Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 54
Filter
1.
Sci Rep ; 14(1): 9396, 2024 04 24.
Article in English | MEDLINE | ID: mdl-38658615

ABSTRACT

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.


Subject(s)
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 in English | MEDLINE | ID: mdl-38582260

ABSTRACT

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.


Subject(s)
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.
Mol Ther Nucleic Acids ; 35(2): 102160, 2024 Jun 11.
Article in English | MEDLINE | ID: mdl-38495845

ABSTRACT

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.

4.
Circulation ; 149(9): 707-716, 2024 02 27.
Article in English | MEDLINE | ID: mdl-38408142

ABSTRACT

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.


Subject(s)
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 in English | MEDLINE | ID: mdl-38112080

ABSTRACT

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.


Subject(s)
Artificial Intelligence , Hypertension , Humans , Hypertension/genetics , Hypertension/therapy , Hypertension/complications , Antihypertensive Agents , Genomics , Proteomics
7.
J Biol Chem ; 299(5): 104694, 2023 05.
Article in English | MEDLINE | ID: mdl-37044217

ABSTRACT

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.


Subject(s)
Cellular Reprogramming , Myocytes, Cardiac , Transcription Factors , Animals , Mice , Fibroblasts/cytology , Gene Expression Regulation , Myocytes, Cardiac/cytology , Transcription Factors/metabolism , Animals, Newborn
8.
Sci Rep ; 13(1): 3920, 2023 03 09.
Article in English | MEDLINE | ID: mdl-36894665

ABSTRACT

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.


Subject(s)
Induced Pluripotent Stem Cells , Myocytes, Cardiac , Humans , Cell Differentiation , Electrophysiological Phenomena , Membrane Proteins
10.
Am J Physiol Cell Physiol ; 324(4): C843-C855, 2023 04 01.
Article in English | MEDLINE | ID: mdl-36847443

ABSTRACT

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.


Subject(s)
Nucleosomes , Receptors, Retinoic Acid , Carrier Proteins/metabolism , Fibroblasts/metabolism , Myocytes, Cardiac/metabolism , Receptors, Retinoic Acid/genetics , Receptors, Retinoic Acid/metabolism , Signal Transduction , Humans , Animals
11.
Biochem Biophys Rep ; 31: 101310, 2022 Sep.
Article in English | MEDLINE | ID: mdl-35860436

ABSTRACT

There is considerable interest in regenerating the injured heart by reprogramming resident fibroblasts into new functional cardiomyocytes. Cardiac reprogramming has been achieved via transcription factors or miRNAs. Transcription factor combinations appear to be species-specific as evidenced by the fact that combinations of transcription factors which are effective for the reprogramming of mouse fibroblasts are ineffective in pigs and humans. Whether miRNA based cardiac reprogramming suffers from the same limitation is unknown. We have previously demonstrated that mouse cardiac fibroblasts can be directly converted into cardiomyocytes both in vitro and in vivo via a combination of four microRNAs (miR-1, miR-133a, miR-208a and miR-499) termed "miR combo." To assess species-specificity, miR combo was transfected into cardiac fibroblasts isolated from the left ventricle of dogs, pigs and humans. QPCR analysis indicated that miR combo effectively reprogrammed fibroblasts from all of the tested mammalian species. Significant upregulation of cardiac developmental, sarcomere, and cardiac ion channel genes was observed. Through Actn2+ staining, we also found that miR combo transfection induced dog, pig and human cardiac fibroblasts to develop into cardiomyocyte-like cells. In conclusion, we have demonstrated that in contrast to transcription factor based approaches, miR combo effectively reprograms mammalian cardiac fibroblasts into cardiomyocyte-like cells.

12.
J Biol Chem ; 298(6): 102053, 2022 06.
Article in English | MEDLINE | ID: mdl-35605661

ABSTRACT

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.


Subject(s)
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
13.
Proc Natl Acad Sci U S A ; 118(33)2021 08 17.
Article in English | MEDLINE | ID: mdl-34380738

ABSTRACT

Cardiomyogenesis, the process by which the body generates cardiomyocytes, is poorly understood. We have recently shown that Sfrp2 promotes cardiomyogenesis in vitro. The objective of this study was to determine if Sfrp2 would similarly promote cardiomyogenesis in vivo. To test this hypothesis, we tracked multipotent cKit(+) cells in response to Sfrp2 treatment. In control adult mice, multipotent cKit(+) cells typically differentiated into endothelial cells but not cardiomyocytes. In contrast, Sfrp2 switched the fate of these cells. Following Sfrp2 injection, multipotent cKit(+) cells differentiated solely into cardiomyocytes. Sfrp2-derived cardiomyocytes integrated into the myocardium and exhibited identical physiological properties to preexisting native cardiomyocytes. The ability of Sfrp2 to promote cardiomyogenesis was further supported by tracking EdU-labeled cells. In addition, Sfrp2 did not promote the formation of new cardiomyocytes when the cKit(+) cell population was selectively ablated in vivo using a diphtheria toxin receptor-diphtheria toxin model. Notably, Sfrp2-induced cardiomyogenesis was associated with significant functional improvements in a cardiac injury model. In summary, our study further demonstrates the importance of Sfrp2 in cardiomyogenesis.


Subject(s)
Membrane Proteins/pharmacology , Myocardial Infarction/therapy , Animals , Calcium/metabolism , Cell Differentiation , Gene Expression Regulation , Membrane Proteins/metabolism , Mice , Mice, Transgenic , Myocardial Contraction/physiology , Myocytes, Cardiac
15.
Physiol Rep ; 9(3): e14678, 2021 02.
Article in English | MEDLINE | ID: mdl-33587322

ABSTRACT

In this review, we summarize the role of Wnt proteins in cardiomyogenesis. More specifically, we focus on how the development of cardiomyocytes from precursor cells involves a complex interplay between Wnt canonical ß-catenin signaling pathways and Wnt noncanonical signaling pathways involving PCP and JNK. We also describe recent literature which suggests that endogenous Wnt inhibitors such as the Sfrp and DKK proteins play important roles in regulating the cardiomyocyte differentiation.


Subject(s)
Cell Differentiation , Heart Diseases/metabolism , Heart/growth & development , Intercellular Signaling Peptides and Proteins/metabolism , Myocytes, Cardiac/metabolism , Wnt Signaling Pathway , Animals , Gene Expression Regulation, Developmental , Heart Diseases/genetics , Heart Diseases/pathology , Heart Diseases/physiopathology , Humans , Intercellular Signaling Peptides and Proteins/genetics , Morphogenesis , Myocytes, Cardiac/pathology , Phenotype , Recovery of Function , Regeneration
16.
Mol Ther Nucleic Acids ; 23: 55-62, 2021 Mar 05.
Article in English | MEDLINE | ID: mdl-33335792

ABSTRACT

Reprogramming scar fibroblasts into new heart muscle cells has the potential to restore function to the injured heart. However, the effectiveness of reprogramming is notably low. We have recently demonstrated that the effectiveness of reprogramming fibroblasts into heart muscle cells (cardiomyocytes) is increased by the addition of RNA-sensing receptor ligands. Clinical use of these ligands is problematic due to their ability to induce adverse inflammatory events. To overcome this issue, we sought to determine whether synthetic analogs of natural RNA-sensing receptor ligands, which avoid generating inflammatory insults and are nuclease resistant, would similarly enhance fibroblast reprogramming into cardiomyocytes. Indeed, one such stabilized RNA, ICR2, increased the expression of cardiomyocyte-specific mRNAs in reprogrammed fibroblasts. Moreover, ICR2 enhanced the ability of reprogramming factors to produce cardiomyocytes with mature sarcomeres. Knockdown assays indicated that the effects of ICR2 were mediated by the RNA-sensing receptors Rig-I and TLR3. In addition, ICR2 reduced the effective dose and number of reprogramming factors needed for efficient reprogramming. In summary, the synthetic RNA oligonucleotide ICR2 is a potential therapeutic agent to enhance cardiac reprogramming efficiency.

17.
Methods Mol Biol ; 2239: 47-59, 2021.
Article in English | MEDLINE | ID: mdl-33226612

ABSTRACT

There are currently no effective treatments to regenerate the heart after cardiac injury. Following cardiac injury, heart muscle cells, also known as cardiomyocytes, die in large numbers. The adult mammalian heart does not have the ability to replace these dead cardiomyocytes. In their place, fibroblasts invade the injury zone and generate a scar. The scar impairs cardiac function. An important approach to cardiac regeneration is direct cardiac reprogramming, whereby cardiac fibroblasts within the scar are directly converted into functional cardiomyocytes. Several laboratories have achieved direct cardiac reprogramming via overexpression of the cardiac transcription factors. In contrast, we utilize a combination of four miRNAs (miR-1, miR-133, miR-208, miR-499) that we call miR Combo. One common issue regarding direct cardiac reprogramming strategies is low efficiency. Recently, we have demonstrated that the efficiency of direct cardiac reprogramming is enhanced in the chemically defined reprogramming media.


Subject(s)
Cellular Reprogramming/genetics , Culture Media/chemistry , Fibroblasts/cytology , MicroRNAs/metabolism , Myocytes, Cardiac/cytology , Animals , Ascorbic Acid/chemistry , Cell Culture Techniques/methods , Cells, Cultured , Fibroblasts/metabolism , Flow Cytometry , Fluorescent Antibody Technique , Mice , MicroRNAs/genetics , Myocytes, Cardiac/metabolism , Real-Time Polymerase Chain Reaction , Regeneration , Selenium/chemistry , Transfection/methods
18.
Am J Physiol Cell Physiol ; 319(6): C1141-C1150, 2020 12 01.
Article in English | MEDLINE | ID: mdl-33026832

ABSTRACT

Stem cell injections are an attractive therapeutic tool. It has been demonstrated that injected stem cells promote tissue repair and regeneration via paracrine mechanisms. However, the effects of injected stem cells continue for far longer than they are present. We hypothesized that the effects of injected stem cells are prolonged because of a sequential paracrine relay mechanism. Conditioned media was collected from mesenchymal stem cells (MSCs) after 24 h. This media was then added to RAW264.7. Media was collected from the macrophages after 24 h and was then added to endothelial cells (ECs). This conditioned macrophage media, but not control media, promoted wound healing and induced EC differentiation. Similar results were observed with primary macrophages. To identify the active paracrine factors released by macrophages in response to stimulation by MSC conditioned media we used an antibody array, identifying increased expression of the angiogenesis-related proteins stromal cell-derived factor 1 (SDF1) and plasminogen activator inhibitor-1 (PAI-1). Knockdown of either protein inhibited the ability of conditioned media derived from MSC paracrine factor-stimulated macrophages to induce EC differentiation both in vitro and in vivo. Conditioned media derived from postnatal day 7 (P7) mouse macrophages induced EC differentiation. Moreover, SDF1 and PAI-1 levels were >120 higher in P7 macrophages compared with adult macrophages, suggesting that MSC paracrine factors promote adult macrophages to adopt a juvenile phenotype. These results indicate that MSC paracrine factors induce macrophages to secrete SDF1 and PAI-1, in-turn inducing endothelial cells to differentiate. Identification of a sequential paracrine mechanism opens new therapeutic avenues for stem cell therapy.


Subject(s)
Culture Media, Conditioned/pharmacology , Endothelial Cells/drug effects , Guided Tissue Regeneration/methods , Mesenchymal Stem Cell Transplantation , Paracrine Communication/physiology , Wound Healing/drug effects , Animals , Cell Differentiation/drug effects , Cell- and Tissue-Based Therapy/methods , Chemokine CXCL12/metabolism , Macrophages/cytology , Male , Mesenchymal Stem Cells/cytology , Mice , Mice, Inbred C57BL , Neovascularization, Physiologic/physiology , RAW 264.7 Cells , Serpin E2/metabolism
19.
Biochem Biophys Res Commun ; 533(1): 9-16, 2020 11 26.
Article in English | MEDLINE | ID: mdl-32917363

ABSTRACT

Following heart injury, cardiomyocytes, are lost and are not regenerated. In their place, fibroblasts invade the dead tissue where they generate a scar, which reduces cardiac function. We and others have demonstrated that combinations of specific miRNAs (miR combo) or transcription factors (GMT), delivered by individual lenti-/retro-viruses in vivo, can convert fibroblasts into cardiomyocytes and improve cardiac function. However, the effects are relatively modest due to the low efficiency of delivery of miR combo or GMT. We hypothesized that efficiency would be improved by optimizing delivery. In the first instance, we developed a multicistronic system to express all four miRNAs of miR combo from a single construct. The order of each miRNA in the multicistronic construct gave rise to different levels of miRNA expression. A combination that resulted in equivalent expression levels of each of the four miRNAs of miR combo showed the highest reprogramming efficiency. Further efficiency can be achieved by directly targeting fibroblasts. Screening of several AAV serotypes indicated that AAV1 displayed tropism towards cardiac fibroblasts. Combining multicistronic expression with AAV1 delivery robustly reprogrammed cardiac fibroblasts into cardiomyocytes in vivo.


Subject(s)
Cellular Reprogramming Techniques/methods , Fibroblasts/cytology , MicroRNAs/genetics , Myocytes, Cardiac/cytology , Transfection/methods , Animals , Cells, Cultured , Cellular Reprogramming , Dependovirus/genetics , Fibroblasts/metabolism , Male , Mice, Inbred C57BL , Myocardial Infarction/therapy , Myocytes, Cardiac/metabolism , Plasmids/genetics
20.
Am J Physiol Renal Physiol ; 318(2): F285-F297, 2020 02 01.
Article in English | MEDLINE | ID: mdl-31760770

ABSTRACT

Juxtaglomerular (JG) cells, major sources of renin, differentiate from metanephric mesenchymal cells that give rise to JG cells or a subset of smooth muscle cells of the renal afferent arteriole. During periods of dehydration and salt deprivation, renal mesenchymal stromal cells (MSCs) differentiate from JG cells. JG cells undergo expansion and smooth muscle cells redifferentiate to express renin along the afferent arteriole. Gene expression profiling comparing resident renal MSCs with JG cells indicates that the transcription factor Sox6 is highly expressed in JG cells in the adult kidney. In vitro, loss of Sox6 expression reduces differentiation of renal MSCs to renin-producing cells. In vivo, Sox6 expression is upregulated after a low-Na+ diet and furosemide. Importantly, knockout of Sox6 in Ren1d+ cells halts the increase in renin-expressing cells normally seen during a low-Na+ diet and furosemide as well as the typical increase in renin. Furthermore, Sox6 ablation in renin-expressing cells halts the recruitment of smooth muscle cells along the afferent arteriole, which normally express renin under these conditions. These results support a previously undefined role for Sox6 in renin expression.


Subject(s)
Arterioles/metabolism , Juxtaglomerular Apparatus/blood supply , Mesenchymal Stem Cells/metabolism , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Renin/metabolism , SOXD Transcription Factors/metabolism , Animals , Arterioles/drug effects , Blood Pressure , Cell Differentiation , Cell Proliferation , Cells, Cultured , Diet, Sodium-Restricted , Diuretics/pharmacology , Furosemide/pharmacology , Gene Expression Regulation , Male , Mesenchymal Stem Cells/drug effects , Mice, Inbred C57BL , Mice, Knockout , Muscle, Smooth, Vascular/drug effects , Myocytes, Smooth Muscle/drug effects , Renin/genetics , SOXD Transcription Factors/deficiency , SOXD Transcription Factors/genetics , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...