Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 91
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Am J Respir Cell Mol Biol ; 70(2): 94-109, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-37874230

RESUMEN

Oxidative stress, inflammation, and endoplasmic reticulum (ER) stress sequentially occur in bronchopulmonary dysplasia (BPD), and all result in DNA damage. When DNA damage becomes irreparable, tumor suppressors increase, followed by apoptosis or senescence. Although cellular senescence contributes to wound healing, its persistence inhibits growth. Therefore, we hypothesized that cellular senescence contributes to BPD progression. Human autopsy lungs were obtained. Sprague-Dawley rat pups exposed to 95% oxygen between Postnatal Day 1 (P1) and P10 were used as the BPD phenotype. N-acetyl-lysyltyrosylcysteine-amide (KYC), tauroursodeoxycholic acid (TUDCA), and Foxo4 dri were administered intraperitoneally to mitigate myeloperoxidase oxidant generation, ER stress, and cellular senescence, respectively. Lungs were examined by histology, transcriptomics, and immunoblotting. Cellular senescence increased in rat and human BPD lungs, as evidenced by increased oxidative DNA damage, tumor suppressors, GL-13 stain, and inflammatory cytokines with decreased cell proliferation and lamin B expression. Cellular senescence-related transcripts in BPD rat lungs were enriched at P10 and P21. Single-cell RNA sequencing showed increased cellular senescence in several cell types, including type 2 alveolar cells. In addition, Foxo4-p53 binding increased in BPD rat lungs. Daily TUDCA or KYC, administered intraperitoneally, effectively decreased cellular senescence, improved alveolar complexity, and partially maintained the numbers of type 2 alveolar cells. Foxo4 dri administered at P4, P6, P8, and P10 led to outcomes similar to TUDCA and KYC. Our data suggest that cellular senescence plays an essential role in BPD after initial inducement by hyperoxia. Reducing myeloperoxidase toxic oxidant production, ER stress, and attenuating cellular senescence are potential therapeutic strategies for halting BPD progression.


Asunto(s)
Displasia Broncopulmonar , Hiperoxia , Ácido Tauroquenodesoxicólico , Recién Nacido , Animales , Ratas , Humanos , Displasia Broncopulmonar/patología , Hiperoxia/metabolismo , Ratas Sprague-Dawley , Pulmón/patología , Senescencia Celular , Peroxidasa/metabolismo , Oxidantes , Animales Recién Nacidos , Modelos Animales de Enfermedad
2.
J Biol Chem ; 292(6): 2369-2378, 2017 02 10.
Artículo en Inglés | MEDLINE | ID: mdl-28028182

RESUMEN

Stress-inducible heat shock protein 70 (hsp70) interacts with superoxide dismutase 2 (SOD2) in the cytosol after synthesis to transfer the enzyme to the mitochondria for subsequent activation. However, the structural basis for this interaction remains to be defined. To map the SOD2-binding site in hsp70, mutants of hsp70 were made and tested for their ability to bind SOD2. These studies showed that SOD2 binds in the amino acid 393-537 region of the chaperone. To map the hsp70-binding site in SOD2, we used a series of pulldown assays and showed that hsp70 binds to the amino-terminal domain of SOD2. To better define the binding site, we used a series of decoy peptides derived from the primary amino acid sequence in the SOD2-binding site in hsp70. This study shows that SOD2 specifically binds to hsp70 at 445GERAMT450 Small peptides containing GERAMT inhibited the transfer of SOD2 to the mitochondria and decreased SOD2 activity in vitro and in vivo To determine the amino acid residues in hsp70 that are critical for SOD2 interactions, we substituted each amino acid residue for alanine or more conservative residues, glutamine or asparagine, in the GERAMT-binding site. Substitutions of E446A/Q and R447A/Q inhibited the ability of the GERAMT peptide to bind SOD2 and preserved SOD2 function more than other substitutions. Together, these findings indicate that the GERAMT sequence is critical for hsp70-mediated regulation of SOD2 and that Glu446 and Arg447 cooperate with other amino acid residues in the GERAMT-binding site for proper chaperone-dependent regulation of SOD2 antioxidant function.


Asunto(s)
Arginina/metabolismo , Ácido Glutámico/metabolismo , Proteínas HSP70 de Choque Térmico/metabolismo , Superóxido Dismutasa/metabolismo , Sustitución de Aminoácidos , Animales , Sitios de Unión , Células Cultivadas , Proteínas HSP70 de Choque Térmico/química , Mitocondrias/metabolismo , Ratas , Ovinos , Superóxidos/metabolismo
4.
Cell Immunol ; 317: 1-8, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28511921

RESUMEN

Myeloperoxidase (MPO) is the most toxic enzyme found in the azurophilic granules of neutrophils. MPO utilizes H2O2 to generate hypochlorous acid (HClO) and other reactive moieties, which kill pathogens during infections. In contrast, in the setting of sterile inflammation, MPO and MPO-derived oxidants are thought to be pathogenic, promoting inflammation and causing tissue damage. In contrast, evidence also exists that MPO can limit the extent of immune responses. Elevated MPO levels and activity are observed in a number of autoimmune diseases including in the central nervous system (CNS) of multiple sclerosis (MS) and the joints of rheumatoid arthritis (RA) patients. A pathogenic role for MPO in driving autoimmune inflammation was demonstrated using mouse models. Mechanisms whereby MPO is thought to contribute to disease pathogenesis include tuning of adaptive immune responses and/or the induction of vascular permeability.


Asunto(s)
Enfermedades Autoinmunes/inmunología , Autoinmunidad , Células Dendríticas/inmunología , Inflamación/inmunología , Peroxidasa/metabolismo , Animales , Permeabilidad Capilar , Humanos , Ratones , Terapia Molecular Dirigida
5.
J Neurochem ; 136(4): 826-836, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26560636

RESUMEN

Oxidative stress is thought to contribute to disease pathogenesis in the central nervous system (CNS) disease multiple sclerosis (MS). Myeloperoxidase (MPO), a potent peroxidase that generates toxic radicals and oxidants, is increased in the CNS during MS. However, the exact mechanism whereby MPO drives MS pathology is not known. We addressed this question by inhibiting MPO in mice with experimental autoimmune encephalomyelitis (EAE) using our non-toxic MPO inhibitor N-acetyl lysyltyrosylcysteine amide (KYC). We found that therapeutic administration of KYC for 5 days starting at the peak of disease significantly attenuated EAE disease severity, reduced myeloid cell numbers and permeability of the blood-brain barrier. These data indicate that inhibition of MPO by KYC restores blood-brain barrier integrity thereby limiting migration of myeloid cells into the CNS that drive EAE pathogenesis. In addition, these observations indicate that KYC may be an effective therapeutic agent for the treatment of MS. We propose that during experimental autoimmune encephalomyelitis (EAE) onset macrophages and neutrophils migrate into the CNS and upon activation release myeloperoxidase (MPO) that promotes disruption of the blood-brain barrier (BBB) and disease progression. KYC restores BBB function by inhibiting MPO activity and in so doing ameliorates disease progression.

6.
J Neuroinflammation ; 13(1): 119, 2016 05 24.
Artículo en Inglés | MEDLINE | ID: mdl-27220420

RESUMEN

BACKGROUND: Oxidative stress plays an important and causal role in the mechanisms by which ischemia/reperfusion (I/R) injury increases brain damage after stroke. Accordingly, reducing oxidative stress has been proposed as a therapeutic strategy for limiting damage in the brain after stroke. Myeloperoxidase (MPO) is a highly potent oxidative enzyme that is capable of inducing both oxidative and nitrosative stress in vivo. METHODS: To determine if and the extent to which MPO-generated oxidants contribute to brain I/R injury, we treated mice subjected to middle cerebral artery occlusion (MCAO) with N-acetyl lysyltyrosylcysteine amide (KYC), a novel, specific and non-toxic inhibitor of MPO. Behavioral testing, ischemic damage, blood-brain-barrier disruption, apoptosis, neutrophils infiltration, microglia/macrophage activation, and MPO oxidation were analyzed within a 7-day period after MCAO. RESULTS: Our studies show that KYC treatment significantly reduces neurological severity scores, infarct size, IgG extravasation, neutrophil infiltration, loss of neurons, apoptosis, and microglia/macrophage activation in the brains of MCAO mice. Immunofluorescence studies show that KYC treatment reduces the formation of chlorotyrosine (ClTyr), a fingerprint biomarker of MPO oxidation, nitrotyrosine (NO2Tyr), and 4-hydroxynonenal (4HNE) in MCAO mice. All oxidative products colocalized with MPO in the infarcted brains, suggesting that MPO-generated oxidants are involved in forming the oxidative products. CONCLUSIONS: MPO-generated oxidants play detrimental roles in causing brain damage after stroke which is effectively reduced by KYC.


Asunto(s)
Lesiones Encefálicas , Infarto de la Arteria Cerebral Media/complicaciones , Fármacos Neuroprotectores/uso terapéutico , Oligopéptidos/uso terapéutico , Peroxidasa/metabolismo , Animales , Apoptosis/efectos de los fármacos , Barrera Hematoencefálica/efectos de los fármacos , Barrera Hematoencefálica/fisiología , Infarto Encefálico/tratamiento farmacológico , Infarto Encefálico/etiología , Lesiones Encefálicas/tratamiento farmacológico , Lesiones Encefálicas/etiología , Lesiones Encefálicas/metabolismo , Proteínas de Unión al Calcio/metabolismo , Modelos Animales de Enfermedad , Regulación de la Expresión Génica/efectos de los fármacos , Macrófagos/efectos de los fármacos , Macrófagos/patología , Ratones , Ratones Endogámicos C57BL , Proteínas de Microfilamentos/metabolismo , Microglía/efectos de los fármacos , Microglía/patología , Actividad Motora/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Infiltración Neutrófila/efectos de los fármacos , Infiltración Neutrófila/fisiología , Óxido Nítrico Sintasa de Tipo I/metabolismo , Oligopéptidos/farmacología , Oxidantes/metabolismo , Oxidantes/farmacología , Proteína p53 Supresora de Tumor/metabolismo
7.
Blood ; 124(26): 3978-81, 2014 Dec 18.
Artículo en Inglés | MEDLINE | ID: mdl-25339362

RESUMEN

High mobility group box 1 (HMGB1) is a chromatin-binding protein that maintains DNA structure. On cellular activation or injury, HMGB1 is released from activated immune cells or necrotic tissues and acts as a damage-associated molecular pattern to activate Toll-like receptor 4 (TLR4). Little is known concerning HMGB1 release and TLR4 activity and their role in the pathology of inflammation of sickle cell disease (SCD). Circulating HMGB1 levels were increased in both humans and mice with SCD compared with controls. Furthermore, sickle plasma increased HMGB1-dependent TLR4 activity compared with control plasma. HMGB1 levels were further increased during acute sickling events (vasoocclusive crises in humans or hypoxia/reoxygenation injury in mice). Anti-HMGB1 neutralizing antibodies reduced the majority of sickle plasma-induced TLR4 activity both in vitro and in vivo. These findings show that HMGB1 is the major TLR4 ligand in SCD and likely plays a critical role in SCD-mediated inflammation.


Asunto(s)
Anemia de Células Falciformes/metabolismo , Proteína HMGB1/metabolismo , Inflamación/metabolismo , Receptor Toll-Like 4/metabolismo , Anemia de Células Falciformes/inmunología , Animales , Regulación de la Expresión Génica , Humanos , Hipoxia/patología , Ligandos , Ratones , Ratones Endogámicos C57BL , Oxígeno/metabolismo , Transducción de Señal
8.
Am J Physiol Lung Cell Mol Physiol ; 309(9): L1009-17, 2015 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-26320159

RESUMEN

An increase in oxygen tension at birth is one of the key signals that initiate pulmonary vasodilation in the fetal lung. We investigated the hypothesis that targeting endothelial nitric oxide synthase (eNOS) to the mitochondrial outer membrane regulates reactive oxygen species (ROS) formation in the fetal pulmonary artery endothelial cells (PAEC) during this transition. We isolated PAEC and pulmonary arteries from 137-day gestation fetal lambs (term = 144 days). We exposed PAEC to a simulated transition from fetal to (3% O2) to normoxic (21%) or hyperoxic (95% O2) postnatal Po2 or to the nitric oxide synthase (NOS) agonist ATP. We assessed the effect of O2 and ATP on eNOS interactions with the mitochondrial outer membrane protein porin and with the chaperone hsp90. We also investigated the effect of decoy peptides that blocked eNOS interactions with porin or hsp90 on PAEC angiogenesis and vasodilator function of pulmonary arteries. Transition of fetal PAEC from 3 to 21% O2 but not to 95% O2 or exposure to ATP increased eNOS association with hsp90 and porin. Decoy peptides that blocked eNOS interactions decreased NO release, increased O2 consumption and mitochondrial ROS levels, and impaired PAEC angiogenesis. Decoy peptides also inhibited the relaxation responses of pulmonary artery rings and dilation of resistance size pulmonary arteries to ATP. The mitochondrial-antioxidant mito-ubiquinone restored the response to ATP in decoy peptide-treated pulmonary arteries. These data indicate that targeting eNOS to mitochondria decreases endothelial oxidative stress and facilitates vasodilation in fetal pulmonary circulation at birth.


Asunto(s)
Células Endoteliales/metabolismo , Feto/metabolismo , Mitocondrias/metabolismo , Óxido Nítrico Sintasa de Tipo III/metabolismo , Estrés Oxidativo/fisiología , Arteria Pulmonar/metabolismo , Animales , Células Cultivadas , Células Endoteliales/citología , Feto/citología , Proteínas HSP90 de Choque Térmico/metabolismo , Óxido Nítrico/metabolismo , Oxígeno/metabolismo , Arteria Pulmonar/citología , Ovinos , Vasodilatación/fisiología
9.
J Stroke Cerebrovasc Dis ; 24(12): 2759-73, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26433438

RESUMEN

BACKGROUND: Insidious cumulative brain injury from motor vehicle-induced whole-body vibration (MV-WBV) has not yet been studied. The objective of the present study is to validate whether whole-body vibration for long periods causes cumulative brain injury and impairment of the cerebral function. We also explored a preventive method for MV-WBV injury. METHODS: A study simulating whole-body vibration was conducted in 72 male Sprague-Dawley rats divided into 9 groups (N = 8): (1) 2-week normal control; (2) 2-week sham control (in the tube without vibration); (3) 2-week vibration (exposed to whole-body vibration at 30 Hz and .5 G acceleration for 4 hours/day, 5 days/week for 2 weeks; vibration parameters in the present study are similar to the most common driving conditions); (4) 4-week sham control; (5) 4-week vibration; (6) 4-week vibration with human apolipoprotein A-I molecule mimetic (4F)-preconditioning; (7) 8-week sham control; (8) 8-week vibration; and (9) 8-week 4F-preconditioning group. All the rats were evaluated by behavioral, physiological, and histological studies of the brain. RESULTS: Brain injury from vibration is a cumulative process starting with cerebral vasoconstriction, squeezing of the endothelial cells, increased free radicals, decreased nitric oxide, insufficient blood supply to the brain, and repeated reperfusion injury to brain neurons. In the 8-week vibration group, which indicated chronic brain edema, shrunken neuron numbers increased and whole neurons atrophied, which strongly correlated with neural functional impairment. There was no prominent brain neuronal injury in the 4F groups. CONCLUSIONS: The present study demonstrated cumulative brain injury from MV-WBV and validated the preventive effects of 4F preconditioning.


Asunto(s)
Lesiones Encefálicas/tratamiento farmacológico , Péptidos/uso terapéutico , Vibración , Accidentes de Tránsito , Animales , Lesiones Encefálicas/prevención & control , Masculino , Ratas , Ratas Sprague-Dawley
10.
Free Radic Biol Med ; 215: 112-126, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38336101

RESUMEN

Murine sickle cell disease (SCD) results in damage to multiple organs, likely mediated first by vasculopathy. While the mechanisms inducing vascular damage remain to be determined, nitric oxide bioavailability and sterile inflammation are both considered to play major roles in vasculopathy. Here, we investigate the effects of high mobility group box-1 (HMGB1), a pro-inflammatory damage-associated molecular pattern (DAMP) molecule on endothelial-dependent vasodilation and lung morphometrics, a structural index of damage in sickle (SS) mice. SS mice were treated with either phosphate-buffered saline (PBS), hE-HMGB1-BP, an hE dual-domain peptide that binds and removes HMGB1 from the circulation via the liver, 1-[4-(aminocarbonyl)-2-methylphenyl]-5-[4-(1H-imidazol-1-yl)phenyl]-1H-pyrrole-2-propanoic acid (N6022) or N-acetyl-lysyltyrosylcysteine amide (KYC) for three weeks. Human umbilical vein endothelial cells (HUVEC) were treated with recombinant HMGB1 (r-HMGB1), which increases S-nitrosoglutathione reductase (GSNOR) expression by ∼80%, demonstrating a direct effect of HMGB1 to increase GSNOR. Treatment of SS mice with hE-HMGB1-BP reduced plasma HMGB1 in SS mice to control levels and reduced GSNOR expression in facialis arteries isolated from SS mice by ∼20%. These changes were associated with improved endothelial-dependent vasodilation. Treatment of SS mice with N6022 also improved vasodilation in SS mice suggesting that targeting GSNOR also improves vasodilation. SCD decreased protein nitrosothiols (SNOs) and radial alveolar counts (RAC) and increased GSNOR expression and mean linear intercepts (MLI) in lungs from SS mice. The marked changes in pulmonary morphometrics and GSNOR expression throughout the lung parenchyma in SS mice were improved by treating with either hE-HMGB1-BP or KYC. These data demonstrate that murine SCD induces vasculopathy and chronic lung disease by an HMGB1- and GSNOR-dependent mechanism and suggest that HMGB1 and GSNOR might be effective therapeutic targets for reducing vasculopathy and chronic lung disease in humans with SCD.


Asunto(s)
Anemia de Células Falciformes , Benzamidas , Proteína HMGB1 , Enfermedades Pulmonares , Lesión Pulmonar , Pirroles , Enfermedades Vasculares , Humanos , Animales , Ratones , Lesión Pulmonar/etiología , Proteína HMGB1/genética , Células Endoteliales/metabolismo , Anemia de Células Falciformes/tratamiento farmacológico , Anemia de Células Falciformes/genética , Inflamación , Enfermedades Vasculares/etiología
11.
J Lipid Res ; 54(11): 3009-15, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23956444

RESUMEN

Activated leukocytes and polymorphonuclear neutrophils (PMN) release myeloperoxidase (MPO), which binds to endothelial cells (EC), is translocated, and generates oxidants that scavenge nitric oxide (NO) and impair EC function. To determine whether MPO impairs EC function in sickle cell disease (SCD), control (AA) and SCD mice were treated with N-acetyl-lysyltyrosylcysteine-amide (KYC). SCD humans and mice have high plasma MPO and soluble L-selectin (sL-selectin). KYC had no effect on MPO but decreased plasma sL-selectin and malondialdehyde in SCD mice. MPO and 3-chlorotyrosine (3-ClTyr) were increased in SCD aortas. KYC decreased MPO and 3-ClTyr in SCD aortas to the levels in AA aortas. Vasodilatation in SCD mice was impaired. KYC increased vasodilatation in SCD mice more than 2-fold, to ∼60% of levels in AA mice. KYC inhibited MPO-dependent 3-ClTyr formation in EC proteins. SCD mice had high plasma alanine transaminase (ALT), which tended to decrease in KYC-treated SCD mice (P = 0.07). KYC increased MPO and XO/XDH and decreased 3-ClTyr and 3-nitrotyrosine (3-NO2Tyr) in SCD livers. These data support the hypothesis that SCD increases release of MPO, which generates oxidants that impair EC function and injure livers. Inhibiting MPO is an effective strategy for decreasing oxidative stress and liver injury and restoring EC function in SCD.


Asunto(s)
Anemia de Células Falciformes/fisiopatología , Vasos Sanguíneos/efectos de los fármacos , Vasos Sanguíneos/metabolismo , Inhibidores Enzimáticos/farmacología , Estrés Oxidativo/efectos de los fármacos , Peroxidasa/antagonistas & inhibidores , Vasodilatación/efectos de los fármacos , Anemia de Células Falciformes/enzimología , Anemia de Células Falciformes/metabolismo , Animales , Vasos Sanguíneos/patología , Vasos Sanguíneos/fisiopatología , Relación Dosis-Respuesta a Droga , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Femenino , Humanos , Ácido Hipocloroso/metabolismo , Selectina L/química , Selectina L/metabolismo , Hígado/efectos de los fármacos , Hígado/metabolismo , Masculino , Ratones , Oligopéptidos/farmacología , Peroxidasa/sangre , Solubilidad
12.
J Lipid Res ; 54(11): 3016-29, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23883583

RESUMEN

Myeloperoxidase (MPO) plays important roles in disease by increasing oxidative and nitrosative stress and oxidizing lipoproteins. Here we report N-acetyl lysyltyrosylcysteine amide (KYC) is an effective inhibitor of MPO activity. We show KYC inhibits MPO-mediated hypochlorous acid (HOCl) formation and nitration/oxidation of LDL. Disulfide is the major product of MPO-mediated KYC oxidation. KYC (≤4,000 µM) does not induce cytotoxicity in bovine aortic endothelial cells (BAECs). KYC inhibits HOCl generation by phorbol myristate acetate (PMA)-stimulated neutrophils and human promyelocytic leukemia (HL-60) cells but not superoxide generation by PMA-stimulated HL-60 cells. KYC inhibits MPO-mediated HOCl formation in BAEC culture and protects BAECs from MPO-induced injury. KYC inhibits MPO-mediated lipid peroxidation of LDL whereas tyrosine (Tyr) and tryptophan (Trp) enhance oxidation. KYC is unique as its isomers do not inhibit MPO activity, or are much less effective. Ultraviolet-visible spectral studies indicate KYC binds to the active site of MPO and reacts with compounds I and II. Docking studies show the Tyr of KYC rests just above the heme of MPO. Interestingly, KYC increases MPO-dependent H2O2 consumption. These data indicate KYC is a novel and specific inhibitor of MPO activity that is nontoxic to endothelial cell cultures. Accordingly, KYC may be useful for treating MPO-mediated vascular disease.


Asunto(s)
Oligopéptidos/farmacología , Peroxidasa/antagonistas & inhibidores , Animales , Aorta/citología , Biocatálisis , Bovinos , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Células HL-60 , Halogenación/efectos de los fármacos , Humanos , Ácido Hipocloroso/metabolismo , Peroxidación de Lípido/efectos de los fármacos , Neutrófilos/enzimología , Nitratos/metabolismo , Oligopéptidos/metabolismo , Oligopéptidos/toxicidad , Oxidación-Reducción , Peroxidasa/metabolismo
13.
Am J Physiol Heart Circ Physiol ; 304(2): H328-36, 2013 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-23125208

RESUMEN

Hemolysis can saturate the hemoglobin (Hb)/heme scavenging system, resulting in increased circulating cell-free Hb (CF-Hb) in hereditary and acquired hemolytic disease. While recent studies have suggested a central role for intravascular hemolysis and CF-Hb in the development of vascular dysfunction, this concept has stimulated considerable debate. This highlights the importance of determining the contribution of CF-Hb to vascular complications associated with hemolysis. Therefore, a novel Hb-binding peptide was synthesized and linked to a small fragment of apolipoprotein E (amino acids 141-150) to facilitate endocytic clearance. Plasma clearance of hE-Hb-b10 displayed a rapid phase t(1/2) of 16 min and slow phase t(1/2) of 10 h, trafficking primarily through the liver. Peptide hE-Hb-B10 decreased CF-Hb in mice treated with phenylhydrazine, a model of acute hemolysis. Administration of hE-Hb-B10 also attenuated CF-Hb in two models of chronic hemolysis: Berkeley sickle cell disease (SS) mice and mice with severe hereditary spherocytosis (HS). The hemolytic rate was unaltered in either chronic hemolysis model, supporting the conclusion that hE-Hb-B10 promotes CF-Hb clearance without affecting erythrocyte lysis. Interestingly, hE-Hb-B10 also decreased plasma ALT activity in SS and HS mice. Although acetylcholine-mediated facialis artery vasodilation was not improved by hE-Hb-B10 treatment, the peptide shifted vascular response in favor of NO-dependent vasodilation in SS mice. Taken together, these data demonstrate that hE-Hb-B10 decreases CF-Hb with a concomitant reduction in liver injury and changes in vascular response. Therefore, hE-Hb-B10 can be used to investigate the different roles of CF-Hb in hemolytic pathology and may have therapeutic benefit in the treatment of CF-Hb-mediated tissue damage.


Asunto(s)
Anemia Hemolítica/tratamiento farmacológico , Apolipoproteínas E/farmacología , Endocitosis/efectos de los fármacos , Hemoglobinas/metabolismo , Hemólisis , Hígado/efectos de los fármacos , Enfermedad Aguda , Anemia Hemolítica/sangre , Anemia Hemolítica/etiología , Anemia Hemolítica/fisiopatología , Anemia de Células Falciformes/sangre , Anemia de Células Falciformes/complicaciones , Anemia de Células Falciformes/tratamiento farmacológico , Animales , Apolipoproteínas E/sangre , Apolipoproteínas E/farmacocinética , Enfermedad Crónica , Modelos Animales de Enfermedad , Semivida , Humanos , Hígado/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Óxido Nítrico/metabolismo , Fragmentos de Péptidos/sangre , Fragmentos de Péptidos/farmacología , Péptidos/sangre , Péptidos/farmacología , Fenilhidrazinas , Unión Proteica , Transporte de Proteínas , Esferocitosis Hereditaria/sangre , Esferocitosis Hereditaria/complicaciones , Esferocitosis Hereditaria/tratamiento farmacológico , Vasodilatación/efectos de los fármacos , Vasodilatadores/farmacología
14.
J Surg Res ; 180(1): 21-6, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23158403

RESUMEN

BACKGROUND: Necrotizing enterocolitis (NEC) is the most common surgical emergency in neonates, with an incidence of 0.5-2.4 cases per 1000 live births and a mortality rate between 10% and 50%. Neonates affected by NEC develop a septic injury that is associated with increased risk of neurological impairment due to intraventricular bleeding and chronic lung disease. Intestinal alkaline phosphatase (IAP) is an endogenous protein that has been shown to inactivate the endotoxin lipopolysaccharide (LPS), and has recently been used successfully as an adjunct to treat sepsis in adult patients. We tested the hypothesis that systemic, exogenous IAP will mitigate the inflammatory response as measured by serum levels of proinflammatory cytokines in a rat model of NEC. METHODS: Newborn Sprague-Dawley rats were divided into groups. Control pups were dam fed. NEC was induced by feeding formula containing LPS and exposure to intermittent hypoxia. NEC pups were given intraperitoneal injections of 4 or 40 glycine units (U) of IAP or placebo twice daily. Intestine and serum was collected for cytokine analysis as well as measurement of alkaline phosphatase activity. RESULTS: Systemic IAP administration significantly increased serum alkaline phosphatase activity in a dose- and time-dependent fashion. The proinflammatory cytokines tumor necrosis factor α, interleukin 6, and interleukin 1ß were significantly increased in NEC rats versus controls on days 2 and 3. Importantly, treatment with 40 U systemic IAP decreased these proinflammatory cytokines back to near-control levels. CONCLUSIONS: Systemic IAP administration appears effective in mitigating the systemic inflammatory response associated with NEC, and may prove to be a valuable adjunctive treatment for NEC.


Asunto(s)
Fosfatasa Alcalina/uso terapéutico , Enterocolitis Necrotizante/tratamiento farmacológico , Intestinos/enzimología , Síndrome de Respuesta Inflamatoria Sistémica/prevención & control , Fosfatasa Alcalina/sangre , Animales , Animales Recién Nacidos , Citocinas/sangre , Enterocolitis Necrotizante/inmunología , Ratas , Ratas Sprague-Dawley
15.
Am J Respir Cell Mol Biol ; 46(3): 389-96, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22033263

RESUMEN

Experimental asthma increases eosinophil and collagen deposition in the lungs of sickle cell disease (SCD) mice to a greater extent than in control mice. However, the effects of asthma on inflammation and airway physiology remain unclear. To determine effects of asthma on pulmonary inflammation and airway mechanics in SCD mice, hematopoietic stem cell transplantation was used to generate chimeric SCD and hemoglobin A mice. Experimental asthma was induced by sensitizing mice with ovalbumin (OVA). Airway mechanics were assessed using forced oscillation techniques. Mouse lungs were examined histologically and physiologically. Cytokine, chemokine, and growth factors in bronchoalveolar lavage fluid were determined by multiplex. IgE was quantified by ELISA. LDH was quantified using a colorimetric enzymatic assay. At baseline (nonsensitized), chimeric SCD mice developed hemolytic anemia with sickled red blood cells, mild leukocytosis, and increased vascular endothelial growth factor and IL-13 compared with chimeric hemoglobin A mice. Experimental asthma increased perialveolar eosinophils, plasma IgE, and bronchoalveolar lavage fluid IL-1ß, IL-4, IL-6, and monocyte chemotactic protein 1 in chimeric hemoglobin A and SCD mice. IFN-γ levels were reduced in both groups. IL-5 was preferentially increased in chimeric SCD mice but not in hemoglobin A mice. Positive end-expiratory pressures and methacholine studies revealed that chimeric SCD mice had greater resistance in large and small airways compared with hemoglobin A mice at baseline and after OVA sensitization. SCD alone induces a baseline lung pathology that increases large and small airway resistance and primes the lungs to increased inflammation and airway hyperresponsiveness after OVA sensitization.


Asunto(s)
Resistencia de las Vías Respiratorias , Anemia de Células Falciformes/complicaciones , Asma/complicaciones , Hiperreactividad Bronquial/etiología , Pulmón/fisiopatología , Neumonía/etiología , Anemia de Células Falciformes/sangre , Anemia de Células Falciformes/genética , Anemia de Células Falciformes/fisiopatología , Animales , Asma/inmunología , Asma/fisiopatología , Hiperreactividad Bronquial/sangre , Hiperreactividad Bronquial/genética , Hiperreactividad Bronquial/inmunología , Hiperreactividad Bronquial/fisiopatología , Pruebas de Provocación Bronquial , Líquido del Lavado Bronquioalveolar/química , Líquido del Lavado Bronquioalveolar/inmunología , Broncoconstrictores , Colorimetría , Citocinas/metabolismo , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática , Eosinófilos/inmunología , Hemoglobina A/genética , Hemoglobina A/metabolismo , Hemoglobina Falciforme/genética , Hemoglobina Falciforme/metabolismo , Humanos , Inmunoglobulina E/sangre , Mediadores de Inflamación/metabolismo , L-Lactato Deshidrogenasa/metabolismo , Pulmón/inmunología , Pulmón/patología , Cloruro de Metacolina , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Ovalbúmina , Neumonía/sangre , Neumonía/genética , Neumonía/inmunología , Neumonía/fisiopatología , Respiración con Presión Positiva , Factor A de Crecimiento Endotelial Vascular/metabolismo
16.
J Surg Res ; 177(2): 228-34, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22687880

RESUMEN

BACKGROUND: Supplementation of intestinal alkaline phosphatase (IAP), an endogenous protein expressed in the intestines, decreases the severity of necrotizing enterocolitis (NEC)-associated intestinal injury and permeability. We hypothesized that IAP administration is protective in a dose-dependent manner of the inflammatory response in a neonatal rat model. MATERIALS AND METHODS: Pre- and full-term newborn Sprague-Dawley rat pups were sacrificed on day of life 3. Control pups were vaginally delivered and dam fed. Preterm pups were delivered via cesarean section and exposed to intermittent hypoxia and formula feeds containing lipopolysaccharide (NEC) with and without IAP. Three different standardized doses were administered to a group of pups treated with 40, 4, and 0.4U/kg of bovine IAP (NEC+IAP40, IAP4, or IAP0.4U). Reverse transcription-real-time polymerase chain reaction (RT-PCR) for inducible nitric oxide synthase (iNOS) and tumor necrosis factor (TNF)-α on liver and lung tissues and serum cytokine analysis for interleukin (IL)-1ß, IL-6, IL-10, and TNF-α were performed. Data were analyzed by Kruskal-Wallis and Mann-Whitney tests, expressed as mean±standard error of the mean and P≤0.05 considered significant. RESULTS: Levels of cytokines IL-1ß, IL-6, and TNF-α increased significantly in NEC versus control, returning to control levels with increasing doses of supplemental enteral IAP. Hepatic and pulmonary TNF-α and iNOS messenger ribonucleic acid expressions increased in NEC, and the remaining elevated despite IAP supplementation. CONCLUSIONS: Proinflammatory cytokine expression is increased systemically with intestinal NEC injury. Administration of IAP significantly reduces systemic proinflammatory cytokine expression in a dose-dependent manner. Early supplemental enteral IAP may reduce NEC-related injury and be useful for reducing effects caused by a proinflammatory cascade.


Asunto(s)
Fosfatasa Alcalina/uso terapéutico , Citocinas/sangre , Enterocolitis Necrotizante/prevención & control , Animales , Animales Recién Nacidos , Evaluación Preclínica de Medicamentos , Enterocolitis Necrotizante/sangre , Enterocolitis Necrotizante/complicaciones , Femenino , Hepatitis/etiología , Hepatitis/prevención & control , Neumonía/etiología , Neumonía/prevención & control , Embarazo , Ratas , Ratas Sprague-Dawley
17.
PLoS One ; 17(8): e0269564, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36018859

RESUMEN

Myeloperoxidase (MPO), oxidative stress (OS), and endoplasmic reticulum (ER) stress are increased in the lungs of rat pups raised in hyperoxia, an established model of bronchopulmonary dysplasia (BPD). However, the relationship between OS, MPO, and ER stress has not been examined in hyperoxia rat pups. We treated Sprague-Dawley rat pups with tunicamycin or hyperoxia to determine this relationship. ER stress was detected using immunofluorescence, transcriptomic, proteomic, and electron microscopic analyses. Immunofluorescence observed increased ER stress in the lungs of hyperoxic rat BPD and human BPD. Proteomic and morphometric studies showed that tunicamycin directly increased ER stress of rat lungs and decreased lung complexity with a BPD phenotype. Previously, we showed that hyperoxia initiates a cycle of destruction that we hypothesized starts from increasing OS through MPO accumulation and then increases ER stress to cause BPD. To inhibit ER stress, we used tauroursodeoxycholic acid (TUDCA), a molecular chaperone. To break the cycle of destruction and reduce OS and MPO, we used N-acetyl-lysyltyrosylcysteine amide (KYC). The fact that TUDCA improved lung complexity in tunicamycin- and hyperoxia-treated rat pups supports the idea that ER stress plays a causal role in BPD. Additional support comes from data showing TUDCA decreased lung myeloid cells and MPO levels in the lungs of tunicamycin- and hyperoxia-treated rat pups. These data link OS and MPO to ER stress in the mechanisms mediating BPD. KYC's inhibition of ER stress in the tunicamycin-treated rat pup's lung provides additional support for the idea that MPO-induced ER stress plays a causal role in the BPD phenotype. ER stress appears to expand our proposed cycle of destruction. Our results suggest ER stress evolves from OS and MPO to increase neonatal lung injury and impair growth and development. The encouraging effect of TUDCA indicates that this compound has the potential for treating BPD.


Asunto(s)
Displasia Broncopulmonar , Hiperoxia , Neumonía , Animales , Animales Recién Nacidos , Modelos Animales de Enfermedad , Estrés del Retículo Endoplásmico , Humanos , Recién Nacido , Pulmón , Proteómica , Ratas , Ratas Sprague-Dawley , Tunicamicina
18.
Am J Physiol Cell Physiol ; 300(3): C550-6, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21160034

RESUMEN

Systemic sclerosis (SSc) is an autoimmune connective tissue disorder characterized by oxidative stress, impaired vascular function, and attenuated angiogenesis. The tight-skin (Tsk(-/+)) mouse is a model of SSc that displays many of the cellular features of the clinical disease. We tested the hypotheses that abnormal fibrillin-1 expression and chronic phospholipid oxidation occur in Tsk(-/+) mice and, furthermore, that these factors precipitate a prooxidant state, collagen-related protein expression, apoptosis, and mesenchymal transition in endothelial cells cultured on Tsk(-/+) extracellular matrix. Human umbilical vein endothelial cells were seeded on microfibrils isolated from skin of C57BL/6J (control) and Tsk(-/+) mice in the presence or absence of chronic pretreatment with the apolipoprotein Apo A-I mimetic D-4F (1 mg·kg(-1)·day(-1) ip for 6 to 8 wk). Nitric oxide-to-superoxide anion ratio was assessed 12 h after culture, and cell proliferation, apoptosis, and phenotype were studied 72 h after culture. Tsk(-/+) mice demonstrated abnormal "big fibrillin" expression (405 kDa) by Western blot analysis compared with control. Endothelial cells cultured on microfibrils prepared from Tsk(-/+) mice demonstrated reduced proliferation, a prooxidant state (reduced nitric oxide-to-superoxide anion ratio), increased apoptosis, and collagen-related protein expression associated with mesenchymal transition. Chronic D-4F pretreatment of Tsk(-/+) mice attenuated many of these adverse effects. The findings demonstrate that abnormal fibrillin-1 expression and chronic oxidative stress mediate endothelial mesenchymal transition in Tsk(-/+) mice. This mesenchymal transition may contribute to the reduction in angiogenesis that is known to occur in this model of SSc.


Asunto(s)
Células Endoteliales/metabolismo , Mesodermo/metabolismo , Proteínas de Microfilamentos/biosíntesis , Proteínas de Microfilamentos/genética , Estrés Oxidativo , Esclerodermia Sistémica/metabolismo , Animales , Proliferación Celular , Células Cultivadas , Enfermedad Crónica , Modelos Animales de Enfermedad , Células Endoteliales/patología , Femenino , Fibrilina-1 , Fibrilinas , Humanos , Masculino , Mesodermo/patología , Ratones , Ratones Endogámicos C57BL , Proteínas de Microfilamentos/fisiología , Peso Molecular , Neovascularización Fisiológica/genética , Estrés Oxidativo/genética , Esclerodermia Sistémica/genética , Esclerodermia Sistémica/patología
19.
Am J Physiol Lung Cell Mol Physiol ; 301(3): L334-45, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21622842

RESUMEN

Persistent pulmonary hypertension of the newborn (PPHN) is associated with decreased blood vessel density that contributes to increased pulmonary vascular resistance. Previous studies showed that uncoupled endothelial nitric oxide (NO) synthase (eNOS) activity and increased NADPH oxidase activity resulted in marked decreases in NO bioavailability and impaired angiogenesis in PPHN. In the present study, we hypothesize that loss of tetrahydrobiopterin (BH4), a critical cofactor for eNOS, induces uncoupled eNOS activity and impairs angiogenesis in PPHN. Pulmonary artery endothelial cells (PAEC) isolated from fetal lambs with PPHN (HTFL-PAEC) or control lambs (NFL-PAEC) were used to investigate the cellular mechanisms impairing angiogenesis in PPHN. Cellular mechanisms were examined with respect to BH4 levels, GTP-cyclohydrolase-1 (GCH-1) expression, eNOS dimer formation, and eNOS-heat shock protein 90 (hsp90) interactions under basal conditions and after sepiapterin (Sep) supplementation. Cellular levels of BH4, GCH-1 expression, and eNOS dimer formation were decreased in HTFL-PAEC compared with NFL-PAEC. Sep supplementation decreased apoptosis and increased in vitro angiogenesis in HTFL-PAEC and ex vivo pulmonary artery sprouting angiogenesis. Sep also increased cellular BH4 content, NO production, eNOS dimer formation, and eNOS-hsp90 association and decreased the superoxide formation in HTFL-PAEC. These data demonstrate that Sep improves NO production and angiogenic potential of HTFL-PAEC by recoupling eNOS activity. Increasing BH4 levels via Sep supplementation may be an important therapy for improving eNOS function and restoring angiogenesis in PPHN.


Asunto(s)
Óxido Nítrico Sintasa de Tipo III/metabolismo , Pterinas/farmacología , Animales , Apoptosis/efectos de los fármacos , Biopterinas/análogos & derivados , Biopterinas/metabolismo , Movimiento Celular/efectos de los fármacos , Endotelio Vascular/metabolismo , GTP Ciclohidrolasa/biosíntesis , Proteínas HSP90 de Choque Térmico/metabolismo , Humanos , Recién Nacido , Neovascularización Fisiológica/efectos de los fármacos , Óxido Nítrico Sintasa de Tipo III/efectos de los fármacos , Síndrome de Circulación Fetal Persistente , Multimerización de Proteína/efectos de los fármacos , Oveja Doméstica
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA