• No results found

University of Groningen Lipocalin 2 and the pathophysiology of Alzheimer's disease Dekens, Doortje W.

N/A
N/A
Protected

Academic year: 2021

Share "University of Groningen Lipocalin 2 and the pathophysiology of Alzheimer's disease Dekens, Doortje W."

Copied!
49
0
0

Bezig met laden.... (Bekijk nu de volledige tekst)

Hele tekst

(1)

Lipocalin 2 and the pathophysiology of Alzheimer's disease Dekens, Doortje W.

IMPORTANT NOTE: You are advised to consult the publisher's version (publisher's PDF) if you wish to cite from it. Please check the document version below.

Document Version

Publisher's PDF, also known as Version of record

Publication date: 2019

Link to publication in University of Groningen/UMCG research database

Citation for published version (APA):

Dekens, D. W. (2019). Lipocalin 2 and the pathophysiology of Alzheimer's disease. Rijksuniversiteit Groningen.

Copyright

Other than for strictly personal use, it is not permitted to download or to forward/distribute the text or part of it without the consent of the author(s) and/or copyright holder(s), unless the work is under an open content license (like Creative Commons).

Take-down policy

If you believe that this document breaches copyright please contact us providing details, and we will remove access to the work immediately and investigate your claim.

Downloaded from the University of Groningen/UMCG research database (Pure): http://www.rug.nl/research/portal. For technical reasons the number of authors shown on this cover page is limited to 10 maximum.

(2)

Chapter 6

Lipocalin 2 as a link between aging, risk factor conditions and

age-related brain diseases

D.W. Dekensa,b, U.L.M. Eiselb,c, L. Gouweleeuwb, R.G. Schoemakerb, P.P. De Deyna,d*, P.J.W. Naudéa,b*

a

Department of Neurology and Alzheimer Center, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands.

bDepartment of Molecular Neurobiology, Groningen Institute for Evolutionary Life Sciences (GELIFES), University of Groningen, Groningen, the Netherlands.

c

University Center of Psychiatry & Interdisciplinary Center of Psychopathology of Emotion Regulation, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands.

d

Laboratory of Neurochemistry and Behaviour, Biobank, Institute Born-Bunge, University of Antwerp, Antwerp, Belgium.

*Shared last author.

(3)

98

Abstract

Chronic (neuro)inflammation is known to play an important role in most if not all age-related diseases of the central nervous system (CNS), such as Alzheimer’s disease (AD), Parkinson’s disease (PD) and vascular dementia (VaD). Inflammation also characterizes many conditions that form a risk factor for these CNS disorders, such as physical inactivity, obesity and cardiovascular disease. Lipocalin 2 (Lcn2) is an inflammatory protein that may be involved in different age-related CNS diseases, as well as different risk factor conditions thereof. Lcn2 levels are increased in the brain in different age-related CNS diseases, including AD and PD. Experimental studies indicate that Lcn2 may contribute to neuropathological processes by affecting for example neuroinflammation, cell survival/cell death and iron metabolism. Interestingly, also in different risk factor conditions, Lcn2 levels are increased in the periphery and the brain. We hypothesize that rising Lcn2 levels during aging and risk factor conditions may sensitize the brain, and increase the risk to develop age-related CNS diseases. In this review, we firstly summarize the current evidence for a role of Lcn2 in different CNS disorders and neuropathological processes. Secondly, we discuss the possibility that Lcn2 may form an inflammatory link between different risk factor conditions and the development of age-related CNS disorders, including AD, PD and VaD.

(4)

99

6

1. Introduction

With aging comes an increased risk to develop age-related brain diseases, such as Alzheimer’s disease (AD), Parkinson’s disease (PD) and vascular dementia (VaD). Although it is evident that aging is the major risk factor for these central nervous system (CNS) conditions, the causation of these conditions remains poorly understood. However, it is becoming increasingly clear that chronic neuroinflammatory processes may play an important rolein the pathogenesis of age-related CNS diseases [1–4].

Neuroinflammation is mediated by microglia - the primary immune cells of the brain - and astrocytes, which are key players in brain inflammation as well [3]. Activated microglia and astrocytes exert multiple protective functions, including the clearance of pathogens, cellular debris and harmful protein aggregates [3,5]. However, when their activation becomes chronic, microglia and astrocytes may lose some of their physiological functions. Instead, chronically activated microglia and astrocytes can provoke brain damage by persistently secreting pro-inflammatory cytokines, thereby sensitizing neurons to cell death and promoting formation of toxic protein aggregates [3,6–11]. Chronic neuroinflammation characterizes most (if not all) age-related CNS diseases, and is fundamental in the development and progression of these pathologies [7]. Identification of novel inflammatory factors and mechanisms involved in chronic neuroinflammation is essential to improve our understanding of the pathology of age-related brain diseases, and subsequently to provide new therapeutic targets.

Neuroinflammation occurs already during early stages of age-related brain diseases. Notably, aging itself is accompanied by the gradual development of a chronic low-grade inflammatory state (termed inflammaging) [12]. Moreover, (neuro)inflammation is known to occur and to play a role in several risk factor conditions (such as obesity and cardiovascular disease) for age-related brain diseases [3]. Inflammatory processes in the periphery and CNS may be a link between aging, risk factor conditions, and development of age-related CNS diseases.

Lipocalin 2 (Lcn2) is an age-associated (neuro)inflammatory factor and may be involved in several risk factor conditions and age-related CNS disorders. Systemic levels of Lcn2 were found to increase with age. The expression of Lcn2 is further increased in several age-related CNS diseases, as well as in different conditions that are risk factors for these age-related CNS disorders. Evidence suggests that Lcn2 may contribute to these risk conditions and age-related brain diseases, by affecting several processes such as inflammation, cell death/cell survival signaling, and iron dysregulation [13,14]. Hence, we hypothesize that chronically increased Lcn2 levels as a result of unhealthy aging might prime and sensitize the brain, rendering the brain more vulnerable to develop different age-related brain diseases.

In this review, we will discuss Lcn2 as an inflammatory component linking aging, risk factor conditions and the development of age-related brain diseases, including: AD, PD and VaD. Of note, Lcn2 is also known as neutrophil gelatinase-associated lipocalin (NGAL), 24p3,

(5)

100

siderocalin, uterocalin, 24 kDa superinducible protein (SIP24), and neu-related lipocalin, and is generally referred to as Lcn2 or 24p3 in mice, and as NGAL in humans. For consistency, Lcn2 will be used as further reference throughout this review.

2. Biochemical characteristics and functions of Lcn2

Biochemical characteristics and binding partners of Lcn2

Lcn2 is a 25 kDa member of the lipocalin protein family, which comprises a group of more than 20 small secretory proteins that are involved in transport of hydrophobic ligands. Members of the lipocalin family are notably heterogeneous regarding amino acid sequence, with a sequence homology that can be lower than 20% [15–17]. However, all lipocalins share one to three characteristic conserved sequence motifs, and present a comparable structural hallmark named the lipocalin fold. The lipocalin fold is formed by a single eight-stranded antiparallel β-sheet which closes on itself by hydrogen bonds to form a β-barrel [18]. The internal cavity of this cup-shaped β-barrel, together with an external loop scaffold, presents the site where ligands can bind. The specific composition of this ligand-binding site varies between different lipocalin members, explaining the variation in ligands that different lipocalins can bind to and carry [15,16,19,20]. Lcn2 contains a more polar binding cavity in comparison to other lipocalins, large enough to allow Lcn2 to bind small hydrophobic molecules as well as certain bigger soluble macromolecules [21–23].

Ligands of Lcn2

Different ligands have been identified for Lcn2 (also illustrated in Fig. 1). As was anticipated for a lipocalin family member, different small hydrophobic ligands were reported to be bound by Lcn2. For example, Lcn2 was reported to bind to the hydrophobic molecules cholesteryl oleate, retinol, linoleic acid, platelet‐activating factor, leukotriene B4 and the chemotactic peptide N-formyl-Met-Leu-Phe (fMLP) [23–26]. Other mediators of inflammation have been proposed as potential ligands of Lcn2 as well, including lipopolysaccharide (LPS) [22,27].

Interestingly, a new view of Lcn2’s ligands and functions was obtained when Goetz and colleagues in 2002 discovered the bacterial siderophore enterobactin as a ligand of Lcn2 [21,23]. Siderophores such as enterobactin are small ferric (Fe3+) iron-chelating molecules that can be secreted by several microbial species (including different bacteria and fungi) in order to collect the iron that is required for their growth. As Goetz et al. discovered, the host innate immune system can defend itself against bacterial infection by producing and releasing Lcn2. Lcn2 can bind to iron-loaded (and iron-free) bacterial siderophores, and as such interfere with siderophore-mediated bacterial iron acquisition. Lcn2 is able to bind to catecholate- and mixed phenolate-type bacterial siderophores, thereby providing protection against bacteria that depend on these siderophore types, including for example E. coli and mycobacteria such as M. tuberculosis [13,21,28–33]. Accordingly, Lcn2 may not be able to interfere with the iron-thievery and growth of microbes that use other types of siderophores,

(6)

101

6

Figure 1. Ligands, receptors and post-translational modifications of Lcn2, and effects of Lcn2. Lcn2 can bind

various ligands, including (iron-bound and iron-free) bacterial and mammalian siderophores, different small hydrophobic molecules, and membrane phosphatidylethanolamine (PE). Lcn2 can also form complexes with other Lcn2 molecules, as well as with matrix metalloproteinase 9 (MMP-9), MMP-2 and hepatocyte growth factor (HGF). Lcn2 can be post-translationally modified by polyamination, phosphorylation and glycosylation. Known receptors for Lcn2 include 24p3R, megalin, MC4R, and possibly MC1R and MC3R. Lcn2 is involved in different processes, ranging from bacterial defense and iron regulation to inflammation and regulation of cell death/cell survival mechanisms.

or utilize siderophore-independent pathways to scavenge iron [23,34–39]. In addition, bacteria may interfere with the antibacterial functions of Lcn2, for example by producing factors (such as cyclic diguanylate monophosphate (c-di-GMP)) that can strongly bind Lcn2, thereby competing with the binding between Lcn2 and bacterial siderophores [40].

Importantly, it became clear that Lcn2 may not only influence bacterial iron regulation, but may also play a role in physiological mammalian iron regulation [41,42]. Indeed, mammalian siderophores were identified, which enable Lcn2 to bind to ferric iron [23]. Mammalian siderophores known so far include for example simple catechols (including catechol, 3-methylcatechol, 4-methylcatechol and pyrogallol), which are diet-derived metabolic products [23,43–46]. These simple catechols are abundantly present in e.g. the blood circulation and urine, and catechol-iron-Lcn2 complexes can for example be detected in human urine where they may influence Lcn2’s antimicrobial activity in the urinary tract [43–45]. In addition to these simple catechols, the neuroendocrine catecholamine L-norepinephrine and the iron-binding moiety 2,5-dihydroxybenzoic acid (2,5-DHBA) were both found to form a complex with iron and Lcn2 [47,48]. By binding iron-loaded L-norepinephrine, Lcn2 could inhibit L-norepinephrine-mediated bacterial iron scavenging [48].

(7)

102

Production of 2,5-DHBA (via synthesis by the enzyme 3-hydroxybutyrate dehydrogenase-2 (BDH2)) was shown to be required for normal iron metabolism [36,49,50]. Finally, certain polyphenols present in green tea were reported to form a complex with Lcn2 and iron, representing another diet-derived type of Lcn2-binding siderophore in the body [23,51,52]. Taken together, next to interfering with bacterial iron acquisition by catching away bacterial siderophores, Lcn2 may also play an important role in physiological mammalian iron homeostasis, via binding mammalian siderophores. Of interest, Lcn2 may also use siderophores to bind more exotic metal ions including radioactive plutonium and curium [53].

Other binding partners of Lcn2

Besides the small hydrophobic and siderophoric ligands described above, some additional binding partners of Lcn2 are known. Firstly, Lcn2 can covalently bind matrix metalloproteinase 9 (MMP-9), which is involved in extracellular matrix degradation and tissue remodeling [54–57]. Lcn2 can protect MMP-9 against degradation, and as such may increase the stability and activity of MMP-9 [55,58–60]. Secondly, Lcn2 was reported to bind to and to possibly promote the stability and activity of matrix metalloproteinase 2 (MMP-2) as well [61–64]. Thirdly, it was reported that Lcn2 can bind to and partly inhibit the activity of hepatocyte growth factor (HGF), thereby reducing HGF-stimulated cell branching in kidney tubular epithelial cells [65]. Fourth, Lcn2 was found to attach to the sperm membrane by binding to membrane phosphatidylethanolamine (PE), which resulted in stimulation of lipid raft movement and cholesterol efflux [66–68]. The mechanisms that underlie these Lcn2-mediated effects in the sperm membrane however are not clear yet [66]. Finally, Lcn2 can bind itself: Lcn2 not only exists as a monomer, but may also form homodimers and homotrimers [54,55]. While monomeric Lcn2 appears to be the most common form, certain cell types seem to be related with formation of homo-multimeric forms of Lcn2 [55,69–71].

Receptors for Lcn2

Lcn2 can bind to the multi-ligand receptors 24p3R and megalin, which were both shown to mediate internalization of Lcn2 into cells [42,72]. 24p3R (also known as SLC22A17, LCN2R, NGALR and brain type organic cation transporter (BOCT)) also binds albumin, metallothionein, phytochelatins and possibly transferrin, when they are not outcompeted by Lcn2 [73–75]. Other ligands for megalin (also known as Lrp2 and gp330) include albumin, insulin, insulin-like growth factor 1, MMP-9 and hemoglobin [76,77]. Of note, different alternative splice variants were found for 24p3R, which all appeared to be functional receptors for Lcn2, yet possibly with different affinities for Lcn2 [42,78].

Furthermore, it was recently shown that Lcn2 can also bind to and activate the melanocortin 4 receptor (MC4R) [79]. In addition, it was suggested that MC1R and MC3R may be activated by Lcn2 [79]. This adds Lcn2 to the list of melanocortin receptor ligands, which otherwise consists of melanocortins such as the agonistic α- and β-melanocyte-stimulating hormones (α-MSH and β-MSH) and adrenocorticotropic hormone (ACTH), and the antagonistic Agouti and Agouti-related peptide [80].

(8)

103

6

Altogether, at least three functional Lcn2 receptors are currently known (including 24p3R, megalin, MC4R and possibly MC1R and MC3R). The signaling mechanisms mediated by these receptors upon Lcn2 binding, and their potential differential affinities for different ligand-bound states of Lcn2, should be further elucidated in future studies. For example, both 24p3R and megalin are able to take up iron-free Lcn2 (apo-Lcn2) as well as iron-bound Lcn2 (holo-Lcn2). However, while megalin does not appear to have a binding preference for either form, there is evidence that 24p3R does present a differential affinity for iron-free versus iron-bound ligands [41,42,72,81].

Post-translational modifications

Lcn2 can also be subject to post-translational modifications. Firstly, Lcn2 is a glycoprotein and may be secreted in at least two different glycosylation isoforms (glycoforms), due to different N-linked glycosylation patterns [82–86]. Different glycosylation patterns may relate to the tissue in which Lcn2 is produced and to the stimulus that induces its production, and might affect its stability and solubility [82,87]. Secondly, Lcn2 can be polyaminated, resulting in faster clearance of Lcn2 from the circulation. On the other hand, deamidation (removal of polyamine groups) of Lcn2 in adipose tissue can delay its clearance and promote its detrimental accumulation in arteries [25,88]. Lastly, Lcn2 can also be phosphorylated [89,90]. Phosphorylation of Lcn2 by protein kinase C delta (PKCδ) was found to influence the released levels of Lcn2 from neutrophils upon stimulation, possibly via affecting the secretory mechanisms that underlie Lcn2 secretion from neutrophils [89].

Taken together, Lcn2 can exist in different ligand-bound, complexed and post-translationally modified states, and is able to bind to different receptors. More research is required to clarify how these various states of Lcn2 in the body may influence its functions and effects.

Functions and expression of Lcn2

Lcn2 is known to be involved in a wide range of processes, including the defense against specific bacterial infections, regulation of mammalian iron homeostasis, anti- and pro-apoptotic signaling, anti- and pro-inflammatory responses, chemotaxis, cell migration, cell differentiation and energy metabolism (Fig. 1) [13,14,79].

Lcn2 in the periphery

Under normal physiological conditions in adults, expression of Lcn2 is low and limited to specific cell types. Constitutive low expression of Lcn2 under healthy circumstances is found in neutrophils, bone marrow, bone osteoblast cells, adipose tissue, heart, blood vessels, uterus, prostate and salivary gland, as well as in tissues that are normally exposed to microorganisms, including epithelial cells in most parts of the respiratory, urinary and gastrointestinal tracts [72,79,91–96]. Continuous Lcn2 expression in these epithelial cell types and constitutive Lcn2 storage in neutrophils are likely related to the readiness of these cell types to respond to pathogenic, inflammatory and tissue damage-related stimuli. Bone-

(9)

104

and adipose tissue-derived Lcn2 plays a role in metabolic regulation (including glucose and insulin homeostasis, appetite and food intake) [79,97]. Moreover, basic Lcn2 levels may be involved in maintaining iron homeostasis [98]. The known receptors for Lcn2 (including megalin and 24p3R) are widely expressed throughout the body [14,42,72,99], indicating that many tissues may be sensitive for and able to respond to Lcn2. Of note, MC4R may be primarily expressed in the CNS [80].

While Lcn2 expression is low and limited to specific cell-types under healthy conditions, Lcn2 expression can increase manifold in various cell types upon different acute and chronic challenges. Lcn2 is an acute-phase protein, and is rapidly produced in response to pathogen exposure, tissue injury and inflammatory stimuli such as LPS [13,28,91]. In addition, Lcn2 levels are significantly increased in various chronic conditions, such as in different types of cancer, metabolic diseases (including obesity and diabetes), heart failure, arthritic diseases and chronic kidney disease [13,88,96,97,100–102]. Depending on the condition, Lcn2 can be produced by many different tissues, including for example: neutrophils, macrophages, adipose tissue, cardiomyocytes, epithelial cells in the respiratory tract, gut, peritoneum, kidney, liver and vascular endothelium [13,28,96,97,100,102–106]. Lcn2 was shown to play an important role in several of these acute and chronic conditions, for example via its effects on iron homeostasis, cell survival and cell death, cell proliferation and inflammation [13,14,100,102,103,107]. Depending on the condition, also the antibacterial effects of Lcn2 (via hijacking bacterial iron acquisition as well as promoting neutrophil functioning) can be essential [28,29,108,109].

Lcn2 in the CNS

The expression and functions of Lcn2 in the CNS have only been explored more recently. Studies in mice illustrated that Lcn2 mRNA and protein expressions in the brain are low during normal physiological conditions [110–114]. Also in human post mortem brain tissue low Lcn2 protein expression was observed in healthy controls, in comparison to patients with gliomas and AD [93,115,116]. Of interest, as suggested by Ferreira et al. the basal levels of Lcn2 in the brain might not only originate from the brain itself but also from the blood [117]. The low basal levels of Lcn2 in the brain may be involved in different processes, including maintenance of normal brain iron homeostasis, adult neurogenesis and synaptic activity/plasticity [110,117–120]. Moreover, basal Lcn2 levels may play a role in the control of behavior and cognition [13,117,118,120]. It can be hypothesized that Lcn2 may exert effects on different brain cell types and in multiple brain regions, considering the widespread expression of the known receptors of Lcn2 throughout the brain. Megalin is primarily present in the choroid plexus, ependymal cells of the lateral ventricles, and brain capillaries [121– 123]. Megalin expression on astrocytes and neurons has also been described [124–126]. Anatomical localization of 24p3R in the brain with in situ hybridization in mice showed that it is widely distributed throughout the gray matter regions, including cortex, thalamus and neurons in the granule layer of the cerebellum [112]. The highest expression levels were found in the choroid plexus, dentate gyrus and pyramidal cells of the hippocampus [99,112].

(10)

105

6

Furthermore, 24p3R can be expressed by neurons, microglia, astrocytes and endothelial cells [112,127,128]. MC4R is widely expressed throughout the brain as well, with expression found in various brain regions including the hypothalamus, thalamus, hippocampus, cortex, amygdala, brain stem and spinal cord [80]. MC4R expression was shown in neurons, astrocytes, microglia and oligodendrocytes [80,129].

Similar to many other tissues in the body, the mRNA expression and protein production of Lcn2 in the brain have been found to increase greatly upon various acute stimuli and chronic pathologies. For example, Lcn2 expression in the brain was found to be increased in different animal models of acute neuronal injury [130,131], as well as in mice that received peripheral or central injection with LPS [13,14,112,132]. Microarray analyses from several studies have identified Lcn2 as one of the highest upregulated genes in the brain and brain cells upon acute pro-inflammatory stimuli and neuronal damage [115,132– 136]. Increased brain Lcn2 levels are also present in different chronic CNS diseases, including multiple sclerosis (MS) and neurodegenerative conditions such as AD and PD [13,14,115,137– 139]. In most of the studied pathological CNS conditions, astrocytes appeared to be major producers of Lcn2 [128,137,138,140]. However, Lcn2 may also be produced by other brain cell types, including choroid plexus epithelial cells, brain endothelial cells, neurons, infiltrating neutrophils and possibly microglia [110,115,127,133,141–146]. The cell type(s) that produce Lcn2 may depend on the specific disease and disease stage that is being studied. Multiple studies so far have implicated that Lcn2 may significantly affect different CNS conditions, for example by influencing neuroinflammation, brain iron metabolism, and survival/death of brain cells [13,14].

Taken together, Lcn2 is induced in many different cell types throughout the body, in both acute and chronic conditions. By affecting multiple processes such as inflammation and iron regulation, Lcn2 may play a significant role in the pathophysiology of various diseases in the periphery and CNS. However, it is important to note that the effects of Lcn2 in many disorders are not fully understood yet. As mentioned above, Lcn2 may exert contrasting effects, including anti- and pro-apoptotic signaling, and anti- and pro-inflammatory responses. The effects exerted by Lcn2 may depend on a complex combination of several factors, including the pathology that is studied, the cell types that are involved, the states in which Lcn2 is present (e.g. iron-free or iron-bound), the relative expression of the different receptors for Lcn2, and the chronicity of Lcn2 exposure.

In the next chapter of this review, the potential role of Lcn2 in different age-related brain conditions will be discussed, including the neurotoxic and neuroprotective effects that have been reported for Lcn2.

(11)

106

3. The role of Lcn2 in age-related CNS conditions, and potential involved

mechanisms

Lcn2 in human and animal studies of AD, PD and VaD

So far, a few human and animal studies point to the possible involvement of Lcn2 in AD, PD and VaD (also see Table 1a). First of all, altered Lcn2 levels have been found in human tissues of AD, PD and VaD patients. In human post-mortem brain tissue of AD patients, elevated Lcn2 protein levels were detected in multiple brain regions that are affected by AD pathology, such as the hippocampus and prefrontal cortex [115,138]. Also in post-mortem brain tissue of PD patients, increased Lcn2 levels were measured in the substantia nigra [137]. No analyses of Lcn2 levels have been reported yet for VaD brain tissue. Concerning circulating Lcn2 levels, increased serum and plasma Lcn2 levels were found in PD and VaD patients [128,147]. Serum Lcn2 levels were not significantly increased in AD patients [115,138,148]. However, Lcn2 levels in cerebrospinal fluid (CSF) were significantly decreased in AD patients as compared to healthy age-matched control subjects, mimicking the characteristically decreased CSF amyloid-β (Aβ) concentrations in AD [115,138].

Corresponding to the upregulated Lcn2 expression seen in patients, Lcn2 levels were also found to be significantly elevated in cell culture and animal models of AD, PD and VaD [120,128,137,146,149]. Importantly, from these cell culture and animal studies it appeared that Lcn2 may significantly affect different pathological processes, such as neuroinflammation, cell death and iron metabolism, depending on the used experimental model. As such, it is possible that increased Lcn2 levels play an important role in the development and progression of AD, PD and VaD.

However, the evidence for the involvement of Lcn2 in these CNS diseases is limited. Moreover, the overall understanding of the functions and effects of Lcn2 in the healthy and diseased brain is still far from complete. To gain more insight into the potential neurotoxic and neuroprotective effects of Lcn2, we will summarize the current knowledge regarding Lcn2’s functions in the healthy and unhealthy brain. In this regard, we will not only discuss findings from cell culture and animal models of AD, PD and VaD, but also from other CNS conditions in which Lcn2 may play a role, such as stroke, MS and spinal cord injury (also see Table 1b). Even though these CNS conditions may not be linked with older age or may not be neurodegenerative in nature, different neuropathological processes may overlap with those in AD, PD and VaD. For example, neuroinflammation, iron dysregulation, white matter damage and blood-brain barrier (BBB) disruption are neuropathological processes that are shared between many CNS conditions. Nevertheless, it should be kept in mind that effects found for Lcn2 in one specific tested condition may not translate to similar effects in another condition. For example, contradictory (e.g. neurotoxic versus neuroprotective) effects of Lcn2 have been found even in similar study set-ups, as will be described below. Therefore, it is important to note that the effects of Lcn2 may depend strongly on, amongst others, the

(12)

107

6

specific disease, disease stage and the acute versus chronic nature of the disease that is studied.

Table 1. Evidence for the involvement of Lcn2 in AD, PD and VaD (Table 1a) and other CNS conditions (Table 1b),

including data from human subjects and overall findings from cell culture and animal studies.

Table 1a. Lcn2 in AD, PD and VaD CNS

condition Model Involvement/effects of Lcn2

Cell types in which Lcn2

was located Ref.

AD Human AD

patients

Lcn2 protein levels are increased in affected brain regions (including hippocampus, prefrontal cortex, amygdala, anterior cingulate cortex). Lcn2 levels are decreased in CSF, compared to healthy age-matched controls. Serum Lcn2 levels were not significantly different.

Neurons and astrocytes. [115,13 8,148]

AD mouse models

Lcn2 protein levels are increased in the brain in the J20 (12 mo.) AD mouse model. Also, hippocampus and choroid plexus Lcn2 mRNA expression levels are increased in APP/PS1 (~12 wk) mice, and WT mice that received i.c.v. oligomeric Aβ injection. Increased Lcn2 mRNA levels were also found in brain tissue in young (2 mo) but not older (12 mo) Tg2576/PS-1P264L/P264L AD mice. Astrocytes, choroid plexus cells. [120,14 9,249] AD mouse model

Lcn2 does not significantly affect AD-like behavioral changes, cognitive impairment, plaque load and glial activation, but does contribute to brain iron accumulation, in the J20 mouse model of AD (12 mo.).

Astrocytes. [120]

Cultured primary brain cells

Lcn2 production is induced in astrocytes and choroid plexus epithelial cells upon Aβ1-42 exposure. Treatment with Lcn2

sensitizes astrocytes and neurons to Aβ- and glutamate-induced cell death.

Astrocytes, choroid plexus epithelial cells.

[115,14 6] and Dekens et al., 2019 in prep. PD Human PD patients

Lcn2 protein levels are significantly increased in the substantia nigra of PD patients.

Lcn2 levels are significantly increased in serum of PD patients. Also, PD subjects demonstrate a positive

relationship between CSF NGAL and CSF α-synuclein, as well as between CSF NGAL and CSF Aβ40 levels.

[137,14 7] PD mouse models (MPTP and 6-OHDA models)

Lcn2 mRNA and protein expression is significantly increased in the substantia nigra and striatum in two neurotoxin mouse models of PD. Lcn2 promotes neuronal death and neuroinflammation, thereby contributing to disruption of the nigrostriatal dopaminergic pathway and disturbance of locomotor behavior. Mostly astrocytes, microglia. [137] Cultured primary brain cells

Conditioned medium from differentiated SH-SY5Y neurons pretreated with 1-methyl-4-phenylpyridinium (MPP+), induces Lcn2 production in cultured glia. Astrocyte-derived Lcn2 promotes neurotoxicity of MPP+, in co-cultures of mesencephalic neurons and astrocytes.

(13)

108

VaD Human VaD

patients

Plasma Lcn2 levels are significantly higher in VaD patients. [128]

VaD mouse model (tBCCAo and cUCCAo models)

Hippocampal Lcn2 levels are significantly increased in two mouse models of VaD. Lcn2 contributes significantly to neuronal loss, neuroinflammation, white matter damage, BBB disruption and cognitive impairment, as shown by comparing WT and Lcn2 KO VaD mice. In addition, i.c.v. injection with recombinant Lcn2 caused neuronal death in the hippocampal CA1 region, and caused cognitive deficits.

Astrocytes. [128]

Cultured primary brain cells

Recombinant Lcn2 reduces the viability of cultured hippocampal neurons. Treatment with Lcn2 induces production of NO, IL‐1β, and TNF‐α by primary microglia. Lcn2 promotes microglia-mediated neurotoxicity. Chemically induced hypoxia by CoCl2 treatment induces

Lcn2 production in primary astrocytes, which was dependent on HIF‐1α.

Astrocytes. [128]

Table 1b. Lcn2 in other CNS conditions CNS

condition Model Involvement/effects of Lcn2

Cell types in which Lcn2

was located Ref.

AD in Down syndrome (DS) Human DS individuals that did/did not develop AD dementia

Serum Lcn2 levels are significantly increased in DS

individuals compared to non-DS people. Serum Lcn2 levels are not associated with clinical dementia symptoms in DS. Serum Lcn2 levels are associated with distinct Aβ species, depending on the progression of dementia (DS without AD, DS with AD at baseline, DS with conversion to AD).

[250]

Human DS individuals

Serum Lcn2 levels are increased in DS individuals, and correlate positively with increasing age.

[251] Hemorrhagic stroke Human hemorrhagic stroke patients

Serum Lcn2 levels are significantly increased in hemorrhagic stroke patients, when compared to healthy controls and ischemic stroke patients (pilot study).

[186]

Mouse and rat models of hemorrhagic stroke

Lcn2 production is increased upon hemorrhagic stroke in the brain. Lcn2 contributes to brain injury and neurological deficits after hemorrhagic stroke (including increased neuroinflammation, BBB disruption and neuronal death).

Astrocytes (most clearly), neurons, microglia and endothelial cells. [143,15 1,166,1 82,194, 252,25 3] Ischemic stroke Human ischemic stroke patients

Lcn2 levels are increased in human brain tissue and blood after ischemic stroke. Higher plasma levels of Lcn2 in the first week after ischemic stroke are associated with a worse clinical outcome at 90 days, a higher cardiovascular mortality in the 4 years after stroke, and with the presence of post-stroke infections.

Neurons. [145,17

8,252,2 54– 258]

Mouse and rat models of ischemic stroke (ischemia-reperfusion

Lcn2 production is increased during ischemia-reperfusion injury in the brain, and increased Lcn2 levels are also detectable in the blood. Lcn2 contributes significantly to ischemia-reperfusion injury in the brain (including increased neuroinflammation, BBB disruption and neurotoxicity) and worsens neurological deficits.

Astrocytes, endothelial cells, macrophages/ microglia, infiltrating neutrophils (23h after stroke, mouse). Neurons (3 days after

[127,13 4,141,1 45,178, 259,26 0]

(14)

109

6

injury) stroke, rat).

Cultured primary brain cells

Lcn2 stimulates glia to adopt a ‘pro-recovery’ phenotype, thereby providing neuronal protection against oxygen-glucose deprivation.

Neurons. [145]

FTLD/ALS Human

FTLD/ALS patients

Lcn2 expression is upregulated in the brains of FTLD patients. Lcn2 plasma protein levels are significantly increased in ALS patients.

Astrocytes. [140,26

1]

Transgenic rat models of FTLD/ALS

Lcn2 levels are increased in brain tissue of transgenic rats expressing mutant TDP-43, FUS or SOD1. Lcn2 levels in CSF increased with disease progression in mutant TDP-43 expressing rats. Astrocytes. [140,26 2] Cultured primary brain cells

Lcn2 is neurotoxic, and primary neurons expressing ALS-related mutant TDP-43 or FUS display a further increased sensitivity to Lcn2-induced cell death compared to WT neurons.

[140]

Progranulin-deficient mice

Lcn2 expression is increased in the brain of progranulin-deficient mice; in which features of FTLD are mimicked.

[263]

MS Human MS

patients

Increased Lcn2 expression is present around lesions in the brain of MS patients. Lcn2 levels are significantly increased in plasma and CSF of (perhaps especially progressive) MS patients. However, in early stages of MS Lcn2 levels in plasma and CSF may be decreased. In clinically isolated syndrome patients, higher CSF Lcn2 levels are associated with conversion to clinically definite MS.

Monocyte/macrophages and granulocytes (neutrophils?), in the blood vessel lumen and perivascular cuffs of active lesions. [135,13 9,179,1 95,264] Mouse model of MS (EAE)

Lcn2 mRNA and protein expression is upregulated in spinal cord and choroid plexus in the EAE mouse model. Lcn2 levels are also increased in the CSF, and corresponded with active disease phases. Lcn2 may significantly affect EAE severity, yet both detrimental and beneficial effects have been reported.

Infiltrating neutrophils (choroid plexus), astrocytes (brain parenchyma and spinal cord), microglia and monocytes (spinal cord).

[135,13 9,150,2 65] Traumatic brain injury (TBI) Human TBI patients

Lcn2 levels are significantly increased in the brain and serum of TBI patients. Increased serum Lcn2 levels are associated with trauma severity, and are a predictor of mortality after head trauma.

Neurons. [266,26

7]

Mouse and rat models of TBI

Lcn2 mRNA and protein expression is increased in the brain after TBI. Astrocytes. [136,26 8,269] Spinal cord injury Mouse model of spinal cord contusion injury

Lcn2 protein levels are significantly increased after spinal cord contusion injury. Lcn2 worsens locomotor recovery, and increases inflammation, neuronal loss, myelin loss and tissue damage after spinal cord contusion injury.

Astrocytes, neurons and infiltrating neutrophils in the spinal cord.

[130]

Abbreviations: Aβ; Amyloid-β, AD; Alzheimer’s disease, ALS; amyotrophic lateral sclerosis, BBB; blood-brain barrier, CoCl2; cobalt chloride, CSF; cerebrospinal fluid, cUCCAo; chronic unilateral common carotid artery

occlusion, DS; Down syndrome, EAE; experimental autoimmune encephalomyelitis, FTLD; frontotemporal lobar degeneration, FUS; fused in sarcoma, HIF‐1α; hypoxia-inducible factor 1 alpha, i.c.v.; intracerebroventricular, IL‐ 1β; interleukin 1 beta, MCI; mild cognitive impairment, MPTP; 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine, MS; Multiple Sclerosis, NO; nitric oxide, PD; Parkinson’s disease, SOD1; superoxide dismutase 1, TDP-43; TAR DNA-binding protein 43, tBCCAo; transient bilateral common carotid artery occlusion, TNF‐α; tumor necrosis factor alpha, VaD; vascular dementia, 6-OHDA; 6-hydroxydopamine.

(15)

110

Potential neuroprotective and neurotoxic effects of Lcn2

Lcn2 was suggested to play a role in different neuro(patho)physiological processes, which will be discussed here (also see Figure 2).

Neuroinflammation

Neuroinflammation is an important player in many if not all CNS conditions. As found in several cell culture and animal models, Lcn2 expression is greatly induced by various inflammatory and pathological factors, after which Lcn2 may influence the activation of astrocytes and microglia. Many studies – as performed for example in models of AD, VaD, PD, amyotrophic lateral sclerosis (ALS), frontotemporal lobar degeneration (FTLD), stroke, MS, spinal cord injury and LPS-induced sepsis – have found that Lcn2 promotes pro-inflammatory activation of glia, and may in certain conditions enhance infiltration of neutrophils and macrophages into the brain [127,128,130,137,141–143,145,150–159].

Some contradictory findings should be mentioned. Firstly, no differences in glial activation were found in 12 months old transgenic AD mice in which Lcn2 was either present or knocked out [120]. Also, Lcn2 did not affect astrocyte activation in a mouse model of systemic lupus erythematosus (SLE) [160], and did not influence neuroinflammation in mice with West Nile virus encephalitis [111], or in a mouse model of neuroinflammation-induced cerebellar degeneration [161]. Moreover, while the majority of research indicated that Lcn2 aggravates LPS-induced neuroinflammation (modelling sepsis) [142,152,153,156], there is one study in which no effect of Lcn2 on glial activation was found upon LPS stimulation [112], and one study in which even opposite, anti-inflammatory, effects of Lcn2 were reported [162]. In addition, while one group found significant pro-inflammatory effects of Lcn2 in the spinal cord in the experimental autoimmune encephalomyelitis (EAE) mouse model of MS [150], another group found anti-inflammatory effects of Lcn2 in EAE [139]. The reasons behind these contradictory effects of Lcn2 on neuroinflammation are not clear, but may depend on the specific CNS condition that was modeled, the exact experimental set-up that was used, the disease stage, the involved cell types, the chronicity of Lcn2 overexpression and the age of the animals. More work is required to clarify the conditions that determine whether Lcn2 may exert e.g. pro- or anti-inflammatory effects. Moreover, the exact mechanisms underlying the potential immunomodulatory effects of Lcn2 warrant further investigation. For instance, Lcn2 was recently shown to induce activation of the NLRP3 (nucleotide-binding oligomerization , leucine-rich repeat- and pyrin domain-containing 3) inflammasome in cardiac fibroblasts, resulting in caspase-1 activation and interleukin 1 beta (IL-1β) production [163]. It would be of interest to assess whether Lcn2 may also influence glial activity via this mechanism.

(16)

111

6

Figure 2. Neurotoxic and neuroprotective effects of Lcn2. Lcn2 may exert various neurotoxic and

neuroprotective effects in the brain, which may depend on the specific CNS disease and disease stage that is studied. The effects of Lcn2 on e.g. neuronal survival/death, neuroinflammation, neutrophil infiltration, brain iron metabolism, blood-brain barrier (BBB) disruption and white matter damage (and additional effects as described in Chapter 3 of this review) may play a role in multiple CNS disorders, such as AD, PD, VaD, stroke-induced brain injury, FTLD, ALS, MS.

Cell death

Death of brain cells occurs in neurodegenerative diseases, as well as in CNS injury. Lcn2 was found to sensitize different brain cell types (including neurons, astrocytes and microglia, and possibly infiltrating cell types such as neutrophils) to cell death [115,127,128,130,137,140– 143,146,152–154,164–166]. Lcn2 may reduce the survival of at least certain cell types when iron-free Lcn2 is taken up by (24p3R expressing) cells, after which Lcn2 binds intracellular iron and mediates the export of iron from the cell [42]. This Lcn2-mediated cellular iron deprivation was found to induce expression of the pro-apoptotic protein BCL2-interacting mediator of cell death (BIM) in different cell types (including astrocytes, neurons and hematopoietic cell types), leading to apoptotic cell death [42,155,165,167]. However, other studies indicated that induction of BIM is not always required for Lcn2-mediated cell death [115,154]. As such, it is likely that Lcn2 may mediate sensitization to cell death in multiple ways. For example, besides inducing cellular iron deprivation, Lcn2 may also affect cell viability via promoting pro-inflammatory glial activation and secretion of pro-inflammatory

(17)

112

cytokines, and via inhibiting certain protective signaling pathways (such as signaling via tumor necrosis factor receptor 2 (TNFR2) [115]). Moreover, while Lcn2 may induce apoptosis in certain cell types by exporting iron, Lcn2 may also promote cell death in certain cells types by inducing cellular iron accumulation [143,168].

Interestingly, some contradictory findings have been reported regarding the pro-apoptotic effects of Lcn2 on brain cells. Firstly, disagreement exists whether Lcn2 is able to induce cell death by itself, or whether Lcn2 only sensitizes cells to cell death in the presence of other inflammatory or toxic stimuli, such as tumor necrosis factor alpha (TNF-α), glutamate, nitric oxide, hydrogen peroxide or Aβ. While different studies suggest the latter [115,146,154,155,165], Lcn2 was found to induce significant toxic effects on its own in other studies [128,137,140,141]. Secondly, some inconsistent findings have been reported concerning the brain cell types that are sensitive to Lcn2-mediated toxicity. While some studies concluded that Lcn2 only affects the viability of neurons and not that of glia [140], other studies found that Lcn2 also capably sensitizes astrocytes and microglia to cell death [146,154,155,160]. These differences might depend on whether studies were performed in

vitro or in vivo, and on the specific toxic factors that were present (e.g. TAR DNA-binding

protein 43 (TDP-43) or Aβ). Lastly, while most studies indicate that Lcn2 exerts cytotoxic effects, one study reported that Lcn2 may be secreted by endangered neurons as a ‘help-me’ signal upon oxygen-glucose deprivation. Increased Lcn2 levels subsequently stimulate glia to adopt a pro-recovery phenotype, resulting in neuroprotection and a decrease in neuronal cell death [145]. Hence, it should be taken into account that Lcn2 may induce neuroprotective effects in certain situations. In addition, no effect of Lcn2 on cell death was found in a mouse model of neuroinflammation-induced cerebellar degeneration [161].

Iron dysregulation

Iron dysregulation and accumulation is found in many neurodegenerative diseases and types of CNS damage and may significantly contribute to CNS injury, for example via increasing oxidative stress (via the Fenton reaction), inducing ferroptosis, and promoting aggregation of pathogenic proteins such as Aβ [169–176]. Lcn2 is involved in the regulation of iron homeostasis, and is able to mediate both import and export of iron into and out of cells [42]. Absence of Lcn2 in healthy unchallenged Lcn2 KO mice appeared to cause intracellular iron accumulation in certain cell types, including macrophages, hippocampal neurons and neural stem cells [98,117,120,177]. Thus, complete absence of Lcn2 may induce accumulation of intracellular iron in certain cell types. Oppositely, elevated Lcn2 levels as present in different CNS conditions may promote iron accumulation as well. For example, we recently found increased hippocampal iron accumulation (especially in plaques and hippocampal pyramidal and granular neurons) in transgenic AD mice as compared to wildtype (WT) mice, but significantly decreased hippocampal iron accumulation in Lcn2-deficient AD mice as compared to AD mice [120]. This difference between AD and Lcn2-deficient AD mice also seems to correspond with in vitro data, in which WT astrocytes presented increased ferritin mRNA expression upon Aβ-treatment (indicating elevated iron storage facilities, possibly

(18)

113

6

resulting in iron accumulation), whereas Lcn2 KO astrocytes did not [146]. However, these effects of Lcn2 on astrocytic ferritin mRNA expression may not translate to the protein level (Dekens et al., 2019, in preparation). This indicates that Lcn2 might not mediate significant iron accumulation in astrocytes, but preferably in other brain cell types (possibly including neurons and microglia) and structures such as Aβ plaques. Further support for Lcn2-mediated brain iron accumulation under pathological CNS conditions comes from findings in a mouse model of intracerebral hemorrhage, showing that upregulation of ferritin upon hemorrhage was lower in Lcn2 KO mice as compared to WT mice, indicating less iron accumulation in Lcn2 KO mice [143]. Moreover, intranigral iron administration aggravated Lcn2-induced loss of dopaminergic neurons in the substantia nigra, while administration of an iron chelator reduced Lcn2-induced cell death. These findings support the possibility that intracellular iron accumulation is an important mechanism involved in Lcn2-mediated loss of dopaminergic neurons [137]. In addition, in a mouse model of ischemic stroke, increasing Lcn2 levels in the brain were paralleled by cellular iron accumulation, which appeared to occur mostly in macrophages/ microglia [178]. Another notable finding is that CSF Lcn2 levels were found to correlate with CSF transferrin levels and with iron accumulation in the basal ganglia in clinically stable MS patients [179].

Taken together, it appears that iron dysregulation might occur both when Lcn2 is completely absent (in Lcn2 KO mice) and when Lcn2 production is increased, such as in several CNS conditions. While several findings indicate that high Lcn2 levels may increase cellular iron accumulation, it is important to note that this effect may be cell type- and disease-specific. It is possible that Lcn2 promotes pathological iron accumulation in certain cell types, and at the same time mediates iron export and iron deprivation in other cell types (which might e.g. depend on the relative expression of Lcn2 receptors). Both iron accumulation and iron deficiency in cells can have significant pathological effects, including induction of inflammatory changes, oxidative stress and cell death. Thus, Lcn2-mediated iron dysregulation might significantly affect cellular health and viability, potentially via promoting both cellular iron accumulation and iron deprivation, depending on the specific cell type [42,117,137,143,155,168].

Blood-brain barrier (BBB) disruption

BBB disruption may play an important role in different CNS injuries and diseases including AD and PD [180,181]. In mouse models of ischemic and hemorrhagic stroke, Lcn2 was found to aggravate BBB disruption [127,128,143,166,182]. The mechanisms underlying this effect of Lcn2 may include Lcn2-mediated induction of vascular endothelial growth factor A VEGFA [128], which has been implicated in BBB disruption. Another possibility is that the interaction of Lcn2 with MMP-9 is of importance in Lcn2-mediated BBB damage. Namely, MMP-9 may contribute significantly to BBB damage in several CNS conditions [183–185]. Since Lcn2 may protect MMP-9 from degradation and may prolong its activity, it could be that Lcn2 and MMP-9 form a toxic couple in the process of BBB disintegration [186].

(19)

114

Induction of inducible nitric oxide synthase (iNOS) and nitric oxide (NO)

Lcn2 was found to promote synthesis of inducible nitric oxide synthase (iNOS) and nitric oxide (NO) upon LPS injection and spinal cord injury in mice, and in LPS-stimulated astrocyte cultures [130,152,153,155]. Lcn2-mediated induction of iNOS and NO may be a potential mechanism via which Lcn2 can contribute to both neuroinflammatory and pro- and anti-apoptotic signaling pathways (and possibly BBB disruption) [187–190].

White matter damage

White matter damage occurs in many brain disorders, and is known to play a significant role in cognitive decline and dementia [191–193]. In mouse models of ischemic and hemorrhagic stroke, spinal cord injury and MS, Lcn2 was found to significantly aggravate the myelin loss that occurs in these models [128,130,150,182,194]. Accordingly, Lcn2 was reported to exacerbate axonal damage after subarachnoid hemorrhage, possibly due to loss of protective myelin [194]. In addition, Lcn2 inhibited myelination in neuroglial co-cultures [195].

Synaptic impairment

Synaptic impairment occurs early on in different CNS conditions [196–199], and it is possible that increased Lcn2 levels may affect synaptic and neuronal functioning. Lcn2 was proposed to act as a ‘help-me’ signal upon oxygen-glucose deprivation in a study by Xing et al., 2014 [145]. In this study, conditioned medium of Lcn2-treated glia had protective effects on neurons, and induced the neuronal expression of different synaptic markers (including synaptotagmin, synaptophysin and post-synaptic density 95) [145]. This finding may indicate a protective effect of Lcn2 on synapses. On the other hand, exposure of cultured hippocampal neurons to Lcn2 reduced the mobility of actin in dendritic spines, caused retraction of mushroom spines, and inhibited spine maturation [110]. Interestingly, it appeared that iron-free Lcn2 (rather than iron-bound Lcn2) was especially potent in inducing these effects [110]. Accordingly, it was shown that Lcn2 KO mice had a higher spine density in the hippocampus and basolateral amygdala as compared to WT control mice, and consequently neurons from the hippocampus and amygdala of Lcn2 KO mice were more excitable and fired more action potentials than WT neurons [110,119]. These findings indicate that Lcn2 may play an important physiological role in spine elimination in the hippocampus and amygdala, and may as such be involved in controlling and adapting behavior (including anxiety) under basic physiological conditions and in response to stimuli such as psychological stress [110,119]. Of note, it was reported that healthy unchallenged Lcn2 KO mice present reduced long-term potentiation in the dorsal hippocampus [118]. The potential protective (e.g. via promoting specific glial pro-recovery phenotypes) and/or damaging (e.g. via excessive spine elimination) effects on synapses by increased Lcn2 levels in different CNS conditions should be explored further in future studies.

(20)

115

6

Defects in neurogenesis

Adult neurogenesis is impaired in different CNS conditions including AD and PD, thereby reducing the chance that newborn neurons replace damaged or dead neurons [200–202]. One study has investigated the role of Lcn2 in adult neurogenesis in WT and Lcn2 KO mice under basal physiological conditions. It was found that absence of Lcn2 in Lcn2 KO mice caused a G0/G1 cell cycle arrest in neural stem cells, resulting in deficits in the proliferation, differentiation and maturation of neural stem/progenitor cells [117]. The cell cycle arrest in Lcn2 KO neural stem cells was likely due to intracellular iron accumulation and oxidative stress [117]. The results from this study indicate that normal physiological levels of Lcn2 are important for successful adult neurogenesis [117,203]. However, whether increased Lcn2 levels in CNS disorders may (positively or negatively) affect adult neurogenesis as well requires further investigation.

Oxidative stress

Oxidative stress plays a common role in many CNS conditions [204,205]. It is possible that Lcn2 is involved herein. However, data on Lcn2-mediated oxidative stress in CNS disorders is limited. It was shown that reactive iron accumulated in neural stem cells in Lcn2 KO mice, causing an increase in oxidative stress [117]. It is imaginable that increased Lcn2 levels as seen in different CNS conditions might also increase oxidative stress, for example by mediating iron accumulation and possibly by promoting NO production (which both may increase oxidative stress when in excess [188]), but this remains to be investigated. Interestingly, in rat primary cardiomyocytes iron-bound Lcn2 was shown to increase generation of mitochondrial reactive oxygen species (ROS), which relied strongly on the siderophore component present in iron-bound Lcn2 and not on Lcn2 alone [206]. Conversely, Lcn2 was found to have significant anti-oxidant properties in the liver of LPS-treated mice and in different peripheral cell lines, potentially in part by inducing the expression of heme oxygenase 1 (HO-1, which notably may also take on pro-oxidant and toxic properties e.g. in chronic conditions [207–209]) [146,210–214]. Hence, it appears that Lcn2 may exert both anti- and pro-oxidant effects in the periphery, which might also be the case in the CNS. The differential effects of Lcn2 on oxidative stress might depend on multiple factors, including the abundance of Lcn2-mediated NO production, the acute or chronic state of disease, the amount of iron in the microenvironment and the iron-free or iron-bound state of Lcn2. These factors may depend on the involved cell types and the specific disease studied.

Other effects: Mitochondrial dysfunction, defects in autophagy and insulin resistance

Besides the discussed processes, there may be other important neuroprotective/neurotoxic processes in which Lcn2 may be involved, for which only little evidence – mostly resulting from research in non-CNS tissue – is present at this moment. For example, it has been suggested that Lcn2 affects mitochondrial functioning, autophagy and insulin sensitivity. Impairments in all these processes play a role in various neurodegenerative diseases as well as in CNS injury due to for example ischemia [215–222]. Similar to several other effects of

(21)

116

Lcn2, also here differential effects of Lcn2 have been described, including findings of Lcn2-mediated protection against as well as Lcn2-Lcn2-mediated aggravation of mitochondrial dysfunction [168,206,223–225] and deficits in autophagy [226–228]. Also, several studies have reported that Lcn2 can dysregulate energy metabolism, by for instance promoting insulin resistance and decreasing glucose tolerance [226,229–234]. However, other investigations showed beneficial effects of Lcn2 on insulin sensitivity and glucose tolerance [79,235], and yet other studies did not detect a significant relationship between Lcn2 and insulin resistance/sensitivity [236–238]. As such, although results indicate that Lcn2 may indeed affect these processes, the direction and magnitude of its effects may depend on multiple factors such as the involved cell types, specific disease, and ligand-bound state of Lcn2.

Cognitive and behavioral changes

Many CNS disorders are accompanied by cognitive and behavioral changes. Lcn2 may be involved in these changes, perhaps via affecting some of the (patho)physiological processes described above. Interestingly, increased Lcn2 levels aggravated abnormal locomotor behavior (including locomotor activity and coordination) in a mouse model of PD, as well as in a mouse model of SLE [137,160]. Moreover, elevated Lcn2 levels were found to exacerbate memory impairment in mouse models of VaD, sepsis and SLE [128,153,160], and were correlated with cognitive decline in other studies [113,239,240]. Contrarily, no differences in working memory and long-term hippocampus-dependent memory functioning were found between AD mice and Lcn2-deficient AD mice [120]. Possibly, the absence of Lcn2-mediated effects on memory functioning in this study may be explained by the fact that a transgenic (‘J20’) mouse model of AD was investigated, which is a chronic (instead of acute) model, with perhaps relatively mild neuropathological and degenerative features. Of note, while increased Lcn2 levels may promote memory impairment in at least certain models of CNS disorders, complete absence of Lcn2 may disturb memory as well. It was shown that unchallenged Lcn2 KO mice under basal physiological conditions may present mild memory problems [117,118,120]. However, other findings showed that unchallenged WT and Lcn2 KO mice at baseline perform at the same level [120,128,153]. These differences may amongst others depend on the specific cognitive tests that were used, and the age of the studied mice.

Besides a role for Lcn2 in cognitive dysfunction, Lcn2 might also affect behavioral and neuropsychiatric changes that often accompany CNS disorders, such as anxiety and depression [241,242]. Lcn2 KO mice were shown to display anxiety-like behavior under unchallenged control conditions as well as upon psychological stress. This may be due to a lack in Lcn2-mediated spine deletion (resulting in a higher spine density and increased neuronal excitability) in the hippocampus and amygdala [110,118,119]. Lcn2 KO mice were also found to have increased circulating corticosterone levels [118]. Interestingly, although young Lcn2 KO mice (2-3 months old) were reported to present anxiety-like behavior under control conditions, no anxiety-like behavior was found in older Lcn2 KO mice (12 months old)

(22)

117

6

[118,120]. This finding poses the possibility that behavioral differences in unchallenged Lcn2 KO mice may normalize with increasing age [120]. Besides increased anxiety-like behavior, Lcn2 KO mice were also suggested to display depressive-like behavior, when tested in the forced-swim test at 2-3 months of age [118]. However, Lcn2 KO mice of 7 months old that were tested in the saccharin-preference test did not show a decreased preference for saccharin water, indicating no depressive-like anhedonia in Lcn2 KO mice as compared to WT mice [160]. As such, it might again be possible that young and older Lcn2 KO mice may differ in behavior, with depressive-like behavior subsiding with increasing age. However, to confirm this it would be essential to test young and old Lcn2 KO mice simultaneously, in exactly the same experimental set-ups. Notably, while absence of Lcn2 may thus affect depressive-like behavior, increased Lcn2 levels have been associated with depression as well [113,160,243– 245]. In humans, increased Lcn2 levels in serum and plasma have been associated with symptoms of depression [243–245]. Also, it was found that hippocampal Lcn2 levels are significantly increased in AD patients with co-existing depression as compared to AD patients without depression [138].

Notably, it seems that absence as well as overexpression of Lcn2 may induce changes in for example energy metabolism, iron metabolism, behavior and cognition. Thus, it appears that certain physiological processes depend on Lcn2 levels following an inverted U-shaped curve, with both absence and overexpression of Lcn2 resulting in the disruption of these processes. This characteristic of Lcn2 is comparable to other known inflammatory cytokines, including IL-1β, TNF-α and interleukin 6 (IL-6) [246–248].

Taken together, Lcn2 production is upregulated in different CNS diseases and injuries, in human patients as well as in animal models for these CNS conditions. In vitro and animal studies have shown that Lcn2 may strongly influence several pathological processes (including neuroinflammation and cell death). Moreover, the effects of Lcn2 may reach up to the level of overall disease outcome/prognosis, behavior and cognition. Although the functions and effects of Lcn2 in the healthy and diseased brain are not fully understood or predictable yet, it is clear that elevated levels of Lcn2 may significantly contribute to neuropathological processes in different age-related CNS conditions, including AD, PD and VaD.

(23)

118

4. Lcn2 levels in risk factor conditions for age-related brain diseases

Besides the increased Lcn2 production in multiple age-related CNS diseases, Lcn2 levels are also increased in several conditions that are risk factors for age-related CNS diseases. By being upregulated in these risk factor conditions (including for example aging, unhealthy lifestyle and chronic inflammatory diseases), it might be possible that Lcn2 is a biological link between risk factor conditions and the development of different age-related CNS disorders, such as AD, PD and VaD. This possibility will be explored in this chapter, by discussing several risk factor conditions in which Lcn2 may play a role (Figure 3).

Figure 3. Increasing Lcn2 levels during aging and risk factor conditions may contribute to the development of related brain diseases? Lcn2 levels increase during aging and several other risk factor conditions for

age-related CNS disorders. These risk factor conditions include for example physical inactivity and other unhealthy lifestyle factors, obesity, diabetes and different chronic inflammatory diseases. Lcn2 levels may increase in various peripheral tissues such as bone, adipose tissue, kidneys, gut, heart, blood vessels and the blood, depending on the present risk factor condition(s). Moreover, Lcn2 levels were also found to increase in the brain in certain risk factor conditions, such as depression, chronic stress, unhealthy diet, obesity and surgery. Possibly, rising peripheral and brain Lcn2 levels during aging and risk factor conditions may gradually drive the brain into a primed inflammatory and sensitive state, which may contribute to the development of different age-related CNS disease, such as AD, PD and VaD.

Aging

Aging is the major risk factor for the CNS conditions discussed here [270,271]. Interestingly, Lcn2 levels in blood were found to increase with age in different human cohorts [92,243,272– 275]. Notably, in the mouse brain, Lcn2 mRNA expression levels were found to rise with age (when comparing 4.5 months vs 26.5 months old mice) [276]. Further studies are required to confirm age-related upregulation of Lcn2 mRNA and protein levels in the body and brain. This would be in line with the chronic low-grade inflammatory state that gradually develops with increasing age, also known as inflammaging.

(24)

119

6

Mild cognitive impairment

Amnestic mild cognitive impairment (aMCI) is often a prodromal stage and risk factor of dementia, particularly AD dementia [277,278]. Blood Lcn2 levels were shown to be significantly increased in aMCI patients as compared to healthy age-matched control subjects, while Lcn2 levels in AD patients were comparable to those in the controls [115,148,279]. Furthermore, significantly decreased Lcn2 levels were found in CSF of MCI patients (as compared to healthy control), mimicking the decreased CSF Lcn2 levels seen in AD [115,138]. Taken together, Lcn2 levels are dysregulated in MCI. Whether dysregulated Lcn2 levels in MCI may contribute to or predict the conversion to AD should be investigated in future longitudinal studies, including MCI patients that do or do not convert to AD.

Depression

Depression is a risk factor for many CNS conditions including AD, PD and VaD [270,280–285]. Increased Lcn2 levels in plasma were found to be associated with depression in older persons (≥ 60 years of age), independent of age, male sex, use of anti-inflammatory drugs and lifestyle factors [243]. People with a recurrent depression had higher plasma Lcn2 levels than subjects with a first episode [243], and Lcn2 levels were especially increased in depressed persons with a higher waist circumference [286]. Notably, higher plasma Lcn2 concentrations in depressed subjects were associated with the sex-specific impairment of specific cognitive functions [240]. In women, higher Lcn2 levels were associated with impaired verbal memory and lower processing speed. In men, higher Lcn2 levels were associated with worse interference control [240]. Plasma Lcn2 levels in male rats were also associated with depressive-like behavior and cognitive dysfunction [113]. Of note, depression is a frequently observed co-morbidity in other diseases such as heart failure and AD, and is known to worsen the prognosis of these diseases [246,287–289]. Interestingly, depression and depression scores in heart failure patients were associated with increased serum Lcn2 levels [244–246]. Moreover, depression in AD patients was associated with increased Lcn2 levels in the hippocampus (and, intriguingly, with decreased Lcn2 levels in certain other brain regions, such as the prefrontal cortex) [138]. It remains to be investigated whether Lcn2 levels in the brain are also increased in depressed persons that do not suffer from AD. Studies in mice have suggested that both absence and overexpression of Lcn2 might contribute to development of depressive-like symptoms [118,160].

Physical inactivity and osteoporosis

Physical inactivity is associated with an increased risk to develop age-related CNS disorders including AD, PD and VaD [270,290,291]. Of interest, sedentary behavior was correlated with increased blood Lcn2 levels in humans [273,292,293]. For instance, in healthy human participants, bed rest was associated with a time-dependent rise in blood Lcn2 levels [292]. Accordingly, exercise (e.g. resistance and endurance training) was associated with decreased plasma Lcn2 concentrations in young men and elderly women [273,294]. In mice it was shown that Lcn2 is a mechanoresponsive protein, with strong induction in bone osteoblasts

Referenties

GERELATEERDE DOCUMENTEN

Therefore our objectives were (1) to investigate NGAL protein levels in serum, CSF and human post-mortem brain tissue (nine different brain regions) of control

Results were tested by one-way ANOVA followed by Tukey’s multiple comparisons post-hoc test, or by Kruskal-Wallis test followed by Dunn’s multiple comparisons test when data was not

The aim of this study was to explore (1) whether the iron chelators deferoxamine and deferiprone are able to inhibit Aβ 1-42 -induced Lcn2 production in

The main aim of this study was to characterise if the dynamics of NGAL in serial measures of plasma and CSF is reduced in older male volunteers by (i) investigating

For example, the effects of Lcn2 may depend significantly on the cell types by which it is expressed, the cell types that respond to it, the receptors via which it

To investigate whether increased Lcn2 levels may actually contribute to Alzheimer’s pathology, in chapter 3 we studied possible damaging or protective effects of

Therefore our objectives were (1) to investigate NGAL protein levels in serum, CSF and human post-mortem brain tissue (nine different brain regions) of control

Lipocalin 2 contributes to brain iron accumulation in J20 Alzheimer mice, and also plays a role in brain iron homeostasis under healthy conditions.. It is increasingly recognized