next, rats were orally administrated by sorafenib for 4 weeks (7.5 and 15 mg/kg/day), and their blood pressure, plasma ET-1, the ETB and ETA receptors and the phosphorylation of MAPK kinases in the mesenteric arteries were investigated. The results showed that sorafenib impairs endothelium-dependent vasodilation due to decreased NO levels and the low expression of eNOS and iNOS. Weak staining for CD31 indicated that sorafenib induced endothelial damage. Moreover, sorafenib caused the upregulation of vasoconstrictive ETB receptors, the enhancement of ETB receptor-mediated vasoconstriction and the activation of JNK/MAPK. Blocking the JNK, ERK1/2 and p38/MAPK signaling pathways by using the inhibitors significantly abolished ETB receptor-mediated vasoconstriction. Furthermore, it was observed that the oral administration of sorafenib caused an increase in blood pressure and plasma ET-1, upregulation of the ETB receptor and the activation of JNK in the mesenteric arteries. In conclusion, sorafenib not only impairs endothelium-dependent vasodilatation but also enhances ETB receptor-mediated vasoconstriction, which may be the causal factors for hypertension and other adverse vascular effects in patients treated with sorafenib.


Subject(s)
Angiogenesis Inhibitors/toxicity , Endothelium, Vascular/drug effects , Hypertension/chemically induced , Mesenteric Artery, Superior/drug effects , Receptor, Endothelin B/metabolism , Sorafenib/toxicity , Vasoconstriction/drug effects , Vasodilation/drug effects , Animals , Blood Pressure/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Endothelium, Vascular/metabolism , Endothelium, Vascular/physiopathology , Extracellular Signal-Regulated MAP Kinases/metabolism , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Hypertension/metabolism , Hypertension/physiopathology , JNK Mitogen-Activated Protein Kinases/metabolism , Male , Mesenteric Artery, Superior/metabolism , Mesenteric Artery, Superior/physiopathology , Nitric Oxide/metabolism , Rats, Sprague-Dawley , Receptor, Endothelin B/genetics , Signal Transduction , Tissue Culture Techniques , Up-Regulation , p38 Mitogen-Activated Protein Kinases/metabolism
5.
Am J Respir Crit Care Med ; 202(7): 983-995, 2020 10 01.
Article in English | MEDLINE | ID: mdl-32515984

ABSTRACT

Rationale: Endothelial injury may provoke emphysema, but molecular pathways of disease development require further discernment. Emphysematous lungs exhibit decreased expression of HIF-2α (hypoxia-inducible factor-2α)-regulated genes, and tobacco smoke decreases pulmonary HIF-2α concentrations. These findings suggest that decreased HIF-2α expression is important in the development of emphysema.Objectives: The objective of this study was to evaluate the roles of endothelial-cell (EC) HIF-2α in the pathogenesis of emphysema in mice.Methods: Mouse lungs were examined for emphysema after either the loss or the overexpression of EC Hif-2α. In addition, SU5416, a VEGFR2 inhibitor, was used to induce emphysema. Lungs were evaluated for HGF (hepatocyte growth factor), a protein involved in alveolar development and homeostasis. Lungs from patients with emphysema were measured for endothelial HIF-2α expression.Measurements and Main Results: EC Hif-2α deletion resulted in emphysema in association with fewer ECs and pericytes. After SU5416 exposure, EC Hif-2α-knockout mice developed more severe emphysema, whereas EC Hif-2α-overexpressing mice were protected. EC Hif-2α-knockout mice demonstrated lower levels of HGF. Human emphysema lung samples exhibited reduced EC HIF-2α expression.Conclusions: Here, we demonstrate a unique protective role for pulmonary endothelial HIF-2α and how decreased expression of this endogenous factor causes emphysema; its pivotal protective function is suggested by its ability to overcome VEGF antagonism. HIF-2α may maintain alveolar architecture by promoting vascular survival and associated HGF production. In summary, HIF-2α may be a key endogenous factor that prevents the development of emphysema, and its upregulation has the potential to foster lung health in at-risk patients.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Endothelial Cells/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Lung/metabolism , Pulmonary Emphysema/genetics , Angiogenesis Inhibitors/toxicity , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Deferoxamine/pharmacology , Disease Models, Animal , Endothelial Cells/pathology , Hepatocyte Growth Factor/genetics , Hepatocyte Growth Factor/metabolism , Humans , Indoles/toxicity , Iron Chelating Agents/pharmacology , Lung/blood supply , Lung/cytology , Lung/drug effects , Mice , Mice, Knockout , Microvessels , Pericytes/metabolism , Pulmonary Circulation , Pulmonary Disease, Chronic Obstructive/metabolism , Pulmonary Emphysema/chemically induced , Pulmonary Emphysema/metabolism , Pulmonary Emphysema/pathology , Pyrroles/toxicity , Smoke/adverse effects
6.
Circ Res ; 123(12): e51-e64, 2018 12 07.
Article in English | MEDLINE | ID: mdl-30566041

ABSTRACT

RATIONALE: Pulmonary arterial hypertension is a deadly disease of the pulmonary vasculature for which no disease-modifying therapies exist. Small-vessel stiffening and remodeling are fundamental pathological features of pulmonary arterial hypertension that occur early and drive further endovascular cell dysfunction. Bone marrow (BM)-derived proangiogenic cells (PACs), a specialized heterogeneous subpopulation of myeloid lineage cells, are thought to play an important role in pathogenesis. OBJECTIVE: To determine whether BM-derived PACs directly contributed to experimental pulmonary hypertension (PH) by promoting small-vessel stiffening through 5-HT2B (serotonin 2B receptor)-mediated signaling. METHODS AND RESULTS: We performed BM transplants using transgenic donor animals expressing diphtheria toxin secondary to activation of an endothelial-specific tamoxifen-inducible Cre and induced experimental PH using hypoxia with SU5416 to enhance endovascular injury and ablated BM-derived PACs, after which we measured right ventricular systolic pressures in a closed-chest procedure. BM-derived PAC lineage tracing was accomplished by transplanting BM from transgenic donor animals with fluorescently labeled hematopoietic cells and treating mice with a 5-HT2B antagonist. BM-derived PAC ablation both prevented and reversed experimental PH with SU5416-enhanced endovascular injury, reducing the number of muscularized pulmonary arterioles and normalizing arteriole stiffness as measured by atomic force microscopy. Similarly, treatment with a pharmacological antagonist of 5-HT2B also prevented experimental PH, reducing the number and stiffness of muscularized pulmonary arterioles. PACs accelerated pulmonary microvascular endothelial cell injury response in vitro, and the presence of BM-derived PACs significantly correlated with stiffer pulmonary arterioles in pulmonary arterial hypertension patients and mice with experimental PH. RNA sequencing of BM-derived PACs showed that 5-HT2B antagonism significantly altered biologic pathways regulating cell proliferation, locomotion and migration, and cytokine production and response to cytokine stimulus. CONCLUSIONS: Together, our findings illustrate that BM-derived PACs directly contribute to experimental PH with SU5416-enhanced endovascular injury by mediating small-vessel stiffening and remodeling in a 5-HT2B signaling-dependent manner.


Subject(s)
Hypertension, Pulmonary/pathology , Myeloid Progenitor Cells/metabolism , Receptor, Serotonin, 5-HT2B/metabolism , Vascular Stiffness , Angiogenesis Inhibitors/toxicity , Animals , Arterioles/pathology , Cell Lineage , Cells, Cultured , Hypertension, Pulmonary/blood , Hypertension, Pulmonary/etiology , Indoles/toxicity , Lung/blood supply , Mice , Mice, Inbred C57BL , Myeloid Progenitor Cells/cytology , Myeloid Progenitor Cells/transplantation , Pyrroles/toxicity
7.
Am J Physiol Lung Cell Mol Physiol ; 317(5): L639-L652, 2019 11 01.
Article in English | MEDLINE | ID: mdl-31461316

ABSTRACT

Pulmonary arterial hypertension (PAH) is a morbid disease characterized by progressive right ventricle (RV) failure due to elevated pulmonary artery pressures (PAP). In PAH, histologically complex vaso-occlusive lesions in the pulmonary vasculature contribute to elevated PAP. However, the mechanisms underlying dysfunction of the microvascular endothelial cells (MVECs) that comprise a significant portion of these lesions are not well understood. We recently showed that MVECs isolated from the Sugen/hypoxia (SuHx) rat experimental model of PAH (SuHx-MVECs) exhibit increases in migration/proliferation, mitochondrial reactive oxygen species (ROS; mtROS) production, intracellular calcium levels ([Ca2+]i), and mitochondrial fragmentation. Furthermore, quenching mtROS with the targeted antioxidant MitoQ attenuated basal [Ca2+]i, migration and proliferation; however, whether increased mtROS-induced [Ca2+]i entry affected mitochondrial morphology was not clear. In this study, we sought to better understand the relationship between increased ROS, [Ca2+]i, and mitochondrial morphology in SuHx-MVECs. We measured changes in mitochondrial morphology at baseline and following inhibition of mtROS, with the targeted antioxidant MitoQ, or transient receptor potential vanilloid-4 (TRPV4) channels, which we previously showed were responsible for mtROS-induced increases in [Ca2+]i in SuHx-MVECs. Quenching mtROS or inhibiting TRPV4 attenuated fragmentation in SuHx-MVECs. Conversely, inducing mtROS production in MVECs from normoxic rats (N-MVECs) increased fragmentation. Ca2+ entry induced by the TRPV4 agonist GSK1017920A was significantly increased in SuHx-MVECs and was attenuated with MitoQ treatment, indicating that mtROS contributes to increased TRPV4 activity in SuHx-MVECs. Basal and maximal respiration were depressed in SuHx-MVECs, and inhibiting mtROS, but not TRPV4, improved respiration in these cells. Collectively, our data show that, in SuHx-MVECs, mtROS production promotes TRPV4-mediated increases in [Ca2+]i, mitochondrial fission, and decreased mitochondrial respiration. These results suggest an important role for mtROS in driving MVEC dysfunction in PAH.


Subject(s)
Endothelial Cells/pathology , Hypoxia/complications , Indoles/toxicity , Lung/pathology , Mitochondria/pathology , Pulmonary Arterial Hypertension/pathology , Pyrroles/toxicity , Reactive Oxygen Species/metabolism , Angiogenesis Inhibitors/toxicity , Animals , Calcium/metabolism , Cells, Cultured , Endothelial Cells/metabolism , Lung/metabolism , Male , Mitochondria/metabolism , Oxygen Consumption , Pulmonary Arterial Hypertension/etiology , Pulmonary Arterial Hypertension/metabolism , Rats , Rats, Wistar , Vascular Remodeling
8.
Chembiochem ; 20(18): 2390-2401, 2019 09 16.
Article in English | MEDLINE | ID: mdl-31026110

ABSTRACT

Class 1 myosins (Myo1s) were the first unconventional myosins identified and humans have eight known Myo1 isoforms. The Myo1 family is involved in the regulation of gene expression, cytoskeletal rearrangements, delivery of proteins to the cell surface, cell migration and spreading. Thus, the important role of Myo1s in different biological processes is evident. In this study, we have investigated the effects of pentachloropseudilin (PClP), a reversible and allosteric potent inhibitor of Myo1s, on angiogenesis. We demonstrated that treatment of cells with PClP promoted a decrease in the number of vessels. The observed inhibition of angiogenesis is likely to be related to the inhibition of cell proliferation, migration and adhesion, as well as to alteration of the actin cytoskeleton pattern, as shown on a PClP-treated HUVEC cell line. Moreover, we also demonstrated that PClP treatment partially prevented the delivery of integrins to the plasma membrane. Finally, we showed that PClP caused DNA strand breaks, which are probably repaired during the cell cycle arrest in the G1 phase. Taken together, our results suggest that Myo1s participate directly in the angiogenesis process.


Subject(s)
Actin Cytoskeleton/drug effects , Angiogenesis Inhibitors/pharmacology , Cell Cycle/drug effects , Hydrocarbons, Chlorinated/pharmacology , Integrins/metabolism , Pyrroles/pharmacology , Angiogenesis Inhibitors/toxicity , Cell Adhesion/drug effects , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , G1 Phase Cell Cycle Checkpoints/drug effects , Human Umbilical Vein Endothelial Cells , Humans , Hydrocarbons, Chlorinated/toxicity , Integrins/genetics , Myosin Type I/metabolism , Pyrroles/toxicity , RNA, Messenger/metabolism
9.
J Biol Inorg Chem ; 24(3): 395-404, 2019 05.
Article in English | MEDLINE | ID: mdl-30927090

ABSTRACT

Based on recent researches, bio synthesized silver nanoparticles (Ag-NPs) seem to have the potential in declining angiogenesis and oxidative stress. In the current study, rapeseed flower pollen (RFP) water extract was triggered to synthesize RFP-silver nanoparticles (RFP/Ag-NPs). Moreover, antioxidant, antiangiogenesis and cytotoxicity of the RFP/Ag-NPs against MDA-MB-231, MCF7 and carcinoma cell lines and normal human skin fibroblast HDF were compared. Results indicated that RFP/Ag-NPs have a peak at 430 nm, spherical shape and an average size of 24 nm. According to the results of FTIR, rapeseed pollen capped Ag-NPs. RFP/Ag-NPs have cytotoxicity on MDA-MB-231 and MCF7 cells and decrease cancerous cell viability (IC50 = 3 µg/ml and 2 µg/ml, respectively) in a dose- and time-dependent manner. The morphological data showed that the RFP/Ag-NPs increase the percentage of apoptotic cells compared to the control group and normal cells (human skin fibroblast cells). The apoptotic morphological change was also confirmed with a flow cytometric analysis. RFP/ Ag-NPs' antioxidant activity was evaluated by measuring their ability to scavenge ABTS and DPPH free radicals. The IC50 values were determined at 800 and 830 µg/ml for ABTS and DPPH tests, respectively. According to the results, green-synthesized RFP/Ag-NPs as a safe efficient apoptosis inducer and strong antioxidant compound have the potential to suppress breast cancer carcinogenesis by VEGF down-regulatiion and thus sensitizing them against apoptosis. However, further researches are required to clarify RFP/Ag-NPs' cell specificity and therapeutic doses in in vivo conditions.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Antioxidants/pharmacology , Brassica napus/chemistry , Metal Nanoparticles/chemistry , Plant Extracts/chemistry , Pollen/chemistry , Angiogenesis Inhibitors/chemical synthesis , Angiogenesis Inhibitors/toxicity , Antioxidants/chemical synthesis , Antioxidants/toxicity , Apoptosis/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Green Chemistry Technology/methods , Humans , Metal Nanoparticles/toxicity , Silver/chemistry
10.
Bioorg Chem ; 92: 103251, 2019 11.
Article in English | MEDLINE | ID: mdl-31525526

ABSTRACT

Worldwide, Hepatocellular Carcinoma (HCC) endures to be a prominent cause of cancer death. Treatment of HCC follows multiple therapies which are not entirely applicable for treatment of all patients. HCC usually arises contextual to chronic liver diseases and is often discovered at later stages which makes treatment options more complex. The present study aimed at design, synthesis & evaluation of new pyridazinoquinazoline derivatives as potential nontoxic anti-hepatocellular carcinoma (HCC) agents, through inhibition of Vascular endothelial growth factor -2 (VEGFR-2). Novel Pyridazino[3, 4, 5-de]quinazoline derivatives (2-6) were designed & synthesized. Their structures were confirmed via spectral and microanalytical data. They were tested for their in vitro VEGFR-2 inhibition & anticancer activity against human liver cancer cell line (HEPG-2). Molecular docking was investigated into VEGFR-2 site. In vivo studies of VEGRF-2 inhibition and the anti-apoptotic effect of the new compounds were determined in liver of irradiated rats. Toxicity of synthesized compounds was also assessed. The results showed that compounds 3-6 have significant antitumor activity and proved to be non-toxic. The ethoxy aniline derivative 6, exhibited the highest activity both in vitro and in vivo compared to the reference drug used, sorafenib. Compound 6 could be considered a promising nontoxic anti HCC agent and this could be partially attributed to its VEGFR-2 inhibition. Future preclinical investigation would be carried out to confirm the specific and exact mechanism of action of these derivatives especially compound 6 as an effective pharmaceutical agent after full toxicological and pharmacological assessment.


Subject(s)
Angiogenesis Inhibitors/chemical synthesis , Drug Design , Liver/drug effects , Protein Kinase Inhibitors/chemical synthesis , Pyridazines/chemistry , Quinazolines/chemistry , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors , Angiogenesis Inhibitors/chemistry , Angiogenesis Inhibitors/pharmacology , Angiogenesis Inhibitors/toxicity , Animals , Apoptosis/drug effects , Apoptosis/radiation effects , Gamma Rays , Hep G2 Cells , Humans , Lethal Dose 50 , Liver/metabolism , Liver/radiation effects , Male , Mice , Molecular Docking Simulation , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/toxicity , Rats, Wistar , Toxicity Tests, Acute , Whole-Body Irradiation
11.
Doc Ophthalmol ; 138(3): 181-194, 2019 06.
Article in English | MEDLINE | ID: mdl-30809742

ABSTRACT

PURPOSE: Investigate the potential application of corosolic acid (CA) in the treatment of diseases causing retinal neovascularization. METHODS: CA cytotoxicity effect was evaluated in ARPE-19 cells by sulforhodamine B colorimetric method, and antiangiogenic activity was studied using chorioallantoic membrane (CAM) assay. An amount of 0.01 mL of CA formulations at 5, 10 and 25 µM was injected in the right eyes of Wistar rats, and the contralateral eyes received the vehicle to verify the safety of ophthalmic use. Electroretinography (ERG) was performed before, 7 and 15 days after CA administration. Animals were killed on the 15th day, and the histological analysis of retina was carried out under light microscopy. RESULTS: CA did not present cytotoxicity at concentrations below 35.5 µM after 48 h of treatment. The antiangiogenic activity was confirmed by CAM assay, since CA (range from 5 to 25 µM) induced a significant reduction in vascularity without any signs of toxicity. ERG recordings and histological evaluation did not show any signs of retinal toxicity. CONCLUSIONS: CA was effective in reducing vascularity in a CAM model and was found to be safe for potential ophthalmic use.


Subject(s)
Angiogenesis Inhibitors/administration & dosage , Retina/drug effects , Triterpenes/administration & dosage , Angiogenesis Inhibitors/toxicity , Animals , Cell Line , Chorioallantoic Membrane/blood supply , Electroretinography/drug effects , Intravitreal Injections , Male , Neovascularization, Pathologic/drug therapy , Rats , Rats, Wistar , Retinal Pigment Epithelium/drug effects , Triterpenes/toxicity , Vitreous Body/drug effects
12.
Am J Physiol Lung Cell Mol Physiol ; 314(5): L893-L907, 2018 05 01.
Article in English | MEDLINE | ID: mdl-29388466

ABSTRACT

Pulmonary arterial hypertension (PAH) is a lethal disease characterized by elevations in pulmonary arterial pressure, in part due to formation of occlusive lesions in the distal arterioles of the lung. These complex lesions may comprise multiple cell types, including endothelial cells (ECs). To better understand the molecular mechanisms underlying EC dysfunction in PAH, lung microvascular endothelial cells (MVECs) were isolated from normoxic rats (N-MVECs) and rats subjected to SU5416 plus hypoxia (SuHx), an experimental model of PAH. Compared with N-MVECs, MVECs isolated from SuHx rats (SuHx-MVECs) appeared larger and more spindle shaped morphologically and expressed canonical smooth muscle cell markers smooth muscle-specific α-actin and myosin heavy chain in addition to endothelial markers such as Griffonia simplicifolia and von Willebrand factor. SuHx-MVEC mitochondria were dysfunctional, as evidenced by increased fragmentation/fission, decreased oxidative phosphorylation, and increased reactive oxygen species (ROS) production. Functionally, SuHx-MVECs exhibited increased basal levels of intracellular calcium concentration ([Ca2+]i) and enhanced migratory and proliferative capacity. Treatment with global (TEMPOL) or mitochondria-specific (MitoQ) antioxidants decreased ROS levels and basal [Ca2]i in SuHx-MVECs. TEMPOL and MitoQ also decreased migration and proliferation in SuHx-MVECs. Additionally, inhibition of ROS-induced Ca2+ entry via pharmacologic blockade of transient receptor potential vanilloid-4 (TRPV4) attenuated [Ca2]i, migration, and proliferation. These findings suggest a role for mitochondrial ROS-induced Ca2+ influx via TRPV4 in promoting abnormal migration and proliferation in MVECs in this PAH model.


Subject(s)
Calcium/metabolism , Endothelium, Vascular/pathology , Hypertension, Pulmonary/pathology , Hypoxia/physiopathology , Indoles/toxicity , Pulmonary Artery/pathology , Pyrroles/toxicity , TRPV Cation Channels/metabolism , Vascular Diseases/pathology , Angiogenesis Inhibitors/toxicity , Animals , Cells, Cultured , Endothelium, Vascular/metabolism , Hypertension, Pulmonary/etiology , Hypertension, Pulmonary/metabolism , Male , Microvessels/metabolism , Microvessels/pathology , Pulmonary Artery/metabolism , Rats , Rats, Wistar , TRPV Cation Channels/genetics , Vascular Diseases/etiology , Vascular Diseases/metabolism
13.
J Anat ; 232(4): 568-574, 2018 04.
Article in English | MEDLINE | ID: mdl-29023763

ABSTRACT

Thalidomide notoriously caused severe birth defects, particularly to the limbs, in those exposed in utero following maternal use of the drug to treat morning sickness. How the drug caused these birth defects remains unclear. Many theories have been proposed including actions on the forming blood vessels. However, thalidomide survivors also have altered nerve patterns and the drug is known for its neurotoxic actions in adults following prolonged use. We have previously shown that CPS49, an anti-angiogenic analog of thalidomide, causes a range of limb malformations in a time-sensitive manner in chicken embryos. Here we investigated whether CPS49 also is neurotoxic and whether effects on nerve development impact upon limb development. We found that CPS49 is neurotoxic, just like thalidomide, and can cause some neuronal loss late developing chicken limbs, but only when the limb is already innervated. However, CPS49 exposure does not cause defects in limb size when added to late developing chicken limbs. In contrast, in early limb buds which are not innervated, CPS49 exposure affects limb area significantly. To investigate in more detail the role of neurotoxicity and its impact on chicken limb development we inhibited nerve innervation at a range of developmental timepoints through using ß-bungarotoxin. We found that neuronal inhibition or ablation before, during or after limb outgrowth and innervation does not result in obvious limb cartilage patterning or number changes. We conclude that while CPS49 is neurotoxic, given the late innervation of the developing limb, and that neuronal inhibition/ablation throughout limb development does not cause similar limb patterning anomalies to those seen in thalidomide survivors, nerve defects are not the primary underlying cause of the severe limb patterning defects induced by CPS49/thalidomide.


Subject(s)
Angiogenesis Inhibitors/toxicity , Body Patterning/drug effects , Embryonic Development/drug effects , Limb Deformities, Congenital/chemically induced , Neuronal Outgrowth/drug effects , Teratogens/toxicity , Thalidomide/analogs & derivatives , Animals , Bungarotoxins/pharmacology , Chick Embryo , Extremities/embryology , Extremities/innervation , Female , Limb Buds/innervation , Mice , Mice, Inbred C57BL , Neurotoxicity Syndromes , Thalidomide/toxicity
14.
Exp Eye Res ; 171: 1-11, 2018 06.
Article in English | MEDLINE | ID: mdl-29522724

ABSTRACT

Anti-Vascular Endothelial Growth Factor (VEGF) therapy is given repeatedly for an extended period of time to patients when treated for age-related macular degeneration. While short-term effects of anti-VEGF agents on retinal pigment epithelial (RPE) cells have been investigated, the effects of long-term and repeated treatment on these cells are scarce. In this study, we have investigated the effects of anti-VEGF treatment after long-term, repeated treatment on cell aging and morphology. The experiments were conducted in primary porcine RPE cells passage one and two. Cells were treated with 125 µg/ml bevacizumab, ranibizumab, aflibercept or rituximab once a week for 1 day, 4 days, 7 days, 4 weeks and 12 weeks. Cell survival was evaluated with 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide tetrazolium (MTT) and trypan blue exclusion assay. Activity of ß-galactosidase was assessed in a commercially available assay. Influence of these compounds was investigated on the expression of cathepsin D and amyloid ß, and the expression and phosphorylation of mechanistic target of rapamycin (mTOR), all proteins involved in senescence and aging, in Western blot. The secretion of Pigment Epithelium Derived Factor (PEDF) and Transforming Growth Factor (TGF)-ß was investigated in Enzyme-linked Immunosorbent Assay (ELISA). The cellular morphology was investigated with electron microscopy, investigating the number and area of mitochondria and autophagosomes. Statistical analysis was conducted using a mixed linear model. Weekly treatment up to 12 weeks displayed no toxic effects on RPE cells in any of the substances tested. Ranibizumab showed a significant increase in ß-galactosidase signal on day 4 (p < 0.05) and 7 (p < 0.05) after treatment. In long-term, however, ranibizumab displayed no significant difference to untreated cells. Bevacizumab displayed a significant reduction of the ß-galactosidase signal after 12 weeks (p < 0.05). Aflibercept significantly decreased ß-galactosidase after 1 day (p < 0.01) and 12 weeks (p < 0.05). Rituximab and bevacizumab also decreased ß-galactosidase signal after 12 weeks (p < 0.05). The expression of mTOR, phospho-mTOR, amyloid ß and cathepsin D was not significantly altered by any of the compounds tested. RPE cells secreted considerate amounts of TGF-ß. Bevacizumab treated cells showed significantly lower TGF-ß secretion than ranibizumab and rituximab (p < 0.05). In contrast, only small amounts of PEDF were secreted which were not altered by any substance tested. Ultrastructural analysis showed no alterations in mitochondria after long-term treatment with either substance. Autophagosomes were not reduced by long-term anti-VEGF treatment compared to control. However, the area of autophagosomes in bevacizumab and aflibercept treated cells was significantly less compared to both ranibizumab and rituximab treated cells (all p < 0.05). Taken together, weekly treatment with VEGF-antagonists up to 3 months does not induce premature aging in primary RPE cells in any tested compound. A significant difference can be found between bevacizumab and aflibercept on the one hand, and ranibizumab (and rituximab) on the other hand, with more autophagosomal area in ranibizumab and (rituximab). Taken together, our data provide indications for long-term safety of anti-VEGF compounds. Further research is warranted.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Cellular Senescence/drug effects , Retinal Pigment Epithelium/drug effects , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Amyloid beta-Peptides/metabolism , Angiogenesis Inhibitors/toxicity , Animals , Bevacizumab/pharmacology , Blotting, Western , Cathepsin D/metabolism , Cell Survival , Cells, Cultured , Enzyme-Linked Immunosorbent Assay , Eye Proteins/metabolism , Follow-Up Studies , Microscopy, Electron , Nerve Growth Factors/metabolism , Phosphorylation , Ranibizumab/pharmacology , Receptors, Vascular Endothelial Growth Factor , Recombinant Fusion Proteins/pharmacology , Retinal Pigment Epithelium/metabolism , Retinal Pigment Epithelium/ultrastructure , Rituximab/pharmacology , Serpins/metabolism , Swine , TOR Serine-Threonine Kinases/metabolism , Transforming Growth Factor beta/metabolism , beta-Galactosidase/metabolism
15.
J Vasc Interv Radiol ; 29(8): 1078-1084, 2018 08.
Article in English | MEDLINE | ID: mdl-29910164

ABSTRACT

PURPOSE: To assess feasibility and efficacy of CKD-516, a vascular disrupting agent, in transarterial chemoembolization in a liver tumor model. MATERIALS AND METHODS: A VX2 carcinoma strain was implanted in rabbit liver (n = 40) and incubated for 2 weeks. After confirmation of tumor growth using computed tomography, transarterial chemoembolization was performed. CKD-516 was dissolved in ethiodized oil, and animals were allocated to 4 treatment groups (n = 10 in each): group A, ethiodized oil; group B, ethiodized oil/CKD-516; group C, ethiodized oil + doxorubicin; group D, ethiodized oil/CKD-516 + doxorubicin. To assess hepatic damage, serum aspartate transaminase and alanine transaminase levels were measured on day 1, 3, and 7 after delivery. To assess tumor necrosis, animals were euthanized on day 7, and explanted tumors were stained with hematoxylin and eosin and a terminal deoxynucleotidyl transferase deoxyuridine triphosphate nick end labeling assay. Percentage areas of viable tumors were calculated using digitalized histopathologic specimen images. RESULTS: Tumor viability rates were 47.1% ± 11.4%, 27.5% ± 13.6%, 14.4% ± 12.5%, and 0.7% ± 1.0% in groups A, B, C, and D (P < .001). Liver enzyme levels were elevated after drug delivery but recovered during follow-up. Significant between-group differences were observed on days 1, 3, and 7 (aspartate transaminase and alanine transaminase: P = .0135 and P = .0134, P = .0390 and P = .0084, and P = .8260 and P = .0440). CONCLUSIONS: Treatment with a combination of CKD-516 and conventional transarterial chemoembolization showed therapeutic benefit in a liver tumor model.


Subject(s)
Angiogenesis Inhibitors/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Benzophenones/administration & dosage , Chemoembolization, Therapeutic/methods , Doxorubicin/administration & dosage , Ethiodized Oil/administration & dosage , Liver Neoplasms, Experimental/blood supply , Liver Neoplasms, Experimental/therapy , Valine/analogs & derivatives , Alanine Transaminase/blood , Angiogenesis Inhibitors/toxicity , Animals , Antineoplastic Combined Chemotherapy Protocols/toxicity , Aspartate Aminotransferases/blood , Benzophenones/toxicity , Cell Line, Tumor , Cell Survival/drug effects , Chemoembolization, Therapeutic/adverse effects , Doxorubicin/toxicity , Ethiodized Oil/toxicity , Liver Neoplasms, Experimental/diagnostic imaging , Liver Neoplasms, Experimental/pathology , Male , Necrosis , Rabbits , Time Factors , Tomography, X-Ray Computed , Tumor Burden/drug effects , Valine/administration & dosage , Valine/toxicity
16.
BMC Ophthalmol ; 18(1): 316, 2018 Dec 11.
Article in English | MEDLINE | ID: mdl-30537942

ABSTRACT

BACKGROUND: Ingrowth of newly formed blood and lymph vessels (angiogenesis) from the limbus region into the cornea can be treated successfully by subconjunctival application of antiangiogenic agents. Currently, there are several angiogenesis inhibitors from various manufacturers available, such as vascular endothelial growth factor (VEGF) antibodies. The aim of the study was to investigate potential cytotoxic effects of two anti-VEGF agents, ranibizumab (Lucentis®) and bevacizumab (Avastin®) on the human corneal endothelium. METHODS: Human donor corneas, not suitable for corneal transplantation, were organ-cultured in the presence of either ranibizumab (Lucentis®) or bevacizumab (Avastin®) at different concentrations (group 1: 250 µg / ml, group 2: 25 µg / ml, group 3: 2.5 µg / ml) for a period of up to 4 weeks. Microscopic imaging for endothelial cell counting, detection of morphologic alterations of the endothelium, and molecular biology testing (Enzyme-linked Immunosorbent Assay [ELISA]) for metabolic changes was performed. RESULTS: Background-corrected results showed neither a significant lactate dehydrogenase (LDH) change with increasing culturing time nor a significant difference between ranibizumab (Lucentis®) and bevacizumab (Avastin®) treatment. The endothelial cell density revealed also no statistically significant difference between the two treatment groups with ranibizumab (Lucentis®) and bevacizumab (Avastin®) at all concentrations tested in this study. CONCLUSIONS: In this study, the anti-angiogenic agents ranibizumab (Lucentis®) and bevacizumab (Avastin®) demonstrated no cytotoxic effects on the corneal endothelium of human organ-cultured donor corneas over the limited study time period of 4 weeks. However, based on the study design (in-vitro) and the limited follow-up period, no conclusions on potential long-term effects can be drawn.


Subject(s)
Angiogenesis Inhibitors/toxicity , Bevacizumab/toxicity , Endothelial Cells/drug effects , Endothelium, Corneal/drug effects , Ranibizumab/toxicity , Aged , Aged, 80 and over , Cell Count , Cells, Cultured , Enzyme-Linked Immunosorbent Assay , Female , Humans , Male , Middle Aged
17.
Regul Toxicol Pharmacol ; 95: 236-243, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29574193

ABSTRACT

Bevacizumab, a recombinant humanized monoclonal antibody targeting vascular endothelial growth factor (VEGF), is approved for treatment of metastatic colorectal cancer, nonsquamous non-small-cell lung cancer, metastatic kidney cancer, and glioblastoma. To support clinical development of the potential bevacizumab biosimilar PF-06439535, nonclinical studies evaluated structural, functional, toxicological, and toxicokinetic similarity to bevacizumab sourced from the European Union (bevacizumab-EU) and United States (bevacizumab-US). Peptide mapping demonstrated the amino acid sequence of PF-06439535 was identical to bevacizumab-EU and bevacizumab-US. Biologic activity, measured via inhibition of VEGF-induced cell proliferation in human umbilical vein endothelial cells and binding to VEGF isoforms, was similar across the three drugs. In vivo similarity was demonstrated in cynomolgus monkeys administered intravenous PF-06439535 or bevacizumab-EU (0 or 10 mg/kg/dose twice weekly for 1 month; total of nine doses). Systemic exposure appeared similar and test article-related effects were limited to physeal dysplasia of the distal femur. The potential for non-target-mediated toxicity of PF-06439535 was evaluated in rats administered intravenous PF-06439535 (15 or 150 mg/kg/dose twice weekly for 15 days; total of five doses). Nonadverse higher liver weights and minimal sinusoidal cell hyperplasia were observed. Collectively, these studies demonstrated similarity of PF-06439535 to bevacizumab, supporting entry into clinical development.


Subject(s)
Angiogenesis Inhibitors/toxicity , Antineoplastic Agents, Immunological/toxicity , Bevacizumab/toxicity , Biosimilar Pharmaceuticals/toxicity , Angiogenesis Inhibitors/blood , Angiogenesis Inhibitors/pharmacokinetics , Angiogenesis Inhibitors/pharmacology , Animals , Antineoplastic Agents, Immunological/blood , Antineoplastic Agents, Immunological/pharmacokinetics , Antineoplastic Agents, Immunological/pharmacology , Bevacizumab/blood , Bevacizumab/pharmacokinetics , Bevacizumab/pharmacology , Biosimilar Pharmaceuticals/pharmacokinetics , Biosimilar Pharmaceuticals/pharmacology , Cell Proliferation/drug effects , Female , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/physiology , Humans , Liver/drug effects , Liver/pathology , Macaca fascicularis , Male , Molecular Structure , Organ Size/drug effects , Protein Binding , Protein Isoforms/metabolism , Rats, Sprague-Dawley , Vascular Endothelial Growth Factor A/metabolism
18.
Doc Ophthalmol ; 135(3): 175-185, 2017 12.
Article in English | MEDLINE | ID: mdl-28825191

ABSTRACT

PURPOSE: To assess the retinal toxicity of an intravitreal injection of infliximab, a monoclonal antibody to tumor necrosis factor α, in a rabbit model. MATERIALS AND METHODS: Two groups of adult albino rabbits (n = 5) received intravitreal injections of infliximab (0.1 ml) in the study eye and balanced salt solution (BSS, 0.1 ml) in the control eye at baseline. Group 1 was administered with 1.5 mg/0.1 ml, and group 2 was injected with 7.5 mg/0.1 ml of infliximab solution. Electroretinography (ERG) was performed at baseline and at 1, 7, 30, and 45 days after the injection. Visual evoked potentials (VEPs) were recorded at 7 and 45 days after the injection. After the last electrophysiological assessment, the rabbits were euthanized and retinal histopathology and immunhistochemistry for glial fibrillary acidic protein (GFAP) were performed. RESULTS: ERG responses demonstrated no significant deficit in retinal function in eyes injected with infliximab. Mean dark-adapted a-wave and b-wave maximal amplitude and semi-saturation constant values at baseline and throughout the 45 days of follow-up after the injection indicated no remarkable difference in outer retinal function between the control and experimental eyes. VEP responses were similar at each time point (7 and 45 days). No difference was seen in retinal histopathology and immunocytochemistry sections in eyes receiving the 1.5 mg/0.1 ml dose compared to the control eyes. However, increased GFAP labeling in retinal Müller cells was detected in rabbit eyes treated with the 7.5 mg/0.1 ml dose. CONCLUSIONS: Intravitreal injection of 1.5 mg/0.1 ml infliximab dose has no toxic effect on the integrity (functional or structural) of the retina in rabbits. A higher dose of 7.5 mg/0.1 ml may be slightly toxic as suggested by positive Müller cell GFAP expression. Additional studies of retinal toxicity at higher doses and after multiple injections are needed to establish the retinal safety of intravitreal infliximab therapy in humans.


Subject(s)
Angiogenesis Inhibitors/toxicity , Infliximab/toxicity , Retina/drug effects , Angiogenesis Inhibitors/administration & dosage , Animals , Antibodies, Monoclonal , Electroretinography/drug effects , Ependymoglial Cells/metabolism , Evoked Potentials, Visual/drug effects , Eye Diseases/physiopathology , Glial Fibrillary Acidic Protein/metabolism , Infliximab/administration & dosage , Intravitreal Injections , Rabbits , Retina/metabolism , Retina/pathology , Tumor Necrosis Factor-alpha , Vitreous Body/metabolism
19.
Chem Biodivers ; 14(3)2017 Mar.
Article in English | MEDLINE | ID: mdl-27936296

ABSTRACT

Angiogenesis plays a major role in the normal embryonic development and in diseases such as cancer. Drugs that control angiogenesis are an alternative way to tackle this disease. The polyphenols usnic acid (3), genistein (5), and daidzein (6) were tested for antiangiogenic and unwanted effects in zebrafish embryos whose blood vessel system resembles that of mammals. The established tyrosine kinase inhibitors axitinib (1) and tyrphostin AG490 (2) were included for comparison. All compounds except 6 caused distinct antiangiogenic effects such as a concentration-dependent reduction of intersegmental vessels, dorsal longitudinal anastomotic vessels, subintestinal veins and secondary sprouts. As side effects, pericardial oedema and the impairment of blood flow were observed. Usnic acid (3), genistein (5) and Cu(II)-genisteinate (7) gave rise to a curvature of the spine. Compounds 5 and 7 also induced cell death in the head of the embryos at higher doses. All effects were more pronounced when the compounds had been applied at an early stage (24 hpf) rather than at 48 hpf. The copper complexes 4 and 7 showed a stronger antiangiogenic effect than the free ligands 3 and 5. The genistein complex 7 was antiangiogenic at doses so low that side effects were tolerable, and thus it may be a potential anticancer drug candidate.


Subject(s)
Benzofurans/chemistry , Coordination Complexes/pharmacology , Coordination Complexes/toxicity , Copper/chemistry , Genistein/chemistry , Zebrafish/growth & development , Angiogenesis Inhibitors/chemistry , Angiogenesis Inhibitors/pharmacology , Angiogenesis Inhibitors/toxicity , Animals , Axitinib , Cell Line, Tumor , Cell Movement/drug effects , Cell Survival/drug effects , Coordination Complexes/chemistry , Embryo, Nonmammalian/blood supply , Embryo, Nonmammalian/drug effects , Embryo, Nonmammalian/metabolism , Embryonic Development/drug effects , Humans , Imidazoles/pharmacology , Imidazoles/toxicity , Indazoles/pharmacology , Indazoles/toxicity , Matrix Metalloproteinases/metabolism , Microscopy, Fluorescence , Neovascularization, Physiologic/drug effects , Zebrafish/metabolism
20.
Nephrol Dial Transplant ; 31(6): 914-21, 2016 06.
Article in English | MEDLINE | ID: mdl-26681729

ABSTRACT

BACKGROUND: Antiangiogenic treatment with the multitargeted vascular endothelial growth factor (VEGF) receptor inhibitor sunitinib associates with a blood pressure (BP) rise and glomerular renal injury. Recent evidence indicates that VEGF derived from tubular cells is required for maintenance of the peritubular vasculature. In the present study, we focussed on tubular and glomerular pathology induced by sunitinib and explored whether a high salt (HS) diet augments the BP rise and renal abnormalities. METHODS: Normotensive Wistar Kyoto (WKY) rats were exposed to a normal salt (NS) or HS diet for 2 weeks and subsequently for 8 days to sunitinib or vehicle administration after which the rats were euthanized and kidneys excised. Mean arterial pressure (MAP) was telemetrically measured. Urine was sampled for proteinuria and endothelinuria, and blood for measurement of endothelin-1, creatinine and cystatin C. RESULTS: Compared with the NS diet, MAP rapidly rose by 27 ± 3 mmHg with the HS diet. On sunitinib, MAP rose further by 15 ± 1 with the NS and by 23 ± 4 mmHg with the HS diet (P < 0.05). The HS diet itself had no effect on proteinuria, endothelinuria or the plasma levels of endothelin-1, creatinine and cystatin C. Only with the HS diet, sunitinib administration massively increased proteinuria and endothelinuria and these two parameters were related (r = 0.50, P < 0.01). Likewise, renal glomerular pathology was enhanced during sunitinib with the HS diet, whereas tubulointerstitial injury or reduced peritubular capillary density did not occur. CONCLUSIONS: An HS diet induces a marked BP rise in WKY rats and exacerbates both the magnitude of the BP rise and glomerular injury induced by sunitinib.


Subject(s)
Angiogenesis Inhibitors/toxicity , Blood Pressure/drug effects , Indoles/toxicity , Kidney Diseases/pathology , Pyrroles/toxicity , Sodium Chloride, Dietary/toxicity , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Animals , Kidney Diseases/drug therapy , Kidney Diseases/etiology , Male , Rats , Rats, Inbred WKY , Sunitinib
SELECTION OF CITATIONS
SEARCH DETAIL