Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 200
1.
Sci Rep ; 14(1): 9396, 2024 04 24.
Article En | MEDLINE | ID: mdl-38658615

In a previous report, we demonstrated that Cbx1, PurB and Sp3 inhibited cardiac muscle differentiation by increasing nucleosome density around cardiac muscle gene promoters. Since cardiac and skeletal muscle express many of the same proteins, we asked if Cbx1, PurB and Sp3 similarly regulated skeletal muscle differentiation. In a C2C12 model of skeletal muscle differentiation, Cbx1 and PurB knockdown increased myotube formation. In contrast, Sp3 knockdown inhibited myotube formation, suggesting that Sp3 played opposing roles in cardiac muscle and skeletal muscle differentiation. Consistent with this finding, Sp3 knockdown also inhibited various muscle-specific genes. The Cbx1, PurB and Sp3 proteins are believed to influence gene-expression in part by altering nucleosome position. Importantly, we developed a statistical approach to determine if changes in nucleosome positioning were significant and applied it to understanding the architecture of muscle-specific genes. Through this novel statistical approach, we found that during myogenic differentiation, skeletal muscle-specific genes undergo a set of unique nucleosome changes which differ significantly from those shown in commonly expressed muscle genes. While Sp3 binding was associated with nucleosome loss, there appeared no correlation with the aforementioned nucleosome changes. In summary, we have identified a novel role for Sp3 in skeletal muscle differentiation and through the application of quantifiable MNase-seq have discovered unique fingerprints of nucleosome changes for various classes of muscle genes during myogenic differentiation.


Cell Differentiation , Muscle Development , Muscle, Skeletal , Nucleosomes , Promoter Regions, Genetic , Nucleosomes/metabolism , Nucleosomes/genetics , Animals , Cell Differentiation/genetics , Mice , Muscle, Skeletal/metabolism , Muscle Development/genetics , Cell Line , Sp3 Transcription Factor/metabolism , Sp3 Transcription Factor/genetics , Muscle Fibers, Skeletal/metabolism
2.
J Mol Cell Cardiol ; 190: 48-61, 2024 May.
Article En | MEDLINE | ID: mdl-38582260

We have demonstrated that directly reprogramming cardiac fibroblasts into new cardiomyocytes via miR combo improves cardiac function in the infarcted heart. However, major challenges exist with delivery and efficacy. During a screening based approach to improve delivery, we discovered that C166-derived EVs were effective delivery agents for miR combo both in vitro and in vivo. In the latter, EV mediated delivery of miR combo induced significant conversion of cardiac fibroblasts into cardiomyocytes (∼20%), reduced fibrosis and improved cardiac function in a myocardial infarction injury model. When compared to lipid-based transfection, C166 EV mediated delivery of miR combo enhanced reprogramming efficacy. Improved reprogramming efficacy was found to result from a miRNA within the exosome: miR-148a-3p. The target of miR-148a-3p was identified as Mdfic. Over-expression and targeted knockdown studies demonstrated that Mdfic was a repressor of cardiomyocyte specific gene expression. In conclusion, we have demonstrated that C166-derived EVs are an effective method for delivering reprogramming factors to cardiac fibroblasts and we have identified a novel miRNA contained within C166-derived EVs which enhances reprogramming efficacy.


Cellular Reprogramming , Fibroblasts , MicroRNAs , Myocytes, Cardiac , MicroRNAs/genetics , MicroRNAs/metabolism , Animals , Cellular Reprogramming/genetics , Myocytes, Cardiac/metabolism , Fibroblasts/metabolism , Mice , Myocardial Infarction/metabolism , Myocardial Infarction/genetics , Myocardial Infarction/pathology , Myocardial Infarction/therapy , Extracellular Vesicles/metabolism , Extracellular Vesicles/genetics , Exosomes/metabolism , Gene Expression Regulation , Humans
3.
JAMA Intern Med ; 184(5): 465-466, 2024 May 01.
Article En | MEDLINE | ID: mdl-38497941

This Viewpoint discusses highlights from the National Academy of Medicine 2023 Annual Meeting Scientific Symposium that are representative of key gaps, trends, and opportunities in women's health.


Women's Health , Humans , Female
4.
Mol Ther Nucleic Acids ; 35(2): 102160, 2024 Jun 11.
Article En | MEDLINE | ID: mdl-38495845

Reprogramming scar fibroblasts into cardiomyocytes has been proposed to reverse the damage associated with myocardial infarction. However, the limited improvement in cardiac function calls for enhanced strategies. We reported enhanced efficacy of our miR reprogramming cocktail miR combo (miR-1, miR-133a, miR-208a, and miR-499) via RNA-sensing receptor stimulation. We hypothesized that we could combine RNA-sensing receptor activation with fibroblast reprogramming by chemically modifying miR combo. To test the hypothesis, miR combo was modified to enhance interaction with the RNA-sensing receptor Rig1 via the addition of a 5'-triphosphate (5'ppp) group. Importantly, when compared with unmodified miR combo, 5'ppp-modified miR combo markedly improved reprogramming efficacy in vitro. Enhanced reprogramming efficacy correlated with a type-I interferon immune response with strong and selective secretion of interferon ß (IFNß). Antibody blocking studies and media replacement experiments indicated that 5'ppp-miR combo utilized IFNß to enhance fibroblast reprogramming efficacy. In conclusion, miRs can acquire powerful additional roles through chemical modification that potentially increases their clinical applications.

5.
Circulation ; 149(9): 707-716, 2024 02 27.
Article En | MEDLINE | ID: mdl-38408142

RNA therapeutics hold significant promise in the treatment of cardiovascular diseases. RNAs are biologically diverse and functionally specific and can be used for gain- or loss-of-function purposes. The effectiveness of mRNA-based vaccines in the recent COVID-19 pandemic has undoubtedly proven the benefits of an RNA-based approach. RNA-based therapies are becoming more common as a treatment modality for cardiovascular disease. This is most evident in hypertension where several small interfering RNA-based drugs have proven to be effective in managing high blood pressure in several clinical trials. As befits a rapidly burgeoning field, there is significant interest in other classes of RNA. Revascularization of the infarcted heart through an mRNA drug is under clinical investigation. mRNA technology may provide the platform for the expression of paracrine factors for myocardial protection and regeneration. Emergent technologies on the basis of microRNAs and gene editing are tackling complex diseases in a novel fashion. RNA-based gene editing offers hope of permanent cures for monogenic cardiovascular diseases, and long-term control of complex diseases such as essential hypertension, as well. Likewise, microRNAs are proving effective in regenerating cardiac muscle. The aim of this review is to provide an overview of the current landscape of RNA-based therapies for the treatment of cardiovascular disease. The review describes the large number of RNA molecules that exist with a discussion of the clinical development of each RNA type. In addition, the review also presents a number of avenues for future development.


Cardiovascular Diseases , Cardiovascular System , MicroRNAs , Humans , Cardiovascular Diseases/therapy , Cardiovascular Diseases/drug therapy , Pandemics , MicroRNAs/genetics , MicroRNAs/therapeutic use , RNA, Small Interfering/genetics , RNA, Messenger/genetics , RNA, Messenger/therapeutic use
6.
Hypertension ; 81(4): 702-708, 2024 Apr.
Article En | MEDLINE | ID: mdl-38112080

Hypertension affects >1 billion people worldwide. Complications of hypertension include stroke, renal failure, cardiac hypertrophy, myocardial infarction, and cardiac failure. Despite the development of various antihypertensive drugs, the number of people with uncontrolled hypertension continues to rise. While the lack of compliance associated with frequent side effects to medication is a contributory issue, there has been a failure to consider the diverse nature of hypertensive populations. Instead, we propose that hypertension can only be truly managed by precision. A precision medicine approach would consider each patient's unique factors. In this review, we discuss the progress toward precision medicine for hypertension with more predictiveness and individualization of treatment. We will highlight the advances in data science, omics (genomics, metabolomics, proteomics, etc), artificial intelligence, gene therapy, and gene editing and their application to precision hypertension.


Artificial Intelligence , Hypertension , Humans , Hypertension/genetics , Hypertension/therapy , Hypertension/complications , Antihypertensive Agents , Genomics , Proteomics
8.
Science ; 381(6662): 1029, 2023 Sep 08.
Article En | MEDLINE | ID: mdl-37676960

The social benefit of technologies is frequently unevenly realized across the United States. Rural communities, individuals with disabilities, and historically marginalized groups face out-of-reach costs or lack access to products that meet their needs. Blame is typically placed on complicated regulatory processes or complex delivery systems, but this response neglects the problem that equity is not baked into the nation's innovation process at any stage. The United States needs to rethink its entire innovation ecosystem to incorporate equity as a foundational guiding principle-from research design and funding requirements to policies and regulations that govern the delivery and oversight of new products to the public.

10.
J Biol Chem ; 299(5): 104694, 2023 05.
Article En | MEDLINE | ID: mdl-37044217

Directly reprogramming fibroblasts into cardiomyocytes improves cardiac function in the infarcted heart. However, the low efficacy of this approach hinders clinical applications. Unlike the adult mammalian heart, the neonatal heart has an intrinsic regenerative capacity. Consequently, we hypothesized that birth imposes fundamental changes in cardiac fibroblasts which limit their regenerative capabilities. In support, we found that reprogramming efficacy in vitro was markedly lower with fibroblasts derived from adult mice versus those derived from neonatal mice. Notably, fibroblasts derived from adult mice expressed significantly higher levels of pro-angiogenic genes. Moreover, under conditions that promote angiogenesis, only fibroblasts derived from adult mice differentiated into tube-like structures. Targeted knockdown screening studies suggested a possible role for the transcription factor Epas1. Epas1 expression was higher in fibroblasts derived from adult mice, and Epas1 knockdown improved reprogramming efficacy in cultured adult cardiac fibroblasts. Promoter activity assays indicated that Epas1 functions as both a transcription repressor and an activator, inhibiting cardiomyocyte genes while activating angiogenic genes. Finally, the addition of an Epas1 targeting siRNA to the reprogramming cocktail markedly improved reprogramming efficacy in vivo with both the number of reprogramming events and cardiac function being markedly improved. Collectively, our results highlight differences between neonatal and adult cardiac fibroblasts and the dual transcriptional activities of Epas1 related to reprogramming efficacy.


Cellular Reprogramming , Myocytes, Cardiac , Transcription Factors , Animals , Mice , Fibroblasts/cytology , Gene Expression Regulation , Myocytes, Cardiac/cytology , Transcription Factors/metabolism , Animals, Newborn
11.
Sci Rep ; 13(1): 3920, 2023 03 09.
Article En | MEDLINE | ID: mdl-36894665

Current methods to generate cardiomyocytes from induced pluripotent stem cells (iPSc) utilize broad-spectrum pharmacological inhibitors. These methods give rise to cardiomyocytes which are typically immature. Since we have recently demonstrated that cardiomyogenesis in vitro and in vivo requires Sfrp2, we asked if Sfrp2 would drive differentiation of human iPSc into cardiomyocytes. Indeed, we found that Sfrp2 induced robust cardiac differentiation. Importantly, replacement of broad spectrum pharmacological inhibitors with Sfrp2 gave rise to mature cardiomyocytes as evidenced by their sarcomere structure, electrophysiological profiles, and ability to form gap junctions.


Induced Pluripotent Stem Cells , Myocytes, Cardiac , Humans , Cell Differentiation , Electrophysiological Phenomena , Membrane Proteins
13.
JAMA ; 329(17): 1445-1446, 2023 05 02.
Article En | MEDLINE | ID: mdl-36939751

This Viewpoint from the president of the National Academy of Medicine looks back at the academy's accomplishments and looks forward to the possibilities it hopes to achieve to better medicine and society.


Medicine , National Academies of Science, Engineering, and Medicine, U.S., Health and Medicine Division , Academies and Institutes , United States , Health/trends , Forecasting
14.
Am J Physiol Cell Physiol ; 324(4): C843-C855, 2023 04 01.
Article En | MEDLINE | ID: mdl-36847443

We discovered that innate immunity plays an important role in the reprogramming of fibroblasts into cardiomyocytes. In this report, we define the role of a novel retinoic acid-inducible gene 1 Yin Yang 1 (Rig1:YY1) pathway. We found that fibroblast to cardiomyocyte reprogramming efficacy was enhanced by specific Rig1 activators. To understand the mechanism of action, we performed various transcriptomic, nucleosome occupancy, and epigenomic approaches. Analysis of the datasets indicated that Rig1 agonists had no effect on reprogramming-induced changes in nucleosome occupancy or loss of inhibitory epigenetic motifs. Instead, Rig1 agonists were found to modulate cardiac reprogramming by promoting the binding of YY1 specifically to cardiac genes. To conclude, these results show that the Rig1:YY1 pathway plays a critical role in fibroblast to cardiomyocyte reprogramming.


Nucleosomes , Receptors, Retinoic Acid , Carrier Proteins/metabolism , Fibroblasts/metabolism , Myocytes, Cardiac/metabolism , Receptors, Retinoic Acid/genetics , Receptors, Retinoic Acid/metabolism , Signal Transduction , Humans , Animals
16.
Milbank Q ; 100(3): 673-701, 2022 09.
Article En | MEDLINE | ID: mdl-36148893

Policy Points Hospital-at-Home (HaH) is a home-based alternative for acute care that has expanded significantly under COVID-19 regulatory flexibilities. The post-pandemic policy agenda for HaH will require consideration of multistakeholder perspectives, including patient, caregiver, provider, clinical operations, technology, equity, legal, quality, and payer. Key policy challenges include reaching a consensus on program standards, clarifying caregivers' issues, creating sustainable reimbursement mechanisms, and mitigating potential equity concerns. Key policy prescriptions include creating a national surveillance system for quality and safety, clarifying legal standards for care in the home, and deploying payment reforms through value-based models.


COVID-19 , COVID-19/epidemiology , Caregivers , Hospitals , Humans , Reimbursement Mechanisms
19.
J Biol Chem ; 298(6): 102053, 2022 06.
Article En | MEDLINE | ID: mdl-35605661

miRNA-based cellular fate reprogramming offers an opportunity to investigate the mechanisms of long-term gene silencing. To further understand how genes are silenced in a tissue-specific manner, we leveraged our miRNA-based method of reprogramming fibroblasts into cardiomyocytes. Through screening approaches, we identified three proteins that were downregulated during reprogramming of fibroblasts into cardiomyocytes: heterochromatin protein Cbx1, transcriptional activator protein PurB, and transcription factor Sp3. We show that knockdown of Cbx1, PurB, and Sp3 was sufficient to induce cardiomyocyte gene expression in fibroblasts. Similarly, gene editing to ablate Cbx1, PurB, and Sp3 expression induced fibroblasts to convert into cardiomyocytes in vivo. Furthermore, high-throughput DNA sequencing and coimmunoprecipitation experiments indicated that Cbx1, PurB, and Sp3 also bound together as a complex and were necessary to localize nucleosomes to cardiomyocyte genes on the chromosome. Finally, we found that the expression of these genes led to nucleosome modification via H3K27me3 (trimethylated histone-H3 lysine-27) deposition through an interaction with the polycomb repressive PRC2 complex. In summary, we conclude that Cbx1, PurB, and Sp3 control cell fate by actively repressing lineage-specific genes.


Cellular Reprogramming , Chromobox Protein Homolog 5 , DNA-Binding Proteins , Gene Silencing , Sp3 Transcription Factor , Animals , Chromobox Protein Homolog 5/genetics , Chromobox Protein Homolog 5/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Heterochromatin/metabolism , Humans , Mice , MicroRNAs/genetics , Myocytes, Cardiac/metabolism , Polycomb-Group Proteins/genetics , Polycomb-Group Proteins/metabolism , Sp3 Transcription Factor/genetics , Sp3 Transcription Factor/metabolism
...