The innate defences of the body · tious agents. Many bacteria fail to survive for long on the skin...

15
SECTION TWO The adversaries – host defences 67 9 The body has both ‘innate’ and ‘adaptive’ immune defences When an organism infects the body, the defence systems already in place may well be adequate to prevent replica- tion and spread of the infectious agent, thereby prevent- ing development of disease. These established mechanisms are referred to as constituting the ‘innate’ immune system. However, should innate immunity be insufficient to parry the invasion by the infectious agent, the so-called ‘adaptive’ immune system then comes into action, although it takes time to reach its maximum efficiency (Fig. 9.1). When it does take effect, it generally eliminates the infective organism, allowing recovery from disease. The main feature distinguishing the adaptive response from the innate mechanism is that specific memory of infec- tion is imprinted on the adaptive immune system, so that should there be a subsequent infection by the same agent, a particularly effective response comes into play with remark- able speed. It is worth emphasizing, however, that there is close synergy between the two systems, with the adaptive mechanism greatly improving the efficiency of the innate response. The contrasts between these two systems are set out in Table 9.1. On the one hand, the soluble factors such as lysozyme and complement, together with the phagocytic cells, contribute to the innate system, while on the other the lymphocyte-based mechanisms that produce antibody and T lymphocytes are the main elements of the adap- tive immune system. Not only do these lymphocytes pro- vide improved resistance by repeated contact with a given infectious agent, but the memory with which they become endowed shows very considerable specificity to that infec- tion. For instance, infection with measles virus will induce a memory to that microorganism alone and not to another virus such as rubella. DEFENCES AGAINST ENTRY INTO THE BODY A variety of biochemical and physical barriers operate at the body surfaces Before an infectious agent can penetrate the body, it must overcome biochemical and physical barriers that operate at the body surfaces. One of the most important of these is the skin, which is normally impermeable to the majority of infec- tious agents. Many bacteria fail to survive for long on the skin because of the direct inhibitory effects of lactic acid and fatty acids present in sweat and sebaceous secretions and the lower pH to which they give rise (Fig. 9.2). However, should there be skin loss, as can occur in burns, for example, infec- tion becomes a major problem. The membranes lining the inner surfaces of the body secrete mucus, which acts as a protective barrier, inhibit- ing the adherence of bacteria to the epithelial cells, thereby preventing them from gaining access to the body. Microbial and other foreign particles trapped within this adhesive mucus may be removed by mechanical means such as cili- ary action, coughing and sneezing. The flushing actions of tears, saliva and urine are other mechanical strategies that help to protect the epithelial surfaces. In addition, many of the secreted body fluids contain microbicidal factors, Introduction In the preceding chapters, we have outlined some of the fundamental characteristics of the myriad types of microparasites and macroparasites that may infect the body. We now turn to consider the ways in which the body seeks to defend itself against infection by these organisms. The innate defences of the body 9 infection re-infection innate immunity adaptive immunity specific immunologic memory disease recovery no disease 1 2 3 4 6 5 Figure 9.1 Innate and adaptive immunity. An infectious agent first encounters elements of the innate immune system. These may be sufficient (1) to prevent disease but if not, disease may result (2). The adaptive immune system is then activated (3) to produce recovery (4) and a specific immunologic memory (5). Following re-infection with the same agent, no disease results (6) and the individual has acquired immunity to the infectious agent.

Transcript of The innate defences of the body · tious agents. Many bacteria fail to survive for long on the skin...

  • SECTION TWO The adversaries – host defences

    67

    9

    The body has both ‘innate’ and ‘adaptive’ immune defences When an organism infects the body, the defence systems

    already in place may well be adequate to prevent replica-

    tion and spread of the infectious agent, thereby prevent-

    ing development of disease. These established mechanisms

    are referred to as constituting the ‘innate’ immune system.

    However, should innate immunity be insufficient to parry

    the invasion by the infectious agent, the so-called ‘adaptive’

    immune system then comes into action, although it takes

    time to reach its maximum efficiency ( Fig. 9.1 ). When it does

    take effect, it generally eliminates the infective organism,

    allowing recovery from disease.

    The main feature distinguishing the adaptive response

    from the innate mechanism is that specific memory of infec-

    tion is imprinted on the adaptive immune system, so that

    should there be a subsequent infection by the same agent, a

    particularly effective response comes into play with remark-

    able speed. It is worth emphasizing, however, that there is

    close synergy between the two systems, with the adaptive

    mechanism greatly improving the efficiency of the innate

    response.

    The contrasts between these two systems are set out

    in Table 9.1 . On the one hand, the soluble factors such as

    lysozyme and complement, together with the phagocytic

    cells, contribute to the innate system, while on the other

    the lymphocyte-based mechanisms that produce antibody

    and T lymphocytes are the main elements of the adap-

    tive immune system. Not only do these lymphocytes pro-

    vide improved resistance by repeated contact with a given

    infectious agent, but the memory with which they become

    endowed shows very considerable specificity to that infec-

    tion. For instance, infection with measles virus will induce

    a memory to that microorganism alone and not to another

    virus such as rubella.

    DEFENCES AGAINST ENTRY INTO THE BODY

    A variety of biochemical and physical barriers operate at the body surfaces Before an infectious agent can penetrate the body, it must

    overcome biochemical and physical barriers that operate at

    the body surfaces. One of the most important of these is the

    skin, which is normally impermeable to the majority of infec-

    tious agents. Many bacteria fail to survive for long on the

    skin because of the direct inhibitory effects of lactic acid and

    fatty acids present in sweat and sebaceous secretions and the

    lower pH to which they give rise ( Fig. 9.2 ). However, should

    there be skin loss, as can occur in burns, for example, infec-

    tion becomes a major problem.

    The membranes lining the inner surfaces of the body

    secrete mucus, which acts as a protective barrier, inhibit-

    ing the adherence of bacteria to the epithelial cells, thereby

    preventing them from gaining access to the body. Microbial

    and other foreign particles trapped within this adhesive

    mucus may be removed by mechanical means such as cili-

    ary action, coughing and sneezing. The flushing actions of

    tears, saliva and urine are other mechanical strategies that

    help to protect the epithelial surfaces. In addition, many

    of the secreted body fluids contain microbicidal factors,

    Introduction

    In the preceding chapters, we have outlined some of the fundamental characteristics of the myriad types of microparasites and macroparasites that may infect the body. We now turn to consider the ways in which the body seeks to defend itself against infection by these organisms.

    The innate defences of the body 9

    infection re-infection

    innateimmunity

    adaptive immunity

    specificimmunologic

    memory

    disease recovery no disease

    1 2

    3

    4 6

    5

    Figure 9.1 Innate and adaptive immunity. An infectious agent first encounters elements of the innate immune system. These may be sufficient (1) to prevent disease but if not, disease may result (2). The adaptive immune system is then activated (3) to produce recovery (4) and a specific immunologic memory (5). Following re-infection with the same agent, no disease results (6) and the individual has acquired immunity to the infectious agent.

  • 68

    CHAPTERCHAPTER9 The innate defences of the body

    e.g. the acid in gastric juice, spermine and zinc in semen,

    lactoperoxidase in milk, and lysozyme in tears, nasal secre-

    tions and saliva.

    The phenomenon of microbial antagonism is associ-

    ated with the normal bacterial flora of the body. These

    commensal organisms suppress the growth of many

    potentially pathogenic bacteria and fungi at superficial

    sites, first by virtue of their physical advantage of previ-

    ous occupancy, especially on epithelial surfaces, second by

    competing for essential nutrients, or third by producing

    inhibitory substances such as acid or colicins. The latter are

    a class of bactericidins that bind to the negatively charged

    surface of susceptible bacteria and form a voltage-depen-

    dent channel in the membrane, which kills by destroying

    the cell's energy potential.

    DEFENCES ONCE THE MICROORGANISM PENETRATES THE BODY Despite the general effectiveness of the various barriers, micro-

    organisms successfully penetrate the body on many occasions.

    When this occurs, two main defensive strategies come into

    play, based on:

    • the mechanism of phagocytosis, involving engulfment

    and killing of microorganisms by specialized cells, the

    ‘professional phagocytes’

    • the destructive effect of soluble chemical factors, such as

    bactericidal enzymes.

    Two types of professional phagocyte Perhaps because of the belief that professionals do a better

    job than amateurs, the cells that shoulder the main burden

    of our phagocytic defences have been labelled ‘professional

    phagocytes’. These consist of two major cell families, as orig-

    inally defined by Elie Metchnikoff, the Russian zoologist

    ( Box 9.1 ; Fig. 9.3 ):

    • the large macrophages

    • the smaller polymorphonuclear granulocytes, which

    are generally referred to as polymorphs or neutrophils

    because their cytoplasmic granules do not stain with

    haematoxylin and eosin.

    As a very crude generalization, it may be said that the poly-

    morphs provide the major defence against pyogenic (pus-

    forming) bacteria, while the macrophages are thought to be

    at their best in combating organisms capable of living within

    the cells of the host.

    Macrophages are widespread throughout the tissues Macrophages originate as bone marrow promonocytes,

    which develop into circulating blood monocytes ( Fig. 9.4 )

    and finally become the mature macrophages, which are

    widespread throughout the tissues and collectively termed

    the ‘mononuclear phagocyte system’ ( Fig. 9.5 ). These

    macrophages are present throughout the connective tissue

    Innate immune system Adaptive immune system

    Major elements

    Soluble factors Lysozyme, complement, acute phase proteins, e.g. C-reactive protein, interferon

    Antibody

    Cells Phagocytes Natural killer cells

    T lymphocytes

    Response to microbial infection

    First contact + + +

    Second contact + + + + +

    Non-specific; no memory Resistance not improved by repeated contact

    Specific; memory Resistance improved by repeated contact

    Table 9.1 Comparison of innate and adaptive effector immune systems

    Innate immunity is sometimes referred to as ‘natural’, and adaptive as ‘acquired’. There is considerable interaction between the two systems. ‘Humoral’ immunity due to soluble factors contrasts with immunity mediated by cells. Primary contact with antigen produces both adaptive and innate responses, but if the same antigen persists or is encountered a second time the specific adaptive response to that antigen is much enhanced.

    lysozymein most

    tears, nasal secretions and

    saliva

    sebaceousgland

    secretions

    commensalorganisms in

    gut and vagina

    spermine insemen

    mucus

    cilia liningtrachea

    skin

    acid instomach

    biochemical chemical and physical

    Figure 9.2 Exterior defences. Most of the infectious agents encountered by an individual are prevented from entering the body by a variety of biochemical and physical barriers. The body tolerates a variety of commensal organisms, which compete effectively with many potential pathogens.

  • 69

    SECTION TWO The adversaries – host defences

    and are associated with the basement membrane of small

    blood vessels. They are particularly concentrated in the

    lung (alveolar macrophages), liver (Kupffer cells) and the

    lining of lymph node medullary sinuses and splenic sinu-

    soids ( Fig. 9.6 ), where they are well placed to filter off

    foreign material ( Fig. 9.7 ). Other examples are the brain

    microglia, kidney mesangial cells, synovial A cells and

    osteoclasts in bone. In general, these are long-lived cells that

    depend upon mitochondria for their metabolic energy and

    show elements of rough-surfaced endoplasmic reticulum

    ( Fig. 9.8 ) related to the formidable array of different secre-

    tory proteins that these cells generate.

    Polymorphs possess a variety of enzyme-containing granules The polymorph is the dominant white cell in the blood-

    stream and, like the macrophage, shares a common haemo-

    poietic stem cell precursor with the other formed elements

    of the blood. It has no mitochondria, but uses its abundant

    cytoplasmic glycogen stores for its energy requirements;

    therefore, glycolysis enables these cells to function under

    anaerobic conditions, such as those in an inflammatory

    focus. The polymorph is a non-dividing, short-lived cell,

    with a segmented nucleus; the cytoplasm is characterized by

    an array of granules, which are illustrated in Figure 9.9 .

    A B

    Figure 9.4 Phagocytic cells. (A) Blood monocyte and (B) polymorphonuclear neutrophil, both derived from bone marrow stem cells. (Courtesy of P.M. Lydyard.)

    4. alveolarmacrophages

    7. splenicmacrophages

    1. bloodmonocytes

    8. lymph noderesident andrecirculating

    macrophages

    precursors inbone marrow

    6. brainmicroglial cells

    2. liverKupffer cells

    3. kidneymesangialphagocytes

    synovialA cells

    osteoclasts

    5. connectivetissuehistiocytes

    Figure 9.5 The mononuclear phagocyte system. Tissue macrophages are derived from blood monocytes, which are manufactured in the bone marrow. (The numbers relate to those in Fig. 9.6 .)

    Elie Metchnikoff (1845–1916)

    This perceptive Russian zoologist can legitimately be regarded as the father of the concept of cellular immunity, in which it is recognized that certain specialized cells mediate the defence against microbial infections. He was intrigued by the motile cells of transparent starfish larvae and made the critical observation that a few hours after introducing a rose thorn into the larvae, the rose thorn became surrounded by the motile cells. He extended his investigations to mammalian leukocytes, showing their ability to engulf microorganisms, a process that he termed ‘phagocytosis’ (literally, eating by cells).

    Because he found this process to be even more effective in animals recovering from an infection, he came to the conclusion that phagocytosis provided the main defence against infection. He defined the existence of two types of circulating phagocytes: the polymorphonuclear leukocyte, which he termed a ‘microphage’, and the larger ‘macrophage’.

    Although Metchnikoff held the somewhat polarized view that cellular immunity based upon phagocytosis provided the main, if not the only, defence mechanism against infectious microorganisms, we now know that the efficiency of the phagocytic system is enormously enhanced through cooperation with humoral factors, in particular antibody and complement.

    Figure 9.3 Elie Metchnikoff (1845–1916). (Courtesy of the Wellcome Institute Library, London.)

    Box 9.1 Lessons in Microbiology

  • 70

    CHAPTERCHAPTER9 The innate defences of the body

    capillaryendothelial cells

    sinusoidal space

    hepatocyte

    Kupffer cell

    endothelial cell

    basementmembrane

    mesangialmacrophage

    endotheliumpodocyte

    pneumocytetype II

    macrophage

    pneumocytetype I

    air space

    basementmembrane

    capillary

    capillary

    mesotheliumbasement membrane

    reticular fibres

    macrophage

    capillary

    microglial cell

    ependyma

    nerve cell basementmembrane

    macrophage

    sinus endothelium

    erythrocytesmacrophages

    endothelialcell

    circulatingblood monocyte

    1Kupffer cellsin the liver

    intraglomerularmesangial cells of the kidney

    alveolar macrophagesin the lung

    connective tissuehistiocytes

    brain microglia spleen sinusmacrophages

    lymph node sinusmacrophages

    2 3 4

    8765

    Figure 9.6 Tissue location of mononuclear phagocytes.

    V

    NORMAL INJECTED

    L

    SG

    Figure 9.7 Localization of intravenously injected particles in the mononuclear phagocyte system. ( Right ) A mouse was injected with fine carbon particles and killed 5 min later. Carbon accumulates in organs rich in mononuclear phagocytes: lungs (L), liver (V), spleen (S) and areas of the gut wall (G). ( Left ) Normal organ colour shown in an uninjected control mouse. (Courtesy of P.M. Lydyard.)

    LM

    E

    PN

    Figure 9.8 Monocyte ( × 8000), with ‘horseshoe’ nucleus (N). Phagocytic and pinocytic vesicles (P), lysosomal granules (L), mitochondria (M) and isolated profiles of rough-surfaced endoplasmic reticulum (E) are evident. (Courtesy of B. Nichols; © Rockefeller University Press.)

  • 71

    SECTION TWO The adversaries – host defences

    Azurophil granules Specific granules

    0.5 µ m 0.2 µ m1500/cell 3000/cell

    Lysozyme Lysozyme

    Myeloperoxidase Cytochrome b 558

    Elastase Alkaline phosphatase

    Cathepsin G Lactoferrin

    Acid hydrolases

    Defensins Vitamin B12 binding protein

    BPI (bactericidal permeability increasing protein)

    Phagocytosis and killing Phagocytes recognize pathogen-associated molecular patterns (PAMPs) The first event in the uptake and digestion of a microorgan-

    ism by the professional phagocyte involves the attachment

    of the microbe to the surface of the cell through the recog-

    nition of repeating pathogen-associated molecular patterns

    (PAMPs) on the microbe by pattern recognition receptors

    (PRRs) on the phagocyte surface ( Fig. 9.10 ). A major sub-

    set of these PRRs belongs to the class of so-called ‘Toll-

    like receptors’ (TLRs) because of their similarity to the Toll

    receptor in the fruit fly, Drosophila , which, in the adult, trig-

    gers an intracellular cascade generating the expression of

    antimicrobial peptides in response to microbial infection.

    A series of cell surface TLRs acting as sensors for extra-

    cellular infections have been identified ( Fig. 9.11 ) which

    are activated by microbial elements such as peptidogly-

    can, lipoproteins, mycobacterial lipoarabinomannan, yeast

    zymosan and flagellin. Other PRRs displayed by phago-

    cytes include the cell bound ‘C-type (calcium-dependent)

    lectins’, of which the macrophage mannose receptor is an

    example, and ‘scavenger receptors’, which recognize a vari-

    ety of anionic polymers and acetylated low density proteins.

    Examples of intracellular PAMPs are the unmethylated

    guanosine-cytosine (CpG) sequences of bacterial DNA and

    double-stranded RNA from RNA viruses.

    The phagocyte is activated through PAMP recognition The attached microbe may then signal through the phago-

    cyte receptors to initiate the ingestion phase by activating

    an actin-myosin contractile system, which sends arms of

    cytoplasm around the particle until it is completely enclosed

    within a vacuole (phagosome; Fig. 9.12 ; see Fig. 9.10 ). Shortly

    afterwards, the cytoplasmic granules fuse with a phago-

    some and discharge their contents around the incarcerated

    microorganism.

    The internalized microbe is the target for a fearsome array of killing mechanisms As phagocytosis is initiated, the attached microbes also signal

    through one of the PRRs to engineer an appropriate defensive

    response to the different types of infection through a number of

    NF κ B-mediated responses. This activation of a unique plasmamembrane reduced nicotinamide adenine dinucleotide phos-

    phate (NADPH) oxidase reduces oxygen to a series of power-

    ful microbicidal agents, namely superoxide anion, hydrogen

    Figure 9.9 Neutrophil. The multi-lobed nucleus and primary azurophilic, secondary specific and tertiary lysosomal granules are well displayed. In some granules there is an overlap in the contents between azurophilic and secondary granules. Typical conventional lysosomes with acid hydrolase are also seen. (Courtesy of D. McLaren.)

    attachment by patternrecognition receptors

    pseudopodia forminga phagosome

    granule fusionand killing

    release ofmicrobial products

    phagosomeforming

    granule damage anddigestion

    phagocyte

    receptor

    PAMP

    phagolysosome

    complete formationof the phagolysosome

    A B

    C D

    Figure 9.10 Phagocytosis. (A) Phagocytes attach to microorganisms (blue icon) via their cell surface receptors which recognize pathogen-associated molecular patterns (PAMPs) such as lipopolysaccharide. (B) If the membrane now becomes activated by the attached infectious agent, the pathogen is taken into a phagosome by pseudopodia, which extend around it. (C) Once inside the cell, the various granules fuse with the phagosome to form a phagolysosome. (D) The infectious agent is then killed by a battery of microbicidal degradation mechanisms, and the microbial products are released.

  • 72

    CHAPTERCHAPTER9 The innate defences of the body

    peroxide, singlet oxygen and hydroxyl radicals ( Box 9.2 ;

    see also Ch. 14 ). Subsequently, the peroxide, in association with

    myeloperoxidase, generates a potent halogenating system from

    halide ions, which is capable of killing both bacteria and viruses.

    As superoxide anion is formed, the enzyme superoxide

    dismutase acts to convert it to molecular oxygen and

    hydrogen peroxide, but in the process consumes hydrogen

    ions. Therefore initially there is a small increase in pH,

    which facilitates the antibacterial function of the families

    of cationic proteins derived from the phagocytic granules.

    These molecules damage microbial membranes by the

    proteolytic action of cathepsin G and by direct adherence

    to the microbial surface. The defensins have an amphipa-

    thic structure which allows them to insert into microbial

    membranes to form destabilizing voltage-regulated ion

    channels. These antibiotic peptides reach extraordinarily

    high concentrations within the phagosome and act as dis-

    infectants against a wide spectrum of bacteria, fungi and

    enveloped viruses. Other important factors are:

    • lactoferrin, which complexes iron to deprive bacteria of

    essential growth elements

    • lysozyme, which splits the proteoglycan cell wall of

    bacteria

    • nitric oxide, which can lead not only to iron seclusion

    but, together with its derivative, the peroxynitrite

    radical, can also be directly microbicidal.

    The pH now falls so that the dead or dying microorganisms

    are extensively degraded by acid hydrolytic enzymes, and

    the degradation products released to the exterior.

    NF κ B activation can also lead to the release of proinflam-matory mediators. These include the antiviral interferons,

    PAMPsPRRs TLR1 TLR2 TLR4 TLR5 TLR6 TLR10 TLR11

    BacterialLipopeptidesLipoproteins Bacterial LPS

    Bacterialflagellin

    TLR3

    Viral dsRNA

    TLR7

    Viral ssRNANucleotide analogues

    TLR8 TLR9

    Bacterialunmethylated

    CpG DNA

    BacterialLipopeptidesLipoproteins Unknown

    Plasmamembrane

    Endosome

    Toxoplasma gondiiProfilin

    NF!B IRF-5

    Transcription

    Chromosome

    IRF-3

    IRF-7 NFκB IRF-5

    Figure 9.11 Recognition of PAMPs by a subset of pattern recognition receptors (PRRs) termed Toll-like receptors (TLRs). TLRs reside within plasma membrane or endosomal membrane compartments, as shown. All TLRs have multiple N-terminal leucine-rich repeats forming a horseshoe-shaped structure which acts as the PAMP-binding domain. Upon engagement of the TLR ectodomain with an appropriate PAMP (some examples are shown), signals are propagated into the cell that activate the nuclear factor kB (NFkB) and/or interferon regulated factor (IRF) transcription factors, as shown. NFkB and IRF transcription factors then direct the expression of numerous antimicrobial gene products such as cytokines and chemokines, as well as proteins that are involved in altering the activation state of the cell.

    Lt

    Lt

    Lt

    Lt

    Lt

    Figure 9.12 Electron micrographic study of phagocytosis. These two micrographs show human phagocytes engulfing latex particles (Lt). (A) × 3000; (B) × 4500. (Courtesy of C.H.W. Horne.)

  • 73

    SECTION TWO The adversaries – host defences

    the small protein cytokines interleukin-1 β (IL-1 β ), IL-6, IL-12and TNF (TNF α ), which activate other cells through bindingto specific receptors, and chemokines such as IL-8, which rep-

    resent a subset of chemoattractant cytokines.

    Microbial nucleotide breakdown products of infectious

    agents that have succeeded in gaining access to the interior

    of a cell can be recognized by the so-called NOD proteins

    and the typical CpG DNA motif which binds to the endo-

    somal TLR9. Other endosomal Toll-like receptors, TLR3 and

    TLR7/8, are responsive to intracellular viral RNA sequences

    and engender production of antiviral interferon.

    Phagocytes are mobilized and targeted onto the microorganism by chemotaxis Phagocytosis cannot occur unless the bacterium first

    attaches to the surface of the phagocyte, and clearly this

    cannot happen unless both have become physically close

    to each other. There is therefore a need for a mechanism

    that mobilizes phagocytes from afar and targets them

    onto the bacterium. Many bacteria produce chemical

    substances, such as formyl methionyl peptides, which

    directionally attract leukocytes, a process known as ‘che-

    motaxis’. However, this is a relatively weak signalling

    system, and evolution has provided the body with a far

    more effective ‘magnet’ that uses a complex series of pro-

    teins collectively termed ‘complement’.

    Activation of the complement system Complement resembles blood clotting, fibrinolysis and kinin

    formation in being a major triggered enzyme cascade sys-

    tem. Such systems are characterized by their ability to pro-

    duce a rapid, highly amplified response to a trigger stimulus

    mediated by a cascade phenomenon in which the product

    of one reaction is the enzymic catalyst of the next. The most

    abundant and most central component is C3 (complement

    components are designated by the letter ‘C’ followed by a

    number), and the cleavage of this molecule is at the heart of

    all complement-mediated phenomena.

    Oxygen-independent antimicrobial mechanisms

    Cathepsin G and elastase

    Damage to microbial membranesLow molecular weight defensins High molecular weight cationic proteins Bactericidal permeability-increasing protein Lactoferrin Complex with iron Lysozyme Splits proteoglycanAcid hydrolases Degrade dead microbes

    Oxygen-dependent antimicrobial mechanisms

    Reaction Sequence Generated by NADPH Oxidase:

    Glucose + NADP + pentose phosphate + NADPH

    } O 2 burst plus generation of superoxide anionNADPH + O 2 −++NADP 2O − ++22O 2H +

    12 2 2H O O Spontaneous formation

    of further microbial agents

    H 2 O 2 + CI − Myeloperoxidase generation

    of microbicidal moleculesOCI − + H 2 O 2

    O 2 + H 2 O 2 Protective mechanisms used by host and many microbes

    2H 2 O 2 2H 2 O + O 2

    Nitric Oxide Reaction Sequence

    O 2 + L –arginine NO· Reactive species −+⋅ ⋅ 2 NO O ·ONOO −

    NO· + Fe/RSH Fe(RS) 2 (NO) 2 Complexes iron

    Microbicidal species in bold letters. Fe/RSH, a complex of iron with a general sulfhydryl molecule; Fe (RS) 2 , oxidized Fe/RSH; 2O− ,

    superoxide anion; 1 O 2 , singlet (activated) oxygen; OH, hydroxyl free radical; NADPH, reduced nicotinamide adenine dinucleotide phosphate; NADP + , oxidized NADPH; H 2 O 2 , hydrogen peroxide; OCI

    – , hypochlorite anion; NO; nitric oxide; ONOO – , peroxynitrite radical.

    }

    Hexose monophosphate shunt

    NADPH oxidase>

    Spontaneous dismutation > }

    − +2 2 2O H O > �−+ +1 2OH OOH

    Myeloperoxidase>

    − + 2H OOCI }> −+ +1 2O 2CI H O

    22O 2H− ++ Superoxide dismutase

    > }Catalase >NO synthase

    > }>>

    Box 9.2 Antimicrobial Mechanisms in Phagocytic Vacuoles

  • 74

    CHAPTERCHAPTER9 The innate defences of the body In normal plasma, C3 undergoes spontaneous activation

    at a very slow rate to generate the split product C3b. This is

    able to complex with another complement component, fac-

    tor B, which is then acted upon by a normal plasma enzyme,

    factor D, to produce the C3-splitting enzyme C3bBb. ThisC3 convertase can then split new molecules of C3 to give C3a

    (a small fragment) and further C3b. This represents a posi-

    tive feedback circuit with potential for runaway amplifica-

    tion; however, the overall process is restricted to a tick-over

    level by powerful regulatory mechanisms, which break the

    unstable soluble-phase C3 convertase into inactive cleavage

    products ( Fig. 9.13 ).

    In the presence of certain molecules, such as the carbohy-

    drates on the surface of many bacteria, the C3 convertase can

    become attached and stabilized against breakdown. Under

    these circumstances, there is active generation of new C3

    convertase molecules, and what is known as the ‘alternative’

    complement pathway can swing into full tempo (see Ch. 10 ).

    Complement synergizes with phagocytic cells to produce an acute inflammatory response Activation of the alternative complement pathway with

    the consequent splitting of very large numbers of C3 mole-

    cules has important consequences for the orchestration of an

    integrated antimicrobial defense strategy ( Fig. 9.14 ). Large

    numbers of C3b produced in the immediate vicinity of the

    microbial membrane bind covalently to that surface and act

    as opsonins (molecules that make the particle they coat more

    susceptible to engulfment by phagocytic cells; see below).

    This C3b, together with the C3 convertase, acts on the next

    component in the sequence, C5, to produce a small fragment,

    C5a which, together with C3a, has a direct effect on mast

    cells to cause their degranulation ( Fig. 9.15 ). This results in

    the release not only of mediators of vascular permeability,

    but also of factors chemotactic for polymorphs ( Table 9.2 ).

    The circulating equivalent of the tissue mast cell, the baso-

    phil, is shown in Figure 9.16 .

    The vascular permeability mediators increase the perme-

    ability of capillaries by modifying the intercellular forces

    between the endothelial cells of the vessel wall. This allows

    the exudation of fluid and plasma components, including

    more complement, to the site of the infection. These media-

    tors ( Table 9.2 ) also up-regulate molecules such as intercel-

    lular adhesion molecule-1 (ICAM-1) and endothelial cell

    leukocyte adhesion molecule-1 (ELAM-1), which bind to

    specific complementary molecules on the polymorphs and

    encourage them to stick to the walls of the capillaries, a pro-

    cess termed ‘margination’.

    The chemotactic factors, on the other hand, provide a

    chemical gradient which attracts marginated polymorpho-

    nuclear leukocytes from their intravascular location, through

    the walls of the blood vessels, and eventually leads them to

    the site of the C3b-coated bacteria that initiated the whole

    activation process. Polymorphs have a well-defined recep-

    tor for C3b on their surface, and as a result, the opsonized

    bacteria adhere very firmly to the surface of these newly

    arrived cells.

    The processes of capillary dilation (erythema), exudation

    of plasma proteins and of fluid (oedema) due to hydro-

    static and osmotic pressure changes, and the accumulation

    of neutrophils are collectively termed the ‘acute inflam-

    matory response’, and result in a highly effective way of

    focusing phagocytic cells onto complement-coated micro-

    bial targets.

    It also seems clear that the macrophage can be stimu-

    lated by certain bacterial toxins such as the lipopolysaccha-

    rides (LPS), by the action of C5a, and by the phagocytosis

    of C3b-coated bacteria, to secrete other potent mediators of

    acute inflammation, independently of the mast cell-directed

    pathway ( Fig. 9.17 ).

    C9 molecules form the ‘membrane attack complex’, which is involved in cell lysis We have already introduced the idea that following the

    activation of C3 the next component to be cleaved is C5;

    the larger C5b fragment that results becomes membrane

    bound. This subsequently binds components C6, C7 and

    C8, which form a complex capable of inducing a criti-

    cal conformational change in the terminal component C9.

    The unfolded C9 molecules become inserted into the lipid

    bilayer and polymerize to form an annular ‘membrane

    attack complex’ (MAC) ( Figs 9.18, 9.19 ). This behaves as a

    transmembrane channel that is fully permeable to electro-

    lytes and water; because of the high internal colloid osmotic

    pressure of cells, there is a net influx of sodium (Na + ) and

    this frequently leads to lysis.

    microbialpolysaccharide

    properdin

    C3 convertasestabilization

    C3bB

    factor BC3b

    C3a

    C3

    C3bBb

    iC3b

    C3dC3c +(inactive products)

    proteases

    factor D

    factors H and I

    C3b regulation

    (C3 convertase)

    feedbackloop

    Figure 9.13 Activation of complement by microorganisms. C3b is formed by the spontaneous breakdown of C3 complexes with factor B to form C3bB which is split by factor D to produce a C3 convertase C3bBb, capable of further cleaving C3. The convertase is heavily regulated by factors H and I but can be stabilized on the surface of microbes and properdin. The horizontal bar indicates an enzymically active complex. iC3b, inactive C3b.

  • 75

    SECTION TWO The adversaries – host defences

    Acute phase proteins Certain proteins in the plasma, collectively termed ‘acute

    phase proteins’, increase in concentration in response to

    early ‘alarm’ mediators such as the cytokines interleukin-1

    (IL-1), IL-6 and tumour necrosis factor (TNF), released as

    a result of infection or tissue injury. Many acute phase

    reactants such as mannose binding lectin and C-reactive

    protein (CRP) increase dramatically during inflamma-

    tion ( Fig. 9.20 ). Like the professional phagocytes, both

    use pattern recognition receptors to bind to molecular

    patterns on the pathogen (PAMPs), to generate defen-

    sive effector functions ( Fig. 9.21 ). Other acute phase reac-

    tants show more moderate rises, usually less than fivefold

    (see Table 9.3 ). In general, these proteins are thought to

    have defensive roles.

    capillary

    C3bBb

    C3b C3

    1

    8

    93

    4

    2

    4

    57

    7

    6

    C5

    C5a/C3a

    vascularpermeability

    mediators

    chemotacticfactors

    exudation

    polymorph

    C3b receptor

    C3b

    bacterium initiation

    MC

    Figure 9.14 The defensive strategy of the acute inflammatory reaction initiated by bacterial activation of the alternative complement pathway. Activation of the C3bBb C3 convertase by the bacterium (1) leads to the generation of C3b (2) (which binds to the bacterium (3)), C3a and C5a (4), which recruit mast cell (MC) mediators. These in turn cause capillary dilation (5), exudation of plasma proteins (6), and chemotactic attraction (7) and adherence of polymorphs to the C3b-coated bacterium (8). Note that C5a itself is also chemotactic. The polymorphs are then activated for phagocytosis and the final kill (9).

    Figure 9.15 Electron micrographs of rat peritoneal mast cells. These show (A) the resting cell with its electron-dense granules ( × 6000) and (B) a granule in the process of exocytosis ( × 30 000). (Courtesy of T.S.C. Orr.)

    A

    B

  • 76

    CHAPTERCHAPTER9 The innate defences of the bodyInfl ammatory mediators

    Mediator Main source Actions

    Histamine Mast cells, basophils Increased vascular permeability, smooth muscle contraction, chemokinesis

    5-hydroxytryptamine (5HT – serotonin)

    Platelets, mast cells (rodent) Increased vascular permeability, smooth muscle contraction

    Platelet activating factor (PAF) Basophils, neutrophils, macrophages Mediator release from platelets, increased vascular permeability, smooth muscle contraction, neutrophil activation

    IL-8 (CXCL8) Mast cells, endothelium, monocytes and lymphocytes

    Polymorph and monocyte localization

    C3a Complement C3 Mast cell degranulation, smooth muscle contraction

    C5a Complement C5 Mast cell degranulation, neutrophil and macrophage chemotaxis, neutrophil activation, smooth muscle contraction, increased capillary permeability

    Bradykinin Kinin system (kininogen) Vasodilation, smooth muscle contraction, increased capillary permeability, pain

    Fibrinopeptides and fibrin breakdown products

    Clotting system Increased vascular permeability, neutrophil and macrophage chemotaxis

    Prostaglandin E 2 (PGE 2 ) Cyclo-oxygenase pathway, mast cells Vasodilation, potentiates increased vascular permeability produced by histamine and bradykinin

    Leukotriene B 4 (LTB 4 ) Lipoxygenase pathway, mast cells Neutrophil chemotaxis, synergizes with PGE 2 in increasing vascular permeability

    Leukotriene D 4 (LTD 4 ) Lipoxygenase pathway Smooth muscle contraction, increasing vascular permeability

    Table 9.2 The major inflammatory mediators that control blood supply and vascular permeability or modulate cell movement

    Other mediators are generated from the coagulation process. Chemotaxis refers to directed migration of granulocytes up the concentration gradient of the mediator, whereas chemokinesis describes randomly increased motility of these cells. (Reproduced from Male D, Brostoff J, Roth DB, Roitt I. Immunology , 7th edition, 2006. Mosby Elsevier, with permission.)

    Figure 9.16 Morphology of the basophil. (A) This blood smear shows a typical basophil with its deep violet-blue granules. Wright's stain ( × 1500). (B) Electron micrograph showing the ultrastructure of the basophil. Basophils in guinea pig skin showing the nuclei (N) and characteristic randomly distributed granules (G) ( × 6000). (Courtesy of D. McLaren.)

    A

    N

    G

    B

  • 77

    SECTION TWO The adversaries – host defences

    Other extracellular antimicrobial factors There are many microbicidal agents that operate at short range

    within phagocytic cells, but also appear in various body fluids

    in sufficient concentration to have direct inhibitory effects on

    infectious agents. For example, lysozyme is present in fluids

    such as tears and saliva in amounts capable of acting against

    the proteoglycan wall of susceptible bacteria. Similarly, lacto-

    ferrin may appear in the blood in sufficient concentration to

    complex iron and deprive bacteria of this important growth

    factor. Whether agents that normally act over a short range,

    such as reactive oxygen metabolites or TNF (a cytotoxic mol-

    ecule produced by macrophages and other cell types), can

    reach concentrations in the body fluids that are adequate to

    allow them to act at a distance from the cell producing them

    will be discussed in Chapter 14 , particularly when consider-

    ing the mechanisms by which the blood-borne forms of para-

    sites such as malaria are attacked.

    Interferons are a family of broad spectrum antiviral molecules Interferons (IFNs) are widespread throughout the animal

    kingdom and are again discussed further in Chapter 14 .

    They were first recognized by the phenomenon of viral inter-

    ference, in which a cell infected with one virus is found to

    be resistant to superinfection by a second unrelated virus.

    Leukocytes produce many different α -interferons (IFN α ),

    Acute phase reactant Function

    Dramatic increases in concentration

    C-reactive protein Fixes complement, opsonizes

    Mannose binding lectin Fixes complement, opsonizes

    α 1 acid glycoprotein Transports proteinSerum amyloid A protein

    Complexes chondroitin sulphate

    Moderate increases in concentration

    α 1 proteinase inhibitors Inhibit bacterial proteases α 1 anti-chymotrypsin Inhibits bacterial proteasesSurfactant protein A Binds influenza virus

    haemagglutinin

    C3, C9, factor B Increase complement function

    Ceruloplasmin O 2 scavenger

    Fibrinogen Coagulation

    Angiotensin Blood pressure

    Haptoglobin Binds haemoglobin

    Fibronectin Cell attachment

    Table 9.3 Acute phase proteins produced in response to infection in the human

    ICAM-1

    bacterial toxin C5a (?C3a)

    microbe

    PGE2 , LTB4 IL-1, TNFα NAP-2

    induce adhesion molecules

    ELAM-1

    PMNactivation andchemotaxis

    neutrophil

    NAP-1

    C3b

    basementmembrane

    endothelial cell

    IL-8

    increasevascular

    permeability

    Figure 9.17 A role for the macrophage (Mø) in the initiation of acute inflammation. Stimulation induces macrophage secretion of mediators. Blood neutrophils stick to the adhesion molecules on the endothelial cell and use them to provide traction as they force their way between the cells, through the basement membrane (with the help of secreted elastase) and up the chemotactic gradient. During this process they become progressively activated by neutrophil activating peptide-2 (NAP-2). PGE 2 , prostaglandin E 2 ; LTB 4 , leukotriene B 4 ; IL-1, interleukin-1; PMN, polymorphonuclear neutrophil; TNF α , tumour necrosis factor alpha; ELAM-1, endothelial cell leukocyte adhesion molecule-1; ICAM-1, intercellular adhesion molecule-1.

  • 78

    CHAPTERCHAPTER9 The innate defences of the body

    while fibroblasts and probably all cell types synthesize IFN β . A third type (IFN γ ) is not a component of the innate immunesystem and will be discussed in Chapter 10 as a member of

    the important cytokine family.

    When cells are infected by a virus, they synthesize and

    secrete IFNs α and β , which bind to specific receptors onnearby uninfected cells. The bound IFN exerts its antivi-

    ral effect by facilitating the synthesis of two new enzymes,

    which interfere with the machinery used by the virus for its

    own replication. The mechanism of action of IFN is discussed

    more fully in Chapter 14; the net result is to set up a cordon

    of infection-resistant cells around the site of virus infection, so

    restraining its spread ( Fig. 9.22 ). IFN is highly effective in vivo,

    as supported by experiments in which mice injected with an

    antiserum to murine IFN were found to be killed by several

    hundred times less virus than was needed to kill the controls.

    It should be emphasized, however, that IFN seems to play a

    significant role in recovery from, rather than prevention of,

    viral infections.

    Extracellular killing Natural killer cells attach to virally infected cells, allowing them to be differentiated from normal cells There is a widely held view that viruses represent fragments

    of the genome of multicellular organisms that have achieved

    the ability to exist in an extracellular state. The small num-

    ber of genes present in the viral genome, however, does not

    C5b

    C6

    C7 C8

    C5b678

    C9C9

    membraneattack

    complex

    C5aC3bBb

    C6 C7 C8

    solutes

    C9 C9

    C5b678

    C3b C5b C3b C5b C3bC5b

    etc.

    membrane

    MAC

    etc.

    1.2. 3. 4.

    Figure 9.18 Assembly of the C5b-9 membrane attack complex (MAC). (1) Recruitment of a further C3b into the C3bBb enzymic complex generates a C5 convertase which cleaves C5a from C5 and leaves the remaining C5b attached to the membrane. (2) Once C5b is membrane bound, C6 and C7 attach themselves to form the stable complex C5b67, which interacts with C8 to yield C5b678. (3) This unit has some effect in disrupting the membrane, but primarily causes the polymerization of C9 to form tubules traversing the membrane. The resulting tubule is referred to as a MAC. (4) Disruption of the membrane by this structure permits the free exchange of solutes, which are primarily responsible for cell lysis.

    Figure 9.19 Electron micrograph of the MAC. The funnel-shaped lesion ( arrowed ) is due to a human C5b–9 complex that has been reincorporated into lecithin liposomal membranes ( × 234 000). (Courtesy of J. Tranum-Jensen and S. Bhakdi.)

    2 4 6 8 10 12 140

    10

    100

    1

    days

    acute phaseprotein titre

    (arbitrary units)

    infection disease recovery

    C-reactive protein (CRP)

    complement

    opsonization

    Ca2+

    Figure 9.20 Acute phase proteins, here exemplified by C-reactive protein (CRP), are serum proteins that increase rapidly in concentration (sometimes up to 100-fold) following infection (graph). They are important in innate immunity to infection. CRP recognizes and binds in a calcium (Ca 2 + )-dependent fashion to molecular groups found on a wide variety of bacteria and fungi. In particular, it uses its pattern recognition to bind the phosphocholine moiety of pneumococci. The CRP acts as an opsonin and activates complement with all the associated sequelae. Mannose binding protein reacts not only with mannose but several other sugars, enabling it to bind to a wide variety of Gram-negative and -positive bacteria, yeasts, viruses and parasites, subsequently activating the complement system and phagocytic cells. The structurally related ficolins typically recognize PAMPs containing N -acetylglucosamine and can also activate the lectin complement pathway.

  • 79

    SECTION TWO The adversaries – host defences

    include those required for viral replication. Accordingly, it is

    essential for viruses to penetrate the cells of an infected host

    in order to subvert the cells' replicative machinery towards

    viral replication. Clearly, it is in the interests of the host to try

    to kill such infected cells before the virus has had a chance

    to reproduce. Natural killer (NK) cells are cytotoxic cells that

    appear to have evolved to carry out just such a task. These

    are large granular lymphocytes (LGLs) ( Fig. 9.23 ) that rec-

    ognize virus-infected or stressed cells and allow them to be

    differentiated from normal cells; this clever discrimination

    is mediated by activating receptors on the NK cells such as

    NKG2D that recognize ligands on the infected cell that are

    related to MHC Class I molecules, and inhibitory receptors

    which bind to MHC Class I molecules on normal cells,

    generating signals that counteract those from the activating

    receptors. Activation of the NK cell results in the extracel-

    lular release of its granule contents into the space between

    the target and effector cells. These contents include perfo-

    rin molecules, which resemble C9 in many respects, espe-

    cially in their ability to insert into the membrane of the target

    cell and polymerize to form annular transmembrane pores,

    like the MAC. This permits the entry of another granule pro-

    tein, granzyme B, which leads to death of the target cell by

    apoptosis (programmed cell death), a process mediated by

    a cascade of proteolytic enzymes termed caspases, which

    terminates with the ultimate fragmentation of DNA by a

    Ca-dependent endonuclease ( Fig. 9.24 ).

    Subsidiary mechanisms that can activate the caspase path-

    way include engagement of Fas on the target cell by the NK

    Fas ligand, and binding of tumour necrosis factor (TNF)

    released from the NK granules to surface receptors. TNF

    was first recognized as a product of activated macrophages

    known to be capable of killing certain other cells, particu-

    larly some tumour cells.

    Yet a further mode of cytotoxicity can be turned on by the

    activated macrophage, involving the direct ‘burning’ of the

    surface of another cell by means of a stream of reactive oxy-

    gen intermediates, produced at the macrophage membrane

    by the respiratory oxygen burst, as discussed previously

    (see Box 9.2 ).

    Eosinophils act against large parasites It takes little imagination to realize that professional

    phagocytes are far too small to be capable of physically

    engulfing large parasites such as helminths. An alterna-

    tive strategy, such as killing by an extracellular broad-

    side of the type discussed above would seem to be a more

    appropriate form of defence. Eosinophils appear to have

    evolved to fulfil this role. These polymorphonuclear rela-

    tives of the neutrophil have distinctive cytoplasmic gran-

    ules, which stain strongly with acidic dyes ( Fig. 9.25 ) and

    have a characteristic ultrastructural appearance. A major

    microbe

    PRR

    transducer

    effectorfunction:

    complementactivation

    phagocytosis

    PAMP

    receptor on solublemolecule or phagocyte

    Figure 9.21 A major defensive strategy in which soluble factors, such as CRP (C reactive protein) and mannose binding protein, and professional phagocytes use their pattern recognition receptors (PRR) to bind to the pathogen-associated molecular patterns (PAMPs) on the microbial surface and signal through their transducer structures to initiate appropriate effector functions.

    virus DNA mRNA

    mRNA

    IFNreceptor

    protein synthesis

    'antiviral' state

    DNA

    IFNsynthesis

    Increased NK activityand expression of

    MHC products

    Figure 9.22 The action of interferon (IFN). Virus infecting a cell induces the production of IFN α / β . This is released and binds to IFN receptors on other cells. The IFN induces the production of antiviral proteins, which are activated if virus enters the second cell, and increased synthesis of surface MHC molecules which enhance susceptibility to cytotoxic T cells (cf. Ch. 10). NK, natural killer; MHC, major histocompatibility complex.

    TC

    NK

    Figure 9.23 Electron micrograph of an NK cell killing a tumour cell (TC). NK cells bind to and kill IgG antibody-coated (see Fig. 10.13 ), and non-coated, tumour cells. It is essential for the membranes of the two cells to be closely apposed in order for the NK cell to deliver the ‘kiss of death’ ( × 4500). (Courtesy of P. Lydyard.)

  • 80

    CHAPTERCHAPTER9 The innate defences of the body

    basic protein (MBP) has been identified in the core of the

    granule, while the matrix has been shown to contain an

    eosinophilic cationic protein, a peroxidase and a perfo-

    rin-like molecule. Eosinophils have surface receptors for

    C3b and when activated generate copious amounts of

    active oxygen metabolites.

    Many helminths can activate the alternative complement

    pathway but, although resistant to C9 attack, their coat-

    ing with C3b allows adherence to the eosinophils through

    their C3b surface receptors. Once activated, the eosinophil

    launches its extracellular ammunition, which includes the

    release of major basic proteins and the cationic protein to

    damage the parasite membrane, with a possibility of a fur-

    ther ‘chemical burn’ from the oxygen metabolites and ‘leaky

    pore’ formation by the perforins.

    G

    B

    A

    Figure 9.25 The eosinophil granulocyte is capable of extracellular killing of parasites (e.g. worms) by releasing its granule contents. (A) Morphology of the eosinophil. This blood smear enriched for granulocytes shows an eosinophil with its multilobed nucleus and heavily stained cytoplasmic granules. Leishman's stain ( × 1800). (Courtesy of P. Lydyard.) (B) Electron micrograph showing the ultrastructure of a guinea pig eosinophil. The mature eosinophil contains granules (G) with central crystalloids ( × 8000). (Courtesy of D. McLaren.)

    NK cell

    target cellnucleus

    nucleus

    granules

    pore

    virally induced structures

    fluid phase perforin

    NK cell receptor

    membrane-bound perforin

    granzyme B

    TNFα

    FasL

    Fas

    death signal

    caspase cascade

    Figure 9.24 Schematic model of lysis of virally infected target cell by a natural killer (NK) cell. As the NK cell receptors bind to the surface of the virally infected cell, and if signals from activation receptors exceed those from the inhibitory receptors that recognize normal MHC Class I molecules, there is exocytosis of granules and release of cytolytic mediators into the intercellular cleft. A calcium (Ca 2 + )-dependent conformational change in the perforin enables it to insert and polymerize within the membrane of the target cell to form a transmembrane pore, which allows entry of granzyme B into the target cell, where it causes programmed cell death (apoptosis). A back-up cytolytic system using engagement of the Fas receptor with its ligand (FasL), can also trigger apoptosis as can binding of granule-derived tumour necrosis factor alpha (TNF α ) to its receptor. Unlike the PRR-mediated activation of phagocytes by intracellular components – so-called danger-associated molecular patterns (DAMPs) – released on necrotic cell-death typically caused by tissue trauma, burns and other non-physiological stimuli, cells undergoing apoptotic death do not activate the immune system because they express surface molecules such as phosphatidyl serine which mark them out for phagocytic removal before they release their intracellular DAMPs.

  • 81

    SECTION TWO The adversaries – host defences

    mast cell

    acute phaseproteins

    phagocytosis

    acute inflammatory response

    microbe mediators

    PMN

    margination

    soluble factors

    lysozymecomplement

    interferon

    transudation

    mobilization

    separation

    endothelium

    Figure 9.26 Mobilization of defensive components of innate immunity. Microbes, either through complement activation or through direct effects on macrophages, release mediators which increase capillary permeability to allow transudation of plasma bactericidal molecules, and chemotactically attract plasma polymorphs from the bloodstream to the infection site. PMN, polymorphonuclear neutrophil.

    The innate system of immune defence consists of a formidable barrier to entry and second-line defence by phagocytes and circulating soluble factors. Colonization of the body by normally non-pathogenic (‘opportunistic’) microorganisms occurs whenever there is a hereditary or acquired deficiency in any of these functions.

    The main phagocytic cells are polymorphonuclear neutrophils and macrophages. They adhere to the surface of the microbe by receptors which recognize pathogen-associated molecular patterns (PAMPs). This activates the engulfment process so that the organisms are taken inside the cell in a phagocytic vacuole which fuses with cytoplasmic granules. A formidable array of oxygen-dependent and oxygen-independent microbicidal mechanisms then comes into play.

    The complement system, a multicomponent triggered enzyme cascade, is used to attract phagocytic cells to the microbes and engulf them.

    The most abundant complement component, C3, is split by a convertase enzyme formed from its own cleavage product C3b and factor B and stabilized against breakdown caused by factors H and I through association with the microbial surface. As it is formed, C3b becomes covalently linked to the microorganism.

    The next most abundant component, C5, is activated to yield a small peptide, C5a, while residual C5b binds to the surface of the microorganism and assembles the terminal components C6–9 into a membrane attack complex (MAC), which is freely permeable to solutes and can lead to osmotic lysis. In addition, C5a is a potent chemotactic agentfor polymorphs and greatly increases capillary permeability.

    C3a and C5a act on mast cells, causing the release of further mediators such as histamine, LTB 4 and TNF α , with effects on capillary permeability and adhesiveness and

    neutrophil chemotaxis. They also activate neutrophils, which bind to the C3b-coated microbes by their surface C3b receptors and then ingest them.

    The influx of polymorphs and increase in vascular permeability constitute the potent antimicrobial acute inflammatory response.

    Inflammation can also be initiated by tissue macrophages, which subserve a similar role to that of the mast cell since signalling by bacterial toxins C5a or by C3b-coated bacteria adhering to surface complement receptors on tissue macrophages causes the release of TNF α , LTB 4 , PGE 2 , the neutrophil chemotactic factor, IL-8, and a neutrophil-activating peptide.

    Other humoral defences include the acute phase proteins such as CRP, and the IFNs, which can block viral replication.

    Virally infected cells can be killed by NK cells, following increased recognition by activation receptors that overcomes inhibitory signals from normal MHC Class I recognition.

    Extracellular killing can also be effected by C3b-bound eosinophils, which may be responsible for the failure of many large parasites to establish a foothold in potential hosts.

    It is probably true to say that engulfment and killing by phagocytic cells is the mechanism used to dispose of the majority of microbes, and the mobilization and activation of these cells by orchestrated responses such as the acute inflammatory response ( Fig. 9.26 ) is a key feature of innate immunity. However, not every organism is readily susceptible to phagocytosis or even to killing by complement or lysozyme, and this brings us to the role of the adaptive immune response, which is explored in Chapter 10.

    KEY FACTS