Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 537
Filtrar
1.
Radiat Res ; 196(3): 284-296, 2021 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-34153091

RESUMO

Thrombocytopenia is a major complication in hematopoietic-acute radiation syndrome (H-ARS) that increases the risk of mortality from uncontrolled hemorrhage. There is a great demand for new therapies to improve survival and mitigate bleeding in H-ARS. Thrombopoiesis requires interactions between megakaryocytes (MKs) and endothelial cells. 16, 16-dimethyl prostaglandin E2 (dmPGE2), a longer-acting analogue of PGE2, promotes hematopoietic recovery after total-body irradiation (TBI), and various angiotensin-converting enzyme (ACE) inhibitors mitigate endothelial injury after radiation exposure. Here, we tested a combination therapy of dmPGE2 and lisinopril to mitigate thrombocytopenia in murine models of H-ARS following TBI. After 7.75 Gy TBI, dmPGE2 and lisinopril each increased survival relative to vehicle controls. Importantly, combined dmPGE2 and lisinopril therapy enhanced survival greater than either individual agent. Studies performed after 4 Gy TBI revealed reduced numbers of marrow MKs and circulating platelets. In addition, sublethal TBI induced abnormalities both in MK maturation and in in vitro and in vivo platelet function. dmPGE2, alone and in combination with lisinopril, improved recovery of marrow MKs and peripheral platelets. Finally, sublethal TBI transiently reduced the number of marrow Lin-CD45-CD31+Sca-1- sinusoidal endothelial cells, while combined dmPGE2 and lisinopril treatment, but not single-agent treatment, accelerated their recovery. Taken together, these data support the concept that combined dmPGE2 and lisinopril therapy improves thrombocytopenia and survival by promoting recovery of the MK lineage, as well as the MK niche, in the setting of H-ARS.


Assuntos
16,16-Dimetilprostaglandina E2/uso terapêutico , Síndrome Aguda da Radiação/tratamento farmacológico , Inibidores da Enzima Conversora de Angiotensina/uso terapêutico , Plaquetas/efeitos dos fármacos , Células Endoteliais/efeitos dos fármacos , Transtornos Hemorrágicos/tratamento farmacológico , Lisinopril/uso terapêutico , Megacariócitos/efeitos dos fármacos , Trombocitopenia/tratamento farmacológico , Trombopoese/efeitos dos fármacos , Síndrome Aguda da Radiação/complicações , Animais , Plaquetas/efeitos da radiação , Medula Óssea/efeitos dos fármacos , Medula Óssea/efeitos da radiação , Proteína C-Reativa/análise , Radioisótopos de Césio , Avaliação Pré-Clínica de Medicamentos , Células Endoteliais/efeitos da radiação , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/efeitos da radiação , Feminino , Raios gama/efeitos adversos , Transtornos Hemorrágicos/etiologia , Megacariócitos/efeitos da radiação , Camundongos , Camundongos Endogâmicos C57BL , Selectina-P/análise , Agregação Plaquetária/efeitos dos fármacos , Agregação Plaquetária/efeitos da radiação , Fator Plaquetário 4/análise , Lesões Experimentais por Radiação/tratamento farmacológico , Lesões Experimentais por Radiação/etiologia , Trombocitopenia/etiologia , Trombopoese/efeitos da radiação , Irradiação Corporal Total , Fator de von Willebrand/análise
2.
Am J Physiol Gastrointest Liver Physiol ; 318(4): G613-G623, 2020 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-32068440

RESUMO

Esophageal injury from acid exposure related to gastroesophageal reflux disease is a common problem and a risk factor for development of Barrett's esophagus and esophageal adenocarcinoma. Our previous work highlights the benefits of using porcine esophagus to study human esophageal disease because of the similarities between porcine and human esophagus. In particular, esophageal submucosal glands (ESMGs) are present in human esophagus and proximal porcine esophagus but not in rodent esophagus. Although CFTR is expressed in the ducts of ESMGs, very little is known about CFTR and alternate anion channels, including ClC-2, in the setting of acid-related esophageal injury. After finding evidence of CFTR and ClC-2 in the basal layers of the squamous epithelium, and in the ducts of the ESMGs, we developed an ex vivo porcine model of esophageal acid injury. In this model, esophageal tissue was placed in Ussing chambers to determine the effect of pretreatment with the ClC-2 agonist lubiprostone on tissue damage related to acid exposure. Pretreatment with lubiprostone significantly reduced the level of acid injury and significantly augmented the recovery of the injured tissue (P < 0.05). Evaluation of the interepithelial tight junctions showed well-defined membrane localization of occludin in lubiprostone-treated injured tissues. Pretreatment of tissues with the Na+-K+-2Cl- cotransporter inhibitor bumetanide blocked lubiprostone-induced increases in short-circuit current and inhibited the reparative effect of lubiprostone. Furthermore, inhibition of ClC-2 with ZnCl2 blocked the effects of lubiprostone. We conclude that ClC-2 contributes to esophageal protection from acid exposure, potentially offering a new therapeutic target.NEW & NOTEWORTHY This research is the first to describe the presence of anion channels ClC-2 and CFTR localized to the basal epithelia of porcine esophageal mucosa and the esophageal submucosal glands. In the setting of ex vivo acid exposure, the ClC-2 agonist lubiprostone reduced acid-related injury and enhanced recovery of the epithelial barrier. This work may ultimately provide an alternate mechanism for treating gastroesophageal reflux disease.


Assuntos
Mucosa Esofágica/efeitos dos fármacos , Lubiprostona/farmacologia , 16,16-Dimetilprostaglandina E2/farmacologia , Animais , Bumetanida/farmacologia , Agonistas dos Canais de Cloreto/farmacologia , Canais de Cloreto/genética , Canais de Cloreto/metabolismo , Cloretos/farmacologia , Regulador de Condutância Transmembrana em Fibrose Cística/genética , Regulador de Condutância Transmembrana em Fibrose Cística/metabolismo , Relação Dose-Resposta a Droga , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Ácido Clorídrico/farmacologia , Masculino , Ocludina/metabolismo , Suínos , Fatores de Tempo , Compostos de Zinco/farmacologia
3.
Am J Physiol Gastrointest Liver Physiol ; 317(6): G862-G871, 2019 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-31545918

RESUMO

Nonsteroidal anti-inflammatory drugs (NSAIDs) such as diclofenac (DFN) and indomethacin (INDO) are extensively used worldwide. Their main side effects are injury of the gastrointestinal tract, including erosions, ulcers, and bleeding. Since gastric epithelial cells (GEPCs) are crucial for mucosal defense and are the major target of injury, we examined the extent to which DFN- and INDO-induced GEPC injury can be reversed by nerve growth factor (NGF), 16,16 dimethyl prostaglandin E2 (dmPGE2), and 5-aminoimidazole-4-carboxamide ribonucleotide (AICAR), the pharmacological activator of the metabolic sensor AMP kinase (AMPK). Cultured normal rat gastric mucosal epithelial (RGM1) cells were treated with PBS (control), NGF, dmPGE2, AICAR, and/or NSAID (DFN or INDO) for 1-4 h. We examined cell injury by confocal microscopy, cell death/survival using calcein AM, mitochondrial membrane potential using MitoTracker, and phosphorylation of AMPK by Western blotting. DFN and INDO treatment of RGM1 cells for 2 h decreased mitochondrial membrane potential and cell viability. NGF posttreatment (initiated 1 or 2 h after DFN or INDO) reversed the dissipation of mitochondrial membrane potential and cell injury caused by DFN and INDO and increased cell viability versus cells treated for 4 h with NSAID alone. Pretreatment with dmPGE2 and AICAR significantly protected these cells from DFN- and INDO-induced injury, whereas dmPGE2 and AICAR posttreatment (initiated 1 h after NSAID treatment) reversed cell injury and significantly increased cell viability and rescued the cells from NSAID-induced mitochondrial membrane potential reduction. DFN and INDO induce extensive mitochondrial injury and GEPC death, which can be significantly reversed by NGF, dmPGE2, and AICAR.NEW & NOTEWORTHY This study demonstrated that mitochondria are key targets of diclofenac- and indomethacin-induced injury of gastric epithelial cells and that diclofenac and indomethacin injury can be prevented and, importantly, also reversed by treatment with nerve growth factor, 16,16 dimethyl prostaglandin E2, and 5-aminoimidazole-4-carboxamide ribonucleotide.


Assuntos
16,16-Dimetilprostaglandina E2/farmacologia , Aminoimidazol Carboxamida/análogos & derivados , Diclofenaco/efeitos adversos , Mucosa Gástrica , Indometacina/efeitos adversos , Mitocôndrias , Fator de Crescimento Neural/farmacologia , Ribonucleosídeos/farmacologia , Aminoimidazol Carboxamida/farmacologia , Animais , Anti-Inflamatórios não Esteroides/efeitos adversos , Anti-Inflamatórios não Esteroides/farmacologia , Antiulcerosos/farmacologia , Morte Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Mucosa Gástrica/efeitos dos fármacos , Mucosa Gástrica/metabolismo , Mucosa Gástrica/patologia , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Ratos
4.
Pulm Pharmacol Ther ; 48: 62-70, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-28951192

RESUMO

A feature of cough variant asthma is a heightened cough response to bronchoconstriction. The mediators of this response are unknown. This study was designed to elucidate the role of lipid mediators in bronchoconstriction-triggered cough response in an experimental animal model. We examined the influence of bronchoconstriction on cell components and mediators including prostaglandin E2 (PGE2) in bronchoalveolar lavage fluid (BALF). We studied the cough response to bronchoconstriction (CRB) by measuring the correlation between the increase in enhanced pause (Penh), an index of bronchoconstriction, and cough counts induced by methacholine (Mch) inhalation in conscious guinea pigs. We then examined the effects of intraperitoneal pretreatment with 16, 16-dimethyl-prostaglandin E2 (dm-PGE2) on CRB and cough counts. The total number of cells and cell components in the BALF were not influenced by bronchoconstriction. While levels of PGE2, prostaglandin I2, and cysteinyl leukotrienes were significantly increased, levels of prostaglandin D2, thromboxane B2, and substance P in the BALF were not. Dm-PGE2 significantly decreased the Mch-induced increase in Penh. Following bronchoconstriction by additional Mch inhalation, dm-PGE2 produced an increase in CRB and cough counts in a dose-dependent manner. Additionally, the heightened CRB following dm-PGE2 treatment was suppressed by pretreatment with PGE2 receptor (E-prostanoid EP) -1 and EP-3 antagonists in a dose-dependent manner, but not by EP-2 and EP-4 antagonists. The EP-1 antagonist also decreased cough counts. These results suggest that PGE2 acts as an exacerbating factor for bronchoconstriction-triggered cough. EP1 and EP3 may provide new therapeutic targets for cough variant asthma.


Assuntos
16,16-Dimetilprostaglandina E2/farmacologia , Broncoconstrição , Tosse/fisiopatologia , Dinoprostona/metabolismo , 16,16-Dimetilprostaglandina E2/administração & dosagem , Animais , Líquido da Lavagem Broncoalveolar , Cisteína/metabolismo , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Epoprostenol/metabolismo , Cobaias , Leucotrienos/metabolismo , Masculino , Cloreto de Metacolina/administração & dosagem , Receptores de Prostaglandina E/efeitos dos fármacos , Receptores de Prostaglandina E/metabolismo
5.
Exp Hematol ; 56: 64-68, 2017 12.
Artigo em Inglês | MEDLINE | ID: mdl-28822771

RESUMO

Monocytes/macrophages differentiating from bone marrow (BM) cells pulsed for 2 hours at 37°C with a stabilized derivative of prostaglandin E2, 16,16-dimethyl PGE2 (dmPGE2), migrated less efficiently toward a chemoattractant than monocytes/macrophages differentiated from BM cells pulsed with vehicle. To confirm that the effect on BM cells was long lasting and to replicate human BM transplantation, chimeric mice were established with donor BM cells pulsed for 2 hours with dmPGE2 before injection into marrow-ablated congenic recipient mice. After 12 weeks, when high levels (90%) of engraftment were obtained, regenerated BM-derived monocytes/macrophages differentiating in vitro or in vivo migrated inefficiently toward the chemokines colony-stimulating factor-1 (CSF-1) and chemokine (C-C motif) ligand 2 (CCL2) or thioglycollate, respectively. Our results reveal long-lasting changes to progenitor cells of monocytes/macrophages by a 2-hour dmPGE2 pulse that, in turn, limits the migration of their daughter cells to chemoattractants and inflammatory mediators.


Assuntos
Células da Medula Óssea/metabolismo , Movimento Celular/efeitos dos fármacos , Dinoprostona/farmacologia , Macrófagos/metabolismo , Monócitos/metabolismo , 16,16-Dimetilprostaglandina E2/farmacologia , Animais , Células da Medula Óssea/citologia , Quimiocina CCL2/metabolismo , Fator Estimulador de Colônias de Macrófagos/metabolismo , Macrófagos/citologia , Camundongos , Monócitos/citologia
6.
Proc Natl Acad Sci U S A ; 113(20): 5552-7, 2016 May 17.
Artigo em Inglês | MEDLINE | ID: mdl-27140638

RESUMO

The incidence of obesity, which is recognized by the American Medical Association as a disease, has nearly doubled since 1980, and obesity-related comorbidities have become a major threat to human health. Given that adipose tissue expansion and transformation require active growth of new blood vasculature, angiogenesis offers a potential target for the treatment of obesity-associated disorders. Here we construct two peptide-functionalized nanoparticle (NP) platforms to deliver either Peroxisome Proliferator-Activated Receptor gamma (PPARgamma) activator rosiglitazone (Rosi) or prostaglandin E2 analog (16,16-dimethyl PGE2) to adipose tissue vasculature. These NPs were engineered through self-assembly of a biodegradable triblock polymer composed of end-to-end linkages between poly(lactic-coglycolic acid)-b-poly(ethylene glycol) (PLGA-b-PEG) and an endothelial-targeted peptide. In this system, released Rosi promotes both transformation of white adipose tissue (WAT) into brown-like adipose tissue and angiogenesis, which facilitates the homing of targeted NPs to adipose angiogenic vessels, thereby amplifying their delivery. We show that i.v. administration of these NPs can target WAT vasculature, stimulate the angiogenesis that is required for the transformation of adipose tissue, and transform WAT into brown-like adipose tissue, by the up-regulation of angiogenesis and brown adipose tissue markers. In a diet-induced obese mouse model, these angiogenesis-targeted NPs have inhibited body weight gain and modulated several serological markers including cholesterol, triglyceride, and insulin, compared with the control group. These findings suggest that angiogenesis-targeting moieties with angiogenic stimulator-loaded NPs could be incorporated into effective therapeutic regimens for clinical treatment of obesity and other metabolic diseases.


Assuntos
16,16-Dimetilprostaglandina E2/administração & dosagem , Tecido Adiposo Marrom/metabolismo , Tecido Adiposo Branco/irrigação sanguínea , Nanopartículas/administração & dosagem , Neovascularização Fisiológica , Obesidade/prevenção & controle , Tiazolidinedionas/administração & dosagem , Animais , Metabolismo dos Carboidratos , Dieta , Sistemas de Liberação de Medicamentos , Metabolismo dos Lipídeos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Nanomedicina , Rosiglitazona
7.
Int J Biochem Cell Biol ; 59: 52-61, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25462156

RESUMO

Prostaglandin E2 (PGE2) and hypoxia-inducible factor-1α (HIF-1α) affect many mechanisms that have been shown to play a role in prostate cancer. In PGE2-treated LNCaP cells, up-regulation of HIF-1α requires the internalization of PGE2, which is in sharp contrast with the generally accepted view that PGE2 acts through EP receptors located at the cell membrane. Here we aimed to study in androgen-independent PC3 cells the role of intracellular PGE2 in several events linked to prostate cancer progression. To this end, we used bromocresol green, an inhibitor of prostaglandin uptake that blocked the immediate rise in intracellular immunoreactive PGE2 following treatment with 16,16-dimethyl-PGE2. Bromocresol green prevented the stimulatory effect of 16,16-dimethyl-PGE on cell proliferation, adhesion, migration and invasion and on HIF-1α expression and activity, the latter assessed as the HIF-dependent activation of (i) a hypoxia response element-luciferase plasmid construct, (ii) production of angiogenic factor vascular endothelial growth factor-A and (iii) in vitro angiogenesis. The basal phenotype of PC3 cells was also affected by bromocresol green, that substantially lowered expression of HIF-1α, production of vascular endothelial growth factor-A and cell proliferation. These results, and the fact that we found functional intracellular EP receptors in PC3 cells, suggest that PGE2-dependent intracrine mechanisms play a role in prostate cancer Therefore, inhibition of the prostaglandin uptake transporter might be a novel therapeutic approach for the treatment of prostate cancer.


Assuntos
Dinoprostona/metabolismo , Espaço Intracelular/metabolismo , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/patologia , 16,16-Dimetilprostaglandina E2/farmacologia , Transporte Biológico/efeitos dos fármacos , Verde de Bromocresol/farmacologia , Adesão Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Colágeno/farmacologia , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Masculino , Invasividade Neoplásica , Fenótipo , Receptores de Prostaglandina E/metabolismo
8.
Exp Neurol ; 261: 354-66, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24910202

RESUMO

Prostaglandin E2 (PGE2), a well-known pain mediator enriched in inflamed tissues, plays a pivotal role in the genesis of chronic pain conditions such as inflammatory and neuropathic pain. PGE2-prolonged sensitization of nociceptive dorsal root ganglion (DRG) neurons (nociceptors) may contribute to the transition from acute to chronic pain. However, the underlying cellular mechanisms are poorly understood. In this study, we tested the hypothesis that facilitating synthesis and anterograde axonal trafficking of EP receptors contribute to PGE2-prolonged nociceptor sensitization. Intraplantar (i.pl.) injection of a stabilized PGE2 analog, 16,16 dimethyl PGE2 (dmPGE2), in a dose- and time-dependent manner, not only elicited primary tactile allodynia which lasted for 1d, but also prolonged tactile allodynia evoked by a subsequent i.pl. injection of dmPGE2 from 1d to 4d. Moreover, the duration of tactile allodynia was progressively prolonged following multiple sequential i.pl. injections of dmPGE2. Co-injection of the selective EP1 or EP4 receptor antagonist, the inhibitors of cAMP, PKA, PKC, PKCε or PLC as well as an interleukin-6 (IL-6) neutralizing antiserum differentially blocked primary tactile allodynia elicited by the 1st dmPGE2 and the prolonged tactile allodynia evoked by the 2nd dmPGE2, suggesting the involvement of these signaling events in dmPGE2-induced nociceptor activation and sensitization. Co-injection of a selective COX2 inhibitor or two EP4 antagonists prevented or shortened inflammagen-prolonged nociceptor sensitization. I.pl. injection of dmPGE2 or carrageenan time-dependently increased EP4 levels in L4-6 DRG neurons and peripheral nerves. EP4 was expressed in almost half of IB4-binding nociceptors of L4-6 DRG. Taken together, our data suggest that stimulating the synthesis and anterograde axonal trafficking to increase EP4 availability at the axonal terminals of nociceptors is likely a novel mechanism underlying PGE2-prolonged nociceptor sensitization. Blocking COX2/PGE2/EP4 signaling at an earlier stage of inflammation or injury is crucial for preventing the transition from acute pain to a chronic state.


Assuntos
Transporte Axonal/efeitos dos fármacos , Gânglios Espinais/citologia , Neuralgia/patologia , Receptores de Prostaglandina E Subtipo EP4/metabolismo , Células Receptoras Sensoriais/efeitos dos fármacos , 16,16-Dimetilprostaglandina E2/toxicidade , Animais , Células Cultivadas , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/farmacologia , Glicoproteínas/metabolismo , Hiperalgesia/induzido quimicamente , Hiperalgesia/metabolismo , Lectinas/metabolismo , Masculino , Neuralgia/induzido quimicamente , Neuralgia/metabolismo , Medição da Dor , Limiar da Dor/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Receptores de Prostaglandina E Subtipo EP1/metabolismo , Fatores de Tempo , Versicanas
9.
Cell Stem Cell ; 14(2): 146-8, 2014 Feb 06.
Artigo em Inglês | MEDLINE | ID: mdl-24506882

RESUMO

Translating basic research findings into therapeutic settings presents many scientific, logistic, and financial challenges for academic researchers. Here, I highlight some key insights for navigating such challenges based on recent clinical trials initiated by basic research from my lab.


Assuntos
Ensaios Clínicos como Assunto , Pesquisa Translacional Biomédica , 16,16-Dimetilprostaglandina E2/farmacologia , Animais , Humanos , Camundongos , Transplante de Células-Tronco , Estados Unidos , United States Food and Drug Administration , Peixe-Zebra
10.
Acta Biochim Biophys Sin (Shanghai) ; 46(1): 40-7, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24296051

RESUMO

The effect of prostaglandin E2 (PGE2) on bone mass has been well-established in vivo. Previous studies have showed that PGE2 increases differentiation, proliferation, and regulates cell morphology through F-actin stress fiber in statically cultured osteoblasts. However, the effect of PGE2 on osteoblasts in the presence of fluid shear stress (FSS), which could better uncover the anabolic effect of PGE2 in vivo, has yet to be examined. Here, we hypothesized that PGE2 modulates F-actin stress fiber in FSS-stimulated MC3T3-E1 osteoblastic cells through protein kinase A (PKA) pathway. Furthermore, this PGE2-induced F-actin remodeling was associated with the recovery of cellular mechanosensitivity. Our data showed that treatment with 10 nM dmPGE2 for 15 min significantly suppressed the F-actin stress fiber intensity in FSS-stimulated cells in a PKA-dependent manner. In addition, dmPGE2 treatment enhanced the cells' calcium peak magnitude and the percentage of responding cells in the second FSS stimulation, though these effects were abolished and attenuated by co-treatment with phalloidin. Our results demonstrated that 10 nM dmPGE2 was able to accelerate the 'reset' process of F-actin stress fiber to its pre-stimulated level partially through PKA pathway, and thus promoted the recovery of cellular mechanosensitivity. Our finding provided a novel cellular mechanism by which PGE2 increased bone formation as shown in vivo, suggesting that PGE2 could be a potential target for treatments of bone formation-related diseases.


Assuntos
16,16-Dimetilprostaglandina E2/farmacologia , Actinas/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/fisiologia , Osteoblastos/metabolismo , Fibras de Estresse/metabolismo , Células Cultivadas , Modelos Biológicos , Osteoblastos/efeitos dos fármacos , Osteogênese/efeitos dos fármacos , Estresse Mecânico
11.
Blood ; 122(17): 3074-81, 2013 Oct 24.
Artigo em Inglês | MEDLINE | ID: mdl-23996087

RESUMO

Umbilical cord blood (UCB) is a valuable source of hematopoietic stem cells (HSCs) for use in allogeneic transplantation. Key advantages of UCB are rapid availability and less stringent requirements for HLA matching. However, UCB contains an inherently limited HSC count, which is associated with delayed time to engraftment, high graft failure rates, and early mortality. 16,16-Dimethyl prostaglandin E2 (dmPGE2) was previously identified to be a critical regulator of HSC homeostasis, and we hypothesized that brief ex vivo modulation with dmPGE2 could improve patient outcomes by increasing the "effective dose" of HSCs. Molecular profiling approaches were used to determine the optimal ex vivo modulation conditions (temperature, time, concentration, and media) for use in the clinical setting. A phase 1 trial was performed to evaluate the safety and therapeutic potential of ex vivo modulation of a single UCB unit using dmPGE2 before reduced-intensity, double UCB transplantation. Results from this study demonstrated clear safety with durable, multilineage engraftment of dmPGE2-treated UCB units. We observed encouraging trends in efficacy, with accelerated neutrophil recovery (17.5 vs 21 days, P = .045), coupled with preferential, long-term engraftment of the dmPGE2-treated UCB unit in 10 of 12 treated participants.


Assuntos
16,16-Dimetilprostaglandina E2/farmacologia , Plaquetas/efeitos dos fármacos , Transplante de Células-Tronco de Sangue do Cordão Umbilical/métodos , Sangue Fetal/efeitos dos fármacos , Sobrevivência de Enxerto/imunologia , Neoplasias Hematológicas/terapia , Adulto , Idoso , Plaquetas/citologia , Plaquetas/imunologia , Células Cultivadas , Criopreservação , Feminino , Sangue Fetal/citologia , Sangue Fetal/imunologia , Sangue Fetal/transplante , Perfilação da Expressão Gênica , Neoplasias Hematológicas/imunologia , Neoplasias Hematológicas/patologia , Humanos , Masculino , Pessoa de Meia-Idade , Quimeras de Transplante , Transplante Homólogo , Resultado do Tratamento
12.
PLoS One ; 7(1): e29331, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22276108

RESUMO

BACKGROUND: Prostaglandin E(2) (PGE(2)) is an important mediator in tumor-promoting inflammation. High expression of cyclooxygenase-2 (COX-2) has been detected in the embryonic childhood tumor neuroblastoma, and treatment with COX inhibitors significantly reduces tumor growth. Here, we have investigated the significance of a high COX-2 expression in neuroblastoma by analysis of PGE(2) production, the expression pattern and localization of PGE(2) receptors and intracellular signal transduction pathways activated by PGE(2). PRINCIPAL FINDINGS: A high expression of the PGE(2) receptors, EP1, EP2, EP3 and EP4 in primary neuroblastomas, independent of biological and clinical characteristics, was detected using immunohistochemistry. In addition, mRNA and protein corresponding to each of the receptors were detected in neuroblastoma cell lines. Immunofluorescent staining revealed localization of the receptors to the cellular membrane, in the cytoplasm, and in the nuclear compartment. Neuroblastoma cells produced PGE(2) and stimulation of serum-starved neuroblastoma cells with PGE(2) increased the intracellular concentration of calcium and cyclic AMP with subsequent phosphorylation of Akt. Addition of 16,16-dimethyl PGE(2) (dmPGE(2)) increased cell viability in a time, dose- and cell line-dependent manner. Treatment of neuroblastoma cells with a COX-2 inhibitor resulted in a diminished cell growth and viability that was reversed by the addition of dmPGE(2). Similarly, PGE(2) receptor antagonists caused a decrease in neuroblastoma cell viability in a dose-dependent manner. CONCLUSIONS: These findings demonstrate that PGE(2) acts as an autocrine and/or paracrine survival factor for neuroblastoma cells. Hence, specific targeting of PGE(2) signaling provides a novel strategy for the treatment of childhood neuroblastoma through the inhibition of important mediators of tumor-promoting inflammation.


Assuntos
16,16-Dimetilprostaglandina E2/farmacologia , Comunicação Autócrina/efeitos dos fármacos , Dinoprostona/metabolismo , Neuroblastoma/metabolismo , Compostos de Bifenilo/farmacologia , Western Blotting , Cálcio/metabolismo , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , AMP Cíclico/metabolismo , Inibidores de Ciclo-Oxigenase 2/farmacologia , Humanos , Imuno-Histoquímica , Técnicas In Vitro , Fosforilação/efeitos dos fármacos , Receptores de Prostaglandina E Subtipo EP1/antagonistas & inibidores , Receptores de Prostaglandina E Subtipo EP1/metabolismo , Receptores de Prostaglandina E Subtipo EP2/antagonistas & inibidores , Receptores de Prostaglandina E Subtipo EP2/metabolismo , Receptores de Prostaglandina E Subtipo EP3/antagonistas & inibidores , Receptores de Prostaglandina E Subtipo EP3/metabolismo , Receptores de Prostaglandina E Subtipo EP4/antagonistas & inibidores , Receptores de Prostaglandina E Subtipo EP4/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Espectrometria de Massas em Tandem , Tiofenos/farmacologia , Triazóis/farmacologia
13.
Br J Pharmacol ; 162(1): 193-209, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20804494

RESUMO

BACKGROUND AND PURPOSE: Changes in airway smooth muscle (ASM) phenotype may contribute to the pathogenesis of airway disease. Platelet-derived growth factor (PDGF) switches ASM from a contractile to a proliferative, hypo-contractile phenotype, a process requiring activation of extracellular signal-regulated kinase (ERK) and p70(S6) Kinase (p70(S6K) ). The effects of cAMP-elevating agents on these processes is unknown. Here, we investigated the effects of cAMP elevation by prostaglandin E(2) (PGE(2) ) and the activation of the cAMP effectors, protein kinase A (PKA) and exchange protein activated by cAMP (Epac) on PDGF-induced phenotype switching in bovine tracheal smooth muscle (BTSM). EXPERIMENTAL APPROACH: Effects of long-term treatment with the PGE(2) analogue 16,16-dimethyl-PGE(2) , the selective Epac activator, 8-pCPT-2'-O-Me-cAMP and the selective PKA activator, 6-Bnz-cAMP were assessed on the induction of a hypo-contractile, proliferative BTSM phenotype and on activation of ERK and p70(S6K) , both induced by PDGF. KEY RESULTS: Treatment with 16,16-dimethyl-PGE(2) inhibited PDGF-induced proliferation of BTSM cells and maintained BTSM strip contractility and contractile protein expression in the presence of PDGF. Activation of both Epac and PKA similarly prevented PDGF-induced phenotype switching and PDGF-induced activation of ERK. Interestingly, only PKA activation resulted in inhibition of PDGF-induced phosphorylation of p70(S6K) . CONCLUSIONS AND IMPLICATIONS: Our data indicate for the first time that both Epac and PKA regulated switching of ASM phenotype via differential inhibition of ERK and p70(S6K) pathways. These findings suggest that cAMP elevation may be beneficial in the treatment of long-term changes in airway disease.


Assuntos
Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , AMP Cíclico/farmacologia , Músculo Liso/efeitos dos fármacos , Traqueia/efeitos dos fármacos , 16,16-Dimetilprostaglandina E2/farmacologia , Animais , Western Blotting , Bovinos , Linhagem Celular , Ativação Enzimática , Técnicas In Vitro , Músculo Liso/enzimologia , Músculo Liso/fisiologia , Fosforilação , Fator de Crescimento Derivado de Plaquetas/farmacologia , Proteínas Quinases S6 Ribossômicas/metabolismo , Traqueia/enzimologia , Traqueia/fisiologia
14.
J Neurochem ; 115(2): 363-72, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20666934

RESUMO

Prostaglandin E2 (PGE2) is a well known pain and pro-inflammatory mediator abundantly produced in inflamed tissue. It causes pain by directly exciting nociceptive primary sensory neurons (nociceptors) and indirectly stimulating the release of pain-related peptide substance P (SP) and calcitonin gene-related peptide (CGRP). In an ex vivo culture of sensory ganglion explants, we tested the hypothesis that PGE2 could induce the synthesis of SP and CGRP in nociceptors. A stabilized PGE2 analog, 16,16-dimethyl PGE2, in a concentration- and time-dependent manner, significantly increased mRNA and peptide levels of SP and CGRP. The agonists of EP1 and EP4 receptors also significantly increased SP and CGRP levels. Moreover, 16,16-dimethyl PGE2-induced SP and CGRP were blocked by EP1 and EP4 antagonists as well as the inhibitors of both protein kinase A and protein kinase C. Nerve growth factor was partially involved in PGE2-induced SP and CGRP synthesis. Taken together, these results indicate that PGE2 contributes to the synthesis of SP and CGRP in nociceptors, an event mediated by EP1 and EP4 receptors, nerve growth factor and protein kinase A and protein kinase C signalling pathways. We thus conclude that facilitating the synthesis of pain-related peptides in nociceptors is a novel mechanism underlying the role of PGE2 in nociception and chronic pain states.


Assuntos
16,16-Dimetilprostaglandina E2/farmacologia , Antiulcerosos/farmacologia , Peptídeo Relacionado com Gene de Calcitonina/metabolismo , Gânglios Espinais/efeitos dos fármacos , Substância P/metabolismo , Animais , Peptídeo Relacionado com Gene de Calcitonina/genética , Ácido Dibenzo(b,f)(1,4)oxazepina-10(11H)-carboxílico, 8-cloro-, 2-acetilidrazida/farmacologia , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/farmacologia , Ensaio de Imunoadsorção Enzimática/métodos , Regulação da Expressão Gênica/efeitos dos fármacos , Masculino , Fator de Crescimento Neural/metabolismo , Técnicas de Cultura de Órgãos , Antagonistas de Prostaglandina/farmacologia , Prostaglandinas Sintéticas/farmacologia , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Receptores de Prostaglandina E/agonistas , Receptores de Prostaglandina E/genética , Receptores de Prostaglandina E/metabolismo , Transdução de Sinais/efeitos dos fármacos , Substância P/genética , Fatores de Tempo
15.
J Gastroenterol Hepatol ; 25 Suppl 1: S111-8, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20586851

RESUMO

BACKGROUND AND AIMS: We examined the effect of lafutidine, a histamine H(2) receptor antagonist with a mucosal protective action mediated by capsaicin-sensitive sensory neurons (CSN), on intestinal lesions produced by loxoprofen administration in rats. METHODS: Animals were given loxoprofen (10-100 mg/kg p.o.) and killed 24 h later. Lafutidine (10 and 30 mg/kg), cimetidine (100 mg/kg) or famotidine (30 mg/kg) was given twice p.o. at 0.5 h before and 6 h after loxoprofen. Omeprazole (100 mg/kg) was given p.o. once 0.5 h before. Ampicillin (800 mg/kg) was given p.o. twice at 24 h and 0.5 h before loxoprofen, while 16,16-dimethyl prostaglandin E(2) (dmPGE(2); 0.01 mg/kg) was given i.v. twice at 5 min before and 6 h after. RESULTS: Loxoprofen dose-dependently produced hemorrhagic lesions in the small intestine, accompanied by invasion of enterobacteria and increased inducible nitric oxide synthase (iNOS) expression as well as myeloperoxidase activity in the mucosa. The ulcerogenic response to loxoprofen (60 mg/kg) was significantly prevented by lafutidine (30 mg/kg), similar to dmPGE(2) and ampicillin, and the effect of lafutidine was totally attenuated by ablation of CSN. Neither cimetidine, famotidine nor omeprazole had a significant effect against these lesions. Lafutidine alone increased mucus secretion and reverted the decreased mucus response to loxoprofen, resulting in suppression of bacterial invasion and iNOS expression. In addition, loxoprofen downregulated Muc2 expression, and this response was totally reversed by lafutidine mediated by CSN. CONCLUSION: Lafutidine protects the small intestine against loxoprofen-induced lesions, essentially mediated by the CSN, and this effect may be functionally associated with increased Muc2 expression/mucus secretion, an important factor in the suppression of bacterial invasion.


Assuntos
Acetamidas/farmacologia , Antiulcerosos/farmacologia , Antagonistas dos Receptores H2 da Histamina/farmacologia , Mucosa Intestinal/efeitos dos fármacos , Intestino Delgado/efeitos dos fármacos , Úlcera Péptica/prevenção & controle , Fenilpropionatos , Piperidinas/farmacologia , Piridinas/farmacologia , 16,16-Dimetilprostaglandina E2/farmacologia , Ampicilina/farmacologia , Animais , Antibacterianos/farmacologia , Capsaicina/farmacologia , Cimetidina/farmacologia , Modelos Animais de Doenças , Enterobacteriaceae/patogenicidade , Famotidina/farmacologia , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiologia , Mucosa Intestinal/patologia , Intestino Delgado/metabolismo , Intestino Delgado/microbiologia , Intestino Delgado/patologia , Masculino , Mucina-2/genética , Óxido Nítrico Sintase Tipo II/genética , Omeprazol/farmacologia , Úlcera Péptica/induzido quimicamente , Úlcera Péptica/metabolismo , Úlcera Péptica/microbiologia , Úlcera Péptica/patologia , Peroxidase/metabolismo , Inibidores da Bomba de Prótons/farmacologia , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Células Receptoras Sensoriais/efeitos dos fármacos
16.
J Gastroenterol Hepatol ; 25 Suppl 1: S15-22, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20586859

RESUMO

BACKGROUND AND AIMS: Enteroscopic observation has clearly demonstrated that non-steroidal anti-inflammatory drugs/low-dose aspirin (usually enteric-coated) induces hemorrhagic lesions, including ulcers and bleeding, in the small intestine of patients at a high incidence. Such intestinal lesions induced by NSAIDs have been confirmed in animal experiments. With aspirin, however, it has long been believed that it is difficult to induce any damage in the intestinal mucosa of laboratory animals. Therefore, we established a new method of inducing intestinal hemorrhagic lesions in rats by injecting aspirin into the proximal duodenum. METHODS: Under ether anesthesia, aspirin (50-200 mg/body), suspended in 2% methylcellulose (with or without 0.1 N HCl), was injected into the proximal duodenum of normally fed or 20-h non-fed rats (male Sprague-Dawley, 9 weeks old). At 1 h after treatment, the animals were killed with ether and the entire small intestine was removed for histological examination. In some experiments, 1% Evans blue was injected (i.v.) into the rats 1 h after aspirin treatment to visualize the lesions. An image analyzer determined the total area of the intestinal lesions. Oral proton pump inhibitors and histamine H(2)-receptor blockers were given 1 h before aspirin injection. 16,16-dimethyl prostaglandin E(2) (dmPGE(2)) was given s.c. 30 min before aspirin injection. RESULTS: Aspirin alone clearly induced severe lesions (including bleeding and ulcers) mainly in the jejunum at 100% incidence. Total score of lesions per rat obtained by histological examination was similar to the damaged area quantified with the dye method. Dose-related induction of lesions by aspirin was confirmed both by the histological and dye methods. The irritable effect of aspirin suspended in 0.1 N HCl solution was the same as that of aspirin alone; 0.1 N HCl alone induced only minor lesions in the intestine. Both proton pump inhibitors and histamine H(2)-receptor blockers, at doses that inhibit gastric acid secretion, had no or little effect on aspirin-induced intestinal lesions. Pretreatment with dmPGE(2) (3, 10, 30 microg/kg) showed significant prevention of both aspirin- and HCl/aspirin-induced intestinal lesions. CONCLUSION: This new aspirin lesion model will be useful for screening defensive drugs against aspirin-induced intestinal lesions and to elucidate the underlying mechanism.


Assuntos
Anti-Inflamatórios não Esteroides/administração & dosagem , Aspirina/administração & dosagem , Úlcera Duodenal/induzido quimicamente , Duodeno/patologia , Jejuno/patologia , Úlcera Péptica Hemorrágica/induzido quimicamente , 16,16-Dimetilprostaglandina E2/farmacologia , Animais , Antiulcerosos/farmacologia , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Úlcera Duodenal/patologia , Úlcera Duodenal/prevenção & controle , Duodeno/efeitos dos fármacos , Duodeno/ultraestrutura , Antagonistas dos Receptores H2 da Histamina/farmacologia , Injeções , Mucosa Intestinal/patologia , Jejuno/efeitos dos fármacos , Jejuno/ultraestrutura , Masculino , Microscopia Eletrônica de Varredura , Úlcera Péptica Hemorrágica/patologia , Úlcera Péptica Hemorrágica/prevenção & controle , Inibidores da Bomba de Prótons/farmacologia , Ratos , Ratos Sprague-Dawley
17.
J Gastroenterol Hepatol ; 25 Suppl 1: S67-74, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20586869

RESUMO

BACKGROUND AND AIMS: The effects of an EP4 agonist/antagonist on the healing of lesions produced by indomethacin in the small intestine were examined in rats, especially in relation to the expression of vascular endothelial growth factor (VEGF) and angiogenesis. METHODS: Animals were given indomethacin (10 mg/kg s.c.) and killed at various time points. To impair the healing of these lesions, a small dose of indomethacin (2 mg/kg p.o.) or AE3-208 (EP4 antagonist: 3 mg/kg i.p.) was given once daily for 6 days after the ulceration was induced, with or without the co-administration of AE1-329 (EP4 agonist: 0.1 mg/kg i.p.). RESULTS: Indomethacin (10 mg/kg) caused severe damage in the small intestine, but the lesions healed rapidly decreasing to approximately one-fifth of their initial size within 7 days. The healing process was significantly impaired by indomethacin (2 mg/kg) given once daily for 6 days after the ulceration. This effect of indomethacin was mimicked by the EP4 antagonist and reversed by co-administration of the EP4 agonist. Mucosal VEGF expression was upregulated after the ulceration, reaching a peak on day 3 followed by a decrease. The changes in VEGF expression paralleled those in mucosal cyclooxygenase-2 expression, as well as prostaglandin E(2) (PGE(2)) content. Indomethacin (2 mg/kg) downregulated both VEGF expression and angiogenesis in the mucosa during the healing process, and these effects were significantly reversed by co-treatment with the EP4 agonist. CONCLUSION: The results suggest that endogenous PGE(2) promotes the healing of small intestinal lesions by stimulating angiogenesis through the upregulation of VEGF expression mediated by the activation of EP4 receptors.


Assuntos
Dinoprostona/metabolismo , Indometacina , Mucosa Intestinal/efeitos dos fármacos , Intestino Delgado/metabolismo , Úlcera Péptica/metabolismo , Receptores de Prostaglandina E/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Cicatrização , 16,16-Dimetilprostaglandina E2/farmacologia , Animais , Ciclo-Oxigenase 1/genética , Ciclo-Oxigenase 1/metabolismo , Ciclo-Oxigenase 2/genética , Ciclo-Oxigenase 2/metabolismo , Modelos Animais de Doenças , Regulação Enzimológica da Expressão Gênica , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patologia , Intestino Delgado/efeitos dos fármacos , Intestino Delgado/patologia , Masculino , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Naftalenos/farmacologia , Neovascularização Fisiológica/efeitos dos fármacos , Úlcera Péptica/induzido quimicamente , Úlcera Péptica/tratamento farmacológico , Úlcera Péptica/patologia , Fenilbutiratos/farmacologia , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Receptores de Prostaglandina E/agonistas , Receptores de Prostaglandina E/antagonistas & inibidores , Receptores de Prostaglandina E Subtipo EP4 , Índice de Gravidade de Doença , Fatores de Tempo , Regulação para Cima , Cicatrização/efeitos dos fármacos
18.
Pharmacology ; 84(5): 294-9, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19816089

RESUMO

Rat gastric mucosal damage was induced by ischemia-reperfusion. The 5-lipoxygenase inhibitors MK886 and A63162, the 12-lipoxygenase inhibitor baicalein, the 15-lipoxygenase inhibitor PD146176 and the lipoxin (LX) A(4)/annexin 1 antagonist Boc1 increased mucosal damage in a dose-dependent manner. Low doses of these compounds, which have no effects on mucosal integrity, cause severe damage when combined with low doses of indomethacin, celecoxib or dexamethasone. 16,16-Dimethylprostaglandin (PG) E(2) and LXA(4) can replace each other in preventing mucosal injury induced by either cyclooxygenase or lipoxygenase inhibitors. The results suggest that not only cyclooxygenases, but also lipoxygenases have a role in limiting gastric mucosal damage during ischemia-reperfusion.


Assuntos
Anexinas/metabolismo , Mucosa Gástrica/efeitos dos fármacos , Lipoxigenase/fisiologia , Receptores de Formil Peptídeo/fisiologia , Receptores de Lipoxinas/fisiologia , Traumatismo por Reperfusão/tratamento farmacológico , 16,16-Dimetilprostaglandina E2/farmacologia , Acetamidas/farmacologia , Animais , Antiulcerosos/farmacologia , Celecoxib , Inibidores de Ciclo-Oxigenase/farmacologia , Dexametasona/farmacologia , Sinergismo Farmacológico , Flavanonas/farmacologia , Fluorenos/farmacologia , Mucosa Gástrica/irrigação sanguínea , Mucosa Gástrica/patologia , Glucocorticoides/farmacologia , Indóis/farmacologia , Indometacina/farmacologia , Inibidores de Lipoxigenase/farmacologia , Masculino , Oligopeptídeos/farmacologia , Éteres Fenílicos , Antagonistas de Prostaglandina/farmacologia , Pirazóis/farmacologia , Ratos , Ratos Wistar , Receptores de Lipoxinas/antagonistas & inibidores , Traumatismo por Reperfusão/metabolismo , Traumatismo por Reperfusão/patologia , Sulfonamidas/farmacologia
19.
Proc Natl Acad Sci U S A ; 106(31): 13094-9, 2009 Aug 04.
Artigo em Inglês | MEDLINE | ID: mdl-19622732

RESUMO

Carbonic anhydrase (CA) is strongly expressed in the duodenum and has been implicated in a variety of physiological functions including enterocyte HCO(3)(-) supply for secretion and the "sensing" of luminal acid and CO(2). Here, we report the physiological role of the intracellular CAII isoform involvement in acid-, PGE(2,) and forskolin-induced murine duodenal bicarbonate secretion (DBS) in vivo. CAII-deficient and WT littermates were studied in vivo during isoflurane anesthesia. An approximate 10-mm segment of the proximal duodenum with intact blood supply was perfused under different experimental conditions and DBS was titrated by pH immediately. Two-photon confocal microscopy using the pH-sensitive dye SNARF-1F was used to assess duodenocyte pH(i) in vivo. After correction of systemic acidosis by infusion of isotonic Na(2)CO(3), basal DBS was not significantly different in CAII-deficient mice and WT littermates. The duodenal bicarbonate secretory response to acid was almost abolished in CAII-deficient mice, but normal to forskolin- or 16,16-dimethyl PGE(2) stimulation. The complete inhibition of tissue CAs by luminal methazolamide and i.v. acetazolamide completely blocked the response to acid, but did not significantly alter the response to forskolin. While duodenocytes acidified upon luminal perfusion with acid, no significant pH(i) change occurred in CAII-deficient duodenum in vivo. The results suggest that CA II is important for duodenocyte acidification by low luminal pH and for eliciting the acid-mediated HCO(3)(-) secretory response, but is not important in the generation of the secreted HCO(3)(-) ions.


Assuntos
Bicarbonatos/metabolismo , Anidrase Carbônica II/fisiologia , Duodeno/metabolismo , 16,16-Dimetilprostaglandina E2/farmacologia , Acetazolamida/farmacologia , Animais , Dióxido de Carbono/metabolismo , Anidrase Carbônica II/antagonistas & inibidores , Colforsina/farmacologia , Concentração de Íons de Hidrogênio , Metazolamida/farmacologia , Camundongos , Camundongos Endogâmicos C57BL
20.
Am J Respir Cell Mol Biol ; 41(1): 50-8, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19059888

RESUMO

In this study, we evaluated the protective effect and therapeutic potential of the prostaglandin E(2) (PGE(2)) synthetic analog 16,16-dimethyl-PGE(2) (dmPGE(2)) in the animal model of pulmonary fibrosis induced by bleomycin. Mice subjected to intratracheal administration of bleomycin (1 mg/kg) received a dmPGE(2) dose of 30 microg/kg/day by continuous subcutaneous infusion. Bronchoalveolar lavage (BAL); immunohistochemical analysis for IL-1, TNF-alpha, and nitrotyrosine; measurement of fluid content in lung; myeloperoxidase activity assay; and lung histology were performed 1 week later. Lung histology and Sircol assay for collagen deposition were performed 3 weeks after treatments. Changes of body weight and survival rate were also evaluated at 1 and 3 weeks. Compared with bleomycin-treated mice, dmPGE(2) co-treated mice exhibited a reduced degree of body weight loss and mortality rate as well as of lung damage and inflammation, as shown by the significant reduction of: (1) lung infiltration by leukocytes; (2) myeloperoxidase activity; (3) IL-1, TNF-alpha, and nitrotyrosine immunostaining; (4) lung edema; and (5) histologic evidence of lung injury and collagen deposition. In a separate set of experiments, dmPGE(2) treatment was started 3 days after bleomycin administration, and the evaluation of lung damage and inflammation was assessed 4 days later. Importantly, delayed administration of dmPGE(2) also was able to protect from inflammation and lung injury induced by bleomycin. These results, indicating that dmPGE(2) is able to prevent and to reduce bleomycin-induced lung injury through its regulatory and anti-inflammatory properties, encourage further research to find new options for the treatment of pulmonary fibrosis.


Assuntos
16,16-Dimetilprostaglandina E2/farmacologia , Lesão Pulmonar/prevenção & controle , Pulmão/efeitos dos fármacos , Substâncias Protetoras/farmacologia , Fibrose Pulmonar/prevenção & controle , 16,16-Dimetilprostaglandina E2/administração & dosagem , Animais , Bleomicina , Peso Corporal/efeitos dos fármacos , Líquido da Lavagem Broncoalveolar/citologia , Colágeno/metabolismo , Modelos Animais de Doenças , Infusões Subcutâneas , Interleucina-1beta/metabolismo , Pulmão/imunologia , Pulmão/patologia , Lesão Pulmonar/induzido quimicamente , Lesão Pulmonar/imunologia , Lesão Pulmonar/patologia , Masculino , Camundongos , Peroxidase/metabolismo , Pneumonia/induzido quimicamente , Pneumonia/prevenção & controle , Substâncias Protetoras/administração & dosagem , Edema Pulmonar/induzido quimicamente , Edema Pulmonar/prevenção & controle , Fibrose Pulmonar/induzido quimicamente , Fibrose Pulmonar/imunologia , Fibrose Pulmonar/patologia , Fatores de Tempo , Fator de Necrose Tumoral alfa/metabolismo , Tirosina/análogos & derivados , Tirosina/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...