首页 | 本学科首页   官方微博 | 高级检索  
相似文献
 共查询到20条相似文献,搜索用时 46 毫秒
1.

Background and Purpose

An influx drug/proton antiporter of unknown structure has been functionally demonstrated at the blood–brain barrier. This transporter, which handles some psychoactive drugs like diphenhydramine, clonidine, oxycodone, nicotine and cocaine, could represent a new pharmacological target in drug addiction therapy. However, at present there are no known drugs/inhibitors that effectively inhibit/modulate this transporter in vivo.

Experimental Approach

The FLAPpharm approach was used to establish a pharmacophore model for inhibitors of this transporter. The inhibitory potency of 44 selected compounds was determined against the specific substrate, [3H]‐clonidine, in the human cerebral endothelial cell line hCMEC/D3 and ranked as good, medium, weak or non‐inhibitor.

Key Results

The pharmacophore model obtained was used as a template to screen xenobiotic and endogenous compounds from databases [Specs, Recon2, Human Metabolome Database (HMDB), human intestinal transporter database], and hypothetical candidates were tested in vitro to determine their inhibitory capacity with [3H]‐clonidine. According to the transporter database, 80% of the proton antiporter inhibitor candidates could inhibit P‐glycoprotein/MDR1/ABCB1 and specificity is improved by reducing inhibitor size/shape and increasing water solubility. Virtual screening results using HMDB and Recon2 for endogenous compounds appropriately scored tryptamine as an inhibitor.

Conclusions and Implications

The pharmacophore model for the proton‐antiporter inhibitors was a good predictor of known inhibitors and allowed us to identify new good inhibitors. This model marks a new step towards the discovery of this drug/proton antiporter and will be of great use for the discovery and design of potent inhibitors that could potentially help to assess and validate its pharmacological role in drug addiction in vivo.

Abbreviations

ADE
absorption, distribution and elimination
BBB
blood–brain barrier
DPBS
Dulbecco''s PBS
DPH
diphenhydramine
G‐I
good inhibitor
Glob‐Prod flap
global product similarity score
Glob‐Sum flap
global sum similarity score
KH
Krebs–HEPES buffer
M‐I
medium inhibitor
N‐I
non‐inhibitor
PCA
principal component analysis
P‐gp
P‐glycoprotein
PK
pharmacokinetics
W‐I
weak inhibitor
  相似文献   

2.

Background and Purpose

Ischaemic heart disease can lead to serious, life‐threatening complications. Traditional therapies for ischaemia aim to increase oxygen delivery and reduce the myocardial ATP consumption by increasing the coronary perfusion and by suppressing cardiac contractility, heart rate or blood pressure. An adjunctive treatment option for ischaemia is to improve or optimize myocardial metabolism.

Experimental Approach

Metabolic suppression in the ischaemic heart is characterized by reduced levels of high‐energy molecules: ATP and NAD+. Because NAD+ is required for most metabolic processes that generate ATP, we hypothesized that restoration of NAD+ would be a prerequisite for ATP regeneration and examined the role of the major NAD+ anabolic and catabolic pathways in the bioenergetic restoration process following oxygen–glucose deprivation injury in a cardiomyocyte cell line (H9c2 cells).

Key Results

Salvage of NAD+ via nicotinamide phosphoribosyl transferase was essential for bioenergetic recovery in cardiomyocytes. Blockade of nicotinamide phosphoribosyl transferase prevented the restoration of the cellular ATP pool following oxygen–glucose deprivation injury by inhibiting both the aerobic and anaerobic metabolism in the cardiomyocytes. NAD+ consumption by PARP‐1 also undermined the recovery processes, and PARP inhibition significantly improved the metabolism and increased cellular ATP levels in cardiomyocytes.

Conclusions and Implications

We conclude that the NAD+ salvage pathway is essential for bioenergetic recovery in post‐hypoxic cardiomyocytes and PARP inhibition may represent a potential future therapeutic intervention in ischaemic heart disease.

Abbreviations

CVD
cardiovascular disease
FK866
(E)‐N‐[4‐(1‐benzoylpiperidin‐4‐yl)butyl]‐3‐(pyridin‐3‐yl)acrylamide
JC‐1
5,5′,6,6′‐tetrachloro‐1,1′,3,3′‐tetraethyl‐imidacarbocyanine iodide
MitoSOX Red
MitoSOX™ Red mitochondrial superoxide indicator
MTT
3‐(4,5‐dimethyl‐2‐thiazolyl)‐2,5‐diphenyl‐2H‐tetrazolium bromide
NamPRT
nicotinamide phosphoribosyltransferase
NMNAT
nicotinamide mononucleotide adenylyl transferase
OGD
oxygen–glucose deprivation
PJ34
N‐(6‐oxo‐5,6‐dihydrophenanthridin‐2‐yl)‐(N,N‐dimethylamino) acetamide hydrochloride
RNF146
ring finger protein 146
  相似文献   

3.

Background and Purpose

The respective impact and interplay between ABC (P‐glycoprotein/P‐gp/Abcb1a, BCRP/ABCG2, MRP/ABCC) and SLC transporter functions at the blood–brain barrier (BBB) and blood–retinal barriers (BRB) are incompletely understood.

Experimental Approach

We measured the initial cerebral and retinal distribution of selected ABC substrates by in situ carotid perfusion using P‐gp/Bcrp knockout mice and chemical ABC/SLC modulation strategies. P‐gp, Bcrp, Mrp1 and Mrp4 were studied by confocal retina imaging.

Key Results

Chemical or physical disruption of P‐gp increased [3H]‐verapamil transport by ~10‐fold at the BBB and ~1.5‐fold at the BRB. [3H]‐Verapamil transport involved influx‐mediated by an organic cation clonidine‐sensitive/diphenhydramine‐sensitive proton antiporter at both barriers; this effect was unmasked when P‐gp was partially or fully inhibited/disrupted at the BBB. Studies of [3H]‐mitoxantrone and [3H]‐zidovudine transport suggested, respectively, that Bcrp efflux was less involved at the BRB than BBB, whereas Mrps were significantly and similarly involved at both barriers. Confocal imaging showed that P‐gp and Bcrp were expressed in intra‐retinal vessels (inner BRB/iBRB) but absent from the blood/basal membrane of cells of the retinal pigment epithelium (outer BRB/oBRB/RPE) where, in contrast, Mrp1 and Mrp4 were localized.

Conclusions and Implications

P‐gp, Bcrp, Mrp1 and Mrp4 are differentially expressed at the outer and inner BRB, resulting in an altered ability to limit substrate distribution at the retina as compared with the BBB. [3H]‐Verapamil distribution is not P‐gp‐specific and involves a proton antiporter at both the BBB and BRB. However, this transport is concealed by P‐gp at the BBB, but not at the BRB, where P‐gp activity is reduced.

Abbreviations

ABC
ATP‐binding cassette
BBB
blood–brain barrier
BRB
blood–retina barrier
CV
choroidal vessels
DPH
diphenhydramine; Elac, elacridar
iBRB
inner blood–retina barrier
KO
knockout
oBRB
outer blood–retina barrier
PD
pharmacodynamics
P‐gp
P‐glycoprotein
pHe
extracellular/vascular pH
pHi
intracellular pH
PK
pharmacokinetics
RPE
retinal pigment epithelial cells
SLC
solute carrier
TKO
triple knockout [Abcb1a−/−, Abcb1b−/−, Abcg2−/−] mice
WT
wild type
  相似文献   

4.
Despite the progress being made toward development of less-toxic and simpler alternatives to the current peg-IFN standard-of-care therapy for chronic hepatitis C, the highly replicative nature of HCV infection and the error-prone nature of its viral RNA synthesis pose extraordinary challenges to drug development. Peg-IFN is likely to remain a mainstay of therapy for the foreseeable future, or until such time that multiple direct-acting STAT-C inhibitors are available and shown to provide a sufficiently high barrier to resistance when used in combination.

References and recommended reading

Papers of particular interest, published within the period of review, have been highlighted as:
• of special interest
•• of outstanding interest

References

1 S.M. Lemon, C. Walker, M.J. Alter and M. Yi, Hepatitis C viruses. In: D. Knipe, P. Howley, D.E. Griffin, M.A. Martin, R.A. Lamb, B. Roizman and S.E. Straus, Editors, Fields Virology (edn 5), Lippincott Williams & Wilkins, Philadelphia (2007), pp. 1253–1304.
2 V. Lohmann, F. Korner, J. Koch, U. Herian, L. Theilmann and R. Bartenschlager, Replication of subgenomic hepatitis C virus RNAs in a hepatoma cell line, Science 285 (1999), pp. 110–113. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (1159)
3 T. Wakita, T. Pietschmann, T. Kato, T. Date, M. Miyamoto, Z. Zhao, K. Murthy, A. Habermann, H.-G. Krauslich and M. Mizokami et al., Production of infectious hepatitis C virus in tissue culture from a cloned viral genome, Nat Med 11 (2005), pp. 791–796. View Record in Scopus | Cited By in Scopus (630)
4 M. Yi, R.A. Villanueva, D.L. Thomas, T. Wakita and S.M. Lemon, Production of infectious genotype 1a hepatitis C virus (Hutchinson strain) in cultured human hepatoma cells, Proc Natl Acad Sci U S A 103 (2006), pp. 2310–2315. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (113)
5•• A.U. Neumann, N.P. Lam, H. Dahari, D.R. Gretch, T.E. Wiley, T.J. Layden and A.S. Perelson, Hepatitis C viral dynamics in vivo and the antiviral efficacy of interferon-alpha therapy, Science 282 (1998), pp. 103–107. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (891)The authors modeled decreases in circulating viral RNA in patients receiving therapy with IFN, and in doing so provided novel insights into both the dynamics of HCV replication in infected persons as well as the mechanism of IFN action.  相似文献   

5.
6.
7.

Background and Purpose

In the phase III clinical trial, RELAX‐AHF, serelaxin caused rapid and long‐lasting haemodynamic changes. However, the cellular mechanisms involved are unclear in humans.

Experimental Approach

This study examined the effects of serelaxin in co‐cultures of human primary endothelial cells (ECs) and smooth muscle cells (SMCs) on cAMP and cGMP signalling.

Key Results

Stimulation of HUVECs or human coronary artery endothelial cells (HCAECs) with serelaxin, concentration‐dependently increased cGMP accumulation in co‐cultured SMCs to a greater extent than in monocultures of either cell type. This was not observed in human umbilical artery endothelial cells (HUAECs) that do not express the relaxin receptor, RXFP1. Treatment of ECs with l‐NG‐nitro arginine (NOARG; 30 μM, 30 min) inhibited serelaxin‐mediated (30 nM) cGMP accumulation in HUVECs, HCAECs and co‐cultured SMCs. In HCAECs, but not HUVECs, pre‐incubation with indomethacin (30 μM, 30 min) also inhibited cGMP accumulation in SMCs. Pre‐incubation of SMCs with the guanylate cyclase inhibitor ODQ (1 μM, 30 min) had no effect on serelaxin‐mediated (30 nM) cGMP accumulation in HUVECs and HCAECs but inhibited cGMP accumulation in SMCs. Serelaxin stimulation of HCAECs, but not HUVECs, increased cAMP accumulation concentration‐dependently in SMCs. Pre‐incubation of HCAECs with indomethacin, but not l‐NOARG, abolished cAMP accumulation in co‐cultured SMCs, suggesting involvement of prostanoids.

Conclusions and Implications

In co‐cultures, treatment of ECs with serelaxin caused marked cGMP accumulation in SMCs and with HCAEC also cAMP accumulation. Responses involved EC‐derived NO and with HCAEC prostanoid production. Thus, serelaxin differentially modulates vascular tone in different vascular beds.

Abbreviations

AHF
acute heart failure
DEA
diethylamine NONOate
ECs
endothelial cells
HCAEC
human coronary artery endothelial cell
HUAEC
human umbilical artery endothelial cell
HUASMC
human umbilical artery smooth muscle cell
HUVSMC
human umbilical vein smooth muscle cell
l‐NOARG
l‐NG‐nitro arginine
SMCs
smooth muscle cells
  相似文献   

8.

Background and Purpose

Meningeal blood flow is controlled by CGRP released from trigeminal afferents and NO mainly produced in arterial endothelium. The vasodilator effect of NO may be due to the NO–derived compound, nitroxyl (HNO), generated through reaction with endogenous H2S. We investigated the involvement of HNO in CGRP release and meningeal blood flow.

Experimental Approach

Blood flow in exposed dura mater of rats was recorded by laser Doppler flowmetry. CGRP release from the dura mater in the hemisected rat head was quantified using an elisa. NO and H2S were localized histochemically with specific sensors.

Key Results

Topical administration of the NO donor diethylamine‐NONOate increased meningeal blood flow by 30%. Pretreatment with oxamic acid, an inhibitor of H2S synthesis, reduced this effect. Administration of Na2S increased blood flow by 20%, an effect abolished by the CGRP receptor antagonist CGRP 8‐37 or the TRPA1 channel antagonist HC030031 and reduced when endogenous NO synthesis was blocked. Na2S dose‐dependently increased CGRP release two‐ to threefold. Co‐administration of diethylamine‐NONOate facilitated CGRP release, while inhibition of endogenous NO or H2S synthesis lowered basal CGRP release. NO and H2S were mainly localized in arterial vessels, HNO additionally in nerve fibre bundles. HNO staining was lost after treatment with LNMMA and oxamic acid.

Conclusions and Implications

NO and H2S cooperatively increased meningeal blood flow by forming HNO, which activated TRPA1 cation channels in trigeminal fibres, inducing CGRP release. This HNOTRPA1‐CGRP signalling pathway may be relevant to the pathophysiology of headaches.

Abbreviations

CBS
cystathionine β‐synthase
CSE
cystathionine γ‐lyase
Cy3
cyanine dye 3
DAF
4‐amino‐5‐methylamino‐2′,7′‐difluoresceine diacetate
HNO
nitroxyl
iNOS
inducible NOS
L‐NMMA
L‐NG‐monomethylarginine acetate
MMA
middle meningeal artery
MST
mercaptopyruvate sulfurtransferase
nNOS
neuronal NOS
NONOate
diethylamine‐NONOate, DEANONOate
ODQ
1H‐[1,2,4]oxadiazole[4,3‐a]quinoxalin‐1‐one
sGC
soluble GC
SIF
synthetic interstitial fluid
TRPA1
transient receptor potential ankyrin 1 channel
Tables of Links
TARGETS
Ion channels a Enzymes c
TRPA1 channel CBS, cystathionine β‐synthase
GPCRs b CSE, cystathionine γ‐lyase
CGRP receptor MST, mercaptopyruvate sulfur transferase
nNOS
eNOS
Open in a separate window
LIGANDS
CGRP
CGRP8‐37
HC030031
NO
ODQ, 1H‐[1,2,4]oxadiazole[4,3‐a]quinoxalin‐1‐one
Open in a separate windowThese Tables list key protein targets and ligands in this article which are hyperlinked to corresponding entries in http://www.guidetopharmacology.org, the common portal for data from the IUPHAR/BPS Guide to PHARMACOLOGY (Pawson et al., 2014) and are permanently archived in the Concise Guide to PHARMACOLOGY 2013/14 (a,b,cAlexander et al., 2013a, 2013b, 2013c).  相似文献   

9.
In order to better inform scientific decision making in the occupational environment, we need a better understanding of the toxicology of mixed exposures. In particular, we need an understanding of the dose–response relationship from the level of individual or population exposure down to the molecular level (and then back up again from the molecular level to the specific health-related response of the organism as a whole). Mixtures toxicology is proving to be different from single-chemical toxicology in several fundamental but barely recognized ways:
• complex chemical mixtures may consist of thousands of (often unidentified) components, each often at very low doses, but together constituting significant exposure levels;
• exposure is nearly always via multiple routes, pathways;
• other stressors such as noise, heat, infection, etc., may play a significant role in the overall environmental health response;
• interactions are potentially many and varied: pharmacokinetic and pharmacodynamic interactions may occur at the same site, or at different sites via complex physiological processes (including defense mechanisms);
• cumulative effects of different exposures/stresses over time need to be considered (altering the “baseline” susceptibility of the individual).
Knowledge gained in mixtures research should be able to improve current risk assessment and mitigation or intervention methods. In NIOSH's National Occupational Research Agenda (NORA) three priority areas have been identified:
Epidemiology: Improvement of statistical tools to identify mixed effects from available epidemiological data (“confounding factors are mixtures effects”). Awareness of disease states with significant environmental components (Gulf War Syndrome (GWS), Chronic Fatigue Syndrome (CFS), Multiple Chemical Sensitivity (MCS), etc.). Recognition and investigation of the complexity of disease related responses to multiple (simultaneous and serial) stressors (immune system, endocrine system, nervous system, etc.). Wider emphasis on relatively new concepts such as susceptibility, which links the genetic and environmental components of a disease. Disease itself is of course also a stressor.
Laboratory approaches: Methods need to be developed to understand and integrate experimental data from the molecular to the whole organism level for understanding multiple data (proteomics, genomics, and metabolomics/metabonomics) from mixed exposures. Understanding and prediction of precursors to adverse health effects will inevitably lead to identification of useful biomarkers of effect, and to earlier and more effective intervention strategies. We also need to improve our ability to forecast interaction effects from mixed exposures using less costly cellular-based screening tools and computer modeling (e.g. QSAR), and develop improved models for large-scale studies of the nature of chemical interactions that lump responses by chemical classes.
Modeling as integrator of data: Development and validation of mechanism-based models and predictive tools are essential for improving current risk assessment processes for mixtures. For example, current linked physiologically-based pharmacokinetic/pharmacodynamic (PBPK/PD) models for multiple simultaneous exposures to chemicals (such as BTEX) need to be extended to more complex mixtures, and new statistical methods of dealing with possibly thousands of components need to be developed. Ultimately, such models may become an integral part of a model of the “virtual human” via computer simulation.
Keywords: Modeling; Risk assessment; PBPK; Mixtures interactions; Chronic disease; Susceptibility; Toxicokinetics  相似文献   

10.

Background and Purpose

α7 nicotinic acetylcholine receptors (α7 nAChRs) may represent useful targets for cognitive improvement. The aim of this study is to compare the pro‐cognitive activity of selective α7‐nAChR ligands, including the partial agonists, DMXBA and A‐582941, as well as the positive allosteric modulator, 3‐furan‐2‐yl‐N‐p‐tolyl‐acrylamide (PAM‐2).

Experimental Approach

The attentional set‐shifting task (ASST) and the novel object recognition task (NORT) in rats, were used to evaluate the pro‐cognitive activity of each ligand [i.e., PAM‐2 (0.5, 1.0, and 2.0 mg·kg−1), DMXBA and A‐582941 (0.3 and 1.0 mg·kg−1)], in the absence and presence of methyllycaconitine (MLA), a selective competitive antagonist. To determine potential drug interactions, an inactive dose of PAM‐2 (0.5 mg·kg−1) was co‐injected with inactive doses of either agonist ‐ DMXBA: 0.1 (NORT); 0.3 mg·kg−1 (ASST) or A‐582941: 0.1 mg·kg−1.

Key Results

PAM‐2, DMXBA, and A‐582941 improved cognition in a MLA‐dependent manner, indicating that the observed activities are mediated by α7 nAChRs. Interestingly, the co‐injection of inactive doses of PAM‐2 and DMXBA or A‐582941 also improved cognition, suggesting drug interactions. Moreover, PAM‐2 reversed the scopolamine‐induced NORT deficit. The electrophysiological results also support the view that PAM‐2 potentiates the α7 nAChR currents elicited by a fixed concentration (3 μM) of DMXBA with apparent EC50 = 34 ± 3 μM and Emax = 225 ± 5 %.

Conclusions and Implications

Our results support the view that α7 nAChRs are involved in cognition processes and that PAM‐2 is a novel promising candidate for the treatment of cognitive disorders.

Abbreviations

α7 nAChR
nicotinic acetylcholine receptor with α7 subunit
AD
Alzheimer''s disease
apparent EC50
enhancement potency
ASST
attentional set‐shifting task
CD
compound discrimination
DI
discrimination index
E
exploration time
ED
extra‐dimensional
Emax
ligand efficacy
ID
intra‐dimensional
ITI
inter‐trial interval
MLA
methyllycaconitine
NORT
novel object recognition task
nH
Hill coefficient
PAM
positive allosteric modulator
PAM‐2
3‐furan‐2‐yl‐N‐p‐tolyl‐acrylamide
Rev
reversal of discrimination
SD
simple discrimination
T1
familiarisation trial
T2
retention trial
  相似文献   

11.
Mitochondrial permeability transition pore (mPTP) opening plays a critical role in cardiac reperfusion injury and its prevention is cardioprotective. Tumour cell mitochondria usually have high levels of hexokinase isoform 2 (HK2) bound to their outer mitochondrial membranes (OMM) and HK2 binding to heart mitochondria has also been implicated in resistance to reperfusion injury. HK2 dissociates from heart mitochondria during ischaemia, and the extent of this correlates with the infarct size on reperfusion. Here we review the mechanisms and regulations of HK2 binding to mitochondria and how this inhibits mPTP opening and consequent reperfusion injury. Major determinants of HK2 dissociation are the elevated glucose‐6‐phosphate concentrations and decreased pH in ischaemia. These are modulated by the myriad of signalling pathways implicated in preconditioning protocols as a result of a decrease in pre‐ischaemic glycogen content. Loss of mitochondrial HK2 during ischaemia is associated with permeabilization of the OMM to cytochrome c, which leads to greater reactive oxygen species production and mPTP opening during reperfusion. Potential interactions between HK2 and OMM proteins associated with mitochondrial fission (e.g. Drp1) and apoptosis (B‐cell lymphoma 2 family members) in these processes are examined. Also considered is the role of HK2 binding in stabilizing contact sites between the OMM and the inner membrane. Breakage of these during ischaemia is proposed to facilitate cytochrome c loss during ischaemia while increasing mPTP opening and compromising cellular bioenergetics during reperfusion. We end by highlighting the many unanswered questions and discussing the potential of modulating mitochondrial HK2 binding as a pharmacological target.

Linked Articles

This article is part of a themed section on Conditioning the Heart – Pathways to Translation. To view the other articles in this section visit http://dx.doi.org/10.1111/bph.2015.172.issue‐8

Abbreviations

Akt
also known as PKB
AMPK
AMP‐activated PK
ANT
adenine nucleotide translocase
Bad
Bcl‐2‐associated death promoter
Bak
Bcl‐2 homologous antagonist/killer
Bax
Bcl‐2‐like protein 4
Bcl‐2
B‐cell lymphoma 2
Bcl‐xL
Bcl‐2‐extra large
CK
creatine kinase
CsA
cyclosporine A
CyP‐D
cyclophilin D
Drp1
dynamin‐related protein 1
G‐6‐P
glucose‐6‐phosphate
GSK3β
glycogen synthase kinase 3β
HK
hexokinase
I/R
ischaemia/reperfusion
IF1
ATP synthase inhibitor factor 1
IMM
inner mitochondrial membrane
IP
ischaemic preconditioning
mPTP
mitochondrial permeability transition pore
OMM
outer mitochondrial membrane
Opa‐1
optic athrophy 1
PCr
phosphocreatine
PPIase
peptidylprolyl isomerase
ROS
reactive oxygen species
SR
sarcoplasmic reticulum
T0
time of ischaemic rigor start
TAT‐HK2
cell‐permeable peptide of HK2 binding domain
TP
temperature preconditioning
TSPO
translocator protein of the outer membrane
VDAC
voltage‐dependent anion channel
Tables of Links
TARGETS
Ion channels a Enzymes c
Connexin 43 Akt (PKB) GSK3β
VDAC AMPK PKA
Transporters b Caspases PKCε
NHE1 Calpains Peptidylprolyl isomerase
F‐type ATPase
Open in a separate window
LIGANDS
Carboxyatractyloside
Clotrimazole
Cyclosporine A
H2O2
Metformin
Open in a separate windowThese Tables list key protein targets and ligands in this article which are hyperlinked to corresponding entries in http://www.guidetopharmacology.org, the common portal for data from the IUPHAR/BPS Guide to PHARMACOLOGY (Pawson et al., 2014) and are permanently archived in the Concise Guide to PHARMACOLOGY 2013/14 (a,b,cAlexander et al., 2013a, 2013b, 2013c).  相似文献   

12.

Background and Purpose

The activation of M 3 cholinoceptors (M 3 receptors) by choline reduces cardiovascular risk, but it is unclear whether these receptors can regulate ischaemia/reperfusion (I/R)‐induced vascular injury. Thus, the primary goal of the present study was to explore the effects of choline on the function of mesenteric arteries following I/R, with a major focus on Ca2+/calmodulin‐dependent protein kinase II (CaMKII) regulation.

Experimental Approach

Rats were given choline (10 mg·kg−1, i.v.) and then the superior mesenteric artery was occluded for 60 min (ischaemia), followed by 90 min of reperfusion. The M 3 receptor antagonist, 4‐diphenylacetoxy‐N‐methylpiperidine methiodide (4‐DAMP), was injected (0.12 μg·kg−1, i.v.) 5 min prior to choline treatment. Vascular function was examined in rings of mesenteric arteries isolated after the reperfusion procedure. Vascular superoxide anion production, CaMKII and the levels of Ca2+‐cycling proteins were also assessed.

Key Results

Choline treatment attenuated I/R‐induced vascular dysfunction, blocked elevations in the levels of reactive oxygen species (ROS) and decreased the up‐regulated expression of oxidised CaMKII and phosphorylated CaMKII. In addition, choline reversed the abnormal expression of Ca2+‐cycling proteins, including Na+/Ca2+ exchanger, inositol 1,4,5‐trisphosphate receptor, sarcoplasmic reticulum Ca2+ATPase and phospholamban. All of these cholinergic effects of choline were abolished by 4‐DAMP.

Conclusions and Implications

Our data suggest that inhibition of the ROS‐mediated CaMKII pathway and modulation of Ca2+‐cycling proteins may be novel mechanisms underlying choline‐induced vascular protection. These results represent a significant addition to the understanding of the pharmacological roles of M 3 receptors in the vasculature, providing a new therapeutic strategy for I/R‐induced vascular injury.

Linked Articles

This article is part of a themed section on Chinese Innovation in Cardiovascular Drug Discovery. To view the other articles in this section visit http://dx.doi.org/10.1111/bph.2015.172.issue-23

Abbreviations

4‐DAMP
4‐diphenylacetoxy‐N‐methylpiperidine methiodide
CaMKII
Ca2+/calmodulin‐dependent protein kinase II
DHE
dihydroethidium
I/R
ischaemia/reperfusion
IP3R
inositol 1,4,5‐trisphosphate receptor
NAC
N‐acetyl‐L‐cysteine
NCX
Na+/Ca2+ exchanger
PLB
phospholamban
ROS
reactive oxygen species
SERCA
sarcoplasmic reticulum Ca2+‐ATPase
SNP
sodium nitroprusside
Tables of Links
TARGETS
GPCRs a
M3 receptors
Enzymes b
SERCA 2, sarcoplasmic reticulum Ca2+‐ATPase
Ion channels c
NCX1, Na+/Ca2+ exchanger
Ligand‐gated ion channels d
IP3R, inositol 1,4,5‐trisphosphate receptor
Open in a separate window
LIGANDS
4‐DAMP, 4‐diphenylacetoxy‐N‐methylpiperidine methiodide
5‐HT
ACh
Caffeine
Choline
Darifenacin
KN‐93
L‐NAME, NG‐nitro‐L‐arginine methyl ester
Phenylephrine
Open in a separate windowThese Tables list key protein targets and ligands in this article which are hyperlinked to corresponding entries in http://www.guidetopharmacology.org, the common portal for data from the IUPHAR/BPS Guide to PHARMACOLOGY (Pawson et al., 2014) and are permanently archived in the Concise Guide to PHARMACOLOGY 2013/14 (a,b,c,dAlexander et al., 2013a, 2013b, 2013c, 2013d).  相似文献   

13.
To predict the safety of a drug at an early stage in its development is a major challenge as there is a lack of in vitro heart models that correlate data from preclinical toxicity screening assays with clinical results. A biophysically detailed computer model of the heart, the virtual heart, provides a powerful tool for simulating drug–ion channel interactions and cardiac functions during normal and disease conditions and, therefore, provides a powerful platform for drug cardiotoxicity screening. In this article, we first review recent progress in the development of theory on drug–ion channel interactions and mathematical modelling. Then we propose a family of biomarkers that can quantitatively characterize the actions of a drug on the electrical activity of the heart at multi‐physical scales including cellular and tissue levels. We also conducted some simulations to demonstrate the application of the virtual heart to assess the pro‐arrhythmic effects of cisapride and amiodarone. Using the model we investigated the mechanisms responsible for the differences between the two drugs on pro‐arrhythmogenesis, even though both prolong the QT interval of ECGs. Several challenges for further development of a virtual heart as a platform for screening drug cardiotoxicity are discussed.

Linked Articles

This article is part of a themed section on Chinese Innovation in Cardiovascular Drug Discovery. To view the other articles in this section visit http://dx.doi.org/10.1111/bph.2015.172.issue-23

Abbreviations

AE
allosteric effector
APD
action potential duration
APD90
APD at 90% repolarization
APs
action potentials
BCL
basic cycle length
CV
conduction velocity
CVR
conduction velocity restitution
ERP
effective refractory period
GR
guarded receptor
HH
Hodgkin–Huxley
ICaL
L‐type Ca2+ current
IKr
delayed rectifier K+ channel current
IKtof
fast component of the cardiac transient outward current
INa
Na+ channel current
LQTs
long QT syndrome
MR
modulated receptor
QTc
corrected QT interval
VW
vulnerable window
WL
wavelength
Tables of Links
TARGETS
GPCRs a Ion channels b
β‐adrenoceptors hERG (KV11.1) channels
L‐type Ca2+ channels
Voltage‐gated K+ channels
Voltage‐gated Na+ channels
Open in a separate window
LIGANDS
Amiodarone Flecainide
Cisapride Lidocaine
Clozapine Mexiletine
E‐4031 Ranolazine
Quinidine
Open in a separate windowThese Tables list key protein targets and ligands in this article which are hyperlinked to corresponding entries in http://www.guidetopharmacology.org, the common portal for data from the IUPHAR/BPS Guide to PHARMACOLOGY (Pawson et al., 2014) and are permanently archived in the Concise Guide to PHARMACOLOGY 2013/14 (a,bAlexander et al., 2013a, 2013b).  相似文献   

14.

Background and Purpose

11β‐hydroxysteroid dehydrogenase type I (11β‐HSD1), a target for Type 2 diabetes mellitus, converts inactive glucocorticoids into bioactive forms, increasing tissue concentrations. We have compared the pharmacokinetic‐pharmacodynamic (PK/PD) relationship of target inhibition after acute and repeat administration of inhibitors of 11β‐HSD1 activity in human, rat and mouse adipose tissue (AT).

Experimental Approach

Studies included abdominally obese human volunteers, rats and mice. Two specific 11β‐HSD1 inhibitors (AZD8329 and COMPOUND‐20) were administered as single oral doses or repeat daily doses for 7–9 days. 11β‐HSD1 activity in AT was measured ex vivo by conversion of 3H‐cortisone to 3H‐cortisol.

Key Results

In human and rat AT, inhibition of 11β‐HSD1 activity was lost after repeat dosing of AZD8329, compared with acute administration. Similarly, in rat AT, there was loss of inhibition of 11β‐HSD1 activity after repeat dosing with COMPOUND‐20 with continuous drug cover, but effects were substantially reduced if a ‘drug holiday’ period was maintained daily. Inhibition of 11β‐HSD1 activity was not lost in mouse AT after continuous cover with COMPOUND‐20 for 7 days.

Conclusions and Implications

Human and rat AT, but not mouse AT, exhibited tachyphylaxis for inhibition of 11β‐HSD1 activity after repeat dosing. Translation of observed efficacy in murine disease models to human for 11β‐HSD1 inhibitors may be misleading. Investigators of the effects of 11β‐HSD1 inhibitors should confirm that desired levels of enzyme inhibition in AT can be maintained over time after repeat dosing and not rely on results following a single dose.

Abbreviations

11β‐HSD1
11β‐hydroxysteroid dehydrogenase type I
PK/PD
pharmacokinetic‐pharmacodynamic
AT
adipose tissue
DIO
diet induced obese
IHC
International Conference on Harmonisation
GCP
Good Clinical Practice
b.i.d.
twice daily
u.i.d.
once daily
HPMC
hydroxypropylmethylcellulose
IC70
concentration that delivers 70% of the maximum effect
IC90
concentration that delivers 90% of the maximum effect
fu
fraction unbound
Cmax
maximum achieved concentration
Cmin
minimum or trough concentration
E0
baseline
Emax
maximum effect
ANCOVA
analysis of covariance
  相似文献   

15.
16.

Background and Purpose

Allosteric modulation of the mGlu2 receptor is a potential strategy for treatment of various neurological and psychiatric disorders. Here, we describe the in vitro characterization of the mGlu2 positive allosteric modulator (PAM) JNJ‐46281222 and its radiolabelled counterpart [3H]‐JNJ‐46281222. Using this novel tool, we also describe the allosteric effect of orthosteric glutamate binding and the presence of a bound G protein on PAM binding and use computational approaches to further investigate the binding mode.

Experimental Approach

We have used radioligand binding studies, functional assays, site‐directed mutagenesis, homology modelling and molecular dynamics to study the binding of JNJ‐46281222.

Key Results

JNJ‐46281222 is an mGlu2‐selective, highly potent PAM with nanomolar affinity (K D = 1.7 nM). Binding of [3H]‐JNJ‐46281222 was increased by the presence of glutamate and greatly reduced by the presence of GTP, indicating the preference for a G protein bound state of the receptor for PAM binding. Its allosteric binding site was visualized and analysed by a computational docking and molecular dynamics study. The simulations revealed amino acid movements in regions expected to be important for activation. The binding mode was supported by [3H]‐JNJ‐46281222 binding experiments on mutant receptors.

Conclusion and Implications

Our results obtained with JNJ‐46281222 in unlabelled and tritiated form further contribute to our understanding of mGlu2 allosteric modulation. The computational simulations and mutagenesis provide a plausible binding mode with indications of how the ligand permits allosteric activation. This study is therefore of interest for mGlu2 and class C receptor drug discovery.

Abbreviations

JNJ‐46281222
3‐(Cyclopropylmethyl)‐7‐[(4‐phenyl‐1‐piperidinyl)methyl]‐8‐(trifluoromethyl)‐1,2,4‐triazolo[4,3‐a]pyridine
NAM
negative allosteric modulator
PAM
positive allosteric modulator
VFT
Venus Flytrap domain
  相似文献   

17.

Background and Purpose

Angiotensin II (AngII) induces migration and growth of vascular smooth muscle cell (VSMC), which is responsible for vascular remodelling in some cardiovascular diseases. Ang II also activates a Cl current, but the underlying mechanism is not clear.

Experimental Approach

The A10 cell line and primary cultures of VSMC from control, ClC‐3 channel null mice and WT mice made hypertensive with AngII infusions were used. Techniques employed included whole‐cell patch clamp, co‐immunoprecipitation, site‐specific mutagenesis and Western blotting,

Key Results

In VSMC, AngII induced Cl currents was carried by the chloride ion channel ClC‐3. This current was absent in VSMC from ClC‐3 channel null mice. The AngII‐induced Cl current involved interactions between ClC‐3 channels and Rho‐kinase 2 (ROCK2), shown by N‐ or C‐terminal truncation of ClC‐3 protein, ROCK2 siRNA and co‐immunoprecipitation assays. Phosphorylation of ClC‐3 channels at Thr532 by ROCK2 was critical for AngII‐induced Cl current and VSMC migration. The ClC‐3 T532D mutant (mutation of Thr532 to aspartate), mimicking phosphorylated ClC‐3 protein, significantly potentiated AngII‐induced Cl current and VSMC migration, while ClC‐3 T532A (mutation of Thr532 to alanine) had the opposite effects. AngII‐induced cell migration was markedly decreased in VSMC from ClC‐3 channel null mice that was insensitive to Y27632, an inhibitor of ROCK2. In addition, AngII‐induced cerebrovascular remodelling was decreased in ClC‐3 null mice, possibly by the ROCK2 pathway.

Conclusions And Implications

ClC‐3 protein phosphorylation at Thr532 by ROCK2 is required for AngII‐induced Cl current and VSMC migration that are involved in AngII‐induced vascular remodelling in hypertension.

Abbreviations

ΔCT
C‐terminal truncated ClC‐3
ΔNT
N‐terminal truncated ClC‐3
BASMC
basilar artery smooth muscle cells
caROCK2
constitutively active ROCK2
dnROCK2
dominant negative ROCK2
N1
EGFP‐N1 plasmid
VRCC
volume‐regulated chloride channel
VSMC
vascular smooth muscle cells
  相似文献   

18.

Background and Purpose

Nuciferine, a constituent of lotus leaf, is an aromatic ring‐containing alkaloid, with antioxidative properties. We hypothesize nuciferine might affect vascular reactivity. This study aimed at determining the effects of nuciferine on vasomotor tone and the underlying mechanism

Experimental Approach

Nuciferine‐induced relaxations in rings of rat main mesenteric arteries were measured by wire myographs. Endothelial NOS (eNOS) was determined by immunoblotting. Intracellular NO production in HUVECs and Ca2+ level in both HUVECs and vascular smooth muscle cells (VSMCs) from rat mesenteric arteries were assessed by fluorescence imaging.

Key Results

Nuciferine induced relaxations in arterial segments pre‐contracted by KCl or phenylephrine. Nuciferine‐elicited arterial relaxations were reduced by removal of endothelium or by pretreatment with the eNOS inhibitor LNAME or the NO‐sensitive guanylyl cyclase inhibitor ODQ. In HUVECs, the phosphorylation of eNOS at Ser1177 and increase in cytosolic NO level induced by nuciferine were mediated by extracellular Ca2+ influx. Under endothelium‐free conditions, nuciferine attenuated CaCl2‐induced contraction in Ca2+‐free depolarizing medium. In the absence of extracellular calcium, nuciferine relieved the vasoconstriction induced by phenylephrine and the addition of CaCl2. Nuciferine also suppressed Ca2+ influx in Ca2+‐free K+‐containing solution in VSMCs.

Conclusions and Implications

Nuciferine has a vasorelaxant effect via both endothelium‐dependent and ‐independent mechanisms. These results suggest that nuciferine may have a therapeutic effect on vascular diseases associated with aberrant vasoconstriction.

Linked Articles

This article is part of a themed section on Chinese Innovation in Cardiovascular Drug Discovery. To view the other articles in this section visit http://dx.doi.org/10.1111/bph.2015.172.issue-23

Abbreviations

[Ca2+]i
intracellular calcium
[NO]i
intracellular NO
DAF‐FM DA
4‐amino‐5‐methylamino‐2′,7′‐difluorofluorescein diacetate
EDHF
endothelium‐derived hyperpolarizing factor
eNOS
endothelial NOS
Fluo‐4 AM
fluo‐4‐acetoxymethylester
iNOS
inducible NOS
L‐NAME
Nω‐nitro‐L‐arginine methyl ester
ODQ
1H‐[1,2,4]oxadizolo[4,3‐a]quinoxalin‐1‐one
VDCCs
voltage‐dependent Ca2+ channels
VSMCs
vascular smooth muscle cells
Tables of Links
TARGETS
Enzymes a
eNOS
iNOS
Ion channels b
KATP channels, Kir6.2
VDCC, voltage‐dependent calcium channels
GPCRs c
α1‐adrenoceptors
β‐adrenoceptors
Open in a separate window
LIGANDS
1400W
Indomethacin
L‐NAME
Nifedipine
NO
ODQ
Phenylephrine
Propranolol
Open in a separate windowThese Tables list key protein targets and ligands in this article which are hyperlinked to corresponding entries in http://www.guidetopharmacology.org, the common portal for data from the IUPHAR/BPS Guide to PHARMACOLOGY (Pawson et al., 2014) and are permanently archived in the Concise Guide to PHARMACOLOGY 2013/14 (a,b,cAlexander et al., 2013a, 2013b, 2013c).  相似文献   

19.
Cardiovascular disease has become the most serious health threat and represents the major cause of morbidity and mortality in China, as in other industrialized nations. During the past few decades, China''s economic boom has tremendously improved people''s standard of living but has also changed their lifestyle, increasing the prevalence of cardiovascular disease, the so‐called ‘disease of modern civilization’. This new trend has attracted a significant amount of research. Many of the studies conducted by Chinese investigators are orientated towards understanding the molecular mechanisms of cardiovascular disease. At the molecular level, the long‐standing consensus is that cardiovascular disease is associated with a sequence mutation (genetic anomaly) and expression deregulation (epigenetic disorder) of protein‐coding genes. However, new research data have established the non‐protein‐coding genes microRNAs (miRNAs) as a central regulator of the pathogenesis of cardiac disease and a potential new therapeutic target for cardiovascular disease. These small non‐coding RNAs have also been subjected to extensive, rigorous investigations by Chinese researchers. Over the years, a large body of studies on miRNAs in cardiovascular disease has been conducted by Chinese investigators, yielding fruitful research results and a better understanding of miRNAs as a new level of molecular mechanisms for the pathogenesis of cardiac disease. In this review, we briefly summarize the current status of research in the field of miRNAs and cardiovascular disease in China, highlighting the advances made in elucidating the role of miRNAs in various cardiac conditions, including cardiac arrhythmia, myocardial ischaemia, cardiac hypertrophy and heart failure. We have also examined the potential of miRNAs as novel diagnostic biomarkers and therapeutic targets.

Linked Articles

This article is part of a themed section on Chinese Innovation in Cardiovascular Drug Discovery. To view the other articles in this section visit http://dx.doi.org/10.1111/bph.2015.172.issue-23

Abbreviations

AF
atrial fibrillation
AMI
acute myocardial infarction
APD
action potential duration
Cx43
connexin 43 (also known as GJA1, gap junction protein, α1)
Drp1
dynamin‐related protein‐1
E‐C
excitation‐contraction
HSP60
heat shock protein 60
ICaL
L type calcium current
JP2
junctophilin‐2
KCNJ2
gene for potassium inwardly rectifying channel, subfamily J, member 2 also known as Kir2.1, inwardly rectifying potassium channel 2.1
LNA
locked nucleic acid
MI
myocardial infarct
miRNAs
microRNAs
NFAT
nuclear factor of activated T‐cells
SERCA2a
sarcoplasmic reticulum calcium ATPase
KCa2.3 (also known as SK3)
small conductance calcium‐activated potassium channels 3
TGF‐β1
transforming growth factor‐β1
TGFBR2
transforming growth factor‐β receptor II
Tables of Links
TARGETS
GPCRs a Ion channels c Enzymes e
β‐adrenoceptors Connexin 43 (Cx43) Caspase 8
Ligand‐gated ion channels b Cav1.2 Furin
Ryanodine receptor Cav1.3 PKA
Catalytic receptors d KCa2.1 PKCε
CCK4 (serum response factor) KCa2.3 (SK3) SERCA2
TGFBR2 Kir2.1
Open in a separate window
LIGANDS
Aldosterone HSP60
Angiotensin II Isoprenaline
cAMP Propranolol
H2O2 TGF‐β1
Open in a separate windowThese Tables list key protein targets and ligands in this article which are hyperlinked to corresponding entries in http://www.guidetopharmacology.org, the common portal for data from the IUPHAR/BPS Guide to PHARMACOLOGY (Pawson et al., 2014) and are permanently archived in the Concise Guide to PHARMACOLOGY 2013/14 (a,b,c,d,eAlexander et al., 2013a, 2013b, 2013c, 2013d, 2013e).  相似文献   

20.

Background and Purpose

7‐[2‐[4‐(2‐Chlorophenyl)piperazinyl]ethyl]‐1,3‐dimethylxanthine (KMUP‐1) is a xanthine‐based derivative. It has soluble GC activation and K+‐channel opening activity. Effects of this compound on ion currents in pituitary GH3 cells were investigated in this study.

Experimental Approach

The aim of this study was to evaluate effects of KMUP‐1 on the amplitude and gating of voltage‐gated Na+ current (I Na) in pituitary GH3 cells and in HEKT293T cells expressing SCN5A. Both the amplitude of Ca2+‐activated K+ current and the activity of large‐conductance Ca2+‐activated K+ (BKCa) channels were also studied.

Key Results

KMUP‐1 depressed the transient and late components of I Na with different potencies. The IC50 values required for its inhibitory effect on transient and late I Na were 22.5 and 1.8 μM respectively. KMUP‐1 (3 μM) shifted the steady‐state inactivation of I Na to a hyperpolarized potential by −10 mV, despite inability to alter the recovery of I Na from inactivation. In cell‐attached configuration, KMUP‐1 applied to bath increased BKCa‐channel activity; however, in inside‐out patches, this compound applied to the intracellular surface had no effect on it. It prolonged the latency in the generation of action currents elicited by triangular voltage ramps. Additionally, KMUP‐1 decreased the peak I Na with a concomitant increase of current inactivation in HEKT293T cells expressing SCN5A.

Conclusions and Implications

Apart from activating BKCa channels, KMUP‐1 preferentially suppresses late I Na. The effects of KUMP‐1 on ion currents presented here constitute an underlying ionic mechanism of its actions.

Abbreviations

AC
action current
AP
action potential
BKCa
channel large‐conductance Ca2+‐activated K+ channel
IK(Ca)
Ca2+‐activated K+ current
INa
voltage‐gated Na+ current
I–V
current versus voltage
KATP
channel ATP‐sensitive K+ channel
ODQ
1H‐[1,2,4]oxadiazolo‐[4,3‐a] quinoxalin‐1‐one
TEA
tetraethylammonium chloride
τinact(S)
slow component of inactivation time constant for I Na
YC‐1
3‐(5′‐hydroxymethyl‐2′‐furyl)‐1‐benzylindazole
  相似文献   

设为首页 | 免责声明 | 关于勤云 | 加入收藏

Copyright©北京勤云科技发展有限公司  京ICP备09084417号