Oxidative Stress in the Brain Novel Cellular Targets That Govern 2005

Embed Size (px)

Citation preview

  • 7/31/2019 Oxidative Stress in the Brain Novel Cellular Targets That Govern 2005

    1/40

    Oxidative stress in the brain: Novel cellular targets that governsurvival during neurodegenerative disease

    Zhao Zhong Chong a, Faqi Li a, Kenneth Maiese a,b,c,*

    aDivision of Cellular and Molecular Cerebral Ischemia, Wayne State University School of Medicine, Detroit, MI 48201, USAbDepartment of Neurology and Anatomy & Cell Biology, Center for Molecular Medicine and Genetics, Institute of Environmental Health Sciences,

    Wayne State University School of Medicine, 8C-1 UHC, 4201 St. Antoine, Detroit, MI 48201, USAc

    Center for Molecular Medicine and Genetics, Institute of Environmental Health Sciences,

    Wayne State University School of Medicine, Detroit, MI 48201, USA

    Received 29 July 2004; accepted 16 February 2005

    Abstract

    Despite our present knowledge of some of the cellular pathways that modulate central nervous system injury, complete therapeutic

    prevention or reversal of acute or chronic neuronal injury has not been achieved. The cellular mechanisms that precipitate these diseases are

    more involved than initially believed. As a result, identification of novel therapeutic targets for the treatment of cellular injury would be

    extremely beneficial to reduce or eliminate disability from nervous system disorders. Current studies have begun to focus on pathways of

    oxidative stress that involve a variety of cellular pathways. Here we discuss novel pathways that involve the generation of reactive oxygen

    species and oxidative stress, apoptotic injury that leads to nuclear degradation in both neuronal and vascular populations, and the early loss of

    cellular membrane asymmetry that mitigates inflammation and vascular occlusion. Current work has identified exciting pathways, such as the

    Wnt pathway and the serinethreonine kinase Akt, as central modulators that oversee cellular apoptosis and their downstream substrates that

    include Forkhead transcription factors, glycogen synthase kinase-3b, mitochondrial dysfunction, Bad, and Bcl-xL. Other closely integrated

    pathways control microglial activation, release of inflammatory cytokines, and caspase and calpain activation. New therapeutic avenues that

    are just open to exploration, such as with brain temperature regulation, nicotinamide adenine dinucleotide modulation, metabotropicglutamate system modulation, and erythropoietin targeted expression, may provide both attractive and viable alternatives to treat a variety of

    disorders that include stroke, Alzheimers disease, and traumatic brain injury.

    # 2005 Elsevier Ltd. All rights reserved.

    www.elsevier.com/locate/pneurobioProgress in Neurobiology 75 (2005) 207246

    Abbreviations: Ab, b-amyloid; AD, Alzheimers disease; AIF, apoptosis-inducing factor; ALS, amyotrophic lateral sclerosis; Apaf-1, apoptotic protease-

    activating factor; APC, adenomatous polyposis coli; APP, amyloid precursor protein; BrdU, bromodeoxyuridine; CARD, caspase recruitment domain; CDK,

    cyclin-dependent kinase; CNS, central nervous system; CPCR, G protein-coupled receptor; CREB, cAMP-response element-binding protein; CTMP, carboxy-

    terminal modulator protein; EC, endothelial cell; eIF2B, the translation initiation factor 2B; EPO, erythropoietin; FADD, Fas-associated protein with death

    domain; FLIP, Fas-associated death domain-like interleukins 1b converting enzyme-like inhibitory protein; FRAT1, frequently rearranged in advanced T-cell

    lymphoma type 1; GSK-3b, glycogen synthase kinase-b; HD, Huntingtons disease; 4-HNE, 4-hydroxynonenal; IAP, inhibitor of apoptosis protein; IKK, IkB

    kinase; JNK, c-Jun-amino terminal kinases; Lef, lymphocyte enhancer factor; LPR,lipoprotein related protein; MPP

    +

    , 1-methyl-4-phenylpyridinium; MPTP, 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine; NF-kB, nuclear factor-kB; NO, nitric oxide; 6-OHDA, 6-hydroxydopamine; OHdG, 8-hydroxy-2-deoxyguano-

    sine; OGD, oxygen-glucose deprivation; PARP, poly(ADP-ribose) polymerase; PCD, programmed cell death; PCNA, proliferating cell nuclear antigen; PD,

    Parkinsons disease; PDK1, phosphoinositide-dependent kinase-1; PI 3-K, phosphoinositide 3 kinase; PIP 2, phosphatidylinositol 3,4-bisphosphate; PIP3,

    phosphatidylinositol 3,4,5-trisphosphate; PKB, protein kinase B; PKC, protein kinase C; PP2A, protein phosphatase 2A; PS, phosphatidylserine; PS1,

    presenilin 1; PTEN, the phosphatase and tensin homolog deleted from chromosome 10; ROS, reactive oxygen species; RTK, receptor tyrosine kinase; SN,

    substantia nigra; SOD, superoxide dismutase; Tcf, T cell factor; TCL1, the T cell leukemia/lymphoma 1; TNF, tumor necrosis factor; WISP-1, Wnt-1 induced

    secreted preotein-1

    * Corresponding author at: Department of Neurology, Department of Anatomy & Cell Biology, Center for Molecular Medicine and Genetics, Institute of

    Environmental Health Sciences, Wayne State University School of Medicine, 8C-1 UHC, 4201 St. Antoine, Detroit, MI 48201, USA. Tel.: +1 313 966 0833;

    fax: +1 313 966 0486.

    E-mail address: [email protected] (K. Maiese).

    0301-0082/$ see front matter # 2005 Elsevier Ltd. All rights reserved.

    doi:10.1016/j.pneurobio.2005.02.004

  • 7/31/2019 Oxidative Stress in the Brain Novel Cellular Targets That Govern 2005

    2/40

    Contents

    1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 208

    1.1. The population at risk. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 208

    1.2. Elucidating novel targets within the cell . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 208

    1.3. The biology of oxidative stress . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 209

    2. Oxidative stress and neurodegenerative disease . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 210

    2.1. Acute . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2102.2. Chronic. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 210

    3. Early and late apoptotic programs . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 212

    4. Microglial activation and inflammation. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 213

    5. Attempted cell cycle induction in post-mitotic cells . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 214

    6. Induction of the Wnt pathway. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 215

    7. Akt as an essential regulatory element . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 216

    7.1. Activation and expression of Akt . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 216

    7.2. Akt as a modulator apoptotic injury and inflammation during ROS exposure. . . . . . . . . . . . . . . . . . . . . . . . . . . . . 218

    7.3. Akt can provide the stimulus for altering the course of neurodegenerative disease . . . . . . . . . . . . . . . . . . . . . . . . . 219

    8. Downstream cellular targets . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 220

    8.1. The Forkhead transcription factors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 220

    8.2. GSK-3b . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 222

    8.3. Bad, Bcl-xL, and NF-kB . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2238.4. Mitochondrial dysfunction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 224

    8.5. Caspases . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 227

    8.6. Calpains . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 229

    9. Future directions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 230

    Acknowledgements . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 233

    References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 233

    1. Introduction

    1.1. The population at risk

    At present, over 23 million people in the United Statessuffer from central nervous system (CNS) disorders.

    Globally, this number reaches a level of 368 million people.

    These disorders predominantly consist of neurodegenerative

    diseases that include presenile dementia, Alzheimers

    disease (AD), and Parkinsons disease (PD). Intimately

    linked to the development of CNS degeneration are also a

    variety of injuries associated with traumatic brain injury

    (TBI). For example, both penetrating head injuries and blast

    injuries without direct head trauma have been shown to

    result in subsequent neurotrauma as a result of potential

    elevations in nervous system oxidative stress and free radical

    levels (Cernak et al., 2000). In addition to direct head

    trauma, diffuse neuronal degeneration can ensue as a result

    of an increased load of kinetic energy from the original

    insult (Carey et al., 1984). Furthermore, tangential cranial

    injuries are susceptible to acute ischemic neuronal injury

    with intracerebral hemorrhage (Elron et al., 1998). Finally,

    environmental toxin exposure also may foster oxidative

    neuronal and vascular damage (Miller et al., 2002) (Table 1).

    In the general population, the cost of physician services,

    hospital and nursing home care, and medications continues

    to rise dramatically. In addition, these medical costs for

    neurodegenerative disease parallel a progressive loss of

    economic productivity with rising morbidity and mortality,

    ultimately resulting in an annual deficit to the economy that

    is greater than $ 380 billion. Interestingly, the most

    significant portion of this economic loss is composed of

    only a few neurodegenerative disease entities, such as

    ischemic disease and AD. The annual cost per patient withAD is estimated at $ 174,000 with an annual population

    aggregate cost of $ 100 billion (McCormick et al., 2001;

    Mendiondo et al., 2001).

    1.2. Elucidating novel targets within the cell

    Despite our present knowledge of some of the cellular

    pathways that modulate CNS injury, complete therapeutic

    prevention or reversal of acute or chronic neuronal injury has

    not been achieved. As a result, identification of novel

    therapeutic targets for the treatment of neuronal injury

    would be extremely beneficial to reduce or eliminate

    disability from CNS disorders. Current studies have begun to

    focus on pathways of oxidative stress that involve a variety

    of cellular pathways. Here we describe the unique capacity

    of intrinsic cellular mechanisms that may offer novel therapy

    for a variety of acute and chronic disorders in both neuronal

    and vascular systems. Oxidative stress leads to apoptotic

    injury that involves early loss of cellular membrane

    asymmetry as well as the eventual destruction of genomic

    DNA. These dynamic stages of apoptosis can be associated

    with an ill-fated attempt to enter the cell cycle, particularly

    in post-mitotic neurons. Subsequent cellular pathways can

    originate from the proto-oncogene Wnt and the serine

    Z.Z. Chong et al. / Progress in Neurobiology 75 (2005) 207246208

  • 7/31/2019 Oxidative Stress in the Brain Novel Cellular Targets That Govern 2005

    3/40

    threonine kinase Akt and involve mechanisms linked to

    inflammatory activation of microglia, Forkhead transcrip-

    tion factors, glycogen synthase kinase-3b activation, loss of

    mitochondrial membrane permeability, and the eventual

    induction of caspases and calpains. Understanding these

    processes may ultimately serve to elucidate robust

    therapeutic strategies linked to brain temperature, cellular

    metabolism, genomic DNA repair, metabotropic glutamate

    modulation, and cytokine regulation that allow future

    clinical strategies to mature from bench side prediction

    to daily practice.

    1.3. The biology of oxidative stress

    Oxidative stress occurs when oxygen free radicals are

    generated in excess through the reduction of oxygen.

    Reactive oxygen species (ROS) consist of oxygen free

    radicals and associated entities that include superoxide free

    radicals, hydrogen peroxide, singlet oxygen, nitric oxide

    (NO), and peroxynitrite. Several of these species are

    produced at low levels during normal physiological

    conditions and are scavenged by endogenous antioxidant

    systems that include superoxide dismutase (SOD), glu-

    tathione peroxidase, catalase, and small molecule sub-

    stances such as Vitamins C and E. Superoxide radical is the

    most commonly occurring oxygen free radical that produces

    hydrogen peroxide by dismutation. Hydroxyl radical is the

    most active oxygen free radical and is generated from

    hydrogen peroxide through the HaberWeiss reaction in the

    presence of ferrous iron. Hydroxyl radical alternatively may

    be formed through an interaction between superoxide

    radical and NO (Fubini and Hubbard, 2003). NO interacts

    with superoxide radical to form peroxynitrite that can further

    lead to the generation of peroxynitrous acid. Hydroxyl

    radical is produced from the spontaneous decomposition of

    peroxynitrous acid. NO itself and peroxynitrite are also

    recognized as active oxygen free radicals. In addition to

    directly altering cellular function, NO may work through

    peroxynitrite that is potentially considered a more potent

    radical than NO itself (Pfeiffer et al., 2001).

    Oxidative stress in the brain occurs when the generation

    of ROS overrides the ability of the endogenous antioxidant

    system to remove excess ROS subsequently leading to

    cellular damage. Several cellular features of the brain

    suggest that it is highly sensitive to oxidative stress. For

    example, the brain is known to possess the highest oxygenmetabolic rate of any organ in the body (Maiese, 2002). The

    brain consumes approximately twenty percent of the total

    amount of oxygen in the body. This enhanced metabolic rate

    leads to an increased probability that excessive levels of

    ROS will be produced. In addition, the brain tissues contain

    increased amounts of unsaturated fatty acids that can be

    metabolized by oxygen free radicals. Finally, the brain

    contains high levels of iron which have been associated with

    free radical injury (Herbert et al., 1994). Liposoluble iron

    chelators have been reported to lead to a reduction in ROS

    and protect neurons from permanent focal cerebral ischemia

    (Demougeot et al., 2004). Yet, given the increased risk

    factors for the generation of elevated levels of ROS in the

    brain, it is interesting to note that the brain also may suffer

    from an inadequate defense system against oxidative stress.

    Catalase activity in the brain is significantly below other

    body organs. If one compares the catalase activity of the

    brain to the catalase activity in the liver, the brain has been

    shown to contain only 10% of the catalase activity present in

    the liver (Floyd and Carney, 1992).

    Oxidative stress represents a significant pathway that

    leads to the destruction of both neuronal and vascular cells in

    the CNS. The production of ROS can lead to cell injury

    through cell membrane lipid destruction and cleavage of

    Z.Z. Chong et al. / Progress in Neurobiology 75 (2005) 207246 209

    Table 1

    Oxidative stress in central nervous system disorders

    Diseases Demonstration Selected references

    Acute

    Cerebral ischemia/

    reperfusion

    Superoxide radical and peroxynitrite increased on microvessels;

    impaired mitochondrial function; protection with

    reactive oxygen species reduction

    Bazan et al. (2002); Yamato et al. (2003);

    Gursoy-Ozdemir et al. (2004) and

    Demougeot et al. (2004)

    Traumatic brain injury Reactive oxygen species increased; lipid peroxidation and protein

    oxidation increased; antioxidant reserve decreased

    Awasthi et al. (1997); Tyurin et al. (2000);

    Marklund et al. (2001) and Bayir et al. (2002)

    Chronic

    Alzheimers disease Oxidation of lipids, DNA, and proteins increased; induction of reactive

    oxygen species by amyloid-b; metal ion reduction in senile plaques;

    formation of ion-permeable channels

    Behl et al. (1994); Montine et al. (1999);

    McGrath et al. (2001); Monji et al. (2001)

    and Boland and Campbell (2003)

    Parkinsons disease Oxidation of lipid, DNA, and proteins increased in substantia nigra Alam et al. (1997); Groc et al. (2001);

    Zigmond et al. (2002) and Basso et al. (2004)

    Huntingtons disease Oxidative DNA damage increased in the basal ganglia; reactive

    oxygen species present

    Browne et al. (1997); Bogdanov et al. (2001)

    and Perez-Severiano et al. (2004)

    Amyotrophic lateral sclerosis Reactive oxygen species increased; oxidation of lipids, DNA, and

    proteins increased; mutant in copper zinc superoxide dismutase;

    protection with reactive oxygen species reduction

    Rosen et al. (1993); Liu et al. (1999)

    and Jung et al. (2001)

  • 7/31/2019 Oxidative Stress in the Brain Novel Cellular Targets That Govern 2005

    4/40

    DNA (Vincent and Maiese, 1999b; Wang et al., 2003). ROS

    result in the peroxidation of cellular membrane lipids (Siu

    and To, 2002), peroxidation of docosahexaenoic acid, a

    precursor of neuroprotective docosanoids (Mukherjee et al.,

    2004), the cleavage of DNA during the hydroxylation of

    guanine and methylation of cytosine (Lee et al., 2002), and

    the oxidation of proteins that yield protein carbonylderivatives and nitrotyrosine (Adams et al., 2001). In

    addition to the detrimental effects to cellular integrity, ROS

    can inhibit complex enzymes in the electron transport chain

    of the mitochondria resulting in the blockade of mitochon-

    drial respiration (Yamamoto et al., 2002). In cerebral

    vascular system, the cellular effects of ROS may lead to the

    destruction of endothelial cell (EC) membranes and an

    increase in endothelial cell permeability (Sakamaki, 2004).

    2. Oxidative stress and neurodegenerative disease

    2.1. Acute

    Oxidative brain damage is considered to be a signi ficant

    contributor to ischemic brain injury (Chong et al., 2004b).

    During cerebral ischemia, ROS, such as superoxide radicals,

    are released in significant quantities and have been

    demonstrated at the interface of the cerebrovascular cell

    membrane (Yamato et al., 2003). Sources such as

    cyclooxygenase-2 (COX-2) and impaired mitochondrial

    function can lead to the release of ROS in the brain during

    cerebral ischemia and reperfusion (Bazan et al., 2002).

    Oxygen free radicals subsequently lead to reperfusion-

    induced injury following cerebral ischemia and areassociated with delayed ischemic neuronal damage (Kita-

    gawa et al., 1990). Several mechanisms may account for the

    cellular injury that results during exposure of ROS. Both

    ischemia and the subsequent failure of energy metabolism in

    the brain lead to the calcium-dependent activation of

    phospholipase A2. Phospholipase A2 can then cleave

    membrane phospholipids and release arachidonic acid

    (Mrsic-Pelcic et al., 2002). Superoxide radical is then

    produced with the metabolism of arachidonic acid by

    cyclooxygenase and lipooxygenase that are activated during

    reperfusion. Mitochondrial injury and the electron transport

    impairment also contribute to the production of superoxide

    radicals during focal cerebral ischemia and exacerbate brain

    infarction. ROS can precipitate endoplasmic reticulum

    damage during global brain ischemia that can be attenuated

    by copper zinc SOD overexpression (Hayashi et al., 2003).

    Cerebral ischemia also leads to NO production in the brain

    (Zhu et al., 2002). Superoxide readily reacts with NO

    leading to the formation of peroxynitrite that has been

    considered as a main product of NO contributing to

    reperfusion-induced brain damage following cerebral

    ischemia (Eliasson et al., 1999). Alternatively, NO may

    involve other signal transduction pathways such as protein

    kinase A and protein kinas C (Maiese and Boccone, 1995;

    Maiese et al., 1993). Following cerebral ischemia, reperfu-

    sion leads to the significant formation of superoxide, NO,

    and peroxynitrite on microvessels and surrounding end-feet.

    These ROS are believed to disrupt microvascular integrity

    resulting in cerebral hemorrhage and edema (Gursoy-

    Ozdemir et al., 2004).

    In addition to the evidence for the production of ROSduring the acute onset of cerebral ischemia and subsequent

    reperfusion injury, the ability to protect both neuronal and

    vascular tissue during cerebral ischemia with antioxidants or

    scavengers of ROS offers further support for the involve-

    ment of ROS during acute cerebral ischemia. For example,

    biologically active SOD fusion proteins can prevent

    hippocampal neuronal injury during transient forebrain

    ischemia (Sik Eum et al., 2004). Furthermore, novel free

    radical scavengers, such as 8-(4-fluorophenyl)-2-((2E)-3-

    phenyl-2-propenoyl)-1,2,3,4-tetra-hydropyrazolo[5,1-

    c][1,2,4]triazine (FR210575), can significantly reduce

    cortical damage by almost 40% in a transient model of

    cerebral ischemia and protect against apoptotic injury during

    permanent cerebral injury (Iwashita et al., 2003).

    Oxidative stress also has been suggested to play a crucial

    role in the pathology of TBI. Following TBI, increased

    ascorbyl free radical signals and reduced ascorbic acid has

    been demonstrated in rats (Awasthi et al., 1997). Additional

    investigations have shown an increase in lipid peroxidation,

    production of peroxynitrite, and impairment of the

    endogenous antioxidant system following TBI (Hall et al.,

    2004; Tyurin et al., 2000). Similarly, a sustained decrease in

    the total antioxidant reserve including ascorbate and

    glutathione has been observed in the cerebrospinal fluid

    in infants and children after severe TBI (Bayir et al., 2002).The level of free radical induced products of lipid

    peroxidation and protein oxidation in the cerebrospinal

    fluid also were increased following TBI (Bayir et al., 2002).

    In contrast, scavenging of ROS after TBI can improve

    neurological function and reduce cerebral injury (Marklund

    et al., 2001).

    2.2. Chronic

    Oxidative stress is considered to play a significant role in

    the onset and progression of AD (Maiese and Chong, 2004;

    Mattson, 2004). AD leads to a progressive deterioration of

    cognitive function with memory loss and is characterized by

    two pathologic hallmarks that consist of extracellular

    plaques of amyloid-b peptide aggregates and intracellular

    neurofibrillary tangles composed of hyperphosphorylated

    microtubular protein tau. The b-amyloid deposition that

    constitutes the plaques is composed of a 3942 amino acid

    peptide (Ab), which is the proteolytic product of the

    amyloid precursor protein (APP) (Maiese and Chong, 2004).

    The association of oxidative stress with AD is dependent

    on several lines of evidence. The oxidative products of

    lipids, protein, and DNA have been reported in patients with

    AD. In the neocortex of the brain of individuals with AD, the

    Z.Z. Chong et al. / Progress in Neurobiology 75 (2005) 207246210

  • 7/31/2019 Oxidative Stress in the Brain Novel Cellular Targets That Govern 2005

    5/40

    end product of lipid peroxidation, malondialdehyde (MDA),

    has been observed to be in significantly higher quantities

    than in aged matched controls (Palmer and Burns, 1994).

    Elevated levels of another product of lipid peroxidation,

    4-hydroxynonenal (4-HNE), also has been shown to be

    increased in the plasma of patients with AD (McGrath et al.,

    2001). 4-HNE is an aldehyde product of lipid peroxidationthat can lead to caspase activation and apoptosis (Liu et al.,

    2000). In addition, HNE can become conjugated to the

    neuronal glucose transporters (Mark et al., 1997) and as a

    result has been suggested to be linked to impaired cellular

    glucose transport activity in AD (Masliah et al., 1996). Loss

    of specific plasma proteins, such as apolipoprotein E (apoE),

    also may play a pivotal role during oxidative stress induced

    injury during AD. In studies that examined cortical

    synaptosomes or neurons from transgenic mice lacking

    apoE, samples from apoE knockout mice possessed

    increased levels of oxidative stress and caspase activity

    during Ab exposure (Keller et al., 2000) as well as enhanced

    NO synthase activity (Law et al., 2003), suggesting a

    protective role for apoE. Although some investigators argue

    that observed lipid peroxidation in the brain of AD patients

    does not appear to correlate with the extent of neuritic

    plaques, neurofibrillary tangles, or apoE genotype, lipid

    peroxidation does appear to directly coincide with pro-

    gressive neuronal degeneration in AD patients (Montine

    et al., 1999). Other observations further support a role for

    ROS during AD. Selective oxidative modification of

    intracellular proteins, such as increased protein carbonyl

    levels in creatine kinase BB and b-actin, can be seen in AD

    (Chong et al., 2005b).

    Other evidence exists that suggests cellular injury duringAD may result from both ROS as well as from impaired

    cellular repair mechanisms following oxidative injury. In

    one study, 8-hydroxy-20-deoxyguanosine (8-OHdG), a

    marker of oxidative damage in intact DNA and as a free

    repair product during DNA repair mechanisms, was

    examined in the cerebrospinal fluid of AD patients.

    Significant elevations of 8-OHdG linked to intact DNA

    were observed in the cerebrospinal fluid of AD patients,

    suggesting that these patients suffer from impaired DNA

    mechanisms. Yet, levels of free 8-OHdG, which are

    generated during normal cellular repair mechanisms, were

    found to be significantly depleted in the cerebrospinal fluid

    of AD patients, further supporting the premise of deficient

    DNA repair mechanisms in these patients (Lovell et al.,

    1999).

    The neurotoxicity of Ab, a major component of AD

    pathogenesis, also is associated with cellular injury

    following ROS exposure. In mice overexpressing APP,

    the Ab deposits that are characteristically found in AD co-

    localize with several oxidative stress markers (Smith et al.,

    1998), suggesting that there exists a close correlation

    between oxidative stress and Ab deposition. In addition,

    agents that modulate ROS have been shown to reduce

    cellular injury during Ab exposure. Application of the free

    radical antioxidant Vitamin E has been demonstrated to

    prevent neurotoxicity from Ab (Subramaniam et al., 1998).

    Over the last decade, a body of work has been generated

    to support the premise that Ab can directly lead to the

    generation of ROS. Early studies have demonstrated that Ab

    can lead to the generation of hydrogen peroxide and cell

    death in primary neuronal cultures (Behl et al., 1994). Theability of Ab to generate ROS may be a result of its

    methionine composition, since the substitution of methio-

    nine by valine, or the removal of the methionine in Ab,

    blocks ROS production, protein oxidation, and toxicity to

    primary hippocampal neurons (Varadarajan et al., 1999).

    Furthermore, free radical generation by Ab appears to be

    strongly influenced by the aggregational state of the

    peptides, such that inhibition of Ab aggregation can reduce

    neuronal toxicity and free radical generation (Monji et al.,

    2001; Tomiyama et al., 1996). The generation of hydrogen

    peroxide by Ab may be mediated through mechanisms that

    are related to metal ion reduction (Huang et al., 1999) and

    the cellular ions of copper, zinc, and iron (Liu et al., 1999b)

    that are significantly elevated in the senile plaques of

    patients with AD and can accelerate aggregation of Ab

    (Deibel et al., 1996).

    Channel formation during oxidative stress also may be a

    significant factor in the pathogenesis of AD and Ab toxicity.

    Ab is able to spontaneously insert into planar lipid

    membranes to form selective, voltage-dependent, ion-

    permeable channels (Arispe et al., 1993; Mirzabekov

    et al., 1994). The subsequent channels formed may be

    calcium-permeable and lead to cellular toxicity through

    impaired calcium homeostasis (Lin et al., 1999; Sanderson

    et al., 1997) as well as through calpain activation (Bolandand Campbell, 2003). Aggregates of Ab can further interact

    with the lipid bilayer and reduce membrane fluidity to

    potentially impair cell function and promote cell injury

    (Kremer et al., 2001). Association with membrane

    phospholipids by Ab can be extensive in nature to disrupt

    both endosomal and plasma membranes through a pH

    dependent mechanism (McLaurin and Chakrabartty, 1996).

    Cellular injury as a result of ROS appears to proceed

    through apoptotic or programmed cell death (PCD)

    mechanisms. Accumulating evidence has been obtained

    from human and in vitro models of AD suggesting that

    apoptosis contributes to the neuronal loss during the disease.

    Data from in situ TUNEL (terminal deoxynucleotidyl

    transferase nick-end labeling) assays of brain tissues from

    individuals with AD demonstrate neuronal demise consis-

    tent with PCD. A correlation between the incidence of

    TUNEL-positive cells and plaque density also exists

    (Colurso et al., 2003). Levels of the apoptotic marker

    prostate apoptosis response-4 (Par-4) has been shown to be

    significantly increased in the brains of patients with AD

    (Guo et al., 1998). Other lines of evidence link apoptotic

    cellular injury with APP and its proteolytic product Ab. I n i n

    vitro studies, expression of familial AD mutants of APP

    results in apoptotic neuronal injury (McPhie et al., 2003). It

    Z.Z. Chong et al. / Progress in Neurobiology 75 (2005) 207246 211

  • 7/31/2019 Oxidative Stress in the Brain Novel Cellular Targets That Govern 2005

    6/40

    is the cytoplasmic domain of APP that can lead to sustained

    apoptosis through c-Jun N-terminal kinase pathways

    (Hashimoto et al., 2003b). Additional studies have

    illustrated that direct application of Ab to neuronal cells

    can lead to chromatin condensation characteristic of

    apoptosis in cultured neurons.

    PD is a movement disorder characterized by restingtremor, rigidity, and bradykinesia. The pathophysiological

    basis of the symptoms rests upon the degeneration of

    dopaminergic neurons in the substantia nigra (SN). In some

    scenarios, it has been hypothesized that dopamine may even

    be a culprit in precipitating disease progression (Maiese

    et al., 2003). Dopamine may increase the rate of the

    generation of ROS species and subsequent oxidative

    products as well as decrease the reserve capacity of the

    brain to inactivate ROS (Zigmond et al., 2002). Other

    observations also support the premise that PD is a result of

    ROS generation. Cerebral iron, which can be a catalyst for

    the formation of hydroxyl radicals has been demonstrated to

    be increased in the basal ganglia of individuals with PD

    (Griffiths et al.,1999). Elevations in oxidative products, such

    as lipid peroxides (Groc et al., 2001), protein carbonyls

    (Alam et al., 1997), and products of nucleic acid 8-

    hydroxyguanosine (Zhang et al., 1999), have been observed

    in the SN of PD patients. Furthermore, when protein

    expression was compared in the SN from patients with PD

    and from controls, a total of 44 proteins expressed in the SN

    were identified by peptide mass fingerprinting with several

    representing mitochondrial and ROS scavenging proteins

    supporting oxidative stress involvement (Basso et al., 2004).

    As a correlation to the increased levels of these products in

    the brain, a systemic increase of the oxidized products ofDNA, RNA, 8-hydroxyguanosine, and 8-hydroxy-20-deox-

    yguanosine has been found in the serum and cerebrospinal

    fluid of individuals with PD (Kikuchi et al., 2002). Given

    these studies, new approaches to treat patients with PD

    advocate the use of neuroprotective monoamine oxidase

    inhibitors combined with iron chelation therapy (Youdim

    et al., 2004).

    Huntingtons disease (HD) is an autosomal dominant

    neurodegenerative disease characterized by impairment of

    involuntary movement and cognitive impairment. Selective

    loss of neurons in the basal ganglia and cerebral cortex is one of

    the anatomical hallmarks of this disease. In patients with HD,

    the basal ganglia has increased levels of OHdG, suggestive of

    oxidative DNA damage (Browne et al., 1997). Furthermore,

    transgenic models of HD with R6/2 mice reveal increased

    OHdG in urine, plasma, and striatal microdialysates (Bogda-

    nov et al., 2001). In other studies with transgenic R6/1 mice,

    dichlorofluorescein (DCF), an index of ROS formation, was

    significantly increased in R6/1 mice at 11, 19, and 35 weeks of

    age while the antioxidant catalase enzyme was significantly

    depressed, suggesting an active role for ROS during the onset

    and progression of HD (Perez-Severiano et al., 2004).

    Amyotrophic lateral sclerosis (ALS), a disabling and

    fatal neurodegenerative disease, is characterized by the

    progressive loss of muscle power as a result of the selective

    loss of motor neurons in the motor cortex, brainstem, and

    spinal cord. Although approximately 10% of the reported

    cases are associated with inheritance, approximately 23%

    of observed ALS cases can be related to a mutation in the

    antioxidant enzyme copper zinc SOD (Rosen et al., 1993).

    ROS has been found to be increased in ALS in mice(Bogdanov et al., 1998). Additionally, the presence of

    oxidative products of protein, DNA, and lipid in the brains of

    ALS patients supports an involvement of ROS in the

    pathology of ALS (Liu et al., 1999a). Reduction in ROS may

    offer hope in providing some form of therapy for ALS. For

    example, mice expressing human mutant SOD1 G93A with

    EUK-8 and EUK-134, two synthetic SOD/catalase

    mimetics, have been shown to reduce oxidative stress and

    potentially prolong survival in animal models of ALS (Jung

    et al., 2001).

    3. Early and late apoptotic programs

    Apoptosis, or PCD, is considered to be important for

    tissue re-modeling during development. Yet, this active

    process is recognized as a central pathway that can lead to a

    cells demise in a variety of tissues and has recently been

    identified in organisms as diverse as plants (Hatsugai et al.,

    2004). PCD consists of two independent processes that

    involve membrane phosphatidylserine (PS) exposure and

    DNA fragmentation (Maiese et al., 2004). Apoptotic injury

    is believed to contribute significantly to a variety of disease

    states that especially involve the nervous system such as

    ischemic stroke, AD, PD, and spinal cord injury (Chong andMaiese, 2004; Li et al., 2004b). Outside of the nervous

    system, such as during cardiovascular injury, PCD also may

    be a significant precipitant of cell death. Ischemic-

    reperfusion injury can lead to apoptosis in cardiomyocytes

    (Cai et al., 2003).

    As an early event in the dynamics of cellular apoptosis,

    the biological role of membrane PS externalization can vary

    in different cell populations. In some cell systems, PS may

    be required for embryogenesis (Bose et al., 2004). Yet, in

    mature tissues, membrane PS externalization can become a

    signal for the phagocytosis of cells (Hong et al., 2004).In the

    nervous system, cells expressing externalized PS may be

    removed by microglia (Chong et al., 2003c; Li et al., 2004b).

    An additional role for membrane PS externalization in the

    vascular cell system is the activation of coagulation

    cascades. The externalization of membrane PS residues in

    ECs can promote the formation of a procoagulant surface

    (Chong et al., 2004a).

    In contrast to the early externalization of membrane PS

    residues, the cleavage of genomic DNA into fragments is

    considered to be a delayed event that occurs late during

    apoptosis (Dombroski et al., 2000; Jessel et al., 2002; Kang

    et al., 2003b; Maiese and Vincent, 2000). Several enzymes

    responsible for DNA degradation have been differentiated

    Z.Z. Chong et al. / Progress in Neurobiology 75 (2005) 207246212

  • 7/31/2019 Oxidative Stress in the Brain Novel Cellular Targets That Govern 2005

    7/40

    based on their ionic sensitivities to zinc (Torriglia et al.,

    1997) and magnesium (Sun and Cohen, 1994). Calcium, a

    critical independent component that can determine cell

    survival (Weber, 2004), also may determine endonuclease

    activity through calcium/magnesium-dependent endonu-

    cleases such as DNase I (Madaio et al., 1996). Other

    enzymes that may disassemble DNA include the acidic,cation independent endonuclease (DNase II) (Torriglia et al.,

    1995), cyclophilins (Montague et al., 1997), and the 97 kDa

    magnesium-dependent endonuclease (Pandey et al., 1997).

    In the nervous system, three separate endonuclease activities

    are present that include a constitutive acidic cation-

    independent endonuclease, a constitutive calcium/magne-

    sium-dependent endonuclease, and an inducible magne-

    sium-dependent endonuclease (Vincent and Maiese, 1999b).

    The physiologic characteristics of the magnesium-depen-

    dent endonuclease, such as a pH range of 7.48.0, a

    dependence on magnesium, and a molecular weight of 95

    108 kDa, are consistent with a recently described consti-

    tutive 97 kDa endonuclease in non-neuronal tissues.

    Exposure to ROS can precipitate apoptosis in neurons and

    ECs through multiple cellular pathways. Oxidative stress,

    such as NO or hydrogen peroxide, results in nuclei

    condensation and DNA fragmentation (Chong et al.,

    2003b; Goldshmit et al., 2001; Pugazhenthi et al., 2003;

    Vincent and Maiese, 1999b). In neurons, NO exposure

    produces apoptotic death in hippocampal and dopaminergic

    neurons (Chong et al., 2003a; Sharma and Ebadi, 2003;

    Vincent and Maiese, 1999a; Witting et al., 2000). Injury

    during NO exposure also can become synergistic with

    hydrogen peroxide to render neurons more sensitive to

    oxidative injury (de la Monte et al., 2003; Wang et al., 2003).Hydrogen peroxide also results in neuronal injury through

    impaired mitochondrial function and increased levels of pro-

    apoptotic gene products, such as CD95/Fas (de la Monte

    et al., 2000; Pugazhenthi et al., 2003; Vaudry et al., 2002).

    Externalization of membrane PS residues also occurs in

    neurons during anoxia (Chong et al., 2002b), NO exposure

    (Chong et al., 2003f), or during the administration of agents

    that induce the production of ROS, such as 6-hydroxydo-

    pamine (Salinas et al., 2003).

    4. Microglial activation and inflammation

    Modulation of extrinsic cell homeostasis through micro-

    glial activation is as vital to cellular survival as the

    maintenance of cellular DNA integrity. Microglia are

    monocyte-derived immunocompetent cells that enter the

    CNS during embryonic development and function similar to

    peripheral macrophages for the phagocytic removal of

    apoptotic cells. Some studies identify the generation of

    annexin I and membrane PS exposure that appears to be

    necessary to connect an apoptotic cell with a phagocyte

    (Arur et al., 2003). Secreted factors by either apoptotic or

    phagocytic cells, such as milk fat globule-EGF-factor 8

    (Hanayama et al., 2004), fractalkine (Hatori et al., 2002),

    and lipid lysophosphatidylcholine (Lauber et al., 2003), also

    have been shown to assist with the phagocytic removal of

    injured cells. Yet, the translocation of membrane PS residues

    from the inner cellular membrane to the outer surface

    appears to be essential for the removal of apoptotic cells

    (Fadok et al., 2001; Kang et al., 2003b; Maiese and Vincent,2000). The phospholipids of the plasma membrane are

    normally in an asymmetric pattern with the outer leaflet of

    the plasma membrane consisting primarily of choline-

    containing lipids, such as phosphatidylcholine and sphin-

    gomyelin, and the inner leaflets consisting of aminopho-

    spholipids that include phosphatidylethanolamine and PS.

    The loss of membrane phospholipid asymmetry leads to the

    externalization of membrane PS residues and serves to

    identify cells for phagocytosis (Chong et al., 2003d;

    Hoffmann et al., 2001; Kang et al., 2003b; Maiese and

    Chong, 2003).

    Expression of the phosphatidylserine receptor (PSR) on

    microglia also functions with cellular membrane PS

    externalization to activate microglia. Cells, such as neurons

    or ECs, exposed to ROS can lead to the induction of both

    microglial activation and microglial PSR expression.

    Treatment with an anti-PSR neutralizing antibody in

    microglia prevents this microglial activation (Chong

    et al., 2003b; Kang et al., 2003a) and application of PS

    directly results in microglial activation that can be blocked

    by a PSR neutralizing antibody (Chong et al., 2003b; Kang

    et al., 2003b), suggesting that both PS exposure in target

    cells and PSR expression in microglia are necessary for

    microglial recognition of apoptotic cells in the nervous

    system. Recognition of cellular membrane PS by the PS-specific receptors on microglia may require cofactors, such

    as Gas6 (Nakano et al., 1997) or other agents, such as

    integrin and lectin (Witting et al., 2000).

    Although microglia may assist with the removal of

    injured cells and cellular debri, these cellular scavengers of

    the brain may sometimes aggravate tissue inflammation.

    Studies with microglia stimulated by phorbol myristate

    acetate have demonstrated the release of superoxide

    radicals. Application of scavenger agents for ROS, such

    as SOD or deferoxamine mesylate, in the presence of

    activated microglia can prevent cellular injury. These studies

    suggest that oxidative stress generated by microglia can be

    responsible for cellular injury (Tanaka et al., 1994).

    Microglia may lead to cellular damage in disease entities,

    not only through the generation of ROS products

    (Sankarapandi et al., 1998) but also through the production

    of cytokines and the demise of neighboring neurons and ECs

    (Benzing et al., 1999; Mehlhorn et al., 2000). In HD and

    ALS, significant microglial activation has been reported in

    regions of the nervous system that are specific for these

    disease entities (Obal et al., 2001; Singhrao et al., 1999).

    During ischemic injury to cells, activation of microglia can

    parallel the induction of cellular apoptosis and correlate well

    with the severity of the ischemic insult (Chong et al., 2004a;

    Z.Z. Chong et al. / Progress in Neurobiology 75 (2005) 207246 213

  • 7/31/2019 Oxidative Stress in the Brain Novel Cellular Targets That Govern 2005

    8/40

    Kang et al., 2003b). Microglia promote the production of

    pro-inflammatory cytokines such as tumor necrosis factor-a

    (TNF-a) and interleukin-1b, free radicals such as NO and

    superoxide (Sankarapandi et al., 1998), and fatty acid

    metabolites such as eicosanoids that can precipitate cell

    death (Liu and Hong, 2003). TNF-a production by microglia

    may be linked to neurodegeneration by increasing thesensitivity of neurons to free radical exposure (Combs et al.,

    2001).

    In several neurodegenerative diseases, microglial activa-

    tion has been identified through glial cultures in autopsy

    specimens (Lue et al., 1996). For example, expression of

    markers that are indicative of microglial activation was

    found to be significantly increased in patients with AD

    (Rogers and Lue, 2001). Application of a position emission

    tomography marker [11C](R)-PK11195 for microglial

    activation in patients with mild and early AD also has

    demonstrated microglial activation in regions of the

    entorhinal, parietal, and cingulate cortex, suggesting that

    microglial activation is an early event in the pathogenesis of

    the disease (Cagnin et al., 2001).

    One of the major pathogens of AD, namely Ab, has been

    shown to lead to inflammatory cell injury through a variety

    of routes. Ab can not only precipitate a significant

    inflammatory response with microglial activation and the

    secretion of TNF-a (Bornemann et al., 2001), but also Ab

    can elicit the neuronal expression of inducible nitric oxide

    synthase, peroxynitrite production, and neuronal apoptosis

    during an acute inflammatory response (Combs et al., 2001).

    Microglial cells also co-localize with the perivascular

    deposits of Ab and microglial activation correlates with the

    development of amyloid plaques (Sheng et al., 1997).Ultrastructural three-dimensional reconstruction of human

    amyloid plaques in different stages of development

    illustrates that the number of microglia parallels a

    progressive increase in fibrillar deposition and the size of

    fibrillar plaque (Wegiel et al., 2000). The generation of ROS

    by microglia during events such as Ab deposition suggests

    that microglia may play an important role during the

    development of neurodegenerative diseases.

    5. Attempted cell cycle induction in post-mitotic cells

    Theattempted reentrance into thecell cyclein post-mitotic

    neurons can trigger apoptosis (Becker and Bonni, 2004). In

    the CNS, post-mitotic neurons are incapable of differentia-

    tion, but they continue to possess the ability to enter into the

    cell cycle. During a cellular insult, deregulation of cell cycle

    proteins, such as cyclin, cyclin-dependent kinase (CDK), and

    the retinoblastoma protein, can ensue (Padmanabhan et al.,

    1999).The deficiency of several essential components for the

    complete execution of the cell cycle in post-mitotic neurons is

    believed to be deleterious to neurons. Several studies have

    provided direct evidence that cell cycle induction in post-

    mitotic neurons can activate cellular mechanisms that lead to

    neuronal apoptosis (El-Khodor et al., 2003; Ino and Chiba,

    2001; Konishiand Bonni,2003; Lin et al., 2001; Rideout et al.,

    2003; Tetsu and McCormick, 1999). Investigations that

    examine ROS as a stimulus for cell cycle induction reveal that

    distinct components of apoptotic injury, membrane PS

    exposure and genomic DNA fragmentation, occur in concert

    with early and late phases of cell cycle induction (Lin et al.,2001).

    Oxidative injury associated with ROS may lead to

    attempted cell cycle induction in neurons. The induction of

    oxidative stress in sympathetic neurons by either dopamine,

    which produces free radicals during its metabolism, or by

    hydrogen peroxide leads to the increased expression of cell

    cycle related genes that include cyclin B and CDK5 prior to

    the induction of neuronal apoptosis (Shirvan et al., 1998).

    Furthermore, antioxidants that include N-acetyl-L-cysteine

    (LNAC) and N-acetyl-D-cysteine (DNAC) can prevent DNA

    fragmentation during trophic factor deprivation through

    mechanisms that may involve the inhibition of cell cycle

    progression in neuronal cell lines (Ferrari et al., 1995).

    Acute injury paradigms have suggested a potential role for

    ill-fated cell cycle induction in neurons. Cell cycle proteins

    (cyclin A, cyclin D, CDK2, CDK4) have been co-localized

    with apoptotic cells following middle cerebral artery

    occlusion (Li et al., 1997b). Although evidence for cell cycle

    induction during cerebral ischemia may be partially

    associated with neurogenesis (Taguchi et al., 2004), ischemic

    insults also can lead to aberrant cell cycle induction that

    may have ramifications for both acute and long-term

    consequences on cellular function (Wen et al., 2004).

    Other neurodegenerative diseases, such as AD, also

    appear to rely upon attempted cell cycle induction, at least inpart, to yield subsequent neuronal cell loss (Arendt et al.,

    2000; Busser et al., 1998; Maiese, 2001; Raina et al., 2000 ).

    In clinical specimens from AD patients, the cell cycle

    regulators P16 and CDK4 have increased expression in

    regions such as the hippocampus (McShea et al., 1997). In

    addition, expression of other components of the cell cycle,

    such as cyclin D, CDK4, proliferating cell nuclear antigen

    (PCNA), and cyclin B1 have been shown to be present in

    patients with AD in regions that include the hippocampus,

    subiculum, locus coeruleus, and dorsal raphe nuclei. A close

    association appears to exist between injured cells and cell

    cycle protein expression, since staining for cell cycle

    proteins have been shown to be absent in brain regions

    without neuronal injury of AD patients and in age-matched

    brains (Busser et al., 1998). Increased accumulation of cell

    cycle kinases, such as CDK5, also has been found in neurons

    that are developing neurofibrillary tangles (Pei et al., 1998).

    Interestingly, in patients with mild cognitive impairment,

    many of which can progress to develop AD (Bennett et al.,

    2002), cell cycle proteins, such as cyclin D, cyclin B, and

    PCNA, are significantly increased in the hippocampus and

    basal nucleus (Yang et al., 2003).

    Experimental models of AD have provided further

    evidence that cell cycle induction in post-mitotic neurons

    Z.Z. Chong et al. / Progress in Neurobiology 75 (2005) 207246214

  • 7/31/2019 Oxidative Stress in the Brain Novel Cellular Targets That Govern 2005

    9/40

    can activate cellular mechanisms that lead to neuronal

    apoptosis. For example, application of Ab (140), Ab (1

    42), and its active fragment Ab (2535) in neurons can result

    in the induction of cyclin D1, cyclins E and A, and the

    phosphorylation of the retinoblastoma protein. The activa-

    tion of the upstream cyclin-dependent kinases (CDK)4/5/6

    appears to be required for the induction of apoptosis inneurons by Ab, since inhibition of CDKs can prevent Abinduced neuronal apoptosis (Alvarez et al., 2001; Giovanni

    et al., 1999).

    Cell cycle proteins can contribute to neurofibrillary

    tangle development. Expression of familial AD mutants of

    the APP in primary neurons can precipitate apoptotic injury

    through cell cycle induction and p21 mediated pathways

    (McPhie et al., 2003). CDK5 also has been identified as a

    critical regulator of the tau protein which leads to

    neurofibrillary tangles. CDK5 can phosphorylate tau

    directly (Flaherty et al., 2000). Furthermore, phosphoryla-

    tion of tau by Ab can be blocked by treatment with antisense

    against p35, a protein that is cleaved to the truncated form

    p25 which can activate CDK5. This work provides evidence

    that Ab requires both the cleavage of p35 and the activation

    of CDK5 to lead to tau phosphorylation (Town et al., 2002).

    Correlative work has shown that p25 also accumulates in

    neurons of patients with AD (Patrick et al., 1999). In

    addition, overexpression of the p25/Cdk5 complex in

    cultured primary neurons leads to cytoskeletal disruption,

    the hyperphosphorylation of tau, and apoptosis (Patrick

    et al., 1999), suggesting that induction of cell cycle proteins

    can be a significant precipitant for neuronal degeneration.

    6. Induction of the Wnt pathway

    Wnt proteins, named after the Drosophilia protein

    wingless and the mouse protein Int-1, represent a

    large family of secreted cysteine-rich glycosylated proteins.

    This novel family of proteins are intimately involved in

    cellular signaling pathways that play a role in a variety of

    processes that involve embryonic cell patterning, prolifera-

    tion, differentiation, orientation, adhesion, survival, and

    apoptosis (Chong and Maiese, 2004; Nelson and Nusse,

    2004; Patapoutian and Reichardt, 2000).

    Nineteen of the 24 Wnt genes that express Wnt proteins

    have been identified in the human. In addition, greater than

    80 target genes of Wnt signaling pathways have been

    demonstrated in human, mouse, Drosophilia, Xenopus, and

    Zebrafish. This representation encompasses several cellular

    populations, such as neurons, cardiomyocytes, endothelial

    cells, cancer cells, and pre-adipocytes (Nusse, 1999). Wnt

    binds to Frizzled transmembrane receptors on the cell

    surface to activate downstream signaling events (Fig. 1).

    These involve at least two intracellular signaling pathways

    that are considered of particular importance. One pathway

    controls target gene transcription through b-catenin,

    generally referred to as the canonical pathway that involves

    Wnt1, Wnt3a, and Wnt8 and functions through b-catenin-

    dependent pathways. Another pathway pertains to intracel-

    lular calcium (Ca2+) release which is termed the non-

    canonical or Wnt/Ca2+ pathway consisting primarily of

    Wnt4, Wnt5a, and Wnt11 that functions through non-b-

    catenin-dependent pathways, such as the planar cell polarity

    (PCP) pathway and the Wnt-Ca2+-dependent pathways

    (Kuhl et al., 2000; Nusse, 1999; Patapoutian and Reichardt,

    2000).

    As one of the best characterized members of the Wnt

    family, Wnt1 was first identified as a proto-oncogene in

    mammary carcinomas, but recently has been illustrated to

    play a critical role in neuronal development (Tang et al.,

    2002). Wnt functions by binding to the transmembrane

    receptor Frizzled and the co-receptor lipoprotein related

    proteins 5 and 6 (LRP-5/6) (Wehrli et al., 2000) followed by

    recruitment of disheveled, the cytoplasmic bridging mole-

    cule, to inhibit glycogen synthase kinase (GSK-3b) (Ikeda

    et al., 1998; Papkoff and Aikawa, 1998). The inhibition of

    GSK-3b prevents phosphorylation of b-catenin and its

    Z.Z. Chong et al. / Progress in Neurobiology 75 (2005) 207246 215

    Fig. 1. Modulation of apoptotic injury by Wnt and Akt pathways. The Wnt

    canonical signaling pathway is initiated by activation of its transmembrane

    receptor Frizzled (Friz) and the co-receptor lipoprotein related proteins 5

    and 6 (LRP-5/6), resulting in the recruitment and activation of disheveled

    which inhibits glycogen synthase kinase (GSK)-3b. When active, GSK-3bfunctions with adenomatous polyposis coli (APC) and the phosphorylation

    of Axin to result in b-catenin phosphorylation and its subsequent degrada-

    tion. In contrast, free b-catenin translocates to the nucleus and activates

    lymphocyte enhancer factor (Lef) and T cell factor (Tcf) to stimulate Wnt-

    responsive genes. The serinethreonine kinase Akt functions as a down-

    stream target of phosphoinositide 3 kinase (PI 3-K). PI 3-K phosphorylates

    glycerophospholipid phosphatidylinositol 4,5-bisphosphate, yielding phos-

    phatidylinositol 3,4-bisphosphate (PIP2) and phosphatidylinositol 3,4,5-

    trisphosphate (PIP3). As a cytosolic protein, Akt translocates to the cell

    membrane following its binding to PIP2 and PIP3 and becomes activated

    through phosphorylation by phosphoinositide-dependent kinase 1 (PDK1).

    Wnt also may activate Akt through the Wnt-1 induced secreted protein

    (WISP-1). Akt targets GSK-3b through phosphorylation, resulting in the

    inactivation of GSK-3b and blocking the degradation ofb-catenin. Further-

    more, phosphorylation of the translation initiation factor 2B (eIF2B) isprevented to prevent the release of cytochrome c (Cyto c). In addition, Akt

    inactivates FOXO3a and Bad to inhibit induction of Bim and restore Bcl-xLfunction.

  • 7/31/2019 Oxidative Stress in the Brain Novel Cellular Targets That Govern 2005

    10/40

    degradation. The free b-catenin translocates to the nucleus

    where it activates lymphocyte enhancer factor (Lef) and T

    cell factor (Tcf) (Ishitani et al., 2003) leading to stimulation

    of Wnt-response genes (Fig. 1).

    In some cell systems, Wnt1 signaling has been associated

    with the control of apoptosis. Wnt-1 prevents apoptosis

    through b-catenin/Tcf transcription mediated pathways(Chen et al., 2001; Rhee et al., 2002). Overexpression of

    exogenous Wnt1 results in the protection of cells against

    c-myc induced apoptosis through induction of b-catenin,

    cyclooxygenase-2, and Wnt1 induced secreted protein

    (WISP-1) (You et al., 2002). Wnt1 signaling also can

    inhibit apoptosis through prevention of cytochrome c release

    from mitochondria and the subsequent inhibition of caspase

    9 activation (Chen et al., 2001). The adenomatous polyposis

    coli (APC) gene, a member of the Wnt pathway, appears to

    represent another mechanism that regulates PCD. The APC

    gene functions to cleave b-catenin leading to the down-

    regulation of transactivation of Tcf/Lef (Tetsu and McCor-

    mick, 1999). Without Tcf/Lef activity, APC is then

    permitted to increase the activities of caspase 3, caspase

    7, and caspase 9 and lead to the cleavage of poly(ADP-

    ribose) polymerase (PARP) to enhance the vulnerability of

    cells to apoptosis (Chen et al., 2003).

    In the nervous system, the non-canonical Wnt pathway

    has been shown to be expressed in the hippocampus of mice

    and can increase dendritic branching in cultured neurons

    (Rosso et al., 2005). Wnt signaling through Wnt1 also is able

    to guide early neural crest stem cells to develop into sensory

    neural cells rather than maturing into other potential neural

    crest cell derivatives (Lee et al., 2004). Yet, in regards to

    cytoprotection in the brain that involves the Wnt pathway,limited studies are available. The work that is presently

    available suggests that enhanced Wnt activity may function

    through several cellular pathways to prevent apoptosis

    during neuronal or vascular injury. Conditioned media with

    Wnt3a activity or the application of a GSK-3b inhibitor can

    block hydrogen peroxide induced mitochondrial dysfunc-

    tion and apoptotic DNA fragmentation (Shin et al., 2004).

    Other work illustrates that Wnt signaling may foster specific

    protection against cellular destruction and inflammatory

    injury by maintaining genomic DNA integrity and cellular

    membrane PS asymmetry (Chong et al., 2004b; Maiese and

    Vincent, 2000). Wnt1 overexpression in primary hippo-

    campal neurons protects cells against oxidative stress or Ab

    toxicity that increases cell survival and prevents PS exposure

    and DNA degradation (Chong et al., 2004b). In addition,

    agents that combine non-steroidal anti-inflammatory com-

    pounds with a cholinesterase inhibitor are believed to

    prevent neurotoxicity against Ab. The mechanism of

    protection has been suggested to involve the enhancement

    of non-amyloidogenic APP cleavage that leads to a

    decreased production of endogenous Ab through the Wnt

    pathway (Farias et al., 2005).

    Loss of Wnt activity may lead to cellular injury or

    dysfunction in the CNS during oxidative stress. Wnt1

    expression has been demonstrated in the brains of

    individuals affected by neuropsychiatric disorders (Miyaoka

    et al., 1999). Furthermore, retinal degeneration during

    retinitis pigmentosa with the progressive loss of photo-

    receptors has been associated with increased secretion of

    Frizzled-related protein-2, a Wnt inhibitory protein,

    suggesting that loss of Wnt signaling may contribute toretinal neurodegeneration (Jones et al., 2000). Additional

    work demonstrates that a mutation in the membrane-type

    Frizzled-related protein gene may be involved in retinal

    photoreceptor degeneration (Kameya et al., 2002).

    During AD, neurotoxicity of Ab in hippocampal neurons

    has been linked to increased levels of GSK-3b and loss ofb-

    catenin. Decreased production of Ab can occur during the

    enhancement of protein kinase C (PKC) activity (Savage

    et al., 1998) which may be controlled by the Wnt pathway

    (Garrido et al., 2002). The proteolytic processing of APP

    during AD also has been closely linked to the Wnt pathway

    through presenilin 1 (PS1) and disheveled. PS1 is required

    for the processing of APP and has been shown to down-

    regulate Wnt signaling and interact with b-catenin to

    promote its turnover (Soriano et al., 2001). Disheveled, a

    known downstream transducer of Wnt signaling pathway,

    also can regulate the a-secretase cleavage of APP through

    PKC/mitogen-activated protein kinase dependent pathways,

    increasing soluble production of APP (sAPP) (Mudher et al.,

    2001). Overexpression of mouse disheveled-1 and -2

    inhibits GSK-3b mediated phosphorylation of tau protein

    and may thus prevent formation of neurofibrillary tangles

    during AD (Wagner et al., 1997). Thus, disheveled may

    increase neuronal protection during neurodegenerative

    disorders through sAPP production and reduction in tauphosphorylation.

    7. Akt as an essential regulatory element

    7.1. Activation and expression of Akt

    Protein kinase B (PKB) is ubiquitously expressed in

    mammals but is initially present at low levels in the adult

    brain (Owada et al., 1997). Three family members of this

    serine/threonine kinase are now known to exist that were

    termed Akt after the molecular cloning of the oncogene

    v-Akt and two human homologs (Staal, 1987; Staal et al.,

    1988). Theyare PKBa or Akt1, PKBb or Akt2, and PKBg or

    Akt3 (Chong et al., 2005a). Akt is part of the AGC (cAMP-

    dependent kinase/protein kinase G/protein kinase C) super-

    family of protein kinases and consists of three functional

    domains. The N-terminal pleckstrin homology (PH) domain

    provides binding sites for membrane phospholipids, which

    are involved in the recruitment of Akt to the plasma

    membrane (Frech et al., 1997). The catalytic domain of Akt

    has specificity for serine or threonine residues of proteins

    that are substrates for Akt. It is interesting to note that the

    three isoforms of Akt share the same regulatory phosphor-

    Z.Z. Chong et al. / Progress in Neurobiology 75 (2005) 207246216

  • 7/31/2019 Oxidative Stress in the Brain Novel Cellular Targets That Govern 2005

    11/40

    ylation sites but that splice variants of Akt that lack the

    C-terminal hydrophobic motif (HM) possess lower specific

    activity than full-length isoforms, suggesting that the

    C-terminal HM is vital to stimulate Akt activity (Brodbeck

    et al., 2001; Yang et al., 2002) (Table 2).

    Activation of Akt is dependent upon PI 3-K (Fig. 1). The

    activation of the receptor tyrosine kinase (RTK) and the G

    protein-coupled receptor (CPCR) are required to activate PI

    3-K. Trophic factors or cytokines can stimulate the

    recruitment of PI 3-K to the plasma membrane. Following

    activation, PI 3-K phosphorylates membrane glyceropho-

    spholipid phosphatidylinositol 4,5-bisphosphate [PI(4,5)P2]

    resulting in the production of phosphatidylinositol 3,4,5-

    trisphosphate (PIP3) and phosphatidylinositol 3,4-bispho-

    sphate (PIP2). Both PIP2 and PIP3 bind with equal affinity to

    Akt and are required for Akt activation (Thomas et al.,

    2002). The critical step for activation of Akt is its transitionfrom the cytosol to the plasma membrane, which is

    accomplished by the binding of Akt to PIP2 and PIP3through its PH domain (Stephens et al., 1998). As a result of

    this sequence of events, Akt becomes available for

    phosphorylation by several upstream kinases.

    The phosphorylation of two major residues, Thr308 and

    Ser473, are considered necessary for the activation of Akt.

    The site of Thr308 is located within the activation T-loop of

    Akt1. For Akt2 and Akt3, the equivalent residues are Thr309

    and Thr305, respectively (Walker et al., 1998). These

    phosphorylation sites are believed to be critical for the

    activation of Akt. Yet, the phosphorylation of Ser473 at the

    C-terminal HM domain also is necessary for the complete

    activation of Akt (Bellacosa et al., 1998). The phosphoryla-

    tion of Thr308 is dependent upon its upstream kinase, 3-

    phosphoinositide-dependent kinase-1 (PDK1) (Wick et al.,

    2000). PDK1 cannot directly phosphorylate Ser473, but a

    distinct phosphoinositide-dependent kinase PDK2 (Ser473

    kinase) has been postulated to promote Akt phosphorylation

    on Ser473. The existence of PDK2 is pending further

    confirmation.

    A number of pathways can control the biological activity

    of Akt. Some lipid phosphatases have been shown to

    negatively modulate the activity of Akt. The phosphatase

    and tensin homolog deleted from chromosome 10 (PTEN)

    appears to be a critical regulator of PI 3-K signaling. PTEN

    can dephosphorylate tyrosine-, serine-, and threonine-

    phosphorylated peptides (Lee et al., 1999). PTEN negatively

    regulates PI 3-K pathways by specifically dephosphorylat-

    ing PIP2 and PIP3 at the D3 position (Maehama and Dixon,

    1998). As a result, a reduction in the membrane

    phospholipid pool that is necessary for the recruitment of

    Akt can ensue during PTEN activity.

    Other lipid phosphatases, such as SHIP (SH2 domain-

    containing inositol phosphatase), can regulate Akt activity.

    SHIP is an inositol 50-phosphatase that dephosphorylates

    inositides and phosphoinositides on the 50-position resulting

    in the transformation of PIP3 into PIP2. The SHIP2 gene

    appears to modulate insulin signaling, since targeted

    disruption of this gene leads to increased insulin sensitivity

    that occurs as a result of enhanced phosphorylation of Akt2at the plasma membrane (Sasaoka et al., 2004). In other cell

    systems that involve hematopoietic proliferation, SHIP also

    functions to block activation of Akt (Carver et al., 2000).

    The Src homology domain 2 (SH2)-containing tyrosine

    phosphatases (SHP) also have been implicated in the control

    of the Akt pathway. In regards to SHP1 and SHP2, SHP1 is

    predominantly expressed in hematopoietic cells, but SHP2 is

    more ubiquitously expressed and occurs in the nervous

    system (Chong et al., 2003f). Through the activation of Akt,

    SHP1 can selectively bind and dephosphorylate PTEN to

    reduce the stability of this protein (Lu et al., 2003). SHP2

    also appears to modulate the activation of Akt (Ivins Zito

    et al., 2004) to prevent cellular death from apoptosis through

    inhibition of either caspase 1- or 3-like activities (Chong

    et al., 2003f; Ivins Zito et al., 2004).

    Alternate cellular systems are responsible for the

    enhancement of Akt activity. Carboxyl-terminal modulator

    protein (CTMP) also can negatively regulate the activity of

    Akt. CTMP is a 27 kDa protein that binds specifically to the

    carboxyl-terminal regulatory domain of Akt1 at the plasma

    membrane (Maira et al., 2001). The binding of CTMP to

    Akt1 decreases the activity of Akt1 by inhibiting the

    phosphorylation of Akt1 on Ser473 and Thr308 (Maira et al.,

    2001). The T cell leukemia/lymphoma 1 (TCL1) protein

    Z.Z. Chong et al. / Progress in Neurobiology 75 (2005) 207246 217

    Table 2

    Substrates of Akt that determine apoptotic cell injury

    Substrate Function Selected references

    FOXO3a Activation leads to apoptotic injury, cell cycle progression;

    contributes to oxidative stress; possesses caspase 3 cleavage sequence

    Brunet et al. (1999); Medema et al. (2000);

    Kops et al. (2002) and Chong et al. (2004c)

    GSK-3b Phosphorylates b-catenin, eIF2B, CREB, and tau protein to result

    in apoptosis and the formation of neurofibrillary tangles; promotescytochrome c release, caspase activation

    Somervaille et al. (2001); Kirschenbaum et al. (2001);

    Pap and Cooper (2002) and Koh et al. (2003)

    Bad Oxidative stress activates Bad;, phosphorylation of Bad by Akt blocks

    apoptotic injury, prevents cytochrome c release

    Datta et al. (1997); Simakajornboon et al. (2001);

    Chong et al. (2003b) and Uchiyama et al. (2004)

    NF-kB Leads to the induction of multiple anti-apoptotic genes; blocks caspase

    activity; protects through activation of Bcl-xL

    Wang et al. (1998); Chen et al., (2000);

    De Smaele et al. (2001) and Tang et al. (2001)

    Note: CREB, cAMP-response element-binding protein; eIF2B, the translation initiation factor 2B; GSK-3b, glycogen synthase kinase-b; IKK, IkB kinase;

    JNK, c-Jun-amino terminal kinase; NF-kB, nuclear factor-kB.

  • 7/31/2019 Oxidative Stress in the Brain Novel Cellular Targets That Govern 2005

    12/40

    functions as a co-activator of Akt. TCL1 can stabilize

    mitochondrial membrane potential and promote cell

    proliferation and survival (Laine et al., 2000). TCL1 binds

    to Akt1 and increases Akt1 kinase activity to promote its

    nuclear translocation (Pekarsky et al., 2000). Additional

    work has shown that TCL1 binds to the PH domain of Akt

    and the formation of TCL1 trimers facilitate the formation ofthe Akt/TCL1 complex. Within this complex, Akt is

    phosphorylated and activated in vivo (Laine et al., 2000).

    Akt activity also can be facilitated by a 90 kDa heat shock

    protein (Hsp90). Hsps are characterized by their mass in

    kilodaltons, are induced in response to heat in essentially all

    organisms, and are highly conserved between different

    species. Hsps, such as Hsp90, can be cytoprotective, such

    as preventing cell injury against heat thermal stress (Beere

    et al., 2000; Kalwy et al., 2003; Latchman,2004).Aktbindsto

    Hsp90 through its 229309 residues resulting in stabilization

    of thephosphorylatedAkt. Inhibition of Akt binding to Hsp90

    leads to dephosphorylation of Akt by protein phosphatase 2A

    (PP2A) and induction of apoptosis (Sato et al., 2000).

    Intracellular Akt also can become complexed with Hsp90 and

    Cdc37. As a result of this association,increased Akt activity is

    present but is closely dependentuponthe presence of Hsp90in

    the complex (Basso et al., 2002).

    The cellular expression of Akt can vary in a variety of

    tissues and cells. Akt1 is the most highly expressed isoform.

    Although Akt2 is expressed at a lower level than Akt1,

    significant expression of Akt2 occurs in insulin-responsive

    tissues, such as skeletal muscle, liver, heart, kidney, and

    adipose tissue (Altomare et al., 1995). In the CNS, the

    expression of Akt1 and Akt2 can be observed at increased

    levels during development but is gradually decreased duringpostnatal development (Owada et al., 1997). Yet, in the adult

    brain, expression of Akt1 and Akt2 is initially weak with a

    dramatic increase in the expression of Akt1 mRNA and Akt1

    protein in cells that are subjected to injury (Chong et al.,

    2004a; Kang et al., 2003b; Owada et al., 1997), suggesting

    that Akt may play an important role during cell injury. In

    contrast to Akt1 and Akt2, Akt3 is expressed only in a

    limited number of tissues, such as in the brain and testes,

    with lower expression evident in skeletal muscle, pancreas,

    heart, and kidney (Nakatani et al., 1999).

    7.2. Akt as a modulator apoptotic injury and

    inflammation during ROS exposure

    Akt is a critical survival factor that can modulate cellular

    pathways in both the central and peripheral nervous systems.

    Early studies have demonstrated that overexpression of Akt

    in CNS neurons prevents apoptosis during growth factor

    withdrawal (Datta et al., 1997). Similar investigations that

    employed superior cervical ganglion neurons also illustrated

    that Akt was necessary to prevent cell death during nerve

    growth factor withdraw (Philpott et al., 1997). Additional

    studies have shown that Akt can be both necessary and

    sufficient for the survival of neurons, since expression of a

    dominant-negative Akt or inhibition of PI 3-K yields

    apoptotic cell death during trophic factor administration

    (Crowder and Freeman, 1998) and precipitates cell death

    during oxidative stress (Kang et al., 2003a,b). Akt also

    impacts upon the function and survival of cerebral vascular

    ECs. Recent investigations have shown that Akt modulates

    cerebral blood flow and vasomotor tone (Luo et al., 2000)and prevents apoptotic injury during compromises in

    mitochondrial function and caspase regulation (Chong

    et al., 2002b, 2004a). Further work has illustrated an

    important role for Akt for the survival of cells during a

    number of injury paradigms. Enhanced Akt activity can

    foster cell survival during free radical exposure (Chong

    et al., 2003b; Matsuzaki et al., 1999), matrix detachment

    (Rytomaa et al., 2000), neuronal axotomy (Namikawa et al.,

    2000), DNA damage (Chong et al., 2002b, 2004a; Henry

    et al., 2001; Kang et al., 2003a), anti-Fas antibody

    administration (Suhara et al., 2001), oxidative stress (Chong

    et al., 2003b; Kang et al., 2003a,b; Yamaguchi and Wang,

    2001), hypoxic preconditioning (Wick et al., 2002), Ab

    exposure (Martin et al., 2001), and transforming growth

    factor-b (TGF-b) application (Conery et al., 2004).

    Akt possesses the ability to offer a broad level of

    cytoprotection in cells through both intrinsic cell mechan-

    isms that involve the maintenance of genomic DNA and the

    exposure of membrane PS residues. Through the over-

    expression of a myristoylated (active) form of Akt and a

    kinase-deficient dominant-negative Akt, recent work has

    shown that Akt is both necessary and sufficient to protect

    cells, such as neurons and ECs from injury associated with

    oxidative stress (Chong et al., 2003b; Kang et al., 2003a,b).

    Overexpression of myr-Akt significantly protects cells fromfree radical injury and prevents degradation of genomic

    DNA (Fig. 2). Yet, cells with a dominant-negative

    overexpression that lack kinase activity suffer a significant

    loss in cell survival during oxidative stress. Further studies

    have suggested that through the inhibition of PI 3-K

    phosphorylation of Akt or through the overexpression of a

    kinase-deficient dominant-negative Akt, endogenous cellu-

    lar reserves of Akt also can provide an additional level of

    protection during cell injury that can function in concert

    with the exogenous activation of Akt to achieve increased

    cellular protection (Chong et al., 2004a; Kang et al.,

    2003a,b). It is important to note that activation of Akt is not

    always desirable. Under some conditions, enhanced cellular

    survival during Akt activation in cells that are targeted for

    destruction, such as in neoplastic cells, could undermine

    treatment as well as foster the growth of a neoplasm. As a

    result, recent work has identified Akt as a potential target to

    block during the treatment of non-small cell lung cancers

    that contain mutations in the epidermal growth factor

    (Sordella et al., 2004).

    Akt prevents inflammatory cell demise through extrinsic

    cellular mechanisms that involve membrane PS exposure

    and the subsequent activation of microglia. Enhanced Akt

    activity can prevent cellular membrane PS externalization in

    Z.Z. Chong et al. / Progress in Neurobiology 75 (2005) 207246218

  • 7/31/2019 Oxidative Stress in the Brain Novel Cellular Targets That Govern 2005

    13/40

    both neurons and ECs during a variety of insults that involve

    anoxia, free radical exposure, and oxygen-glucose depriva-

    tion (Chong et al., 2002b, 2004a; Maiese et al., 2004). In

    addition, Akt appears to employ the modulation of

    membrane PS externalization to prevent microglial activa-

    tion (Kang et al., 2003b). Activation of Akt can prevent

    membrane PS exposure on injured cells and block the

    activation of microglia that are exposed to media taken from

    cells that overexpress active, phosphorylated Akt during

    cellular injury (Kang et al., 2003a,b). Cytoprotective agents,

    such as nicotinamide and erythropoietin (EPO), also employ

    mechanisms that involve Akt to regulate microglial

    activation and proliferation (Chong et al., 2003d; Liet al., 2004b; Maiese et al., 2004). These protective agents

    block membrane PS exposure on cells and possibly prevent

    the shedding of membrane PS residues that is known to

    occur during apoptosis (Simak et al., 2002). In addition to

    targeting the activity of membrane PS exposure and

    microglial activation, Akt also may directly address cellular

    inflammation by inhibiting several pro-inflammatory cyto-

    kines, such as TNF-a (Fontaine et al., 2002).

    In addition to its ability to protect cells against apoptotic

    and inflammatory injury, Akt can function to either reduce or

    prevent cellular destruction from ROS. For example, ROS

    generated by hydrogen peroxide can lead to the endogenous

    activation of Akt in several cell lines such as Hela, A549, and

    MCF-7 cells (Wang et al., 2000). In human glioblastoma cell

    lines, hydrogen peroxide also leads to a marked phosphor-

    ylation of Akt (Sonoda et al., 1999). Generation of

    peroxynitrite by sodium nitrite and acidic hydrogen

    peroxide also results in a time and dose-dependent activation

    of Akt followed by inactivation of GSK-3b in human skin

    primary fibroblast cells (Klotz et al., 2000). Akt activation

    during ROS in several neuronal and vascular cell systems

    has been demonstrated in neuronal cell lines (Kang et al.,

    2003a,b; Salinas et al., 2003), primary hippocampal and

    cortical neurons (Chong et al., 2003b,e; Crossthwaite et al.,

    2002; Matsuzaki et al., 1999) and cerebral vascular ECs

    (Chong et al., 2002b, 2004a).

    7.3. Akt can provide the stimulus for altering the

    course of neurodegenerative disease

    As a result of the broad protective nature of Akt, it may

    come as no surprise to learn that many agents or growth

    factors appear to prevent apoptotic cellular injury through

    Akt activation. In the vascular system, angiopoietin-1 is an

    endothelium-specific ligand essential for embryonic vas-

    cular stabilization, branching, morphogenesis, and post-

    natal angiogenesis. Angiopoietin-1 also supports endothelialcell survival and prevents apoptosis through the activation of

    Akt that requires a PI 3-K dependent pathway (Papape-

    tropoulos et al., 2000). Furthermore, in the cardiovascular

    system, myocardial protection by insulin during myocardial

    ischemia/reperfusion is abolished by PI 3-K inhibition,

    suggesting that cardioprotection of insulin is mediated by

    Akt activation (Jonassen et al., 2001; Li et al., 2004a; Maiese

    et al., 2005). The involvement of the PI 3-K/Akt pathway

    also has been demonstrated during the protection of retinal

    ganglion cells from axotomy (Kermer et al., 2000).

    A number of trophic factors and cytokines, such as EPO,

    may depend upon Akt to offer cellular protection ( Maiese

    et al., 2003). EPO can phosphorylate Akt and is dependent

    upon the activation of PI 3-K and Janus kinase 2 (Jak2)

    (Chong et al., 2002b; Witthuhn et al., 1993). Activation of

    Jak2 promotes the phosphorylation of tyrosine residues in

    the intracellular portion of the EPO receptor (Witthuhn et al.,

    1993). Phosphorylation of the last tyrosine of the EPO

    receptor initiates binding of the 85 kDa regulatory subunit of

    PI 3-K, a heterodimer consisting of a 110 kDa catalytic

    subunit and an 85 kDa regulatory subunit. As a result of the

    binding of the 85 kDa regulatory subunit, the 110 kDa

    catalytic subunit becomes active and leads to the phosphor-

    ylation of Akt.

    Z.Z. Chong et al. / Progress in Neurobiology 75 (2005) 207246 219

    Fig. 2. Overexpression of Akt1 prevents cellular injury and DNAdegradation during oxidative stress. Representative images illustrate DNAfragmentationwith

    TUNEL and cell survival with a trypan blue dye exclusion (TB) methods in both wild-type and myristolated (myr)-Akt1 (active Akt1) transfected cerebral

    microvascular endothelial cells (ECs) 24 h followingexposure to a NO donor (NOC-9, 1000mM).NO induced DNA fragmentation andTB stainingwas evident

    in wild-type cells (wild-type/NO), but is absent in cells overexpressing Akt1 (myr-Akt1/NO).

  • 7/31/2019 Oxidative Stress in the Brain Novel Cellular Targets That Govern 2005

    14/40

    Central to the ability of EPO to prevent cellular apoptosis

    is the activation of Akt by EPO (Maiese et al., 2004). During

    anoxia or free radical exposure, expression of the active

    form of Akt (phospho-Akt) is increased (Kang et al.,

    2003a,b). EPO can significantly enhance the activity of Akt

    during oxidative stress and prevent inflammatory activation

    of microglia (Chong et al., 2003a,b,e). This up-regulation ofAkt activity during injury paradigms appears to be vital for

    EPO protection, since prevention of Akt phosphorylation

    blocks cellular protection by EPO (Chong et al., 2003a,b,e).

    Through the regulation of the PI 3-K/Akt dependent

    pathway, EPO can prevent cellular apoptosis following N-

    methyl-D-aspartate toxicity (Dzietko et al., 2004), hypoxia

    (Chong et al., 2002b), and oxidative stress (Chong et al.,

    2003a,b,e).

    Given the intimate association between Akt and

    cytoprotective agents, Akt may be viewed as an essential

    target for therapeutic strategies against a number of diseases

    that involve apoptotic cell death. The association of familial

    AD with mutations in APP suggests that wild-type APP may

    have a protective ability against toxic insults to cells, since

    mutations in APP impair its ability to offer resistance against

    oxidative stress. Recent work has suggested that protection

    by wild-type APP against ROS may require the PI 3-K/Akt

    pathway, since dominant-negative forms of Akt eliminated

    the protective capacity of wild-type APP (Kashour et al.,

    2003). Furthermore, overexpression of Akt1 can attenuate

    apoptosis during Ab exposure (Martin et al., 2001). In

    models of Parkinsons disease that employ the neurotoxin 1-

    methyl-4-phenylpyridinium (MPP+), apoptosis was pre-

    vented and a reduction in ROS was observed in cells

    overexpressing an active form of Akt (Salinas et al., 2001).Activation of Akt during acute cellular insults also

    appears to be necessary to foster cell survival. Phosphoryla-

    tion of Akt has been observed in the brain following either

    focal or global cerebral ischemia (Friguls et al., 2001; Yano

    et al., 2001). Sublethal ischemic induction during pre-

    conditioning experiments leads to the phosphorylation of

    Akt in the hippocampal CA1 region. This activation of Akt

    was not present in ischemic animals that did not receive

    sublethal ischemic preconditioning and led to a greater

    degree of cerebral infarction, suggesting that Akt activation

    provides an important mechanism for ischemic precondi-

    tioning (Yano et al., 2001). Cell culture experiments also

    have supported that hypoxic preconditioning may be

    mediated by the activation of Akt (Wick et al., 2002).

    8. Downstream cellular targets

    At this point of time, no definitive therapy for either acute

    or chronic neurodegenerative diseases is available. Yet,

    investigations into the cellular pathways that determine

    oxidative stress and apoptotic injury have begun to elucidate

    pathways that provide us with a clearer understandin