Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
1.
Endocrinology ; 162(8)2021 08 01.
Article in English | MEDLINE | ID: mdl-33955458

ABSTRACT

Pulmonary fibrosis is an irreversible, potentially fatal disease. Adrenomedullin (AM) is a multifunctional peptide whose activity is regulated by receptor activity-modifying protein 2 (RAMP2). In the present study, we used the bleomycin (BLM)-induced mouse pulmonary fibrosis model to investigate the pathophysiological significance of the AM-RAMP2 system in the lung. In heterozygous AM knockout mice (AM+/-), hydroxyproline content and Ashcroft scores reflecting the fibrosis severity were significantly higher than in wild-type mice (WT). During the acute phase after BLM administration, FACS analysis showed significant increases in eosinophil, monocyte, and neutrophil infiltration into the lungs of AM+/-. During the chronic phase, fibrosis-related molecules were upregulated in AM+/-. Notably, nearly identical changes were observed in RAMP2+/-. AM administration reduced fibrosis severity. In the lungs of BLM-administered AM+/-, the activation level of Smad3, a receptor-activated Smad, was higher than in WT. In addition, Smad7, an antagonistic Smad, was downregulated and microRNA-21, which targets Smad7, was upregulated compared to WT. Isolated AM+/- lung fibroblasts showed less proliferation and migration capacity than WT fibroblasts. Stimulation with TGF-ß increased the numbers of α-SMA-positive myofibroblasts, which were more prominent among AM+/- cells. TGF-ß-stimulated AM+/- myofibroblasts were larger and exhibited greater contractility and extracellular matrix production than WT cells. These cells were α-SMA (+), F-actin (+), and Ki-67(-) and appeared to be nonproliferating myofibroblasts (non-p-MyoFbs), which contribute to the severity of fibrosis. Our findings suggest that in addition to suppressing inflammation, the AM-RAMP2 system ameliorates pulmonary fibrosis by suppressing TGF-ß-Smad3 signaling, microRNA-21 activity and differentiation into non-p-MyoFbs.


Subject(s)
Adrenomedullin/therapeutic use , Myofibroblasts/drug effects , Pulmonary Fibrosis/drug therapy , Receptor Activity-Modifying Protein 2/metabolism , Transforming Growth Factor beta/metabolism , Adrenomedullin/metabolism , Adrenomedullin/pharmacology , Animals , Bleomycin , Cell Differentiation/drug effects , Disease Models, Animal , Drug Evaluation, Preclinical , Infusions, Intravenous , Male , Mice, Inbred C57BL , Mice, Knockout , MicroRNAs/metabolism , Myofibroblasts/metabolism , Pulmonary Fibrosis/etiology , Pulmonary Fibrosis/metabolism , Pulmonary Fibrosis/prevention & control , Smad7 Protein/metabolism , Transforming Growth Factor beta/pharmacology
2.
Nefrologia ; 34(6): 724-31, 2014 Nov 17.
Article in English, Spanish | MEDLINE | ID: mdl-25335086

ABSTRACT

BACKGROUND AND AIMS: Contrast-induced nephropathy (CIN) has a growing incidence in which renal vasoconstriction and medullary hypoxia are important mechanisms. Therapeutic approaches are very restricted and there is a considerable interest in advancing preventive strategies. Adrenomedullin is a relatively novel peptide having antioxidant, vasoactive and vasodilatory properties. We aimed to investigate whether adrenomedullin might have a preventive role against the development of experimental CIN. METHODS: Wistar albino rats (n=24) were allocated randomly into four equal groups of 6 each; Control (C), Adrenomedullin (A), Contrast Media (CM) and Adrenomedullin plus Contrast Media (ACM). All rats were deprived of water from day 1 to day 4 during 72 hours. Then, intravenous administrations of chemicals were performed. Adrenomedullin was given at dose of 12µg/kg to groups A and ACM. A single dose of high-osmolar contrast media; diatrizoate (Urografin 76%, Schering AG, Germany) was injected to groups CM and ACM at dose of 10mL/kg. On day 1 and 6 blood samples were drawn for renal function tests and inflammatory markers including TNF-α IL-1β, IL-6 and IL-18. After sacrification, kidney histologies were examined with hematoxylin-eosin staining. RESULTS: Compared to CM group, serum cystatin-C levels on 6th day were found significantly lower in ACM group (p<0.05). Additionally, daily protein excretion rates, absolute changes in daily urine output and creatinine clearance values were significantly lower in ACM group than those in CM group (p<0.05). In histopathological evaluation, regarding the degree of tubular damage and medullary congestion scores, ACM group had slightly better scores compared to CM group; however the differences did not reach significance as shown in inflammatory markers. CONCLUSION: This study demonstrated a beneficial impact of adrenomedullin on deteriorated renal function tests in an experimental CIN model. Adrenomedullin might be a candidate agent for prophylaxis of CIN. However, further studies are needed to shed more light on this issue.


Subject(s)
Acute Kidney Injury/prevention & control , Adrenomedullin/therapeutic use , Anti-Inflammatory Agents/therapeutic use , Contrast Media/toxicity , Diatrizoate/toxicity , Vasodilator Agents/therapeutic use , Acute Kidney Injury/blood , Acute Kidney Injury/chemically induced , Acute Kidney Injury/pathology , Animals , Cytokines/blood , Drug Evaluation, Preclinical , Female , Inflammation Mediators/blood , Kidney/pathology , Random Allocation , Rats , Rats, Wistar , Water Deprivation
3.
Arthritis Rheum ; 64(4): 1069-81, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22006509

ABSTRACT

OBJECTIVE: Adrenomedullin(22-52) is a truncated peptide derived from adrenomedullin, a growth factor with antiapoptotic and immunoregulatory properties. It can act as an agonist or an antagonist depending on cell type. Its in vivo effects are unknown, but adrenomedullin(22-52) could possess immunomodulatory properties. This study was undertaken to evaluate the effect of adrenomedullin(22-52) in a mouse model of arthritis. METHODS: DBA/1 mice with collagen-induced arthritis (CIA) were treated with 1.2 µg/gm adrenomedullin(22-52) , adrenomedullin, or saline at arthritis onset. Bone mineral density was measured at the beginning of the experiment and when mice were killed. Mouse joints were processed for histologic analysis and protein studies, and spleens were examined for Treg cell expression. Cytokine expression was studied in mouse joint tissue and serum. RESULTS: In mice with CIA, adrenomedullin and adrenomedullin(22-52) reduced clinical and histologic arthritis scores and shifted the pattern of articular and systemic cytokine expression from Th1 to Th2, as compared to untreated mice with CIA (controls). Tumor necrosis factor α, interleukin-6 (IL-6), and IL-17A levels were significantly decreased in the joints of mice with CIA treated with adrenomedullin or adrenomedullin(22-52) as compared to controls, whereas IL-4 and IL-10 levels were increased. Adrenomedullin(22-52) was more effective than adrenomedullin in modulating cytokine content and enhanced Treg cell function without changing Treg cell expression compared to controls. Adrenomedullin receptor binding and transcriptional adrenomedullin receptor expression were markedly increased in joints from controls, whereas adrenomedullin receptor binding was considerably decreased in treated animals. Mice with CIA treated with adrenomedullin or adrenomedullin(22-52) had considerably fewer apoptotic chondrocytes and diminished cartilage degradation. Adrenomedullin(22-52) completely prevented systemic bone loss by preserving osteoblastic activity, but without changes in osteoclastic activity. CONCLUSION: Our findings indicate that adrenomedullin(22-52) , which has no vasoactive or tumor-inducing effects, is a potent antiinflammatory and bone-protective agent in this arthritis model.


Subject(s)
Adrenomedullin/therapeutic use , Arthritis, Experimental/drug therapy , Bone Resorption/drug therapy , Inflammation/drug therapy , Joints/drug effects , Peptide Fragments/therapeutic use , Adrenomedullin/administration & dosage , Animals , Apoptosis/drug effects , Arthritis, Experimental/metabolism , Arthritis, Experimental/pathology , Bone Resorption/metabolism , Bone Resorption/pathology , Chondrocytes/drug effects , Chondrocytes/metabolism , Chondrocytes/pathology , Cytokines/metabolism , Inflammation/metabolism , Inflammation/pathology , Joints/metabolism , Joints/pathology , Male , Mice , Peptide Fragments/administration & dosage , Receptors, Adrenomedullin/metabolism , Severity of Illness Index
4.
Thorax ; 65(12): 1077-84, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20971983

ABSTRACT

BACKGROUND: Mechanical ventilation (MV) is a life-saving intervention in acute respiratory failure without any alternative. However, even protective ventilation strategies applying minimal mechanical stress may evoke ventilator-induced lung injury (VILI). Adjuvant pharmacological strategies in addition to lung-protective ventilation to attenuate VILI are lacking. Adrenomedullin exhibited endothelial barrier-stabilising properties in vitro and in vivo. METHODS: In untreated mice (female C57/Bl6 mice, 11-15 weeks old) and animals treated with adrenomedullin, lung permeability, local and systemic inflammation and markers of distal organ function were assessed following 2 or 6 h of mechanical ventilation with 100% oxygen and protective or moderately injurious ventilator settings, respectively. RESULTS: Adrenomedullin dramatically reduced lung permeability in VILI in mice, leading to improved oxygenation. Adrenomedullin treatment reduced myosin light chain phosphorylation, attenuated the accumulation of leucocytes in the lung and prevented the increase in lactate and creatinine levels in mice ventilated with high tidal volumes. Moreover, adrenomedullin protected against VILI even when treatment was initiated 2 h after the beginning of mechanical ventilation in a 6 h VILI mouse model. CONCLUSION: Adjuvant treatment with adrenomedullin may be a promising new pharmacological approach to attenuate VILI.


Subject(s)
Adrenomedullin/therapeutic use , Bronchodilator Agents/therapeutic use , Ventilator-Induced Lung Injury/prevention & control , Animals , Capillary Permeability/drug effects , Cytokines/biosynthesis , Drug Evaluation, Preclinical/methods , Female , Kidney Diseases/prevention & control , Lactic Acid/blood , Leukocyte Count , Lung/blood supply , Mice , Mice, Inbred C57BL , Oxygen Consumption/drug effects , Phosphorylation/drug effects , Pulmonary Alveoli/pathology , Respiration, Artificial/adverse effects , Respiration, Artificial/methods , Ventilator-Induced Lung Injury/physiopathology
5.
J Surg Res ; 163(1): 110-7, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20538296

ABSTRACT

BACKGROUND: Acute renal failure secondary to ischemia and reperfusion (I/R) injury poses a significant burden on both surgeons and patients. It carries a high morbidity and mortality rate and no specific treatment currently exists. Major causes of renal I/R injury include trauma, sepsis, hypoperfusion, and various surgical procedures. We have demonstrated that adrenomedullin (AM), a novel vasoactive peptide, combined with AM binding protein-1 (AMBP-1), which augments the activity of AM, is beneficial in various disease conditions. However, it remains unknown whether human AM/AMBP-1 provides any beneficial effects in renal I/R injury. The objective of our study therefore was to determine whether administration of human AM/AMBP-1 can prevent and/or minimize damage in a rat model of renal I/R injury. METHODS: Male adult rats were subjected to renal I/R injury by bilateral renal pedicle clamping with microvascular clips for 60 min followed by reperfusion. Human AM (12 microg/kg BW) and human AMBP-1 (40 microg/kg BW) or vehicle (52 microg/kg BW human albumin) were given intravenously over 30 min immediately following the clip removal (i.e., reperfusion). Rats were allowed to recover for 24 h post-treatment, and blood and renal tissue samples were collected. Plasma levels of AM were measured using a radioimmunoassay specific for rat AM. Plasma AMBP-1 was measured by Western analysis. Renal water content and serum levels of systemic markers of tissue injury were measured. Serum and renal TNF-alpha levels were also assessed. RESULTS: At 24 h after renal I/R injury, plasma levels of AM were significantly increased while plasma AMBP-1 was markedly decreased. Renal water content and systemic markers of tissue injury (e.g., creatinine, BUN, AST, and ALT) were significantly increased following renal I/R injury. Serum and renal TNF-alpha levels were also increased post injury. Administration of human AM/AMBP-1 decreased renal water content, and plasma levels of creatinine, BUN, AST, and ALT. Serum and renal TNF-alpha levels were also significantly decreased after AM/AMBP-1 treatment. CONCLUSION: Treatment with human AM/AMBP-1 in renal I/R injury significantly attenuated organ injury and the inflammatory response. Thus, human AM combined with human AMBP-1 may be developed as a novel treatment for patients with acute renal I/R injury.


Subject(s)
Adrenomedullin/therapeutic use , Complement Factor H/therapeutic use , Kidney Diseases/prevention & control , Reperfusion Injury/prevention & control , Vasodilator Agents/therapeutic use , Adrenomedullin/blood , Alpha-Globulins , Animals , Humans , Kidney/metabolism , Kidney Diseases/blood , Male , Proteins/metabolism , Rats , Reperfusion Injury/blood , Tumor Necrosis Factor-alpha/metabolism , Water/metabolism
6.
Endocrinology ; 150(3): 1450-5, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19008310

ABSTRACT

We recently reported that long-term treatment with pioglitazone restored cardiac Akt phosphorylation in response to hyperthermia (HT) and subsequent cardiac heat-shock protein 72 (HSP72) expression, in heredity insulin resistance rats via improvement of insulin sensitivity. Because adrenomedullin (AM) promotes Akt phosphorylation and attenuates myocardial ischemia/reperfusion injury, we tested the hypothesis that pretreatment with AM before HT could restore depressed Akt activation and cardiac HSP72 expression, thereby enhancing protection against ischemia/reperfusion injury in this model. At 16 wk of age, male insulin-resistant Otsuka Long-Evans Tokushima Fatty (OLETF) rats and control Long-Evans Tokushima Otsuka (LETO) rats were treated with AM (0.05 microg/kg . /min iv) or vehicle for 60 min. Thereafter, HT (43 C for 20 min) or normothermia (NT; 37 C for 20 min) was applied. The heart was isolated 1 and 24 h after HT. 1) Either AM or HT induced myocardial Akt phosphorylation in a phosphatidylinositol 3-kinase-dependent manner, which was augmented by their combination. 2) Akt phosphorylation induced by HT, or a combination of HT and AM, was attenuated in insulin-resistant OLETF rat hearts. 3) The levels of Akt phosphorylation in response to AM and/or HT correlated with reperfusion-induced left ventricular functional recovery and amount of released creatine kinase during reperfusion. 4) AM protected the hearts of OLETF rats and LETO rats. Our results suggest that AM pretreatment could enhance HT-induced myocardial Akt phosphorylation and subsequent HSP72 expression in a phosphatidylinositol 3-kinase-dependent manner, in association with tolerance against ischemia/reperfusion injury. This intervention was effective even in insulin-resistant hearts.


Subject(s)
Adrenomedullin/therapeutic use , HSP72 Heat-Shock Proteins/metabolism , Insulin Resistance , Myocardium/metabolism , Reperfusion Injury/prevention & control , Adrenomedullin/pharmacology , Animals , Blood Pressure/drug effects , Cardiotonic Agents/pharmacology , Cardiotonic Agents/therapeutic use , Drug Evaluation, Preclinical , Heart/drug effects , Insulin Resistance/physiology , Male , Oncogene Protein v-akt/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation/drug effects , Rats , Rats, Inbred OLETF , Rats, Long-Evans , Recovery of Function/drug effects , Reperfusion Injury/rehabilitation , Ventricular Function, Left/drug effects
7.
Br J Anaesth ; 99(6): 830-6, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17962242

ABSTRACT

BACKGROUND: Hypodynamic septic shock is associated with a poor prognosis. The present randomized-controlled laboratory experiment was designed to test the hypothesis that the vasodilatory peptide hormone adrenomedullin (ADM) is a useful agent to prevent and reverse the development of hypodynamic circulation in ovine endotoxaemia. METHODS: Twenty-four healthy ewes were chronically instrumented for haemodynamic monitoring and randomly allocated to either the control, treatment, or prophylaxis group (n = 8 each). After a baseline (BL) measurement in the healthy state, all sheep were subjected to a continuous endotoxin infusion started at 10 ng kg(-1) min(-1) and doubled every hour six times. After 4 h of endotoxin challenge, the treatment group received ADM (50 ng kg(-1) min(-1)) for the remaining 3 h of the experiment. The prophylaxis group received a simultaneous infusion of endotoxin and ADM (50 ng kg(-1) min(-1)) from the beginning to the end of the 7 h intervention period. RESULTS: In the control and treatment groups, the ewes exhibited a hypodynamic circulation at 4 h (>20% reduction in cardiac index, both P < 0.01 vs BL). Endotoxin also increased mean pulmonary arterial pressure (MPAP) and arterial lactate concentrations. Prophylactic infusion of ADM prevented the occurrence of pulmonary hypertension and hypodynamic circulation and thereby blunted the increase in arterial lactate concentrations. In the treatment group, ADM administration increased CI (P < 0.001) and reduced both MPAP (P = 0.023) and arterial lactate concentrations (P < 0.001 each at 7 h) when compared with the control group. CONCLUSIONS: This study demonstrates that exogenous ADM prevents and reverses hypodynamic circulation, attenuates pulmonary hypertension, and limits lactic acidosis in ovine endotoxaemia.


Subject(s)
Adrenomedullin/therapeutic use , Endotoxemia/complications , Hypertension, Pulmonary/prevention & control , Vasodilator Agents/therapeutic use , Acidosis, Lactic/etiology , Acidosis, Lactic/prevention & control , Animals , Cardiac Output/drug effects , Drug Evaluation, Preclinical , Female , Hypertension, Pulmonary/etiology , Lactic Acid/blood , Sheep, Domestic , Shock, Septic/complications
8.
Mol Med ; 13(9-10): 488-94, 2007.
Article in English | MEDLINE | ID: mdl-17932560

ABSTRACT

Downregulation of vascular endothelial constitutive nitric oxide synthase (ecNOS) contributes to the vascular hyporesponsiveness in sepsis. Although coadministration of the potent vasodilatory peptide adrenomedulin (AM) and the newly discovered AM binding protein (AMBP-1) maintains cardiovascular stability and reduces mortality in sepsis, it remains unknown whether AM/AMBP-1 prevents endothelial cell dysfunction. To investigate this possibility, we subjected adult male rats to sepsis by cecal ligation and puncture (CLP), with or without subsequent intravenous administration of the combination of AM (12 microg/kg) and AMBP-1 (40 microg/kg). Thoracic aortae were harvested 20 h after CLP (i.e., the late stage of sepsis) and endothelium-dependent vascular relaxation was determined by the addition of acetylcholine (ACh) in an organ bath system. In addition, ecNOS gene and protein expression was assessed by RT-PCR and immunohistochemistry, respectively. The results indicate that ACh-induced (i.e., endothelium-dependent) vascular relaxation was significantly reduced 20 h after CLP. Administration of AM/AMBP-1 prevented the reduction of vascular relaxation. In addition, ecNOS gene expression in aortic and pulmonary tissues was downregulated 20 h after CLP and AM/AMBP-1 attenuated such a reduction. Moreover, the decreased ecNOS staining in thoracic aortae of septic animals was prevented by the treatment with AM/AMBP-1. These results, taken together, indicate that AM/AMBP-1 preserves ecNOS and prevents reduced endothelium-dependent vascular relaxation (i.e., endothelial cell dysfunction) in sepsis. In light of our recent finding that AM/AMBP-1 improves organ function and reduces mortality in sepsis, it is most likely that the protective effect of these compounds on ecNOS is a mechanism responsible for the salutary effect of AM/AMBP-1 in sepsis.


Subject(s)
Adrenomedullin/therapeutic use , Endothelium, Vascular/drug effects , Proteins/therapeutic use , Sepsis/drug therapy , Vasodilation/drug effects , Acetylcholine/pharmacology , Adrenomedullin/administration & dosage , Animals , Aorta, Thoracic/enzymology , Aorta, Thoracic/metabolism , Dose-Response Relationship, Drug , Immunohistochemistry , Infusions, Intravenous , Intracellular Signaling Peptides and Proteins , Male , Nitric Oxide Synthase Type III/analysis , Nitric Oxide Synthase Type III/metabolism , Proteins/administration & dosage , Rats , Rats, Sprague-Dawley , Sepsis/etiology , Vasodilator Agents/pharmacology
9.
Exp Toxicol Pathol ; 58(5): 367-74, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17267195

ABSTRACT

Recent studies have demonstrated that chronic cadmium administration induces oxidative stress. In the present study, we investigated the possible therapeutic effect of adrenomedullin, a potent antioxidant, in cadmium-induced morphological, ultrastructural and biochemical alterations. Two groups of rats were exposed to 100 ppm of CdCl(2) in drinking water for four weeks. One of these groups received 3000 ng/kg body weight of adrenomedullin (AdM) intraperitoneally during the last week. Hepatic oxidative stress markers were evaluated by changes in the amount of lipid peroxides and changes in the antioxidant enzyme activities, superoxide dismutase (SOD) and glutathione peroxidase (GPx) and glutathione reductase (GSH) levels. Hepatic damage score was significantly higher in Cd-administered rats than those of controls (p<0.005). Cd-induced ultrastructural changes in hepatocytes included focal parenchymal cell necrosis, dilatation of rough endoplasmic reticulum, proliferation of lysosomes and mitochondrial degeneration. Hepatic damage was accompanied by significant increase in tissue MDA level (p<0.05) and significant decrease in tissue GSH level (p<0.05), and SOD and GPx activities (p>0.05, p>0.005, respectively). Adrenomedullin failed to restore the light and electron microscopic, and biochemical changes. We conclude that although we administered a high dose of adrenomedullin, it failed to reduce cadmium-induced hepatic damage probably because of the irreversibility of Cd-induced hepatic injury.


Subject(s)
Adrenomedullin/therapeutic use , Antioxidants/therapeutic use , Cadmium Chloride/toxicity , Chemical and Drug Induced Liver Injury , Liver , Oxidative Stress/drug effects , Adrenomedullin/pharmacology , Animals , Antioxidants/pharmacology , Chemical and Drug Induced Liver Injury/metabolism , Chemical and Drug Induced Liver Injury/pathology , Chemical and Drug Induced Liver Injury/prevention & control , Glutathione Reductase/metabolism , Lipid Peroxides/metabolism , Liver/drug effects , Liver/enzymology , Liver/metabolism , Liver/ultrastructure , Male , Microscopy, Electron , Rats , Rats, Wistar , Superoxide Dismutase/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL