Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 154
Filter
1.
Sci Rep ; 11(1): 14961, 2021 07 22.
Article in English | MEDLINE | ID: mdl-34294757

ABSTRACT

Influenza and other respiratory viruses present a significant threat to public health, national security, and the world economy, and can lead to the emergence of global pandemics such as from COVID-19. A barrier to the development of effective therapeutics is the absence of a robust and predictive preclinical model, with most studies relying on a combination of in vitro screening with immortalized cell lines and low-throughput animal models. Here, we integrate human primary airway epithelial cells into a custom-engineered 96-device platform (PREDICT96-ALI) in which tissues are cultured in an array of microchannel-based culture chambers at an air-liquid interface, in a configuration compatible with high resolution in-situ imaging and real-time sensing. We apply this platform to influenza A virus and coronavirus infections, evaluating viral infection kinetics and antiviral agent dosing across multiple strains and donor populations of human primary cells. Human coronaviruses HCoV-NL63 and SARS-CoV-2 enter host cells via ACE2 and utilize the protease TMPRSS2 for spike protein priming, and we confirm their expression, demonstrate infection across a range of multiplicities of infection, and evaluate the efficacy of camostat mesylate, a known inhibitor of HCoV-NL63 infection. This new capability can be used to address a major gap in the rapid assessment of therapeutic efficacy of small molecules and antiviral agents against influenza and other respiratory viruses including coronaviruses.


Subject(s)
Antiviral Agents/pharmacology , Coronavirus Infections/virology , Influenza, Human/virology , Microbial Sensitivity Tests/instrumentation , Microfluidic Analytical Techniques/instrumentation , Respiratory Mucosa/cytology , Bronchi/cytology , Bronchi/virology , COVID-19/virology , Cell Culture Techniques/instrumentation , Cell Line , Coronavirus/drug effects , Coronavirus Infections/drug therapy , Equipment Design , High-Throughput Screening Assays/instrumentation , Humans , Influenza A virus/drug effects , Influenza, Human/drug therapy , Respiratory Mucosa/virology , Respiratory Tract Infections/drug therapy , Respiratory Tract Infections/virology , SARS-CoV-2/drug effects , COVID-19 Drug Treatment
2.
Lab Chip ; 21(8): 1454-1474, 2021 04 20.
Article in English | MEDLINE | ID: mdl-33881130

ABSTRACT

Drug development suffers from a lack of predictive and human-relevant in vitro models. Organ-on-chip (OOC) technology provides advanced culture capabilities to generate physiologically appropriate, human-based tissue in vitro, therefore providing a route to a predictive in vitro model. However, OOC technologies are often created at the expense of throughput, industry-standard form factors, and compatibility with state-of-the-art data collection tools. Here we present an OOC platform with advanced culture capabilities supporting a variety of human tissue models including liver, vascular, gastrointestinal, and kidney. The platform has 96 devices per industry standard plate and compatibility with contemporary high-throughput data collection tools. Specifically, we demonstrate programmable flow control over two physiologically relevant flow regimes: perfusion flow that enhances hepatic tissue function and high-shear stress flow that aligns endothelial monolayers. In addition, we integrate electrical sensors, demonstrating quantification of barrier function of primary gut colon tissue in real-time. We utilize optical access to the tissues to directly quantify renal active transport and oxygen consumption via integrated oxygen sensors. Finally, we leverage the compatibility and throughput of the platform to screen all 96 devices using high content screening (HCS) and evaluate gene expression using RNA sequencing (RNA-seq). By combining these capabilities in one platform, physiologically-relevant tissues can be generated and measured, accelerating optimization of an in vitro model, and ultimately increasing predictive accuracy of in vitro drug screening.


Subject(s)
Drug Development , Lab-On-A-Chip Devices , Humans , Liver , Perfusion , Workflow
3.
Acta Physiol (Oxf) ; 220(1): 72-82, 2017 05.
Article in English | MEDLINE | ID: mdl-27636900

ABSTRACT

AIM: Increasing evidence demonstrates that circadian clock proteins are important regulators of physiological functions including blood pressure. An established risk factor for developing cardiovascular disease is the absence of a blood pressure dip during the inactive period. The goal of the present study was to determine the effects of a high salt diet plus mineralocorticoid on PER1-mediated blood pressure regulation in a salt-resistant, normotensive mouse model, C57BL/6J. METHODS: Blood pressure was measured using radiotelemetry. After control diet, wild-type (WT) and Per1 (KO) knockout mice were given a high salt diet (4% NaCl) and the long-acting mineralocorticoid deoxycorticosterone pivalate. Blood pressure and activity rhythms were analysed to evaluate changes over time. RESULTS: Blood pressure in WT mice was not affected by a high salt diet plus mineralocorticoid. In contrast, Per1 KO mice exhibited significantly increased mean arterial pressure (MAP) in response to a high salt diet plus mineralocorticoid. The inactive/active phase ratio of MAP in WT mice was unchanged by high salt plus mineralocorticoid treatment. Importantly, this treatment caused Per1 KO mice to lose the expected decrease or 'dip' in blood pressure during the inactive compared to the active phase. CONCLUSION: Loss of PER1 increased sensitivity to the high salt plus mineralocorticoid treatment. It also resulted in a non-dipper phenotype in this model of salt-sensitive hypertension and provides a unique model of non-dipping. Together, these data support an important role for the circadian clock protein PER1 in the modulation of blood pressure in a high salt/mineralocorticoid model of hypertension.


Subject(s)
Hypertension/metabolism , Hypertension/physiopathology , Period Circadian Proteins/metabolism , Animals , Blood Pressure/physiology , Desoxycorticosterone/analogs & derivatives , Desoxycorticosterone/pharmacology , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Hypertension/chemically induced , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mineralocorticoids/pharmacology , Period Circadian Proteins/deficiency , Real-Time Polymerase Chain Reaction , Sodium Chloride, Dietary/pharmacology
4.
Br J Pharmacol ; 168(2): 327-34, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22220553

ABSTRACT

Control of gene transcription is a major regulatory determinant for function of the endothelin pathway. Epigenetic mechanisms act on tissue-specific gene expression during development and in response to physiological stimuli. Most of the limited evidence available on epigenetic regulation of the endothelin pathway focuses on the EDN1 and EDNRB genes. Examination of whole genome databases suggests that both genes are influenced by histone modifications and DNA methylation. This interpretation is supported by studies directed at detecting epigenetic action on the two genes. The clearest illustration of epigenetic factors altering endothelin signalling is DNA methylation-associated EDNRB silencing during tumourigenesis. This review summarizes our current understanding of epigenetic regulation of the endothelin pathway genes. LINKED ARTICLES This article is part of a themed section on Endothelin. To view the other articles in this section visit http://dx.doi.org/10.1111/bph.2013.168.issue-1.


Subject(s)
Endothelins/genetics , Animals , DNA Methylation , Epigenesis, Genetic , Histones/metabolism , Humans
5.
J Neurochem ; 89(2): 307-13, 2004 Apr.
Article in English | MEDLINE | ID: mdl-15056274

ABSTRACT

Prohormone convertase (PC1) is found in endocrine cell lines that express cholecystokinin (CCK) mRNA and process pro CCK to biologically active products. Other studies have demonstrated that PC1 may be a one of the enzymes responsible for the endoproteolytic cleavages that occur in pro CCK during its biosynthesis and processing. Prohormone convertase 1 (PC1) has a distribution that is similar to cholecystokinin (CCK) in rat brain. A moderate to high percentage of CCK mRNA-positive neurons express PC1 mRNA. CCK levels were measured in PC1 knockout and control mice to assess the degree to which loss of PC1 changed CCK content. CCK levels were decreased 62% in hippocampus, 53% in amygdala and 57% in pons-medulla in PC1 knockout mice as compared to controls. These results are highly correlated with the colocalization of CCK and PC1. The majority of CCK mRNA-positive neurons in the pyramidal cell layer of the hippocampus express PC1 mRNA and greater than 50% of CCK mRNA-positive neurons in several nuclei of the amygdala also express PC1. These results demonstrate that PC1 is important for CCK processing. PC2 and PC5 are also widely colocalized with CCK. It may be that PC2, PC5 or another non-PC enzyme are able to substitute for PC1 and sustain production of some amidated CCK. Together these enzymes may represent a redundant system to insure the production of CCK.


Subject(s)
Brain/metabolism , Cholecystokinin/metabolism , Proprotein Convertases/genetics , RNA, Messenger/metabolism , Amygdala/metabolism , Animals , Cholecystokinin/genetics , Female , Hippocampus/metabolism , Male , Medulla Oblongata/metabolism , Mice , Mice, Knockout , Neurons/metabolism , Pons/metabolism , Proprotein Convertases/deficiency , Protein Processing, Post-Translational , Rats
6.
Acad Emerg Med ; 8(12): 1122-7, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11733288

ABSTRACT

OBJECTIVES: To measure the effect of tricyclic antidepressant drugs (TCAs) on human myocardial contractility. METHODS: Human atrial tissue was obtained during cardiac bypass surgery. The tissue was harvested, suspended in a Tyrode buffer at 37 degrees C, and perfused with a 95%/5% oxygen-carbon dioxide mixture. Developed force was continuously measured using a force transducer and recorded by computer. After an equilibration period, escalating doses of amitriptyline or desipramine were added to the bath. All strips were exposed to the following five concentrations of each drug: 0 (control) 0.4, 4, 40, and 400 microM. The results for each experiment were expressed as the difference between the developed force measured prior to the addition of each concentration of drug and the developed force measured after a 30-minute exposure to the drug. RESULTS: Desipramine decreased the developed force by 27%, 49%, and 74% at concentrations of 0.4, 40, and 400 microM, respectively. Amitriptyline decreased the developed force by 38% at the 40-microM concentration and by 89% at the 400-microM concentration. Untreated strips retained 94% of baseline developed force at 150 minutes. CONCLUSIONS: Tricyclic antidepressants depress human myocardial function in a dose-dependent fashion independent of the effects on the cardiac conduction system. While previous work has demonstrated the effect of therapies for the reversal of impaired cardiac conduction following TCA poisoning, to the best of the authors' knowledge, no reports have documented the effects of therapy on direct myocardial depression. Additional therapies targeted at reversing the direct cardiodepressive effects of TCA may improve outcome following TCA poisoning.


Subject(s)
Amitriptyline/pharmacology , Antidepressive Agents, Tricyclic/pharmacology , Desipramine/pharmacology , Heart Atria/drug effects , Myocardial Contraction/drug effects , Analysis of Variance , Culture Techniques , Dose-Response Relationship, Drug , Humans , Neural Conduction/drug effects , Neural Conduction/physiology , Probability , Reference Values , Sensitivity and Specificity
8.
Peptides ; 22(8): 1271-7, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11457520

ABSTRACT

Endocrine tumor cells in culture and in vitro cleavage assays have shown that PC1 and PC2 are capable of processing pro-CCK into smaller, intermediate and final, bioactive forms. Similar studies have shown that PC5 has the ability to process a number of propeptides. Here, we use GT1-7 (mouse hypothalamic) and SK-N-MC and SK-N-SH (human neuroblastoma) tumor cell lines to study the ability of PC5 to process pro-CCK. RT-PCR and Western blot analysis showed that the cells express PC5 mRNA and protein, but not PC1 or PC2. They were engineered to stably overexpress CCK and cell media was analyzed for pro-CCK expression and cleavage of the prohormone. Radioimmunoassays showed that pro-CCK was expressed, but no amidated CCK was detected. Lack of production of amidated CCK may be due to the lack of the appropriate carboxypeptidase and amidating enzymes. Production of glycine-extended CCK processing products was evaluated by treatment of media with carboxypeptidase B followed by analysis with a CCK Gly RIA. Glycine-extended forms of the peptide were found in the media. The predominant forms co-eluted with CCK 12 Gly and CCK 22 Gly on gel filtration chromatography. The results demonstrate that these cell lines which express PC5 and not PC1 or PC2 have the ability to process pro-CCK into intermediate, glycine-extended forms more closely resembling pro-CCK products in intestine than in brain.


Subject(s)
Aspartic Acid Endopeptidases/biosynthesis , Cholecystokinin/metabolism , Glycine/metabolism , Neurons/metabolism , Peptide Fragments/metabolism , Proprotein Convertase 1 , Protein Precursors/metabolism , Serine Endopeptidases/biosynthesis , Subtilisins/biosynthesis , Animals , Blotting, Western , Brain/metabolism , Cell Line , Chromatography, Gel , Genetic Vectors , Glycine/chemistry , Humans , Intestinal Mucosa/metabolism , Mice , Proprotein Convertase 2 , Proprotein Convertase 5 , Proprotein Convertases , RNA, Messenger/metabolism , Radioimmunoassay , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured
9.
Am J Physiol Renal Physiol ; 281(2): F357-65, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11457728

ABSTRACT

The rabbit kidney possesses mRNA for the H-K-ATPase alpha(1)-subunit (HKalpha(1)) and two splice variants of the H-K-ATPase alpha(2)-subunit (HKalpha(2)). The purpose of this study was to determine the specific distribution of one of these, the H-K-ATPase alpha(2c)-subunit isoform (HKalpha(2c)), in rabbit kidney by immunohistochemistry. Chicken polyclonal antibodies against a peptide based on the NH(2) terminus of HKalpha(2c) were used to detect HKalpha(2c) immunoreactivity in tissue sections. Immunohistochemical localization of HKalpha(2c) revealed intense apical immunoreactivity in a subpopulation of cells in the connecting segment, cortical collecting duct, and outer medullary collecting duct in both the outer and inner stripe. An additional population of cells exhibited a thin apical band of immunolabel. Immunohistochemical colocalization of HKalpha(2c) with carbonic anhydrase II, the Cl(-)/HCO exchanger AE1, and HKalpha(1) indicated that both type A and type B intercalated cells possessed intense apical HKalpha(2c) immunoreactivity, whereas principal cells and connecting segment cells had only a thin apical band of HKalpha(2c). Labeled cells were evident through the middle third of the inner medullary collecting duct in the majority of animals. Immunolabel was also present in papillary surface epithelial cells, cells in the cortical thick ascending limb of Henle's loop (cTAL), and the macula densa. Thus in the rabbit kidney, apical HKalpha(2c) is present and may contribute to acid secretion or potassium uptake throughout the connecting segment and collecting duct in both type A and type B intercalated cells, principal cells, and connecting segment cells, as well as in cells in papillary surface epithelium, cTAL, and macula densa.


Subject(s)
H(+)-K(+)-Exchanging ATPase/analysis , Kidney/enzymology , Animals , Antibodies, Monoclonal/immunology , Antiporters/analysis , Carbonic Anhydrases/analysis , Chloride-Bicarbonate Antiporters , Female , H(+)-K(+)-Exchanging ATPase/immunology , Immunoenzyme Techniques , Isoenzymes/analysis , Kidney/chemistry , Kidney/cytology , Kidney Cortex/chemistry , Kidney Cortex/enzymology , Kidney Medulla/chemistry , Kidney Medulla/enzymology , Kidney Tubules/chemistry , Kidney Tubules/enzymology , Protein Subunits , Rabbits
10.
Nature ; 409(6818): 327-31, 2001 Jan 18.
Article in English | MEDLINE | ID: mdl-11201738

ABSTRACT

Except in regions of recent crustal extension, the dominant origin of carbon dioxide in fluids in sedimentary basins has been assumed to be from crustal organic matter or mineral reactions. Here we show, by contrast, that Rayleigh fractionation caused by partial degassing of a magma body can explain the CO2/3He ratios and delta13C(CO2) values observed in CO2-rich natural gases in the west Texas Val Verde basin and also the mantle 3He/22Ne ratios observed in other basin systems. Regional changes in CO2/3He and CO2/CH4 ratios can be explained if the CO2 input pre-dates methane generation in the basin, which occurred about 280 Myr ago. Uplift to the north of the Val Verde basin between 310 and 280 Myr ago appears to be the only tectonic event with appropriate timing and location to be the source of the magmatic CO2. Our identification of magmatic CO2 in a foreland basin indicates that the origin of CO2 in other mid-continent basin systems should be re-evaluated. Also, the inferred closed-system preservation of natural gas in a trapping structure for approximately 300 Myr is far longer than the residence time predicted by diffusion models.

11.
Arch Biochem Biophys ; 380(1): 201-7, 2000 Aug 01.
Article in English | MEDLINE | ID: mdl-10900150

ABSTRACT

A large number of mutations affecting the F(0) sector of Escherichia coli F(1)F(0) ATP synthase have been constructed and characterized. A subset of the missense mutations resulted in fully assembled enzyme complexes blocked in proton translocation and displaying marked decreases in ATP hydrolysis activity. The catalytic activities of one such mutant enzyme, a(ala-217-->arg), have been determined using both multisite and unisite catalysis conditions. As expected, the V(max) of the a(ala-217-->arg) enzyme was reduced under conditions of saturating substrate concentration. However, the F(0) sector amino acid substitution did not affect nucleotide occupancy of the noncatalytic sites. Moreover, the microscopic rate constants measured using unisite methods yielded no significant differences between the intact wild type F(1)F(0) ATP synthase and the a(ala-217-->arg) mutant enzyme. In general, the values for unisite activities in both preparations were very similar to numbers reported in the literature for E. coli F(1)-ATPase. The results suggest that the a(ala-217-->arg) substitution resulted in a defect in catalytic cooperativity and most likely altered the enzyme by inhibiting the rotational mechanism of F(1)F(0) ATP synthase.


Subject(s)
Alanine/chemistry , Arginine/chemistry , Proton-Translocating ATPases/metabolism , Adenosine Triphosphate/pharmacology , Catalysis , Dose-Response Relationship, Drug , Escherichia coli/enzymology , Hydrolysis , Kinetics , Mutagenesis , Mutation, Missense , Protein Conformation , Proton-Translocating ATPases/chemistry , Proton-Translocating ATPases/genetics , Time Factors
13.
Biochem Biophys Res Commun ; 273(1): 188-91, 2000 Jun 24.
Article in English | MEDLINE | ID: mdl-10873584

ABSTRACT

Analysis of CCK content in extracts of whole forebrain from PC2 and 7B2 null mouse brain showed a significant decrease relative to wild-type brains. More detailed analysis revealed that CCK 8 amide levels in cerebral cortex and forebrain regions were more decreased than in hypothalamus. CCK 8 content in PC2 null mouse intestines was identical to control. Null mutant brains contained less CCK 8 than wild type and no other forms were seen when analyzed by gel filtration chromatography. No brain area examined was completely devoid of CCK, suggesting that other enzymes can partially compensate for the loss of PC2. This is the first demonstration that any endoprotease is important for CCK processing but also suggest the presence of a redundant system to ensure production of active CCK in the brain.


Subject(s)
Cholecystokinin/metabolism , Gene Deletion , Nerve Tissue Proteins/metabolism , Pituitary Hormones/metabolism , Protein Precursors/metabolism , Protein Processing, Post-Translational , Subtilisins/metabolism , Animals , Cerebral Cortex/enzymology , Cerebral Cortex/metabolism , Chromatography, Gel , Female , Hypothalamus/enzymology , Hypothalamus/metabolism , Intestinal Mucosa/metabolism , Intestines/enzymology , Male , Mice , Mice, Knockout , Nerve Tissue Proteins/deficiency , Nerve Tissue Proteins/genetics , Neuroendocrine Secretory Protein 7B2 , Peptide Fragments/metabolism , Pituitary Hormones/deficiency , Pituitary Hormones/genetics , Proprotein Convertase 2 , Prosencephalon/enzymology , Prosencephalon/metabolism , Radioimmunoassay , Subtilisins/deficiency , Subtilisins/genetics
15.
J Bioenerg Biomembr ; 32(4): 365-71, 2000 Aug.
Article in English | MEDLINE | ID: mdl-11768298

ABSTRACT

The a and b subunits constitute the stator elements in the F0 sector of F1F0-ATP synthase. Both subunits have been difficult to study by physical means, so most of the information on structure and function relationships in the a and b subunits has been obtained using mutagenesis in combination with biochemical methods. These approaches were used to demonstrate that the a subunit in association with the ring of c subunits houses the proton channel through F1F0-ATP synthase. The map of the amino acids contributing to the proton channel is probably complete. The two b subunits dimerize, forming an extended flexible unit in the peripheral stalk linking the F1 and F0 sectors. The unique characteristics of specific amino acid substitutions affecting the a and b subunits suggested differential effects on rotation during F1F0-ATPase activity.


Subject(s)
Mutagenesis , Proton-Translocating ATPases/chemistry , Bacteria/enzymology , Humans , Mutation , Protein Subunits , Proton-Translocating ATPases/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism
16.
J Biol Chem ; 274(51): 36261-6, 1999 Dec 17.
Article in English | MEDLINE | ID: mdl-10593914

ABSTRACT

In Escherichia coli F(1)F(0) ATP synthase, the two b subunits dimerize forming the peripheral second stalk linking the membrane F(0) sector to F(1). Previously, we have demonstrated that the enzyme could accommodate relatively large deletions in the b subunits while retaining function (Sorgen, P. L., Caviston, T. L., Perry, R. C., and Cain, B. D. (1998) J. Biol. Chem. 273, 27873-27878). The manipulations of b subunit length have been extended by construction of insertion mutations into the uncF(b) gene adding amino acids to the second stalk. Mutants with insertions of seven amino acids were essentially identical to wild type strains, and mutants with insertions of up to 14 amino acids retained biologically significant levels of activity. Membranes prepared from these strains had readily detectable levels of F(1)F(0)-ATPase activity and proton pumping activity. However, the larger insertions resulted in decreasing levels of activity, and immunoblot analysis indicated that these reductions in activity correlated with reduced levels of b subunit in the membranes. Addition of 18 amino acids was sufficient to result in the loss of F(1)F(0) ATP synthase function. Assuming the predicted alpha-helical structure for this area of the b subunit, the 14-amino acid insertion would result in the addition of enough material to lengthen the b subunit by as much as 20 A. The results of both insertion and deletion experiments support a model in which the second stalk is a flexible feature of the enzyme rather than a rigid rod-like structure.


Subject(s)
Bacterial Proteins/metabolism , Escherichia coli/enzymology , Proton-Translocating ATPases/metabolism , Amino Acid Sequence , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Base Sequence , Molecular Sequence Data , Mutagenesis, Insertional , Mutation , Proton-Translocating ATPases/chemistry , Proton-Translocating ATPases/genetics
17.
Semin Nephrol ; 19(5): 431-7, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10511383

ABSTRACT

The pharmacological properties of H+,K+-ATPase activity described in the kidney were not necessarily consistent with the properties of the well-characterized gastric H+,K+-ATPase. Recent molecular biology experiments suggest that renal H+,K+-ATPase activity may be the product of several closely related P-type ATPases. At least 3 different pumps containing the HKalpha1, HKalpha2a, and HKalpha2c subunits have been detected in rabbit kidney. The current view is that these HKalpha subunits arose through gene duplication early in evolution and the proteins evolved their differing activities over time. The HKbeta protein associates with HKalpha1 in gastric tissues and is the likely mate for the HKalpha1 subunit in renal tissues. Three distinct beta subunits have been implicated as possible partners for the HKalpha2 subunits, but it remains to be determined which beta subunit predominantly associates with the HKalpha2 subunits in vivo. Sequence analysis suggests the beta subunit was constrained by size and shape of the protein rather than specific amino acid content during the course of evolution. Multiple H+,K+-ATPases in the kidney may be an important adaptation providing redundancy for the essential physiological function of maintaining ionic balance.


Subject(s)
H(+)-K(+)-Exchanging ATPase/genetics , Kidney Tubules, Collecting/enzymology , Amino Acid Sequence , Animals , DNA, Complementary/analysis , Dogs , H(+)-K(+)-Exchanging ATPase/metabolism , Humans , Ion Transport/physiology , Kidney Tubules, Collecting/physiology , Mice , Molecular Biology , Molecular Sequence Data , Polymerase Chain Reaction , Rabbits , Rats , Species Specificity , Water-Electrolyte Balance
18.
J Surg Res ; 86(2): 171-6, 1999 Oct.
Article in English | MEDLINE | ID: mdl-10534420

ABSTRACT

We have previously reported that atrial trabeculae from patients taking oral sulfonylurea hypoglycemic agents cannot be preconditioned by transient ischemia, which may, in part, explain the increased cardiovascular mortality historically associated with the use of these agents (J. C. Cleveland et al., 1997, Circulation 96, 29-32). Recently, we reported that clinically accessible and acceptable exogenous Ca(2+) pretreatment protects human atrial trabeculae from subsequent ischemia (B. S. Cain et al., 1998, Ann. Thoracic Surg. 65, 1065-1070). It remains unknown whether this preconditioning strategy could confer protection to trabeculae from patients taking oral sulfonylurea drugs. We therefore hypothesized that exogenous Ca(2+) confers ischemic protection to trabeculae from patients taking oral sulfonylureas. Human atrial trabeculae were suspended in organ baths and field stimulated at 1 Hz, and force development was recorded. Following 90 min equilibration, trabeculae from patients taking oral sulfonylurea agents (n = 6 patients) were subjected to ischemia/reperfusion (I/R; 45/120 min) with or without Ca(2+) (1 mM increase x 5 min) 10 min prior to I/R. I/R decreased postischemic human myocardial contractility in trabeculae from patients on oral hypoglycemics to 15.3 +/- 2.0% baseline developed force (%BDF). Ca(2+) pretreatment increased postischemic human myocardial developed force to 35.3 +/- 2.9 %BDF in these patients (P < 0.05 vs I/R, ANOVA and Bonferroni/Dunn). We conclude that atrial muscle from patients taking oral hypoglycemic agents can be preconditioned with exogenous Ca(2+). This therapy may offer a clinically relevant means to precondition the myocardium of diabetics taking oral hypoglycemic agents prior to clinical interventions such as coronary angioplasty or cardiac bypass.


Subject(s)
Atrial Function/drug effects , Calcium/pharmacology , Hypoglycemic Agents/therapeutic use , Ischemic Preconditioning, Myocardial/methods , Sulfonylurea Compounds/therapeutic use , Administration, Oral , Humans , In Vitro Techniques , Myocardial Contraction/drug effects , Myocardial Ischemia/physiopathology , Myocardial Reperfusion Injury/physiopathology
19.
Int J Biochem Cell Biol ; 31(7): 769-76, 1999 Jul.
Article in English | MEDLINE | ID: mdl-10467733

ABSTRACT

The mutations in human mitochondrial DNA at nt8993 are associated with a range of neuromuscular disorders. One mutation encodes a proline in place of a leucine conserved in all animal mitochondrial ATPase-6 subunits and bacterial a subunits of F1F0 ATP synthases. This conserved site is leu-156 and leu-207 in humans and Escherichia coli, respectively. An aleu-207-->pro substitution mutation has been constructed in the E. coli F1F0 ATP synthase in order to model the biochemical basis of the human disease mutation. The phenotype of the aleu-207-->pro substitution has been compared to that of the previously studied aleu-207-->arg substitution (Hartzog and Cain, 1993, Journal of Biological Chemistry 268, 12250-12252). The leu-207-->pro mutation resulted in approximately a 35% decrease in the number of intact enzyme complexes as determined by N, N'-dicyclohexylcarbodiimide-sensitive membrane associated ATP hydrolysis activity and western analysis using an anti-a subunit antibody. A 75% reduction in the efficiency of proton translocation through F1F0 ATP synthase was observed in ATP-driven proton pumping assays. Interestingly, the loss in F1F0 ATP synthase activity resulting from the leu-207-->pro substitution was markedly less dramatic than had been observed for the leu-207-->arg mutation studied earlier. By analogy, the human enzyme may also be affected by the leu-156-->pro substitution to a lesser extent than the leu-156-->arg substitution, and this would account for the milder clinical manifestations of the human leu-156-->pro disease mutations.


Subject(s)
Escherichia coli/enzymology , Leigh Disease/enzymology , Leigh Disease/genetics , Point Mutation , Proton-Translocating ATPases/genetics , Base Sequence , DNA, Mitochondrial/genetics , Escherichia coli/genetics , Genetic Complementation Test , Humans , Plasmids/genetics , Proton-Translocating ATPases/chemistry , Proton-Translocating ATPases/metabolism , Syndrome
20.
Crit Care Med ; 27(7): 1309-18, 1999 Jul.
Article in English | MEDLINE | ID: mdl-10446825

ABSTRACT

OBJECTIVE: Proinflammatory cytokines such as tumor necrosis factor (TNF)-alpha and interleukin (IL)-1beta have been implicated in the pathogenesis of myocardial dysfunction in ischemia-reperfusion injury, sepsis, chronic heart failure, viral myocarditis, and cardiac allograft rejection. Although circulating TNF-alpha and IL-1beta are both often elevated in septic shock, it remains unknown whether TNF-alpha or IL-1beta are the factors induced during sepsis that directly depress human myocardial function, and if so, whether the combination synergistically depresses myocardial function. Furthermore, the mechanism(s) by which these cytokines induce human myocardial depression remain unknown. We hypothesized the following: a) TNF-alpha and IL-1beta directly depress human myocardial function; b) together, TNF-alpha and IL-1beta act synergistically to depress human myocardial function; and c) inhibition of ceramidase or nitric oxide synthase attenuates myocardial depression induced by TNF-alpha or IL-1beta by limiting proximal cytokine signaling or production of myocardial nitric oxide (NO). DESIGN: Prospective, randomized, controlled study. SETTING: Experimental laboratory in a university hospital. SUBJECTS: Freshly obtained human myocardial trabeculae. INTERVENTIONS: Human atrial trabeculae were obtained at the time of cardiac surgery, suspended in organ baths, and field simulated at 1 Hz, and the developed force was recorded. After a 90-min equilibration, TNF-alpha (1.25, 12.5, 125, or 250 pg/mL for 20 mins), IL-1beta (6.25, 12.5, 50, or 200 pg/mL for 20 mins), or TNF-alpha (1.25 pg/mL) plus IL-1beta (6.25 pg/mL) were added to the bath, and function was measured for the subsequent 100 mins after the 20-min exposure. To assess the roles of the sphingomyelin and NO pathways in TNF-alpha and IL-1beta cross-signaling, the ceramidase inhibitor N-oleoyl ethanolamine (1 microM) or the NO synthase inhibitor N(G)-monomethyl-L-arginine (10 microM) was added before TNF-alpha (125 pg/mL) or IL-1beta (50 pg/mL). MEASUREMENTS AND MAIN RESULTS: TNF-alpha and IL-1beta each depressed human myocardial function in a dose-dependent fashion (maximally depressing to 16.2 + 1.9% baseline developed force for TNF-alpha and 25.7 + 6.3% baseline developed force for IL-1beta), affecting systolic relatively more than diastolic performance (each p < .05). However, when combined, TNF-alpha and IL-1beta at concentrations that did not individually result in depression (p > .05 vs. control) resulted in contractile depression (p < .05 vs. control). Inhibition of myocardial sphingosine or NO release abolished the myocardial depressive effects of either TNF-alpha or IL-1beta. CONCLUSIONS: TNF-alpha and IL-1beta separately and synergistically depress human myocardial function. Sphingosine likely participates in the TNF-alpha and IL-1beta signal leading to human myocardial functional depression. Therapeutic strategies to reduce production or signaling of either TNF-alpha or IL-1beta may limit myocardial dysfunction in sepsis.


Subject(s)
Cardiomyopathies/metabolism , Cardiomyopathies/pathology , Interleukin-1/physiology , Sepsis/physiopathology , Tumor Necrosis Factor-alpha/physiology , Amidohydrolases/antagonists & inhibitors , Ceramidases , Coronary Disease/physiopathology , Heart Atria/pathology , Humans , Interleukin-1/pharmacokinetics , Nitric Oxide Synthase/antagonists & inhibitors , Prospective Studies , Sphingosine/metabolism , Tumor Necrosis Factor-alpha/pharmacokinetics
SELECTION OF CITATIONS
SEARCH DETAIL
...