Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
Eur Heart J ; 45(15): 1355-1367, 2024 Apr 14.
Article in English | MEDLINE | ID: mdl-38385506

ABSTRACT

BACKGROUND AND AIMS: Thromboxane (TX) A2, released by activated platelets, plays an important role in atherothrombosis. Urinary 11-dehydro-TXB2 (U-TXM), a stable metabolite reflecting the whole-body TXA2 biosynthesis, is reduced by ∼70% by daily low-dose aspirin. The U-TXM represents a non-invasive biomarker of in vivo platelet activation and is enhanced in patients with diabetes. This study assessed whether U-TXM is associated with the risk of future serious vascular events or revascularizations (SVE-R), major bleeding, or cancer in patients with diabetes. METHODS: The U-TXM was measured pre-randomization to aspirin or placebo in 5948 people with type 1 or 2 diabetes and no cardiovascular disease, in the ASCEND trial. Associations between log U-TXM and SVE-R (n = 618), major bleed (n = 206), and cancer (n = 700) during 6.6 years of follow-up were investigated by Cox regression; comparisons of these associations with the effects of randomization to aspirin were made. RESULTS: Higher U-TXM was associated with older age, female sex, current smoking, type 2 diabetes, higher body size, urinary albumin/creatinine ratio of ≥3 mg/mmol, and higher estimated glomerular filtration rate. After adjustment for these, U-TXM was marginally statistically significantly associated with SVE-R and major bleed but not cancer [hazard ratios per 1 SD higher log U-TXM (95% confidence interval): 1.09 (1.00-1.18), 1.16 (1.01-1.34), and 1.06 (0.98-1.14)]. The hazard ratio was similar to that implied by the clinical effects of randomization to aspirin for SVE-R but not for major bleed. CONCLUSIONS: The U-TXM was log-linearly independently associated with SVE-R in diabetes. This is consistent with the involvement of platelet TXA2 in diabetic atherothrombosis.


Subject(s)
Diabetes Mellitus, Type 2 , Neoplasms , Thrombosis , Humans , Female , Thromboxanes/metabolism , Thromboxanes/therapeutic use , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/drug therapy , Aspirin/therapeutic use , Thromboxane B2/therapeutic use , Thromboxane B2/urine , Thromboxane A2/therapeutic use , Thromboxane A2/urine , Thrombosis/drug therapy , Neoplasms/drug therapy
2.
Respir Res ; 24(1): 262, 2023 Oct 31.
Article in English | MEDLINE | ID: mdl-37907918

ABSTRACT

INTRODUCTION: The standard therapy for bronchial asthma consists of combinations of acute (short-acting ß2-sympathomimetics) and, depending on the severity of disease, additional long-term treatment (including inhaled glucocorticoids, long-acting ß2-sympathomimetics, anticholinergics, anti-IL-4R antibodies). The antidepressant amitriptyline has been identified as a relevant down-regulator of immunological TH2-phenotype in asthma, acting-at least partially-through inhibition of acid sphingomyelinase (ASM), an enzyme involved in sphingolipid metabolism. Here, we investigated the non-immunological role of amitriptyline on acute bronchoconstriction, a main feature of airway hyperresponsiveness in asthmatic disease. METHODS: After stimulation of precision cut lung slices (PCLS) from mice (wildtype and ASM-knockout), rats, guinea pigs and human lungs with mediators of bronchoconstriction (endogenous and exogenous acetylcholine, methacholine, serotonin, endothelin, histamine, thromboxane-receptor agonist U46619 and leukotriene LTD4, airway area was monitored in the absence of or with rising concentrations of amitriptyline. Airway dilatation was also investigated in rat PCLS by prior contraction induced by methacholine. As bronchodilators for maximal relaxation, we used IBMX (PDE inhibitor) and salbutamol (ß2-adrenergic agonist) and compared these effects with the impact of amitriptyline treatment. Isolated perfused lungs (IPL) of wildtype mice were treated with amitriptyline, administered via the vascular system (perfusate) or intratracheally as an inhalation. To this end, amitriptyline was nebulized via pariboy in-vivo and mice were ventilated with the flexiVent setup immediately after inhalation of amitriptyline with monitoring of lung function. RESULTS: Our results show amitriptyline to be a potential inhibitor of bronchoconstriction, induced by exogenous or endogenous (EFS) acetylcholine, serotonin and histamine, in PCLS from various species. The effects of endothelin, thromboxane and leukotrienes could not be blocked. In acute bronchoconstriction, amitriptyline seems to act ASM-independent, because ASM-deficiency (Smdp1-/-) did not change the effect of acetylcholine on airway contraction. Systemic as well as inhaled amitriptyline ameliorated the resistance of IPL after acetylcholine provocation. With the flexiVent setup, we demonstrated that the acetylcholine-induced rise in central and tissue resistance was much more marked in untreated animals than in amitriptyline-treated ones. Additionally, we provide clear evidence that amitriptyline dilatates pre-contracted airways as effectively as a combination of typical bronchodilators such as IBMX and salbutamol. CONCLUSION: Amitriptyline is a drug of high potential, which inhibits acute bronchoconstriction and induces bronchodilatation in pre-contracted airways. It could be one of the first therapeutic agents in asthmatic disease to have powerful effects on the TH2-allergic phenotype and on acute airway hyperresponsiveness with bronchoconstriction, especially when inhaled.


Subject(s)
Asthma , Bronchoconstriction , Mice , Rats , Humans , Animals , Guinea Pigs , Methacholine Chloride/pharmacology , Amitriptyline/pharmacology , Amitriptyline/therapeutic use , Histamine/pharmacology , Bronchodilator Agents/pharmacology , Bronchodilator Agents/therapeutic use , Serotonin/pharmacology , Serotonin/therapeutic use , Acetylcholine/pharmacology , Sympathomimetics/pharmacology , Sympathomimetics/therapeutic use , 1-Methyl-3-isobutylxanthine/pharmacology , 1-Methyl-3-isobutylxanthine/therapeutic use , Dilatation , Lung , Asthma/drug therapy , Albuterol , Endothelins/pharmacology , Endothelins/therapeutic use , Thromboxanes/pharmacology , Thromboxanes/therapeutic use
3.
Br J Cancer ; 129(4): 706-720, 2023 09.
Article in English | MEDLINE | ID: mdl-37420000

ABSTRACT

BACKGROUND: Pre-clinical models demonstrate that platelet activation is involved in the spread of malignancy. Ongoing clinical trials are assessing whether aspirin, which inhibits platelet activation, can prevent or delay metastases. METHODS: Urinary 11-dehydro-thromboxane B2 (U-TXM), a biomarker of in vivo platelet activation, was measured after radical cancer therapy and correlated with patient demographics, tumour type, recent treatment, and aspirin use (100 mg, 300 mg or placebo daily) using multivariable linear regression models with log-transformed values. RESULTS: In total, 716 patients (breast 260, colorectal 192, gastro-oesophageal 53, prostate 211) median age 61 years, 50% male were studied. Baseline median U-TXM were breast 782; colorectal 1060; gastro-oesophageal 1675 and prostate 826 pg/mg creatinine; higher than healthy individuals (~500 pg/mg creatinine). Higher levels were associated with raised body mass index, inflammatory markers, and in the colorectal and gastro-oesophageal participants compared to breast participants (P < 0.001) independent of other baseline characteristics. Aspirin 100 mg daily decreased U-TXM similarly across all tumour types (median reductions: 77-82%). Aspirin 300 mg daily provided no additional suppression of U-TXM compared with 100 mg. CONCLUSIONS: Persistently increased thromboxane biosynthesis was detected after radical cancer therapy, particularly in colorectal and gastro-oesophageal patients. Thromboxane biosynthesis should be explored further as a biomarker of active malignancy and may identify patients likely to benefit from aspirin.


Subject(s)
Aspirin , Colorectal Neoplasms , Female , Humans , Male , Middle Aged , Biomarkers , Colorectal Neoplasms/drug therapy , Creatinine , Thromboxanes/therapeutic use
4.
J Allergy Clin Immunol ; 152(3): 700-710.e3, 2023 09.
Article in English | MEDLINE | ID: mdl-37068712

ABSTRACT

BACKGROUND: Aspirin-exacerbated respiratory disease (AERD) is the triad of asthma, nasal polyposis, and respiratory reactions to COX-1 inhibitors. Overproduction of cysteinyl leukotrienes and underproduction of prostaglandin E2 (PGE2) are hallmarks of AERD. A mouse model predicted a key role for the thromboxane-prostanoid (TP) receptor in AERD. OBJECTIVE: Our aim was to determine whether ifetroban, a TP receptor antagonist, attenuates aspirin-induced respiratory symptoms in patients with AERD. METHODS: A total of 35 patients with AERD completed a 4-week double-blinded, placebo-controlled trial of ifetroban and underwent an oral aspirin challenge. The primary outcome was change in the provocative dose of aspirin that caused a 2-point increase in Total Nasal Symptom Score. Changes in lung function, eicosanoid levels, and platelet and mast cell activation were assessed. Cultured human nasal fibroblasts were stimulated with or without the TP agonist U46619 and assayed for prostanoid production. RESULTS: Ifetroban was well tolerated in AERD and did not change the mean 2-point increase in Total Nasal Symptom Score (P = .763). Participants taking ifetroban had greater aspirin-induced nasal symptoms and a greater decline in FEV1 value than did participants receiving placebo (-18.8% ± 3.6% with ifetroban vs -8.4% ± 2.1% with placebo [P = .017]). Four weeks of ifetroban significantly increased urinary leukotriene E4 levels and decreased nasal PGE2 levels compared with placebo. Peak aspirin-induced urinary thromboxane levels correlated with peak urinary leukotriene E4 and prostaglandin D2 metabolite levels in participants taking ifetroban. U46119 significantly potentiated the production of PGE2 by cultured nasal fibroblasts from subjects with AERD but not by cultured nasal fibroblasts from controls without polypoid sinusitis. CONCLUSION: Contrary to our hypothesis, TP receptor blockade worsened aspirin-induced reactions in AERD, possibly by exacerbating dysregulation of the eicosanoid system. TP signaling on stromal cells may be critical to maintaining PGE2 production when COX-2 function is low.


Subject(s)
Asthma, Aspirin-Induced , Sinusitis , Animals , Mice , Humans , Prostaglandins , Thromboxanes/therapeutic use , Leukotriene E4 , Receptors, Thromboxane/therapeutic use , Asthma, Aspirin-Induced/drug therapy , Asthma, Aspirin-Induced/diagnosis , Aspirin/adverse effects , Eicosanoids , Dinoprostone , Homeostasis , Sinusitis/chemically induced
5.
Clin Transl Sci ; 15(12): 2958-2970, 2022 12.
Article in English | MEDLINE | ID: mdl-36200184

ABSTRACT

Low-dose aspirin is currently recommended for patients with polycythemia vera (PV), a myeloproliferative neoplasm with increased risk of arterial and venous thromboses. Based on aspirin pharmacodynamics in essential thrombocythemia, a twice-daily regimen is recommended for patients with PV deemed at particularly high thrombotic risk. We investigated the effects of low-dose aspirin on platelet cyclooxygenase activity and in vivo platelet activation in 49 patients with PV, as assessed by serum thromboxane (TX) B2 and urinary TXA2 /TXB2 metabolite (TXM) measurements, respectively. A previously described pharmacokinetic-pharmacodynamic in silico model was used to simulate the degree of platelet TXA2 inhibition by once-daily (q.d.) and twice-daily (b.i.d.) aspirin, and to predict the effect of missing an aspirin dose during q.d. and b.i.d. regimens. Serum TXB2 averaged 8.2 (1.6-54.7) ng/ml and significantly correlated with the platelet count (γ = 0.39) and urinary TXM (γ = 0.52) in multivariable analysis. One-third of aspirin-treated patients with PV displayed less-than-maximal platelet TXB2 inhibition, and were characterized by significantly higher platelet counts and platelet-count corrected serum TXB2 than those with adequate inhibition. Eight patients with PV were sampled again after 12 ± 4 months, and had reproducible serum TXB2 and urinary TXM values. The in silico model predicted complete inhibition of platelet-derived TXB2 by b.i.d. aspirin, a prediction verified in a patient with PV with the highest TXB2 value while on aspirin q.d. and treated short-term with a b.i.d. regimen. In conclusion, one in three patients with PV on low-dose aspirin display less-than-maximal inhibition of platelet TXA2 production. Serum TXB2 measurement can be a valuable option to guide precision dosing of antiplatelet therapy in patients with PV.


Subject(s)
Polycythemia Vera , Humans , Polycythemia Vera/drug therapy , Polycythemia Vera/metabolism , Thromboxanes/metabolism , Thromboxanes/pharmacology , Thromboxanes/therapeutic use , Aspirin/pharmacology , Blood Platelets/metabolism , Thromboxane B2 , Thromboxane A2/metabolism , Thromboxane A2/pharmacology , Computer Simulation , Platelet Aggregation Inhibitors
6.
Respir Res ; 23(1): 272, 2022 Oct 01.
Article in English | MEDLINE | ID: mdl-36183104

ABSTRACT

BACKGROUND: Oxidative stress associated with severe cardiopulmonary diseases leads to impairment in the nitric oxide/soluble guanylate cyclase signaling pathway, shifting native soluble guanylate cyclase toward heme-free apo-soluble guanylate cyclase. Here we describe a new inhaled soluble guanylate cyclase activator to target apo-soluble guanylate cyclase and outline its therapeutic potential. METHODS: We aimed to generate a novel soluble guanylate cyclase activator, specifically designed for local inhaled application in the lung. We report the discovery and in vitro and in vivo characterization of the soluble guanylate cyclase activator mosliciguat (BAY 1237592). RESULTS: Mosliciguat specifically activates apo-soluble guanylate cyclase leading to improved cardiopulmonary circulation. Lung-selective effects, e.g., reduced pulmonary artery pressure without reduced systemic artery pressure, were seen after inhaled but not after intravenous administration in a thromboxane-induced pulmonary hypertension minipig model. These effects were observed over a broad dose range with a long duration of action and were further enhanced under experimental oxidative stress conditions. In a unilateral broncho-occlusion minipig model, inhaled mosliciguat decreased pulmonary arterial pressure without ventilation/perfusion mismatch. With respect to airway resistance, mosliciguat showed additional beneficial bronchodilatory effects in an acetylcholine-induced rat model. CONCLUSION: Inhaled mosliciguat may overcome treatment limitations in patients with pulmonary hypertension by improving pulmonary circulation and airway resistance without systemic exposure or ventilation/perfusion mismatch. Mosliciguat has the potential to become a new therapeutic paradigm, exhibiting a unique mode of action and route of application, and is currently under clinical development in phase Ib for pulmonary hypertension.


Subject(s)
Hypertension, Pulmonary , Acetylcholine , Animals , Guanylate Cyclase/metabolism , Guanylate Cyclase/therapeutic use , Nitric Oxide/metabolism , Rats , Soluble Guanylyl Cyclase/metabolism , Soluble Guanylyl Cyclase/therapeutic use , Swine , Swine, Miniature/metabolism , Thromboxanes/therapeutic use , Vasodilator Agents
7.
J Sex Med ; 19(12): 1733-1749, 2022 12.
Article in English | MEDLINE | ID: mdl-36195535

ABSTRACT

BACKGROUND: Stromal interaction molecule (STIM)/Orai calcium entry system appears to have a role in erectile dysfunction (ED) pathophysiology but its specific contribution to diabetic ED was not elucidated. AIM: To evaluate STIM/Orai inhibition on functional alterations associated with diabetic ED in rat and human penile tissues and on in vivo erectile responses in diabetic rats. METHODS: Rat corpus cavernosum (RCC) strips from nondiabetic (No DM) and streptozotocin-induced diabetic (DM) rats and human penile resistance arteries (HPRA) and corpus cavernosum (HCC) from ED patients undergoing penile prosthesis insertion were functionally evaluated in organ chambers and wire myographs. Erectile function in vivo in rats was assessed by intracavernosal pressure (ICP) responses to cavernous nerve electrical stimulation (CNES). Expression of STIM/Orai elements in HCC was determined by immunofluorescence and immunoblot. MAIN OUTCOME MEASURES: Functional responses in RCC, HCC and HPRA and STIM/Orai protein expression in HCC. In vivo erectile responses to CNES. RESULTS: Inhibition of Orai channels with YM-58483 (20 µM) significantly reduced adrenergic contractions in RCC but more effectively in DM. Thromboxane-induced and neurogenic contractions were reduced by STIM/Orai inhibition while defective endothelial, neurogenic and PDE5 inhibitor-induced relaxations were enhanced by YM-58483 (10 µM) in RCC from DM rats. In vivo, YM-58483 caused erections and attenuated diabetes-related impairment of erectile responses. YM-58483 potentiated the effects of PDE5 inhibition. In human tissues, STIM/Orai inhibition depressed adrenergic and thromboxane-induced contractions in ED patients more effectively in those with type 2 diabetes. Diabetes was associated with increased expression of Orai1 and Orai3 in ED patients. CLINICAL TRANSLATION: Targeting STIM/Orai to alleviate diabetes-related functional alterations of penile vascular tissue could improve erectile function and potentiate therapeutic effects of PDE5 inhibitors in diabetic ED. STRENGTHS AND LIMITATIONS: Improving effects of STIM/Orai inhibition on diabetes-related functional impairment was evidenced in vitro and in vivo in an animal model and validated in human tissues from ED patients. Functional findings were complemented with expression results. Main limitation was low numbers of human experiments due to limited human tissue availability. CONCLUSIONS: STIM/Orai inhibition alleviated alterations of functional responses in vitro and improved erectile responses in vivo in diabetic rats, potentiating the effects of PDE5 inhibition. STIM/Orai inhibition was validated as a target to modulate functional alterations of human penile vascular tissue in diabetic ED where Orai1 and Orai3 channels were upregulated. STIM/Orai inhibition could be a potential therapeutic strategy to overcome poor response to conventional ED therapy in diabetic patients. Sevilleja-Ortiz A, El Assar M, García-Gómez B, et al. STIM/Orai Inhibition as a Strategy for Alleviating Diabetic Erectile Dysfunction Through Modulation of Rat and Human Penile Tissue Contractility and in vivo Potentiation of Erectile Responses. J Sex Med 2022;19:1733-1749.


Subject(s)
Diabetes Mellitus, Experimental , Diabetes Mellitus, Type 2 , Erectile Dysfunction , Stromal Interaction Molecules , Animals , Humans , Male , Rats , Adrenergic Agents/metabolism , Adrenergic Agents/pharmacology , Adrenergic Agents/therapeutic use , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Type 2/complications , Erectile Dysfunction/drug therapy , Erectile Dysfunction/etiology , Penile Erection , Penis/blood supply , Phosphodiesterase 5 Inhibitors/therapeutic use , Stromal Interaction Molecules/metabolism , Thromboxanes/metabolism , Thromboxanes/pharmacology , Thromboxanes/therapeutic use
8.
Int J Biochem Cell Biol ; 151: 106281, 2022 10.
Article in English | MEDLINE | ID: mdl-35995387

ABSTRACT

Excessive mitochondrial fission in podocytes serves as a central hub for the pathogenesis of diabetic nephropathy (DN), and the thromboxane/prostaglandin receptor (TP receptor) plays a potential role in DN. However, regulation of the TP receptor during mitochondrial dynamics disorder in podocytes remains unknown. Here, we firstly reported novel mechanistic details of TP receptor effects on mitochondrial dynamics in podocytes under diabetic conditions. Expression of the TP receptor was significantly upregulated in podocytes under diabetic conditions both in vivo and in vitro. S18886 attenuated podocyte mitochondrial fission, glomerular injury and renal dysfunction in diabetic mice. Furthermore, inhibition of the TP receptor by both genetic and pharmacological methods dramatically reduced mitochondrial fission and attenuated podocyte injury induced by high glucose through regulating dynamin-related protein 1 (Drp1) phosphorylation and its subsequent translocation to mitochondria. In contrast, TP receptor overexpression and application of TP receptor agonist U46619 in these podocytes showed the opposite effect on mitochondrial fission and podocyte injury. Furthermore, treatment with Y27632, an inhibitor of Rho-associated kinase1 (ROCK1), significantly blunted more fragmented mitochondria and reduced podocyte injuries in podocytes with TP receptor overexpression or after U46619 treatment. Finally, pharmacological inhibition of Drp1 alleviated excessive mitochondrial fragmentation and podocyte damage in TP receptor overexpressing podocytes. Our data suggests that increased expression of the TP receptor can occur in a human cultured podocyte cell line and in podocytes derived from streptozotocin (STZ)-induced diabetic mice, which contributes to mitochondrial excessive fission and podocyte injury via ROCK1-Drp1 signaling.


Subject(s)
Diabetes Mellitus, Experimental , Diabetic Nephropathies , Mitochondrial Diseases , Podocytes , 15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid/metabolism , 15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid/pharmacology , 15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid/therapeutic use , Animals , Diabetes Mellitus, Experimental/pathology , Diabetic Nephropathies/pathology , Dynamins/metabolism , Glucose/metabolism , Glucose/pharmacology , Humans , Mice , Mitochondrial Diseases/metabolism , Mitochondrial Dynamics , Prostaglandins/metabolism , Prostaglandins/pharmacology , Prostaglandins/therapeutic use , Receptors, Prostaglandin/metabolism , Receptors, Prostaglandin/therapeutic use , Receptors, Thromboxane/metabolism , Receptors, Thromboxane/therapeutic use , Streptozocin , Thromboxanes/metabolism , Thromboxanes/pharmacology , Thromboxanes/therapeutic use , rho-Associated Kinases/metabolism
9.
Acta pediatr. esp ; 60(9): 535-544, nov. 2002. ilus
Article in Es | IBECS | ID: ibc-15007

ABSTRACT

Con el término autocoide se define un grupo de pequeñas moléculas liberadas por diversos tipos celulares y que actúan como hormonass autocrinas y paracrinas. La mayoría de estas moléculas se liberan como parte de la respuesta inflamatoria y están implicadas en la patogénesis de la sepsis y el choque séptico. Entre los autocoides se encuentran los eicosanoides, el factor de activación plaquetaria (PAF) y los mediadores peptídicos bradicinina y calidina. En numerosos modelos animales la administración de fármacos que bloquean la síntesis o los receptores de los autocoides ha mejorado las condiciones de sepsis o shock séptico; no obstante, los resultados son contradictorios cuando se habla de ensayos clínicos (AU)


Subject(s)
Animals , Dogs , Rats , Mice , Clinical Trials as Topic/methods , Platelet Activation , Sepsis/diagnosis , Sepsis/therapy , Eicosanoids/administration & dosage , Eicosanoids/therapeutic use , Eicosanoids/biosynthesis , Shock, Septic/complications , Shock, Septic/diagnosis , Shock, Septic/epidemiology , Thromboxanes/administration & dosage , Thromboxanes/therapeutic use
11.
New Horiz ; 1(4): 623-30, 1993 Nov.
Article in English | MEDLINE | ID: mdl-8087582

ABSTRACT

The adult respiratory distress syndrome is now thought to be caused by or complicated by a variety of mediators for which potential antagonists exist. Lung dysfunction could be prevented by antagonists to metabolites of membrane phospholipids. Examples of such metabolites include thromboxane, prostacyclin, leukotrienes, and platelet-activating factor. Oxidant stress can also produce cytotoxicity through membrane lipid peroxidation, as evidenced by the generation of isoprostanes. N-acetylcysteine, by repletion of the endogenous antioxidant glutathione, may represent a novel approach to the therapy of acute lung injury.


Subject(s)
Antioxidants/therapeutic use , Respiratory Distress Syndrome/therapy , Acetylcysteine/therapeutic use , Adrenal Cortex Hormones/therapeutic use , Antioxidants/pharmacology , Clinical Trials as Topic , Disease Models, Animal , Epoprostenol/therapeutic use , Humans , Inflammation , Intensive Care Units , Leukotrienes/therapeutic use , Lipid Peroxidation , Middle Aged , Platelet Activating Factor/therapeutic use , Respiratory Distress Syndrome/etiology , Respiratory Distress Syndrome/metabolism , Respiratory Distress Syndrome/mortality , Thromboxanes/therapeutic use , Treatment Failure
13.
Am J Med ; 80(1A): 34-45, 1986 Jan 17.
Article in English | MEDLINE | ID: mdl-3080878

ABSTRACT

The renal prostaglandins and thromboxanes are powerful autacoids with potential effects on renal hemodynamics, salt and water metabolism, and the immune system. The possibility of adverse effects on renal function in certain patients with renal disease due to cyclooxygenase inhibition with nonsteroidal anti-inflammatory drugs has long been appreciated. Experimental evidence indicates that renal prostaglandin and thromboxane production is increased in several models of renal disease and that similar decrements in renal function occur with cyclooxygenase inhibition and may be due to inhibition of vasodilator prostaglandins. Additionally, several investigators have shown that administration of prostaglandins may be therapeutic in some forms of renal disease, particularly immunologically mediated diseases. Dietary modification to affect prostaglandin production has also been promising in certain experimental models. In contrast to vasodilator prostaglandins, thromboxane is a potent vasoconstrictor and would be expected to have adverse effects on renal function. Despite demonstration of elevated glomerular thromboxane, studies using inhibitors of thromboxane synthesis in immunologically mediated glomerular disease have been disappointing. There is some evidence, however, that these drugs may be of benefit in ureteric obstruction and renal transplant rejection.


Subject(s)
Kidney Diseases/physiopathology , Prostaglandins/physiology , Thromboxanes/physiology , Acute Kidney Injury/physiopathology , Animals , Anti-Inflammatory Agents/pharmacology , Cyclooxygenase Inhibitors , Dietary Fats/metabolism , Fatty Acids/metabolism , Glomerulonephritis/drug therapy , Glomerulonephritis/metabolism , Glomerulonephritis/physiopathology , Graft Rejection/drug effects , Humans , Kidney Diseases/drug therapy , Kidney Diseases/metabolism , Kidney Transplantation , Nephrectomy , Prostaglandins/metabolism , Prostaglandins/therapeutic use , Renal Circulation/drug effects , Thromboxane-A Synthase/antagonists & inhibitors , Thromboxanes/metabolism , Thromboxanes/therapeutic use
18.
J Trauma ; 20(9): 722-5, 1980 Sep.
Article in English | MEDLINE | ID: mdl-6997504

ABSTRACT

Decreasing progressive dermal ischemia after burning could theoretically limit the amount of skin necrosis to the zone of coagulation. Methylprednisolone, aspirin, indomethacin, imidazole, dipyridamole, and methimazole have been shown to prevent dermal ischemia, suggesting that prostaglandins and/or thromboxanes may play a role in its pathogenesis. Specific antiprostaglandin antibodies (anti-PgE2, PgF2 alpha, PgI2, and TxA2) were reacted with tissue biopsies of burned guinea pig skin at various time intervals postburn. An immunoperoxidase technique with goat anti-rabbit immunoglobulin and horseradish peroxidase demonstrated the presence of the specific arachidonic acid metabolites. The burned tissue showed high levels of PgE2 and TxA2. The effects of three thromboxane inhibitors, imidazole, methimazole, and dipyridamole, on dermal ischemia were studied. Xenon133 washout studies were performed in burned and unburned areas. Tissue half-life of Xenon was prolonged in burned, untreated areas but this rapidly decreased in antithromboxane-treated burns. Repeated antiprostaglandin and antithromboxane antibody-immunoperoxidase studies on tissue from the thromboxane inhibitor-treated animals showed that PgE2, PgF2 alpha, and PgI2 were at the same levels as in untreated animals, but thromboxane (TxA2) was essentially absent, suggesting that thromboxane may be responsible for the progressive dermal ischemia after burning and that decreasing its production can increase dermal perfusion.


Subject(s)
Burns/drug therapy , Ischemia/prevention & control , Skin/blood supply , Thromboxanes/antagonists & inhibitors , Thromboxanes/therapeutic use , Animals , Burns/physiopathology , Epoprostenol/physiology , Female , Guinea Pigs , Prostaglandins E/physiology , Prostaglandins F/physiology , Thromboxanes/biosynthesis , Xenon
20.
Prostaglandins Med ; 3(3): 139-46, 1979 Sep.
Article in English | MEDLINE | ID: mdl-552102

ABSTRACT

Pinane thromboxane A2 (PTA2) an analog of thromboxane A2 that inhibits the formation of thromboxanes as well as antagonizes their biological actions, at an infusion rate of 1.0 mumole . kg-1 . h-1, prolonged survival in traumatic shock in rats. PTA2 also prevented the accumulation of thromboxane B2, the lysosomal protease, cathepsin D, and the cardiotoxic peptide MDF in the circulating blood.


Subject(s)
Shock, Traumatic/drug therapy , Thromboxane A2/therapeutic use , Thromboxanes/therapeutic use , Animals , Bicyclic Monoterpenes , Cathepsins/blood , Myocardial Depressant Factor/blood , Rats , Shock, Traumatic/blood , Thromboxane A2/analogs & derivatives , Thromboxane B2/blood , Thromboxanes/antagonists & inhibitors
SELECTION OF CITATIONS
SEARCH DETAIL
...