Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Circ Res ; 131(11): e152-e168, 2022 11 11.
Artigo em Inglês | MEDLINE | ID: mdl-36263775

RESUMO

BACKGROUND: The pioneer transcription factor (TF) GATA4 (GATA Binding Protein 4) is expressed in multiple cardiovascular lineages and is essential for heart development. GATA4 lineage-specific occupancy in the developing heart underlies its lineage specific activities. Here, we characterized GATA4 chromatin occupancy in cardiomyocyte and endocardial lineages, dissected mechanisms that control lineage specific occupancy, and analyzed GATA4 regulation of endocardial gene expression. METHODS: We mapped GATA4 chromatin occupancy in cardiomyocyte and endocardial cells of embryonic day 12.5 (E12.5) mouse heart using lineage specific, Cre-activated biotinylation of GATA4. Regulation of GATA4 pioneering activity was studied in cell lines stably overexpressing GATA4. GATA4 regulation of endocardial gene expression was analyzed using single cell RNA sequencing and luciferase reporter assays. RESULTS: Cardiomyocyte-selective and endothelial-selective GATA4 occupied genomic regions had features of lineage specific enhancers. Footprints within cardiomyocyte- and endothelial-selective GATA4 regions were enriched for NKX2-5 (NK2 homeobox 5) and ETS1 (ETS Proto-Oncogene 1) motifs, respectively, and both of these TFs interacted with GATA4 in co-immunoprecipitation assays. In stable NIH3T3 cell lines expressing GATA4 with or without NKX2-5 or ETS1, the partner TFs re-directed GATA4 pioneer binding and augmented its ability to open previously inaccessible regions, with ETS1 displaying greater potency as a pioneer partner than NKX2-5. Single-cell RNA sequencing of embryonic hearts with endothelial cell-specific Gata4 inactivation identified Gata4-regulated endocardial genes, which were adjacent to GATA4-bound, endothelial regions enriched for both GATA4 and ETS1 motifs. In reporter assays, GATA4 and ETS1 cooperatively stimulated endothelial cell enhancer activity. CONCLUSIONS: Lineage selective non-pioneer TFs NKX2-5 and ETS1 guide the activity of pioneer TF GATA4 to bind and open chromatin and create active enhancers and mechanistically link ETS1 interaction to GATA4 regulation of endocardial development.


Assuntos
Endocárdio , Fator de Transcrição GATA4 , Proteína Proto-Oncogênica c-ets-1 , Animais , Camundongos , Cromatina/metabolismo , Endocárdio/metabolismo , Fator de Transcrição GATA4/genética , Fator de Transcrição GATA4/metabolismo , Miócitos Cardíacos/metabolismo , Células NIH 3T3 , Proteína Proto-Oncogênica c-ets-1/metabolismo
2.
Am J Physiol Lung Cell Mol Physiol ; 324(2): L89-L101, 2023 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-36472329

RESUMO

Clinical observation indicates that exercise capacity, an important determinant of survival in patients with congenital heart disease (CHD), is most decreased in children with reduced pulmonary blood flow (RPF). However, the underlying mechanism remains unclear. Here, we obtained human RPF lung samples from children with tetralogy of Fallot as well as piglet and rat RPF lung samples from animals with pulmonary artery banding surgery. We observed impaired alveolarization and vascularization, the main characteristics of pulmonary dysplasia, in the lungs of RPF infants, piglets, and rats. RPF caused smaller lungs, cyanosis, and body weight loss in neonatal rats and reduced the number of alveolar type 2 cells. RNA sequencing demonstrated that RPF induced the downregulation of metabolism and migration, a key biological process of late alveolar development, and the upregulation of immune response, which was confirmed by flow cytometry and cytokine detection. In addition, the immunosuppressant cyclosporine A rescued pulmonary dysplasia and increased the expression of the Wnt signaling pathway, which is the driver of postnatal lung development. We concluded that RPF results in pulmonary dysplasia, which may account for the reduced exercise capacity of patients with CHD with RPF. The underlying mechanism is associated with immune response activation, and immunosuppressants have a therapeutic effect in CHD-associated pulmonary dysplasia.


Assuntos
Cardiopatias Congênitas , Alvéolos Pulmonares , Lactente , Criança , Animais , Humanos , Ratos , Suínos , Alvéolos Pulmonares/metabolismo , Pulmão/metabolismo , Cardiopatias Congênitas/complicações , Cardiopatias Congênitas/metabolismo , Cardiopatias Congênitas/patologia , Circulação Pulmonar , Hiperplasia/metabolismo , Hiperplasia/patologia , Animais Recém-Nascidos
3.
Respir Res ; 24(1): 12, 2023 Jan 11.
Artigo em Inglês | MEDLINE | ID: mdl-36631871

RESUMO

BACKGROUND: Pulmonary hypoperfusion is common in children with congenital heart diseases (CHDs) or pulmonary hypertension (PH) and causes adult pulmonary dysplasia. Systematic reviews have shown that some children with CHDs or PH have mitigated clinical outcomes with COVID-19. Understanding the effects of pulmonary hypoperfusion on postnatal alveolar development may aid in the development of methods to improve the pulmonary function of children with CHDs or PH and improve their care during the COVID-19 pandemic, which is characterized by cytokine storm and persistent inflammation. METHODS AND RESULTS: We created a neonatal pulmonary hypoperfusion model through pulmonary artery banding (PAB) surgery at postnatal day 1 (P1). Alveolar dysplasia was confirmed by gross and histological examination at P21. Transcriptomic analysis of pulmonary tissues at P7(alveolar stage 2) and P14(alveolar stage 4) revealed that the postnatal alveolar development track had been changed due to pulmonary hypoperfusion. Under the condition of pulmonary hypoperfusion, the cell-cell communication and axon guidance, which both determine the final number of alveoli, were lost; instead, there was hyperactive cell cycle activity. The transcriptomic results were further confirmed by the examination of axon guidance and cell cycle markers. Because axon guidance controls inflammation and immune cell activation, the loss of axon guidance may explain the lack of severe COVID-19 cases among children with CHDs or PH accompanied by pulmonary hypoperfusion. CONCLUSIONS: This study suggested that promoting cell-cell communication or supplementation with guidance molecules may treat pulmonary hypoperfusion-induced alveolar dysplasia, and that COVID-19 is less likely to cause a cytokine storm in children with CHD or PH accompanied by pulmonary hypoperfusion.


Assuntos
COVID-19 , Hipertensão Pulmonar , Criança , Recém-Nascido , Humanos , Orientação de Axônios , Síndrome da Liberação de Citocina/metabolismo , Síndrome da Liberação de Citocina/patologia , Pandemias , COVID-19/metabolismo , Alvéolos Pulmonares/patologia , Hipertensão Pulmonar/metabolismo , Comunicação Celular
4.
J Cell Biochem ; 120(6): 10106-10117, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30592323

RESUMO

The Rho family plays crucial roles in O2 -induced vasoconstriction, cell proliferation, and migration. Rho GTPase-activating protein 26 (ARHGAP26) is a GTPase-activating protein for the small GTPases of the Rho family. Our previous studies have demonstrated that ARHGAP26 expression was significantly downregulated in patent human ductus arteriosus (DA) tissue. However, its role underlying the maintenance of DA patency is unclear. In this study, patent (fetal) and constricted (newborn) mouse DA tissues were harvested to confirm the differences in the levels of expression of ARHGAP26. Human DA smooth muscle cells (DASMCs) were isolated and cultured in vitro and used to test the function of ARHGAP26. The expression of ARHGAP26 was significantly lower in patent (fetal) than constricted (newborn) mouse DA. ARHGAP26-knocked-down human DASMCs showed reduced proliferation and migration, which are both crucial to anatomic closure of DA. Moreover, after culturing under hypoxic conditions, the expression of ARHGAP26 in human DASMCs was significantly lower and hypoxia-induced ARHGAP26 deficiency activated the phosphorylation level of phosphatase and tensin homolog (PTEN) in DASMCs by mediating the activity of RhoA and RhoA-associated kinase 1 (ROCK1). Use of Y27632, an inhibitor of ROCK which further reduces the phospholipid activity of PTEN can reverse the inhibitory effect of PTEN on the proliferation and migration of human DASMCs. This provides insight into the molecular regulation of the RhoA-ROCK-PTEN pathway in DA smooth muscle cells, which may be a suitable therapeutic target or diagnostic biomarker for perinatal DA tone management.


Assuntos
Movimento Celular/fisiologia , Proliferação de Células/fisiologia , Canal Arterial/metabolismo , Enzimas/metabolismo , Proteínas Ativadoras de GTPase/metabolismo , Miócitos de Músculo Liso/metabolismo , Animais , Animais Recém-Nascidos , Hipóxia Celular , Movimento Celular/genética , Proliferação de Células/genética , Células Cultivadas , Canal Arterial/citologia , Canal Arterial/embriologia , Permeabilidade do Canal Arterial/genética , Permeabilidade do Canal Arterial/metabolismo , Proteínas Ativadoras de GTPase/deficiência , Proteínas Ativadoras de GTPase/genética , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Camundongos Endogâmicos C57BL , Miócitos de Músculo Liso/citologia , PTEN Fosfo-Hidrolase/metabolismo , Interferência de RNA , Transdução de Sinais/fisiologia , Quinases Associadas a rho/metabolismo , Proteína rhoA de Ligação ao GTP/metabolismo
5.
J Cardiovasc Pharmacol ; 68(6): 425-432, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-27575008

RESUMO

BACKGROUND: The renewal capacity of neonate human cardiomyocytes provides an opportunity to manipulate endogenous cardiogenic mechanisms for supplementing the loss of cardiomyocytes caused by myocardial infarction or other cardiac diseases. GSK-3ß inhibitors have been recently shown to promote cardiomyocyte proliferation in rats and mice, thus may be ideal candidates for inducing human cardiomyocyte proliferation. METHODS: Human cardiomyocytes were isolated from right atrial specimens obtained during routine surgery for ventricle septal defect and cultured with either GSK-3ß inhibitor (CHIR-99021) or ß-catenin inhibitor (IWR-1). Immunocytochemistry was performed to visualize 5-ethynyl-2'-deoxyuridine (EdU)-positive or Ki67-positive cardiomyocytes, indicative of proliferative cardiomyocytes. RESULTS: GSK-3ß inhibitor significantly increased ß-catenin accumulation in cell nucleus, whereas ß-catenin inhibitor significantly reduced ß-catenin accumulation in cell plasma. In parallel, GSK-3ß inhibitor increased EdU-positive and Ki67-positive cardiomyocytes, whereas ß-catenin inhibitor decreased EdU-positive and Ki67-positive cardiomyocytes. CONCLUSIONS: These results indicate that GSK-3ß inhibitor can promote human atrial cardiomyocyte proliferation. Although it remains to be determined whether the observations in atrial myocytes could be directly applicable to ventricular myocytes, the current findings imply that Wnt/ß-catenin pathway may be a valuable pathway for manipulating endogenous human heart regeneration.


Assuntos
Proliferação de Células/efeitos dos fármacos , Glicogênio Sintase Quinase 3 beta/antagonistas & inibidores , Miócitos Cardíacos/efeitos dos fármacos , Piridinas/farmacologia , Pirimidinas/farmacologia , Regulação para Cima/efeitos dos fármacos , beta Catenina/biossíntese , Proliferação de Células/fisiologia , Células Cultivadas , Feminino , Glicogênio Sintase Quinase 3 beta/metabolismo , Átrios do Coração/citologia , Átrios do Coração/efeitos dos fármacos , Átrios do Coração/metabolismo , Humanos , Lactente , Recém-Nascido , Masculino , Miócitos Cardíacos/metabolismo , Inibidores de Proteínas Quinases/farmacologia , Regulação para Cima/fisiologia
6.
Tumour Biol ; 36(12): 9179-88, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26088446

RESUMO

Despite the fact that great advances have been made in the management of non-small cell lung cancer (NSCLC), the prognosis of advanced NSCLC remains very poor. HOX transcript antisense intergenic RNA (HOTAIR) has been identified as an oncogenic long noncoding RNA (lncRNA) that is involved in the progression of a variety of carcinomas and acts as a negative prognostic biomarker. Yet, little is known about the effect of HOTAIR in the hypoxic microenvironment of NSCLC. The expression and promoter activity of HOTAIR were measured by quantitative real-time polymerase chain reaction (qRT-PCR) and luciferase reporter assay. The function of the hypoxia-inducible factor-1α (HIF-1α) binding site to hypoxia-responsive elements (HREs) in the HOTAIR promoter region was tested by luciferase reporter assay with nucleotide substitutions. The binding of HIF-1α to the HOTAIR promoter in vivo was confirmed by chromatin immunoprecipitation assay (CHIP) and electrophoretic mobility shift assay (EMSA). The effect of HIF-1α suppression by small interference RNA or YC-1 on HOTAIR expression was also determined. In the present study, we demonstrated that HOTAIR was upregulated by hypoxia in NSCLC cells. HOTAIR is a direct target of HIF-1α through interaction with putative HREs in the upstream region of HOTAIR in NSCLC cells. Furthermore, HIF-1α knockdown or inhibition could prevent HOTAIR upregulation under hypoxic conditions. Under hypoxic conditions, HOTAIR enhanced cancer cell proliferation, migration, and invasion. These data suggested that suppression of HOTAIR upon hypoxia of NSCLC could be a novel therapeutic strategy.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/genética , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , RNA Longo não Codificante/biossíntese , Apoptose/genética , Carcinoma Pulmonar de Células não Pequenas/patologia , Hipóxia Celular/genética , Linhagem Celular Tumoral , Movimento Celular/genética , Proliferação de Células/genética , Regulação Neoplásica da Expressão Gênica , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/biossíntese , Invasividade Neoplásica/genética , Invasividade Neoplásica/patologia , RNA Longo não Codificante/genética
7.
J Cardiovasc Pharmacol ; 66(2): 204-13, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25915513

RESUMO

We aimed to evaluate global changes in protein expression associated with patency by undertaking proteomic analysis of human constricted and patent ductus arteriosus (DA). Ten constricted and 10 patent human DAs were excised from infants with ductal-dependent heart disease during surgery. Using isobaric tags for relative and absolute quantitation-based quantitative proteomics, 132 differentially expressed proteins were identified. Of 132 proteins, voltage-gated sodium channel 1.3 (SCN3A), myosin 1d (Myo1d), Rho GTPase activating protein 26 (ARHGAP26), and retinitis pigmentosa 1 (RP1) were selected for validation by Western blot and quantitative real-time polymerase chain reaction analyses. Significant upregulation of SCN3A, Myo1d, and RP1 messenger RNA, and protein levels was observed in the patent DA group (all P ≤ 0.048). ARHGAP26 messenger RNA and protein levels were decreased in patent DA tissue (both P ≤ 0.018). Immunohistochemistry analysis revealed that Myo1d, ARHGAP26, and RP1 were specifically expressed in the subendothelial region of constricted DAs; however, diffuse expression of these proteins was noted in the patent group. Proteomic analysis revealed global changes in the expression of proteins that regulate oxygen sensing, ion channels, smooth muscle cell migration, nervous system, immune system, and metabolism, suggesting a basis for the systemic regulation of DA patency by diverse signaling pathways, which will be confirmed in further studies.


Assuntos
Permeabilidade do Canal Arterial/genética , Permeabilidade do Canal Arterial/cirurgia , Proteômica/métodos , Reação em Cadeia da Polimerase em Tempo Real/métodos , Vasoconstrição/genética , Permeabilidade do Canal Arterial/diagnóstico , Feminino , Humanos , Lactente , Recém-Nascido , Masculino
8.
Pediatr Cardiol ; 36(4): 743-51, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25500693

RESUMO

Patency of the ductus arteriosus (DA) after birth is essential in ductal-dependent congenital heart disease. The Na(+)/Ca(2+) exchanger (NCX) has been demonstrated to play a key role in regulating vascular tone. The potassium-dependent Na(+)/Ca(2+) exchanger (NCKX) is a related family of NCX depending on the K(+) gradients which triggers DA constriction. The present study investigated the comparative expression of NCX and NCKX between a constricted DA and patent DA in human ductal-dependant congenital heart disease. Human DAs, which were patent (n = 10, age = 20.2 ± 4.3 days) or constricted (n = 10, age = 18.3 ± 3.9 days), were excised during surgery from neonates with ductal-dependent congenital heart disease. Western blotting analysis, real-time quantitative polymerase chain reaction analysis and immunofluorescence studies were performed to detect the protein and mRNA levels of NCX1, NCKX3, and NCKX4. The expressions of NCX1 and NCKX4 were significantly higher in the patent DA group at both the protein and mRNA levels, and expression was localized to the smooth muscle layer. These findings indicate that NCX1 and NCKX4 are up-regulated in human postnatal patent DAs and may represent potential therapeutic targets for maintaining DA patency in ductal-dependent congenital heart disease.


Assuntos
Antiporters/metabolismo , Permeabilidade do Canal Arterial/metabolismo , Canal Arterial/anormalidades , Trocador de Sódio e Cálcio/metabolismo , Antiporters/genética , Western Blotting , Canal Arterial/patologia , Canal Arterial/cirurgia , Permeabilidade do Canal Arterial/genética , Permeabilidade do Canal Arterial/cirurgia , Feminino , Cardiopatias Congênitas/genética , Cardiopatias Congênitas/metabolismo , Cardiopatias Congênitas/fisiopatologia , Humanos , Lactente , Recém-Nascido , Masculino , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Trocador de Sódio e Cálcio/genética , Regulação para Cima
9.
J Obstet Gynaecol Res ; 40(6): 1578-83, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24888919

RESUMO

AIM: Constriction of small pulmonary arteries and high resistance of pulmonary circulation are important for maintaining fetal circulation before birth. In this study, we investigated how cytosolic free calcium concentration ([Ca(2+)]i) in fetal lamb small pulmonary artery smooth muscle cells (SPASMC) was affected by hypoxia and regulated by calcium pumps during this process. METHODS: (Ca(2+))i in response to acute hypoxia was determined spectrofluorometrically with fluo-3AM in cultured fetal SPASMC. Chemicals or solutions, including ryanodine, 2-aminoethoxydiphenyl borate, Ca(2+)-free solution with 20 mmol ethyleneglycoltetraacetic (EGTA), nimodipine, Na(+)-free medium and KB-R7943, were administrated at the same time point when samples were exposed to acute hypoxia. RESULTS: (Ca(2+))i in fetal lamb SPASMC increased under acute hypoxia. 2-Aminoethoxydiphenyl borate, an inhibitor of inositol triphosphate calcium store, partially attenuated the (Ca(2+))i increase after 6-min treatment. Ryanodine, an inhibitor of ryanodine-sensitive calcium stores, had no effect on the (Ca(2+))i increase. Ca(2+)-free solution with EGTA completely abolished this increase. Both nimodipine, that blocks the voltage-gated calcium channel, and KB-R7943, that inhibits the reverse mode of Na(+)/Ca(2+) exchanger, greatly diminished the hypoxia-induced (Ca(2+))i increase. The inhibitory effect of KB-R7943 was stronger than nimodipine, evidenced by the fact that (Ca(2+))i dropped near to the baseline level in the presence of KB-R7943 at a later time point. Low extracellular Na(+) concentration enhanced the hypoxia-induced increase of (Ca(2+))i. CONCLUSION: These results suggest that hypoxia-induced Ca(2+) increase in fetal SPASMC results from cytosolic Ca(2+) influx mediated primarily by the reverse mode of Na(+)/Ca(2+) exchanger.


Assuntos
Cálcio/metabolismo , Hipóxia/metabolismo , Miócitos de Músculo Liso/metabolismo , Artéria Pulmonar/metabolismo , Trocador de Sódio e Cálcio/metabolismo , Animais , Canais de Cálcio/metabolismo , Células Cultivadas , Citosol/metabolismo , Feto/fisiologia , Artéria Pulmonar/embriologia , Circulação Pulmonar , Ovinos , Vasoconstrição
10.
Biology (Basel) ; 13(4)2024 Mar 31.
Artigo em Inglês | MEDLINE | ID: mdl-38666846

RESUMO

Hemodynamics is the eternal theme of the circulatory system. Abnormal hemodynamics and cardiac and pulmonary development intertwine to form the most important features of children with congenital heart diseases (CHDs), thus determining these children's long-term quality of life. Here, we review the varieties of hemodynamic abnormalities that exist in children with CHDs, the recently developed neonatal rodent models of CHDs, and the inspirations these models have brought us in the areas of cardiomyocyte proliferation and maturation, as well as in alveolar development. Furthermore, current limitations, future directions, and clinical decision making based on these inspirations are highlighted. Understanding how CHD-associated hemodynamic scenarios shape postnatal heart and lung development may provide a novel path to improving the long-term quality of life of children with CHDs, transplantation of stem cell-derived cardiomyocytes, and cardiac regeneration.

11.
J Heart Lung Transplant ; 43(3): 496-507, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-37839791

RESUMO

BACKGROUD: Diseased animal models play an extremely important role in preclinical research. Lacking the corresponding animal models, many basic research studies cannot be carried out, and the conclusions obtained are incomplete or even incorrect. Right ventricular (RV) outflow tract (RVOT) obstruction leads to RV pressure overload (PO) and reduced pulmonary blood flow (RPF), which are 2 of the most important pathophysiological characteristics in pediatric cardiovascular diseases and seriously affect the survival rate and long-term quality of life of many children. Due to the lack of a neonatal mouse model for RVOT obstruction, it is largely unknown how RV PO and RPF regulate postnatal RV and pulmonary development. The aim of this study was to construct a neonatal RVOT obstruction mouse model. METHODS AND RESULTS: Here, we first introduced a neonatal mouse model of RVOT obstruction by pulmonary artery banding (PAB) on postnatal day 1. PAB induced neonatal RVOT obstruction, RV PO, and RPF. Neonatal RV PO induced cardiomyocyte proliferation, and neonatal RPF induced pulmonary dysplasia, the 2 features that are not observed in adult RVOT obstruction. As a result, PAB neonates exhibited overall developmental dysplasia, a sign similar to that of children with RVOT obstruction. CONCLUSIONS: Because many pediatric cardiovascular diseases are associated with RV PO and RPF, the introduction of a neonatal mouse model of RVOT obstruction may greatly enhance our understanding of these diseases and eventually improve or save the lives of many children.


Assuntos
Procedimentos Cirúrgicos Cardíacos , Doenças Cardiovasculares , Tetralogia de Fallot , Obstrução da Via de Saída Ventricular Direita , Obstrução do Fluxo Ventricular Externo , Humanos , Criança , Adulto , Recém-Nascido , Animais , Camundongos , Tetralogia de Fallot/cirurgia , Procedimentos Cirúrgicos Cardíacos/métodos , Artéria Pulmonar/cirurgia , Qualidade de Vida , Obstrução do Fluxo Ventricular Externo/etiologia , Obstrução do Fluxo Ventricular Externo/cirurgia
13.
J Vis Exp ; (196)2023 06 09.
Artigo em Inglês | MEDLINE | ID: mdl-37358276

RESUMO

Right ventricular (RV) volume overload (VO) is common in children with congenital heart disease. In view of distinct developmental stages,the RV myocardium may respond differently to VO in children compared to adults. The present study aims to establish a postnatal RV VO model in mice using a modified abdominal arteriovenous fistula. To confirm the creation of VO and the following morphological and hemodynamic changes of the RV, abdominal ultrasound, echocardiography, and histochemical staining were performed for 3 months. As a result, the procedure in postnatal mice showed an acceptable survival and fistula success rate. In VO mice, the RV cavity was enlarged with a thickened free wall, and the stroke volume was increased by about 30%-40% within 2 months after surgery. Thereafter, the RV systolic pressure increased, corresponding pulmonary valve regurgitation was observed, and small pulmonary artery remodeling appeared. In conclusion, modified arteriovenous fistula (AVF) surgery is feasible to establish the RV VO model in postnatal mice. Considering the probability of fistula closure and elevated pulmonary artery resistance, abdominal ultrasound and echocardiography must be performed to confirm the model status before application.


Assuntos
Insuficiência Cardíaca , Disfunção Ventricular Direita , Camundongos , Animais , Ventrículos do Coração/diagnóstico por imagem , Modelos Animais de Doenças , Volume Sistólico , Hemodinâmica , Função Ventricular Direita
14.
Cell Biosci ; 13(1): 112, 2023 Jun 19.
Artigo em Inglês | MEDLINE | ID: mdl-37337290

RESUMO

OBJECTIVES: Pulmonary vein stenosis (PVS), one of the most challenging clinical problems in congenital heart disease, leads to secondary pulmonary arterial hypertension (PAH) and right ventricular (RV) hypertrophy. Due to the lack of a rodent model, the mechanisms underlying PVS and its associated secondary effects are largely unknown, and treatments are minimally successful. This study developed a neonatal rat PVS model with the aim of increasing our understanding of the mechanisms and developing possible treatments for PVS. METHODS: PVS was created at postnatal day 1 (P1) by banding pulmonary veins that receive blood from the right anterior and mid lobes. The condition was confirmed using echocardiography, computed tomography (CT), gross anatomic examination, hematoxylin and eosin (H&E) staining, fibrosis staining, and immunofluorescence. Lung and RV remodeling under the condition of PVS were evaluated using H&E staining, fibrosis staining, and immunofluorescence. RESULTS: At P21, echocardiography revealed a change in wave form and a decrease in pulmonary artery acceleration time-indicators of PAH-at the transpulmonary valve site in the PVS group. CT at P21 showed a decrease in pulmonary vein diameter in the PVS group. At P30 in the PVS group, gross anatomic examination showed pulmonary congestion, H&E staining showed wall thickening and lumen narrowing in the upstream pulmonary veins, and immunofluorescence showed an increase in the smooth muscle layers in the upstream pulmonary veins. In addition, at P30 in the PVS group, lung remodeling was evidenced by hyperemia, thickening of pulmonary small vessel walls and smooth muscle layers, and reduction of the number of alveoli. RV remodeling was evidenced by an increase in RV free wall thickness. CONCLUSIONS: A neonatal rat model of PVS was successfully established, showing secondary lung and RV remodeling. This model may serve as a useful platform for understanding the mechanisms and treatments for PVS.

15.
Front Physiol ; 14: 1237187, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37908335

RESUMO

Introduction: Adult patients with atrial septal defects (ASD), the most common form of adult congenital heart disease, often die of arrhythmias, and the immaturity of cardiomyocytes contributes significantly to arrhythmias. ASD typically induces a left-to-right shunt, which then leads to the right atrium (RA) volume overload (VO). Whether or not VO contributes to RA cardiomyocyte immaturity and thereby causes arrhythmias in adult patients with ASD remains unclear. Methods: Here, we developed the first neonatal RA VO mouse model by creating a fistula between the inferior vena cava and abdominal aorta on postnatal day 7. RA VO was confirmed by increases in the mean flow velocity, mean pressure gradient, and velocity time integral across the tricuspid valve, and an increase in the RA diameter and RA area middle section. Results: We found that VO decreased the regularity and length of sarcomeres, and decreased the T-element density, regularity, and index of integrity of T-tubules in RA cardiomyocytes, suggesting that the two most important maturation hallmarks (sarcomere and T-tubules) of RA cardiomyocytes were impaired by VO. Accordingly, the calcium handling capacity of cardiomyocytes from postnatal day 21 (P21) RA was decreased by VO. VO caused a significant elongation of the PR interval. The expression of connexin 43 (Cx43) was decreased in RA VO. Moreover, gene ontology (GO) analysis of the downregulated genes in RA demonstrated that there was an abundance of enriched terms associated with sarcomeres and T-tubules exposed to VO. The results were further verified by qRT-PCR. Conclusions: In conclusion, the first neonatal RA VO mouse model was developed; furthermore, using this neonatal RA VO mouse model, we revealed that VO impeded RA sarcomere and T-tubule maturation, which may be the underlying causes of atrial arrhythmias in adult patients with ASD.

16.
Front Immunol ; 14: 1153573, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37449198

RESUMO

Objective: Inflammation is recognized as a contributor in the development of pulmonary arterial hypertension (PAH), and the recruitment and functional capacity of immune cells are well-orchestrated by chemokines and their receptors. This study is aimed at identification of critical chemokines in the progression of PAH via transcriptomic analysis. Methods: Differentially expressed genes (DEGs) from lungs of PAH patients were achieved compared to controls based on Gene Expression Omnibus (GEO) database. Gene set enrichment analysis (GSEA) was applied for functional annotation and pathway enrichement. The abundance of immune cells was estimated by the xCell algorithm. Weighted correlation network analysis (WGCNA) was used to construct a gene expression network, based on which a diagnostic model was generated to determine its accuracy to distinguish PAH from control subjects. Target genes were then validated in lung of hypoxia-induce pulmonary hypertension (PH) mouse model. Results: ACKR4 (atypical chemokine receptor 4) was downregulated in PAH lung tissues in multiple datasets. PAH relevant biological functions and pathways were enriched in patients with low-ACKR4 level according to GSEA enrichment analysis. Immuno-infiltration analysis revealed a negative correlation of activated dendritic cells, Th1 and macrophage infiltration with ACKR4 expression. Three gene modules were associated with PAH via WGCNA analysis, and a model for PAH diagnosis was generated using CXCL12, COL18A1 and TSHZ2, all of which correlated with ACKR4. The ACKR4 expression was also downregulated in lung tissues of our experimental PH mice compared to that of controls. Conclusions: The reduction of ACKR4 in lung tissues of human PAH based on transcriptomic data is consistent with the alteration observed in our rodent PH. The correlation with immune cell infiltration and functional annotation indicated that ACKR4 might serve as a protective immune checkpoint for PAH.


Assuntos
Hipertensão Pulmonar , Hipertensão Arterial Pulmonar , Humanos , Camundongos , Animais , Hipertensão Arterial Pulmonar/genética , Hipertensão Pulmonar Primária Familiar , Hipertensão Pulmonar/genética , Perfilação da Expressão Gênica , Pulmão
17.
Front Cardiovasc Med ; 10: 1164577, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37293289

RESUMO

Background: Copy number variations (CNVs) have been shown to be overrepresented in children with congenital heart disease (CHD). Genetic evaluation of CHD is currently underperformed in China. We sought to determine the occurrence of CNVs in CNV regions with disease-causing potential among a large cohort of Chinese pediatric CHD patients and investigate whether these CNVs could be the important critical modifiers of surgical intervention. Methods: CNVs screenings were performed in 1,762 Chinese children who underwent at least one cardiac surgery. CNV status at over 200 CNV locus with disease-causing potential was analyzed with a high-throughput ligation-dependent probe amplification (HLPA) assay. Results: We found 378 out of 1,762 samples (21.45%) to have at least one CNV and 2.38% of them were carrying multiple CNVs. The detection rates of ppCNVs (pathogenic and likely pathogenic CNVs) were 9.19% (162/1,762), significantly higher than that of the healthy Han Chinese individuals from The Database of Genomic Variants archive (9.19% vs. 3.63%; P = 0.0012). CHD cases with ppCNVs had a significantly higher proportion of complex surgeries compared to CHD patients with no ppCNVs (62.35% vs. 37.63%, P < 0.001). Duration of cardiopulmonary bypass and aortic cross clamp procedures were significantly longer in CHD cases with ppCNVs (all P < 0.05), while no group differences were identified for complications of surgery and one-month mortality after surgery. The detection rate of ppCNVs in the atrioventricular septal defect (AVSD) subgroup was significantly higher than that in other subgroups (23.10% vs. 9.70%, P = 0.002). Conclusions: CNV burden is an important contributor to Chinese children with CHD. Our study demonstrated the robustness and diagnostic efficiency of HLPA method in the genetic screening of CNVs in CHD patients.

18.
Dev Cell ; 58(10): 898-914.e7, 2023 05 22.
Artigo em Inglês | MEDLINE | ID: mdl-37071996

RESUMO

Cardiomyocyte differentiation continues throughout murine gestation and into the postnatal period, driven by temporally regulated expression changes in the transcriptome. The mechanisms that regulate these developmental changes remain incompletely defined. Here, we used cardiomyocyte-specific ChIP-seq of the activate enhancer marker P300 to identify 54,920 cardiomyocyte enhancers at seven stages of murine heart development. These data were matched to cardiomyocyte gene expression profiles at the same stages and to Hi-C and H3K27ac HiChIP chromatin conformation data at fetal, neonatal, and adult stages. Regions with dynamic P300 occupancy exhibited developmentally regulated enhancer activity, as measured by massively parallel reporter assays in cardiomyocytes in vivo, and identified key transcription factor-binding motifs. These dynamic enhancers interacted with temporal changes of the 3D genome architecture to specify developmentally regulated cardiomyocyte gene expressions. Our work provides a 3D genome-mediated enhancer activity landscape of murine cardiomyocyte development.


Assuntos
Elementos Facilitadores Genéticos , Miócitos Cardíacos , Animais , Camundongos , Cromatina , Regiões Promotoras Genéticas , Transcriptoma
19.
J Cardiol ; 79(1): 110-120, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34518077

RESUMO

BACKGROUND: Metabolic maturation is one of the primary processes of postnatal cardiomyocyte development. How volume overload (VO), a pathological state of the right ventricle (RV) in children with congenital heart disease (CHD) and patients with heart failure, affects cardiomyocyte metabolic maturation is unclear. METHODS AND RESULTS: A fistula between the abdominal aorta and inferior vena cava on postnatal day 7 (P7) was created in a mouse model to induce a young-aged RV VO. RNA sequencing revealed that the most enriched gene ontology (GO) terms of the upregulated transcriptome had been changed from metabolic maturation to heart contraction by VO. Transmission electron microscopy imaging showed that metabolic maturation marker-mitochondria were converted into the maturation style in the sham group while remaining unchanged in VO group. Calcium imaging showed that the calcium handling ability had slightly increased in the sham group but dramatically increased in the VO group, even with irregular contraction. Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis showed that the top three enriched KEGG pathways for the upregulated transcriptome during normal RV development were the citrate cycle, cardiac muscle contraction, and protein processing in the endoplasmic reticulum. VO changed those to arrhythmogenic RV cardiomyopathy, dilated cardiomyopathy, and hypertrophic cardiomyopathy. CONCLUSIONS: Metabolic maturation of postnatal RV development was partly interrupted by VO, and the underlining mechanism was associated with the activation of cardiomyopathy pathways.


Assuntos
Insuficiência Cardíaca , Ventrículos do Coração , Idoso , Animais , Humanos , Camundongos , Contração Miocárdica , Miócitos Cardíacos/metabolismo , Função Ventricular Direita/fisiologia
20.
Front Biosci (Landmark Ed) ; 27(10): 279, 2022 09 30.
Artigo em Inglês | MEDLINE | ID: mdl-36336858

RESUMO

BACKGROUND: This study aimed to investigate the mechanisms of acute rejection for vascularized composite allotransplantation (VCA) using microRNAs (miRNAs) differential expression in a VCA animal model. METHODS: Brown Norway rats were used as transplant donors and Lewis rats as VCA receptors. The changes were divided into different stages before and after transplantation in Lewis rats, and all appearance changes were recorded. Also, histological evaluations were performed on all recipients, and the expression of microRNAs was analyzed when acute immune rejection occurred. Then, we used GO and KEGG Pathway enrichment analyses to predict miRNA targets. Finally, differentially expressed miRNAs were detected by RT-qPCR. RESULTS: Compared to pre-operation, 22 miRNAs were differentially expressed after operations. Among them, nine were upregulated, and 13 were downregulated in skin tissues. The RT-qPCR results revealed that rno-miR-340-5p and rno-miR-21-5p were significantly upregulated and enriched in the PI3K-Akt signaling pathway. Moreover, rno-miR-145-5p and rno-miR-195-5p were significantly downregulated, and most of their target genes were enriched in the Hippo signaling pathway. The histological evaluations showed that, after VCA, the skin tissue presented severe acute rejection. CONCLUSIONS: The miRNAs rno-miR-340-5p, rno-miR-21-5p, rno-miR-145-5p, and rno-miR-195-5p were significantly regulated during VCA acute rejection, when the four miRNAs analyses were done on skin biopsies. These miRNAs might be potential biomarkers for objective, early, and minimally invasive rejection diagnosis.


Assuntos
MicroRNAs , Alotransplante de Tecidos Compostos Vascularizados , Animais , Ratos , Fosfatidilinositol 3-Quinases , Ratos Endogâmicos Lew , MicroRNAs/genética , MicroRNAs/metabolismo , Biomarcadores
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA