Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 17 de 17
2.
JCI Insight ; 8(10)2023 05 22.
Article En | MEDLINE | ID: mdl-37022786

To improve our limited understanding of the pathogenesis of thoracic aortic aneurysm (TAA) that leads to acute aortic dissection, single-cell RNA sequencing (scRNA-seq) was employed to profile disease-relevant transcriptomic changes of aortic cell populations in a well-characterized mouse model of the most commonly diagnosed form of Marfan syndrome (MFS). As result, 2 discrete subpopulations of aortic cells (SMC3 and EC4) were identified only in the aorta of Fbn1mgR/mgR mice. SMC3 cells highly express genes related to extracellular matrix formation and nitric oxide signaling, whereas the EC4 transcriptional profile is enriched in smooth muscle cell (SMC), fibroblast, and immune cell-related genes. Trajectory analysis predicted close phenotypic modulation between SMC3 and EC4, which were therefore analyzed together as a discrete MFS-modulated (MFSmod) subpopulation. In situ hybridization of diagnostic transcripts located MFSmod cells at the intima of Fbn1mgR/mgR aortas. Reference-based data set integration revealed transcriptomic similarity between MFSmod- and SMC-derived cell clusters modulated in human TAA. Consistent with the angiotensin II type I receptor (At1r) contribution to TAA development, MFSmod cells were absent in the aorta of Fbn1mgR/mgR mice treated with the At1r antagonist losartan. Altogether, our findings indicate that a discrete dynamic alteration of aortic cell identity is associated with dissecting TAA in MFS mice and increased risk of aortic dissection in MFS patients.


Aortic Aneurysm, Thoracic , Aortic Aneurysm , Aortic Dissection , Marfan Syndrome , Humans , Mice , Animals , Transcriptome , Losartan/pharmacology , Marfan Syndrome/complications , Marfan Syndrome/genetics , Marfan Syndrome/pathology , Aortic Aneurysm/genetics , Aortic Aneurysm, Thoracic/genetics , Aortic Aneurysm, Thoracic/pathology , Aorta/pathology , Aortic Dissection/genetics
3.
Hum Mol Genet ; 31(19): 3281-3289, 2022 09 29.
Article En | MEDLINE | ID: mdl-35567544

A disproportionate tall stature is the most evident manifestation in Marfan syndrome (MFS), a multisystem condition caused by mutations in the extracellular protein and TGFß modulator, fibrillin-1. Unlike cardiovascular manifestations, there has been little effort devoted to unravel the molecular mechanism responsible for long bone overgrowth in MFS. By combining the Cre-LoxP recombination system with metatarsal bone cultures, here we identify the outer layer of the perichondrium as the tissue responsible for long bone overgrowth in MFS mice. Analyses of differentially expressed genes in the fibrillin-1-deficient perichondrium predicted that loss of TGFß signaling may influence chondrogenesis in the neighboring epiphyseal growth plate (GP). Immunohistochemistry revealed that fibrillin-1 deficiency in the outer perichondrium is associated with decreased accumulation of latent TGFß-binding proteins (LTBPs)-3 and -4, and reduced levels of phosphorylated (activated) Smad2. Consistent with these findings, mutant metatarsal bones grown in vitro were longer and released less TGFß than the wild-type counterparts. Moreover, addition of recombinant TGFß1 normalized linear growth of mutant metatarsal bones. We conclude that longitudinal bone overgrowth in MFS is accounted for by diminished sequestration of LTBP-3 and LTBP-4 into the fibrillin-1-deficient matrix of the outer perichondrium, which results in less TGFß signaling locally and improper GP differentiation distally.


Marfan Syndrome , Animals , Fibrillin-1/genetics , Fibrillin-2 , Fibrillins , Latent TGF-beta Binding Proteins/genetics , Latent TGF-beta Binding Proteins/metabolism , Marfan Syndrome/genetics , Mice , Microfilament Proteins/genetics , Microfilament Proteins/metabolism , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism
4.
Int J Mol Sci ; 23(3)2022 Feb 08.
Article En | MEDLINE | ID: mdl-35163812

Fibrillin-1 is the major structural component of the 10 nm-diameter microfibrils that confer key physical and mechanical properties to virtually every tissue, alone and together with elastin in the elastic fibers. Mutations in fibrillin-1 cause pleiotropic manifestations in Marfan syndrome (MFS), including dissecting thoracic aortic aneurysms, myocardial dysfunction, progressive bone loss, disproportionate skeletal growth, and the dislocation of the crystalline lens. The characterization of these MFS manifestations in mice, that replicate the human phenotype, have revealed that the underlying mechanisms are distinct and organ-specific. This brief review summarizes relevant findings supporting this conclusion.


Fibrillin-1/genetics , Marfan Syndrome/pathology , Animals , Disease Models, Animal , Humans , Marfan Syndrome/genetics , Mice , Mutation , Organ Specificity
5.
Biomolecules ; 12(1)2022 01 14.
Article En | MEDLINE | ID: mdl-35053276

About 20% of individuals afflicted with thoracic aortic disease have single-gene mutations that predispose the vessel to aneurysm formation and/or acute aortic dissection often without associated syndromic features. One widely studied exception is Marfan syndrome (MFS) in which mutations in the extracellular protein fibrillin-1 cause additional abnormalities in the heart, eyes, and skeleton. Mouse models of MFS have been instrumental in delineating major cellular and molecular determinants of thoracic aortic disease. In spite of research efforts, translating experimental findings from MFS mice into effective drug therapies for MFS patients remains an unfulfilled promise. Here, we describe a series of studies that have implicated endothelial dysfunction and improper angiotensin II and TGFß signaling in driving thoracic aortic disease in MFS mice. We also discuss how these investigations have influenced the way we conceptualized possible new therapies to slow down or even halt aneurysm progression in this relatively common connective tissue disorder.


Aortic Aneurysm, Thoracic , Aortic Dissection , Marfan Syndrome , Aortic Dissection/genetics , Angiotensin II , Animals , Aorta/metabolism , Aortic Aneurysm, Thoracic/genetics , Aortic Aneurysm, Thoracic/metabolism , Humans , Marfan Syndrome/complications , Marfan Syndrome/drug therapy , Marfan Syndrome/genetics , Mice
6.
Hypertension ; 75(5): 1279-1288, 2020 05.
Article En | MEDLINE | ID: mdl-32172624

Ceramides are sphingolipids that modulate a variety of cellular processes via 2 major mechanisms: functioning as second messengers and regulating membrane biophysical properties, particularly lipid rafts, important signaling platforms. Altered sphingolipid levels have been implicated in many cardiovascular diseases, including hypertension, atherosclerosis, and diabetes mellitus-related conditions; however, molecular mechanisms by which ceramides impact endothelial functions remain poorly understood. In this regard, we generated mice defective of endothelial sphingolipid de novo biosynthesis by deleting the Sptlc2 (long chain subunit 2 of serine palmitoyltransferase)-the first enzyme of the pathway. Our study demonstrated that endothelial sphingolipid de novo production is necessary to regulate (1) signal transduction in response to NO agonists and, mainly via ceramides, (2) resting eNOS (endothelial NO synthase) phosphorylation, and (3) blood pressure homeostasis. Specifically, our findings suggest a prevailing role of C16:0-Cer in preserving vasodilation induced by tyrosine kinase and GPCRs (G-protein coupled receptors), except for Gq-coupled receptors, while C24:0- and C24:1-Cer control flow-induced vasodilation. Replenishing C16:0-Cer in vitro and in vivo reinstates endothelial cell signaling and vascular tone regulation. This study reveals an important role of locally produced ceramides, particularly C16:0-, C24:0-, and C24:1-Cer in vascular and blood pressure homeostasis, and establishes the endothelium as a key source of plasma ceramides. Clinically, specific plasma ceramides ratios are independent predictors of major cardiovascular events. Our data also suggest that plasma ceramides might be indicative of the diseased state of the endothelium.


Blood Pressure/physiology , Ceramides/physiology , Endothelial Cells/metabolism , Nitric Oxide/physiology , Signal Transduction , Sphingolipids/biosynthesis , Acetylcholine/pharmacology , Animals , Cell Adhesion Molecules/metabolism , Cells, Cultured , Histamine/pharmacology , Homeostasis , Male , Mice , Mice, Transgenic , Microfilament Proteins/metabolism , Nitric Oxide/agonists , Nitric Oxide Synthase Type III/metabolism , Nitroprusside/pharmacology , Phosphoproteins/metabolism , Serine C-Palmitoyltransferase/deficiency , Vascular Endothelial Growth Factor Receptor-2/physiology , Vasoconstriction/drug effects , Vasoconstriction/physiology , Vasodilation/drug effects , Vasodilation/physiology
7.
Sci Signal ; 10(492)2017 Aug 15.
Article En | MEDLINE | ID: mdl-28811382

Endothelial dysfunction, a hallmark of vascular disease, is restored by plasma high-density lipoprotein (HDL). However, a generalized increase in HDL abundance is not beneficial, suggesting that specific HDL species mediate protective effects. Apolipoprotein M-containing HDL (ApoM+HDL), which carries the bioactive lipid sphingosine 1-phosphate (S1P), promotes endothelial function by activating G protein-coupled S1P receptors. Moreover, HDL-bound S1P is limiting in several inflammatory, metabolic, and vascular diseases. We report the development of a soluble carrier for S1P, ApoM-Fc, which activated S1P receptors in a sustained manner and promoted endothelial function. In contrast, ApoM-Fc did not modulate circulating lymphocyte numbers, suggesting that it specifically activated endothelial S1P receptors. ApoM-Fc administration reduced blood pressure in hypertensive mice, attenuated myocardial damage after ischemia/reperfusion injury, and reduced brain infarct volume in the middle cerebral artery occlusion model of stroke. Our proof-of-concept study suggests that selective and sustained targeting of endothelial S1P receptors by ApoM-Fc could be a viable therapeutic strategy in vascular diseases.


Endothelium, Vascular/drug effects , Hypertension/prevention & control , Lysophospholipids/pharmacology , Receptors, Lysosphingolipid/metabolism , Reperfusion Injury/prevention & control , Sphingosine/analogs & derivatives , Animals , Apolipoproteins M/metabolism , Endothelium, Vascular/metabolism , Human Umbilical Vein Endothelial Cells , Humans , Hypertension/metabolism , Hypertension/pathology , Lipoproteins, HDL/metabolism , Male , Mice , Mice, Knockout , Protein Binding , Receptors, Fc/metabolism , Reperfusion Injury/metabolism , Reperfusion Injury/pathology , Signal Transduction/drug effects , Sphingosine/pharmacology
8.
Hypertension ; 70(2): 426-434, 2017 08.
Article En | MEDLINE | ID: mdl-28607130

Nitric oxide is one of the major endothelial-derived vasoactive factors that regulate blood pressure (BP), and the bioactive lipid mediator S1P (sphingosine-1-phosphate) is a potent activator of endothelial nitric oxide synthase through G protein-coupled receptors. Endothelial-derived S1P and the autocrine/paracrine activation of S1PR (S1P receptors) play an important role in preserving vascular functions and BP homeostasis. Furthermore, FTY720 (fingolimod), binding to 4 out of 5 S1PRs recently approved by the Food and Drug Administration to treat autoimmune conditions, induces a modest and transient decrease in heart rate in both animals and humans, suggesting that drugs targeting sphingolipid signaling affect cardiovascular functions in vivo. However, the role of specific S1P receptors in BP homeostasis remains unknown. The aim of this study is to determine the role of the key vascular S1P receptors, namely, S1PR1 and S1PR3, in BP regulation in physiological and hypertensive conditions. The specific loss of endothelial S1PR1 decreases basal and stimulated endothelial-derived nitric oxide and resets BP to a higher-than-normal value. Interestingly, we identified a novel and important role for S1PR1 signaling in flow-mediated mechanotransduction. FTY720, acting as functional antagonist of S1PR1, markedly decreases endothelial S1PR1, increases BP in control mice, and exacerbates hypertension in angiotensin II mouse model, underlining the antihypertensive functions of S1PR1 signaling. Our study identifies S1P-S1PR1-nitric oxide signaling as a new regulatory pathway in vivo of vascular relaxation to flow and BP homeostasis, providing a novel therapeutic target for the treatment of hypertension.


Blood Pressure/physiology , Fingolimod Hydrochloride/pharmacology , Hypertension , Nitric Oxide Synthase Type III/metabolism , Nitric Oxide/metabolism , Receptors, Lysosphingolipid , Animals , Blood Pressure/drug effects , Disease Models, Animal , Endothelial Cells/metabolism , Hypertension/drug therapy , Hypertension/metabolism , Hypertension/physiopathology , Immunosuppressive Agents/pharmacology , Mice , Receptors, Lysosphingolipid/antagonists & inhibitors , Receptors, Lysosphingolipid/metabolism , Signal Transduction/drug effects , Signal Transduction/physiology , Sphingosine-1-Phosphate Receptors
9.
J Pharmacol Exp Ther ; 358(2): 359-70, 2016 08.
Article En | MEDLINE | ID: mdl-27317800

Initially discovered as abundant components of eukaryotic cell membranes, sphingolipids are now recognized as important bioactive signaling molecules that modulate a variety of cellular functions, including those relevant to cancer and immunologic, inflammatory, and cardiovascular disorders. In this review, we discuss recent advances in our understanding of the role of sphingosine-1-phosphate (S1P) receptors in the regulation of vascular function, and focus on how de novo biosynthesized sphingolipids play a role in blood pressure homeostasis. The therapeutic potential of new drugs that target S1P signaling is also discussed.


Blood Pressure , Homeostasis , Lysophospholipids/biosynthesis , Lysophospholipids/metabolism , Signal Transduction , Sphingosine/analogs & derivatives , Animals , Endothelium, Vascular/metabolism , Humans , Sphingosine/biosynthesis , Sphingosine/metabolism , Vasodilation
10.
JCI Insight ; 1(5)2016 Apr 21.
Article En | MEDLINE | ID: mdl-27158676

We recently discovered that endothelial Nogo-B, a membrane protein of the ER, regulates vascular function by inhibiting the rate-limiting enzyme, serine palmitoyltransferase (SPT), in de novo sphingolipid biosynthesis. Here, we show that endothelium-derived sphingolipids, particularly sphingosine-1-phosphate (S1P), protect the heart from inflammation, fibrosis, and dysfunction following pressure overload and that Nogo-B regulates this paracrine process. SPT activity is upregulated in banded hearts in vivo as well as in TNF-α-activated endothelium in vitro, and loss of Nogo removes the brake on SPT, increasing local S1P production. Hence, mice lacking Nogo-B, systemically or specifically in the endothelium, are resistant to the onset of pathological cardiac hypertrophy. Furthermore, pharmacological inhibition of SPT with myriocin restores permeability, inflammation, and heart dysfunction in Nogo-A/B-deficient mice to WT levels, whereas SEW2871, an S1P1 receptor agonist, prevents myocardial permeability, inflammation, and dysfunction in WT banded mice. Our study identifies a critical role of endothelial sphingolipid biosynthesis and its regulation by Nogo-B in the development of pathological cardiac hypertrophy and proposes a potential therapeutic target for the attenuation or reversal of this clinical condition.

11.
Clin Sci (Lond) ; 130(1): 35-44, 2016 Jan.
Article En | MEDLINE | ID: mdl-26460077

Hydrogen sulfide is an endogenous gasotransmitter and its mechanism of action involves activation of ATP-sensitive K(+) channels and phosphodiesterase inhibition. As both mechanisms are potentially involved in malignant hyperthermia (MH), in the present study we addressed the involvement of the L-cysteine/hydrogen sulfide pathway in MH. Skeletal muscle biopsies obtained from 25 MH-susceptible (MHS) and 56 MH-negative (MHN) individuals have been used to perform the in vitro contracture test (IVCT). Quantitative real-time PCR (qPCR) and Western blotting studies have also been performed. Hydrogen sulfide levels are measured in both tissue samples and plasma. In MHS biopsies an increase in cystathionine ß-synthase (CBS) occurs, as both mRNA and protein expression compared with MHN biopsies. Hydrogen sulfide biosynthesis is increased in MHS biopsies (0.128±0.12 compared with 0.943±0.13 nmol/mg of protein per min for MHN and MHS biopsies, respectively; P<0.01). Addition of sodium hydrosulfide (NaHS) to MHS samples evokes a response similar, in the IVCT, to that elicited by either caffeine or halothane. Incubation of MHN biopsies with NaHS, before caffeine or halothane challenge, switches an MHN to an MHS response. In conclusion we demonstrate the involvement of the L-cysteine/hydrogen sulfide pathway in MH, giving new insight into MH molecular mechanisms. This finding has potential implications for clinical care and could help to define less invasive diagnostic procedures.


Cystathionine beta-Synthase/metabolism , Gasotransmitters/metabolism , Hydrogen Sulfide/metabolism , Malignant Hyperthermia/enzymology , Muscle, Skeletal/enzymology , Biopsy , Caffeine/pharmacology , Case-Control Studies , Cystathionine beta-Synthase/genetics , Dose-Response Relationship, Drug , Gene Expression Regulation, Enzymologic , Glyburide/pharmacology , Halothane/pharmacology , Humans , In Vitro Techniques , KATP Channels/antagonists & inhibitors , KATP Channels/metabolism , Malignant Hyperthermia/genetics , Malignant Hyperthermia/physiopathology , Muscle Contraction , Muscle, Skeletal/drug effects , Muscle, Skeletal/physiopathology , Potassium Channel Blockers/pharmacology , RNA, Messenger/metabolism , Signal Transduction , Sulfides/metabolism , Sulfides/pharmacology , Up-Regulation
12.
Nat Med ; 21(9): 1028-1037, 2015 Sep.
Article En | MEDLINE | ID: mdl-26301690

Endothelial dysfunction is a critical factor in many cardiovascular diseases, including hypertension. Although lipid signaling has been implicated in endothelial dysfunction and cardiovascular disease, specific molecular mechanisms are poorly understood. Here we report that Nogo-B, a membrane protein of the endoplasmic reticulum, regulates endothelial sphingolipid biosynthesis with direct effects on vascular function and blood pressure. Nogo-B inhibits serine palmitoyltransferase, the rate-limiting enzyme of the de novo sphingolipid biosynthetic pathway, thereby controlling production of endothelial sphingosine 1-phosphate and autocrine, G protein-coupled receptor-dependent signaling by this metabolite. Mice lacking Nogo-B either systemically or specifically in endothelial cells are hypotensive, resistant to angiotensin II-induced hypertension and have preserved endothelial function and nitric oxide release. In mice that lack Nogo-B, pharmacological inhibition of serine palmitoyltransferase with myriocin reinstates endothelial dysfunction and angiotensin II-induced hypertension. Our study identifies Nogo-B as a key inhibitor of local sphingolipid synthesis and shows that autocrine sphingolipid signaling within the endothelium is critical for vascular function and blood pressure homeostasis.


Blood Pressure , Endothelium, Vascular/physiology , Homeostasis , Myelin Proteins/physiology , Sphingolipids/metabolism , Animals , COS Cells , Chlorocebus aethiops , HEK293 Cells , Humans , Lysophospholipids/physiology , Male , Mice , Nitric Oxide Synthase Type III/physiology , Nogo Proteins , Receptors, Lysosphingolipid/physiology , Sphingosine/analogs & derivatives , Sphingosine/physiology
13.
Cardiovasc Res ; 102(1): 138-47, 2014 Apr 01.
Article En | MEDLINE | ID: mdl-24501330

AIMS: Therapeutic use of sulfhydrylated inhibitor S-zofenopril has raised different hypotheses regarding the role played by its thiol group in the beneficial clinical effects exerted compared with other angiotensin-converting enzyme (ACE) inhibitors. Here, we investigated hydrogen sulfide (H2S) pathway as accountable for extra-beneficial effects in vascular function. METHODS AND RESULTS: Spontaneously hypertensive rat (SHRs) and control Wistar Kyoto (WKY) rats were treated with either S-zofenopril or enalapril in vivo. Aorta and carotid were harvested and ex vivo vascular reactivity to acetylcholine (Ach) and L-cysteine (L-cys) assessed. Cystathionine-ß-synthase (CBS), cystathionine-γ-lyase (CSE), and 3-mercaptosulfur-transferase (3MST) expression, as well as H2S levels, were evaluated in both vascular tissues. The vascular response to Ach in both carotid and aorta was impaired in SHR (~30%, P < 0.001). S-zofenopril, but not enalapril, restored this response, while L-cys-induced relaxation was enhanced. CSE expression in vessels and tissue/plasma H2S levels were restored to WKY values in SHRs receiving S-zofenopril. In contrast, CBS and 3MST expression were not modified by treatments. S-zofenoprilat, an active metabolite of S-zofenopril, releases H2S in a 'cell-free' assay and it directly relaxed vessels in vitro in a concentration-dependent manner (P < 0.001). In vivo administration of R-zofenoprilat diasteroisomer, which does not inhibit ACE, did not modify blood pressure; nonetheless, it retained the beneficial effect on SHR vascular function as well as restored plasma/tissue H2S levels. CONCLUSION: Our findings establish that S-zofenopril improves vascular function by potentiating the H2S pathway in a model of spontaneous hypertension. This novel mechanism, unrelated to ACE inhibition and based on H2S release, could explain the beneficial effects of sulfhydrylated ACE inhibitors reported in the clinical literature.


Angiotensin-Converting Enzyme Inhibitors/pharmacology , Aorta/drug effects , Blood Pressure/drug effects , Captopril/analogs & derivatives , Hydrogen Sulfide/pharmacology , Animals , Captopril/chemistry , Captopril/pharmacology , Cystathionine gamma-Lyase/metabolism , Hydrogen Sulfide/metabolism , Male , Rats, Inbred SHR , Rats, Inbred WKY
14.
Hypertension ; 62(3): 634-40, 2013 Sep.
Article En | MEDLINE | ID: mdl-23918749

Hypertension is a major worldwide health issue for which only a small proportion of cases have a known mechanistic pathogenesis. Of the defined causes, none have been directly linked to heightened vasoconstrictor responsiveness, despite the fact that vasomotor tone in resistance vessels is a fundamental determinant of blood pressure. Here, we reported a previously undescribed role for smooth muscle hypoxia-inducible factor-1α (HIF-1α) in controlling blood pressure homeostasis. The lack of HIF-1α in smooth muscle caused hypertension in vivo and hyperresponsiveness of resistance vessels to angiotensin II stimulation ex vivo. These data correlated with an increased expression of angiotensin II receptor type I in the vasculature. Specifically, we show that HIF-1α, through peroxisome proliferator-activated receptor-γ, reciprocally defined angiotensin II receptor type I levels in the vessel wall. Indeed, pharmacological blockade of angiotensin II receptor type I by telmisartan abolished the hypertensive phenotype in smooth muscle cell-HIF-1α-KO mice. These data revealed a determinant role of a smooth muscle HIF-1α/peroxisome proliferator-activated receptor-γ/angiotensin II receptor type I axis in controlling vasomotor responsiveness and highlighted an important pathway, the alterations of which may be critical in a variety of hypertensive-based clinical settings.


Blood Pressure/physiology , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Muscle, Smooth, Vascular/metabolism , PPAR gamma/metabolism , Receptor, Angiotensin, Type 1/metabolism , Signal Transduction/physiology , Angiotensin II/pharmacology , Angiotensin II Type 1 Receptor Blockers/pharmacology , Animals , Benzimidazoles/pharmacology , Benzoates/pharmacology , Blood Pressure/drug effects , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Mesenteric Arteries/drug effects , Mesenteric Arteries/metabolism , Mice , Mice, Knockout , Muscle, Smooth, Vascular/drug effects , PPAR gamma/genetics , Receptor, Angiotensin, Type 1/genetics , Signal Transduction/drug effects , Telmisartan
15.
PLoS One ; 8(3): e57801, 2013.
Article En | MEDLINE | ID: mdl-23554869

Here we have characterized perthamide C, a cyclopeptide from a Solomon Lithistid sponge Theonella swinhoei, which displays an anti-inflammatory/immunomodulatory activity. The study has been performed using the carragenan-induced mouse paw edema that displays an early (0-6 h) and a late phase (24-96 h). Perthamide C significantly inhibits neutrophils infiltration in tissue both in the early and late phases. This effect was coupled to a reduced expression of the endothelial nitric oxide synthase (eNOS) in the early phase while cyclooxygenase-1 and 2 (COX-1, COX-2), and inducible NOS (iNOS) expression were unaffected. In the late phase perthamide C reduced expression of both NOS isoforms without affecting COXs expression. This peculiar selectivity toward the two enzymes deputed to produce NO lead us to investigate on a possible action of perthamide C on lymphocytes infiltration and activation. We found that perthamide C inhibited the proliferation of peripheral lymphocytes, and that this effect was secondary to its metabolic activation in vivo. Indeed, in vitro perthamide C did not inhibit proliferation as opposite to its metabolite perthamide H. In conclusion, perthamide C selectively interferes with NO generation triggered by either eNOS or iNOS without affecting either COX-1 or COX-2. This in turn leads to modulation of the inflammatory response through a reduction of vascular permeability, neutrophil infiltration as well as lymphocyte proliferation.


Gene Expression Regulation, Enzymologic/drug effects , Immunologic Factors/pharmacology , Nitric Oxide Synthase Type III/biosynthesis , Nitric Oxide Synthase Type II/biosynthesis , Nitric Oxide/biosynthesis , Peptides, Cyclic/pharmacology , Animals , Capillary Permeability/drug effects , Capillary Permeability/immunology , Cell Proliferation/drug effects , Cyclooxygenase 1/biosynthesis , Cyclooxygenase 1/immunology , Cyclooxygenase 2/biosynthesis , Cyclooxygenase 2/immunology , Disease Models, Animal , Edema/drug therapy , Edema/enzymology , Edema/immunology , Edema/pathology , Gene Expression Regulation, Enzymologic/immunology , Lymphocytes/enzymology , Lymphocytes/immunology , Lymphocytes/pathology , Male , Membrane Proteins/biosynthesis , Membrane Proteins/immunology , Mice , Neutrophil Infiltration/drug effects , Neutrophil Infiltration/immunology , Nitric Oxide/immunology , Nitric Oxide Synthase Type II/immunology , Nitric Oxide Synthase Type III/immunology
16.
Pflugers Arch ; 465(4): 509-16, 2013 Apr.
Article En | MEDLINE | ID: mdl-23334408

Functional ß3-adrenoceptors have been found in skeletal muscle where they mediate metabolic oxidation and glucose utilization. Whether ß3-adrenoceptors (ARs) also play any role in muscle protein metabolism still remains uncertain. By using rat L6 myocyte cultures, we found that CL316,243, a ß3-AR selective agonist, at the concentration of 10(-6) M for 24 h, induced a significant increase of skeletal muscle constitutive proteins such as H- and L-myosin and ß-actin. Such effect was correlated to an increased expression of phosphorylated p70(S6K) that was significantly inhibited by ß3-AR antagonist, SR 59230A, but not by ß2-AR antagonist, ICI-118,551. The CL316,243-induced activation of p70(S6K) was markedly inhibited by wortmannin, a PI3K inhibitor, and rapamycin, a specific inhibitor of mTOR, suggesting a critical involvement of the PI3K-mTOR-p70(S6K) signaling cascade in the anabolic response of L6 cells to ß3-AR agonist. Taken together, these results suggest that stimulation of ß3-AR in skeletal muscle cells activates a specific signaling pathway leading to protein synthesis and, eventually, muscle growth.


Adrenergic beta-3 Receptor Agonists/pharmacology , Dioxoles/pharmacology , Muscle Fibers, Skeletal/metabolism , Protein Biosynthesis/drug effects , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/metabolism , Actins/genetics , Actins/metabolism , Adrenergic beta-Antagonists/pharmacology , Androstadienes/pharmacology , Animals , Cells, Cultured , Gene Expression/drug effects , Muscle Fibers, Skeletal/drug effects , Muscle Fibers, Skeletal/enzymology , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Phosphorylation , Propanolamines/pharmacology , Rats , Ribosomal Protein S6 Kinases, 70-kDa/genetics , Sirolimus/pharmacology , Skeletal Muscle Myosins/genetics , Skeletal Muscle Myosins/metabolism , TOR Serine-Threonine Kinases/antagonists & inhibitors , TOR Serine-Threonine Kinases/genetics , Wortmannin
17.
PLoS One ; 7(12): e53319, 2012.
Article En | MEDLINE | ID: mdl-23285278

A growing body of evidence suggests that hydrogen sulfide (H2S) is a signaling molecule in mammalian cells. In the cardiovascular system, H2S enhances vasodilation and angiogenesis. H2S-induced vasodilation is hypothesized to occur through ATP-sensitive potassium channels (K(ATP)); however, we recently demonstrated that it also increases cGMP levels in tissues. Herein, we studied the involvement of cGMP-dependent protein kinase-I in H2S-induced vasorelaxation. The effect of H2S on vessel tone was studied in phenylephrine-contracted aortic rings with or without endothelium. cGMP levels were determined in cultured cells or isolated vessel by enzyme immunoassay. Pretreatment of aortic rings with sildenafil attenuated NaHS-induced relaxation, confirming previous findings that H2S is a phosphodiesterase inhibitor. In addition, vascular tissue levels of cGMP in cystathionine gamma lyase knockouts were lower than those in wild-type control mice. Treatment of aortic rings with NaHS, a fast releasing H2S donor, enhanced phosphorylation of vasodilator-stimulated phosphoprotein in a time-dependent manner, suggesting that cGMP-dependent protein kinase (PKG) is activated after exposure to H2S. Incubation of aortic rings with a PKG-I inhibitor (DT-2) attenuated NaHS-stimulated relaxation. Interestingly, vasodilatory responses to a slowly releasing H2S donor (GYY 4137) were unaffected by DT-2, suggesting that this donor dilates mouse aorta through PKG-independent pathways. Dilatory responses to NaHS and L-cysteine (a substrate for H2S production) were reduced in vessels of PKG-I knockout mice (PKG-I⁻/⁻). Moreover, glibenclamide inhibited NaHS-induced vasorelaxation in vessels from wild-type animals, but not PKG-I⁻/⁻, suggesting that there is a cross-talk between K(ATP) and PKG. Our results confirm the role of cGMP in the vascular responses to NaHS and demonstrate that genetic deletion of PKG-I attenuates NaHS and L-cysteine-stimulated vasodilation.


Cyclic GMP-Dependent Protein Kinases/physiology , Hydrogen Sulfide/pharmacology , Vasodilation/drug effects , Animals , Aorta/drug effects , Aorta/physiology , Cells, Cultured , Cyclic GMP-Dependent Protein Kinases/antagonists & inhibitors , Cyclic GMP-Dependent Protein Kinases/genetics , Cyclic GMP-Dependent Protein Kinases/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 5/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 5/physiology , Endothelium, Vascular/drug effects , Endothelium, Vascular/physiology , Female , Male , Mice , Mice, Knockout , Phosphodiesterase 5 Inhibitors/pharmacology , Rats , Rats, Wistar , Vasodilation/genetics
...