Galley - Inflammation and Immunity

Embed Size (px)

Citation preview

  • 7/30/2019 Galley - Inflammation and Immunity

    1/119

    Critical Care Focus10: Inflammation and

    Immunity

    EDITOR

    DR. HELEN F. GALLEY

    BMJ Books

  • 7/30/2019 Galley - Inflammation and Immunity

    2/119

    Critical Care Focus

    10: Inflammation and Immunity

  • 7/30/2019 Galley - Inflammation and Immunity

    3/119

  • 7/30/2019 Galley - Inflammation and Immunity

    4/119

    Critical Care Focus

    10: Inflammation and Immunity

    EDITOR

    DR HELEN F GALLEY

    Senior Lecturer in Anaesthesia and Intensive Care

    University of Aberdeen

    EDITORIAL BOARD

    PROFESSOR NIGEL R WEBSTER

    Professor of Anaesthesia and Intensive Care

    University of Aberdeen

    DR PAUL G P LAWLER

    Clinical Director of Intensive Care

    University of Aberdeen

    DR NEIL SONI

    Consultant in Anaesthesia and Intensive Care

    Chelsea and Westminster Hospital

    DR MERVYN SINGER

    Reader in Intensive Care

    University College Hospital, London

  • 7/30/2019 Galley - Inflammation and Immunity

    5/119

    BMJ Publishing Group 2003BMJ Books is an imprint of the BMJ Publishing Group

    All rights reserved. No part of this publication may be reproduced, stored in aretrieval system, or transmitted, in any form or by any means, electronic,

    mechanical, photocopying, recording and/or otherwise, without the prior writtenpermission of the publishers.

    First published in 2003by BMJ Books, BMA House,Tavistock Square,

    London WC1H 9JR

    www.bmjbooks.comwww.ics.ac.uk

    British Library Cataloguing in Publication Data

    A catalogue record for this book is available from the British Library

    ISBN 0-7279-1689-0

    Typeset by Newgen Imaging Systems (P) Ltd, Chennai.Printed and bound in Spain by GraphyCems, Navarra

  • 7/30/2019 Galley - Inflammation and Immunity

    6/119

    v

    Contents

    Contributors vii

    Preface viii

    Introduction ix

    1 Immunoparalysis 1

    JEAN-MARC CAVAILLON, HELEN F GALLEY

    2 Apoptosis and the inflammatory process 18NIGEL R WEBSTER

    3 Virus interaction with host immunity 33

    LAWRENCE S YOUNG

    4 The double-edged role of the neutrophil in inflammatory

    responses 47

    PAUL G HELLEWELL

    5 T cell immunity and sepsis 65

    EGBERT PRAVINKUMAR

    6 Metalloproteinases and inflammation 77

    ANDREW J GEARING

    7 Glucocorticoid therapy in septic shock 90

    PIERRE-EDOUARD BOLLAERT

    Index 99

  • 7/30/2019 Galley - Inflammation and Immunity

    7/119

    Critical Care Focus series

    Also available:

    H F Galley (ed) Critical Care Focus 1: Renal Failure, 1999.

    H F Galley (ed) Critical Care Focus 2: Respiratory Failure, 1999.

    H F Galley (ed) Critical Care Focus 3:Neurological Injury, 2000.

    H F Galley (ed) Critical Care Focus 4: Endocrine Disturbance, 2000.

    H F Galley (ed) Critical Care Focus 5:Antibiotic Resistance and Infection

    Control, 2001.

    H F Galley (ed) Critical Care Focus 6: Cardiology in Critical Illness, 2001.

    H F Galley (ed) Critical Care Focus 7:Nutritional Issues, 2001.

    H F Galley (ed) Critical Care Focus 8: Blood and Blood Transfusion, 2002.

    H F Galley (ed) Critical Care Focus 9: The Gut, 2002.

  • 7/30/2019 Galley - Inflammation and Immunity

    8/119

    vii

    Contributors

    Pierre-Edouard BollaertProfesseur des Universits, Service de Ranimation Mdicale, Centre

    Hospitalier Universitaire, Nancy, France

    Jean-Marc Cavaillon

    Unit dImmunology-Allergie, Institute Pasteur, Paris, France

    Helen F Galley

    Senior Lecturer in Anaesthesia and Intensive Care, University of

    Aberdeen, UK

    Andrew J Gearing

    Chief Executive Officer, Biocomm International, Melbourne, Victoria,

    Australia

    Paul G Hellewell

    Professor of Vascular Biology, University of Sheffield, UK

    Egbert PravinkumarLecturer in Intensive Care Medicine, University of Aberdeen, UK

    Nigel R Webster

    Professor of Anaesthesia and Intensive Care and Honorary Consultant,

    University of Aberdeen, UK

    Lawrence S Young

    Director of Institute and Head of Division of Cancer Research,

    UK Institute for Cancer Studies, University of Birmingham, UK

  • 7/30/2019 Galley - Inflammation and Immunity

    9/119

    viii

    Preface to the Critical Care

    Focus series

    The Critical Care Focus series aims to provide a snapshot of currentthoughts and practice, by renowned experts. The complete series should

    provide a comprehensive guide for all health professionals on key issues in

    todays field of critical care. The volumes are deliberately concise and easy

    to read, designed to inform and provoke. Most chapters are produced from

    transcriptions of lectures given at the Intensive Care Society meetings and

    represent the views of world leaders in their fields.

    Helen F Galley

  • 7/30/2019 Galley - Inflammation and Immunity

    10/119

    ix

    Introduction

    Immunoparalysis

    Jean-Marc Cavaillon and Helen F Galley

    Several studies indicate that depression of immune function induced by

    traumatic injury is aetiologically involved in the development of infection

    or sepsis. Nevertheless, the mechanisms behind the maintenance of the

    sustained suppression of immune function remain incompletely

    understood. Alterations of immune responses have been regularly reported

    in patients with systemic inflammatory responses syndrome (SIRS).

    The observation that some patients have apparent immune paralysisled to the concept of compensatory anti-inflammatory response

    syndrome or CARS. In this article, we describe this phenomenon of

    immunoparalysis but, although alterations in immune response are

    probably associated with an enhanced sensitivity to nosocomial infections,

    there is no clear demonstration that they are directly responsible for poor

    outcome in sepsis.

    Apoptosis and the inflammatory process

    Nigel R Webster

    Cells that are damaged by injury undergo swelling and leakage of cell

    contents, leading to inflammation of surrounding tissues. This process

    is called necrosis. Cells that are induced to commit suicide, in contrast,

    shrink, and the mitochondrial membrane becomes breached such that

    release of cytochrome c occurs. Chromatin (DNA and protein) in the

    nucleus becomes degraded into small, membrane-wrapped fragments, and

    the phospholipid phosphatidylserine, which is normally hidden within theplasma membrane, is exposed on the surface. This is then bound by

    receptors on phagocytic cells, such as macrophages, which engulf the cell

    fragments, leading to a quiet orderly removal of dead cells. This pattern

  • 7/30/2019 Galley - Inflammation and Immunity

    11/119

    x

    CRITICAL CARE FOCUS 10: INFLAMMATION AND IMMUNITY

    of events is called programmed cell death or apoptosis. The cellular

    machinery of programmed cell death is as intrinsic to the cell as, for

    example, mitosis. This article will describe the regulation and process of

    apoptosis and its relevance to disease, including the inflammatory response

    in patients with sepsis.

    Virus interaction with host immunity

    Lawrence S Young

    Resistance to and recovery from viral infections depends on the

    interactions between virus and host. The defences mounted by the host

    may act directly on the virus or indirectly on virus replication by altering or

    killing the infected cell. The non-specific host defences function early inthe encounter with virus to prevent or limit infection, while the specific

    host defences function after infection in initiation of immune responses

    to subsequent challenges. Viruses have evolved complex strategies to

    manipulate host immune defences to their advantage, permitting viral

    replication without massive inflammatory responses integrating their

    needs with that of their host man. However, some persistent and latent

    viral infections can lead to serious disease and malignancy.This article will

    describe the hostvirus interactions and particularly focus on the role of

    latent infection with the EpsteinBarr virus in tumour development thekiller within.

    The double-edged role of the neutrophil in

    inflammatory responses

    Paul G Hellewell

    Accumulation of leucocytes in tissues is essential for effective host

    defence. The major role of neutrophils is to phagocytose and destroy

    infectious agents but they can also cause host damage, and neutrophil-

    mediated injury has been implicated in several inflammatory conditions

    seen on the Intensive Care Unit (ICU). This article provides an overview

    of the vital role of neutrophils in host defence, and the consequences of

    host damage. An increased understanding of neutrophil biology is likely to

    result in intelligent intervention strategies.

    T cell immunity and sepsis

    Egbert Pravinkumar

    Immune responses essential for defeating systemic microbial infections

    depend on intact innate and acquired immune responses. Recognition

  • 7/30/2019 Galley - Inflammation and Immunity

    12/119

    xi

    molecules, inflammatory cells, and the cytokines allow host tissues to

    recognise invading microbes and to initiate intercellular communication

    between the innate and acquired immune systems. However, activation of

    innate immunity may also occur in the absence of microbial recognition,

    through expression of internal signals produced by tissue ischaemia andnecrosis. Induction of the innate immune system can have catastrophic

    effects on patients with sepsis. Exaggerated production of cytokines and

    the induction of mediators such as nitric oxide, platelet activation factor,

    and prostaglandins have been implicated in the endothelial changes and

    induction of a procoagulant state, leading to hypotension, inadequate organ

    perfusion, and necrotic cell death, associated with multiorgan dysfunction

    syndrome. This article provides an overview of the T cell immune system,

    its regulation, and the influence of sepsis.

    Metalloproteinases and inflammation

    Andrew J Gearing

    Matrix metalloproteinases (MMPs) are a large family of zinc-containing

    endoproteinases, which have similar structures but differ in substrate

    specificity, cellular sources, and inducibility. MMP activity is controlled at

    the transcriptional level and by a family of at least four endogenous natural

    inhibitors (tissue inhibitors) of MMPs called TIMPs. MMPs cleaveprotein components of the extracellular matrix, membrane receptors, and

    cytokines, and have a role in cell extravasation. This article describes the

    action, regulation, and roles of MMPs in inflammatory and immune

    responses.

    Glucocorticoid therapy in septic shock

    Pierre-Edouard Bollaert

    Recent findings highlighting the role of the ability of the hypothalamic

    pituitaryadrenal axis to respond appropriately to a septic insult have led

    to a reappraisal of the use of steroids in septic shock. Recent work has

    suggested that physiological doses of corticosteroids given for a longer

    duration may be beneficial in catecholamine-dependent septic shock

    leading to a more rapid withdrawal of vasopressor therapy and a trend

    toward improved survival. A recent multicentre study of patients in septic

    shock has suggested a reduction in mortality in patients with relative

    adrenal insufficiency receiving replacement therapy with a combinationof hydrocortisone and fludrocortisone. This article describes studies of

    corticosteroid therapy in patients with septic shock and comments on the

    possible benefits of corticosteroid therapy in this population.

    INTRODUCTION

  • 7/30/2019 Galley - Inflammation and Immunity

    13/119

  • 7/30/2019 Galley - Inflammation and Immunity

    14/119

    1

    1: ImmunoparalysisJEAN-MARC CAVAILLON, HELEN F GALLEY

    Introduction

    Several studies indicate that depression of immune function induced by

    traumatic injury is aetiologically involved in the development of infection

    or sepsis. Nevertheless, the mechanisms behind the maintenance of the

    sustained suppression of immune function remain incompletely under-

    stood. Alterations of immune responses have been regularly reported in

    patients with systemic inflammatory response syndrome (SIRS).

    Trauma, haemorrhage, burns, surgery, or sepsis are associated with events

    such as tissue injury, blood loss, hypoxia, transfusion, microbial infection, andbacterial translocation, which contribute to an inflammatory response and

    affect the quality of the immune response. Drugs (for example, anaesthetics,

    opioids) also influence immune responses (Figure 1.1). Depressed immune

    status including decreased blood cell counts, low expression of surface

    markers (for example, MHC Class II antigen), altered natural killer (NK) cell

    activity, reduced cellular cytotoxicity and antigen presentation, poor

    proliferation in response to mitogens, and depressed cytokine production, are

    seen in vitro, and illustrated in vivo by anergy to skin test antigens. These

    observations led Roger Bone to coin the concept of compensatory anti-

    inflammatory response syndrome or CARS.1 Bone postulated that when the

    SIRS response predominated it was associated with an organ dysfunction and

    cardiovascular compromise leading to shock; in contrast, when CARS

    predominated it was characterised by anti-inflammatory responses associated

    with a suppression of the immune system termed immunoparalysis.1 It was

    initially accepted that the SIRS response occurred first and was followed in

    some patients by the CARS response. However, the two syndromes most

    probably occur concomitantly.2 In this article, we will describe the

    phenomenon of immunoparalysis in SIRS patients. Although alterations in

    immune response are probably associated with an enhanced sensitivity to

    nosocomial infections, there is no clear demonstration that they are directly

    responsible for poor outcome in sepsis. Indeed, since the investigations of

  • 7/30/2019 Galley - Inflammation and Immunity

    15/119

    2

    immune function may depend upon numerous parameters (for example,

    nature of the activators, cell types used, initial compartment of the cells, the

    cytokine investigated), interpretation of findings is not easy.

    Measures of immune dysfunction

    How can this modification of immune status be monitored? Lymphocyte

    and monocyte population changes and also HLA-DR expression are simple

    examples. Immune suppression can be assessed in vitro as poor lymphocyte

    proliferation in response to mitogens, reduced NK cell activity, reduced

    neutrophil function, reduced cytokine production, and in vivo anergy to

    skin test antigens (Figure 1.2).

    CRITICAL CARE FOCUS 10: INFLAMMATION AND IMMUNITY

    Figure 1.2 Approaches to monitoring immune status resulting from trauma, haemorrhage, burns,surgery, or sepsis.

    IMMUNE STATUS

    Immune suppression

    Lymphocyte

    proliferation

    Anergy to skin

    test antigens

    Natural killer

    cell activity Cytokineproduction

    Lymphocyte/monocyte

    population changes

    Anaesthesia

    Opioids

    Blood loss

    Transfusion

    Hypoxia

    Tissue injury

    Bacterial translocation

    Inflammatory response

    IMMUNE STATUS

    Figure 1.1 Contributory factors to altered immune status, resulting from trauma, haemorrhage,burns, surgery, or sepsis.

  • 7/30/2019 Galley - Inflammation and Immunity

    16/119

    3

    HLA-DR expression

    Abnormal antigen presentation has been observed in SIRS patients,3 and

    decreased HLA-DR expression on monocytes may contribute to this

    defect.4

    In a study by Hershman and colleagues,5

    60 trauma patients wereretrospectively divided into three groups: those with an uneventful recovery

    (n 17), those with recovery after major sepsis (n 27), and those who

    did not survive (n 16). HLA-DR expression on peripheral blood

    monocytes was compared with that of 77 healthy volunteers. After

    the initial injury, there was a significant decrease from normal in the three

    groups of trauma patients, and this returned to normal after one week in

    the group of patients who recovered uneventfully. In those who developed

    sepsis, HLA-DR expression took three weeks to return to normal and in

    the patients who did not survive, expression never returned to normal.Thisstudy demonstrated that monocyte HLA-DR antigen expression was able

    to distinguish those patients who survived severe trauma, from those who

    died (Figure 1.3). HLA-DR antigen expression correlated directly with the

    clinical course and identified a group of patients at high risk of infection

    and death following trauma.5 Low HLA-DR expression is now widely

    recognised as a good marker of the intensity of the immune depression and

    of increased risk of bacterial infection.6,7

    IMMUNOPARALYSIS

    uneventful recovery

    major sepsis

    death

    80

    60

    40

    %HLA-DRpo

    sitivemonocytes

    20

    Days after injury

    1 3 6 9 12 15 18

    Figure 1.3 Percentage of monocytes expressing HLA-DR expression in 60 trauma patients, 17 of

    whom had an uneventful recovery, 27 developed sepsis and 16 died. Dotted line represents mean

    HLA-DR expression in 77 healthy controls. Reproduced with permission from Hershman Met al.5

  • 7/30/2019 Galley - Inflammation and Immunity

    17/119

    4

    Lymphocyte proliferation

    Impaired lymphocyte transformation in SIRS patients was reported more

    than three decades ago.8 The impairment was proportional to the severity

    of the injury. In vitro lymphocyte proliferative response to antigens andmitogens, and in mixed lymphocyte reactions, are all significantly

    decreased in trauma patients.8,9 The length of depressed lymphocyte

    responses may exceed two weeks, and lower responses and longer depression

    have been observed in patients who become infected.9,10

    Natural killer cell activity

    The ability of the cell to mount an NK cell response provides another

    means to monitor immune status. NK cell activity was studied in burn andtrauma patients11 and was shown to be significantly depressed over a very

    long period of time for the more severely burned patients. Patients with

    lesser burns and traumatically injured patients had an altered NK activity

    for a shorter period. Interestingly, Blazar et al. further showed that stress-

    induced mediators (cortisol, epinephrine, glucagon) had the capacity to

    reduce NK activity in healthy volunteers.11 In the study by Maturana

    et al.,12 patients with septic shock (n 11) had also a markedly lower NK

    activity than healthy controls (n 10), independently of the effector:target

    cell ratio in the experimental system. In another study, NK cell activityin patients with septic shock (n 20) was also lower than in healthy

    volunteers (n 15). Pre-incubation of peripheral blood lymphocytes with

    either interferon- (IFN) or interleukin-2 (IL-2) enhanced NK cell

    activity in healthy controls but not in patients with sepsis indicating the

    difficulty in reversing the depressed immune responsiveness.13

    Neutrophil functions

    Although apoptosis of circulating neutrophils (PMN) is delayed in patientswith SIRS or sepsis,14 function of the cells is altered. This is the case of

    phagocytosis and bactericidal activity15 and of migration.16 The reduced

    responsiveness of PMN to chemoattractant agents may reflect the action of

    nitric oxide,16 the decreased expression of certain chemokine receptors,17

    or a deactivation occurring in the bloodstream after interacting with large

    amounts of circulating chemokines as indicated, for example, by the huge

    amounts of IL-8 found associated to PMN in septic patients.18

    Delayed hypersensitivity

    The in vitro evidence of immune depression is also reflected in vivo by

    tests of delayed type hypersensitivity. Several years ago, Christou and

    CRITICAL CARE FOCUS 10: INFLAMMATION AND IMMUNITY

  • 7/30/2019 Galley - Inflammation and Immunity

    18/119

    5

    colleagues19 skin tested 727 surgical patients with recall antigens prior to

    operation. Patients who had normal skin test responses were of similar age

    and had equal degrees of surgical procedures performed compared with

    those patients who were anergic (that is, had depressed skin test responses).

    Postoperatively, sepsis, mortality, and death from sepsis were significantlyhigher in the anergic population, reconfirming the hypothesis that skin test

    anergy in patients preoperatively is a signal of increased risk for septic

    complications and death in such patients. These authors20 also reported

    that surgical patients who were anergic to a battery of five skin test antigens

    had a two-fold higher rate of postoperative infection than those who

    reacted to only two antigens, and were more than five times more likely to

    die in the postoperative period.

    Cytokine production

    The analysis of sepsis and SIRS patients reveals a paradoxical situation: an

    overwhelming production of cytokines as assessed by their concentrations

    within the bloodstream21 and a profound reduction of the capacity of

    circulating cells to produce cytokines upon in vitro activation. Among

    pioneering work is the study by Wood and co-workers.22 They studied the

    production of IL-1 and IL-2 by peripheral blood mononuclear cells from

    23 burn patients and 23 matched controls. Serial measurements were made

    of IL-1 production by monocytes after stimulation with lipopolysaccharide

    (LPS), and of IL-2 production by lymphocytes after stimulation with the

    mitogen phytohaemagglutinin (PHA). Lymphocyte IL-2 production from

    12 patients with more than 30% body surface area burns revealed lower

    IL-2 production compared with patients with less than 30% burns.

    Patients with systemic sepsis also had lower IL-2 production than non-

    septic patients. IL-1 production was increased compared with controls

    early after injury, but was subsequently within the normal range regardless

    of burn size. The percentage of circulating helper T lymphocytes, the

    principal source of IL-2, was also reduced, although this did not always

    correlate with IL-2 production, which remained depressed after recovery of

    the T cell population. This study indicated that failure to produce IL-2,

    which is a powerful mediator of cellular immune responses, is an important

    mechanism underlying the defective cell-mediated immunity seen in burn

    patients.22 Surgery also leads to significant modulation of the immune

    system, and cytokine release in particular. Cabie et al. investigated the

    consequences of surgery on in vitro cytokine production by human

    monocytes stimulated by LPS.23 The responsiveness of cells obtained the

    day before, during and after surgery was compared in patients undergoingabdominal aortic surgery (n 9), carotid surgery (n 4), and spinal

    surgery (n 4). A significant decrease in monocyte tumour necrosis

    factor- (TNF), interleukin-1 (IL-1), and IL-1 production during

    IMMUNOPARALYSIS

  • 7/30/2019 Galley - Inflammation and Immunity

    19/119

    6

    surgery was reported, whereas IL-6 production remained unchanged.

    By day 2 following surgery, a significant increase in monocyte

    responsiveness was observed and levels of cytokine production were similar

    to initial values (Figure 1.4).

    CRITICAL CARE FOCUS 10: INFLAMMATION AND IMMUNITY

    ng/ml

    TNF IL-1

    10

    8

    6

    4

    2

    0

    ng/ml

    15

    12

    9

    6

    3

    0pre post pre post

    Figure 1.4 Influence of surgery on in vitro monocyte tumour necrosis factor (TNF) and

    interleukin-1 (IL-1) production in nine patients undergoing aortic surgery, four patients

    undergoing surgery for atheromatous lesions of the carotid artery,and four patients undergoing spinal

    surgery. Isolated cells were stimulated for 24 hours with 2 g/ml lipopolysaccharide (LPS).

    Pre one day before surgery and post 3 hours into the surgical procedure. Reproduced withpermission from Cabie A et al.23

    Characterisation of the ex vivo cytokine

    production in sepsis and SIRS

    Inflammation is characterised by an interplay between pro- and anti-

    inflammatory cytokines. Cytokines are commonly classified in one or theother category: IL-1, TNF, interferon- (IFN), IL-12, IL-18, and

    granulocyte-macrophage colony stimulating factor (GM-CSF) are well

    characterised as pro-inflammatory cytokines whereas IL-4, IL-10, IL-13,

  • 7/30/2019 Galley - Inflammation and Immunity

    20/119

    7

    IFN, and transforming growth factor- (TGF) are recognised as anti-

    inflammatory cytokines. However, this dichotomy may be too simplistic

    and it should be remembered that the amount of cytokine produced, the

    target cell, the activating signal, the timing and sequence of cytokine action,

    and even the experimental model, are parameters that greatly influencecytokine properties.24

    There have been several studies investigating in vitro stimulated cytokine

    release in patients with sepsis, in stimulated whole blood or isolated cell

    preparations.

    Monocyte-derived cytokines

    Several years ago, Munoz et al. studied the capacity of monocytes from

    septic patients to produce cytokines in response to LPS.25 Monocyteproduction of IL-1, IL-1, IL-6 and TNF in patients with sepsis

    syndrome (n 23) or non-infectious shock (n 6) was measured at

    admission and at regular intervals during intensive care unit (ICU) stay.

    Reduced LPS-induced production of cytokines was most pronounced in

    patients with Gram-negative infections. Recovery of cytokine production

    was observed among surviving patients but not in non-surviving patients.

    The data suggest that complex regulatory mechanisms can downregulate

    the production of cytokines by monocytes during severe infections.

    The suppression of lymphocyte and monocyte responses may reflectpotential defects in the upregulation of the IL-12 and IFN pathway.These

    cytokines exert protective effects during experimental endotoxaemia

    through upregulation of cellular immunity and phagocytic functions and are

    part of a positive regulatory feedback loop that enhances the production of

    the other. In a study by Ertel et al.,26 LPS-stimulated whole blood from 25

    critically ill patients and 12 healthy individuals was incubated with either

    recombinant human (rh) IL-12 or rhIFN.They found that, although IFN

    increased the release of IL-12 from LPS-stimulated whole blood from

    healthy subjects in a dose-dependent manner, this effect was not seen incritically ill patients. IL-12 enhanced the secretion of IFN in healthy

    subjects, but was ineffective in critically ill patients. Although the anti-

    inflammatory cytokine, IL-10, but not IL-4, mimicked suppression of the

    IL-12-IFN pathway similar to that observed during critical illness, the

    release of anti-inflammatory cytokines (IL-4, IL-10, TGF) was decreased

    in LPS-stimulated blood from critically ill patients. This study suggested

    that deactivation of IL-12 and IFN-producing leukocytes occurred in vivo.

    Lymphocyte-derived cytokines

    The T lymphocyte population comprises both T helper 1 (Th1) and T helper 2

    (Th2) subsets. Th1 cells produce cytokines predominantly concerned with

    IMMUNOPARALYSIS

  • 7/30/2019 Galley - Inflammation and Immunity

    21/119

    8

    pro-inflammatory responses (IFN, TNF, IL-2) and Th2 produce

    cytokines concerned with anti-inflammatory responses (IL-4, IL-10, and

    IL-13). It has been regularly reported that the production of Th1 cytokines

    was mainly altered in SIRS patients, whereas this was not the case for the

    Th2 cytokines.27,28 However, the study of purified T cells from 37 severelyinjured trauma patients showed that T cell anergy was a global depression of

    both Th1 and Th2 cytokine profile. Interestingly, depressed T cell

    proliferation and depressed cytokine production correlate to poor clinical

    outcome.29 Muret et al. further illustrated that infectious and non-infectious

    SIRS exert a more subtle modulation on circulating cell reactivity. They

    investigated the production of IL-2, IL-4, IL-5, and IL-10 by peripheral

    blood mononuclear cells in 13 patients with sepsis and 13 patients with non-

    infectious inflammation (patients undergoing cardiac surgery with

    cardiopulmonary bypass).30

    Cytokine release after activation of lymphocyteswith either concanavalin A (ConA), PHA, or anti-CD3 antibodies was

    studied. ConA-induced IL-10 was reduced in both groups of patients

    compared with healthy controls. In sepsis patients, ConA-induced IL-2,

    IL-5, and IL-10 production was decreased but not that released in response

    to PHA or anti-CD3. In cardiac patients, only anti-CD3-induced IL-10

    production was reduced.These data indicate that subtle modifications of the

    reactivity of circulating cells occur during infectious and non-infectious

    inflammation, dependent on the cell stimulus. This suggests that regulation

    of both Th1 and Th2 responses is occurring in patients with sepsis and SIRS,and that the cell stimulant used determines the results achieved.

    Neutrophil-derived cytokines

    McCall and co-workers31 reported that neutrophils from patients with

    the sepsis syndrome were consistently resistant to LPS stimulation such

    that synthesis of IL-1 was depressed. This downregulation occurred

    concomitant with an upregulation in expression of the type 2 IL-1 receptor

    (IL-1r2). Similar findings were not seen in uninfected patients with severe

    trauma or shock from other causes. In another study,32 Marie et al. reported

    depressed IL-8 release from neutrophils in patients who had undergone

    cardiac surgery with cardiopulmonary bypass and patients with sepsis. Cells

    were activated with either LPS or heat-killed streptococci. Compared with

    healthy controls, the release of IL-8 in both groups of patients was

    significantly reduced whether activated by LPS or by heat-killed

    streptococci.These observations suggest that stressful conditions related to

    inflammation, independently of infection, resulted in hyporeactivity of

    circulating neutrophils, suggestive of LPS tolerance. However, in vitroexperiments suggested that neutrophils from healthy controls (in contrast to

    monocytes) could not be rendered tolerant to LPS. Interestingly, while

    IL-1 receptor antagonist (IL-1ra) was shown to be enhanced in whole

    CRITICAL CARE FOCUS 10: INFLAMMATION AND IMMUNITY

  • 7/30/2019 Galley - Inflammation and Immunity

    22/119

    9

    blood assays in meningococcal infection,33 Marie et al. found that the release

    of IL-1ra by isolated PMN was diminished in SIRS patients.34

    Not a universal defect

    It is noteworthy that the diminished capacity of leukocytes from SIRS

    patients to produce cytokines as compared with healthy donors is not

    obtained with all activating signals. For example, McCall et al.,31 in their

    studies of patients with sepsis, failed to observe a decreased capacity of

    PMN to release IL-1 when they used heat-killed staphylococci, while

    immune-depression was revealed with LPS. In sepsis, IL-2, IL-5, and

    IL-10 production in response to conA was reduced, but not when

    phytohaemagglutinin or anti-CD3 were used.30

    More recently, in cardiac arrest and resuscitated patients, hyporeactivity,assessed in terms of TNF production, was observed with LPS stimulation,

    but not with heat-killed staphylococci (Adrie et al., personal communication).

    A similar dissociation between stimuli that reveal hyporeactivity (LPS, CpG,

    IL-1,TNF) and those that do not (for example, staphylococci, streptococci)

    has also been observed in trauma patients (Adib-Conquy et al., personal

    communication). These observations suggest that differential alteration of

    signalling pathways may occur within the cells of SIRS patients, depending

    upon the nature of the activating agent and the nature of the cytokine being

    analysed.

    Mechanisms of hyporesponsiveness of monocytes

    Desensitising agents

    The presence of deactivating or immunosuppressive agents within the

    bloodstream may contribute to the hyporeactivity of circulating leukocytes.

    IL-10 has been identified as a major functional deactivator of monocytes in

    human septic shock plasma,35 and TGF was shown in animal models of

    haemorrhagic shock and of sepsis to be the causative agent of the depressed

    splenocyte responsiveness.36,37 Furthermore, there is accumulating

    evidence for a strong interaction between components of the nervous and

    the immune systems, and numerous neuromediators have been shown to

    behave as immunosuppressors. Catecholamines suppress the activity of

    immunocompetent cells and are found at higher concentrations in stressful

    situations.38 Catecholamines are known to inhibit TNF production39 and

    to favour IL-10 release.40 Similarly, alpha-melanocyte-stimulating

    hormone contributes to immunosuppression by inducing IL-10 productionby human monocytes.41 In addition, vasoactive intestinal peptide and

    pituitary adenylate cyclase-activating polypeptide directly inhibit endotoxin

    induced pro-inflammatory cytokine secretion.42 SIRS is also associated

    IMMUNOPARALYSIS

  • 7/30/2019 Galley - Inflammation and Immunity

    23/119

    10

    with an activation of the hypothalamuspituitaryadrenal axis, which leads

    to the release of glucocorticoids, well known for their potent ability to limit

    cytokine production.43 Finally, prostaglandins are produced during sepsis

    and can also contribute to the downregulation of cytokine production.44

    Endotoxin neutralising molecules

    As mentioned previously, the reduced capacity of monocytes to produce

    inflammatory cytokines has been established, particularly in experimental

    systems using LPS as a triggering agent. Since numerous recent studies have

    analysed the hyporeactivity phenomenon using whole blood cultures, it is

    possible that endotoxin-neutralising molecules have interfered in these

    studies. Indeed, Adrie et al. have shown that the hyporeactivity to LPS was

    both an intrinsic property of circulating monocytes, as well as the reflectionof a specific neutralising activity within the plasma of SIRS patients (personal

    communication). It has been reported that plasma of septic patients

    contains large amounts of LPS binding protein (LBP), which can either

    inhibit the LPS molecules,45 or transfer LPS to lipoproteins,46 known for

    their inhibitory activity towards LPS.47 Furthermore, sera from septic

    patients contain amounts of soluble CD14, which also favours the shuttle of

    LPS towards lipoproteins.48

    Toll-like receptors

    Toll-like receptors (TLR) are a family of receptors that recognise

    components of bacteria, virus, parasites, and fungi, and induce a pro-

    inflammatory response by several cell types. So far, 10 human TLRs

    differing in their specificity for microbial components have been cloned,

    which respond to various components, including LPS from Gram-negative

    bacteria, lipopeptides of Gram-positive cell walls, bacterial DNA, and

    flagella. TLR4 was identified as the receptor for LPS and requires the

    presence of an extracellular accessory protein called MD-2. CD14

    physically associates with LPS complexed with LBP and transfers the

    endotoxin to the TLR4 and MD-2 dimer; each component of this complex

    is required for efficient LPS-induced signalling.

    Many parameters of immunoparalysis observed in SIRS patients are

    reminiscent of the endotoxin tolerance phenomenon, which characterises the

    refractoriness of cells or the inability of whole animals to respond to a second

    endotoxin challenge shortly after a first encounter.49 Because recent studies

    reported a downregulation of surface expression of TLR4 in endotoxin-

    tolerant macrophages,50,51 it was of interest to investigate the expression ofthis molecule on the surface of monocytes from SIRS patients. A decreased

    expression of TLR4, but not TLR2, on CD14 positive cells was found in

    11 trauma patients compared with 6 healthy subjects (Adib-Conquy et al.,

    CRITICAL CARE FOCUS 10: INFLAMMATION AND IMMUNITY

  • 7/30/2019 Galley - Inflammation and Immunity

    24/119

    11

    personal communication). However, this lower expression of TLR4 is not

    sufficient to explain the decreased capacity of the cells to respond to stimuli.

    Indeed, while LPS-induced TNF is decreased, this was not the case of LPS-

    induced IL-1ra and IL-10. This latter observation suggests that the defect

    may occur at the different signalling pathways within the cell rather than atthe initiation of the signalling cascade on the cell surface.

    Nuclear factor kappa B

    Transcription factors are DNA-binding proteins which regulate gene

    expression. Nuclear factor kappa B (NFB) is one such transcription factor

    which is critical for maximal expression of many cytokines involved in the

    pathogenesis of inflammation. Activation and regulation of NF

    B is tightlycontrolled by a group of inhibitory proteins (IB), which maintain NFB in

    an inhibited state in the cytoplasm of effector cells.The sequence of events

    leading to NFB activation involves phosphorylation, ubiquitination, and

    proteolysis of IB, allowing exposure of a nuclear recognition site. NFB

    then migrates to the nucleus, binds to specific promoter sites, and activates

    transcription of target genes (for example,TNF, IL-1, IL-6, IL-8). NFB,

    part of the Rel/NF-B family of transcription factors, is involved in the

    regulation of immune and acute-phase responses at the transcriptional level.

    Rel proteins can be divided into two groups based on their structures,

    functions, and modes of synthesis. The first group of Rel proteins consists

    of p65 (also known as RelA), c-Rel, and RelB, each of which contains one

    or more transcriptional-activation domain necessary for gene induction.The

    second group consists of p105 and p100, which, upon proteolytic

    processing, give rise to p50 and p52, respectively. Members of both groups

    of Rel proteins can form homo- or heterodimers. Studies have shown that

    the transactivator form of NFB is the p65 unit, whereas the p50 unit has

    shown no or minimal activation capacity.

    To investigate the role of NFB in the mechanism of endotoxin tolerance

    in macrophages, Blackwell and co-workers52 used a rat alveolar macrophage

    cell line made endotoxin tolerant by exposure to low concentrations of

    LPS for 48 hours. This treatment induced a state of tolerance such that

    subsequent exposure to high-dose LPS resulted in decreased production

    of cytokines compared with LPS-sensitive cells. This decreased cytokine

    production was associated with impaired activation of NFB with depletion

    of both RelA and p50. This study suggested endotoxin tolerance might be

    mediated by depletion of RelA/p50, which could limit the amount of

    NFB available for activation and inhibit transcription of NFB-dependent

    genes. On the other hand, Ziegler-Heitbrock and colleagues demonstratedthat endotoxin tolerance of monocytes was associated with an increase of

    the inactive p50 homodimer and a decrease of the p50/p65 active

    heterodimer.53

    IMMUNOPARALYSIS

  • 7/30/2019 Galley - Inflammation and Immunity

    25/119

    12

    Accordingly, Adib-Conquy et al. studied NFB expression and dimer

    characteristics in mononuclear cells of patients with sepsis and major

    trauma and healthy controls.54,55 The expression of p50/p65 heterodimer

    was significantly reduced in all patients as compared with controls. The

    p50/p50 homodimer was reduced in the survivors of sepsis. Subsequentin vitro stimulation of mononuclear cells with LPS did not induce further

    NFB nuclear translocation: the survivors of sepsis showed low expression

    of both p50/p65 and p50/p50, while non-survivors of sepsis showed a

    predominance of the inactive homodimer and a low p50/p65:p50/p50

    ratio when compared with controls. In the latter group of patients there

    was a negative correlation between plasma IL-10 levels and the

    p50/p65:p50/p50 ratio after in vitro LPS stimulation (r 0.8,

    P 0.04). The reduced expression of nuclear NFB was not due to its

    inhibition by IB since very low expression of IB as well as low levels ofp65 and p50, were found in the cytoplasm of mononuclear cells from

    sepsis patients when compared with controls. These results demonstrate

    that upon LPS activation, mononuclear cells of systemic inflammatory

    response syndrome patients show patterns of NFB expression that

    resemble those reported during LPS-tolerance: global downregulation of

    NFB in survivors of sepsis and presence of large amounts of the inactive

    homodimer in the non-survivors of sepsis.54

    In trauma patients, after 1, 3, 5, and 10 days following admission of

    patients in the intensive care units, expression of both p50/p65 heterodimersand p50/p50 homodimers was significantly reduced compared with

    controls. After LPS stimulation in vitro, the p50/p65:p50/p50 ratio was

    significantly lower in cells from trauma patients than from healthy controls

    and the ex vivo expression of IB was higher. Although no direct

    correlation was found between levels of IL-10 or TGF and NFB, these

    immunosuppressive cytokines were significantly elevated in the trauma

    patients by 10 days after admission. This long-term low basal and LPS-

    induced activation of NFB might be linked to immunoparalysis.55

    Signalling pathways

    There are still very few studies in humans addressing whether some

    alterations of the signalling pathways might explain part of the

    immunodepression seen in circulating cells in SIRS patients. Most

    interestingly, Learn et al.56 reported that in septic patients the repressed

    production of IL-1 and the selective elevation of the secreted form of

    IL-1ra in response to LPS, was linked to a probable alteration in the

    interleukin-1 receptor-associated kinase (IRAK) signalling pathway anda maintained efficient phosphatidylinositol 3-kinase-dependent signalling

    pathway. In murine model of sepsis, it was reported that inhibition of p59fyn

    phosphorylation and kinase activity was associated with T lymphocyte

    CRITICAL CARE FOCUS 10: INFLAMMATION AND IMMUNITY

  • 7/30/2019 Galley - Inflammation and Immunity

    26/119

    13

    IL-2 production and proliferation,57 whilst activation of MAPK p38 was

    associated with T cell immune dysfunction.58

    Cells derived from inflammatory foci

    The hyporesponsiveness of peripherally derived cells in terms of the capacity

    of the cells to produce cytokines is not a generalised phenomenon, and cells

    derived from inflammatory foci are, in contrast, activated. In a baboon model

    of inflammation, the capacity of alveolar macrophages after unilateral lung

    irradiation was studied.2 Over a 1 month period, bronchoalveolar lavage

    was undertaken in five baboons, macrophages were recovered and

    production of TNF was measured.This study showed a significant increase

    of spontaneous, LPS-, staphylococci- and streptococci-induced TNF

    release in macrophages from the inflamed lung compared with the initialvalues. Macrophages from lungs of patients with acute respiratory distress

    syndrome (ARDS) also clearly demonstrated no deactivation of the cells.

    Schwartz et al.59 investigated activation of the transcription factors NFB,

    nuclear factor-IL-6 (NF-IL-6), cyclic adenosine monophosphate (cAMP)-

    responsive element binding protein, serum protein-1 (SP-1), and activating

    protein-1 (AP-1) in alveolar macrophages from six patients with ARDS and

    from six control patients without lung injury. Activation of NFB in alveolar

    macrophages observed in patients with ARDS had increased compared with

    control patients, but there was no increase in the activation of the othertranscription factors. Moine and colleagues60 subsequently showed

    decreased cytoplasmic levels of p50, p65 and c-Rel in alveolar macrophages

    from patients with ARDS, consistent with enhanced migration of liberated

    NFB dimers from the cytoplasm to the nucleus.

    When leukocytes are derived from the peritoneal cavity or the gut

    of patients suffering peritonitis,61 inflammatory bowel diseases,62 or

    endometriosis,63,64 LPS-induced cytokine production by peritoneal

    macrophages or mononuclear cells from the lamina propria was enhanced

    as compared with healthy controls. Following injection of endotoxin, theproduction of IFN by intra-epithelial lymphocytes was enhanced upon

    stimulation as compared with control animals.65 A similar enhanced

    activity of intra-epithelial T lymphocytes after endotoxaemia was observed

    when cellular cytotoxicity and proliferation were monitored. Altogether,

    these examples illustrate that local inflammation is associated with an

    enhanced activity of resident and/or infiltrating leukocytes, whilst systemic

    inflammation is associated with a reduced activity of circulating leukocytes.

    Conclusion

    Sepsis and non-infectious SIRS are paradoxically associated with an

    exacerbated production of cytokines, as assessed by their presence in

    IMMUNOPARALYSIS

  • 7/30/2019 Galley - Inflammation and Immunity

    27/119

    14

    biological fluids, and a diminished ability of circulating cells to produce

    cytokine upon in vitro activation.This might represent a protective response

    against an overwhelming dysregulation of the pro-inflammatory process, but

    on the other hand it may induce a state of immune paralysis (endogenous

    immunosuppression) leading to an increased risk of subsequentnosocomial infections.66 However, cellular hyporeactivity is not a global

    phenomenon and some signalling pathways are unaltered and allow the cells

    to respond normally to certain stimuli. Furthermore, during sepsis and

    SIRS, cells derived from tissues or inflammatory foci are either fully

    responsive to ex vivo stimuli or even primed, in contrast to cells derived from

    haematopoietic compartments (blood), which are hyporeactive. In addition

    to cytokine production, NFB activity within leukocytes reflects cellular

    hyporeactivity. Thus the immunoparalysis reported in sepsis and SIRS

    patients, often revealed by a diminished capacity of leukocytes to respondto LPS, is not a generalised phenomenon, and SIRS is associated with a

    compartmentalised responsiveness involving either anergic or primed cells.

    References

    1 Bone RC, Grodzin CJ, Balk RA. Sepsis: a new hypothesis for pathogenesis ofthe disease process. Chest1997;112:23543.

    2 Cavaillon J-M, Adib-Conquy M, Cloz-Tayarani I, Fitting C. Immuno-

    depression in sepsis and SIRS assessed by ex vivo cytokine production is nota generalized phenomenon: a review.J Endotoxin Res 2001;7:8593.

    3 Polk HC Jr, George CD,Wellhausen SRet al. A systematic study of host defense

    processes in badly injured patients.Ann Surg1986;204:28299.4 Livingston DH, Appel SH,Wellhausen SR, Sonnenfeld G, Polk HC. Depressed

    interferon gamma production and monocyte HLA DR expression after severeinjury.Arch Surg1988;123:130912.

    5 Hershman MJ, Cheadle WG, Wellhausen SR, Davidson PF, Polk HC Jr.Monocyte HLA-DR antigen expression characterizes clinical outcome in thetrauma patient. Br J Surg1990;77:2047.

    6 Cheadle WG, Hershman MJ, Wellhausen SR, Polk HC Jr. HLA-DR antigen

    expression on peripheral blood monocytes correlates with surgical infection.Am J Surg1991;161:63945.

    7 Van den Berk JMM, Oldenburger RHJ, van den Berg AP et al. Low HLA-DRexpression on monocytes as a prognostic marker for bacterial sepsis after livertransplantation. Transplantation 1997;63:18468.

    8 Salo M, Merikanto J, Eskola J, Nieminen S, Aho AJ. Impaired lymphocytetransformation after accidental trauma.Acta Chir Scand1970;145:36772.

    9 Keane RM, Birmingham W, Shatney CM, Winchurch RA, Munster AM.Prediction of sepsis in the multitraumatic patients by assays of lymphocyte

    responsiveness. Surg Gynecol Obst1983;156:1637.10 Levy EM, Alharbi SA, Grindlinger G, Black PH. Changes in mitogen

    responsiveness lymphocyte subsets after traumatic injury: relation todevelopment of sepsis. Clin Immunol Immunopathol1984;32:22433.

    11 Blazar BA, Rodrick ML, OMahony JB et al. Suppresion of natural killer-cellfunction in humans following thermal and traumatic injury. J Clin Immunol1986;6:2636.

    CRITICAL CARE FOCUS 10: INFLAMMATION AND IMMUNITY

  • 7/30/2019 Galley - Inflammation and Immunity

    28/119

    15

    12 Maturana P, Puente J, Miranda D, Sepulveda C, Wolf ME, Mosnaim AD.Natural killer cell activity in patients with septic shock.J Crit Care 1991;6:425.

    13 Puente J, Carvajal T, Parra S et al. In vitro studies of natural killer cell activity inseptic shock patients. Response to a challenge with alpha-interferon andinterleukin-2. Int J Clin Pharmacol Ther Toxicol1993;31:2715.

    14 Jimenez MF,Watson WG, Parodo J et al. Dysregulated expression of neutrophilapoptosis in the systemic inflammatory response syndrome. Arch Surg1997;132:126370.

    15 van Dijk WC, Verbrugh HA, van der Tol ME et al. Interactions of phagocyticand bacterial cells in patients with bacteremia caused by Gram-negative rods.

    J Infect Dis 1980;141:4419.

    16 Benjamin CF, Ferreira SH, Cunha FDQ. Role of nitric oxide in the failure ofneutrophil migration in sepsis.J Infect Dis 2000;182:21423.

    17 Cummings CJ, Martin TR, Frevert CW et al. Expression and function of thechemokine receptor CXCR1 and CXCR2 in sepsis. J Immunol 1999;162:23416.

    18 Marie C, Fitting C, Cheval C et al. High levels of leukocyte-associatedinterleukin-8 upon cell-activation and in patients with sepsis syndrome. InfectImmun 1997;65:86571.

    19 Christou NV. Host-defence mechanism in surgical patients: a correlative studyof the delayed hypersensitivity skin-test response, granulocyte function andsepsis. Can J Surg1985;28:3946.

    20 Christou NV, Meakins JL, MacLean LD. The predictive role of delayedhypersensitivity in preoperative patients. Surg Gynecol Obstet1981;152:297301.

    21 Cavaillon J-M. Possibilities and problems of cytokine measurements. In RedlH, Schlag G, eds. Cytokines in Severe Sepsis and Septic Shock, Prog. Inflam. Res.,Basel: Birkhuser Publishing Ltd. 1998, 95119.

    22 Wood JJ, Rodrick ML, OMahony JB et al. Inadequate interleukin 2 production.A fundamental immunological deficiency in patients with major burns. AnnSurg1984;200:31120.

    23 Cabie A, Fitting C, Farkas JC et al. Influence of surgery on in-vitro cytokineproduction by human monocytes. Cytokine 1992;4:57680.

    24 Cavaillon J-M. Pro- versus anti-inflammatory cytokines: myth or reality. Cell MolBiol2001;47:695702.

    25 Munoz C, Carlet J, Fitting C, Misset B, Bleriot JP, Cavaillon J-M. Dysregulationof in vitro cytokine production by monocytes during sepsis. J Clin Invest1991;88:174754.

    26 Ertel W, Keel M, Neidhardt Ret al. Inhibition of the defense system stimulating

    interleukin-12 interferon-gamma pathway during critical Illness. Blood1997;89:161220.

    27 OSullivan ST, Lederer, JA, Horgan AF, Chin DHL, Mannick JA, Rodrick ML.Major injury leads to predominance of the T helper-2 lymphocyte phenotypeand diminished interleukin-12 production associated with decreased resistanceto infection.Ann Surg1995;222:48292.

    28 Mack VE, McCarter MD, Naana HA, Calvano SE, Daly J-M. Dominance of

    T helper 2 type cytokine after severe injury.Arch Surg1996;131:13039.29 Puyana JC, Pellegrini JD, De AK, Kodys K, Silva WE, Miller CL. Both

    T-helper-1- and T-helper-2-type lymphokines are depressed in posttraumaanergy.J Trauma 1998;44:103745.

    30 Muret J, Marie C, Fitting C, Payen D, Cavaillon J-M. Ex vivo T-lymphocytederived cytokine production in SIRS patients is influenced by experimentalprocedures. Shock 2000;13:16974.

    31 McCall CE, Grosso-Wilmoth LM, LaRue K, Guzman RN, Cousart SL.Tolerance to endotoxin-induced expression of the interleukin-1 beta gene in

    IMMUNOPARALYSIS

  • 7/30/2019 Galley - Inflammation and Immunity

    29/119

    16

    blood neutrophils of humans with the sepsis syndrome. J Clin Invest1993;91:85361.

    32 Marie C, Muret J, Fitting C, Losser MR, Payen D, Cavaillon J-M. Reducedex vivo interleukin-8 production by neutrophils in septic and nonseptic systemicinflammatory response syndrome. Blood1998;91:343946.

    33 Van Deuren M,Van Der Ven-Jongekrijg H, Demacker PNM et al. Differentialexpression of proinflammatory cytokines and their inhibitors during the courseof meningococcal infections.J Infect Dis 1994;169:15761.

    34 Marie C, Muret J, Fitting C, Payen D, Cavaillon J-M. Interleukin-1 receptorantagonist production during infectious and non-infectious systemicinflammatory response syndrome. Crit Care Med2000;28:227783.

    35 Brandtzaeg P, Osnes L, vsteb R, Jo GB, Westwik AB, Kierulf P. Netinflammatory capacity of human septic shock plasma evaluated by a monocyte-based target cell assay: identification of interleukin-10 as a major functionaldeactivator of human monocytes.J Exp Med1996;184:5160.

    36 Ayala A, Meldrum DR, Perrin MM, Chaudry IH.The release of transforming

    growth factor-beta following haemorrhage: its role as a mediator of hostimmunosuppression. Immunology 1993;79:47984.

    37 Ayala A, Knotts JB, Ertel W et al. Role of interleukin 6 and transforming growthfactor-beta in the induction of depressed splenocyte responses following sepsis.

    Arch Surg1993;128:8994.38 Jones S, Romano F. Dose- and time-dependent changes in plasma catecholamines

    in response to endotoxin in conscious rats. Circ Shock 1989;28:5968.39 Severn A, Rapson NT, Hunter CA, Liew FY. Regulation of tumor necrosis

    factor production by adrenaline and by -adrenergic agonists. J Immunol1992;148:34415.

    40 van der Poll T, Coyle SM, Barbosa K, Braxton CC, Lowry SF. Epinephrine

    inhibits tumor necrosis factor-alpha and potentiates interleukin-10 productionduring human endotoxemia.J Clin Invest1996;97:71319.

    41 Luger TA, Kalden DH, Scholzen TE, Brzoska T. -melanocyte-stimulatinghormone as a mediator of tolerance induction. Pathobiology 1999;67:31821.

    42 Delgado M, Pozo D, Martinez C et al.Vasoactive intestinal peptide and pituitaryadenylate cyclase-activating polypeptide inhibit endotoxin-induced TNFproduction by macrophages: in vitro and in vivo studies. J Immunol1999;162:235867.

    43 Chrousos GP. The hypothalamicpituitaryadrenal axis and immune-mediatedinflammation.N Engl J Med1995;332:135162.

    44 Choudhry MA, Ahmad S, Ahmed Z, Sayeed MM. Prostaglandin E2 down-

    regulation of T cell IL-2 production is independent of IL-10 during Gramnegative sepsis. Immunol Lett1999;67:12530.

    45 Zweigner J, Gramm HJ, Singer OC, Wegscheider K, Schumann RR. Highconcentration of lipopolysaccharide-binding protein in serum of patients withsevere sepsis or septic shock inhibit the lipopolysaccharide response in humanmonocytes. Blood2001;98:38008.

    46 Vreugdenhil ACE, Snoeck AMP, vant Veer C, Greve JWM, Buurman WA.LPS-binding protein circulates in association with apoB-containing lipoproteinsand enhances endotoxin-LDL/VLDL interaction. J Clin Invest 2001;107:22534.

    47 Cavaillon J-M, Fitting C, Haeffner-Cavaillon N, Kirsch J, Warren HS.

    Cytokine response by monocytes/macrophages to free and lipoprotein-bound lipopolysaccharide. Infect Immun 1990;58:237582.

    48 Kitchens RL,Thompson PA,Viriyakosol S, OKeefe GE, Munford RS. PlasmaCD14 decreases monocyte responses to LPS by transferring cell-bound LPS toplasma lipoproteins.J Clin Invest2001;108:48593.

    CRITICAL CARE FOCUS 10: INFLAMMATION AND IMMUNITY

  • 7/30/2019 Galley - Inflammation and Immunity

    30/119

    17

    49 Cavaillon J-M.The non specific nature of endotoxin tolerance. Trends Microbiol1995;3:3204.

    50 Nomura F, Akashi S, Sakao Y et al. Endotoxin tolerance in mouse peritonealmacrophages correlates with down-regulation of surface Toll-like receptor 4expression.J Immunol2000;164:34769.

    51 Medvedev AE, Kopydlowski KM, Vogel SN. Inhibition of lipopolysaccharide-induced signal transduction in endotoxin-tolerized mouse macrophages:dysregulation of cytokine, chemokine, and Toll-like receptor 2 and 4 geneexpression.J Immunol2000;164:556474.

    52 Blackwell TS, Blackwell TR, Christman JW. Induction of endotoxin tolerancedepletes nuclear factor-kappaB and suppresses its activation in rat alveolar

    macrophages.J Leukocyte Biol1997;62:88591.53 Ziegler-Heitbrock L, Wedel A, Schraut W et al. Tolerance to lipopolysaccharide

    involves mobilization of nuclear factorB with predominance of p50homodimers.J Biol Chem 1994;269:170014.

    54 Adib-Conquy M, Adrie C, Moine P et al. Nuclear factorB expression in

    mononuclear cells of septic patients resembles that observed in LPS-tolerance.Am J Respir Crit Care Med2000;162:187783.

    55 Adib-Conquy M, Asehnoune K, Moine P, Cavaillon J-M. Long term impaired

    expression of nuclear factorB and IB in peripheral blood mononuclearcells of patients with major trauma.J Leukocyte Biol2001;70:308.

    56 Learn CA, Boger MS, Li L, McCall CE. The phosphatidylinositol 3 kinasepathway selectively controls sIL-1ra not interleukin-1 production in the septicleukocytes.J Biol Chem 2001;276:202349.

    57 Choudhry MA, Uddin S, Sayeed MM. Prostaglandin E2 modulation of p59fyn

    tyrosine kinase in T lymphocytes during sepsis.J Immunol1998;160:92935.58 Song GY, Chung CS, Chaudry IH, Ayala A. MAPK p38 antagonism as a novel

    method of inhibiting lymphoid immune suppression in polymicrobial sepsis.Am J Physiol Cell Physiol2001;281:C6629.

    59 Schwartz MD, Moore EE, Moore FA et al. Nuclear factor kappa B is activatedin alveolar macrophages from patients with acute respiratory distress syndrome.Crit Care Med1996;24:128592.

    60 Moine P, McIntyre R, Schwartz MD et al. NF-kappaB regulatory mechanismsin alveolar macrophages from patients with acute respiratory distress syndrome.

    Shock 2000;13:8591.61 Fieren MWJ, Van Den Bemd GJ, Bonta IL. Endotoxin-stimulated peritoneal

    macrophages obtained from continuous ambulatory peritoneal dialysis patients

    show an increased capacity to release interleukin-1 in vitro during infectious

    peritonitis. Eur J Clin Invest1990;B4537.62 Rugtveit J, Nilsen EM, Bakka A, Carlsen H, Brandtzaeg P, Scott H. Cytokine

    profiles differ in newly recruited and resident subsets of mucosal macrophagesfrom inflammatory bowel disease. Gastroenterol1997;112:1493505.

    63 Rana N, Braun DP, House R, Gebel H, Rotman C, Dmowski WP. Basal andstimulated secretion of cytokines by peritoneal macrophages in women withendometriosis. Fertil Steril1996;65:92530.

    64 Wu MY, Ho HN, Chen SU, Chao KH, Chen CD, Yang YS. Increase in theproduction of IL-6, IL-10 and IL-12 by LPS stimulated peritoneal macrophagesfrom women with endometriosis.Am J Reprod Immunol1999;41:10611.

    65 Nssler NC, Stange B, Nussler AK et al. Upregulation of intraepithelial

    lymphocyte function in the small intestinal mucosa in sepsis. Shock2001;16:4548.

    66 Munford RS, Pugin J. Normal responses to injury prevent systemicinflammation and can be immunosuppressive. Am J Respir Crit Care Med2001;163:31621.

    IMMUNOPARALYSIS

  • 7/30/2019 Galley - Inflammation and Immunity

    31/119

    2: Apoptosis and the

    inflammatory processNIGEL R WEBSTER

    Introduction

    Cells that are damaged by injury, such as by mechanical damage or

    exposure to toxic chemicals, undergo swelling, from disruption of the

    ability of the plasma membrane to control the passage of ions and water,

    with consequent leakage of cell contents, leading to inflammation of

    surrounding tissues.This process is called necrosis. Cells which are induced

    to commit suicide, in contrast, shrink, and the mitochondrial membrane

    becomes breached, such that release of cytochrome c occurs. Chromatin

    (DNA and protein) in the nucleus becomes degraded into small,membrane-wrapped fragments, and the phospholipid phosphatidylserine,

    which is normally hidden within the plasma membrane, is exposed on the

    surface. This is then bound by receptors on phagocytic cells such as

    macrophages, which engulf the cell fragments, leading to a quiet orderly

    removal of dead cells. This pattern of events is called programmed cell

    death or apoptosis.The cellular machinery of programmed cell death is as

    intrinsic to the cell as, for example, mitosis. This article will describe the

    regulation and process of apoptosis and its relevance to disease, including

    the inflammatory response in patients with sepsis.

    Identification of apoptosis

    A series of careful observational studies in the 1950s and 1960s

    demonstrated the importance of physiological cell death in development. By

    the 1970s, a process of cell death, characterised by a rigid set of structural

    changes, was also observed in a wide variety of physiological circumstances:

    negative selection in the immune system, cytotoxic T cell killing, atrophy

    induced by hormones and other stimuli, the growth and regression oftumours, and tissue development after exposure to teratogens.

    These distinctive structural changes characterising cell death were

    identical to those found in cell death during normal development and

    18

  • 7/30/2019 Galley - Inflammation and Immunity

    32/119

    19

    raised the possibility that a programmed death pathway, similar to that in

    development, might also occur in adult tissues in response to a variety of

    stimuli. This type of death was called apoptosis a term derived from the

    Greek word meaning the dropping of leaves from the trees. The word

    apoptosis is often mispronounced it derives from apo- and -ptosis andtherefore the second p is silent. This term is applied to a group of

    characteristic structural and molecular events which separate this type of

    cell deletion from necrosis (Box 2.1). In contrast to necrosis, which involves

    a group of cells simultaneously, apoptosis may occur in a single cell

    surrounded by a group of viable cells. Apoptosis is a selective process for

    deletion of cells in various biological systems and, in a similar manner to

    proliferation, is tightly regulated, with both processes playing essential roles

    in the homeostasis of renewable tissues.13

    APOPTOSIS AND THE INFLAMMATORY PROCESS

    Box 2.1 Key facts about apoptosis

    Normal process modelling in vertebrate development cell loss accompanying atrophy in adult tissues deletion of B and T lymphocytes occurs widely in tumours

    Characteristic morphological changes

    Characteristic biochemical changes

    The process of apoptosis

    Structurally, the dying cell loses contact with its neighbours, undergoes a

    dramatic process of bubbling, blebbing, and shrinkage, and disintegrates

    into a cluster of membrane-bound fragments. Inside, there are compacted

    organelles and prominent and characteristic chromatin condensation.

    Apoptotic cells in tissues are rapidly recognised and phagocytosed by their

    neighbours, or by specialised phagocytes, in whose phagosomes they are

    safely destroyed within a few hours.Tissues can shrink to half their original

    cell number in a day or two, with little disturbance in structure, no

    inflammatory process, and few accumulating dead cells.

    Apoptosis versus necrosis

    All of this is very different from the changes in cells exposed to

    severe toxicological injury or major degrees of hypoxia, where damage to

  • 7/30/2019 Galley - Inflammation and Immunity

    33/119

    20

    CRITICAL CARE FOCUS 10: INFLAMMATION AND IMMUNITY

    Box 2.2 Morphological features of apoptosis versus necrosis

    Necrosis

    Loss of membrane integrity

    Swelling of cytoplasm and mitochondria

    Ends with total cell lysis

    No vesicle formation, complete lysis

    Disintegration of organelles

    Apoptosis

    Mitochondria becomes leaky

    Membrane blebbing, no loss of integrity

    Aggregation of chromatin at the nuclear membrane

    Shrinking of cytoplasm and condensation of nucleus

    Ends with fragmentation of cell into smaller bodies

    Formation of membrane bound vesicles (apoptotic bodies)

    Box 2.3 Biochemical features of apoptosis versus necrosis

    Necrosis

    Loss of regulation of ion homeostasis

    No energy requirement

    Random digestion of DNA

    Postlytic DNA fragmentation (late event of death)

    Apoptosis Tightly regulated process

    Energy (ATP)-dependent

    Non-random fragmentation of DNA (ladder pattern)

    Pre-lytic DNA fragmentation

    Release of cytochrome c into cytoplasm by mitochondria

    Activation of caspase cascade

    Translocation of membrane phosphatidyl-serine

  • 7/30/2019 Galley - Inflammation and Immunity

    34/119

    21

    energy-dependent membrane ion pumps leads to progressive cellular

    swelling and rupture necrosis. When this occurs there is usually an acute

    inflammatory reaction, apparently stimulated by neutrophil chemotactic

    factors originating from intracellular proteins lost from the necroticcells. The characteristic morphological and biochemical features and

    physiological significance of necrosis versus apoptosis are given in Boxes 2.2,

    2.3, and 2.4. Figure 2.1 shows a normal eosinophil and one undergoing

    apoptosis.

    APOPTOSIS AND THE INFLAMMATORY PROCESS

    Box 2.4 Physiological significance of apoptosis

    versus necrosis

    Necrosis

    Affects groups of contiguous cells

    Evoked by non-physiological disturbances

    Phagocytosis by macrophages

    Significant inflammatory response

    Apoptosis

    Affects individual cells

    Induced by physiological stimuli (lack of growth factors,

    hormonal environment) Phagocytosis by adjacent cells or macrophages

    No inflammatory response

    A B

    Figure 2.1 A transmission electron photomicrograph of (A) a normal eosinophil and (B) an

    eosinophil undergoing apoptosis showing aggregation of chromatin, blebbing, and shedding of

    intracytoplasmic granules.

  • 7/30/2019 Galley - Inflammation and Immunity

    35/119

    22

    Why do we need apoptosis?

    There are two reasons why apoptosis is physiologically vital.The first is the

    role it plays in fetal development and other key processes. This is most

    eloquently seen in the change from a tadpole to a frog. The resorption ofthe tadpole tail at the time of its metamorphosis into a frog occurs by

    apoptosis. In man the formation of the fingers and toes of the fetus requires

    the removal, by apoptosis, of the tissue between them. The formation of

    synapses between neurones requires that surplus cells be eliminated by

    apoptosis, and the sloughing off of the endometrium at the start of

    menstruation also occurs by apoptosis.

    The second reason for apoptosis is the need to destroy cells that

    represent a threat to the integrity of the organism. This might include, for

    example, cells infected with viruses one of the methods by whichcytotoxic T (Tc) lymphocytes kill virus-infected cells is by inducing

    apoptosis and some viruses are able to mount countermeasures

    (see Chapter 3 in this volume). Apoptosis is also important in cells with

    DNA damage where disruption of proper embryonic development leading

    to birth defects can occur, or the cell can become cancerous. Cells respond

    to DNA damage by increasing their production of p53 a potent inducer

    of apoptosis. Mutations in the p53 gene an oncogene producing a

    defective protein, are often found in cancer cells, and radiation and

    chemotherapeutic agents used in cancer therapy induce apoptosis in sometypes of cancer cells. Mice with both copies of p53 deleted develop multiple

    malignancies, and p53 mutation is associated with many human cancers.

    Following DNA damage, for example, by radiation, p53 levels rise, and

    proliferating cells arrest in the G1 phase of mitosis. This allows time for

    DNA repair prior to the next round of replication. This arrest is mediated

    by stimulation of expression of p21CIP1, a cyclin kinase inhibitor.

    Why can apoptosis be a problem?

    Although the process of apoptosis is physiologically essential for both

    development and removal of dangerous cells, initiation of the sequence

    of events leading to cell death through apoptosis can lead to both unwanted

    removal of healthy cells, and propagation of inflammatory responses

    through release of cytokines (see description of the actions of caspase

    enzymes below).

    Caenorhabditis elegans and the genetics of apoptosis

    C. elegans is a hermaphrodite nematode worm with a life cycle from egg to

    sexual maturity of about 3 days. The genome of this organism has been

    CRITICAL CARE FOCUS 10: INFLAMMATION AND IMMUNITY

  • 7/30/2019 Galley - Inflammation and Immunity

    36/119

    23

    fully sequenced and contains 19 099 genes. The adult hermaphrodite

    consists of exactly 959 somatic cells of very precisely determined lineage

    and function. Development control occurs through apoptotic removal of

    exactly 131 cells, and thus C. elegans is an ideal organism to study the

    genetics of apoptosis. It has been found that three key genes triggerapoptosis: ced-3 and ced-4 (C.elegans cell death genes) and egl-1 (C.elegans

    egg laying defective gene) (Figure 2.2).

    Ced-3 has a mammalian counterpart, originally known as interleukin 1

    converting enzyme or ICE, but now termed caspase 1 (Cys catalytic Asp

    targeting protease). Thirteen caspases are known in mammalian systems

    and have conserved sequence and subunit structure; of these four play key

    effector roles in apoptosis and four are initiators in the activation process.4

    Ced-4 acts as an adapter for caspase activation in C. elegans; the

    mammalian counterpart is called apoptosis activating factor (Apaf-1).A fourth gene in C. elegans promotes survival, that is, it acts as a negative

    regulator of apoptosis, ced-9. The mammalian equivalent is the BCL-2

    family of genes. Bcl-2 and ced-9 proteins are 23% identical, and bcl-2 can

    substitute for ced-9 in C. elegans. However, in higher animals, bcl-2 is a

    member of a large family of closely related proteins, some of which promote

    survival and some death (Box 2.5). The similarities between ced-3 and

    ICE, ced-4 and Apaf-1, and between ced-9 and bcl-2 strongly suggest that

    programmed cell death in C. elegans parallels apoptosis in higher animals,

    but in a much more simplified form. Figure 2.2 shows the C. elegans

    apoptosis genes and their human equivalents.5

    APOPTOSIS AND THE INFLAMMATORY PROCESS

    Pro-apoptotic

    ced-3

    ced-4Anti-apoptotic

    ced-9

    caspase-1(ICE)

    Apaf-1

    BCL-2 gene family

    Figure 2.2 A transmission electron micrograph of the nematode worm Caenorhabditis elegans.

    Its apoptosis genes ( left) with the mammalian equivalents (right) are indicated.

    Signals for apoptosis

    The cascade of events leading to apoptosis takes place as a result of either

    the withdrawal of positive signals (that is, signals needed for continuedsurvival), or the initiation of negative signals (that is, those which instigate

    cell death). Signals can arise within the cell or from so-called death

    activators binding to receptors at the cell surface.6

  • 7/30/2019 Galley - Inflammation and Immunity

    37/119

    24

    CRITICAL CARE FOCUS 10: INFLAMMATION AND IMMUNITY

    Box 2.5 The bcl-2 protein family

    Pro-apoptotic

    Bad

    Bak

    Bax

    Bcl-Xs

    Bid

    Bik

    Anti-apoptotic

    AL

    Bcl-2

    Bcl-W

    Bcl-XL

    Mcl-1

    Withdrawal of positive signals

    The continued survival of most cells requires that they receive continuous

    stimulation from other cells and, for many, continued adhesion to the

    surface on which they are growing. Some examples of positive signals

    include specific growth factors for neurones, and interleukin-2 (IL-2), an

    essential factor for the mitosis of lymphocytes.

    Receipt of negative signals

    Internal signals

    In a healthy cell, the outer membranes of mitochondria express the protein

    bcl-2 on their surface. The role of the mitochondrion in apoptosis is

    discussed further below. Bcl-2 is bound to a molecule of the protein

    Apaf-1, and internal damage in the cell causes bcl-2 to release Apaf-1.This

    results in leakage of cytochrome c from mitochondria into the cytoplasm.

    The released cytochrome c and Apaf-1 bind to molecules of caspase 9.

    The resulting complex of cytochrome c, Apaf-1 and caspase 9 (with ATP)

    is called an apoptosome and these aggregate in the cytosol. Caspase 9activates other caspases, which leads to digestion of structural proteins in

    the cytoplasm, degradation of chromosomal DNA, and ultimately

    phagocytosis of the cell (see caspases below).

  • 7/30/2019 Galley - Inflammation and Immunity

    38/119

    25

    External signals

    External signals which initiate apoptosis include increased levels of oxidants

    within the cell or damage to DNA by these oxidants or other agents, such as

    ultraviolet light, x rays, and chemotherapeutic drugs. In addition there are

    several molecules that bind to specific receptors on the cell surface and signalthe start of apoptosis. Signalling pathways link key receptors to the caspase

    proteolytic cascade.These death activators include tumour necrosis factor

    (TNF) which binds to the TNF receptor; lymphotoxin (also known as

    TNF) which also binds to the TNF receptor; and Fas ligand (FasL), a

    molecule that binds to the Fas cell-surface receptor (also called CD95).

    Fas expression and immune function

    Fas is a transmembrane receptor protein which is very widely distributed,

    and is constitutively expressed in some cells for example, liver, or induced

    in cells such as lymphocytes on activation. The activating ligand, Fas-L,

    is expressed in a narrower range of cells, including antigen-activated

    T lymphocytes, testis, eye, and central nervous system. Persistent and strong

    stimulation of CD4T helper lymphocytes results in expression of Fas-L.

    During ablation of the immune repertoire in the fetal/neonatal period, self-

    antigen stimulation causes killing of self-reactive B lymphocytes and self or

    mutual killing of the Fas-L expressing T lymphocytes. The development of

    tolerance against a potential antibody represents a similar process. Sometumours also express Fas-L constitutively for example, melanoma and

    lung tumours. This renders them resistant to immune intervention any

    visiting lymphocytes are induced to commit suicide through apoptosis.

    Expression of Fas-L in testis, eye, and central nervous system gives rise to

    the phenomenon of immune privilege, in which these tissues do not reject

    allografts, but kill invading lymphocytes instead.

    Mechanism of Fas-L and TNF-induced apoptosis

    When Fas binds Fas-L, changes occur in the cell membrane; its cytoplasmic

    domain contains a sequence called the death domain (DD), a sequence

    critically required for stimulated apoptosis.The changes in the cell membrane

    result in incorporation of an intracellular DD-containing protein called

    Fas-associated death domain (FADD) protein (synonymous with a protein

    previously named MORT1). The N-terminal region of FADD contains

    another domain referred to as death effector domain or DED.This associates

    with another DED at the N-terminus of pro-caspase-8 (the pro-caspase

    formerly known as pro-FLICE or MACH1, or Mort-associated ced-3homologue). Recruitment into the complex results in dimerisation of pro-

    caspase-8, releasing the active caspase-8. Caspase-8 then initiates the whole

    effector protease cascade by acting on pro-caspase-3 to liberate caspase 3.

    APOPTOSIS AND THE INFLAMMATORY PROCESS

  • 7/30/2019 Galley - Inflammation and Immunity

    39/119

    26

    A similar sequence involves the TNF receptor (TNFR), signalled by

    TNF, and DR3 or Apo3 signalled by Apo3L. TNF binds to TNFR,

    recruiting the DD protein TRADD, and acts as an intermediary for binding

    FADD, activating pro-caspase 8 in the same way as FAS. However,

    TRADD can also be occupied by another DD protein RIP, which signalsto the nuclear factor B (NFB) and Jun pathways, which are inhibitory

    to apoptosis. TNF is expressed mainly in activated lymphocytes and

    macrophages, while TNFR is expressed ubiquitously. DR3 is expressed by

    spleen and thymus cells, and Apo3L by T cells. This complex pathway is

    represented in a simplified form in Figure 2.3.

    CRITICAL CARE FOCUS 10: INFLAMMATION AND IMMUNITY

    Apo2L

    TRAIL

    Death adaptor

    ICE/

    Caspase 1 Caspase 8

    Caspase 3m

    Bad, Bax

    Cytochrome c

    APOPTOSIS Nucleus

    Bcl2/Bcl-XL

    FADD TRADD TRAF 1

    FLIPNFB

    activation

    FasL Apo3L TNF

    DR4/

    DR5

    Fas/

    CD95

    DR3 TNF R1Free radicals

    NO Steroids

    +/

    Figure 2.3 The complex pathway involved in the signalling of apoptosis events.For explanation and

    abbreviations/acronyms see text.

    Caspases

    Caspases are a group of proteases, named because they cleave proteins

    mostly each other at aspartic acid (Asp) residues. Caspases initially exist

    as immature pro-caspases (zymogens) and require processing to be

    activated, in a similar way to the proteins of the clotting or complement

    cascades. There are three basic domains in the immature form: the pro-

    domain, the large subunit, and the small subunit. Some caspases are

    initiators that is, their targets are downstream effector caspases. The

    initiator caspases have a large pro-domain, since these are regulated by

    proteins other than caspases.The effector caspases have small pro-domains

    since they are directly regulated by other caspases. In other words, the

    pro-domain is important for proteinprotein interactions. The large

    pro-domain interacts with other proteins in the cell, containing caspaserecruitment domains (CARD domains). These interactions lead to

    the cleavage and activation of the inhibitor caspases, which then go on to

    cleave the effector caspases. Once activated the long pro-domain caspases

  • 7/30/2019 Galley - Inflammation and Immunity

    40/119

    27

    may then cleave caspases with short pro-domains to achieve their

    activation.7

    The full process of apoptosis in mammalian cells involves several caspases,

    some of which have specialised purposes; for example a whole subgroup is

    involved in cytokine activation rather than apoptosis (Figure 2.4).

    APOPTOSIS AND THE INFLAMMATORY PROCESS

    m

    cytochrome c

    cytochrome c

    Apaf-1

    pro-caspase 9

    pro-caspase 3 caspase 3

    APOPTOSIS

    caspase 9

    (ced-4 equivalent)

    mitochondrion

    Figure 2.4 A simplified representation of the role of caspases in apoptosis.For abbreviations/acronyms

    see text.

    The initiator caspases include caspases 2, 8, 9, and 10, and act on pro-

    caspase 3. The effector caspases are 3, 6, and 7, which act on proteins

    involved in cell structure.The inflammatory caspases mentioned earlier in

    this chapter are 1, 4, 5, 11, 12, and 13. Substrates for these enzymes are

    pro-IL-1 and pro-IL-18. IL-18 is a relatively recently elucidated cytokine,

    which has similar effects to IL-1. Thus the signals for apoptosis can also

    result in propagation of inflammatory events via pro-inflammatory

    cytokine release. The net effects of caspases are outlined in Box 2.6.

    Box 2.6 Net effect of caspase activity

    Halts cell cycle progression

    Disables homeostatic and repair mechanisms

    Initiates detachment of the cell from surrounding tissue structure

    Disassembles structured components Marks dying cell for engulfment by other cells such as

    macrophages

  • 7/30/2019 Galley - Inflammation and Immunity

    41/119

    28

    CRITICAL CARE FOCUS 10: INFLAMMATION AND IMMUNITY

    Redundancy of caspases

    Caspase-1 or ICE is not a single enzyme, but one of a family of related

    proteases, which are co-expressed.This appears to provide redundancy for

    an important function and circumvents what was once an embarrassment,that caspase-1 knockout mice still undergo apoptosis.

    The role of mitochondria in apoptosis

    Recently, the mitochondrion has been identified as playing a central role in

    apoptosis. Several findings support this idea: members of the BCL-2 gene

    family localise into the mitochondrial membrane and inhibit apoptosis and

    a mutant BCL-2, which cannot insert into the mitochondrial membrane,is a less potent inhibitor of apoptosis. BCL-2/-XL can also recruit Apaf-1

    into the mitochondrial membrane and may prevent Apaf-1 from activating

    caspases, thereby inhibiting apoptosis. In addition some mitochondrial

    proteins induce apoptosis when leaked into the cytosol: during apoptosis,

    cytochrome c and apoptosis inducing factor (AIF) are released from the

    mitochondria and, with other factors, such as Apaf-1 and Apaf-3, lead to

    caspase activation and apoptosis. Increased levels of BCL-2 can prevent

    the release of these molecules, whereas caspase inhibitors cannot. This

    indicates the release of cytochromec

    and AIF is downstream of BCL-2function but upstream of the caspases. Apoptosis is also associated

    with a change in the mitochondrial membrane potential, a phenomenon

    known as permeability transition. The permeability transition can be

    blocked by excess BCL-2 but not by inhibitors of caspases, indicating that

    the permeability transition is downstream of BCL-2 but upstream

    of caspase activation. BCL-2, bcl-XL, and another apoptosis-related

    gene, BAX, are capable of forming selective ion pores in membranes, thus

    forming channels in the mitochondrial membrane that could regulate

    permeability transition and the release of molecules such as cytochrome c

    and AIF.

    Apoptosis and disease

    Apoptosis and cancer

    Cancer cells are known to use inhibition of apoptosis.8 For example, one

    of the two human papilloma viruses (HPV) that have been implicated

    in causing cervical cancer produces a protein (E6) which binds andinactivates the apoptosis promoter p53. EpsteinBarr Virus (EBV), the

    cause of mononucleosis and a cause of Burkitts lymphoma, secretes a

    protein that stimulates cells to increase endogenous production of bcl-2.

  • 7/30/2019 Galley - Inflammation and Immunity

    42/119

    29

    Both these actions make the cell more resistant to apoptosis and therefore

    enables cancer cells to continue to proliferate. Some B-cell leukaemias and

    lymphomas express high levels of bcl-2, thus blocking apoptotic signals

    that they may receive. Melanoma cells avoid apoptosis by inhibiting the

    expression of the gene encoding Apaf-1, and some cancer cells, especiallylung and colon cancer cells, secrete elevated levels of a soluble decoy

    molecule that binds to FasL, preventing binding to Fas and blocking

    cytotoxic T cell killing mechanisms.

    Apoptosis and AIDS

    The hallmark of acquired immunodeficiency syndrome (AIDS) is the

    decline in CD4 T cell numbers. These cells are responsible, directly

    or indirectly (as T helper cells), for all immune responses. Human

    immunodeficiency virus (HIV) invades CD4 and one might assume that

    it is this infection by HIV that causes loss of CD4T cells. However, fewer

    than 1 in 100 000 CD4T cells in the blood of AIDS patients are actually

    infected with the virus and, although the mechanism is not clear, apoptosis

    appears to be involved. Since all T cells, both infected and uninfected,

    express Fas, expression of an HIV gene (termed Nef) in a HIV-infected cell

    causes the cell to express high levels of FasL at its surface while preventing

    an interaction with self-Fas preventing self-elimination. However, when the

    infected T cell encounters an uninfected cell (for example, in a lymphnode), the interaction of FasL with Fas on the uninfected cell kills it by

    apoptosis.

    Apoptosis and organ transplants

    For many years it has been known that certain tissues including the anterior

    chamber of the eye and the testes are immunologically privileged sites

    such that antigens within these sites fail to elicit an immune response fromhigh constitutive expression of high levels of FasL. This finding raises the

    possibility of a new way of preventing graft rejection. However, animal

    studies have produced mixed results. Allografts that have been genetically

    modified to express FasL have shown increased survival for kidneys but not

    for hearts or islets of Langerhans.

    Apoptosis and sepsis

    Apoptosis is also relevant to inflammatory responses in sepsis. Theimmune/inflammatory balance in sepsis reflects the balance between pro- and

    anti-inflammatory responses, and the balance between hypermetabolic/

    hyperdynamic versus hypometabolic/hypodynamic responses.There is also

    APOPTOSIS AND THE INFLAMMATORY PROCESS

  • 7/30/2019 Galley - Inflammation and Immunity

    43/119

    30

    probably a balance between ischaemic or necrotic cell death and apoptotic

    cell death. Ultimately, the balance between all these components probably

    then governs death or survival of the patient (Figure 2.5).

    CRITICAL CARE FOCUS 10: INFLAMMATION AND IMMUNITY

    Hypermetabolic

    Hyperdynamic

    Hypometabolic

    Hypodynamic

    Exaggerated inflammatory

    response

    SEPSIS

    Immune hyporesponsiveness

    Ischaemic/

    necrotic

    Apoptotic

    cell death