Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 326
Filtrer
1.
Clin Toxicol (Phila) ; 62(6): 357-363, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38946467

RÉSUMÉ

INTRODUCTION: The opioid receptor mu1 is a protein coding gene that can have different codes for a protein and may have variations (polymorphisms) affecting how opioids work. The aim of this study was to investigate the prevalence of the most common opioid receptor mu1 polymorphism (A118G) and any relationship between this polymorphism and features following tramadol overdose. MATERIALS AND METHODS: This was a cross-sectional study of patients admitted with tramadol poisoning to an Iranian hospital. These patients were not taking any other drugs or medications and had no history of seizures. RESULTS: The results showed that among the 83 patients included in the study, 57 (69 per cent) had the AA genotype, 25 (30 per cent) had the AG genotype, and one (1 per cent) had the GG genotype for the opioid receptor mu1 A118G polymorphism. Nausea and/or vomiting occurred in nine (11 per cent) patients and dizziness in 38 (46 per cent) patients. Serious adverse events included seizures in 51 (60 per cent) patients and respiratory failure requiring mechanical ventilation in 21 (25 per cent) patients. However, there was no significant association between the opioid receptor mu1 A118G polymorphism and these adverse events. DISCUSSION: In our study, the frequency of the A allele was greater than the G allele, and the AA genotype was more prevalent than AG. The GG genotype was the least common among the polymorphisms of opioid receptor mu1 rs1799971. There was no significant association between the opioid receptor mu1 A118G polymorphism and symptoms in tramadol-poisoned patients. Although these allele proportions are similar to the results reported in other Caucasian populations, they are dissimilar to the findings in Chinese and Singaporean populations. In these Asian studies, the predominant allele was the G allele. It has been suggested that a mutated G allele will decrease the production of opioid receptor mu1-related messenger ribonucleic acid and related proteins, leading to fewer mu-opioid receptors in the brain. CONCLUSIONS: This study found no significant association between the opioid receptor mu1 A118G polymorphism and adverse outcomes in tramadol-poisoned patients. However, more research is needed to draw more definitive conclusions due to the limited evidence and variability of opioid receptor mu1 polymorphisms in different populations.


Sujet(s)
Analgésiques morphiniques , Récepteur mu , Crises épileptiques , Tramadol , Humains , Tramadol/intoxication , Études transversales , Récepteur mu/génétique , Mâle , Femelle , Adulte , Iran , Analgésiques morphiniques/intoxication , Analgésiques morphiniques/effets indésirables , Adulte d'âge moyen , Crises épileptiques/génétique , Crises épileptiques/induit chimiquement , Jeune adulte , Polymorphisme de nucléotide simple , Mauvais usage des médicaments prescrits/génétique , Génotype , Nausée/induit chimiquement , Nausée/génétique , Insuffisance respiratoire/induit chimiquement , Insuffisance respiratoire/génétique , Vomissement/induit chimiquement , Vomissement/génétique , Adolescent , Sensation vertigineuse/induit chimiquement , Sensation vertigineuse/génétique
2.
Physiol Rep ; 12(8): e16015, 2024 Apr.
Article de Anglais | MEDLINE | ID: mdl-38653581

RÉSUMÉ

Adaptation of humans to challenging environmental conditions, such as extreme temperature, malnutrition, or hypoxia, is an interesting phenomenon for both basic and applied research. Identification of the genetic factors contributing to human adaptation to these conditions enhances our understanding of the underlying molecular and physiological mechanisms. In our study, we analyzed the exomes of 22 high altitude mountaineers to uncover genetic variants contributing to hypoxic adaptation. To our surprise, we identified two putative loss-of-function variants, rs1385101139 in RTEL1 and rs1002726737 in COL6A1 in two extremely high altitude (personal record of more than 8500 m) professional climbers. Both variants can be interpreted as pathogenic according to medical geneticists' guidelines, and are linked to inherited conditions involving respiratory failure (late-onset pulmonary fibrosis and severe Ullrich muscular dystrophy for rs1385101139 and rs1002726737, respectively). Our results suggest that a loss of gene function may act as an important factor of human adaptation, which is corroborated by previous reports in other human subjects.


Sujet(s)
Altitude , Collagène de type VI , Insuffisance respiratoire , Adulte , Femelle , Humains , Mâle , Adulte d'âge moyen , Mal de l'altitude/génétique , Collagène de type VI/génétique , Exome Sequencing/méthodes , Alpinisme , Insuffisance respiratoire/génétique
3.
Neonatology ; 121(1): 133-136, 2024.
Article de Anglais | MEDLINE | ID: mdl-38035569

RÉSUMÉ

Acinar dysplasia (AcDys) is one of the three main diffuse developmental disorders of the lung. The transcription factor NK2 homeobox 1 (NKX2.1) partly controls the synthesis of surfactant proteins by type 2 alveolar epithelial cells (AEC2), and germline mutations are known to be associated with brain-lung thyroid syndrome. We report the case of a full-term neonate who developed refractory respiratory failure with pulmonary hypertension requiring venoarterial extracorporeal membrane oxygenation. Histological examination of the lung biopsy specimen was consistent with the diagnosis of AcDys. Molecular analyses led to the identification of the missense heterozygous variant in NKX2.1 (NM_001079668) c.731A>G p.(Tyr244Cys), which is predicted to be pathogenic. After 5 weeks, because AcDys is a fatal disorder and the patient's status worsened, life-sustaining therapies were withdrawn, and she died after a few hours. This study is the first to extend the phenotype of NKX2.1 pathogenic variant, to a fatal form of AcDys.


Sujet(s)
Syndrome de détresse respiratoire du nouveau-né , Insuffisance respiratoire , Nouveau-né , Femelle , Humains , Mutation , Poumon/métabolisme , Facteurs de transcription/génétique , Facteurs de transcription/métabolisme , Insuffisance respiratoire/génétique , Insuffisance respiratoire/thérapie , Syndrome de détresse respiratoire du nouveau-né/diagnostic , Syndrome de détresse respiratoire du nouveau-né/génétique
5.
Nat Commun ; 14(1): 7443, 2023 Nov 17.
Article de Anglais | MEDLINE | ID: mdl-37978185

RÉSUMÉ

The transcriptional and phenotypic characteristics that define alveolar monocyte and macrophage subsets in acute hypoxemic respiratory failure (AHRF) are poorly understood. Here, we apply CITE-seq (single-cell RNA-sequencing and cell-surface protein quantification) to bronchoalveolar lavage and blood specimens longitudinally collected from participants with AHRF to identify alveolar myeloid subsets, and then validate their identity in an external cohort using flow cytometry. We identify alveolar myeloid subsets with transcriptional profiles that differ from other lung diseases as well as several subsets with similar transcriptional profiles as reported in healthy participants (Metallothionein) or patients with COVID-19 (CD163/LGMN). We use information from CITE-seq to determine cell-surface proteins that distinguish transcriptional subsets (CD14, CD163, CD123, CD71, CD48, CD86 and CD44). In the external cohort, we find a higher proportion of CD163/LGMN alveolar macrophages are associated with mortality in AHRF. We report a parsimonious set of cell-surface proteins that distinguish alveolar myeloid subsets using scalable approaches that can be applied to clinical cohorts.


Sujet(s)
Maladies pulmonaires , Insuffisance respiratoire , Humains , Macrophages alvéolaires/métabolisme , Macrophages/métabolisme , Monocytes/métabolisme , Maladies pulmonaires/métabolisme , Insuffisance respiratoire/génétique
6.
Pediatr Int ; 65(1): e15672, 2023.
Article de Anglais | MEDLINE | ID: mdl-37888536

RÉSUMÉ

BACKGROUND: Interactions among single nucleotide polymorphisms (SNPs) of surfactant protein (SP) are associated with acute respiratory failure (ARF) and its short-term outcome, pulmonary dysfunction at discharge (PDAD) in children. However, genetic association studies using individual SNPs have not been conducted before. We hypothesize that SP genetic variants are associated with pediatric ARF and its short-term complications by themselves. METHODS: We used available genotype and clinical data in the Floros biobank consisting of 248 children aged ≤24 months with ARF; 86 developed PDAD. A logistic regression analysis was performed for each of the 14 selected SNPs, SP-A1 and SP-A2 genotypes. A p-value less than the Bonferroni correction threshold was considered significant. A likelihood ratio test was done to compare two models (one with demographic data and another with genetic variants). RESULTS: Before Bonferroni correction, female sex is associated with a decreased risk of ARF. Black race and the rs721917 of the SFTPD are associated with increased risk of ARF. After Bonferroni correction, the 1A0 1A1 genotype of SFTPA2 was associated with decreased risk of ARF. The likelihood ratio test showed that the model of the genotype information with demographic data was a better fit to predict ARF risk. None of the SP SNPs and SP-A1, SP-A2 genotypes were associated with PDAD. CONCLUSION: Our results indicate that SNPs and genotypes of SPs involved in innate immunity and host defense play an important role in ARF and, in the future, may be used as biomarkers.


Sujet(s)
Surfactants pulmonaires , Insuffisance respiratoire , Humains , Enfant , Femelle , Protéine A associée au surfactant pulmonaire/génétique , Polymorphisme de nucléotide simple , Tensioactifs , Insuffisance respiratoire/génétique
7.
J Extra Corpor Technol ; 55(3): 147-152, 2023 Sep.
Article de Anglais | MEDLINE | ID: mdl-37682215

RÉSUMÉ

Recent data describe an increasing use of extracorporeal membrane oxygenation (ECMO) in neonates with various clinical conditions besides primary respiratory or cardiac diagnoses. Infants with underlying genetic disorders characterized by cardiopulmonary failure pose unique management challenges. When pathognomonic dysmorphic features for common genetic diagnoses are not present, the prognosis is uncertain at best when determining ECMO candidacy. Lengthy turnaround times of genetic testing often delay definitive diagnosis during the ECMO course. Clinical management pathways to guide practice and evidence to support the use of ECMO in rare genetic conditions are lacking. The decision to initiate ECMO is daunting but may be of benefit if the subsequent genetic diagnosis is non-lethal. In lethal genetic cases warranting discontinuation of care, the time spent on ECMO may still be advantageous as a bridge to diagnosis while allowing for parental bonding with the terminally ill infant. Diagnostic confirmation may also facilitate the attainment of closure for these parents. Here, we report our experience providing ECMO to three neonates presenting with cardiorespiratory failure and later diagnosed with rare genetic syndromes. We share the challenges faced, lessons learned, and outcomes of these critically ill neonates.


Sujet(s)
Oxygénation extracorporelle sur oxygénateur à membrane , Insuffisance respiratoire , Nourrisson , Nouveau-né , Humains , Coeur , Insuffisance respiratoire/diagnostic , Insuffisance respiratoire/génétique , Insuffisance respiratoire/thérapie
8.
Arch Toxicol ; 97(10): 2575-2585, 2023 Oct.
Article de Anglais | MEDLINE | ID: mdl-37537419

RÉSUMÉ

The increasing use of opioids in pregnant women has led to an alarming rise in the number of cases of neonates with drug-induced withdrawal symptoms known as neonatal opioid withdrawal syndrome (NOWS). NOWS is a toxic heterogeneous condition with many neurologic, autonomic, and gastrointestinal symptoms including poor feeding, irritability, tachycardia, hypertension, respiratory defects, tremors, hyperthermia, and weight loss. Paradoxically, for the management of NOWS, low doses of morphine, methadone, or buprenorphine are administered. NOWS is a polygenic disorder supported by studies of genomic variation in opioid-related genes. Single-nucleotide polymorphisms (SNPs) in CYP2B6 are associated with variations in NOWS infant responses to methadone and SNPs in the OPRM1, ABCB1, and COMT genes are associated with need for treatment and length of hospital stay. Epigenetic gene changes showing higher methylation levels in infants and mothers have been associated with more pharmacologic treatment in the case of newborns, and for mothers, longer infant hospital stays. Respiratory disturbances associated with NOWS are not well characterized. Little is known about the effects of opioids on developing neonatal respiratory control and respiratory distress (RD), a potential problem for survival of the neonate. In a rat model to test the effect of maternal opioids on the developing respiratory network and neonatal breathing, maternal-derived methadone increased apneas and lessened RD in neonates at postnatal (P) days P0 and P1. From P3, breathing normalized with age suggesting reorganization of respiratory rhythm-generating circuits at a time when the preBötC becomes the dominant inspiratory rhythm generator. In medullary slices containing the preBötC, maternal opioid treatment plus exposure to exogenous opioids showed respiratory activity was maintained in younger but not older neonates. Thus, maternal opioids blunt centrally controlled respiratory frequency responses to exogenous opioids in an age-dependent manner. In the absence of maternal opioid treatment, exogenous opioids abolished burst frequencies at all ages. Prenatal opioid exposure in children stunts growth rate and development while studies of behavior and cognitive ability reveal poor performances. In adults, high rates of attention deficit disorder, hyperactivity, substance abuse, and poor performances in intelligence and memory tests have been reported.


Sujet(s)
Syndrome de sevrage néonatal , Troubles liés aux opiacés , Insuffisance respiratoire , Syndrome de sevrage , Humains , Nouveau-né , Nourrisson , Adulte , Enfant , Femelle , Grossesse , Animaux , Rats , Analgésiques morphiniques/toxicité , Pharmacogénétique , Troubles liés aux opiacés/génétique , Troubles liés aux opiacés/complications , Troubles liés aux opiacés/traitement médicamenteux , Syndrome de sevrage/complications , Syndrome de sevrage/traitement médicamenteux , Méthadone/effets indésirables , Syndrome de sevrage néonatal/génétique , Syndrome de sevrage néonatal/complications , Syndrome de sevrage néonatal/traitement médicamenteux , Insuffisance respiratoire/induit chimiquement , Insuffisance respiratoire/génétique
11.
Acta Neuropathol ; 145(1): 127-143, 2023 01.
Article de Anglais | MEDLINE | ID: mdl-36264506

RÉSUMÉ

DNAJ/HSP40 co-chaperones are integral to the chaperone network, bind client proteins and recruit them to HSP70 for folding. We performed exome sequencing on patients with a presumed hereditary muscle disease and no genetic diagnosis. This identified four individuals from three unrelated families carrying an unreported homozygous stop gain (c.856A > T; p.Lys286Ter), or homozygous missense variants (c.74G > A; p.Arg25Gln and c.785 T > C; p.Leu262Ser) in DNAJB4. Affected patients presented with axial rigidity and early respiratory failure requiring ventilator support between the 1st and 4th decade of life. Selective involvement of the semitendinosus and biceps femoris muscles was seen on MRI scans of the thigh. On biopsy, muscle was myopathic with angular fibers, protein inclusions and occasional rimmed vacuoles. DNAJB4 normally localizes to the Z-disc and was absent from muscle and fibroblasts of affected patients supporting a loss of function. Functional studies confirmed that the p.Lys286Ter and p.Leu262Ser mutant proteins are rapidly degraded in cells. In contrast, the p.Arg25Gln mutant protein is stable but failed to complement for DNAJB function in yeast, disaggregate client proteins or protect from heat shock-induced cell death consistent with its loss of function. DNAJB4 knockout mice had muscle weakness and fiber atrophy with prominent diaphragm involvement and kyphosis. DNAJB4 knockout muscle and myotubes had myofibrillar disorganization and accumulated Z-disc proteins and protein chaperones. These data demonstrate a novel chaperonopathy associated with DNAJB4 causing a myopathy with early respiratory failure. DNAJB4 loss of function variants may lead to the accumulation of DNAJB4 client proteins resulting in muscle dysfunction and degeneration.


Sujet(s)
Maladies musculaires , Insuffisance respiratoire , Animaux , Souris , Mutation/génétique , Maladies musculaires/imagerie diagnostique , Maladies musculaires/génétique , Chaperons moléculaires/génétique , Chaperons moléculaires/métabolisme , Mutation faux-sens , Insuffisance respiratoire/génétique , Insuffisance respiratoire/complications , Insuffisance respiratoire/anatomopathologie , Muscles squelettiques/anatomopathologie
12.
Clin Respir J ; 16(6): 460-466, 2022 Jun.
Article de Anglais | MEDLINE | ID: mdl-35665444

RÉSUMÉ

OBJECTIVE: Severe pneumonia occurs commonly in children and is the main cause of clinical infant mortality. This study tested the expression pattern of long noncoding RNA cancer susceptibility candidate 2 (CASC2) in the serum of children with severe pneumonia and explored its clinical values. METHODS: Serum levels of CASC2 were detected in 145 children with severe pneumonia. All cases were divided into two groups based on their respiratory failure (RF) condition. Receiver operating characteristic (ROC) and Kaplan-Meier (K-M) curves were plotted for the diagnostic and prognostic ability evaluation. Multivariate cox regression analysis was done for the examination of independent influence factors. RESULTS: The serum levels of CASC2 significantly decreased in children with severe pneumonia in contrast with healthy individuals and reached the lowest value in those with RF. Serum CASC2 can distinguish severe pneumonia and predicted the development of RF. Based on the 28-day survival data, cases with low CASC2 levels had a poor survival rate. CASC2 (hazard ratio [HR] = 0.068, 95% confidence interval [CI] = 0.016-0.292, P < 0.001) and age (HR = 2.806, 95% CI = 1.240-6.394, P < 0.001) were independent influence factor for the poor prognosis of children with severe pneumonia. CONCLUSION: Downregulation of serum CASC2 was related to the occurrence of RF in children with severe pneumonia and may be a predictor of the poor prognosis. This study will provide a potential biomarker for severe pneumonia treatment in clinic.


Sujet(s)
Tumeurs , Pneumopathie infectieuse , ARN long non codant , Insuffisance respiratoire , Protéines suppresseurs de tumeurs/génétique , Lignée cellulaire tumorale , Prolifération cellulaire/génétique , Enfant , Humains , Pneumopathie infectieuse/complications , Pneumopathie infectieuse/génétique , ARN long non codant/génétique , Insuffisance respiratoire/génétique
13.
Cell Cycle ; 21(17): 1879-1886, 2022 09.
Article de Anglais | MEDLINE | ID: mdl-35587261

RÉSUMÉ

lncRNA CASC9 expression was involved in a variety of diseases and exerted a protective role against inflammation and sepsis-induced injury. However, the role of CASC9 in severe pneumonia remains unclear. This study aimed to explore the potential diagnostic role of lncRNA CASC9 in severe pneumonia. The CASC9 expression levels were measured by RT-qPCR. The receiver operating characteristic curve (ROC) was conducted to evaluate the clinical diagnostic value of CASC9 in severe pneumonia. LPS-induced human lung fibroblast MRC-5 was used to establish the pneumonia model and then transfected with CASC9 overexpression vectors to evaluate the influence of CASC9 on cell viability and apoptosis. The inflammatory cytokines IL-1ß, TNF-α, IL-6 levels were detected using a commercial enzyme-linked immunosorbent assay (ELISA). Pearson correlation analysis was used to explore the correlation between CASC9 expression and clinical data. The relative expression of CASC9 was downregulated in serum samples of severe pneumonia patients. The low expression of CASC9 in severe pneumonia was negatively correlated with several clinical data. The CASC9 had the relatively high area under ROC curve (AUC) values for distinguishing severe pneumonia from pneumonia children and healthy control. The elevated expression of CASC9 accelerated cell viability and diminished apoptosis in LPS-induced MRC-5 cells. The CASC9 expression was decreased in serum samples of severe pneumonia, and upregulation of CASC9 facilitated LPS-induced cell viability and inhibited apoptosis. In summary, CASC9 might be a diagnostic predictor and might act as a crucial regulatory roles in the progression of severe pneumonia.


Sujet(s)
Pneumopathie infectieuse , ARN long non codant , Insuffisance respiratoire , Apoptose/génétique , Lignée cellulaire tumorale , Enfant , Régulation de l'expression des gènes tumoraux , Humains , Lipopolysaccharides , Pneumopathie infectieuse/diagnostic , Pneumopathie infectieuse/génétique , ARN long non codant/génétique , Insuffisance respiratoire/génétique
14.
Genes (Basel) ; 13(5)2022 04 21.
Article de Anglais | MEDLINE | ID: mdl-35627110

RÉSUMÉ

LAS1L encodes a nucleolar ribosomal biogenesis protein and is also a component of the Five Friends of Methylated CHTOP (5FMC) complex. Mutations in the LAS1L gene can be associated with Wilson−Turner syndrome (WTS) and, much more rarely, severe infantile hypotonia with respiratory failure. Here, we present an eighteen-month old boy with a phenotype of spinal muscular atrophy with respiratory distress (SMARD). By applying WES, we identified a novel hemizygous synonymous variant in the LAS1L gene inherited from an unaffected mother (c.846G > C, p.Thr282=). We suggest that the identified variant impairs the RNA splicing process. Furthermore, we proved the absence of any coding regions by qPCR and sequencing cDNA using amplicon deep sequencing and Sanger sequencing methods. According to the SMARD phenotype, severe breathing problems causing respiratory insufficiency, hypotonia, and feeding difficulties were observed in our patient from the first days of life. Remarkably, our case is the second described patient with a SMARD-like phenotype due to a mutation in the LAS1L gene and the first with a variant impacting splicing.


Sujet(s)
Amyotrophie spinale , Insuffisance respiratoire , Humains , Hypotonie musculaire , Amyotrophie spinale/génétique , Mutation , Protéines nucléaires/génétique , Polyneuropathies , Syndrome de détresse respiratoire du nouveau-né , Insuffisance respiratoire/génétique
15.
Turk J Pediatr ; 64(2): 364-374, 2022.
Article de Anglais | MEDLINE | ID: mdl-35611426

RÉSUMÉ

BACKGROUND: Spinal muscular atrophy with respiratory distress type 1 (SMARD1) is a very rare autosomal recessive disorder caused by mutations in the immunoglobulin µ-binding protein-2 (IGHMBP2) gene on chromosome 11q13.2-q13.4. The initial symptoms of patients with SMARD1 are respiratory distress and distal muscle weakness manifesting in the infantile period due to progressive degeneration of α-motor neurons. Preterm birth, intrauterine growth retardation, feet deformities, sensory and autonomic neuropathy are other main features. CASE: Herein, we report the characteristics of a 6-year-old Turkish girl with a diagnosis of SMARD1 confirmed by homozygous c.1738G > A (p.Val580Ile) missense IGHMBP2 variant. She had unusual features such as vocal cord paralysis, nystagmus, and lack of congenital foot deformities besides typical findings including hypotonia, respiratory distress, and diaphragmatic weakness in the early infantile period. Epileptic seizures, cognitive impairment, and brain magnetic resonance imaging (MRI) abnormalities were other, unexpected, features which developed during the course of the disorder possibly due to several hypoxic episodes. CONCLUSIONS: SMARD1 should be kept in mind in hypotonic infants with diaphragmatic weakness and respiratory failure during the early infantile period, even in the presence of unexpected findings including vocal cord paralysis, nystagmus, epileptic seizures, and brain MRI abnormalities.


Sujet(s)
Naissance prématurée , Syndrome de détresse respiratoire du nouveau-né , Insuffisance respiratoire , Paralysie des cordes vocales , Enfant , Protéines de liaison à l'ADN/génétique , Femelle , Humains , Nourrisson , Nouveau-né , Hypotonie musculaire/génétique , Faiblesse musculaire/génétique , Amyotrophie spinale , Syndrome de détresse respiratoire du nouveau-né/diagnostic , Syndrome de détresse respiratoire du nouveau-né/génétique , Insuffisance respiratoire/génétique , Crises épileptiques , Facteurs de transcription/génétique
16.
Pharmacogenomics J ; 22(2): 124-129, 2022 03.
Article de Anglais | MEDLINE | ID: mdl-35169303

RÉSUMÉ

We have identified an underrecognized severe adverse drug reaction (ADR) of trimethoprim-sulfamethoxazole (TMP-SMX) associated respiratory failure in previously healthy children and young adults. We investigated potential genetic risk factors associated with TMP-SMX induced respiratory failure in a cohort of seven patients. We explored whole genome sequence among seven patients representing nearly half of all reported cases worldwide and 63 unrelated control individuals in two stages: (1) human leukocyte antigen (HLA) locus variation as several other ADRs have been associated HLA genetic variants and (2) coding variation to catalog and explore potential rare variants contributing to this devastating reaction. All cases were either heterozygous (carriers) or homozygous for the common HLA-B*07:02-HLA-C*07:02 haplotype. Despite the small sample size, this observation is statistically significant both in conservative comparison to maximum reported population frequencies (binomial P = 0.00017 for HLA-B and P = 0.00028 for HLA-C) and to our control population assessed by same HLA genotyping approach (binomial P = 0.000001 for HLA-B and P = 0.000018 for HLA-C). No gene elsewhere in the genome harnessed shared rare case enriched coding variation. Our results suggests that HLA-B*07:02 and HLA-C*07:02 are necessary for a patient to develop respiratory failure due to TMP-SMX.


Sujet(s)
Insuffisance respiratoire , Association triméthoprime-sulfaméthoxazole , Enfant , Antigènes HLA-B/génétique , Antigène HLA-B7 , Antigènes HLA-C/génétique , Humains , Insuffisance respiratoire/induit chimiquement , Insuffisance respiratoire/génétique , Association triméthoprime-sulfaméthoxazole/effets indésirables
17.
Pediatr Crit Care Med ; 23(5): 371-377, 2022 05 01.
Article de Anglais | MEDLINE | ID: mdl-35213412

RÉSUMÉ

OBJECTIVES: We have previously shown that patients with a chromosome 22q11 microdeletion are at risk for prolonged respiratory failure after pulmonary artery reconstruction surgery compared with those with normal genotype. We sought to describe preexisting airway abnormalities in this patient population and examine relationships between airway abnormalities and outcomes. DESIGN: Single-center retrospective chart review from Society of Thoracic Surgery and Pediatric Cardiac Critical Care Consortium databases and the electronic medical record. SETTING: Lucile Packard Children's Hospital at Stanford from September 2017 to February 2019. PATIENTS: All patients undergoing pulmonary artery reconstruction surgery were considered for inclusion. INTERVENTIONS: We identified 127 patients meeting study inclusion criteria. Thirty-nine patients met specific criteria and underwent screening preoperative bronchoscopy including microdirect laryngoscopy and lower airway examination. Postoperative bronchoscopy was performed at the discretion of the intensive care team. MEASUREMENTS AND MAIN RESULTS: Airway abnormalities were detected in 25/26 of children (96%) with a chromosome 22q11 deletion who underwent preoperative bronchoscopy. Upper and lower airway pathologies were found in 19/25 (73%) and 21/25 (81%) patients, respectively, and it was common for patients to have more than one abnormality. Presence of 22q11 deletion was associated with longer duration of mechanical ventilation (9.1 vs 4.3 d; p = 0.001), use of noninvasive positive pressure support (13 vs 6 d; p = 0.001), and longer hospital stays (30 vs 14 d; p = 0.002). These outcomes were worse when compared with patients with known airway abnormalities who did not have 22q11 deletion. CONCLUSIONS: Preexisting upper and lower airway pathologies are common in patients with a chromosome 22q11 deletion who undergo pulmonary artery reconstruction surgery. Despite similar postoperative hemodynamics and outcomes as their counterparts without 22q11 deletion, 22q11 deletion is associated with more postoperative respiratory complications not entirely explained by preexisting airway abnormalities.


Sujet(s)
Artère pulmonaire , Insuffisance respiratoire , Bronchoscopie , Enfant , Délétion de segment de chromosome , Humains , Poumon , Complications postopératoires/épidémiologie , Artère pulmonaire/chirurgie , Insuffisance respiratoire/génétique , Études rétrospectives
18.
Neurol Sci ; 43(5): 3371-3380, 2022 May.
Article de Anglais | MEDLINE | ID: mdl-34839411

RÉSUMÉ

Hereditary myopathy with early respiratory failure (HMERF) is a subtype of myofibrillar myopathy. Mutations located on exon 344 of the titin-A band, the 119th fibronectin-3 domain (FN3 119), are responsible for HMERF. In this article, we retrospectively analyzed the clinical features, findings of muscle imaging, muscle pathology, immunohistochemistry, and ultrastructural characteristics of seven patients diagnosed with HMERF at a single center in China. Muscle MRI showed the involvement of semitendinosus in four patients. The common pathological features were variability in fiber diameter, increased internal nuclei, endomysial fibrosis, and cytoplasmic bodies. On immunohistochemical examination, the cytoplasmic bodies stained positive for calpain-3, p53, and programmed death-ligand 1. Electron microscopy showed cytoplasmic bodies, distorted sarcomere architecture, glycogen pool, and subsarcolemmal accumulation of mitochondria and lysosomes. We retrospectively reviewed four reported HMERF patients in China. Among the 11 patients, the median age at onset was 34 years (range 14-54). Allelic frequency of mutation c.95195C > T was 36.36%. This study characterizes the phenotype and genotype spectrum of HMERF in China.


Sujet(s)
Maladies musculaires , Insuffisance respiratoire , Maladies génétiques congénitales , Humains , Biologie moléculaire , Muscles squelettiques/anatomopathologie , Maladies musculaires/diagnostic , Mutation/génétique , Insuffisance respiratoire/diagnostic , Insuffisance respiratoire/génétique , Études rétrospectives
19.
Neuropharmacology ; 202: 108858, 2022 01 01.
Article de Anglais | MEDLINE | ID: mdl-34715121

RÉSUMÉ

Because of increased opioid misuse, there is a need to identify new targets for minimizing opioid tolerance, and physical and psychological dependence. Previous studies showed that fibroblast growth factor 21 (FGF21) decreased alcohol and sweet preference in mice. In this study, FGF21-transgenic (FGF21-Tg) mice, expressing high FGF21 serum levels, and wildtype (WT) C57BL/6J littermates were treated with morphine and saline to determine if differences exist in their physiological and behavioral responses to opioids. FGF21-Tg mice displayed reduced preference for morphine in the conditioned place preference assay compared to WT littermates. Similarly, FGF21-Tg mice had an attenuation of the magnitude and rate of acute morphine antinociceptive tolerance development, and acute and chronic morphine physical dependence, but exhibited no change in chronic morphine antinociceptive tolerance. The ED50 values for morphine-induced antinociception in the 55 °C hot plate and the 55 °C warm-water tail withdrawal assays were similar in both strains of mice. Likewise, FGF21-Tg and WT littermates had comparable responses to morphine-induced respiratory depression. Overall, FGF21-Tg mice had a decrease in the development of acute analgesic tolerance, and the development of physical dependence, and morphine preference. FGF21 and its receptor have therapeutic potential for reducing opioid withdrawal symptoms and craving, and augmenting opioid therapeutics for acute pain patients to minimize tolerance development.


Sujet(s)
Tolérance aux médicaments/génétique , Facteurs de croissance fibroblastique/sang , Facteurs de croissance fibroblastique/physiologie , Dépendance à la morphine/génétique , Morphine/effets indésirables , Nociception/effets des médicaments et des substances chimiques , Animaux , Comportement animal/effets des médicaments et des substances chimiques , Souris de lignée C57BL , Souris transgéniques , Insuffisance respiratoire/induit chimiquement , Insuffisance respiratoire/génétique , Syndrome de sevrage/génétique , Syndrome de sevrage/thérapie
20.
Intern Med ; 61(10): 1587-1592, 2022 May 15.
Article de Anglais | MEDLINE | ID: mdl-34670883

RÉSUMÉ

Hereditary myopathy with early respiratory failure (HMERF) is caused by titin A-band mutations in exon 344 and is considered quite rare. Respiratory insufficiency can be the sole symptom in the disease course. We herein report the first Japanese HMERF patient with a p.P31732L mutation in titin. The patient manifested respiratory failure and mild weakness of the neck flexor muscle at 69 years old and showed fatty replacement of the bilateral semitendinosus muscles on muscle imaging. Our case indicates that HMERF with a heterozygous p.P31732L mutation should be included in the differential diagnosis of muscular diseases presenting with early respiratory failure.


Sujet(s)
Connectine , Maladies musculaires , Insuffisance respiratoire , Sujet âgé , Connectine/génétique , Maladies génétiques congénitales , Humains , Japon , Muscles squelettiques , Maladies musculaires/complications , Maladies musculaires/imagerie diagnostique , Maladies musculaires/génétique , Mutation/génétique , Insuffisance respiratoire/diagnostic , Insuffisance respiratoire/génétique
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE