Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 62
1.
J Extracell Vesicles ; 13(5): e12445, 2024 May.
Article En | MEDLINE | ID: mdl-38711334

Small extracellular vesicles (sEV) derived from various cell sources have been demonstrated to enhance cardiac function in preclinical models of myocardial infarction (MI). The aim of this study was to compare different sources of sEV for cardiac repair and determine the most effective one, which nowadays remains limited. We comprehensively assessed the efficacy of sEV obtained from human primary bone marrow mesenchymal stromal cells (BM-MSC), human immortalized MSC (hTERT-MSC), human embryonic stem cells (ESC), ESC-derived cardiac progenitor cells (CPC), human ESC-derived cardiomyocytes (CM), and human primary ventricular cardiac fibroblasts (VCF), in in vitro models of cardiac repair. ESC-derived sEV (ESC-sEV) exhibited the best pro-angiogenic and anti-fibrotic effects in vitro. Then, we evaluated the functionality of the sEV with the most promising performances in vitro, in a murine model of MI-reperfusion injury (IRI) and analysed their RNA and protein compositions. In vivo, ESC-sEV provided the most favourable outcome after MI by reducing adverse cardiac remodelling through down-regulating fibrosis and increasing angiogenesis. Furthermore, transcriptomic, and proteomic characterizations of sEV derived from hTERT-MSC, ESC, and CPC revealed factors in ESC-sEV that potentially drove the observed functions. In conclusion, ESC-sEV holds great promise as a cell-free treatment for promoting cardiac repair following MI.


Extracellular Vesicles , Mesenchymal Stem Cells , Myocardial Infarction , Myocytes, Cardiac , Extracellular Vesicles/metabolism , Extracellular Vesicles/transplantation , Humans , Animals , Mice , Myocardial Infarction/therapy , Myocardial Infarction/metabolism , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/cytology , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/cytology , Embryonic Stem Cells/metabolism , Embryonic Stem Cells/cytology , Human Embryonic Stem Cells/cytology , Human Embryonic Stem Cells/metabolism , Fibroblasts/metabolism , Male , Myocardial Reperfusion Injury/therapy , Myocardial Reperfusion Injury/metabolism , Disease Models, Animal , Neovascularization, Physiologic , Cells, Cultured
3.
Circulation ; 149(2): 135-154, 2024 01 09.
Article En | MEDLINE | ID: mdl-38084582

BACKGROUND: Endothelial cell (EC) generation and turnover by self-proliferation contributes to vascular repair and regeneration. The ability to accurately measure the dynamics of EC generation would advance our understanding of cellular mechanisms of vascular homeostasis and diseases. However, it is currently challenging to evaluate the dynamics of EC generation in large vessels such as arteries because of their infrequent proliferation. METHODS: By using dual recombination systems based on Cre-loxP and Dre-rox, we developed a genetic system for temporally seamless recording of EC proliferation in vivo. We combined genetic recording of EC proliferation with single-cell RNA sequencing and gene knockout to uncover cellular and molecular mechanisms underlying EC generation in arteries during homeostasis and disease. RESULTS: Genetic proliferation tracing reveals that ≈3% of aortic ECs undergo proliferation per month in adult mice during homeostasis. The orientation of aortic EC division is generally parallel to blood flow in the aorta, which is regulated by the mechanosensing protein Piezo1. Single-cell RNA sequencing analysis reveals 4 heterogeneous aortic EC subpopulations with distinct proliferative activity. EC cluster 1 exhibits transit-amplifying cell features with preferential proliferative capacity and enriched expression of stem cell markers such as Sca1 and Sox18. EC proliferation increases in hypertension but decreases in type 2 diabetes, coinciding with changes in the extent of EC cluster 1 proliferation. Combined gene knockout and proliferation tracing reveals that Hippo/vascular endothelial growth factor receptor 2 signaling pathways regulate EC proliferation in large vessels. CONCLUSIONS: Genetic proliferation tracing quantitatively delineates the dynamics of EC generation and turnover, as well as EC division orientation, in large vessels during homeostasis and disease. An EC subpopulation in the aorta exhibits more robust cell proliferation during homeostasis and type 2 diabetes, identifying it as a potential therapeutic target for vascular repair and regeneration.


Diabetes Mellitus, Type 2 , Vascular Endothelial Growth Factor A , Animals , Mice , Vascular Endothelial Growth Factor A/metabolism , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/metabolism , Aorta/metabolism , Endothelial Cells/metabolism , Homeostasis , Ion Channels/metabolism
4.
Org Lett ; 26(1): 416-420, 2024 Jan 12.
Article En | MEDLINE | ID: mdl-38160397

A reductive amidation of triazine esters with nitroarenes by using cheap iron as a reducing metal in the presence of TMSCl in DMF was developed. The reactions proceeded efficiently under transition metal-free conditions to give the corresponding amides in moderate to good yields with good functional group compatibility. Preliminary mechanistic investigations indicated that nitrosobenzene, N-phenyl hydroxylamine, azoxybenzene, azobenzene, aniline, and N-arylformamide possibly served as the intermediates of the reaction.

5.
Sci Rep ; 13(1): 15022, 2023 09 12.
Article En | MEDLINE | ID: mdl-37699967

The heart depends on a functional vasculature for oxygenation and transport of nutrients, and it is of interest to learn how primary impairment of the vasculature can indirectly affect cardiac function and heart morphology. Notch3-deficiency causes vascular smooth muscle cell (VSMC) loss in the vasculature but the consequences for the heart remain largely elusive. Here, we demonstrate that Notch3-/- mice have enlarged hearts with left ventricular hypertrophy and mild fibrosis. Cardiomyocytes were hypertrophic but not hyperproliferative, and the expression of several cardiomyocyte markers, including Tnt2, Myh6, Myh7 and Actn2, was altered. Furthermore, expression of genes regulating the metabolic status of the heart was affected: both Pdk4 and Cd36 were downregulated, indicating a metabolic switch from fatty acid oxidation to glucose consumption. Notch3-/- mice furthermore showed lower liver lipid content. Notch3 was expressed in heart VSMC and pericytes but not in cardiomyocytes, suggesting that a perturbation of Notch signalling in VSMC and pericytes indirectly impairs the cardiomyocytes. In keeping with this, Pdgfbret/ret mice, characterized by reduced numbers of VSMC and pericytes, showed left ventricular and cardiomyocyte hypertrophy. In conclusion, we demonstrate that reduced Notch3 or PDGFB signalling in vascular mural cells leads to cardiomyocyte dysfunction.


Cardiomegaly , Hypertrophy, Left Ventricular , Animals , Mice , Becaplermin , Lipid Metabolism , Myocytes, Cardiac , Proto-Oncogene Proteins c-sis
6.
NPJ Regen Med ; 8(1): 41, 2023 Aug 03.
Article En | MEDLINE | ID: mdl-37537178

A genetic system, ProTracer, has been recently developed to record cell proliferation in vivo. However, the ProTracer is initiated by an infrequently used recombinase Dre, which limits its broad application for functional studies employing floxed gene alleles. Here we generated Cre-activated functional ProTracer (fProTracer) mice, which enable simultaneous recording of cell proliferation and tissue-specific gene deletion, facilitating broad functional analysis of cell proliferation by any Cre driver.

7.
Comput Biol Med ; 164: 107357, 2023 09.
Article En | MEDLINE | ID: mdl-37586205

BACKGROUND: Lumbar/lumbosacral fusion supplemented with topping-off devices has been proposed with the aim of avoiding adjacent segment degeneration proximal to the fusion construct. However, it remains unclear how the biomechanics of the sacroiliac joint (SIJ) are altered after topping-off surgery. The objective of this study was to investigate the biomechanical effects of topping-off instrumentation on SIJ after lumbosacral fusion. METHODS: The validated finite element model of an intact lumbar spine-pelvis segment was modified to simulate L5-S1 interbody fusion fixed with a pedicle screw system. An interspinous spacer, Device for Intervertebral Assisted Motion (DIAM), was used as a topping-off device and placed between interspinous processes of the L4 and L5 segments. Range of motion (ROM), von-Mises stress distribution, and ligament strain at SIJ were compared between fusion (without DIAM) and topping-off (fusion with DIAM) models under moments of four physiological motions. RESULTS: ROM at the left and right SIJs in the topping-off model was higher by 26.9% and 27.5% in flexion, 16.8% and 16.1% in extension, 18.8% and 15.8% in lateral bending, and 3.7% and 7.4% in axial rotation, respectively, compared to those in the fusion model. The predicted stress and strain data showed that under all physiological loads, the topping-off model exhibited higher stress and ligament strain at the SIJs than the fusion model. CONCLUSIONS: Motion, stress, and ligament strain at SIJ increase when supplementing lumbosacral fusion with topping-off devices, suggesting that topping-off surgery may be associated with higher risks of SIJ degeneration and pain than fusion alone.


Sacroiliac Joint , Spinal Fusion , Sacroiliac Joint/surgery , Sacroiliac Joint/physiology , Biomechanical Phenomena/physiology , Lumbosacral Region , Pelvis , Lumbar Vertebrae/surgery , Lumbar Vertebrae/physiology , Range of Motion, Articular/physiology , Finite Element Analysis
8.
Kaohsiung J Med Sci ; 39(4): 354-363, 2023 Apr.
Article En | MEDLINE | ID: mdl-36919585

Laryngeal cancer is a usual malignant tumor of the head and neck. The role and mechanism of deubiquitinase USP21 in laryngeal cancer are still unclear. We aimed to explore whether USP21 affected laryngeal cancer progress through deubiquitinating AURKA. USP21 and AURKA levels were evaluated by qRT-PCR and Western blot. Kaplan-Meier analysis was conducted by survival package. MTT was performed to detect cell proliferation. The wound healing assay was applied to evaluate cell migration. Transwell was used to measure cell invasion. Co-IP and GST-pull down determined the interaction between USP21 and AURKA. In addition, AURKA ubiquitination levels were analyzed. USP21 was signally elevated in laryngeal cancer tissues and cells. USP21 level in clinical stages III-IV was higher than that in clinical stages I-II, and high levels of USP21 were highly correlated with poor prognosis in laryngeal cancer. USP21 inhibition suppressed AMC-HN-8 and TU686 cell proliferation, migration and invasion. Co-IP and GST-pull down confirmed the interaction between USP21 and AURKA. Knockdown of USP21 markedly increased the ubiquitination level of AURKA, and USP21 restored AURKA activity through deubiquitination. In addition, overexpression of AURKA reversed the effects of USP21 knockdown on cell growth, migration, and invasion. USP21 stabilized AURKA through deubiquitination to promote laryngeal cancer progression.


Laryngeal Neoplasms , Humans , Laryngeal Neoplasms/genetics , Laryngeal Neoplasms/pathology , Aurora Kinase A/genetics , Cell Proliferation/genetics , Cell Line, Tumor , Ubiquitination , Ubiquitin Thiolesterase/genetics , Ubiquitin Thiolesterase/metabolism
9.
Nat Genet ; 55(4): 665-678, 2023 04.
Article En | MEDLINE | ID: mdl-36959363

After severe heart injury, fibroblasts are activated and proliferate excessively to form scarring, leading to decreased cardiac function and eventually heart failure. It is unknown, however, whether cardiac fibroblasts are heterogeneous with respect to their degree of activation, proliferation and function during cardiac fibrosis. Here, using dual recombinase-mediated genetic lineage tracing, we find that endocardium-derived fibroblasts preferentially proliferate and expand in response to pressure overload. Fibroblast-specific proliferation tracing revealed highly regional expansion of activated fibroblasts after injury, whose pattern mirrors that of endocardium-derived fibroblast distribution in the heart. Specific ablation of endocardium-derived fibroblasts alleviates cardiac fibrosis and reduces the decline of heart function after pressure overload injury. Mechanistically, Wnt signaling promotes activation and expansion of endocardium-derived fibroblasts during cardiac remodeling. Our study identifies endocardium-derived fibroblasts as a key fibroblast subpopulation accounting for severe cardiac fibrosis after pressure overload injury and as a potential therapeutic target against cardiac fibrosis.


Heart Diseases , Fibroblasts/metabolism , Heart Diseases/genetics , Heart Diseases/pathology , Fibrosis/genetics , Animals , Mice , Aging , Cell Proliferation , Wnt Signaling Pathway , Mice, Transgenic
10.
J Biomech Eng ; 145(6)2023 06 01.
Article En | MEDLINE | ID: mdl-36695754

Rigid interspinous process fixation (RIPF) has been recently discussed as an alternative to pedicle screw fixation (PSF) for reducing trauma in lumbar interbody fusion (LIF) surgery. This study aimed to investigate biomechanics of the lumbar spine with RIPF, and also to compare biomechanical differences between two postoperative stages (before and after bony fusion). Based on an intact finite-element model of lumbosacral spine, the models of single-level LIF with RIPF or conventional PSF were developed and were computed for biomechanical responses to the moments of four physiological motions using hybrid testing protocol. It was found that compared with PSF, range of motion (ROM), intradiscal pressure (IDP), and facet joint forces (FJF) at adjacent segments of the surgical level for RIPF were decreased by up to 8.4%, 2.3%, and 16.8%, respectively, but ROM and endplate stress at the surgical segment were increased by up to 285.3% and 174.3%, respectively. The results of comparison between lumbar spine with RIPF before and after bony fusion showed that ROM and endplate stress at the surgical segment were decreased by up to 62.6% and 40.4%, respectively, when achieved to bony fusion. These findings suggest that lumbar spine with RIPF as compared to PSF has potential to decrease the risk of adjacent segment degeneration but might have lower stability of surgical segment and an increased risk of cage subsidence; When achieved bony fusion, it might be helpful for the lumbar spine with RIPF in increasing stability of surgical segment and reducing failure of bone contact with cage.


Pedicle Screws , Spinal Fusion , Spinal Fusion/adverse effects , Spinal Fusion/methods , Pedicle Screws/adverse effects , Range of Motion, Articular/physiology , Lumbar Vertebrae/surgery , Lumbar Vertebrae/physiology , Biomechanical Phenomena , Finite Element Analysis
11.
Pathol Int ; 73(3): 109-119, 2023 Mar.
Article En | MEDLINE | ID: mdl-36285444

Laryngeal cancer (LC) is a rare and challenging clinical problem. Our aim was to investigate the mechanism of salt-like transcription factor 4 (SALL4) in LC. LC tissue and paracancerous tissue were collected. Relative mRNA or protein levels were measured by quantitative real-time polymerase chain reaction or Western blot. MTT, wound healing, and transwell assay were performed to evaluate cell proliferation, migration and invasion. The binding relationship between SALL4 and USP21 promoter was verified by dual-luciferase assay and ChIP. Co-IP and glutathione-S-transferase (GST)-pull down were performed to measure the protein interaction between USP21 and YY1. Additionally, YY1 ubiquitination level was analyzed. It was found that SALL4 mRNA and SALL4 protein levels were elevated in LC clinical tissues and various LC cells. Knockdown of SALL4 inhibited epithelial-mesenchymal transition (EMT) of LC cells. USP21 was transcriptionally activated by SALL4. Co-IP and GST-pull down confirmed USP21 interacted with YY1. USP21 protected YY1 from degradation through deubiquitination. Furthermore, overexpression of USP21 reversed the effect of knockdown of SALL4 on YY1 and EMT in LC cells. In general, SALL4 facilitated EMT of LC cells through modulating USP21/YY1 axis.


Laryngeal Neoplasms , Transcription Factors , Humans , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Gene Expression Regulation, Neoplastic , Laryngeal Neoplasms/genetics , RNA, Messenger , Transcription Factor 4/genetics , Transcription Factor 4/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Transcriptional Activation , Ubiquitin Thiolesterase/genetics , Ubiquitin-Specific Proteases/genetics , Ubiquitin-Specific Proteases/metabolism , Yin-Yang
12.
J Mater Sci Mater Med ; 33(6): 52, 2022 Jun 03.
Article En | MEDLINE | ID: mdl-35657438

Under whole body vibration, how the cement augmentation affects the vibration characteristic of the osteoporotic fusion lumbar spine, complications, and fusion outcomes is unclear. A L1-L5 lumbar spine finite element model was developed to simulate a transforaminal lumbar interbody fusion (TLIF) model with bilateral pedicle screws at L4-L5 level, a polymethylmethacrylate (PMMA) cement-augmented TLIF model (TLIF-PMMA) and an osteoporotic TLIF model. A 40 N sinusoidal vertical load at 5 Hz and a 400 N preload were utilized to simulate a vertical vibration of the human body and the physiological compression caused by muscle contraction and the weight of human body. The results showed that PMMA cement augmentation may produce a stiffer pedicle screw/rod construct and decrease the risk of adjacent segment disease, subsidence, and rod failure under whole-body vibration(WBV). Cement augmentation might restore the disc height and segmental lordosis and decrease the risk of poor outcomes, but it might also increase the risk of cage failure and prolong the period of lumbar fusion under WBV. The findings may provide new insights for performing lumbar interbody fusion in patients affected by osteoporosis of the lumbar spine. Graphical abstract.


Lumbar Vertebrae , Spinal Fusion , Biomechanical Phenomena , Finite Element Analysis , Humans , Lumbar Vertebrae/surgery , Polymethyl Methacrylate , Spinal Fusion/methods , Vibration/therapeutic use
13.
Org Lett ; 24(27): 4919-4924, 2022 07 15.
Article En | MEDLINE | ID: mdl-35771670

An efficient Sonogashira coupling of a heterocyclic phosphonium salt with a terminal alkyne via C-P bond cleavage was developed. The reactions proceeded smoothly in the presence of palladium catalyst, copper(I) iodide, and N,N-diisopropylethylamine (DIPEA) in N-methyl-2-pyrrolidone (NMP) at 100 °C for 12 h, producing the corresponding alkynyl-substituted pyridine, quinoline, pyrazine, and quinoxaline in moderate to good yields with wide substrate scope and broad functional group tolerance. In addition, gram-scale synthesis could also be achieved, and the reaction could be applied to the functionalization of alkyne-containing complex molecules derived from sugars and pharmaceutical and naturally occurring products (e.g., estrone, d-galactopyranose, menthol, and ibuprofen).


Alkynes , Palladium , Alkynes/chemistry , Catalysis , Copper , Palladium/chemistry
14.
Bioorg Med Chem ; 65: 116782, 2022 07 01.
Article En | MEDLINE | ID: mdl-35512484

Achieving pharmacological control over cardiomyocyte proliferation represents a prime goal in therapeutic cardiovascular research. Here, we identify a novel chemical tool compound for the expansion of human induced pluripotent stem cell (hiPSC)-derived cardiomyocytes. The forkhead box O (FOXO) inhibitor AS1842856 was identified as a significant hit from an unbiased proliferation screen in early, immature hiPSC- cardiomyocytes (eCMs). The mitogenic effects of AS1842856 turned out to be robust, dose-dependent, sustained, and reversible. eCM numbers increased >30-fold as induced by AS1842856 over three passages. Phenotypically as well as by marker gene expression, the compound interestingly appeared to counteract cellular maturation both in immature hiPSC-CMs as well as in more advanced ones. Thus, FOXO inhibitor AS1842856 presents a novel proliferation inducer for the chemically defined, xeno-free expansion of hiPSC-derived CMs, while its de-differentiation effect might as well bear potential in regenerative medicine.


Induced Pluripotent Stem Cells , Cell Differentiation , Cell Proliferation , Cells, Cultured , Extracellular Matrix , Humans , Induced Pluripotent Stem Cells/metabolism , Myocytes, Cardiac
15.
Nat Cell Biol ; 24(5): 659-671, 2022 05.
Article En | MEDLINE | ID: mdl-35550611

Heart regeneration is an unmet clinical need, hampered by limited renewal of adult cardiomyocytes and fibrotic scarring. Pluripotent stem cell-based strategies are emerging, but unravelling cellular dynamics of host-graft crosstalk remains elusive. Here, by combining lineage tracing and single-cell transcriptomics in injured non-human primate heart biomimics, we uncover the coordinated action modes of human progenitor-mediated muscle repair. Chemoattraction via CXCL12/CXCR4 directs cellular migration to injury sites. Activated fibroblast repulsion targets fibrosis by SLIT2/ROBO1 guidance in organizing cytoskeletal dynamics. Ultimately, differentiation and electromechanical integration lead to functional restoration of damaged heart muscle. In vivo transplantation into acutely and chronically injured porcine hearts illustrated CXCR4-dependent homing, de novo formation of heart muscle, scar-volume reduction and prevention of heart failure progression. Concurrent endothelial differentiation contributed to graft neovascularization. Our study demonstrates that inherent developmental programmes within cardiac progenitors are sequentially activated in disease, enabling the cells to sense and counteract acute and chronic injury.


Nerve Tissue Proteins , Pluripotent Stem Cells , Animals , Cell Differentiation , Cicatrix/pathology , Cicatrix/prevention & control , Fibrosis , Humans , Myocardium/pathology , Myocytes, Cardiac/pathology , Pluripotent Stem Cells/pathology , Receptors, Immunologic , Swine
16.
Circulation ; 144(25): 2004-2020, 2021 12 21.
Article En | MEDLINE | ID: mdl-34797683

BACKGROUND: Cardiac fibrosis is a lethal outcome of excessive formation of myofibroblasts that are scar-forming cells accumulated after heart injury. It has been reported that cardiac endothelial cells (ECs) contribute to a substantial portion of myofibroblasts through endothelial to mesenchymal transition (EndoMT). Recent lineage tracing studies demonstrate that myofibroblasts are derived from the expansion of resident fibroblasts rather than from the transdifferentiation of ECs. However, it remains unknown whether ECs can transdifferentiate into myofibroblasts reversibly or EndoMT genes were just transiently activated in ECs during cardiac fibrosis. METHODS: By using the dual recombination technology based on Cre-loxP and Dre-rox, we generated a genetic lineage tracing system for tracking EndoMT in cardiac ECs. We used it to examine if there is transiently activated mesenchymal gene expression in ECs during cardiac fibrosis. Activation of the broadly used marker gene in myofibroblasts, αSMA (α-smooth muscle actin), and the transcription factor that induces epithelial to mesenchymal transition, Zeb1 (zinc finger E-box-binding homeobox 1), was examined. RESULTS: The genetic system enables continuous tracing of transcriptional activity of targeted genes in vivo. Our genetic fate mapping results revealed that a subset of cardiac ECs transiently expressed αSMA and Zeb1 during embryonic valve formation and transdifferentiated into mesenchymal cells through EndoMT. Nonetheless, they did not contribute to myofibroblasts, nor transiently expressed αSMA or Zeb1 after heart injury. Instead, expression of αSMA was activated in resident fibroblasts during cardiac fibrosis. CONCLUSIONS: Mesenchymal gene expression is activated in cardiac ECs through EndoMT in the developing heart, but ECs do not transdifferentiate into myofibroblasts, nor transiently express some known mesenchymal genes during homeostasis and fibrosis in the adult heart. Resident fibroblasts that are converted to myofibroblasts by activating mesenchymal gene expression are the major contributors to cardiac fibrosis.


Endothelial Cells/metabolism , Fibrosis/genetics , Gene Expression/genetics , Myofibroblasts/metabolism , Animals , Female , Humans , Male , Mice
17.
Front Cardiovasc Med ; 8: 753470, 2021.
Article En | MEDLINE | ID: mdl-34722683

Objective: After myocardial infarction (MI), the non-infarcted left ventricle (LV) ensures appropriate contractile function of the heart. Metabolic disturbance in this region greatly exacerbates post-MI heart failure (HF) pathology. This study aimed to provide a comprehensive understanding of the metabolic derangements occurring in the non-infarcted LV that could trigger cardiovascular deterioration. Methods and Results: We used a pig model that progressed into chronic HF over 3 months following MI induction. Integrated gene and metabolite signatures revealed region-specific perturbations in amino acid- and lipid metabolism, insulin signaling and, oxidative stress response. Remote LV, in particular, showed impaired glutamine and arginine metabolism, altered synthesis of lipids, glucose metabolism disorder, and increased insulin resistance. LPIN1, PPP1R3C, PTPN1, CREM, and NR0B2 were identified as the main effectors in metabolism dysregulation in the remote zone and were found differentially expressed also in the myocardium of patients with ischemic and/or dilated cardiomyopathy. In addition, a simultaneous significant decrease in arginine levels and altered PRCP, PTPN1, and ARF6 expression suggest alterations in vascular function in remote area. Conclusions: This study unravels an array of dysregulated genes and metabolites putatively involved in maladaptive metabolic and vascular remodeling in the non-infarcted myocardium and may contribute to the development of more precise therapies to mitigate progression of chronic HF post-MI.

18.
Nat Commun ; 12(1): 5784, 2021 10 01.
Article En | MEDLINE | ID: mdl-34599161

Cardiac regeneration involves the generation of new cardiomyocytes from cycling cardiomyocytes. Understanding cell-cycle activity of pre-existing cardiomyocytes provides valuable information to heart repair and regeneration. However, the anatomical locations and in situ dynamics of cycling cardiomyocytes remain unclear. Here we develop a genetic approach for a temporally seamless recording of cardiomyocyte-specific cell-cycle activity in vivo. We find that the majority of cycling cardiomyocytes are positioned in the subendocardial muscle of the left ventricle, especially in the papillary muscles. Clonal analysis revealed that a subset of cycling cardiomyocytes have undergone cell division. Myocardial infarction and cardiac pressure overload induce regional patterns of cycling cardiomyocytes. Mechanistically, cardiomyocyte cell cycle activity requires the Hippo pathway effector YAP. These genetic fate-mapping studies advance our basic understanding of cardiomyocyte cell cycle activity and generation in cardiac homeostasis, repair, and regeneration.


Myocytes, Cardiac/cytology , Animals , Blotting, Southern , Cell Cycle/physiology , Cell Proliferation/physiology , Female , Flow Cytometry , Heart/physiology , Male , Mice , Microscopy, Fluorescence , Myocardium/metabolism , Myocardium/pathology , Myocytes, Cardiac/metabolism
19.
Nat Commun ; 12(1): 5180, 2021 08 30.
Article En | MEDLINE | ID: mdl-34462437

Heart failure (HF) is a major cause of morbidity and mortality worldwide, highlighting an urgent need for novel treatment options, despite recent improvements. Aberrant Ca2+ handling is a key feature of HF pathophysiology. Restoring the Ca2+ regulating machinery is an attractive therapeutic strategy supported by genetic and pharmacological proof of concept studies. Here, we study antisense oligonucleotides (ASOs) as a therapeutic modality, interfering with the PLN/SERCA2a interaction by targeting Pln mRNA for downregulation in the heart of murine HF models. Mice harboring the PLN R14del pathogenic variant recapitulate the human dilated cardiomyopathy (DCM) phenotype; subcutaneous administration of PLN-ASO prevents PLN protein aggregation, cardiac dysfunction, and leads to a 3-fold increase in survival rate. In another genetic DCM mouse model, unrelated to PLN (Cspr3/Mlp-/-), PLN-ASO also reverses the HF phenotype. Finally, in rats with myocardial infarction, PLN-ASO treatment prevents progression of left ventricular dilatation and improves left ventricular contractility. Thus, our data establish that antisense inhibition of PLN is an effective strategy in preclinical models of genetic cardiomyopathy as well as ischemia driven HF.


Calcium-Binding Proteins/genetics , Cardiomyopathies/genetics , Cardiomyopathies/therapy , Genetic Therapy , Heart Failure/genetics , Heart Failure/therapy , Oligonucleotides, Antisense/genetics , Animals , Calcium/metabolism , Calcium-Binding Proteins/metabolism , Cardiomyopathies/metabolism , Female , Heart Failure/metabolism , Humans , Male , Mice , Mice, Inbred C57BL , Myocytes, Cardiac/metabolism , Oligonucleotides, Antisense/metabolism , Rats , Rats, Inbred Lew
20.
J Org Chem ; 86(12): 8236-8247, 2021 06 18.
Article En | MEDLINE | ID: mdl-34061530

An unprecedented and expeditious tandem bisannulation of polyfluoroalkylated tetralones with benzamidines to access various fluoroalkyl tetracyclic [1,3]-diazepines through multiple C-N bond formation and C(sp3)-F bond cleavage is reported. The process features high regio-/chemoselectivities, broad substrate scope, good functional group tolerance, procedural simplicity, mild reaction conditions, and scale-up synthesis. Mechanistic studies showed that the distinctive fluorine effect of polyfluoroalkyl tetralone plays a vital role for the aza-tetracycle construction.


Fluorine
...