Вы находитесь на странице: 1из 11

Pathophysiology 7 (27) 153 163

www.elsevier.com/locate/pathophys

Review

Oxidative stress and apoptosis


Krishnaswamy Kannan a, Sushil K. Jain b,*
a

Department of Medicine, Centre of Excellence for Arthritis and Rheumatology, Lousiana State Uni6ersity Health Sciences Center, Shre6eport,
LA 71130 -3932, USA
b
Department of Pediatrics, Lousiana State Uni6ersity Health Sciences Center, 1501 Kings Highway, Shre6eport, LA71130 -3932, USA
Received 9 March 2000; accepted 15 May 2000

Abstract
Apoptosis or programmed cell death, is essential for the normal functioning and survival of most multi-cellular organisms. The
morphological and biochemical characteristics of apoptosis, however, are highly conserved during the evolution. It is currently
believed that apoptosis can be divided into at least three functionally distinct phases, i.e. induction, effector and execution phase.
Recent studies have demonstrated that reactive oxygen species (ROS) and the resulting oxidative stress play a pivotal role in
apoptosis. Antioxidants and thiol reductants, such as N-acetylcysteine, and overexpression of manganese superoxide (MnSOD)
can block or delay apoptosis. Bcl-2, an endogenously produced protein, has been shown to prevent cells from dying of apoptosis
apparently by an antioxidative mechanism. Taken together ROS, and the resulting cellular redox change, can be part of signal
transduction pathway during apoptosis. It is now established that mitochondria play a prominent role in apoptosis. During
mitochondrial dysfunction, several essential players of apoptosis, including pro-caspases, cytochrome C, apoptosis-inducing factor
(AIF), and apoptotic protease-activating factor-1 (APAF-1) are released into the cytosol. The multimeric complex formation of
cytochrome C, APAF-1 and caspase 9 activates downstream caspases leading to apoptotic cell death. All the three functional
phases of apoptosis are under the influence of regulatory controls. Thus, increasing evidences provide support that oxidative stress
and apoptosis are closely linked physiological phenomena and are implicated in pathophysiology of some of the chronic diseases
including AIDS, autoimmunity, cancer, diabetes mellitus, Alzheimers and Parkinsons and ischemia of heart and brain. 2000
Elsevier Science Ireland Ltd. All rights reserved.
Keywords: Reactive oxygen species; Apoptosis-inducing factor; Apoptotic protease-activating factor-1

1. Introduction
Apoptosis, or programmed cell death (PCD), is a
naturally occurring cell death process, essential for the
normal development and homeostasis of all multicellular organisms [1]. This process is also important for
removing damaged, infected, or potentially neoplastic
cells. However, both too little and too much apoptotic
cell death can lead to adverse biological consequences
[24]. Rheumatoid arthritis and cancer are the best examples for too little apoptosis. Ischemic heart disease,
AIDS and neurodegenerative diseases such as
* Corresponding author. Tel.: +1-318-6756086; fax: + 1-3186756059.
E-mail address: sjain@lsuhsc.edu (S.K. Jain).

Alzheimers, and Parkinsons are the best examples of


too much apoptosis [26]. The commercial availability
of an ever-growing list of reagents, an explosive number
of research publications, and a number of scientific
meetings devoted to apoptosis signifies the relative importance of this emerging field [6].
Apoptosis is a cell death process that is clearly distinct from necrosis. The integrity of the cell membrane
is severely compromised or damaged in necrosis, leading to cell swelling and cell lysis. Often, necrosis occurs
in a group of cells, or in tissue at a particular locus. In
contrast, apoptosis occurs at the single cell level. During this process, an individual cell undergoes an active
process of cell death, set in motion by a genetic program and culminating in DNA fragmentation and the
formation of membrane-packaged bits called apoptotic

0928-4680/00/$ - see front matter 2000 Elsevier Science Ireland Ltd. All rights reserved.
PII: S 0 9 2 8 - 4 6 8 0 ( 0 0 ) 0 0 0 5 3 - 5

154

K. Kannan, S.K. Jain / Pathophysiology 7 (2000) 153163

bodies. The plasma membrane of early apoptotic cells


and the apoptotic bodies maintain their integrity, preventing leakage of cellular material and thereby an
inflammatory response and local tissue damage. Biochemical changes on the outer surface of plasma membrane are recognized by tissue macrophages and several
other neighboring cell types. Apoptotic cells are efficiently cleared in vivo by antigen processing cells within
2 4 h [1,3,5].
Apoptosis can be triggered by numerous factors including receptor-mediated signals, withdrawal of
growth factors, anti-tumor drugs and, under certain
conditions, damage to DNA (1 7). A partial list of
some of these inducers is included in Table 1. Each of
these stimuli has its own specific pathway that leads to
activation of apoptotic process, however, all appear to
converge at a highly conserved sequence of events. The
caspases are central component of this apoptotic program [8]. While the initial signal for apoptotic programming may vary, the morphological and biochemical
characteristics of PCD are uniformly similar, and are
Table 1
A partial list of inducers of oxidative stress and or apoptosis
Inducers of oxidative stress
Reactive oxygen
species
Metals
Pathophysiologic
condition

Superoxide radical, hydroxyl


radial, hydrogen peroxide
Iron, cadmium, mercuric
chloride
Hyperglycemia, ischemic heart
disease, Alzheimers and
Parkinsons

Inducers of apoptosis that in most cases, involve severe oxidative


stress
Pro-oxidants
H2O2, diamide, etoposide and
semiquinones
Ionizing radiation
Gamma UV radiation
Protein synthesis inhibitor
Cycloheximide
Apoptotic stimuli
Fas (CD95), TNFa, ceramide,
glutamate, growth factor
withdrawl (IL-2, IL-3, nerve
growth factor and serum
starvation)
Physiologic stimuli
Glucocorticosteroids, calcium,
TNF-a, glutamate
Pathophysiologic
Serum starvation, IL-2 and
conditions
IL-3 withdrawal,
hyperglycemia, ischemia and
reperfusion
Pro-apoptotic genes
p53, Bax, c-myc
Organic solvents and
Benzene and their metabolites
metabolites
and 2,5-hexanedione
Pesticides
DDT, endosulfan, dieldrin,
and 2,3,7,8
tetrachrolorodibenzo-p-dioxin
Drugs
Actinomycin D, cisplatin,
cycloheximide, taxol,
camptothecin and
staurosporine

highly conserved during the evolution of species. These


observations raise the intriguing possibility of the coexistence of multiple signaling pathways that converge
upstream of a common mechanism of events, predisposing the cell to apoptosis [15]. It is currently believed that one such convergent scheme involves the
active participation of mitochondria [912].
It is now well established that mitochondria is the
main site of the generation of oxygen radicals, such as,
superoxide anion, hydroxyl radical, singlet oxygen and
hydrogen peroxide [7,13]. It is estimated that 1 4% of
oxygen reacting with the respiratory chain leads to the
formation of superoxide radicals (O2). Other sources
of reactive oxygen species include radiation, cytotoxic
chemicals and drugs. The formation and the sources of
free radicals have been the subject of many reviews
[9,1417].
Excess oxidative stress kills cells either by necrosis or
by apoptosis [9,10]. In many models of apoptosis, alterations in the redox status of the cell to a more oxidizing
environment occurs prior to activation of the final
phase of caspase activation [1015]. This argument is
further supported by the ability of various anti-oxidants
such as N-acetylcysteine (NAC) to block apoptosis in a
similar way that caspase inhibitors do [18]. The anti-oxidant properties of Bcl-2, a potent inhibitor of apoptosis, further supports this view [1922]. Under normal
conditions, aerobic cells are endowed with extensive
anti-oxidant defense mechanisms to counteract the
damaging effects of ROS [7,13,14]. When pro-oxidants
overwhelm anti-oxidant defense mechanisms, oxidative
stress occurs. Interestingly, apoptosis may serve as a
fail-safe device to prevent cells from running amok and
proliferating uncontrollably in the face of a persistent
oxidative stress [23]. This review presents an overview
of the relationship between oxidative stress and apoptosis and its relevance to human health.

2. Mediators of apoptosis
Most of the mediators of apoptosis can be broadly
placed in one of five categories based on the primary
perturbation within the cell, i.e. (1) the cell surface, (2)
the cytosol, (3) the cytoskeleton, (4) the mitochondrion,
(5) and the nucleus. At the cell surface, some of the
well-characterized death receptors are Fas (CD95), tumor necrosis factor receptor-1 (TNF-Rl), CAR1, DR3,
DR4, and DR5 [2429]. The ligands that activate these
death receptors are structurally related molecules that
belong to the tumor necrosis factor (TNF) gene superfamily [1,30]. Interaction of death receptors with ligands may lead to the initiation of a death signal,
depending upon the presence of intracellular death
domains and its physical association with the adapter
protein called Fas-associated death domain (FADD)

K. Kannan, S.K. Jain / Pathophysiology 7 (2000) 153163

[28,29]. Additionally, FADD also contains another


death effector domain that communicates with caspase
8, one of the key enzymes in the death machinery. This,
in turn, activates downstream caspases, thus committing the cell to apoptosis. Further examples of ligands
that act at the cell surface include the CD40/CD40
ligand on T and B cells, and the glutamate/glutamate
receptor in the nervous system [28,31 33].
A major shift in the focus of research on apoptosis
from the nucleus to the cytoplasm has occurred in
recent years. A substantial fraction of the pro-apoptotic
members is localized in the cytosol or cytoskeleton
[12,13,30]. Following a death signal, the pro-apoptotic
members undergo a conformational change that enables them to target and integrate various subcellular
systems including mitochondrial outer membrane [34].
Another major player in cell death by apoptosis is a
class of enzymes called caspases, which are cysteinedependent enzymes and are sensitive to the redox status
of the cell [8,30,35,36]. These cysteine proteases are
synthesized as precursors that have little, if any, catalytic activity. The precursor is usually converted to the
active protease by proteolytic processing. One role of
caspases is to inactivate proteins that are vital to cell
survival. These enzymes cut off the cell cell contact to
the surrounding cells, disorganize the cytoskeleton, shut
down DNA replication and repair, interrupt splicing,
destroy DNA, disrupt the nuclear structure, induce
biochemical changes on the cell surface for easy recognition by phagocytes, and finally disintegrate the cell to
vesicular bodies [35,37 42]. At present, 13 different
caspases have been identified and more are being added
to the list [37]. Initiation of apoptotic events, however,
depends on the ability of the signaling complexes to
generate an active protease [8].
Poly (ADP-ribose) polymerase (PARP) is a cellular
substrate for caspase 3 and 7. Caspase 3 has also been
demonstrated to cleave all of the three following substrates, DNA dependent protein kinase (DNA-PKcs,
the 70 kDa protein component of the U1-ribonucleoprotein (U1-70 kDa), and PARP, at very similar sites,
defining the DXXD motif as the key determinant for
cleavage specificity. Specific inactivation of these substrates initiate an irreversible stress on the organization
of nuclear DNA structure and the regulation of gene
expression [45,46,51].
Under normal conditions, mitochondria possess an
efficient biochemical defense mechanism to neutralize
the effect mediated by ROS. This system is composed
of GSH, glutathione peroxidase, glutathione reductase,
superoxide
dismutase,
NADP
dehydrogenase
(NADPH), Vitamin E and C [13,18,43 46]. For example, superoxide radicals are scavenged by superoxide
dismutase, leading to the production of hydrogen peroxide, which again is detoxified by glutathione peroxidase or catalase. Oxidative stress can occur under

155

conditions when oxygen radicals production is greater


than the detoxification capacity of the cell [47,48].
Several lines of experimental evidence recognize the
mitochondrial dysfunction as one of the important
mediators of apoptosis [915,23,4956].

3. Oxidative stress and its significance in biology


ROS has been shown to play both beneficial as well
as deleterious roles. At very low concentration, it may
act as a second messenger in some of the signal transduction pathways [57]. However, when produced in
excess, it can cause oxidative damage to many vital
components of the cell. There exists a dynamic relationship between ROS production and antioxidant capacity
of the given cell system. Some oxidation processes such
as cysteine oxidation play a role in a dynamic regulatory process within the cell. Such a variation may cause
a drastic modulation of the oxidized or reduced ratio of
signaling proteins, such as, transcription factors. One
mechanism through which these effectors may elicit
oxidative stress is the small G-protein Ras. Indeed,
Ras is suspected to activate a cascade of kinases via
ROS production [58]. Similarly, transcriptional factors
such as NF-kB, p53 and AP-1 have been shown to be
modulated by oxygen species (reviewed in [13]). Sublethal ROS production, therefore, can interfere with
signal transduction pathways. ROS, in particular H2O2,
are indeed second messengers for various physiological
stimuli, such as, angiotensin inflammatory cytokines
and growth factors or transforming factors [59,60].
There is a paradox surrounding the physiological and
patho-physiological roles played by reactive oxygen
species such as the superoxide radical. It is universally
accepted that the production of superoxide radical by
activated neutrophils and other phagocytes is an essential component of immunological defense mechanisms
against bacteria [61].
Oxidative stress occurs when redox homeostasis
within the cell is altered. It has been suggested that the
global shutdown of mitochondrial function under conditions of oxidative stress could contribute to apoptosis
because of the dramatic decrease in cellular energy
supply. Injury to cells occurs only when the ROS
overwhelm the biochemical defenses of the cell [43,62
65]. Reactive oxygen species, (in particular, hydroxy
radicals) can react with all biological macromolecules,
i.e. lipids, proteins, nucleic acids and carbohydrates.
The initial reaction generates a second radical, which
reacts with a second macromolecule and so on in a
continuing chain reaction. Among the more susceptible
targets are polyunsaturated fatty acids [48,6365].
The polyunsaturated fatty acyl side chains, because
of their susceptibility to oxidative damage in membrane
phopholipids, pose a constant threat to cellular in-

156

K. Kannan, S.K. Jain / Pathophysiology 7 (2000) 153163

Table 2
Diseases that are influenced by oxidative stress or apoptosis
Neurodegenerative diseases (Alzheimers, Parkinsons)
Autoimmune disease (e.g. rheumatoid arthritis)
Human immunodeficiency virus (AIDS)
Diabetes mellitus
Cancers of lung, colon, breast and others
Alcohol induced liver disease
Hepatitis-C induced liver disease
Ischemic reperfusion damage-heart, liver

tegrity and function [62 72]. The lipid radicals generated during the early encounter with an oxidant add
molecular oxygen to produce lipid dioxyl radical. This
pro-oxidant abstracts, an allylic hydrogen from another
unsaturated side chain, producing a lipid hydroperoxide
(LOOH) and thus propagating the chain [62 64]. Iron,
a transition metal, is also well known for its crucial role
in the initiation of new lipid-radical chain reactions
[65].
Lipid peroxidation is an important biological consequence of oxidative cellular damage and aging [6673].
A partial list of conditions or diseases that are likely to
involve oxidative stress is given in Table 2. This includes several neurodegenerative diseases and AIDS.
Other sources of ROS include radiation (e.g. UV), toxic
chemicals (e.g. paraquat and endosulfan) and
chemotherapeutic agents (e.g. adriamycin and
bleomycin) [13,74,75]. The classical view of ROS as
villains that indiscriminately destroy biological macromolecules has undergone a shift, in which, positive
physiological roles are considered as well. In summary,
oxidative stress can have positive responses, such as,
proliferation or activation, as well as negative responses, such as, lipid peroxidation, DNA damage, cell
growth inhibition or cell death.

4. Redox imbalance and apoptosis


Oxidative stress has been implicated in the pathogenesis of several disease processes including, ischemia/
reperfusion injury, Alzheimers, Parkinsons and
diabetes mellitus. In cancer, ROS has been implicated
in damage to DNA resulting in altered gene expression.
Consequently, changes in cell cycle-related protein expression, activation of proto-oncogenes, and the inactivation of some tumor suppressor genes have been
reported [7678]. Yet another modulation that deserves
mention here is the increased expression of Bcl-2
protein that favors prolonged cell survival. Recently, a
therapeutic approach led to the yet incomplete investigation of antisense Bcl-2 oligonucleotides in the treatment of B-cell lymphomas [79].
In colon cancer, it is believed that a mutated p53
gene allows pre-cancerous cells to accumulate tumor-

promoting genetic changes that favor the origin of


tumor cells of malignant type [78]. Interestingly, current
chemotherapeutic agents such as anthracycline-derivatives, which are frequently used as chemotherapeutics in
the treatment of numerous types of cancers, target
some of these apoptotic pathways. For example, adriamycin is known to chelate iron and generate ROS that
result in apoptosis of cancer cells [13,80]. As similar to
cancer, oxidative stress has also been implicated in
AIDS disease [81]. In particular, the bystander effect
in uninfected lymphocytes, in HIV infection, seems to
involve increased apoptosis. This increased cell death
has been linked to the effects of free extracellular gp120
and Tat protein that may have a major impact on the
progression of AIDS [81,82].
Although several risk factors can trigger the development of insulin-dependent diabetes (IDDM), several
studies suggest a role for ROS in beta-cell death and
disease progression [83,84]. In a pancreatic beta-cell
line, stable transfectants of aldose reductase gene induced apoptosis by causing a redox imbalance [85].
Taken together, these observations show that oxidative
stress can negatively modulate the expression of genes
that control carbohydrate metabolism by repressing the
insulin signaling pathway. Accumulating evidence indicates that increased antioxidant defense systems and
ingestion of antioxidants reduce the susceptibility to
IDDM in animal models or in human study [84]
[86,87]. In vitro studies have demonstrated inhibition of
apoptosis by the antisense nucleotide to the p65NF-kB
[88]. Whether the intervention with antisense therapy
can prevent apoptosis and the progression of the disease, it needs further investigation. Gene therapy, therefore, remains a big challenge in translating some of the
recent advances made in molecular biology to patient
care.
Ischemia and reperfusion injury are other manifestations of ROS-mediated injury during surgery [89]. Ischemic injury occurs when the blood supply to an area
of tissue is cut off under physiological conditions, or
during surgery when blood vessels are cross-clamped,
and in organs for transplant. Oxygen deprivation during this period leads to necrotic lesions depending on
duration. On the other hand, restoration of blood
supply leads to reperfusion injury, which is due to
sudden increase of ROS in the target tissue or organ.
Under the in vivo conditions, a complex interplay
between endothelial cells, neutrophils and other cells of
immune system occurs after anoxia-reoxygenation.
Some of the consequences of this interaction are the
oxidative cell damage including lipid peroxidation, activation of inflammatory cytokines, followed by neutrophil and macrophage attack on the reperfused tissue.
A second consequence being accelerated apoptotic cell
death in these tissues resulting in various clinical complications [90]. Oxidative stress can also repress the

K. Kannan, S.K. Jain / Pathophysiology 7 (2000) 153163

activity of T-lymphocytes, and thus alter the immune


response. Depletion of intracellular GSH pool elicits
oxidative stress by raising the intracellular redox potential, and causes a marked inhibition of the T cell-mediated immune response. In particular, IL-2 expression,
an important cytokine, is decreased by oxidative stress.
This could contribute to the alteration of protective
immune response as in patients suffering from rheumatoid arthritis and HIV-infected people [13,91].

5. Genes, oxidative stress and apoptosis


Gene expression, in general, is modulated by both
physiological and environmental stimuli. It appears that
oxidative stress acts as a pleiotropic modulator in both
these pathways. Indeed, ROS have been described as
second messengers for several growth factors and cytokines. It has been shown that the transcription factors
such as NF-kB and AP-1, which are stimulated by
ROS, could mediate such inductions [13]. Most of the
literature has historically focused on gene induction and
less on gene repression by ROS. Under conditions of
mild oxidative stress, cell cycle-related genes are repressed to increase the lengthening of G1-phase. A cell
cycle arrest is required in order to assess the amount of
macromolecule alterations, and, if necessary, to enter
the apoptotic pathway instead of carrying on the cellular division process. This delay in cell cycle is important, since a base alteration could be converted into an
irreversible mutation if a mismatch escaped the repair
systems before replication. Hence, there is a necessity
not to activate S-phase too quickly when a cellular
stress occurs.
As compared with genomic DNA, mitochondrial
genome appears to be more sensitive to oxidative damage. The mitochondrion possesses its own genome and
produces its own RNAs that are necessary to its function. Crawford et al., in 1998 [92] have shown that
mitochondrial RNAs undergo specific degradation
upon oxidative stress. Similarly, a direct relationship
between glutathione oxidation and mtDNA damage in
apoptosis has been suggested by Esteve and co-workers
[93]. Mitochondrial DNA is also heavily damaged by
ROS at the bases, as indicated by the high steady-state
level of 8-hydroxydeoxyguanosine, i.e. the presence of
which causes mispairing and point mutations [94].
These mutations can be detected by long-extension
PCR, a method for detecting a variety of mutations of
mitochondrial genome [95]. In mammalian cells, UVB
radiation has been reported to repress mitochondrial
function by strongly inhibiting its transcription [96].
Following the treatment of hamster fibroblasts with
H2O2, these authors observed that the 16S rRNAs, a
major component of mitochondrial ribosomes, were
specifically degraded, whereas cytosolic mRNAs were

157

not. This resulted in a dramatic shutdown of mitochondrial protein biosynthesis. Transcription factors such as
NF-kB, p53 and AP-1 are also sensitive to redox
changes in mammalian cells, primarily through redox
regulation of their DNA binding regions [13,84,88,97].
Cell death in the nematode Caenorhabditis elegans is
undoubtedly the best system for the study the genetics
of programmed cell death. During development, a process resembling apoptosis deletes 131 cells out of 1090
cells of this nematode. A total of 14 suicide genes that
are important for development have been identified so
far, of which three genes Ced3, Ced4 and Ced9
have received most attention. The Ced9 gene negatively
regulates the Ced3 and Ced4 genes [1,35,9799]. The
homologues of Ced9 in humans have been identified as
Bcl-2, which protect mammalian cells. Caspase 3
(YAMA/CPP32), a proteolytic enzyme in humans is
believed to be an equivalent of Ced3 in nematode. The
homologue to Ced4 in human has been identified to be
APAF-1 (apoptotic protease-activating factor-1) [35].
Other genes of importance are c-fos, c-jun as well as
c-myc, p53, clusterin, RP-2 and RP-8 [100,101]. However, the complex mechanisms controlling gene expression that determines cell survival and cell death are still
unresolved.

6. Bcl-2 family members and apoptosis


Bcl-2 is the first mammalian regulator gene that was
identified to have anti-apoptotic potential in a variety
of cell systems. At least 15 Bcl-2 family members have
been identified in mammalian cells [107]. All members
possess at least one of four conserved motifs known as
Bcl-2 homology domains (BH1BH4). Generally
speaking, family members that act as inhibitors of cell
death harbor at least three domains (BH1, BH2, and
BH3), which are important for proteinprotein interaction and the suppression of apoptosis, whereas BH3
serves as the minimal death domain in the prop-apoptotic members studied so far [11]. Some of the family
members, such as Bax, Bak, and Bok, work in a
manner opposite to that of Bcl-2, though they resemble
Bcl-2 closely. Pro- and anti-apoptotic family members
of Bcl-2 can homodimerize or heterodimerize and seemingly can neutralize one anothers function, suggesting
that their relative concentration in the given cell may
act as a rheostat for the suicide program. Bcl-2 is
localized on the cytoplasmic face of the mitochondrial
outer membrane, endoplasmic reticulum and nuclear
envelope. It is currently believed that this strategic
localization allows it to register and counterbalance the
oxidative damage done to these compartments and
affect their behavior [19,108,109]. This pro-survival
protein also seems to maintain the membrane integrity
of mitochondria, by directly or indirectly preventing the

158

K. Kannan, S.K. Jain / Pathophysiology 7 (2000) 153163

release of cytochrome C, which, along with dATP and


APAF-1, facilitates the activation of pro-caspase 9 to
its active form of caspase 9 [11,108,109]. Bcl-2 protects
cells against diverse cytotoxic insults, for example,
gamma radiation, cytokine withdrawal, hypoxia, ROS,
dexamethasone, staurosporine, and cytotoxic drugs. All
pro-survival Bcl-2-like genes are potentially oncogenic,
whereas pro-apoptotic family members may act as tumor suppressors. Clarifying how Bcl-2 family members
govern apoptosis under various conditions might
provide clues for the development of novel therapies in
clinical settings.

7. Oxidative stress and mitochondria


The mitochondria is sensitive to changes in the redox
state of the cell. Several studies have shown that the
global shutdown of mitochondrial function under conditions of oxidative stress could contribute to apoptosis
[9 13]. Maintenance of mitochondrial membrane integrity is a dynamic process. Under severe oxidative
stress, the mitochondrial permeability transition (PT)
occurs. PT involves a sudden increase of the inner
mitochondrial membrane permeability to solutes
greater than 1500 Da (protons, calcium, GSH etc.). It is
currently believed that PT functions as a voltage sensor,
a thiol sensor, a sensor of oxidation reduction equilibrium of adenine nucleotide pool, and as a sensor of
divalent cations. As a consequence, defective PT pore
opening to larger molecules causes uncoupling of the
respiratory chain resulting in hypergeneration of ROS,
cessation of ATP synthesis, matrix Ca++ outflow and
depletion of reduced glutathione and other reductants.
Following the inner membrane permeability and the
release of matrix solutes, a colloidal osmotic pressure
arises in the mitochondrial matrix due to the high
concentrations of proteins, which are slow to equilibrate [101103]. In order to correct the osmotic balance, the diffusion of H2O results in a massive swelling
of the mitochondria [102]. This change in membrane
potential predisposes these cells to oxidative damage by
impairment of endogenous antioxidant defense mechanisms [4956]. Another crucial step in the early changes
in mitochondria is alteration of mitochondrial
transmembrane potential (DCm) [9 13]. A decline in
the DCm alone can induce oxidative stress and cell
death [12]. The measurement of cell respiration, performed with whole cell populations, showed that the
lower DCm correlates, as expected, with an uncoupling
of electron transport from ATP production [104,105].
These events are detected at early stages of the apoptotic process before most of the cells are irreversibly
committed to death suggesting that mitochondria could
be a primary target during apoptosis. Recently, the

release of cytochrome C from mitochondria has been


shown as another important effector molecule in the
mediation of programmed cell death [12,56,106]. It is
postulated that cytochrome C is somehow able to interact with pre-existing cytoplasmic factor(s), which subsequently mediate the cleavage of zymogens and resulting
in the activation of caspase 9.
Permeability transition is also associated with the
release of apoptotic inducing factor (AIF), which has
recently been identified and characterized [11,53,54].
This is a  50 kDa protein located in the inner mitochondrial membrane whose functions are inhibited by
Bcl-2. Under in vitro conditions, purified AIF induces
dose and temperature-dependent apoptosis in isolated
nuclei. This is an important finding in that membrane
permeability transition predisposes the mitochondria to
lose protein thiols and other factors including AIF to
the cytosol for the initiation of a biochemical cascade
resulting in programmed cell death [11]. It is not clear
at this time whether the release of AIF and cytochrome
C is interrelated, or occurs as biochemical events of
separate pathways. In both instances, Bcl-2, an antiapoptotic protein, prevented the actions of AIF and
cytochrome C release. This partly explains the importance of the physical location of Bcl-2 on the outer
membrane of the mitochondria, nuclear envelope and
endoplasmic reticulum [53,54,110]. A schematic representation of the association between oxidative stress
and apoptosis is presented in Fig. 1.
Calcium is another important mediator of apoptosis
[111113]. Also, a close relationship exists between
calcium status and oxidative stress in many isolated cell
systems, the best example being isolated rat hepatocytes
[114]. When cells are deprived of calcium, mitochondria
become sensitive to this calcium-deficient state and their
calcium regulatory mechanism is disturbed. Calcium
omission results in a marked decline of mitochondrial
transmembrane potential (DCm), leading to oxidative
stress and induction of programmed cell death. Many
of the antioxidants can prevent this collapse, thus indicating that the DCm is critical to the process and is
reversible in the early phases. However, it remains to be
determined what dictates the efflux of mitochondrial
calcium. It must be emphasized here that oxidative
stress induced by the absence of extracellular calcium is
distinct from that generated by ROS, in that it appears
to induce calcium cycling in and out of mitochondrial
matrix, which serves to promote oxidative stress
[15,64,115]. In the human erythrocyte model, pretreatment of erythrocytes with calcium increases their sensitivity to calcium [115]. Thus, it appears that calcium
can promote lipid peroxidation both under conditions
of excess and of deficiency. Calcium homeostasis is,
thus, vital to the function and survival of the cell [116].

K. Kannan, S.K. Jain / Pathophysiology 7 (2000) 153163

8. Mitochondria and apoptosis


While the central role of mitochondria in apoptosis
has been widely accepted, experimental evidence now
points to the existence of yet another pathway independent of mitochondria. However, it appears that both
these pathways involve pro-caspase 8 as one of the
important intermediary step. Using the Xenopus cellfree system as the experimental model, Kuwana et al.,

159

in 1998 [117] have provided experimental evidence that


suggests that caspase 8 participates in both pathways.
In the absence of mitochondria, activation of a caspase
cascade by caspase 8 produces only a partial apoptotic
phenotype in nuclei added to the Xenopus cell extract.
In contrast, the mitochondria-independent pathway,
which involves the release of cytochrome C from mitochondria into cytosol, triggers full nuclear apoptosis.
Moreover, engagement of the mitochondria provides an

Fig. 1. Schematic model of mammalian cell death pathway. Death signal may be delivered at the cell surface by direct ligand-receptor interaction.
This followed by clustering of death receptors and activation of caspase-8. Alternatively, cytotoxic drugs, ionizing radiation may directly activate
caspase-9, otherwise a later event. Mitochondria plays a central role in apoptosis by releasing apoptogenic factors and proteases into the cytosol.
A major checkpoint in this pathway is the ratio between pro-apoptotic (Bax) to anti-apoptotic (Bcl-2) members. Miitochondrial dysfunction
includes PT, change in mitochondrial membrane potential, production of ROS, release of cytochrome C, AIPF-1 and caspases-2, -3, and -9 into
the cytosol, where they form multimeric complexes. This activates downstream caspases and degradation of death substrates in the nucleus, which
ultimately leads to cell death.

160

K. Kannan, S.K. Jain / Pathophysiology 7 (2000) 153163

efficient means of amplifying the apoptotic signal transduced by caspase 8 even under very low concentration.
In the absence of mitochondria, high concentrations of
caspase 8 were required to activate downstream caspase
cascade. Interestingly, caspase 8 can activate the mitochondria-dependent pathway even when the Bcl-2
protein is present. This helps explain the failure of Bcl-2
to inhibit CD95-dependent apoptosis consistently [117].
Scaffidi et al., in 1998 [118] have identified two types of
CD95-mediated cell death. Recently, it has been
demonstrated that Apaf-1 / and caspase 9 / T
cells remain sensitive to Fas-induced killing [119,120].
However, Fas-induced apoptosis was markedly reduced
in embryonic fibroblasts with Apaf-1 / phenotype
[121], suggesting Fas (CD95) can activate different
pathways in different cell types. These observations in
T cells indicate that the apoptotic function of mitochondria (at least the cytochrome C release part) can be
bypassed in these cells.
Other less defined pathways that lead to apoptosis
are the ones initiated by potent cytotoxic chemicals or
gamma irradiation. These pathways seem to rely on the
apoptotic function of mitochondria, since cells with
Apaf-1 / and caspase-9 / phenotypes are principally resistant to these death-inducing agents [119
121]. Further research in this area may help us to better
understand some of the missing links.

9. Conclusions
The role of oxidative stress as a mediator of apoptotic cell death in diverse cell systems is now better
understood. In recent years, mitochondria have gained
considerable importance both as a site for ROS production and as a major player in apoptosis. The involvement of mitochondria or ROS in apoptosis is not
without controversy. In some experimental systems,
apoptosis can occur at very low oxygen tension, when
ROS are unlikely to occur. The protooncogene
product, Bcl-2, can inhibit apoptosis both in the presence and in the absence of reactive oxygen products.
Further research is needed to clarify the underlying
mechanisms.
Recent developments suggest that mtDNA may be a
good indicator of oxidative damage in apoptosis. It is
becoming apparent that the redox status of a cell can
have a complex and multilayered effect on cell survival
and cell death. Future investigations will unravel
whether oxidative stress is a pre-requisite for all apoptotic events. This would be an attractive mechanism
whereby a panoply of seemingly diverse injuries could
rapidly converge on common apoptotic pathways.
More in-depth studies are needed to elucidate the role
of BH3-domain-only subset of molecules and their interplay with ROS. This could open new sites of drug

targeting. Future studies may also improve our understanding whether the oxidation states of mitochondrial
thiols alone function as a prominent apoptotic sensor.
A better understanding of the molecular machinery of
apoptosis in the pathophysiology will undoubtedly
provide novel therapeutic interventions and care for a
large number of patients with chronic diseases.

Acknowledgements
We thank Georgia Morgn First for the critical reading of the manuscript. This work was supported in part
by the funds by Feist Weiller Cancer Center and the
Center of Excellence for Arthritis and Rheumatology of
the Louisiana State University Health Sciences Center
at Shreveport, and a grant-in aid from the American
Heart Association (Southeast).

References
[1] R.A. Schwartzman, L.A. Cidlowski, Apoptosis: the biochemistry and molecular biology of programmed cell death, Endocrine Rev. 14 (1993) 133 151.
[2] C.B. Thompson, Apoptosis in the pathogenesis and treatment
of disease, Science 267 (1995) 1456 1462.
[3] L.F.R. Kerr, C.M. Winterford, B.N. Harmon, Apoptosis: its
significance in cancer and cancer therapy, Cancer 73 (1994)
2013 2026.
[4] R.C. Duke, D.M. Ojcius, D.E. Young, Cell suicide in health
and disease, Sci. Am. 275 (1996) 80 87.
[5] L.F.R. Kerr, A.H. Wyllie, A.R. Currie, Apoptosis: a basic
biological phenomenon with wide ranging implications in tissue
kinetics, Br. J. Cancer 26 (1972) 239 257.
[6] Editorial: Cell Death in Us and Others. Science 281 (1998)
1283.
[7] T.M. Burtke, P.A. Sandstrom, Oxidative stress as a mediator of
apoptosis, Immunol. Today 15 (1994) 7 10.
[8] G.S. Salvesen, V.M. Dixit, Caspase activation: the inducedproximity model, Proc. Natl. Acad. Sci. USA 96 (1999) 10964
10967.
[9] N. Zamzami, P. Marchetti, M. Castedo, D. Decaudin, A.
Macho, T. Hirsch, S.A. Susin, P.X. Petit, B. Mignotte, G.
Kroemer, Sequential reduction of mitochondrial transmembrane potential and generation of reactive oxygen species in
early programmed cell death, J. Exp. Med. 182 (1995) 367377.
[10] N. Zamzami, S.A. Susin, P. Marchetti, T. Hirsch, I. Gomez
Monterrey, M. Castedo, G. Kroemer, Mitochondrial control of
nuclear apoptosis, J. Exp. Med. 183 (1996) 1533 1544.
[11] D.R. Green, J.C. Reed, Mitochondria and apoptosis, Science
281 (1988) 1309 1311.
[12] S.A. Susin, N. Zamzami, G. Kroemer, Mitochondria as regulators of apoptosis; doubt no more, Biochim. Biophys. Acta 1366
(1998) 151 165.
[13] Y. Morel, Barouki. Repression of gene expression by oxidative
stress, Biochem J (1999) 342: 481 496.
[14] A. Fernandez, J. Kiefer, L. Fosdick, D.J. McConkey, Oxygen
radical production and thiol depletion are required for Ca2 + mediated endogenous endonuclease activation in apoptotic thymocytes, J. Immunol. 155 (1995) 5133 5139.

K. Kannan, S.K. Jain / Pathophysiology 7 (2000) 153163


[15] C. Richter, Pro-oxidants and mitochondrial Ca2 + and their
relationship to apoptosis and oncogenesis, FEBS 325 (1993)
104 107.
[16] J.Z. Byczkowski, T. Gessner, Biological role of superoxide ion
radical, Int. J. Biochem. 20 (1988) 569580.
[17] I. Fridovich, Superoxide radical: an endogenous toxicant, Ann.
Rev. Pharmacol. Toxicol. 23 (1983) 239257.
[18] A.J. MeGowan, R.S. Fernandes, A. Samali, T.G. Cotter, Antioxidants and apoptosis, Biochem. Soc. Trans. 24 (1996) 229
267.
[19] D.M. Hockenberry, Bcl-2, a novel regulator of cell death,
BioEssays 17 (1995) 631638.
[20] S.J. Korsmeyer, X. Yin, Z.N. Oltvai, D.J. Veis-Novack, G.P.
Linette, Reactive oxygen species and the regulation of cell death
by the Bcl-2 gene family, Biochim. Biophys. Acta 1271 (1995)
63 66.
[21] D.M. Hockenbery, Z.N. Oltvai, X.M. Yin, C.L. Millman, S.J.
Korsmeyer, Bcl-2 functions in an antioxidant pathway to prevent apoptosis, Immunol. Today 18 (1993) (1993) 4451.
[22] D.W. Voehringer, Bcl-2 and glutathione: alterations in cellular
redox state that regulate apoptosis sensitivity, Free Rad. Biol.
Med. 27 (1999) 945950.
[23] G. Kroemer, N. Zamzami, S.A. Susin, Mitochondrial control
of apoptosis, Immunol. Today 18 (1997) 4451.
[24] S. Nagata, Fas-mediated apoptosis, Adv. Exp. Med. Biol. 406
(1996) 119 124.
[25] H. Hsu, H.B. Shu, M.G. Pan, D.V. Goeddel, TRADD-TRAF2
and TRADD-FADD interactions define two distinct TNF receptor 1 signal transduction pathways, Cell 84 (1996) 299 308.
[26] R. Schwandner, K. Wiegmann, K. Bernardo, D. Kreder, M.
Kronke, TNF receptor death domain-associated proteins
TRADD and FADD signal activation of acid sphingomyelinase, J. Biol. Chem. 273 (1998) 59165922.
[27] L. Kitson, T. Raven, Y.P. Jiang, D.V. Goeddel, K.M. Giles,
K.T. Pun, C.J. Grinham, R. Brown, S.N. Farrow, A death-domain-containing receptor that mediates apoptosis, Nature 384
(1996) 372 375.
[28] A. Ashkenazi, V.M. Dixit, Death receptors: signaling and modulation, Science 281 (1998) 13051308.
[29] P. Schneider, M. Thome, K. Burns, L.L. Bodmer, K. Hofmann,
T. Kataoka, N. Holler, J. Tschopp, TRAIL receptors 1 (DR4)
and 2 (DR5) signal FADD-dependent apoptosis and activate
NF-kappa-B, Immunity 7 (1997) 831836.
[30] I.E. Wertz, M.R. Hanley, Diverse molecular provocation of
programmed cell death, Trends Biochem. Sci. 21 (1996) 359
364.
[31] S. Lederman, A.M. Cleary, M.J. Yellin, D.M. Frank, M.
Karpusas, D.W. Thomas, L. Chess, The central role of the
CD40-ligand and CD40 pathway in T-lymphocyte-mediated
diffirentiation of B lymphocytes, Curr. Opin. Hematol. 3 (1996)
77 86.
[32] G.G. Klaus, M.S. Choi, E.W. Lam, C. Johnson-Leger, J. Cliff,
CD40: a pivotal receptor in the determination of life/death
decisions in B lymphocytes, Int. Rev. Immunol. 15 (1997) 5 31.
[33] Y. Du, K.R. Bales, R.C. Dodel, E. Hamilton-Byrd, L.W. Horn,
D.L. Czilli, L.K. Simmons, B. Ni, S.M. Paul, Activation of a
caspase 3-related cysteine protease is required for glutamatemediated apoptosis of cultured cerebellar granule neurons,
Proc. Natl. Acad. Sci. USA 94 (1997) 1165711662.
[34] A. Gross, J.M. McDonnell, S.J. Korsmeyer, Bcl-2 family members and the mitochondria in apoptosis, Genes Develop. 13
(1999) 1899 1911.
[35] N.A. Thornberry, Y. Lazebnik, Caspases: enemies within, Science 281 (1998) 13121316.
[36] M.B. Hampton, S. Orrenius, Redox regulation of apoptotic cell
death in the immune system, Toxicol. Lett. 102-103 (1998)
355 358.

161

[37] E.S. Alnemri, D.J. Livingston, D.W. Nicholson, G. Salvesen,


N.A. Thomberry, W.W. Wong, J. Yuan, Human ICE/CED-3
protease nomenclature, Cell 87 (1996) 171.
[38] S.L. Martin, D.R. Green, Protease activation during apoptosis:
death by a thousand cuts?, Cell 82 (1995) 349 352.
[39] B. Zhivotovsky, D.H. Burgess, D.M. Vanags, S. Orrenius,
Involvement of proteolytic machinery in apoptosis, Biochem.
Biophys. Res. Comm. 230 (1997) 481 488.
[40] L. Casciola-Rosen, D.W. Nicholson, T. Chong, K.R. Rowan,
N.A. Thornberry, D.X. Miller, A. Rosen, Apopain/CPP32
cleaves proteins that are essential cellular repair: a fundamental
principle of apoptotic death, J. Exp. Med. 183 (1996) 1957
1964.
[41] B. Zhivotovsky, A. Gahm, M. Ankarcrona, P. Nicotera, S.
Orrenius, Multiple proteases are involved in thymocyte apoptosis, Exp. Cell. Res. 221 (1995) 404 412.
[42] P.A. Henkart, ICE family proteases: mediators of all apoptotic
cell death?, Immunity 4 (1996) 195 201.
[43] H. Sies, Oxidative Stress, Academic Press, London, 1985.
[44] N. Sato, S. Iwata, K. Nakamura, T. Hori, K. Mori, J. Yodoi,
Thiol-mediated redox regulation of apoptosis. Possible roles of
cellular thiols other than glutathione in T cell apoptosis, J.
Immunol. 154 (1995) 3194 3203.
[45] L.P. Kehrer, L.G. Lund, Cellular reducing equivalents and
oxidative stress, Free Radic. Biol. Med. 17 (1994) 65 75.
[46] D.J. Reed, M.K. Savage, Influence of metabolic inhibitors on
mitochondrial permeability transition and glutathione status,
Biochim. Biophys. Acta 1271 (1995) 43 50.
[47] A. Kowaltowski, A.E. Vercesi, Mitochondrial damage induced
by conditions of oxidative stress, Free Radic. Biol. 26 (1999)
463 471.
[48] B Halliwell, J.M.C. Gutteridge, Free Radicals in Biology and
Medicine, Oxford University Press, Oxford, 1989.
[49] P.X. Petit, S.A. Susin, N. Zamzami, B. Mignotte, G. Kroemer,
Mitochondria and programmed cell death: back to the future,
FEBS Lett. 396 (1996) 7 13.
[50] P. Marchetti, M. Castedo, S.A. Susin, N. Zamzami, T. Hirsch,
A. Macho, A. Haeffner, F. Hirsch, M. Geuskens, G. Kroemer,
Mitochondrial permeability transition is a central coordinating
event of apoptosis, J. Exp. Med. 184 (1996) 1155 1160.
[51] M. Castedo, A. Macho, N. Zamzami, T. Hirsch, P. Marchetti,
J. Uriel, G. Kroemer, Mitochondrial perturbations define
lymphocytes undergoing apoptotic depletion in vivo, Eur. J.
Immunol. 25 (1995) 3277 3284.
[52] N. Zamzami, T. Hirsch, B. Dallaporta, P.X. Petit, G. Kroemer,
Mitochondrial implication in accidental and programmed cell
death: apoptosis and necrosis, J. Bioenerg. Biomembr. 29
(1997) 185 193.
[53] S.A. Susin, N. Zamzami, M. Castedo, T. Hirsch, P. Marchetti,
A. Macho, E. Daugas, M. Geuskens, G. Kroemer, Bcl-2 inhibits the mitochondrial release of an apoptogenic protease, J.
Exp. Med. 184 (1996) 1331 1341.
[54] I. Marzo, S.A. Susin, P.X. Petit, L. Ravagnan, C. Brenner, N.
Larochette, N. Zamzami, G. Kroemer, Caspases disrupt mitochondrial membrane barrier function, FEBS Lett. 427 (1998)
198 202.
[55] L.L. Scarlett, M.P. Murphy, Release of apoptogenic proteins
from the mitochondrial intermembrane space during the mitochondrial permeability transition, FEBS Lett. 418 (1997) 282
286.
[56] J.C. Yang, G.A. Cortopassi, 1998. Induction of the mitochondrial permeability transition causes release of the apoptogenic
factor cytochrome C, Free Radic. Biol. Med. 24 (1998) 624-631.
[57] Y.J. Suzuki, H.J. Forman, A. Sevanian, Oxidants as stimulators of signal transduction, Free Radic. Biol. Med. 22 (1997)
269 285.

162

K. Kannan, S.K. Jain / Pathophysiology 7 (2000) 153163

[58] E. Pennesi, Superoxide relay Ras proteins oncogenic message,


Science 275 (1997) 15671568.
[59] M. Ushio-Fukai, R.W. Alexander, M. Akers, K.K. Griendling,
p38 Mitogen-activated protein kinase is a critical component of
the redox-sensitive signaling pathways activated by angiotensin
II. Role in vascular smooth muscle cell hypertrophy, J. Biol.
Chem. 273 (1998) 1502215029.
[60] Y.S. Bae, S.W. Kang, M.S. Seo, I.C. Bainers, E. Tekle, P.B.
Chock, S.G. Rhee, Epidermal growth factor (EGF)-induced
generation of hydrogen peroxide. Role in EW receptormediated
tyrosine phosphorylation, J. Biol. Chem. 272 (1997) 217 221.
[61] B.M. Babier, Oxygen-dependent microbial killing by phagocytes, New Engl. J. Med. 298 (1978) 659668.
[62] A.A. Horton, S. Fairhurst, Lipid peroxidation and mechanisms
of toxicity, CRC Crit. Rev. Toxicol. 18 (1987) 2779.
[63] H. Jaescheke, Mechanisms of oxidant stress-induced acute tissue injury, Proc. Soc. Exp. Biol. Med 209 (1995) 104111.
[64] D.J. Reed, Toxicity of oxygen, in: F. De Matteis, L.L Smith
(Eds.), Molecular and Cellular Mechanisms of Toxicity, CRC
Press, New York, 1995, pp. 3568.
[65] H. Sies, Biochemistry of oxidant stress, Agrew. Chem. Int. Ed.
25 (1986) 1058 1071.
[66] K. Kannan, S.K. Jain, Effect of high glucose on cellular
proliferation and lipid peroxidation in cultured Vero cells,
Horm. Metab. Res. 26 (1994) 322325.
[67] S.K. Jain, The accumulation of Malonyldialdehyde, a product
of fatty acid peroxidation, can disturb aminophospholipid organization in the membrane bilayer of human erythrocytes, J.
Biol. Chem. 25 (1984) 33913394.
[68] S.K. Jain, In vivo externalization of phosphatidylserine and
phosphatidylethanolamine in the membrane bilayer and hypercoagulability by the lipid peroxidation of erythrocytes in rats, J
Clin. Invest. 76 (1985) 281286.
[69] S.K. Jain, The neonatal erythrocyte and its oxidative susceptibility, Semin. Hernatol. 26 (1989) 286300.
[70] S.K. Jain, Evidence for membrane lipid peroxidation during the
in vivo aging of human erytbrocytes, Biochim. Biophys. Acta
937 (1988) 205 210.
[71] S.K. Jain, K. Kannan, G. Lim, Ketosis (acetoacetate) can
generate oxygen radicals and cause increased lipid peroxidation
and growth inhibition in human endothelial cells, Free Radic.
Biol. Med. 25 (1998) 10831088.
[72] R.J. Reiter, J.M. Guerrero, J.J. Garcia, D. Acuna-Castroviejo,
Reactive oxygen intermediates, molecular damage, and aging.
Relation to melatonin, Ann. N.Y.Acad. Sci. 854 (1998) 410
424.
[73] Y. Christen, Oxidative stress and Alzhieimer disease, Am. J.
Clin. Nutr. 71 (2000) 621S629S.
[74] K. Kannan, R.F. Holcombe, S.K. Jain, R. Chervenak, R.E.
Wolf, J. Glass, Evidence for the induction of apoptosis by
endosulfan in a human T-cell leukemic line, Mol. Cell Biochem.
205 (2000) 53 66.
[75] H. Kappus, Oxidative stress in chemical toxicity, Arch. Toxicol.
60 (1987) 144 149.
[76] J.M. Mates, F.M. Sanchez-Jimenez, Role of reactive oxygen
species in apoptosis: implications for cancer therapy, Int. J.
Biochem. Cell Biol. 32 (2000) 157170.
[77] S. Ambs, S.P. Hussain, A.J. Maarrogi, C.C. Harris, Cancerprone oxyradical overload disease, IARC Sci. Publ. 150 (1999)
295 302.
[78] J.M. Lee, A. Bernstein, Apoptosis, cancer and the p53 tumour
suppressor gene, Cancer Metastasis Rev. 14 (1995) 149 161.
[79] F.E. Cotter, Antisense therapy of hematologic malignancies,
Semin. Hematol. 36 (1999) 914.
[80] I. Muller, D. Niethammer, G. Bruchelt, Anthracycline-derived
chemotherapeutics in apoptosis and free radical cytotoxicity,
Int. J. Mol. Med. 1 (1998) 491494.

[81] D. Romero-Alvira, E. Roche, The keys of oxidative stress in


acquired immune deficiency syndrome apoptosis, Med. Hypotheses 51 (1998) 169 173.
[82] S.R. Bartz, M. Emerman, Human immunodeficiency virus type
1 Tat induces apoptosis and increases sensitivity to apoptotic
signals by up-regulating FLICE/caspase-8, J. Virol. 73 (1999)
1956 1963.
[83] O. Guigliano, A. Ceriello, Oxidative stress and diabetic vascular
complications, Diab. Care 19 (1996) 257 267.
[84] E. Ho, T.M. Bray, Antioxidants, NFkappaB activation, and
diabetogenesis, Proc. Soc. Exp. Biol. Med. 222 (1999) 205213.
[85] R. Hamaoki, J. Fujii, J. Miyagawa, M. Takahashi, M. Kishimoto, M. Moriwaki, K. Yamamoto, Y. Kajimoto, Y. Yamasaki, T. Hanafusa, Y. Matsuzawa, N. Taniguchi,
Overexpression of the aldose reductase gene induces apoptosis
in pancreatic beta-cells by causing redox imbalance, J. Biochem.
(Tokyo) 126 (1999) 41 47.
[86] R.A. Kowluru, R.L. Engerman, T.S. Kern, Diabetes-induced
metabolic abnormalities in myocardium: effect of antioxidant
therapy, Free Radic. Res. 32 (2000) 67 74.
[87] H. Kaneto, Y. Kajimoto, J. Miyagawa, T. Matsuoka, Y. Fujitani, Y. Umayahara, T. Hanafusa, Y. Matsuzawa, Y. Yamasaki, M. Hori, Beneficial effects of antioxidants in diabetes:
possible protection of pancreatic b-cells against glucose toxicity,
Diabetes 48 (1999) 2398 2406.
[88] X. Du, K. Stockklauser-Farber, P. Rosen, Generation of reactive oxygen intermediates, activation of NF-KB and induction
of aoptosis in human endothelial cells by glucose: role of nitric
oxide synthase, Free Radic. Biol. Med. 27 (1999) 752763.
[89] D.L. Carden, D.N. Granger, Pathophysiology of ischeamiareperfusion injury, J. Pathol. 190 (2000) 255 266.
[90] G.L. Semenza, Cellular and molecular dissection of reperfusion
injury: ROS within and without, Circ. Res. 86 (2000) 117118.
[91] A.C. WaJsh, S.G. Michaud, L.A. Malossi, D.A. Lawrence,
Glutathione depletion in human T lymphocytes: analysis of
activation-associated gene expression and the stress response,
Toxicol. Appl. Pharmacol. 133 (1995) 249 261.
[92] D.R. Crawford, N.E. Abramova, K.J. Davies, Oxidative stress
causes a general, calcium-dependent degradation of mitochondrial polynucleotides, Free Radic. Biol. Med. 25 (1998) 1106
1111.
[93] J.M. Esteve, J. Mompo, J. Garcia, J.G.D. Asuncion, J. Sastre,
M. Asensi, J. Boix, J.R. Vina, F.V. Pallardo, Oxidative damage
to mitochondrial DNA and glutathione oxidation in apoptosis:
studies in vivo and in vitro, FASEB J. 13 (1999) 10551064.
[94] C. Richter, Reactive oxygen and DNA damage in mitochondria, Mutat. Res. 275 (1992) 249 255.
[95] D.C. Wallace, Mitochondrial diseases in man and disease,
Science 283 (1999) (1999) 1482 1487.
[96] T.M. Vogt, L. Welsh, W. Stolz, E. Kullmann, B. Jung, N.
Landthaler, M. McClelland, RNA fingerprinting displays UVBspecific disruption of transcriptional control in human
melanocytes, Cancer Res. 57 (1997) 3554 3561.
[97] G.T. Williams, C.A. Smith, Molecular regulation of apoptosis:
genetic controls on cell death, Cell 74 (1993) 777 779.
[98] H. Steller, Mechanisms and genes of cellular suicide, Science
267 (1995) 1445 1448.
[99] A.M. Chinnaiyan, K. ORourke, B.R. Lane, V.M. Dixit, Interaction of CED-4 with CED-3 and CED-9: A molecular framework for cell death, Science 275 (1977) 1122 1126.
[100] M. Oren, The involvement of oncogenes and tumor suppressor
genes in the control of apoptosis, Cancer Metastasis Rev. 11
(1992) 141 148.
[101] U. lgbavboa, C.N. Zwizinski, D.R. Pfeiffer, Release of mitochondrial matrix proteins through a Ca2 + requiring, cyclosporin-sensitive pathway, Biochem. Biophys. Res. Commun.
161 (1989) 619 625.

K. Kannan, S.K. Jain / Pathophysiology 7 (2000) 153163


[102] T.E. Gunter, D.R. Pfeiffer, Mechanisms by which initochondria
transport calcium, Am. J. Physiol. 258 (1990) C755C786.
[103] C. Richter, V. Gogvadze, R. Laffranchi, R. Schlapbach, M.
Schweizer, M. Suter, R. Walter, M. Yaffee, Oxidants in mitochondria: from physiology to disease, Biochim. Biophys. Acta
1127 (1995) 67 74.
[104] M. Leist, B. Single, A.F. Castoldi, S. Kuhnle, P. Nicotera,
Intracellular adenosine triphosphate (ATP) concentration: a
switch in the decision between apoptosis and necrosis, J. Exp.
Med. 185 (8) (1997) 14811486.
[105] C. Richter, M. Schweizer, A. Cossarizza, C. Franceschi, Control of apoptosis by the cellular ATP level, FEBS Lett. 378
(1996) 107 110.
[106] M. Oren, Relationship of p53 to the control of apoptotic cell
death, Semin. Cancer Biol. 5 (1994) 221227.
[107] L.M. Adams, S. Cory, The Bcl-2 protein family: arbiters of cell
survival, Science 281 (1998) 13221325.
[108] B. Antonsson, E. Conti, A. Ciavatta, S. Montessuit, et al.,
Inhibition of Bax channel forming activity by Bcl-2, Science 277
(1997) 370 372.
[109] E. Yang, S.J. Korsmeyer, Molecular thanatopsis: a discourse
on the Bcl-2 family and cell death, Blood 88 (1996) 386 401.
[110] R.M. Kluck, E. Bossy-Wetzel, D.R. Green, D.D. Newmeyer,
The release of cytochrome c from mitochondria: a primary site
for Bcl-2 regulation of apoptosis, Science 275 (1997) 1132
1136.
[111] L. Soabo, M. Zoratti, The mitochondrial megachannel is the
permeability transition pore, J. Bioenerg. Biomembr. 24 (1992)
111 117.
[112] L. Al-Nasser, M. Crompton, The reversible Ca2 + induced
permeabilization of rat liver mitochondria, Biochem. J. 239
(1986) 19 29.
[113] V. Petronilli, C. Cola, P. Bernardi, Modulation of the mitochondrial cyclosporin Asensitive permeability transition

[114]

[115]

[116]

[117]

[118]

[119]

[120]

[121]

163

pore. II. The minimal requirements for pore induction underscore a key role for transmembrane electrical potential, matrix
pH, and matrix Ca2 + , J. Biol. Chem. 268 (1993) 10111016.
P.L. Gutierrez, N.R. Bachur, Free radicals in quinone containing antitumor agents. The nature of the diaziquone (3,6-diaziridinyl-2,5-bis(carboethoxyamino)-1,4-benzoquinone)
free
radical, Biochim. Biophys. Acta 758 (1983) 37 41.
S.K. Jain, S.B. Shohet, Calcium potentiates the peroxidation of
erythrocyte membrane lipids, Biochim. Biophys. Acta 642
(1981) 46 54.
D. Thomas, M.R. Hanley, Pharmacological tools for perturbing intracellular calcium storage, Methods Cell Biol. 40
(1994) 65 89.
T. Kuwana, J.J. Smith, M. Muzio, V. Dixit, D.D. Newmeyer,
S. Kornbluth, Apoptosis induction by caspase-8 is amplified
through the mitochondrial release of cytochrome C, J. Biol.
Chem. 26 (1998) 16589 16594.
C.S. Scaffidi, S. Fulda, A. Srinivasan, C. Friesen, F. Li, K.J.
Tomaselli, K.M. Debatin, P.H. Krammer, M.E. Peter, Two
CD95 APOA/Fas signaling pathways, EMBO J. 17 (1998)
1675 1687.
R. Hakem, A. Hakem, G.S. Duncan, J.T. Henderson, M. Woo,
M.S. Soengas, A. Elia, J.L. de la Pompa, D. Kagi, W. Khoo, J.
Potter, R. Yoshida, S.A. Kaufman, S.W. Lowe, J.M. Penninger, T.W. Mak, Differential requirement for caspase 9 in
apoptotic pathways in vivo, Cell 94 (1998) 339 352.
K. Kuida, T.F. Hayder, C.Y. Ktian, Y. Gu, C. Taya, H.
Karasuyama, M.S. Su, P. Rakic, R.A. Flavell, Reduced apoptosis and cytochrome c-mediated caspase activation in mice
lacking caspase 9, Cell 94 (1998) 325 337.
F. Cecconi, G. Alvarez Bolado, B.I. Meyer, K.A. Roth, P.
Gruss, Apaf 1 (CED-4 homolog) regulates programmed cell
death in mammalian development, Cell 94 (1998) 727737.

Вам также может понравиться