Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
Children (Basel) ; 11(5)2024 May 20.
Article in English | MEDLINE | ID: mdl-38790606

ABSTRACT

In neonates with acute lung injury (ALI), targeting lower oxygenation saturations is suggested to limit oxygen toxicity while maintaining vital organ function. Although thresholds for cerebral autoregulation are studied for the management of premature infants, the impact of hypoxia on hemodynamics, tissue oxygen consumption and extraction is not well understood in term infants with ALI. We examined hemodynamics, cerebral autoregulation and fractional oxygen extraction, as measured by near-infrared spectroscopy (NIRS) and blood gases, in a neonatal porcine oleic acid injury model of moderate ALI. We hypothesized that in ALI animals, cerebral oxygen extraction would be increased to a greater degree than kidney or gut oxygen extraction as indicative of the brain's adaptive efforts to increase cerebral oxygen extraction at the expense of splanchnic end organs. Fifteen anesthetized, ventilated 5-day-old neonatal piglets were divided into moderate lung injury by treatment with oleic acid or control (sham injection). The degree of lung injury was quantified at baseline and after establishment of ALI by blood gases, ventilation parameters and calculated oxygenation deficit, hemodynamic indices by echocardiography and lung injury score by ultrasound. PaCO2 was maintained constant during ventilation. Cerebral, renal and gut oxygenation was determined by NIRS during stepwise decreases in inspired oxygen from 50% to 21%, correlated with PaO2 and PvO2; changes in fractional oxygen extraction (ΔFOE) were calculated from NIRS and from regional blood gas samples. The proportion of cerebral autoregulation impairment attributable to blood pressure, and to hypoxemia, was calculated from autoregulation nomograms. ALI manifested as hypoxemia with increasing intrapulmonary shunt fraction, decreased lung compliance and increased resistance, and marked increase in lung ultrasound score. Brain, gut and renal NIRS, obtained from probes placed over the anterior skull, central abdomen and flank, respectively, correlated with concurrent SVC (brain) or IVC (gut, renal) PvO2 and SvO2. Cerebral autoregulation was impaired after ALI as a function of blood pressure at all FiO2 steps, but predominantly by hypoxemia at FiO2 < 40%. Cerebral ΔFOE was higher in ALI animals at all FiO2 steps. We conclude that in an animal model of neonatal ALI, cerebrovascular blood flow regulation is primarily dependent on oxygenation. There is not a defined oxygenation threshold below which cerebral autoregulation is impaired in ALI. Cerebral oxygen extraction is enhanced in ALI, reflecting compensation for exhausted cerebral autoregulation due to the degree of hypoxemia and/or hypotension, thereby protecting against tissue hypoxia.

2.
Am J Perinatol ; 41(S 01): e3401-e3412, 2024 May.
Article in English | MEDLINE | ID: mdl-38211608

ABSTRACT

OBJECTIVE: To evaluate the sensitivity and specificity of clinical, laboratory, and radiological markers and the neonatologist-performed intestinal ultrasound (NP-IUS) for treatment interventions in preterm neonates who developed necrotizing enterocolitis (NEC). STUDY DESIGN: This was a case-control study of preterm neonates < 35 weeks with a diagnostic workup for NEC. The diagnostic workup included NP-IUS performed by trained neonatologists using a standard protocol, abdominal roentgenogram (AXR), and laboratory investigations. Intestinal ultrasound (IUS) performed by two neonatologists was standardized to detect 11 injury markers. AXRs were read independently by experienced pediatric radiologists. The investigators who retrospectively interpreted the IUS were blinded to the clinical and treatment outcomes. RESULTS: A total of 111 neonates were assessed. Fifty-four did not require intervention and formed the control group. Twenty cases were treated medically, 21 cases were treated with late surgery for stricture or adhesions, and 16 were treated with early surgery. The integrated model of cumulative severity of ultrasound markers, respiratory and hemodynamic instability, abdominal wall cellulitis, and C- reactive protein > 16 mg/L had an area under the curve (AUC) of 0.89 (95% confidence interval [CI]: 0.83-0.94%, p < 0.0001) for diagnosing NEC requiring surgical intervention. We also investigated the utility of Bell's classification to diagnose either the need for surgery or death, and it had an AUC of 0.74 (95% CI: 0.65-0.83%, p < 0.0001). CONCLUSION: In this cohort, a combination of specific IUS markers and clinical signs of instability, abdominal wall cellulitis, plus laboratory markers were diagnostic of NEC requiring interventions. KEY POINTS: · The diagnosis of necrotizing enterocolitis requires a combination of markers.. · The combination of specific ultrasound markers, clinical signs, and laboratory markers were diagnostic of NEC requiring intervention.. · The intestinal ultrasound performed by a trained neonatologist was the most sensitive diagnostic marker of NEC requiring surgical intervention..


Subject(s)
Enterocolitis, Necrotizing , Infant, Premature , Ultrasonography , Humans , Enterocolitis, Necrotizing/diagnostic imaging , Enterocolitis, Necrotizing/classification , Infant, Newborn , Case-Control Studies , Male , Female , Retrospective Studies , Sensitivity and Specificity , Biomarkers/blood , C-Reactive Protein/analysis , ROC Curve , Radiography, Abdominal , Intestines/diagnostic imaging , Severity of Illness Index
3.
Front Physiol ; 14: 1144574, 2023.
Article in English | MEDLINE | ID: mdl-37064915

ABSTRACT

Introduction: Hypoxic persistent pulmonary hypertension in the newborn (PPHN) is usually treated with oxygen and inhaled nitric oxide (NO), both pulmonary arterial relaxants. But treatment failure with NO occurs in 25% of cases. We previously demonstrated that 72 h exposure to hypoxia, modeling PPHN, sensitized pulmonary artery smooth muscle cells (PASMC) to the contractile agonist thromboxane and inhibited relaxant adenylyl cyclase (AC) activity. Methods: In this study, we examined the effects of sodium nitroprusside (SNP), as NO donor, on the thromboxane-mediated contraction and NO-independent relaxation pathways and on reactive oxygen species (ROS) accumulation in PASMC. In addition, we examined the effect of the peroxynitrite scavenger 5,10,15,20-Tetrakis (4-sulfonatophenyl)porphyrinato Iron (III) (FeTPPS) on these processes. Results: Exposure of PASMC to 72 h hypoxia increased total intracellular ROS compared to normoxic control cells and this was mitigated by treatment of cells with either SNP or FeTPPS. Total protein nitrosylation was increased in hypoxic PASMC compared to controls. Both normoxic and hypoxic cells treated with SNP exhibited increased total protein nitrosylation and intracellular nitrite; this was reduced by treatment with FeTPPS. While cell viability and mitochondrial number were unchanged by hypoxia, mitochondrial activity was decreased compared to controls; addition of FeTPPS did not alter this. Basal and maximal mitochondrial metabolism and ATP turnover were reduced in hypoxic PASMC compared to controls. Hypoxic PASMC had higher basal Ca2+, and a heightened peak Ca2+ response to thromboxane challenge compared to controls. Addition of SNP further elevated the peak Ca2+ response, while addition of FeTPPS brought peak Ca2+ response down to control levels. AC mediated relaxation was impaired in hypoxic PASMC compared to controls but was normalized following treatment with FeTPPS. Addition of SNP inhibited adenylyl cyclase activity in both normoxic and hypoxic PASMC. Moreover, addition of the Ca2+ chelator BAPTA improved AC activity, but the effect was minimal. Discussion: We conclude that NO independently augments contraction and inhibits relaxation pathways in hypoxic PASMC, in part by a mechanism involving nitrogen radical formation and protein nitrosylation. These observations may partially explain impaired effectiveness of NO when treating hypoxic pulmonary hypertension.

4.
Biology (Basel) ; 12(4)2023 Apr 10.
Article in English | MEDLINE | ID: mdl-37106773

ABSTRACT

BACKGROUND: The adenylyl cyclase (AC) pathway, crucial for pulmonary vasodilation, is inhibited by hypoxia. Forskolin (FSK) binds allosterically to AC, stimulating ATP catalysis. As AC6 is the primary AC isoform in the pulmonary artery, selective reactivation of AC6 could provide targeted reinstatement of hypoxic AC activity. This requires elucidation of the FSK binding site in AC6. METHODS: HEK293T cells stably overexpressing AC 5, 6, or 7 were incubated in normoxia (21% O2) or hypoxia (10% O2) or exposed to s-nitrosocysteine (CSNO). AC activity was measured using terbium norfloxacin assay; AC6 structure built by homology modeling; ligand docking to examine FSK-interacting amino acids; roles of selected residues determined by site-directed mutagenesis; FSK-dependent cAMP generation measured in wild-type and FSK-site mutants by biosensor-based live cell assay. RESULTS: Only AC6 is inhibited by hypoxia and nitrosylation. Homology modeling and docking revealed residues T500, N503, and S1035 interacting with FSK. Mutation of T500, N503, or S1035 decreased FSK-stimulated AC activity. FSK site mutants were not further inhibited by hypoxia or CSNO; however, mutation of any of these residues prevented AC6 activation by FSK following hypoxia or CSNO treatment. CONCLUSIONS: FSK-interacting amino acids are not involved in the hypoxic inhibition mechanism. This study provides direction to design FSK derivatives for selective activation of hypoxic AC6.

5.
FASEB Bioadv ; 4(3): 180-196, 2022 Mar.
Article in English | MEDLINE | ID: mdl-35664968

ABSTRACT

Activation of adenylyl cyclases (ACs) by G-protein Gαs catalyzes the production of cyclic adenosine monophosphate (cAMP), a key second messenger that regulates diverse physiological responses. There are 10 AC isoforms present in humans, with AC5 and AC6 proposed to play vital roles in cardiac function. We have previously shown that under hypoxic conditions, AC6 is amenable to post-translational modification by nitrosylation, resulting in decreased AC catalytic activity. Using a computational model of the AC6-Gαs complex, we predicted key nitrosylation-amenable cysteine residues involved in the interaction of AC6 with Gαs and pursued a structure-function analysis of these cysteine residues in both AC6 and Gαs. Our results based on site-directed mutagenesis of AC6 and Gαs, a constitutively active Gαs, AC activity, and live cell intracellular cAMP assays suggest that Cys1004 in AC6 (subunit C2) and Cys237 in Gαs are present at the AC-Gαs interface and are important for the activation of AC6 by Gαs. We further provide mechanistic evidence to show that mutating Cys 1004 in the second catalytic domain of AC6 makes it amenable to inhibition by Gαi, which may account for decreased functional activity of AC6 when this residue is unavailable.

6.
Biochem Biophys Rep ; 27: 101080, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34368469

ABSTRACT

Persistent pulmonary hypertension of the newborn (PPHN) is characterized by hypoxemia and arterial remodeling. Dynamic stretch and recoil of the arterial wall during pulsation (in normal conduit arteries, stretch 20% above diastolic diameter) maintains homeostasis; a static arterial wall is associated with remodeling. PPHN is diagnosed by echocardiography as decreased pulmonary artery wall displacement during systole, causing decreased pulmonary arterial pressure acceleration time in a stiff artery. We hypothesized that a 'normal' amplitude of pulsatile stretch is protective against ER stress, while the loss of stretch is a trigger for hypoxia-induced stress responses. Using a novel in vitro model of pulmonary arterial myocytes subject to repetitive stretch-relaxation cycles within a normoxic or hypoxic environment, we examined the relative impact of hypoxia (pulmonary circuit during unresolved PPHN) and cyclic mechanical stretch (diminished in PPHN) on myocyte homeostasis, specifically on signaling proteins for autophagy and endoplasmic reticulum (ER) stress. Stretch induced autophagosome abundance under electron microscopy. Hypoxia, in presence or absence of pulsatile stretch, decreased unfolded protein response (UPR) hallmark BIP (GRP78) in contractile phenotype pulmonary arterial myocytes. Inositol requiring enzyme-1 α (IRE1α) was not activated; but hypoxia induced eif2α phosphorylation, increasing expression of ATF4 (activating transcription factor-4). This was sensitive to inhibition by autophagy inhibitor bafilomycin A1. We conclude that in the pulmonary circuit, hypoxia induces one arm of the UPR pathway and causes ER stress. Pulsatile stretch ameliorates the hypoxic UPR response, and while increasing presence of autophagosomes, does not activate canonical autophagy signaling pathways. We propose that simultaneous application of hypoxia and graded levels of cyclic stretch can be used to distinguish myocyte signaling in the deformable pulmonary artery of early PPHN, versus the inflexible late stage PPHN artery.

7.
Eur J Pediatr ; 180(6): 1747-1754, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33486603

ABSTRACT

The impact of patent ductus arteriosus (PDA) on vital sign trends represented as histograms, and perfusion index in particular, is unknown. This study aimed to split continuously obtained PI and other vital signs before, during, and after medical treatment of PDA, into histogram bins, and determine the utility of PI and other vital sign histograms in the early prediction of hemodynamically significant PDA (hsPDA). In 34 infants at a mean gestational age of 26 ± 2.1 weeks, we prospectively collected vital signs for three different periods, 24 h before starting treatment of PDA, during PDA treatment, and 24 h after completion of the course of treatment, and confirmed PDA closure by echo. Histograms with three comparable periods were obtained from preterm infants who did not require treatment for PDA and analyzed for comparison. The duration of time spent in each histogram bin was determined for each time epoch. Episodes of low PI < 0.4 and high PI > 2 were significantly longer in duration in infants with PDA before treatment compared to those in infants with PDA during and after treatment. The arterial oxygen saturation (SpO2) < 80% was also longer in duration in infants with PDA before compared to that in infants with PDA during and after treatment. Low PI < 0.4 correlated with most echocardiography indices of hsPDA.Conclusion: We conclude that a patent ductus arteriosus requiring treatment in preterm infants ≤ 29 weeks GA was associated with significant fluctuations between a low PI < 0.4 alternating with a high PI > 2, reflecting the dynamic nature of hsPDA shunt volume. PI variability may be an early marker of hsPDA. What is Known: • The perfusion index is a continuous underutilized parameter provided by pulse oximetry to assess the peripheral perfusion. • The perfusion index helps predict conditions with hemodynamic instability. What is New: • The perfusion index assessed as daily histogram trends can predict patent ductus arteriosus requiring treatment.


Subject(s)
Ductus Arteriosus, Patent , Ductus Arteriosus, Patent/diagnostic imaging , Ductus Arteriosus, Patent/therapy , Gestational Age , Hemodynamics , Humans , Infant , Infant, Newborn , Infant, Premature , Perfusion Index
8.
Front Oral Health ; 2: 735634, 2021.
Article in English | MEDLINE | ID: mdl-35048051

ABSTRACT

Pregnancy is a tightly regulated immunological state. Mild environmental perturbations can affect the developing fetus significantly. Infections can elicit severe immunological cascades in the mother's body as well as the developing fetus. Maternal infections and resulting inflammatory responses can mediate epigenetic changes in the fetal genome, depending on the developmental stage. The craniofacial development begins at the early stages of embryogenesis. In this review, we will discuss the immunology of pregnancy and its responsive mechanisms on maternal infections. Further, we will also discuss the epigenetic effects of pathogens, their metabolites and resulting inflammatory responses on the fetus with a special focus on craniofacial development. Understanding the pathophysiological mechanisms of infections and dysregulated inflammatory responses during prenatal development could provide better insights into the origins of craniofacial birth defects.

9.
Pediatr Pulmonol ; 55(11): 2913-2923, 2020 11.
Article in English | MEDLINE | ID: mdl-32741109

ABSTRACT

RATIONALE: Point-of-care ultrasound (POCUS) is used to evaluate pulmonary edema in adults with acute respiratory distress syndrome (ARDS). Its use has not been validated in neonatal models. OBJECTIVES: We compared an in vivo lung ultrasound score against clinical and histological markers of acute lung injury, in a neonatal animal model, hypothesizing that POCUS would sensitively diagnose early acute lung injury in neonates and discern its severity. METHODS: Fifteen anesthetized, ventilated 3-day-old neonatal piglets were divided into controls, moderate lung injury, or severe lung injury by graded treatment with oleic acid. Degree of lung injury was quantified at baseline, immediately after oleic acid administration, and 1 hour after the evolution of acute lung injury, by blood gases, ventilation parameters and calculated oxygenation deficit; hemodynamic indices by echocardiography, and lung ultrasound obtained in an 8-region grid of anterior and posterior zones, semi-quantitatively analyzed by a blinded observer. Lungs were inflation-fixed postmortem at last mean airway pressure, for histological assessment. RESULTS: Acute lung injury manifested in oleic acid-treated groups as dose-dependent capillary leak causing intravascular depletion and cardiac failure, hypoxemia with increasing intrapulmonary shunt fraction, decreased lung compliance, and resistance. Ultrasound scores of anterior regions distinguished moderate from severe injury; scores in posterior regions reached maximum values immediately after lung injury. POCUS score correlated with calculated intrapulmonary shunt fraction (R2 = .65) and with histological injury score (R2 = .61), P < .01. CONCLUSION: We conclude that POCUS may be valuable in neonates for early quantification of acute lung injury or ARDS; and that nondependent ultrasound regions clearly distinguish severity of pulmonary edema.


Subject(s)
Acute Lung Injury/diagnostic imaging , Lung/diagnostic imaging , Pulmonary Edema/diagnostic imaging , Respiratory Distress Syndrome/diagnostic imaging , Acute Lung Injury/blood , Acute Lung Injury/physiopathology , Animals , Animals, Newborn , Blood Gas Analysis , Female , Hemodynamics , Lung/physiopathology , Male , Oleic Acid/administration & dosage , Pulmonary Edema/physiopathology , Respiratory Distress Syndrome/blood , Respiratory Distress Syndrome/physiopathology , Swine , Ultrasonography
10.
Methods Mol Biol ; 1947: 389-401, 2019.
Article in English | MEDLINE | ID: mdl-30969430

ABSTRACT

G protein-coupled receptors (GPCR) are crucial transducers of extracellular signals into changes in vascular tone. Vasoactive GPCR stimulation in the pulmonary circuit may be elicited by agonists released in acute tissue hypoxia or inflammation, as well as chronic disease. Acute responses involve activation of smooth muscle contraction or relaxation machinery causing changes in actomyosin interaction, thereby altering lumen diameter. Chronic responses may typically include activation of proliferation or fibrosis. Using pulmonary artery myocytes and pulmonary artery rings, we describe a general strategy for quantification of vasoconstrictor or vasodilator GPCR responses, and for comparison of signaling pathways in cultured cells and in contracted vessels using immunohistochemistry of contracting vessels.


Subject(s)
Muscle Contraction/physiology , Myocytes, Smooth Muscle/metabolism , Pulmonary Artery/metabolism , Receptors, G-Protein-Coupled/metabolism , Cells, Cultured , Humans , Muscle Contraction/drug effects , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/drug effects , Pulmonary Artery/cytology , Pulmonary Artery/drug effects , Vasoconstrictor Agents/pharmacology , Vasodilator Agents/pharmacology
11.
Pediatr Pulmonol ; 54(3): 319-332, 2019 03.
Article in English | MEDLINE | ID: mdl-30644649

ABSTRACT

BACKGROUND: Persistent pulmonary hypertension of the newborn (PPHN) is due to a failure of pulmonary vascular relaxation. Vasopressin, a systemic vasoconstrictor acting on smooth muscle AVPR1a receptors, is used in treatment of PPHN. We sought to determine acute effects of vasopressin infusion on pulmonary hemodynamics in a large animal model of hypoxic PPHN. METHODS: PPHN was induced in 6 newborn piglets by 72 h normobaric hypoxia (FiO2 = 0.10); controls were 7 age-matched 3-day-old piglets. Animals were anesthetized and ventilated with central venous and arterial lines, and after stabilization, randomized using a crossover design to normoxic or hypoxic ventilation, then 30 min infusion of 0.0012 U/kg/min vasopressin, followed by 45 min vasopressin washout period. Echocardiographic parameters and oxygen consumption were measured before and after vasopressin. Relaxation to vasopressin was tested in isolated PPHN and control pulmonary arteries by isometric myography. Expression of AVPR1a receptor mRNA was quantified in arterial and myocardial tissues. RESULTS: Vasopressin did not alleviate hypoxia-responsiveness of PPHN pulmonary circuit. There were no significant differences in pulmonary hypertension, cardiac function indices, or oxygenation indices after vasopressin infusion. Vasopressin did not dilate control or PPHN pulmonary arteries, and AVPR1 was minimally expressed. CONCLUSIONS: Vasopressin does not have a direct pulmonary vasodilator effect in PPHN, within the timeframe studied.


Subject(s)
Hypertension, Pulmonary/drug therapy , Vasoconstrictor Agents/therapeutic use , Vasopressins/therapeutic use , Animals , Animals, Newborn , Disease Models, Animal , Female , Hemodynamics/drug effects , Hypertension, Pulmonary/physiopathology , Male , Oxygen Consumption/drug effects , Pulmonary Artery/drug effects , Pulmonary Artery/physiology , Random Allocation , Respiration, Artificial , Swine
12.
Pulm Pharmacol Ther ; 48: 195-202, 2018 02.
Article in English | MEDLINE | ID: mdl-29246840

ABSTRACT

Infants with lung disease may be exposed to high O2 concentrations, and may have transient hypoxic episodes due to worsening lung pathophysiology, aggravating pulmonary arterial (PA) oxidative stress. NADPH oxidase (NOX) converts O2 to superoxide. Mitochondrial antioxidants such as superoxide dismutase (SOD) are sensitive to O2 tension. We previously reported decreased SOD2 activity in hypoxic PA myocytes due to nitration. In this study, we examined whether a transient hypoxic episode exposes antioxidant defense defects in hyperoxic PA myocytes. PA myocytes of term newborn piglets were cultured in hyperoxia (60% O2) or normoxia (21% O2) for 72 h; cells from both groups were challenged with transient hypoxia (10% O2) for 2 h. We measured NOX activity, SOD activities (fractionated by centrifugation and concavalin A- Sepharose chromatography), total ROS and superoxide generation, 8-isoprostane, and calcium responses to thromboxane mimetic. NOX activity increased in hyperoxic myocytes. Hyperoxia increased SOD1 activity but decreased SOD2 activity. Total ROS were reduced in hyperoxia, and hyperoxia + hypoxia groups. While hyperoxia alone did not alter superoxide content, superoxide increased after a hypoxic challenge of both normoxic and hyperoxic myocytes. Increased 8-isoprostane was seen only in hyperoxic myocytes challenged by transient hypoxia. We conclude that hyperoxic PA myocytes can limit total ROS despite increased NOX activity, but with inhibited SOD2 activity. Transient hypoxia increases superoxide formation; in the face of impaired SOD2, despite induction of SOD1, this oxidative stress causes increased 8-isoprostane generation. This may contribute to the mechanism of pulmonary arterial reactivity in infants with severe lung disease.


Subject(s)
Antioxidants/metabolism , Oxidative Stress , Pulmonary Artery/metabolism , Superoxide Dismutase/metabolism , Animals , Animals, Newborn , Cell Hypoxia/physiology , Dinoprost/analogs & derivatives , Dinoprost/metabolism , Hyperoxia/metabolism , Hypoxia/metabolism , Mitochondria/metabolism , Myocytes, Smooth Muscle/metabolism , NADPH Oxidases/metabolism , Pulmonary Artery/cytology , Swine
13.
Methods Cell Biol ; 142: 101-110, 2017.
Article in English | MEDLINE | ID: mdl-28964329

ABSTRACT

G protein-coupled receptors (GPCRs) signal in response to various external stimuli including stress. GPCR signaling has been shown to play a critical role in the adaptation of cell response to limited oxygen supply. Hypoxia has been implicated in cardiovascular diseases, human pulmonary arterial responses, and persistent pulmonary hypertension in newborns. One of the key GPCRs implicated in hypoxia is the prostanoid receptor, thromboxane A2 receptor (TP). Hypoxia can affect TP localization, stability, and activity both in vivo and in vitro. To elucidate hypoxia-mediated GPCR signaling in vitro, we lay out a general strategy to perform hypoxic experiments using both primary pulmonary artery smooth muscle cells and TP expressed in HEK293T cells. We describe assay for measuring moderate tissue hypoxia using static cell cultures, monitoring pericellular media oxygen content, and signaling of TP.


Subject(s)
Biological Assay/methods , Hypoxia/metabolism , Receptors, Thromboxane A2, Prostaglandin H2/metabolism , Signal Transduction , Calcium/metabolism , HEK293 Cells , Humans , Myocytes, Smooth Muscle , Primary Cell Culture , Pulmonary Artery/cytology
14.
Article in English | MEDLINE | ID: mdl-26583045

ABSTRACT

BACKGROUND: Persistent pulmonary hypertension of the newborn (PPHN) is characterized by vasoconstriction and pulmonary vascular remodeling. Remodeling is believed to be a response to physical or chemical stimuli including pro-mitotic inflammatory mediators such as thromboxane. Our objective was to examine the effects of hypoxia and thromboxane signaling ex vivo and in vitro on phenotype commitment, cell cycle entry, and proliferation of PPHN and control neonatal pulmonary artery (PA) myocytes in tissue culture. METHODS: To examine concurrent effects of hypoxia and thromboxane on myocyte growth, serum-fed first-passage newborn porcine PA myocytes were randomized into normoxic (21 % O2) or hypoxic (10 % O2) culture for 3 days, with daily addition of thromboxane mimetic U46619 (10(-9) to 10(-5) M) or diluent. Cell survival was detected by MTT assay. To determine the effect of chronic thromboxane exposure (versus whole serum) on activation of arterial remodeling, PPHN was induced in newborn piglets by a 3-day hypoxic exposure (FiO2 0.10); controls were 3 day-old normoxic and day 0 piglets. Third-generation PA were segmented and cultured for 3 days in physiologic buffer, Ham's F-12 media (in the presence or absence of 10 % fetal calf serum), or media with 10(-6) M U46619. DNA synthesis was measured by (3)H-thymidine uptake, protein synthesis by (3)H-leucine uptake, and proliferation by immunostaining for Ki67. Cell cycle entry was studied by laser scanning cytometry of nuclei in arterial tunica media after propidium iodide staining. Phenotype commitment was determined by immunostaining tunica media for myosin heavy chain and desmin, quantified by laser scanning cytometry. RESULTS: Contractile and synthetic myocyte subpopulations had differing responses to thromboxane challenge. U46619 decreased proliferation of synthetic and contractile myocytes. PPHN arteries exhibited decreased protein synthesis under all culture conditions. Serum-supplemented PA treated with U46619 had decreased G1/G0 phase myocytes and an increase in S and G2/M. When serum-deprived, PPHN PA incubated with U46619 showed arrested cell cycle entry (increased G0/G1, decreased S and G2/M) and increased abundance of contractile phenotype markers. CONCLUSIONS: We conclude that thromboxane does not initiate phenotypic dedifferentiation and proliferative activation in PPHN PA. Exposure to thromboxane triggers cell cycle exit and myocyte commitment to contractile phenotype.

15.
Am J Respir Cell Mol Biol ; 50(1): 135-43, 2014 Jan.
Article in English | MEDLINE | ID: mdl-23962128

ABSTRACT

Pulmonary arterial vasoconstriction is a hallmark of persistent pulmonary hypertension of the newborn (PPHN). We reported increased calcium responses to thromboxane and selectively increased thromboxane prostanoid (TP) association with Gαq in hypoxic pulmonary artery. Palmitoylation of Gαq is important for efficient receptor-Gαq-phospholipase-C interactions. TPα receptor is not itself amenable to palmitoylation. We studied the role of Gαq palmitoylation in constriction of hypoxic pulmonary artery using pharmacological palmitoylation inhibition, the effects of hypoxia on palmitoylation, and the effects of site-specific cysteine substitution mutations of Gαq on Gαq membrane targeting, TPα association, and calcium dose-response curve to a TP agonist. PPHN pulmonary artery and HEK293T cells expressing TPα were exposed to irreversible palmitoylation inhibitor 2-bromopalmitate before challenge with TP agonist U46619. Palmitate uptake was studied in hypoxic and normoxic myocytes. Wild-type Gαq and Gαq cysteine-to-alanine mutants C9A, C10A, and C9A/C10A were transiently coexpressed in HEK293T cells stably expressing TPα. We examined membrane localization of Gαq, TP receptor-Gαq association by coimmunoprecipitation, and Ca(2+) responses to U46619 in hypoxic and normoxic cells. Gαq palmitoylation is essential for the Ca(2+) response to TPα stimulation. Inhibition of palmitoylation reduces contractile force to thromboxane in PPHN but not in control pulmonary artery. Hypoxia increases palmitoylation of Gαq; the hypoxic. but not the normoxic, response to thromboxane is palmitoylation sensitive. Palmitoylation of one N-terminal cysteine is required for physical association of Gαq with the TPα receptor. Palmitoylation of both cysteines is required for Gαq membrane localization and Ca(2+) mobilization. Depalmitoylation of any one Gαq cysteine reduces the hypoxic response to thromboxane challenge to equal that of normoxic cells.


Subject(s)
GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , Hypertension, Pulmonary/metabolism , Hypoxia/metabolism , Lipoylation/physiology , Receptors, Thromboxane/metabolism , Animals , Animals, Newborn , Calcium/metabolism , Cells, Cultured , Cysteine/metabolism , HEK293 Cells , Humans , Muscle Cells/metabolism , Prostaglandins/metabolism , Pulmonary Artery/metabolism , Swine/metabolism , Thromboxanes/metabolism , Vasoconstriction/physiology
16.
Pediatr Pulmonol ; 49(9): 885-97, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24167160

ABSTRACT

BACKGROUND: Hypoxia and reactive oxygen species (ROS) including H(2)O(2) play major roles in triggering and progression of pulmonary vascular remodeling in persistent pulmonary hypertension. Catalase (CAT), the major endogenous enzyme scavenging H(2)O(2), is regulated in a tissue- and context-specific manner. OBJECTIVE: To investigate mechanisms by which hypoxia and H(2)O(2) regulate catalase expression, and the role of AMPK-FoxO pathway, in neonatal porcine pulmonary artery smooth muscle (PASMC). DESIGN/METHODS: PASMC were grown in hypoxia (10% O(2)) or normoxia (21% O(2)) for 72 hr. We measured catalase activity and lipid peroxidation; CAT, FoxO1, and FoxO3a expression by qPCR; protein contents of CAT, FoxOs, p-AMPK, p-AKT, p-JNK, p-ERK1/2 in whole lysates, and FoxOs in nuclear extracts, by immunoblot; and FoxO-1 nuclear localization by immunocytochemistry, quantified by laser scanning cytometry. RESULTS: Hypoxia upregulated CAT transcription, content and activity, by increasing CAT transcription factors FoxO1 and FoxO3a mRNA, and promoting nuclear translocation of FoxO1. However, lipid peroxidation increased in hypoxic PASMC. Among candidate FoxO regulatory kinases, hypoxia activated AMPK, and decreased p-Akt and ERK1/2. AMPK activation increased FoxO1 (total and nuclear) and CAT, while AMPK inhibition inhibited FoxO1 and CAT, but not FoxO3a. Exogenous H(2)O(2) decreased p-AMPK and increased p-AKT in hypoxic PASMC. This decreased active FoxO1, and reduced mRNA and protein content of CAT. Hypoxic induction of CAT, AKT inhibition (LY294002), or addition of PEG-catalase partly ameliorated the H(2)O(2) -mediated loss of nuclear FoxO1. CONCLUSIONS: Hypoxia induces catalase expression, though this adaptation is insufficient to protect PASMC from hypoxia-induced lipid peroxidation. This occurs via hypoxic activation of AMPK, which promotes nuclear FoxO1 and thus catalase expression. Exogenous ROS may downregulate cellular antioxidant defenses; H(2)O(2) activates survival factor Akt, decreasing nuclear FoxO1 and thus catalase.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Forkhead Transcription Factors/metabolism , Hypoxia/metabolism , Muscle, Smooth/metabolism , Pulmonary Artery/metabolism , Animals , Catalase/genetics , Catalase/metabolism , Lipid Peroxidation , RNA, Messenger/metabolism , Reactive Oxygen Species/metabolism , Swine , Up-Regulation
17.
Neurourol Urodyn ; 30(8): 1633-8, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21826715

ABSTRACT

AIMS: The objective of the present study was to evaluate drug efflux transporter interactions of darifenacin and examine the impact of such transporter interactions on darifenacin permeability in an in vitro model of the blood-brain barrier (BBB) and blood-ocular barrier (BOB). METHODS: Cell membranes expressing human P-glycoprotein (P-gp), multidrug resistance-associated protein (MRP), and breast cancer resistance protein (BCRP) were examined for ATPase activity following darifenacin exposure (0-10 µM). Primary cultured bovine brain microvessel endothelial cells (BBMEC) and P-gp transfected Manin-Darby canine kidney epithelial cells (MDCKMDR1) were used to examine darifenacin permeability and drug efflux transporter responses. RESULTS: Concentration-dependent increases in ATPase activity was observed in P-gp membranes following darifenacin exposure. Both MRP and BCRP membrane preparations were unresponsive to darifenacin. Studies in both BBMEC and MDCKMDR1 monolayers confirmed a P-gp interaction for darifenacin and significantly greater efflux (basolateral to apical) permeability for darifenacin that was reduced by the P-gp inhibitor, elacridar. CONCLUSIONS: Darifenacin is a substrate for the P-gp drug efflux transporter present in both BBB and BOB. The P-gp drug efflux transporter liabilities of darifenacin may limit its penetration into brain and ocular tissue thereby reducing side effect potential.


Subject(s)
Benzofurans/metabolism , Blood-Aqueous Barrier/metabolism , Blood-Brain Barrier/metabolism , Endothelial Cells/metabolism , Epithelial Cells/metabolism , Muscarinic Antagonists/metabolism , Pyrrolidines/metabolism , ATP Binding Cassette Transporter, Subfamily B , ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 2 , ATP-Binding Cassette Transporters/metabolism , Adenosine Triphosphatases/metabolism , Animals , Benzofurans/pharmacology , Biological Transport , Blood-Aqueous Barrier/drug effects , Blood-Brain Barrier/drug effects , Cattle , Cells, Cultured , Dogs , Dose-Response Relationship, Drug , Endothelial Cells/drug effects , Epithelial Cells/drug effects , Humans , Kinetics , Multidrug Resistance-Associated Proteins/metabolism , Muscarinic Antagonists/pharmacology , Neoplasm Proteins/metabolism , Permeability , Pyrrolidines/pharmacology , Transfection
18.
Pharm Res ; 28(10): 2505-15, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21598079

ABSTRACT

PURPOSE: To examine functional activity of P-glycoprotein (P-gp) in the blood-brain barrier (BBB) using near infrared fluorescence (NIRF) imaging techniques. METHODS: Cellular accumulation and bi-directional permeability of the NIRF probe, rhodamine 800 (R800) was determined in MDCKMDR1 and MDCKwt monolayers under normal conditions and following P-gp inhibition with GF120918. Functional P-gp activity was also assessed in mice following administration of R800 alone and with GF230918. Quantitative analysis of R800 fluorescence in brain tissue and blood was measured ex-vivo using Odyssey Near Infrared imaging. RESULTS: R800 accumulation was reduced in MDCKMDR1 compared to MDCKwt monolayers. Addition of GF120918, resulted in increased R800 accumulation in MDCKMDR1 monolayers. Permeability of R800 in MDCKMDR1 monolayers was significantly enhanced (4-fold) in the basolateral to apical direction under control conditions and was abolished following treatment with GF120918. With the exception of the choriod plexus, there was very little penetration of R800 into the brain under control conditions. Treatment of mice with GF120918 resulted in a nearly 4-fold increase in R800 fluorescence in the brain. In contrast, GF120918 had no effect on brain penetration of a vascular permeability marker. CONCLUSIONS: In vitro studies demonstrate the P-gp transporter properties of the NIRF probe R800. Preliminary in vivo studies confirm the P-gp transporter liabilities of R800 and suggest this probe may be useful as a molecular imaging agent for examining P-gp activity in the BBB.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/cerebrospinal fluid , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Blood-Brain Barrier/metabolism , Spectroscopy, Near-Infrared/methods , ATP Binding Cassette Transporter, Subfamily B , ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors , ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , Acridines/pharmacology , Adenosine Triphosphatases/metabolism , Animals , Blood-Brain Barrier/drug effects , Brain/drug effects , Brain/metabolism , Cell Line , Dogs , Humans , Mice , Permeability , Rhodamines/blood , Rhodamines/cerebrospinal fluid , Rhodamines/metabolism , Rhodamines/pharmacology , Tetrahydroisoquinolines/pharmacology , Tissue Distribution
19.
Am J Physiol Lung Cell Mol Physiol ; 293(6): L1406-18, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17890325

ABSTRACT

Contractile responses of airway smooth muscle (ASM) determine airway resistance in health and disease. Caveolae microdomains in the plasma membrane are marked by caveolin proteins and are abundant in contractile smooth muscle in association with nanospaces involved in Ca(2+) homeostasis. Caveolin-1 can modulate localization and activity of signaling proteins, including trimeric G proteins, via a scaffolding domain. We investigated the role of caveolae in contraction and intracellular Ca(2+) ([Ca(2+)](i)) mobilization of ASM induced by the physiological muscarinic receptor agonist, acetylcholine (ACh). Human and canine ASM tissues and cells predominantly express caveolin-1. Muscarinic M(3) receptors (M(3)R) and Galpha(q/11) cofractionate with caveolin-1-rich membranes of ASM tissue. Caveolae disruption with beta-cyclodextrin in canine tracheal strips reduced sensitivity but not maximum isometric force induced by ACh. In fura-2-loaded canine and human ASM cells, exposure to methyl-beta-cyclodextrin (mbetaCD) reduced sensitivity but not maximum [Ca(2+)](i) induced by ACh. In contrast, both parameters were reduced for the partial muscarinic agonist, pilocarpine. Fluorescence microscopy revealed that mbetaCD disrupted the colocalization of caveolae-1 and M(3)R, but [N-methyl-(3)H]scopolamine receptor-binding assay revealed no effect on muscarinic receptor availability or affinity. To dissect the role of caveolin-1 in ACh-induced [Ca(2+)](i) flux, we disrupted its binding to signaling proteins using either a cell-permeable caveolin-1 scaffolding domain peptide mimetic or by small interfering RNA knockdown. Similar to the effects of mbetaCD, direct targeting of caveolin-1 reduced sensitivity to ACh, but maximum [Ca(2+)](i) mobilization was unaffected. These results indicate caveolae and caveolin-1 facilitate [Ca(2+)](i) mobilization leading to ASM contraction induced by submaximal concentrations of ACh.


Subject(s)
Calcium Signaling , Caveolae/metabolism , Intracellular Space/metabolism , Muscle Contraction/physiology , Muscle, Smooth/metabolism , Receptor, Muscarinic M3/metabolism , Respiratory System/metabolism , Acetylcholine/pharmacology , Animals , Calcium Signaling/drug effects , Caveolae/drug effects , Caveolin 1/chemistry , Cells, Cultured , Cytosol/drug effects , Cytosol/metabolism , Dogs , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , Humans , Intracellular Space/drug effects , Muscle Cells/cytology , Muscle Cells/drug effects , Muscle Contraction/drug effects , Muscle, Smooth/cytology , Muscle, Smooth/drug effects , Muscle, Smooth/ultrastructure , N-Methylscopolamine/metabolism , Protein Structure, Tertiary , RNA, Small Interfering/metabolism , Respiratory System/cytology , Respiratory System/drug effects , Respiratory System/ultrastructure , Trachea/cytology , Trachea/drug effects , Trachea/metabolism , Tritium/metabolism , beta-Cyclodextrins/pharmacology
20.
Am J Physiol Lung Cell Mol Physiol ; 292(3): L654-63, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17085527

ABSTRACT

Hypoxia-induced neonatal persistent pulmonary hypertension (PPHN) is characterized by sustained vasospasm and increased thromboxane (TxA2)-to-prostacyclin ratio. We previously demonstrated that moderate hypoxia induces myocyte TxA2 hypersensitivity. Here, we examined TxA2 prostanoid receptor (TP-R) localization and kinetics following hypoxia to determine the mechanism of hypoxia-induced TxA2 hypersensitivity. Primary cultured neonatal pulmonary artery myocytes were exposed to 10% O2 (hypoxic myocytes; HM) or 21% O2 (normoxic myocytes; NM) for 3 days. PPHN was induced in neonatal piglets by in vivo exposure to 10% FiO2 for 3 days. TP-R was studied in whole lung sections from pigs with hypoxic PPHN- and age-matched controls; intracellular localization was studied by immunocytochemistry. TP-R affinity was studied in cultured myocytes by saturation binding kinetics using 3H-SQ-29548 and competitive binding kinetics by coincubation with U-46619. Phosphorylation and coupling were examined in immunoprecipitated TP-R. We report distal propagation of TP-R expression in PPHN, extending to pulmonary arteries <50 microm. In HM, intracellular TP-R moves towards the perinuclear region, mirroring a change in endoplasmic reticulum (ER) morphology. TP-R kinetics also alter in HM membranes, with decreased Kd and Bmax (maximal binding sites). Additionally, in hypoxia, 3H-SQ-29548 is displaced at lower concentration of U-46619 than in normoxia, suggesting increased agonist affinity. Phosphorylation of serine residues on HM TP-R was significantly decreased compared with NM; this difference correlated with increased Galphaq coupling in hypoxia and was ablated by incubation with PKA. We conclude that the TP-R is normally desensitized in the neonatal pulmonary circuit by PKA-mediated regulatory phosphorylation, decreasing ligand affinity and coupling to Galphaq; this protection is lost following hypoxic exposure. Also, the appearance of TP-R in resistance arteries after development of hypoxic PPHN may contribute to increased pulmonary arterial pressure.


Subject(s)
Hypoxia/physiopathology , Muscle, Smooth, Vascular/drug effects , Myocytes, Smooth Muscle/drug effects , Pulmonary Artery/drug effects , Receptors, Thromboxane A2, Prostaglandin H2/metabolism , Thromboxane A2/metabolism , Vasoconstriction/drug effects , Animals , Animals, Newborn , Binding, Competitive , Calcium/metabolism , Cells, Cultured , Disease Models, Animal , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , Immunoenzyme Techniques , Immunoprecipitation , Kinetics , Ligands , Muscle, Smooth, Vascular/cytology , Myocytes, Smooth Muscle/physiology , Phosphorylation , Pulmonary Artery/cytology , Pulmonary Artery/physiology , Swine
SELECTION OF CITATIONS
SEARCH DETAIL
...