You are on page 1of 13

Anaesthesia, 2000, 55, pages 10811093

................................................................................................................................................................................................................................................

R E V I E W A RT I C L E

Apoptosis: mechanisms and clinical implications


P. C. A. Kam1 and N. I. Ferch2
1 Clinical Associate Professor and Senior Staff Anaesthetist, Department of Anaesthesia and Pain Management,
University of Sydney, Royal North Shore Hospital, St Leonards, NSW 2065, Australia
2 Anaesthetic Registrar, Department of Anaesthetics, University of Sydney, Royal Prince Alfred Hospital, Camperdown,
NSW 2050, Australia
Summary

The balance between cell survival and death is under tight genetic control. A multiplicity of
extracellular signals and intracellular mediators is involved in maintaining this balance. When the cell
is exposed to physical, biochemical or biological injury, or deprived of necessary substances, it
activates a series of stress-response genes. With minimal insults, the cell may recover. With greater
insults, single cell death, or apoptosis, results; the cell dies and is recycled to its neighbours. If the
insult overwhelms a large number of cells then necrosis ensues, with an accompanying inflammatory
response. Dysregulation of the controlling mechanisms of this system results in disease. Deficient
apoptosis is associated with cancer, auto-immunity and viral infections. Excessive apoptosis is
associated with ischaemic heart disease, stroke, neurodegenerative disease, sepsis and multiple organ
dysfunction syndrome. There are myriad therapeutic options unfolding as understanding is gained of
apoptosis and its control.
Keywords Apoptosis. Cell death: morphology; mechanisms; clinical implications.
Multiple organ dysfunction syndrome.
.................................................................................................
Correspondence to: Dr P. C. A. Kam
Accepted: 4 April 2000

Cell death, a tightly controlled, finely orchestrated event,


may be described either as apoptosis or nonapoptotic cell
death, traditionally called `necrosis' [1]. Apoptosis is a
process of cell suicide, the mechanisms of which are
encoded in the chromosomes of all nucleated cells.
Physiological cell death that removes unwanted cells plays
an important role in development, tissue homeostasis and
defence against viral infection and mutation. Apoptosis is
regulated by complex molecular signalling systems. Tissue
ischaemia and reperfusion activate these molecular
systems, which therefore represent a therapeutic target
for novel treatment to preserve cellular integrity in critical
organs such as the brain and heart. Apoptotic cells
undergo orderly, energy-dependent enzymatic breakdown into characteristic molecular fragments, deoxyribonucleic acid (DNA), lipids and other macromolecules,
which are packaged into small vesicles that may be
phagocytosed and reused. The cells involute and die with
minimal harm to nearby cells [2]. In contrast `necrotic'
q 2000 Blackwell Science Ltd

cell death is characterised by inflammation and widespread damage.


Apoptosis has a central role in the pathogenesis of
human disease when the genes controlling the apoptotic
process are suppressed, overexpressed or altered by
mutation [3]. Disordered apoptosis is implicated in a
variety of human diseases (Table 1). Research into
apoptosis is proceeding at a fast pace and this has led to
the possibility of new therapeutic approaches to some
intractable human diseases [4].
The purpose of this review is to provide a background to
the molecular components that activate apoptosis and the
relevance of these to a variety of human diseases, and to
discuss the potential for novel therapies based on our
understanding of them. An understanding of the role of cell
injury and death in the pathophysiology of organ dysfunction is relevant to anaesthetists and intensivists particularly in
the management of ischaemic and reperfusion injury, and
multiple organ dysfunction syndrome (MODS).
1081

P. C. A. Kam and N. I. Ferch  Apoptosis


Anaesthesia, 2000, 55, pages 10811093
................................................................................................................................................................................................................................................

Table 1 Human diseases associated with


Increased apoptosis
Central nervous system
Myocardium
Lymphocytes
Macrophages
Decreased apoptosis
Epithelial tissues
Blood vessels
Lymphocytes
Haemopoietic system

Degenerative diseases (Alzheimer's and Parkinson's disease)


Cerebral ischaemia
Peri-infarct border zones
Lymphocyte depletion in sepsis and HIV infection
Bacillary dysentery (Shigella dysenteriae)

disordered apoptosis.

Carcinogenesis
Intimal hyperplasia
Autoimmune disorders
Leukaemia, lymphoma

History

In the 1880s Weigert & Cohnheim described the


microscopic appearance of cell death in necrotic tissue
as coagulation necrosis [5]. In 1885, Flemming described
the process of `chromatolysis' in which the nuclei of
mammalian ovarian follicles broke up and ultimately
disappeared in spontaneous cell death [68].
In the early 1970s, Kerr described the electron
microscopic appearance of singe-cell death in the livers
of animals [9, 10] treated with toxins (heliotrine and
albitocin) and ischaemia (by ligation of a large branch of
the portal vein) and called it `shrinkage necrosis' [11].
`Single cell death' in carcinogen-treated rat adrenal cortex
[12] and in tumour cells treated with actinomycin D,
mitomycin C, cytosine arabinose and cycloheximide [13]
was also observed. In 1971, Wyllie found that single cell
death in the adrenal gland was induced by hypophysectomy, and suggested a common pathway for cell death
initiated by hormone regulation and carcinogen-induced
injury [14, 15]. In a landmark paper in 1972, Kerr et al.
described the characteristic sequential changes occurring
in cell structure during the death process in healthy
tissues, normal development, tumour regression, atrophy
and involution [1]. The term `apoptosis' (derived from the
Greek word for `falling off ', a reference to the falling of
leaves from trees in autumn in response to the impending
threat of freezing and damage in winter) was coined [1].
Further momentum was gained in the mid-1980s
when Horvitz reported specific cell death during the

development of the nematode Caenorhabditis elegans and


cloned the C. elegans death genes (ced genes) responsible
[16]. The nematode anti-apoptotic gene ced-9 shared
homology with the mammalian proto-oncogene, Bcl-2
[17], and was shown to preserve B lymphocytes [18, 19].
During the 1990s, the molecular mechanisms that keep
apoptosis in check and the molecular events involved in
disordered apoptosis were unravelled. Rodent fibroblast
tumours expressing human c-myc genes had high rates of
apoptosis [20], suggesting that cancer can occur when a
mutation results in dysfunctional apoptosis [21].
Definitions

Apoptosis specifically refers to an energy-dependent,


asynchronous, genetically controlled process by which
unnecessary or damaged single cells self-destruct when the
apoptosis genes are activated [22, 23]. Briefly, the cell
shrinks and detaches from neighbouring cells and the
nucleus is broken down. The nuclear fragments and
organelles condense and are ultimately packaged in
membrane-bound vesicles, exocytosed and ingested by
surrounding cells. Membrane integrity is preserved and
dyes are only taken up late in the process. The absence of
inflammation differentiates apoptosis from necrosis. The
differences between apoptotic and necrotic cell deaths are
summarised in Table 2.
Ischaemic and accidental (caused by toxins) cell death
are characterised by cell swelling or `oncosis' (from the
Greek word `onkos' meaning swelling), resulting in
Table 2 Differences between apoptosis and

Apoptosis

Necrosis

Physiological or pathological
Asynchronous process in single cells
Genetically controlled
Late loss of membrane integrity
Cell shrinkage
Condensation of nuclear contents
(`ladder' formation of chromatin)
No inflammatory reaction

Always pathological
Occurs synchronously in multiple cells
Caused by overwhelming noxious stimuli
Early loss of membrane integrity
Generalised cell and nucleus swelling
Nuclear chromatin disintegration

1082

necrosis.

Inflammatory reaction

q 2000 Blackwell Science Ltd

Anaesthesia, 2000, 55, pages 10811093


P. C. A. Kam and N. I. Ferch  Apoptosis
................................................................................................................................................................................................................................................

cytoplasmic and nuclear swelling and karyolysis (loss of


affinity for basic dyes). After either oncosis or apoptosis,
cells reach the stage of necrosis where phagocytosis
occurs, and in the case of oncosis, this is accompanied by
inflammation. Apoptosis and oncosis/necrosis are part of
an overlapping spectrum [5, 8, 2426]. Cells exposed to
an overwhelming noxious stimulus, such as ischaemia or
toxins, undergo oncosis and necrosis [27]. Programmed
cell death (PCD) refers to cell suicide occurring during
normal embryological development of an immature
organism and the maturation of tissues or organs [28,
29] and does not require de novo gene expression [30].

Pathophysiology

Morphological features
There are three distinct phases of apoptosis [31]. During
the first phase, the cell detaches from its substratum and
adjacent cells with a loss of microvilli and junctional
complexes or desmosomes [32]. The DNA is digested by
specific endonucleases into fragments and ultimately
packed into vesicles. The changes in DNA include strand
breakage (karyorhexis) and condensation of nuclear
chromatin (pyknosis). This pyknotic chromatin appears
as characteristic crescent-shaped `caps' under light
microscopy. The endoplasmic reticulum swells and
exocytoses its contents. The cell becomes denser as the
cytoplasm shrinks and involutes. In the second phase, the
cell produces pseudopodia (budding) which contain
organelles or nuclear fragments, and these break off into
multiple membrane-bound vesicles. The remaining cell
becomes a round, smooth membrane-bound remnant
(apoptotic body) [3, 4, 32]. In the third phase, the cell
membrane becomes permeable to dyes such as Tryphan
Blue. The apoptotic body and membrane-bound buds
may then be phagocytosed by macrophages, epithelial
cells, vascular endothelium or tumour cells. The entire
process occurs may take only 15 min, and therefore may
be undetectable on tissue sections [5, 23, 33].
In contrast, oncosis is characterised by cellular and
organelle swelling with late nuclear fragmentation and
breakdown by lysosomal enzymes. The swelling is caused

by a deficit in adenosine triphosphate (ATP) production


that leads to membrane ionic pump failure and increased
plasma membrane permeability. This results in bleb
(blister-like, fluid-filled structures) formation of the
plasma membrane and ultimate rupture [5]. There is an
influx of neutrophils and macrophages in the surrounding
tissues, leading to generalised inflammation.
Techniques to identify and quantify apoptosis include
staining with nuclear stains such as Hoechst 33258 that
allow visualisation of nuclear chromatin clumping.
Modern video microscopy allows visualisation of the
temporal sequence of events that occur over 1560 min
to 24 h [23, 34, 35].
Fluorescent microscopy using acridine orange allows
characteristic chromatin clumping to be observed soon
after staining. A further refinement is the comet assay,
which shows up DNA degradation [2]. More accurate
identification of apoptosis is achieved with methods that
specifically target the characteristic DNA cleavages [36].
Agarose gel electrophoresis of extracted DNA fragments
yields a characteristic `ladder' pattern which can be used
as a marker for apoptosis. A lesser extent of DNA
degradation produces hexameric structures called
`rosettes' [4]. Necrotic cells leave a nondescript smear
[3]. The terminal transferase deoxyuridine nick-end
labelling of DNA breaking points (TUNEL) method
[37], which labels the uridine residues of the nuclear
DNA fragments, can be applied to tissue sections and
cell cultures to quantify apoptosis. Flow cytometry assays
[3840] may prove to be most accurate in quantifying
apoptosis.
Mechanism of cell death
Following an appropriate stimulus, the first stage or
`decision phase' of apoptosis is the genetic control point
of cell death. This is followed by the second stage or
`execution' phase, which is responsible for the morphological changes of apoptosis.
There are four main groups of stimuli for apoptosis
[4, 30, 32, 34, 4145]. A wide variety of physiological
and pathological stimuli can initiate apoptosis and these
stimuli are summarised in Table 3. The first group of

Table 3 Stimuli for apoptosis.


DNA (genome) damage
Activation of death receptors
Stimulation of apoptotic pathway
Direct physical cell damage

Ionising radiation
Anti-cancer drugs (e.g. alkylating agents)
Binding of `death receptors' (e.g. Fas receptor, TNF receptor)
Withdrawal of growth factors (e.g. nerve growth factor, IL-3)
Phosphatases, kinase inhibitors
Heat, ultraviolet light, oxygen free radicals, hydrogen peroxide

DNA, deoxyribonucleic acid; TNF, tumour necrosis factor; IL-3, interleukin-3.

q 2000 Blackwell Science Ltd

1083

P. C. A. Kam and N. I. Ferch  Apoptosis


Anaesthesia, 2000, 55, pages 10811093
................................................................................................................................................................................................................................................

stimuli causes DNA damage and include ionising


radiation and alkylating anticancer drugs. The second
group induces apoptosis via receptor mechanisms, either
by receptor activation mediated by glucocorticoids (acting
on the thymus) [34], tumour necrosis factor-a (TNF-a),
or by withdrawal of growth factors (nerve growth factor
and interleukin (IL)-3) [41, 42]. The third group
comprises biochemical agents that enhance the downstream components of the apoptotic pathway and includes
phosphatases [30] and kinase inhibitors (e.g. calphostin C,
staurosporine). The fourth group comprises agents that
cause direct cell membrane damage and includes heat,
ultraviolet light and oxidising agents (superoxide anion,
hydrogen peroxide). Excessive production of reactive
oxygen species (ROS), such as superoxide, hydrogen
peroxide and the hydroxyl radicals, produces free radicals
that damage lipid membranes, proteins, nucleic acids and
extracellular matrix glycosaminoglycans. Many of these
stimuli cause necrosis in larger doses. Injury to cell
membranes induces apoptosis by activating acid sphingomyelinase that generates the second messenger ceramide
from membrane lipids [43, 44].
The signal that initiates apoptosis may result from
binding of a cell-surface `death' receptor or from damage
to the genome. Death receptors that initiate apoptosis
include the Fas receptor and the TNF receptor system
[41]. The Fas receptor, initially known as CD95 or
APO-1, is a transmembrane glycoprotein death receptor
that is activated by binding of Fas ligand (Fas-L) to cell
membranes [41]. Intracellular molecules known as Fasassociated death domain (FADD) are produced. Fas
receptors are found in epithelial tissues, tumours and
haemopoietic tissues, and may be induced in other tissues
that do not express them. The Fas pathway is important in
controlling the immune response. Cytotoxic T lymphocytes expressing Fas ligands activate cells bearing Fas
receptors and induce apoptosis.
The TNF receptor system mediates different biochemical pathways [41, 42]. A TNF-related apoptosis-inducing
ligand (TRAIL) has been discovered. Cancer cells are
susceptible to TRAIL-induced apoptosis. Following
binding of the TNF receptor, intracellular molecules
called `death domains' are produced. A TNF receptor
associated death domain (TRADD) has been identified.
Tumour necrosis factor may suppress apoptosis by binding
to the receptor, TNFR2, which activates a protein known
as Nuclear Factor kB (NF-kB), classed as an inhibitor of
apoptosis protein (IAP) that prevents the execution phase
of apoptosis [43]. It is a DNA binding protein that
regulates many pro-inflammatory genes for the production of cytokines and other pro-inflammatory molecules.
There is increasing evidence that NF-kB is important in
the pathogenesis of systemic inflammatory response
1084

syndrome (SIRS), MODS and acute respiratory distress


syndrome (ARDS) [28].
Decision phase (genetic control)
Apoptosis is controlled genetically and two genes, Bcl-2
and p53 are important. The first, Bcl-2, is a family of genes
that regulates apoptosis [4649]; found on the mitochondrial membrane, endoplasmic reticulum it may
control calcium channels. It is now recognised that
there is a family of mammalian proteins similar to Bcl-2
that promotes or inhibits apoptosis [50, 51]. Proteins such
as Bcl-2 and Bcl-xL prevent apoptosis, whereas Bcl-2
associated x proteins (Bax) such as Bax, Bad, Bak and
Bcl-xS promote apoptosis [30, 52, 53].
The gene p53 is a 53-kDa nuclear phospho-protein
that binds to DNA to act as a transcription factor, and
controls cell proliferation and DNA repair [54]. Mutations
of p53 have been found in . 50% of human cancers (e.g.
colon carcinoma) and are associated with resistance to
treatment [55]. The gene c-myc is a proto-oncogene that
encodes a sequence-specific DNA-binding protein that
acts as a transcription factor and induces apoptosis in the
presence of p53. The c-myc protein is elevated in many
tumours [4].
Mutation of the gene for neuronal apoptosis inhibitory
protein (NAIP) occurs in people with spinal muscular
atrophy [56]. NAIP protects various cells from apoptosis
caused by TNF-a, free radicals and growth factor
deprivation.
Execution phase
The central events in apoptosis are proteolysis and
mitochondrial inactivation. Cellular disruption results
from activation of a family of cysteine proteases called
caspases (CASP) [30, 32, 43, 5759]. Caspases are
proenzymes that have been conserved from nematodes
to humans. Ten human caspases (CASP 110) have been
described. Early molecular studies on apoptosis focused
on the nematode C. elegans and the gene required for the
execution of apoptosis, called ced-3, has been identified
and coded. There are two subfamilies of caspases, the
ced-3 subfamily (produced by ced-3 gene) and the ICE
(IL-1b-converting enzyme) subfamily [60, 61]. Caspase 1,
which is related to ICE, is mainly involved in inflammation [60, 61]. The ced-3 caspases are important effectors
of apoptosis. Caspase 8 or FADD-like interleukin converting enzyme (FLICE) is the most important enzyme of
the ced-3 subfamily [61, 62]. The actions of the caspases
are varied; some are endonucleases that cleave DNA,
some cleave cytoskeletal proteins and others cause a loss of
cell adhesion.
The integrity of the plasma membrane of the apoptotic
cell is maintained initially, although `budding' of the cell
q 2000 Blackwell Science Ltd

Anaesthesia, 2000, 55, pages 10811093


P. C. A. Kam and N. I. Ferch  Apoptosis
................................................................................................................................................................................................................................................

membrane can occur later. There is no leakage of


lysosomal enzymes that can damage nearby cells or elicit
immune responses [63]. The apoptotic cell expresses
membrane signals that induce phagocytosis [64]. Macrophages can recognise neutrophils undergoing apoptosis
via complexes involving thrombospondin receptors
(CD36) and the avb3 integrin [6567].
Clinical relevance of apoptosis

Central nervous system


In the developing organism there is a specialised form of
cell death previously called apoptosis but now known as
`programmed cell death' (PCD) [30]. During embryonic
development of the nervous system a surplus of cells is
produced. PCD eliminates those neurons whose axons fail
to reach the target. It occurs with the withdrawal of
trophic substances, such as nerve growth factor, or with a
loss of synaptic contact or afferent input. Cytokines (e.g.
TNF-a) and ROS may trigger PCD [68].
Oxidative stress, glutamate excitotoxicity and calcium
influx can induce apoptosis in the mature central nervous
system. Excessive production of ROS causes `oxidative
stress', damaging lipid membranes, proteins, nucleic acids
and extracellular matrix glycosaminoglycans. At low
levels of ROS or depletion of antioxidants (e.g. superoxide dismutase, catalase, glutathione peroxidase) apoptosis occurs. At high levels ROS produce more damage
and cause necrosis [30].
Glutamate receptor-mediated neuronal injury is an
important cause of `excitotoxic` neuronal death following
ischaemia, trauma, epileptic seizures or neurodegeneration [30]. Glutamate produces either necrosis associated
with influx of Na1, Cl2 and water leading to cell swelling
or delayed neuronal death (DND) which appears to be
apoptotic, occurring several hours after exposure and
associated with calcium influx via channels linked to
glutamate receptors [6971].
Calcium is an important second messenger, instrumental in inducing apoptosis by stimulating neurotransmitter release, gene induction and the activation of
enzymes (proteases, phosphatases, protein kinases, endonucleases, phospholipases and nitric oxide synthase) [72].
Phospholipase A2 produces superoxide anion, and nitric
oxide synthase produces nitric oxide, both of which can
cause oxidative stress leading to apoptosis.
Increased apoptosis caused by excessive intracellular
Ca21 has been implicated in cerebral ischaemia, traumatic
brain injury, epilepsy and neurodegenerative disease.
Cerebral ischaemia results in both necrosis and DND.
Cells undergoing DND as a result of less severe insult are
seen on the periphery of the infarct within the penumbra.
There is accumulating evidence that apoptosis plays a role
q 2000 Blackwell Science Ltd

in DND [30]. Gene transfer using herpes simplex viruses


containing a Bcl-2 vector may offer neuroprotection
against ischaemia [73]. In traumatic brain injury apoptosis
occurs in < 10% of dying neurons, peaking 2448 h after
injury [74]. With severe trauma, the proportion of
necrotic cell death increases and antiapoptotic agents
may minimise cell death following trauma [73].
Enhanced apoptosis is implicated in several neurodegenerative diseases. In Alzheimer's disease accumulation
of b-amyloid peptide in plaques in the brain and cerebral
vasculature causes apoptosis of cortical neurones [75],
possibly via nitric oxide (NO) [76]. Parkinson's disease is
associated with the loss of dopaminergic neurones in the
substantia nigra, and dopamine is thought to cause
apoptosis in exposed neurons [30]. Some forms of familial
amyotrophic lateral sclerosis may be caused by a mutant
gene coding for the antioxidant superoxide dismutase
resulting in motor neuron loss via apoptosis. Future
therapy may involve the use of growth factors or
inhibitors of macromolecular synthesis to block apoptosis
[56].
Defective apoptosis caused by mutant apoptotic genes
may contribute to the development of neural tumours.
Mutated p53 is seen in astrocytic tumours [77, 78].
Protein kinase C inhibitors (e.g. hypericin and calphostin)
can cause apoptosis in glioma cell lines. Gene transfer
techniques have also been used to introduce Bcl-xs into
neuroblastoma cells and ICE retrovirus into gliosarcoma
cells with induction of apoptosis in these tumours
[79, 80].
Cardiovascular system
Recently it has been shown that myocyte death with
cardiac disease occurs by both apoptosis and necrosis in
response to hypoxia and ischaemia [45]. Apoptosis has
been documented in myocardial ischaemia, reperfusion
and infarction, heart failure induced by rapid ventricular
pacing or coronary microemboli, several models of
pressure overload hypertrophy (by passive load on papillary muscle, aortic banding) [42] and ageing [53].
Hypoxia, nutritional deficiency and toxins (e.g.
chemotherapy) that can cause necrosis may induce
apoptosis at lower doses [45, 52]. Infarcted areas have a
central area of necrosis with surrounding apoptosis in the
peri-infarct border zone [42]. Autopsy studies of
myocardial infarction have found apoptotic cells scattered
between the necrotic cells centrally and the normal cells
around the periphery. Coronary artery ligation induces
marked increases in proto-oncogenes (Bcl-2 and Fas) in
myocytes. The initial event during the early hours of
ischaemia appears to be apoptosis. As myocardial cells are
overwhelmed by severe ischaemia, apoptotic genes can no
longer be expressed and necrosis supervenes [81]. In in vivo
1085

P. C. A. Kam and N. I. Ferch  Apoptosis


Anaesthesia, 2000, 55, pages 10811093
................................................................................................................................................................................................................................................

reperfusion studies in rabbit models, apoptosis was seen


predominantly in reperfused hearts, whereas necrosis was
seen mainly with persistent occlusion [82]. Reperfusion
increases free radical production and intracellular calcium,
which are potent inducers of apoptosis.
Apoptosis in vascular smooth muscle cells in atherosclerotic plaques in coronary arteries may be involved in
plaque destabilisation and rupture [83]. Apoptosis is
increased by 43% in primary atherosclerotic lesions and
93% in restenoses [84, 85]. It may be triggered by NO
and is associated with the expression of caspase 1 [84].
Arrhythmias may be associated with deficient apoptosis
during cardiac development. Normally after birth there is
apoptosis of the small round pacemaker cells in the central
and lower part of the AV node. Delay or failure of
apoptosis in these cells may lead to life-threatening
arrhythmias that may resolve spontaneously. Excessive
apoptosis can lead to bradyarrhythmias and sudden death
[45]. Patients with long QT syndrome have increased
rates of apoptosis in the sinus node [85].
A common end-stage pathway of various cardiovascular
diseases results in deteriorating function and cardiac
failure associated with dilated cardiomyopathy [45, 46].
Myocardial hypertrophy may be an initial compensatory
response to overloading of adult myocytes. This is
associated with c-myc, c-fos, and transforming growth
factor, which are also involved in initiating apoptosis.
Dilated cardiomyopathy can be induced by chronic
infusion of TNF-a, which causes apoptosis [43]. A
progressive decline in function in end-stage cardiac failure
may be due to apoptosis and this is supported by the
finding of increased levels of Bcl-2 in the myocytes of
patients with cardiac failure. Although apoptosis itself is
irreversible, minimising it with growth factors and cytokines may prevent the progressive decline in left
ventricular function [45].
Arrhythmogenic right ventricular dysplasia is a cardiomyopathy associated with sudden death. It is postulated
that repetitive episodes of ischaemia and reperfusion
associated with ventricular arrhythmias produce apoptosis, which is found in myocardial biopsies in these
patients [45].
Immune system
Dysfunction of the apoptotic pathway causes autoimmune
disease, immunodeficiencies and lymphoid malignancies.
During development large numbers of precursor cells
from the bone marrow migrate to the thymus. The
majority (9095%) fail to produce T-cell receptor (TCR)
and die via the apoptotic pathway [86]. Thymocyte
apoptosis can be induced by endogenously produced
glucocorticoids and a lack of TCR [87, 88]. Death of a
cell clone (clonal deletion) due to TCR-induced
1086

apoptosis takes place in immature B cells that produce


antiself antibodies in the fetal liver and later in the bone
marrow [89, 90]. Animal studies indicate that autoimmune diseases, such as rheumatoid arthritis, systemic
lupus erythematosis, inflammatory bowel disease and
insulin-dependent diabetes, may be caused by dysfunctional apoptosis [91, 92]. There is evidence that cytotoxic
T cells kill their target cells by the induction of apoptosis
in the target [93]. Activated peripheral cytotoxic T cells
and peripheral B cells are removed by apoptosis [89].
Granulocyte apoptosis is also important for the
resolution of the inflammatory response. Inflammatory
agents such as lipopolysaccharide (LPS) and granulocyte
colony-stimulating factor (G-CSF) inhibit neutrophil
apoptosis. During apoptosis induced by TNF-a and
NO donors the neutrophil loses its ability to degranulate,
thus limiting the inflammation. Intact senescent neutrophils are engulfed by macrophages and degraded within
minutes, with no release of pro-inflammatory mediators.
When necrotic granulocytes are ingested, the macrophages release mediators leading to inflammation [94].
Haematological diseases, such as myelodysplastic syndromes, aplastic anaemia, chronic neutropenia or severe
b-thalassaemia, are associated with increased apoptosis
within the bone marrow [95].
Viral infection
Many viruses inhibit apoptosis in their target cells to
prolong host cell life and permit viral replication. Viruses
may encode anti-apoptotic proteins, such as the baculovirus IAPs, the baculovirus p35 and cowpox serpin
protein crmA, to promote the development of certain
cancers [96]. DNA viruses also contain anti-apoptotic
genes. For example, the papilloma virus and adenovirus
can encode a p53 inhibitor. Ribonucleic acid (RNA)
viruses may also contain anti-apoptotic genes [97].
Human immunodeficiency virus (HIV) infection is
characterised by a decreased proliferation of T cells with
loss of CD41 cells initially, and loss of CD81 cells, natural
killer cells and neurons later. Inappropriate induction of
apoptosis in HIV-infected CD41 cells is triggered by the
virus [98]. Another viral product, the HIV-1 transactivating protein, Tat, is produced by infected cells and taken
up by uninfected T cells. It enhances apoptosis by
reducing intracellular antioxidant levels, producing oxidative stress [99].
Sepsis and multiple organ dysfunction syndrome
Sepsis is often accompanied by MODS, which may
caused by apoptosis [100]. Systemic release of cytokines
such as TNF-a and IL-1b by bacterial LPS, resulting in
increased intracellular calcium and enhanced production
of oxygen free radicals, is thought to produce apoptosis in
q 2000 Blackwell Science Ltd

Anaesthesia, 2000, 55, pages 10811093


P. C. A. Kam and N. I. Ferch  Apoptosis
................................................................................................................................................................................................................................................

various organs [101104]. TNF-a may act by causing


macrophage release of IL-1b [105]. In a murine model of
septic shock, direct application of TNF-a caused
apoptosis in hepatocytes [106]. Further support for the
role of apoptosis in endotoxic shock is provided by the
finding that mice deficient in ICE have a marked
resistance to septic shock [107]. It is postulated that
apoptotic (as opposed to necrotic) cell death may confer
advantages to the septic organism by limiting the
inflammatory response [28]. Apoptosis is an important
cause of death of lymphocytes in the immune system
(thymus, lymph nodes and spleen) during bacterial and
viral infections [101, 108113]. Furthermore, apoptotic
cell death has been shown in the lung, ileum, colon and
skeletal muscle during sepsis [109]. Apoptosis may be an
important regulator of a balance between pro- and antiinflammatory factors and this balance is achieved through
widespread lymphocyte apoptosis and subsequently
diminished cytokine load [114].
Trauma patients with and without septic complications
show decreased neutrophil apoptosis associated with
increased tyrosine phosphorylation leading to excessive
tissue damage [115, 116]. Patients with SIRS and
following major elective aortic surgery also show
decreased neutrophil apoptosis, possibly due to circulating
pro-inflammatory anti-apoptotic factors such as LPS,
TNF-a, interferon (IFN)-g, G-CSF and granulocyte/
monocyte colony-stimulating factor [117]. The persistence of neutrophils in such conditions may contribute to
the pathophysiology of MODS through the excessive
release of toxic granule components and ROS leading to
tissue destruction. Apoptosis can be partially restored by
the addition of anti-TNF antibody and IL-10 [118].
Macrophages show increased rates of apoptosis in
polymicrobial sepsis, associated with NO and caspase
activation, leading to loss of phagocytic function [119].
Cancer
There is evidence that a failure to initiate apoptosis
following DNA damage may cause cancer. Elevated levels
of c-myc are found in many tumours [120122]. In
human follicular lymphoma, translocation of chromosomes 14 and 18 causes up-regulation of Bcl-2 `oncogene'
expression [48, 123]. Levels of Bcl-2 are elevated in
various other human cancers, for example lymphomas,
leukaemias, adenocarcinomas, renal and lung cancers,
neuroblastomas and melanomas [123126].
Other mutations may also be involved in carcinogenesis. A notable example involves the tumour suppressor p53 that represses Bcl-2 expression. The p53 gene is
deficient in over half of human cancers [127130]. For
example, in Wilm's tumour p53 mutations occur in
anaplastic areas.
q 2000 Blackwell Science Ltd

Renal system
Embryological development of the kidney involves
periods of growth and apoptosis which are reflected by
the levels of Bcl-2 present [131]. Mice deficient in Bcl-2
develop polycystic kidney disease [132] whereas, Bcl-2
levels are high in all renal tumours [133].
Gastrointestinal system
Gastrointestinal diseases may be associated with excessive
or defective apoptosis. Shigella dysenteriae causes excessive
apoptosis of macrophages in the lamina propria of the
intestine by the release of IL-1b. Mice expressing a
mutant nonfunctional N-cadherin in intestinal villi
develop changes similar to Crohn's disease and show
increased rates of apoptosis in both villi and crypts, as well
as higher rates of adenomas.
Progressive inhibition of apoptosis appears to be
involved in the pathogenesis of gastrointestinal neoplasia,
in particular colorectal cancer [134]. Genes that regulate
apoptosis are mutant in colonic and gastric cancers. Wildtype p53, when introduced into human colon cancer cell
lines, inhibits cell growth and induces apoptosis [135].
However, p53 expression is associated with a poorer
prognosis in both colorectal and gastric cancers [4]
possibly owing to mutant or nonfunctional p53 that fails
to induce the usual apoptosis. The Bcl-2 protein can also
be detected in human cancers and is highest in adenomas
[136].
Hepatic cells develop apoptosis when infected with
viruses as in chronic hepatitis [137]. Abnormal activation
of cytotoxic T cells may be involved in human fulminant
hepatitis [95]. Paracetamol stimulates increases in intracellular calcium which activates Ca21-dependent nucleases
[138]. Apoptosis also appears to mediate allograft rejection
in a model of liver transplantation [139].
Reproductive system
Apoptosis is continually inhibited in many tissues of the
reproductive system owing to the presence of trophic
hormones from the pituitary, gonads and uterus. When
the hormones are removed, the tissues undergo atrophy.
Ovarian follicles undergo growth or atresia in response to
cyclic changes in luteinizing hormone and follicle
stimulating hormone; the endometrium, breast and
prostate are dependent on the steroid hormones and
regress when these are removed [140].
Therapeutic possibilities and future directions

The widespread involvement of apoptosis in the pathophysiology of disease lends itself to therapeutic intervention. In diseases caused by increased cell loss, such as
viral hepatitis and neurodegenerative disease, the aim will
1087

P. C. A. Kam and N. I. Ferch  Apoptosis


Anaesthesia, 2000, 55, pages 10811093
................................................................................................................................................................................................................................................

be to minimise apoptosis by modifying the signals which


trigger the response (e.g. Ca21, ROS) or interfering with
the effectors (e.g. caspases and endonucleases). However,
inhibition of apoptosis may be deleterious because new
tumours may arise when damaged cells are prevented
from committing suicide. In diseases caused by deficient
apoptosis, such as cancer, viral latency and autoimmunity,
methods of producing selective apoptosis are being
sought.
Agents targeting receptors or regulatory molecules and
agents targeting the final common pathway are attractive
possibilities. The soluble form of Fas could prove useful
for increasing apoptosis (e.g. in tumours). Antibodies to
Fas or Fas ligand may be useful in preventing apoptosis
(e.g. in neurodegenerative disease). Preliminary successes
in treating chronic inflammatory diseases, such as
rheumatoid arthritis and ulcerative colitis, with TNF-a
inhibitors have been reported, but these therapies have
proved disappointing in sepsis [141].
The regulatory molecule Bcl-2 may protect normal
cells from death induced by cytotoxic agents [142]. In
contrast, decreasing Bcl-2 in cancer cells can reinstate
chemo- and radio-sensitivity. This can be achieved by
treatment with synthetic short DNA single strands called
antisense oligonucleotides, which bind to specific messenger RNA sequences and prevent production of the
offending protein [143].
The caspases are of interest from a therapeutic point of
view as specific inhibitors exist. Tetrapeptide aldehydes
are potent inhibitors of ICE but are toxic [4]. They act by
specifically binding to the protease and preventing
cleavage of the target proteins. Inhibitors of ICE can
inhibit apoptosis in a number of cell systems. They may
have a role in sepsis and chronic inflammatory and
neurodegenerative disease [144].
Cancer
Kerr et al. [145] found that anticancer agents induce
apoptosis in tumours. Chemotherapeutic agents reported
to induce apoptosis include the alkylating agents (cyclophosphamide, mitomycin C, nitrogen mustard), topoisomerase II inhibitors (daunorubicin, adriamycin),
dexamethasone, antimetabolites (methotrexate, 5-fluorouracil, 5-azacytidine), cisplatin, microtubule disrupters
(vincristine, vinblastine, taxol), cycloheximide, bleomycin,
cisplatin, tamoxifen and cytosine arabinose [30, 90].
Irradiation and cytotoxic agents produce DNA damage
which predisposes to p53 enhancement of apoptosis. If
p53 is defective, then resistance to chemotherapy may
result [129]. The activation of the p53 pathway in neoplasms to regain chemosensitivity is a potentially powerful
therapeutic tool that can render the tumour apoptotic.
This may be possible through p53 gene-specific therapy.
1088

Such therapy has been attempted using retrovirally


introduced wild-type p53 on non-small cell-lung cancer
with encouraging results [129]. Nicotine has been shown
to suppress apoptosis in lung cancer in humans [146].
Inflammatory disease
Corticosteroids induce eosinophil apoptosis but inhibit
neutrophil apoptosis. The treatment of asthmatic patients
with corticosteroids can cause eosinophil death and
macrophage engulfment [147]. The detection of this
process in airway secretions of asthmatic patients is
associated with clinical improvement [148].
Ischaemia and reperfusion
Apoptosis has been demonstrated in areas of brief
ischaemia and reperfusion in the brain, heart, liver and
kidney [11, 82]. Further understanding of the molecular
mechanisms should allow novel protective strategies to be
developed.
Gastrointestinal tract
Cytotoxic drugs induce apoptosis in studies of human
gastrointestinal cancer cells as well as normal mouse
intestine, which may account for their therapeutic action.
Chronic ingestion of nonsteroidal anti-inflammatory
drugs may be useful in preventing colonic cancer, possibly
by induction of apoptosis. Cyclo-oxygenase-2 (COX-2)
may enhance formation of cancer by changes in cellular
adhesion and by inhibition of apoptosis via enhanced
Bcl-2 expression [4, 134]. Non-steroidal anti-inflammatory drugs, which inhibit COX-1 and COX-2, can
prevent up-regulation of Bcl-2 by prostaglandins and
prevent colorectal carcinoma. The protective effects of
dietary fibre may be via its fermentation by bacteria in the
colon to form short-chain fatty acids (e.g. butyrate) which
promote apoptosis [4].
Conclusion

Cell injury, as a result of physical, biochemical or


biological insults, or from a deficiency of vital substances,
induces expression of adaptive stress response genes. The
interactions of the acute phase, heat shock and oxidative
stress responses appear to determine the fate of the cell.
The intracellular response to injury leads to distinct
genetic expression that mediates the cellular changes
which are usually specific to cell type and injury. These
cellular responses are usually cytoprotective but can
precipitate apoptosis. Reducing agents, antioxidants,
anti-TNF antibodies, steroid antagonists and inhibitors
of protein synthesis can modulate apoptosis. Novel genedirected treatment for various disease states, such as
multiple organ dysfunction syndrome and some cancers,
q 2000 Blackwell Science Ltd

Anaesthesia, 2000, 55, pages 10811093


P. C. A. Kam and N. I. Ferch  Apoptosis
................................................................................................................................................................................................................................................

may be available as the understanding of the impact and


mechanism of stress gene responses to injury are
elucidated.
References
1 Kerr JFR, Wyllie AH, Currie AR. Apoptosis: a basic
biological phenomenon with wide-ranging implications in
tissue kinetics. British Journal of Cancer 1972; 26: 23957.
2 Bar PR. Apoptosis the cell's silent exit. Life Science 1996;
59: 36978.
3 Thatte U, Dahanukar S. Apoptosis clinical relevance and
pharmacological manipulation. Drugs 1997; 54: 51132.
4 Pritchard DM, Watson AJM. Apoptosis and gastrointestinal
pharmacology. Pharmacological Therapeutics 1996; 72: 149
69.
5 Majno G, Joris I. Apoptosis, oncosis and necrosis an
overview of cell death. American Journal of Pathology 1995;
146: 315.
6 Graper L. Eine neue Anschauung uber physiologische
Zellausschaltung. Archive Zellforsch 1914; 12: 37394.
7 Glucksmann A. Cell deaths in normal vertebrate ontogeny.
Biological Reviews of the Cambridge Philosophical Society 1951;
26: 5986.
8 Farber E. Programmed cell death: necrosis versus apoptosis.
Modern Pathology 1994; 7: 6059.
9 Kerr JFR. An electron-microscope study of liver cell
necrosis due to Heliotrine. Journal of Pathology 1969; 97:
55762.
10 Kerr JFR. An electron-microscope study of liver cell
necrosis due to Albitocin. Pathology 1970; 2: 2519.
11 Kerr JFR. Shrinkage necrosis: a distinct mode of cellular
death. Journal of Pathology 1971; 105: 1320.
12 Wyllie AH, Kerr JFR, Currie AR. Cell death in the
normal neonatal rat adrenal cortex. Journal of Pathology
1973; 111: 25561.
13 Searle J, Lawson TA, Abbott PJ, Harmon B, Kerr JF. An
electron microscope study of the mode of cell death
induced by cancer-chemotherapeutic agents in populations
of proliferating normal and neoplastic cells. Journal of
Pathology 1975; 116: 12938.
14 Wyllie AH, Kerr JFR, Macaskill IAM, Currie AR.
Adrenocortical cell deletion: the role of ACTH. Journal of
Pathology 1973; 111: 8594.
15 Wyllie AH, Beattie GJ, Hargreaves AD. Chromatin changes
in apoptosis. Histochemistry Journal 1981; 13: 68192.
16 Ellis EM, Horvitz HR. Genetic control of programmed cell
death in the nematode Caenorhabditis elegans. Cell 1986; 4:
81729.
17 Korsmeyer SJ. Bcl-2 initiates a new category of oncogenes:
regulators of cell death. Blood 1992; 80: 87986.
18 Vaux DL, Cory S, Adams JM. Bcl-2 gene promotes
haemopoetic cell survival and cooperates with C-myc to
immortalize pre-B cells. Nature 1988; 335: 4402.
19 Nagata S, Goldstein P. The Fas death factor. Science 1995;
267: 144956.
20 Wyllie AH, Rose KA, Morris RG, Stell CM, Forster E,
q 2000 Blackwell Science Ltd

21
22
23
24
25
26
27

28
29
30
31
32
33
34
35
36
37

38
39

Spandidis DA. Rodent fibroblast tumours expressing


human myc and ras genes: growth, metastasis and
endogenous oncogene expression. British Journal of Cancer
1987; 56: 2519.
Evan GI, Wyllie AH, Gilbert CS, et al. Induction of
apoptosis in fibroblasts by C-myc protein. Cell 1992; 69:
11928.
Martin SJ. Apoptosis: suicide, execution or murder? Trends
in Cell Biology 1993; 3: 1414.
Earnshaw WC. Nuclear changes in apoptosis. Current
Opinions in Cell Biology 1995; 7: 33743.
Farber E. Chemical carcinogenesis. New England Journal of
Medicine 1981; 305: 137989.
Hockenbery D. Defining apoptosis. American Journal of
Pathology 1995; 146: 1619.
Kane AB. Redefining cell death. American Journal of
Pathology 1995; 146: 12.
Lennon SV, Martin SJ, Cotter TG. Dose-dependent
induction of apoptosis in human tumour cell lines by
widely divergent stimuli. Cell Proliferation 1991; 24: 203
14.
Cobb JP, Hotchkiss RS, Karl IE, Buchman TG. Mechanisms of cell injury and death. British Journal of Anaesthesia
1996; 77: 310.
Sulston JE, Horvitz HR. Post-embryonic cell lineages of
the nematode C. elegans. Developmental Biology 1977; 82:
11056.
Savitz SI, Daniel BA, Rosenbaum MD. Apoptosis in
neurological disease. Neurosurgery 1998; 42: 55572.
Arends MJ, Wyllie AH. Apoptosis: mechanisms and roles in
pathology. International Reviews of Experimental Pathology
1991; 32: 22354.
Wyllie AH. Apoptosis: an overview. British Medical Bulletin
1997; 53: 45165.
Weedon D, Searle J, Kerr JFR. Apoptosis: its nature and
implications for dermatopathology. American Journal of
Dermatopathology 1979; 1: 13344.
Wyllie A. Glucocorticoid-induced thymocyte apoptosis is
associated with endogenous endonuclease activation.
Nature 1980; 284: 5556.
Kontogeorgos G, Kovacs K. Apoptosis in endocrine glands.
Endocrine Pathology 1995; 6: 25765.
Bortner CD, Oldenburg NBS, Cedlowski JA. The role of
DNA fragmentation in apoptosis. Trends in Cell Biology
1995; 5: 216.
Gavrieli Y, Sherman Y, Benasson SA. Identification of
programmed cell death in situ via special labelling of nuclear
DNA fragments. Journal of Cell Biology 1992; 119: 493
501.
Hamel W, Dazin P, Israel M. Adaptation of a simple flow
cytometric assay to identify different stages during
apoptosis. Cytometry 1996; 25: 17381.
Dive C, Gregory CD, Phipps DJ, Evans DL, Milner AE,
Wyllie AH. Analysis and discrimination of necrosis and
apoptosis (programmed cell death) by multiparameter flow
cytometry. Biochemica et Biophysica Acta 1992; 1133: 275
85.
1089

P. C. A. Kam and N. I. Ferch  Apoptosis


Anaesthesia, 2000, 55, pages 10811093
................................................................................................................................................................................................................................................

40 Gong J, Traganos F, Darsynkiewicz Z. A selective


procedure for DNA extracting form apoptotic cells
applicable for gel electrophoresis and flow cytometry.
Analytical Biochemistry 1994; 218: 31419.
41 Rudin CM, Thompson CB. Apoptosis and disease:
regulation and clinical relevance of programmed cell death.
Annual Review of Medicine 1997; 48: 26781.
42 MacLellan WR, Schneider MD. Death by design
programmed cell death in cardiovascular biology and
disease. Circulation Research 1997; 81: 13744.
43 Haimovitz-Friedman A, Kan C, Enleiter D, et al. Ionizing
radiation acts on cellular membranes to generate ceramide
and initiate apoptosis. Journal of Experimental Medicine 1994;
180: 52535.
44 Santana P, Pena LA, Haimovitz-Friedman A, et al. Acid
sphingomyelinase deficient lymphoblasts and mice are
defective in radiation-induced apoptosis. Cell 1996; 86:
18999.
45 Narula J, Kharbanda S, Khaw B-A. Apoptosis and the
heart. Chest 1997; 112: 135862.
46 Vaux DL, Weissman IL, Kim SK. Prevention of programmed cell death in Caenorhabditis elegans by human Bcl2. Science 1992; 258: 19557.
47 Brown R. The Bcl-2 family of proteins. British Medical
Bulletin 1996; 53: 46677.
48 Tsujimoto Y, Cossman J, Jaffe E, Croce C. Involvement of
the Bcl-2 gene in human follicular lymphoma. Science 1985;
228: 14403.
49 Cleary ML, Smith SD, Sklar J. Cloning and structural
analysis of cDNAs for Bcl-2 and a hybrid Bcl-2
immunoglobulin transcript resulting from the T (14; 18)
translocation. Cell 1986; 47: 1928.
50 Hockenbery D, Oltavi Z, Yin X, Milliman C, Kersmeyer
B. Bcl-2 functions in an antioxidant pathway to prevent
apoptosis. Cell 1993; 75: 24151.
51 Yang J, Liu X, Bhalla K, et al. Prevention of apoptosis by
Bcl-2: release of cytochrome C from mitochondria
blocked. Science 1997; 275: 112932.
52 Haunstetter A, Izumo S. Apoptosis basic mechanisms and
implications for cardiovascular disease. Circulation Research
1998; 82: 111129.
53 Olivetti G, Abbi R, Quaini F, et al. Apoptosis in the failing
human heart. New England Journal of Medicine 1997; 336:
113141.
54 Miyashita T, Reed JC. Tumour suppressor p53 is a direct
transcriptional activator of the human bax gene. Cell 1995;
80: 2939.
55 Donehower LA, Harvey M, Slagle BL, et al. Mice deficient
for p53 are developmentally normal but susceptible to
spontaneous tumours. Nature 1992; 356: 21521.
56 Roy N, Mahadevan MS, McLean M, et al. The gene for
neuronal apoptosis inhibitory protein is partially deleted in
individuals with spinal muscular atrophy. Cell 1995; 80:
16778.
57 Oberhammer F, Wilson JW, Dive C, et al. Apoptotic death
in epithelial cells: cleavage of DNA to 300 and/or 50kb
1090

58
59
60
61
62

63
64
65
66

67
68
69
70
71

72

73

74

75

fragments prior to or in the absence of internucleosomal


fragmentation. EMBO Journal 1993; 12: 367984.
Whyte M. ICE/CED-3 proteases in apoptosis. Trends in
Cell Biology 1996; 6: 2458.
Thornberry NA, Rosen A, Nicholson DW. Control of
apoptosis by proteases. Advances in Pharmacology 1997; 41:
15577.
Enari M, Hug H, Nagata S. Involvement of an ICE-like
protease in Fas-mediated apoptosis. Nature 1995; 375: 78
81.
Thornberry NA. The caspase family of cysteine proteases.
British Medical Bulletin 1996; 53: 47890.
Knight CR, Ress RC, Griffin M. Apoptosis: a potential
role for cytosolic transglutaminase and its importance in
tumour progression. Biochemica et Biophysica Acta 1991;
1096: 31218.
Savill J. Recognition and phagocytosis of cells undergoing
apoptosis. British Medical Bulletin 1997; 53: 491508.
Savill J. Phagocytic docking without shocking. Nature
1998; 392: 4423.
Savill J, Haslett C. Granulocyte clearance by apoptosis in
the resolution of inflammation. Seminars in Cell Biology
1995; 6: 38593.
Savill J, Hogg N, Ren Y, Haslett C. Thrombospondin
cooperates with CD36 and the vitronectin receptor in
macrophage recognition of neutrophils undergoing apoptosis. Journal of Clinical Investigation 1992; 90: 151322.
Devitt A, Moffatt OD, Raykundalia C, Capra JD, Simmons
DL, Gregory CD. Human CD14 mediates recognition and
phagocytosis of apoptotic cells. Nature 1998; 392: 5058.
Louis J, Magal E, Takayama S, Varon S. CNTF protection
of oligodendrocytes against natural and tumour necrosis
factor-induces death. Science 1993; 259: 68992.
Choi DW. Ionic dependence of glutamate neurotoxicity in
cortical cell culture. Journal of Neuroscience 1987; 7: 36989.
Choi DW, Malucci-Gedde M, Kriegstein SR. Glutamate
neurotoxicity in cortical cell culture. Journal of Neuroscience
1987; 7: 35768.
Kure S, Tominaga T, Yoshimoto T, Tada K, Narisawa K.
Glutamate triggers internucleosomal DNA cleavage in
neuronal cells. Biochemistry and Biophysics Research Communications (New York) 1991; 179: 3945.
Tymianski M, Tator CH. Normal and abnormal calcium
homeostasis in neurones: a basis for the pathophysiology of
traumatic and ischaemic central nervous system injury.
Neurosurgery 1996; 38: 117695.
Linnick MD, Zahos P, Geschwind MD, Fedoroff HJ.
Expression of bcl-2 from a defective herpes simplex virus-1
vector limits neuronal death in focal cerebral ischaemia.
Stroke 1995; 26: 16705.
Rink A, Fung KM, Trojanowski JQ, Lee VM, Neugebauer
E, McIntosh TK. Evidence of apoptotic cell death after
experimental traumatic brain injury in the rat. American
Journal of Pathology 1995; 147: 157583.
Loo D, Copani A, Pike C, Whittemore E, Walencewicz A,
Cotman C. Apoptosis is induced by -amyloid in cultured
q 2000 Blackwell Science Ltd

Anaesthesia, 2000, 55, pages 10811093


P. C. A. Kam and N. I. Ferch  Apoptosis
................................................................................................................................................................................................................................................

76

77
78
79

80

81

82

83

84
85

86
87

88

89
90

central nervous system neurones. Proceedings of the National


Academy of Sciences of the USA 1993; 90: 79515.
Lee WD, Colom LV, Xie WJ, Smith RG, Alexianu M,
Appel SH. Cell death induced by -amyloid 140 in MES
23.5 hybrid clone: the role of nitric oxide and NMDAgated channel attraction leading to apoptosis. Brain Research
1995; 686: 4960.
Wu JYeZ, Darras B. Frequency of p53 tumor suppressor
gene mutations in human primary brain tumors. Neurosurgery 1993 33: 82430.
Bogler O, Huang HJ, Kleihues P, Cavenee WK. The p53
gene and its role in human brain tumors. Glia 1995; 15:
30827.
Clarke MF, Apel IJ, Benedict MA, et al. A recombinant bclxs adenovirus selectively induces apoptosis in cancer cells
but not in normal bone marrow cells. Proceedings of the
National Academy of Sciences of the USA 1995; 92: 110248.
Yu J, Sena-Esteves M, Paulus W, Breakfield X, Reeve DS.
Retroviral delivery and tetracycline-dependent expression
of IL-1b-converting enzyme (ICE) in a rat glioma model
provides controlled induction of apoptotic death in tumor
cells. Cancer Research 1996; 56: 54237.
Tanaka M, Ito H, Adachi S, et al. Hypoxia induces
apoptosis with enhanced expression of Fas antigen
messenger RNA in cultured neonatal rat cardiomyocytes.
Circulation Research 1994; 75: 42633.
Gottlieb RA, Burleson KO, Kloner RA, Babior BM,
Engler RA. Reperfusion injury induced apoptosis in rabbit
cardiomyocytes. Journal of Clinical Investigation 1994; 94:
16218.
Bennett MR, Evan GI, Schwartz SM. Apoptosis of human
vascular smooth muscle cells derived from normal vessels
and coronary atherosclerotic plaques. Journal of Clinical
Investigation 1995; 95: 226674.
Isner JM, Kearney M, Bortram S, Passeri J. Apoptosis in
human atherosclerosis and restenosis. Circulation 1995; 91:
270311.
James TN. Normal and abnormal consequences of
apoptosis in the human heart: from postnatal morphogenesis to paroxysmal arrhythmias. Transactions of the American
Clinical and Climatological Association 1993; 105: 14577.
Surh CD, Sprent J. T-cell apoptosis detected in situ during
positive and negative selection in the thymus. Nature 1994;
372: 1003.
Zacharchuk CM, Mercep M, Ashwell JD. Programmed T
lymphocyte death: cell activation- and steroid-induced
pathways are mutually antagonistic. Journal of Immunology
1990; 145: 403745.
Iwata M, Hanaoka S, Sato K. Rescue of thymocytes and T
cell hybridomas from glucocorticoid-induced apoptosis by
stimulation via the T cell receptor/CD3 complex: a
possible in vitro model for positive selection of the T cell
repertoire. European Journal of Immunology 1991; 21: 6438.
Osborne BA. Apoptosis and the maintenance of homeostasis in the immune system. Current Opinion in Immunology 1996; 8: 24554.
McConkey DJ, Zhivotovsky B, Orrenius S. Apoptosis

q 2000 Blackwell Science Ltd

91
92
93
94
95
96
97
98

99
100
101

102
103

104
105
106

107

108

109

molecular mechanisms and biomedical implications. Molecular Aspects of Medicine 1996; 17: 1110.
Elkon KB. Mechanisms of autoantibody production and
their role in disease. Mount Sinai Journal of Medicine 1994;
61: 28390.
Emlen W, Niebur J, Kadera R. Accelerated in vitro
apoptosis of lymphocytes from patients with systemic lupus
erythematosis. Journal of Immunology 1994; 152: 368592.
Eckert PG, Vaux DL. Apoptosis and the immune system.
British Medical Bulletin 1997; 53: 591603.
Haslett C. Granulocyte apoptosis and inflammatory disease.
British Medical Bulletin 1997; 53: 66983.
Solary E, Dubrez L, Eymin B. The role of apoptosis in the
pathogenesis and treatment of diseases. European Respiratory
Journal 1996; 9: 1293305.
Clem RJ, Hardwick JM, Miller LK. Anti-apoptotic genes
of baculovirus. Cell Death 1996; 3: 916.
Young LS, Dawson CW, Eliopoulos A. Viruses and
apoptosis. British medical Bulletin 1997; 53: 50921.
Terai C, Kornbluth TS, Pauza CD, Richman DD, Carson
AD. Apoptosis as a mechanism of cell death in cultured T
lymphoblasts acutely infected with HIV-1. Journal of Clinical
Investigation 1991; 87: 171015.
Peter ME, Ehret A, Berndt C, Krammer PH. AIDS and the
death receptors. British Medical Bulletin 1997; 53: 60416.
Carrico CJ, Meakins JL, Marshall JC, Fry D, Maier RV.
Multiple organ failure syndrome. Archives of Surgery 1986;
121: 196208.
Ayala A, Herdon C, Lehman D, DeMaso CM, Ayala CA,
Chaudry IH. The induction of accelerated thymic
programmed cell death during polymicrobial sepsis: control
by corticosteroids but not tumor necrosis factor. Shock
1995; 3: 25967.
Zaloga GP, Washburn D, Black KW, Prielipp R. Human
sepsis increases lymphocyte intracellular calcium. Critical
Care Medicine 1993; 21: 196202.
Bautista AP, Meszaros K, Bojta J, Spitzer J. Superoxide
anion generation in the liver during the early stage of
endotoxaemia in rats. Journal of Leukemia Biology 1990; 48:
1238.
Vassali P. The pathophysiology of tumor necrosis factors.
Annual Review of Immunology 1992; 10: 41152.
Dinarello CA. The biological properties of interleukin-1.
European Cytokine Network 1994; 5: 51731.
Leist M, Gantner F, Jilg S, Wendel A. Activation of the
55 kDa TNF receptor is necessary and sufficient for TNFinduced liver failure, hepatocyte apoptosis, and nitrite
release. Journal of Immunology 1995; 154: 130716.
Li P, Allen H, Banerjee S. Mice deficient in IL-1bconverting enzyme are defective in production of mature
IL-1b and resistant to endotoxic shock. Cell 1995; 80:
40111.
Ayala A, Herdon C, Lehman D, Ayala CA, Chaudry IH.
Differential induction of apoptosis in lymphoid tissues
during sepsis: variation in onset, frequency and nature of
the mediators. Blood 1996; 87: 426175.
Hotchkiss RS, Swanson PE, Cobb JP, Jacobson A,
1091

P. C. A. Kam and N. I. Ferch  Apoptosis


Anaesthesia, 2000, 55, pages 10811093
................................................................................................................................................................................................................................................

110
111

112

113
114
115

116

117

118

119

120
121

122

123

Buchman TG, Karl IE. Apoptosis in lymphoid and


parenchymal cells during sepsis: findings in normal and
T- and B-cell deficient mice. Critical Care Medicine 1997;
25: 1298307.
Wang SD, Huang KJ, Lin YS, Lei HY. Sepsis-induced
apoptosis of the thymocytes in mice. Journal of Immunology
1994; 152: 501421.
Barke RA, Roy S, Chapin RB, Charboneau R. The role
of programmed cell death (apoptosis) in thymic
involution following sepsis. Archives of Surgery 1994; 129:
125662.
Zhang XM, Morikawa A, Takahashi K, et al. Localization
of apoptosis (programmed cell death) in mice by administration of lipopolysaccharide. Microbiology and Immunology
1994; 38: 66971.
Rogers HW, Callery MP, Deck B, Unanue ER. Listeria
monocytogenes induce apoptosis of infected hepatocytes.
Journal of Immunology 1996; 156: 67986.
Bone RC. Sir Isaac Newton, sepsis. SIRS and CARS.
Critical Care Medicine 1996; 24: 11259.
Ertel W, Keel M, Infanger M, Ungethum U, Steckholzer
U, Trentz O. Circulating mediators in serum of injured
patients with septic complications inhibit neutrophil
apoptosis through up-regulation of proteintyrosine
phosphorylation. Journal of Trauma 1998; 44: 76775.
Ertel W, Keel M, Ungethum U, Trentz O. Pro-inflammatory cytokines regulate apoptosis of granulocytes during
systemic inflammation. Langenbecks Archive Fur Chirugie
1997; 114: 6279.
Jimenez MF, Watson RW, Parodo J, et al. Dysregulated
expression of neutrophil apoptosis in the systemic inflammatory response syndrome. Archives of Surgery 1997; 132:
12639.
Keel M, Ungethum U, Steckholzer U, et al. Interleukin-10
counter-regulates proinflammatory cytokine-induced
inhibition of neutrophil apoptosis during severe sepsis.
Blood 1997; 90: 335663.
Williams TE, Ayala A, Chaudry IH. Inducible macrophage
apoptosis following sepsis is mediated by cysteine protease
activation and nitric oxide release. Journal of Surgical Research
1997; 70: 11318.
Spencer CA, Groudine M. Control of C-myc regulation in
normal and neoplastic cells. Advances in Cancer Research
1991; 56: 148.
Loke SL, Stein C, Zhang X, Avigan M, Cohen J, Neckers
LM. Delivery of C-myc antisense phosphorothioate
oligodeoxynucleotides to haematopoietic cells in culture by
liposome fusion: specific reduction in C-myc protein
expression correlates with inhibition of cell growth and
DNA synthesis. Current Topics in Microbiology and Immunology 1988; 141: 2829.
Bennet M, Anglin A, McEwan J, Jagoe R, Newby A, Evan
G. Inhibition of vascular smooth muscle cell proliferation in
vitro and in vivo by C-myc antisense oligonucleotides.
Journal of Clinical Investigation 1993; 93: 8208.
Reed JC. Bcl-2 and the regulation of cell death. Journal of
Cell Biology 1994; 124: 16.

1092

124 Allsop TE, Wyatt S, Paterson HF, Davies AM. The


proto-oncogene bcl-2 can selectively rescue neurotrophic
factor-dependent neurons from apoptosis. Cell 1993; 73:
295307.
125 Levine B, Huang Q, Isaacs JT, Reed JC, Griffin DE,
Hardwick JM. Conversion of lytic to persistent alphavirus
infection by the bcl-2 cellular oncogene. Nature 1993; 361:
73942.
126 Miyashita T, Reed JC. Bcl-2 oncoprotein blocks chemotherapy-induced apoptosis in a human leukemia cell
line. Blood 1993; 81: 1517.
127 Harris CC, Holstein M. Clinical implications of the p53
tumor suppressor gene. New England Journal of Medicine
1993; 329: 131827.
128 Lowe SW, Jacks T, Housman DE, Ruley HE. Abrogation
of oncogene-associated apoptosis allows transformation of
p53-deficient cells. Proceedings of the National Academy of
Sciences of the USA 1994; 91: 202630.
129 Bellamy CO. P53 and apoptosis. British Medical Bulletin
1997; 53: 52238.
130 Strand S, Hoffman WJ, Hug H, et al. Lymphocyte apoptosis
induced by CD 45 (APO-1/fas) ligand-expressing tumour
cells a mechanism of immune-evasion? Nature Medicine
1996; 2: 13616.
131 Savill J. Apoptosis and the kidney. Journal of the American
Society of Nephrology 1994; 5: 1221.
132 Veis DJ, Sorenson CM, Shutter JR, Korsmeyer SJ. Bcl-2deficient mice demonstrate fulminant lymphoid apoptosis,
polycystic kidneys, and hypopigmented hair. Cell 1993; 75:
22940.
133 Chandler D, El-Naggar AK, Brisbay S, Redline RW,
McDonnel TJ. Apoptosis and expression of the Bcl-2
proto-oncogene in the fetal and adult human kidney:
evidence for the contribution of bcl-2 expression to renal
carcinogenesis. Human Pathology 1994; 25: 78996.
134 Bedi A, Pasricha PJ, Akhtar AJ, et al. Inhibition of apoptosis
during development of colorectal cancer. Cancer Research
1995; 55: 181116.
135 Shaw P, Bovey R, Tardy S, Sahli R, Sordat B, Costa J.
Induction of apoptosis by wild-type p53 in a human colon
tumour-derived cell line. Proceedings of the National Academy
of Sciences of the USA 1992; 89: 44959.
136 Bosari S, Moneghini L, Graziani D, et al. Bcl-2 oncoprotein
in colorectal hyperplastic polyps, adenomas, and adenocarcinomas. Human Pathology 1995; 26: 53440.
137 Hiramatsu N, Hayashi N, Katayama K, et al. Immunohistochemical detection of Fas antigen in liver tissue of
patients with chronic hepatitis C. Hepatology 1994; 19:
13549.
138 Ray SD, Kamendulis LM, Gurule MW, Yorkin RD,
Corcoran GB. Ca21 antagonists inhibit DNA fragmentation and toxic cell death induced by acetaminophen.
FASEB Journal 1993; 7: 45363.
139 Krams SM, Egawa H, Quinn MB, Villanueva JC, GarciaKennedy R, Martinez OM. Apoptosis as a mechanism of
cell death in liver allograft rejection. Transplantation 1995;
59: 6215.
q 2000 Blackwell Science Ltd

Anaesthesia, 2000, 55, pages 10811093


P. C. A. Kam and N. I. Ferch  Apoptosis
................................................................................................................................................................................................................................................

140 Gosden R, Spears N. Programmed cell death in the


reproductive system. British Medical Bulletin 1997; 53: 644
61.
141 Natanson C, Hoffman WD, Suffredini AF, Eichacker PQ,
Danner RL. Selected treatment strategies for septic shock
based on proposed mechanisms of pathogenesis. Annals of
Internal Medicine 1994; 120: 77183.
142 Kondo S, Takeuchi J, Morimura T, Oda Y, Kikuchi H. Bcl2 gene enables rescue from in vitro myelosupression (bone
marrow cell death) induced by chemotherapy. British
Journal of Cancer 1994; 70: 4216.
143 Campos L, Sabido O, Rouault JP, Guyotat D. Effects of
Bcl-2 antisense oligonucleotides on in vitro proliferation
and survival of normal bone marrow progenitors and
leukemic cells. Blood 1994; 84: 595600.

q 2000 Blackwell Science Ltd

144 Livinston DJ. In vitro and in vivo studies of ICE inhibitors.


Journal of Cell Biochemistry 1997; 64: 1926.
145 Kerr J, Witerford C, Harmon B. Apoptosis: its significance
in cancer and cancer therapy. Cancer 1994; 73: 201326.
146 Maneckgee R, Minna JD. Opioids induce while nicotine
suppresses apoptosis in human lung cancer cells. Cell
Growth and Differentiation 1994; 5: 103340.
147 Meagher L, Cousin JM, Seckl JR, Haslett C. Opposing
effects of glucocorticoids on the rate of apoptosis in
neutrophilic and eosinophilic granulocytes. Journal of
Immunology 1996; 156: 44228.
148 Wooley KL, Gibson PG, Carty K, Wilson AJ, Twaddel SH,
Wooley MJ. Eosinophil apoptosis and the resolution of
airway inflammation in asthma. American Journal of
Respiratory and Critical Care Medicine 1996; 154: 23743.

1093

You might also like