Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
1.
Am J Physiol Heart Circ Physiol ; 320(5): H1873-H1886, 2021 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-33739154

RESUMO

Compared with acyanotic congenital heart disease (CHD), cyanotic CHD has an increased risk of lifelong mortality and morbidity. These adverse outcomes may be attributed to delayed cardiomyocyte maturation, since the transition from a hypoxic fetal milieu to oxygen-rich postnatal environment is disrupted. We established a rodent model to replicate hypoxic myocardial conditions spanning perinatal development, and tested the hypothesis that chronic hypoxia impairs cardiac development. Pregnant mice were housed in hypoxia beginning at embryonic day 16. Pups stayed in hypoxia until postnatal day (P)8 when cardiac development is nearly complete. Global gene expression was quantified at P8 and at P30, after recovering in normoxia. Phenotypic testing included electrocardiogram, echocardiogram, and ex vivo electrophysiology study. Hypoxic P8 animals were 47% smaller than controls with preserved heart size. Gene expression was grossly altered by hypoxia at P8 (1,427 genes affected), but normalized after recovery (P30). Electrocardiograms revealed bradycardia and slowed conduction velocity in hypoxic animals at P8, with noticeable resolution after recovery (P30). Notable differences that persisted after recovery (P30) included a 65% prolongation in ventricular effective refractory period, sinus node dysfunction, 23% reduction in ejection fraction, and 16% reduction in fractional shortening in animals exposed to hypoxia. We investigated the impact of chronic hypoxia on the developing heart. Perinatal hypoxia was associated with changes in gene expression and cardiac function. Persistent changes to the electrophysiological substrate and contractile function warrant further investigation and may contribute to adverse outcomes observed in the cyanotic CHD population.NEW & NOTEWORTHY We utilized a new mouse model of chronic perinatal hypoxia to simulate the hypoxic myocardial conditions present in cyanotic congenital heart disease. Hypoxia caused numerous abnormalities in cardiomyocyte gene expression, the electrophysiologic substrate of the heart, and contractile function. Taken together, alterations observed in the neonatal period suggest delayed cardiac development immediately following hypoxia.


Assuntos
Cianose/etiologia , Coração Fetal/crescimento & desenvolvimento , Cardiopatias Congênitas/etiologia , Hipóxia/complicações , Fatores Etários , Animais , Animais Recém-Nascidos , Doença Crônica , Cianose/genética , Cianose/metabolismo , Cianose/fisiopatologia , Modelos Animais de Doenças , Feminino , Coração Fetal/metabolismo , Hipóxia Fetal/complicações , Hipóxia Fetal/genética , Hipóxia Fetal/metabolismo , Hipóxia Fetal/fisiopatologia , Regulação da Expressão Gênica no Desenvolvimento , Idade Gestacional , Cardiopatias Congênitas/genética , Cardiopatias Congênitas/metabolismo , Cardiopatias Congênitas/fisiopatologia , Frequência Cardíaca , Hipóxia/genética , Hipóxia/metabolismo , Hipóxia/fisiopatologia , Camundongos , Contração Miocárdica , Miócitos Cardíacos/metabolismo , Organogênese , Gravidez , Efeitos Tardios da Exposição Pré-Natal
2.
Ultrasound Med Biol ; 47(3): 751-758, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33293111

RESUMO

In vivo micro-imaging of mice is useful in studying the genetic basis of cardiac development in mutant embryos. We examined Phox2b-/- mutant mice, which lack autonomic innervation to the heart and die in utero, and investigated whether this lack of innervation causes cardiac dysfunction during embryogenesis. A VisualSonics Vevo 2100 ultrahigh-frequency linear array ultrasound machine with 30- and 40-MHz probes was used to analyze embryo size, gross characteristics, ventricular contractility and rhythm. Phox2b-/- mutant embryos underwent cessation of heartbeat and death at a greater rate than wild-type controls. We did not observe a hydrops phenotype or congenital heart defects in Phox2b-/- mutants. Analysis of heart rhythm revealed no significant correlation with genotype. Absent these signs of a progressive pathology, we suggest that Phox2b-/- mutant embryos likely die of sudden death secondary to acute arrhythmia. These data provide insight into the role of cardiac autonomic innervation during development.


Assuntos
Ecocardiografia/métodos , Perda do Embrião/genética , Coração/diagnóstico por imagem , Coração/embriologia , Proteínas de Homeodomínio/genética , Fatores de Transcrição/genética , Animais , Homozigoto , Camundongos , Camundongos Endogâmicos C57BL , Fenótipo
3.
Dis Model Mech ; 12(1)2019 01 11.
Artigo em Inglês | MEDLINE | ID: mdl-30578278

RESUMO

The development of the aortic arch is a complex process that involves remodeling of the bilaterally symmetrical pharyngeal arch arteries (PAAs) into the mature asymmetric aortic arch. Retinoic acid signaling is a key regulator of this process by directing patterning of the second heart field (SHF), formation of the caudal PAAs and subsequent remodeling of the PAAs to form the aortic arch. Here, we identify the HECTD1 ubiquitin ligase as a novel modulator of retinoic acid signaling during this process. Hectd1opm/opm homozygous mutant embryos show a spectrum of aortic arch abnormalities that occur following loss of 4th PAAs and increased SHF marker expression. This sequence of defects is similar to phenotypes observed in mutant mouse models with reduced retinoic acid signaling. Importantly, HECTD1 binds to and influences ubiquitination of the retinoic acid receptor, alpha (RARA). Furthermore, reduced activation of a retinoic acid response element (RARE) reporter is detected in Hectd1 mutant cells and embryos. Interestingly, Hectd1opm/+ heterozygous embryos exhibit reduced retinoic acid signaling, along with intermediate increased expression of SHF markers; however, heterozygotes show normal development of the aortic arch. Decreasing retinoic acid synthesis by reducing Raldh2 (also known as Aldh1a2) gene dosage in Hectd1opm/+ heterozygous embryos reveals a genetic interaction. Double heterozygous embryos show hypoplasia of the 4th PAA and increased incidence of a benign aortic arch variant, in which the transverse arch between the brachiocephalic and left common carotid arteries is shortened. Together, our data establish that HECTD1 is a novel regulator of retinoic acid signaling required for proper aortic arch development.


Assuntos
Aorta Torácica/embriologia , Aorta Torácica/metabolismo , Transdução de Sinais , Tretinoína/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Aldeído Oxirredutases/genética , Animais , Aorta Torácica/anormalidades , Aorta Torácica/patologia , Padronização Corporal , Região Branquial/irrigação sanguínea , Embrião de Mamíferos/metabolismo , Embrião de Mamíferos/patologia , Feminino , Dosagem de Genes , Coração/embriologia , Camundongos , Mutação/genética , Fenótipo , Ligação Proteica , Receptor alfa de Ácido Retinoico/metabolismo , Ubiquitinação
5.
Nat Genet ; 49(7): 1152-1159, 2017 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-28530678

RESUMO

Congenital heart disease (CHD) affects up to 1% of live births. Although a genetic etiology is indicated by an increased recurrence risk, sporadic occurrence suggests that CHD genetics is complex. Here, we show that hypoplastic left heart syndrome (HLHS), a severe CHD, is multigenic and genetically heterogeneous. Using mouse forward genetics, we report what is, to our knowledge, the first isolation of HLHS mutant mice and identification of genes causing HLHS. Mutations from seven HLHS mouse lines showed multigenic enrichment in ten human chromosome regions linked to HLHS. Mutations in Sap130 and Pcdha9, genes not previously associated with CHD, were validated by CRISPR-Cas9 genome editing in mice as being digenic causes of HLHS. We also identified one subject with HLHS with SAP130 and PCDHA13 mutations. Mouse and zebrafish modeling showed that Sap130 mediates left ventricular hypoplasia, whereas Pcdha9 increases penetrance of aortic valve abnormalities, both signature HLHS defects. These findings show that HLHS can arise genetically in a combinatorial fashion, thus providing a new paradigm for the complex genetics of CHD.


Assuntos
Heterogeneidade Genética , Síndrome do Coração Esquerdo Hipoplásico/genética , Sequência de Aminoácidos , Animais , Aorta/embriologia , Sistemas CRISPR-Cas , Mapeamento Cromossômico , Cromossomos Humanos/genética , Modelos Animais de Doenças , Exoma , Feminino , Edição de Genes , Técnicas de Inativação de Genes , Ventrículos do Coração/embriologia , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Mutação , Mutação de Sentido Incorreto , Miócitos Cardíacos/patologia , Penetrância , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos , Obstrução do Fluxo Ventricular Externo/genética , Peixe-Zebra/genética
6.
PLoS Genet ; 12(2): e1005821, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26918822

RESUMO

Heterotaxy, a birth defect involving left-right patterning defects, and primary ciliary dyskinesia (PCD), a sinopulmonary disease with dyskinetic/immotile cilia in the airway are seemingly disparate diseases. However, they have an overlapping genetic etiology involving mutations in cilia genes, a reflection of the common requirement for motile cilia in left-right patterning and airway clearance. While PCD is a monogenic recessive disorder, heterotaxy has a more complex, largely non-monogenic etiology. In this study, we show mutations in the novel dynein gene DNAH6 can cause heterotaxy and ciliary dysfunction similar to PCD. We provide the first evidence that trans-heterozygous interactions between DNAH6 and other PCD genes potentially can cause heterotaxy. DNAH6 was initially identified as a candidate heterotaxy/PCD gene by filtering exome-sequencing data from 25 heterotaxy patients stratified by whether they have airway motile cilia defects. dnah6 morpholino knockdown in zebrafish disrupted motile cilia in Kupffer's vesicle required for left-right patterning and caused heterotaxy with abnormal cardiac/gut looping. Similarly DNAH6 shRNA knockdown disrupted motile cilia in human and mouse respiratory epithelia. Notably a heterotaxy patient harboring heterozygous DNAH6 mutation was identified to also carry a rare heterozygous PCD-causing DNAI1 mutation, suggesting a DNAH6/DNAI1 trans-heterozygous interaction. Furthermore, sequencing of 149 additional heterotaxy patients showed 5 of 6 patients with heterozygous DNAH6 mutations also had heterozygous mutations in DNAH5 or other PCD genes. We functionally assayed for DNAH6/DNAH5 and DNAH6/DNAI1 trans-heterozygous interactions using subthreshold double-morpholino knockdown in zebrafish and showed this caused heterotaxy. Similarly, subthreshold siRNA knockdown of Dnah6 in heterozygous Dnah5 or Dnai1 mutant mouse respiratory epithelia disrupted motile cilia function. Together, these findings support an oligogenic disease model with broad relevance for further interrogating the genetic etiology of human ciliopathies.


Assuntos
Síndrome de Heterotaxia/genética , Síndrome de Kartagener/genética , Animais , Dineínas do Axonema/genética , Dineínas do Axonema/metabolismo , Padronização Corporal , Cílios/fisiologia , Embrião não Mamífero , Técnicas de Silenciamento de Genes , Heterozigoto , Humanos , Células de Kupffer/patologia , Camundongos Knockout , Mutação , RNA Interferente Pequeno/genética , Peixe-Zebra/embriologia , Peixe-Zebra/genética , Proteínas de Peixe-Zebra/genética , Proteínas de Peixe-Zebra/metabolismo
7.
J Thorac Cardiovasc Surg ; 151(4): 1126-35.e2, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26611747

RESUMO

OBJECTIVES: Patients with single ventricle congenital heart disease often form aortopulmonary collateral vessels via an unclear mechanism. To gain insights into the pathogenesis of aortopulmonary collateral vessels, we correlated angiogenic factor levels with in vitro activity and angiographic aortopulmonary collateral assessment and examined whether patients with single ventricle physiology have increased angiogenic factors that can stimulate endothelial cell sprouting in vitro. METHODS: In patients with single ventricle physiology (n = 27) and biventricular acyanotic control patients (n = 21), hypoxia-inducible angiogenic factor levels were measured in femoral venous and arterial plasma at cardiac catheterization. To assess plasma angiogenic activity, we used a 3-dimensional in vitro cell sprouting assay that recapitulates angiogenic sprouting. Aortopulmonary collateral angiograms were graded using a 4-point scale. RESULTS: Compared with controls, patients with single ventricle physiology had increased vascular endothelial growth factor (artery: 58.7 ± 1.2 pg/mL vs 35.3 ± 1.1 pg/mL, P < .01; vein: 34.8 ± 1.1 pg/mL vs 21 ± 1.2 pg/mL, P < .03), stromal-derived factor 1-alpha (artery: 1901.6 ± 1.1 pg/mL vs 1542.6 ± 1.1 pg/mL, P < .03; vein: 2092.8 pg/mL ± 1.1 vs 1752.9 ± 1.1 pg/mL, P < .02), and increased arterial soluble fms-like tyrosine kinase-1, a regulatory vascular endothelial growth factor receptor (612.3 ± 1.2 pg/mL vs 243.1 ± 1.2 pg/mL, P < .003). Plasma factors and sprout formation correlated poorly with aortopulmonary collateral severity. CONCLUSIONS: We are the first to correlate plasma angiogenic factor levels with angiography and in vitro angiogenic activity in patients with single ventricle disease with aortopulmonary collaterals. Patients with single ventricle disease have increased stromal-derived factor 1-alpha and soluble fms-like tyrosine kinase-1, and their roles in aortopulmonary collateral formation require further investigation. Plasma factors and angiogenic activity correlate poorly with aortopulmonary collateral severity in patients with single ventricles, suggesting complex mechanisms of angiogenesis.


Assuntos
Proteínas Angiogênicas/sangue , Aorta/fisiopatologia , Circulação Colateral , Células Endoteliais/metabolismo , Cardiopatias Congênitas/sangue , Neovascularização Fisiológica , Artéria Pulmonar/fisiopatologia , Circulação Pulmonar , Adolescente , Aortografia , Estudos de Casos e Controles , Células Cultivadas , Quimiocina CXCL12/sangue , Criança , Pré-Escolar , Feminino , Cardiopatias Congênitas/diagnóstico , Cardiopatias Congênitas/fisiopatologia , Humanos , Lactente , Masculino , Fator A de Crescimento do Endotélio Vascular/sangue , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/sangue
8.
Respir Med ; 109(9): 1126-30, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26233707

RESUMO

INTRODUCTION: Primary ciliary dyskinesia (PCD), a disease of impaired respiratory cilia motility, is often difficult to diagnose. Recent studies show low nasal nitric oxide (nNO) is closely linked to PCD, allowing the use of nNO measurement for PCD assessments. Nasal NO cutoff values for PCD are stratified by age, given nNO levels normally increase with age. However, normative values for nNO have not been established for infants less than 1 year old. In this study, we aim to establish normative values for nNO in infants and determine their utility in guiding infant PCD assessment. METHODS AND RESULTS: We obtained 42 nNO values from infants less than 1 year old without a history of PCD or recurrent sinopulmonary disease. Using regression analysis, we estimated the mean age-adjusted nNO values and established a 95% prediction interval (PI) for normal nNO. Using these findings, we were able to show 14 of 15 infant PCD patients had abnormally low nNO with values below the 95% PI. CONCLUSIONS: In this study we determined a regression model that best fits normative nNO values for infants less than 1 year old. This model identified the majority of PCD infants as having abnormally low nNO. These findings suggest nNO measurement can help guide PCD assessment in infants, and perhaps other pulmonary diseases with a link to low nNO. With early assessments, earlier clinical intervention may be possible to slow disease progression and help reduce pulmonary morbidity.


Assuntos
Síndrome de Kartagener/diagnóstico , Óxido Nítrico/metabolismo , Envelhecimento/metabolismo , Testes Respiratórios/métodos , Estudos de Viabilidade , Humanos , Lactente , Recém-Nascido , Cavidade Nasal , Valores de Referência
9.
Nature ; 521(7553): 520-4, 2015 May 28.
Artigo em Inglês | MEDLINE | ID: mdl-25807483

RESUMO

Congenital heart disease (CHD) is the most prevalent birth defect, affecting nearly 1% of live births; the incidence of CHD is up to tenfold higher in human fetuses. A genetic contribution is strongly suggested by the association of CHD with chromosome abnormalities and high recurrence risk. Here we report findings from a recessive forward genetic screen in fetal mice, showing that cilia and cilia-transduced cell signalling have important roles in the pathogenesis of CHD. The cilium is an evolutionarily conserved organelle projecting from the cell surface with essential roles in diverse cellular processes. Using echocardiography, we ultrasound scanned 87,355 chemically mutagenized C57BL/6J fetal mice and recovered 218 CHD mouse models. Whole-exome sequencing identified 91 recessive CHD mutations in 61 genes. This included 34 cilia-related genes, 16 genes involved in cilia-transduced cell signalling, and 10 genes regulating vesicular trafficking, a pathway important for ciliogenesis and cell signalling. Surprisingly, many CHD genes encoded interacting proteins, suggesting that an interactome protein network may provide a larger genomic context for CHD pathogenesis. These findings provide novel insights into the potential Mendelian genetic contribution to CHD in the fetal population, a segment of the human population not well studied. We note that the pathways identified show overlap with CHD candidate genes recovered in CHD patients, suggesting that they may have relevance to the more complex genetics of CHD overall. These CHD mouse models and >8,000 incidental mutations have been sperm archived, creating a rich public resource for human disease modelling.


Assuntos
Cílios/patologia , Cardiopatias Congênitas/genética , Cardiopatias Congênitas/patologia , Animais , Cílios/diagnóstico por imagem , Cílios/genética , Cílios/fisiologia , Análise Mutacional de DNA , Eletrocardiografia , Exoma/genética , Genes Recessivos , Testes Genéticos , Cardiopatias Congênitas/diagnóstico por imagem , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Mutação/genética , Transdução de Sinais , Ultrassonografia
10.
Ann Am Thorac Soc ; 11(9): 1426-32, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25302410

RESUMO

RATIONALE: Patients with congenital heart disease with heterotaxy exhibit a high prevalence of abnormal airway ciliary motion and low nasal nitric oxide, characteristics associated with primary ciliary dyskinesia, a reflection of the role of motile cilia in airway clearance and left-right patterning. OBJECTIVES: To assess the potential broader clinical significance of airway ciliary dysfunction in congenital heart disease, we assessed the prevalence of ciliary dysfunction versus respiratory symptoms in patients with congenital heart disease with or without heterotaxy. METHODS: Patients with a broad spectrum of congenital heart disease were recruited (n = 218), 39 with heterotaxy. Nasal nitric oxide measurements and nasal biopsies for ciliary motion video microscopy were conducted. Sinopulmonary symptoms were reviewed by questionnaire. MEASUREMENTS AND MAIN RESULTS: A high prevalence of ciliary motion defects (51.8%) and low or borderline low nasal nitric oxide levels (35.5%) were observed in patients with congenital heart disease with or without heterotaxy. Patients with ciliary motion defects or low nasal nitric oxide showed increased sinopulmonary symptoms, with most respiratory symptoms seen in those with both abnormal ciliary motion and low nitric oxide. Multivariate analysis showed that abnormal ciliary motion and low nasal nitric oxide were more important in determining risk of sinopulmonary symptoms than heterotaxy status. CONCLUSIONS: Patients with congenital heart disease without heterotaxy exhibit a high prevalence of abnormal ciliary motion and low nasal nitric oxide. This was associated with more sinopulmonary symptoms. These findings suggest that patients with a broad spectrum of congenital heart disease and respiratory symptoms may benefit from screening for ciliary dysfunction and implementation of medical interventions to reduce sinopulmonary morbidities.


Assuntos
Transtornos da Motilidade Ciliar/diagnóstico , Cardiopatias Congênitas/complicações , Síndrome de Heterotaxia/complicações , Adolescente , Adulto , Testes Respiratórios , Criança , Transtornos da Motilidade Ciliar/complicações , Feminino , Humanos , Masculino , Microscopia de Vídeo , Pessoa de Meia-Idade , Análise Multivariada , Óxido Nítrico/metabolismo , Otite Média/complicações , Hipersensibilidade Respiratória/complicações , Doenças Respiratórias/complicações , Adulto Jovem
11.
Pediatr Res ; 76(6): 500-7, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25167202

RESUMO

BACKGROUND: Mouse mutants are used to model human congenital cardiovascular disease. Few studies exist comparing normal cardiovascular development in mice vs. humans. We carried out a systematic comparative analysis of mouse and human fetal cardiovascular development. METHODS: Episcopic fluorescence image capture (EFIC) was performed on 66 wild-type mouse embryos from embryonic day (E) 9.5 to birth; 2-dimensional and 3-dimensional datasets were compared with EFIC and magnetic resonance images from a study of 52 human fetuses (Carnegie stage 13-23). RESULTS: Time course of atrial, ventricular, and outflow septation were outlined and followed a similar sequence in both species. Bilateral venae cavae and prominent atrial appendages were seen in the mouse fetus; in human fetuses, atrial appendages were small, and a single right superior vena cava was present. In contrast to humans with separate pulmonary vein orifices, a pulmonary venous confluence with one orifice enters the left atrium in mice. CONCLUSION: The cardiac developmental sequences observed in mouse and human fetuses are comparable, with minor differences in atrial and venous morphology. These comparisons of mouse and human cardiac development strongly support that mouse morphogenesis is a good model for human development.


Assuntos
Coração Fetal/embriologia , Coração/embriologia , Animais , Apêndice Atrial/embriologia , Septo Interatrial/embriologia , Idade Gestacional , Valvas Cardíacas/embriologia , Ventrículos do Coração/embriologia , Humanos , Imageamento por Ressonância Magnética , Camundongos , Morfogênese , Imagem Óptica , Especificidade da Espécie , Septo Interventricular/embriologia
12.
Circ Cardiovasc Imaging ; 7(1): 31-42, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24319090

RESUMO

BACKGROUND: Congenital heart disease (CHD) has a multifactorial pathogenesis, but a genetic contribution is indicated by heritability studies. To investigate the spectrum of CHD with a genetic pathogenesis, we conducted a forward genetic screen in inbred mice using fetal echocardiography to recover mutants with CHD. Mice are ideally suited for these studies given that they have the same four-chamber cardiac anatomy that is the substrate for CHD. METHODS AND RESULTS: Ethylnitrosourea mutagenized mice were ultrasound-interrogated by fetal echocardiography using a clinical ultrasound system, and fetuses suspected to have cardiac abnormalities were further interrogated with an ultrahigh-frequency ultrasound biomicroscopy. Scanning of 46 270 fetuses revealed 1722 with cardiac anomalies, with 27.9% dying prenatally. Most of the structural heart defects can be diagnosed using ultrasound biomicroscopy but not with the clinical ultrasound system. Confirmation with analysis by necropsy and histopathology showed excellent diagnostic capability of ultrasound biomicroscopy for most CHDs. Ventricular septal defect was the most common CHD observed, whereas outflow tract and atrioventricular septal defects were the most prevalent complex CHD. Cardiac/visceral organ situs defects were observed at surprisingly high incidence. The rarest CHD found was hypoplastic left heart syndrome, a phenotype never seen in mice previously. CONCLUSIONS: We developed a high-throughput, 2-tier ultrasound phenotyping strategy for efficient recovery of even rare CHD phenotypes, including the first mouse models of hypoplastic left heart syndrome. Our findings support a genetic pathogenesis for a wide spectrum of CHDs and suggest that the disruption of left-right patterning may play an important role in CHD.


Assuntos
Ecocardiografia Doppler , Coração Fetal/diagnóstico por imagem , Testes Genéticos , Cardiopatias Congênitas/diagnóstico por imagem , Cardiopatias Congênitas/genética , Microscopia Acústica , Mutação , Ultrassonografia Pré-Natal/métodos , Animais , Modelos Animais de Doenças , Ecocardiografia Doppler em Cores , Etilnitrosoureia/toxicidade , Feminino , Coração Fetal/anormalidades , Predisposição Genética para Doença , Cardiopatias Congênitas/embriologia , Hereditariedade , Ensaios de Triagem em Larga Escala , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Linhagem , Fenótipo
13.
J Thorac Cardiovasc Surg ; 147(4): 1291-1298.e2, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23886032

RESUMO

OBJECTIVE(S): Congenital heart disease (CHD) and heterotaxy patients have increased postoperative and respiratory complications. We recently showed CHD-heterotaxy patients can have respiratory ciliary dysfunction (CD) similar to that associated with primary ciliary dyskinesia, including low nasal nitric oxide and abnormal ciliary motion. In this study, we investigated whether CHD-heterotaxy patients with CD may have worse postsurgical outcomes. METHODS: We examined postsurgical outcome in 13 heterotaxy-CHD patients with CD (25 surgeries), compared with 14 heterotaxy-CHD patients without CD (27 surgeries). Outcome data were collected for each surgery, including respiratory complications, tracheostomy, use of inhaled ß-agonists or nitric oxide, length of hospital stay, days on ventilator, and death. RESULTS: The CD versus the no-CD CHD cohorts had similar Risk Adjustment in Congenital Heart Surgery-1 risk categories, repair track, age at surgery, and follow-up evaluation times. Respiratory complications (76% vs 37%; P = .006), need for tracheostomy (16% vs 0%; P = .047), and use of inhaled ß-agonists (64% vs 11%; P = .0001) all were increased significantly in heterotaxy-CHD patients with CD. No significant differences were detected in postoperative hospital stay, days on mechanical ventilation, or surgical mortality. A trend toward increased mortality for the CD group beyond the postoperative period was observed (33% vs 0%; P = .055) in patients younger than age 10 years. CONCLUSIONS: Our findings showed that heterotaxy-CHD patients with CD may have increased risks for respiratory deficiencies. Overall, there was a trend toward increased mortality in CD patients with intermediate follow-up evaluation. Because ß-agonists are known to increase ciliary beat frequency, presurgical screening for CD and perioperative treatment of CD patients with inhaled ß-agonists may improve postoperative outcomes and survival.


Assuntos
Transtornos da Motilidade Ciliar/complicações , Síndrome de Heterotaxia/complicações , Síndrome de Heterotaxia/cirurgia , Complicações Pós-Operatórias/epidemiologia , Complicações Pós-Operatórias/etiologia , Transtornos Respiratórios/epidemiologia , Transtornos Respiratórios/etiologia , Adolescente , Adulto , Criança , Pré-Escolar , Feminino , Cardiopatias Congênitas/complicações , Cardiopatias Congênitas/cirurgia , Humanos , Lactente , Recém-Nascido , Masculino , Pessoa de Meia-Idade , Estudos Prospectivos , Resultado do Tratamento , Adulto Jovem
14.
Circ Cardiovasc Imaging ; 6(4): 551-9, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23759365

RESUMO

BACKGROUND: Mice are well suited for modeling human congenital heart disease (CHD), given their 4-chamber cardiac anatomy. However, mice with CHD invariably die prenatally/neonatally, causing CHD phenotypes to be missed. Therefore, we investigated the efficacy of noninvasive microcomputed tomography (micro-CT) to screen for CHD in stillborn/fetal mice. These studies were performed using chemically mutagenized mice expected to be enriched for birth defects, including CHD. METHODS AND RESULTS: Stillborn/fetal mice obtained from the breeding of N-ethyl-N-nitrosourea mutagenized mice were formalin-fixed and stained with iodine, then micro-CT scanned. Those diagnosed with CHD and some CHD-negative pups were necropsied. A subset of these were further analyzed by histopathology to confirm the CHD/no-CHD diagnosis. Micro-CT scanning of 2105 fetal/newborn mice revealed an abundance of ventricular septal defects (n=307). Overall, we observed an accuracy of 89.8% for ventricular septal defect diagnosis. Outflow tract anomalies identified by micro-CT included double outlet right ventricle (n=36), transposition of the great arteries (n=14), and persistent truncus arteriosus (n=3). These were diagnosed with a 97.4% accuracy. Aortic arch anomalies also were readily detected with an overall 99.6% accuracy. This included right aortic arch (n=28) and coarctation/interrupted aortic arch (n=12). Also detected by micro-CT were atrioventricular septal defects (n=22), tricuspid hypoplasia/atresia (n=13), and coronary artery fistulas (n=16). They yielded accuracies of 98.9%, 100%, and 97.8%, respectively. CONCLUSIONS: Contrast enhanced micro-CT imaging in neonatal/fetal mice can reliably detect a wide spectrum of CHD. We conclude that micro-CT imaging can be used for routine rapid assessments of structural heart defects in fetal/newborn mice.


Assuntos
Coração Fetal/anormalidades , Coração Fetal/diagnóstico por imagem , Cardiopatias Congênitas/diagnóstico por imagem , Diagnóstico Pré-Natal/métodos , Microtomografia por Raio-X , Animais , Animais Recém-Nascidos , Meios de Contraste , Modelos Animais de Doenças , Coração Fetal/patologia , Genótipo , Idade Gestacional , Cardiopatias Congênitas/genética , Cardiopatias Congênitas/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Variações Dependentes do Observador , Fenótipo , Valor Preditivo dos Testes , Reprodutibilidade dos Testes
15.
Circulation ; 125(18): 2232-42, 2012 May 08.
Artigo em Inglês | MEDLINE | ID: mdl-22499950

RESUMO

BACKGROUND: Patients with congenital heart disease (CHD) and heterotaxy show high postsurgical morbidity/mortality, with some developing respiratory complications. Although this finding is often attributed to the CHD, airway clearance and left-right patterning both require motile cilia function. Thus, airway ciliary dysfunction (CD) similar to that of primary ciliary dyskinesia (PCD) may contribute to increased respiratory complications in heterotaxy patients. METHODS AND RESULTS: We assessed 43 CHD patients with heterotaxy for airway CD. Videomicrocopy was used to examine ciliary motion in nasal tissue, and nasal nitric oxide (nNO) was measured; nNO level is typically low with PCD. Eighteen patients exhibited CD characterized by abnormal ciliary motion and nNO levels below or near the PCD cutoff values. Patients with CD aged >6 years show increased respiratory symptoms similar to those seen in PCD. Sequencing of all 14 known PCD genes in 13 heterotaxy patients with CD, 12 without CD, 10 PCD disease controls, and 13 healthy controls yielded 0.769, 0.417, 1.0, and 0.077 novel variants per patient, respectively. One heterotaxy patient with CD had the PCD causing DNAI1 founder mutation. Another with hyperkinetic ciliary beat had 2 mutations in DNAH11, the only PCD gene known to cause hyperkinetic beat. Among PCD patients, 2 had known PCD causing CCDC39 and CCDC40 mutations. CONCLUSIONS: Our studies show that CHD patients with heterotaxy have substantial risk for CD and increased respiratory disease. Heterotaxy patients with CD were enriched for mutations in PCD genes. Future studies are needed to assess the potential benefit of prescreening and prophylactically treating heterotaxy patients for CD.


Assuntos
Transtornos da Motilidade Ciliar/epidemiologia , Cardiopatias Congênitas/epidemiologia , Síndrome de Heterotaxia/epidemiologia , Anormalidades do Sistema Respiratório/epidemiologia , Adolescente , Adulto , Dineínas do Axonema/genética , Testes Respiratórios , Criança , Pré-Escolar , Transtornos da Motilidade Ciliar/genética , Proteínas do Citoesqueleto , Feminino , Cardiopatias Congênitas/genética , Síndrome de Heterotaxia/genética , Humanos , Lactente , Masculino , Microscopia de Vídeo , Pessoa de Meia-Idade , Mutação , Óxido Nítrico/análise , Prevalência , Proteínas/genética , Anormalidades do Sistema Respiratório/genética , Adulto Jovem
16.
J Thorac Cardiovasc Surg ; 141(3): 637-44, 644.e1-3, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20884020

RESUMO

OBJECTIVE: Patients with heterotaxy and complex congenital heart disease underwent cardiac surgery with high mortality and morbidity. Recent studies have revealed an association among heterotaxy, congenital heart disease, and primary ciliary dyskinesia. We undertook a retrospective review of patients undergoing cardiac surgery at Children's National Medical Center between 2004 and 2008 to explore the hypothesis that there is increased mortality and respiratory complications in heterotaxy patients. METHODS: Retrospective review was performed on postsurgical outcomes of 87 patients with heterotaxy and congenital heart disease exhibiting the full spectrum of situs abnormalities associated with heterotaxy. As controls patients, 634 cardiac surgical patients with congenital heart disease, but without laterality defects, were selected, and surgical complexities were similar with a median Risk Adjustment in Congenital Heart Surgery-1 score of 3.0 for both groups. RESULTS: We found the mean length of postoperative hospital stay (17 vs 11 days) and mechanical ventilation (11 vs 4 days) were significantly increased in the heterotaxy patients. Also elevated were rates of tracheostomies (6.9% vs 1.6%; odds ratio, 4.6), extracorporeal membrane oxygenation support (12.6% vs 4.9%: odds ratio, 2.8), prolonged ventilatory courses (23% vs 12.3%; odds ratio, 2.1) and postsurgical deaths (16.1% vs 4.7%; odds ratio, 3.9). CONCLUSIONS: Our findings show heterotaxy patients had more postsurgical events with increased postsurgical mortality and risk for respiratory complications as compared to control patients with similar Risk Adjustment in Congenital Heart Surgery-1 surgical complexity scores. We speculate that increased respiratory complications maybe due to ciliary dysfunction. Further studies are needed to explore the basis for the increased surgical risks for heterotaxy patients undergoing cardiac surgery.


Assuntos
Anormalidades Múltiplas , Procedimentos Cirúrgicos Cardíacos/efeitos adversos , Cardiopatias Congênitas/cirurgia , Transtornos Respiratórios/etiologia , Adolescente , Adulto , Procedimentos Cirúrgicos Cardíacos/mortalidade , Estudos de Casos e Controles , Distribuição de Qui-Quadrado , Criança , Pré-Escolar , Dextrocardia/complicações , Dextrocardia/mortalidade , District of Columbia , Oxigenação por Membrana Extracorpórea , Feminino , Doenças Genéticas Ligadas ao Cromossomo X/complicações , Doenças Genéticas Ligadas ao Cromossomo X/mortalidade , Cardiopatias Congênitas/complicações , Cardiopatias Congênitas/mortalidade , Síndrome de Heterotaxia , Humanos , Lactente , Recém-Nascido , Estimativa de Kaplan-Meier , Síndrome de Kartagener/complicações , Síndrome de Kartagener/mortalidade , Tempo de Internação , Masculino , Razão de Chances , Transtornos Respiratórios/mortalidade , Transtornos Respiratórios/terapia , Respiração Artificial , Estudos Retrospectivos , Medição de Risco , Fatores de Risco , Situs Inversus/complicações , Situs Inversus/mortalidade , Traqueostomia , Resultado do Tratamento , Adulto Jovem
17.
J Am Soc Echocardiogr ; 23(3): 315-23, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20097527

RESUMO

BACKGROUND: The authors conducted an ultrasound interrogation of a mutant mouse model with a Dnah5 mutation to determine whether cardiac mechanics may be affected by reversal of cardiac situs. This mutant is a bona fide model of primary ciliary dyskinesia, with surviving homozygous mice showing either situs solitus (SS) or situs inversus totalis (SI). METHODS: High-frequency ultrasound interrogations of 27 neonatal and infant Dnah5 mutant mice, 16 with SS and 11 with SI, were conducted using an ultra-high-frequency biomicroscope. Electrocardiographic and respiratory gating were used to reconstruct high-resolution two-dimensional cines at 1,000 Hz, with speckle-tracking echocardiography used to further analyze midchamber and apical rotation. RESULTS: All SS mice exhibited the expected counterclockwise apical rotation as viewed caudocranially, and surprisingly, the same counterclockwise motion was also observed in SI mice. Speckle-tracking analysis confirmed counterclockwise systolic rotation in both SS and SI mice, and this increased in magnitude from the subepicardium to the endocardium and from the papillary muscles to the apex. The magnitude of apical endocardial rotation was not different for SS and SI mice (5.64+/-0.75 degrees and 5.76+/-1.90 degrees, respectively, P=.93). The anatomic segments responsible for the largest components of apical endocardial systolic rotation differed between the SS and SI hearts (P=.004). In both, the two largest contributors to rotation were offset 180 degrees from each other, but the anatomic regions differed between them. In SS hearts, maximal regional rotation occurred at the anterior mid-septum and posterolateral free wall, while in SI hearts, it was derived from the posterior septum and the anterolateral free wall. Analysis by episcopic fluorescence image capture histology of representative SI and SS mice showed normal intracardiac and segmental anatomy ({S,D,S} or {I,L,I}) without intracardiac defects. CONCLUSIONS: These results show that mirror-image cardiac looping did not result in mirror-image rotation of the morphologic left ventricle. These findings suggest that further studies are warranted to evaluate whether fiber orientation and cardiac mechanics may be abnormal in individuals with reversal of cardiac situs. The results of this study indicate that cardiac looping and myofiber orientation may be independently regulated.


Assuntos
Ecocardiografia/métodos , Técnicas de Imagem por Elasticidade/métodos , Interpretação de Imagem Assistida por Computador/métodos , Situs Inversus/diagnóstico por imagem , Anormalidade Torcional/diagnóstico por imagem , Disfunção Ventricular Esquerda/diagnóstico por imagem , Animais , Camundongos , Camundongos Knockout , Mutação , Reprodutibilidade dos Testes , Rotação , Sensibilidade e Especificidade , Situs Inversus/complicações , Anormalidade Torcional/etiologia , Disfunção Ventricular Esquerda/etiologia
19.
Ultrasound Med Biol ; 34(5): 741-52, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18328616

RESUMO

To establish a developmental profile of fetal mouse cardiovascular parameters, we analyzed a large body of ultrasound measurements obtained by in utero echocardiography of C57BL/6J fetal mice from embryonic day 12.5 to 19.5 (term). Measurements were obtained using two-dimensional (2D), spectral Doppler and M-mode imaging with standard clinical cardiac ultrasound imaging planes. As these studies were conducted as part of a large scale mouse mutagenesis screen, stringent filtering criteria were used to eliminate potentially abnormal fetuses. Our analysis showed heart rate increased from 190 to 245 beats per minute as the mouse fetus grew from 8 mm at embryonic day 12.5 to 18.7 mm at term. This was accompanied by increases in peak outflow velocity, E-wave, E/A ratio and ventricular dimensions. In contrast, the A-wave, myocardial performance index and isovolemic contraction time decreased gradually. Systolic function remained remarkably stable at 80% ejection fraction. Analysis of intra- and interobserver variabilities showed these parameters were reproducible, with most comparing favorably to clinical ultrasound measurements in human fetuses. A comprehensive database was generated comprising 23 echocardiographic parameters delineating fetal mouse cardiovascular function from embryonic day 12.5 to term. This database can serve as a standard for evaluating cardiovascular pathophysiology in genetically altered and mutant mouse models.


Assuntos
Coração Fetal/diagnóstico por imagem , Ultrassonografia Pré-Natal/métodos , Animais , Ecocardiografia Doppler , Feminino , Coração Fetal/embriologia , Idade Gestacional , Frequência Cardíaca Fetal , Camundongos , Camundongos Endogâmicos C57BL , Modelos Animais , Gravidez , Fluxo Sanguíneo Regional , Processamento de Sinais Assistido por Computador , Volume Sistólico , Sístole
20.
Pediatr Res ; 63(1): 9-14, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18043505

RESUMO

Heterotaxy arises from a failure of the embryo to establish normal left-right asymmetry and is known to affect 3% of infants with congenital heart disease. A recessive mutation causing heterotaxy was recovered in a mouse mutagenesis screen focused on congenital heart defects. Homozygote mutants exhibit abnormal situs in the thoracic and abdominal cavities. Dextrocardia, levocardia, or mesocardia was seen together with right pulmonary isomerism and complex structural heart defects in the single ventricle spectrum. A dominant chamber of left ventricular morphology positioned on the left or right is seen together with transposition of the great arteries. Right atrial isomerism with or without total anomalous pulmonary venous connection was observed in half of the mutants. Because ciliary motion at the embryonic node is required for the specification of laterality, we examined the tracheal epithelia of newborn mice as a proxy for the nodal cilia. However, videomicroscopy showed no defect in ciliary motion. Genome scanning using polymorphic microsatellite markers mapped the mutation to a 3.3 Mb interval on mouse chromosome 7. None of the genes previously described for familial heterotaxy were found in this interval, indicating a novel mutation in this mouse model of heterotaxy.


Assuntos
Anormalidades Múltiplas/patologia , Padronização Corporal/genética , Cromossomos de Mamíferos , Cardiopatias Congênitas/patologia , Mutação , Anormalidades Múltiplas/embriologia , Anormalidades Múltiplas/genética , Animais , Animais Recém-Nascidos , Mapeamento Cromossômico , Cílios/patologia , Dextrocardia/genética , Dextrocardia/patologia , Modelos Animais de Doenças , Átrios do Coração/patologia , Cardiopatias Congênitas/embriologia , Cardiopatias Congênitas/genética , Ventrículos do Coração/anormalidades , Homozigoto , Interpretação de Imagem Assistida por Computador , Imageamento Tridimensional , Levocardia/genética , Levocardia/patologia , Camundongos , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Microscopia de Fluorescência , Microscopia de Vídeo , Fenótipo , Polidactilia/genética , Polidactilia/patologia , Veias Pulmonares/anormalidades , Mucosa Respiratória/patologia , Transposição dos Grandes Vasos/genética , Transposição dos Grandes Vasos/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA