Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 506
Filtrar
1.
Cell Physiol Biochem ; 55(4): 489-504, 2021 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-34416105

RESUMEN

BACKGROUND/AIMS: Diaphragm dysfunction with increased reactive oxygen species (ROS) occurs within 72 hrs post-myocardial infarction (MI) in mice and may contribute to loss of inspiratory maximal pressure and endurance in patients. METHODS: We used wild-type (WT) and whole-body Nox4 knockout (Nox4KO) mice to measure diaphragm bundle force in vitro with a force transducer, mitochondrial respiration in isolated fiber bundles with an O2 sensor, mitochondrial ROS by fluorescence, mRNA (RT-PCR) and protein (immunoblot), and fiber size by histology 72 hrs post-MI. RESULTS: MI decreased diaphragm fiber cross-sectional area (CSA) (~15%, p = 0.015) and maximal specific force (10%, p = 0.005), and increased actin carbonylation (5-10%, p = 0.007) in both WT and Nox4KO. Interestingly, MI did not affect diaphragm mRNA abundance of MAFbx/atrogin-1 and MuRF-1 but Nox4KO decreased it by 20-50% (p < 0.01). Regarding the mitochondria, MI and Nox4KO decreased the protein abundance of citrate synthase and subunits of electron transport system (ETS) complexes and increased mitochondrial O2 flux (JO2) and H2O2 emission (JH2O2) normalized to citrate synthase. Mitochondrial electron leak (JH2O2/JO2) in the presence of ADP was lower in Nox4KO and not changed by MI. CONCLUSION: Our study shows that the early phase post-MI causes diaphragm atrophy, contractile dysfunction, sarcomeric actin oxidation, and decreases citrate synthase and subunits of mitochondrial ETS complexes. These factors are potential causes of loss of inspiratory muscle strength and endurance in patients, which likely contribute to the pathophysiology in the early phase post-MI. Whole-body Nox4KO did not prevent the diaphragm abnormalities induced 72 hrs post-MI, suggesting that systemic pharmacological inhibition of Nox4 will not benefit patients in the early phase post-MI.


Asunto(s)
Diafragma/enzimología , Mitocondrias Musculares/enzimología , Contracción Muscular , Atrofia Muscular/enzimología , Infarto del Miocardio/enzimología , NADPH Oxidasa 4/deficiencia , Animales , Diafragma/patología , Masculino , Ratones , Ratones Noqueados , Mitocondrias Musculares/genética , Mitocondrias Musculares/patología , Atrofia Muscular/genética , Atrofia Muscular/patología , Infarto del Miocardio/genética , Infarto del Miocardio/patología , NADPH Oxidasa 4/metabolismo
2.
Int J Mol Sci ; 22(11)2021 May 25.
Artículo en Inglés | MEDLINE | ID: mdl-34070501

RESUMEN

Our goal was to analyze postmortem tissues of an adult patient with late-onset thymidine kinase 2 (TK2) deficiency who died of respiratory failure. Compared with control tissues, we found a low mtDNA content in the patient's skeletal muscle, liver, kidney, small intestine, and particularly in the diaphragm, whereas heart and brain tissue showed normal mtDNA levels. mtDNA deletions were present in skeletal muscle and diaphragm. All tissues showed a low content of OXPHOS subunits, and this was especially evident in diaphragm, which also exhibited an abnormal protein profile, expression of non-muscular ß-actin and loss of GAPDH and α-actin. MALDI-TOF/TOF mass spectrometry analysis demonstrated the loss of the enzyme fructose-bisphosphate aldolase, and enrichment for serum albumin in the patient's diaphragm tissue. The TK2-deficient patient's diaphragm showed a more profound loss of OXPHOS proteins, with lower levels of catalase, peroxiredoxin 6, cytosolic superoxide dismutase, p62 and the catalytic subunits of proteasome than diaphragms of ventilated controls. Strong overexpression of TK1 was observed in all tissues of the patient with diaphragm showing the highest levels. TK2 deficiency induces a more profound dysfunction of the diaphragm than of other tissues, which manifests as loss of OXPHOS and glycolytic proteins, sarcomeric components, antioxidants and overactivation of the TK1 salvage pathway that is not attributed to mechanical ventilation.


Asunto(s)
ADN Mitocondrial/genética , ADN Mitocondrial/metabolismo , Diafragma/metabolismo , Mitocondrias/metabolismo , Insuficiencia Respiratoria/metabolismo , Timidina Quinasa/deficiencia , Timidina Quinasa/genética , Actinas/metabolismo , Adulto , Autopsia , Encéfalo/metabolismo , Catalasa/metabolismo , Diafragma/enzimología , Femenino , Fructosa-Bifosfato Aldolasa/metabolismo , Gliceraldehído-3-Fosfato Deshidrogenasa (Fosforilante)/metabolismo , Humanos , Intestino Delgado/metabolismo , Riñón/metabolismo , Hígado/metabolismo , Espectrometría de Masas , Mitocondrias/enzimología , Mitocondrias/genética , Músculo Esquelético/metabolismo , Fosforilación Oxidativa , Peroxiredoxina VI/metabolismo , Complejo de la Endopetidasa Proteasomal , Proteoma/genética , Proteoma/metabolismo , Insuficiencia Respiratoria/genética , Insuficiencia Respiratoria/mortalidad , Superóxido Dismutasa/metabolismo , Timidina Quinasa/metabolismo , Regulación hacia Arriba
3.
Food Chem Toxicol ; 140: 111322, 2020 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-32289335

RESUMEN

The perinatal period is characterized by developmental stages with high sensitivity to environmental factors. Among the risk factors, maternal High-Fat Diet (HFD) consumption and early-life pesticide exposure can induce metabolic disorders at adulthood. We established the effects of perigestational exposure to Chlorpyrifos (CPF) and/or HFD on respiratory parameters, sleep apnea and diaphragm contractility in adult rats. Four groups of female rats were exposed starting from 4 months before gestation till the end of lactation period to CPF (1 mg/kg/day vs. vehicle) with or without HFD. Sleep apnea and respiratory parameters were measured by whole-body plethysmography in male offspring at postnatal day 60. Then diaphragm strips were dissected for the measurement of contractility, acetylcholinesterase (AChE) activity, and gene expression. The perigestational exposure to CPF and/or HFD increased the sleep apnea index but decreased the respiratory frequency. The twitch tension and the fatigability index were also increased, associated with reduced AChE activity and elevated mRNA expression of AChE, ryanodine receptor, and myosin heavy chain isoforms. Therefore, the perigestational exposure to either CPF and/or HFD could program the risks for altered ventilatory parameters and diaphragm contractility in young adult offspring despite the lack of direct contact to CPF and/or HFD.


Asunto(s)
Cloropirifos/toxicidad , Diafragma/efectos de los fármacos , Dieta Alta en Grasa , Insecticidas/toxicidad , Contracción Muscular/efectos de los fármacos , Efectos Tardíos de la Exposición Prenatal , Respiración/efectos de los fármacos , Acetilcolinesterasa/metabolismo , Animales , Diafragma/enzimología , Diafragma/fisiología , Femenino , Perfilación de la Expresión Génica , Masculino , Embarazo , Ratas , Ratas Wistar
4.
J Appl Physiol (1985) ; 128(1): 70-77, 2020 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-31774353

RESUMEN

In aged rats, diaphragm muscle (DIAm) reduced specific force and fiber cross-sectional area, sarcopenia, is selective for vulnerable type IIx and/or IIb DIAm fibers, with type I and IIa fibers being resilient. In humans, the oxidative capacity [as measured by maximum succinate dehydrogenase (SDHmax) activity] of fast-type muscle is reduced with aging, with slow-type muscle being unaffected. We hypothesized that in aged Fischer rat DIAm exhibiting sarcopenia, reduced SDHmax activity would occur in type IIx and/or IIb fibers. Rats obtained from the NIA colony (6, 18, and 24 mo old) were euthanized, and ~2-mm-wide DIAm strips were obtained. For SDHmax and fiber type assessments, DIAm strips were stretched (approximately optimal length), fresh frozen in isopentane, and sectioned on a cryostat at 6 µm. SDHmax, quantified by intensity of nitroblue tetrazolium diformazan precipitation, was assessed in a fiber type-specific manner by comparing serial sections labeled with myosin heavy chain (MyHC) antibodies differentiating type I (MyHCSlow), IIa (MyHC2A), and IIx and/or IIb fibers. Isometric DIAm force and fatigue were assessed in DIAm strips by muscle stimulation with supramaximal pulses at a variety of frequencies (5-100 Hz) delivered in 1-s trains. By 24 mo, DIAm sarcopenia was apparent and SDHmax in type IIx and/or IIb fibers activity was reduced ~35% compared with 6-mo-old control DIAm. These results underscore the remarkable fiber type selectivity of type IIx and/or IIb fibers to age-associated perturbations and suggest that reduced mitochondrial oxidative capacity is associated with DIAm sarcopenia.NEW & NOTEWORTHY We examined the oxidative capacity as measured by maximum succinate dehydrogenase activity in older (18 or 24 mo old) Fischer 344 rat diaphragm muscle (DIAm) compared with young rats (6 mo old). In 24-mo-old rats, SDH activity was reduced in type IIx/b DIAm fibers. These SDH changes were concomitant with sarcopenia (reduced specific force and atrophy of type IIx/b DIAm fibers) at 24 mo old. At 18 mo old, there was no change in SDH activity and no evidence of sarcopenia.


Asunto(s)
Envejecimiento/metabolismo , Diafragma/enzimología , Fibras Musculares Esqueléticas/enzimología , Sarcopenia/enzimología , Succinato Deshidrogenasa/metabolismo , Envejecimiento/patología , Animales , Femenino , Masculino , Fibras Musculares Esqueléticas/patología , Ratas , Ratas Endogámicas F344 , Sarcopenia/patología , Succinato Deshidrogenasa/genética
5.
Zhonghua Shao Shang Za Zhi ; 35(7): 507-511, 2019 Jul 20.
Artículo en Chino | MEDLINE | ID: mdl-31357820

RESUMEN

Objective: To explore the occurrence of oxidative stress and antioxidases expression in diaphragm of severely burned rats, so that the mechanism of respiratory muscle atrophy and dysfunction post-burn injury will be further clarified. Methods: Eighty male Wistar rats (aged 7 to 8 weeks) were divided into sham injury group and burn injury group according to the random number table, with 40 rats in each group. Rats in burn injury group were inflicted with 50% total body surface area full-thickness scald (hereinafter referred to as burn) on the back and abdomen by immersing into 80 ℃ water for 15 s and 8 s respectively. Immediately after injury, 40 mL/kg normal saline was injected through abdomen for resuscitation, and the wounds were treated with iodine. Except for immersing into 37 ℃ warm water and no resuscitation, the other treatments of rats in sham injury group were the same as those of burn injury group. Whole diaphragms of 8 rats per time point per group were collected after anesthesia at post injury hour (PIH) 2 and on post injury day (PID) 1, 3, 7, and 14, and muscle mass was determined. The protein carbonyl content was determined by microplate reader. The protein expressions of catalase, superoxide dismutase 2 (SOD2), and glutathione peroxidase 1 were determined by Western blotting. Data were processed with analysis of variance of factorial design, t test, and Bonferroni correction. Results: (1) There were no statistically significant differences in the diaphragm mass of rats between the 2 groups at PIH 2 and on PID 1 (t=0.453, 0.755, P>0.05). The diaphragm mass of rats in burn injury group started to decrease from PID 3, which was significantly lower than that of sham injury group (t=3.321, P<0.01). The diaphragm mass of rats in burn injury group started to increase from PID 7 to PID 14, which was significantly lower than that of sham injury group (t=4.622, 4.380, P<0.01). (2) Protein carbonyl content in diaphragm of rats in burn injury group at PIH 2, and on PID 1, 3, 7, and 14 [(2.7±0.3), (2.5±0.5), (2.4±0.4), (2.5±0.4), (3.2±0.6) pg/mL] was significantly higher than that of sham injury group respectively [(1.2±0.4), (1.6±0.3), (1.5±0.7), (1.7±0.3), (1.8±0.4) pg/mL, t=5.994, 3.263, 3.666, 3.158, 5.763, P<0.05 or P<0.01]. (3) Protein expressions of catalase in diaphragm of rats in burn injury group on PID 1 and 3 were close to those of sham injury group (t=0.339, 0.324, P>0.05). There were no statistically significant differences in protein expressions of SOD2 in diaphragm of rats between the 2 groups at PIH 2 and on PID 1, 3, 7, and 14 (t=1.446, 1.385, 0.757, 1.561, 0.531, P>0.05). There were no statistically significant differences in protein expressions of glutathione peroxidase 1 in diaphragm of rats in the 2 groups at PIH 2 and on PID 1, 3, and 7 (t=0.200, 0.729, 0.385, 1.559, P>0.05). Conclusions: Continuous oxidative stress and relatively insufficient expression of antioxidases in diaphragm induced by burn injury could be a contributor to diaphragm atrophy.


Asunto(s)
Quemaduras/fisiopatología , Diafragma/enzimología , Estrés Oxidativo , Animales , Quemaduras/enzimología , Masculino , Carbonilación Proteica , Distribución Aleatoria , Ratas , Ratas Wistar
6.
Oxid Med Cell Longev ; 2019: 2561929, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31178955

RESUMEN

Cytosolic phospholipase A2 (cPLA2) has been reported to be critical for infection-induced mitochondrial reactive oxygen species (ROS) production and diaphragm dysfunction (DD). In the present study, we aim to investigate whether cPLA2 was involved in ventilator-induced diaphragm dysfunction (VIDD). Our results showed that mechanical ventilation (MV) induced cPLA2 activation in the diaphragm with excessive mitochondrial ROS generation and muscle weakness. Specific inhibition of cPLA2 with CDIBA resulted in decreased mitochondrial ROS levels and improved diaphragm forces. In addition, mitochondria-targeted antioxidant MitoTEMPO attenuated ventilator-induced mitochondrial oxidative stress and downregulated cPLA2 activation in vivo. Both CDIBA and MitoTEMPO were able to attenuate protein degradation, muscle atrophy, and weakness following prolonged MV. Furthermore, laser Doppler imaging showed that MV decreased diaphragm tissue perfusion and induced subsequent hypoxia. An in vitro study also demonstrated a positive association between cPLA2 activation and mitochondrial ROS generation in C2C12 cells cultured under hypoxic condition. Collectively, our study showed that cPLA2 activation positively interacts with mitochondrial ROS generation in the development of VIDD, and ventilator-induced diaphragm hypoxia serves as a possible contributor to this positive feedback loop.


Asunto(s)
Diafragma/fisiopatología , Fosfolipasas A2/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Respiración Artificial/efectos adversos , Respiración Artificial/métodos , Animales , Diafragma/enzimología , Masculino , Ratas , Ratas Wistar
8.
Am J Physiol Lung Cell Mol Physiol ; 316(4): L679-L690, 2019 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-30702345

RESUMEN

Heart failure with reduced ejection fraction (HFREF) increases neutral sphingomyelinase (NSMase) activity and mitochondrial reactive oxygen species (ROS) emission and causes diaphragm weakness. We tested whether a systemic pharmacological NSMase inhibitor or short-hairpin RNA (shRNA) targeting NSMase isoform 3 (NSMase3) would prevent diaphragm abnormalities induced by HFREF caused by myocardial infarction. In the pharmacological intervention, we used intraperitoneal injection of GW4869 or vehicle. In the genetic intervention, we injected adeno-associated virus serotype 9 (AAV9) containing shRNA targeting NSMase3 or a scrambled sequence directly into the diaphragm. We also studied acid sphingomyelinase-knockout mice. GW4869 prevented the increase in diaphragm ceramide content, weakness, and tachypnea caused by HFREF. For example, maximal specific forces (in N/cm2) were vehicle [sham 31 ± 2 and HFREF 26 ± 2 ( P < 0.05)] and GW4869 (sham 31 ± 2 and HFREF 31 ± 1). Respiratory rates were (in breaths/min) vehicle [sham 61 ± 3 and HFREF 84 ± 11 ( P < 0.05)] and GW4869 (sham 66 ± 2 and HFREF 72 ± 2). AAV9-NSMase3 shRNA prevented heightening of diaphragm mitochondrial ROS and weakness [in N/cm2, AAV9-scrambled shRNA: sham 31 ± 2 and HFREF 27 ± 2 ( P < 0.05); AAV9-NSMase3 shRNA: sham 30 ± 1 and HFREF 30 ± 1] but displayed tachypnea. Both wild-type and ASMase-knockout mice with HFREF displayed diaphragm weakness. Our study suggests that activation of NSMase3 causes diaphragm weakness in HFREF, presumably through accumulation of ceramide and elevation in mitochondrial ROS. Our data also reveal a novel inhibitory effect of GW4869 on tachypnea in HFREF likely mediated by changes in neural control of breathing.


Asunto(s)
Diafragma/fisiopatología , Insuficiencia Cardíaca/fisiopatología , Debilidad Muscular/prevención & control , ARN Interferente Pequeño/genética , Esfingomielina Fosfodiesterasa/antagonistas & inhibidores , Esfingomielina Fosfodiesterasa/genética , Compuestos de Anilina/farmacología , Animales , Compuestos de Bencilideno/farmacología , Diafragma/enzimología , Modelos Animales de Enfermedad , Insuficiencia Cardíaca/genética , Insuficiencia Cardíaca/terapia , Humanos , Masculino , Ratones , Ratones Noqueados , Debilidad Muscular/genética , Debilidad Muscular/fisiopatología , Ratas , Ratas Wistar , Esfingomielina Fosfodiesterasa/deficiencia , Volumen Sistólico/genética , Volumen Sistólico/fisiología
9.
Toxicology ; 408: 113-124, 2018 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-30176331

RESUMEN

BACKGROUND: Physostigmine and its analogues neostigmine, pyridostigmine and rivastigmine are carbamates nowadays used in many indications, including antidotal effects against antimuscarinic poisonings, reversal of competitive neuromuscular block, myasthenia gravis, Alzheimer's disease and prophylaxis against nerve agent intoxications. Use of these medicinal carbamates, but also of carbamate insecticides, created need for research into the potential and mechanisms of action of several antidotes against carbamate poisonings, including anticholinergics and oximes. AIM: The goal of this experimental study was to ascertain the life-preserving potential of anticholinergics atropine, hexamethonium and d-tubocurarine, oxime HI-6 and their combinations in rats poisoned with physostigmine or pyridostigmine. MATERIALS AND METHODS: Experiments were performed in Wistar rats. Carbamates were injected subcutaneously (sc) and antidotes intramuscularly (im). Median lethal dose (LD50) in animals treated with antidotes were compared to the ones in saline-treated rats and protective ratios (PRs) were calculated. Atropine (5, 10 and 20 mg/kg), hexamethonium (5, 10 and 20 mg/kg), d-tubocurarine (0.005, 0.010 and 0.020 mg/kg) and oxime HI-6 (25, 50 and 100 mg/kg) were used as monotherapies and in dual combinations, where atropine was the obligatory antidote. Biochemical experiments consisted in measuring of the cholinesterase activities in brain, whole blood and diaphragm in rats 5, 15, 30, 60, 120 and 240 min after poisoning with 0.8 LD50 of physostigmine or pyridostigmine. RESULTS: All the tested antidotes assured some degree of protection against the two carbamates. Atropine and hexamethonium produced better protection in physostigmine-poisoned rats, while d-tubocurarine and HI-6 were more efficacious in pyridostigmine-intoxicated animals. Oxime HI-6 50 mg/kg reactivated acetylcholinesterase (AChE) in brain inhibited by physostigmine and in diaphragm inhibited by pyridostigmine. CONCLUSIONS: Mechanism of physostigmine-induced lethal effect is predominantly central and it involves inhibition of brain AChE, while pyridostigmine produces the same effect exclusively outside the central nervous system, by inhibiting AChE in the respiratory muscles. As a consequence, increasing doses of atropine and their combination with hexamethonium assure excellent protection against physostigmine toxicity, while the best protection against pyridostigmine is provided by a strictly peripherally acting antinicotinic d-tubocurarine and bispyridinium oxime HI-6. The oxime acts as antidote against physostigmine and pyridostigmine poisoning by reactivating AChE in the brain and diaphragm, respectively.


Asunto(s)
Antídotos/farmacología , Antagonistas Colinérgicos/farmacología , Reactivadores de la Colinesterasa/farmacología , Síndromes de Neurotoxicidad/tratamiento farmacológico , Fisostigmina , Bromuro de Piridostigmina , Acetilcolinesterasa/metabolismo , Animales , Atropina/farmacología , Encéfalo/efectos de los fármacos , Encéfalo/enzimología , Diafragma/efectos de los fármacos , Diafragma/enzimología , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Quimioterapia Combinada , Activación Enzimática , Proteínas Ligadas a GPI/metabolismo , Hexametonio/farmacología , Masculino , Síndromes de Neurotoxicidad/enzimología , Síndromes de Neurotoxicidad/etiología , Oximas/farmacología , Compuestos de Piridinio/farmacología , Ratas Wistar , Tubocurarina/farmacología
10.
J Pathol ; 244(3): 323-333, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29214629

RESUMEN

Chronic muscle inflammation is a critical feature of Duchenne muscular dystrophy and contributes to muscle fibre injury and disease progression. Although previous studies have implicated T cells in the development of muscle fibrosis, little is known about their role during the early stages of muscular dystrophy. Here, we show that T cells are among the first cells to infiltrate mdx mouse dystrophic muscle, prior to the onset of necrosis, suggesting an important role in early disease pathogenesis. Based on our comprehensive analysis of the kinetics of the immune response, we further identify the early pre-necrotic stage of muscular dystrophy as the relevant time frame for T-cell-based interventions. We focused on protein kinase C θ (PKCθ, encoded by Prkcq), a critical regulator of effector T-cell activation, as a potential target to inhibit T-cell activity in dystrophic muscle. Lack of PKCθ not only reduced the frequency and number of infiltrating T cells but also led to quantitative and qualitative changes in the innate immune cell infiltrate in mdx/Prkcq-/- muscle. These changes were due to the inhibition of T cells, since PKCθ was necessary for T-cell but not for myeloid cell infiltration of acutely injured muscle. Targeting T cells with a PKCθ inhibitor early in the disease process markedly diminished the size of the inflammatory cell infiltrate and resulted in reduced muscle damage. Moreover, diaphragm necrosis and fibrosis were also reduced following treatment. Overall, our findings identify the early T-cell infiltrate as a therapeutic target and highlight the potential of PKCθ inhibition as a therapeutic approach to muscular dystrophy. Copyright © 2017 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Asunto(s)
Quimiotaxis de Leucocito/efectos de los fármacos , Diafragma/efectos de los fármacos , Distrofia Muscular Animal/prevención & control , Proteína Quinasa C-theta/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/farmacología , Linfocitos T/efectos de los fármacos , Animales , Diafragma/enzimología , Diafragma/inmunología , Diafragma/patología , Modelos Animales de Enfermedad , Fibrosis , Inmunidad Innata/efectos de los fármacos , Ratones Endogámicos C57BL , Ratones Endogámicos mdx , Ratones Noqueados , Distrofia Muscular Animal/enzimología , Distrofia Muscular Animal/inmunología , Distrofia Muscular Animal/patología , Necrosis , Proteína Quinasa C-theta/deficiencia , Proteína Quinasa C-theta/genética , Proteína Quinasa C-theta/metabolismo , Índice de Severidad de la Enfermedad , Transducción de Señal/efectos de los fármacos , Linfocitos T/enzimología , Linfocitos T/inmunología , Factores de Tiempo
11.
Mol Med Rep ; 16(4): 5263-5268, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28849127

RESUMEN

Our previous study demonstrated that sepsis may decrease the activity of acetylcholinesterase (AChE) at the neuromuscular junction (NMJ) of the diaphragm at 24 h, and thus improve the antagonistic action of neostigmine on rocuronium. The present study aimed to determine the effects of sepsis on AChE activity over 2 weeks, which is a more clinically relevant time period. Furthermore, the present study aimed to elucidate the association between AChE activity and its expression at the NMJ during sepsis. Male adult Sprague­Dawley rats were randomly divided into the sham or sepsis groups. Sepsis was induced by cecal ligation and puncture. On days 1, 3, 7 and 14 after surgery, AChE activity at the NMJ of the diaphragm was detected using a modified Karnovsky and Roots method. Furthermore, AChE expression levels at the NMJ, and in the whole muscle fibers of the diaphragm, were detected by immunohistofluorescence staining and western blot analysis, respectively. AChE activity was significantly decreased in the sepsis group, with its lowest level detected on day 7; however, its activity had partially recovered on day 14 (P<0.01). AChE activity was positively correlated (r=0.975, P=0.025) with its expression at the NMJ, which showed a similar trend over 2 weeks of sepsis. The protein expression levels of AChE in the whole muscle fibers of the diaphragm were significantly decreased on days 1, 3 and 7 in the sepsis group (P<0.01), with the lowest level observed on day 3. In conclusion, sepsis decreased AChE activity by reducing its expression at the NMJ over 14 days; the reduced expression of AChE at the NMJ might be as a result of its reduced muscular production.


Asunto(s)
Acetilcolinesterasa/metabolismo , Unión Neuromuscular/enzimología , Sepsis/enzimología , Sepsis/patología , Animales , Diafragma/enzimología , Diafragma/patología , Masculino , Fibras Musculares Esqueléticas/enzimología , Fibras Musculares Esqueléticas/patología , Ratas Sprague-Dawley
12.
Braz J Cardiovasc Surg ; 32(2): 104-110, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28492791

RESUMEN

INTRODUCTION:: Enhanced respiratory muscle strength in patients with heart failure positively alters the clinical trajectory of heart failure. In an experimental model, respiratory muscle training in rats with heart failure has been shown to improve cardiopulmonary function through mechanisms yet to be entirely elucidated. OBJECTIVE:: The present report aimed to evaluate the respiratory muscle training effects in diaphragm citrate synthase activity and hemodynamic function in rats with heart failure. METHODS:: Wistar rats were divided into four experimental groups: sedentary sham (Sed-Sham, n=8), trained sham (RMT-Sham, n=8), sedentary heart failure (Sed-HF, n=7) and trained heart failure (RMT-HF, n=7). The animals were submitted to a RMT protocol performed 30 minutes a day, 5 days/week, for 6 weeks. RESULTS:: In rats with heart failure, respiratory muscle training decreased pulmonary congestion and right ventricular hypertrophy. Deleterious alterations in left ventricular pressures, as well as left ventricular contractility and relaxation, were assuaged by respiratory muscle training in heart failure rats. Citrate synthase activity, which was significantly reduced in heart failure rats, was preserved by respiratory muscle training. Additionally, a negative correlation was found between citrate synthase and left ventricular end diastolic pressure and positive correlation was found between citrate synthase and left ventricular systolic pressure. CONCLUSION:: Respiratory muscle training produces beneficial adaptations in the diaphragmatic musculature, which is linked to improvements in left ventricular hemodynamics and blood pressure in heart failure rats. The RMT-induced improvements in cardiac architecture and the oxidative capacity of the diaphragm may improve the clinical trajectory of patients with heart failure.


Asunto(s)
Ejercicios Respiratorios/métodos , Citrato (si)-Sintasa/metabolismo , Diafragma/enzimología , Insuficiencia Cardíaca/enzimología , Insuficiencia Cardíaca/fisiopatología , Hemodinámica/fisiología , Animales , Presión Sanguínea/fisiología , Diafragma/fisiología , Masculino , Modelos Animales , Infarto del Miocardio/fisiopatología , Ratas Wistar , Mecánica Respiratoria/fisiología
13.
J Pediatr Surg ; 52(6): 1035-1039, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28363469

RESUMEN

BACKGROUND: Congenital diaphragmatic hernia (CDH) has been attributed to various developmental abnormalities of the underlying tissue components. N-deacetylase-N-sulfotransferase-1 (Ndst1) is a strongly expressed biosynthetic enzyme in endothelial cells, which has recently been identified as an important factor during diaphragmatic vascularization. Loss of endothelial Ndst1 has been demonstrated to cause angiogenic defects in the developing diaphragm and disrupt normal diaphragmatic development. Furthermore, deficiency of Ndst1 diminishes the expression of slit homolog 3 (Slit3), a known CDH-related gene that has been associated with reduced vascular density and muscle defects in the diaphragm of Slit3-/- mice. We hypothesized that expression of Ndst1 and Slit3 is decreased in the diaphragmatic vasculature of fetal rats with nitrofen-induced CDH. METHODS: Time-mated rats received either nitrofen or vehicle on gestational day 9 (D9). Fetal diaphragms were microdissected on D13, D15 and D18, and divided into control and nitrofen-exposed specimens. Gene expression levels of Ndst1 and Slit3 were assessed using qRT-PCR. Immunofluorescence-double-staining for Ndst1 and Slit3 was performed to evaluate protein expression and localization. RESULTS: Relative mRNA expression of Ndst1 and Slit3 was significantly decreased in pleuroperitoneal folds (D13), developing diaphragms (D15) and fully muscularized diaphragms (D18) of nitrofen-exposed fetuses compared to controls. Confocal-laser-scanning-microscopy revealed markedly diminished Ndst1 and Slit3 expression in endothelial cells within the diaphragmatic vasculature on D13, D15 and D18 compared to controls. CONCLUSIONS: Down-regulation of Ndst1 signaling in the developing diaphragm may impair endothelial cell migration and angiogenesis, thus leading to defective diaphragmatic vascular development and CDH. LEVEL OF EVIDENCE: Ib.


Asunto(s)
Diafragma/enzimología , Regulación hacia Abajo , Regulación del Desarrollo de la Expresión Génica , Hernias Diafragmáticas Congénitas/enzimología , Proteínas de la Membrana/genética , Transducción de Señal , Sulfotransferasas/genética , Animales , Biomarcadores/metabolismo , Diafragma/irrigación sanguínea , Diafragma/embriología , Células Endoteliales/enzimología , Femenino , Marcadores Genéticos , Hernias Diafragmáticas Congénitas/inducido químicamente , Hernias Diafragmáticas Congénitas/embriología , Hernias Diafragmáticas Congénitas/genética , Proteínas de la Membrana/metabolismo , Éteres Fenílicos , Distribución Aleatoria , Ratas , Ratas Sprague-Dawley , Sulfotransferasas/metabolismo
14.
Rev. bras. cir. cardiovasc ; 32(2): 104-110, Mar.-Apr. 2017. tab, graf
Artículo en Inglés | LILACS | ID: biblio-843478

RESUMEN

Abstract INTRODUCTION: Enhanced respiratory muscle strength in patients with heart failure positively alters the clinical trajectory of heart failure. In an experimental model, respiratory muscle training in rats with heart failure has been shown to improve cardiopulmonary function through mechanisms yet to be entirely elucidated. OBJECTIVE: The present report aimed to evaluate the respiratory muscle training effects in diaphragm citrate synthase activity and hemodynamic function in rats with heart failure. METHODS: Wistar rats were divided into four experimental groups: sedentary sham (Sed-Sham, n=8), trained sham (RMT-Sham, n=8), sedentary heart failure (Sed-HF, n=7) and trained heart failure (RMT-HF, n=7). The animals were submitted to a RMT protocol performed 30 minutes a day, 5 days/week, for 6 weeks. RESULTS: In rats with heart failure, respiratory muscle training decreased pulmonary congestion and right ventricular hypertrophy. Deleterious alterations in left ventricular pressures, as well as left ventricular contractility and relaxation, were assuaged by respiratory muscle training in heart failure rats. Citrate synthase activity, which was significantly reduced in heart failure rats, was preserved by respiratory muscle training. Additionally, a negative correlation was found between citrate synthase and left ventricular end diastolic pressure and positive correlation was found between citrate synthase and left ventricular systolic pressure. CONCLUSION: Respiratory muscle training produces beneficial adaptations in the diaphragmatic musculature, which is linked to improvements in left ventricular hemodynamics and blood pressure in heart failure rats. The RMT-induced improvements in cardiac architecture and the oxidative capacity of the diaphragm may improve the clinical trajectory of patients with heart failure.


Asunto(s)
Animales , Masculino , Ejercicios Respiratorios/métodos , Diafragma/enzimología , Citrato (si)-Sintasa/metabolismo , Insuficiencia Cardíaca/enzimología , Insuficiencia Cardíaca/fisiopatología , Hemodinámica/fisiología , Presión Sanguínea/fisiología , Diafragma/fisiología , Mecánica Respiratoria/fisiología , Ratas Wistar , Modelos Animales , Infarto del Miocardio/fisiopatología
15.
J Cell Physiol ; 232(12): 3744-3761, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28177129

RESUMEN

Strategies to treat cachexia are still at its infancy. Enhanced muscle protein breakdown and ubiquitin-proteasome system are common features of cachexia associated with chronic conditions including lung cancer (LC). Poly(ADP-ribose) polymerases (PARP), which play a major role in chromatin structure regulation, also underlie maintenance of muscle metabolism and body composition. We hypothesized that protein catabolism, proteolytic markers, muscle fiber phenotype, and muscle anabolism may improve in respiratory and limb muscles of LC-cachectic Parp-1-deficient (Parp-1-/- ) and Parp-2-/- mice. In diaphragm and gastrocnemius of LC (LP07 adenocarcinoma) bearing mice (wild type, Parp-1-/- , and Parp-2-/- ), PARP activity (ADP-ribose polymers, pADPr), redox balance, muscle fiber phenotype, apoptotic nuclei, tyrosine release, protein ubiquitination, muscle-specific E3 ligases, NF-κB signaling pathway, markers of muscle anabolism (Akt, mTOR, p70S6K, and mitochondrial DNA) were evaluated along with body and muscle weights, and limb muscle force. Compared to wild type cachectic animals, in both respiratory and limb muscles of Parp-1-/- and Parp-2-/- cachectic mice: cancer induced-muscle wasting characterized by increased PARP activity, protein oxidation, tyrosine release, and ubiquitin-proteasome system (total protein ubiquitination, atrogin-1, and 20S proteasome C8 subunit) were blunted, the reduction in contractile myosin and atrophy of the fibers was attenuated, while no effects were seen in other structural features (inflammatory cells, internal or apoptotic nuclei), and markers of muscle anabolism partly improved. Activation of either PARP-1 or -2 is likely to play a role in muscle protein catabolism via oxidative stress, NF-κB signaling, and enhanced proteasomal degradation in cancer-induced cachexia. Therapeutic potential of PARP activity inhibition deserves attention.


Asunto(s)
Caquexia/etiología , Neoplasias Pulmonares/complicaciones , Fibras Musculares Esqueléticas/enzimología , Proteínas Musculares/metabolismo , Músculo Esquelético/enzimología , Estrés Oxidativo , Poli(ADP-Ribosa) Polimerasa-1/metabolismo , Poli(ADP-Ribosa) Polimerasas/metabolismo , Proteolisis , Animales , Apoptosis , Biomarcadores/metabolismo , Caquexia/enzimología , Caquexia/genética , Caquexia/patología , Línea Celular Tumoral , Diafragma/enzimología , Diafragma/patología , Femenino , Genotipo , Neoplasias Pulmonares/enzimología , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Ratones de la Cepa 129 , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Mitocondrias Musculares/metabolismo , Mitocondrias Musculares/patología , Fibras Musculares Esqueléticas/patología , Músculo Esquelético/patología , FN-kappa B/metabolismo , Tamaño de los Órganos , Fenotipo , Poli(ADP-Ribosa) Polimerasa-1/deficiencia , Poli(ADP-Ribosa) Polimerasa-1/genética , Poli(ADP-Ribosa) Polimerasas/deficiencia , Poli(ADP-Ribosa) Polimerasas/genética , Complejo de la Endopetidasa Proteasomal/metabolismo , Transducción de Señal , Factores de Tiempo , Ubiquitinación
16.
Am J Physiol Lung Cell Mol Physiol ; 310(10): L975-84, 2016 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-26968769

RESUMEN

Calpain activation contributes to the development of infection-induced diaphragm weakness, but the mechanisms by which infections activate calpain are poorly understood. We postulated that skeletal muscle calcium-dependent phospholipase A2 (cPLA2) is activated by cytokines and has downstream effects that induce calpain activation and muscle weakness. We determined whether cPLA2 activation mediates cytokine-induced calpain activation in isolated skeletal muscle (C2C12) cells and infection-induced diaphragm weakness in mice. C2C12 cells were treated with the following: 1) vehicle; 2) cytomix (TNF-α 20 ng/ml, IL-1ß 50 U/ml, IFN-γ 100 U/ml, LPS 10 µg/ml); 3) cytomix + AACOCF3, a cPLA2 inhibitor (10 µM); or 4) AACOCF3 alone. At 24 h, we assessed cell cPLA2 activity, mitochondrial superoxide generation, calpain activity, and calpastatin activity. We also determined if SS31 (10 µg/ml), a mitochondrial superoxide scavenger, reduced cytomix-mediated calpain activation. Finally, we determined if CDIBA (10 µM), a cPLA2 inhibitor, reduced diaphragm dysfunction due to cecal ligation puncture in mice. Cytomix increased C2C12 cell cPLA2 activity (P < 0.001) and superoxide generation; AACOCF3 and SS31 blocked increases in superoxide generation (P < 0.001). Cytomix also activated calpain (P < 0.001) and inactivated calpastatin (P < 0.01); both AACOCF3 and SS31 prevented these changes. Cecal ligation puncture reduced diaphragm force in mice, and CDIBA prevented this reduction (P < 0.001). cPLA2 modulates cytokine-induced calpain activation in cells and infection-induced diaphragm weakness in animals. We speculate that therapies that inhibit cPLA2 may prevent diaphragm weakness in infected, critically ill patients.


Asunto(s)
Diafragma/fisiopatología , Fosfolipasas A2 Grupo IV/fisiología , Sepsis/enzimología , Sepsis/fisiopatología , Animales , Línea Celular , Diafragma/enzimología , Diafragma/microbiología , Ratones , Debilidad Muscular , Superóxidos/metabolismo
17.
Am J Physiol Regul Integr Comp Physiol ; 310(8): R707-10, 2016 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-26864813

RESUMEN

Respiratory dysfunction is prevalent in critically ill patients and can lead to adverse clinical outcomes, including respiratory failure and increased mortality. Respiratory muscles, which normally sustain respiration through inspiratory muscle contractions, become weakened during critical illness, and recent studies suggest that respiratory muscle weakness is related to systemic inflammation. Here, we investigate the pathophysiological role of the inflammatory JAK1/3 signaling pathway in diaphragm weakness in two distinct experimental models of critical illness. In the first experiment, mice received subcutaneous injections of PBS or C26 cancer cells and were fed chow formulated with or without the JAK1/3 inhibitor R548 for 26 days. Diaphragm specific force was significantly reduced in tumor-bearing mice receiving standard chow; however, treatment with the JAK1/3 inhibitor completely prevented diaphragm weakness. Diaphragm cross-sectional area was diminished by ∼25% in tumor-bearing mice but was similar to healthy mice in tumor-bearing animals treated with R548. In the second study, mice received sham surgery or coronary artery ligation, leading to myocardial infarction (MI), and were treated with R548 or vehicle 1 h postsurgery, and once daily for 3 days. Diaphragm specific force was comparable between sham surgery/vehicle, sham surgery/R548 and MI/R548 groups, but significantly decreased in the MI/vehicle group. Markers of oxidative damage and activated caspase-3, mechanisms previously identified to reduce muscle contractility, were not elevated in diaphragm extracts. These experiments implicate JAK1/3 signaling in cancer- and MI-mediated diaphragm weakness in mice, and provide a compelling case for further investigation.


Asunto(s)
Neoplasias del Colon/tratamiento farmacológico , Diafragma/efectos de los fármacos , Janus Quinasa 1/antagonistas & inhibidores , Janus Quinasa 3/antagonistas & inhibidores , Fuerza Muscular/efectos de los fármacos , Debilidad Muscular/prevención & control , Infarto del Miocardio/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/farmacología , Trastornos Respiratorios/prevención & control , Animales , Caquexia/enzimología , Caquexia/etiología , Caquexia/fisiopatología , Neoplasias del Colon/complicaciones , Neoplasias del Colon/enzimología , Neoplasias del Colon/fisiopatología , Diafragma/enzimología , Diafragma/fisiopatología , Modelos Animales de Enfermedad , Janus Quinasa 1/metabolismo , Janus Quinasa 3/metabolismo , Masculino , Ratones Endogámicos C57BL , Contracción Muscular/efectos de los fármacos , Debilidad Muscular/enzimología , Debilidad Muscular/etiología , Debilidad Muscular/fisiopatología , Infarto del Miocardio/complicaciones , Infarto del Miocardio/enzimología , Infarto del Miocardio/fisiopatología , Respiración/efectos de los fármacos , Trastornos Respiratorios/enzimología , Trastornos Respiratorios/etiología , Trastornos Respiratorios/fisiopatología , Transducción de Señal/efectos de los fármacos
18.
Hum Pathol ; 49: 27-32, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26826406

RESUMEN

Complex I deficiency causes Leigh syndrome, fatal infant lactic acidosis, and neonatal cardiomyopathy. Mutations in more than 100 nuclear DNA and mitochondrial DNA genes miscode for complex I subunits or assembly factors. ACAD9 is an acyl-CoA dehydrogenase with a novel function in assembly of complex I; biallelic mutations cause progressive encephalomyopathy, recurrent Reye syndrome, and fatal cardiomyopathy. We describe the first autopsy in fatal neonatal lethal lactic acidosis due to mutations in ACAD9 that reduced complex I activity. We identified mitochondrial hyperplasia in cardiac myocytes, diaphragm muscle, and liver and renal tubules in formalin-fixed, paraffin-embedded tissue using immunohistochemistry for mitochondrial antigens. Whole-exome sequencing revealed compound heterozygous variants in the ACAD9 gene: c.187G>T (p.E63*) and c.941T>C (p.L314P). The nonsense mutation causes late infantile lethality; the missense variant is novel. Autopsy-derived fibroblasts had reduced complex I activity (53% of control) with normal activity in complexes II to IV, similar to reported cases of ACAD9 deficiency.


Asunto(s)
Acidosis Láctica/diagnóstico , Acidosis/diagnóstico , Acil-CoA Deshidrogenasa/deficiencia , Acil-CoA Deshidrogenasas/genética , Errores Innatos del Metabolismo de los Aminoácidos/diagnóstico , Cardiomiopatía Hipertrófica/diagnóstico , Codón sin Sentido , Diafragma/patología , Complejo I de Transporte de Electrón/deficiencia , Túbulos Renales/patología , Enfermedad de Leigh/diagnóstico , Mitocondrias Cardíacas/patología , Mitocondrias Hepáticas/patología , Mitocondrias Musculares/patología , Enfermedades Mitocondriales/diagnóstico , Insuficiencia Multiorgánica/diagnóstico , Debilidad Muscular/diagnóstico , Acidosis/enzimología , Acidosis/genética , Acidosis/patología , Acidosis Láctica/enzimología , Acidosis Láctica/genética , Acidosis Láctica/patología , Acil-CoA Deshidrogenasa/genética , Acil-CoA Deshidrogenasas/deficiencia , Errores Innatos del Metabolismo de los Aminoácidos/enzimología , Errores Innatos del Metabolismo de los Aminoácidos/genética , Errores Innatos del Metabolismo de los Aminoácidos/patología , Autopsia , Cardiomiopatía Hipertrófica/enzimología , Cardiomiopatía Hipertrófica/genética , Cardiomiopatía Hipertrófica/patología , Causas de Muerte , Células Cultivadas , Análisis Mutacional de ADN , ADN Mitocondrial/genética , Diafragma/enzimología , Complejo I de Transporte de Electrón/genética , Resultado Fatal , Fibroblastos/enzimología , Fibroblastos/patología , Predisposición Genética a la Enfermedad , Humanos , Hiperplasia , Inmunohistoquímica , Recién Nacido , Túbulos Renales/enzimología , Enfermedad de Leigh/enzimología , Enfermedad de Leigh/genética , Enfermedad de Leigh/patología , Masculino , Mitocondrias Cardíacas/enzimología , Mitocondrias Hepáticas/enzimología , Mitocondrias Musculares/enzimología , Enfermedades Mitocondriales/enzimología , Enfermedades Mitocondriales/genética , Enfermedades Mitocondriales/patología , Insuficiencia Multiorgánica/enzimología , Insuficiencia Multiorgánica/genética , Insuficiencia Multiorgánica/patología , Debilidad Muscular/enzimología , Debilidad Muscular/genética , Debilidad Muscular/patología , Fenotipo , Transfección
19.
Muscle Nerve ; 53(1): 91-5, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25965940

RESUMEN

INTRODUCTION: Duchenne muscular dystrophy (DMD) is a degenerative disease of skeletal, respiratory, and cardiac muscles caused by defects in the dystrophin gene. More recently, brain involvement has been verified. Mitochondrial dysfunction and oxidative stress may underlie the pathophysiology of DMD. In this study we evaluate Krebs cycle enzymes activity in the cerebral cortex, diaphragm, and quadriceps muscles of mdx mice. METHODS: Cortex, diaphragm, and quadriceps tissues from male dystrophic mdx and control mice were used. RESULTS: We observed increased malate dehydrogenase activity in the cortex; increased malate dehydrogenase and succinate dehydrogenase activities in the diaphragm; and increased citrate synthase, isocitrate dehydrogenase, and malate dehydrogenase activities in the quadriceps of mdx mice. CONCLUSION: This study showed increased activity of Krebs cycle enzymes in cortex, quadriceps, and diaphragm in mdx mice.


Asunto(s)
Citrato (si)-Sintasa/metabolismo , Ciclo del Ácido Cítrico/fisiología , Isocitrato Deshidrogenasa/metabolismo , Malato Deshidrogenasa/metabolismo , Distrofia Muscular de Duchenne/enzimología , Animales , Corteza Cerebral/enzimología , Diafragma/enzimología , Modelos Animales de Enfermedad , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos mdx , Músculo Esquelético/enzimología , Distrofia Muscular de Duchenne/genética
20.
Life Sci ; 142: 1-7, 2015 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-26459051

RESUMEN

AIMS: Lymphangiogenesis is frequently observed during inflammation, and this inflammation-induced lymphangiogenesis (IL) is a phenomenon actively involved in the pathophysiology of inflammation. We explored the roles of an inducible prostaglandin E synthase, mPGES-1, in IL elicited by lipopolysaccharide (LPS). MAIN METHODS: Peritonitis was induced in mice by intraperitoneal injection of LPS (E. coli 0111-B4; 25µg/mouse every 2days), and IL was evaluated by LYVE-1 immunostaining of whole-mount diaphragm tissues. KEY FINDINGS: Compared to vehicle-treated wild-type (WT) mice, lymphatics in the diaphragms of mice injected with LPS were widened and the number of LYVE-1-positive ladder-structured lymphatics increased temporally. This increase in lymphangiogenesis was accompanied by increased expression of vascular endothelial growth factor (VEGF)-C/D in the diaphragms. In mice treated with celecoxib, a cyclooxygenase-2 inhibitor, IL was suppressed with reduced expression of VEGF-C/D. This was also observed in mPGES-1 knockout mice (KO). Immunoreactive COX-2 and mPGES-1 were detected in both CD11b-positive and CD3ε-positivecells in the diaphragm. When FITC-dextran was injected into the peritoneal cavities, the amount of residual FITC-dextran was reduced significantly in WT mice injected with LPS, and this reduction was significantly decreased in mPGES-1 KO mice. SIGNIFICANCE: The present results suggest that mPGES-1 plays a significant role in lymphangiogenesis during inflammation, and represents a novel target for controlling IL.


Asunto(s)
Oxidorreductasas Intramoleculares/metabolismo , Lipopolisacáridos/toxicidad , Linfangiogénesis/efectos de los fármacos , Peritonitis/inducido químicamente , Peritonitis/enzimología , Animales , Celecoxib/farmacología , Diafragma/enzimología , Diafragma/patología , Modelos Animales de Enfermedad , Glicoproteínas/genética , Glicoproteínas/metabolismo , Oxidorreductasas Intramoleculares/antagonistas & inhibidores , Oxidorreductasas Intramoleculares/genética , Linfangiogénesis/genética , Masculino , Proteínas de Transporte de Membrana , Ratones , Ratones Noqueados , Peritonitis/genética , Peritonitis/patología , Prostaglandina-E Sintasas , Factor C de Crecimiento Endotelial Vascular/genética , Factor C de Crecimiento Endotelial Vascular/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...