Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 59
Filter
1.
Arterioscler Thromb Vasc Biol ; 44(1): e1-e18, 2024 01.
Article in English | MEDLINE | ID: mdl-38031839

ABSTRACT

BACKGROUND: Heart failure with preserved ejection fraction is proposed to be caused by endothelial dysfunction in cardiac microvessels. Our goal was to identify molecular and cellular mechanisms underlying the development of cardiac microvessel disease and diastolic dysfunction in the setting of type 2 diabetes. METHODS: We used Leprdb/db (leptin receptor-deficient) female mice as a model of type 2 diabetes and heart failure with preserved ejection fraction and identified Hhipl1 (hedgehog interacting protein-like 1), which encodes for a decoy receptor for HH (hedgehog) ligands as a gene upregulated in the cardiac vascular fraction of diseased mice. RESULTS: We then used Dhh (desert HH)-deficient mice to investigate the functional consequences of impaired HH signaling in the adult heart. We found that Dhh-deficient mice displayed increased end-diastolic pressure while left ventricular ejection fraction was comparable to that of control mice. This phenotype was associated with a reduced exercise tolerance in the treadmill test, suggesting that Dhh-deficient mice do present heart failure. At molecular and cellular levels, impaired cardiac relaxation in DhhECKO mice was associated with a significantly decreased PLN (phospholamban) phosphorylation on Thr17 (threonine 17) and an alteration of sarcomeric shortening ex vivo. Besides, as expected, Dhh-deficient mice exhibited phenotypic changes in their cardiac microvessels including a prominent prothrombotic phenotype. Importantly, aspirin therapy prevented the occurrence of both diastolic dysfunction and exercise intolerance in these mice. To confirm the critical role of thrombosis in the pathophysiology of diastolic dysfunction, we verified Leprdb/db also displays increased cardiac microvessel thrombosis. Moreover, consistently, with Dhh-deficient mice, we found that aspirin treatment decreased end-diastolic pressure and improved exercise tolerance in Leprdb/db mice. CONCLUSIONS: Altogether, these results demonstrate that microvessel thrombosis may participate in the pathophysiology of heart failure with preserved ejection fraction.


Subject(s)
Cardiomyopathies , Diabetes Mellitus, Type 2 , Heart Failure , Thrombosis , Ventricular Dysfunction, Left , Animals , Female , Mice , Ventricular Function, Left , Stroke Volume , Diabetes Mellitus, Type 2/complications , Ventricular Dysfunction, Left/genetics , Hedgehog Proteins , Microvessels , Thrombosis/complications , Aspirin
2.
J Am Heart Assoc ; 12(13): e029279, 2023 07 04.
Article in English | MEDLINE | ID: mdl-37345826

ABSTRACT

Background Although the critical role of pericytes in maintaining vascular integrity has been extensively demonstrated in the brain and the retina, little is known about their role in the heart. We aim to investigate structural and functional consequences of partial pericyte depletion (≈60%) in the heart of adult mice. Methods and Results To deplete pericytes in adult mice, we used platelet-derived growth factor receptor ß-Cre/ERT2; RosaDTA mice and compared their phenotype with that of control mice (RosaDTA) chosen among their littermates. Cardiac function was assessed via echocardiography and left ventricular catheterization 1 month after the first tamoxifen injection. We found mice depleted with pericytes had a reduced left ventricular ejection fraction and an increased end-diastolic pressure, demonstrating both systolic and diastolic dysfunction. Consistently, mice depleted with pericytes presented a decreased left ventricular contractility and an increased left ventricular relaxation time (dP/dtmin). At the tissue level, mice depleted of pericytes displayed increased coronary endothelium leakage and activation, which was associated with increased CD45+ cell infiltration. Consistent with systolic dysfunction, pericyte depletion was associated with an increased expression of myosin heavy chain 7 and decreased expression of ATPase sarcoplasmic/endoplasmic reticulum Ca2+ transporting 2 and connexin 43. More important, coculture assays demonstrated, for the first time, that the decreased expression of connexin 43 is likely attributable to a direct effect of pericytes on cardiomyocytes. Besides, this study reveals that cardiac pericytes may undergo strong remodeling on injury. Conclusions Cardiac pericyte depletion induces both systolic and diastolic dysfunction, suggesting that pericyte dysfunction may contribute to the occurrence of cardiac diseases.


Subject(s)
Cardiomyopathies , Connexin 43 , Mice , Animals , Connexin 43/metabolism , Stroke Volume , Ventricular Function, Left , Cardiomyopathies/metabolism , Heart , Pericytes
3.
Nat Commun ; 14(1): 3766, 2023 06 24.
Article in English | MEDLINE | ID: mdl-37355632

ABSTRACT

Successful muscle regeneration relies on the interplay of multiple cell populations. However, the signals required for this coordinated intercellular crosstalk remain largely unknown. Here, we describe how the Hedgehog (Hh) signaling pathway controls the fate of fibro/adipogenic progenitors (FAPs), the cellular origin of intramuscular fat (IMAT) and fibrotic scar tissue. Using conditional mutagenesis and pharmacological Hh modulators in vivo and in vitro, we identify DHH as the key ligand that acts as a potent adipogenic brake by preventing the adipogenic differentiation of FAPs. Hh signaling also impacts muscle regeneration, albeit indirectly through induction of myogenic factors in FAPs. Our results also indicate that ectopic and sustained Hh activation forces FAPs to adopt a fibrogenic fate resulting in widespread fibrosis. In this work, we reveal crucial post-developmental functions of Hh signaling in balancing tissue regeneration and fatty fibrosis. Moreover, they provide the exciting possibility that mis-regulation of the Hh pathway with age and disease could be a major driver of pathological IMAT formation.


Subject(s)
Adipogenesis , Hedgehog Proteins , Adipogenesis/genetics , Cell Differentiation/physiology , Fibrosis , Hedgehog Proteins/genetics , Hedgehog Proteins/metabolism , Ligands , Muscle, Skeletal/metabolism , Signal Transduction , Animals
4.
Circ Res ; 132(1): 34-48, 2023 01 06.
Article in English | MEDLINE | ID: mdl-36448444

ABSTRACT

BACKGROUND: Lower-limb peripheral artery disease is one of the major complications of diabetes. Peripheral artery disease is associated with poor limb and cardiovascular prognoses, along with a dramatic decrease in life expectancy. Despite major medical advances in the treatment of diabetes, a substantial therapeutic gap remains in the peripheral artery disease population. Praliciguat is an orally available sGC (soluble guanylate cyclase) stimulator that has been reported both preclinically and in early stage clinical trials to have favorable effects in metabolic and hemodynamic outcomes, suggesting that it may have a potential beneficial effect in peripheral artery disease. METHODS: We evaluated the effect of praliciguat on hind limb ischemia recovery in a mouse model of type 2 diabetes. Hind limb ischemia was induced in leptin receptor-deficient (Leprdb/db) mice by ligation and excision of the left femoral artery. Praliciguat (10 mg/kg/day) was administered in the diet starting 3 days before surgery. RESULTS: Twenty-eight days after surgery, ischemic foot perfusion and function parameters were better in praliciguat-treated mice than in vehicle controls. Improved ischemic foot perfusion was not associated with either improved traditional cardiovascular risk factors (ie, weight, glycemia) or increased angiogenesis. However, treatment with praliciguat significantly increased arteriole diameter, decreased ICAM1 (intercellular adhesion molecule 1) expression, and prevented the accumulation of oxidative proangiogenic and proinflammatory muscle fibers. While investigating the mechanism underlying the beneficial effects of praliciguat therapy, we found that praliciguat significantly downregulated Myh2 and Cxcl12 mRNA expression in cultured myoblasts and that conditioned medium form praliciguat-treated myoblast decreased ICAM1 mRNA expression in endothelial cells. These results suggest that praliciguat therapy may decrease ICAM1 expression in endothelial cells by downregulating Cxcl12 in myocytes. CONCLUSIONS: Our results demonstrated that praliciguat promotes blood flow recovery in the ischemic muscle of mice with type 2 diabetes, at least in part by increasing arteriole diameter and by downregulating ICAM1 expression.


Subject(s)
Diabetes Mellitus, Type 2 , Peripheral Arterial Disease , Mice , Animals , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/drug therapy , Receptors, Leptin/genetics , Endothelial Cells/metabolism , Ischemia/metabolism , Disease Models, Animal , Reperfusion , Peripheral Arterial Disease/complications , Hindlimb/blood supply , Neovascularization, Physiologic , Muscle, Skeletal/metabolism , Mice, Inbred C57BL
5.
Front Physiol ; 13: 906272, 2022.
Article in English | MEDLINE | ID: mdl-35874523

ABSTRACT

Heart failure with preserved ejection fraction (HFpEF) has been recognized as the greatest single unmet need in cardiovascular medicine. Indeed, the morbi-mortality of HFpEF is high and as the population ages and the comorbidities increase, so considerably does the prevalence of HFpEF. However, HFpEF pathophysiology is still poorly understood and therapeutic targets are missing. An unifying, but untested, theory of the pathophysiology of HFpEF, proposed in 2013, suggests that cardiovascular risk factors lead to a systemic inflammation, which triggers endothelial cells (EC) and coronary microvascular dysfunction. This cardiac small vessel disease is proposed to be responsible for cardiac wall stiffening and diastolic dysfunction. This paradigm is based on the fact that microvascular dysfunction is highly prevalent in HFpEF patients. More specifically, HFpEF patients have been shown to have decreased cardiac microvascular density, systemic endothelial dysfunction and a lower mean coronary flow reserve. Importantly, impaired coronary microvascular function has been associated with the severity of HF. This review discusses evidence supporting the causal role of endothelial dysfunction in the pathophysiology of HFpEF in human and experimental models.

6.
Arterioscler Thromb Vasc Biol ; 42(6): 745-763, 2022 06.
Article in English | MEDLINE | ID: mdl-35510550

ABSTRACT

BACKGROUND: While endothelial dysfunction is suggested to contribute to heart failure with preserved ejection fraction pathophysiology, understanding the importance of the endothelium alone, in the pathogenesis of diastolic abnormalities has not yet been fully elucidated. Here, we investigated the consequences of specific endothelial dysfunction on cardiac function, independently of any comorbidity or risk factor (diabetes or obesity) and their potential effect on cardiomyocyte. METHODS: The ubiquitine ligase Pdzrn3, expressed in endothelial cells (ECs), was shown to destabilize tight junction. A genetic mouse model in which Pdzrn3 is overexpressed in EC (iEC-Pdzrn3) in adults was developed. RESULTS: EC-specific Pdzrn3 expression increased cardiac leakage of IgG and fibrinogen blood-born molecules. The induced edema demonstrated features of diastolic dysfunction, with increased end-diastolic pressure, alteration of dP/dt min, increased natriuretic peptides, in addition to limited exercise capacity, without major signs of cardiac fibrosis and inflammation. Electron microscopic images showed edema with disrupted EC-cardiomyocyte interactions. RNA sequencing analysis of gene expression in cardiac EC demonstrated a decrease in genes coding for endothelial extracellular matrix proteins, which could be related to the fragile blood vessel phenotype. Irregularly shaped capillaries with hemorrhages were found in heart sections of iEC-Pdzrn3 mice. We also found that a high-fat diet was not sufficient to provoke diastolic dysfunction; high-fat diet aggravated cardiac inflammation, associated with an altered cardiac metabolic signature in EC-Pdzrn3 mice, reminiscent of heart failure with preserved ejection fraction features. CONCLUSIONS: An increase of endothelial permeability is responsible for mediating diastolic dysfunction pathophysiology and for aggravating detrimental effects of a high-fat diet on cardiac inflammation and metabolism.


Subject(s)
Cardiomyopathies , Heart Failure , Animals , Capillary Permeability , Endothelial Cells/metabolism , Fibrosis , Heart Failure/genetics , Heart Failure/metabolism , Inflammation/metabolism , Mice , Myocytes, Cardiac/metabolism , Stroke Volume/physiology , Ubiquitin-Protein Ligases
8.
Cardiovasc Res ; 117(12): 2489-2501, 2021 11 01.
Article in English | MEDLINE | ID: mdl-33063110

ABSTRACT

AIMS: The therapeutic potential of Hedgehog (Hh) signalling agonists for vascular diseases is of growing interest. However, molecular and cellular mechanisms underlying the role of the Hh signalling in vascular biology remain poorly understood. The purpose of the present article is to clarify some conflicting literature data. METHODS AND RESULTS: With this goal, we have demonstrated that, unexpectedly, ectopically administered N-terminal Sonic Hh (N-Shh) and endogenous endothelial-derived Desert Hh (Dhh) induce opposite effects in endothelial cells (ECs). Notably, endothelial Dhh acts under its full-length soluble form (FL-Dhh) and activates Smoothened in ECs, while N-Shh inhibits it. At molecular level, N-Shh prevents FL-Dhh binding to Patched-1 (Ptch1) demonstrating that N-Shh acts as competitive antagonist to FL-Dhh. Besides, we found that even though FL-Hh ligands and N-Hh ligands all bind Ptch1, they induce distinct Ptch1 localization. Finally, we confirmed that in a pathophysiological setting, i.e. brain inflammation, astrocyte-derived N-Shh acts as a FL-Dhh antagonist. CONCLUSION: The present study highlights for the first time that FL-Dhh and N-Hh ligands have antagonistic properties especially in ECs.


Subject(s)
Astrocytes/metabolism , Capillary Permeability , Cerebral Cortex/blood supply , Corneal Neovascularization/metabolism , Encephalomyelitis, Autoimmune, Experimental/metabolism , Endothelial Cells/metabolism , Hedgehog Proteins/metabolism , Neovascularization, Pathologic , Animals , Astrocytes/drug effects , Astrocytes/pathology , Capillary Permeability/drug effects , Cells, Cultured , Corneal Neovascularization/genetics , Corneal Neovascularization/pathology , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/pathology , Endothelial Cells/drug effects , Endothelial Cells/pathology , Female , Hedgehog Proteins/administration & dosage , Hedgehog Proteins/genetics , Ligands , Male , Mice, Knockout , Patched-1 Receptor/metabolism , Protein Binding , Signal Transduction , Smoothened Receptor/genetics , Smoothened Receptor/metabolism
9.
Cell Surf ; 6: 100046, 2020 Dec.
Article in English | MEDLINE | ID: mdl-33204900

ABSTRACT

Cell walls are essential in the interaction of fungi with the (a)biotic environment and are also key to hyphal morphogenesis and mechanical strength. Here, we used solid-state NMR (ssNMR) spectroscopy combined with HPLC and GC-MS to study the structural organization of the cell wall of a representative of the Basidiomycota, one of the two main phyla of fungi. Based on the data we propose a refined model for the cell wall of a basidiomycete. In this model, the rigid core is built from α- and ß-(1,3)-glucan, ß-(1,3)-(1,6)-glucan, highly branched and single stranded ß-(1,4)-chitin as well as polymeric fucose. The mobile fraction of the cell wall is composed of ß-(1,3)-glucan, ß-(1,3)-(1,6)-glucan, ß-(1,6)-glucan, α-linked reducing and non-reducing ends and polymeric mannose. Together, these findings provide novel insights into the structural organization of the cell wall of the model basidiomycete S. commune that was previously based on destructive chemical and enzymatic analysis.

10.
PLoS Biol ; 18(11): e3000946, 2020 11.
Article in English | MEDLINE | ID: mdl-33253145

ABSTRACT

Inflammation of the central nervous system (CNS) induces endothelial blood-brain barrier (BBB) opening as well as the formation of a tight junction barrier between reactive astrocytes at the Glia Limitans. We hypothesized that the CNS parenchyma may acquire protection from the reactive astrocytic Glia Limitans not only during neuroinflammation but also when BBB integrity is compromised in the resting state. Previous studies found that astrocyte-derived Sonic hedgehog (SHH) stabilizes the BBB during CNS inflammatory disease, while endothelial-derived desert hedgehog (DHH) is expressed at the BBB under resting conditions. Here, we investigated the effects of endothelial Dhh on the integrity of the BBB and Glia Limitans. We first characterized DHH expression within endothelial cells at the BBB, then demonstrated that DHH is down-regulated during experimental autoimmune encephalomyelitis (EAE). Using a mouse model in which endothelial Dhh is inducibly deleted, we found that endothelial Dhh both opens the BBB via the modulation of forkhead box O1 (FoxO1) transcriptional activity and induces a tight junctional barrier at the Glia Limitans. We confirmed the relevance of this glial barrier system in human multiple sclerosis active lesions. These results provide evidence for the novel concept of "chronic neuroinflammatory tolerance" in which BBB opening in the resting state is sufficient to stimulate a protective barrier at the Glia Limitans that limits the severity of subsequent neuroinflammatory disease. In summary, genetic disruption of the BBB generates endothelial signals that drive the formation under resting conditions of a secondary barrier at the Glia Limitans with protective effects against subsequent CNS inflammation. The concept of a reciprocally regulated CNS double barrier system has implications for treatment strategies in both the acute and chronic phases of multiple sclerosis pathophysiology.


Subject(s)
Blood-Brain Barrier/physiology , Blood-Brain Barrier/physiopathology , Adherens Junctions/pathology , Adherens Junctions/physiology , Animals , Antigens, CD/genetics , Antigens, CD/physiology , Astrocytes/pathology , Astrocytes/physiology , Cadherins/genetics , Cadherins/physiology , Capillary Permeability/genetics , Capillary Permeability/physiology , Claudin-5/genetics , Claudin-5/physiology , Down-Regulation , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/pathology , Encephalomyelitis, Autoimmune, Experimental/physiopathology , Endothelial Cells/pathology , Endothelial Cells/physiology , Female , Hedgehog Proteins/deficiency , Hedgehog Proteins/genetics , Hedgehog Proteins/physiology , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Multiple Sclerosis/pathology , Multiple Sclerosis/physiopathology , Neuroglia/pathology , Neuroglia/physiology , Tight Junctions/pathology , Tight Junctions/physiology
11.
Circ Res ; 127(12): 1473-1487, 2020 12 04.
Article in English | MEDLINE | ID: mdl-33012251

ABSTRACT

RATIONALE: Tamoxifen prevents the recurrence of breast cancer and is also beneficial against bone demineralization and arterial diseases. It acts as an ER (estrogen receptor) α antagonist in ER-positive breast cancers, whereas it mimics the protective action of 17ß-estradiol in other tissues such as arteries. However, the mechanisms of these tissue-specific actions remain unclear. OBJECTIVE: Here, we tested whether tamoxifen is able to accelerate endothelial healing and analyzed the underlying mechanisms. METHODS AND RESULTS: Using 3 complementary mouse models of carotid artery injury, we demonstrated that both tamoxifen and estradiol accelerated endothelial healing, but only tamoxifen required the presence of the underlying medial smooth muscle cells. Chronic treatment with 17ß-estradiol and tamoxifen elicited differential gene expression profiles in the carotid artery. The use of transgenic mouse models targeting either whole ERα in a cell-specific manner or ERα subfunctions (membrane/extranuclear versus genomic/transcriptional) demonstrated that 17ß-estradiol-induced acceleration of endothelial healing is mediated by membrane ERα in endothelial cells, while the effect of tamoxifen is mediated by the nuclear actions of ERα in smooth muscle cells. CONCLUSIONS: Whereas tamoxifen acts as an antiestrogen and ERα antagonist in breast cancer but also on the membrane ERα of endothelial cells, it accelerates endothelial healing through activation of nuclear ERα in smooth muscle cells, inviting to revisit the mechanisms of action of selective modulation of ERα.


Subject(s)
Carotid Artery Injuries/drug therapy , Endothelial Cells/drug effects , Estrogen Receptor alpha/drug effects , Muscle, Smooth, Vascular/drug effects , Myocytes, Smooth Muscle/drug effects , Selective Estrogen Receptor Modulators/pharmacology , Tamoxifen/pharmacology , Wound Healing/drug effects , Animals , Carotid Arteries/drug effects , Carotid Arteries/metabolism , Carotid Arteries/pathology , Carotid Artery Injuries/metabolism , Carotid Artery Injuries/pathology , Disease Models, Animal , Endothelial Cells/metabolism , Endothelial Cells/pathology , Estradiol/pharmacology , Estrogen Receptor alpha/genetics , Estrogen Receptor alpha/metabolism , Female , Mice, Inbred C57BL , Mice, Transgenic , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/pathology , Signal Transduction , Time Factors
12.
Arterioscler Thromb Vasc Biol ; 40(12): e336-e349, 2020 12.
Article in English | MEDLINE | ID: mdl-33028094

ABSTRACT

OBJECTIVE: Evidences accumulated within the past decades identified hedgehog signaling as a new regulator of endothelium integrity. More specifically, we recently identified Dhh (desert hedgehog) as a downstream effector of Klf2 (Kruppel-like factor 2) in endothelial cells (ECs). The purpose of this study is to investigate whether hedgehog coreceptors Gas1 (growth arrest-specific 1) and Cdon (cell adhesion molecule-related/downregulated by oncogenes) may be used as therapeutic targets to modulate Dhh signaling in ECs. Approach and Results: We demonstrated that both Gas1 and Cdon are expressed in adult ECs and relied on either siRNAs- or EC-specific conditional knockout mice to investigate their role. We found that Gas1 deficiency mainly phenocopies Dhh deficiency especially by inducing VCAM-1 (vascular cell adhesion molecule 1) and ICAM-1 (intercellular adhesion molecule 1) overexpression while Cdon deficiency has opposite effects by promoting endothelial junction integrity. At a molecular level, Cdon prevents Dhh binding to Ptch1 (patched-1) and thus acts as a decoy receptor for Dhh, while Gas1 promotes Dhh binding to Smo (smoothened) and as a result potentiates Dhh effects. Since Cdon is upregulated in ECs treated by inflammatory cytokines, including TNF (tumor necrosis factor)-α and Il (interleukin)-1ß, we then tested whether Cdon inhibition would promote endothelium integrity in acute inflammatory conditions and found that both fibrinogen and IgG extravasation were decreased in association with an increased Cdh5 (cadherin-5) expression in the brain cortex of EC-specific Cdon knockout mice administered locally with Il-1ß. CONCLUSIONS: Altogether, these results demonstrate that Gas1 is a positive regulator of Dhh in ECs while Cdon is a negative regulator. Interestingly, Cdon blocking molecules may then be used to promote endothelium integrity, at least in inflammatory conditions.


Subject(s)
Blood-Brain Barrier/metabolism , Cell Adhesion Molecules/metabolism , Cell Cycle Proteins/metabolism , Corneal Neovascularization/metabolism , Endothelial Cells/metabolism , Endothelium, Corneal/metabolism , Hedgehog Proteins/metabolism , Inflammation/metabolism , Animals , Antigens, CD/genetics , Antigens, CD/metabolism , Blood-Brain Barrier/pathology , Cadherins/genetics , Cadherins/metabolism , Cell Adhesion Molecules/deficiency , Cell Adhesion Molecules/genetics , Cell Cycle Proteins/deficiency , Cell Cycle Proteins/genetics , Cells, Cultured , Corneal Neovascularization/genetics , Corneal Neovascularization/pathology , Disease Models, Animal , Endothelial Cells/pathology , Endothelium, Corneal/pathology , Female , GPI-Linked Proteins/deficiency , GPI-Linked Proteins/genetics , GPI-Linked Proteins/metabolism , Hedgehog Proteins/genetics , Humans , Inflammation/genetics , Inflammation/pathology , Male , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Patched-1 Receptor/metabolism , Signal Transduction , Smoothened Receptor/metabolism
13.
Dev Biol ; 463(1): 26-38, 2020 07 01.
Article in English | MEDLINE | ID: mdl-32151560

ABSTRACT

Non-syndromic mitral valve prolapse (MVP) is the most common heart valve disease affecting 2.4% of the population. Recent studies have identified genetic defects in primary cilia as causative to MVP, although the mechanism of their action is currently unknown. Using a series of gene inactivation approaches, we define a paracrine mechanism by which endocardially-expressed Desert Hedgehog (DHH) activates primary cilia signaling on neighboring valve interstitial cells. High-resolution imaging and functional assays show that DHH de-represses smoothened at the primary cilia, resulting in kinase activation of RAC1 through the RAC1-GEF, TIAM1. Activation of this non-canonical hedgehog pathway stimulates α-smooth actin organization and ECM remodeling. Genetic or pharmacological perturbation of this pathway results in enlarged valves that progress to a myxomatous phenotype, similar to valves seen in MVP patients. These data identify a potential molecular origin for MVP as well as establish a paracrine DHH-primary cilium cross-talk mechanism that is likely applicable across developmental tissue types.


Subject(s)
Cilia/metabolism , Hedgehog Proteins/metabolism , Mitral Valve/embryology , Actins/metabolism , Animals , Extracellular Matrix/metabolism , Heart Valve Diseases , Hedgehog Proteins/physiology , Mice , Mitral Valve Prolapse/genetics , Mitral Valve Prolapse/metabolism , Muscle, Smooth/metabolism , Muscle, Smooth/physiology , Myocytes, Smooth Muscle/metabolism , Neuropeptides/metabolism , Phenotype , Signal Transduction , Transcription Factors/metabolism , rac1 GTP-Binding Protein/metabolism
14.
Spine (Phila Pa 1976) ; 44(19): 1390-1395, 2019 Oct 01.
Article in English | MEDLINE | ID: mdl-31261281

ABSTRACT

STUDY DESIGN: Prospective study. OBJECTIVE: Determine risk factors and consider impact of low back pain for medical students. SUMMARY OF BACKGROUND DATA: Low back pain (LBP) is one of the most prevalent complaints among students. The vulnerability of medical students due to stress and numerous hours of studying and training makes them at risk of LBP. METHODS: We submitted an online self-administered modified version of the Standardized Nordic Questionnaire to 1800 medical students from 2nd to 6th year from December 2017 to March 2018. RESULTS: A total of 1243/1800 (68.9%) students responded to our survey. Mean age was 23.3 ±â€Š2.9 years ranging from 18 to 44 years. 835 (72.1%) students reported suffering from LBP. In multivariate analysis with logistic regression analysis model, the third year of medical studies (odds ratio [OR]: 0.558, 95% confidence interval [CI] 0.387-0.805; P = 0.002) was identified as an independent prognostic factor of LBP. Moreover, exercising weekly (OR: 1.835, 95% CI 0.933-2.5; P = 0.01) and walking at least 30 minutes a day (OR: 1.458, 95% CI 1.129-1.876; P = 0.01) significantly improve LBP. LBP generate higher monthly consumption of an analgesic (OR: 32.8, 95% CI 4.271-252.2; P < 0.001). Finally, LBP had a severe repercussion on student work (OR: 18.89, 95% CI 10.122-35.253; P < 0.0001), on the quality of sleep (OR: 12.162, 95% CI 6.917-21.386; P < 0.0001) and on their personal life (OR: 12.343, 95% CI 5.681-26.8; P < 0.0001). CONCLUSION: Medical students reported high prevalence of LBP with severe consequences. Our educational perspective is to identify the risk factors of LBP, fight them, to improve the medical student' work, and welfare. LEVEL OF EVIDENCE: 3.


Subject(s)
Low Back Pain/epidemiology , Students, Medical/statistics & numerical data , Adolescent , Adult , Humans , Prospective Studies , Risk Factors , Young Adult
15.
Int J Mol Sci ; 20(12)2019 Jun 24.
Article in English | MEDLINE | ID: mdl-31238510

ABSTRACT

The role of Hedgehog (Hh) signaling in vascular biology has first been highlighted in embryos by Pepicelli et al. in 1998 and Rowitch et al. in 1999. Since then, the proangiogenic role of the Hh ligands has been confirmed in adults, especially under pathologic conditions. More recently, the Hh signaling has been proposed to improve vascular integrity especially at the blood-brain barrier (BBB). However, molecular and cellular mechanisms underlying the role of the Hh signaling in vascular biology remain poorly understood and conflicting results have been reported. As a matter of fact, in several settings, it is currently not clear whether Hh ligands promote vessel integrity and quiescence or destabilize vessels to promote angiogenesis. The present review relates the current knowledge regarding the role of the Hh signaling in vasculature development, maturation and maintenance, discusses the underlying proposed mechanisms and highlights controversial data which may serve as a guideline for future research. Most importantly, fully understanding such mechanisms is critical for the development of safe and efficient therapies to target the Hh signaling in both cancer and cardiovascular/cerebrovascular diseases.


Subject(s)
Blood Vessels/embryology , Blood Vessels/metabolism , Hedgehog Proteins/metabolism , Organogenesis , Signal Transduction , Animals , Blood-Brain Barrier/metabolism , Cell Differentiation , Humans , Ligands , Neovascularization, Physiologic
17.
J Thromb Haemost ; 17(5): 827-840, 2019 05.
Article in English | MEDLINE | ID: mdl-30801958

ABSTRACT

Essentials To reliably study the respective roles of blood and endothelial cells in hemostasis, mouse models with a strong and specific endothelial expression of the Cre recombinase are needed. Using mT/mG reporter mice and conditional JAK2V617F/WT mice, we compared Pdgfb-iCreERT2 and Cdh5(PAC)-CreERT2 with well-characterized Tie2-Cre mice. Comparison of recombination efficiency and specificity towards blood lineage reveals major differences between endothelial transgenic mice. Cre-mediated recombination occurs in a small number of adult hematopoietic stem cells in Pdgfb-iCreERT2;JAK2V617F/WT transgenic mice. SUMMARY: Background The vessel wall, and particularly blood endothelial cells (BECs), are intensively studied to better understand hemostasis and target thrombosis. To understand the specific role of BECs, it is important to have mouse models that allow specific and homogeneous expression of genes of interest in all BEC beds without concomitant expression in blood cells. Inducible Pdgfb-iCreERT2 and Cdh5(PAC)-CreERT2 transgenic mice are widely used for BEC targeting. However, issues remain in terms of recombination efficiency and specificity regarding hematopoietic cells. Objectives To determine which mouse model to choose when strong expression of a transgene is required in adult BECs from various organs, without concomitant expression in hematopoietic cells. Methods Using mT/mG reporter mice to measure recombination efficiency and conditional JAK2V617F/WT mice to assess specificity regarding hematopoietic cells, we compared Pdgfb-iCreERT2 and Cdh5(PAC)-CreERT2 with well-characterized Tie2-Cre mice. Results Adult Cdh5(PAC)-CreERT2 mice are endothelial specific but require a dose of 10 mg of tamoxifen to allow constant Cre expression. Pdgfb-iCreERT2 mice injected with 5 mg of tamoxifen are appropriate for most endothelial research fields except liver studies, as hepatic sinusoid ECs are not recombined. Surprisingly, 2 months after induction of Cre-mediated recombination, all Pdgfb-iCreERT2;JAK2V617F/WT mice developed a myeloproliferative neoplasm that is related to the presence of JAK2V617F in hematopoietic cells, showing for the first time that Cre-mediated recombination occurs in a small number of adult hematopoietic stem cells in Pdgfb-iCreERT2 transgenic mice. Conclusion This study provides useful guidelines for choosing the best mouse line to study the role of BECs in hemostasis and thrombosis.


Subject(s)
Endothelial Cells/cytology , Hematopoietic Stem Cells/cytology , Lymphokines/genetics , Lymphokines/metabolism , Platelet-Derived Growth Factor/genetics , Platelet-Derived Growth Factor/metabolism , Alleles , Animals , Brain/metabolism , Hemostasis , Integrases/metabolism , Kidney/metabolism , Liver/metabolism , Lung/metabolism , Mice , Mice, Transgenic , Myocardium/metabolism , Polymerase Chain Reaction , Retina/metabolism , Tamoxifen/pharmacology , Thrombosis/metabolism
18.
Haematologica ; 104(1): 70-81, 2019 01.
Article in English | MEDLINE | ID: mdl-30171023

ABSTRACT

Thrombosis is the main cause of morbidity and mortality in patients with JAK2V617F myeloproliferative neoplasms. Recent studies have reported the presence of JAK2V617F in endothelial cells of some patients with myeloproliferative neoplasms. We investigated the role of endothelial cells that express JAK2V617F in thrombus formation using an in vitro model of human endothelial cells overexpressing JAK2V617F and an in vivo model of mice with endothelial-specific JAK2V617F expression. Interestingly, these mice displayed a higher propensity for thrombus. When deciphering the mechanisms by which JAK2V617F-expressing endothelial cells promote thrombosis, we observed that they have a pro-adhesive phenotype associated with increased endothelial P-selectin exposure, secondary to degranulation of Weibel-Palade bodies. We demonstrated that P-selectin blockade was sufficient to reduce the increased propensity of thrombosis. Moreover, treatment with hydroxyurea also reduced thrombosis and decreased the pathological interaction between leukocytes and JAK2V617F-expressing endothelial cells through direct reduction of endothelial P-selectin expression. Taken together, our data provide evidence that JAK2V617F-expressing endothelial cells promote thrombosis through induction of endothelial P-selectin expression, which can be reversed by hydroxyurea. Our findings increase our understanding of thrombosis in patients with myeloproliferative neoplasms, at least those with JAK2V617F-positive endothelial cells, and highlight a new role for hydroxyurea. This novel finding provides the proof of concept that an acquired genetic mutation can affect the pro-thrombotic nature of endothelial cells, suggesting that other mutations in endothelial cells could be causal in thrombotic disorders of unknown cause, which account for 50% of recurrent venous thromboses.


Subject(s)
Endothelial Cells/metabolism , Janus Kinase 2/biosynthesis , P-Selectin/biosynthesis , Thrombosis/metabolism , Animals , Disease Models, Animal , Endothelial Cells/pathology , Gene Expression Regulation, Enzymologic/drug effects , Humans , Hydroxyurea/pharmacology , Janus Kinase 2/genetics , Mice , Mice, Transgenic , P-Selectin/genetics , Thrombosis/drug therapy , Thrombosis/genetics , Thrombosis/pathology
19.
Circ Res ; 123(9): 1053-1065, 2018 10 12.
Article in English | MEDLINE | ID: mdl-30355159

ABSTRACT

RATIONALE: Klf (kruppel-like factor) 2 is critical to establish and maintain endothelial integrity. OBJECTIVE: Therefore, determining upstream and downstream mediators of Klf2 would lead to alternative therapeutic targets in cardiovascular disease management. METHODS AND RESULTS: Here we identify Dhh (desert hedgehog) as a downstream effector of Klf2, whose expression in endothelial cells (ECs) is upregulated by shear stress and decreased by inflammatory cytokines. Consequently, we show that Dhh knockdown in ECs promotes endothelial permeability and EC activation and that Dhh agonist prevents TNF-α (tumor necrosis factor alpha) or glucose-induced EC dysfunction. Moreover, we demonstrate that human critical limb ischemia, a pathological condition linked to diabetes mellitus and inflammation, is associated to major EC dysfunction. By recreating a complex model of critical limb ischemia in diabetic mice, we found that Dhh-signaling agonist significantly improved EC function without promoting angiogenesis, which subsequently improved muscle perfusion. CONCLUSION: Restoring EC function leads to significant critical limb ischemia recovery. Dhh appears to be a promising target, downstream of Klf2, to prevent the endothelial dysfunction involved in ischemic vascular diseases.


Subject(s)
Endothelial Cells/metabolism , Hedgehog Proteins/metabolism , Ischemia/metabolism , Kruppel-Like Transcription Factors/metabolism , Muscle, Skeletal/blood supply , Neovascularization, Physiologic , Animals , Autocrine Communication , Capillary Permeability , Cells, Cultured , Critical Illness , Cyclohexylamines/pharmacology , Cytokines/metabolism , Disease Models, Animal , Endothelial Cells/drug effects , Gene Expression Regulation , Hedgehog Proteins/deficiency , Hedgehog Proteins/genetics , Hindlimb , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Inflammation Mediators/metabolism , Ischemia/drug therapy , Ischemia/genetics , Ischemia/physiopathology , Kruppel-Like Transcription Factors/genetics , Male , Mice, Inbred C57BL , Mice, Knockout , Neovascularization, Physiologic/drug effects , Regional Blood Flow , Signal Transduction , Stress, Mechanical , Thiophenes/pharmacology
20.
Physiol Rep ; 6(12): e13736, 2018 06.
Article in English | MEDLINE | ID: mdl-29932507

ABSTRACT

This study combines two well-known vascular research models, hyperoxia and hind limb ischemia, aiming to better characterize capacities of the hyperoxia challenge. We studied two groups of C57/BL6 male mice, a control (C) and a hind limb ischemia (HLI) group. Perfusion from both limbs was recorded in all animals by laser Doppler techniques under an oxygen (O2 ) saturated atmosphere, once for control and, during 35 days for the HLI group. We used a third set of normoxic animals for HLI morphometric control. The expected variability of responses was higher for the younger animals. In the HLI group, capillary density normalized at Day 21 as expected, but not microcirculatory physiology. In the operated limb, perfusion decreased dramatically following surgery (Day 4), as a slight reduction in the non-operated limb was also noted. Consistently, the response to hyperoxia was an increased perfusion in the ischemic limb and decreased perfusion in the contralateral limb. Only at Day 35, both limbs exhibited similar flows, although noticeably lower than Day 0. These observations help to understand some of the functional variability attributed to the hyperoxia model, by showing (i) differences in the circulation of the limb pairs to readjust a new perfusion set-point even after ischemia, an original finding implying that (ii) data from both limbs should be recorded when performing distal measurements in vivo. Our data demonstrate that the new vessels following HLI are not functionally normal, and this also affects the non-operated limb. These findings confirm the discriminative capacities of the hyperoxia challenge and suggest its potential utility to study other pathologies with vascular impact.


Subject(s)
Hindlimb/blood supply , Hyperoxia/physiopathology , Ischemia/physiopathology , Neovascularization, Physiologic/physiology , Animals , Disease Models, Animal , Laser-Doppler Flowmetry , Male , Mice, Inbred C57BL , Microcirculation/physiology , Regeneration/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...