Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
Pediatr Res ; 91(7): 1677-1685, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-34285351

RESUMO

BACKGROUND: Pathologic ocular neovascularization in retinopathy of prematurity (ROP) and other proliferative retinopathies are characterized by dysregulation of vascular endothelial growth factor-A (VEGF-A). A study of Vegfa isoform expression during oxygen-induced ischemic retinopathy (OIR) may enhance our understanding of Vegf dysregulation. METHODS: Following induction of OIR, immunohistochemistry and polymerase chain reaction (PCR) was performed on room air (RA) and OIR mice. RESULTS: Total Vegfa messenger RNA (mRNA) expression was stable in RA mice, but increased in OIR mice with a peak at postnatal day 17 (P17), before returning to RA levels. Vegfa164a expression was similar in both OIR and RA mice at P10 (Phase 1 OIR), but 2.4-fold higher in OIR mice compared to RA mice at P16 (Phase 2 OIR). At P10, Vegfa164b mRNA was similar in OIR vs RA mice, but was expressed 2.5-fold higher in OIR mice compared to RA mice at P16. At P10 and P16, Vegfr2/Vegfr1 expression was increased in OIR mice compared to RA mice. Increased activation of microglia was seen in OIR mice. CONCLUSIONS: Vegfa164a, Vegfa164b, and Vegfr1 were overexpressed in OIR mice, leading to abnormal signaling and angiogenesis. Further studies of mechanisms of Vegf dysregulation may lead to novel therapies for ROP and other proliferative retinopathies. IMPACT: Vegfa164 has two major isoforms, a proangiogenic, Vegfa164a, and an antiangiogenic, Vegfa164b, with opposing receptors, inhibitory Vegfr1, and stimulatory Vegfr2, but their role in OIR is unclear. In Phase 1 OIR, both isoforms and receptors are expressed similarly. In Phase 2 OIR, both isoforms are overexpressed, with an increased ratio of inhibitory Vegfr1. Modulation of angiogenesis by Vegf regulation enables pruning of excess angiogenesis during physiology, but results in ineffective angiogenesis during OIR. Knowledge of VEGF dysregulation may have novel therapeutic implications in the management of ROP and retinal proliferative diseases.


Assuntos
Neovascularização Retiniana , Retinopatia da Prematuridade , Fator A de Crescimento do Endotélio Vascular/metabolismo , Animais , Animais Recém-Nascidos , Modelos Animais de Doenças , Camundongos , Camundongos Endogâmicos C57BL , Neovascularização Patológica/metabolismo , Oxigênio/uso terapêutico , Isoformas de Proteínas , RNA Mensageiro/metabolismo , Neovascularização Retiniana/genética , Retinopatia da Prematuridade/genética , Retinopatia da Prematuridade/patologia
2.
Pediatr Res ; 90(3): 513-523, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-33070164

RESUMO

In the fetus and the neonate, altered macrophage function has been implicated not only in inflammatory disorders but also in developmental abnormalities marked by altered onset, interruption, or imbalance of key structural changes. The developmental role of macrophages were first noted nearly a century ago, at about the same time when these cells were being identified as central effectors in phagocytosis and elimination of microbes. Since that time, we have made considerable progress in understanding the diverse roles that these cells play in both physiology and disease. Here, we review the role of fetal and neonatal macrophages in immune surveillance, innate immunity, homeostasis, tissue remodeling, angiogenesis, and repair of damaged tissues. We also discuss the possibility of therapeutic manipulation of the relative abundance and activation status of macrophage subsets in various diseases. This article combines peer-reviewed evidence from our own studies with results of an extensive literature search in the databases PubMed, EMBASE, and Scopus. IMPACT: We have reviewed the structure, differentiation, and classification of macrophages in the neonatal period. Neonatal macrophages are derived from embryonic, hepatic, and bone marrow precursors. Macrophages play major roles in tissue homeostasis, innate immunity, inflammation, tissue repair, angiogenesis, and apoptosis of various cellular lineages in various infectious and inflammatory disorders. Macrophages and related inflammatory mediators could be important therapeutic targets in several neonatal diseases.


Assuntos
Desenvolvimento Fetal , Recém-Nascido/crescimento & desenvolvimento , Macrófagos/citologia , Diferenciação Celular , Humanos
3.
Pediatr Res ; 89(7): 1619-1626, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33027803

RESUMO

Integrins are heterodimeric transmembrane cell adhesion molecules made up of alpha (α) and beta (ß) subunits arranged in numerous dimeric pairings. These complexes have varying affinities to extracellular ligands. Integrins regulate cellular growth, proliferation, migration, signaling, and cytokine activation and release and thereby play important roles in cell proliferation and migration, apoptosis, tissue repair, as well as in all processes critical to inflammation, infection, and angiogenesis. This review presents current evidence from human and animal studies on integrin structure and molecular signaling, with particular emphasis on signal transduction in infants. We have included evidence from our own laboratory studies and from an extensive literature search in databases PubMed, EMBASE, Scopus, and the electronic archives of abstracts presented at the annual meetings of the Pediatric Academic Societies. To avoid bias in identification of existing studies, key words were short-listed prior to the actual search both from anecdotal experience and from PubMed's Medical Subject Heading (MeSH) thesaurus. IMPACT: Integrins are a family of ubiquitous αß heterodimeric receptors that interact with numerous ligands in physiology and disease. Integrins play a key role in cell proliferation, tissue repair, inflammation, infection, and angiogenesis. This review summarizes current evidence from human and animal studies on integrin structure and molecular signaling and promising role in diseases of inflammation, infection, and angiogenesis in infants. This review shows that integrin receptors and ligands are novel therapeutic targets of clinical interest and hold promise as novel therapeutic targets in the management of several neonatal diseases.


Assuntos
Inflamação/fisiopatologia , Integrinas/fisiologia , Neovascularização Patológica , Animais , Criança , Humanos , Recém-Nascido , Recém-Nascido Prematuro , Ligantes , Ligação Proteica
4.
Mol Vis ; 26: 257-276, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32256029

RESUMO

Purpose: Retinopathy of prematurity (ROP) is a condition of aberrant retinal vascularization in premature infants in response to high levels of oxygen used for critical care that can potentially cause blindness. Although therapies to mitigate vascular abnormalities are being evaluated, functional deficits often remain in patients with treated or regressed ROP. This study investigated long-term outcomes of hyperoxia on retinal morphology and function using a mouse model of oxygen-induced ischemic retinopathy (OIR). Methods: Twenty-two mice were exposed to 77% oxygen to induce OIR, while 23 age-matched control mice were raised in room air (RA). In vivo fluorescein angiography (FA), spectral-domain optical coherence tomography (SD-OCT), and focal electroretinography (fERG) were performed at P19, P24, P32, and P47, followed by histological assessments of retinal morphology, gliosis, microglia activation, and apoptosis. Results: FA in OIR mice showed capillary attrition despite peripheral revascularization. Inner retina thinning was detected with SD-OCT; outer and inner retinal dysfunction were demonstrated with fERG. Histology of the OIR mice exhibited a thin, disorganized structure. Immunohistochemistry showed increased gliosis, microglial activation, and apoptosis with increasing age from P19 to P47. The synapses between rod photoreceptor cells and rod bipolar cells were ectopically localized in the OIR mice. Conclusions: We demonstrated histological evidence of persistent ectopic synapses, prolonged cellular apoptosis, and gliosis in the OIR retina that corresponded with long-term in vivo evidence of capillary attrition, inner retinal thinning, and dysfunction despite full peripheral revascularization. Further studies on the mechanisms underlying these persistent phenotypes could enhance our understanding of ROP pathogenesis and lead to new therapeutic targets to preserve visual function in premature infants.


Assuntos
Apoptose , Hiperóxia/complicações , Oxigênio/efeitos adversos , Retina/crescimento & desenvolvimento , Retina/patologia , Neovascularização Retiniana/patologia , Retinopatia da Prematuridade/fisiopatologia , Animais , Animais Recém-Nascidos , Dendritos/metabolismo , Modelos Animais de Doenças , Eletrorretinografia , Angiofluoresceinografia , Gliose/patologia , Hiperóxia/patologia , Hiperóxia/fisiopatologia , Imuno-Histoquímica , Camundongos , Camundongos Endogâmicos C57BL , Microglia/metabolismo , Microglia/patologia , Células Fotorreceptoras/metabolismo , Proteína Quinase C-alfa/metabolismo , Retina/metabolismo , Células Bipolares da Retina/metabolismo , Retinopatia da Prematuridade/diagnóstico por imagem , Sinapses/metabolismo , Tomografia de Coerência Óptica
5.
J Pediatr ; 206: 56-65.e8, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30471715

RESUMO

OBJECTIVE: To investigate recombinant human insulin-like growth factor 1 complexed with its binding protein (rhIGF-1/rhIGFBP-3) for the prevention of retinopathy of prematurity (ROP) and other complications of prematurity among extremely preterm infants. STUDY DESIGN: This phase 2 trial was conducted from September 2014 to March 2016. Infants born at a gestational age of 230/7 weeks to 276/7 weeks were randomly allocated to rhIGF-1/rhIGFBP-3 (250 µg/kg/ 24 hours, continuous intravenous infusion from <24 hours of birth to postmenstrual age 296/7 weeks) or standard neonatal care, with follow-up to a postmenstrual age of 404/7 weeks. Target exposure was ≥70% IGF-1 measurements within 28-109 µg/L and ≥70% intended therapy duration. The primary endpoint was maximum severity of ROP. Secondary endpoints included time to discharge from neonatal care, bronchopulmonary dysplasia, intraventricular hemorrhage, and growth measures. RESULTS: Overall, 61 infants were allocated to rhIGF-1/rhIGFBP-3, 60 to standard care (full analysis set); 24 of 61 treated infants achieved target exposure (evaluable set). rhIGF-1/rhIGFBP-3 did not decrease ROP severity or ROP occurrence. There was, however, a 53% decrease in severe bronchopulmonary dysplasia in the full analysis set (21.3% treated vs 44.9% standard care), and an 89% decrease in the evaluable set (4.8% vs 44.9%; P = .04 and P = .02, respectively) for severity distribution between groups. There was also a nonsignificant trend toward decrease in grades 3-4 intraventricular hemorrhage in the full analysis set (13.1% vs 23.3%) and in the evaluable set (8.3% vs 23.3%). Fatal serious adverse events were reported in 19.7% of treated infants (12/61) and 11.7% of control infants (7/60). No effect was observed on time to discharge from neonatal care/growth measures. CONCLUSIONS: rhIGF-1/rhIGFBP-3 did not affect development of ROP, but decreased the occurrence of severe bronchopulmonary dysplasia, with a nonsignificant decrease in grades 3-4 intraventricular hemorrhage. TRIAL REGISTRATION: ClinicalTrials.gov: NCT01096784.


Assuntos
Hemorragia Cerebral/prevenção & controle , Fator de Crescimento Insulin-Like I/uso terapêutico , Proteínas Recombinantes/uso terapêutico , Retinopatia da Prematuridade/prevenção & controle , Displasia Broncopulmonar/prevenção & controle , Hemorragia Cerebral/terapia , Feminino , Idade Gestacional , Humanos , Lactente Extremamente Prematuro , Recém-Nascido , Recém-Nascido Prematuro , Infusões Intravenosas , Proteína 3 de Ligação a Fator de Crescimento Semelhante à Insulina/uso terapêutico , Masculino , Retinopatia da Prematuridade/mortalidade , Retinopatia da Prematuridade/terapia , Índice de Gravidade de Doença , Resultado do Tratamento
6.
Optom Vis Sci ; 93(10): 1268-1279, 2016 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-27668493

RESUMO

PURPOSE: Retinopathy of prematurity (ROP) is a potentially blinding vasoproliferative disease. There is no standardized way to quantify plus disease (tortuous and dilated retinal vessels) or characterize abnormal recovery during ROP monitoring. This study objectively studies vascular features in live mice during development using noninvasive retinal imaging. METHODS: Using fluorescein angiography (FA), retinal vascular features were quantified in live mice with oxygen induced retinopathy (OIR). A total of 105 wild-type mice were exposed to 77% oxygen from postnatal day 7 (P7) till P12 (OIR mice). Also, 105 age-matched pups were raised in room air (RA mice). In vivo FA was performed at early (P16 to P20), mid (P23 to P27), late (P30 to P34), and mature (P47) phases of retinal vascular development. Retinal vascular area, retinal vein width, and retinal artery tortuosity were quantified. RESULTS: Retinal artery tortuosity was higher in OIR than RA mice at early (p < 0.0001), mid (p < 0.0001), late (p < 0.0001), and mature (p < 0.0001) phases. Retinal vascular area in OIR mice increased from early to mid-phase (p < 0.0001), but remained unchanged from mid to late (p = 0.23), and from late to mature phase (p = 0.98). Retinal vein width was larger in OIR mice compared to RA mice during early phase only. Arteries in OIR mice were more tortuous from early to mid-phase (p < 0.0001), but tortuosity remained stable from mid through mature phase. RA mice had an increase in retinal vascular area from early to late phase, but maintained uniform retinal vein width and retinal artery tortuosity in all phases. CONCLUSIONS: In vivo FA distinguished arterial and venous features, similar to plus disease, and revealed aberrant recovery of OIR mice (arterial tortuosity, reduced capillary density, and absent neovascular buds) that persisted into adulthood. Retinal artery tortuosity may be a reliable, objective marker of severity of ROP. Infants with abnormal retinal vascular recovery may need extended monitoring.

7.
Exp Eye Res ; 122: 119-22, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24703909

RESUMO

The aberrantly vascularized peripheral retina in retinopathy of prematurity (ROP) may be associated with visual field constriction, retinal dysfunction, and abnormalities in retinal thickness which is commonly assessed by spectral domain optical coherence tomography (SDOCT). However, due to the limitation of SDOCT for peripheral retinal imaging, retinal thickness in avascular peripheral retina in ROP has not been evaluated. Oxygen induced retinopathy (OIR) in mice has features of vasculopathy similar to those in human ROP. These features occur in the posterior retina and thereby are accessible by standard imaging methods. The purpose of the current study was to determine the correspondence between abnormalities in retinal thickness and vasculopathy in neonatal OIR mice by simultaneous SDOCT imaging and fluorescein angiography (FA). Newborn mice (N = 19; C57BL/6J strain) were exposed to 77% oxygen from postnatal day 7 (P7) to P12. Age-matched control mice (N = 12) were raised in room air. FA and SDOCT were performed in mice between P17 and P19 to visualize retinal vasculature and measure retinal thickness, respectively. Retinal thickness measurements in vascular regions of interest (ROIs) of control mice, and in hypovascular and avascular ROIs of OIR mice were compared. In control mice, FA showed uniformly dense retinal capillary networks between major retinal vessels and retinal thickness of vascular ROIs was 260 ± 7 µm (N = 12). In OIR mice, FA displayed hypovascular regions with less dense and fewer capillaries and avascular regions devoid of visible capillaries. Retinal thickness measurements of hypovascular and avascular ROIs were 243 ± 21 µm and 209 ± 11 µm (N = 19), respectively. Retinal thickness in hypovascular and avascular ROIs of OIR mice was significantly lower than in vascular ROIs of control mice (p ≤ 0.01). Likewise, retinal thickness in avascular ROIs was significantly lower than in hypovascular ROIs (p < 0.001). Retinal thinning in hypovascular and avascular regions may be due to arrested retinal development and/or ischemia induced apoptosis.


Assuntos
Modelos Animais de Doenças , Oxigênio/toxicidade , Retina/patologia , Vasos Retinianos/patologia , Retinopatia da Prematuridade/diagnóstico , Animais , Animais Recém-Nascidos , Angiofluoresceinografia , Camundongos , Camundongos Endogâmicos C57BL , Retinopatia da Prematuridade/induzido quimicamente , Retinopatia da Prematuridade/fisiopatologia , Tomografia de Coerência Óptica
8.
Newborn (Clarksville) ; 1(1): 44-57, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35754998

RESUMO

In both fetal and neonatal physiologic and pathologic processes in most organs, endothelial cells are known to play critical roles. Although the endothelium is one of the most ubiquitous cell type in the body, the tight adherence to the blood vessel wall has made it difficult to study their diverse function and structure. In this article, we have reviewed endothelial cell origins and explored their heterogeneity in terms of structure, function, developmental changes, and their role in inflammatory and infectious diseases. We have also attempted to evaluate the untapped therapeutic potentials of endothelial cells in neonatal disease. This article comprises various peer-reviewed studies, including ours, and an extensive database literature search from EMBASE, PubMed, and Scopus.

9.
Ann Glob Health ; 88(1): 105, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36474896

RESUMO

Objectives: Iron-deficiency anemia (IDA) is a global cause of morbidity in children under five, particularly in sub-Saharan Africa. In southeast Nigeria, poor dietary intake and caregiver knowledge about childhood anemia are observed; however, there is no consensus on how to best prevent it. This study seeks to test the effectiveness of caregiver education on improving anemia knowledge and dietary prevention strategies and promoting sustainable lifestyle changes to reduce the prevalence of childhood IDA. Study Design: A questionnaire was administered to the primary caregivers of 41 patients under age five with anemia in southeast Nigeria regarding socioeconomic status (SES), diet diversity, and risk factors for anemia. Caregivers were administered a preeducation questionnaire, poster education on anemia and iron-rich foods, and a posteducation questionnaire. All patients underwent a medical exam to confirm a diagnosis of anemia or anemia-related conditions. Results: Ninety-five percent of patients had moderate diet diversity, but there was no correlation between diet diversity and SES. Barriers to healthier diets were associated with SES. Preeducation scores were not associated with caregivers' education levels; however, posteducation scores were significantly higher in university-educated than technical-trained caregivers. Caregiver-reported self-efficacy increased after the education program. Conclusion: Caregivers' SES was associated with financial and knowledge barriers to a healthier diet but not diet diversity, suggesting that nutritional education could benefit all SES groups. Overall, the education program increased caregivers' anemia knowledge across educational levels. A community-based health education program could improve caregivers' anemia knowledge and self-efficacy in applying this information and potentially reduce this area's pediatric IDA.


Assuntos
Anemia Ferropriva , Humanos , Criança , Anemia Ferropriva/epidemiologia , Anemia Ferropriva/prevenção & controle , Educação em Saúde , Classe Social , Universidades , Escolaridade
10.
Brain Res ; 1796: 148093, 2022 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-36116486

RESUMO

OBJECTIVE: Hypoxic ischemic encephalopathy (HIE) secondary to perinatal asphyxia leads to long-term visual disabilities. Dilated retinal exams in human newborns with HIE is an emerging diagnostic tool, but phenotypes of hypoxia ischemia (HI) related retinal vascular injury are unclear. 7,8-Dihydroxyflavone (7,8-DHF) is a TrkB agonist with protective effects on HI-related brain damage. We studied retinal vessels in a mouse model of neonatal HIE and the efficacy of 7,8-DHF in ameliorating HI-related retinal vascular injury. METHODS: C57BL6/J mice at post-natal day (P) 9 received unilateral left carotid artery ligation followed by exposure to 10 % oxygen for 50 min. Phosphate buffered saline or 7,8-DHF (5 mg/kg) were administered daily for 7 days intraperitoneally. Control groups of naïve or carotid artery ligation only mice were studied. Fluorescein angiography was performed in acute (two weeks post-exposure) and chronic (four weeks post-exposure) time points. Retinal artery width, retinal vein width, and collateral vessel length were quantified. RESULTS: Ligation of the common carotid artery alone caused retinal artery dilation in acute and chronic time points, but had no effect on retinal veins. At acute time point, HI caused increased retinal artery vasodilation, but was reversed by 7,8-DHF. HI caused short collateral vessel formation in ipsilateral eyes, rescued by 7,8-DHF treatment. CONCLUSION: Retinal artery vasodilation and collateral vessel formation due to HI were rescued by 7,8-DHF treatment. Retinal and collateral vessel monitoring could be diagnostic biomarkers for HI severity. Studies to elucidate mechanisms of 7,8-DHF action on retinal vessels could aid development of therapies for neonatal HI.


Assuntos
Asfixia Neonatal , Hipóxia-Isquemia Encefálica , Doenças Retinianas , Lesões do Sistema Vascular , Animais , Animais Recém-Nascidos , Biomarcadores , Humanos , Hipóxia , Hipóxia-Isquemia Encefálica/complicações , Hipóxia-Isquemia Encefálica/diagnóstico por imagem , Hipóxia-Isquemia Encefálica/tratamento farmacológico , Recém-Nascido , Isquemia/complicações , Camundongos , Camundongos Endogâmicos C57BL , Oxigênio , Fosfatos , Receptores Proteína Tirosina Quinases , Lesões do Sistema Vascular/complicações
11.
Eye (Lond) ; 33(3): 363-373, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30209267

RESUMO

BACKGROUND: Retinopathy of prematurity is a condition of abnormal retinal vascularization in premature infants. The effect of abnormal vascularization on retinal structure and function is unclear. In vivo studies of retinal vascularization, thickness, and function were performed in mice with oxygen-induced retinopathy (OIR mice). METHODS: Eighteen mice were exposed to hyperoxia at postnatal day (P) 7, whereas 18 mice were raised in room air (RA). At P20 and 40, electroretinogram was performed for a-wave and b-wave amplitudes and peak times, followed by simultaneous fluorescein angiography for retinal avascular area, arterial tortuosity, and vein dilation assessments, and spectral domain optical coherence tomography for retinal thickness. RESULTS: Capillary density appeared sparser in OIR mice, but retinal avascular area similar to RA mice. Retinal artery tortuosity was higher at P20 and P40 (P = 0.0001) in OIR than RA mice. OIR mice had dilated retinal veins at P20 and thinner inner retinas at P40. Retinal vein width positively correlated with inner retinal thickness (P = 0.008). b-wave amplitude was decreased in avascular retinal areas, and correlated with inner retinal thinning. b-wave peak time was prolonged in adult OIR mice at high intensities (P = 0.03). CONCLUSIONS: Focal variations in retinal vascularization of OIR mice correlate with thickness and function. Adult OIR mice had increased retinal artery tortuosity, prolonged b-wave peak time, and decreased retinal vein width with inner retina attrition. These suggest abnormalities in inner retinal morphology or post-receptor signaling. Studying interactions between retinal vascular, structural, and functional changes could enhance knowledge of OIR pathogenesis and potential therapies.


Assuntos
Hiperóxia/complicações , Oxigênio/toxicidade , Retina/patologia , Neovascularização Retiniana/patologia , Vasos Retinianos/patologia , Retinopatia da Prematuridade/patologia , Animais , Animais Recém-Nascidos , Modelos Animais de Doenças , Eletrorretinografia , Angiofluoresceinografia , Hiperóxia/fisiopatologia , Camundongos , Camundongos Endogâmicos C57BL , Retina/ultraestrutura , Neovascularização Retiniana/fisiopatologia , Retinopatia da Prematuridade/induzido quimicamente , Retinopatia da Prematuridade/fisiopatologia
12.
Int J Pediatr ; 2019: 9457981, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31281394

RESUMO

INTRODUCTION: Child mortality rate in sub-Saharan Africa is 29 times higher than that in industrialized countries. Anemia is one of the preventable causes of child morbidity. During a humanitarian medical mission in rural South-Eastern Nigeria, the prevalence and risk factors of anemia were determined in the region in order to identify strategies for reduction. METHODS: A cross-sectional study was done on 96 children aged 1-7 years from 50 randomly selected families. A study questionnaire was used to collect information regarding socioeconomic status, family health practices, and nutrition. Anemia was diagnosed clinically or by point of care testing of hemoglobin (Hb) levels. RESULTS: 96 children were selected for the study; 90 completed surveys were analyzed (43% male and 57% females). Anemia was the most prevalent clinical morbidity (69%), followed by intestinal worm infection (53%) and malnutrition (29%). Mean age (months) at which breastfeeding was stopped was 11.8 (±2.2) in children with Hb <11mg/dl (severe anemia), 10.5±2.8 in those with Hb = 11-11.9mg/dl (mild-moderate anemia), and 9.4±3.9 in children with Hb >12mg/dl (no anemia) (P=0.0445). CONCLUSIONS: The longer the infant was breastfed, the worse the severity of childhood anemia was. Childhood anemia was likely influenced by the low iron content of breast milk in addition to maternal anemia and poor nutrition. A family-centered preventive intervention for both maternal and infant nutrition may be more effective in reducing childhood anemia and child mortality rate in the community.

13.
Curr Eye Res ; 44(3): 275-286, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30383455

RESUMO

PURPOSE: Retinopathy of prematurity (ROP) is a condition of abnormal retinal vascularization with reduced levels of vascular endothelial growth factor (VEGF) causing vaso-obliteration (Phase I), followed by abnormal neovascularization from increased VEGF (Phase II). We hypothesized that intravitreal pro-angiogenic VEGF-A in microparticle form would promote earlier retinal revascularization in an oxygen-induced ischemic retinopathy (OIR) mouse model. MATERIALS AND METHODS: Wildtype mice (39) were exposed to 77% oxygen from postnatal day 7 (P7) to P12. VEGF-A165-loaded poly(lactic-co-glycolic acid) (PLGA) (n = 15) or empty PLGA (n = 14) microparticles were fabricated using a water-in-oil-in-water double emulsion method, and injected intravitreally at P13 into mice right eyes (RE). Left eyes (LE) were untreated. At P20, after retinal fluorescein angiography, vascular parameters were quantified. Retinal VEGF levels at P13 and flatmounts at P20 were performed separately. RESULTS: VEGF-A165-loaded microparticles had a mean diameter of 4.2 µm. with a loading level of 8.6 weight.%. Retinal avascular area was reduced in VEGF-treated RE (39.5 ± 9.0%) compared to untreated LE (52.6 ± 6.1%, p < 0.0001) or empty microparticle-treated RE (p < 0.001) and untreated LEs (p = 0.001). Retinal arteries in VEGF-treated RE were less tortuous than untreated LE (1.08 ± 0.05 vs. 1.18 ± 0.08, p < 0.001) or empty-microparticles-treated RE (p = 0.02). Retinal arterial tortuosity was similar in the LE of VEGF and empty microparticle-treated mice (P > 0.05). Retinal vein width was similar in VEGF-treated and empty microparticle-treated RE (P > 0.9), which were each less dilated than their contralateral LE (p < 0.01). VEGF levels were higher in P13 OIR mice than RA mice (p < 0.0001). Retinal flatmounts showed vaso-obliteration and neovascularization. CONCLUSIONS: Endogenous retinal VEGF is suppressed in OIR mice. Exogenous intravitreal VEGF-A165-loaded microparticles in OIR mice reduced retinal vaso-obliteration and accelerated recovery from vein dilation and arterial tortuosity. This may be beneficial in preventing Phase II ROP without systemic effects.


Assuntos
Modelos Animais de Doenças , Sistemas de Liberação de Medicamentos , Copolímero de Ácido Poliláctico e Ácido Poliglicólico/química , Neovascularização Retiniana/tratamento farmacológico , Retinopatia da Prematuridade/tratamento farmacológico , Fator A de Crescimento do Endotélio Vascular/uso terapêutico , Animais , Portadores de Fármacos , Ensaio de Imunoadsorção Enzimática , Angiofluoresceinografia , Injeções Intravítreas , Camundongos Endogâmicos C57BL , Microesferas , Oxigênio/toxicidade , Neovascularização Retiniana/metabolismo , Vasos Retinianos/efeitos dos fármacos , Vasos Retinianos/metabolismo , Vasos Retinianos/patologia , Retinopatia da Prematuridade/metabolismo , Fator A de Crescimento do Endotélio Vascular/química , Fator A de Crescimento do Endotélio Vascular/metabolismo
14.
J Ophthalmol ; 2017: 9620876, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28573047

RESUMO

BACKGROUND: Retinopathy of prematurity (ROP) is a condition of abnormal retinal vascular development (RVD) in premature infants. Fluorescein angiography (FA) has depicted phases (early, mid, late, and mature) of RVD in oxygen-induced retinopathy (OIR) mice. We sought to establish the relationship between retinal structural and vascular changes using simultaneous FA and spectral domain optical coherence tomography (SD-OCT). METHOD: 63 mice were exposed to 77% oxygen at postnatal day 7 (P7) for 5 days, while 63 mice remained in room air (RA). Total retinal thickness (TRT), inner retinal thickness (IRT), and outer retinal thickness (ORT) were calculated at early (P19), mid (P24), late (P32), and mature (P47) phases of RVD. RESULTS: TRT was reduced in OIR (162.66 ± 17.75 µm, n = 13) compared to RA mice at P19 (197.57 ± 3.49 µm, n = 14), P24, P32, and P49 (P < 0.0001). ORT was similar in RA and OIR mice at all ages (P > 0.05). IRT was reduced in OIR (71.60 ± 17.14 µm) compared to RA (103.07 ± 3.47 µm) mice at P19 and all ages (P < 0.0001). CONCLUSION: We have shown the spatial and temporal relationship between retinal structure and vascular development in OIR. Significant inner retinal thinning in OIR mice persisted despite revascularization of the capillary network; further studies will elucidate its functional implications in ROP.

16.
Invest Ophthalmol Vis Sci ; 54(10): 6968-72, 2013 Oct 23.
Artigo em Inglês | MEDLINE | ID: mdl-24052641

RESUMO

PURPOSE: Oxygenation abnormalities are implicated in the development of retinopathy of prematurity (ROP). The purpose of this study is to report in vivo retinal vascular oxygen tension (PO2) measurements and fluorescein angiography (FA) findings in the mouse model of oxygen-induced retinopathy (OIR). METHODS: We exposed 19 neonatal mice to 77% oxygen from postnatal day 7 (P7) to P12 (OIR), while 11 neonatal mice were kept under room air (control). Using phosphorescence lifetime imaging, retinal vascular PO2 was measured followed by FA. Repeated measures ANOVA was performed to determine the effects of blood vessel type (artery and vein) and group (OIR and control) on PO2. Avascular retinal areas were measured from FA images in OIR mice. RESULTS: There was a significant effect of vessel type on PO2 (P < 0.001). The effect of group on PO2 was not significant (P = 0.3), indicating similar PO2 between OIR and control mice. The interaction between group and vessel type was significant (P = 0.03), indicating a larger arteriovenous PO2 difference in OIR mice than control mice. In control mice, FA displayed normal vascularization, while FA of OIR mice showed abnormalities, including dilation and tortuosity of major retinal blood vessels, and avascular regions. In OIR mice, the mean percent avascular retinal area was 33% ± 18%. CONCLUSIONS: In vivo assessment of retinal vascular oxygen tension and vascularization patterns demonstrated abnormalities in the mouse model of OIR. This approach has the potential to improve understanding of retinal vascular development and oxygenation alterations due to ROP and other ischemic retinal diseases.


Assuntos
Oxigênio/metabolismo , Vasos Retinianos/metabolismo , Retinopatia da Prematuridade/metabolismo , Análise de Variância , Animais , Modelos Animais de Doenças , Angiofluoresceinografia , Camundongos , Camundongos Endogâmicos C57BL , Oxigênio/farmacologia , Vasos Retinianos/patologia , Retinopatia da Prematuridade/patologia
17.
Drugs ; 72(7): 907-16, 2012 May 07.
Artigo em Inglês | MEDLINE | ID: mdl-22564132

RESUMO

Persistent patency of the ductus arteriosus is a major cause of morbidity and mortality in premature infants. In infants born prior to 28 weeks of gestation, a haemodynamically significant patent ductus arteriosus (PDA) can cause cardiovascular instability, exacerbate respiratory distress syndrome, prolong the need for assisted ventilation and increase the risk of bronchopulmonary dysplasia, intraventricular haemorrhage, renal dysfunction, cerebral palsy and mortality. We review the pathophysiology, clinical features and assessment of haemodynamic significance, and provide a rigorous appraisal of the quality of evidence to support current medical and surgical management of PDA of prematurity. Cyclo-oxygenase inhibitors such as indomethacin and ibuprofen remain the mainstay of medical therapy for PDA, and can be used both for prophylaxis as well as for rescue therapy to achieve PDA closure. Surgical ligation is also effective and is used in infants who do not respond to medical management. Although both medical and surgical treatment have proven efficacy in closing the ductus, both modalities are associated with significant adverse effects. Because the ductus does undergo spontaneous closure in some premature infants, improved and early identification of infants most likely to develop a symptomatic PDA could help in directing treatment to the at-risk infants and allow others to receive expectant management.


Assuntos
Permeabilidade do Canal Arterial/complicações , Recém-Nascido Prematuro , Inibidores de Ciclo-Oxigenase/uso terapêutico , Permeabilidade do Canal Arterial/diagnóstico , Permeabilidade do Canal Arterial/terapia , Humanos , Ibuprofeno/uso terapêutico , Indometacina/uso terapêutico , Recém-Nascido , Ligadura/métodos , Fatores de Risco
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA