Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
1.
Sci Adv ; 10(7): eadj7481, 2024 Feb 16.
Article in English | MEDLINE | ID: mdl-38354249

ABSTRACT

Exercise promotes pulsatile shear stress in the arterial circulation and ameliorates cardiometabolic diseases. However, exercise-mediated metabolic transducers for vascular protection remain under-investigated. Untargeted metabolomic analysis demonstrated that wild-type mice undergoing voluntary wheel running exercise expressed increased endothelial stearoyl-CoA desaturase 1 (SCD1) that catalyzes anti-inflammatory lipid metabolites, namely, oleic (OA) and palmitoleic acids (PA), to mitigate NF-κB-mediated inflammatory responses. In silico analysis revealed that exercise augmented time-averaged wall shear stress but mitigated flow recirculation and oscillatory shear index in the lesser curvature of the mouse aortic arch. Following exercise, endothelial Scd1-deleted mice (Ldlr-/- Scd1EC-/-) on high-fat diet developed persistent VCAM1-positive endothelium in the lesser curvature and the descending aorta, whereas SCD1 overexpression via adenovirus transfection mitigated endoplasmic reticulum stress and inflammatory biomarkers. Single-cell transcriptomics of the aorta identified Scd1-positive and Vcam1-negative endothelial subclusters interacting with other candidate genes. Thus, exercise mitigates flow recirculation and activates endothelial SCD1 to catalyze OA and PA for vascular endothelial protection.


Subject(s)
Aorta , Motor Activity , Animals , Mice , Aorta/metabolism , Diet, High-Fat , Endothelium, Vascular/metabolism , Stearoyl-CoA Desaturase/genetics , Stearoyl-CoA Desaturase/metabolism
2.
bioRxiv ; 2023 May 03.
Article in English | MEDLINE | ID: mdl-37205360

ABSTRACT

Exercise modulates vascular plasticity in multiple organ systems; however, the metabolomic transducers underlying exercise and vascular protection in the disturbed flow-prone vasculature remain under-investigated. We simulated exercise-augmented pulsatile shear stress (PSS) to mitigate flow recirculation in the lesser curvature of the aortic arch. When human aortic endothelial cells (HAECs) were subjected to PSS ( τ ave = 50 dyne·cm -2 , ∂τ/∂t = 71 dyne·cm -2 ·s -1 , 1 Hz), untargeted metabolomic analysis revealed that Stearoyl-CoA Desaturase (SCD1) in the endoplasmic reticulum (ER) catalyzed the fatty acid metabolite, oleic acid (OA), to mitigate inflammatory mediators. Following 24 hours of exercise, wild-type C57BL/6J mice developed elevated SCD1-catalyzed lipid metabolites in the plasma, including OA and palmitoleic acid (PA). Exercise over a 2-week period increased endothelial SCD1 in the ER. Exercise further modulated the time-averaged wall shear stress (TAWSS or τ ave) and oscillatory shear index (OSI ave ), upregulated Scd1 and attenuated VCAM1 expression in the disturbed flow-prone aortic arch in Ldlr -/- mice on high-fat diet but not in Ldlr -/- Scd1 EC-/- mice. Scd1 overexpression via recombinant adenovirus also mitigated ER stress. Single cell transcriptomic analysis of the mouse aorta revealed interconnection of Scd1 with mechanosensitive genes, namely Irs2 , Acox1 and Adipor2 that modulate lipid metabolism pathways. Taken together, exercise modulates PSS ( τ ave and OSI ave ) to activate SCD1 as a metabolomic transducer to ameliorate inflammation in the disturbed flow-prone vasculature.

3.
Adv Sci (Weinh) ; 8(23): e2103266, 2021 12.
Article in English | MEDLINE | ID: mdl-34687279

ABSTRACT

Activation of endothelial cells following severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection is thought to be the primary driver for the increasingly recognized thrombotic complications in coronavirus disease 2019 patients, potentially due to the SARS-CoV-2 Spike protein binding to the human angiotensin-converting enzyme 2 (hACE2). Vaccination therapies use the same Spike sequence or protein to boost host immune response as a protective mechanism against SARS-CoV-2 infection. As a result, cases of thrombotic events are reported following vaccination. Although vaccines are generally considered safe, due to genetic heterogeneity, age, or the presence of comorbidities in the population worldwide, the prediction of severe adverse outcome in patients remains a challenge. To elucidate Spike proteins underlying patient-specific-vascular thrombosis, the human microcirculation environment is recapitulated using a novel microfluidic platform coated with human endothelial cells and exposed to patient specific whole blood. Here, the blood coagulation effect is tested after exposure to Spike protein in nanoparticles and Spike variant D614G in viral vectors and the results are corroborated using live SARS-CoV-2. Of note, two potential strategies are also examined to reduce blood clot formation, by using nanoliposome-hACE2 and anti-Interleukin (IL) 6 antibodies.


Subject(s)
Blood Coagulation/physiology , SARS-CoV-2/isolation & purification , Spike Glycoprotein, Coronavirus/metabolism , Angiotensin-Converting Enzyme 2/chemistry , Angiotensin-Converting Enzyme 2/genetics , Angiotensin-Converting Enzyme 2/metabolism , Antibodies/chemistry , Antibodies/immunology , Antibodies/metabolism , COVID-19/diagnosis , COVID-19/virology , Endothelial Cells/chemistry , Endothelial Cells/cytology , Endothelial Cells/metabolism , Fibrin/chemistry , Fibrin/metabolism , Genetic Vectors/genetics , Genetic Vectors/metabolism , Humans , Interleukin-6/immunology , Liposomes/chemistry , Microfluidics/methods , Mutation , Nanoparticles/chemistry , Platelet Aggregation , SARS-CoV-2/metabolism , Spike Glycoprotein, Coronavirus/analysis , Spike Glycoprotein, Coronavirus/genetics
4.
Curr Top Membr ; 87: 131-151, 2021.
Article in English | MEDLINE | ID: mdl-34696883

ABSTRACT

Living cells are exposed to multiple mechanical stimuli from the extracellular matrix or from surrounding cells. Mechanoreceptors are molecules that display status changes in response to mechanical stimulation, transforming physical cues into biological responses to help the cells adapt to dynamic changes of the microenvironment. Mechanical stimuli are responsible for shaping the tridimensional development and patterning of the organs in early embryonic stages. The development of the heart is one of the first morphogenetic events that occur in embryos. As the circulation is established, the vascular system is exposed to constant shear stress, which is the force created by the movement of blood. Both spatial and temporal variations in shear stress differentially modulate critical steps in heart development, such as trabeculation and compaction of the ventricular wall and the formation of the heart valves. Zebrafish embryos are small, transparent, have a short developmental period and allow for real-time visualization of a variety of fluorescently labeled proteins to recapitulate developmental dynamics. In this review, we will highlight the application of zebrafish models as a genetically tractable model for investigating cardiovascular development and regeneration. We will introduce our approaches to manipulate mechanical forces during critical stages of zebrafish heart development and in a model of vascular regeneration, as well as advances in imaging technologies to capture these processes at high resolution. Finally, we will discuss the role of molecules of the Plexin family and Piezo cation channels as major mechanosensors recently implicated in cardiac morphogenesis.


Subject(s)
Mechanotransduction, Cellular , Zebrafish , Animals , Models, Animal , Morphogenesis , Stress, Mechanical
5.
Mol Metab ; 25: 50-63, 2019 07.
Article in English | MEDLINE | ID: mdl-31027994

ABSTRACT

OBJECTIVE: Obesity has increased to pandemic levels and enhanced understanding of adipose regulation is required for new treatment strategies. Although bone morphogenetic proteins (BMPs) influence adipogenesis, the effect of BMP antagonists such as Noggin is largely unknown. The aim of the study was to define the role of Noggin, an extracellular BMP inhibitor, in adipogenesis. METHODS: We generated adipose-derived progenitor cells and a mouse model with adipocyte-specific Noggin deletion using the AdiponectinCre transgenic mouse, and determined the adipose phenotype of Noggin-deficiency. RESULTS: Our studies showed that Noggin is expressed in progenitor cells but declines in adipocytes, possibly allowing for lipid accumulation. Correspondingly, adipocyte-specific Noggin deletion in vivo promoted age-related obesity in both genders with no change in food intake. Although the loss of Noggin caused white adipose tissue hypertrophy, and whitening and impaired function in brown adipose tissue in both genders, there were clear gender differences with the females being most affected. The females had suppressed expression of brown adipose markers and thermogenic genes including peroxisome proliferator activated receptor gamma coactivator 1 alpha (PGC1alpha) and uncoupling protein 1 (UCP1) as well as genes associated with adipogenesis and lipid metabolism. The males, on the other hand, had early changes in a few BAT markers and thermogenic genes, but the main changes were in the genes associated with adipogenesis and lipid metabolism. Further characterization revealed that both genders had reductions in VO2, VCO2, and RER, whereas females also had reduced heat production. Noggin was also reduced in diet-induced obesity in inbred mice consistent with the obesity phenotype of the Noggin-deficient mice. CONCLUSIONS: BMP signaling regulates female and male adipogenesis through different metabolic pathways. Modulation of adipose tissue metabolism by select BMP antagonists may be a strategy for long-term regulation of age-related weight gain and obesity.


Subject(s)
Adipocytes/metabolism , Adipogenesis/physiology , Bone Morphogenetic Proteins/metabolism , Carrier Proteins/metabolism , Obesity/metabolism , Adipocytes/pathology , Adipogenesis/genetics , Adipose Tissue/metabolism , Adipose Tissue, Brown/metabolism , Adipose Tissue, White/metabolism , Animals , Bone Morphogenetic Proteins/drug effects , Carrier Proteins/genetics , Disease Models, Animal , Eating , Female , Gene Deletion , Gene Expression Regulation , Genetic Association Studies , Lipid Accumulation Product , Lipid Metabolism , Male , Mice , Mice, Obese , Mice, Transgenic , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/genetics , Signal Transduction/genetics , Thermogenesis/genetics , Thermogenesis/physiology , Transcriptome , Uncoupling Protein 1/genetics
6.
Obes Rev ; 20(5): 648-658, 2019 05.
Article in English | MEDLINE | ID: mdl-30609449

ABSTRACT

The bone morphogenetic proteins (BMPs) belong to the same superfamily as related to transforming growth factor ß (TGFß), growth and differentiation factors (GDFs), and activins. They were initially described as inducers of bone formation but are now known to be involved in morphogenetic activities and cell differentiation throughout the body, including the development of adipose tissue and adipogenic differentiation. BMP4 and BMP7 are the most studied BMPs in adipose tissue, with major roles in white adipogenesis and brown adipogenesis, respectively, but other BMPs such as BMP2, BMP6, and BMP8b as well as some inhibitors and modulators have been shown to also affect adipogenesis. It has become ever more important to understand adipose regulation, including the BMP pathways, in light of the strong links between obesity and metabolic and cardiovascular disease. In this review, we summarize the available information on BMP signaling in adipose tissue using preferentially articles that have appeared in the last decade, which together demonstrate the importance of BMP signaling in adipose biology.


Subject(s)
Adipogenesis/physiology , Adipose Tissue/metabolism , Bone Morphogenetic Proteins/metabolism , Signal Transduction/physiology , Animals , Humans
7.
J Cell Biol ; 216(10): 3369-3385, 2017 10 02.
Article in English | MEDLINE | ID: mdl-28838957

ABSTRACT

The vascular endothelium is critical for induction of appropriate lineage differentiation in organogenesis. In this study, we report that dysfunctional pulmonary endothelium, resulting from the loss of matrix Gla protein (MGP), causes ectopic hepatic differentiation in the pulmonary epithelium. We demonstrate uncontrolled induction of the hepatic growth factor (HGF) caused by dysregulated cross talk between pulmonary endothelium and epithelium in Mgp-null lungs. Elevated HGF induced hepatocyte nuclear factor 4 α (Hnf4a), which competed with NK2 homeobox 1 (Nkx2.1) for binding to forkhead box A2 (Foxa2) to drive hepatic differentiation in Mgp-null airway progenitor cells. Limiting endothelial HGF reduced Hnf4a, abolished interference of Hnf4a with Foxa2, and reduced hepatic differentiation in Mgp-null lungs. Together, our results suggest that endothelial-epithelial interactions, maintained by MGP, are essential in pulmonary cell differentiation.


Subject(s)
Cell Communication , Cell Differentiation , Endothelium, Vascular/metabolism , Epithelial Cells/metabolism , Respiratory Mucosa/metabolism , Animals , Endothelium, Vascular/cytology , Epithelial Cells/cytology , Hepatocyte Growth Factor/genetics , Hepatocyte Growth Factor/metabolism , Hepatocyte Nuclear Factor 3-beta/genetics , Hepatocyte Nuclear Factor 3-beta/metabolism , Hepatocyte Nuclear Factor 4/genetics , Hepatocyte Nuclear Factor 4/metabolism , Mice , Mice, Knockout , Proteins/genetics , Proteins/metabolism , Respiratory Mucosa/cytology
8.
PLoS One ; 11(12): e0167936, 2016.
Article in English | MEDLINE | ID: mdl-27936229

ABSTRACT

Endothelial-mesenchymal transition (EndMT) drives endothelium to contribute to normal development and disease processes. Here, we report that EndMTs occur in the diabetic endothelium of Ins2Akita/wt mouse, and show that induction of sex determining region Y-box 2 (Sox2) is a mediator of excess BMP signaling that results in activation of EndMTs and increased vascular calcification. We also find an induction of a complex of serine proteases in the diabetic endothelium, required for the up-regulation of Sox2. Our results suggest that EndMTs contribute to vascular calcification in diabetic arteries.


Subject(s)
Blood Vessels/pathology , Calcinosis/pathology , Endothelium, Vascular/pathology , Insulin/genetics , Mesoderm/pathology , Animals , Diabetes Mellitus, Experimental/genetics , Endothelium, Vascular/enzymology , Mice , Mice, Transgenic , SOXB1 Transcription Factors/physiology , Serine Proteases/metabolism , Up-Regulation
9.
Atherosclerosis ; 253: 124-127, 2016 10.
Article in English | MEDLINE | ID: mdl-27615595

ABSTRACT

BACKGROUND AND AIMS: Endothelial-mesenchymal transitions (EndMTs) in endothelial cells (ECs) contribute to vascular disease. METHODS: We used ApoE-/- mice fed a high-fat/high-cholesterol diet. RESULTS: We reported evidence of EndMT in atherosclerotic lesions contributing to calcification. Stem cell and mesenchymal markers, including sex-determining region Y-box 2 (Sox2), were upregulated in aortic ECs of fat-fed ApoE-/- mice. Limiting Sox2 decreased marker expression and calcification in ApoE-/- aortas. Furthermore, a complex of serine proteases was upregulated in ApoE-/- aortic ECs. Blockade of these proteases reduced expression of Sox2 and atherosclerotic lesion calcification. CONCLUSIONS: Together, our data suggest that EndMTs contribute to atherosclerotic lesion calcification.


Subject(s)
Aorta/cytology , Atherosclerosis/blood , Calcinosis/blood , Endothelial Cells/cytology , Epithelial-Mesenchymal Transition , SOXB1 Transcription Factors/genetics , Animals , Aorta/metabolism , Cholesterol/metabolism , Cytokines/metabolism , Endothelium, Vascular/metabolism , Humans , Mice , Mice, Knockout, ApoE , Plaque, Atherosclerotic/pathology , SOXB1 Transcription Factors/metabolism , Serine Endopeptidases/metabolism , Stem Cells/cytology , Up-Regulation
10.
Angiogenesis ; 19(1): 1-7, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26364300

ABSTRACT

Matrix Gla protein (MGP) is an antagonist of bone morphogenetic proteins and expressed in vascular endothelial cells. Lack of MGP causes vascular abnormalities in multiple organs in mice. The objective of this study is to define the role of MGP in early endothelial differentiation. We find that expression of endothelial markers is highly induced in Mgp null organs, which, in wild type, contain high MGP expression. Furthermore, Mgp null embryonic stem cells express higher levels of endothelial markers than wild-type controls and an abnormal temporal pattern of expression. We also find that the Mgp-deficient endothelial cells adopt characteristics of mesenchymal stem cells. We conclude that loss of MGP causes dysregulation of early endothelial differentiation.


Subject(s)
Calcium-Binding Proteins/metabolism , Cell Differentiation , Endothelial Cells/cytology , Endothelial Cells/metabolism , Extracellular Matrix Proteins/metabolism , Mouse Embryonic Stem Cells/cytology , Animals , Biomarkers/metabolism , Calcium-Binding Proteins/deficiency , Cell Count , Extracellular Matrix Proteins/deficiency , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Mice , Mice, Inbred C57BL , Matrix Gla Protein
11.
Oxid Med Cell Longev ; 2016: 9124676, 2016.
Article in English | MEDLINE | ID: mdl-26635913

ABSTRACT

Oxidative stress is associated with cardiac and vascular defects leading to hypertension and atherosclerosis, being superoxide dismutase (SOD) one of the main intracellular antioxidant defence mechanisms. Although several parameters of vascular function and structure have a predictive value for cardiovascular morbidity-mortality in hypertensive patients, there are no studies on the involvement of SOD serum levels with these vascular parameters. Thus, we assessed if SOD serum levels are correlated with parameters of vascular function and structure and with cardiovascular risk in hypertensive and type 2 diabetic patients. We enrolled 255 consecutive hypertensive and diabetic patients and 52 nondiabetic and nonhypertensive controls. SOD levels were measured with an enzyme-linked immunosorbent assay kit. Vascular function and structure were evaluated by pulse wave velocity, augmentation index, ambulatory arterial stiffness index, and carotid intima-media thickness. We detected negative correlations between SOD and pressure wave velocity, peripheral and central augmentation index and ambulatory arterial stiffness index, pulse pressure, and plasma HDL-cholesterol, as well as positive correlations between SOD and plasma uric acid and triglycerides. Our study shows that SOD is a marker of cardiovascular alterations in hypertensive and diabetic patients, since changes in its serum levels are correlated with alterations in vascular structure and function.


Subject(s)
Diabetes Mellitus, Type 2/blood , Hypertension/blood , Oxidative Stress , Superoxide Dismutase/blood , Adult , Aged , Female , Humans , Male , Middle Aged , Pulse Wave Analysis , Triglycerides/blood , Uric Acid/blood
12.
Medicine (Baltimore) ; 94(41): e1617, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26469898

ABSTRACT

Early detection of hypertensive end-organ damage and secondary diseases are key determinants of cardiovascular prognosis in patients suffering from arterial hypertension. Presently, there are no biomarkers for the detection of hypertensive target organ damage, most outstandingly including blood vessels, the heart, and the kidneys.We aimed to validate the usefulness of the urinary excretion of the serine protease kallikrein-related peptidase 9 (KLK9) as a biomarker of hypertension-induced target organ damage.Urinary, plasma, and renal tissue levels of KLK9 were measured by the Western blot in different rat models of hypertension, including angiotensin-II infusion, DOCA-salt, L-NAME administration, and spontaneous hypertension. Urinary levels were associated to cardiovascular and renal injury, assessed by histopathology. The origin of urinary KLK9 was investigated through in situ renal perfusion experiments.The urinary excretion of KLK9 is increased in different experimental models of hypertension in rats. The ACE inhibitor trandolapril significantly reduced arterial pressure and the urinary level of KLK9. Hypertension did not increase kidney, heart, liver, lung, or plasma KLK9 levels. Hypertension-induced increased urinary excretion of KLK9 results from specific alterations in its tubular reabsorption, even in the absence of overt nephropathy. KLK9 urinary excretion strongly correlates with cardiac hypertrophy and aortic wall thickening.KLK9 appears in the urine in the presence of hypertension as a result of subtle renal handling alterations. Urinary KLK9 might be potentially used as an indicator of hypertensive cardiac and vascular damage.


Subject(s)
Hypertension/metabolism , Kallikreins/blood , Kallikreins/urine , Kidney Diseases/blood , Angiotensin-Converting Enzyme Inhibitors/pharmacology , Animals , Arterial Pressure , Biomarkers , Blood Pressure , Cardiovascular Diseases/blood , Disease Models, Animal , Gene Expression , Indoles/pharmacology , Kidney/metabolism , Male , Polymerase Chain Reaction , Rats , Rats, Wistar
13.
Circ Res ; 117(9): 758-69, 2015 Oct 09.
Article in English | MEDLINE | ID: mdl-26265629

ABSTRACT

RATIONALE: Endothelial cells have the ability to undergo endothelial-mesenchymal transitions (EndMTs), by which they acquire a mesenchymal phenotype and stem cell-like characteristics. We previously found that EndMTs occurred in the endothelium deficient in matrix γ-carboxyglutamic acid protein enabling endothelial cells to contribute cells to vascular calcification. However, the mechanism responsible for initiating EndMTs is not fully understood. OBJECTIVE: To determine the role of specific serine proteases and sex determining region Y-box 2 (Sox2) in the initiation of EndMTs. METHODS AND RESULTS: In this study, we used in vivo and in vitro models of vascular calcification to demonstrate that serine proteases and Sox2 are essential for the initiation of EndMTs in matrix γ-carboxyglutamic acid protein-deficient endothelium. We showed that expression of a group of specific serine proteases was highly induced in endothelial cells at sites of vascular calcification in Mgp null aortas. Treatment with serine protease inhibitors decreased both stem cell marker expression and vascular calcification. In human aortic endothelial cells, this group of serine proteases also induced EndMTs, and the activation of proteases was mediated by Sox2. Knockdown of the serine proteases or Sox2 diminished EndMTs and calcification. Endothelial-specific deletion of Sox2 decreased expression of stem cell markers and aortic calcification in matrix γ-carboxyglutamic acid protein-deficient mice. CONCLUSIONS: Our results suggest that Sox2-mediated activation of specific serine proteases is essential for initiating EndMTs, and thus, may provide new therapeutic targets for treating vascular calcification.


Subject(s)
Calcinosis , Endothelium, Vascular/metabolism , Mesoderm/metabolism , Serine Endopeptidases/metabolism , Animals , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Cells, Cultured , Endothelium, Vascular/pathology , Endothelium, Vascular/ultrastructure , Enzyme Activation , Extracellular Matrix Proteins/genetics , Extracellular Matrix Proteins/metabolism , Gene Expression Profiling/methods , Humans , Immunoblotting , Kallikreins/genetics , Kallikreins/metabolism , Mesoderm/pathology , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Microscopy, Electron, Scanning , Microscopy, Electron, Transmission , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Pancreatic Elastase/genetics , Pancreatic Elastase/metabolism , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , SOXB1 Transcription Factors/genetics , SOXB1 Transcription Factors/metabolism , Serine Endopeptidases/genetics , Twist-Related Protein 1/genetics , Twist-Related Protein 1/metabolism , Matrix Gla Protein
14.
FEBS Open Bio ; 5: 257-63, 2015.
Article in English | MEDLINE | ID: mdl-25893161

ABSTRACT

Mutations in ABCC6 (ATP-binding cassette, subfamily C, member 6), an orphan transporter expressed in the liver, are the cause of pseudoxanthoma elasticum. Since ABCC6 was reported to affect matrix Gla protein (MGP), an inhibitor of bone morphogenetic proteins (BMPs), we studied BMP signaling and expression in various tissues of mice with and without functional ABCC. Enhanced BMP signaling was found in all examined tissues in the absence of ABCC6. Despite this, the expression of particular BMP proteins varied widely between tissues. Interestingly, the expression of most BMP proteins in the liver moved in the opposite direction to the same BMP proteins in kidneys in response to ABCC6 alterations. Thus, ABCC6 deficiency stimulates BMP signaling by acting on the expression of multiple BMPs.

16.
PLoS One ; 9(8): e105988, 2014.
Article in English | MEDLINE | ID: mdl-25148248

ABSTRACT

BACKGROUND: Hypertension and diabetes are the two leading causes of chronic kidney disease (CKD) eventually leading to end stage renal disease (ESRD) and the need of renal replacement therapy. Mortality among CKD and ESRD patients is high, mostly due to cardiovascular events. New early markers of risk are necessary to better anticipate the course of the disease, to detect the renal affection of additive risk factors, and to appropriately handle patients in a pre-emptive and personalized manner. METHODS: Renal function and NGAL urinary excretion was monitored in rats with spontaneous (SHR) or L-NAME induced hypertension rendered hyperglycemic (or not as controls). RESULTS: Combination of hypertension and hyperglycemia (but not each of these factors independently) causes an increased urinary excretion of neutrophil gelatinase-associated lipocalin (NGAL) in the rat, in the absence of signs of renal damage. Increased NGAL excretion is observed in diabetic animals with two independent models of hypertension. Elevated urinary NGAL results from a specific alteration in its tubular handling, rather than from an increase in its renal expression. In fact, when kidneys of hyperglycaemic-hypertensive rats are perfused in situ with Krebs-dextran solution containing exogenous NGAL, they excrete more NGAL in the urine than hypertensive rats. We also show that albuminuria is not capable of detecting the additive effect posed by the coexistence of these two risk factors. CONCLUSIONS: Our results suggest that accumulation of hypertension and hyperglycemia induces an incipient and quite specific alteration in the tubular handling of NGAL resulting in its increased urinary excretion.


Subject(s)
Acute-Phase Proteins/urine , Diabetes Mellitus, Experimental/physiopathology , Hyperglycemia/physiopathology , Hypertension/physiopathology , Kidney Tubules/physiopathology , Lipocalins/urine , Proto-Oncogene Proteins/urine , Acute-Phase Proteins/genetics , Animals , Biomarkers/urine , Blood Pressure , Hypertension/chemically induced , Kidney Tubules/metabolism , Lipocalin-2 , Lipocalins/genetics , Male , NG-Nitroarginine Methyl Ester/toxicity , Perfusion , Proto-Oncogene Proteins/genetics , Rats, Inbred SHR , Rats, Wistar
17.
Proc Natl Acad Sci U S A ; 110(47): 19071-6, 2013 Nov 19.
Article in English | MEDLINE | ID: mdl-24191040

ABSTRACT

Cerebral arteriovenous malformations (AVMs) are common vascular malformations, which may result in hemorrhagic strokes and neurological deficits. Bone morphogenetic protein (BMP) and Notch signaling are both involved in the development of cerebral AVMs, but the cross-talk between the two signaling pathways is poorly understood. Here, we show that deficiency of matrix Gla protein (MGP), a BMP inhibitor, causes induction of Notch ligands, dysregulation of endothelial differentiation, and the development of cerebral AVMs in MGP null (Mgp(-/-)) mice. Increased BMP activity due to the lack of MGP induces expression of the activin receptor-like kinase 1, a BMP type I receptor, in cerebrovascular endothelium. Subsequent activation of activin receptor-like kinase 1 enhances expression of Notch ligands Jagged 1 and 2, which increases Notch activity and alters the expression of Ephrin B2 and Ephrin receptor B4, arterial and venous endothelial markers, respectively. Reducing the expression of Jagged 1 and 2 in the Mgp(-/-) mice by crossing them with Jagged 1 or 2 deficient mice reduces Notch activity, normalizes endothelial differentiation, and prevents cerebral AVMs, but not pulmonary or renal AVMs. Our results suggest that Notch signaling mediates and can modulate changes in BMP signaling that lead to cerebral AVMs.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Calcium-Binding Proteins/deficiency , Calcium-Binding Proteins/metabolism , Extracellular Matrix Proteins/deficiency , Guanylate Kinases/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Intracranial Arteriovenous Malformations/etiology , Intracranial Arteriovenous Malformations/prevention & control , Membrane Proteins/metabolism , Receptors, Notch/metabolism , Analysis of Variance , Animals , Bone Morphogenetic Proteins/antagonists & inhibitors , Calcium-Binding Proteins/genetics , Cell Differentiation/physiology , Endothelial Cells/physiology , Ephrin-B2/metabolism , Extracellular Matrix Proteins/genetics , Gene Deletion , Immunoblotting , Jagged-1 Protein , Jagged-2 Protein , Mice , Mice, Knockout , RNA, Small Interfering/genetics , Real-Time Polymerase Chain Reaction , Receptors, Eph Family/metabolism , Serrate-Jagged Proteins , X-Ray Microtomography , Matrix Gla Protein
18.
Biomed Res Int ; 2013: 397391, 2013.
Article in English | MEDLINE | ID: mdl-23956981

ABSTRACT

The edema formation in nephrotic syndrome (NS) is associated with a blunted response to atrial natriuretic peptide (ANP). The natriuretic effects of ANP have been related to renal dopamine D1-receptors (D1R). We examined the interaction between ANP and renal D1R in rats with puromycin aminonucleoside-induced NS (PAN-NS). Urinary sodium, cyclic guanosine monophosphate (cGMP) excretion, and D1R protein expression and localization in renal tubules were evaluated in PAN-NS and control rats before and during volume expansion (VE). The effects of zaprinast (phosphodiesterase type 5 inhibitor), alone or in combination with Sch-23390 (D1R antagonist), were examined in both groups. The increased natriuresis and urinary cGMP excretion evoked by acute VE were blunted in PAN-NS despite increased levels of circulating ANP. This was accompanied in PAN-NS by a marked decrease of D1R expression in the renal tubules. Infusion of zaprinast in PAN-NS resulted in increased urinary excretion of cGMP and sodium to similar levels of control rats and increased expression of D1R in the plasma membrane of renal tubular cells. Combined administration of Sch-23390 and zaprinast prevented natriuresis and increased cGMP excretion induced by zaprinast alone. We conclude that D1R may play a major role in the ANP resistance observed in PAN-NS.


Subject(s)
Atrial Natriuretic Factor/metabolism , Natriuresis/drug effects , Receptors, Dopamine D1/biosynthesis , Sodium/metabolism , Animals , Benzazepines/administration & dosage , Cyclic GMP/urine , Glomerular Filtration Rate , Homeostasis , Kidney/metabolism , Kidney/pathology , Male , Nephrotic Syndrome/chemically induced , Nephrotic Syndrome/drug therapy , Nephrotic Syndrome/pathology , Purinones/administration & dosage , Puromycin Aminonucleoside/toxicity , Rats , Receptors, Dopamine D1/antagonists & inhibitors , Receptors, Dopamine D1/metabolism
19.
Am J Physiol Cell Physiol ; 302(4): C686-97, 2012 Feb 15.
Article in English | MEDLINE | ID: mdl-22094331

ABSTRACT

Ras GTPases are ubiquitous plasma membrane transducers of extracellular stimuli. In addition to their role as oncogenes, Ras GTPases are key regulators of cell function. Each of the Ras isoforms exhibits specific modulatory activity on different cellular pathways. This has prompted researchers to determine the pathophysiological roles of each isoform. There is a proven relationship between the signaling pathways of transforming growth factor-ß1 (TGF-ß1) and Ras GTPases. To assess the individual role of H-Ras oncogene in basal and TGF-ß1-mediated extracellular matrix (ECM) synthesis, proliferation, and migration in fibroblasts, we analyzed these processes in embryonic fibroblasts obtained from H-Ras knockout mice (H-ras(-/-)). We found that H-ras(-/-) fibroblasts exhibited a higher basal phosphatidylinositol-3-kinase (PI3K)/Akt activation than wild-type (WT) fibroblasts, whereas MEK/ERK 1/2 activation was similar in both types of cells. Fibronectin and collagen synthesis were higher in H-ras(-/-) fibroblasts and proliferation was lower in H-ras(-/-) than in WT fibroblasts. Moreover, H-Ras appeared indispensable to maintain normal fibroblast motility, which was highly restricted in H-ras(-/-) cells. These results suggest that H-Ras (through downregulation of PI3K/Akt activation) could modulate fibroblast activity by reducing ECM synthesis and upregulating both proliferation and migration. TGF-ß1 strongly increased ERK and Akt activation in WT but not in H-ras(-/-) fibroblasts, suggesting that H-Ras is necessary to increase ERK 1/2 activation and to maintain PI3K downregulation in TGF-ß1-stimulated fibroblasts. TGF-ß1 stimulated ECM synthesis and proliferation, although ECM synthesis was higher and proliferation lower in H-ras(-/-) than in WT fibroblasts. Hence, H-Ras activation seems to play a key role in the regulation of these effects.


Subject(s)
Fibroblasts/metabolism , Gene Expression Regulation/drug effects , Oncogene Protein p21(ras)/deficiency , Protein Isoforms/deficiency , Signal Transduction/drug effects , Transforming Growth Factor beta1/pharmacology , Animals , Cell Movement , Cell Proliferation , Cells, Cultured , Collagen/genetics , Collagen/metabolism , Extracellular Matrix/genetics , Extracellular Matrix/metabolism , Fibroblasts/cytology , Fibronectins/genetics , Fibronectins/metabolism , Gene Deletion , MAP Kinase Kinase Kinases/genetics , MAP Kinase Kinase Kinases/metabolism , Mice , Mice, Knockout , Oncogene Protein p21(ras)/genetics , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation/drug effects , Protein Isoforms/genetics , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/genetics
20.
Eur J Clin Invest ; 42(5): 548-56, 2012 May.
Article in English | MEDLINE | ID: mdl-22050177

ABSTRACT

BACKGROUND: Osteoprotegerin (OPG), a secreted member of the tumour necrosis factor receptor superfamily of cytokines, has been associated with endothelial dysfunction. We studied in type 2 diabetic and/or hypertensive patients the relationship between serum OPG and vascular alterations associated with these pathologies. MATERIALS AND METHODS: We analysed 191 consecutive patients (52 with type 2 diabetes and 139 hypertensive nondiabetic patients) and 54 healthy controls. We assessed the relationship of OPG serum levels measured by ELISA with basal glycaemia, glycosylated haemoglobin, blood pressure, endothelial dysfunction (assessed by pulse wave velocity), retinopathy (by Keith-Wagener classification), left ventricular hypertrophy (by Cornell index), cardiovascular risk and target organs (heart, vascular, kidney) damage. RESULTS: Serum OPG levels were higher in either hypertensive or diabetic patients and in patients with non-dipper and riser circadian blood pressure patterns. We found significant correlations between OPG levels and age, height, glycaemia, systolic, diastolic and pulse blood pressure, pulse wave velocity and left ventricular hypertrophy in both hypertensive and diabetic patients. OPG levels were also higher in hypertensive patients with retinopathy, patients with high probability of 10-year cardiovascular risk, patients with three or more damaged target organs (heart, vessels, kidneys) and patients with previous episodes of ischaemic cardiopathy or hypercholesterolaemia. CONCLUSIONS: Osteoprotegerin is an indicator of diabetes- and hypertension-associated vascular pathologies as endothelial dysfunction and cardiovascular risk.


Subject(s)
Cardiovascular Diseases/blood , Diabetes Mellitus, Type 2/blood , Hypertension/blood , Osteoprotegerin/blood , Adult , Aged , Biomarkers/blood , Blood Pressure , Case-Control Studies , Cross-Sectional Studies , Enzyme-Linked Immunosorbent Assay , Female , Humans , Male , Middle Aged , Risk Factors , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...