Hayley curr pharm design

download Hayley curr pharm design

of 49

Transcript of Hayley curr pharm design

  • 8/7/2019 Hayley curr pharm design

    1/49

    Multiple mechanisms of cytokine action in neurodegenerative and psychiatric states:

    neurochemical and molecular substrates.

    *S. Hayley & H. Anisman

    Institute of Neuroscience, Carleton University, Ottawa, Ontario, Canada

    Correspondence to: S.H., Carleton University, A511 Loeb Bld, 1125 Colonel By Drive, Ottawa,

    ON, Canada, K1S 5B6;

    Phone: (613) 520-2600x6314; FAX: (613) 520-4052; email: [email protected]

    Key words: cytokine, sensitization, depression, Parkinsons disease, neurodegeneration,

    microglia, neurochemical, neurotoxin

  • 8/7/2019 Hayley curr pharm design

    2/49

    Hayley 2

    Abstract

    Neuroinflammatory processes appear to play a fundamental role in the pathology associated with

    a number of neurodegenerative and psychiatric conditions. In this respect, the immunocompetent

    brain microglia and peripheral macrophages release a host of proinflammatory cytokines that not

    only modulate immunological processes but also influence neuronal functioning and even

    survival. For instance, alterations of the cytokines, tumor necrosis factor-, as well as several of

    the interferons and interleukins have been associated with Parkinsons disease (PD) and clinical

    depression. Importantly, anti-inflammatory treatments that block these cytokines may impart

    protection against behavioural pathology and neuronal damage in animal models of PD and

    depression involving exposure to environmental toxins and stressors, respectively. The present

    review highlights the involvement of inflammatory cells and cytokines in depression and PD and

    explores some of the potential cellular and molecular mechanisms through which the

    immunotransmitters affect neuronal functioning. Attention is also devoted to the possibility that

    cytokines may sensitize neuroinflammatory pathways that, in turn, favour long-term pathology.

  • 8/7/2019 Hayley curr pharm design

    3/49

    Hayley 3

    Introduction

    The articles presented in this issue of Current Pharmacological Therapeutics deals with the

    influence of immune system messengers (cytokines) on central neuronal functioning, and

    whether such effects are responsible for centrally mediated clinical conditions. Inasmuch as

    major depressive illness and Parkinsons disease (PD) are highly comorbid, the present series of

    articles focus on these states. It ought to be underscored from the outset that comorbidity may

    occur for any number of reasons. For instance, one illness may directly or indirectly favour the

    development of the second pathology, or alternatively, common processes give rise to both

    illnesses. In the case of major depressive disorder and PD, inflammatory immune factors have

    been implicated as a provocative factor, and there is reason to suspect that these come about

    through a variety of environmental triggers, including physical or psychological distress. In the

    present review we offer the position that the immune messengers, pro-inflammatory cytokines,

    contribute to both these conditions (Figure 1), and that their sensitizing effects on central

    processes may be particularly important.

    We have argued that, like stressors, cytokine exposure or cytokine inducing immune

    challenges can sensitize central nervous system (CNS) reactivity to subsequent insults. For

    example, a single injection of either tumour necrosis factor- (TNF-) or the potent pro-

    inflammatory bacterial endotoxin, lipopolysaccharide (LPS), sensitized CNS functioning, such

    that later reexposure to these challenges provoked greatly augmented neurochemical and

    behavioural disturbances [1,2,3]. Interestingly, many of these effects were reminiscent of the

    neurovegatative and biological effects associated with depression and stressor induced disorders.

    In addition to the potential cognitive and mood alterations elicited, cytokines and

    neuroinflammatory processes have been implicated in the neurodegeneration observed in

  • 8/7/2019 Hayley curr pharm design

    4/49

    Hayley 4

    response to acute trauma as well as progressive disease states, including stroke, head injury,

    seizure, Alzheimers disease, Multiple sclerosis (MS) and Parkinsons disease [4,5,6,7].

    Furthermore, experimental findings and epidemiological studies have raised the possibility that

    viral and/or bacterial insults encountered early in life may increase the likelihood of

    subsequently developing PD [8,9], possibly by sensitizing neurons to the deleterious effects of

    various environmental insults. In this regard, cytokines may be acting as a mediator of the

    protracted consequences of diverse inflammatory challenges. It is our contention that if cytokine

    exposure is coupled with other environmental insults, then neuronal systems may become

    overly taxed (allostatic overload) favoring the evolution of neurodegeneration or behavioural

    pathology. In this review we highlight some of the possible molecular pathways (e.g. MAP

    kinases) involved in transducing cytokine signals into messages that influence the decision

    making processes within cells, and which may be fundamental in promoting pathology related

    to neuroinflammation.

    Cytokines and psychiatric states: Depression

    Stress-cytokine connection

    Through disturbances of neuroendocrine and neurotransmitter functioning, stressors are

    thought to be fundamental in the provocation or exacerbation of affective disorders [10,11]. Like

    other stressors, cytokines such as IL-1, IL-6 and TNF- engender a host of behavioural,

    neuroendocrine and central neurotransmitter changes, including increased plasma levels of

    glucocorticoids and ACTH (adrenocorticotropin) coupled with augmented turnover of

    monoamines within hypothalamic and extrahypothalamic regions [10, 12, 13]. It has been our

    contention that by virtue of such neurochemical alterations, cytokines may contribute to

    depressive illness.

  • 8/7/2019 Hayley curr pharm design

    5/49

    Hayley 5

    Given the similarity of the neurochemical changes elicited by cytokines and by traditional

    stressors, Herman & Cullinan (1997) suggested that systemic insults, which include cytokines

    and other insults that promote circulatory, respiratory or hemodynamic alterations, may have

    stressor-like characteristics, and could potentially influence disease processes in ways that

    stressors ordinarily have such effects. Of course, the effects of systemic and processive stressors

    (the latter include events or stimuli that involve information processing) are not identical, and

    cytokines may stimulate HPA activity and other central functions through pathways somewhat

    different from those utilized by psychogenic (e.g. predator exposure, restraint) or neurogenic

    (e.g., painful stimuli) stressors. In fact, both IL-1 and footshock provoked a similar activation (as

    indicated by c-fos expression) of hypothalamic and cortical nuclei, however, the pattern of c-fos

    expression varied greatly between the challenges within several limbic system regions, including

    the amygdala and bed nucleus of the stria terminals [14].

    Besides acting in a manner similar to that of stressors, cytokines may in fact mediate some

    of the neurochemical changes elicited by stressors. For instance, administration of the IL-1

    antagonist, IL-1ra, attenuated the hypothalamic NE, DA and 5-HT alterations, as well as the

    ACTH elevation elicited by immobilization stress [15], suggesting that IL-1 mediates the effects

    of stressors on these neuronal processes. Furthermore, hypothalamic levels of IL-1 are increased

    in response to both immobilization and electric shock [15, 16] as well as in the hippocampus and

    NTS in response to acute inescapable shock [17, 18] .

    Given that proinflammatory cytokines alter monoamine functioning within several mood

    regulatory brain regions, such as the hypothalamus, central amygdala and medial prefrontal

    cortex, the proposition has been made that these factors may be fundamental in depressive

    illnesses [19,20]. Interestingly, these monoamine changes are subject to the synergistic effects of

  • 8/7/2019 Hayley curr pharm design

    6/49

    Hayley 6

    stressors and cytokines, such that amine release from limbic neurons is dramatically increased in

    IL-1 treated rodents also exposed to a mild stressor [21]. Curiously, however, the bulk of the

    available data have been limited to the effects of acute cytokine/immune activation and limited

    data are available concerning the influence of chronic activation of the inflammatory immune

    system, despite the fact that immune activation rarely occurs on a transient basis.

    Animal models: Sickness and Depression

    Animal studies have shown that immunogenic challenges provoke a sickness syndrome that

    is though to reflect many of the neurovegatative symptoms of depression. Along these lines,

    Smith (1991) proposed that cytokines released from macrophages instigate the onset of

    depression, and may account for the high comorbidity between depression and inflammatory

    related illnesses, such as heart disease, arthritis, stroke, Alzheimers disease and Parkinsons

    disease [22]. As well, the greater incidence of depression in women than men (3:1) may be

    associated with the fact that estrogen is a potent macrophage activator [22]. Thus, it follows that

    infections, tissue injury and environmental insults that promote inflammation may all impact

    upon depression through macrophage related functioning.

    Activation of macrophages and other inflammatory immune cells following systemic LPS

    treatment results in the manifestation of a constellation of behavioral symptoms collectively

    referred to as sickness behavior [23], which may mimic some of the neurovegatative symptoms

    of depression. Symptoms include reduced locomotion, increased sleep and curled body posture

    thereby minimizing energy expenditure, as well as elevated body temperature coupled with

    anorexia, which may serve to make the physiological environment unfavourable to an invading

    pathogen [23]. The cytokines IL-1 and TNF- also elicit sickness behaviors and IL-1 antagonists

    attenuate these effects [12]. Moreover, cytokines disrupt operant responding for reinforcement

  • 8/7/2019 Hayley curr pharm design

    7/49

    Hayley 7

    and reduce social exploration, with the latter effects being secondary to illness or stemming from

    changes in motivational state (e.g., anhedonia) [24]. However, studies employing progressive

    ratio schedules of reinforcement (i.e., these tap into the degree of to which an animal is willing to

    respond in order to gain a reward) indicated that cytokines act on incentive motivational forces

    independent of effects related to sickness [24]. Moreover, antidepressant treatment effectively

    attenuated the decreased responding for a palatable snack elicited by IL-1, but without

    influencing the reduced chow intake ordinarily elicited by the cytokine [24].

    Animal models: Neurochemical effects of cytokines

    As indicated earlier, cytokine administration provokes hypothalamic pituitary adrenal

    (HPA) activation as well as augmented monoamine utilization in several brain regions implicated

    in depression. Indeed, systemic and central IL-1 administration increased the expression and

    secretion of CRH and arginine vasopressin (AVP) from PVN neurons of the hypothalamus [25]

    and direct infusion of IL-1 into the median eminence (site of CRH terminals from neurons

    originating within the PVN) increased AVP and CRH secretion [26]. Correspondingly, IL-1

    increased ACTH secretion from the pituitary corticotrophes and subsequent adrenal

    corticosterone release [26]. Similar to IL-1, systemic TNF- elevated median eminence CRH

    release, as well as circulating ACTH and corticosterone levels [1,2,3,20, 27]. Although central

    infusion of TNF- was reported to dose-dependently increase circulating ACTH levels, this

    cytokine had relatively modest effects on plasma corticosterone [2,3]. While much evidence

    suggests that TNF- may stimulate HPA activity by direct actions upon the corticotropic cells

    within the PVN [28,29], it was reported that the cytokine may affect HPA processes through

  • 8/7/2019 Hayley curr pharm design

    8/49

    Hayley 8

    actions outside the brain [30]. In this regard, we found that the vasoactive amine, histamine, may

    be important for some of the protracted effects of TNF- upon HPA activity [31].

    Like psychogenic and neurogenic stressors, subpyrogenic doses of IL-1 increased

    accumulation of the 5-HT metabolite, 5-HIAA, within the PVN, central amygdala, and medial

    prefrontal cortex [32]. Likewise, in vivo studies indicated that systemic IL-1 increased

    hypothalamic NE release as well as that of 5-HT at the nucleus accumbens and the hippocampus

    [21]. When centrally administered, IL-1 increased hypothalamic release of NE, 5-HT and DA,

    as well as that of NE and 5-HT within the prefrontal cortex and hippocampus, respectively

    [33,34]. Although less data are available concerning TNF-, systemic administration of the

    cytokine was demonstrated to increase NE activity within the PVN, central amygdala, dorsal

    hippocampus and locus coeruleus, while 5-HT utilization was increased within the PVN, medial

    prefrontal cortex, hippocampus and central amygdala [35, 32]. In vivo, icv TNF- administration

    increased plasma corticosterone, but did not influence hippocampal 5-HT release [36].

    Clinical evidence for cytokines in depression

    Depressed patients, particularly those presenting with melancholic illness, exhibit

    disturbances of several aspects of immune functioning. For instance, severely depressed mood

    was accompanied by altered circulating lymphocyte subsets, reduced mitogen-stimulated

    lymphocyte proliferation, and impaired natural killer (NK) cytotoxicity, although equivocal

    results have been reported [37]. However, depression was also associated with immune

    activation reminiscent of an acute phase response, with increased plasma concentrations of

    complement proteins, C3 and C4, IgM, and positive acute phase proteins, haptoglobin, 1-

    antitrypsin, 1 and2 macroglobulin, and reduced negative acute phase proteins [38]. Thus,

  • 8/7/2019 Hayley curr pharm design

    9/49

    Hayley 9

    affective disturbances might be secondary to activation of some components of the immune

    response. Consistent with this view, depressive illness was associated with elevated circulating

    levels of cytokines and/or their soluble receptors, including IL-2, soluble IL-2 receptors (sIL-

    2R), IL-1, IL-1 receptor antagonist (IL-1Ra), IL-6, soluble IL-6 receptor (sIL-6R), and -

    interferon (IFN) [38, 39]. Moreover, increased production of IL-1, IL-6 and TNF- wasevident in mitogen-stimulated lymphocytes [40]. As already indicated, the cytokine changes may

    have been related to severity of illness, and may have been a reflection of the duration of illness

    or the age of illness onset [41].

    While the relationship between cytokines and depression in the aforementioned studies are

    based on correlational analyses, it has also been shown that direct administration of cytokines or

    immune challenges to humans provokes depressive-like symptomatology. Indeed, healthy

    volunteers administered a low dose of LPS or vaccinated with live attenuated rubella virus

    displayed depressed mood up to 10 weeks following the challenge and in the case of the

    endotoxin protracted anxiety and memory deficits were provoked [42]. Of considerable clinical

    importance, is the use of cytokines as immunotherapeutic treatments for various cancers and

    viral conditions. In this respect, cancer patients undergoing IL-2 or IFN- immunotherapy often

    display depressive-like symptoms [43]. This point has been emphasized by Maes (1992,1999) in

    which he indicates that IFN- treatment for hepatitis C or melanoma resulted in neuropsychiatric

    symptoms, including fatigue, sleep disturbances, irritability, appetite suppression and depressed

    mood. As well, IFN- as well as several other proinflammatory cytokines (TNF-, IL-1, IFN-)

    were reported to increase levels of the 5-HT transporter which may decrease extracellular 5-HT

    levels [44,45]. Alternatively, IFN- may reduce 5-HT levels through its enzymatic alterations

    (e.g. stimulation of indoleamine 2,3-dioxygenase) that favour reduced plasma levels of the 5-HT

  • 8/7/2019 Hayley curr pharm design

    10/49

    Hayley 10

    precursor, trypthophan (see Figure 1) [46]. Hence, these cytokines can potentially mimic the

    neurotransmitter changes observed in clinical depression. It is particularly significant, from both

    a heuristic and a practical perspective, that depressive symptoms elicited by IFN- are attenuated

    among patients that received conventional antidepressant treatments [43].

    Neurodegenerative aspects of Depression

    The enduring belief that depression is a biochemical disorder unrelated to neuronal survival

    is changing with the demonstration of some degree of impaired neuronal survival evident in this

    disorder. Indeed, imaging studies have demonstrated reduced hippocampal volume in the brains

    of depressed patients and post-mortem analyses revealed a positive relationship between duration

    of depression and atrophy of the hippocampus [47,48]. The exact mechanisms responsible for

    such hippocampal atrophy are not clear, but several possibilities have been advanced, including a

    reduction of neurogenesis, reduced dendritic branching as well as excitotoxic and apoptotic death

    mechanisms. Although many of these effects are linked to the excessive glucocorticoid levels

    associated with depression, cytokine mediated neuroinflammatory processes may also be

    involved.

    Neurodegenerative aspects of Depression: Mechanisms of death

    Several studies reported a reduction of neurogenesis (or new cellular growth) within the

    hippocampus of postmortem tissue obtained from patients suffering from major depression [49].

    Similarly, rodents faced with chronic stressors or corticosterone treatments displayed impaired

    hippocampal neurogenesis [50]. The fact that this impairment was reversed by chronic

    antidepressant or a single electroconvulsive shock (ECS) treatment indicates that induction of

    neurogenesis may be a clinically important event [51]. Although corticoids have been reported to

    reduce neurogenesis [52], other factors may also provoke such effects in depressed individuals.

  • 8/7/2019 Hayley curr pharm design

    11/49

    Hayley 11

    Indeed, chronic ECS increased neurogenesis above basal levels, even in the presence of elevated

    corticoids [53]. As well, the antidepressant, fluoxetine, normalized hippocampal neurogenesis in

    animals exposed to inescapable electric shock independent of any actions upon corticosterone

    [54]. Interestingly, drugs that inhibited either of the CRH1 and AVP1b receptors prevented the

    reduction of neurogenesis evident in the chronic mild stress model of depression [55], suggesting

    an important role for these neuropeptides in hippocampal changes evident in depression.

    Hippocampal atrophy associated with clinical depression may be related to the direct actions

    of glucocorticoids. Indeed, these hormones have been reported to impair glucose transport into

    hippocampal pyramidal neurons resulting in an energetic compromise of these cells [56].

    Likewise, glucocorticoids can elevate free cytocolic calcium concentrations and provoke

    alterations of NMDA and/or AMPA glutamate receptors linked to excitotoxic processes [56].

    Excessive glucocorticoid levels have also been reported to contribute to free radical

    accumulation by reducing the capacity of antioxidant enzymes [57]. Each of these direct

    corticoid mediated effects may lead to either hippocampal neuronal demise or a regression of

    pyramidal neuron dendritic branching [57]. In any case, both situations would result in a

    reduction of hippocampal volume similar to that observed in chronically depressed patients.

    Indirect evidence has indicated the possibility that apoptotic processes may be operative in

    depression. In this respect, postmortem analysis revealed that 11 out of 15 depressed patients

    displayed, albeit modest, in situ end-labeling for DNA fragmentation [58]. However, data

    concerning the expression of the prototypical apoptotic initiators, Fas, p53, Bax and downstream

    caspases are lacking for clinical depression. A recent report did indicate an increased Bax/bcl-2

    ratio in the brains of schizophrenic patients suggested in increased vulnerability to apoptotic

    activation [59]. Animal studies have also indicated that stressor exposure reduced expression of

  • 8/7/2019 Hayley curr pharm design

    12/49

    Hayley 12

    the anti-apoptotic factor, bcl-2 [52,60], and that exposure to a severe stressor exacerbated infarct

    size in response to ischemia through the suppression of bcl-2 expression [52].

    Neurodegenerative aspects of Depression: Molecular pathways

    Stressor provoked reductions of bcl-2 expression have been linked to altered neurotrophic

    factor levels and activation of mitogen activated protein (MAP) kinase pathways [60]. In this

    regard, the neurotrophic cytokine, brain derived neurotrophic factor (BDNF), was reduced in the

    serum of depressed patients and the reduction negatively correlated with the degree of clinical

    impairment, as determined by the Hamilton Rating Scale for Depression [61]. Likewise, rodents

    exposed to a restraint stressor displayed reduced BDNF expression and this effect was prevented

    by antidepressant treatment [62]. Attesting to the importance of BDNF in depression, it has

    become clear that several clinically beneficial treatments for depression increase BDNF

    expression, including selective serotonin reuptake inhibitors (SSRIs), tricyclics and ECS therapy

    [63]. An upregulation of BDNF expression was also associated with improved performance of

    rats in the forced swim test, which is used as a model of behavioural despair [63].

    The underlying molecular pathways operative in models of depression are currently being

    elucidated. As alluded to earlier, MAP kinase signaling pathways may be important for BDNF

    and bcl-2 functioning in depression. The three MAP kinase pathways, (1) extracellular signal-

    regulated kinase (ERK), (2) c-Jun N-terminal kinase (JNK; also know as stress-activated protein

    kinase) and (3) p38, play an important role in responding to environmental events through the

    transmission of synaptic signals to the nucleus [64]. Essentially, these pathways involve a series

    of enzymes that sequentially phosphorylate each other to promote transcriptional activation and

    synthesis of proteins important for cellular survival/death as well as inflammatory processes.

    Importantly, BDNF and bcl-2 promote many of their physiological functions through stimulation

  • 8/7/2019 Hayley curr pharm design

    13/49

    Hayley 13

    of MAP kinase MAP signaling pathways [65,66]. Likewise, antidepressants, which can

    normalize the stressor-induced reductions of BDNF, bcl-2 and cerebral catecholamines, also

    modulate MAP kinase activity [67]. Thus, MAP kinase signaling has obvious implications for

    depressive conditions, which are often precipitated by chronic stressors and characterized by

    altered brain amine levels.

    Neurodegenerative aspects of Depression: Cytokine involvement

    As already indicated, it remains to be determined if true neurodegeneration occurs in

    depression. Certainly, cytokines and other neuroinflammatory factors would be in a position to

    influence such cellular viability. Cytokines may mediate neurodegeneration in depression

    through the promotion of oxidative or excitototic factors derived from metabolic toxins [46]. In

    this respect, IFN-, IFN- and TNF- have been reported to promote tryptophan metabolism

    into kynurenine and subsequently into the oxidative metabolites, 3-hydroxy-kynurenine and

    quinolinc acid, which themselves are increased in depression [68]. Chronic IFN- treatment

    produced depressive symptomology that was associated with increased kynurenine and

    decreased troptophan serum levels (Figure 1) [68]. Likewise, enhanced circulating IL-6 and IL-8

    concentrations were correlated with elevated levels of kynurenine toxic metabolites [46].

    Interestingly, these kynurenine metabolites can synergistically induce free radical generation and

    have been implicated in a number of neurodegenerative disease including Huntingtons disease,

    Parkinsons disease and AIDS dementia [46].

    It will be recalled that macrophage activity was posited to influence mood states through

    the release of cytokines following inflammatory challenges [22]. Accordingly, the common

    myeloid lineage and striking similarity of functioning between peripheral macrophages and brain

    microglia raise the possibility of common involvement of these cell types in depression. Indeed,

  • 8/7/2019 Hayley curr pharm design

    14/49

    Hayley 14

    microglia are the main central reservoirs of proinflammatory cytokines, and like macrophages,

    microglia can act as antigen presenting cells within the CNS [69], thereby potentially

    orchestrating central immune responses that may have deleterious consequences to local tissue.

    Consistent with a role for microglia involvement in cognitive and mood processes, they release

    several interleukins (IL-1, IL-6, IL-8), TNF- and IFN- in response to a host of stressful and

    traumatic stimuli (e.g. stroke, head injury, seizure), as well during the course of

    neurodegenerative diseases (e.g. PD, Alzheimers disease, MS) [69,70].

    Unfortunately, postmortem analysis of cytokines and inflammatory factors in depressed

    individuals is lacking. However, it is interesting that a strong link exists between

    neurodegenerative disorders with an inflammatory component and depression. In fact, a high

    degree of co-morbidity is evident between depression eeeand Parkinsons disease (~40%) [71].

    Although it may be the case that the depressive symptomatology is a reaction to the stress and

    uncertainty associated with facing and coping with a debilitating disease, degeneration of

    neurons involved in regulation of mood may also be important. Indeed, epidemiological

    evidence suggests that the onset of depression often precedes the diagnosis of PD [72]. Thus,

    low levels of degeneration in monoaminergic regions important for emotionality and reward

    processes (e.g. within locus coeruleus, ventral tegmental area) that occur long before the onset of

    PD motoric disturbances may precipitate depressive pathology.

    Neurodegenerative aspects of Depression: Conclusions

    Although it seems likely that some degree of neuronal atrophy accompanies depression the

    question still remains as to whether such changes are a cause or consequence of the disorder. In

    this respect, it seems highly probable that the high circulating levels of glucocorticoids and

  • 8/7/2019 Hayley curr pharm design

    15/49

    Hayley 15

    sympathetic transmitters (epinephrine) associated with the disorder would eventually induce

    some degree of cellular loss. Likewise, chronically dysregulated brain monoaminergic systems

    would also be a potential candidate for eventual energetic, metabolic or other derangements that

    would leave cells vulnerable to degeneration. In such situations the degeneration would be

    secondary to the depressive condition, however, the possibility should not be dismissed that an

    initial slowly developing, mild degree of degeneration would eventually produce disturbances in

    mood regulatory circuits producing depressive symptomatology. In this respect,

    neurodegenerative disorders, such as Parkinsons disease, are often associated with depression at

    early stages before widespread degeneration and subsequent motor difficulties [72].

    Cytokines and Neurodegeneration: Parkinsons disease:

    Cytokines have been implicated in acute and chronic cell death [6,73]. Clinical studies

    revealed increased levels of the proinflammatory cytokines in postmortem brain as well as in

    blood of patients with stroke, head injury, multiple sclerosis, Alzheimers and Parkinsons

    disease [6,73,74,75,76]. Although these findings have been recapitulated in animal models, as

    indicated earlier it is still uncertain whether these cytokines play a neuroprotective or

    neurodestructive role. It may be that relatively low endogenous cytokine levels act in a protective

    capacity to buffer against damage related to death processes, whereas relatively high levels of

    these factors may contribute to neuronal damage [76]. Indeed, low levels of cytokines can

    provoke the release of potentially beneficial trophic factors and free radical scavengers, but

    elevated levels may activate inflammatory cascades or even induce apoptotic death (self

    destructive programmed death mechanism). For instance, mice genetically lacking TNF-

    receptors (thereby removing the influence of endogenous TNF-) were more susceptible to

    ischemic injury, but administration of exogenous TNF- at the time of ischemia exacerbated

  • 8/7/2019 Hayley curr pharm design

    16/49

    Hayley 16

    neuronal death [76]. Likewise, administration of the endogenous IL-1 antagonist, IL-1ra, reduce

    infarct size in response to middle cerebral artery occlusion and prevented the accumulation of

    inflammatory infiltrates within the area of damage [73], suggesting a prominent destructive role

    for IL-1 in acute cerebrovascular insults. In effect, the concentration as well as timing of

    cytokine exposure likely determines whether primarily protective or deleterious consequences

    arise from these immunotransmitters.

    Environmental stressors in PD: Animal models

    Parkinsons disease is characterized by degeneration of the dopaminergic neurons within

    the SNc thereby promoting a reduction of dopamine release from the terminals within the

    striatum [77, 78]. The clinical features of PD, including bradykinesia, tremor and rigidity, stem

    from the dysregulation of basal ganglia functioning associated with the reduced dopamine levels

    [79]. Indeed, a dis-inhibition of striatal interneurons provides faulty input to the globus pallidus

    and thalamus ultimately culminating in reduced drive to motor regulatory cortical regions [80].

    The two most commonly used and widely validated animal models of PD are those involving

    MPTP and 6-OHDA administration. Essentially, MPTP is a thermal breakdown product of a

    meperidine-like form of synthetic heroin, that was accidentally discovered to induce

    Parkinsonism in a group of drug users in the early 1980s [79,81]. Systemic exposure to MPTP

    has been demonstrated in numerous studies over the past two and a half decades to provoke SNc

    dopaminergic degeneration coupled with depletion of striatal dopamine in mice and primates

    [79,82]. Although MPTP elicits behavioral disturbances (e.g. akinesia, tremor, impaired gait)

    similar to those evident in clinical PD, a threshold of neuronal loss (estimated around 80%) may

    have to be evident before such effects are manifested [83]. In contrast to MPTP, 6-OHDA is not

    able cross the BBB and is consequently typically directly infused into the either the SNc or

  • 8/7/2019 Hayley curr pharm design

    17/49

    Hayley 17

    striatum where it exerts substantial destructive effects on local neurons and terminals [84]. 6-

    OHDA is a hydroxylated analogue of dopamine that may be generated by auto-oxidation of

    endogenous dopamine [85]. Consequently, the highly reactive metabolic nature of dopamine

    itself may contribute to PD neurodegeneration.

    Although rare familial forms of PD appear to have a strong genetic component [86], the

    majority of cases are idiopathic and environmental events may act as causative agents. In this

    respect, a provocative role of pesticides (e.g. rotenone and paraquat) as well as heavy metals (e.g.

    iron, manganese) has been suggested [87,88,89]. In particular, epidemiological evidence

    revealed a high incidence of PD in rural areas that use substantial amounts of agrochemicals

    [90]. Consistent with a role for these chemicals in PD, all of the most commonly used pesticides,

    namely, rotenone, paraquat and maneb have been linked to dopaminergic death in rodents

    [88,89]. Indeed, continuous infusion of rotenone, using osmotic minipumps, elicited

    degeneration of SNc dopaminergic neurons and destruction of non-dopaminergic neurons within

    the basal ganglia and brainstem [84,91]. The fact that non-dopaminergic neurons were affected is

    consistent with the modest degeneration observed within these areas in PD patients. Indeed, loss

    of noradrenergic locus coeruleus neurons often occurs in PD and, as alluded to earlier, may

    contribute to some of the depressive symptoms evident in PD [92].

    Although less evidence is available concerning the impact of paraquat in PD, at least one

    study demonstrated that repeated systemic administration of the pesticide provoked selective

    destruction of SNc dopamine neurons [88]. However, striatal dopamine levels were not altered

    by paraquat, suggesting that compensatory mechanisms may have been provoked by the

    surviving neurons [88]. Although the fungicidal agent, maneb, alone did not influence

    dopaminergic neurons, it did augment the neurodegenerative actions of paraquat [89], suggesting

  • 8/7/2019 Hayley curr pharm design

    18/49

    Hayley 18

    that potential synergistic interactions among these agents.

    Environmental stressors in PD: Inflammatory death mechanisms

    Three primary mechanisms may underlie the neurodegenerative actions of MPTP, 6-

    OHDA and the pesticides: (1) inhibition of complex I of the mitochondrial respiratory chain, (2)

    direct oxidative stress factors and (3) provocation of neuroinflammatory cascades. Each of these

    chemicals shares the common property of being inhibitors of complex I of the mitochondrial

    respiratory chain. Rotenone and MPTP are particularly potent complex I inhibitors, promoting

    reduced oxidation of NAD+ substrates and a-ketoglutarate dehydrogenase, culminating in

    decreased ATP levels, loss of mitochondrial membrane potential, faulty intracellular calcium

    buffering and free radical generation [85]. Any one of these mitochondrial-mediated outcomes

    could induce neuronal degeneration. For instance, reduced ATP levels may leave the cell unable

    to meet energy demands and would be especially vulnerable to alternate metabolically

    challenging insults (such as other toxins or stressors).

    Although mitochondrial dysfunction itself can elicit oxidative neuronal damage, 6-

    OHDA and MPTP have been demonstrated to provoke oxidative stress independent of the

    mitochondria. Indeed, like endogenous dopamine, through auto-oxidation or through interactions

    with the catecholaminergic enzyme, monoamine oxidase, 6-OHDA can generate toxic

    metabolites, such as quinones, superoxide radicals, hydrogen peroxide and the hydroxy radical

    which can directly damage neurons [85]. Likewise, rotenone damaged dopaminergic neurons

    through the induction of free radicals and anti-oxidant treatments (e.g. alpha-tocopherol)

    protected these neurons [93]. Systemic MPTP can induce superoxide as well as nitric oxide (NO)

    formation, which together can create the incredibly reactive and destructive peroxynitrite radical

    [94]. As well, MPTP may also impair the functioning of endogenous protective free radical

  • 8/7/2019 Hayley curr pharm design

    19/49

    Hayley 19

    scavengers, such as GSH, metallothionein and manganese superoxide dismutase [94].

    Interestingly, as Czlonkowska and colleagues point out in this issue, inflammatory

    processes associated with microglial activation likely contribute to the oxidative damage

    provoked by MPTP [95,96]. In this respect, it is of interest that each of the environmental agents

    used to induce experimental Parkinsonism also elicit profound immune activation and

    neuroinflammation. This is not surprising given that a primary role of immunologial functioning

    is to rid the body of such environmental antigens. Accordingly, excessive activation of central

    and peripheral immune factors (such as cytokines) engendered by these challenges may

    contribute to tissue damage evident in PD.

    Microglial activation in PD neurotoxin models

    Both in vivo and in vitro procedures have demonstrated that 6-OHDA, MPTP (or its

    metabolite MPP+) and rotenone can induce substantial activation of microglia, the primary CNS

    immunocompetent cell [95,96,97]. Indeed, systemic MPTP treatment promoted profound

    microgliosis that was detected in the SNc of monkeys exposed to the toxin 5 to 14 years earlier,

    suggesting a progressive, long term neuroinflammatory process was associated with relatively

    brief toxin exposure [98]. Mice treated acutely with systemic MPTP (four 10 mg/kg doses

    spaced 1 hr apart) displayed an increased number of microglia and morphological changes

    indicative of activation (e.g. cellular thickening) that was evident for up to 4 and 14 days within

    the striatum and SNc, respectively [96]. It of interest to note that microglial responses occurred

    long before dopaminergic neuronal death was evident (14-21 days), providing evidence for a

    primary role for the inflammatory process. Importantly, advancing age, which is a clear risk

    factor for PD, has also been associated with profound microglia activation, with older mice (9-12

    months) displayed greatly enhanced microglial reactivity following MPTP relative to young

  • 8/7/2019 Hayley curr pharm design

    20/49

    Hayley 20

    animals (3 months) [99].

    In order to determine the mechanistic role of activated microglia in dopaminergic loss,

    several co-culture systems have been established using embryonic ventral mesencephalic

    neurons and postnatally obtained microglia. Using this approach, microglia (but not astrocytes)

    co-cultured with mesencephalic neurons were found to contribute to MPTP provoked neuronal

    injury [97]. Their deleterious effects were linked to NADPH oxidase, the main reactive oxygen

    species-producing enzyme during inflammation [100]. In this regard, neurons obtained from

    mice genetically lacking NADPH or treated with the pharmacological inhibitor, apocynin, were

    largely resistant to MPTP toxicity [101]. Likewise, knockout mice lacking molecular subunits

    (gp91pnox) required for functioning of NADPH oxidase, were resistant to rotenone induced

    dopaminergic loss [101].

    Corresponding to the in vitro data, NADPH oxidase is increased within the SNc of

    human PD patients and MPTP treated rodents [100]. Likewise, NADPH oxidase deficient mice

    displayed substantially less dopaminergic neuron loss in response to systemically delivered

    MPTP compared to wild type animals [100]. Additionally, it has also become apparent that

    systemic MPTP promotes microglial iNOS expression and that mice lacking this oxidative

    enzyme displayed substantially reduced dopaminergic loss [82]. Treatment with minocycline, a

    tetracycline derivative that inhibits microglial activation, prevented MPTP induced nigrostriatal

    degeneration as well as NADPH oxidase, iNOS and nitrotyrosine (marker of NO activity)

    expression [100]. Thus, inflammatory microglia reactivity likely contributes to ongoing

    degeneration through the release of highly reactive oxidative species (see Figure 1).

    It is exceedingly difficult to disentangle the interrelationships among inflammation,

    oxidative stress, apoptosis and excitotoxic death mechanisms. From our perspective, it is

  • 8/7/2019 Hayley curr pharm design

    21/49

    Hayley 21

    important to note that microglia serve as pivotal regulators of each of these diverse molecular

    processes. Thus, it should be underscored that microglia are the most prominent CNS cells

    expressing proinflammatory cytokines, such as IL-1, IL-6, TNF- and IFN-, that have the

    ability to influence death processes (see Figure 1). Indeed, TNF- and its related receptor family

    member, Fas, have well established caspase mediated neuronal apoptotic consequences

    stemming from their receptor linked intracellular death domain complexes [102]. Likewise, IL-

    1 has been reported to promote neuronal apoptosis when found at elevated concentrations; in

    fact, its synthetic enzyme, interleukin converting enzyme, is actually a member of the pro-

    apoptotic caspase family [103]. In terms of excitotoxic death, IL-1 exacerbated the degree of

    neuronal demise promoted by glutamate through NMDA and AMPA receptors [103], whereas

    infusion of the endogenous IL-1 antagonist, IL-1ra, prevented striatal excitotoxicty [104].

    Interestingly, TNF- was recently demonstrated to alter communication between microglia and

    astrocytes to favor development of excitotoxicity [105]. Thus, cytokines may promote neuronal

    death directly or though their impact upon glial cells. In fact, through their potent autocrine

    stimulatory effects, cytokines may also amplify the release of any oxidative or other death

    factors released from the microglia in which they originate.

    Viral and bacterial involvement in PD

    In addition to chemical agents, another environmental culprit has been implicated in PD,

    namely pathogenic microorganisms. Cases of parkinsonian-like syndromes have been associated

    with infections including, poliovirus, arbovirus, herpes simplex virus and encephalitis

    [106,107,108]. A viral hypothesis proposed for PD has suggested the possibility that infection

    prenatally or early in life with some (yet to be discovered) latent virus(es) may instigate the

    disease [106]. The long incubation period and slow evolution of damage provoked by the virus

  • 8/7/2019 Hayley curr pharm design

    22/49

    Hayley 22

    could certainly be envisaged to correspond with the insidious time-course for PD onset. For

    instance, cases of Parkinsonism associated with von Economo encephalitis have been reported to

    occur years after infection [109]. Likewise, postencephalic cases of Parksinsonism that were

    associated with the influenza epidemic of 1918 [108] have been attributed to cytotoxic effects of

    the virus on the developing SNc within the intra-uterine environment [110]. In addition to

    viruses, prenatal or early life exposure to a pathogen of bacterial origin may also play a role in

    PD. In fact, as will be discussed shortly, recent animal studies have demonstrated that rats

    receiving prenatal administration of the bacterial antigen, lipopolysaccharide (LPS), displayed

    substantial degeneration of dopaminergic neurons [8]. It was also noted that rodents exposed to

    low concentrations of pesticides early in life were much more susceptible to the neurotoxic

    consequences of dopaminergic toxins later in life [89]. It may be that early exposure to

    immunogenic events (viral, bacterial or chemical) provokes mild neuroinflammation (e.g.

    microglial activation, cytokine release) that over time may cause neurodegeneration or render

    dopamine neurons vulnerable to degeneration in response to normally low grade insults [8].

    Cases of Parkinsonism have been reported in HIV infected individuals and it was

    suggested that accompanying infections, such as toxoplasmosis, may exacerbate the impact of

    the virus upon basal ganglia functioning [111]. HIV may directly impair dopamine neurons

    through the envelope protein, gp120, or associated HIV protein, Tat, both of which inhibit

    dopamine synthesis and have been found to promote nigrostriatal degeneration in exposed

    rodents [111,112]. Another virus implicated in PD, the Japanese encephalitis virus, reduced the

    number of dopamine neurons and provoked marked gliosis within the SNc of infected rats [113].

    The PD-like behavioral symptoms provoked by the Japanese encephalitis virus, most notably

    bradykinesia, were significantly improved by l-DOPA treatment [113].

  • 8/7/2019 Hayley curr pharm design

    23/49

    Hayley 23

    Neuroimmune mechanisms of PD

    Examination of postmortem PD tissue revealed numerous signs of inflammation,

    including microglial activation and increased levels of several proinflammatory cytokines (IL-1,

    IL-2, IL-6 and TNF-), as well as expression of elements of the complement cascade; an

    important mechanism mediating antibody dependent cytotoxicity [95,114]. In contrast to the role

    of glial mediated inflammatory processes in PD, much less attention has focused upon the impact

    of adaptive immune responses in the disease. In this respect, cytokines may orchestrate

    lymphocyte activity and effector adaptive responses (e.g. antibody mediated complement

    deposition, cell mediated cytotoxicity) that may damage CNS tissue.

    Unlike several other neurological conditions, including MS, stroke and to a lesser degree

    Alzheimers disease, definitive evidence of T lymphocytes within the PD brain is lacking.

    However, reduced levels of T cells within the bloodstream and impaired proliferative responses

    to mitogens in PD patients indicate some degree of altered peripheral lymphocyte immunity [95].

    It has been estimated that 30% of PD patients have autoantibodies reactive against basal ganglia

    neurons [115]. Likewise, examination of cases of Parkinsonism related to encephalitis revealed

    that 95% of patients had autoantibodies reactive against basal ganglia antigens compared to 2%

    of controls [116]. The suggestion has also been made that antibody dependent cell mediated

    cytotoxicity mediated by natural killer (NK) cells may contribute to the pathogenesis of PD

    [157]. This assertion largely stems from the finding that elevations of circulating NK cell activity

    positively correlated with disease severity in PD patients [157].

    One interesting emerging neuroimmune theory of PD suggests that formation of

    neoepitopes (new antigens) within the basal ganglia recruits a specific destructive immune

    reaction [117,118]. Specifically, accumulation of toxic dopamine auto-oxidation metabolites,

  • 8/7/2019 Hayley curr pharm design

    24/49

    Hayley 24

    particularly quinone, may cause tissue alterations favoring neoepitope creation [117]. Consistent

    with this proposition, antibodies from a subset of PD patients (7/21) but not subjects with other

    neurological diseases (0/21), recognized epitopes from dopamine quinone modified proteins

    [118]. Thus, PD patients displayed antibodies against neoepitopes produced by altered dopamine

    metabolism, which could contribute to or amplify ongoing inflammatory and degenerative

    response [118]. Animal studies revealed that direct intra-SNc infusion of purified antibodies

    from PD patients but not age matched disease controls, induced complement activation and

    dopaminergic neuronal death in several rodent species [119]. Importantly, these effects were

    prevented in mice lacking the Fc receptors, which are critical for antibody mediated activation of

    microglial cells [119]. Indeed, through binding and clustering of the Fc membrane antibody

    receptors, specific antibodies can provoke the release of oxidative species, such as superoxide

    radicals, from microglia.

    Neuroinflammatory models of PD: Cytokine involvement

    Further support for a role of immune factors in PD comes from recent studies

    demonstrating that central administration of the bacterial endotoxin, LPS, provoked a loss of

    dopaminergic neurons within the SNc [120]. However, infusion of LPS into the hippocampus,

    thalamus and cortex of rats did not induce substantial neuronal loss [120], suggesting that SNc

    dopaminergic neurons are especially vulnerable to immunogenic insults. It was suggested that

    the particularly high concentration of microglia within the SNc may contribute to the enhanced

    vulnerability of these dopamine neurons [120]. In vitro experiments revealed that the

    neurodegenerative consequences of LPS on mesencephalic neurons were only evident in the

    presence of co-cultures including microglia [97,101]. Since LPS potently induces circulating

    cytokine production and may also stimulate central cytokine expression [121], this may be one

  • 8/7/2019 Hayley curr pharm design

    25/49

    Hayley 25

    mechanism through which the endotoxin causes dopaminergic degeneration. In fact, as will be

    discussed shortly, release of some of the typical proinflammatory cytokines elicited by LPS,

    including IL-1, IL-6, TNF- and IFN-, can synergistically promote a variety of central

    consequences, including cellular death [122,123].

    As alluded to earlier, PD postmortem brain tissue often contains increased expression of

    cytokines, such as IL-1, IL-6, IFN-, TNF-, as well as the TNF- receptor superfamily

    member, Fas [124]. Likewise, cDNA microarray studies indicated that MPTP treated mice

    displayed similar alterations of proinflammatory cytokine genes within basal ganglia brain

    regions [125]. Although 6-OHDA stimulated TNF- and IL-1 expression within the basal

    ganglia [124,126], there is a lack of data on the impact of pesticides, such as rotenone, on

    cytokine levels.

    Although correlative evidence exists, few studies have assessed the mechanistic role of

    cytokines in PD; it is even unclear as to whether these immunotransmitters primarily act in a

    protective or destructive capacity. However, two laboratories have recently reported altered basal

    ganglia responses to MPTP in TNF- deficient knockout mice [127,128]. Although one report

    indicated that TNF- deletion protected striatal terminals and normalized dopamine levels in

    MPTP treated mice [127], the other found increased dopamine metabolism in the absence of any

    evidence of neuroprotection in the MPTP null mice [128]. As depicted in Figure 2, our own

    recent findings found that mice lacking the TNF- receptor superfamily receptor, Fas, displayed

    attenuated dopaminergic neurodegeneration and associated microgliosis [129]. Interestingly, IL-

    6 knockout mice displayed increased SNc dopaminergic soma and striatal terminal degeneration

    following MPTP, suggesting enhanced sensitivity to the toxin in the absence of the cytokine

    [130]. Thus, in keeping with the trophic actions reported for IL-6, endogenous levels of the

  • 8/7/2019 Hayley curr pharm design

    26/49

    Hayley 26

    cytokine may actually protect neurons against insults.

    There are several mechanisms through which cytokines may influence the survival or

    death of dopaminergic neurons. Although this section will evaluate some of these pro-death

    mechanisms, it should be underscored that many cytokines (at least within the immune system)

    act as growth factors promoting cellular differentiation and proliferation. However, it has been

    well established that pro-inflammatory cytokines, such as TNF-, Fas and IFN- as well as

    several chemokines (subcategory of chemoattractant cytokines), can promote cellular death

    through apoptotic, excitotoxic or oxidative processes [131]. For instance, the recently reported

    low levels of the intracellular Fas death domain (FADD) in PD patients prompted the assertion

    that FADD expressing neurons may selectively die through apoptosis in PD [132]. Other

    evidence for classical apoptotic pathways operative in PD includes reports of increased levels of

    caspase-3 and 8 which act as downstream effectors of FADD, Bax and related death pathways

    [133]. However, evidence also exists for inflammatory mechanisms of action for Fas. For

    instance, as shown in Figure 2, we reported that Fas knockout mice displayed diminished

    microglial activation within the SNc and striatum in response to MPTP compared to wild type

    mice [129]. Fas also promoted central expression of the chemokine, IL-8 and has numerous

    inflammatory effects in common with its superfamily member, TNF- [134].

    Neuroimmune-neurotoxin interactions in PD: synergistic effects

    The fact that PD often occurs in distinct clusters suggests that environmental factors (e.g.,

    toxins) related to certain geographic areas may confer vulnerability to disease. Environmental

    distribution of many chemical agents often overlap and it is certainly conceivable that multiple

    toxins may synergistically influence neuronal processes. Likewise, one can surmise that the

  • 8/7/2019 Hayley curr pharm design

    27/49

    Hayley 27

    combination of various toxins (e.g. pesticides) with immunological events (e.g. viral, bacterial

    pathogens) may interact in a similar fashion. Consistent with this proposition, co-administration

    of LPS with rotenone synergistically augmented dopaminergic degeneration in mesencephalon-

    microglia co-cultures, through the release of reactive oxygen species [97,101]. In vivo studies

    indicated that although the pesticide, maneb, had no effect on dopaminergic neurons, its co-

    administration with another pesticide, paraquat, synergistically enhanced the degree of

    nigrostriatal damage and gliosis [89].

    Interestingly, many of the behavioral deficits produced by MPTP are only evident in the

    presence of other chemical agents or stressors [83], suggesting possible synergistic interactions

    among these treatments. For instance, it has been reported that MPTP only induced akinesia and

    cataplepsy deficits when co-administered with the pesticides, diethyldithiocarbamate (DDC) or

    maneb [135, 136]. It was also reported that the imposition of a stressor (transportation stress)

    was necessary to realize the behavioral effects of MPTP [83]. Taken together, these studies

    indicate that consideration of the interactive effects of multiple factors is warranted when

    considering the environmental triggers operative for PD.

    Cytokine-provoked neuronal sensitization: Implications for Depression and PD

    Cytokines may influence the neuronal responses to later challenges although the

    processes leading to such outcomes do not involve recognition in the same way that typical

    immune responses to antigens elicit such an effect. Sensitization from a neuronal perspective is a

    fundamental process relevant to stressor related pathologies, and to neural plasticity in general.

    Just as stressor experiences may increase the likelihood of a depressive episode developing given

    a subsequent stressor event, cytokines, even when given at low concentrations that do not

  • 8/7/2019 Hayley curr pharm design

    28/49

    Hayley 28

    produce noticeable behavioural effects, may elicit central neuronal sensitization upon their

    reexposure. From this perspective, stressors and cytokines have protracted effects on

    psychological and neurological processes well after the initial actions of the treatment have

    waned.

    Cytokine induced HPA and behavioural sensitization: Peripheral mechanisms

    Just as certain psychosocial and neurogenic stressors may provoke exaggerated

    neurochemical responses upon subsequent stressor exposure [137,138,139,140,141], IL-1 and

    TNF sensitized activity of the HPA axis so that later cytokine treatment provoked particularly

    marked responses [1,142]. These cytokines induced a phenotypic change in the co-localization

    of the hypothalamic neuropeptides, AVP and CRH, such that increased co-expression of AVP

    occurred within CRH terminals in the external zone of the median eminence (ZEME). In keeping

    with the synergistic impact of these neuropeptides on pituitary ACTH release, their increased co-

    expression engendered by IL-1 was associated with enhanced ACTH and corticosterone

    secretion [141,142].

    Interestingly, the corticosterone and CRH/AVP sensitization effects of TNF- followed

    different time courses. Although increased CRH and AVP co-storage within the ZEME peaked

    7-14 days following the initial TNF- treatment, the sensitized corticosterone response was only

    evident in mice re-exposed to the cytokine 28 days after pre-treatment with the cytokine [1].

    Thus, mechanisms other than hypothalamic neuropeptide secretion may underlie the corticoid

    sensitization. Indeed, we recently found evidence that TNF- may be acting as an adjuvant with

    the well known immunogenic factor, bovine serum albumin (BSA), which was used as a carrier

    protein in the injection mixture [143]. As well, systemic pre-treatment with an antihistamine

  • 8/7/2019 Hayley curr pharm design

    29/49

    Hayley 29

    cocktail (diphenhydramine + cimetidine, H1 and H2 antagonists, respectively) abrogated the

    corticosterone sensitization, suggesting a histamine dependent mechanism was involved [31].

    Paralleling the corticosterone sensitization, TNF- reexposure 28 days following initial

    cytokine elicited profound sickness symptoms reminiscent of those associated with

    systemic/anaphylactic shock [31]. Indeed, mice displayed pronounced reddening of the tail, ears

    and nose (cyanosis; indicating increased accumulation of blood cells and inflammation), coupled

    with a marked reduction of blood volume and pressure, as well as signs of hypothermia [31].

    Although co-administration of the H1 and H2 antagonists, diphenhydramine and cimetidine, did

    not appreciably influence the acute effects of TNF-, the sensitization of sickness associated

    with reexposure to the cytokine was prevented [31]. Together, these findings raise the possibility

    that events that induce TNF- or other cytokines (e.g. infections, stressors) may render

    organisms vulnerable to subsequent exposure to the same or sufficiently similar insult. In the

    case of TNF-, such reexposure may induce an acute phase reaction that could progress to full-

    blown shock. Alternatively, exposure to relatively low endogenous levels of TNF- (as would be

    expected in humans) over time may elicit modest acute phase reactions that could not only

    influence visceral processes, but may also promote exaggerated corticoid release, inducing

    detrimental effects as chronically high levels of the hormone may influence the development of

    diabetes, heart disease, stroke and memory impairments.

    Cytokine induced neurochemical sensitization: Central mechanisms

    Although the corticosterone and sickness sensitization elicited by TNF- appears to be

    related to peripheral immune processes, the cytokine also evoked sensitized brain neurochemical

    responses independent of peripheral factors. Indeed, it will be recalled that the augmented

  • 8/7/2019 Hayley curr pharm design

    30/49

    Hayley 30

    CRH/AVP co-localization within the ZEME occurred independent of sickness and corticoid

    variations. Likewise, reexposure to TNF- 1 day following pre-treatment with the cytokine

    induced a sensitization of CRH expression within the central nucleus of the amygdala, at a time

    when no signs of illness or corticoid activation were apparent [144]. These neuropeptide

    variations presumably involved some sensitized central mechanism that was closely linked to the

    timing of reexposure. Importantly, activation of CRH within the central amygdala contributes to

    anxiety and responses to fear-related stimuli and it is thus possible that this mechanism

    contributes to anxiety associated with endotoxin challenges (which induce TNF-) in humans

    [145]. In fact, central infusion of TNF- was reported to induce anxiogenic effects in rodents

    testing using an elevated plus maze [146].

    Systemic TNF- markedly influenced monoamine activity within several brain regions [1,

    32, 35], and re-exposure to the cytokine increased activity and/or levels of NE, DA and 5-HT in

    a region-specific and time-dependent fashion [1, 3, 20]. As was the case for CRH/AVP, these

    time-dependent brain neurochemical alterations occurred earlier than the sickness or HPA

    changes [1]. Sensitized utilization of NE and 5-HT that was elicited by systemic TNF- were

    reliably detected within the amygdala and prefrontal cortex [1], the former of which is important

    for emotional responding and the latter involved in cognitive appraisal. Interestingly, as depicted

    in Figure 3, when TNF- was central infused into the lateral ventricles, monoamine sensitization

    effects were most apparent within the hypothalamus [147]. Such an effect may stem from region

    specific difference in cytokine diffusion and uptake.

    Cytokine sensitization and the development of depression.

  • 8/7/2019 Hayley curr pharm design

    31/49

    Hayley 31

    It will be recalled that cytokines and their soluble receptors are increased in depressive

    conditions and antidepressant treatments often normalize these variations, at least for TNF-

    [148]. As well, cytokine immunotherapy with IFN- or IL-2 induced depressive conditions that

    were amendable to antidepressant treatments [68]. Interestingly, the assertion was made that

    individuals vulnerable to developing depression have sensitized HPA responses when challenged

    with immune agents [149]. In this regard, IFN- immunotherapy initially provoked a greatly

    augmented ACTH responses in melanoma patients that subsequently developed depressive

    pathology, relative to those that did not develop such symptoms [149]. Others have demonstrated

    that daily IFN- treatment for three weeks reduced HPA responses to the cytokine but a greatly

    augmented response to a sub-threshold challenge with CRH [150]. Thus, although repeated IFN-

    administration may induce a tolerance to its own neuroendocrine actions may sensitize HPA

    functioning to alternate challenges.

    It is important to underscore that clinical cytokine administration involves quite high

    concentrations of these immunotransmitters, certainly within the pathophysiological range [151],

    unlike the relatively low levels detected in depressive individuals not undergoing such

    treatments. We propose the possibility that modestly elevated cytokine levels that persist over

    long time intervals may come to sensitize neural functioning in stressor sensitive brain regions.

    Along these lines, it is also important to consider that the schedule or pattern of cytokine

    exposure likely plays a critical role in its neurochemical consequences. Indeed,

    immunotherepeutic cancer treatment schedules often involve repeated administration of the

    cytokine for many months, often with delays of several days between injections. The chronicity

    of such therapy would give ample time for the development of time dependent sensitization

    effects. The intermittent nature of treatment may also favour the development of central

  • 8/7/2019 Hayley curr pharm design

    32/49

    Hayley 32

    sensitized effects, since we have found that TNF- sensitized CRH and monoamine activity

    within brain regions controlling emotional, cognitive and endocrine responses when the cytokine

    injections were spaced 1-7 days apart [144,147].

    Like TNF-, administration of a bacterial endotoxin, such as LPS, may proactively influence

    the HPA and central monoamine responses to subsequent challenges of a different sort (i.e.,

    cross-sensitization developed). Unlike the effects of TNF-, however, the proactive effects of

    LPS were relatively transient, being evident 1-day after the initial treatment but not at 28 days

    [2]. Interestingly, the developmental timing of the immunological insult may be of relevance,

    since rats exposed to endotoxin perinatally (1 and 3 days post partum) displayed sensitized CNS

    activity in response to subsequent endotoxin or stressor exposure during adulthood [152]. These

    data raise the possibility that early life infectious events may also impact upon psychological

    responses to stressors encountered in adulthood [152].

    One possibility that has been considered is that early insults may be affecting the plasticity of

    neuronal processes. For example, as indicated earlier, recent theories of depression have

    entertained the view that aberrant plasticity of certain neuronal pathways may be involved in the

    etiology of depression and that the trophic cytokine, brain derived neurotrophic factor (BDNF)

    may be important in this respect. Indeed, stressful events have been shown to reduce BDNF

    expression, whereas chronic antidepressant treatments increased brain levels of BDNF and

    attenuated the reduced BDNF ordinarily provoked by stressors [61,62,63]. While it is certainly

    possible that the impact of stressors (and antidepressants) on BDNF and other cytokines, such as

    IL-1 and TNF-, have additive or interactive effects with respect to depressive states, at present

    data are unavailable concerning such potential interactions. Likewise, it is unclear whether cross-

    sensitization occurs between these varied growth factors and stressors.

  • 8/7/2019 Hayley curr pharm design

    33/49

    Hayley 33

    Cytokine sensitization and the development of PD

    Just as early life immune challenges may confer increased vulnerability to the

    development stress-related anxiety, there is reason to believe that this treatment influences

    vulnerability to other pathological conditions, such as PD. In this regard, prenatal exposure to

    LPS at embryonic days 10-11 resulted in a substantial reduction of the number of SNc

    dopaminergic neurons evident in adult rats [8]. In addition, prenatal LPS exposure engendered a

    long-term increase of striatal TNF- that was even evident in mice 120 days of age [153]. As

    the overall number of neurons within the SNc, as revealed by Map-2 immunoreactivity, was not

    reduced by LPS, it appears that at the dose used the endotoxin selectively impacted dopamine

    neurons. It was suggested that dopamine containing neurons may be particularly vulnerable to

    the effects of early TNF- exposure, possibly through its inhibitory effects on important growth

    factors, such as nurr-1 or sonic hedgehog [8].

    Early exposure to proinflammatory environmental toxins may also act to sensitize

    neurons to the deleterious actions of subsequent nigrostriatal insults. For instance, combined

    treatment with the pesticides, paraquat + maneb, from postnatal days 5-19 sensitized rodents to

    the damaging effects of these challenges months later [89]. Indeed, reexposure to the

    combination of these pesticides in adulthood provoked a significantly greater SNc neuronal loss

    and striatal dopamine depletion coupled with pronounced motor impairment relative to animals

    not exposed to the toxins early in life [89]. Although not measured in this study, it will be

    recalled that these pesticides readily instigate neuroinflammatory activation, particularly as

    indicated by microgliosis.

    The time course for neurodegeneration following inflammatory challenges is consistent

    with the slow progressive nature of PD. For instance, continuous intra-SNc infusion of a low

  • 8/7/2019 Hayley curr pharm design

    34/49

    Hayley 34

    dose of LPS for several weeks (closely mimicking a typical neuroinflammatory state) elicited a

    maximal microglia response after 2 weeks but SNc degeneration was not evident until 4-6 weeks

    later [97]. Likewise, exposure to MPTP and closed head injury have been associated with

    protracted elevations (often for years) of microglia and cytokines [154]. The latter may explain

    cases of PD linked to repeated blows to the head, as observed in boxers. In any case, it is

    conceivable that such ongoing neuroinflammation would sensitize individuals to the

    degenerative consequences of subsequent environmental insults. We believe that microglia

    activation can lead to cytokine release that contributes to pathology through two primary

    mechanisms: (1) the promotion of glial derived reactive oxygen species and (2) activation of

    intracellular neuronal apoptotic or excitotoxic death processes, possibly through stimulation of

    MAP kinase pathways. Alternatively, the possibility should not be dismissed that low-grade

    dopaminergic injury may recruit peripheral immune responses (e.g. antibody dependent

    cytotoxicity) that may further amplify any ongoing degeneration.

    In conclusion, as summarized in Figure 1, insults that affect immune and inflammatory

    system functioning may have profound effects on CNS mechanisms implicated in the regulation

    of neurochemical, behavioral and neurodegenerative processes. The fact that cytokines act as

    common messengers between and within the CNS and immune system, coupled with the

    possible involvement of these systems in both psychiatric and neurodegenerative conditions,

    raises the possibility that manipulation of cytokine responses may have beneficial effects for

    these clinical conditions. Indeed, anti-inflammatory clinical trials (e.g. using NSAIDs and

    cytokine antagonists) are currently being explored for the treatment of MS and Alzheimers

    disease [155]. Since cytokine immunotherapy (e.g. IFN- for melanoma) can induce depressive

    symptomatology that is ameliorated by antidepressants, the possibility of using cytokine

  • 8/7/2019 Hayley curr pharm design

    35/49

    Hayley 35

    antagonists in certain cases of depression (particularly those associated with medical conditions)

    should also be considered. In any case, the temporal pattern of exposure to stressors and immune

    challenges over the life span sets the tone for whether or not such individuals develop a

    sensitized state for CNS pathology.

  • 8/7/2019 Hayley curr pharm design

    36/49

    Hayley 36

    Acknowledgements:

    Supported by the Canadian Institutes of Health Research and the Natural Science and Engineering

    Research Council of Canada. S.H. and H.A. hold Canada Research Chairs in Neuroscience.

    References

    1. Hayley S, Brebner K, Lacosta S., Merali Z & Anisman H. Sensitization effects of TumorNecrosis Factor-a: neuroendocrine, central monoamine and behavioral variations. J Neurosci1999; 19 (13): 5654-5665.

    2. Hayley S, Lacosta S, Merali Z, van Rooijen N & Anisman H. Central Monoamine and PlasmaCorticosterone Changes Induced by a Bacterial Endotoxin: Sensitization and Cross-sensitizationEffects. Eur J Neurosci 2001; 13: 1155-1165.

    3. Hayley S, Merali Z & AnismanH. The acute and sensitization effects of tumor necrosis factor-: implications for immunotherapy as well as psychiatric and neurological conditions. ActaNeuropsychiat 2002; 14: 322-335.

    4. Mogi M, Harada M, Narabayashi H, Inagaki H, Minami M, Nagatsu T.Interleukin (IL)-1 beta, IL-2, IL-4, IL-6 and transforming growth factor-alpha levels are elevated

    in ventricular cerebrospinal fluid in juvenile parkinsonism and Parkinson's disease. Neurosci Lett

    1996; 211:1 13-6.

    5. Mogi M, Harada M, Riederer P, Narabayashi H, Fujita K, Nagatsu T. Tumor necrosis factor-

    alpha (TNF-alpha) increases both in the brain and in the cerebrospinal fluid from parkinsonianpatients. Neurosci Lett 1994;165(1-2):208-10.

    6. Rothwell NJ. Cytokines killers in the brain? J Physiol 1999;514: 3-17.

    7. Griffin WS, Sheng JG, Roberts GW, Mrak RE. Interleukin-1 expression in different plaque

    types in Alzheimer's disease: significance in plaque evolution. J Neuropathol Exp Neurol 1995;

    54(2):276-81.

    8. Carvey PM, Chang Q, Lipton JW, Ling Z. Prenatal exposure to the bacteriotoxin

    lipopolysaccharide leads to long-term losses of dopamine neurons in offspring: a potential, new

    model of Parkinson's disease. Front Biosci 2003; 8:s826-37.

    9. Ringheim GE, Conant K. Neurodegenerative disease and the neuroimmune axis (Alzheimer's

    and Parkinson's disease, and viral infections). J Neuroimmunol 2004; 147:43-9.

    10. Anisman H, Hayley S, Turrin N & Merali Z (2002). Stress, cytokines and depression. Inter. J.

    Neuropsychiat. 5, 357-373.

  • 8/7/2019 Hayley curr pharm design

    37/49

    Hayley 37

    11. Nemeroff CB. The role of corticotrophin-releasing factor in the pathogenesis of major

    depression. Pharmacopsychiatry 1988; 21: 76-82.

    12. Bluthe RM, Dantzer R, Kelley KW. Central mediation of the effects of interleukin-1 on

    social exploration and body weight in mice. Psychoneuroendocrinology. 1997; 22(1):1-11.

    13. Dunn A.J. Interactions between the nervous system and the immune system. 1995 In:

    Psychopharmacology: The Fourth Generation of Progress (Bloom, F.E., Kupfer, D.J., eds).Raven Press, New York. pp 719-731

    14. Ericsson A, Kovacs KJ, Sawchenko PE. A functional anatomical analysis of centralpathways subserving the effects of interleukin-1 on stress-related neuroendocrine neurons. J

    Neurosci. 1994; 14(2):897-913.

    15. Shintani, F., Nakaki, T., Kanba ,S., Sato, K., Yagi, G., Shiozawa, M., Aiso, S., Kato, R. and Asai,

    M. Involvement of interleukin-1 in immibilization stress-induced increase in plasma

    adrenocorticotropic hormone and in release of hypothalamic monoamines in the rat. J Neurosci 1995;15: 1961-1970.

    16. Maier SF, Watkins LR. Cytokines for psychologists: Implications of bidirectional immune-

    to-brain communication for understanding behavior, mood, and cognition. Psychol Rev 1998;105: 83-107.

    17. Nguyen KT, Deak T, Owens SM, Kohno T, Fleshner M, Watkins LR, Maier SF. Exposure to

    acute stress induces brain interleukin-1beta protein in the rat. J Neurosci 1998; 18(6):2239-46.

    18. Pugh CR, Nguyen KT, Gonyea JL, Fleshner M, Watkins LR, Maier SF, Rudy JW. Role of

    interleukin-1 beta in impairment of contextual fear conditioning caused by social isolation.Behav Brain Res 1999; 106: 109118.

    19. Anisman H, Merali Z. Anhedonic and anxiogenic effects of cytokine exposure. Adv ExpMed Biol, 1999; 461:199-233.

    20. Hayley S, Merali Z, Anisman H. Stress and cytokine-elicited neuroendocrine andneurotransmitter sensitization: implications for depressive illness. Stress 2003;6(1):19-32.

    21. Merali Z, Lacosta S, Anisman H. Effects of interleukin-1beta and mild stress on alterationsof norepinephrine, dopamine and serotonin neurotransmission: a regional microdialysis study.

    Brain Res. 1997; 761(2):225-35.

    22. Smith RS. The macrophage theory of depression. Med Hypotheses 1991; 35(4):298-306.

    23. Hart BL. Biological basis of the behavior of sick animals. Neurosci Biobehav Rev 1988;12(2):123-37.

  • 8/7/2019 Hayley curr pharm design

    38/49

  • 8/7/2019 Hayley curr pharm design

    39/49

    Hayley 39

    36. Pauli S, Linthorst AC, Reul JM. Tumour necrosis factor-alpha and interleukin-2 differentiallyaffect hippocampal serotonergic neurotransmission, behavioural activity, body temperature and

    hypothalamic-pituitary-adrenocortical axis activity in the rat. Eur J Neurosci. 1998; 10(3):868-

    878.

    37. Kronfol Z. Immune dysregulation in major depression: a critical review of existing evidence.

    Int J Neuropsychopharmacol. 2002;5(4):333-43.

    38. Maes M, Scharpe S, Bosmans E, Vandewoude M, Suy E, Uyttenbroeck W, Cooreman W,

    Vandervorst C, Raus J. Disturbances in acute phase plasma proteins during melancholia:additional evidence for the presence of an inflammatory process during that illness. Prog

    Neuropsychopharmacol Biol Psychiatry. 1992;16(4):501-15.

    39. Anisman, H. Stress, Immunity, Cytokines, and Depression. Acta Neuropsychiatrica 2002; 14:251-262.

    40.Anisman H, Ravindran AV, Griffiths J, Merali Z. Endocrine and cytokine correlates of majordepression and dysthymia with typical or atypical features. Mol Psychiatry. 1999; 4(2):182-188.

    41. Nunes SO, Reiche EM, Morimoto HK, Matsuo T, Itano EN, Xavier EC, Yamashita CM,Vieira VR, Menoli AV, Silva SS, Costa FB, Reiche FV, Silva FL, Kaminami MS. Immune andhormonal activity in adults suffering from depression.Braz J Med Biol Res 2002;35(5):581-587.

    42. Yirmiya R, Pollak Y, Morag M, Reichenberg A, Barak O, Avitsur R, Shavit Y, Ovadia H,Weidenfeld J, Morag A, Newman ME, Pollmacher T. Illness, cytokines, and depression. Ann NY Acad Sci 2000; 917:478-487.

    43. Capuron L, Ravaud A, Dantzer R. Timing and specificity of the cognitive changes inducedby interleukin-2 and interferon-alpha treatments in cancer patients. Psychosom Med 2001;63(3):376-86.

    44. Mossner R, Heils A, Stober G, Okladnova O, Daniel S, Lesch KP. Enhancement of serotonintransporter function by tumor necrosis factor alpha but not by interleukin-6. Neurochem Int

    1998; 33(3):251-4.

    45. Ramamoorthy S, Blakely RD. Phosphorylation and sequestration of serotonin transporters

    differentially modulated by psychostimulants. Science 1999; 285(5428):763-6.

    46. Wichers MC, Maes M. The role of indoleamine 2,3-dioxygenase (IDO) in thepathophysiology of interferon-alpha-induced depression. J Psychiatry Neurosci 2004; 29(1):11-7.

    47. Sheline YI, Gado MH, Kraemer HC. Untreated depression and hippocampal volume loss.Am J Psychiatry 2003; 160(8):1516-8

    48. Bremner JD, Narayan M, Anderson ER, Staib LH, Miller HL, Charney DS. Hippocampalvolume reduction in major depression. Am J Psychiatr 2000; 157(1):115-8.

  • 8/7/2019 Hayley curr pharm design

    40/49

    Hayley 40

    49. Kempermann G, Kronenberg G. Depressed new neurons--adult hippocampal neurogenesisand a cellular plasticity hypothesis of major depression. Biol Psychiatry 2003; 54(5):499-503.

    50. Coyle JT, Duman RS. Finding the intracellular signaling pathways affected by mood disordertreatments. Neuron 2003; 38(2):157-60.

    51. Jacobs BL. Adult brain neurogenesis and depression. Brain Behav Immun 2002;16(5):602-9.

    52. Manji HK, Quiroz JA, Sporn J, Payne JL, Denicoff K, A Gray N, Zarate CA Jr, Charney DS.Enhancing neuronal plasticity and cellular resilience to develop novel, improved therapeutics fordifficult-to-treat depression. Biol Psychia 2003; 53(8):707-42.

    53. Hellsten J, Wennstrom M, Mohapel P, Ekdahl CT, Bengzon J, Tingstrom A.

    Electroconvulsive seizures increase hippocampal neurogenesis after chronic corticosterone

    treatment. Eur J Neurosci 2002; 16(2):283-90.

    54. Malberg JE, Duman RS. Cell proliferation in adult hippocampus is decreased by inescapablestress: reversal by fluoxetine treatment. Neuropsychopharmacol 2003; 28(9):1562-71.

    55. Alonso R, Griebel G, Pavone G, Stemmelin J, Le Fur G, Soubrie P.

    Blockade of CRF(1) or V(1b) receptors reverses stress-induced suppression of neurogenesis in a

    mouse model of depression. Mol Psychiatry. 2003; Dec 23 [Epub ahead of print].

    56. McEwen BS. Plasticity of the hippocampus: adaptation to chronic stress and allostatic load.

    Ann N Y Acad Sci 2001; 933:265-77.

    57. Sapolsky RM. The possibility of neurotoxicity in the hippocampus in major depression: a

    primer on neuron death. Biol Psychiatry 2000; 48(8):755-65.

    58. Lucassen PJ, Muller MB, Holsboer F, Bauer J, Holtrop A, Wouda J, Hoogendijk WJ, De

    Kloet ER, Swaab DF. Hippocampal apoptosis in major depression is a minor event and absent

    from subareas at risk for glucocorticoid overexposure. Am J Pathol 2003; 158(2):453-68.

    59. Jarskog LF, Selinger ES, Lieberman JA, Gilmore JH. Apoptotic proteins in the temporal

    cortex in schizophrenia: high Bax/Bcl-2 ratio without caspase-3 activation. Am J Psychiatry2004;161(1):109-15.

    60. Manji HK, Chen G. PKC, MAP kinases and the bcl-2 family of proteins as long-term targets

    for mood stabilizers. Mol Psychia 2002; 7 Suppl 1:S46-56.

    61. Shimizu E, Hashimoto K, Okamura N, Koike K, Komatsu N, Kumakiri C, Nakazato M,

    Watanabe H, Shinoda N, Okada S, Iyo M. Alterations of serum levels of brain-derivedneurotrophic factor (BDNF) in depressed patients with or without antidepressants. Biol

    Psychiatry 2003; 54(1):70-5.

  • 8/7/2019 Hayley curr pharm design

    41/49

    Hayley 41

    62. Duman RS, Malberg J, Thome J. Neural plasticity to stress and antidepressant treatment. Biol

    Psychiatry 1999; 46(9):1181-91.

    63. Shirayama Y, Chen AC, Nakagawa S, Russell DS, Duman RS. Brain-derived neurotrophic

    factor produces antidepressant effects in behavioral models of depression. J Neurosci 2002;

    22(8):3251-61.

    64. Curtis J, Finkbeiner S. Sending signals from the synapse to the nucleus: possible roles forCaMK, Ras/ERK, and SAPK pathways in the regulation of synaptic plasticity and neuronal

    growth. Neurosci Res 1999; 58(1):88-95.

    65. Eom DS, Choi WS, Oh YJ.. Bcl-2 enhances neurite extension via activation of c-Jun N-

    terminal kinase. Biochem Biophys Res Commun 2004; 314(2):377-81.

    66. Ying SW, Futter M, Rosenblum K, Webber MJ, Hunt SP, Bliss TV, Bramham CR.Brain-derived neurotrophic factor induces long-term potentiation in intact adult hippocampus:

    requirement for ERK activation coupled to CREB and upregulation of Arc synthesis. J Neurosci2002; 22(5):1532-40.

    67. Manji HK, Duman RS. Impairments of neuroplasticity and cellular resilience in severe mood

    disorders: implications for the development of novel therapeutics. Psychopharmacol Bull 2003;35(2):5-49.

    68. Capuron L, Neurauter G, Musselman DL, Lawson DH, Nemeroff CB, Fuchs D, Miller AH.

    Interferon-alpha-induced changes in tryptophan metabolism. relationship to depression andparoxetine treatment. Biol Psychiatry 2003; 54(9):906-14.

    69. Hanisch UK. Microglia as a source and target of cytokines. Glia 2002; 40(2):140-55.

    70. Pocock JM, Liddle AC. Microglial signalling cascades in neurodegenerative disease. Prog

    Brain Res 2001; 132:555-65.

    71. Cummings JL, Masterman DL. Depression in patients with Parkinson's disease. Int J Geriatr

    Psychiatry 1999; 14(9):711-8.

    72. Leentjens AF, Van den Akker M, Metsemakers JF, Lousberg R, Verhey FR.. Higher

    incidence of depression preceding the onset of Parkinson's disease: a register study. Mov Disord

    2003; 18(4):414-8.

    73. Barone FC, White RF, Spera PA, Ellison J, Currie RW, Wang X, Feuerstein GZ. Ischemic

    preconditioning and brain tolerance: temporal histological and functional outcomes, proteinsynthesis requirement, and interleukin-1 receptor antagonist and early gene expression. Stroke

    1998; 29(9):1937-50.

  • 8/7/2019 Hayley curr pharm design

    42/49

    Hayley 42

    74. Sheng JG, Griffin WS, Royston MC, Mrak RE. Distribution of interleukin-1-immunoreactive

    microglia in cerebral cortical layers: implications for neuritic plaque formation in Alzheimer'sdisease. Neuropathol Appl Neurobiol 1998; 24(4):278-83.

    75. Woodroofe MN. Cytokine production in the central nervous system. Neurology1999; 45(6

    Suppl 6):S6-10.

    76. Bruce AJ, Boling W, Kindy MS, Peschon J, Kraemer PJ, Carpenter MK, Holtsberg FW,Mattson MP. Altered neuronal and microglial responses to excitotoxic and ischemic brain injury

    in mice lacking TNF receptors. Nat Med 1996; 2(7):788-94.

    77. Hirsch E, Graybiel AM, Agid YA. Melanized dopaminergic neurons are differentially

    susceptible to degeneration in Parkinson's disease. Nature 1998; 334(6180):345-48.

    78. Bonnet AM, Houeto JL. Pathophysiology of Parkinson's disease. Biomed Pharmacother1999; 53(3):117-21.

    79. Langston JW, Ballard P, Tetrud JW, Irwin I. Chronic Parkinsonism in humans due to aproduct of meperidine-analog synthesis. Science 1983; 25;219(4587):979-80.

    80. Blandini F, Nappi G, Tassorelli C, Martignoni E. Functional changes of the basal gangliacircuitry in Parkinson's disease. Prog Neurobiol. 2000; 62(1):63-88.

    81. Salach JI, Singer TP, Castagnoli N Jr, Trevor A. Oxidation of the neurotoxic amine 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) by monoamine oxidases A and B and

    suicide inactivation of the enzymes by MPTP. Biochem Biophys Res Commun 1984;

    125(2):831-5.

    82. Przedborski S, Jackson-Lewis V, Yokoyama R, Shibata T, Dawson VL, Dawson TM.

    Role of neuronal nitric oxide in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced

    dopaminergic neurotoxicity. Proc Natl Acad Sci 1996; 14;93(10):4565-71.

    83. Sedelis M, Schwarting RK, Huston JP. Behavioral phenotyping of the MPTP mouse model

    of Parkinson's disease. Behav Brain Res 2001; 125(1-2):109-125.

    84. Hirsch EC, Hoglinger G, Rousselet E, Breidert T, Parain K, Feger J, Ruberg M, Prigent A,

    Cohen-Salmon C, Launay JM. Animal models of Parkinson's disease in rodents induced by

    toxins: an update. J Neural Transm Suppl. 2003; 65:89-100.

    85. Blum D, Torch S, Lambeng N, Nissou M, Benabid AL, Sadoul R, Verna JM. Molecular

    pathways involved in the neurotoxicity of 6-OHDA, dopamine and MPTP: contribution to theapoptotic theory in Parkinson's disease. Prog Neurobiol. 2001; 65(2):135-72.

    86. Lansbury PT, Brice A. Genetics of Parkinson's disease and biochemical studies of implicatedgene products. Curr Opin Genet Dev 2002; 12(3):299-306.

  • 8/7/2019 Hayley curr pharm design

    43/49

    Hayley 43

    87. Jenner P. Parkinson's disease, pesticides and mitochondrial dysfunction. Trends Neurosci

    2001; 24(5):245-7.

    88. McCormack AL, Thiruchelvam M, Manning-Bog AB, Thiffault C, Langston JW, Cory-

    Slechta DA, Di Monte DA. Environmental risk factors and Parkinson's disease: selective

    degeneration of nigral dopaminergic neurons caused by the herbicide paraquat. Neurobiol Dis2002; 10(2):119-127.

    89. Thiruchelvam M, Richfield EK, Baggs RB, Tank AW, Cory-Slechta DA.The nigrostriatal dopaminergic system as a preferential target of repeated exposures to combined

    paraquat and maneb: implications for Parkinson's disease. J Neurosci 2000; 20, 9207-14.

    90. Di Monte DA. The environment and Parkinson's disease: is the nigrostriatal system

    preferentially targeted by neurotoxins? Lancet Neurol. 2003; 2(9):531-8.

    91. Betarbet R, Sherer TB, MacKenzie G, Garcia-Osuna M, Panov AV, Greenamyre JT. Chronicsystemic pesticide exposure reproduces features of Parkinson's disease. Nat Neurosci. 2000;

    3(12):1301-6.

    92. Chan-Palay V. Depression and dementia in Parkinson's disease. Catecholamine changes in

    the locus ceruleus, a basis for therapy. Adv Neurol. 1993; 60:438-46.

    93. Sherer TB, Betarbe