Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
1.
Article in English | MEDLINE | ID: mdl-36901556

ABSTRACT

With urban expansion and traffic environment improvement, travel chains continue to grow, and the combination of travel purposes and modes becomes more complex. The promotion of mobility as a service (MaaS) has positive effects on facilitating the public transport traffic environment. However, public transport service optimization requires an accurate understanding of the travel environment, selection preferences, demand prediction, and systematic dispatch. Our study focused on the relationship between the trip-chain complexity environment and travel intention, combining the Theory of Planned Behavior (TPB) with travelers' preferences to construct a bounded rationality theory. First, this study used K-means clustering to transform the characteristics of the travel trip chain into the complexity of the trip chain. Then, based on the partial least squares structural equation model (PLS-SEM) and the generalized ordered Logit model, a mixed-selection model was established. Finally, the travel intention of PLS-SEM was compared with the travel sharing rate of the generalized ordered Logit model to determine the trip-chain complexity effects for different public transport modes. The results showed that (1) the proposed model, which transformed travel-chain characteristics into travel-chain complexity using K-means clustering and adopted a bounded rationality perspective, had the best fit and was the most effective with comparison to the previous prediction approaches. (2) Compared with service quality, trip-chain complexity negatively affected the intention of using public transport in a wider range of indirect paths. Gender, vehicle ownership, and with children/without children had significant moderating effects on certain paths of the SEM. (3) The research results obtained by PLS-SEM indicated that when travelers were more willing to travel by subway, the subway travel sharing rate corresponding to the generalized ordered Logit model was only 21.25-43.49%. Similarly, the sharing rate of travel by bus was only 32-44% as travelers were more willing to travel by bus obtained from PLS-SEM. Therefore, it is necessary to combine the qualitative results of PLS-SEM with the quantitative results of generalized ordered Logit. Moreover, when service quality, preferences, and subjective norms were based on the mean value, with each increase in trip-chain complexity, the subway travel sharing rate was reduced by 3.89-8.30%, while the bus travel sharing rate was reduced by 4.63-6.03%.


Subject(s)
Railroads , Travel , Child , Humans , Transportation , Intention , Cluster Analysis
2.
Article in English | MEDLINE | ID: mdl-36981759

ABSTRACT

The links between built environments (BE) and commute durations have been extensively studied. However, relatively few studies have considered the effects of BEs at different spatial levels within a unified framework, or identified the gendered relationships between BEs and commute durations. Using survey data from 3209 household couples in 97 Chinese cities, this study investigates the effects of neighborhood- and city-level BEs on commute durations and the potential differences in these effects between the male and female members of the same household couple. A multi-group generalized multilevel structural equation model is applied to reveal the gendered relationships between neighborhood- and city-level BEs and commute durations. The findings suggest that the BE variables at two levels have significant effects on the commute duration. The mediating roles that the traffic congestion, car ownership, and commuting modes play in linking these BEs and commute durations are confirmed. Both levels of the BE variables are more influential factors for males' commuting durations. These findings have policy implications for the design of gender-equal transportation systems.


Subject(s)
Built Environment , Transportation , Male , Humans , Female , Cities , Surveys and Questionnaires
3.
Article in English | MEDLINE | ID: mdl-36361027

ABSTRACT

The literature has offered much evidence regarding associations between the built environment (BE) and commuting behavior. However, most prior studies are conducted based on cross-sectional samples from developed countries, and little is known about the longitudinal link between BE and commuting behavior. Based on two rounds of survey data from China, this study examines relationships of BE with commuting mode choice from both cross-sectional and longitudinal perspectives. The effects of life-cycle events are considered within a unified framework. Results of the longitudinal examination of BE and commuting mode shift largely support the cross-sectional analysis. Specifically, promoting more balanced land use and improving residential density are important for car use reductions and active travel initiatives. Meanwhile, more balanced land use improves the probability of commuting by motorcycle and electric bike, but reduces the probability of commuting by public transit. This study also highlights the remarkable role played by life-cycle events in affecting commuting mode shifts.


Subject(s)
Transportation , Walking , Cross-Sectional Studies , Built Environment , Bicycling
4.
Sci Transl Med ; 12(566)2020 10 21.
Article in English | MEDLINE | ID: mdl-33087505

ABSTRACT

Direct reprogramming of fibroblasts to alternative cell fates by forced expression of transcription factors offers a platform to explore fundamental molecular events governing cell fate identity. The discovery and study of induced cardiomyocytes (iCMs) not only provides alternative therapeutic strategies for heart disease but also sheds lights on basic biology underlying CM fate determination. The iCM field has primarily focused on early transcriptome and epigenome repatterning, whereas little is known about how reprogramming iCMs remodel, erase, and exit the initial fibroblast lineage to acquire final cell identity. Here, we show that autophagy-related 5 (Atg5)-dependent autophagy, an evolutionarily conserved self-digestion process, was induced and required for iCM reprogramming. Unexpectedly, the autophagic factor Beclin1 (Becn1) was found to suppress iCM induction in an autophagy-independent manner. Depletion of Becn1 resulted in improved iCM induction from both murine and human fibroblasts. In a mouse genetic model, Becn1 haploinsufficiency further enhanced reprogramming factor-mediated heart function recovery and scar size reduction after myocardial infarction. Mechanistically, loss of Becn1 up-regulated Lef1 and down-regulated Wnt inhibitors, leading to activation of the canonical Wnt/ß-catenin signaling pathway. In addition, Becn1 physically interacts with other classical class III phosphatidylinositol 3-kinase (PI3K III) complex components, the knockdown of which phenocopied Becn1 depletion in cardiac reprogramming. Collectively, our study revealed an inductive role of Atg5-dependent autophagy as well as a previously unrecognized autophagy-independent inhibitory function of Becn1 in iCM reprogramming.


Subject(s)
Cellular Reprogramming , Phosphatidylinositol 3-Kinases , Animals , Autophagy , Beclin-1/metabolism , Down-Regulation , Fibroblasts/metabolism , Mice , Myocytes, Cardiac/metabolism
5.
Circulation ; 139(14): 1725-1740, 2019 04 02.
Article in English | MEDLINE | ID: mdl-30636447

ABSTRACT

BACKGROUND: Hypertrophic response to pathological stimuli is a complex biological process that involves transcriptional and epigenetic regulation of the cardiac transcriptome. Although previous studies have implicated transcriptional factors and signaling molecules in pathological hypertrophy, the role of RNA-binding protein in this process has received little attention. METHODS: Here we used transverse aortic constriction and in vitro cardiac hypertrophy models to characterize the role of an evolutionary conserved RNA-binding protein Lin28a in pathological cardiac hypertrophy. Next-generation sequencing, RNA immunoprecipitation, and gene expression analyses were applied to identify the downstream targets of Lin28a. Epistatic analysis, metabolic assays, and flux analysis were further used to characterize the effects of Lin28a and its downstream mediator in cardiomyocyte hypertrophic growth and metabolic remodeling. RESULTS: Cardiac-specific deletion of Lin28a attenuated pressure overload-induced hypertrophic growth, cardiac dysfunction, and alterations in cardiac transcriptome. Mechanistically, Lin28a directly bound to mitochondrial phosphoenolpyruvate carboxykinase 2 ( Pck2) mRNA and increased its transcript level. Increasing Pck2 was sufficient to promote hypertrophic growth similar to that caused by increasing Lin28a, whereas knocking down Pck2 attenuated norepinephrine-induced cardiac hypertrophy. Epistatic analysis demonstrated that Pck2 mediated, at least in part, the role of Lin28a in cardiac hypertrophic growth. Furthermore, metabolomic analyses highlighted the role for Lin28a and Pck2 in promoting cardiac biosynthesis required for cell growth. CONCLUSIONS: Our study demonstrates that Lin28a promotes pathological cardiac hypertrophy and glycolytic reprograming, at least in part, by binding to and stabilizing Pck2 mRNA.


Subject(s)
Cell Proliferation , Energy Metabolism , Hypertrophy, Left Ventricular/enzymology , Mitochondria, Heart/enzymology , Myocytes, Cardiac/enzymology , Phosphoenolpyruvate Carboxykinase (ATP)/metabolism , RNA-Binding Proteins/metabolism , Animals , Cells, Cultured , Disease Models, Animal , Glycolysis , Hypertrophy, Left Ventricular/genetics , Hypertrophy, Left Ventricular/pathology , Hypertrophy, Left Ventricular/physiopathology , Mice, Knockout , Mitochondria, Heart/pathology , Myocytes, Cardiac/pathology , Phosphoenolpyruvate Carboxykinase (ATP)/genetics , Protein Binding , RNA Stability , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA-Binding Proteins/genetics , Rats, Sprague-Dawley , Ventricular Function, Left , Ventricular Remodeling
6.
Article in English | MEDLINE | ID: mdl-30158467

ABSTRACT

Although the impacts of built environment on car ownership and use have been extensively studied, limited evidence has been offered for the role of spatial effects in influencing the interaction between built environment and travel behavior. Ignoring the spatial effects may lead to misunderstanding the role of the built environment and providing inconsistent transportation policies. In response to this, we try to employ a two-step modeling approach to investigate the impacts of built environment on car ownership and use by combining multilevel Bayesian model and conditional autocorrelation (CAR) model to control for spatial autocorrelation. In the two-step model, the predicting car ownership status in the first-step model is used as a mediating variable in the second-step car use model. Taking Changchun as a case study, this paper identifies the presence of spatial effects in influencing the effects of built environment on car ownership and use. Meanwhile, the direct and cascading effects of built environment on car ownership and use are revealed. The results show that the spatial autocorrelation exists in influencing the interaction between built environment and car dependency. The results suggest that it is necessary for urban planners to pay attention to the spatial effects and make targeted policy according to local land use characteristics.


Subject(s)
Automobiles/statistics & numerical data , Built Environment/statistics & numerical data , Ownership/statistics & numerical data , Bayes Theorem , China , Humans , Spatial Analysis , Travel
7.
JACC Basic Transl Sci ; 3(3): 350-362, 2018 Jun.
Article in English | MEDLINE | ID: mdl-30062222

ABSTRACT

Management for patients with diabetes experiencing myocardial infarction remains a challenge. Here the authors show that hyperglycemia- and hyperinsulinemia-induced microRNA-24 (miR-24) reduction and O-GlcNAcylation in the diabetic heart contribute to poor survival and increased infarct size in diabetic myocardial ischemia/reperfusion (I/R). In a mouse model of myocardial I/R, pharmacological or genetic overexpression of miR-24 in hearts significantly reduced myocardial infarct size. Experimental validation revealed that miR-24 targets multiple key proteins, including O-GlcNac transferase, ATG4A, and BIM, to coordinately protect the myocardium from I/R injury. These results establish miR-24 as a promising therapeutic candidate for diabetic I/R injury.

8.
Stem Cells Int ; 2018: 3814747, 2018.
Article in English | MEDLINE | ID: mdl-29743891

ABSTRACT

Direct reprogramming of cardiac fibroblasts (CFs) to induced cardiomyocytes (iCMs) is a newly emerged promising approach for cardiac regeneration, disease modeling, and drug discovery. However, its potential has been drastically limited due to the low reprogramming efficiency and largely unknown underlying molecular mechanisms. We have previously screened and identified epigenetic factors related to histone modification during iCM reprogramming. Here, we used shRNAs targeting an additional battery of epigenetic factors involved in chromatin remodeling and RNA splicing factors to further identify inhibitors and facilitators of direct cardiac reprogramming. Knockdown of RNA splicing factors Sf3a1 or Sf3b1 significantly reduced the percentage and total number of cardiac marker positive iCMs accompanied with generally repressed gene expression. Removal of another RNA splicing factor Zrsr2 promoted the acquisition of CM molecular features in CFs and mouse embryonic fibroblasts (MEFs) at both protein and mRNA levels. Moreover, a consistent increase of reprogramming efficiency was observed in CFs and MEFs treated with shRNAs targeting Bcor (component of BCOR complex superfamily) or Stag2 (component of cohesin complex). Our work thus reveals several additional epigenetic and splicing factors that are either inhibitory to or required for iCM reprogramming and highlights the importance of epigenetic regulation and RNA splicing process during cell fate conversion.

9.
Nature ; 551(7678): 100-104, 2017 11 02.
Article in English | MEDLINE | ID: mdl-29072293

ABSTRACT

Direct lineage conversion offers a new strategy for tissue regeneration and disease modelling. Despite recent success in directly reprogramming fibroblasts into various cell types, the precise changes that occur as fibroblasts progressively convert to the target cell fates remain unclear. The inherent heterogeneity and asynchronous nature of the reprogramming process renders it difficult to study this process using bulk genomic techniques. Here we used single-cell RNA sequencing to overcome this limitation and analysed global transcriptome changes at early stages during the reprogramming of mouse fibroblasts into induced cardiomyocytes (iCMs). Using unsupervised dimensionality reduction and clustering algorithms, we identified molecularly distinct subpopulations of cells during reprogramming. We also constructed routes of iCM formation, and delineated the relationship between cell proliferation and iCM induction. Further analysis of global gene expression changes during reprogramming revealed unexpected downregulation of factors involved in mRNA processing and splicing. Detailed functional analysis of the top candidate splicing factor, Ptbp1, revealed that it is a critical barrier for the acquisition of cardiomyocyte-specific splicing patterns in fibroblasts. Concomitantly, Ptbp1 depletion promoted cardiac transcriptome acquisition and increased iCM reprogramming efficiency. Additional quantitative analysis of our dataset revealed a strong correlation between the expression of each reprogramming factor and the progress of individual cells through the reprogramming process, and led to the discovery of new surface markers for the enrichment of iCMs. In summary, our single-cell transcriptomics approaches enabled us to reconstruct the reprogramming trajectory and to uncover intermediate cell populations, gene pathways and regulators involved in iCM induction.


Subject(s)
Cellular Reprogramming/genetics , Fibroblasts/cytology , Fibroblasts/metabolism , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Single-Cell Analysis , Transcriptome , Algorithms , Animals , Cell Lineage/genetics , Down-Regulation/genetics , GATA4 Transcription Factor/genetics , Heterogeneous-Nuclear Ribonucleoproteins/deficiency , Heterogeneous-Nuclear Ribonucleoproteins/genetics , Heterogeneous-Nuclear Ribonucleoproteins/metabolism , MEF2 Transcription Factors/genetics , Mice , Polypyrimidine Tract-Binding Protein/deficiency , Polypyrimidine Tract-Binding Protein/genetics , Polypyrimidine Tract-Binding Protein/metabolism , RNA Splicing/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , T-Box Domain Proteins/genetics
10.
Cell Rep ; 20(13): 3014-3024, 2017 Sep 26.
Article in English | MEDLINE | ID: mdl-28954220

ABSTRACT

Cardiomyocytes derived from induced pluripotent stem cells (iPSC-CMs) or directly reprogrammed from non-myocytes (induced cardiomyocytes [iCMs]) are promising sources for heart regeneration or disease modeling. However, the similarities and differences between iPSC-CMs and iCMs are still unknown. Here, we performed transcriptome analyses of beating iPSC-CMs and iCMs generated from cardiac fibroblasts (CFs) of the same origin. Although both iPSC-CMs and iCMs establish CM-like molecular features globally, iPSC-CMs exhibit a relatively hyperdynamic epigenetic status, whereas iCMs exhibit a maturation status that more closely resembles that of adult CMs. Based on gene expression of metabolic enzymes, iPSC-CMs primarily employ glycolysis, whereas iCMs utilize fatty acid oxidation as the main pathway. Importantly, iPSC-CMs and iCMs exhibit different cell-cycle status, alteration of which influenced their maturation. Therefore, our study provides a foundation for understanding the pros and cons of different reprogramming approaches.


Subject(s)
Gene Expression Profiling/methods , Gene Expression/genetics , Myocytes, Cardiac/metabolism , Cell Differentiation , Cellular Reprogramming , Humans
11.
Gene ; 592(2): 325-30, 2016 Nov 05.
Article in English | MEDLINE | ID: mdl-27390088

ABSTRACT

It has been long recognized that the mammalian heart loses its proliferative capacity soon after birth, yet, the molecular basis of this loss of cardiac proliferation postnatally is largely unknown. In this study, we found that cardiac ErbB2, a member of the epidermal growth factor receptor family, exhibits a rapid and dramatic decline in expression at the neonatal stage. We further demonstrate that conditional ablation of ErbB2 in the ventricular myocardium results in upregulation of negative cell cycle regulators and a significant reduction in cardiomyocyte proliferation during the narrow neonatal proliferative time window. Together, our data reveal a positive correlation between the expression levels of ErbB2 with neonatal cardiomyocyte proliferation and suggest that reduction in cardiac ErbB2 expression may contribute to the loss of postnatal cardiomyocyte proliferative capacity.


Subject(s)
Cell Proliferation , Heart/growth & development , Myocytes, Cardiac/metabolism , Receptor, ErbB-2/metabolism , Animals , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Gene Expression Regulation, Developmental , Mice , Myocytes, Cardiac/physiology , Receptor, ErbB-2/genetics
12.
Genesis ; 54(7): 398-406, 2016 07.
Article in English | MEDLINE | ID: mdl-27194122

ABSTRACT

Direct reprogramming of fibroblasts into induced cardiomyocytes (iCMs) through forced expression of cardiac-lineage specific transcription factors holds promise as an alternative strategy for cardiac regeneration. To facilitate research in iCM reprogramming, we generated a suite of new tools. We developed a transformed cell line derived from mouse embryonic fibroblasts (MEF). This fibroblast cell line (MEF-T) harbors an αMHC-eGFP reporter transgene for rapid detection of newly derived iCMs. The MEF-T cell line is highly proliferative and easily transfected and transduced, making it an ideal tool for transgene expression and genetic manipulation. Additionally, we generated a Tet-On inducible polycistronic iCM reprogramming construct for the temporal regulation of reprogramming factor expression. Furthermore, we introduced this construct into MEF-T and created an inducible reprogrammable fibroblast cell line. These tools will facilitate future research in cell fate reprogramming by enabling the temporal control of reprogramming factor expression as well as high-throughput screening using libraries of small molecules, noncoding RNAs, and siRNAs. genesis 54:398-406, 2016. © 2016 Wiley Periodicals, Inc.


Subject(s)
Cell Differentiation/genetics , Cellular Reprogramming/genetics , Myocytes, Cardiac/metabolism , Regeneration/genetics , Animals , Fibroblasts/metabolism , Gene Expression Regulation, Developmental , Heart/growth & development , Mice , Mice, Transgenic , Myocardium/metabolism
13.
Stem Cell Res ; 16(2): 507-18, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26957038

ABSTRACT

Direct conversion of fibroblasts into induced cardiomyocytes (iCMs) offers an alternative strategy for cardiac disease modeling and regeneration. During iCM reprogramming, the starting fibroblasts must overcome existing epigenetic barriers to acquire the CM-like chromatin pattern. However, epigenetic dynamics along this reprogramming process have not been studied. Here, we took advantage of our recently generated polycistronic system and determined the dynamics of two critical histone marks, H3K27me3 and H3K4me3, in parallel with gene expression at a set of carefully selected cardiac and fibroblast loci during iCM reprogramming. We observed reduced H3K27me3 and increased H3K4me3 at cardiac promoters as early as day 3, paralleled by a rapid significant increase in their mRNA expression. In contrast, H3K27me3 at loci encoding fibroblast marker genes did not increase until day 10 and H3K4me3 progressively decreased along the reprogramming process; these changes were accompanied by a gradual decrease in the mRNA expression of fibroblast marker genes. Further analyses of fibroblast-enriched transcription factors revealed a similarly late deposition of H3K27me3 and decreased mRNA expression of Sox9, Twist1 and Twist2, three important players in epithelial-mesenchymal transition. Our data suggest early rapid activation of the cardiac program and later progressive suppression of fibroblast fate at both epigenetic and transcriptional levels. Additionally, we determined the DNA methylation states of representative cardiac promoters and found that not every single CpG was equally demethylated during early stages of iCM reprogramming. Rather, there are specific CpGs, whose demethylation states correlated tightly with transcription activation, that we propose are the major contributing CpGs. Our work thus reveals a differential re-patterning of H3K27me3, H3K4me3 at cardiac and fibroblast loci during iCM reprogramming and could provide future genome-wide epigenetic studies with important guidance such as the appropriate time window and loci to be utilized as positive and negative controls.


Subject(s)
DNA Methylation , Fibroblasts/cytology , Histones/metabolism , Myocytes, Cardiac/cytology , Animals , Cell Line , Cellular Reprogramming , Epigenesis, Genetic , Fibroblasts/metabolism , Flow Cytometry , Mice , Mice, Transgenic , Microscopy, Fluorescence , Myocytes, Cardiac/metabolism , Promoter Regions, Genetic , RNA, Messenger/metabolism , Real-Time Polymerase Chain Reaction , SOX9 Transcription Factor/genetics , SOX9 Transcription Factor/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Transcriptional Activation , Twist-Related Protein 1/genetics , Twist-Related Protein 1/metabolism , Twist-Related Protein 2/genetics , Twist-Related Protein 2/metabolism
14.
Cell Stem Cell ; 18(3): 382-95, 2016 Mar 03.
Article in English | MEDLINE | ID: mdl-26942853

ABSTRACT

Direct reprogramming of induced cardiomyocytes (iCMs) suffers from low efficiency and requires extensive epigenetic repatterning, although the underlying mechanisms are largely unknown. To address these issues, we screened for epigenetic regulators of iCM reprogramming and found that reducing levels of the polycomb complex gene Bmi1 significantly enhanced induction of beating iCMs from neonatal and adult mouse fibroblasts. The inhibitory role of Bmi1 in iCM reprogramming is mediated through direct interactions with regulatory regions of cardiogenic genes, rather than regulation of cell proliferation. Reduced Bmi1 expression corresponded with increased levels of the active histone mark H3K4me3 and reduced levels of repressive H2AK119ub at cardiogenic loci, and de-repression of cardiogenic gene expression during iCM conversion. Furthermore, Bmi1 deletion could substitute for Gata4 during iCM reprogramming. Thus, Bmi1 acts as a critical epigenetic barrier to iCM production. Bypassing this barrier simplifies iCM generation and increases yield, potentially streamlining iCM production for therapeutic purposes.


Subject(s)
Cell Proliferation , Cellular Reprogramming , Epigenesis, Genetic , Gene Deletion , Myocytes, Cardiac/metabolism , Polycomb Repressive Complex 1/metabolism , Proto-Oncogene Proteins/metabolism , Animals , GATA4 Transcription Factor/genetics , GATA4 Transcription Factor/metabolism , Mice , Myocytes, Cardiac/cytology , Polycomb Repressive Complex 1/genetics , Proto-Oncogene Proteins/genetics
15.
J Vis Exp ; (105)2015 Nov 13.
Article in English | MEDLINE | ID: mdl-26649751

ABSTRACT

Direct conversion of cardiac fibroblasts (CFs) into induced cardiomyocytes (iCMs) holds great potential for regenerative medicine by offering alternative strategies for treatment of heart disease. This conversion has been achieved by forced expression of defined factors such as Gata4 (G), Mef2c (M) and Tbx5 (T). Traditionally, iCMs are generated by a cocktail of viruses expressing these individual factors. However, reprogramming efficiency is relatively low and most of the in vitro G,M,T-transduced fibroblasts do not become fully reprogrammed, making it difficult to study the reprogramming mechanisms. We recently have shown that the stoichiometry of G,M,T is crucial for efficient iCM reprogramming. An optimal stoichiometry of G,M,T with relative high level of M and low levels of G and T achieved by using our polycistronic MGT vector (hereafter referred to as MGT) significantly increased reprogramming efficiency and improved iCM quality in vitro. Here we provide a detailed description of the methodology used to generate iCMs with MGT construct from cardiac fibroblasts. Isolation of cardiac fibroblasts, generation of virus for reprogramming and evaluation of the reprogramming process are also included to provide a platform for efficient and reproducible generation of iCMs.

16.
Elife ; 4: e09406, 2015 Oct 16.
Article in English | MEDLINE | ID: mdl-26473617

ABSTRACT

Truncating mutations in the giant sarcomeric protein Titin result in dilated cardiomyopathy and skeletal myopathy. The most severely affected dilated cardiomyopathy patients harbor Titin truncations in the C-terminal two-thirds of the protein, suggesting that mutation position might influence disease mechanism. Using CRISPR/Cas9 technology, we generated six zebrafish lines with Titin truncations in the N-terminal and C-terminal regions. Although all exons were constitutive, C-terminal mutations caused severe myopathy whereas N-terminal mutations demonstrated mild phenotypes. Surprisingly, neither mutation type acted as a dominant negative. Instead, we found a conserved internal promoter at the precise position where divergence in disease severity occurs, with the resulting protein product partially rescuing N-terminal truncations. In addition to its clinical implications, our work may shed light on a long-standing mystery regarding the architecture of the sarcomere.


Subject(s)
Cardiomyopathy, Dilated/pathology , Connectin/genetics , Muscular Diseases/pathology , Promoter Regions, Genetic , Sequence Deletion , Animals , Connectin/metabolism , Disease Models, Animal , Humans , Mutant Proteins/genetics , Mutant Proteins/metabolism , Zebrafish
18.
Circ Res ; 116(2): 237-44, 2015 Jan 16.
Article in English | MEDLINE | ID: mdl-25416133

ABSTRACT

RATIONALE: Generation of induced cardiac myocytes (iCMs) directly from fibroblasts offers great opportunities for cardiac disease modeling and cardiac regeneration. A major challenge of iCM generation is the low conversion rate of fibroblasts to fully reprogrammed iCMs, which could in part be attributed to unbalanced expression of reprogramming factors Gata4 (G), Mef2c (M), and Tbx5 (T) using the current gene delivery approach. OBJECTIVE: We aimed to establish a system to express distinct ratios of G, M, T proteins in fibroblasts and determine the effect of G, M, T stoichiometry on iCM reprogramming. METHODS AND RESULTS: We took advantage of the inherent feature of the polycistronic system and generated all possible combinations of G, M, T with identical 2A sequences in a single transgene. We demonstrated that each splicing order of G, M, T gave rise to distinct G, M, T protein expression levels. Combinations that resulted in higher protein level of Mef2c with lower levels of Gata4 and Tbx5 significantly enhanced reprogramming efficiency compared with separate G, M, T transduction. Importantly, after further optimization, the MGT vector resulted in more than 10-fold increase in the number of mature beating iCM loci. Molecular characterization revealed that more optimal G, M, T stoichiometry correlated with higher expression of mature cardiac myocyte markers. CONCLUSIONS: Our results demonstrate that stoichiometry of G, M, T protein expression influences the efficiency and quality of iCM reprogramming. The established optimal G, M, T expression condition will provide a valuable platform for future iCM studies.


Subject(s)
Cellular Reprogramming/physiology , GATA4 Transcription Factor/biosynthesis , Myocytes, Cardiac/physiology , T-Box Domain Proteins/biosynthesis , Animals , Cells, Cultured , GATA4 Transcription Factor/genetics , MEF2 Transcription Factors/biosynthesis , MEF2 Transcription Factors/genetics , Mice , Mice, Transgenic , T-Box Domain Proteins/genetics
19.
Proc Natl Acad Sci U S A ; 110(44): 17933-8, 2013 Oct 29.
Article in English | MEDLINE | ID: mdl-24114272

ABSTRACT

Glioblastoma (GBM), the most common brain malignancy, remains fatal with no effective treatment. Analyses of common aberrations in GBM suggest major regulatory pathways associated with disease etiology. However, 90% of GBMs are diagnosed at an advanced stage (primary GBMs), providing no access to early disease stages for assessing disease progression events. As such, both understanding of disease mechanisms and the development of biomarkers and therapeutics for effective disease management are limited. Here, we describe an adult-inducible astrocyte-specific system in genetically engineered mice that queries causation in disease evolution of regulatory networks perturbed in human GBM. Events yielding disease, both engineered and spontaneous, indicate ordered grade-specific perturbations that yield high-grade astrocytomas (anaplastic astrocytomas and GBMs). Impaired retinoblastoma protein RB tumor suppression yields grade II histopathology. Additional activation of v-Ki-ras2 Kirsten rat sarcoma viral oncogene homolog (KRAS) network drives progression to grade III disease, and further inactivation of phosphatase and tensin homolog (PTEN) yields GBM. Spontaneous missense mutation of tumor suppressor Trp53 arises subsequent to KRAS activation, but before grade III progression. The stochastic appearance of mutations identical to those observed in humans, particularly the same spectrum of p53 amino acid changes, supports the validity of engineered lesions and the ensuing interpretations of etiology. Absence of isocitrate dehydrogenase 1 (IDH1) mutation, asymptomatic low grade disease, and rapid emergence of GBM combined with a mesenchymal transcriptome signature reflect characteristics of primary GBM and provide insight into causal relationships.


Subject(s)
Astrocytoma/etiology , Biological Evolution , Disease Models, Animal , Genetic Engineering/methods , Glioblastoma/etiology , Animals , Base Sequence , Disease Progression , Gene Expression Profiling , Gene Regulatory Networks/genetics , Mice , Mice, Transgenic , Microarray Analysis , Molecular Sequence Data , Mutation, Missense/genetics , PTEN Phosphohydrolase/metabolism , Proto-Oncogene Proteins p21(ras)/genetics , Sequence Analysis, DNA , Tumor Suppressor Protein p53/genetics
20.
Mol Ther ; 21(12): 2205-16, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23939025

ABSTRACT

A drawback of gene therapy using adeno-associated virus (AAV) is the DNA packaging restriction of the viral capsid (<4.7 kb). Recent observations demonstrate oversized AAV genome transduction through an unknown mechanism. Herein, AAV production using an oversized reporter (6.2 kb) resulted in chloroform and DNase-resistant particles harboring distinct "fragment" AAV (fAAV) genomes (5.0, 2.4, and 1.6 kb). Fractionation experiments determined that only the larger "fragments" mediated transduction in vitro, and relatively efficient transduction was also demonstrated in the muscle, the eye, and the liver. In contrast with concatemerization-dependent large-gene delivery by split AAV, fAAV transduction is independent of the catalytic subunit of DNA-dependent protein kinase (DNA-PKcs) in vitro and in vivo while disproportionately reliant on the DNA strand-annealing protein Rad51C. Importantly, fAAV's unique dependence on DNA repair proteins, compared with intact AAV, strongly suggests that the majority of oversized AAV transduction is mediated by fragmented genomes. Although fAAV transduction is less efficient than intact AAV, it is enhanced fourfold in muscle and sevenfold in the retina compared with split AAV transduction. Furthermore, fAAV carrying codon-optimized therapeutic dysferlin cDNA in a 7.5 kb expression cassette restored dysferlin levels in a dystrophic model. Collectively, oversized AAV genome transduction requires unique DNA repair pathways and offers an alternative, more efficient strategy for large-gene therapy.


Subject(s)
Capsid/metabolism , DNA-Binding Proteins/genetics , Dependovirus/physiology , Genetic Therapy , Liver/virology , Muscles/virology , Rad51 Recombinase/genetics , Transduction, Genetic , Animals , CHO Cells , Cricetulus , DNA Packaging , DNA, Viral/genetics , DNA-Binding Proteins/metabolism , Dependovirus/genetics , Disease Models, Animal , Eye/virology , Genes, Reporter , Genetic Vectors , Genome, Viral , HEK293 Cells , HeLa Cells , Humans , Mice , Muscle Proteins/genetics , Muscle Proteins/metabolism , Muscle, Skeletal/virology , Nucleic Acid Conformation , Rad51 Recombinase/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...