Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
Blood Sci ; 6(4): e00199, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39027902

ABSTRACT

Hematopoietic stem progenitor cells (HSPCs) are derived from a specialized subset of endothelial cells named hemogenic endothelial cells (HECs) via a process of endothelial-to-hematopoietic transition during embryogenesis. Recently, with the usage of multiple single-cell technologies and advanced genetic lineage tracing techniques, namely, "TIF" approaches that combining transcriptome, immunophenotype and function/fate analyses, massive new insights have been achieved regarding the cellular and molecular evolution underlying the emergence of HSPCs from embryonic vascular beds. In this review, we focus on the most recent advances in the enrichment markers, functional characteristics, developmental paths, molecular controls, and the embryonic site-relevance of the key intermediate cell populations bridging embryonic vascular and hematopoietic systems, namely HECs and pre-hematopoietic stem cells, the immediate progenies of some HECs, in mouse and human embryos. Specifically, using expression analyses at both transcriptional and protein levels and especially efficient functional assays, we propose that the onset of Kit expression is at the HEC stage, which has previously been controversial.

2.
Cell ; 187(12): 3090-3107.e21, 2024 Jun 06.
Article in English | MEDLINE | ID: mdl-38749423

ABSTRACT

Platelet dysregulation is drastically increased with advanced age and contributes to making cardiovascular disorders the leading cause of death of elderly humans. Here, we reveal a direct differentiation pathway from hematopoietic stem cells into platelets that is progressively propagated upon aging. Remarkably, the aging-enriched platelet path is decoupled from all other hematopoietic lineages, including erythropoiesis, and operates as an additional layer in parallel with canonical platelet production. This results in two molecularly and functionally distinct populations of megakaryocyte progenitors. The age-induced megakaryocyte progenitors have a profoundly enhanced capacity to engraft, expand, restore, and reconstitute platelets in situ and upon transplantation and produce an additional platelet population in old mice. The two pools of co-existing platelets cause age-related thrombocytosis and dramatically increased thrombosis in vivo. Strikingly, aging-enriched platelets are functionally hyper-reactive compared with the canonical platelet populations. These findings reveal stem cell-based aging as a mechanism for platelet dysregulation and age-induced thrombosis.


Subject(s)
Aging , Blood Platelets , Cell Differentiation , Hematopoietic Stem Cells , Thrombosis , Animals , Hematopoietic Stem Cells/metabolism , Blood Platelets/metabolism , Thrombosis/pathology , Thrombosis/metabolism , Mice , Humans , Megakaryocytes/metabolism , Mice, Inbred C57BL , Megakaryocyte Progenitor Cells/metabolism , Male
3.
Cell Stem Cell ; 30(11): 1549-1558.e5, 2023 11 02.
Article in English | MEDLINE | ID: mdl-37794588

ABSTRACT

The maternal liver undergoes dramatic enlargement to adapt to the increased metabolic demands during pregnancy. However, the cellular sources for liver growth during pregnancy remain largely elusive. Here, we employed a proliferation recording system, ProTracer, to examine the spatial-temporal proliferation of hepatocytes during pregnancy. We discovered that during early to late pregnancy, hepatocyte proliferation initiated from zone 1, to zone 2, and lastly to zone 3, with the majority of new hepatocytes being generated in zone 2. Additionally, using single-cell RNA sequencing, we observed that Ccnd1 was highly enriched in zone 2 hepatocytes. We further applied dual-recombinase-mediated genetic lineage tracing to reveal that Ccnd1+ hepatocytes expanded preferentially during pregnancy. Moreover, we demonstrated that estrogen induces liver enlargement during pregnancy, which was abolished in Ccnd1 knockout mice. Our work revealed a unique spatial-temporal hepatocyte proliferation pattern during pregnancy, with Ccnd1+ hepatocytes in zone 2 serving as the major cellular source for hepatic enlargement.


Subject(s)
Hepatocytes , Liver Regeneration , Mice , Animals , Female , Pregnancy , Hepatocytes/metabolism , Liver/metabolism , Cell Proliferation , Mice, Knockout
4.
Cell Mol Life Sci ; 80(10): 300, 2023 Sep 23.
Article in English | MEDLINE | ID: mdl-37740736

ABSTRACT

AIMS: Mesenchymal stem cells (MSCs) present in the heart cannot differentiate into cardiomyocytes, but may play a role in pathological conditions. Therefore, the aim of this study was to scrutinise the role and mechanism of MSC differentiation in vivo during heart failure. METHODS AND RESULTS: We performed single-cell RNA sequencing of total non-cardiomyocytes from murine and adult human hearts. By analysing the transcriptomes of single cells, we illustrated the dynamics of the cell landscape during the progression of heart hypertrophy, including those of stem cell antigen-1 (Sca1)+ stem/progenitor cells and fibroblasts. By combining genetic lineage tracing and bone marrow transplantation models, we demonstrated that non-bone marrow-derived Sca1+ cells give rise to fibroblasts. Interestingly, partial depletion of Sca1+ cells alleviated the severity of myocardial fibrosis and led to a significant improvement in cardiac function in Sca1-CreERT2;Rosa26-eGFP-DTA mice. Similar non-cardiomyocyte cell composition and heterogeneity were observed in human patients with heart failure. Mechanistically, our study revealed that Sca1+ cells can transform into fibroblasts and affect the severity of fibrosis through the Wnt4-Pdgfra pathway. CONCLUSIONS: Our study describes the cellular landscape of hypertrophic hearts and reveals that fibroblasts derived from Sca1+ cells with a non-bone marrow source largely account for cardiac fibrosis. These findings provide novel insights into the pathogenesis of cardiac fibrosis and have potential therapeutic implications for heart failure. Non-bone marrow-derived Sca1+ cells differentiate into fibroblasts involved in cardiac fibrosis via Wnt4-PDGFRα pathway.

5.
Front Cardiovasc Med ; 10: 1085629, 2023.
Article in English | MEDLINE | ID: mdl-36923960

ABSTRACT

The Cre-loxP-mediated genetic lineage tracing system is essential for constructing the fate mapping of single-cell progeny or cell populations. Understanding the structural hierarchy of cardiac progenitor cells facilitates unraveling cell fate and origin issues in cardiac development. Several prospective Cre-loxP-based lineage-tracing systems have been used to analyze precisely the fate determination and developmental characteristics of endocardial cells (ECs), epicardial cells, and cardiomyocytes. Therefore, emerging lineage-tracing techniques advance the study of cardiovascular-related cellular plasticity. In this review, we illustrate the principles and methods of the emerging Cre-loxP-based genetic lineage tracing technology for trajectory monitoring of distinct cell lineages in the heart. The comprehensive demonstration of the differentiation process of single-cell progeny using genetic lineage tracing technology has made outstanding contributions to cardiac development and homeostasis, providing new therapeutic strategies for tissue regeneration in congenital and cardiovascular diseases (CVDs).

6.
Sci China Life Sci ; 66(1): 51-66, 2023 01.
Article in English | MEDLINE | ID: mdl-36322324

ABSTRACT

Arrhythmogenic cardiomyopathy (ACM) is one of the most common inherited cardiomyopathies, characterized by progressive fibrofatty replacement in the myocardium. However, the cellular origin of cardiac adipocytes in ACM remains largely unknown. Unraveling the cellular source of cardiac adipocytes in ACM would elucidate the underlying pathological process and provide a potential target for therapy. Herein, we generated an ACM mouse model by inactivating desmosomal gene desmoplakin in cardiomyocytes; and examined the adipogenic fates of several cell types in the disease model. The results showed that SOX9+, PDGFRa+, and PDGFRb+ mesenchymal cells, but not cardiomyocytes or smooth muscle cells, contribute to the intramyocardial adipocytes in the ACM model. Mechanistically, Bmp4 was highly expressed in the ACM mouse heart and functionally promoted cardiac mesenchymal-to-adipose transition in vitro.


Subject(s)
Cardiomyopathies , Heart , Mice , Animals , Myocardium/metabolism , Cardiomyopathies/genetics , Cardiomyopathies/metabolism , Cardiomyopathies/pathology , Adipocytes/metabolism , Adipocytes/pathology , Adipogenesis/physiology , Obesity/metabolism
7.
Cell Tissue Res ; 388(1): 63-73, 2022 Apr.
Article in English | MEDLINE | ID: mdl-35072792

ABSTRACT

Pericytes are pluripotent cells that enclose the endothelium of small blood vessels in the whole body. These cells are thought to play a limited role in vascular development and blood pressure regulation; however, current evidence from numerous studies suggests several significant biologic aspects of pericytes in animals. One viewpoint is that pericytes are also known as potential cellular origin of multiple soft tissue tumors. Experimental evidence of the cellular origin of pericytic tumors is still insufficient, however, and their molecular pathogenesis is poorly understood. Here, we used a conditional constitutively active Smoothened allele (Rosa-SmoM2) and Cre recombinase mice to activate hedgehog (Hh) signaling, exclusively in the monocyte/macrophage and osteoclast lineage (LysMcre) or in RANK expressing cells (RANKcre) that are recognized as osteoclast precursor cells. Mice conditionally expressing SmoM2 with LysMcre displayed no significant skeletal phenotype; surprisingly, however, RANKcre; Rosa-SmoM2 mice frequently developed progressive soft tissue tumors in regions of the leg. Genetic lineage tracing analysis uncovered a new domain of RANKcre-expressing cells in the skeletal muscle interstitial cells that display markers consistent with vascular pericytes. Neoplasms arising from these cells showed increased expression of Matrix metalloproteinases (MMPs) that are molecular indicators of malignancy. Moreover, the tumors displayed strong bone invasive potency associated with osteoclastic bone resorption. Thus, these findings provide a novel insight into tumor pathology: Hh signal activated-pericytes can be a potential cellular origin of multiple soft tissue tumors.


Subject(s)
Pericytes , Soft Tissue Neoplasms , Animals , Disease Models, Animal , Hedgehog Proteins/metabolism , Mice , Pericytes/metabolism , Signal Transduction , Soft Tissue Neoplasms/pathology
8.
Front Cell Dev Biol ; 9: 728057, 2021.
Article in English | MEDLINE | ID: mdl-34589491

ABSTRACT

In the aorta-gonad-mesonephros (AGM) region of mouse embryos, pre-hematopoietic stem cells (pre-HSCs) are generated from rare and specialized hemogenic endothelial cells (HECs) via endothelial-to-hematopoietic transition, followed by maturation into bona fide hematopoietic stem cells (HSCs). As HECs also generate a lot of hematopoietic progenitors not fated to HSCs, powerful tools that are pre-HSC/HSC-specific become urgently critical. Here, using the gene knockin strategy, we firstly developed an Hlf-tdTomato reporter mouse model and detected Hlf-tdTomato expression exclusively in the hematopoietic cells including part of the immunophenotypic CD45- and CD45+ pre-HSCs in the embryonic day (E) 10.5 AGM region. By in vitro co-culture together with long-term transplantation assay stringent for HSC precursor identification, we further revealed that unlike the CD45- counterpart in which both Hlf-tdTomato-positive and negative sub-populations harbored HSC competence, the CD45+ E10.5 pre-HSCs existed exclusively in Hlf-tdTomato-positive cells. The result indicates that the cells should gain the expression of Hlf prior to or together with CD45 to give rise to functional HSCs. Furthermore, we constructed a novel Hlf-CreER mouse model and performed time-restricted genetic lineage tracing by a single dose induction at E9.5. We observed the labeling in E11.5 AGM precursors and their contribution to the immunophenotypic HSCs in fetal liver (FL). Importantly, these Hlf-labeled early cells contributed to and retained the size of the HSC pool in the bone marrow (BM), which continuously differentiated to maintain a balanced and long-term multi-lineage hematopoiesis in the adult. Therefore, we provided another valuable mouse model to specifically trace the fate of emerging HSCs during development.

9.
Cells ; 10(7)2021 07 06.
Article in English | MEDLINE | ID: mdl-34359886

ABSTRACT

Cardiac fibrosis is the excess deposition of extracellular matrix (ECM), such as collagen. Myofibroblasts are major players in the production of collagen, and are differentiated primarily from resident fibroblasts. Collagen can compensate for the dead cells produced by injury. The appropriate production of collagen is beneficial for preserving the structural integrity of the heart, and protects the heart from cardiac rupture. However, excessive deposition of collagen causes cardiac dysfunction. Recent studies have demonstrated that myofibroblasts can change their phenotypes. In addition, myofibroblasts are found to have functions other than ECM production. Myofibroblasts have macrophage-like functions, in which they engulf dead cells and secrete anti-inflammatory cytokines. Research into fibroblasts has been delayed due to the lack of selective markers for the identification of fibroblasts. In recent years, it has become possible to genetically label fibroblasts and perform sequencing at single-cell levels. Based on new technologies, the origins of fibroblasts and myofibroblasts, time-dependent changes in fibroblast states after injury, and fibroblast heterogeneity have been demonstrated. In this paper, recent advances in fibroblast and myofibroblast research are reviewed.


Subject(s)
Endomyocardial Fibrosis/pathology , Extracellular Matrix/pathology , Fibroblasts/pathology , Macrophages/pathology , Myofibroblasts/pathology , Animals , Cardiotonic Agents/therapeutic use , Cell Differentiation , Cell Lineage , Collagen/genetics , Collagen/metabolism , Cytokines/genetics , Cytokines/metabolism , Discoidin Domain Receptor 2/genetics , Discoidin Domain Receptor 2/metabolism , Endomyocardial Fibrosis/drug therapy , Endomyocardial Fibrosis/genetics , Endomyocardial Fibrosis/metabolism , Extracellular Matrix/drug effects , Extracellular Matrix/metabolism , Fibroblasts/classification , Fibroblasts/drug effects , Fibroblasts/metabolism , Gene Expression Regulation , Humans , Macrophages/drug effects , Macrophages/metabolism , Myocardium/metabolism , Myocardium/pathology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Myofibroblasts/drug effects , Myofibroblasts/metabolism , Signal Transduction
10.
J Gastroenterol Hepatol ; 36(12): 3418-3428, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34151462

ABSTRACT

BACKGROUND AND AIM: In chronic hepatic diseases where treatment strategies are not available, deposited fibrotic tissues deteriorate the intrinsic regeneration capacity of the liver by creating special restrictions. Thus, if the anti-fibrosis modality is efficiently applied, the regeneration capacity of the liver should be reactivated even in such refractory hepatic diseases. METHODS: Rat liver fibrosis was induced by dimethyl-nitrosamine (DMN). Another liver fibrosis model was established in CCl4 treated Sox9CreERT2ROSA26: YFP mice. To resolve hepatic fibrosis, vitamin A-coupled liposomes containing siRNA HSP47 (VA-liposome siHSP47) were employed. EpCAM + hepatic progenitor cells from GFP rats were transplanted to DMN rat liver to examine their trans-differentiation into hepatic cells after resolution of liver fibrosis. RESULTS: Even under continuous exposure to such strong hepatotoxin as DMN, rats undergoing VA-liposome siHSP47 treatment showed an increment of DNA synthesis of hepatocytes with the concomitant restoration of impaired liver weight and normalization of albumin levels. These results were consistent with the observation that GFP + EpCAM hepatic progenitor cells transplanted to DMN rat liver, trans-differentiated into GFP + mature hepatic cells after VA-liposome siHSP47 treatment. Another rodent model also proved regeneration potential of the fibrotic liver in CCl4 administered Sox9CreERT2ROSA26: YFP mice, VA-liposome siHSP47 treatment-induced restoration of liver weight and trans-differentiation of YEP + Sox9 + cells into YFP + hepatic cells, although because of relatively mild hepatotoxicity of CCl4, undamaged hepatocytes also proliferated. CONCLUSIONS: These results demonstrated that regeneration of chronically damaged liver indeed occurs after anti-fibrosis treatment even under continuous exposure to hepatotoxin, which promises a significant benefit of the anti-fibrosis therapy for refractory liver diseases.


Subject(s)
Liposomes , Liver Cirrhosis , RNA, Small Interfering , Vitamin A , Animals , Fibrosis , Liposomes/pharmacology , Liver Cirrhosis/drug therapy , Liver Cirrhosis/pathology , Liver Regeneration/drug effects , Lung Injury/pathology , Mice , RNA, Small Interfering/pharmacology , Rats , Treatment Outcome , Vitamin A/pharmacology
11.
Int J Mol Sci ; 22(3)2021 Jan 26.
Article in English | MEDLINE | ID: mdl-33530552

ABSTRACT

The reproductive tract in mammals emerges from two ductal systems during embryogenesis: Wolffian ducts (WDs) and Mullerian ducts (MDs). Most of the female reproductive tract (FRT) including the oviducts, uterine horn and cervix, originate from MDs. It is widely accepted that the formation of MDs depends on the preformed WDs within the urogenital primordia. Here, we found that the WD mesenchyme under the regulation of Hedgehog (Hh) signaling is closely related to the developmental processes of the FRT during embryonic and postnatal periods. Deficiency of Sonic hedgehog (Shh), the only Hh ligand expressed exclusively in WDs, prevents the MD mesenchyme from affecting uterine growth along the radial axis. The in vivo cell tracking approach revealed that after WD regression, distinct cells responding to WD-derived Hh signal continue to exist in the developing FRT and gradually contribute to the formation of various tissues such as smooth muscle, endometrial stroma and vascular vessel, in the mouse uterus. Our study thus provides a novel developmental mechanism of FRT relying on WD.


Subject(s)
Genitalia, Female/embryology , Genitalia, Female/metabolism , Hedgehog Proteins/metabolism , Organogenesis , Signal Transduction , Uterus/embryology , Uterus/metabolism , Animals , Biomarkers , Cell Differentiation , Female , Gene Expression Regulation, Developmental , Immunohistochemistry , Mice , Mice, Knockout , Models, Biological , Mullerian Ducts/embryology , Mullerian Ducts/metabolism , Organogenesis/genetics
12.
Elife ; 102021 01 04.
Article in English | MEDLINE | ID: mdl-33393460

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) is a devastating disease. Although rigorous efforts identified the presence of 'cancer stem cells (CSCs)' in PDAC and molecular markers for them, stem cell dynamics in vivo have not been clearly demonstrated. Here we focused on Doublecortin-like kinase 1 (Dclk1), known as a CSC marker of PDAC. Using genetic lineage tracing with a dual-recombinase system and live imaging, we showed that Dclk1+ tumor cells continuously provided progeny cells within pancreatic intraepithelial neoplasia, primary and metastatic PDAC, and PDAC-derived spheroids in vivo and in vitro. Furthermore, genes associated with CSC and epithelial mesenchymal transition were enriched in mouse Dclk1+ and human DCLK1-high PDAC cells. Thus, we provided direct functional evidence for the stem cell activity of Dclk1+ cells in vivo, revealing the essential roles of Dclk1+ cells in expansion of pancreatic neoplasia in all progressive stages.


Subject(s)
Adenocarcinoma/physiopathology , Carcinoma, Pancreatic Ductal/physiopathology , Cell Lineage/genetics , Neoplastic Stem Cells/metabolism , Pancreatic Neoplasms/physiopathology , Adenocarcinoma/pathology , Animals , Carcinoma, Pancreatic Ductal/pathology , Humans , Mice , Neoplasm Metastasis , Pancreatic Neoplasms/pathology
13.
Methods Mol Biol ; 2158: 323-336, 2021.
Article in English | MEDLINE | ID: mdl-32857384

ABSTRACT

Genetic lineage tracing is accomplished using bi-transgenic mice, where one allele is altered to express Cre recombinase, and another allele encodes a Cre-dependent genetic reporter protein. Once Cre is activated (constitutive or in response to tamoxifen), the marker gene-expressing cells become indelibly labeled by the reporter protein. Therefore, daughter cells derived from labeled cells are permanently labeled even if the marker gene that drove Cre recombinase expression is no longer expressed in these cells. This system is commonly used to label putative progenitor cells and determine the fate of their progeny. Here, we describe the use of c-kit-based genetic lineage-tracing mouse line as an example and discuss caveats for performing these types of experiments.


Subject(s)
Cell Lineage/genetics , Cell Tracking/methods , Stem Cells/chemistry , Stem Cells/metabolism , Animals , Gene Expression , Genes, Reporter , Genetic Linkage , Green Fluorescent Proteins/genetics , Integrases/genetics , Integrases/metabolism , Mice , Mice, Transgenic , Models, Animal , Myocytes, Cardiac/chemistry , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Proto-Oncogene Proteins c-kit/analysis , Proto-Oncogene Proteins c-kit/genetics , Stem Cells/cytology , Tamoxifen/pharmacology
14.
Front Physiol ; 11: 601923, 2020.
Article in English | MEDLINE | ID: mdl-33192618

ABSTRACT

During embryonic development, symmetric ectodermal thickenings [olfactory placodes (OP)] give rise to several cell types that comprise the olfactory system, such as those that form the terminal nerve ganglion (TN), gonadotropin releasing hormone-1 neurons (GnRH-1ns), and other migratory neurons in rodents. Even though the genetic heterogeneity among these cell types is documented, unidentified cell populations arising from the OP remain. One candidate to identify placodal derived neurons in the developing nasal area is the transcription factor Isl1, which was recently identified in GnRH-3 neurons of the terminal nerve in fish, as well as expression in neurons of the nasal migratory mass (MM). Here, we analyzed the Isl1 genetic lineage in chemosensory neuronal populations in the nasal area and migratory GnRH-1ns in mice using in situ hybridization, immunolabeling a Tamoxifen inducible Isl1CreERT and a constitutive Isl1Cre knock-in mouse lines. In addition, we also performed conditional Isl1 ablation in developing GnRH neurons. We found Isl1 lineage across non-sensory cells of the respiratory epithelium and sustentacular cells of OE and VNO. We identified a population of transient embryonic Isl1 + neurons in the olfactory epithelium and sparse Isl1 + neurons in postnatal VNO. Isl1 is expressed in almost all GnRH neurons and in approximately half of the other neuron populations in the MM. However, Isl1 conditional ablation alone does not significantly compromise GnRH-1 neuronal migration or GnRH-1 expression, suggesting compensatory mechanisms. Further studies will elucidate the functional and mechanistic role of Isl1 in development of migratory endocrine neurons.

15.
J Genet Genomics ; 46(10): 489-498, 2019 10 20.
Article in English | MEDLINE | ID: mdl-31776062

ABSTRACT

The functional heterogeneity of hematopoietic stem cells (HSCs) has been comprehensively investigated by single-cell transplantation assay. However, the heterogeneity regarding their physiological contribution remains an open question, especially for those with life-long hematopoietic fate of rigorous self-renewing and balanced differentiation capacities. In this study, we revealed that Procr expression was detected principally in phenotypical vascular endothelium co-expressing Dll4 and CD44 in the mid-gestation mouse embryos, and could enrich all the HSCs of the embryonic day 11.5 (E11.5) aorta-gonad-mesonephros (AGM) region. We then used a temporally restricted genetic tracing strategy to irreversibly label the Procr-expressing cells at E9.5. Interestingly, most labeled mature HSCs in multiple sites (such as AGM) around E11.5 were functionally categorized as lymphomyeloid-balanced HSCs assessed by direct transplantation. Furthermore, the labeled cells contributed to an average of 7.8% of immunophenotypically defined HSCs in E14.5 fetal liver (FL) and 6.9% of leukocytes in peripheral blood (PB) during one-year follow-up. Surprisingly, in aged mice of 24 months, the embryonically tagged cells displayed constant contribution to leukocytes with no bias to myeloid or lymphoid lineages. Altogether, we demonstrated, for the first time, the existence of a subtype of physiologically long-lived balanced HSCs as hypothesized, whose precise embryonic origin and molecular identity await further characterization.


Subject(s)
Endothelial Protein C Receptor/metabolism , Hematopoietic Stem Cells/metabolism , Animals , Aorta/cytology , Aorta/metabolism , Embryo, Mammalian , Endothelial Protein C Receptor/genetics , Female , Hematopoiesis/genetics , Hematopoiesis/physiology , Hematopoietic Stem Cells/cytology , Male , Mesonephros/cytology , Mesonephros/metabolism , Mice , Platelet Endothelial Cell Adhesion Molecule-1/genetics , Platelet Endothelial Cell Adhesion Molecule-1/metabolism
17.
J Mol Cell Cardiol ; 129: 92-104, 2019 04.
Article in English | MEDLINE | ID: mdl-30771308

ABSTRACT

Serum response factor (SRF) and the SRF co-activators myocardin-related transcription factors (MRTFs) are essential for epicardium-derived progenitor cell (EPDC)-mobilization during heart development; however, the impact of developmental EPDC deficiencies on adult cardiac physiology has not been evaluated. Here, we utilize the Wilms Tumor-1 (Wt1)-Cre to delete Mrtfs or Srf in the epicardium, which reduced the number of EPDCs in the adult cardiac interstitium. Deficiencies in Wt1-lineage EPDCs prevented the development of cardiac fibrosis and diastolic dysfunction in aged mice. Mice lacking MRTF or SRF in EPDCs also displayed preservation of cardiac function following myocardial infarction partially due to the depletion of Wt1 lineage-derived cells in the infarct. Interestingly, depletion of Wt1-lineage EPDCs allows for the population of the infarct with a Wt1-negative cell lineage with a reduced fibrotic profile. Taken together, our study conclusively demonstrates the contribution of EPDCs to both ischemic cardiac remodeling and the development of diastolic dysfunction in old age, and reveals the existence of an alternative Wt1-negative source of resident fibroblasts that can populate the infarct.


Subject(s)
Aging/pathology , Fibroblasts/pathology , Myocardial Ischemia/pathology , Pericardium/pathology , Animals , Cell Lineage , Diastole , Fibrosis , Heart/physiopathology , Mice, Knockout , Myocardial Ischemia/physiopathology , Serum Response Factor/metabolism , Stem Cells/metabolism , Trans-Activators/metabolism , Ventricular Remodeling , WT1 Proteins/metabolism
18.
Vasc Biol ; 1(1): H67-H73, 2019.
Article in English | MEDLINE | ID: mdl-32923956

ABSTRACT

Vascular remodeling is a complex and dynamic pathological process engaging many different cell types that reside within the vasculature. Mesenchymal stromal/stem cells (MSCs) refer to a heterogeneous cell population with the plasticity to differentiate toward multiple mesodermal lineages. Various types of MSC have been identified within the vascular wall that actively contribute to the vascular remodeling process such as atherosclerosis. With the advances of genetic mouse models, recent findings demonstrated the crucial roles of MSCs in the progression of vascular diseases. This review aims to provide an overview on the current knowledge of the characteristics and behavior of vascular resident MSCs under quiescence and remodeling conditions, which may lead to the development of novel therapeutic approaches for cardiovascular diseases.

19.
Sichuan Da Xue Xue Bao Yi Xue Ban ; 49(4): 560-565, 2018 Jul.
Article in Chinese | MEDLINE | ID: mdl-30378310

ABSTRACT

OBJECTIVE: To establish Gli1-CreERt2; tdTomato genetic lineage-tracing mice for studies on hepatic fibrosis. METHODS: Offspring of ROSA26 td Tomato (tdTomato) mice and Gli1-CreERt2 mice (Gli1 mice) were obtained, with Gli1-CreERt2; tdTomato genotype being identified by PCR. The mice model of hepatic fibrosis was induced with CCl4. Their liver samples were taken. The formalin-fixed and paraffin-embedded samples were prepared for HE staining and Masson staining. The expression of tdTomato was observed under immunofluorescent microscope. RESULTS: An ideal number of Gli1-CreERt2; tdTomato genetic lineage-tracing mice were harvested. The differences in fertility between the parental and the offspring mice were not significant (P>0.05). Pseudolobular formation occurred in the CCl4-induced hepatic fibrosis model mice. Enhanced red fluoresce was observed in the model mice. CONCLUSION: Gli1-CreERt2; tdTomato genetic lineage-tracing mice can be used to monitor the cell source of fibrous tissues, its transition as well as the underlying mechanism of pathogenesis of hepatic fibrosis.


Subject(s)
Liver Cirrhosis/genetics , Zinc Finger Protein GLI1/genetics , Animals , Liver Cirrhosis/pathology , Mice , Mice, Transgenic
20.
Development ; 145(19)2018 10 02.
Article in English | MEDLINE | ID: mdl-30201686

ABSTRACT

The vertebrate body forms by continuous generation of new tissue from progenitors at the posterior end of the embryo. The study of these axial progenitors has proved to be challenging in vivo largely because of the lack of unique molecular markers to identify them. Here, we elucidate the expression pattern of the transcription factor Nkx1-2 in the mouse embryo and show that it identifies axial progenitors throughout body axis elongation, including neuromesodermal progenitors and early neural and mesodermal progenitors. We create a tamoxifen-inducible Nkx1-2CreERT2 transgenic mouse and exploit the conditional nature of this line to uncover the lineage contributions of Nkx1-2-expressing cells at specific stages. We show that early Nkx1-2-expressing epiblast cells contribute to all three germ layers, mostly neuroectoderm and mesoderm, excluding notochord. Our data are consistent with the presence of some self-renewing axial progenitors that continue to generate neural and mesoderm tissues from the tail bud. This study identifies Nkx1-2-expressing cells as the source of most trunk and tail tissues in the mouse and provides a useful tool to genetically label and manipulate axial progenitors in vivo.


Subject(s)
Cell Lineage , Homeodomain Proteins/metabolism , Integrases/metabolism , Nuclear Proteins/metabolism , Stem Cells/cytology , Tail/embryology , Torso/embryology , Transcription Factors/metabolism , Animals , Body Patterning , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Genes, Reporter , Mesoderm/cytology , Mice, Inbred C57BL , Mice, Transgenic , Neurons/cytology , Neurons/metabolism , SOXB1 Transcription Factors/metabolism , Tail/cytology
SELECTION OF CITATIONS
SEARCH DETAIL