Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 63
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
2.
Am J Physiol Lung Cell Mol Physiol ; 322(3): L385-L400, 2022 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-34986654

RESUMO

Idiopathic pulmonary fibrosis (IPF) is a chronic interstitial lung disease. The pathogenesis of IPF is not completely understood. However, numerous genes are associated with the development and progression of pulmonary fibrosis, indicating there is a significant genetic component to the pathogenesis of IPF. Epigenetic influences on the development of human disease, including pulmonary fibrosis, remain to be fully elucidated. In this paper, we identify miR-338-3p as a microRNA severely downregulated in the lungs of patients with pulmonary fibrosis and in experimental models of pulmonary fibrosis. Treatment of primary human lung fibroblasts with miR-338-3p inhibits myofibroblast differentiation and matrix protein production. Published and proposed targets of miR-338-3p such as TGFß receptor 1, MEK/ERK 1/2, Cdk4, and Cyclin D are also not responsible for the regulation of pulmonary fibroblast behavior by miR-338-3p. miR-338-3p inhibits myofibroblast differentiation by preventing TGFß-mediated downregulation of phosphatase and tensin homolog (PTEN), a known antifibrotic mediator.


Assuntos
Fibrose Pulmonar Idiopática , MicroRNAs , Fibroblastos/metabolismo , Humanos , Fibrose Pulmonar Idiopática/patologia , Pulmão/metabolismo , MicroRNAs/genética , MicroRNAs/metabolismo , Miofibroblastos/metabolismo , PTEN Fosfo-Hidrolase/genética , PTEN Fosfo-Hidrolase/metabolismo , Fator de Crescimento Transformador beta/metabolismo
3.
Curr Opin Ophthalmol ; 33(3): 219-227, 2022 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-35220328

RESUMO

PURPOSE OF REVIEW: Despite advancement in the surgical instrumentation and techniques, proliferative vitreoretinopathy (PVR) remains the most common cause for failure of rhegmatogenous retinal detachment (RRD) repair. This review discusses ongoing translational and clinical advancements in PVR. RECENT FINDINGS: PVR represents an exaggerated and protracted scarring process that can occur after RRD. The primary cell types involved are retinal pigment epithelium, glial, and inflammatory cells. They interact with growth factors and cytokines derived from the breakdown of the blood-retinal barrier that trigger a cascade of cellular processes, such as epithelial-mesenchymal transition, cell migration, chemotaxis, proliferation, elaboration of basement membrane and collagen and cellular contraction, leading to overt retinal pathology. Although there are currently no medical therapies proven to be effective against PVR in humans, increased understanding of the risks factors and pathophysiology have helped guide investigations for molecular targets of PVR. The leading therapeutic candidates are drugs that mitigate growth factors, inflammation, and proliferation are the leading therapeutic candidates. SUMMARY: Although multiple molecular targets have been investigated to prevent and treat PVR, none have yet demonstrated substantial evidence of clinical benefit in humans though some show promise. Advancements in our understanding of the pathophysiology of PVR may help develop a multipronged approach for this condition.


Assuntos
Descolamento Retiniano , Vitreorretinopatia Proliferativa , Transição Epitelial-Mesenquimal , Humanos , Descolamento Retiniano/complicações , Epitélio Pigmentado da Retina/patologia , Vitreorretinopatia Proliferativa/metabolismo , Vitreorretinopatia Proliferativa/cirurgia , Corpo Vítreo/patologia
4.
FASEB J ; 33(3): 3353-3363, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30376360

RESUMO

The obesity epidemic is developing into the most costly health problem facing the world. Obesity, characterized by excessive adipogenesis and enlarged adipocytes, promotes morbidities, such as diabetes, cardiovascular disease, and cancer. Regulation of adipogenesis is critical to our understanding of how fat cell formation causes obesity and associated health problems. Thy1 (also called CD90), a widely used stem cell marker, blocks adipogenesis and reduces lipid accumulation. Thy1-knockout mice are prone to diet-induced obesity. Although the importance of Thy1 in adipogenesis and obesity is now evident, how its expression is regulated is not. We hypothesized that DNA methylation has a role in promoting adipogenesis and affects Thy1 expression. Using the methylation inhibitor 5-aza-2'-deoxycytidine (5-aza-dC), we investigated whether DNA methylation alters Thy1 expression during adipogenesis in both mouse 3T3-L1 preadipocytes and mouse mesenchymal stem cells. Thy1 protein and mRNA levels were decreased dramatically during adipogenesis. However, 5-aza-dC treatment prevented that phenomenon. Methylation-sensitive pyrosequencing analysis showed that CpG sites at the Thy1 locus have increased methylation during adipogenesis, as well as increased methylation in adipose tissue from diet-induced obese mice. These new findings highlight the potential role of Thy1 and DNA methylation in adipogenesis and obesity.-Flores, E. M., Woeller, C. F., Falsetta, M. L., Susiarjo, M., Phipps, R. P. Thy1 (CD90) expression is regulated by DNA methylation during adipogenesis.


Assuntos
Adipogenia/genética , Metilação de DNA/genética , Antígenos Thy-1/genética , Células 3T3-L1 , Adipócitos/fisiologia , Tecido Adiposo/fisiologia , Animais , Diferenciação Celular/genética , Linhagem Celular , Células-Tronco Mesenquimais/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Obesos , Obesidade/genética , RNA Mensageiro/genética , Células-Tronco/fisiologia
5.
J Immunol ; 201(11): 3343-3351, 2018 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-30348736

RESUMO

Vaccination has been the most effective way to prevent or reduce infectious diseases; examples include the eradication of smallpox and attenuation of tetanus and measles. However, there is a large segment of the population that responds poorly to vaccines, in part because they are immunocompromised because of disease, age, or pharmacologic therapy and are unable to generate long-term protection. Specialized proresolving mediators are endogenously produced lipids that have potent proresolving and anti-inflammatory activities. Lipoxin B4 (LXB4) is a member of the lipoxin family, with its proresolving effects shown in allergic airway inflammation. However, its effects on the adaptive immune system, especially on human B cells, are not known. In this study, we investigated the effects of LXB4 on human B cells using cells from healthy donors and donors vaccinated against influenza virus in vitro. LXB4 promoted IgG Ab production in memory B cells and also increased the number of IgG-secreting B cells. LXB4 enhanced expression of two key transcription factors involved in plasma cell differentiation, BLIMP1 and XBP1. Interestingly, LXB4 increased expression of cyclooxygenase-2 (COX2), an enzyme that is required for efficient B cell Ab production. The effects of LXB4 are at least partially COX2-dependent as COX2 inhibitors attenuated LXB4-stimulated BLIMP1 and Xpb-1 expression as well as IgG production. Thus, our study reveals for the first time, to our knowledge, that LXB4 boosts memory B cell activation through COX2 and suggests that LXB4 can serve as a new vaccine adjuvant.


Assuntos
Adjuvantes Imunológicos/metabolismo , Anticorpos Antivirais/metabolismo , Linfócitos B/imunologia , Ciclo-Oxigenase 2/metabolismo , Vacinas contra Influenza/imunologia , Influenza Humana/imunologia , Lipoxinas/metabolismo , Imunidade Adaptativa , Formação de Anticorpos , Diferenciação Celular , Células Cultivadas , Ciclo-Oxigenase 2/genética , Humanos , Memória Imunológica , Fator 1 de Ligação ao Domínio I Regulador Positivo/genética , Fator 1 de Ligação ao Domínio I Regulador Positivo/metabolismo , Regulação para Cima , Vacinação , Proteína 1 de Ligação a X-Box/genética , Proteína 1 de Ligação a X-Box/metabolismo
6.
Am J Respir Cell Mol Biol ; 60(3): 269-278, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30265126

RESUMO

The differentiation of interstitial lung fibroblasts into contractile myofibroblasts that proliferate and secrete excessive extracellular matrix is critical for the pathogenesis of pulmonary fibrosis. Certain lipid signaling molecules, such as prostaglandins (PGs), can inhibit myofibroblast differentiation. However, the sources and delivery mechanisms of endogenous PGs are undefined. Activated primary human lung fibroblasts (HLFs) produce PGs such as PGE2. We report that activation of primary HLFs with IL-1ß inhibited transforming growth factor ß-induced myofibroblast differentiation in both the IL-1ß-treated cells themselves (autocrine signal) and adjacent naive HLFs in cocultures (paracrine signal). Additionally, we demonstrate for the first time that at least some of the antifibrotic effect of activated fibroblasts on nearby naive fibroblasts is carried by exosomes and other extracellular vesicles that contain several PGs, including high levels of the antifibrotic PGE2. Thus, activated fibroblasts communicate with surrounding cells to limit myofibroblast differentiation and maintain homeostasis. This work opens the way for future research into extracellular vesicle-mediated intercellular signaling in the lung and may inform the development of novel therapies for fibrotic lung diseases.


Assuntos
Antifibrinolíticos/farmacologia , Vesículas Extracelulares/efeitos dos fármacos , Fibroblastos/efeitos dos fármacos , Prostaglandinas/farmacologia , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Dinoprostona/metabolismo , Exossomos/efeitos dos fármacos , Exossomos/metabolismo , Matriz Extracelular/efeitos dos fármacos , Matriz Extracelular/metabolismo , Vesículas Extracelulares/metabolismo , Feminino , Fibroblastos/metabolismo , Humanos , Interleucina-1beta/metabolismo , Pulmão/efeitos dos fármacos , Pulmão/metabolismo , Masculino , Miofibroblastos/efeitos dos fármacos , Miofibroblastos/metabolismo , Fibrose Pulmonar/tratamento farmacológico , Fibrose Pulmonar/metabolismo , Transdução de Sinais/efeitos dos fármacos , Fator de Crescimento Transformador beta/metabolismo
7.
FASEB J ; 32(6): 3174-3183, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29401595

RESUMO

Thy1 (CD90), a glycosylated, glycophosphatidylinositol-anchored membrane protein highly expressed by subsets of mesenchymal stem cells and fibroblasts, inhibits adipogenesis. The role of Thy1 on bone structure and function has been poorly studied and represents a major knowledge gap. Therefore, we analyzed the long bones of wild-type (WT) and Thy1 knockout (KO) mice with micro-computed tomography (micro-CT) and histomorphometry to compare changes in bone architecture and overall bone structure. micro-CT analysis of long bones revealed Thy1 KO and WT mice fed a high-fat diet demonstrated bone structural parameters at 4 mo that differed significantly between WT and KO mice. A significant reduction in trabecular bone volume was noted in Thy1 KO mice. The most prominent differences were observed in trabecular bone volume ratio and trabecular bone connectivity density. Consistent with micro-CT measurements, histomorphometric analysis also showed decreased bone volume in the obese Thy1 KO mice compared to obese WT mice. In vitro assays revealed that osteogenic conditions increased Thy1 expression during OB differentiation and absence of Thy1 attenuated osteoblastogenesis. Together, these findings support the concept that Thy1 serves as a major mechanistic link to regulate bone formation and negatively regulate adipogenesis.-Paine, A., Woeller, C. F., Zhang, H., Garcia-Hernandez, M. L., Huertas, N., Xing, L., Phipps, R. P., Ritchlin, C. T. Thy1 is a positive regulator of osteoblast differentiation and modulates bone homeostasis in obese mice.


Assuntos
Osso Esponjoso/metabolismo , Diferenciação Celular , Homeostase , Obesidade/metabolismo , Osteoblastos/metabolismo , Antígenos Thy-1/biossíntese , Adipogenia/genética , Animais , Osso Esponjoso/diagnóstico por imagem , Osso Esponjoso/patologia , Camundongos , Camundongos Knockout , Camundongos Obesos , Obesidade/genética , Obesidade/patologia , Osteoblastos/patologia , Antígenos Thy-1/genética , Microtomografia por Raio-X
8.
Am J Physiol Lung Cell Mol Physiol ; 314(4): L569-L582, 2018 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-29351444

RESUMO

In pulmonary fibrosis (PF), fibroblasts and myofibroblasts proliferate and deposit excessive extracellular matrix in the interstitium, impairing normal lung function. Because most forms of PF have a poor prognosis and limited treatment options, PF represents an urgent unmet need for novel, effective therapeutics. Although the role of immune cells in lung fibrosis is unclear, recent studies suggest that T lymphocyte (T cell) activation may be impaired in PF patients. Furthermore, we have previously shown that activated T cells can produce prostaglandins with anti-scarring potential. Here, we test the hypothesis that activated T cells directly inhibit myofibroblast differentiation using a coculture system. Coculture with activated primary blood-derived T cells, from both healthy human donors and PF patients, inhibited transforming growth factor ß-induced myofibroblast differentiation in primary human lung fibroblasts isolated from either normal or PF lung tissue. Coculture supernatants contained anti-fibrotic prostaglandins D2 and E2, and the inhibitory effect of coculture on myofibroblast differentiation was largely reversed when prostaglandin production was abrogated either by resting the T cells before coculture or via specific pharmacological inhibitors. Moreover, coculture conditions induced COX-2 in HLFs but not in T cells, suggesting that T cells deliver an activating signal to HLFs, which in turn produce anti-fibrotic prostaglandins. We show for the first time that coculture with activated primary human T lymphocytes strongly inhibits myofibroblast differentiation, revealing a novel cell-to-cell communication network with therapeutic implications for fibrotic lung diseases.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Dinoprostona/metabolismo , Fibroblastos/patologia , Miofibroblastos/patologia , Prostaglandina D2/metabolismo , Linfócitos T/imunologia , Fator de Crescimento Transformador beta/farmacologia , Células Cultivadas , Técnicas de Cocultura , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Humanos , Miofibroblastos/efeitos dos fármacos , Miofibroblastos/metabolismo , Fibrose Pulmonar/metabolismo , Fibrose Pulmonar/patologia , Linfócitos T/metabolismo , Linfócitos T/patologia
9.
J Low Genit Tract Dis ; 22(1): 52-57, 2018 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-29271858

RESUMO

OBJECTIVES: Localized provoked vulvodynia (LPV) afflicts approximately 8% of women in the United States and represents a huge financial, physical, and psychological burden. Women with LPV experience intense pain localized to the vulvar vestibule (area immediately surrounding vaginal opening). We have identified mechanisms involved in the development of LPV whereby vulvar fibroblasts respond to proinflammatory stimuli to perpetuate an inflammatory response that causes pain. However, these mechanisms are not fully elucidated. Therefore, we explored the role of toll-like receptors (TLRs), a class of innate immune receptors that rapidly respond to microbial assaults. MATERIALS AND METHODS: To determine whether TLRs are expressed by vulvar fibroblasts and whether these contribute to proinflammatory mediator production and pain in LPV, we examined TLR expression and innate immune responses in fibroblasts derived from painful vestibular regions compared with nonpainful external vulvar regions. RESULTS: Human vulvar fibroblasts express functional TLRs that trigger production of inflammatory mediators associated with chronic pain. We focused on the TLR-7-imiquimod proinflammatory interaction, because imiquimod, a ligand of TLR-7, may exacerbate pain in women during treatment of human papillomavirus-associated disease. CONCLUSIONS: Human vulvar fibroblasts express a broad spectrum of TLRs (a new finding). A significantly higher TLR-mediated proinflammatory response was observed in LPV case vestibular fibroblasts, and with respect to the imiquimod-TLR 7 interaction, development of chronic vestibular pain and inflammation may be a possible sequelae of treatment of vulvar human papillomavirus-associated disease. Suppressing enhanced TLR-associated innate immune responses to a spectrum of pathogen-associated molecular patterns may represent a new/effective therapeutic approach for vulvodynia.


Assuntos
Aminoquinolinas/metabolismo , Fibroblastos/imunologia , Imunidade Inata , Mediadores da Inflamação/metabolismo , Transdução de Sinais , Receptor 7 Toll-Like/análise , Vulvodinia/induzido quimicamente , Células Cultivadas , Feminino , Fibroblastos/metabolismo , Perfilação da Expressão Gênica , Humanos , Imiquimode , Receptor 7 Toll-Like/genética , Vulvodinia/patologia
10.
Eur J Immunol ; 46(1): 81-91, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26474728

RESUMO

Specialized proresolving mediators (SPMs) constitute a recently recognized class of bioactive molecules thatpromote the resolution of inflammation. We recently reported that the SPMs resolvin D1 (RvD1) and 17-hydroxydocosahexaenoic acid (17-HDHA) promote the differentiation of IgG-secreting B cells and enhance antibody-mediated immune responses. However, there is an important knowledge gap regarding whether or not SPMs regulate human B-cell IgE production, which is the key effector in diseases such as asthma and allergy. Therefore, we investigated whether a panel of diverse SPMs influences B-cell IgE production. An important finding was that 17-HDHA and RvD1 inhibit IgE production by human B cells and suppress the differentiation of naïve B cells into IgE-secreting cells by specifically blocking epsilon germline transcript. This effect is specific to human IgE, as the SPMs do not inhibit production of IgM and IgG and did not suppress other IL-4-upregulated genes. 17-HDHA and RvD1 act by stabilizing the transcriptional repressor B-cell lymphoma 6, which competes with STAT6 for binding at the epsilon germline transcript promoter. Overall, these new findings demonstrate that certain SPMs inhibit the differentiation of IgE-producing B cells, without being broadly immune suppressive, representing a novel class of potential therapeutics for IgE-driven diseases such as asthma and allergy.


Assuntos
Linfócitos B/metabolismo , Ácidos Docosa-Hexaenoicos/imunologia , Regulação da Expressão Gênica/imunologia , Imunoglobulina E/biossíntese , Ativação Linfocitária/imunologia , Linfócitos B/imunologia , Western Blotting , Diferenciação Celular/imunologia , Células Cultivadas , Imunoprecipitação da Cromatina , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Humanos , Switching de Imunoglobulina/imunologia , Reação em Cadeia da Polimerase em Tempo Real
11.
Am J Pathol ; 186(12): 3189-3202, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27842700

RESUMO

Thyroid eye disease (TED) is a degenerative disease that manifests with detrimental tissue remodeling, myofibroblast accumulation, and scarring in the orbit of affected individuals. Currently, there are no effective therapies for TED that target or prevent the excessive tissue remodeling caused by myofibroblast formation and activation. The canonical cytokine that induces myofibroblast formation is transforming growth factor (TGF)-ß. The TGF-ß signaling pathway is influenced by aryl hydrocarbon receptor (AHR) signaling pathways. We hypothesized that AHR agonists can prevent myofibroblast formation in fibroblasts from patients with TED, and thus AHR ligands are potential therapeutics for the disease. Orbital fibroblasts explanted from patients with TED were treated with TGF-ß to induce myofibroblast formation, contraction, and proliferation. We found that AHR ligands prevent TGF-ß-dependent myofibroblast formation, and this ability is dependent on AHR expression. The AHR and AHR ligands block profibrotic Wnt signaling by inhibiting the phosphorylation of GSK3ß to prevent myofibroblast formation. These results provide new insight into the molecular pathways underlying orbital scarring in TED. These novel studies highlight the potential of the AHR and AHR ligands as future therapeutic options for eye diseases and possibly also for other scarring conditions.


Assuntos
Doença de Graves/fisiopatologia , Miofibroblastos/imunologia , Doenças Orbitárias/fisiopatologia , Receptores de Hidrocarboneto Arílico/imunologia , Fator de Crescimento Transformador beta/uso terapêutico , Via de Sinalização Wnt , Células Cultivadas , Fibroblastos/imunologia , Técnicas de Silenciamento de Genes , Doença de Graves/imunologia , Humanos , Ligantes , Miofibroblastos/metabolismo , Doenças Orbitárias/imunologia , RNA Interferente Pequeno , Receptores de Hidrocarboneto Arílico/genética , Receptores de Hidrocarboneto Arílico/metabolismo , Glândula Tireoide/imunologia , Glândula Tireoide/fisiopatologia , Fator de Crescimento Transformador beta/imunologia
12.
Genes Dev ; 23(1): 54-66, 2009 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-19095803

RESUMO

UPF1 functions in both Staufen 1 (STAU1)-mediated mRNA decay (SMD) and nonsense-mediated mRNA decay (NMD), which we show here are competitive pathways. STAU1- and UPF2-binding sites within UPF1 overlap so that STAU1 and UPF2 binding to UPF1 appear to be mutually exclusive. Furthermore, down-regulating the cellular abundance of STAU1, which inhibits SMD, increases the efficiency of NMD, whereas down-regulating the cellular abundance of UPF2, which inhibits NMD, increases the efficiency of SMD. Competition under physiological conditions is exemplified during the differentiation of C2C12 myoblasts to myotubes: The efficiency of SMD increases and the efficiency of NMD decreases, consistent with our finding that more STAU1 but less UPF2 bind UPF1 in myotubes compared with myoblasts. Moreover, an increase in the cellular level of UPF3X during myogenesis results in an increase in the efficiency of an alternative NMD pathway that, unlike classical NMD, is largely insensitive to UPF2 down-regulation. We discuss the remarkable balance between SMD and the two types of NMD in view of data indicating that PAX3 mRNA is an SMD target whose decay promotes myogenesis whereas myogenin mRNA is a classical NMD target encoding a protein required for myogenesis.


Assuntos
Desenvolvimento Muscular/fisiologia , Miogenina/metabolismo , Fatores de Transcrição Box Pareados/metabolismo , Estabilidade de RNA/fisiologia , RNA Mensageiro/metabolismo , Aminoácidos/metabolismo , Animais , Sítios de Ligação , Células COS , Diferenciação Celular , Linhagem Celular , Chlorocebus aethiops , Proteínas do Citoesqueleto/metabolismo , Regulação para Baixo , Células HeLa , Humanos , Camundongos , Microtúbulos/metabolismo , Mioblastos/citologia , Mioblastos/metabolismo , Fator de Transcrição PAX3 , Proteínas de Ligação a RNA/metabolismo , Transativadores/metabolismo
13.
J Biol Chem ; 290(6): 3563-75, 2015 Feb 06.
Artigo em Inglês | MEDLINE | ID: mdl-25538236

RESUMO

Although scarring is a component of wound healing, excessive scar formation is a debilitating condition that results in pain, loss of tissue function, and even death. Many tissues, including the lungs, heart, skin, and eyes, can develop excessive scar tissue as a result of tissue injury, chronic inflammation, or autoimmune disease. Unfortunately, there are few, if any, effective treatments to prevent excess scarring, and new treatment strategies are needed. Using HEK293FT cells stably transfected with a TGFß-dependent luciferase reporter, we performed a small molecule screen to identify novel compounds with antiscarring activity. We discovered that the polyether ionophore salinomycin potently inhibited the formation of scar-forming myofibroblasts. Salinomycin (250 nm) blocked TGFß-dependent expression of the cardinal myofibroblast products α smooth muscle actin, calponin, and collagen in primary human fibroblasts without causing cell death. Salinomycin blocked phosphorylation and activation of TAK1 and p38, two proteins fundamentally involved in signaling myofibroblast and scar formation. Expression of constitutively active mitogen activated kinase kinase 6, which activates p38 MAPK, attenuated the ability of salinomycin to block myofibroblast formation, demonstrating that salinomycin targets the p38 kinase pathway to disrupt TGFß signaling. These data identify salinomycin and other polyether ionophores as novel potential antiscarring therapeutics.


Assuntos
Miofibroblastos/efeitos dos fármacos , Piranos/farmacologia , Bibliotecas de Moléculas Pequenas/farmacologia , Cicatrização/efeitos dos fármacos , Actinas/genética , Actinas/metabolismo , Proteínas de Ligação ao Cálcio/genética , Proteínas de Ligação ao Cálcio/metabolismo , Proliferação de Células , Células Cultivadas , Colágeno/genética , Colágeno/metabolismo , Células HEK293 , Humanos , MAP Quinase Quinase Quinases/metabolismo , Sistema de Sinalização das MAP Quinases , Proteínas dos Microfilamentos/genética , Proteínas dos Microfilamentos/metabolismo , Miofibroblastos/metabolismo , Miofibroblastos/fisiologia , Fator de Crescimento Transformador beta/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Calponinas
14.
Am J Physiol Lung Cell Mol Physiol ; 311(5): L855-L867, 2016 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-27612965

RESUMO

Human lung fibroblasts (HLFs) act as innate immune sentinel cells that amplify the inflammatory response to injurious stimuli. Here, we use targeted lipidomics to explore the hypothesis that HLFs also play an active role in the resolution of inflammation. We detected cyclooxygenase-2 (COX-2)-dependent production of both proinflammatory and proresolving prostaglandins (PGs) in conditioned culture medium from HLFs treated with a proinflammatory stimulus, IL-1ß. Among the proresolving PGs in the HLF lipidome were several known ligands for peroxisome proliferator-activated receptor-γ (PPARγ), a transcription factor whose activation in the lung yields potent anti-inflammatory, antifibrotic, and proresolving effects. Next, we used a cell-based luciferase reporter to confirm the ability of HLF supernatants to activate PPARγ, demonstrating, for the first time, that primary HLFs activated with proinflammatory IL-1ß or cigarette smoke extract produce functional PPARγ ligands; this phenomenon is temporally regulated, COX-2- and lipocalin-type PGD synthase-dependent, and enhanced by arachidonic acid supplementation. Finally, we used luciferase reporter assays to show that several of the PGs in the lipidome of activated HLFs independently activate PPARγ and/or inhibit NFκB. These results indicate that HLFs, as immune sentinels, regulate both proinflammatory and proresolving responses to injurious stimuli. This novel endogenous resolution pathway represents a new therapeutic target for globally important inflammatory diseases such as chronic obstructive pulmonary disease.


Assuntos
Ciclo-Oxigenase 2/metabolismo , Fibroblastos/metabolismo , Pulmão/citologia , PPAR gama/metabolismo , Ácidos Araquidônicos/farmacologia , Meios de Cultivo Condicionados/farmacologia , Dinoprostona/metabolismo , Eicosanoides/metabolismo , Feminino , Fibroblastos/efeitos dos fármacos , Fibroblastos/enzimologia , Células HEK293 , Humanos , Mediadores da Inflamação/metabolismo , Interleucina-1beta/farmacologia , Interleucina-6/metabolismo , Interleucina-8/metabolismo , Oxirredutases Intramoleculares/metabolismo , Ligantes , Lipocalinas/metabolismo , Masculino , NF-kappa B/metabolismo , Prostaglandina-E Sintases , Fumar , Regulação para Cima/efeitos dos fármacos
15.
FASEB J ; 29(3): 920-31, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25416548

RESUMO

Worldwide obesity rates are at epidemic levels, and new insight into the regulation of obesity and adipogenesis are required. Thy1 (CD90), a cell surface protein with an enigmatic function, is expressed on subsets of fibroblasts and stem cells. We used a diet-induced obesity model to show that Thy1-null mice gain weight at a faster rate and gain 30% more weight than control C57BL/6 mice. During adipogenesis, Thy1 expression is lost in mouse 3T3-L1 cells. Overexpression of Thy1 blocked adipocyte formation and reduced mRNA and protein expression of an adipocyte marker, fatty acid-binding protein 4, by 5-fold. Although preadipocyte fibroblasts expressed Thy1 mRNA and protein, adipocytes from mouse and human fat tissue had almost undetectable Thy1 levels. Thy1 decreases the activity of the adipogenic transcription factor PPARγ by more than 60% as shown by PPARγ-dependent reporter assays. Using both genetic and pharmacologic approaches, we show Thy1 expression dampens PPARγ by inhibiting the activity of the Src-family kinase, Fyn. Thus, these studies reveal Thy1 blocks adipogenesis and PPARγ by inhibiting Fyn and support the idea that Thy1 is a novel therapeutic target in obesity.


Assuntos
Adipogenia/fisiologia , Regulação Enzimológica da Expressão Gênica , Obesidade/fisiopatologia , Proteínas Proto-Oncogênicas c-fyn/fisiologia , Antígenos Thy-1/metabolismo , Células 3T3-L1 , Adipócitos/citologia , Adipócitos/metabolismo , Animais , Western Blotting , Diferenciação Celular , Proliferação de Células , Células Cultivadas , Dieta Hiperlipídica , Proteínas de Ligação a Ácido Graxo/genética , Proteínas de Ligação a Ácido Graxo/metabolismo , Fibroblastos/citologia , Fibroblastos/metabolismo , Citometria de Fluxo , Humanos , Imunoprecipitação , Masculino , Camundongos , Camundongos Endogâmicos C57BL , PPAR gama/genética , PPAR gama/metabolismo , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Antígenos Thy-1/genética
17.
Am J Pathol ; 184(5): 1429-45, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24650561

RESUMO

Corneal scarring, whether caused by trauma, laser refractive surgery, or infection, remains a significant problem for humans. Certain ligands for peroxisome proliferator-activated receptor gamma (PPARγ) have shown promise as antiscarring agents in a variety of body tissues. In the cornea, their relative effectiveness and mechanisms of action are still poorly understood. Here, we contrasted the antifibrotic effects of three different PPARγ ligands (15-deoxy-Δ12,14-prostaglandin J2, troglitazone, and rosiglitazone) in cat corneal fibroblasts. Western blot analyses revealed that all three compounds reduced transforming growth factor (TGF)-ß1-driven myofibroblast differentiation and up-regulation of α-smooth muscle actin, type I collagen, and fibronectin expression. Because these effects were independent of PPARγ, we ascertained whether they occurred by altering phosphorylation of Smads 2/3, p38 mitogen-activated protein kinase, stress-activated protein kinase, protein kinase B, extracellular signal-regulated kinase, and/or myosin light chain 2. Only p38 mitogen-activated protein kinase phosphorylation was significantly inhibited by all three PPARγ ligands. Finally, we tested the antifibrotic potential of troglitazone in a cat model of photorefractive keratectomy-induced corneal injury. Topical application of troglitazone significantly reduced α-smooth muscle actin expression and haze in the stromal ablation zone. Thus, the PPARγ ligands tested here showed great promise as antifibrotics, both in vitro and in vivo. Our results also provided new evidence for the signaling pathways that may underlie these antifibrotic actions in corneal fibroblasts.


Assuntos
Córnea/patologia , Miofibroblastos/patologia , PPAR gama/metabolismo , Fator de Crescimento Transformador beta1/farmacologia , Actinas/metabolismo , Animais , Gatos , Linhagem Celular Transformada , Núcleo Celular/efeitos dos fármacos , Núcleo Celular/metabolismo , Cromanos/farmacologia , Colágeno Tipo I/metabolismo , Fibronectinas/metabolismo , Fibrose , Humanos , Ligantes , Miofibroblastos/efeitos dos fármacos , Miofibroblastos/enzimologia , Fosforilação/efeitos dos fármacos , Prostaglandina D2/análogos & derivados , Prostaglandina D2/farmacologia , Inibidores de Proteínas Quinases/farmacologia , Transporte Proteico/efeitos dos fármacos , Rosiglitazona , Transdução de Sinais/efeitos dos fármacos , Proteínas Smad/metabolismo , Tiazolidinedionas/farmacologia , Troglitazona , Cicatrização/efeitos dos fármacos , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
18.
Am J Obstet Gynecol ; 213(1): 38.e1-38.e12, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25683963

RESUMO

OBJECTIVE: Our goal was to gain a better understanding of the inflammatory pathways affected during localized vulvodynia, a poorly understood, common, and debilitating condition characterized by chronic pain of the vulvar vestibule. STUDY DESIGN: In a control matched study, primary human fibroblast strains were generated from biopsies collected from localized provoked vulvodynia (LPV) cases and from age- and race-matched controls. We then examined intracellular mechanisms by which these fibroblasts recognize pathogenic Candida albicans; >70% of vulvodynia patients report the occurrence of prior chronic Candida infections, which is accompanied by localized inflammation and elevated production of proinflammatory/pain-associated interleukin (IL)-6 and prostaglandin E2 (PGE2). We focused on examining the signaling pathways involved in recognition of yeast components that are present and abundant during chronic infection. RESULTS: Dectin-1, a surface receptor that binds C albicans cell wall glucan, was significantly elevated in vestibular vs external vulvar cells (from areas without pain) in both cases and controls, while its abundance was highest in LPV cases. Blocking Dectin-1 signaling significantly reduced pain-associated IL-6 and PGE2 production during the response to C albicans. Furthermore, LPV patient vestibular cells produced inflammatory mediators in response to low numbers of C albicans cells, while external vulvar fibroblasts were nonresponsive. Inhibition of nuclear factor kappa-light-chain-enhancer of activated B cells (proinflammatory transcription factor) nearly abrogated IL-6 and PGE2 production induced by C albicans, in keeping with observations that Dectin-1 signals through the nuclear factor kappa-light-chain-enhancer of activated B cells pathway. CONCLUSION: These findings implicate that a fibroblast-mediated proinflammatory response to C albicans contributes to the induction of pain in LPV cases. Targeting this response may be an ideal strategy for the development of new vulvodynia therapies.


Assuntos
Vulvodinia/fisiopatologia , Adulto , Candidíase Vulvovaginal/fisiopatologia , Dinoprostona/metabolismo , Feminino , Fibroblastos/fisiologia , Humanos , Inflamação/fisiopatologia , Interleucina-6/metabolismo , Lectinas Tipo C/metabolismo , NF-kappa B/metabolismo , Dor/etiologia , Dor/fisiopatologia , Reação em Cadeia da Polimerase em Tempo Real , Vulvodinia/microbiologia
19.
Exp Eye Res ; 127: 290-8, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25245083

RESUMO

Selective glucocorticoid receptor agonists (SEGRAs) are a new class of compounds under clinical evaluation for treatment of ocular inflammation. Widely prescribed therapeutics, such as glucocorticoids, are effective at reducing ocular inflammation, but their long term use predisposes to undesirable side effects. The purpose of this study was to investigate a novel SEGRA, mapracorat (BOL-303242-X), and the differences in mapracorat's mechanism of action compared with traditional steroids (i.e. dexamethasone). Keratocytes from three different humans were cultured and treated with mapracorat or dexamethasone, with and without a strong provoking agent, interleukin (IL)-1ß. The effects of mapracorat compared to dexamethasone were determined by measuring protein levels (Western blotting) and DNA binding (ELISA) for two nuclear factor-kappaB (NF-κB) family members, RelA and RelB. Cytokine production (i.e. IL-6, IL-8, prostaglandin E2 (PGE2)) was characterized by immunoassay. Our findings reveal mechanistic differences between mapracorat and traditional steroid therapies. Mapracorat showed partial attenuation of the classical NF-κB pathway, consistent with traditional steroids. However, mapracorat uniquely potentiated a novel anti-inflammatory mechanism through rapid upregulation of RelB, an anti-inflammatory member of the NF-κB alternative pathway. Mapracorat potently inhibits ocular inflammation in vitro and is a promising new treatment for ocular inflammatory disease. Mapracorat acts, in part, by a novel mechanism via upregulation of RelB in the NF-κB alternative pathway.


Assuntos
Anti-Inflamatórios/farmacologia , Benzofuranos/farmacologia , Ceratócitos da Córnea/efeitos dos fármacos , NF-kappa B/metabolismo , Pentanóis/farmacologia , Quinolinas/farmacologia , Receptores de Glucocorticoides/agonistas , Fator de Transcrição RelB/metabolismo , Western Blotting , Células Cultivadas , Ceratócitos da Córnea/metabolismo , Citocinas/metabolismo , Dexametasona/farmacologia , Ensaio de Imunoadsorção Enzimática , Glucocorticoides/farmacologia , Humanos , Fator de Transcrição RelA/metabolismo , Regulação para Cima
20.
J Biol Chem ; 287(10): 7051-62, 2012 Mar 02.
Artigo em Inglês | MEDLINE | ID: mdl-22235121

RESUMO

The de novo thymidylate biosynthetic pathway in mammalian cells translocates to the nucleus for DNA replication and repair and consists of the enzymes serine hydroxymethyltransferase 1 and 2α (SHMT1 and SHMT2α), thymidylate synthase, and dihydrofolate reductase. In this study, we demonstrate that this pathway forms a multienzyme complex that is associated with the nuclear lamina. SHMT1 or SHMT2α is required for co-localization of dihydrofolate reductase, SHMT, and thymidylate synthase to the nuclear lamina, indicating that SHMT serves as scaffold protein that is essential for complex formation. The metabolic complex is enriched at sites of DNA replication initiation and associated with proliferating cell nuclear antigen and other components of the DNA replication machinery. These data provide a mechanism for previous studies demonstrating that SHMT expression is rate-limiting for de novo thymidylate synthesis and indicate that de novo thymidylate biosynthesis occurs at replication forks.


Assuntos
Replicação do DNA/fisiologia , DNA/biossíntese , Glicina Hidroximetiltransferase/metabolismo , Complexos Multienzimáticos/metabolismo , Lâmina Nuclear/enzimologia , Timidina Monofosfato/biossíntese , DNA/genética , Reparo do DNA/fisiologia , Glicina Hidroximetiltransferase/genética , Células HeLa , Humanos , Complexos Multienzimáticos/genética , Lâmina Nuclear/metabolismo , Tetra-Hidrofolato Desidrogenase/genética , Tetra-Hidrofolato Desidrogenase/metabolismo , Timidina Monofosfato/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA