Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 96
Filtrar
1.
Pediatr Res ; 2024 Sep 19.
Artigo em Inglês | MEDLINE | ID: mdl-39300277

RESUMO

BACKGROUND: Intermittent hypoxia (IH) and oxidative stress play key roles in gut dysbiosis and inflammation. We tested the hypothesis that increasing numbers of daily IH episodes cause microbiome dysbiosis and severe gut injury. METHODS: Neonatal rats were exposed to hyperoxia (Hx), growth restriction, and IH. For IH, pups were exposed to 2-12 daily episodes from birth (P0) to postnatal day 7 (7D) or P0-P14 (14D), with or without recovery in room air (RA) until P21. Animals raised in RA from P0 to P21 served as normoxia controls. Stool was expressed from the large intestines for microbiome analysis, and tissue samples were assessed for histopathology and biomarkers of inflammation. RESULTS: Hx and IH caused a significant reduction in the number and diversity of organisms. The severity of gut injury and levels of inflammatory cytokines and TLR4 increased, while total glutathione (tGSH) declined, with increasing daily IH episodes. The number of organisms correlated with the villi number (p < 0.05) and tGSH depletion (p < 0.001). CONCLUSIONS: The critical number of daily IH episodes that the newborn gut may sustain is 6, beyond which irreversible damage occurs. The immature gut is highly susceptible to IH-induced injury, and IH may contribute to pathological outcomes in the immature gut. IMPACT STATEMENT: 1. The neonatal gut at birth is highly susceptible to intermittent hypoxia (IH) injury. 2. IH causes gut dysbiosis, inflammation, and glutathione depletion. 3. The severity of gut injury worsens as a function of increasing daily IH episodes. 4. The critical number of daily IH episodes that the newborn gut may sustain is 6, beyond which irreversible damage occurs.

2.
Pediatr Res ; 92(1): 109-117, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-34455420

RESUMO

BACKGROUND: Preterm infants frequently experience intermittent hypoxia (IH) episodes, rendering them susceptible to oxidative stress and gut dysbiosis. We tested the hypothesis that early supplementation with antioxidants and/or fish oil promotes gut biodiversity and mitigates IH-induced gut injury. METHODS: Newborn rats were exposed to neonatal IH from birth (P0) to P14 during which they received daily oral supplementation with: (1) coenzyme Q10 (CoQ10) in olive oil, (2) fish oil, (3) glutathione nanoparticles (nGSH), (4) CoQ10 + fish oil, or (5) olive oil (placebo control). Pups were placed in room air (RA) from P14 to P21 with no further treatment. RA controls were similarly treated. Stool samples were assessed for microbiota and terminal ileum for histopathology and morphometry, total antioxidant capacity, lipid peroxidation, and biomarkers of gut injury. RESULTS: Neonatal IH induced histopathologic changes consistent with necrotizing enterocolitis, which were associated with increased lipid peroxidation, toll-like receptor, transforming growth factor, and nuclear factor kappa B. Combination of CoQ10 + fish oil and nGSH were most effective for preserving gut integrity, reducing biomarkers of gut injury, and increasing commensal organisms. CONCLUSIONS: Combination of antioxidants and fish oil may confer synergistic benefits to mitigate IH-induced injury in the terminal ileum. IMPACT: Antioxidant and fish oil (PUFA) co-treatment was most beneficial for reducing neonatal IH-induced gut injury. The synergistic effects of antioxidant and fish oil is likely due to prevention of IH-induced ROS attack on lipids, thus preserving and augmenting its therapeutic benefits. Combination treatment was also effective for increasing the abundance of the non-pathogenic Firmicutes phylum, which is associated with a healthy gastrointestinal system of the newborn. Extremely low gestational age neonates who are at high risk for frequent, repetitive neonatal IH and oxidative stress-induced diseases may benefit from this combination therapy.


Assuntos
Asfixia Neonatal , Microbioma Gastrointestinal , Animais , Animais Recém-Nascidos , Antioxidantes/farmacologia , Biomarcadores , Suplementos Nutricionais , Óleos de Peixe/farmacologia , Humanos , Hipóxia/metabolismo , Recém-Nascido , Recém-Nascido Prematuro , Azeite de Oliva , Ratos
3.
Respir Res ; 22(1): 196, 2021 Jul 05.
Artigo em Inglês | MEDLINE | ID: mdl-34225702

RESUMO

BACKGROUND: Neonatal intermittent hypoxia (IH) results in oxidative distress in preterm infants with immature antioxidant systems, contributing to lung injury. Coenzyme Q10 (CoQ10) and fish oil protect against oxidative injury. We tested the hypothesis that CoQ10 is more effective than fish oil for prevention of IH-induced lung injury in neonatal rats. METHODS: Newborn rats were exposed to two clinically relevant IH paradigms at birth (P0): (1) 50% O2 with brief hypoxia (12% O2); or (2) room air (RA) with brief hypoxia (12% O2), until P14 during which they were supplemented with daily oral CoQ10, fish oil, or olive oil from P0 to P14. Pups were studied at P14 or placed in RA until P21 with no further treatment. Lungs were assessed for histopathology and morphometry; biomarkers of oxidative stress and lipid peroxidation; and antioxidants. RESULTS: Of the two neonatal IH paradigms 21%/12% O2 IH resulted in the most severe outcomes, evidenced by histopathology and morphometry. CoQ10 was effective for preserving lung architecture and reduction of IH-induced oxidative stress biomarkers. In contrast, fish oil resulted in significant adverse outcomes including oversimplified alveoli, hemorrhage, reduced secondary crest formation and thickened septae. This was associated with elevated oxidants and antioxidants activities. CONCLUSIONS: Data suggest that higher FiO2 may be needed between IH episodes to curtail the damaging effects of IH, and to provide the lungs with necessary respite. The negative outcomes with fish oil supplementation suggest oxidative stress-induced lipid peroxidation.


Assuntos
Óleos de Peixe/uso terapêutico , Hipóxia/tratamento farmacológico , Pulmão/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Ubiquinona/análogos & derivados , Animais , Animais Recém-Nascidos , Feminino , Óleos de Peixe/farmacologia , Hipóxia/metabolismo , Hipóxia/patologia , Pulmão/metabolismo , Pulmão/patologia , Masculino , Estresse Oxidativo/fisiologia , Gravidez , Ratos , Ratos Sprague-Dawley , Ubiquinona/farmacologia , Ubiquinona/uso terapêutico
4.
Prostaglandins Other Lipid Mediat ; 155: 106565, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34051366

RESUMO

Intermittent hypoxia (IH) is associated with the pathogenesis of necrotizing enterocolitis (NEC). We tested the hypothesis that early supplementation with antioxidants and/or fish oil protects the terminal ileum from oxidative injury induced by neonatal IH. Newborn rats were exposed to neonatal IH from birth (P0) until P14 during which they received daily fish oil, coenzyme Q10 (CoQ10), glutathione nanoparticles (nGSH), fish oil + CoQ10, or olive oil. Pups were then placed in room air from P14 to P21 with no further supplementation. Terminal ileum was assessed for IH-induced injury and inflammatory biomarkers. Neonatal IH induced severe damage consistent with NEC, and was associated with oxidative stress and elevations in PGE2, PGF2α, TxB2, NOS-2 and TLR-4, effects that were ameliorated with nGSH and combination CoQ10+fish oil. Early postnatal supplementation with antioxidants and/or fish oil during neonatal IH may be favorable for preserving gut integrity and reducing oxidative injury.


Assuntos
Óleos de Peixe
5.
Exp Lung Res ; 47(3): 121-135, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33377400

RESUMO

PURPOSE/AIM: Intravitreal bevacizumab (Avastin) is an irreversible vascular endothelial growth factor (VEGF) inhibitor used to treat severe retinopathy of prematurity (ROP) in extremely low gestational age neonates (ELGANs). ELGANs who are at the highest risk for developing severe ROP often experience brief intermittent hypoxia (IH) episodes which may cause oxidative damage. We tested the hypothesis that intravitreal Avastin leaks into the systemic circulation during exposure to IH and has adverse effects on biomarkers of pulmonary microvascular maturation, thus leading to pulmonary hemorrhage and long-term pulmonary sequelae. METHODS: Neonatal rats at postnatal day (PN) 0 (birth) were exposed to either: 1) hyperoxia (50% O2) or 2) neonatal IH (50% O2 with brief episodes of 12% O2) from PN0 to PN14. Room air (RA) littermates served as controls. At PN14, the time of eye opening in rats, a single dose of Avastin (0.125 mg in 5 µL) was injected into the vitreous cavity of the left eyes. A control group received equivalent volume saline. At PN23 and PN45, blood gases, lung-to-body weight ratios, histology, immunofluorescence, and lung biomarkers of angiogenesis were examined. RESULTS: At PN23, Avastin increased lung VEGF, nitric oxide derivatives (NOx), and hypoxia-inducible factor (HIF)1a in the hyperoxia-exposed groups, but decreased soluble VEGFR-1 (sVEGFR-1). At PN45, lungs from animals exposed to neonatal IH and treated with Avastin were severely hemorrhagic with morphologic changes in lung architecture consistent with chronic lung disease. This was associated with higher VEGF and NOx levels, and lower insulin-like growth factor (IGF)-I and sVEGFR-1. CONCLUSIONS: These findings prove our hypothesis that intravitreal Avastin penetrates the blood-ocular barrier in IH and alters lung biomarkers of angiogenesis. Avastin targeting of VEGF could affect normal lung development which may be exaggerated under pathologic conditions such as IH, ultimately leading to vascular permeability, vessel rupture, and pulmonary hemorrhage.


Assuntos
Bevacizumab/uso terapêutico , Hipóxia , Pulmão , Neovascularização Patológica/tratamento farmacológico , Animais , Animais Recém-Nascidos , Biomarcadores , Pulmão/efeitos dos fármacos , Pulmão/patologia , Ratos , Ratos Sprague-Dawley , Retinopatia da Prematuridade , Fator A de Crescimento do Endotélio Vascular
6.
BMC Nephrol ; 22(1): 299, 2021 09 04.
Artigo em Inglês | MEDLINE | ID: mdl-34481475

RESUMO

BACKGROUND: We tested the hypotheses that: 1) early exposure to increasing episodes of clinically relevant intermittent hypoxia (IH) is detrimental to the developing kidneys; and 2) there is a critical number of daily IH episodes which will result in irreparable renal damage that may involve angiotensin (Ang) II and endothelin (ET)-1. METHODS: At birth (P0), neonatal rat pups were exposed to brief IH episodes from the first day of life (P0) to P7 or from P0-P14. Pups were either euthanized immediately or placed in room air (RA) until P21. RA littermates served as controls. Kidneys were harvested at P7, P14, and P21 for histopathology; angiotensin converting enzyme (ACE), ACE-2, ET-1, big ET-1, and malondialdehyde (MDA) levels; immunoreactivity of ACE, ACE-2, ET-1, ET-2, ET receptors (ETAR, ETBR), and hypoxia inducible factor (HIF)1α; and apoptosis (TUNEL stain). RESULTS: Histopathology showed increased renal damage with 8-12 IH episodes/day, and was associated with Ang II, ACE, HIF1α, and apoptosis. ACE-2 was not expressed at P7, and minimally increased at P14. However, a robust ACE-2 response was seen during recovery with maximum levels noted in the groups recovering from 8 IH episodes/day. ET-1, big ET-1, ETAR, ETBR, and MDA increased with increasing levels of neonatal IH. CONCLUSIONS: Chronic neonatal IH causes severe damage to the developing kidney with associated elevations in vasoconstrictors, suggesting hypertension, particularly with 8 neonatal IH episodes. ACE-2 is not activated in early postnatal life, and this may contribute to IH-induced vasoconstriction. Therapeutic targeting of ACE and ET-1 may help decrease the risk for kidney injury in the developing neonate to prevent and/or treat neonatal acute kidney injury and/or chronic kidney disease.


Assuntos
Injúria Renal Aguda/etiologia , Biomarcadores/urina , Hipóxia/complicações , Rim/patologia , Injúria Renal Aguda/patologia , Injúria Renal Aguda/urina , Enzima de Conversão de Angiotensina 2/urina , Animais , Animais Recém-Nascidos , Apoptose , Estresse Oxidativo , Peptidil Dipeptidase A/urina , Ratos , Ratos Sprague-Dawley
7.
Int J Mol Sci ; 22(7)2021 Mar 27.
Artigo em Inglês | MEDLINE | ID: mdl-33801707

RESUMO

(1) Background: Caffeine citrate, at standard doses, is effective for reducing the incidence of apnea of prematurity (AOP) and may confer neuroprotection and decrease neonatal morbidities in extremely low gestational age neonates (ELGANs) requiring oxygen therapy. We tested the hypothesis that high-dose caffeine (HiC) has no adverse effects on the neonatal brain. (2) Methods: Newborn rat pups were randomized to room air (RA), hyperoxia (Hx) or neonatal intermittent hypoxia (IH), from birth (P0) to P14 during which they received intraperitoneal injections of LoC (20 mg/kg on P0; 5 mg/kg/day on P1-P14), HiC (80 mg/kg; 20 mg/kg), or equivalent volume saline. Blood gases, histopathology, myelin and neuronal integrity, and adenosine receptor reactivity were assessed. (3) Results: Caffeine treatment in Hx influenced blood gases more than treatment in neonatal IH. Exposure to neonatal IH resulted in hemorrhage and higher brain width, particularly in layer 2 of the cerebral cortex. Both caffeine doses increased brain width in RA, but layer 2 was increased only with HiC. HiC decreased oxidative stress more effectively than LoC, and both doses reduced apoptosis biomarkers. In RA, both caffeine doses improved myelination, but the effect was abolished in Hx and neonatal IH. Similarly, both doses inhibited adenosine 1A receptor in all oxygen environments, but adenosine 2A receptor was inhibited only in RA and Hx. (4) Conclusions: Caffeine, even at high doses, when administered in normoxia, can confer neuroprotection, evidenced by reductions in oxidative stress, hypermyelination, and increased Golgi bodies. However, varying oxygen environments, such as Hx or neonatal IH, may alter and modify pharmacodynamic actions of caffeine and may even override the benefits caffeine.


Assuntos
Encéfalo/efeitos dos fármacos , Encéfalo/crescimento & desenvolvimento , Cafeína/farmacologia , Hipóxia/tratamento farmacológico , Animais , Animais Recém-Nascidos , Apoptose , Biomarcadores/metabolismo , Gasometria , Cafeína/administração & dosagem , Córtex Cerebral/efeitos dos fármacos , Esquema de Medicação , Feminino , Hemorragia/tratamento farmacológico , Hiperóxia , Masculino , Estresse Oxidativo , Oxigênio/química , Ratos , Ratos Sprague-Dawley , Receptores Purinérgicos P1/metabolismo
8.
Int J Mol Sci ; 21(3)2020 Feb 02.
Artigo em Inglês | MEDLINE | ID: mdl-32024231

RESUMO

Aquaporins (AQPs) are involved in hypoxia-induced angiogenesis and retinal damage. Bumetanide is a diuretic agent, Na+/K+/Cl- cotransporter (NKCC1), and AQP 1-4 inhibitor. We tested the hypothesis that early postnatal treatment with bumetanide suppresses biomarkers of angiogenesis and decreases severe retinopathy oxygen-induced retinopathy (OIR). Neonatal rats were exposed at birth (P0) to either (1) room air (RA); (2) hyperoxia (50% O2); or (3) intermittent hypoxia (IH) consisting of 50% O2 with brief, clustered episodes of 12% O2 from P0 to postnatal day 14 (P14), during which they were treated intraperitoneally (IP) with bumetanide (0.1 mg/kg/day) or an equivalent volume of saline, on P0-P2. Pups were examined at P14 or allowed to recover in RA from P14-P21. Retinal angiogenesis, morphometry, pathology, AQPs, and angiogenesis biomarkers were determined at P14 and P21. Bumetanide reduced vascular abnormalities associated with severe OIR. This was associated with reductions in AQP-4 and VEGF. Bumetanide suppressed sVEGFR-1 in the serum and vitreous fluid, but levels were increased in the ocular tissues during recovery. Similar responses were noted for IGF-I. In this model, early systemic bumetanide administration reduces severe OIR, the benefits of which appear to be mediated via suppression of AQP-4 and VEGF. Further studies are needed to determine whether bumetanide at the right doses may be considered a potential pharmacologic agent to treat retinal neovascularization.


Assuntos
Bumetanida/farmacologia , Modelos Animais de Doenças , Diuréticos/farmacologia , Neovascularização Patológica/prevenção & controle , Oxigênio/efeitos adversos , Neovascularização Retiniana/prevenção & controle , Retinopatia da Prematuridade/tratamento farmacológico , Animais , Animais Recém-Nascidos , Aquaporina 4/metabolismo , Feminino , Masculino , Neovascularização Patológica/etiologia , Neovascularização Patológica/patologia , Ratos , Ratos Sprague-Dawley , Neovascularização Retiniana/etiologia , Neovascularização Retiniana/patologia , Retinopatia da Prematuridade/etiologia , Retinopatia da Prematuridade/patologia , Fator A de Crescimento do Endotélio Vascular/metabolismo
9.
Pediatr Res ; 2024 Apr 20.
Artigo em Inglês | MEDLINE | ID: mdl-38643263
10.
Prostaglandins Other Lipid Mediat ; 134: 93-107, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-28923362

RESUMO

Topical ocular ketorolac improves the outcomes of severe retinopathy of prematurity and when administered with systemic caffeine, decreases the severity of oxygen-induced retinopathy. We tested the hypothesis that co-cultures of human retinal endothelial cells (HRECs) and human retinal astrocytes (HRAs) on 3-dimensional (3-D) hydrogel scaffolds is a more representative biomimetic paradigm of the blood-retinal-barrier (BRB) than 2-D cultures, and should be utilized for preclinical drug discovery and development. Mono- and co-cultures of HRECs and HRAs were treated with standard doses of ketorolac, ibuprofen, and/or caffeine, and exposed to hyperoxia, intermittent hypoxia (IH), or normoxia on 2-D surfaces or 3-D biodegradable hydrogel scaffolds (AlgiMatrix or Geltrex). Media and cells were collected at 72h post treatment for arachidonic acid metabolites. Cells cultured on 3-D scaffolds exhibited less oxidative stress and variability in drug responses. HRAs enhanced the responses of HRECs to drugs and changes in oxygen environment. PGE2 and PGI2 were the predominant prostanoids produced in response to IH, reflecting COX-2 immunoreactivity. We conclude that HRECs and HRAs co-cultured on 3-D scaffolds may recapitulate drug responses of the dynamic BRB and therefore should be implemented for preclinical ocular drug discovery and development.


Assuntos
Astrócitos/citologia , Astrócitos/efeitos dos fármacos , Descoberta de Drogas , Células Endoteliais/citologia , Células Endoteliais/efeitos dos fármacos , Retina/citologia , Astrócitos/metabolismo , Biomimética , Técnicas de Cocultura , Ciclo-Oxigenase 1/metabolismo , Inibidores de Ciclo-Oxigenase 2/metabolismo , Dinoprosta/análogos & derivados , Dinoprosta/metabolismo , Dinoprostona/metabolismo , Interações Medicamentosas , Células Endoteliais/metabolismo , Humanos , Ibuprofeno/farmacologia , Cetorolaco/farmacologia , Tromboxano B2/metabolismo
11.
Artigo em Inglês | MEDLINE | ID: mdl-29107023

RESUMO

Caffeine, one of the most commonly prescribed drugs in preterm neonates, is given in standard or suprapharmacologic doses. Although known as a diuretic, its effects in the neonatal kidneys are not well studied. We tested the hypothesis that neonatal intermittent hypoxia (IH) and high caffeine doses (HCD) alter renal regulators of vasomotor tone and water balance. Newborn rats were randomized to room air, hyperoxia, or IH and treated with standard or high caffeine doses; or placebo saline. Renal prostanoids; histopathology; and cyclooxygenase (COX), prostanoid receptor, and aquaporin (AQP) immunoreactivity were determined. HCD in IH caused severe pathological changes in the glomeruli and proximal tubules, consistent with acute kidney injury. This was associated with reductions in anthropometric growth, PGI2, and IP, DP, and AQP-4 immunoreactivity, well as a robust increase in COX-2, suggesting that the use of HCD should be avoided in preterm infants who experience frequent IH episodes.


Assuntos
Cafeína/farmacologia , Rim/efeitos dos fármacos , Rim/metabolismo , Prostaglandinas/metabolismo , Receptores de Prostaglandina/metabolismo , Animais , Animais Recém-Nascidos , Aquaporinas/metabolismo , Hipóxia Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Feminino , Gravidez , Prostaglandina-Endoperóxido Sintases/metabolismo , Ratos , Ratos Sprague-Dawley
12.
Int J Mol Sci ; 19(5)2018 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-29724000

RESUMO

Preterm infants often experience intermittent hypoxia (IH) with resolution in room air (RA) or hyperoxia (Hx) between events. Hypoxia is a major inducer of vascular endothelial growth factor, which plays a key role in normal and aberrant retinal angiogenesis. This study tested the hypothesis that neonatal IH which resolved with RA is less injurious to the immature retina than IH resolved by Hx between events. Newborn rats were exposed to: (1) Hx (50% O2) with brief hypoxia (12% O2); (2) RA with 12% O2; (3) Hx with RA; (4) Hx only; or (5) RA only, from P0 to P14. Pups were examined at P14 or placed in RA until P21. Retinal vascular and astrocyte integrity; retinal layer thickness; ocular and systemic biomarkers of angiogenesis; and somatic growth were determined at P14 and P21. All IH paradigms resulted in significant retinal vascular defects, disturbances in retinal astrocyte template, retinal thickening, and photoreceptor damage concurrent with elevations in angiogenesis biomarkers. These data suggest that the susceptibility of the immature retina to changes in oxygen render no differences in the outcomes between RA or O2 resolution. Interventions and initiatives to curtail O2 variations should remain a high priority to prevent severe retinopathy.


Assuntos
Hipóxia/metabolismo , Oxigênio/efeitos adversos , Retina/patologia , Retinopatia da Prematuridade/metabolismo , Ar , Animais , Animais Recém-Nascidos , Astrócitos/patologia , Biomarcadores/sangue , Hiperóxia/metabolismo , Fator de Crescimento Insulin-Like I/metabolismo , Células Fotorreceptoras de Vertebrados/patologia , Ratos , Ratos Sprague-Dawley , Neovascularização Retiniana/induzido quimicamente , Neovascularização Retiniana/metabolismo , Retinopatia da Prematuridade/induzido quimicamente , Fator A de Crescimento do Endotélio Vascular/sangue
13.
Exp Lung Res ; 43(3): 120-133, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-28409646

RESUMO

Purpose/Aim: Intravitreal bevacizumab (Avastin) is an irreversible vascular endothelial growth factor (VEGF) inhibitor used off-label to treat severe retinopathy of prematurity in extremely low gestational age neonates. VEGF and matrix metalloproteinases (MMPs) and the tissue inhibitors of metalloproteinases (TIMPs) participate in lung maturation. We tested the hypothesis that intravitreal bevacizumab enters the systemic circulation and has long-lasting effects on lung MMPs. MATERIALS AND METHODS: Neonatal rats were exposed to: (1) hyperoxia (50% O2); (2) intermittent hypoxia (IH) (50% O2 with brief episodes of 12% O2); or (3) room air (RA) from birth (P0) to P14. At P14, the time of eye opening in rats, a single dose of Avastin (0.125 mg) was injected into the vitreous cavity of the left eye. A control group received equivalent volume saline. At P23 and P45, lung MMP-2 and MMP-9, and TIMP-1, and TIMP-2 were assessed in the lungs. RESULTS: At P23, Avastin increased MMP-2, MMP-9, and TIMP-1 levels in the hyperoxia group but decreased TIMP-1 levels in the IH group. The ratios of MMP-2/TIMP-1 and MMP-9/TIMP-1 were significantly elevated at P23 in the IH group treated with Avastin. At P45, the levels of MMP-2 and MMP-9 remained elevated in the hyperoxia and IH groups treated with Avastin, while a rebound increase in TIMP-1 levels was noted in the IH group. CONCLUSIONS: Avastin treatment in IH has lasting alterations in the balance between MMPs and their tissue inhibitors. These changes may lead to impaired alveologenesis and tissue damage consistent with bronchopulmonary dysplasia/chronic lung disease.


Assuntos
Bevacizumab/farmacologia , Colagenases/metabolismo , Pulmão/crescimento & desenvolvimento , Alvéolos Pulmonares/crescimento & desenvolvimento , Animais , Animais Recém-Nascidos , Displasia Broncopulmonar , Colágeno Tipo IV/metabolismo , Hiperóxia/metabolismo , Hipóxia/metabolismo , Pulmão/enzimologia , Metaloproteinases da Matriz/análise , Metaloproteinases da Matriz/efeitos dos fármacos , Ratos , Inibidor Tecidual de Metaloproteinase-1/análise , Inibidor Tecidual de Metaloproteinase-1/efeitos dos fármacos , Inibidor Tecidual de Metaloproteinase-2/análise , Inibidor Tecidual de Metaloproteinase-2/efeitos dos fármacos , Fator A de Crescimento do Endotélio Vascular
14.
J Biol Chem ; 290(38): 23264-81, 2015 Sep 18.
Artigo em Inglês | MEDLINE | ID: mdl-26242736

RESUMO

The response of the retina to ischemic insult typically leads to aberrant retinal neovascularization, a major cause of blindness. The epigenetic regulation of angiogenic gene expression by miRNAs provides new prospects for their therapeutic utility in retinal neovascularization. Here, we focus on miR-155, a microRNA functionally important in inflammation, which is of paramount importance in the pathogenesis of retinal neovascularization. Whereas constitutive miR-155-deficiency in mice results in mild vascular defects, forced expression of miR-155 causes endothelial hyperplasia and increases microglia count and activation. The mouse model of oxygen-induced retinopathy, which recapitulates ischemia-induced aberrant neovessel growth, is characterized by increased expression of miR-155 and localized areas of microglia activation. Interestingly, miR-155 deficiency in mice reduces microglial activation, curtails abnormal vessel growth, and allows for rapid normalization of the retinal vasculature following ischemic insult. miR-155 binds to the 3'-UTR and represses the expression of the CCN1 gene, which encodes an extracellular matrix-associated integrin-binding protein that both promotes physiological angiogenesis and harnesses growth factor-induced abnormal angiogenic responses. Single CCN1 deficiency or double CCN1 and miR-155 knock-out in mice causes retinal vascular malformations typical of faulty maturation, mimicking the vascular alterations of miR-155 gain of function. During development, the miR-155/CCN1 regulatory axis balances the proangiogenic and proinflammatory activities of microglia to allow for their function as guideposts for sprout fusion and anastomosis. Under ischemic conditions, dysregulated miR-155 and CCN1 expression increases the inflammatory load and microglial activation, prompting aberrant angiogenic responses. Thus, miR-155 functions in tandem with CCN1 to modulate inflammation-induced vascular homeostasis and repair.


Assuntos
Proteína Rica em Cisteína 61/metabolismo , MicroRNAs/metabolismo , Microglia/metabolismo , Retina/metabolismo , Neovascularização Retiniana/metabolismo , Vasos Retinianos/metabolismo , Regiões 3' não Traduzidas/genética , Animais , Proteína Rica em Cisteína 61/genética , Isquemia/genética , Isquemia/metabolismo , Isquemia/patologia , Camundongos , Camundongos Knockout , MicroRNAs/genética , Microglia/patologia , Retina/patologia , Neovascularização Retiniana/genética , Neovascularização Retiniana/patologia , Vasos Retinianos/patologia
15.
Pediatr Res ; 80(4): 554-65, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27438224

RESUMO

BACKGROUND: Caffeine or ketorolac decrease the risk of retinopathy of prematurity and may act synergistically to improve beneficial effect. Combination of caffeine (Caff) and ketorolac (Keto) to prevent oxygen-induced retinopathy was studied. METHODS: Newborn rats exposed to room air (RA) or intermittent hypoxia (IH) consisting of 12% O2 during hyperoxia (50% O2) from birth (P0) had single daily IP injections of Caff from P0-P13 or saline; and/or ocular Keto (Acuvail, 0.45% ophthalmic solution) administered subcutaneously over the eyes from P5-P7. Pups were studied at P14 or placed in RA for recovery from IH (IHR) until P21. Eyes were examined for neovascularization, histopathology, growth factors, and VEGF-signaling genes. RESULTS: Severe retinal damage noted during IHR in the untreated groups evidenced by hemorrhage, neovascularization, and oxygen-induced retinopathy (OIR) pathologies were prevented with Keto/Caff treatment. Keto and/or Caff treatment in IH also promoted retinal neural development evidenced by eye opening (92%, P < 0.001 vs. 31% in the placebo-treated IH group). No corneal pathologies were noted with Keto. CONCLUSION: Caff or Keto given individually reduced retinal neovascularization, but the two drugs given together prevented severe OIR.


Assuntos
Cafeína/administração & dosagem , Citratos/administração & dosagem , Cetorolaco/administração & dosagem , Oxigênio/efeitos adversos , Retinopatia da Prematuridade/tratamento farmacológico , Animais , Animais Recém-Nascidos , Anti-Inflamatórios não Esteroides/administração & dosagem , Apirase/metabolismo , Artérias/metabolismo , Peso Corporal , Estimulantes do Sistema Nervoso Central/administração & dosagem , Corioide/metabolismo , Dinoprosta/análogos & derivados , Dinoprosta/metabolismo , Sinergismo Farmacológico , Feminino , Hemorragia , Subunidade alfa do Fator 1 Induzível por Hipóxia/sangue , Fator de Crescimento Insulin-Like I/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Neovascularização Patológica , Ratos , Ratos Sprague-Dawley , Retina/metabolismo , Retinopatia da Prematuridade/induzido quimicamente , Transdução de Sinais , Fatores de Tempo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/sangue
16.
J Proteome Res ; 14(5): 2109-2120, 2015 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-25780855

RESUMO

Investigation of the retina proteome during hypoxia-induced retinal neovascularization is valuable for understanding pathogenesis of retinopathy of prematurity (ROP). Here we employed a reproducible ion-current-based MS1 quantification approach (ICB) to explore the retinal proteomic changes in early stage of ROP in a rat model of oxygen-induced retinopathy (OIR). Retina proteins, which are rich in membrane proteins, were efficiently extracted by a detergent-cocktail and subjected to precipitation/on-pellet-digestion, followed by nano-LC-MS analysis on a 75-cm column with a 7-h gradient. The high reproducibility of sample preparation and chromatography separation enabled excellent peak alignment and contributed to the superior performance of ICB over parallel label-free approaches. In this study, sum-of-intensity with rejection was incorporated to determine the protein ratios. In total, 1325 unique protein groups were quantified from rat retinas (n = 4/group) with at least two distinct peptides at a protein FDR of 1%. Thirty-two significantly altered proteins were observed with confidence, and the elevated glial fibrillary acidic protein and decreased crystalline proteins in OIR retinas agree well with previous studies. Selected key alterations were further validated by Western blot analysis. Interestingly, Rab21/RhoA/ROCK2/moesin signaling pathway was found to be involved in retinal neovascularization of OIR. Moreover, highly elevated annexin A3, a potential angiogenic mediator, was observed in OIR retinas and may serve as a potential therapeutic target. In conclusion, reproducible ICB profiling enabled reliable discovery of many altered mediators and pathways in OIR retinas, thereby providing new insights into molecular mechanisms involved in pathogenesis of ROP.


Assuntos
Proteínas do Olho/isolamento & purificação , Espectrometria de Massas/métodos , Proteoma/isolamento & purificação , Retina/química , Degeneração Retiniana/genética , Animais , Animais Recém-Nascidos , Anexina A3/genética , Anexina A3/isolamento & purificação , Anexina A3/metabolismo , Clusterina/genética , Clusterina/isolamento & purificação , Clusterina/metabolismo , Modelos Animais de Doenças , Proteínas do Olho/genética , Proteínas do Olho/metabolismo , Regulação da Expressão Gênica , Proteína Glial Fibrilar Ácida/genética , Proteína Glial Fibrilar Ácida/isolamento & purificação , Proteína Glial Fibrilar Ácida/metabolismo , Humanos , Proteínas de Membrana/genética , Proteínas de Membrana/isolamento & purificação , Proteínas de Membrana/metabolismo , Proteínas dos Microfilamentos/genética , Proteínas dos Microfilamentos/isolamento & purificação , Proteínas dos Microfilamentos/metabolismo , Neovascularização Patológica/genética , Oxigênio , Proteoma/genética , Proteoma/metabolismo , Ratos , Ratos Sprague-Dawley , Retina/metabolismo , Retina/patologia , Degeneração Retiniana/induzido quimicamente , Degeneração Retiniana/metabolismo , Degeneração Retiniana/patologia , Retinopatia da Prematuridade/genética , Retinopatia da Prematuridade/metabolismo , Retinopatia da Prematuridade/patologia , Fator de Transcrição STAT1/genética , Fator de Transcrição STAT1/isolamento & purificação , Fator de Transcrição STAT1/metabolismo , Proteínas rab de Ligação ao GTP/genética , Proteínas rab de Ligação ao GTP/isolamento & purificação , Proteínas rab de Ligação ao GTP/metabolismo , Quinases Associadas a rho/genética , Quinases Associadas a rho/isolamento & purificação , Quinases Associadas a rho/metabolismo , Proteína rhoA de Ligação ao GTP/genética , Proteína rhoA de Ligação ao GTP/isolamento & purificação , Proteína rhoA de Ligação ao GTP/metabolismo
17.
BMC Pediatr ; 15: 18, 2015 Mar 08.
Artigo em Inglês | MEDLINE | ID: mdl-25884954

RESUMO

BACKGROUND: Since an objective description is essential to determine infant's postnatal condition and efficacy of interventions, two scores were suggested in the past but weren't tested yet: The Specified-Apgar uses the 5 items of the conventional Apgar score; however describes the condition regardless of gestational age (GA) or resuscitative interventions. The Expanded-Apgar measures interventions needed to achieve this condition. We hypothesized that the combination of both (Combined-Apgar) describes postnatal condition of preterm infants better than either of the scores alone. METHODS: Scores were assessed in preterm infants below 32 completed weeks of gestation. Data were prospectively collected in 20 NICU in 12 countries. Prediction of poor outcome (death, severe/moderate BPD, IVH, CPL and ROP) was used as a surrogate parameter to compare the scores. To compare predictive value the AUC for the ROC was calculated. RESULTS: Of 2150 eligible newborns, data on 1855 infants with a mean GA of 28(6/7) ± 2(3/7) weeks were analyzed. At 1 minute, the Combined-Apgar was significantly better in predicting poor outcome than the Specified- or Expanded-Apgar alone. Of infants with a very low score at 5 or 10 minutes 81% or 100% had a poor outcome, respectively. In these infants the relative risk (RR) for perinatal mortality was 24.93 (13.16-47.20) and 31.34 (15.91-61.71), respectively. CONCLUSION: The Combined-Apgar allows a more appropriate description of infant's condition under conditions of modern neonatal care. It should be used as a tool for better comparison of group of infants and postnatal interventions. TRIAL REGISTRATION: clinicaltrials.gov Protocol Registration System (NCT00623038). Registered 14 February 2008.


Assuntos
Índice de Apgar , Recém-Nascido Prematuro , Salas de Parto , Feminino , Idade Gestacional , Humanos , Lactente , Mortalidade Infantil , Recém-Nascido , Gravidez , Prognóstico , Fatores de Risco
18.
Am J Perinatol ; 32(9): 895-904, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25715313

RESUMO

OBJECTIVE: Immature antioxidant and oxygen-sensing mechanisms are involved in the pathogenesis of the patent ductus arteriosus (PDA). We conducted a prospective, observational, pilot study to test the hypothesis that antioxidant activity is low at birth in preterm infants at risk for symptomatic PDA. STUDY DESIGN: Blood and urine samples were collected within 24 to 48 hours of life in 53 preterm infants (≤32 weeks' gestation) who developed early PDA symptoms and in 30 term (≥37 weeks' gestation) control infants. Thirty preterm infants developed hemodynamically significant PDA (hsPDA) and required pharmacologic treatment and/or PDA ligation. For these infants, blood and urine samples were also collected at 24 hours posttreatment. Samples were analyzed for biomarkers of antioxidant activity, oxidative stress, and lipid peroxidation. RESULTS: At 24 to 48 hours after birth, plasma superoxide dismutase (SOD), urinary catalase, and plasma and urinary 8-isoPGF2α were significantly lower in preterm infants who developed hsPDA. Plasma 8-isoPGF2α levels rebounded post-PDA treatment, while urinary prostaglandin E2, plasma and urinary thromboxane B2, and plasma SOD declined. CONCLUSION: The antioxidant status is low in preterm infants at risk for developing hsPDA. SOD may be a key antioxidant regulating functional ductus arteriosus closure. Therefore, low levels may result in persistence of a hsPDA.


Assuntos
Antioxidantes/uso terapêutico , Biomarcadores/sangue , Biomarcadores/urina , Inibidores de Ciclo-Oxigenase/uso terapêutico , Permeabilidade do Canal Arterial/terapia , Estresse Oxidativo/efeitos dos fármacos , Permeabilidade do Canal Arterial/sangue , Permeabilidade do Canal Arterial/urina , Feminino , Humanos , Ibuprofeno/uso terapêutico , Indometacina/uso terapêutico , Lactente , Recém-Nascido , Recém-Nascido Prematuro/sangue , Recém-Nascido Prematuro/urina , Ligadura , Masculino , Projetos Piloto , Estudos Prospectivos
19.
Pharmaceuticals (Basel) ; 17(3)2024 Mar 17.
Artigo em Inglês | MEDLINE | ID: mdl-38543167

RESUMO

Notch ligands and receptors are important for cell specification and angiogenesis, but their role in oxygen-induced retinopathy (OIR) is not well studied. Delta-like ligand (DLL)-4/Notch inhibits angiogenesis, while Jagged-1/Notch promotes angiogenesis. We tested the hypothesis that early supplementation with antioxidants and/or fish oil curtails severe OIR by inducing DLL-4/Notch and reducing Jagged-1/Notch. Newborn rats were exposed to brief intermittent hypoxia (IH) during hyperoxia, during which they received daily oral supplements of (1) fish oil, (2) coenzyme Q10 (CoQ10) in olive oil (OO), (3) glutathione nanoparticles (nGSH), (4) fish oil + CoQ10, or (5) OO (controls) from birth (P0) to P14. At P14, the pups were placed in room air (RA) until P21, with no further treatment. Oxidative stress, apoptosis, ocular histopathology, and Notch signaling were assessed. Neonatal IH resulted in severe retinal damage consistent with retinopathy of prematurity (ROP). Retinal damage was associated with induced oxidative stress and Jagged-1/Notch signaling, as well as reduced DLL-4/Notch signaling. All treatments reversed these outcomes, but nGSH produced the most beneficial outcomes. Severe OIR promoted the induction of Jagged-1/Notch and curtailed DLL-4/Notch, which was an effect that could be reversed with nGSH supplementation. These findings may indicate a potential alternate pathway for ROP treatment and/or prevention.

20.
Int J Dev Neurosci ; 84(3): 227-250, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38459740

RESUMO

Preterm infants often experience frequent intermittent hypoxia (IH) episodes which are associated with neuroinflammation. We tested the hypotheses that early caffeine and/or non-steroidal inflammatory drugs (NSAIDs) confer superior therapeutic benefits for protection against IH-induced neuroinflammation than late treatment. Newborn rats were exposed to IH or hyperoxia (50% O2) from birth (P0) to P14. For early treatment, the pups were administered: 1) daily caffeine (Caff) citrate (Cafcit, 20 mg/kg IP loading on P0, followed by 5 mg/kg from P1-P14); 2) ketorolac (Keto) topical ocular solution in both eyes from P0 to P14; 3) ibuprofen (Ibu, Neoprofen, 10 mg/kg loading dose on P0 followed by 5 mg/kg/day on P1 and P2); 4) Caff+Keto co-treatment; 5) Caff+Ibu co-treatment; or 6) equivalent volume saline (Sal). On P14, animals were placed in room air (RA) with no further treatment until P21. For late treatment, pups were exposed from P0 to P14, then placed in RA during which they received similar treatments from P15-P21 (Sal, Caff, and/or Keto), or P15-P17 (Ibu). RA controls were similarly treated. At P21, whole brains were assessed for histopathology, apoptosis, myelination, and biomarkers of inflammation. IH caused significant brain injury and hemorrhage, inflammation, reduced myelination, and apoptosis. Early treatment with Caff alone or in combination with NSAIDs conferred better neuroprotection against IH-induced damage than late treatment. Early postnatal treatment during a critical time of brain development, may be preferable for the prevention of IH-induced brain injury in preterm infants.


Assuntos
Animais Recém-Nascidos , Anti-Inflamatórios não Esteroides , Cafeína , Ratos Sprague-Dawley , Animais , Ratos , Anti-Inflamatórios não Esteroides/farmacologia , Cafeína/farmacologia , Cafeína/uso terapêutico , Doenças Neuroinflamatórias/prevenção & controle , Doenças Neuroinflamatórias/tratamento farmacológico , Hipóxia/complicações , Feminino , Masculino , Modelos Animais de Doenças , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Encéfalo/patologia , Ibuprofeno/farmacologia , Ibuprofeno/uso terapêutico , Cetorolaco/farmacologia , Cetorolaco/uso terapêutico
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa