Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
Add more filters










Publication year range
1.
Microvasc Res ; 101: 118-26, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26215931

ABSTRACT

Recent data has indicated that, besides its classical therapeutic indication in hyperurecemia and gout, xanthine oxidase inhibitors can be used to various forms of ischemia and other types of tissue and vascular injuries. We tested the hypothesis that allopurinol, an inhibitor of xanthine oxidase (XO), might modulate acute and/or chronic inflammatory angiogenesis induced by subcutaneous implantation of synthetic matrix in mice. C57/BL6 male mice (6-7 weeks) were implanted with polyether-polyurethane sponge discs. The animals received by oral gavage 1.0mg/kg of allopurinol for six consecutive days in two treatment regimen. In the first series of experiments, the treatment was initiated 24h post-implantation and the implants were removed at day 7 post-implantation. For the assessment of the effect of the compound on chronic inflammation, the treatment was initiated at day 8 post-implantation and the implants removed 14days post-implantation. Angiogenesis as determined by hemoglobin content, VEGF levels and number of vessels intraimplant, and inflammation (myeloperoxidase -MPO, n-acetyl-ß-d-glucosaminidase -NAG, TNF-α and CCL2 levels) were reduced by allopurinol treatment in acute phase. Similarly, the treatment inhibited nitric oxide and H2O2 production. However, fibrogenesis determined by collagen deposition and levels of TGF-ß1 increased in the implants after allopurinol treatment. In marked contrast with the effects when the treatment initiated 24h post-implantation, allopurinol increased angiogenesis and inflammation but reduced collagen and TGF-ß1 levels intra-implant, when the treatment was started during the chronic inflammatory process. The dual effects of allopurinol described here, extend its range of actions as a potential agent able to modulate the components of the fibrovascular tissue present in both physiological (healing processes) as well as in chronic fibroproliferative diseases. These modulatory effects depended on the phase at which the treatment was initiated.


Subject(s)
Allopurinol/chemistry , Acetylglucosaminidase/metabolism , Animals , Collagen/chemistry , Ether/chemistry , Hemoglobins/analysis , Hemoglobins/metabolism , Hydrogen Peroxide/chemistry , Inflammation , Male , Mice , Mice, Inbred C57BL , Models, Animal , Neovascularization, Pathologic/drug therapy , Nitric Oxide/chemistry , Peroxidase/metabolism , Polyurethanes/chemistry , Time Factors , Transforming Growth Factor beta1/metabolism , Tumor Necrosis Factor-alpha/metabolism , Vascular Endothelial Growth Factor A/metabolism , Xanthine Oxidase/antagonists & inhibitors
2.
Microvasc Res ; 86: 44-51, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23253264

ABSTRACT

Inflammation, angiogenesis and cytokine production are common features of almost, if not all tumors. However, the extent of these processes induced by different types of tumors has not been evaluated. We investigated the growth pattern of the experimental metastatic tumors, B16F10 melanoma, CT26.WT colon and 4T1 mammary cells inoculated in the flank of syngeneic mice and determined the degree of inflammation, angiogenesis, and production level of pro-inflammatory and pro-angiogenic cytokines within the tumors. In addition, we have analyzed vascular changes in the interface between the tumors and the adjacent cutaneous tissue and levels of relevant pro-inflammatory and pro-angiogenic cytokines systemically. The weight of tumors 15 days post-inoculation of 10(6) cells was markedly different. Melanomas were 2 and 10-fold heavier than colon and mammary tumors, respectively. Locally, CT26.WT tumor cells induced more vessels in cutaneous tissue adjacent to the tumors but systemically, the plasma levels of VEGF were higher (approximately 2-fold) in 4T1 tumor-bearing mice compared with the other two tumors. Mammary tumors presented the most prominent inflammatory content as assessed by a range of markers (inflammatory enzymes and cytokines). The vascular index, as determined by the intra-tumor amount of hemoglobin and number of vessels in hot spot areas, was also higher (approximately 2-fold) in melanomas compared with the other two tumors. These findings showing that distinct tumor types determine differential grade of inflammation, angiogenesis and host interaction in mice may provide new insights to tailor differential therapeutic approach based on the status of tumor biomarkers.


Subject(s)
Colonic Neoplasms/blood supply , Inflammation/etiology , Mammary Neoplasms, Experimental/blood supply , Melanoma, Experimental/blood supply , Neovascularization, Pathologic/etiology , Animals , Biomarkers , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Cytokines/biosynthesis , Cytokines/genetics , Female , Hemoglobins/analysis , Inflammation/blood , Intercellular Signaling Peptides and Proteins/biosynthesis , Intercellular Signaling Peptides and Proteins/genetics , Lymphocytes, Tumor-Infiltrating , Male , Mammary Neoplasms, Experimental/metabolism , Mammary Neoplasms, Experimental/pathology , Melanoma, Experimental/metabolism , Melanoma, Experimental/pathology , Mice , Mice, Inbred BALB C , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Nitric Acid/metabolism , Skin/blood supply , Tumor Burden , Tumor Necrosis Factor-alpha/analysis , Vascular Endothelial Growth Factor A/blood
3.
Br J Pharmacol ; 165(2): 436-54, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21718305

ABSTRACT

BACKGROUND AND PURPOSE: DF 2156A is a new dual inhibitor of IL-8 receptors CXCR1 and CXCR2 with an optimal pharmacokinetic profile. We characterized its binding mode, molecular mechanism of action and selectivity, and evaluated its therapeutic potential. EXPERIMENTAL APPROACH: The binding mode, molecular mechanism of action and selectivity were investigated using chemotaxis of L1.2 transfectants and human leucocytes, in addition to radioligand and [(35) S]-GTPγS binding approaches. The therapeutic potential of DF 2156A was evaluated in acute (liver ischaemia and reperfusion) and chronic (sponge-induced angiogenesis) experimental models of inflammation. KEY RESULTS: A network of polar interactions stabilized by a direct ionic bond between DF 2156A and Lys(99) on CXCR1 and the non-conserved residue Asp(293) on CXCR2 are the key determinants of DF 2156A binding. DF 2156A acted as a non-competitive allosteric inhibitor blocking the signal transduction leading to chemotaxis without altering the binding affinity of natural ligands. DF 2156A effectively and selectively inhibited CXCR1/CXCR2-mediated chemotaxis of L1.2 transfectants and leucocytes. In a murine model of sponge-induced angiogenesis, DF 2156A reduced leucocyte influx, TNF-α production and neovessel formation. In vitro, DF 2156A prevented proliferation, migration and capillary-like organization of HUVECs in response to human IL-8. In a rat model of liver ischaemia and reperfusion (I/R) injury, DF 2156A decreased PMN and monocyte-macrophage infiltration and associated hepatocellular injury. CONCLUSION AND IMPLICATIONS: DF 2156A is a non-competitive allosteric inhibitor of both IL-8 receptors CXCR1 and CXCR2. It prevented experimental angiogenesis and hepatic I/R injury in vivo and, therefore, has therapeutic potential for acute and chronic inflammatory diseases.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Receptors, Interleukin-8A/antagonists & inhibitors , Receptors, Interleukin-8B/antagonists & inhibitors , Sulfonamides/pharmacology , Animals , Anti-Inflammatory Agents/pharmacokinetics , Anti-Inflammatory Agents/therapeutic use , Cell Membrane/metabolism , Cell Proliferation/drug effects , Chemotaxis, Leukocyte/drug effects , Disease Models, Animal , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Interleukin-8/metabolism , Leukocytes/drug effects , Leukocytes/immunology , Leukocytes/metabolism , Liver/drug effects , Liver/immunology , Liver/pathology , Male , Mice , Mice, Inbred C57BL , Models, Molecular , Mutagenesis, Site-Directed , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Rats , Rats, Sprague-Dawley , Receptors, Interleukin-8A/genetics , Receptors, Interleukin-8A/metabolism , Receptors, Interleukin-8B/genetics , Receptors, Interleukin-8B/metabolism , Reperfusion Injury/drug therapy , Reperfusion Injury/immunology , Reperfusion Injury/pathology , Skin/blood supply , Sulfonamides/pharmacokinetics , Sulfonamides/therapeutic use
4.
Br J Pharmacol ; 161(7): 1458-71, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20942845

ABSTRACT

BACKGROUND AND PURPOSE: Vasculopathies represent the main cause of morbidity and mortality in diabetes. Vascular malfunctioning in diabetes is associated with abnormal vasoconstriction and Ca(2+) handling by smooth muscle cells (SMC). Phosphatidylinositol 3-kinases (PI3K) are key mediators of insulin action and have been shown to modulate the function of voltage-dependent L-type Ca(2+) channels (Ca(V) 1.2). In the present work, we investigated the involvement of PI3K signalling in regulating Ca(2+) current through Ca(V) 1.2 (I(Ca,L) ) and vascular dysfunction in a mouse model of type I diabetes. EXPERIMENTAL APPROACH: Changes in isometric tension were recorded on myograph. Ca(2+) currents in freshly dissociated mice aortic SMCs were measured using the whole-cell patch-clamp technique. Antisense techniques were used to knock-down the PI3Kδ isoform. KEY RESULTS Contractile responses to phenylephrine and KCl were strongly enhanced in diabetic aorta independent of a functional endothelium. The magnitude of phenylephrine-induced I(Ca,L) was also greatly augmented. PI3Kδ expression, but not PI3Kα, PI3Kß, PI3Kγ, was increased in diabetic aortas and treatment of vessels with a selective PI3Kδ inhibitor normalized I(Ca,L) and contractile response of diabetic vessels. Moreover, knock-down of PI3Kδin vivo decreased PI3Kδ expression and normalized I(Ca,L) and contractile response of diabetic vessels ex vivo. CONCLUSIONS AND IMPLICATIONS: Phosphatidylinositol 3-kinase δ was essential to the increased vascular contractile response in our model of type I diabetes. PI3Kδ signalling was up-regulated and most likely accounted for the increased I(Ca,L,) leading to increased vascular contractility. Blockade of PI3Kδ may represent a novel therapeutic approach to treat vascular dysfunction in diabetic patients.


Subject(s)
Calcium Channels, L-Type/metabolism , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Type 1/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Animals , Aorta/physiopathology , Calcium/metabolism , Class I Phosphatidylinositol 3-Kinases , Diabetes Mellitus, Experimental/physiopathology , Diabetes Mellitus, Type 1/physiopathology , Male , Mice , Mice, Inbred C57BL , Muscle Contraction , Muscle, Smooth, Vascular/physiopathology , Myocytes, Smooth Muscle/physiology , Patch-Clamp Techniques , Phosphoinositide-3 Kinase Inhibitors , Receptors, Adrenergic, alpha-1/metabolism , Signal Transduction , Up-Regulation , Vasoconstriction , Vasodilation
5.
Biomed Pharmacother ; 64(3): 220-5, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20053525

ABSTRACT

To investigate the effects of metformin on angiogenesis, on inflammatory cell accumulation and on production of endogenous cytokines in sponge implant in mice. Polyester-polyurethane sponges were implanted in Swiss mice and metformin (40 or 400mg/kg/day) was given orally for six days. The implants collected at day 7 postimplantation were processed for the assessment of hemoglobin (Hb), myeloperoxidase (MPO), N-acetylglucosaminidase (NAG) e collagen used as indexes for angiogenesis, neutrophil and macrophage accumulation and extracellular matrix deposition, respectively. Relevant inflammatory, angiogenic and fibrogenic cytokines were also determined. Metformin treatment attenuated the main components of the fibrovascular tissue, wet weight, vascularization (Hb content), macrophage recruitment (NAG activity), collagen deposition and the levels of transforming growth factor (TGF-beta1) intraimplant. A regulatory function of metformin on multiple parameters of main components of inflammatory angiogenesis has been revealed giving insight into the potential therapeutic underlying the actions of metformin.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Anti-Inflammatory Agents/therapeutic use , Foreign-Body Reaction/drug therapy , Inflammation/prevention & control , Metformin/therapeutic use , Neovascularization, Pathologic/prevention & control , Angiogenesis Inhibitors/pharmacology , Animals , Anti-Inflammatory Agents/pharmacology , Chemokine CCL2/biosynthesis , Chemokine CCL2/genetics , Chemotaxis, Leukocyte/drug effects , Collagen/biosynthesis , Collagen/genetics , Drug Evaluation, Preclinical , Foreign-Body Reaction/metabolism , Foreign-Body Reaction/physiopathology , Inflammation/physiopathology , Male , Metformin/pharmacology , Mice , Peroxidase/analysis , Surgical Sponges , Transforming Growth Factor beta1/biosynthesis , Transforming Growth Factor beta1/genetics , Tumor Necrosis Factor-alpha/biosynthesis , Tumor Necrosis Factor-alpha/genetics , Vascular Endothelial Growth Factor A/biosynthesis , Vascular Endothelial Growth Factor A/genetics
6.
Biomed Pharmacother ; 64(1): 29-34, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19811885

ABSTRACT

While compelling evidence indicates beneficial effects of statins on inflammatory processes, besides their cholesterol-lowering activities, the actions on angiogenesis are less clear-cut. Our aim was to investigate the effects of atorvastatin on key components of inflammatory angiogenesis in the murine sponge model. Polyester-polyurethane sponges, used as a framework for fibrovascular tissue growth, were implanted in Swiss mice. Atorvastatin (0.6, 3 mg/kg/day) was given orally for 8 days in drinking water. The implants collected at day 9 postimplantation were processed for the assessment of hemoglobin, myeloperoxidase (MPO), N-acetylglucosaminidase (NAG) and collagen. Relevant inflammatory, angiogenic and fibrogenic cytokines were also determined. Atorvastatin treatment resulted in significant decrease in sponge vascularization (Hb content) and in VEGF levels at both doses. Neutrophil influx (MPO activity) was not affected by the compound whereas macrophage recruitment (NAG activity) was inhibited, suggesting a degree of selectivity by atorvastatin for this cell population. The level of CCL2 (MCP1-JE) was decreased only with 0.6 mg/kg. Atorvastatin was also able to reduce collagen deposition and the levels of transforming growth factor (TGF-beta1) intraimplant, dose-dependently. The inhibitory function of atorvastatin on multiple parameters of main components of inflammatory angiogenesis revealed in this study is clearly associated with the modulatory effects of HMG-CoA reductase on VEGF, TNF-alpha and TGF-beta1 production.


Subject(s)
Down-Regulation/drug effects , Heptanoic Acids/pharmacology , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Neovascularization, Pathologic/drug therapy , Pyrroles/pharmacology , Administration, Oral , Animals , Atorvastatin , Disease Models, Animal , Dose-Response Relationship, Drug , Heptanoic Acids/administration & dosage , Hydroxymethylglutaryl-CoA Reductase Inhibitors/administration & dosage , Inflammation/drug therapy , Inflammation/physiopathology , Macrophages/drug effects , Macrophages/metabolism , Male , Mice , Neovascularization, Pathologic/physiopathology , Pyrroles/administration & dosage , Transforming Growth Factor beta1/drug effects , Transforming Growth Factor beta1/genetics , Tumor Necrosis Factor-alpha/drug effects , Tumor Necrosis Factor-alpha/genetics , Vascular Endothelial Growth Factor A/drug effects , Vascular Endothelial Growth Factor A/genetics
7.
Life Sci ; 81(3): 210-7, 2007 Jun 27.
Article in English | MEDLINE | ID: mdl-17588613

ABSTRACT

Tumor growth is associated with angiogenesis and inflammation and the endogenous lipid, platelet activating factor (PAF), is a pro-inflammatory and pro-angiogenic mediator. We therefore measured tumor growth, angiogenesis and inflammation in normal (WT) mice and those lacking the receptor for PAF, through gene deletion (PAFR-KO). Growth of solid tumors derived from colon 26 cells was not altered but that from Ehrlich cells was markedly (5-fold) increased in the PAFR-KO mice, relative to the WT strain. Angiogenesis, as tumor content of VEGF or hemoglobin, was increased in both tumors from the mutant strain. Inflammation, as neutrophil and macrophage accumulation and chemokine (CXCL2 and CCL2) content of tumors, was decreased or unchanged in the tumors implying an overall decrease in the inflammatory response in the PAFR-KO strain. We also assessed growth of the Ehrlich tumor in its ascites form, after i.p. injection. Here growth (ascites volume) was inhibited by about 30%, but neutrophil and macrophage numbers were increased in the ascites fluid from the PAFR-KO mice. Angiogenesis in the peritoneal wall, which is not invaded by the tumor cells, was increased but leukocyte infiltration decreased in the mutant strain. Our results show, unexpectedly, that tumor-induced angiogenesis was increased in mice lacking response to PAF, from which we infer that in normal (WT) mice, PAF is anti-angiogenic. Further, although growth was still associated with angiogenesis in PAFR-KO mice, growth was not correlated with inflammation (leukocyte accumulation).


Subject(s)
Inflammation/pathology , Neoplasms/pathology , Neovascularization, Pathologic/pathology , Platelet Membrane Glycoproteins/physiology , Receptors, G-Protein-Coupled/physiology , Acetylglucosaminidase/metabolism , Animals , Ascites/metabolism , Carcinoma, Ehrlich Tumor/pathology , Chemokine CCL2/biosynthesis , Chemokines, CXC/biosynthesis , Inflammation/genetics , Mice , Mice, Inbred BALB C , Mice, Knockout , Neoplasm Transplantation , Neoplasms/blood supply , Neoplasms/genetics , Neovascularization, Pathologic/genetics , Peritoneal Cavity , Peroxidase/metabolism , Platelet Activating Factor/pharmacology , Platelet Activating Factor/physiology , Platelet Membrane Glycoproteins/genetics , Receptors, G-Protein-Coupled/genetics , Regional Blood Flow/genetics , Regional Blood Flow/physiology , Tumor Necrosis Factor-alpha/metabolism , Vascular Endothelial Growth Factor A/metabolism
8.
Histol Histopathol ; 21(12): 1263-70, 2006 12.
Article in English | MEDLINE | ID: mdl-16977576

ABSTRACT

The integration of implanted material to host organism requires spatial and temporal organization of several cellular processes, such as proliferation, differentiation and apoptosis. Despite the clinical relevance of these processes, there is little information regarding the sequence of such events in synthetic matrices. Here, we present a combination of techniques used to characterize the fibrovascular response in subcutaneous polyether-polyurethane sponge implants in mice at days 4, 7, 10 and 14 postimplantation. The AgNOR technique was modified and used as a surrogate marker for proliferating and activated cells invading the implant. The number of AgNOR-stained cells increased progressively from day 4 (606+/-76) to day 14 (2146+/-71) postimplantation. The number of TUNEL-positive (apoptotic index) cells also increased progressively from day 4 (459+/-40) to day 14 (1157+/-119) postimplantation. However, the ratio of TUNEL-labeled/proliferating cells had its highest peak in the early phase of the process remaining stable until day 14. Using Picrosirius staining it was shown that thin collagen increased from day 4, peaking at day 10 and falling markedly at day 14, whereas dense collagen increased progressively during the whole period. These experiments hold potential to investigate not only distinct phases of tissue repair induced by synthetic matrices but also to study underlying mechanisms involved.


Subject(s)
Collagen/metabolism , Endothelium, Vascular/cytology , Fibroblasts/cytology , Inflammation/pathology , Surgical Sponges/adverse effects , Wound Healing/immunology , Animals , Apoptosis , Biomarkers/analysis , Cell Differentiation , Cell Proliferation , Chemotaxis, Leukocyte , Implants, Experimental/adverse effects , Male , Mice , Mice, Inbred BALB C , Models, Animal , Neovascularization, Physiologic , Neutrophils , Polyurethanes
9.
Life Sci ; 78(14): 1637-45, 2006 Feb 28.
Article in English | MEDLINE | ID: mdl-16313924

ABSTRACT

This study addresses the interaction between Ehrlich ascites tumor and skeletal abdominal muscle, presenting quantitative analysis of ascites-induced angiogenesis and inflammation in this tissue of mice bearing-tumor. Time-dependent changes in the muscle (cellular activity, angiogenesis, inflammation and cytokines production) were assessed by morphometric, functional, and biochemical parameters at days 1, 4 and 8 after i.p. inoculation of Ehrlich tumor cells (2.5 x 10(7)). The number of cells stained with AgNOR technique (argyrophilic nucleolar organizer region) in the muscle, together with MTS assay used as markers of cellular activity increased progressively in parallel with the out flow rate of sodium fluorescein (blood flow index), hemoglobin content (vascular index) and VEGF production. Likewise, the inflammatory process in the muscle, as assessed by myeloperoxidase (MPO) and n-acethylglucosaminidase (NAG) activities and the levels of the chemokines, keratinocyte-derived chemokine (CXC1-3/KC) and macrophage-chemoattractant protein (CCL2/MCP-1) increased with tumor development. The combination of techniques used to describe angiogenesis and inflammation in a muscle model system has proved to be suited for quantitative measurements of microvascular changes and cellular infiltration occurring in the abdominal muscle wall of ascites-bearing mice. This study holds potential for investigating events and mechanisms associated with skeletal muscle response to neoplasic stimulus.


Subject(s)
Carcinoma, Ehrlich Tumor/complications , Muscle, Skeletal/blood supply , Myositis/etiology , Neovascularization, Pathologic/etiology , Animals , Cytokines/analysis , Cytokines/metabolism , Hemoglobins/analysis , Male , Mice , Mice, Inbred BALB C , Muscle, Skeletal/chemistry , Muscle, Skeletal/pathology , Myositis/pathology , Neovascularization, Pathologic/pathology
10.
Microcirculation ; 12(7): 597-606, 2005.
Article in English | MEDLINE | ID: mdl-16207631

ABSTRACT

OBJECTIVES: A wide range of compounds inhibit formation of new blood vessels in a variety of models, accompanied by decreases in pro-angiogenic cytokines. The authors sought a surrogate marker for the complex process of neovascularization by correlating inhibition of cytokine production with anti-angiogenic effect. METHODS: Three anti-angiogenic compounds, clotrimazole (120 mg kg(-1) day(-1)), thalidomide (100 mg kg(-1) day(-1)), and rosiglitazone (10 mg kg(-1) day(-1)), were used to inhibit angiogenesis developing over 9 days, in sponges implanted subcutaneously in Swiss mice. Angiogenesis was assessed by hemoglobin content and by histology. Content of cytokines in implants was measured by specific immunoassays and accumulation of neutrophils or macrophages in implants by measuring myeloperoxidase or N-acetylglucosaminidase activity, respectively. RESULTS: These compounds caused equal inhibition of angiogenesis (about 40%). However, implant levels of vascular endothelial growth factor (VEGF), tumor necrosis factor (TNF-alpha) or the macrophage chemoattractant cytokine, CCL2/MCP-1/JE, and accumulation of macrophages were more variably inhibited. Only the neutrophil chemokine, CXCL2/KC, was inhibited equally by the three compounds, in this model. CONCLUSIONS: Anti-angiogenic effect was most clearly and closely correlated with levels of the chemokine KC. Thus, measurement of the chemokine KC might provide an adequate surrogate marker for the functional process of neovascularization in our model.


Subject(s)
Angiogenesis Inhibitors/administration & dosage , Cytokines/metabolism , Granulation Tissue/metabolism , Neovascularization, Pathologic/metabolism , Animals , Biomarkers/metabolism , Chemokine CXCL1 , Chemokines , Chemokines, CXC , Inflammation/metabolism , Male , Mice , Neutrophil Infiltration/drug effects
11.
J Leukoc Biol ; 78(2): 352-8, 2005 Aug.
Article in English | MEDLINE | ID: mdl-15894588

ABSTRACT

The majority of biological responses classically attributed to tumor necrosis factor alpha (TNF-alpha) is mediated by p55 receptor (TNFR1). Here, we aimed to clarify the biological role of TNFR1-mediated signals in an in vivo inflammatory angiogenesis model. Polyester-polyurethane sponges, used as a framework for tissue growth, were implanted in C57Bl/6 mice. These implants were collected at days 1, 7, and 14 post-implant for enzyme-linked immunosorbent assay or at days 7 and 14 for hemoglobin, myeloperoxidase, and N-acetylglucosaminidase measurements, used as indexes for angiogenesis, neutrophil, and macrophage accumulation, respectively. In TNFR1-deficient C57Bl/6 mice, there was a significant decrease in sponge vascularization but not in late inflammatory cell influx. It is interesting that levels of vascular endothelial growth factor were significantly lower in TNFR1-deficient than in wild-type mice at days 1 and 7. Levels of angiogenic chemokines, CC chemokine ligand 2/murine homologue of monocyte chemoattractant protein-1 and CXC chemokine ligand 1-3/keratinocyte-derived chemokine, were significantly lower in TNFR1-deficient mice at days 1 and 7 after implantation, respectively. These observations suggest that TNFR1-mediated signals have a critical role in sponge-induced angiogenesis, possibly by influencing the effector state of inflammatory cells and hence, modulating the angiogenic molecular network.


Subject(s)
Neovascularization, Pathologic/metabolism , Receptors, Tumor Necrosis Factor, Type I/metabolism , Signal Transduction , Acetylglucosaminidase/analysis , Animals , Chemokines/metabolism , Female , Hemoglobins/analysis , Inflammation/genetics , Inflammation/metabolism , Inflammation/pathology , Macrophages/metabolism , Mice , Mice, Knockout , Neutrophil Infiltration/genetics , Peroxidase/analysis , Polyesters , Polyurethanes , Prostheses and Implants , Receptors, Tumor Necrosis Factor, Type I/genetics , Signal Transduction/genetics , Tumor Necrosis Factor-alpha/metabolism
12.
Inflamm Res ; 53(10): 576-84, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15597153

ABSTRACT

OBJECTIVE: Using the murine sponge model, we investigated the temporal relationship between angiogenesis, leukocyte accumulation and endogenous generation of the pro-inflammatory chemokines CXCL1-3/KC and CCL2/JE. Furthermore, the effects of exogenous administration of these chemokines were studied. METHODS: Angiogenesis in the implants was assessed by measuring the hemoglobin content (vascular index) and leukocyte accumulation quantified by evaluating MPO and NAG enzyme activities. RESULTS: A progressive increase in hemoglobin content and in enzymatic activities was observed during the whole period. The levels of CXCL1-3/KC and CCL2/JE in the implants peaked at days 7 and 1, respectively. Exogenous administration of CXCL1-3/KC (100 ng/day intra-implant) applied at days 1-3 resulted in increased neovascularization and macrophage accumulation. Intra-implant injections of CCL2/JE (100 ng/day) also resulted in increased angiogenesis and macrophage accumulation. CONCLUSIONS: These results demonstrated that the chemokines, CXCL1-3/KC and CCL2/JE, are generated within the sponge compartment and that neovascularization and inflammatory cells influx can be modulated by exogenous administration of the chemokines.


Subject(s)
Chemokine CCL2/biosynthesis , Chemokines, CXC/biosynthesis , Intercellular Signaling Peptides and Proteins/biosynthesis , Neovascularization, Pathologic , Acetylglucosaminidase/chemistry , Animals , Chemokine CXCL1 , Chemokines/metabolism , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Hemoglobins/metabolism , Immunohistochemistry , Inflammation , Kinetics , Leukocytes/cytology , Leukocytes/immunology , Male , Mice , Mice, Inbred BALB C , Neutrophils/metabolism , Peroxidase/metabolism , Recombinant Proteins/chemistry , Time Factors
13.
Life Sci ; 74(23): 2827-37, 2004 Apr 23.
Article in English | MEDLINE | ID: mdl-15050421

ABSTRACT

We investigated angiogenesis, inflammatory cells accumulation and endogenous production of cytokines in sponge implants of tumor-bearing mice. Seven days after inoculation of Ehrlich tumor cells (2.5 x 10(6)), sponge discs were implanted subcutaneously in the dorsa of mice to induce the formation of fibrovascular tissue. The implants of tumor-bearing and non tumor-bearing animals were assessed for neovascularization and leukocyte accumulation, together with levels of relevant cytokines, vascular endothelial growth factor VEGF), tumor necrosis factor alpha (TNF-alpha), CXCL1-3/KC and CCL2/JE. In the implants of tumor-bearing animals angiogenesis (assessed by hemoglobin content and VEGF levels in the implants) and leukocyte accumulation (assessed by myeloperoxidase -MPO- and N- acetylglucosaminidase-NAG-enzyme activities) were all significantly less than those in the implants of non tumor-bearing animals. Although the chemokine CXCL1-3/KC was lower in the implants of tumor-bearing animals, the chemokine CCL2/JE was increased in this group. The production of TNF-alpha in the implants was not modified by the presence of the subcutaneous tumor. The combination of the methodologies used in this study has provided a novel approach to investigate the interaction between two distinct proliferating tissues that share common features (angiogenesis, cell recruitment, inflammation) and has shown that the predominant inhibitory effect of a tumor mass over repair process is associated with altered cytokine production.


Subject(s)
Angiogenesis Inhibitors/metabolism , Carcinoma, Ehrlich Tumor/metabolism , Chemotaxis, Leukocyte/physiology , Granulation Tissue/blood supply , Neovascularization, Pathologic , Animals , Carcinoma, Ehrlich Tumor/pathology , Chemokines/metabolism , Granulation Tissue/metabolism , Granulation Tissue/pathology , Leukocytes, Mononuclear/enzymology , Leukocytes, Mononuclear/pathology , Male , Mice , Prostheses and Implants , Surgical Sponges , Vascular Endothelial Growth Factor A/metabolism
14.
Regul Pept ; 107(1-3): 105-13, 2002 Jul 15.
Article in English | MEDLINE | ID: mdl-12137972

ABSTRACT

Angiotensin-(1-7) (Ang-(1-7)), a peptide constituent of the renin-angiotensin system, has been shown to act as a vasodilator mediator in pre-existing (skin) and newly formed vasculatures (14-day-old sponge implants). Blood flow was determined by the outflow rate of sodium fluorescein applied intradermally or intraimplant and the results were expressed in t(1/2) values (time taken for the fluorescence to reach 50% of the peak in the systemic circulation). We showed that the t(1/2) value was significantly lower (4.1+/-0.46) in the implants compared with the cutaneous vasculature (5.7+/-0.5). Ang-(1-7) 20 ng was able to decrease t(1/2) values in both vasculatures. The specific receptor antagonist, D-Ala7-Ang-(1-7) (A-779), prevented Ang-(1-7)-induced vasodilation and altered the basal vascular tone of the implants. The vasodilator effect was also abolished by nitric oxide (NO) synthase inhibitors in both vasculatures and by indomethacin in the implant. Selective AT(1) and AT(2) receptor antagonists did not alter the vasodilation induced by the peptide. These results establish the vasodilator effect of Ang-(1-7) in the cutaneous and implant vasculature and that the peptide is produced endogenously by the fibrovascular tissue, and suggest that this peptide contributes for the vasodilation found in newly formed vascular beds (wound healing, chronic inflammatory processes and tumors).


Subject(s)
Angiotensin I/pharmacology , Neovascularization, Physiologic/drug effects , Peptide Fragments/pharmacology , Skin/blood supply , Vasodilator Agents/pharmacology , Angiotensin Receptor Antagonists , Animals , Blood Flow Velocity/drug effects , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Fluorescein/pharmacokinetics , Male , Mice , Neovascularization, Physiologic/physiology , Surgical Sponges , Tissue Distribution , Vasodilation/drug effects , Vasomotor System/drug effects
15.
Inflammation ; 25(2): 91-6, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11321364

ABSTRACT

Thalidomide, clinically used as an antiinflammatory and antitumoral drug, inhibited sponge-induced angiogenesis when administered systemically (100 mg/kg(-1)) in mice. However, it failed to inhibit solid Ehrlich tumor in the same mouse strain. We have used functional, biochemical and histological parameters to assess neovascularization and fibrovascular tissue infiltration of the mice sponge granuloma. The neovascularization growth as detected by development of blood flow and hemoglobin content extracted from the implants showed that thalidomide inhibited fibrovascular tissue formation by 40%. The functional and biochemical parameters correlated well with the histological study. Thalidomide had no inhibitory effect in the development of Ehrlich tumor. The detection of this selective action using the same animal strain bearing two different processes, supports the hypothesis that rather than species specificity, thalidomide is tissue specific. This approach may be used to identify the specificity of other therapeutic agents against distinct angiogenesis-dependent diseases.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Carcinoma, Ehrlich Tumor/blood supply , Carcinoma, Ehrlich Tumor/drug therapy , Neovascularization, Pathologic/drug therapy , Thalidomide/pharmacology , Animals , Carcinoma, Ehrlich Tumor/pathology , Granuloma, Foreign-Body/drug therapy , Granuloma, Foreign-Body/etiology , Granuloma, Foreign-Body/pathology , Male , Mice , Neovascularization, Pathologic/pathology
16.
Am J Physiol Regul Integr Comp Physiol ; 280(4): R994-R1000, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11247819

ABSTRACT

Angiotensin-(1-7) [ANG-(1-7)], an endogenous bioactive peptide constituent of the renin-angiotensin system, acts as an inhibitory growth factor in vitro and in vivo. In this study, we evaluated whether the antiangiogenic effect of ANG-(1-7) in the mouse sponge model of angiogenesis might be receptor mediated and involved in the release of nitric oxide (NO). The hemoglobin content (microg/mg wet tissue) of 7-day-old sponge implants was used as an index of the vascularization and showed that daily injections of ANG-(1-7) (20 ng) inhibited significantly the angiogenesis in the implants relative to the saline-treated group. The specific receptor antagonist D-Ala(7)-ANG-(1-7); A-779 prevented ANG-(1-7)-induced inhibition of angiogenesis. The antiangiogenic effect was also abolished by pretreatment with NO synthase inhibitors aminoguanidine (1 mg/ml) or N(G)-nitro-L-arginine methyl ester (0.3 mg/ml). Selective AT1 and AT2 angiotensin-receptor antagonists and an angiotensin-converting enzyme inhibitor, in combination with ANG-(1-7) or alone, did not alter angiogenesis in the implants. These results establish that the regulation of the vascular tissue growth by ANG-(1-7) is associated with NO release by activation of an angiotensin receptor distinct from AT1 and AT2.


Subject(s)
Angiotensin II/analogs & derivatives , Angiotensin I/pharmacology , Antihypertensive Agents/pharmacology , Neovascularization, Pathologic/prevention & control , Neovascularization, Physiologic/drug effects , Peptide Fragments/pharmacology , Analysis of Variance , Angiogenesis Inhibitors/pharmacology , Angiotensin II/pharmacology , Angiotensin Receptor Antagonists , Animals , Imidazoles/pharmacology , Losartan/pharmacology , Male , Mice , Neovascularization, Physiologic/physiology , Prostheses and Implants , Pyridines/pharmacology , Receptor, Angiotensin, Type 1 , Receptor, Angiotensin, Type 2
17.
Methods Mol Med ; 46: 77-86, 2001.
Article in English | MEDLINE | ID: mdl-21340913

ABSTRACT

The process of capillary growth, angiogenesis, is an integral part of wound healing and repair mechanisms. When it occurs during these conditions, it is tightly controlled and strictly delimited. However, in tumor growth and in a variety of vascular diseases, unrestrained angiogenesis can contribute significantly to the pathology and persistence of these manifestations (1). Research on angiogenesis was initiated with the development of several bioassays that have permitted direct observations of the microvasculature in the living animal. The bioassays have been used for a variety of purposes; for example, to detect angiogenesis activity in malignant and normal cells and tissue, to screen purified test substances for angiogenic activity, and to elucidate the cellular events that accompany vessel growth. The response observed after the introduction of an appropriate stimulus such as mechanical injury or injection of neoplastic tissue implants has allowed the cataloging of the main events of the angiogenic cascade as well as the characterization of pro- and antiangiogenic factors.

18.
Regul Pept ; 91(1-3): 45-62, 2000 Jul 28.
Article in English | MEDLINE | ID: mdl-10967201

ABSTRACT

The renin-angiotensin system is a major physiological regulator of arterial pressure and hydro-electrolyte balance. Evidence has now been accumulated that in addition to angiotensin (Ang) II other Ang peptides [Ang III, Ang IV and Ang-(1-7)], formed in the limited proteolysis processing of angiotensinogen, are importantly involved in mediating several actions of the RAS. In this article we will review our knowledge of the biological actions of Ang-(1-7) with focus on the puzzling aspects of the mediation of its effects and the interaction Ang-(1-7)-kinins. In addition, we will attempt to summarize the evidence that Ang-(1-7) takes an important part of the mechanisms aimed to counteract the vasoconstrictor and proliferative effects of Ang II.


Subject(s)
Angiotensin II/physiology , Peptide Fragments/physiology , Angiotensin I , Angiotensin II/metabolism , Animals , Blood Vessels/metabolism , Central Nervous System/metabolism , Humans , Kidney/metabolism , Losartan/pharmacology , Models, Biological , Myocardium/metabolism , Peptide Fragments/metabolism , Peptidyl-Dipeptidase A/metabolism , Protein Binding , Receptor, Angiotensin, Type 1 , Receptor, Angiotensin, Type 2 , Receptors, Angiotensin/metabolism , Signal Transduction , Vasoconstriction/physiology
19.
Microvasc Res ; 59(2): 278-85, 2000 Mar.
Article in English | MEDLINE | ID: mdl-10684733

ABSTRACT

Angiogenesis and tumor growth in conscious mice have been determined using the kinetics of appearance of a fluorescent tracer in the bloodstream after application to subcutaneously implanted sponges bearing tumor cells. The functional parameter expressed in terms of half-time (t(1/2); time taken for the fluorescence to reach 50% of the peak in the systemic circulation), which is inversely proportional to blood flow, showed that in the tumor-free implants t(1/2) values decreased from 11.55 +/- 1 min at day 1 to 5.7 +/- 0. 44 min by day 14. In the tumor-bearing implants, this process was accelerated and maximum vascularization was achieved by day 7 (3 days after tumor cell inoculation). Increases in t(1/2) values were observed at days 10 and 14, which paralleled the tumor growth as indicated by wet weight. The hemoglobin content (microg Hb/mg wet weight) in the tumor-free group increased during the 14-day period. In contrast, in the tumor-bearing implants. Hb concentration decreased per unit of tissue weight. Dexamethasone treatment for 13 days prevented fibrovascular tissue infiltration in tumor-free implants, but was unable to delay tumor growth, indicating that this procedure can be used to exclude the inflammatory reaction induced by the implantation technique, thus allowing tumor angiogenesis to be studied without the confounding influence of the host inflammatory cells. The results of our experimental observation indicate the suitability of this combination of techniques for analyzing angiogenesis induced by tumor cells and several hemodynamic features of Ehrlich tumor growth in awake animals.


Subject(s)
Carcinoma, Ehrlich Tumor/blood supply , Fluorometry/methods , Neovascularization, Pathologic , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Carcinoma, Ehrlich Tumor/pathology , Consciousness , Dexamethasone/pharmacology , Dexamethasone/therapeutic use , Fluorescein , Fluorescent Dyes , Hemodynamics , Male , Mice , Prostheses and Implants , Surgical Sponges
20.
Life Sci ; 66(1): 67-76, 2000.
Article in English | MEDLINE | ID: mdl-10658925

ABSTRACT

Using the murine sponge model of angiogenesis, associated to functional and morphological parameters we have demonstrated opposing actions of angiotensin II (Ang II) and angiotensin-(1-7;Ang-1-7) in modulating fibrovascular tissue growth. Angiogenesis in the implants was assessed at day 7 postimplantation by extracting the hemoglobin content, by determining the outflow rate of sodium fluorescein applied intraimplant and by histological analysis. Furthermore, the proliferative activity of control and angiotensin-treated implants was established using the MTS (3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2(4 -sulfonyl)2H-tetrazolium)assay. The hemoglobin content in the control implants was 2.4 +/- 0.14 (microg/mg wet weight) versus 3.6 +/- 0.27(Ang II;100 ng) and 0.86 +/- 0.07 Ang-(1-7); 20 ng. Blood flow in the implants as determined by t1/2 values (time taken for the fluorescence to reach 50% of the peak in the systemic circulation) showed that Ang II stimulated angiogenesis, whereas Ang-(1-7) inhibited it. The proliferative activity of the sponge-induced fibrovascular tissue was enhanced by Ang II and diminished by Ang-(1-7). These results show the pro-versus anti-angiogenic effects of these angiotensin molecules, providing evidence for their opposing effects on vascular tissue growth and wound healing in vivo.


Subject(s)
Angiotensin II/physiology , Neovascularization, Physiologic , Wound Healing/physiology , Angiotensin I , Animals , Cell Division , Male , Mice , Peptide Fragments/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...