Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
1.
Int J Mol Sci ; 22(16)2021 Aug 16.
Artigo em Inglês | MEDLINE | ID: mdl-34445520

RESUMO

Endocytic trafficking is an under-appreciated pathway in cardiac development. Several genes related to endocytic trafficking have been uncovered in a mutagenic ENU screen, in which mutations led to congenital heart defects (CHDs). In this article, we review the relationship between these genes (including LRP1 and LRP2) and cardiac neural crest cells (CNCCs) during cardiac development. Mice with an ENU-induced Lrp1 mutation exhibit a spectrum of CHDs. Conditional deletion using a floxed Lrp1 allele with different Cre drivers showed that targeting neural crest cells with Wnt1-Cre expression replicated the full cardiac phenotypes of the ENU-induced Lrp1 mutation. In addition, LRP1 function in CNCCs is required for normal OFT lengthening and survival/expansion of the cushion mesenchyme, with other cell lineages along the NCC migratory path playing an additional role. Mice with an ENU-induced and targeted Lrp2 mutation demonstrated the cardiac phenotype of common arterial trunk (CAT). Although there is no impact on CNCCs in Lrp2 mutants, the loss of LRP2 results in the depletion of sonic hedgehog (SHH)-dependent cells in the second heart field. SHH is known to be crucial for CNCC survival and proliferation, which suggests LRP2 has a non-autonomous role in CNCCs. In this article, other endocytic trafficking proteins that are associated with CHDs that may play roles in the NCC pathway during development, such as AP1B1, AP2B1, FUZ, MYH10, and HECTD1, are reviewed.


Assuntos
Etilnitrosoureia/efeitos adversos , Cardiopatias Congênitas/genética , Proteína-1 Relacionada a Receptor de Lipoproteína de Baixa Densidade/genética , Proteína-2 Relacionada a Receptor de Lipoproteína de Baixa Densidade/genética , Animais , Linhagem da Célula , Modelos Animais de Doenças , Endocitose , Cardiopatias Congênitas/induzido quimicamente , Camundongos , Mutação , Crista Neural/metabolismo , Transdução de Sinais
2.
Am J Crit Care ; 32(3): 216-220, 2023 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-37121895

RESUMO

BACKGROUND: Children with congenital heart disease have unique risk factors associated with the pathophysiology of an abnormal heart; hence, this population is most likely at increased risk of acquiring a pressure injury during hospitalization. Few studies have included patients with congenital heart disease or examined the factors unique to these patients. OBJECTIVE: To identify risk factors associated with pressure injury development in children with congenital heart disease. METHODS: This retrospective study used a convenience sample from hospital-acquired data at an urban, tertiary, free-standing children's hospital. Patients were admitted to the intensive care unit between 2011 and 2018 with a diagnosis of congenital heart disease. Chi-square analysis was done to compare risk factors between patients, and logistic regression analysis was used to predict the probability that a patient would acquire a pressure injury. RESULTS: Eighty-two (30.5%) of the 269 patients in this study acquired pressure injuries. Sixty-six patients with pressure injuries met the inclusion criteria for analysis; 82% of those patients had had corticosteroids prescribed, and 71% were receiving anticoagulants. The overall predictive model for acquiring a pressure injury indicated an odds ratio of 3.25 (95% CI, 1.58-6.65) with an anticoagulant and an odds ratio of 9.98 (95% CI, 4.68-21.3) with a prescribed corticosteroid (P < .001 for both factors). Inpatient mortality was significantly associated with pressure injuries. CONCLUSIONS: Corticosteroid and anticoagulant use were contributing factors in the development of pressure injuries in children with congenital heart disease.


Assuntos
Cardiopatias Congênitas , Úlcera por Pressão , Humanos , Criança , Úlcera por Pressão/epidemiologia , Estudos Retrospectivos , Estado Terminal , Fatores de Risco , Cardiopatias Congênitas/complicações , Cardiopatias Congênitas/epidemiologia , Anticoagulantes
3.
J Am Heart Assoc ; 12(18): e029340, 2023 09 19.
Artigo em Inglês | MEDLINE | ID: mdl-37681527

RESUMO

Background Chromosomal microarray analysis (CMA) provides an opportunity to understand genetic causes of congenital heart disease (CHD). The methods for describing cardiac phenotypes in patients with CMA abnormalities have been inconsistent, which may complicate clinical interpretation of abnormal testing results and hinder a more complete understanding of genotype-phenotype relationships. Methods and Results Patients with CHD and abnormal clinical CMA were accrued from 9 pediatric cardiac centers. Highly detailed cardiac phenotypes were systematically classified and analyzed for their association with CMA abnormality. Hierarchical classification of each patient into 1 CHD category facilitated broad analyses. Inclusive classification allowing multiple CHD types per patient provided sensitive descriptions. In 1363 registry patients, 28% had genomic disorders with well-recognized CHD association, 67% had clinically reported copy number variants (CNVs) with rare or no prior CHD association, and 5% had regions of homozygosity without CNV. Hierarchical classification identified expected CHD categories in genomic disorders, as well as uncharacteristic CHDs. Inclusive phenotyping provided sensitive descriptions of patients with multiple CHD types, which occurred commonly. Among CNVs with rare or no prior CHD association, submicroscopic CNVs were enriched for more complex types of CHD compared with large CNVs. The submicroscopic CNVs that contained a curated CHD gene were enriched for left ventricular obstruction or septal defects, whereas CNVs containing a single gene were enriched for conotruncal defects. Neuronal-related pathways were over-represented in single-gene CNVs, including top candidate causative genes NRXN3, ADCY2, and HCN1. Conclusions Intensive cardiac phenotyping in multisite registry data identifies genotype-phenotype associations in CHD patients with abnormal CMA.


Assuntos
Cardiopatias Congênitas , Criança , Humanos , Cardiopatias Congênitas/diagnóstico , Cardiopatias Congênitas/genética , Coração , Genômica , Ventrículos do Coração , Análise em Microsséries
4.
Cell Stem Cell ; 29(5): 840-855.e7, 2022 05 05.
Artigo em Inglês | MEDLINE | ID: mdl-35395180

RESUMO

Hypoplastic left heart syndrome (HLHS) is a severe congenital heart disease with 30% mortality from heart failure (HF) in the first year of life, but the cause of early HF remains unknown. Induced pluripotent stem-cell-derived cardiomyocytes (iPSC-CM) from patients with HLHS showed that early HF is associated with increased apoptosis, mitochondrial respiration defects, and redox stress from abnormal mitochondrial permeability transition pore (mPTP) opening and failed antioxidant response. In contrast, iPSC-CM from patients without early HF showed normal respiration with elevated antioxidant response. Single-cell transcriptomics confirmed that early HF is associated with mitochondrial dysfunction accompanied with endoplasmic reticulum (ER) stress. These findings indicate that uncompensated oxidative stress underlies early HF in HLHS. Importantly, mitochondrial respiration defects, oxidative stress, and apoptosis were rescued by treatment with sildenafil to inhibit mPTP opening or TUDCA to suppress ER stress. Together these findings point to the potential use of patient iPSC-CM for modeling clinical heart failure and the development of therapeutics.


Assuntos
Cardiopatias Congênitas , Insuficiência Cardíaca , Células-Tronco Pluripotentes Induzidas , Antioxidantes/metabolismo , Cardiopatias Congênitas/metabolismo , Insuficiência Cardíaca/metabolismo , Humanos , Poro de Transição de Permeabilidade Mitocondrial , Miócitos Cardíacos/metabolismo , Estresse Oxidativo
5.
Front Cardiovasc Med ; 8: 734388, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34631832

RESUMO

Background: Congenital heart disease (CHD) with single-ventricle (SV) physiology is now survivable with a three-stage surgical course ending with Fontan palliation. However, 10-year transplant-free survival remains at 39-50%, with ventricular dysfunction progressing to heart failure (HF) being a common sequela. For SV-CHD patients who develop HF, undergoing the surgical course would not be helpful and could even be detrimental. As HF risk cannot be predicted and metabolic defects have been observed in Ohia SV-CHD mice, we hypothesized that respiratory defects in peripheral blood mononuclear cells (PBMCs) may allow HF risk stratification in SV-CHD. Methods: SV-CHD (n = 20), biventricular CHD (BV-CHD; n = 16), or healthy control subjects (n = 22) were recruited, and PBMC oxygen consumption rate (OCR) was measured using the Seahorse Analyzer. Respiration was similarly measured in Ohia mouse heart tissue. Results: Post-Fontan SV-CHD patients with HF showed higher maximal respiratory capacity (p = 0.004) and respiratory reserve (p < 0.0001), parameters important for cell stress adaptation, while the opposite was found for those without HF (reserve p = 0.037; maximal p = 0.05). This was observed in comparison to BV-CHD or healthy controls. However, respiration did not differ between SV patients pre- and post-Fontan or between pre- or post-Fontan SV-CHD patients and BV-CHD. Reminiscent of these findings, heart tissue from Ohia mice with SV-CHD also showed higher OCR, while those without CHD showed lower OCR. Conclusion: Elevated mitochondrial respiration in PBMCs is correlated with HF in post-Fontan SV-CHD, suggesting that PBMC respiration may have utility for prognosticating HF risk in SV-CHD. Whether elevated respiration may reflect maladaptation to altered hemodynamics in SV-CHD warrants further investigation.

6.
Dev Cell ; 55(4): 432-449.e12, 2020 11 23.
Artigo em Inglês | MEDLINE | ID: mdl-32966817

RESUMO

The etiology of congenital heart defects (CHDs), which are among the most common human birth defects, is poorly understood because of its complex genetic architecture. Here, we show that two genes implicated in CHDs, Megf8 and Mgrn1, interact genetically and biochemically to regulate the strength of Hedgehog signaling in target cells. MEGF8, a transmembrane protein, and MGRN1, a RING superfamily E3 ligase, assemble to form a receptor-like ubiquitin ligase complex that catalyzes the ubiquitination and degradation of the Hedgehog pathway transducer Smoothened. Homozygous Megf8 and Mgrn1 mutations increased Smoothened abundance and elevated sensitivity to Hedgehog ligands. While mice heterozygous for loss-of-function Megf8 or Mgrn1 mutations were normal, double heterozygous embryos exhibited an incompletely penetrant syndrome of CHDs with heterotaxy. Thus, genetic interactions can arise from biochemical mechanisms that calibrate morphogen signaling strength, a conclusion broadly relevant for the many human diseases in which oligogenic inheritance is emerging as a mechanism for heritability.


Assuntos
Coração/embriologia , Proteínas Hedgehog/metabolismo , Transdução de Sinais , Ubiquitinação , Alelos , Animais , Embrião de Mamíferos/metabolismo , Epistasia Genética , Dosagem de Genes , Proteínas de Membrana/metabolismo , Camundongos , Mutação/genética , Células NIH 3T3 , Fenótipo , Ligação Proteica , Receptor Smoothened/metabolismo , Ubiquitina-Proteína Ligases/metabolismo
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa