Skip to main content
Erschienen in: Alzheimer's Research & Therapy 1/2015

Open Access 01.12.2015 | Review

‘Hearts and minds’: association, causation and implication of cognitive impairment in heart failure

verfasst von: Jane A Cannon, John JV McMurray, Terry J Quinn

Erschienen in: Alzheimer's Research & Therapy | Ausgabe 1/2015

Abstract

The clinical syndrome of heart failure is one of the leading causes of hospitalisation and mortality in older adults. An association between cognitive impairment and heart failure is well described but our understanding of the relationship between the two conditions remains limited. In this review we provide a synthesis of available evidence, focussing on epidemiology, the potential pathogenesis, and treatment implications of cognitive decline in heart failure. Most evidence available relates to heart failure with reduced ejection fraction and the syndromes of chronic cognitive decline or dementia. These conditions are only part of a complex heart failure-cognition paradigm. Associations between cognition and heart failure with preserved ejection fraction and between acute delirium and heart failure also seem evident and where data are available we will discuss these syndromes. Many questions remain unanswered regarding heart failure and cognition. Much of the observational evidence on the association is confounded by study design, comorbidity and insensitive cognitive assessment tools. If a causal link exists, there are several potential pathophysiological explanations. Plausible underlying mechanisms relating to cerebral hypoperfusion or occult cerebrovascular disease have been described and it seems likely that these may coexist and exert synergistic effects. Despite the prevalence of the two conditions, when cognitive impairment coexists with heart failure there is no specific guidance on treatment. Institution of evidence-based heart failure therapies that reduce mortality and hospitalisations seems intuitive and there is no signal that these interventions have an adverse effect on cognition. However, cognitive impairment will present a further barrier to the often complex medication self-management that is required in contemporary heart failure treatment.
Hinweise
This is an Open Access article distributed under the terms of the Creative Commons Attribution License (http://​creativecommons.​org/​licenses/​by/​4.​0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated.

Competing interests

JC has no competing interests. JJVM has no competing interests. TJQ has received modest honoraria, research funding and travel support from: Astra-Zeneca; Bayer; Boehringer Eingelheim; Bristol Meyers Squibb; Merck; Pfizer. He holds grants relating to cognitive assessment from British Geriatric Society; Chest Heart and Stroke Scotland; Chief Scientists Office Scotland; The Stroke Association.
Abkürzungen
ACE-(i)
Angiotensin converting enzyme-(inhibitor)
AD
Alzheimer’s disease
AF
Atrial fibrillation
CAD
Coronary artery disease
CI
Cognitive impairment
EF
Ejection fraction
HF
Heart failure
HF-PEF
Heart failure-preserved ejection fraction
HF-REF
Heart failure-reduced ejection fraction
RAAS
Renin-angiotensin-aldosterone system.

Definitions and burden of heart failure

The term 'heart failure' (HF) is used to describe a condition wherein cardiac output is insufficient to meet metabolic requirements [1]. Clinically, it is defined as a syndrome where patients have typical signs and symptoms resulting from an abnormality of cardiac structure or function [2]. Contemporary terminology used to describe HF is based on left ventricular ejection fraction (EF). This is considered important not only because of prognosis (the lower the EF the poorer the survival) but also because the major trials that inform the evidence base have almost exclusively focussed on patients who have HF with reduced ejection fraction (HF-REF) [2]. A subgroup of patients also present with classical signs and symptoms but in the context of preserved ejection fraction (HF-PEF). These patients often have evidence of diastolic dysfunction and this is considered by many as the cause of HF symptoms.
It is estimated that 1 to 2% of the adult population in developed countries have HF with the prevalence increasing to ≥10% among patients aged over 70 years; more than half of these patients have HF-REF [3]. The most common underlying aetiology in HF-REF is coronary artery disease (CAD) resulting in myocardial damage. Other common causes include hypertension, valvular pathology, viral infection and alcohol excess [2]. HF-PEF is more common in older, female patients. It is less frequently due to CAD and more often linked to hypertension and atrial fibrillation (AF), with the diagnosis being one of exclusion of other non-cardiac causes of breathlessness [2].
HF admissions account for 5% of all medical admissions (making it the commonest cause of unscheduled admission in older adults) and 2% of the total UK National Health Service budget [4]. Societal and demographic changes, including aging of the general population and improved survival from CAD, will increase HF prevalence (Figure 1) with a potential doubling in HF prevalence within the next 40 years [2].

Heart failure and cognitive impairment – strength of association

The co-existence of symptomatic 'heart failure' and 'brain failure' has been recognised for decades, with a description of 'cardiogenic dementia' first introduced in the 1970s. While the co-occurrence of HF and cognitive problems will be familiar to most clinicians, the topic has received relatively little research interest compared with other aspects of cardiac disease. In collating and offering a synthesis of the available literature describing the association of HF and cognition, we have found a disparate and inconsistent literature, characterised by small sample sizes, heterogeneity and multiple potential biases. We provide a brief narrative overview of the field and have tabulated a more detailed summary of findings from available cross-sectional and prospective studies (Tables 1 to 3).
Table 1
Studies examining the prevalence of cognitive impairment in patients with heart failure
Study
Sample
Population
Median age in years (SD)
Study methodology
Inclusion criteria
Exclusion criteria
CV measures/criteria
Cognitive assessment tool(s) used
Results
Zuccalà 1997 [6]
57 HF pts
Consecutive admissions to hospital
77
Cross-sectional
Not specified
Co-morbid psychiatric or physical illness and previous diagnosis of CI
LVEF (mean EF 45%)
NYHA II-III
MMSE, MDBandRCPM
53% of HF pts showed global CI with MMSE less than 24
Callegari 2002 [7]
64 HF pts, 321 healthy controls
Age <65 years and consecutive admissions to hospital
52 (8)
Cross-sectional
Not specified
Co-morbid psychiatric or neurological illness. Previous diagnosis of CI and female sex
LVEF <50%
Multidomain neuropsychiatric battery
HF pts scored lower than control group in short-term verbal memory, short-term visuospatial memory and visual spatial logical ability
NYHA I-III
Cardiopulmonary testing with treadmill
Right heart catheterisation
Trojano 2003 [8]
149 HF NYHA II pts
Age >65 years and consecutive admissions to hospital
HF NYHA II: 75 (7)
Cross-sectional
Not specified
Co-morbid psychiatric, neurological or physical illness. Previous diagnosis of CI
No measure of LV function
Multidomain neuropsychiatric battery
HF pts scored worse than those without HF in domains of: attention, verbal fluency, verbal learning
159 HF NYHA III/IV
HF NYHA III/IV: 77 (7)
NYHA II-IV
No significant difference between pts with moderate or severe HF
207 non-HF controls
Non-HF controls: 74 (7)
 
Zuccalà 2005 [9]
1,511 HF pts, 11,790 control patients
All geriatric or general medical hospital admissions
79 (9)
Cross-sectional
Not specified
Not specified
HF diagnosis based on discharge documentation
Hodkinson abbreviated mental test
35% of HF pts showed global CI
29% of non-HF pts showed global CI
Feola 2007 [10]
60 HF-REF
Consecutive admissions to hospital
66
Cross-sectional
HF-REF: clinical HF, NYHA II-IV, LVEF ≤50%
Not specified
LVEF
Multidomain neuropsychiatric battery
23% of HF pts showed global CI
12 HF-PEF
HF-PEF: diagnosed based on E/A ratio, deceleration time and LV dilatation
NYHA II-IV
BNP
Debette 2007 [11]
83 HF pts
Consecutive admissions to hospital
62
Cross-sectional
Not specified
Hearing/visual impairment
LVEF <45%
MMSE
61% of HF pts showed global CI
NYHA I-IV
Dodson 2013 [12]
282 decompensated HF pts
Age >65 years and non-consecutive admissions to hospital
80 (8)
Prospective
English speaking
Co-morbid psychiatric illness
HF diagnosis based on documentation in medical records
MMSE
25% of HF pts showed evidence of mild CI 22% of HF pts showed moderate to severe CI
Schmidt 1991 [13]
20 iDCM pts
Age <50 years and ambulatory outpatients only
iDCM: 38 (5)
Cross-sectional
Not specified
Co-morbid psychiatric, neurological or physical illness
LVEF 14-45%
LGT-3 and ALID
Systolic HF pts performed worse than the control group in domains of attention, learning and memory and reaction time
20 healthy controls
Healthy controls: 41 (8)
NYHA II-IV
Grubb 2000 [14]
20 HF pts with CADs
Ambulatory outpatients only
HF: 68
Cross-sectional
Not specified
Co-morbid psychiatric or neurological illness. Previous hospital admission within 6 months
HF: LVEF <40%, NYHA III/IV
RBMT and WMS
No difference between HF pts and control group
20 CAD control
CAD controls: 67
CAD controls: LVEF >55%, no CHF
Riegel 2002 [15]
42 HF pts
Ambulatory outpatients only
75 (12)
Cross-sectional
English speaking
Co-morbid physical or psychiatric illness
No measure of LV function
MMSE and CIMS
29% of HF pts showed evidence of global CI
NYHA I-IV
Vogels 2007 [16]
62 HF pts
Age >50 years and ambulatory outpatients only
HF: 69 (9))
Case control
HF pts: diagnosis of HF >6 months and stable on medication >4 weeks
Co-morbid physical, neurological or psychiatric illness. Previous diagnosis of CI
LVEF <40%
Multidomain neuropsychiatric battery
HF pts scored lower than the healthy control group in all domains
53 CAD controls
CAD controls: 69 (10)
CAD controls: IHD but no clinical CHF and EF >40%
NYHA II-IV
HF pts scored lower than the IHD control group in domains of memory and mental speed
42 healthy controls
Healthy controls: 67 (9
IHD control group scored lower than the healthy control group in language only
Hoth 2008 [17]
31 HF pts
Age >55 years and ambulatory outpatients only
HF: 69 (9)
Cross-sectional
English speaking
Co-morbid physical, neurological or psychiatric illness. Previous diagnosis of CI
LVEF <40%
Multidomain neuropsychiatric battery
Systolic HF pts scored lower than the IHD control group in domains of executive function and cognitive flexibility
31 CAD controls
CAD controls: 69 (9)
Minimum of 8th grade education
NYHA II-IV
CAD controls: angina/previous MI/PCI/PVD and HF excluded on basis of clinical examination
Beer 2009 [18]
31 HF pts
Ambulatory outpatients only
HF: 54 (11)
Case control
Not specified
Co-morbid neurological illness or previous diagnosis of CI
LVEF <40%
Block design, CVLT and 'F,A,S test'
Systolic HF pts scored lower than control group in all cognitive domains
24 healthy controls
Healthy controls: 56 (8)
NYHA II
LWHFQ
Stanek 2009 [19]
40 HF pts, 35 CAD controls
Ambulatory outpatients only
70 (8)
Prospective
English speaking
Co-morbid psychiatric or neurological illness. Previous diagnosis of CI
NYHA II-III
DRS
No difference between systolic HF pts and CAD control patients in all domains
CAD controls: history of MI, CAD, cardiac surgery, hypertension
CO <4 L/minute on echo
Sauvé 2009 [20]
50 HF pts
50 healthy controls
Age >30 years in HF pts and >55 years in controls. Ambulatory outpatients only
HF: 63 (14)
Healthy controls: 63 (14)
Case control
Diagnosis of HF >6 months
Co-morbid psychiatric or neurological illness
LVEF ≤40%
NYHA II-IV
Multidomain neuropsychiatric battery
Systolic HF pts scored lower than control group in domain of verbal memory
Pressler 2010 [21]
249 HF pts
Ambulatory outpatients only
HF: 63 (15)
Cross-sectional
HF: LVEF ≤40% and clinical HF
Co-morbid psychiatric, neurological or physical illness. Previous diagnosis of CI
NYHA
Multidomain neuropsychiatric battery
HF group performed worse than healthy and general medical groups in domains of memory, executive function and psychomotor speed
63 healthy controls
Healthy controls: 53 (17)
Healthy controls: absence of any medical condition or controlled CV risk factors
LVEF
102 general medical pts
Medical group: 63 (12)
Medical group: major chronic disorder other than HF
Bauer 2012 [22]
51 HF-REF, 29 HF-PEF
Age >21 years and ambulatory outpatients only
72 (12)
Cross-sectional
HF-REF: history of HF-REF >6 months, stable on medication >4 weeks, LVEF ≤40%
Co-morbid psychiatric, neurological or physical illness. Previous diagnosis of CI
LVEF
Multidomain neuropsychiatric battery
HF-REF and HF-PEF pts performed worse than age- and educated-adjusted healthy control groups in executive function, attention, language, memory and psychomotor speed
HF-PEF: history of HF-PEF >6 months, stable on medication >4 weeks, LVEF >41%
NYHA
Festa 2011 [23]
169 HF-REF, 38 HF-PEF
Age >17 years and ambulatory outpatients only
69
Retrospective
On medical treatment for HF
Co-morbid neurological illness
LVEF
Multidomain neuropsychiatric battery
Low EF was associated with poor memory in pts over 63 years old
Haemodynamically stable
Pts <63 years old had preserved memory function regardless of EF.
Not receiving mechanical circulatory support
Steinberg 2011 [24]
55 HF pts
Ambulatory outpatients only
55 (8)
Cross-sectional
Stable clinical status
Co-morbid neurological or physical illness. Previous diagnosis of CI
LVEF ≤45%
Multidomain neuropsychiatric battery
44% of HF pts showed evidence of global CI
NYHA I-III
6 minute walk test
Jefferson 2011 [25]
1,114 pts from Framingham Heart Study
Age >40 and <89 years and ambulatory outpatients only
67 (9)
Cross-sectional
Not specified
Co-morbid neurological illness or previous diagnosis of CI
LVEF
Multidomain neuropsychiatric battery
U-shaped association between LVEF and cognitive performance
Cardiac MRI
Miller 2012 [26]
140 HF pts
Age >50 and <85 years and ambulatory outpatients only
69 (9)
Cross-sectional
English speaking
Co-morbid psychiatric or neurological illness
No measure of LV function
Multidomain neuropsychiatric battery
62% of HF pts showed evidence of global CI
No NYHA classification
2 minute step test
Almeida 2012 [27]
35 HF pts
Age >45 years and ambulatory outpatients only
HF: 69 (9)
Cross-sectional
HF: EF <40%, clinical HF ≥6 months, English speaking, NYHA I-III
Co-morbid psychiatric, neurological or physical illness. Previous diagnosis of CI
LVEF
Multidomain neuropsychiatric battery
HF pts scored lower than the healthy control group in domains of immediate/long-term memory and psychomotor speed
56 CAD controls
CAD controls: 67 (10)
CAD controls: previous MI, English speaking, EF ≥60%, no clinical HF
NYHA
No difference between the HF group and IHD control group in cognition
64 healthy controls
Healthy controls: 69 (11)
Healthy controls: English speaking, no previous MI/angina, EF ≥60%
 
Hawkins 2012 [28]
251 HF pts
Ambulatory outpatients only
66 (10)
Cross-sectional
English speaking
Co-morbid psychiatric illness. Previous diagnosis of CI
LVEF ≤40%
Multidomain neuropsychiatric battery
58% of HF pts had CI with poor scores in the domains of verbal learning and verbal memory
Bratzke-Bauer 2013 [29]
47 HF-REF
Age >50 years and ambulatory outpatients only
HF-REF: 75 (9)
Cross-sectional
History of HF >6 months
Co-morbid psychiatric, neurological or physical illness. Previous diagnosis of CI
LVEF
Multidomain neuropsychiatric battery
23% of the HF-REF cohort showed evidence of CI
33 HF-PEF
HF-PEF: 68 (15)
Stable on medication ≥4 weeks
NYHA
3% of the HF-PEF cohort showed evidence of CI
HF-PEF based on AHA criteria
Huijts 2013 [30]
491 HF-REF
Age >60 years and ambulatory outpatients only
77 (8)
Prospective
HF-REF: hospitalization within past year
Co-morbid physical illness
HF-REF: LVEF <45%, NYHA II-IV, NT-proBNP >400 pg/ml
AMT
8% of HF-REF group showed evidence of severe CI (AMT ≤7)
120 HF-PEF
HF-PEF: NT-proBNP ≥400 pg/ml if pt <75 years or ≥800 pg/ml if pt ≥75 years
HF-PEF: LVEF ≥45%
13% of HF-PEF group showed evidence of severe CI (AMT ≤7)
Kindermann 2012 [31]
20 decompensated HF pts
Decompensated HF: non-consecutive admissions to hospital
Decompensated HF: 60 (16)
Prospective
Decompensated HF: caused by ischaemic or DCM, symptomatic HF for ≥6 months, clinical signs of decompensation, for example, raised JVP
Co-morbid psychiatric, neurological or physical illness. Previous diagnosis of CI
LVEF <45%
Multidomain neuropsychiatric battery
Decompensated HF group scored lower than stable HF group in domains of memory, executive control and processing speed
20 stable HF pts
Stable HF: outpatients
Stable HF: 61 (17)
Stable HF pts: CHF of ischaemic or DCM, NYHA III-IV, no clinical signs/history of decompensation for ≥3 months
NYHA III/IV
Stable HF group scored lower than the healthy control group in domains of intelligence and episodic memory
20 healthy controls
Healthy controls: 62 (15)
AHA, American Heart Association; ALID, adjective list of Janke and Debus; AMT, Abbreviated Mental Test; BNP, brain natriuretic peptide; CAD, coronary artery disease; CHF, congestive heart failure; CI, cognitive impairment; CIMS, complex ideational material subset; CO, cardiac output; CV, cardiovascular; CVLT, California Verbal Learning Test; DCM, dilated cardiomyopathy; DRS, Disability Rating Scale; E/A ratio, ratio of mitral peak velocity of early filling (E) to mitral peak velocity of late filling (A); EF, ejection fraction; HF, heart failure; HF-REF, heart failure-reduced ejection fraction; HF-PEF, heart failure-preserved ejection fraction; iDCM, idiopathic dilated cardiomyopathy; IHD, ischaemic heart disease; JVP, jugular venous pressure; LGT-3, Lern und Gedachtnistest; LV, left ventricular; LVEF, left ventricular ejection fraction; LWHFQ, Living With Heart Failure Questionnaire; MDB, mental deterioration battery; MI, myocardial infarction; MMSE, Mini-Mental State Examination; MRI, magnetic resonance imaging; NT-pro BNP, N-terminal prohormone brain natriuretic peptide; NYHA, New York Heart Association; pts, patients; PCI, percutaneous coronary intervention; PVD, peripheral vascular disease; RBMT, Rivermead Behavioural Memory Test; RCPM, raven coloured progressive matrices; SD, standard deviation; WMS, Weschler Memory Scale.
Table 2
Studies examining cognitive changes over time in the heart failure population
Study
Sample
Population
Median age in years (SD)
Study methodology
Inclusion criteria
Exclusion criteria
CV measures
Cognitive assessment tool used
Follow-up period
Results
Karlsson 2005 [32]
146 CHF pts
Age >60 years and outpatients
76 (8)
Prospective
EF <45%
Co-morbid psychiatric, neurological or physical illness. Previous diagnosis of CI
LVEF
MMSE
6 months
12% of HF patients had MMSE scores <24 at baseline
NYHA II-IV
NYHA
And 4% had MMSE scores <24 at 6 months
Tanne 2005 [33]
20 CHF underwent exercise programme
5 CHF pts as control pts
Outpatients
63 (13)
Prospective
EF ≤35%
Co-morbid psychiatric, neurological or physical illness
LVEF
Multidomain neuropsychiatric battery
18 weeks
Improvement in executive function post-exercise programme
NYHA III
NYHA
History of HF for ≥6 months
Mod-Bruce ETT
No change in cognition in control group with time
Stable on medication ≥6 weeks
6 minute walk test
Stanek 2009 [19]
40 HF pts, 35 CAD controls
Age >53 and <84 years. Outpatients
70 (8)
Prospective
HF: English speaking
Co-morbid psychiatric or neurological illness. Previous diagnosis of CI
NYHA
DRS
12 months
HF patients improved at 12 months, particularly in attention
NYHA II or III
CO <4 L/minute
CO
Cardiac controls stable at 12 months
CAD controls: CO ≥4 L/minute, history of MI/CAD
Almeida 2013 [34]
77 HF pts
Age >45 years and outpatients
HF: 68 (10)
Prospective
HF: EF <40%, English speaking
Co-morbid psychiatric or neurological illness. Previous diagnosis of CI
NYHA
Multidomain neuropsychiatric battery
2 years
CHF group showed cognitive decline compared with CAD and healthy controls
73 CAD controls
CAD controls: 68 (10)
CAD controls: previous MI and EF >60%, English speaking
LVEF
81 healthy controls
Healthy controls: 69 (11)
Healthy controls: no history of CAD, English speaking
6 minute walk test
Hjelm 2011 [35]
95 HF pts
607 non-CHF controls
Age >80 years and outpatients
84 (3)
Prospective
Not specified
Not specified
HF diagnosis based on documentation in medical records
Multidomain neuropsychiatric battery
10 years
HF patients showed significant decline in episodic memory and spatial performance compared with controls
Riegel 2012 [36]
279 consecutive HF pts (HF-REF and HF-PEF)
Age <80 years and outpatients
62 (12)
Prospective
Stage C HF and English speaking
Co-morbid psychiatric or physical illness. Previous diagnosis of CI
NYHA I-IV
Multidomain neuropsychiatric battery
6 months
No significant change in cognition over 6 months (HF-REF and HF-PEF)
LVEF
Minimal improvement in DSST in both groups (likely due to learned effect)
Higher LVEF associated with lower DSST score
Huijts 2013 [30]
491 HF-REF
120 HF-PEF
Age >60 years and outpatients
77 (8)
Prospective
HF-REF: hospitalization within past year
Co-morbid physical illness
HF-REF: LVEF <45%, NYHA II-IV, NT-proBNP >400 pg/ml
AMT
18 months
HF-REF: 23% of HF pts showed decline of ≥1 point in AMT over 18 months
HF-PEF: NT-proBNP ≥400 pg/ml if pt <75 years or ≥800 pg/ml if pt ≥75 years
120 HF-PEF: LVEF ≥45%
HF-PEF: 24% of HF pts showed improvement of ≥1 point in AMT over 18 months
AMT, Abbreviated Mental Test; CAD, coronary artery disease; CHF, congestive heart failure; CI, cognitive impairment; CO, cardiac output; CV, cardiovascular; DRS, Disability Rating Scale; DSST, digit symbol substitution test; EF, ejection fraction; ETT, exercise tolerance test; HF, heart failure; HF-REF, heart failure-reduced ejection fraction; HF-PEF, heart failure-preserved ejection fraction; LVEF, left ventricular ejection fraction; MI, myocardial infarction; MMSE, Mini-Mental State Examination; NT-pro BNP, N-terminal prohormone brain natriuretic peptide; NYHA, New York Heart Association; pts, patients; SD, standard deviation.
Table 3
Studies examining the relationship between cognitive impairment and outcomes in patients with heart failure
Study
Sample
Population
Median age in years (SD)
Study methodology
Inclusion criteria
Exclusion criteria
Measures
Results
Zuccalà 2003 [37]
1511 HF pts 11,790 controls
All geriatric or general medical admissions
79 (9)
Prospective
Not specified
Not specified
Hodkinson abbreviated mental test
Mean length of hospital stay: pts with CI = 15 ± 10 days; pts without CI = 15 ± 9 days
Length of hospital stay
Inpatient mortality: pts with CI, 18%; pts without CI, 3%
1 year mortality
1-year mortality: pts with CI, 27%; pts without CI, 15%
Karlsson 2005 [32]
146 CHF pts
Age >60 years and outpatients
76 (8)
Prospective
LVEF <45%
Co-morbid psychiatric, neurological or physical illness. Previous diagnosis of CI
HF self-care
Self-care scores were significantly higher in those with MMSE >24 compared to those ≤24
NYHA II–IV
questionnaire
MMSE
Riegel 2007 [38]
29 CHF pts
Outpatients
64 (10)
Cross-sectional
LVSD on echo
Co-morbid psychiatric or physical illness. Previous diagnosis of CI
Self-care of HF index
CI was worse in the poor self-care group compared to the good and expert self-care groups but did not reach level of significance
Clinical HF
DSST
English speaking
Probed memory recall
Cameron 2009 [39]
50 CHF pts
Age >45 years and consecutive hospital admissions
73 (11)
Cross-sectional
Clinical CHF
Co-morbid neurological illness. Previous diagnosis of CI
Self-care of HF index
CI was not a predictor of self-care
LVSD on echo
Cardiac depression scale
English speaking
MMSE
Cameron 2010 [40]
93 CHF pts
Age >45 years and consecutive hospital admissions
73 (11)
Cross-sectional
Clinical CHF
Co-morbid neurological illness. Previous diagnosis of CI
Self-care HF index
CI and self-care management were significantly associated (t = 2.7; P < 0.01)
LVSD on echo
MMSE
English speaking
MoCA
Pulignano 2010 [41]
93 CHF pts
Consecutive outpatients
77 (6)
Cross-sectional
Not specified
Not specified
The European heart failure self-care behaviour scale
MMSE was negatively correlated with self-care behavioural scores (r = 0.58, P < 0.001)
MMSE
Alosco 2013 [42]
110 CHF pts
Age >50 years and <85 years. Outpatients
70 (9)
Prospective
NYHA II-IV
Co-morbid psychiatric, neurological or physical illness. Previous diagnosis of CI
Lawton-Brody instrumental activities of daily living
Poorer performance on 3MS was associated with worse total activities of daily living performance
English speaking
Modified MMSE (3MS)
Harkness 2013 [43]
100 CHF pts
Age >55 years and outpatients
72 (10)
Cross-sectional
Confirmed HF using the Boston criteria
Co-morbid psychiatric illness or previous diagnosis of CI
MoCA
MoCA score of <26 was significantly associated with worse self-care management
LVEF ≤45%
Self-care in HF index
Change in symptoms on previous 3 months
Geriatric Depression Scale
English speaking
Alosco 2013 [42]
175 CHF pts
Age >50 years and <85 years. Outpatients
68 (10)
Cross-sectional
NYHA II-IV
Co-morbid psychiatric, neurological or physical illness. Previous diagnosis of CI
Lawton-Brody instrumental activities of daily living
Poorer executive function was independently associated with poorer total activities of daily living performance
English speaking
Executive function assessed by FAB and LNS
CHF, congestive heart failure; CI, cognitive impairment; DSST, digit symbol substitution test; FAB, frontal assessment battery; HF, heart failure; LNS, letter number sequencing; LVEF, left ventricular ejection fraction; LVSD, left ventricular systolic dysfunction; MMSE, Mini-Mental State Examination; MoCA, Montreal Cognitive Assessment Tool; NYHA, New York Heart Association; SD, standard deviation.
Studies describing cognitive impairment (CI) in HF-REF have estimated prevalence at anywhere between 30 and 80% of patients (Table 1). This heterogeneity results from differences in study designs, case mix and cognitive assessments employed. Accepting the limitations of the evidence, even at the more conservative estimates of prevalence, the literature would suggest that CI frequently co-exists with HF-REF (Table 1).
Cross-sectional studies of cognition in HF have value in quantifying the burden of prevalent disease but give no clues as to temporal relationship or causation. To describe the incidence and 'natural history' of cognition in HF ideally requires prospective follow-up of a cohort free from CI at inception. Few studies have utilised this design and, where data are available, the validity is limited by small sample sizes, limited follow-up with substantial attrition and use of cognitive assessment tools that may not be sensitive to modest but clinically meaningful change (Table 2). Inherent in this study design is the assumption that CI follows or is a consequence of the HF pathology [16]. A literature around 'reverse causation' in heart disease has been described. In brief, early studies describing association of psychological or 'personality' factors and heart disease assumed that the neuropsychological traits pre-dated and were probably causative in the development of the cardiac condition. Subsequent data have questioned this temporality and suggest that subclinical (undiagnosed) vascular disease may cause psychological distress phenotypes [44]. Such arguments may also hold for HF and neuropsychological disease, where both cognitive change and psychological distress may be the cause or effect of HF. Investigating reverse causation is challenging but possible; to avoid biases from early mortality, large datasets with sufficient prospective follow-up are required [44].
Association does not imply causation and we must be mindful that both HF and CI are diseases of older age with many shared pathologies. Recognising this, many HF studies have defined an age-related inclusion criterion. With all the caveats that come with the heterogeneity of the available data, it would seem that association of CI and HF is present at all ages (Table 1). Studies that have attempted more sophisticated adjustment for confounders illustrate the inherent difficulty in teasing out what is contributory to cognitive decline and what is association or epi-phenomenon. In general, HF patients tend to have poorer scores on cognitive tests when compared with a 'healthy' (no cardiac disease) control group [34], but this comparator is still potentially confounded by cardiovascular comorbidity in the HF group. Inclusion of a cohort with common vascular risk factors but no HF may allow determination of whether HF per se is associated with CI. Where attempts have been made to utilise this design, studies have been modest in size and results contradictory [16,19]. Some authors have described little difference between groups and others have described increased rates of CI in HF-REF groups, particularly in 'executive function' domains.
A direct 'dose response' relationship between severity of HF and severity of CI would strengthen arguments for a causal link. HF-REF can be quantified in terms of EF or symptom burden. For both measures there is an independent association with increasing prevalence of CI [6,8,13,16,17,20] and the poorest scores on cognitive testing are most often seen in those with the severest disease [23]. Interestingly, an association with CI is also seen in those with echocardiographic evidence of reduced EF but without symptoms of HF (that is, patients with asymptomatic left ventricular systolic dysfunction) [7].
Few studies have described cognitive function in patients with HF-PEF [10,22,23,29,30,45], but the pattern seems to be that CI is a substantial problem in all HF regardless of EF. Whether the prevalence or phenotype of cognitive change differs between HF-PEF and HF-REF is not clear as there have been few comparative studies. In keeping with much of the HF and cognition literature, where data are available, there is substantial potential for bias and results are contradictory. Some authors have described higher proportion of cognitive problems in HF-REF [29], while secondary analyses of clinical trials have suggested either an equal proportion of CI across the groups or an excess of CI in those with HF-PEF [30,45].

Heart failure and delirium

Two patterns of cognitive problems in HF are recognised: a chronic, progressive decline in cognitive ability and a more acute change in cognition often in association with decompensated disease. The acute delirium and HF relationship has not been well described. Delirium is a common sequela of decompensated HF; one study estimated that 17% of unscheduled HF hospitalisations had features of delirium [46]. Where delirium accompanies HF, outcomes are generally poor with increased mortality and length of stay [46]. However, delirium is a frequent complication of most medical emergencies in older adults and the delirium of decompensated HF may be no more or less frequent than the delirium that accompanies other medical conditions such as stroke or pneumonia.

Impact of cognitive impairment in heart failure

There is a literature describing the relationship between CI and 'classical cardiovascular trial' outcomes (Table 3). In general the presence of CI in HF is associated with poorer clinical outcomes, including longer hospital admissions, increased inpatient mortality and increased 1-year mortality [37]. However, as CI seems to be associated with more severe HF and with other medical comorbidities, we should not assume that poorer outcomes are directly attributable to the cognitive state. Several other important metrics have been described in HF cohorts and all seem to be worsened by the presence of CI, including functional ability, medication adherence and institutionalisation (Table 3). Cognitive decline tends not to occur in isolation and, as with other diseases of older age, the presence of impaired cognition in HF is often associated with concomitant functional decline and poor levels of self-care [32,37,38,40-43,47].

Potential pathophysiological explanations of cognitive impairment in heart failure

Historically, research describing the pathology of the dementias has been polarised, with vocal proponents for 'amyloid' and 'cerebral small vessel disease' aetiologies. Increasingly these processes are recognised as co-existing with complex biological interactions [48]. The same is likely true of the pathogenesis of CI in HF. Chronic cerebral hypoperfusion and occult cardioembolic disease are exemplar mechanistic explanations that have dominated the literature on cognition in HF. Both processes have face validity, have strong supporting scientific and observational data and yet have traditionally been studied in isolation [49]. For ease of understanding, we will keep this dichotomy and discuss the potential pathological mechanisms separately; however, it seems likely that both processes frequently coexist in patients with HF and may exert pathological synergy.
Although most of the postulated mechanisms we will discuss have been described in the context of HF-REF, issues of cerebral hypoperfusion, thrombotic disease and concomitant cardiovascular disease are also seen in HF-PEF [2] and it seems likely they will factor in the pathogenesis of any cognitive decline seen in this syndrome.

Confounding from other diseases

Co-existence of dementia and CI has been reported in a variety of cardiovascular disorders, including CAD, myocardial infarction and valvular heart disease. Midlife exposure to the common vascular risk factors of diabetes, hypertension and smoking is associated with later life cognitive decline [16]. This background is relevant to the study of patients with HF as many have a history of one or more of these co-morbidities. As discussed previously, dissecting the contribution of HF from concomitant vascular risk and disease is challenging but is essential for future studies that wish to describe the cognitive component of HF.
AF is a potential confounding condition worthy of separate discussion. The association of AF with cognitive decline is compelling [50]. Much of the CI associated with AF will be driven by cardioembolic stroke. However, cognitive decline is also seen in patients with AF and no history of clinical stroke, possibly representing occult embolic disease [50]. AF is common in HF and prevalence increases with severity of disease. Up to 50% of patients with 'end-stage' HF have AF [51]. Increasing use of ambulatory monitors is discovering substantial undetected paroxysmal AF and so these figures may be underestimates. While AF will be a factor in the pathogenesis of some HF-related CI, it is probably not the sole explanation. Where studies have controlled for the presence of AF in their HF patient population, there remains substantial prevalent CI [10,11,13,16,31].
Any discussion of cognition in cardiac disease has to consider the effect of invasive and instrumental procedures. The interventional toolkit available to cardiologists is increasingly sophisticated, with new indications emerging. Acute and chronic neurological deficits associated with cardiac surgery are well described [52] while interventions such as cardiac catheterisation and transcatheter aortic valve replacement have also been associated with post-procedure CI [53]. The mechanism of neurological insult associated with these procedures is likely a combination of reduced cerebral perfusion and embolic disease.
As well as 'physical' conditions, mood disorder may also represent an important confounder of association between HF and CI. Clinically important depression and anxiety are common in patients with HF. Depression is found in nearly 30% of HF patients and is associated with poor outcomes [54]. There is a complex interplay between cognitive decline (particularly in the context of 'small vessel disease'), mood disorder and systemic vascular disease that is poorly understood but likely to be relevant to HF. Mood disorders are particularly important to detect as they can respond to intervention, making mood disorder in HF a potentially treatable form of cognitive decline.

Shared pathophysiology (systemic inflammation and amyloid)

Several recent studies have demonstrated the formation of tangle and plaque-like structures and fibrillar deposits (that is, the 'hallmark' lesions of Alzheimer’s disease (AD) dementia) within the myocardium of patients with hypertrophic cardiomyopathy and idiopathic dilated cardiomyopathy [55]. Mis-folded proteins in the form of intermediate oligomers have also been described in cardiac tissue, with a distribution similar to that observed in the brain of patients with AD [55], raising the possibility of a common myocardial and cerebral pathology in a subset of patients with HF.
The systemic inflammatory state recognised in patients with HF may also contribute to CI [56]. It is postulated that inflammatory mediators influence cognition via diverse cytokine-mediated interactions between neurons and glial cells. In vitro and animal models support the inflammation and cognitive decline hypothesis and studies in humans with HF are emerging, although data are far from definitive at present [56].

Acute and chronic hypoperfusion

A mechanistic link between hypotension and CI, mediated via chronic cerebral hypoperfusion and loss of the normal autoregulation of cerebral perfusion pressures, has been postulated. Many diseases, including diabetes mellitus and depression, are associated with impaired reactivity of cerebrovascular perfusion autoregulatory systems and this state seems to confer a higher risk of cognitive decline [57]. HF patients often have systemic hypotension and in the context of disordered autoregulation this could lead to further insults to cerebral perfusion. Cerebral perfusion abnormalities have been demonstrated in HF patients, with reactivity more impaired in patients with greater severity of HF.
These hypoperfusion cognitive problems are not necessarily 'vascular' dementia. In animal models, reduced cerebral blood flow triggers a neurotoxic cascade that culminates in accumulation of amyloid and hyperphosphorylated tau proteins, the classical precursors of AD. If chronic hypoperfusion is causative, then improving cerebral blood flow should reduce cognitive decline. There is some evidence to support this view in patients with severe HF who have undergone cardiac transplant, pacemaker or cardiac resynchronisation therapy, and in whom measures of cognition have stabilised or improved post-procedure [58].

Thrombosis and cerebral infarction

The potential importance of AF-related cardioembolism has been discussed. Cardioembolism is also seen in HF with sinus rhythm where ventricular function is the most important determinant of thrombus formation and potential embolic cerebral infarction [59] (Figure 2). Downregulation of thrombomodulin, structural changes in the cardiac chambers and potential blood stasis in the context of reduced myocardial contractility are associated with thrombus formation that may in turn lead to arterial events of clinical stroke or occult cerebral infarction [59]. This systemic prothrombotic phenotype increases risk of all thrombo-embolic diseases and HF is also associated with venous thromboembolism [60,61]. This is not surprising, as abnormalities in all three constituents of Virchow’s Triad (abnormal blood constituents, abnormal vessel wall and abnormal blood flow) are present in HF. Neurohormonal activation seen in HF is associated with increased production of thrombogenic factors such as von Willebrand factor, thromboxane A2 and endothelin. The end result is a hypercoagulable state with increased serum levels of circulating fibrinogen, fibrinopeptide A and D-dimer (amongst others) resulting in platelet and thrombin activation and ultimately leading to plasma hyperviscosity and thrombosis [1]. A relationship between all these circulating markers of thrombosis and haemostasis and cognitive decline, particularly 'vascular dementia', has been described [62]. It would seem intuitive that anticoagulation may prevent sequelae of thrombosis; however, studies of formal anticoagulation in HF with sinus rhythm have been equivocal. To date, no large study of anticoagulation in HF describing cognitive outcomes has been published.

Cognitive screening in heart failure services

Given the prevalence and potential impact of CI in HF, a case could be made for routine cognitive screening of HF patients. This is a controversial area with strongly held views on both sides. Recent observational data suggest that informal assessment of cognition by a cardiologist is insufficiently sensitive, with around three in four HF patients with important cognitive problems not recognised as such in routine consultations [63]. To date, routine screening for CI has not been incorporated into HF clinical guidelines; this may be due in part to the lack of a standardised screening technique that is feasible and acceptable for use in the cardiology outpatient setting. A recent systematic review of cognitive screening questionnaires utilised in HF studies concluded that the accuracy of traditional cognitive assessment measures is questionable in HF populations and appropriate thresholds/normative values need to be established [64]. In this regard we welcome ongoing work by the Cochrane Dementia and Cognitive Improvement Group to offer synthesis of test accuracy of cognitive assessments in various healthcare contexts [65].

Treatment implications of cognitive impairment in heart failure

There is an impressive evidence base to support pharmacological interventions in HF-REF. Historically HF trials have described clinical outcomes such as death, vascular events and hospitalisation with decompensated HF. There has been little focus on cognition or dementia as trial outcome or as a case mix adjuster. In fact for many of the trials that inform the HF evidence base, dementia or CI will have been an exclusion criterion. Where trialists have attempted to describe cognitive effects of HF treatment, results have been neutral [30].
Central to the treatment of HF is relatively complex multi-drug pharmacological treatment with attendant need for careful biochemical surveillance and self- monitoring. To achieve optimal outcomes requires strict adherence to prescribed evidence-based therapy [2]. Poor adherence is linked to an elevated risk of hospitalisation and death, whereas appropriate self-management may reduce these risks [2]. It seems intuitive that ensuring adherence and self-management would be especially challenging in the context of CI.
Interventions with angiotensin converting enzyme inhibitors (ACE-is), which have effects on the renin-angiotensin-aldosterone system (RAAS), have been a mainstay of HF-REF therapy for decades. ACE is also important in neurotransmitter modulation and there are theoretical reasons to believe that ACE-is may have an effect on cognitive decline. Cognitive sub-studies of the Cardiovascular Health Study and the Italian Longitudinal Study on Ageing [66,67] both reported that subjects treated with ACE-is had equivalent rates of incident dementia compared with those treated with other antihypertensives. However, there were intriguing within-class differences in cognitive outcomes - for example, between centrally and non-centrally active agents and between differing drug potencies [67]. The other pillars of HF-REF therapy, beta-blockers and mineralocorticoid receptor antagonists, may also influence cognition. Although no studies specific to HF are available, there is hypertension literature suggesting theoretical cognitive effects of beta-blockade but inconclusive evidence that this is clinically important [68]. Cognitive effects of mineralocorticoid receptor antagonists have been demonstrated in animal models but human data are limited [69].
Novel approaches to pharmacological intervention in HF are being developed, with the natriuretic peptide system a key therapeutic target. These peptides possess differing degrees of haemodynamic, neurohormonal, renal and cardiac effects which may be favourable in the HF setting and may augment the effects of RAAS blockade. Preliminary studies using inhibitors of neprilysin (also known as neutral endopeptidase), an enzyme involved in the breakdown of endogenous natriuretic peptides, have yielded encouraging results [70]. Based on this experience a phase III trial comparing the angiotensin receptor neprilysin inhibitor molecule LCZ696 to the ACE-i enalapril was undertaken in chronic HF-REF (PARADIGM-HF). This trial was recently stopped for benefit of LCZ696 over enalapril [71]. However, cardiac optimism must be tempered by caution regarding potential non-cardiac, cognitive adverse effects. Mutations in the neprilysin gene have been associated with familial forms of AD and neprilysin-deficient mice show an AD phenotype [72].
In the light of non-definitive data, how should we treat a patient with HF and CI? Cognitive enhancing medication such as acetylcholinesterase inhibitors have recognised effects on the cardiac conduction system, occasionally causing bradycardia, sick sinus syndrome or other arrhythmias (including torsades de pointes) resulting from QT prolongation through excessive cholinergic stimulation. One recent study showed donepezil to be safe in patients without symptomatic heart disease and actually reduced levels of plasma brain natriuretic peptide in patients with subclinical HF [73].
Although there are no data to suggest cognitive benefits of standard HF therapy, there are equally no signals of harm. Given the beneficial effects of pharmacological therapy on mortality and hospitalisation, it would seem sensible to consider these evidence-based medical interventions for all HF patients, tailoring the intervention to suit the patient. A multidisciplinary approach with frequent review and medication titration seems to work well. Prescribers need to be alert to the potential effects of CI on concordance with sometimes complex drug regimens. Early use of compliance aids and involvement of family or carers may help in this regard. The goal of management of HF is to provide 'seamless care' in both the community and hospital to ensure the treatment of every patient is optimal. Despite the plethora of publications and guidelines, the data consistently show a lower uptake of evidence-based investigations and therapies in older patients with consequent higher rates of HF hospitalizations and mortality [43]. The current shift away from concentration on individual drug therapies to a focus on systems of care that allow effective treatment delivery is welcomed.

Conclusion

Recurrent themes in our synthesis of the literature regarding CI and HF are a lack of primary data, methodological limitations in available research, and conflicting results. To progress our understanding we recommend increasing use of cognitive assessment using standardised screening tools in all future HF studies. Although we found numerous studies assessing prevalence, there is a dearth of studies investigating the incidence of CI in HF. Once the incidence and prevalence of CI in HF are better defined we need to evaluate the consequences of CI in HF. Identifying underlying mechanisms for CI in HF may present targets for intervention, the 'holy grail' of cognitive research. A number of processes have been postulated, and we now need confirmatory studies using new developments in neuroimaging and biomarkers in representative populations of HF patients. All of this will require a multidisciplinary approach between HF and dementia research teams. Such collaborative activity is urgently needed given the projected increases in both CI and HF.

Note

This article is part of a series on The impact of acute and chronic medical disorders on accelerated cognitive decline’, edited by Carol Brayne and Daniel Davis. Other articles in this series can be found at http://​alres.​com/​series/​medicaldisorders​.

Competing interests

JC has no competing interests. JJVM has no competing interests. TJQ has received modest honoraria, research funding and travel support from: Astra-Zeneca; Bayer; Boehringer Eingelheim; Bristol Meyers Squibb; Merck; Pfizer. He holds grants relating to cognitive assessment from British Geriatric Society; Chest Heart and Stroke Scotland; Chief Scientists Office Scotland; The Stroke Association.
Literatur
1.
Zurück zum Zitat Braunwald E. Heart Disease: A Textbook of Cardiovascular Medicine, vol. 2. Philadelphia, US: WB Saunders; 2005. Braunwald E. Heart Disease: A Textbook of Cardiovascular Medicine, vol. 2. Philadelphia, US: WB Saunders; 2005.
2.
Zurück zum Zitat McMurray JJ, Adamopoulos S, Anker SD, Auricchio A, Böhm M, Dickstein K, et al. ESC Guidelines for the diagnosis and treatment of acute and chronic heart failure 2012: The Task Force for the Diagnosis and Treatment of Acute and Chronic Heart Failure 2012 of the European Society of Cardiology. Developed in collaboration with the Heart Failure Association (HFA) of the ESC. Eur Heart J. 2012;33:1787–847.CrossRefPubMed McMurray JJ, Adamopoulos S, Anker SD, Auricchio A, Böhm M, Dickstein K, et al. ESC Guidelines for the diagnosis and treatment of acute and chronic heart failure 2012: The Task Force for the Diagnosis and Treatment of Acute and Chronic Heart Failure 2012 of the European Society of Cardiology. Developed in collaboration with the Heart Failure Association (HFA) of the ESC. Eur Heart J. 2012;33:1787–847.CrossRefPubMed
3.
Zurück zum Zitat Jhund PS, Macintyre K, Simpson CR, Lewsey JD, Stewart S, Redpath A, et al. Long-term trends in first hospitalization for heart failure and subsequent survival between 1986 and 2003: a population study of 5.1 million people. Circulation. 2009;119:515–23.CrossRefPubMed Jhund PS, Macintyre K, Simpson CR, Lewsey JD, Stewart S, Redpath A, et al. Long-term trends in first hospitalization for heart failure and subsequent survival between 1986 and 2003: a population study of 5.1 million people. Circulation. 2009;119:515–23.CrossRefPubMed
4.
Zurück zum Zitat Stewart S, MacIntyre K, Capewell S, McMurray JJ. Heart failure and the aging population: an increasing burden in the 21st century? Heart. 2003;89:49–53.CrossRefPubMedCentralPubMed Stewart S, MacIntyre K, Capewell S, McMurray JJ. Heart failure and the aging population: an increasing burden in the 21st century? Heart. 2003;89:49–53.CrossRefPubMedCentralPubMed
5.
Zurück zum Zitat Ho KKL, Pinsky JL, Kannel WB, Levy D, Pitt B. The epidemiology of heart failure: The Framingham Study. J Am Coll Cardiol. 1993;Suppl 4:A6–A13.CrossRef Ho KKL, Pinsky JL, Kannel WB, Levy D, Pitt B. The epidemiology of heart failure: The Framingham Study. J Am Coll Cardiol. 1993;Suppl 4:A6–A13.CrossRef
6.
Zurück zum Zitat Zuccalà G, Cattel C, Manes-Gravina E, Di Niro MG, Cocchi A, Bernabei R. Left ventricular dysfunction: a clue to cognitive impairment in older patients with heart failure. J Neurol Neurosurg Psychiatry. 1997;63:509–12.CrossRefPubMedCentralPubMed Zuccalà G, Cattel C, Manes-Gravina E, Di Niro MG, Cocchi A, Bernabei R. Left ventricular dysfunction: a clue to cognitive impairment in older patients with heart failure. J Neurol Neurosurg Psychiatry. 1997;63:509–12.CrossRefPubMedCentralPubMed
7.
Zurück zum Zitat Callegari S, Majani G, Giardini A, Pierobon A, Opasich C, Cobelli F, et al. Relationship between cognitive impairment and clinical status in chronic heart failure patients. Monaldi Arch Chest Dis. 2002;58:19–25.PubMed Callegari S, Majani G, Giardini A, Pierobon A, Opasich C, Cobelli F, et al. Relationship between cognitive impairment and clinical status in chronic heart failure patients. Monaldi Arch Chest Dis. 2002;58:19–25.PubMed
8.
Zurück zum Zitat Trojano L, Antonelli Inc, Acanfora D, Picone C, Mecocci P, Rengo F. Cognitive impairment: a key feature of congestive heart failure in the elderly. J Neurol. 2003;250:1456–63.CrossRefPubMed Trojano L, Antonelli Inc, Acanfora D, Picone C, Mecocci P, Rengo F. Cognitive impairment: a key feature of congestive heart failure in the elderly. J Neurol. 2003;250:1456–63.CrossRefPubMed
9.
Zurück zum Zitat Zuccalà G, Marzetti E, Cesari M, Lo Monaco MR, Antonica L, Cocchi A, et al. Correlates of cognitive impairment among patients with heart failure: results of a multicenter survey. Am J Med. 2005;118:496–502.CrossRefPubMed Zuccalà G, Marzetti E, Cesari M, Lo Monaco MR, Antonica L, Cocchi A, et al. Correlates of cognitive impairment among patients with heart failure: results of a multicenter survey. Am J Med. 2005;118:496–502.CrossRefPubMed
10.
Zurück zum Zitat Feola M, Rosso GL, Peano M, Agostini M, Aspromonte N, Carena G, et al. Correlation between cognitive impairment and prognostic parameters in patients with congestive heart failure. Arch Med Res. 2007;38:234–9.CrossRefPubMed Feola M, Rosso GL, Peano M, Agostini M, Aspromonte N, Carena G, et al. Correlation between cognitive impairment and prognostic parameters in patients with congestive heart failure. Arch Med Res. 2007;38:234–9.CrossRefPubMed
11.
Zurück zum Zitat Debette S, Bauters C, Leys D, Lamblin N, Pasquier F, de Groote P. Prevalence and determinants of cognitive impairment in chronic heart failure patients. Congest Heart Fail. 2007;13:205–8.CrossRefPubMed Debette S, Bauters C, Leys D, Lamblin N, Pasquier F, de Groote P. Prevalence and determinants of cognitive impairment in chronic heart failure patients. Congest Heart Fail. 2007;13:205–8.CrossRefPubMed
12.
Zurück zum Zitat Dodson J. Cognitive impairment in older adults with heart failure: prevalence, documentation, and impact on outcomes. Am J Med. 2013;126:12026.CrossRef Dodson J. Cognitive impairment in older adults with heart failure: prevalence, documentation, and impact on outcomes. Am J Med. 2013;126:12026.CrossRef
13.
Zurück zum Zitat Schmidt R, Fazekas F, Offenbacher H, Dusleag J, Lechner H. Brain magnetic resonance imaging and neuropsychologic evaluation of patients with idiopathic dilated cardiomyopathy. Stroke. 1991;22:195–9.CrossRefPubMed Schmidt R, Fazekas F, Offenbacher H, Dusleag J, Lechner H. Brain magnetic resonance imaging and neuropsychologic evaluation of patients with idiopathic dilated cardiomyopathy. Stroke. 1991;22:195–9.CrossRefPubMed
14.
Zurück zum Zitat Grubb NR, Simpson C, Fox KA. Memory function in patients with stable, moderate to severe cardiac failure. Am Heart J. 2000;140:E1–5.CrossRefPubMed Grubb NR, Simpson C, Fox KA. Memory function in patients with stable, moderate to severe cardiac failure. Am Heart J. 2000;140:E1–5.CrossRefPubMed
15.
Zurück zum Zitat Riegel B, Bennett JA, Davis A, Carlson B, Montague J, Robin H, et al. Cognitive impairment in heart failure: issues of measurement and etiology. Am J Crit Care. 2002;11:520–8.PubMed Riegel B, Bennett JA, Davis A, Carlson B, Montague J, Robin H, et al. Cognitive impairment in heart failure: issues of measurement and etiology. Am J Crit Care. 2002;11:520–8.PubMed
16.
Zurück zum Zitat Vogels RL, Oosterman JM, van Harten B, Scheltens P, van der Flier WM, Schroeder-Tanka JM, et al. Profile of cognitive impairment in chronic heart failure. J Am Geriatr Soc. 2007;55:1764–70.CrossRefPubMed Vogels RL, Oosterman JM, van Harten B, Scheltens P, van der Flier WM, Schroeder-Tanka JM, et al. Profile of cognitive impairment in chronic heart failure. J Am Geriatr Soc. 2007;55:1764–70.CrossRefPubMed
17.
Zurück zum Zitat Hoth KF, Poppas A, Moser DJ, Paul RH, Cohen RA. Cardiac dysfunction and cognition in older adults with heart failure. Cogn Behav Neurol. 2008;21:65–72.CrossRefPubMed Hoth KF, Poppas A, Moser DJ, Paul RH, Cohen RA. Cardiac dysfunction and cognition in older adults with heart failure. Cogn Behav Neurol. 2008;21:65–72.CrossRefPubMed
18.
Zurück zum Zitat Beer C, Ebenezer E, Fenner S, Lautenschlager NT, Arnolda L, Flicker L, et al. Contributors to cognitive impairment in congestive heart failure: a pilot case–control study. Intern Med J. 2009;39:600–5.CrossRefPubMed Beer C, Ebenezer E, Fenner S, Lautenschlager NT, Arnolda L, Flicker L, et al. Contributors to cognitive impairment in congestive heart failure: a pilot case–control study. Intern Med J. 2009;39:600–5.CrossRefPubMed
19.
Zurück zum Zitat Stanek KM, Gunstad J, Paul RH, Poppas A, Jefferson AL, Sweet LH, et al. Longitudinal cognitive performance in older adults with cardiovascular disease: evidence for improvement in heart failure. J Cardiovasc Nurs. 2009;24:192–7.CrossRefPubMedCentralPubMed Stanek KM, Gunstad J, Paul RH, Poppas A, Jefferson AL, Sweet LH, et al. Longitudinal cognitive performance in older adults with cardiovascular disease: evidence for improvement in heart failure. J Cardiovasc Nurs. 2009;24:192–7.CrossRefPubMedCentralPubMed
20.
Zurück zum Zitat Sauve MJ, Lewis WR, Blankenbiller M, Rickabaugh B, Pressler SJ. Cognitive impairments in chronic heart failure: a case controlled study. J Card Fail. 2009;15:1–10.CrossRefPubMed Sauve MJ, Lewis WR, Blankenbiller M, Rickabaugh B, Pressler SJ. Cognitive impairments in chronic heart failure: a case controlled study. J Card Fail. 2009;15:1–10.CrossRefPubMed
22.
Zurück zum Zitat Bauer L, Pozehl B, Hertzog M, Johnson J, Zimmerman L, Filipi M. A brief neuropsychological battery for use in the chronic heart failure population. Eur J Cardiovasc Nurs. 2012;11:223–30.PubMedCentralPubMed Bauer L, Pozehl B, Hertzog M, Johnson J, Zimmerman L, Filipi M. A brief neuropsychological battery for use in the chronic heart failure population. Eur J Cardiovasc Nurs. 2012;11:223–30.PubMedCentralPubMed
23.
Zurück zum Zitat Festa JR, Jia X, Cheung K, Marchidann A, Schmidt M, Shapiro PA, et al. Association of low ejection fraction with impaired verbal memory in older patients with heart failure. Arch Neurol. 2011;68:1021–6.CrossRefPubMed Festa JR, Jia X, Cheung K, Marchidann A, Schmidt M, Shapiro PA, et al. Association of low ejection fraction with impaired verbal memory in older patients with heart failure. Arch Neurol. 2011;68:1021–6.CrossRefPubMed
24.
Zurück zum Zitat Steinberg G, Lossnitzer N, Schellberg D, Mueller-Tasch T, Krueger C, Haass M, et al. Peak oxygen uptake and left ventricular ejection fraction, but not depressive symptoms, are associated with cognitive impairment in patients with chronic heart failure. Int J Gen Med. 2011;4:879–87.PubMedCentralPubMed Steinberg G, Lossnitzer N, Schellberg D, Mueller-Tasch T, Krueger C, Haass M, et al. Peak oxygen uptake and left ventricular ejection fraction, but not depressive symptoms, are associated with cognitive impairment in patients with chronic heart failure. Int J Gen Med. 2011;4:879–87.PubMedCentralPubMed
25.
Zurück zum Zitat Jefferson AL, Himali JJ, Au R, Seshadri S, Decarli C, O'Donnell CJ, et al. Relation of left ventricular ejection fraction to cognitive aging (from the Framingham Heart Study). Am J Cardiol. 2011;108:1346–51.CrossRefPubMedCentralPubMed Jefferson AL, Himali JJ, Au R, Seshadri S, Decarli C, O'Donnell CJ, et al. Relation of left ventricular ejection fraction to cognitive aging (from the Framingham Heart Study). Am J Cardiol. 2011;108:1346–51.CrossRefPubMedCentralPubMed
26.
Zurück zum Zitat Miller LA, Spitznagel MB, Alosco ML, Cohen RA, Raz N, Sweet LH, et al. Cognitive profiles in heart failure: a cluster analytic approach. J Clin Exp Neuropsychol. 2012;34:509–20.CrossRefPubMedCentralPubMed Miller LA, Spitznagel MB, Alosco ML, Cohen RA, Raz N, Sweet LH, et al. Cognitive profiles in heart failure: a cluster analytic approach. J Clin Exp Neuropsychol. 2012;34:509–20.CrossRefPubMedCentralPubMed
27.
Zurück zum Zitat Almeida OP, Garrido GJ, Beer C, Lautenschlager NT, Arnolda L, Flicker L. Cognitive and brain changes associated with ischaemic heart disease and heart failure. Eur Heart J. 2012;33:1769–76.CrossRefPubMed Almeida OP, Garrido GJ, Beer C, Lautenschlager NT, Arnolda L, Flicker L. Cognitive and brain changes associated with ischaemic heart disease and heart failure. Eur Heart J. 2012;33:1769–76.CrossRefPubMed
28.
Zurück zum Zitat Hawkins LA, Kilian S, Firek A, Kashner TM, Firek CJ, Silvet H. Cognitive impairment and medication adherence in outpatients with heart failure. Heart Lung. 2012;41:572–82.CrossRefPubMed Hawkins LA, Kilian S, Firek A, Kashner TM, Firek CJ, Silvet H. Cognitive impairment and medication adherence in outpatients with heart failure. Heart Lung. 2012;41:572–82.CrossRefPubMed
29.
Zurück zum Zitat Bratzke-Bauer L. Neuropsychological patterns differ by type of left ventricular dysfunction in heart failure. Arch Clin Neuropsychol. 2013;28:114–24.CrossRefPubMedCentralPubMed Bratzke-Bauer L. Neuropsychological patterns differ by type of left ventricular dysfunction in heart failure. Arch Clin Neuropsychol. 2013;28:114–24.CrossRefPubMedCentralPubMed
30.
Zurück zum Zitat Huijts M, van Oostenbrugge RJ, Duits A, Burkard T, Muzzarelli S, Maeder MT, et al. Cognitive impairment in heart failure: results from the Trial of Intensified versus standard Medical therapy in Elderly patients with Congestive Heart Failure (TIME-CHF) randomized trial. Eur J Heart Fail. 2013;15:699–707.CrossRefPubMed Huijts M, van Oostenbrugge RJ, Duits A, Burkard T, Muzzarelli S, Maeder MT, et al. Cognitive impairment in heart failure: results from the Trial of Intensified versus standard Medical therapy in Elderly patients with Congestive Heart Failure (TIME-CHF) randomized trial. Eur J Heart Fail. 2013;15:699–707.CrossRefPubMed
31.
Zurück zum Zitat Kindermann I, Fischer D, Karbach J, Link A, Walenta K, Barth C, et al. Cognitive function in patients with decompensated heart failure: the Cognitive Impairment in Heart Failure (CogImpair-HF) study. Eur J Heart Fail. 2012;14:404–13.CrossRefPubMed Kindermann I, Fischer D, Karbach J, Link A, Walenta K, Barth C, et al. Cognitive function in patients with decompensated heart failure: the Cognitive Impairment in Heart Failure (CogImpair-HF) study. Eur J Heart Fail. 2012;14:404–13.CrossRefPubMed
32.
Zurück zum Zitat Karlsson MR, Edner M, Henriksson P, Mejhert M, Persson H, Grut M. Billing, A nurse-based management program in heart failure patients affects females and persons with cognitive dysfunction most. Patient Educ Couns. 2005;58:146–53.CrossRefPubMed Karlsson MR, Edner M, Henriksson P, Mejhert M, Persson H, Grut M. Billing, A nurse-based management program in heart failure patients affects females and persons with cognitive dysfunction most. Patient Educ Couns. 2005;58:146–53.CrossRefPubMed
33.
Zurück zum Zitat Tanne D. Cognitive functions in severe congestive heart failure before and after an exercise training program. Int J Cardiol. 2005;103:145–9.CrossRefPubMed Tanne D. Cognitive functions in severe congestive heart failure before and after an exercise training program. Int J Cardiol. 2005;103:145–9.CrossRefPubMed
34.
Zurück zum Zitat Almeida O. Brain and mood changes over 2 years in healthy controls and adults with heart failure and ischaemic heart disease. Eur J Heart Failure. 2013;15:850–8.CrossRef Almeida O. Brain and mood changes over 2 years in healthy controls and adults with heart failure and ischaemic heart disease. Eur J Heart Failure. 2013;15:850–8.CrossRef
35.
Zurück zum Zitat Hjelm C, Dahl A, Brostrom A, Martensson J, Johansson B, Stromberg A. The influence of heart failure on longitudinal changes in cognition among individuals 80 years of age and older. J Clin Nurs. 2012;21:994–1003.CrossRefPubMed Hjelm C, Dahl A, Brostrom A, Martensson J, Johansson B, Stromberg A. The influence of heart failure on longitudinal changes in cognition among individuals 80 years of age and older. J Clin Nurs. 2012;21:994–1003.CrossRefPubMed
36.
Zurück zum Zitat Riegel B, Lee CS, Glaser D, Moelter ST. Patterns of change in cognitive function over six months in adults with chronic heart failure. Cardiol Res Pract. 2012;2012:631075.PubMedCentralPubMed Riegel B, Lee CS, Glaser D, Moelter ST. Patterns of change in cognitive function over six months in adults with chronic heart failure. Cardiol Res Pract. 2012;2012:631075.PubMedCentralPubMed
37.
Zurück zum Zitat Zuccalà G, Pedone C, Cesari M, Onder G, Pahor M, Marzetti E, et al. The effects of cognitive impairment on mortality among hospitalized patients with heart failure. Am J Med. 2003;115:97–103.CrossRefPubMed Zuccalà G, Pedone C, Cesari M, Onder G, Pahor M, Marzetti E, et al. The effects of cognitive impairment on mortality among hospitalized patients with heart failure. Am J Med. 2003;115:97–103.CrossRefPubMed
38.
Zurück zum Zitat Riegel B, Vaughan Dickson V, Goldberg LR, Deatrick JA. Factors associated with the development of expertise in heart failure self-care. J Nurs Res. 2007;56:235–43.CrossRef Riegel B, Vaughan Dickson V, Goldberg LR, Deatrick JA. Factors associated with the development of expertise in heart failure self-care. J Nurs Res. 2007;56:235–43.CrossRef
39.
Zurück zum Zitat Cameron J. Testing a model of patient characteristics, psychological status, and cognitive function as predictors of self-care in persons with chronic heart failure. J Acute Crit Care. 2009;38:410–8.CrossRef Cameron J. Testing a model of patient characteristics, psychological status, and cognitive function as predictors of self-care in persons with chronic heart failure. J Acute Crit Care. 2009;38:410–8.CrossRef
40.
Zurück zum Zitat Cameron J, Worrall-Carter L, Page K, Riegel B, Lo SK, Stewart S. Does cognitive impairment predict poor self-care in patients with heart failure. Eur J Heart Fail. 2010;12:508–15.CrossRefPubMed Cameron J, Worrall-Carter L, Page K, Riegel B, Lo SK, Stewart S. Does cognitive impairment predict poor self-care in patients with heart failure. Eur J Heart Fail. 2010;12:508–15.CrossRefPubMed
41.
Zurück zum Zitat Pulignano G, Del Sindaco D, Minardi G, Tarantini L, Cioffi G, Bernardi L, et al. Translation and validation of the Italian version of the European Heart Failure Self-Care Behaviour Scale. J Cardiovasc Med. 2010;11:493–8.CrossRef Pulignano G, Del Sindaco D, Minardi G, Tarantini L, Cioffi G, Bernardi L, et al. Translation and validation of the Italian version of the European Heart Failure Self-Care Behaviour Scale. J Cardiovasc Med. 2010;11:493–8.CrossRef
42.
Zurück zum Zitat Alosco ML, Spitznagel MB, Cohen R, Sweet LH, Colbert LH, Josephson R, et al. Reduced cognitive function predicts functional decline in patients with heart failure over 12 months. Eur J Cardiovasc Nurs. 2013;13:304–10.CrossRefPubMed Alosco ML, Spitznagel MB, Cohen R, Sweet LH, Colbert LH, Josephson R, et al. Reduced cognitive function predicts functional decline in patients with heart failure over 12 months. Eur J Cardiovasc Nurs. 2013;13:304–10.CrossRefPubMed
43.
Zurück zum Zitat Harkness K, Heckman GA, Akhtar-Danesh N, Demers C, Gunn E, McKelvie RS. Cognitive function and self-care management in older patients with heart failure. Eur J Cardiovasc Nurs. 2013;13:277–84.CrossRefPubMed Harkness K, Heckman GA, Akhtar-Danesh N, Demers C, Gunn E, McKelvie RS. Cognitive function and self-care management in older patients with heart failure. Eur J Cardiovasc Nurs. 2013;13:277–84.CrossRefPubMed
44.
Zurück zum Zitat Russ TC, Stamatakis E, Hamer M, Starr JM, Kivimaki M, Batty GD. Association between psychological distress and mortality: individual participant pooled analysis of 10 prospective cohort studies. BMJ. 2012;345:e4933.CrossRefPubMedCentralPubMed Russ TC, Stamatakis E, Hamer M, Starr JM, Kivimaki M, Batty GD. Association between psychological distress and mortality: individual participant pooled analysis of 10 prospective cohort studies. BMJ. 2012;345:e4933.CrossRefPubMedCentralPubMed
45.
Zurück zum Zitat van den Hurk K, Reijmer YD, van den Berg E, Alssema M, Nijpels G, Kostense PJ, et al. Heart failure and cognitive function in the general population: the Hoorn Study. Eur J Heart Fail. 2011;13:1362–9.CrossRefPubMed van den Hurk K, Reijmer YD, van den Berg E, Alssema M, Nijpels G, Kostense PJ, et al. Heart failure and cognitive function in the general population: the Hoorn Study. Eur J Heart Fail. 2011;13:1362–9.CrossRefPubMed
46.
Zurück zum Zitat Uthamalingham S. Usefulness of acute delirium as a predictor of adverse outcomes in patients >65 years of age with acute decompensated heart failure. Am J Cardiol. 2011;108:402–8.CrossRef Uthamalingham S. Usefulness of acute delirium as a predictor of adverse outcomes in patients >65 years of age with acute decompensated heart failure. Am J Cardiol. 2011;108:402–8.CrossRef
47.
Zurück zum Zitat Alosco ML, Spitznagel MB, Raz N, Cohen R, Sweet LH, Colbert LH, et al. Executive dysfunction is independently associated with reduced functional independence in heart failure. J Clin Nurs. 2014;23:829–36.CrossRefPubMed Alosco ML, Spitznagel MB, Raz N, Cohen R, Sweet LH, Colbert LH, et al. Executive dysfunction is independently associated with reduced functional independence in heart failure. J Clin Nurs. 2014;23:829–36.CrossRefPubMed
48.
Zurück zum Zitat Jicha GA, Parisi JE, Dickson DW, Johnson K, Cha R, Ivnik RJ, et al. Neuropathologic outcome of mild cognitive impairment following progression to clinical dementia. Arch Neurol. 2006;63:674–81.CrossRefPubMed Jicha GA, Parisi JE, Dickson DW, Johnson K, Cha R, Ivnik RJ, et al. Neuropathologic outcome of mild cognitive impairment following progression to clinical dementia. Arch Neurol. 2006;63:674–81.CrossRefPubMed
49.
Zurück zum Zitat Georgiadis D, Sievert M, Cencetti S, Uhlmann F, Krivokuca M, Zierz S, et al. Cerebrovascular reactivity is impaired in patients with cardiac failure. Eur Heart J. 2000;21:407–13.CrossRefPubMed Georgiadis D, Sievert M, Cencetti S, Uhlmann F, Krivokuca M, Zierz S, et al. Cerebrovascular reactivity is impaired in patients with cardiac failure. Eur Heart J. 2000;21:407–13.CrossRefPubMed
50.
Zurück zum Zitat Kalantarian S, Stern TA, Mansour M, Ruskin JN. Cognitive impairment associated with atrial fibrillation: a meta-analysis. Ann Intern Med. 2013;158:338–46.CrossRefPubMed Kalantarian S, Stern TA, Mansour M, Ruskin JN. Cognitive impairment associated with atrial fibrillation: a meta-analysis. Ann Intern Med. 2013;158:338–46.CrossRefPubMed
51.
Zurück zum Zitat Neuberger HR, Mewis C, van Veldhuisen DJ, Schotten U, van Gelder IC, Allessie MA, et al. Management of atrial fibrillation in patients with heart failure. Eur Heart J. 2007;28:2568–77.CrossRefPubMed Neuberger HR, Mewis C, van Veldhuisen DJ, Schotten U, van Gelder IC, Allessie MA, et al. Management of atrial fibrillation in patients with heart failure. Eur Heart J. 2007;28:2568–77.CrossRefPubMed
52.
Zurück zum Zitat Caplan LR. Translating what is known about neurological complications of coronary artery bypass graft surgery into action. Arch Neurol. 2009;66:1062–4.CrossRefPubMed Caplan LR. Translating what is known about neurological complications of coronary artery bypass graft surgery into action. Arch Neurol. 2009;66:1062–4.CrossRefPubMed
53.
Zurück zum Zitat Kahlert P, Knipp SC, Schlamann M, Thielmann M, Al-Rashid F, Weber M, et al. Silent and apparent cerebral ischemia after percutaneous transfemoral aortic valve implantation: a diffusion-weighted magnetic resonance imaging study. Circulation. 2010;121:870–8.CrossRefPubMed Kahlert P, Knipp SC, Schlamann M, Thielmann M, Al-Rashid F, Weber M, et al. Silent and apparent cerebral ischemia after percutaneous transfemoral aortic valve implantation: a diffusion-weighted magnetic resonance imaging study. Circulation. 2010;121:870–8.CrossRefPubMed
54.
Zurück zum Zitat Diez-Quevedo C, Lupón J, González B, Urrutia A, Cano L, Cabanes R, et al. Depression, antidepressants, and long-term mortality in heart failure. Int J Cardiol. 2013;167:1217–25.CrossRefPubMed Diez-Quevedo C, Lupón J, González B, Urrutia A, Cano L, Cabanes R, et al. Depression, antidepressants, and long-term mortality in heart failure. Int J Cardiol. 2013;167:1217–25.CrossRefPubMed
55.
Zurück zum Zitat Willis MS, Patterson C. Proteotoxicity and cardiac dysfunction - Alzheimer's disease of the heart? N Engl J Med. 2013;368:455–64.CrossRefPubMed Willis MS, Patterson C. Proteotoxicity and cardiac dysfunction - Alzheimer's disease of the heart? N Engl J Med. 2013;368:455–64.CrossRefPubMed
56.
Zurück zum Zitat Athilingam P, Moynihan J, Chen L, D'Aoust R, Groer M, Kip K. Elevated Levels of Interleukin 6 and C-reactive protein associated with cognitive impairment in heart failure. Congest Heart Fail. 2013;19:92–8.CrossRefPubMedCentralPubMed Athilingam P, Moynihan J, Chen L, D'Aoust R, Groer M, Kip K. Elevated Levels of Interleukin 6 and C-reactive protein associated with cognitive impairment in heart failure. Congest Heart Fail. 2013;19:92–8.CrossRefPubMedCentralPubMed
57.
Zurück zum Zitat Tchistiakova E, Anderson ND, Greenwood CE, MacIntosh BJ. Combined effects of type 2 diabetes and hypertension associated with cortical thinning and impaired cerebrovascular reactivity relative to hypertension alone in older adults. Neuroimage Clin. 2014;5:36–41.CrossRefPubMedCentralPubMed Tchistiakova E, Anderson ND, Greenwood CE, MacIntosh BJ. Combined effects of type 2 diabetes and hypertension associated with cortical thinning and impaired cerebrovascular reactivity relative to hypertension alone in older adults. Neuroimage Clin. 2014;5:36–41.CrossRefPubMedCentralPubMed
58.
Zurück zum Zitat Conti JB, Sears SF. Cardiac resynchronization therapy: can we make our heart failure patients smarter? Trans Am Clin Climatol Assoc. 2007;118:153–64.PubMedCentralPubMed Conti JB, Sears SF. Cardiac resynchronization therapy: can we make our heart failure patients smarter? Trans Am Clin Climatol Assoc. 2007;118:153–64.PubMedCentralPubMed
59.
Zurück zum Zitat Kalaria VG, Passannante MR, Shah T, Modi K, Weisse AB. Effect of mitral regurgitation on left ventricular thrombus formation in dilated cardiomyopathy. Am Heart J. 1998;135:215–20.CrossRefPubMed Kalaria VG, Passannante MR, Shah T, Modi K, Weisse AB. Effect of mitral regurgitation on left ventricular thrombus formation in dilated cardiomyopathy. Am Heart J. 1998;135:215–20.CrossRefPubMed
60.
Zurück zum Zitat Al-Khadra AS, Salem DN, Rand WM, Udelson JE, Smith JJ, Konstam MA. Antiplatelet agents and survival: a cohort analysis from the Studies of Left Ventricular Dysfunction (SOLVD) trial. J Am Coll Cardiol. 1998;31:419–25.CrossRefPubMed Al-Khadra AS, Salem DN, Rand WM, Udelson JE, Smith JJ, Konstam MA. Antiplatelet agents and survival: a cohort analysis from the Studies of Left Ventricular Dysfunction (SOLVD) trial. J Am Coll Cardiol. 1998;31:419–25.CrossRefPubMed
61.
Zurück zum Zitat Freudenberger RS, Hellkamp AS, Halperin JL, Poole J, Anderson J, Johnson G, et al. Risk of thromboembolism in heart failure: an analysis from the Sudden Cardiac Death in Heart Failure Trial (SCD-HeFT). Circulation. 2007;115:2637–41.CrossRefPubMed Freudenberger RS, Hellkamp AS, Halperin JL, Poole J, Anderson J, Johnson G, et al. Risk of thromboembolism in heart failure: an analysis from the Sudden Cardiac Death in Heart Failure Trial (SCD-HeFT). Circulation. 2007;115:2637–41.CrossRefPubMed
62.
Zurück zum Zitat Quinn TJ, Gallacher J, Deary IJ, Lowe GD, Fenton C, Stott DJ. Association between circulating hemostatic measures and dementia or cognitive impairment: systematic review and meta-analyzes. J Thromb Haemost. 2011;9:1475–82.CrossRefPubMed Quinn TJ, Gallacher J, Deary IJ, Lowe GD, Fenton C, Stott DJ. Association between circulating hemostatic measures and dementia or cognitive impairment: systematic review and meta-analyzes. J Thromb Haemost. 2011;9:1475–82.CrossRefPubMed
63.
Zurück zum Zitat Hanon O, Vidal JS, de Groote P, Galinier M, Isnard R, Logeart D, et al. Prevalence of memory disorders in ambulatory patients aged >/=70 years with chronic heart failure (from the EFICARE Study). Am J Cardiol. 2014;113:1205–10.CrossRefPubMed Hanon O, Vidal JS, de Groote P, Galinier M, Isnard R, Logeart D, et al. Prevalence of memory disorders in ambulatory patients aged >/=70 years with chronic heart failure (from the EFICARE Study). Am J Cardiol. 2014;113:1205–10.CrossRefPubMed
64.
Zurück zum Zitat Davis KK, Allen JK. Identifying cognitive impairment in heart failure: a review of screening measures. Heart Lung. 2013;42:92–7.CrossRefPubMed Davis KK, Allen JK. Identifying cognitive impairment in heart failure: a review of screening measures. Heart Lung. 2013;42:92–7.CrossRefPubMed
65.
Zurück zum Zitat Quinn TJ, Fearon P, Noel-Storr AH, Young C, McShane R, Stott DJ. Informant Questionnaire on Cognitive Decline in the Elderly (IQCODE) for the diagnosis of dementia within community dwelling populations. Cochrane Database Syst Rev. 2014;4:CD010079.PubMed Quinn TJ, Fearon P, Noel-Storr AH, Young C, McShane R, Stott DJ. Informant Questionnaire on Cognitive Decline in the Elderly (IQCODE) for the diagnosis of dementia within community dwelling populations. Cochrane Database Syst Rev. 2014;4:CD010079.PubMed
66.
Zurück zum Zitat Sink KM, Leng X, Williamson J, Kritchevsky SB, Yaffe K, Kuller L, et al. Angiotensin-converting enzyme inhibitors and cognitive decline in older adults with hypertension: results from the Cardiovascular Health Study. Arch Intern Med. 2009;169:1195–202.CrossRefPubMedCentralPubMed Sink KM, Leng X, Williamson J, Kritchevsky SB, Yaffe K, Kuller L, et al. Angiotensin-converting enzyme inhibitors and cognitive decline in older adults with hypertension: results from the Cardiovascular Health Study. Arch Intern Med. 2009;169:1195–202.CrossRefPubMedCentralPubMed
67.
Zurück zum Zitat Solfrizzi V, Scafato E, Frisardi V, Seripa D, Logroscino G, Kehoe PG, et al. Angiotensin-converting enzyme inhibitors and incidence of mild cognitive impairment. The Italian Longitudinal Study on Aging. Age (Dordr). 2013;35:441–53.CrossRef Solfrizzi V, Scafato E, Frisardi V, Seripa D, Logroscino G, Kehoe PG, et al. Angiotensin-converting enzyme inhibitors and incidence of mild cognitive impairment. The Italian Longitudinal Study on Aging. Age (Dordr). 2013;35:441–53.CrossRef
68.
Zurück zum Zitat Levi MN, Macquin-Mavier I, Tropeano AI, Bachoud-Levi AC, Maison P. Antihypertensive classes, cognitive decline and incidence of dementia: a network meta-analysis. J Hypertens. 2013;31:1073–82.CrossRef Levi MN, Macquin-Mavier I, Tropeano AI, Bachoud-Levi AC, Maison P. Antihypertensive classes, cognitive decline and incidence of dementia: a network meta-analysis. J Hypertens. 2013;31:1073–82.CrossRef
69.
Zurück zum Zitat Korte SM, Korte-Bouws GA, Koob GF, De Kloet ER, Bohus B. Mineralocorticoid and glucocorticoid receptor antagonists in animal models of anxiety. Pharmacol Biochem Behav. 1996;54:261–7.CrossRefPubMed Korte SM, Korte-Bouws GA, Koob GF, De Kloet ER, Bohus B. Mineralocorticoid and glucocorticoid receptor antagonists in animal models of anxiety. Pharmacol Biochem Behav. 1996;54:261–7.CrossRefPubMed
70.
Zurück zum Zitat Packer M, Califf RM, Konstam MA, Krum H, McMurray JJ, Rouleau JL, et al. Comparison of omapatrilat and enalapril in patients with chronic heart failure: the Omapatrilat Versus Enalapril Randomized Trial of Utility in Reducing Events (OVERTURE). Circulation. 2002;106:920–6.CrossRefPubMed Packer M, Califf RM, Konstam MA, Krum H, McMurray JJ, Rouleau JL, et al. Comparison of omapatrilat and enalapril in patients with chronic heart failure: the Omapatrilat Versus Enalapril Randomized Trial of Utility in Reducing Events (OVERTURE). Circulation. 2002;106:920–6.CrossRefPubMed
71.
Zurück zum Zitat McMurray JJV, Packer M, Desai AS, Gong J, Lefkowitz MP, Rizkala AR, et al. Angiotensin–neprilysin inhibition versus enalapril in heart failure. N Engl J Med. 2014;371:993–1004.CrossRefPubMed McMurray JJV, Packer M, Desai AS, Gong J, Lefkowitz MP, Rizkala AR, et al. Angiotensin–neprilysin inhibition versus enalapril in heart failure. N Engl J Med. 2014;371:993–1004.CrossRefPubMed
72.
Zurück zum Zitat Madani R, Poirier R, Wolfer DP, Welzl H, Groscurth P, Lipp HP, et al. Lack of neprilysin suffices to generate murine amyloid-like deposits in the brain and behavioral deficit in vivo. J Neurosci Res. 2006;84:1871–8.CrossRefPubMed Madani R, Poirier R, Wolfer DP, Welzl H, Groscurth P, Lipp HP, et al. Lack of neprilysin suffices to generate murine amyloid-like deposits in the brain and behavioral deficit in vivo. J Neurosci Res. 2006;84:1871–8.CrossRefPubMed
73.
Zurück zum Zitat Kubo T, Sato T, Noguchi T, Kitaoka H, Yamasaki F, Kamimura N, et al. Influences of donepezil on cardiovascular system - possible therapeutic benefits for heart failure - donepezil cardiac test registry (DOCTER) study. J Cardiovasc Pharmacol. 2012;60:310–4.CrossRefPubMed Kubo T, Sato T, Noguchi T, Kitaoka H, Yamasaki F, Kamimura N, et al. Influences of donepezil on cardiovascular system - possible therapeutic benefits for heart failure - donepezil cardiac test registry (DOCTER) study. J Cardiovasc Pharmacol. 2012;60:310–4.CrossRefPubMed
Metadaten
Titel
‘Hearts and minds’: association, causation and implication of cognitive impairment in heart failure
verfasst von
Jane A Cannon
John JV McMurray
Terry J Quinn
Publikationsdatum
01.12.2015
Verlag
BioMed Central
Erschienen in
Alzheimer's Research & Therapy / Ausgabe 1/2015
Elektronische ISSN: 1758-9193
DOI
https://doi.org/10.1186/s13195-015-0106-5

Weitere Artikel der Ausgabe 1/2015

Alzheimer's Research & Therapy 1/2015 Zur Ausgabe

Leitlinien kompakt für die Neurologie

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Update Neurologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.