Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
J Clin Invest ; 134(8)2024 Mar 11.
Artigo em Inglês | MEDLINE | ID: mdl-38466355

RESUMO

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.


Assuntos
Face , Histonas , Animais , Humanos , Epigênese Genética , Histonas/genética , Histonas/metabolismo , Lactatos/metabolismo , Mamíferos/metabolismo , Morfogênese , Crista Neural
2.
bioRxiv ; 2023 Nov 22.
Artigo em Inglês | MEDLINE | ID: mdl-38045303

RESUMO

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.
Artigo em Inglês | MEDLINE | ID: mdl-37555328

RESUMO

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.


Assuntos
Doenças das Valvas Cardíacas , Prolapso da Valva Mitral , Animais , Humanos , Camundongos , Células Endoteliais/metabolismo , Doenças das Valvas Cardíacas/genética , Doenças das Valvas Cardíacas/prevenção & controle , Doenças das Valvas Cardíacas/metabolismo , Valva Mitral/metabolismo , Prolapso da Valva Mitral/metabolismo , Fatores de Transcrição/metabolismo , Proteínas Proto-Oncogênicas c-sis/metabolismo
4.
Development ; 149(19)2022 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-36245218

RESUMO

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.


Assuntos
Ligamento Periodontal , Transcriptoma , Animais , Proteínas de Ligação ao Cálcio/metabolismo , Diferenciação Celular/genética , Proteínas da Matriz Extracelular/metabolismo , Camundongos , Osteoblastos , RNA/metabolismo
5.
Cell Rep ; 40(7): 111192, 2022 08 16.
Artigo em Inglês | MEDLINE | ID: mdl-35977484

RESUMO

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.


Assuntos
Miofibroblastos , Receptor alfa de Fator de Crescimento Derivado de Plaquetas , Animais , Diferenciação Celular/fisiologia , Fibroblastos/metabolismo , Fibrose , Camundongos , Miofibroblastos/patologia , Fator de Crescimento Derivado de Plaquetas/metabolismo , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/genética , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/metabolismo , Cicatrização
6.
Development ; 148(23)2021 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-34738614

RESUMO

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.


Assuntos
Mutação com Ganho de Função , Mioblastos Esqueléticos/metabolismo , Mutação Puntual , Receptor beta de Fator de Crescimento Derivado de Plaquetas/metabolismo , Substituição de Aminoácidos , Animais , Condrogênese/genética , Regulação da Expressão Gênica , Camundongos , Camundongos Transgênicos , Mioblastos Esqueléticos/patologia , Osteogênese/genética , Receptor beta de Fator de Crescimento Derivado de Plaquetas/genética , Fatores de Transcrição SOX9/genética , Fatores de Transcrição SOX9/metabolismo , Transdução de Sinais/genética
7.
Cell Stem Cell ; 28(6): 989-990, 2021 06 03.
Artigo em Inglês | MEDLINE | ID: mdl-34087159

RESUMO

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.


Assuntos
Integrases , Recombinases , Animais , Sequência de Bases , Movimento Celular , Integrases/metabolismo , Camundongos , Recombinases/genética , Recombinases/metabolismo
8.
Cell Stem Cell ; 26(5): 707-721.e5, 2020 05 07.
Artigo em Inglês | MEDLINE | ID: mdl-32229310

RESUMO

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.


Assuntos
Adipogenia , Receptor alfa de Fator de Crescimento Derivado de Plaquetas , Receptor beta de Fator de Crescimento Derivado de Plaquetas , Adipócitos , Adipogenia/genética , Animais , Diferenciação Celular/genética , Técnicas de Introdução de Genes , Camundongos , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/genética , Receptor beta de Fator de Crescimento Derivado de Plaquetas/genética
9.
Wound Repair Regen ; 28(4): 448-459, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32175700

RESUMO

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.


Assuntos
Artérias Carótidas/metabolismo , Fibroblastos/metabolismo , Miócitos de Músculo Liso/metabolismo , Miofibroblastos/metabolismo , Reepitelização/genética , Fator de Transcrição STAT1/genética , Pele/metabolismo , Actinas/metabolismo , Animais , Lesões das Artérias Carótidas/metabolismo , Movimento Celular/genética , Proliferação de Células/genética , Colágeno/metabolismo , Regulação da Expressão Gênica/genética , Tecido de Granulação/metabolismo , Camundongos , Camundongos Knockout , Músculo Liso Vascular/metabolismo , Fenótipo , Cicatrização/genética
10.
EMBO Mol Med ; 12(3): e11021, 2020 03 06.
Artigo em Inglês | MEDLINE | ID: mdl-31943786

RESUMO

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.


Assuntos
Nefropatias , Receptor beta de Fator de Crescimento Derivado de Plaquetas/metabolismo , Animais , Fibroblastos/patologia , Fibrose , Humanos , Rim/patologia , Nefropatias/patologia , Camundongos , Camundongos Transgênicos , Miofibroblastos/patologia
11.
Genes Dev ; 31(16): 1666-1678, 2017 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-28924035

RESUMO

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.


Assuntos
Transtornos do Crescimento/genética , Mutação , Receptor beta de Fator de Crescimento Derivado de Plaquetas/genética , Fator de Transcrição STAT1/genética , Tecido Adiposo/patologia , Animais , Aorta/patologia , Atrofia , Osso e Ossos/anormalidades , Feminino , Fibroblastos/metabolismo , Fibrose , Transtornos do Crescimento/metabolismo , Transtornos do Crescimento/patologia , Hiperplasia , Inflamação/metabolismo , Interferons/fisiologia , Masculino , Camundongos , Camundongos Knockout , Músculo Liso Vascular/patologia , Células NIH 3T3 , Fenótipo , Receptor beta de Fator de Crescimento Derivado de Plaquetas/metabolismo , Fator de Transcrição STAT1/metabolismo , Transdução de Sinais , Pele/patologia
12.
Development ; 144(1): 83-94, 2017 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-28049691

RESUMO

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.


Assuntos
Adipócitos/fisiologia , Adipogenia/genética , Tecido Adiposo/embriologia , Organogênese/genética , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/fisiologia , Células Estromais/fisiologia , Tecido Adiposo/crescimento & desenvolvimento , Tecido Adiposo/fisiologia , Substituição de Aminoácidos , Animais , Animais Recém-Nascidos , Linhagem da Célula/genética , Células Cultivadas , Embrião de Mamíferos , Feminino , Lipodistrofia/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Mutação Puntual , Gravidez
13.
Kidney Int ; 91(3): 642-657, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-27914705

RESUMO

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.


Assuntos
Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Células Epiteliais/metabolismo , Proteínas do Olho/metabolismo , Nefropatias/prevenção & controle , Túbulos Renais Proximais/metabolismo , Fatores de Crescimento Neural/metabolismo , Serpinas/metabolismo , Obstrução Ureteral/metabolismo , Via de Sinalização Wnt , Animais , Apoptose , Proteína Axina/genética , Proteína Axina/metabolismo , Linhagem Celular , Diabetes Mellitus Tipo 1/complicações , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/patologia , Diabetes Mellitus Tipo 2/complicações , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/patologia , Nefropatias Diabéticas/genética , Nefropatias Diabéticas/metabolismo , Nefropatias Diabéticas/patologia , Nefropatias Diabéticas/prevenção & controle , Modelos Animais de Doenças , Células Epiteliais/patologia , Proteínas do Olho/genética , Fibrose , Regulação da Expressão Gênica , Predisposição Genética para Doença , Humanos , Mediadores da Inflamação/metabolismo , Nefropatias/genética , Nefropatias/metabolismo , Nefropatias/patologia , Túbulos Renais Proximais/patologia , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fatores de Crescimento Neural/deficiência , Fatores de Crescimento Neural/genética , Estresse Oxidativo , Fenótipo , Serpinas/deficiência , Serpinas/genética , Fatores de Tempo , Transfecção , Obstrução Ureteral/complicações , Obstrução Ureteral/genética , Obstrução Ureteral/patologia
14.
BMC Bioinformatics ; 16 Suppl 13: S10, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26423047

RESUMO

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.


Assuntos
Sequenciamento de Nucleotídeos em Larga Escala/métodos , RNA/genética , Análise de Sequência de RNA/métodos , Humanos
15.
Nat Commun ; 6: 7770, 2015 Jul 17.
Artigo em Inglês | MEDLINE | ID: mdl-26183159

RESUMO

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.


Assuntos
Aterosclerose/genética , Hipercolesterolemia/genética , Placa Aterosclerótica/genética , Receptor beta de Fator de Crescimento Derivado de Plaquetas/genética , Animais , Apolipoproteínas E/genética , Aterosclerose/metabolismo , Western Blotting , Quimiocinas/metabolismo , Colesterol/metabolismo , Citometria de Fluxo , Técnicas de Introdução de Genes , Hipercolesterolemia/metabolismo , Imunoprecipitação , Inflamação/metabolismo , Camundongos , Camundongos Knockout , Microscopia Eletrônica de Transmissão , Músculo Liso Vascular , Miócitos de Músculo Liso , Placa Aterosclerótica/metabolismo , Placa Aterosclerótica/patologia , Reação em Cadeia da Polimerase em Tempo Real , Receptores de LDL/genética , Fator de Transcrição STAT1/genética , Fator de Transcrição STAT1/metabolismo , Transdução de Sinais , Triglicerídeos/metabolismo
16.
Genes Dev ; 29(11): 1106-19, 2015 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-26019175

RESUMO

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.


Assuntos
Tecido Adiposo/citologia , Tecido Adiposo/fisiopatologia , Fibrose/fisiopatologia , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/metabolismo , Transdução de Sinais , Adipogenia/genética , Animais , Diferenciação Celular , Proliferação de Células , Transplante de Células , Células Cultivadas , Regulação Neoplásica da Expressão Gênica , Camundongos , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/genética , Células-Tronco/patologia
17.
Sci Rep ; 5: 8468, 2015 Feb 16.
Artigo em Inglês | MEDLINE | ID: mdl-25683249

RESUMO

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.


Assuntos
Neoplasias Encefálicas/patologia , Amplificação de Genes , Glioma/patologia , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/genética , Fator 4 Ativador da Transcrição/deficiência , Fator 4 Ativador da Transcrição/genética , Fator 4 Ativador da Transcrição/metabolismo , Alelos , Animais , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/veterinária , Células Cultivadas , Inibidor p16 de Quinase Dependente de Ciclina/genética , Inibidor p16 de Quinase Dependente de Ciclina/metabolismo , DNA Circular/química , Modelos Animais de Doenças , Técnicas de Introdução de Genes , Glioma/metabolismo , Glioma/veterinária , Hibridização in Situ Fluorescente , Estimativa de Kaplan-Meier , Camundongos , Camundongos Endogâmicos C57BL , Oligodendroglia/citologia , Oligodendroglia/metabolismo , Mutação Puntual , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/metabolismo , Índice de Gravidade de Doença
18.
Curr Rheumatol Rep ; 15(2): 304, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23307576

RESUMO

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.


Assuntos
Fator de Crescimento Derivado de Plaquetas/fisiologia , Escleroderma Sistêmico/fisiopatologia , Animais , Benzamidas/uso terapêutico , Fibrose/tratamento farmacológico , Fibrose/metabolismo , Fibrose/fisiopatologia , Humanos , Mesilato de Imatinib , Camundongos , Modelos Animais , Piperazinas/uso terapêutico , Inibidores de Proteínas Quinases/uso terapêutico , Pirimidinas/uso terapêutico , Receptores do Fator de Crescimento Derivado de Plaquetas/metabolismo , Escleroderma Sistêmico/tratamento farmacológico , Escleroderma Sistêmico/metabolismo
19.
Dev Dyn ; 241(4): 663-74, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22354888

RESUMO

BACKGROUND: Intercellular communication between epithelial and mesenchymal cells is central to mammalian craniofacial development. ß-catenin is the gateway of canonical Wnt signaling, one of the major evolutionarily conserved cell-cell communication pathways in metazoa. In this study, we report an unexpected stage- and tissue-specific function of ß-catenin during mammalian jaw development. RESULTS: Using a unique mouse genetic tool, we have discovered that epithelial ß-catenin is essential for lower jaw formation, while attenuation of ß-catenin is required for proper upper jaw development. Changes in ß-catenin in vivo alter major epithelial Fgf8, Bmp4, Shh, and Edn1 signals, resulting in partial transcriptional reprogramming of the neural crest-derived mesenchyme, the primary source of jawbones. CONCLUSIONS: The Wnt/ß-catenin signal coordinates expression of multiple epithelial signals and has stage-specific asymmetric functions during mammalian upper and lower jaw development. In addition, these findings suggest that evolutionary changes of the canonical Wnt/ß-catenin signaling pathway may lead to innovation of jaws.


Assuntos
Arcada Osseodentária/embriologia , beta Catenina/fisiologia , Animais , Epitélio/embriologia , Epitélio/fisiologia , Regulação da Expressão Gênica no Desenvolvimento , Arcada Osseodentária/fisiologia , Mesoderma/embriologia , Mesoderma/fisiologia , Camundongos , Especificidade de Órgãos , Transdução de Sinais , Proteínas Wnt/metabolismo , beta Catenina/genética
20.
Dev Cell ; 20(6): 815-26, 2011 Jun 14.
Artigo em Inglês | MEDLINE | ID: mdl-21664579

RESUMO

Mural cells (pericytes and vascular smooth muscle cells) provide trophic and structural support to blood vessels. Vascular smooth muscle cells alternate between a synthetic/proliferative state and a differentiated/contractile state, but the dynamic states of pericytes are poorly understood. To explore the cues that regulate mural cell differentiation and homeostasis, we have generated conditional knockin mice with activating mutations at the PDGFRß locus. We show that increased PDGFRß signaling drives cell proliferation and downregulates differentiation genes in aortic vascular smooth muscle. Increased PDGFRß signaling also induces a battery of immune response genes in pericytes and mesenchymal cells and inhibits differentiation of white adipocytes. Mural cells are emerging as multipotent progenitors of pathophysiological importance, and we identify PDGFRß signaling as an important in vivo regulator of their progenitor potential.


Assuntos
Adipócitos Brancos/metabolismo , Encéfalo/metabolismo , Músculo Liso Vascular/citologia , Músculo Liso Vascular/metabolismo , Pericitos/citologia , Pericitos/metabolismo , Receptor beta de Fator de Crescimento Derivado de Plaquetas/fisiologia , Adipócitos Brancos/citologia , Animais , Encéfalo/citologia , Encéfalo/imunologia , Diferenciação Celular , Proliferação de Células , Células Cultivadas , Citocinas/metabolismo , Feminino , Citometria de Fluxo , Técnicas de Introdução de Genes , Homeostase , Integrases/metabolismo , Mesoderma/citologia , Mesoderma/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Fenótipo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA