Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
1.
Drug Metab Dispos ; 47(4): 419-423, 2019 04.
Article in English | MEDLINE | ID: mdl-30733251

ABSTRACT

It is well recognized that nonspecific binding of a drug within an in vitro assay (f u) can have a large impact on in vitro to in vivo correlations of intrinsic clearance. Typically, this value is determined experimentally across multiple species in the drug-discovery stage. Herein we examine the feasibility of using a single species (rat) as a surrogate for other species using a panel of small molecules representing highly diverse structures and physiochemical classes. The study demonstrated that 86% and 92% of the tested compounds measured in the mouse, dog, monkey, and human were within 2-fold of rat values for f u in microsomes and hepatocytes, respectively. One compound, amiodarone, exhibited unique species-dependent binding where the f u was approximately 10-fold higher in human microsomes and 20-fold higher in human hepatocytes compared with the average of the other species tested. Overall, these data indicate that using a single species (rat) f u as a surrogate for other major species, including humans, is a means to increase the throughput of measuring nonspecific binding in vitro.


Subject(s)
Hepatocytes/metabolism , Microsomes, Liver/metabolism , Animals , Dogs , Drug Discovery/methods , Female , Haplorhini , Humans , Male , Metabolic Clearance Rate/physiology , Mice , Rats , Rats, Sprague-Dawley , Species Specificity
2.
Drug Metab Dispos ; 47(10): 1174-1182, 2019 10.
Article in English | MEDLINE | ID: mdl-31097425

ABSTRACT

Understanding small interfering RNA (siRNA) fraction unbound (f u) in relevant physiologic compartments is critical for establishing pharmacokinetic-pharmacodynamic relationships for this emerging modality. In our attempts to isolate the equilibrium free fraction of N-acetylgalactosamine-conjugated siRNA using classic small-molecule in vitro techniques, we found that the hydrodynamic radius was critical in determining the size exclusion limit requirements for f u isolation, largely validating the siRNA "rigid rod" hypothesis. With this knowledge, we developed an orthogonally validated 50 kDa molecular-mass cutoff ultrafiltration assay to quantify f u in biologic matrices including human, nonhuman primate, rat, and mouse plasma, and human liver homogenate. To enhance understanding of the siRNA-plasma interaction landscape, we examined the effects of various common oligonucleotide therapeutic modifications to the ribose and helix backbone on siRNA f u in plasma (f u,plasma) and found that chemical modifications can alter plasma protein binding by at least 20%. Finally, to gain insight into which specific plasma proteins bind to siRNA, we developed a qualitative screen to identify binding "hits" across a panel of select purified human plasma proteins.


Subject(s)
Acetylgalactosamine/pharmacokinetics , Blood Proteins/metabolism , Liver/metabolism , RNA, Small Interfering/pharmacokinetics , Acetylgalactosamine/chemistry , Adult , Animals , Female , Humans , Macaca fascicularis , Mice , Protein Binding , RNA, Small Interfering/chemistry , Rats
3.
Am J Respir Cell Mol Biol ; 50(6): 1010-20, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24328827

ABSTRACT

Pulmonary arterial smooth muscle cell (PASMC) proliferation and migration are important contributors to the vascular remodeling that occurs during development of pulmonary hypertension. We previously demonstrated that aquaporin (AQP)1, a member of the water channel family of proteins, was expressed in PASMCs and was necessary for hypoxia-induced migration; however, the mechanism by which AQP1 controls this response is unclear. The C-terminal tail of AQP1 contains putative calcium (EF-hand) and protein binding sites. Thus, we wanted to explore whether the C-terminal tail or the EF-hand motif of AQP1 was required for migration and proliferation. Rat PASMCs were isolated from distal pulmonary arteries, and proliferation and migration were measured using BrdU incorporation and transwell filters, respectively. To deplete AQP1, PASMCs were transfected with AQP1 small interference RNA (siRNA) or nontargeting siRNA. Knockdown of AQP1 reduced basal proliferation and hypoxia-induced migration and proliferation in PASMCs. In subsequent experiments, wild-type AQP1, AQP1 lacking the entire cytoplasmic C-terminal tail, or AQP1 with a mutation in the EF-hand motif were expressed in PASMCs using adenoviral constructs. For all AQP1 constructs, infection increased AQP1 protein levels, water permeability, and the change in cell volume induced by hypotonic challenge. Infection with wild-type and EF-hand mutated AQP1, but not C-terminal-deleted AQP1, increased PASMC migration and proliferation. Our results suggest that AQP1 controls proliferation and migration in PASMCs and that the mechanism requires the C-terminal tail of the protein but is independent of water transport or the EF-hand motif.


Subject(s)
Aquaporin 1/metabolism , Cell Movement/physiology , Muscle Cells/physiology , Pulmonary Artery/physiology , Animals , Aquaporin 1/genetics , Calcium/metabolism , Cell Growth Processes/genetics , Cell Growth Processes/physiology , Cell Hypoxia/physiology , Cell Movement/genetics , Fluoresceins/chemistry , Gene Knockdown Techniques , Male , Muscle Cells/metabolism , Mutation , Pulmonary Artery/metabolism , Rats , Rats, Wistar , Water/metabolism
4.
Hepatol Commun ; 8(5)2024 May 01.
Article in English | MEDLINE | ID: mdl-38696369

ABSTRACT

BACKGROUND: Human genetic studies have identified several mitochondrial amidoxime-reducing component 1 (MTARC1) variants as protective against metabolic dysfunction-associated steatotic liver disease. The MTARC1 variants are associated with decreased plasma lipids and liver enzymes and reduced liver-related mortality. However, the role of mARC1 in fatty liver disease is still unclear. METHODS: Given that mARC1 is mainly expressed in hepatocytes, we developed an N-acetylgalactosamine-conjugated mouse Mtarc1 siRNA, applying it in multiple in vivo models to investigate the role of mARC1 using multiomic techniques. RESULTS: In ob/ob mice, knockdown of Mtarc1 in mouse hepatocytes resulted in decreased serum liver enzymes, LDL-cholesterol, and liver triglycerides. Reduction of mARC1 also reduced liver weight, improved lipid profiles, and attenuated liver pathological changes in 2 diet-induced metabolic dysfunction-associated steatohepatitis mouse models. A comprehensive analysis of mARC1-deficient liver from a metabolic dysfunction-associated steatohepatitis mouse model by metabolomics, proteomics, and lipidomics showed that Mtarc1 knockdown partially restored metabolites and lipids altered by diet. CONCLUSIONS: Taken together, reducing mARC1 expression in hepatocytes protects against metabolic dysfunction-associated steatohepatitis in multiple murine models, suggesting a potential therapeutic approach for this chronic liver disease.


Subject(s)
Disease Models, Animal , Gene Knockdown Techniques , Hepatocytes , Animals , Mice , Hepatocytes/metabolism , Liver/metabolism , Male , RNA, Small Interfering/genetics , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Non-alcoholic Fatty Liver Disease/genetics , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/prevention & control , Mice, Inbred C57BL
5.
Antimicrob Agents Chemother ; 57(10): 5067-79, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23917319

ABSTRACT

Rilpivirine is a nonnucleoside reverse transcriptase inhibitor used to treat HIV-1. In the present study, the pathways responsible for the biotransformation of rilpivirine were defined. Using human liver microsomes, the formation of two mono- and two dioxygenated metabolites were detected via ultra high-performance liquid chromatography-tandem mass spectrometry (UHPLC-MS/MS). Mass spectral analysis of the products suggested that these metabolites resulted from oxygenation of the 2,6-dimethylphenyl ring and methyl groups of rilpivirine. Chemical inhibition studies and cDNA-expressed cytochrome P450 (CYP) assays indicated that oxygenations were catalyzed primarily by CYP3A4 and CYP3A5. Glucuronide conjugates of rilpivirine and a monomethylhydroxylated metabolite of rilpivirine were also detected and were found to be formed by UDP-glucuronosyltransferases (UGTs) UGT1A4 and UGT1A1, respectively. All metabolites that were identified in vitro were detectable in vivo. Further, targeted UHPLC-MS/MS-based in vivo metabolomics screening revealed that rilpivirine treatment versus efavirenz treatment may result in differential levels of endogenous metabolites, including tyrosine, homocysteine, and adenosine. Rilpivirine biotransformation was also assessed across species using liver microsomes isolated from a range of mammals, and the metabolite profile identified using human liver microsomes was largely conserved for both oxidative and glucuronide metabolite formation. These studies provide novel insight into the metabolism of rilpivirine and the potential differential effects of rilpivirine- and efavirenz-containing antiretroviral regimens on the endogenous metabolome.


Subject(s)
Nitriles/metabolism , Pyrimidines/metabolism , Animals , Chromatography, Liquid , Cricetinae , Dogs , Guinea Pigs , Humans , Mice , Microsomes, Liver/metabolism , Rabbits , Rats , Rilpivirine , Tandem Mass Spectrometry
6.
Front Physiol ; 14: 1108304, 2023.
Article in English | MEDLINE | ID: mdl-36926194

ABSTRACT

Exposure to hypoxia, due to high altitude or chronic lung disease, leads to structural changes in the pulmonary vascular wall, including hyperplasia and migration of pulmonary arterial smooth muscle cells (PASMCs). Previous studies showed that hypoxia upregulates the expression of Na+/H+ exchanger isoform 1 (NHE1) and that inhibition or loss of NHE1 prevents hypoxia-induced PASMC migration and proliferation. The exact mechanism by which NHE1 controls PASMC function has not been fully delineated. In fibroblasts, NHE1 has been shown to act as a membrane anchor for actin filaments, via binding of the adaptor protein, ezrin. Thus, in this study, we tested the role of ezrin and NHE1/actin interactions in controlling PASMC function. Using rat PASMCs exposed to in vitro hypoxia (4% O2, 24 h) we found that hypoxic exposure increased phosphorylation (activation) of ezrin, and promoted interactions between NHE1, phosphorylated ezrin and smooth muscle specific α-actin (SMA) as measured via immunoprecipitation and co-localization. Overexpression of wild-type human NHE1 in the absence of hypoxia was sufficient to induce PASMC migration and proliferation, whereas inhibiting ezrin phosphorylation with NSC668394 suppressed NHE1/SMA co-localization and migration in hypoxic PASMCs. Finally, overexpressing a version of human NHE1 in which amino acids were mutated to prevent NHE1/ezrin/SMA interactions was unable to increase PASMC migration and proliferation despite exhibiting normal Na+/H+ exchange activity. From these results, we conclude that hypoxic exposure increases ezrin phosphorylation in PASMCs, leading to enhanced ezrin/NHE1/SMA interaction. We further speculate that these interactions promote anchoring of the actin cytoskeleton to the membrane to facilitate the changes in cell movement and shape required for migration and proliferation.

7.
J Med Chem ; 65(13): 8713-8734, 2022 07 14.
Article in English | MEDLINE | ID: mdl-35790118

ABSTRACT

The "free drug hypothesis" assumes that, in the absence of transporters, the steady state free plasma concentrations equal to that at the site of action that elicit pharmacologic effects. While it is important to utilize the free drug hypothesis, exceptions exist that the free plasma exposures, either at Cmax, Ctrough, and Caverage, or at other time points, cannot represent the corresponding free tissue concentrations. This "drug concentration asymmetry" in both total and free form can influence drug disposition and pharmacological effects. In this review, we first discuss options to assess total and free drug concentrations in tissues. Then various drug design strategies to achieve concentration asymmetry are presented. Last, the utilities of tissue concentrations in understanding exposure-effect relationships and translational projections to humans are discussed for several therapeutic areas and modalities. A thorough understanding in plasma and tissue exposures correlation with pharmacologic effects can provide insightful guidance to aid drug discovery.


Subject(s)
Drug Discovery , Plasma , Humans , Membrane Transport Proteins
8.
Biochem Pharmacol ; 189: 114374, 2021 07.
Article in English | MEDLINE | ID: mdl-33358826

ABSTRACT

We report here the evaluation of a novel in vitro experimental model, prolonged cultured human hepatocytes (PCHC), as an experimental system to evaluate the potency and duration of effects of oligonucleotide therapeutics. A novel observation was made on the redifferentiation of PCHC upon prolonged culturing based on mRNA profiling of characteristic hepatic differentiation marker genes albumin, transferrin, and transthyretin. Consistent with the known de-differentiation of cultured human hepatocytes, decreases in marker gene expression were observed upon culturing of the hepatocytes for 2 days. A novel observation of re-differentiation was observed on day 7 as demonstrated by an increase in expression of the marker genes to levels similar to that observed on the first day of culture. The expression of the differentiation marker genes was highest on day 7, followed by a gradual decrease but remained higher than that on day 2 for up to the longest culture duration evaluated of 41 days. The redifferentiation phenomenon suggests that PCHC may be useful for the evaluation of the duration of effects of oligonucleotide therapeutics on gene expression in human hepatocytes. A proof of concept study was thereby conducted with PCHC with a GalNAc-conjugated siRNA targeting human hypoxanthine phosphoribosyl transferase1 (HPRT1). HPRT1 mRNA expression in siRNA-treated cultures decreased to 21% of that in untreated hepatocytes on day 1, <10% from days 2 to 12, <20% from days 16 to 33, and eventually recovered to 64% by day 41. Our results suggest that PCHC represent a clinically-relevant cost- and time-efficient experimental tool to aid in the evaluation of GalNAc-siRNA silencing activity, providing information on both efficacy and duration of efficacy. PCHC may be applicable in the drug development setting as a species- and cell type-relevant experimental tool to aid the development of oligonucleotide therapeutics.


Subject(s)
Acetylgalactosamine/biosynthesis , Cell Culture Techniques/methods , Gene Silencing/physiology , Hepatocytes/metabolism , Hypoxanthine Phosphoribosyltransferase/biosynthesis , RNA, Small Interfering/administration & dosage , Acetylgalactosamine/antagonists & inhibitors , Acetylgalactosamine/genetics , Cells, Cultured , Drug Evaluation, Preclinical/methods , Gene Silencing/drug effects , Hepatocytes/drug effects , Humans , Hypoxanthine Phosphoribosyltransferase/antagonists & inhibitors , Hypoxanthine Phosphoribosyltransferase/genetics , RNA, Small Interfering/genetics
9.
AIDS Res Hum Retroviruses ; 37(3): 173-183, 2021 03.
Article in English | MEDLINE | ID: mdl-33191765

ABSTRACT

A long-acting injectable formulation of rilpivirine (RPV), a non-nucleoside reverse transcriptase inhibitor, is currently under investigation for use in human immunodeficiency virus (HIV) maintenance therapy. We previously characterized RPV metabolism after oral dosing and identified seven metabolites: four metabolites resulting from mono- or dioxygenation of the 2,6-dimethylphenyl ring itself or either of the two methyl groups located on that ring, one N-linked RPV glucuronide conjugate, and two O-linked RPV glucuronides produced via glucuronidation of mono- and dihydroxymethyl metabolites. However, as is true for most drugs, the metabolism of RPV after injection has yet to be reported. The phase II clinical trial HPTN 076 enrolled 136 HIV-uninfected women and investigated the safety and acceptability of long-acting injectable RPV for use in HIV pre-exposure prophylaxis. Through the analysis of plasma samples from 80 of these participants in the active product arm of the study, we were able to detect 2 metabolites after intramuscular injection of long-acting RPV, 2-hydroxymethyl-RPV, and RPV N-glucuronide. Of the total of 80 individuals, 72 participants exhibited detectable levels of 2-hydroxymethyl-RPV in plasma samples whereas RPV N-glucuronide was detectable in plasma samples of 78 participants. In addition, RPV N-glucuronide was detectable in rectal fluid, cervicovaginal fluid, and vaginal tissue. To investigate potential genetic variation in genes encoding enzymes relevant to RPV metabolism, we isolated genomic DNA and performed next-generation sequencing of CYP3A4, CYP3A5, UGT1A1 and UGT1A4. From these analyses, four missense variants were detected for CYP3A4 whereas one missense variant and one frameshift variant were detected for CYP3A5. A total of eight missense variants of UGT1A4 were detected, whereas two variants were detected for UGT1A1; however, these variants did not appear to account for the observed interindividual variability in metabolite levels. These findings provide insight into the metabolism of long-acting RPV and contribute to an overall understanding of metabolism after oral dosing versus injection. ClinicalTrials.gov Identifier: NCT02165202.


Subject(s)
Anti-HIV Agents , HIV Infections , Anti-HIV Agents/therapeutic use , Female , HIV Infections/drug therapy , HIV Infections/prevention & control , Humans , Injections, Intramuscular , Reverse Transcriptase Inhibitors/therapeutic use , Rilpivirine/therapeutic use
10.
J Med Chem ; 63(12): 6407-6422, 2020 06 25.
Article in English | MEDLINE | ID: mdl-32352779

ABSTRACT

After two decades teetering at the intersection of laboratory tool and therapeutic reality, with two siRNA drugs now clinically approved, this modality has finally come into fruition. Consistent with other emerging modalities, initial proof-of-concept efforts concentrated on coupling pharmacologic efficacy with desirable safety profiles. Consequently, thorough investigations of siRNA absorption, distribution, metabolism, and excretion (ADME) properties are lacking. Advancing ADME knowledge will aid establishment of in vitro-in vivo correlations and pharmacokinetic-pharmacodynamic relationships to optimize candidate selection through discovery and translation. Here, we outline the emerging siRNA design principles and discuss the consequences for siRNA disposition and biotransformation. We propose a conceptual framework for siRNA ADME evaluation, contextualizing the site of biotransformation product formation with PK-PD modulation, and end with a discussion around safety and regulatory considerations and future directions for this modality.


Subject(s)
Biotransformation , Drug Design , Drug Development , Drug Evaluation, Preclinical , Pharmaceutical Preparations/chemistry , RNA, Small Interfering/chemistry , Animals , Humans , Pharmaceutical Preparations/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/pharmacokinetics , Tissue Distribution
11.
Sci Rep ; 10(1): 10425, 2020 06 26.
Article in English | MEDLINE | ID: mdl-32591626

ABSTRACT

Oligonucleotide therapeutics use short interfering RNA (siRNA) or antisense oligonucleotide (ASO) molecules to exploit endogenous systems-neutralizing target RNA to prevent subsequent protein translation. While the potential clinical application is vast, delivery efficiency and extrahepatic targeting is challenging. Bioanalytical assays are important in building understanding of these complex relationships. The literature currently lacks description of robust and sensitive methods to measure siRNA and ASOs in complex biological matrices. Described herein is a non-enzymatic hybridization-based immunoassay that enables quantification of individual siRNA strands (antisense or sense) in serum, urine, bile, and liver and kidney homogenates. Assay utility is also demonstrated in ASOs. The assay improves upon previous works by abolishing enzymatic steps and further incorporating Locked Nucleic Acid (LNA) nucleotide modifications to increase analyte hybridization affinity and improve sensitivity, specificity, and robustness. We report an assay with an ultrasensitive dynamic range of 0.3 to 16,700 pM for siRNA in serum. The assay was submitted to full qualification for accuracy and precision in both serum and tissue matrices and assay performance was assessed with single and mixed analytes. The reliable LNA-hybridization-based approach removes the need for matrix sample extraction, enrichment or amplification steps which may be impeded by more advanced chemical modifications.


Subject(s)
Immunoassay , Nucleic Acids/analysis , RNA, Small Interfering/metabolism , Animals , Humans , Mice , Oligonucleotides , Rats , Rats, Sprague-Dawley
12.
Mol Ther Nucleic Acids ; 21: 725-736, 2020 Sep 04.
Article in English | MEDLINE | ID: mdl-32771924

ABSTRACT

There has been a renewed interest in therapeutic small interfering RNAs (siRNAs) over the past few years. This is particularly the result of successful and efficient delivery of N-acetylgalactosamine (GalNAc)-conjugated siRNAs to the liver. In general, the lead selection process for siRNA drugs is faster and more straightforward than traditional small molecules. Nevertheless, many siRNAs of different sequences and chemical modification patterns must still be evaluated before arriving at a final candidate. One of the major difficulties in streamlining this workflow is the well-known phenomenon that the in vitro data obtained from oligonucleotides transfected into cells are not directly predictive of their in vivo activity. Consequently, all oligonucleotides with some degree of in vitro activity are typically screened in vivo before final lead selection. Here, we demonstrate that the stability of liver-targeting GalNAc-conjugated siRNAs in a mouse liver homogenate shows an acceptable correlation to their in vivo target knockdown efficacy. Therefore, we suggest the incorporation of an in vitro liver homogenate stability assay during the lead optimization process for siRNAs. The addition of this assay to a flow scheme may decrease the need for animal studies, and it could bring cost savings and increase efficiency in siRNA drug development.

13.
Sci Rep ; 9(1): 3566, 2019 03 05.
Article in English | MEDLINE | ID: mdl-30837588

ABSTRACT

Despite the exquisite potential of siRNA as a therapeutic, the mechanism(s) responsible for the robust indirect exposure-response relationships have not been fully elucidated. To understand the siRNA properties linked to potent activity, requires the disposition of siRNA to be characterized. A technical challenge in the characterization is the detection and quantitation of siRNA from biological samples. Described herein, a Locked Nucleic Acid (LNA) Hybridization-Ligation ECL ELISA was designed for ultra-sensitive quantification of both sense and antisense strands of siRNA independent of structural modifica-tions. This assay was applied to measure siRNA in serum and tissue homogenate in preclinical species. We observed rapid clearance of siRNA from the systemic circulation which contrasted the prolonged accumulation within the tissue. The assay was also able to distinguish and quantify free siRNA from RNA-induced silencing complex (RISC) and Argonaute 2 (Ago2) associated with therapeutic siRNA. We utilized an orthogonal method, LC-MS, to investigate 3' exonuclease activity toward the antisense strand metabolism. Taken together, we have demonstrated that the LNA Hybridization-Ligation ECL ELISA is arobust analytical method with direct application to measuring the exposure of siRNA therapeutics seamlessly across biological matrices.


Subject(s)
Enzyme-Linked Immunosorbent Assay/methods , Oligonucleotides/genetics , RNA, Small Interfering/analysis , Animals , Intracellular Space/metabolism , Nucleic Acid Hybridization , Oligonucleotides, Antisense/genetics , RNA, Small Interfering/genetics
14.
ChemMedChem ; 13(7): 736-747, 2018 04 06.
Article in English | MEDLINE | ID: mdl-29430850

ABSTRACT

Efavirenz (EFV), an antiretroviral that interacts clinically with co-administered drugs via activation of the pregnane X receptor (PXR), is extensively metabolized by the cytochromes P450. We tested whether its primary metabolite, 8-hydroxyEFV (8-OHEFV) can activate PXR and potentially contribute to PXR-mediated drug-drug interactions attributed to EFV. Luciferase reporter assays revealed that despite only differing from EFV by an oxygen atom, 8-OHEFV does not activate PXR. Corroborating this, treatment with EFV for 72 h elevated the mRNA abundance of the PXR target gene, Cyp3a11, by approximately 28-fold in primary hepatocytes isolated from PXR-humanized mice, whereas treatment with 8-OHEFV did not result in a change in Cyp3A11 mRNA levels. FRET-based competitive binding assays and isothermal calorimetry demonstrated that even with the lack of ability to activate PXR, 8-OHEFV displays an affinity for PXR (IC50 12.1 µm; KD 7.9 µm) nearly identical to that of EFV (IC50 18.7 µm; KD 12.5 µm). The use of 16 EFV analogues suggest that other discreet changes to the EFV structure beyond the 8-position are well tolerated. Molecular docking simulations implicate an 8-OHEFV binding mode that may underlie its divergence in PXR activation from EFV.


Subject(s)
Benzoxazines/metabolism , Pregnane X Receptor/metabolism , Alkynes , Animals , Benzoxazines/chemistry , Benzoxazines/pharmacology , Binding Sites , Cyclopropanes , Hepatocytes/drug effects , Humans , Ligands , Mice , Molecular Docking Simulation , Molecular Structure , Pregnane X Receptor/agonists , Pregnane X Receptor/chemistry , Protein Binding , Structure-Activity Relationship
15.
Physiol Rep ; 6(9): e13698, 2018 05.
Article in English | MEDLINE | ID: mdl-29756391

ABSTRACT

Excessive production of endothelin-1 (ET-1) has been observed in almost all forms of pulmonary hypertension. ET-1, a highly potent vasoconstrictor, can also potentiate pulmonary arterial smooth muscle cell (PASMC) growth and migration, both of which contribute to the vascular remodeling that occurs during the development of pulmonary hypertension. Increasing evidence indicates that alkalinization of intracellular pH (pHi ), typically due to activation of Na+ /H+ exchange (NHE), is associated with enhanced PASMC proliferation and migration. We recently demonstrated that application of exogenous ET-1 increased NHE activity in murine PASMCs via a mechanism requiring Rho kinase (ROCK). However, whether ROCK and/or increased NHE activity mediate ET-1-induced migration and proliferation in PASMCs remains unknown. In this study, we used fluorescent microscopy in transiently cultured PASMCs from distal pulmonary arteries of the rat and the pH-sensitive dye, BCECF-AM, to measure changes in resting pHi and NHE activity induced by exposure to exogenous ET-1 (10-8  mol/L) for 24 h. Cell migration and proliferation in response to ET-1 were also measured using Transwell assays and BrdU incorporation, respectively. We found that application of exogenous ET-1 had no effect on NHE1 expression, but increased pHi , NHE activity, migration, and proliferation in rat PASMCs. Pharmacologic inhibition of NHE or ROCK prevented the ET-1-induced changes in cell function (proliferation and migration). Our results indicate that ET-1 modulates PASMC migration and proliferation via changes in pHi homeostasis through a pathway involving ROCK.


Subject(s)
Cell Movement , Cell Proliferation , Endothelin-1/metabolism , Muscle, Smooth, Vascular/metabolism , Pulmonary Artery/metabolism , Sodium-Hydrogen Exchangers/metabolism , rho-Associated Kinases/metabolism , Animals , Cell Movement/drug effects , Cell Proliferation/drug effects , Endothelin-1/administration & dosage , Hydrogen-Ion Concentration , Male , Muscle, Smooth, Vascular/drug effects , Primary Cell Culture , Pulmonary Artery/drug effects , Rats, Wistar
16.
Physiol Rep ; 4(6)2016 Mar.
Article in English | MEDLINE | ID: mdl-27009277

ABSTRACT

Abnormal proliferation and migration of pulmonary arterial smooth muscle cells (PASMCs) are hallmark characteristics of vascular remodeling in pulmonary hypertension induced by chronic hypoxia. In this study, we investigated the role of the Na(+)/H(+)exchanger (NHE) and alterations in intracellularpH(pHi) homeostasis in meditating increased proliferation and migration inPASMCs isolated from resistance-sized pulmonary arteries from chronically hypoxic rats or from normoxic rats that were exposed to hypoxia ex vivo (1% or 4% O2, 24-96 h). We found thatPASMCs exposed to either in vivo or ex vivo hypoxia exhibited greater proliferative and migratory capacity, elevated pHi, and enhancedNHEactivity. TheNHEinhibitor, ethyl isopropyl amiloride (EIPA), normalized pHiin hypoxicPASMCs and reduced migration by 73% and 45% in cells exposed to in vivo and in vitro hypoxia, respectively. Similarly,EIPAreduced proliferation by 97% and 78% in cells exposed to in vivo and in vitro hypoxia, respectively. We previously demonstrated thatNHEisoform 1 (NHE1) is the predominant isoform expressed inPASMCs. The development of hypoxia-induced pulmonary hypertension and alterations inPASMC pHihomeostasis were prevented in mice deficient forNHE1. We found that short-term (24 h) ex vivo hypoxic exposure did not alter the expression ofNHE1, so we tested the role of Rho kinase (ROCK) as a possible means of increasingNHEactivity. In the presence of theROCKinhibitor, Y-27632, we found that pHiandNHEactivity were normalized and migration and proliferation were reduced inPASMCs exposed to either in vivo (by 68% for migration and 22% for proliferation) or ex vivo (by 43% for migration and 17% for proliferation) hypoxia. From these results, we conclude that during hypoxia, activation ofROCKenhancesNHEactivity and promotesPASMCmigration and proliferation.


Subject(s)
Cation Transport Proteins/metabolism , Cell Movement , Cell Proliferation , Hypertension, Pulmonary/etiology , Hypoxia/complications , Muscle, Smooth, Vascular/enzymology , Myocytes, Smooth Muscle/enzymology , Sodium-Hydrogen Exchangers/metabolism , Vascular Remodeling , rho-Associated Kinases/metabolism , Amides/pharmacology , Amiloride/analogs & derivatives , Amiloride/pharmacology , Animals , Cation Transport Proteins/antagonists & inhibitors , Cation Transport Proteins/deficiency , Cation Transport Proteins/genetics , Cell Hypoxia , Cell Movement/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Disease Models, Animal , Enzyme Activation , Hydrogen-Ion Concentration , Hypertension, Pulmonary/enzymology , Hypertension, Pulmonary/pathology , Hypertension, Pulmonary/physiopathology , Hypoxia/enzymology , Hypoxia/pathology , Hypoxia/physiopathology , Male , Mice, Inbred C57BL , Mice, Knockout , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/pathology , Muscle, Smooth, Vascular/physiopathology , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/pathology , Protein Kinase Inhibitors/pharmacology , Pulmonary Artery/enzymology , Pulmonary Artery/pathology , Pulmonary Artery/physiopathology , Pyridines/pharmacology , Rats, Wistar , Signal Transduction , Sodium-Hydrogen Exchanger 1 , Sodium-Hydrogen Exchangers/antagonists & inhibitors , Sodium-Hydrogen Exchangers/genetics , Vascular Remodeling/drug effects
17.
EBioMedicine ; 2(9): 1145-52, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26501112

ABSTRACT

Tenofovir (TFV) is used in combination with other antiretroviral drugs for human immunodeficiency virus (HIV) treatment and prevention. TFV requires two phosphorylation steps to become pharmacologically active; however, the kinases that activate TFV in cells and tissues susceptible to HIV infection have yet to be identified. Peripheral blood mononuclear cells (PBMC), vaginal, and colorectal tissues were transfected with siRNA targeting nucleotide kinases, incubated with TFV, and TFV-monophosphate (TFV-MP) and TFV-diphosphate (TFV-DP) were measured using mass spectrometry-liquid chromatography. Adenylate kinase 2 (AK2) performed the first TFV phosphorylation step in PBMC, vaginal, and colorectal tissues. Interestingly, both pyruvate kinase isozymes, muscle (PKM) or liver and red blood cell (PKLR), were able to phosphorylate TFV-MP to TFV-DP in PBMC and vaginal tissue, while creatine kinase, muscle (CKM) catalyzed this conversion in colorectal tissue. In addition, next-generation sequencing of the Microbicide Trials Network MTN-001 clinical samples detected 71 previously unreported genetic variants in the genes encoding these kinases. In conclusion, our results demonstrate that TFV is activated in a compartment-specific manner. Further, genetic variants have been identified that could negatively impact TFV activation, thereby compromising TFV efficacy in HIV treatment and prevention.


Subject(s)
Cell Compartmentation , Genetic Variation , Protein Kinases/genetics , Tenofovir/pharmacology , Adult , Anti-HIV Agents/pharmacology , Anti-HIV Agents/therapeutic use , Demography , Ethnicity , Female , Gene Knockdown Techniques , HIV Infections/drug therapy , High-Throughput Nucleotide Sequencing , Humans , Male , Middle Aged , Mutation, Missense/genetics , RNA, Small Interfering/metabolism , Tenofovir/therapeutic use , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL