Skip to main content
Erschienen in: BMC Medicine 1/2023

Open Access 01.12.2023 | Research article

A network medicine approach to study comorbidities in heart failure with preserved ejection fraction

verfasst von: Jan D. Lanzer, Alberto Valdeolivas, Mark Pepin, Hauke Hund, Johannes Backs, Norbert Frey, Hans-Christoph Friederich, Jobst-Hendrik Schultz, Julio Saez-Rodriguez, Rebecca T. Levinson

Erschienen in: BMC Medicine | Ausgabe 1/2023

Abstract

Background

Comorbidities are expected to impact the pathophysiology of heart failure (HF) with preserved ejection fraction (HFpEF). However, comorbidity profiles are usually reduced to a few comorbid disorders. Systems medicine approaches can model phenome-wide comorbidity profiles to improve our understanding of HFpEF and infer associated genetic profiles.

Methods

We retrospectively explored 569 comorbidities in 29,047 HF patients, including 8062 HFpEF and 6585 HF with reduced ejection fraction (HFrEF) patients from a German university hospital. We assessed differences in comorbidity profiles between HF subtypes via multiple correspondence analysis. Then, we used machine learning classifiers to identify distinctive comorbidity profiles of HFpEF and HFrEF patients. Moreover, we built a comorbidity network (HFnet) to identify the main disease clusters that summarized the phenome-wide comorbidity. Lastly, we predicted novel gene candidates for HFpEF by linking the HFnet to a multilayer gene network, integrating multiple databases. To corroborate HFpEF candidate genes, we collected transcriptomic data in a murine HFpEF model. We compared predicted genes with the murine disease signature as well as with the literature.

Results

We found a high degree of variance between the comorbidity profiles of HFpEF and HFrEF, while each was more similar to HFmrEF. The comorbidities present in HFpEF patients were more diverse than those in HFrEF and included neoplastic, osteologic and rheumatoid disorders. Disease communities in the HFnet captured important comorbidity concepts of HF patients which could be assigned to HF subtypes, age groups, and sex. Based on the HFpEF comorbidity profile, we predicted and recovered gene candidates, including genes involved in fibrosis (COL3A1, LOX, SMAD9, PTHL), hypertrophy (GATA5, MYH7), oxidative stress (NOS1, GSST1, XDH), and endoplasmic reticulum stress (ATF6). Finally, predicted genes were significantly overrepresented in the murine transcriptomic disease signature providing additional plausibility for their relevance.

Conclusions

We applied systems medicine concepts to analyze comorbidity profiles in a HF patient cohort. We were able to identify disease clusters that helped to characterize HF patients. We derived a distinct comorbidity profile for HFpEF, which was leveraged to suggest novel candidate genes via network propagation. The identification of distinctive comorbidity profiles and candidate genes from routine clinical data provides insights that may be leveraged to improve diagnosis and identify treatment targets for HFpEF patients.

Graphical Abstract

Hinweise

Supplementary Information

The online version contains supplementary material available at https://​doi.​org/​10.​1186/​s12916-023-02922-7.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Abkürzungen
AUROC
Area under the receiver operating characteristic
BH
Benjamini–Hochberg
DC
Disease clusters
GO
Gene Ontology
HF
Heart failure
HFnet
Heart failure comorbidity network
HFhetnet
Heart failure multilayer heterogeneous network
HFmrEF
Heart failure with mid-range ejection fraction
HFpEF
Heart failure with preserved ejection fraction
HFrEF
Heart failure with reduced ejection fraction
ICD
Implantable cardioverter defibrillator
ICD-10
International classification of disease, version 10
LvEF
Left ventricular ejection fraction
MCA
Multiple correspondence analysis
NTproBNP
N-terminal pro b-type natriuretic peptide
PheCodes
Phenome-wide association scan codes
PPI
Protein–protein interaction
UMLS
Unified Medical Language System

Background

Heart failure (HF) with preserved ejection fraction (HFpEF) represents an unmet public health concern with rising prevalence [1, 2]. Accumulating evidence indicates HFpEF is clinically and biologically distinct from HF with reduced ejection fraction (HFrEF), as reflected by missing therapy response in HFpEF patients to drugs effective in HFrEF [3]. HF patients suffer from a wide range of comorbidities, which are considered important for HF development and progression [4]. In the pathogenesis of HFpEF, comorbidities have been suggested as causal factors [3, 5] and could possibly be linked to genetic etiology. Treatment of comorbidity has also been shown to have beneficial effects of cardiac physiology [6], emphasizing the potential to address HF subtypes through their comorbidities.
Systems medicine attempts to model disease in a holistic manner. One facet of this, network medicine, is used to analyze complex systems such as patients, organs, or cells via network representation [7, 8]. Comorbidity networks represent diseases as nodes, connected via edges based on co-occurrence in patients. These networks can be used to define disease modules or explore topological changes between patient cohorts [912]. Previous work has shown that disease comorbidity is also often linked to shared disease genes that locate close together in gene-based networks like protein–protein interaction networks [12, 13]. This observation is often the basis of network-based gene prediction, where novel disease genes are predicted based on network proximity to known disease genes.
Cardiovascular diseases are particularly suited for system medicine approaches due to the typical multiorgan involvement [14] and multifactorial etiology [15]. To date, such approaches to study HFpEF have been limited, though the comorbidity-driven pathophysiology of HFpEF makes it a promising subject. In addition, despite the technological advances in multi-omics, knowledge of molecular characteristics of HFpEF remains limited, possibly due to difficulties of biopsy acquisition in HFpEF patients [16] and heterogeneity of HFpEF patients [17].
In this study, we applied a network medicine approach to describe comorbidity patterns in HFpEF and investigate a shared genetic background associated with these patterns. We first demonstrated that comorbidity profiles vary between HFpEF and HFrEF patients and derived distinct comorbidity profiles for each cohort. Then, we built a comorbidity network that contained disease clusters relevant for HF patients. The construction of a multilayer heterogeneous network by integration of prior knowledge resources allowed us to translate the comorbidity profiles into a gene signature for HFpEF. We corroborated this signature in the cardiac transcriptome of a murine HFpEF model. This network medicine approach allowed us to identify distinct comorbidity profiles and novel genetic patterns in HFpEF.

Methods

Study population

The study population was derived from a research data warehouse containing data from patients that visited the Department of Cardiology, Angiology, and Pneumology at Heidelberg University Hospital, Heidelberg, Germany [18]. Heidelberg University Hospital acts as a tertiary care center for the surrounding region, specializing in the treatment of cardiomyopathy. From this data warehouse, we identified patients with HF, visiting between 01.01.2008 and 01.01.2021. The study protocol was approved by the local ethics committee. HF was defined as two or more HF-relevant International Classification of Disease, version 10 (ICD-10) diagnosis codes (I50*, I11.0, I13.0, I13.2, I42.0, I42.5, I42.8, I42.9, I25.5) or at least one HF-relevant diagnosis and at least one of the following criteria: (i) elevated N-terminal pro b-type natriuretic peptide (NTproBNP) (> 120 ng/ml), (ii) recorded New York Heart Association functional class, (iii) echocardiography based E/e’ > 15 ( ratio of early diastolic mitral inflow velocity to early diastolic mitral annulus velocity), (iv) echocardiography or MRI-based left ventricular ejection fraction (LvEF) < 50%, and (v) documented loop diuretic. Patients with HF before age 40, those with a diagnosis of inheritable cardiomyopathy (I42.1-I42.4, I42.6, I42.7), and heart transplant patients (Z94.3) were excluded from the HF cohort. Within the HF cohort, HF subtypes were identified, based on echocardiographic or MRI-based LvEF. Patients with LvEF ≥ 50% were labeled HFpEF, LvEF 40–50% HFmrEF (HF with mid-range ejection fraction), and ≤ 40% HFrEF (Fig. 1). For all patients in the HF cohort, demographics, ICD-10 codes, operational and procedural codes, and targeted clinically relevant measurements were processed (Additional file 1: Fig. S1A, B) [11, 1938].

Multiple correspondence analysis (MCA)

Disease profiles of HFpEF, HFrEF, and HFmrEF cohorts were captured as binary variables (0—patient has no record, 1—patient has a record of disease) of 569 phenome-wide association scan codes (PheCodes) (Additional file 1: Supplementary Methods). In this feature space (569 comorbidities × 17,665 HF patients), we performed MCA (R-package FactoMiner [39]). Each MCA dimension was then tested for association with clinical covariates with linear regression models (e.g., MCA-dimension 1 ~ age). For each covariate, we then summed the variance associated to all significantly associated dimensions (p-value < 0.05) as an estimate for the total associated variance.

Patient classifier

We trained random forest and regularized logistic regression (elastic net) models to predict HFpEF and HFrEF cohort labels on 569 PheCodes. The highest achieved mean area under the receiver operating characteristic (AUROC) in hyperparameter tuning was reported as an estimate for the model test error (Additional file 1: Supplementary Methods).
To derive the comorbidity profiles for HFpEF and HFrEF, we performed a forward selection with an L1-regularized logistic regression model of the 196 non-zero features from the elastic net model. Models were trained in R with R packages tidymodels using model engines from glmnet [40] and ranger [41].

HF comorbidity network (HFnet) construction

In disease comorbidity networks, nodes represent diseases while edges represent statistical association of co-occurrence, resulting in the graphical depiction of comorbidities as diseases that are statistically dependent. In detail, we selected edges using Fisher’s exact test for estimating statistical dependence and its Benjamini–Hochberg (BH) corrected p-value (< 0.0001) to discard non-significant disease pairs and keep a more sparse network structure. To determine strength of association, we calculated ɸ correlation, which can be interpreted as a Pearson correlation for binary variables. We selected all edges with positive correlation. To account for bias in ɸ correlations, we scaled the values by dividing by mean correlation values for every disease and assigned these values as edge weights [42].
Network node characteristics, such as betweenness, closeness, and degree centrality, and transitivity were calculated with the igraph R package. To calculate metrics based on graph distance, we replaced weights for edge i (Wi) with a new edge score Si: \(S_i= \mathrm{max}(W)-W_i\). The constructed network was then compared to other comorbidity networks (Additional file 1: Supplementary Methods).

HF heterogeneous network (HFhetnet) construction

Disease-gene association

We used disease–gene associations provided by DisGeNet v7.0 [43, 44] and mapped the ICD-10 codes in DisGeNet to PheCodes (Additional file 1: Supplementary Methods). To ensure that the most frequent diseases in our cohort were mapped, we selected the most frequent 3-digit ICD-10 codes that were not mapped to DisGeNet and performed manual annotation via Unified Medical Language System (UMLS) IDs for 23 disease entities (Additional file 2: Supp. Table 1), e.g., PheCode 427.2 (atrial fibrillation) was manually mapped to the UMLS ID C000423. We only included disease–gene associations with a DisGeNet confidence score > 0.29. This cut-off was chosen, such that either one curated source or multiple experimental sources were necessary for disease–gene associations. Details on DisGeNET score calculation can be found at https://​www.​disgenet.​org/​dbinfo.

Gene–gene association

To consider multiple layers of gene organization, we constructed a multilayer gene network from different sources.
Omnipath [45, 46] is a meta resource of a multitude of biological knowledge databases, and we curated a network by connecting two genes if a resource provides a co-membership for a signaling pathway. We used the number of resources that reported a relationship as an estimate for the confidence in the relationship, which we introduced as edge weights in the Omnipath layer.
The protein–protein interaction (PPI) network was constructed based on the union of publicly available data from experimental and literature curated data (HuRi-union [47]) [48].
Gene Ontology (GO) gene networks have been constructed before, and we used the GO networks constructed by [49].
Each gene network was reduced to remove loops and multiple edges. To filter for genes relevant in cardiac tissue, gene networks were subset to genes expressed in the human heart on RNA or protein level. For protein expression, we used proteomic data [50, 51], where we selected all peptides that were detected in the human heart and used the leading gene associated with each peptide. For gene expression, we selected genes that were detected in the heart tissue in the Genotype-Tissue Expression (GTEx) Project v8 with a transcript per million value > 1 [52]. We chose this threshold to discard non-expressed genes but include lowly expressed genes. To ensure that gene programs only active in diseased hearts were also captured, we also included genes that were captured in a meta-analysis of HF transcriptomes [32, 33].

Disease–gene prediction and prioritization

To predict genes from diseases within the HFhetnet, we relied on a network propagation algorithm developed for multilayer networks (random walk with restart on multiplex heterogeneous networks; RWR-MH [53]). This algorithm is an extension of the random walk algorithm that tries to find a stationary distribution of probabilities that a node is visited when a random walk on the network is initiated in a set of seed nodes.
We assessed the performance of the link prediction task within the HFhetnet. For a given disease that was present in the HFhetnet and directly linked to two or more genes, we attempted to predict those genes after removing the direct links from the HFhetnet and running RWR-MH with the disease as seed node. The position of the target genes in the resulting probability ranking was then assessed with multiple metrics to estimate success of disease gene recovery (Additional file 1: Supplementary Methods).
For HFpEF and HFrEF gene prediction, we applied the RWR-MH, using the comorbidity profiles from the patient classifier as the seed nodes in the HFhetnet. This yielded two vectors of RW probabilities for each comorbidity profile. The top 500 genes yielded non-zero probability values for each profile. To select gene candidates that were differently ranked, we calculated a prioritization score for HFpEF and HFrEF. For this, we calculated \(G_i= {P_i}*|\Delta R_i|\).
G is the gene prioritization score, P is the RW-based probability, ΔR is the rank difference between HFpEF and HFrEF rankings for gene i.

Transcriptome analysis

We filtered lowly expressed genes and normalized samples using the Trimmed mean of M-values (edgeR [54]) and subsequent variance-stabilizing transformation (limma voom) and performed differential expression analysis (limma [55]. We performed principal component analysis and Gene Ontology enrichment with the enrichr [56] R package. For the overrepresentation analysis, we ranked genes by t-statistic and performed gene set enrichment analysis (fgsea R package [57]) of the top predicted HFpEF and HFrEF genes using different cut-offs.

Results

The study population

The study population consisted of 29,047 patients with HF (Fig. 1). Within this cohort, we identified three sub cohorts, HFpEF (8062 patients), HFrEF (6585 patients), and HFmrEF (3018 patients) based on LvEF. LvEF was not recorded in 11,382 HF patients, preventing subcohort labeling (i.e., unlabelled HF cohort). HFpEF patients were more often female compared to HFrEF patients (35 vs 25%, p < 0.01) (Table 1). However, we did not observe a significant difference in body mass index (median [IQR] = 26.8 [24.2, 30.0] vs 26.5 [24.1, 30.1] for HFpEF vs HFrEF, p = 0.9) or age (median [IQR] = 70 [61, 88] for HFpEF vs 70 [60, 70] for HFrEF, p = 0.5). When phenotypic data were available, cholesterol, LDL, HDL, and blood pressure values were higher in HFpEF patients compared to HFrEF, while NT-proBNP values were higher in HFrEF patients. Comorbidity burden measured by Elixhauser index was slightly lower in HFpEF than HFrEF patients, as previously reported [58]. HFpEF patients were intubated (8.5% vs 15%, p < 0.001) or received an implantable cardioverter-defibrillator (16% vs 26%, p < 0.001) less frequently than HFrEF patients, suggesting that the HFrEF cohort be a later stage of HF.
Table 1
Clinical characteristics of HFrEF, HFmrEF, and HFpEF cohorts. Descriptive statistics of HFrEF, HFmrEF and HFpEF cohorts. F female, m male, BMI body mass index, BP blood pressure, LDL low-density lipoprotein, HDL high-density lipoprotein, ICD implantable cardioverter defibrillator, PCI percutaneous coronary intervention, NT-BNP N-terminal pro b-type natriuretic peptide. All numerical values are median (IQR), Elixhauser index is mean (SD)
   
HF subtypes
  
Variable
N
Overall
HFpEF
HFmrEF
HFrEF
p-value*
p-value+
  
(N = 17,665)
(N = 8062)
(N = 3018)
(N = 6585)
  
Sex
17,617
    
 < 0.001
 < 0.001
 Female
 
5247 (30%)
2822 (35%)
790 (26%)
1635 (25%)
  
 Male
 
12,370 (70%)
5228 (65%)
2218 (74%)
4924 (75%)
  
Age (years)
17,665
70 (60, 78)
70 (61, 78)
70 (59, 77)
70 (60, 78)
0.093
0.5
BMI
9132
26.8 (24.2, 30.0)
26.8 (24.2, 30.0)
26.9 (24.2, 29,9)
26.5 (24.1, 30.1)
0.08
0.9
Systolic BP (mmHg)
5146
148 (134, 160)
150 (139, 164)
148 (135, 160)
140 (127, 154)
 < 0.001
 < 0.001
Diastolic BP (mmHg)
5146
84 (76, 92)
85 (78, 93)
84 (76, 93)
82 (73, 90)
 < 0.001
 < 0.001
LDL (mg/dL)
12,270
87 (69, 110)
88 (69, 110)
91 (72, 113)
84 (67, 106)
 < 0.001
 < 0.001
HDL (mg/dL)
12,368
44 (36, 54)
46 (38, 56)
43 (36, 53)
40 (34, 50)
 < 0.001
 < 0.001
Triglycerides (mg/dL)
13,859
112 (85, 153)
112 (85, 151)
112 (85, 156)
113 (85, 156)
0.11
0.006
Cholesterol (mg/dL)
13,577
160 (135, 188)
163 (138, 190)
164 (140, 192)
153 (129, 183)
 < 0.001
 < 0.001
N PheCodes
17,665
13 (8, 21)
13 (9,21)
12 (7, 19)
14 (9, 22)
 < 0.001
0.088
Elixhauser Index
17,665
5.39 (2.72)
5.36 (2.68)
5.09 (2.70)
5.56 (2.76)
 < 0.001
 < 0.001
Intubated
17,665
1766 (10.0%)
552 (6.8)
257 (8.5%)
957 (15%)
 < 0.001
 < 0.001
ICD Implantation
17,665
3213 (18%)
1007 (12%)
468 (16%)
1738 (26%)
 < 0.001
 < 0.001
PCI
17,665
9116 (52%)
4267 (53%)
1554 (51%)
3295 (50%)
0.002
 < 0.001
log (NT-BNP)
6169
2.99 (2.45, 3.53)
3.07 (2.53, 3.55)
3.07 (2.53, 3.55)
3.45 (2.96, 3.88)
 < 0.001
 < 0.001
All continuous values displayed as median (IQR) except for Elixhauser index which is mean (SD). All dichotomous values displayed as N (%)
*Kruskal–Wallis p-value across all subtypes
+Wilcoxon–rank sum or chi-squared p-value for HFpEF vs HFrEF

High variation in comorbidity profiles is associated with HFpEF/HFrEF subtype

We expected differences in the composition of comorbidity profiles between HF subtype cohorts. To quantify this variance, we applied MCA and estimated the variance associated with sub-cohort labels and clinical features (Fig. 2A). Device implantation was the feature most strongly associated with variance in comorbidity profiles (Fig. 2B). When comparing HF subtypes, HFpEF and HFrEF cohort labels were associated with a high degree of explained variation (39.5%). HFmrEF patients seemed to be in an intermediate state, as they displayed lower variance when compared to HFpEF (25.2%) and HFrEF (18.6%). Sex and age were each associated with high variance (37.9% and 44.4%, respectively) as expected. In summary, this analysis approach identified a pronounced contrast between comorbidities in HFpEF and HFrEF patients.

Distinctive comorbidity profiles derived for HFpEF and HFrEF

Next, to explain and interpret the variance between HFpEF and HFrEF, we derived distinct comorbidity profiles for both cohorts. For this purpose, we fit random forest and elastic net classifier models with the 569 comorbidities as predictors to distinguish between HFpEF and HFrEF (Fig. 2A, Additional file 1: Fig. S2A,B). The highest achieved AUROCs were 0.778 for the random forest and 0.777 for the elastic net model, indicating that the random forest’s ability to model more complex interactions between comorbidities did not improve classifier performance substantially. The most important features were shared in both models (Additional file 1: Fig. S2C).
Next, because elastic net parameter estimates can provide both magnitude and direction, we selected the elastic net model to assign HFpEF and HFrEF a distinctive set of comorbidities. To select the most discriminant comorbidities, we performed forward selection. We found that the model with 100 comorbidities yielded a cross-validated AUROC of 0.780 (Additional file 1: Fig. S2D); 71 and 29 comorbidities from this model were assigned to HFpEF or HFrEF, respectively, which we will refer to as their comorbidity profiles.
These comorbidity profiles might be influenced by age, sex, time of visit, or time relative to HF diagnosis. We therefore investigated whether these factors influence the assignment of these 100 comorbidities to HF subtype by fitting a series of logistic regression models in different data subsets (Additional file 1: Supplementary Methods). We found that the derived comorbidity profiles of HFpEF and HFrEF yielded mostly consistent patterns independent of these factors (Additional file 1: Fig. S4).
The HFpEF profile (15 disease categories) was more diverse than the HFrEF profile (10 disease categories) and included comorbidities from the digestive disease, hematopoietic and neoplastic disease categories (Fig. 2C). Cardiovascular disease was the most important category in both profiles, accounting for 48.2% of the sum of parameter estimates in HFrEF and 38.3% in HFpEF. In HFpEF, important comorbidities included hypertensive and pulmonary heart disease, essential hypertension, inflammatory cardiac conditions (pericarditis, myocarditis), sleep apnea, osteopenia, neoplasms (multiple myeloma, breast cancer, metastasis in digestive systems), and rheumatoid disorders. The HFrEF comorbidity profile was characterized among others by myocardial infarction, ischemic heart disease, tobacco abuse, mitral valve disease, coma and cardiogenic shock, neurological disorders (vascular dementia, cerebral edema), chronic kidney disease, and diabetes type II (Fig. 2D, Additional file 1: Fig. S3).
In conclusion, the observed variation in comorbidity profiles between HFpEF and HFrEF was analyzed by interpreting patient classifiers. The derived features captured known subtype associations such as typical etiologies of HF including hypertensive heart disease (with HFpEF) and ischemic heart disease (with HFrEF) but also more novel and understudied comorbidities associated with HFpEF such as breast cancer or rheumatoid arthritis with HFpEF.

The HF comorbidity network (HFnet) captures HF specific disease relationships

To analyze patterns of disease co-occurrence in the HF-patient cohort, we constructed a comorbidity network as previously described [11, 12, 5961]). This network was built by calculating pairwise disease correlations for the general HF-patient cohort (Fig. 3A) (Additional file 1: Fig. S6A,B). The resulting significant disease–disease relationships were assembled to form an undirected and weighted HF comorbidity network (HFnet) consisting of 569 nodes and 19,347 edges (Additional file 1: Fig. S6C), with edge weights defined by a statistical dependency of co-occurrence for each disease pair.
While certain comorbidities were distinctive for HFpEF or HFrEF, it was unclear whether the disease relationships that built the HFnet also depended on the HF subtype (Fig. 3A). When comparing odds ratios for each disease pair from both cohorts, we found a high concordance (Additional file 1: Fig. S5A,B). Only 33 disease pairs had significantly different odds ratios between HFpEF and HFrEF (Breslow–Dayes test with BH correction p < 0.01) (Additional file 1: Fig. S5C), suggesting that in the vast majority of cases, the co-occurrence of two diseases did not depend on whether it was assessed in HFpEF or HFrEF patients.
Whether the HFnet constituted a unique wiring of diseases or predominantly captured generalizable disease relationships was unclear. To investigate this, we analyzed and compared two additional disease networks: a human phenotype ontology network (HPOnet), where two diseases are connected if they are phenotypically similar, and Morbinet [11], another comorbidity network from a large patient cohort but without a cohort defining disease (Additional file 1: Supplementary Methods).
Jaccard index-based edge similarity of HFnet and Morbinet was 0.18 and of HFnet and HPOnet was 0.12 (Additional file 1: Fig. S7A,B,C). We then calculated network similarities with the DeltaCon algorithm to capture conserved node affinities between networks [28]. HFnet and Morbinet displayed again a higher similarity (0.46) than HFnet and HPOnet (0.39) (Additional file 1: Fig. S7D). This suggested that comorbidity correlation was not completely redundant with phenotype similarity. The differences between Morbinet and HFnet indicated that many disease relationships in the HFnet could be specific for HF patients.
Finally, we analyzed the centrality of diseases. Diseases which were most frequently reported could be considered the network hubs, as indicated by their high node degree and their closeness and betweenness centrality scores (Additional file 1: Fig. S8A). Our network captured well-known HF comorbidities [41, 42], like chronic kidney disease, which by multiple metrics was the main HFnet hub (Additional file 1: Fig. S8B, Additional file 3: Supp. Table 2). We found that closeness and degree centrality were both significantly associated with the disease category (Additional file 1: Fig. S8C) (Kruskal–Wallis p < 0.01). Infectious and hematopoietic diseases had the highest median centrality scores (betweenness, closeness, and degree), indicating that patients with diseases from these categories were typically suffering from many comorbidities. Diseases affecting the circulatory system had the highest prevalence as was expected with a HF centered cohort (Additional file 1: Fig. S8C).
In summary, we found that comorbid relationships were mostly independent of the HF subtype. However, when comparing with other disease networks, many comorbid relationships were specific to the HFnet. This indicated that the constructed HFnet captured disease patterns relevant for HF patients, which only in part can be generalized to other cohorts.

The HFnet contains 9 disease clusters that represent important comorbidity groups in HF

Network communities represent densely connected subgraphs and can be helpful to summarize network topology. Network clustering algorithms identified nine disease clusters (DCs) (Additional file 1: Supplementary Methods, Fig. S9A-F). DCs were partially grouped by disease categories (Fig. 3B, Additional file 4: Supp. Table 3) and we labeled DCs by manually reviewing disease composition (Table 2). For instance, DC1 and DC3 contained the majority of cardiovascular diseases. While DC1 contained cardiovascular diseases with vascular etiology (EH, CAD, MI) and included metabolic and endocrine diseases, DC3 contained valve disorders and arrhythmias (Fig. 3C).
Table 2
Overview of disease clusters. Manual labeling of disease clusters (DC) by characterizing most central and prevalent diseases in each cluster. EH essential hypertension, MI myocardial infarction, COPD chronic obstructive pulmonary disease, CAD coronary artery disease, DM II diabetes mellitus type II, CKD chronic kidney disease, RA rheumatoid arthritis
DC
Label
Important nodes
DC1
Cardiac/endocrine/respiratory diseases
EH, MI, COPD, hyperlipidemia, hypothyroidism, CAD
DC2
Sensory/ophthalmologic/skin disease
Cataract, macular degeneration, melanomas of skin
DC3
Cardiovascular disease with heart focus
Valve disease, congenital anomalies, arrhythmias
DC4
Vascular/renal/diabetic diseases
DM II, CKD, atherosclerosis
DC5
Critical illness/complications
Infectious disease, organ failures
DC6
Rheumatoid/osteological/psychiatric diseases
Osteoporosis, osteopenia, RA, depression
DC7
Gastroenterological diseases
Gastritis, diverticulitis, cirrhosis
DC8
Neoplastic/hematopoietic diseases
Breast cancer, aplastic anemia, lymphomas
DC9
Neurological/vascular neurological diseases
Stroke, dementias, epilepsy
We hypothesized that DCs represent facets of the subcohort specific HF comorbidity spectrum, and we therefore tested whether DCs capture demographic or HF subtype-related characteristics. We quantified the similarity of an individual patient's comorbidity profile with each DC by calculating Jaccard indices and tested for differences between patient cohorts (Fig. 3D). In age-stratified analyses, we found that all DCs, except DC7, were more similar to 60–80-year-old (n = 16,54) compared to 40–59-year-old patients (n = 5973) comorbidity profiles. This could indicate a general increase of comorbidity burden with age or that with age come increasingly consistent comorbidity profiles between individuals. The 80 + cohort (n = 6,527) had less similarity with DC1 and significantly more similarity with DC3, DC5, and DC9 profiles compared to 40–60-year-old patients. When comparing female and male patients, we found that DC6 and DC2 yielded the highest similarity differences, respectively. Comparing HFpEF with HFrEF patients, we found that DC1, DC2, DC6, and DC8 were more similar to HFpEF patients, while DC3, DC4, and DC5 were suggested to be similar to HFrEF patients. As DC1 and DC6 also captured sex-related comorbidity differences, we investigated further, whether DC6 diseases were more prevalent in HFpEF independent of sex. For this, we fit logistic regression models for each disease predicting HFpEF/HFrEF while controlling for sex (Additional file 1: Fig. S9G). Again, DC1, DC2, and DC6 contained more diseases prevalent in HFpEF while DC3, DC4, and DC5 diseases were more prevalent in HFrEF. In addition, this analysis also suggested that many diseases in DC7 and DC8 too were distinctive for HFpEF.
We further compared the comorbidity profiles from the patient classifier by mapping them to DCs which yielded a qualitatively similar DC to HF subtype association (Additional file 1: Fig. S9H). No DC was positively associated with HFmrEF. Instead, HFmrEF patients were less similar to DC1 and DC6 than HFpEF patients and less similar to DC3, DC4, and DC5 than HFrEF patients.
In general, we found that aggregating comorbidity profiles (569 dimensions) to DC similarity (9 dimensions) allowed us to capture differences among patient cohorts in regard to sex, age, and HF subtype in meaningful disease groups.

Building the HF heterogeneous network (HFhetnet)

Biomedical research has yielded significant knowledge of disease gene associations, which can be harnessed to extrapolate novel disease gene relationships. HFpEF is a comorbidity-driven syndrome and we hypothesized that the identified HFpEF comorbidity profile could be translated to a genetic profile consisting of recurrent genetic associations to these comorbidities. In this part of our study, we first integrated multiple biomedical databases to construct a cardiac specific multilayer disease and gene network. We then estimated the success of this network to recover known disease–gene associations and, finally, used the HFpEF comorbidity profile to identify the most commonly associated genes.
To construct a gene network that reflected different hierarchies of gene function (i.e., pathway memberships, PPI, and ontological similarity), we integrated multiple databases and represented gene–gene relationships as networks (“Methods”) (Fig. 4A). To focus on genes relevant in cardiac tissue, we subset the resulting gene networks to protein coding genes expressed in the heart (Additional file 1: Fig. S10A). Next, we used DisGeNET, a resource containing disease–gene associations, to connect the HFnet with the gene network. We connected 400 diseases of the HFnet with a total of 4044 genes via 20,170 edges. As the HPOnet constructed earlier had a small intersection with the HFnet and captured a different type of disease relationship, it was included as an additional disease layer in our network.
The presented HFhetnet is an assembly of the data-driven comorbidity relationships (HFnet) and six biomedical databases resulting in a total of 13,572 nodes and 181,529 edges (Additional file 5: Supp. Table 4). Its main structure is set up by two biological networks (disease layer and gene layer) that each consist of two or four network layers, respectively (Fig. 4A). The two disease networks were the smallest when comparing node numbers (Fig. 4B). However, edge density was much higher resulting in centralization of these networks compared to the gene layers. Within gene layers, the ontological layers displayed the highest transitivity, as well as tendency to connect to hub genes (degree assortativity). To assess research bias in the gene networks, we calculated Pearson correlation between the number of abstracts in PubMed mentioning a gene and the gene’s network degree per layer and found that only the pathway layer (Omnipath) displayed significant correlation (p-value < 0.05). This is related to a biomedical research bias towards the investigation of a small number of genes [62]. Thus, the integration of experimental and ontological data can ameliorate the centrality of overstudied genes.
In summary, we constructed the HFhetnet by integrating various prior knowledge resources to incorporate genetic information. The different network layers of the HFhetnet captured unique node relationships and displayed particular network topologies.

Estimating the success of disease–gene prediction within the HFhetnet

To estimate the potential of the HFhetnet to predict disease–gene relationships, we estimated the success of predicting known disease genes. The rationale behind this approach is the guilt-by-association principle that assumes that functionally related genes are also associated in the network context. Extending this notion to heterogeneous networks, this principle can be interpreted as a disease being associated with relevant disease genes through its position in the network. To quantify this property, we applied a leave-one-out validation design to assess whether known disease genes can be recovered after removing the direct edges that connected them to a disease. After edge removal, the gene recovery was performed by applying the RWR-MH algorithm which considers each network layer and its topology ( Additional file 1: Supplementary Methods).
We performed this analysis by comparing the impact of three variations of the disease layer: (i) HFnet + HPOnet (original HFhetnet), (ii) only HFnet, and (iii) a rewired HFnet. Gene prediction worked best in the original HFhetnet (median AUROC 0.91, median AUC-PR 0.07, and median rank ratio 0.03) (Fig. 4C). This performance dropped for every metric when removing the HPO layer or when rewiring the HFnet (paired, two-sided Wilcoxon’s rank sum test p < e − 10). The rewired HFnet still performed better than random, which might be explained by (i) high edge density in the HFnet and (ii) the large size of the unaltered gene–gene and disease–gene network in comparison to the smaller HFnet.
Prediction success correlated weakly but significantly with gene set size (Additional file 1: Fig. S10C). In addition, neither disease prevalence nor DisGeNET confidence scores were significantly correlated with prediction success, suggesting that frequent diseases could not be predicted better than less frequent diseases. Prediction performance depended on disease category (Additional file 1: Fig. S10D) (Kruskal–Wallis test p-value < 0.01 for all metrics) with respiratory, neurological, genitourinary, and cardiovascular diseases performing best.
In summary, we found that within the HFhetnet, the disease genes remained close via the disease’s connection through its comorbidities or phenotypically similar neighbors. Thus, we concluded that HFnet and its extension, HFhetnet, captured meaningful disease–disease, disease–gene, and gene–gene relationships, which can be exploited for predicting a disease’s genetic profile through its comorbidities.

Predicting genes associated with comorbidity profiles of HFpEF and HFrEF

In the first part of our study, we found that HFpEF and HFrEF patients were distinguishable based on their comorbidity profiles. We then demonstrated that diseases within the HFhetnet were located in network proximity to their respective disease–genes. Leveraging both insights, we hypothesized that genes located close to the HFpEF and HFrEF comorbidity profiles could yield novel candidates for the respective HF subtype. In this section, we applied the RWR-MH algorithm with the HFpEF and HFrEF comorbidity profiles as seed nodes resulting in gene ranking based on network proximity (Additional file 1: Fig. S11A-C).
To assess whether the resulting gene rankings recapitulated known HF genes, we curated a set of HF-related genes from various prior knowledge sources and independent datasets (Additional file 1: Supplementary Methods), which had only little intersection (Additional file 1: Fig. S11D). We found that prior knowledge gene sets were well recovered within the HFpEF and HFrEF gene rankings (Fig. 5A, Additional file 1: Fig. S11E). Gene sets that were retrieved from experimental data (gene expression, PheWAS, GWAS) performed worse in the predictions. Next, we compared these prediction results with random comorbidity profiles and found that the HFrEF profile associated with Kegg’s dilated cardiomyopathy (DCM) (z-score AUROC 1.77; z-score PR-AUC 6.7) and DisGeNETs HF genes (z-score AUROC 1.76; z-score PR-AUC 2.46) (Additional file 1: Fig. S11F). This indicated that the HFrEF comorbidity profile which was more cardiac centered was closer to prior knowledge of HF genes within the HFhetnet. In general, well-known genes relevant for HF were recovered for both, HFpEF and HFrEF comorbidity profiles including NPPA, NPPB, TNFa, NOS2, NOS3, CCL2, IL1B, LMNA, and TTN (Additional file 1: Fig. S11D).
To emphasize genes that might be HFpEF specific, we prioritized genes that were close to the HFpEF but not to the HFrEF comorbidity profile within the HFhetnet (Fig. 5B, Additional file 6: Supp. Table 5, Additional file 1: Supplementary Methods). We found that MMP1, MHY7, and DAPK1 received the highest scores and other candidates included genes functionally involved in fibrosis (e.g., LOX), metabolism (MVK), transcriptional regulation (ATF6), coagulation (THBD), and oxidative stress (NOS1, XDH) (Table 3).
Table 3
Potential HFpEF candidates based on network proximity to comorbidity profiles. Genes are sorted by functional groups. Rank indicates prediction rank for HFpEF. TF transcription factor, CM cardiomyopathy
Rank
Gene symbol
Gene name
Functional group
Gene function
Role in HF (exemplary or putative)
References
17
PCSK5
Proprotein convertase subtilisin/kexin type 5
Cell differentiation
Mediates post translational endoproteolytic processing
Cleaves GDF1, heart development
[63, 64]
9
NKX2-5
NK2 Homeobox 5
Cell differentiation
TF expressed in precursor cardiac cells, involved in cardiac development
Heart development, activated in HF involved in hypertrophy
[65, 66]
31
GATA5
GATA binding protein 5
Cell differentiation
TF, involved in embryonic development of the heart
Possible role in cardiac hypertrophy, linked to dilated CM
[67]
42
GATA3
GATA binding protein 3
Cell differentiation
TF, involved in embryonic development of the heart and immune cell differentiation
Role in T-cell recruitment
[68, 69]
67
JAG1
Jagged canonical Notch ligand 1
Cell differentiation
Interacts with four receptors in the mammalian Notch signaling pathway
Possible protective role against PAH and diabetic CM, involved in regenerative capacity of the heart
[7072]
62
NOTCH1
Notch receptor 1
Cell differentiation
Regulates interactions between physically adjacent cells through binding of Notch family receptors to their cognate ligands
Possible protective role against PAH and diabetic CM, involved in regenerative capacity of the heart
[7072]
13
CITED2
Cbp/p300 interacting transactivator with Glu/Asp rich carboxy-terminal domain 2
Cell differentiation
TF that controls pluripotency among others
Cardiomyocyte pluripotency
[73]
23
KLKB1
Kallikrein B1
Coagulation
Factor XII activation
Interacts with LDL, possible HF biomarker
[7476]
27
TBXA2R
Thromboxane A2 receptor
Coagulation
G protein-coupled thromboxane A2 receptor that induces platelet aggregation and regulate hemostasis
Involved in endothelial homeostasis, angiogenesis
[7779]
34
THBD
Thrombomodulin
Coagulation
Endothelial-specific type I membrane receptor that binds thrombin
Angiogenesis and anticoagulant, possible HF biomarker
[80, 81]
11
DAB2IP
DAB2 interacting protein
Endothelial dysfunction
Ras GTPase-activating protein (GAP)
Associated with endothelial dysfunction in atherosclerosis
[82, 83]
77
ESR1
Estrogen receptor1
Endothelial dysfunction
Estrogen receptor and ligand-activated TF
Regulates NOS activity among many other effects
[84]
7
ATF6
Activating transcription factor 6
ER-stress
Unfolded protein response
Activates XBP1 which modulates ER-stress
[8587]
3
MMP1
Matrix metallopeptidase 1
Fibrosis
Proteases involved in degrading of ECM
involved in cardiac fibrosis
[88]
25
FGFR2
Fibroblast growth factor receptor 2
Fibrosis
Receptor tyrosine kinases that promote mitogenic signal mediators that induce cell proliferation and survival
Involved in cardiac fibrosis
[89]
33
LOX
Lysyl oxidase
Fibrosis
Cross-linking of collagens
Possible role in myocardial stiffness
[90]
2
MYH7
Myosin heavy chain 7
Hypertrophy
Part of the thick filament in cardiac muscle, involved in contraction
Cardiac hypertrophy marker
[91, 92]
4
SUZ12
SUZ12 polycomb repressive complex 2 subunit
Hypertrophy
Core PRC2 (polycomb repressive complex 2) protein
Mediates long non-coding RNA Ahit induced cardiac hypertrophy
[93]
65
FSTL1
Follistatin like 1
Hypertrophy
Extracellular glycoprotein
Protective against hypertrophy
[94, 95]
79
RGS5
Regulator of G protein signaling 5
Hypertrophy
RGS proteins are involved in the regulation of heterotrimeric G proteins by acting as GTPase activators
Protects from cardiac hypertrophy and fibrosis
[96, 97]
89
CD14
Cluster of differentiation 14
Inflammation
Membrane glycoprotein primarily expressed by myeloid cells that plays a key role in inflammation
Monocytes/macrophages marker in immune disease
[98]
47
SHARPIN
SHANK associated RH domain interactor
Inflammation
Displacing talin from the integrin cytoplasmic domain
Involved in integrin Inactivation and NF-κB Signaling
[99, 100]
90
CD209
Cluster of differentiation 209
Inflammation
Expressed by macrophages and dendritic cells
Involved in rheumatoid arthritis
[101]
36
MVK
Mevalonate kinase
Metabolism
Cholesterol metabolism
Fibrotic effects, Rho family small GTPase activity modulation
[102, 103]
1
DAPK1
Death associated protein kinase 1
Oxidative stress
Calcium/calmodulin-dependent serine/threonine kinase
Protection from oxidative stress in MI
[101]
18
XDH
Xanthine dehydrogenase
Oxidative stress
Oxidative metabolism of purines
Linked to nitroso-redox balance, also possible plasma marker
[104]
69
NOS1
Nitric oxide synthase 1
Oxidative stress
Synthesizing nitric oxide from L-arginine
Inhibition possibly linked to protection of diastolic dysfunction
[105, 106]
14,19
GSTZ1, GSTT1
Glutathione-S-transferases
Oxidative stress
Catalyzing the conjugation of the reduced form of glutathione to xenobiotic substrates for the purpose of detoxification
Related to oxidative stress in cardiac tissue
[107, 108]

Corroboration of HFpEF gene candidates in the transcriptome of a murine HFpEF model

After translating the comorbidity profile of HFpEF patients to an associated genetic profile, the functional relevance of this predicted profile remained unclear. We hypothesized that the relevance of the gene predictions could be suggested by transcriptional dysregulation in myocardial tissue of HFpEF. However, human molecular data of HFpEF is sparse and mechanistic insights are typically derived from mouse models [3]. Hence, we collected myocardial bulk transcriptomics from a murine HFpEF model, induced via high-fat diet and nitric oxide synthase inhibitor N[w]-nitro-l-arginine methyl ester (L-NAME) treatment [36] (Fig. 5C, Additional file 1: Supplementary Methods).
First, we confirmed important HFpEF phenotype characteristics including preserved ejection fraction, increased body weight, blood glucose levels, and blood pressure as well as diastolic dysfunction (increased E/e') (nCT = 4, nHFpEF = 4, Student’s t-test, p < 0.05, Additional file 1: Fig. S12).
Second, we assessed transcriptomic changes in HFpEF via differential gene expression analysis (Additional file 1: Fig. S13A-D). Upregulated genes confirmed processes involving fibrosis and metabolic stress (Additional file 1: Fig. S13C, D).
After confirming the phenotypic and molecular resemblance of the HFpEF model, we investigated gene expression dysregulation of the comorbidity-based gene profiles by performing enrichment analysis of the HFpEF and HFrEF gene predictions (Fig. 5D, Additional file 1: Fig. S13E). We found that the top 50 to 100 predicted HFpEF genes displayed significant enrichment in overexpressed genes in the murine HFpEF model, while the HFrEF predicted genes were not enriched (p-value < 0.05, Fig. 5E, Additional file 1: Fig. S13E). Fibrosis-related genes like LOX, SMAD9, and PTHL and hypertrophy-related genes like GATA5, GATA3, and MYH7 could be recovered together with XDH, among others. This suggested that the genetic profile derived from human HFpEF comorbidities associated with relevant gene expression dysregulation during myocardial remodeling in murine HFpEF.

Discussion

In this study, we provided a retrospective systems level analysis of comorbidities in a large cohort of HF patients. We derived clinically relevant insights by comparing comorbidity profiles between HFpEF and HFrEF patients and biological insights by defining genes associated with HFpEF and HFrEF comorbidity profiles.
Patient clustering has been previously shown to yield novel subgroups of HFpEF defined by multivariate similarity [109111]. In contrast, the clustering of features (i.e., comorbidities) can inform about patterns of co-occurring disease groups. Our study demonstrated that this approach can be useful to interpret comorbidity profiles: the aggregation of co-occurrence patterns of diseases can help to organize illness into different levels of clinical concepts like organs (DC7—gastrointestinal tract), illness severity (DC5—intensive care), or disease categories (DC8—cancer). This aggregation via network clustering may also reduce multiple testing burdens and provide insights into the relevance of low prevalence diseases where comparisons for a single disease may be problematic.
In the patient classifier, HFpEF was characterized by a larger number of comorbidities with lesser emphasis on cardiac disorders. This supports the hypothesis of HFpEF as a comorbidity-driven systemic syndrome [112, 113]. We found that hypertensive heart disease was the most discriminant feature for HFpEF, which has been viewed as a major etiology for diastolic HF involving cardiac hypertrophy and myocardial stiffness [114, 115]. In contrast, ischemic etiologies including myocardial infarction characterized HFrEF consistent with other studies [116].
We identified more novel disease associations with HFpEF such as neoplastic diseases including breast cancer. HF related hospitalizations in breast cancer survivors have been recently associated more with HFpEF than with HFrEF [117], though the reasons for this remain incompletely elucidated [118]. The association to other cancerous diseases remains largely unexplored and should be addressed in future studies. Another interesting aspect of the HFpEF comorbidity profile was the high similarity to DC6, which contained rheumatic, osteologic, and mental diseases. Systemic inflammatory diseases could be a driving factor for HFpEF and rheumatic disease could constitute a pathophysiologic linkage [112, 119121]. Bone mineralization also has been reported to be lowered in HFpEF patients [122] and is a symptomatic link to postmenopausal endocrinology [123]. While mental health has been studied in the context of HF extensively, differences between HFpEF and HFrEF are largely unexplored. The joint clustering of these disease complexes and their similarity to female patients provides a potential link between female sex and HFpEF. Future work should further explore these relationships.
HFpEF and HFrEF clearly displayed distinguishable comorbidity profiles. By contrast, HFmrEF, introduced as a unique form of HF in 2016 [124], appeared to be a combination of attributes from HFrEF and HFpEF. Thus, from the comorbidity perspective, it may be a transitional state instead of a unique syndrome as suggested before [125].
We predicted an associated genetic profile from data-driven HFpEF comorbidity profiles. This genetic profile indicates that HFpEF comorbidities are associated with recurrent patterns of genes involved in fibrosis, inflammation, cell differentiation, metabolism, and oxidative stress. As an example, the glutathione-S-transferases, NOS1 and Xanthine dehydrogenase (XDH), were identified by our network. XDH catalyzes the rate limiting step in purine metabolism producing uric acid [126] and previous literature supports both the role of serum uric acids in HF [104] and plasma XDH activity as relevant for adverse clinical outcomes in HFpEF [127]. Nitric oxide synthase (NOS) has been proposed to contribute to endothelial dysfunction in HFpEF [105, 106], and NOS1 inhibition was recently associated with recovery of diastolic dysfunction in a murine model resembling HFpEF [128]. Glutathione-S-transferases (GSTM1, GSTT1, GSTZ1) are antioxidant enzymes and polymorphisms of these genes have been reported as potentially relevant to HF and diastolic dysfunction [107, 108]. This group of genes could constitute crucial gene candidates involved in comorbidity-based HFpEF pathophysiology.
In general, HFpEF is likely to be a disease in which multiple genes and pathways contribute to the spectrum of phenotypes. Therefore, instead of using the disease–gene prediction to identify and validate individual genes, we have corroborated the overall effect of a spectrum of identified genes in murine gene expression data. While this provided additional evidence for the relevance of comorbidity-based gene prediction, further experimental validation is necessary to explore the functional role and reproducible validity of candidate genes. In real-world populations, it is likely that the genetic heterogeneity of the HFpEF syndrome will be influenced by the specific comorbidities that are well represented in each population. In previous disease–gene prediction studies, gene prediction was performed either by selecting multiple seed genes or single seed diseases [129, 130]. We propose that our approach for gene inference based on data-driven comorbidity profiles might be suitable for systemic syndromes where multimorbidity plays an important role like HF and especially HFpEF.
In addition, several data resources were generated in this study: (i) the HFpEF gene predictions, (ii) HFhetnet, and (iii) murine HFpEF transcriptome to help facilitate future efforts to understand HFpEF-related pathophysiology and benefit the research community.
This study had important strengths and was subject to several limitations. An important strength of this study is that we analyzed clinical care data, which is a real-world representation of patients and therefore allowed us to perform a data-driven analysis of comorbidities in this patient population. However, as a result, this analysis is limited to the information captured (i) in our hospital system and (ii) at the hospital visits of a patient. Therefore, obtained results could be subject to some common biases found in medical record-derived data, such as non-random interaction with the health care system resulting in some patient populations having more data than others [131], incomplete documentation [132], selection bias [133], missing data, and lack of documentation of potential confounders [134]. In addition, possible non-observed confounders like socioeconomic status or health-related behavior could not be taken into account due to lack of documentation in the medical record. We determined subtypes using LvEF, which can be error prone [135] and might not fully provide a sufficient criterion for the HFpEF diagnosis [136]. Patients with more serious conditions will tend to visit a tertiary health care provider more often and thus could be overrepresented. In our study, at a tertiary care center with a focus on cardiomyopathy, this seemed to affect the contrast between HFrEF and HFpEF, as HFrEF patients had higher intubation prevalence and DC 5 similarity. This may also have contributed to differences between this study population and other reports of HFpEF population characteristics. However, given the known heterogeneity of HFpEF and HFrEF [109111], we believe these differences are plausible and a more granular approach to study HFpEF subtypes could be necessary to address inconsistent patient characteristics [137]. Another limitation of our study is the use of ICD-10 codes to capture comorbidities. Different ICD-10 codes are known to have different predictive value for disease, and therefore, it is likely that some diseases are over- or underrepresented in our data [138]. Moreover, we performed a cross-sectional analysis and therefore did not consider the timing and sequence of comorbidities when generating comorbidity profiles.
Given these limitations, future studies are necessary to address the generalizability of our findings to other HF populations and to delineate different disease trajectories by considering the time of events. Nevertheless, our study recapitulated known HF comorbidity patterns, as discussed above, that could substantiate more novel comorbidity patterns identified in this work.
Many open questions remain regarding HFpEF pathophysiology and genetics [16]. Interdisciplinary and translational approaches are needed to account for the cross-organ disease involvement that is suggested to be critical in HFpEF. The increasing abundance of routine clinical care data and novel approaches like network medicine can provide novel insights and guidance for future experimental approaches.

Conclusions

In our study, we found evidence for greater diversity of comorbidity profiles in patients with HFpEF compared to HFrEF. We further identified nine co-occurring disease groups which capture differences of disease prevalence regarding age, sex, and HF subtype. Here, we find that multimorbidity in HFpEF extends to disease clusters beyond typical HF comorbidities and includes rheumatoid, neoplastic, and gastrointestinal diseases. We further provided a biological interpretation of the HFpEF comorbidity profile, capturing overexpressed gene programs observed in murine HFpEF models. Oxidative stress, hypertrophy, cell differentiation, and fibrosis-related genes are recurrent patterns in genes associated to comorbidities of HFpEF and could constitute a link for the comorbid relationships of HFpEF resulting in a multiorgan disease state. Thus, our work highlights that comorbidity profiles are an important characteristic of HFpEF patients and should be incorporated into both clinical and genomic approaches to the study of HFpEF.

Acknowledgements

We thank Ricardio Ramirez-Flores and Jovan Tanevski for helpful discussions that shaped the analysis. Laura Wienecke provided valuable feedback on the manuscript.

Declarations

The study was conducted in accordance with the Declaration of Helsinki. The ethics committee of the University Hospital Heidelberg approved this study (approval ID: S-571/2019).
All authors provide consent for the publication of this manuscript.

Competing interests

JSR has received funding from GSK and Sanofi and consultant fees from Travere Therapeutics. AV is currently employed by F. Hoffmann-La Roche Ltd.
Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​. 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 in a credit line to the data.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Literatur
1.
Zurück zum Zitat Oktay AA, Rich JD, Shah SJ. The emerging epidemic of heart failure with preserved ejection fraction. Curr Heart Fail Rep. 2013;10:401–10.PubMedCrossRef Oktay AA, Rich JD, Shah SJ. The emerging epidemic of heart failure with preserved ejection fraction. Curr Heart Fail Rep. 2013;10:401–10.PubMedCrossRef
2.
Zurück zum Zitat Dunlay SM, Roger VL, Redfield MM. Epidemiology of heart failure with preserved ejection fraction. Nat Rev Cardiol. 2017;14:591–602.PubMedCrossRef Dunlay SM, Roger VL, Redfield MM. Epidemiology of heart failure with preserved ejection fraction. Nat Rev Cardiol. 2017;14:591–602.PubMedCrossRef
3.
4.
Zurück zum Zitat Pandey A, Vaduganathan M, Arora S, Qamar A, Mentz RJ, Shah SJ, et al. Temporal trends in prevalence and prognostic implications of comorbidities among patients with acute decompensated heart failure: the ARIC study community surveillance. Circulation. 2020;142:230–43.PubMedPubMedCentralCrossRef Pandey A, Vaduganathan M, Arora S, Qamar A, Mentz RJ, Shah SJ, et al. Temporal trends in prevalence and prognostic implications of comorbidities among patients with acute decompensated heart failure: the ARIC study community surveillance. Circulation. 2020;142:230–43.PubMedPubMedCentralCrossRef
5.
Zurück zum Zitat Simmonds SJ, Cuijpers I, Heymans S, Jones EAV. Cellular and molecular differences between HFpEF and HFrEF: a step ahead in an improved pathological understanding. Cells. 2020;9(1):242. Simmonds SJ, Cuijpers I, Heymans S, Jones EAV. Cellular and molecular differences between HFpEF and HFrEF: a step ahead in an improved pathological understanding. Cells. 2020;9(1):242.
6.
Zurück zum Zitat Palmiero G, Cesaro A, Vetrano E, Pafundi PC, Galiero R, Caturano A, et al. Impact of SGLT2 inhibitors on heart failure: from pathophysiology to clinical effects. Int J Mol Sci. 2021;22(11):5863. Palmiero G, Cesaro A, Vetrano E, Pafundi PC, Galiero R, Caturano A, et al. Impact of SGLT2 inhibitors on heart failure: from pathophysiology to clinical effects. Int J Mol Sci. 2021;22(11):5863.
7.
Zurück zum Zitat Maron BA, Altucci L, Balligand J-L, Baumbach J, Ferdinandy P, Filetti S, et al. A global network for network medicine. NPJ Syst Biol Appl. 2020;6:29.PubMedPubMedCentralCrossRef Maron BA, Altucci L, Balligand J-L, Baumbach J, Ferdinandy P, Filetti S, et al. A global network for network medicine. NPJ Syst Biol Appl. 2020;6:29.PubMedPubMedCentralCrossRef
8.
Zurück zum Zitat Hu JX, Thomas CE, Brunak S. Network biology concepts in complex disease comorbidities. Nat Rev Genet. 2016;17:615–29.PubMedCrossRef Hu JX, Thomas CE, Brunak S. Network biology concepts in complex disease comorbidities. Nat Rev Genet. 2016;17:615–29.PubMedCrossRef
9.
Zurück zum Zitat Khan A, Uddin S, Srinivasan U. Comorbidity network for chronic disease: a novel approach to understand type 2 diabetes progression. Int J Med Inform. 2018;115:1–9.PubMedCrossRef Khan A, Uddin S, Srinivasan U. Comorbidity network for chronic disease: a novel approach to understand type 2 diabetes progression. Int J Med Inform. 2018;115:1–9.PubMedCrossRef
10.
Zurück zum Zitat Guo M, Yu Y, Wen T, Zhang X, Liu B, Zhang J, et al. Analysis of disease comorbidity patterns in a large-scale China population. BMC Med Genomics. 2019;12(Suppl 12):177.PubMedPubMedCentralCrossRef Guo M, Yu Y, Wen T, Zhang X, Liu B, Zhang J, et al. Analysis of disease comorbidity patterns in a large-scale China population. BMC Med Genomics. 2019;12(Suppl 12):177.PubMedPubMedCentralCrossRef
11.
Zurück zum Zitat Aguado A, Moratalla-Navarro F, López-Simarro F, Moreno V. MorbiNet: multimorbidity networks in adult general population. Analysis of type 2 diabetes mellitus comorbidity. Sci Rep. 2020;10:2416.PubMedPubMedCentralCrossRef Aguado A, Moratalla-Navarro F, López-Simarro F, Moreno V. MorbiNet: multimorbidity networks in adult general population. Analysis of type 2 diabetes mellitus comorbidity. Sci Rep. 2020;10:2416.PubMedPubMedCentralCrossRef
12.
Zurück zum Zitat Hidalgo CA, Blumm N, Barabási A-L, Christakis NA. A dynamic network approach for the study of human phenotypes. PLoS Comput Biol. 2009;5: e1000353.PubMedPubMedCentralCrossRef Hidalgo CA, Blumm N, Barabási A-L, Christakis NA. A dynamic network approach for the study of human phenotypes. PLoS Comput Biol. 2009;5: e1000353.PubMedPubMedCentralCrossRef
13.
15.
Zurück zum Zitat Bousquet J, Anto JM, Sterk PJ, Adcock IM, Chung KF, Roca J, et al. Systems medicine and integrated care to combat chronic noncommunicable diseases. Genome Med. 2011;3:43.PubMedPubMedCentralCrossRef Bousquet J, Anto JM, Sterk PJ, Adcock IM, Chung KF, Roca J, et al. Systems medicine and integrated care to combat chronic noncommunicable diseases. Genome Med. 2011;3:43.PubMedPubMedCentralCrossRef
16.
Zurück zum Zitat Shah SJ, Borlaug BA, Kitzman DW, McCulloch AD, Blaxall BC, Agarwal R, et al. Research priorities for heart failure with preserved ejection fraction: national heart, lung, and blood institute working group summary. Circulation. 2020;141:1001–26.PubMedPubMedCentralCrossRef Shah SJ, Borlaug BA, Kitzman DW, McCulloch AD, Blaxall BC, Agarwal R, et al. Research priorities for heart failure with preserved ejection fraction: national heart, lung, and blood institute working group summary. Circulation. 2020;141:1001–26.PubMedPubMedCentralCrossRef
17.
Zurück zum Zitat Joseph J, Liu C, Hui Q, Aragam K, Wang Z, Charest B, et al. Genetic architecture of heart failure with preserved versus reduced ejection fraction. Nat Commun. 2022;13:7753.PubMedPubMedCentralCrossRef Joseph J, Liu C, Hui Q, Aragam K, Wang Z, Charest B, et al. Genetic architecture of heart failure with preserved versus reduced ejection fraction. Nat Commun. 2022;13:7753.PubMedPubMedCentralCrossRef
20.
Zurück zum Zitat Wu P, Gifford A, Meng X, Li X, Campbell H, Varley T, et al. Mapping ICD-10 and ICD-10-CM Codes to Phecodes: Workflow Development and Initial Evaluation JMIR Med Inform. 2019;7(4):e14325. Wu P, Gifford A, Meng X, Li X, Campbell H, Varley T, et al. Mapping ICD-10 and ICD-10-CM Codes to Phecodes: Workflow Development and Initial Evaluation JMIR Med Inform. 2019;7(4):e14325.
21.
Zurück zum Zitat Gasparini A. comorbidity: an R package for computing comorbidity scores. JOSS. 2018;3:648.CrossRef Gasparini A. comorbidity: an R package for computing comorbidity scores. JOSS. 2018;3:648.CrossRef
25.
Zurück zum Zitat Köhler S, Gargano M, Matentzoglu N, Carmody LC, Lewis-Smith D, Vasilevsky NA, et al. The human phenotype ontology in 2021. Nucleic Acids Res. 2021;49:D1207–17.PubMedCrossRef Köhler S, Gargano M, Matentzoglu N, Carmody LC, Lewis-Smith D, Vasilevsky NA, et al. The human phenotype ontology in 2021. Nucleic Acids Res. 2021;49:D1207–17.PubMedCrossRef
26.
Zurück zum Zitat Greene D, Richardson S, Turro E. ontologyX: a suite of R packages for working with ontological data. Bioinformatics. 2017;33:1104–6.PubMedCrossRef Greene D, Richardson S, Turro E. ontologyX: a suite of R packages for working with ontological data. Bioinformatics. 2017;33:1104–6.PubMedCrossRef
27.
Zurück zum Zitat Serrano MA, Boguñá M, Vespignani A. Extracting the multiscale backbone of complex weighted networks. Proc Natl Acad Sci USA. 2009;106:6483–8.PubMedPubMedCentralCrossRef Serrano MA, Boguñá M, Vespignani A. Extracting the multiscale backbone of complex weighted networks. Proc Natl Acad Sci USA. 2009;106:6483–8.PubMedPubMedCentralCrossRef
28.
Zurück zum Zitat Koutra D, Vogelstein JT, Faloutsos C. DELTACON: a principled massive-graph similarity function. arXiv:1304.4657. Koutra D, Vogelstein JT, Faloutsos C. DELTACON: a principled massive-graph similarity function. arXiv:1304.4657.
30.
Zurück zum Zitat Kanehisa M, Furumichi M, Sato Y, Kawashima M, Ishiguro-Watanabe M. KEGG for taxonomy-based analysis of pathways and genomes. Nucleic Acids Res. 2023;51:D587–92.PubMedCrossRef Kanehisa M, Furumichi M, Sato Y, Kawashima M, Ishiguro-Watanabe M. KEGG for taxonomy-based analysis of pathways and genomes. Nucleic Acids Res. 2023;51:D587–92.PubMedCrossRef
33.
Zurück zum Zitat Ramirez Flores RO, Lanzer JD, Holland CH, Leuschner F, Most P, Schultz J-H, et al. Consensus transcriptional landscape of human end-stage heart failure. J Am Heart Assoc. 2021;10: e019667.PubMedPubMedCentralCrossRef Ramirez Flores RO, Lanzer JD, Holland CH, Leuschner F, Most P, Schultz J-H, et al. Consensus transcriptional landscape of human end-stage heart failure. J Am Heart Assoc. 2021;10: e019667.PubMedPubMedCentralCrossRef
35.
Zurück zum Zitat Denny JC, Bastarache L, Ritchie MD, Carroll RJ, Zink R, Mosley JD, et al. Systematic comparison of phenome-wide association study of electronic medical record data and genome-wide association study data. Nat Biotechnol. 2013;31:1102–10.PubMedPubMedCentralCrossRef Denny JC, Bastarache L, Ritchie MD, Carroll RJ, Zink R, Mosley JD, et al. Systematic comparison of phenome-wide association study of electronic medical record data and genome-wide association study data. Nat Biotechnol. 2013;31:1102–10.PubMedPubMedCentralCrossRef
36.
Zurück zum Zitat Schiattarella GG, Altamirano F, Tong D, French KM, Villalobos E, Kim SY, et al. Nitrosative stress drives heart failure with preserved ejection fraction. Nature. 2019;568:351–6.PubMedPubMedCentralCrossRef Schiattarella GG, Altamirano F, Tong D, French KM, Villalobos E, Kim SY, et al. Nitrosative stress drives heart failure with preserved ejection fraction. Nature. 2019;568:351–6.PubMedPubMedCentralCrossRef
37.
Zurück zum Zitat Tong D, Schiattarella GG, Jiang N, May HI, Lavandero S, Gillette TG, et al. Female sex is protective in a preclinical model of heart failure with preserved ejection fraction. Circulation. 2019;140:1769–71.PubMedPubMedCentralCrossRef Tong D, Schiattarella GG, Jiang N, May HI, Lavandero S, Gillette TG, et al. Female sex is protective in a preclinical model of heart failure with preserved ejection fraction. Circulation. 2019;140:1769–71.PubMedPubMedCentralCrossRef
38.
Zurück zum Zitat Dobin A, Davis CA, Schlesinger F, Drenkow J, Zaleski C, Jha S, et al. STAR: ultrafast universal RNA-seq aligner. Bioinformatics. 2013;29:15–21.PubMedCrossRef Dobin A, Davis CA, Schlesinger F, Drenkow J, Zaleski C, Jha S, et al. STAR: ultrafast universal RNA-seq aligner. Bioinformatics. 2013;29:15–21.PubMedCrossRef
39.
Zurück zum Zitat Lê S, Josse J, Husson F. FactoMineR: An R package for multivariate analysis. J Stat Softw. 2008;25(1):1–18. Lê S, Josse J, Husson F. FactoMineR: An R package for multivariate analysis. J Stat Softw. 2008;25(1):1–18.
40.
41.
Zurück zum Zitat Wright MN, Ziegler A. ranger: a fast implementation of random forests for high dimensional data inC++ and R. J Stat Softw. 2017;77:1–17.CrossRef Wright MN, Ziegler A. ranger: a fast implementation of random forests for high dimensional data inC++ and R. J Stat Softw. 2017;77:1–17.CrossRef
42.
Zurück zum Zitat Chmiel A, Klimek P, Thurner S. Spreading of diseases through comorbidity networks across life and gender. New J Phys. 2014;16: 115013.CrossRef Chmiel A, Klimek P, Thurner S. Spreading of diseases through comorbidity networks across life and gender. New J Phys. 2014;16: 115013.CrossRef
43.
Zurück zum Zitat Piñero J, Ramírez-Anguita JM, Saüch-Pitarch J, Ronzano F, Centeno E, Sanz F, et al. The DisGeNET knowledge platform for disease genomics: 2019 update. Nucleic Acids Res. 2020;48:D845–55.PubMed Piñero J, Ramírez-Anguita JM, Saüch-Pitarch J, Ronzano F, Centeno E, Sanz F, et al. The DisGeNET knowledge platform for disease genomics: 2019 update. Nucleic Acids Res. 2020;48:D845–55.PubMed
45.
Zurück zum Zitat Türei D, Valdeolivas A, Gul L, Palacio‐Escat N, Klein M, Ivanova O, et al. Integrated intra‐ and intercellular signaling knowledge for multicellular omics analysis. Mol Syst Biol. 2021;17:e9923. Türei D, Valdeolivas A, Gul L, Palacio‐Escat N, Klein M, Ivanova O, et al. Integrated intra‐ and intercellular signaling knowledge for multicellular omics analysis. Mol Syst Biol. 2021;17:e9923.
48.
Zurück zum Zitat Luck K, Kim D-K, Lambourne L, Spirohn K, Begg BE, Bian W, et al. A reference map of the human binary protein interactome. Nature. 2020;580:402–8.PubMedPubMedCentralCrossRef Luck K, Kim D-K, Lambourne L, Spirohn K, Begg BE, Bian W, et al. A reference map of the human binary protein interactome. Nature. 2020;580:402–8.PubMedPubMedCentralCrossRef
49.
Zurück zum Zitat Buphamalai P, Kokotovic T, Nagy V, Menche J. Network analysis reveals rare disease signatures across multiple levels of biological organization. Nat Commun. 2021;12:6306.PubMedPubMedCentralCrossRef Buphamalai P, Kokotovic T, Nagy V, Menche J. Network analysis reveals rare disease signatures across multiple levels of biological organization. Nat Commun. 2021;12:6306.PubMedPubMedCentralCrossRef
50.
Zurück zum Zitat Doll S, Dreßen M, Geyer PE, Itzhak DN, Braun C, Doppler SA, et al. Region and cell-type resolved quantitative proteomic map of the human heart. Nat Commun. 2017;8:1469.PubMedPubMedCentralCrossRef Doll S, Dreßen M, Geyer PE, Itzhak DN, Braun C, Doppler SA, et al. Region and cell-type resolved quantitative proteomic map of the human heart. Nat Commun. 2017;8:1469.PubMedPubMedCentralCrossRef
53.
Zurück zum Zitat Valdeolivas A, Tichit L, Navarro C, Perrin S, Odelin G, Levy N, et al. Random walk with restart on multiplex and heterogeneous biological networks. Bioinformatics. 2019;35:497–505.PubMedCrossRef Valdeolivas A, Tichit L, Navarro C, Perrin S, Odelin G, Levy N, et al. Random walk with restart on multiplex and heterogeneous biological networks. Bioinformatics. 2019;35:497–505.PubMedCrossRef
54.
Zurück zum Zitat Robinson MD, McCarthy DJ, Smyth GK. edgeR: a bioconductor package for differential expression analysis of digital gene expression data. Bioinformatics. 2010;26:139–40.PubMedCrossRef Robinson MD, McCarthy DJ, Smyth GK. edgeR: a bioconductor package for differential expression analysis of digital gene expression data. Bioinformatics. 2010;26:139–40.PubMedCrossRef
55.
Zurück zum Zitat Ritchie ME, Phipson B, Wu D, Hu Y, Law CW, Shi W, et al. limma powers differential expression analyses for RNA-sequencing and microarray studies. Nucleic Acids Res. 2015;43: e47.PubMedPubMedCentralCrossRef Ritchie ME, Phipson B, Wu D, Hu Y, Law CW, Shi W, et al. limma powers differential expression analyses for RNA-sequencing and microarray studies. Nucleic Acids Res. 2015;43: e47.PubMedPubMedCentralCrossRef
56.
Zurück zum Zitat Chen EY, Tan CM, Kou Y, Duan Q, Wang Z, Meirelles GV, et al. Enrichr: interactive and collaborative HTML5 gene list enrichment analysis tool. BMC Bioinformatics. 2013;14:128.PubMedPubMedCentralCrossRef Chen EY, Tan CM, Kou Y, Duan Q, Wang Z, Meirelles GV, et al. Enrichr: interactive and collaborative HTML5 gene list enrichment analysis tool. BMC Bioinformatics. 2013;14:128.PubMedPubMedCentralCrossRef
57.
Zurück zum Zitat Korotkevich G, Sukhov V, Budin N, Shpak B, Artyomov MN, Sergushichev A. Fast gene set enrichment analysis. BioRxiv. 2016. Korotkevich G, Sukhov V, Budin N, Shpak B, Artyomov MN, Sergushichev A. Fast gene set enrichment analysis. BioRxiv. 2016.
58.
Zurück zum Zitat Ergatoudes C, Schaufelberger M, Andersson B, Pivodic A, Dahlström U, Fu M. Non-cardiac comorbidities and mortality in patients with heart failure with reduced vs. preserved ejection fraction: a study using the Swedish Heart Failure Registry. Clin Res Cardiol. 2019;108:1025–33.PubMedPubMedCentralCrossRef Ergatoudes C, Schaufelberger M, Andersson B, Pivodic A, Dahlström U, Fu M. Non-cardiac comorbidities and mortality in patients with heart failure with reduced vs. preserved ejection fraction: a study using the Swedish Heart Failure Registry. Clin Res Cardiol. 2019;108:1025–33.PubMedPubMedCentralCrossRef
59.
Zurück zum Zitat Cruz-Ávila HA, Vallejo M, Martínez-García M, Hernández-Lemus E. Comorbidity networks in cardiovascular diseases. Front Physiol. 2020;11:1009.PubMedPubMedCentralCrossRef Cruz-Ávila HA, Vallejo M, Martínez-García M, Hernández-Lemus E. Comorbidity networks in cardiovascular diseases. Front Physiol. 2020;11:1009.PubMedPubMedCentralCrossRef
60.
Zurück zum Zitat Klimek P, Aichberger S, Thurner S. Disentangling genetic and environmental risk factors for individual diseases from multiplex comorbidity networks. Sci Rep. 2016;6:39658.PubMedPubMedCentralCrossRef Klimek P, Aichberger S, Thurner S. Disentangling genetic and environmental risk factors for individual diseases from multiplex comorbidity networks. Sci Rep. 2016;6:39658.PubMedPubMedCentralCrossRef
61.
Zurück zum Zitat Divo MJ, Casanova C, Marin JM, Pinto-Plata VM,de-Torres JP, Zulueta JJ, et al. COPD comorbidities network. Eur Respir J. 2015;46:640–50.PubMedCrossRef Divo MJ, Casanova C, Marin JM, Pinto-Plata VM,de-Torres JP, Zulueta JJ, et al. COPD comorbidities network. Eur Respir J. 2015;46:640–50.PubMedCrossRef
62.
Zurück zum Zitat Kustatscher G, Collins T, Gingras A-C, Guo T, Hermjakob H, Ideker T, et al. Understudied proteins: opportunities and challenges for functional proteomics. Nat Methods. 2022;19:774–9.PubMedCrossRef Kustatscher G, Collins T, Gingras A-C, Guo T, Hermjakob H, Ideker T, et al. Understudied proteins: opportunities and challenges for functional proteomics. Nat Methods. 2022;19:774–9.PubMedCrossRef
63.
Zurück zum Zitat Wesseling M, de Poel JHC, de Jager SCA. Growth differentiation factor 15 in adverse cardiac remodelling: from biomarker to causal player. ESC Heart Fail. 2020;7:1488–501.PubMedPubMedCentralCrossRef Wesseling M, de Poel JHC, de Jager SCA. Growth differentiation factor 15 in adverse cardiac remodelling: from biomarker to causal player. ESC Heart Fail. 2020;7:1488–501.PubMedPubMedCentralCrossRef
64.
Zurück zum Zitat Szumska D, Cioroch M, Keeling A, Prat A, Seidah NG, Bhattacharya S. Pcsk5 is required in the early cranio-cardiac mesoderm for heart development. BMC Dev Biol. 2017;17:6.PubMedPubMedCentralCrossRef Szumska D, Cioroch M, Keeling A, Prat A, Seidah NG, Bhattacharya S. Pcsk5 is required in the early cranio-cardiac mesoderm for heart development. BMC Dev Biol. 2017;17:6.PubMedPubMedCentralCrossRef
65.
Zurück zum Zitat Warren SA, Terada R, Briggs LE, Cole-Jeffrey CT, Chien W-M, Seki T, et al. Differential role of Nkx2-5 in activation of the atrial natriuretic factor gene in the developing versus failing heart. Mol Cell Biol. 2011;31:4633–45.PubMedPubMedCentralCrossRef Warren SA, Terada R, Briggs LE, Cole-Jeffrey CT, Chien W-M, Seki T, et al. Differential role of Nkx2-5 in activation of the atrial natriuretic factor gene in the developing versus failing heart. Mol Cell Biol. 2011;31:4633–45.PubMedPubMedCentralCrossRef
66.
Zurück zum Zitat Arasaratnam D, Bell KM, Sim CB, Koutsis K, Anderson DJ, Qian EL, et al. The role of cardiac transcription factor NKX2-5 in regulating the human cardiac miRNAome. Sci Rep. 2019;9:15928.PubMedPubMedCentralCrossRef Arasaratnam D, Bell KM, Sim CB, Koutsis K, Anderson DJ, Qian EL, et al. The role of cardiac transcription factor NKX2-5 in regulating the human cardiac miRNAome. Sci Rep. 2019;9:15928.PubMedPubMedCentralCrossRef
68.
69.
Zurück zum Zitat Blanton RM, Carrillo-Salinas FJ, Alcaide P. T-cell recruitment to the heart: friendly guests or unwelcome visitors? Am J Physiol Heart Circ Physiol. 2019;317:H124–40.PubMedPubMedCentralCrossRef Blanton RM, Carrillo-Salinas FJ, Alcaide P. T-cell recruitment to the heart: friendly guests or unwelcome visitors? Am J Physiol Heart Circ Physiol. 2019;317:H124–40.PubMedPubMedCentralCrossRef
70.
Zurück zum Zitat Croquelois A, Domenighetti AA, Nemir M, Lepore M, Rosenblatt-Velin N, Radtke F, et al. Control of the adaptive response of the heart to stress via the Notch1 receptor pathway. J Exp Med. 2008;205:3173–85.PubMedPubMedCentralCrossRef Croquelois A, Domenighetti AA, Nemir M, Lepore M, Rosenblatt-Velin N, Radtke F, et al. Control of the adaptive response of the heart to stress via the Notch1 receptor pathway. J Exp Med. 2008;205:3173–85.PubMedPubMedCentralCrossRef
71.
Zurück zum Zitat Fernández-Ruiz I. Selective JAG1-NOTCH3 targeting shows potential for treating PAH. Nat Rev Cardiol. 2022;19:433.PubMedCrossRef Fernández-Ruiz I. Selective JAG1-NOTCH3 targeting shows potential for treating PAH. Nat Rev Cardiol. 2022;19:433.PubMedCrossRef
72.
Zurück zum Zitat Zhao Q, Huang J, Wang D, Chen L, Sun D, Zhao C. Endothelium-specific CYP2J2 overexpression improves cardiac dysfunction by promoting angiogenesis via Jagged1/Notch1 signaling. J Mol Cell Cardiol. 2018;123:118–27.PubMedCrossRef Zhao Q, Huang J, Wang D, Chen L, Sun D, Zhao C. Endothelium-specific CYP2J2 overexpression improves cardiac dysfunction by promoting angiogenesis via Jagged1/Notch1 signaling. J Mol Cell Cardiol. 2018;123:118–27.PubMedCrossRef
74.
Zurück zum Zitat Wang J-K, Li Y, Zhao X-L, Liu Y-B, Tan J, Xing Y-Y, et al. Ablation of plasma prekallikrein decreases low-density lipoprotein cholesterol by stabilizing low-density lipoprotein receptor and protects against atherosclerosis. Circulation. 2022;145:675–87.PubMedCrossRef Wang J-K, Li Y, Zhao X-L, Liu Y-B, Tan J, Xing Y-Y, et al. Ablation of plasma prekallikrein decreases low-density lipoprotein cholesterol by stabilizing low-density lipoprotein receptor and protects against atherosclerosis. Circulation. 2022;145:675–87.PubMedCrossRef
75.
Zurück zum Zitat Dixit G, Blair J, Ozcan C. Plasma proteomic analysis of association between atrial fibrillation, coronary microvascular disease and heart failure. Am J Cardiovasc Dis. 2022;12:81–91.PubMedPubMedCentral Dixit G, Blair J, Ozcan C. Plasma proteomic analysis of association between atrial fibrillation, coronary microvascular disease and heart failure. Am J Cardiovasc Dis. 2022;12:81–91.PubMedPubMedCentral
76.
Zurück zum Zitat Lau ES, Paniagua SM, Zarbafian S, Hoffman U, Long MT, Hwang S-J, et al. Cardiovascular biomarkers of obesity and overlap with cardiometabolic dysfunction. J Am Heart Assoc. 2021;10: e020215.PubMedPubMedCentralCrossRef Lau ES, Paniagua SM, Zarbafian S, Hoffman U, Long MT, Hwang S-J, et al. Cardiovascular biomarkers of obesity and overlap with cardiometabolic dysfunction. J Am Heart Assoc. 2021;10: e020215.PubMedPubMedCentralCrossRef
77.
Zurück zum Zitat Eckenstaler R, Ripperger A, Hauke M, Petermann M, Hemkemeyer SA, Schwedhelm E, et al. A thromboxane A2 receptor-driven COX-2-dependent feedback loop that affects endothelial homeostasis and angiogenesis. Arterioscler Thromb Vasc Biol. 2022;42:444–61.PubMedPubMedCentralCrossRef Eckenstaler R, Ripperger A, Hauke M, Petermann M, Hemkemeyer SA, Schwedhelm E, et al. A thromboxane A2 receptor-driven COX-2-dependent feedback loop that affects endothelial homeostasis and angiogenesis. Arterioscler Thromb Vasc Biol. 2022;42:444–61.PubMedPubMedCentralCrossRef
78.
Zurück zum Zitat Butenas ALE, Rollins KS, Williams AC, Parr SK, Hammond ST, Ade CJ, et al. Thromboxane A2 receptors contribute to the exaggerated exercise pressor reflex in male rats with heart failure. Physiol Rep. 2021;9: e15052.PubMedPubMedCentralCrossRef Butenas ALE, Rollins KS, Williams AC, Parr SK, Hammond ST, Ade CJ, et al. Thromboxane A2 receptors contribute to the exaggerated exercise pressor reflex in male rats with heart failure. Physiol Rep. 2021;9: e15052.PubMedPubMedCentralCrossRef
79.
Zurück zum Zitat Hariri E, Kakouros N, Bunsick DA, Russell SD, Mudd JO, Laws K, et al. Nonplatelet thromboxane generation is associated with impaired cardiovascular performance and mortality in heart failure. Am J Physiol Heart Circ Physiol. 2022;323:H248–55.PubMedPubMedCentralCrossRef Hariri E, Kakouros N, Bunsick DA, Russell SD, Mudd JO, Laws K, et al. Nonplatelet thromboxane generation is associated with impaired cardiovascular performance and mortality in heart failure. Am J Physiol Heart Circ Physiol. 2022;323:H248–55.PubMedPubMedCentralCrossRef
80.
Zurück zum Zitat Alonso F, Dong Y, Génot E. Thrombomodulin, an unexpected new player in endothelial cell invasion during angiogenesis. Arterioscler Thromb Vasc Biol. 2021;41:1672–4.PubMedCrossRef Alonso F, Dong Y, Génot E. Thrombomodulin, an unexpected new player in endothelial cell invasion during angiogenesis. Arterioscler Thromb Vasc Biol. 2021;41:1672–4.PubMedCrossRef
81.
Zurück zum Zitat Eidizadeh A, Schnelle M, Leha A, Edelmann F, Nolte K, Werhahn SM, et al. Biomarker profiles in heart failure with preserved vs. reduced ejection fraction: results from the DIAST-CHF study. ESC Heart Fail. 2023;10(1):200–10. Eidizadeh A, Schnelle M, Leha A, Edelmann F, Nolte K, Werhahn SM, et al. Biomarker profiles in heart failure with preserved vs. reduced ejection fraction: results from the DIAST-CHF study. ESC Heart Fail. 2023;10(1):200–10.
82.
83.
Zurück zum Zitat Huang Q, Qin L, Dai S, Zhang H, Pasula S, Zhou H, et al. AIP1 suppresses atherosclerosis by limiting hyperlipidemia-induced inflammation and vascular endothelial dysfunction. Arterioscler Thromb Vasc Biol. 2013;33:795–804.PubMedPubMedCentralCrossRef Huang Q, Qin L, Dai S, Zhang H, Pasula S, Zhou H, et al. AIP1 suppresses atherosclerosis by limiting hyperlipidemia-induced inflammation and vascular endothelial dysfunction. Arterioscler Thromb Vasc Biol. 2013;33:795–804.PubMedPubMedCentralCrossRef
84.
Zurück zum Zitat Sickinghe AA, Korporaal SJA, den Ruijter HM, Kessler EL. Estrogen contributions to microvascular dysfunction evolving to heart failure with preserved ejection fraction. Front Endocrinol (Lausanne). 2019;10:442.PubMedCrossRef Sickinghe AA, Korporaal SJA, den Ruijter HM, Kessler EL. Estrogen contributions to microvascular dysfunction evolving to heart failure with preserved ejection fraction. Front Endocrinol (Lausanne). 2019;10:442.PubMedCrossRef
85.
Zurück zum Zitat Yoshida H, Matsui T, Yamamoto A, Okada T, Mori K. XBP1 mRNA is induced by ATF6 and spliced by IRE1 in response to ER stress to produce a highly active transcription factor. Cell. 2001;107:881–91.PubMedCrossRef Yoshida H, Matsui T, Yamamoto A, Okada T, Mori K. XBP1 mRNA is induced by ATF6 and spliced by IRE1 in response to ER stress to produce a highly active transcription factor. Cell. 2001;107:881–91.PubMedCrossRef
86.
Zurück zum Zitat Fu F, Doroudgar S. IRE1/XBP1 and endoplasmic reticulum signaling — from basic to translational research for cardiovascular disease. Curr Opin Physiol. 2022;28: 100552.PubMedPubMedCentralCrossRef Fu F, Doroudgar S. IRE1/XBP1 and endoplasmic reticulum signaling — from basic to translational research for cardiovascular disease. Curr Opin Physiol. 2022;28: 100552.PubMedPubMedCentralCrossRef
87.
Zurück zum Zitat Schiattarella GG, Altamirano F, Kim SY, Tong D, Ferdous A, Piristine H, et al. Xbp1s-FoxO1 axis governs lipid accumulation and contractile performance in heart failure with preserved ejection fraction. Nat Commun. 2021;12:1684.PubMedPubMedCentralCrossRef Schiattarella GG, Altamirano F, Kim SY, Tong D, Ferdous A, Piristine H, et al. Xbp1s-FoxO1 axis governs lipid accumulation and contractile performance in heart failure with preserved ejection fraction. Nat Commun. 2021;12:1684.PubMedPubMedCentralCrossRef
88.
Zurück zum Zitat Li L, Zhao Q, Kong W. Extracellular matrix remodeling and cardiac fibrosis. Matrix Biol. 2018;68–69:490–506.PubMedCrossRef Li L, Zhao Q, Kong W. Extracellular matrix remodeling and cardiac fibrosis. Matrix Biol. 2018;68–69:490–506.PubMedCrossRef
90.
Zurück zum Zitat Rodríguez C, Martínez-González J. The role of lysyl oxidase enzymes in cardiac function and remodeling. Cells. 2019;8(12):1483. Rodríguez C, Martínez-González J. The role of lysyl oxidase enzymes in cardiac function and remodeling. Cells. 2019;8(12):1483.
91.
Zurück zum Zitat Agrawal V, Fortune N, Yu S, Fuentes J, Shi F, Nichols D, et al. Natriuretic peptide receptor C contributes to disproportionate right ventricular hypertrophy in a rodent model of obesity-induced heart failure with preserved ejection fraction with pulmonary hypertension. Pulm Circ. 2019;9:2045894019878599.PubMedPubMedCentralCrossRef Agrawal V, Fortune N, Yu S, Fuentes J, Shi F, Nichols D, et al. Natriuretic peptide receptor C contributes to disproportionate right ventricular hypertrophy in a rodent model of obesity-induced heart failure with preserved ejection fraction with pulmonary hypertension. Pulm Circ. 2019;9:2045894019878599.PubMedPubMedCentralCrossRef
92.
93.
Zurück zum Zitat Yu J, Yang Y, Xu Z, Lan C, Chen C, Li C, et al. Long noncoding RNA ahit protects against cardiac hypertrophy through SUZ12 (suppressor of Zeste 12 protein homolog)-mediated downregulation of MEF2A (myocyte enhancer factor 2A). Circ Heart Fail. 2020;13: e006525.PubMedPubMedCentralCrossRef Yu J, Yang Y, Xu Z, Lan C, Chen C, Li C, et al. Long noncoding RNA ahit protects against cardiac hypertrophy through SUZ12 (suppressor of Zeste 12 protein homolog)-mediated downregulation of MEF2A (myocyte enhancer factor 2A). Circ Heart Fail. 2020;13: e006525.PubMedPubMedCentralCrossRef
94.
Zurück zum Zitat Tanaka K, Valero-Muñoz M, Wilson RM, Essick EE, Fowler CT, Nakamura K, et al. Follistatin like 1 regulates hypertrophy in heart failure with preserved ejection fraction. JACC Basic Transl Sci. 2016;1:207–21.PubMedPubMedCentralCrossRef Tanaka K, Valero-Muñoz M, Wilson RM, Essick EE, Fowler CT, Nakamura K, et al. Follistatin like 1 regulates hypertrophy in heart failure with preserved ejection fraction. JACC Basic Transl Sci. 2016;1:207–21.PubMedPubMedCentralCrossRef
95.
Zurück zum Zitat Seki M, Powers JC, Maruyama S, Zuriaga MA, Wu C-L, Kurishima C, et al. Acute and chronic increases of circulating FSTL1 normalize energy substrate metabolism in pacing-induced heart failure. Circ Heart Fail. 2018;11: e004486.PubMedPubMedCentralCrossRef Seki M, Powers JC, Maruyama S, Zuriaga MA, Wu C-L, Kurishima C, et al. Acute and chronic increases of circulating FSTL1 normalize energy substrate metabolism in pacing-induced heart failure. Circ Heart Fail. 2018;11: e004486.PubMedPubMedCentralCrossRef
96.
Zurück zum Zitat Qin M, Huang H, Wang T, Hu H, Liu Y, Cao H, et al. Absence of Rgs5 prolongs cardiac repolarization and predisposes to ventricular tachyarrhythmia in mice. J Mol Cell Cardiol. 2012;53:880–90.PubMedCrossRef Qin M, Huang H, Wang T, Hu H, Liu Y, Cao H, et al. Absence of Rgs5 prolongs cardiac repolarization and predisposes to ventricular tachyarrhythmia in mice. J Mol Cell Cardiol. 2012;53:880–90.PubMedCrossRef
97.
Zurück zum Zitat Li H, He C, Feng J, Zhang Y, Tang Q, Bian Z, et al. Regulator of G protein signaling 5 protects against cardiac hypertrophy and fibrosis during biomechanical stress of pressure overload. Proc Natl Acad Sci USA. 2010;107:13818–23.PubMedPubMedCentralCrossRef Li H, He C, Feng J, Zhang Y, Tang Q, Bian Z, et al. Regulator of G protein signaling 5 protects against cardiac hypertrophy and fibrosis during biomechanical stress of pressure overload. Proc Natl Acad Sci USA. 2010;107:13818–23.PubMedPubMedCentralCrossRef
98.
Zurück zum Zitat Al-Kindi SG, Buzkova P, Shitole SG, Reiner AP, Garg PK, Gottdiener JS, et al. Soluble CD14 and risk of heart failure and its subtypes in older adults. J Card Fail. 2020;26:410–9.PubMedPubMedCentralCrossRef Al-Kindi SG, Buzkova P, Shitole SG, Reiner AP, Garg PK, Gottdiener JS, et al. Soluble CD14 and risk of heart failure and its subtypes in older adults. J Card Fail. 2020;26:410–9.PubMedPubMedCentralCrossRef
99.
Zurück zum Zitat De Franceschi N, Peuhu E, Parsons M, Rissanen S, Vattulainen I, Salmi M, et al. Mutually exclusive roles of SHARPIN in integrin inactivation and NF-κB signaling. PLoS ONE. 2015;10: e0143423.PubMedPubMedCentralCrossRef De Franceschi N, Peuhu E, Parsons M, Rissanen S, Vattulainen I, Salmi M, et al. Mutually exclusive roles of SHARPIN in integrin inactivation and NF-κB signaling. PLoS ONE. 2015;10: e0143423.PubMedPubMedCentralCrossRef
100.
Zurück zum Zitat Lim S, Sala C, Yoon J, Park S, Kuroda S, Sheng M, et al. Sharpin, a novel postsynaptic density protein that directly interacts with the shank family of proteins. Mol Cell Neurosci. 2001;17:385–97.PubMedCrossRef Lim S, Sala C, Yoon J, Park S, Kuroda S, Sheng M, et al. Sharpin, a novel postsynaptic density protein that directly interacts with the shank family of proteins. Mol Cell Neurosci. 2001;17:385–97.PubMedCrossRef
101.
Zurück zum Zitat Corker A, Neff LS, Broughton P, Bradshaw AD, DeLeon-Pennell KY. Organized chaos: deciphering immune cell heterogeneity’s role in inflammation in the heart. Biomolecules. 2021;12(1):11. Corker A, Neff LS, Broughton P, Bradshaw AD, DeLeon-Pennell KY. Organized chaos: deciphering immune cell heterogeneity’s role in inflammation in the heart. Biomolecules. 2021;12(1):11.
102.
Zurück zum Zitat Essandoh K, Auchus RJ, Brody MJ. Cardiac decompensation and promiscuous prenylation of small GTPases in cardiomyocytes in response to local mevalonate pathway disruption. J Pathol. 2022;256:249–52.PubMedCrossRef Essandoh K, Auchus RJ, Brody MJ. Cardiac decompensation and promiscuous prenylation of small GTPases in cardiomyocytes in response to local mevalonate pathway disruption. J Pathol. 2022;256:249–52.PubMedCrossRef
103.
Zurück zum Zitat Xu H, Shen Y, Liang C, Wang H, Huang J, Xue P, et al. Inhibition of the mevalonate pathway improves myocardial fibrosis. Exp Ther Med. 2021;21:224.PubMedPubMedCentralCrossRef Xu H, Shen Y, Liang C, Wang H, Huang J, Xue P, et al. Inhibition of the mevalonate pathway improves myocardial fibrosis. Exp Ther Med. 2021;21:224.PubMedPubMedCentralCrossRef
104.
Zurück zum Zitat Nishizawa H, Maeda N, Shimomura I. Impact of hyperuricemia on chronic kidney disease and atherosclerotic cardiovascular disease. Hypertens Res. 2022;45:635–40.PubMedCrossRef Nishizawa H, Maeda N, Shimomura I. Impact of hyperuricemia on chronic kidney disease and atherosclerotic cardiovascular disease. Hypertens Res. 2022;45:635–40.PubMedCrossRef
105.
Zurück zum Zitat Mátyás C, Németh BT, Oláh A, Török M, Ruppert M, Kellermayer D, et al. Prevention of the development of heart failure with preserved ejection fraction by the phosphodiesterase-5A inhibitor vardenafil in rats with type 2 diabetes. Eur J Heart Fail. 2017;19:326–36.PubMedCrossRef Mátyás C, Németh BT, Oláh A, Török M, Ruppert M, Kellermayer D, et al. Prevention of the development of heart failure with preserved ejection fraction by the phosphodiesterase-5A inhibitor vardenafil in rats with type 2 diabetes. Eur J Heart Fail. 2017;19:326–36.PubMedCrossRef
106.
Zurück zum Zitat Cornuault L, Rouault P, Duplàa C, Couffinhal T, Renault M-A. Endothelial dysfunction in heart failure with preserved ejection fraction: what are the experimental proofs? Front Physiol. 2022;13: 906272.PubMedPubMedCentralCrossRef Cornuault L, Rouault P, Duplàa C, Couffinhal T, Renault M-A. Endothelial dysfunction in heart failure with preserved ejection fraction: what are the experimental proofs? Front Physiol. 2022;13: 906272.PubMedPubMedCentralCrossRef
107.
Zurück zum Zitat Simeunovic D, Odanovic N, Pljesa-Ercegovac M, Radic T, Radovanovic S, Coric V, et al. Glutathione transferase P1 polymorphism might be a risk determinant in heart failure. Dis Markers. 2019;2019:6984845.PubMedPubMedCentralCrossRef Simeunovic D, Odanovic N, Pljesa-Ercegovac M, Radic T, Radovanovic S, Coric V, et al. Glutathione transferase P1 polymorphism might be a risk determinant in heart failure. Dis Markers. 2019;2019:6984845.PubMedPubMedCentralCrossRef
108.
Zurück zum Zitat Singh MM, Kumar R, Tewari S, Agarwal S. Association of GSTT1/GSTM1 and ApoE variants with left ventricular diastolic dysfunction in thalassaemia major patients. Hematology. 2019;24:20–5.PubMedCrossRef Singh MM, Kumar R, Tewari S, Agarwal S. Association of GSTT1/GSTM1 and ApoE variants with left ventricular diastolic dysfunction in thalassaemia major patients. Hematology. 2019;24:20–5.PubMedCrossRef
109.
Zurück zum Zitat Nagamine T, Gillette B, Pakhomov A, Kahoun J, Mayer H, Burghaus R, et al. Multiscale classification of heart failure phenotypes by unsupervised clustering of unstructured electronic medical record data. Sci Rep. 2020;10:21340.PubMedPubMedCentralCrossRef Nagamine T, Gillette B, Pakhomov A, Kahoun J, Mayer H, Burghaus R, et al. Multiscale classification of heart failure phenotypes by unsupervised clustering of unstructured electronic medical record data. Sci Rep. 2020;10:21340.PubMedPubMedCentralCrossRef
110.
Zurück zum Zitat Gulea C, Zakeri R, Quint JK. Model-based comorbidity clusters in patients with heart failure: association with clinical outcomes and healthcare utilization. BMC Med. 2021;19:9.PubMedPubMedCentralCrossRef Gulea C, Zakeri R, Quint JK. Model-based comorbidity clusters in patients with heart failure: association with clinical outcomes and healthcare utilization. BMC Med. 2021;19:9.PubMedPubMedCentralCrossRef
111.
Zurück zum Zitat Woolley RJ, Ceelen D, Ouwerkerk W, Tromp J, Figarska SM, Anker SD, et al. Machine learning based on biomarker profiles identifies distinct subgroups of heart failure with preserved ejection fraction. Eur J Heart Fail. 2021;23:983–91.PubMedCrossRef Woolley RJ, Ceelen D, Ouwerkerk W, Tromp J, Figarska SM, Anker SD, et al. Machine learning based on biomarker profiles identifies distinct subgroups of heart failure with preserved ejection fraction. Eur J Heart Fail. 2021;23:983–91.PubMedCrossRef
112.
Zurück zum Zitat Franssen C, Chen S, Hamdani N, Paulus WJ. From comorbidities to heart failure with preserved ejection fraction: a story of oxidative stress. Heart. 2016;102:320–30.PubMedCrossRef Franssen C, Chen S, Hamdani N, Paulus WJ. From comorbidities to heart failure with preserved ejection fraction: a story of oxidative stress. Heart. 2016;102:320–30.PubMedCrossRef
114.
Zurück zum Zitat Tam MC, Lee R, Cascino TM, Konerman MC, Hummel SL. Current perspectives on systemic hypertension in heart failure with preserved ejection fraction. Curr Hypertens Rep. 2017;19:12.PubMedPubMedCentralCrossRef Tam MC, Lee R, Cascino TM, Konerman MC, Hummel SL. Current perspectives on systemic hypertension in heart failure with preserved ejection fraction. Curr Hypertens Rep. 2017;19:12.PubMedPubMedCentralCrossRef
115.
Zurück zum Zitat Hicklin HE, Gilbert ON, Ye F, Brooks JE, Upadhya B. Hypertension as a road to treatment of heart failure with preserved ejection fraction. Curr Hypertens Rep. 2020;22:82.PubMedCrossRef Hicklin HE, Gilbert ON, Ye F, Brooks JE, Upadhya B. Hypertension as a road to treatment of heart failure with preserved ejection fraction. Curr Hypertens Rep. 2020;22:82.PubMedCrossRef
116.
Zurück zum Zitat Vedin O, Lam CSP, Koh AS, Benson L, Teng THK, Tay WT, et al. Significance of ischemic heart disease in patients with heart failure and preserved, midrange, and reduced ejection fraction: a nationwide cohort study. Circ Heart Fail. 2017;10(6):e003875. Vedin O, Lam CSP, Koh AS, Benson L, Teng THK, Tay WT, et al. Significance of ischemic heart disease in patients with heart failure and preserved, midrange, and reduced ejection fraction: a nationwide cohort study. Circ Heart Fail. 2017;10(6):e003875.
117.
Zurück zum Zitat Reding KW, Cheng RK, Vasbinder A, Ray RM, Barac A, Eaton CB, et al. Lifestyle and cardiovascular risk factors associated with heart failure subtypes in postmenopausal breast cancer survivors. JACC CardioOncol. 2022;4:53–65.PubMedPubMedCentralCrossRef Reding KW, Cheng RK, Vasbinder A, Ray RM, Barac A, Eaton CB, et al. Lifestyle and cardiovascular risk factors associated with heart failure subtypes in postmenopausal breast cancer survivors. JACC CardioOncol. 2022;4:53–65.PubMedPubMedCentralCrossRef
118.
Zurück zum Zitat Saiki H, Petersen IA, Scott CG, Bailey KR, Dunlay SM, Finley RR, et al. Risk of heart failure with preserved ejection fraction in older women after contemporary radiotherapy for breast cancer. Circulation. 2017;135:1388–96.PubMedPubMedCentralCrossRef Saiki H, Petersen IA, Scott CG, Bailey KR, Dunlay SM, Finley RR, et al. Risk of heart failure with preserved ejection fraction in older women after contemporary radiotherapy for breast cancer. Circulation. 2017;135:1388–96.PubMedPubMedCentralCrossRef
119.
Zurück zum Zitat Huang S, Cai T, Weber BN, He Z, Dahal KP, Hong C, et al. The association between inflammation, incident heart failure, and heart failure subtypes in patients with rheumatoid arthritis. Arthritis Care Res (Hoboken). 2021;75(5):1036–45. Huang S, Cai T, Weber BN, He Z, Dahal KP, Hong C, et al. The association between inflammation, incident heart failure, and heart failure subtypes in patients with rheumatoid arthritis. Arthritis Care Res (Hoboken). 2021;75(5):1036–45.
120.
Zurück zum Zitat Packer M. Link between synovial and myocardial inflammation: conceptual framework to explain the pathogenesis of heart failure with preserved ejection fraction in patients with systemic rheumatic diseases. Card Fail Rev. 2020;6:e10. Packer M. Link between synovial and myocardial inflammation: conceptual framework to explain the pathogenesis of heart failure with preserved ejection fraction in patients with systemic rheumatic diseases. Card Fail Rev. 2020;6:e10.
121.
Zurück zum Zitat Gevaert AB, Boen JRA, Segers VF, Van Craenenbroeck EM. Heart failure with preserved ejection fraction: a review of cardiac and noncardiac pathophysiology. Front Physiol. 2019;10:638.PubMedPubMedCentralCrossRef Gevaert AB, Boen JRA, Segers VF, Van Craenenbroeck EM. Heart failure with preserved ejection fraction: a review of cardiac and noncardiac pathophysiology. Front Physiol. 2019;10:638.PubMedPubMedCentralCrossRef
122.
Zurück zum Zitat Gao H, Patel S, Fohtung RB, Cawthon PM, Newman AB, Cauley JA, et al. Sex- and race-specific associations of bone mineral density with incident heart failure and its subtypes in older adults. J Am Geriatr Soc. 2023;71(3):742–55. Gao H, Patel S, Fohtung RB, Cawthon PM, Newman AB, Cauley JA, et al. Sex- and race-specific associations of bone mineral density with incident heart failure and its subtypes in older adults. J Am Geriatr Soc. 2023;71(3):742–55.
123.
Zurück zum Zitat Sabbatini AR, Kararigas G. Menopause-related estrogen decrease and the pathogenesis of HFpEF: JACC review topic of the week. J Am Coll Cardiol. 2020;75:1074–82.PubMedCrossRef Sabbatini AR, Kararigas G. Menopause-related estrogen decrease and the pathogenesis of HFpEF: JACC review topic of the week. J Am Coll Cardiol. 2020;75:1074–82.PubMedCrossRef
124.
Zurück zum Zitat Ponikowski P, Voors AA, Anker SD, Bueno H, Cleland JGF, Coats AJS, et al. 2016 ESC Guidelines for the diagnosis and treatment of acute and chronic heart failure: The Task Force for the diagnosis and treatment of acute and chronic heart failure of the European Society of Cardiology (ESC)Developed with the special contribution of the Heart Failure Association (HFA) of the ESC. Eur Heart J. 2016;37:2129–200.PubMedCrossRef Ponikowski P, Voors AA, Anker SD, Bueno H, Cleland JGF, Coats AJS, et al. 2016 ESC Guidelines for the diagnosis and treatment of acute and chronic heart failure: The Task Force for the diagnosis and treatment of acute and chronic heart failure of the European Society of Cardiology (ESC)Developed with the special contribution of the Heart Failure Association (HFA) of the ESC. Eur Heart J. 2016;37:2129–200.PubMedCrossRef
125.
Zurück zum Zitat Savarese G, Stolfo D, Sinagra G, Lund LH. Heart failure with mid-range or mildly reduced ejection fraction. Nat Rev Cardiol. 2022;19:100–16.PubMedCrossRef Savarese G, Stolfo D, Sinagra G, Lund LH. Heart failure with mid-range or mildly reduced ejection fraction. Nat Rev Cardiol. 2022;19:100–16.PubMedCrossRef
126.
Zurück zum Zitat Bortolotti M, Polito L, Battelli MG, Bolognesi A. Xanthine oxidoreductase: One enzyme for multiple physiological tasks. Redox Biol. 2021;41: 101882.PubMedPubMedCentralCrossRef Bortolotti M, Polito L, Battelli MG, Bolognesi A. Xanthine oxidoreductase: One enzyme for multiple physiological tasks. Redox Biol. 2021;41: 101882.PubMedPubMedCentralCrossRef
127.
Zurück zum Zitat Watanabe K, Watanabe T, Otaki Y, Shishido T, Murase T, Nakamura T, et al. Impact of plasma xanthine oxidoreductase activity in patients with heart failure with preserved ejection fraction. ESC Heart Fail. 2020;7:1735–43.PubMedPubMedCentralCrossRef Watanabe K, Watanabe T, Otaki Y, Shishido T, Murase T, Nakamura T, et al. Impact of plasma xanthine oxidoreductase activity in patients with heart failure with preserved ejection fraction. ESC Heart Fail. 2020;7:1735–43.PubMedPubMedCentralCrossRef
128.
Zurück zum Zitat Yoon S, Kim M, Lee H, Kang G, Bedi K, Margulies KB, et al. S-nitrosylation of histone deacetylase 2 by neuronal nitric oxide synthase as a mechanism of diastolic dysfunction. Circulation. 2021;143:1912–25.PubMedCrossRef Yoon S, Kim M, Lee H, Kang G, Bedi K, Margulies KB, et al. S-nitrosylation of histone deacetylase 2 by neuronal nitric oxide synthase as a mechanism of diastolic dysfunction. Circulation. 2021;143:1912–25.PubMedCrossRef
129.
Zurück zum Zitat van Dam S, Võsa U, van der Graaf A, Franke L, de Magalhães JP. Gene co-expression analysis for functional classification and gene-disease predictions. Brief Bioinformatics. 2018;19:575–92.PubMed van Dam S, Võsa U, van der Graaf A, Franke L, de Magalhães JP. Gene co-expression analysis for functional classification and gene-disease predictions. Brief Bioinformatics. 2018;19:575–92.PubMed
130.
Zurück zum Zitat Ata SK, Wu M, Fang Y, Ou-Yang L, Kwoh CK, Li X-L. Recent advances in network-based methods for disease gene prediction. Brief Bioinformatics. 2021;22(4):bbaa303. Ata SK, Wu M, Fang Y, Ou-Yang L, Kwoh CK, Li X-L. Recent advances in network-based methods for disease gene prediction. Brief Bioinformatics. 2021;22(4):bbaa303.
131.
Zurück zum Zitat Weiskopf NG, Rusanov A, Weng C. Sick patients have more data: the non-random completeness of electronic health records. AMIA Annu Symp Proc. 2013;2013:1472–7.PubMedPubMedCentral Weiskopf NG, Rusanov A, Weng C. Sick patients have more data: the non-random completeness of electronic health records. AMIA Annu Symp Proc. 2013;2013:1472–7.PubMedPubMedCentral
132.
Zurück zum Zitat Wells BJ, Chagin KM, Nowacki AS, Kattan MW. Strategies for handling missing data in electronic health record derived data. EGEMS (Wash DC). 2013;1:1035.PubMed Wells BJ, Chagin KM, Nowacki AS, Kattan MW. Strategies for handling missing data in electronic health record derived data. EGEMS (Wash DC). 2013;1:1035.PubMed
133.
Zurück zum Zitat Haneuse S, Daniels M. A general framework for considering selection bias in EHR-based studies: what data are observed and why? EGEMS (Wash DC). 2016;4:1203.PubMed Haneuse S, Daniels M. A general framework for considering selection bias in EHR-based studies: what data are observed and why? EGEMS (Wash DC). 2016;4:1203.PubMed
134.
Zurück zum Zitat Farmer R, Mathur R, Bhaskaran K, Eastwood SV, Chaturvedi N, Smeeth L. Promises and pitfalls of electronic health record analysis. Diabetologia. 2018;61:1241–8.PubMedCrossRef Farmer R, Mathur R, Bhaskaran K, Eastwood SV, Chaturvedi N, Smeeth L. Promises and pitfalls of electronic health record analysis. Diabetologia. 2018;61:1241–8.PubMedCrossRef
135.
Zurück zum Zitat Pellikka PA, She L, Holly TA, Lin G, Varadarajan P, Pai RG, et al. Variability in ejection fraction measured by echocardiography, gated single-photon emission computed tomography, and cardiac magnetic resonance in patients with coronary artery disease and left ventricular dysfunction. JAMA Netw Open. 2018;1: e181456.PubMedPubMedCentralCrossRef Pellikka PA, She L, Holly TA, Lin G, Varadarajan P, Pai RG, et al. Variability in ejection fraction measured by echocardiography, gated single-photon emission computed tomography, and cardiac magnetic resonance in patients with coronary artery disease and left ventricular dysfunction. JAMA Netw Open. 2018;1: e181456.PubMedPubMedCentralCrossRef
136.
Zurück zum Zitat Pieske B, Tschöpe C, de Boer RA, Fraser AG, Anker SD, Donal E, et al. How to diagnose heart failure with preserved ejection fraction: the HFA-PEFF diagnostic algorithm: a consensus recommendation from the Heart Failure Association (HFA) of the European Society of Cardiology (ESC). Eur Heart J. 2019;40:3297–317.PubMedCrossRef Pieske B, Tschöpe C, de Boer RA, Fraser AG, Anker SD, Donal E, et al. How to diagnose heart failure with preserved ejection fraction: the HFA-PEFF diagnostic algorithm: a consensus recommendation from the Heart Failure Association (HFA) of the European Society of Cardiology (ESC). Eur Heart J. 2019;40:3297–317.PubMedCrossRef
137.
Zurück zum Zitat Triposkiadis F, Butler J, Abboud FM, Armstrong PW, Adamopoulos S, Atherton JJ, et al. The continuous heart failure spectrum: moving beyond an ejection fraction classification. Eur Heart J. 2019;40:2155–63.PubMedPubMedCentralCrossRef Triposkiadis F, Butler J, Abboud FM, Armstrong PW, Adamopoulos S, Atherton JJ, et al. The continuous heart failure spectrum: moving beyond an ejection fraction classification. Eur Heart J. 2019;40:2155–63.PubMedPubMedCentralCrossRef
138.
Zurück zum Zitat Thygesen SK, Christiansen CF, Christensen S, Lash TL, Sørensen HT. The predictive value of ICD-10 diagnostic coding used to assess Charlson comorbidity index conditions in the population-based Danish National Registry of Patients. BMC Med Res Methodol. 2011;11:83.PubMedPubMedCentralCrossRef Thygesen SK, Christiansen CF, Christensen S, Lash TL, Sørensen HT. The predictive value of ICD-10 diagnostic coding used to assess Charlson comorbidity index conditions in the population-based Danish National Registry of Patients. BMC Med Res Methodol. 2011;11:83.PubMedPubMedCentralCrossRef
Metadaten
Titel
A network medicine approach to study comorbidities in heart failure with preserved ejection fraction
verfasst von
Jan D. Lanzer
Alberto Valdeolivas
Mark Pepin
Hauke Hund
Johannes Backs
Norbert Frey
Hans-Christoph Friederich
Jobst-Hendrik Schultz
Julio Saez-Rodriguez
Rebecca T. Levinson
Publikationsdatum
01.12.2023
Verlag
BioMed Central
Erschienen in
BMC Medicine / Ausgabe 1/2023
Elektronische ISSN: 1741-7015
DOI
https://doi.org/10.1186/s12916-023-02922-7

Weitere Artikel der Ausgabe 1/2023

BMC Medicine 1/2023 Zur Ausgabe

Leitlinien kompakt für die Allgemeinmedizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Facharzt-Training Allgemeinmedizin

Die ideale Vorbereitung zur anstehenden Prüfung mit den ersten 49 von 100 klinischen Fallbeispielen verschiedener Themenfelder

Mehr erfahren

Nach Herzinfarkt mit Typ-1-Diabetes schlechtere Karten als mit Typ 2?

29.05.2024 Herzinfarkt Nachrichten

Bei Menschen mit Typ-2-Diabetes sind die Chancen, einen Myokardinfarkt zu überleben, in den letzten 15 Jahren deutlich gestiegen – nicht jedoch bei Betroffenen mit Typ 1.

Wie der Klimawandel gefährliche Pilzinfektionen begünstigt

24.05.2024 Candida-Mykosen Nachrichten

Dass sich invasive Pilzinfektionen in letzter Zeit weltweit häufen, liegt wahrscheinlich auch am Klimawandel. Ausbrüche mit dem Hefepilz Candida auris stellen eine zunehmende Gefahr für Immungeschwächte dar – auch in Deutschland.

So wirken verschiedene Alkoholika auf den Blutdruck

23.05.2024 Störungen durch Alkohol Nachrichten

Je mehr Alkohol Menschen pro Woche trinken, desto mehr steigt ihr Blutdruck, legen Daten aus Dänemark nahe. Ob es dabei auch auf die Art des Alkohols ankommt, wurde ebenfalls untersucht.

Das sind die führenden Symptome junger Darmkrebspatienten

Darmkrebserkrankungen in jüngeren Jahren sind ein zunehmendes Problem, das häufig längere Zeit übersehen wird, gerade weil die Patienten noch nicht alt sind. Welche Anzeichen Ärzte stutzig machen sollten, hat eine Metaanalyse herausgearbeitet.

Update Allgemeinmedizin

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