Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
J Vis Exp ; (195)2023 May 19.
Article in English | MEDLINE | ID: mdl-37318237

ABSTRACT

Self-renewal capacity and multi-lineage differentiation potential are generally regarded as the defining characteristics of hematopoietic stem cells (HSCs). However, numerous studies have suggested that functional heterogeneity exists in the HSC compartment. Recent single-cell analyses have reported HSC clones with different cell fates within the HSC compartment, which are referred to as biased HSC clones. The mechanisms underlying heterogeneous or poorly reproducible results are little understood, especially regarding the length of self-renewal when purified HSC fractions are transplanted by conventional immunostaining. Therefore, establishing a reproducible isolation method for long-term HSCs (LT-HSCs) and short-term HSCs (ST-HSCs), defined by the length of their self-renewal, is crucial for overcoming this issue. Using unbiased multi-step screening, we identified a transcription factor, Hoxb5, which may be an exclusive marker of LT-HSCs in the mouse hematopoietic system. Based on this finding, we established a Hoxb5 reporter mouse line and successfully isolated LT-HSCs and ST-HSCs. Here we describe a detailed protocol for the isolation of LT-HSCs and ST-HSCs using the Hoxb5 reporter system. This isolation method will help researchers better understand the mechanisms of self-renewal and the biological basis for such heterogeneity in the HSC compartment.


Subject(s)
Gene Expression Regulation , Hematopoietic Stem Cells , Mice , Animals , Cell Differentiation/physiology , Homeodomain Proteins/metabolism
2.
Nat Commun ; 14(1): 550, 2023 02 08.
Article in English | MEDLINE | ID: mdl-36754961

ABSTRACT

Myofibroblasts cause tissue fibrosis by producing extracellular matrix proteins, such as collagens. Humoral factors like TGF-ß, and matrix stiffness are important for collagen production by myofibroblasts. However, the molecular mechanisms regulating their ability to produce collagen remain poorly characterised. Here, we show that vestigial-like family member 3 (VGLL3) is specifically expressed in myofibroblasts from mouse and human fibrotic hearts and promotes collagen production. Further, substrate stiffness triggers VGLL3 translocation into the nucleus through the integrin ß1-Rho-actin pathway. In the nucleus, VGLL3 undergoes liquid-liquid phase separation via its low-complexity domain and is incorporated into non-paraspeckle NONO condensates containing EWS RNA-binding protein 1 (EWSR1). VGLL3 binds EWSR1 and suppresses miR-29b, which targets collagen mRNA. Consistently, cardiac fibrosis after myocardial infarction is significantly attenuated in Vgll3-deficient mice, with increased miR-29b expression. Overall, our results reveal an unrecognised VGLL3-mediated pathway that controls myofibroblasts' collagen production, representing a novel therapeutic target for tissue fibrosis.


Subject(s)
MicroRNAs , Myocardium , Humans , Mice , Animals , Myocardium/metabolism , Transforming Growth Factor beta1/metabolism , Fibrosis , Collagen/metabolism , Myofibroblasts/metabolism , Transcription Factors/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Fibroblasts/metabolism , Cells, Cultured
3.
FEBS Open Bio ; 13(2): 380-391, 2023 02.
Article in English | MEDLINE | ID: mdl-36633120

ABSTRACT

During myocardial infarction (MI), cardiac cells at the infarcted area undergo cell death. In response, cardiac myofibroblasts, which are mainly differentiated from resident fibroblasts upon inflammation, produce extracellular matrix proteins such as collagen to fill the damaged areas of the heart to prevent cardiac rupture. In this study, we identified a cardioprotective role of G-protein-coupled receptor kinase 5 (GRK5) in MI. GRK5 expression was found to increase in the mouse heart after MI and was highly expressed in cardiac fibroblasts/myofibroblasts. In fibroblasts/myofibroblasts, GRK5 promoted the expression of inflammation-related genes through nuclear factor-κB activation, leading to an increase in the expression levels of fibrosis-related genes. Bone marrow transfer experiments confirmed that GRK5 in fibroblasts/myofibroblasts, but not in infiltrated macrophages in the infarcted area, is mainly responsible for GRK5-mediated inflammation in infarcted hearts. In addition, inflammation and fibrosis at the infarcted area were significantly suppressed in GRK5 knockout mice, resulting in increased mortality compared with that in wild-type mice. These data indicate that GRK5 in cardiac fibroblasts/myofibroblasts promotes inflammation and fibrosis to ameliorate the damage after MI.


Subject(s)
Myocardial Infarction , Myocardium , Animals , Mice , Collagen/metabolism , Fibrosis , Inflammation/metabolism , Mice, Knockout , Myocardial Infarction/genetics , Myocardium/metabolism
4.
Biochem Biophys Res Commun ; 561: 180-186, 2021 07 05.
Article in English | MEDLINE | ID: mdl-34023784

ABSTRACT

Fibrosis is a condition characterized by the overproduction of extracellular matrix (ECM) components (e.g., collagen) in the myofibroblasts, causing tissue hardening and eventual organ dysfunction. Currently, the molecular mechanisms that regulate ECM production in the myofibroblasts are still obscure. In this study, we investigated the function of GPRC5B in the cardiac and lung myofibroblasts using real-time RT-PCR and siRNA-mediated knockdown. We discovered a significantly high expression of Gprc5b in the tissues of the fibrosis mice models and confirmed that Gprc5b was consistently expressed in the myofibroblasts of fibrotic hearts and lungs. We also found that Gprc5b expression was associated and may be dependent on the actin-MRTF-SRF signaling pathway. Notably, we observed that Gprc5b knockdown reduced the expression of collagen genes in the cardiac and lung myofibroblasts. Therefore, our findings reveal that GPRC5B enhances collagen production in the myofibroblasts, which directly promotes fibrosis in the tissues.


Subject(s)
Collagen/metabolism , Fibrosis/pathology , Heart/physiopathology , Liver/metabolism , Lung/metabolism , Myofibroblasts/metabolism , Receptors, G-Protein-Coupled/metabolism , Animals , Disease Models, Animal , Extracellular Matrix/metabolism , Extracellular Matrix/pathology , Fibrosis/metabolism , Liver/pathology , Lung/pathology , Male , Mice , Myofibroblasts/pathology , Signal Transduction
5.
Biochem Biophys Res Commun ; 529(2): 224-230, 2020 08 20.
Article in English | MEDLINE | ID: mdl-32703415

ABSTRACT

Fibrosis is attributed to excess deposition of extracellular matrix (ECM) proteins including collagen and is associated with various organ dysfunction. This excessive ECM is produced by myofibroblasts, which are differentiated from various cells by a variety of stimuli, represented by TGF-ß. However, molecular mechanisms for the regulation of ECM production in myofibroblasts remain obscure. In this study, we demonstrate that the expression of drebrin, which binds to and increases the stability of actin filament in neurons, is increased in mouse hearts and lungs upon fibrosis. Drebrin is mainly expressed in myofibroblasts in the fibrotic hearts and lungs and promotes the expression of fibrosis-related genes, such as Acta2 and Col1a1. Taken together, our study identifies drebrin as a molecule that promotes the production of fibrosis-related genes in myofibroblasts.


Subject(s)
Lung/pathology , Myocardium/pathology , Myofibroblasts/pathology , Neuropeptides/genetics , Animals , Cell Differentiation , Cells, Cultured , Fibrosis , Lung/metabolism , Mice , Mice, Inbred C57BL , Myocardium/metabolism , Myofibroblasts/metabolism , NIH 3T3 Cells , Pulmonary Fibrosis/genetics , Pulmonary Fibrosis/pathology , Up-Regulation
6.
FASEB J ; 34(6): 8749-8763, 2020 06.
Article in English | MEDLINE | ID: mdl-32385915

ABSTRACT

Leukotriene B4 receptor 1 (BLT1), a high-affinity G-protein-coupled receptor for leukotriene B4 (LTB4 ), is expressed on various inflammatory cells and plays critical roles in several inflammatory diseases. In myocardial infarction (MI), various inflammatory cells are known to be recruited to the infarcted area, but the function of BLT1 in MI is poorly understood. Here, we investigated the role of BLT1 in MI and the therapeutic effect of a BLT1 antagonist, ONO-4057, on MI. Mice with infarcted hearts showed increased BLT1 expression and LTB4 levels. BLT1-knockout mice with infarcted hearts exhibited attenuated leukocyte infiltration, proinflammatory cytokine production, and cell death, which led to reduced mortality and improved cardiac function after MI. Bone-marrow transplantation studies showed that BLT1 expressed on bone marrow-derived cells was responsible for the exacerbation of inflammation in infarcted hearts. Furthermore, ONO-4057 administration attenuated the inflammatory responses in hearts surgically treated for MI, which resulted in reduced mortality and improved cardiac function after MI. Our study demonstrated that BLT1 contributes to excessive inflammation after MI and could represent a new therapeutic target for MI.


Subject(s)
Inflammation/metabolism , Myocardial Infarction/metabolism , Receptors, Leukotriene B4/metabolism , Animals , Disease Models, Animal , Leukotriene B4/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Signal Transduction/physiology
7.
J Biochem ; 168(1): 1-6, 2020 Jul 01.
Article in English | MEDLINE | ID: mdl-32302392

ABSTRACT

Myocardial infarction is one of the major causes of death worldwide. Many heart cells die during myocardial infarction through various processes such as necrosis, apoptosis, necroptosis, autophagy-related cell death, pyroptosis and ferroptosis. These dead cells in infarcted hearts expose the so-called 'eat-me' signals, such as phosphatidylserine, on their surfaces, enhancing their removal by professional and non-professional phagocytes. Clearance of dead cells by phagocytes in the diseased hearts plays a crucial role in the pathology of myocardial infarction by inhibiting the inflammatory responses caused by the leakage of contents from dead cells. This review focuses on the rapidly growing understanding of the molecular mechanisms of dead cell phagocytosis, termed efferocytosis, during myocardial infarction, which contributes to the pathophysiology of myocardial infarction.


Subject(s)
Apoptosis , Inflammation/physiopathology , Macrophages/physiology , Myocardial Infarction/physiopathology , Phagocytes/physiology , Phagocytosis , Humans , Signal Transduction
8.
Sci Rep ; 7(1): 7812, 2017 08 10.
Article in English | MEDLINE | ID: mdl-28798316

ABSTRACT

Myocardial infarction (MI) is an ischaemic heart condition caused by the occlusion of coronary arteries. Following MI, lactic acid from anaerobic glycolysis increases and infiltrating immune cells produce severe inflammation, which leads to acidosis in the ischaemic heart. However, the physiological implication of this pH reduction remains largely unknown. T-cell death-associated gene 8 (TDAG8) is a proton-sensing G protein-coupled receptor found on cardiac macrophages that recognise increases in extracellular protons. We demonstrated that TDAG8 negatively regulates the transcription of the chemokine Ccl20. The infarcted hearts of TDAG8 KO mice showed an increase in CCL20 expression and the number of infiltrating IL-17A-producing γδT cells that express CCR6, a receptor for CCL20. Accordingly, excessive IL-17A production, which is linked to the functional deterioration after MI, was observed in MI-operated TDAG8 KO mice. The survival rate and cardiac function significantly decreased in TDAG8 KO mice compared with those in wild-type mice after MI. Thus, our results suggest that TDAG8 is a key regulator of MI and a potential therapeutic target.


Subject(s)
Chemokine CCL20/genetics , Myocardial Infarction/genetics , Animals , Chemokine CCL20/metabolism , Disease Models, Animal , Gene Expression Regulation , Interleukin-17/metabolism , Intraepithelial Lymphocytes/immunology , Mice , Mice, Knockout , Myocardial Infarction/immunology , Myocardial Infarction/metabolism , Receptors, CCR6/metabolism , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Survival Analysis , Transcription, Genetic
9.
Bio Protoc ; 7(18): e2552, 2017 Sep 20.
Article in English | MEDLINE | ID: mdl-34541198

ABSTRACT

In myocardial infarction (MI), a plenty of cardiomyocytes undergo necrosis and necroptosis due to the lack of oxygen and nutrients. The dead cardiomyocytes are promptly engulfed by phagocytes. When the dead cells are not engulfed, the noxious contents of the cells are released outside, and thus, induce inflammation, and obstruct the function of organs. Therefore, phagocytosis is crucial for maintaining homeostasis of organs. Herein, we describe a protocol of an in vitro phagocytosis assay of necroptotic cells.

10.
Bio Protoc ; 7(18): e2553, 2017 Sep 20.
Article in English | MEDLINE | ID: mdl-34541199

ABSTRACT

In myocardial infarction (MI), a number of cardiomyocytes undergo apoptosis. These apoptotic cardiomyocytes are promptly engulfed by phagocytes. If the dead cells are not engulfed, their noxious contents are released outside, resulting in induction of inflammation. Therefore, the removal of these dead cells is necessary. However, the contribution of each phagocyte type to the removal of apoptotic cells in infarcted hearts remains unresolved. Here, we describe an in vitro protocol for a phagocytosis assay to compare the engulfment ability of cardiac macrophages and cardiac myofibroblasts.

11.
J Clin Invest ; 127(1): 383-401, 2017 01 03.
Article in English | MEDLINE | ID: mdl-27918308

ABSTRACT

Myocardial infarction (MI) results in the generation of dead cells in the infarcted area. These cells are swiftly removed by phagocytes to minimize inflammation and limit expansion of the damaged area. However, the types of cells and molecules responsible for the engulfment of dead cells in the infarcted area remain largely unknown. In this study, we demonstrated that cardiac myofibroblasts, which execute tissue fibrosis by producing extracellular matrix proteins, efficiently engulf dead cells. Furthermore, we identified a population of cardiac myofibroblasts that appears in the heart after MI in humans and mice. We found that these cardiac myofibroblasts secrete milk fat globule-epidermal growth factor 8 (MFG-E8), which promotes apoptotic engulfment, and determined that serum response factor is important for MFG-E8 production in myofibroblasts. Following MFG-E8-mediated engulfment of apoptotic cells, myofibroblasts acquired antiinflammatory properties. MFG-E8 deficiency in mice led to the accumulation of unengulfed dead cells after MI, resulting in exacerbated inflammatory responses and a substantial decrease in survival. Moreover, MFG-E8 administration into infarcted hearts restored cardiac function and morphology. MFG-E8-producing myofibroblasts mainly originated from resident cardiac fibroblasts and cells that underwent endothelial-mesenchymal transition in the heart. Together, our results reveal previously unrecognized roles of myofibroblasts in regulating apoptotic engulfment and a fundamental importance of these cells in recovery from MI.


Subject(s)
Antigens, Surface/metabolism , Apoptosis , Epithelial-Mesenchymal Transition , Milk Proteins/metabolism , Myocardial Infarction/metabolism , Myocardium/metabolism , Myofibroblasts/metabolism , Animals , Antigens, Surface/genetics , Cell Survival/genetics , Male , Mice , Mice, Knockout , Milk Proteins/genetics , Myocardial Infarction/genetics , Myocardial Infarction/pathology , Myocardium/pathology , Myofibroblasts/pathology
12.
Biochem Biophys Res Commun ; 461(2): 307-13, 2015 May 29.
Article in English | MEDLINE | ID: mdl-25881508

ABSTRACT

G protein-coupled receptor kinases (GRKs) comprise a family of seven serine/threonine kinases that phosphorylate agonist-activated G protein-coupled receptors (GPCRs). It has recently been reported that GRKs regulate GPCR-independent signaling through the phosphorylation of intracellular proteins. To date, several intracellular substrates for GRK2 and GRK5 have been reported. However, those for GRK6 are poorly understood. Here we identified IκBα, a negative regulator of NF-κB signaling, as a substrate for GRK6. GRK6 directly phosphorylated IκBα at Ser(32)/Ser(36), and the kinase activity of GRK6 was required for the promotion of NF-κB signaling after TNF-α stimulation. Knockout of GRK6 in peritoneal macrophages remarkably attenuated the transcription of inflammatory genes after TNF-α stimulation. In addition, we developed a bioluminescence resonance energy transfer (BRET) probe to monitor GRK6 activity. Using this probe, we revealed that the conformational change of GRK6 was induced by TNF-α. In summary, our study demonstrates that TNF-α induces GRK6 activation, and GRK6 promotes inflammatory responses through the phosphorylation of IκBα.


Subject(s)
G-Protein-Coupled Receptor Kinases/immunology , I-kappa B Proteins/immunology , Inflammation/immunology , Tumor Necrosis Factor-alpha/immunology , Animals , Cells, Cultured , G-Protein-Coupled Receptor Kinases/chemistry , G-Protein-Coupled Receptor Kinases/genetics , G-Protein-Coupled Receptor Kinases/metabolism , Gene Knockdown Techniques , Gene Knockout Techniques , I-kappa B Proteins/chemistry , I-kappa B Proteins/metabolism , Inflammation/metabolism , Mice , Mice, Inbred C57BL , NF-KappaB Inhibitor alpha , NF-kappa B/immunology , NIH 3T3 Cells , Phosphorylation , Protein Conformation
13.
FEBS J ; 274(12): 3055-64, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17509075

ABSTRACT

The eye lens is composed of fiber cells that differentiate from epithelial cells on its anterior surface. In concert with this differentiation, a set of proteins essential for lens function is synthesized, and the cellular organelles are degraded. DNase II-like acid DNase, also called DNase IIbeta, is specifically expressed in the lens, and degrades the DNA in the lens fiber cells. Here we report that DNase II-like acid DNase is synthesized as a precursor with a signal sequence, and is localized to lysosomes. DNase II-like acid DNase mRNA was found in cortical fiber cells but not epithelial cells, indicating that its expression is induced during the differentiation of epithelial cells into fiber cells. Immunohistochemical and immunocytochemical analyses indicated that DNase II-like acid DNase was colocalized with Lamp-1 in the lysosomes of fiber cells in a relatively narrow region bordering the organelle-free zone, and was often found in degenerating nuclei. A comparison by microarray analysis of the gene expression profiles between epithelial and cortical fiber cells of young mouse lens indicated that some genes for lysosomal enzymes (cathepsins and lipases) were strongly expressed in the fiber cells. These results suggest that the lysosomal system plays a role in the degradation of cellular organelles during lens cell differentiation.


Subject(s)
DNA/metabolism , Endodeoxyribonucleases/metabolism , Epithelial Cells/metabolism , Lens Cortex, Crystalline/metabolism , RNA, Messenger/metabolism , Amino Acid Sequence , Animals , Cathepsins/metabolism , Cell Differentiation/physiology , Cell Line , Cell Nucleus/metabolism , Endodeoxyribonucleases/genetics , Epithelial Cells/cytology , Humans , Lens Cortex, Crystalline/cytology , Lipase/metabolism , Lysosomal Membrane Proteins/metabolism , Lysosomes/metabolism , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Oligonucleotide Array Sequence Analysis , Protein Sorting Signals/physiology , Recombinant Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...