Potential of p38MAPK inhibitors in the treatment of ischaemic heart disease

15
UNCORRECTED PROOF 1 Associate editor: Madhani Q1 2 Potential of p38-MAPK inhibitors in the treatment of ischaemic heart disease 3 James E. Clark, Negin Sarafraz, Michael S. Marber 4 The Cardiovascular Division, Kings College London, The Rayne Institute, St Thomas' Hospital, London, SE1 7EH, United Kingdom 5 Abstract 6 Chronic heart failure is debilitating, often fatal, expensive to treat and common. In most patients it is a late consequence of myocardial 7 infarction (MI). The intracellular signals following infarction that lead to diminished contractility, apoptosis, fibrosis and ultimately heart failure 8 are not fully understood but probably involve p38-mitogen activated protein kinases (p38), a family of serine/threonine kinases which, when 9 activated, cause cardiomyocyte contractile dysfunction and death. Pharmacological inhibitors of p38 suppress inflammation and are undergoing 10 clinical trials in rheumatoid arthritis, Chrohn's disease, psoriasis and surgery-induced tissue injury. In this review, we discuss the mechanisms, 11 circumstances and consequences of p38 activation in the heart. The purpose is to evaluate p38 inhibition as a potential therapy for ischaemic heart 12 disease. 13 © 2007 Elsevier Inc. All rights reserved. 14 15 Keywords: p38; MAPK; Inhibitors; Heart failure; Ischaemia; Infarction 16 17 Contents 18 1. Introduction .............................................. 0 19 2. p38-Mitogen-activated protein kinase ................................. 0 20 3. Structure and function of p38-MAPK ................................. 0 21 4. Mechanisms of p38-MAPK activation................................. 0 22 4.1. Mitogen-activated protein kinase kinases............................ 0 23 4.2. Autophosphorylation ...................................... 0 24 5. p38, myocardial ischaemia and ischaemic heart disease ........................ 0 25 6. Pharmacological inhibitors of p38-MAPK............................... 0 26 7. Clinical trials of p38-MAPK inhibition ................................ 0 27 8. Summary and conclusions ....................................... 0 28 References ................................................. 0 29 30 1. Introduction 31 Atherosclerotic vascular disease manifests predominantly as 32 heart disease and stroke, which are the most frequent causes 33 of death in the United Kingdom. Collectively atherosclerotic 34 vascular disease was responsible for 40% of total mortality in the 35 United Kingdom in 2004. Despite recent reductions in this high 36 mortality, morbidity is increasing as more patients survive 37 myocardial infarction (MI) and stroke. For example, about 38 1.3 million people in the United Kingdom have survived acute 39 MI, 2 million have angina and 0.9 million have heart failure. The 40 extraordinarily high impact of atherosclerosis on the Nation's 41 health is reflected in its economic cost. Atherosclerotic disease 42 of just the coronary arteries costs the United Kingdom healthcare 43 system £3500 million with an estimated further £4400 million 44 lost to the economy through premature death, illness and infor- 45 mal care (The United Kingdom Heart Attack Study (UKHAS) 46 Collaborative Group, 1998; Office for National Statistics, 2000). Pharmacology & Therapeutics xx (2007) xxx xxx + MODEL JPT-05965; No of Pages 15 www.elsevier.com/locate/pharmthera Corresponding author. Tel.: +44 20 7188 1008. E-mail address: [email protected] (M.S. Marber). 0163-7258/$ - see front matter © 2007 Elsevier Inc. All rights reserved. doi:10.1016/j.pharmthera.2007.06.013 ARTICLE IN PRESS Please cite this article as: Clark, J. E. et al. Potential of p38-MAPK inhibitors in the treatment of ischaemic heart disease. Pharmacol Ther (2007), doi:10.1016/j. pharmthera.2007.06.013

Transcript of Potential of p38MAPK inhibitors in the treatment of ischaemic heart disease

1Q1

2

3

4

5

6

7

8

9

10

11

12

13

14

15

16

17

18

19

20

21

22

23

24

25

26

27

28

29

30

31

32

33

34

35

Pharmacology & Therapeutics xx (2007) xxx–xxx

+ MODEL

JPT-05965; No of Pages 15

www.elsevier.com/locate/pharmthera

ARTICLE IN PRESS

F

Associate editor: Madhani

Potential of p38-MAPK inhibitors in the treatment of ischaemic heart disease

James E. Clark, Negin Sarafraz, Michael S. Marber ⁎

The Cardiovascular Division, Kings College London, The Rayne Institute, St Thomas' Hospital, London, SE1 7EH, United Kingdom

O

DPR

OAbstract

Chronic heart failure is debilitating, often fatal, expensive to treat and common. In most patients it is a late consequence of myocardialinfarction (MI). The intracellular signals following infarction that lead to diminished contractility, apoptosis, fibrosis and ultimately heart failureare not fully understood but probably involve p38-mitogen activated protein kinases (p38), a family of serine/threonine kinases which, whenactivated, cause cardiomyocyte contractile dysfunction and death. Pharmacological inhibitors of p38 suppress inflammation and are undergoingclinical trials in rheumatoid arthritis, Chrohn's disease, psoriasis and surgery-induced tissue injury. In this review, we discuss the mechanisms,circumstances and consequences of p38 activation in the heart. The purpose is to evaluate p38 inhibition as a potential therapy for ischaemic heartdisease.© 2007 Elsevier Inc. All rights reserved.

EKeywords: p38; MAPK; Inhibitors; Heart failure; Ischaemia; Infarction

⁎ Corresponding aE-mail address:

0163-7258/$ - see fdoi:10.1016/j.pharm

Please cite this artpharmthera.2007.

T

Contents

ORRE

C

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 02. p38-Mitogen-activated protein kinase . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 03. Structure and function of p38-MAPK . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 04. Mechanisms of p38-MAPK activation. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0

4.1. Mitogen-activated protein kinase kinases. . . . . . . . . . . . . . . . . . . . . . . . . . . . 04.2. Autophosphorylation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0

5. p38, myocardial ischaemia and ischaemic heart disease . . . . . . . . . . . . . . . . . . . . . . . . 06. Pharmacological inhibitors of p38-MAPK. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 07. Clinical trials of p38-MAPK inhibition . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 08. Summary and conclusions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0

C

36

37

38

39

40

41

42

UN1. Introduction

Atherosclerotic vascular disease manifests predominantly asheart disease and stroke, which are the most frequent causesof death in the United Kingdom. Collectively atheroscleroticvascular disease was responsible for 40% of total mortality in theUnited Kingdom in 2004. Despite recent reductions in this high

43

44

45

46

uthor. Tel.: +44 20 7188 [email protected] (M.S. Marber).

ront matter © 2007 Elsevier Inc. All rights reserved.thera.2007.06.013

icle as: Clark, J. E. et al. Potential of p38-MAPK inhibitors in th06.013

mortality, morbidity is increasing as more patients survivemyocardial infarction (MI) and stroke. For example, about1.3 million people in the United Kingdom have survived acuteMI, 2 million have angina and 0.9 million have heart failure. Theextraordinarily high impact of atherosclerosis on the Nation'shealth is reflected in its economic cost. Atherosclerotic diseaseof just the coronary arteries costs the United Kingdom healthcaresystem £3500 million with an estimated further £4400 millionlost to the economy through premature death, illness and infor-mal care (The United Kingdom Heart Attack Study (UKHAS)Collaborative Group, 1998; Office for National Statistics, 2000).

e treatment of ischaemic heart disease. Pharmacol Ther (2007), doi:10.1016/j.

James Clark
Inserted Text
M.
James Clark
Inserted Text
'

UNCORRECTED PROOF

Table 1

t1:1 Study Model End point Inhibitor Outcome

t1:2 (Weinbrenneret al., 1997)

Ex vivo: buffer perfused rabbit heart subjectedto global ischaemia–reperfusion

p38 phosphorylation andcell viability (trypan blue)

SB203580 5 μM during ischaemia Inhibition of p38 activation abolishes protection inpreconditioned hearts and cardiomyocytes

In vitro: isolated rabbit cardiomyocytest1:4 (Wang et al., 1998a) In vitro: neonatal rat cardiac myocytes

cotransfected with p38α/β, activeMKK3/MKK6 or dominant negative isoforms

Cell survival, hypertrophicresponse, apoptosis

Modulation of p38α/β usingoverexpression of dominantnegative isoforms orupstream activators

The hypertrophic response in myocytes is mediated byp38β isoform, whereas over expression of p38α resultsin increase cell death

t1:5 (Nagarkatti & Afi, 1998) In vitro: ischaemia in rat myoblast cellline H9C2

Cell viability (MTT) SB203580 15 μM before/duringischaemia

SB203580 administered prior to ischaemia blockspreconditioning, but is protective during prolongedischaemia

t1:6 (Meldrum et al., 1998) Ex vivo: buffer perfused rat heart exposedto H2O2

LV function, coronary flow,CK release and tissue TNF

SB203580 1 mmol/min prior to insult p38 inhibition decreases myocardial TNF production,cardiomyocyte death and dysfunction

t1:7 (Mackay & Mochly-Rosen, 1999)

In vitro: simulated ischaemia in neonatalrat cardiomyocytes

LDH release SB203580 10 μM during ischaemia p38 inhibition, by SB203580, during ischaemia protectsagainst cardiomyocyte apoptosis

t1:8 (Ma et al., 1999) Ex vivo: buffer perfused rat heart subjectedto global ischaemia–reperfusion

LV Function, apoptosis andCK release

SB203580 10 μM before/duringischaemia

Inhibition of p38 attenuates reperfusion injury byreducing apoptosis and improving cardiac function

t1:9 (Craig et al., 2000) In vitro: neonatal rat cardiac myocytescotransfected with MKK6, TNF-α, TAK-1and IL-6

Apoptosis and IL-1 translationand transcription

SB203580 5 μM Activating both p38 and TNF-α augments myocardialsurvival during stress which is inhibited by SB203580

t1:10 (Hoover et al., 2000) In vitro: cardiac myocytes (transfectedwith MKK6) exposed to sorbitol

α-B crystallin expressionand phosphorylation;MAPKAP-K2 andp38 activation and apoptosis

SB203580 5 μM during insult Inhibition of p38 with SB203580 increased sorbitol-mediated apoptosis

t1:11 (Saurin et al., 2000) In vitro: simulated ischaemia in culturedrat neonatal cardiac myocytes

p38 phosphorylation, CKand LDH release andcell viability (MTT)

SB203580 10 μM during ischaemia Inhibition of p38 activation during prolonged ischaemiareduces injury and contributes to preconditioning-induced cardioprotection p38α is phosphorylated duringischaemia whereas p38β is deactivated

t1:12 (Yue et al., 2000) In vitro: simulated ischaemia–reperfusion inrat neonatal cardiomyocytes

LV function and apoptosis(TUNEL)

SB242719 10 μM Inhibition of p38/JNK leads to cardioprotection, whereasinhibition of ERK pathway exacerbates injuryt1:13 SB203580 10 μM

t1:14 (Mackay & Mochly-Rosen, 2000)

In vitro: simulated ischaemia–reperfusion inrat neonatal cardiomyocytes

p38 phosphorylation, apoptosisand LDH release

SB203580 10 μM during ischaemia Incubation with SB203580 during ischaemia–reperfusion attenuates cell death

t1:15 (Barancik et al., 2000) In vivo: regional ischaemia–reperfusion inpig heart

p38 phosphorylation andATF-2 phosphorylationand Infarct size

SB203580 1 μM during ischaemia Inhibition of p38 during ischaemia decrease infarct sizeand delays cell death

t1:16 (Marais et al., 2001) Ex vivo: buffer perfused rat heart subjectedto global ischaemia–reperfusion

p38 phosphorylation, LVfunction and apoptosis

SB203580 1-10 μM duringischaemia/reperfusion

p38 inhibition by SB203580 during ischaemia andreperfusion is cardioprotective

In vitro: neonatal cardiac myocytest1:18 (Gysembergh

et al., 2001)Ex vivo: buffer perfused rabbit heart subjectedto focal ischaemia–reperfusion

p38 activity and infarct size SB203580 1 μM during ischaemia Inhibition of p38 activity during coronary arteryocclusion is cardioprotective

t1:19 (Schneider et al., 2001) Ex vivo: buffer perfused rat heart subjected toglobal ischaemia–reperfusion

LV function and necrosis SB202190 10 μM before ischaemia p38 inhibition reduces ischaemic injury and does notblock protective effect of preconditioning

t1:20 (Martin et al., 2001) In vitro: simulated ischaemia in neonatal/adultrat cardiocytes overexpressing wild-typep38α MAPK

LDH release SB203580 10 μM during ischaemia SB203580 is cardioprotective through inhibition of p38isoform and not due to inhibition or activation of otherkinases

t1:21 (Rakhit et al., 2001) In vitro: simulated ichaemia–reperfusion injuryin neonatal rat cardiomyocytes

p42/44 and p38 MAPKphosphorylation

SB203580 1 μM during ischaemia SB203580 protected against injury, but p38β isoformdoes not contribute to survival

t1:22 (Sanada et al., 2001) In vivo: regional ischaemic preconditioning indog heart

HSP27 phosphorylation,arterial blood pressure,infarct size and collateral flow

SB203580 1 μM during ischaemia/reperfusion

SB203580 treatment during the preischemic andpostischaemic periods had no significant effect oninfarct size

2J.E

.Clark

etal.

/Pharm

acology&

Therapeutics

xx(2007)

xxx–xxx

ARTICLE

INPRESS

Please

citethis

articleas:C

lark,J.E.etal.P

otentialofp38-M

APK

inhibitorsin

thetreatm

entofischaem

icheartdisease.P

harmacolT

her(2007),doi:10.1016/j.

pharmthera.2007.06.013

UNCORRECTED PROOF

t1:24 (Tanno et al., 2003) Ex vivo: buffer perfused mouse heart subjectedto global ischaemia–reperfusion (mkk3−/−

and mkk3+/+)

Infarction/risk volume,p38, TAB1 and HSP27phosphorylation

SB203580 1 μM during ischaemia SB203580 sensitive ischaemic activation of p38 byTAB1-associated autophosphorylation contributes tomyocardial injury

In vitro: global ischaemia in H9c2 myoblastsexpressing wild-type and drug-resistant p38α

t1:26 (Sharov et al., 2003) In vitro: cardiomyocytes isolated from dogswith heart failure simulated by hypoxia,Angiotensin-II or nor-epinephrine

Apoptosis and Fas-L cyclinD1 expression

SB203580 10 μM during stress Inhibition of p38 activity attenuated stress-inducedapoptosis and reversed changes in Fas-L and cyclinD1 expression

t1:27 (Clanachan et al., 2003) Ex vivo: buffer perfused rat heart subjected tohypothermia-rewarming and global ischaemia–reperfusion

LV function SB202190 10 μM during rewarming/reperfusion

SB202190, when present during reperfusion, improvesrecovery of LV function Inhibition of p38 did not protectagainst rewarming-induced injury

t1:28 (Otsu et al., 2003) In vivo: regional Ichaemia–reperfusion inp38α+/+ and p38α+/− mice

p38α activity, infarct size andLV function

p38α+/− mice were used toknockdown p38 activity

Activation of p38α during ichaemia–reperfusion isdetrimental; reduction in p38α expression resultsin protection

t1:29 (Gorog et al., 2004) Ex vivo: buffer perfused mouse heart subjectedto low-flow global ischaemia–reperfusion

LV function, Coronary flowand apoptosis

SB203580 1 μM during ischaemia The p38 activation that accompanies short-termhibernation does not appear to contribute to thecontractile deficit

t1:30 (Koike et al., 2004) In vivo: canine heart transplantation from non-heart-beating donors

Cardiac output (CO) andLV function

FR167653 Dose not reported duringcold storage

Inhibition of p38 activation attenuates ischaemia–reperfusion injury in heart transplantation from non-heart-beating donors

t1:31 (See et al., 2004) In vivo: regional ischaemia in rat hearts LV function and postinfarctionremodelling

RWJ-67657 50 mg/day 7 days postMI for 21 days

RWJ-67657 treatment post-MI had beneficial effects onLV remodelling and dysfunction

t1:32 (Yada et al., 2004) In vivo: regional ichaemia–reperfusion inmouse hearts

Protein kinase activation andkinase activity, Nuclear factorκB activity, inflammatorycytokines and infarct size

FR167653 2 mg/kg i.p.before ischaemia

FR167653-mediated inhibition of p38 activity duringregional ichaemia–reperfusion injury reduces infarct size

t1:33 (Kaiser et al., 2004) In vitro: simulated ischaemia–reperfusion inneonatal cardiomyocytes expressing dominant-negative p38

Cell death, apoptosis, DNAfragmentation and Infarct size

Reduction of endogenous p38 usingoverexpression of dominant negativep38 isoform or upstream activation

p38α functions as a prodeath signalling effector in bothcultured myocytes as well as in the intact heart

In vivo: regional ischaemia–reperfusion indominant-negative MKK6 transgenic mice ordominant-negative p38α transgenic mice

t1:35 (Aleshin et al., 2004) Ex vivo: buffer perfused rat heart subjected toglobal ischaemia–reperfusion

TNFα mRNA expression; LVfunction and CK release

FR167653 1.0 mg/kg i.p. before I/R,and 1.0 mg/L during perfusion

FR167653 inhibited ichaemia–reperfusion-mediatedmyocardial TNFα production and p38 activation andimproved functional recovery

t1:36 (Martindale et al., 2005) In vivo: regional ischaemia–reperfusion inhearts from mice overexpressing cardiac-restricted wild type MKK6

LV dimensions (by echo);αB-crystallin expression;DNA fragmentation

p38 activity was modified by cardiacoverexpression of MKK6

Overexpression of MKK6 resulted in less myocardialdamage following ischaemia–reperfusion and enhancedfunctional recovery

t1:37 (Kabir et al., 2005) Ex vivo: buffer perfused mouse heart subjectedto global ischaemia–reperfusion in presence/absence of antimycin A

p38 and HSP27phosphorylation andinfarct size

SB203580 1 μM at the same time asantimycin A

Cardioprotection initiated by antimycin A is dependantupon p38 activation but independent of the upstreamkinase MKK3; however, during lethal ischaemia,inhibition of p38 activity was protective

t1:38 (Wang et al., 2005) Ex vivo: buffer perfused rat heart subjected toglobal ischaemia–reperfusion

p38, activation and cytokineexpression, activation ofcaspases and LV function

SB203580 20 μM before ichaemia Inhibition of p38 activation during ischaemia resulted inless MAPKAPK2, caspase-1, caspase-3 and caspase-11activation, and TNF, IL-1beta, IL-6 production aftermyocardial ichaemia as well as increasing functionalrecovery of the heart

t1:39 (Okada et al., 2005) In vitro: hypoxia-reoxygenation in neonatal ratcardiomyocyte

MAPKAP phosphorylation;Cyt. C release frommitochondria, caspase-3activation and LDH release

SB203580 10 μM throughout theexperiment

SB203580 abrogated activation of p38 MAPK,translocation of HSP27, and F-actin reorganization,prevented cytochrome C release, caspase-3 activation,and DNA fragmentation

(continued on next page) 3J.E

.Clark

etal.

/Pharm

acology&

Therapeutics

xx(2007)

xxx–xxx

ARTICLE

INPRESS

Please

citethis

articleas:C

lark,J.E.etal.P

otentialofp38-M

APK

inhibitorsin

thetreatm

entofischaem

icheartdisease.P

harmacolT

her(2007),doi:10.1016/j.

pharmthera.2007.06.013

UNCORRECTED PROOF

t1:40 Table 1 (continued )

t1:41 Study Model End point Inhibitor Outcome

t1:42 (Sumida et al., 2005) Ex vivo: buffer perfused rat heart subjected toglobal ischaemia–reperfusion

p38 and JNK activities LVcontractility, CK release,mitochondrial ATP generationand infarct size

SB203580 10 μM during reperfusion p38 inhibition exerts cardioprotection only whencontractile force-induced necrosis is prevented

t1:43 (Liu et al., 2005) In vivo: mouse hearts subjected to regionalischaemia–reperfusion

LV function, cardiacremodelling

SC-409 30 mg/kg/day given after MIfor 12 weeks

Inhibition of p38 MAPK attenuates cardiac remodellingand improves cardiac function in mice with heart failureafter infarction

t1:44 (House et al., 2005) Ex vivo: buffer perfused mouse heart subjectedto global low-flow ischaemia–reperfusion

Infarct size and p38 and HSP27phosphorylation

SB203580 2 μM during ischaemia/reperfusion

Inhibition of p38 during ichaemia–reperfusion injuryprotects against myocardial cell death

t1:45 (Li et al., 2005) In vitro: rat neonatal cardiomyocytesoverexpressing MKK6

LV remodelling, cytokinerelease and LV function

SB239068 20 μM and 1200 ppm indrinking water or culture mediumduring exposure

p38 inhibition prevents induction of inflammatorycytokines in cardiomyocytes and extracellularremodelling in heartIn vivo: hearts from MKK6bE transgenic mice

subjected to ischaemia–reperfusiont1:47 (Gupta et al., 2005) In vitro: adult rat ventricular myocytes

(ARVMs) during sepsisCell fractional shortening, cellviability (MTT), caspase-3activity

SB203580 10 μM before insult SB203580 pretreatment followed by bigET-1administration decreased p38 phosphorylation and down-regulated ET(B) receptor expression in sepsis group

t1:48 (Kim et al., 2006) In vitro:Hypoxia–re–oxygination in neonatalrat cardiomyocyte

Cell viability (trypan blue),apoptosis (TUNEL), necrosis,ROS generation

SB203580 1 μM during ischaemia/

reperfusion

Inhibition of p38α prevented hypoxia re–oxygenationinduced apoptosis of cardiomyocytes

t1:49 (Khan et al., 2006) Ex vivo: buffer perfused rat heart subjected toglobal ischaemia–reperfusion

Coronary flow, LV function,LDH release, infarct size andapoptosis (TUNNEL)

SB203580 10 μM before TNF-α Inhibition of p38 MAPK improved cardiac function afterreperfusion and attenuated ischaemic reperfusion-induced myocardial apoptosis and necrosis

t1:50 (Bellahcene et al., 2006) Ex vivo: buffer perfused mouse hearts frommkk3−/− mice subjected to TNF-α

LV function, p38 and HSP27phosphorylation

SB203580 1 μM before ischaemia Activation of p38 contributes to TNF-α inducedcontractile depression in intact heart and in isolatedcardiac myocytes through MKK3; inhibition of p38abolished contractile depression caused by TNFα

t1:51 (Engel et al., 2006) In vivo: regional ischaemia in rat hearts Left ventricular remodelling,fractional shortening andneovascularisation

SB203580 2 mg/kg at time of surgery SB203580 and FGF-1 induces cardiomyocyte mitosis,reduces scarring, and rescues function after MI

t1:52 (Li et al., 2006) In vivo: angiotensin and L-NAME-mediatedcardiac hypertrophy in rats

LV function, arterialinflammatory cell infiltration,and cardiomyocyte apoptosis

SD-282 60 mg/kg administered for4 days during treatment

SD-282, reduces inflammatory response and apoptosis,resulting in a reduction of myocardial damage, which, inturn, improves cardiac function following angiotensin IIand L-NAME treatment

t1:53 (Vahebi et al., 2007) In vitro: isolated skinned cardiac muscle fibrebundles

Cardiac myofilament function,Phosphorylation of α-Tmand TNI

Reduction of endogenous p38 usingoverexpression of dominant negativep38α isoform or upstream activation

Activation of p38α directly depresses saromeric functionby decreased phosphorylation of α-tropomyosin, whichis reversed by inhibition of p38α activity or by overexpressing dominant negative isoform

t1:54 (Riad et al., 2007) In vivo: diabetic mellitus induced by a singleinjection of streptozotocin

LV function, p38phosphorylation and peripheralICAM-1 and VCAM-1

SB239063 40 mg/kg/day for 43 daysafter induction of diabetes mellitus

Inhibition of p38MAPK reduced cardiac cell adhesionmolecules expression indicating both antiinflammatoryand vasculoprotective effects in the diabetic heart

t1:55 (Clark et al., 2007) In vivo: regional ischaemia in hearts frommkk3−/− and mkk3+/+ mice

LV function, LV remodelling,p38 and HSP27phosphorylation

Knockout of MKK3 Postinfarction LV remodelling continues in the absenceof MKK3 as does p38 activation

4J.E

.Clark

etal.

/Pharm

acology&

Therapeutics

xx(2007)

xxx–xxx

ARTICLE

INPRESS

Please

citethis

articleas:C

lark,J.E.etal.P

otentialofp38-M

APK

inhibitorsin

thetreatm

entofischaem

icheartdisease.P

harmacolT

her(2007),doi:10.1016/j.

pharmthera.2007.06.013

47

48

49

50

51

52

53

54

55

56

57

58

59

60

61

62

63

64

65

66

67

68

69

70

71

72

73

74

75

76

77

78

79

80

81

82

83

84

85

86

87

88

89

90

91

92

93

94

95

96

97

98

99

100

101

102

103

104

105

106

107

108

109

110

111

112

113

114

115

116

117

118

119

120

121

122

123

124

125

126

127

128

129

130

131

132

133

134

135

136

137

138

139

140

141

142

143

144

145

146

147

148

149

150

151

152

153

154

155

156

5J.E. Clark et al. / Pharmacology & Therapeutics xx (2007) xxx–xxx

ARTICLE IN PRESS

UNCO

RREC

Atherosclerotic plaque rupture/erosion results inMI, which ischaracterised by necrosis and apoptosis of cardiomyocytes.Although the acute condition alone may result in death due toventricular arrhythmias or pump failure, in the patients withsubstantial infarction that survive, a chronic phase of ventricularremodelling occurs. Remodelling, is a maladaptive processcharacterised by cardiomyocyte apoptosis, fibrosis, thinning ofthe ventricular wall at the site of infarction, ventricular chamberenlargement and hypertrophy of surviving cardiomyocytes(Pfeffer & Braunwald, 1990; Swynghedauw, 1999; Udelsonet al., 2003). These events may, eventually, lead to heart failurewhich is frequently lethal despite current best care. Therefore,intervention to minimise pathological cardiac remodelling ishighly desirable to reduce the mortality and the incidence andseverity of congestive heart failure after MI.

Various intracellular signalling pathways are thought to playa critical role in the myocardial response to ischaemia andconsequent pathological remodelling. Multiple mitogen-acti-vated protein kinase (MAPK) are activated during ischaemiaand may contribute to the structural and functional changes.MAPK are highly conserved serine/threonine kinases that areactivated by a dual phosphorylation of a Thr-X-Tyr motif, inresponse to wide a variety of stimuli such as cytokines, osmoticand other environmental stresses and consequently play a role innumerous cell functions including growth and proliferation(English et al., 1999; Pearson et al., 2001). Three of the fivemajor MAPK cascades have been extensively studied in theheart: extracellular signal-regulated kinase (ERK1 and ERK2),c-Jun N-terminal kinases (JNK1 and JNK2) and p38 kinases. Ithas been shown that JNK and p38 contribute to, whereas ERK/ERK2 protect against, apoptotic cell death. Although the mech-anisms by which p38 and JNK induce apoptosis may be cell andstimulus specific, there is overwhelming evidence that theactivation of p38-MAPK (or p38) that occurs during prolongedischaemia accelerates injury since its inhibition by pharmaco-logical or genetic means slows the rate of infarction/death(Saurin et al., 2000; Martin et al., 2001; see Table 1). Althoughthis evidence is based on animal data, it seems likely similarmechanisms operate in the human heart since p38 is identicallyactivated by ischaemia (Han et al., 1995; Cain et al., 1999; Cooket al., 1999; Lee et al., 2000; Lemke et al., 2001) and earlyclinical trials indicate a potential benefit (de Winter et al., 2005).Thus, superficially at least, inhibitors of p38 have therapeuticpotential in ischemic heart disease (Force et al., 2004).

2. p38-Mitogen-activated protein kinase

p38-MAPK are activated by a wide range of extracellularinfluences, including radiation, ultraviolet light, heat shock,osmotic stress, proinflammatory cytokines such as interleukin(IL)-1 and tumour necrosis factor (TNF)-α, and certain mitogens(Sugden & Clerk, 1998) in addition to myocardial ischaemia(Bogoyevitch et al., 1996; Saurin et al., 2000; Luss et al., 2000;Ping & Murphy, 2000). Furthermore, the consequent activationof p38-MAPK is intimately involved in multiple cellular re-sponses, including growth, proliferation, differentiation, anddeath (English et al., 1999; Ono & Han, 2000). Perhaps not

Please cite this article as: Clark, J. E. et al. Potential of p38-MAPK inhibitors in thpharmthera.2007.06.013

TEDPR

OOF

surprisingly these cellular effects have clear consequence(s),translating into involvement in complex pathophysiologies,such as wound healing (Lim et al., 1998), inflammatory arthritis(Badger et al., 1996), sepsis (Kotlyarov et al., 1999), acute res-piratory distress syndrome (Carter et al., 1999), and malignanthypertension (Behr et al., 2001).

Four p38 isoforms (α,β, δ and γ) exist, which have preservedstructure but variable sensitivity to pharmacological inhibition.All 4 isoforms have a Thr180-Gly181-Tyr182 (TGY) dual phos-phorylation motif which is used by investigators to infer acti-vation. p38α and β have high sequence homology and sharesensitivity to pharmacological inhibition by prydinyl imidazolemolecules (such as SB203580) but have only 60% homologywith p38γ and δ, which are resistant to SB203580 (SB) inhi-bition (Eyers et al., 1999). Of the SB-sensitive isoforms, p38α isthe predominant form in human and rodent myocardium (Lemkeet al., 2001; Rakhit et al., 2001; Sanada et al., 2001; Braz et al.,2003). Studies with knockout mice and cells have shown thatp38α is essential for embryonic development as knockout of theα isoform results in embryonal lethality, but mice lacking p38β,p38γ, and p38δ are viable (Allen et al., 2000; Adams et al., 2000;Tamura et al., 2000; Brancho et al., 2003).

3. Structure and function of p38-MAPK

The catalytic site of p38 lies in a pocket between the N- andC-terminal domains. These domains are connected by a singlehinge and the L16 loop of the C-terminal domain which wrapsback around the N-terminal domain and controls the relationshipbetween the relatively rigid domains (see Fig. 2). In addition, inthe inhibitor bound nonphosphorylated state, there is a mis-alignment between the N- and C-lobes which prevents thecooperation between a lysine residue (Lys53) in the N-terminallobe and aspartic acid residue (Asp168) in the C-terminal lobe,imperative to binding and stabilization of the α phosphate groupand adjacent ribose of ATP, respectively (Wilson et al., 1996;Gum et al., 1998). Therefore, it is widely thought that thenondual phospho- form of p38 is inactive as a result of stericobstruction of the peptide-binding channel and low ATP affinity.

Thr180 and Tyr182 are located on a flexible “activation loop”that guards the active site. Dual phosphorylation of these 2amino acids in response to exotoxin, cytokines, physical stress(such as hyperosmolarity), and chemical oxidant stress, such ashydrogen peroxide (Han et al., 1994; Freshney et al., 1994;Rouse et al., 1994; Raingeaud et al., 1995) is thought to cause theactivation loop to refold and move out of the peptide-bindingchannel. This movement is then thought to exert a “crank-handle” effect on the overall tertiary structure of the kinasereorienting the N- and C-, terminal lobes so that Lys53 andAsp168 move towards one another by 2.5–5 Å. This alters theconformation of the catalytic site enabling the cooperationnecessary for ATP binding and allowing substrate access(Wilson et al., 1996; Diskin et al., 2007). The docking groovesused by substrates and activators consist of 2 regions (see Fig. 2),the CD region and the ED region (Tanoue et al., 2000). The CDregion is part of a shallow groove formed by the acidic residuesAsp313, Asp316, Glu81, and the aromatic residues Phe129 and

e treatment of ischaemic heart disease. Pharmacol Ther (2007), doi:10.1016/j.

James Clark
Cross-Out
James Clark
Replacement Text
endo

157

158

159

160

161

162

163

164

165

166

167

168

169

170

171

72

73

74

75

76

77

78

79

80

81

82

83

84

85

86

87

88

89

6 J.E. Clark et al. / Pharmacology & Therapeutics xx (2007) xxx–xxx

ARTICLE IN PRESS

Tyr311. The ED region is part of a deeper groove formed byresidues 159–163 at one side and residues Gln120, His126 andPhe129 at the opposite side (Haar et al., 2007). It is believed thatthese 2 binding regions facilitate activator (MAPK kinase 3,MKK3) and substrate (MK2 and MEF2) binding (Chang et al.,2002; Haar et al., 2007).

4. Mechanisms of p38-MAPK activation

4.1. Mitogen-activated protein kinase kinases

Although the intracellular activation cascade for p38 undermost physiological conditions is still unclear, several upstreamMAPK kinases (MKK) have been identified from in vitroanalysis, including MKK3 and MKK6 (Derijard et al., 1995;Han et al., 1996). MKK4 is predominately involved in JNKactivation but is able to activate p38-MAPK, at least, in vitro(Deacon & Blank, 1997). Using MKK-targeted mouse lines, it

UNCO

RREC

Fig. 1. Mechanisms of p38-MAPK activation. Classical activation by MKK3/MKmechanism ②. TCR-mediated Tyr323 phorphorylation by ZAP70 is depicted as maddition, TAB1 is a p38 substrate. PhosphoTAB1 is less able to activate TAK1 (★).and phosphoTAB-1. Heavy lines represent an interaction; dotted lines represent a m

Please cite this article as: Clark, J. E. et al. Potential of p38-MAPK inhibitors in thpharmthera.2007.06.013

OOF

1has been shown that, in response to most stress stimuli, MKK31and MKK6 are the principal MKK activating p38α and β,1respectively (see Fig. 1). MKK3 and MKK6 are in turn acti-1vated by phosphorylation by a MKK kinase (MKKK). The1MKKK, being responsible for activation of the p38 cascade,1appears to be cell type and stimulus specific, and several have1been implicated (Yamaguchi et al., 1995; Moriguchi et al.,11996; Ichijo et al., 1997; Hutchison et al., 1998; Gallo &1Johnson, 2002; Ge et al., 2002; Cheung et al., 2003).1However, p38 activation is not limited to this traditional1phospho-relay signalling cascade. Since SB203508 (the most1widely used p38 kinase inhibitor) occupies the catalytic site,1without inhibiting upstream MKK, it should only inhibit the1phosphorylation events downstream of p38 without inhibiting1the dual-phosphorylation of p38 itself (Young et al., 1997).1However, certain conditions, such as myocardial ischaemia,1cause a SB-sensitive form of p38 dual phosphorylation. Two1mutually exclusive explanations for these observations are

TEDPR

K6 is depicted as mechanism ①. TAB1-induced autoactivation is depicted asechanism ③. TAB1 activates TAK1, which in turn activates MKK3/MKK6. InPharmacological inhibition of p38-MAPK diminishes p38 dual phosphorylationodification (phosphorylation); open arrows represent a multi-element pathway.

e treatment of ischaemic heart disease. Pharmacol Ther (2007), doi:10.1016/j.

190

191

192

193

194

195

196

197

198

199

200

201

202

203

204

205

206

207

208

209

210

211

212

213

214

215

216

217

218

219

220

221

222

223

224

225

226

227

228

229

230

231

232

233

234

235

236

237

238

239

240

241

242

243

244

245

246

247

248

249

250

251

252

253

254

255

256

257

258

259

260

261

262

263

264

265

266

267

268

269

270

271

272

273

274

275

276

277

278

279

280

281

282

283

284

285

286

287

288

289

290

291

292

293

294

295

296

297

298

299

7J.E. Clark et al. / Pharmacology & Therapeutics xx (2007) xxx–xxx

ARTICLE IN PRESS

UNCO

RREC

(i) that p38 is able to autophosphorylate its activation loop or(ii) that SB203580 inhibits a kinase upstream of p38 involved inits activation by trans-phosphorylation during ischaemia.

4.2. Autophosphorylation

Ge and co-workers elegantly reported that auto-phosphory-lation of p38 can occur, facilitated by an interaction with the non-enzymatic adaptor protein transforming growth factor-β-activated protein kinase-1 (TAK1) binding protein-1 (TAB1;Ge et al., 2002). TAB1 is known to perform a similar function byinducing the autophosphorylation of TAK1, which in turnactivates MKK3/MKK6. In vitro co-expression experimentshave shown that the interaction of TAB1 and p38α leads tophosphorylation of the TGY activation motif. TAB1-dependentp38α activation appears to play a role in the injury responseduring myocardial ischaemia (Tanno et al., 2003; Fiedler et al.,2006), myocyte-derived dendritic cell maturation (Matsuyamaet al., 2003), and peripheral T-cell anergymaintenance (Ohkusu-Tsukada et al., 2005). The interpretation of the TAB1-p38interaction was, however, complicated by Cohen's group whodemonstrated that the phosphorylation of TAB1 on Ser423 andTyr431 was p38-MAPK-dependent and hence prevented bySB203580. The authors proposed a feedback control mechanismof TAK1 activity, whereby p38 activity inhibits TAK1, throughthe phosphorylation of TAB1. Inhibition of p38 activity (bySB203580) abolishes this feedback control of TAK1, causingunopposed activation of the parallel JNK pathway andconsequently IKK (Cheung et al., 2003). This is depicted inFig. 1. Although TAB1 andMKK3/MKK6 mechanisms interactthrough the potential modulation of TAK1, other possibilitiesalso exist. For example there is some evidence to suggest thatTAB1 causes p38 redistribution to the cytoplasm and mayrestrict access to downstream targets, such as MAPK-activatedprotein kinase 2 (MAPKAPK2). This is in direct contrast to thepattern seen with MKK3/MKK6 (Lu et al., 2005).

Using MKK3/MKK6 double knockout and MKK4/MKK7double knock out mouse embryonic fibroblasts (MEF), Kanget al. have shown that peroxynitrite-induced phosphorylation ofp38α is associated with an ∼85 kDa disulfide complex in wildtype MEF (Kang et al., 2006). This association was diminishedin MKK3/MKK6 knockout MEF (Kang et al., 2006). Theauthors suggested that phosphorylation of p38 mediated byTAB-1 can be modulated by a yet unknown binding partner(s) ina manner dependent on a disulfide complex (Kang et al., 2006).

In addition to TAB-1-mediated activation, p38 can alsoautophosphorylate through an alternative pathway in responseto T-cell antigen receptor (TCR) activation (Dong et al., 2002;Rincon & Pedraza-Alva, 2003). In this pathway, activation ofTCR leads to recruitment of a Syk family kinase, ZAP-70,which directly phosphorylates p38 on Tyr323 (Rouse et al.,1994). In a recent study it was shown that the phosphorylationof p38 on Try323 can be blocked in the presence of the DNAdamage inducible gene Gadd45a, an autoimmune suppressor.Absence of Gadd45a has been shown to result in chronicphosphorylation of p38, T-cell hyperproliferation and autoim-munity (Salvador et al., 2005a, 2005b). Following ZAP-70

Please cite this article as: Clark, J. E. et al. Potential of p38-MAPK inhibitors in thpharmthera.2007.06.013

TEDPR

OOF

phosphorylation of p38, an autophosphorylation event similarto that induced by interaction with TAB-1 occurs resulting indual phosphorylation, and activation of the kinase.

However, regulation of p38 kinase activity in vitro, at least isnot solely dependent on upstream kinases and binding partners.Diskin and co-workers, using a in vitro mutation approach, madeintrinsically active p38 isoforms based on activating mutationspreviously found in the yeast MAPK kinase p38/Hog1 (Bellet al., 2001). Single andmultiple point mutations of human p38αresulted in high intrinsic activity independent of activation bydual phosphorylation. Structural analysis of these p38 mutantshas identified a hydrophobic core stabilised by 3 aromaticresidues, Tyr69, Phe327 and Trp337, in the vicinity of the L16Loop region. It is believed that the hydrophobic core is an in-herent stabiliser that maintains the low basal activity level ofunphosphorylated p38 (Diskin et al., 2004). Upon activation,however, a segment of the L16 Loop, including Phe327 becomesdisordered allowing ATP and substrate binding. The mutation ofthese amino acids involved in the hydrophobic core results in theconformational changes imposed naturally by dual phosphory-lation, namely destabilising the hydrophobic core and lockingthe kinase in a constitutively active state. In addition, in thisactive state, p38 is able to autophosphorylate in an invitro kinaseassay (Diskin et al., 2004). More recently p38β, p38γ and p38δmutants were similarly constructed (Askari et al., 2007). In thesemutants, a highly conserved aspartic acid located in the acti-vation loop (Asp170 in Hog-1; Asp176 in p38α, p38β and p38δ;and Asp179 in p38) was mutated. The spontaneous kinaseactivity of p38β, p38γ and p38δ appeared to be lower than thedual phosphorylated wild-type isoforms whereas the p38αisoform presented the highest spontaneous activity. Therefore, itis apparent that modifications of the amino acids in the hydro-phobic core along with the mutations in Asp176 are capable ofactivating p38s (Askari et al., 2007), the former likely explainingthe mechanism by which ZAP-70 induces autoactivation(Mittelstadt et al., 2005). In addition, these mutants provide atool to dissect isoform-specific downstream signalling (Askariet al., 2007).

5. p38, myocardial ischaemia and ischaemic heart disease

Ischaemic heart disease remains the leading cause of death,accounting for approximately 1 quarter of all deaths in theUnited Kingdom. Currently, the most effective method ofreducing mortality in such patients is to achieve rapidreperfusion by lysis or mechanical disruption of the occlusivecoronary thrombus and plaque. The mortality from acute MIunder these circumstances is inversely related to the amount ofmyocardial salvage achieved by reperfusion.

There is increasing evidence from preclinical investigationsthat inhibition of p38 during prolonged ischaemia slows the rateof infarction/death and inhibits the production of inflammatorycytokines, such as TNF-α, IL-1 and IL-8, which aggravateischaemic injury (Young et al., 1997; see Table 1 for summary).It was fist demonstrated as early as 1996 that p38α and βare activated in response to ischaemia and reperfusion in theheart (Bogoyevitch et al., 1996). Since then, using gene transfer

e treatment of ischaemic heart disease. Pharmacol Ther (2007), doi:10.1016/j.

C

300

301

302

303

304

305

306

307

308

309

310

311

312

313

314

315

316

317

318

319

320

321

322

323

324

325

326

327

328

329

330

331

332

333

334

335

336

337

338

339

340

341

342

343

344

345

346

347

348

349

350

351

352

353

354

355

356

57

58

59

60

61

62

63

64

65

66

67

68

69

70

71

72

73

74

75

76

77

78

79

80

81

82

83

84

85

86

87

88

89

90

91

92

93

94

95

96

97

98

99

00

01

02

03

04

05

06

07

08

09 Q2

10

11

8 J.E. Clark et al. / Pharmacology & Therapeutics xx (2007) xxx–xxx

ARTICLE IN PRESS

UNCO

RRE

techniques, the α isoform that has been implicated in myocyteapoptosis, consistent with the findings that this isoform alonecontributes to cell death following ischaemia (Saurin et al.,2000; Martin et al., 2001). p38s phosphorylate a number ofknown transcription factors to alter their transactivating po-tential influencing gene expression. However, the immediatedownstream targets of p38 that aggravate myocardial injuryare still largely unknown. One downstream substrate of p38αis MAPKAP2, which can, in turn, phosphorylate HSP27, aheat shock protein, which is thought to confer a number of pro-tective effects (Kim et al., 2005). In addition, phosphorylation ofMAPKAP2 can also result in phosphorylation of factors thattransactivate cytokine genes, such as TNF-α, a cytokine im-plicated in chronic heart failure. Interestingly, TNF-α alsoactivates p38 and thus p38 has been considered as the keystonein an autoamplifying cytokine cascade by most investigators andan attractive target for antiinflammatory drug development (Leeet al., 2000; Kuma et al., 2005).

Furthermore, a proapoptotic role for p38α and/or p38βduring myocardial ischaemia is suggested by protection of car-diac myocytes from ischaemic damage using a selective p38α/p38β isoform inhibitor, SB203580 (Wang et al., 1998a). Usingadenoviral-mediated expression of p38α and p38β in rat neo-natal cardiomyocytes our group have previously shown that after2.5 hr simulated ischaemia p38α was activated, whereas p38βactivation was significantly inhibited (Saurin et al., 2000).Inhibition of p38α activation during prolonged ischaemia, butnotβ, resulted in an increase in cell viability (Saurin et al., 2000).This strongly supported Wang et al. who suggested that p38αactivation in cardiac myocytes is sufficient to cause apoptosiswhereas activation of the β isoforms leads to protection andhypertrophy (Wang et al., 1998a). However, there is some evi-dence to suggest that p38 isoforms may have potential protectivefunction and suggest a possible adverse effect of prolonged p38inhibition in the heart. Glembotski's laboratory have demon-strated chronic activation of p38 through overexpression ofMKK6 in the heart can result in improved functional recoveryfrom ischaemia and MI (Martindale et al., 2005). The protectiverole of p38β has also been investigated in a recent study by Kimand co-workers who have shown that activation of p38β bycarbon monoxide promotes the nuclear translocation of heatshock factor-1 (HSF-1), which regulates the expression ofcytoprotective HSP70 in cells and tissues (Kim et al., 2005).HSF-1 can also serve as a negative regulator of proinflammatorygenes, including IL-1β, and TNF-α (Xie et al., 2002). The roleof p38s (the α isoform predominantly) in myocardial ischaemicinjury has been studied extensively since the findings of (Wanget al. 1998a). These studies, which in the main suggest p38activation during ischaemia worsens injury and depresses LVfunction, are too numerous to review and appear in Table 1.

There is little information in the literature regarding the rolesof either the γ and δ isoform of p38 in the myocardium duringischaemia. Conserved cardiac expression of p38γ amongst sev-eral different species suggests that this isoform may play animportant role in the heart and therefore is unlikely to be func-tionally redundant (Court NW et al., 2002). p38γ is localised inthe cytoplasm of the cardiac myocyte and is reported to have a

Please cite this article as: Clark, J. E. et al. Potential of p38-MAPK inhibitors in thpharmthera.2007.06.013

TEDPR

OOF

3punctate distribution (Court NW et al., 2002), and p38δ mRNA3is broadly expressed in a wide variety of mouse and human3tissues including the heart (Wang et al., 1997; Beardmore et al.,32005). The C-terminal tail of p38γ allows its interaction with3PDZ domains of its substrate protein(s) and association with3α1-syntrophin and SAP90/PSD95 in skeletal muscle and3neuronal synapses, respectively (Zhang et al., 2003; Hasegawa3& Cahill, 2004; Sabio et al., 2004). p38γ-catalysed phosphory-3lation of hDlg (the mammalian homologue of the Drosophila3tumour suppressor Dlg) triggers its dissociation from the3cytoskeleton, indicating that this may regulate the integrity of3intercellular-junctional complexes, cell shape, volume and cell3polarity in response to many kinds of external stimuli. In support3of these findings, Parker et al. identified a novel p38δ substrate as3stathmin, a cytoplasmic protein that was previously reported to be3a substrate of several intercellular signalling kinases which have3been linked to regulation of microtubule (MT) dynamics in a3phosphorylation-dependent manner (Belmont & Mitchison,31996). This may suggest that a common theme in p38 pathway3activation may be the re-organisation of the cytoskeletal frame-3work to enhance cell survival in times of stress such as ischaemia3(Parker et al., 1998). Moreover, both p38γ and p38δ phos-3phorylate the MT-associated protein Tau in neurons in vivo.3Hyperphosphorylated Tau is the major component of the paired3helical filaments, which constitute one of the main neuropatho-3logical hallmarks of many neurodegenerative disorders (Sabio3et al., 2005). So, although there is only circumstantial evidence to3support a role for γ and δ isoforms in the heart, this is likely an3emerging area of research. In the absence of isoform-selective3pharmacological inhibitors, this is, to some extent, aided by the3availability of a number of p38 isoform-targeted mouse lines3(Beardmore et al., 2005; Sabio et al., 2005) and spontaneously3active mutants (Askari et al., 2007).

36. Pharmacological inhibitors of p38-MAPK

3Early efforts in drug discovery of small molecule inhibitors of3kinases were met with scepticism that selectivity could ever be3accomplished, due to the high degree of structural similarity in3the adenosine binding pocket among the entire kinome. Thus, it3was somewhat of a surprise when SB203580, the first reported3p38 inhibitor, emerged showing selectivity over the closely3related JNK and ERKMAPK families (Lantos et al., 1984). The3pyridinyl imidazole antiinflammatory agents were soon shown3to be highly selective p38 inhibitors and the bi-cyclic pyridinyl4imidazole SKF-86002 was the first compound reported to inhibit4LPS-stimulated cytokine production (Lee et al., 1988, 1994). It4was not long before investigators explored dual 5-lipooxygen-4ase/cyclooxygenase (LO/COX) and cytokine inhibition as4potential mechanisms for the potent anti-inflammatory activity4of these compounds (Lee et al., 1993), and subsequently,4SB203580 was used as a pharmacological inhibitor to study the4cascade of kinases (via p38) involved in cytokine production4(Gallagher et al., 1997). The structures of representative classes4of p38 inhibitors are shown in Fig. 3.4The crystal structures of pyridinyl imidazole-p38α complexes4have recently become available and suggest that SB203580 binds

e treatment of ischaemic heart disease. Pharmacol Ther (2007), doi:10.1016/j.

412

413

414

415

416

417

418

419

420

421

422

423

424

425

426

427

428

429

430

431

432

433

434

435

436

437

438

439

440

441

442

443

444

445

446

447

448

449

450

451

452

453

454

455

456

457

458

459

460

461

462

463

464

465

9J.E. Clark et al. / Pharmacology & Therapeutics xx (2007) xxx–xxx

ARTICLE IN PRESS

to the active site of both phosphorylated (active) and unpho-sphorylated (inactive) p38 in an ATP-competitive manner (seeFigs. 2 and 3). These inhibitors bind to an aryl-specificity pocketbehind the site, which is normally occupied by the adenine ring ofATP. The interaction occurs between the 4-pyridinyl group(analogous to the N-1 adenine of ATP) and the N-H of Met109. Inaddition, studies have also have implicated Thr106 as a key residueconferring selectivity (Wilson et al., 1996; Gum et al., 1998). The2 adjacent residues, His107 and Leu108, alongwith Thr106 lie at theback of the ATP pocket and are identical in p38α and p38β, butare different in p38γ and p38δ (Met106, Pro107 and Phe108, re-spectively) which are insensitive to SB203580 inhibition. Using amutagenesis approach it has been shown that if these 3 residues inp38α and p38β are changed to Met-Pro-Phe (as found in p38γand p38δ) the mutant kinase is no longer inhibited by SB203580.By contrast, introduction of the Thr-His-Leu sequence of p38αinto p38γ or p38δ confers sensitivity to SB203580. Takentogether, these studies have identified Thr106 as the key residueforming the aryl-specificity pocket (Saccani et al., 2002).

In addition to novel prydinyl imidazole compounds, a newgroup of selective p38 inhibitors are the arly-pyridinyle-hetero-cylces. In these compounds, the imidazole core is replaced byother heteroaryl scaffolding, and consequently these compoundsgenerally exhibit in vitro potency similar to prydinyl imidazoles(Boehm et al., 2001). Other structurally diverse p38 inhibitorsinclude a subset of novel non-aryl-pyridinyls such as triaza-napthalenones, N,N′-diary ureas, benzzophenones, pyrazole

UNCO

RREC

466

467

468

469

470

471

472

473

474

475

476

477

478

479

480

481

482

483

484

485

486

487

488

489

490

491

492

493

494

495

Fig. 2. Crystal structure of p38 with SB203580 occupying the ATP binding site.The Thr106 residue (4), which is important for binding of pyridinyl imidazoleinhibitors, and the 2 residues within the activation loop that are phosphorylated(Thr180 (2) and Try182 (1)) are highlighted. Tyr323 (3), which has been implicatedin TCR-mediated activation of p38 is also shown. SB203580 is shown in green.The activation loop is shown in orange. ED and CD activator/substrate bindingregions are highlighted. The C-terminal extension that forms the L16 loopbridging the domains is also indicated (created in JenaLib Jmol; PDB entry1a9u; Wang et al., 1998b).

Please cite this article as: Clark, J. E. et al. Potential of p38-MAPK inhibitors in thpharmthera.2007.06.013

TEDPR

OOF

ketones, indole amides, diamides, quinazolinones, and pyridy-laino-quinazolines (Cirillo et al., 2002). Unlike the imidazole-based p38α inhibitors, the urea-containing inhibitors act in anoncompetitive manner (Kulkarni et al., 2007). Crystallographicstudies of urea-containing p38α inhibitors, such as BIRB-796,have revealed that these compounds bind to at a site remote fromATP pocket, and induce a significant movement of Phe169, suchthat this residue fills the ATP pocket, preventing ATP binding(Pargellis et al., 2002). Thus, inhibitors of p38 can be dividedinto 2 groups dependent upon their mode of binding to p38; theseare (i) active site or “gatekeeping” inhibitors (such as SB203580)and (ii) those which bind remotely and interfere with ATP bind-ing indirectly (such as BIRB-796).

Enthusiasm for “blanket” pharmacological inhibition of p38is tempered by the fact that this kinase is involved in innu-merable biological processes and therefore not surprising thatunder many circumstances its activation leads to myocardialprotection rather than injury (Weinbrenner et al., 1997; Craiget al., 2000; Hoover et al., 2000; Communal et al., 2000; Forceet al., 2004; Zheng & Zuo, 2004; Martindale et al., 2005). Thisparticularly seems to be the case when p38 activation occurs as aconsequence of an intervention that precedes lethal myocardialischaemia, such as ischaemic or pharmacological precondition-ing (Nagarkatti & Afi, 1998; Marais et al., 2001; Sanada et al.,2001). However, in these studies the same inhibitor, at the sameconcentration, reduces injury if present solely during lethalischaemic injury (Nagarkatti & Afi, 1998; Marais et al., 2001;Sanada et al., 2001; Tanno et al., 2003). The cause of this ap-parent paradoxical observation may relate to an attenuation ofp38 activation during lethal ischaemia by its prior transientactivation. Thus there is ample evidence from cardiac as well asother research fields that p38 activation can have beneficialconsequences whilst it is also incontrovertible that restrictingp38 inhibition to the activation that accompanies lethalmyocardial ischaemia reduces infarction (Nagarkatti & Afi,1998; Marais et al., 2001; Sanada et al., 2001; Marais et al.,2005; Table 1).

However, if we are to consider currently available p38inhibitors with the aim of treating chronic conditions, it is likelygreater levels of selectivity will be required to avoid the inhi-bition of beneficial forms of activation. As more kinases areinvestigated, a better understanding of selectivity over thekinome has followed. With expanded kinase panels, it has nowemerged that “classic” p38α inhibitors like SB203580, whichwere once described as being selective over certain kinases, nowhave been shown to have similar, and sometimes lower IC50s.These actions are a result of similarities in the inhibitory bindingsite or in particular the hydrophobic pocket equivalent to thatformed by Thr106 (Fabian et al., 2005). So if broad inhibition ofp38 is not the answer, what is? At present, the majority ofpharmacological inhibitors of p38 are selective for the α and βisoforms of the kinase. It is clear from the published data that theduring prolonged ischaemia the α isoform plays an importantrole in the progression of dysfunction. Perhaps a more rationalapproach to inhibit p38 in a site- and condition-specific mannermight be to target the activation of the kinase pathway upstreamof p38 itself, such as TAB1 or MKK3/MKK6. Using a model of

e treatment of ischaemic heart disease. Pharmacol Ther (2007), doi:10.1016/j.

UNCO

RREC

TEDPR

OOF

496

497

498

99

00

01

Fig. 3. Structure of representative classes of p38 MAPK inhibitors. p38 inhibitors can be divided into 2 groups dependant upon their mode of binding to p38; active siteinhibitors, such as SB203580 and RJW-67657, bind competitively to the ATP site of the enzyme whereas others bind remotely and interfere with ATP bindingindirectly (such as BIRB-796).

10 J.E. Clark et al. / Pharmacology & Therapeutics xx (2007) xxx–xxx

ARTICLE IN PRESS

coronary artery ligation in a mkk3-targetted mouse line, we haverecently demonstrated that removing MKK3 does not alterpathological remodelling and progression to ventricular dys-

Please cite this article as: Clark, J. E. et al. Potential of p38-MAPK inhibitors in thpharmthera.2007.06.013

4function after MI (Clark et al., 2007). Maybe this is not sur-5prising considering the multitude of pathways involved in5ischaemia and inflammation but it does, perhaps, highlight the

e treatment of ischaemic heart disease. Pharmacol Ther (2007), doi:10.1016/j.

502

503

504

505

506

507

508

509

510

511

512

513

514

515

516

517

518

519

520

521

522

523

524

525

526

527

528

529

530

531

532

533

534

535

536

537

538

539

540

541

542

543

544

545

546

547

548

549

550

551

552

553

554

555

556

557

558

559

560

561

562

563

564

565

566

567

568

569

570

571

572

573

574

575

576

577

578

579

580

581

582

583

584

585

586

587

588

589

590

591

592

593

594

595

596

597

598

599

600

601

602

603

604

605

606

607

608

609

610

611

612

613

614

615

11J.E. Clark et al. / Pharmacology & Therapeutics xx (2007) xxx–xxx

ARTICLE IN PRESS

UNCO

RREC

potential importance of other pathways which warrant furtherinvestigation.

7. Clinical trials of p38-MAPK inhibition

Despite the apparent dichotomy in preclinical research of theconsequences of p38 inhibition, 2 types of molecular inhibitorsof p38, aryl-prydinyl heterocycles (namely SB242235 and RWJ-67657) and non-aryl-prydinyl heterocycles (VX-745, BIRB-796and RO3201195), have so far advanced to clinical trials. Resultsof phase I and early phase II trials were also promising. In onestudy, orally administered SB242235 (1–500 mg), which hasbeen shown to have potent antiinflammatory effects in a ratmodel of arthritis (Badger et al., 2000), was well tolerated andsuppressed production of TNF-α, IL-1β, IL-6, and IL-8 in adose-dependent manner within 3 hr (Adams et al., 2001). Arandomised clinical study to examine the efficacy of RWJ-67657to combat the effects of endotoxin on normal healthy volunteerwas recently carried out. Development of flu-like symptoms,which were associated with raised serum levels of TNF-α, IL-6and IL-8, were reduced in a dose dependent manner by RWJ-67657 (Fijen et al., 2001).

However, clinical studies have not been limited to healthycontrols; VX-745 (Vertex) has been given to patients with activerheumatoid arthritis (Haddad, 2001; Weisman, 2002), and al-though the drug is well tolerated, it was associated with adverseeffects, such as elevation in liver transaminases. Preclinical safetyevaluations of this drug in animals have revealed that at thatconcentrations VX-745 can cross the blood–brain barrier andexert adverse neurological side effects (Weisman, 2002). For thesereasons, further investigation onVX-745 has been suspended. Therise in the level of liver transaminases has also been observed in aseries of double-blinded, randomised, placebo-controlled studiesof BIRB-796 in healthy volunteers. This compound also sup-pressed neutrophil activation ex vivo, but no inhibition of LPS-induced TNF-α production was observed (Wood et al., 2002).

Currently, the most clinically advanced p38 inhibitors are theScios compound (now Johnson & Johnson) SCIO-323 for treat-ment of stroke and the Vertex compound VX-702, which hasbeen tested in patients with acute coronary syndromes in whompercutaneous coronary intervention (PCI) is planned. Therewere no adverse side effects and the serum level of C-reactiveprotein (CRP), which is considered a risk factor in MI patients,was suppressed (de Winter et al., 2005). However, althoughmany p38 inhibitors have advanced to phase I, II or III clinicaltrial, many of these studies have been stopped prematurely dueto adverse side effects. One reason for this might be the cross-reactivity against other kinases or other cellular signalling mole-cules. An alternative explanation is that p38-dependant sig-nalling is vital to normal cell function necessitating a greaterunderstanding of mechanisms of activation in the hope that thiswill reveal circumstance-specific targets.

8. Summary and conclusions

The p38 kinase pathway has been studied intensely since itsdiscovery in the early 1990s. It has been implicated in various

Please cite this article as: Clark, J. E. et al. Potential of p38-MAPK inhibitors in thpharmthera.2007.06.013

TEDPR

OOF

biological processes, such as cell growth, apoptosis and inflam-mation. In the heart p38 is activated by various pathologicalconditions, such as ischaemia and pressure overload. Studiesusing genetic and pharmacological inhibitors of p38 suggeststhat this kinase plays a key role in left ventricular matrix re-modelling (Petrich & Wang, 2004), cell survival followingischaemia–reperfusion (Ma et al., 1999; Shao et al., 2006) andpossibly, in post MI remodelling (Ren et al., 2005). Investigatorshave shown that multiple isoforms of p38 are activated inresponse to specific stimuli and take part in distinct signallingpathways, which result in activation of specific downstreamsubstrates. From the existing evidence, it appears that p38α andp38β are differentially regulated during myocardial stresses andthat the consequences of activation of each isoformmay differ bycell type. This highlights the likelihood that different memberswithin a single kinase family can play distinct roles in the heartduring ischaemia. Despite continued interest in the p38 pathwayfew studies to date have addressed the role of p38 isoforms otherthan p38α during ischaemia. Furthermore, since there are noisoform-specific pharmacological inhibitors of p38 activity, thecontribution of each isoform remains unclear.

Understanding the physiological roles of each p38 isoformand identifying their mechanism(s) of activation and potentialsubstrates are important avenues that may lead to pharmaco-logical inhibitors with greater circumstance selectivity therebyavoiding the potential pitfalls of chronic systemic inhibition.

References

Adams, R. H., Porras, A., Alonso, G., Jones, M., Vintersten, K., Panelli, S., et al.(2000). Essential role of p38[alpha] MAP kinase in placental but notembryonic cardiovascular development. Mol Cell 6(1), 109−116.

Adams, J. L., Badger, A. M., Kumar, S., & Lee, J. C. (2001). p38 MAP kinase:molecular target for the inhibition of pro-inflammatory cytokines. Prog MedChem 38, 1−60.

Aleshin, A., Sawa, Y., Ono, M., Funatsu, T., Miyagawa, S., &Matsuda, H. (2004).Myocardial protective effect of FR167653; a novel cytokine inhibitor inischemic-reperfused rat heart. Eur J Cardio-Thorac Surg 26(5), 974−980.

Allen, M., Svensson, L., Roach, M., Hambor, J., McNeish, J., & Gabel, C. A.(2000). Deficiency of the stress kinase p38{alpha} results in embryoniclethality: characterization of the kinase dependence of stress responses ofenzyme-deficient embryonic stem cells. J Exp Med 191(5), 859−870.

Askari, N., Diskin, R., Avitzour, M., Capone, R., Livnah, O., & Engelberg, D.(2007). Hyperactive variants of p38alpha induce, whereas hyperactivevariants of p38gamma suppress, activating protein 1-mediated transcription.J Biol Chem 282(1), 91−99.

Badger, A.M., Bradbeer, J. N., Votta, B., Lee, J. C., Adams, J. L., &Griswold, D. E.(1996). Pharmacological profile of SB 203580, a selective inhibitor of cyto-kine suppressive binding protein/p38 kinase, in animal models of arthritis,bone resorption, endotoxin shock and immune function. J Pharmacol ExpTher 279(3), 1453−1461.

Badger, A. M., Griswold, D. E., Kapadia, R., Blake, S., Swift, B. A., Hoffman,S. J., et al. (2000). Disease-modifying activity of SB 242235, a selectiveinhibitor of p38 mitogen-activated protein kinase, in rat adjuvant-inducedarthritis. Arthritis Rheum 43(1), 175−183.

Barancik, M., Htun, P., Strohm, C., Kilian, S., & Schaper, W. (2000). Inhibitionof the cardiac p38-MAPK pathway by SB203580 delays ischemic cell death.J Cardiovasc Pharmacol 35(3), 474−483.

Beardmore, V. A., Hinton, H. J., Eftychi, C., Apostolaki, M., Armaka, M.,Darragh, J., et al. (2005). Generation and characterization of p38beta(MAPK11) gene-targeted mice. Mol Cell Biol 25(23), 10454−10464.

Behr, T. M., Nerurkar, S. S., Nelson, A. H., Coatney, R. W., Woods, T. N.,Sulpizio, A., et al. (2001). Hypertensive end-organ damage and premature

e treatment of ischaemic heart disease. Pharmacol Ther (2007), doi:10.1016/j.

C

616

617

618

619

620

621

622

623

624

625

626

627

628

629

630

631

632

633

634

635

636

637

638

639

640

641

642

643

644

645

646

647

648

649

650

651

652

653

654

655

656

657

658

659

660

661

662

663

664

665

666

667Q3668

669

670

671

672

673

674

675

676

677

678

679

680

681

682

683

84

85

86

87

88

89

90

91

92

93

94

95

96

97

98

99

00

01

02

03

04

05

06

07

08

09

10

11

12

13

14

15

16

17

18

19

20

21

22

23

24

25

26

27

28

29

30

31

32

33

34

35

36

37

38

39

40

41

42

43

44

45

46

47

48

49

50

51

12 J.E. Clark et al. / Pharmacology & Therapeutics xx (2007) xxx–xxx

ARTICLE IN PRESS

UNCO

RRE

mortality are p38 mitogen-activated protein kinase-dependent in a rat modelof cardiac hypertrophy and dysfunction. Circulation 104(11), 1292−1298.

Bell, M., Capone, R., Pashtan, I., Levitzki, A., & Engelberg, D. (2001). Isolationof hyperactive mutants of the MAPK p38/Hog1 that are independent ofMAPK kinase activation. J Biol Chem 276(27), 25351−25358.

Bellahcene, M., Jacquet, S., Cao, X. B., Tanno, M., Haworth, R. S., Layland, J.,et al. (2006). Activation of p38 mitogen-activated protein kinase contributesto the early cardiodepressant action of tumor necrosis factor. J Am CollCardiol 48, 545−555.

Belmont, L. D., &Mitchison, T. J. (1996). Identification of a protein that interactswith tubulin dimers and increases the catastrophe rate of microtubules. Cell84(4), 623−631.

Boehm, J. C., Bower, M. J., Gallagher, T. F., Kassis, S., Johnson, S. R., &Adams, J. L. (2001). Phenoxypyrimidine inhibitors of p38alpha kinase:synthesis and statistical evaluation of the p38 inhibitory potencies of a seriesof 1-(piperidin-4-yl)-4-(4-fluorophenyl)-5-(2-phenoxypyrimidin-4-yl) imi-dazoles. Bioorg Med Chem Lett 11(9), 1123−1126.

Bogoyevitch, M. A., Gillespie-Brown, J., Ketterman, A. J., Fuller, S. J., Ben-Levy, R., Ashworth, A., et al. (1996). Stimulation of the stress-activatedmitogen-activated protein kinase subfamilies in perfused heart. p38/RKmitogen-activated protein kinases and c-Jun N-terminal kinases are activatedby ischemia/reperfusion. Circ Res 79(2), 162−173.

Brancho, D., Tanaka, N., Jaeschke, A., Ventura, J. J., Kelkar, N., Tanaka, Y., et al.(2003). Mechanism of p38 MAP kinase activation in vivo.Genes Dev 17(16),1969−1978.

Braz, J. C., Bueno, O. F., Liang, Q., Wilkins, B. J., Dai, Y. S., Parsons, S., et al.(2003). Targeted inhibition of p38 MAPK promotes hypertrophic cardio-myopathy through upregulation of calcineurin-NFATsignaling. J Clin Invest111(10), 1475−1486.

Cain, B. S., Meldrum, D. R., Meng, X., Dinarello, C. A., Shames, B. D.,Banerjee, A., et al. (1999). p38 MAPK inhibition decreases TNF-[alpha]production and enhances postischemic human myocardial function. J SurgRes 83(1), 7−12.

Carter, A. B., Monick, M. M., & Hunninghake, G. W. (1999). Both Erk and p38kinases are necessary for cytokine gene transcription. Am J Respir Cell MolBiol 20(4), 751−758.

Chang, C. I., Xu, B. E., Akella, R., Cobb, M. H., & Goldsmith, E. J. (2002).Crystal structures of MAP kinase p38 complexed to the docking sites on itsnuclear substrate MEF2A and activator MKK3b.Mol Cell 9(6), 1241−1249.

Cheung, P. C., Campbell, D. G., Nebreda, A. R., & Cohen, P. (2003). Feedbackcontrol of the protein kinase TAK1 by SAPK2a/p38alpha. EMBO J 22(21),5793−5805.

Cirillo, P. F., Pargellis, C., & Regan, J. (2002). The non-diaryl heterocycle classesof p38 MAP kinase inhibitors. Curr Top Med Chem 2(9), 1021−1035.

Clanachan, A. S., Jaswal, J. S., Gandhi, M., Bottorff, D. A., Coughlin, J.,Finegan, B. A., et al. (2003). Effects of inhibition ofmyocardial extracellular-responsive kinase and P38 mitogen-activated protein kinase on mechanicalfunction of rat hearts after prolonged hypothermic ischemia. Transplantation75(2), 173−180.

Clark, J. E., Flavell, R. A., Faircloth, M. E., Davis, R. J., Heads, R. J., &Marber,M. S. (2007). Post-infarction remodeling is independent of mitogen-activated protein kinase kinase 3 (MKK3). Cardiovasc Res.

Communal, C., Colucci, W. S., & Singh, K. (2000). p38 mitogen-activatedprotein kinase pathway protects adult rat ventricular myocytes against beta-adrenergic receptor-stimulated apoptosis. Evidence for Gi-dependentactivation. J Biol Chem 275(25), 19395−19400.

Cook, S. A., Sugden, P. H., & Clerk, A. (1999). Activation of c-Jun N-terminalkinases and p38-mitogen-activated protein kinases in human heart failuresecondary to ischaemic heart disease. J Mol Cell Cardiol 31(8), 1429−1434.

Court, N. W., dos Remedios, C. G., Cordell, J., & Bogoyevitch, M. A. (2002).Cardiac expression and subcellular localization of the p38 mitogen-activatedprotein kinase member, stress-activated protein kinase-3 (SAPK3). J MolCell Cardiol 34(4), 413−426.

Craig, R., Larkin, A., Mingo, A. M., Thuerauf, D. J., Andrews, C., McDonough,P. M., et al. (2000). p38 MAPK and NF-kappa B collaborate to induceinterleukin-6 gene expression and release. Evidence for a cytoprotectiveautocrine signaling pathway in a cardiac myocyte model system. J BiolChem 275(31), 23814−23824.

Please cite this article as: Clark, J. E. et al. Potential of p38-MAPK inhibitors in thpharmthera.2007.06.013

TEDPR

OOF

6de Winter, R. J., Tijssen, J. G. P., Windhausen, F., Krasznai, K., Michels, H. R.,6Ziekenhuis, C., et al. (2005). A major determinant of C-reactive protein6production in patients with acute coronary syndromes undergoing PCI: the6p38 mitogen activated protein kinase signalling pathway. American Heart6Association Scientific Sessions.6Deacon, K., & Blank, J. L. (1997). Characterization of the mitogen-activated6protein kinase kinase 4(MKK4)/c-Jun NH2-terminal kinase 1 and MKK3/6p38 pathways regulated by MEK kinases 2á and 3. MEK kinase 3 activates6MKK3 but does not cause activation of p38 kinase in vivo. J Biol Chem6272(22), 14489−14496.6Derijard, B., Raingeaud, J., Barrett, T., Wu, I. H., Han, J., Ulevitch, R. J., et al.6(1995). Independent human MAP-kinase signal transduction pathways6defined by MEK and MKK isoforms. Science 267(5198), 682−685.6Diskin, R., Askari, N., Capone, R., Engelberg, D., & Livnah, O. (2004). Active6mutants of the human p38alpha mitogen-activated protein kinase. J Biol6Chem 279(45), 47040−47049.7Diskin, R., Lebendiker, M., Engelberg, D., & Livnah, O. (2007). Structures of7p38alpha active mutants reveal conformational changes in L16 loop that7induce autophosphorylation and activation. J Mol Biol 365(1), 66−76.7Dong, C., Davis, R. J., & Flavell, R. A. (2002). MAP kinases in the immune7response. Annu Rev Immunol 20, 55−72.7Engel, F. B., Hsieh, P. C., Lee, R. T., & Keating, M. T. (2006). FGF1/p38 MAP7kinase inhibitor therapy induces cardiomyocyte mitosis, reduces scarring,7and rescues function after myocardial infarction. Proc Natl Acad Sci U S A7103(42), 15546−15551.7English, J., Pearson, G., Wilsbacher, J., Swantek, J., Karandikar, M., Xu, S., et7al. (1999). New insights into the control of MAP kinase pathways. Exp Cell7Res 253(1), 255−270.7Eyers, P. A., van, d. I., Quinlan, R. A., Goedert, M., & Cohen, P. (1999). Use of a7drug-resistant mutant of stress-activated protein kinase 2a/p38 to validate the7in vivo specificity of SB 203580. FEBS Lett 451(2), 191−196.7Fabian,M.A., Biggs,W. H., III, Treiber, D. K., Atteridge, C. E., Azimioara,M.D.,7Benedetti, M. G., et al. (2005). A small molecule-kinase interaction map for7clinical kinase inhibitors. Nat Biotechnol 23(3), 329−336.7Fiedler, B., Feil, R., Hofmann, F., Willenbockel, C., Drexler, H., Smolenski, A.,7et al. (2006). cGMP-dependent protein kinase type I inhibits TAB1-p387mitogen-activated protein kinase apoptosis signaling in cardiac myocytes.7J Biol Chem 281(43), 32831−32840.7Fijen, J. W., Zijlstra, J. G., De, B. P., Spanjersberg, R., Tervaert, J. W., van der7Werf, T. S., et al. (2001). Suppression of the clinical and cytokine response to7endotoxin by RWJ-67657, a p38 mitogen-activated protein-kinase inhibitor,7in healthy human volunteers. Clin Exp Immunol 124(1), 16−20.7Force, T., Kuida, K., Namchuk, M., Parang, K., & Kyriakis, J. M. (2004).7Inhibitors of protein kinase signaling pathways: emerging therapies for7cardiovascular disease. Circulation 109(10), 1196−1205.7Freshney, N.W., Rawlinson, L., Guesdon, F., Jones, E., Cowley, S., Hsuan, J., et al.7(1994). Interleukin-1 activates a novel protein kinase cascade that results in the7phosphorylation of Hsp27. Cell 78(6), 1039−1049.7Gallagher, T. F., Seibel, G. L., Kassis, S., Laydon, J. T., Blumenthal,M. J., Lee, J. C.,7et al. (1997). Regulation of stress-induced cytokine production by pyridiny-7limidazoles; inhibition of CSBP kinase. Bioorg Med Chem 5(1), 49−64.7Gallo, K. A., & Johnson, G. L. (2002). Mixed-lineage kinase control of JNK and7p38 MAPK pathways. Nat Rev Mol Cell Biol 3(9), 663−672.7Ge, B., Gram, H., Di, P. F., Huang, B., New, L., Ulevitch, R. J., et al. (2002).7MAPKK-independent activation of p38alpha mediated by TAB1-dependent7autophosphorylation of p38alpha. Science 295(5558), 1291−1294.7Gorog, D. A., Tanno, M., Cao, X., Bellahcene, M., Bassi, R., Kabir, A. M., et al.7(2004). Inhibition of p38 MAPK activity fails to attenuate contractile7dysfunction in a mouse model of low-flow ischemia. Cardiovasc Res 61(1),7123−131.7Gum, R. J., McLaughlin, M. M., Kumar, S., Wang, Z., Bower, M. J., Lee, J. C.,7et al. (1998). Acquisition of sensitivity of stress-activated protein kinases to7the p38 inhibitor, SB 203580, by alteration of one or more amino acids7within the ATP binding pocket. J Biol Chem 273(25), 15605−15610.7Gupta, A., Aberle, N. S., Kapoor, R., Ren, J., & Sharma, A. C. (2005).7Bigendothelin-1 via p38-MAPK-dependent mechanism regulates adult rat7ventricular myocyte contractility in sepsis. Biochim Biophys Acta 1741(1-2),7127−139.

e treatment of ischaemic heart disease. Pharmacol Ther (2007), doi:10.1016/j.

James Clark
Inserted Text
74(3), 466-70.

752

753

754

755

756

757

758

759

760

761

762

763

764

765

766

767

768

769

770

771

772

773

774

775

776

777

778

779

780

781

782

783

784

785

786

787

788

789

790

791

792

793

794

795

796

797

798

799

800

801

802

803

804

805

806

807

808

809

810

811

812

813

814

815

816

817

818

819Q4

820

821

822

823

824

825

826

827

828

829

830

831

832

833

834

835

836

837

838

839

840

841

842

843

844

845

846

847

848

849

850

851

852

853

854

855

856

857

858

859

860

861

862

863

864

865

866

867

868

869

870

871

872

873

874

875

876

877

878

879

880

881

882

883

884

885

886

887

13J.E. Clark et al. / Pharmacology & Therapeutics xx (2007) xxx–xxx

ARTICLE IN PRESS

UNCO

RREC

Gysembergh, A., Simkhovich, B. Z., Kloner, R. A., & Przyklenk, K. (2001). p38MAPK activity is not increased early during sustained coronary arteryocclusion in preconditioned versus control rabbit heart. J Mol Cell Cardiol33(4), 681−690.

Haar, E. T., Prabakhar, P., Liu, X., & Lepre, C. (2007). Crystal structure of the P38{alpha}-MAPKAP kinase 2 heterodimer. J Biol Chem 282(13), 9733−9739.

Haddad, J. J. (2001). VX-745. Vertex Pharmaceuticals. Curr Opin InvestigDrugs 2(8), 1070−1076.

Han, J., Lee, J. D., Bibbs, L., & Ulevitch, R. J. (1994). A MAP kinase targetedby endotoxin and hyperosmolarity in mammalian cells. Science 265(5173),808−811.

Han, J., Richter, B., Li, Z., Kravchenko, V., & Ulevitch, R. J. (1995). Molecularcloning of human p38 MAP kinase. Biochim Biophys Acta 1265(2-3),224−227.

Han, J., Lee, J. D., Jiang, Y., Li, Z., Feng, L., & Ulevitch, R. J. (1996).Characterization of the structure and function of a novel MAP kinase kinase(MKK6). J Biol Chem 271(6), 2886−2891.

Hasegawa, M., & Cahill, G. M. (2004). Regulation of the circadian oscillator inXenopus retinal photoreceptors by protein kinases sensitive to the stress-activated protein kinase inhibitor, SB 203580. J Biol Chem 279(21),22738−22746.

Hoover, H. E., Thuerauf, D. J., Martindale, J. J., & Glembotski, C. C. (2000).alpha B-crystallin gene induction and phosphorylation by MKK6-activatedp38. A potential role for alpha B-crystallin as a target of the p38 branch ofthe cardiac stress response. J Biol Chem 275(31), 23825−23833.

House, S. L., Branch, K., Newman, G., Doetschman, T., & Schultz, J. J. (2005).Cardioprotection induced by cardiac-specific overexpression of fibroblastgrowth factor-2 is mediated by the MAPK cascade. Am J Physiol Heart CircPhysiol 289(5), H2167−H2175.

Hutchison, M., Berman, K. S., & Cobb, M. H. (1998). Isolation of TAO1, aprotein kinase that activates MEKs in stress-activated protein kinasecascades. J Biol Chem 273(44), 28625−28632.

Ichijo, H., Nishida, E., Irie, K., ten, D. P., Saitoh, M., Moriguchi, T., et al. (1997).Induction of apoptosis by ASK1, a mammalian MAPKKK that activatesSAPK/JNK and p38 signaling pathways. Science 275(5296), 90−94.

Kabir, A. M., Cao, X., Gorog, D. A., Tanno, M., Bassi, R., Bellahcene, M., et al.(2005). Antimycin A induced cardioprotection is dependent on pre-ischemicp38-MAPK activation but independent of MKK3. J Mol Cell Cardiol 39(4),709−717.

Kaiser, R.A., Bueno, O. F., Lips, D. J., Doevendans, P. A., Jones, F., Kimball, T. F.,et al. (2004). Targeted inhibition of p38 mitogen-activated protein kinaseantagonizes cardiac injury and cell death following ischemia-reperfusion invivo. J Biol Chem 279(15), 15524−15530.

Kang, Y. J., Seit-Nebi, A., Davis, R. J., & Han, J. (2006). Multiple activationmechanisms of p38alpha mitogen-activated protein kinase. J Biol Chem 281(36), 26225−26234.

Khan, M., Varadharaj, S., Ganesan, L. P., Shobha, J. C., Naidu, M. U., Parinandi,N. L., et al. (2006). C-phycocyanin protects against ischemia–reperfusioninjury of heart through involvement of p38 MAPK and ERK signaling. Am JPhysiol Heart Circ Physiol 290(5), H2136−H2145.

Kim, H. P., Wang, X., Zhang, J., Suh, G. Y., Benjamin, I. J., Ryter, S. W., et al.(2005). Heat shock protein-70 mediates the cytoprotective effect of carbonmonoxide: involvement of p38{beta} MAPK and heat shock factor-1.J Immunol 175(4), 2622−2629.

Kim, J. K., Pedram, A., Razandi, M., & Levin, E. R. (2006). Estrogen preventscardiomyocyte apoptosis through inhibition of reactive oxygen species anddifferential regulation of p38 kinase isoforms. J Biol Chem 281(10),6760−6767.

Koike, N., Takeyoshi, I., Ohki, S., Tokumine, M., Matsumoto, K., & Morishita,Y. (2004). Effects of adding P38 mitogen-activated protein-kinase inhibitorto celsior solution in canine heart transplantation from non-heart-beatingdonors. Transplantation 77(2), 286−292.

Kotlyarov, A., Neininger, A., Schubert, C., Eckert, R., Birchmeier, C., Volk, H. D.,et al. (1999). MAPKAP kinase 2 is essential for LPS-induced TNF-alphabiosynthesis. Nat Cell Biol 1(2), 94−97.

Kulkarni, R. G., Srivani, P., Achaiah, G., & Sastry, G. N. (2007). Strategies todesign pyrazolyl urea derivatives for p38 kinase inhibition: a molecularmodeling study. J Comput Aided Mol Des.

Please cite this article as: Clark, J. E. et al. Potential of p38-MAPK inhibitors in thpharmthera.2007.06.013

TEDPR

OOF

Kuma, Y., Sabio, G., Bain, J., Shpiro, N., Marquez, R., & Cuenda, A. (2005).BIRB796 inhibits all p38 MAPK isoforms in vitro and in vivo. J Biol Chem280(20), 19472−19479.

Lantos, I., Bender, P. E., Razgaitis, K. A., Sutton, B. M., DiMartino, M. J.,Griswold, D. E., et al. (1984). Antiinflammatory activity of 5,6-diaryl-2,3-dihydroimidazo[2,1-b]thiazoles. Isomeric 4-pyridyl and 4-substituted phe-nyl derivatives. J Med Chem 27(1), 72−75.

Lee, J. C., Griswold, D. E., Votta, B., & Hanna, N. (1988). Inhibition ofmonocyte IL-1 production by the anti-inflammatory compound, SK&F86002. Int J Immunopharmacol 10(7), 835−843.

Lee, J. C., Badger, A. M., Griswold, D. E., Dunnington, D., Truneh, A., Votta,B., et al. (1993). Bicyclic imidazoles as a novel class of cytokinebiosynthesis inhibitors. Ann N Y Acad Sci 696, 149−170.

Lee, J. C., Laydon, J. T., McDonnell, P. C., Gallagher, T. F., Kumar, S., Green,D., et al. (1994). A protein kinase involved in the regulation of inflammatorycytokine biosynthesis. Nature 372(6508), 739−746.

Lee, J. C., Kumar, S., Griswold, D. E., Underwood, D. C., Votta, B. J., &Adams, J. L. (2000). Inhibition of p38 MAP kinase as a therapeutic strategy.Immunopharmacology 47(2-3), 185−201.

Lemke, L. E., Bloem, L. J., Fouts, R., Esterman, M., Sandusky, G., & Vlahos, C. J.(2001). Decreased p38 MAPK activity in end-stage failing human myocar-dium: p38MAPK alpha is the predominant isoform expressed in human heart.J Mol Cell Cardiol 33(8), 1527−1540.

Li, M., Georgakopoulos, D., Lu, G., Hester, L., Kass, D. A., Hasday, J., et al.(2005). p38 MAP kinase mediates inflammatory cytokine induction incardiomyocytes and extracellular matrix remodeling in heart. Circulation111(19), 2494−2502.

Li, Z., Ma, J. Y., Kerr, I., Chakravarty, S., Dugar, S., Schreiner, G., et al. (2006).Selective inhibition of p38alpha MAPK improves cardiac function andreduces myocardial apoptosis in rat model of myocardial injury. Am JPhysiol Heart Circ Physiol 291(4), H1972−H1977.

Lim, M., Martinez, T., Jablons, D., Cameron, R., Guo, H., Toole, B., et al.(1998). Tumor-derived EMMPRIN (extracellular matrix metalloproteinaseinducer) stimulates collagenase transcription through MAPK p38. FEBSLett 441(1), 88−92.

Liu, Y. H., Wang, D., Rhaleb, N. E., Yang, X. P., Xu, J., Sankey, S. S., et al.(2005). Inhibition of p38 mitogen-activated protein kinase protects the heartagainst cardiac remodeling in mice with heart failure resulting frommyocardial infarction. J Card Fail 11(1), 74−81.

Lu, G., Kang, Y. J., Han, J., Herschman, H. R., Stefani, E., & Wang, Y. (2005).TAB-1 modulates intracellular localization of p38 MAP kinase anddownstream signaling. J Biol Chem, M507610200.

Luss, H., Neumann, J., Schmitz, W., Schulz, R., & Heusch, G. (2000). Thestress-responsive MAP kinase p38 is activated by low-flow ischemia in thein situ porcine heart. J Mol Cell Cardiol 32(10), 1787−1794.

Ma, X. L., Kumar, S., Gao, F., Louden, C. S., Lopez, B. L., Christopher, T. A., et al.(1999). Inhibition of p38 mitogen-activated protein kinase decreasescardiomyocyte apoptosis and improves cardiac function after myocardialischemia and reperfusion. Circulation 99(13), 1685−1691.

Mackay, K., &Mochly-Rosen, D. (1999). An inhibitor of p38 mitogen-activatedprotein kinase protects neonatal cardiac myocytes from ischemia. J BiolChem 274(10), 6272−6279.

Mackay, K., & Mochly-Rosen, D. (2000). Involvement of a p38 mitogen-activated protein kinase phosphatase in protecting neonatal rat cardiacmyocytes from ischemia. J Mol Cell Cardiol 32(8), 1585−1588.

Marais, E., Genade, S., Huisamen, B., Strijdom, J. G., Moolman, J. A., &Lochner, A. (2001). Activation of p38 MAPK induced by a multi-cycleischaemic preconditioning protocol is associated with attenuated p38 MAPKactivity during sustained ischaemia and reperfusion. J Mol Cell Cardiol 33(4),769−778.

Marais, E., Genade, S., Salie, R., Huisamen, B., Maritz, S., Moolman, J. A., et al.(2005). The temporal relationship between p38 MAPK and HSP27 activationin ischaemic and pharmacological preconditioning. Basic Res Cardiol 100(1),35−47.

Martin, J. L., Avkiran, M., Quinlan, R. A., Cohen, P., & Marber, M. S. (2001).Antiischemic effects of SB203580 are mediated through the inhibition ofp38{alpha} mitogen-activated protein kinase: evidence from ectopicexpression of an inhibition-resistant kinase. Circ Res 89(9), 750−752.

e treatment of ischaemic heart disease. Pharmacol Ther (2007), doi:10.1016/j.

James Clark
Inserted Text
21(4), 155-66.

C

888

889

890

891

892

893

894

895

896

897

898

899

900

901

902

903

904

905

906

907

908

909

910

911

912

913

914

915

916

917

918

919

920

921

922

923

924

925

926

927

928

929

930

931

932

933

934

935

936

937

938

939

940

941

942

943

944

945

946

947

948

949

950

951

952

953

954

955

56

57

58

59

60

61

62

63

64

65

66

67

68

69

70

71

72

73

74

75

76

77

78

79

80

81

82

83

84

85

86

87

88

89

90

91

92

93

94

95

96

97

98

99

000

001

002

003

004

005

006

007

008

009

010

011

012

013

014

015

016

017

018

019

020

021

022

023

14 J.E. Clark et al. / Pharmacology & Therapeutics xx (2007) xxx–xxx

ARTICLE IN PRESS

UNCO

RRE

Martindale, J. J., Wall, J. A., Martinez-Longoria, D. M., Aryal, P., Rockman,H. A., Guo, Y., et al. (2005). Overexpression of mitogen-activated proteinkinase kinase 6 in the heart improves functional recovery from ischemia invitro and protects against myocardial infarction in vivo. J Biol Chem 280(1),669−676.

Matsuyama, W., Faure, M., & Yoshimura, T. (2003). Activation of discoidindomain receptor 1 facilitates the maturation of human monocyte-deriveddendritic cells through the TNF receptor associated factor 6/TGF-beta-activated protein kinase 1 binding protein 1 beta/p38 alpha mitogen-activated protein kinase signaling cascade. J Immunol 171(7), 3520−3532.

Meldrum, D. R., Dinarello, C. A., Cleveland, J. C., Jr., Cain, B. S., Shames, B. D.,Meng, X., et al. (1998). Hydrogen peroxide induces tumor necrosis factoralpha-mediated cardiac injury by a P38 mitogen-activated protein kinase-dependent mechanism. Surgery 124(2), 291−296.

Mittelstadt, P. R., Salvador, J. M., Fornace, A. J., Jr., & Ashwell, J. D. (2005).Activating p38MAPK: new tricks for an old kinase.Cell Cycle 4(9), 1189−1192.

Moriguchi, T., Kuroyanagi, N., Yamaguchi, K., Gotoh, Y., Irie, K., Kano, T., et al.(1996). A novel kinase cascade mediated by mitogen-activated protein kinasekinase 6 and MKK3. J Biol Chem 271(23), 13675−13679.

Nagarkatti, D. S., & Afi, R. I. (1998). Role of p38 MAP kinase in myocardialstress. J Mol Cell Cardiol 30(8), 1651−1664.

Office for National Statistics (2000). Key Health Statistics from GeneralPractice. London: The Stationery Office.

Ohkusu-Tsukada, K., Tominaga, N., Udono, H., & Yui, K. (2005). Regulation ofthe maintenance of peripheral T-cell anergy by TAB1-1 mediated p38alphaactivation. Mol Cell Biol 25(19), 8763.

Okada, T., Otani, H., Wu, Y., Kyoi, S., Enoki, C., Fujiwara, H., et al. (2005).Role of F-actin organization in p38 MAP kinase-mediated apoptosis andnecrosis in neonatal rat cardiomyocytes subjected to simulated ischemia andreoxygenation. Am J Physiol Heart Circ Physiol 289(6), H2310−H2318.

Ono, K., & Han, J. (2000). The p38 signal transduction pathway: activation andfunction. Cell Signal 12(1), 1−13.

Otsu, K., Yamashita, N., Nishida, K., Hirotani, S., Yamaguchi, O., Watanabe, T.,et al. (2003). Disruption of a single copy of the p38alpha MAP kinase geneleads to cardioprotection against ischemia–reperfusion. Biochem BiophysRes Commun 302(1), 56−60.

Pargellis, C., Tong, L., Churchill, L., Cirillo, P. F., Gilmore, T., Graham,A. G., et al.(2002). Inhibition of p38 MAP kinase by utilizing a novel allosteric bindingsite. Nat Struct Biol 9(4), 268−272.

Parker, C. G., Hunt, J., Diener, K.,McGinley,M., Soriano, B., Keesler, G. A., et al.(1998). Identification of stathmin as a novel substrate for p38 delta. BiochemBiophys Res Commun 249(3), 791−796.

Pearson, G., Robinson, F., Beers, G. T., Xu, B. E., Karandikar, M., Berman, K.,et al. (2001). Mitogen-activated protein (MAP) kinase pathways: regulationand physiological functions. Endocr Rev 22(2), 153−183.

Petrich, B. G., & Wang, Y. (2004). Stress-activated MAP kinases in cardiacremodeling and heart failure: new insights from transgenic studies. TrendsCardiovasc Med 14(2), 50−55.

Pfeffer, M. A., & Braunwald, E. (1990). Ventricular remodeling after myocardialinfarction. Experimental observations and clinical implications. Circulation81(4), 1161−1172.

Ping, P., & Murphy, E. (2000). Role of p38 mitogen-activated protein kinases inpreconditioning: a detrimental factor or a protective kinase? Circ Res 86(9),921−922.

Raingeaud, J., Gupta, S., Rogers, J. S., Dickens, M., Han, J., Ulevitch, R. J., et al.(1995). Pro-inflammatory cytokines and environmental stress cause p38mitogen-activated protein kinase activation by dual phosphorylation ontyrosine and threonine. J Biol Chem 270(13), 7420−7426.

Rakhit, R. D., Kabir, A. N. M., Mockridge, J. W., Saurin, A., & Marber, M. S.(2001). Role of G proteins and modulation of p38 MAPK activation in theprotection by nitric oxide against ischemia-reoxygenation injury. BiochemBiophys Res Commun 286(5), 995−1002.

Ren, J., Zhang, S., Kovacs, A., Wang, Y., & Muslin, A. J. (2005). Role ofp38alpha MAPK in cardiac apoptosis and remodeling after myocardialinfarction. J Mol Cell Cardiol 38(4), 617−623.

Riad, A., Unger, D., Du, J., Westermann, D., Mohr, Z., Sobirey, M., et al. (2007).Chronic inhibition of p38MAPK improves cardiac and endothelial functionin experimental diabetes mellitus. Eur J Pharmacol 554(1), 40−45.

Please cite this article as: Clark, J. E. et al. Potential of p38-MAPK inhibitors in thpharmthera.2007.06.013

TEDPR

OOF

9Rincon, M., & Pedraza-Alva, G. (2003). JNK and p38 MAP kinases in CD4+9and CD8+T cells. Immunol Rev 192, 131−142.9Rouse, J., Cohen, P., Trigon, S., Morange, M., Onso-Llamazares, A., Zamanillo,9D., et al. (1994). A novel kinase cascade triggered by stress and heat shock9that stimulates MAPKAP kinase-2 and phosphorylation of the small heat9shock proteins. Cell 78(6), 1027−1037.9Sabio, G., Reuver, S., Feijoo, C., Hasegawa,M., Thomas, G.M., Centeno, F., et al.9(2004). Stress- and mitogen-induced phosphorylation of the synapse-as-9sociated protein SAP90/PSD-95 by activation of SAPK3/p38gamma and9ERK1/ERK2. Biochem J 380(Pt 1), 19−30.9Sabio, G., Arthur, J. S., Kuma, Y., Peggie, M., Carr, J., Murray-Tait, V., et al.9(2005). p38gamma regulates the localisation of SAP97 in the cytoskeleton9by modulating its interaction with GKAP. EMBO J 24(6), 1134−1145.9Saccani, S., Pantano, S., & Natoli, G. (2002). p38-Dependent marking of9inflammatory genes for increased NF-kappa B recruitment.Nat Immunol 3(1),969−75.9Salvador, J. M., Mittelstadt, P. R., Belova, G. I., Fornace, A. J., Jr., & Ashwell,9J. D. (2005). The autoimmune suppressor Gadd45alpha inhibits the T cell9alternative p38 activation pathway. Nat Immunol 6(4), 396−402.9Salvador, J. M., Mittelstadt, P. R., Guszczynski, T., Copeland, T. D., Yamaguchi,9H., Appella, E., et al. (2005). Alternative p38 activation pathway mediated9by T cell receptor-proximal tyrosine kinases. Nat Immunol 6(4), 390−395.9Sanada, S., Kitakaze, M., Papst, P. J., Hatanaka, K., Asanuma, H., Aki, T., et al.9(2001). Role of Phasic Dynamism of p38 Mitogen-Activated Protein Kinase9Activation in Ischemic Preconditioning of the Canine Heart. Circ Res 88(2),9175−180.9Saurin, A. T., Martin, J. L., Heads, R. J., Foley, C. L. A. I., Mockridge, J. W.,9Wright, M. J., et al. (2000). The role of differential activation of p38-9mitogen-activated protein kinase in preconditioned ventricular myocytes.9FASEB J 14(14), 2237−2246.9Schneider, S., Chen, W., Hou, J., Steenbergen, C., & Murphy, E. (2001).9Inhibition of p38 MAPK {alpha}/{beta} reduces ischemic injury and does9not block protective effects of preconditioning. Am J Physiol Heart Circ9Physiol 280(2), H499−H508.9See, F., Thomas, W., Way, K., Tzanidis, A., Kompa, A., Lewis, D., et al. (2004).9p38 mitogen-activated protein kinase inhibition improves cardiac function9and attenuates left ventricular remodeling following myocardial infarction in9the rat. J Am Coll Cardiol 44(8), 1679−1689.9Shao, Z., Bhattacharya, K., Hsich, E., Park, L., Walters, B., Germann, U., et al.9(2006). c-Jun N-Terminal Kinases Mediate Reactivation of Akt and9Cardiomyocyte Survival After Hypoxic Injury In Vitro and In Vivo. Circ9Res 98(1), 111−118.9Sharov, V. G., Todor, A., Suzuki, G., Morita, H., Tanhehco, E. J., & Sabbah, H. N.9(2003). Hypoxia, angiotensin-II, and norepinephrine mediated apoptosis is1stimulus specific in canine failed cardiomyocytes: a role for p38MAPK, Fas-L1and cyclin D1. Eur J Heart Fail 5(2), 121−129.1Sugden, P. H., & Clerk, A. (1998). “Stress-responsive” mitogen-activated1protein kinases (c-Jun N-terminal kinases and p38 mitogen-activated protein1kinases) in the myocardium. Circ Res 83(4), 345−352.1Sumida, T., Otani, H., Kyoi, S., Okada, T., Fujiwara, H., Nakao, Y., et al. (2005).1Temporary blockade of contractility during reperfusion elicits a cardiopro-1tective effect of the p38 MAP kinase inhibitor SB-203580. Am J Physiol1Heart Circ Physiol 288(6), H2726−H2734.1Swynghedauw, B. (1999). Molecular mechanisms of myocardial remodeling.1Physiol Rev 79(1), 215−262.1Tamura, K., Sudo, T., Senftleben, U., Dadak, A. M., Johnson, R., & Karin, M.1(2000). Requirement for p38alpha in erythropoietin expression: a role for1stress kinases in erythropoiesis. Cell 102(2), 221−231.1Tanno, M., Bassi, R., Gorog, D. A., Saurin, A. T., Jiang, J., Heads, R. J., et al.1(2003). Diverse mechanisms of myocardial p38 mitogen-activated protein1kinase activation: evidence for MKK-independent activation by a TAB1-1associated mechanism contributing to injury during myocardial ischemia.1Circ Res 93(3), 254−261.1Tanoue, T., Adachi, M., Moriguchi, T., & Nishida, E. (2000). A conserved1docking motif in MAP kinases common to substrates, activators and1regulators. Nat Cell Biol 2(2), 110−116.1The United Kingdom Heart Attack Study (UKHAS) Collaborative Group. (1998).1The falling mortality from coronary heart disease: a clinic-pathological

e treatment of ischaemic heart disease. Pharmacol Ther (2007), doi:10.1016/j.

D

1024

1025

1026

1027

1028

1029

1030

1031

1032

1033

1034

1035

1036

1037

1038

1039

1040

1041

1042

1043

1044

1045

1046

1047

1048

1049

1050

1051

1052

1053

1054

1055

1056

1057

1058

1059

1060

1061

1062

1063

1064

1065

1066

1067

1068

1069

1070

1071

1072

1073

1074

1075

1076

1077

1078

1079

1080

1081

1082

1083

15J.E. Clark et al. / Pharmacology & Therapeutics xx (2007) xxx–xxx

ARTICLE IN PRESS

perspective. Heart 80, 121−126 BHF Coronary heart disease statistics atwww.heartstats.org

Udelson, J. E., Patten, R. D., & Konstam, M. A. (2003). New concepts in post-infarction ventricular remodeling. Rev Cardiovasc Med 4(Suppl 3), S3−S12.

Vahebi, S., Ota, A., Li, M., Warren, C. M., de Tombe, P. P., Wang, Y., et al.(2007). p38-MAPK induced dephosphorylation of alpha-tropomyosin isassociated with depression of myocardial sarcomeric tension and ATPaseactivity. Circ Res 100(3), 408−415.

Wang, R., Wang, Z., & Wu, L. (1997). Carbon monoxide-induced vasorelaxa-tion and the underlying mechanisms. Br J Pharmacol 121(5), 927−934.

Wang, Y., Huang, S., Sah, V. P., Ross, J., Jr., Brown, J. H., Han, J., et al. (1998).Cardiac muscle cell hypertrophy and apoptosis induced by distinct membersof the p38 mitogen-activated protein kinase family. J Biol Chem 273(4),2161−2168.

Wang, Z., Canagarajah, B. J., Boehm, J. C., Kassisa, S., Cobb,M. H., Young, P. R.,et al. (1998). Structural basis of inhibitor selectivity in MAP kinases.Structure 6(9), 1117−1128.

Wang, M., Tsai, B. M., Turrentine, M. W., Mahomed, Y., Brown, J. W., &Meldrum, D. R. (2005). p38 mitogen activated protein kinase mediates bothdeath signaling and functional depression in the heart. Ann Thorac Surg 80(6),2235−2241.

Weinbrenner, C., Liu, G. S., Cohen, M. V., & Downey, J. M. (1997).Phosphorylation of tyrosine 182 of p38 mitogen-activated protein kinasecorrelates with the protection of preconditioning in the rabbit heart. J MolCell Cardiol 29(9), 2383−2391.

Weisman, M. (2002). Double-blind, placebo-controlled trial of VX-745, an oralp38 mitogen activated protein kinase inhibitor, in patients with rheumatoidarthritis (RA). Ann. European Congress Rheumatol.

Wilson, K. P., Fitzgibbon, M. J., Caron, P. R., Griffith, J. P., Chen, W.,McCaffrey, P. G., et al. (1996). Crystal structure of p38 mitogen-activatedprotein kinase. J Biol Chem 271(44), 27696−27700.

UNCO

RREC

Please cite this article as: Clark, J. E. et al. Potential of p38-MAPK inhibitors in thpharmthera.2007.06.013

PROO

F

Wood, C. C., Yong, C. L., Madwed, J. B., & Gupta, A. (2002). Pharmacoki-netics and Pharmacodynamics of an Oral p38 MAP Kinase Inhibitor (BIRB768 BS), Administered once Daily for 7 Days (pp. A993).

Xie, Y., Chen, C., Stevenson, M. A., Auron, P. E., & Calderwood, S. K. (2002).Heat shock factor 1 represses transcription of the IL-1beta gene throughphysical interaction with the nuclear factor of interleukin 6. J Biol Chem277(14), 11802−11810.

Yada, M., Shimamoto, A., Hampton, C. R., Chong, A. J., Takayama, H.,Rothnie, C. L., et al. (2004). FR167653 diminishes infarct size in a murinemodel of myocardial ischemia–reperfusion injury. J Thorac CardiovascSurg 128(4), 588−594.

Yamaguchi, K., Shirakabe, K., Shibuya, H., Irie, K., Oishi, I., Ueno, N., et al.(1995). Identification of a member of the MAPKKK family as a potentialmediator of TGF-beta signal transduction. Science 270(5244), 2008−2011.

Young, P. R., McLaughlin, M. M., Kumar, S., Kassis, S., Doyle, M. L., McNulty,D., et al. (1997). Pyridinyl imidazole inhibitors of p38 mitogen-activatedprotein kinase bind in the ATP site. J Biol Chem 272(18), 12116−12121.

Yue, T. L., Wang, C., Gu, J. L., Ma, X. L., Kumar, S., Lee, J. C., et al. (2000).Inhibition of extracellular signal-regulated kinase enhances ischemia/reoxygenation-induced apoptosis in cultured cardiac myocytes andexaggerates reperfusion injury in isolated perfused heart. Circ Res 86(6),692−699.

Zhang, S., Weinheimer, C., Courtois, M., Kovacs, A., Zhang, C. E., Cheng, A. M.,et al. (2003). The role of the Grb2-p38 MAPK signaling pathway in cardiachypertrophy and fibrosis. J Clin Invest 111(6), 833−841.

Zheng, S., & Zuo, Z. (2004). Isoflurane preconditioning induces neuroprotec-tion against ischemia via activation of P38 mitogen-activated proteinkinases. Mol Pharmacol 65(5), 1172−1180.

E T

e treatment of ischaemic heart disease. Pharmacol Ther (2007), doi:10.1016/j.