Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 202
Filter
1.
Contemp Clin Trials Commun ; 38: 101257, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38298917

ABSTRACT

Background: Registry-based trials have the potential to reduce randomized clinical trial (RCT) costs. However, observed cost differences also may be achieved through pragmatic trial designs. A systematic comparison of trial costs across different designs has not been previously performed. Methods: We conducted a study to compare the current Steroids to Reduce Systemic inflammation after infant heart surgery (STRESS) registry-based RCT vs. two established designs: pragmatic RCT and explanatory RCT. The primary outcome was total RCT design costs. Secondary outcomes included: RCT duration and personnel hours. Costs were estimated using the Duke Clinical Research Institute's pricing model. Results: The Registry-Based RCT estimated duration was 31.9 weeks greater than the other designs (259.5 vs. 227.6 weeks). This delay was caused by the Registry-Based design's periodic data harvesting that delayed site closing and statistical reporting. Total personnel hours were greatest for the Explanatory design followed by the Pragmatic design and the Registry-Based design (52,488 vs 29,763 vs. 24,480 h, respectively). Total costs were greatest for the Explanatory design followed by the Pragmatic design and the Registry-Based design ($10,140,263 vs. $4,164,863 vs. $3,268,504, respectively). Thus, Registry-Based total costs were 32 % of the Explanatory and 78 % of the Pragmatic design. Conclusion: Total costs for the STRESS RCT with a registry-based design were less than those for a pragmatic design and much less than an explanatory design. Cost savings reflect design elements and leveraging of registry resources to improve cost efficiency, but delays to trial completion should be considered.

2.
N Engl J Med ; 387(23): 2138-2149, 2022 12 08.
Article in English | MEDLINE | ID: mdl-36342116

ABSTRACT

BACKGROUND: Although perioperative prophylactic glucocorticoids have been used for decades, whether they improve outcomes in infants after heart surgery with cardiopulmonary bypass is unknown. METHODS: We conducted a multicenter, prospective, randomized, placebo-controlled, registry-based trial involving infants (<1 year of age) undergoing heart surgery with cardiopulmonary bypass at 24 sites participating in the Society of Thoracic Surgeons Congenital Heart Surgery Database. Registry data were used in the evaluation of outcomes. The infants were randomly assigned to receive prophylactic methylprednisolone (30 mg per kilogram of body weight) or placebo, which was administered into the cardiopulmonary-bypass pump-priming fluid. The primary end point was a ranked composite of death, heart transplantation, or any of 13 major complications. Patients without any of these events were assigned a ranked outcome based on postoperative length of stay. In the primary analysis, the ranked outcomes were compared between the trial groups with the use of odds ratios adjusted for prespecified risk factors. Secondary analyses included an unadjusted odds ratio, a win ratio, and safety outcomes. RESULTS: A total of 1263 infants underwent randomization, of whom 1200 received either methylprednisolone (599 infants) or placebo (601 infants). The likelihood of a worse outcome did not differ significantly between the methylprednisolone group and the placebo group (adjusted odds ratio, 0.86; 95% confidence interval [CI], 0.71 to 1.05; P = 0.14). Secondary analyses (unadjusted for risk factors) showed an odds ratio for a worse outcome of 0.82 (95% CI, 0.67 to 1.00) and a win ratio of 1.15 (95% CI, 1.00 to 1.32) in the methylprednisolone group as compared with the placebo group, findings suggestive of a benefit with methylprednisolone; however, patients in the methylprednisolone group were more likely than those in the placebo group to receive postoperative insulin for hyperglycemia (19.0% vs. 6.7%, P<0.001). CONCLUSIONS: Among infants undergoing surgery with cardiopulmonary bypass, prophylactic use of methylprednisolone did not significantly reduce the likelihood of a worse outcome in an adjusted analysis and was associated with postoperative development of hyperglycemia warranting insulin in a higher percentage of infants than placebo. (Funded by the National Center for Advancing Translational Sciences and others; STRESS ClinicalTrials.gov number, NCT03229538.).


Subject(s)
Cardiac Surgical Procedures , Methylprednisolone , Humans , Methylprednisolone/adverse effects , Prospective Studies , Insulin
3.
Front Cardiovasc Med ; 9: 857581, 2022.
Article in English | MEDLINE | ID: mdl-35600483

ABSTRACT

Throughout the continuum of heart formation, myocardial growth and differentiation occurs in concert with the development of a specialized population of endothelial cells lining the cardiac lumen, the endocardium. Once the endocardial cells are specified, they are in close juxtaposition to the cardiomyocytes, which facilitates communication between the two cell types that has been proven to be critical for both early cardiac development and later myocardial function. Endocardial cues orchestrate cardiomyocyte proliferation, survival, and organization. Additionally, the endocardium enables oxygenated blood to reach the cardiomyocytes. Cardiomyocytes, in turn, secrete factors that promote endocardial growth and function. As misregulation of this delicate and complex endocardial-myocardial interplay can result in congenital heart defects, further delineation of underlying genetic and molecular factors involved in cardiac paracrine signaling will be vital in the development of therapies to promote cardiac homeostasis and regeneration. Herein, we highlight the latest research that has advanced the elucidation of endocardial-myocardial interactions in early cardiac morphogenesis, including endocardial and myocardial crosstalk necessary for cellular differentiation and tissue remodeling during trabeculation, as well as signaling critical for endocardial growth during trabeculation.

4.
Article in English | MEDLINE | ID: mdl-34250501

ABSTRACT

Background: Pregestational diabetes complicates one million pregnancies in the United States and is associated with placental dysfunction. Placental dysfunction can manifest as stillbirth, spontaneous abortions, fetal growth restriction, and preeclampsia in the mother. However, the underlying mechanisms of placental dysfunction are not well understood. Objective: We hypothesize that maternal hyperglycemia disrupts cellular processes important for normal vascular development and function. Study Design: Hyperglycemia, defined as a non-fasting glucose concentration of >250 mg/dL was induced in eight-week-old female CD1 mice by injecting a one-time intraperitoneal dose of 150mg/kg streptozotocin. Control mice received an equal volume of normal saline. Hyperglycemic and control females were mated with CD-1 males. At Embryonic Day 17.5, the pregnant mice were euthanized. Sixty-eight placentas were harvested from the six euglycemic dams and twenty-six placentas were harvested from three hyperglycemic dams. RNA was extracted from homogenized placental tissue (N=12/group; 2-4 placentas per litter of each group). Total RNA was prepared and sequenced. Differentially expressed genes that were >2-fold change was considered significant. Placentas (9-20/group) were fixed in paraffin wax and sectioned at 6 µm. Cross-sectional areas of placental zones were evaluated using slides stained for hematoxylin and eosin, glycogen, collagen, proliferation and apoptosis. Quantification of staining intensity and percent positive nuclei was done using Leica Image Hub Data software. Data were compared between the control and experimental group using t-tests. Values of p < 0.05 were considered to be statistically significant. Results: The average maternal blood glucose concentrations for control and diabetic dams were 112+/-24 and 473+/-47 respectively (p<0.0001). A higher rate of resorptions was noted in the hyperglycemia exposed placentas compared to euglycemic exposed placentas (24% vs 7%; p=0.04). A total of 24 RNA libraries (12/group) were prepared. Placentas from hyperglycemic pregnancies exhibited 1374 differentially expressed genes (DEGs). The 10 most significantly differentially expressed genes are Filip 1, Prom 2, Fam 78a, Pde4d, Pou3f1, Kcnk5, Dusp4, Cxcr4, Slc6a4 and D430019H16Rik. Their corresponding biologic functions are related to chemotaxis, ossification, cellular and vascular development. Histologically, we found that hyperglycemia exposed placentas demonstrated increased proliferation, apoptosis, and glycogen content and decreased collagen deposition. Conclusion: There was a higher rate of resorptions in the pregnancies of hyperglycemic dams. Pregestational diabetes resulted in significant changes in placental morphology, including increased glycogen content in the spongiotrophoblast, decreased collagen deposition, increased apoptosis and proliferation in the junction zone. Maternal diabetes causes widespread disruption in multiple cellular processes important for normal vascular development and sets the platform for placenta dysfunction.

5.
Front Cell Dev Biol ; 9: 676543, 2021.
Article in English | MEDLINE | ID: mdl-34239874

ABSTRACT

Dopa decarboxylase (DDC) synthesizes serotonin in the developing mouse heart where it is encoded by Ddc_exon1a, a tissue-specific paternally expressed imprinted gene. Ddc_exon1a shares an imprinting control region (ICR) with the imprinted, maternally expressed (outside of the central nervous system) Grb10 gene on mouse chromosome 11, but little else is known about the tissue-specific imprinted expression of Ddc_exon1a. Fluorescent immunostaining localizes DDC to the developing myocardium in the pre-natal mouse heart, in a region susceptible to abnormal development and implicated in congenital heart defects in human. Ddc_exon1a and Grb10 are not co-expressed in heart nor in brain where Grb10 is also paternally expressed, despite sharing an ICR, indicating they are mechanistically linked by their shared ICR but not by Grb10 gene expression. Evidence from a Ddc_exon1a gene knockout mouse model suggests that it mediates the growth of the developing myocardium and a thinning of the myocardium is observed in a small number of mutant mice examined, with changes in gene expression detected by microarray analysis. Comparative studies in the human developing heart reveal a paternal expression bias with polymorphic imprinting patterns between individual human hearts at DDC_EXON1a, a finding consistent with other imprinted genes in human.

6.
Am Heart J ; 226: 188-197, 2020 08.
Article in English | MEDLINE | ID: mdl-32599259

ABSTRACT

BACKGROUND: Randomized controlled trials (RCTs) in children with heart disease are challenging and therefore infrequently performed. We sought to improve feasibility of perioperative RCTs for this patient cohort using data from a large, multicenter clinical registry. We evaluated potential enrollment and end point frequencies for various inclusion cohorts and developed a novel global rank trial end point. We then performed trial simulations to evaluate power gains with the global rank end point and with use of planned covariate adjustment as an analytic strategy. METHODS: Data from the Society of Thoracic Surgery-Congenital Heart Surgery Database (STS-CHSD, 2011-2016) were used to support development of a consensus-based global rank end point and for trial simulations. For Monte Carlo trial simulations (n = 50,000/outcome), we varied the odds of outcomes for treatment versus placebo and evaluated power based on the proportion of trial data sets with a significant outcome (P < .05). RESULTS: The STS-CHSD study cohort included 35,967 infant index cardiopulmonary bypass operations from 103 STS-CHSD centers, including 11,411 (32%) neonatal cases and 12,243 (34%) high-complexity (Society of Thoracic Surgeons-European Association for Cardio-Thoracic Surgery mortality category ≥4) cases. In trial simulations, study power was 21% for a mortality-only end point, 47% for a morbidity and mortality composite, and 78% for the global rank end point. With covariate adjustment, power increased to 94%. Planned covariate adjustment was preferable to restricting to higher-risk cohorts despite higher event rates in these cohorts. CONCLUSIONS: Trial simulations can inform trial design. Our findings, including the newly developed global rank end point, may be informative for future perioperative trials in children with heart disease.


Subject(s)
Cardiopulmonary Bypass , Heart Diseases/surgery , Randomized Controlled Trials as Topic/statistics & numerical data , Randomized Controlled Trials as Topic/standards , Research Design/statistics & numerical data , Child , Heart Diseases/congenital , Humans
7.
Am Heart J ; 220: 192-202, 2020 02.
Article in English | MEDLINE | ID: mdl-31855716

ABSTRACT

For decades, physicians have administered corticosteroids in the perioperative period to infants undergoing heart surgery with cardiopulmonary bypass (CPB) to reduce the postoperative systemic inflammatory response to CPB. Some question this practice because steroid efficacy has not been conclusively demonstrated and because some studies indicate that steroids could have harmful effects. STRESS is a randomized, placebo-controlled, double-blind, multicenter trial designed to evaluate safety and efficacy of perioperative steroids in infants (age < 1 year) undergoing heart surgery with CPB. Participants (planned enrollment = 1,200) are randomized 1:1 to methylprednisolone (30 mg/kg) administered into the CPB pump prime versus placebo. The trial is nested within the existing infrastructure of the Society of Thoracic Surgeons Congenital Heart Surgery Database. The primary outcome is a global rank score of mortality, major morbidities, and hospital length of stay with components ranked commensurate with their clinical severity. Secondary outcomes include several measures of major postoperative morbidity, postoperative hospital length of stay, and steroid-related safety outcomes including prevalence of hyperglycemia and postoperative infectious complications. STRESS will be one of the largest trials ever conducted in children with heart disease and will answer a decades-old question related to safety and efficacy of perioperative steroids in infants undergoing heart surgery with CPB. The pragmatic "trial within a registry" design may provide a mechanism for conducting low-cost, high-efficiency trials in a heretofore-understudied patient population.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Cardiopulmonary Bypass/adverse effects , Heart Defects, Congenital/surgery , Methylprednisolone/therapeutic use , Postoperative Complications/prevention & control , Systemic Inflammatory Response Syndrome/prevention & control , Anti-Inflammatory Agents/administration & dosage , Anti-Inflammatory Agents/adverse effects , Double-Blind Method , Humans , Hyperglycemia/epidemiology , Infant , Infant, Newborn , Infections/epidemiology , Length of Stay , Methylprednisolone/administration & dosage , Methylprednisolone/adverse effects , Outcome Assessment, Health Care , Placebos/therapeutic use , Postoperative Complications/epidemiology , Postoperative Complications/mortality , Registries , Research Design , Systemic Inflammatory Response Syndrome/mortality , United States
8.
Dev Biol ; 455(1): 73-84, 2019 11 01.
Article in English | MEDLINE | ID: mdl-31319059

ABSTRACT

The mechanisms regulating endothelial cell response to hemodynamic forces required for heart valve development, especially valve remodeling, remain elusive. Tie1, an endothelial specific receptor tyrosine kinase, is up-regulated by oscillating shear stress and is required for lymphatic valve development. In this study, we demonstrate that valvular endothelial Tie1 is differentially expressed in a dynamic pattern predicted by disturbed flow during valve remodeling. Following valvular endocardial specific deletion of Tie1 in mice, we observed enlarged aortic valve leaflets, decreased valve stiffness and valvular insufficiency. Valve abnormalities were only detected in late gestation and early postnatal mutant animals and worsened with age. The mutant mice developed perturbed extracellular matrix (ECM) deposition and remodeling characterized by increased glycosaminoglycan and decreased collagen content, as well as increased valve interstitial cell expression of Sox9, a transcription factor essential for normal ECM maturation during heart valve development. This study provides the first evidence that Tie1 is involved in modulation of late valve remodeling and suggests that an important Tie1-Sox9 signaling axis exists through which disturbed flows are converted by endocardial cells to paracrine Sox9 signals to modulate normal matrix remodeling of the aortic valve.


Subject(s)
Aortic Valve/metabolism , Gene Expression Profiling/methods , Gene Expression Regulation, Developmental , Organogenesis/genetics , Receptor, TIE-1/genetics , Animals , Aortic Valve/embryology , Aortic Valve/growth & development , Endothelial Cells/metabolism , Extracellular Matrix/metabolism , Female , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Pregnancy , Receptor, TIE-1/metabolism , SOX9 Transcription Factor/genetics , SOX9 Transcription Factor/metabolism , Vascular Remodeling/genetics
9.
JCI Insight ; 52019 05 21.
Article in English | MEDLINE | ID: mdl-31112136

ABSTRACT

The ang1-Tie2 pathway is required for normal vascular development, but its molecular effectors are not well-defined during cardiac ontogeny. Here we show that endocardial specific attenuation of Tie2 results in mid-gestation lethality due to heart defects associated with a hyperplastic but simplified trabecular meshwork (fewer but thicker trabeculae). Reduced proliferation and production of endocardial cells (ECs) following endocardial loss of Tie2 results in decreased endocardial sprouting required for trabecular assembly and extension. The hyperplastic trabeculae result from enhanced proliferation of trabecular cardiomyocyte (CMs), which is associated with upregulation of Bmp10, increased retinoic acid (RA) signaling, and Erk1/2 hyperphosphorylation in the myocardium. Intriguingly, myocardial phenotypes in Tie2-cko hearts could be partially rescued by inhibiting in utero RA signaling with pan-retinoic acid receptor antagonist BMS493. These findings reveal two complimentary functions of endocardial Tie2 during ventricular chamber formation: ensuring normal trabeculation by supporting EC proliferation and sprouting, and preventing hypertrabeculation via suppression of RA signaling in trabecular CMs.


Subject(s)
Embryonic Development/physiology , Heart Defects, Congenital/metabolism , Heart/embryology , Heart/growth & development , Receptor, TIE-2/metabolism , Animals , Cell Proliferation , Disease Models, Animal , Embryonic Development/genetics , Endocardium/embryology , Endocardium/growth & development , Endocardium/metabolism , Endocardium/pathology , Female , Gene Expression Regulation, Developmental , Heart Defects, Congenital/genetics , Heart Defects, Congenital/pathology , Male , Mice , Receptor, TIE-2/genetics , Signal Transduction
10.
Development ; 146(9)2019 05 08.
Article in English | MEDLINE | ID: mdl-31023876

ABSTRACT

The endocardium interacts with the myocardium to promote proliferation and morphogenesis during the later stages of heart development. However, the role of the endocardium in early cardiac ontogeny remains under-explored. Given the shared origin, subsequent juxtaposition, and essential cell-cell interactions of endocardial and myocardial cells throughout heart development, we hypothesized that paracrine signaling from the endocardium to the myocardium is crucial for initiating early differentiation of myocardial cells. To test this, we generated an in vitro, endocardial-specific ablation model using the diphtheria toxin receptor under the regulatory elements of the Nfatc1 genomic locus (NFATc1-DTR). Early treatment of NFATc1-DTR mouse embryoid bodies with diphtheria toxin efficiently ablated endocardial cells, which significantly attenuated the percentage of beating EBs in culture and expression of early and late myocardial differentiation markers. The addition of Bmp2 during endocardial ablation partially rescued myocyte differentiation, maturation and function. Therefore, we conclude that early stages of myocardial differentiation rely on endocardial paracrine signaling mediated in part by Bmp2. Our findings provide novel insight into early endocardial-myocardial interactions that can be explored to promote early myocardial development and growth.


Subject(s)
Cell Differentiation/physiology , Endocardium/cytology , Endocardium/metabolism , Myocardium/cytology , Myocardium/metabolism , Animals , Cell Differentiation/genetics , Female , Flow Cytometry , Male , Mice , Mice, Inbred C57BL , Microscopy, Fluorescence , NFATC Transcription Factors/genetics , NFATC Transcription Factors/metabolism , Organogenesis/genetics , Organogenesis/physiology , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction
11.
Article in English | MEDLINE | ID: mdl-32719828

ABSTRACT

Maternal mortality is on the rise in the United States and it disproportionately affects black women. The reasons for this staggering discrepancy hinge on three central issues: First, black women are more likely to have pre-existing cardiovascular morbidity that increase the risk of maternal mortality. Second, black women are more likely to experience adverse pregnancy outcomes which puts them at risk for developing long-term cardiovascular disease. Third, racial bias of providers and perceived racial discrimination from patients (the elephant in the room) impacts black patients' trust in their providers and the medical community at large. Reducing black maternal mortality involves a multi-tiered approach involving the patient, provider and public health policy.

12.
Br J Dermatol ; 180(5): 1050-1057, 2019 05.
Article in English | MEDLINE | ID: mdl-30500065

ABSTRACT

BACKGROUND: Once-daily topical oxymetazoline cream 1·0% significantly reduced persistent facial erythema of rosacea in trials requiring live, static patient assessments. OBJECTIVES: To evaluate critically the methodology of clinical trials that require live, static patient assessments by determining whether assessment of erythema is different when reference to the baseline photograph is allowed. METHODS: In two identically designed, randomized, phase III trials, adults with persistent facial erythema of rosacea applied oxymetazoline or vehicle once daily. This phase IV study evaluated standardized digital facial photographs from the phase III trials to record ≥ 1-grade Clinician Erythema Assessment (CEA) improvement at 1, 3, 6, 9 and 12 h postdose. RESULTS: Among 835 patients (oxymetazoline n = 415, vehicle n = 420), significantly greater proportions of patients treated with oxymetazoline vs. vehicle achieved ≥ 1-grade CEA improvement. For the comparison between phase IV study results and the original phase III analysis, when reference to baseline photographs was allowed while evaluating post-treatment photographs, the results for oxymetazoline were similar to results of the phase III trials (up to 85.7%), but a significantly lower proportion of vehicle recipients achieved ≥ 1-grade CEA improvement (up to 29.7% [phase 4] vs. 52.3% [phase 3]; P<0.001). In the phase IV study, up to 80·2% of patients treated with oxymetazoline achieved at least moderate erythema improvement vs. up to 22·9% of patients treated with vehicle. The association between patients' satisfaction with facial skin redness and percentage of erythema improvement was statistically significant. CONCLUSIONS: Assessment of study photographs, with comparison to baseline, confirmed significant erythema reduction with oxymetazoline on the first day of application. Compared with the phase III trial results, significantly fewer vehicle recipients attained ≥ 1-grade CEA improvement, suggesting a mitigated vehicle effect. This methodology may improve the accuracy of clinical trials evaluating erythema severity.


Subject(s)
Erythema/diagnosis , Oxymetazoline/administration & dosage , Photography/standards , Rosacea/diagnosis , Severity of Illness Index , Erythema/drug therapy , Face , Female , Humans , Male , Middle Aged , Patient Reported Outcome Measures , Patient Satisfaction , Research Design/standards , Rosacea/drug therapy , Skin/diagnostic imaging , Skin/drug effects , Skin Cream/administration & dosage , Treatment Outcome
14.
J Clin Invest ; 128(2): 834-845, 2018 02 01.
Article in English | MEDLINE | ID: mdl-29355844

ABSTRACT

The endothelial tyrosine kinase receptor Tie1 remains poorly characterized, largely owing to its orphan receptor status. Global Tie1 inactivation causes late embryonic lethality, thereby reflecting its importance during development. Tie1 also plays pivotal roles during pathologies such as atherosclerosis and tumorigenesis. In order to study the contribution of Tie1 to tumor progression and metastasis, we conditionally deleted Tie1 in endothelial cells at different stages of tumor growth and metastatic dissemination. Tie1 deletion during primary tumor growth in mice led to a decrease in microvessel density and an increase in mural cell coverage with improved vessel perfusion. Reduced angiogenesis and enhanced vascular normalization resulted in a progressive increase of intratumoral necrosis that caused a growth delay only at later stages of tumor progression. Concomitantly, surgical removal of the primary tumor decreased the number of circulating tumor cells, reduced metastasis, and prolonged overall survival. Additionally, Tie1 deletion in experimental murine metastasis models prevented extravasation of tumor cells into the lungs and reduced metastatic foci. Taken together, the data support Tie1 as a therapeutic target by defining its regulatory functions during angiogenesis and vascular abnormalization and identifying its role during metastasis.


Subject(s)
Endothelial Cells/metabolism , Lung Neoplasms/pathology , Neoplasm Metastasis , Neovascularization, Pathologic , Receptor, TIE-1/metabolism , Animals , Cell Line, Tumor , Disease Progression , Female , Gene Deletion , Human Umbilical Vein Endothelial Cells , Humans , Lung Neoplasms/metabolism , Male , Melanoma, Experimental , Mice , Mice, Knockout , Necrosis , Neoplasm Transplantation
15.
Ann Biomed Eng ; 45(11): 2548-2562, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28812215

ABSTRACT

Aortic valve (AV) disease involves stiffening of the AV cusp with progression characterized by inflammation, fibrosis, and calcification. Here, we examine the relationship between biomechanical valve function and proteomic changes before and after the development of AV pathology in the Emilin1-/- mouse model of latent AV disease. Biomechanical studies were performed to quantify tissue stiffness at the macro (micropipette) and micro (atomic force microscopy (AFM)) levels. Micropipette studies showed that the Emilin1-/- AV annulus and cusp regions demonstrated increased stiffness only after the onset of AV disease. AFM studies showed that the Emilin1-/- cusp stiffens before the onset of AV disease and worsens with the onset of disease. Proteomes from AV cusps were investigated to identify protein functions, pathways, and interaction network alterations that occur with age- and genotype-related valve stiffening. Protein alterations due to Emilin1 deficiency, including changes in pathways and functions, preceded biomechanical aberrations, resulting in marked depletion of extracellular matrix (ECM) proteins interacting with TGFB1, including latent transforming growth factor beta 3 (LTBP3), fibulin 5 (FBLN5), and cartilage intermediate layer protein 1 (CILP1). This study identifies proteomic dysregulation is associated with biomechanical dysfunction as early pathogenic processes in the Emilin1-/- model of AV disease.


Subject(s)
Heart Defects, Congenital/metabolism , Heart Valve Diseases/metabolism , Membrane Glycoproteins/genetics , Animals , Aortic Valve/metabolism , Aortic Valve/physiology , Bicuspid Aortic Valve Disease , Biomechanical Phenomena , Disease Models, Animal , Extracellular Matrix Proteins/metabolism , Female , Male , Mice, Knockout , Proteomics , Transforming Growth Factor beta1/metabolism
16.
Biochim Biophys Acta Proteins Proteom ; 1865(7): 927-935, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28341601

ABSTRACT

Significant progress has been made for tissue imaging of proteins using matrix-assisted laser desorption ionization imaging mass spectrometry (MALDI IMS). These advancements now facilitate mapping of a wide range of proteins, peptides, and post-translational modifications in a wide variety of tissues; however, the use of MALDI IMS to detect proteins from cardiac tissue is limited. This review discusses the most recent advances in protein imaging and demonstrates application to cardiac tissue, including the heart valve. Protein imaging by MALDI IMS allows multiplexed histological mapping of proteins and protein components that are inaccessible by antibodies and should be considered an important tool for basic and clinical cardiovascular research. This article is part of a Special Issue entitled: MALDI Imaging, edited by Dr. Corinna Henkel and Prof. Peter Hoffmann.


Subject(s)
Heart Valves/metabolism , Proteins/metabolism , Humans , Protein Processing, Post-Translational/physiology , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization/methods
17.
JCI Insight ; 2(4): e90656, 2017 02 23.
Article in English | MEDLINE | ID: mdl-28239655

ABSTRACT

It remains unclear how perturbations in cardiomyocyte sarcomere function alter postnatal heart development. We utilized murine models that allowed manipulation of cardiac myosin-binding protein C (MYBPC3) expression at critical stages of cardiac ontogeny to study the response of the postnatal heart to disrupted sarcomere function. We discovered that the hyperplastic to hypertrophic transition phase of mammalian heart development was altered in mice lacking MYBPC3 and this was the critical period for subsequent development of cardiomyopathy. Specifically, MYBPC3-null hearts developed evidence of increased cardiomyocyte endoreplication, which was accompanied by enhanced expression of cell cycle stimulatory cyclins and increased phosphorylation of retinoblastoma protein. Interestingly, this response was self-limited at later developmental time points by an upregulation of the cyclin-dependent kinase inhibitor p21. These results provide valuable insights into how alterations in sarcomere protein function modify postnatal heart development and highlight the potential for targeting cell cycle regulatory pathways to counteract cardiomyopathic stimuli.


Subject(s)
Carrier Proteins/genetics , Cell Enlargement , Cell Proliferation/genetics , Gene Expression Regulation, Developmental/genetics , Heart/growth & development , Myocytes, Cardiac/metabolism , Sarcomeres/metabolism , Animals , Cardiomyopathies/genetics , Cardiomyopathies/metabolism , Cell Cycle , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Cyclins/metabolism , Hyperplasia , Hypertrophy , Mice , Myocytes, Cardiac/physiology , Phosphorylation , Retinoblastoma Protein/metabolism , Sarcomeres/physiology , Up-Regulation
18.
J Heart Valve Dis ; 25(2): 240-252, 2016 03.
Article in English | MEDLINE | ID: mdl-27989075

ABSTRACT

BACKGROUND: Valvular disease is characterized in part by lipid deposition, but systematic analysis of the patterns of global lipid expression in healthy and diseased valve tissues are unknown. This is due in part to tissue limitations for lipidomic preparations and technologies for evaluating lipid distribution in tissues. The study aim was to examine the application of matrixassisted laser desorption ionization imaging mass spectrometry (MALDI IMS) to the aortic valve during development and disease, as an approach to detect and map lipids and ultimately better understand valve structure and function. METHODS: Established MALDI IMS strategies were applied to thin tissue sections of heart valves to map lipids to corresponding morphological features. Healthy prenatal and adult ovine aortic valve tissues were evaluated using the developed techniques. Lipid expression levels were compared between prenatal and adult valves using Wilcoxon rank sum testing and area under the receiver operating curves. A classification algorithm was used to determine distinct lipid signatures in adult extracellular matrix (ECM) substructures, including fibrosa and spongiosa layers. Lipid patterns were examined in heart valve tissue from pediatric patients with congenital aortic valve stenosis (CAVS). RESULTS: Lipid levels were decreased in adult ovine aortic valves when compared with prenatal valves. Classification algorithms applied to lipid signatures reported distinct lipid signatures mapping to ECM substructures in the adult aortic valve, but could not distinguish amorphous structures at pre-natal day 5. In CAVS, the in-situ lipid aggregation of distinct lipid species showed unique patterning both concurrent and divergent with ECM disarray. Fatty acid content varied between normal and diseased human aortic valves. CONCLUSIONS: MALDI IMS provides a new and useful approach to evaluate lipid biology in heart valve tissue. These findings define a role for lipid regulation in aortic valve development and demonstrate patterns of lipid deregulation in congenital disease.


Subject(s)
Aortic Valve Stenosis/metabolism , Aortic Valve/chemistry , Lipids/analysis , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Age Factors , Algorithms , Animals , Animals, Newborn , Aortic Valve/pathology , Aortic Valve Stenosis/congenital , Aortic Valve Stenosis/pathology , Biomarkers/analysis , Humans , Sheep, Domestic
19.
PLoS One ; 11(8): e0159710, 2016.
Article in English | MEDLINE | ID: mdl-27505173

ABSTRACT

The epicardium plays an important role in coronary vessel formation and Tgfbr3-/- mice exhibit failed coronary vessel development associated with decreased epicardial cell invasion. Immortalized Tgfbr3-/- epicardial cells display the same defects. Tgfbr3+/+ and Tgfbr3-/- cells incubated for 72 hours with VEH or ligands known to promote invasion via TGFßR3 (TGFß1, TGFß2, BMP2), for 72 hours were harvested for RNA-seq analysis. We selected for genes >2-fold differentially expressed between Tgfbr3+/+ and Tgfbr3-/- cells when incubated with VEH (604), TGFß1 (515), TGFß2 (553), or BMP2 (632). Gene Ontology (GO) analysis of these genes identified dysregulated biological processes consistent with the defects observed in Tgfbr3-/- cells, including those associated with extracellular matrix interaction. GO and Gene Regulatory Network (GRN) analysis identified distinct expression profiles between TGFß1-TGFß2 and VEH-BMP2 incubated cells, consistent with the differential response of epicardial cells to these ligands in vitro. Despite the differences observed between Tgfbr3+/+ and Tgfbr3-/- cells after TGFß and BMP ligand addition, GRNs constructed from these gene lists identified NF-ĸB as a key nodal point for all ligands examined. Tgfbr3-/- cells exhibited decreased expression of genes known to be activated by NF-ĸB signaling. NF-ĸB activity was stimulated in Tgfbr3+/+ epicardial cells after TGFß2 or BMP2 incubation, while Tgfbr3-/- cells failed to activate NF-ĸB in response to these ligands. Tgfbr3+/+ epicardial cells incubated with an inhibitor of NF-ĸB signaling no longer invaded into a collagen gel in response to TGFß2 or BMP2. These data suggest that NF-ĸB signaling is dysregulated in Tgfbr3-/- epicardial cells and that NF-ĸB signaling is required for epicardial cell invasion in vitro. Our approach successfully identified a signaling pathway important in epicardial cell behavior downstream of TGFßR3. Overall, the genes and signaling pathways identified through our analysis yield the first comprehensive list of candidate genes whose expression is dependent on TGFßR3 signaling.


Subject(s)
Pericardium/cytology , Pericardium/embryology , Proteoglycans/metabolism , Receptors, Transforming Growth Factor beta/metabolism , Signal Transduction , Transcriptome , Animals , Cell Line , Ligands , Mice , Mice, Inbred C57BL , NF-kappa B/metabolism , Proteoglycans/deficiency , Receptors, Transforming Growth Factor beta/deficiency , Sequence Analysis, RNA
20.
Curr Opin Pediatr ; 28(5): 584-9, 2016 10.
Article in English | MEDLINE | ID: mdl-27428484

ABSTRACT

PURPOSE OF REVIEW: The study of cardiac development is critical to inform management strategies for congenital and acquired heart disease. This review serves to highlight some of the advances in this field over the past year. RECENT FINDINGS: Three main areas of study are included that have been particularly innovative and progressive. These include more precise gene targeting in animal models of disease and in moving from animal models to human disease, more precise in-vitro models including three-dimensional structuring and inclusion of hemodynamic components, and expanding the concepts of genetic regulation of heart development and disease. SUMMARY: Targeted genetics in animal models are able to make use of tissue and time-specific promotors that drive gene expression or knockout with high specificity. In-vitro models can recreate flow patterns in blood vessels and across cardiac valves. Noncoding RNAs, once thought to be of no consequence to gene transcription and translation, prove to be key regulators of genetic function in health and disease.


Subject(s)
Heart Defects, Congenital/embryology , Heart/embryology , Animals , DNA Copy Number Variations , Disease Models, Animal , Gene Expression Regulation, Developmental , Heart/physiology , Heart Defects, Congenital/genetics , Heart Defects, Congenital/physiopathology , Hemodynamics , Humans , In Vitro Techniques , Models, Anatomic , RNA, Untranslated
SELECTION OF CITATIONS
SEARCH DETAIL
...