Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 85
Filtrar
2.
iScience ; 27(6): 109975, 2024 Jun 21.
Artículo en Inglés | MEDLINE | ID: mdl-38827398

RESUMEN

Severe COVID-19 often leads to secondary infections and sepsis that contribute to long hospital stays and mortality. However, our understanding of the precise immune mechanisms driving severe complications after SARS-CoV-2 infection remains incompletely understood. Here, we provide evidence that the SARS-CoV-2 envelope (E) protein initiates innate immune inflammation, via toll-like receptor 2 signaling, and establishes a sustained state of innate immune tolerance following initial activation. Monocytes in this tolerant state exhibit reduced responsiveness to secondary stimuli, releasing lower levels of cytokines and chemokines. Mice exposed to E protein before secondary lipopolysaccharide challenge show diminished pro-inflammatory cytokine expression in the lung, indicating that E protein drives this tolerant state in vivo. These findings highlight the potential of the SARS-CoV-2 E protein to induce innate immune tolerance, contributing to long-term immune dysfunction that could lead to susceptibility to subsequent infections, and uncovers therapeutic targets aimed at restoring immune function following SARS-CoV-2 infection.

3.
J Physiol ; 602(10): 2265-2285, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38632887

RESUMEN

The mechanisms governing brain vascularization during development remain poorly understood. A key regulator of developmental vascularization is delta like 4 (DLL4), a Notch ligand prominently expressed in endothelial cells (EC). Exposure to hyperoxia in premature infants can disrupt the development and functions of cerebral blood vessels and lead to long-term cognitive impairment. However, its role in cerebral vascular development and the impact of postnatal hyperoxia on DLL4 expression in mouse brain EC have not been explored. We determined the DLL4 expression pattern and its downstream signalling gene expression in brain EC using Dll4+/+ and Dll4+/LacZ mice. We also performed in vitro studies using human brain microvascular endothelial cells. Finally, we determined Dll4 and Cldn5 expression in mouse brain EC exposed to postnatal hyperoxia. DLL4 is expressed in various cell types, with EC being the predominant one in immature brains. Moreover, DLL4 deficiency leads to persistent abnormalities in brain microvasculature and increased vascular permeability both in vivo and in vitro. We have identified that DLL4 insufficiency compromises endothelial integrity through the NOTCH-NICD-RBPJ-CLDN5 pathway, resulting in the downregulation of the tight junction protein claudin 5 (CLDN5). Finally, exposure to neonatal hyperoxia reduces DLL4 and CLDN5 expression in developing mouse brain EC. We reveal that DLL4 is indispensable for brain vascular development and maintaining the blood-brain barrier's function and is repressed by neonatal hyperoxia. We speculate that reduced DLL4 signalling in brain EC may contribute to the impaired brain development observed in neonates exposed to hyperoxia. KEY POINTS: The role of delta like 4 (DLL4), a Notch ligand in vascular endothelial cells, in brain vascular development and functions remains unknown. We demonstrate that DLL4 is expressed at a high level during postnatal brain development in immature brains and DLL4 insufficiency leads to abnormal cerebral vasculature and increases vascular permeability both in vivo and in vitro. We identify that DLL4  regulates endothelial integrity through NOTCH-NICD-RBPJ-CLDN5 signalling. Dll4 and Cldn5 expression are decreased in mouse brain endothelial cells exposed to postnatal hyperoxia.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales , Animales Recién Nacidos , Proteínas de Unión al Calcio , Claudina-5 , Células Endoteliales , Hiperoxia , Receptores Notch , Animales , Humanos , Ratones , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Encéfalo/metabolismo , Encéfalo/irrigación sanguínea , Encéfalo/crecimiento & desarrollo , Proteínas de Unión al Calcio/metabolismo , Proteínas de Unión al Calcio/genética , Células Cultivadas , Claudina-5/metabolismo , Claudina-5/genética , Células Endoteliales/metabolismo , Hiperoxia/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Péptidos y Proteínas de Señalización Intracelular/genética , Proteínas de la Membrana/metabolismo , Proteínas de la Membrana/genética , Ratones Endogámicos C57BL , Receptores Notch/metabolismo , Receptores Notch/genética , Transducción de Señal
8.
Front Pediatr ; 11: 1284085, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-38130941

RESUMEN

Background: Necrotizing enterocolitis (NEC) is a devastating disease in premature infants, and 50% of infants with surgical NEC develop neurodevelopmental defects. The mechanisms by which NEC-induced cytokine release and activation of inflammatory cells in the brain mediate neuronal injury, and whether enteral immunotherapy attenuates NEC-associated brain injury remain understudied. Based on our prior work, which demonstrated that experimental NEC-like intestinal injury is attenuated by the short-chain fatty acid, butyrate, in this study, we hypothesize that NEC-induced brain injury would be suppressed by enteral butyrate supplementation. Methods: A standardized NEC mouse model [enteral formula feeding, lipopolysaccharide (LPS), and hypoxia] was used. Mice were randomized into the following groups: control, NEC, butyrate pretreated NEC, and butyrate control. NEC scoring (1-4 with 4 representing severe injury) was performed on ileal sections using a validated scoring system. Intestinal and brain lysates were used to assess inflammation, proinflammatory signaling, and apoptosis. Results: NEC-induced intestinal injury was attenuated by butyrate supplementation. NEC-induced microglial activation in the cerebral cortex and hippocampus was suppressed with butyrate. NEC increased the number of activated microglial cells but decreased the number of oligodendrocytes. Butyrate pretreatment attenuated these changes. Increased activation of proinflammatory Toll-like receptor signaling, cytokine expression, and induction of GFAP and IBA1 in the cerebral cortex observed with NEC was suppressed with butyrate. Conclusion: Experimental NEC induced inflammation and activation of microglia in several regions of the brain, most prominently in the cortex. NEC-induced neuroinflammation was suppressed with butyrate pretreatment. The addition of short-chain fatty acids to diet may be used to attenuate NEC-induced intestinal injury and neuroinflammation in preterm infants.

9.
Cells ; 12(22)2023 11 17.
Artículo en Inglés | MEDLINE | ID: mdl-37998383

RESUMEN

Endothelial cells are the crucial inner lining of blood vessels, which are pivotal in vascular homeostasis and integrity. However, these cells are perpetually subjected to a myriad of mechanical, chemical, and biological stresses that can compromise their plasma membranes. A sophisticated repair system involving key molecules, such as calcium, annexins, dysferlin, and MG53, is essential for maintaining endothelial viability. These components orchestrate complex mechanisms, including exocytosis and endocytosis, to repair membrane disruptions. Dysfunctions in this repair machinery, often exacerbated by aging, are linked to endothelial cell death, subsequently contributing to the onset of atherosclerosis and the progression of cardiovascular diseases (CVD) and stroke, major causes of mortality in the United States. Thus, identifying the core machinery for endothelial cell membrane repair is critically important for understanding the pathogenesis of CVD and stroke and developing novel therapeutic strategies for combating CVD and stroke. This review summarizes the recent advances in understanding the mechanisms of endothelial cell membrane repair. The future directions of this research area are also highlighted.


Asunto(s)
Enfermedades Cardiovasculares , Accidente Cerebrovascular , Humanos , Células Endoteliales , Membrana Celular/metabolismo , Membranas , Enfermedades Cardiovasculares/metabolismo , Accidente Cerebrovascular/metabolismo
10.
Am J Perinatol ; 2023 Dec 29.
Artículo en Inglés | MEDLINE | ID: mdl-38011861

RESUMEN

OBJECTIVE: Physiologic breast milk production in the first 24 hours is estimated to be between 2 and 10 mL per feed. Many mothers intending to breastfeed use formula supplementation (FS) early on, which can affect successful breastfeeding. Whether the volume and timing of FS introduced in the first 24 hours of life (24 HOL) impacts the rate of "breastfeeding at discharge" (BFAD) is not well-studied and was investigated herein. STUDY DESIGN: Single-center, retrospective, chart review of breastfeeding infants born at ≥35 weeks who received supplementation in the first 24 HOL. Comprehensive demographic data pertaining to maternal and infant characteristics, along with infant feeding data, were collected. Four supplementation characteristics, (timing, rate, volume [mL/kg per feed], and type [expressed breast milk (EBM) or formula]) were correlated with BFAD. RESULTS: Among 3,102 supplemented infants in whom mothers intended to breastfeed, 1,031 (33.2%) infants were BFAD. At baseline, African American, Medicaid-insured, and single mothers had lower odds of BFAD. The overall maximum volume of FS per feed was 11.0 mL/kg (interquartile range 8.0-14.4). With each hour of delay in first supplementation, the odds of BFAD increased by 2.8% (95% confidence interval [CI] 0.022, 0.035). With every 1 mL/kg increase in the first formula volume, subsequent supplementation frequency increased by 4.5%. A positive association was observed between BFAD and a lower rate of supplementation (cutoff value ≤35.1%). However, among infants with these lower rates of supplementation, each unit increase in maximum FS, from 2 to 15 mL/kg, decreased the probability of BFAD by 4.2% (3.6-4.7%). Additionally, we observed that infants who were given at least one EBM supplementation (n = 223; 7.2%) had substantially increased rates of BFAD (odds ratio [OR] = 9.8, 95% CI 7.2-13.3). CONCLUSION: Early and higher volumes of FS negatively impacted BFAD. Birthweight-based FS of feeding with physiological volumes may increase breastfeeding rates at discharge. KEY POINTS: · Higher volumes of first supplementation increases subsequent supplementation frequency.. · For each unit increase in maximum supplementation, BFAD probability decreases by 4.2%.. · Even one EBM supplementation increases rates of BFAD..

11.
Front Pediatr ; 11: 1245947, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37705598

RESUMEN

Objective: This study investigates whether volumes of intake in the first 24 h of life (24 HOL), in relation to birth weight (BW) and gestational age (GA), impact neonatal feeding intolerance (FI). Methods: This study employed a retrospective chart review of 6,650 infants born at ≥35 weeks. The volumes of each formula feed per kg BW in the first 24 HOL were assessed. FI was defined as evidenced by chart documentation of emesis, abdominal distension, abdominal x-ray, and/or switching to a sensitive formula. Results: Overall, the maximum volume of formula intake per feed was inversely correlated with GA and was higher in infants with FI (ß = -1.39, p < 0.001) compared with infants without FI (ß = -1.28, p < 0.001). The odds of emesis in late preterm infants with first feeding of >8 ml/kg [adjusted odds ratio (AOR) = 2.5, 95% confidence interval (CI): 1.4-4.6] and formula switching in the exclusively formula-fed group with volumes >10.5 ml/kg [AOR = 2.2, 95% CI (1.8-2.6)] were high. In the breastfeeding group, the odds of FI increased by 2.8-, 4.6-, and 5.2-fold with 5-10, 10-15, and >15 ml/kg of supplementations, respectively. Conclusion: A higher volume of intake in relation to BW often exceeds the physiological stomach capacity of newborns and is associated with early FI. Optimizing early feeding volumes based on infant BW and GA may decrease FI, which may be an issue of volume intolerance.

12.
Cells ; 12(18)2023 Sep 08.
Artículo en Inglés | MEDLINE | ID: mdl-37759454

RESUMEN

Angiogenesis plays a critical role in various physiological and pathological processes and is regulated by VEGF. Histone Deacetylase 6 (HDAC6) is a class IIB HDAC that regulates cytoplasmic signaling through deacetylation and is emerging as a target for modulating angiogenesis. We investigated the hypothesis that VEGF-induced endothelial cell (EC) NOTCH signaling is regulated by HDAC6 through acetylation of NOTCH intracellular cytoplasmic domain (NICD). In pulmonary endothelial cells (EC), VEGF-induced activation of the NICD transcriptional response was regulated by ERK1/2 and ADAM 17 and required DLL4. While HDAC6 inhibition induced the acetylation of NICD and stabilized NICD, it repressed NICD-SNW1 binding required for the NOTCH transcriptional responses. In vitro experiments showed that HDAC6 inhibition inhibited lung EC angiogenesis, and neonatal mice treated with a systemic HDAC6 inhibitor had significantly altered angiogenesis and alveolarization. These findings shed light on the role of HDAC6 in modulating VEGF-induced angiogenesis through acetylation and repression of the transcriptional regulators, NICD and SNW1.

13.
Microorganisms ; 11(6)2023 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-37374984

RESUMEN

Peripartum antibiotics can negatively impact the developing gut microbiome and are associated with necrotizing enterocolitis (NEC). The mechanisms by which peripartum antibiotics increase the risk of NEC and strategies that can help mitigate this risk remain poorly understood. In this study, we determined mechanisms by which peripartum antibiotics increase neonatal gut injury and evaluated whether probiotics protect against gut injury potentiated by peripartum antibiotics. To accomplish this objective, we administered broad-spectrum antibiotics or sterile water to pregnant C57BL6 mice and induced neonatal gut injury to their pups with formula feeding. We found that pups exposed to antibiotics had reduced villus height, crypt depth, and intestinal olfactomedin 4 and proliferating cell nuclear antigen compared to the controls, indicating that peripartum antibiotics impaired intestinal proliferation. When formula feeding was used to induce NEC-like injury, more severe intestinal injury and apoptosis were observed in the pups exposed to antibiotics compared to the controls. Supplementation with the probiotic Lactobacillus rhamnosus GG (LGG) reduced the severity of formula-induced gut injury potentiated by antibiotics. Increased intestinal proliferating cell nuclear antigen and activation of the Gpr81-Wnt pathway were noted in the pups supplemented with LGG, suggesting partial restoration of intestinal proliferation by probiotics. We conclude that peripartum antibiotics potentiate neonatal gut injury by inhibiting intestinal proliferation. LGG supplementation decreases gut injury by activating the Gpr81-Wnt pathway and restoring intestinal proliferation impaired by peripartum antibiotics. Our results suggest that postnatal probiotics may be effective in mitigating the increased risk of NEC associated with peripartum antibiotic exposure in preterm infants.

14.
EMBO Rep ; 24(7): e56030, 2023 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-37154714

RESUMEN

Intestinal ischemia underlies several clinical conditions and can result in the loss of the intestinal mucosal barrier. Ischemia-induced damage to the intestinal epithelium is repaired by stimulation of intestinal stem cells (ISCs), and paracrine signaling from the vascular niche regulates intestinal regeneration. Here, we identify FOXC1 and FOXC2 as essential regulators of paracrine signaling in intestinal regeneration after ischemia-reperfusion (I/R) injury. Vascular endothelial cell (EC)- and lymphatic EC (LEC)-specific deletions of Foxc1, Foxc2, or both in mice worsen I/R-induced intestinal damage by causing defects in vascular regrowth, expression of chemokine CXCL12 and Wnt activator R-spondin 3 (RSPO3) in blood ECs (BECs) and LECs, respectively, and activation of Wnt signaling in ISCs. Both FOXC1 and FOXC2 directly bind to regulatory elements of the CXCL12 and RSPO3 loci in BECs and LECs, respectively. Treatment with CXCL12 and RSPO3 rescues the I/R-induced intestinal damage in EC- and LEC-Foxc mutant mice, respectively. This study provides evidence that FOXC1 and FOXC2 are required for intestinal regeneration by stimulating paracrine CXCL12 and Wnt signaling.


Asunto(s)
Intestinos , Daño por Reperfusión , Ratones , Animales , Células Endoteliales/metabolismo , Vía de Señalización Wnt , Mucosa Intestinal , Daño por Reperfusión/genética , Daño por Reperfusión/metabolismo
15.
J Clin Invest ; 133(11)2023 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-37097753

RESUMEN

Neonatal herpes simplex virus (HSV) infection is a devastating disease with substantial morbidity and mortality. The genetic basis of susceptibility to HSV in neonates remains undefined. We evaluated a male infant with neonatal skin/eye/mouth (SEM) HSV-1 disease, who had complete recovery after acyclovir but developed HSV-1 encephalitis at 1 year of age. An immune workup showed an anergic PBMC cytokine response to TLR3 stimulation but no other TLRs. Exome sequencing identified rare missense variants in IFN-regulatory factor 7 (IRF7) and UNC-93 homolog B1 (UNC93B1). PBMC single-cell RNA-Seq done during childhood revealed decreased expression of several innate immune genes and a repressed TLR3 pathway signature at baseline in several immune cell populations, including CD14 monocytes. Functional studies in fibroblasts and human leukemia monocytic THP1 cells showed that both variants individually suppressed TLR3-driven IRF3 transcriptional activity and the type I IFN response in vitro. Furthermore, fibroblasts expressing the IRF7 and UNC93B1 variants had higher intracellular viral titers with blunting of the type I IFN response upon HSV-1 challenge. This study reports an infant with recurrent HSV-1 disease complicated by encephalitis associated with deleterious variants in the IRF7 and UNC93B1 genes. Our results suggest that TLR3 pathway mutations may predispose neonates to recurrent, severe HSV.


Asunto(s)
Encefalitis por Herpes Simple , Herpes Simple , Herpesvirus Humano 1 , Interferón Tipo I , Humanos , Lactante , Recién Nacido , Masculino , Encefalitis por Herpes Simple/genética , Herpes Simple/genética , Leucocitos Mononucleares/metabolismo , Proteínas de Transporte de Membrana , Receptor Toll-Like 3/genética
16.
Front Pediatr ; 11: 1112812, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36865691

RESUMEN

While prompt initiation of antibiotics at birth due to concerns for early onset sepsis is common, it often leads to many preterm infants being exposed to treatment despite negative blood cultures. Such exposure to early antibiotics can impact the developing gut microbiome putting infants at increased risk of several diseases. Necrotizing enterocolitis (NEC), a devastating inflammatory bowel disease that affects preterm infants, is among the most widely studied neonatal disease that has been linked to early antibiotics. While some studies have demonstrated an increased risk of NEC, other studies have demonstrated seemingly contrary findings of decreased NEC with early antibiotics. Studies using animal models have also yielded differing findings of benefit vs. harm of early antibiotic exposure on subsequent NEC susceptibility. We thus sought to conduct this narrative review to help clarify the relationship between early antibiotics exposure and future risk of NEC in preterm infants. Our objectives are to: (1) summarize findings from human and animal studies that investigated the relationship between early antibiotics and NEC, (2) highlight important limitations of these studies, (3) explore potential mechanisms that can explain why early antibiotics may increase or decrease NEC risk, and (4) identify future directions for research.

17.
Am J Physiol Lung Cell Mol Physiol ; 324(5): L722-L736, 2023 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-36976925

RESUMEN

SARS-CoV-2 viremia is associated with increased acute lung injury (ALI) and mortality in children and adults. The mechanisms by which viral components in the circulation mediate ALI in COVID-19 remain unclear. We tested the hypothesis that the SARS-CoV-2 envelope (E) protein induces Toll-like receptor (TLR)-mediated ALI and lung remodeling in a model of neonatal COVID-19. Neonatal C57BL6 mice given intraperitoneal E protein injections revealed a dose-dependent increase in lung cytokines [interleukin 6 (Il6), tumor necrosis factor (Tnfα), and interleukin 1 beta (Il1ß)] and canonical proinflammatory TLR signaling. Systemic E protein induced endothelial immune activation, immune cell influx, and TGFß signaling and lung matrix remodeling inhibited alveolarization in the developing lung. E protein-mediated ALI and transforming growth factor beta (TGFß) signaling was repressed in Tlr2-/-, but not Tlr4-/- mice. A single dose of intraperitoneal E protein injection induced chronic alveolar remodeling as evidenced by a decrease in radial alveolar counts and increase in mean linear intercepts. Ciclesonide, a synthetic glucocorticoid, inhibited E protein-induced proinflammatory TLR signaling and ALI. In vitro, E protein-mediated inflammation and cell death were TLR2-dependent in human primary neonatal lung endothelial cells and were rescued by ciclesonide. This study provides insight into the pathogenesis of ALI and alveolar remodeling with SARS-CoV-2 viremia in children, whereas revealing the efficacy of steroids.NEW & NOTEWORTHY We reveal that the envelope protein of SARS-CoV-2 mediates acute lung injury (ALI) and alveolar remodeling through Toll-like receptor activation, which is rescued by the glucocorticoid, ciclesonide.


Asunto(s)
Lesión Pulmonar Aguda , COVID-19 , Animales , Niño , Humanos , Ratones , Lesión Pulmonar Aguda/inducido químicamente , COVID-19/complicaciones , Células Endoteliales/metabolismo , Glucocorticoides , Lipopolisacáridos/efectos adversos , Ratones Endogámicos C57BL , SARS-CoV-2/metabolismo , Receptor Toll-Like 2 , Receptor Toll-Like 4/metabolismo , Receptores Toll-Like , Factor de Crecimiento Transformador beta , Viremia/complicaciones , Envoltura Viral/metabolismo
18.
Int J Mol Sci ; 24(4)2023 Feb 04.
Artículo en Inglés | MEDLINE | ID: mdl-36834525

RESUMEN

Bacterial infections are common in the etiology of human diseases owing to the ubiquity of bacteria. Such infections promote the development of periodontal disease, bacterial pneumonia, typhoid, acute gastroenteritis, and diarrhea in susceptible hosts. These diseases may be resolved using antibiotics/antimicrobial therapy in some hosts. However, other hosts may be unable to eliminate the bacteria, allowing them to persist for long durations and significantly increasing the carrier's risk of developing cancer over time. Indeed, infectious pathogens are modifiable cancer risk factors, and through this comprehensive review, we highlight the complex relationship between bacterial infections and the development of several cancer types. For this review, searches were performed on the PubMed, Embase, and Web of Science databases encompassing the entirety of 2022. Based on our investigation, we found several critical associations, of which some are causative: Porphyromonas gingivalis and Fusobacterium nucleatum are associated with periodontal disease, Salmonella spp., Clostridium perfringens, Escherichia coli, Campylobacter spp., and Shigella are associated with gastroenteritis. Helicobacter pylori infection is implicated in the etiology of gastric cancer, and persistent Chlamydia infections present a risk factor for the development of cervical carcinoma, especially in patients with the human papillomavirus (HPV) coinfection. Salmonella typhi infections are linked with gallbladder cancer, and Chlamydia pneumoniae infection is implicated in lung cancer, etc. This knowledge helps identify the adaptation strategies used by bacteria to evade antibiotic/antimicrobial therapy. The article also sheds light on the role of antibiotics in cancer treatment, the consequences of their use, and strategies for limiting antibiotic resistance. Finally, the dual role of bacteria in cancer development as well as in cancer therapy is briefly discussed, as this is an area that may help to facilitate the development of novel microbe-based therapeutics as a means of securing improved outcomes.


Asunto(s)
Antiinfecciosos , Infecciones Bacterianas , Proteínas de Escherichia coli , Gastroenteritis , Infecciones por Helicobacter , Helicobacter pylori , Neoplasias , Fiebre Tifoidea , Humanos , Infecciones Bacterianas/microbiología , Antibacterianos , Gastroenteritis/microbiología , Escherichia coli , Proteínas Portadoras
19.
Mucosal Immunol ; 16(2): 208-220, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36804483

RESUMEN

A key aspect of postnatal intestinal adaptation is the establishment of symbiotic relationships with co-evolved gut microbiota. Necrotizing enterocolitis (NEC) is the most severe disease arising from failure in postnatal gut adaptation in premature infants. Although pathological activation of intestinal Toll-like receptors (TLRs) is believed to underpin NEC pathogenesis, the mechanisms are incompletely understood. We postulate that unregulated aberrant TLR activation in NEC arises from a failure in intestinal-specific mechanisms that tamponade TLR signaling (the brakes). In this review, we discussed the human and animal studies that elucidate the developmental mechanisms inhibiting TLR signaling in the postnatal intestine (establishing the brakes). We then evaluate evidence from preclinical models and human studies that point to a defect in the inhibition of TLR signaling underlying NEC. Finally, we provided a framework for the assessment of NEC risk by screening for signatures of TLR signaling and for NEC prevention by TLR-targeted therapy in premature infants.


Asunto(s)
Enterocolitis Necrotizante , Enfermedades del Recién Nacido , Lactante , Animales , Recién Nacido , Humanos , Enterocolitis Necrotizante/etiología , Recien Nacido Prematuro , Receptores Toll-Like , Transducción de Señal
20.
Pediatr Pulmonol ; 58(3): 753-762, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36377273

RESUMEN

OBJECTIVE: To describe the survival rate, timing of liberation from the ventilator, and factors favorable for decannulation among infants with severe bronchopulmonary dysplasia (sBPD) who received tracheostomy. METHODS: Demographics and clinical outcomes were obtained through retrospective chart review of 98 infants with sBPD who were born between 2004 and 2017, received tracheostomy at <1 year of age, and were followed in the Infant Tracheostomy and Home Ventilator clinic up to 4 years of age. RESULTS: The number of infants with sBPD who received tracheostomy increased significantly over the study period. The median age at tracheostomy was 4 months (IQR 3, 5) or 43 weeks corrected gestational age; the median age at NICU discharge was 7 months (IQR 6, 9). At 48 months of age, all subjects had been liberated from the ventilator, at a median age of 24 months (IQR 18, 29); 52% had been decannulated with a median age at decannulation of 32 months (IQR 26, 39). Only 1 (1%) infant died. Multivariate logistic regression showed infants who were White, liberated from the ventilator by 24 months of age and have public insurance had significantly greater odds of being decannulated by 48 months of age. Tracheobronchomalacia was associated with decreased odds of decannulation. CONCLUSION: Infants with sBPD who received tracheostomy had an excellent survival rate. Liberation from home ventilation and decannulation are likely to occur by 4 years of age.


Asunto(s)
Displasia Broncopulmonar , Traqueostomía , Recién Nacido , Lactante , Humanos , Displasia Broncopulmonar/complicaciones , Estudios Retrospectivos , Respiración Artificial , Tasa de Supervivencia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...