1
|
Timofeeva AV, Fedorov IS, Nikonets AD, Tarasova AM, Balashova EN, Degtyarev DN, Sukhikh GT. Increased Levels of hsa-miR-199a-3p and hsa-miR-382-5p in Maternal and Neonatal Blood Plasma in the Case of Placenta Accreta Spectrum. Int J Mol Sci 2024; 25:13309. [PMID: 39769074 PMCID: PMC11678653 DOI: 10.3390/ijms252413309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 12/08/2024] [Accepted: 12/09/2024] [Indexed: 01/11/2025] Open
Abstract
Despite the increasing number of placenta accreta spectrum (PAS) cases in recent years, its impact on neonatal outcomes and respiratory morbidity, as well as the underlying pathogenetic mechanism, has not yet been extensively studied. Moreover, no study has yet demonstrated the effectiveness of antenatal corticosteroid therapy (CT) for the prevention of respiratory distress syndrome (RDS) in newborns of mothers with PAS at the molecular level. In this regard, microRNA (miRNA) profiling by small RNA deep sequencing and quantitative real-time PCR was performed on 160 blood plasma samples from preterm infants (gestational age: 33-36 weeks) and their mothers who had been diagnosed with or without PAS depending on the timing of the antenatal RDS prophylaxis. A significant increase in hsa-miR-199a-3p and hsa-miR-382-5p levels was observed in the blood plasma of the newborns from mothers with PAS compared to the control group. A clear trend toward the normalization of hsa-miR-199a-3p and hsa-miR-382-5p levels in the neonatal blood plasma of the PAS groups was observed when CT was administered within 14 days before delivery, but not beyond 14 days. Direct correlations were found among the hsa-miR-382-5p level in neonatal blood plasma and the hsa-miR-199a-3p level in the same sample (r = 0.49; p < 0.001), the oxygen requirements in the NICU (r = 0.41; p = 0.001), the duration of the NICU stay (r = 0.31; p = 0.019), and the severity of the newborn's condition based on the NEOMOD scale (r = 0.36; p = 0.005). Logistic regression models based on the maternal plasma levels of hsa-miR-199a-3p and hsa-miR-382-5p predicted the need for cardiotonic therapy, invasive mechanical ventilation, or high-frequency oscillatory ventilation in newborns during the early neonatal period, with a sensitivity of 95-100%. According to the literary data, these miRNAs regulate fetal organogenesis via IGF-1, the formation of proper lung tissue architecture, surfactant synthesis in alveolar cells, and vascular tone.
Collapse
Affiliation(s)
- Angelika V. Timofeeva
- National Medical Research Center for Obstetrics, Gynecology and Perinatology Named After Academician Kulakov V.I., 117997 Moscow, Russia
| | | | | | | | | | | | | |
Collapse
|
2
|
Cave C, Samano D, Sharma AM, Dickinson J, Salomon J, Mahapatra S. Acute respiratory distress syndrome: A review of ARDS across the life course. J Investig Med 2024; 72:798-818. [PMID: 39092841 DOI: 10.1177/10815589241270612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/04/2024]
Abstract
Acute respiratory distress syndrome (ARDS) is a multifactorial, inflammatory lung disease with significant morbidity and mortality that predominantly requires supportive care in its management. Although initially described in adult patients, the diagnostic definitions for ARDS have evolved over time to accurately describe this disease process in pediatric and, more recently, neonatal patients. The management of ARDS in each age demographic has converged in the application of lung-protective ventilatory strategies to mitigate the primary disease process and prevent its exacerbation by limiting ventilator-induced lung injury. However, differences arise in the preferred ventilatory strategies or adjunctive pulmonary therapies used to mitigate each type of ARDS. In this review, we compare and contrast the epidemiology, common etiologies, pathophysiology, diagnostic criteria, and outcomes of ARDS across the lifespan. Additionally, we discuss in detail the different management strategies used for each subtype of ARDS and spotlight how these strategies were applied to mitigate poor outcomes during the COVID-19 pandemic. This review is geared toward both clinicians and clinician-scientists as it not only summarizes the latest information on disease pathogenesis and patient management in ARDS across the lifespan but also highlights knowledge gaps for further investigative efforts. We conclude by projecting how future studies can fill these gaps in research and what improvements may be envisioned in the management of NARDS and PARDS based on the current breadth of literature on adult ARDS treatment strategies.
Collapse
Affiliation(s)
- Caleb Cave
- Division of Neonatology, and Division of Pulmonology, Department of Pediatrics, Children's Hospital and Medical Center, Omaha, University of Nebraska Medical Center, Omaha, NE, USA
| | - Dannielle Samano
- Division of Pulmonary, Sleep, and Critical Care Medicine, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Abhineet M Sharma
- Division of Neonatology, and Division of Pulmonology, Department of Pediatrics, Children's Hospital and Medical Center, Omaha, University of Nebraska Medical Center, Omaha, NE, USA
| | - John Dickinson
- Division of Pulmonary, Sleep, and Critical Care Medicine, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Jeffrey Salomon
- Division of Critical Care Medicine, Department of Pediatrics, Children's Hospital and Medical Center, Omaha, University of Nebraska Medical Center, Omaha, NE, USA
| | - Sidharth Mahapatra
- Division of Critical Care Medicine, Department of Pediatrics, Children's Hospital and Medical Center, Omaha, University of Nebraska Medical Center, Omaha, NE, USA
| |
Collapse
|
3
|
Baker AK, Beardsley AL, Leland BD, Moser EA, Lutfi RL, Cristea AI, Rowan CM. Predictors of Failure of Noninvasive Ventilation in Critically Ill Children. J Pediatr Intensive Care 2023; 12:196-202. [PMID: 37565011 PMCID: PMC10411242 DOI: 10.1055/s-0041-1731433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Accepted: 05/14/2021] [Indexed: 10/21/2022] Open
Abstract
Noninvasive ventilation (NIV) is a common modality employed to treat acute respiratory failure. Most data guiding its use is extrapolated from adult studies. We sought to identify clinical predictors associated with failure of NIV, defined as requiring intubation. This single-center retrospective observational study included children admitted to pediatric intensive care unit (PICU) between July 2014 and June 2016 treated with NIV, excluding postextubation. A total of 148 patients was included. Twenty-seven (18%) failed NIV. There was no difference between the two groups with regard to age, gender, comorbidities, or etiology of acute respiratory failure. Those that failed had higher admission pediatric risk of mortality ( p = 0.01) and pediatric logistic organ dysfunction ( p = 0.002) scores and higher fraction of inspired oxygen (FiO 2 ; p = 0.009) at NIV initiation. Failure was associated with lack of improvement in tachypnea. At 6 hours of NIV, the failure group had worsening tachypnea with a median increase in respiratory rate of 8%, while the success group had a median reduction of 18% ( p = 0.06). Multivariable Cox's proportional hazard models revealed FiO 2 at initiation and worsening respiratory rate at 1- and 6-hour significant risks for failure of NIV. Failure was associated with a significantly longer PICU length of stay (success [2.8 days interquartile range (IQR): 1.7, 5.5] vs. failure [10.6 days IQR: 5.6, 13.2], p < 0.001). NIV can be successfully employed to treat acute respiratory failure in pediatric patients. There should be heightened concern for NIV failure in hypoxemic patients whose tachypnea is unresponsive to NIV. A trend toward improvement should be closely monitored.
Collapse
Affiliation(s)
- Alyson K. Baker
- Division of Pediatric Critical Care, Riley Hospital for Children, Indiana University, Indianapolis, Indiana, United States
| | - Andrew L. Beardsley
- Division of Pediatric Critical Care, Riley Hospital for Children, Indiana University, Indianapolis, Indiana, United States
| | - Brian D. Leland
- Division of Pediatric Critical Care, Riley Hospital for Children, Indiana University, Indianapolis, Indiana, United States
| | - Elizabeth A. Moser
- Department of Biostatistics, Indiana University, Indianapolis, Indiana, United States
| | - Riad L. Lutfi
- Division of Pediatric Critical Care, Riley Hospital for Children, Indiana University, Indianapolis, Indiana, United States
| | - A. Ioana Cristea
- Division of Pediatric Pulmonology, Riley Hospital for Children, Indiana University, Indianapolis, Indiana, United States
| | - Courtney M. Rowan
- Division of Pediatric Critical Care, Riley Hospital for Children, Indiana University, Indianapolis, Indiana, United States
| |
Collapse
|
4
|
McCahill C, Laycock HC, Guris RJD, Chigaru L. State-of-the-art management of the acutely unwell child. Anaesthesia 2022; 77:1288-1298. [PMID: 36089884 PMCID: PMC9826095 DOI: 10.1111/anae.15816] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/05/2022] [Indexed: 01/11/2023]
Abstract
Children make up around one-fifth of all emergency department visits in the USA and UK, with an increasing trend of emergency admissions requiring intensive care. Anaesthetists play a vital role in the management of paediatric emergencies contributing to stabilisation, emergency anaesthesia, transfers and non-technical skills that optimise team performance. From neonates to adolescents, paediatric patients have diverse physiology and present with a range of congenital and acquired pathologies that often differ from the adult population. With increasing centralisation of paediatric services, staff outside these centres have less exposure to caring for children, yet are often the first responders in managing these high stakes situations. Staying abreast of the latest evidence for managing complex low frequency emergencies is a challenge. This review focuses on recent evidence and pertinent clinical updates within the field. The challenges of maintaining skills and training are explored as well as novel advancements in care.
Collapse
Affiliation(s)
- C. McCahill
- Department of AnaesthesiaGreat Ormond Street HospitalLondonUK
| | - H. C. Laycock
- Department of AnaesthesiaGreat Ormond Street HospitalLondonUK,Department of Surgery and CancerImperial CollegeLondonUK
| | - R. J. Daly Guris
- Department of Anesthesiology and Critical Care MedicineChildren's Hospital of PhiladelphiaPhiladelphiaPAUSA,Department of Anesthesiology and Critical CareUniversity of Pennsylvania Perelman School of MedicinePhiladelphiaPAUSA
| | - L. Chigaru
- Department of AnaesthesiaGreat Ormond Street HospitalLondonUK,Children's Acute Transport ServiceLondonUK
| |
Collapse
|
5
|
Gong X, He Y, Lu G, Zhang Y, Qiu Y, Qiao L, Li Y. Exome sequencing contributes to identify comorbidities in a rare case of infant ARDS induced by the CD40LG mutation. BMC Med Genomics 2022; 15:153. [PMID: 35804376 PMCID: PMC9264746 DOI: 10.1186/s12920-022-01303-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Accepted: 07/01/2022] [Indexed: 11/22/2022] Open
Abstract
Background Acute respiratory distress syndrome (ARDS) causes significant mortality in young children with certain diseases. Early diagnosis and treatment can reduce infant mortality. Here, we report a rare case of exome sequencing in the early diagnosis of immunodeficiency in an infant. Case presentation A four-month-old full-term male infant presented with severe shortness of breath, hypoxemia, and unexplained parenchymal lung lesions. A series of examinations were performed to search for potential culprit viruses but negative results were obtained with the only exception being the rhinovirus that tested positive. The child’s family history revealed he had a brother who died of severe infection at the age of two years. We performed an exome sequencing analysis and a mutation analysis of CD40LG to obtain genetic data on the patient. Besides, we used flow cytometry to measure the CD40LG expression levels of activated T cells. A retrospective review of all the CD40LG mutant-induced X-linked hyper IgM syndromes (XHIGM) had been conducted to assess the differences between clinical and genetic molecular features. Finally, a regular intravenous immunoglobulin (IVIG) regimen led to steady breathing, the correction of hypoxemia, and a progressive improvement of lung CT scans. During follow-up, the patient received an IVIG regimen and his CT images improved. Moreover, his parents took advantage of pre-implantation genetic testing with in vitro fertilization to have a healthy twin offspring who did not carry such a mutation according to the early exome sequencing for the proband. Compared with other CD40LG mutant cases in our center, this proband displayed a normal plasma immunoglobulin level and he should be the youngest infant to have a molecular diagnosis of XHIGM. Conclusion Usually, XHIGM would not be suspected with a normal plasma immunoglobulin concentration. However, as we could not identify a potential comorbidity or risk factor, exome sequencing helps target this patient's real facts. Thus, this case report calls for exome sequencing to be performed in the case of unexplained infections when immunodeficiency is suspected after general immunological tests, especially for cases with a contributive family history among infants as the maternal transfused immunoglobulin might mask immune deficiency. Supplementary Information The online version contains supplementary material available at 10.1186/s12920-022-01303-y.
Collapse
Affiliation(s)
- Xue Gong
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Pediatrics, West China Second University Hospital, Sichuan University, No. 20, 3rd Section, South Renmin Road, Chengdu, 610041, Sichuan, China
| | - Yunru He
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Pediatrics, West China Second University Hospital, Sichuan University, No. 20, 3rd Section, South Renmin Road, Chengdu, 610041, Sichuan, China
| | - Guoyan Lu
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Pediatrics, West China Second University Hospital, Sichuan University, No. 20, 3rd Section, South Renmin Road, Chengdu, 610041, Sichuan, China
| | - Yulin Zhang
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Pediatrics, West China Second University Hospital, Sichuan University, No. 20, 3rd Section, South Renmin Road, Chengdu, 610041, Sichuan, China
| | - Yu Qiu
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Pediatrics, West China Second University Hospital, Sichuan University, No. 20, 3rd Section, South Renmin Road, Chengdu, 610041, Sichuan, China
| | - Lina Qiao
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Pediatrics, West China Second University Hospital, Sichuan University, No. 20, 3rd Section, South Renmin Road, Chengdu, 610041, Sichuan, China
| | - Yifei Li
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Pediatrics, West China Second University Hospital, Sichuan University, No. 20, 3rd Section, South Renmin Road, Chengdu, 610041, Sichuan, China.
| |
Collapse
|
6
|
Lederer CW, Koniali L, Buerki-Thurnherr T, Papasavva PL, La Grutta S, Licari A, Staud F, Bonifazi D, Kleanthous M. Catching Them Early: Framework Parameters and Progress for Prenatal and Childhood Application of Advanced Therapies. Pharmaceutics 2022; 14:pharmaceutics14040793. [PMID: 35456627 PMCID: PMC9031205 DOI: 10.3390/pharmaceutics14040793] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 03/29/2022] [Accepted: 04/01/2022] [Indexed: 01/19/2023] Open
Abstract
Advanced therapy medicinal products (ATMPs) are medicines for human use based on genes, cells or tissue engineering. After clear successes in adults, the nascent technology now sees increasing pediatric application. For many still untreatable disorders with pre- or perinatal onset, timely intervention is simply indispensable; thus, prenatal and pediatric applications of ATMPs hold great promise for curative treatments. Moreover, for most inherited disorders, early ATMP application may substantially improve efficiency, economy and accessibility compared with application in adults. Vindicating this notion, initial data for cell-based ATMPs show better cell yields, success rates and corrections of disease parameters for younger patients, in addition to reduced overall cell and vector requirements, illustrating that early application may resolve key obstacles to the widespread application of ATMPs for inherited disorders. Here, we provide a selective review of the latest ATMP developments for prenatal, perinatal and pediatric use, with special emphasis on its comparison with ATMPs for adults. Taken together, we provide a perspective on the enormous potential and key framework parameters of clinical prenatal and pediatric ATMP application.
Collapse
Affiliation(s)
- Carsten W. Lederer
- The Molecular Genetics Thalassemia Department, The Cyprus Institute of Neurology & Genetics, Nicosia 2371, Cyprus; (L.K.); (P.L.P.); (M.K.)
- Correspondence: ; Tel.: +357-22-392764
| | - Lola Koniali
- The Molecular Genetics Thalassemia Department, The Cyprus Institute of Neurology & Genetics, Nicosia 2371, Cyprus; (L.K.); (P.L.P.); (M.K.)
| | - Tina Buerki-Thurnherr
- Empa, Swiss Federal Laboratories for Materials Science and Technology, 9014 St. Gallen, Switzerland;
| | - Panayiota L. Papasavva
- The Molecular Genetics Thalassemia Department, The Cyprus Institute of Neurology & Genetics, Nicosia 2371, Cyprus; (L.K.); (P.L.P.); (M.K.)
| | - Stefania La Grutta
- Institute of Translational Pharmacology, IFT National Research Council, 90146 Palermo, Italy;
| | - Amelia Licari
- Pediatric Clinic, Department of Clinical, Surgical, Diagnostic and Pediatric Sciences, Fondazione IRCCS Policlinico San Matteo, University of Pavia, 27100 Pavia, Italy;
| | - Frantisek Staud
- Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Králové, Charles University, 50005 Hradec Králové, Czech Republic;
| | - Donato Bonifazi
- Consorzio per Valutazioni Biologiche e Farmacologiche (CVBF) and European Paediatric Translational Research Infrastructure (EPTRI), 70122 Bari, Italy;
| | - Marina Kleanthous
- The Molecular Genetics Thalassemia Department, The Cyprus Institute of Neurology & Genetics, Nicosia 2371, Cyprus; (L.K.); (P.L.P.); (M.K.)
| |
Collapse
|
7
|
Wang Q, Wu Q. Knockdown of receptor interacting protein 140 (RIP140) alleviated lipopolysaccharide-induced inflammation, apoptosis and permeability in pulmonary microvascular endothelial cells by regulating C-terminal binding protein 2 (CTBP2). Bioengineered 2022; 13:3981-3992. [PMID: 35113002 PMCID: PMC8973626 DOI: 10.1080/21655979.2022.2031403] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
The main pathological feature of acute lung injury (ALI) is pulmonary edema caused by increased permeability of pulmonary microvascular endothelial cells (PMVECs). LPS was has been confirmed to lead to cell damage and barrier dysfunction in PMVECs. Furthermore, receptor interacting protein 140 (RIP140) was discovered to be increased in LPS-induced human pulmonary microvascular endothelial cells (HPMECs), but the mechanism of RIP140 on LPS-induced HPMECs has not been investigated. In this study, an acute lung injury model was constructed in LPS-induced HPMECs. After RIP140 was downregulated, inflammation, apoptosis and cell permeability levels were detected by RT-qPCR, TUNEL staining and FITC-Dextran, respectively. Western blotting was used to detect the protein levels of related factors. The binding of RIP140 and C-terminal binding protein 2 (CTBP2) was predicted by database and verified by Co-IP. Subsequently, CTBP2 overexpression was transfected into cells and the above experiments were performed again. The results showed that inflammation, apoptosis and permeability levels of LPS-induced HPMECs were remarkably increased compared to the untreated control group. However, these levels were suppressed after RIP140 was silenced compared to the LPS-induced HPMECs group. Notably, the Co-IP study demonstrated that RIP140 and CTBP2 interacted with each other. Moreover, CTBP2 overexpression reversed the inhibitory effects of RIP140 silencing on LPS-induced inflammation, apoptosis and permeability levels in HPMECs. Together, the study found that interference of RIP140 could alleviate LPS-induced inflammation, apoptosis and permeability in HPMECs by regulating CTBP2.
Collapse
Affiliation(s)
- Qizheng Wang
- Department of Pediatrics, Huai'an Maternal and Child Health Care Hospital, Huai'an, Jiangu, China
| | - Qiong Wu
- Department of Pediatrics, The People's Hospital of China Three Gorges University (The First People's Hospital of Yichang), Yichang, Hubei, China
| |
Collapse
|
8
|
Rai E, Alaraimi R, Al Aamri I. Pediatric lower respiratory tract infection: Considerations for the anesthesiologist. Paediatr Anaesth 2022; 32:181-190. [PMID: 34927318 DOI: 10.1111/pan.14382] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 12/06/2021] [Accepted: 12/13/2021] [Indexed: 11/29/2022]
Abstract
Neonatal and childhood infectious diseases continue to be a global health problem. Acute respiratory tract infections are typically classified as upper respiratory tract infection and lower respiratory tract infections. The most common lower respiratory infections in childhood are pneumonia and bronchiolitis. Vaccination against measles, diphtheria, pertussis, Haemophilus influenzae, pneumococcus, and influenza resulted in a significant reduction in the incidence of acute respiratory tract infection globally. Though the global burden of the disease has decreased, the mortality rates still are higher in developing countries. Patients with severe lower respiratory tract infections and their complications are often evaluated for elective or emergency procedures. In this review article, the authors aim to discuss the etiology, pathogenesis, preoperative evaluation of lower respiratory tract infections, and the anesthesia implications pertinent to the practice of anesthesia.
Collapse
Affiliation(s)
- Ekta Rai
- Christian Medical College, Vellore, India
| | - Rashid Alaraimi
- Emergency Physician & Neurocritical Care Fellow, Montreal Neurological Institute and Hospital, Montreal, QC, Canada
| | - Is'haq Al Aamri
- Adult and Pediatric Cardiac Anesthesia Consultant, National Heart Center, Muscat, Oman
| |
Collapse
|
9
|
Zoulikha M, Xiao Q, Boafo GF, Sallam MA, Chen Z, He W. Pulmonary delivery of siRNA against acute lung injury/acute respiratory distress syndrome. Acta Pharm Sin B 2022; 12:600-620. [PMID: 34401226 PMCID: PMC8359643 DOI: 10.1016/j.apsb.2021.08.009] [Citation(s) in RCA: 132] [Impact Index Per Article: 44.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 06/14/2021] [Accepted: 07/02/2021] [Indexed: 02/08/2023] Open
Abstract
The use of small interfering RNAs (siRNAs) has been under investigation for the treatment of several unmet medical needs, including acute lung injury/acute respiratory distress syndrome (ALI/ARDS) wherein siRNA may be implemented to modify the expression of pro-inflammatory cytokines and chemokines at the mRNA level. The properties such as clear anatomy, accessibility, and relatively low enzyme activity make the lung a good target for local siRNA therapy. However, the translation of siRNA is restricted by the inefficient delivery of siRNA therapeutics to the target cells due to the properties of naked siRNA. Thus, this review will focus on the various delivery systems that can be used and the different barriers that need to be surmounted for the development of stable inhalable siRNA formulations for human use before siRNA therapeutics for ALI/ARDS become available in the clinic.
Collapse
Key Words
- AAV, adeno-associated virus
- ALI/ARDS
- ALI/ARDS, acute lung injury/acute respiratory distress syndrome
- AM, alveolar macrophage
- ATI, alveolar cell type I
- ATII, alveolar cell type II
- AV, adenovirus
- Ago-2, argonaute 2
- CFDA, China Food and Drug Administration
- COPD, chronic obstructive pulmonary disease
- CPP, cell-penetrating peptide
- CS, cigarette smoke
- CXCR4, C–X–C motif chemokine receptor type 4
- Cellular uptake
- DAMPs, danger-associated molecular patterns
- DC-Chol, 3β-(N-(N′,N′-dimethylethylenediamine)-carbamoyl) cholesterol
- DDAB, dimethyldioctadecylammonium bromide
- DODAP, 1,2-dioleyl-3-dimethylammonium-propane
- DODMA, 1,2-dioleyloxy-N,N-dimethyl-3-aminopropane
- DOGS, dioctadecyl amido glycin spermine
- DOPC, 1,2-dioleoyl-sn-glycero-3-phosphocholine
- DOPE, 1,2-dioleoyl-l-α-glycero-3-phosphatidylethanolamine
- DOSPA, 2,3-dioleyloxy-N-[2-(sperminecarboxamido)ethyl]-N,N-dimethyl-1-propanaminium
- DOTAP, 1,2-dioleoyl-3-trimethylammonium-propane
- DOTMA, N-[1-(2,3-dioleoyloxy)propyl]-N,N,N-trimethylammonium
- DPI, dry powder inhaler
- DPPC, 1,2-dipalmitoyl-sn-glycero-3-phosphocholine
- Drug delivery
- EC, endothelial cell
- EPC, egg phosphatidylcholine
- EXOs, exosomes
- Endosomal escape
- EpiC, epithelial cell
- FDA, US Food and Drug Administration
- HALI, hyperoxic acute lung injury
- HMGB1, high-mobility group box 1
- HMVEC, human primary microvascular endothelial cell
- HNPs, hybrid nanoparticles
- Hem-CLP, hemorrhagic shock followed by cecal ligation and puncture septic challenge
- ICAM-1, intercellular adhesion molecule-1
- IFN, interferons
- Inflammatory diseases
- LPS, lipopolysaccharides
- MEND, multifunctional envelope-type nano device
- MIF, macrophage migration inhibitory factor
- Myd88, myeloid differentiation primary response 88
- N/P ratio, nitrogen /phosphate ratio
- NETs, neutrophil extracellular traps
- NF-κB, nuclear factor kappa B
- NPs, nanoparticles
- Nanoparticles
- PAI-1, plasminogen activator inhibitor-1
- PAMAM, polyamidoamine
- PAMPs, pathogen-associated molecular patterns
- PD-L1, programmed death ligand-1
- PDGFRα, platelet-derived growth factor receptor-α
- PEEP, positive end-expiratory pressure
- PEG, polyethylene glycol
- PEI, polyethyleneimine
- PF, pulmonary fibrosis
- PFC, perfluorocarbon
- PLGA, poly(d,l-lactic-co-glycolic acid)
- PMs, polymeric micelles
- PRR, pattern recognition receptor
- PS, pulmonary surfactant
- Pulmonary administration
- RIP2, receptor-interacting protein 2
- RISC, RNA-induced silencing complex
- RNAi, RNA interference
- ROS, reactive oxygen species
- SLN, solid lipid nanoparticle
- SNALP, stable nucleic acid lipid particle
- TGF-β, transforming growth factor-β
- TLR, Toll-like receptor
- TNF-α, tumor necrosis factor-α
- VALI, ventilator-associated lung injury
- VILI, ventilator-induced lung injury
- dsDNA, double-stranded DNA
- dsRNA, double-stranded RNA
- eggPG, l-α-phosphatidylglycerol
- mRNA, messenger RNA
- miRNA, microRNA
- pDNA, plasmid DNA
- shRNA, short RNA
- siRNA
- siRNA, small interfering RNA
Collapse
Affiliation(s)
- Makhloufi Zoulikha
- Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Qingqing Xiao
- Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - George Frimpong Boafo
- Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Marwa A. Sallam
- Department of Industrial Pharmacy, Faculty of Pharmacy, Alexandria University, Alexandria 21521, Egypt
| | - Zhongjian Chen
- Shanghai Skin Disease Hospital, Tongji University School of Medicine, Shanghai 200443, China
| | - Wei He
- Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
- Shanghai Skin Disease Hospital, Tongji University School of Medicine, Shanghai 200443, China
| |
Collapse
|
10
|
Nagaraju YH, Sapare A. A comprehensive review on the management of ARDS among pediatric patients. INDIAN JOURNAL OF RESPIRATORY CARE 2022. [DOI: 10.4103/ijrc.ijrc_158_22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
|
11
|
Tsao PC, Lin CH, Lee YS, Chen WY, Jeng MJ, Kou YR. Efficacy of intratracheal budesonide-surfactant combined therapy in surfactant-insufficient rat lungs with lipopolysaccharide insult. J Chin Med Assoc 2021; 84:783-790. [PMID: 34155174 DOI: 10.1097/jcma.0000000000000567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
BACKGROUND Intratracheal steroid therapy for lipopolysaccharide (LPS)-induced acute lung injury (ALI) remains challenging particularly in surfactant-insufficient lungs, a common problem of neonatal or pediatric ALI. Surfactant has been used as a vehicle for intratracheal steroid in the treatment of other types of ALI. This study investigated the efficacy of intratracheal budesonide (BUD) delivered by two concentrations of surfactant in the treatment of LPS-induced ALI in surfactant-insufficient rat lungs. METHODS Male adult rats were anesthetized and ventilated. Our ALI model was established by repeated saline lavage to produce surfactant insufficiency, followed by intratracheal LPS instillation. Five study groups (n = 5 for each) with different intratracheal treatments following ALI were used: control (no treatment), BUD (NS-BUD; BUD in saline), DS-BUD (BUD in diluted surfactant), FS-BUD (BUD in full-strength surfactant), FS (full-strength surfactant). Cardiopulmonary variables were monitored 4 hours post injury. Histological and immunohistochemical assessments of the lungs were performed. RESULTS The FS-BUD and FS groups presented better gas exchange, less metabolic acidosis, less oxygen index, and more stable hemodynamic changes than the DS-BUD, NS-BUD, and control groups. The total lung injury scores assessed by histological examination were ordered as follows: FS-BUD < DS-BUD or FS < NS-BUD < control. The immunostaining intensities of lung myeloperoxidase showed the following order: NS-BUD, DS-BUD, or FS-BUD < control or FS. Only the FS-BUD group displayed a smaller immunostaining intensity of lung tumor necrosis factor (TNF)-α than the control group. CONCLUSION Among our therapeutic strategies, intratracheal BUD delivered by full-strength surfactant confers an optimal protection against LPS-induced ALI in surfactant-insufficient rat lungs.
Collapse
Affiliation(s)
- Pei-Chen Tsao
- Department of Pediatrics, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
- Department and Institute of Physiology, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan, ROC
- Department of Pediatrics, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan, ROC
| | - Chih-Hsueh Lin
- Department and Institute of Physiology, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan, ROC
- Department of Nutrition, Hung-Kuang University, Taichung, Taiwan, ROC
| | - Yu-Sheng Lee
- Department of Pediatrics, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
- Department of Pediatrics, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan, ROC
| | - Wei-Yu Chen
- Department of Pediatrics, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
- Department of Pediatrics, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan, ROC
- Institute of Emergency and Critical Care Medicine, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan, ROC
| | - Mei-Jy Jeng
- Department of Pediatrics, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
- Department of Pediatrics, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan, ROC
- Institute of Emergency and Critical Care Medicine, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan, ROC
| | - Yu Ru Kou
- Department and Institute of Physiology, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan, ROC
- Institute of Emergency and Critical Care Medicine, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan, ROC
| |
Collapse
|
12
|
Rong Z, Mo L, Pan R, Zhu X, Cheng H, Li M, Yan L, Lang Y, Zhu X, Chen L, Xia S, Han J, Chang L. Bovine surfactant in the treatment of pneumonia-induced-neonatal acute respiratory distress syndrome (NARDS) in neonates beyond 34 weeks of gestation: a multicentre, randomized, assessor-blinded, placebo-controlled trial. Eur J Pediatr 2021; 180:1107-1115. [PMID: 33084980 PMCID: PMC7575859 DOI: 10.1007/s00431-020-03821-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 09/21/2020] [Accepted: 09/24/2020] [Indexed: 11/30/2022]
Abstract
Neonatal acute respiratory distress syndrome (NARDS) reflects pulmonary surfactant dysfunction, and the usage of bovine surfactant (Calsurf) supplement may therefore be beneficial. To determine whether bovine surfactant given in NARDS can improve oxygenation and survival rate, we conducted a multicenter, randomized trial between January 2018 and June 2019, and we compared Calsurf treatment to controls in neonates with pneumonia accompanied by NARDS. Neonates who met the Montreux criteria definition of NARDS were included, and those with congenital heart and lung malformations were excluded. Primary outcomes were oxygenation index (OI) after Calsurf administration, and secondary outcomes were mortality, and duration of ventilator and oxygen between the two groups, and also other morbidities. Cumulatively, 328 neonates were recruited and analyzed, 162 in the control group, and 166 in the Calsurf group. The results shows that OI in the Calsurf group were significantly lower than that in the control group at 4 h (7.2 ± 2.7 and 11.4 ± 9.1, P = 0.001); similarly, OI in the Calsurf group were significantly lower than in the control group at 12 h ( 7.5 ± 3.1 and 11.2 ± 9.2, P = 0.001). Mortality and duration of ventilator support or oxygen use between the two groups were not significantly different.Conclusion: Calsurf acutely improved OI immediately after administration in pneumonia-induced NARDS; although, we observed no significant decrease in mortality, duration of ventilator or oxygen, or major morbidity. What is known: • The definition proposed as the Monteux criteria for neonatal acute respiratory distress syndrome (NARDS). • Surfactant acutely improved oxygenation and significantly decreased mortality in children and adolescents with acute lung injury. What is new: • This is the first large randomized controlled trail to study on surfactant treatment of neonates with acute respiratory distress syndromes. • Surfactant acutely improved oxygenation immediately after administration in pneumonia-induced NARDS at a gestational age beyond 34 weeks.
Collapse
Affiliation(s)
- Zhihui Rong
- Department of Neonatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 China
| | - Luxia Mo
- Department of Neonatology, Wuhan Maternal and Child Healthcare Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, China
| | - Rui Pan
- Department of Neonatology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, China
| | - Xiaofang Zhu
- Department of Neonatology, Jingzhou Central Hospital, Jingzhou, China
| | - Hongbin Cheng
- Department of Neonatology, HuangShi Maternal and Child Healthcare Hospital, Huangshi, China
| | - Maojun Li
- Department of Neonatology, Sichuan Academy of Medical Sciences and Sichuan Provincial People’s Hospital, Sichuan, China
| | - Lubiao Yan
- Department of Neonatology, Maternal and Child Healthcare Hospital, Nanjing Medical University, Nanjing, China
| | - Yujie Lang
- Department of Neonatology, Children’s Hospital of Jinan, Jinan, China
| | - Xiaoshan Zhu
- Department of Neonatology, Anhui Provincial Children’s Hospital, Hefei, China
| | - Liping Chen
- Department of Neonatology, Jiangxi Provincial Children’s Hospital, Jiangxi, China
| | - Shiwen Xia
- Department of Neonatology, Hubei Maternity and Child Heath Hospital, Wuhan, China
| | - Jun Han
- Department of Neonatology, The First Hospital of Jilin University, Changchun, China
| | - Liwen Chang
- Department of Neonatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
13
|
Zhang Q, Hu WT, Yin F, Qian H, Wang Y, Li BR, Qian J, Tang YJ, Ning BT. The Clinical Characteristics of ARDS in Children With Hematological Neoplasms. Front Pediatr 2021; 9:696594. [PMID: 34307258 PMCID: PMC8295493 DOI: 10.3389/fped.2021.696594] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Accepted: 05/31/2021] [Indexed: 12/27/2022] Open
Abstract
In order to explore the clinical characteristics of pediatric patients admitted to the pediatric intensive care unit (PICU) who suffered from hematological neoplasms complicated with acute respiratory distress syndrome (ARDS), we retrospectively analyzed 45 ARDS children with hematological neoplasms who were admitted to the PICU of Shanghai Children's Medical Center from January 1, 2014, to December 31, 2020. The 45 children were divided into a survival group and a non-survival group, a pulmonary ARDS group and an exogenous pulmonary ARDS group, and an agranulocytosis group and a non-agranulocytosis group, for statistical analysis. The main clinical manifestations were fever, cough, progressive dyspnea, and hypoxemia; 55.6% (25/45) of the children had multiple organ dysfunction syndrome (MODS). The overall mortality rate was 55.6% (25/45). The vasoactive inotropic score (VIS), pediatric critical illness scoring (PCIS), average fluid volume in the first 3 days and the first 7 days, and the incidence of MODS in the non-survival group were all significantly higher than those in the survival group (P < 0.05). However, total length of mechanical ventilation and length of hospital stay and PICU days in the non-survival group were significantly lower than those in the survival group (P < 0.05). The PCIS (OR = 0.832, P = 0.004) and the average fluid volume in the first 3 days (OR = 1.092, P = 0.025) were independent risk factors for predicting death. Children with exogenous pulmonary ARDS were more likely to have MODS than pulmonary ARDS (P < 0.05). The mean values of VIS, C-reactive protein (CRP), and procalcitonin (PCT) in children with exogenous pulmonary ARDS were also higher (P < 0.05). After multivariate analysis, PCT was independently related to exogenous pulmonary ARDS. The total length of hospital stay, peak inspiratory pressure (PIP), VIS, CRP, and PCT in the agranulocytosis group were significantly higher than those in the non-agranulocytosis group (P < 0.05). Last, CRP and PIP were independently related to agranulocytosis. In conclusion, children with hematological neoplasms complicated with ARDS had a high overall mortality and poor prognosis. Children complicated with MODS, positive fluid balance, and high VIS and PCIS scores were positively correlated with mortality. In particular, PCIS score and average fluid volume in the first 3 days were independent risk factors for predicting death. Children with exogenous pulmonary ARDS and children with agranulocytosis were in a severely infected status and more critically ill.
Collapse
Affiliation(s)
- Qiao Zhang
- Department of Intensive Care Medicine, Shanghai Children's Medical Center, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Wen-Ting Hu
- Department of Hematology and Oncology, Shanghai Children's Medical Center, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Fan Yin
- Department of Intensive Care Medicine, Shanghai Children's Medical Center, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Han Qian
- Department of Intensive Care Medicine, Shanghai Children's Medical Center, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Ying Wang
- Department of Intensive Care Medicine, Shanghai Children's Medical Center, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Bi-Ru Li
- Department of Intensive Care Medicine, Shanghai Children's Medical Center, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Juan Qian
- Department of Intensive Care Medicine, Shanghai Children's Medical Center, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Yan-Jing Tang
- Department of Hematology and Oncology, Shanghai Children's Medical Center, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Bo-Tao Ning
- Department of Intensive Care Medicine, Shanghai Children's Medical Center, Shanghai Jiaotong University School of Medicine, Shanghai, China.,Shanghai Engineering Research Center of Intelligence Pediatrics (SERCIP), Shanghai, China
| |
Collapse
|
14
|
COVID-19 lockdown: de-risking exit by protecting the lung with leukaemia inhibitory factor (LIF). MEDICINE IN DRUG DISCOVERY 2020. [PMCID: PMC7227543 DOI: 10.1016/j.medidd.2020.100043] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
There are two key needs in COVID-19 management: (i) to reduce SARS-CoV-2 viral infection rate; and (ii) to reduce death rate of those infected - the subject of this commentary. The current WHO estimated global mortality rate is 3.4% (March 2020) and the global death toll has now past 200,000 (April 2020). Without therapy the COVID-19 pandemic is escalating exponentially: from the first reported death in Wuhan China 10th January 2020, it took 91 days for the global death toll to pass 100,000 - then a further 16 days to reach 200,000. A vaccination program will take 1–2 years to roll out, once safety and efficacy is proven. Anti-virals are being sought mainly amongst repurposed drug candidates but also with combinatorial screening of libraries, for example to block virus binding angiotensin converting enzyme 2 (ACE2) - ACE2 providing the receptor on cells that allows viral entry. Cell-based approaches include stem cells and exosomes but these will never meet scale of need whilst also carrying risk of viral transmission if contaminated. Countries have introduced Population control with social distancing and lockdown to isolate individuals: this has reduced infectivity rate - “R” - where R denotes the average number of people an infected person will spread the illness to. But, after lockdown, the virus remains: the probability of R increasing again is high. The new danger is exit from lockdown. Here, leukaemia inhibitory factor (LIF) represents an untapped resource to boost the lung's own resistance to developing COVID-19 - reducing risk of severe disease as nations cautiously leave lockdown to return to normality. Leukaemia Inhibitory Factor (LIF) protects the lung from SARS-induced viral Inflammation Therapeutic LIF will reduce mortality from the COVID-19 pandemic Exit from Lock-Down will be de-risked, benefiting global recovery both in health and the economy.
Collapse
|
15
|
Tobias JD. Sugammadex: Applications in Pediatric Critical Care. J Pediatr Intensive Care 2020; 9:162-171. [PMID: 32685243 DOI: 10.1055/s-0040-1705133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Accepted: 01/27/2020] [Indexed: 10/24/2022] Open
Abstract
Sugammadex is a novel pharmacologic agent, which reverses neuromuscular blockade with a mechanism that differs from acetylcholinesterase inhibitors such as neostigmine. There is a growing body of literature demonstrating its efficacy in pediatric patients of all ages. Prospective trials have demonstrated a more rapid and more complete reversal of rocuronium-induced neuromuscular blockade than the acetylcholinesterase inhibitor, neostigmine. Unlike the acetylcholinesterase inhibitors, sugammadex effectively reverses intense or complete neuromuscular blockade. It may also be effective in situations where reversal of neuromuscular blockade is problematic including patients with neuromyopathic conditions or when acetylcholinesterase inhibitors are contraindicated. This article reviews the physiology of neuromuscular transmission as well as the published literature, regarding the use of sugammadex in pediatric population including the pediatric intensive care unit population. Clinical applications are reviewed, adverse effects are discussed, and dosing algorithms are presented.
Collapse
Affiliation(s)
- Joseph D Tobias
- Department of Anesthesiology and Pain Medicine, Nationwide Children's Hospital, Columbus, Ohio, United States.,Department of Anesthesiology and Pain Medicine, The Ohio State University College of Medicine, Columbus, Ohio, United States.,Department of Pediatrics, The Ohio State University College of Medicine, Columbus, Ohio, United States
| |
Collapse
|
16
|
Rosenberg L, Traube C. Sedation strategies in children with pediatric acute respiratory distress syndrome (PARDS). ANNALS OF TRANSLATIONAL MEDICINE 2019; 7:509. [PMID: 31728362 DOI: 10.21037/atm.2019.09.16] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
In this review, we discuss the changing landscape of sedation in mechanically ventilated children with pediatric acute respiratory distress syndrome (PARDS). While previous approaches advocated for early and deep sedation with benzodiazepines, emerging literature has highlighted the benefits of light sedation and use of non-benzodiazepine sedating agents, such as dexmedetomidine. Recent studies have emphasized the importance of monitoring multiple factors including, but not limited to, sedation depth, analgesia efficacy, opiate withdrawal, and development of delirium. Through this approach, we hope to improve PARDS outcomes. Overall, more research is needed to further our understanding of the best sedation strategies in children with PARDS.
Collapse
Affiliation(s)
- Lynne Rosenberg
- Department of Pediatrics, Weill Cornell Medical College, New York, NY, USA
| | - Chani Traube
- Department of Pediatrics, Weill Cornell Medical College, New York, NY, USA
| |
Collapse
|
17
|
Hanudel MR, Zinter MS, Chen L, Gala K, Lim M, Guglielmo M, Deshmukh T, Vangala S, Matthay M, Sapru A. Plasma total fibroblast growth factor 23 levels are associated with acute kidney injury and mortality in children with acute respiratory distress syndrome. PLoS One 2019; 14:e0222065. [PMID: 31487315 PMCID: PMC6728039 DOI: 10.1371/journal.pone.0222065] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Accepted: 08/21/2019] [Indexed: 11/24/2022] Open
Abstract
Acute respiratory distress syndrome (ARDS) has high rates of mortality and multisystem morbidity. Pre-clinical data suggest that fibroblast growth factor 23 (FGF23) may contribute to pulmonary pathology, and FGF23 is associated with mortality and morbidity, including acute kidney injury (AKI), in non-ARDS cohorts. Here, we assess whether FGF23 is associated with AKI and/or mortality in a cohort of 161 pediatric ARDS patients. Plasma total (intact + C-terminal) FGF23 and intact FGF23 concentrations were measured within 24 hours of ARDS diagnosis (Day 1), and associations with Day 3 AKI and 60-day mortality were evaluated. 35 patients (22%) developed AKI by 3 days post-ARDS diagnosis, and 25 (16%) died by 60 days post-ARDS diagnosis. In unadjusted models, higher Day 1 total FGF23 was associated with Day 3 AKI (odds ratio (OR) 2.22 [95% confidence interval (CI) 1.62, 3.03], p<0.001), but Day 1 intact FGF23 was not. In a model adjusted for demographics and disease severity, total FGF23 remained associated with AKI (OR 1.52 [95% CI 1.02, 2.26], p = 0.039). In unadjusted models, both higher Day 1 total and intact FGF23 were associated with 60-day mortality (OR 1.43 [95% CI 1.07, 1.91], p = 0.014; and OR 1.44 [95% CI 1.02, 2.05], p = 0.039, respectively). In the adjusted model, only total FGF23 remained associated with 60-day mortality (OR 1.62 [95% CI 1.07, 2.45], p = 0.023). In a subgroup analysis of patients with Day 1 plasma IL-6 concentrations available, inflammation partially mediated the association between total FGF23 and AKI. Our data suggest both inflammation-dependent and inflammation-independent associations between total FGF23 and clinical outcomes in pediatric ARDS patients.
Collapse
Affiliation(s)
- Mark R. Hanudel
- Department of Pediatrics, David Geffen School of Medicine at UCLA, Los Angeles, CA, United States of America
- * E-mail:
| | - Matthew S. Zinter
- Department of Pediatrics, UCSF School of Medicine, San Francisco, CA, United States of America
| | - Lucia Chen
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, United States of America
| | - Kinisha Gala
- Department of Pediatrics, David Geffen School of Medicine at UCLA, Los Angeles, CA, United States of America
| | - Michelle Lim
- Department of Pediatrics, David Geffen School of Medicine at UCLA, Los Angeles, CA, United States of America
| | - Mona Guglielmo
- Department of Pediatrics, David Geffen School of Medicine at UCLA, Los Angeles, CA, United States of America
| | - Tanaya Deshmukh
- Department of Pediatrics, David Geffen School of Medicine at UCLA, Los Angeles, CA, United States of America
| | - Sitaram Vangala
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, United States of America
| | - Michael Matthay
- Department of Medicine, UCSF School of Medicine, San Francisco, CA, United States of America
| | - Anil Sapru
- Department of Pediatrics, David Geffen School of Medicine at UCLA, Los Angeles, CA, United States of America
| |
Collapse
|
18
|
Orloff KE, Turner DA, Rehder KJ. The Current State of Pediatric Acute Respiratory Distress Syndrome. PEDIATRIC ALLERGY IMMUNOLOGY AND PULMONOLOGY 2019; 32:35-44. [PMID: 31236307 PMCID: PMC6589490 DOI: 10.1089/ped.2019.0999] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Accepted: 03/24/2019] [Indexed: 12/16/2022]
Abstract
Pediatric acute respiratory distress syndrome (PARDS) is a significant cause of morbidity and mortality in children. Children with PARDS often require intensive care admission and mechanical ventilation. Unfortunately, beyond lung protective ventilation, there are limited data to support our management strategies in PARDS. The Pediatric Acute Lung Injury Consensus Conference (PALICC) offered a new definition of PARDS in 2015 that has improved our understanding of the true epidemiology and heterogeneity of the disease as well as risk stratification. Further studies will be crucial to determine optimal management for varying disease severity. This review will present the physiologic basis of PARDS, describe the unique pediatric definition and risk stratification, and summarize the current evidence for current standards of care as well as adjunctive therapies.
Collapse
Affiliation(s)
- Kirsten E Orloff
- Division of Pediatric Critical Care Medicine, Department of Pediatrics, Duke Children's Hospital, Durham, North Carolina
| | - David A Turner
- Division of Pediatric Critical Care Medicine, Department of Pediatrics, Duke Children's Hospital, Durham, North Carolina
| | - Kyle J Rehder
- Division of Pediatric Critical Care Medicine, Department of Pediatrics, Duke Children's Hospital, Durham, North Carolina
| |
Collapse
|
19
|
Kim BR, Kim SY, Sol IS, Kim YH, Kim KW, Sohn MH, Kim KE. Clinical application of the Pediatric Acute Lung Injury Consensus Conference definition of acute respiratory distress syndrome. ALLERGY ASTHMA & RESPIRATORY DISEASE 2019. [DOI: 10.4168/aard.2019.7.1.44] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Affiliation(s)
- Byuh Ree Kim
- Department of Pediatrics, Severance Children's Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Soo Yeon Kim
- Department of Pediatrics, Severance Children's Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - In Suk Sol
- Department of Pediatrics, Severance Children's Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Yoon Hee Kim
- Department of Pediatrics, Severance Children's Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Kyung Won Kim
- Department of Pediatrics, Severance Children's Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Myung Hyun Sohn
- Department of Pediatrics, Severance Children's Hospital, Yonsei University College of Medicine, Seoul, Korea
| | | |
Collapse
|
20
|
Kamat PP, Kudchadkar SR, Simon HK. Sedative and Anesthetic Neurotoxicity in Infants and Young Children: Not Just an Operating Room Concern. J Pediatr 2019; 204:285-290. [PMID: 30270160 DOI: 10.1016/j.jpeds.2018.08.039] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Revised: 07/15/2018] [Accepted: 08/17/2018] [Indexed: 02/07/2023]
Affiliation(s)
- Pradip P Kamat
- Department of Pediatrics, Division of Critical Care Medicine, Emory University School of Medicine, Atlanta, GA; Children's Sedation Services at Egleston, Children's Healthcare of Atlanta at Egleston, Atlanta, GA.
| | - Sapna R Kudchadkar
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Charlotte R. Bloomberg Children's Center, Baltimore, MD; Department of Pediatrics, Johns Hopkins University School of Medicine, Charlotte R. Bloomberg Children's Center, Baltimore, MD; Department of Physical Medicine and Rehabilitation, Johns Hopkins University School of Medicine, Charlotte R. Bloomberg Children's Center, Baltimore, MD
| | - Harold K Simon
- Department of Pediatrics, Division of Pediatric Emergency Medicine, Emory University School of Medicine, Atlanta, GA; Department of Emergency Medicine, Emory University School of Medicine, Atlanta, GA
| |
Collapse
|