Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 78
1.
PLoS One ; 15(12): e0242445, 2020.
Article En | MEDLINE | ID: mdl-33301490

Acyl-CoA dehydrogenase 10 (Acad10)-deficient mice develop impaired glucose tolerance, peripheral insulin resistance, and abnormal weight gain. In addition, they exhibit biochemical features of deficiencies of fatty acid oxidation, such as accumulation of metabolites consistent with abnormal mitochondrial energy metabolism and fasting induced rhabdomyolysis. ACAD10 has significant expression in mouse brain, unlike other acyl-CoA dehydrogenases (ACADs) involved in fatty acid oxidation. The presence of ACAD10 in human tissues was determined using immunohistochemical staining. To characterize the effect of ACAD10 deficiency on the brain, micro-MRI and neurobehavioral evaluations were performed. Acad10-deficient mouse behavior was examined using open field testing and DigiGait analysis for changes in general activity as well as indices of gait, respectively. ACAD10 protein was shown to colocalize to mitochondria and peroxisomes in lung, muscle, kidney, and pancreas human tissue. Acad10-deficient mice demonstrated subtle behavioral abnormalities, which included reduced activity and increased time in the arena perimeter in the open field test. Mutant animals exhibited brake and propulsion metrics similar to those of control animals, which indicates normal balance, stability of gait, and the absence of significant motor impairment. The lack of evidence for motor impairment combined with avoidance of the center of an open field arena and reduced vertical and horizontal exploration are consistent with a phenotype characterized by elevated anxiety. These results implicate ACAD10 function in normal mouse behavior, which suggests a novel role for ACAD10 in brain metabolism.


Acyl-CoA Dehydrogenase/genetics , Anxiety/genetics , Brain/enzymology , Energy Metabolism/genetics , Mitochondria/enzymology , Acyl-CoA Dehydrogenase/deficiency , Acyl-CoA Dehydrogenase/metabolism , Animals , Anxiety/enzymology , Anxiety/physiopathology , Behavior, Animal , Brain/diagnostic imaging , Carnitine/analogs & derivatives , Carnitine/metabolism , Gait/physiology , Humans , Kidney/enzymology , Liver/enzymology , Lung/enzymology , Magnetic Resonance Imaging , Maze Learning , Mice , Mice, Knockout , Muscle, Skeletal/enzymology , Pancreas/enzymology , Peroxisomes/enzymology
2.
J Cardiovasc Dev Dis ; 7(2)2020 Jun 12.
Article En | MEDLINE | ID: mdl-32545681

The goal of this review is to provide a broad overview of the biomechanical maturation and regulation of vertebrate cardiovascular (CV) morphogenesis and the evidence for mechanistic relationships between function and form relevant to the origins of congenital heart disease (CHD). The embryonic heart has been investigated for over a century, initially focusing on the chick embryo due to the opportunity to isolate and investigate myocardial electromechanical maturation, the ability to directly instrument and measure normal cardiac function, intervene to alter ventricular loading conditions, and then investigate changes in functional and structural maturation to deduce mechanism. The paradigm of "Develop and validate quantitative techniques, describe normal, perturb the system, describe abnormal, then deduce mechanisms" was taught to many young investigators by Dr. Edward B. Clark and then validated by a rapidly expanding number of teams dedicated to investigate CV morphogenesis, structure-function relationships, and pathogenic mechanisms of CHD. Pioneering studies using the chick embryo model rapidly expanded into a broad range of model systems, particularly the mouse and zebrafish, to investigate the interdependent genetic and biomechanical regulation of CV morphogenesis. Several central morphogenic themes have emerged. First, CV morphogenesis is inherently dependent upon the biomechanical forces that influence cell and tissue growth and remodeling. Second, embryonic CV systems dynamically adapt to changes in biomechanical loading conditions similar to mature systems. Third, biomechanical loading conditions dynamically impact and are regulated by genetic morphogenic systems. Fourth, advanced imaging techniques coupled with computational modeling provide novel insights to validate regulatory mechanisms. Finally, insights regarding the genetic and biomechanical regulation of CV morphogenesis and adaptation are relevant to current regenerative strategies for patients with CHD.

3.
Cell Rep ; 31(9): 107711, 2020 06 02.
Article En | MEDLINE | ID: mdl-32492423

The availability of an autologous transplantable auxiliary liver would dramatically affect the treatment of liver disease. Assembly and function in vivo of a bioengineered human liver derived from induced pluripotent stem cells (iPSCs) has not been previously described. By improving methods for liver decellularization, recellularization, and differentiation of different liver cellular lineages of human iPSCs in an organ-like environment, we generated functional engineered human mini livers and performed transplantation in a rat model. Whereas previous studies recellularized liver scaffolds largely with rodent hepatocytes, we repopulated not only the parenchyma with human iPSC-hepatocytes but also the vascular system with human iPS-endothelial cells, and the bile duct network with human iPSC-biliary epithelial cells. The regenerated human iPSC-derived mini liver containing multiple cell types was tested in vivo and remained functional for 4 days after auxiliary liver transplantation in immunocompromised, engineered (IL2rg-/-) rats.


Hepatocytes/transplantation , Tissue Engineering , Activins/genetics , Activins/metabolism , Animals , Bone Morphogenetic Protein 4/genetics , Bone Morphogenetic Protein 4/metabolism , Cell Differentiation , Cells, Cultured , Cellular Reprogramming , Fetus/cytology , Fibroblast Growth Factor 2/genetics , Fibroblast Growth Factor 2/metabolism , Fibroblasts/cytology , Fibroblasts/metabolism , Hepatocytes/cytology , Hepatocytes/metabolism , Humans , Immunocompromised Host , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Male , Rats , Rats, Sprague-Dawley , Tissue Scaffolds/chemistry , Transcription Factors/genetics , Transcription Factors/metabolism
4.
Int J Neonatal Screen ; 5(4): 41, 2019 Dec.
Article En | MEDLINE | ID: mdl-33072999

Spinal muscular atrophy (SMA) is a neuromuscular disorder caused by SMN1 gene deletion/mutation. The drug nusinersen modifies SMN2 mRNA splicing, increasing the production of the full-length SMN protein. Recent studies have demonstrated the beneficial effects of nusinersen in patients with SMA, particularly when treated in early infancy. Because nusinersen treatment can alter disease trajectory, there is a strong rationale for newborn screening. In the current study, we validated the accuracy of a new system for detecting SMN1 deletion (Japanese patent application No. 2017-196967, PCT/JP2018/37732) using dried blood spots (DBS) from 50 patients with genetically confirmed SMA and 50 controls. Our system consists of two steps: (1) targeted pre-amplification of SMN genes by direct polymerase chain reaction (PCR) and (2) detection of SMN1 deletion by real-time modified competitive oligonucleotide priming-PCR (mCOP-PCR) using the pre-amplified products. Compared with PCR analysis results of freshly collected blood samples, our system exhibited a sensitivity of 1.00 (95% confidence interval [CI] 0.96-1.00) and a specificity of 1.00 (95% CI 0.96-1.00). We also conducted a prospective SMA screening study using DBS from 4157 Japanese newborns. All DBS tested negative, and there were no screening failures. Our results indicate that the new system can be reliably used in SMA newborn screening.

5.
Nihon Yakurigaku Zasshi ; 152(3): 147-159, 2018.
Article Ja | MEDLINE | ID: mdl-30185733

Nusinersen (Spinraza®) was approved as Japan's first antisense oligonucleotide (ASO) drug for treatment of SMA (spinal muscular atrophy) patients with a deletion or mutation of the survival motor neuron (SMN) 1 gene and ≥1 copy of the SMN2 gene. Nuseinersen is a fully modified 2'-O-(2-methoxyethyl) (2'-MOE) ASO designed to bind the SMN2 pre-mRNA and alter splicing, such that a mature mRNA is produced and is translated as full-length SMN protein. In 4 types of mouse SMA disease models, treatment with nusinersen improved the form of the neuromuscular junction, increased myofiber size, improved righting reflex and grip, and prolonged survival. The efficacy of nusinersen was verified in 2 multinational, randomized, double-blind, sham-controlled clinical studies in SMA patients with differing ages of onset and ages (ENDEAR study and CHERISH study), and improvement and maintenance of motor function by nusinersen were demonstrated regardless of the type of SMA. Moreover, both studies showed that greater efficacy may be obtained with early initiation of nusinersen treatment. Therefore, treatment with nusinersen should be started as early as possible to delay or halt progression of the disease and maximize therapeutic effect. As nusinersen is the only ASO currently available for SMA, it will be widely used, therefore we will expect that nusinersen will contribute to improve patients' QOL and reduce the burden of caregivers and the healthcare system by improving motor function of patients with SMA.


Muscular Atrophy, Spinal/drug therapy , Oligonucleotides/therapeutic use , Double-Blind Method , Humans
6.
Lipids Health Dis ; 16(1): 121, 2017 Jun 17.
Article En | MEDLINE | ID: mdl-28623954

BACKGROUND: Statins are generally well-tolerated and serious side effects are infrequent, but some patients experience adverse events and reduce their statin dose or discontinue treatment altogether. Alirocumab is a highly specific, fully human monoclonal antibody to proprotein convertase subtilisin/kexin type 9 (PCSK9), which can produce substantial and sustained reductions of low-density lipoprotein cholesterol (LDL-C). METHODS: The randomized, double-blind, placebo-controlled, parallel-group, phase 3 ODYSSEY NIPPON study will explore alirocumab 150 mg every 4 weeks (Q4W) in 163 Japanese patients with hypercholesterolemia who are on the lowest-strength dose of atorvastatin (5 mg/day) or are receiving a non-statin lipid-lowering therapy (LLT) (fenofibrate, bezafibrate, ezetimibe, or diet therapy alone). Hypercholesterolemia is defined as LDL-C ≥ 100 mg/dL (2.6 mmol/L) in patients with heterozygous familial hypercholesterolemia or non-familial hypercholesterolemia with a history of documented coronary heart disease, or ≥120 mg/dL (3.1 mmol/L) in patients with non-familial hypercholesterolemia classified as primary prevention category III (i.e. high-risk patients). During the 12-week double-blind treatment period, patients will be randomized (1:1:1) to receive alirocumab subcutaneously (SC) 150 mg Q4W alternating with placebo for alirocumab Q4W, or alirocumab 150 mg SC every 2 weeks (Q2W), or SC placebo Q2W. The primary efficacy endpoint is the percentage change in calculated LDL-C from baseline to week 12. The long-term safety and tolerability of alirocumab will also be investigated. DISCUSSION: The ODYSSEY NIPPON study will provide insights into the efficacy and safety of alirocumab 150 mg Q4W or 150 mg Q2W among Japanese patients with hypercholesterolemia who are on the lowest-strength dose of atorvastatin, or are receiving a non-statin LLT (including diet therapy alone). TRIAL REGISTRATION: ClinicalTrials.gov number: NCT02584504.


Antibodies, Monoclonal/therapeutic use , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Hypercholesterolemia/drug therapy , Adult , Antibodies, Monoclonal/adverse effects , Antibodies, Monoclonal, Humanized , Atorvastatin/adverse effects , Atorvastatin/therapeutic use , Cholesterol, LDL/blood , Double-Blind Method , Female , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/adverse effects , Hypercholesterolemia/blood , Hypercholesterolemia/metabolism , Male , PCSK9 Inhibitors , Young Adult
7.
Biomaterials ; 83: 182-93, 2016 Mar.
Article En | MEDLINE | ID: mdl-26774561

Intramyocardial injection of various injectable hydrogel materials has shown benefit in positively impacting the course of left ventricular (LV) remodeling after myocardial infarction (MI). However, since LV remodeling is a complex, time dependent process, the most efficacious time of hydrogel injection is not clear. In this study, we injected a relatively stiff, thermoresponsive and bioabsorbable hydrogel in rat hearts at 3 different time points - immediately after MI (IM), 3 d post-MI (3D), and 2 w post-MI (2W), corresponding to the beginnings of the necrotic, fibrotic and chronic remodeling phases. The employed left anterior descending coronary artery ligation model showed expected infarction responses including functional loss, inflammation and fibrosis with distinct time dependent patterns. Changes in LV geometry and contractile function were followed by longitudinal echocardiography for 10 w post-MI. While all injection times positively affected LV function and wall thickness, the 3D group gave better functional outcomes than the other injection times and also exhibited more local vascularization and less inflammatory markers than the earlier injection time. The results indicate an important role for injection timing in the increasingly explored concept of post-MI biomaterial injection therapy and suggest that for hydrogels with mechanical support as primary function, injection at the beginning of the fibrotic phase may provide improved outcomes.


Hydrogel, Polyethylene Glycol Dimethacrylate/administration & dosage , Hydrogel, Polyethylene Glycol Dimethacrylate/therapeutic use , Myocardial Infarction/drug therapy , Myocardial Infarction/physiopathology , Ventricular Remodeling , Actins/metabolism , Animals , Cytokines/metabolism , Female , Heart Ventricles/diagnostic imaging , Heart Ventricles/drug effects , Heart Ventricles/pathology , Heart Ventricles/physiopathology , Hydrogel, Polyethylene Glycol Dimethacrylate/pharmacology , Inflammation Mediators/metabolism , Injections , Macrophages/drug effects , Macrophages/pathology , Myocardial Infarction/diagnostic imaging , Myocardial Infarction/pathology , Myocardium/pathology , Neutrophil Infiltration/drug effects , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Rats, Inbred Lew , Time Factors , Ventricular Remodeling/drug effects
8.
Cardiovasc Res ; 107(4): 487-98, 2015 Sep 01.
Article En | MEDLINE | ID: mdl-26156497

AIMS: After injury, the adult zebrafish can regenerate the heart. This requires the activation of the endocardium and epicardium as well as the proliferation of pre-existing cardiomyocytes to replace the lost tissue. However, the molecular mechanisms involved in this process are not completely resolved. In this work, we aim to identify the proteins involved in zebrafish heart regeneration and to explore their function. METHODS AND RESULTS: Using a proteomic approach, we identified Hyaluronan-mediated motility receptor (Hmmr), a hyaluronic acid (HA) receptor, to be expressed following ventricular resection in zebrafish. Moreover, enzymes that produce HA, hyaluronic acid synthases (has), were also expressed following injury, suggesting that this pathway may serve important functions in the regenerating heart. Indeed, suppression of HA production, as well as depletion of Hmmr, blocked cardiac regeneration. Mechanistically, HA and Hmmr are required for epicardial cell epithelial-mesenchymal transition (EMT) and their subsequent migration into the regenerating ventricle. Furthermore, chemical inhibition of Focal Adhesion Kinase (FAK) or inhibition of Src kinases, downstream effectors of Hmmr, also prevented epicardial cell migration, implicating a HA/Hmmr/FAK/Src pathway in this process. In a rat model of myocardial infarction, both HA and HMMR were up-regulated and localized in the infarct area within the first few days following damage, suggesting that this pathway may also play an important role in cardiac repair in mammals. CONCLUSION: HA and Hmmr are required for activated epicardial cell EMT and migration involving the FAK/Src pathway for proper heart regeneration.


Epithelial-Mesenchymal Transition/physiology , Extracellular Matrix Proteins/metabolism , Heart/growth & development , Hyaluronan Receptors/metabolism , Myocytes, Cardiac/metabolism , Regeneration/physiology , Zebrafish/metabolism , Animals , Animals, Genetically Modified , Cell Adhesion/genetics , Cell Movement/physiology , Extracellular Space/metabolism , Myocytes, Cardiac/cytology , Proteomics/methods , Zebrafish/genetics , Zebrafish/growth & development
9.
Nature ; 521(7553): 520-4, 2015 May 28.
Article En | MEDLINE | ID: mdl-25807483

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.


Cilia/pathology , Heart Defects, Congenital/genetics , Heart Defects, Congenital/pathology , Animals , Cilia/diagnostic imaging , Cilia/genetics , Cilia/physiology , DNA Mutational Analysis , Electrocardiography , Exome/genetics , Genes, Recessive , Genetic Testing , Heart Defects, Congenital/diagnostic imaging , Humans , Male , Mice , Mice, Inbred C57BL , Mutation/genetics , Signal Transduction , Ultrasonography
11.
Nat Commun ; 6: 6023, 2015 Jan 20.
Article En | MEDLINE | ID: mdl-25599650

The ciliary kinase NEK8 plays a critical role in situs determination and cystic kidney disease, yet its exact function remains unknown. In this study, we identify ANKS6 as a target and activator of NEK8. ANKS6 requires NEK8 for localizing to the ciliary inversin compartment (IC) and activates NEK8 by binding to its kinase domain. Here we demonstrate the functional importance of this interaction through the analysis of two novel mouse mutations, Anks6(Streaker) and Nek8(Roc). Both display heterotaxy, cardiopulmonary malformations and cystic kidneys, a syndrome also characteristic of mutations in Invs and Nphp3, the other known components of the IC. The Anks6(Strkr) mutation decreases ANKS6 interaction with NEK8, precluding NEK8 activation. The Nek8(Roc) mutation inactivates NEK8 kinase function while preserving ANKS6 localization to the IC. Together, these data reveal the crucial role of NEK8 kinase activation within the IC, promoting proper left-right patterning, cardiopulmonary development and renal morphogenesis.


Body Patterning/physiology , Carrier Proteins/metabolism , Nuclear Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Animals , Blotting, Western , Body Patterning/genetics , Carrier Proteins/genetics , Cell Line , Female , Humans , Kinesins/genetics , Kinesins/metabolism , Male , Mice , Morphogenesis/genetics , Morphogenesis/physiology , Mutation , NIMA-Related Kinases , Nuclear Proteins/genetics , Protein Serine-Threonine Kinases/genetics , Rats , Transcription Factors/genetics , Transcription Factors/metabolism
12.
Tissue Eng Part A ; 21(1-2): 75-84, 2015 Jan.
Article En | MEDLINE | ID: mdl-24980864

Tracheal loss is a source of significant morbidity for affected patients with no acceptable solution. Interest in engineering tracheal transplants has created a demand for small animal models of orthotopic tracheal transplantation. Here, we examine the use of a decellularized graft in a murine model of tracheal replacement. Fresh or decellularized tracheas harvested from age-matched female donor C57BL/6 mice were transplanted into syngeneic recipients. Tracheas were decellularized using repeated washes of water, 3% Triton X-100, and 3 M NaCl under cyclic pressure changes, followed by disinfection with 0.1% peracetic acid/4% ethanol, and terminal sterilization by gamma irradiation. Tracheas were explanted for immunolabeling at 1, 4, and 8 weeks following surgery. Video microscopy and computed tomography were performed to assess function and structure. Decellularized grafts supported complete reepithelialization by 8 weeks and motile cilia were observed. Cartilaginous portions of the trachea were maintained in mice receiving fresh transplants, but repopulation of the cartilage was not seen in mice receiving decellularized transplants. We observed superior postsurgical survival, weight gain, and ciliary function in mice receiving fresh transplants compared with those receiving decellularized transplants. The murine orthotopic tracheal transplant provides an appropriate model to assess the repopulation and functional regeneration of decellularized tracheal grafts.


Cell Differentiation , Cell Movement , Epithelial Cells/cytology , Extracellular Matrix/transplantation , Trachea/cytology , Trachea/transplantation , Animals , Cilia/metabolism , Female , Mice, Inbred C57BL , Plastic Surgery Procedures , Tissue Scaffolds/chemistry , Trachea/diagnostic imaging , Trachea/surgery , Vacuum , X-Ray Microtomography
13.
Dev Cell ; 31(3): 279-290, 2014 Nov 10.
Article En | MEDLINE | ID: mdl-25446516

Vertebrate hedgehog signaling is coordinated by the differential localization of the receptors patched-1 and Smoothened in the primary cilium. Cilia assembly is mediated by intraflagellar transport (IFT), and cilia defects disrupt hedgehog signaling, causing many structural birth defects. We generated Ift25 and Ift27 knockout mice and show that they have structural birth defects indicative of hedgehog signaling dysfunction. Surprisingly, ciliary assembly is not affected, but abnormal hedgehog signaling is observed in conjunction with ciliary accumulation of patched-1 and Smoothened. Similarly, Smoothened accumulates in cilia on cells mutated for BBSome components or the BBS binding protein/regulator Lztfl1. Interestingly, the BBSome and Lztfl1 accumulate to high levels in Ift27 mutant cilia. Because Lztfl1 mutant cells accumulate BBSome but not IFT27, it is likely that Lztfl1 functions downstream of IFT27 to couple the BBSome to the IFT particle for coordinated removal of patched-1 and Smoothened from cilia during hedgehog signaling.


Cilia/metabolism , Signal Transduction , rab GTP-Binding Proteins/metabolism , Animals , Biological Transport , Flagella/metabolism , Hedgehog Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Receptors, Cell Surface/metabolism , Transcription Factors/metabolism
14.
Sci Rep ; 4: 6614, 2014 Oct 14.
Article En | MEDLINE | ID: mdl-25310989

Cellular cardiomyoplasty has emerged as a novel therapy to restore contractile function of injured failing myocardium. Human multipotent muscle derived stem cells (MDSC) can be a potential abundant, autologous cell source for cardiac repair. However, robust conditions for cardiomyocyte (CM) differentiation are not well established for this cell type. We have developed a new method for CM differentiation from human MDSC that combines 3-dimensional artificial muscle tissue (AMT) culture with temporally controlled biophysical cell aggregation and delivery of 4 soluble factors (microRNA-206 inhibitor, IWR-1, Lithium Chloride, and BMP-4) (4F-AG-AMT). The 4F-AG-AMT displayed cardiac-like response to ß-adrenergic stimulation and contractile properties. 4F-AG-AMT expressed major cardiac (NKX2-5, GATA4, TBX5, MEF2C) transcription factors and structural proteins. They also express cardiac gap-junction protein, connexin-43, similar to CMs and synchronized spontaneous calcium transients. These results highlight the importance of temporal control of biophysical and soluble factors for CM differentiation from MDSCs.


Cell Differentiation , Muscle, Skeletal/cytology , Myocytes, Cardiac/cytology , Stem Cells/cytology , Biophysical Phenomena , Cell Aggregation , Gene Expression Regulation, Developmental , Humans , Transcription Factors/biosynthesis
15.
Cardiovasc Res ; 104(2): 258-69, 2014 Nov 01.
Article En | MEDLINE | ID: mdl-25209314

AIMS: Familial hypertrophic cardiomyopathy (HCM) is one the most common heart disorders, with gene mutations in the cardiac sarcomere. Studying HCM with patient-specific induced pluripotent stem-cell (iPSC)-derived cardiomyocytes (CMs) would benefit the understanding of HCM mechanism, as well as the development of personalized therapeutic strategies. METHODS AND RESULTS: To investigate the molecular mechanism underlying the abnormal CM functions in HCM, we derived iPSCs from an HCM patient with a single missense mutation (Arginine442Glycine) in the MYH7 gene. CMs were next enriched from HCM and healthy iPSCs, followed with whole transcriptome sequencing and pathway enrichment analysis. A widespread increase of genes responsible for 'Cell Proliferation' was observed in HCM iPSC-CMs when compared with control iPSC-CMs. Additionally, HCM iPSC-CMs exhibited disorganized sarcomeres and electrophysiological irregularities. Furthermore, disease phenotypes of HCM iPSC-CMs were attenuated with pharmaceutical treatments. CONCLUSION: Overall, this study explored the possible patient-specific and mutation-specific disease mechanism of HCM, and demonstrates the potential of using HCM iPSC-CMs for future development of therapeutic strategies. Additionally, the whole methodology established in this study could be utilized to study mechanisms of other human-inherited heart diseases.


Cardiomyopathy, Hypertrophic, Familial/genetics , Induced Pluripotent Stem Cells/metabolism , Myocytes, Cardiac/metabolism , Action Potentials , Adult , Animals , Calcium Signaling/genetics , Cardiac Myosins/genetics , Cardiomyopathy, Hypertrophic, Familial/metabolism , Cardiomyopathy, Hypertrophic, Familial/pathology , Case-Control Studies , Cell Proliferation/genetics , Cell Separation/methods , Cells, Cultured , Cellular Reprogramming , Cellular Reprogramming Techniques , Female , Gene Expression Profiling/methods , Gene Expression Regulation , Gene Regulatory Networks , Genetic Predisposition to Disease , Humans , Induced Pluripotent Stem Cells/drug effects , Induced Pluripotent Stem Cells/ultrastructure , Mice, Inbred NOD , Mice, SCID , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/ultrastructure , Myosin Heavy Chains/genetics , Phenotype , Sarcomeres/metabolism , Sarcomeres/ultrastructure , Transcriptome
16.
Toxicol Sci ; 141(1): 112-9, 2014 Sep.
Article En | MEDLINE | ID: mdl-24924401

The transcription factor Nrf2 (Nfe2l2 nuclear factor, erythroid 2-like 2) regulates gene expression directly, controlling pharmacological and toxicological responses. These processes may also be influenced by the structure of the hepatic vasculature, which distributes blood flow through compartmentalized microenvironments to maintain organismal stability. Castings of the hepatic portal vasculature of albino C57BL/6J but not ICR Nrf2(-/-) mice revealed a congenital intrahepatic shunt that was present in two thirds of Nrf2-disrupted mice. This shunt directly connected the portal vein to the inferior vena cava and displayed characteristics of a patent ductus venosus. Immunohistochemistry revealed that Nrf2(-/-) mice with an intrahepatic shunt manifest changes to hepatic oxygen and protein expression gradients when compared with wild-type (WT) and non-shunted Nrf2(-/-) mice. Centrilobular hypoxia found in WT and Nrf2(-/-) mice without shunts was reduced in Nrf2(-/-) livers with a shunt. Hepatic protein expression of phosphoenolpyruvate carboxykinase (Pepck), normally confined to the periportal zone, exhibited both periportal and centrilobular zonal expression in livers from Nrf2(-/-) mice with an intrahepatic shunt. Centrilobular expression of Cytochrome P450 2E1 (Cyp2e1) was diminished in shunted Nrf2(-/-) livers compared with WT and Nrf2(-/-) livers without shunts. The intrahepatic shunt in Nrf2(-/-) mice was further found to diminish acetaminophen hepatoxicity compared with WT and Nrf2(-/-) non-shunted mice following a 6 h challenge with 250 mg/kg acetaminophen. The presence of an intrahepatic shunt influences several physiological and pathophysiological properties of Nrf2(-/-) mice through changes in blood flow, hepatic oxygenation, and protein expression that extent beyond loss of canonical transactivation of Nrf2 target genes.


Arteriovenous Malformations/metabolism , Chemical and Drug Induced Liver Injury/metabolism , Cytochrome P-450 CYP2E1/genetics , Disease Models, Animal , Liver Circulation , NF-E2-Related Factor 2/deficiency , Oxygen/metabolism , Acetaminophen/pharmacokinetics , Acetaminophen/toxicity , Animals , Arteriovenous Malformations/genetics , Arteriovenous Malformations/pathology , Chemical and Drug Induced Liver Injury/etiology , Chemical and Drug Induced Liver Injury/genetics , Liver/blood supply , Liver/enzymology , Liver/pathology , Liver Circulation/genetics , Mice, Inbred C57BL , Mice, Inbred ICR , Mice, Knockout , NF-E2-Related Factor 2/genetics
17.
PLoS One ; 9(3): e92263, 2014.
Article En | MEDLINE | ID: mdl-24642611

In experimental models of pancreatic growth and recovery, changes in pancreatic size are assessed by euthanizing a large cohort of animals at varying time points and measuring organ mass. However, to ascertain this information in clinical practice, patients with pancreatic disorders routinely undergo non-invasive cross-sectional imaging of the pancreas using magnetic resonance imaging (MRI) or computed tomography (CT). The aim of the current study was to develop a thin-sliced, optimized sequence protocol using a high field MRI to accurately calculate pancreatic volumes in the most common experimental animal, the mouse. Using a 7 Telsa Bruker micro-MRI system, we performed abdominal imaging in whole-fixed mice in three standard planes: axial, sagittal, and coronal. The contour of the pancreas was traced using Vitrea software and then transformed into a 3-dimensional (3D) reconstruction, from which volumetric measurements were calculated. Images were optimized using heart perfusion-fixation, T1 sequence analysis, and 0.2 to 0.4 mm thick slices. As proof of principle, increases in pancreatic volume among mice of different ages correlated tightly with increasing body weight. In summary, this is the first study to measure pancreatic volumes in mice, using a high field 7 Tesla micro-MRI and a thin-sliced, optimized sequence protocol. We anticipate that micro-MRI will improve the ability to non-invasively quantify changes in pancreatic size and will dramatically reduce the number of animals required to serially assess pancreatic growth and recovery.


Magnetic Resonance Imaging/methods , Pancreas/growth & development , Aging , Animals , Magnetic Resonance Imaging/instrumentation , Male , Mice , Organ Size , Pancreas/anatomy & histology , Phantoms, Imaging
18.
Circ Cardiovasc Imaging ; 7(1): 31-42, 2014 Jan.
Article En | MEDLINE | ID: mdl-24319090

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.


Echocardiography, Doppler , Fetal Heart/diagnostic imaging , Genetic Testing , Heart Defects, Congenital/diagnostic imaging , Heart Defects, Congenital/genetics , Microscopy, Acoustic , Mutation , Ultrasonography, Prenatal/methods , Animals , Disease Models, Animal , Echocardiography, Doppler, Color , Ethylnitrosourea/toxicity , Female , Fetal Heart/abnormalities , Genetic Predisposition to Disease , Heart Defects, Congenital/embryology , Heredity , High-Throughput Screening Assays , Male , Mice , Mice, Inbred C57BL , Pedigree , Phenotype
19.
Stem Cells Transl Med ; 3(2): 183-93, 2014 Feb.
Article En | MEDLINE | ID: mdl-24371329

Valuable and ample resources have been spent over the last two decades in pursuit of interventional strategies to treat the unmet demand of heart failure patients to restore myocardial structure and function. At present, it is clear that full restoration of myocardial structure and function is outside our reach from both clinical and basic research studies, but it may be achievable with a combination of ongoing research, creativity, and perseverance. Since the 1990s, skeletal myoblasts have been extensively investigated for cardiac cell therapy of congestive heart failure. Whereas the Myoblast Autologous Grafting in Ischemic Cardiomyopathy (MAGIC) trial revealed that transplanted skeletal myoblasts did not integrate into the host myocardium and also did not transdifferentiate into cardiomyocytes despite some beneficial effects on recipient myocardial function, recent studies suggest that skeletal muscle-derived stem cells have the ability to adopt a cardiomyocyte phenotype in vitro and in vivo. This brief review endeavors to summarize the importance of skeletal muscle stem cells and how they can play a key role to surpass current results in the future and enhance the efficacious implementation of regenerative cell therapy for heart failure.


Cardiomyopathies/therapy , Heart Failure/therapy , Muscle Fibers, Skeletal/cytology , Muscle, Skeletal/cytology , Myocardium/cytology , Stem Cells/cytology , Animals , Cardiomyopathies/pathology , Cell Lineage/physiology , Heart Failure/pathology , Humans
20.
Nat Commun ; 4: 2307, 2013.
Article En | MEDLINE | ID: mdl-23942048

Heart disease is the leading cause of death in the world. Heart tissue engineering holds a great promise for future heart disease therapy by building personalized heart tissues. Here we create heart constructs by repopulating decellularized mouse hearts with human induced pluripotent stem cell-derived multipotential cardiovascular progenitor cells. We show that the seeded multipotential cardiovascular progenitor cells migrate, proliferate and differentiate in situ into cardiomyocytes, smooth muscle cells and endothelial cells to reconstruct the decellularized hearts. After 20 days of perfusion, the engineered heart tissues exhibit spontaneous contractions, generate mechanical force and are responsive to drugs. In addition, we observe that heart extracellular matrix promoted cardiomyocyte proliferation, differentiation and myofilament formation from the repopulated human multipotential cardiovascular progenitor cells. Our novel strategy to engineer personalized heart constructs could benefit the study of early heart formation or may find application in preclinical testing.


Cell- and Tissue-Based Therapy/methods , Guided Tissue Regeneration/methods , Myocardium/cytology , Pluripotent Stem Cells/metabolism , Tissue Engineering/methods , Animals , Cell Differentiation , Cell Movement , Cell Proliferation , Cells, Cultured , Endothelial Cells/cytology , Heart , Heart Diseases/therapy , Humans , Mice , Mice, Inbred C57BL , Myocardium/metabolism , Myocytes, Cardiac/cytology , Pluripotent Stem Cells/cytology
...