Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 71
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Am J Physiol Lung Cell Mol Physiol ; 326(3): L303-L312, 2024 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-38226605

RESUMEN

Idiopathic pulmonary fibrosis (IPF) is an incurable interstitial lung disease characterized by fibrosis. Two FDA-approved drugs, pirfenidone and nintedanib, only modestly prolong survival. In this study, we asked whether levels of select circulating biomarkers in patients with IPF demonstrated changes in response to treatment over time and whether treatment with pirfenidone and nintedanib led to differential biomarker expression. Serial plasma samples from 48 patients with IPF on usual treatment and six healthy volunteers were analyzed to identify differentially expressed blood protein. Hypothesis-driven potential biomarker selection was based on recent literature, internal preclinical data, and the PROLIFIC Consortium (Schafer P. 6th Annual IPF Summit. Boston, MA, 2022) proposed biomarkers of pulmonary fibrosis. We compared our findings to public databases to provide insights into relevant signaling pathways in IPF. Of the 26 proteins measured, we found that 11 (SP-D, TIMP1, MMP7, CYFRA21-1, YKL40, CA125, sICAM, IP-10, MDC, CXCL13) were significantly elevated in patients with IPF compared with healthy volunteers but their levels did not significantly change over time. In the IPF samples, seven proteins were elevated in the treatment group compared with the no-treatment group. However, protein profiles were not distinguishable between patients on pirfenidone versus nintedanib. We demonstrated that most proteins differentially detected in our samples were predicted to be secreted from the lung epithelial or interstitial compartments. However, a significant minority of the proteins are not known to be transcriptionally expressed by lung cells, suggesting an ongoing systemic response. Understanding the contributions of the systemic response in IPF may be important as new therapeutics are developed.NEW & NOTEWORTHY In this study, we confirmed protein expression differences in only a subset of predicted biomarkers from IPF and control subjects. Most differentially expressed proteins were predicted to be secreted from lung cells. However, a significant minority of the proteins are not known to be transcriptionally expressed by lung cells, suggesting an ongoing systemic response. The contributions of the systemic response in IPF may be important as new therapeutics are developed.


Asunto(s)
Antígenos de Neoplasias , Fibrosis Pulmonar Idiopática , Queratina-19 , Humanos , Fibrosis Pulmonar Idiopática/patología , Pulmón/metabolismo , Fibrosis , Biomarcadores
2.
Am J Physiol Lung Cell Mol Physiol ; 324(3): L335-L344, 2023 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-36719987

RESUMEN

Nephronectin (NPNT) is a basement membrane (BM) protein and high-affinity ligand of integrin α8ß1 that is required for kidney morphogenesis in mice. In the lung, NPNT also localizes to BMs, but its potential role in pulmonary development has not been investigated. Mice with a floxed Npnt allele were used to generate global knockouts (KOs). Staged embryos were obtained by timed matings of heterozygotes and lungs were isolated for analysis. Although primary and secondary lung bud formation was normal in KO embryos, fusion of right lung lobes, primarily the medial and caudal, was first detected at E13.5 and persisted into adulthood. The lung parenchyma of KO mice was indistinguishable from wild-type (WT) and lobe fusion did not alter respiratory mechanics in adult KO mice. Interrogation of an existing single-cell RNA-seq atlas of embryonic and adult mouse lungs identified Npnt transcripts in mesothelial cells at E12.5 and into the early postnatal period, but not in adult lungs. KO embryonic lungs exhibited increased expression of laminin α5 and deposition of collagen IV in the mesothelial BM, accompanied by abnormalities in collagen fibrils in the adjacent stroma. Cranial and accessory lobes extracted from KO embryonic lungs fused ex vivo when cultured in juxtaposition, with the area of fusion showing loss of the mesothelial marker Wilms tumor 1. Because a similar pattern of lobe fusion was previously observed in integrin α8 KO embryos, our results suggest that NPNT signaling through integrin α8, likely in the visceral pleura, maintains right lung lobe separation during embryogenesis.


Asunto(s)
Proteínas de la Matriz Extracelular , Proteínas de la Membrana , Animales , Ratones , Proteínas de la Matriz Extracelular/genética , Desarrollo Embrionario/genética , Pulmón/metabolismo , Colágeno
3.
Am J Physiol Lung Cell Mol Physiol ; 322(4): L607-L616, 2022 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-35196901

RESUMEN

We previously showed that pericyte-like cells derived from the FoxD1-lineage contribute to myofibroblasts following bleomycin-induced lung injury. However, their functional significance in lung fibrosis remains unknown. In this study, we used a model of lung pericyte-like cell ablation to test the hypothesis that pericyte-like cell ablation attenuates lung fibrosis in bleomycin-induced lung injury. Lung fibrosis was induced by intratracheal instillation of bleomycin. To ablate pericyte-like cells in the lung, diphtheria toxin (DT) was administered to Foxd1-Cre;Rosa26-iDTR mice at two different phases of bleomycin-induced lung injury. For early ablation, we coadministered bleomycin with DT and harvested mice at days 7 and 21. To test the effect of ablation after acute injury, we delivered DT 7 days after bleomycin administration. We assessed fibrosis by lung hydroxyproline content and semiquantitative analysis of picrosirius red staining. We performed bronchoalveolar lavage to determine cell count and differential. We also interrogated mRNA expression of fibrosis-related genes in whole lung RNA. Compared with DT-insensitive littermates where pericyte-like cells were not ablated, DT-sensitive animals exhibited no difference in fibrosis at day 21 both in the early and late pericyte ablation models. However, early ablation of pericytes reduced acute lung inflammation, as indicated by decreased inflammatory cells. Our data confirm a role for pericytes in regulating pulmonary inflammation in early lung injury.


Asunto(s)
Lesión Pulmonar , Fibrosis Pulmonar , Animales , Bleomicina/farmacología , Líquido del Lavado Bronquioalveolar , Hidroxiprolina , Pulmón/metabolismo , Lesión Pulmonar/inducido químicamente , Lesión Pulmonar/patología , Lesión Pulmonar/terapia , Ratones , Ratones Endogámicos C57BL , Pericitos/metabolismo , Fibrosis Pulmonar/patología
4.
Lab Invest ; 101(5): 625-635, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33446892

RESUMEN

Pericytes apposed to the capillary endothelium are known to stabilize and promote endothelial integrity. Recent studies indicate that lung pericytes play a prominent role in lung physiology, and they are involved in the development of various lung diseases including lung injury in sepsis, pulmonary fibrosis, asthma, and pulmonary hypertension. Accordingly, human lung pericyte studies are important for understanding the mechanistic basis of lung physiology and pathophysiology; however, human lung pericytes can only be cultured for a few passages and no immortalized human lung pericyte cell line has been established so far. Thus, our study aims to establish an immortalized human lung pericyte cell line. Developed using SV40 large T antigen lentivirus, immortalized pericytes exhibit stable SV40T expression, sustained proliferation, and have significantly higher telomerase activity compared to normal human lung pericytes. In addition, these cells retained pericyte characteristics, marked by similar morphology, and expression of pericyte cell surface markers such as PDGFRß, NG2, CD44, CD146, CD90, and CD73. Furthermore, similar to that of primary pericytes, immortalized pericytes promoted endothelial cell tube formation and responded to different stimuli. Our previous data showed that friend leukemia virus integration 1 (Fli-1), a member of the ETS transcription factor family, is a key regulator that modulates inflammatory responses in mouse lung pericytes. We further demonstrated that Fli-1 regulates inflammatory responses in immortalized human lung pericytes. To summarize, we successfully established an immortalized human lung pericyte cell line, which serves as a promising tool for in vitro pericyte studies to understand human lung pericyte physiology and pathophysiology.


Asunto(s)
Línea Celular , Pulmón/citología , Pericitos , Proteínas de Microfilamentos/metabolismo , Transactivadores/metabolismo
5.
Am J Physiol Lung Cell Mol Physiol ; 319(5): L848-L853, 2020 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-32901522

RESUMEN

Antiretroviral therapy in HIV patients has lengthened lifespan but led to an increased risk for secondary comorbidities, such as pulmonary complications characterized by vascular dysfunction. In the lung, PDGFRß+ mesenchymal cells known as pericytes intimately associate with endothelial cells and are key for their survival both structurally and through the secretion of prosurvival factors. We hypothesize that in HIV infection there are functional changes in pericytes that may lead to destabilization of the microvasculature and ultimately to pulmonary abnormalities. Our objective in this study was to determine whether lung pericytes could be directly infected with HIV. We leveraged lung samples from macaque lungs with or without SIV infection and normal human lung for in vitro experiments. Pericytes were isolated based on the marker platelet-derived growth factor receptor-ß (PDGFRß). We determined that lung PDGFRß-positive (PDGFRß+) pericytes from both macaques and humans express CD4, the primary receptor for SIV/HIV, as well as the major coreceptors CXCR4 and CCR5. We found cells positive for both PDGFRß and SIV in lungs from infected macaques. Lung pericytes isolated from these animals also harbored detectable SIV. To confirm relevance to human disease, we demonstrated that human lung pericytes are capable of being productively infected by HIV in vitro, with the time course of infection suggesting development of viral latency. In summary, we show for the first time that SIV/HIV directly infects lung pericytes, implicating these cells as a novel target and potential reservoir for the virus in vivo.


Asunto(s)
Células Endoteliales/virología , Infecciones por VIH/virología , Pulmón/virología , Macrófagos/virología , Linfocitos T CD4-Positivos/virología , Humanos , Pulmón/inmunología , Macrófagos/inmunología , Receptores CXCR4/inmunología , Virus de la Inmunodeficiencia de los Simios/patogenicidad , Latencia del Virus/fisiología , Replicación Viral
8.
Adv Exp Med Biol ; 1122: 41-58, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30937862

RESUMEN

The lung has numerous roles, including gas exchange, immune surveillance, and barrier function. Being a highly vascularized organ, the lung receives dual blood supply from both the pulmonary and bronchial circulation. Therefore, pericytes likely play a prominent role in lung physiology given their localization in the perivascular niche. New genetic approaches have increased our understanding of the origin and the diverse functions of lung pericytes. Lung pericytes are myofibroblast progenitors, contributing to development of fibrosis in mouse models. Lung pericytes are also capable of responding to danger signals and amplify the inflammatory response through elaboration of cytokines and adhesion molecules. In this chapter, we describe the molecular, anatomical, and phenotypical characterization of lung pericytes. We further highlight their potential roles in the pathogenesis of lung diseases including pulmonary fibrosis, asthma, and pulmonary hypertension. Finally, current gaps in knowledge and areas of ongoing investigation in lung pericyte biology are also discussed.


Asunto(s)
Pulmón/citología , Miofibroblastos/citología , Pericitos/citología , Animales , Asma , Humanos , Hipertensión Pulmonar , Ratones , Fibrosis Pulmonar
9.
J Infect Dis ; 218(12): 1995-2005, 2018 11 05.
Artículo en Inglés | MEDLINE | ID: mdl-30053030

RESUMEN

Background: Pericytes are vascular mural cells and are embedded in the basement membrane of the microvasculature. Recent studies suggest a role for pericytes in lipopolysaccharide (LPS)-induced microvascular dysfunction and mortality, but the mechanisms of pericyte loss in sepsis are largely unknown. Methods: By using a cecal ligation and puncture (CLP)-induced murine model of sepsis, we observed that CLP led to lung and renal pericyte loss and reduced lung pericyte density and pericyte/endothelial cell (EC) coverage. Results: Up-regulated Friend leukemia virus integration 1 (Fli-1) messenger ribonucleic acid (RNA) and protein levels were found in lung pericytes from CLP mice in vivo and in LPS-stimulated lung pericytes in vitro. Knockout of Fli-1 in Foxd1-derived pericytes prevented CLP-induced pericyte loss, vascular leak, and improved survival. Disrupted Fli-1 expression by small interfering RNA inhibited LPS-induced inflammatory cytokines and chemokines in cultured lung pericytes. Furthermore, CLP-induced pericyte pyroptosis was mitigated in pericyte Fli-1 knockout mice. Conclusions: Our findings suggest that Fli-1 is a potential therapeutic target in sepsis.


Asunto(s)
Pericitos/fisiología , Proteína Proto-Oncogénica c-fli-1/metabolismo , Sepsis/metabolismo , Animales , Ciego , Células Cultivadas , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Inflamación/metabolismo , Ligadura , Lipopolisacáridos , Pulmón/citología , Ratones , Ratones Noqueados , Piroptosis , Sepsis/inmunología , Regulación hacia Arriba
10.
Am J Physiol Lung Cell Mol Physiol ; 315(6): L991-L1002, 2018 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-30335500

RESUMEN

Pericytes are key regulators of the microvasculature through their close interactions with the endothelium. However, pericytes play additional roles in tissue homeostasis and repair, in part by transitioning into myofibroblasts. Accumulation of myofibroblasts is a hallmark of fibrotic diseases such as idiopathic pulmonary fibrosis (IPF). To understand the contribution and role of pericytes in human lung fibrosis, we isolated these cells from non-IPF control and IPF lung tissues based on expression of platelet-derived growth factor receptor-ß (PDGFR-ß), a common marker of pericytes. When cultured in a specialized growth medium, PDGFR-ß+ cells retain the morphology and marker profile typical of pericytes. We found that IPF pericytes migrated more rapidly and invaded a basement membrane matrix more readily than control pericytes. Exposure of cells to transforming growth factor-ß, a major fibrosis-inducing cytokine, increased expression of α-smooth muscle actin and extracellular matrix genes in both control and IPF pericytes. Given that pericytes are uniquely positioned in vivo to respond to danger signals of both systemic and tissue origin, we stimulated human lung pericytes with agonists having pathogen-associated molecular patterns (PAMPs) or damage-associated molecular patterns (DAMPs). Both control and IPF lung pericytes increased expression of proinflammatory chemokines in response to specific PAMPs and DAMPs released from necrotic cells. Our results suggest that control and IPF lung pericytes are poised to react to tissue damage, as well as microbial and fibrotic stimuli. However, IPF pericytes are primed for migration and matrix invasion, features that may contribute to the function of these cells in lung fibrosis.


Asunto(s)
Fibrosis Pulmonar Idiopática/metabolismo , Fibrosis Pulmonar Idiopática/patología , Pulmón/metabolismo , Pulmón/patología , Pericitos/metabolismo , Pericitos/patología , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Adulto , Anciano , Matriz Extracelular/metabolismo , Matriz Extracelular/patología , Femenino , Fibroblastos/metabolismo , Fibroblastos/patología , Fibrosis/metabolismo , Fibrosis/patología , Humanos , Masculino , Persona de Mediana Edad , Miofibroblastos/metabolismo , Miofibroblastos/patología , Factor de Crecimiento Transformador beta/metabolismo , Adulto Joven
11.
Am J Physiol Lung Cell Mol Physiol ; 314(4): L583-L592, 2018 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-29351445

RESUMEN

Emphysema is one of the most common lung diseases in HIV+ individuals. The pathogenesis of HIV-associated emphysema remains unclear; however, radiographic distribution and earlier age of presentation of emphysema in the lungs of HIV+ patients are similar to deficiency of α1-antitrypsin (A1AT), a key elastase inhibitor in the lung. Reduced levels of circulating A1AT in HIV+ patients suggest a potential mechanism for emphysema development. In the present study we asked if A1AT levels and activity in the bronchoalveolar lavage fluid (BALF) differ in HIV+ and HIV- patients with and without emphysema. A1AT levels were measured by ELISA in plasma and BALF from a cohort of 21 HIV+ and 29 HIV- patients with or without emphysematous changes on chest CT scan. To analyze A1AT function, we measured elastase activity in the BALF and assessed oxidation and polymerization of A1AT by Western blotting. Total A1AT was increased in the BALF, but not in the plasma, of HIV+ compared with HIV- patients, regardless of the presence or absence of emphysema. However, antielastase activity was decreased in BALF from HIV+ patients, suggesting impaired A1AT function. Higher levels of the oxidized form of A1AT were detected in BALF from HIV+ than HIV- patients, which may account for the decreased antielastase activity. These findings suggest that, in the lungs of HIV+ patients, posttranslational modifications of A1AT produce a "functional deficiency" of this critical elastase inhibitor, which may contribute to emphysema development.


Asunto(s)
Biomarcadores/sangre , Infecciones por VIH/complicaciones , Enfisema Pulmonar/sangre , Inhibidores de Tripsina/sangre , alfa 1-Antitripsina/sangre , Líquido del Lavado Bronquioalveolar , Estudios de Cohortes , Femenino , VIH/aislamiento & purificación , Infecciones por VIH/virología , Humanos , Masculino , Persona de Mediana Edad , Enfisema Pulmonar/etiología , Enfisema Pulmonar/patología
12.
Am J Physiol Lung Cell Mol Physiol ; 314(4): L617-L625, 2018 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-29212802

RESUMEN

Mohan A, Malur A, McPeek M, Barna BP, Schnapp LM, Thomassen MJ, Gharib SA. Transcriptional survey of alveolar macrophages in a murine model of chronic granulomatous inflammation reveals common themes with human sarcoidosis. Am J Physiol Lung Cell Mol Physiol 314: L617-L625, 2018. First published December 6, 2017; doi: 10.1152/ajplung.00289.2017 . To advance our understanding of the pathobiology of sarcoidosis, we developed a multiwall carbon nanotube (MWCNT)-based murine model that shows marked histological and inflammatory signal similarities to this disease. In this study, we compared the alveolar macrophage transcriptional signatures of our animal model with human sarcoidosis to identify overlapping molecular programs. Whole genome microarrays were used to assess gene expression of alveolar macrophages in six MWCNT-exposed and six control animals. The results were compared with the transcriptional profiles of alveolar immune cells in 15 sarcoidosis patients and 12 healthy humans. Rigorous statistical methods were used to identify differentially expressed genes. To better elucidate activated pathways, integrated network and gene set enrichment analysis (GSEA) was performed. We identified over 1,000 differentially expressed between control and MWCNT mice. Gene ontology functional analysis showed overrepresentation of processes primarily involved in immunity and inflammation in MCWNT mice. Applying GSEA to both mouse and human samples revealed upregulation of 92 gene sets in MWCNT mice and 142 gene sets in sarcoidosis patients. Commonly activated pathways in both MWCNT mice and sarcoidosis included adaptive immunity, T-cell signaling, IL-12/IL-17 signaling, and oxidative phosphorylation. Differences in gene set enrichment between MWCNT mice and sarcoidosis patients were also observed. We applied network analysis to differentially expressed genes common between the MWCNT model and sarcoidosis to identify key drivers of disease. In conclusion, an integrated network and transcriptomics approach revealed substantial functional similarities between a murine model and human sarcoidosis particularly with respect to activation of immune-specific pathways.


Asunto(s)
Modelos Animales de Enfermedad , Perfilación de la Expresión Génica , Enfermedad Granulomatosa Crónica/metabolismo , Inflamación/metabolismo , Macrófagos Alveolares/metabolismo , Sarcoidosis/metabolismo , Animales , Estudios de Casos y Controles , Femenino , Regulación de la Expresión Génica , Enfermedad Granulomatosa Crónica/genética , Enfermedad Granulomatosa Crónica/patología , Humanos , Inflamación/genética , Inflamación/patología , Macrófagos Alveolares/patología , Ratones , Ratones Endogámicos C57BL , Nanotubos de Carbono/química , Sarcoidosis/genética , Sarcoidosis/patología , Transcripción Genética
13.
Am J Respir Cell Mol Biol ; 56(2): 160-167, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-27779900

RESUMEN

We demonstrated previously that FoxD1-derived cells in the lung are enriched in pericyte-like cells in mouse lung. These cells express the common pericyte markers and are located adjacent to endothelial cells. In this study, we demonstrate the feasibility of administering diphtheria toxin (DT) by oropharyngeal aspiration as an approach to ablating FoxD1-derived cells. We crossed mice expressing Cre-recombinase under the FoxD1 promoter to Rosa26-loxP-STOP-loxP-iDTR mice and generated a bitransgenic line (FoxD1-Cre;Rs26-iDTR) in which FoxD1-derived cells heritably express simian or human diphtheria toxin receptor and are sensitive to DT. We delivered low-dose (0.5 ng/g) and high-dose (1ng/g × 2) to FoxD1-Cre;Rs26-iDTR mice and littermate control mice by oropharyngeal aspiration and evaluated ablation by flow cytometry and immunohistochemistry. FoxD1-Cre mice showed a 40-50% reduction in PDGFRß+ cells by flow cytometry at Days 2 and 7 after DT administration, with a return of PDGFRß+ cells at Day 28. Confocal microscopy revealed an observable reduction in pericyte markers. Bronchoalveolar lavage fluid analysis revealed no significant differences in total protein, bronchoalveolar lavage fluid red blood cell, or white blood cell counts at low dose. However, at high-dose DT, there was a proinflammatory effect in the control mice and increased mortality associated with systemic toxicity in Cre+ mice. Low-dose DT reduced lung PDGFRß+ stromal cells in the FoxD1-Cre;iDTR transgenic model without a differential effect on lung inflammation in DT-sensitive and DT-insensitive animals. Low-dose DT is a viable method for transient lineage-specific stromal cell ablation in the lung that minimizes systemic toxicity.


Asunto(s)
Toxina Diftérica/administración & dosificación , Pulmón/citología , Boca/fisiología , Pericitos/citología , Faringe/fisiología , Succión/métodos , Animales , Líquido del Lavado Bronquioalveolar , Permeabilidad Capilar/efectos de los fármacos , Toxina Diftérica/farmacología , Ratones Transgénicos , Modelos Animales , Pericitos/efectos de los fármacos , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Células del Estroma/efectos de los fármacos , Células del Estroma/metabolismo
14.
Am J Physiol Lung Cell Mol Physiol ; 312(4): L556-L567, 2017 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-28188224

RESUMEN

Pericytes are perivascular PDGF receptor-ß+ (PDGFRß+) stromal cells required for vasculogenesis and maintenance of microvascular homeostasis in many organs. Because of their unique juxtaposition to microvascular endothelium, lung PDGFRß+ cells are well situated to detect proinflammatory molecules released following epithelial injury and promote acute inflammatory responses. Thus we hypothesized that these cells represent an unrecognized immune surveillance or injury-sentinel interstitial cell. To evaluate this hypothesis, we isolated PDGFRß+ cells from murine lung and demonstrated that they have characteristics consistent with a pericyte population (referred to as pericyte-like cells for simplicity hereafter). We showed that pericyte-like cells expressed functional Toll-like receptors and upregulated chemokine expression following exposure to bronchoalveolar lavage fluid (BALF) collected from mice with sterile lung injury. Interestingly, BALF from mice without lung injury also induced chemokine expression in pericyte-like cells, suggesting that pericyte-like cells are primed to sense epithelial injury (permeability changes). Following LPS-induced lung inflammation, increased numbers of pericyte-like cells expressed IL-6, chemokine (C-X-C motif) ligand-1, chemokine (C-C motif) ligand 2/ monocyte chemotactic protein-1, and ICAM-1 in vivo. Sterile lung injury in pericyte-ablated mice was associated with decreased inflammation compared with normal mice. In summary, we found that pericyte-like cells are immune responsive and express diverse chemokines in response to lung injury in vitro and in vivo. Furthermore, pericyte-like cell ablation attenuated inflammation in sterile lung injury, suggesting that these cells play an important functional role in mediating lung inflammatory responses. We propose a model in which pericyte-like cells function as interstitial immune sentinels, detecting proinflammatory molecules released following epithelial barrier damage and participating in recruitment of circulating leukocytes.


Asunto(s)
Sistema Inmunológico/citología , Pulmón/citología , Pericitos/citología , Animales , Líquido del Lavado Bronquioalveolar , Células Cultivadas , Inflamación/patología , Mediadores de Inflamación/metabolismo , Lesión Pulmonar/patología , Ratones Endogámicos C57BL , Ratones Transgénicos , Modelos Biológicos , Pericitos/metabolismo , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Células del Estroma/metabolismo
16.
Respir Res ; 17(1): 93, 2016 07 26.
Artículo en Inglés | MEDLINE | ID: mdl-27460362

RESUMEN

BACKGROUND: Sarcoidosis is a multisystem immuno-inflammatory disorder of unknown etiology that most commonly involves the lungs. We hypothesized that an unbiased approach to identify pathways activated in bronchoalveolar lavage (BAL) cells can shed light on the pathogenesis of this complex disease. METHODS: We recruited 15 patients with various stages of sarcoidosis and 12 healthy controls. All subjects underwent bronchoscopy with lavage. For each subject, total RNA was extracted from BAL cells and hybridized to an Affymetrix U133A microarray. Rigorous statistical methods were applied to identify differential gene expression between subjects with sarcoidosis vs. CONTROLS: To better elucidate pathways differentially activated between these groups, we integrated network and gene set enrichment analyses of BAL cell transcriptional profiles. RESULTS: Sarcoidosis patients were either non-smokers or former smokers, all had lung involvement and only two were on systemic prednisone. Healthy controls were all non-smokers. Comparison of BAL cell gene expression between sarcoidosis and healthy subjects revealed over 1500 differentially expressed genes. Several previously described immune mediators, such as interferon gamma, were upregulated in the sarcoidosis subjects. Using an integrative computational approach we constructed a modular network of over 80 gene sets that were highly enriched in patients with sarcoidosis. Many of these pathways mapped to inflammatory and immune-related processes including adaptive immunity, T-cell signaling, graft vs. host disease, interleukin 12, 23 and 17 signaling. Additionally, we uncovered a close association between the proteasome machinery and adaptive immunity, highlighting a potentially important and targetable relationship in the pathobiology of sarcoidosis. CONCLUSIONS: BAL cells in sarcoidosis are characterized by enrichment of distinct transcriptional programs involved in immunity and proteasomal processes. Our findings add to the growing evidence implicating alveolar resident immune effector cells in the pathogenesis of sarcoidosis and identify specific pathways whose activation may modulate disease progression.


Asunto(s)
Líquido del Lavado Bronquioalveolar/citología , Regulación de la Expresión Génica/genética , Sarcoidosis Pulmonar/genética , Sarcoidosis Pulmonar/metabolismo , Adulto , Anciano , Broncoscopía , Recuento de Células , Citocinas/metabolismo , Femenino , Redes Reguladoras de Genes/genética , Humanos , Inmunidad/genética , Masculino , Análisis por Micromatrices , Persona de Mediana Edad , Complejo de la Endopetidasa Proteasomal/genética , ARN/biosíntesis , ARN/aislamiento & purificación , Sarcoidosis Pulmonar/inmunología , Fumar/genética
17.
Am J Physiol Lung Cell Mol Physiol ; 308(8): L759-65, 2015 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-25713320

RESUMEN

Cysteine-rich protein-61 (CYR61), also known as connective tissue growth factor, CYR61, and nephroblastoma overexpressed gene 1 (CCN1), is a heparin-binding protein member of the CCN family of matricellular proteins. Gene expression profiles showed that Cyr61 is upregulated in human acute lung injury (ALI), but its functional role is unclear. We hypothesized that CYR61 contributes to ALI in mice. First, we demonstrated that CYR61 expression increases after bleomycin-induced lung injury. We then used adenovirus-mediated gene transfer to determine whether CYR61 overexpression in the lungs was sufficient to cause ALI. Mice instilled with CYR61 adenovirus showed greater weight loss, increased bronchoalveolar lavage total neutrophil counts, increased protein concentrations, and increased mortality compared with mice instilled with empty-vector adenovirus. Immunohistochemical studies in lungs from humans with idiopathic pulmonary fibrosis revealed CYR61 expression on the luminal membrane of alveolar epithelial cells in areas of injury. We conclude that CYR61 is upregulated in ALI and that CYR61 overexpression exacerbates ALI in mice.


Asunto(s)
Lesión Pulmonar Aguda/metabolismo , Proteína 61 Rica en Cisteína/metabolismo , Expresión Génica , Animales , Proteína 61 Rica en Cisteína/genética , Humanos , Pulmón/metabolismo , Masculino , Ratones Endogámicos C57BL , Síndrome de Dificultad Respiratoria/metabolismo
18.
Am J Physiol Lung Cell Mol Physiol ; 306(1): L35-42, 2014 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-24213920

RESUMEN

The lung is an important reservoir of human immunodeficiency virus (HIV). Individuals infected with HIV are more prone to pulmonary infections and chronic lung disorders. We hypothesized that comprehensively profiling the proteomic landscape of bronchoalveolar lavage fluid (BALF) in patients with HIV would provide insights into how this virus alters the lung milieu and contributes to pathogenesis of HIV-related lung diseases. BALF was obtained from five HIV-negative (HIV(-)) and six asymptomatic HIV-positive (HIV(+)) subjects not on antiretroviral therapy. Each sample underwent shotgun proteomic analysis based on HPLC-tandem mass spectrometry. Differentially expressed proteins between the groups were identified using statistical methods based on spectral counting. Mechanisms of disease were explored using functional annotation to identify overlapping and distinct pathways enriched between the BALF proteome of HIV(+) and HIV(-) subjects. We identified a total of 318 unique proteins in BALF of HIV(-) and HIV(+) subjects. Of these, 87 were differentially up- or downregulated between the two groups. Many of these differentially expressed proteins are known to interact with key HIV proteins. Functional analysis of differentially regulated proteins implicated downregulation of immune responses in lungs of HIV(+) patients. Combining shotgun proteomic analysis with computational methods demonstrated that the BALF proteome is significantly altered during HIV infection. We found that immunity-related pathways are underrepresented in HIV(+) patients. These findings implicate mechanisms whereby HIV invokes local immunosuppression in the lung and increases the susceptibility of HIV(+) patients to develop a wide range of infectious and noninfectious pulmonary diseases.


Asunto(s)
Infecciones por VIH/metabolismo , VIH-1 , Proteoma/metabolismo , Adulto , Líquido del Lavado Bronquioalveolar , Estudios de Casos y Controles , Citocinas/metabolismo , Femenino , Interacciones Huésped-Patógeno , Humanos , Pulmón/metabolismo , Pulmón/virología , Masculino , Persona de Mediana Edad , Adulto Joven
19.
Am J Respir Crit Care Med ; 188(7): 820-30, 2013 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-23924232

RESUMEN

RATIONALE: The origin of cells that make pathologic fibrillar collagen matrix in lung disease has been controversial. Recent studies suggest mesenchymal cells may contribute directly to fibrosis. OBJECTIVES: To characterize discrete populations of mesenchymal cells in the normal mouse lung and to map their fate after bleomycin-induced lung injury. METHODS: We mapped the fate of Foxd1-expressing embryonic progenitors and their progeny during lung development, adult homeostasis, and after fibrosing injury in Foxd1-Cre; Rs26-tdTomato-R mice. We studied collagen-I(α)1-producing cells in normal and diseased lungs using Coll-GFP(Tg) mice. MEASUREMENTS AND MAIN RESULTS: Foxd1-expressing embryonic progenitors enter lung buds before 13.5 days post-conception, expand, and form an extensive lineage of mesenchymal cells that have characteristics of pericytes. A collagen-I(α)1-expressing mesenchymal population of distinct lineage is also found in adult lung, with features of a resident fibroblast. In contrast to resident fibroblasts, Foxd1 progenitor-derived pericytes are enriched in transcripts for innate immunity, vascular development, WNT signaling pathway, and cell migration. Foxd1 progenitor-derived pericytes expand after bleomycin lung injury, and activate expression of collagen-I(α)1 and the myofibroblast marker αSMA in fibrotic foci. In addition, our studies suggest a distinct lineage of collagen-I(α)1-expressing resident fibroblasts that also expands after lung injury is a second major source of myofibroblasts. CONCLUSIONS: We conclude that the lung contains an extensive population of Foxd1 progenitor-derived pericytes that are an important lung myofibroblast precursor population.


Asunto(s)
Bleomicina/efectos adversos , Factores de Transcripción Forkhead/efectos de los fármacos , Lesión Pulmonar/inducido químicamente , Células Madre Mesenquimatosas/efectos de los fármacos , Pericitos/patología , Fibrosis Pulmonar/patología , Animales , Lesión Pulmonar/patología , Células Madre Mesenquimatosas/fisiología , Ratones , Miofibroblastos/efectos de los fármacos , Pericitos/efectos de los fármacos , Fibrosis Pulmonar/inducido químicamente
20.
iScience ; 27(6): 110010, 2024 Jun 21.
Artículo en Inglés | MEDLINE | ID: mdl-38868196

RESUMEN

Systemic sclerosis (SSc) is a chronic disease characterized by fibrosis and vascular abnormalities in the skin and internal organs, including the lung. SSc-associated pulmonary fibrosis (SSc-PF) is the leading cause of death in SSc patients. Pericytes are key regulators of vascular integrity and endothelial function. The role that pericytes play in SSc-PF remains unclear. We compared the transcriptome of pericytes from SSc-PF lungs (SScL) to pericytes from normal lungs (NORML). We identified 1,179 differentially expressed genes in SScL pericytes. Pathways enriched in SScL pericytes included prostaglandin, PI3K-AKT, calcium, and vascular remodeling signaling. Decreased cyclic AMP production and altered phosphorylation of AKT in response to prostaglandin E2 in SScL pericytes demonstrate the functional consequence of changes in the prostaglandin pathway that may contribute to fibrosis. The transcriptomic signature of SSc lung pericytes suggests that they promote vascular dysfunction and contribute to the loss of protection against lung inflammation and fibrosis.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA