Enhanced levels of TGFβ are found in patients with heart failure Khan et al., 2014, in various animal models of cardiac remodelling and during the transition from compensated hypertrophy
Trang 2Reviews Editor
Stephen Alexander Nottingham, UK Annette Gilchrist Senior Online EditorDowners Grove, USA
Senior Editors
Editorial Board
Amrita Ahluwalia London, UK
Michael J Curtis London, UK
James Docherty Dublin, Ireland
Mark Giembycz Calgary, Canada
Daniel Hoyer Melbourne, Australia
Paul Insel La Jolla, USA
Angelo A Izzo Naples, Italy
David MacEwan Liverpool, UK
Clare Stanford London, UK
Susan Wonnacott Bath, UK
Ruth Andrew Edinburgh, UK
Alexis Bailey Guildford, UK
Chris Bailey Bath, UK
Phillip Beart Melbourne, Australia
Tamás Bíró Budapest, Hungary
Tom Blackburn Leigh on Sea, UK
Heather Bradshaw Bloomington, USA
Keith Brain Birmingham, UK
James Alexander Brock Melbourne, Australia
Gillian Burgess Slough, UK
John Challiss Leicester, UK
Diana Chow Houston, USA
Macdonald Christie Sydney, Australia
Sandy Clanachan Edmonton, Canada
John Cryan Cork, Ireland
Anthony Davenport Cambridge, UK
Martin Diener Giessen, Germany
Peter Doris Houston, USA
Pedro D’Orléans-Juste Sherbrooke, Canada
Grant Drummond Clayton, Australia
Claire Edwards Oxford, UK
Michael Emerson London, UK
Liana Fattore Cagliari, Italy
Peter Ferdinandy Szeged, Hungary
Anthony Ford San Mateo, USA
Chris George Cardiff, UK
Jon Gibbons Reading, UK
Gary Gintant Illinois, USA
Michelle Glass Auckland, New Zealand
Jules Hancox Bristol, UK
Deborah L Hay Auckland, New Zealand
Jackie Hunter Weston, UK
Ryuji Inoue Fukuoka, Japan
Yong Ji Nanjing, China
Marcel Jiménez Barcelona, Spain
Eamonn Kelly Bristol, UK
Melanie Kelly Halifax, Canada
Terry Kenakin Durham, USA
Dave Kendall Nottingham, UK
Charles Kennedy Glasgow, UK
Simon Kennedy Glasgow, UK
Chris Langmead Welwyn Garden City, UK
Andy Lawrence Melbourne, Australia
Eliot Lilley Redhill, UK
Jon Lundberg Stockholm, Sweden
Mhairi Macrae Glasgow, UK
Karen McCloskey Belfast, UK
Barbara McDermott Belfast, UK
Alister McNeish Reading, UK
Jo De Mey Odense, Denmark
Olivier Micheau Dijon, France
Paula Moreira Coimbra, Portugal
Maria Moro Madrid, Spain
Fiona Murray San Diego, USA
Anne Negre-Salvayre Toulouse, France
Janet Nicholson Biberach an der Riss, Germany
Eliot Ohlstein Pennsylvania, USA
Saoirse O’Sullivan Nottingham, UK
Hiroshi Ozaki Tokyo, Japan
Reynold Panettieri Jr Philadelphia, USA
Andreas Papapetropoulos Athens, Greece
Clare Parish Melbourne, Australia
Adam Pawson Edinburgh, UK
Roger Phillips Bradford, UK
Michael Pugsley Jersey City, USA
Susan Pyne Strathclyde, UK
Jelena Radulovic Chicago, USA
Chris Sobey Monash, Australia
Michael Spedding Suresnes, France
Beata Sperlagh Budapest, Hungary
Shiva Sruti Pittsburgh, USA
Katarzyna Starowicz Krakow, Poland
Barbara Stefanska Quebec, Canada
Gary Stephens Reading, UK
Csaba Szabo Budapest, Hungary
Kenneth Takeda Strasbourg, France
Paolo Tammaro Oxford, UK
Anna Teti L’Aquila, Italy
Ekaterini Tiligada Athens, Greece
Jean-Pierre Valentin Macclesfield, UK
Paul Vanhoutte Hong Kong, China
Christopher Vaughan Sydney, Australia
Harald Wajant Würzburg, Germany
Julia Walker Durham, USA
Xin Wang Manchester, UK
Nina Weber Amsterdam, the Netherlands
James Whiteford London, UK
Baofeng Yang Heilongjiang, China
The British Journal of Pharmacology is a broad-based journal giving leading
international coverage of all aspects of experimental pharmacology The
Edito-rial Board represents a wide range of expertise and ensures that well-presented
work is published as promptly as possible, consistent with maintaining the
overall quality of the journal
Disclaimer
The Publisher, British Pharmacological Society and Editors cannot be held
responsible for errors or any consequences arising from the use of information
contained in this journal; the views and opinions expressed do not necessarily
reflect those of the Publisher, British Pharmacological Society and Editors
Neither does the publication of advertisements constitute any endorsement by
the Publisher, British Pharmacological Society and Editors of the products
advertised
Copyright and Copying
Copyright © 2016 The British Pharmacological Society All rights reserved No part of this publication may be reproduced, stored or transmitted in any form or
by any means without the prior permission in writing from the copyright holder Authorization to copy items for internal and personal use is granted by the copyright holder for libraries and other users registered with their localReproduction Rights Organisation (RRO), e.g Copyright Clearance Center (CCC),
222 Rosewood Drive, Danvers, MA 01923, USA (www.copyright.com), providedthe appropriate fee is paid directly to the RRO This consent does not extend to other kinds of copying, such as copying for general distribution for advertising
or promotional purposes, for creating new collective works, or for resale.Special requests should be addressed to: permissions@wiley.com
Trang 3Molecular switches under
progression from cardiac
hypertrophy to heart failure
J Heger, R Schulz and G Euler
Institute of Physiology, Justus Liebig University, Giessen, Germany
Correspondence
Gerhild Euler, Institute of Physiology,Justus Liebig University, Giessen,Germany
E-mail: Gerhild.Euler@physiologie.med.uni-giessen.de
Commissioning Editor: PeterFerdinandy
we will describe examples of these molecular switches that change compensated hypertrophy to the development of heart failureand will focus on the importance of the signalling cascades of the TGFβ superfamily in this process In this context, potentialtherapeutic targets for pharmacological interventions that could attenuate the progression of heart failure will be discussed
Abbreviations
ALK, activin receptor-like kinase; AMPK, AMP kinase; ANT1, adenine nucleotide translocator 1; AP-1, activator protein 1;Hsp, heat shock protein; IGF2R, insulin-like growth factor receptor II; JDP2, jun dimerization protein 2; LNA, locked nucleicacid; LV, left ventricle; miRNA, microRNA; MPTP, mitochondrial permeability transition pore; NLRP3, nucleotide-bindingdomain and leucine-rich repeat containing PYD-3; PAH, pulmonary hypertension; RIP, receptor interacting protein; RV,right ventricle; siRNA, silencing RNA; SIRT1, sirtuin 1; SMAD, small mothers against decapentaplegic; TAC, transverse aorticconstriction; TAK1, TGFβ activated kinase 1; TGFBR1, TGFβ receptor I; TGFBR2, TGFβ receptor II; TOM, translocase of themitochondrial outer membrane; UPS, ubiquitin proteasome system; VDAC1, voltage-dependent anion channel-1
BJP British Journal of Pharmacology www.brjpharmacol.org
Trang 4The main causes of heart failure are on the one hand chronic
pressure overload of the left ventricle (LV) resulting in
hypertension and on the other the impairment of
myocar-dial perfusion resulting in acute myocarmyocar-dial infarction or
While pressure overload creates hypertension and results
in cardiac hypertrophy, myocardial infarction primarily
results in a loss of cardiomyocytes that is compensated
for by hypertrophy of the remaining cells, the generation
of fibrosis and ventricular dilatation Thus, the
remodel-ling processes, caused by pressure overload or ischaemia
are different, but both eventually result in heart failure
This has to be considered when using different animal
models, which are induced either by chronic pressure
overload by aortic banding or by direct damage after
myo-cardial infarction
In spite of these differences, in both situations, the
organism reacts by activation of the sympathetic nervous
system and the release of local mediators (cytokines and
natriuretic peptides) to ensure a sufficient blood supply
under these conditions, thereby resulting in intra- and
intercellular remodelling processes Temporarily, this leads
to compensated hypertrophy and preserved heart
func-tion However, in the long run, progressive myocardial
dysfunction develops (Narula et al., 1996), either resulting
in around 50% of the patients having an impaired
diastolic function with preserved ejection fraction, while
the other 50% of patients develop systolic dysfunction
with reduced ejection fraction (Abbate et al., 2015) With
regard to the reduced ejection fraction, apoptotic and
necroptotic loss of cardiomyocytes, contractile
dysfunc-tion of cardiomyocytes and massive fibrosis are significant
factors for the transition from compensated hypertrophy
to decompensation and deterioration of systolic heart
function, which will be the main focus of the current
review
Enhanced levels of TGFβ are found in patients with
heart failure (Khan et al., 2014), in various animal models
of cardiac remodelling and during the transition from
compensated hypertrophy to heart failure (Boluyt et al.,
1994; Lijnen et al., 2000; Rosenkranz, 2004) Therefore,
there is a huge drive to clarify the role of TGFβ in heart
failure progression Interestingly, TGFβ modulates nearly
all processes that are engaged in heart failure development,
that is, cardiac hypertrophy, fibrosis, apoptosis,
inflamma-tion and differentiainflamma-tion of cardiac progenitor cells In spite
of this, broad inhibition of TGFβ signalling does not only
have positive effects on heart failure progression
Adminis-tration of the TGFβ receptor I (TGFBR1) inhibitor (SM16)
after aortic banding prevented cardiac fibrosis and
attenu-ated cardiac dysfunction However, mortality rates increased
due to enhanced left ventricular dilatation and inflammation
(Engebretsen et al., 2014) Similar results have been found
when soluble TGFBR2 was applied after myocardial
in-farction In this case, the increase in mortality rates was
probably due to reduced inflammatory responses (Ikeuchi
et al., 2004) Therefore, a more target-orientated approach
pathways
TGFβ signals through binding at a heterotetrameric receptorcomplex of type II and type I receptor serine/threonine kinases.Upon TGFβ binding, TGFBR2 phosphorylates and therebyactivates type I receptor serine/threonine kinases that in turnphosphorylates and activates SMAD transcription factors De-pending on the subtype of type I receptor serine/threonine ki-nases, also known as activin receptors or activin receptor-likekinases (ALKs), different receptor-activated SMADs (R-SMADs)become activated In cardiomyocytes and also many other celltypes, TGFβ1 signalling is attributed to TGFBR1, also calledALK5, which then results in SMAD2/3 activation In endothe-lial cells TGFβ has also been shown to signal via ALK1 andSMAD1/5/8 (Goumans et al., 2002) However, this appears to
be no longer exclusive for endothelial cells; In other cells,TGFbeta1 stimulation has been found to activate SMAD1/5and SMAD2/3 as well (Wharton and Derynck, 2009) We alsoidentified both responses in cardiomyocytes After stimulation
of ventricular cardiomyocytes, from adult rats, with 1 ngml 1TGFβ1 for 2 h, enhanced phosphorylation of SMAD2/3,SMAD1/3 and SMAD1/5 was detected in Western blots (n = 5,
P < 0.05 vs unstimulated controls) (Figure 1), thereby ing that TGFβ signalling is even more complicated than origi-nally thought Activated R-SMADs form a complex withSMAD4 that translocates into the nucleus and acts as a tran-scription factor The binding specificity of SMADs to promoterscan be influenced by their association with other transcriptionfactors like activator protein 1 (AP-1) In addition to this canon-ical SMAD pathway, another prominent signalling molecule ofTGFβ is TGFβ-activated kinase 1 (TAK1) TAK1 activation is alsomediated by TGFBR2 Downstream targets of TAK1 are c-Jun, N-terminal kinase (JNK) and p38 Furthermore, via binding to its
phosphoinositide 3-kinase (PI3K) or small GTPases like Rho(reviewed by Zhang, 2009) This huge variety of TGFβ signallingpathways already implies that the effects of TGFβ in tissues will
be complex In this review, we highlight the signallingmolecules that are induced by TGFβ and modulate adverse car-diac remodelling by interfering with adrenoceptor-mediatedsignalling, mitochondrial proteins, cell death, microRNAs(miRNAs), contractile function or fibrosis (Figure 2)
The TAK1 pathway is pro-hypertrophic and prevents cell death while SMADs promote apoptotic signalling in the heart
TGFβ itself is known to be a pro-hypertrophic, pro-apoptoticand pro-fibrotic factor in the heart TAK1 and not SMADsseems to be the main mediator of TGFβ-induced hypertrophicgrowth effects TAK1 is found to be up-regulated in vivo afteraortic banding, and TAK1 overexpression promotes cardiachypertrophy in transgenic mice (Zhang et al., 2000) (Figure 3).Furthermore, in neonatal cardiomyocytes, angiotensin II-induced hypertrophic growth could be prevented by knock-down of TAK1 with silencing RNA (siRNA), but not withsiRNA against SMAD2/3 (Watkins et al., 2012) This indicatesthat SMAD signalling is not involved in angiotensin II –TGFβ1-induced hypertrophic growth In addition to its pro-hypertrophic effects, TAK1 antagonizes the apoptosis and
BJP J Heger et al.
4 British Journal of Pharmacology (2016)173 3–14
Trang 5necroptosis induced by TNFα stimulation and prevents
ad-verse cardiac remodelling (Li et al., 2014a) Necroptosis is a
form of cell death, which combines features of necrotic and
apoptotic cell death, it is a death receptor-mediated process
which is executed via receptor activating protein (RIP)
com-plexes (Zhang et al., 2009) During TNFα stimulation, TAK1
associates with RIP1 thereby preventing RIP1 interactionwith other death signalling proteins, that is, with caspase 8
or RIP3 This results in a reduction in apoptosis andnecroptosis (Li et al., 2014a) As TAK1 is not only induced byTGFβ and TNFα but also by other cytokines (Besse et al.,2007), strong TAK1 activation may act as a pro-survival factor
in the heart
In contrast to TAK1, SMAD signalling seems to
cardiomyocytes induced by stimulation ofα-adrenoceptorswas hampered by simultaneous overexpression of SMADs(Heger et al., 2009) Hypertrophic growth of cardiomyocytesinduced by stimulation of α-adrenoceptors is mediated viathe transcription factor AP-1 (Taimor et al., 2004) Under si-multaneous SMAD4 overexpression, AP-1/SMAD complexesare formed, which may detract AP-1 from its hypertrophy-promoting target genes Indeed, a shift from hypertrophy
to the induction of apoptosis is found in
(Heger et al., 2009) Furthermore, cardiac-specific SMAD4knock-out mice displayed cardiac hypertrophy (Wang et al.,2005) This indicates that SMAD4 acts as a molecular switchfor transition from hypertrophy to apoptosis In addition,TGFβ induces apoptosis in adult cardiomyocytes via en-hancement of SMAD and AP-1 activity (Schneiders et al.,2005) Similar to these findings, inhibition of SMAD signal-ling in vivo may preserve the compensating character of hy-pertrophic growth in cardiac remodelling while preventingthe transition to apoptosis (Figure 3)
That AP-1 is a mediator of hypertrophy and apoptosis inadrenoceptor stimulated cardiomyocytes has been shown byuse of transgenic mice overexpressing the AP-1 inhibitor jun
prevented isoprenaline (ISO)-induced hypertrophy as well
as TGFβ-induced apoptosis in cardiomyocytes (Hill et al.,2013) But AP-1 is also required to preserve the contractilefunction of cardiomyocytes because AP-1 inhibition byJDP2 overexpression attenuated contractile responses in-duced byβ-adrenoceptor stimulation (Hill et al., 2013) There-fore, to prevent adverse remodelling, inhibition of SMADsignalling seems to be the better choice than inhibition ofAP-1, because this would negatively influence the contractilefunction of the heart
Modulation of β-adrenoceptor responses in the presence of TGF β
During heart failure, progressive desensitization of adrenoceptors occurs β-adrenoceptors are members of theGPCR superfamily whose stimulation results in activation
β-of PKA via AC and cAMP, which regulate different lar, sarcolemmal and myofibrillar substrates Thus, cAMPexerts the cellular effects on cardiac contractile function in-duced by activation of β-adrenoceptors However, stimula-tion of β-adrenoceptors also results in agonist-dependentdesensitization of these receptors, a phenomenon foundduring the development of heart failure This process is me-diated by the receptor adapter proteinβ-arrestin that binds
intracellu-to β-adrenoceptors This binding either results in direct
Figure 2
Overview about TGFβ influence on components of cardiac
remodel-ling in left ventricular systolic heart failure TGFβ has been shown to
promote the transition from cardiac hypertrophy to apoptosis and
to regulate mitochondrial signalling molecules, miRNA expression
and contractile function andfibrosis All these processes are involved
in heart failure progression
Figure 1
TGFβ signals via the SMAD2/3 and SMAD1/5 pathway in
cardiomyocytes Ventricular cardiomyocytes of adult rat were
stimu-lated with 1 ngml 1TGFβ1 for 2 h Protein extracts of these cells were
analysed by Western blots with antibodies specific against
phosphoSMAD2, phosphoSMAD1/3 or phosphoSMAD1/5
Phosphor-ylation, which is indicative of SMAD activation, was detected for all
these SMADs *P< 0.05 versus unstimulated controls, n = 5
indepen-dent culture preparations
TGFβ-guided switches to heart failure
BJP
Trang 6inhibition ofβ-adrenoceptors, known as functional
desensi-tization, or in internalization of β-adrenoceptors that
re-duces their density (reviewed by Lymperopoulos and
Negussie, 2013) Studies inβ-arrestin1 knock-out mice
dem-onstrated a major role for β-arrestin1 in cardiac
stimulation were enhanced in these knock-out animals
β-arrestin1 prevented adverse cardiac remodelling after
myo-cardial infarction by inhibiting apoptosis and preserving
cardiac function (Bathgate-Siryk et al., 2014) Interestingly,
an increase in myocardial β-adrenoceptor density and a
β-adrenoceptor-kinase-1 were demonstrated in transgenic
TGFβ-overexpressing mice (Rosenkranz et al., 2002) And
in isolated cardiomyocytes of adult rat, TGFβ enhanced
the hypertrophic response to β-adrenoceptor stimulation
(Schlüter et al., 1995) These findings indicate that TGFβ
can preventβ-adrenoceptor desensitization in cardiomyocytes
and thereby promote pro-hypertrophic signalling Whether
this response is mediated by the down-regulation of
β-arrestin1 by TGFβ has not yet been clarified But TGFβ
may be a plausible target in order to preventβ-adrenoceptor
desensitization
So far, a connection between β-arrrestin expression
and TGFβ signalling has been shown in cardiac fibroblasts
β-Arrestins were found to be up-regulated in cardiac
fibroblasts during heart failure Overexpression ofβ-arrestin
in cardiac fibroblasts results in the uncoupling of adrenoceptors and activation of SMAD2/3, thereby promoting
β-a pro-fibrotic phenotype This mβ-ay cβ-ause enhβ-anced stiffness
of the ventricular wall and contribute to the development ofheart failure
Although TGFβ stimulation prevents the uncoupling ofβ-adrenoceptors and enhances the pro-hypertrophic signal-ling, the inotropic β-adrenoceptor-mediated response wasdiminished in TGFβ-overexpressing mice This is due to anup-regulation of mitochondrial uncoupling proteins duringβ-adrenoceptor stimulation, which results in a decreased mi-tochondrial energy production Thus, TGFβ-overexpressingmice resemble a phenotype occurring at the transition toheart failure, namely, displaying cardiomyocytes hypertro-phy and promoting apoptosis as well as mitochondrialand contractile dysfunction (Schneiders et al., 2005;Huntgeburth et al., 2011)
That these interacting pathways of ADRB-TGFβ signallingare even more complex was indicated by the findings thatGPCRs not only activate TK receptors but also alsotransactivate the serine/threonine kinase TGFBR1 in differentcell types (Burch et al., 2012) The proposed mechanism forthis transactivation is activation of integrin by GPCRs Subse-quently, integrin binding to the large latent TGFβ complexcauses a conformational change and allows TGFβ to bindand activate TGFBR2/TGFBR1, thereby resulting in SMAD
Figure 3
Influence of TGFβ-SMAD and TGFβ-TAK1 signalling on adrenoceptor-mediated pathways in LV heart failure progression Adrenoceptors (AR) ulate the expression of genes promoting hypertrophic growth via the transcription factor AP-1 Under simultaneous presence of SMAD4, the pro-hypertrophic response to adrenoceptor stimulation is shifted to a pro-apoptotic gene transcription via AP-1/SMAD complexes Also, under TGFβstimulation of cardiomyocytes, AP-1 and SMADs mediate apoptosis In addition to these effects on cardiomyocytes, activation of the TGFβ/SMADpathway or induction of SMADs viaβ-arrestins induces the transcription of fibrotic genes In contrast to the SMAD pathway, TAK1 activation stim-ulates hypertrophic growth while inhibiting cardiac necroptosis and apoptosis by interacting with RIP1 Strongβ-adrenoceptor (ADRB) activationresults inβ-adrenoceptor desensitization via β-adrenoceptor /β-arrestin complexes This process can be inhibited by TGFβ Depicted in red areswitch molecules that can modulate the response of the cell to receptor stimulation and thereby influence the outcome of this stimulation onthe remodelling process
stim-BJP J Heger et al.
6 British Journal of Pharmacology (2016)173 3–14
Trang 7activation (Munger et al., 1999) Whether this
cardiomyocytes has yet to be analysed
The ubiquitin system in the context of
β-adrenoceptor and TGFβ stimulation
Another focus for identification of the triggers contributing
to heart failure development or progression relies on the
anal-ysis of the proteasome, as degradation of proteins is changed
in cardiac hypertrophy The primary cellular response to
β-adrenoceptor stimulation in the heart is an increased pool
of 20S subunits with catalytic activity, while chronic
β-adrenoceptor stimulation enhanced the 26S proteasome but
decreased 20S proteasomal activity, accompanied by a
de-crease in ubiquitinated proteins (Drews et al., 2010) Elevated
26S proteasome activities were also observed in a pressure
overload model stimulating ventricular hypertrophy (Depre
et al., 2006) The switch in proteasome subpopulations,
which is facilitated by differentβ-subunits of the proteasome,
is decisive for the development of hypertrophy and depends
again on the strength ofβ-adrenoceptor activation Proteins
involved in cardiac hypertrophy are targeted by
muscle-specific ubiquitin ligase atrogin-1 for degradation (Zaglia
et al., 2014) Atrogin-1 KO hearts revealed increased apoptosis
and hypertrophy The effects were mediated by the
endosomal sorting complex required for transport III
(ESCRT-III) family protein charged multivesicular body
pro-tein 2B (CHMP2B) Thus, Zaglia et al (2014) demonstrated
the interplay between the ubiquitin proteasome system
(UPS) and autophagy and the importance of controlled
degra-dation of proteins for the control of cardiac hypertrophy and
apoptosis
UPS regulates important signalling pathways in the heart,
including MAPK, JNK and calcineurin (Portbury et al., 2012)
Huang et al (2014) suggested a proteasome-dependent
mech-anism for angiotensin II–induced apoptosis in hearts that is
accompanied by activation of insulin-like growth factor
re-ceptor II (IGF2R) signalling Heat shock transcription factor
1 (HSF1) acts as a repressor of IGF2R gene expression only if
deacetylated by sirtuin 1 However, angiotensin II and
subse-quently JNK activation mediates sirtuin 1 degradation via the
proteasome This results in an increase in the acetylation of
HSF1 that is then not able to bind to the IGF2R promoter
So, sirtuin is a negative regulator of IGF2R, thereby protecting
cardiomyocytes from apoptosis In this context, it is
remark-able that the IGF2R is required for the activation of latent
TGFβ (Dennis and Rifkin, 1991) In human umbilical-vein
endothelial cells, the association of IGF2R and the urokinase
receptor–converts plasminogen (uPAR) to active plasmin –
is essential for the activation of latent TGFβ, the release of
TGFβ and induction of apoptosis (Leksa et al., 2005) Whether
this also holds true for cardiomyocytes remains to be
evalu-ated, but we have already shown that angiotensin II induces
the release of TGFβ and SMAD-dependent apoptosis in
cardiomyocytes (Schröder et al., 2006) Not only is the
intra-cellular activity of TGFβ controlled by UPS but also the
stability and levels of TGFβ receptor complexes are mined by ubiquitination (Xu et al., 2012)
deter-In fluence of TGFβ on mitochondria, energy metabolism and heart failure
Mitochondria are the power houses of the cell, generating ATPvia oxidative phosphorylation On average, 30% of thecardiomyocytes volume is filled with mitochondria (Barth
et al., 1992) One side product of the major respiratory zyme complexes is the generation of reactive oxygen species(ROS) that modifies the redox potential of the cell and is es-sential for numerous signalling pathways (Chen and Zweier,2014) Mitochondrial dysfunction occurs under pathophysi-ological conditions and involves malfunction of complexes
en-of oxidative phosphorylation, and an increase in ROS duction that leads to cell death contributing to the develop-ment of heart failure The enzymes of the respiratory chainseem to be the main site of ROS formation, but many otherenzymes contribute to ROS production in failing hearts, in-cluding monoamine oxidases and the cytosolic adaptor pro-tein p66Shc(Di Lisa et al., 2009) Cellular stress signals lead
pro-to translocation of p66Shcinto the mitochondrial brane space, where it oxidizes cytochrome c and generatesROS (Heusch, 2015) Factors that influence ROS production,therefore, critically determine the cell’s fate
intermem-A newly identified signalling molecule in the control ofmitochondrial ROS production that is under the control ofTGFβ signalling is nucleotide-binding domain and leucine-rich repeat containing PYD-3 (NLRP3), a pattern recognitionreceptor that is involved in the pathogenesis of chronic dis-eases and inflammation NLRP3 is expressed in the heart, lo-calized in mitochondria, and interacts with components ofthe redox system (Figure 4) Upon TGFβ stimulation of car-diac fibroblasts, NLRP3 increases mitochondrial ROS produc-tion, which supports SMAD2 phosphorylation and results inthe differentiation of cardiac fibroblasts into myofibroblasts,
an important process in adverse cardiac remodelling (Bracey
et al., 2014) The involvement of NLRP3 in cardiac fibrosishas been confirmed in an in vivo model of hypertension: an-giotensin II infusion for 28 days resulted in TGFβ-mediatedfibrosis in wild-type mice, but NLRP3-deficient mice wereprotected against this angiotensin II-induced fibrosis NLRP3,therefore, is a newly identified mitochondrial signalling fac-tor in TGFβ-induced cardiac remodelling that may promotethe transition to heart failure as it facilitates ROS-mediatedfibrosis
Increased ROS production induces the opening of the tochondrial permeability transition pore (MPTP) (Figure 4)that changes the permeability of the inner mitochondrialmembrane, leading to mitophagy, fusion/fission events andbiogenesis (Brenner and Moulin, 2012) Opening of theMPTP facilitates the release of pro-apoptotic factors fromthe mitochondria that stimulates the activation of caspasesand finally leads to cell death (Kinnally et al., 2011) The addi-tion of noradrenaline induced a concentration-dependentdecrease in mitochondrial membrane potential that was asso-ciated with a switch from compensated hypertrophy to apo-ptosis, thereby indicating that MPTP opening is involved in
mi-TGFβ-guided switches to heart failure
BJP
Trang 8adverse remodelling (Jain et al., 2015) Inhibiting MPTP
opening by overexpression of adenine nucleotide translocase
1 (ANT1) prevented TGFβ1-induced apoptosis in ventricular
cardiomyocytes (Heger et al., 2012) and improved cardiac
function in rats with an activated renin–angiotensin system
(Walther et al., 2007) These findings highlight the
contribu-tion MPTP opening has to the adverse cardiac remodelling
in-duced by TGFβ stimulation and indicate that ANT1 is a
critical component at the inner mitochondria membrane
for regulating MPTP opening (Figure 4)
Besides modulation of MPTP opening, the B-cell lymphoma
2 (Bcl-2) family is a well-known gate keeper in
mitochondria-mediated apoptosis TGFβ can stimulate or inhibit the
expression of pro-apoptotic and anti-apoptotic Bcl-2 family
members (Grünenfelder et al., 2002) After renal artery ligation,
a model for angiotensinII/TGFβ-mediated cardiac hypertrophy,
up-regulation of the pro-apoptotic family member Bax and the
voltage-dependent anion channel-1 (VDAC1) occurred (Figure 4)
Together, they lead to permeabilization of the outer
mito-chondrial membrane, release of cytochrome c from the
intermembrane space into the cytosol, formation of the
apoptosome, activation of caspases and finally the induction
of apoptosis (Mitra et al., 2013) The small heat shock protein,
crystalline B, is able to block the pro-apoptotic action of
VDAC1, and thereby acts as a molecular key that guides
VDAC1 to be anti-apoptotic Therefore, crystalline B may
become an interesting therapeutic target for the prevention
of the transition from compensated hypertrophy to heart
failure Another heat shock protein (Hsp) with anti-apoptotic
action on the mitochondrial level is Hsp22 (Qiu et al., 2011).Overexpression of Hsp22 results in physiological hypertro-phy via up-regulation of NFκB, and binding of Hsp22 to signaltransducer and activator of transcription 3 (STAT3), which is amarker of cardiac stress responses Down-regulation of Hsp22leads to an increased remodelling of the heart and death inknock-out mice after transverse aortic constriction (TAC) bymodulating the nuclear and mitochondrial function of STAT3and STAT3-dependent genes
A further mitochondria-associated candidate, mediating aswitch to pathophysiological hypertrophy is TOM70, atranslocase of the mitochondrial outer membrane (TOM)complex that mediates the import of mitochondrialpreproteins (Figure 4) Li et al (2014b) nicely showed adown-regulation of TOM70 in pathophysiological hypertro-phy in humans as well as animal models This results in thereduced import of optical atrophy-1 (OPA1)– a protein impor-tant for mitochondrial fusion–, a reduction in complex I ac-tivity and finally in ROS production As a consequence,changes in the outer mitochondrial membrane and/or innermitochondrial membrane occurred, followed by apoptoticevents as discussed above In addition, increased TOM70levels made cardiomyocytes completely resistant to the ef-fects of various pro-hypertrophic stimuli
These findings explain the significance of the modulation ofmitochondrial pores by, for example, VDAC1, crystalline B orTOM70 for cardiac hypertrophy to progress to heart failure
At the onset of the development of heart failure, a bolic shift from fatty acid to glucose metabolism has been
meta-Figure 4
The central role of mitochondria in LV heart failure can be modulated by TGFβ Hypertrophy, fibrosis and apoptosis can be controlled by chondria via generation of ROS NLRP3 is a newly identified molecule that enhances mitochondrial ROS production and that is controlled by TGFβ
mito-or angiotensin II (AngII) miR181c enhances ROS production via modulation of complex IV of the respiratmito-ory chain TOM70, acting as a repressmito-or
of mitochondrial ROS production, is found to be reduced in heart failure This reduction then provokes enhancement of ROS Enhancement ofmitochondrial uncoupling protein during stimulation ofβ-adrenoceptors by noradrenaline (NA) and TGFβ-receptor activation results in reducedenergy production and impaired contractile function Opening of the MPTP plays a central role in the induction of apoptosis Opening of this porecan be modulated by the accessory proteins VDAC and ANT1 Their expression is regulated by crystalline B, TGFβ, AngII, ROS and β-adrenoceptors(ADRB) Central molecules that modulate mitochondrial processes in heart failure are depicted in red
BJP J Heger et al.
8 British Journal of Pharmacology (2016)173 3–14
Trang 9described This shift is due to the down-regulation of
en-zymes for fatty acid oxidation, whereas glycolytic enen-zymes
are up-regulated (Sack et al., 1996) This enables the heart to
increase its metabolic substrate efficiency in relation to O2
consumption However, the metabolic shift seems to be
re-lated to adverse cardiac remodelling A key regulator of
en-ergy homeostasis induced by stimulation of glycolysis and
glycogen accumulation is AMP-activated kinase (AMPK),
which is activated during cardiac remodelling (Dolinsky and
Dyck, 2006; Kolwicz and Tian, 2011) Just recently,
myostatin, a member of the TGFβ superfamily, was identified
as a repressor of AMPK (Biesemann et al., 2014) Myostatin
reduces muscle growth (skeletal or cardiac), and thus
pro-tects the heart against hypertrophy and failure, and this
function of myostatin is, in part, mediated via repression
of AMPK and the prevention of a metabolic switch towards
glycolysis
In addition to the metabolic shift, a down-regulation of
transporters for glucose (GLUT1/4) and fatty acid (CD36)
up-take into cardiomyocytes as well as a reduction of
trans-porters for pyruvate (PDH) or the carnitine shuttle (CPT1/2)
in mitochondria contribute to heart failure development, as
deletions of these transporters provoked cardiac remodelling
and or dysfunction (Bersin et al., 1994; Liao et al., 2002;
Domenighetti et al., 2010; Lai et al., 2014)
Thus, enhancing the uptake mechanisms for glucose and
fatty acids into cardiomyocytes, as well as for metabolized
substrates into mitochondria can attenuate heart failure
pro-gression Furthermore, prevention of the metabolic switch,
probably via AMPK, is a promising target for therapeutic
approaches against heart failure development
MicroRNAs in heart failure
An increased ability to regulate the processes involved in cardiacremodelling is attributed to miRNAs miRNAs are small non-coding RNAs that target the 3´-untranslated region or 5’-untranslated region of mRNA transcripts This results in the de-stabilization or translational repression of mRNAs (Bartel, 2004).Furthermore, miRNAs can regulate gene transcription by induc-ing histone modifications or DNA methylations (Hawkins andMorris, 2008) In fact, one single miRNA can affect many targetgenes generating a broad network of miRNA-controlled gene ex-pression that has a huge effect on different biological processesincluding cardiac remodelling Analysing the role of miRNAs
in heart failure development has already identified some ising new therapeutic targets
prom-The RNase III endonuclease Dicer is essential for the ing of pre-miRNA into its mature form In the adult myocar-dium, a loss of Dicer-induced biventricular enlargement isaccompanied by hypertrophic growth of cardiomyocytes, ven-tricular fibrosis and functional defects (da Costa Martins et al.,2008) A similar study by Chen et al (2008) revealed signs of di-lated cardiomyopathy and heart failure after cardiac-specific de-letion of Dicer Furthermore, they found that the level of Dicerprotein was significantly reduced in in human patients with di-lated cardiomyopathy and failing hearts These findings indi-cate that miRNAs have a major function in the control ofheart failure development and progression
process-Either an up-regulation or down-regulation of miRNAsunder pressure overload can mediate cardiac remodelling,for example, when miR25 is increased the activity of the sar-coplasmic reticulum (SR) Ca2+ATPase (SERCA2A) is reduced
Figure 5
Influence of miRNAs on LV adverse remodelling can be modulated by β-adrenoceptors (ADRBs) or TGFβ miRNAs that have been demonstrated toreverse or promote adverse cardiac remodelling are depicted Up-regulation of miR15 or miR22 prevents the induction offibrosis or apoptosisunder pressure overload (TAC) orβ-adrenoceptor stimulation (ISO) while preserving effects on moderate, compensatory hypertrophy, as whenthese miRs are inhibited adverse remodelling develops In contrast, up-regulation of miR25 or miR21 under TAC enhances adverse cardiac remod-elling, and down-regulation of miR133a under TAC preserves cardiac function, whereas the overexpression of miR133a results in the development
of adverse remodelling Black arrows indicate the responses of the cell to TAC or ISO Switch molecules in the process of adverse remodelling aredepicted in red Green arrows and symbols indicate interference of miR expression by anti-miRs or transgenic overexpression
TGFβ-guided switches to heart failure
BJP
Trang 10and Ca-handling is impaired Anti-miR25 reverses hypertrophy,
fibrosis and heart failure progression after TAC (Wahlquist et al.,
2014) (Figure 5) miR133a is down-regulated under pressure
over-load and when this down-regulation is prevented in transgenic
mice TAC-induced fibrosis and apoptosis are attenuated, whereas
hypertrophy is not affected (Matkovich et al., 2010); hence, these
diverse processes of cardiac remodelling are differentiated
(Figure 5) This indicates that by altering the levels of miR133a,
it may be possible to stop the adverse remodelling processes
while maintaining the compensatory effects of hypertrophy
The development of moderate hypertrophy and physiological
cardiac remodelling induced by an infusion of isoprenaline is
converted to adverse remodelling in miR22 knock-out mice with
a marked enhancement of fibrosis and apoptosis that finally
leads to dilated cardiomyopathy (Huang et al., 2013) The effects
of miR22 seem to be mediated by the inhibition of histone
deacetylases, which indicates that miR22 has a role in the
epige-netic regulation of gene expression during cardiac hypertrophy
These findings indicate that duringβ-adrenoceptor stimulation
miR22 prevents the transition from compensated hypertrophy
to heart failure progression (Figure 5)
A new aspect of miRNA-controlled signalling relates to
the translocation of nuclear-encoded miR181c to
mitochon-dria (Das et al., 2014) This results in the remodelling of
mitochondrial complex IV and an enhanced production of
ROS (Figure 4) The overexpression of miR181c induces
ventricular dysfunction indicating that miRNAs can
modu-late exercise capacity directly at the mitochondrial level
Interestingly, this miR181c-induced dysfunction was not
ac-companied by cardiac hypertrophy Unfortunately, these
ob-servations were only carried out in vitro with a simulated
overexpression of miR181c and its role in cardiovascular
dis-ease in vivo has still to be proven
With regard to TGFβ signalling, the miR15 family needs to
be mentioned This family comprises six highly conserved
fam-ily members that are up-regulated in human heart failure and
can inhibit numerous components of the TGFβ signalling
path-way (Tijsen et al., 2014) Inhibition of one family member by
anti-miR15b in mice also resulted in the down-regulation of
other family members and predominantly enhanced
SMAD-signalling and cardiac fibrosis, especially after TAC (Figure 5)
Therefore, an up-regulation of miR15 members in heart failure
acts as negative feedback mechanism on TGFβ signalling in
or-der to restrict adverse remodelling However, due to the
ubiqui-tous expression of miR15 and because miR15 is also involved
in inducing apoptosis after acute myocardial infarction
(Hullinger et al., 2012), the therapeutic application of miR15
against heart failure progression should be treated with caution
as a substantial amount of additional work needs to be done to
exclude negative side effects In addition to miRs modifiying
TGFβ signalling pathways, TGFβ itself is also known as a
regula-tor of miRs In the context of heart failure progression, miR21
should be highlighted; miR21 is selectively up-regulated in
fibroblasts upon TGFβ stimulation, and in the failing
myocar-dium (Thum et al., 2008; Topkara and Mann, 2011) It has been
shown to induce cardiac fibrosis by enhancing the proliferation
of fibroblasts and to stimulate endothelial mesenchymal
transi-tion during TGFβ stimulatransi-tion (Kumarswamy et al., 2012) and
also to act as a mediator of adverse cardiac remodelling after
TAC (Thum et al., 2011) (Figure 5) Just recently, cardiac
fibroblasts were shown to secrete miRNA-enriched exosomes
(including miR21) Fibroblast-derived miR21 acts as a potentparacrine RNA molecule that induces cardiomyocyte hypertro-phy (Bang et al., 2014) miR155, secreted by macrophages, alsohas paracrine effects on the heart, because miR155 knockout
in macrophages prevented angiotensin II-induced or induced cardiac hypertrophy and dysfunction althoughfibrosis was still present
TAC-These findings showing that miRNAs can exert paracrineeffects implies that systemic pharmacological interference
of miRNA signalling by the use of anti-miRNAs might be ful for preventing heart failure progression Such a systemictherapeutic intervention would be easy to apply clinically.However, in cardiac pathophysiology, the systemic applica-tion of anti-miRNAs is still restricted to basic science studies
use-A promising approach in this direction has been shown byMontgomery et al (2011) using locked nucleic acid (LNA)-modified miR208a-antisense oligonucleotides Systemic de-livery of these oligos silenced miR208a-expression in theheart, thereby preventing hypertension-induced heart failure
in Dahl hypertensive rats by reducing cardiomyocyte trophy, cardiac fibrosis and improving cardiac function Be-cause cardiac miR208 overexpression in transgenic miceinduced cardiac hypertrophy, the reduction of miR208a inthe heart by LNA-antisense oligos is at least in part responsi-ble for cardioprotection in hypertensive Dahl rats However,reductions in the levels of circulating miR499 and miR208bwere also found during the treatment with LNA-modified208a antisense oligos Therefore, a combination of local andsystemic effects may contribute to the protective effects.All these studies demonstrate the enormity of the miRNAnetwork and its influence on heart failure progression Finetuning of specific miRNAs is essential for physiological hypertro-phy or decompensation, and thus has great therapeutic potentialfor the treatment of heart failure patients
hyper-Right heart failure
For many years, analysis of left ventricular systolic tion was at the centre of heart failure research, which is alsothe focus of our review However, in recent years, some prom-ising advances in the analysis of right heart failure have beenmade that should be discussed here
dysfunc-One major cause for right ventricular dysfunction is nary hypertension (PAH) Due to an increase in pulmonary vas-cular resistance, afterload on the right ventricle (RV) increases,leading to right heart failure which determines the prognosis
pulmo-of patients with PAH Therefore, it is pulmo-of utmost importance todefine new therapeutic strategies to reduce RV remodelling inorder to improve patient prognosis (Ryan et al., 2015).The RV, similar to the LV, compensates an increased workload due to hypertension by hypertrophic growth processes.However, compensatory remodelling is limited and over time,the RV decompensates, finally leading to heart failure Thereseem to be chamber-specific responses, and thus, a simple ex-tension of LV findings to the RV is not possible An interestingnew finding in this respect comes from Schreckenberg et al.(2015) who analysed the effect of chronic NO deprivation onthe remodelling processes in the LV and RV Treatment of ratswith the NOS inhibitorL-NAME resulted in moderate ventricu-lar hypertrophy without signs of dysfunction However, the
BJP J Heger et al.
10 British Journal of Pharmacology (2016)173 3–14
Trang 11RV responded with dilatation and dysfunction A massive
increase in oxygen radicals due to a down-regulation of
the anti-oxidative protein, SOD, was only found in the RV,
and this was the cause of RV remodelling, whereas in the
LV, anti-oxidative enzymes were even increased under the
L-NAME treatment Thus, oxidative stress is a much greater
risk factor in the RV compared with the LV The reduction
in SOD and the development of RV dysfunction could be
attenuated by captopril, an angiotensin-converting enzyme
inhibitor with high anti-oxidative capacity Interestingly,
enhanced ROS production in the RV is responsible for
HIF1α inhibition and suppression of angiogenesis, thereby
resulting in capillary rarefication of the RV and chronic
ischaemia (Bogaard et al., 2009)
A further positive effect of therapeutics against the renin–
angiotensin system was demonstrated by Friedberg et al (2013)
Pulmonary artery banding in rabbits induced RV hypertrophy
and biventricular up-regulation of the TGFβ pathways, including
activation of SMAD3, fibrosis and apoptosis These responses
were blocked by losartan, an inhibitor of the AT1receptor In
ad-dition, TGFβ had negative effects on vascular remodelling in PAH
(Zaiman et al., 2008) Therefore, blocking this pathway has
posi-tive effects on the vasculature as well as on both ventricles and
seems to be a promising therapeutic target
Further interesting targets in RVare (i) the up-regulation of
pyruvate dehydrogenase kinase (PDK), as it is responsible for
the metabolic shift to inefficient cytosolic glyocolysis and RV
dysfunction (Piao et al., 2010) and (ii) the inhibition of
G-protein receptor kinase 2 (GRK2) to prevent the
down-regulation of β-adrenoceptors and preserve RV function in
PAH (Piao et al., 2012)
Conclusion and outlook
Cardiac remodelling is a multifactorial-induced process The
present pharmacological interventions are able to postpone
the onset of heart failure but are unable to prevent the
ongo-ing process of cardiac remodellongo-ing in hypertrophied hearts
Newly identified signalling molecules might have the
poten-tial to serve as therapeutic targets in the treatment of heart
failure Therefore, different and common pathways of left
and RVs should be considered Among the decisive signalling
molecules for LV heart failure progression are modulators of
mitochondrial pores, such as NLRP3, ANT1, VDAC1 or
TOM70 and various miRNAs; for example, miR22 specifically
impairs the progression from compensated hypertrophy to
left ventricular heart failure and miR21 has significant
potential to induce fibrosis Furthermore, prevention of the
down-regulation of mitochondrial transporters might improve
the metabolic situation in the remodelled myocardium
Other switch molecules control the adrenoceptor-mediated
signalling pathway, like SMAD4, TAK1 orβ-arrestin, thereby
modulating hypertrophic, apoptotic necroptotic and
con-tractile responses of the cell In the RV, a higher
susceptibil-ity to oxygen radical production has been found, thereby
indicating the importance of using therapeutic ROS
scaven-gers Furthermore, PDK and GRK2 have been identified as
new targets in RV
Many of these targets are regulated by TGFβ, and
ad-ministration of an ALK5 inhibitor after aortic banding
prevented cardiac fibrosis and attenuated cardiac tion However, mortality rates of animals increased due toenhanced left ventricular dilatation and inflammation(Engebretsen et al., 2014) Therefore, a more target-orientated approach needs to be used to inhibit the detri-mental TGFβ pathways but preserve the protective ones
dysfunc-As exemplified in this review, newly identified switchmolecules may offer novel options in the therapeutic ap-proach against heart failure development and progression
Con flict of interest
None
ReferencesAbbate A, Arena R, Abouzaki N, Van Tassell BW, Canada J, Shah K et al.(2015) Heart failure with preserved ejection fraction: refocusing ondiastole Int J Cardiol 179: 430–440 doi:10.1016/j.ijcard.2014.11.106.Alexander SP, Benson HE, Faccenda E, Pawson AJ, Sharman JL,Spedding M et al (2013a) The Concise Guide to
PHARMACOLOGY 2013/14: G protein-coupled receptors Br JPharmacol 170: 1459–1581 doi:10.1111/bph.12445
Alexander SPH, Benson HE, Faccenda E, Pawson AJ, Sharman JL,Spedding M et al (2013b) The concise guide to
PHARMACOLOGY 2013/14: catalytic receptors Br J Pharmacol170: 1676–1075
Alexander SPH, Benson HE, Faccenda E, Pawson AJ, Sharman JL,Spedding M et al (2013c) The concise guide to PHARMACOLOGY2013/14: transporters Br J Pharmacol 170: 1706–1796
Alexander SPH, Benson HE, Faccenda E, Pawson AJ, Sharman JL,Spedding M et al (2013d) The concise guide to PHARMACOLOGY2013/14: enzymes Br J Pharmacol 170: 1797–1867
Bang C, Batkai S, Dangwal S, Gupta SK, Foinquinos A, Holzmann A
et al (2014) Cardiac fibroblast-derived microRNA passenger enriched exosomes mediate cardiomyocyte hypertrophy J ClinInvest 124: 2136–2146 doi:10.1172/JCI70577
strand-Bartel DP (2004) MicroRNAs: genomics, biogenesis, mechanism, andfunction Cell 116: 281–297 doi:10.1016/S0092-8674(04)00045-5.Barth E, Stämmler G, Speiser B, Schaper J (1992) Ultrastructuralquantitation of mitochondria and myofilaments in cardiac musclefrom 10 different animal species including man J Mol Cell Cardiol24: 669–681 doi:10.1016/0022-2828(92)93381-S
Bathgate-Siryk A, Dabul S, Pandya K, Walklett K, Rengo G, Cannavo A
et al (2014) Negative impact of β-arrestin-1 on post-myocardialinfarction heart failure via cardiac and adrenal-dependentneurohormonal mechanisms Hypertension 63: 404–412
et al (2007) TAK1-dependent signaling requires functionalinteractionwith TAB1/TAB3 J Biol Chem 282: 3918–3928
doi:10.1074/jbc.M608867200
TGFβ-guided switches to heart failure
BJP
Trang 12Biesemann N, Mendler L, Wietelmann A, Hermann S, Schäfers M,
Krüger M et al (2014) Myostatin regulates energy homeostasis in the
heart and prevents heart failure Circ Res 115: 296–310 doi:10.1161/
CIRCRESAHA.115.304185
Boluyt MO, O’Neill L, Meredith AL, Bing OH, Brooks WW, Conrad
CH et al (1994) Alterations in cardiac gene expression during the
transition from stable hypertrophy to heart failure Circ Res 75:
23–32 doi:10.1161/01.RES.75.1.23
Bogaard HJ, Natarajan R, Henderson SC, Long CS, Kraskauskas D,
Smithson L et al (2009) Chronic pulmonary artery pressure
elevation is insufficient to explain right heart failure Circulation
120: 1951–1960 doi:10.1161/CIRCULATIONAHA.109.883843
Bracey NA, Gershkovich B, Chun J, Vilaysane A, Meijndert HC,
Wright JR Jr et al (2014) Mitochondrial NLRP3 protein induces
reactive oxygen species to promote Smad protein signaling and
fibrosis independent from the inflammasome J Biol Chem 289:
19571–19584 doi:10.1074/jbc.M114.550624
Brenner C, Moulin M (2012) Physiological roles of the permeability
transition pore Circ Res 111: 1237–1247 doi:10.1161/
CIRCRESAHA 112.265942
Burch ML, Osman N, Getachew R, Al-Aryahi S, Poronnik P, Zheng WG
et al (2012) Protein coupled receptor transactivation: extending the
paradigm to include serine/threonine kinase receptors Int J Biochem
Cell Biol 44: 722–727 doi:10.1016/j.biocel.2012.01.018
Chen JF, Murchison EP, Tang R, Callis TE, Tatsuguchi M, Deng Z et al
(2008) Targeted deletion of Dicer in the heart leads to dilated
cardiomyopathy and heart failure Proc Natl Acad Sci U S A 105:
2111–2116 doi:10.1073/pnas.0710228105
Chen Y-R, Zweier JL (2014) Cardiac mitochondria and reactive
oxygen species generation Circ Res 114: 524–537 doi:10.1161/
CIRCRESAHA 114.300559
Conner DA, Mathier MA, Mortensen RM, Christe M, Vatner SF,
Seidman CE et al (1997) beta-Arrestin1 knockout mice appear
normal but demonstrate altered cardiac responses to beta-adrenergic
stimulation Circ Res 81: 1021–1026 doi:10.1161/01.RES81.6.1021
da Costa Martins PA, Bourajjaj M, Gladka M, Kortland M, van Oort RJ,
Pinto YM et al (2008) Conditional dicer gene deletion in the postnatal
myocardium provokes spontaneous cardiac remodeling Circulation
118: 1567–1576 doi:10.1161/CIRCULATIONAHA.108.769984
Das S, Bedja D, Campbell N, Dunkerly B, Chenna V, Maitra A et al
(2014) miR-181c regulates the mitochondrial genome, bioenergetics,
and propensity for heart failure in vivo PLoSOne 9: e96820
doi:10.1371/journal.pone.0096820
Dennis PA, Rifkin DB (1991) Cellular activation of latent
transforming growth factor beta requires binding to the
cation-independent mannose 6-phosphate/insulin-like growth factor type II
receptor Proc Natl Acad Sci U S A 88: 580–584, PMID: 1846448
Depre C, Wang Q, Yan L, Hedhli N, Peter P, Chen L et al (2006)
Activation of the cardiac proteasome during pressure overload
promotes ventricular hypertrophy Circulation 14: 1821–1828
doi:10.1161/CIRCULATIONAHA.106.637827
Di Lisa F, Kaludercic N, Carpi A, Menabò R, Giorgio M (2009)
Mitochondria and vascular pathology Pharmacol Rep 61: 123–130,
PMID: 19307700
Dolinsky VW, Dyck JR (2006) Role of AMP-activated protein kinase in
healthy and diseased hearts Am J Physiol Heart Circ Physiol 291:
H2557–H2569 doi:10.1152/ajpheart.00329.2006
Domenighetti AA, Danes VR, Curl CL, Favaloro JM, Proietto J,
Delbridge LM (2010) Targeted GLUT-4 deficiency in the heart induces
cardiomyocyte hypertrophy and impaired contractility linked withCa(2+) and protonflux dysregulation J Mol Cell Cardiol 48: 663–672.doi:10.1016/j.yjmcc.2009.11.017
Drews O, Tsukamoto O, Liem D, Streicher J, Wang Y, Ping P (2010).Differential regulation of proteasome function in isoproterenol-induced cardiac hypertrophy Circ Res 107: 1094–1101 doi:10.1161/CIRCRESAHA.110.222364
Engebretsen KV, Skårdal K, Bjørnstad S, Marstein HS, Skrbic B,Sjaastad I et al (2014) Attenuated development of cardiacfibrosis
in left ventricular pressure overload by SM16, an orally active inhibitor
of ALK5 J Mol Cell Cardiol 76: 148–157 doi:10.1016/j
yjmcc.2014.08.008
Friedberg MK, Cho MY, Li J, Assad RS, Sun M, Rohailla S et al (2013).Adverse biventricular remodeling in isolated right ventricularhypertension is mediated by increased transforming growth factor-β1signaling and is abrogated by angiotensin receptor blockade Am JRespir Cell Mol Biol 49: 1019–1028 doi:10.1165/rcmb.2013-0149OC.Goumans MJ, Valdimarsdottir G, Itoh S, Rosendahl A, Sideras P, tenDijke P et al (2002) Balancing the activation state of the
endothelium via two distinct TGF-beta type I receptors EMBO J 21:
1743–1753, PMID:11927558
Grünenfelder J, Miniati DN, Murata S, Falk V, Hoyt EG, Robbins RC(2002) Up-regulation of Bcl-2 through hyperbaric pressuretransfection of TGF-beta1 ameliorates ischemia-reperfusion injury inrat cardiac allografts J Heart Lung Transplant 21: 244–250
Heusch G (2015) Mitochondria at the heart of cardiovascularprotection: p66shc-friend or foe? Eur Heart J 36: 469–471
doi:10.1093/eurheartj/ehu409
Hill C, Würfel A, Heger J, Meyering B, Schlüter KD, Weber M
et al (2013) Inhibition of AP-1 signaling by JDP2 overexpressionprotects cardiomyocytes against hypertrophy and apoptosisinduction Cardiovasc Res 99: 121–128 doi:10.1093/cvr/cvt094.Hoppe UC, Erdmann E (2009) Kapitel 5 Chronische Herzinsuffizienz,aus Erdmann E Klinische Kardiologie Krankheiten des Herzens, desKreislaufs und herznaher Gefäße 7 Auflage, Springer VerlagHuang CY, Kuo WW, Yeh YL, Ho TJ, Lin JY, Lin DY et al (2014) ANG IIpromotes IGF-IIR expression and cardiomyocyte apoptosis byinhibiting HSF1 via JNK activation and SIRT1 degradation Cell DeathDiffer 21: 1262–1274 doi:10.1038/cdd.2014.46
Huang ZP, Chen J, Seok HY, Zhang Z, Kataoka M, Hu X et al (2013).MicroRNA-22 regulates cardiac hypertrophy and remodeling inresponse to stress Circ Res 112: 1234–1243 doi:10.1161/
CIRCRESAHA.112.300682
Hullinger TG, Montgomery RL, Seto AG, Dickinson BA, Semus HM,Lynch JM et al (2012) Inhibition of miR-15 protects against cardiacischemic injury Circ Res 110: 71–81 doi:10.1161/
CIRCRESAHA.111.244442
BJP J Heger et al.
12 British Journal of Pharmacology (2016)173 3–14
Trang 13Huntgeburth M, Tiemann K, Shahverdyan R, Schlüter KD,
Schreckenberg R, Gross ML et al (2011) Transforming growth factor
β1oppositely regulates the hypertrophic and contractile response to
β-adrenergic stimulation in the heart PLoS One 6: e26628
doi:10.1371/journal.pone.0026628
Ikeuchi M, Tsutsui H, Shiomi T, Matsusaka H, Matsushima S, Wen J
et al (2004) Inhibition of TGF-beta signaling exacerbates early
cardiac dysfunction but prevents late remodeling after infarction
Cardiovasc Res 64: 526–535 doi:10.1016/j.cardiores.2004.07.017
Jain A, Atale N, Kohli S, Bhattacharya S, Sharma M, Rani V (2015) An
assessment of norepinephrine mediated hypertrophy to apoptosis
transition in cardiac cells: a signal for cell death Chem Biol Interact
225: 54–62 doi:10.1016/ j.cbi.2014.11.017
Khan S, Joyce J, Margulies KB, Tsuda T (2014) Enhanced bioactive
myocardial transforming growth factor-β in advanced human heart
failure Circ J 78: 2711–2718 doi:10.1253/circj.CJ-14-0511
Kinnally KW, Peixoto PM, Ryu SY, Dejean LM (2011) Is mPTP the
gatekeeper for necrosis, apoptosis, or both? Biochim Biophys Acta 4:
616–624 doi:10.1016/j.bbamcr.2010.09.013
Kolwicz SC Jr, Tian R (2011) Glucose metabolism and cardiac
hypertrophy Cardiovasc Res 90: 194–201 doi:10.1093/cvr/cvr071
Kumarswamy R, Volkmann I, Jazbutyte V, Dangwal S, Park DH,
Thum T (2012) Transforming growth factor-β-induced
endothelial-to-mesenchymal transition is partly mediated by microRNA-21
Arterioscler Thromb Vasc Biol 32: 361–369 doi:10.1161/
ATVBAHA.111.234286
Lai L, Leone TC, Keller MP, Martin OJ, Broman AT, Nigro J et al
(2014) Energy metabolic reprogramming in the hypertrophied and
early stage failing heart: a multisystems approach Circ Heart Fail 7:
1022–1031 doi:10.1161/CIRCHEARTFAILURE.114.001469
Leksa V, Godar S, Schiller HB, Fuertbauer E, Muhammad A,
Slezakova K (2005) TGF-beta-induced apoptosis in endothelial
cells mediated by M6P/IGFII-R and mini-plasminogen J Cell Sci
118 (Pt 19): 4577–4586 doi:10.1242/ jcs.02587
Li L, Chen Y, Doan J, Murray J, Molkentin JD, Liu Q (2014a)
Transforming growth factorβ-activating kinase 1 signaling pathway
critically regulated myocardial survival and remodeling Circulation
130: 2162–2172 doi:10.1161/CIRCULATIONAHA.114.011195
Li J, Qi M, Li C, Shi D, Zhang D, Xie D et al (2014b) Tom70 serves as a
molecular switch to determine pathological cardiac hypertrophy Cell
Res 24: 977–993 doi:10.1038/Cr.2014.9
Liao R, Jain M, Cui L, D’Agostino J, Aiello F, Luptak I et al (2002)
Cardiac-specific overexpression of GLUT1 prevents the development
of heart failure attributable to pressure overload in mice Circulation
106: 2125–2131 doi:10.1161/01.CIR.0000034049.61181
Lijnen PJ, Petrov VV, Fagard RH (2000) Induction of cardiac
fibrosis by transforming growth factor-beta(1) Mol Genet Metab
71: 418–435 PMID: 11001836
Lymperopoulos A, Negussie S (2013).βArrestins in cardiac G
protein-coupled receptor signaling and function: partners in crime or "good cop,
bad cop"? Int J Mol Sci 14: 24726–24741 doi:10.3390/ijms141224726
Matkovich SJ, Wang W, Tu Y, Eschenbacher WH, Dorn LE, Condorelli
G et al (2010) MicroRNA-133a protects against myocardialfibrosis
and modulates electrical repolarization without affecting
hypertrophy in pressure-overloaded adult hearts Circ Res 106:
166–175 doi:10.1161/CIRCRESAHA.109.202176
Mitra A, Basak T, Datta K, Naskar S, Sengupta S, Sarkar S (2013) Role of
α-crystallin B as a regulatory switch in modulating cardiomyocyte
apoptosis by mitochondria or endoplasmic reticulum during cardiac
hypertrophy and myocardial infarction Cell Death Dis 4: e582.doi:10.1038/cddis.2013.114
Montgomery RL, Hullinger TG, Semus HM, Dickinson BA, Seto AG,Lynch JM et al (2011) Therapeutic inhibition of miR-208a improvescardiac function and survival during heart failure Circulation 124:
1537–1547 doi:10.1161/CIRCULATIONAHA.111.030932
Munger JS, Huang X, Kawakatsu H, Griffiths MJ, Dalton SL, Wu J
et al (1999) The integrin alpha v beta 6 binds and activateslatent TGF beta 1: a mechanism for regulating pulmonaryinflammation and fibrosis Cell 6: 319–328 doi:10.1016/S0092-8674(00)80545-0
Narula J, Haider N, Virmani R, DiSalvo TG, Kolodgie FD, Hajjar RJ(1996) Apoptosis in myocytes in end-stage heart failure N Engl JMed 335: 1182–1189 doi:10.1056/NEJM199610173351603.Pawson AJ, Sharman JL, Benson HE, Faccenda E, Alexander SP,Buneman OP et al (2014) The IUPHAR/BPS Guide toPHARMACOLOGY: an expert-driven knowledgebase of drug targetsand their ligands Nucl Acids Res 42 (Database Issue): D1098–106.Piao L, Fang YH, Cadete VJ, Wietholt C, Urboniene D, Toth PT
et al (2010) The inhibition of pyruvate dehydrogenase kinaseimproves impaired cardiac function and electrical remodeling intwo models of right ventricular hypertrophy: resuscitating thehibernating right ventricle J Mol Med (Berl) 88: 47–60
doi:10.1007/s00109-009-0524-6
Piao L, Fang YH, Parikh KS, Ryan JJ, D’Souza KM, Theccanat T et al.(2012) GRK2-mediated inhibition of adrenergic and dopaminergicsignaling in right ventricular hypertrophy: therapeutic implications
in pulmonary hypertension Circulation 126 (24): 2859–2869.doi:10.1161/CIRCULATIONAHA.112.109868
Portbury AL, Ronnebaum SM, Zungu M, Patterson C, Willis MS(2012) Back to your heart: ubiquitin proteasome system-regulatedsignal transduction J Mol Cell Cardiol 52: 526–537 doi:10.1016/j.yjmcc.2011.10.023
Qiu H, Lizano P, Laure L, Sui X, Rashed E, Park JY et al (2011) H11kinase/heat shock protein 22 deletion impairs both nuclear andmitochondrial functions of STAT3 and accelerates the transition intoheart failure on cardiac overload Circulation 24: 406–415
doi:10.1161/CIRCULATIONAHA.110.013847
Rosenkranz S, Flesch M, Amann K, Haeuseler C, Kilter H, Seeland U
et al (2002) Alterations of beta-adrenergic signaling and cardiachypertrophy in transgenic mice overexpressing TGF-beta(1) Am JPhysiol Heart Circ Physiol 283: H1253–H1262 doi:10.1152/ajpheart.00578.2001
Rosenkranz S (2004) TGF-beta1 and angiotensin networking incardiac remodeling Cardiovasc Res 94: 2837–2842 doi:10.1016/j.cardiores.2004.04.030
Ryan JJ, Huston J, Kutty S, Hatton ND, Bowman L, Tian L et al (2015).Right ventricular adaptation and failure in pulmonary arterialhypertension Can J Cardiol 31: 391–406 doi:10.1016/j
cjca.2015.01.023
Sack MN, Rader TA, Park S, Bastin J, McCune SA, Kelly DP (1996) Fattyacid oxidation enzyme gene expression is downregulated in thefailing heart Circulation 94: 2837–2842 doi:10.1161/01
Trang 14cardiomyocytes Cardiovasc Res 67: 87–96 doi:10.1016/j.
cardiores.2005.02.021
Schreckenberg R, Rebelo M, Deten A, Weber M, Rohrbach S, Pipicz M
et al (2015) Specific Mechanisms Underlying Right Heart Failure:
The Missing Upregulation of Superoxide Dismutase-2 and Its Decisive
Role in Antioxidative Defense Antioxid Redox Signal Epub ahead of
print, PMID:25978844
Schröder D, Heger J, Piper HM, Euler G (2006) Angiotensin II
stimulates apoptosis via TGF-beta1 signaling in ventricular
cardiomyocytes of rat J Mol Med (Berl) 84: 975–983 doi:10.1007/
s00109-006-0090-0
Taimor G, Schlüter KD, Best P, Helmig S, Piper HM (2004) Transcription
activator protein 1 mediates alpha- but not beta-adrenergic hypertrophic
growth responses in adult cardiomyocytes Am J Physiol Heart Circ
Physiol 286: H2369–H2375 doi:10.1152/ajpheart.00741.2003
Thum T, Gross C, Fiedler J, Fischer T, Kissler S, Bussen M et al (2008)
MicroRNA-21 contributes to myocardial disease by stimulating MAP
kinase signalling infibroblasts Nature 456: 980–984 doi:10.1038/
nature07511
Thum T, Chau N, Bhat B, Gupta SK, Linsley PS, Bauersachs J, et al
(2011) Comparison of different miR-21 inhibitor chemistries in a
cardiac disease model J Clin Invest 121:461-462: 462–463 doi:
10.1172/JCI45938
Tijsen AJ, van der Made I, van den Hoogenhof MM, Wijnen WJ, van
Deel ED, de Groot NE et al (2014) The microRNA-15 family inhibits
the TGFβ-pathway in the heart Cardiovasc Res 104: 61–71
doi:10.1093/cvr/cvu184
Topkara VK, Mann DL (2011) Role of microRNAs in cardiac
remodeling and heart failure Cardiovasc Drugs Ther 25: 171–182
doi:10.1007/s10557-011-6289-5
Wahlquist C, Jeong D, Rojas-Muñoz A, Kho C, Lee A, Mitsuyama S
et al (2014) Inhibition of miR-25 improves cardiac contractility in
the failing heart Nature 508: 531–535 doi:10.1038/nature13073
Walther T, Tschope C, Sterner-Kock A, Westermann D,
Heringer-Walther S, Riad A et al (2007) Accelerated mitochondrial adenosine
diphosphate/adenosine triphosphate transport improveshypertension-induced heart disease Circulation 115: 333–344.doi:10.1161/CIRCULATIONAHA.106.643296
Wang J, Xu N, Feng X, Hou N, Zhang J, Cheng X et al (2005) Targeteddisruption of Smad4 in cardiomyocytes results in cardiac
hypertrophy and heart failure Circ Res 97: 821–828 doi:10.1161/01.RES.0000185833.42544.06
Watkins SJ, Borthwick GM, Oakenfull R, Robson A, Arthur HM (2012).Angiotensin II-induced cardiomyocyte hypertrophy in vitro is TAK1-dependent and Smad2/3-independent Hypertens Res 35: 393–398.doi:10.1038/hr.2011
Wharton K, Derynck R (2009) TGFbeta family signaling: novelinsights in development and disease Development 136: 3691–3697.doi:10.1242/dev.040584
Xu P, Liu J, Derynck R (2012) Post-translational regulation of TGF-βreceptor and Smad signaling FEBS Lett 586: 1871–1884 doi:10.1016/j.febslet.2012.05.010
Zaglia T, Milan G, Ruhs A, Franzoso M, Bertaggia E, Pianca N et al.(2014) Atrogin-1 deficiency promotes cardiomyopathy andpremature death via impaired autophagy J Clin Invest 124:
2410–2424 doi:10.1172/JCI66339
Zhang D, Gaussin V, Taffet GE, Belaguli NS, Yamada M, Schwartz RJ
et al (2000) TAK1 is activated in the myocardium after pressureoverload and is sufficient to provoke heart failure in transgenic mice.Nat Med 6: 556–563 doi:10.1038/75037
Zhang YE (2009) Non-Smad pathways in TGF-beta signaling Cell Res19: 128–139 doi:10.1038/cr.2008.328
Zhang DW, Shao J, Lin J, Zhang N, Lu BJ, Lin SC et al (2009) RIP3, anenergy metabolism regulator that switches TNF-induced cell deathfrom apoptosis to necrosis Science 325: 332–336 doi:10.1126/science.1172308
Zaiman AL, Podowski M, Medicherla S, Gordy K, Xu F, Zhen L et al.(2008) Role of the TGF-beta/Alk5 signaling pathway in
monocrotaline-induced pulmonary hypertension Am J Respir CritCare Med 177: 896–905 doi:10.1164/rccm.200707-1083OC
BJP J Heger et al.
14 British Journal of Pharmacology (2016)173 3–14
Trang 15Seung-Hyun Jung†, Young Sook Kim†, Yu-Ri Lee and Jin Sook Kim
Korean Medicine Convergence Research Division, Korea Institute of Oriental Medicine (KIOM),
Daejeon 305-811, Korea
Correspondence
Jin Sook Kim, Korean MedicineConvergence Research Division,Korea Institute of Oriental Medicine(KIOM), 1672 Yuseongdae-ro,Yuseong-gu, Daejeon 305-811, Korea.E-mail: jskim@kiom.re.kr
†These authors contributed equally tothis work
BACKGROUND AND PURPOSE
Although a variety of animal models have been used to test drug candidates and examine the pathogenesis of diabetic
retinopathy, time-saving and inexpensive models are still needed to evaluate the increasing number of therapeutic approaches.EXPERIMENTAL APPROACH
We developed a model for diabetic retinopathy using the early stage of transgenic zebrafish (flk:EGFP) by treating embryos with
130 mM glucose, from 3-6 days post fertilisation (high-glucose model) On day 6, lenses from zebrafish larvae were isolated andtreated with 3% trypsin, and changes in hyaloid-retinal vessels were analysed usingfluorescent stereomicroscopy In addition,expression of tight junction proteins (such as zonula occludens-1), effects of hyperosmolar solutions and of hypoxia, and Vegfexpression were assessed by RT–PCR NO production was assessed with a fluorescent substrate Effects of inhibitors of the VEGFreceptor, NO synthesis and a VEGF antibody (ranibizumab) were also measured
KEY RESULTS
In this high-glucose model, dilation of hyaloid-retinal vessels, on day 6, was accompanied by morphological lesions with ruption of tight junction proteins, overproduction of Vegf mRNA and increased NO production Treatment of this high-glucosemodel with an inhibitor of VEGF receptor tyrosine kinase or an inhibitor of NO synthase or ranibizumab decreased dilation ofhyaloid-retinal vessels
dis-CONCLUSIONS AND IMPLICATIONS
Thesefindings suggest that short-term exposure of zebrafish larvae to high-glucose conditions could be used for screening anddrug discovery for diabetic retinopathy and particularly for disorders of retinal vessels related to disruption of tight junctionproteins and excessive VEGF and NO production
Trang 16Hyperglycaemia is involved in retinal vascular dysfunction
asso-ciated with the development of diabetic retinopathy (DR), which
results in worsening vision and eventual blindness Nearly all
pa-tients with type 1 and type 2 diabetes exhibit some lesions after
20 years with disease (Kempen et al., 2004; Roy et al., 2004;
Calcutt et al., 2009) As early DR progresses, non-proliferative
DR enters an advanced proliferative stage Loss of pericytes,
reti-nal vasculature thickening and disruption of tight junction
pro-teins such as zonula occludens-1 (ZO-1) are found in the early
stages of DR (Checchin et al., 2006; Pfister et al., 2008)
Further-more, these events result in decreased circulation, blood vessel
dilation and obstruction, and increased leakage of blood from
the microvascular circulation, thereby increasing VEGF
signal-ling Increased VEGF signalling causes the formation of new
blood vessels from retinal tissues (Guillemin and Brew, 2004;
Armulik et al., 2005; Bergers and Song, 2005; Checchin et al.,
2006; Alvarez et al., 2010) These vascular changes, collectively,
cause a breakdown of the blood-retinal barrier and compromise
retinal function Neovascularization on the retina and posterior
surface of the vitreous, and macular oedema characterized by
retinal thickening from leaky blood vessels, develop during
vari-ous stages of retinopathy (Fong et al., 2003) Moreover,
expres-sion of endothelial NOS (eNOS), a downstream mediator of
VEGF activity, was increased in diabetic retina, and eNOS
inhibi-tion also potently reduced retinal leukocyte adhesion (Joussen
et al., 2002) Several studies indicate that NO plays a crucial role
in VEGF-induced vascular hyperpermeability and angiogenesis
(Morbidelli et al., 1996; Lakshminarayanan et al., 2000)
Addi-tionally, VEGF induces the expression of NOS and stimulates
production of NO (Abu El-Asrar et al., 2004) Therapeutic
ap-proaches such as VEGF inhibitors, aldose reductase inhibitors,
NOS inhibitors, antioxidants, anti-inflammatory agents and
the receptor for advanced glycation end products are proposed
interventions for treating DR (Greene et al., 1999; Joussen et al.,
2002; Starita et al., 2007; Calcutt et al., 2009)
Many species, including dogs, hamsters, rats, mice and
zebrafish, have been used to provide research models for DR
(Engerman and Kern, 1995; Jorgens et al., 2012) These models
employ chemical or genetic modifications to induce early stages
of DR, including degeneration of retinal capillaries (Kempen
et al., 2004) Zebrafish are an attractive animal model for
molec-ular, toxicological and drug development studies because of
their fecundity, as well as their genetic and physiological
similar-ities to mammals (Barros et al., 2008) They are generally suited
to high-throughput screening because of their small size, highproductivity and optical transparency of the embryo (Kitambi
et al., 2009) Furthermore, techniques for generating transgeniclines and gene-targeting mutations have been developed forzebrafish, enabling the establishment of disease models for drugdiscovery The immersion of adult or larvae zebrafish in glucose(0.7–25%) results in diabetic complications, which share similari-ties with streptozotocin (STZ)-induced diabetic mice and diabeticpatients (Gleeson et al., 2007; Alvarez et al., 2010; Liang et al.,2010; Jorgens et al., 2012) Additionally, in retinas of adultzebrafish exposed for 30 days to hyperglycaemia, there are mor-phological changes such as thickening of the vessels, breakdown
of interendothelial cell–cell junction integrity and vessel ment membrane thickening (Gleeson et al., 2007; Alvarez et al.,2010) In larval zebrafish, anti-diabetic compounds reduce expres-sion of phosphoenolpyruvate carboxykinase, which catalyses arate-limiting step in gluconeogenesis and is transcriptionallyregulated by glucagon and insulin Data have shown that larvaezebrafish are an appropriate model for the study of glucose metab-olism (Elo et al., 2007) Our previous study, we tested the inhibi-tory effect of single compounds isolated from herbal extracts onhyaloid-retinal vessel dilation in glucose-induced larval zebrafish(Lee et al., 2013; Yu et al., 2013) However, changes of tight junc-tions in retinal vessels in glucose-induced zebrafish larvae havenot previously been described in a short-term model for selection
base-of effective agents against DR
Here, to examine the potential of larval zebrafish as a modelfor DR, we exposed zebrafish embryos, from flk:EGFP transgenicfish, to a range of concentrations of glucose for 3 days, and iso-lated the retina Alterations in thickness of the hyaloid-retinalvessels, expression of the tight junction protein ZO-1, vascularleakage and mRNA expression for Vegf and nos were measured
in glucose-exposed zebrafish larvae We also tested the effects
of an inhibitor of VEGFR tyrosine kinase, an inhibitor of NOS(L-NAME) and the clinically used VEGF antibody ranibizumab(FDA approved drug for DR)
Methods
Zebrafish maintenanceAll animal husbandry and experimental protocols complied withinstitutional guidelines and were approved by local ethical boards(Korea Institute of Oriental Medicine Animal Care and Use Com-mittee) Studies involving animals are reported in accordance with
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 ConciseGuide to PHARMACOLOGY 2013/14 (a bAlexander et al., 2013a,b)
BJP S-H Jung et al.
16 British Journal of Pharmacology (2016)173 15–26
Trang 17the ARRIVE guidelines for reporting experiments involving
ani-mals (Kilkenny et al., 2010; McGrath et al., 2010) Adult zebrafish
were maintained under standard conditions at 28.5°C with a
14 h light/10 h dark cycle (Nusslein and Dahm, 2002) Embryos
were obtained from crosses between flk:EGFP transgenicfish and
raised in embryonic water Transgenic zebrafish (flk:EGFP) were
provided by Professor C-H Kim (Chungnam National University)
Embryonic stages were determined by days post-fertilization (dpf)
Fluorescent images were collected at 6 dpf using a fluorescent
dissecting microscope SZX16 (Olympus, Tokyo, Japan)
Treatments and measurement of glucose levels
Zebrafish transgenic (flk:EGFP) embryos (3 dpf) were placed in
24-well plates (five embryos per well) and maintained in 2 mL
embryonic water (sea salt, Sigma, USA, 0.06 g·L 1) containing
a range of concentrations of glucose (30, 60, 90, 120, 125, 130,
135 and 150 mM) Media were maintained for 3 days, and
embryos werefixed, and survival rates were analysed at 6 dpf
In later experiments, embryos were exposed to a single
concen-tration of glucose (130mM) from 3 to 6 dpf and are referred to as
high glucose (HG)-treated larvae
To assess the effects of hyperosmolarity, embryos were
exposed to mannitol (130 mM; Sigma, USA, Cat No C8661),
in-stead of glucose, but under the same conditions To assess effects
of hypoxia, embryos were exposed to CoCl2 (120μM; Sigma,
USA, Cat No M9546) instead of glucose, but under the same
conditions The effects of the VEGF receptor tyrosine kinase
in-hibitor (0.5 or 1μM; Calbiochem, Germany, Cat No 676500),
or ranibizumab (1 or 2.5μg·mL 1
; Lucentis, Novatris, Stein,Switzerland) or L-NAME (10 or 20μM; Sigma, USA, Cat No
N5751) were tested by addition to the HG treatment for 3 days
Quantitative analysis of glucose levels was performed from
whole body lysates using a glucose assay kit (Sigma, USA) Briefly,
20 zebrafish larvae in each experimental group were sonicated in
500μL deionized water on ice According to the instructions,
standard curves were generated using glucose standard solution
A total of 2 mL assay reagent (Sigma) was added and incubated
for 30 min at 37°C Fluorescence (540 nm) was measured using
a BioTek plate reader equipped with GEN5 software (v.2.04,
BioTek, Winooski, VT, USA)
Lens isolation and measurement of
hyaloid-retinal vessel diameter
At 6 dpf, zebrafish larvae were fixed with 4%
paraformalde-hyde and stored overnight at 4°C They were then washed
with distilled water (1.5 ml per well, 3 times within 1 h) and
lenses containing hyaloid-retinal vessels were isolated after
incubation with 3% trypsin in Tris–HCl buffer (pH 7.8), for
80 min at 37°C (Figure 1) Images of hyaloid-retinal vessels,
optic disc (OD) branches and lens size were obtained using
an Olympus stereomicroscope (SZX16) or a laser scanning
confocal microscope (FV10i, Olympus, Tokyo, Japan) Their
diameters were determined byIMAGEJsoftware (Figure S1)
Total RNA extraction and RT-PCR
Total RNA from whole embryos was extracted using TRIzol reagent
according to the manufacturer’s protocol (iNtRON, Korea) and
reverse transcribed using M-MLV reverse transcriptase (Invitrogen,
USA) The reverse transcription (RT) product (1μL) was used as
a template for PCR amplification of Vegf165 and β-actin using
specific primers (5 pM; Genotech, Korea), Taq DNA polymerase(Elpis, Korea) and the following cycles: 95°C for 30 s, 55°C for
30 s and 68°C for 40 s for 30 cycles Primer pairs were as follows:Vegf165, forward primer 5′-CTC CTC CAT CTG TCT GCT GTAAAG-3′ and reverse primer 5′-CTC TCT GAG CAA GGC TCACAG-3′; nos2a, forward primer 5′-GTG TTC CCT CAG AGAACA GAT-3′ and reverse primer 5′-GAT CAG TCC TTT GAA GCTGAC-3′; and β-actin, forward primer 5′-GAG AAG ATC TGG CATCAC AC-3′ and reverse primer 5′-ATC AGG TAG TCT GTC AGGTC-3′ PCR products were separated by electrophoresis on 1.2%agarose gels and visualized by ethidium bromide staining.Whole-mount immunostaining
Whole-mount immunostaining was performed as previouslydescribed (Jung et al., 2010) using primary antibodies againstmouse ZO-1 (1:500, Invitrogen, USA) Forfluorescent detection
of antibody labelling, we used Alexa Fluor 568 anti-mouse jugate (1:500, Molecular Probes, Eugene, OR, USA) and Hoechst
con-33342 (1:1000, Thermo Fisher Scientific, Waltham, MA, USA).Fluorescence images were examined using an Olympus laserscanning confocal microscope (FV10i)
NO detectionLive imaging detection of NO production was carried out asdescribed by Lepiller et al (2007) Briefly, wild-type larvae treatedwith control and HG at 6 dpf were incubated in 5μM 4,5-diamino-fluorescein diacetate (DAF-FM DA) (Molecular Probes)for 30 min, rinsed in egg water, anaesthetized and imaged usingOlympus laser scanning confocal microscopy (FV10i)
Data analysisResults are expressed as the mean ± SEM of multiple experi-ments Experimental data are from 40–60 embryos per treat-ment, from 2–3 independent clutches Data were analysedusing one-way ANOVAwith Tukey’s post hoc analysis or unpairedStudent’s t-tests andPRISMsoftware (GraphPad, San Diego, CA,USA) P< 0.05 was considered statistically significant
to DR models Thus, retinal vascular changes in HG zebrafishmay reflect similar changes observed in mammalian models
To investigate changes in hyaloid-retinal vessels induced by
HG in zebrafish Tg (flk:EGFP), we exposed embryos to anrange of concentrations of glucose (30–150 mM) from 3 to
6 dpf and analysed the diameter of the hyaloid-retinal vesselsobtained at 6dpf (Figure 2) The hyaloid-retinal vasculature be-gins to be generated on 2.5 dpf in normal zebrafish development(Alvarez et al., 2007) Thus, we exposed embryos for 3 days, from
3 to 6 dpf to the different levels of glucose The experimentalprotocols used to measure glucose-induced changes in hyaloid-retinal vessel diameter in zebrafish embryos are summarised inFigure 1 (and S1) Using retinae isolated at 6 dpf, we found that
HG-induced changes in hyaloid-retinal vessels in ZF
BJP
Trang 18the diameter of hyaloid-retinal vessels increased
concentration-dependently, from 30 to 135 mM glucose (Figure 2A)
Further-more, we observed that the proportion of lenses containing
vessels with diameter more than 1.5 AU (artificial unit) also
increased with the concentration of glucose, from about 20%
of lenses with large vessels at 30mM glucose to just over 80%
of lenses at 130mM (Figure 2B) This effect is referred to as
inductivity The effects of these conditions on the survival of
the larvae is shown in Figure 2C Survival was unaffected,
com-pared with control (0 mM glucose) values, from 30 to 120 mM
glucose However, at 135 and 150 mM glucose there was a icant decrease in survival at 6 dpf
signif-On the basis of the results shown in Figure 2, we chose130mM glucose as a standard treatment to induce changes
in the retinal vessels and this is referred to as the HG tion Under these HG conditions, we examined the vesseldiameter, the rate of induction , overall survival rates andglucose levels in the zebrafish larvae, along with other aspects
condi-of embryonic development, as described below (Figure 3) In
10 preparations, HG treatment did not induce the dilated
Figure 1
Overall strategy for rapid screening to identify therapeutic drugs for DR in zebrafish larvae (A) Fertilized embryos from zebrafish Tg (flk:EGFP) pairs wereproduced GFP-positive embryos at 3 dpf were placed in 24-well plates and maintained in embryonic water containing high concentrations of glucose(130 mM; HG) and candidate drugs (B) At 6 dpf, larvae deposited in 24-well plates werefixed with 4% paraformaldehyde and stored overnight at 4°C.After washing three times for 1 h with distilled water (DW), larvae were incubated for 80 min with 3% trypsin [Tris–HCl (pH 7.8)] at 37°C and washedthree times for 1 h with DW on an orbital shaker at room temperature Using a stereomicroscope, lenses with hyaloid-retinal vessels were detached using
a single channel pipette (P200) by strongly pipetting buffer up and down in the 24-well plates Lenses were transferred to cover glasses in dishes(35 × 10 mm) Images of hyaloid-retinal vessels were observed usingfluorescence stereomicroscopy
BJP S-H Jung et al.
18 British Journal of Pharmacology (2016)173 15–26
Trang 19retinal vessels in 3 samples (Figure 3A) This phenomenon is
thought to be modulated by different individuals In the 7
prep-arations that did show these changes, the inductivity was 75%
(Figure 3B) Under the HG conditions, the overall survival rate
was approximately 80% (Figure 3C) Apart from the changes
in the hyaloid-retinal vessels, there were no obvious differences
in the structural development of the embryos, as assessed by
mi-croscopic observation, between the HG group and the control
group (Figure 3B, C and E) In addition, glucose levels in whole
embryo lysates were measured and found to be much higher
in the HG group than those in the control group (Figure 3D)
Because changes in the size of the retinal vessels could reflect
changes in the size of the lens itself, we measured the lens size in
the HG and control groups As shown in Figure S2A, there was
no significant difference in lens size between these two
experi-mental groups Moreover there was no difference in the number
of OD branches in the HG and control groups (Figure S2B)
As the HG conditions affected the hyaloid-retinal vessels, we
looked for changes in other parts of the vasculature, using
con-focal microscopy With this method we were able to see through
the lens to the hyaloid-retinal vessels in zebrafish larvae and
were able to confirm that HG treatment increased the diameter,
compared with control conditions (Figures S3A, 3B and 3C)
However, the diameters of blood vessels in the body parts
present from the ninth segment to the 14 segment and of the
intersomitic vessels (ISVs) and the dorsal aorta (DA) showed
no significant differences between the HG and control groups(Figures S3A and 3B)
Effects of mannitol and CoCl2on zebrafish hyaloid vessels Next, weassessed the contribution of the osmotic pressure of the HGtreatment (the glucose concentration is 130 mM) to the changes
in the hyaloid-retinal vessels in zebrafish larvae, by using 130mMmannitol instead of glucose for the 3 days from 3-6dpf Asshown in Figure 4, the hyaloid-retinal vessels of mannitol-treatedlarvae were not affected compared with the control vessels,whereas the HG treatment clearly increased the vessel diameter.Another physical change associated with DR is hypoxia(Geoffrey and Sobha, 2011) and hypoxic conditions haveinduced retinopathy in adult zebrafish (Cao et al., 2010) Wetherefore evaluated the effects of hypoxia induced by CoCl2
on the hyaloid-retinal vessels In these experiments, weexposed the zebrafish embryos to 120 μM CoCl2, instead ofglucose, from 3-6dpf and found that this condition of hyp-oxia was as effective as HG in increasing the diameter of thehyaloid-retinal vessels (Figure 4A, B and D)
HG-induced vessel dilation leads to a defect in tight-junctionproteins In DR, pericyte loss and vessel thickening lead tocompromised integrity of the vascular walls, resulting inincreased permeability and changes in tight junctionprotein formation in retinal vessels (Bergers and Song, 2005;
HG-induced changes in hyaloid-retinal vessels in ZF
BJP
Trang 20Pfister et al., 2008) As shown in Figure 5A and D, at 6 dpf, after 3
days of HG treatment, the overall diameter of hyaloid-retinal
vessels in the HG-treated zebrafish larvae was thicker than that
in the control vessels The tight junction protein ZO-1 is an
intracellular adaptor protein that regulates permeability in the
retinal vasculature When we measured expression of this tight
junction protein in hyaloid-retinal vessels, using a ZO-1-specific
antibody, we found that expression of this protein in the vessels
from from HG-treated larvae showed markedly irregular and
discontinuous staining, compared with the staining in control
vessels (Figure 5B and E)
Effects of VEGF signalling and the VEGFR
inhibitor in HG-induced larvae
The mitogenic cytokine VEGF exhibits potent pro-angiogenic
activity and also increases vascular permeability Moreover,
VEGF has been implicated in the pathogenesis of DR throughangiogenesis induced by binding to VEGFR2 in endothelialcells (Sheetz and King, 2002; Caldwell et al., 2003; Scheppke
et al., 2008) We therefore assessed the role of VEGF in ourmodel of DR
First, we examined the effects of inhibiting VEGF signallingusing a VEGFR tyrosine kinase inhibitor (0.5 or 1μM), added tothe HG treatment, from 3 to 6 dpf, of zebrafish larvae As shown
in Figure 6A and 6B, the addition of the VEGF inhibitor blockedthe effects of HG treatment on the vessels and, at the higher con-centration (1μM), treated vessels were identical to those in con-trol larvae Next, we wanted to evaluate the expression of theVegf165 gene in hyaloid-retinal vessels However, due to the smallbody size of zebrafish larvae, it is not possible to isolate specifictissues for tissue-specific mRNA quantifications without notice-able contamination from surrounding tissues So we carried outour evaluation using RT-PCR and mRNAs extracted from whole
Figure 3
Changes in hyaloid-retinal vessel diameter induced by 130 mM glucose(HG) (A) The average diameter andfluorescent images of hyaloid-retinal vessels inHG-treated larvae The diameter of hyaloid-retinal vessels was measured at locations proximal (red circle) to the OD from each group The graph displaysthe mean AU for vessel diameter The vessel diameter of each lens was measured three times ***P< 0.001, significantly different from control, n = 8–10embryos per group Scale bar = 40μm (B) Inductivity for the vessel diameter was increased in the HG-treated group, compared with the control group
***P< 0.001, significantly different from control, n = 8–10 embryos per group (C) Survival rates in HG-treated zebrafish larvae *P < 0.05, significantlydifferent from control, n = 10 embryos per group (D) After HG-treatment for 3 days, the zebrafish displayed increased levels of glucose ***P < 0.001,significantly different from control, n = 20 embryos per group (E) Control and HG-treated larvae did not show gross morphological changes at 6dpf Scalebar = 300μm
BJP S-H Jung et al.
20 British Journal of Pharmacology (2016)173 15–26
Trang 21body lysates As shown in Figure 6C and D, Vegf165 mRNA was
up-regulated in larvae after HG-treatment from 3-6dpf, compared
with control larvae and this increase was clearly prevented by
addition of the VEGFR inhibitor
To further validate our model, we used a monoclonal
anti-body to VEGF, ranibizumab (Lucentis), which has been used
clinically for the treatment of retinal neovascularization This
antibody was added to the HG treatment for 3 days and
substan-tially prevented the increase in vessel diameter induced by HG
treatment alone (Figure 6E) The effects of the ranibizumab were
significant only at the higher concentration used (2.5μg mL-1
;Figure 6F)
NO activation and the effect of L-NAME on the dilation of hyaloid-retinal vessels after
HG-treatmentSignalling by NO is important for regulating blood vessel dila-tion and HG conditions are known to increase NOS expression(Francesco et al., 1997) Zebrafish larvae produce NO, and this
Figure 4
Effects of mannitol and CoCl2on hyaloid vessels of zebrafish larvae [(A)–(D)] To assess possible effects of the hyperosmolarity of the HG treatment,mannitol (130 mM) was added instead of glucose HG-treated larvae (B) showed dilated hyaloid vessels compared to the control (A), whereas hyaloidvessels of the mannitol-treated larvae (C) were not affected (D) The effects of hypoxic conditions on hyaloid vessel development from 3-6 dpf wasstudied by adding CoCl2(120μM) instead of glucose (130 mM) for three days Under these conditions of hypoxia, we found dilated hyaloid vessels,similar to those after HG exposure (B) Scale bar = 40μm (E) Graph data are displayed as the mean AU for vessel diameter The diameter of each lenswas measured three times ###P< 0.001, significantly different from control, n = 16–20 embryos per group
Figure 5
Expression of the tight junction protein ZO-1ion hyaloid-retinal vessels of HG-treated larvae Confocal immunofluorescence microscopy images ofhyaloid-retinal vessels on the dissected lens after treatment with trypsin The overall morphology of hyaloid-retinal vessels was viewed with a confocalmicroscope in control and HG-treated larvae Scale bar = 40μm Hyaloid-retinal vessels in HG-induced larvae were significantly thicker than control[(A) and (D)] The expression of ZO-1 showed irregular and discontinuous staining on hyaloid-retinal vessels from HG-treated larvae (E) compared tocontrol (B) To identify cells, Hoechst nuclear stain was used [(C) and (F)]
HG-induced changes in hyaloid-retinal vessels in ZF
BJP
Trang 22signal can be live imaged in larvae incubated with a DAF-FM DA
probe (Lepiller et al., 2007) In our system, , we exposed the control
and experimental groups of larvae at 6 dpf to DAF-FM DA and
observed the results using confocal microscopy We were able to
identify NO production in the region of the notochord, cleithrum,
and heart (data not shown) Interestingly, in hyaloid-retinal
vessels, NO production was induced in HG-treated larvae but
was not induced in control larvae (Figure 7A) Next, we used an
NO inhibitor,,L-NAME, (10 or 20μM) added to the HG treatmentfor 3 days and found that the hyaloid-retinal vessels from larvaetreated with HG andL-NAME showed decreased vessel diameter,similar to the control group (Figure 7B and C) We also analysedthe expression of mRNA for nos2a in the larvae As shown inFigure 7D and E, mRNA expression of nos2a was increased, relative
to control, in the larvae treated with HG and that this increase wassignificantly reduced by treatment withL-NAME
Figure 6
Vegf expression and the effect of the VEGFR inhibitor and the VEGF antibody ranibizumab in HG-treated larvae (A) The effect of the VEGFR inhibitor on lated hyaloid-retinal vessels Vessel dilation induced by HG was blocked by treatment with the VEGFR tyrosin kinase inhibitor (0.5 or 1μM) Scale bar = 40 μm.(B) The diameter of hyaloid-retinal vessels in HG-treated larvae with VEGFR inhibition was similar to control Graph data are displayed as the mean AU forvessel diameter ###P< 0.001 versus control; **P < 0.01, ***P < 0.001, significantly different from HG, n = 8–10 embryos per group The vessel diameter
di-of each lens was measured three times [(C) and (D)] Expression di-of Vegf in HG-treated larvae increased significantly compared with control and was reduced
by treatment with the VEGFR inhibitor No changes in the transcript levels ofβ-actin were detected between control and HG-treated larvae #P < 0.05,significantly different from control; ***P < 0.001, significantly different from HG (E) The effect of the VEGF antibody, ranibizumab, on dilated hyaloid-retinalvessels HG-induced vessel dilation was morphologically rescued by ranibizumab treatment (2.5μg·mL 1
) Scale bar = 40μm (F) The diameter of retinal vessels in HG-treated larvae with ranibizumab treatment was significantly reduced Graph data are displayed as the mean AU for vesseldiameter ###P< 0.001, significantly different from control; **P < 0.01, significantly different from HG, n = 8–10 embryos per group The vesseldiameter of each lens was measured three times
hyaloid-BJP S-H Jung et al.
22 British Journal of Pharmacology (2016)173 15–26
Trang 23Discussion and conclusions
During the last two decades, models of diabetes have been
devel-oped in many species, including pigs, dogs, cats, rats and mice,
using different inducing agents such as chemicals (STZ or alloxan)
or genetic modifications (Feit-Leichman et al., 2005; Robinson
et al., 2012) Studies of the molecular pathways involved in DR
have used STZ- or alloxan-induced diabetes models or transgenic
and knockout mouse models such as ob/ob and db/db (Midena
et al., 1989; Cheung et al., 2005; Zhang et al., 2007) The retinae
of STZ-induced diabetic animals exhibit biochemical and
physio-logical changes 1–2 months after the onset of hyperglycaemia
This model reproduces early symptoms of DR, such as thickening
of the vascular basement membrane, loss of retinal pericytes andcapillaries, vascular occlusion and increased vascular permeability(Robinson et al., 2012) A recent study suggested an animal modelfor non-proliferative DR in adult zebrafish, in which the zebrafishare subjected to 2% glucose (110 mM) immersion for 30 days(Alvarez et al., 2010) In this model, visual function is diminished,and cone photoreceptors are disrupted Hypoxia-induced retinalvascular disorder in the adult zebrafish model takes 11 days toshow retinopathy and, consequently, to assess therapeutic effects(Cao et al., 2010) In addition, these adult zebrafish models re-quire testing several drug compounds in the plastic containers,and isolation of the retina vessels is time-consuming, requiringsurgical microscissors and forceps resulting in physical stress
Figure 7
NO production by HG in hyaloid-retinal vessels (A) Comparison of NO production using a DAF-FA DA probe in hyaloid-retinal vessels in control larvae at 6 dpf
NO production is not observed in control larvae but is detected in HG-treated larvae (red asterisk) Scale bar = 50μm (B) The effect of the NO inhibitor ondilated hyaloid-retinal vessels HG-induced vessel dilation was rescued by the NOS inhibitor (L-NAME, 10 and 20μM) (C) Graph data are displayed as the mean
AU for vessel diameter The vessel diameter of each lens was measured three times Scale bar = 40μm ###P < 0.001 versus control; **P < 0.01, ***P < 0.001,significantly different from HG, n = 10 embryos per group [(D) and (E)] Expression of nos2a in HG-treated larvae increased significantly compared to controland reduced by treatment withL-NAME No changes in the transcript levels ofβ-actin were detected between control and HG-treated larvae ###P < 0.001,significantly different from control; *P < 0.05, **P < 0.01, significantly different from HG
HG-induced changes in hyaloid-retinal vessels in ZF
BJP
Trang 24Recently, Wang et al (2013) reported that combination of a
high cholesterol diet and immersion in a 3% glucose (166 mM)
solution applied to 5 to 15 dpf zebrafish larvae could be used as
a model for diabetic vasculopathy Earlier studies showed that
1 dpf zebrafish larvae could be exposed to 30 mM glucose and
drugs for to 6 days (Lee et al., 2013; Yu et al., 2013) In the present
study, we have attempted to optimize and characterize a
short-term in zebrafish to study diabetic retinal vascular dysfunction
and provide a system for rapid drug screening We induced
hyperglycaemia by immersing 3 dpf zebrafish larvae for 3 days
in 130 mM glucose (HG; Figures 2 and 3) Although there were
clear changes in the hyaloid-retinal vessels, the lens size and
number of OD branches were not affected (Figure S2) Moreover,
other bloods vessels such as ISVs and DA did not show any
differences between the control and HG groups (Figure S3)
In the present model, the hyaloid-retinal vessels were
analysed after only 3 days incubation in 24-well plates This
pro-cedure provides two advantages, a low incubation volume means
that less drug is needed per test and the 3 days is clearly much
shorter than 11 (Cao et al., 2010) or 30 days (Alvarez et al.,
2010), as used previously The structural changes in the
hyaloid-retinal vessels after HG were not osmotic events but were
influ-enced by in vivo glucose metabolism in zebrafish larvae Gleeson
et al (2007) and Wang et al (2013) reported that larvae treated
with HG regulate glucose metabolism through direct secretion
of insulin, phosphoenolpyruvate carboxykinase, somatostatin
and glucagon into the bloodstream These results suggest that
HG-induced changes in zebrafish larvae hyaloid-retinal vessels
closely mimic the molecular mechanism for human DR
Reduction and redistribution of tight junction proteins in
retinal endothelial cells under hyperglycaemic conditions
increases vascular permeability (Antonetti et al., 1998) Kim
et al (2011) did not observe occludin and ZO-1 expression
on retinal vessels when viewed on whole mounts of frozen
sections with immunofluorescence staining in zebrafish
However, using our isolation method and quantitative
analy-ses (Figure 1 and S1), we were able to demonstrate expression
of the tight junction protein ZO-1 and show that its
expres-sion was irregular or discontinuous on hyaloid-retinal vessels
from HG-treated larvae (Figure 5)
At present, VEGF constitutes an exciting target for
thera-peutic intervention in DR VEGF-induced angiogenesis and
vascular hyperpermeability are mediated by NO (Van der Zee
et al., 1997) Also NO was an essential downstream
compo-nent of VEGF-induced angiogenesis in vivo and NOS
inhibi-tors blocked the VEGF-induced proliferation of endothelial
cells (Shizukuda et al., 1999) In the present experiments, we
have confirmed that this model was sensitive to VEGF-related
and NO-related interventions Although early (0–1 dpf)
expo-sure to a VEGFR inhibitor induced severe developmental
defects, including heart and eye oedema, short body and
de-velopmental retardation (data not shown), later application,
at 3 to 6 dpf, blocked the increase in diameter of retinal
vessels, induced by HG treatment (Figure 6A and B)
As 75% of the protein sequence in zebrafish VEGFAA is
similar to that of human VEGF-A 165 isoform (Wu et al., 2015)
and the recombinant humanized monoclonal IgG antibody
for VEGF, ranibizumab, has been used to treat human DR by
blocking angiogenesis, it was reasonable to test ranibizumab in
our model We also assessed NO production with afluorescent
indicator dye (DAF-FA DA) and the expression of NOS mRNA,
in our model These results and those after application of theNOS inhibitor L-NAME together demonstrated clearly that
NO was an important mediator of the structural changes inthe hyaloid-retinal vessels of zebrafish larvae following HGtreatment Overall, our results suggest that NO and VEGF inHG-treated zebrafish larvae display regulatory patterns andactivities, very similar to those in mammals
Although several animal models for DR are well established,there are no short-term in vivo models to test the effects of can-didate therapeutic compounds In this study, we have described
a novel, short-term, in vivo screening method for compoundsaffecting DR, using zebrafish larvae exposed to HG conditions
In this model, dilation of hyaloid-retinal vessels was nied by morphological lesions with disruption of tight junctionproteins, overproduction of Vegf mRNA and NO production Wealso showed that the VEGFR tyrosine kinase inhibitor, the VEGFantibody ranibizumab and inhibition of endogenous NO syn-thesis could all decrease or reverse the HG-induced dilation ofhyaloid-retinal vessels These results suggest that this zebrafishmodel system will be a powerful novel tool for the screening
accompa-of therapeutic drug candidates for DR
Con flict of interest
The authors declare they have no conflict of interest
ReferencesAlexander SPH, Benson HE, Faccenda E, Pawson AJ, Sharman JL,Spedding M et al (2013a) The Concise Guide to PHARMACOLOGY2013/14: Catalytic Receptors Br J Pharmacol 170: 1676–1705.Alexander SPH, Benson HE, Faccenda E, Pawson AJ, Sharman JL,Spedding M et al (2013b) The Concise Guide to PHARMACOLOGY2013/14: Enzymes Br J Pharmacol 170: 1797–1867
Alvarez Y, Cederlund ML, Cottell DC, Bill BR, Ekker SC, Torres-Vazquez J
et al (2007) Genetic determinants of hyaloid and retinal vasculature inzebrafish BMC Dev Biol 7: 114–130
Alvarez Y, Chen K, Reynolds AL, Waghorne N, O’Connor JJ, Kennedy
BN (2010) Predominant cone photoreceptor dysfunction in ahyperglycaemic model of non-proliferative diabetic retinopathy DisModel Mech 3: 236–245
BJP S-H Jung et al.
24 British Journal of Pharmacology (2016)173 15–26
Trang 25Abu El-Asrar AM, Meersschaert A, Dralands L, Geboes K (2004) Inducible
nitric oxide synthase and vascular endothelial growth factor are colocalized
in the retinas of human subjects with diabetes Eye 18: 306–313
Antonetti DA, Barber AJ, Khin S, Lieth E, Tarbell JM, Gardner TW
(1998) Vascular permeability in experimental diabetes is associated
with reduced endothelial occludin content: vascular endothelial
growth factor decreases occludin in retinal endothelial cells Penn
State Retina Res Group Diab 47: 1953–1959
Armulik A, Abramsson A, Betsholtz C (2005) Endothelial/pericyte
interactions Circ Res 97: 512–523
Barros TP, Alderton WK, Reynolds HM, Roach AG, Berghmans S (2008)
Zebrafish: an emerging technology for in vivo pharmacological
assessment to identify potential safety liabilities in early drug discovery
Br J Pharmacol 154: 1400–1413
Bergers G, Song S (2005) The role of pericytes in blood-vessel
formation and maintenance Neuro Oncol 7: 452–464
Calcutt NA, Cooper ME, Kern TS, Schmidt AM (2009) Therapies for
hyperglycaemia-induced diabetic complications: from animal
models to clinical trials Nat Rev Drug Discov 8: 417–429
Caldwell RB, Bartoli M, Behzadian MA, EI-Remessy AE, AI-Shabrawey
M, Platt DH et al (2003) Vascular endothelial growth factor and
diabetic retinopathy: pathophysiological mechanisms and treatment
perspectives Diabetes Metab Res Rev 19: 442–455
Cao Z, Jensen LD, Rouhi P, Hosaka K, Lanne T, Steffensen JF et al
(2010) Hypoxia-induced retinopathy model in adult zebrafish Nat
Protoc 5: 1903–1910
Checchin D, Sennlaub F, Levavasseur E, Leduc M, Chemtob S (2006)
Potential role of microglia in retinal blood vessel formation Invest
Ophthalmol Vis Sci 47: 3595–3602
Cheung AK, Fung MK, Lo AC, Lam TT, So KF, Chung SS et al (2005)
Aldose reductase deficiency prevents diabetes-induced blood-retinal
barrier breakdown, apoptosis, and glial reactivation in the retina of
db/db mice Diabetes 54: 3119–3125
Elo B, Villano CM, Govorko D, White LA (2007) Larval zebrafish as a
model for glucose metabolism: expression of phosphoenolpyruvate
carboxykinase as a marker for exposure to anti-diabetic compounds
J Mol Endocrinol 38: 433–440
Engerman RL, Kern TS (1995) Retinopathy in animal models of
diabetes Diabetes Metab Rev 11: 109–120
Feit-Leichman RA, Kinouchi R, Takeda M, Fan Z, Mohr S, Kern TS et al
(2005) Vascular damage in a mouse model of diabetic retinopathy: relation
to neuronal and glial changes Invest Ophthalmol Vis Sci 46: 4281–4287
Fong DS, Aiello L, Gardner TW, King GL, Blankenship G, Cavallerano
JD et al (2003) Diabetic retinopathy Diabetes Care 26: 226–229
Francesco C, Keiichi H, Zvonimir SK, Thomas FL (1997) High glucose
increases nitric oxide synthase expression and superoxide anion
generation in human aortic endothelial cells Circulation 96: 25–28
Geoffrey BA, Sobha S (2011) Hypoxia and oxidative stress in the
causation of diabetic retinopathy Curr Diabetes Rev 7: 291–304
Gleeson M, Connaughton V, Arneson LS (2007) Induction of
hyperglycaemia in zebrafish (Danio rerio) leads to morphological
changes in the retina Acta Diabetol 44: 157–163
Greene DA, Arezzo JC, Brown MB (1999) Effect of aldose reductase
inhibition on nerve conduction and morphometry in diabetic
neuropathy Zenarestat Study Group Neurology 53: 580–591
Guillemin GJ, Brew BJ (2004) Microglia, macrophages, perivascular
macrophages, and pericytes: a review of function and identification
J Leukoc Biol 75: 388–397
Jorgens K, Hillebrands JL, Hammes HP, Kroll J (2012) Zebrafish: amodel for understanding diabetic complications Exp ClinEndocrinol Diabetes 120: 186–187
Joussen AM, Poulaki V, Qin W, Kirchhof B, Mitsiades N, Wiegand SJ
et al (2002) Retinal vascular endothelial growth factor inducesintercellular adhesion molecule-1 and endothelial nitric oxidesynthase expression and initiates early diabetic retinal leukocyteadhesion in vivo Am J Pathol 2: 501–509
Jung SH, Kim S, Chung AY, Kim HT, So JH, Ryu J et al (2010) Visualization
of myelination in GFP-transgenic zebrafish Dev Dyn 2: 592–597.Kempen JH, O’Colmain BJ, Leske MC, Haffner SM, Klein R, Moss SE et al.(2004) The prevalence of diabetic retinopathy among adults in theUnited States Arch Ophthalmol 122: 552–563
Kilkenny C, Browne WJ, Cuthill IC, Emerson M, Altman DG (2010).Animal research: Reporting in vivo experiments: The ARRIVEguidlines Br J Pharmacol 160: 1577–1579
Kim JH, Yu YS, Kim KW, Kim JH (2011) Investigation of barriercharacteristics in the hyaloid-retinal vessel of zebrafish J NeurosciRes 89: 921–928
Kitambi SS, McCulloch KJ, Peterson RT, Malicki JJ (2009) Smallmolecule screen for compounds that affect vascular development inthe zebrafish retina Mech Dev 126: 464–477
Lakshminarayanan S, Antonetti DA, Gardner TW, Tarbell JM (2000).Effect of VEGF on retinal microvascular endothelial hydraulicconductivity: the role of NO Invest Ophthalmol Vis Sci 41: 4256–4261.Lee IS, Yu SY, Jung SH, Lee YR, Lee YM, Kim JH et al (2013)
Proanthocyanidins from Spenceria ramalana and their effects on AGEformation in vitro and hyaloid-retinal vessel dilation in larvalzebrafish in vivo J Nat Prod 76: 1881–1888
Lepiller S, Laurens V, Bouchot A, Herbomel P, Solary E, Chluba J (2007).Imaging of nitric oxide in a living vertebrate using a diamino-fluoresceinprobe Free Radic Biol Med 43: 619–627
Liang J, Gui Y, Wang W, Gao S, Li J, Song H (2010) Elevated glucoseinduces congenital heart defects by altering the expression of tbx5,tbx20, and has2 in developing zebrafish embryos Birth Defects Res AClin Mol Teratol 88: 480–486
McGrath JC, Drummond GB, McLachlan EM, KilKenny C,Wainwright CL (2010) Guidelins for reporteing experimentsinvolving animals: the ARRIVE guidlines Br J Pharmacol 160:
1573–1576
Midena E, Segato T, Radin S, di Giorgio G, Meneghini F, Piermarocchi S
et al (1989) Studies on the retina of the diabetic db/db mouse I.Endothelial cell-pericyte ratio Ophthalmic Res 21: 106–111
Morbidelli L, Chang CH, Douglas JG, Granger HJ, Ledda F, Ziche M(1996) Nitric oxide mediates mitogenic effect of VEGF on coronaryvascular endothelium Am J Physiol 270: H411–415
Nusslein VC, Dahm R (eds) (2002) Zebrafish: A Practical Approach.Oxford University Press: Oxford, UK
Pawson AJ, Sharman JL, Benson HE, Faccenda E, Alexander SP,Buneman OP et al (2014) The IUPHAR/BPS Guide toPHARMACOLOGY: an expert-driven knowledge base of drug targetsand their ligands Nucl Acids Res 42 (Database Issue): D1098–1106.Pfister F, Feng Y, vom Hagen F, Hoffmann S, Molema G, Hillebrands JL
et al (2008) Pericyte migration: a novel mechanism of pericyte loss
in experimental diabetic retinopathy Diabetes 57: 2495–2502.Robinson R, Barathi VA, Chaurasia SS, Wong TY, Kern TS (2012) Update
on animal models of diabetic retinopathy: from molecular approaches tomice and higher mammals Dis Model Mech 5: 444–456
HG-induced changes in hyaloid-retinal vessels in ZF
BJP
Trang 26Roy MS, Klein R, O’Colmain BJ, Klein BE, Moss SE, Kempen JH
(2004) The prevalence of diabetic retinopathy among adult type 1
diabetic persons in the United States Arch Ophthalmol 122:
546–551
Scheppke L, Aguilar E, Gariano RF, Jacobson R, Hood J, Doukas J et al
(2008) Retinal vascular permeability suppression by topical
application of a novel VEGFR2/Src kinase inhibitor in mice and
rabbits J Clin Invest 118: 2337–2346
Sheetz MJ, King GL (2002) Molecular understanding of
hyperglycemia’s adverse effects for diabetic complications JAMA
288: 2579–2588
Shizukuda Y, Tang S, Yokota R, Ware JA (1999) Vascular endothelial
growth factor-induced endothelial cell migration and proliferation
depend on a nitric oxide-mediated decrease in protein kinase Cδactivity
Circ Res 85: 247–256
Starita C, Patel M, Katz B, Adamis AP (2007) Vascular endothelial
growth factor and the potential therapeutic use of pegaptanib
(macugen) in diabetic retinopathy Dev Ophthalmol 39: 122–148
Van der Zee R, Murohara T, Luo Z, Zollmann F, Passeri J, LeKutat C et al
(1997) Vascular endothelial growth factor/vascular permeability
factor augments nitric oxide release from quiescent rabbit and
human vascular endothelium Circulation 95: 1030–1037
Wang Z, Mao Y, Cui T, Tang D, Wang XL (2013) Impact of a combined
high cholesterol diet and high glucose environment on vasculature
PLoS One 12: e81485
Wu YC, Chang CY, Kao A, Hsi B, Lee SH, Chen YH et al (2015)
Hypoxia-induced retinal neovascularization in zebrafish embryos: a potential
model of retinopathy of prematurity PLoS One 15: e0126750
Yu SY, Lee IS, Jung SH, Lee YM, Lee YR, Kim JH et al (2013)
Caffeoylated phenylpropanoid glycosides from Brandisia hancei
inhibit advanced glycation end product formation and aldose
reductase in vitro and vessel dilation in larval zebrafish in vivo Planta
Med 79: 1705–1709
Zhang H, Sonoda KH, Qiao H, Oshima T, Hisatomi T, Ishibashi T (2007).Development of a new mouse model of branch retinal vein occlusionand retinal neovascularization Jpn J Ophthalmol 51: 251–257
Supporting Information
Additional Supporting Information may be found in the onlineversion of this article at the publisher’s web-site:
http://dx.doi.org/10.1111/bph.13279Figure S1Diagram depicting measurement of the hyaloid-retinal vessels diameter (A) Lens in a dish with a coverglass.(B) Lens alignments under thefluorescence microscope Theoptic disc (OD) was displayed up (C) Fluorescence micros-copy images (D) The diameter of hyaloid vessels was mea-sured in three main branches (red arrow) from the OD intothefirst branch (~30 μm) using Image J software
Figure S2Morphological effects on the patterning of thehyaloid vessels and lens size by HG (A) The size of lenses werenormal in control and HG-treated larvae Control larvae,n=38; HG-treated larvae, n=40 Scale bar = 40μm (B) Thenumber of main branches from the optic disc (OD) wascounted in control and glucose-treated larvae Control larvae,n=76; HGtreated larvae, n=72 Scale bar = 40μm
Figure S3Repersentative images of hyaloid-retinal vessels andtrunk vessels in 6 dpf zebrafish (A,A`,B,B`) There were no differ-ences in intersomite vessels (ISVs) and dorsal aorta (DA) betweenthe control (A, A`) and HG-treated groups (B, B`) 250´ magnifica-tion, n=4 in each group Scale bar = 50μm (C-E) The graph displaysthe mean artificial unit (AU) for diameter of ISVs and DA Thevessel diameter of each region was measured three times Theexperiment was repeated triplicate *P< 0.05 vs control
BJP S-H Jung et al.
26 British Journal of Pharmacology (2016)173 15–26
Trang 27RESEARCH PAPER
human GIP receptors, but a
partial agonist and
competitive antagonist at rat
and mouse GIP receptors
A H Sparre-Ulrich1,2†, L S Hansen1,2,4†, B Svendsen2, M Christensen4,
F K Knop4, B Hartmann2,3, J J Holst2,3and M M Rosenkilde1
1Department of Neuroscience and Pharmacology, Faculty of Health and Medical Sciences, The
Panum Institute, University of Copenhagen, Copenhagen, Denmark,2NNF Center for Basic
Metabolic Research, Copenhagen, Denmark,3Department of Biomedical Sciences Faculty of Health
and Medical Sciences, University of Copenhagen, Copenhagen, Denmark, and4Center for Diabetes
Research, Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
BACKGROUND AND PURPOSE
Specific, high potency receptor antagonists are valuable tools when evaluating animal and human physiology Within theglucose-dependent, insulinotropic polypeptide (GIP) system, considerable attention has been given to the presumed GIP re-ceptor antagonist, (Pro3)GIP, and its effect in murine studies We conducted a pharmacological analysis of this ligand includinginterspecies differences between the rodent and human GIP system
EXPERIMENTAL APPROACH
Transiently transfected COS-7 cells were assessed for cAMP accumulation upon ligand stimulation and assayed in competitionbinding using125I-human GIP Using isolated perfused pancreata both from wild type and GIP receptor-deficient rodents, insulin-releasing, glucagon-releasing and somatostatin-releasing properties in response to species-specific GIP and (Pro3)GIP analogueswere evaluated
CONCLUSIONS AND IMPLICATIONS
When evaluating new compounds, it is important to consider interspecies differences both at the receptor and ligand level Thus,
in rodent models, human GIP is a comparatively weak partial agonist Human (Pro3)GIP was not an antagonist at human GIPreceptors, so there is still a need for a potent antagonist in order to elucidate the physiology of human GIP
Abbreviations
GIP, glucose-dependent insulinotropic polypeptide (gastric inhibitory peptide); GLP-1, glucagon-like peptide-1; HBS,HEPES-buffered saline
BJP British Journal of Pharmacology www.brjpharmacol.org
© 2015 The Authors British Journal of Pharmacology
published by John Wiley & Sons Ltd on behalf of British Pharmacological Society.
British Journal of Pharmacology (2016) 173 27–38 27
Trang 28Glucose-dependent insulinotropic polypeptide (also gastric
inhibitory peptide; GIP) is a hormone secreted from the K
cells of the gut following a meal (Baggio and Drucker,
2007) Like the related hormone glucagon-like peptide-1
(GLP-1), GIP is a potent insulin secretagogue (Holst, 2004)
In contrast to the glucagonostatic effect of GLP-1 (Gutniak
et al., 1992; de Heer et al., 2008), GIP has glucagon-releasing
properties (Kreymann et al., 1987; Christensen et al., 2011)
The incentive to understand the biology of GIP was
intensi-fied by the discovery of an association between rodent GIP
receptors and adiposity (Miyawaki et al., 1999; Miyawaki
et al., 2002; Nasteska et al., 2014) In humans, although less
clear, there is likewise evidence for a role of GIP in fat
metab-olism with the demonstration of GIP receptor expression in
adipose tissue (Ahlqvist et al., 2013), an association between
high body mass index and increased GIP levels (Ahlqvist
et al., 2013; Calanna et al., 2013), and increased adipose
tis-sue bloodflow and triacylglycerol deposition following GIP
administration in a state of high insulin and high glucose
(Asmar et al., 2010) Furthermore, obese children decrease
their basal and postprandial GIP levels, when put on a
low-calorie diet (Deschamps et al., 1980), and healthy young
men increase their fasting GIP levels, when put on a
high-fat diet (Brøns et al., 2009)
The availability of the GLP-1 receptor antagonist
exendin-(9–39) (Raufman et al., 1991; Jørgensen et al., 2013)
has been helpful for our understanding of the physiology of
GLP-1, which underlies the pharmaceutical development of
GLP-1 receptor agonists as anti-diabetic and anti-obesity
agents Inspired by this, researchers have tried to develop a
potent GIP receptor antagonist Many different strategies
have been undertaken in order to inhibit GIP function,
including prepro-GIP gene truncations (Nasteska et al.,
2014), administration of low MW receptor antagonists
(Nakamura et al., 2012), immunization against GIP (Ebert
et al., 1979; Fulurija et al., 2008; Irwin et al., 2009), various
truncations and amino acid substitutions of the GIP molecule
thought to provide antagonistic properties (Tseng et al., 1996;
Gelling et al., 1997; Hinke et al., 2001; Gault et al., 2002;
Deacon et al., 2006; Irwin et al., 2006; Kerr et al., 2011), and
recently, a potent antagonistic antibody against the GIP
receptor was reported (Ravn et al., 2013) However, most
attention has been given to the analogue (Pro3)GIP
follow-ing demonstrations that chronic treatment with this
peptide improved diabetic parameters in ob/ob mice (Irwin
et al., 2007), improved diabetic parameters and inducedweight loss in obese mice previously on a high-fat diet(McClean, 2007), and reduced weight gain and improveddiabetic parameters following the induction of a high-fatdiet (Gault et al., 2007)
Animal models are widely used as basis for predicting logical properties of given receptor-ligand systems to human(patho)physiology Often, rodent models are thefirst choicedue to reduced costs and space requirements compared withlarger animal models (pigs, dogs and primates) Initial drugscreening are similarly often carried out in rodent models.This creates the classical dilemma of whether the chosenrodent model is representative for human physiology orwhether the inter-species differences (stemming from struc-tural, functional, spatial or temporal differences) are too great.Among the seven-transmembrane receptor systems, such dif-ferences have been described for instance for the lipid-activated G protein-coupled receptor 119 (GPR119), where aseries of compounds, with similar binding properties to hu-man and mouse GPR119, displayed large differences in po-tency and efficacy (Scott et al., 2013) A similar example can
bio-be found within the 5-HT receptor system, where a singleamino acid difference between the human and mouse recep-tor can account for large differences in binding affinities andpotencies (Canal et al., 2013)
Other receptor-ligand systems, however, display very tle interspecies variations, as seen in the GLP-1 system with
lit-a llit-arge degree of sequence homology both in terms of theligand (GLP-1) and its receptor in humans, mice and ratsand where both liraglutide (a long-acting GLP-1 receptor ago-nist) and native GLP-1 have similar potencies across species-specific receptors (Knudsen et al., 2012) (Figure 1) Eventhough the GIP system is closely related to GLP-1 and theGLP-1 receptor, the GIP system is noticeably less conservedacross species (Figure 1)
In an attempt to develop GIP receptor antagonists to beused in humans, for instance, as a putative therapeuticagainst obesity (Asmar et al., 2010; Ahlqvist et al., 2013), weset out to investigate functional differences between rodentsand human within the GIP system in terms of ligand bindingand signalling properties As agonists, we chose the endoge-nous agonist GIP(1–42) from human, rat and mouse, andcharacterized this in parallel with the species-correspondingpreviously described antagonist (Pro3)GIP (Gault et al.,2002) on all three GIP receptors (from human, rat and
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 (Pawsonet al., 2014)and are permanently archived in the Concise Guide to PHARMACOLOGY 2013/14 (Alexander et al., 2013)
BJP A H Sparre-Ulrich et al.
28 British Journal of Pharmacology (2016) 173 27–38
Trang 29mouse) The goal was to determine whether the beneficial
an-tagonistic effects of (Pro3)GIP in rodents could possibly be
transferred to humans
Methods
Animals
All animal care and experimental procedures complied with
institutional guidelines and were approved by the Danish
Animal Experiments Inspectorate (2013-15-2934-00833)
Studies involving animals are reported in accordance with
the ARRIVE guidelines for reporting experiments involving
animals (Kilkenny et al., 2010; McGrath et al., 2010) A total
of 18 animals were used in the experiments described here
Male C57Black/6 (B6) mice (25–30 g) or male Wistar rats
(220–250 g) were purchased from Taconic (Lille Skensved,
Denmark) The animals were housed in plastic-bottomed,
wire-lidded cages in air-conditioned (21C) and
humidity-controlled (55%) rooms with a 12:12 h light–dark cycle
and free access to standard rat chow and water Animals
were acclimatized for at least 1 week before use
Transfections and tissue culture
COS-7 cells (ATCC, Virginia, USA) were cultured at 10% CO2
and 37 °C in DMEM 1885 supplemented with 10% FBS,
2 mM glutamine, 180 units mL 1 penicillin and 45 g mL 1
streptomycin Transient transfection of the COS-7 cells for
cAMP accumulation and competition binding was performed
using the calcium phosphate precipitation method with the
addition of chloroquine (Kissow et al., 2012)
cAMP assay
Transiently transfected COS-7 cells were seeded out in white
96-well plates at a density of 3 * 104cells per well The day after, the
cells were washed twice with HEPES-buffered saline (HBS) buffer
and incubated with HBS and 1 mM IBMX for 30 min at 37 °C To
test agonists, ligands were added and incubated for 30 min at
37 °C In order to test for antagonistic properties, the antagonistwas pre-incubated for 10 min, and then the agonist was addedand incubated for an additional 20 min The HitHunterTMcAMP XS assay (DiscoveRx, Herlev, Denmark) was carried out ac-cording to the manufacturer’s instructions
Competition binding assayCOS-7 cells were seeded in clear 96-well plates the day aftertransient transfection The number of cells seeded per well,which is determined by the apparent expression efficiency
of the receptor, was aimed to result in 5–10% specific binding
of the added radioactive ligand (125I-human GIP) The ing day, cells were assayed by competition binding for 4 h at
follow-4 °C using 15–40 pM125
I-human GIP as well as unlabelledligand in 50 mM HEPES buffer (pH 7.2) supplemented with0.5% BSA (binding buffer) After incubation, the cells werewashed twice in ice-cold binding buffer and lysed using
200 mM NaOH with 1% SDS for 30 min Nonspecific bindingwas determined as the binding in the presence of 100 nMunlabelled human GIP The samples were analysed by theWallac Wizard 1470 Gamma Counter
Rat or mouse isolated perfused pancreasMale C57B6 mice (25–30 g) or male Wistar rats (220–250 g) wereanaesthetized (mice: ketamine/xylazine 100/10 mgkg 1
i.p.;rats: 0.0158 mg fentanyl citrate + 0.5 mgfluanisone + 0.25 mgmidazolam/100 g; Pharmacy Service, Denmark), and the pan-creas was dissected and perfused in situ as described previously(Deacon et al., 2006) Briefly, the pancreas was perfused in asingle-pass system through both the coeliac and the superiormesenteric artery via a catheter inserted into the aorta All otheraortic branches were ligated The venous effluent was collectedfor 1 min intervals via a catheter in the portal vein, and stored
at 20 °C until analysis The pancreas was perfused with amodified Krebs Ringer bicarbonate buffer [composition:
fu-marate, 5 mM pyruvate, 5 mM glutamate, 7 mM glucose,
-1 mouse
Figure 1
The GIP system is less conserved than the GLP-1 hormone system A) Sequence identity between rodent and human GIP receptors (GIPR) and GLP-1receptors (GLP-1R), as well as GIP and GLP-1 given in percent The receptors were compared without signalling peptide sequences The alignment wasdone in Geneious 6.0.5 B) Sequence alignment between human, rat, and mouse GIP (top) and GLP-1 (bottom) Grey indicates a mismatch and the blackspiral indicates predicted alpha helix structure
Species-specific activity of (Pro3)GIP
BJP
Trang 300.1% BSA, 5% dextran (Pharmacosmos, Holbaek, Denmark)].
Flow rate was kept constant at 1.5 mLmin 1
(mouse) or
4 mLmin 1
(rat), perfusion buffer was heated and
oxygen-ated (95% O2, 5% CO2), and pressure was continuously
measured throughout the experiment Rodent (rat/mouse)
forms of (Pro3)GIP and of GIP were infused as test
sub-stances through a sidearm infusion pump at a flow rate of
(rat) Arginine(10 mM) was infused at the end of each experiment as a
positive control
Hormone analysis
Hormone concentrations in the perfusion effluent were
measured using in-house radioimmunoassays Glucagon
was measured using a midregion-directed glucagon
rum (code no 4304) or a COOH-terminally directed
antise-rum (code no 4305), which measures fully processed
glucagon as well as N-terminally extended molecular forms, in
addition, a synthetic glucagon for standards, and 125I-labelled
glucagon (a gift from Novo Nordisk A/S) as tracer (Orskov
et al., 1991) Insulin was measured using an antibody
cross-reacting strongly with rat insulin I and II (code no 2006–3)
As the standard, we used human insulin, and the tracer was
125
I-labelled human insulin (gift from Novo Nordisk A/S)
(Brand et al., 1995) Somatostatin concentrations were
deter-mined using a rabbit antiserum (code no 1758) raised against
synthetic cyclic somatostatin, recognizing both somatostatin
14 and somatostatin 28 (Baldissera et al., 1983), somatostatin
14 as standard and 125I-labelled Tyr11-somatostatin (NEX389,
Perkin-Elmer, Skovlunde, Denmark) as tracer
Sequence alignments
The amino acid sequences of the GIP and GLP-1 systems were
acquired from GenBank of NCBI The alignment was
per-formed in Geneious 6.0.5 using MAFFT v6.814b The
BLOSUM62 matrix was applied with gap open penalty and
offset value of 1.53 and 0.123 respectively
Data analysis
Pharmacological analyses including Schild plot analyses and
statistical analyses were carried out with the GraphPad Prism
6.0 software (GraphPad, San Diego, CA, USA) and Microsoft
ExcelTM Sigmoid curves werefitted logistically with a Hillslope
of 1.0 The calculations of Kivalues were based on the
Cheng-Prusoff formula (DeBlasi et al., 1989) Dose ratios (DR) for the
Schild analyses (Lazareno and Birdsall, 1993) were based on
the potency shift of GIP(1–42) in the presence of a given
antag-onist concentration, relative to the absence of antagantag-onist
Materials
Rat and mouse (Pro3)GIP (cat no 027–29 and 027–49,
re-spectively) and rat and mouse GIP (cat no 027–12 and
027–27, respectively) were purchased from Phoenix
Phar-maceuticals (Karlsruhe, Germany) Human (Pro3)GIP was
synthesized by CASLO ApS (Lyngby, Denmark) and human
GIP was purchased from Bachem (H5645: Bubendorf,
Switzerland) All peptides had a purity of more than 95%
by HPLC analyses and an MS-controlled molecular weight
purchased from Origene (Rockville, MD, USA) (SC110906,RN212314 and MC216211, respectively) and cloned into
purchased from PerkinElmer Life Sciences (Skovlunde,Denmark; NEX402)
(Pro3)GIP is an agonist with efficacies dependent on the species providing GIP receptors
Due to the reported antagonistic properties and the effects served in obese and/or diabetic animal models (Gault et al.,2007; Irwin et al., 2007; McClean, 2007), the Pro3-analogues wereinvestigated on the human, rat and mouse GIP receptors, in par-allel with the respective endogenous GIP agonists As GIPreceptor-induced cAMP accumulation is well established as an im-portant signalling pathway for GIP function, the effects ofspecies-specific GIP and (Pro3)GIP analogues were evaluated bymeasuring cAMP accumulation Compared with human GIP, hu-man (Pro3)GIP was an efficacious agonist on the human GIP re-ceptor approaching the Emaxof human GIP (Figure 2A), whileboth rat (Pro3)GIP (Figure 2B) and mouse (Pro3)GIP (Figure 2C)were partial agonists with Emaxvalues of ~50 and ~30%, respec-tively, on their respective receptors In addition to the differ-ences in efficacy, there was a dramatic shift in potencybetween GIP and the corresponding (Pro3)GIP within the threespecies On the human GIP receptor, we observed 181-fold dif-ference between human GIP and human (Pro3)GIP, while muchlarger differences (1300-fold and 2636-fold in rat and mouse, re-spectively) were observed in the rodent systems These differ-ences were due to both higher species-specific GIP potencies(from 26 pM for human GIP on human GIP receptors to 10and 11 pM for rat and mouse GIP on their GIP receptors) andlower (Pro3)GIP species-specific potencies (from 4.7 nM on hu-man GIP receptors to 13 and 29 nM on rat and mouse GIP recep-tors respectively)
ob-(Pro3)GIP is a partial agonist with competitive antagonistic properties in rodent GIP receptors
To determine the potential of the two partial agonists (rat andmouse (Pro3)GIP) as antagonists of GIP-induced activation,
BJP A H Sparre-Ulrich et al.
30 British Journal of Pharmacology (2016) 173 27–38
Trang 31cAMP production was measured as a function of increasing
concentration of rat and mouse GIP in the absence or
pres-ence of various concentrations of rat and mouse (Pro3)GIP
on the corresponding GIP receptors (Figure 3A and B
respectively) Reflected by the agonistic properties, (Pro3)
GIP increased the cAMP production in the absence or at low
GIP concentrations on both receptors However, a
concomi-tant rightward shift in potency of GIP was observed, which
is an indication of a competitive antagonistic nature At 10,
100, and 1000 nM of rat (Pro3)GIP, the potency of rat GIP
was shifted 2-fold, 4-fold and 16-fold compared with the
absence of (Pro3)GIP (Figure 3A) Using the calculated EC50
values from these curves, a Schild plot analysis was made
(Figure 3C) The Hill coefficient was 0.55 ± 0.20, and the
X-axis intercept, which represents the dissociation
con-stants (Ki) for rat (Pro3)GIP, was 13 nM A similar pattern
was observed in the mouse system Here, the highest
concen-tration of mouse (Pro3)GIP (1 uM) resulted in an 11-fold
right-shift in the potency of mouse GIP The Hill coefficient
in the Schild Plot in the mouse system was 0.79 ± 0.16 and
Kiof rat (Pro3)GIP was calculated to be 61 nM (Figure 3D)
As the Hill coefficient was <1 in both cases, this indicates
confounding factors, presumably determined by the
con-comitant (partial) agonist and antagonist effects of (Pro3)GIP
Lower activities of all three (Pro3)GIP in the
rodent GIP systems compared with the human
system
To determine whether the species-specific differences of
(Pro3)GIPs were due to ligand or receptor differences, we
investigated the effects of the three (Pro3)GIP ligands
across all three GIP receptors The approaching full
agonism as observed for human (Pro3)GIP at the human
GIP receptor was not seen with the two rodent (Pro3)GIP
li-gands, which both demonstrated an Emaxof ~70% as
com-pared with human GIP (Figure 4A) In contrast, the (Pro3)
GIP analogues were equally potent at the human GIP
re-ceptor (Table 1) At the rat GIP rere-ceptor, there was a lower
efficacy of all three (Pro3)GIP analogues, with human
(Pro3)GIP again showing the highest efficacy (67 vs
~40%), but with similar potencies for the three (Pro3)GIPanalogues (Figure 4B, Table 1) The same tendency was seen
at the mouse GIP receptor with an even lower efficacy ofhuman (Pro3)GIP (41%) and rodent (Pro3)GIP (~30%) com-pared with mouse GIP (Figure 4C)
Higher activities of GIP in the rodent GIP system as compared with the human systemInspired by thefindings that the glutamic acid to proline sub-stitution at the third position in GIP resulted in a lower activ-ity in the rodent systems, we decided to examine whethersuch species-dependent differences were present for nativeGIP Interestingly, the opposite picture arose here, as thehuman GIP displayed a lower potency and efficacy on allthree receptors compared with the rodent GIP ligands(Figure 5A–C respectively) The rodent GIPs were equallypotent and efficacious on all tested receptors, without anyconsiderable change in potency between the receptors(EC50~ 10 pM) (Table 1) In contrast, human GIP had a lower
efficacy on all three receptors with the largest difference inthe rodent receptors with an Emax of 75% on the rat GIPreceptor and 60% on mouse GIP receptors compared withthe corresponding rodent GIP Likewise, human GIP had alower potency than the rodent GIPs on all three receptorswith the largest difference being 23-fold on the mouse GIPreceptor followed by 10-fold and 5-fold on the rat and humanGIP receptors respectively
No inter-species differences in the binding affinities of GIP analogues or (Pro3)GIP analogues
In order to determine whether the differences at both the tor and the ligand level in terms of cAMP accumulation weredue to differences in binding affinity, competition binding ex-periments were conducted using 125I-labelled human GIP asthe radioligand Human GIP was found to bind to all three
recep-Figure 2
Human (Pro3)GIP is a full agonist, rat and mouse (Pro3)GIP are partial agonists COS-7 cells were transiently transfected with either human GIPreceptors (GIPR; A), rat GIPR (B), or mouse GIPR (C) and assayed for cAMP accumulation following increasing concentrations of WT GIP and(Pro3)GIP from the corresponding species The data was normalised to Emaxof the endogenous GIP on every receptor and shown as mean
±SEM, N≥3 Nonlinear regression was used to calculate the EC50value and Emax
Species-specific activity of (Pro3)GIP
BJP
Trang 32Figure 3
The rodent partial agonist peptides, (Pro3)GIP, are functional antagonists at their corresponding GIP receptors (GIPR) (A) rat GIP and (B) mouseGIP cAMP accumulation concentration–response curves with or without increasing concentrations (10, 100 and 1000nM) of rat (Pro3)GIP ormouse (Pro3)GIP The results were normalized to Emaxof the species-specific GIP in the absence of (Pro3)GIP and shown as mean + SEM, N = 3.Nonlinear regression was used to calculate EC50values Schild plot analysis of the dose–response curves of (C) rat GIP and (D) mouse GIP revealed
Ki-values of 13 and 61 nM respectively
Figure 4
Human (Pro3)GIP is a partial agonist on rodent receptors COS-7 cells were transiently transfected with either human GIPR (A), rat GIPR (B), ormouse GIPR (C) and assayed for cAMP accumulation following increasing concentrations of human, rat, and mouse (Pro3)GIP The data was nor-malised to Emaxof the species specific GIP on every receptor and shown as mean±SEM, N≥3 Nonlinear regression was used to calculate the EC50
value and Emax
BJP A H Sparre-Ulrich et al.
32 British Journal of Pharmacology (2016) 173 27–38
Trang 33receptors with equally high affinities (Kdof 0.90 to 1.1 nM)
(Table 2) Likewise, both rodent GIP analogues were similar to
human GIP on the tested receptors with a Kiin the nM range
from 0.27 nM of rat GIP on the mouse GIP receptor to 2.0 nM
for mouse GIP on the human GIP receptor Thus, the functional
discrepancies between human and rodent GIP are not due to
ac-tual differences in affinity The (Pro3)GIP analogues displayed in
average 10-fold lower affinities than the native GIP analogues
on all three receptors with the lowest affinity observed for the
human ligand on mouse GIP receptor (Kiof 39 nM) whereas
the rest displayed affinities (Ki) between 4.8 and 14 nM Thus,
similar to the native GIP ligands, the functional differences
be-tween the human (Pro3)GIP and the rodent counterparts are
not caused by differences in binding affinities
(Pro3)GIP stimulates insulin, glucagon and
somatostatin release
As most of the studies describing (Pro3)GIP were carried out
in rodent models (Gault et al., 2007; Irwin et al., 2007;
McClean, 2007), we determined how the receptor bindingand activation would translate into hormone secretion, usingperfused pancreata from rodents In brief, the tested ana-logues were perfused through the pancreata of the rodents,and the venous effluent was collected at 1 min interval andanalysed for insulin, glucagon and somatostatin content Inboth perfusion models, we used 100-fold higher concentra-tions of (Pro3)GIP (100 nM), compared with that of GIP(1 nM), because of the 1000-fold difference in potencies(Table 1) and the 10-fold difference in affinity (Table 2) Inthe mouse pancreas, (Pro3)GIP as well as GIP-induced secre-tion of insulin, glucagon and somatostatin (Figure 6A–C) asexpected from the agonistic nature of (Pro3)GIP (Figure 2C).However, compared with GIP, there were differences in themagnitude of secretion Although given at 100-fold higherconcentration, (Pro3)GIP did not reach the same levels ofinsulin secretion as GIP (Figure 6A), while there was no differ-ence in somatostatin and glucagon release (Figure 6B and C)
Table 1
cAMP accumulation induced by human, rat, and mouse GIP and (Pro3)GIP
Human GIPRlogEC50±SEM
EC50
(nM)
Emax±SEM (N)
Rat GIPRlogEC50±SEM
EC50
(nM)
Emax±SEM (N)
Mouse GIPRlogEC50±SEM
EC50
(nM)
Emax±SEM (N)
Figure 5
Human GIP is less efficacious and has a lower potency than rodent GIP COS-7 cells were transiently transfected with either human GIP receptors(GIPR; A), rat GIPR (B), or mouse GIPR (C) and assayed for cAMP accumulation following increasing concentrations of human, rat and mouse GIP.The data was normalized to Emaxof the species specific GIP on every receptor and shown as mean±SEM, N≥3 Nonlinear regression was used tocalculate the EC50value and Emax
Species-specific activity of (Pro3)GIP
BJP
Trang 34Similar results were obtained in the rat perfused pancreas
model with a significantly lower insulin and somatostatin
secretion by (Pro3)GIP compared with GIP (Figure 6D and F,
respectively) but with an equal glucagon release by both
ligands (Figure 6E)
In order to confirm that the observed hormone secretion
from perfused pancreata was mediated through the GIP
receptors, the same experimental set-up was repeated using
GIP receptor KO mice As seen in Figure 7, GIP
receptor-deficient mice did not respond to mouse GIP or mouse
(Pro3)GIP with either insulin, glucagon or somatostatin
secretion (Figure 7A–C respectively) As with the pancreata
from wild-type mice and rats, the positive control, arginine,
induced secretion of the respective hormones, validatingthe experimental set-up
Discussion
Our study demonstrated that human (Pro3)GIP was not anantagonist at human GIP receptors, but rather approachesfull agonism At rodent GIP receptors, (Pro3)GIP was a par-tial agonist with competitive antagonistic properties whenevaluated in terms of cAMP accumulation In terms of its ef-fect on pancreatic hormone secretion, (Pro3)GIP followedthe partial agonistic pattern seen in cAMP with regard to in-
Figure 6
(Pro3)GIP is a agonist on rodent pancreata (A/D) insulin, (B/E) glucagon, and (C/F) somatostatin secretion from perfused mouse (A, B, C) and rat(D, E, F) pancreata following stimulation with either species specific 100 nM (Pro3)GIP, 1 nM GIP, or 10 mM arginine (N=6-9) The glucose con-centration was 7 mM and data are mean±SEM
Table 2
Similar displacement of125I-labeled hGIP by GIP analogues or (Pro3)GIP analogues
Human GIPRlogIC50± SEM Ki/Kd(nM) (N)
Rat GIPRlogIC50± SEM Ki/Kd(nM) (N)
Mouse GIPRlogIC50± SEM Ki/Kd(nM) (N)
BJP A H Sparre-Ulrich et al.
34 British Journal of Pharmacology (2016) 173 27–38
Trang 35(Pro3)GIP induced a glucagon release equal to that induced
by native GIP in rats
(Pro3)GIP is not an effective GIP receptor
antagonist for future studies of the GIP system
Thefirst GIP receptor antagonists that showed promise were
the truncated and amidated forms, GIP(6–30)-NH2and GIP
200 nM obtained in heterologous binding experiments
(Tseng et al., 1996; Gelling et al., 1997; Hinke et al., 2001)
However, elaborate in vivo characterization has not been
car-ried out The naturally occurring GIP metabolite GIP(3–42)
binds with similar affinity (IC50of 22 nM); however, no tagonistic effect could be demonstrated in vivo in pigs(Deacon et al., 2006) Low MW compounds have also beenpresented, e.g the antagonist SKL-14959 with an affinity (Kivalue) of 55 nM and the ability to increase blood glucoselevels and inhibit insulin secretion during an oral glucose tol-erance test in mice (Nakamura et al., 2012) Further develop-ment of this compound has not been published Recently, apromising GIP receptor antibody, Gipg013, with a Ki of6.8 nM was reported (Ravn et al., 2013) Based on the numer-ous rodent studies that displayed promising improvements inthe diabetic and/or obese state, (Pro3)GIP showed potential
an-tofill the role as a promising antagonist in the context of
Figure 7
The effects seen in perfused pancreata are mediated through GIP receptors (GIPR) A) insulin, B) glucagon, and C) somatostatin secretion ing stimulation of perfused pancreata from GIPR KO mice by either 100 nM mouse (Pro3)GIP, 1 nM mouse GIP, or 10 mM arginine (N=3) Theglucose concentration was 7 mM and data are mean±SEM
follow-Figure 8
The changes in potency and efficacy of the tested GIP receptors (GIPR) and GIP analogues The calculated EC50(A/C) and Emax(B/D) values termined from the cAMP accumulation assays and normalised to human GIP on each species receptor (A, B, and D) or human (Pro3)GIP on eachspecies receptor
de-Species-specific activity of (Pro3)GIP
BJP
Trang 36GIP physiology (Gault et al., 2007; Irwin et al., 2007;
McClean, 2007; De Toro-Martin et al., 2014) In ob/ob mice,
chronic treatment with (Pro3)GIP improved glucose
toler-ance and insulin sensitivity (Irwin et al., 2007), while
treat-ment in mice previously on a high-fat diet resulted in
weight loss, improved insulin sensitivity and glucose
toler-ance (McClean, 2007)
Consistent with previous rodent studies (Gault et al.,
2002; Gault et al., 2003; Gault Victor et al., 2007), wefind
that rodent (Pro3)GIP ligands act as competitive
antago-nists on the rodent receptors (Figure 3) But when it comes
to the human GIP system, human (Pro3)GIP acted as a
effi-cacious agonist (Figure 2A), which is in line with very
re-cent studies that reported substantial agonist activity of
human (Pro3)GIP with efficacies up to 83% of human GIP
in cAMP release from transiently transfected HEK293 cells
(Ravn et al., 2013) or CHL cells (Pathak et al., 2015), and
in the reporter gene expression for cAMP-response element
(Al-Sabah et al., 2014) However, this contrasts to a previous
study demonstrating (Pro3)GIP to have 9% of GIP’s efficacy
on transiently transfected Chinese hamster lung cells (CHL)
expressing the human GIP receptor (Gault Victor et al.,
2007) These efficacy discrepancies may rely on differences
in cell types (CHL, HEK283 and COS-7 cells); however,
con-sistent for all studies is thefinding that (Pro3)GIP was not a
neutral antagonist, but has agonist properties in
cAMP-dependent pathways
Structural GIP divergence between species has
markedly affects the pharmacology
Our study emphasizes important interspecies variations
within the GIP system The rodent GIP receptorligands were
more potent and efficacious than human GIP on all the tested
receptors (Figure 5), with up to 22-fold and 25-fold increase in
potency of rat GIP and mouse GIP on the mouse GIP
recep-tors (Figure 8A and B) These changes were not matched by
a similar increase in binding affinities (Table 2) Only few
amino acids are altered among the three GIP receptor ligands
(Figure 1A) The most dramatic change is located at position
18 where human GIP has a histidine instead of an arginine,
which is found in rat and mouse GIP Among the few
non-identities observed (in positions 18, 30, 34 and 40), this is
the only case where the rodent sequences are identical and,
because of the non-conservative nature (Arg/His), a
signifi-cant structural divergence is possible A previous study
sup-ports the functional importance of this position, as an
alanine substitution in human GIP increased insulin
secre-tion from BRIN-BD11 cells compared with native GIP
(Venneti et al., 2011) Thus, position 18 seems to have a role
in the activation mechanism of the GIP receptor, as also
sup-ported by a recent study predicting a direct interaction
be-tween His18 of human GIP and Ala32 of the N-terminus of
the GIP receptor (Tikhele et al., 2010) Thus, thesefindings
add some detail to the general view that hydrophobic
interac-tions between the C-terminal part of GIP and the N-terminus
receptor parts account for binding, while the N-terminus of
GIP interacts with other extracellular receptor domains
in-ducing activation (Malde et al., 2007; Parthier et al., 2007)
This so-called two-step receptor activation not only describes
GIP receptor activation but presents a global mechanism for
ligand-binding and receptor activation among class B1 tors (e.g glucagon, GLP-1, secretin, vasoactive intestinalpolypeptide and parathyroid hormone receptor) (Gourlet
recep-et al., 1996; Hjorth and Schwartz, 1996; Gardella and Jüppner,2001; Castro et al., 2005; Vilardaga et al., 2011) It also extendsbeyond class B1, as also some class A seven-transmembranereceptors (e.g chemokine, C3a and C5a receptors) are acti-vated by their endogenous peptide ligands in a two (ormulti)-step model involving receptor recognition and activa-tion by different regions in the ligands (Allen et al., 2007; Klos
et al., 2013; Thiele and Rosenkilde, 2014)
Taken together, the rodent GIP systems seem more activecompared with the human system, in terms of GIP (1–42)-mediated activation and more sensitive to GIP ligandmodifications, as judged by the lower activity of (Pro3)GIP.This is relevant and should be considered in studies using ro-dent models As our study indicates, when using human GIP
in rodent models, researchers are in fact injecting a less tent partial agonist (compared with rodent GIP) that maylead to underestimation of the true GIP activity in such exper-imental systems
po-The therapeutic potential of targeting the GIP receptor
In the wake of the GIP receptor KO studies demonstrating sistance to diet-induced obesity (Miyawaki et al., 2002), manyhave tried to antagonize the GIP receptors as a potential tar-get for treating obesity Accumulating evidence links GIP bi-ology to adiposity and due to the lack of an effective GIPreceptor antagonist, the therapeutic potential of antagoniz-ing this receptor in obese patients remains to be assessed Inthis context, our study brings attention to a significant inter-species difference within the GIP system, and thus, pharma-cological evaluation of novel compounds in rodent modelsshould interpreted cautiously with respect to their relevancefor humans
re-Acknowledgements
This work was supported by the Novo Nordisk FoundationCenter for Basic Metabolic Research and the University ofCopenhagen
Con flicts of interest
A H S U., L S H., B S., M C., B H and M M R declare thatthey have no conflict of interest J J H has served as a consul-tant or advisor to Novartis Pharmaceuticals, Novo Nordisk,Merck Sharp & Dohme and Roche and has received fees forlectures from Novo Nordisk, Merck Sharp & Dohme andGlaxoSmithKline F K K has received fees for consultancy,lectures or being part of an advisory board from AstraZeneca,Boehringer Ingelheim, Bristol-Myers Squibb, Eli Lilly, Gilead,Merck Sharp & Dohme, Novo Nordisk, Sanofi and ZealandPharma, and has received research support from Sanofi andNovo Nordisk
BJP A H Sparre-Ulrich et al.
36 British Journal of Pharmacology (2016) 173 27–38
Trang 37Ahlqvist E, Osmark P, Kuulasmaa T, Pilgaard K, Omar B, Brøns C et al
(2013) Link between GIP and osteopontin in adipose tissue and
insulin resistance Diabetes 62: 2088–2094
Alexander SPH, Benson HE, Faccenda E, Pawson AJ, Sharman JL,
Spedding M et al (2013) The Concise Guide to PHARMACOLOGY
2013/14: G Protein-Coupled Receptors Br J Pharmacol 170:
1459–1581
Allen SJ, Crown SE, Handel TM (2007) Chemokine:receptor structure,
interactions, and antagonism Annu Rev Immunol 25: 787–820
Al-Sabah S, Al-Fulaij M, Ahmed HA (2014) Selectivity of peptide
ligands for the human incretin receptors expressed in HEK-293 cells
Eur J Pharmacol 741: 311–315
Asmar M, Simonsen L, Madsbad S, Stallknecht B, Holst JJ, Bülow J
(2010) Glucose-dependent insulinotropic polypeptide may enhance
fatty acid re-esterification in subcutaneous abdominal adipose tissue
in lean humans Diabetes 59: 2160–2163
Baggio LL, Drucker DJ (2007) Biology of incretins: GLP-1 and GIP
Gastroenterology 132: 2131–2157
Baldissera FGA, Munoz-Perez MA, Holst JJ (1983) Somatostatin 1–28
circulates in human plasma Regul Pept 6: 63–69
Brand CL, Jorgensen PN, Knigge U, Warberg J, Svendsen I, Kristensen
JS et al (1995) Role of glucagon in maintenance of euglycemia in fed
and fasted rats Am J Physiol 269: E469–E477
Brøns C, Jensen CB, Storgaard H, Hiscock NJ, White A, Appel JS et al
(2009) Impact of short-term high-fat feeding on glucose and insulin
metabolism in young healthy men J Physiol 587: 2387–2397
Calanna S, Christensen M, Holst JJ, Laferrère B, Gluud LL, Vilsbøll T
et al (2013) Secretion of glucose-dependent insulinotropic
polypeptide in patients with type 2 diabetes: systematic review and
meta-analysis of clinical studies Diabetes Care 36: 3346–3352
Canal CE, Cordova-Sintjago T, Liu Y, Kim MS, Morgan D, Booth RG
(2013) Molecular pharmacology and ligand docking studies reveal a
single amino acid difference between mouse and human serotonin
5-HT2A receptors that impacts behavioral translation of novel
4-phenyl-2-dimethylaminotetralin ligands J Pharmacol Exp
Therapeut 347: 705–716
Castro M, Nikolaev VO, Palm D, Lohse MJ, Vilardaga J-P (2005) Turn-on
switch in parathyroid hormone receptor by a two-step parathyroid
hormone binding mechanism Proc Natl Acad Sci U S A 102: 16084–16089
Christensen M, Vedtofte L, Holst JJ, Vilsboell T, Knop FK (2011)
Glucose-Dependent insulinotropic polypeptide: a bifunctional
glucose-dependent regulator of glucagon and insulin secretion in
humans Diabetes 60: 3103–3109
De Toro-Martin J, Fernandez-Millan E, Lizarraga-Mollinedo E,
Lopez-Oliva E, Serradas P, Escriva F et al (2014) Predominant role of
GIP in the development of a metabolic syndrome-like phenotype in
female Wistar rats submitted to forced catch-up growth
Endocrinology 155: 3769–3780
Deacon CF, Plamboeck A, Rosenkilde MM, deHeer J, Holst JJ (2006)
GIP-(3–42) does not antagonize insulinotropic effects of GIP at physiological
concentrations Am J Physiol Endocrinol Metabol 291: E468–E475
Deblasi A, O’reilly K, Motulsky HJ (1989) Calculating receptor
number from binding experiments using same compound as
radioligand and competitor Trends Pharmacol Sci 10: 227–229
Deschamps I, Heptner W, Desjeux JF, Baltakse V, Machinot S, Lestradet
H (1980) Effects of diet on insulin and gastric inhibitory polypeptide
levels in obese children Pediatr Res 14: 300–303
Ebert R, Illmer K, Creutzfeldt W (1979) Release of gastric inhibitorypolypeptide (GIP) by intraduodenal acidification in rats and humansand abolishment of the incretin effect of acid by GIP-antiserum inrats Gastroenterology 76: 515–523
Fulurija A, Lutz TA, Sladko K, Osto M, Wielinga PY, Bachmann MF et al.(2008) Vaccination against GIP for the treatment of obesity PLoSOne 3: e3163
Gardella TJ, Jüppner H (2001) Molecular properties of thePTH/PTHrP receptor Trends Endocrinol Metabol 12: 210–217.Gault Victor A, Hunter K, Irwin N, Greer B, Green Brian D, Harriott P
et al (2007) Characterisation and glucoregulatory actions of a novelacylated form of the (Pro3)GIP receptor antagonist in type 2 diabetes.Biol Chem 388: 173
Gault VA, O’harte FPM, Harriott P, Flatt PR (2002) Characterization ofthe cellular and metabolic effects of a novel enzyme-resistantantagonist of glucose-dependent insulinotropic polypeptide.Biochem Biophys Res Commun 290: 1420–1426
Gault VA, O’harte FPM, Harriott P, Mooney MH, Green BD, Flatt PR(2003) Effects of the novel (Pro3)GIP antagonist and exendin(9–39)amide on GIP- and GLP-1-induced cyclic AMP generation, insulinsecretion and postprandial insulin release in obese diabetic (ob/ob)mice: evidence that GIP is the major physiological incretin.Diabetologia 46: 222–230
Gault VA, Mcclean PL, Cassidy RS, Irwin N, Flatt PR (2007) Chemicalgastric inhibitory polypeptide receptor antagonism protects againstobesity, insulin resistance, glucose intolerance and associated disturbances
in mice fed high-fat and cafeteria diets Diabetologia 50: 1752–1762.Gelling RW, Coy DH, Pederson RA, Wheeler MB, Hinke S, Kwan T et al.(1997) GIP(6-30amide) contains the high affinity binding region ofGIP and is a potent inhibitor of GIP1-42 action in vitro Regul Pept 69:151–154
Gourlet P, Vilardaga J-P, De Neef P, Waelbroeck M, Vandermeers A,Robberecht P (1996) The C-terminus ends of secretin and VIP interactwith the N-terminal domains of their receptors Peptides 17: 825–829.Gutniak M, Orskov C, Holst JJ, Ahrén B, Efendic S (1992)
Antidiabetogenic effect of glucagon-like peptide-1 (7–36)amide innormal subjects and patients with diabetes mellitus New Engl JMedMassachusetts Medical Society 326: 1316–1322
de Heer J, Rasmussen C, Coy DH, Holst JJ (2008) Glucagon-likepeptide-1, but not glucose-dependent insulinotropic peptide,inhibits glucagon secretion via somatostatin (receptor subtype 2) inthe perfused rat pancreas Diabetologia 51: 2263–2270
Hinke SA, Manhart S, Pamir N, Demuth HU, Gelling W, Pederson RA
et al (2001) Identification of a bioactive domain in the terminus of glucose-dependent insulinotropic polypeptide (GIP).Biochim Biophys Acta Protein Struct Mol Enzymol 1547: 143–155.Hjorth SA, Schwartz TW (1996) Glucagon and GLP-1 receptors: lessonsfrom chimeric ligands and receptors Acta Physiol Scand 157: 343–345.Holst JJ (2004) On the Physiology of GIP and GLP-1 Horm Metab Res36: 747–754
amino-Irwin N, Green BD, Parker JC, Gault VA, O’harte FPM, Flatt PR (2006).Biological activity and antidiabetic potential of synthetic fragmentpeptides of glucose-dependent insulinotropic polypeptide, GIP(1–16)and (Pro3)GIP(1–16) Regul Pept 135: 45–53
Irwin N, Mcclean PL, O’harte FPM, Gault VA, Harriott P, Flatt PR (2007).Early administration of the glucose-dependent insulinotropicpolypeptide receptor antagonist (Pro3)GIP prevents the development ofdiabetes and related metabolic abnormalities associated with geneticallyinherited obesity in ob/ob mice Diabetologia 50: 1532–1540
Species-specific activity of (Pro3)GIP
BJP
Trang 38Irwin N, Mcclean PL, Patterson S, Hunter K, Flatt PR (2009) Active
immunisation against gastric inhibitory polypeptide (GIP) improves
blood glucose control in an animal model of obesity-diabetes Biol
Chem 390: 75
Jørgensen NB, Dirksen C, Bojsen-Møller KN, Jacobsen SH, Worm D,
Hansen DL et al (2013) Exaggerated glucagon-like peptide 1
response is important for improvedβ-cell function and glucose
tolerance after roux-en-Y gastric bypass in patients with type 2
diabetes Diabetes 62: 3044–3052
Kerr BD, Flatt AJS, Flatt PR, Gault VA (2011) Characterization and
biological actions of N-terminal truncated forms of
glucose-dependent insulinotropic polypeptide Biochem Biophys Res
Commun 404: 870–876
Kilkenny C, Browne W, Cuthill IC, Emerson M, Altman DG (2010)
NC3Rs Reporting Guidelines Working Group Br J Pharmacol 160:
1577–1579
Kissow H, Hartmann B, Holst JJ, Viby NE, Hansen LRS, Rosenkilde
MM et al (2012) Glucagon-like peptide-1 (GLP-1) receptor agonism
or DPP-4 inhibition does not accelerate neoplasia in carcinogen
treated mice Regul Pept 179: 91–100
Klos A, Wende E, Wareham KJ, Monk PN (2013) International Union
of Basic and Clinical Pharmacology LXXXVII Complement Peptide
C5a, C4a, and C3a Receptors Pharmacol Rev 65: 500–543
Knudsen LB, Hastrup S, Underwood CR, Wulff BS, Fleckner J (2012)
Functional importance of GLP-1 receptor species and expression
levels in cell lines Regul Pept 175: 21–29
Kreymann B, Ghatei MA, Williams G, Bloom SR (1987) Glucagon-like
peptide-1 7–36: A physiological incretin in man Lancet 330:
1300–1304
Lazareno S, Birdsall NJ (1993) Estimation of competitive antagonist
affinity from functional inhibition curves using the Gaddum, Schild
and Cheng-Prusoff equations Br J Pharmacol 109: 1110–1119
Malde AK, Srivastava SS, Coutinho EC (2007) Understanding
interactions of gastric inhibitory polypeptide (GIP) with its G-protein
coupled receptor through NMR and molecular modeling J Pept Sci
13: 287–300
Mcclean PLI (2007) GIP receptor antagonism reverses obesity, insulin
resistance, and associated metabolic disturbances induced in mice by
prolonged consumption of high-fat diet Am J Physiol Endocrinol
Metabol 293: E1746–E1755
McGrath J, Drummond G, Kilkenny C, Wainwright C (2010)
Guidelines for reporting experiments involving animals: the ARRIVE
guidelines Br J Pharmacol 160: 1573–1576
Miyawaki K, Yamada Y, Yano H, Niwa H, Ban N, Ihara Y et al (1999)
Glucose intolerance caused by a defect in the entero-insular axis: a
study in gastric inhibitory polypeptide receptor knockout mice Proc
Natl Acad Sci 96: 14843–14847
Miyawaki K, Yamada Y, Ban N, Ihara Y, Tsukiyama K, Zhou H et al
(2002) Inhibition of gastric inhibitory polypeptide signaling
prevents obesity Nat Med 8: 738–742
Nakamura T, Tanimoto H, Mizuno Y, Tsubamoto Y, Noda H (2012)
Biological and functional characteristics of a novel low-molecular
weight antagonist of glucose-dependent insulinotropic polypeptide
receptor, SKL-14959, in vitro and in vivo Diabetes Obes Metabol 14:
511–517
Nasteska D, Harada N, Suzuki K, Yamane S, Hamasaki A, Joo E et al (2014).Chronic reduction of GIP secretion alleviates obesity and insulin resistanceunder high-fat diet conditions Diabetes 63: 2332–2343
Orskov C, Jeppesen J, Madsbad S, Holst JJ (1991) Proglucagonproducts in plasma of noninsulin-dependent diabetics andnondiabetic controls in the fasting state and after oral glucose andintravenous arginine J Clin Invest 87: 415–423
Parthier C, Kleinschmidt M, Neumann P, Rudolph R, Manhart S,Schlenzig D et al (2007) Crystal structure of the incretin-boundextracellular domain of a G protein-coupled receptor Proc Natl AcadSci 104: 13942–13947
Pathak V, Gault VA, Flatt PR, Irwin N (2015) Antagonism of gastricinhibitory polypeptide (GIP) by palmitoylation of GIP analogues with N-and C-terminal modifications improves obesity and metabolic control inhigh fat fed mice Mol Cell Endocrinol 401: 120–129
Pawson AJ, Sharman JL, Benson HE, Faccenda E, Alexander SP,Buneman OP et al (2014) The IUPHAR/BPS Guide toPHARMACOLOGY: an expert-driven knowledge base of drug targetsand their ligands Nucl Acids Res 42 (Database Issue): D1098–D1106.Raufman JP, Singh L, Eng J (1991) Exendin-3, a novel peptide fromHeloderma horridum venom, interacts with vasoactive intestinalpeptide receptors and a newly described receptor on dispersed acinifrom guinea pig pancreas Description of exendin-3(9–39) amide, aspecific exendin receptor antagonist J Biol Chem 266: 2897–2902.Ravn P, Madhurantakam C, Kunze S, Matthews E, Priest C, O’brien S
et al (2013) Structural and pharmacological characterization ofnovel potent and selective monoclonal antibody antagonists ofglucose-dependent insulinotropic polypeptide receptor J Biol Chem288: 19760–19772
Scott JS, Brocklehurst KJ, Brown HS, Clarke DS, Coe H, Groombridge
SD et al (2013) Conformational restriction in a series of GPR119agonists: differences in pharmacology between mouse and human.Bioorg Med Chem Lett 23: 3175–3179
Thiele S, Rosenkilde M (2014) Interaction of chemokines with theirreceptors– from initial chemokine binding to receptor activatingsteps Curr Med Chem 21: 3594–3614
Tikhele SH, Pissurlenkar RRS, Srivastava S, Saran A, Coutinho EC(2010) Mapping interactions of gastric inhibitory polypeptide withGIPR N-terminus using NMR and molecular dynamics simulations JPept Sci 16: 383–391
Tseng CC, Kieffer TJ, Jarboe LA, Usdin TB, Wolfe MM (1996).Postprandial stimulation of insulin release by glucose-dependentinsulinotropic polypeptide (GIP) Effect of a specific glucose-dependent insulinotropic polypeptide receptor antagonist in the rat
J Clin Invest 98: 2440–2445
Venneti KC, Malthouse JP, O’harte FPM, Hewage CM (2011).Conformational, receptor interaction and alanine scan studies ofglucose-dependent insulinotropic polypeptide Biochim BiophysActa Protein Proteonomics 1814: 882–888
Vilardaga J-P, Romero G, Friedman P, Gardella T (2011) Molecularbasis of parathyroid hormone receptor signaling and trafficking: afamily B GPCR paradigm Cell Mol Life Sci 68: 1–13
BJP A H Sparre-Ulrich et al.
38 British Journal of Pharmacology (2016) 173 27–38
Trang 39RESEARCH PAPER
Contractile function
assessment by intraventricular
balloon alters the ability of
regional ischaemia to evoke
Catherine D E Wilder, Radwa Masoud, Duygu Yazar, Brett A O ’Brien,
Thomas R Eykyn and Michael J Curtis
Cardiovascular Division, King‘s College London, London, UK
Correspondence
Michael J Curtis, CardiovascularDivision, Faculty of Life Sciences andMedicine, The Rayne Institute, StThomas’ Hospital, London SE17EH,UK
E-mail: michael.curtis@kcl.ac.uk -
BACKGROUND AND PURPOSE
In drug research using the rat Langendorff heart preparation, it is possible to study left ventricular (LV) contractility using anintraventricular balloon (IVB), and arrhythmogenesis during coronary ligation-induced regional ischaemia Assessing both con-currently would halve animal requirements We aimed to test the validity of this approach
CONCLUSIONS AND IMPLICATIONS
IVB inflation does not inhibit VF suppression by a standard drug, but it has profound antiarrhythmic effects of its own, likely to bedue to inflation-induced localized ischaemia This means rhythm and contractility cannot be assessed concurrently by this ap-proach, with implications for drug discovery and safety assessment
Abbreviations
BG, bigeminy; IVB, intraventricular balloon; IZ, ischaemic zone; LV, left ventricular; NMR, nuclear magnetic resonance; NO,nitric oxide; PCr, phosphocreatine; Pi, inorganic phosphate; TVW, total ventricular weight; UZ, uninvolved zone; VF, ven-tricularfibrillation; VPB, ventricular premature beat; VT, ventricular tachycardia
BJP British Journal of Pharmacology www.brjpharmacol.org
Trang 40The present study reports a refinement of a common model
for studying drug actions on the heart, identifies an
unex-pected alteration in the model’s bioassay characteristics,
explains this mechanistically and provides guidance on
future usage
An intraventricular balloon (IVB) is commonly used in
the rat isolated Langendorff perfused heart in order to
measure left ventricular (LV) contractile function (Ridley
and Curtis, 1992; Farkas et al., 1999; Crook and Curtis,
2012) In addition, the rat isolated heart is also used to
evalu-ate ischaemia-induced and reperfusion-induced cardiac
arrhythmias such as ventricular fibrillation (VF) (Ridley
et al., 1992; Clements-Jewery et al., 2006; Crook and Curtis,
2012) Assessment of rhythm and LV contractility
simulta-neously in a single experiment is a strategy that would give
a more comprehensive assessment of drug effects with
reduced animal usage Indeed, the growing interest in the
rat Langendorff preparation with an IVB for cardiac safety
assessment (Henderson et al., 2013) coupled with its
longstanding value for examining drug effects on regional
ischaemia-induced arrhythmias (Curtis et al., 2013) indicates
that combining assessment into a single experiment is a
log-ical next step
It is not known whether it is feasible to do this, and there
are reasons to suspect that IVB assessment of function may
affect arrhythmogenesis owing to effects of LV stretch Studies
on electrically induced arrhythmias with and without
ischae-mia suggest an increase in VF susceptibility and complexity
with LV stretch (Coronel et al., 2002; Parker et al., 2004;
Barrabés et al., 2013), and importantly, from a
pharmacologi-cal perspective, possible alteration of the effects of
antiar-rhythmic drugs (Chorro et al., 2000) However, the effects of
LV stretch on VF induced by ischaemia (rather than by
electri-cal stimulation) have not been systematielectri-cally established,
especially in the versatile rat Langendorff preparation
In rat isolated hearts subjected to regional ischaemia, the
incidence of VF is dependent on the size of the ischaemic
territory (Ridley et al., 1992) as it is in larger species (Austin
et al., 1982) Moreover, in the rat isolated heart, the size of
the territory, that is, the ischaemic zone (IZ), can be varied
easily by selecting the position of the coronary ligature (Ridley
et al., 1992) Proximal ligation of the left coronary artery in rat
isolated hearts results in approximately 80% of hearts
devel-oping VF, whereas placing the ligature more distally to the
atrial appendage reduces VF incidence proportionately (Ridley
et al., 1992) This allows evaluation of an alteration in the tionship between VF and IZ by drugs, or by other manipula-tion such as IVB inflation
rela-In order to determine the feasibility of combining IVBassessment of contractility with assessment of ischaemia-induced arrhythmias, and assess whether an IVB facilitatesischaemia-induced arrhythmias in the rat isolated heart, wedeliberately made the IZ smaller (~35% mean of the ventricu-lar mass, with individual values<40%) than is typically usedfor antiarrhythmic drug studies, thereby generating a moremoderate baseline incidence of VF (~33%) to permit detec-tion of any increase in VF incidence In separate studies, weexamined whether IVB inflation affected the antiarrhythmicaction of verapamil, a drug well characterized in the unloadedpreparation (Farkas et al., 1999) and which has been reported
to alter the pattern of VF in rabbit hearts subjected to IVB flation (Chorro et al., 2000) Coronary effluent lactate releaseand 31P nuclear magnetic resonance (NMR) spectroscopy(providing data on intracellular pH, ATP and other variablessusceptible to ischaemia) were analysed in two separate stud-ies in order to explore whether IVB-induced myocardial in-jury was the mechanism that determined outcome
in-Methods
Concordance with ARRIVE, ethical and legal requirements and new guidance on design and analysis
Animal housing and husbandry were exactly as previouslydescribed (Andrag and Curtis, 2013), in full compliance withARRIVE (Kilkenny et al., 2010) and United Kingdom HomeOffice Guide on the Operation of the Animals (Scientific Proce-dures) Act 1986, and the new guidance on design and analysisfor British Journal of Pharmacology (Curtis et al., 2015) withblinding and randomization throughout, and are thereforenot described in detail here A total of 164 animals were used
in the experiments described here
Animals and general experimental methodsRats (male Wistar; Harlan UK; 280–400 g) were anaesthetizedwith a lethal dose of sodium pentobarbitone (170 mg kg 1i.p.) and given sodium heparin (160 IU kg 1 i.p.) in order
to preclude blood clot formation within the coronary ture The high dose of pentobarbitone obtained a rapid onset
BJP C D E Wilder et al.
40 British Journal of Pharmacology (2016) 173 39–52