Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
Exp Lung Res ; 42(3): 121-30, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-27093376

RESUMO

UNLABELLED: Purpose/Aim of Study: The renin angiotensin system is involved in experimentally induced lung fibrosis. Angiotensin (ANG)-II is profibrotic. Angiotensin converting enzyme-2 (ACE-2) cleaves ANG-II and is thus protective. ACE-2 has recently been reported to be significantly decreased under hyperoxic conditions. Hyperoxia is linked to Bronchopulmonary Dysplasia and lung fibrosis. Fetal lung cells normally do not undergo fibrotic changes with physiologic hypoxemia. We hypothesized that hypoxia prior to hyperoxic exposure in fetal lung fibroblasts (IMR-90 cell line) might be protective by preventing ACE-2 downregulation. MATERIALS AND METHODS: IMR-90 cells were exposed to hypoxia (1%O2/99%N2) followed by hyperoxia (95%O2/5%CO2) or normoxia (21%O2) in vitro. Cells and culture media were recovered separately for assays of ACE-2, TNF-α-converting enzyme (TACE), αSmooth muscle actin (αSMA)-myofibroblast marker-, N-cadherin, and ß-catenin immunoreactive protein. RESULTS: ACE-2 significantly increased when IMR-90 were hypoxic prior to hyperoxic exposure with no recovery. In contrast to hyperoxia alone, ACE-2 did not decrease when IMR-90 were hypoxic prior to hyperoxic exposure with recovery. TACE/ADAM17 protein and mRNA were significantly decreased under these conditions. αSMA N-cadherin, and ß-catenin proteins were significantly decreased with or without normoxic recovery. CONCLUSIONS: Hypoxia prior to hyperoxic exposure of fetal lung fibroblasts prevented ACE-2 downregulation and decreased ADAM17/TACE protein and mRNA. αSMA, N-cadherin, and ß-catenin were also significantly decreased under these conditions.


Assuntos
Regulação para Baixo/fisiologia , Fibroblastos/fisiologia , Hiperóxia/fisiopatologia , Hipóxia/fisiopatologia , Pulmão/fisiologia , Peptidil Dipeptidase A/metabolismo , Actinas/metabolismo , Angiotensina II/metabolismo , Enzima de Conversão de Angiotensina 2 , Caderinas/metabolismo , Células Cultivadas , Fibroblastos/metabolismo , Humanos , Hiperóxia/metabolismo , Hipóxia/metabolismo , Pulmão/metabolismo , Fibrose Pulmonar/metabolismo , Fibrose Pulmonar/fisiopatologia , Fator de Necrose Tumoral alfa/metabolismo , beta Catenina/metabolismo
2.
Bioengineering (Basel) ; 11(6)2024 May 21.
Artigo em Inglês | MEDLINE | ID: mdl-38927760

RESUMO

Biological aging is defined as a progressive decline in tissue function that eventually results in cell death. Accelerated biologic aging results when the telomere length is shortened prematurely secondary to damage from biological or environmental stressors, leading to a defective reparative mechanism. Stem cells therapy may have a potential role in influencing (counteract/ameliorate) biological aging and maintaining the function of the organism. Mesenchymal stem cells, also called mesenchymal stromal cells (MSCs) are multipotent stem cells of mesodermal origin that can differentiate into other types of cells, such as adipocytes, chondrocytes, and osteocytes. MSCs influence resident cells through the secretion of paracrine bioactive components such as cytokines and extracellular vesicles (EVs). This review examines the changes in telomere length, cellular senescence, and normal biological age, as well as the factors contributing to telomere shortening and accelerated biological aging. The role of MSCs-especially those derived from gestational tissues-in prevention of telomere shortening (TS) and accelerated biological aging is explored. In addition, the strategies to prevent MSC senescence and improve the antiaging therapeutic application of MSCs and MSC-derived EVs in influencing telomere length and cellular senescence are reviewed.

3.
Eur Respir J ; 42(1): 198-210, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23100504

RESUMO

Alveolar epithelial type II cells, a major source of angiotensin-converting enzyme (ACE)-2 in the adult lung, are normally quiescent but actively proliferate in lung fibrosis and downregulate this protective enzyme. It was, therefore, hypothesised that ACE-2 expression might be related to cell cycle progression. To test this hypothesis, ACE-2 mRNA levels, protein levels and enzymatic activity were examined in fibrotic human lungs and in the alveolar epithelial cell lines A549 and MLE-12 studied at postconfluent (quiescent) versus subconfluent (proliferating) densities. ACE-2 mRNA, immunoreactive protein and enzymatic activity were all high in quiescent cells, but were severely downregulated or absent in actively proliferating cells. Upregulation of the enzyme in cells that were progressing to quiescence was completely inhibited by the transcription blocker actinomycin D or by SP600125, an inhibitor of c-Jun N-terminal kinase (JNK). In lung biopsy specimens obtained from patients with idiopathic pulmonary fibrosis, immunoreactive enzyme was absent in alveolar epithelia that were positive for proliferation markers, but was robustly expressed in alveolar epithelia devoid of proliferation markers. These data explain the loss of ACE-2 in lung fibrosis and demonstrate cell cycle-dependent regulation of this protective enzyme by a JNK-mediated transcriptional mechanism.


Assuntos
Ciclo Celular , Fibrose Pulmonar Idiopática/metabolismo , Fibrose Pulmonar Idiopática/fisiopatologia , Pulmão/patologia , Peptidil Dipeptidase A/metabolismo , Enzima de Conversão de Angiotensina 2 , Animais , Apoptose , Linhagem Celular Tumoral , Proliferação de Células , Regulação para Baixo , Citometria de Fluxo , Regulação da Expressão Gênica , Humanos , Imuno-Histoquímica , Pulmão/metabolismo , Camundongos , Peptídeos/química , Alvéolos Pulmonares/metabolismo , RNA Mensageiro/metabolismo
4.
Cells ; 12(2)2023 01 07.
Artigo em Inglês | MEDLINE | ID: mdl-36672185

RESUMO

BACKGROUND: Mesenchymal stem cells (MSCs) have been used for ex vivo expansion of umbilical cord blood (UCB) hematopoietic stem cells (HSCs) to maintain their primitive characters and long-term reconstitution abilities during transplantation. Therapeutic effects of MSCs mainly rely on paracrine mechanisms, including secretion of exosomes (Exos). The objective of this study was to examine the effect of cord blood plasma (CBP)-derived Exos (CBP Exos) and Placental MSCs-derived Exos (MSCs Exos) on the expansion of UCB HSCs to increase their numbers and keep their primitive characteristics. METHODS: CD34+ cells were isolated from UCB, cultured for 10 days, and the expanded HSCs were sub-cultured in semisolid methylcellulose media for primitive colony forming units (CFUs) assay. MSCs were cultured from placental chorionic plates. RESULTS: CBP Exos and MSCs Exos compared with the control group significantly increased the number of total nucleated cells (TNCs), invitro expansion of CD34+ cells, primitive subpopulations of CD34+38+ and CD34+38-Lin- cells (p < 0.001). The expanded cells showed a significantly higher number of total CFUs in the Exos groups (p < 0.01). CONCLUSION: CBP- and placental-derived exosomes are associated with significant ex vivo expansion of UCB HSCs, while maintaining their primitive characters and may eliminate the need for transplantation of an additional unit of UCB.


Assuntos
Exossomos , Células-Tronco Mesenquimais , Humanos , Feminino , Gravidez , Sangue Fetal , Placenta , Proliferação de Células , Células-Tronco Hematopoéticas
5.
Cells ; 11(8)2022 04 09.
Artigo em Inglês | MEDLINE | ID: mdl-35455954

RESUMO

Premature newborns are at a higher risk for the development of respiratory distress syndrome (RDS), acute lung injury (ALI) associated with lung inflammation, disruption of alveolar structure, impaired alveolar growth, lung fibrosis, impaired lung angiogenesis, and development of bronchopulmonary dysplasia (BPD) with severe long-term developmental adverse effects. The current therapy for BPD is limited to supportive care including high-oxygen therapy and pharmacotherapy. Recognizing more feasible treatment options to improve lung health and reduce complications associated with BPD is essential for improving the overall quality of life of premature infants. There is a reduction in the resident stem cells in lungs of premature infants with BPD, which strongly suggests a critical role of stem cells in BPD pathogenesis; this warrants the exploration of the potential therapeutic use of stem-cell therapy. Stem-cell-based therapies have shown promise for the treatment of many pathological conditions including acute lung injury and BPD. Mesenchymal stem cells (MSCs) and MSC-derived extracellular vesicles (EVs) including exosomes are promising and effective therapeutic modalities for the treatment of BPD. Treatment with MSCs and EVs may help to reduce lung inflammation, improve pulmonary architecture, attenuate pulmonary fibrosis, and increase the survival rate.


Assuntos
Lesão Pulmonar Aguda , Displasia Broncopulmonar , Transplante de Células-Tronco Mesenquimais , Fibrose Pulmonar , Animais , Displasia Broncopulmonar/patologia , Displasia Broncopulmonar/terapia , Modelos Animais de Doenças , Humanos , Lactente , Recém-Nascido , Qualidade de Vida
6.
Am J Physiol Lung Cell Mol Physiol ; 301(3): L269-74, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21665960

RESUMO

Earlier work from this laboratory demonstrated that apoptosis of alveolar epithelial cells (AECs) requires autocrine generation of angiotensin (ANG) II. More recent studies showed that angiotensin converting enzyme-2 (ACE-2), which degrades ANGII to form ANG1-7, is protective but severely downregulated in human and experimental lung fibrosis. Here it was theorized that ACE-2 and its product ANG1-7 might therefore regulate AEC apoptosis. To evaluate this hypothesis, the AEC cell line MLE-12 and primary cultures of rat AECs were exposed to the profibrotic apoptosis inducers ANGII or bleomycin (Bleo). Markers of apoptosis (caspase-9 or -3 activation and nuclear fragmentation), steady-state ANGII and ANG1-7, and JNK phosphorylation were measured thereafter. In the absence of Bleo, inhibition of ACE-2 by small interfering RNA or by a competitive inhibitor (DX600 peptide) caused a reciprocal increase in autocrine ANGII and corresponding decrease in ANG1-7 in cell culture media (both P < 0.05) and, moreover, induced AEC apoptosis. At baseline (without inhibitor), ANG1-7 in culture media was 10-fold higher than ANGII (P < 0.01). Addition of purified ANGII or bleomycin-induced caspase activation, nuclear fragmentation, and JNK phosphorylation in cultured AECs. However, preincubation with ANG1-7 (0.1 µM) prevented JNK phosphorylation and apoptosis. Moreover, pretreatment with A779, a specific blocker of the ANG1-7 receptor mas, prevented ANG1-7 blockade of JNK phosphorylation, caspase activation, and nuclear fragmentation. These data demonstrate that ACE-2 regulates AEC survival by balancing the proapoptotic ANGII and its antiapoptotic degradation product ANG1-7. They also suggest that ANG1-7 inhibits AEC apoptosis through the ANG1-7 receptor mas.


Assuntos
Angiotensina I/fisiologia , Sobrevivência Celular/efeitos dos fármacos , Células Epiteliais/fisiologia , Fragmentos de Peptídeos/fisiologia , Peptidil Dipeptidase A/fisiologia , Proteínas Proto-Oncogênicas/fisiologia , Receptores Acoplados a Proteínas G/fisiologia , Enzima de Conversão de Angiotensina 2 , Animais , Apoptose/efeitos dos fármacos , Bleomicina/antagonistas & inibidores , Bleomicina/farmacologia , Linhagem Celular , Células Epiteliais/efeitos dos fármacos , Camundongos , Proto-Oncogene Mas , Alvéolos Pulmonares/citologia , Ratos
7.
J Lung Pulm Respir Res ; 8: 47-52, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34825051

RESUMO

BACKGROUND: Bronchopulmonary Dysplasia (BPD) occurs in premature neonates with respiratory distress who require supplemental oxygen in the first days after birth. BPD involves uniform arrest of alveolar development and variable interstitial cellularity and/or fibroproliferation. Previous studies by our lab showed that the enzyme, angiotensin converting enzyme-2 (ACE-2) and its product Ang1-7 exerting action on the receptor Mas oncogene in what is known as ACE-2/Mas axis is protective to lung cells. We also showed that ACE-2 is expressed in fetal human lung fibroblasts but is significantly decreased by hyperoxic gas lung injury, an effect caused by ACE-2 enzyme shedding mediated by TNF-alpha-converting enzyme (TACE/ADAM17). However, no reports yet exist about the regulation of ACE-2 in the alveolar epithelia in hyperoxic lung injury. OBJECTIVE: In this study we aim to define the effects of hyperoxic lung injury on the protective ACE-2 enzyme in the human lung alveolar epithelial cell line A549. DESIGN/METHODS: Cultured A549 cells were exposed to hyperoxia (95% O2) or normoxia (21% O2) for 3 or 7 days in serum-free nutrient media. Cells were lysed and culture media were collected to test for cellular ACE-2 enzymatic activity and for ACE-2, Mas receptor, TACE/ADAM17, and ubiquitin proteins abundance by immunoblotting. Cells were harvested in Trizol for RNA extraction and ACE-2 qRT-PCR. Whole cell extracts of A549 cell line was used for ACE-2 immunoprecipitation and subsequent ubiquitin immunoblotting. RESULTS: Total ubiquitinated proteins were increased by hyperoxia treatment, while ACE-2 and Mas receptor proteins abundance and ACE-2 enzymatic activity were decreased significantly in A549 cells exposed to hyperoxia relative to the normoxia controls. The percent decrease in ACE-2 activity corresponded with increased time of hyperoxic gas exposure. However, in contrast to our data from lung fibroblasts, no significant change was noted in ACE-2 protein released into the media or in ACE-2 mRNA levels by the hyperoxic treatment. Ubiquitin immunoreactive bands were detectable in the ACE-2 immunoprecipitate. CONCLUSIONS: These data suggest that hyperoxic exposure of the lung epithelial cells decreases the protective enzyme ACE-2 by cell type specific mechanisms independent of shedding by TACE/ADAM17. The data also suggest a regulatory level of ACE-2 downstream of transcription may involve ACE-2 ubiquitination and targeting for degradation.

8.
J Lung Pulm Respir Res ; 8(2): 54-60, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34414260

RESUMO

Coronavirus Disease 2019 (COVID-19) is caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Like the 2002-2003 epidemic severe acute respiratory syndrome coronavirus (SARS-CoV), angiotensin converting enzyme-2 (ACE-2) has been identified as the SARS-CoV-2 receptor.1-3 The virus docks into host cell via its spike protein binding to ACE-2 and undergoes proteolytic cleavage by TMPRSS2 protease to facilitate membrane fusion. The spike protein binding to ACE-2 has been shown to be stronger in the novel SARS-CoV-2 virus.1 This review will present an overview of ACE-2 biology.

9.
FASEB J ; 23(6): 1655-62, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19211927

RESUMO

Earlier work showed that TGF-beta1 potently increases angiotensinogen (AGT) gene mRNA in primary human lung fibroblasts. Here the mechanism of TGF-beta1-induced AGT expression was studied in the IMR90 human lung fibroblast cell line. The increase in AGT mRNA induced by TGF-beta1 was completely blocked by actinomycin-D. TGF-beta1 increased the activity of a full-length human AGT promoter-luciferase reporter (AGT-LUC) but did not alter AGT mRNA half-life. Serial deletion analyses revealed that 67% of TGF-beta-inducible AGT-LUC activity resides in a small domain of the AGT core promoter; this domain contains binding sites for hypoxia-inducible factor (HIF)-1 and activation protein-1 (AP-1) transcription factors. TGF-beta1 increased HIF-1alpha protein abundance and the activity of a hypoxia-responsive element reporter; overexpression of HIF-1 increased basal AGT-LUC activity. Both oligonucleotide pulldown and chromatin immunoprecipitation assays revealed increased binding of JunD and HIF-1alpha to the AGT core promoter in response to TGF-beta1. TGF-beta1-inducible AGT-LUC was reduced by an AP-1 dominant negative or by mutation of the AP-1 site. Knockdown of either JunD or HIF-1alpha individually by siRNA partially reduced AGT-LUC. In contrast, simultaneous knockdown of both JunD and HIF-1alpha completely eliminated TGF-beta1-inducible AGT-LUC activity. These data suggest that TGF-beta1 up-regulates AGT transcription in human lung fibroblasts through a mechanism that requires both JunD and HIF-1alpha binding to the AGT core promoter. They also suggest a molecular mechanism linking hypoxia signaling and fibrogenic stimuli in the lungs.


Assuntos
Angiotensinogênio/metabolismo , Fibroblastos/fisiologia , Pulmão/citologia , Ativação Transcricional , Fator de Crescimento Transformador beta1/metabolismo , Angiotensinogênio/genética , Animais , Sequência de Bases , Linhagem Celular , Dactinomicina/metabolismo , Fibroblastos/citologia , Técnicas de Silenciamento de Genes , Genes Reporter , Humanos , Fator 1 Induzível por Hipóxia/genética , Fator 1 Induzível por Hipóxia/metabolismo , Dados de Sequência Molecular , Regiões Promotoras Genéticas , Inibidores da Síntese de Proteínas/metabolismo , Proteínas Proto-Oncogênicas c-jun , Estabilidade de RNA , Suínos , Fator de Transcrição AP-1/genética , Fator de Transcrição AP-1/metabolismo , Fator de Crescimento Transformador beta1/genética
10.
J Lung Pulm Respir Res ; 7(4): 85-91, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-34414259

RESUMO

BACKGROUND: Hyperoxia in pre-term neonates is a known risk factor of bronchopulmonary dysplasia (BPD). Hyperoxia is known to cause oxidative stress, inflammatory changes that leads to surfactant deactivation, and decreased surfactant expression. The previous research has shown short term exposure to hyperoxia increases surfactant protein expression but decreased expression in long term exposure. Local tissue renin-angiotensin system (RAS) is associated with tissue injury and repair and it may play a role in BPD. Endogenous peptide angiotensin 1-7 acts on the MAS receptor. The activation of the MAS receptor was previously shown to have protective pulmonary responses. However, the effect of MAS receptor activation on surfactant proteins in hyperoxic conditions has not been tested. OBJECTIVE: To determine the effects of hyperoxia with or without MAS receptor activation on Surfactant proteins. METHODS: Human epithelial cell line A549 and human primary alveolar epithelial cells (AECs) were cultured to sub-confluence (60-75%) and treated with hyperoxia (95% oxygen) and normoxia (21% oxygen) for 72 hours with or without the MAS receptor agonist (AVE0991) in serum-free F-12 nutrient media. Cells were lysed and cell lysates were collected for western blot. The statistical analysis was done using Student-Newman-Keuls Multiple comparison test. RESULTS: Surfactant protein concentration increased in AVE treated group under the hyperoxic condition when compared to the control group in both A549 cells and human primary AECs. Surfactant protein was in higher concentration in AVE0991 treated cells in both hyperoxic and normoxic conditions when compared to the non-treated control group. CONCLUSIONS: MAS receptor activation via AVE0991 causes an increase in Surfactant protein concentration in both hyperoxic and normoxic conditions. As per our experiments, hyperoxic conditions decrease the production of surfactant protein when compared to normoxic conditions. These results may reveal a novel potential drug for BPD treatment and decrease its severity.

11.
J Lung Pulm Respir Res ; 7(1): 4-13, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-34337150

RESUMO

Premature birth results in an increased risk of respiratory distress and often requires oxygen therapy. While the supplemental oxygen has been implicated as a cause of bronchopulmonary dysplasia (BPD), in clinical practice this supplementation usually only occurs after the patient's oxygen saturation levels have dropped. The effect of hyperoxia on neonates has been extensively studied. However, there is an unanswered fundamental question: which has the most impact-hyperoxia, hypoxia or fluctuating oxygen levels? In this review, we will summarize the reported effect of hypoxia, hyperoxia or a fluctuation of oxygen levels (hypoxia/hyperoxia cycling) in preterm neonates, with special emphasis on the lungs.

12.
J Lung Pulm Respir Res ; 6(3): 58-62, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-32632378

RESUMO

BACKGROUND: Neonatal therapy with a high concentration of oxygen (hyperoxia) is a known cause of bronchopulmonary dysplasia (BPD). BPD is characterized by increased pulmonary permeability and diffuse infiltration of various inflammatory cells. Disruption of the epithelial barrier may lead to altered pulmonary permeability and airways fluid accumulation. Mas receptor is a component of the renin angiotensin system and is the receptor for the protective endogenous peptide angiotensin 1-7. The activation of the Mas receptor was previously shown to have protective pulmonary responses. However, the effect of Mas receptor activation on epithelial barrier integrity has not been tested. OBJECTIVE: To determine the effects of hyperoxia with or without Mas receptor activation on epithelial cell barrier integrity. DESIGN/METHODS: Human epithelial cell line A549 was cultured on transwell polycarbonate porous membrane to confluence and treated with 95% oxygen (hyperoxia) for 72 hours with or without the Mas receptor agonist (AVE0991), or the apoptotic inhibitors Z-VAD-FMK or aurintricarboxylic acid. The cells were then challenged with Rhodamine labeled bovine serum albumin (Rh-BSA) on one side of the membrane. Fluorescent quantitation of Rh-BSA (albumin flux) was performed on the media in the other side of the membrane 3 hours later and was compared with 21% oxygen (Normoxia) control group. A549 cells were also cultured with or without AVE0991 in hyperoxia or normoxia and used for nuclear fragmentation apoptosis assay using propidium iodide staining. RESULTS: Hyperoxia induced an increase in albumin flux that was significantly prevented by AVE0991 treatment and by the apoptosis inhibitors. AVE0991 also significantly decreased the hyperoxia-induced nuclear fragmentation. CONCLUSION: These results suggest that hyperoxia causes a disruption in the epithelial barrier integrity, and that this disruption is inhibited by the Mas receptor agonist AVE0991 through inhibition of epithelial apoptosis. These results reveal a novel potential drug for BPD and pulmonary edema treatment.

13.
Artigo em Inglês | MEDLINE | ID: mdl-30175235

RESUMO

Liver and lung fibrosis are two main organ diseases that are of particular importance in both Egypt and the US. Hepatitis C Virus "HCV" infection and idiopathic pulmonary fibrosis (IPF) are fibrotic diseases of the liver and lung respectively. The liver and lung are reported in literature to share many immune/inflammatory responses to damage through the lung-liver axis. Most importantly, HCV was shown to enhance the development of IPF and is considered one of the risk factors for IPF. The renin angiotensin system (RAS) plays a critical role in the fibrogenesis and inflammation damage of many organs including liver and lung. The relatively recently identified component of RAS, angiotensin converting enzyme-2 (ACE-2), has shown a promising therapeutic potential in models of liver and pulmonary fibrosis. This article reviews the role of RAS in organ fibrosis with focus on role of ACE-2 in fibrotic diseases of the liver and the lung.

14.
Basic Clin Pharmacol Toxicol ; 100(1): 59-66, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17214612

RESUMO

Previous work from this laboratory has shown that amiodarone induces alveolar epithelial cell apoptosis that was abrogated by antagonists of angiotensin II. In this study, amiodarone up-regulated angiotensinogen mRNA and protein in primary cultures of rat type II pneumocytes and in the human A549 cell line. The mechanism of amiodarone-induced angiotensinogen expression was studied in A549 cells with a human angiotensinogen promoter-luciferase reporter (angiotensinogen/luciferase). Amiodarone (3 microg/ml) induced both angiotensinogen/luciferase and endogenous angiotensinogen mRNA; the latter was completely blocked by actinomycin-D. Amiodarone did not affect the half-life of endogenous angiotensinogen mRNA. Deletion analyses of angiotensinogen/luciferase identified at least two amiodarone-responsive domains in the angiotensinogen promoter between -350 to -260 bp and -203 to -46 bp. DNA/Protein array and electrophoretic mobility shift assays showed that amiodarone increases DNA binding of both activation protein-1 and STAT-5 transcription factors. Site-directed mutagenesis of three IL-6-responsive signal transducer activator of transcription (STAT) binding sites within the amiodarone-response domains had no effect on amiodarone-induced angiotensinogen/luciferase expression. In contrast, amiodarone-induced angiotensinogen/luciferase expression was abrogated by a dominant-negative fos construct and was stimulated over five times by c-fos and c-jun expressed together but not separately. Mutagenesis of the activation protein-1 binding site at -15 to -12 bp completely eliminated the response to amiodarone. These data show that activation protein-1 family transcription factors mediate amiodarone-induced angiotensinogen expression in human alveolar epithelial cells and identify an activation protein-1 site, located between the TATA (DNA sequence) box and the transcription initiation site, that is required for the response.


Assuntos
Amiodarona/farmacologia , Angiotensinas/genética , Antiarrítmicos/farmacologia , Expressão Gênica/efeitos dos fármacos , Alvéolos Pulmonares/efeitos dos fármacos , Fator de Transcrição AP-1/metabolismo , Adenocarcinoma , Angiotensinas/metabolismo , Sequência de Bases , Sítios de Ligação , Linhagem Celular Tumoral , Relação Dose-Resposta a Droga , Epitélio/efeitos dos fármacos , Epitélio/metabolismo , Humanos , Neoplasias Pulmonares , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Análise de Sequência com Séries de Oligonucleotídeos , Análise Serial de Proteínas , Alvéolos Pulmonares/metabolismo , RNA Mensageiro/metabolismo , Fator de Transcrição STAT5/metabolismo , Fator de Transcrição AP-1/genética
15.
Int J Pept ; 2012: 875910, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22500179

RESUMO

An established body of literature supports the hypothesis that activation of a local tissue angiotensin (ANG) system in the extravascular tissue compartment of the lungs is required for lung fibrogenesis. Transcriptional activation of the angiotensinogen (AGT) gene is believed to be a critical and necessary step in this activation. This paper summarizes the data in support of this theory and discusses transcriptional regulation of AGT, with an emphasis on lung AGT synthesis as a determinant of fibrosis severity. Genetic data linking AGT polymorphisms to the severity of disease in Idiopathic Pulmonary Fibrosis are also discussed.

16.
Am J Physiol Lung Cell Mol Physiol ; 295(1): L178-85, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18441099

RESUMO

Earlier work from this laboratory showed that local generation of angiotensin (ANG) II is required for the pathogenesis of experimental pulmonary fibrosis and that ANG peptides are expressed robustly in the lungs of patients with idiopathic pulmonary fibrosis (IPF). Angiotensin converting enzyme-2 (ACE-2) degrades the octapeptide ANG II to form the heptapeptide ANG1-7 and thereby limits ANG II accumulation. On this basis, we hypothesized that ACE-2 would be protective against experimental lung fibrogenesis and might be downregulated in human and experimental lung fibrosis. In lung biopsy specimens from patients with IPF, ACE-2 mRNA and enzyme activity were decreased by 92% (P<0.01) and 74% (P<0.05), respectively. ACE-2 mRNA and activity were also decreased similarly in the lungs of bleomycin-treated rats and C57-BL6 mice. In mice exposed to low doses of bleomycin, lung collagen accumulation was enhanced by intratracheal administration of either ACE-2-specific small interfering RNAs (siRNAs) or the peptide DX(600), a competitive inhibitor of ACE-2 (P<0.05). Administration of either ACE-2 siRNA or DX(600) significantly increased the ANG II content of mouse lung tissue above the level induced by bleomycin alone. Coadministration of the ANG II receptor antagonist saralasin blocked the DX(600)-induced increase in lung collagen. Moreover, purified recombinant human ACE-2, delivered to mice systemically by osmotic minipump, attenuated bleomycin-induced lung collagen accumulation. Together, these data show that ACE-2 mRNA and activity are severely downregulated in both human and experimental lung fibrosis and suggest that ACE-2 protects against lung fibrogenesis by limiting the local accumulation of the profibrotic peptide ANG II.


Assuntos
Angiotensina II/metabolismo , Regulação Enzimológica da Expressão Gênica , Pulmão/enzimologia , Peptidil Dipeptidase A/biossíntese , Fibrose Pulmonar/enzimologia , Adulto , Idoso , Enzima de Conversão de Angiotensina 2 , Animais , Antibióticos Antineoplásicos/toxicidade , Bleomicina/toxicidade , Regulação para Baixo/efeitos dos fármacos , Feminino , Colágenos Fibrilares/metabolismo , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Humanos , Masculino , Camundongos , Pessoa de Meia-Idade , Peptidil Dipeptidase A/farmacologia , Fibrose Pulmonar/induzido quimicamente , Fibrose Pulmonar/prevenção & controle , RNA Mensageiro/biossíntese , RNA Interferente Pequeno/farmacologia , Ratos
17.
Am J Physiol Lung Cell Mol Physiol ; 291(5): L887-95, 2006 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16844946

RESUMO

Previous work from this laboratory demonstrated de novo synthesis of angiotensin (ANG) peptides by apoptotic pulmonary alveolar epithelial cells (AEC) and by lung myofibroblasts in vitro and in bleomycin-treated rats. To determine whether these same cell types also synthesize ANG peptides de novo within the fibrotic human lung in situ, we subjected paraffin sections of normal and fibrotic (idiopathic pulmonary fibrosis, IPF) human lung to immunohistochemistry (IHC) and in situ hybridization to detect ANG peptides and angiotensinogen (AGT) mRNA. These were analyzed both alone and in combination with cell-specific markers of AEC [monoclonal antibody (MAb) MNF-116] and myofibroblasts [alpha-smooth muscle actin (alpha-SMA) MAb] and an in situ DNA end labeling (ISEL) method to detect apoptosis. In normal human lung, IHC detected AGT protein in smooth muscle underlying normal bronchi and vessels, but not elsewhere. Real-time RT-PCR and Western blotting revealed that AGT mRNA and protein were 21-fold and 3.6-fold more abundant, respectively, in IPF lung biopsies relative to biopsies of normal human lung (both P < 0.05). In IPF lung, both AGT protein and mRNA were detected in AEC that double-labeled with MAb MNF-116 and with ISEL, suggesting AGT expression by apoptotic epithelia in situ. AGT protein and mRNA also colocalized to myofibroblast foci detected by alpha-SMA MAb, but AGT mRNA was not detected in smooth muscle. These data are consistent with earlier data from isolated human lung cells in vitro and bleomycin-induced rat lung fibrosis models, and they suggest that apoptotic AEC and myofibroblasts constitute key sources of locally derived ANG peptides in the IPF lung.


Assuntos
Angiotensinogênio/genética , Angiotensinas/metabolismo , Pulmão/patologia , Fibrose Pulmonar/patologia , Fibrose Pulmonar/fisiopatologia , Angiotensinogênio/metabolismo , Antibióticos Antineoplásicos , Apoptose/fisiologia , Bleomicina , Linhagem Celular , Água Extravascular Pulmonar/metabolismo , Fibroblastos/metabolismo , Fibroblastos/patologia , Humanos , Hibridização In Situ , Pulmão/metabolismo , Alvéolos Pulmonares/metabolismo , Alvéolos Pulmonares/patologia , Fibrose Pulmonar/induzido quimicamente , Fibrose Pulmonar/metabolismo , RNA Mensageiro/metabolismo , Mucosa Respiratória/metabolismo , Mucosa Respiratória/patologia
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa