Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 110
1.
Pulm Circ ; 11(4): 20458940211049948, 2021.
Article En | MEDLINE | ID: mdl-34646499

Upon sensing a reduction in local oxygen partial pressure, pulmonary vessels constrict, a phenomenon known as hypoxic pulmonary vasoconstriction. Excessive hypoxic pulmonary vasoconstriction can occur with ascent to high altitude and is a contributing factor to the development of high-altitude pulmonary edema. The carbonic anhydrase inhibitor, acetazolamide, attenuates hypoxic pulmonary vasoconstriction through stimulation of alveolar ventilation via modulation of acid-base homeostasis and by direct effects on pulmonary vascular smooth muscle. In pulmonary arterial smooth muscle cells (PASMCs), acetazolamide prevents hypoxia-induced increases in intracellular calcium concentration ([Ca2+]i), although the exact mechanism by which this occurs is unknown. In this study, we explored the effect of acetazolamide on various calcium-handling pathways in PASMCs. Using fluorescent microscopy, we tested whether acetazolamide directly inhibited store-operated calcium entry or calcium release from the sarcoplasmic reticulum, two well-documented sources of hypoxia-induced increases in [Ca2+]i in PASMCs. Acetazolamide had no effect on calcium entry stimulated by store-depletion, nor on calcium release from the sarcoplasmic reticulum induced by either phenylephrine to activate inositol triphosphate receptors or caffeine to activate ryanodine receptors. In contrast, acetazolamide completely prevented Ca2+-release from the sarcoplasmic reticulum induced by hypoxia (4% O2). Since these results suggest the acetazolamide interferes with a mechanism upstream of the inositol triphosphate and ryanodine receptors, we also determined whether acetazolamide might prevent hypoxia-induced changes in reactive oxygen species production. Using roGFP, a ratiometric reactive oxygen species-sensitive fluorescent probe, we found that hypoxia caused a significant increase in reactive oxygen species in PASMCs that was prevented by 100 µM acetazolamide. Together, these results suggest that acetazolamide prevents hypoxia-induced changes in [Ca2+]i by attenuating reactive oxygen species production and subsequent activation of Ca2+-release from sarcoplasmic reticulum stores.

2.
Vet J ; 202(2): 236-43, 2014 Nov.
Article En | MEDLINE | ID: mdl-24973006

The objective of this study was to measure whole-body protein kinetics in weanling horses receiving forage and one of two different concentrates: (1) commercial crude protein (CCP) concentrate, which with the forage provided 4.1 g CP/kg bodyweight (BW)/day (189 mg lysine (Lys)/kg BW/day), and (2) recommended crude protein (RCP) concentrate which, with the same forage, provided 3.1 g CP/kg BW/day (194 mg Lys/kg BW/day). Blood samples were taken to determine the response of plasma amino acid concentrations to half the daily concentrate allocation. The next day, a 2 h-primed, constant infusion of [(13)C]sodium bicarbonate and a 4 h-primed, constant infusion of [1-(13)C]phenylalanine were used with breath and blood sampling to measure breath (13)CO2 and blood [(13)C]phenylalanine enrichment. Horses on the CCP diet showed an increase from baseline in plasma isoleucine, leucine, lysine, threonine, valine, alanine, arginine, asparagine, glutamine, ornithine, proline, serine, and tyrosine at 120 min post-feeding. Baseline plasma amino acid concentrations were greater with the CCP diet for histidine, isoleucine, leucine, threonine, valine, asparagine, proline, and serine. Phenylalanine, lysine, and methionine were greater in the plasma of horses receiving the RCP treatment at 0 and 120 min. Phenylalanine intake was standardized between groups; however, horses receiving the RCP diet had greater rates of phenylalanine oxidation (P = 0.02) and lower rates of non-oxidative phenylalanine disposal (P = 0.04). Lower whole-body protein synthesis indicates a limiting amino acid in the RCP diet.


Dietary Proteins/metabolism , Horses/metabolism , Protein Biosynthesis , Amino Acids/blood , Animal Feed/analysis , Animals , Diet/veterinary , Horses/growth & development , Weaning
3.
Am J Physiol Lung Cell Mol Physiol ; 304(8): L549-61, 2013 Apr 15.
Article En | MEDLINE | ID: mdl-23418090

Numerous cellular responses to hypoxia are mediated by the transcription factor hypoxia-inducible factor-1 (HIF-1). HIF-1 plays a central role in the pathogenesis of hypoxic pulmonary hypertension. Under certain conditions, HIF-1 may utilize feedforward mechanisms to amplify its activity. Since hypoxia increases endothelin-1 (ET-1) levels in the lung, we hypothesized that during moderate, prolonged hypoxia ET-1 might contribute to HIF-1 signaling in pulmonary arterial smooth muscle cells (PASMCs). Primary cultures of rat PASMCs were treated with ET-1 or exposed to moderate, prolonged hypoxia (4% O(2) for 60 h). Levels of the oxygen-sensitive HIF-1α subunit and expression of HIF target genes were increased in both hypoxic cells and cells treated with ET-1. Both hypoxia and ET-1 also increased HIF-1α mRNA expression and decreased mRNA and protein expression of prolyl hydroxylase 2 (PHD2), which is the protein responsible for targeting HIF-1α for O(2)-dependent degradation. The induction of HIF-1α by moderate, prolonged hypoxia was blocked by BQ-123, an antagonist of ET-1 receptor subtype A. The effects of ET-1 were mediated by increased intracellular calcium, generation of reactive oxygen species, and ERK1/2 activation. Neither ET-1 nor moderate hypoxia induced the expression of HIF-1α or HIF target genes in aortic smooth muscle cells. These results suggest that ET-1 induces a PASMC-specific increase in HIF-1α levels by upregulation of HIF-1α synthesis and downregulation of PHD2-mediated degradation, thereby amplifying the induction of HIF-1α in PASMCs during moderate, prolonged hypoxia.


Endothelin-1/pharmacology , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Pulmonary Artery/drug effects , Pulmonary Artery/metabolism , Animals , Base Sequence , Calcium/metabolism , Cell Hypoxia/genetics , Cell Hypoxia/physiology , Cells, Cultured , Endothelin A Receptor Antagonists , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Expression/drug effects , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor-Proline Dioxygenases , Male , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , Peptides, Cyclic/pharmacology , Procollagen-Proline Dioxygenase/genetics , Procollagen-Proline Dioxygenase/metabolism , Pulmonary Artery/cytology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Wistar , Reactive Oxygen Species/metabolism
5.
Am J Physiol Lung Cell Mol Physiol ; 303(4): L343-53, 2012 Aug 15.
Article En | MEDLINE | ID: mdl-22683574

Pulmonary arterial smooth muscle cell (PASMC) migration is a key component of the vascular remodeling that occurs during the development of hypoxic pulmonary hypertension, although the mechanisms governing this phenomenon remain poorly understood. Aquaporin-1 (AQP1), an integral membrane water channel protein, has recently been shown to aid in migration of endothelial cells. Since AQP1 is expressed in certain types of vascular smooth muscle, we hypothesized that AQP1 would be expressed in PASMCs and would be required for migration in response to hypoxia. Using PCR and immunoblot techniques, we determined the expression of AQPs in pulmonary vascular smooth muscle and the effect of hypoxia on AQP levels, and we examined the role of AQP1 in hypoxia-induced migration in rat PASMCs using Transwell filter assays. Moreover, since the cytoplasmic tail of AQP1 contains a putative calcium binding site and an increase in intracellular calcium concentration ([Ca(2+)](i)) is a hallmark of hypoxic exposure in PASMCs, we also determined whether the responses were Ca(2+) dependent. Results were compared with those obtained in aortic smooth muscle cells (AoSMCs). We found that although AQP1 was abundant in both PASMCs and AoSMCs, hypoxia selectively increased AQP1 protein levels, [Ca(2+)](i), and migration in PASMCs. Blockade of Ca(2+) entry through voltage-dependent Ca(2+) or nonselective cation channels prevented the hypoxia-induced increase in PASMC [Ca(2+)](i), AQP1 levels, and migration. Silencing AQP1 via siRNA also prevented hypoxia-induced migration of PASMCs. Our results suggest that hypoxia induces a PASMC-specific increase in [Ca(2+)](i) that results in increased AQP1 protein levels and cell migration.


Aquaporin 1/genetics , Calcium/metabolism , Cell Movement , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/pathology , Pulmonary Artery/pathology , Up-Regulation/genetics , Animals , Aorta/pathology , Aquaporin 1/metabolism , Cell Hypoxia , Cell Proliferation , Intracellular Space/metabolism , Male , Muscle, Smooth, Vascular/pathology , Rats , Rats, Wistar
6.
Am J Physiol Lung Cell Mol Physiol ; 303(2): L161-8, 2012 Jul.
Article En | MEDLINE | ID: mdl-22582116

In pulmonary arterial smooth muscle cells (PASMC), acute hypoxia increases intracellular Ca(2+) concentration ([Ca(2+)](i)) by inducing Ca(2+) release from the sarcoplasmic reticulum (SR) and Ca(2+) influx through store- and voltage-operated Ca(2+) channels in sarcolemma. To evaluate the mechanisms of hypoxic Ca(2+) release, we measured [Ca(2+)](i) with fluorescent microscopy in primary cultures of rat distal PASMC. In cells perfused with Ca(2+)-free Krebs Ringer bicarbonate solution (KRBS), brief exposures to caffeine (30 mM) and norepinephrine (300 µM), which activate SR ryanodine and inositol trisphosphate receptors (RyR, IP(3)R), respectively, or 4% O(2) caused rapid transient increases in [Ca(2+)](i), indicating intracellular Ca(2+) release. Preexposure of these cells to caffeine, norepinephrine, or the SR Ca(2+)-ATPase inhibitor cyclopiazonic acid (CPA; 10 µM) blocked subsequent Ca(2+) release to caffeine, norepinephrine, and hypoxia. The RyR antagonist ryanodine (10 µM) blocked Ca(2+) release to caffeine and hypoxia but not norepinephrine. The IP(3)R antagonist xestospongin C (XeC, 0.1 µM) blocked Ca(2+) release to norepinephrine and hypoxia but not caffeine. In PASMC perfused with normal KRBS, acute hypoxia caused a sustained increase in [Ca(2+)](i) that was abolished by ryanodine or XeC. These results suggest that in rat distal PASMC 1) the initial increase in [Ca(2+)](i) induced by hypoxia, as well as the subsequent Ca(2+) influx that sustained this increase, required release of Ca(2+) from both RyR and IP(3)R, and 2) the SR Ca(2+) stores accessed by RyR, IP(3)R, and hypoxia functioned as a common store, which was replenished by a CPA-inhibitable Ca(2+)-ATPase.


Calcium Signaling , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Myocytes, Smooth Muscle/metabolism , Pulmonary Artery/cytology , Ryanodine Receptor Calcium Release Channel/metabolism , Sarcoplasmic Reticulum/metabolism , Animals , Caffeine/pharmacology , Calcium/physiology , Calcium Channel Agonists/pharmacology , Calcium Channel Blockers/pharmacology , Calcium-Transporting ATPases/antagonists & inhibitors , Calcium-Transporting ATPases/metabolism , Cell Hypoxia , Cells, Cultured , Indoles/pharmacology , Inositol 1,4,5-Trisphosphate Receptors/agonists , Inositol 1,4,5-Trisphosphate Receptors/antagonists & inhibitors , Macrocyclic Compounds/pharmacology , Male , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/physiology , Norepinephrine/pharmacology , Oxazoles/pharmacology , Rats , Rats, Wistar , Ryanodine/pharmacology , Sarcoplasmic Reticulum/drug effects
7.
Am J Physiol Lung Cell Mol Physiol ; 302(10): L1128-39, 2012 May 15.
Article En | MEDLINE | ID: mdl-22387294

Exposure to chronic hypoxia (CH) causes pulmonary hypertension. The vasoconstrictor endothelin-1 (ET-1) is thought to play a role in the development of hypoxic pulmonary hypertension. In pulmonary arterial smooth muscle cells (PASMCs) from chronically hypoxic rats, ET-1 signaling is altered, with the ET-1-induced change in intracellular calcium concentration (Δ[Ca(2+)](i)) occurring through activation of voltage-dependent Ca(2+) channels (VDCC) even though ET-1-induced depolarization via inhibition of K(+) channels is lost. The mechanism underlying this response is unclear. We hypothesized that activation of VDCCs by ET-1 following CH might be mediated by protein kinase C (PKC) and/or Rho kinase, both of which have been shown to phosphorylate and activate VDCCs. To test this hypothesis, we examined the effects of PKC and Rho kinase inhibitors on the ET-1-induced Δ[Ca(2+)](i) in PASMCs from rats exposed to CH (10% O(2), 3 wk) using the Ca(2+)-sensitive dye fura 2-AM and fluorescent microscopy techniques. We found that staurosporine and GF109203X, inhibitors of PKC, and Y-27632 and HA 1077, Rho kinase inhibitors, reduced the ET-1-induced Δ[Ca(2+)](i) by >70%. Inhibition of tyrosine kinases (TKs) with genistein or tyrphostin A23, or combined inhibition of PKC, TKs, and Rho kinase, reduced the Δ[Ca(2+)](i) to a similar extent as inhibition of either PKC or Rho kinase alone. The ability of PKC or Rho kinase to activate VDCCs in our cells was verified using phorbol 12-myristate 13-acetate and GTP-γ-S. These results suggest that following CH, the ET-1-induced Δ[Ca(2+)](i) in PASMCs occurs via Ca(2+) influx through VDCCs mediated primarily by PKC, TKs, and Rho kinase.


Calcium Signaling , Endothelin-1/metabolism , Hypoxia/metabolism , Muscle Cells/metabolism , Protein Kinase C/metabolism , rho-Associated Kinases/metabolism , Animals , Calcium/metabolism , Calcium Channels/drug effects , Calcium Channels/metabolism , Chronic Disease , Endothelin-1/pharmacology , Fluorescent Dyes , Fura-2/analogs & derivatives , Gene Expression , Hypertension, Pulmonary/etiology , Hypertension, Pulmonary/metabolism , Hypertension, Pulmonary/physiopathology , Hypoxia/complications , Hypoxia/physiopathology , Ion Channel Gating/drug effects , Male , Microscopy, Fluorescence , Muscle Cells/drug effects , Muscle Cells/pathology , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/pathology , Protein Kinase C/genetics , Protein Kinase Inhibitors/pharmacology , Rats , Rats, Wistar , rho-Associated Kinases/genetics
8.
Physiol Rev ; 92(1): 367-520, 2012 Jan.
Article En | MEDLINE | ID: mdl-22298659

It has been known for more than 60 years, and suspected for over 100, that alveolar hypoxia causes pulmonary vasoconstriction by means of mechanisms local to the lung. For the last 20 years, it has been clear that the essential sensor, transduction, and effector mechanisms responsible for hypoxic pulmonary vasoconstriction (HPV) reside in the pulmonary arterial smooth muscle cell. The main focus of this review is the cellular and molecular work performed to clarify these intrinsic mechanisms and to determine how they are facilitated and inhibited by the extrinsic influences of other cells. Because the interaction of intrinsic and extrinsic mechanisms is likely to shape expression of HPV in vivo, we relate results obtained in cells to HPV in more intact preparations, such as intact and isolated lungs and isolated pulmonary vessels. Finally, we evaluate evidence regarding the contribution of HPV to the physiological and pathophysiological processes involved in the transition from fetal to neonatal life, pulmonary gas exchange, high-altitude pulmonary edema, and pulmonary hypertension. Although understanding of HPV has advanced significantly, major areas of ignorance and uncertainty await resolution.


Hypoxia/physiopathology , Pulmonary Alveoli/blood supply , Vasoconstriction/physiology , Altitude Sickness/physiopathology , Cell Communication , Humans , Hypertension, Pulmonary/physiopathology , Infant, Newborn , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/physiology , Pulmonary Alveoli/embryology , Pulmonary Alveoli/growth & development , Pulmonary Gas Exchange/physiology
9.
Proc Natl Acad Sci U S A ; 109(4): 1239-44, 2012 Jan 24.
Article En | MEDLINE | ID: mdl-22232678

Chronic hypoxia is an inciting factor for the development of pulmonary arterial hypertension. The mechanisms involved in the development of hypoxic pulmonary hypertension (HPH) include hypoxia-inducible factor 1 (HIF-1)-dependent transactivation of genes controlling pulmonary arterial smooth muscle cell (PASMC) intracellular calcium concentration ([Ca(2+)](i)) and pH. Recently, digoxin was shown to inhibit HIF-1 transcriptional activity. In this study, we tested the hypothesis that digoxin could prevent and reverse the development of HPH. Mice were injected daily with saline or digoxin and exposed to room air or ambient hypoxia for 3 wk. Treatment with digoxin attenuated the development of right ventricle (RV) hypertrophy and prevented the pulmonary vascular remodeling and increases in PASMC [Ca(2+)](i), pH, and RV pressure that occur in mice exposed to chronic hypoxia. When started after pulmonary hypertension was established, digoxin attenuated the hypoxia-induced increases in RV pressure and PASMC pH and [Ca(2+)](i). These preclinical data support a role for HIF-1 inhibitors in the treatment of HPH.


Digoxin/pharmacology , Hypertension, Pulmonary/prevention & control , Hypoxia-Inducible Factor 1/metabolism , Hypoxia/complications , Transcriptional Activation/physiology , Analysis of Variance , Animals , Blood Pressure/drug effects , Calcium/metabolism , Digoxin/blood , Hypertension, Pulmonary/etiology , Hypertrophy, Right Ventricular/prevention & control , Hypoxia-Inducible Factor 1/antagonists & inhibitors , Mice , Microscopy, Confocal , Myocytes, Smooth Muscle/metabolism , Pulmonary Artery/cytology , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Transcriptional Activation/drug effects
10.
Am J Physiol Lung Cell Mol Physiol ; 301(3): L380-7, 2011 Sep.
Article En | MEDLINE | ID: mdl-21665962

Hypoxic contraction of pulmonary arterial smooth muscle is thought to require increases in both intracellular Ca(2+) concentration ([Ca(2+)](i)) and myofilament Ca(2+) sensitivity, which may or may not be endothelium-dependent. To examine the effects of hypoxia and endothelium on Ca(2+) sensitivity in pulmonary arterial smooth muscle, we measured the relation between [Ca(2+)](i) and isometric force at 37°C during normoxia (21% O(2)-5% CO(2)) and after 30 min of hypoxia (1% O(2)-5% CO(2)) in endothelium-intact (E+) and -denuded (E-) rat distal intrapulmonary arteries (IPA) permeabilized with staphylococcal α-toxin. Endothelial denudation enhanced Ca(2+) sensitivity during normoxia but did not alter the effects of hypoxia, which shifted the [Ca(2+)](i)-force relation to higher force in E+ and E- IPA. Neither hypoxia nor endothelial denudation altered Ca(2+) sensitivity in mesenteric arteries. In E+ and E- IPA, hypoxic enhancement of Ca(2+) sensitivity was abolished by the nitric oxide synthase inhibitor N(ω)-nitro-l-arginine methyl ester (30 µM), which shifted normoxic [Ca(2+)](i)-force relations to higher force. In E- IPA, the Rho kinase antagonist Y-27632 (10 µM) shifted the normoxic [Ca(2+)](i)-force relation to lower force but did not alter the effects of hypoxia. These results suggest that acute hypoxia enhanced myofilament Ca(2+) sensitivity in rat IPA by decreasing nitric oxide production and/or activity in smooth muscle, thereby revealing a high basal level of Ca(2+) sensitivity, due in part to Rho kinase, which otherwise did not contribute to Ca(2+) sensitization by hypoxia.


Actin Cytoskeleton/drug effects , Calcium/pharmacology , Hypoxia/physiopathology , Pulmonary Artery/drug effects , Animals , Male , NG-Nitroarginine Methyl Ester/pharmacology , Rats , Rats, Wistar , Vasoconstriction , rho-Associated Kinases/metabolism
11.
J Dairy Sci ; 92(8): 3849-60, 2009 Aug.
Article En | MEDLINE | ID: mdl-19620669

Methane is an end product of ruminal fermentation that is energetically wasteful and contributes to global climate change. Bromoethanesulfonate, animal-vegetable fat, and monensin were compared with a control treatment to suppress different functional groups of ruminal prokaryotes in the presence or absence of protozoa to evaluate changes in fermentation, digestibility, and microbial N outflow. Four dual-flow continuous culture fermenter systems were used in 4 periods in a 4 x 4 Latin square design split into 2 subperiods. In subperiod 1, a multistage filter system (50-microm smallest pore size) retained most protozoa. At the start of subperiod 2, conventional filters (300-microm pore size) were substituted to efflux protozoa via filtrate pumps over 3 d; after a further 7 d of adaptation, the fermenters were sampled for 3 d. Treatments were retained during both subperiods. Flow of total N and digestibilities of NDF and OM were 18, 16, and 9% higher, respectively, for the defaunated subperiod but were not different among treatments. Ammonia concentration was 33% higher in the faunated fermenters but was not affected by treatment. Defaunation increased the flow of nonammonia N and bacterial N from the fermenters. Protozoal counts were not different among treatments, but bromoethanesulfonate increased the generation time from 43.2 to 55.6 h. Methanogenesis was unaffected by defaunation but tended to be increased by unsaturated fat. Defaunation did not affect total volatile fatty acid production but decreased the acetate:propionate ratio; monensin increased production of isovalerate and valerate. Biohydrogenation of unsaturated fatty acids was impaired in the defaunated fermenters because effluent flows of oleic, linoleic, and linolenic acids were 60, 77, and 69% higher, and the ratio of vaccenic acid:unsaturated FA ratio was decreased by 34% in the effluent. This ratio was increased in both subperiods with the added fat diet, indicating an accumulation of intermediates of biohydrogenation. However, the flow of 18:2 conjugated linoleic acid was unaffected by defaunation or by treatments other than added fat. The flows of trans-10, trans-11, and total trans-18:1 fatty acids were not affected by monensin or faunation status.


Alkanesulfonates/pharmacology , Dietary Fats, Unsaturated/metabolism , Eukaryota , Fermentation/drug effects , Hydrocarbons, Brominated/pharmacology , Monensin/pharmacology , Protein Biosynthesis/drug effects , Rumen , Ammonia/analysis , Animals , Anti-Infective Agents/pharmacology , Antiprotozoal Agents/pharmacology , Bacteria/drug effects , Bacteria/metabolism , Cattle , Culture Techniques , Eukaryota/drug effects , Eukaryota/metabolism , Eukaryota/physiology , Fatty Acids, Volatile/analysis , Female , Gastrointestinal Contents/chemistry , Hydrogenation/drug effects , Methane/metabolism , Nitrogen/analysis , Nitrogen/metabolism , Rumen/metabolism , Rumen/parasitology
12.
Am J Physiol Lung Cell Mol Physiol ; 297(1): L17-25, 2009 Jul.
Article En | MEDLINE | ID: mdl-19395668

Stromal interaction molecule 1 (STIM1) is a recently discovered membrane-spanning protein thought to sense lumenal Ca(2+) in sarco(endo)plasmic reticulum (SR/ER) and transduce activation of Ca(2+)-permeable store-operated channels (SOC) in plasmalemma in response to SR/ER Ca(2+) depletion. To evaluate the role of STIM1 and a closely related protein, STIM2, in Ca(2+) signaling of rat distal pulmonary arterial smooth muscle cells (PASMC) during hypoxia, we used fluorescent microscopy and the Ca(2+)-sensitive dye, fura 2, to measure basal intracellular Ca(2+) concentration ([Ca(2+)](i)), store-operated Ca(2+) entry (SOCE), and increases in [Ca(2+)](i) caused by acute hypoxia (4% O(2)) or depolarization (60 mmol/l KCl) in cells treated with small interfering RNA targeted to STIM1 (siSTIM1) or STIM2 (siSTIM2). As determined by real-time quantitative PCR analysis (qPCR), STIM1 mRNA was 200-fold more abundant than STIM2 in untreated control PASMC. siSTIM1 and siSTIM2 caused specific and significant knockdown of both mRNA measured by qPCR and protein measured by Western blotting. siSTIM1 markedly inhibited SOCE and abolished the sustained [Ca(2+)](i) response to hypoxia but did not alter the initial transient [Ca(2+)](i) response to hypoxia, the [Ca(2+)](i) response to depolarization, or basal [Ca(2+)](i). The only effect of siSTIM2 was a smaller inhibition of SOCE. We conclude that STIM1 was quantitatively more important than STIM2 in activation of SOC in rat distal PASMC and that the increase in [Ca(2+)](i) induced by acute hypoxia in these cells required SR Ca(2+) release and STIM1-dependent activation of SOC.


Calcium Signaling , Gene Knockdown Techniques , Hypoxia/metabolism , Membrane Glycoproteins/metabolism , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/pathology , Pulmonary Artery/cytology , Acute Disease , Animals , Blotting, Western , Calcium Signaling/drug effects , Chlorides/pharmacology , Fluorescence , Fura-2/metabolism , Gene Expression Regulation/drug effects , Hypoxia/pathology , Indoles/pharmacology , Manganese Compounds/pharmacology , Membrane Glycoproteins/genetics , Myocytes, Smooth Muscle/drug effects , Potassium Chloride/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Stromal Interaction Molecule 1 , TRPC Cation Channels/genetics , TRPC Cation Channels/metabolism , Time Factors
13.
J Dairy Sci ; 92(1): 256-69, 2009 Jan.
Article En | MEDLINE | ID: mdl-19109285

Defaunation studies have documented decreased ammonia concentrations associated with reduced microbial protein recycling and wastage of dietary protein, whereas many methods to suppress protozoa can reduce feed intake or depress ruminal organic matter or fiber digestibility. Therefore, more research is needed to optimize dietary conditions that improve protozoal growth and ruminal outflow relative to autolysis and recycling. Response in growth rate to ruminal outflow was simulated by abrupt changes in transfer interval of batch cultures, and substrate availability was evaluated by feeding without or with abrupt addition of monensin, which was postulated to inhibit digestive vacuole function. In experiment 1, Entodinium caudatum, a mix of Entodinium species, Epidinium caudatum, or Ophryoscolex caudatus cultures rapidly adjusted their generation times to approach respective changes in transfer interval from 3 to 2 or 1 d (cultures were always fed at 24-h intervals). Monensin (0.25 microM) consistently delayed this response. To evaluate a metabolic upshift associated with feeding or a downshift associated with substrate depletion, experiment 2 used real-time PCR to quantify protozoal 18S rRNA gene (rDNA) copies that were expressed relative to cell numbers or to the cellular constituents N and nucleic acids after feeding without or with monensin (0.5 microM). The 18S rDNA copies per milligram of nucleic acids were least for Ophryoscolex compared with the other cultures. When averaged over cultures (no culture x treatment interaction), 18S rDNA copies per unit of nucleic acids decreased at 16 h when cultures were starved but increased with feeding unless monensin uncoupled availability of consumed substrate. Rumen protozoal growth increased in response to decreased transfer interval in experiment 1. Substrate availability appeared to initiate metabolic responses preparing for cell growth, explaining how cultures could rapidly adjust to decreasing transfer interval in experiment 2. Because feeding was not coupled with transfer in experiment 2, however, a metabolic control probably arrested cell division to prevent overgrowth relative to substrate availability.


Antiprotozoal Agents/pharmacology , Ciliophora/drug effects , Ciliophora/physiology , Gene Dosage/genetics , Monensin/pharmacology , Rumen/parasitology , Starvation , Animals , Cattle/parasitology , Cell Division/drug effects , Cell Division/physiology , Ciliophora/cytology , Ciliophora/genetics , Culture Techniques , DNA, Ribosomal/genetics , RNA, Ribosomal, 18S/genetics , Regression Analysis , Time Factors
14.
Am J Physiol Lung Cell Mol Physiol ; 295(1): L104-13, 2008 Jul.
Article En | MEDLINE | ID: mdl-18424621

Hypoxic pulmonary vasoconstriction (HPV) requires Ca(2+) influx through store-operated Ca(2+) channels (SOCC) in pulmonary arterial smooth muscle cells (PASMC) and is greater in distal than proximal pulmonary arteries (PA). SOCC may be composed of canonical transient receptor potential (TRPC) proteins and activated by stromal interacting molecule 1 (STIM1). To assess the possibility that HPV is greater in distal PA because store-operated Ca(2+) entry (SOCE) is greater in distal PASMC, we measured intracellular Ca(2+) concentration ([Ca(2+)](i)) and SOCE in primary cultures of PASMC using fluorescent microscopy and the Ca(2+)-sensitive dye fura 2. Both hypoxia (4% O(2)) and KCl (60 mM) increased [Ca(2+)](i). Responses to hypoxia, but not KCl, were greater in distal cells. We measured SOCE in PASMC perfused with Ca(2+)-free solutions containing cyclopiazonic acid to deplete Ca(2+) stores in sarcoplasmic reticulum and nifedipine to prevent Ca(2+) entry through L-type voltage-operated Ca(2+) channels. Under these conditions, the increase in [Ca(2+)](i) caused by restoration of extracellular Ca(2+) and the decrease in fura 2 fluorescence caused by Mn(2+) were greater in distal PASMC, indicating greater SOCE. Moreover, the increase in SOCE caused by hypoxia was also greater in distal cells. Real-time quantitative polymerase chain reaction analysis of PASMC and freshly isolated deendothelialized PA tissue demonstrated expression of STIM1 and five of seven known TRPC isoforms (TRPC1 > TRPC6 > TRPC4 >> TRPC3 approximately TRPC5). For both protein, as measured by Western blotting, and mRNA, expression of STIM1, TRPC1, TRPC6, and TRPC4 was greater in distal than proximal PASMC and PA. These results provide further support for the importance of SOCE in HPV and suggest that HPV is greater in distal than proximal PA because greater numbers and activation of SOCC in distal PASMC generate bigger increases in [Ca(2+)](i).


Calcium Signaling , Calcium/metabolism , Hypoxia/metabolism , Membrane Glycoproteins/biosynthesis , Myocytes, Smooth Muscle/metabolism , Pulmonary Artery/metabolism , TRPC Cation Channels/biosynthesis , Animals , Gene Expression Regulation , Hypoxia/pathology , Male , Myocytes, Smooth Muscle/pathology , Organ Specificity , Protein Isoforms/biosynthesis , Pulmonary Artery/pathology , RNA, Messenger/biosynthesis , Rats , Rats, Wistar , Sarcoplasmic Reticulum/metabolism , Sarcoplasmic Reticulum/pathology , Stromal Interaction Molecule 1
15.
Am J Physiol Lung Cell Mol Physiol ; 294(2): L309-18, 2008 Feb.
Article En | MEDLINE | ID: mdl-18065659

Prolonged exposure to decreased oxygen tension causes contraction and proliferation of pulmonary arterial smooth muscle cells (PASMCs) and pulmonary hypertension. Hypoxia-induced inhibition of voltage-gated K(+) (K(v)) channels may contribute to the development of pulmonary hypertension by increasing intracellular calcium concentration ([Ca(2+)](i)). The peptide endothelin-1 (ET-1) has been implicated in the development of pulmonary hypertension and acutely decreases K(v) channel activity. ET-1 also activates several transcription factors, although whether ET-1 alters K(V) channel expression is unclear. The hypoxic induction of ET-1 is regulated by the transcription factor hypoxia-inducible factor-1 (HIF-1), which we demonstrated to regulate hypoxia-induced decreases in K(V) channel activity. In this study, we tested the hypothesis that HIF-1-dependent increases in ET-1 lead to decreased K(v) channel expression and subsequent elevation in [Ca(2+)](i). Resting [Ca(2+)](i) and K(v) channel expression were measured in cells exposed to control (18% O(2), 5% CO(2)) and hypoxic (4% O(2), 5% CO(2)) conditions. Hypoxia caused a decrease in expression of K(v)1.5 and K(v)2.1 and a significant increase in resting [Ca(2+)](i). The increase in [Ca(2+)](i) was reduced by nifedipine, an inhibitor of voltage-dependent calcium channels, and removal of extracellular calcium. Treatment with BQ-123, an ET-1 receptor inhibitor, prevented the hypoxia-induced decrease in K(v) channel expression and blunted the hypoxia-induced increase in [Ca(2+)](i) in PASMCs, whereas ET-1 mimicked the effects of hypoxia. Both hypoxia and overexpression of HIF-1 under normoxic conditions increased ET-1 expression. These results suggest that the inhibition of K(v) channel expression and rise in [Ca(2+)](i) during chronic hypoxia may be the result of HIF-1-dependent induction of ET-1.


Endothelin-1/metabolism , Hypoxia/metabolism , Kv1.5 Potassium Channel/metabolism , Myocytes, Smooth Muscle/metabolism , Pulmonary Artery/cytology , Pulmonary Artery/metabolism , Shab Potassium Channels/metabolism , Animals , Calcium Signaling/drug effects , Gene Expression Regulation/drug effects , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Kv1.5 Potassium Channel/genetics , Male , Mice , Models, Biological , Myocytes, Smooth Muscle/drug effects , Oligopeptides/pharmacology , Peptides, Cyclic/pharmacology , Perfusion , Piperidines/pharmacology , Pulmonary Artery/drug effects , Rats , Rats, Wistar , Shab Potassium Channels/genetics
16.
Am J Physiol Lung Cell Mol Physiol ; 293(3): L674-85, 2007 Sep.
Article En | MEDLINE | ID: mdl-17575009

Antagonists of myosin light chain (MLC) kinase (MLCK) and Rho kinase (ROK) are thought to inhibit hypoxic pulmonary vasoconstriction (HPV) by decreasing the concentration of phosphorylated MLC at any intracellular Ca(2+) concentration ([Ca(2+)](i)) in pulmonary arterial smooth muscle cells (PASMC); however, these antagonists can also decrease [Ca(2+)](i). To determine whether MLCK and ROK antagonists alter Ca(2+) signaling in HPV, we measured the effects of ML-9, ML-7, Y-27632, and HA-1077 on [Ca(2+)](i), Ca(2+) entry, and Ca(2+) release in rat distal PASMC exposed to hypoxia or depolarizing concentrations of KCl. We performed parallel experiments in isolated rat lungs to confirm the inhibitory effects of these agents on pulmonary vasoconstriction. Our results demonstrate that MLCK and ROK antagonists caused concentration-dependent inhibition of hypoxia-induced increases in [Ca(2+)](i) in PASMC and HPV in isolated lungs and suggest that this inhibition was due to blockade of Ca(2+) release from the sarcoplasmic reticulum and Ca(2+) entry through store- and voltage-operated Ca(2+) channels in PASMC. Thus MLCK and ROK antagonists might block HPV by inhibiting Ca(2+) signaling, as well as the actin-myosin interaction, in PASMC. If effects on Ca(2+) signaling were due to decreased phosphorylated myosin light chain concentration, their diversity suggests that MLCK and ROK antagonists may have acted by inhibiting myosin motors and/or altering the cytoskeleton in a manner that prevented achievement of required spatial relationships among the cellular components of the response.


Calcium Signaling/drug effects , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Lung/blood supply , Myosin-Light-Chain Kinase/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Vasoconstriction/drug effects , Animals , Blood Pressure/drug effects , Calcium Channels, L-Type/metabolism , Cell Hypoxia , In Vitro Techniques , Lung/drug effects , Lung/enzymology , Male , Membrane Potentials/drug effects , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/enzymology , Pulmonary Artery/drug effects , Pulmonary Artery/enzymology , Rats , Rats, Wistar , rho-Associated Kinases
17.
Am J Physiol Lung Cell Mol Physiol ; 292(4): L1002-12, 2007 Apr.
Article En | MEDLINE | ID: mdl-17209136

Hypoxic pulmonary vasoconstriction (HPV) occurs with ascent to high altitude and can contribute to development of high altitude pulmonary edema (HAPE). Vascular smooth muscle contains carbonic anhydrase (CA), and acetazolamide (AZ), a CA inhibitor, blunts HPV and might be useful in the prevention of HAPE. The mechanism by which AZ impairs HPV is uncertain. Originally developed as a diuretic, AZ also has direct effects on systemic vascular smooth muscle, including modulation of pH and membrane potential; however, the effect of AZ on pulmonary arterial smooth muscle cells (PASMCs) is unknown. Since HPV requires Ca2+ influx into PASMCs and can be modulated by pH, we hypothesized that AZ alters hypoxia-induced changes in PASMC intracellular pH (pH(i)) or Ca2+ concentration ([Ca2+](i)). Using fluorescent microscopy, we tested the effect of AZ as well as two other potent CA inhibitors, benzolamide and ethoxzolamide, which exhibit low and high membrane permeability, respectively, on hypoxia-induced responses in PASMCs. Hypoxia caused a significant increase in [Ca2+](i) but no change in pH(i). All three CA inhibitors slightly decreased basal pH(i), but only AZ caused a concentration-dependent decrease in the [Ca2+](i) response to hypoxia. AZ had no effect on the KCl-induced increase in [Ca2+](i) or membrane potential. N-methyl-AZ, a synthesized compound lacking the unsubstituted sulfonamide group required for CA inhibition, had no effect on pH(i) but inhibited hypoxia-induced Ca2+ responses. These results suggest that AZ attenuates HPV by selectively inhibiting hypoxia-induced Ca2+ responses via a mechanism independent of CA inhibition, changes in pH(i), or membrane potential.


Acetazolamide/pharmacology , Calcium/physiology , Carbonic Anhydrase Inhibitors/pharmacology , Carbonic Anhydrases/metabolism , Hypoxia/physiopathology , Muscle, Smooth, Vascular/physiology , Pulmonary Artery/physiology , Benzolamide/pharmacology , Calcium Signaling/drug effects , Ethoxzolamide/pharmacology , Hydrogen-Ion Concentration , Methazolamide/pharmacology
18.
J Dairy Sci ; 90(2): 798-809, 2007 Feb.
Article En | MEDLINE | ID: mdl-17235157

Methionine supplemented as 2-hydroxy-4-(methylthio)-butanoic acid (HMB) has been suggested to alter bacterial or protozoal populations in the rumen. Our objective was to determine if source of Met would change microbial populations in the rumen and to compare those results to samples from the omasum. The ruminal and omasal samples were collected from cows fed control (no Met), dl-Met, HMB, or the isopropyl ester of HMB (HMBi; estimated 50% rumen protection) in a replicated 4 x 4 Latin square design. In one square, changes in protozoal populations were determined using microscopic counts and denaturing gradient gel electrophoresis (DGGE), whereas changes in bacterial populations were determined using DGGE and ribosomal intergenic spacer length polymorphism (RIS-LP). Neither the protozoal counts nor the DGGE banding patterns derived from protozoa were different among the dietary treatments or for ruminal vs. omasal samples. As revealed by both DGGE and RIS-LP, bacterial populations clustered by treatments in ruminal and especially in omasal samples. Using cows from both Latin squares, the flow of protozoal cells from the rumen was quantified by multiplying protozoal cell count in omasal fluid by the omasal fluid flow (using CoEDTA as a liquid flow marker) or was estimated by rumen pool size of cells multiplied by either the ruminal dilution rate of CoEDTA (after termination of CoEDTA dosing) or the passage rate of Yb-marked particles. Compared with the omasal fluid flow measurement (16.4 h), protozoal generation time was approximated much more closely using the particulate than the fluid passage rate from the rumen (generation times of 15.7 and 7.5 h, respectively). There seems to be minimal selective retention of protozoal genera in the rumen in dairy cattle fed every 2 h. Data support the validity of the omasal sampling technique under our conditions.


Bacteria/growth & development , Cattle , Eukaryota/growth & development , Methionine/administration & dosage , Rumen/microbiology , Rumen/parasitology , Animals , Bacteria/genetics , Cattle/microbiology , Cattle/parasitology , DNA, Bacterial/analysis , Diet , Dietary Supplements , Duodenum/microbiology , Duodenum/parasitology , Electrophoresis , Female , Hydrogen-Ion Concentration , Lactobacillus/genetics , Lactobacillus/growth & development , Methionine/analogs & derivatives , Omasum/microbiology , Omasum/parasitology
19.
Microcirculation ; 13(8): 657-70, 2006 Dec.
Article En | MEDLINE | ID: mdl-17085426

Many chronic lung diseases are associated with prolonged exposure to alveolar hypoxia, resulting in the development of pulmonary hypertension. While the exact mechanisms underlying the pathogenesis of hypoxic pulmonary hypertension remain poorly understood, a key role for changes in Ca2+ homeostasis has emerged. Intracellular Ca2+ concentration controls a variety of pulmonary vascular cell functions, including contraction, gene expression, growth, barrier function and synthesis of vasoactive substances. Several studies indicate that prolonged exposure to hypoxia causes alterations in the expression and activity of several Ca2+ handling pathways in pulmonary arterial smooth muscle cells. In contrast, the effect of chronic hypoxia on Ca2+ homeostasis in pulmonary arterial endothelial cells is relatively unexplored. In this review, we discuss data from our laboratory and others describing the effects of prolonged hypoxia on pulmonary vascular smooth muscle and endothelial cell Ca2+ homeostasis and the various Ca2+ channels and handling pathways involved in these responses. We will also highlight future directions of investigation that might improve our understanding of the response of pulmonary vascular cells to chronic hypoxia.


Calcium Channels/metabolism , Calcium Signaling , Hypertension, Pulmonary/metabolism , Hypoxia/metabolism , Lung/metabolism , Animals , Chronic Disease , Endothelial Cells/metabolism , Endothelial Cells/pathology , Humans , Hypertension, Pulmonary/pathology , Hypoxia/pathology , Lung/blood supply , Lung/pathology , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/pathology
20.
Circ Res ; 98(12): 1528-37, 2006 Jun 23.
Article En | MEDLINE | ID: mdl-16709899

Chronic hypoxia (CH) causes pulmonary vasoconstriction because of increased pulmonary arterial smooth muscle cell (PASMC) contraction and proliferation. We previously demonstrated that intracellular Ca(2+) concentration ([Ca(2+)](i)) was elevated in PASMCs from chronically hypoxic rats because of Ca(2+) influx through pathways other than L-type Ca(2+) channels and that development of hypoxic pulmonary hypertension required full expression of the transcription factor hypoxia inducible factor 1 (HIF-1). In this study, we examined the effect of CH on the activity and expression of store-operated Ca(2+) channels (SOCCs) and the regulation of these channels by HIF-1. Capacitative Ca(2+) entry (CCE) was enhanced in PASMCs from intrapulmonary arteries of rats exposed to CH (10% O(2); 21 days), and exposure to Ca(2+)-free extracellular solution or SOCC antagonists (SKF96365 or NiCl(2)) decreased resting [Ca(2+)](i) in these cells. Expression of TRPC1 and TRPC6, but not TRPC4, mRNA and protein was increased in PASMCs from rats and wild-type mice exposed to CH, in PASMCs from normoxic animals cultured under hypoxic conditions (4% O(2); 60 hours), and in PASMCs in which HIF-1 was overexpressed under nonhypoxic conditions. Hypoxia-induced increases in basal [Ca(2+)](i) and TRPC expression were absent in mice partially deficient for HIF-1. These results suggest that increased TRPC expression, leading to enhanced CCE through SOCCs, may contribute to hypoxic pulmonary hypertension by facilitating Ca(2+) influx and increasing basal [Ca(2+)](i) in PASMCs and that this response is mediated by HIF-1.


Calcium/metabolism , Hypoxia-Inducible Factor 1/metabolism , Hypoxia/metabolism , Intracellular Membranes/metabolism , Myocytes, Smooth Muscle/metabolism , Pulmonary Artery/metabolism , TRPC Cation Channels/metabolism , Animals , Calcium Channel Blockers/pharmacology , Calcium Channels/metabolism , Cells, Cultured , Chronic Disease , Hypoxia-Inducible Factor 1/deficiency , In Vitro Techniques , Mice , Osmolar Concentration , Rats , TRPC6 Cation Channel , Vasoconstriction/drug effects
...