51
|
Paeoniflorin protects 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-induced Parkinson's disease mice by inhibiting oxidative stress and neuronal apoptosis through activating the Nrf2/HO-1 signaling pathway. Neuroreport 2023; 34:255-266. [PMID: 36881748 DOI: 10.1097/wnr.0000000000001884] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/09/2023]
Abstract
OBJECTIVES This study aimed to explore the neuroprotective effects of paeoniflorin on oxidative stress and apoptosis in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced Parkinson's disease (PD) mice. METHODS The effects of paeoniflorin on motor function in mice were evaluated by behavioral test. Then substantia nigra of mice were collected and neuronal damage was assessed using Nissl staining. Positive expression of tyrosine hydroxylase (TH) was detected by immunohistochemistry. Levels of malondialdehyde, superoxide dismutase (SOD) and glutathione were measured by biochemical method. terminal deoxynucleotidyl transferase-mediated dUTP-biotin nick end labeling assay was used to detect apoptosis of dopaminergic neurons. Western blotting and real-time fluorescence quantitative PCR were used to detect the protein and mRNA expressions of Nrf2, heme oxygenase-1 (HO-1), B-cell lymphoma-2(Bcl-2), Bax and cleaved caspase-3. RESULTS Paeoniflorin treatment significantly ameliorated the motor performance impairment in MPTP-induced PD mice. Moreover, it notably increased the positive expression rate of TH and reduced the damage and apoptosis of dopaminergic neurons in the substantia nigra. Furthermore, paeoniflorin increased the levels of SOD and glutathione and decreased the malondialdehyde content. It also promoted Nrf2 nuclear translocation, increased the protein and mRNA expressions of HO-1 and Bcl-2 and reduced the protein and mRNA expressions of BCL2-Associated X2 (Bax) and cleaved caspase-3. Treatment with the Nrf2 inhibitor, ML385, notably reduced the effects of paeoniflorin in MPTP-induced PD mice. CONCLUSIONS Neuroprotective effects of paeoniflorin in MPTP-induced PD mice may be mediated via inhibition of oxidative stress and apoptosis of dopaminergic neurons in substantia nigra through activation of the Nrf2/HO-1 signaling pathway.
Collapse
|
52
|
D'Alessandro A, Pastore A, Amadio P, D'Agostini M, Terreri S, Carsetti R, Argentieri M, Bernaschi P, Onetti Muda A, Porzio O, Dotta A, Salvatori G. Influence of Defatting and Pasteurization on Nutrients and Oxidative Stress Markers in Human Milk. J Hum Lact 2023; 39:278-287. [PMID: 36945737 DOI: 10.1177/08903344231156894] [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] [Indexed: 03/23/2023]
Abstract
BACKGROUND It is well known that the best nutritional option for infants is human milk, and that when breastfeeding is not possible, human milk banks are a possible alternative. However, in the case of infants with fat transport disorder like chylothorax, defatting of human milk is mandatory. RESEARCH AIM The aim of the study was to reduce milk fat content without reducing other nutrients, increasing oxidative stress, or introducing harmful microorganisms. METHODS In this prospective, cross-sectional, observational study, we examined the influence of defatting and pasteurization of 50 donor samples on fat, macro- and micronutrients, as well as on oxidative stress markers. RESULTS Low-temperature centrifugation proved to be very efficient in defatting, reducing the concentration of triglycerides by 85% and cholesterol by 50%. The macronutrients (proteins, albumin, and Immunoglobulin A) did not undergo significant changes due to defatting and pasteurization procedures, while iron decreased by 36%. However, as the majority of iron is retained, this result does not remarkably change the milk composition. Furthermore, oxidative stress markers and antioxidant levels were unchanged, and the milk result was microbiologically safe. CONCLUSIONS Cold milk centrifugation proved to be an effective technique that allows the reduction of human milk lipids. The determination of triglycerides and cholesterol can be used as an indicator of skimming. This procedure is not accompanied by substantial modifications of other components present in the milk.
Collapse
Affiliation(s)
| | - Anna Pastore
- Research Unit of Diagnostic and Management Innovations, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Patrizia Amadio
- Neonatal Intensive Care Unit and Human Milk Bank, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Matteo D'Agostini
- Clinical Laboratory Unit, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Sara Terreri
- Diagnostic Immunology Research Unit, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Rita Carsetti
- Diagnostic Immunology Research Unit, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Marta Argentieri
- Microbiology Laboratory, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Paola Bernaschi
- Microbiology Laboratory, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Andrea Onetti Muda
- Research Unit of Diagnostic and Management Innovations, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Ottavia Porzio
- Clinical Laboratory Unit, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
- Department of Experimental Medicine, University of Rome "Tor Vergata", Rome, Italy
| | - Andrea Dotta
- Neonatal Intensive Care Unit and Human Milk Bank, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Guglielmo Salvatori
- Neonatal Intensive Care Unit and Human Milk Bank, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| |
Collapse
|
53
|
Sankaran D, Giusto EM, Lesneski AL, Hardie ME, Joudi HM, Lane ECA, Hammitt VL, Tully KC, Vali P, Lakshminrusimha S. Randomized Trial of 21% versus 100% Oxygen during Chest Compressions Followed by Gradual versus Abrupt Oxygen Titration after Return of Spontaneous Circulation in Neonatal Lambs. CHILDREN 2023; 10:children10030575. [PMID: 36980132 PMCID: PMC10047452 DOI: 10.3390/children10030575] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 03/15/2023] [Accepted: 03/16/2023] [Indexed: 03/19/2023]
Abstract
The combination of perinatal acidemia with postnatal hyperoxia is associated with a higher incidence of hypoxic-ischemic encephalopathy (HIE) in newborn infants. In neonatal cardiac arrest, current International Liaison Committee on Resuscitation (ILCOR) and Neonatal Resuscitation Program (NRP) guidelines recommend increasing inspired O2 to 100% during chest compressions (CC). Following the return of spontaneous circulation (ROSC), gradual weaning from 100% O2 based on pulse oximetry (SpO2) can be associated with hyperoxia and risk for cerebral tissue injury owing to oxidative stress. We hypothesize that compared to gradual weaning from 100% O2 with titration based on preductal SpO2, abrupt or rapid weaning of inspired O2 to 21% after ROSC or use of 21% O2 during CC followed by upward titration of inspired O2 to achieve target SpO2 after ROSC will limit hyperoxia after ROSC. Nineteen lambs were randomized before delivery and asphyxial arrest was induced by umbilical cord occlusion. There was no difference in oxygenation during chest compressions between the three groups. Gradual weaning of inspired O2 from 100% O2 after ROSC resulted in supraphysiological PaO2 and higher cerebral oxygen delivery compared to 21% O2 during CC or 100% O2 during CC followed by abrupt weaning to 21% O2 after ROSC. The use of 21% O2 during CC was associated with very low PaO2 after ROSC and higher brain tissue lactic acid compared to other groups. Our findings support the current recommendations to use 100% O2 during CC and additionally suggest the benefit of abrupt decrease in inspired oxygen to 21% O2 after ROSC. Clinical studies are warranted to investigate optimal oxygen titration after chest compressions and ROSC during neonatal resuscitation.
Collapse
Affiliation(s)
- Deepika Sankaran
- Department of Pediatrics, University of California, Davis, Sacramento, CA 95817, USA
- Correspondence:
| | - Evan M. Giusto
- Department of Pediatrics, University of California, Davis, Sacramento, CA 95817, USA
| | - Amy L. Lesneski
- Department of Stem Cell Research, University of California, Davis, Sacramento, CA 95817, USA
| | - Morgan E. Hardie
- Department of Pediatrics, University of California, Davis, Sacramento, CA 95817, USA
| | - Houssam M. Joudi
- Department of Pediatrics, University of California, Davis, Sacramento, CA 95817, USA
| | - Emily C. A. Lane
- Department of Pediatrics, University of California, Davis, Sacramento, CA 95817, USA
| | - Victoria L. Hammitt
- Department of Stem Cell Research, University of California, Davis, Sacramento, CA 95817, USA
| | - Kirstie C. Tully
- Department of Stem Cell Research, University of California, Davis, Sacramento, CA 95817, USA
| | - Payam Vali
- Department of Pediatrics, University of California, Davis, Sacramento, CA 95817, USA
| | | |
Collapse
|
54
|
Relationship between large and small for gestational age and hospital readmission after postpartum discharge: a population-based, data-linkage study. Eur J Pediatr 2023; 182:2245-2252. [PMID: 36869901 DOI: 10.1007/s00431-023-04908-2] [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: 12/20/2022] [Revised: 02/17/2023] [Accepted: 02/26/2023] [Indexed: 03/05/2023]
Abstract
This study aims to determine the association of small for gestational age (SGA) and large for gestational age (LGA) at birth with hospital readmission after postpartum discharge for up to 28 days of delivery. This is a population-based, data-linkage study using the French National Uniform Hospital Discharge Database. "Healthy" singleton term infants born between January 1st, 2017, and November 30th, 2018, in the French South region were included. SGA and LGA were defined as birth weight < 10th and > 90th percentiles, respectively, according to sex and gestational age. A multivariable regression analysis was performed. Among 67,359 included infants, 2441 (3.6%) were readmitted, and 61% of them were hospitalized within 14 days postpartum. Hospitalized infants were more likely to be LGA at birth (10.3% vs. 8.6% in non-hospitalized infants, p < 0.01); the proportion of SGA infants did not differ between both groups. Compared to appropriate birth weight for GA (AGA) infants, LGA infants were more often hospitalized for infectious diseases (57.7% vs. 51.3%, p = 0.05). After regression analysis, LGA infants had a 20% higher odds of being hospitalized than those born AGA (aOR (95%CI) = 1.21 (1.06-1.39)), while aOR (95%CI) for SGA was 1.11 (0.96-1.28). CONCLUSION In contrast to SGA, LGA was associated with hospital readmission during the first month of life. Follow-up protocols that include LGA should be evaluated. WHAT IS KNOWN • Newborns are at high risk of hospital readmission during the postpartum period. • However, the influence of appropriateness for gestational age at birth, i.e. being born small for gestational age (SGA) or large for gestational age (LGA), has been little evaluated. WHAT IS NEW • In contrast to SGA born infants, we found that infants born LGA were at high risk of hospital admission and the main cause was infectious diseases. • This population should be considered at risk of early adverse outcomes and should require attentive medical follow-up after postpartum discharge.
Collapse
|
55
|
Hundscheid TM, Huizing MJ, Villamor-Martinez E, Bartoš F, Villamor E. Association of Funisitis with Short-Term Outcomes of Prematurity: A Frequentist and Bayesian Meta-Analysis. Antioxidants (Basel) 2023; 12:534. [PMID: 36830092 PMCID: PMC9951960 DOI: 10.3390/antiox12020534] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 02/08/2023] [Accepted: 02/15/2023] [Indexed: 02/22/2023] Open
Abstract
The fetal systemic inflammatory response associated with intra-amniotic inflammation may play a key role in the pathogenesis of complications of preterm birth. Funisitis is the histologic equivalent of the fetal inflammatory response, whereas chorioamnionitis represents a maternal inflammatory response. We conducted a frequentist and Bayesian model average (BMA) meta-analysis of studies investigating the effects of funisitis on short-term outcomes of prematurity. Thirty-three studies (12,237 infants with gestational age ≤ 34 weeks) were included. Frequentist meta-analysis showed that funisitis was associated with an increased risk of any bronchopulmonary dysplasia (BPD), moderate/severe BPD, retinopathy of prematurity (ROP), intraventricular hemorrhage (IVH), periventricular leukomalacia (PVL), any sepsis, early-onset sepsis (EOS), and mortality. However, Bayesian meta-analysis showed that the evidence in favor of the alternative hypothesis (i.e., funisitis is associated with an increased risk of developing the outcome) was strong for any IVH, moderate for severe IVH and EOS, and weak for the other outcomes. When the control group was restricted to infants having chorioamnionitis without funisitis, the only outcome associated with funisitis was any IVH. In conclusion, our data suggest that the presence of funisitis does not add an additional risk to preterm birth when compared to chorioamnionitis in the absence of fetal inflammatory response.
Collapse
Affiliation(s)
- Tamara Maria Hundscheid
- Department of Pediatrics, University Medical Center (MUMC+), School for Oncology and Reproduction (GROW), Maastricht University, 6202 AZ Maastricht, The Netherlands
| | - Maurice Jacob Huizing
- Department of Pediatrics, University Medical Center (MUMC+), School for Oncology and Reproduction (GROW), Maastricht University, 6202 AZ Maastricht, The Netherlands
| | | | - František Bartoš
- Department of Psychology, University of Amsterdam, 1001 NK Amsterdam, The Netherlands
| | - Eduardo Villamor
- Department of Pediatrics, University Medical Center (MUMC+), School for Oncology and Reproduction (GROW), Maastricht University, 6202 AZ Maastricht, The Netherlands
| |
Collapse
|
56
|
Talisman S, Guedalia J, Farkash R, Avitan T, Srebnik N, Kasirer Y, Schimmel MS, Ghanem D, Unger R, Granovsky SG. Neonatal intensive care admission for term neonates and subsequent childhood mortality: a retrospective linkage study. BMC Med 2023; 21:44. [PMID: 36747227 PMCID: PMC9903506 DOI: 10.1186/s12916-023-02744-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 01/18/2023] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Neonatal intensive care unit (NICU) admission among term neonates is a rare event. The aim of this study was to study the association of the NICU admission of term neonates on the risk of long-term childhood mortality. METHODS A single-center case-control retrospective study between 2005 and 2019, including all in-hospital ≥ 37 weeks' gestation singleton live-born neonates. The center perinatal database was linked with the birth and death certificate registries of the Israeli Ministry of Internal Affairs. The primary aim of the study was to study the association between NICU admission and childhood mortality throughout a 15-year follow-up period. RESULTS During the study period, 206,509 births were registered; 192,527 (93.22%) term neonates were included in the study; 5292 (2.75%) were admitted to NICU. Throughout the follow-up period, the mortality risk for term neonates admitted to the NICU remained elevated; hazard ratio (HR), 19.72 [14.66, 26.53], (p < 0.001). For all term neonates, the mortality rate was 0.16% (n = 311); 47.9% (n = 149) of those had records of a NICU admission. The mortality rate by time points (ratio1:10,0000 births) related to the age at death during the follow-up period was as follows: 29, up to 7 days; 20, 7-28 days; 37, 28 days to 6 months; 21, 6 months to 1 year; 19, 1-2 years; 9, 2-3 years; 10, 3-4 years; and 27, 4 years and more. Following the exclusion of congenital malformations and chromosomal abnormalities, NICU admission remained the most significant risk factor associated with mortality of the study population, HRs, 364.4 [145.3; 913.3] for mortality in the first 7 days of life; 19.6 [12.1; 32.0] for mortality from 28 days through 6 months of life and remained markedly elevated after age 4 years; HR, 7.1 [3.0; 17.0]. The mortality risk related to the NICU admission event, adjusted for admission diagnoses remained significant; HR = 8.21 [5.43; 12.4]. CONCLUSIONS NICU admission for term neonates is a pondering event for the risk of long-term childhood mortality. This group of term neonates may benefit from focused health care.
Collapse
Affiliation(s)
- Shahar Talisman
- Department of Obstetrics & Gynecology, Shaare Zedek Medical Center, Affiliated with the Hebrew University-Hadassah School of Medicine, Jerusalem, Israel
| | - Joshua Guedalia
- The Mina and Everard Goodman Faculty of Life Sciences, Bar Ilan University, Ramat-Gan, Israel
| | - Rivka Farkash
- Department of Obstetrics & Gynecology, Shaare Zedek Medical Center, Affiliated with the Hebrew University-Hadassah School of Medicine, Jerusalem, Israel
| | - Tehila Avitan
- Department of Obstetrics & Gynecology, Shaare Zedek Medical Center, Affiliated with the Hebrew University-Hadassah School of Medicine, Jerusalem, Israel.
| | - Naama Srebnik
- Department of Obstetrics & Gynecology, Shaare Zedek Medical Center, Affiliated with the Hebrew University-Hadassah School of Medicine, Jerusalem, Israel
| | - Yair Kasirer
- Department of Pediatrics, Shaare Zedek Medical Center, Affiliated with the Hebrew University-Hadassah School of Medicine, Jerusalem, Israel
| | - Michael S Schimmel
- Department of Pediatrics, Shaare Zedek Medical Center, Affiliated with the Hebrew University-Hadassah School of Medicine, Jerusalem, Israel
| | - Donia Ghanem
- Department of Obstetrics & Gynecology, Shaare Zedek Medical Center, Affiliated with the Hebrew University-Hadassah School of Medicine, Jerusalem, Israel
| | - Ron Unger
- The Mina and Everard Goodman Faculty of Life Sciences, Bar Ilan University, Ramat-Gan, Israel
| | - Sorina Grisaru Granovsky
- Department of Obstetrics & Gynecology, Shaare Zedek Medical Center, Affiliated with the Hebrew University-Hadassah School of Medicine, Jerusalem, Israel
| |
Collapse
|
57
|
Abstract
Bronchopulmonary dysplasia (BPD) in neonates is the most common pulmonary disease that causes neonatal mortality, has complex pathogenesis, and lacks effective treatment. It is associated with chronic obstructive pulmonary disease, pulmonary hypertension, and right ventricular hypertrophy. The occurrence and development of BPD involve various factors, of which premature birth is the most crucial reason for BPD. Under the premise of abnormal lung structure and functional product, newborns are susceptible to damage to oxides, free radicals, hypoxia, infections and so on. The most influential is oxidative stress, which induces cell death in different ways when the oxidative stress balance in the body is disrupted. Increasing evidence has shown that programmed cell death (PCD), including apoptosis, necrosis, autophagy, and ferroptosis, plays a significant role in the molecular and biological mechanisms of BPD and the further development of the disease. Understanding the mode of PCD and its signaling pathways can provide new therapeutic approaches and targets for the clinical treatment of BPD. This review elucidates the mechanism of BPD, focusing on the multiple types of PCD in BPD and their molecular mechanisms, which are mainly based on experimental results obtained in rodents.
Collapse
|
58
|
Protective Effects of Early Caffeine Administration in Hyperoxia-Induced Neurotoxicity in the Juvenile Rat. Antioxidants (Basel) 2023; 12:antiox12020295. [PMID: 36829854 PMCID: PMC9952771 DOI: 10.3390/antiox12020295] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 01/12/2023] [Accepted: 01/24/2023] [Indexed: 01/31/2023] Open
Abstract
High-risk preterm infants are affected by a higher incidence of cognitive developmental deficits due to the unavoidable risk factor of oxygen toxicity. Caffeine is known to have a protective effect in preventing bronchopulmonary dysplasia associated with improved neurologic outcomes, although very early initiation of therapy is controversial. In this study, we used newborn rats in an oxygen injury model to test the hypothesis that near-birth caffeine administration modulates neuronal maturation and differentiation in the hippocampus of the developing brain. For this purpose, newborn Wistar rats were exposed to 21% or 80% oxygen on the day of birth for 3 or 5 days and treated with vehicle or caffeine (10 mg/kg/48 h). Postnatal exposure to 80% oxygen resulted in a drastic reduction of associated neuronal mediators for radial glia, mitotic/postmitotic neurons, and impaired cell-cycle regulation, predominantly persistent even after recovery to room air until postnatal day 15. Systemic caffeine administration significantly counteracted the effects of oxygen insult on neuronal maturation in the hippocampus. Interestingly, under normoxia, caffeine inhibited the transcription of neuronal mediators of maturing and mature neurons. The early administration of caffeine modulated hyperoxia-induced decreased neurogenesis in the hippocampus and showed neuroprotective properties in the neonatal rat oxygen toxicity model.
Collapse
|
59
|
Saugstad OD. Oxygenation of the newborn. The impact of one molecule on newborn lives. J Perinat Med 2023; 51:20-26. [PMID: 35848535 DOI: 10.1515/jpm-2022-0259] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 06/06/2022] [Indexed: 01/21/2023]
Abstract
Hypoxanthine is a purine metabolite which increases during hypoxia and therefore is an indicator of this condition. Further, when hypoxanthine is oxidized to uric acid in the presence of xanthine oxidase, oxygen radicals are generated. This was the theoretical basis for suggesting and studying, beginning in the 1990s, resuscitation of newborn infants with air instead of the traditional 100% O2. These studies demonstrated a 30% reduction in mortality when resuscitation of term and near term infants was carried out with air compared to pure oxygen. The mechanism for this is not fully understood, however the hypoxanthine -xanthine oxidase system increases oxidative stress and plays a role in regulation of the perinatal circulation. Further, hyperoxic resuscitation inhibits mitochondrial function, and one reason may be that genes involved in ATP production are down-regulated. Thus, the study of one single molecule, hypoxanthine, has contributed to the global prevention of an estimated 2-500,000 annual infant deaths.
Collapse
Affiliation(s)
- Ola Didrik Saugstad
- Department of Pediatric Research, University of Oslo and Oslo University hospital, Oslo, Norway
- Anne and Robert H. Lurie Children's Hospital of Chicago, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| |
Collapse
|
60
|
Wang X, Lv S, Sun J, Zhang M, Zhang L, Sun Y, Zhao Z, Wang D, Zhao X, Zhang J. Caffeine reduces oxidative stress to protect against hyperoxia-induced lung injury via the adenosine A2A receptor/cAMP/PKA/Src/ERK1/2/p38MAPK pathway. Redox Rep 2022; 27:270-278. [DOI: 10.1080/13510002.2022.2143114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Xijuan Wang
- Department of Paediatrics, Henan Provincial People’s Hospital, Zhengzhou University People’s Hospital, Henan University People’s Hospital, Zhengzhou, People’s Republic of China
| | - Shuai Lv
- Department of Gastroenterology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, People’s Republic of China
| | - Jianwei Sun
- Department of Paediatrics, Henan Provincial People’s Hospital, Zhengzhou University People’s Hospital, Henan University People’s Hospital, Zhengzhou, People’s Republic of China
| | - Meihui Zhang
- Department of Paediatrics, Henan Provincial People’s Hospital, Zhengzhou University People’s Hospital, Henan University People’s Hospital, Zhengzhou, People’s Republic of China
| | - Lei Zhang
- Department of Paediatrics, Henan Provincial People’s Hospital, Zhengzhou University People’s Hospital, Henan University People’s Hospital, Zhengzhou, People’s Republic of China
| | - Yan Sun
- Department of Paediatrics, Henan Provincial People’s Hospital, Zhengzhou University People’s Hospital, Henan University People’s Hospital, Zhengzhou, People’s Republic of China
| | - Ziyan Zhao
- Department of Paediatrics, Henan Provincial People’s Hospital, Zhengzhou University People’s Hospital, Henan University People’s Hospital, Zhengzhou, People’s Republic of China
| | - Dandan Wang
- Department of Paediatrics, Henan Provincial People’s Hospital, Zhengzhou University People’s Hospital, Henan University People’s Hospital, Zhengzhou, People’s Republic of China
| | - Xinjing Zhao
- Department of Paediatrics, Henan Provincial People’s Hospital, Zhengzhou University People’s Hospital, Henan University People’s Hospital, Zhengzhou, People’s Republic of China
| | - Jiajie Zhang
- Department of Paediatrics, Henan Provincial People’s Hospital, Zhengzhou University People’s Hospital, Henan University People’s Hospital, Zhengzhou, People’s Republic of China
| |
Collapse
|
61
|
Perinatal Oxidative Stress and Kidney Health: Bridging the Gap between Animal Models and Clinical Reality. Antioxidants (Basel) 2022; 12:antiox12010013. [PMID: 36670875 PMCID: PMC9855228 DOI: 10.3390/antiox12010013] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2022] [Revised: 12/02/2022] [Accepted: 12/19/2022] [Indexed: 12/24/2022] Open
Abstract
Oxidative stress arises when the generation of reactive oxygen species or reactive nitrogen species overwhelms antioxidant systems. Developing kidneys are vulnerable to oxidative stress, resulting in adult kidney disease. Oxidative stress in fetuses and neonates can be evaluated by assessing various biomarkers. Using animal models, our knowledge of oxidative-stress-related renal programming, the molecular mechanisms underlying renal programming, and preventive interventions to avert kidney disease has grown enormously. This comprehensive review provides an overview of the impact of perinatal oxidative stress on renal programming, the implications of antioxidant strategies on the prevention of kidney disease, and the gap between animal models and clinical reality.
Collapse
|
62
|
Teofili L, Papacci P, Orlando N, Bianchi M, Pasciuto T, Mozzetta I, Palluzzi F, Giacò L, Giannantonio C, Remaschi G, Santosuosso M, Beccastrini E, Fabbri M, Valentini CG, Bonfini T, Cloclite E, Accorsi P, Dragonetti A, Cresi F, Ansaldi G, Raffaeli G, Villa S, Pucci G, Mondello I, Santodirocco M, Ghirardello S, Vento G. BORN study: a multicenter randomized trial investigating cord blood red blood cell transfusions to reduce the severity of retinopathy of prematurity in extremely low gestational age neonates. Trials 2022; 23:1010. [PMID: 36514106 PMCID: PMC9746198 DOI: 10.1186/s13063-022-06949-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2022] [Accepted: 11/22/2022] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Extremely low gestational age neonates (ELGANs, i.e., neonates born before 28 weeks of gestation) are at high risk of developing retinopathy of prematurity (ROP), with potential long-life visual impairment. Due to concomitant anemia, ELGANs need repeated red blood cell (RBC) transfusions. These produce a progressive replacement of fetal hemoglobin (HbF) by adult hemoglobin (HbA). Furthermore, a close association exists between low levels of HbF and severe ROP, suggesting that a perturbation of the HbF-mediated oxygen release may derange retinal angiogenesis and promote ROP. METHODS/DESIGN BORN (umBilical blOod to tRansfuse preterm Neonates) is a multicenter double-blinded randomized controlled trial in ELGANs, to assess the effect of allogeneic cord blood RBC transfusions (CB-RBCs) on severe ROP development. Recruitment, consent, and randomization take place at 10 neonatology intensive care units (NICUs) of 8 Italian tertiary hospitals. ELGANs with gestational age at birth comprised between 24+0 and 27+6 weeks are randomly allocated into two groups: (1) standard RBC transfusions (adult-RBCs) (control arm) and (2) CB-RBCs (intervention arm). In case of transfusion need, enrolled patients receive transfusions according to the allocation arm, unless an ABO/RhD CB-RBC is unavailable. Nine Italian public CB banks cooperate to make available a suitable amount of CB-RBC units for all participating NICUs. The primary outcome is the incidence of severe ROP (stage 3 or higher) at discharge or 40 weeks of postmenstrual age, which occurs first. DISCUSSION BORN is a groundbreaking trial, pioneering a new transfusion approach dedicated to ELGANs at high risk for severe ROP. In previous non-randomized trials, this transfusion approach was proven feasible and able to prevent the HbF decrease in patients requiring multiple transfusions. Should the BORN trial confirm the efficacy of CB-RBCs in reducing ROP severity, this transfusion strategy would become the preferential blood product to be used in severely preterm neonates. TRIAL REGISTRATION ClinicalTrials.gov NCT05100212. Registered on October 29, 2021.
Collapse
Affiliation(s)
- Luciana Teofili
- grid.414603.4Fondazione Policlinico A. Gemelli IRCCS, Largo Gemelli 8, 00168 Rome, Italy ,grid.8142.f0000 0001 0941 3192Università Cattolica del Sacro Cuore, Rome, Italy
| | - Patrizia Papacci
- grid.414603.4Fondazione Policlinico A. Gemelli IRCCS, Largo Gemelli 8, 00168 Rome, Italy ,grid.8142.f0000 0001 0941 3192Università Cattolica del Sacro Cuore, Rome, Italy
| | - Nicoletta Orlando
- grid.414603.4Fondazione Policlinico A. Gemelli IRCCS, Largo Gemelli 8, 00168 Rome, Italy
| | - Maria Bianchi
- grid.414603.4Fondazione Policlinico A. Gemelli IRCCS, Largo Gemelli 8, 00168 Rome, Italy
| | - Tina Pasciuto
- grid.414603.4Fondazione Policlinico A. Gemelli IRCCS, Largo Gemelli 8, 00168 Rome, Italy
| | - Iolanda Mozzetta
- grid.414603.4Fondazione Policlinico A. Gemelli IRCCS, Largo Gemelli 8, 00168 Rome, Italy
| | - Fernando Palluzzi
- grid.414603.4Fondazione Policlinico A. Gemelli IRCCS, Largo Gemelli 8, 00168 Rome, Italy
| | - Luciano Giacò
- grid.414603.4Fondazione Policlinico A. Gemelli IRCCS, Largo Gemelli 8, 00168 Rome, Italy
| | - Carmen Giannantonio
- grid.414603.4Fondazione Policlinico A. Gemelli IRCCS, Largo Gemelli 8, 00168 Rome, Italy
| | - Giulia Remaschi
- grid.24704.350000 0004 1759 9494Azienda Ospedaliero Universitaria Careggi, Florence, Italy
| | - Michela Santosuosso
- grid.24704.350000 0004 1759 9494Azienda Ospedaliero Universitaria Careggi, Florence, Italy
| | - Enrico Beccastrini
- grid.24704.350000 0004 1759 9494Azienda Ospedaliero Universitaria Careggi, Florence, Italy
| | - Marco Fabbri
- grid.144189.10000 0004 1756 8209Azienda Ospedaliero Universitaria Pisana, Pisa, Italy
| | | | - Tiziana Bonfini
- Azienda Sanitaria Locale-Presidio Ospedaliero di Pescara, Pescara, Italy
| | - Eleonora Cloclite
- Azienda Sanitaria Locale-Presidio Ospedaliero di Pescara, Pescara, Italy
| | - Patrizia Accorsi
- Azienda Sanitaria Locale-Presidio Ospedaliero di Pescara, Pescara, Italy
| | | | - Francesco Cresi
- Città della Salute e della Scienza, Turin, Italy ,grid.7605.40000 0001 2336 6580Department of Public Health and Pediatrics, University of Turin, Turin, Italy
| | | | - Genny Raffaeli
- grid.414818.00000 0004 1757 8749Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy ,grid.4708.b0000 0004 1757 2822Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - Stefania Villa
- grid.414818.00000 0004 1757 8749Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Giulia Pucci
- grid.414504.00000 0000 9051 0784Azienda Ospedaliera Bianchi Melacrino Morelli, Reggio Calabria, Italy
| | - Isabella Mondello
- grid.414504.00000 0000 9051 0784Azienda Ospedaliera Bianchi Melacrino Morelli, Reggio Calabria, Italy
| | - Michele Santodirocco
- grid.413503.00000 0004 1757 9135Ospedale Casa Sollievo della Sofferenza, San Giovanni Rotondo, Foggia, Italy
| | - Stefano Ghirardello
- grid.419425.f0000 0004 1760 3027Fondazione IRCCS Policlinico S. Matteo, Pavia, Italy
| | - Giovanni Vento
- grid.414603.4Fondazione Policlinico A. Gemelli IRCCS, Largo Gemelli 8, 00168 Rome, Italy ,grid.8142.f0000 0001 0941 3192Università Cattolica del Sacro Cuore, Rome, Italy
| |
Collapse
|
63
|
Srivastava A, Srivastava P, Mathur S, Mishra S, Abbas S, Gupta A, Mishra P, Tiwari M, Sharma LK. Analysis of cellular and cell free mitochondrial DNA content and reactive oxygen species levels in maternal blood during normal pregnancy: a pilot study. BMC Pregnancy Childbirth 2022; 22:845. [PMID: 36384496 PMCID: PMC9670558 DOI: 10.1186/s12884-022-05156-2] [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] [Received: 08/11/2022] [Accepted: 10/26/2022] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Alterations in mitochondrial signatures such as mitochondrial DNA (mtDNA) content in maternal blood have been linked to pregnancy-related complications. However, changes in maternal mtDNA content, their distribution and associated signaling during normal pregnancies are not clear; which could suggest their physiological role in maternal adaptation to pregnancy related changes and a reference threshold. THE AIM OF THIS STUDY to assess the distribution of mtDNA in peripheral blood and their association with circulatory ROS levels across different trimesters of healthy pregnancy. METHODS In this pilot cross sectional study, blood samples of normal pregnant women from each trimester (total = 60) and age-matched non-pregnant (NP) women as control group (n = 20) were analyzed for a) the relative distribution of mtDNA content in cellular and cell free (plasma) fractions using relative quantitative polymerase chain reaction (qPCR) and b) the levels of circulating reactive oxygen species (ROS) by measurement of plasma H2O2. The results were compared between pregnant and NP groups and within trimesters for significant differences, and were also analyzed for their correlation between groups using statistical methods. RESULTS While, we observed a significant decline in cellular mtDNA; plasma mtDNA was significant increased across all trimesters compared to NP. However, from comparisons within trimesters; only cellular mtDNA content in 3rd trimester was significantly reduced compared to 1st trimester, and plasma mtDNA did not differ significantly among different trimesters. A significantly higher level of plasma H2O2 was also observed during 3rd trimester compared to NP and to 1st trimester. Correlation analysis showed that, while cellular mtDNA content was negatively correlated to plasma mtDNA and to plasma H2O2 levels; plasma mtDNA was positively correlated with plasma H2O2 content. CONCLUSIONS This study suggested that normal pregnancy is associated with an opposing trend of reduced cellular mtDNA with increased circulatory mtDNA and H2O2 levels, which may contribute to maternal adaptation, required during different stages of pregnancy. Estimation of mtDNA distribution and ROS level in maternal blood could show mitochondrial functionality during normal pregnancy, and could be exploited to identify their prognostic/ diagnostic potential in pregnancy complications.
Collapse
Affiliation(s)
- Anubhav Srivastava
- Department of Molecular Medicine & Biotechnology, Sanjay Gandhi Post Graduate Institute of Medical Sciences, 4th Floor, PMSSY Building, Raebareli Road, Lucknow, Uttar Pradesh, 226014, India
| | - Pransu Srivastava
- Department of Molecular Medicine & Biotechnology, Sanjay Gandhi Post Graduate Institute of Medical Sciences, 4th Floor, PMSSY Building, Raebareli Road, Lucknow, Uttar Pradesh, 226014, India
| | - Shashank Mathur
- Department of Molecular Medicine & Biotechnology, Sanjay Gandhi Post Graduate Institute of Medical Sciences, 4th Floor, PMSSY Building, Raebareli Road, Lucknow, Uttar Pradesh, 226014, India
| | - Suman Mishra
- Department of Molecular Medicine & Biotechnology, Sanjay Gandhi Post Graduate Institute of Medical Sciences, 4th Floor, PMSSY Building, Raebareli Road, Lucknow, Uttar Pradesh, 226014, India
| | - Sabiya Abbas
- Department of Molecular Medicine & Biotechnology, Sanjay Gandhi Post Graduate Institute of Medical Sciences, 4th Floor, PMSSY Building, Raebareli Road, Lucknow, Uttar Pradesh, 226014, India
| | - Amrit Gupta
- Department of Maternal & Reproductive Health, Sanjay Gandhi Post Graduate Institute of Medical Sciences, Lucknow, 226014, India
| | - Prabhaker Mishra
- Department of Biostatistics and Health Informatics, Sanjay Gandhi Post Graduate Institute of Medical Sciences, Lucknow, 226014, India
| | - Meenakshi Tiwari
- Department of Pathology and Laboratory Medicine, All India Institute of Medical Sciences, Patna, 801507, India
| | - Lokendra Kumar Sharma
- Department of Molecular Medicine & Biotechnology, Sanjay Gandhi Post Graduate Institute of Medical Sciences, 4th Floor, PMSSY Building, Raebareli Road, Lucknow, Uttar Pradesh, 226014, India.
| |
Collapse
|
64
|
Karlsson V, Sporre B, Fredén F, Ågren J. Randomized controlled trial of low vs high oxygen during neonatal anesthesia: Oxygenation, feasibility, and oxidative stress. Paediatr Anaesth 2022; 32:1062-1069. [PMID: 35791748 PMCID: PMC9546133 DOI: 10.1111/pan.14519] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 06/07/2022] [Accepted: 06/09/2022] [Indexed: 11/27/2022]
Abstract
BACKGROUND To reduce risk for intermittent hypoxia a high fraction of inspired oxygen is routinely used during anesthesia induction. This differs from the cautious dosing of oxygen during neonatal resuscitation and intensive care and may result in significant hyperoxia. AIM In a randomized controlled trial, we evaluated oxygenation during general anesthesia with a low (23%) vs a high (80% during induction and recovery, and 40% during maintenance) fraction of inspired oxygen, in newborn infants undergoing surgery. METHOD Thirty-five newborn infants with postconceptional age of 35-44 weeks were included (17 infants in low and 18 in high oxygen group). Oxygenation was monitored by transcutaneous partial pressure of oxygen, pulse oximetry, and cerebral oxygenation. Predefined SpO2 safety targets dictated when to increase inspired oxygen. RESULTS At start of anesthesia, oxygenation was similar in both groups. Throughout anesthesia, the high oxygen group displayed significant hyperoxia with higher (difference-20.3 kPa, 95% confidence interval (CI)-28.4 to 12.2, p < .001) transcutaneous partial pressure of oxygen values than the low oxygen group. While SpO2 in the low oxygen group was lower (difference - 5.8%, 95% CI -9.3 to -2.4, p < .001) during anesthesia, none of the infants spent enough time below SpO2 safety targets to mandate supplemental oxygen, and cerebral oxygenation was within the normal range and not statistically different between the groups. Analysis of the oxidative stress biomarker urinary F2 -Isoprostane revealed no differences between the low and high oxygen group. CONCLUSION We conclude that in healthy newborn infants, use of low oxygen during general anesthesia was feasible, while the prevailing practice of using high levels of inspired oxygen resulted in significant hyperoxia. The trade-off between careful dosing of oxygen and risks of hypo- and hyperoxia in neonatal anesthesia should be further examined.
Collapse
Affiliation(s)
- Victoria Karlsson
- Department of Women's and Children's HealthUppsala UniversityUppsalaSweden,Neonatology DivisionUniversity Children's HospitalUppsalaSweden
| | - Bengt Sporre
- Anaesthesiology and Intensive Care DivisionUppsala University HospitalUppsalaSweden
| | - Filip Fredén
- Department of Anaesthesiology and Intensive CareUppsala UniversityUppsalaSweden
| | - Johan Ågren
- Department of Women's and Children's HealthUppsala UniversityUppsalaSweden,Neonatology DivisionUniversity Children's HospitalUppsalaSweden
| |
Collapse
|
65
|
Moon K, Mckinnon E, Croft K, Hendrie D, Patole S, Simmer K, Rao S. Early versus late parenteral nutrition in term and late preterm infants: study protocol for a randomised controlled trial. BMC Pediatr 2022; 22:514. [PMID: 36042439 PMCID: PMC9429301 DOI: 10.1186/s12887-022-03569-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Accepted: 08/21/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Despite the wide use of parenteral nutrition (PN) in neonatal intensive care units (NICU), there is limited evidence regarding the optimal time to commence PN in term and late preterm infants. The recommendations from the recently published ESPGHAN/ESPEN/ESPR/CPEN and NICE guidelines are substantially different in this area, and surveys have reported variations in clinical practice. The aim of this randomised controlled trial (RCT) is to evaluate the benefits and risks of early versus late PN in term and late preterm infants. METHODS/DESIGN This study is a single-centre, non-blinded RCT in the NICU of Perth Children's Hospital, Western Australia.A total of 60 infants born ≥34 weeks of gestation who have a high likelihood of intolerance to enteral nutrition (EN) for at least 3-5 days will be randomised to early (day 1 or day 2 of admission) or late commencement (day 6 of admission) of PN after informed parental consent. In both groups, EN will be commenced as early as clinically feasible. Primary outcomes are plasma phenylalanine and plasma F2-isoprostane levels on Day 4 and Day 8 of admission. Secondary outcomes are total and individual plasma amino acid profiles, plasma and red blood cell fatty acid profiles, in-hospital all-cause mortality, hospital-acquired infections, length of hospital/NICU stay, z scores and changes in z scores at discharge for weight, height and head circumference, time to full EN, duration of respiratory (mechanical, non-invasive) support, duration of inotropic support, the incidence of hyper and hypoglycaemia, incidence of metabolic acidosis, liver function, blood urea nitrogen, and C-reactive protein (CRP). DISCUSSION This RCT will examine the effects of early versus late PN in term and late preterm infants by comparing key biochemical and clinical outcomes and has the potential to identify underlying pathways for beneficial or harmful effects related to the timing of commencement of PN in such infants. TRIAL REGISTRATION ANZCTR; ACTRN12620000324910 (3rd March 2020).
Collapse
Affiliation(s)
- Kwi Moon
- Pharmacy Department, Perth Children's Hospital, 15 Hospital Ave, Nedlands, WA, 6009, Australia.
- Medical School, The University of Western Australia, Perth, WA, Australia.
| | | | - Kevin Croft
- School of Biomedical Sciences, The University of Western Australia, Perth, WA, Australia
| | - Delia Hendrie
- School of Public Health Sciences, Curtin University, Perth, WA, Australia
| | - Sanjay Patole
- Medical School, The University of Western Australia, Perth, WA, Australia
- Department of Neonatology, King Edward Memorial Hospital, Subiaco, WA, Australia
| | - Karen Simmer
- Medical School, The University of Western Australia, Perth, WA, Australia
- Telethon Kids Institute, Nedlands, WA, Australia
| | - Shripada Rao
- Medical School, The University of Western Australia, Perth, WA, Australia
- Department of Neonatology, Perth Children's Hospital, Nedlands, WA, Australia
| |
Collapse
|
66
|
Machado RS, Tenfen L, Joaquim L, Lanzzarin EVR, Bernardes GC, Bonfante SR, Mathias K, Biehl E, Bagio É, Stork SDS, Denicol T, de Oliveira MP, da Silva MR, Danielski LG, de Quadros RW, Rezin GT, Terra SR, Balsini JN, Gava FF, Petronilho F. Hyperoxia by short-term promotes oxidative damage and mitochondrial dysfunction in rat brain. Respir Physiol Neurobiol 2022; 306:103963. [PMID: 36041716 DOI: 10.1016/j.resp.2022.103963] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 07/29/2022] [Accepted: 08/21/2022] [Indexed: 11/17/2022]
Abstract
Oxygen (O2) therapy is used as a therapeutic protocol to prevent or treat hypoxia. However, a high inspired fraction of O2 (FIO2) promotes hyperoxia, a harmful condition for the central nervous system (CNS). The present study evaluated parameters of oxidative stress and mitochondrial dysfunction in the brain of rats exposed to different FIO2. Male Wistar rats were exposed to hyperoxia (FIO2 40 % and 60 %) compared to the control group (FIO2 21 %) for 2 h. Oxidative stress, neutrophilic infiltration, and mitochondrial respiratory chain enzymes were determined in the hippocampus, striatum, cerebellum, cortex, and prefrontal cortex after O2 exposure. The animals exposed to hyperoxia showed increased lipid peroxidation, formation of carbonyl proteins, N/N concentration, and neutrophilic infiltration in some brain regions, like hippocampus, striatum, and cerebellum being the most affected. Furthermore, CAT activity and activity of mitochondrial enzyme complexes were also altered after exposure to hyperoxia. Rats exposed to hyperoxia showed increase in oxidative stress parameters and mitochondrial dysfunction in brain structures.
Collapse
Affiliation(s)
- Richard Simon Machado
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Graduate Program in Health Sciences, Health Sciences Unit, University of South Santa Catarina, Tubarão, SC, Brazil
| | - Leonardo Tenfen
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Graduate Program in Health Sciences, Health Sciences Unit, University of South Santa Catarina, Tubarão, SC, Brazil
| | - Larissa Joaquim
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Graduate Program in Health Sciences, Health Sciences Unit, University of South Santa Catarina, Tubarão, SC, Brazil
| | - Everton Venicius Rosa Lanzzarin
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Graduate Program in Health Sciences, Health Sciences Unit, University of South Santa Catarina, Tubarão, SC, Brazil
| | - Gabriela Costa Bernardes
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Graduate Program in Health Sciences, Health Sciences Unit, University of South Santa Catarina, Tubarão, SC, Brazil
| | - Sandra Regina Bonfante
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Graduate Program in Health Sciences, Health Sciences Unit, University of South Santa Catarina, Tubarão, SC, Brazil
| | - Khiany Mathias
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Graduate Program in Health Sciences, Health Sciences Unit, University of South Santa Catarina, Tubarão, SC, Brazil
| | - Erica Biehl
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Graduate Program in Health Sciences, Health Sciences Unit, University of South Santa Catarina, Tubarão, SC, Brazil
| | - Érick Bagio
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Graduate Program in Health Sciences, Health Sciences Unit, University of South Santa Catarina, Tubarão, SC, Brazil
| | - Solange de Souza Stork
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Graduate Program in Health Sciences, Health Sciences Unit, University of South Santa Catarina, Tubarão, SC, Brazil
| | - Tais Denicol
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Graduate Program in Health Sciences, Health Sciences Unit, University of South Santa Catarina, Tubarão, SC, Brazil
| | - Mariana Pacheco de Oliveira
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Graduate Program in Health Sciences, Health Sciences Unit, University of South Santa Catarina, Tubarão, SC, Brazil
| | - Mariella Reinol da Silva
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Graduate Program in Health Sciences, Health Sciences Unit, University of South Santa Catarina, Tubarão, SC, Brazil
| | - Lucinéia Gainski Danielski
- Laboratory of Experimental Neurology, Graduate Program in Health Sciences, University of Southern Santa Catarina, Criciuma, SC, Brazil
| | | | - Gislaine Tezza Rezin
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Graduate Program in Health Sciences, Health Sciences Unit, University of South Santa Catarina, Tubarão, SC, Brazil
| | - Silvia Resende Terra
- Department of Biochemistry, Federal University of Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Jairo Nunes Balsini
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Graduate Program in Health Sciences, Health Sciences Unit, University of South Santa Catarina, Tubarão, SC, Brazil
| | - Fernanda Frederico Gava
- Laboratory of Experimental Neurology, Graduate Program in Health Sciences, University of Southern Santa Catarina, Criciuma, SC, Brazil
| | - Fabricia Petronilho
- Laboratory of Experimental Neurology, Graduate Program in Health Sciences, University of Southern Santa Catarina, Criciuma, SC, Brazil.
| |
Collapse
|
67
|
Bellach L, Eigenschink M, Hassanein A, Savran D, Salzer U, Müllner EW, Repa A, Klebermass-Schrehof K, Wisgrill L, Giordano V, Berger A. Packed red blood cell transfusion in preterm infants. Lancet Haematol 2022; 9:e615-e626. [PMID: 35901846 DOI: 10.1016/s2352-3026(22)00207-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 06/03/2022] [Accepted: 06/09/2022] [Indexed: 06/15/2023]
Abstract
Premature infants commonly receive adult packed red blood cells (pRBCs) during their hospital stay. As adult erythrocytes differ substantially from those of preterm infants, transfusion of adult pRBCs into preterm infants can be considered inappropriate for the physiology of a preterm infant. An absence of standardisation of transfusion protocols makes it difficult to compare and interpret pertinent clinical data, as reflected by unclear associations between pRBC transfusion and complications related to prematurity, such as bronchopulmonary dysplasia, neurodevelopmental impairment, retinopathy of prematurity, or necrotising enterocolitis. The difficulty in interpreting clinical data is further increased by differences in study designs that either overestimate pRBC-associated complications of prematurity or have not yet been designed to directly link pRBC transfusions to their respective complications. Thus, neonatal transfusion practice has become an ongoing difficulty, in which differences in transfusion guidelines hinder the ability to generate comparable clinical data, and heterogeneity in clinical data prevents the implementation of standardised transfusion protocols. To overcome these issues, novel approaches with biochemical-clinical translational designs could enable clinicians to gather causal evidence instead of circumstantial correlation.
Collapse
Affiliation(s)
- Luise Bellach
- Department of Pediatrics and Adolescent Medicine, Comprehensive Center for Pediatrics, Medical University of Vienna, Vienna, Austria
| | - Michael Eigenschink
- Center for Medical Biochemistry, Max Perutz Labs, Medical University of Vienna, Vienna, Austria
| | - Abtin Hassanein
- Department of Pediatrics and Adolescent Medicine, Comprehensive Center for Pediatrics, Medical University of Vienna, Vienna, Austria
| | - Danylo Savran
- Center for Medical Biochemistry, Max Perutz Labs, Medical University of Vienna, Vienna, Austria
| | - Ulrich Salzer
- Center for Medical Biochemistry, Max Perutz Labs, Medical University of Vienna, Vienna, Austria
| | - Ernst W Müllner
- Center for Medical Biochemistry, Max Perutz Labs, Medical University of Vienna, Vienna, Austria
| | - Andreas Repa
- Department of Pediatrics and Adolescent Medicine, Comprehensive Center for Pediatrics, Medical University of Vienna, Vienna, Austria
| | - Katrin Klebermass-Schrehof
- Department of Pediatrics and Adolescent Medicine, Comprehensive Center for Pediatrics, Medical University of Vienna, Vienna, Austria
| | - Lukas Wisgrill
- Department of Pediatrics and Adolescent Medicine, Comprehensive Center for Pediatrics, Medical University of Vienna, Vienna, Austria
| | - Vito Giordano
- Department of Pediatrics and Adolescent Medicine, Comprehensive Center for Pediatrics, Medical University of Vienna, Vienna, Austria
| | - Angelika Berger
- Department of Pediatrics and Adolescent Medicine, Comprehensive Center for Pediatrics, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
68
|
Özer Bekmez B, Tayman C, Çakır U, Koyuncu İ, Büyüktiryaki M, Türkmenoğlu TT, Çakır E. Glucocorticoids in a Neonatal Hyperoxic Lung Injury Model: Pulmonary and Neurotoxic effects. Pediatr Res 2022; 92:436-444. [PMID: 34725500 DOI: 10.1038/s41390-021-01777-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2019] [Revised: 06/18/2021] [Accepted: 09/12/2021] [Indexed: 11/08/2022]
Abstract
BACKGROUND We aimed to compare the effect of dexamethasone (Dex), hydrocortisone (Hc), and methylprednisolone (Mpz) at equivalent doses on somatic growth, lung healing, and neurotoxicity in a hyperoxic rat model. We hypothesized that Mpz and Hc would be superior to Dex with less neurotoxicity by exerting similar therapeutic efficacy on the injured lung. METHODS Neonatal rats were randomized to control, bronchopulmonary dysplasia (BPD), Dex, Hc, and Mpz groups. All drugs were administered daily following day 15 over 7 days. Histopathological and immunohistochemical analyses of the lung and brain were performed on day 22. RESULTS All types had much the same impact on lung repair. Oxidative markers in the lung were similar in the steroid groups. While nuclear factor erythroid 2-related factor and heat-shock protein 70 dropped following steroid treatment, no difference was noted in other biochemical markers in the brain between the study groups. Apoptotic activity and neuron loss in the parietal cortex and hippocampus were noted utmost in Dex, but alike in other BPD groups. CONCLUSIONS Mpz does not appear to be superior to Dex or Hc in terms of pulmonary outcomes and oxidative damage in the brain, but safer than Dex regarding apoptotic neuron loss. IMPACT This is the first study that compared the pulmonary efficacy and neurotoxic effects of Dex, Hc, and Mpz simultaneously in an established BPD model. This study adds to the literature on the importance of possible antioxidant and protective effects of glucocorticoid therapy in an oxidative stress-exposed brain. Mpz ended up with no more additional neuron loss or apoptosis risk by having interchangeable effects with others for the treatment of established BPD. Mpz and Hc seem safe as a rescue therapy in terms of adverse outcomes for established BPD in which lung and brain tissue is already impaired.
Collapse
Affiliation(s)
- Buse Özer Bekmez
- Division of Neonatology, Sariyer Hamidiye Etfal Education and Research Hospital, Istanbul, Turkey.
| | - Cüneyt Tayman
- Division of Neonatology, Ankara City Hospital, Ankara, Turkey
| | - Ufuk Çakır
- Division of Neonatology, Ankara City Hospital, Ankara, Turkey
| | - İsmail Koyuncu
- Division of Biochemistry, Faculty of Medicine, Harran University, Şanlıurfa, Turkey
| | - Mehmet Büyüktiryaki
- Division of Neonatology, Faculty of Medicine, Medipol Univerisity, Istanbul, Turkey
| | | | - Esra Çakır
- Division of Anesthesia and Reanimation, Numune Education and Research Hospital, Ankara, Turkey
| |
Collapse
|
69
|
Rawat M, Lakshminrusimha S, Vento M. Pulmonary hypertension and oxidative stress: Where is the link? Semin Fetal Neonatal Med 2022; 27:101347. [PMID: 35473693 PMCID: PMC11151383 DOI: 10.1016/j.siny.2022.101347] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Oxidative stress can be associated with hyperoxia and hypoxia and is characterized by an increase in reactive oxygen (ROS) and nitrogen (RNS) species generated by an underlying disease process or by supplemental oxygen that exceeds the neutralization capacity of the organ system. ROS and RNS acting as free radicals can inactive several enzymes and vasodilators in the nitric oxide pathway promoting pulmonary vasoconstriction resulting in persistent pulmonary hypertension of the newborn (PPHN). Studies in animal models of PPHN have shown high ROS/RNS that is further increased by hyperoxic ventilation. In addition, antioxidant therapy increased PaO2 in these models, but clinical trials are lacking. We recommend targeting preductal SpO2 between 90 and 97%, PaO2 between 55 and 80 mmHg and avoiding FiO2 > 0.6-0.8 if possible during PPHN management. This review highlights the role of oxidative and nitrosative stress markers on PPHN and potential therapeutic interventions that may alleviate the consequences of increased oxidant stress during ventilation with supplemental oxygen.
Collapse
Affiliation(s)
- Munmun Rawat
- Department of Pediatrics, University at Buffalo, Buffalo, NY, USA
| | | | - Maximo Vento
- Division of Neonatology, University & Polytechnic Hospital La Fe and Instituto de Investigación Sanitaria La Fe (IISLAFE), Valencia, Spain.
| |
Collapse
|
70
|
Ekhaguere OA, Okonkwo IR, Batra M, Hedstrom AB. Respiratory distress syndrome management in resource limited settings-Current evidence and opportunities in 2022. Front Pediatr 2022; 10:961509. [PMID: 35967574 PMCID: PMC9372546 DOI: 10.3389/fped.2022.961509] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2022] [Accepted: 06/30/2022] [Indexed: 01/19/2023] Open
Abstract
The complications of prematurity are the leading cause of neonatal mortality worldwide, with the highest burden in the low- and middle-income countries of South Asia and Sub-Saharan Africa. A major driver of this prematurity-related neonatal mortality is respiratory distress syndrome due to immature lungs and surfactant deficiency. The World Health Organization's Every Newborn Action Plan target is for 80% of districts to have resources available to care for small and sick newborns, including premature infants with respiratory distress syndrome. Evidence-based interventions for respiratory distress syndrome management exist for the peripartum, delivery and neonatal intensive care period- however, cost, resources, and infrastructure limit their availability in low- and middle-income countries. Existing research and implementation gaps include the safe use of antenatal corticosteroid in non-tertiary settings, establishing emergency transportation services from low to high level care facilities, optimized delivery room resuscitation, provision of affordable caffeine and surfactant as well as implementing non-traditional methods of surfactant administration. There is also a need to optimize affordable continuous positive airway pressure devices able to blend oxygen, provide humidity and deliver reliable pressure. If the high prematurity-related neonatal mortality experienced in low- and middle-income countries is to be mitigated, a concerted effort by researchers, implementers and policy developers is required to address these key modalities.
Collapse
Affiliation(s)
- Osayame A. Ekhaguere
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Ikechukwu R. Okonkwo
- Department of Pediatrics, University of Benin Teaching Hospital, Benin City, Nigeria
| | - Maneesh Batra
- Departments of Pediatrics and Global Health, University of Washington, Seattle, WA, United States
| | - Anna B. Hedstrom
- Departments of Pediatrics and Global Health, University of Washington, Seattle, WA, United States
| |
Collapse
|
71
|
Oxidative Stress and Antioxidative Therapy in Pulmonary Arterial Hypertension. Molecules 2022; 27:molecules27123724. [PMID: 35744848 PMCID: PMC9229274 DOI: 10.3390/molecules27123724] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 06/06/2022] [Accepted: 06/07/2022] [Indexed: 02/04/2023] Open
Abstract
Pulmonary arterial hypertension (PAH) is clinically characterized by a progressive increase in pulmonary artery pressure, followed by right ventricular hypertrophy and subsequently right heart failure. The underlying mechanism of PAH includes endothelial dysfunction and intimal smooth muscle proliferation. Numerous studies have shown that oxidative stress is critical in the pathophysiology of PAH and involves changes in reactive oxygen species (ROS), reactive nitrogen (RNS), and nitric oxide (NO) signaling pathways. Disrupted ROS and NO signaling pathways cause the proliferation of pulmonary arterial endothelial cells (PAECs) and pulmonary vascular smooth muscle cells (PASMCs), resulting in DNA damage, metabolic abnormalities, and vascular remodeling. Antioxidant treatment has become a main area of research for the treatment of PAH. This review mainly introduces oxidative stress in the pathogenesis of PAH and antioxidative therapies and explains why targeting oxidative stress is a valid strategy for PAH treatment.
Collapse
|
72
|
Polyphenols and IUGR Pregnancies: Effects of the Antioxidant Hydroxytyrosol on the Hippocampus Proteome in a Porcine Model. Antioxidants (Basel) 2022; 11:antiox11061135. [PMID: 35740029 PMCID: PMC9219860 DOI: 10.3390/antiox11061135] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 06/04/2022] [Accepted: 06/07/2022] [Indexed: 12/13/2022] Open
Abstract
Supplementation of a mother’s diet with antioxidants such as hydroxytyrosol (HTX) has been proposed to ameliorate the adverse phenotypes of foetuses affected by intrauterine growth restriction (IUGR). Our previous studies showed, in a porcine model of IUGR, an effect of maternal HTX supplementation on the neurotransmitter profile of several brain areas and the morphology of the hippocampus in 100 days old foetuses. The present study analyzed the impact of maternal HTX supplementation on the hippocampus proteome at this foetal age by TMT10plex labelling. Eleven differentially abundant proteins were identified by comparing both conditions, and eight of them downregulated and three upregulated in the HTX-treated group. The downregulated proteins were mainly involved in protein synthesis and RNA metabolism and may explain the differences in neuron differentiation in the HTX-treated group. The upregulated proteins were related to cell detoxification and could represent a potential mechanism to explain the neuroprotective effect of HTX.
Collapse
|
73
|
Kumova OK, Galani IE, Rao A, Johnson H, Triantafyllia V, Matt SM, Pascasio J, Gaskill PJ, Andreakos E, Katsikis PD, Carey AJ. Severity of neonatal influenza infection is driven by type I interferon and oxidative stress. Mucosal Immunol 2022; 15:1309-1320. [PMID: 36352099 PMCID: PMC9724789 DOI: 10.1038/s41385-022-00576-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 09/26/2022] [Accepted: 10/23/2022] [Indexed: 11/11/2022]
Abstract
Neonates exhibit increased susceptibility to respiratory viral infections, attributed to inflammation at the developing pulmonary air-blood interface. IFN I are antiviral cytokines critical to control viral replication, but also promote inflammation. Previously, we established a neonatal murine influenza virus (IV) model, which demonstrates increased mortality. Here, we sought to determine the role of IFN I in this increased mortality. We found that three-day-old IFNAR-deficient mice are highly protected from IV-induced mortality. In addition, exposure to IFNβ 24 h post IV infection accelerated death in WT neonatal animals but did not impact adult mortality. In contrast, IFN IIIs are protective to neonatal mice. IFNβ induced an oxidative stress imbalance specifically in primary neonatal IV-infected pulmonary type II epithelial cells (TIIEC), not in adult TIIECs. Moreover, neonates did not have an infection-induced increase in antioxidants, including a key antioxidant, superoxide dismutase 3, as compared to adults. Importantly, antioxidant treatment rescued IV-infected neonatal mice, but had no impact on adult morbidity. We propose that IFN I exacerbate an oxidative stress imbalance in the neonate because of IFN I-induced pulmonary TIIEC ROS production coupled with developmentally regulated, defective antioxidant production in response to IV infection. This age-specific imbalance contributes to mortality after respiratory infections in this vulnerable population.
Collapse
Affiliation(s)
- Ogan K. Kumova
- Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA, United States
| | - Ioanna-Evdokia Galani
- Laboratory of Immunobiology, Center for Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation Academy of Athens, Athens, Greece
| | - Abhishek Rao
- Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA, United States
| | - Hannah Johnson
- Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA, United States
| | - Vasiliki Triantafyllia
- Laboratory of Immunobiology, Center for Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation Academy of Athens, Athens, Greece
| | - Stephanie M. Matt
- Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA, United States
| | - Judy Pascasio
- Pathology, Drexel University College of Medicine, Philadelphia, PA, United States
| | - Peter J. Gaskill
- Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA, United States
| | - Evangelos Andreakos
- Laboratory of Immunobiology, Center for Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation Academy of Athens, Athens, Greece
| | - Peter D. Katsikis
- Immunology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Alison J. Carey
- Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA, United States.,Pediatrics, Drexel University College of Medicine, Philadelphia, PA, United States
| |
Collapse
|
74
|
Ferrante G, Montante C, Notarbartolo V, Giuffrè M. Antioxidants: Role the in prevention and treatment of bronchopulmonary dysplasia. Paediatr Respir Rev 2022; 42:53-58. [PMID: 35177319 DOI: 10.1016/j.prrv.2022.01.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 01/01/2022] [Accepted: 01/06/2022] [Indexed: 10/19/2022]
Abstract
Bronchopulmonary dysplasia (BPD) is one of the major causes of chronic respiratory diseases among infants. Both pharmacological and nonpharmacological approaches have been proposed for its management. Since oxidative stress is known to play a pivotal role in the pathogenesis of BPD, it is reasonable to consider the potential of antioxidant strategies in the prevention and treatment of this condition. Indeed, antioxidants can prevent or inhibit substrate oxidation. Some studies have evaluated the efficacy of the exogenous administration of vitamins and micronutrients in reducing the propagation of free radicals through their scavenging capacity. Nonetheless, encouraging preclinical results did not translate into effective preventive and/or therapeutic interventions. This narrative review evaluates the current evidence about the antioxidants that are potentially useful for preventing and treating BPD and explores the most relevant issues affecting their implementation in clinical practice, as well as their associated evidence gaps and research limitations.
Collapse
Affiliation(s)
- Giuliana Ferrante
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, University of Palermo, Palermo, Italy.
| | - Claudio Montante
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, University of Palermo, Palermo, Italy
| | - Veronica Notarbartolo
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, University of Palermo, Palermo, Italy.
| | - Mario Giuffrè
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, University of Palermo, Palermo, Italy
| |
Collapse
|
75
|
In Vivo Assessment of Antioxidant Potential of Human Milk Treated by Holder Pasteurization or High Hydrostatic Pressure Processing: A Preliminary Study on Intestinal and Hepatic Markers in Adult Mice. Antioxidants (Basel) 2022; 11:antiox11061091. [PMID: 35739988 PMCID: PMC9220199 DOI: 10.3390/antiox11061091] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 05/20/2022] [Accepted: 05/27/2022] [Indexed: 12/10/2022] Open
Abstract
Preterm infants are highly susceptible to oxidative stress due to an imbalance between endogenous oxidant and antioxidant systems. In addition, these newborns are frequently fed with donor milk (DM) treated by Holder pasteurization (HoP) at 62.5 °C for 30 min, which is known to alter numerous heat-sensitive factors, including some antioxidants. High hydrostatic pressure (HHP) processing was recently proposed as an innovative method for the treatment of DM. The present study aimed to measure the redox balance of HoP- and HHP-DM and to study, in vivo, the effects of HoP- and HHP-DM on the gut and liver. H2O2, vitamin A and vitamin E (α- and γ-tocopherols) concentrations, as well as the total antioxidant capacity (TAC), were measured in raw-, HoP- and HHP-DM. The gene expression level of antioxidant systems and inflammatory response were quantified in the ileum and liver of adult mice after 7 days of oral administration of HoP- or HHP-DM. HoP reduced the γ-tocopherol level, whereas HHP treatment preserved all vitamins close to the raw milk level. The milk H2O2 content was reduced by HHP but not by HoP. The total antioxidant capacity of DM was reduced after HHP processing measured by PAOT-Liquid® technology but was unaffected after measurement by ORAC assay. In mice, HHP-DM administration induced a stimulation of antioxidant defenses and reduced some inflammatory markers in both the ileum and liver compared to HoP-DM treatment. Our preliminary study suggests that the HHP processing of DM may better protect preterm infants from gut and liver pathologies compared to HoP, which is currently used in most human milk banks.
Collapse
|
76
|
Damiano S, Longobardi C, Salzano A, D’Angelo L, Amenta M, Maggiolino A, De Palo P, Claps S, Rufrano D, Iannaccone F, Matera R, Ciarcia R. Red orange and lemon extract preserve from oxidative stress, DNA damage and inflammatory status in lambs. ITALIAN JOURNAL OF ANIMAL SCIENCE 2022. [DOI: 10.1080/1828051x.2022.2056527] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Sara Damiano
- Dipartimento di Medicina Veterinaria e Produzioni Animali, Università degli Studi di Napoli Federico II, Napoli, Italia
| | - Consiglia Longobardi
- Dipartimento di Salute Mentale, Fisica e Medicina Preventiva, Università della Campania "Luigi Vanvitelli", Napoli Italia
| | - Angela Salzano
- Dipartimento di Medicina Veterinaria e Produzioni Animali, Università degli Studi di Napoli Federico II, Napoli, Italia
| | - Livia D’Angelo
- Dipartimento di Medicina Veterinaria e Produzioni Animali, Università degli Studi di Napoli Federico II, Napoli, Italia
| | - Margherita Amenta
- Consiglio per la Ricerca e l’Economia Agraria (CREA)— Centro di Ricerca per l’Olivo, Frutticultura e Agrumi, Acireale, Italia
| | - Aristide Maggiolino
- Dipartimento di Medicina Veterinaria, Università degli Studi di Bari“Aldo Moro”, Valenzano (BA), Italy
| | - Pasquale De Palo
- Dipartimento di Medicina Veterinaria, Università degli Studi di Bari“Aldo Moro”, Valenzano (BA), Italy
| | - Salvatore Claps
- Consiglio per la Ricerca e l’Economia Agraria (CREA) – Centro di Ricerca per le Produzioni Animali e l’Acquacoltura, Bella Muro (PZ), Italia
| | - Domenico Rufrano
- Consiglio per la Ricerca e l’Economia Agraria (CREA) – Centro di Ricerca per le Produzioni Animali e l’Acquacoltura, Bella Muro (PZ), Italia
| | - Francesco Iannaccone
- Dipartimento di Scienze Agro-Ambientali e Territoriali (DISAAT), Università degli Studi di Bari “Aldo Moro”, Bari, Italia
| | - Roberta Matera
- Dipartimento di Medicina Veterinaria e Produzioni Animali, Università degli Studi di Napoli Federico II, Napoli, Italia
| | - Roberto Ciarcia
- Dipartimento di Medicina Veterinaria e Produzioni Animali, Università degli Studi di Napoli Federico II, Napoli, Italia
| |
Collapse
|
77
|
Jhaveri Sanghvi U, Wright CJ, Hernandez TL. Pulmonary Resilience: Moderating the Association between Oxygen Exposure and Pulmonary Outcomes in Extremely Preterm Newborns. Neonatology 2022; 119:433-442. [PMID: 35551136 PMCID: PMC9296587 DOI: 10.1159/000524438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2021] [Accepted: 03/29/2022] [Indexed: 11/19/2022]
Abstract
Bronchopulmonary dysplasia (BPD) is a chronic lung disease of infancy associated with high morbidity and mortality. Although most prevalent following extremely preterm birth, BPD is diagnosed at 36 weeks post-menstrual age, when the disease trajectory is underway, and long-term physiological implications may be irreversible. There is an urgent and unmet need to identify how early exposures can be modified to decrease the risk of developing BPD before disease progression becomes irreversible. Extremely preterm newborns encounter a paradox at birth: oxygen is a life-sustaining component of ex utero life yet is undeniably toxic. Attempts at minimizing supplemental oxygen exposure by targeting lower oxygen saturations appear to decrease BPD but may increase mortality. Given the potential association between lower oxygen saturations and increased mortality, practice guidelines favor targeting higher saturations. This uniformly increases oxygen exposure, prompting a cascade of pathogenic mechanisms implicated in BPD development. In this review, we introduce the concept of pulmonary resilience: a homeostatic process driven by the autonomic nervous system (ANS) as a moderator of physiologic stress that when functional, could inform successful environmental adaptation following extremely preterm birth. We hypothesize that infants with early-life ANS dysfunction require a higher oxygen dose for survival; conversely, oxygen exposure could be safely limited in infants with more robust early-life ANS function, an indicator of pulmonary resilience. Characterizing the pulmonary resilience continuum to guide individualized supplemental oxygen dosing may reduce morbidity and mortality in this growing population of extremely preterm infants at risk for BPD.
Collapse
Affiliation(s)
- Urvi Jhaveri Sanghvi
- College of Nursing; Anschutz Medical Campus, University of
Colorado, Aurora, CO, USA
| | - Clyde J. Wright
- Department of Pediatrics, Section of Neonatology; Anschutz
Medical Campus; University of Colorado, Aurora, CO, USA
| | - Teri L. Hernandez
- College of Nursing; Anschutz Medical Campus, University of
Colorado, Aurora, CO, USA
- Department of Medicine, Division of Endocrinology,
Metabolism, and Diabetes; Anschutz Medical Campus, University of Colorado, Aurora,
CO, USA
- Department of Research, Innovation, and Clinical Practice;
Children’s Hospital Colorado, Aurora, CO, USA
| |
Collapse
|
78
|
Castro TDF, de Matos NA, de Souza ABF, Costa GDP, Perucci LO, Talvani A, Cangussú SD, Chianca-Jr DA, de Menezes RCA, Bezerra FS. Protein restriction during pregnancy affects lung development and promotes oxidative stress and inflammation in C57BL/6 mice offspring. Nutrition 2022; 101:111682. [DOI: 10.1016/j.nut.2022.111682] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 03/22/2022] [Accepted: 04/04/2022] [Indexed: 11/16/2022]
|
79
|
Abstract
In evaluating vitamin E (VE) nutritional status of preterm infants, it is essential that any data should be compared with those of healthy term infants, and never with those of adults. Moreover, it should be evaluated in terms of gestational age (GA), not birth weight (BW), because placental transfer of most nutrients from mother to fetus is dependent on GA, not BW. Judging from the limited data during the last 75 years, there was no significant correlation between GA and VE concentrations in circulation or in the red blood cells (RBCs), leukocytes, and buccal mucosal cells. In addition, the oxidizability of polyunsaturated fatty acids (PUFAs) in plasma or RBCs, as targets for protection by VE chain-breaking ability, was lower in preterm infants. However, because of the minimal information available about hepatic VE levels, which is considered a key determinant of whole body VE status, the decision on whether VE status of preterm infants is comparable with that of term infants should be postponed. Clinical trials of VE supplementation in preterm infants were repeatedly undertaken to investigate whether VE reduces severity or inhibits development of several diseases specific to preterm infants, namely retinopathy of prematurity (ROP), bronchopulmonary dysplasia (BPD), and germinal matrix hemorrhage - intraventricular hemorrhage (GMH-IVH). Most of these trials resulted in a misfire, with a few exceptions for IVH prevention. However, almost all these studies were performed from 1980s to early 1990s, in the pre-surfactant era, and the study populations were composed of mid-preterm infants with GAs of approximately 30 weeks (wks). There is considerable difference in 'preterm infants' between the pre- and post-surfactant eras; modern neonatal medicine mainly treats preterm infants of 28 wks GA or less. Therefore, these results are difficult to apply in modern neonatal care. Before considering new trials of VE supplementation, we should fully understand modern neonatal medicine, especially the recent method of oxygen supplementation. Additionally, a deeper understanding of recent progress in pathophysiology and therapies for possible target diseases is necessary to decide whether VE administration is still worth re-challenging in modern neonatal intensive care units (NICUs). In this review, we present recent concepts and therapeutic trends in ROP, BPD, and GMH-IVH for those unfamiliar with neonatal medicine. Numerous studies have reported the possible involvement of reactive oxygen species (ROS)-induced damage in relation to supplemental oxygen use, inflammation, and immature antioxidant defense in the development of both BPD and ROP. Various antioxidants effectively prevented the exacerbation of BPD and ROP in animal models. In the future, VE should be re-attempted as a complementary factor in combination with various therapies for BPD, ROP, and GMH-IVH. Because VE is a natural and safe supplement, we are certain that it will attract attention again in preterm medicine.
Collapse
Affiliation(s)
- Tohru Ogihara
- Division of Neonatology, Department of Pediatrics, Osaka Medical and Pharmaceutical University, Takatsuki, Osaka, Japan.
| | - Makoto Mino
- Division of Neonatology, Department of Pediatrics, Osaka Medical and Pharmaceutical University, Takatsuki, Osaka, Japan
| |
Collapse
|
80
|
Ordóñez-Díaz MD, Gil-Campos M, Flores-Rojas K, Muñoz-Villanueva MC, Mesa MD, de la Torre-Aguilar MJ, Gil Á, Pérez-Navero JL. Impaired Antioxidant Defence Status Is Associated With Metabolic-Inflammatory Risk Factors in Preterm Children With Extrauterine Growth Restriction: The BIORICA Cohort Study. Front Nutr 2022; 8:793862. [PMID: 34993223 PMCID: PMC8724254 DOI: 10.3389/fnut.2021.793862] [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: 10/12/2021] [Accepted: 11/29/2021] [Indexed: 11/13/2022] Open
Abstract
Introduction: An impaired antioxidant status has been described during foetal growth restriction (FGR). Similarly, the antioxidant defence system can be compromised in preterm children with extrauterine growth restriction (EUGR). The aim of this prospective study was to evaluate the antioxidant status in prepubertal children with a history of prematurity without FGR, with and without EUGR, compared to a healthy group. Methods: In total, 211 children were recruited and classified into three groups: 38 with a history of prematurity and EUGR; 50 with a history of prematurity and adequate extrauterine growth (AEUG); and 123 control children born at term. Catalase (CAT), superoxide dismutase (SOD), glutathione peroxidase (GPx) and glutathione reductase (GR) activities were assessed in lysed erythrocytes with spectrophotometric methods. Plasma levels of the antioxidants α-tocopherol, retinol and β-carotene were determined through solvent extraction and ultra-high-pressure liquid chromatography coupled to mass spectrometry. Results: Children with the antecedent of EUGR and prematurity had lower CAT activity than the other two groups and lower GPx activity than the control children. Lower SOD, GPx and GR activities were observed in the AEUG group compared to the controls. However, higher concentrations of α-tocopherol and β-carotene were found in the EUGR group compared to the other groups; retinol levels were also higher in EUGR than in AEUG children. In EUGR and AEUG children, enzymatic antioxidant activities and plasma antioxidants were associated with metabolic syndrome components and pro-inflammatory biomarkers. Conclusions: This study reveals, for the first time, that the EUGR condition and prematurity appear to be linked to an impairment of the antioxidant defence status, which might condition an increased risk of adverse metabolic outcomes later in life.
Collapse
Affiliation(s)
- María Dolores Ordóñez-Díaz
- Unit of Neonatology, Department of Paediatrics, Maimonides Biomedical Research Institute of Cordoba, Reina Sofia Hospital, University of Córdoba, Córdoba, Spain
| | - Mercedes Gil-Campos
- Unit of Metabolism and Paediatric Research, Maimonides Biomedical Research Institute of Cordoba, Reina Sofia University Hospital, University of Córdoba, Córdoba, Spain.,Biomedical Research Center-Pathophysiology of Obesity and Nutrition, Carlos III Health Institute, Madrid, Spain
| | - Katherine Flores-Rojas
- Unit of Metabolism and Paediatric Research, Maimonides Biomedical Research Institute of Cordoba, Reina Sofia University Hospital, University of Córdoba, Córdoba, Spain.,Biomedical Research Center-Pathophysiology of Obesity and Nutrition, Carlos III Health Institute, Madrid, Spain
| | | | - María Dolores Mesa
- Department of Biochemistry and Molecular Biology II, Center of Biomedical Research, Institute of Nutrition and Food Technology, University of Granada, Granada, Spain.,Granada Biosanitary Research Institute (ibs.Granada), Granada, Spain
| | - María José de la Torre-Aguilar
- Unit of Metabolism and Paediatric Research, Maimonides Biomedical Research Institute of Cordoba, Reina Sofia University Hospital, University of Córdoba, Córdoba, Spain
| | - Ángel Gil
- Biomedical Research Center-Pathophysiology of Obesity and Nutrition, Carlos III Health Institute, Madrid, Spain.,Department of Biochemistry and Molecular Biology II, Center of Biomedical Research, Institute of Nutrition and Food Technology, University of Granada, Granada, Spain.,Granada Biosanitary Research Institute (ibs.Granada), Granada, Spain
| | - Juan Luis Pérez-Navero
- Unit of Metabolism and Paediatric Research, Maimonides Biomedical Research Institute of Cordoba, Reina Sofia University Hospital, University of Córdoba, Córdoba, Spain.,Biomedical Research Center-Rare Diseases (CIBERER), Carlos III Health Institute, Madrid, Spain
| |
Collapse
|
81
|
Teofili L, Papacci P, Bartolo M, Molisso A, Orlando N, Pane L, Giannantonio C, Serrao F, Bianchi M, Valentini CG, Pellegrino C, Baldascino A, Carducci B, Lepore D, Vento G. Transfusion-Free Survival Predicts Severe Retinopathy in Preterm Neonates. Front Pediatr 2022; 10:814194. [PMID: 35223696 PMCID: PMC8866869 DOI: 10.3389/fped.2022.814194] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Accepted: 01/04/2022] [Indexed: 12/23/2022] Open
Abstract
Repeated red blood cell (RBC) transfusions are thought to increase the risk for retinopathy of prematurity (ROP), likely due to a critical fetal hemoglobin (HbF) reduction. In this study, we investigated if the postmenstrual age (PMA) of neonates at transfusion influences the risk for ROP. We estimated the cumulative transfusion-free survival (TFS) in a series of 100 preterm neonates receiving one or more RBC units. TFS was calculated by censoring patients at first transfusion and expressing the time between birth and transfusion as either PMA or postnatal day. Then, we investigated if TFS predicted the occurrence of severe ROP, defined as ROP stage 3 or higher. We found that neonates with severe ROP displayed a significantly shorter TFS expressed according to their PMA (p = 0.001), with similar TFS according to postnatal days. At receiver operating characteristic (ROC) curve analysis, receiving an RBC unit before week 28 of PMA predicted severe ROP with a sensitivity of 64% and a specificity of 78%. In addition, receiving a second RBC unit before the PMA of 29 weeks predicted severe ROP with a sensitivity of 75% and a specificity of 69%. At multivariate analysis, PMA at the second transfusion was even more informative than at first transfusion and outperformed all other variables in predicting severe ROP, with an odds ratio of 4.554 (95% CI 1.332-15.573, p = 0.016). Since HbF decrease is greater after multiple RBC transfusions, it is conceivable that neonates receiving more than one unit before the PMA of 29 weeks may be exposed to a greater disturbance of retinal vascularization. Any strategy aimed at preventing the critical HbF decrease at this low age might potentially reduce the risk for severe ROP.
Collapse
Affiliation(s)
- Luciana Teofili
- Divisione di Medicina Trasfusionale, Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica, ed Ematologia, Fondazione Policlinico Universitario A. Gemelli Istituto di Ricerca e Cura a Carattere Scientifico, Rome, Italy.,Sezione di Ematologia, Dipartimento di Scienze Radiologiche ed Ematologiche, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Patrizia Papacci
- Divisione di Neonatologia, Dipartimento per le Scienze della Salute della Donna, del Bambino e di Sanità Pubblica, Fondazione Policlinico Universitario A. Gemelli, Istituto di Ricerca e Cura a Carattere Scientifico, Rome, Italy.,Dipartimento di Scienze della Vita e Sanità Pubblica, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Martina Bartolo
- Divisione di Medicina Trasfusionale, Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica, ed Ematologia, Fondazione Policlinico Universitario A. Gemelli Istituto di Ricerca e Cura a Carattere Scientifico, Rome, Italy
| | - Anna Molisso
- Divisione di Neonatologia, Dipartimento per le Scienze della Salute della Donna, del Bambino e di Sanità Pubblica, Fondazione Policlinico Universitario A. Gemelli, Istituto di Ricerca e Cura a Carattere Scientifico, Rome, Italy
| | - Nicoletta Orlando
- Divisione di Medicina Trasfusionale, Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica, ed Ematologia, Fondazione Policlinico Universitario A. Gemelli Istituto di Ricerca e Cura a Carattere Scientifico, Rome, Italy
| | - Lucia Pane
- Divisione di Neonatologia, Dipartimento per le Scienze della Salute della Donna, del Bambino e di Sanità Pubblica, Fondazione Policlinico Universitario A. Gemelli, Istituto di Ricerca e Cura a Carattere Scientifico, Rome, Italy
| | - Carmen Giannantonio
- Divisione di Neonatologia, Dipartimento per le Scienze della Salute della Donna, del Bambino e di Sanità Pubblica, Fondazione Policlinico Universitario A. Gemelli, Istituto di Ricerca e Cura a Carattere Scientifico, Rome, Italy
| | - Francesca Serrao
- Divisione di Neonatologia, Dipartimento per le Scienze della Salute della Donna, del Bambino e di Sanità Pubblica, Fondazione Policlinico Universitario A. Gemelli, Istituto di Ricerca e Cura a Carattere Scientifico, Rome, Italy
| | - Maria Bianchi
- Divisione di Medicina Trasfusionale, Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica, ed Ematologia, Fondazione Policlinico Universitario A. Gemelli Istituto di Ricerca e Cura a Carattere Scientifico, Rome, Italy
| | - Caterina Giovanna Valentini
- Divisione di Medicina Trasfusionale, Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica, ed Ematologia, Fondazione Policlinico Universitario A. Gemelli Istituto di Ricerca e Cura a Carattere Scientifico, Rome, Italy
| | - Claudio Pellegrino
- Divisione di Medicina Trasfusionale, Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica, ed Ematologia, Fondazione Policlinico Universitario A. Gemelli Istituto di Ricerca e Cura a Carattere Scientifico, Rome, Italy.,Sezione di Ematologia, Dipartimento di Scienze Radiologiche ed Ematologiche, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Antonio Baldascino
- Divisione di Oculistica, Dipartimento di Scienze dell'Invecchiamento, Neurologiche, Ortopediche e della Testa-Collo, Fondazione Policlinico Universitario A. Gemelli Istituto di Ricerca e Cura a Carattere Scientifico, Rome, Italy
| | - Brigida Carducci
- Divisione di Ostetricia e Patologia Ostetrica, Dipartimento per le Scienze della Salute della Donna, del Bambino e di Sanità Pubblica, Fondazione Policlinico Universitario A. Gemelli, Istituto di Ricerca e Cura a Carattere Scientifico, Rome, Italy
| | - Domenico Lepore
- Divisione di Oculistica, Dipartimento di Scienze dell'Invecchiamento, Neurologiche, Ortopediche e della Testa-Collo, Fondazione Policlinico Universitario A. Gemelli Istituto di Ricerca e Cura a Carattere Scientifico, Rome, Italy.,Dipartimento Testa-Collo e Organi di Senso, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Giovanni Vento
- Divisione di Neonatologia, Dipartimento per le Scienze della Salute della Donna, del Bambino e di Sanità Pubblica, Fondazione Policlinico Universitario A. Gemelli, Istituto di Ricerca e Cura a Carattere Scientifico, Rome, Italy.,Dipartimento di Scienze della Vita e Sanità Pubblica, Università Cattolica del Sacro Cuore, Rome, Italy
| |
Collapse
|
82
|
Antioxidant Effect of Melatonin in Preterm Newborns. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:6308255. [PMID: 34840669 PMCID: PMC8626170 DOI: 10.1155/2021/6308255] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 07/08/2021] [Accepted: 10/29/2021] [Indexed: 01/13/2023]
Abstract
Introduction Preterm infants are at risk of free radical-mediated diseases from oxidative stress (OS) injury. Increased free radical generation has been demonstrated in preterm infants during the first seven days of life. Melatonin (MEL) is a powerful antioxidant and scavenger of free radicals. In preterm neonates, melatonin deficiency has been reported. Exogenous melatonin administration appears a promising strategy in the treatment of neonatal morbidities in which OS has a leading role. Objective The aim was to evaluate plasma MEL concentrations and OS biomarkers in preterm newborns after early administration of melatonin. Methods A prospective, randomized double-blind placebo-controlled pilot study was conducted from January 2019 to September 2020. Thirty-six preterm newborns were enrolled. Starting from the first day of life, 21 received a single dose of oral melatonin 0.5 mg/kg once a day, in the morning (MEL group); 15 newborns received an equivalent dose of placebo (placebo group). Samples of 0.2 mL of plasma were collected at 24 and 48 hours after MEL administration. Plasma concentrations of melatonin, non-protein-bound iron (NPBI), advanced oxidation protein products (AOPP), and F2-isoprostanes (F2-Isopr) were measured. Babies were clinically followed until discharge. Results At 24 and 48 hours after MEL administration, the MEL concentrations were significantly higher in the MEL group than in the placebo group (52759.30 ± 63529.09 vs. 28.57 ± 46.24 pg/mL and 279397.6 ± 516344.2 vs. 38.50 ± 44.01 pg/mL, respectively). NPBI and AOPP did not show any statistically significant differences between the groups both at 24 and 48 hours. At 48 hours, the mean blood concentrations of F2-Isopr were significantly lower in the MEL group than in the placebo group (36.48 ± 33.85 pg/mL vs.89.97 ± 52.01 pg/mL). Conclusions Early melatonin administration in preterm newborns reduces lipid peroxidation in the first days of life showing a potential role to protect high-risk newborns. Trial Registration. This trial is registered with NCT04785183, Early Supplementation of Melatonin in Preterm Newborns: the Effects on Oxidative Stress.
Collapse
|
83
|
Chełchowska M, Gajewska J, Ambroszkiewicz J, Mazur J, Ołtarzewski M, Maciejewski TM. Influence of Oxidative Stress Generated by Smoking during Pregnancy on Glutathione Status in Mother-Newborn Pairs. Antioxidants (Basel) 2021; 10:antiox10121866. [PMID: 34942969 PMCID: PMC8698311 DOI: 10.3390/antiox10121866] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 11/15/2021] [Accepted: 11/21/2021] [Indexed: 11/16/2022] Open
Abstract
Glutathione plays a key role in maintaining a physiological balance between prooxidants and antioxidants in the human body. Therefore, we examined the influence of maternal smoking as a source of oxidative stress measured by total oxidant capacity (TOC) on reduced glutathione (GSH), oxidized glutathione (GSSG), glutathione peroxidase (GPx-3), and reductase (GR) amount in maternal and umbilical cord blood in 110 (45 smoking and 65 non-smoking) mother-newborn pairs. Concentrations of glutathione status markers and TOC were evaluated by competitive inhibition enzyme immunoassay technique. Plasma TOC levels were significantly higher and the GSH/GSSG ratio, which is considered an index of the cell’s redox status, were significantly lower in smoking women and their offspring than in non-smoking pairs. Decreased GR levels were found in smoking mothers and their newborns compared with similar non-smoking groups. Although plasma GPx-3 concentrations were similar in both maternal groups, in the cord blood of newborns exposed to tobacco smoke in utero they were reduced compared with the levels observed in children of tobacco abstinent mothers. Oxidative stress generated by tobacco smoke impairs glutathione homeostasis in both the mother and the newborn. The severity of oxidative processes in the mother co-existing with the reduced potential of antioxidant systems may have a negative effect on the oxidative-antioxidant balance in the newborn.
Collapse
Affiliation(s)
- Magdalena Chełchowska
- Department of Screening Tests and Metabolic Diagnostics, Institute of Mother and Child, Kasprzaka 17a, 01-211 Warsaw, Poland; (J.G.); (J.A.); (M.O.)
- Correspondence: ; Tel./Fax: +48-2-2327-7260
| | - Joanna Gajewska
- Department of Screening Tests and Metabolic Diagnostics, Institute of Mother and Child, Kasprzaka 17a, 01-211 Warsaw, Poland; (J.G.); (J.A.); (M.O.)
| | - Jadwiga Ambroszkiewicz
- Department of Screening Tests and Metabolic Diagnostics, Institute of Mother and Child, Kasprzaka 17a, 01-211 Warsaw, Poland; (J.G.); (J.A.); (M.O.)
| | - Joanna Mazur
- Department of Humanization in Medicine and Sexology, Collegium Medicum University of Zielona Góra, 65-729 Zielona Góra, Poland;
| | - Mariusz Ołtarzewski
- Department of Screening Tests and Metabolic Diagnostics, Institute of Mother and Child, Kasprzaka 17a, 01-211 Warsaw, Poland; (J.G.); (J.A.); (M.O.)
| | - Tomasz M. Maciejewski
- Clinic of Obstetrics and Gynaecology, Institute of Mother and Child, Kasprzaka 17a, 01-211 Warsaw, Poland;
| |
Collapse
|
84
|
Cannavò L, Perrone S, Viola V, Marseglia L, Di Rosa G, Gitto E. Oxidative Stress and Respiratory Diseases in Preterm Newborns. Int J Mol Sci 2021; 22:ijms222212504. [PMID: 34830385 PMCID: PMC8625766 DOI: 10.3390/ijms222212504] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 11/08/2021] [Accepted: 11/17/2021] [Indexed: 01/01/2023] Open
Abstract
Premature infants are exposed to increased generation of reactive oxygen species, and on the other hand, they have a deficient antioxidant defense system. Oxidative insult is a salient part of lung injury that begins as acute inflammatory injury in respiratory distress disease and then evolves into chronic and structural scarring leading to bronchopulmonary dysplasia. Oxidative stress is also involved in the pathogenesis of pulmonary hypertension in newborns through the modulation of the vascular tone and the response to pulmonary vasodilators, with consequent decrease in the density of the pulmonary vessels and thickening of the pulmonary arteriolar walls. Oxidative stress has been recognized as both a trigger and an endpoint for several events, including inflammation, hypoxia, hyperoxia, drugs, transfusions, and mechanical ventilation, with impairment of pulmonary function and prolonged lung damage. Redoxomics is the most fascinating new measure to address lung damage due to oxidative stress. The new challenge is to use omics data to discover a set of biomarkers useful in diagnosis, prognosis, and formulating optimal and individualized neonatal care. The aim of this review was to examine the most recent evidence on the relationship between oxidative stress and lung diseases in preterm newborns. What is currently known regarding oxidative stress-related lung injury pathogenesis and the available preventive and therapeutic strategies are also discussed.
Collapse
Affiliation(s)
- Laura Cannavò
- Neonatal and Pediatric Intensive Care Unit, Department of Human Pathology of the Adult and Developmental Age “Gaetano Barresi”, University of Messina, 98125 Messina, Italy; (L.C.); (V.V.); (L.M.); (E.G.)
| | - Serafina Perrone
- Neonatology Unity, Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy
- Correspondence: ; Tel.: +39-0521-703518
| | - Valeria Viola
- Neonatal and Pediatric Intensive Care Unit, Department of Human Pathology of the Adult and Developmental Age “Gaetano Barresi”, University of Messina, 98125 Messina, Italy; (L.C.); (V.V.); (L.M.); (E.G.)
| | - Lucia Marseglia
- Neonatal and Pediatric Intensive Care Unit, Department of Human Pathology of the Adult and Developmental Age “Gaetano Barresi”, University of Messina, 98125 Messina, Italy; (L.C.); (V.V.); (L.M.); (E.G.)
| | - Gabriella Di Rosa
- Unit of Child Neurology and Psychiatry, Department of Human Pathology of the Adult and Developmental Age “Gaetano Barresi”, University of Messina, 98125 Messina, Italy;
| | - Eloisa Gitto
- Neonatal and Pediatric Intensive Care Unit, Department of Human Pathology of the Adult and Developmental Age “Gaetano Barresi”, University of Messina, 98125 Messina, Italy; (L.C.); (V.V.); (L.M.); (E.G.)
| |
Collapse
|
85
|
Fetal hemoglobin levels in premature newborns. Should we reconsider transfusion of adult donor blood? J Pediatr Surg 2021; 56:1944-1948. [PMID: 34052004 DOI: 10.1016/j.jpedsurg.2021.04.018] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 03/26/2021] [Accepted: 04/12/2021] [Indexed: 01/07/2023]
Abstract
PURPOSE The aim of this study was to assess the percent decrease in fetal hemoglobin (HbF) after transfusion of adult-derived donor packed red blood cell (pRBC) units in extremely low gestational age newborns (ELGANs). METHODS Control percent fetal hemoglobin (%HbF) levels were measured in newborn cord blood or peripheral blood samples in non-transfused patients prior to elective surgery. ELGANs were followed prospectively and %HbF was measured on residual post-test complete blood count (CBC) specimens. ELGAN %HbF values were compared to the control population and transfusions were recorded. RESULTS Initial mean %HbF in ELGANs (n=16) was 92.2±1.3% (range 90.2-95.1%), which is similar to the control group (n=25). Mean levels dropped to 61.1±11.1% (range 34.2-73.2%) after a single pRBC transfusion (n=9) and to a mean of 35.6±6.3% after an additional transfusion (n=5). %HbF levels trended upwards if no additional transfusions were given, but levels still remained lower than expected for gestational age through discharge (n=85 samples). CONCLUSIONS Percent fetal hemoglobin concentrations in ELGANs decrease precipitously after transfusion with adult donor pRBCs. Further studies are needed to evaluate the benefit of maintaining higher fetal hemoglobin concentrations in these patients and whether administration of HbF rather than adult donor pRBCs would improve patient outcomes.
Collapse
|
86
|
Behnke J, Dippel CM, Choi Y, Rekers L, Schmidt A, Lauer T, Dong Y, Behnke J, Zimmer KP, Bellusci S, Ehrhardt H. Oxygen Toxicity to the Immature Lung-Part II: The Unmet Clinical Need for Causal Therapy. Int J Mol Sci 2021; 22:10694. [PMID: 34639034 PMCID: PMC8508961 DOI: 10.3390/ijms221910694] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 09/30/2021] [Accepted: 09/30/2021] [Indexed: 12/11/2022] Open
Abstract
Oxygen toxicity continues to be one of the inevitable injuries to the immature lung. Reactive oxygen species (ROS) production is the initial step leading to lung injury and, subsequently, the development of bronchopulmonary dysplasia (BPD). Today, BPD remains the most important disease burden following preterm delivery and results in life-long restrictions in lung function and further important health sequelae. Despite the tremendous progress in the pathomechanistic understanding derived from preclinical models, the clinical needs for preventive or curative therapies remain unmet. This review summarizes the clinical progress on guiding oxygen delivery to the preterm infant and elaborates future directions of research that need to take into account both hyperoxia and hypoxia as ROS sources and BPD drivers. Many strategies have been tested within clinical trials based on the mechanistic understanding of ROS actions, but most have failed to prove efficacy. The majority of these studies were tested in an era before the latest modes of non-invasive respiratory support and surfactant application were introduced or were not appropriately powered. A comprehensive re-evaluation of enzymatic, antioxidant, and anti-inflammatory therapies to prevent ROS injury is therefore indispensable. Strategies will only succeed if they are applied in a timely and vigorous manner and with the appropriate outcome measures.
Collapse
Affiliation(s)
- Judith Behnke
- Department of General Pediatrics and Neonatology, Justus-Liebig-University, Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Lung Research Center (DZL), Feulgenstrasse 12, 35392 Giessen, Germany; (J.B.); (C.M.D.); (Y.C.); (L.R.); (A.S.); (T.L.); (Y.D.); (K.-P.Z.)
| | - Constanze M. Dippel
- Department of General Pediatrics and Neonatology, Justus-Liebig-University, Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Lung Research Center (DZL), Feulgenstrasse 12, 35392 Giessen, Germany; (J.B.); (C.M.D.); (Y.C.); (L.R.); (A.S.); (T.L.); (Y.D.); (K.-P.Z.)
| | - Yesi Choi
- Department of General Pediatrics and Neonatology, Justus-Liebig-University, Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Lung Research Center (DZL), Feulgenstrasse 12, 35392 Giessen, Germany; (J.B.); (C.M.D.); (Y.C.); (L.R.); (A.S.); (T.L.); (Y.D.); (K.-P.Z.)
| | - Lisa Rekers
- Department of General Pediatrics and Neonatology, Justus-Liebig-University, Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Lung Research Center (DZL), Feulgenstrasse 12, 35392 Giessen, Germany; (J.B.); (C.M.D.); (Y.C.); (L.R.); (A.S.); (T.L.); (Y.D.); (K.-P.Z.)
| | - Annesuse Schmidt
- Department of General Pediatrics and Neonatology, Justus-Liebig-University, Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Lung Research Center (DZL), Feulgenstrasse 12, 35392 Giessen, Germany; (J.B.); (C.M.D.); (Y.C.); (L.R.); (A.S.); (T.L.); (Y.D.); (K.-P.Z.)
| | - Tina Lauer
- Department of General Pediatrics and Neonatology, Justus-Liebig-University, Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Lung Research Center (DZL), Feulgenstrasse 12, 35392 Giessen, Germany; (J.B.); (C.M.D.); (Y.C.); (L.R.); (A.S.); (T.L.); (Y.D.); (K.-P.Z.)
| | - Ying Dong
- Department of General Pediatrics and Neonatology, Justus-Liebig-University, Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Lung Research Center (DZL), Feulgenstrasse 12, 35392 Giessen, Germany; (J.B.); (C.M.D.); (Y.C.); (L.R.); (A.S.); (T.L.); (Y.D.); (K.-P.Z.)
| | - Jonas Behnke
- Department of Internal Medicine V, Justus-Liebig-University, Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Lung Research Center (DZL), Klinikstrasse 33, 35392 Giessen, Germany;
| | - Klaus-Peter Zimmer
- Department of General Pediatrics and Neonatology, Justus-Liebig-University, Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Lung Research Center (DZL), Feulgenstrasse 12, 35392 Giessen, Germany; (J.B.); (C.M.D.); (Y.C.); (L.R.); (A.S.); (T.L.); (Y.D.); (K.-P.Z.)
| | - Saverio Bellusci
- Department of Internal Medicine II, Universities of Giessen and Marburg Lung Center (UGMLC), Cardiopulmonary Institute (CPI), Member of the German Center for Lung Research (DZL), Justus-Liebig-University, Aulweg 130, 35392 Giessen, Germany;
| | - Harald Ehrhardt
- Department of General Pediatrics and Neonatology, Justus-Liebig-University, Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Lung Research Center (DZL), Feulgenstrasse 12, 35392 Giessen, Germany; (J.B.); (C.M.D.); (Y.C.); (L.R.); (A.S.); (T.L.); (Y.D.); (K.-P.Z.)
| |
Collapse
|
87
|
Joram N, Beqiri E, Pezzato S, Andrea M, Robba C, Liet JM, Chenouard A, Bourgoin P, Czosnyka M, Léger PL, Smielewski P. Impact of Arterial Carbon Dioxide and Oxygen Content on Cerebral Autoregulation Monitoring Among Children Supported by ECMO. Neurocrit Care 2021; 35:480-490. [PMID: 33686559 DOI: 10.1007/s12028-021-01201-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Accepted: 01/29/2021] [Indexed: 02/05/2023]
Abstract
BACKGROUND Cerebral autoregulation (CA) impairment is associated with neurological complications among children supported by extracorporeal membrane oxygenation (ECMO). Severe variations of arterial CO2 (PaCO2) and O2 (PaO2) tension after ECMO onset are common and associate with mortality and poor neurological outcome. The impact of gas exchange on CA among critically ill patients is poorly studied. METHODS Retrospective analysis of data collected prospectively from 30 children treated with veno-arterial or veno-venous ECMO in the PICU of Nantes University Hospital, France. A correlation coefficient between the variations of regional cerebral oxygen saturation (rSO2) and the variations of mean arterial blood pressure (MAP) was calculated as an index of CA (cerebral oxygenation reactivity index, COx). Cox-MAP plots were investigated allowing determining lower limit of autoregulation (LLA) and upper limit of autoregulation (ULA) limits of autoregulation. Age-based normal blood pressure was used to adjust the MAP, LLA, and ULA data from each patient and then reported as percentage (nMAP, nLLA, and nULA, respectively). RSO2, COx, nMAP, nLLA, and nULA values were averaged over one hour before each arterial blood gas (ABG) sample during ECMO run. RESULTS Thirty children (median age 4.8 months [Interquartile range (IQR) 0.7-39.1], median weight 5 kg [IQR 4-15]) experiencing 31 ECMO runs were included in the study. Three hundred and ninety ABGs were analyzed. The highest values of COx were observed on day 1 (D1) of ECMO. The relationship between COx and PaCO2 was nonlinear, but COx values tended to be lower in case of hypercapnia compared to normocapnia. During the whole ECMO run, a weak but significant correlation between PaCO2 and nULA was observed (R = 0.432, p = 0.02). On D1 of ECMO, this correlation was stronger (R = 0.85, p = 0.03) and a positive correlation between nLLA and PaCO2 was also found (R = 0.726, p < 0.001). A very weak negative correlation between PaO2 and nULA was observed within the whole ECMO run and on D1 of ECMO (R = -0.07 p = 0.04 and R = -0.135 p = <0.001, respectively). The difference between nULA and nLLA representing the span of the autoregulation plateau was positively correlated with PaCO2 and negatively correlated with PaO2 (R = 0.224, p = 0.01 and R = -0.051, p = 0.004, respectively). CONCLUSIONS We observed a complex relationship between PaCO2 and CA, influenced by the level of blood pressure. Hypercapnia seems to be globally protective in normotensive or hypertensive condition, while, in case of very low MAP, hypercapnia may disturb CA as it increases LLA. These data add additional arguments for very cautiously lower PaCO2, especially after ECMO start.
Collapse
Affiliation(s)
- Nicolas Joram
- Pediatric Intensive Care Unit, University Hospital of Nantes, Nantes, France. .,Clinical Investigation Center (CIC) 1413, University Hospital of Nantes, Nantes, France. .,INSERM U955-ENVA, University Paris 12, Paris, France.
| | - Erta Beqiri
- Brain Physics Laboratory, Division of Neurosurgery, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK.,Department of Physiology and Transplantation, Milan University, Milan, Italy
| | - Stefano Pezzato
- Pediatric Intensive Care Unit, IRCCS Giannina Gaslini Institute, Genoa, Italy
| | - Moscatelli Andrea
- Pediatric Intensive Care Unit, IRCCS Giannina Gaslini Institute, Genoa, Italy
| | - Chiara Robba
- Brain Physics Laboratory, Division of Neurosurgery, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK.,Policlinico San Martino IRCCS for Oncology and Neuroscience, Genova, Italy
| | - Jean-Michel Liet
- Pediatric Intensive Care Unit, University Hospital of Nantes, Nantes, France.,Clinical Investigation Center (CIC) 1413, University Hospital of Nantes, Nantes, France
| | - Alexis Chenouard
- Pediatric Intensive Care Unit, University Hospital of Nantes, Nantes, France.,Clinical Investigation Center (CIC) 1413, University Hospital of Nantes, Nantes, France
| | - Pierre Bourgoin
- Pediatric Intensive Care Unit, University Hospital of Nantes, Nantes, France.,Clinical Investigation Center (CIC) 1413, University Hospital of Nantes, Nantes, France
| | - Marek Czosnyka
- Brain Physics Laboratory, Division of Neurosurgery, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Pierre-Louis Léger
- INSERM U955-ENVA, University Paris 12, Paris, France.,Pediatric Intensive Care Unit, Trousseau University Hospital, Paris, France
| | - Peter Smielewski
- Brain Physics Laboratory, Division of Neurosurgery, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| |
Collapse
|
88
|
Paiva WS, Queiroz MF, Araujo Sabry D, Santiago ALCMA, Sassaki GL, Batista ACL, Rocha HAO. Preparation, Structural Characterization, and Property Investigation of Gallic Acid-Grafted Fungal Chitosan Conjugate. J Fungi (Basel) 2021; 7:812. [PMID: 34682234 PMCID: PMC8540519 DOI: 10.3390/jof7100812] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 09/23/2021] [Accepted: 09/23/2021] [Indexed: 02/06/2023] Open
Abstract
Oxidative stress is the cause of numerous diseases in humans; therefore, there has been a continuous search for novel antioxidant molecules. Fungal chitosan is an attractive molecule that has several applications (antifungal, antibacterial, anticancer and antiparasitic action) owing to its unique characteristics; however, it exhibits low antioxidant activity. The aim of this study was to obtain fungal chitosan (Chit-F) from the fungus Rhizopus arrhizus and synthesize its derivative, fungal chitosan-gallic acid (Chit-FGal), as a novel antioxidant chitosan derivative for biomedical use. A low molecular weight Chi-F (~3.0 kDa) with a degree of deacetylation of 86% was obtained from this fungus. Chit-FGal (3.0 kDa) was synthesized by an efficient free radical-mediated method using hydrogen peroxide (H2O2) and ascorbic acid. Both Chit-F and Chit-FGal showed similar copper chelating activities; however, Chit-FGal was more efficient as an antioxidant, exhibiting twice the total antioxidant capacity than Chi-F (p < 0.05). Furthermore, H2O2 (0.06 M) promoted a 50% decrease in the viabilities of the 3T3 fibroblast cells. However, this effect was abolished in the presence of Chit-FGal (0.05-0.25 mg/mL), indicating that Chit-FGal protected the cells from oxidative damage. These results suggest that Chit-FGal may be a promising agent to combat oxidative stress.
Collapse
Affiliation(s)
- Weslley Souza Paiva
- Postgraduate Programe in Biotechnology (RENORBIO), Federal University of Rio Grande do Norte (UFRN), Natal 59078-970, RN, Brazil
| | - Moacir Fernandes Queiroz
- Biomedicine Departament, Medical Sciences College, Potiguar University (UNP), Natal 59056-000, RN, Brazil;
| | - Diego Araujo Sabry
- Laboratorio de Biotecnologia de Polímeros Naturais-BIOPOL, Departament of Biochemistry, Federal University of Rio Grande do Norte (UFRN), Natal 59078-970, RN, Brazil;
| | | | - Guilherme Lanzi Sassaki
- Department of Biochemistry and Molecular Biology, Federal University of Paraná, Curitiba 81531-980, PR, Brazil;
| | | | - Hugo Alexandre Oliveira Rocha
- Postgraduate Programe in Biotechnology (RENORBIO), Federal University of Rio Grande do Norte (UFRN), Natal 59078-970, RN, Brazil
- Laboratorio de Biotecnologia de Polímeros Naturais-BIOPOL, Departament of Biochemistry, Federal University of Rio Grande do Norte (UFRN), Natal 59078-970, RN, Brazil;
| |
Collapse
|
89
|
Male Disadvantage in Oxidative Stress-Associated Complications of Prematurity: A Systematic Review, Meta-Analysis and Meta-Regression. Antioxidants (Basel) 2021; 10:antiox10091490. [PMID: 34573122 PMCID: PMC8465696 DOI: 10.3390/antiox10091490] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 09/14/2021] [Indexed: 11/17/2022] Open
Abstract
A widely accepted concept is that boys are more susceptible than girls to oxidative stress-related complications of prematurity, including bronchopulmonary dysplasia (BPD), retinopathy of prematurity (ROP), necrotizing enterocolitis (NEC), intraventricular hemorrhage (IVH), and periventricular leukomalacia (PVL). We aimed to quantify the effect size of this male disadvantage by performing a systematic review and meta-analysis of cohort studies exploring the association between sex and complications of prematurity. Risk ratios (RRs) and 95% CIs were calculated by a random-effects model. Of 1365 potentially relevant studies, 41 met the inclusion criteria (625,680 infants). Male sex was associated with decreased risk of hypertensive disorders of pregnancy, fetal distress, and C-section, but increased risk of low Apgar score, intubation at birth, respiratory distress, surfactant use, pneumothorax, postnatal steroids, late onset sepsis, any NEC, NEC > stage 1 (RR 1.12, CI 1.06–1.18), any IVH, severe IVH (RR 1.28, CI 1.22–1.34), severe IVH or PVL, any BPD, moderate/severe BPD (RR 1.23, CI 1.18–1.27), severe ROP (RR 1.14, CI 1.07–1.22), and mortality (RR 1.23, CI 1.16–1.30). In conclusion, preterm boys have higher clinical instability and greater need for invasive interventions than preterm girls. This leads to a male disadvantage in mortality and short-term complications of prematurity.
Collapse
|
90
|
Tagliaferro T, Cayabyab R, Ramanathan R. Association between blood carboxyhemoglobin level and bronchopulmonary dysplasia in extremely low birthweight infants. J Investig Med 2021; 70:68-72. [PMID: 34493626 DOI: 10.1136/jim-2021-001967] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/27/2021] [Indexed: 11/04/2022]
Abstract
Carboxyhemoglobin (CO-Hb) can be endogenously formed in the presence of oxidative stress and may be elevated in inflammatory lung disease. There is lack of evidence of its relationship with the development of bronchopulmonary dysplasia (BPD) in extremely low birthweight (ELBW) infants. The objective of the study is to evaluate the relationship between blood CO-Hb levels in the first 14 days of life (DOL) in ELBW infants and the development of BPD at 36 weeks postmenstrual age (PMA). This is a retrospective cohort study of 58 ELBW infants born at LAC-USC Medical Center between June 2015 and and June 2019 who survived to 36 weeks PMA. CO-Hb values were collected daily from DOL 1 to DOL 14. BPD definition using the recent 2019 NICHD criteria was used. Multivariate logistic regression was performed to determine the association between blood CO-Hb levels and BPD. Receiver operator curve was used to evaluate the ability of the median fraction of inspired oxygen (FiO2) level used at DOL 11-14 in discriminating absent to mild BPD versus moderate to severe BPD. 58 ELBW infants were included in the study. 24 (41%) were diagnosed with moderate to severe BPD, while 34 (59%) were diagnosed with no to mild BPD. Severity of BPD was fairly discriminated by FiO2 at DOL 11-14, but not with CO-Hb levels at any point within the first 14 DOL. The role and mechanism of CO-Hb production in this population need to be further studied.
Collapse
Affiliation(s)
- Thea Tagliaferro
- Keck School of Medicine/Pediatrics/Division of Neonatology, USC, Los Angeles, California, USA
| | - Rowena Cayabyab
- Keck School of Medicine/Pediatrics/Division of Neonatology, USC, Los Angeles, California, USA
| | - Rangasamy Ramanathan
- Keck School of Medicine/Pediatrics/Division of Neonatology, USC, Los Angeles, California, USA
| |
Collapse
|
91
|
Dong W, Zhu X, Liu X, Zhao X, Lei X, Kang L, Liu L. Role of the SENP1-SIRT1 pathway in hyperoxia-induced alveolar epithelial cell injury. Free Radic Biol Med 2021; 173:142-150. [PMID: 34311030 DOI: 10.1016/j.freeradbiomed.2021.07.027] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 07/12/2021] [Accepted: 07/20/2021] [Indexed: 12/29/2022]
Abstract
Bronchopulmonary dysplasia (BPD) is a common complication in preterm infants, and its main pathogenesis partly involves oxidative stress. A large number of studies have shown that silent information regulator 1 (SIRT1) plays a protective role in oxidative stress. SUMO-specific protease 1 (SENP1) is vital in the nucleoplasmic distribution of SIRT1 under stress. However, whether the SENP1-SIRT1 pathway is involved in the hyperoxic lung injury is unknown. Therefore, this study aimed to explore the role and related mechanisms of the SENP1-SIRT1 pathway in hyperoxic lung injury. Peripheral blood mononuclear cells (PBMCs) from infants with BPD and SENP1-silenced alveolar epithelial cells were used as research models. PBMCs were isolated from the peripheral blood of premature infants. Next, the SENP1-silenced human alveolar epithelial cells were used to verify the role of the SENP1-SIRT1 pathway in vitro. The results indicated that the ROS level and the mRNA and protein expression of SENP1 increased in PBMCs of infants with BPD, but the expression of SIRT1 decreased in the nucleus and increased in the cytoplasm, and then the expression of acetyl-p53 (Ac-p53) increased. In the hyperoxic alveolar epithelial cell injury model, it seemed that hyperoxia could induce the same variation trend in the SENP1-SIRT1 pathway as in infants with BPD and then increased the expression of Ac-p53 and BAX, and cell apoptosis. Furthermore, silencing SENP1 could alleviate these hyperoxia-induced changes. These results suggested that SENP1 played an important role in hyperoxia-induced lung injury. It could regulate the expression and nucleoplasmic distribution of SIRT1 to inhibit its deacetylase activity, and then promoted cell apoptosis. Hence, SENP1 may become a potential intervention target of BPD in the future.
Collapse
Affiliation(s)
- Wenbin Dong
- Department of Neonatology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China; Division of Neonatology, Department of Pediatrics, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Xiaodan Zhu
- Division of Neonatology, Department of Pediatrics, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Xingling Liu
- Division of Neonatology, Department of Pediatrics, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Xu Zhao
- Division of Neonatology, Department of Pediatrics, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Xiaoping Lei
- Division of Neonatology, Department of Pediatrics, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Lan Kang
- Division of Neonatology, Department of Pediatrics, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Li Liu
- Department of Neonatology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China.
| |
Collapse
|
92
|
Sankaran D, Vali P, Chen P, Lesneski AL, Hardie ME, Alhassen Z, Wedgwood S, Wyckoff MH, Lakshminrusimha S. Randomized trial of oxygen weaning strategies following chest compressions during neonatal resuscitation. Pediatr Res 2021; 90:540-548. [PMID: 33941864 PMCID: PMC8530847 DOI: 10.1038/s41390-021-01551-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 02/08/2021] [Accepted: 04/10/2021] [Indexed: 02/08/2023]
Abstract
BACKGROUND The Neonatal Resuscitation Program (NRP) recommends using 100% O2 during chest compressions and adjusting FiO2 based on SpO2 after return of spontaneous circulation (ROSC). The optimal strategy for adjusting FiO2 is not known. METHODS Twenty-five near-term lambs asphyxiated by umbilical cord occlusion to cardiac arrest were resuscitated per NRP. Following ROSC, lambs were randomized to gradual decrease versus abrupt wean to 21% O2 followed by FiO2 titration to achieve NRP SpO2 targets. Carotid blood flow and blood gases were monitored. RESULTS Three minutes after ROSC, PaO2 was 229 ± 32 mmHg in gradual wean group compared to 57 ± 13 following abrupt wean to 21% O2 (p < 0.001). PaO2 remained high in the gradual wean group at 10 min after ROSC (110 ± 10 vs. 67 ± 12, p < 0.01) despite similar FiO2 (~0.3) in both groups. Cerebral O2 delivery (C-DO2) was higher above physiological range following ROSC with gradual wean (p < 0.05). Lower blood oxidized/reduced glutathione ratio (suggesting less oxidative stress) was observed with abrupt wean. CONCLUSION Weaning FiO2 abruptly to 0.21 with adjustment based on SpO2 prevents surge in PaO2 and C-DO2 and minimizes oxidative stress compared to gradual weaning from 100% O2 following ROSC. Clinical trials with neurodevelopmental outcomes comparing post-ROSC FiO2 weaning strategies are warranted. IMPACT In a lamb model of perinatal asphyxial cardiac arrest, abrupt weaning of inspired oxygen to 21% prevents excessive oxygen delivery to the brain and oxidative stress compared to gradual weaning from 100% oxygen following return of spontaneous circulation. Clinical studies assessing neurodevelopmental outcomes comparing abrupt and gradual weaning of inspired oxygen after recovery from neonatal asphyxial arrest are warranted.
Collapse
Affiliation(s)
- Deepika Sankaran
- Division of Neonatology, Department of Pediatrics, University of California Davis, Sacramento, CA, USA.
| | - Payam Vali
- Division of Neonatology, Department of Pediatrics, University of California Davis, Sacramento, CA, USA
| | - Peggy Chen
- Division of Neonatology, Department of Pediatrics, University of California Davis, Sacramento, CA, USA
| | - Amy L Lesneski
- Division of Neonatology, Department of Pediatrics, University of California Davis, Sacramento, CA, USA
| | - Morgan E Hardie
- Division of Neonatology, Department of Pediatrics, University of California Davis, Sacramento, CA, USA
| | - Ziad Alhassen
- Division of Neonatology, Department of Pediatrics, University of California Davis, Sacramento, CA, USA
| | - Stephen Wedgwood
- Division of Neonatology, Department of Pediatrics, University of California Davis, Sacramento, CA, USA
| | - Myra H Wyckoff
- Division of Neonatology, Department of Pediatrics, University of Texas South Western (UTSW), Dallas, TX, USA
| | - Satyan Lakshminrusimha
- Division of Neonatology, Department of Pediatrics, University of California Davis, Sacramento, CA, USA
| |
Collapse
|
93
|
The newborn sheep translational model for pulmonary arterial hypertension of the neonate at high altitude. J Dev Orig Health Dis 2021; 11:452-463. [PMID: 32705972 DOI: 10.1017/s2040174420000616] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Chronic hypoxia during gestation induces greater occurrence of perinatal complications such as intrauterine growth restriction, fetal hypoxia, newborn asphyxia, and respiratory distress, among others. This condition may also cause a failure in the transition of the fetal to neonatal circulation, inducing pulmonary arterial hypertension of the neonate (PAHN), a syndrome that involves pulmonary vascular dysfunction, increased vasoconstrictor tone and pathological remodeling. As this syndrome has a relatively low prevalence in lowlands (~7 per 1000 live births) and very little is known about its prevalence and clinical evolution in highlands (above 2500 meters), our understanding is very limited. Therefore, studies on appropriate animal models have been crucial to comprehend the mechanisms underlying this pathology. Considering the strengths and weaknesses of any animal model of human disease is fundamental to achieve an effective and meaningful translation to clinical practice. The sheep model has been used to study the normal and abnormal cardiovascular development of the fetus and the neonate for almost a century. The aim of this review is to highlight the advances in our knowledge on the programming of cardiopulmonary function with the use of high-altitude newborn sheep as a translational model of PAHN.
Collapse
|
94
|
D'Agrosa C, Cai CL, Siddiqui F, Deslouches K, Wadowski S, Aranda JV, Beharry KD. Comparison of coenzyme Q10 or fish oil for prevention of intermittent hypoxia-induced oxidative injury in neonatal rat lungs. Respir Res 2021; 22:196. [PMID: 34225702 PMCID: PMC8256540 DOI: 10.1186/s12931-021-01786-w] [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: 04/27/2021] [Accepted: 06/28/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Neonatal intermittent hypoxia (IH) results in oxidative distress in preterm infants with immature antioxidant systems, contributing to lung injury. Coenzyme Q10 (CoQ10) and fish oil protect against oxidative injury. We tested the hypothesis that CoQ10 is more effective than fish oil for prevention of IH-induced lung injury in neonatal rats. METHODS Newborn rats were exposed to two clinically relevant IH paradigms at birth (P0): (1) 50% O2 with brief hypoxia (12% O2); or (2) room air (RA) with brief hypoxia (12% O2), until P14 during which they were supplemented with daily oral CoQ10, fish oil, or olive oil from P0 to P14. Pups were studied at P14 or placed in RA until P21 with no further treatment. Lungs were assessed for histopathology and morphometry; biomarkers of oxidative stress and lipid peroxidation; and antioxidants. RESULTS Of the two neonatal IH paradigms 21%/12% O2 IH resulted in the most severe outcomes, evidenced by histopathology and morphometry. CoQ10 was effective for preserving lung architecture and reduction of IH-induced oxidative stress biomarkers. In contrast, fish oil resulted in significant adverse outcomes including oversimplified alveoli, hemorrhage, reduced secondary crest formation and thickened septae. This was associated with elevated oxidants and antioxidants activities. CONCLUSIONS Data suggest that higher FiO2 may be needed between IH episodes to curtail the damaging effects of IH, and to provide the lungs with necessary respite. The negative outcomes with fish oil supplementation suggest oxidative stress-induced lipid peroxidation.
Collapse
Affiliation(s)
- Christina D'Agrosa
- Department of Pediatrics and Ophthalmology, Division of Neonatal-Perinatal Medicine Clinical and Translational Research Labs, State University of New York, Downstate Medical Center, 450 Clarkson Avenue, Box 49, Brooklyn, NY, 11203, USA
| | - Charles L Cai
- Department of Pediatrics and Ophthalmology, Division of Neonatal-Perinatal Medicine Clinical and Translational Research Labs, State University of New York, Downstate Medical Center, 450 Clarkson Avenue, Box 49, Brooklyn, NY, 11203, USA
| | - Faisal Siddiqui
- Department of Pediatrics and Ophthalmology, Division of Neonatal-Perinatal Medicine Clinical and Translational Research Labs, State University of New York, Downstate Medical Center, 450 Clarkson Avenue, Box 49, Brooklyn, NY, 11203, USA
| | - Karen Deslouches
- Department of Pediatrics and Ophthalmology, Division of Neonatal-Perinatal Medicine Clinical and Translational Research Labs, State University of New York, Downstate Medical Center, 450 Clarkson Avenue, Box 49, Brooklyn, NY, 11203, USA
| | - Stephen Wadowski
- Department of Pediatrics and Ophthalmology, Division of Neonatal-Perinatal Medicine Clinical and Translational Research Labs, State University of New York, Downstate Medical Center, 450 Clarkson Avenue, Box 49, Brooklyn, NY, 11203, USA
| | - Jacob V Aranda
- Department of Pediatrics and Ophthalmology, Division of Neonatal-Perinatal Medicine Clinical and Translational Research Labs, State University of New York, Downstate Medical Center, 450 Clarkson Avenue, Box 49, Brooklyn, NY, 11203, USA.,Department of Ophthalmology, State University of New York, Downstate Medical Center, Brooklyn, NY, 11203, USA.,State University of New York Eye Institute, New York, NY, USA
| | - Kay D Beharry
- Department of Pediatrics and Ophthalmology, Division of Neonatal-Perinatal Medicine Clinical and Translational Research Labs, State University of New York, Downstate Medical Center, 450 Clarkson Avenue, Box 49, Brooklyn, NY, 11203, USA. .,Department of Ophthalmology, State University of New York, Downstate Medical Center, Brooklyn, NY, 11203, USA. .,State University of New York Eye Institute, New York, NY, USA.
| |
Collapse
|
95
|
Polyphenols and IUGR Pregnancies: Effects of the Antioxidant Hydroxytyrosol on Brain Neurochemistry and Development in a Porcine Model. Antioxidants (Basel) 2021; 10:antiox10060884. [PMID: 34073097 PMCID: PMC8227239 DOI: 10.3390/antiox10060884] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 05/19/2021] [Accepted: 05/22/2021] [Indexed: 12/30/2022] Open
Abstract
Supplementation of a mother’s diet with antioxidants, such as hydroxytyrosol (HTX), has been proposed to ameliorate the adverse phenotypes of fetuses at risk of intrauterine growth restriction. In the present study, sows were treated daily with or without 1.5 mg of HTX per kilogram of feed from day 35 of pregnancy (at 30% of total gestational period), and individuals were sampled at three different ages: 100-day-old fetuses and 1-month- and 6-month-old piglets. After euthanasia, the brain was removed and the hippocampus, amygdala, and prefrontal cortex were dissected. The profile of the catecholaminergic and serotoninergic neurotransmitters (NTs) was characterized and an immunohistochemical study of the hippocampus was performed. The results indicated that maternal supplementation with HTX during pregnancy affected the NT profile in a brain-area-dependant mode and it modified the process of neuron differentiation in the hippocampal CA1 and GD areas, indicating that cell differentiation occurred more rapidly in the HTX group. These effects were specific to the fetal period, concomitantly with HTX maternal supplementation, since no major differences remained between the control and treated groups in 1-month- and 6-month-old pigs.
Collapse
|
96
|
Di Fiore JM, Raffay TM. The relationship between intermittent hypoxemia events and neural outcomes in neonates. Exp Neurol 2021; 342:113753. [PMID: 33984336 DOI: 10.1016/j.expneurol.2021.113753] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 04/06/2021] [Accepted: 05/07/2021] [Indexed: 12/20/2022]
Abstract
This brief review examines 1) patterns of intermittent hypoxemia in extremely preterm infants during early postnatal life, 2) the relationship between neonatal intermittent hypoxemia exposure and outcomes in both human and animal models, 3) potential mechanistic pathways, and 4) future alterations in clinical care that may reduce morbidity. Intermittent hypoxemia events are pervasive in extremely preterm infants (<28 weeks gestation at birth) during early postnatal life. An increased frequency of intermittent hypoxemia events has been associated with a range of poor neural outcomes including language and cognitive delays, motor impairment, retinopathy of prematurity, impaired control of breathing, and intraventricular hemorrhage. Neonatal rodent models have shown that exposure to short repetitive cycles of hypoxia induce a pathophysiological cascade. However, not all patterns of intermittent hypoxia are deleterious and some may even improve neurodevelopmental outcomes. Therapeutic interventions include supplemental oxygen, pressure support and pharmacologic drugs but prolonged hyperoxia and pressure exposure have been associated with cardiopulmonary morbidity. Therefore, it becomes imperative to distinguish high risk from neutral and/or even beneficial patterns of intermittent hypoxemia during early postnatal life. Identification of such patterns could improve clinical care with targeted interventions for high-risk patterns and minimal or no exposure to treatment modalities for low-risk patterns.
Collapse
Affiliation(s)
- Juliann M Di Fiore
- Division of Neonatology, Rainbow Babies and Children's Hospital, Cleveland, OH, United States of America; Department of Pediatrics, Case Western Reserve University, Cleveland, OH, United States of America.
| | - Thomas M Raffay
- Division of Neonatology, Rainbow Babies and Children's Hospital, Cleveland, OH, United States of America; Department of Pediatrics, Case Western Reserve University, Cleveland, OH, United States of America
| |
Collapse
|
97
|
Krizova J, Hulkova M, Capek V, Mlejnek P, Silhavy J, Tesarova M, Zeman J, Hansikova H. Microarray and qPCR Analysis of Mitochondrial Metabolism Activation during Prenatal and Early Postnatal Development in Rats and Humans with Emphasis on CoQ 10 Biosynthesis. BIOLOGY 2021; 10:418. [PMID: 34066731 PMCID: PMC8150536 DOI: 10.3390/biology10050418] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Revised: 05/02/2021] [Accepted: 05/03/2021] [Indexed: 11/16/2022]
Abstract
At the end of the mammalian intra-uterine foetal development, a rapid switch from glycolytic to oxidative metabolism must proceed. Using microarray techniques, qPCR, enzyme activities and coenzyme Q content measurements, we describe perinatal mitochondrial metabolism acceleration in rat liver and skeletal muscle during the perinatal period and correlate the results with those in humans. Out of 1546 mitochondrial genes, we found significant changes in expression in 1119 and 827 genes in rat liver and skeletal muscle, respectively. The most remarkable expression shift occurred in the rat liver at least two days before birth. Coenzyme Q-based evaluation in both the rat model and human tissues showed the same trend: the total CoQ content is low prenatally, significantly increasing after birth in both the liver and skeletal muscle. We propose that an important regulator of rat coenzyme Q biosynthesis might be COQ8A, an atypical kinase involved in the biosynthesis of coenzyme Q. Our microarray data, a total of 16,557 RefSeq (Entrez) genes, have been deposited in NCBI's Gene Expression Omnibus and are freely available to the broad scientific community. Our microarray data could serve as a suitable background for finding key factors regulating mitochondrial metabolism and the preparation of the foetus for the transition to extra-uterine conditions.
Collapse
Affiliation(s)
- Jana Krizova
- Laboratory for Study of Mitochondrial Disorders, Department of Pediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, General University Hospital in Prague, Charles University, Ke Karlovu 2, 128 08 Prague 2, Czech Republic; (J.K.); (M.H.); (V.C.); (M.T.); (J.Z.)
| | - Martina Hulkova
- Laboratory for Study of Mitochondrial Disorders, Department of Pediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, General University Hospital in Prague, Charles University, Ke Karlovu 2, 128 08 Prague 2, Czech Republic; (J.K.); (M.H.); (V.C.); (M.T.); (J.Z.)
| | - Vaclav Capek
- Laboratory for Study of Mitochondrial Disorders, Department of Pediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, General University Hospital in Prague, Charles University, Ke Karlovu 2, 128 08 Prague 2, Czech Republic; (J.K.); (M.H.); (V.C.); (M.T.); (J.Z.)
| | - Petr Mlejnek
- Department of Genetics of Model Diseases, Institute of Physiology AS CR, v.v.i., Videnska 1083, 142 20 Prague 4, Czech Republic; (P.M.); (J.S.)
| | - Jan Silhavy
- Department of Genetics of Model Diseases, Institute of Physiology AS CR, v.v.i., Videnska 1083, 142 20 Prague 4, Czech Republic; (P.M.); (J.S.)
| | - Marketa Tesarova
- Laboratory for Study of Mitochondrial Disorders, Department of Pediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, General University Hospital in Prague, Charles University, Ke Karlovu 2, 128 08 Prague 2, Czech Republic; (J.K.); (M.H.); (V.C.); (M.T.); (J.Z.)
| | - Jiri Zeman
- Laboratory for Study of Mitochondrial Disorders, Department of Pediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, General University Hospital in Prague, Charles University, Ke Karlovu 2, 128 08 Prague 2, Czech Republic; (J.K.); (M.H.); (V.C.); (M.T.); (J.Z.)
| | - Hana Hansikova
- Laboratory for Study of Mitochondrial Disorders, Department of Pediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, General University Hospital in Prague, Charles University, Ke Karlovu 2, 128 08 Prague 2, Czech Republic; (J.K.); (M.H.); (V.C.); (M.T.); (J.Z.)
| |
Collapse
|
98
|
Go H, Ohto H, Nollet KE, Sato K, Ichikawa H, Kume Y, Kanai Y, Maeda H, Kashiwabara N, Ogasawara K, Sato M, Hashimoto K, Hosoya M. Red cell distribution width as a predictor for bronchopulmonary dysplasia in premature infants. Sci Rep 2021; 11:7221. [PMID: 33790386 PMCID: PMC8012706 DOI: 10.1038/s41598-021-86752-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Accepted: 03/19/2021] [Indexed: 01/21/2023] Open
Abstract
Bronchopulmonary dysplasia (BPD) is the most common morbidity complicating preterm birth. Red blood cell distribution width (RDW), a measure of the variation of red blood cell size, could reflect oxidative stress and chronic inflammation in many diseases such as cardiovascular, pulmonary, and other diseases. The objectives of the present study were to evaluate perinatal factors affecting RDW and to validate whether RDW could be a potential biomarker for BPD. A total of 176 preterm infants born at < 30 weeks were included in this study. They were categorized into BPD (n = 85) and non-BPD (n = 91) infants. RDW at birth and 14 days and 28 days of life (DOL 14, DOL 28) were measured. Clinical data were obtained from all subjects at Fukushima Medical University (Fukushima, Japan). The mean RDW at birth, DOL 14 and DOL 28 were 16.1%, 18.6%, 20.1%, respectively. Small for gestational age (SGA), chorioamnionitis (CAM), hypertensive disorders of pregnancy (HDP), gestational age and birth weight were significantly associated with RDW at birth. SGA, BPD and red blood cell (RBC) transfusion before DOL 14 were associated with RDW at DOL 14. BPD and RBC transfusion before DOL 14 were associated with RDW at DOL 28. Compared with non-BPD infants, mean RDW at DOL 14 (21.1% vs. 17.6%, P < 0.001) and DOL 28 (22.2% vs. 18.2%, P < 0.001) were significantly higher in BPD infants. Multivariate analysis revealed that RDW at DOL 28 was significantly higher in BPD infants (P = 0.001, odds ratio 1.63; 95% CI 1.22–2.19). Receiver operating characteristic analysis for RDW at DOL 28 in infants with and without BPD yielded an area under the curve of 0.87 (95% CI 0.78–0.91, P < 0.001). RDW at DOL 28 with mild BPD (18.1% vs. 21.3%, P < 0.001), moderate BPD (18.1% vs. 21.2%, P < 0.001), and severe BPD (18.1% vs. 24.0%, P < 0.001) were significantly higher than those with non-BPD, respectively. Furthermore, there are significant differences of RDW at DOL 28 among mild, moderate, and severe BPD. In summary, we conclude that RDW at DOL 28 could serve as a biomarker for predicting BPD and its severity. The mechanism by which RDW at DOL 28 is associated with the pathogenesis of BPD needs further elucidation.
Collapse
Affiliation(s)
- Hayato Go
- Department of Pediatrics, Fukushima Medical University School of Medicine, Hikarigaoka 1, Fukushima, Japan.
| | | | - Kenneth E Nollet
- Department of Blood Transfusion and Transplantation Immunology, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Kenichi Sato
- Department of Pediatrics, Fukushima Medical University School of Medicine, Hikarigaoka 1, Fukushima, Japan
| | - Hirotaka Ichikawa
- Department of Pediatrics, Fukushima Medical University School of Medicine, Hikarigaoka 1, Fukushima, Japan
| | - Yohei Kume
- Department of Pediatrics, Fukushima Medical University School of Medicine, Hikarigaoka 1, Fukushima, Japan
| | - Yuji Kanai
- Department of Pediatrics, Fukushima Medical University School of Medicine, Hikarigaoka 1, Fukushima, Japan
| | - Hajime Maeda
- Department of Pediatrics, Fukushima Medical University School of Medicine, Hikarigaoka 1, Fukushima, Japan
| | - Nozomi Kashiwabara
- Department of Pediatrics, Fukushima Medical University School of Medicine, Hikarigaoka 1, Fukushima, Japan
| | - Kei Ogasawara
- Department of Pediatrics, Fukushima Medical University School of Medicine, Hikarigaoka 1, Fukushima, Japan
| | - Maki Sato
- Department of Pediatrics, Fukushima Medical University School of Medicine, Hikarigaoka 1, Fukushima, Japan
| | - Koichi Hashimoto
- Department of Pediatrics, Fukushima Medical University School of Medicine, Hikarigaoka 1, Fukushima, Japan
| | - Mitsuaki Hosoya
- Department of Pediatrics, Fukushima Medical University School of Medicine, Hikarigaoka 1, Fukushima, Japan
| |
Collapse
|
99
|
Pharmacodynamic Effects of Standard versus High Caffeine Doses in the Developing Brain of Neonatal Rats Exposed to Intermittent Hypoxia. Int J Mol Sci 2021; 22:ijms22073473. [PMID: 33801707 PMCID: PMC8037517 DOI: 10.3390/ijms22073473] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 03/11/2021] [Accepted: 03/23/2021] [Indexed: 02/06/2023] Open
Abstract
(1) Background: Caffeine citrate, at standard doses, is effective for reducing the incidence of apnea of prematurity (AOP) and may confer neuroprotection and decrease neonatal morbidities in extremely low gestational age neonates (ELGANs) requiring oxygen therapy. We tested the hypothesis that high-dose caffeine (HiC) has no adverse effects on the neonatal brain. (2) Methods: Newborn rat pups were randomized to room air (RA), hyperoxia (Hx) or neonatal intermittent hypoxia (IH), from birth (P0) to P14 during which they received intraperitoneal injections of LoC (20 mg/kg on P0; 5 mg/kg/day on P1-P14), HiC (80 mg/kg; 20 mg/kg), or equivalent volume saline. Blood gases, histopathology, myelin and neuronal integrity, and adenosine receptor reactivity were assessed. (3) Results: Caffeine treatment in Hx influenced blood gases more than treatment in neonatal IH. Exposure to neonatal IH resulted in hemorrhage and higher brain width, particularly in layer 2 of the cerebral cortex. Both caffeine doses increased brain width in RA, but layer 2 was increased only with HiC. HiC decreased oxidative stress more effectively than LoC, and both doses reduced apoptosis biomarkers. In RA, both caffeine doses improved myelination, but the effect was abolished in Hx and neonatal IH. Similarly, both doses inhibited adenosine 1A receptor in all oxygen environments, but adenosine 2A receptor was inhibited only in RA and Hx. (4) Conclusions: Caffeine, even at high doses, when administered in normoxia, can confer neuroprotection, evidenced by reductions in oxidative stress, hypermyelination, and increased Golgi bodies. However, varying oxygen environments, such as Hx or neonatal IH, may alter and modify pharmacodynamic actions of caffeine and may even override the benefits caffeine.
Collapse
|
100
|
Developmental programming of cardiovascular function: a translational perspective. Clin Sci (Lond) 2021; 134:3023-3046. [PMID: 33231619 DOI: 10.1042/cs20191210] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 11/07/2020] [Accepted: 11/10/2020] [Indexed: 12/11/2022]
Abstract
The developmental origins of health and disease (DOHaD) is a concept linking pre- and early postnatal exposures to environmental influences with long-term health outcomes and susceptibility to disease. It has provided a new perspective on the etiology and evolution of chronic disease risk, and as such is a classic example of a paradigm shift. What first emerged as the 'fetal origins of disease', the evolution of the DOHaD conceptual framework is a storied one in which preclinical studies played an important role. With its potential clinical applications of DOHaD, there is increasing desire to leverage this growing body of preclinical work to improve health outcomes in populations all over the world. In this review, we provide a perspective on the values and limitations of preclinical research, and the challenges that impede its translation. The review focuses largely on the developmental programming of cardiovascular function and begins with a brief discussion on the emergence of the 'Barker hypothesis', and its subsequent evolution into the more-encompassing DOHaD framework. We then discuss some fundamental pathophysiological processes by which developmental programming may occur, and attempt to define these as 'instigator' and 'effector' mechanisms, according to their role in early adversity. We conclude with a brief discussion of some notable challenges that hinder the translation of this preclinical work.
Collapse
|