Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
1.
PLoS One ; 12(11): e0188204, 2017.
Article in English | MEDLINE | ID: mdl-29145462

ABSTRACT

Inflammation is associated with every health condition, and is an important component of many pathologies such as cardiovascular diseases. Circulating levels of soluble endoglin have been shown to be higher in the serum of patients with cardiovascular diseases with a significant inflammatory component. The aim of this study was to evaluate the implication of circulating soluble endoglin in the inflammatory response. For this purpose, a transgenic mouse expressing human soluble endoglin (sEng+) was employed, and three different inflammatory approaches were used to mimic inflammatory conditions in different tissues. This study shows that control sEng+ mice have a normal inflammatory state. The lung and kidney injury induced by the inflammatory agents was reduced in sEng+ mice, especially the intra-alveolar and kidney infiltrates, suggesting a possible reduction in inflammation induced by soluble endoglin. To deepen into this possible effect, the leukocyte number in the bronchoalveolar lavage and air pouch lavage was evaluated and a significant reduction of neutrophil infiltration in LPS-treated lungs and ischemic kidneys from sEng+ with respect to WT mice was observed. Additionally, the mechanisms through which soluble endoglin prevents inflammation were studied. We found that in sEng+ animals the increment of proinflammatory cytokines, TNFα, IL1ß and IL6, induced by the inflammatory stimulus was reduced. Soluble endoglin also prevents the augmented adhesion molecules, ICAM, VCAM and E-selectin induced by the inflammatory stimulus. In addition, vascular permeability increased by inflammatory agents was also reduced by soluble endoglin. These results suggest that soluble endoglin modulates inflammatory-related diseases and open new perspectives leading to the development of novel and targeted approaches for the prevention and treatment of cardiovascular diseases.


Subject(s)
Endoglin/blood , Inflammation/blood , Acute Lung Injury/blood , Acute Lung Injury/chemically induced , Animals , Bronchoalveolar Lavage Fluid , Capillary Permeability , Cell Adhesion Molecules/metabolism , Cytokines/metabolism , Inflammation Mediators/metabolism , Lipopolysaccharides/pharmacology , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Transgenic
2.
Pharmacol Ther ; 167: 132-145, 2016 11.
Article in English | MEDLINE | ID: mdl-27490717

ABSTRACT

Pre-renal acute kidney injury (AKI) results from glomerular haemodynamic alterations leading to reduced glomerular filtration rate (GFR) with no parenchymal compromise. Renin-angiotensin system inhibitors, such as angiotensin-converting enzyme inhibitors (ACEIs), angiotensin receptor antagonists (ARAs), non-steroidal anti-inflammatory drugs (NSAIDs) and diuretics, are highly prescribed drugs that are frequently administered together. Double and triple associations have been correlated with increased pre-renal AKI incidence, termed "double whammy" and "triple whammy", respectively. This article presents an integrative analysis of the complex interplay among the effects of NSAIDs, ACEIs/ARAs and diuretics, acting alone and together in double and triple therapies. In addition, we explore how these drug combinations alter the equilibrium of regulatory mechanisms controlling blood pressure (renal perfusion pressure) and GFR to increase the odds of inducing AKI through the concomitant reduction of blood pressure and distortion of renal autoregulation. Using this knowledge, we propose a more general model of pre-renal AKI based on a multi whammy model, whereby several factors are necessary to effectively reduce net filtration. The triple whammy was the only model associated with pre-renal AKI accompanied by a course of other risk factors, among numerous potential combinations of clinical circumstances causing hypoperfusion in which renal autoregulation is not operative or is deregulated. These factors would uncouple the normal BP-GFR relationship, where lower GFR values are obtained at every BP value.


Subject(s)
Acute Kidney Injury/etiology , Blood Pressure/physiology , Models, Theoretical , Acute Kidney Injury/epidemiology , Acute Kidney Injury/physiopathology , Angiotensin Receptor Antagonists/administration & dosage , Angiotensin Receptor Antagonists/adverse effects , Angiotensin-Converting Enzyme Inhibitors/administration & dosage , Angiotensin-Converting Enzyme Inhibitors/adverse effects , Animals , Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Anti-Inflammatory Agents, Non-Steroidal/adverse effects , Blood Pressure/drug effects , Diuretics/administration & dosage , Diuretics/adverse effects , Glomerular Filtration Rate , Humans , Incidence , Renin-Angiotensin System/drug effects , Risk Factors
3.
Cell Physiol Biochem ; 37(4): 1231-9, 2015.
Article in English | MEDLINE | ID: mdl-26431052

ABSTRACT

BACKGROUND/AIMS: Defective tissue repair underlies renal tissue degeneration during chronic kidney disease (CKD) progression. Unbalanced presence of TGF-ß opposes effective cell proliferation and differentiation processes, necessary to replace damaged epithelia. TGF-ß also retains arrested cells in a fibrotic phenotype responsible for irreversible scarring. In order to identify prospective molecular targets to prevent the effect of TGF-ß during CKD, we studied the signaling pathways responsible for the antiproliferative effect of this cytokine. METHODS: Tubule epithelial HK2 and MDCK cells were treated with TGF-ß (or not as control) to study cell proliferation (by MTT), cell signaling (by Western blot), cell cycle (by flow cytometry) and apoptosis (DNA fragmentation). RESULTS: TGF-ß fully activates the ALK-5 receptor pathway, whereas it has no effect on the ALK-1 and MAPK pathways in both HK2 and MDCK cells. Interestingly, TGF-ß exerts an antiproliferative effect only on MDCK cells, through a cytostatic effect in G0/G1. Inhibition of the ALK-5 pathway with SB431542 prevents the cytostatic effect of TGF-ß on MDCK cells. CONCLUSION: Activation of the ALK-5 pathway is not sufficient for the antiproliferative effect of TGF-ß. The presence of undetermined permissive conditions or absence of undetermined inhibitory conditions seems to be necessary for this effect. The ALK-5 pathway appears to provide targets to modulate fibrosis, but further research is necessary to identify critical circumstances allowing or inhibiting its role at modulating tubule epithelial cell proliferation and tubule regeneration in the context of CKD progression.


Subject(s)
Cell Proliferation/drug effects , Protein Serine-Threonine Kinases/metabolism , Receptors, Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta1/pharmacology , Animals , Benzamides/pharmacology , Cell Line , DNA Fragmentation/drug effects , Dioxoles/pharmacology , Dogs , Epithelial Cells/cytology , Epithelial Cells/drug effects , Epithelial Cells/metabolism , G1 Phase Cell Cycle Checkpoints/drug effects , Humans , Kidney Tubules/cytology , Madin Darby Canine Kidney Cells , Protein Serine-Threonine Kinases/antagonists & inhibitors , Receptor, Transforming Growth Factor-beta Type I , Receptors, Transforming Growth Factor beta/antagonists & inhibitors , Signal Transduction/drug effects , Smad2 Protein/metabolism
4.
Toxicol Lett ; 234(2): 99-109, 2015 Apr 16.
Article in English | MEDLINE | ID: mdl-25677510

ABSTRACT

Nephrotoxicity limits the therapeutic efficacy of the antineoplastic drug cisplatin. Due to dosage adjustment and appropriate monitoring, most therapeutic courses with cisplatin produce no or minimal kidney damage. However, we studied whether even sub-nephrotoxic dosage of cisplatin poses a potential risk for the kidneys by predisposing to acute kidney injury (AKI), specifically by lowering the toxicity threshold for a second nephrotoxin. With this purpose rats were treated with a single sub-nephrotoxic dosage of cisplatin (3mg/kg, i.p.) and after two days, with a sub-nephrotoxic regime of gentamicin (50mg/kg/day, during 6 days, i.p.). Control groups received only one of the drugs or the vehicle. Renal function and renal histology were monitored throughout the experiment. Cisplatin treatment did not cause any relevant functional or histological alterations in the kidneys. Rats treated with cisplatin and gentamicin, but not those under single treatments, developed an overt renal failure characterized by both renal dysfunction and massive tubular necrosis. In addition, the urinary excretion of fumarylacetoacetase was increased in cisplatin-treated animals at subtoxic doses, which might be exploited as a cisplatin-induced predisposition marker. In fact, the urinary level of fumarylacetoacetase prior to the second nephrotoxin correlated with the level of AKI triggered by gentamicin in predisposed animals.


Subject(s)
Acute Kidney Injury/chemically induced , Antineoplastic Agents/toxicity , Cisplatin/toxicity , Hydrolases/urine , Kidney/drug effects , Acute Kidney Injury/enzymology , Acute Kidney Injury/pathology , Acute Kidney Injury/physiopathology , Acute Kidney Injury/urine , Animals , Biomarkers/urine , Disease Models, Animal , Gentamicins , Humans , Kidney/enzymology , Kidney/pathology , Kidney/physiopathology , Kidney Tubular Necrosis, Acute/chemically induced , Kidney Tubular Necrosis, Acute/enzymology , Kidney Tubular Necrosis, Acute/urine , Male , Rats, Wistar , Risk Assessment , Risk Factors , Time Factors , Up-Regulation
5.
Nephron Extra ; 4(1): 1-7, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24575118

ABSTRACT

BACKGROUND/AIMS: Chronic kidney disease (CKD) is a progressive deterioration of the kidney function, which may eventually lead to renal failure and the need for dialysis or kidney transplant. Whether initiated in the glomeruli or the tubuli, CKD is characterized by progressive nephron loss, for which the process of tubular deletion is of key importance. Tubular deletion results from tubular epithelial cell death and defective repair, leading to scarring of the renal parenchyma. Several cytokines and signaling pathways, including transforming growth factor-ß (TGF-ß) and the Fas pathway, have been shown to participate in vivo in tubular cell death. However, there is some controversy about their mode of action, since a direct effect on normal tubular cells has not been demonstrated. We hypothesized that epithelial cells would require specific priming to become sensitive to TGF-ß or Fas stimulation and that this priming would be brought about by specific mediators found in the pathological scenario. METHODS: Herein we studied whether the combined effect of several stimuli known to take part in CKD progression, namely TGF-ß, tumor necrosis factor-α, interferon-γ (IFN-γ), and Fas stimulation, on primed resistant human tubular cells caused cell death or reduced proliferation. RESULTS: We demonstrate that these cytokines have no synergistic effect on the proliferation or viability of human kidney (HK2) cells. We also demonstrate that IFN-γ, but not the other stimuli, reduces the proliferation of cycloheximide-primed HK2 cells without affecting their viability. CONCLUSION: Our results point at a potentially important role of IFN-γ in defective repair, leading to nephron loss during CKD.

6.
Biochim Biophys Acta ; 1832(12): 2204-15, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23994610

ABSTRACT

Hepatocyte growth factor (HGF) and its receptor, Met, are key determinants of distinct developmental processes. Although HGF exerts cardio-protective effects in a number of cardiac pathologies, it remains unknown whether HGF/Met signaling is essential for myocardial development and/or physiological function in adulthood. We therefore investigated the requirement of HGF/Met signaling in cardiomyocyte for embryonic and postnatal heart development and function by conditional inactivation of the Met receptor in cardiomyocytes using the Cre-α-MHC mouse line (referred to as α-MHCMet-KO). Although α-MHCMet-KO mice showed normal heart development and were viable and fertile, by 6 months of age, males developed cardiomyocyte hypertrophy, associated with interstitial fibrosis. A significant upregulation in markers of myocardial damage, such as ß-MHC and ANF, was also observed. By the age of 9 months, α-MHCMet-KO males displayed systolic cardiac dysfunction. Mechanistically, we provide evidence of a severe imbalance in the antioxidant defenses in α-MHCMet-KO hearts involving a reduced expression and activity of catalase and superoxide dismutase, with consequent reactive oxygen species accumulation. Similar anomalies were observed in females, although with a slower kinetics. We also found that Met signaling down-regulation leads to an increase in TGF-ß production and a decrease in p38MAPK activation, which may contribute to phenotypic alterations displayed in α-MHCMet-KO mice. Consistently, we show that HGF acts through p38α to upregulate antioxidant enzymes in cardiomyocytes. Our results highlight that HGF/Met signaling in cardiomyocytes plays a physiological cardio-protective role in adult mice by acting as an endogenous regulator of heart function through oxidative stress control.


Subject(s)
Gene Expression Regulation, Developmental , Heart/physiopathology , Myocytes, Cardiac/metabolism , Oxidative Stress , Proto-Oncogene Proteins c-met/metabolism , Animals , Blotting, Western , Catalase/genetics , Catalase/metabolism , Cell Proliferation , Cells, Cultured , Cytochromes c/genetics , Cytochromes c/metabolism , Electrocardiography , Electron Transport Complex IV/genetics , Electron Transport Complex IV/metabolism , Female , Immunoenzyme Techniques , Integrases , Male , Mice , Mice, Knockout , Mice, Transgenic , Mitochondria/genetics , Mitochondria/metabolism , Myocytes, Cardiac/pathology , Proto-Oncogene Proteins c-met/antagonists & inhibitors , Proto-Oncogene Proteins c-met/genetics , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Reactive Oxygen Species/metabolism , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism , p38 Mitogen-Activated Protein Kinases/genetics , p38 Mitogen-Activated Protein Kinases/metabolism
7.
PLoS One ; 8(1): e53624, 2013.
Article in English | MEDLINE | ID: mdl-23341958

ABSTRACT

The functional mechanisms involved in angiogenesis and the potential role of endoglin (ENG), recently described as a new marker for this process, have not been explored in Myelodysplastic Syndromes (MDS). In order to gain insight in MDS angiogenesis a combined analysis in bone marrow (BM) of gene expression levels, angiogenesis-related soluble factors and functional angiogenesis-related studies was carried out. Ninety-seven MDS patients and forty-two normal BM samples were studied. The morphology of the capillary-like structures originated by two endothelial cells lines in the BM environment of patients with refractory cytopenia with multilineage dysplasia (RCMD) was different from those of the remaining MDS. In addition, the BM mononuclear cells from RCMD patients displayed over-expression of VEGF, HIF and FN1 while they showed reduced expression of ENG in contrast to the normal ENG expression of the remaining low-risk MDS and the high expression of ENG in high-risk MDS subtype. Moreover, higher soluble ENG and soluble FLT-1 levels in BM microenvironment were observed in RCMD cases, which distinguished them from other individuals. Therefore, the present study suggests that the patterns of angiogenesis are different between the MDS subtypes. The differences in angiogenesis observed in RCMD patients could be related to ENG abnormalities.


Subject(s)
Endothelial Cells/metabolism , Myelodysplastic Syndromes/pathology , Neovascularization, Pathologic/pathology , Anemia, Refractory/metabolism , Anemia, Refractory/pathology , Angiogenesis Inducing Agents/metabolism , Antigens, CD/genetics , Antigens, CD/metabolism , Bone Marrow/metabolism , Bone Marrow/pathology , Cell Lineage , Cell Proliferation , Cellular Microenvironment , Endoglin , Endothelial Cells/pathology , Humans , Myelodysplastic Syndromes/metabolism , Neovascularization, Pathologic/metabolism , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Solubility , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism
8.
Toxicology ; 304: 83-91, 2013 Feb 08.
Article in English | MEDLINE | ID: mdl-23261757

ABSTRACT

Drug nephrotoxicity is a serious health and economic problem worldwide. Rats can be acutely sensitized to acute kidney injury (AKI) by subnephrotoxic treatments with potentially nephrotoxic drugs. Acquired sensitization to AKI poses a silent risk impossible to diagnose pre-emptively with the technology available at the clinical level. Herein, we hypothesized whether a chronic, subnephrotoxic insult to the kidneys might result in chronically acquired sensitization to AKI, and whether chronic sensitization might be detected through specific urinary markers. To this end, rats were treated with a subtoxic dosage of the experimental nephrotoxin uranyl nitrate (UN) in the drinking water for 21 weeks, or plain water (as control), and then with low-dose gentamicin for 7 days. Renal function and renal tissue damage were evaluated through the experiment. The mild renal damage caused by gentamicin was markedly magnified in rats having received UN chronically, which was evident both at the functional and histological level. Four proteins, namely albumin, hemopexin, transferrin and vitamin D binding protein were increased in the urine in temporal association with the appearance of chronic predisposition. Although further studies are necessary, our results suggest that these proteins might be potentially used as markers of hidden, chronic predisposition to gentamicin nephrotoxicity, in order to appropriately and pre-emptively stratify and handle individuals according to their specific risk in the long term, and to conveniently optimize their life conditions or additional clinical procedures or treatments that might trigger the disease. This might reduce AKI incidence and severity and the associated costs.


Subject(s)
Acute Kidney Injury/chemically induced , Anti-Bacterial Agents/toxicity , Gentamicins/toxicity , Uranyl Nitrate/toxicity , Acute Kidney Injury/physiopathology , Albuminuria/chemically induced , Animals , Anti-Bacterial Agents/administration & dosage , Biomarkers/urine , Disease Susceptibility , Gentamicins/administration & dosage , Hemopexin/urine , Male , Rats , Rats, Sprague-Dawley , Severity of Illness Index , Transferrin/urine , Uranyl Nitrate/administration & dosage , Vitamin D-Binding Protein/urine
9.
ScientificWorldJournal ; 10: 2367-84, 2010 Dec 14.
Article in English | MEDLINE | ID: mdl-21170488

ABSTRACT

Endoglin (CD105) is an auxiliary membrane receptor of transforming growth factor beta (TGF-ß) that interacts with type I and type II TGF-ß receptors and modulates TGF-ß signaling. Endoglin is overexpressed in the tumor-associated vascular endothelium, where it modulates angiogenesis. This feature makes endoglin a promising target for antiangiogenic cancer therapy. In addition, recent studies on human and experimental models of carcinogenesis point to an important tumor cell-autonomous role of endoglin by regulating proliferation, migration, invasion, and metastasis. These studies suggest that endoglin behaves as a suppressor of malignancy in experimental and human epithelial carcinogenesis, although it can also promote metastasis in other types of cancer. In this review, we evaluate the implication of endoglin in tumor development underlying studies developed in our laboratories in recent years.


Subject(s)
Antigens, CD/metabolism , Neoplasms/metabolism , Receptors, Cell Surface/metabolism , Receptors, Transforming Growth Factor beta/metabolism , Animals , Antigens, CD/physiology , Cell Movement , Cell Proliferation , Endoglin , Humans , Neoplasm Invasiveness , Neoplasm Metastasis , Neoplasms/pathology , Protein Binding , Receptors, Cell Surface/physiology , Signal Transduction
10.
Hypertension ; 56(3): 484-9, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20679183

ABSTRACT

The aim of the present experiments was to evaluate the differences in arterial pressure between H-Ras lacking mice and control mice and to analyze the mechanisms involved in the genesis of the differences. H-Ras lacking mice and mouse embryonic fibroblasts from these animals were used. Blood pressure was measured using 3 different methods: direct intraarterial measurement in anesthetized animals, tail-cuff sphygmomanometer, and radiotelemetry. H-Ras lacking mice showed lower blood pressure than control animals. Moreover, the aorta protein content of endothelial nitric oxide synthase, soluble guanylyl cyclase, and cyclic guanosine monophosphate-dependent protein kinase was higher in H-Ras knockout mice than in control animals. The activity of these enzymes was increased, because urinary nitrite excretion, sodium nitroprusside-stimulated vascular cyclic guanosine monophosphate synthesis, and phosphorylated vasoactive-stimulated phosphoprotein in aortic tissue increased in these animals. Furthermore, mouse embryonic fibroblasts from H-Ras lacking mice showed higher cyclic guanosine monophosphate-dependent protein kinase promoter activity than control cells. These results strongly support the upregulation of the nitric oxide-cyclic guanosine monophosphate pathway in H-Ras-deficient mice. Moreover, they suggest that H-Ras pathway could be considered as a therapeutic target for hypertension treatment.


Subject(s)
Cyclic GMP-Dependent Protein Kinases/metabolism , Cyclic GMP/metabolism , Hypotension/metabolism , Nitric Oxide/metabolism , ras Proteins/metabolism , Animals , Aorta/metabolism , Blood Pressure/physiology , Blotting, Northern , Blotting, Western , Cells, Cultured , Cyclic GMP/genetics , Cyclic GMP-Dependent Protein Kinases/genetics , Fibroblasts/cytology , Fibroblasts/metabolism , Guanylate Cyclase/genetics , Guanylate Cyclase/metabolism , Hypotension/genetics , Immunohistochemistry , Mice , Mice, Knockout , Nitric Oxide/genetics , Nitric Oxide Synthase Type III/genetics , Nitric Oxide Synthase Type III/metabolism , Phosphorylation , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Cytoplasmic and Nuclear/metabolism , Signal Transduction/physiology , Soluble Guanylyl Cyclase , Statistics, Nonparametric , Up-Regulation/physiology , ras Proteins/genetics
11.
Toxicol Sci ; 118(2): 324-47, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20554698

ABSTRACT

As in the case of other heavy metals, a considerable body of evidence suggests that overexposure to uranium may cause pathological alterations to the kidneys in both humans and animals. In the present work, our aim was to analyze the available data from a critical perspective that should provide a view of the real danger of the nephrotoxicity of this metal for human beings. A further aim was to elaborate a comparative compilation of the renal pathophysiological data obtained in humans and experimental animals with a view to gaining more insight into our knowledge of the mechanisms of action and renal damage. Finally, we address the existing perspectives for the improvement of diagnostic methods and the treatment of intoxications by uranium, performing an integrated analysis of all these aspects.


Subject(s)
Environmental Pollutants/toxicity , Kidney Diseases/chemically induced , Kidney/drug effects , Uranium Compounds/toxicity , Acute Disease , Animals , Disease Models, Animal , Female , Humans , Kidney/metabolism , Kidney/physiopathology , Kidney Diseases/physiopathology , Kidney Diseases/therapy , Male , Oxidative Stress/drug effects , Toxicity Tests
12.
Menopause ; 17(4): 766-71, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20386345

ABSTRACT

OBJECTIVE: The aim of this study was to evaluate the effect of 17beta-estradiol, raloxifene, and 1-alpha,25-dihydroxycholecalciferol or calcitriol on bone and lipid metabolism in chronic kidney disease and estrogen insufficiency. METHODS: Six-month-old female Sprague-Dawley rats (n = 48) were ovariectomized and nephrectomized (seven eighths). One week after surgery, the rats were divided into six groups and treated with (1) placebo, (2) 17beta-estradiol 10 microg kg day, (3) raloxifene 1 mg kg day, (4) calcitriol 10 ng kg day, (5) 17beta-estradiol + calcitriol, and (6) raloxifene + calcitriol. A group of untreated animals with chronic kidney disease and normal ovarian function was used as a control group (n = 5). The rats were killed after 8 weeks of treatment. Blood samples were drawn for serum analyses; the right tibia was removed to perform histomorphometric analyses, uteri were used as tissue markers of estrogen replacement, and paraffin-embedded sections of the uterus and the fourth breast were used for histopathologic evaluation. RESULTS: Raloxifene, alone or combined with calcitriol, and 17beta-estradiol combined with calcitriol significantly diminished total cholesterol level compared with placebo. Qualitative histological and histomorphometric analyses showed that both the single treatments and their combinations were able to increase the trabecular connectivity compared with placebo. The less beneficial results were obtained with 17beta-estradiol alone, whereas the more beneficial results were obtained with the combined treatments, particularly with raloxifene and calcitriol. CONCLUSIONS: In summary, this experimental study demonstrates the advantages of replacing both hormonal deficiencies together. The combination of calcitriol and raloxifene, a selective estrogen receptor modulator, showed a better lipid, uterus, and bone profile.


Subject(s)
Cholesterol/blood , Estradiol/pharmacology , Estrogens/deficiency , Kidney Diseases/complications , Raloxifene Hydrochloride/pharmacology , Tibia/metabolism , Animals , Atrophy , Bone Density Conservation Agents/pharmacology , Calcitriol/pharmacology , Calcium/blood , Chronic Disease , Estrogens/pharmacology , Female , Hyperplasia , Mammary Glands, Animal/pathology , Organ Size , Parathyroid Hormone/blood , Phosphorus/blood , Rats , Rats, Sprague-Dawley , Selective Estrogen Receptor Modulators/pharmacology , Tibia/pathology , Uterus/pathology
13.
Nephrol Dial Transplant ; 25(9): 2930-7, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20233740

ABSTRACT

BACKGROUND: The aim of this study was to investigate whether nanomolar concentrations of lanthanum could influence the calcium-sensing receptor (CaSR) response. METHODS: Embryonic kidney (HEK-293) cells transiently transfected with the human CaSR were used to test the ability of lanthanum to activate the CaSR, either alone or in combination with calcium. CaSR activation was measured by flow cytometry. Parathyroid glands from 4-month-old male Wistar rats with normal renal function (n = 60) were also cultured ex vivo with different concentrations of lanthanum to measure parathyroid hormone (PTH) secreted to the medium and PTH mRNA. RESULTS: The maximal CaSR activation induced by 1 muM lanthanum chloride (LaCl(3)) was similar to that induced by 16 mM calcium chloride (CaCl(2) 16 mM: 294 +/- 14%; LaCl(3) 1 muM: 303 +/- 11%). Lanthanum half effective concentration (EC(50)) was 77.28 nM, lower than the 2.30 mM obtained for calcium, supporting the concept that this metal is a strong agonist of the CaSR. Moreover, lanthanum was also able to enhance CaSR sensitivity to calcium. The presence of 1 nM LaCl(3) significantly left-shifted the CaSR response curve, changing the EC(50) value for calcium from 2.30 mM (calcium alone) to 1.26 mM (calcium + 1 nM lanthanum). The parathyroid glands cultured with lanthanum showed a trend to secrete less PTH compared to the control glands: 1.51 +/- 0.23 (control), 0.91 +/- 0.17 (La 100 nM) and 1.04 +/- 0.18 (La 400 nM) [(pg/h)/(pg/h), mean +/- SEM] (ANOVA P = 0.0145). A similar trend was also observed in PTH synthesis measured by PTH mRNA levels. CONCLUSIONS: These in vitro findings demonstrate that lanthanum, at nanomolar concentrations, is an agonist of the CaSR able to activate it in the absence of calcium. In addition, it can also enhance CaSR sensitivity to calcium, modulating PTH synthesis and secretion.


Subject(s)
Calcium Chloride/pharmacology , Lanthanum/pharmacology , Parathyroid Glands/drug effects , Receptors, Calcium-Sensing/metabolism , Animals , Blotting, Western , Cells, Cultured , Drug Synergism , Flow Cytometry , Humans , Immunoenzyme Techniques , Kidney/cytology , Kidney/drug effects , Kidney/metabolism , Male , Mass Spectrometry , Parathyroid Glands/cytology , Parathyroid Glands/metabolism , Parathyroid Hormone/genetics , Parathyroid Hormone/metabolism , RNA, Messenger/genetics , Rats , Rats, Wistar , Reverse Transcriptase Polymerase Chain Reaction
14.
Head Neck ; 32(6): 737-43, 2010 Jun.
Article in English | MEDLINE | ID: mdl-19827102

ABSTRACT

BACKGROUND: Endoglin is expressed on endothelium and is implicated in the control of angiogenesis. This study compares the expression of endoglin with vascular endothelial growth factor (VEGF), commonly used as a marker for neoangiogenesis in cervical paragangliomas (CPG). METHODS: The CPG were surgically obtained from 5 patients and compared with nontumoral lung obtained from patients subjected to pulmonary resection. Detection with specific antibodies was used to determine the expression of the proteins VEGF and endoglin. The expressions of hypoxia-inducible factor (HIF) and vascular cell adhesion molecule-1 (VCAM-1) were used to determine the degree of hypoxia and capillarization, respectively. RESULTS: Endoglin is located at the plasma membrane of endothelial cells. The relative expression of endoglin is significantly higher in CPG respect to lung (p < .02), whereas that of VEGF is similar. CONCLUSION: Endoglin expression in CPG is significantly superior to that of VEGF and correlates with tumor vascularization.


Subject(s)
Antigens, CD/metabolism , Biomarkers, Tumor/metabolism , Head and Neck Neoplasms/metabolism , Neovascularization, Pathologic/metabolism , Paraganglioma/metabolism , Receptors, Cell Surface/metabolism , Vascular Endothelial Growth Factor A/metabolism , Adult , Aged , Blotting, Western , Disease Progression , Endoglin , Female , Head and Neck Neoplasms/blood supply , Humans , Hypoxia-Inducible Factor 1/metabolism , Immunoblotting , Immunohistochemistry , Ki-67 Antigen/metabolism , Male , Middle Aged , Paraganglioma/blood supply , Vascular Cell Adhesion Molecule-1/metabolism
15.
Cytotherapy ; 11(8): 1041-51, 2009.
Article in English | MEDLINE | ID: mdl-19929468

ABSTRACT

BACKGROUND AIMS: The aim of this study was to compare prospectively the vasculogenic capacity of two cell sources, monocytes and CD133+ cells. METHODS: Cells were obtained from healthy donors by adherence or magnetic selection. Animals studies were performed in a model of hind limb ischemia and different groups were established according to type and number of cells infused. Revascularization was measured by sequential blood flow analysis using a laser Doppler device and by assessing capillary density in the ischemic muscles. In order to locate the infused cells, immunofluorescence and immunocytochemistry techniques were performed and analyzed by light and confocal microscopy. RESULTS: During the study period there was a significant improvement in both limb perfusion and capillary density in mice treated with either human monocytes or CD133+ cells (P<0.05) compared with non-treated mice. No cells were detected as incorporated into the vessels when 1 x 10(5) cells were used but with higher doses (1 x 10(6)) a few human cells were observed integrated into the vessels in both groups of treated mice. Supernatants of both cell types showed vascular endothelial growth factor (VEGF), epidermal growth factor (EGF) and platelet-derived growth factor- AB (PDGF-AB) expression. CONCLUSIONS: Treatment with human monocytes or CD133+ cells improves blood perfusion and capillary density in a murine model and both cell types seem to stimulate vasculogenesis in a fairly similar way.


Subject(s)
Antigens, CD/metabolism , Glycoproteins/metabolism , Hindlimb/blood supply , Hindlimb/pathology , Ischemia/pathology , Monocytes/cytology , Neovascularization, Physiologic , Peptides/metabolism , AC133 Antigen , Angiogenesis Inducing Agents/metabolism , Animals , Capillaries/pathology , Cell Movement , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Humans , Immunophenotyping , Laser-Doppler Flowmetry , Mice , Microscopy, Confocal , Muscles/pathology , Perfusion , Phenotype , Regional Blood Flow , Staining and Labeling
16.
EMBO Mol Med ; 1(6-7): 303-14, 2009 Sep.
Article in English | MEDLINE | ID: mdl-20049734

ABSTRACT

Recent advances in our understanding of the molecular pathways that govern the association of inflammation with organ fibrosis and cancer point to the epithelial to mesenchymal transition (EMT) as the common link in the progression of these devastating diseases. The EMT is a crucial process in the development of different tissues in the embryo and its reactivation in the adult may be regarded as a physiological attempt to control inflammatory responses and to 'heal' damaged tissue. However, in pathological contexts such as in tumours or during the development of organ fibrosis, this healing response adopts a sinister nature, steering these diseases towards metastasis and organ failure. Importantly, the chronic inflammatory microenvironment common to fibrotic and cancer cells emerges as a decisive factor in the induction of the pathological EMT.


Subject(s)
Fibrosis/etiology , Fibrosis/pathology , Inflammation/complications , Neoplasms/etiology , Neoplasms/pathology , Animals , Disease Progression , Fibrosis/immunology , Humans , Inflammation/immunology , Inflammation/pathology , Neoplasms/immunology , Neoplastic Processes , Transforming Growth Factor beta1/immunology
17.
Pancreas ; 32(4): 335-42, 2006 May.
Article in English | MEDLINE | ID: mdl-16670614

ABSTRACT

OBJECTIVES: To investigate the effect of treatment with several vasodilatory substance on the changes in mean arterial pressure (MAP) of severe acute pancreatitis. METHODS: Pancreatitis was induced in rats by 5% sodium taurocholate retrograde infusion through the pancreatic duct, which produces a significant decrease in arterial blood pressure. RESULTS: Three hours after the induction of pancreatitis, a fall of approximately 25 mm Hg in MAP was observed, with no changes of MAP in untreated controls. The administration of the nitric oxide synthesis inhibitor, N-nitro-L-arginine methyl ester (25 mg/kg), previously to the induction of pancreatitis, produced a marked fall in MAP leading to the death of all the animals. When several vasodilatory substances, S-nitroso-N-acetylpenicillamine (200 microg x kg x h), calcitonin gene-related peptide (10 microg/kg), and vasoactive intestinal polypeptide (8 microg x kg x h) were administered previously to the induction of pancreatitis, the MAP fall induced by pancreatitis was not observed. The improvement of physiological conditions observed in vasodilator-treated animals is in agreement with histological data, which show only minor structural changes in the pancreas from these animals, in contrast with the severe alterations observed in untreated pancreatitic rats. CONCLUSION: : Vasodilation confers protection against the systemic circulatory derangement derived from the development of severe acute pancreatitis.


Subject(s)
Pancreatitis/drug therapy , Shock/prevention & control , Vasodilator Agents/therapeutic use , Acute Disease , Amylases/blood , Animals , Blood Pressure/drug effects , Calcitonin Gene-Related Peptide/therapeutic use , Male , NG-Nitroarginine Methyl Ester/pharmacology , Nitric Oxide/physiology , Pancreatitis/complications , Pancreatitis/pathology , Pancreatitis/physiopathology , Penicillamine/analogs & derivatives , Penicillamine/therapeutic use , Rats , Rats, Wistar , Vasoactive Intestinal Peptide/therapeutic use , Vasodilator Agents/pharmacology
18.
Hepatol Res ; 32(1): 58-65, 2005 May.
Article in English | MEDLINE | ID: mdl-15905118

ABSTRACT

An increased expression and activity of the heme oxygenase-1 (HO-1) in the liver has been observed in models of hepatic damage. Nitric oxide (NO) seems to be involved in HO-1 regulation. The aim of this work is to assess HO-1 induction and heme oxygenase (HO) activity in rats with bile duct ligation (BDL). We have assessed the effect of chronic inhibition of the NO synthesis by N(G)-nitro-l-arginine methyl ester (l-NAME) on HO-1 induction and HO activity. In the BDL animals, compared with sham-operated ones, we found an increased plasma nitrite and bilirubin concentration, and a marked liver expression of inducible nitric oxide synthase and HO-1, assessed by both Western blot and immunohistochemistry. Chronic l-NAME treatment prevented plasma nitrite increase in animals subjected to BDL. BDL animals treated with l-NAME, compared with untreated BDL rats, showed an important decrease in HO-1 expression and in HO activity (assessed as a decreased plasma bilirubin and bilirubin excretion). In conclusion, our experiments show parallel changes in expression and activity of HO-1 and NOS2 activity in the BDL model of liver damage and suggest that increased NO production is involved in HO-1 overexpression.

19.
Oncogene ; 24(27): 4450-61, 2005 Jun 23.
Article in English | MEDLINE | ID: mdl-15806144

ABSTRACT

Endoglin is a transmembrane glycoprotein that acts as an auxiliary receptor for transforming growth factor-beta (TGF-beta) and modulates cellular responses to this pleiotropic cytokine. Endoglin is strongly expressed in endothelial cells, where it appears to exert a crucial role in vascular development and angiogenesis. Two endoglin isoforms (L and S), differing in their cytoplasmic domains, have been previously characterized in human tissues. We now demonstrate the existence of similar L- and S-endoglin variants in murine tissues with 47 and 35 amino acids, respectively, in their cytoplasmic tail. RT-PCR analysis showed that L is the predominant endoglin isoform expressed in mouse tissues, although S-endoglin mRNA is significantly expressed in liver and lung, as well as in endothelial cell lines. Furthermore, a protein of size equivalent to recombinant S-endoglin expressed in mammalian cells was detected in mouse endothelial cells by Western blot analysis. L- and S-endoglin isoforms can form disulfide-linked heterodimers, as demonstrated by cotransfection of L- and S-endoglin constructs. To address the role of S-endoglin in vivo, an S-Eng(+) transgenic mouse model that targets S-endoglin expression to the endothelium was generated. The lethal phenotype of endoglin-null (Eng(-/-)) mice was not rescued by breeding S-Eng(+) transgenic mice into the endoglin-null background. S-Eng(+) mice exhibited reduced tumor growth and neovascularization after transplantation of Lewis lung carcinoma cells. In addition, S-Eng(+) mice showed a drastic inhibition of benign papilloma formation when subjected to two-stage chemical skin carcinogenesis. These results point to S-endoglin as an antiangiogenic molecule, in contrast to L-endoglin which is proangiogenic. Oncogene (2005) 24, 4450-4461. doi:10.1038/sj.onc.1208644 Published online 4 April 2005.


Subject(s)
Intracellular Signaling Peptides and Proteins/metabolism , Neoplasms/metabolism , Neoplasms/pathology , Amino Acid Sequence , Animals , Base Sequence , Cell Line , DNA, Complementary/genetics , Endoglin , Female , Homozygote , Humans , Intracellular Signaling Peptides and Proteins/chemistry , Intracellular Signaling Peptides and Proteins/genetics , Male , Mice , Mice, Knockout , Molecular Sequence Data , Neoplasms/genetics , Phenotype , Protein Isoforms/chemistry , Protein Isoforms/genetics , Protein Isoforms/metabolism , RNA, Messenger/genetics , Sequence Alignment
20.
Proc Natl Acad Sci U S A ; 102(10): 3685-90, 2005 Mar 08.
Article in English | MEDLINE | ID: mdl-15728377

ABSTRACT

To explore the mechanisms by which NO elicits endothelial cell (EC) migration we used murine and bovine aortic ECs in an in vitro wound-healing model. We found that exogenous or endogenous NO stimulated EC migration. Moreover, migration was significantly delayed in ECs derived from endothelial NO synthase-deficient mice compared with WT murine aortic EC. To assess the contribution of matrix metalloproteinase (MMP)-13 to NO-mediated EC migration, we used RNA interference to silence MMP-13 expression in ECs. Migration was delayed in cells in which MMP-13 was silenced. In untreated cells MMP-13 was localized to caveolae, forming a complex with caveolin-1. Stimulation with NO disrupted this complex and significantly increased extracellular MMP-13 abundance, leading to collagen breakdown. Our findings show that MMP-13 is an important effector of NO-activated endothelial migration.


Subject(s)
Cell Movement , Collagenases/physiology , Endothelial Cells/cytology , Nitric Oxide/physiology , Wound Healing , Animals , Cattle , Caveolin 1 , Caveolins/physiology , Matrix Metalloproteinase 13 , Mice , Mice, Inbred C57BL , Nitric Oxide Synthase/physiology , Nitric Oxide Synthase Type II , Nitric Oxide Synthase Type III
SELECTION OF CITATIONS
SEARCH DETAIL
...