Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
J Autoimmun ; 124: 102713, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34390919

RESUMEN

Despite the existence of potent anti-inflammatory biological drugs e.g., anti-TNF and anti IL-6 receptor antibodies, for treating chronic inflammatory and autoimmune diseases, these are costly and not specific. Cheaper oral available drugs remain an unmet need. Expression of the acute phase protein Serum Amyloid A (SAA) is dependent on release of pro-inflammatory cytokines IL-1, IL-6 and TNF-α during inflammation. Conversely, SAA induces pro-inflammatory cytokine secretion, including Th17, leading to a pathogenic vicious cycle and chronic inflammation. 5- MER peptide (5-MP) MTADV (methionine-threonine-alanine-aspartic acid-valine), also called Amilo-5MER, was originally derived from a sequence of a pro-inflammatory CD44 variant isolated from synovial fluid of a Rheumatoid Arthritis (RA) patient. This human peptide displays an efficient anti-inflammatory effects to ameliorate pathology and clinical symptoms in mouse models of RA, Inflammatory Bowel Disease (IBD) and Multiple Sclerosis (MS). Bioinformatics and qRT-PCR revealed that 5-MP, administrated to encephalomyelytic mice, up-regulates genes contributing to chronic inflammation resistance. Mass spectrometry of proteins that were pulled down from an RA synovial cell extract with biotinylated 5-MP, showed that it binds SAA. 5-MP disrupted SAA assembly, which is correlated with its pro-inflammatory activity. The peptide MTADV (but not scrambled TMVAD) significantly inhibited the release of pro-inflammatory cytokines IL-6 and IL-1ß from SAA-activated human fibroblasts, THP-1 monocytes and peripheral blood mononuclear cells. 5-MP suppresses the pro-inflammatory IL-6 release from SAA-activated cells, but not from non-activated cells. 5-MP could not display therapeutic activity in rats, which are SAA deficient, but does inhibit inflammations in animal models of IBD and MS, both are SAA-dependent, as shown by others in SAA knockout mice. In conclusion, 5-MP suppresses chronic inflammation in animal models of RA, IBD and MS, which are SAA-dependent, but not in animal models, which are SAA-independent.


Asunto(s)
Artritis Reumatoide/inmunología , Receptores de Hialuranos/genética , Inflamación/inmunología , Enfermedades Inflamatorias del Intestino/inmunología , Esclerosis Múltiple/inmunología , Péptidos/genética , Proteína Amiloide A Sérica/inmunología , Animales , Antiinflamatorios/uso terapéutico , Autoinmunidad , Células Cultivadas , Biología Computacional , Citocinas/metabolismo , Modelos Animales de Enfermedad , Humanos , Mediadores de Inflamación/metabolismo , Ratones , Ratones Noqueados , Péptidos/uso terapéutico , Proteína Amiloide A Sérica/genética
2.
Int J Mol Sci ; 22(4)2021 Feb 22.
Artículo en Inglés | MEDLINE | ID: mdl-33671651

RESUMEN

By dint of the aging population and further deepened with the Covid-19 pandemic, lung disease has turned out to be a major cause of worldwide morbidity and mortality. The condition is exacerbated when the immune system further attacks the healthy, rather than the diseased, tissue within the lung. Governed by unremittingly proliferating mesenchymal cells and increased collagen deposition, if inflammation persists, as frequently occurs in aging lungs, the tissue develops tumors and/or turns into scars (fibrosis), with limited regenerative capacity and organ failure. Fas ligand (FasL, a ligand of the Fas cell death receptor) is a key factor in the regulation of these processes. FasL is primarily found in two forms: full length (membrane, or mFasL) and cleaved (soluble, or sFasL). We and others found that T-cells expressing the mFasL retain autoimmune surveillance that controls mesenchymal, as well as tumor cell accumulation following an inflammatory response. However, mesenchymal cells from fibrotic lungs, tumor cells, or cells from immune-privileged sites, resist FasL+ T-cell-induced cell death. The mechanisms involved are a counterattack of immune cells by FasL, by releasing a soluble form of FasL that competes with the membrane version, and inhibits their cell death, promoting cell survival. This review focuses on understanding the previously unrecognized role of FasL, and in particular its soluble form, sFasL, in the serum of aged subjects, and its association with the evolution of lung disease, paving the way to new methods of diagnosis and treatment.


Asunto(s)
COVID-19/inmunología , Proteína Ligando Fas/inmunología , Enfermedades Pulmonares/inmunología , Pulmón/inmunología , Factores de Edad , Anciano , COVID-19/sangre , Muerte Celular/inmunología , Proteína Ligando Fas/sangre , Humanos , Inmunidad , Enfermedades Pulmonares/sangre , SARS-CoV-2/inmunología , SARS-CoV-2/aislamiento & purificación , Linfocitos T/inmunología
3.
Biomolecules ; 10(7)2020 07 02.
Artículo en Inglés | MEDLINE | ID: mdl-32630813

RESUMEN

In contrast to normal regenerating tissue, resistance to Fas- and FasL-positive T cell-induced apoptosis were detected in myofibroblasts from fibrotic-lungs of humans and mice following bleomycin (BLM) exposure. In this study we show, decreased FLIP expression in lung-tissues with resolution of BLM-induced fibrosis and in isolated-lung fibroblasts, with decreased resistance to apoptosis. Using a FLIP-expression vector or a shFLIP-RNA, we further confirmed the critical need for FLIP to regain/lose susceptibility of fibrotic-lung myofibroblast to Fas-induced apoptosis. Our study further show that FLIP is regulated by SIRT1 (Sirtuin 1) deacetylase. Chimeric mice, with SIRT1-deficiency in deacetylase domain (H355Y-Sirt1y/y), specifically in mesenchymal cells, were not only protected from BLM-induced lung fibrosis but, as assessed following Ku70 immunoprecipitation, had also decreased Ku70-deacetylation, decreasedKu70/FLIP complex, and decreased FLIP levels in their lung myofibroblasts. In addition, myofibroblasts isolated from lungs of BLM-treated miR34a-knockout mice, exposed to a miR34a mimic, which we found here to downregulate SIRT1 in the luciferase assay, had a decreased Ku70-deacetylation indicating decrease in SIRT1 activity. Thus, SIRT1 may mediate, miR34a-regulated, persistent FLIP levels by deacetylation of Ku70 in lung myofibroblasts, promoting resistance to cell-death and lung fibrosis.


Asunto(s)
Bleomicina/efectos adversos , Proteína Reguladora de Apoptosis Similar a CASP8 y FADD/antagonistas & inhibidores , Regulación hacia Abajo , MicroARNs/genética , Fibrosis Pulmonar/terapia , ARN Interferente Pequeño/farmacología , Sirtuina 1/metabolismo , Acetilación/efectos de los fármacos , Animales , Proteína Reguladora de Apoptosis Similar a CASP8 y FADD/metabolismo , Línea Celular , Supervivencia Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Fibroblastos/citología , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Técnicas de Inactivación de Genes , Humanos , Autoantígeno Ku/metabolismo , Pulmón/citología , Pulmón/efectos de los fármacos , Pulmón/metabolismo , Masculino , Ratones , Especificidad de Órganos , Fibrosis Pulmonar/inducido químicamente , Fibrosis Pulmonar/metabolismo
4.
Biomolecules ; 10(7)2020 07 02.
Artículo en Inglés | MEDLINE | ID: mdl-32630842

RESUMEN

Regenerative capacity in vital organs is limited by fibrosis propensity. Idiopathic pulmonary fibrosis (IPF), a progressive lung disease linked with aging, is a classic example. In this study, we show that in flow cytometry, immunoblots (IB) and in lung sections, FLIP levels can be regulated, in vivo and in vitro, through SIRT1 activity inhibition by CMH (4-(4-Chloro-2-methylphenoxy)-N-hydroxybutanamide), a small molecule that, as we determined here by structural biology calculations, docked into its nonhistone substrate Ku70-binding site. Ku70 immunoprecipitations and immunoblots confirmed our theory that Ku70-deacetylation, Ku70/FLIP complex, myofibroblast resistance to apoptosis, cell survival, and lung fibrosis in bleomycin-treated mice, are reduced and regulated by CMH. Thus, small molecules associated with SIRT1-mediated regulation of Ku70 deacetylation, affecting FLIP stabilization in fibrotic-lung myofibroblasts, may be a useful strategy, enabling tissue regeneration.


Asunto(s)
Proteína Reguladora de Apoptosis Similar a CASP8 y FADD/metabolismo , Ácidos Hidroxámicos/administración & dosificación , Fibrosis Pulmonar Idiopática/tratamiento farmacológico , Autoantígeno Ku/metabolismo , Pulmón/citología , Sirtuina 1/química , Sirtuina 1/metabolismo , Acetilación/efectos de los fármacos , Animales , Sitios de Unión , Proteína Reguladora de Apoptosis Similar a CASP8 y FADD/química , Línea Celular , Supervivencia Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Fibroblastos/citología , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Ácidos Hidroxámicos/química , Ácidos Hidroxámicos/farmacología , Fibrosis Pulmonar Idiopática/metabolismo , Pulmón/efectos de los fármacos , Pulmón/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Modelos Moleculares , Simulación del Acoplamiento Molecular , Conformación Proteica , Estabilidad Proteica/efectos de los fármacos
5.
Int J Mol Sci ; 21(6)2020 Mar 23.
Artículo en Inglés | MEDLINE | ID: mdl-32210149

RESUMEN

MicroRNAs (miRs) are known to limit gene expression at the post-transcriptional level and have important roles in the pathogenesis of various conditions, including acute lung injury (ALI) and fibrotic diseases such as idiopathic pulmonary fibrosis (IPF). In this study, we found increased levels of miR-34 at times of fibrosis resolution following injury, in myofibroblasts from Bleomycin-treated mouse lungs, which correlates with susceptibility to cell death induced by immune cells. On the contrary, a substantial downregulation of miR-34 was detected at stages of evolution, when fibroblasts resist cell death. Concomitantly, we found an inverse correlation between miR-34 levels with that of the survival molecule FLICE-like inhibitory protein (FLIP) in lung myofibroblasts from humans with IPF and the experimental model. Forced upregulation of miR-34 with miR-34 mimic in human IPF fibrotic-lung myofibroblasts led to decreased cell survival through downregulation of FLIP. Using chimeric miR-34 knock-out (KO)-C57BL/6 mice with miR34KO myofibroblasts but wild-type (WT) hematopoietic cells, we found, in contrast to WT mice, increased and persistent FLIP levels with a more severe fibrosis and with no signs of resolution as detected in pathology and collagen accumulation. Moreover, a mimic of miR-34a decreased FLIP expression and susceptibility to cell death was regained in miR-34KO fibroblasts. Through this study, we show for the first time an inverse correlation between miR-34a and FLIP expression in myofibroblasts, which affects survival, and accumulation in lung fibrosis. Reprogramming fibrotic-lung myofibroblasts to regain susceptibility to cell-death by specifically increasing their miR34a and downregulating FLIP, may be a useful strategy, enabling tissue regeneration following lung injury.


Asunto(s)
Susceptibilidad a Enfermedades , Fibrosis Pulmonar Idiopática/etiología , Fibrosis Pulmonar Idiopática/patología , Lesión Pulmonar/complicaciones , Lesión Pulmonar/genética , MicroARNs/genética , Animales , Apoptosis , Bleomicina/efectos adversos , Proteína Reguladora de Apoptosis Similar a CASP8 y FADD/genética , Proteína Reguladora de Apoptosis Similar a CASP8 y FADD/metabolismo , Modelos Animales de Enfermedad , Fibroblastos/metabolismo , Regulación de la Expresión Génica , Humanos , Fibrosis Pulmonar Idiopática/metabolismo , Inmunohistoquímica , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Miofibroblastos/metabolismo , Interferencia de ARN
6.
Cells ; 9(2)2020 02 11.
Artículo en Inglés | MEDLINE | ID: mdl-32053892

RESUMEN

A prominent feature of obstructed tissue regeneration following injury in general, and fibrotic lung tissue in particular, is fibroblast proliferation and accumulation. The Fas/FasL apoptotic pathway has been shown to be involved in human idiopathic pulmonary fibrosis (IPF) and bleomycin-induced lung fibrosis in rodents. We previously showed that in normal injury repair, myofibroblasts' accumulation is followed by their decline by FasL+ T cell-induced cell death. In pathological lung fibrosis, myofibroblasts resist cell death and accumulate. Like other members of the tumor necrosis factor (TNF) family, membrane-bound FasL can be cleaved from the cell surface to generate a soluble form (sFasL). Metalloproteinases (MMPs) are known to convert the membrane-bound form of FasL to sFasL. MMP-7 knockout (KO) mice were shown to be protected from bleomycin (BLM)-induced lung fibrosis. In this study, we detected increased levels of sFasL in their blood serum, as in the lungs of patients with IPF, and IPF-lung myofibroblast culture medium. In this study, using an MMP-inhibitor, we showed that sFasL is decreased in cultures of IPF-lung myofibroblasts and BLM-treated lung myofibroblasts, and in the blood serum of MMP-7KO mice. Moreover, resistant fibrotic-lung myofibroblasts, from the lungs of humans with IPF and of BLM-treated mice, became susceptible to T-cell induced cell death in a co-culture following MMP-inhibition- vs. control-treatment or BLM-treated MMP-7KO vs. wild-type mice, respectively. sFasL may be an unrecognized mechanism for MMP-7-mediated decreased tissue regeneration following injury and the evolution of lung fibrosis.


Asunto(s)
Proteína Ligando Fas/genética , Fibrosis Pulmonar Idiopática/genética , Metaloproteinasa 7 de la Matriz/genética , Receptor fas/genética , Animales , Apoptosis/genética , Bleomicina/toxicidad , Muerte Celular/genética , Femenino , Humanos , Fibrosis Pulmonar Idiopática/inducido químicamente , Fibrosis Pulmonar Idiopática/metabolismo , Fibrosis Pulmonar Idiopática/patología , Pulmón/crecimiento & desarrollo , Pulmón/patología , Masculino , Metaloproteinasas de la Matriz , Ratones , Ratones Noqueados , Miofibroblastos/metabolismo , Miofibroblastos/patología , Regeneración/genética , Transducción de Señal/genética
8.
Cells ; 8(10)2019 10 07.
Artículo en Inglés | MEDLINE | ID: mdl-31591327

RESUMEN

CD44, an adhesion-molecule promoting cell-migration, is shown here to increase in stress conditions following bleomycin-induced apoptosis in alveolar epithelial cells (AECs), a main target of lung injury. In vivo, it inhibits tissue regeneration and leads to fibrosis. We show that some AECs survive by the ataxia-telangiectasia mutated kinase/ATM pathway, and undergo a CD44-mediated epithelial-mesenchymal transdifferentiation (EMT) with migratory capacities in vitro, and in vivo. We assessed apoptosis vs. proliferation of AECs following bleomycin, ATM/P53 signaling pathway in AECs, and CD44 involvement in EMT, cell motility and tissue regeneration in vitro and in vivo. Expression of survival genes, CD44, and ATM/p53 pathway was elevated in AECs surviving bleomycin injury, as were the markers of EMT (downregulation of E-cadherin, upregulation of N-cadherin and vimentin, nuclear translocation of ß-catenin). Inhibition of CD44 decreased AECs transdifferentiation. Bleomycin-treated chimeric CD44KO-mice had decreased EMT markers, ATM, and mesenchymal cells (α-SMA+) accumulation in lung, increased surfactant-b, diminished lung mesenchymal cell motility, and increased lung tissue regenerative capacity following bleomycin injury, as indicated by lung collagen content and semiquantitave morphological index scoring. Thus, AECs surviving lung injury are plastic and undergo ATM-mediated, CD44-dependent transdifferentiation, preventing tissue regeneration and promoting fibrosis. Synthetic or natural compounds that downregulate CD44 may improve tissue regeneration following injury.


Asunto(s)
Células Epiteliales Alveolares/fisiología , Transición Epitelial-Mesenquimal/genética , Receptores de Hialuranos/fisiología , Lesión Pulmonar/patología , Regeneración/genética , Células Epiteliales Alveolares/efectos de los fármacos , Animales , Apoptosis/genética , Biomarcadores , Bleomicina , Línea Celular , Técnicas de Inactivación de Genes , Receptores de Hialuranos/antagonistas & inhibidores , Receptores de Hialuranos/genética , Lesión Pulmonar/inducido químicamente , Masculino , Células Madre Mesenquimatosas/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
9.
Int J Mol Sci ; 20(12)2019 Jun 19.
Artículo en Inglés | MEDLINE | ID: mdl-31248154

RESUMEN

Telomeres are distal chromosome regions associated with specific protein complexes that protect the chromosome against degradation and aberrations. Telomere maintenance capacity is an essential indication of healthy cell populations, and telomere damage is observed in processes such as malignant transformation, apoptosis, or cell senescence. At a cellular level, telomere damage may result from genotoxic stress, decreased activity of telomerase enzyme complex, dysfunction of shelterin proteins, or changes in expression of telomere-associated RNA such as TERRA. Clinical evidence suggests that mutation of telomerase genes (Tert/Terc) are associated with increased risk of congenital as well as age-related diseases (e.g., pneumonitis, idiopathic pulmonary fibrosis (IPF), dyskeratosis congenita, emphysema, nonspecific interstitial pneumonia, etc.). Thus, telomere length and maintenance can serve as an important prognostic factor as well as a potential target for new strategies of treatment for interstitial lung diseases (ILDs) and associated pulmonary pathologies.


Asunto(s)
Enfermedades Pulmonares Intersticiales/genética , Enfermedades Pulmonares Intersticiales/metabolismo , Telomerasa/genética , Telomerasa/metabolismo , Telómero/genética , Animales , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Susceptibilidad a Enfermedades , Regulación de la Expresión Génica , Estudios de Asociación Genética , Humanos , Enfermedades Pulmonares Intersticiales/patología , Mutación , ARN Largo no Codificante , Telómero/metabolismo
10.
Mediators Inflamm ; 2018: 4638437, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30002599

RESUMEN

Lung fibrosis is characterized by abnormal accumulation of Thy-deficient fibroblasts in the interstitium of the alveolar space. We have previously shown in bleomycin-treated chimeric Thy1-deficient mice with wild-type lymphocytes that Thy1-deficient fibroblasts accumulate and promote fibrosis and an "inflammation-free" environment. Here, we aimed to identify the critical effects of Thy1, or the absence of Thy1, in lung myofibroblast profibrotic functions, particularly proliferation and collagen deposition. Using specific Thy1 siRNA in Thy1-positive cells, Thy1 knockout cells, Thy1 cDNA expression vector in Thy1-deficient cells, and Thy1 cross-linking, we evaluated cell proliferation (assessed by cell mass and BrdU uptake), differentiation (using immunofluorescence), and collagen deposition (using Sircol assay). We found that myofibroblast Thy1 cross-linking and genetic manipulation modulate cell proliferation and expression of Fgf (fibroblast growth factor) and Angtl (angiotensin) receptors (using qPCR) that are involved in myofibroblast proliferation, differentiation, and collagen deposition. In conclusion, lung myofibroblast downregulation of Thy1 expression is critical to increase proliferation, differentiation, and collagen deposition.


Asunto(s)
Pulmón/citología , Miofibroblastos/efectos de los fármacos , Miofibroblastos/metabolismo , Antígenos Thy-1/metabolismo , Animales , Bleomicina/farmacología , Ciclo Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Droperidol/farmacología , Citometría de Flujo , Ketamina/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Microscopía Confocal
11.
Mediators Inflamm ; 2015: 942179, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26300593

RESUMEN

Lung fibrosis is characterized by abnormal accumulation of fibroblasts in the interstitium of the alveolar space. Two populations of myofibroblasts, distinguished by Thy1 expression, are detected in human and murine lungs. Accumulation of Thy1-negative (Thy1(-)) myofibroblasts was shown in the lungs of humans with idiopathic pulmonary fibrosis (IPF) and of bleomycin-treated mice. We aimed to identify genetic changes in lung myofibroblasts following Thy1 crosslinking and assess the impact of specific lung myofibroblast Thy1-deficiency, in vivo, in bleomycin-injured mouse lungs. Thy1 increased in mouse lung lymphocytes following bleomycin injury but decreased in myofibroblasts when fibrosis was at the highest point (14 days), as assessed by immunohistochemistry. Using gene chip analysis, we detected that myofibroblast Thy1 crosslinking mediates downregulation of genes promoting cell proliferation, survival, and differentiation, and reduces production of extracellular matrix (ECM) components, while concurrently mediating the upregulation of genes known to foster inflammation and immunological functions. Chimeric Thy1-deficient mice with Thy1(+) lymphocytes and Thy1(-) myofibroblasts showed fibrosis similar to wild-type mice and an increased number of CD4/CD25 regulatory T cells, with a concomitant decrease in inflammation. Lung myofibroblasts downregulate Thy1 expression to increase their proliferation but to diminish the in vivo inflammatory milieu. Inflammation is not essential for evolution of fibrosis as was previously stated.


Asunto(s)
Bleomicina/farmacología , Inflamación/prevención & control , Linfocitos/fisiología , Miofibroblastos/fisiología , Fibrosis Pulmonar/etiología , Antígenos Thy-1/fisiología , Animales , Humanos , Pulmón/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Análisis de Secuencia por Matrices de Oligonucleótidos
12.
PLoS One ; 10(5): e0126730, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25951185

RESUMEN

High doses of bleomycin administered to patients with lymphomas and other tumors lead to significant lung toxicity in general, and to apoptosis of epithelial cells, in particular. Apoptosis of alveolar epithelium is an important step in the pathogenesis of bleomycin-induced pulmonary fibrosis. The Fas-FasL pathway is one of the main apoptotic pathways involved. Telomerase is a ribonucleoprotein RNA-dependent DNA polymerase complex consisting of an RNA template and a catalytic protein, telomerase reverse transcriptase (TERT). Telomerase also possess extra-telomeric roles, including modulation of transcription of anti-apoptotic genes, differentiation signals, and more. We hypothesized that telomerase overexpression affects Fas-induced epithelial cell apoptosis by an extra-telomeric role such as regulation of anti-apoptotic genes, specifically FLICE-like inhibitory protein (FLIP). Telomerase in mouse (MLE) and human (A549) lung epithelial cell lines was upregulated by transient transfection using cDNA hTERT expression vector. Telomerase activity was detected using a real-time PCR-based system. Bleomycin, and bleomycin-induced Fas-mediated apoptosis following treatment with anti-Fas activating mAb or control IgG, were assessed by Annexin V staining, FACS analysis, and confocal microscopy; caspase cleavage by Western blot; FLIP or Fas molecule detection by Western blot and flow cytometry. hTERT transfection of lung epithelial cells resulted in a 100% increase in their telomerase activity. Fas-induced lung epithelial cell apoptosis was significantly reduced in hTERT-transfected cells compared to controls in all experiments. Lung epithelial cells with increased telomerase activity had higher levels of FLIP expression but membrane Fas expression was unchanged. Upregulation of hTERT+ in human lung epithelial cells and subsequent downregulation of FLIP by shFLIP-RNA annulled hTERT-mediated resistance to apoptosis. Telomerase-mediated FLIP overexpression may be a novel mechanism to confer protection from apoptosis in bleomycin-exposed human lung epithelial cells.


Asunto(s)
Antibióticos Antineoplásicos/efectos adversos , Bleomicina/efectos adversos , Proteína Reguladora de Apoptosis Similar a CASP8 y FADD/genética , Células Epiteliales/efectos de los fármacos , Telomerasa/genética , Receptor fas/metabolismo , Animales , Apoptosis/efectos de los fármacos , Proteína Reguladora de Apoptosis Similar a CASP8 y FADD/metabolismo , Línea Celular , Células Epiteliales/metabolismo , Células Epiteliales/patología , Humanos , Pulmón/citología , Pulmón/efectos de los fármacos , Pulmón/metabolismo , Pulmón/patología , Ratones , Telomerasa/metabolismo , Transfección , Regulación hacia Arriba
13.
J Autoimmun ; 59: 67-76, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25812467

RESUMEN

Immune cells, particularly those expressing the ligand of the Fas-death receptor (FasL), e.g. cytotoxic T cells, induce apoptosis in 'undesirable' self- and non-self-cells, including lung fibroblasts, thus providing a means of immune surveillance. We aimed to validate this mechanism in resolution of lung fibrosis. In particular, we elucidated whether FasL(+) immune cells possess antifibrotic capabilities by induction of FasL-dependent myofibroblast apoptosis and whether antagonists of membrane (m) and soluble (s) FasL can inhibit these capabilities. Myofibroblast interaction with immune cells and its FasL-dependency, were investigated in vitro in coculture with T cells and in vivo, following transplantation into lungs of immune-deficient syngeneic Rag-/- as well as allogeneic SCID mice, and into lungs and air pouches of FasL-deficient (gld) mice, before and after reconstitution of the mice with wild-type (wt), FasL(+) immune cells. We found that myofibroblasts from lungs resolving fibrosis undergo FasL-dependent T cell-induced apoptosis in vitro and demonstrate susceptibility to in vivo immune surveillance in lungs of reconstituted, immune- and FasL-deficient, mice. However, immune-deficient Rag-/- and SCID mice, and gld-mice with FasL-deficiency, endure the accumulation of transplanted myofibroblasts in their lungs with subsequent development of fibrosis. Concomitantly, gld mice, in contrast to chimeric FasL-deficient mice with wt immune cells, accumulated transplanted myofibroblasts in the air pouch model. In humans we found that myofibroblasts from fibrotic lungs secrete sFasL and resist T cell-induced apoptosis, whereas normal lung myofibroblasts are susceptible to apoptosis but acquire resistance upon addition of anti-s/mFasL to the coculture. Immune surveillance, particularly functional FasL(+) immune cells, may represent an important extrinsic component in myofibroblast apoptosis and serve as a barrier to fibrosis. Factors interfering with Fas/FasL-immune cell-myofibroblast interaction such as sFasL secreted by fibrotic-lung myofibroblasts, may abrogate immune surveillance during fibrosis. Annulling these factors may pave a new direction to control human lung fibrosis.


Asunto(s)
Apoptosis , Proteína Ligando Fas/metabolismo , Pulmón/patología , Miofibroblastos/metabolismo , Linfocitos T Citotóxicos/inmunología , Traslado Adoptivo , Animales , Anticuerpos Bloqueadores/farmacología , Apoptosis/efectos de los fármacos , Células Cultivadas , Técnicas de Cocultivo , Proteína Ligando Fas/genética , Fibrosis , Genes RAG-1/genética , Humanos , Vigilancia Inmunológica/efectos de los fármacos , Pulmón/inmunología , Ratones , Ratones Noqueados , Ratones SCID , Miofibroblastos/efectos de los fármacos , Miofibroblastos/patología , Linfocitos T Citotóxicos/trasplante
14.
Transpl Immunol ; 27(4): 184-8, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22801052

RESUMEN

Formation of donor-recipient mixed chimerism after nonmyeloablative conditioning allows co-existence of donor and recipient hematopoietic stem cells, with solid organ allograft tolerance and less likeliness of graft versus host development. Using a post-transplant bronchiolitis obliterans murine model, we aimed to test the hypothesis that allograft preservation after mixed chimerism formation is dependent on the presence of a functional Fas ligand (FasL) on donor hematopoietic cells. To form mixed chimerism, two aliquots of 30 × 10(6) whole bone marrow cells (BMC) from either wild-type C57BL/6 in one group, or transgenic gld mice with mutant FasL (d = 0 and 2+) in the other were used, with both groups receiving intravenous busulfan (10mg/kg) on d-1 and intraperitoneal cyclophosphamide (200mg/kg) on d+1. Tracheal allografts obtained from C57BL/6 mice were implanted into recipient BALB/c mice subcutaneously on d = 0. Tracheal allografts were harvested at d+28 post-transplant and were evaluated by histopathology. Mixed chimerism formation was detected in wild type C57BL/6 whole BMC recipients, which was accompanied by tracheal allograft acceptance with near normal structure at d+28 post implantation. However, in recipients of FasL mutant whole BMC, neither mixed chimerism formation nor tracheal allograft acceptance was obtained. We thus conclude that bone marrow cells lacking functional FasL molecules could not be engrafted in allogeneic recipients to form stable mixed chimerism after nonmyeloablative conditioning, thus not allowing tracheal allograft acceptance.


Asunto(s)
Células de la Médula Ósea/inmunología , Proteína Ligando Fas/genética , Proteína Ligando Fas/inmunología , Quimera por Trasplante/inmunología , Acondicionamiento Pretrasplante/métodos , Tolerancia al Trasplante/inmunología , Animales , Trasplante de Células Madre Hematopoyéticas , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Mutantes , Ratones Transgénicos , Mutación , Donantes de Tejidos , Tráquea/patología , Tráquea/trasplante , Trasplante Homólogo
15.
Am J Respir Cell Mol Biol ; 47(3): 271-9, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22582174

RESUMEN

A prominent feature of fibrotic tissue in general and of lungs in particular is fibroblast proliferation and accumulation. In patients overcoming fibrosis, apoptosis limits this excessive cell growth. We have previously shown resistance to Fas-induced apoptosis of primary lung fibroblasts from mice with bleomycin-induced lung fibrosis, their escape from immune surveillance, and continued accumulation in spite of overexpression of the Fas death receptor. Cellular FLICE-like inhibitory protein (c-FLIP) is a regulator of cell death receptor-induced apoptosis in many cell types. We aimed to determine c-FLIP levels in myofibroblasts from fibrotic lungs and to directly assess c-FLIP's role in apoptosis and proliferation of primary lung myofibroblasts. c-FLIP levels were determined by apoptosis gene array, flow cytometry, Western blot, and immunofluorescence before and after down-regulation with a specific small interfering RNA. Apoptosis was assessed by caspase cleavage in Western blot and by Annexin V affinity labeling after FACS and tissue immunofluorescence. Proliferation was assessed by BrdU uptake, also using FACS and immunofluorescence. We show that myofibroblasts from lungs of humans with idiopathic pulmonary fibrosis and from bleomycin-treated versus normal saline-treated mice up-regulate c-FLIP levels. Using the animal model, we show that fibrotic lung myofibroblasts divert Fas signaling from apoptosis to proliferation and that this requires signaling by TNF receptor-associated factor (TRAF) and NF-κB. c-FLIP down-regulation reverses the effect of Fas activation, causing increased apoptosis, decreased proliferation, and diminished recruitment of TRAF to the DISC complex. This indicates that c-FLIP is essential for myofibroblast accumulation and may serve as a potential target to manipulate tissue fibrosis.


Asunto(s)
Apoptosis/fisiología , Proteína Reguladora de Apoptosis Similar a CASP8 y FADD/fisiología , Proliferación Celular , Miofibroblastos/patología , Fibrosis Pulmonar/patología , Receptor fas/fisiología , Animales , Anexina A5/metabolismo , Secuencia de Bases , Western Blotting , Proteína Reguladora de Apoptosis Similar a CASP8 y FADD/metabolismo , Caspasas/metabolismo , Cartilla de ADN , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Ratones , Ratones Endogámicos C57BL , FN-kappa B/metabolismo , Ratas , Ratas Sprague-Dawley , Transducción de Señal
16.
Am J Respir Cell Mol Biol ; 40(2): 231-8, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18676775

RESUMEN

We have previously demonstrated that myofibroblasts from lungs with bleomycin-induced fibrosis overexpress FasL molecules. Two subpopulations of fibroblasts, distinguished by their expression of Thy1 molecules, have been shown in the lungs of both mice and humans. Thy1-mediated FasL induction has been reported in T cells through the use of anti-Thy1 (G7). We therefore sought to determine whether FasL expression in lung myofibroblasts is associated with and/or dependent on Thy1 expression, and to examine the underlying mechanism of regulation. We show that myofibroblast Thy1 expression is associated with increased FasL expression. Moreover, we directly show that Thy1 activation induces FasL up-regulation. Initially, Thy1 activation causes translocation of FasL to the membrane surface, and later induces de novo synthesis of FasL at the mRNA and protein levels. In contrast to Thy1 activation, TNF-alpha and IFN-gamma do not induce FasL myofibroblast up-regulation. Using Src family kinase (SFKs) inhibitor (PP2), we show the general involvement of SFKs in Thy1 signal transduction leading to FasL up-regulation; and, using specific siRNA, we show the particular involvement of Fyn, one protein in the SFK family. These results demonstrate that Thy1 in myofibroblasts is not just a marker, but is a functional protein that transmits signals into the cell, up-regulating its FasL expression.


Asunto(s)
Proteína Ligando Fas/biosíntesis , Fibroblastos/metabolismo , Mioblastos/metabolismo , Transducción de Señal/fisiología , Antígenos Thy-1/metabolismo , Regulación hacia Arriba/fisiología , Animales , Antivirales/farmacología , Fibroblastos/citología , Humanos , Interferón gamma/farmacología , Masculino , Ratones , Mioblastos/citología , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas c-fyn/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-fyn/metabolismo , Pirimidinas/farmacología , ARN Interferente Pequeño , Transducción de Señal/efectos de los fármacos , Factor de Necrosis Tumoral alfa/farmacología , Regulación hacia Arriba/efectos de los fármacos
17.
Semin Cancer Biol ; 18(4): 260-7, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18467123

RESUMEN

Tumor progression is substantially dependent on network of multiple factors, including adhesion and homing molecules, which support the malignant metastatic spread. CD44, one of the adhesion/homing molecules, has attracted much attention not only because it is expressed on many types of tumors, but also owing to its numerous functions, such as supporting cell migration and transmitting survival signals, thereby being pro-oncogenic by nature. We have used the mouse malignant LB lymphoma cell line as a model for comprehensive in vitro and in vivo analyses of the interaction between CD44 and hyaluronic acid (HA), and its relevance to tumor dissemination. The in vitro studies revealed that LB cells could not bind HA, either under static or dynamic (i.e., shear flow) conditions, unless their CD44 is activated by phorbol ester, deglycosylated (to increase the CD44 positive net charge) or transfected with CD44 variants. In parallel, in vivo experiments showed that LB cell dissemination could be controlled by injection of anti-CD44 monoclonal antibodies or hyaluronidase. Furthermore, LB cells transfected with CD44v4-v10 variant, rather than standard CD44, displayed enhanced invasion of the peripheral lymph nodes. This effect was completely lost if the HA binding site of CD44 were mutated. LB cell accumulation in the lymph nodes is caused by enhanced migration via the afferent lymphatics rather than by accelerated proliferation within the lymph node. This information can be exploited to tailor a "therapeutic suit" that should be maximally effective in inducing tumor resistance, while minimizing destructive side effects.


Asunto(s)
Receptores de Hialuranos/fisiología , Linfoma/metabolismo , Animales , Humanos , Ácido Hialurónico/metabolismo
18.
Proc Natl Acad Sci U S A ; 104(51): 20460-5, 2007 Dec 18.
Artículo en Inglés | MEDLINE | ID: mdl-18077384

RESUMEN

Tissue fibrosis evolving from impaired tissue remodeling after injury is characterized by myofibroblast accumulation. We propose that during the development of fibrosis myofibroblasts acquire an immune-privileged cell phenotype, allowing their uninterrupted accumulation. Using the murine model of bleomycin-induced lung fibrosis in mice, we show that myofibroblasts that accumulate in lungs with fibrosis, but not in normal lungs, kill Fas(+) lymphocytes, resist Fas-induced apoptosis, and survive longer when grafted into allogeneic mice. In contrast, bleomycin-treated FasLigand (FasL)-deficient (gld) chimeric mice did not accumulate myofibroblasts or collagen in their lungs, and their FasL(-) myofibroblasts did not survive after alloengraftment. This finding indicates that myofibroblasts possess Fas/FasL-pathway-dependent characteristics that allow them to escape from immune surveillance and resulting organ fibrosis.


Asunto(s)
Fibroblastos/inmunología , Vigilancia Inmunológica , Enfermedades Pulmonares/inmunología , Enfermedades Pulmonares/patología , Mioblastos/inmunología , Actinas/metabolismo , Animales , Apoptosis , Colágeno/metabolismo , Proteína Ligando Fas/genética , Fibrosis , Enfermedades Pulmonares/inducido químicamente , Linfocitos/inmunología , Ratones , Ratones Mutantes , Receptor fas/genética , Receptor fas/metabolismo
19.
Am J Respir Cell Mol Biol ; 36(3): 270-5, 2007 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-16990614

RESUMEN

The Fas/Fas ligand (FasL) apoptotic pathway has been shown to be involved in bleomycin-induced lung fibrosis. We examined the hypothesis that myofibroblasts from fibrotic lungs possess a cytotoxic phenotype that causes apoptosis of epithelial cells via the Fas/FasL pathway. We show in vivo epithelial cell apoptosis and associated upregulation of Fas and apoptotic Fas pathway genes in epithelial cells of lungs with bleomycin-induced fibrosis. In addition, we show that FasL surface molecules are overexpressed on alpha-SMA-positive cells in mice with bleomycin-induced fibrosis, and in humans with idiopathic pulmonary fibrosis. This enables the molecules to kill Fas-positive epithelial cells. In contrast, FasL-deficient myofibroblasts lose this myofibroblast cytotoxic phenotype, both in vivo and in vitro. In vivo, there was no bleomycin-induced epithelial cell apoptosis, as assessed by specific M30 staining in chimeric FasL-deficient mice that lacked FasL-positive myofibroblasts. In vitro, FasL-positive, but not FasL-negative myofibroblasts, induce mouse lung epithelial cell apoptosis. Thus myofibroblast cytotoxicity may underlie the absence of re-epithelialization, resulting in persistent lung fibrosis.


Asunto(s)
Apoptosis , Células Epiteliales/citología , Proteína Ligando Fas/metabolismo , Fibroblastos/patología , Fibrosis Pulmonar/patología , Animales , Apoptosis/efectos de los fármacos , Bleomicina/farmacología , Quimera , Técnicas de Cocultivo , Células Epiteliales/efectos de los fármacos , Proteína Ligando Fas/deficiencia , Proteína Ligando Fas/genética , Fibroblastos/efectos de los fármacos , Expresión Génica/efectos de los fármacos , Humanos , Linfocitos/citología , Linfocitos/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Receptor fas/genética , Receptor fas/metabolismo
20.
Am J Physiol Lung Cell Mol Physiol ; 290(4): L790-L796, 2006 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-16306138

RESUMEN

Epithelial cells are considered to be a main target of bleomycin-induced lung injury, which leads to fibrosis in vivo. We studied the characteristics of in vitro bleomycin-induced apoptosis in a mouse lung epithelial (MLE) cell line. Bleomycin caused an increase of reactive oxygen species (ROS) resulting in oxidative stress, mitochondrial leakage, and apoptosis. These were associated with elevated caspase-8 and resultant caspase-9 activity and with upregulation of Fas expression. Glutathione and inhibitors of caspase-8 or caspase-9, but not of FasL, inhibited these effects, suggesting their dependence on ROS, caspase-8 and -9, in a Fas/FasL-independent pathway. However, postbleomycin-exposed MLE cells were more sensitive to Fas-mediated apoptosis. These results demonstrate that the initial bleomycin-induced oxidative stress causes a direct apoptotic effect in lung epithelial cells involving a regulatory role of caspase-8 on caspase-9. Fas represents an amplification mechanism, and not a direct trigger of bleomycin-induced epithelial cell apoptosis.


Asunto(s)
Antimetabolitos Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Bleomicina/farmacología , Pulmón/fisiología , Glicoproteínas de Membrana/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Factores de Necrosis Tumoral/metabolismo , Receptor fas/metabolismo , Animales , Antimetabolitos Antineoplásicos/administración & dosificación , Bleomicina/administración & dosificación , Caspasa 8 , Caspasa 9 , Inhibidores de Caspasas , Caspasas/metabolismo , Muerte Celular/efectos de los fármacos , Línea Celular , Relación Dosis-Respuesta a Droga , Activación Enzimática/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Células Epiteliales/fisiología , Proteína Ligando Fas , Glutatión/farmacología , Pulmón/citología , Potenciales de la Membrana/efectos de los fármacos , Ratones , Mitocondrias/fisiología , Regulación hacia Arriba
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...