Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 78
Filter
1.
Circ Genom Precis Med ; 17(2): e004397, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38563135

ABSTRACT

BACKGROUND: Basic scientists have used preclinical animal models to explore mechanisms driving human diseases for decades, resulting in thousands of publications, each supporting causative inferences. Despite substantial advances in the mechanistic construct of disease, there has been limited translation from individual studies to advances in clinical care. An integrated approach to these individual studies has the potential to improve translational success. METHODS: Using atherosclerosis as a test case, we extracted data from the 2 most common mouse models of atherosclerosis (ApoE [apolipoprotein E]-knockout and LDLR [low-density lipoprotein receptor]-knockout). We restricted analyses to manuscripts published in 2 well-established journals, Arteriosclerosis, Thrombosis, and Vascular Biology and Circulation, as of query in 2021. Predefined variables including experimental conditions, intervention, and outcomes were extracted from each publication to produce a preclinical atherosclerosis database. RESULTS: Extracted data include animal sex, diet, intervention type, and distinct plaque pathologies (size, inflammation, and lipid content). Procedures are provided to standardize data extraction, attribute interventions to specific genes, and transform the database for use with available transcriptomics software. The database integrates hundreds of genes, each directly tested in vivo for causation in a murine atherosclerosis model. The database is provided to allow the research community to perform integrated analyses that reflect the global impact of decades of atherosclerosis investigation. CONCLUSIONS: This database is provided as a resource for future interrogation of sub-data sets associated with distinct plaque pathologies, cell type, or sex. We also provide the methods and software needed to expand this data set and apply this approach to the extensive repository of peer-reviewed data utilizing preclinical models to interrogate mechanisms of diverse human diseases.


Subject(s)
Atherosclerosis , Plaque, Atherosclerotic , Mice , Humans , Animals , Atherosclerosis/pathology
2.
J Tradit Chin Med ; 44(2): 289-302, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38504535

ABSTRACT

OBJECTIVE: To discuss the influence of Sailuotong (, SLT) on the Neurovascular Unit (NVUs) of amyloid precursor protein (APP)/presenilin-1(PS1) mice and evaluate the role of gas supplementation in activating blood circulation during the progression of Alzheimer's disease (AD). METHODS: The mice were allocated into the following nine groups: (a) the C57 Black (C57BL) sham-operated group (control group), (b) ischaemic treatment in C57BL mice (the C57 ischaemic group), (c) the APP/PS1 sham surgery group (APP/PS1 model group), (d) ischaemic treatment in APP/PS1 mice (APP/PS1 ischaemic group), (e) C57BL mice treated with aspirin following ischaemic treatment (C57BL ischaemic + aspirin group), (f) C57BL mice treated with SLT following ischaemic treatment (C57BL ischaemic + SLT group), (g) APP/PS1 mice treated with SLT (APP/PS1 + SLT group), (h) APP/PS1 mice treated with donepezil hydrochloride following ischaemic treatment (APP/PS1 ischaemic + donepezil hydrochloride group) and (i) APP/PS1 mice treated with SLT following ischaemic treatment (APP/PS1 ischaemic + SLT group). The ischaemic model was established by operating on the bilateral common carotid arteries and creating a microembolism. The Morris water maze and step-down tests were used to detect the spatial behaviour and memory ability of mice. The hippocampus of each mouse was observed by haematoxylin and eosin (HE) and Congo red staining. The ultrastructure of NVUs in each group was observed by electron microscopy, and various biochemical indicators were detected by enzyme-linked immunosorbent assay (ELISA). The protein expression level was detected by Western blot. The mRNA expression was detected by quantitative real-time polymerase chain reaction (qRT-PCR). RESULTS: The results of the Morris water maze and step-down tests showed that ischemia reduced learning and memory in the mice, which were restored by SLT. The results of HE staining showed that SLT restored the pathological changes of the NVUs. The Congo red staining results revealed that SLT also improved the scattered orange-red sediments in the upper cortex and hippocampus of the APP/PS1 and APP/PS1 ischaemic mice. Furthermore, SLT significantly reduced the content of Aß, improved the vascular endothelium and repaired the mitochondrial structures. The ELISA detection, western blot detection and qRT-PCR showed that SLT significantly increased the vascular endothelial growth factor (VEGF), angiopoietin and basic fibroblast growth factor, as well as the levels of gene and protein expression of low-density lipoprotein receptor-related protein-1 (LRP-1) and VEGF in brain tissue. CONCLUSIONS: By increasing the expression of VEGF, SLT can promote vascular proliferation, up-regulate the expression of LRP-1, promote the clearance of Aß and improve the cognitive impairment of APP/PS1 mice. These results confirm that SLT can improve AD by promoting vascular proliferation and Aß clearance to protect the function of NVUs.


Subject(s)
Alzheimer Disease , Amyloid beta-Protein Precursor , Drugs, Chinese Herbal , Mice , Animals , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Alzheimer Disease/drug therapy , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Mice, Transgenic , Vascular Endothelial Growth Factor A , Donepezil , Amyloid beta-Peptides/genetics , Amyloid beta-Peptides/metabolism , Presenilin-1/genetics , Presenilin-1/metabolism , Congo Red , Mice, Inbred C57BL , Aspirin , Disease Models, Animal
3.
Circulation ; 148(1): 47-67, 2023 07 04.
Article in English | MEDLINE | ID: mdl-37199168

ABSTRACT

BACKGROUND: Activation of vascular smooth muscle cell (VSMC) inflammation is vital to initiate vascular disease. The role of human-specific long noncoding RNAs in VSMC inflammation is poorly understood. METHODS: Bulk RNA sequencing in differentiated human VSMCs revealed a novel human-specific long noncoding RNA called inflammatory MKL1 (megakaryoblastic leukemia 1) interacting long noncoding RNA (INKILN). INKILN expression was assessed in multiple in vitro and ex vivo models of VSMC phenotypic modulation as well as human atherosclerosis and abdominal aortic aneurysm. The transcriptional regulation of INKILN was verified through luciferase reporter and chromatin immunoprecipitation assays. Loss-of-function and gain-of-function studies and multiple RNA-protein and protein-protein interaction assays were used to uncover a mechanistic role of INKILN in the VSMC proinflammatory gene program. Bacterial artificial chromosome transgenic mice were used to study INKILN expression and function in ligation injury-induced neointimal formation. RESULTS: INKILN expression is downregulated in contractile VSMCs and induced in human atherosclerosis and abdominal aortic aneurysm. INKILN is transcriptionally activated by the p65 pathway, partially through a predicted NF-κB (nuclear factor kappa B) site within its proximal promoter. INKILN activates proinflammatory gene expression in cultured human VSMCs and ex vivo cultured vessels. INKILN physically interacts with and stabilizes MKL1, a key activator of VSMC inflammation through the p65/NF-κB pathway. INKILN depletion blocks interleukin-1ß-induced nuclear localization of both p65 and MKL1. Knockdown of INKILN abolishes the physical interaction between p65 and MKL1 and the luciferase activity of an NF-κB reporter. Furthermore, INKILN knockdown enhances MKL1 ubiquitination through reduced physical interaction with the deubiquitinating enzyme USP10 (ubiquitin-specific peptidase 10). INKILN is induced in injured carotid arteries and exacerbates ligation injury-induced neointimal formation in bacterial artificial chromosome transgenic mice. CONCLUSIONS: These findings elucidate an important pathway of VSMC inflammation involving an INKILN/MKL1/USP10 regulatory axis. Human bacterial artificial chromosome transgenic mice offer a novel and physiologically relevant approach for investigating human-specific long noncoding RNAs under vascular disease conditions.


Subject(s)
Aortic Aneurysm, Abdominal , RNA, Long Noncoding , Animals , Humans , Mice , Aortic Aneurysm, Abdominal/metabolism , Cell Proliferation , Cells, Cultured , Inflammation/genetics , Inflammation/metabolism , Luciferases/metabolism , Mice, Transgenic , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , NF-kappa B/metabolism , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Ubiquitin Thiolesterase/metabolism
5.
Arterioscler Thromb Vasc Biol ; 43(2): 203-211, 2023 02.
Article in English | MEDLINE | ID: mdl-36519470

ABSTRACT

BACKGROUND: The Myh11 promoter is extensively used as a smooth muscle cell (SMC) Cre-driver and is regarded as the most restrictive and specific promoter available to study SMCs. Unfortunately, in the existing Myh11-CreERT2 mouse, the transgene was inserted on the Y chromosome precluding the study of female mice. Given the importance of including sex as a biological variable and that numerous SMC-based diseases have a sex-dependent bias, the field has been tremendously limited by the lack of a model to study both sexes. Here, we describe a new autosomal Myh11-CreERT2 mouse (referred to as Myh11-CreERT2-RAD), which allows for SMC-specific lineage tracing and gene knockout studies in vivo using both male and female mice. METHODS: A Myh11-CreERT2-RAD transgenic C57BL/6 mouse line was generated using bacterial artificial chromosome clone RP23-151J22 modified to contain a Cre-ERT2 after the Myh11 start codon. Myh11-CreERT2-RAD mice were crossed with 2 different fluorescent reporter mice and tested for SMC-specific labeling by flow cytometric and immunofluorescence analyses. RESULTS: Myh11-CreERT2-RAD transgene insertion was determined to be on mouse chromosome 2. Myh11-CreERT2-RAD fluorescent reporter mice showed Cre-dependent, tamoxifen-inducible labeling of SMCs equivalent to the widely used Myh11-CreERT2 mice. Labeling was equivalent in both male and female Cre+ mice and was limited to vascular and visceral SMCs and pericytes in various tissues as assessed by immunofluorescence. CONCLUSIONS: We generated and validated the function of an autosomal Myh11-CreERT2-RAD mouse that can be used to assess sex as a biological variable with respect to the normal and pathophysiological functions of SMCs.


Subject(s)
Integrases , Myocytes, Smooth Muscle , Mice , Animals , Male , Female , Mice, Transgenic , Gene Knockout Techniques , Integrases/genetics , Integrases/metabolism , Mice, Knockout , Mice, Inbred C57BL , Myocytes, Smooth Muscle/metabolism , Cell Lineage , Tamoxifen
6.
Arterioscler Thromb Vasc Biol ; 42(11): 1378-1397, 2022 11.
Article in English | MEDLINE | ID: mdl-36172865

ABSTRACT

BACKGROUND: Migration of human aortic smooth muscle cells (HASMCs) contributes to the pathogenesis of atherosclerosis. This study aims to functionally characterize long noncoding RNA TPRG1-AS1 (tumor protein p63 regulated 1, antisense 1) in HASMCs and reveal the underlying mechanism of TPRG1-AS1 in HASMCs migration, neointima formation, and subsequent atherosclerosis. METHODS: The expression of TPRG1-AS1 in atherosclerotic plaques was verified a series of in silico analysis and quantitative real-time polymerase chain reaction analysis. Northern blot, rapid amplification of cDNA ends and Sanger sequencing were used to determine its full length. In vitro transcription-translation assay was used to investigate the protein-coding capacity of TPRG1-AS1. RNA fluorescent in situ hybridization was used to confirm its subcellular localization. Loss- and gain-of-function studies were used to investigate the function of TPRG1-AS1. Furthermore, the effect of TPRG1-AS1 on the pathological response was evaluated in carotid balloon injury model, wire injury model, and atherosclerosis model, respectively. RESULTS: TPRG1-AS1 was significantly increased in atherosclerotic plaques. TPRG1-AS1 did not encode any proteins and its full length was 1279nt, which was bona fide a long noncoding RNA. TPRG1-AS1 was mainly localized in cytoplasmic and perinuclear regions in HASMCs. TPRG1-AS1 directly interacted with MYH9 (myosin heavy chain 9) protein in HASMCs, promoted MYH9 protein degradation through the proteasome pathway, hindered F-actin stress fiber formation, and finally inhibited HASMCs migration. Vascular smooth muscle cell-specific transgenic overexpression of TPRG1-AS1 significantly reduced neointima formation, and attenuated atherosclerosis in apolipoprotein E knockout (Apoe-/-) mice. CONCLUSIONS: This study demonstrated that TPRG1-AS1 inhibited HASMCs migration through interacting with MYH9 protein and consequently suppressed neointima formation and atherosclerosis.


Subject(s)
Atherosclerosis , MicroRNAs , Plaque, Atherosclerotic , RNA, Long Noncoding , Humans , Mice , Animals , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Muscle, Smooth, Vascular/metabolism , Neointima/metabolism , Plaque, Atherosclerotic/metabolism , Actins/metabolism , Proteasome Endopeptidase Complex/metabolism , DNA, Complementary/metabolism , DNA, Complementary/pharmacology , In Situ Hybridization, Fluorescence , Myosin Heavy Chains/genetics , Myosin Heavy Chains/metabolism , Cell Proliferation , Myocytes, Smooth Muscle/metabolism , Cell Movement , Atherosclerosis/genetics , Atherosclerosis/prevention & control , Atherosclerosis/metabolism , MicroRNAs/genetics , Cytoskeletal Proteins/metabolism , Apolipoproteins E/genetics , Apolipoproteins/metabolism
7.
Circulation ; 144(23): 1876-1890, 2021 12 07.
Article in English | MEDLINE | ID: mdl-34672678

ABSTRACT

BACKGROUND: The catalytic subunit of telomerase, telomerase reverse transcriptase (TERT), has protective functions in the cardiovascular system. TERT is not only present in the nucleus but also in mitochondria. However, it is unclear whether nuclear or mitochondrial TERT is responsible for the observed protection, and the appropriate tools are missing to dissect this. METHODS: We generated new mouse models containing TERT exclusively in the mitochondria (mitoTERT mice) or the nucleus (nucTERT mice) to finally distinguish between the functions of nuclear and mitochondrial TERT. Outcome after ischemia/reperfusion, mitochondrial respiration in the heart, and cellular functions of cardiomyocytes, fibroblasts, and endothelial cells, as well, were determined. RESULTS: All mice were phenotypically normal. Although respiration was reduced in cardiac mitochondria from TERT-deficient and nucTERT mice, it was increased in mitoTERT animals. The latter also had smaller infarcts than wild-type mice, whereas nucTERT animals had larger infarcts. The decrease in ejection fraction after 1, 2, and 4 weeks of reperfusion was attenuated in mitoTERT mice. Scar size was also reduced and vascularization increased. Mitochondrial TERT protected a cardiomyocyte cell line from apoptosis. Myofibroblast differentiation, which depends on complex I activity, was abrogated in TERT-deficient and nucTERT cardiac fibroblasts and completely restored in mitoTERT cells. In endothelial cells, mitochondrial TERT enhanced migratory capacity and activation of endothelial nitric oxide synthase. Mechanistically, mitochondrial TERT improved the ratio between complex I matrix arm and membrane subunits, explaining the enhanced complex I activity. In human right atrial appendages, TERT was localized in mitochondria and there increased by remote ischemic preconditioning. The telomerase activator TA-65 evoked a similar effect in endothelial cells, thereby increasing their migratory capacity, and enhanced myofibroblast differentiation. CONCLUSIONS: Mitochondrial, but not nuclear TERT, is critical for mitochondrial respiration and during ischemia/reperfusion injury. Mitochondrial TERT improves complex I subunit composition. TERT is present in human heart mitochondria, and remote ischemic preconditioning increases its level in those organelles. TA-65 has comparable effects ex vivo and improves the migratory capacity of endothelial cells and myofibroblast differentiation. We conclude that mitochondrial TERT is responsible for cardioprotection, and its increase could serve as a therapeutic strategy.


Subject(s)
Electron Transport Complex I/metabolism , Mitochondria, Heart/enzymology , Mitochondrial Proteins/metabolism , Myocardial Reperfusion Injury/enzymology , Telomerase/metabolism , Animals , Electron Transport Complex I/genetics , Female , Humans , Male , Mice , Mice, Transgenic , Mitochondria, Heart/genetics , Mitochondrial Proteins/genetics , Myocardial Reperfusion Injury/genetics , Telomerase/genetics
8.
J Breast Cancer ; 24(5): 455-462, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34652081

ABSTRACT

PURPOSE: The breast cancer susceptibility gene, BRCA1, is involved in normal development and carcinogenesis of mammary glands. Here, we aimed to evaluate the relationship between histological findings of mammary gland development and breast cancer risk in BRCA1 mutant mice. METHODS: Five BRCA1 mutant mice and five non-mutant FVB/NJ mice were used for each group of 1-month-old (pubertal), 3-month-old (fertile), and 8-month-old (menopausal) mice. In another experiment, 15 BRCA1 mutant mice were followed up to 8 months after birth and classified into tumor-bearing (11 mice) and tumor-free (4 mice) groups. Excised mammary gland tissues were stained with Carmine Alum, and the number of terminal end buds (or alveolar buds), branching density, and duct elongation were measured using image analysis programs. Differences between the two groups were assessed using paired t-test. RESULTS: One-month-old BRCA1 mutant mice showed a higher number of terminal end buds (23.8 ± 1.0 vs. 15.6 ± 0.8, p = 0.0002), branching density (11.7 ± 0.4 vs. 9.6 ± 0.5%, p = 0.0082), and duct elongation (9.7 ± 0.7 vs. 7.3 ± 0.4 mm, p = 0.0186) than controls. However, there was no difference between the 3- and 8-month-old groups. In BRCA1 mutant mice, the tumor-bearing group showed a significantly higher number of alveolar buds (142.7 ± 5.5 vs. 105.5 ± 5.4, p = 0.0008) and branching density (30.0 ± 1.0 vs. 24.1 ± 1.1%, p = 0.008) than the tumor-free group; however, duct elongation was not different (23.9 ± 0.6 vs. 23.6 ± 0.6 mm, p = 0.8099) between the groups. CONCLUSION: BRCA1 mutant mice exhibited early pubertal mammary gland development and delayed age-related mammary gland involution was associated with breast cancer. Our results may have clinical implications for predicting breast cancer risk and developing prevention strategies for BRCA1 mutation carriers.

9.
Circulation ; 144(3): 229-242, 2021 07 20.
Article in English | MEDLINE | ID: mdl-33910361

ABSTRACT

BACKGROUND: Genetic variants in SCN10A, encoding the neuronal voltage-gated sodium channel NaV1.8, are strongly associated with atrial fibrillation, Brugada syndrome, cardiac conduction velocities, and heart rate. The cardiac function of SCN10A has not been resolved, however, and diverging mechanisms have been proposed. Here, we investigated the cardiac expression of SCN10A and the function of a variant-sensitive intronic enhancer previously linked to the regulation of SCN5A, encoding the major essential cardiac sodium channel NaV1.5. METHODS: The expression of SCN10A was investigated in mouse and human hearts. With the use of CRISPR/Cas9 genome editing, the mouse intronic enhancer was disrupted, and mutant mice were characterized by transcriptomic and electrophysiological analyses. The association of genetic variants at SCN5A-SCN10A enhancer regions and gene expression were evaluated by genome-wide association studies single-nucleotide polymorphism mapping and expression quantitative trait loci analysis. RESULTS: We found that cardiomyocytes of the atria, sinoatrial node, and ventricular conduction system express a short transcript comprising the last 7 exons of the gene (Scn10a-short). Transcription occurs from an intronic enhancer-promoter complex, whereas full-length Scn10a transcript was undetectable in the human and mouse heart. Expression quantitative trait loci analysis revealed that the genetic variants in linkage disequilibrium with genetic variant rs6801957 in the intronic enhancer associate with SCN10A transcript levels in the heart. Genetic modification of the enhancer in the mouse genome led to reduced cardiac Scn10a-short expression in atria and ventricles, reduced cardiac sodium current in atrial cardiomyocytes, atrial conduction slowing and arrhythmia, whereas the expression of Scn5a, the presumed enhancer target gene, remained unaffected. In patch-clamp transfection experiments, expression of Scn10a-short-encoded NaV1.8-short increased NaV1.5-mediated sodium current. We propose that noncoding genetic variation modulates transcriptional regulation of Scn10a-short in cardiomyocytes that impacts NaV1.5-mediated sodium current and heart rhythm. CONCLUSIONS: Genetic variants in and around SCN10A modulate enhancer function and expression of a cardiac-specific SCN10A-short transcript. We propose that noncoding genetic variation modulates transcriptional regulation of a functional C-terminal portion of NaV1.8 in cardiomyocytes that impacts on NaV1.5 function, cardiac conduction velocities, and arrhythmia susceptibility.


Subject(s)
Enhancer Elements, Genetic , Gene Expression Regulation , Heart Conduction System/physiology , Introns , NAV1.8 Voltage-Gated Sodium Channel/genetics , Action Potentials/genetics , Animals , Biomarkers , Cardiac Conduction System Disease/diagnosis , Cardiac Conduction System Disease/genetics , Cardiac Conduction System Disease/physiopathology , Cardiac Electrophysiology , Disease Susceptibility , Electrocardiography , Female , Genetic Association Studies , Male , Mice , NAV1.5 Voltage-Gated Sodium Channel/genetics , Quantitative Trait Loci , Quantitative Trait, Heritable
10.
Circ Res ; 128(2): 155-168, 2021 01 22.
Article in English | MEDLINE | ID: mdl-33146578

ABSTRACT

RATIONALE: Endogenously cycling adult cardiomyocytes increase after myocardial infarction (MI) but remain scarce and are generally thought not to contribute to myocardial function. However, this broadly held assumption has not been tested, mainly because of the lack of transgenic reporters that restrict Cre expression to adult cardiomyocytes that reenter the cell cycle. OBJECTIVE: We created and validated a new transgenic mouse, αMHC (alpha myosin heavy chain)-MerDreMer-Ki67p-RoxedCre (denoted αDKRC [cardiomyocyte-specific αMHC-MerDreMer-Ki67p-RoxedCre]) that restricts Cre expression to cycling adult cardiomyocytes and uniquely integrates spatial and temporal adult cardiomyocyte cycling events based on the DNA specificities of orthologous Dre and Cre recombinases. We then created αDKRC::DTA mice that expressed an inducible diphtheria toxin in adult cycling cardiomyocytes and examined the effects of ablating these endogenously cycling cardiomyocytes on myocardial function after ischemic-reperfusion (I/R) MI. METHODS AND RESULTS: A tandem αDKRC transgene was designed, validated in cultured cells, and used to make transgenic mice. The αDKRC transgene integrated between MYH6 and MYH7 and did not disrupt expression of the surrounding genes. Compared with controls, αDKRC::RLTG (Rox-Lox-tdTomato-eGFP) mice treated with Tamoxifen expressed tdTomato+ in cardiomyocytes with rare Bromodeoxyuridine+, eGFP+ cardiomyocytes, consistent with reentry of the cell cycle. We then pretreated αDKRC::RLTG mice with Tamoxifen to activate the reporter before sham or reperfusion (I/R) MI surgeries. Compared with Sham surgery, the I/R MI group had increased single and paired eGFP+ (enhanced green fluorescent protein)+ cardiomyocytes predominantly in the border zones (5.8±0.5 versus 3.3±0.3 cardiomyocytes per 10-micron section, N=8-9 mice per group, n=16-24 sections per mouse), indicative of cycled cardiomyocytes. The single to paired eGFP+ cardiomyocyte ratio was ≈9 to 1 (5.2±0.4 single versus 0.6±0.2 paired cardiomyocytes) in the I/R MI group after MI, suggesting that cycling cardiomyocytes were more likely to undergo polyploidy than replication. The ablation of endogenously cycling adult cardiomyocytes in αDKRC::DTA (diphtheria) mice caused progressive worsening left ventricular chamber size and function after I/R MI, compared with controls. CONCLUSIONS: Although scarce, endogenously cycling adult cardiomyocytes contribute to myocardial function after injury, suggesting that these cells may be physiologically relevant.


Subject(s)
Cell Cycle , Cell Proliferation , Myocardial Infarction/pathology , Myocardial Reperfusion Injury/pathology , Myocytes, Cardiac/pathology , Animals , Diphtheria Toxin/genetics , Diphtheria Toxin/metabolism , Disease Models, Animal , Female , HEK293 Cells , Humans , Integrases/genetics , Integrases/metabolism , Ki-67 Antigen/genetics , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Male , Mice, Inbred C57BL , Mice, Transgenic , Myocardial Infarction/genetics , Myocardial Infarction/metabolism , Myocardial Infarction/physiopathology , Myocardial Reperfusion Injury/genetics , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/physiopathology , Myocytes, Cardiac/metabolism , Myosin Heavy Chains/genetics , Myosin Heavy Chains/metabolism , Peptide Fragments/genetics , Peptide Fragments/metabolism , Promoter Regions, Genetic , Time Factors , Ventricular Function, Left , Ventricular Remodeling
11.
J Periodontol ; 91(6): 819-827, 2020 06.
Article in English | MEDLINE | ID: mdl-31495928

ABSTRACT

BACKGROUND: Although numerous reports have demonstrated that the junctional epithelium (JE) is derived from the reduced enamel epithelium (REE), the fate of the REE-derived JE remains controversial. The present study elucidated the fate of the REE-derived JE and the cell dynamics of stem/progenitor cells in the JE following tooth eruption. METHODS: Mandibular first molar germs (embryonic days 15 to postnatal 1-day-old) were transplanted into the socket of 2-week-old mice after extraction of the upper first molars of B6 wild-type (WT) and green fluorescent protein (GFP) transgenic mice. After analysis by µ-CT, paraffin sections were processed for immunohistochemistry for Nestin, Ki67 and GFP. We also performed chasing analysis using BrdU-administered TetOP-H2B-GFP mice. RESULTS: Amelogenesis progressed normally in the cervical areas, and the structure of the JE was like that in normal tooth development. The JE was GFP-negative in transplantations using GFP transgenic mice as the host, and the oral epithelium (OE) showed a positive reaction. By contrast, the JE remained GFP-positive throughout the experimental period in transplantations using GFP transgenic mice as the donor. Chasing analysis revealed that H2B-GFP- and 5-Bromo-2'-deoxyuridine (BrdU)-labeled cells in the basal side of the JE translocated to oral side of the JE as the chasing time increased. Some label-retaining cells remained at the supra-basal cell layer in the JE. CONCLUSION: These results suggest that REE-derived cell niche in the JE is maintained for a long time following tooth eruption. Therefore, the JE may be available as the source of the odontogenic epithelium.


Subject(s)
Epithelial Attachment , Tooth , Animals , Dental Enamel , Epithelial Cells , Epithelium , Mice , Tooth Eruption
12.
Regen Ther ; 11: 217-224, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31516919

ABSTRACT

Although insulin-like growth factor binding protein 5 (IGFBP5) may play a crucial role in activating the functions of periodontal and bone marrow stem cells, the factors responsible for regulating the maintenance of dental pulp stem cells (DPSCs) remain to be clarified. This study aimed to elucidate the role of IGFBP5 in maintaining pulpal homeostasis during tooth development and pulpal healing after tooth injury in doxycycline-inducible TetOP-histone 2B (H2B)-green fluorescent protein (GFP) transgenic mice (GFP expression was induced at E14.5 or E15.5) by using TUNEL assay, RT-PCR, in situ hybridization for Igfbp5, and immunohistochemistry for IGFBP5, Nestin, and GFP. To observe the pulpal response to exogenous stimuli, the roots of the maxillary first molars were resected, and the coronal portion was autografted into the sublingual region. Intense IGFBP5/Igfbp5 expression was observed in cells from the center of the pulp tissue and the subodontoblastic layer in developing teeth during postnatal Week 4. Intense H2B-GFP-expressing label-retaining cells (LRCs) were localized in the subodontoblastic layer in addition to the center of the pulp tissue, suggesting that slowly dividing cell populations reside in these areas. During postoperative days 3-7, the LRCs were maintained in the dental pulp, showed an IGFBP5-positve reaction in their nuclei, and lacked a TUNEL-positive reaction. In situ hybridization and RT-PCR analyses confirmed the expression of Igfbp5 in the dental pulp. These findings suggest that IGFBP5 play a pivotal role in regulating the survival and apoptosis of DPSCs during both tooth development and pulpal healing following tooth injury.

13.
Circulation ; 140(9): 765-778, 2019 08 27.
Article in English | MEDLINE | ID: mdl-31315475

ABSTRACT

BACKGROUND: Restrictive cardiomyopathy is a rare heart disease associated with mutations in sarcomeric genes and with phenotypic overlap with hypertrophic cardiomyopathy. There is no approved therapy directed at the underlying cause. Here, we explore the potential of an interfering RNA (RNAi) therapeutic for a human sarcomeric mutation in MYL2 causative of restrictive cardiomyopathy in a mouse model. METHODS: A short hairpin RNA (M7.8L) was selected from a pool for specificity and efficacy. Two groups of myosin regulatory light chain N47K transgenic mice were injected with M7.8L packaged in adeno-associated virus 9 at 3 days of age and 60 days of age. Mice were subjected to treadmill exercise and echocardiography after treatment to determine maximal oxygen uptake and left ventricular mass. At the end of treatment, heart, lung, liver, and kidney tissue was harvested to determine viral tropism and for transcriptomic and proteomic analysis. Cardiomyocytes were isolated for single-cell studies. RESULTS: A one-time injection of AAV9-M7.8L RNAi in 3-day-old humanized regulatory light chain mutant transgenic mice silenced the mutated allele (RLC-47K) with minimal effects on the normal allele (RLC-47N) assayed at 16 weeks postinjection. AAV9-M7.8L RNAi suppressed the expression of hypertrophic biomarkers, reduced heart weight, and attenuated a pathological increase in left ventricular mass. Single adult cardiac myocytes from mice treated with AAV9-M7.8L showed partial restoration of contraction, relaxation, and calcium kinetics. In addition, cardiac stress protein biomarkers, such as calmodulin-dependent protein kinase II and the transcription activator Brg1 were reduced, suggesting recovery toward a healthy myocardium. Transcriptome analyses further revealed no significant changes of argonaute (AGO1, AGO2) and endoribonuclease dicer (DICER1) transcripts, and endogenous microRNAs were preserved, suggesting that the RNAi pathway was not saturated. CONCLUSIONS: Our results show the feasibility, efficacy, and safety of RNAi therapeutics directed towards human restrictive cardiomyopathy. This is a promising step toward targeted therapy for a prevalent human disease.


Subject(s)
Cardiomyopathy, Restrictive/pathology , Myosin Light Chains/metabolism , RNA Interference , Alleles , Animals , Calcium/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Cardiomyopathy, Restrictive/prevention & control , DNA Helicases/genetics , DNA Helicases/metabolism , Disease Models, Animal , Gene Regulatory Networks , Genetic Vectors/metabolism , Humans , Mice , Mice, Transgenic , Muscle Contraction , Mutagenesis, Site-Directed , Myocardium/metabolism , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Myosin Light Chains/antagonists & inhibitors , Myosin Light Chains/genetics , RNA, Small Interfering/metabolism
14.
Endocrinol Metab (Seoul) ; 34(1): 11-22, 2019 03.
Article in English | MEDLINE | ID: mdl-30784243

ABSTRACT

The development of next generation sequencing (NGS) has led to marked advancement of our understanding of genetic events mediating the initiation and progression of thyroid cancers. The NGS studies have confirmed the previously reported high frequency of mutually-exclusive oncogenic alterations affecting BRAF and RAS proto-oncogenes in all stages of thyroid cancer. Initially identified by traditional sequencing approaches, the NGS studies also confirmed the acquisition of alterations that inactivate tumor protein p53 (TP53) and activate phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit alpha (PIK3CA) in advanced thyroid cancers. Novel alterations, such as those in telomerase reverse transcriptase (TERT) promoter and mating-type switching/sucrose non-fermenting (SWI/SNF) complex, are also likely to promote progression of the BRAFV600E-driven thyroid cancers. A number of genetically engineered mouse models (GEMM) of BRAFV600E-driven thyroid cancer have been developed to investigate thyroid tumorigenesis mediated by oncogenic BRAF and to explore the role of genetic alterations identified in the genomic analyses of advanced thyroid cancer to promote tumor progression. This review will discuss the various GEMMs that have been developed to investigate oncogenic BRAFV600E-driven thyroid cancers.


Subject(s)
Promoter Regions, Genetic/genetics , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogenes/genetics , Thyroid Neoplasms/genetics , Animals , Carcinoma, Papillary/genetics , Class I Phosphatidylinositol 3-Kinases/metabolism , Disease Progression , High-Throughput Nucleotide Sequencing/methods , Humans , Mice , Mice, Transgenic/genetics , Mutation , Proto-Oncogene Mas , Telomerase/genetics , Thyroid Neoplasms/veterinary , Tumor Suppressor Protein p53/metabolism
15.
Arterioscler Thromb Vasc Biol ; 39(4): 603-612, 2019 04.
Article in English | MEDLINE | ID: mdl-30727757

ABSTRACT

Smooth muscle cells (SMCs) are a critical component of blood vessel walls that provide structural support, regulate vascular tone, and allow for vascular remodeling. These cells also exhibit a remarkable plasticity that contributes to vascular growth and repair but also to cardiovascular pathologies, including atherosclerosis, intimal hyperplasia and restenosis, aneurysm, and transplant vasculopathy. Mouse models have been an important tool for the study of SMC functions. The development of smooth muscle-expressing Cre-driver lines has allowed for exciting discoveries, including recent advances revealing the diversity of phenotypes derived from mature SMC transdifferentiation in vivo using inducible CreER T2 lines. We review SMC-targeting Cre lines driven by the Myh11, Tagln, and Acta2 promoters, including important technical considerations associated with these models. Limitations that can complicate study of the vasculature include expression in visceral SMCs leading to confounding phenotypes, and expression in multiple nonsmooth muscle cell types, such as Acta2-Cre expression in myofibroblasts. Notably, the frequently employed Tagln/ SM22α- Cre driver expresses in the embryonic heart but can also confer expression in nonmuscular cells including perivascular adipocytes and their precursors, myeloid cells, and platelets, with important implications for interpretation of cardiovascular phenotypes. With new Cre-driver lines under development and the increasing use of fate mapping methods, we are entering an exciting new era in SMC research.


Subject(s)
Gene Targeting/methods , Muscle, Smooth, Vascular/physiology , Promoter Regions, Genetic , Actins/biosynthesis , Actins/genetics , Animals , Cell Line , Cell Lineage , Cell Transdifferentiation , Gene Expression Regulation , Gene Knockout Techniques , Humans , Mice , Microfilament Proteins/biosynthesis , Microfilament Proteins/genetics , Muscle Proteins/biosynthesis , Muscle Proteins/genetics , Myocytes, Smooth Muscle/physiology , Myofibroblasts/physiology , Myosin Heavy Chains/biosynthesis , Myosin Heavy Chains/genetics , Neovascularization, Pathologic/physiopathology , Neovascularization, Physiologic , Phenotype , Recombinant Fusion Proteins/metabolism
16.
Circulation ; 139(4): 533-545, 2019 01 22.
Article in English | MEDLINE | ID: mdl-30586742

ABSTRACT

BACKGROUND: N6-Methyladenosine (m6A) methylation is the most prevalent internal posttranscriptional modification on mammalian mRNA. The role of m6A mRNA methylation in the heart is not known. METHODS: To determine the role of m6A methylation in the heart, we isolated primary cardiomyocytes and performed m6A immunoprecipitation followed by RNA sequencing. We then generated genetic tools to modulate m6A levels in cardiomyocytes by manipulating the levels of the m6A RNA methylase methyltransferase-like 3 (METTL3) both in culture and in vivo. We generated cardiac-restricted gain- and loss-of-function mouse models to allow assessment of the METTL3-m6A pathway in cardiac homeostasis and function. RESULTS: We measured the level of m6A methylation on cardiomyocyte mRNA, and found a significant increase in response to hypertrophic stimulation, suggesting a potential role for m6A methylation in the development of cardiomyocyte hypertrophy. Analysis of m6A methylation showed significant enrichment in genes that regulate kinases and intracellular signaling pathways. Inhibition of METTL3 completely abrogated the ability of cardiomyocytes to undergo hypertrophy when stimulated to grow, whereas increased expression of the m6A RNA methylase METTL3 was sufficient to promote cardiomyocyte hypertrophy both in vitro and in vivo. Finally, cardiac-specific METTL3 knockout mice exhibit morphological and functional signs of heart failure with aging and stress, showing the necessity of RNA methylation for the maintenance of cardiac homeostasis. CONCLUSIONS: Our study identified METTL3-mediated methylation of mRNA on N6-adenosines as a dynamic modification that is enhanced in response to hypertrophic stimuli and is necessary for a normal hypertrophic response in cardiomyocytes. Enhanced m6A RNA methylation results in compensated cardiac hypertrophy, whereas diminished m6A drives eccentric cardiomyocyte remodeling and dysfunction, highlighting the critical importance of this novel stress-response mechanism in the heart for maintaining normal cardiac function.


Subject(s)
Adenosine/analogs & derivatives , Hypertrophy, Left Ventricular/enzymology , Methyltransferases/metabolism , Myocytes, Cardiac/enzymology , Ventricular Function, Left , Ventricular Remodeling , Adenosine/metabolism , Animals , Cells, Cultured , Disease Models, Animal , Female , Hypertrophy, Left Ventricular/genetics , Hypertrophy, Left Ventricular/pathology , Hypertrophy, Left Ventricular/physiopathology , Male , Methyltransferases/deficiency , Methyltransferases/genetics , Mice, Inbred C57BL , Mice, Knockout , Myocytes, Cardiac/pathology , RNA Processing, Post-Transcriptional , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Signal Transduction
17.
Article in English | WPRIM (Western Pacific) | ID: wpr-739224

ABSTRACT

The development of next generation sequencing (NGS) has led to marked advancement of our understanding of genetic events mediating the initiation and progression of thyroid cancers. The NGS studies have confirmed the previously reported high frequency of mutually-exclusive oncogenic alterations affecting BRAF and RAS proto-oncogenes in all stages of thyroid cancer. Initially identified by traditional sequencing approaches, the NGS studies also confirmed the acquisition of alterations that inactivate tumor protein p53 (TP53) and activate phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit alpha (PIK3CA) in advanced thyroid cancers. Novel alterations, such as those in telomerase reverse transcriptase (TERT) promoter and mating-type switching/sucrose non-fermenting (SWI/SNF) complex, are also likely to promote progression of the BRAF(V600E)-driven thyroid cancers. A number of genetically engineered mouse models (GEMM) of BRAF(V600E)-driven thyroid cancer have been developed to investigate thyroid tumorigenesis mediated by oncogenic BRAF and to explore the role of genetic alterations identified in the genomic analyses of advanced thyroid cancer to promote tumor progression. This review will discuss the various GEMMs that have been developed to investigate oncogenic BRAF(V600E)-driven thyroid cancers.


Subject(s)
Animals , Mice , Carcinogenesis , Catalytic Domain , Mice, Transgenic , Negotiating , Proto-Oncogene Proteins B-raf , Proto-Oncogenes , Telomerase , Thyroid Gland , Thyroid Neoplasms
19.
Zhonghua Yan Ke Za Zhi ; 54(12): 911-917, 2018 Dec 11.
Article in Chinese | MEDLINE | ID: mdl-30526790

ABSTRACT

Objective: To investigate the differential expression of small leucine-rich proteoglycans at mRNA level in Lumican transgenic mouse cornea with Real-time Quantitative PCR Detecting System. Methods: Experimental research. Ten Lumican transgenic mice (5 male and 5 female) were chosen as experimental group and 10 wild mice (5 male and 5 female) were chosen as control group. All the mice were killed and enucleated both eyes at eight weeks of age. Gene expression levels of Lumican, Decorin, Biglycan, Keratocan, Fibromodulin in the excised corneas were analyzed by real-time quantitative polymerase chain reaction (RT-Q-PCR) using Real-time Quantitative PCR Detecting System. Differential expression within each group were analysed by fold changes and independent t-test. Results: There were statistic different expression level of Lumican, Decorin, Biglycan and Keratocan mRNA between experimental and control group. The expression level of Lumican RNA was found to be 1.497-fold increased relative to the control (t=4.34, P<0.05) , while Decorin, Biglycan, Keratocan were 0.648-fold (t=-9.98, P<0.05) , 0.522-fold (t=-7.74,P<0.05), 0.323-fold (t=-95.94, P<0.05)decreased in transgenic mice. Fibromodulin mRNA up regulated 1.193-fold in transgenic mice without statistic difference (t=1.66, P>0.05). Conclusions: Lumican gene mutation(cDNA 569T>C) results in abnormal SLRP expression in transgenic mouse cornea at mRNA level, which may indicate that this mutation changes the structure of Lumican and impairs the function of regulating SLRP expression. Also, Lumican gene mutation leads to amio acid exchanging(L199P), which may hinder Lumican from binding to collagens and result in abnormal expression of SLRP at mRNA level. (Chin J Ophthalmol, 2018, 54:911-917).


Subject(s)
Cornea , Small Leucine-Rich Proteoglycans , Animals , Chondroitin Sulfate Proteoglycans , Cornea/metabolism , Extracellular Matrix Proteins , Female , Keratan Sulfate , Lumican/genetics , Lumican/metabolism , Male , Mice , Mice, Transgenic , Small Leucine-Rich Proteoglycans/genetics , Small Leucine-Rich Proteoglycans/metabolism
20.
Circulation ; 138(10): 1012-1024, 2018 09 04.
Article in English | MEDLINE | ID: mdl-29666070

ABSTRACT

BACKGROUND: Although c-Kit+ adult progenitor cells were initially reported to produce new cardiomyocytes in the heart, recent genetic evidence suggests that such events are exceedingly rare. However, to determine if these rare events represent true de novo cardiomyocyte formation, we deleted the necessary cardiogenic transcription factors Gata4 and Gata6 from c-Kit-expressing cardiac progenitor cells. METHODS: Kit allele-dependent lineage tracing and fusion analysis were performed in mice following simultaneous Gata4 and Gata6 cell type-specific deletion to examine rates of putative de novo cardiomyocyte formation from c-Kit+ cells. Bone marrow transplantation experiments were used to define the contribution of Kit allele-derived hematopoietic cells versus Kit lineage-dependent cells endogenous to the heart in contributing to apparent de novo lineage-traced cardiomyocytes. A Tie2CreERT2 transgene was also used to examine the global impact of Gata4 deletion on the mature cardiac endothelial cell network, which was further evaluated with select angiogenesis assays. RESULTS: Deletion of Gata4 in Kit lineage-derived endothelial cells or in total endothelial cells using the Tie2CreERT2 transgene, but not from bone morrow cells, resulted in profound endothelial cell expansion, defective endothelial cell differentiation, leukocyte infiltration into the heart, and a dramatic increase in Kit allele-dependent lineage-traced cardiomyocytes. However, this increase in labeled cardiomyocytes was an artefact of greater leukocyte-cardiomyocyte cellular fusion because of defective endothelial cell differentiation in the absence of Gata4. CONCLUSIONS: Past identification of presumed de novo cardiomyocyte formation in the heart from c-Kit+ cells using Kit allele lineage tracing appears to be an artefact of labeled leukocyte fusion with cardiomyocytes. Deletion of Gata4 from c-Kit+ endothelial progenitor cells or adult endothelial cells negatively impacted angiogenesis and capillary network integrity.


Subject(s)
Cell Lineage , Cell Proliferation , Endothelial Cells/metabolism , GATA4 Transcription Factor/metabolism , Myocytes, Cardiac/metabolism , Neovascularization, Physiologic , Proto-Oncogene Proteins c-kit/metabolism , Regeneration , Animals , Bone Marrow Transplantation , Cell Fusion , Cell Tracking/methods , Cells, Cultured , Female , GATA4 Transcription Factor/deficiency , GATA4 Transcription Factor/genetics , GATA6 Transcription Factor/genetics , GATA6 Transcription Factor/metabolism , Leukocytes/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...