Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 88
Filter
Add more filters










Publication year range
1.
Int J Biomed Imaging ; 2023: 5366733, 2023.
Article in English | MEDLINE | ID: mdl-37362614

ABSTRACT

We propose an enhanced method to accurately retrieve time-activity curves (TACs) of blood and tissue from dynamic 2-deoxy-2-[18F]fluoro-D-glucose ([18F]FDG) positron emission tomography (PET) cardiac images of mice. The method is noninvasive and consists of using a constrained nonnegative matrix factorization algorithm (CNMF) applied to the matrix (A) containing the intensity values of the voxels of the left ventricle (LV) PET image. CNMF factorizes A into nonnegative matrices H and W, respectively, representing the physiological factors (blood and tissue) and their associated weights, by minimizing an extended cost function. We verified our method on 32 C57BL/6 mice, 14 of them with acute myocardial infarction (AMI). With CNMF, we could break down the mouse LV into myocardial and blood pool images. Their corresponding TACs were used in kinetic modeling to readily determine the [18F]FDG influx constant (Ki) required to compute the myocardial metabolic rate of glucose. The calculated Ki values using CNMF for the heart of control mice were in good agreement with those published in the literature. Significant differences in Ki values for the heart of control and AMI mice were found using CNMF. The values of the elements of W agreed well with the LV structural changes induced by ligation of the left coronary artery. CNMF was compared with the recently published method based on robust unmixing of dynamic sequences using regions of interest (RUDUR). A clear improvement of signal separation was observed with CNMF compared to the RUDUR method.

2.
J Am Heart Assoc ; 12(4): e028056, 2023 02 21.
Article in English | MEDLINE | ID: mdl-36752268

ABSTRACT

Background Deep vein thrombosis (DVT) is the primary cause of pulmonary embolism and the third most life-threatening cardiovascular disease in North America. Post-DVT anticoagulants, such as warfarin, heparin, and direct oral anticoagulants, reduce the incidence of subsequent venous thrombi. However, all currently used anticoagulants affect bleeding time at various degrees, and there is therefore a need for improved therapeutic regimens in DVT. It has recently been shown that mast cells play a crucial role in a DVT murine model. The underlying mechanism involved in the prothrombotic properties of mast cells, however, has yet to be identified. Methods and Results C57BL/6 mice and mouse mast cell protease-4 (mMCP-4) genetically depleted mice (mMCP-4 knockout) were used in 2 mouse models of DVT, partial ligation (stenosis) and ferric chloride-endothelial injury model of the inferior vena cava. Thrombus formation and impact of genetically repressed or pharmacologically (specific inhibitor TY-51469) inhibited mMCP-4 were evaluated by morphometric measurements of thrombi immunochemistry (mouse and human DVT), color Doppler ultrasound, bleeding times, and enzymatic activity assays ex vivo. Recombinant chymases, mMCP-4 (mouse) and CMA-1 (human), were used to characterize the interaction with murine and human plasmin, respectively, by mass spectrometry and enzymatic activity assays. Inhibiting mast cell-generated mMCP-4, genetically or pharmacologically, resolves and prevents venous thrombus formation in both DVT models. Inferior vena cava blood flow obstruction was observed in the stenosis model after 6 hours of ligation, in control- but not in TY-51469-treated mice. In addition, chymase inhibition had no impact on bleeding times of healthy or DVT mice. Furthermore, endogenous chymase limits plasmin activity in thrombi ex vivo. Recombinant mouse or human chymase degrades/inactivates purified plasmin in vitro. Finally, mast cell-containing immunoreactive chymase was identified in human DVT. Conclusions This study identified a major role for mMCP-4, a granule-localized protease of chymase type, in DVT formation. These findings support a novel pharmacological strategy to resolve or prevent DVT without affecting the coagulation cascade through the inhibition of chymase activity.


Subject(s)
Fibrinolysin , Venous Thrombosis , Mice , Humans , Animals , Chymases/metabolism , Bleeding Time , Disease Models, Animal , Constriction, Pathologic , Mice, Inbred C57BL , Venous Thrombosis/prevention & control , Anticoagulants
3.
J Pharmacol Exp Ther ; 376(2): 213-221, 2021 02.
Article in English | MEDLINE | ID: mdl-33154104

ABSTRACT

Mouse mast cell protease 4 (mMCP-4), the murine functional analog to the human chymase, is a serine protease synthesized and stored in mast cell secretory granules. Our previous studies reported physiologic and pathologic roles for mMCP-4 in the maturation and synthesis of the vasoactive peptide endothelin-1 (ET-1) from its precursor, big ET-1. The aim of this study was to investigate the impact of mast cell degranulation or stabilization on mMCP-4-dependent pressor responses after the administration of big ET-1 or angiotensin I (Ang I). In anesthetized mice, mast cell degranulation induced by compound 48/80 (C48/80) or stabilization by cromolyn enhanced or repressed, respectively, the dose-dependent vasopressor responses to big ET-1 in wild-type (WT) mice but not in mMCP-4 knockout mice in a chymase inhibitor (TY-51469)-sensitive fashion. In addition, mMCP-4-dependent hydrolysis of the fluorogenic substrate Suc-Leu-Leu-Val-Tyr-7-amino-4-methylcoumarin was depleted or enhanced in peritoneal mast cells isolated from mice pretreated with C48/80 or cromolyn, respectively. Furthermore, C48/80 or cromolyn markedly increased or abolished, respectively, ET-1 (1-31) conversion from exogenous big ET-1 in WT mice peritoneal fluid-isolated mast cells, in vitro. Finally, the vasopressor responses to Ang I were unaffected by mast cell activation or stabilization, whereas those induced by the angiotensin-converting enzyme-resistant Ang I analog, [Pro11, D-Ala12] Ang I, were potentiated by C48/80. Altogether, the present study shows that mast cell activation enhances the mMCP-4-dependent vasoactive properties of big ET-1 but not Ang I in the mouse model. SIGNIFICANCE STATEMENT: The current work demonstrates a significant role for mast cell stability in the cardiovascular pharmacology of big endothelin-1 but not angiotensin I in the murine systemic circulation.


Subject(s)
Angiotensin I/pharmacology , Blood Pressure , Cell Degranulation , Endothelin-1/pharmacology , Mast Cells/physiology , Serine Endopeptidases/metabolism , Animals , Cells, Cultured , Chymases/antagonists & inhibitors , Cromolyn Sodium/pharmacology , Enzyme Inhibitors/pharmacology , Male , Mast Cell Stabilizers/pharmacology , Mast Cells/drug effects , Mast Cells/metabolism , Mice , Mice, Inbred C57BL , Peritoneum/cytology , Serine Endopeptidases/genetics , Sulfonamides/pharmacology , Thiophenes/pharmacology
4.
J Pharmacol Exp Ther ; 370(3): 437-446, 2019 09.
Article in English | MEDLINE | ID: mdl-31248979

ABSTRACT

Multiple sclerosis is a neurodegenerative disease affecting predominantly female patients between 20 and 45 years of age. We previously reported the significant contribution of mouse mast cell protease 4 (mMCP-4) in the synthesis of endothelin-1 (ET-1) in healthy mice and in a murine model of experimental autoimmune encephalomyelitis (EAE). In the current study, the cardiovascular effects of ET-1 and big endothelin-1 (big-ET-1) administered systemically or intrathecally were assessed in the early preclinical phase of EAE in telemetry instrumented/conscious mice. Chymase-specific enzymatic activity was also measured in the lung, brain, and mast cell extracts in vitro. Finally, the impact of EAE immunization was studied on the pulmonary and brain mRNA expression of different genes of the endothelin pathway, interleukin-33 (IL-33), and monitoring of immunoreactive tumor necrosis factor-α (TNF-α). Systemically or intrathecally administered big-ET-1 triggered increases in blood pressure in conscious mice. One week post-EAE, the pressor responses to big-ET-1 were potentiated in wild-type (WT) mice but not in mMCP-4 knockout (KO) mice. EAE triggered mMCP-4-specific activity in cerebral homogenates and peritoneal mast cells. Enhanced pulmonary, but not cerebral preproendothelin-1 and IL-33 mRNA were found in KO mice and further increased 1 week post-EAE immunization, but not in WT animals. Finally, TNF-α levels were also increased in serum from mMCP-4 KO mice, but not WT, 1 week post-EAE. Our study suggests that mMCP-4 activity is enhanced both centrally and systemically in a mouse model of EAE.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/metabolism , Endothelin-1/administration & dosage , Endothelin-1/pharmacology , Serine Endopeptidases/metabolism , Animals , Brain/drug effects , Brain/metabolism , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/pathology , Encephalomyelitis, Autoimmune, Experimental/physiopathology , Gene Knockout Techniques , Hemodynamics/drug effects , Injections, Spinal , Interleukin-33/deficiency , Interleukin-33/genetics , Lung/drug effects , Lung/metabolism , Mast Cells/drug effects , Mast Cells/metabolism , Mice , RNA, Messenger/genetics , RNA, Messenger/metabolism , Serine Endopeptidases/deficiency , Serine Endopeptidases/genetics , Up-Regulation/drug effects
5.
Pharmacol Ther ; 194: 145-160, 2019 02.
Article in English | MEDLINE | ID: mdl-30291906

ABSTRACT

Endothelins were discovered more than thirty years ago as potent vasoactive compounds. Beyond their well-documented cardiovascular properties, however, the contributions of the endothelin pathway have been demonstrated in several neuroinflammatory processes and the peptides have been reported as clinically relevant biomarkers in neurodegenerative diseases. Several studies report that endothelin-1 significantly contributes to the progression of neuroinflammatory processes, particularly during infections in the central nervous system (CNS), and is associated with a loss of endothelial integrity at the blood brain barrier level. Because of the paucity of clinical trials with endothelin-1 antagonists in several infectious and non-infectious neuroinflammatory diseases, it remains an open question whether the 21 amino acid peptide is a mediator/modulator rather than a biomarker of the progression of neurodegeneration. This review focuses on the potential roles of endothelins in the pathology of neuroinflammatory processes, including infectious diseases of viral, bacterial or parasitic origin in which the synthesis of endothelins or its pharmacology have been investigated from the cell to the bedside in several cases, as well as in non-infectious inflammatory processes such as neurodegenerative disorders like Alzheimers Disease or central nervous system vasculitis.


Subject(s)
Endothelins/metabolism , Nervous System Diseases/metabolism , Animals , Humans , Inflammation/metabolism
6.
Front Pharmacol ; 9: 868, 2018.
Article in English | MEDLINE | ID: mdl-30233357

ABSTRACT

Chymase, a mast cell serine protease involved in the generation of multiple cardiovascular factors, such as angiotensin II and endothelin-1 (ET-1), is elevated and participates in tissue degeneration after permanent myocardial infarction (PMI). Anesthetized 4-month old male wild-type (WT) C57BL/6J mice and mouse mast cell protease-4 knockout (mMCP-4 KO) congeners were subjected to ligation of the left anterior descending (LAD) coronary artery. A group of mice was then subjected to Kaplan-Meier 28-day survival analysis. In another group of mice, 18F-fluorodeoxyglucose positron emission tomography (PET) was performed to evaluate heart function and the infarcted zone 3 days post-PMI surgery. Cardiac morphology following PMI was evaluated on formalin-fixed heart slices and glycoproteomic analysis was performed using mass spectrometry. Finally, cardiac and lung tissue content of immunoreactive ET-1 was determined. PMI caused 60% mortality in WT mice, due to left ventricular wall rupture, and 7% in mMCP-4 KO mice. Cardiac PET analysis revealed a significant reduction in left ventricular volume (systolic and diastolic) and preserved the ejection fraction in mMCP-4 KO compared to WT animals. The infarcted area, apoptotic signaling and wall remodeling were significantly decreased in mMCP-4 KO mice compared to their WT congeners, while collagen deposition was increased. Glycoproteomic analysis showed an increase in apolipoprotein A1, an established chymase substrate in mMCP-4 KO mice compared to WT mice post-PMI. ET-1 levels were increased in the lungs of WT, but not mMCP-4 KO mice, 24 h post-PMI. Thus, the genetic deletion of mMCP-4 improved survival and heart function post-PMI.

7.
Can J Physiol Pharmacol ; 96(7): 655-661, 2018 Jul.
Article in English | MEDLINE | ID: mdl-29756482

ABSTRACT

The interaction between vascular endothelial cells (VECs) and vascular smooth muscle cells (VSMCs) plays an important role in the modulation of vascular tone. There is, however, no information on whether direct physical communication regulates the intracellular calcium levels of human VECs (hVECs) and (or) human VSMCs (hVSMCs). Thus, the objective of the study is to verify whether co-culture of hVECs and hVSMCs modulates cytosolic ([Ca2+]c) and nuclear calcium ([Ca2+]n) levels via physical contact and (or) factors released by both cell types. Quantitative 3D confocal microscopy for [Ca2+]c and [Ca2+]n measurement was performed in cultured hVECs or hVSMCs or in co-culture of hVECs-hVSMCs. Our results show that: (1) physical contact between hVECs-hVECs or hVSMCs-hVSMCs does not affect [Ca2+]c and [Ca2+]n in these 2 cell types; (2) physical contact between hVECs and hVSMCs induces a significant increase only of [Ca2+]n of hVECs without affecting the level of [Ca2+]c and [Ca2+]n of hVSMCs; and (3) preconditioned culture medium of hVECs or hVSMCs does not affect [Ca2+]c and [Ca2+]n of both types of cells. We concluded that physical contact between hVECs and hVSMCs only modulates [Ca2+]n in hVECs. The increase of [Ca2+]n in hVECs may modulate nuclear functions that are calcium dependent.


Subject(s)
Calcium/metabolism , Cell Communication/physiology , Endothelial Cells/physiology , Myocytes, Smooth Muscle/physiology , Adolescent , Adult , Cations, Divalent/metabolism , Cell Nucleus/metabolism , Cells, Cultured , Coculture Techniques , Cytosol/metabolism , Endothelium, Vascular/cytology , Healthy Volunteers , Humans , Microscopy, Confocal , Muscle, Smooth, Vascular/cytology , Primary Cell Culture , Young Adult
8.
Can J Physiol Pharmacol ; 95(10): 1298-1305, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28732172

ABSTRACT

Our previous work showed the presence of endothelin-1 (ET-1) receptors, ETA and ETB, in human vascular endothelial cells (hVECs). In this study, we wanted to verify whether ET-1 plays a role in the survival of hVECs via the activation of its receptors ETA and (or) ETB (ETAR and ETBR, respectively). Our results showed that treatment of hVECs with ET-1 prevented apoptosis induced by genistein, an effect that was mimicked by treatment with ETBR-specific agonist IRL1620. Furthermore, blockade of ETBR with the selective ETBR antagonist A-192621 prevented the anti-apoptotic effect of ET-1 in hVECs. However, activation of ETA receptor alone did not seem to contribute to the anti-apoptotic effect of ET-1. In addition, the anti-apoptotic effect of ETBR was found to be associated with caspase 3 inhibition and does not depend on the density of this type of receptor. In conclusion, our results showed that ET-1 possesses an anti-apoptotic effect in hVECs and that this effect is mediated, to a great extent, via the activation of ETBR. This study revealed a new role for ETBR in the survival of hVECs.


Subject(s)
Apoptosis/drug effects , Endothelial Cells/drug effects , Endothelin-1/pharmacology , Receptor, Endothelin A/agonists , Receptor, Endothelin B/agonists , Caspase 3/metabolism , Cell Survival/drug effects , Cells, Cultured , Endothelial Cells/metabolism , Endothelial Cells/pathology , Endothelin A Receptor Antagonists/pharmacology , Endothelin B Receptor Antagonists/pharmacology , Genistein/toxicity , Humans , Receptor, Endothelin A/metabolism , Receptor, Endothelin B/metabolism , Signal Transduction/drug effects
9.
Can J Physiol Pharmacol ; 95(10): 1224-1229, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28738162

ABSTRACT

Endocardial endothelial cells (EECs) constitute an important component of the heart. These cells form a monolayer that covers the cavities of the right (EECRs) and left (EECLs) ventricles. They play an important role in cardiac excitation-contraction coupling via their secretion of cardioactive factors such as neuropeptide Y (NPY). They also contribute to cardiac pathology such as arrhythmia, hypertrophy, and heart failure. Differences between EECRs and EECLs contribute to tuning of circulating factors at the entry and exit of the ventricles. NPY, via activation of its receptors, modulates the excitation-secretion coupling of EECs, thus, indirectly modulating cardiac function and remodeling.


Subject(s)
Endocardium/metabolism , Endothelial Cells/metabolism , Heart Ventricles/metabolism , Neuropeptide Y/metabolism , Receptors, Neuropeptide Y/metabolism , Animals , Cardiovascular Diseases/metabolism , Cardiovascular Diseases/pathology , Cardiovascular Diseases/physiopathology , Endocardium/pathology , Endocardium/physiopathology , Endothelial Cells/pathology , Heart Ventricles/pathology , Heart Ventricles/physiopathology , Humans , Signal Transduction
10.
Mediators Inflamm ; 2016: 9797021, 2016.
Article in English | MEDLINE | ID: mdl-27610007

ABSTRACT

Experimental autoimmune encephalomyelitis (EAE) is a mouse model that reproduces cardinal signs of clinical, histopathological, and immunological features found in Multiple Sclerosis (MS). Mast cells are suggested to be involved in the main inflammatory phases occurring during EAE development, possibly by secreting several autacoids and proteases. Among the latter, the chymase mouse mast cell protease 4 (mMCP-4) can contribute to the inflammatory response by producing endothelin-1 (ET-1). The aim of this study was to determine the impact of mMCP-4 on acute inflammatory stages in EAE. C57BL/6 wild type (WT) or mMCP-4 knockout (KO) mice were immunized with MOG35-55 plus complete Freund's adjuvant followed by pertussis toxin. Immunized WT mice presented an initial acute phase characterized by progressive increases in clinical score, which were significantly reduced in mMCP-4 KO mice. In addition, higher levels of spinal myelin were found in mMCP-4 KO as compared with WT mice. Finally, whereas EAE triggered significant increases in brain levels of mMCP-4 mRNA and immunoreactive ET-1 in WT mice, the latter peptide was reduced to basal levels in mMCP-4 KO congeners. Together, the present study supports a role for mMCP-4 in the early inflammatory phases of the disease in a mouse model of MS.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/enzymology , Serine Endopeptidases/metabolism , Animals , Astrocytes/metabolism , Brain/enzymology , Brain/metabolism , Brain/pathology , Encephalomyelitis, Autoimmune, Experimental/metabolism , Encephalomyelitis, Autoimmune, Experimental/pathology , Endothelin-1/metabolism , Female , Fluorescent Antibody Technique , Inflammation/enzymology , Inflammation/metabolism , Inflammation/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microglia/metabolism , Serine Endopeptidases/deficiency , Serine Endopeptidases/genetics
11.
Life Sci ; 159: 71-75, 2016 Aug 15.
Article in English | MEDLINE | ID: mdl-26976326

ABSTRACT

AIMS: To determine the impact of mixed endothelin receptor antagonist and mouse mast cell protease-4 (mMCP-4) in the development of atherosclerosis in the mouse model. MATERIALS AND METHODS: Apolipoprotein E (ApoE) KO mice were crossed with mMCP-4 KO mice to generate ApoE/mMCP-4 double KO mice. Atherosclerosis was induced with a normal- or high-fat diet for 12, 27 or 52weeks. Macitentan (30mg/kg/day), a dual ETA/ETB receptor antagonist, was given orally for 6weeks (27week protocol). At sacrifice, aortas and brachiocephalic arteries (BCAs) were collected. En face Sudan IV staining was performed on aortas and BCA sections were subjected to Masson's trichrome stain and α-smooth muscle actin labeling. KEY FINDINGS: Under normal diet, both macitentan treatment and the absence of mMCP-4 reduced the development of aortic atherosclerotic lesions in 27-week old ApoE KO mice, but mMCP-4 deletion failed to maintain this effect on 52-week old mice. Under high-fat diet (WD), macitentan, but not the absence of mMCP-4, reduced aortic lesion development in ApoE KO mice. On BCA lesions of 27-week old WD mice, macitentan treatment had a small impact while mMCP-4 deletion showed improved features of plaque stability. SIGNIFICANCE: These results suggest that the inhibition of mMCP-4 reduces lesion spreading in the earlier phases of atherosclerosis development and can help stabilise the more advanced plaque. Macitentan treatment was more effective to prevent lesion spreading but did not improve plaque features to the same extent.


Subject(s)
Aorta/pathology , Atherosclerosis/pathology , Endothelin Receptor Antagonists/pharmacology , Pyrimidines/pharmacology , Serine Endopeptidases/metabolism , Sulfonamides/pharmacology , Animals , Mice , Mice, Knockout , Serine Endopeptidases/genetics
12.
PLoS One ; 10(8): e0128881, 2015.
Article in English | MEDLINE | ID: mdl-26248341

ABSTRACT

Increased blood glucose concentrations promote reactions between glucose and proteins to form advanced glycation end-products (AGE). Circulating AGE in the blood plasma can activate the receptor for advanced end-products (RAGE), which is present on both endothelial and vascular smooth muscle cells (VSMC). RAGE exhibits a complex signaling that involves small G-proteins and mitogen activated protein kinases (MAPK), which lead to increased nuclear factor kappa B (NF-κB) activity. While RAGE signaling has been previously addressed in endothelial cells, little is known regarding its impact on the function of VSMC. Therefore, we hypothesized that RAGE signaling leads to alterations in the mechanical and functional properties of VSMC, which could contribute to complications associated with diabetes. We demonstrated that RAGE is expressed and functional in the A7r5 VSMC model, and its activation by AGE significantly increased NF-κB activity, which is known to interfere with the contractile phenotype of VSMC. The protein levels of the contraction-related transcription factor myocardin were also decreased by RAGE activation with a concomitant decrease in the mRNA and protein levels of transgelin (SM-22α), a regulator of VSMC contraction. Interestingly, we demonstrated that RAGE activation increased the overall cell rigidity, an effect that can be related to an increase in myosin activity. Finally, although RAGE stimulation amplified calcium signaling and slightly myosin activity in VSMC challenged with vasopressin, their contractile capacity was negatively affected. Overall, RAGE activation in VSMC could represent a keystone in the development of vascular diseases associated with diabetes by interfering with the contractile phenotype of VSMC through the modification of their mechanical and functional properties.


Subject(s)
Muscle, Smooth, Vascular/cytology , Receptor for Advanced Glycation End Products/metabolism , Signal Transduction , Animals , Biomechanical Phenomena , Calcium/metabolism , Cell Line , Diabetes Complications/etiology , Diabetes Complications/metabolism , Glycation End Products, Advanced/metabolism , Humans , Muscle Contraction , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/physiology , NF-kappa B/metabolism , Rats , Serum Albumin/metabolism , Vascular Diseases/etiology , Vascular Diseases/metabolism , Glycated Serum Albumin
13.
Vascul Pharmacol ; 73: 124-37, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26141931

ABSTRACT

OBJECTIVES: The effects of chronic fluoxetine treatment were investigated on blood pressure and on vascular reactivity in the isolated rat aorta. METHODS AND RESULTS: Male Wistar rats were treated with fluoxetine (10 mg/kg/day) for 21 days. Fluoxetine increased systolic blood pressure. Chronic, but not acute, fluoxetine treatment increased the contractile response induced by phenylephrine, serotonin (5-HT) and KCl in endothelium-intact rat aortas. L-NAME and ODQ did not alter the contraction induced by phenylephrine and 5-HT in aortic rings from fluoxetine-treated rats. Tiron, SC-560 and AH6809 reversed the increase in the contractile response to phenylephrine and 5-HT in aortas from fluoxetine-treated rats. Fluoxetine treatment increased superoxide anion generation (O2(-)) and the expression of cyclooxygenase (COX)-1 in the rat aorta. Reduced expression of nNOS, but not eNOS or iNOS was observed in animals treated with fluoxetine. Fluoxetine treatment increased prostaglandin (PG)F2α levels but did not affect thromboxane (TX)B2 levels in the rat aorta. Reduced hydrogen peroxide (H2O2) levels and increased catalase (CAT) activity were observed after treatment. CONCLUSIONS: The major new finding of our study is that chronic fluoxetine treatment induces endothelial dysfunction, which alters vascular responsiveness by a mechanism that involves increased oxidative stress and the generation of a COX-derived vasoconstrictor prostanoid (PGF2α). Moreover, our results evidenced a relation between the period of treatment with fluoxetine and the magnitude in the increment of blood pressure. Finally, our findings raise the possibility that fluoxetine treatment increases the risk for vascular injury, a response that could predisposes to cardiovascular diseases.


Subject(s)
Aorta/drug effects , Blood Pressure/drug effects , Dinoprost/metabolism , Endothelium, Vascular/drug effects , Fluoxetine/toxicity , Oxidative Stress/drug effects , Vasoconstriction/drug effects , Acetylcholinesterase/metabolism , Animals , Aorta/metabolism , Aorta/physiopathology , Catalase/metabolism , Cyclooxygenase 1/genetics , Cyclooxygenase 1/metabolism , Dose-Response Relationship, Drug , Endothelium, Vascular/metabolism , Endothelium, Vascular/physiopathology , GPI-Linked Proteins/metabolism , Hydrogen Peroxide/metabolism , Male , Membrane Proteins/genetics , Membrane Proteins/metabolism , Nitric Oxide Synthase Type I/genetics , Nitric Oxide Synthase Type I/metabolism , Rats, Wistar , Superoxide Dismutase/metabolism , Superoxides/metabolism , Time Factors , Vasoconstrictor Agents/pharmacology
14.
Anal Biochem ; 478: 73-81, 2015 Jun 01.
Article in English | MEDLINE | ID: mdl-25800564

ABSTRACT

Protein glycation is a ubiquitous process involved in vascular complications observed in diabetes. Glyoxal (GO), an intracellular reactive oxoaldehyde that is one of the most potent glycation agents, readily reacts with amines present on proteins to produce the lysine-derived adduct carboxymethyllysine, which is a prevalent advanced glycation end-product (AGE). Our group previously showed that cell exposure to GO leads to an alteration in the cell contractile activity that could occur as a result of the glycation of various proteins regulating the cell contractile machinery. Here, we measured the extent of glycation on three functionally distinct proteins known to participate in cell contraction and cytoskeletal organization-Rho-kinase (ROCK), actin, and gelsolin (GSN)-using an assay based on the reaction of the cell membrane-permeable fluorescent probe carboxyfluorescein diacetate succinimidyl ester (CFDA-SE), which reacts with primary amine groups of proteins. By combining CFDA-SE fluorescence and Western blot detection, we observed (following GO incubation) increased glycation of actin and ROCK as well as an increased interaction between actin and GSN as observed by co-immunoprecipitation. Thus, we conclude that the use of the fluorescent probe CFDA-SE offers an interesting alternative to perform a comparative analysis of the extent of intracellular protein glycation in live cells.


Subject(s)
Fluoresceins/metabolism , Fluorescent Dyes/metabolism , Glycosylation , Succinimides/metabolism , Actins/analysis , Actins/metabolism , Blotting, Western , Cell Line , Fluoresceins/analysis , Fluorescent Dyes/analysis , Gelsolin/analysis , Gelsolin/metabolism , Glyoxal/metabolism , Humans , Microscopy, Fluorescence , Succinimides/analysis , rho-Associated Kinases/analysis , rho-Associated Kinases/metabolism
15.
Can J Physiol Pharmacol ; 93(4): 291-7, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25741585

ABSTRACT

The objective of this work was to verify whether, as in the case of the plasma membrane of human vascular smooth muscle cells (hVSMCs), cytosolic ET-1-induced increase of nuclear calcium is mediated via the activation of calcium influx through the steady-state R-type calcium channel. Pharmacological tools to identify the R-type calcium channels, as well as real 3-D confocal microscopy imaging techniques coupled to calcium fluorescent probes, were used to study the effect of cytosolic ET-1 on nuclear calcium in isolated nuclei of human hepatocytes and plasma membrane perforated hVSMCs. Our results showed that pre-treatment with pertussis toxin (PTX) or cholera toxin (CTX) prevented cytosolic ET-1 (10(-9) mol/L) from inducing a sustained increase in nuclear calcium. Furthermore, the L-type calcium channel blocker nifedipine did not prevent cytosolic ET-1 from inducing an increase in nuclear calcium, as opposed to the dual L- and R-type calcium channel blocker isradipine (PN200-110) (in the presence of nifedipine). In conclusion, the preventative effect with PTX and CTX, and the absence of an effect with nifedipine, as well as the blockade by isradipine on cytosolic ET-1-induced increase in nuclear calcium, suggest that this nuclear calcium influx in hVSMCs is due to activation of the steady-state R-type calcium channel. The sarcolemmal and nuclear membrane R-type calcium channels in hVSMCs are involved in ET-1 modulation of vascular tone in physiology and pathology.


Subject(s)
Calcium Channels, R-Type/metabolism , Calcium Signaling , Cation Transport Proteins/metabolism , Cell Nucleus/metabolism , Cytosol/metabolism , Endothelin-1/metabolism , Muscle, Smooth, Vascular/metabolism , Nuclear Envelope/metabolism , Aorta , Calcium Channel Blockers/pharmacology , Calcium Channels, R-Type/chemistry , Calcium Signaling/drug effects , Cation Transport Proteins/chemistry , Cell Membrane Permeability/drug effects , Cell Nucleus/drug effects , Cells, Cultured , Cholera Toxin/pharmacology , Cytosol/drug effects , Hepatocytes/cytology , Hepatocytes/drug effects , Hepatocytes/metabolism , Humans , Imaging, Three-Dimensional , Membrane Fusion/drug effects , Membrane Transport Modulators/pharmacology , Microscopy, Confocal , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/drug effects , Nuclear Envelope/drug effects , Pertussis Toxin/pharmacology , Sarcolemma/drug effects , Sarcolemma/metabolism
16.
Biochem Pharmacol ; 94(2): 91-100, 2015 Mar 15.
Article in English | MEDLINE | ID: mdl-25667044

ABSTRACT

Important structural differences imply that human and mouse mast cell chymases may differ with respect to their enzymatic properties. We compared in this study the catalytic efficiencies of recombinant human chymase (rCMA1) and its functional murine homologue recombinant mouse mast cell protease-4 (rmMCP-4) toward a fluorogenic chymase substrate (Suc-Ala-Ala-Pro-Phe-7-amino-4-methylcoumarin (AMC) and by their ability to convert Big-endothelin (ET)-1 into ET-1 (1-31) using a LC/MS/MS system. Activities toward a fluorogenic substrate (Suc-Leu-Leu-Val-Tyr-AMC) and Big ET-1 were also measured in extracts from mouse peritoneal mast cells, LUVA human mast cell-like cells and human aortas. The specificity of these activities was assessed with the chymase inhibitor TY-51469 (2-[4-(5-fluoro-3-methylbenzo[b]thiophen-2-yl)sulfonamido-3-methanesulfonyl-phenyl]thiazole-4-carboxylic acid). For similar affinities, rmMCP-4 showed a higher activity toward the fluorogenic substrate and a higher ability to process Big ET-1 as compared to recombinant CMA1 (chymase activity (kcat/KM in µM(-1)s(-1)): 2.29 × 10(-4)vs. 6.41 × 10(-6); ET-1 (1-31) production: 2.19 × 10(-3)vs. 6.57 × 10(-5)), and both of these activities of mouse and human chymase were sensitive to TY-51469. Furthermore, extracts from mouse peritoneal mast cells, LUVA cells and human aorta homogenates contained processing activities toward the fluorogenic chymase substrate as well as Big ET-1, all of which were sensitive to TY-51469. Finally, the pressor responses to Big ET-1 but not to ET-1 were significantly reduced in conscious and free moving mMCP-4 KO mice when compared to wild type congeners. Our results suggest that both mouse and human chymases have potent ET-1 (1-31)-producing abilities, with the murine isoform being more efficient.


Subject(s)
Chymases/antagonists & inhibitors , Endothelin-1/analogs & derivatives , Enzyme Inhibitors/pharmacology , Peptide Fragments/chemical synthesis , Serine Endopeptidases/metabolism , Animals , Chromatography, Liquid , Chymases/metabolism , Endothelin-1/chemical synthesis , Humans , Mice , Mice, Inbred C57BL , Serine Endopeptidases/genetics , Tandem Mass Spectrometry
18.
J Pharmacol Exp Ther ; 346(1): 31-7, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23596057

ABSTRACT

The serine protease chymase has been reported to generate intracardiac angiotensin-II (Ang-II) from Ang-I as well as an intermediate precursor of endothelin-1 (ET-1), ET-1 (1-31) from Big-ET-1. Although humans possess only one chymase, several murine isoforms are documented, each with its own specific catalytic activity. Among these, mouse mast cell protease 4 (mMCP-4) is the isoform most similar to the human chymase for its activity. The aim of this study was to characterize the capacity of mMCP-4 to convert Big-ET-1 into its bioactive metabolite, ET-1, in vitro and in vivo in the mouse model. Basal mean arterial pressure did not differ between wild-type (WT) and mMCP-4(-/-) mice. Systemic administration of Big-ET-1 triggered pressor responses and increased blood levels of immunoreactive (IR) ET-1 (1-31) and ET-1 that were reduced by more than 50% in mMCP-4 knockout (-/-) mice compared with WT controls. Residual responses to Big-ET-1 in mMCP-4(-/-) mice were insensitive to the enkephalinase/neutral endopeptidase inhibitor thiorphan and the specific chymase inhibitor TY-51469 {2-[4-(5-fluoro-3-methylbenzo[b]thiophen-2-yl)sulfonamido-3-methanesulfonylphenyl]thiazole-4-carboxylic acid}. Soluble fractions from the lungs, left cardiac ventricle, aorta, and kidneys of WT but not mMCP-4(-/-) mice generated ET-1 (1-31) from exogenous Big-ET-1 in a TY-51469-sensitive fashion as detected by high-performance liquid chromatography/ matrix-assisted laser desorption/ionization-mass spectrometry. Finally, pulmonary endogenous levels of IR-ET-1 were reduced by more than 40% in tissues derived from mMCP-4(-/-) mice compared with WT mice. Our results show that mMCP-4 plays a pivotal role in the dynamic conversion of systemic Big-ET-1 to ET-1 in the mouse model.


Subject(s)
Aorta/enzymology , Endothelin-1/metabolism , Heart Ventricles/enzymology , Serine Endopeptidases/metabolism , Animals , Aorta/drug effects , Aorta/metabolism , Carboxypeptidases A/biosynthesis , Carboxypeptidases A/genetics , Carboxypeptidases A/metabolism , Drug Resistance , Endothelin-1/analogs & derivatives , Endothelin-1/blood , Gene Expression Regulation, Enzymologic , Heart Ventricles/drug effects , Heart Ventricles/metabolism , Hemodynamics/drug effects , Isoenzymes/biosynthesis , Isoenzymes/genetics , Isoenzymes/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Organ Specificity , Peptide Fragments/blood , Peptide Fragments/metabolism , Protein Processing, Post-Translational/drug effects , Proteolysis/drug effects , RNA, Messenger/metabolism , Serine Endopeptidases/chemistry , Serine Endopeptidases/genetics , Serine Proteinase Inhibitors/pharmacology , Sulfonamides/pharmacology , Thiophenes/pharmacology , Thiorphan/pharmacology
19.
PLoS One ; 8(2): e58023, 2013.
Article in English | MEDLINE | ID: mdl-23469133

ABSTRACT

Expression of the Endothelin-2 (Edn2) mRNA is greatly increased in the photoreceptors (PRs) of mouse models of inherited PR degeneration (IPD). To examine the role of Edn2 in mutant PR survival, we generated Edn2(-/-) mice carrying homozygous Pde6b(rd1) alleles or the Tg(RHO P347S) transgene. In the Edn2(-/-) background, PR survival increased 110% in Pde6b(rd1/rd1) mice at post-natal (PN) day 15, and 60% in Tg(RHO P347S) mice at PN40. In contrast, PR survival was not increased in retinal explants of Pde6b(rd1/rd1) ; Edn2(-/-) mice. This finding, together with systemic abnormalities in Edn2(-/-) mice, suggested that the increased survival of mutant PRs in the Edn2(-/-) background resulted at least partly from the systemic EDN2 loss of function. To examine directly the role of EDN2 in mutant PRs, we used a scAAV5-Edn2 cDNA vector to restore Edn2 expression in Pde6b(rd1/rd1) ; Edn2(-/-) PRs and observed an 18% increase in PR survival at PN14. Importantly, PR survival was also increased after injection of scAAV5-Edn2 into Pde6b(rd1/rd1) retinas, by 31% at PN15. Together, these findings suggest that increased Edn2 expression is protective to mutant PRs. To begin to elucidate Edn2-mediated mechanisms that contribute to PR survival, we used microarray analysis and identified a cohort of 20 genes with >4-fold increased expression in Tg(RHO P347S) retinas, including Fgf2. Notably, increased expression of the FGF2 protein in Tg(RHO P347S) PRs was ablated in Tg(RHO P347S); Edn2(-/-) retinas. Our findings indicate that the increased expression of PR Edn2 increases PR survival, and suggest that the Edn2-dependent increase in PR expression of FGF2 may contribute to the augmented survival.


Subject(s)
Endothelin-2/metabolism , Mutation , Photoreceptor Cells, Vertebrate/metabolism , Photoreceptor Cells, Vertebrate/pathology , Retinal Diseases/genetics , Retinal Diseases/metabolism , Animals , Cell Hypoxia/genetics , Cell Survival/genetics , Cyclic Nucleotide Phosphodiesterases, Type 6/metabolism , Endothelin-2/genetics , Fibroblast Growth Factor 2/metabolism , Humans , Mice , Mice, Transgenic , RNA, Messenger/genetics , RNA, Messenger/metabolism , Retina/metabolism , Retina/pathology , Retinal Diseases/pathology , Rhodopsin/genetics , Signal Transduction/genetics , Up-Regulation/genetics
20.
Br J Pharmacol ; 169(1): 179-96, 2013 May.
Article in English | MEDLINE | ID: mdl-23425137

ABSTRACT

BACKGROUND AND PURPOSE: Ectonucleotidases control extracellular nucleotide levels and consequently, their (patho)physiological responses. Among these enzymes, nucleoside triphosphate diphosphohydrolase-1 (NTPDase1), -2, -3 and -8 are the major ectonucleotidases responsible for nucleotide hydrolysis at the cell surface under physiological conditions, and NTPDase1 is predominantly located at the surface of vascular endothelial cells and leukocytes. Efficacious inhibitors of NTPDase1 are required to modulate responses induced by nucleotides in a number of pathological situations such as thrombosis, inflammation and cancer. EXPERIMENTAL APPROACH: Here, we present the synthesis and enzymatic characterization of five 8-BuS-adenine nucleotide derivatives as potent and selective inhibitors of NTPDase1. KEY RESULTS: The compounds 8-BuS-AMP, 8-BuS-ADP and 8-BuS-ATP inhibit recombinant human and mouse NTPDase1 by mixed type inhibition, predominantly competitive with Ki values <1 µM. In contrast to 8-BuS-ATP which could be hydrolyzed by other NTPDases, the other BuS derivatives were resistant to hydrolysis by either NTPDase1, -2, -3 or -8. 8-BuS-AMP and 8-BuS-ADP were the most potent and selective inhibitors of NTPDase1 expressed in human umbilical vein endothelial cells as well as in situ in human and mouse tissues. As expected, as a result of their inhibition of recombinant human NTPDase1, 8-BuS-AMP and 8-BuS-ADP impaired the ability of this enzyme to block platelet aggregation. Importantly, neither of these two inhibitors triggered platelet aggregation nor prevented ADP-induced platelet aggregation, in support of their inactivity towards P2Y1 and P2Y12 receptors. CONCLUSIONS AND IMPLICATIONS: The 8-BuS-AMP and 8-BuS-ADP have therefore potential to serve as drugs for the treatment of pathologies regulated by NTPDase1.


Subject(s)
Adenosine Diphosphate/pharmacology , Adenosine Monophosphate/pharmacology , Adenosine Triphosphate/pharmacology , Apyrase/antagonists & inhibitors , Adenosine Diphosphate/analogs & derivatives , Adenosine Diphosphate/chemical synthesis , Adenosine Monophosphate/analogs & derivatives , Adenosine Monophosphate/chemical synthesis , Adenosine Triphosphate/analogs & derivatives , Adenosine Triphosphate/chemical synthesis , Animals , Antigens, CD , COS Cells , Chlorocebus aethiops , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Human Umbilical Vein Endothelial Cells , Humans , Mice , Platelet Aggregation/drug effects , Rats , Species Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...