Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 75
Filter
1.
EMBO Rep ; 24(7): e56030, 2023 Jul 05.
Article in English | MEDLINE | ID: mdl-37154714

ABSTRACT

Intestinal ischemia underlies several clinical conditions and can result in the loss of the intestinal mucosal barrier. Ischemia-induced damage to the intestinal epithelium is repaired by stimulation of intestinal stem cells (ISCs), and paracrine signaling from the vascular niche regulates intestinal regeneration. Here, we identify FOXC1 and FOXC2 as essential regulators of paracrine signaling in intestinal regeneration after ischemia-reperfusion (I/R) injury. Vascular endothelial cell (EC)- and lymphatic EC (LEC)-specific deletions of Foxc1, Foxc2, or both in mice worsen I/R-induced intestinal damage by causing defects in vascular regrowth, expression of chemokine CXCL12 and Wnt activator R-spondin 3 (RSPO3) in blood ECs (BECs) and LECs, respectively, and activation of Wnt signaling in ISCs. Both FOXC1 and FOXC2 directly bind to regulatory elements of the CXCL12 and RSPO3 loci in BECs and LECs, respectively. Treatment with CXCL12 and RSPO3 rescues the I/R-induced intestinal damage in EC- and LEC-Foxc mutant mice, respectively. This study provides evidence that FOXC1 and FOXC2 are required for intestinal regeneration by stimulating paracrine CXCL12 and Wnt signaling.


Subject(s)
Intestines , Reperfusion Injury , Mice , Animals , Endothelial Cells/metabolism , Wnt Signaling Pathway , Intestinal Mucosa , Reperfusion Injury/genetics , Reperfusion Injury/metabolism
2.
J Cell Sci ; 135(19)2022 10 01.
Article in English | MEDLINE | ID: mdl-36073617

ABSTRACT

Nephron development proceeds with reciprocal interactions among three layers: nephron progenitors (NPs), ureteric buds and stromal progenitors (SPs). We found that Foxc1 and Foxc2 (Foxc1/2) are expressed in NPs and SPs. Systemic deletion of Foxc1/2 2 days after the onset of metanephros development (embryonic day 13.5) resulted in the epithelialization of NPs and ectopic formation of renal vesicles. NP-specific deletion did not cause these phenotypes, indicating that Foxc1/2 in other cells (likely in SPs) contributed to the maintenance of NPs. Single-cell RNA-sequencing analysis revealed the existence of NP and SP subpopulations, the border between committed NPs and renewing NPs, and similarity between the cortical interstitium and vascular smooth muscle type cells. Integrated analysis of the control and Foxc1/2 knockout data indicated transformation of some NPs to strange cells expressing markers of the vascular endothelium, reduced numbers of self-renewing NP and SP populations, and downregulation of crucial genes for kidney development, such as Fgf20 and Frem1 in NPs, and Foxd1 and Sall1 in SPs. It also revealed upregulation of genes that were not usually expressed in NPs and SPs. Thus, Foxc1/2 maintain NPs and SPs by regulating the expression of multiple genes.


Subject(s)
Forkhead Transcription Factors , Nephrons , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Kidney/metabolism , Nephrons/metabolism , Organogenesis , RNA/metabolism
3.
Cell ; 139(3): 597-609, 2009 Oct 30.
Article in English | MEDLINE | ID: mdl-19879845

ABSTRACT

Extrinsic signals controlling generation of neocortical neurons during embryonic life have been difficult to identify. In this study we demonstrate that the dorsal forebrain meninges communicate with the adjacent radial glial endfeet and influence cortical development. We took advantage of Foxc1 mutant mice with defects in forebrain meningeal formation. Foxc1 dosage and loss of meninges correlated with a dramatic reduction in both neuron and intermediate progenitor production and elongation of the neuroepithelium. Several types of experiments demonstrate that retinoic acid (RA) is the key component of this secreted activity. In addition, Rdh10- and Raldh2-expressing cells in the dorsal meninges were either reduced or absent in the Foxc1 mutants, and Rdh10 mutants had a cortical phenotype similar to the Foxc1 null mutants. Lastly, in utero RA treatment rescued the cortical phenotype in Foxc1 mutants. These results establish RA as a potent, meningeal-derived cue required for successful corticogenesis.


Subject(s)
Meninges/metabolism , Neurogenesis , Neurons/cytology , Tretinoin/metabolism , Animals , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , In Vitro Techniques , Mice , Prosencephalon/cytology , Prosencephalon/metabolism
4.
Exp Eye Res ; 234: 109599, 2023 09.
Article in English | MEDLINE | ID: mdl-37488009

ABSTRACT

Limbal epithelial stem cells are not only critical for corneal epithelial homeostasis but also have the capacity to change from a relatively quiescent mitotic phenotype to a rapidly proliferating cell in response to population depletion following corneal epithelial wounding. Pax6+/- mice display many abnormalities including corneal vascularization and these aberrations are consistent with a limbal stem cell deficiency (LSCD) phenotype. FoxC1 has an inhibitory effect on corneal avascularity and a positive role in stem cell maintenance in many tissues. However, the role of FoxC1 in limbal epithelial stem cells remains unknown. To unravel FoxC1's role(s) in limbal epithelial stem cell homeostasis, we utilized an adeno-associated virus (AAV) vector to topically deliver human FOXC1 proteins into Pax6 +/- mouse limbal epithelium. Under unperturbed conditions, overexpression of FOXC1 in the limbal epithelium had little significant change in differentiation (PAI-2, Krt12) and proliferation (BrdU, Ki67). Conversely, such overexpression resulted in a marked increase in the expression of putative limbal epithelial stem cell markers, N-cadherin and Lrig1. After corneal injuries in Pax6 +/- mice, FOXC1 overexpression enhanced the behavior of limbal epithelial stem cells from quiescence to a highly proliferative status. Overall, the treatment of AAV8-FOXC1 may be beneficial to the function of limbal epithelial stem cells in the context of a deficiency of Pax6 function.


Subject(s)
Corneal Diseases , Epithelium, Corneal , Limbus Corneae , Animals , Humans , Mice , Cornea , Corneal Diseases/metabolism , Debridement , Epithelial Cells , Epithelium, Corneal/metabolism , Limbus Corneae/metabolism , Stem Cells
5.
J Am Soc Nephrol ; 32(6): 1323-1337, 2021 06 01.
Article in English | MEDLINE | ID: mdl-33771836

ABSTRACT

BACKGROUND: Transcriptional programs control cell fate, and identifying their components is critical for understanding diseases caused by cell lesion, such as podocytopathy. Although many transcription factors (TFs) are necessary for cell-state maintenance in glomeruli, their roles in transcriptional regulation are not well understood. METHODS: The distribution of H3K27ac histones in human glomerulus cells was analyzed to identify superenhancer-associated TFs, and ChIP-seq and transcriptomics were performed to elucidate the regulatory roles of the TFs. Transgenic animal models of disease were further investigated to confirm the roles of specific TFs in podocyte maintenance. RESULTS: Superenhancer distribution revealed a group of potential TFs in core regulatory circuits in human glomerulus cells, including FOXC1/2, WT1, and LMX1B. Integration of transcriptome and cistrome data of FOXC1/2 in mice resolved transcriptional regulation in podocyte maintenance. FOXC1/2 regulated differentiation-associated transcription in mature podocytes. In both humans and animal models, mature podocyte injury was accompanied by deregulation of FOXC1/2 expression, and FOXC1/2 overexpression could protect podocytes in zebrafish. CONCLUSIONS: FOXC1/2 maintain podocyte differentiation through transcriptional stabilization. The genome-wide chromatin resources support further investigation of TFs' regulatory roles in glomeruli transcription programs.


Subject(s)
Forkhead Transcription Factors/genetics , Podocytes/physiology , Transcription Factors/genetics , Transcription, Genetic , Animals , Cell Differentiation/genetics , Chromosome Mapping , Disease Models, Animal , Forkhead Transcription Factors/metabolism , Gene Expression Profiling , Gene Expression Regulation , Histones , Humans , Kidney Diseases/genetics , Kidney Diseases/metabolism , LIM-Homeodomain Proteins/genetics , LIM-Homeodomain Proteins/metabolism , Mice , Mice, Knockout , Mice, Transgenic , Podocytes/pathology , Transcription Factors/metabolism , Transcriptome , WT1 Proteins/genetics , WT1 Proteins/metabolism , Zebrafish , Zebrafish Proteins/genetics
6.
Dev Dyn ; 250(8): 1125-1139, 2021 08.
Article in English | MEDLINE | ID: mdl-33667029

ABSTRACT

BACKGROUND: Foxc2 is a member of the winged helix/forkhead (Fox) box family of transcription factors. Loss of function of Foxc2 causes craniofacial abnormalities such as cleft palate and deformed cranial base, but its role during craniofacial development remains to be elucidated. RESULTS: The contributions of Foxc2-positive and its descendant cells to the craniofacial structure at E18.5 were examined using a tamoxifen-inducible Cre driver mouse (Foxc2-CreERT2) crossed with the R26R-LacZ reporter mouse. Foxc2 expression at E8.5 is restricted to the cranial mesenchyme, contributing to specific components including the cranial base, sensory capsule, tongue, upper incisor, and middle ear. Expression at E10.5 was still positively regulated in most of those regions. In situ hybridization analysis of Foxc2 and its closely related gene, Foxc1, revealed that expression domains of these genes largely overlap in the cephalic mesenchyme. Meanwhile, the tongue expressed Foxc2 but not Foxc1, and its development was affected by the neural crest-specific deletion of Foxc2 in mice (Wnt1-Cre; Foxc2fl/fl ). CONCLUSIONS: Foxc2 is expressed in cranial mesenchyme that contributes to specific craniofacial tissue components from an early stage, and it seems to be involved in their development in cooperation with Foxc1. Foxc2 also has its own role in tongue development.


Subject(s)
Cell Lineage/genetics , Craniofacial Abnormalities/genetics , Forkhead Transcription Factors/genetics , Gene Expression Regulation, Developmental , Organogenesis/genetics , Animals , Craniofacial Abnormalities/metabolism , Forkhead Transcription Factors/metabolism , Mice , Mice, Transgenic , Neural Crest/embryology , Neural Crest/metabolism
7.
Genesis ; 58(7): e23364, 2020 07.
Article in English | MEDLINE | ID: mdl-32259372

ABSTRACT

Cardiac neural crest cells (cNCCs) are required for normal heart development. cNCCs are a multipotent and migratory cell lineage that differentiates into multiple cell types. cNCCs migrate into the developing heart to contribute to the septation of the cardiac outflow tract (OFT). Foxc1 and Foxc2 are closely related members of the FOX (Forkhead box) transcription factor family and are expressed in cNCC during heart development. However, the precise role of Foxc1 and Foxc2 in cNCCs has yet to be fully described. We found that compound NCC-specific Foxc1;Foxc2 mutant embryos exhibited persistent truncus arteriosus (PTA), ventricular septal defects (VSDs), and thinning of the ventricular myocardium. Loss of Foxc1/c2 expression in cNCCs resulted in abnormal patterns of cNCC migration into the OFT without the formation of the aorticopulmonary septum. Further, loss of Foxc1 expression in cNCCs resulted in normal OFT development but abnormal ventricular septal formation. In contrast, loss of Foxc2 expression in NCCs led to no obvious cardiac abnormalities. Together, we provide evidence that Foxc1 and Foxc2 in cNCCs are cooperatively required for proper cNCC migration, the formation of the OFT septation, and the development of the ventricles. Our data also suggests that Foxc1 expression may play a larger role in ventricular development compared to Foxc2.


Subject(s)
Forkhead Transcription Factors/genetics , Neural Crest/metabolism , Truncus Arteriosus, Persistent/genetics , Animals , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Heart Ventricles/abnormalities , Heart Ventricles/growth & development , Heart Ventricles/metabolism , Mice , Mice, Inbred C57BL , Mutation , Neural Crest/cytology , Neural Crest/growth & development , Truncus Arteriosus, Persistent/pathology
8.
Nature ; 508(7497): 536-40, 2014 Apr 24.
Article in English | MEDLINE | ID: mdl-24590069

ABSTRACT

Haematopoietic stem and progenitor cells are maintained by special microenvironments known as niches in bone marrow. Many studies have identified diverse candidate cells that constitute niches for haematopoietic stem cells in the marrow, including osteoblasts, endothelial cells, Schwann cells, α-smooth muscle actin-expressing macrophages and mesenchymal progenitors such as CXC chemokine ligand (CXCL)12-abundant reticular (CAR) cells, stem cell factor-expressing cells, nestin-expressing cells and platelet-derived growth factor receptor-α (PDGFR-α)(+)Sca-1(+)CD45(-)Ter119(-) (PαS) cells. However, the molecular basis of the formation of the niches remains unclear. Here we find that the transcription factor Foxc1 is preferentially expressed in the adipo-osteogenic progenitor CAR cells essential for haematopoietic stem and progenitor cell maintenance in vivo in the developing and adult bone marrow. When Foxc1 was deleted in all marrow mesenchymal cells or CAR cells, from embryogenesis onwards, osteoblasts appeared normal, but haematopoietic stem and progenitor cells were markedly reduced and marrow cavities were occupied by adipocytes (yellow adipose marrow) with reduced CAR cells. Inducible deletion of Foxc1 in adult mice depleted haematopoietic stem and progenitor cells and reduced CXCL12 and stem cell factor expression in CAR cells but did not induce a change to yellow marrow. These data suggest a role for Foxc1 in inhibiting adipogenic processes in CAR progenitors. Foxc1 might also promote CAR cell development, upregulating CXCL12 and stem cell factor expression. This study identifies Foxc1 as a specific transcriptional regulator essential for development and maintenance of the mesenchymal niches for haematopoietic stem and progenitor cells.


Subject(s)
Forkhead Transcription Factors/metabolism , Hematopoietic Stem Cells/cytology , Stem Cell Niche/physiology , Adipocytes/cytology , Adipocytes/metabolism , Animals , Bone Marrow Cells/cytology , Cell Count , Cell Differentiation , Chemokine CXCL12/metabolism , Embryonic Development/genetics , Forkhead Transcription Factors/deficiency , Forkhead Transcription Factors/genetics , Hematopoietic Stem Cells/metabolism , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Mice , Osteoblasts/cytology , Osteoblasts/metabolism , Stem Cell Factor/metabolism
9.
Development ; 143(5): 872-9, 2016 Mar 01.
Article in English | MEDLINE | ID: mdl-26839363

ABSTRACT

Pax3 and Foxc2 have been shown genetically to mutually repress each other in the mouse somite. Perturbation of this balance in multipotent cells of the dermomyotome influences cell fate; upregulation of Foxc2 favours a vascular fate, whereas higher levels of Pax3 lead to myogenesis. Foxc1 has overlapping functions with Foxc2. In Foxc1/2 double-mutant embryos, somitogenesis is severely affected, precluding analysis of somite derivatives. We have adopted a conditional approach whereby mutations in Foxc1 and Foxc2 genes were targeted to Pax3-expressing cells. Inclusion of a conditional reporter allele in the crosses made it possible to follow cells that had expressed Pax3. At the forelimb level, endothelial and myogenic cells migrate from adjacent somites into the limb bud. This population of endothelial cells is compromised in the double mutant, whereas excessive production of myogenic cells is observed in the trunk. However, strikingly, myogenic progenitors fail to enter the limbs, leading to the absence of skeletal muscle. Pax3-positive migratory myogenic progenitors, marked by expression of Lbx1, are specified in the somite at forelimb level, but endothelial progenitors are absent. The myogenic progenitors do not die, but differentiate prematurely adjacent to the somite. We conclude that the small proportion of somite-derived endothelial cells in the limb is required for the migration of myogenic limb progenitors.


Subject(s)
Endothelial Cells/metabolism , Forelimb/embryology , Forkhead Transcription Factors/genetics , Muscle Development/physiology , Paired Box Transcription Factors/metabolism , Somites/metabolism , Animals , Cell Movement , Cell Separation , Crosses, Genetic , Female , Flow Cytometry , Forelimb/metabolism , Gene Expression Regulation, Developmental , Genes, Reporter , In Situ Hybridization , Limb Buds/embryology , Male , Mice , Mice, Transgenic , Muscle Proteins/genetics , Mutation , PAX3 Transcription Factor , Paired Box Transcription Factors/genetics , Phenotype
10.
Biol Pharm Bull ; 42(10): 1609-1619, 2019.
Article in English | MEDLINE | ID: mdl-31582649

ABSTRACT

The vascular system forms the largest surface in our body, serving as a critical interface between blood circulation and our diverse organ/tissue environments. Thus, the vascular system performs a gatekeeper function for organ/tissue homeostasis and the body's adjustment to pathological challenges. The endothelium, as the most inner layer of the vasculature, regulates the tissue microenvironment, which is critical for development, hemostatic balance, inflammation, and angiogenesis, with a role as well in tumor malignancy and metastasis. These multitudinous functions are primarily mediated by organ/tissue-specifically differentiated endothelial cells, in which heterogeneity has long been recognized at the molecular and histological level. Based on these general principles of vascular-bed heterogeneity and characterization, this review largely covers landmark discoveries regarding organ/tissue microenvironment-governed endothelial cell phenotypic changes. These involve the physical features of continuous, discontinuous, fenestrated, and sinusoidal endothelial cells, in addition to the more specialized endothelial cell layers of the lymphatic system, glomerulus, tumors, and the blood brain barrier (BBB). Major signal pathways of endothelial specification are outlined, including Notch as a key factor of tip/stalk- and arterial-endothelial cell differentiation. We also denote the shear stress sensing machinery used to convey blood flow-mediated biophysical forces that are indispensable to maintaining inert and mature endothelial phenotypes. Since our circulatory system is among the most fundamental and emergent targets of study in pharmacology from the viewpoint of drug metabolism and delivery, a better molecular understanding of organ vasculature-bed heterogeneity may lead to better strategies for novel vascular-targeted treatments to fight against hitherto intractable diseases.


Subject(s)
Endothelial Cells , Organ Specificity , Animals , Disease , Endothelium, Vascular , Health , Humans
11.
Proc Natl Acad Sci U S A ; 113(11): E1506-15, 2016 Mar 15.
Article in English | MEDLINE | ID: mdl-26912458

ABSTRACT

Adult tissue stem cells (SCs) reside in niches, which orchestrate SC behavior. SCs are typically used sparingly and exist in quiescence unless activated for tissue growth. Whether parsimonious SC use is essential to conserve long-term tissue-regenerating potential during normal homeostasis remains poorly understood. Here, we examine this issue by conditionally ablating a key transcription factor Forkhead box C1 (FOXC1) expressed in hair follicle SCs (HFSCs). FOXC1-deficient HFSCs spend less time in quiescence, leading to markedly shortened resting periods between hair cycles. The enhanced hair cycling accelerates HFSC expenditure, and impacts hair regeneration in aging mice. Interestingly, although FOXC1-deficient HFs can still form a new bulge that houses HFSCs for the next hair cycle, the older bulge is left unanchored. As the new hair emerges, the entire old bulge, including its reserve HFSCs and SC-inhibitory inner cell layer, is lost. We trace this mechanism first, to a marked increase in cell cycle-associated transcripts upon Foxc1 ablation, and second, to a downstream reduction in E-cadherin-mediated inter-SC adhesion. Finally, we show that when the old bulge is lost with each hair cycle, overall levels of SC-inhibitory factors are reduced, further lowering the threshold for HFSC activity. Taken together, our findings suggest that HFSCs have restricted potential in vivo, which they conserve by coupling quiescence to adhesion-mediated niche maintenance, thereby achieving long-term tissue homeostasis.


Subject(s)
Adult Stem Cells/physiology , Forkhead Transcription Factors/metabolism , Hair Follicle/cytology , Hair Follicle/physiology , Adult Stem Cells/metabolism , Aging , Animals , Cadherins/metabolism , Cell Adhesion/physiology , Cell Proliferation , Forkhead Transcription Factors/genetics , Gene Expression Regulation , Mice, Knockout , Mice, Mutant Strains , Regeneration , Stem Cell Niche
12.
Dev Dyn ; 247(12): 1286-1296, 2018 12.
Article in English | MEDLINE | ID: mdl-30376688

ABSTRACT

BACKGROUND: Proper development of the great vessels of the heart and septation of the cardiac outflow tract requires cardiac neural crest cells. These cells give rise to the parasympathetic cardiac ganglia, the smooth muscle layer of the great vessels, some cardiomyocytes, and the conotruncal cushions and aorticopulmonary septum of the outflow tract. Ablation of cardiac neural crest cells results in defective patterning of each of these structures. Previous studies have shown that targeted deletion of the forkhead transcription factor C2 (Foxc2), results in cardiac phenotypes similar to that derived from cardiac neural crest cell ablation. RESULTS: We report that Foxc2-/- embryos on the 129s6/SvEv inbred genetic background display persistent truncus arteriosus and hypoplastic ventricles before embryonic lethality. Foxc2 loss-of-function resulted in perturbed cardiac neural crest cell migration and their reduced contribution to the outflow tract as evidenced by lineage tracing analyses together with perturbed expression of the neural crest cell markers Sox10 and Crabp1. Foxc2 loss-of-function also resulted in alterations in PlexinD1, Twist1, PECAM1, and Hand1/2 expression in association with vascular and ventricular defects. CONCLUSIONS: Our data indicate Foxc2 is required for proper migration of cardiac neural crest cells, septation of the outflow tract, and development of the ventricles. Developmental Dynamics 247:1286-1296, 2018. © 2018 Wiley Periodicals, Inc.


Subject(s)
Embryo, Mammalian , Forkhead Transcription Factors/physiology , Neural Crest/cytology , Animals , Cell Movement , Coronary Vessels/embryology , Coronary Vessels/growth & development , Heart/innervation , Heart Ventricles/embryology , Heart Ventricles/growth & development , Mice , Myocardium/cytology , Neural Crest/embryology , Organogenesis
13.
J Physiol ; 596(8): 1397-1417, 2018 04 15.
Article in English | MEDLINE | ID: mdl-29380370

ABSTRACT

KEY POINTS: The mechanisms by which bacteria alter endothelial cell phenotypes and programme inflammatory angiogenesis remain unclear. In lung endothelial cells, we demonstrate that toll-like receptor 4 (TLR4) signalling induces activation of forkhead box protein C2 (FOXC2), a transcriptional factor implicated in lymphangiogenesis and endothelial specification, in an extracellular signal-regulated kinase (ERK)-dependent manner. TLR4-ERK-FOXC2 signalling regulates expression of the Notch ligand DLL4 and signals inflammatory angiogenesis in vivo and in vitro. Our work reveals a novel link between endothelial immune signalling (TLR pathway) and a vascular transcription factor, FOXC2, that regulates embryonic vascular development. This mechanism is likely to be relevant to pathological angiogenesis complicating inflammatory diseases in humans. ABSTRACT: Endothelial cells (ECs) mediate a specific and robust immune response to bacteria in sepsis through the activation of toll-like receptor (TLR) signalling. The mechanisms by which bacterial ligands released during sepsis programme EC specification and altered angiogenesis remain unclear. We postulated that the forkhead box protein C2 (FOXC2) transcriptional factor directs EC cell-fate decisions and angiogenesis during TLR signalling. In human lung ECs, lipopolysaccharide (LPS) induced ERK phosphorylation, FOXC2, and delta-like 4 (DLL4, the master regulator of sprouting angiogenesis expression) in a TLR4-dependent manner. LPS-mediated ERK phosphorylation resulted in FOXC2-ERK protein ligation, ERK-dependent FOXC2 serine and threonine phosphorylation, and subsequent activation of DLL4 gene expression. Chemical inhibition of ERK or ERK-2 dominant negative transfection disrupted LPS-mediated FOXC2 phosphorylation and transcriptional activation of FOXC2. FOXC2-siRNA or ERK-inhibition attenuated LPS-induced DLL4 expression and angiogenic sprouting in vitro. In vivo, intraperitoneal LPS induced ERK and FOXC2 phosphorylation, FOXC2 binding to DLL4 promoter, and FOXC2/DLL4 expression in the lung. ERK-inhibition suppressed LPS-induced FOXC2 phosphorylation, FOXC2-DLL4 promoter binding, and induction of FOXC2 and DLL4 in mouse lung ECs. LPS induced aberrant retinal angiogenesis and DLL4 expression in neonatal mice, which was attenuated with ERK inhibition. FOXC2+/- mice treated with LPS showed a mitigated increase in FOXC2 and DLL4 compared to FOXC2+/+ mice. These data reveal a new mechanism (TLR4-ERK-FOXC2-DLL4) by which sepsis-induced EC TLR signalling programmes EC specification and altered angiogenesis.


Subject(s)
Endothelial Cells/immunology , Forkhead Transcription Factors/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Neovascularization, Physiologic , Signal Transduction , Toll-Like Receptor 4/metabolism , Adaptor Proteins, Signal Transducing , Animals , Calcium-Binding Proteins , Cell Differentiation , Cells, Cultured , Endothelial Cells/cytology , Endothelial Cells/drug effects , Humans , Intracellular Signaling Peptides and Proteins/genetics , Lipopolysaccharides/toxicity , Lung/blood supply , Lung/embryology , Lung/metabolism , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism
14.
Exp Cell Res ; 352(2): 265-272, 2017 03 15.
Article in English | MEDLINE | ID: mdl-28223138

ABSTRACT

Foxc1 and Foxc2 (Foxc1/2) are transcription factors involved in many biological processes. In adult kidneys, expression of Foxc1/2 is confined to the glomerular epithelial cells, i.e., podocytes. To bypass embryonic lethality of Foxc1/2 null mice, mice ubiquitously expressing inducible-Cre (ROSA26-CreERT2) or mice expressing Cre in podocytes (Nephrin-Cre) were mated with floxed-Foxc1 and floxed-Foxc2 mice. The CreERT2 was activated in adult mice by administrations of tamoxifen. Eight weeks after tamoxifen treatment, ROSA26-CreERT2; Foxc1+/flox; Foxc2flox/flox mice developed microalbuminuria, while ROSA26-Cre ERT2; Foxc1flox/flox; Foxc2+/flox mice had no microalbuminuria. The kidneys of conditional-Foxc1/2 null mice showed proteinaceous casts, protein reabsorption droplets in tubules and huge vacuoles in podocytes, indicating severe podocyte injury and massive proteinuria. Comparison of gene expression profiles revealed that Foxc1/2 maintain expression of genes necessary for podocyte function such as podocin and Cxcl12. In addition, mice with an innate podocyte-specific deletion of Foxc1/2 by Nephrin-Cre develop similar podocyte injury. These results demonstrate dose-dependence of Foxc1/2 gene in maintaining the podocyte with a more critical role for Foxc2 than Foxc1 and a critical role of Foxc1/2 in regulating expression of genes that maintain podocyte integrity.


Subject(s)
Albuminuria/metabolism , Forkhead Transcription Factors/metabolism , Kidney Glomerulus/metabolism , Podocytes/metabolism , Animals , Cells, Cultured , Chemokine CXCL12/metabolism , Forkhead Transcription Factors/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Kidney Glomerulus/cytology , Kidney Glomerulus/pathology , Membrane Proteins/metabolism , Mice , Podocytes/pathology
15.
Blood ; 125(9): 1418-26, 2015 Feb 26.
Article in English | MEDLINE | ID: mdl-25587036

ABSTRACT

Hematopoietic and vascular development share many common features, including cell surface markers and sites of origin. Recent lineage-tracing studies have established that definitive hematopoietic stem and progenitor cells arise from vascular endothelial-cadherin(+) hemogenic endothelial cells of the aorta-gonad-mesonephros region, but the genetic programs underlying the specification of hemogenic endothelial cells remain poorly defined. Here, we discovered that Notch induction enhances hematopoietic potential and promotes the specification of hemogenic endothelium in differentiating cultures of mouse embryonic stem cells, and we identified Foxc2 as a highly upregulated transcript in the hemogenic endothelial population. Studies in zebrafish and mouse embryos revealed that Foxc2 and its orthologs are required for the proper development of definitive hematopoiesis and function downstream of Notch signaling in the hemogenic endothelium. These data establish a pathway linking Notch signaling to Foxc2 in hemogenic endothelial cells to promote definitive hematopoiesis.


Subject(s)
Embryonic Stem Cells/cytology , Endothelium, Vascular/cytology , Forkhead Transcription Factors/metabolism , Gene Expression Regulation, Developmental , Hematopoiesis/physiology , Hematopoietic Stem Cells/cytology , Receptor, Notch1/metabolism , Animals , Apoptosis , Blotting, Western , Cell Differentiation , Cell Lineage , Cell Proliferation , Cells, Cultured , Embryonic Stem Cells/metabolism , Endothelium, Vascular/metabolism , Forkhead Transcription Factors/genetics , Hematopoietic Stem Cells/metabolism , Mice , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Receptor, Notch1/genetics , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Zebrafish/embryology , Zebrafish/genetics , Zebrafish/metabolism
16.
Stem Cells ; 34(6): 1487-500, 2016 06.
Article in English | MEDLINE | ID: mdl-26824887

ABSTRACT

Embryonic Stem Cells (ESCs) hold great potential for regeneration of damaged myocardium, however the molecular circuitry that guides ESC differentiation into cardiomyocytes remains poorly understood. This is exemplified by the elusive role of the transcription factor, Foxc1, during cardiac development. The only known Foxc1 target during heart development is Tbx1. Because Foxc1 null mice contain heart mutations that are far more severe than Tbx1 null mice, it is likely that Foxc1 has additional regulatory roles during heart development. The goal of our study was to test whether Foxc1 is critical for ESC differentiation into functional cardiomyocytes through proper regulation of specific downstream gene networks. Converging evidence from Foxc1 deficient and overexpression ESC models reveals a close relationship between Foxc1 levels and early cardiomyogenic factors Isl1, Mef2c, and Nkx2.5 and also the production of functional cardiomyocytes. We show Foxc1 regulates early cardiomyogenesis during a specific window of differentiation, D4-D6. Through whole transcriptome RNA-sequencing analysis, we report pathways regulated by Foxc1 involved in cardiac function including actin cytoskeleton, cell adhesion, tight and gap junctions, and calcium signaling. Our data indicate a novel Foxc1 direct gene target, Myh7, which encodes the predominant myosin heavy chain isoform, MHCß, expressed during cardiac development. These data lead us to conclude that Foxc1 regulates both early cardiomyogenesis and the functional properties of ESC-derived cardiomyocytes. Our findings shed light on the molecular circuitry governing cardiomyogenesis that may lead to the development of better translational strategies for the use of pluripotent stem cells in regenerative medicine towards repairing damaged myocardium. Stem Cells 2016;34:1487-1500.


Subject(s)
Forkhead Transcription Factors/metabolism , Mouse Embryonic Stem Cells/cytology , Mouse Embryonic Stem Cells/metabolism , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Organogenesis , Animals , Cell Differentiation/drug effects , Cell Differentiation/genetics , Doxycycline/pharmacology , Endoderm/drug effects , Endoderm/metabolism , Forkhead Transcription Factors/deficiency , Homeobox Protein Nkx-2.5/metabolism , Mesoderm/drug effects , Mesoderm/metabolism , Mice , Mouse Embryonic Stem Cells/drug effects , Myocytes, Cardiac/drug effects , Myosin Heavy Chains/genetics , Myosin Heavy Chains/metabolism , Organogenesis/drug effects , Organogenesis/genetics , Sequence Analysis, RNA , Transcriptome/genetics , Up-Regulation/drug effects , Up-Regulation/genetics
17.
Dev Dyn ; 245(7): 751-61, 2016 07.
Article in English | MEDLINE | ID: mdl-26890576

ABSTRACT

Heart disease is the number one cause of morbidity and mortality in the world and is a major health and economic burden, costing the United States Health Care System more than $200 billion annually. A major cause of heart disease is the massive loss or dysfunction of cardiomyocytes caused by myocardial infarctions and hypertension. Due to the limited regenerative capacity of the heart, much research has focused on better understanding the process of differentiation toward cardiomyocytes. This review will highlight what is currently known about cardiac cell specification during mammalian development, areas of controversy, cellular sources of cardiomyocytes, and current and potential uses of stem cell derived cardiomyocytes for cardiac therapies. Developmental Dynamics 245:751-761, 2016. © 2016 Wiley Periodicals, Inc.


Subject(s)
Adult Stem Cells/cytology , Myocytes, Cardiac/cytology , Adult Stem Cells/metabolism , Adult Stem Cells/physiology , Animals , Cell Differentiation/genetics , Cell Differentiation/physiology , Cellular Reprogramming/genetics , Cellular Reprogramming/physiology , Humans , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/physiology
18.
Cells Tissues Organs ; 201(5): 380-9, 2016.
Article in English | MEDLINE | ID: mdl-27193493

ABSTRACT

Foxc1 and Foxc2 play key roles in mouse development. Foxc1 mutant mice develop duplex kidneys with double ureters, and lack calvarial and sternal bones. Foxc2 null mice have been reported to have glomerular abnormalities in the kidney and axial skeletal anomalies. Expression patterns of Foxc1 and Foxc2 overlap extensively and are believed to have interactive roles. However, cooperative roles of these factors in glomerular and skeletal development are unknown. Therefore, we examined the kidneys and skeleton of mice that were double heterozygous for Foxc1 and Foxc2. Double heterozygotes were generated by mating single heterozygotes for Foxc1 and Foxc2. Newborn double heterozygous mice showed many anomalies in the kidney and urinary tract resembling Foxc1 phenotypes, including duplex kidneys, double ureters, hydronephrosis and mega-ureter. Some mice had hydronephrosis alone. In addition to these macroscopic anomalies, some mice had abnormal glomeruli and disorganized glomerular capillaries observed in Foxc2 phenotypes. Interestingly, these mice also showed glomerular cysts not observed in the single-gene knockout of either Foxc1 or Foxc2 but observed in conditional knockout of Foxc2 in the kidney. Serial section analysis revealed that all cystic glomeruli were connected to proximal tubules, precluding the possibility of atubular glomeruli resulting in cyst formation. Dorsally opened vertebral arches and malformations of sternal bones in the double heterozygotes were phenotypes similar to Foxc1 null mice. Absent or split vertebral bodies in the double heterozygotes were phenotypes similar to Foxc2 null mice, whilst hydrocephalus noted in the Foxc1 phenotype was not observed. Thus, Foxc1 and Foxc2 have a role in kidney and axial skeleton development. These transcription factors might interact in the regulation of the embryogenesis of these organs.


Subject(s)
Bone and Bones/pathology , Forkhead Transcription Factors/metabolism , Kidney/pathology , Animals , Bone and Bones/abnormalities , Bone and Bones/metabolism , Choristoma/pathology , Heterozygote , Kidney/abnormalities , Kidney/metabolism , Kidney Diseases, Cystic/pathology , Kidney Glomerulus/pathology , Kidney Tubules/pathology , Mesangial Cells/pathology , Mice, Knockout , Phenotype
19.
PLoS Genet ; 9(12): e1003949, 2013.
Article in English | MEDLINE | ID: mdl-24385915

ABSTRACT

Syngnathia (bony fusion of the upper and lower jaw) is a rare human congenital condition, with fewer than sixty cases reported in the literature. Syngnathia typically presents as part of a complex syndrome comprising widespread oral and maxillofacial anomalies, but it can also occur in isolation. Most cartilage, bone, and connective tissue of the head and face is derived from neural crest cells. Hence, congenital craniofacial anomalies are often attributed to defects in neural crest cell formation, survival, migration, or differentiation. The etiology and pathogenesis of syngnathia however remains unknown. Here, we report that Foxc1 null embryos display bony syngnathia together with defects in maxillary and mandibular structures, and agenesis of the temporomandibular joint (TMJ). In the absence of Foxc1, neural crest cell derived osteogenic patterning is affected, as osteoblasts develop ectopically in the maxillary prominence and fuse with the dentary bone. Furthermore, we observed that the craniofacial musculature is also perturbed in Foxc1 null mice, which highlights the complex tissue interactions required for proper jaw development. We present evidence that Foxc1 and Fgf8 genetically interact and that Fgf8 dosage is associated with variation in the syngnathic phenotype. Together our data demonstrates that Foxc1 - Fgf8 signaling regulates mammalian jaw patterning and provides a mechanistic basis for the pathogenesis of syngnathia. Furthermore, our work provides a framework for understanding jaw patterning and the etiology of other congenital craniofacial anomalies, including temporomandibular joint agenesis.


Subject(s)
Body Patterning/genetics , Fibroblast Growth Factor 8/genetics , Forkhead Transcription Factors/genetics , Maxillofacial Abnormalities/genetics , Animals , Cartilage/growth & development , Cartilage/pathology , Embryo, Mammalian , Embryonic Development/genetics , Fibroblast Growth Factor 8/metabolism , Forkhead Transcription Factors/metabolism , Humans , Jaw/pathology , Maxillofacial Abnormalities/pathology , Mice , Neural Crest/growth & development , Neural Crest/pathology , Protein Interaction Maps/genetics , Temporomandibular Joint/growth & development , Temporomandibular Joint/pathology
20.
Proc Natl Acad Sci U S A ; 109(6): 2015-20, 2012 Feb 07.
Article in English | MEDLINE | ID: mdl-22171010

ABSTRACT

Normal vision requires the precise control of vascular growth to maintain corneal transparency. Here we provide evidence for a unique mechanism by which the Forkhead box transcription factor FoxC1 regulates corneal vascular development. Murine Foxc1 is essential for development of the ocular anterior segment, and in humans, mutations have been identified in Axenfeld-Rieger syndrome, a disorder characterized by anterior segment dysgenesis. We show that FOXC1 mutations also lead to corneal angiogenesis, and that mice homozygous for either a global (Foxc1(-/-)) or neural crest (NC)-specific (NC-Foxc1(-/-)) null mutation display excessive growth of corneal blood and lymphatic vessels. This is associated with disorganization of the extracellular matrix and increased expression of multiple matrix metalloproteinases. Heterozygous mutants (Foxc1(+/-) and NC-Foxc1(+/-)) exhibit milder phenotypes, such as disrupted limbal vasculature. Moreover, environmental exposure to corneal injury significantly increases growth of both blood and lymphatic vessels in both Foxc1(+/-) and NC-Foxc1(+/-) mice compared with controls. Notably, this amplification of the angiogenic response is abolished by inhibition of VEGF receptor 2. Collectively, these findings identify a role for FoxC1 in inhibiting corneal angiogenesis, thereby maintaining corneal transparency by regulating VEGF signaling.


Subject(s)
Blood Vessels/growth & development , Cornea/blood supply , Forkhead Transcription Factors/metabolism , Alkalies , Animals , Anterior Eye Segment/abnormalities , Anterior Eye Segment/metabolism , Anterior Eye Segment/pathology , Blood Vessels/pathology , Burns/pathology , Cornea/pathology , Corneal Stroma/enzymology , Corneal Stroma/pathology , Eye Abnormalities/complications , Eye Abnormalities/metabolism , Eye Abnormalities/pathology , Eye Diseases, Hereditary , Haploinsufficiency/genetics , Heterozygote , Lymphangiogenesis , Matrix Metalloproteinases/metabolism , Mice , Mutation/genetics , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Neural Crest/metabolism , Neural Crest/pathology , Phenotype , Pupil , Signal Transduction , Vascular Endothelial Growth Factor A/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL