Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 4.549
1.
J Am Heart Assoc ; 13(9): e034731, 2024 May 07.
Article En | MEDLINE | ID: mdl-38700011

BACKGROUND: Cardiac damage induced by ischemic stroke, such as arrhythmia, cardiac dysfunction, and even cardiac arrest, is referred to as cerebral-cardiac syndrome (CCS). Cardiac macrophages are reported to be closely associated with stroke-induced cardiac damage. However, the role of macrophage subsets in CCS is still unclear due to their heterogeneity. Sympathetic nerves play a significant role in regulating macrophages in cardiovascular disease. However, the role of macrophage subsets and sympathetic nerves in CCS is still unclear. METHODS AND RESULTS: In this study, a middle cerebral artery occlusion mouse model was used to simulate ischemic stroke. ECG and echocardiography were used to assess cardiac function. We used Cx3cr1GFPCcr2RFP mice and NLRP3-deficient mice in combination with Smart-seq2 RNA sequencing to confirm the role of macrophage subsets in CCS. We demonstrated that ischemic stroke-induced cardiac damage is characterized by severe cardiac dysfunction and robust infiltration of monocyte-derived macrophages into the heart. Subsequently, we identified that cardiac monocyte-derived macrophages displayed a proinflammatory profile. We also observed that cardiac dysfunction was rescued in ischemic stroke mice by blocking macrophage infiltration using a CCR2 antagonist and NLRP3-deficient mice. In addition, a cardiac sympathetic nerve retrograde tracer and a sympathectomy method were used to explore the relationship between sympathetic nerves and cardiac macrophages. We found that cardiac sympathetic nerves are significantly activated after ischemic stroke, which contributes to the infiltration of monocyte-derived macrophages and subsequent cardiac dysfunction. CONCLUSIONS: Our findings suggest a potential pathogenesis of CCS involving the cardiac sympathetic nerve-monocyte-derived macrophage axis.


Disease Models, Animal , Ischemic Stroke , Macrophages , Mice, Inbred C57BL , NLR Family, Pyrin Domain-Containing 3 Protein , Animals , Macrophages/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/deficiency , Ischemic Stroke/physiopathology , Ischemic Stroke/metabolism , Ischemic Stroke/pathology , Receptors, CCR2/genetics , Receptors, CCR2/metabolism , Male , Mice, Knockout , Mice , Infarction, Middle Cerebral Artery/physiopathology , Infarction, Middle Cerebral Artery/pathology , Sympathetic Nervous System/physiopathology , Myocardium/pathology , Myocardium/metabolism , Heart Diseases/etiology , Heart Diseases/physiopathology , Heart Diseases/pathology , CX3C Chemokine Receptor 1/genetics , CX3C Chemokine Receptor 1/metabolism , CX3C Chemokine Receptor 1/deficiency
2.
J Am Heart Assoc ; 13(9): e032067, 2024 May 07.
Article En | MEDLINE | ID: mdl-38700010

BACKGROUND: Doxorubicin and other anthracyclines are crucial cancer treatment drugs. However, they are associated with significant cardiotoxicity, severely affecting patient care and limiting dosage and usage. Previous studies have shown that low carbon monoxide (CO) concentrations protect against doxorubicin toxicity. However, traditional methods of CO delivery pose complex challenges for daily administration, such as dosing and toxicity. To address these challenges, we developed a novel oral liquid drug product containing CO (HBI-002) that can be easily self-administered by patients with cancer undergoing doxorubicin treatment, resulting in CO being delivered through the upper gastrointestinal tract. METHODS AND RESULTS: HBI-002 was tested in a murine model of doxorubicin cardiotoxicity in the presence and absence of lung or breast cancer. The mice received HBI-002 twice daily before doxorubicin administration and experienced increased carboxyhemoglobin levels from a baseline of ≈1% to 7%. Heart tissue from mice treated with HBI-002 had a 6.3-fold increase in CO concentrations and higher expression of the cytoprotective enzyme heme oxygenase-1 compared with placebo control. In both acute and chronic doxorubicin toxicity scenarios, HBI-002 protected the heart from cardiotoxic effects, including limiting tissue damage and cardiac dysfunction and improving survival. In addition, HBI-002 did not compromise the efficacy of doxorubicin in reducing tumor volume, but rather enhanced the sensitivity of breast 4T1 cancer cells to doxorubicin while simultaneously protecting cardiac function. CONCLUSIONS: These findings strongly support using HBI-002 as a cardioprotective agent that maintains the therapeutic benefits of doxorubicin cancer treatment while mitigating cardiac damage.


Antibiotics, Antineoplastic , Carbon Monoxide , Cardiotoxicity , Doxorubicin , Membrane Proteins , Animals , Doxorubicin/toxicity , Carbon Monoxide/metabolism , Antibiotics, Antineoplastic/toxicity , Female , Administration, Oral , Mice , Heme Oxygenase-1/metabolism , Heart Diseases/chemically induced , Heart Diseases/prevention & control , Heart Diseases/metabolism , Heart Diseases/pathology , Disease Models, Animal , Mice, Inbred C57BL , Carboxyhemoglobin/metabolism , Ventricular Function, Left/drug effects , Humans
3.
Physiol Rep ; 12(6): e15989, 2024 Mar.
Article En | MEDLINE | ID: mdl-38538007

Cardiac fibroblasts (CFs) are an attractive target for reducing pathological cardiac remodeling, and understanding the underlying mechanisms of these processes is the key to develop successful therapies for treating the pressure-overloaded heart. CF-specific knockout (KO) mouse lines with a Cre recombinase under the control of human TCF21 (hTCF21) promoter and/or an adeno-associated virus serotype 9 (AAV9)-hTCF21 system provide a powerful tool for understanding CF biology in vivo. Although a variety of rat disease models are vital for the research of cardiac fibrosis similar to mouse models, there are few rat models that employ cardiac cell-specific conditional gene modification, which has hindered the development and translational relevance of cardiac disease models. In addition, to date, there are no reports of gene manipulation specifically in rat CFs in vivo. Here, we report a simplified CF-specific rat transgenic model using an AAV9-hTCF21 system that achieved a CF-specific expression of transgene in adult rat hearts. Moreover, we successfully applied this approach to specifically manipulate mitochondrial morphology in quiescent CFs. In summary, this model will allow us to develop fast and simple rat CF-specific transgenic models for studying cardiovascular diseases in vivo.


Cardiomyopathies , Heart Diseases , Mice , Animals , Rats , Humans , Myocytes, Cardiac/metabolism , Dependovirus/genetics , Heart Diseases/pathology , Mice, Knockout , Fibroblasts/metabolism , Basic Helix-Loop-Helix Transcription Factors/metabolism
4.
Nature ; 627(8005): 854-864, 2024 Mar.
Article En | MEDLINE | ID: mdl-38480880

The heart, which is the first organ to develop, is highly dependent on its form to function1,2. However, how diverse cardiac cell types spatially coordinate to create the complex morphological structures that are crucial for heart function remains unclear. Here we integrated single-cell RNA-sequencing with high-resolution multiplexed error-robust fluorescence in situ hybridization to resolve the identity of the cardiac cell types that develop the human heart. This approach also provided a spatial mapping of individual cells that enables illumination of their organization into cellular communities that form distinct cardiac structures. We discovered that many of these cardiac cell types further specified into subpopulations exclusive to specific communities, which support their specialization according to the cellular ecosystem and anatomical region. In particular, ventricular cardiomyocyte subpopulations displayed an unexpected complex laminar organization across the ventricular wall and formed, with other cell subpopulations, several cellular communities. Interrogating cell-cell interactions within these communities using in vivo conditional genetic mouse models and in vitro human pluripotent stem cell systems revealed multicellular signalling pathways that orchestrate the spatial organization of cardiac cell subpopulations during ventricular wall morphogenesis. These detailed findings into the cellular social interactions and specialization of cardiac cell types constructing and remodelling the human heart offer new insights into structural heart diseases and the engineering of complex multicellular tissues for human heart repair.


Body Patterning , Heart , Myocardium , Animals , Humans , Mice , Heart/anatomy & histology , Heart/embryology , Heart Diseases/metabolism , Heart Diseases/pathology , Heart Ventricles/anatomy & histology , Heart Ventricles/cytology , Heart Ventricles/embryology , In Situ Hybridization, Fluorescence , Models, Animal , Myocardium/cytology , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Single-Cell Gene Expression Analysis
5.
J Vet Cardiol ; 52: 61-67, 2024 Apr.
Article En | MEDLINE | ID: mdl-38430823

A dog was presented for lameness, fever, and extreme lethargy. On physical exam, a new heart murmur, arrhythmia, and joint effusion were detected. These findings were not detected two months prior. A diagnostic work-up confirmed septic suppurative inflammation in multiple joints. Echocardiogram revealed aortic valvular endocarditis along with a communication, as a consequence of a fistula, that extended from just below the aortic sinotubular junction to the left atrial lumen. Due to a poor prognosis, humane euthanasia was elected. Necropsy and histopathology confirmed infective endocarditis of the aortic valve and an aorto-left atrial fistulous tract extending from the left coronary sinus of the aortic valve to the lumen of left atrium.


Dog Diseases , Echocardiography , Heart Atria , Animals , Dogs , Dog Diseases/pathology , Dog Diseases/diagnostic imaging , Heart Atria/pathology , Heart Atria/diagnostic imaging , Echocardiography/veterinary , Fistula/veterinary , Fistula/diagnostic imaging , Endocarditis, Bacterial/veterinary , Endocarditis, Bacterial/complications , Endocarditis, Bacterial/diagnostic imaging , Endocarditis, Bacterial/pathology , Vascular Fistula/veterinary , Vascular Fistula/diagnostic imaging , Vascular Fistula/complications , Male , Aortic Diseases/veterinary , Aortic Diseases/diagnostic imaging , Aortic Diseases/pathology , Aortic Diseases/complications , Endocarditis/veterinary , Endocarditis/complications , Endocarditis/diagnostic imaging , Endocarditis/pathology , Heart Diseases/veterinary , Heart Diseases/diagnostic imaging , Heart Diseases/pathology , Heart Diseases/etiology , Heart Diseases/complications , Female
6.
Cardiovasc Toxicol ; 24(2): 184-198, 2024 Feb.
Article En | MEDLINE | ID: mdl-38324115

Advancements in cancer treatments have improved survival rates but have also led to increased cardiotoxicities, which can cause adverse cardiovascular events or worsen pre-existing conditions. Herein, cardiotoxicity is a severe adverse effect of 5-fluorouracil (5-FU) therapy in cancer patients, with reported incidence rates ranging from 1 to 20%. Some studies have also suggested subclinical effects and there are reports which have documented instances of cardiac arrest or sudden death during 5-FU treatment, highlighting the importance of timely management of cardiovascular symptoms. However, despite being treated with conventional medical approaches for this cardiotoxicity, a subset of patients has demonstrated suboptimal or insufficient responses. The frequent use of 5-FU in chemotherapy and its association with significant morbidity and mortality indicates the need for a greater understanding of 5-FU-associated cardiotoxicity. It is essential to reduce the adverse effects of anti-tumor medications while preserving their efficacy, which can be achieved through drugs that mitigate toxicity associated with these drugs. Underpinning cardiotoxicity associated with 5-FU therapy also has the potential to offer valuable guidance in pinpointing pharmacological approaches that can be employed to prevent or ameliorate these effects. The present study provides an overview of management strategies for cardiac events induced by fluoropyrimidine-based cancer treatments. The review encompasses the underlying molecular and cellular mechanisms of cardiotoxicity, associated risk factors, and diagnostic methods. Additionally, we provide information on several available treatments and drug choices for angina resulting from 5-FU exposure, including nicorandil, ranolazine, trimetazidine, ivabradine, and sacubitril-valsartan, which have demonstrated potential in mitigating or protecting against chemotherapy-induced adverse cardiac effects.


Heart Diseases , Neoplasms , Humans , Cardiotoxicity , Fluorouracil/adverse effects , Heart , Heart Diseases/pathology , Neoplasms/drug therapy
7.
Nat Commun ; 15(1): 606, 2024 Jan 19.
Article En | MEDLINE | ID: mdl-38242884

Hematopoietic mutations in epigenetic regulators like DNA methyltransferase 3 alpha (DNMT3A), play a pivotal role in driving clonal hematopoiesis of indeterminate potential (CHIP), and are associated with unfavorable outcomes in patients suffering from heart failure (HF). However, the precise interactions between CHIP-mutated cells and other cardiac cell types remain unknown. Here, we identify fibroblasts as potential partners in interactions with CHIP-mutated monocytes. We used combined transcriptomic data derived from peripheral blood mononuclear cells of HF patients, both with and without CHIP, and cardiac tissue. We demonstrate that inactivation of DNMT3A in macrophages intensifies interactions with cardiac fibroblasts and increases cardiac fibrosis. DNMT3A inactivation amplifies the release of heparin-binding epidermal growth factor-like growth factor, thereby facilitating activation of cardiac fibroblasts. These findings identify a potential pathway of DNMT3A CHIP-driver mutations to the initiation and progression of HF and may also provide a compelling basis for the development of innovative anti-fibrotic strategies.


DNA Methyltransferase 3A , Heart Failure , Humans , Clonal Hematopoiesis , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA Methyltransferase 3A/genetics , Fibroblasts , Fibrosis/genetics , Fibrosis/pathology , Heart Failure/genetics , Hematopoiesis/genetics , Leukocytes, Mononuclear , Mutation , Heart Diseases/genetics , Heart Diseases/pathology
8.
Hypertension ; 81(2): 218-228, 2024 Feb.
Article En | MEDLINE | ID: mdl-38084597

Hypertensive heart disease (HHD) can no longer be considered as the beneficial adaptive result of the hypertrophy of cardiomyocytes in response to pressure overload leading to the development of left ventricular hypertrophy. The current evidence indicates that in patients with HHD, pathological lesions in the myocardium lead to maladaptive structural remodeling and subsequent alterations in cardiac function, electrical activity, and perfusion, all contributing to poor outcomes. Diffuse myocardial interstitial fibrosis is probably the most critically involved lesion in these disorders. Therefore, in this review, we will focus on the histological characteristics, the mechanisms, and the clinical consequences of myocardial interstitial fibrosis in patients with HHD. In addition, we will consider the most useful tools for the noninvasive diagnosis of myocardial interstitial fibrosis in patients with HHD, as well as the most effective available therapeutic strategies to prevent its development or facilitate its regression in this patient population. Finally, we will issue a call to action for the need for more fundamental and clinical research on myocardial interstitial fibrosis in HHD.


Cardiomyopathies , Heart Diseases , Hypertension , Humans , Heart Diseases/pathology , Myocardium/pathology , Hypertrophy, Left Ventricular , Hypertension/complications , Hypertension/drug therapy , Hypertension/pathology , Fibrosis
9.
Cardiovasc Res ; 120(3): 262-272, 2024 03 14.
Article En | MEDLINE | ID: mdl-38084908

AIMS: Physiological cardiac hypertrophy occurs in response to exercise and can protect against pathological stress. In contrast, pathological hypertrophy occurs in disease and often precedes heart failure. The cardiac pathways activated in physiological and pathological hypertrophy are largely distinct. Our prior work demonstrated that miR-222 increases in exercised hearts and is required for exercise-induced cardiac hypertrophy and cardiomyogenesis. Here, we sought to define the role of miR-222 in pathological hypertrophy. METHODS AND RESULTS: We found that miR-222 also increased in pathological hypertrophy induced by pressure overload. To assess its functional significance in this setting, we generated a miR-222 gain-of-function model through cardiac-specific constitutive transgenic miR-222 expression (TgC-miR-222) and used locked nucleic acid anti-miR specific for miR-222 to inhibit its effects. Both gain- and loss-of-function models manifested normal cardiac structure and function at baseline. However, after transverse aortic constriction (TAC), miR-222 inhibition accelerated the development of pathological hypertrophy, cardiac dysfunction, and heart failure. Conversely, miR-222-overexpressing mice had less pathological hypertrophy after TAC, as well as better cardiac function and survival. We identified p53-up-regulated modulator of apoptosis, a pro-apoptotic Bcl-2 family member, and the transcription factors, Hmbox1 and nuclear factor of activated T-cells 3, as direct miR-222 targets contributing to its roles in this context. CONCLUSION: While miR-222 is necessary for physiological cardiac growth, it inhibits cardiac growth in response to pressure overload and reduces adverse remodelling and cardiac dysfunction. These findings support the model that physiological and pathological hypertrophy are fundamentally different. Further, they suggest that miR-222 may hold promise as a therapeutic target in pathological cardiac hypertrophy and heart failure.


Heart Diseases , Heart Failure , MicroRNAs , Mice , Animals , MicroRNAs/genetics , Cardiomegaly/metabolism , Heart Failure/metabolism , Heart , Heart Diseases/pathology , Myocytes, Cardiac/metabolism , Disease Models, Animal , Homeodomain Proteins/metabolism
10.
Int Immunopharmacol ; 126: 111297, 2024 Jan 05.
Article En | MEDLINE | ID: mdl-38039718

OBJECTIVE AND DESIGN: We aimed to investigate the molecular mechanism underlying formaldehyde (FA)-induced congenital heart disease (CHD) using in vitro and in vivo models. MATERIALS AND SUBJECTS: Neonatal rat heart tissues and H9C2 cells were used for in vitro studies, while FA-exposed new-born rats were used for in vivo studies. TREATMENT: H9C2 cells were exposed to FA concentrations of 0, 50, 100 and 150 µM/mL for 24 h. METHODS: Whole transcriptome gene sequencing identified differentially expressed miRNAs in neonatal rat heart tissues, while Real-time quantitative PCR (RT-qPCR) assessed miR-871-3p and Megf8 expression. RNA pull-down and dual-luciferase reporter assays determined miR-871-3p and Megf8 relationships. Inflammatory cytokine expression was assessed by western blotting. A FA-induced CHD model was used to validate miR-871-3p regulatory effects in vivo. RESULTS: We identified 89 differentially expressed miRNAs, with 28 up-regulated and 61 down-regulated (fold change ≥ 2.0, P < 0.05). Inflammation (interleukin) and signalling pathways were found to control FA-induced cardiac dysplasia. miR-871-3p was upregulated in FA-exposed heart tissues, modulated inflammation, and directly targeted Megf8. In vivo experiments showed miR-871-3p knockdown inhibited FA-induced inflammation and CHD. CONCLUSION: We demonstrated miR-871-3p's role in FA-induced CHD by targeting Megf8, providing potential targets for CHD intervention and improved diagnosis and treatment strategies.


Formaldehyde , Heart Diseases , Membrane Proteins , MicroRNAs , Animals , Female , Humans , Infant , Infant, Newborn , Male , Rats , Air Pollutants/metabolism , Air Pollutants/toxicity , Disease Models, Animal , Formaldehyde/metabolism , Formaldehyde/toxicity , Gene Expression , Gene Knockdown Techniques , Heart/drug effects , Heart/physiopathology , Heart Diseases/congenital , Heart Diseases/metabolism , Heart Diseases/pathology , Inflammation/metabolism , Membrane Proteins/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Rats, Sprague-Dawley
11.
Vet Clin Pathol ; 52(4): 681-690, 2023 Dec.
Article En | MEDLINE | ID: mdl-37788981

BACKGROUND: Eosinophilic effusions are commonly defined as effusions with ≥10% eosinophils. Eosinophilic cavitary effusions are infrequently observed in cats, with few case reports comprising the majority of the recent literature. OBJECTIVE: The objective was to review disease associations of cats with eosinophilic cavitary effusions and to assess if a lower threshold of eosinophils (5%-9%) may warrant consideration of similar etiologies associated with effusions with ≥10% eosinophils. METHODS: Cytology reports were retrospectively reviewed for all feline cavitary effusions submitted for fluid analysis from 2010 to 2020 at a veterinary teaching hospital. Cases were included if the manual leukocyte differential included ≥5% eosinophils and separated into 5%-9% and ≥10% eosinophils groups. Patient records were reviewed for associated medical conditions. RESULTS: A total of 669 effusions were submitted from 579 cats; 50 effusions from 48 cats had a leukocyte differential with ≥5% eosinophils. The eosinophil proportion was ≥10% in 22 cats; the most common underlying cause was neoplasia (10/22, 45%), followed by inflammatory disease (4/22, 18%), cardiac disease (3/22, 14%), suspect neoplasia (3/22, 14%), and undetermined (2/22, 9%). The underlying causes for the 26 cats with 5%-9% eosinophils were similar; neoplasia (8/26, 31%), cardiac disease (6/26, 23%), inflammatory disease (4/26, 15%), suspect neoplasia (3/26, 12%), undetermined (3/26, 12%), and idiopathic chylothorax (2/26, 8%). Cats with eosinophil proportions ≥10% in the fluid exhibited peripheral eosinophilia more frequently (35%) compared to those with 5%-9% eosinophils (5%). CONCLUSIONS: Consistent with the current literature, neoplasia, particularly lymphoma, remains a primary consideration for cats with eosinophilic effusions. Previously unreported associated diseases included cardiovascular and inflammatory disorders. Our findings suggest an eosinophil differential of 5%-9% is seen with similar etiologies considered for classically defined eosinophilic effusions.


Cat Diseases , Heart Diseases , Neoplasms , Pleural Effusion , Cats , Animals , Pleural Effusion/veterinary , Retrospective Studies , Hospitals, Animal , Hospitals, Teaching , Eosinophils/pathology , Neoplasms/pathology , Neoplasms/veterinary , Heart Diseases/complications , Heart Diseases/pathology , Heart Diseases/veterinary , Cat Diseases/pathology
12.
Am J Physiol Cell Physiol ; 325(5): C1244-C1251, 2023 Nov 01.
Article En | MEDLINE | ID: mdl-37746696

Myopathic processes affect skeletal muscle and heart. In the muscular dystrophies, which are a subset of myopathies, muscle cells are gradually replaced by fibrosis and fat, impairing muscle function as well as regeneration and repair. In addition to skeletal muscle, these genetic disorders often also affect the heart, where fibrofatty infiltration progressively accumulates in the myocardium, impairing heart function. Although considerable effort has focused on gene-corrective and gene-replacement approaches to stabilize myofibers and cardiomyocytes, the continual and ongoing deposition of extracellular matrix itself contributes to tissue and organ dysfunction. Transcriptomic and proteomic profiling, along with high-resolution imaging and biophysical measurements, have been applied to define extracellular matrix components and their role in contributing to cardiac and skeletal muscle weakness. More recently, decellularization methods have been adapted to an on-slide format to preserve the spatial geography of the extracellular matrix, allowing new insight into matrix remodeling and its direct role in suppressing regeneration in muscle. This review highlights recent literature with focus on the extracellular matrix and molecular mechanisms that contribute to muscle and heart fibrotic disorders. We will also compare how the myopathic matrix differs from healthy matrix, emphasizing how the pathological matrix contributes to disease.


Heart Diseases , Muscular Diseases , Humans , Proteomics , Extracellular Matrix/pathology , Muscular Diseases/genetics , Muscular Diseases/pathology , Muscle, Skeletal/pathology , Myocytes, Cardiac/pathology , Heart Diseases/pathology , Disease Progression , Fibrosis
13.
Int J Cardiovasc Imaging ; 39(11): 2237-2246, 2023 Nov.
Article En | MEDLINE | ID: mdl-37682417

The evaluation of cardiac magnetic resonance feature tracking may have great diagnostic value in hypertrophic cardiomyopathy and hypertensive heart disease. Exploring the diagnostic and clinical research value of cardiac magnetic resonance feature tracks in evaluation of myocardium deformation in patients with subclinical hypertrophic cardiomyopathy(SHCM)and subclinical hypertensive heart disease(SHHD). Cardiovascular Magnetic Resonance (CMR) scans were performed on a 1.5 T MR scanner in 33 patients with SHCM, 31 patients with SHHD, and 27 controls(NS). The CMR image post-processing software was used to analyze the characteristics of routine cardiac function, different global and regional myocardial strain in each group. Analysis of variance (ANOVA) was used to compare age, blood pressure, heart rate, routine cardiac function, body mass index (BMI), as well as the strain between different segments within each of the three groups. Once a significant difference was detected, a least significant difference (LSD) comparison would be performed. The diagnostic efficacy of different parameters in differentiating SHHD from SHCM was evaluated through receiver operating characteristic (ROC) curve analysis, and the best cut-off value was determined. There was no statistical difference among three groups (P>0.05) in routine cardiac function while significant statistical differences were found in the global myocardial strain parameters and the peak strain parameters of some segments (especially basal segments) (P < 0.05). The global radial peak strain (GRPS) was most effective (AUC = 0.885, 95% CI: 0.085-0.971, P<0.001) with a sensitivity and specificity of 84% and 88% at a cut-off value of 40.105, contributing to distinguishing SHCM from SHHD group. Cardiac magnetic resonance feature tracking could detect left ventricular deformation in patients with SHCM and SHHD group. The abnormality of strain has important research value for subclinical diagnosis and clinical evaluation.


Cardiomyopathy, Hypertrophic , Heart Diseases , Hypertension , Humans , Magnetic Resonance Imaging, Cine/methods , Predictive Value of Tests , Myocardium/pathology , Cardiomyopathy, Hypertrophic/complications , Cardiomyopathy, Hypertrophic/diagnostic imaging , Cardiomyopathy, Hypertrophic/pathology , Heart Diseases/pathology , Magnetic Resonance Spectroscopy , Hypertension/complications , Hypertension/diagnostic imaging , Hypertension/pathology , Ventricular Function, Left
14.
Am J Physiol Heart Circ Physiol ; 325(4): H702-H719, 2023 10 01.
Article En | MEDLINE | ID: mdl-37539452

Maternal hypothyroidism (MH) could adversely affect the cardiac disease responses of the progeny. This study tested the hypothesis that MH reduces early postnatal cardiomyocyte (CM) proliferation so that the adult heart of MH progeny has a smaller number of larger cardiac myocytes, which imparts adverse cardiac disease responses following injury. Thyroidectomy (TX) was used to establish MH. The progeny from mice that underwent sham or TX surgery were termed Ctrl (control) or MH (maternal hypothyroidism) progeny, respectively. MH progeny had similar heart weight (HW) to body weight (BW) ratios and larger CM size consistent with fewer CMs at postnatal day 60 (P60) compared with Ctrl (control) progeny. MH progeny had lower numbers of EdU+, Ki67+, and phosphorylated histone H3 (PH3)+ CMs, which suggests they had a decreased CM proliferation in the postnatal timeframe. RNA-seq data showed that genes related to DNA replication were downregulated in P5 MH hearts, including bone morphogenetic protein 10 (Bmp10). Both in vivo and in vitro studies showed Bmp10 treatment increased CM proliferation. After transverse aortic constriction (TAC), the MH progeny had more severe cardiac pathological remodeling compared with the Ctrl progeny. Thyroid hormone (T4) treatment for MH mothers preserved their progeny's postnatal CM proliferation capacity and prevented excessive pathological remodeling after TAC. Our results suggest that CM proliferation during early postnatal development was significantly reduced in MH progeny, resulting in fewer CMs with hypertrophy in adulthood. These changes were associated with more severe cardiac disease responses after pressure overload.NEW & NOTEWORTHY Our study shows that compared with Ctrl (control) progeny, the adult progeny of mothers who have MH (MH progeny) had fewer CMs. This reduction of CM numbers was associated with decreased postnatal CM proliferation. Gene expression studies showed a reduced expression of Bmp10 in MH progeny. Bmp10 has been linked to myocyte proliferation. In vivo and in vitro studies showed that Bmp10 treatment of MH progeny and their myocytes could increase CM proliferation. Differences in CM number and size in adult hearts of MH progeny were linked to more severe cardiac structural and functional remodeling after pressure overload. T4 (synthetic thyroxine) treatment of MH mothers during their pregnancy, prevented the reduction in CM number in their progeny and the adverse response to disease stress.


Heart Diseases , Hypothyroidism , Pregnancy , Female , Mice , Animals , Myocytes, Cardiac/metabolism , Heart Diseases/pathology , Hypertrophy/metabolism , Hypertrophy/pathology , Hypothyroidism/complications , Hypothyroidism/metabolism , Hypothyroidism/pathology , Bone Morphogenetic Proteins/metabolism , Cell Proliferation , Cardiomegaly/metabolism
16.
Cardiovasc Res ; 119(15): 2550-2562, 2023 11 25.
Article En | MEDLINE | ID: mdl-37648651

AIMS: Cardiac fibrosis drives the progression of heart failure in ischaemic and hypertrophic cardiomyopathy. Therefore, the development of specific anti-fibrotic treatment regimens to counteract cardiac fibrosis is of high clinical relevance. Hence, this study examined the presence of persistent fibroblast activation during longstanding human heart disease at a single-cell resolution to identify putative therapeutic targets to counteract pathological cardiac fibrosis in patients. METHODS AND RESULTS: We used single-nuclei RNA sequencing with human tissues from two samples of one healthy donor, and five hypertrophic and two failing hearts. Unsupervised sub-clustering of 7110 nuclei led to the identification of 7 distinct fibroblast clusters. De-convolution of cardiac fibroblast heterogeneity revealed a distinct population of human cardiac fibroblasts with a molecular signature of persistent fibroblast activation and a transcriptional switch towards a pro-fibrotic extra-cellular matrix composition in patients with established cardiac hypertrophy and heart failure. This sub-cluster was characterized by high expression of POSTN, RUNX1, CILP, and a target gene adipocyte enhancer-binding protein 1 (AEBP1) (all P < 0.001). Strikingly, elevated circulating AEBP1 blood level were also detected in a validation cohort of patients with confirmed cardiac fibrosis and hypertrophic cardiomyopathy by cardiac magnetic resonance imaging (P < 0.01). Since endogenous AEBP1 expression was increased in patients with established cardiac hypertrophy and heart failure, we assessed the functional consequence of siRNA-mediated AEBP1 silencing in human cardiac fibroblasts. Indeed, AEBP1 silencing reduced proliferation, migration, and fibroblast contractile capacity and α-SMA gene expression, which is a hallmark of fibroblast activation (all P < 0.05). Mechanistically, the anti-fibrotic effects of AEBP1 silencing were linked to transforming growth factor-beta pathway modulation. CONCLUSION: Together, this study identifies persistent fibroblast activation in patients with longstanding heart disease, which might be detected by circulating AEBP1 and therapeutically modulated by its targeted silencing in human cardiac fibroblasts.


Cardiomyopathies , Cardiomyopathy, Hypertrophic , Heart Diseases , Heart Failure , Humans , Heart Failure/metabolism , Heart Diseases/pathology , Cardiomegaly/metabolism , Cardiomyopathy, Hypertrophic/metabolism , Cardiomyopathies/metabolism , Fibrosis , Fibroblasts/metabolism , Gene Expression Profiling , Carboxypeptidases/metabolism , Repressor Proteins/metabolism
17.
J Pak Med Assoc ; 73(Suppl 4)(4): S221-S227, 2023 Apr.
Article En | MEDLINE | ID: mdl-37482862

Objectives: To identify patients with occult cardiac dysfunction and itsrelationship with the severity of liver impairment. Method: This is a Judgment (Purposive) Sampling, cross-sectionalstudy that was conducted at Kafrelsheikh University Hospital, Egypt, from November 2019 to December 2020, and comprised adult patients of either gender with liver cirrhosis. After detailed history, a clinical examination, pathological assessment and cardiac evaluation based on electrocardiogram and echocardiography, the patients were divided into three groups. Patients who had dyspnoea or cyanosis were in group A, those who did not have dyspnoea or cyanosis but had electrocardiogram and echocardiography abnormalities were in group B, and patients who did not have dyspnoea, cyanosis or electrocardiogram and echocardiography abnormalities were in group C. The severity of the liver disease was evaluated using Child-Pugh and Model of End Liver Disease scores. Data was analysed using SPSS 20. RESULTS: Of the 300 patients, 153(51%) were males and 147(49%) were females. The overall mean age was 55.1±5.1 years(range: 20-60 years). There were 58(19.33%) patientsin group A, 108(36%) in group B and 134(44.66%) in group C. Group A patientsshowed higher Child-Pugh and Model of End Liver Disease scoresthan the other groups(p<0.05). Child-Pugh score >6 and Model of End Liver Disease score >37 yielded the best accuracy for detecting cardiac abnormalities in group B (p<0.05). CONCLUSIONS: There were significant cardiac changes in cirrhotic patients.


Heart Diseases , Liver Cirrhosis , Male , Adult , Female , Humans , Middle Aged , Liver Cirrhosis/diagnosis , Liver Cirrhosis/diagnostic imaging , Heart Diseases/pathology , Echocardiography , Dyspnea , Cyanosis , Severity of Illness Index
18.
Ageing Res Rev ; 88: 101963, 2023 07.
Article En | MEDLINE | ID: mdl-37245633

Ageing is a physiological/pathological process accompanied by the progressive damage of cell function, triggering various ageing-related disorders. Phosphatidylinositol 3-kinase (PI3K), which serves as one of the central regulators of ageing, is closely associated with cellular characteristics or molecular features, such as genome instability, telomere erosion, epigenetic alterations, and mitochondrial dysfunction. In this review, the PI3K signalling pathway was firstly thoroughly explained. The link between ageing pathogenesis and the PI3K signalling pathway was then summarized. Finally, the key regulatory roles of PI3K in ageing-related illnesses were investigated and stressed. In summary, we revealed that drug development and clinical application targeting PI3K is one of the focal points for delaying ageing and treating ageing-related diseases in the future.


Aging , Phosphatidylinositol 3-Kinase , Phosphatidylinositol 3-Kinase/metabolism , Humans , Animals , Signal Transduction , Aging/pathology , Aging/physiology , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/pathology , Metabolic Diseases/metabolism , Metabolic Diseases/pathology , Heart Diseases/metabolism , Heart Diseases/pathology , Neoplasms/metabolism , Neoplasms/pathology
19.
Free Radic Biol Med ; 204: 38-53, 2023 08 01.
Article En | MEDLINE | ID: mdl-37100355

Doxorubicin (DOX) is a potent chemotherapeutic drug for various cancers. Yet, the cardiotoxic side effects limit its application in clinical uses, in which ferroptosis serves as a crucial pathological mechanism in DOX-induced cardiotoxicity (DIC). A reduction of Na+/K + ATPase (NKA) activity is closely associated with DIC progression. However, whether abnormal NKA function was involved in DOX-induced cardiotoxicity and ferroptosis remains unknown. Here, we aim to decipher the cellular and molecular mechanisms of dysfunctional NKA in DOX-induced ferroptosis and investigate NKA as a potential therapeutic target for DIC. A decrease activity of NKA further aggravated DOX-triggered cardiac dysfunction and ferroptosis in NKAα1 haploinsufficiency mice. In contrast, antibodies against the DR-region of NKAα-subunit (DR-Ab) attenuated the cardiac dysfunction and ferroptosis induced by DOX. Mechanistically, NKAα1 interacted with SLC7A11 to form a novel protein complex, which was directly implicated in the disease progression of DIC. Furthermore, the therapeutic effect of DR-Ab on DIC was mediated by reducing ferroptosis by promoting the association of NKAα1/SLC7A11 complex and maintaining the stability of SLC7A11 on the cell surface. These results indicate that antibodies targeting the DR-region of NKA may serve as a novel therapeutic strategy to alleviate DOX-induced cardiotoxicity.


Cardiotoxicity , Heart Diseases , Mice , Animals , Cardiotoxicity/drug therapy , Cardiotoxicity/etiology , Adenosine Triphosphatases/metabolism , Myocytes, Cardiac/metabolism , Doxorubicin/pharmacology , Heart Diseases/pathology , Antibodies/metabolism , Apoptosis , Oxidative Stress
20.
Acta Pharmacol Sin ; 44(8): 1521-1535, 2023 Aug.
Article En | MEDLINE | ID: mdl-36914852

Heart disease is a worldwide health menace. Both intractable primary and secondary cardiomyopathies contribute to malignant cardiac dysfunction and mortality. One of the key cellular processes associated with cardiomyopathy is cardiomyocyte death. Cardiomyocytes are terminally differentiated cells with very limited regenerative capacity. Various insults can lead to irreversible damage of cardiomyocytes, contributing to progression of cardiac dysfunction. Accumulating evidence indicates that majority of cardiomyocyte death is executed by regulating molecular pathways, including apoptosis, ferroptosis, autophagy, pyroptosis, and necroptosis. Importantly, these forms of regulated cell death (RCD) are cardinal features in the pathogenesis of various cardiomyopathies, including dilated cardiomyopathy, diabetic cardiomyopathy, sepsis-induced cardiomyopathy, and drug-induced cardiomyopathy. The relevance between abnormity of RCD with adverse outcome of cardiomyopathy has been unequivocally evident. Therefore, there is an urgent need to uncover the molecular and cellular mechanisms for RCD in order to better understand the pathogenesis of cardiomyopathies. In this review, we summarize the latest progress from studies on RCD pathways in cardiomyocytes in context of the pathogenesis of cardiomyopathies, with particular emphasis on apoptosis, necroptosis, ferroptosis, autophagy, and pyroptosis. We also elaborate the crosstalk among various forms of RCD in pathologically stressed myocardium and the prospects of therapeutic applications targeted to various cell death pathways.


Diabetic Cardiomyopathies , Heart Diseases , Regulated Cell Death , Humans , Apoptosis/physiology , Myocardium/pathology , Diabetic Cardiomyopathies/metabolism , Heart Diseases/pathology
...