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.
The Concise Guide to PHARMACOLOGY 2015/16 provides concise overviews of the key properties of over 1750 human drug targets with their pharmacology, plus links to an open access knowledgebase of drug targets and their ligands (www.guidetopharmacology.org), which provides more detailed views of target and ligand properties. The full contents can be found at http://onlinelibrary.wiley.com/doi/10.1111/bph.13351/full. Other ion channels are one of the eight major pharmacological targets into which the Guide is divided, with the others being: G protein‐coupled receptors, ligand‐gated ion channels, voltage‐gated ion channels, nuclear hormone receptors, catalytic receptors, enzymes and transporters. These are presented with nomenclature guidance and summary information on the best available pharmacological tools, alongside key references and suggestions for further reading. The Concise Guide is published in landscape format in order to facilitate comparison of related targets. It is a condensed version of material contemporary to late 2015, which is presented in greater detail and constantly updated on the website www.guidetopharmacology.org, superseding data presented in the previous Guides to Receptors & Channels and the Concise Guide to PHARMACOLOGY 2013/14. It is produced in conjunction with NC‐IUPHAR and provides the official IUPHAR classification and nomenclature for human drug targets, where appropriate. It consolidates information previously curated and displayed separately in IUPHAR‐DB and GRAC and provides a permanent, citable, point‐in‐time record that will survive database updates.

Conflict of interest

The authors state that there are no conflicts of interest to declare.

Family structure

5943 Aquaporins 5944 Chloride channels 5944 ClC family 5947 CFTR 5948 Calcium activated chloride channel 5949 Maxi chloride channel 5950 Volume regulated chloride channels 5952 Connexins and Pannexins 5954 Sodium leak channel, non‐selective  相似文献   

2.
Class A1 scavenger receptors (SRA1) are membrane glycoproteins that can form homotrimers. This receptor was originally defined by its ability to mediate the accumulation of lipids in macrophages. Subsequent studies reveal that SRA1 plays critical roles in innate immunity, cell apoptosis and proliferation. This review highlights recent advances in understanding the structure, receptor pathway and regulation of SRA1. Although its role in atherosclerosis is disputable, recent discoveries suggest that SRA1 function in anti‐inflammatory responses by promoting an M2 macrophage phenotype in cardiovascular diseases. Therefore, SRA1 may be a potential target for therapeutic intervention of cardiovascular diseases.

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

acLDL
acetylated low‐density lipoprotein
ER
endoplasmic reticulum
I/R
ischaemia/reperfusion
LTA
lipoteichoic acid
MI
myocardial infarction
mLDL
modified low‐density lipoprotein
oxLDL
oxidized low‐density lipoprotein
PRR
pattern recognition receptor
RAGE
receptor for advanced glycated end‐products
SR‐A1
class A1 scavenger receptor
TLR4
Toll‐like receptor 4
TRAF6
TNF receptor‐associated factor 6
Tables of Links
TARGETS
Catalytic receptors a
Mer receptor tyrosine kinase
TLR4
Enzymes b
Caspase 3
ERK
JNK
Mitogen‐activated protein kinase kinase 7 (MKK7)
p38
PI3K
PKC
PLC‐γ1
Open in a separate window
LIGANDS
Amyloid β
IFN‐γ
IL‐1
IL‐10
LPS
Lysophosphatidylcholine
Macrophage colony‐stimulating factor (M‐CSF)
MMP‐9
Phorbol ester (PMA)
Phosphatidylserine
TGF‐β1
TNF‐α
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). Scavenger receptors are cell surface receptors that are structurally diverse but they typically recognize many different ligands to participate in diverse biological functions. The functional mechanisms of scavenger receptors include endocytosis, phagocytosis, adhesion and signalling, which ultimately leads to the removal of non‐self‐ or altered self‐targets. There are 10 classes of scavenger receptors according to a unified nomenclature system for scavenger receptors (Prabhudas et al., 2014). Class A scavenger receptors have several structural features in common, including a cytoplasmic tail, a transmembrane domain, a spacer region, a helical coiled coil domain, a collagenous domain and a C‐terminal cysteine‐rich domain (Figure 1). Class A1 scavenger receptor (SR‐A1), also known as SCARA1, CD204 or macrophage scavenger receptor 1, is the prototypical SR‐A molecule and was the first scavenger receptor to be identified (Goldstein et al., 1979; Kodama et al., 1990; Rohrer et al., 1990)Open in a separate windowFigure 1Members of class A scavenger receptor family. The members of class A scavenger receptor family have a similar structure that is composed of a cytoplasmic tail, a transmembrane domain, a spacer region, a helical coiled coil domain, a collagenous domain and a C‐terminal cysteine‐rich domain.SR‐A1 was initially identified by its ability to mediate the formation of foam cells, a characteristic component of atherosclerotic lesions (Goldstein et al., 1979; Kodama et al., 1990; Krieger and Herz, 1994; Bowdish and Gordon, 2009). However, observations from various SR‐A1 gene knockout mouse models have yielded discrepant results concerning its role in the occurrence and development of atherosclerotic lesions (Suzuki et al., 1997; Kuchibhotla et al., 2008; Manning‐Tobin et al., 2009). A role beyond the handling of cholesterol is emerging for SR‐A1 in the pathogenesis of cardiovascular diseases. It not only functions as a phagocytic receptor and an innate immune recognition receptor but also plays an important role in cell apoptosis and cell proliferation. An overview of the recent progress of SR‐A1 structure, signal transduction and its roles in cardiovascular diseases will be provided in this review.  相似文献   

3.

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   

4.
The body is constantly faced with a dynamic requirement for blood flow. The heart is able to respond to these changing needs by adjusting cardiac output based on cues emitted by circulating catecholamine levels. Cardiac β‐adrenoceptors transduce the signal produced by catecholamine stimulation via Gs proteins to their downstream effectors to increase heart contractility. During heart failure, cardiac output is insufficient to meet the needs of the body; catecholamine levels are high and β‐adrenoceptors become hyperstimulated. The hyperstimulated β1‐adrenoceptors induce a cardiotoxic effect, which could be counteracted by the cardioprotective effect of β2‐adrenoceptor‐mediated Gi signalling. However, β2‐adrenoceptor‐Gi signalling negates the stimulatory effect of the Gs signalling on cardiomyocyte contraction and further exacerbates cardiodepression. Here, further to the localization of β1‐ and β2‐adrenoceptors and β2‐adrenoceptor‐mediated β‐arrestin signalling in cardiomyocytes, we discuss features of the dysregulation of β‐adrenoceptor subtype signalling in the failing heart, and conclude that Gi‐biased β2‐adrenoceptor signalling is a pathogenic pathway in heart failure that plays a crucial role in cardiac remodelling. In contrast, β2‐adrenoceptor‐Gs signalling increases cardiomyocyte contractility without causing cardiotoxicity. Finally, we discuss a novel therapeutic approach for heart failure using a Gs‐biased β2‐adrenoceptor agonist and a β1‐adrenoceptor antagonist in combination. This combination treatment normalizes the β‐adrenoceptor subtype signalling in the failing heart and produces therapeutic effects that outperform traditional heart failure therapies in animal models. The present review illustrates how the concept of biased signalling can be applied to increase our understanding of the pathophysiology of diseases and in the development of novel therapies.

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

ACEI
ACE inhibitors
CaMKII
Ca2+/calmodulin‐dependent kinase II
ct
carboxy terminus
EGFR
epidermal growth receptor
Epac
exchange protein directly activated by cAMP
Gi
inhibitory G protein
GRK
GPCR kinase
Gs
stimulatory G protein
HF
heart failure
PKA
cAMP‐dependent protein kinase
SNS
sympathetic nervous system
Tables of Links
TARGETS
GPCRs a Enzymes d
β1‐adrenoceptor Adenylyl cyclase (AC)
β2‐adrenoceptor Akt (PKB)
Angiotensin receptors CaMKII
Nuclear hormone receptors b Epac
Aldosterone receptor ERK
Catalytic receptors c GRK2
EGFR PKA
PI3K
Open in a separate window
LIGANDS
Carvedilol Fenoterol
Digoxin Metoprolol
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   

5.

Background and Purpose

Oestrogen inhibits cardiac hypertrophy and bone morphogenetic protein‐4 (BMP4) induces cardiac hypertrophy. Here we have studied the inhibition by oestrogen of BMP4 expression in cardiomyocytes.

Experimental Approach

Cultures of neonatal rat cardiomyocytes were used in in vitro experiments. Bilatαl ovariectomy (OVX) was carried out in female Kunming mice and cardiac hypertrophy was induced by transverse aortic constriction (TAC).

Key Results

Oestrogen inhibited BMP4‐induced cardiomyocyte hypertrophy and BMP4 expression in vitro. The inhibition of BMP4‐induced BMP4 protein expression by oestrogen was prevented by the inhibitor of oestrogen receptor‐β, PHTPP, but not by the inhibitor of oestrogen receptor‐α MPP. BMP4 induced smad1/5/8 activation, which was not affected by oestrogen in cardiomyocytes. BMP4 induced JNK but not ERK1/2 and p38 activation, and activated JNK was inhibited by oestrogen. Treatment with the p38 inhibitor SB203580 or the JNK inhibitor SP600125 inhibited BMP4‐induced BMP4 expression in cardiomyocytes, but the ERK1/2 inhibitor U0126 increased BMP4‐induced BMP4 expression, indicating that JNK, ERK1/2 and p38 MAPKs were all involved, although only JNK activation contributed to the inhibition of BMP4‐induced BMP4 expression by oestrogen. TAC induced significant heart hypertrophy in OVX mice in vivo and oestrogen replacement inhibited TAC‐induced heart hypertrophy in OVX mice. In parallel with the data of heart hypertrophy, oestrogen replacement significantly reduced the increased BMP4 protein expression in TAC‐treated OVX mice.

Conclusions and Implications

Oestrogen treatment inhibited BMP4‐induced BMP4 expression in cardiomyocytes through stimulating oestrogen receptor‐β and inhibiting JNK activation. Our results provide a novel mechanism underlying oestrogen‐mediated protection against cardiac hypertrophy.

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

BMP4
bone morphogenetic protein‐4
ER‐α
oestrogen receptor‐α
ER‐β
oestrogen receptor‐β
MPP
1,3‐bis(4‐hydroxyphenyl)‐4‐methyl‐5‐[4‐(2‐piperidinylethoxy)phenol]‐1H‐pyrazole dihydrochloride
OVX
ovariectomy
PHTPP
4‐(2‐phenyl‐5,7‐bis(trifluoromethyl)pyrazolo[1,5‐a]pyrimidin‐3‐yl)phenol
TAC
transverse aortic constriction
Tables of Links
TARGETS
Nuclear hormone receptors a
ER‐α, oestrogen receptor‐α
ER‐β, oestrogen receptor‐β
Enzymes b
ERK1/2
Furin
Histone deacetylase
JNK
p38
Ion channels c
Cav3.1 channels
Kv4.3 channels
Open in a separate window
LIGANDS
ANP, atrial natriuretic peptide
BNP, brain natriuretic peptide
MPP
PHTPP
SB203580
SP600125
β‐oestradiol
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, 2013c, 2010).  相似文献   

6.
Traumatic brain injury (TBI) has been linked to dementia and chronic neurodegeneration. Described initially in boxers and currently recognized across high contact sports, the association between repeated concussion (mild TBI) and progressive neuropsychiatric abnormalities has recently received widespread attention, and has been termed chronic traumatic encephalopathy. Less well appreciated are cognitive changes associated with neurodegeneration in the brain after isolated spinal cord injury. Also under‐recognized is the role of sustained neuroinflammation after brain or spinal cord trauma, even though this relationship has been known since the 1950s and is supported by more recent preclinical and clinical studies. These pathological mechanisms, manifested by extensive microglial and astroglial activation and appropriately termed chronic traumatic brain inflammation or chronic traumatic inflammatory encephalopathy, may be among the most important causes of post‐traumatic neurodegeneration in terms of prevalence. Importantly, emerging experimental work demonstrates that persistent neuroinflammation can cause progressive neurodegeneration that may be treatable even weeks after traumatic injury.

Linked Articles

This article is part of a themed section on Inflammation: maladies, models, mechanisms and molecules. To view the other articles in this section visit http://dx.doi.org/10.1111/bph.2016.173.issue-4

Abbreviations

AD
Alzheimer''s disease
CCI
controlled cortical impact
CTBI
chronic traumatic brain inflammation
CTE
chronic traumatic encephalopathy
CTIE
chronic traumatic inflammatory encephalopathy
DG
dentate gyrus
ERPs
event‐related potentials
SCI
spinal cord injury
TBI
traumatic brain injury
Tables of Links
TARGETS
Catalytic receptors a
IL‐4 receptor α
Enzymes b
Arginase 1
GPCR c
mGlu5 receptor
Open in a separate window
LIGANDS
Aβ, β‐amyloid IL‐1β
CCL2 TGFβ
CCL3 TNFα
CCL21
CHPG, 2‐chloro‐5‐hydroxyphenylglycine
Ibudilast
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   

7.
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   

8.

Background and Purpose

Cardiac fibrosis is a common feature of advanced coronary heart disease and is characteristic of heart disease. However, currently available drugs against cardiac fibrosis are still very limited. Here, we have assessed the role of isopropyl 3‐(3,4‐dihydroxyphenyl)‐2‐hydroxylpropanoate (IDHP), a new metabolite of Danshen Dripping Pills, in cardiac fibrosis mediated by the β‐adrenoceptor agonist, isoprenaline, and its underlying mechanisms.

Experimental Approach

Identification of IDHP was identified by mass spectrometry, and proton and carbon nuclear magnetic resonance spectra. Myocardial collagen was quantitatively assessed with Picrosirius Red staining. Expression of mRNA for collagen was evaluated with real‐time PCR. Phosphorylated and total p38 MAPK, NADPH oxidase (NOX) and superoxide dismutase (SOD) were analysed by Western blot. Generation of reactive oxygen species (ROS) generation was evaluated by dihydroethidium (DHE) fluorescent staining. NOX2 was knocked down using specific siRNA.

Key Results

IDHP attenuated β‐adrenoceptor mediated cardiac fibrosis in vivo and inhibited isoprenaline‐induced proliferation of neonatal rat cardiac fibroblasts (NRCFs) and collagen I synthesis in vitro. Phosphorylation of p38 MAPK, which is an important mediator in the pathogenesis of isoprenaline‐induced cardiac fibrosis, was inhibited by IDHP. This inhibition of phospho‐p38 by IDHP was dependent on decreased generation of ROS. These effects of IDHP were abolished in NRCFs treated with siRNA for NOX2.

Conclusions and Implications

IDHP attenuated the cardiac fibrosis induced by isoprenaline through a NOX2/ROS/p38 pathway. These novel findings suggest that IDHP is a potential pharmacological candidate for the treatment of cardiac fibrosis, induced by β‐adrenoceptor agonists.

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

CCK‐8
Cell Counting Kit‐8
DHE
dihydroethidium
ECM
extracellular matrix
IDHP
isopropyl 3‐(3,4‐dihydroxyphenyl)‐2‐hydroxylpropanoate
NAC
N‐acetylcysteine
NOX
NADPH oxidase
NRCFs
neonatal rat cardiac fibroblasts
ROS
reactive oxygen species
SOD
superoxide dismutase
Tables of Links
TARGETS
GPCRs a
β2‐adrenoceptors
Enzymes b
p38
Open in a separate window
LIGANDS
Collagen
Isoprenaline
SB202190
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 Guideto PHARMACOLOGY 2013/14 (a,bAlexander et al., 2013a, [Link]).  相似文献   

9.

Background and Purpose

There is emerging evidence that the mineralocorticoid hormone aldosterone is associated with arrhythmias in cardiovascular disease. However, the effect of aldosterone on the slowly activated delayed rectifier potassium current (IK s) remains poorly understood. The present study was designed to investigate the modulation of IK s by aldosterone.

Experimental Approach

Adult guinea pigs were treated with aldosterone for 28 days via osmotic pumps. Standard glass microelectrode recordings and whole‐cell patch‐clamp techniques were used to record action potentials in papillary muscles and IK s in ventricular cardiomyocytes.

Key Results

The aldosterone‐treated animals exhibited a prolongation of the QT interval and action potential duration with a higher incidence of early afterdepolarizations. Patch‐clamp recordings showed a significant down‐regulation of IK s density in the ventricular myocytes of these treated animals. These aldosterone‐induced electrophysiological changes were fully prevented by a combined treatment with spironolactone, a mineralocorticoid receptor (MR) antagonist. In addition, in in vitro cultured ventricular cardiomyocytes, treatment with aldosterone (sustained exposure for 24 h) decreased the IK s density in a concentration‐dependent manner. Furthermore, a significant corresponding reduction in the mRNA/protein expression of IKs channel pore and auxiliary subunits, KCNQ1 and KCNE1 was detected in ventricular tissue from the aldosterone‐treated animals.

Conclusions and Implications

Aldosterone down‐regulates IK s by inhibiting the expression of KCNQ1 and KCNE1, thus delaying the ventricular repolarization. These results provide new insights into the mechanism underlying K + channel remodelling in heart disease and may explain the highly beneficial effects of MR antagonists in HF.

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

APD
action potential duration
APD90
APD at 90% of repolarization
EAD
early afterdepolarization
ERG
ether‐a‐go‐go related gene
HF
heart failure
HSP90
heat shock protein 90
IK
delayed rectifier K+ current
IKr
rapid component of IK
IKs
slow component of IK
MR
mineralocorticoid receptor
Nim
nimodipine
RAAS
renin‐angiotensin‐aldosterone system
Tables of Links
TARGETS
Ion channels a Nuclear hormone receptors b
Cav1.2 Kir2.3 MR
Cav3.1 Kv1.5 Transporters c
ERG Kv4.2 Na+/K+‐ATPase
KCNQ1 Kv4.3
Kir2.1 Nav1.5
Open in a separate window
LIGANDS
Aldosterone E4031
Chromanol Ranolazine
Dofetilide Spirolactone
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   

10.

Background and Purpose

Cholecystokinin (CCK) is secreted by intestinal I cells and regulates important metabolic functions. In pancreatic islets, CCK controls beta cell functions primarily through CCK1 receptors, but the signalling pathways downstream of these receptors in pancreatic beta cells are not well defined.

Experimental Approach

Apoptosis in pancreatic beta cell apoptosis was evaluated using Hoechst‐33342 staining, TUNEL assays and Annexin‐V‐FITC/PI staining. Insulin secretion and second messenger production were monitored using ELISAs. Protein and phospho‐protein levels were determined by Western blotting. A glucose tolerance test was carried out to examine the functions of CCK‐8s in streptozotocin‐induced diabetic mice.

Key Results

The sulfated carboxy‐terminal octapeptide CCK26‐33 amide (CCK‐8s) activated CCK1 receptors and induced accumulation of both IP3 and cAMP. Whereas Gq‐PLC‐IP3 signalling was required for the CCK‐8s‐induced insulin secretion under low‐glucose conditions, Gs‐PKA/Epac signalling contributed more strongly to the CCK‐8s‐mediated insulin secretion in high‐glucose conditions. CCK‐8s also promoted formation of the CCK1 receptor/β‐arrestin‐1 complex in pancreatic beta cells. Using β‐arrestin‐1 knockout mice, we demonstrated that β‐arrestin‐1 is a key mediator of both CCK‐8s‐mediated insulin secretion and of its the protective effect against apoptosis in pancreatic beta cells. The anti‐apoptotic effects of β‐arrestin‐1 occurred through cytoplasmic late‐phase ERK activation, which activates the 90‐kDa ribosomal S6 kinase‐phospho–Bcl‐2‐family protein pathway.

Conclusions and Implications

Knowledge of different CCK1 receptor‐activated downstream signalling pathways in the regulation of distinct functions of pancreatic beta cells could be used to identify biased CCK1 receptor ligands for the development of new anti‐diabetic drugs.

Abbreviations

CCK
cholecystokinin
CCK‐8s
sulfated cholecystokinin fragment 26‐33 amide
p90RSK
90‐kDa ribosomal S6 kinase
siRNA
small interfering RNA
STZ
streptozotocin
Tables of Links
TARGETS
Bad
Bcl2
GPCRs a
CCK1 receptor
Enzymes b
Epac
ERK
MEK
p90RSK, 90‐kDa ribosomal S6 kinase
PKA
PLC
Open in a separate window
LIGANDS
Carbamylcholine
CCK, cholecystokinin
CCK‐8s
GLP‐1, glucagon‐like peptide‐1
H‐89
Insulin
IP3
Lorglumide
Rp‐cAMPS
U0126
U73122
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   

11.
12.

Background and Purpose

Disruptions of executive function, including attentional deficits, are a hallmark of a number of diseases. ACh in the prefrontal cortex regulates attentive behaviour; however, the role of α7 nicotinic ACh receptor (α7nAChR) in attention is contentious.

Experimental Approach

In order to probe attention, we trained both wild‐type and α7nAChR knockout mice on a touch screen‐based five‐choice serial reaction time task (5‐CSRT). Following training procedures, we then tested sustained attention using a probe trial experiment. To further differentiate the role of specific nicotinic receptors in attention, we then tested the effects of both α7nAChR and β2nAChR agonists on the performance of both wild‐type and knockout mice on the 5‐CSRT task.

Key Results

At low doses, α7nAChR agonists improved attentional performance of wild‐type mice, while high doses had deleterious effects on attention. α7nAChR knockout mice displayed deficits in sustained attention that were not ameliorated by α7nAChR agonists. However, these deficits were completely reversed by the administration of a β2nAChR agonist. Furthermore, administration of a β2nAChR agonist in α7nAChR knockout mice elicited similar biochemical response in the prefrontal cortex as the administration of α7nAChR agonists in wild‐type mice.

Conclusions and Implications

Our experiments reveal an intricate relationship between distinct nicotinic receptors to regulate attentional performance and provide the basis for targeting β2nAChRs pharmacologically to decrease attentional deficits due to a dysfunction in α7nAChRs.

Abbreviations

5‐CSRT
five‐choice serial reaction time task
VAChT
vesicular ACh transporter
Tables of Links
TARGETS
Ligand‐gated ion channels a Transporters b Enzymes c
α7nAChR (CHRNA7) VAChT AChE
β2nAChR ChAT
CHRNA2 ERK1
CHRNA4 ERK2
Open in a separate window
LIGANDS
Enzymes c
ACh
Methyllycaconitine
Nicotine
PHA‐543,613
Open in a separate window These 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   

13.

Background and Purpose

Although aspirin (acetylsalicylic acid) is commonly used to prevent ischaemic events in patients with coronary artery disease, many patients fail to respond to aspirin treatment. Dietary fish oil (FO), containing ω3 polyunsaturated fatty acids (PUFAs), has anti‐inflammatory and cardio‐protective properties, such as lowering cholesterol and modulating platelet activity. The objective of the present study was to investigate the potential additional effects of aspirin and FO on platelet activity and vascular response to injury.

Experimental Approach

Femoral arterial remodelling was induced by wire injury in mice. Platelet aggregation, and photochemical‐ and ferric chloride‐induced carotid artery thrombosis were employed to evaluate platelet function.

Key Results

FO treatment increased membrane ω3 PUFA incorporation, lowered plasma triglyceride and cholesterol levels, and reduced systolic BP in mice. FO or aspirin alone inhibited platelet aggregation; however, when combined, they exhibited synergistic suppression of platelet activity in mice, independent of COX‐1 inhibition. FO alone, but not aspirin, attenuated arterial neointimal growth in response to injury. Strikingly, a combination of FO and aspirin synergistically inhibited injury‐induced neointimal hyperplasia and reduced perivascular inflammatory reactions. Moreover, co‐administration of FO and aspirin decreased the expression of pro‐inflammatory cytokines and adhesion molecules in inflammatory cells. Consistently, a pro‐resolution lipid mediator‐Resolvin E1, was significantly elevated in plasma in FO/aspirin‐treated mice.

Conclusions and Implications

Co‐administration of FO and low‐dose aspirin may act synergistically to protect against thrombosis and injury‐induced vascular remodelling in mice. Our results support further investigation of adjuvant FO supplementation for patients with stable coronary artery disease.

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

AA
arachidonic acid
CO
coconut oil
CVD
cardiovascular disease
DHA
docosahexaenoic acid
EPA
eicosapentaenoic acid
FO
fish oil
HDLC
high‐density lipoprotein cholesterol
LDLC
low‐density lipoprotein cholesterol
PCI
percutaneous coronary intervention
PUFA
polyunsaturated fatty acid; TG, triglyceride
Tables of Links
TARGETS
Catalytic receptors a Enzymes b
CD11b (integrin, alpha M subunit) COX‐1
LFA‐1 (integrin, beta 2 subunit)
PDGFRβ
VLA‐4 (integrin α4β1)
Open in a separate window
LIGANDS
α‐linolenic acid IL‐6
ADP IL‐10
Arachidonic acid (AA) Linoleic acid
Aspirin PDGF
Clopidogrel Resolvin E1
Docosahexaenoic acid (DHA) Rose bengal
Eicosapentaenoic acid (EPA) TNF‐α
ICAM‐1 TxA2
VCAM‐1
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

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   

15.

Background and Purpose

Pressure overload‐induced cardiac interstitial fibrosis is viewed as a major cause of heart failure in patients with hypertension or aorta atherosclerosis. The purpose of this study was to investigate the effects and the underlying mechanisms of genistein, a natural phytoestrogen found in soy bean extract, on pressure overload‐induced cardiac fibrosis.

Experimental Approach

Genisten was administered to mice with pressure overload induced by transverse aortic constriction. Eight weeks later, its effects on cardiac dysfunction, hypertrophy and fibrosis were determined. Its effects on proliferation, collagen production and myofibroblast transformation of cardiac fibroblasts (CFs) and the signalling pathways were also assessed in vitro.

Key Results

Pressure overload‐induced cardiac dysfunction, hypertrophy and fibrosis were markedly attenuated by genistein. In cultured CFs, genistein inhibited TGFβ1‐induced proliferation, collagen production and myofibroblast transformation. Genistein suppressed TGFβ‐activated kinase 1 (TAK1) expression and produced anti‐fibrotic effects by blocking the TAK1/MKK4/JNK pathway. Further analysis indicated that it up‐regulated oestrogen‐dependent expression of metastasis‐associated gene 3 (MTA3), which was found to be a negative regulator of TAK1. Silencing MTA3 by siRNA, or inhibiting the activity of the MTA3‐NuRD complex with trichostatin A, abolished genistein''s anti‐fibrotic effects.

Conclusions and Implications

Genistein improved cardiac function and inhibited cardiac fibrosis in response to pressure overload. The underlying mechanism may involve regulation of the MTA3/TAK1/MKK4/JNK signalling pathway. Genistein may have potential as a novel agent for prevention and therapy of cardiac disorders associated with fibrosis.

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

CFs
cardiac fibroblasts
CTGF
connective tissue growth factor
ECM
extracellular matrix
EdU
5‐ethynyl‐2′‐deoxyuridine
EF
ejection fraction
ER
oestrogen receptors
FS
fractional shortening
MTA3
metastasis‐associated gene 3
NuRD
nucleosome remodelling and deacetylase
TAC
transverse aortic constriction
TAK1
TGFβ‐activated kinase 1
TSA
trichostatin A
Tables of Links
TARGETS
Nuclear hormone receptors a Enzymes b
Oestrogen receptor: ERα Endothelial (e) NOS
Oestrogen receptor: ERβ Histone deacetylase (HDAC)
Other protein targets Inducible (i) NOS
Kelch‐like ECH‐associated protein 1 (Keap1) JNK
P38‐MAPK
Smad2/3
TAK1
Open in a separate window
LIGANDS
Aliskiren ICI182780
Col1a1 Isoprenaline
Col3a1 TGFβ1
Curcumin Trichostatin A (TSA)
Genistein Wnt4
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   

16.

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   

17.

Background and Purpose

Cryptotanshinone (CTS) is a major bioactive diterpenoid isolated from Danshen, an eminent medicinal herb that is used to treat cardiovascular disorders in Asian medicine. However, it is not known whether CTS can prevent experimental atherosclerosis. The present study was designed to investigate the protective effects of CTS on atherosclerosis and its molecular mechanisms of action.

Experimental Approach

Apolipoprotein E‐deficient (ApoE −/−) mice, fed an atherogenic diet, were dosed daily with CTS (15, 45 mg kg−1 day−1) by oral gavage. In vitro studies were carried out in oxidized LDL (oxLDL)‐stimulated HUVECs treated with or without CTS.

Key Results

CTS significantly attenuated atherosclerotic plaque formation and enhanced plaque stability in ApoE −/− mice by inhibiting the expression of lectin‐like oxLDL receptor‐1 (LOX‐1) and MMP‐9, as well as inhibiting reactive oxygen species (ROS) generation and NF‐κB activation. CTS treatment significantly decreased the levels of serum pro‐inflammatory mediators without altering the serum lipid profile. In vitro, CTS decreased oxLDL‐induced LOX‐1 mRNA and protein expression and, thereby, inhibited LOX‐1‐mediated adhesion of monocytes to HUVECs, by reducing the expression of adhesion molecules (intracellular adhesion molecule 1 and vascular cellular adhesion molecule 1). Furthermore, CTS inhibited NADPH oxidase subunit 4 (NOX4)‐mediated ROS generation and consequent activation of NF‐κB in HUVECs.

Conclusions and Implications

CTS was shown to have anti‐atherosclerotic activity, which was mediated through inhibition of the LOX‐1‐mediated signalling pathway. This suggests that CTS is a vasculoprotective drug that has potential therapeutic value for the clinical treatment of atherosclerotic cardiovascular diseases.

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

ApoE−/−
apolipoprotein E deficient
CD36
cluster of differentiation 36
CTS
cryptotanshinone
HCD
high cholesterol diet
ICAM‐1
intracellular adhesion molecule 1
LOX‐1
lectin‐like oxidized low‐density lipoprotein receptor‐1
NOX4
NADPH oxidase subunit 4
oxLDL
oxidized low‐density lipoprotein
ROS
reactive oxygen species
SMCs
smooth muscle cells
SR‐A
scavenger receptor‐A
VCAM‐1
vascular cellular adhesion molecule 1
Tables of Links
TARGETS
Nuclear hormone receptors a
PPARγ
Enzymes b
MMP‐9
Open in a separate window
LIGANDS
Angiotensin II IFN‐γ IL‐17A
Homocysteine IL‐1β TNF‐α
ICAM‐1 IL‐6 VCAM‐1
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   

18.

Background and Purpose

High‐salt diet induces cardiac remodelling and leads to heart failure, which is closely related to cardiac mitochondrial dysfunction. Transient receptor potential (TRP) channels are implicated in the pathogenesis of cardiac dysfunction. We investigated whether activation of TRP vanilloid (subtype 1) (TRPV1) channels by dietary capsaicin can, by ameliorating cardiac mitochondrial dysfunction, prevent high‐salt diet‐induced cardiac hypertrophy.

Experimental Approach

Male wild‐type (WT) and TRPV1 −/− mice were fed a normal or high‐salt diet with or without capsaicin for 6 months. Their cardiac parameters and endurance capacity were assessed. Mitochondrial respiration and oxygen consumption were measured using high‐resolution respirometry. The expression levels of TRPV1, sirtuin 3 and NDUFA9 were detected in cardiac cells and tissues.

Key Results

Chronic high‐salt diet caused cardiac hypertrophy and reduced physical activity in mice; both effects were ameliorated by capsaicin intake in WT but not in TRPV1 −/− mice. TRPV1 knockout or high‐salt diet significantly jeopardized the proficiency of mitochondrial Complex I oxidative phosphorylation (OXPHOS) and reduced Complex I enzyme activity. Chronic dietary capsaicin increased cardiac mitochondrial sirtuin 3 expression, the proficiency of Complex I OXPHOS, ATP production and Complex I enzyme activity in a TRPV1‐dependent manner.

Conclusions and Implications

TRPV1 activation by dietary capsaicin can antagonize high‐salt diet‐mediated cardiac lesions by ameliorating its deleterious effect on the proficiency of Complex I OXPHOS. TRPV1‐mediated amendment of mitochondrial dysfunction may represent a novel target for management of early cardiac dysfunction.

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

AngII
angiotensin II
CI
complex I
CII
complex II
CS
citrate synthase
EF
ejection fraction
ETC
electronic transport chain
FS
shortening fraction
HS
high‐salt diet
iRTX
5′‐iodoresiniferatoxin
LVH
left ventricular hypertrophy
LVID
left ventricular internal diameter
LVPW
left ventricular posterior wall
ND
normal‐salt diet
OXPHOS
oxidative phosphorylation
RER
respiration exchange ratio
RES
resveratrol
ROS
reactive oxygen species
TRPCs
transient potential receptor channels
TRPV1
transient receptor potential vanilloid subfamily member 1
TRPV1−/−
TRPV1 knockout
WT
wild‐type
Tables of Links
TARGETS
Ion channels a Enzymes b
TRPC3 Endothelial NOS
TRPC6 ERK1/2
TRPV1 MEK
VDAC PKA
Sirtuin 3
Open in a separate window
LIGANDS
5′‐iodoresiniferatoxin Capsaicin
Angiotensin II Hydrogen peroxide
ATP NADH
Baicalein Resveratrol
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   

19.
Hydrogen sulfide (H2S) has traditionally been viewed as a highly toxic gas; however, recent studies have implicated H2S as a third member of the gasotransmitter family, exhibiting properties similar to NO and carbon monoxide. Accumulating evidence has suggested that H2S influences a wide range of physiological and pathological processes, among which blood vessel relaxation, cardioprotection and atherosclerosis have been particularly studied. In the cardiovascular system, H2S production is predominantly catalyzed by cystathionine γ‐lyase (CSE). Decreased endogenous H2S levels have been found in hypertensive patients and animals, and CSE −/− mice develop hypertension with age, suggesting that a deficiency in H2S contributes importantly to BP regulation. H2S supplementation attenuates hypertension in different hypertensive animal models. The mechanism by which H2S was originally proposed to attenuate hypertension was by virtue of its action on vascular tone, which may be related to effects on different ion channels. Both H2S and NO cause vasodilatation and there is cross‐talk between these two molecules to regulate BP. Suppression of oxidative stress may also contribute to antihypertensive effects of H2S. This review also summarizes the state of research on H2S and hypertension in China. A better understanding of the role of H2S in hypertension and related cardiovascular diseases will allow novel strategies to be devised for their treatment.

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

Abbreviation

2K1C
two‐kidney‐one‐clip
Ang II
angiotensin II
AOA
aminooxyacetic acid
AT1 receptor
angiotensin II type 1 receptor
CBS
cystathionine‐β‐synthase
CO
carbon monoxide
CSE
cystathionine‐γ‐lyase
DBP
diastolic BP
eNOS
endothelial NOS
H2S
hydrogen sulfide
I/R
ischaemia/reperfusion
L‐NAME
NG‐nitro‐l‐arginine methyl ester
MAP
mean arterial pressure
MPST
3‐mercaptopyruvate sulfurtransferase
MWT
medial wall thickness
PAAT
pulmonary arterial acceleration time
PAG
DL‐propargylglycine
PHT
pulmonary hypertension
RVET
right ventricular ejection time
RVH
right ventricular hypertrophy
SBP
systolic BP
SHR
spontaneously hypertensive rat
SMCs
smooth muscle cells
VEGFR‐1
soluble fms‐like tyrosine kinase 1
VD
vas deferens
Tables of Links
TARGETS
GPCR a Catalytic receptors d
AT1 receptor VEGFR‐1
Muscarinic receptors Enzymes e
Thromboxane A2 receptor CBS
Ligand‐gated ion channels b CSE
Epithelial sodium channels (ENaC) eNOS
Ion channels c ERK1/2
BKCa channels HO1
CaV channels p38MAPK
CaV1.1‐1.4 (L‐type Ca) channels PKG
KATP channels PTEN
Kir channels MPST
KV channels
KV7.x (KCNQ) channels
Open in a separate window
LIGANDS
ACh L‐NAME
Angiotensin II NaHS
AOA Nifedipine
Carbachol Nitric oxide (NO)
Glibenclamide Noradrenaline
Homocysteine Phenylephrine
Iberiotoxin Tetraethylammonium
L‐arginine U46619
L‐cysteine XE991
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.
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   

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