-
Mesenchymal stromal cells modulate neutrophil phenotype via paracrine signals.3 weeks agoDespite the recognized importance of neutrophils in cardiac repair following myocardial infarction (MI), their interaction with mesenchymal stromal cells (MSCs), particularly regarding polarization phenotypes and functional impacts, remains unclear. Here, we investigated these interactions across controlled in vitro systems and an in vivo MI model.
Human HL-60 cells were differentiated into neutrophil-like cells (dHL-60) and polarized toward N1/N2 states to test the MSC paracrine effects using indirect transwell coculture. Readouts included gene expression, cytokine profiling and functional assays. To increase translational relevance, indirect ex vivo cocultures of human MSCs with primary neutrophils isolated from MI mice or from MI patients were analysed for gene expression and cytokine profiles in conditioned media. In vivo, syngeneic mouse MSCs were transplanted subcutaneously immediately after MI, and early cardiac function was evaluated by echocardiography. Cardiac neutrophils where quantified by flow cytometry, and Ly6G+ neutrophils from infarcted hearts and peripheral blood were purified by MACS for bulk RNA-seq with targeted RT-qPCR validation.
In vitro, MSCs suppressed pro-inflammatory mediators in N1-like neutrophils and enhanced reparative factors in N2-like cells. In vivo, remote MSC transplantation improved early cardiac performance, and reduced neutrophil accumulation in the infarct. Paradoxically, cardiac neutrophils showed transcriptomic enrichment of inflammatory pathways, whereas blood neutrophils showed reduced interferon-related programs.
Our findings indicate that MSCs can modulate neutrophil responses, underscoring the nuanced effects of MSC-based approaches in ischemic heart disease. These results suggest that anti-inflammatory effects observed under controlled conditions may not fully translate in vivo, highlighting the importance of context when evaluating MSC-based therapies.Cardiovascular diseasesCare/Management -
Impact of digoxin versus beta-blocker in patients with coexistent atrial fibrillation and heart failure: a target trial emulation.3 weeks agoThis study aimed to compare the impact of digoxin versus beta-blocker on adverse clinical outcomes in patients with coexisting atrial fibrillation (AF) and heart failure (HF).
This study employed a target trial emulation with a clone-censor-weight approach to analyze data from 28,377 patients diagnosed with both AF and HF in the Clinical Data Analysis and Reporting System (CDARS) in Hong Kong between January 1, 2005, and December 31, 2017. Patients were followed up for up to 3 years or until the occurrence of clinical outcomes. The exposures were digoxin (N = 5351) versus beta-blocker (N = 7655) within a 90-day grace period. Absolute risks (ARs), risk differences, and risk ratios (RRs) with 95% confidence intervals (CIs) were estimated using weighted pooled logistic regression adjusted for demographic characteristics, comorbidities, and medication use. The primary outcome was all-cause mortality, while secondary outcomes included cardiovascular (CV) mortality, heart failure hospitalization, acute ischemic stroke, acute myocardial infarction, and pacemaker implantation.
Over 3 years, digoxin was associated with a significantly higher risk of all-cause mortality (AR: 51.2% vs. 42.2%; RR: 1.21; 95% CI: 1.17 to 1.26), CV mortality (AR: 25.1% vs. 21.0%; RR: 1.20; 95% CI: 1.11 to 1.29), and heart failure hospitalization (AR: 29.0% vs. 26.4%; RR: 1.10; 95% CI: 1.04 to 1.16). No significant differences were observed for acute ischemic stroke (AR: 4.3% vs. 4.3%; RR: 1.00; 95% CI: 0.85 to 1.18), acute myocardial infarction (AR: 4.6% vs. 4.3%; RR: 1.04; 95% CI: 0.88 to 1.23), or pacemaker implantation (AR: 1.0% vs. 1.3%; RR: 0.74; 95% CI: 0.54 to 1.01).
In patients with coexisting AF and HF, digoxin was associated with significantly higher risks of all-cause mortality, cardiovascular mortality, and heart failure hospitalization compared to beta-blocker.Cardiovascular diseasesCare/Management -
An optimized variant prioritization process for rare disease diagnostics: recommendations for Exomiser and Genomiser.3 weeks agoExome sequencing (ES) and genome sequencing (GS) are increasingly used as standard genetic tests to identify diagnostic variants in rare disease cases. However, prioritizing these variants to reduce the time and burden of manual interpretation by clinical teams remains a significant challenge. The Exomiser/Genomiser software suite is the most widely adopted open-source software for prioritizing coding and noncoding variants. Despite its ubiquitous use, limited data-driven guidelines currently exist to optimize its performance for diagnostic variant prioritization. Based on detailed analyses of Undiagnosed Diseases Network (UDN) probands, this study presents optimized parameters and practical recommendations for deploying the Exomiser and Genomiser tools. We also highlight scenarios where diagnostic variants may be missed and propose alternative workflows to improve diagnostic success in such complex cases.
We analyzed 386 diagnosed probands from the UDN, including cases with coding and noncoding diagnostic variants. We systematically evaluated how tool performance was affected by key parameters, including gene:phenotype association data, variant pathogenicity predictors, phenotype term quality and quantity, and the inclusion and accuracy of family variant data.
Parameter optimization significantly improved Exomiser's performance over default parameters. For GS data, the percentage of coding diagnostic variants ranked within the top 10 candidates increased from 49.7% to 85.5%, and for ES, from 67.3% to 88.2%. For noncoding variants prioritized with Genomiser, the top 10 rankings improved from 15.0% to 40.0%. We also explored refinement strategies for Exomiser outputs, including using p-value thresholds and flagging genes that are frequently ranked in the top 30 candidates but rarely associated with diagnoses.
This study provides an evidence-based framework for variant prioritization in ES and GS data using Exomiser and Genomiser. These recommendations have been implemented in the Mosaic platform to support the ongoing analysis of undiagnosed UDN participants and provide efficient, scalable reanalysis to improve diagnostic yield. Our work also highlights the importance of tracking solved cases and diagnostic variants that can be used to benchmark bioinformatics tools. Exomiser and Genomiser are available at https://github.com/exomiser/Exomiser/ .Cardiovascular diseasesCare/Management -
Mechanism of DKK3 protecting brain tissues in ischemic stroke by regulating LGI1 expression and activity.3 weeks agoTo investigate the mechanism of Dickkopf-3 (DKK3) in protecting brain tissues in ischemic stroke (IS) by regulating LGI1 expression and activity.
220 patients with acute ischemic stroke (AIS) were enrolled to measure the serum DKK3 levels and analyze the predictive ability of serum DKK3 for early neurological deterioration (END) in AIS patients. A murine model of permanent middle cerebral artery occlusion (pMCAO) was established. Neurological function recovery in mice was assessed using neurological function scores and behavioral experiments, and the volume of cerebral infarction in mice was measured. The histopathological morphology of the cortex in mice was observed by hematoxylin‒eosin staining; apoptotic neurons were evaluated by terminal deoxynucleotidyl transferase dUTP nick end labeling staining. DKK3 and leucine-rich glioma-inactivated protein 1 (LGI1) expression levels were measured by immunofluorescence, RT-qPCR, or Western blot, and their interaction was verified by bioinformatics analysis and Co-IP. A cellular model of oxygen-glucose deprivation (OGD) injury was established using HT22 murine neurons for in vitro investigation.
DKK3 was downregulated in IS and shows potential as a biomarker for predicting END in AIS patients. DKK3 overexpression contributed to neurological recovery and attenuated brain injury in pMCAO mice. DKK3 co-localized and interacted with LGI1. LGI1 downregulation weakened the protective effect of DKK3 against pMCAO-induced brain damage and OGD-induced neuronal injury.
DKK3 protects against ischemic injury in permanent cerebral ischemia by regulating LGI1 expression and activity.Cardiovascular diseasesCare/Management -
ACOX2 destabilizes the MRE11-RAD50-NBS1 complex and boosts anticancer immunity via the cGAS-STING pathway in clear cell renal cell carcinoma.3 weeks agoThe rapid development of ICI-based immunotherapy has ushered in a promising era for clear cell renal cell carcinoma (ccRCC). However, durable clinical responses remain limited to a subset of patients. Therefore, identifying novel predictive biomarkers and developing effective combination immunotherapies are critical for advancing personalized ccRCC management. In this study, we report that ccRCC patients exhibiting elevated ACOX2 expression may benefit from PARPi in combination with ICI. Multi-omics cohorts show ACOX2 is significantly downregulated in ccRCC and correlated with improved clinical prognosis. ACOX2 inhibits the growth of ccRCC both in vitro and in vivo. Mechanistically, ACOX2 interacts with MRE11 and inhibits the binding of MRE11 and RAD50, thereby destabilizing the MRE11-RAD50-NBS1 (MRN) complex. Furthermore, ACOX2 activates the cGAS-STING pathway, correlates with more mature tertiary lymphoid structures (TLS), and enhances CD8+ T cell infiltration and activity. Therapeutically, preclinical ccRCC models with high ACOX2 expression, including ccRCC cells, cell-derived xenograft (CDX), patient-derived organoid (PDO), patient-derived xenograft (PDX), and immunocompetent mouse models show increased sensitivity to PARPi plus anti-PD-1 therapy. In conclusion, our findings elucidate a pivotal role of ACOX2 in inhibiting HRR and propose that PARPi, either alone or in combination with anti-PD-1 therapy, represents a promising treatment strategy for ccRCC with elevated ACOX2 expression.
[Image: see text]
The online version contains supplementary material available at 10.1186/s12943-025-02420-9.Cardiovascular diseasesCare/Management -
Emerging therapy strategies for energy metabolism in acute myocardial infarction.3 weeks agoAcute myocardial infarction (AMI) is a life-threatening cardiovascular event caused by the sudden blockage of a coronary artery, usually triggered by thrombotic events. This leads to significant myocardial ischaemia and hypoxia, disrupting the heart's energy metabolism and ultimately resulting in irreversible injury to cardiomyocytes. Despite advancements in reperfusion therapies, ischaemia-reperfusion injury (IRI) remains a critical contributor to complications such as arrhythmias and heart failure. This review explores the pivotal role of myocardial energy metabolism in AMI pathogenesis, focusing on the dysregulation of fatty acid oxidation (FAO), glycolysis, and mitochondrial dysfunction during ischaemia-reperfusion. Key mechanisms, including the overproduction of reactive oxygen species (ROS), succinate accumulation, and the opening of the mitochondrial permeability transition pore (mPTP), are highlighted as drivers of cellular injury. Emerging therapeutic strategies targeting metabolic reprogramming, mitochondrial protection, and ischaemic conditioning are discussed, highlighting their potential to mitigate IRI. By integrating preclinical and clinical evidence, this review highlights the promise of metabolic modulation as a significant approach to enhancing outcomes in AMI patients.Cardiovascular diseasesCare/Management
-
Association between anxiety disorders and medication adherence in patients with cardiovascular disease: a systematic review and meta-analysis of observational studies.3 weeks agoThe management of cardiovascular (CV) diseases is considerably hindered by the presence of anxiety disorders, which act as a significant barrier to medication adherence. This systematic review and meta-analysis aim to synthesize existing evidence on the relationship between anxiety and medication adherence in individuals with CV diseases.
A systematic search was conducted across five major databases from inception to 2025. The methodological quality of the included studies was assessed using the National Institutes of Health (NIH) quality assessment tool. A random-effects meta-analysis was conducted utilizing Comprehensive Meta-Analysis software (version 3), with with p < 0.05 indicating significance.
A total of 23 studies were selected, including 18 with cross-sectional and five with a longitudinal study design. The combined sample size was 72,815 participants, with an average age of 60.7 years and 51.7% were male participants. Majority of studies (n = 15) reported a negative association between anxiety and cardiovascular (CV) medication adherence. Meta-analysis (n = 20) further supported this overall negative association (SMD = -1.087, 95% CI [-1.469, -0.705], p < 0.001) which was also evident across clinical subgroups: hypertension (SMD = -0.648, 95% CI [-1.293, -0.004], p = 0.049), congestive heart failure (SMD = -0.51, 95% CI [-0.92, -0.10], p = 0.02), and ischemic heart disease (SMD = -2.322, 95% CI [-3.498, -1.146], p < 0.001).
This review primarily contributes to the understanding of negative association between anxiety and medication adherence in the context of CV diseases. However, the predominance of cross-sectional designs, and substantial heterogeneity necessitate cautious interpretation of these findings.
The online version contains supplementary material available at 10.1186/s12888-025-07350-w.Cardiovascular diseasesCare/Management -
Indole-3-lactic acid protects the gut vascular barrier following intestinal ischemia injury through AhR/Nrf2/STAT3 mediated claudin 2 downregulation.3 weeks agoDysfunction of the intestinal epithelial barrier (IEB) and gut vascular barrier (GVB) contributes to the development of intestinal ischemia/reperfusion (IR) injury. Tryptophan (TRP), an essential amino acid, plays a crucial role in maintaining intestinal homeostasis, yet its regulatory effects on the GVB following IR remain unexplored. We aimed to better define the role of TRP in intestinal IR in vivo and in vitro.
Mice underwent intestinal ischemia/reperfusion (IR) and were fed control, TRP-recommended (TRP-r), or TRP-sufficient (TRP-s) diets. Fecal metagenomic sequencing analyzed microbial composition, and targeted metabolomics quantified tryptophan and its metabolites in intestinal and serum samples. ILA's effects on barrier integrity were assessed via tight junction protein expression and FITC-dextran permeability assays. RNA sequencing of intestinal endothelial cells elucidated mechanisms by which ILA modulated GVB function. The STAT3-claudin 2 relationship was validated in vitro by ChIP-qPCR.
TRP supplementation significantly reshaped the gut microbiota, mitigated tissue damage and enhanced the integrity of both the IEB and GVB. Indole-3-lactic acid (ILA), a key tryptophan metabolite, was identified as an important factor in preserving GVB function. Mechanistically, our results show that the aryl hydrocarbon receptor (AhR)/Nrf2/signal transducer and activator of transcription 3 (STAT3) pathway is essential for ILA-mediated improvement of GVB integrity and downregulation of the pore-forming protein claudin 2.
Our findings highlight the dual role of ILA in reinforcing both IEB and GVB functions and shed light on the molecular mechanisms underlying ILA's GVB-protective effects. This study implicates that ILA or other AhR-activating metabolites may serve as promising pharmacological agents for alleviating IR-induced intestinal damage.Cardiovascular diseasesCare/ManagementPolicy -
Runx1 drives cardiac fibrosis and heart failure through epigenetic activation of myofibroblasts in pressure overload-induced cardiac remodeling.3 weeks agoHeart failure (HF) is often accompanied by cardiac fibrosis, a pathological process driven by activated cardiac fibroblasts (CFs) transitioning to a myofibroblast phenotype. The Runt-related transcription factor 1 (Runx1) has been implicated in various fibrotic diseases, but its role in cardiac fibrosis and HF progression remains unclear. This study aimed to elucidate the role of Runx1 in CF activation, cardiac fibrosis, and HF development.
HF was induced in mice using transverse aortic constriction (TAC). Runx1 expression was assessed in failing hearts via Western blot and qPCR, with immunostaining to localize Runx1 in CFs. In vitro, CFs were treated with TGF-β, and Runx1 knockdown was achieved using siRNA or adenoviral-mediated deletion. Myofibroblast-specific Runx1 knockout mice (PostnCre, Runx1F/F) were used to investigate the in vivo effects of Runx1 on cardiac function, fibrosis, and hypertrophy post-TAC. Chromatin immunoprecipitation (ChIP) and luciferase assays were conducted to evaluate Runx1's regulation of Postn transcription.
TAC-induced HF was associated with significant upregulation of Runx1 protein and mRNA levels, particularly in CFs. In vitro, Runx1 knockdown suppressed TGF-β-induced markers of CF activation, including α-smooth muscle actin (α-SMA), periostin (Postn), and collagen type I (Col1a1), and reduced CF migration and proliferation. PostnCre-Runx1F/F mice exhibited improved cardiac function, reduced hypertrophy, and decreased fibrosis compared to control mice post-TAC. Mechanistically, Runx1 was found to bind the Postn promoter and recruit the transcriptional coactivator P300, enhancing histone acetylation and promoting Postn transcription.
Runx1 plays a pivotal role in cardiac fibroblast activation and fibrosis, likely through epigenetic regulation of Postn expression, thereby driving heart failure progression. Targeting Runx1 may represent a promising therapeutic strategy for heart failure.Cardiovascular diseasesCare/ManagementPolicy -
Differential cognitive, behavioral, and neurochemical responses to acute chlorpyrifos exposure in normotensive compared to hypertensive adult rats.3 weeks agoClinical and preclinical evidence points to a bilateral association between cardiovascular diseases (CVD) and mental disorders such as anxiety and depression. We previously reported that exposure to organophosphate (OP) compounds, such as chlorpyrifos (CPF), promotes cardiovascular damage and behavioral alterations in normotensive rats. Also, spontaneously hypertensive rats (SHR), a well-established rodent model of hypertension, exhibit more severe symptoms of acute CPF toxicosis and higher mortality rates, likely due to lower plasma butyrylcholinesterase (BuChE) activity. The potential role of pre-existing hypertension in increasing susceptibility to acute OP toxicity, particularly in relation to psychiatric disorders, remains an open question. Given this, we investigated whether SHR are more susceptible than normotensive Wistar rats to the damage caused by acute CPF exposure on innate (elevated plus maze, EPM; light-dark transition, LDT; and open field tests) and learned (contextual fear conditioning) anxiety-like behaviors. A single dose of CPF (20 mg/kg) induced an anxiolytic-like behavior in SHR exposed to the EPM and no effect in Wistar rats. CPF acute intoxication increased fear expression in both strains, but impaired memory extinction only in Wistar rats. CPF inhibited BuChE in Wistar at all tested doses (10, 20 and 30 mg/kg), whereas inhibition occurred only at the highest dose in SHR. CPF also decreased acetylcholinesterase (AChE) activity in the hippocampus and prefrontal cortex of both strains. In summary, acute intoxication with CPF induces strain-dependent behavioral changes. SHRs intoxicated with CPF may not be the most suitable model for studying anxiety susceptibility to OP intoxication in previously hypertensive rats.Cardiovascular diseasesMental HealthCare/Management