• No results found

University of Groningen Neuroinflammation as common denominator in heart failure associated mental dysfunction Gouweleeuw, Leonie

N/A
N/A
Protected

Academic year: 2021

Share "University of Groningen Neuroinflammation as common denominator in heart failure associated mental dysfunction Gouweleeuw, Leonie"

Copied!
15
0
0

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

Hele tekst

(1)

Neuroinflammation as common denominator in heart failure associated mental dysfunction

Gouweleeuw, Leonie

DOI:

10.33612/diss.122192415

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:

2020

Link to publication in University of Groningen/UMCG research database

Citation for published version (APA):

Gouweleeuw, L. (2020). Neuroinflammation as common denominator in heart failure associated mental

dysfunction: Studies in animal models. University of Groningen. https://doi.org/10.33612/diss.122192415

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 1

(3)

General introduction

Myocardial infarction and mental dysfunction - epidemics

Myocardial infarction (MI) and heart failure are associated with a number of different comorbidities, including renal disease, diabetes and hypertension. Another important comorbidity is mental dysfunction; often overlooked or regarded as ”natural response to a life-threatening condition”. Cardiac patients have a higher risk of developing depression, anxiety or cognitive impairment compared to the general population. Vice versa, patients with mental illness are more prone to MI and other forms of cardiac disease. The scope of this thesis, however, is focused on one side of this bilateral neurocardiac interaction; MI leading to mental dysfunction.

The percentage of MI patients suffering from depression varies widely, depending on the study population and the criteria by which depression was scored. Major depression was found in 15-22% of MI patients [1, 2], while a literature review reported symptoms of depression in up to 65% of MI patients [3]. Depression following myocardial infarction should not be considered as an innocent complication. Different studies have shown that depression following MI is associated with higher risk of comorbidities, hospitalizations and all-cause mortality [4, 5]. Unfortunately, treating the depression does not lead to an improved cardiac prognosis. In fact, there is evidence that in women the use of anti-depressant drugs and anxiolytics even worsens cardiac prognosis [6].

Similarly to depression, cognitive impairment is common in MI patients. A recent meta-analysis of 24 studies found an average odds ratio (OR) of 1.45 for cognitive impairment or dementia in patients with coronary heart disease, including MI patients [7]. Results of the separate studies did show high variability, with some studies showing no difference and others a high OR (ranging from 0.97 – 2.58) [8-10]. Studies also showed that OR was sex and age dependant, with higher dementia risk in the male sex and with increasing age [11, 12]. Cognitive impairment is associated with unfavourable disease outcome [13], and MI patients that also have cognitive impairment are more likely to develop dementia [14].

Treatment of mental dysfunction in patients with cardiovascular disease has proven to be difficult. One of the difficulties is that the pathophysiology of the interaction has not been fully elucidated.

(4)

Inflammation hypothesis

There are many hypotheses regarding the association between cardiac disease and decreased mental wellbeing. No doubt that psychological and behavioural factors can contribute to mental dysfunction in MI patients. However, many biological factors are proposed to contribute as well. MI evokes a strong neuroendocrine response in order to preserve cardiac function. This can give rise to platelet aggregation, endothelial dysfunction, hypoxia in the central nervous system (CNS) and autonomic nervous system dysfunction [15-17]. These phenomena have been linked to depression, anxiety and cognitive problems. Another major player in the field is inflammation. A schematic overview of mechanisms that contribute to the relationship between cardiovascular disease and depression is giving in Figure 1. Both cardiac disease and mental dysfunction have been linked to elevated levels of cytokines. Several of these cytokines overlap, occurring both in depression and in cardiac disease [18]. Rather than one pathology causing the other, the link between cardiac and mental dysfunction could hence be attributed to inflammation as common underlying factor. This would also explain why treating one of the diseases does not change the prognosis for the other.

Figure 1: Proposed mechanisms involved in the relationship between cardiovascular disease and mental dysfunction.

Inflammation, altered platelet function, endothelial dysfunction and autonomic nervous system dysfunction have been mentioned as involved in both pathologies and can influence and aggravate each other.

(5)

The inflammatory reaction following MI is a response to tissue damage, and is needed for proper scar formation [19, 20]. However, circulatory cytokines are raised to long after the healing phase following MI. Clinical studies have found specific inflammatory markers that were more elevated in MI patients with depression compared to MI patients without depression. Among these markers are tumour necrosis factor alpha (TNF-α), interleukin 6 (IL-6), C-reactive protein (CRP) and Neutrophil-gelatinase-associated lipocalin (NGAL) [21-24]. There are several proposed mechanism by which these factors could exert effects on the CNS, leading to behavioural changes. Either they could enter the CNS at locations not lined by the blood brain barrier, circumventricular organs, or at leaky spots in the blood brain barrier; they could be transported through active transport mechanisms over the blood brain barrier, or they can interact with afferent nerves to reflect the signal into the brain [25]. Moreover, it has been reported that circulatory cytokines can cause local increases in permeability of the blood brain barrier, facilitating the entry of cytokines in the CNS [26]. The latter has been especially well described in the case of TNF-α [27]. MI causes blood brain barrier leakage in rats that can be mimicked by TNF-α infusion [26, 28]. Mice showed blood brain barrier leakage for at least 3 months after MI [29].

Neutrophil gelatinase associated lipocalin

An interesting protein in the association between cardiac disease and mental dysfunction is Neutrophil gelatinase associated lipocalin (NGAL), also known as Lipocalin-2. NGAL is important in anti-microbial defence, primarily through sequestering iron, depleting iron levels needed for bacterial growth [30]. In recent years, NGAL was linked to various disease states and was found to have several pro- and anti-inflammatory actions. Increased levels of NGAL were found in patients with cardiac disease, as well as in animal models of cardiac disease [31-34]. In heart failure patients, higher NGAL levels were associated with unfavourable prognosis [35]. In the nervous system, NGAL expression is altered in relation to different pathologic states, including stroke, MS and Alzheimer’s disease [36-38]. We know that CNS NGAL is upregulated in response to peripheral inflammation, as it is increased upon administration of lipopolysaccharide [39, 40]. NGAL expression by neurons, astrocytes and microglia is upregulated upon stimulation with TNF-α in cell culture experiments, again indicating it responds to inflammatory stimuli [41].

A number of papers regarding NGAL from our group also studied the role of NGAL in the brain and its link with depression. NGAL was identified as a protein highly upregulated in neurons, astrocytes and microglia upon stimulation with TNF-α in cell culture experiments [42]. This indicates NGAL acts downstream of TNF-α and we know that TNF-α is elevated

(6)

in heart failure and that TNF-α administration is associated with depressive behaviour [43]. Circulatory NGAL was also found to be elevated in elderly patients with depression [44]. Additionally, NGAL levels measured in post-mortem brain tissue showed differences in expression between Alzheimer’s disease patients with and without depression [36]. Three of our previous studies also focused on the link between NGAL and depression in heart failure patients [24, 45, 46]. These clinical results provide a good foundation to build the experimental work in this thesis on.

In the 2014 publication [24], we measured NGAL and depression in a cohort of heart failure patients, along with cardiovascular parameters at baseline and at 12 months follow-up. Results indicated NGAL was significantly associated with signs depression, both in the self-reported Beck Depression Inventory (BDI), as well as the Hamilton Depression Rating Scale (HAMD) interview (Figure 2). These results were independent of other factors, including age, sex, severity of heart disease and parameters for renal disease. In the multivariate analysis, symptoms of depression according to the BDI were subcategorized into cognitive and somatic symptoms of depression, and NGAL was only statistically associated with total BDI score and BDI somatic score, but not BDI cognitive score. Furthermore, both NGAL levels and depression scores correlated with 6-minute walk test, but not left ventricular ejection fraction. This is an indication that NGAL and depression are both associated with the experienced burden of heart failure, rather than with the clinical measure of disease severity [24].

In 2015, we published another paper on the same cohort of patients [45]. This time, other markers of inflammation (TNF-α, sTNFR1, sTNFR2, IL-6, and hsCRP) were also measured and we investigated whether NGAL correlated with these other inflammatory markers, and whether the correlation between NGAL and somatic depression still held after correcting for these other factors. Results showed that NGAL correlated significantly with TNF-α, sTNF-R1, sTNF-R2, hsCRP and leukocytes. Results also showed that NGAL was significantly associated with somatic symptoms of depression, also after correction for these other inflammatory markers.

The publication from 2016 [46] followed this group of patients for a longer period of time (on average 6.1 years) and investigated inflammatory markers and depression and its association with all-cause mortality. Results showed that depression, NGAL and several other inflammatory markers were associated with all-cause mortality. This association remained after correcting for age, sex and comorbidities.

(7)

Figure 2: Correlations between NGAL and depressive symptoms in heart failure patients. BDI baseline r = 0.22,

p = 0.03; 12 months r = 0.391, p = 0.001; HAMD baseline r = 0.25, p = 0.02; 12 months r = 0.18, p = 0.12. Derived from Naudé et al., 2014 [24].

The clinical data presented in these papers shows some very interesting correlations between heart disease, depression and inflammation. To collect more evidence for the role of inflammation linking depressive symptoms to cardiovascular disease, we have set up several animal experiments described in the further chapters of this thesis.

Animal models of MI and neuroinflammation (cardioneural interaction)

Animal models of MI and heart failure are a valuable research tool, most research is carried out in mice and rats. Usually myocardial infarction is induced by ligation of the left coronary artery, leading to an arrest in blood flow to part of the myocardium, which is comparable to the damage that is caused in MI patients. Following the surgery, animals show elevated plasma cytokines, remodelling of the myocardium and scar formation.

Animal models of MI have also been used to study changes in the central nervous system. A publication from 1993 already shows heart failure leads to selectively increased metabolic activity in the paraventricular nucleus of the hypothalamus (PVN) and locus coeruleus in rats [47]. These specific brain areas are involved in tasks including the regulation of blood pressure and sympathetic activity, processes highly involved in MI and heart failure.

Besides increased metabolic activity, there is also evidence for local inflammation in the CNS. The levels of TNF-α were measured over a longer period of time [48]. The investigators found

(8)

elevated levels of TNF-α in the heart, plasma and hypothalamus for up to 9 weeks post-MI. TNF-α was also measured in the cortex, here TNF-α levels seemed unaltered, indicating localized rather than general responses.

More proof for neuroinflammation following MI was found in the form of local activation of microglia [49]. The activity score of microglia was increased in the PVN for up to 5 weeks after MI. Notably, this change was only found in the portion of the PVN adjacent to the third ventricle, as there was no change observed in the area of the PVN further away from the third ventricle, where the magnocellular cells are localized.

Neuroinflammatory changes are not confined to areas involved in cardiovascular activity. Rinaldi and colleagues also found changes in microglia and astrocyte morphology in the prefrontal cortex, thalamus and hippocampus [50]. This is interesting, since the prefrontal cortex and hippocampus are also linked to behaviour. Hippocampus is known to be important for cognition and mood [51, 52]. Prefrontal cortex is involved in emotions and decision making and has been linked to the cognitive symptoms of depression [53]. Apoptosis was also found in hippocampus and amygdala areas in rats that received MI [54]. Like the hippocampus and prefrontal cortex, the amygdala has been linked to depression, it is known to play a role in emotions and anxiety [55].

Animal models of MI and behaviour

Animal models have also been used to measure behavioural changes after MI. One of the early studies on behaviour in MI rats revealed that MI rats showed more anxious behaviour and less exploratory behaviour and mobility [56]. Rats also showed decreased social interaction. The combination of these behavioural changes matches the range of behavioural symptoms seen in MI patients. Others also found signs of depressive behaviour in rats [57, 58]. There is also a report showing that depressive like behaviour only occurs in a subset of MI rats [59]. Behavioural changes following MI in rats can be modified by medication, as Schoemaker et al. showed that effective cardiovascular drug treatment could prevent part of the behavioural changes associated with MI [60]. A later study found signs of depression in infarcted rats that could be prevented by treatment with the antidepressant drug sertraline [54]. Similarly, MI induced anhedonia in rats could be reversed by treatment with etanercept, a TNF-α blocker [58].

Experimental heart failure in mice also led to behavioural changes associated with decreased exploration and an impaired short term memory, though changes were mild [61]. Signs of

(9)

depression after MI were also found by Ito et al., where behavioural changes were linked to higher sympathoactivation and decreased expression of the sigma-1-receptor [62]. In a study by Hong and colleagues, MI in mice led to cognitive impairment that was associated with changes in β-amyloid metabolism and blood brain barrier leakage [29].

Aim of this thesis

In this thesis we investigated behavioural and inflammatory changes in experimental MI in mice and rats. Special attention was paid to microglia activation as measure for neuroinflammation, and the markers TNF-α and neutrophil-gelatinase-associated lipocalin (NGAL).

Both TNF-α and microglia activation were previously shown to be elevated in experimental MI. However, these studies mostly focused their attention on the PVN and the alterations in blood volume regulation and sympathoactivation, rather than on behavioural changes. TNF-α is an interesting candidate to study, since we know there is elevated expression following MI, and in patients with depression plasma TNF-α is often elevated, while in animal MI models, TNF-α in the brain is also elevated. It is hypothesized that the increased permeability of the blood brain barrier by TNF-α is partly responsible for neuroinflammation and behavioural effects following MI [27]. NGAL is interesting to us, since a study in our group found NGAL as highly upregulated following TNF-α stimulation in neurons, astrocytes and microglia [41]. We later showed plasma NGAL was associated with depressive symptoms in heart failure patients [24].

This thesis will describe changes in behaviour, TNF-α, microglia and NGAL following MI in animal models. The goal is to shed light on neuroinflammatory pathways that might be critically involved in the development of mental dysfunction following MI.

(10)

References

1. Frasure-Smith N, Lesperance F, Talajic M: Depression following myocardial infarction. Impact on 6-month survival. JAMA 1993, 270(15):1819-1825.

2. Hance M, Carney RM, Freedland KE, Skala J: Depression in patients with coronary heart disease. A 12-month follow-up. Gen Hosp Psychiatry 1996, 18(1):61-65.

3. Carney RM, Freedland KE, Sheline YI, Weiss ES: Depression and coronary heart disease: a review for cardiologists. Clin Cardiol 1997, 20(3):196-200.

4. Meijer A, Conradi HJ, Bos EH, Thombs BD, van Melle JP, de Jonge P: Prognostic association of depression following myocardial infarction with mortality and cardiovascular events: a meta-analysis of 25 years of research. Gen Hosp Psychiatry 2011, 33(3):203-216.

5. Sherwood A, Blumenthal JA, Trivedi R, Johnson KS, O’Connor CM, Adams KF,Jr, Dupree CS, Waugh RA, Bensimhon DR, Gaulden L, Christenson RH, Koch GG, Hinderliter AL: Relationship of depression to death or hospitalization in patients with heart failure. Arch Intern Med 2007, 167(4):367-373.

6. Krantz DS, Whittaker KS, Francis JL, Rutledge T, Johnson BD, Barrow G, McClure C, Sheps DS, York K, Cornell C, Bittner V, Vaccarino V, Eteiba W, Parashar S, Vido DA, Merz CN: Psychotropic medication use and risk of adverse cardiovascular events in women with suspected coronary artery disease: outcomes from the Women’s Ischemia Syndrome Evaluation (WISE) study. Heart 2009, 95(23):1901-1906.

7. Deckers K, Schievink SHJ, Rodriquez MMF, van Oostenbrugge RJ, van Boxtel MPJ, Verhey FRJ, Kohler S: Coronary heart disease and risk for cognitive impairment or dementia: Systematic review and meta-analysis. PLoS One 2017, 12(9):e0184244.

8. Aronson MK, Ooi WL, Morgenstern H, Hafner A, Masur D, Crystal H, Frishman WH, Fisher D, Katzman R: Women, myocardial infarction, and dementia in the very old. Neurology 1990, 40(7):1102-1106.

9. Hayden KM, Zandi PP, Lyketsos CG, Khachaturian AS, Bastian LA, Charoonruk G, Tschanz JT, Norton MC, Pieper CF, Munger RG, Breitner JC, Welsh-Bohmer KA, Cache County Investigators: Vascular risk factors for incident Alzheimer disease and vascular dementia: the Cache County study. Alzheimer Dis Assoc Disord 2006, 20(2):93-100.

10. Kalmijn S, Feskens EJ, Launer LJ, Kromhout D: Cerebrovascular disease, the apolipoprotein e4 allele, and cognitive decline in a community-based study of elderly men. Stroke 1996, 27(12):2230-2235.

11. Kivipelto M, Helkala EL, Laakso MP, Hanninen T, Hallikainen M, Alhainen K, Iivonen S, Mannermaa A, Tuomilehto J, Nissinen A, Soininen H: Apolipoprotein E epsilon4 allele, elevated midlife total cholesterol level, and high midlife systolic blood pressure are independent risk factors for late-life Alzheimer disease. Ann Intern Med 2002, 137(3):149-155.

12. Ikram MA, van Oijen M, de Jong FJ, Kors JA, Koudstaal PJ, Hofman A, Witteman JC, Breteler MM: Unrecognized myocardial infarction in relation to risk of dementia and cerebral small vessel disease. Stroke 2008, 39(5):1421-1426.

13. Zuccala G, Pedone C, Cesari M, Onder G, Pahor M, Marzetti E, Lo Monaco MR, Cocchi A, Carbonin P, Bernabei R: The effects of cognitive impairment on mortality among hospitalized patients with heart failure. Am J Med 2003, 115(2):97-103.

14. Gharacholou SM, Reid KJ, Arnold SV, Spertus J, Rich MW, Pellikka PA, Singh M, Holsinger T, Krumholz HM, Peterson ED, Alexander KP: Cognitive impairment and outcomes in older adult survivors of acute myocardial infarction: findings from the translational research investigating underlying disparities in acute myocardial infarction patients’ health status registry. Am Heart J 2011, 162(5):860-869.e1.

15. Pizzi C, Mancini S, Angeloni L, Fontana F, Manzoli L, Costa GM: Effects of selective serotonin reuptake inhibitor therapy on endothelial function and inflammatory markers in patients with coronary heart disease. Clin Pharmacol Ther 2009, 86(5):527-532.

16. Schins A, Hamulyak K, Scharpe S, Lousberg R, Van Melle J, Crijns H, Honig A: Whole blood serotonin and platelet activation in depressed post-myocardial infarction patients. Life Sci 2004, 76(6):637-650.

(11)

17. Kop WJ, Stein PK, Tracy RP, Barzilay JI, Schulz R, Gottdiener JS: Autonomic nervous system dysfunction and inflammation contribute to the increased cardiovascular mortality risk associated with depression. Psychosom Med 2010, 72(7):626-635.

18. Pasic J, Levy WC, Sullivan MD: Cytokines in depression and heart failure. Psychosom Med 2003, 65(2):181-193. 19. Frangogiannis NG: Inflammation in cardiac injury, repair and regeneration. Curr Opin Cardiol 2015,

30(3):240-245.

20. Frangogiannis NG, Smith CW, Entman ML: The inflammatory response in myocardial infarction. Cardiovasc Res 2002, 53(1):31-47.

21. Basaran Y, Basaran MM, Babacan KF, Ener B, Okay T, Gok H, Ozdemir M: Serum tumor necrosis factor levels in acute myocardial infarction and unstable angina pectoris. Angiology 1993, 44(4):332-337.

22. Ikeda U, Ohkawa F, Seino Y, Yamamoto K, Hidaka Y, Kasahara T, Kawai T, Shimada K: Serum interleukin 6 levels become elevated in acute myocardial infarction. J Mol Cell Cardiol 1992, 24(6):579-584.

23. Kushner I, Broder ML, Karp D: Control of the acute phase response. Serum C-reactive protein kinetics after acute myocardial infarction. J Clin Invest 1978, 61(2):235-242.

24. Naude PJ, Mommersteeg PM, Zijlstra WP, Gouweleeuw L, Kupper N, Eisel UL, Kop WJ, Schoemaker RG: Neutrophil Gelatinase-Associated Lipocalin and depression in patients with chronic heart failure. Brain Behav Immun 2014, 38:59-65.

25. Quan N: In-depth conversation: spectrum and kinetics of neuroimmune afferent pathways. Brain Behav Immun 2014, 40:1-8.

26. Van der Werf YD, De Jongste MJL, Ter Horst GJ: The immune system mediates blood–brain barrier damage: possible implications for pathophysiology of neuropsychiatric illnesses. Acta Neuropsychiat 1995, 7:114-121. 27. Liu H, Luiten PG, Eisel UL, Dejongste MJ, Schoemaker RG: Depression after myocardial infarction:

TNF-alpha-induced alterations of the blood-brain barrier and its putative therapeutic implications. Neurosci Biobehav Rev 2013, .

28. Ter Horst G, Nagel J, De Jongste M, Van der Werf Y: Selective Blood Brain Barrier Dysfunction After Intravenous Injections of RTNFα in the Rat. In Neurochemistry. Edited by Anonymous New York: Springer; 1997:141-146. 29. Hong X, Bu L, Wang Y, Xu J, Wu J, Huang Y, Liu J, Suo H, Yang L, Shi Y, Lou Y, Sun Z, Zhu G, Behnisch T, Yu M,

Jia J, Hai W, Meng H, Liang S, Huang F, Zou Y, Ge J: Increases in the risk of cognitive impairment and alterations of cerebral beta-amyloid metabolism in mouse model of heart failure. PLoS One 2013, 8(5):e63829. 30. Goetz DH, Holmes MA, Borregaard N, Bluhm ME, Raymond KN, Strong RK: The neutrophil lipocalin NGAL is a

bacteriostatic agent that interferes with siderophore-mediated iron acquisition. Mol Cell 2002, 10(5):1033-1043. 31. Damman K, van Veldhuisen DJ, Navis G, Voors AA, Hillege HL: Urinary neutrophil gelatinase associated lipocalin (NGAL), a marker of tubular damage, is increased in patients with chronic heart failure. Eur J Heart Fail 2008, 10(10):997-1000.

32. Shrestha K, Borowski AG, Troughton RW, Klein AL, Tang WH: Association between systemic neutrophil gelatinase-associated lipocalin and anemia, relative hypochromia, and inflammation in chronic systolic heart failure. Congest Heart Fail 2012, 18(5):239-244.

33. Sahinarslan A, Kocaman SA, Bas D, Akyel A, Ercin U, Zengin O, Timurkaynak T: Plasma neutrophil gelatinase-associated lipocalin levels in acute myocardial infarction and stable coronary artery disease. Coron Artery Dis 2011, 22(5):333-338.

34. Hemdahl AL, Gabrielsen A, Zhu C, Eriksson P, Hedin U, Kastrup J, Thoren P, Hansson GK: Expression of neutrophil gelatinase-associated lipocalin in atherosclerosis and myocardial infarction. Arterioscler Thromb Vasc Biol 2006, 26(1):136-142.

35. van Deursen VM, Damman K, Voors AA, van der Wal MH, Jaarsma T, van Veldhuisen DJ, Hillege HL: Prognostic value of plasma neutrophil gelatinase-associated lipocalin for mortality in patients with heart failure. Circ Heart Fail 2014, 7(1):35-42.

36. Dekens DW, Naude PJ, Engelborghs S, Vermeiren Y, Van Dam D, Oude Voshaar RC, Eisel UL, De Deyn PP: Neutrophil Gelatinase-Associated Lipocalin and its Receptors in Alzheimer’s Disease (AD) Brain Regions: Differential Findings in AD with and without Depression. J Alzheimers Dis 2017, 55(2):763-776.

(12)

37. Al Nimer F, Elliott C, Bergman J, Khademi M, Dring AM, Aeinehband S, Bergenheim T, Romme Christensen J, Sellebjerg F, Svenningsson A, Linington C, Olsson T, Piehl F: Lipocalin-2 is increased in progressive multiple sclerosis and inhibits remyelination. Neurol Neuroimmunol Neuroinflamm 2016, 3(1):e191.

38. Xing C, Wang X, Cheng C, Montaner J, Mandeville E, Leung W, van Leyen K, Lok J, Wang X, Lo EH: Neuronal production of lipocalin-2 as a help-me signal for glial activation. Stroke 2014, 45(7):2085-2092.

39. Flo TH, Smith KD, Sato S, Rodriguez DJ, Holmes MA, Strong RK, Akira S, Aderem A: Lipocalin 2 mediates an innate immune response to bacterial infection by sequestrating iron. Nature 2004, 432(7019):917-921. 40. Marques F, Rodrigues AJ, Sousa JC, Coppola G, Geschwind DH, Sousa N, Correia-Neves M, Palha JA: Lipocalin

2 is a choroid plexus acute-phase protein. J Cereb Blood Flow Metab 2008, 28(3):450-455.

41. Naude PJ, Nyakas C, Eiden LE, Ait-Ali D, van der Heide R, Engelborghs S, Luiten PG, De Deyn PP, den Boer JA, Eisel UL: Lipocalin 2: novel component of proinflammatory signaling in Alzheimer’s disease. FASEB J 2012, 26(7):2811-2823.

42. Naude PJ, Nyakas C, Eiden LE, Ait-Ali D, van der Heide R, Engelborghs S, Luiten PG, De Deyn PP, den Boer JA, Eisel UL: Lipocalin 2: novel component of proinflammatory signaling in Alzheimer’s disease. FASEB J 2012, 26(7):2811-2823.

43. van Heesch F, Prins J, Korte-Bouws GA, Westphal KG, Lemstra S, Olivier B, Kraneveld AD, Korte SM: Systemic tumor necrosis factor-alpha decreases brain stimulation reward and increases metabolites of serotonin and dopamine in the nucleus accumbens of mice. Behav Brain Res 2013, 253:191-195.

44. Naude PJ, Eisel UL, Comijs HC, Groenewold NA, De Deyn PP, Bosker FJ, Luiten PG, den Boer JA, Oude Voshaar RC: Neutrophil gelatinase-associated lipocalin: a novel inflammatory marker associated with late-life depression. J Psychosom Res 2013, 75(5):444-450.

45. Naude PJ, Mommersteeg PM, Gouweleeuw L, Eisel UL, Denollet J, Westerhuis LW, Schoemaker RG: NGAL and other markers of inflammation as competitive or complementary markers for depressive symptom dimensions in heart failure. World J Biol Psychiatry 2015, :1-6.

46. Mommersteeg PMC, Schoemaker RG, Naude PJW, Eisel ULM, Garrelds IM, Schalkwijk CG, Westerhuis BWJJM, Kop WJ, Denollet J: Depression and markers of inflammation as predictors of all-cause mortality in heart failure. Brain Behav Immun 2016, 57:144-150.

47. Patel KP, Zhang PL, Krukoff TL: Alterations in brain hexokinase activity associated with heart failure in rats. Am J Physiol 1993, 265(4 Pt 2):R923-8.

48. Francis J, Chu Y, Johnson AK, Weiss RM, Felder RB: Acute myocardial infarction induces hypothalamic cytokine synthesis. Am J Physiol Heart Circ Physiol 2004, 286(6):H2264-71.

49. Rana I, Stebbing M, Kompa A, Kelly DJ, Krum H, Badoer E: Microglia activation in the hypothalamic PVN following myocardial infarction. Brain Res 2010, 1326:96-104.

50. Rinaldi B, Guida F, Furiano A, Donniacuo M, Luongo L, Gritti G, Urbanek K, Messina G, Maione S, Rossi F, de Novellis V: Effect of Prolonged Moderate Exercise on the Changes of Nonneuronal Cells in Early Myocardial Infarction. Neural Plast 2015, 2015:265967.

51. Small SA, Schobel SA, Buxton RB, Witter MP, Barnes CA: A pathophysiological framework of hippocampal dysfunction in ageing and disease. Nat Rev Neurosci 2011, 12(10):585-601.

52. Femenia T, Gomez-Galan M, Lindskog M, Magara S: Dysfunctional hippocampal activity affects emotion and cognition in mood disorders. Brain Res 2012, 1476:58-70.

53. Krishnan V, Nestler EJ: The molecular neurobiology of depression. Nature 2008, 455(7215):894-902. 54. Wann BP, Bah TM, Kaloustian S, Boucher M, Dufort AM, Le Marec N, Godbout R, Rousseau G: Behavioural

signs of depression and apoptosis in the limbic system following myocardial infarction: effects of sertraline. J Psychopharmacol 2009, 23(4):451-459.

55. Beauregard M, Paquette V, Levesque J: Dysfunction in the neural circuitry of emotional self-regulation in major depressive disorder. Neuroreport 2006, 17(8):843-846.

56. Schoemaker RG, Smits JF: Behavioral changes following chronic myocardial infarction in rats. Physiol Behav 1994, 56(3):585-589.

(13)

57. Bah TM, Benderdour M, Kaloustian S, Karam R, Rousseau G, Godbout R: Escitalopram reduces circulating pro-inflammatory cytokines and improves depressive behavior without affecting sleep in a rat model of post-cardiac infarct depression. Behav Brain Res 2011, 225(1):243-251.

58. Grippo AJ, Francis J, Weiss RM, Felder RB, Johnson AK: Cytokine mediation of experimental heart failure-induced anhedonia. Am J Physiol Regul Integr Comp Physiol 2003, 284(3):R666-73.

59. Wang Y, Liu X, Zhang D, Chen J, Liu S, Berk M: The effects of apoptosis vulnerability markers on the myocardium in depression after myocardial infarction. BMC Med 2013, 11(1):32.

60. Schoemaker RG, Kalkman EA, Smits JF: ‘Quality of life’ after therapy in rats with myocardial infarction: dissociation between hemodynamic and behavioral improvement. Eur J Pharmacol 1996, 298(1):17-25. 61. Frey A, Popp S, Post A, Langer S, Lehmann M, Hofmann U, Siren AL, Hommers L, Schmitt A, Strekalova T, Ertl

G, Lesch KP, Frantz S: Experimental heart failure causes depression-like behavior together with differential regulation of inflammatory and structural genes in the brain. Front Behav Neurosci 2014, 8:376.

62. Ito K, Hirooka Y, Sunagawa K: Brain sigma-1 receptor stimulation improves mental disorder and cardiac function in mice with myocardial infarction. J Cardiovasc Pharmacol 2013, 62(2):222-228.

(14)
(15)

Referenties

GERELATEERDE DOCUMENTEN

It turned out that these feedback invariants have significance not only for problems not directly related to feedback but also for realization theory, in

Verder wordt het vlak nog doorsneden door twee evenwijdige noordwest-zuidoost verlopende perceelsgrachten die opnieuw een bruine kleivulling kennen.. 17 Bij zone 3 is

Neuroinflammation as common denominator in heart failure associated mental dysfunction Studies in animal models. The studies described in this thesis were carried out at

It is reported that patients with cardiovascular disease, including heart failure and acute myocardial infarction (AMI), have an increased risk of developing depression.. On the

Neuroinflammation in different brain areas was measured as microglia activation and TNF-α expression, while neurogenesis was obtained from DCX staining of immature neurons in the

This indicates that microglia could indeed be a mediator in myocardial infarction or surgery related behavioural changes, including anxiety and depression and that the

Indeed in our study plasma NGAL levels in male rats were positively correlated to infarct size and parameters of heart failure.. One reason that we failed to find this effect

For each experiment plasma and hippocampal NGAL concentrations, hippocampal cell body to cell size ratios, and the AUC of the spatial learning paradigm were expressed as percentage