Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters










Publication year range
1.
J Clin Invest ; 134(8)2024 Mar 11.
Article in English | MEDLINE | ID: mdl-38466355

ABSTRACT

Craniofacial anomalies, especially midline facial defects, are among the most common birth defects in patients and are associated with increased mortality or require lifelong treatment. During mammalian embryogenesis, specific instructions arising at genetic, signaling, and metabolic levels are important for stem cell behaviors and fate determination, but how these functionally relevant mechanisms are coordinated to regulate craniofacial morphogenesis remain unknown. Here, we report that bone morphogenetic protein (BMP) signaling in cranial neural crest cells (CNCCs) is critical for glycolytic lactate production and subsequent epigenetic histone lactylation, thereby dictating craniofacial morphogenesis. Elevated BMP signaling in CNCCs through constitutively activated ACVR1 (ca-ACVR1) suppressed glycolytic activity and blocked lactate production via a p53-dependent process that resulted in severe midline facial defects. By modulating epigenetic remodeling, BMP signaling-dependent lactate generation drove histone lactylation levels to alter essential genes of Pdgfra, thus regulating CNCC behavior in vitro as well as in vivo. These findings define an axis wherein BMP signaling controls a metabolic/epigenetic cascade to direct craniofacial morphogenesis, thus providing a conceptual framework for understanding the interaction between genetic and metabolic cues operative during embryonic development. These findings indicate potential preventive strategies of congenital craniofacial birth defects via modulating metabolic-driven histone lactylation.


Subject(s)
Face , Histones , Animals , Humans , Epigenesis, Genetic , Histones/genetics , Histones/metabolism , Lactates/metabolism , Mammals/metabolism , Morphogenesis , Neural Crest
2.
bioRxiv ; 2023 Nov 22.
Article in English | MEDLINE | ID: mdl-38045303

ABSTRACT

Adipocytes have diverse roles in energy storage and metabolism, inflammation, and tissue repair. Mature adipocytes have been assumed to be terminally differentiated cells. However, recent evidence suggests that adipocytes retain substantial phenotypic plasticity, with potential to dedifferentiate into fibroblast-like cells under physiological and pathological conditions. Here, we develop a two-step lineage tracing approach based on the observation that fibroblasts express platelet-derived growth factor receptor alpha ( Pdgfra ) while adipocytes express Adiponectin ( Adipoq ) but not Pdgfra . Our approach specifically traces Pdgfra + cells that originate from Adipoq + adipocytes. We find many traced adipocytes and fibroblast-like cells surrounding skin wounds, but only a few traced cells localize to the wound center. In agreement with adipocyte plasticity, traced adipocytes incorporate EdU, downregulate Plin1 and PPARγ, and upregulate αSMA. We also investigate the role of potential dedifferentiation signals using constitutively active PDGFRα mutation, Pdgfra knockout, or Tgfbr2 knockout models. We find that PDGF and TGFß signaling both promote dedifferentiation, and PDGFRα does so independently of TGFßR2. These results demonstrate an intersectional genetic approach to trace the hybrid cell phenotype of Pdgfra + adipocytes, which may be important for wound repair, regeneration and fibrosis.

3.
Circ Res ; 133(6): 463-480, 2023 09.
Article in English | MEDLINE | ID: mdl-37555328

ABSTRACT

BACKGROUND: Cardiac valve disease is observed in 2.5% of the general population and 10% of the elderly people. Effective pharmacological treatments are currently not available, and patients with severe cardiac valve disease require surgery. PROX1 (prospero-related homeobox transcription factor 1) and FOXC2 (Forkhead box C2 transcription factor) are transcription factors that are required for the development of lymphatic and venous valves. We found that PROX1 and FOXC2 are expressed in a subset of valvular endothelial cells (VECs) that are located on the downstream (fibrosa) side of cardiac valves. Whether PROX1 and FOXC2 regulate cardiac valve development and disease is not known. METHODS: We used histology, electron microscopy, and echocardiography to investigate the structure and functioning of heart valves from Prox1ΔVEC mice in which Prox1 was conditionally deleted from VECs. Isolated valve endothelial cells and valve interstitial cells were used to identify the molecular mechanisms in vitro, which were tested in vivo by RNAScope, additional mouse models, and pharmacological approaches. The significance of our findings was tested by evaluation of human samples of mitral valve prolapse and aortic valve insufficiency. RESULTS: Histological analysis revealed that the aortic and mitral valves of Prox1ΔVEC mice become progressively thick and myxomatous. Echocardiography revealed that the aortic valves of Prox1ΔVEC mice are stenotic. FOXC2 was downregulated and PDGF-B (platelet-derived growth factor-B) was upregulated in the VECs of Prox1ΔVEC mice. Conditional knockdown of FOXC2 and conditional overexpression of PDGF-B in VECs recapitulated the phenotype of Prox1ΔVEC mice. PDGF-B was also increased in mice lacking FOXC2 and in human mitral valve prolapse and insufficient aortic valve samples. Pharmacological inhibition of PDGF-B signaling with imatinib partially ameliorated the valve defects of Prox1ΔVEC mice. CONCLUSIONS: PROX1 antagonizes PDGF-B signaling partially via FOXC2 to maintain the extracellular matrix composition and prevent myxomatous degeneration of cardiac valves.


Subject(s)
Heart Valve Diseases , Mitral Valve Prolapse , Animals , Humans , Mice , Endothelial Cells/metabolism , Heart Valve Diseases/genetics , Heart Valve Diseases/prevention & control , Heart Valve Diseases/metabolism , Mitral Valve/metabolism , Mitral Valve Prolapse/metabolism , Transcription Factors/metabolism , Proto-Oncogene Proteins c-sis/metabolism
4.
Development ; 149(19)2022 10 01.
Article in English | MEDLINE | ID: mdl-36245218

ABSTRACT

Periodontal tissue supports teeth in the alveolar bone socket via fibrous attachment of the periodontal ligament (PDL). The PDL contains periodontal fibroblasts and stem/progenitor cells, collectively known as PDL cells (PDLCs), on top of osteoblasts and cementoblasts on the surface of alveolar bone and cementum, respectively. However, the characteristics and lineage hierarchy of each cell type remain poorly defined. This study identified periodontal ligament associated protein-1 (Plap-1) as a PDL-specific extracellular matrix protein. We generated knock-in mice expressing CreERT2 and GFP specifically in Plap-1-positive PDLCs. Genetic lineage tracing confirmed the long-standing hypothesis that PDLCs differentiate into osteoblasts and cementoblasts. A PDL single-cell atlas defined cementoblasts and osteoblasts as Plap-1-Ibsp+Sparcl1+ and Plap-1-Ibsp+Col11a2+, respectively. Other populations, such as Nes+ mural cells, S100B+ Schwann cells, and other non-stromal cells, were also identified. RNA velocity analysis suggested that a Plap-1highLy6a+ cell population was the source of PDLCs. Lineage tracing of Plap-1+ PDLCs during periodontal injury showed periodontal tissue regeneration by PDLCs. Our study defines diverse cell populations in PDL and clarifies the role of PDLCs in periodontal tissue homeostasis and repair.


Subject(s)
Periodontal Ligament , Transcriptome , Animals , Calcium-Binding Proteins/metabolism , Cell Differentiation/genetics , Extracellular Matrix Proteins/metabolism , Mice , Osteoblasts , RNA/metabolism
5.
Cell Rep ; 40(7): 111192, 2022 08 16.
Article in English | MEDLINE | ID: mdl-35977484

ABSTRACT

Fibroblasts differentiate into myofibroblasts by acquiring new contractile function. This is important for tissue repair, but it also contributes to organ fibrosis. Platelet-derived growth factor (PDGF) promotes tissue repair and fibrosis, but the relationship between PDGF and myofibroblasts is unclear. Using mice with lineage tracing linked to PDGF receptor α (PDGFRα) gene mutations, we examine cell fates during skin wound healing. Elevated PDGFRα signaling increases proliferation but unexpectedly delays the fibroblast-to-myofibroblast transition, suggesting that PDGFRα must be downregulated for myofibroblast differentiation. In contrast, deletion of PDGFRα decreases proliferation and myofibroblast differentiation by reducing serum response factor (SRF) nuclear localization. Consequences of SRF deletion resemble PDGFRα deletion, but deletion of two SRF coactivators, MRTFA and MRTFB, specifically eliminates myofibroblasts. Our findings suggest a scenario where PDGFRα signaling initially supports proliferation of fibroblast progenitors to expand their number during early wound healing but, later, PDGFRα downregulation facilitates fibroblast differentiation into myofibroblasts.


Subject(s)
Myofibroblasts , Receptor, Platelet-Derived Growth Factor alpha , Animals , Cell Differentiation/physiology , Fibroblasts/metabolism , Fibrosis , Mice , Myofibroblasts/pathology , Platelet-Derived Growth Factor/metabolism , Receptor, Platelet-Derived Growth Factor alpha/genetics , Receptor, Platelet-Derived Growth Factor alpha/metabolism , Wound Healing
6.
Development ; 148(23)2021 12 01.
Article in English | MEDLINE | ID: mdl-34738614

ABSTRACT

Autosomal dominant PDGFRß gain-of-function mutations in mice and humans cause a spectrum of wasting and overgrowth disorders afflicting the skeleton and other connective tissues, but the cellular origin of these disorders remains unknown. We demonstrate that skeletal stem cells (SSCs) isolated from mice with a gain-of-function D849V point mutation in PDGFRß exhibit colony formation defects that parallel the wasting or overgrowth phenotypes of the mice. Single-cell RNA transcriptomics with SSC-derived polyclonal colonies demonstrates alterations in osteogenic and chondrogenic precursors caused by PDGFRßD849V. Mutant cells undergo poor osteogenesis in vitro with increased expression of Sox9 and other chondrogenic markers. Mice with PDGFRßD849V exhibit osteopenia. Increased STAT5 phosphorylation and overexpression of Igf1 and Socs2 in PDGFRßD849V cells suggests that overgrowth in mice involves PDGFRßD849V activating the STAT5-IGF1 axis locally in the skeleton. Our study establishes that PDGFRßD849V causes osteopenic skeletal phenotypes that are associated with intrinsic changes in SSCs, promoting chondrogenesis over osteogenesis.


Subject(s)
Gain of Function Mutation , Myoblasts, Skeletal/metabolism , Point Mutation , Receptor, Platelet-Derived Growth Factor beta/metabolism , Amino Acid Substitution , Animals , Chondrogenesis/genetics , Gene Expression Regulation , Mice , Mice, Transgenic , Myoblasts, Skeletal/pathology , Osteogenesis/genetics , Receptor, Platelet-Derived Growth Factor beta/genetics , SOX9 Transcription Factor/genetics , SOX9 Transcription Factor/metabolism , Signal Transduction/genetics
7.
Cell Stem Cell ; 28(6): 989-990, 2021 06 03.
Article in English | MEDLINE | ID: mdl-34087159

ABSTRACT

Specific cell targeting with one site-specific recombinase is challenging. In this issue of Cell Stem Cell, Han et al. (2021) released a collection of Dre drivers and demonstrate how two recombinases can be combined to improve the cell specificity of lineage tracing and gene inactivation in mice.


Subject(s)
Integrases , Recombinases , Animals , Base Sequence , Cell Movement , Integrases/metabolism , Mice , Recombinases/genetics , Recombinases/metabolism
8.
Matrix Biol ; 97: 40-57, 2021 03.
Article in English | MEDLINE | ID: mdl-33454424

ABSTRACT

Little is known about extracellular matrix (ECM) contributions to formation of the earliest cell lineages in the embryo. Here, we show that the proteoglycan versican and glycosaminoglycan hyaluronan are associated with emerging Flk1+ hematoendothelial progenitors at gastrulation. The mouse versican mutant Vcanhdf lacks yolk sac vasculature, with attenuated yolk sac hematopoiesis. CRISPR/Cas9-mediated Vcan inactivation in mouse embryonic stem cells reduced vascular endothelial and hematopoietic differentiation within embryoid bodies, which generated fewer blood colonies, and had an impaired angiogenic response to VEGF165. Hyaluronan was severely depleted in Vcanhdf embryos, with corresponding upregulation of the hyaluronan-depolymerase TMEM2. Conversely, hyaluronan-deficient mouse embryos also had vasculogenic suppression but with increased versican proteolysis. VEGF165 and Indian hedgehog, crucial vasculogenic factors, utilized the versican-hyaluronan matrix, specifically versican chondroitin sulfate chains, for binding. Versican-hyaluronan ECM is thus an obligate requirement for vasculogenesis and primitive hematopoiesis, providing a vasculogenic factor-enriching microniche for Flk1+ progenitors from their origin at gastrulation.


Subject(s)
Extracellular Matrix/metabolism , Hyaluronic Acid/metabolism , Mouse Embryonic Stem Cells/cytology , Vascular Endothelial Growth Factor Receptor-2/metabolism , Versicans/genetics , Animals , CRISPR-Cas Systems , Cell Differentiation , Cells, Cultured , Hedgehog Proteins/metabolism , Hematopoiesis , Membrane Proteins/metabolism , Mice , Mouse Embryonic Stem Cells/metabolism , Stem Cell Niche , Up-Regulation , Versicans/metabolism
9.
Cell Stem Cell ; 26(5): 707-721.e5, 2020 05 07.
Article in English | MEDLINE | ID: mdl-32229310

ABSTRACT

Adipocyte progenitors (APs) express platelet-derived growth factor receptors (PDGFRs), PDGFRα and PDGFRß. Elevated PDGFRα signaling inhibits adipogenesis and promotes fibrosis; however, the function of PDGFRs in APs remains unclear. We combined lineage tracing and functional analyses in a sequential dual-recombinase approach that creates mosaic Pdgfr mutant cells by Cre/lox recombination with a linked Flp/frt reporter to track individual cell fates. Using mosaic lineage labeling, we show that adipocytes are derived from the Pdgfra lineage during postnatal growth and adulthood. In contrast, adipocytes are only derived from the mosaic Pdgfrb lineage during postnatal growth. Functionally, postnatal mosaic deletion of PDGFRα enhances adipogenesis and adult deletion enhances ß3-adrenergic-receptor-induced beige adipocyte formation. Mosaic deletion of PDGFRß also enhances white, brown, and beige adipogenesis. These data show that both PDGFRs are cell-autonomous inhibitors of adipocyte differentiation and implicate downregulation of PDGF signaling as a critical event in the transition from AP to adipocyte.


Subject(s)
Adipogenesis , Receptor, Platelet-Derived Growth Factor alpha , Receptor, Platelet-Derived Growth Factor beta , Adipocytes , Adipogenesis/genetics , Animals , Cell Differentiation/genetics , Gene Knock-In Techniques , Mice , Receptor, Platelet-Derived Growth Factor alpha/genetics , Receptor, Platelet-Derived Growth Factor beta/genetics
10.
Wound Repair Regen ; 28(4): 448-459, 2020 07.
Article in English | MEDLINE | ID: mdl-32175700

ABSTRACT

Signal transducer and activator of transcription 1 (Stat1) is a ubiquitously expressed latent transcription factor that is activated by many cytokines and growth factors. Global Stat1 knockout mice are prone to chemical-induced lung and liver fibrosis, suggesting roles for Stat1 in tissue repair. However, the importance of Stat1 in fibroblast-mediated and vascular smooth muscle cell (VSMC)-mediated injury response has not been directly evaluated in vivo. Here, we focused on two models of tissue repair in conditional Stat1 knockout mice: excisional skin wounding in mice with Stat1 deletion in dermal fibroblasts, and carotid artery ligation in mice with global Stat1 deletion or deletion specific to VSMCs. In the skin model, dermal wounds closed at a similar rate in mice with fibroblast Stat1 deletion and controls, but collagen and α-smooth muscle actin (αSMA) expression were increased in the mutant granulation tissue. Cultured Stat1 -/- and Stat1 +/- dermal fibroblasts exhibited similar αSMA+ stress fiber assembly, collagen gel contraction, proliferation, migration, and growth factor-induced gene expression. In the artery ligation model, there was a significant increase in fibroblast-driven perivascular fibrosis when Stat1 was deleted globally. However, VSMC-driven remodeling and neointima formation were unchanged when Stat1 was deleted specifically in VSMCs. These results suggest an in vivo role for Stat1 as a suppressor of fibroblast mediated, but not VSMC mediated, injury responses, and a suppressor of the myofibroblast phenotype.


Subject(s)
Carotid Arteries/metabolism , Fibroblasts/metabolism , Myocytes, Smooth Muscle/metabolism , Myofibroblasts/metabolism , Re-Epithelialization/genetics , STAT1 Transcription Factor/genetics , Skin/metabolism , Actins/metabolism , Animals , Carotid Artery Injuries/metabolism , Cell Movement/genetics , Cell Proliferation/genetics , Collagen/metabolism , Gene Expression Regulation/genetics , Granulation Tissue/metabolism , Mice , Mice, Knockout , Muscle, Smooth, Vascular/metabolism , Phenotype , Wound Healing/genetics
11.
EMBO Mol Med ; 12(3): e11021, 2020 03 06.
Article in English | MEDLINE | ID: mdl-31943786

ABSTRACT

Kidney fibrosis is characterized by expansion and activation of platelet-derived growth factor receptor-ß (PDGFR-ß)-positive mesenchymal cells. To study the consequences of PDGFR-ß activation, we developed a model of primary renal fibrosis using transgenic mice with PDGFR-ß activation specifically in renal mesenchymal cells, driving their pathological proliferation and phenotypic switch toward myofibroblasts. This resulted in progressive mesangioproliferative glomerulonephritis, mesangial sclerosis, and interstitial fibrosis with progressive anemia due to loss of erythropoietin production by fibroblasts. Fibrosis induced secondary tubular epithelial injury at later stages, coinciding with microinflammation, and aggravated the progression of hypertensive and obstructive nephropathy. Inhibition of PDGFR activation reversed fibrosis more effectively in the tubulointerstitium compared to glomeruli. Gene expression signatures in mice with PDGFR-ß activation resembled those found in patients. In conclusion, PDGFR-ß activation alone is sufficient to induce progressive renal fibrosis and failure, mimicking key aspects of chronic kidney disease in humans. Our data provide direct proof that fibrosis per se can drive chronic organ damage and establish a model of primary fibrosis allowing specific studies targeting fibrosis progression and regression.


Subject(s)
Kidney Diseases , Receptor, Platelet-Derived Growth Factor beta/metabolism , Animals , Fibroblasts/pathology , Fibrosis , Humans , Kidney/pathology , Kidney Diseases/pathology , Mice , Mice, Transgenic , Myofibroblasts/pathology
12.
Cell Rep ; 25(3): 571-584.e5, 2018 10 16.
Article in English | MEDLINE | ID: mdl-30332639

ABSTRACT

Wnt/ß-catenin signaling is necessary for lymphatic vascular development. Oscillatory shear stress (OSS) enhances Wnt/ß-catenin signaling in cultured lymphatic endothelial cells (LECs) to induce expression of the lymphedema-associated transcription factors GATA2 and FOXC2. However, the mechanisms by which OSS regulates Wnt/ß-catenin signaling and GATA2 and FOXC2 expression are unknown. We show that OSS activates autocrine Wnt/ß-catenin signaling in LECs in vitro. Tissue-specific deletion of Wntless, which is required for the secretion of Wnt ligands, reveals that LECs and vascular smooth muscle cells are complementary sources of Wnt ligands that regulate lymphatic vascular development in vivo. Further, the LEC master transcription factor PROX1 forms a complex with ß-catenin and the TCF/LEF transcription factor TCF7L1 to enhance Wnt/ß-catenin signaling and promote FOXC2 and GATA2 expression in LECs. Thus, our work defines Wnt sources, reveals that PROX1 directs cell fate by acting as a Wnt signaling component, and dissects the mechanisms of PROX1 and Wnt synergy.


Subject(s)
Endothelial Cells/cytology , Gene Expression Regulation, Developmental , Homeodomain Proteins/metabolism , Muscle, Smooth, Vascular/cytology , Tumor Suppressor Proteins/metabolism , Wnt Proteins/metabolism , beta Catenin/metabolism , Animals , Cells, Cultured , Endothelial Cells/metabolism , Female , Forkhead Transcription Factors/metabolism , GATA2 Transcription Factor/metabolism , Humans , Male , Mice , Mice, Inbred C57BL , Muscle, Smooth, Vascular/metabolism , Transcription Factor 7-Like 1 Protein/metabolism , Wnt Signaling Pathway
13.
Genes Dev ; 31(16): 1666-1678, 2017 08 15.
Article in English | MEDLINE | ID: mdl-28924035

ABSTRACT

Platelet-derived growth factor (PDGF) acts through two conserved receptor tyrosine kinases: PDGFRα and PDGFRß. Gain-of-function mutations in human PDGFRB have been linked recently to genetic diseases characterized by connective tissue wasting (Penttinen syndrome) or overgrowth (Kosaki overgrowth syndrome), but it is unclear whether PDGFRB mutations alone are responsible. Mice with constitutive PDGFRß signaling caused by a kinase domain mutation (D849V) develop lethal autoinflammation. Here we used a genetic approach to investigate the mechanism of autoinflammation in Pdgfrb+/D849V mice and test the hypothesis that signal transducer and activator of transcription 1 (STAT1) mediates this phenotype. We show that Pdgfrb+/D849V mice with Stat1 knockout (Stat1-/-Pdgfrb+/D849V ) are rescued from autoinflammation and have improved life span compared with Stat1+/-Pdgfrb+/D849V mice. Furthermore, PDGFRß-STAT1 signaling suppresses PDGFRß itself. Thus, Stat1-/-Pdgfrb+/D849V fibroblasts exhibit increased PDGFRß signaling, and mice develop progressive overgrowth, a distinct phenotype from the wasting seen in Stat1+/-Pdgfrb+/D849V mice. Deletion of interferon receptors (Ifnar1 or Ifngr1) does not rescue wasting in Pdgfrb+/D849V mice, indicating that interferons are not required for autoinflammation. These results provide functional evidence that elevated PDGFRß signaling causes tissue wasting or overgrowth reminiscent of human genetic syndromes and that the STAT1 pathway is a crucial modulator of this phenotypic spectrum.


Subject(s)
Growth Disorders/genetics , Mutation , Receptor, Platelet-Derived Growth Factor beta/genetics , STAT1 Transcription Factor/genetics , Adipose Tissue/pathology , Animals , Aorta/pathology , Atrophy , Bone and Bones/abnormalities , Female , Fibroblasts/metabolism , Fibrosis , Growth Disorders/metabolism , Growth Disorders/pathology , Hyperplasia , Inflammation/metabolism , Interferons/physiology , Male , Mice , Mice, Knockout , Muscle, Smooth, Vascular/pathology , NIH 3T3 Cells , Phenotype , Receptor, Platelet-Derived Growth Factor beta/metabolism , STAT1 Transcription Factor/metabolism , Signal Transduction , Skin/pathology
14.
Development ; 144(1): 83-94, 2017 01 01.
Article in English | MEDLINE | ID: mdl-28049691

ABSTRACT

Adipose tissue is distributed in depots throughout the body with specialized roles in energy storage and thermogenesis. PDGFRα is a marker of adipocyte precursors, and increased PDGFRα activity causes adipose tissue fibrosis in adult mice. However, the function of PDGFRα during adipose tissue organogenesis is unknown. Here, by analyzing mice with juxtamembrane or kinase domain point mutations that increase PDGFRα activity (V561D or D842V), we found that PDGFRα activation inhibits embryonic white adipose tissue organogenesis in a tissue-autonomous manner. By lineage tracing analysis, we also found that collagen-expressing precursor fibroblasts differentiate into white adipocytes in the embryo. PDGFRα inhibited the formation of adipocytes from these precursors while favoring the formation of stromal fibroblasts. This imbalance between adipocytes and stromal cells was accompanied by overexpression of the cell fate regulator Zfp521. PDGFRα activation also inhibited the formation of juvenile beige adipocytes in the inguinal fat pad. Our data highlight the importance of balancing stromal versus adipogenic cell expansion during white adipose tissue development, with PDGFRα activity coordinating this crucial process in the embryo.


Subject(s)
Adipocytes/physiology , Adipogenesis/genetics , Adipose Tissue/embryology , Organogenesis/genetics , Receptor, Platelet-Derived Growth Factor alpha/physiology , Stromal Cells/physiology , Adipose Tissue/growth & development , Adipose Tissue/physiology , Amino Acid Substitution , Animals , Animals, Newborn , Cell Lineage/genetics , Cells, Cultured , Embryo, Mammalian , Female , Lipodystrophy/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Point Mutation , Pregnancy
15.
Kidney Int ; 91(3): 642-657, 2017 03.
Article in English | MEDLINE | ID: mdl-27914705

ABSTRACT

Pigment epithelium-derived factor (PEDF) expression is downregulated in the kidneys of diabetic rats, and delivery of PEDF suppressed renal fibrotic factors in these animals. PEDF has multiple functions including anti-angiogenic, anti-inflammatory and antifibrotic activities. Since the mechanism underlying its antifibrotic effect remains unclear, we studied this in several murine models of renal disease. Renal PEDF levels were significantly reduced in genetic models of type 1 and type 2 diabetes (Akita and db/db, respectively), negatively correlating with Wnt signaling activity in the kidneys. In unilateral ureteral obstruction, an acute renal injury model, there were significant decreases of renal PEDF levels. The kidneys of PEDF knockout mice with ureteral obstruction displayed exacerbated expression of fibrotic and inflammatory factors, oxidative stress, tubulointerstitial fibrosis, and tubule epithelial cell apoptosis, compared to the kidneys of wild-type mice with obstruction. PEDF knockout enhanced Wnt signaling activation induced by obstruction, while PEDF inhibited the Wnt pathway-mediated fibrosis in primary renal proximal tubule epithelial cells. Additionally, oxidative stress was aggravated in renal proximal tubule epithelial cells isolated from knockout mice and suppressed by PEDF treatment of renal proximal tubule epithelial cells. PEDF also reduced oxidation-induced apoptosis in renal proximal tubule epithelial cells. Thus, the renoprotective effects of PEDF are mediated, at least partially, by inhibition of the Wnt pathway. Hence, restoration of renal PEDF levels may have therapeutic potential for renal fibrosis.


Subject(s)
Diabetes Mellitus, Type 1/metabolism , Diabetes Mellitus, Type 2/metabolism , Epithelial Cells/metabolism , Eye Proteins/metabolism , Kidney Diseases/prevention & control , Kidney Tubules, Proximal/metabolism , Nerve Growth Factors/metabolism , Serpins/metabolism , Ureteral Obstruction/metabolism , Wnt Signaling Pathway , Animals , Apoptosis , Axin Protein/genetics , Axin Protein/metabolism , Cell Line , Diabetes Mellitus, Type 1/complications , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/pathology , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/pathology , Diabetic Nephropathies/genetics , Diabetic Nephropathies/metabolism , Diabetic Nephropathies/pathology , Diabetic Nephropathies/prevention & control , Disease Models, Animal , Epithelial Cells/pathology , Eye Proteins/genetics , Fibrosis , Gene Expression Regulation , Genetic Predisposition to Disease , Humans , Inflammation Mediators/metabolism , Kidney Diseases/genetics , Kidney Diseases/metabolism , Kidney Diseases/pathology , Kidney Tubules, Proximal/pathology , Mice, Inbred C57BL , Mice, Knockout , Nerve Growth Factors/deficiency , Nerve Growth Factors/genetics , Oxidative Stress , Phenotype , Serpins/deficiency , Serpins/genetics , Time Factors , Transfection , Ureteral Obstruction/complications , Ureteral Obstruction/genetics , Ureteral Obstruction/pathology
16.
BMC Bioinformatics ; 16 Suppl 13: S10, 2015.
Article in English | MEDLINE | ID: mdl-26423047

ABSTRACT

BACKGROUND: Adapter trimming and removal of duplicate reads are common practices in next-generation sequencing pipelines. Sequencing reads ambiguously mapped to repetitive and low complexity regions can also be problematic for accurate assessment of the biological signal, yet their impact on sequencing data has not received much attention. We investigate how trimming the adapters, removing duplicates, and filtering out reads overlapping low complexity regions influence the significance of biological signal in RNA- and ChIP-seq experiments. METHODS: We assessed the effect of data processing steps on the alignment statistics and the functional enrichment analysis results of RNA- and ChIP-seq data. We compared differentially processed RNA-seq data with matching microarray data on the same patient samples to determine whether changes in pre-processing improved correlation between the two. We have developed a simple tool to remove low complexity regions, RepeatSoaker, available at https://github.com/mdozmorov/RepeatSoaker, and tested its effect on the alignment statistics and the results of the enrichment analyses. RESULTS: Both adapter trimming and duplicate removal moderately improved the strength of biological signals in RNA-seq and ChIP-seq data. Aggressive filtering of reads overlapping with low complexity regions, as defined by RepeatMasker, further improved the strength of biological signals, and the correlation between RNA-seq and microarray gene expression data. CONCLUSIONS: Adapter trimming and duplicates removal, coupled with filtering out reads overlapping low complexity regions, is shown to increase the quality and reliability of detecting biological signals in RNA-seq and ChIP-seq data.


Subject(s)
High-Throughput Nucleotide Sequencing/methods , RNA/genetics , Sequence Analysis, RNA/methods , Humans
17.
Nat Commun ; 6: 7770, 2015 Jul 17.
Article in English | MEDLINE | ID: mdl-26183159

ABSTRACT

Platelet-derived growth factor (PDGF) is a mitogen and chemoattractant for vascular smooth muscle cells (VSMCs). However, the direct effects of PDGF receptor ß (PDGFRß) activation on VSMCs have not been studied in the context of atherosclerosis. Here we present a new mouse model of atherosclerosis with an activating mutation in PDGFRß. Increased PDGFRß signalling induces chemokine secretion and leads to leukocyte accumulation in the adventitia and media of the aorta. Furthermore, PDGFRß(D849V) amplifies and accelerates atherosclerosis in hypercholesterolemic ApoE(-/-) or Ldlr(-/-) mice. Intriguingly, increased PDGFRß signalling promotes advanced plaque formation at novel sites in the thoracic aorta and coronary arteries. However, deletion of the PDGFRß-activated transcription factor STAT1 in VSMCs alleviates inflammation of the arterial wall and reduces plaque burden. These results demonstrate that PDGFRß pathway activation has a profound effect on vascular disease and support the conclusion that inflammation in the outer arterial layers is a driving process for atherosclerosis.


Subject(s)
Atherosclerosis/genetics , Hypercholesterolemia/genetics , Plaque, Atherosclerotic/genetics , Receptor, Platelet-Derived Growth Factor beta/genetics , Animals , Apolipoproteins E/genetics , Atherosclerosis/metabolism , Blotting, Western , Chemokines/metabolism , Cholesterol/metabolism , Flow Cytometry , Gene Knock-In Techniques , Hypercholesterolemia/metabolism , Immunoprecipitation , Inflammation/metabolism , Mice , Mice, Knockout , Microscopy, Electron, Transmission , Muscle, Smooth, Vascular , Myocytes, Smooth Muscle , Plaque, Atherosclerotic/metabolism , Plaque, Atherosclerotic/pathology , Real-Time Polymerase Chain Reaction , Receptors, LDL/genetics , STAT1 Transcription Factor/genetics , STAT1 Transcription Factor/metabolism , Signal Transduction , Triglycerides/metabolism
18.
Genes Dev ; 29(11): 1106-19, 2015 Jun 01.
Article in English | MEDLINE | ID: mdl-26019175

ABSTRACT

Fibrosis is a common disease process in which profibrotic cells disturb organ function by secreting disorganized extracellular matrix (ECM). Adipose tissue fibrosis occurs during obesity and is associated with metabolic dysfunction, but how profibrotic cells originate is still being elucidated. Here, we use a developmental model to investigate perivascular cells in white adipose tissue (WAT) and their potential to cause organ fibrosis. We show that a Nestin-Cre transgene targets perivascular cells (adventitial cells and pericyte-like cells) in WAT, and Nestin-GFP specifically labels pericyte-like cells. Activation of PDGFRα signaling in perivascular cells causes them to transition into ECM-synthesizing profibrotic cells. Before this transition occurs, PDGFRα signaling up-regulates mTOR signaling and ribosome biogenesis pathways and perturbs the expression of a network of epigenetically imprinted genes that have been implicated in cell growth and tissue homeostasis. Isolated Nestin-GFP(+) cells differentiate into adipocytes ex vivo and form WAT when transplanted into recipient mice. However, PDGFRα signaling opposes adipogenesis and generates profibrotic cells instead, which leads to fibrotic WAT in transplant experiments. These results identify perivascular cells as fibro/adipogenic progenitors in WAT and show that PDGFRα targets progenitor cell plasticity as a profibrotic mechanism.


Subject(s)
Adipose Tissue/cytology , Adipose Tissue/physiopathology , Fibrosis/physiopathology , Receptor, Platelet-Derived Growth Factor alpha/metabolism , Signal Transduction , Adipogenesis/genetics , Animals , Cell Differentiation , Cell Proliferation , Cell Transplantation , Cells, Cultured , Gene Expression Regulation, Neoplastic , Mice , Receptor, Platelet-Derived Growth Factor alpha/genetics , Stem Cells/pathology
19.
Sci Rep ; 5: 8468, 2015 Feb 16.
Article in English | MEDLINE | ID: mdl-25683249

ABSTRACT

In primary brain tumors, oncogenes are frequently amplified and maintained on extrachromosomal DNA as double minutes (DM), but the underlying mechanisms remain poorly understood. We have generated a mouse model of malignant glioma based on knock-in of a mutant PDGF receptor α (PDGFRα) that is expressed in oligodendrocyte precursor cells (OPCs) after activation by a Cre recombinase. In the tumor suppressor INK4/Arf(-/-) background, mutant animals frequently developed brain tumors resembling anaplastic human gliomas (WHO grade III). Besides brain tumors, most animals also developed aggressive fibrosarcomas, likely triggered by Cre activation of mutant PDGFRα in fibroblastic cell lineages. Importantly, in the brain tumors and cell lines derived from brain tumor tissues, we identified a high prevalence of DM Pdgfra gene amplification, suggesting its occurrence as an early mutational event contributing to the malignant transformation of OPCs. Amplicons extended beyond the Pdgfra locus and included in some cases neighboring genes Kit and Kdr. Our genetically defined mouse brain tumor model therefore supports OPC as a cell of origin for malignant glioma and offers an example of a defined temporal sequence of mutational events, thus providing an entry point for a mechanistic understanding of DM gene amplification and its functionality in gliomagenesis.


Subject(s)
Brain Neoplasms/pathology , Gene Amplification , Glioma/pathology , Receptor, Platelet-Derived Growth Factor alpha/genetics , Activating Transcription Factor 4/deficiency , Activating Transcription Factor 4/genetics , Activating Transcription Factor 4/metabolism , Alleles , Animals , Brain Neoplasms/metabolism , Brain Neoplasms/veterinary , Cells, Cultured , Cyclin-Dependent Kinase Inhibitor p16/genetics , Cyclin-Dependent Kinase Inhibitor p16/metabolism , DNA, Circular/chemistry , Disease Models, Animal , Gene Knock-In Techniques , Glioma/metabolism , Glioma/veterinary , In Situ Hybridization, Fluorescence , Kaplan-Meier Estimate , Mice , Mice, Inbred C57BL , Oligodendroglia/cytology , Oligodendroglia/metabolism , Point Mutation , Receptor, Platelet-Derived Growth Factor alpha/metabolism , Severity of Illness Index
20.
Curr Rheumatol Rep ; 15(2): 304, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23307576

ABSTRACT

Fibrosis is the principal characteristic of the autoimmune disease known as scleroderma or systemic sclerosis (SSc). Studies published within the last three years suggest central involvement of platelet-derived growth factors (PDGFs) in SSc-associated fibrosis. PDGFs may also be involved in SSc-associated autoimmunity and vasculopathy. The PDGF signaling pathway is well understood and PDGF receptors are expressed on collagen-secreting fibroblasts and on mesenchymal stem and/or progenitor cells that may affect SSc in profound and unexpected ways. Although much work remains before we fully understand how PDGFs are involved in SSc, there is much interest in using PDGF inhibitors as a therapeutic approach to SSc.


Subject(s)
Platelet-Derived Growth Factor/physiology , Scleroderma, Systemic/physiopathology , Animals , Benzamides/therapeutic use , Fibrosis/drug therapy , Fibrosis/metabolism , Fibrosis/physiopathology , Humans , Imatinib Mesylate , Mice , Models, Animal , Piperazines/therapeutic use , Protein Kinase Inhibitors/therapeutic use , Pyrimidines/therapeutic use , Receptors, Platelet-Derived Growth Factor/metabolism , Scleroderma, Systemic/drug therapy , Scleroderma, Systemic/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...