Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 514
Filter
Add more filters

Publication year range
1.
Hum Mol Genet ; 33(20): 1800-1814, 2024 Oct 07.
Article in English | MEDLINE | ID: mdl-39128026

ABSTRACT

Spinal Muscular Atrophy with Respiratory Distress (SMARD1) is a lethal infantile disease, characterized by the loss of motor neurons leading to muscular atrophy, diaphragmatic paralysis, and weakness in the trunk and limbs. Mutations in IGHMBP2, a ubiquitously expressed DNA/RNA helicase, have been shown to cause a wide spectrum of motor neuron disease. Though mutations in IGHMBP2 are mostly associated with SMARD1, milder alleles cause the axonal neuropathy, Charcot-Marie-Tooth disease type 2S (CMT2S), and some null alleles are potentially a risk factor for sudden infant death syndrome (SIDS). Variant heterogeneity studied using an allelic series can be informative in order to create a broad spectrum of models that better exhibit the human variation. We previously identified the nmd2J mouse model of SMARD1, as well as two milder CMT2S mouse models. Here, we used CRISPR-Cas9 genome editing to create three new, more severe Ighmbp2 mouse models of SMARD1, including a null allele, a deletion of C495 (C495del) and a deletion of L362 (L362del). Phenotypic characterization of the IGHMBP2L362del homozygous mutants and IGHMBP2C495del homozygous mutants respectively show a more severe disease presentation than the previous nmd2J model. The IGHMBP2L362del mutants lack a clear denervation in the diaphragm while the IGHMBP2C495del mutants display a neurogenic diaphragmatic phenotype as observed in SMARD1 patients. Characterization of the Ighmbp2-null model indicated neo-natal lethality (median lifespan = 0.5 days). These novel strains expand the spectrum of SMARD1 models to better reflect the clinical continuum observed in the human patients with various IGHMBP2 recessive mutations.


Subject(s)
DNA-Binding Proteins , Disease Models, Animal , Muscular Atrophy, Spinal , Respiratory Distress Syndrome, Newborn , Transcription Factors , Animals , Mice , Muscular Atrophy, Spinal/genetics , Muscular Atrophy, Spinal/pathology , Muscular Atrophy, Spinal/physiopathology , DNA-Binding Proteins/genetics , Humans , Respiratory Distress Syndrome, Newborn/genetics , Respiratory Distress Syndrome, Newborn/pathology , Transcription Factors/genetics , Alleles , Mutation , CRISPR-Cas Systems , Gene Editing , Motor Neurons/pathology , Motor Neurons/metabolism , Phenotype
2.
Am J Physiol Lung Cell Mol Physiol ; 321(2): L291-L307, 2021 08 01.
Article in English | MEDLINE | ID: mdl-34132118

ABSTRACT

ATP-binding cassette class A3 (ABCA3) is a lipid transporter that plays a critical role in pulmonary surfactant function. The substitution of valine for glutamic acid at codon 292 (E292V) produces a hypomorphic variant that accounts for a significant portion of ABCA3 mutations associated with lung disorders spanning from neonatal respiratory distress syndrome and childhood interstitial lung disease to diffuse parenchymal lung disease (DPLD) in adults including pulmonary fibrosis. The mechanisms by which this and similar ABCA3 mutations disrupt alveolar type 2 (AT2) cell homeostasis and cause DPLD are largely unclear. The present study, informed by a patient homozygous for the E292V variant, used an in vitro and a preclinical murine model to evaluate the mechanisms by which E292V expression promotes aberrant lung injury and parenchymal remodeling. Cell lines stably expressing enhanced green fluorescent protein (EGFP)-tagged ABCA3 isoforms show a functional deficiency of the ABCA3E292V variant as a lipid transporter. AT2 cells isolated from mice constitutively homozygous for ABCA3E292V demonstrate the presence of small electron-dense lamellar bodies, time-dependent alterations in macroautophagy, and induction of apoptosis. These changes in AT2 cell homeostasis are accompanied by a spontaneous lung phenotype consisting of both age-dependent inflammation and fibrillary collagen deposition in alveolar septa. Older ABCA3E292V mice exhibit increased vulnerability to exogenous lung injury by bleomycin. Collectively, these findings support the hypothesis that the ABCA3E292V variant is a susceptibility factor for lung injury through effects on surfactant deficiency and impaired AT2 cell autophagy.


Subject(s)
ATP-Binding Cassette Transporters , Alveolar Epithelial Cells , Autophagy , Gene Expression Regulation , Lung Injury , Mutation, Missense , ATP-Binding Cassette Transporters/biosynthesis , ATP-Binding Cassette Transporters/genetics , Alveolar Epithelial Cells/metabolism , Alveolar Epithelial Cells/pathology , Amino Acid Substitution , Animals , Lung Injury/genetics , Lung Injury/metabolism , Lung Injury/pathology , Mice , Mice, Mutant Strains , Pulmonary Fibrosis/genetics , Pulmonary Fibrosis/metabolism , Pulmonary Fibrosis/pathology , Respiratory Distress Syndrome, Newborn/metabolism , Respiratory Distress Syndrome, Newborn/pathology
3.
Am J Physiol Lung Cell Mol Physiol ; 321(6): L1036-L1043, 2021 12 01.
Article in English | MEDLINE | ID: mdl-34585605

ABSTRACT

Invasive mechanical ventilation and oxygen toxicity are postnatal contributors to chronic lung disease of prematurity, also known as bronchopulmonary dysplasia (BPD). Cyfra 21-1 is a soluble fragment of cytokeratin 19, which belongs to the cytoskeleton stabilizing epithelial intermediate filaments. As a biomarker of structural integrity, Cyfra 21-1 might be associated with airway injury and lung hypoplasia in neonates. Serum Cyfra 21-1 concentrations for 80 preterm and 80 healthy term newborns were measured within 48 h after birth. Preterm infants with the combined endpoint BPD/mortality had significantly higher Cyfra 21-1 levels compared with those without fulfilling BPD/mortality criteria (P = 0.01). Also, severe RDS (>grade III) was associated with higher Cyfra levels (P = 0.01). Total duration of oxygen therapy was more than five times longer in neonates with high Cyfra 21-1 levels (P = 0.01). Infants with higher Cyfra 21-1 values were more likely to receive mechanical ventilation (50% vs. 17.5%). However, the duration of mechanical ventilation was similar between groups. The median Cyfra value was 1.93 ng/mL (IQR: 1.68-2.53 ng/mL) in healthy term neonates and 8.5 ng/mL (IQR: 3.6-16.0 ng/mL) in preterm infants. Using ROC analysis, we calculated a Cyfra cutoff > 8.5 ng/mL to predict BPD/death with an AUC of 0.795 (P = 0.004), a sensitivity of 88.9%, and a specificity of 55%. Mortality was predicted with a cutoff > 17.4 ng/mL (AUC: 0.94; P = 0.001), a sensitivity of 100%, and a specificity of 84%. These findings suggest that Cyfra 21-1 concentration might be useful to predict poor outcome in premature infants.


Subject(s)
Biomarkers/metabolism , Bronchopulmonary Dysplasia/mortality , Infant, Premature/growth & development , Keratin-19/metabolism , Respiration, Artificial/mortality , Respiratory Distress Syndrome, Newborn/mortality , Bronchopulmonary Dysplasia/metabolism , Bronchopulmonary Dysplasia/pathology , Bronchopulmonary Dysplasia/therapy , Case-Control Studies , Female , Humans , Infant, Newborn , Male , Prognosis , Respiratory Distress Syndrome, Newborn/metabolism , Respiratory Distress Syndrome, Newborn/pathology , Respiratory Distress Syndrome, Newborn/therapy , Survival Rate
4.
Am J Physiol Lung Cell Mol Physiol ; 320(2): L193-L204, 2021 02 01.
Article in English | MEDLINE | ID: mdl-33112186

ABSTRACT

Premature infants, especially those with bronchopulmonary dysplasia (BPD), develop recurrent severe respiratory viral illnesses. We have shown that hyperoxic exposure of immature mice, a model of BPD, increases lung IL-12-producing Clec9a+ CD103+ dendritic cells (DCs), pro-inflammatory responses, and airway hyperreactivity following rhinovirus (RV) infection. However, the requirement for CD103+ DCs and Clec9a, a DAMP receptor that binds necrotic cell cytoskeletal filamentous actin (F-actin), for RV-induced inflammatory responses has not been demonstrated. To test this, 2-day-old C57BL/6J, CD103+ DC-deficient Batf3-/- or Clec9agfp-/- mice were exposed to normoxia or hyperoxia for 14 days. Also, selected mice were treated with neutralizing antibody against CD103. Immediately after hyperoxia, the mice were inoculated with RV intranasally. We found that compared with wild-type mice, hyperoxia-exposed Batf3-/- mice showed reduced levels of IL-12p40, IFN-γ, and TNF-α, fewer IFN-γ-producing CD4+ T cells, and decreased airway responsiveness following RV infection. Similar effects were observed in anti-CD103-treated and Clec9agfp-/- mice. Furthermore, hyperoxia increased airway dead cell number and extracellular F-actin levels. Finally, studies in preterm infants with respiratory distress syndrome showed that tracheal aspirate CLEC9A expression positively correlated with IL12B expression, consistent with the notion that CLEC9A+ cells are responsible for IL-12 production in humans as well as mice. We conclude that CD103+ DCs and Clec9a are required for hyperoxia-induced pro-inflammatory responses to RV infection. In premature infants, Clec9a-mediated activation of CD103+ DCs may promote pro-inflammatory responses to viral infection, thereby driving respiratory morbidity.


Subject(s)
Antigens, CD/metabolism , Dendritic Cells/immunology , Hyperoxia/physiopathology , Integrin alpha Chains/metabolism , Lectins, C-Type/physiology , Lung/immunology , Pneumonia/immunology , Receptors, Immunologic/physiology , Respiratory Distress Syndrome, Newborn/immunology , Animals , Animals, Newborn , Antigens, CD/genetics , Basic-Leucine Zipper Transcription Factors/physiology , Female , Humans , Infant, Newborn , Infant, Premature/immunology , Integrin alpha Chains/genetics , Lung/metabolism , Lung/pathology , Lung/virology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Picornaviridae Infections/complications , Picornaviridae Infections/virology , Pneumonia/virology , Repressor Proteins/physiology , Respiratory Distress Syndrome, Newborn/metabolism , Respiratory Distress Syndrome, Newborn/pathology , Rhinovirus/isolation & purification
5.
Cell Mol Life Sci ; 77(17): 3351-3367, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32123965

ABSTRACT

Spinal muscular atrophy (SMA) with respiratory distress type 1 (SMARD1) is an autosomal recessive motor neuron disease that is characterized by distal and proximal muscle weakness and diaphragmatic palsy that leads to respiratory distress. Without intervention, infants with the severe form of the disease die before 2 years of age. SMARD1 is caused by mutations in the IGHMBP2 gene that determine a deficiency in the encoded IGHMBP2 protein, which plays a critical role in motor neuron survival because of its functions in mRNA processing and maturation. Although it is rare, SMARD1 is the second most common motor neuron disease of infancy, and currently, treatment is primarily supportive. No effective therapy is available for this devastating disease, although multidisciplinary care has been an essential element of the improved quality of life and life span extension in these patients in recent years. The objectives of this review are to discuss the current understanding of SMARD1 through a summary of the presently known information regarding its clinical presentation and pathogenesis and to discuss emerging therapeutic approaches. Advances in clinical care management have significantly extended the lives of individuals affected by SMARD1 and research into the molecular mechanisms that lead to the disease has identified potential strategies for intervention that target the underlying causes of SMARD1. Gene therapy via gene replacement or gene correction provides the potential for transformative therapies to halt or possibly prevent neurodegenerative disease in SMARD1 patients. The recent approval of the first gene therapy approach for SMA associated with mutations in the SMN1 gene may be a turning point for the application of this strategy for SMARD1 and other genetic neurological diseases.


Subject(s)
DNA-Binding Proteins/genetics , Muscular Atrophy, Spinal/pathology , Respiratory Distress Syndrome, Newborn/pathology , Transcription Factors/genetics , Animals , Cell- and Tissue-Based Therapy , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/metabolism , Genetic Therapy , Humans , Muscular Atrophy, Spinal/complications , Muscular Atrophy, Spinal/genetics , Muscular Atrophy, Spinal/therapy , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Neural Stem Cells/transplantation , Respiratory Distress Syndrome, Newborn/complications , Respiratory Distress Syndrome, Newborn/genetics , Respiratory Distress Syndrome, Newborn/therapy , Ribosomes/chemistry , Ribosomes/metabolism , Survival of Motor Neuron 1 Protein/genetics , Transcription Factors/chemistry , Transcription Factors/metabolism
6.
J Korean Med Sci ; 36(11): e81, 2021 Mar 22.
Article in English | MEDLINE | ID: mdl-33754511

ABSTRACT

BACKGROUND: Despite the advances in neonatology, the incidence of bronchopulmonary dysplasia (BPD) is increasing. It is important to prevent the development of BPD in the first place. The online BPD outcome estimator from National Institute of Children Health and Human Development and Neonatal Research Network is available. However, it is not applicable for Asians. Moreover, limits are set for birth weight and gestational weeks excluding those who may still have BPD. The aim of this study was to develop a prediction model for BPD using first hour perinatal and neonatal factors in Korean very low birth weight infants (VLBWIs). METHODS: Data were collected for 8,022 VLBWIs with gestational age (GA) ≥ 22 weeks who were born between January 1, 2013 and December 31, 2016, and admitted to the neonatal intensive care units of the KNN. Multiple logistic regression models reanalyzed by stepwise selection with significant clinical indicators for BPD. PROC package was used to calculate the area under curve (AUC) and corresponding 95% confidence intervals. Moreover, it was used to search the best cut-off value. External validation was performed with the 2017 Korean neonatal network (KNN) data. RESULTS: After all missing data were excluded, 4,600 VLBWIs were included in the training dataset of the prediction model. Predictability of presence of BPD was 90.8% and prediction P value cut off was 0.550. Five-minute Apgar score, birth weight, GA, sex, surfactant use were significant indicators. Predictability of severe BPD was 81.5% and prediction P value cut off was 0.160. Five-minute Apgar score, birth weight, maternal PIH, chronic maternal hypertension, GA, sex, respiratory distress syndrome, need of resuscitation at birth were significant indicators. After external validation, sensitivity and specificity did not change significantly. CONCLUSION: From this study, high predictability was obtained using clinical parameters obtained within one hour of life. P value for prediction of each grade of BPD and equation for calculation was presented. It can be helpful for the early prediction of BPD in Korean VLBWI. This study will contribute to the prediction of BPD in Asians especially Korean VLBWIs, not currently included in the NICHD BPD online BPD predictor. In addition, the predictive power may be continuously increased with the cumulative data of KNN.


Subject(s)
Bronchopulmonary Dysplasia/diagnosis , Infant, Very Low Birth Weight , Algorithms , Apgar Score , Area Under Curve , Body Temperature , Bronchopulmonary Dysplasia/pathology , Databases, Factual , Female , Gestational Age , Humans , Infant, Newborn , Infant, Premature , Logistic Models , Male , Predictive Value of Tests , Pulmonary Surfactants/therapeutic use , ROC Curve , Registries , Respiratory Distress Syndrome, Newborn/drug therapy , Respiratory Distress Syndrome, Newborn/pathology , Time Factors
7.
J Cell Mol Med ; 24(2): 1169-1178, 2020 01.
Article in English | MEDLINE | ID: mdl-31802621

ABSTRACT

Spinal muscular atrophy with respiratory distress type 1 (SMARD1) is a rare autosomal recessive neuromuscular disorder caused by mutations in the IGHMBP2 gene, which encodes immunoglobulin µ-binding protein 2, leading to progressive spinal motor neuron degeneration. We review the data available in the literature about SMARD1. The vast majority of patients show an onset of typical symptoms in the first year of life. The main clinical features are distal muscular atrophy and diaphragmatic palsy, for which permanent supportive ventilation is required. No effective treatment is available yet, but novel therapeutic approaches, such as gene therapy, have shown encouraging results in preclinical settings and thus represent possible methods for treating SMARD1. Significant advancements in the understanding of both the SMARD1 clinical spectrum and its molecular mechanisms have allowed the rapid translation of preclinical therapeutic strategies to human patients to improve the poor prognosis of this devastating disease.


Subject(s)
DNA-Binding Proteins/genetics , Molecular Targeted Therapy , Muscular Atrophy, Spinal/pathology , Muscular Atrophy, Spinal/therapy , Mutation , Respiratory Distress Syndrome, Newborn/pathology , Respiratory Distress Syndrome, Newborn/therapy , Transcription Factors/genetics , Animals , Humans , Muscular Atrophy, Spinal/genetics , Prognosis , Respiratory Distress Syndrome, Newborn/genetics
8.
Hum Mol Genet ; 27(4): 679-690, 2018 02 15.
Article in English | MEDLINE | ID: mdl-29272405

ABSTRACT

Spinal muscular atrophy with respiratory distress type 1 (SMARD1) is an autosomal recessive motor neuron disease causing distal limb muscle atrophy that progresses proximally and is accompanied by diaphragmatic paralysis. Neuromuscular junction (NMJ) alterations have been reported in muscles of SMARD1 model mice, known as nmd mice, with varying degrees of severity, suggesting that different muscles are specifically and selectively resistant or susceptible to denervation. To evaluate the extent of NMJ pathology in a broad range of muscles, a panel of axial and appendicular muscles were isolated and immunostained from nmd mice. These analyses revealed that selective distal appendage muscles were highly vulnerable to denervation. Susceptibility to pathology was not limited to NMJ alterations, but included defects in myelination within those neurons innervating susceptible muscles. Interestingly, end plate fragmentation was present within all muscles independent of the extent of NMJ alterations, suggesting that end plate fragmentation is an early hallmark of SMARD1 pathogenesis. Expressing the full-length IGHMBP2 cDNA using an adeno-associated virus (AAV9) significantly decreased all aspects of muscle and nerve disease pathology. These results shed new light onto the pathogenesis of SMARD1 by identifying specific motor units that are resistant and susceptible to neurodegeneration in an important model of SMARD1.


Subject(s)
Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscular Atrophy, Spinal/metabolism , Neuromuscular Junction/metabolism , Respiratory Distress Syndrome, Newborn/metabolism , Animals , DNA-Binding Proteins/metabolism , Immunohistochemistry , Male , Mice , Motor Neurons/metabolism , Motor Neurons/pathology , Muscular Atrophy, Spinal/pathology , Neuromuscular Junction/pathology , Neurons/metabolism , Respiratory Distress Syndrome, Newborn/pathology , Transcription Factors/metabolism
9.
Hum Genet ; 139(10): 1273-1283, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32367404

ABSTRACT

Unlike disorders of primary cilium, primary ciliary dyskinesia (PCD) has a much narrower clinical spectrum consistent with the limited tissue distribution of motile cilia. Nonetheless, PCD diagnosis can be challenging due to the overlapping features with other disorders and the requirement for sophisticated tests that are only available in specialized centers. We performed exome sequencing on all patients with a clinical suspicion of PCD but for whom no nasal nitric oxide test or ciliary functional assessment could be ordered. Among 81 patients (56 families), in whom PCD was suspected, 68% had pathogenic or likely pathogenic variants in established PCD-related genes that fully explain the phenotype (20 variants in 11 genes). The major clinical presentations were sinopulmonary infections (SPI) (n = 58), neonatal respiratory distress (NRD) (n = 2), laterality defect (LD) (n = 6), and combined LD/SPI (n = 15). Biallelic likely deleterious variants were also encountered in AKNA and GOLGA3, which we propose as novel candidates in a lung phenotype that overlaps clinically with PCD. We also encountered a PCD phenocopy caused by a pathogenic variant in ITCH, and a pathogenic variant in CEP164 causing Bardet-Biedl syndrome and PCD presentation as a very rare example of the dual presentation of these two disorders of the primary and motile cilia. Exome sequencing is a powerful tool that can help "democratize" the diagnosis of PCD, which is currently limited to highly specialized centers.


Subject(s)
Cilia/metabolism , Ciliary Motility Disorders/genetics , Genetic Predisposition to Disease , Pneumonia/genetics , Respiratory Distress Syndrome, Newborn/genetics , Sinusitis/genetics , Autoantigens/genetics , Cilia/pathology , Ciliary Motility Disorders/complications , Ciliary Motility Disorders/diagnosis , Ciliary Motility Disorders/pathology , Consanguinity , DNA-Binding Proteins/genetics , Female , Gene Expression , Golgi Matrix Proteins/genetics , Humans , Male , Microtubule Proteins/genetics , Mutation , Nuclear Proteins/genetics , Pedigree , Phenotype , Pneumonia/complications , Pneumonia/diagnosis , Pneumonia/pathology , Repressor Proteins/genetics , Respiratory Distress Syndrome, Newborn/complications , Respiratory Distress Syndrome, Newborn/diagnosis , Respiratory Distress Syndrome, Newborn/pathology , Respiratory Mucosa/metabolism , Respiratory Mucosa/pathology , Saudi Arabia , Sinusitis/complications , Sinusitis/diagnosis , Sinusitis/pathology , Transcription Factors/genetics , Ubiquitin-Protein Ligases/genetics , Exome Sequencing
10.
Med Sci Monit ; 26: e922281, 2020 Mar 31.
Article in English | MEDLINE | ID: mdl-32231178

ABSTRACT

BACKGROUND Acute respiratory distress syndrome (ARDS) is a sudden and serious disease with increasing morbidity and mortality rates. Phosphodiesterase 4 (PDE4) is a novel target for inflammatory disease, and ibudilast (IBU), a PDE4 inhibitor, inhibits inflammatory response. Our study investigated the effect of IBU on the pathogenesis of neonatal ARDS and the underlying mechanism related to it. MATERIAL AND METHODS Western blotting was performed to analyze the expression levels of PDE4, CXCR4, SDF-1, CXCR5, CXCL1, inflammatory cytokines, and proteins related to cell apoptosis. Hematoxylin-eosin staining was performed to observe the pathological morphology of lung tissue. Pulmonary edema score was used to assess the degree of lung water accumulation after pulmonary injury. Enzyme-linked immunosorbent assay (ELISA) was used to assess levels of inflammatory factors (TNF-alpha, IL-1ß, IL-6, and MCP-1) in serum. TUNEL assay was used to detect apoptotic cells. RESULTS Increased expression of PDE4 was observed in an LPS-induced neonatal ARDS mouse model, and IBU ameliorated LPS-induced pathological manifestations and pulmonary edema in lung tissue. In addition, IBU attenuated the secretion of inflammatory cytokines by inactivating the chemokine axis in the LPS-induced neonatal ARDS mouse model. Finally, IBU significantly reduced LPS-induced cell apoptosis in lung tissue. CONCLUSIONS IBU, a PDE4 inhibitor, protected against ARDS by interfering with pulmonary inflammation and apoptosis. Our findings provide a novel and promising strategy to regulate pulmonary inflammation in ARDS.


Subject(s)
Cyclic Nucleotide Phosphodiesterases, Type 4/metabolism , Inflammation/drug therapy , Phosphodiesterase 4 Inhibitors/pharmacology , Pyridines/pharmacology , Respiratory Distress Syndrome, Newborn/drug therapy , Animals , Animals, Newborn , Apoptosis/drug effects , Apoptosis/immunology , Bronchoalveolar Lavage Fluid , Disease Models, Animal , Humans , Inflammation/diagnosis , Inflammation/immunology , Inflammation/pathology , Injections, Intraperitoneal , Lipopolysaccharides/immunology , Lung/drug effects , Lung/immunology , Lung/pathology , Mice , Phosphodiesterase 4 Inhibitors/therapeutic use , Pyridines/therapeutic use , Respiratory Distress Syndrome, Newborn/diagnosis , Respiratory Distress Syndrome, Newborn/immunology , Respiratory Distress Syndrome, Newborn/pathology , Signal Transduction/drug effects , Signal Transduction/immunology
11.
Biochem Biophys Res Commun ; 516(1): 313-319, 2019 08 13.
Article in English | MEDLINE | ID: mdl-31256932

ABSTRACT

Spinal muscular atrophy with respiratory distress type 1 (SMARD1) is an autosomal recessive disease that causes distal limb muscle atrophy, due to motor neuron degeneration. Similar to other motor neuron diseases, SMARD1 shows differential vulnerability to denervation in various muscle groups, which is recapitulated in the nmd mouse, a model of SMARD1. In multiple neurodegenerative disease models, transcriptomic analysis has identified differentially expressed genes between vulnerable motor neuron populations, but the mechanism leading to susceptibility is largely unknown. To investigate if denervation vulnerability is linked to intrinsic muscle properties, we analyzed muscle fiber-type composition in muscles from motor units that show different degrees of denervation in nmd mice: gastrocnemius, tibialis anterior (TA), and extensor digitorum longus (EDL). Our results revealed that denervation vulnerability correlated with atrophy and loss of MyHC-IIb and MyHC-IIx muscle fiber types. Interestingly, increased vulnerability also correlated with an increased abundance of MyHC-I and MyHC-IIa muscle fibers. These results indicated that MyHC-IIx muscle fibers are the most vulnerable to denervation, followed by MyHC-IIb muscle fibers. Moreover, our data indicate that type MyHC-IIa and MyHC-IIb muscle fibers show resistance to denervation and compensate for the loss of MyHC-IIx and MyHC-IIb muscle fibers in the most vulnerable muscles. Taken together these results provide a basis for the selective vulnerability to denervation of specific muscles in nmd mice and identifies new targets for potential therapeutic intervention.


Subject(s)
Muscle Fibers, Skeletal/pathology , Muscular Atrophy, Spinal/pathology , Respiratory Distress Syndrome, Newborn/pathology , Animals , Disease Models, Animal , Mice , Motor Neurons/pathology , Muscle, Skeletal/innervation , Muscle, Skeletal/pathology , Myosin Heavy Chains/analysis
12.
Med Sci Monit ; 25: 4992-4998, 2019 Jul 06.
Article in English | MEDLINE | ID: mdl-31278248

ABSTRACT

BACKGROUND This study assessed the clinical characteristics of neonatal acute respiratory distress syndrome (ARDS) and differences in therapy in comparison to RDS. MATERIAL AND METHODS The clinical data of 925 preterm infants with respiratory distress were collected and divided into 4 groups. Group A and B both met the diagnosis of neonatal RDS, whereas infants in group B also showed inflammatory response. Group C met the Montreux definition of neonatal ARDS and group D was the control. RESULTS We found that 73.50% of the 925 preterm infants were diagnosed with RDS, of which RDS with inflammatory response accounted for 42.05%. ARDS accounted for 5.29% and control group accounted for 21.19%. Group C infants were the heaviest (2168.16±654.43 g) and had the oldest gestational age. The pregnancy-induced hypertension was highest (30.07%) in group B and lowest in group D (13.26%). Group C had higher iNO and longer invasive ventilator times, but had less frequent surfactant treatment, as well as shorter oxygen time and hospital stay. Group B had significantly longer invasive ventilator use than in Group A. In group A, PDA, ROP, and PPHN were the most common complications, with morbidity rates at 78.35%, 8.4%, and 25.77%, respectively, while group C had higher incidence of PDA (71.42%) and coagulation disorders (38.77%). CONCLUSIONS ARDS mainly occurs in late preterm infants. Its treatment is dependent on iNO and invasive ventilator-assisted therapy, and the surfactant treatment rate was relatively lower in comparison to RDS. RDS accompanied with inflammatory response is also dependent on prolonged use of an invasive ventilator.


Subject(s)
Respiratory Distress Syndrome, Newborn/pathology , Respiratory Distress Syndrome, Newborn/therapy , Respiratory Distress Syndrome/pathology , Respiratory Distress Syndrome/therapy , Female , Humans , Infant, Newborn , Male , Respiratory Distress Syndrome, Newborn/complications , Retrospective Studies
13.
J Korean Med Sci ; 34(25): e175, 2019 Jul 01.
Article in English | MEDLINE | ID: mdl-31243934

ABSTRACT

Neonatal respiratory distress syndrome (RDS) is a disease that is unique to newborn infants. It is caused by a deficiency of pulmonary surfactant (PS), which is usually ready to be activated around the perinatal period. Until RDS was more clearly understood, it was not known why premature infants died from respiratory failure, although pathology revealed hyaline membranes in the alveoli. Surprisingly, the era of PS replacement therapy began only relatively recently. The first clinical trial investigating neonatal RDS was conducted in 1980. Since then, newborn survival has improved dramatically, which has led to significant advances in the field of neonatology. The present comprehensive review addresses PS, from its discovery to the application of artificial PS in newborns with RDS. It also reviews the history of PS in Korea, including its introduction, various commercial products, present and past research, newborn registries, and health insurance issues. Finally, it describes the inception of the Korean Society of Neonatology and future directions of research and treatment.


Subject(s)
Pulmonary Surfactants/therapeutic use , Respiratory Distress Syndrome, Newborn/drug therapy , History of Medicine , Humans , Infant, Very Low Birth Weight , Premature Birth , Republic of Korea , Respiratory Distress Syndrome, Newborn/pathology
14.
Niger J Clin Pract ; 22(3): 298-304, 2019 Mar.
Article in English | MEDLINE | ID: mdl-30837415

ABSTRACT

BACKGROUND: Intraventricular hemorrhage (IVH) is a severe complication among preterm neonates which can result in hydrocephalus, cerebral palsy, behavioural disorders, learning disabilities, or death. It is important to identify the factors associated with IVH in order to prevent these neurological consequences and reduce the resultant burden of neurological disease. Aim: To determine the factors associated with IVH among preterm neonates. DESIGN: The study was prospective cross-sectional in design. SUBJECTS AND METHODS: Ninety-nine preterm neonates who were < 37 completed weeks of gestation were recruited consecutively from the Special Care Baby Unit of a Tertiary Hospital. Transfontanelle ultrasonography was used to detect IVH and the factors associated with IVH were classified into: neonatal, maternal (prenatal), and clinical factors. Data were analyzed using SPSS version 16.0 for windows. Chi-squared test and Fisher's exact probability test were used as appropriate. The level of significance was set at P < 0.05. The association between these factors and IVH was evaluated by univariate and multivariate logistic regression analyses. RESULTS: Among the 99 preterm neonates studied, 36 (36.4%) of them were between 28 and 31 weeks of gestation, whereas 63 (63.6%) were between 32 and 36 weeks of gestation. In univariate analysis, the factors found to be associated with IVH were lower gestational age <32 weeks gestation, low Apgar score of <3 in 1 and 5 min, respectively, outborn status of neonates, lower social class, need for respiratory support, and blood transfusion. However, the lower gestational age (odds ratio [OR]: 10.9, 95% confidence interval [CI]:1.95-61.04) and respiratory support (continuous positive airway pressure (CPAP)) [OR: 52.24; CI: 3.40-721.84] were retained as significant predictors of IVH in the multivariate logistic regression model. CONCLUSION: The lower gestational age and respiratory support (CPAP) are independent predictors for IVH. Prevention of preterm delivery and improvement in interventions of neonatal care (CPAP) are necessary to prevent the risk for IVH especially in the early preterm neonates.


Subject(s)
Cerebral Hemorrhage/diagnostic imaging , Cerebral Hemorrhage/etiology , Gestational Age , Infant, Premature, Diseases/etiology , Infant, Premature , Ultrasonography , Cerebral Hemorrhage/epidemiology , Cross-Sectional Studies , Female , Hospitals, Teaching , Humans , Infant, Newborn , Infant, Premature, Diseases/pathology , Male , Nigeria , Pregnancy , Prevalence , Prospective Studies , Respiratory Distress Syndrome, Newborn/etiology , Respiratory Distress Syndrome, Newborn/pathology
15.
Am J Physiol Lung Cell Mol Physiol ; 314(1): L32-L53, 2018 01 01.
Article in English | MEDLINE | ID: mdl-28860142

ABSTRACT

The biological and immune-protective properties of surfactant-derived phospholipids and phospholipid subfractions in the context of neonatal inflammatory lung disease are widely unknown. Using a porcine neonatal triple-hit acute respiratory distress syndrome (ARDS) model (repeated airway lavage, overventilation, and LPS instillation into airways), we assessed whether the supplementation of surfactant (S; poractant alfa) with inositol derivatives [inositol 1,2,6-trisphosphate (IP3) or phosphatidylinositol 3,5-bisphosphate (PIP2)] or phosphatidylglycerol subfractions [16:0/18:1-palmitoyloleoyl-phosphatidylglycerol (POPG) or 18:1/18:1-dioleoyl-phosphatidylglycerol (DOPG)] would result in improved clinical parameters and sought to characterize changes in key inflammatory pathways behind these improvements. Within 72 h of mechanical ventilation, the oxygenation index (S+IP3, S+PIP2, and S+POPG), the ventilation efficiency index (S+IP3 and S+POPG), the compliance (S+IP3 and S+POPG) and resistance (S+POPG) of the respiratory system, and the extravascular lung water index (S+IP3 and S+POPG) significantly improved compared with S treatment alone. The inositol derivatives (mainly S+IP3) exerted their actions by suppressing acid sphingomyelinase activity and dependent ceramide production, linked with the suppression of the inflammasome nucleotide-binding domain, leucine-rich repeat-containing protein-3 (NLRP3)-apoptosis-associated speck-like protein containing a caspase recruitment domain (ASC)-caspase-1 complex, and the profibrotic response represented by the cytokines transforming growth factor-ß1 and IFN-γ, matrix metalloproteinase (MMP)-1/8, and elastin. In addition, IκB kinase activity was significantly reduced. S+POPG and S+DOPG treatment inhibited polymorphonuclear leukocyte activity (MMP-8 and myeloperoxidase) and the production of interleukin-6, maintained alveolar-capillary barrier functions, and reduced alveolar epithelial cell apoptosis, all of which resulted in reduced pulmonary edema. S+DOPG also limited the profibrotic response. We conclude that highly concentrated inositol derivatives and phosphatidylglycerol subfractions in surfactant preparations mitigate key inflammatory pathways in inflammatory lung disease and that their clinical application may be of interest for future treatment of the acute exudative phase of neonatal ARDS.


Subject(s)
Disease Models, Animal , Inositol/pharmacology , Phosphatidylglycerols/pharmacology , Pulmonary Edema/drug therapy , Pulmonary Surfactants/pharmacology , Respiratory Distress Syndrome, Newborn/drug therapy , Animals , Animals, Newborn , Apoptosis , Bronchoalveolar Lavage Fluid , Cytokines/genetics , Cytokines/metabolism , Female , Humans , Male , NF-kappa B/genetics , NF-kappa B/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Pulmonary Edema/metabolism , Pulmonary Edema/pathology , Pulmonary Gas Exchange , Random Allocation , Respiration, Artificial , Respiratory Distress Syndrome, Newborn/metabolism , Respiratory Distress Syndrome, Newborn/pathology , Swine , Translational Research, Biomedical , Vitamin B Complex/pharmacology
16.
Scand J Clin Lab Invest ; 78(5): 352-357, 2018 Sep.
Article in English | MEDLINE | ID: mdl-29786454

ABSTRACT

Preterm neonates with respiratory distress syndrome (RDS) are at increased risk of acute kidney injury (AKI). Our study aimed at determining whether serum cystatin C (sCysC) on day 3 of life (D3) can early predict AKI in preterm neonates with RDS. This prospective study was conducted on 75 preterm neonates; 50 with RDS and 25 without RDS. On D3, sCysC, serum creatinine (sCr) and blood urea nitrogen (BUN) were measured and estimated glomerular filtration rate (eGFR) was calculated. sCr and BUN levels were measured again on days 5 and 7. Neonates were evaluated for development of AKI during first week of life according to the modified pediatric RIFLE (pRIFLE) criteria. Thirteen neonates with RDS developed AKI (26%).There was no significant difference between RDS and control groups with respect to sCysC. RDS neonates with AKI had significantly higher sCysC than those without AKI (1.62 ± 0.12 versus 1.16 ± 0.09 mg/l; p < .001). RDS grade III-IV neonates had significantly higher sCysC than RDS grade I-II. There was a significant positive correlation between D3 sCysC and (D5 and D7 sCr and BUN). Receiver operating characteristic (ROC) curve showed that D3 sCysC can predict AKI in preterm neonates with RDS at a cutoff point of >1.3 mg/l with sensitivity of 92.30% and specificity of 96%. We conclude that neonates with RDS are at increased risk of AKI. sCysC on day 3 of life can predict AKI earlier than Cr and eGFR.


Subject(s)
Acute Kidney Injury/diagnosis , Creatinine/blood , Cystatin C/blood , Respiratory Distress Syndrome, Newborn/diagnosis , Acute Kidney Injury/blood , Acute Kidney Injury/etiology , Acute Kidney Injury/pathology , Biomarkers/blood , Blood Urea Nitrogen , Female , Glomerular Filtration Rate , Humans , Infant, Newborn , Infant, Premature , Male , Prospective Studies , ROC Curve , Respiratory Distress Syndrome, Newborn/blood , Respiratory Distress Syndrome, Newborn/complications , Respiratory Distress Syndrome, Newborn/pathology , Severity of Illness Index
17.
Rev Chil Pediatr ; 88(2): 280-284, 2017 Apr.
Article in Spanish | MEDLINE | ID: mdl-28542663

ABSTRACT

Adequate pupil dilation is needed to evaluate some neonates at risk of developing illness during this stage. However, this procedure is not free of adverse effects, either local or systemic. One of these complications is the local vasoconstriction of the preterm baby’s skin following the application of mydriatic eye drops. OBJECTIVE: To describe secondary local and systemic complications of pharmacological pupil dilation in 2 newborns. Clinical case 1: Full term baby with diagnosis of low-birth weight and hydrocephalia. An ophtalmological evaluation was performed at 5 days of age due to the presence of corneal opacities. Peri ocular pallor was observed during the procedure, as well as tachycardia and hypertension 2 hours later, spontaneosly recovered. Case 2: Preterm newborn, 27 weeks of gestational age. Neonatal respiratory distress syndrome, patent ductus arteriosus, intraventricular hemorrhage and hydrocephalia were diagnosed at birth. At 28 days of life an ophtalmological evaluation was performed. After 10 minutes of mydriatic drops administration to evaluate preterm retinopathy, peri ocular pallor was observed, with spontaneous resolution; however, 24 hours later, the patient showed abdominal distention and feeding intolerance. Necrotizing enterocolitis was discarded, and symptoms were spontaneosly recovered. CONCLUSION: The establishment of protocols in relation to the number of drops to apply for dilation is needed to reduce deleterious effects on high risk infants, such as premature babies and those with hydrocephalus. Therefore this monitoring practice should be performed during the evaluation.


Subject(s)
Hydrocephalus/physiopathology , Mydriatics/administration & dosage , Vasoconstriction/drug effects , Ductus Arteriosus, Patent/pathology , Humans , Infant, Low Birth Weight , Infant, Newborn , Mydriatics/adverse effects , Respiratory Distress Syndrome, Newborn/pathology
18.
Am J Obstet Gynecol ; 215(3): 384.e1-384.e89, 2016 09.
Article in English | MEDLINE | ID: mdl-27143398

ABSTRACT

BACKGROUND: Neonatal respiratory distress syndrome in preterm infants is a leading cause of neonatal death. Pulmonary insufficiency-related infant mortality rates have improved with antenatal glucocorticoid treatment and neonatal surfactant replacement. However, the mechanism of glucocorticoid-promoted fetal lung maturation is not understood fully, despite decades of clinical use. We previously have shown that genetic deletion of Erk3 in mice results in growth restriction, cyanosis, and early neonatal lethality because of pulmonary immaturity and respiratory distress. Recently, we demonstrated that the addition of postnatal surfactant administration to antenatal dexamethasone treatment resulted in enhanced survival of neonatal Erk3-null mice. OBJECTIVE: To better understand the molecular underpinnings of corticosteroid-mediated lung maturation, we used high-throughput transcriptomic and high-resolution morphologic analysis of the murine fetal lung. We sought to examine the alterations in fetal lung structure and function that are associated with neonatal respiratory distress and antenatal glucocorticoid treatment. STUDY DESIGN: Dexamethasone (0.4 mg/kg) or saline solution was administered to pregnant dams on embryonic days 16.5 and 17.5. Fetal lungs were collected and analyzed by microCT and RNA-seq for differential gene expression and pathway interactions with genotype and treatment. Results from transcriptomic analysis guided further investigation of candidate genes with the use of immunostaining in murine and human fetal lung tissue. RESULTS: Erk3(-/-) mice exhibited atelectasis with decreased overall porosity and saccular space relative to wild type, which was ameliorated by glucocorticoid treatment. Of 596 differentially expressed genes (q < 0.05) that were detected by RNA-seq, pathway analysis revealed 36 genes (q < 0.05) interacting with dexamethasone, several with roles in lung development, which included corticotropin-releasing hormone and surfactant protein B. Corticotropin-releasing hormone protein was detected in wild-type and Erk3(-/-) lungs at E14.5, with significantly temporally altered expression through embryonic day 18.5. Antenatal dexamethasone attenuated corticotropin-releasing hormone at embryonic day 18.5 in both wild-type and Erk3(-/-) lungs (0.56-fold and 0.67-fold; P < .001). Wild type mice responded to glucocorticoid administration with increased pulmonary surfactant protein B (P = .003). In contrast, dexamethasone treatment in Erk3(-/-) mice resulted in decreased surfactant protein B (P = .012). In human validation studies, we confirmed that corticotropin-releasing hormone protein is present in the fetal lung at 18 weeks of gestation and increases in expression with progression towards viability (22 weeks of gestation; P < .01). CONCLUSION: Characterization of whole transcriptome gene expression revealed glucocorticoid-mediated regulation of corticotropin-releasing hormone and surfactant protein B via Erk3-independent and -dependent mechanisms, respectively. We demonstrated for the first time the expression and temporal regulation of corticotropin-releasing hormone protein in midtrimester human fetal lung. This unique model allows the effects of corticosteroids on fetal pulmonary morphologic condition to be distinguished from functional gene pathway regulation. These findings implicate Erk3 as a potentially important molecular mediator of antenatal glucocorticoid action in promoting surfactant protein production in the preterm neonatal lung and expanding our understanding of key mechanisms of clinical therapy to improve neonatal survival.


Subject(s)
Dexamethasone/administration & dosage , Glucocorticoids/administration & dosage , Lung/pathology , Mitogen-Activated Protein Kinase 6/deficiency , Animals , Animals, Newborn , Corticotropin-Releasing Hormone/metabolism , Disease Models, Animal , Female , Insulin-Like Growth Factor II/metabolism , Lung/diagnostic imaging , Lung/metabolism , Lung/physiopathology , Mice, Knockout , Pregnancy , Pulmonary Surfactant-Associated Protein D/metabolism , Respiratory Distress Syndrome, Newborn/pathology , X-Ray Microtomography
19.
J Trop Pediatr ; 62(5): 385-9, 2016 10.
Article in English | MEDLINE | ID: mdl-27118822

ABSTRACT

BACKGROUND: Kenya's neonatal mortality rate remains unacceptably high, at 22 deaths per 1000 live births, with a third of those attributable to prematurity. Respiratory distress syndrome (RDS) is the single most important cause of morbidity and mortality in the premature neonate. Continuous positive airway pressure (CPAP) is a proven modality of therapy but is rarely used in low-resource settings. We report on the introduction of bubble CPAP (BCPAP), a low-cost method of delivering CPAP appropriate to our setting, by comparing survival-to-discharge before and after the technology was introduced. METHODS: The inpatient hospital records of all preterm infants (<37 weeks) diagnosed with RDS in the AIC Kijabe Hospital Nursery during two 18-month periods before and after the introduction of BCPAP (46 infants enrolled from 1 November 2007 to 30 April 2009 vs. 72 infants enrolled from 1 November 2009 to 30 April 2011) were reviewed. Differences in survival-to-discharge rates between the two time periods were analyzed. RESULTS: The survival-to-discharge rate was higher in Period 2 (after the introduction of BCPAP) than in Period 1 (pre-BCPAP) (85% vs. 61%, p = 0.007). Similarly, there were lower referral rates of preterm infants with RDS in Period 2 than Period 1 (4% vs. 17%, p = 0.037). CONCLUSION: BCPAP has contributed significantly to favorable outcomes for preterm infants with RDS at AIC Kijabe Hospital. The use of this simple technology should be considered and studied for expansion to all hospitals in Kenya that care for preterm infants.


Subject(s)
Continuous Positive Airway Pressure/methods , Infant, Premature , Intensive Care Units, Neonatal , Respiratory Distress Syndrome, Newborn/therapy , Continuous Positive Airway Pressure/economics , Continuous Positive Airway Pressure/instrumentation , Female , Hospitals, Rural , Humans , Infant , Infant, Newborn , Infant, Very Low Birth Weight , Kenya/epidemiology , Male , Oxygen Inhalation Therapy , Respiratory Distress Syndrome, Newborn/mortality , Respiratory Distress Syndrome, Newborn/pathology , Retrospective Studies , Rural Population , Survival Analysis , Survival Rate , Treatment Outcome
20.
Am J Forensic Med Pathol ; 37(2): 67-9, 2016 Jun.
Article in English | MEDLINE | ID: mdl-27049660

ABSTRACT

Respiratory distress syndrome (RDS) has a major contribution to neonatal mortality worldwide. Multiple factors associated with increased risk for RDS have been documented to effectively understand the emergence and progression of this disorder. A portion of these parameters has been broadly examined whereas the role of others, despite being clinically described, has not been fully evaluated. In this report, we analyze a forensic RDS case of a late preterm infant. Taking the maternal medical history into account, we focused on 2 not widely established risk factors, oligohydramnios and maternal age, discussing their possible pathophysiological relation to the development of RDS. Simultaneously, the fundamental role of the histopathological examination as a diagnostic tool resurfaces. Following a multidisciplinary approach derived from the collaboration of clinicians and researchers, the identification of factors that precipitate or contribute to this syndrome can be enhanced, leading to novel prognostic and therapeutic strategies against RDS.


Subject(s)
Maternal Age , Oligohydramnios , Respiratory Distress Syndrome, Newborn/pathology , Fatal Outcome , Female , Humans , Infant, Newborn , Infant, Premature , Infant, Premature, Diseases , Lung/pathology , Pregnancy , Risk Factors
SELECTION OF CITATIONS
SEARCH DETAIL