Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 724
Filter
Add more filters

Country/Region as subject
Publication year range
1.
BMC Cardiovasc Disord ; 24(1): 11, 2024 01 02.
Article in English | MEDLINE | ID: mdl-38166688

ABSTRACT

BACKGROUND: Endothelial dysfunction is characterized by an imbalance between endothelium-derived vasodilatory and vasoconstrictive effects and may play an important role in the development of heart failure. An increasing number of studies have shown that endothelial-derived NO-mediated vasodilation is attenuated in heart failure patients. However, the role of endothelin-1 (ET-1) in heart failure remains controversial due to its different receptors including ET-1 receptor type A (ETAR) and ET-1 receptor type B (ETBR). The aim of this study was to determine whether ET-1 and its receptors are activated and to explore the role of ETAR and ETBR in heart failure induced by myocarditis. METHODS: We constructed an animal model of experimental autoimmune myocarditis (EAM) with porcine cardiac myosin. Twenty rats were randomized to the control group (3 weeks, n = 5), the extended control group (8 weeks, n = 5), the EAM group (3 weeks, n = 5), the extended EAM group (8 weeks, n = 5). HE staining was used to detect myocardial inflammatory infiltration and the myocarditis score, Masson's trichrome staining was used to assess myocardial fibrosis, echocardiography was used to evaluate cardiac function, ELISA was used to detect serum NT-proBNP and ET-1 concentrations, and immunohistochemistry and western blotting were used to detect ETAR and ETBR expression in myocardial tissue of EAM-induced heart failure. Subsequently, a model of myocardial inflammatory injury in vitro was constructed to explore the role of ETAR and ETBR in EAM-induced heart failure. RESULTS: EAM rats tended to reach peak inflammation after 3 weeks of immunization and developed stable chronic heart failure at 8 weeks after immunization. LVEDd and LVEDs were significantly increased in the EAM group compared to the control group at 3 weeks and 8 weeks after immunization while EF and FS were significantly reduced. Serum NT-proBNP concentrations in EAM (both 3 weeks and 8 weeks) were elevated. Therefore, EAM can induce acute and chronic heart failure due to myocardial inflammatory injury. Serum ET-1 concentration and myocardial ETAR and ETBR protein were significantly increased in EAM-induced heart failure in vivo. Consistent with the results of the experiments in vivo, ETAR and ETBR protein expression levels were significantly increased in the myocardial inflammatory injury model in vitro. Moreover, ETAR gene silencing inhibited inflammatory cytokine TNF-α and IL-1ß levels, while ETBR gene silencing improved TNF-α and IL-1ß levels. CONCLUSIONS: ET-1, ETAR, and ETBR were activated in both EAM-induced acute heart failure and chronic heart failure. ETAR may positively regulate EAM-induced heart failure by promoting myocardial inflammatory injury, whereas ETBR negatively regulates EAM-induced heart failure by alleviating myocardial inflammatory injury.


Subject(s)
Autoimmune Diseases , Heart Failure , Heart Injuries , Myocarditis , Receptor, Endothelin A , Receptor, Endothelin B , Animals , Rats , Heart Failure/etiology , Heart Failure/metabolism , Myocarditis/chemically induced , Myocardium/metabolism , Swine , Tumor Necrosis Factor-alpha/metabolism , Receptor, Endothelin A/metabolism , Receptor, Endothelin B/metabolism
2.
Am J Physiol Renal Physiol ; 325(2): F235-F247, 2023 08 01.
Article in English | MEDLINE | ID: mdl-37348026

ABSTRACT

Renal nerves have been an attractive target for interventions aimed at lowering blood pressure; however, the specific roles of renal afferent (sensory) versus efferent sympathetic nerves in mediating hypertension are poorly characterized. A number of studies have suggested that a sympathoexcitatory signal conveyed by renal afferents elicits increases in blood pressure, whereas other studies identified sympathoinhibitory afferent pathways. These sympathoinhibitory pathways have been identified as protective against salt-sensitive increases in blood pressure through endothelin B (ETB) receptor activation. We hypothesized that ETB-deficient (ETB-def) rats, which are devoid of functional ETB receptors except in adrenergic tissues, lack appropriate sympathoinhibition and have lower renal afferent nerve activity following a high-salt diet compared with transgenic controls. We found that isolated renal pelvises from high salt-fed ETB-def animals lack a response to a physiological stimulus, prostaglandin E2, compared with transgenic controls but respond equally to a noxious stimulus, capsaicin. Surprisingly, we observed elevated renal afferent nerve activity in intact ETB-def rats compared with transgenic controls under both normal- and high-salt diets. ETB-def rats have been previously shown to have heightened global sympathetic tone, and we also observed higher total renal sympathetic nerve activity in ETB-def rats compared with transgenic controls under both normal- and high-salt diets. These data indicate that ETB receptors are integral mediators of the sympathoinhibitory renal afferent reflex (renorenal reflex), and, in a genetic rat model of ETB deficiency, the preponderance of sympathoexcitatory renal afferent nerve activity prevails and may contribute to hypertension.NEW & NOTEWORTHY Here, we found that endothelin B receptors are an important contributor to renal afferent nerve responsiveness to a high-salt diet. Rats lacking endothelin B receptors have increased afferent nerve activity that is not responsive to a high-salt diet.


Subject(s)
Hypertension , Kidney , Rats , Animals , Receptor, Endothelin B/genetics , Receptor, Endothelin B/metabolism , Kidney/metabolism , Blood Pressure/physiology , Afferent Pathways/metabolism , Sodium Chloride, Dietary/metabolism , Endothelin-1/metabolism , Receptor, Endothelin A/metabolism
3.
FASEB J ; 36(12): e22638, 2022 12.
Article in English | MEDLINE | ID: mdl-36331552

ABSTRACT

Painful crises in sickle cell disease (SCD) are associated with increased plasma cytokines levels, including endothelin-1 (ET-1). Reduced red cell magnesium content, mediated in part by increased Na+ /Mg2+ exchanger (NME) activity, contributes to erythrocyte K+ loss, dehydration and sickling in SCD. However, the relationship between ET-1 and the NME in SCD has remained unexamined. We observed increased NME activity in sickle red cells incubated in the presence of 500 nM ET-1. Deoxygenation of sickle red cells, in contrast, led to decreased red cell NME activity and cellular dehydration that was reversed by the NME inhibitor, imipramine. Increased NME activity in sickle red cells was significantly blocked by pre-incubation with 100 nM BQ788, a selective blocker of ET-1 type B receptors. These results suggest an important role for ET-1 and for cellular magnesium homeostasis in SCD. Consistent with these results, we observed increased NME activity in sickle red cells of three mouse models of sickle cell disease greater than that in red cells of C57BL/J6 mice. In vivo treatment of BERK sickle transgenic mice with ET-1 receptor antagonists reduced red cell NME activity. Our results suggest that ET-1 receptor blockade may be a promising therapeutic approach to control erythrocyte volume and magnesium homeostasis in SCD and may thus attenuate or retard the associated chronic inflammatory and vascular complications of SCD.


Subject(s)
Anemia, Sickle Cell , Endothelin-1 , Mice , Animals , Endothelin-1/metabolism , Magnesium/metabolism , Dehydration/metabolism , Mice, Inbred C57BL , Erythrocytes/metabolism , Anemia, Sickle Cell/drug therapy , Anemia, Sickle Cell/metabolism , Sodium/metabolism , Homeostasis , Receptor, Endothelin B/metabolism , Mice, Transgenic
4.
Chembiochem ; 23(6): e202100340, 2022 03 18.
Article in English | MEDLINE | ID: mdl-34699123

ABSTRACT

Fine-tuning of G protein-coupled receptor (GPCR) signaling is important to maintain cellular homeostasis. Recent studies demonstrated that lateral GPCR interactions in the cell membrane can impact signaling profiles. Here, we report on a one-step labeling method of multiple membrane-embedded GPCRs. Based on short peptide tags, complementary probes transfer the cargo (e. g. a fluorescent dye) by an acyl transfer reaction with high spatial and temporal resolution within 5 min. We applied this approach to four receptors of the cardiovascular system: the endothelin receptor A and B (ETA R and ETB R), angiotensin II receptor type 1, and apelin. Wild type-like G protein activation after N-terminal modification was demonstrated for all receptor species. Using FRET-competent dyes, a constitutive proximity between hetero-receptors was limited to ETA R/ETB R. Further, we demonstrate, that ETA R expression regulates the signaling of co-expressed ETB R. Our orthogonal peptide-templated labeling of different GPCRs provides novel insight into the regulation of GPCR signaling.


Subject(s)
GTP-Binding Proteins , Signal Transduction , GTP-Binding Proteins/metabolism , Peptides/metabolism , Receptor, Endothelin A/metabolism , Receptor, Endothelin B/metabolism , Signal Transduction/physiology
5.
Nature ; 537(7620): 363-368, 2016 09 15.
Article in English | MEDLINE | ID: mdl-27595334

ABSTRACT

Endothelin, a 21-amino-acid peptide, participates in various physiological processes, such as regulation of vascular tone, humoral homeostasis, neural crest cell development and neurotransmission. Endothelin and its G-protein-coupled receptor are involved in the development of various diseases, such as pulmonary arterial hypertension, and thus are important therapeutic targets. Here we report crystal structures of human endothelin type B receptor in the ligand-free form and in complex with the endogenous agonist endothelin-1. The structures and mutation analysis reveal the mechanism for the isopeptide selectivity between endothelin-1 and -3. Transmembrane helices 1, 2, 6 and 7 move and envelop the entire endothelin peptide, in a virtually irreversible manner. The agonist-induced conformational changes are propagated to the receptor core and the cytoplasmic G-protein coupling interface, and probably induce conformational flexibility in TM6. A comparison with the M2 muscarinic receptor suggests a shared mechanism for signal transduction in class A G-protein-coupled receptors.


Subject(s)
Endothelin-1/metabolism , Receptor, Endothelin B/chemistry , Receptor, Endothelin B/metabolism , Allosteric Regulation , Allosteric Site , Cell Membrane/metabolism , Crystallography, X-Ray , Endothelin-1/chemistry , Endothelin-1/pharmacology , Endothelin-3/chemistry , Endothelin-3/metabolism , Humans , Ligands , Models, Molecular , Protein Conformation , Receptor, Endothelin B/agonists , Receptor, Endothelin B/genetics , Receptor, Muscarinic M2/chemistry , Receptor, Muscarinic M2/metabolism , Signal Transduction , Substrate Specificity
6.
Nature ; 531(7592): 105-9, 2016 Mar 03.
Article in English | MEDLINE | ID: mdl-26863197

ABSTRACT

The enteric nervous system (ENS) is the largest component of the autonomic nervous system, with neuron numbers surpassing those present in the spinal cord. The ENS has been called the 'second brain' given its autonomy, remarkable neurotransmitter diversity and complex cytoarchitecture. Defects in ENS development are responsible for many human disorders including Hirschsprung disease (HSCR). HSCR is caused by the developmental failure of ENS progenitors to migrate into the gastrointestinal tract, particularly the distal colon. Human ENS development remains poorly understood owing to the lack of an easily accessible model system. Here we demonstrate the efficient derivation and isolation of ENS progenitors from human pluripotent stem (PS) cells, and their further differentiation into functional enteric neurons. ENS precursors derived in vitro are capable of targeted migration in the developing chick embryo and extensive colonization of the adult mouse colon. The in vivo engraftment and migration of human PS-cell-derived ENS precursors rescue disease-related mortality in HSCR mice (Ednrb(s-l/s-l)), although the mechanism of action remains unclear. Finally, EDNRB-null mutant ENS precursors enable modelling of HSCR-related migration defects, and the identification of pepstatin A as a candidate therapeutic target. Our study establishes the first, to our knowledge, human PS-cell-based platform for the study of human ENS development, and presents cell- and drug-based strategies for the treatment of HSCR.


Subject(s)
Cell Lineage , Cell- and Tissue-Based Therapy , Drug Discovery/methods , Enteric Nervous System/pathology , Hirschsprung Disease/drug therapy , Hirschsprung Disease/pathology , Neurons/pathology , Aging , Animals , Cell Differentiation , Cell Line , Cell Movement , Cell Separation , Cell- and Tissue-Based Therapy/methods , Chick Embryo , Colon/drug effects , Colon/pathology , Disease Models, Animal , Female , Gastrointestinal Tract/drug effects , Gastrointestinal Tract/pathology , Hirschsprung Disease/therapy , Humans , Male , Mice , Neurons/drug effects , Pepstatins/metabolism , Pluripotent Stem Cells/pathology , Receptor, Endothelin B/metabolism , Signal Transduction
7.
Can J Physiol Pharmacol ; 100(7): 679-688, 2022 Jul 01.
Article in English | MEDLINE | ID: mdl-35442801

ABSTRACT

Endothelin-1 (ET-1), the most potent vasoconstrictor identified to date, contributes to cerebrovascular dysfunction and brain ET-1 levels were shown to be related to Alzheimer's disease and related dementias (ADRD) progression. ET-1 also contributes to neuroinflammation, especially in infections of the central nervous system. Recent studies causally linked chronic periodontal infection with an opportunistic anaerobic bacterium Porphyromonas gingivalis (Coykendall et al.) Shah & Collins to AD development. Thus, the goal of the study was to determine the impact of P. gingivalis infection on the ET system and cell senescence in brain microvascular endothelial cells. Cells were infected with a multiplicity of infection 50 P. gingivalis with and without extracellular ATP-induced oxidative stress for 24 h. Cell lysates were collected for analysis of endothelin A receptor (ETA)/endothelin B receptor (ETB) receptor as well as senescence markers. ET-1 levels in cell culture media were measured with enzyme-linked immunosorbent assay. P. gingivalis infection increased ET-1 (pg/mL) secretion, as well as the ETA receptor expression, whereas decreased lamin A/C expression compared to control. Tight junction protein claudin-5 was also decreased under these conditions. ETA or ETB receptor blockade during infection did not affect ET-1 secretion or the expression of cell senescence markers. Current findings suggest that P. gingivalis infection may compromise endothelial integrity and activate the ET system.


Subject(s)
Bacteroidaceae Infections , Endothelial Cells , Porphyromonas gingivalis , Bacteroidaceae Infections/metabolism , Base Composition , Brain/metabolism , Endothelial Cells/metabolism , Endothelial Cells/microbiology , Endothelin-1/metabolism , Endothelins , Phylogeny , Porphyromonas gingivalis/metabolism , RNA, Ribosomal, 16S , Receptor, Endothelin A/metabolism , Receptor, Endothelin B/metabolism , Receptors, Endothelin/metabolism , Sequence Analysis, DNA
8.
Cell Mol Life Sci ; 78(16): 6033-6049, 2021 Aug.
Article in English | MEDLINE | ID: mdl-34274976

ABSTRACT

Melanocytes are pigmented cells residing mostly in the skin and hair follicles of vertebrates, where they contribute to colouration and protection against UV-B radiation. However, the spectrum of their functions reaches far beyond that. For instance, these pigment-producing cells are found inside the inner ear, where they contribute to the hearing function, and in the heart, where they are involved in the electrical conductivity and support the stiffness of cardiac valves. The embryonic origin of such extracutaneous melanocytes is not clear. We took advantage of lineage-tracing experiments combined with 3D visualizations and gene knockout strategies to address this long-standing question. We revealed that Schwann cell precursors are recruited from the local innervation during embryonic development and give rise to extracutaneous melanocytes in the heart, brain meninges, inner ear, and other locations. In embryos with a knockout of the EdnrB receptor, a condition imitating Waardenburg syndrome, we observed only nerve-associated melanoblasts, which failed to detach from the nerves and to enter the inner ear. Finally, we looked into the evolutionary aspects of extracutaneous melanocytes and found that pigment cells are associated mainly with nerves and blood vessels in amphibians and fish. This new knowledge of the nerve-dependent origin of extracutaneous pigment cells might be directly relevant to the formation of extracutaneous melanoma in humans.


Subject(s)
Brain/physiology , Ear, Inner/physiology , Heart/physiology , Meninges/physiology , Nervous System/physiopathology , Schwann Cells/physiology , Amphibians/metabolism , Amphibians/physiology , Animals , Brain/metabolism , Cell Lineage/physiology , Ear, Inner/metabolism , Embryonic Development/physiology , Female , Fishes/metabolism , Fishes/physiology , Melanocytes/metabolism , Melanocytes/physiology , Meninges/metabolism , Mice , Nervous System/metabolism , Pregnancy , Receptor, Endothelin B/metabolism , Schwann Cells/metabolism
9.
Pflugers Arch ; 473(10): 1667-1683, 2021 10.
Article in English | MEDLINE | ID: mdl-34355294

ABSTRACT

Renal interstitial fibrosis is characterized by the development of myofibroblasts, originating from resident renal and immigrating cells. Myofibroblast formation and extracellular matrix production during kidney damage are triggered by various factors. Among these, endothelins have been discussed as potential modulators of renal fibrosis. Utilizing mouse models of adenine nephropathy (AN) and unilateral ureter occlusion (UUO), this study aimed to investigate the contribution of endothelin signaling in stromal mesenchymal resident renal interstitial cells. We found in controls that adenine feeding and UUO caused marked upregulations of endothelin-1 (ET-1) gene expression in endothelial and in tubular cells and a strong upregulation of ETA-receptor (ETA-R) gene expression in interstitial and mesangial cells, while the gene expression of ETB-receptor (ETB-R) did not change. Conditional deletion of ETA-R and ETB-R gene expression in the FoxD1 stromal cell compartment which includes interstitial cells significantly reduced renal ETA-R gene expression and moderately lowered renal ETB-R gene expression. ET receptor (ET-R) deletion exerted no apparent effects on kidney development nor on kidney function. Adenine feeding and UUO led to similar increases in profibrotic and proinflammatory gene expression in control as well as in ETAflflETBflfl FoxD1Cre+ mice (ET-Ko). In summary, our findings suggest that adenine feeding and UUO activate endothelin signaling in interstitial cells which is due to upregulated ETA-R expression and enhanced renal ET-1 production Our data also suggest that the activation of endothelin signaling in interstitial cells has less impact for the development of experimentally induced fibrosis.


Subject(s)
Adenine/toxicity , Fibrosis/physiopathology , Kidney Diseases/etiology , Kidney/cytology , Receptor, Endothelin A/metabolism , Receptor, Endothelin B/metabolism , Animals , Fibrosis/metabolism , Gene Deletion , Gene Expression Regulation , Kidney Diseases/metabolism , Mice , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptor, Endothelin A/genetics , Receptor, Endothelin B/genetics , Up-Regulation , Ureteral Obstruction
10.
Am J Physiol Renal Physiol ; 320(3): F297-F307, 2021 03 01.
Article in English | MEDLINE | ID: mdl-33356953

ABSTRACT

We reported that high salt (HS) intake stimulates renal collecting duct (CD) endothelin (ET) type B receptor (ETBR)/nitric oxide (NO) synthase 1ß (NOS1ß)-dependent NO production inhibiting the epithelial sodium channel (ENaC) promoting natriuresis. However, the mechanism underlying the HS-induced increase of NO production is unclear. Histone deacetylase 1 (HDAC1) responds to increased fluid flow, as can occur in the CD during HS intake. The renal inner medulla (IM), in particular the IMCD, has the highest NOS1 activity within the kidney. Hence, we hypothesized that HS intake provokes HDAC1 activation of NO production in the IM. HS intake for 1 wk significantly increased HDAC1 abundance in the IM. Ex vivo treatment of dissociated IM from HS-fed mice with a selective HDAC1 inhibitor (MS-275) decreased NO production with no change in ET-1 peptide or mRNA levels. We further investigated the role of the ET-1/ETBR/NOS1ß signaling pathway with chronic ETBR blockade (A-192621). Although NO was decreased and ET-1 levels were elevated in the dissociated IM from HS-fed mice treated with A-192621, ex vivo MS-275 did not further change NO or ET-1 levels suggesting that HDAC1-mediated NO production is regulated at the level or downstream of ETBR activation. In split-open CDs from HS-fed mice, patch clamp analysis revealed significantly higher ENaC activity after MS-275 pretreatment, which was abrogated by an exogenous NO donor. Moreover, flow-induced increases in mIMCD-3 cell NO production were blunted by HDAC1 or calcium inhibition. Taken together, these findings indicate that HS intake induces HDAC1-dependent activation of the ETBR/NO pathway contributing to the natriuretic response.


Subject(s)
Histone Deacetylase 1/metabolism , Kidney Tubules, Collecting/enzymology , Natriuresis , Nitric Oxide/metabolism , Renal Elimination , Sodium Chloride, Dietary/administration & dosage , Animals , Endothelin-1/metabolism , Male , Mice, Inbred C57BL , Nitric Oxide Synthase Type I/metabolism , Receptor, Endothelin B/metabolism , Signal Transduction , Sodium Chloride, Dietary/urine
11.
Am J Physiol Heart Circ Physiol ; 321(3): H592-H598, 2021 09 01.
Article in English | MEDLINE | ID: mdl-34415188

ABSTRACT

The endothelin-B (ETB) receptor is a key regulator of vascular endothelial function in women. We have previously shown that the ETB receptor mediates vasodilation in young women, an effect that is lost after menopause. However, the direct impact of changes in estradiol (E2) on ETB receptor function in women remains unclear. Therefore, the purpose of this study was to test the hypothesis that E2 exposure modulates ETB receptor-mediated dilation in young women. Fifteen young women (24 ± 4 yr, 24 ± 3 kg/m2) completed the study. Endogenous sex hormone production was suppressed with daily administration of a gonadotropin-releasing hormone antagonist (GnRHant; Ganirelix) for 10 days; E2 (0.1 mg/day, Vivelle-Dot patch) was added back on days 4-10. We measured vasodilation in the cutaneous microcirculation (microvascular endothelial function) via local heating (42°C) on day 4 (GnRHant) and day 10 (GnRHant + E2) using laser Doppler flowmetry coupled with intradermal microdialysis during perfusions of lactated Ringer's (control) and ETB receptor antagonist (BQ-788, 300 nM). During GnRHant, vasodilatory responses to local heating were enhanced with ETB receptor blockade (control: 83 ± 9 vs. BQ-788: 90 ± 5%CVCmax, P = 0.004). E2 administration improved vasodilation in the control site (GnRHant: 83 ± 9 vs. GnRHant + E2: 89 ± 8%CVCmax, P = 0.036). Furthermore, cutaneous vasodilatory responses during ETB receptor blockade were blunted after E2 administration (control: 89 ± 8 vs. BQ-788: 84 ± 8%CVCmax, P = 0.047). These data demonstrate that ovarian hormones, specifically E2, modulate ETB receptor function and contribute to the regulation of microvascular endothelial function in young women.NEW & NOTEWORTHY The endothelin-B (ETB) receptor mediates vasodilation in young women, an effect lost following menopause. It is unclear whether these alterations are due to aging or changes in estradiol (E2). During endogenous hormone suppression (GnRH antagonist), blockade of ETB receptors enhanced cutaneous microvascular vasodilation. However, during E2 administration, blockade of ETB receptors attenuated vasodilation, indicating that the ETB receptor mediates dilation in the presence of E2. In young women, ETB receptors mediate vasodilation in the presence of E2, an effect that is lost when E2 is suppressed.


Subject(s)
Endothelin B Receptor Antagonists/pharmacology , Estradiol/pharmacology , Estrogens/pharmacology , Receptor, Endothelin B/metabolism , Vasodilation , Adult , Female , Gonadotropin-Releasing Hormone/analogs & derivatives , Gonadotropin-Releasing Hormone/pharmacology , Hormone Antagonists/pharmacology , Humans , Microvessels/drug effects , Microvessels/metabolism , Microvessels/physiology , Oligopeptides/pharmacology , Piperidines/pharmacology , Skin/blood supply
12.
J Pharmacol Exp Ther ; 376(1): 98-105, 2021 01.
Article in English | MEDLINE | ID: mdl-33127751

ABSTRACT

The G protein-coupled estrogen receptor 1 (GPER1) mediates rapid estrogenic signaling. We recently reported that activation of GPER1 in the renal medulla evokes endothelin-1-dependent natriuresis in female, but not male, rats. However, the involvement of the ET receptors, ETA and ETB, underlying GPER1 natriuretic action remain unclear. In this study, we used genetic and pharmacologic methods to identify the contributions of ETA and ETB in mediating this female-specific natriuretic effect of renal medullary GPER1. Infusion of the GPER1-selective agonist G1 (5 pmol/kg per minute) into the renal medulla for 40 minutes increased Na+ excretion and urine flow in anesthetized female ETB-deficient (ETB def) rats and littermate controls but did not affect blood pressure or urinary K+ excretion in either group. Pretreatment with the selective ETA inhibitor ABT-627 (5 mg/kg, intravenous) abolished G1-induced natriuresis in ETB def rats. To further isolate the effects of inhibiting either receptor alone, we conducted the same experiments in anesthetized female Sprague-Dawley (SD) rats pretreated or not with ABT-627 and/or the selective ETB inhibitor A-192621 (10 mg/kg, intravenous). Neither antagonism of ETA nor antagonism of ETB receptor alone affected the G1-induced increase in Na+ excretion and urine flow in SD rats. However, simultaneous antagonism of both receptors completely abolished these effects. These data suggest that ETA and ETB receptors can mediate the natriuretic and diuretic response to renal medullary GPER1 activation in female rats. SIGNIFICANCE STATEMENT: Activation of G protein-coupled estrogen receptor 1 (GPER1) in the renal medulla of female rats evokes natriuresis via endothelin receptors A and/or B, suggesting that GPER1 and endothelin signaling pathways help efficient sodium excretion in females. Thus, GPER1 activation could be potentially useful to mitigate salt sensitivity in females.


Subject(s)
Natriuresis , Receptor, Endothelin A/metabolism , Receptor, Endothelin B/metabolism , Receptors, G-Protein-Coupled/metabolism , Animals , Atrasentan/pharmacology , Endothelin Receptor Antagonists/pharmacology , Female , Kidney Medulla/drug effects , Kidney Medulla/metabolism , Pyrrolidines/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, G-Protein-Coupled/agonists
13.
Toxicol Appl Pharmacol ; 414: 115420, 2021 03 01.
Article in English | MEDLINE | ID: mdl-33503445

ABSTRACT

As a VEGF-targeting agent, sorafenib has been used to treat a number of solid tumors but can easily lead to adverse vascular effects. To elucidate the underlying mechanism, rat mesenteric arteries were subjected to organ cultured in the presence of different concentrations of sorafenib (0, 3, 6 and 9 mg/L) with or without inhibitors (U0126, 10-5 M; SB203580, 10-5 M; SP200126, 10-5 M) of MAPK kinases, and then acetylcholine- or sodium nitroprusside-induced vasodilation and sarafotoxin 6c-induced vasoconstriction were monitored by a sensitive myograph. The NO synthetases, the nitrite levels, the endothelial marker CD31,the ETB and ETA receptors and the phosphorylation of MAPK kinases were studied. Next, rats were orally administrated by sorafenib for 4 weeks (7.5 and 15 mg/kg/day), and their blood pressure, plasma ET-1, the ETB and ETA receptors and the phosphorylation of MAPK kinases in the mesenteric arteries were investigated. The results showed that sorafenib impairs endothelium-dependent vasodilation due to decreased NO levels and the low expression of eNOS and iNOS. Weak staining for CD31 indicated that sorafenib induced endothelial damage. Moreover, sorafenib caused the upregulation of vasoconstrictive ETB receptors, the enhancement of ETB receptor-mediated vasoconstriction and the activation of JNK/MAPK. Blocking the JNK, ERK1/2 and p38/MAPK signaling pathways by using the inhibitors significantly abolished ETB receptor-mediated vasoconstriction. Furthermore, it was observed that the oral administration of sorafenib caused an increase in blood pressure and plasma ET-1, upregulation of the ETB receptor and the activation of JNK in the mesenteric arteries. In conclusion, sorafenib not only impairs endothelium-dependent vasodilatation but also enhances ETB receptor-mediated vasoconstriction, which may be the causal factors for hypertension and other adverse vascular effects in patients treated with sorafenib.


Subject(s)
Angiogenesis Inhibitors/toxicity , Endothelium, Vascular/drug effects , Hypertension/chemically induced , Mesenteric Artery, Superior/drug effects , Receptor, Endothelin B/metabolism , Sorafenib/toxicity , Vasoconstriction/drug effects , Vasodilation/drug effects , Animals , Blood Pressure/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Endothelium, Vascular/metabolism , Endothelium, Vascular/physiopathology , Extracellular Signal-Regulated MAP Kinases/metabolism , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Hypertension/metabolism , Hypertension/physiopathology , JNK Mitogen-Activated Protein Kinases/metabolism , Male , Mesenteric Artery, Superior/metabolism , Mesenteric Artery, Superior/physiopathology , Nitric Oxide/metabolism , Rats, Sprague-Dawley , Receptor, Endothelin B/genetics , Signal Transduction , Tissue Culture Techniques , Up-Regulation , p38 Mitogen-Activated Protein Kinases/metabolism
14.
FASEB J ; 34(9): 11594-11604, 2020 09.
Article in English | MEDLINE | ID: mdl-32687659

ABSTRACT

The endothelin receptor type B (ETBR) regulates water and electrolyte balance and blood pressure, in part, by inhibiting renal sodium transport. Our preliminary study found that the ETBR-mediated diuresis and natriuresis are impaired in hypertension with unknown mechanism. Persistently increased activity of G protein-coupled receptor kinase 4 (GRK4), caused by increased expression or genetic variants (eg, GRKγ142V), impairs the ability of the kidney to excrete a sodium load, in part, by impairing renal dopamine D1 receptor function through persistent phosphorylation. Our present study found that although renal ETBR expression was not different between Wistar-Kyoto (WKY) and spontaneously hypertensive rats (SHRs), renal ETBR phosphorylation was higher in SHRs. The role of hyper-phosphorylation in impaired ETBR-function was supported by results in human (h) GRK4γ transgenic mice. Stimulation of ETBR by BQ3020-induced natriuresis in human (h) GRK4γ wild-type (WT) mice. However, in hGRK4γ 142V transgenic mice, the renal ETBR was hyperphosphorylated and ETBR-mediated natriuresis and diuresis were not evident. There were co-localization and co-immunoprecipitation of ETBR and GRK4 in renal proximal tubule (RPT) cells from both WKY and SHRs but was greater in the latter than the former group. SiRNA-mediated downregulation of GRK4 expression, recovered the impaired inhibitory effect of ETBR on Na+ -K+ -ATPase activity in RPT cells from SHR. In vivo downregulation of renal GRK4 expression, via ultrasound-targeted microbubble destruction, decreased ETBR phosphorylation and restored ETBR-mediated natriuresis and diuresis in SHRs. This study provides a mechanism by which GRK4, via regulation of renal ETBR function, participates in the pathogenesis of hypertension.


Subject(s)
G-Protein-Coupled Receptor Kinase 4/metabolism , Hypertension/metabolism , Kidney/metabolism , Receptor, Endothelin B/metabolism , Animals , Cells, Cultured , Female , G-Protein-Coupled Receptor Kinase 4/genetics , Hypertension/genetics , Kidney Tubules, Proximal/metabolism , Male , Mice, Transgenic , Phosphorylation , Rats , Rats, Inbred SHR , Rats, Inbred WKY , Receptor, Endothelin B/genetics , Sodium/metabolism , Species Specificity
15.
FASEB J ; 34(8): 10931-10947, 2020 08.
Article in English | MEDLINE | ID: mdl-32592286

ABSTRACT

Enteric nervous system (ENS) development is governed by interactions between neural crest cells (NCC) and the extracellular matrix (ECM). Hirschsprung disease (HSCR) results from incomplete NCC migration and failure to form an appropriate ENS. Prior studies implicate abnormal ECM in NCC migration failure. We performed a comparative microarray of the embryonic distal hindgut of wild-type and EdnrBNCC-/- mice that model HSCR and identified laminin-ß1 as upregulated in EdnrBNCC-/- colon. We identified decreased expression of 37/67 kDa laminin receptor (LAMR), which binds laminin-ß1, in human HSCR myenteric plexus and EdnrBNCC-/- NCC. Using a combination of in vitro gut slice cultures and ex vivo organ cultures, we determined the mechanistic role of LAMR in NCC migration. We found that enteric NCC express LAMR, which is downregulated in human and murine HSCR. Binding of LAMR by the laminin-ß1 analog YIGSR promotes NCC migration. Silencing of LAMR abrogated these effects. Finally, applying YIGSR to E13.5 EdnrBNCC-/- colon explants resulted in 80%-100% colonization of the hindgut. This study adds LAMR to the large list of receptors through which NCC interact with their environment during ENS development. These results should be used to inform ongoing integrative, regenerative medicine approaches to HSCR.


Subject(s)
Cell Movement/physiology , Enteric Nervous System/growth & development , Enteric Nervous System/metabolism , Neural Crest/metabolism , Receptors, Laminin/metabolism , Animals , Colon/metabolism , Colon/physiology , Down-Regulation/physiology , Enteric Nervous System/physiology , Hirschsprung Disease/metabolism , Hirschsprung Disease/physiopathology , Humans , Laminin/metabolism , Mice , Mice, Knockout , Neural Crest/physiology , Organogenesis/physiology , Receptor, Endothelin B/metabolism , Up-Regulation/physiology
16.
J Cardiovasc Pharmacol ; 77(6): 699-706, 2021 06 01.
Article in English | MEDLINE | ID: mdl-34001723

ABSTRACT

ABSTRACT: Treatment-resistant hypertension (TRH) is associated with increased cardiovascular risks and progression of chronic kidney disease. The pathophysiology of TRH is multifactorial, including overactivity of the renin-angiotensin-aldosterone system and sympathetic nervous system, endothelial dysfunction, and volume overload. Endothelin-1 is a vasoconstrictive peptide that causes neurohormonal and sympathetic activation, increased aldosterone synthesis and secretion, endothelial dysfunction, vascular hypertrophy and remodeling, and fibrosis. Endothelin-1 acts through 2 receptors, ETA and ETB. Activation of ETA receptors in vascular smooth muscle cells results in vasoconstriction, whereas ETB receptor activation results in vasoconstriction in the vascular smooth muscle cells and vasodilation through nitric oxide release in endothelial cells. Aprocitentan is novel, oral, dual endothelin-receptor antagonist that has demonstrated a more favorable tolerability and safety profile in early clinical trials compared with other endothelin-receptor antagonists studied. Phase 2 trial data support a significant reduction in blood pressure compared to placebo and similar blood pressure reduction compared to a moderately dosed angiotensin-converting enzyme inhibitor in patients with essential hypertension. An ongoing phase 3 randomized clinical trial is evaluating aprocitentan's efficacy and safety in patients with TRH receiving multiple antihypertensives. Additional research is needed to determine aprocitentan's role in therapy, but this agent may be a suitable treatment option for TRH.


Subject(s)
Endothelin Receptor Antagonists/pharmacology , Hypertension/drug therapy , Pyrimidines/pharmacology , Sulfonamides/pharmacology , Animals , Antihypertensive Agents/adverse effects , Antihypertensive Agents/pharmacology , Drug Resistance , Endothelin Receptor Antagonists/adverse effects , Humans , Hypertension/physiopathology , Pyrimidines/adverse effects , Randomized Controlled Trials as Topic , Receptor, Endothelin A/drug effects , Receptor, Endothelin A/metabolism , Receptor, Endothelin B/drug effects , Receptor, Endothelin B/metabolism , Sulfonamides/adverse effects
17.
BMC Cardiovasc Disord ; 21(1): 475, 2021 10 02.
Article in English | MEDLINE | ID: mdl-34600481

ABSTRACT

BACKGROUND: HSCR, a colonic neurocristopathy affecting 1/5000 births, is suggested to associate with cardiac septal defects and conotruncal malformations. However, we question subtle cardiac changes maybe more commonly present due to multi-regulations by HSCR candidate genes, in this instance, ETB. To investigate, we compared the cardiac morphology and quantitative measurements of sl/sl rat to those of the control group. METHODS: Eleven neonatal rats were generated from heterozygote (ETB+/-) crossbreeding. Age and bodyweight were recorded at time of sacrifice. Diffusion-staining protocols with 1.5% iodine solution was completed prior to micro-CT scanning. All rats were scanned using an in vivo micro-CT scanner, Caliper Quantum FX, followed by two quality-control scans using a custom-built ex vivo micro-CT system. All scans were reviewed for gross cardiac dysmorphology. Micro-CT data were segmented semi-automatically post-NLM filtering for: whole-heart, LV, RV, LA, RA, and aortic arch. Measurements were taken with Drishti. Following image analysis, PCR genotyping of rats was performed: five sl/sl rats, three wildtype, and three heterozygotes. Statistical comparisons on organ volume, growth rate, and organ volume/bodyweight ratios were made between sl/sl and the control group. RESULTS: Cardiac morphology and constituents were preserved. However, significant volumetric reductions were recorded in sl/sl rats with respect to the control: whole heart (38.70%, p value = 0.02); LV (41.22%, p value = 0.01), RV (46.15%, p value = 0.02), LA (44.93%, p value = 0.06), and RA (39.49%, p value = 0.02). Consistent trend was observed in growth rate (~ 20%) and organ-volume/bodyweight ratios (~ 25%). On the contrary, measurements on aortic arch demonstrated no significant difference among the two groups. CONCLUSION: Despite the presence of normal morphology, significant cardiac growth retardation was detected in sl/sl rat, supporting the likely association of cardiac anomalies with HSCR, at least in ETB-/- subtype. Structural reduction was likely due to a combination of failure to thrive from enteric dysfunction, alterations to CaNCC colonization, and importantly coronary hypoperfusion from elevated ET-1/ETA-mediated hypervasoconstriction. Little correlation was detected between aortic arch development and sl/sl rat, supporting minor ETB role in large vessels. Although further clinical study is warranted, HSCR patients may likely require cardiac assessment in view of potential congenital cardiac defects.


Subject(s)
Heart Defects, Congenital/genetics , Heart/growth & development , Hirschsprung Disease/genetics , Receptor, Endothelin B/genetics , Animals , Animals, Newborn , Disease Models, Animal , Genetic Predisposition to Disease , Heart/diagnostic imaging , Heart Defects, Congenital/diagnostic imaging , Heart Defects, Congenital/metabolism , Heart Defects, Congenital/physiopathology , Hirschsprung Disease/metabolism , Hirschsprung Disease/physiopathology , Mutation , Myocardium/pathology , Rats, Transgenic , Receptor, Endothelin B/metabolism , Weight Gain , X-Ray Microtomography
18.
Endocr Regul ; 55(2): 72-82, 2021 May 21.
Article in English | MEDLINE | ID: mdl-34020533

ABSTRACT

Objective. The aim of the present investigation was to study the impact of glucose and gluta-mine deprivations on the expression of genes encoding EDN1 (endothelin-1), its cognate receptors (EDNRA and EDNRB), and ECE1 (endothelin converting enzyme 1) in U87 glioma cells in response to knockdown of ERN1 (endoplasmic reticulum to nucleus signaling 1), a major signaling pathway of endoplasmic reticulum stress, for evaluation of their possible implication in the control of glioma growth through ERN1 and nutrient limitations. Methods. The expression level of EDN1, its receptors and converting enzyme 1 in control U87 glioma cells and cells with knockdown of ERN1 treated by glucose or glutamine deprivation by quantitative polymerase chain reaction was studied. Results. We showed that the expression level of EDN1 and ECE1 genes was significantly up-regulated in control U87 glioma cells exposure under glucose deprivation condition in comparison with the glioma cells, growing in regular glucose containing medium. We also observed up-regulation of ECE1 gene expression in U87 glioma cells exposure under glutamine deprivation as well as down-regulation of the expression of EDN1 and EDNRA mRNA, being more significant for EDN1. Furthermore, the knockdown of ERN1 signaling enzyme function significantly modified the response of most studied gene expressions to glucose and glutamine deprivation conditions. Thus, the ERN1 knockdown led to a strong suppression of EDN1 gene expression under glucose deprivation, but did not change the effect of glutamine deprivation on its expression. At the same time, the knockdown of ERN1 signaling introduced the sensitivity of EDNRB gene to both glucose and glutamine deprivations as well as completely removed the impact of glucose deprivation on the expression of ECE1 gene. Conclusions. The results of this study demonstrated that the expression of endothelin-1, its receptors, and ECE1 genes is preferentially sensitive to glucose and glutamine deprivations in gene specific manner and that knockdown of ERN1 significantly modified the expression of EDN1, EDNRB, and ECE1 genes in U87 glioma cells. It is possible that the observed changes in the expression of studied genes under nutrient deprivation may contribute to the suppressive effect of ERN1 knockdown on glioma cell proliferation and invasiveness.


Subject(s)
Endoribonucleases/metabolism , Endothelin-1/metabolism , Endothelin-Converting Enzymes/metabolism , Glioma/metabolism , Glucose/metabolism , Glutamine/metabolism , Protein Serine-Threonine Kinases/metabolism , Receptor, Endothelin A/metabolism , Receptor, Endothelin B/metabolism , Cell Line, Tumor , Gene Expression/genetics , Gene Knockdown Techniques , Humans , RNA, Messenger/metabolism
19.
Int J Mol Sci ; 22(9)2021 Apr 21.
Article in English | MEDLINE | ID: mdl-33919338

ABSTRACT

In brain disorders, reactive astrocytes, which are characterized by hypertrophy of the cell body and proliferative properties, are commonly observed. As reactive astrocytes are involved in the pathogenesis of several brain disorders, the control of astrocytic function has been proposed as a therapeutic strategy, and target molecules to effectively control astrocytic functions have been investigated. The production of brain endothelin-1 (ET-1), which increases in brain disorders, is involved in the pathophysiological response of the nervous system. Endothelin B (ETB) receptors are highly expressed in reactive astrocytes and are upregulated by brain injury. Activation of astrocyte ETB receptors promotes the induction of reactive astrocytes. In addition, the production of various astrocyte-derived factors, including neurotrophic factors and vascular permeability regulators, is regulated by ETB receptors. In animal models of Alzheimer's disease, brain ischemia, neuropathic pain, and traumatic brain injury, ETB-receptor-mediated regulation of astrocytic activation has been reported to improve brain disorders. Therefore, the astrocytic ETB receptor is expected to be a promising drug target to improve several brain disorders. This article reviews the roles of ETB receptors in astrocytic activation and discusses its possible applications in the treatment of brain disorders.


Subject(s)
Astrocytes/metabolism , Brain Diseases/metabolism , Receptor, Endothelin B/metabolism , Alzheimer Disease/metabolism , Alzheimer Disease/physiopathology , Animals , Astrocytes/physiology , Brain Diseases/physiopathology , Brain Injuries/metabolism , Brain Injuries/physiopathology , Brain Ischemia/metabolism , Brain Ischemia/physiopathology , Endothelin-1/metabolism , Humans , Neuralgia/metabolism , Neuralgia/physiopathology , Receptor, Endothelin B/physiology
20.
Int J Mol Sci ; 22(18)2021 Sep 07.
Article in English | MEDLINE | ID: mdl-34575824

ABSTRACT

The development of the enteric nervous system (ENS) is highly modulated by the synchronized interaction between the enteric neural crest cells (ENCCs) and the neural stem cell niche comprising the gut microenvironment. Genetic defects dysregulating the cellular behaviour(s) of the ENCCs result in incomplete innervation and hence ENS dysfunction. Hirschsprung disease (HSCR) is a rare complex neurocristopathy in which the enteric neural crest-derived cells fail to colonize the distal colon. In addition to ENS defects, increasing evidence suggests that HSCR patients may have intrinsic defects in the niche impairing the extracellular matrix (ECM)-cell interaction and/or dysregulating the cellular niche factors necessary for controlling stem cell behaviour. The niche defects in patients may compromise the regenerative capacity of the stem cell-based therapy and advocate for drug- and niche-based therapies as complementary therapeutic strategies to alleviate/enhance niche-cell interaction. Here, we provide a summary of the current understandings of the role of the enteric neural stem cell niche in modulating the development of the ENS and in the pathogenesis of HSCR. Deciphering the contribution of the niche to HSCR may provide important implications to the development of regenerative medicine for HSCR.


Subject(s)
Enteric Nervous System/cytology , Enteric Nervous System/metabolism , Hirschsprung Disease/genetics , Neural Stem Cells/cytology , Neural Stem Cells/physiology , Stem Cell Niche , Animals , Biomarkers , Cell Differentiation , Disease Management , Disease Susceptibility , Endothelin-3/metabolism , Genetic Predisposition to Disease , Glial Cell Line-Derived Neurotrophic Factor/metabolism , Hirschsprung Disease/diagnosis , Hirschsprung Disease/metabolism , Hirschsprung Disease/therapy , Humans , Neural Crest/cytology , Neural Crest/metabolism , Receptor, Endothelin B/metabolism , Regenerative Medicine , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL