1
|
Romero-Lopez M, Naik M, Hartman T, Anderson-Berry A, Thoene M. Evidence and unknowns for the relevancy of applying current parenteral nutrition support recommendations among infants born less than 750 g or younger than 25 weeks' gestation: A narrative review. JPEN J Parenter Enteral Nutr 2025. [PMID: 40235365 DOI: 10.1002/jpen.2761] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Revised: 03/04/2025] [Accepted: 03/24/2025] [Indexed: 04/17/2025]
Abstract
With advancements in neonatal care, the viability of extremely low-birth-weight (ELBW) infants, especially those born extremely preterm, is increasing. However, specific recommendations for managing parenteral nutrition (PN) support in nanopreterm infants (<750 g or <25 weeks' gestation) are lacking. We aim to evaluate current recommendations and highlight considerations for applying them to nanopreterm infants. The author team used English-language articles related to nutrition in ELBW with emphasis on nanopreterm infants, along with studies on fetal growth and metabolism. Current PN support recommendations for ELBW infants may not suit nanopreterm infants due to physiological and developmental differences. Key considerations for nanopreterm infants include: Carbohydrate: They require immediate dextrose provision with low glucose infusion rates because of limited glycogen stores, immature gluconeogenesis, and impaired glucose intolerance. Lipids: Although essential for energy storage and cell membrane integrity, the ability to metabolize them may be limited, requiring careful consideration of lipid injectable emulsion provision and dosing. Protein: Protein is crucial for growth and development. However, achieving euglycemia is essential for proper amino acid utilization, requiring a delicate balance of dextrose and protein provision. Energy: Because of lower muscle and tissue mass and immature metabolic capabilities, existing recommendations may overestimate their energy needs. Micronutrients: Exact micronutrient requirements are unknown during this specific period of fetal development. This review highlights the limitations of available PN support recommendations for nanopreterm infants. Further research is needed to establish precise guidelines that optimally meet their nutrition needs.
Collapse
Affiliation(s)
- Mar Romero-Lopez
- Department of Pediatrics, Division of Perinatal-Neonatal Medicine, University of Texas Health Science Center Houston, McGovern Medical School, Houston, Texas, USA
- Institute for Clinical Research and Learning Health Care, University of Texas Health Science Center Houston, McGovern Medical School, Houston, Texas, USA
| | - Mamta Naik
- Department of Pharmacy Services, Children's Memorial Hermann Hospital - Texas Medical Center, Houston, Texas, USA
| | - Teresa Hartman
- Education & Research Services, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Ann Anderson-Berry
- Department of Pediatrics, Division of Neonatology, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Melissa Thoene
- Department of Pediatrics, Division of Neonatology, University of Nebraska Medical Center, Omaha, Nebraska, USA
| |
Collapse
|
2
|
Nian S, Zeng Y, Heyden KE, Cagnone G, Yagi H, Boeck M, Lee D, Hirst V, Hua Z, Lee J, Wang C, Neilsen K, Joyal JS, Field MS, Fu Z. Folic Acid Supplementation Inhibits Proliferative Retinopathy of Prematurity. Biomolecules 2025; 15:309. [PMID: 40001612 PMCID: PMC11852370 DOI: 10.3390/biom15020309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 02/11/2025] [Accepted: 02/17/2025] [Indexed: 02/27/2025] Open
Abstract
BACKGROUND Retinopathy of prematurity (ROP) is the major cause of blindness in children. It is a biphasic disease with retinal vessel growth cessation and loss (Phase I) followed by uncontrolled retinal vessel growth (Phase II). Folate is an essential nutrient for fetal development and growth. Premature infants have a high risk for folate deficiency. However, the contribution of folate to ROP risk remains unknown. METHODS In mouse oxygen-induced retinopathy (OIR), the nursing dams were fed with a folic acid-deficient or control diet after delivery until the end of hyperoxia. Alternatively, pups received direct injection of either folic acid or vehicle during Phase I hyperoxia. Genes involved in the folate cycle and angiogenic responses were examined using real-time PCR. Total retinal folate levels were measured with the Lactobacillus casei assay. RESULTS Maternal folic acid deficiency in early life exacerbated pathological retinal vessel growth, while supplementation with folic acid suppressed it. Genes involved in the folate cycle were downregulated in Phase I OIR retinas and were highly expressed in Müller glia. Folic acid reduced pro-angiogenic signaling in cultured rat retinal Müller glia in vitro. CONCLUSIONS Appropriate supplementation of folic acid might be a new and safe treatment for ROP at an early stage.
Collapse
Affiliation(s)
- Shen Nian
- Department of Ophthalmology, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (S.N.); (Y.Z.); (H.Y.); (D.L.); (V.H.); (Z.H.); (C.W.); (K.N.)
- Department of Pathology, Xi’an Medical University, Xi’an 710021, China
| | - Yan Zeng
- Department of Ophthalmology, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (S.N.); (Y.Z.); (H.Y.); (D.L.); (V.H.); (Z.H.); (C.W.); (K.N.)
| | - Katarina E. Heyden
- Division of Nutritional Sciences, Cornell University, Ithaca, NY 14853, USA; (K.E.H.); (M.S.F.)
| | - Gaël Cagnone
- Centre Hospitalier Universitaire Sainte-Justine Research Center, Montreal, QC H3T 1C5, Canada (J.-S.J.)
| | - Hitomi Yagi
- Department of Ophthalmology, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (S.N.); (Y.Z.); (H.Y.); (D.L.); (V.H.); (Z.H.); (C.W.); (K.N.)
- Department of Ophthalmology, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Myriam Boeck
- Department of Ophthalmology, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (S.N.); (Y.Z.); (H.Y.); (D.L.); (V.H.); (Z.H.); (C.W.); (K.N.)
- Eye Center, Medical Center, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Deokho Lee
- Department of Ophthalmology, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (S.N.); (Y.Z.); (H.Y.); (D.L.); (V.H.); (Z.H.); (C.W.); (K.N.)
| | - Victoria Hirst
- Department of Ophthalmology, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (S.N.); (Y.Z.); (H.Y.); (D.L.); (V.H.); (Z.H.); (C.W.); (K.N.)
| | - Zhanqing Hua
- Department of Ophthalmology, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (S.N.); (Y.Z.); (H.Y.); (D.L.); (V.H.); (Z.H.); (C.W.); (K.N.)
| | - Jeff Lee
- Department of Ophthalmology, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (S.N.); (Y.Z.); (H.Y.); (D.L.); (V.H.); (Z.H.); (C.W.); (K.N.)
| | - Chaomei Wang
- Department of Ophthalmology, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (S.N.); (Y.Z.); (H.Y.); (D.L.); (V.H.); (Z.H.); (C.W.); (K.N.)
| | - Katherine Neilsen
- Department of Ophthalmology, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (S.N.); (Y.Z.); (H.Y.); (D.L.); (V.H.); (Z.H.); (C.W.); (K.N.)
| | - Jean-Sébastien Joyal
- Centre Hospitalier Universitaire Sainte-Justine Research Center, Montreal, QC H3T 1C5, Canada (J.-S.J.)
- Department of Pediatrics, Faculty of Medicine, Université de Montréal, Montreal, QC H3C 3J7, Canada
| | - Martha S. Field
- Division of Nutritional Sciences, Cornell University, Ithaca, NY 14853, USA; (K.E.H.); (M.S.F.)
| | - Zhongjie Fu
- Department of Ophthalmology, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (S.N.); (Y.Z.); (H.Y.); (D.L.); (V.H.); (Z.H.); (C.W.); (K.N.)
| |
Collapse
|
3
|
Sridharan K, Al Jufairi M. Unveiling the Interplay: Antioxidant Enzyme Polymorphisms and Oxidative Stress in Preterm Neonatal Renal and Hepatic Functions. Curr Drug Metab 2025; 25:605-612. [PMID: 39377382 DOI: 10.2174/0113892002328584240923095216] [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: 05/22/2024] [Revised: 09/03/2024] [Accepted: 09/10/2024] [Indexed: 10/09/2024]
Abstract
AIMS To explore the relationship between oxidative stress biomarkers and the occurrence of acute kidney injury (AKI) alongside notable liver function disturbances in preterm neonates. BACKGROUND Given the immaturity of kidneys and incomplete liver development in preterm neonates, oxidative stress poses a considerable threat to their renal and hepatic health. OBJECTIVE To find out the association between various oxidative stress biomarkers and polymorphisms of antioxidant enzymes with renal and live functions. METHODS In this cross-sectional study, we gathered umbilical cord blood and peripheral blood samples for assessing oxidative stress biomarkers and identifying single nucleotide polymorphisms (SNPs) in antioxidant enzymes. Utilizing enzyme-linked immunosorbent assay kits, we quantified these oxidative stress biomarkers. Receiver-operating characteristics curve analysis was employed to ascertain the predictive capacity of these biomarkers, denoted by the area-under-the-curve (AUC). RESULTS Our findings revealed that umbilical cord heat-shock proteins emerged as robust predictors of neonatal AKI (AUC: 0.92; 95% CI: 0.8-1) with a defined cut-off concentration of 1.8 ng/mL. Likewise, umbilical cord 8-hydroxy-2-deoxy guanosine demonstrated significant predictability for liver function alterations (AUC: 0.7; 95% CI: 0.6-0.9) at a cut-off concentration of 2487.6 pg/mL. CONCLUSIONS We observed significant associations between SNPs in endothelial nitric oxide synthase and catalase with both AKI and impaired liver functions. Prospective studies are warranted to validate these findings, with a particular focus on exploring potential antioxidant interventions aimed at mitigating AKI and liver function abnormalities.
Collapse
Affiliation(s)
- Kannan Sridharan
- Department of Pharmacology & Therapeutics, College of Medicine and Medical Sciences, Arabian Gulf University, Manama, Kingdom of Bahrain
| | - Mona Al Jufairi
- Neonatal Intensive Care Unit, Department of Pediatrics, Salmaniya Medical Complex, Manama, Kingdom of Bahrain
- Department of Pediatrics, College of Medicine and Medical Sciences, Arabian Gulf University, Manama, Kingdom of Bahrain
| |
Collapse
|
4
|
Shvetsova AA, Shateeva VS, Khlystova MA, Makukha YA, Tarasova OS, Gaynullina DK. NADPH oxidase derived ROS promote arterial contraction in early postnatal rats by activation of L-type voltage-gated Ca 2+ channels. Free Radic Res 2025; 59:49-60. [PMID: 39879138 DOI: 10.1080/10715762.2024.2448483] [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: 09/02/2024] [Revised: 12/04/2024] [Accepted: 12/26/2024] [Indexed: 01/31/2025]
Abstract
Reactive oxygen species (ROS) produced by NADPH oxidase promote contraction of peripheral arteries, which is especially pronounced in early postnatal period in comparison to adulthood, but the mechanisms of such vasomotor influence are poorly understood. We tested the hypothesis that Rho-kinase and protein kinase C (PKC) mediate procontractile influence of NADPH oxidase derived ROS in peripheral artery of early postnatal rats. In addition, we evaluated the involvement Src-kinase and L-type voltage-gated Ca2+ channels (LTCC) into procontractile influence of ROS, produced by NADPH oxidase, because of their known interplay with Rho-kinase and PKC pathways. Saphenous arteries from 11- to 15-day-old male rats were studied using quantitative PCR, isometric myography and lucigenin-enhanced chemiluminescence. Arterial tissue of early postnatal rats contained Nox2, Nox4, Duox1 and Duox2 mRNAs, among which Nox2 mRNA was the most abundant. Pan-NADPH oxidase inhibitor VAS2870 (10 µM) significantly reduced arterial contractile responses to methoxamine. The inhibitors of Rho-kinase (Y27632, 3 µM), PKC (GF109203X, 10 µM) and Src-kinase (PP2, 10 µM), as well as LTCC blockers (nimodipine, 0.1 µM, and verapamil, 0.1 μM) also reduced methoxamine-induced contraction. Importantly, the effect of VAS2870 persisted in the presence of Rho-kinase, PKC or Src-kinase inhibitors, but not in the presence of LTCC blocker. Notably, the blockade of LTCC did not affect either basal or NADPH-induced O2•- production. Our data show that LTCC, but not Rho-kinase, PKC or Src-kinase are involved into procontractile effect of ROS, produced by NADPH oxidase, in saphenous artery of young rats. Сalcium influx through LTCC does not activate ROS production by NADPH oxidase.
Collapse
Affiliation(s)
- Anastasia A Shvetsova
- Department of Human and Animal Physiology, Faculty of Biology, M.V. Lomonosov Moscow State University, Moscow, Russia
| | - Valentina S Shateeva
- Department of Human and Animal Physiology, Faculty of Biology, M.V. Lomonosov Moscow State University, Moscow, Russia
| | - Margarita A Khlystova
- Department of Human and Animal Physiology, Faculty of Biology, M.V. Lomonosov Moscow State University, Moscow, Russia
| | - Yulia A Makukha
- Department of Human and Animal Physiology, Faculty of Biology, M.V. Lomonosov Moscow State University, Moscow, Russia
| | - Olga S Tarasova
- Department of Human and Animal Physiology, Faculty of Biology, M.V. Lomonosov Moscow State University, Moscow, Russia
- Department of Physiology and Pathology, Faculty of Basic Medicine, M.V. Lomonosov Moscow State University, Moscow, Russia
| | - Dina K Gaynullina
- Department of Human and Animal Physiology, Faculty of Biology, M.V. Lomonosov Moscow State University, Moscow, Russia
| |
Collapse
|
5
|
Shvetsova AA, Khlystova MA, Makukha YA, Shateeva VS, Borzykh AA, Gaynullina DK, Tarasova OS. Reactive oxygen species augment contractile responses of saphenous artery in 10-15-day-old but not adult rats: Substantial role of NADPH oxidases. Free Radic Biol Med 2024; 216:24-32. [PMID: 38460742 DOI: 10.1016/j.freeradbiomed.2024.03.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 03/02/2024] [Accepted: 03/07/2024] [Indexed: 03/11/2024]
Abstract
Reactive oxygen species (ROS) produced by NADPH oxidases (NOX, a key source of ROS in vascular cells) are involved in the regulation of vascular tone, but this has been explored mainly for adult organisms. Importantly, the mechanisms of vascular tone regulation differ significantly in early postnatal ontogenesis and adulthood, while the vasomotor role of ROS in immature systemic arteries is poorly understood. We tested the hypothesis that the functional contribution of NADPH oxidase-derived ROS to the regulation of peripheral arterial tone is higher in the early postnatal period than in adulthood. We studied saphenous arteries from 10- to 15-day-old ("young") and 3- to 4-month-old ("adult") male rats using lucigenin-enhanced chemiluminescence, quantitative PCR, Western blotting, and isometric myography. We demonstrated that both basal and NADPH-stimulated superoxide anion radical (O2•-) production was significantly higher in the arteries from young in comparison to adult rats. Importantly, pan-inhibitor of NADPH oxidase VAS2870 (10 μM) reduced NADPH-induced O2•- production in arteries of young rats. Saphenous arteries of both young and adult rats demonstrated high levels of Nox2 and Nox4 mRNAs, while Nox1 and Nox3 mRNAs were not detected. The protein contents of NOX2 and NOX4 were significantly higher in arterial tissue of young compared to adult animals. Moreover, VAS2870 (10 μM) had no effect on methoxamine-induced contractile responses of adult arteries but decreased them significantly in young arteries; such effect of VAS2870 persisted after removal of the endothelium. Finally, NOX2 inhibitor GSK2795039 (10 μM), but not NOX1/4 inhibitor GKT137831 (10 μM) weakened methoxamine-induced contractile responses of arteries from young rats. Thus, ROS produced by NOX2 have a pronounced contractile influence in saphenous artery smooth muscle cells of young, but not adult rats, which is associated with the increased vascular content of NOX2 protein at this age.
Collapse
Affiliation(s)
- Anastasia A Shvetsova
- Department of Human and Animal Physiology, Faculty of Biology, M.V. Lomonosov Moscow State University, 119234, Moscow, Russia.
| | - Margarita A Khlystova
- Department of Human and Animal Physiology, Faculty of Biology, M.V. Lomonosov Moscow State University, 119234, Moscow, Russia
| | - Yulia A Makukha
- Department of Human and Animal Physiology, Faculty of Biology, M.V. Lomonosov Moscow State University, 119234, Moscow, Russia
| | - Valentina S Shateeva
- Department of Human and Animal Physiology, Faculty of Biology, M.V. Lomonosov Moscow State University, 119234, Moscow, Russia
| | - Anna A Borzykh
- Laboratory of Exercise Physiology, State Research Center of the Russian Federation-Institute of Biomedical Problems, Russian Academy of Sciences, 123007, Moscow, Russia
| | - Dina K Gaynullina
- Department of Human and Animal Physiology, Faculty of Biology, M.V. Lomonosov Moscow State University, 119234, Moscow, Russia; Department of Physiology, Russian National Research Medical University, 117997, Moscow, Russia
| | - Olga S Tarasova
- Department of Human and Animal Physiology, Faculty of Biology, M.V. Lomonosov Moscow State University, 119234, Moscow, Russia; Laboratory of Exercise Physiology, State Research Center of the Russian Federation-Institute of Biomedical Problems, Russian Academy of Sciences, 123007, Moscow, Russia
| |
Collapse
|
6
|
Zheng S, Ye L. Hemodynamic Melody of Postnatal Cardiac and Pulmonary Development in Children with Congenital Heart Diseases. BIOLOGY 2024; 13:234. [PMID: 38666846 PMCID: PMC11048247 DOI: 10.3390/biology13040234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 03/20/2024] [Accepted: 03/25/2024] [Indexed: 04/28/2024]
Abstract
Hemodynamics is the eternal theme of the circulatory system. Abnormal hemodynamics and cardiac and pulmonary development intertwine to form the most important features of children with congenital heart diseases (CHDs), thus determining these children's long-term quality of life. Here, we review the varieties of hemodynamic abnormalities that exist in children with CHDs, the recently developed neonatal rodent models of CHDs, and the inspirations these models have brought us in the areas of cardiomyocyte proliferation and maturation, as well as in alveolar development. Furthermore, current limitations, future directions, and clinical decision making based on these inspirations are highlighted. Understanding how CHD-associated hemodynamic scenarios shape postnatal heart and lung development may provide a novel path to improving the long-term quality of life of children with CHDs, transplantation of stem cell-derived cardiomyocytes, and cardiac regeneration.
Collapse
Affiliation(s)
- Sixie Zheng
- Department of Thoracic and Cardiovascular Surgery, Shanghai Children’s Medical Center, School of Medicine, Shanghai Jiao Tong University, National Children’s Medical Center, Shanghai 200127, China;
- Shanghai Institute for Pediatric Congenital Heart Disease, Shanghai Children’s Medical Center, School of Medicine, Shanghai Jiao Tong University, National Children’s Medical Center, Shanghai 200127, China
| | - Lincai Ye
- Department of Thoracic and Cardiovascular Surgery, Shanghai Children’s Medical Center, School of Medicine, Shanghai Jiao Tong University, National Children’s Medical Center, Shanghai 200127, China;
- Shanghai Institute for Pediatric Congenital Heart Disease, Shanghai Children’s Medical Center, School of Medicine, Shanghai Jiao Tong University, National Children’s Medical Center, Shanghai 200127, China
| |
Collapse
|
7
|
Zhao T, Chang X, Biswas SK, Balsbaugh JL, Liddle J, Chen MH, Matson AP, Alder NN, Cong X. Pain/Stress, Mitochondrial Dysfunction, and Neurodevelopment in Preterm Infants. Dev Neurosci 2024; 46:341-352. [PMID: 38286121 PMCID: PMC11284246 DOI: 10.1159/000536509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Accepted: 01/10/2024] [Indexed: 01/31/2024] Open
Abstract
INTRODUCTION Preterm infants experience tremendous early life pain/stress during their neonatal intensive care unit (NICU) hospitalization, which impacts their neurodevelopmental outcomes. Mitochondrial function/dysfunction may interface between perinatal stress events and neurodevelopment. Nevertheless, the specific proteins or pathways linking mitochondrial functions to pain-induced neurodevelopmental outcomes in infants remain unidentified. Our study aims to investigate the associations among pain/stress, proteins associated with mitochondrial function/dysfunction, and neurobehavioral responses in preterm infants. METHODS We conducted a prospective cohort study, enrolling 33 preterm infants between September 2017 and July 2022 at two affiliated NICUs located in Hartford and Farmington, CT. NICU Network Neurobehavioral Scale (NNNS) datasets were evaluated to explore potential association with neurobehavioral outcomes. The daily pain/stress experienced by infant's during their NICU stay was documented. At 36-38 weeks post-menstrual age (PMA), neurobehavioral outcomes were evaluated using the NNNS and buccal swabs were collected for further analysis. Mass spectrometry-based proteomics was conducted on epithelial cells obtained from buccal swabs to evaluate protein expression level. Lasso statistical methods were conducted to study the association between protein abundance and infants' NNNS summary scores. Multiple linear regression and Gene Ontology (GO) enrichment analyses were performed to examine how clinical characteristics and neurodevelopmental outcomes may be associated with protein levels and underlying molecular pathways. RESULTS During NICU hospitalization, preterm premature rupture of membrane (PPROM) was negatively associated with neurobehavioral outcomes. The protein functions including leptin receptor binding activity, glutathione disulfide oxidoreductase activity and response to oxidative stress, lipid metabolism, and phosphate and proton transmembrane transporter activity were negatively associated with neurobehavioral outcomes; in contrast, cytoskeletal regulation, epithelial barrier, and protection function were found to be associated with the optimal neurodevelopmental outcomes. In addition, mitochondrial function-associated proteins including SPRR2A, PAIP1, S100A3, MT-CO2, PiC, GLRX, PHB2, and BNIPL-2 demonstrated positive association with favorable neurodevelopmental outcomes, while proteins of ABLIM1, UNC45A, keratins, MUC1, and CYB5B showed positive association with adverse neurodevelopmental outcomes. CONCLUSION Mitochondrial function-related proteins were observed to be associated with early life pain/stress and neurodevelopmental outcomes in infants. Large-scale studies with longitudinal datasets are warranted. Buccal proteins could be used to predict potential neurobehavioral outcomes.
Collapse
Affiliation(s)
- Tingting Zhao
- School of Nursing, Yale University, Orange, Connecticut, USA,
| | - Xiaolin Chang
- Department of Statistics, University of Connecticut, Storrs, Connecticut, USA
| | - Subrata Kumar Biswas
- Department of Molecular and Cell Biology, University of Connecticut, Storrs, Connecticut, USA
| | - Jeremy L Balsbaugh
- Proteomics and Metabolomics Facility, University of Connecticut, Storrs, Connecticut, USA
| | - Jennifer Liddle
- Proteomics and Metabolomics Facility, University of Connecticut, Storrs, Connecticut, USA
| | - Ming-Hui Chen
- Department of Statistics, University of Connecticut, Storrs, Connecticut, USA
| | - Adam P Matson
- Division of Neonatology, Connecticut Children's Medical Center, Hartford, Connecticut, USA
- Department of Pediatrics, University of Connecticut School of Medicine, Farmington, Connecticut, USA
| | - Nathan N Alder
- Department of Molecular and Cell Biology, University of Connecticut, Storrs, Connecticut, USA
| | - Xiaomei Cong
- School of Nursing, Yale University, Orange, Connecticut, USA
| |
Collapse
|
8
|
Zhang L, Buonfiglio F, Fieß A, Pfeiffer N, Gericke A. Retinopathy of Prematurity-Targeting Hypoxic and Redox Signaling Pathways. Antioxidants (Basel) 2024; 13:148. [PMID: 38397746 PMCID: PMC10885953 DOI: 10.3390/antiox13020148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Revised: 01/19/2024] [Accepted: 01/21/2024] [Indexed: 02/25/2024] Open
Abstract
Retinopathy of prematurity (ROP) is a proliferative vascular ailment affecting the retina. It is the main risk factor for visual impairment and blindness in infants and young children worldwide. If left undiagnosed and untreated, it can progress to retinal detachment and severe visual impairment. Geographical variations in ROP epidemiology have emerged over recent decades, attributable to differing levels of care provided to preterm infants across countries and regions. Our understanding of the causes of ROP, screening, diagnosis, treatment, and associated risk factors continues to advance. This review article aims to present the pathophysiological mechanisms of ROP, including its treatment. Specifically, it delves into the latest cutting-edge treatment approaches targeting hypoxia and redox signaling pathways for this condition.
Collapse
Affiliation(s)
| | | | | | | | - Adrian Gericke
- Department of Ophthalmology, University Medical Center, Johannes Gutenberg University Mainz, Langenbeckstrasse 1, 55131 Mainz, Germany; (L.Z.); (F.B.); (A.F.); (N.P.)
| |
Collapse
|
9
|
Seki D, Kirkegaard R, Osvatic J, Hausmann B, Séneca J, Pjevac P, Berger A, J Hall L, Wisgrill L, Berry D. Gut microbiota genome features associated with brain injury in extremely premature infants. Gut Microbes 2024; 16:2410479. [PMID: 39679616 DOI: 10.1080/19490976.2024.2410479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 08/19/2024] [Accepted: 09/24/2024] [Indexed: 12/17/2024] Open
Abstract
Severe brain damage is common among premature infants, and the gut microbiota has been implicated in its pathology. Although the order of colonizing bacteria is well described, the mechanisms underlying aberrant assembly of the gut microbiota remain elusive. Here, we employed long-read nanopore sequencing to assess abundances of microbial species and their functional genomic potential in stool samples from a cohort of 30 extremely premature infants. We identify several key microbial traits significantly associated with severe brain damage, such as the genomic potential for nitrate respiration and iron scavenging. Members of the Enterobacteriaceae were prevalent across the cohort and displayed a versatile metabolic potential, including pathogenic and nonpathogenic traits. Predominance of Enterobacter hormaechei and Klebsiella pneumoniae were associated with an overall loss of genomic functional redundancy as well as poor neurophysiological outcome. These findings reveal microbial traits that may be involved in exacerbating brain injury in extremely premature infants and provide suitable targets for therapeutic interventions.
Collapse
Affiliation(s)
- David Seki
- Centre for Microbiology and Environmental Systems Science, Department of Microbiology and Ecosystem Science, Division of Microbial Ecology, University of Vienna, Vienna, Austria
- Joint Microbiome Facility of the Medical University of Vienna and the University of Vienna, Vienna, Austria
- Intestinal Microbiome, School of Life Sciences, ZIEL - Institute for Food & Health, Technical University of Munich, Freising, Germany
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Rasmus Kirkegaard
- Centre for Microbiology and Environmental Systems Science, Department of Microbiology and Ecosystem Science, Division of Microbial Ecology, University of Vienna, Vienna, Austria
- Joint Microbiome Facility of the Medical University of Vienna and the University of Vienna, Vienna, Austria
| | - Jay Osvatic
- Joint Microbiome Facility of the Medical University of Vienna and the University of Vienna, Vienna, Austria
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Bela Hausmann
- Joint Microbiome Facility of the Medical University of Vienna and the University of Vienna, Vienna, Austria
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Joana Séneca
- Centre for Microbiology and Environmental Systems Science, Department of Microbiology and Ecosystem Science, Division of Microbial Ecology, University of Vienna, Vienna, Austria
- Joint Microbiome Facility of the Medical University of Vienna and the University of Vienna, Vienna, Austria
| | - Petra Pjevac
- Centre for Microbiology and Environmental Systems Science, Department of Microbiology and Ecosystem Science, Division of Microbial Ecology, University of Vienna, Vienna, Austria
- Joint Microbiome Facility of the Medical University of Vienna and the University of Vienna, Vienna, Austria
| | - Angelika Berger
- Department of Pediatrics and Adolescent Medicine, Division of Neonatology, Pediatric Intensive Care and Neuropediatrics, Comprehensive Center for Pediatrics. Medical University of Vienna, Vienna, Austria
| | - Lindsay J Hall
- Intestinal Microbiome, School of Life Sciences, ZIEL - Institute for Food & Health, Technical University of Munich, Freising, Germany
- Food Microbiomes and Health, Quadram Institute Bioscience, Norwich, UK
- Norwich Medical School, University of East Anglia, Norwich, UK
- Microbes, Infections, and Microbiomes, Institute of Microbiology and Infection, University of Birmingham, Birmingham, UK
| | - Lukas Wisgrill
- Department of Pediatrics and Adolescent Medicine, Division of Neonatology, Pediatric Intensive Care and Neuropediatrics, Comprehensive Center for Pediatrics. Medical University of Vienna, Vienna, Austria
| | - David Berry
- Centre for Microbiology and Environmental Systems Science, Department of Microbiology and Ecosystem Science, Division of Microbial Ecology, University of Vienna, Vienna, Austria
- Intestinal Microbiome, School of Life Sciences, ZIEL - Institute for Food & Health, Technical University of Munich, Freising, Germany
| |
Collapse
|
10
|
Demirtas MS, Kilicbay F, Erdal H, Tunc G. Oxidative Stress Levels and Dynamic Thiol-Disulfide Balance in Preterm Newborns with Bronchopulmonary Dysplasia. Lab Med 2023; 54:587-592. [PMID: 36896684 DOI: 10.1093/labmed/lmad010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/11/2023] Open
Abstract
OBJECTIVE The aim of this study was to assess the oxidative stress (OS) levels and dynamic thiol-disulfide balance in preterm newborns with bronchopulmonary dysplasia (BPD). METHODS This prospective study included newborns separated into 2 groups, those with BPD (case) or without BPD (control). The 2 groups were compared by clinical and laboratory findings. The OS parameters total oxidant status (TOS), total antioxidant status (TAS), OS index (OSI), native thiol (NT), and total thiol were measured within the first day after birth. Oxygen requirements were measured using the fraction of inspired oxygen (FIO2) recorded in the first hour after birth/admission and the average FIO2 within 28 days of the birth. RESULTS Infants diagnosed with BPD had a significantly lower gestational age and birth weight and a lower 5-min Apgar score (P < .05). Infants with BPD also had a higher rate of respiratory distress syndrome, rate of use of surfactant therapy, duration of ventilation therapy, and duration of hospital stay compared with control (P = .001, P = .001, P = .001, and P = .001, respectively). Plasma TAS and NT levels of newborns with BPD were significantly lower than newborns without BPD (P < .05). In the BPD group, plasma TOS and OSI levels were significantly higher than in the control group. CONCLUSION We found that OS was increased in newborns with BPD. The clinical significance of this study will provide the clinician with a different perspective on BPD by determining the dynamic thiol disulfide balance.
Collapse
Affiliation(s)
| | - Fatih Kilicbay
- Division of Neonatology, Department of Pediatrics, Sivas Cumhuriyet University Faculty of Medicine, Sivas, Turkey
| | - Huseyin Erdal
- Department of Medical Genetics, Aksaray University Faculty of Medicine, Aksaray, Turkey
| | - Gaffari Tunc
- Division of Neonatology, Department of Pediatrics, Sivas Cumhuriyet University Faculty of Medicine, Sivas, Turkey
| |
Collapse
|
11
|
Bousquet A, Sanderson K, O’Shea TM, Fry RC. Accelerated Aging and the Life Course of Individuals Born Preterm. CHILDREN (BASEL, SWITZERLAND) 2023; 10:1683. [PMID: 37892346 PMCID: PMC10605448 DOI: 10.3390/children10101683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/02/2023] [Revised: 09/29/2023] [Accepted: 10/11/2023] [Indexed: 10/29/2023]
Abstract
Individuals born preterm have shorter lifespans and elevated rates of chronic illness that contribute to mortality risk when compared to individuals born at term. Emerging evidence suggests that individuals born preterm or of low birthweight also exhibit physiologic and cellular biomarkers of accelerated aging. It is unclear whether, and to what extent, accelerated aging contributes to a higher risk of chronic illness and mortality among individuals born preterm. Here, we review accelerated aging phenotypes in adults born preterm and biological pathways that appear to contribute to accelerated aging. We highlight biomarkers of accelerated aging and various resiliency factors, including both pharmacologic and non-pharmacologic factors, that might buffer the propensity for accelerated aging among individuals born preterm.
Collapse
Affiliation(s)
- Audrey Bousquet
- Department of Environmental Sciences and Engineering, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC 27599, USA; (A.B.); (R.C.F.)
| | - Keia Sanderson
- Department of Internal Medicine, School of Medicine, University of North Carolina, Chapel Hill, NC 27599, USA;
| | - T. Michael O’Shea
- Department of Pediatrics, School of Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Rebecca C. Fry
- Department of Environmental Sciences and Engineering, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC 27599, USA; (A.B.); (R.C.F.)
| |
Collapse
|
12
|
Kaltsas A, Zikopoulos A, Moustakli E, Zachariou A, Tsirka G, Tsiampali C, Palapela N, Sofikitis N, Dimitriadis F. The Silent Threat to Women's Fertility: Uncovering the Devastating Effects of Oxidative Stress. Antioxidants (Basel) 2023; 12:1490. [PMID: 37627485 PMCID: PMC10451552 DOI: 10.3390/antiox12081490] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 07/18/2023] [Accepted: 07/24/2023] [Indexed: 08/27/2023] Open
Abstract
Oxidative stress (OS), which arises through an imbalance between the formation of reactive oxygen species (ROS) and antioxidant defenses, plays a key role in the pathophysiology of female infertility, with the latter constituting just one of a number of diseases linked to OS as a potential cause. The aim of the present article is to review the literature regarding the association between OS and female infertility. Among the reproductive diseases considered are endometriosis and polycystic ovary syndrome (PCOS), while environmental pollutants, lifestyle variables, and underlying medical conditions possibly resulting in OS are additionally examined. Current evidence points to OS likely contributing to the pathophysiology of the above reproductive disorders, with the amount of damage done by OS being influenced by such variables as duration and severity of exposure and the individual's age and genetic predisposition. Also discussed are the processes via which OS may affect female fertility, these including DNA damage and mitochondrial dysfunction. Finally, the last section of the manuscript contains an evaluation of treatment options, including antioxidants and lifestyle modification, capable of minimizing OS in infertile women. The prime message underlined by this review is the importance of considering OS in the diagnosis and treatment of female infertility. Further studies are, nevertheless required to identify the best treatment regimen and its ideal duration.
Collapse
Affiliation(s)
- Aris Kaltsas
- Department of Urology, Faculty of Medicine, School of Health Sciences, University of Ioannina, 45110 Ioannina, Greece; (A.K.); (A.Z.); (A.Z.); (N.S.)
| | - Athanasios Zikopoulos
- Department of Urology, Faculty of Medicine, School of Health Sciences, University of Ioannina, 45110 Ioannina, Greece; (A.K.); (A.Z.); (A.Z.); (N.S.)
| | - Efthalia Moustakli
- Laboratory of Medical Genetics in Clinical Practice, Faculty of Medicine, School of Health Sciences, University of Ioannina, 45110 Ioannina, Greece; (E.M.); (G.T.)
| | - Athanasios Zachariou
- Department of Urology, Faculty of Medicine, School of Health Sciences, University of Ioannina, 45110 Ioannina, Greece; (A.K.); (A.Z.); (A.Z.); (N.S.)
| | - Georgia Tsirka
- Laboratory of Medical Genetics in Clinical Practice, Faculty of Medicine, School of Health Sciences, University of Ioannina, 45110 Ioannina, Greece; (E.M.); (G.T.)
| | | | - Natalia Palapela
- Medical Faculty, Medical University of Sofia, 1431 Sofia, Bulgaria;
| | - Nikolaos Sofikitis
- Department of Urology, Faculty of Medicine, School of Health Sciences, University of Ioannina, 45110 Ioannina, Greece; (A.K.); (A.Z.); (A.Z.); (N.S.)
| | - Fotios Dimitriadis
- Department of Urology, Faculty of Medicine, School of Health Sciences, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| |
Collapse
|
13
|
Zhang EY, Bartman CM, Prakash YS, Pabelick CM, Vogel ER. Oxygen and mechanical stretch in the developing lung: risk factors for neonatal and pediatric lung disease. Front Med (Lausanne) 2023; 10:1214108. [PMID: 37404808 PMCID: PMC10315587 DOI: 10.3389/fmed.2023.1214108] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Accepted: 06/01/2023] [Indexed: 07/06/2023] Open
Abstract
Chronic airway diseases, such as wheezing and asthma, remain significant sources of morbidity and mortality in the pediatric population. This is especially true for preterm infants who are impacted both by immature pulmonary development as well as disproportionate exposure to perinatal insults that may increase the risk of developing airway disease. Chronic pediatric airway disease is characterized by alterations in airway structure (remodeling) and function (increased airway hyperresponsiveness), similar to adult asthma. One of the most common perinatal risk factors for development of airway disease is respiratory support in the form of supplemental oxygen, mechanical ventilation, and/or CPAP. While clinical practice currently seeks to minimize oxygen exposure to decrease the risk of bronchopulmonary dysplasia (BPD), there is mounting evidence that lower levels of oxygen may carry risk for development of chronic airway, rather than alveolar disease. In addition, stretch exposure due to mechanical ventilation or CPAP may also play a role in development of chronic airway disease. Here, we summarize the current knowledge of the impact of perinatal oxygen and mechanical respiratory support on the development of chronic pediatric lung disease, with particular focus on pediatric airway disease. We further highlight mechanisms that could be explored as potential targets for novel therapies in the pediatric population.
Collapse
Affiliation(s)
- Emily Y. Zhang
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, MN, United States
| | - Colleen M. Bartman
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, MN, United States
| | - Y. S. Prakash
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, MN, United States
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, United States
| | - Christina M. Pabelick
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, MN, United States
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, United States
| | - Elizabeth R. Vogel
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, MN, United States
| |
Collapse
|
14
|
Li DB, Xu XX, Hu YQ, Cui Q, Xiao YY, Sun SJ, Chen LJ, Ye LC, Sun Q. Congenital heart disease-associated pulmonary dysplasia and its underlying mechanisms. Am J Physiol Lung Cell Mol Physiol 2023; 324:L89-L101. [PMID: 36472329 PMCID: PMC9925164 DOI: 10.1152/ajplung.00195.2022] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Clinical observation indicates that exercise capacity, an important determinant of survival in patients with congenital heart disease (CHD), is most decreased in children with reduced pulmonary blood flow (RPF). However, the underlying mechanism remains unclear. Here, we obtained human RPF lung samples from children with tetralogy of Fallot as well as piglet and rat RPF lung samples from animals with pulmonary artery banding surgery. We observed impaired alveolarization and vascularization, the main characteristics of pulmonary dysplasia, in the lungs of RPF infants, piglets, and rats. RPF caused smaller lungs, cyanosis, and body weight loss in neonatal rats and reduced the number of alveolar type 2 cells. RNA sequencing demonstrated that RPF induced the downregulation of metabolism and migration, a key biological process of late alveolar development, and the upregulation of immune response, which was confirmed by flow cytometry and cytokine detection. In addition, the immunosuppressant cyclosporine A rescued pulmonary dysplasia and increased the expression of the Wnt signaling pathway, which is the driver of postnatal lung development. We concluded that RPF results in pulmonary dysplasia, which may account for the reduced exercise capacity of patients with CHD with RPF. The underlying mechanism is associated with immune response activation, and immunosuppressants have a therapeutic effect in CHD-associated pulmonary dysplasia.
Collapse
Affiliation(s)
- De-Bao Li
- 1Department of Thoracic and Cardiovascular Surgery, Shanghai Children’s Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, People’s Republic of China
| | - Xiu-Xia Xu
- 4Department of Radiology, Huangpu Branch, Shanghai Ninth People’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, People’s Republic of China
| | - Yu-Qing Hu
- 3Department of Cardiology, Shanghai Children’s Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, People’s Republic of China
| | - Qing Cui
- 3Department of Cardiology, Shanghai Children’s Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, People’s Republic of China
| | - Ying-Ying Xiao
- 1Department of Thoracic and Cardiovascular Surgery, Shanghai Children’s Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, People’s Republic of China
| | - Si-Juan Sun
- 5Department of Pediatric Intensive Care Unit, Shanghai Children’s Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, People’s Republic of China
| | - Li-Jun Chen
- 3Department of Cardiology, Shanghai Children’s Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, People’s Republic of China
| | - Lin-Cai Ye
- 2Department of Thoracic and Cardiovascular Surgery, Shanghai Institute for Pediatric Congenital Heart Disease, Institute of Pediatric Translational Medicine, Shanghai Children’s Medical Center, Shanghai School of Medicine, Shanghai Jiao Tong University, Shanghai, People’s Republic of China
| | - Qi Sun
- 1Department of Thoracic and Cardiovascular Surgery, Shanghai Children’s Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, People’s Republic of China
| |
Collapse
|
15
|
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
|
16
|
Mohammadi A, Higazy R, Gauda EB. PGC-1α activity and mitochondrial dysfunction in preterm infants. Front Physiol 2022; 13:997619. [PMID: 36225305 PMCID: PMC9548560 DOI: 10.3389/fphys.2022.997619] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Accepted: 09/09/2022] [Indexed: 11/26/2022] Open
Abstract
Extremely low gestational age neonates (ELGANs) are born in a relatively hyperoxic environment with weak antioxidant defenses, placing them at high risk for mitochondrial dysfunction affecting multiple organ systems including the nervous, respiratory, ocular, and gastrointestinal systems. The brain and lungs are highly affected by mitochondrial dysfunction and dysregulation in the neonate, causing white matter injury (WMI) and bronchopulmonary dysplasia (BPD), respectively. Adequate mitochondrial function is important in providing sufficient energy for organ development as it relates to alveolarization and axonal myelination and decreasing oxidative stress via reactive oxygen species (ROS) and reactive nitrogen species (RNS) detoxification. Peroxisome proliferator-activated receptor gamma coactivator-1 alpha (PGC-1α) is a master regulator of mitochondrial biogenesis and function. Since mitochondrial dysfunction is at the root of WMI and BPD pathobiology, exploring therapies that can regulate PGC-1α activity may be beneficial. This review article describes several promising therapeutic agents that can mitigate mitochondrial dysfunction through direct and indirect activation and upregulation of the PGC-1α pathway. Metformin, resveratrol, omega 3 fatty acids, montelukast, L-citrulline, and adiponectin are promising candidates that require further pre-clinical and clinical studies to understand their efficacy in decreasing the burden of disease from WMI and BPD in preterm infants.
Collapse
Affiliation(s)
- Atefeh Mohammadi
- The Hospital for Sick Children, Division of Neonatology, Department of Pediatrics and Translational Medicine Program, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Randa Higazy
- The Hospital for Sick Children, Division of Neonatology, Department of Pediatrics and Translational Medicine Program, Toronto, ON, Canada
| | - Estelle B. Gauda
- The Hospital for Sick Children, Division of Neonatology, Department of Pediatrics and Translational Medicine Program, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
17
|
Moliner-Calderón E, Verd S, Leiva A, Ponce-Taylor J, Ginovart G, Moll-McCarthy P, Gelabert C, Figueras-Aloy J. Human Milk Feeding for Septic Newborn Infants Might Minimize Their Exposure to Ventilation Therapy. CHILDREN (BASEL, SWITZERLAND) 2022; 9:children9101450. [PMID: 36291386 PMCID: PMC9600066 DOI: 10.3390/children9101450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Revised: 09/12/2022] [Accepted: 09/14/2022] [Indexed: 11/16/2022]
Abstract
Background. It has been well established that human milk feeding contributes to limiting lung diseases in vulnerable neonates. The primary aim of this study was to compare the need for mechanical ventilation between human milk-fed neonates with sepsis and formula-fed neonates with sepsis. Methods. All late preterm and full-term infants from a single center with sepsis findings from 2002 to 2017 were identified. Data on infant feeding during hospital admission were also recorded. Multivariate logistic regression analyses were performed to assess the impact of feeding type on ventilation support and main neonatal morbidities. Results. The total number of participants was 322 (human milk group = 260; exclusive formula group = 62). In the bivariate analysis, 72% of human milk-fed neonates did not require oxygen therapy or respiratory support versus 55% of their formula-fed counterparts (p < 0.0001). Accordingly, invasive mechanical ventilation was required in 9.2% of any human milk-fed infants versus 32% of their exclusively formula-fed counterparts (p = 0.0085). These results held true in multivariate analysis; indeed, any human milk-fed neonates were more likely to require less respiratory support (OR = 0.44; 95% CI:0.22, 0.89) than those who were exclusively formula-fed. Conclusion. Human milk feeding may minimize exposure to mechanical ventilation.
Collapse
Affiliation(s)
- Elisenda Moliner-Calderón
- Neonatal Unit, Department of Paediatrics, Santa Creu i Sant Pau Hospital, 90 Mas Casanovas Street, 08041 Barcelona, Spain
| | - Sergio Verd
- Pediatric Unit, La Vileta Surgery, Department of Primary Care, Matamusinos Street, 07013 Palma de Mallorca, Spain
- Balearic Islands Health Research Institute (IdISBa), 79 Valldemossa Road, 07120 Palma de Mallorca, Spain
- Correspondence: ; Tel.: +34-600-505-246; Fax: +34-(9)-71-799534
| | - Alfonso Leiva
- Research Unit, Department of Primary Care, Escola Graduada Street, 07002 Palma de Mallorca, Spain
| | - Jaume Ponce-Taylor
- A & E Unit, Department of Primary Care, Illes Balears Street, 07014 Palma de Mallorca, Spain
| | - Gemma Ginovart
- Neonatal Unit, Department of Paediatrics, Germans Trias i Pujol Hospital, Canyet Road, 08916 Badalona, Spain
| | - Pia Moll-McCarthy
- A & E Division, Manacor Hospital, Alcudia Road, 07500 Manacor, Spain
| | - Catian Gelabert
- Department of Paediatrics, Son Espases Hospital, 79 Valldemossa Road, 07120 Palma de Mallorca, Spain
| | - Josep Figueras-Aloy
- Neonatal Unit, ICGON, Clinic Hospital, Sabino Arana Street, 08028 Barcelona, Spain
| |
Collapse
|
18
|
Thoene M, Anderson-Berry A. Nutrition Support Practices for Infants Born <750 Grams or <25 Weeks Gestation: A Call for More Research. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:10957. [PMID: 36078670 PMCID: PMC9517820 DOI: 10.3390/ijerph191710957] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Accepted: 08/29/2022] [Indexed: 06/15/2023]
Abstract
With advances in medical care and efforts to care for continually smaller and younger preterm infants, the gestational age of viability has decreased, including as young as 21 or 22 weeks of gestation [...].
Collapse
|
19
|
Pritchard KA, Jing X, Teng M, Wells C, Jia S, Afolayan AJ, Jarzembowski J, Day BW, Naylor S, Hessner MJ, Konduri GG, Teng RJ. Role of endoplasmic reticulum stress in impaired neonatal lung growth and bronchopulmonary dysplasia. PLoS One 2022; 17:e0269564. [PMID: 36018859 PMCID: PMC9417039 DOI: 10.1371/journal.pone.0269564] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 05/24/2022] [Indexed: 11/18/2022] Open
Abstract
Myeloperoxidase (MPO), oxidative stress (OS), and endoplasmic reticulum (ER) stress are increased in the lungs of rat pups raised in hyperoxia, an established model of bronchopulmonary dysplasia (BPD). However, the relationship between OS, MPO, and ER stress has not been examined in hyperoxia rat pups. We treated Sprague-Dawley rat pups with tunicamycin or hyperoxia to determine this relationship. ER stress was detected using immunofluorescence, transcriptomic, proteomic, and electron microscopic analyses. Immunofluorescence observed increased ER stress in the lungs of hyperoxic rat BPD and human BPD. Proteomic and morphometric studies showed that tunicamycin directly increased ER stress of rat lungs and decreased lung complexity with a BPD phenotype. Previously, we showed that hyperoxia initiates a cycle of destruction that we hypothesized starts from increasing OS through MPO accumulation and then increases ER stress to cause BPD. To inhibit ER stress, we used tauroursodeoxycholic acid (TUDCA), a molecular chaperone. To break the cycle of destruction and reduce OS and MPO, we used N-acetyl-lysyltyrosylcysteine amide (KYC). The fact that TUDCA improved lung complexity in tunicamycin- and hyperoxia-treated rat pups supports the idea that ER stress plays a causal role in BPD. Additional support comes from data showing TUDCA decreased lung myeloid cells and MPO levels in the lungs of tunicamycin- and hyperoxia-treated rat pups. These data link OS and MPO to ER stress in the mechanisms mediating BPD. KYC's inhibition of ER stress in the tunicamycin-treated rat pup's lung provides additional support for the idea that MPO-induced ER stress plays a causal role in the BPD phenotype. ER stress appears to expand our proposed cycle of destruction. Our results suggest ER stress evolves from OS and MPO to increase neonatal lung injury and impair growth and development. The encouraging effect of TUDCA indicates that this compound has the potential for treating BPD.
Collapse
Affiliation(s)
- Kirkwood A. Pritchard
- Division of Pediatric Surgery, Department of Surgery, Medical College of Wisconsin, Wauwatosa, Wisconsin, United States of America
- Children’s Research Institute, Medical College of Wisconsin, Wauwatosa, Wisconsin, United States of America
| | - Xigang Jing
- Children’s Research Institute, Medical College of Wisconsin, Wauwatosa, Wisconsin, United States of America
- Department of Pediatrics, Medical College of Wisconsin, Wauwatosa, Wisconsin, United States of America
| | - Michelle Teng
- Department of Pediatrics, Medical College of Wisconsin, Wauwatosa, Wisconsin, United States of America
| | - Clive Wells
- Electron Microscope Facility, Medical College of Wisconsin, Wauwatosa, Wisconsin, United States of America
| | - Shuang Jia
- Department of Pediatrics, Medical College of Wisconsin, Wauwatosa, Wisconsin, United States of America
| | - Adeleye J. Afolayan
- Children’s Research Institute, Medical College of Wisconsin, Wauwatosa, Wisconsin, United States of America
- Department of Pediatrics, Medical College of Wisconsin, Wauwatosa, Wisconsin, United States of America
| | - Jason Jarzembowski
- Children’s Research Institute, Medical College of Wisconsin, Wauwatosa, Wisconsin, United States of America
- Division of Pediatric Pathology, Department of Pathology, Medical College of Wisconsin, Wauwatosa, Wisconsin, United States of America
| | - Billy W. Day
- ReNeuroGen L.L.C. Milwaukee, Elm Grove, Wisconsin, United States of America
| | - Stephen Naylor
- ReNeuroGen L.L.C. Milwaukee, Elm Grove, Wisconsin, United States of America
| | - Martin J. Hessner
- Children’s Research Institute, Medical College of Wisconsin, Wauwatosa, Wisconsin, United States of America
- Department of Pediatrics, Medical College of Wisconsin, Wauwatosa, Wisconsin, United States of America
| | - G. Ganesh Konduri
- Children’s Research Institute, Medical College of Wisconsin, Wauwatosa, Wisconsin, United States of America
- Department of Pediatrics, Medical College of Wisconsin, Wauwatosa, Wisconsin, United States of America
| | - Ru-Jeng Teng
- Children’s Research Institute, Medical College of Wisconsin, Wauwatosa, Wisconsin, United States of America
- Department of Pediatrics, Medical College of Wisconsin, Wauwatosa, Wisconsin, United States of America
| |
Collapse
|
20
|
Zhao T, Alder NN, Starkweather AR, Chen MH, Matson AP, Xu W, Balsbaugh JL, Cong X. Associations of Mitochondrial Function, Stress, and Neurodevelopmental Outcomes in Early Life: A Systematic Review. Dev Neurosci 2022; 44:438-454. [PMID: 35995037 PMCID: PMC9928905 DOI: 10.1159/000526491] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Accepted: 07/18/2022] [Indexed: 11/19/2022] Open
Abstract
Early life stress is commonly experienced by infants, especially preterm infants, and may impact their neurodevelopmental outcomes in their early and later lives. Mitochondrial function/dysfunction may play an important role underlying the linkage of prenatal and postnatal stress and neurodevelopmental outcomes in infants. This review aimed to provide insights on the relationship between early life stress and neurodevelopment and the mechanisms of mitochondrial function/dysfunction that contribute to the neuropathology of stress. The Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) statement was used to develop this systematic review. PubMed, Scopus, PsycINFO, and Biosis databases were searched for primary research articles published between 2010 and 2021 that examined the relationships among mitochondrial function/dysfunction, infant stress, and neurodevelopment. Thirty studies were identified. There is evidence to support that mitochondrial function/dysfunction mediates the relationship between prenatal and postnatal stress and neurodevelopmental outcomes in infants. Maternal transgenerational transmission of mitochondrial bioenergetic patterns influenced prenatal stress induced neurodevelopmental outcomes and behavioral changes in infants. Multiple functionally relevant mitochondrial proteins, genes, and polymorphisms were associated with stress exposure. This is the first review of the role that mitochondrial function/dysfunction plays in the association between stress and neurodevelopmental outcomes in full-term and preterm infants. Although multiple limitations were found based on the lack of data on the influence of biological sex, and due to invasive sampling, and lack of longitudinal data, many genes and proteins associated with mitochondrial function/dysfunction were found to influence neurodevelopmental outcomes in the early life of infants.
Collapse
Affiliation(s)
- Tingting Zhao
- School of Nursing, University of Connecticut, Storrs, Connecticut, USA,
| | - Nathan N Alder
- Department of Molecular and Cell Biology, University of Connecticut, Storrs, Connecticut, USA
| | | | - Ming-Hui Chen
- Department of Statistics, University of Connecticut, Storrs, Connecticut, USA
| | - Adam P Matson
- Division of Neonatology, Connecticut Children's Medical Center, Hartford, Connecticut, USA
- Department of Pediatrics, University of Connecticut School of Medicine, Farmington, Connecticut, USA
| | - Wanli Xu
- School of Nursing, University of Connecticut, Storrs, Connecticut, USA
| | - Jeremy L Balsbaugh
- Proteomics and Metabolomics Facility, University of Connecticut, Storrs, Connecticut, USA
| | - Xiaomei Cong
- School of Nursing, University of Connecticut, Storrs, Connecticut, USA
| |
Collapse
|
21
|
Ryu J. New Aspects on the Treatment of Retinopathy of Prematurity: Currently Available Therapies and Emerging Novel Therapeutics. Int J Mol Sci 2022; 23:8529. [PMID: 35955664 PMCID: PMC9369302 DOI: 10.3390/ijms23158529] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 07/27/2022] [Accepted: 07/29/2022] [Indexed: 02/05/2023] Open
Abstract
Retinopathy of prematurity (ROP) is a rare proliferative ocular disorder in preterm infants. Because of the advancements in neonatal care, the incidence of ROP has increased gradually. Now, ROP is one of the leading causes of blindness in children. Preterm infants with immature retinal development are exposed to supplemental oxygen inside an incubator until their cardiopulmonary system is adequately developed. Once they are returned to room air, the relatively low oxygen level stimulates various angiogenesis factors initiating retinal neovascularization. If patients with ROP are not offered adequate and timely treatment, they can experience vision loss that may ultimately lead to permanent blindness. Although laser therapy and anti-vascular endothelial growth factor agents are widely used to treat ROP, they have limitations. Thus, it is important to identify novel therapeutics with minimal adverse effects for the treatment of ROP. To date, various pharmacologic and non-pharmacologic therapies have been assessed as treatments for ROP. In this review, the major molecular factors involved in the pathogenesis of ROP, currently offered therapies, therapies under investigation, and emerging novel therapeutics of ROP are discussed.
Collapse
Affiliation(s)
- Juhee Ryu
- Vessel-Organ Interaction Research Center, College of Pharmacy, Kyungpook National University, Daegu 41566, Korea; ; Tel.: +82-539508583
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Kyungpook National University, Daegu 41566, Korea
| |
Collapse
|
22
|
Evaluation of pro-oxidant antioxidant balance in retinopathy of prematurity. Eye (Lond) 2022; 36:148-152. [PMID: 33649574 PMCID: PMC8727563 DOI: 10.1038/s41433-021-01465-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 01/06/2021] [Accepted: 02/09/2021] [Indexed: 02/06/2023] Open
Abstract
INTRODUCTION Retinopathy of prematurity (RoP) is a vasoproliferative disorder caused by the abnormal development of retinal vessels in premature neonates. It is one of the major causes of childhood blindness, which is increasing with the increasing survival rate of low birth weight and premature neonates. This study has aimed to evaluate the role of oxidative stress and peroxidant antioxidant balance (PAB) in the pathogenesis and prediction of RoP. MATERIALS AND METHODS A total of 154 neonates weighing <1500 g admitted at the NICU of Ghaem Hospital, Mashhad, Iran, were enrolled in this cross-sectional study between 2018 and 2020. Blood samples were collected on the first day of birth to assess the peroxidant and antioxidant balance. The demographic, prenatal, and clinical course and postnatal problems were also recorded. The neonates were examined for RoP and divided into control (healthy) and affected (ROP) groups. RESULTS There were significant differences between the two groups in gestational age, duration of oxygen therapy, and first and fifth minute Apgar score (P < 0.005). The mean PAB in infants without and with ROP was 19.79 HK (Hamidi-Koliakos) and 38.45 HK, respectively (P < 0.0001). Also, the mean PAB in neonates with ROP grade 1 and 2 was 36.69 HK and 45.53 HK, respectively (P = 0.002). CONCLUSION According to our findings, the PAB level can be helpful in predicting ROP incidence. With increasing PAB, the possibility of ROP severity will increase.
Collapse
|
23
|
Valls-Bellés V, Abad C, Hernández-Aguilar MT, Nacher A, Guerrero C, Baliño P, Romero FJ, Muriach M. Human Milk Antioxidative Modifications in Mastitis: Further Beneficial Effects of Cranberry Supplementation. Antioxidants (Basel) 2021; 11:antiox11010051. [PMID: 35052555 PMCID: PMC8772773 DOI: 10.3390/antiox11010051] [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: 12/01/2021] [Revised: 12/15/2021] [Accepted: 12/16/2021] [Indexed: 12/31/2022] Open
Abstract
Mastitis is the inflammation of one or several mammal lobes which can be accompanied by a mammary gland infection, and is the leading cause of undesired early weaning in humans. However, little information exists regarding the changes that this disease may induce in the biochemical composition of human milk, especially in terms of oxidative status. Given that newborns are subject to a significant increase in total ROS burden in their transition to neonatal life and that their antioxidant defense system is not completely developed, the aim of this study was to evaluate antioxidant defense (glutathione peroxidase (GPx), reduced glutathione (GSH), total polyphenol content (TPP), and total antioxidant capacity (TAC)) in milk samples from mothers suffering from mastitis and controls. We also measured the oxidative damage to lipids (malondyaldehyde (MDA)) and proteins (carbonyl group content (CGC)) in these samples. Finally, we tested whether dietary supplementation with cranberries (a product rich in antioxidants) in these breastfeeding mothers during 21 days could improve the oxidative status of milk. GPx activity, TPP, and TAC were increased in milk samples from mastitis-affected women, providing a protective mechanism to the newborn drinking mastitis milk. MDA concentrations were diminished in the mastitis group, confirming this proposal. Some oxidative damage might occur in the mammary gland since the CGC was increased in mastitis milk. Cranberries supplementation seems to strengthen the antioxidant system, further improving the antioxidative state of milk.
Collapse
Affiliation(s)
- Victoria Valls-Bellés
- Unitat Predepartamental de Medicina, Facultat de Ciencies de la Salud, Universitat Jaume I, 12071 Castellon de la Plana, Spain; (V.V.-B.); (C.A.); (M.T.H.-A.); (A.N.); (C.G.); (P.B.)
| | - Cristina Abad
- Unitat Predepartamental de Medicina, Facultat de Ciencies de la Salud, Universitat Jaume I, 12071 Castellon de la Plana, Spain; (V.V.-B.); (C.A.); (M.T.H.-A.); (A.N.); (C.G.); (P.B.)
| | - María Teresa Hernández-Aguilar
- Unitat Predepartamental de Medicina, Facultat de Ciencies de la Salud, Universitat Jaume I, 12071 Castellon de la Plana, Spain; (V.V.-B.); (C.A.); (M.T.H.-A.); (A.N.); (C.G.); (P.B.)
| | - Amalia Nacher
- Unitat Predepartamental de Medicina, Facultat de Ciencies de la Salud, Universitat Jaume I, 12071 Castellon de la Plana, Spain; (V.V.-B.); (C.A.); (M.T.H.-A.); (A.N.); (C.G.); (P.B.)
| | - Carlos Guerrero
- Unitat Predepartamental de Medicina, Facultat de Ciencies de la Salud, Universitat Jaume I, 12071 Castellon de la Plana, Spain; (V.V.-B.); (C.A.); (M.T.H.-A.); (A.N.); (C.G.); (P.B.)
| | - Pablo Baliño
- Unitat Predepartamental de Medicina, Facultat de Ciencies de la Salud, Universitat Jaume I, 12071 Castellon de la Plana, Spain; (V.V.-B.); (C.A.); (M.T.H.-A.); (A.N.); (C.G.); (P.B.)
| | - Francisco J. Romero
- Hospital General de Requena, Conselleria de Sanitat, Generalitat Valenciana, 46340 Requena, Spain
- Correspondence: (F.J.R.); (M.M.)
| | - María Muriach
- Unitat Predepartamental de Medicina, Facultat de Ciencies de la Salud, Universitat Jaume I, 12071 Castellon de la Plana, Spain; (V.V.-B.); (C.A.); (M.T.H.-A.); (A.N.); (C.G.); (P.B.)
- Correspondence: (F.J.R.); (M.M.)
| |
Collapse
|
24
|
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: 73] [Impact Index Per Article: 18.3] [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
|
25
|
Lembo C, Buonocore G, Perrone S. Oxidative Stress in Preterm Newborns. Antioxidants (Basel) 2021; 10:antiox10111672. [PMID: 34829543 PMCID: PMC8614893 DOI: 10.3390/antiox10111672] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2021] [Revised: 10/20/2021] [Accepted: 10/21/2021] [Indexed: 02/07/2023] Open
Abstract
Preterm babies are highly susceptible to oxidative stress (OS) due to an imbalance between the oxidant and antioxidant systems. The generation of free radicals (FR) induces oxidative damage to multiple body organs and systems. OS is the main factor responsible for the development of typical premature infant diseases, such as bronchopulmonary dysplasia, retinopathy of prematurity, necrotizing enterocolitis, intraventricular hemorrhage, periventricular leukomalacia, kidney damage, eryptosis, and also respiratory distress syndrome and patent ductus arteriosus. Many biomarkers have been detected to early identify newborns at risk of developing a free radical-mediated disease and to investigate new antioxidant strategies. This review reports the current knowledge on OS in the preterm newborns and the newest findings concerning the use of OS biomarkers as diagnostic tools, as well as in implementing antioxidant therapeutic strategies for the prevention and treatment of these diseases and their sequelae.
Collapse
Affiliation(s)
- Chiara Lembo
- Department of Molecular and Developmental Medicine, University of Siena, 53100 Siena, Italy; (C.L.); (G.B.)
| | - Giuseppe Buonocore
- Department of Molecular and Developmental Medicine, University of Siena, 53100 Siena, Italy; (C.L.); (G.B.)
| | - Serafina Perrone
- Department of Medicine and Surgery, Neonatology Unit, University of Parma, 43126 Parma, Italy
- Correspondence:
| |
Collapse
|
26
|
Ozdemir R, Gokce IK, Taslidere AC, Tanbek K, Gul CC, Sandal S, Turgut H, Kaya H, Aslan M. Does Chrysin prevent severe lung damage in Hyperoxia-Induced lung injury Model? Int Immunopharmacol 2021; 99:108033. [PMID: 34343938 DOI: 10.1016/j.intimp.2021.108033] [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: 04/15/2021] [Revised: 07/27/2021] [Accepted: 07/27/2021] [Indexed: 10/20/2022]
Abstract
BACKGROUND Oxidative stress and inflammation play a critical role in the etiopathogenesis of bronchopulmonary dysplasia (BPD). The aim of this study was to evaluate the preventive effect of Chrysin (CH), an antioxidant, antiinflammatory, antiapoptotic and antifibrotic drug, on hyperoxia-induced lung injury in a neonatal rat model. METHODS Forty infant rats were divided into four groups labeled the Control, CH, BPD, and BPD + CH. The control and CH groups were kept in a normal room environment, while the BPD and BPD + CH groups were kept in a hyperoxic (90-95%) environment. At the end of the study, lung tissue was evaluated with respect to apoptosis, histopathological damage and alveolar macrophage score as well as oxidant capacity, antioxidant capacity, and inflammation. RESULTS Compared to the BPD + CH and control groups, the lung tissues of the BPD group displayed substantially higher levels of MDA, TOS, TNF-α, and IL-1β (p < 0.05). While the BPD + CH group showed similar levels of TNF-α and IL-1β as the control group, MDA and TOS levels were higher than the control group, and significantly lower than the BPD group (p < 0.05). The BPD group exhibited considerably lower levels of TAS, SOD, GSH, and GSH-Px in comparison to the control group (p < 0.05). The BPD and BPD + CH groups exhibited higher mean scores of histopathological damage and alveolar macrophage when compared to the control and CH groups (p ≤ 0.0001). Both scores were found to be lower in the BPD + CH group in comparison to the BPD group (p ≤ 0.0001). The BPD + CH group demonstrated a significantly lower average of TUNEL and caspase-3 positive cells than the BPD group. CONCLUSION We found that prophylaxis with CH results in lower histopathological damage score and reduces apoptotic cell count, inflammation and oxidative stress while increasing anti-oxidant capacity.
Collapse
Affiliation(s)
- Ramazan Ozdemir
- Division of Neonatology, Department of Pediatrics, Inonu University School of Medicine, Malatya, Turkey.
| | - Ismail Kursat Gokce
- Division of Neonatology, Department of Pediatrics, Inonu University School of Medicine, Malatya, Turkey
| | - Asli Cetin Taslidere
- Department of Histology and Embryology, Inonu University School of Medicine, Malatya, Turkey
| | - Kevser Tanbek
- Department of Physiology, Inonu University School of Medicine, Malatya, Turkey
| | - Cemile Ceren Gul
- Department of Histology and Embryology, Inonu University School of Medicine, Malatya, Turkey
| | - Suleyman Sandal
- Department of Physiology, Inonu University School of Medicine, Malatya, Turkey
| | - Hatice Turgut
- Division of Neonatology, Department of Pediatrics, Inonu University School of Medicine, Malatya, Turkey
| | - Huseyin Kaya
- Division of Neonatology, Department of Pediatrics, Inonu University School of Medicine, Malatya, Turkey
| | - Mehmet Aslan
- Department of Pediatrics, Inonu University School of Medicine, Malatya, Turkey
| |
Collapse
|
27
|
Nair S, Rocha‐Ferreira E, Fleiss B, Nijboer CH, Gressens P, Mallard C, Hagberg H. Neuroprotection offered by mesenchymal stem cells in perinatal brain injury: Role of mitochondria, inflammation, and reactive oxygen species. J Neurochem 2021; 158:59-73. [PMID: 33314066 PMCID: PMC8359360 DOI: 10.1111/jnc.15267] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 12/03/2020] [Accepted: 12/03/2020] [Indexed: 12/11/2022]
Abstract
Preclinical studies have shown that mesenchymal stem cells have a positive effect in perinatal brain injury models. The mechanisms that cause these neurotherapeutic effects are not entirely intelligible. Mitochondrial damage, inflammation, and reactive oxygen species are considered to be critically involved in the development of injury. Mesenchymal stem cells have immunomodulatory action and exert mitoprotective effects which attenuate production of reactive oxygen species and promote restoration of tissue function and metabolism after perinatal insults. This review summarizes the present state, the underlying causes, challenges and possibilities for effective clinical translation of mesenchymal stem cell therapy.
Collapse
Affiliation(s)
- Syam Nair
- Centre of Perinatal Medicine and Health, Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
- Institute of Neuroscience and PhysiologySahlgrenska Academy, University of GothenburgGothenburgSweden
- Institute of Clinical SciencesSahlgrenska Academy, University of GothenburgGothenburgSweden
| | - Eridan Rocha‐Ferreira
- Centre of Perinatal Medicine and Health, Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
- Institute of Neuroscience and PhysiologySahlgrenska Academy, University of GothenburgGothenburgSweden
- Institute of Clinical SciencesSahlgrenska Academy, University of GothenburgGothenburgSweden
| | - Bobbi Fleiss
- School of Health and Biomedical SciencesRMIT UniversityBundooraVictoriaAustralia
- Université de Paris, NeuroDiderotParisFrance
| | - Cora H Nijboer
- Department for Developmental Origins of DiseaseUniversity Medical Center Utrecht Brain Center and Wilhelmina Children’s Hospital, Utrecht UniversityUtrechtNetherlands
| | | | - Carina Mallard
- Centre of Perinatal Medicine and Health, Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
- Institute of Neuroscience and PhysiologySahlgrenska Academy, University of GothenburgGothenburgSweden
| | - Henrik Hagberg
- Centre of Perinatal Medicine and Health, Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
- Institute of Clinical SciencesSahlgrenska Academy, University of GothenburgGothenburgSweden
| |
Collapse
|
28
|
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
|
29
|
Medical-Grade Honey for the Treatment of Extravasation-Induced Injuries in Preterm Neonates: A Case Series. Adv Neonatal Care 2021; 21:122-132. [PMID: 32675576 DOI: 10.1097/anc.0000000000000781] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
INTRODUCTION Preterm neonates often depend on peripheral intravenous administration of nutrition and medication. Since their skin is not fully developed and very vulnerable, extravasation injury is a risk. Medical-grade honey (MGH) possesses antimicrobial activity and stimulates wound healing; although its use in neonatal patients is limited. CLINICAL FINDINGS We present a case series of 7 preterm neonates (28-36 weeks of gestation) with extravasation injuries secondary to peripheral intravenous administration of total parental nutrition and medication. PRIMARY DIAGNOSIS Extravasation injury following the unintentional leakage of total parenteral nutrition, and medication into the surrounding tissue. Signs of extravasation include local pain, erythema, burning, pruritus, and/or swelling. INTERVENTIONS All extravasation injuries were treated with daily cleaning and application of MGH. Some of the cases needed additional surgical intervention or assisted debridement. OUTCOMES After treatment, all extravasation injury wounds presented with granulation tissue formation progressed to normal epithelialization and closed in 7 to 67 days (median: 32 days). Upon initial application, peripheral edema and inflammation decreased. When present, necrotic tissue was effectively debrided, slough was removed, and no signs of infection were detected, irrespective of initial wound presentations. Cicatrization was minimal, and the full range of motion was preserved in all cases. PRACTICE RECOMMENDATIONS Continuous and thorough assessment of peripheral intravenous line placement for malposition, leaking, and signs of extravasation is needed for fast discovery and prevention of further damage. CONCLUSION Medical-grade honey possesses antimicrobial, anti-inflammatory, and antioxidative activity, enhancing wound healing. Medical-grade honey was safe and effective for treating extravasation-induced injuries, independent of location and severity. We recommend MGH for treating extravasation wounds and consideration for other types of wounds.
Collapse
|
30
|
Neonatal Selenoenzyme Expression Is Variably Susceptible to Duration of Maternal Selenium Deficiency. Antioxidants (Basel) 2021; 10:antiox10020288. [PMID: 33672905 PMCID: PMC7917816 DOI: 10.3390/antiox10020288] [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: 01/11/2021] [Revised: 02/03/2021] [Accepted: 02/08/2021] [Indexed: 12/16/2022] Open
Abstract
Maternal selenium (Se) deficiency is associated with decreased neonatal Se levels, which increases the risk for neonatal morbidities. There is a hierarchy to selenoprotein expression after Se deficiency in adult rodents, depending on the particular protein and organ evaluated. However, it is unknown how limited Se supply during pregnancy impacts neonatal selenoprotein expression. We used an Se-deficient diet to induce perinatal Se deficiency (SeD), initiated 2–4 weeks before onset of breeding and continuing through gestation. Neonatal plasma, liver, heart, kidney, and lung were collected on the day of birth and assessed for selenoproteins, factors required for Se processing, and non-Se containing antioxidant enzymes (AOE). Maternal SeD reduced neonatal circulating and hepatic glutathione peroxidase (GPx) activity, as well as hepatic expression of Gpx1 and selenophosphate synthetase 2 (Sps2). In contrast, the impact of maternal SeD on hepatic thioredoxin reductase 1, hepatic non-Se containing AOEs, as well as cardiac, renal, and pulmonary GPx activity, varied based on duration of maternal exposure to SeD diet. We conclude that the neonatal liver and circulation demonstrate earlier depletion in selenoenzyme activity after maternal SeD. Our data indicate that prolonged maternal SeD may escalate risk to the neonate by progressively diminishing Se-containing AOE across multiple organs.
Collapse
|
31
|
Xu Y, Yu Z, Li Q, Zhou J, Yin X, Ma Y, Yin Y, Jiang S, Zhu R, Wu Y, Han L, Gao Y, Xue M, Qiao Y, Zhu L, Tu W, Wu M, Wan J, Wang W, Deng X, Li S, Wang S, Chen X, Zhou Q, Wang J, Cheng R, Wang J, Han S. Dose-dependent effect of human milk on Bronchopulmonary dysplasia in very low birth weight infants. BMC Pediatr 2020; 20:522. [PMID: 33190629 PMCID: PMC7666971 DOI: 10.1186/s12887-020-02394-1] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Accepted: 10/16/2020] [Indexed: 12/13/2022] Open
Abstract
Background and aim Human milk has potential protective effects against bronchopulmonary dysplasia (BPD). However, studies on the association between the dose of human milk and BPD in China are limited. This study aimed to evaluate the dose-dependent effects of human milk on BPD and other neonatal morbidities in very low birth weight (VLBW) infants. Methods This retrospective cohort study of preterm infants was conducted on preterm infants of gestational age ≤ 34 weeks and birth weight < 1500 g admitted to the multicenter clinical research database for breastfeeding quality improvement in Jiangsu province. The multivariate analysis was performed to compare the effect outcomes of daily graded doses [1–24 mL/(kg · day), 25–49 mL/(kg · day), and ≥ 50 mL/(kg · day) of body weight] of human milk on neonatal outcomes throughout the first 4 weeks of life versus a reference group receiving no human milk. The models were adjusted for potential confounding variables. Results Of 964 included infants, 279 (28.9%) received exclusive preterm formula, 128 (13.3%) received 1–24 ml/(kg · day), 139 (14.4%) received 25–49 ml/(kg · day), and 418 (43.4%) received ≥50 ml/(kg · day) human milk for the first 4 weeks of life. Compared with infants receiving exclusive formula, those receiving the highest volume of human milk daily [≥50 mL/(kg · day)] had lower incidences of BPD [27.5% in ≥50 mL/(kg · day) vs 40.1% in 0 mL/(kg · day) human milk, P = 0.001)], moderate and severe BPD [8.9% in ≥50 mL/(kg · day) vs 16.1% in 0 mL/(kg · day), P = 0.004], necrotizing enterocolitis [NEC; 3.8% in ≥50 mL/(kg · day) vs 10.8% in 0 mL/(kg · day), P = 0.001], late-onset sepsis [LOS; 9.3% in ≥50 mL/(kg · day) vs 19.7% in 0 mL/(kg · day), P <0.01], and extrauterine growth retardation [EUGR; 38.5% in ≥50 mL/(kg · day) vs 57.6% in 0 mL/(kg · day), P <0.01)]. The logistic regression indicated that those receiving ≥50 ml/kg · day human milk had lower odds of BPD [adjusted odds ratio (AOR) 0.453; 95% confidence interval (CI): 0.309, 0.666], moderate and severe BPD (AOR 0.430; 95% CI: 0.249, 0.742), NEC (AOR 0.314; 95% CI: 0.162, 0. 607), LOS (AOR 0.420; 95% CI: 0.263, 0.673), and EUGR (AOR 0.685; 95% CI: 0.479, 0.979). Conclusions A daily threshold amount of ≥50 ml/(kg · day) human milk in the first 4 weeks of life was associated with lower incidence of BPD as well as NEC, LOS, and EUGR in VLBW infants. Trial registration ClinicalTrials.gov Identifier: NCT03453502. Registration date: March 5, 2018. This study was retrospectively registered. Supplementary information Supplementary information accompanies this paper at 10.1186/s12887-020-02394-1.
Collapse
Affiliation(s)
- Yan Xu
- Department of Neonatology, The Affiliated Hospital of Xuzhou Medical University, No. 99, Huaihai West Road, Xuzhou, 221000, Jiangsu Province, China.,Department of Neonatology, The Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, 210004, Jiangsu, China
| | - Zhangbin Yu
- Department of Neonatology, The Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, 210004, Jiangsu, China
| | - Qianqian Li
- Department of Neonatology, Xuzhou Maternity and Child Health Care Hospital, Xuzhou, 210009, Jiangsu, China
| | - Jinjun Zhou
- Department of Neonatology, Nantong Maternity and Child Health Care Hospital, Nantong, 226001, Jiangsu, China
| | - Xiaoguang Yin
- Department of Neonatology, Anhui women and Child Health Care Hospital, Hefei, 230001, Anhui, China
| | - Yuelan Ma
- Department of Neonatology, The Affiliated Suzhou Hospital of Nanjing Medical University and Suzhou Municipal Hospital, Suzhou, 215002, Jiangsu, China
| | - Yujie Yin
- Department of Neonatology, Jiangsu Women and Children Health Hospital, Nanjing, 210036, Jiangsu, China
| | - Shanyu Jiang
- Department of Neonatology, Wuxi Maternity and Child Health Care Hospital, Wuxi, 214002, Jiangsu, China
| | - Rongping Zhu
- Department of Neonatology, Changzhou Maternity and Child Health Care Hospital, Changzhou, 213003, Jiangsu, China
| | - Yue Wu
- Department of Neonatology, Nanjing Children's Hospital, Nanjing, 210008, Jiangsu, China
| | - Liangrong Han
- Department of Neonatology, Huaian Maternity and Child Health Care Hospital, Huaian, 223002, Jiangsu, China
| | - Yan Gao
- Department of Neonatology, Lianyungang Maternity and Child Health Care Hospital, Lianyungang, 222000, Jiangsu, China
| | - Mei Xue
- Department of Neonatology, Taizhou People's Hospital, Taizhou, 225300, Jiangsu, China
| | - Yu Qiao
- Department of Neonatology, Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, Jiangsu, China
| | - Lingling Zhu
- Department of Neonatology, Northern Jiangsu People's Hospital, Yangzhou, 225001, Jiangsu, China
| | - Wenjuan Tu
- Department of Neonatology, Changzhou Children's Hospital, Changzhou, 213003, Jiangsu, China
| | - Mingfu Wu
- Department of Neonatology, Affiliated Hospital of Yangzhou University, Yangzhou, 225001, Jiangsu, China
| | - Jun Wan
- Department of Neonatology, Jiangyin People's Hospital, Jiangyin, 214400, Jiangsu, China
| | - Weiyuan Wang
- Department of Neonatology, Suqian Maternity Hospital, Suqian, 223800, Jiangsu, China
| | - Xiaoyi Deng
- Department of Neonatology, Xuzhou Maternity and Child Health Care Hospital, Xuzhou, 210009, Jiangsu, China
| | - Shuangshuang Li
- Department of Neonatology, Nantong Maternity and Child Health Care Hospital, Nantong, 226001, Jiangsu, China
| | - Sannan Wang
- Department of Neonatology, The Affiliated Suzhou Hospital of Nanjing Medical University and Suzhou Municipal Hospital, Suzhou, 215002, Jiangsu, China
| | - Xiaoqing Chen
- Department of Neonatology, Jiangsu Women and Children Health Hospital, Nanjing, 210036, Jiangsu, China
| | - Qin Zhou
- Department of Neonatology, Wuxi Maternity and Child Health Care Hospital, Wuxi, 214002, Jiangsu, China
| | - Jinxiu Wang
- Department of Neonatology, Changzhou Maternity and Child Health Care Hospital, Changzhou, 213003, Jiangsu, China
| | - Rui Cheng
- Department of Neonatology, Nanjing Children's Hospital, Nanjing, 210008, Jiangsu, China
| | - Jun Wang
- Department of Neonatology, The Affiliated Hospital of Xuzhou Medical University, No. 99, Huaihai West Road, Xuzhou, 221000, Jiangsu Province, China.
| | - Shuping Han
- Department of Neonatology, The Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, 210004, Jiangsu, China. .,Department of Pediatrics, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, No. 123 Tian Fei Xiang, Mo Chou Road, Nanjing, Jiangsu Province, 210004, China.
| |
Collapse
|
32
|
Hill NS, Cormack BE, Little BS, Bloomfield FH. Growth and Clinical Outcome in Very Low-Birth-Weight Infants After the Introduction of a Multicomponent Intravenous Lipid Emulsion. JPEN J Parenter Enteral Nutr 2020; 44:1318-1327. [PMID: 31989691 DOI: 10.1002/jpen.1750] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Revised: 10/18/2019] [Accepted: 11/05/2019] [Indexed: 01/08/2023]
Abstract
BACKGROUND Very low-birth-weight (VLBW; birth weight < 1500 g) infants are often dependent on intravenous nutrition after birth. Conventional soy-based intravenous lipid emulsions (Soy LE) are associated with inflammatory and metabolic complications that may be harmful to preterm infants. Evidence to support any clinical benefit associated with newer multicomponent emulsions (Mixed LE), remains inconsistent and unsubstantiated in appropriate studies. This retrospective study aimed to determine whether growth and clinical outcomes differed between VLBW infants given Mixed LE vs Soy LE at Auckland City Hospital. METHODS Data were collected on nutrition, growth, and neonatal morbidities for the first 4 weeks after birth. Outcomes were compared between 2 lipid cohorts, Soy LE (February 2013 - August 2014) and Mixed LE (August 2014 - December 2015), using univariate and multivariate analysis. RESULTS 207 infants (Soy LE, 105 vs Mixed LE, 102) were included in the study. Significantly fewer infants in the Mixed LE cohort developed any stage retinopathy of prematurity (Soy LE 59% vs Mixed LE 39%, P = .005) or intraventricular hemorrhage (Soy LE 27% vs Mixed LE 15%, P = .03) during their admission. Mixed LE was also associated with significantly lower mean (P = .01), minimum (P = .03), and maximum (P = .04) total bilirubin concentrations across the first 4 weeks after birth. There was no difference in growth velocity or weight, length, and head circumference z-score change. CONCLUSION SMOFlipid may represent a favorable alternative to conventional lipid emulsions in neonatal parenteral nutrition regimens; however, long-term effects should be further evaluated.
Collapse
Affiliation(s)
| | - Barbara E Cormack
- Liggins Institute, The University of Auckland, Auckland, New Zealand.,Newborn Services, Auckland City Hospital, Auckland, New Zealand
| | | | - Frank H Bloomfield
- Liggins Institute, The University of Auckland, Auckland, New Zealand.,Newborn Services, Auckland City Hospital, Auckland, New Zealand
| |
Collapse
|
33
|
Wang SH, Tsao PN. Phenotypes of Bronchopulmonary Dysplasia. Int J Mol Sci 2020; 21:ijms21176112. [PMID: 32854293 PMCID: PMC7503264 DOI: 10.3390/ijms21176112] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 08/20/2020] [Accepted: 08/22/2020] [Indexed: 12/18/2022] Open
Abstract
Bronchopulmonary dysplasia (BPD) is the most common chronic morbidity in preterm infants. In the absence of effective interventions, BPD is currently a major therapeutic challenge. Several risk factors are known for this multifactorial disease that results in disrupted lung development. Inflammation plays an important role and leads to persistent airway and pulmonary vascular disease. Since corticosteroids are potent anti-inflammatory agents, postnatal corticosteroids have been used widely for BPD prevention and treatment. However, the clinical responses vary to a great degree across individuals, and steroid-related complications remain major concerns. Emerging studies on the molecular mechanism of lung alveolarization during inflammatory stress will elucidate the complicated pathway and help discover novel therapeutic targets. Moreover, with the advances in metabolomics, there are new opportunities to identify biomarkers for early diagnosis and prognosis prediction of BPD. Pharmacometabolomics is another novel field aiming to identify the metabolomic changes before and after a specific drug treatment. Through this "metabolic signature," a more precise treatment may be developed, thereby avoiding unnecessary drug exposure in non-responders. In the future, more clinical, genetic, and translational studies would be required to improve the classification of BPD phenotypes and achieve individualized care to enhance the respiratory outcomes in preterm infants.
Collapse
Affiliation(s)
- Shih-Hsin Wang
- Department of Pediatrics, Far Eastern Memorial Hospital, New Taipei City 22060, Taiwan;
| | - Po-Nien Tsao
- Department of Pediatrics, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei 100225, Taiwan
- Center for Developmental Biology & Regenerative Medicine, National Taiwan University, Taipei 100226, Taiwan
- Correspondence: ; Tel.: +886-2-23123456 (ext. 71013)
| |
Collapse
|
34
|
Gila-Díaz A, Herranz Carrillo G, Cañas S, Saenz de Pipaón M, Martínez-Orgado JA, Rodríguez-Rodríguez P, López de Pablo ÁL, Martin-Cabrejas MA, Ramiro-Cortijo D, Arribas SM. Influence of Maternal Age and Gestational Age on Breast Milk Antioxidants During the First Month of Lactation. Nutrients 2020; 12:nu12092569. [PMID: 32854220 PMCID: PMC7551022 DOI: 10.3390/nu12092569] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 08/14/2020] [Accepted: 08/21/2020] [Indexed: 12/15/2022] Open
Abstract
Breast milk (BM) is beneficial due to its content in a wide range of different antioxidants, particularly relevant for preterm infants, who are at higher risk of oxidative stress. We hypothesize that BM antioxidants are adapted to gestational age and are negatively influenced by maternal age. Fifty breastfeeding women from two hospitals (Madrid, Spain) provided BM samples at days 7, 14 and 28 of lactation to assess total antioxidant capacity (ABTS), thiol groups, reduced glutathione (GSH), superoxide dismutase (SOD) and catalase activities, lipid peroxidation (malondialdehyde, MDA + 4-Hydroxy-Trans-2-Nonenal, HNE), protein oxidation (carbonyl groups) (spectrophotometry) and melatonin (ELISA). Mixed random-effects linear regression models were used to study the influence of maternal and gestational ages on BM antioxidants, adjusted by days of lactation. Regression models evidenced a negative association between maternal age and BM melatonin levels (β = -7.4 ± 2.5; p-value = 0.005); and a negative association between gestational age and BM total antioxidant capacity (β = -0.008 ± 0.003; p-value = 0.006), SOD activity (β = -0.002 ± 0.001; p-value = 0.043) and protein oxidation (β = -0.22 ± 0.07; p-value = 0.001). In conclusion, BM antioxidants are adapted to gestational age providing higher levels to infants with lower degree of maturation; maternal ageing has a negative influence on melatonin, a key antioxidant hormone.
Collapse
Affiliation(s)
- Andrea Gila-Díaz
- Department of Physiology, Faculty of Medicine, Universidad Autónoma de Madrid, C/Arzobispo Morcillo 2, 28029 Madrid, Spain; (A.G.-D.); (P.R.-R.); (Á.L.L.d.P.)
| | - Gloria Herranz Carrillo
- Division of Neonatology, Hospital Clínico San Carlos, Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), C/Profesor Martín Lagos s/n, 28040 Madrid, Spain; (G.H.C.); (J.A.M.-O.)
| | - Silvia Cañas
- Department of Agricultural and Food Chemistry-CIAL, Faculty of Sciences, Universidad Autónoma de Madrid, Ciudad Universitaria de Cantoblanco, 28049 Madrid, Spain; (S.C.); (M.A.M.-C.)
| | - Miguel Saenz de Pipaón
- Department of Neonatology, Hospital La Paz, Paseo de la Castellana 216, 28046 Madrid, Spain;
| | - José Antonio Martínez-Orgado
- Division of Neonatology, Hospital Clínico San Carlos, Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), C/Profesor Martín Lagos s/n, 28040 Madrid, Spain; (G.H.C.); (J.A.M.-O.)
| | - Pilar Rodríguez-Rodríguez
- Department of Physiology, Faculty of Medicine, Universidad Autónoma de Madrid, C/Arzobispo Morcillo 2, 28029 Madrid, Spain; (A.G.-D.); (P.R.-R.); (Á.L.L.d.P.)
| | - Ángel Luis López de Pablo
- Department of Physiology, Faculty of Medicine, Universidad Autónoma de Madrid, C/Arzobispo Morcillo 2, 28029 Madrid, Spain; (A.G.-D.); (P.R.-R.); (Á.L.L.d.P.)
| | - María A. Martin-Cabrejas
- Department of Agricultural and Food Chemistry-CIAL, Faculty of Sciences, Universidad Autónoma de Madrid, Ciudad Universitaria de Cantoblanco, 28049 Madrid, Spain; (S.C.); (M.A.M.-C.)
| | - David Ramiro-Cortijo
- Department of Physiology, Faculty of Medicine, Universidad Autónoma de Madrid, C/Arzobispo Morcillo 2, 28029 Madrid, Spain; (A.G.-D.); (P.R.-R.); (Á.L.L.d.P.)
- Division of Gastroenterology, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline avenue, 02215 Boston, MA, USA
- Correspondence: (D.R.-C.); (S.M.A.)
| | - Silvia M. Arribas
- Department of Physiology, Faculty of Medicine, Universidad Autónoma de Madrid, C/Arzobispo Morcillo 2, 28029 Madrid, Spain; (A.G.-D.); (P.R.-R.); (Á.L.L.d.P.)
- Correspondence: (D.R.-C.); (S.M.A.)
| |
Collapse
|
35
|
Yakut HI, Koyuncu E, Cakir U, Tayman C, Koyuncu İ, Taskin Turkmenoglu T, Cakir E, Ozyazici A, Aydogan S, Zenciroglu A. Preventative and therapeutic effects of fennel (Foeniculum vulgare) seed extracts against necrotizing enterocolitis. J Food Biochem 2020; 44:e13284. [PMID: 32510653 DOI: 10.1111/jfbc.13284] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 04/22/2020] [Accepted: 04/27/2020] [Indexed: 02/06/2023]
Abstract
We aimed to understand the efficacy of fennel (Foeniculum vulgare: FV) extract in an experimental necrotizing enterocolitis (NEC) model. Forty-two rat pups were divided into three groups as NEC, NEC treated with fennel extract, and control. At the end of the experiment, tissue samples were taken from the proximal colon and ileum for biochemical and immuno-histological studies including hematoxylin-eosin and Caspase-3-8-9 immunohistochemical staining. Bowel damage and apoptosis were found to be less in the NEC + FV group. Oxidant stress, caspase 3, TNF-α, and IL-6 levels were considerably decreased in the NEC + FV group. Antioxidants were significantly higher in the NEC + FV group more than in the NEC group. Moreover, protein, DNA damage, and lipid peroxidation were found to be decreased in the NEC + FV group compared to the NEC group. PRACTICAL APPLICATIONS: Intense inflammation, oxidant stress, apoptosis, and infection are important in the development of NEC. Fennel has anti-oxidant, anti-inflammatory, antibacterial, antifungal, antiviral, immunomodulatory effects. Fennel extract might be a novel option in the treatment of NEC through its anti-oxidant, anti-inflammatory, anti-apoptotic, and cytoprotective features.
Collapse
Affiliation(s)
- Halil Ibrahim Yakut
- Department of Pediatrics, Ankara Hematology Oncology Children Education and Research Hospital, Health Sciences University, Ankara, Turkey
| | - Ece Koyuncu
- Department of Neonatology, Dr. Sami Ulus Maternity and Children Education and Research Hospital, Health Sciences University, Ankara, Turkey
| | - Ufuk Cakir
- Department of Neonatology, Zekai Tahir Burak Maternity Education and Research Hospital, Health Sciences University, Ankara, Turkey
| | - Cuneyt Tayman
- Department of Neonatology, Zekai Tahir Burak Maternity Education and Research Hospital, Health Sciences University, Ankara, Turkey
| | - İsmail Koyuncu
- Faculty of Medicine, Department of Biochemistry, Harran University, Sanlıurfa, Turkey
| | - Tugba Taskin Turkmenoglu
- Department of Pathology, Ankara Yıldırım Beyazıt Education and Research Hospital, Health Sciences University, Ankara, Turkey
| | - Esra Cakir
- Anesthesiology and Clinical of Critical Care, Ankara Numune Education and Research Hospital, Health Sciences University, Ankara, Turkey
| | - Ahmet Ozyazici
- Department of Neonatology, Dr. Sami Ulus Maternity and Children Education and Research Hospital, Health Sciences University, Ankara, Turkey
| | - Seda Aydogan
- Department of Neonatology, Dr. Sami Ulus Maternity and Children Education and Research Hospital, Health Sciences University, Ankara, Turkey
| | - Aysegul Zenciroglu
- Department of Neonatology, Dr. Sami Ulus Maternity and Children Education and Research Hospital, Health Sciences University, Ankara, Turkey
| |
Collapse
|
36
|
Killian B, Yuan TH, Tsai CH, Chiu THT, Chen YH, Chan CC. Emission-related Heavy Metal Associated with Oxidative Stress in Children: Effect of Antioxidant Intake. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2020; 17:ijerph17113920. [PMID: 32492875 PMCID: PMC7312654 DOI: 10.3390/ijerph17113920] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 05/22/2020] [Accepted: 05/30/2020] [Indexed: 12/13/2022]
Abstract
Heavy metals, the common pollutants emitted from industrial activities, are believed to cause harmful effects, partially through the mechanism of elevated oxidative stress, and antioxidant intake has been hypothesized to provide a potential protective effect against oxidative stress. This study aims to investigate the heavy metal exposure and the associated oxidative damage of young children living near a petrochemical complex and to assess the protective effect of antioxidant intake. There were 168 children recruited from the kindergartens near a huge petrochemical complex, with 87 as the high exposure group and 81 as the low exposure group. Urinary concentrations of eleven metals were detected by inductively coupled plasma mass spectrometry, and four biomarkers of oxidative stress were measured in urine by liquid chromatography-tandem mass spectrometry. The food frequency questionnaire was collected to assess participants’ intake of antioxidants. Multiple linear regression was performed to determine the predictors of metals for oxidative stress and to measure the beneficial effect of antioxidants. Weighted quantile sum regression was performed to determine the contributors among metals to the oxidative stress. Results showed that high exposure group had significantly higher concentrations of chromium, manganese, nickel, arsenic, strontium, cadmium, and lead when compared to those in low exposure group. There was no obviously difference on the total antioxidant intake and dietary profile between two groups. The elevated levels of two oxidative stress markers were significantly associated with most of the urinary metal concentrations in all study subjects after adjusting confounders, while no significant association was found between oxidative stress and antioxidant intake. Among the metals, mercury and strontium showed the dominated contributions for elevated levels of oxidative stress. It concluded that higher metal exposure was associated with elevated oxidative stress but with no protective effect by antioxidant intake among the young children residents near a petrochemical industry.
Collapse
Affiliation(s)
- Brittany Killian
- Master of Public Health Degree Program, College of Public Health, National Taiwan University, No. 17, Xu-Zhou Road, Taipei 10055, Taiwan;
| | - Tzu-Hsuen Yuan
- Institute of Environmental and Occupational Health Science, College of Public Health, National Taiwan University, No. 17, Xu-Zhou Road, Taipei 10055, Taiwan; (T.-H.Y.); (Y.-H.C.)
- Innovation and Policy Center for Population Health and Sustainable Environment (Population Health Research Center, PHRC), College of Public Health, National Taiwan University, No. 17, Xu-Zhou Road, Taipei 10055, Taiwan
| | - Cheng-Hsien Tsai
- National Taiwan University Hospital, Yunlin Branch, No.579, Sec. 2, Yunlin Rd., Douliu City, Yunlin County 64041, Taiwan;
| | - Tina H. T. Chiu
- Department of Nutritional Science, Fu-Jen Catholic University, No. 510, Zhongzheng Rd., Xinzhang Dist., New Taipei City 24205, Taiwan;
| | - Yi-Hsuan Chen
- Institute of Environmental and Occupational Health Science, College of Public Health, National Taiwan University, No. 17, Xu-Zhou Road, Taipei 10055, Taiwan; (T.-H.Y.); (Y.-H.C.)
| | - Chang-Chuan Chan
- Institute of Environmental and Occupational Health Science, College of Public Health, National Taiwan University, No. 17, Xu-Zhou Road, Taipei 10055, Taiwan; (T.-H.Y.); (Y.-H.C.)
- Innovation and Policy Center for Population Health and Sustainable Environment (Population Health Research Center, PHRC), College of Public Health, National Taiwan University, No. 17, Xu-Zhou Road, Taipei 10055, Taiwan
- Correspondence:
| |
Collapse
|
37
|
Baldassarre ME, Di Mauro A, Labellarte G, Pignatelli M, Fanelli M, Schiavi E, Mastromarino P, Capozza M, Panza R, Laforgia N. Resveratrol plus carboxymethyl-β-glucan in infants with common cold: A randomized double-blind trial. Heliyon 2020; 6:e03814. [PMID: 32322697 PMCID: PMC7172624 DOI: 10.1016/j.heliyon.2020.e03814] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 04/02/2020] [Accepted: 04/16/2020] [Indexed: 12/30/2022] Open
Abstract
Objectives To evaluate effectiveness of a nasal resveratrol/carboxymethyl-β-glucan solution compared to nasal saline solution: a) on common cold symptoms by means of a validated measure scale (CARIFS score), b) on Rhinovirus infection and CCL2, CCL5, IL8, IL6, CXCL10 and TLR2 expression in nasal swabs, c) on frequency of relapses after 30 days of follow-up. Methods 89 infants with respiratory infection symptoms were randomly assigned to receive either a nasal resveratrol/carboxymethyl-β-glucan solution or nasal saline solution. All patients were evaluated with CARIFS score at enrollment, after 48 h, 7 and 30 days by physicians and parents. Nasal swabs were obtained at enrollment, after 48 h and after one week. Results CARIFS score improved in both groups. Episodes of sneezing and cough were fewer in study group after 7 days of follow-up (p < 0.05). No significant differences were found on nasopharyngeal swabs in Rhinovirus detection and cytokines expression after 48 h, nor in 30 days relapses. TLR2 expression was significantly higher in Rhinovirus infected children of the study group. No adverse effects occurred. Conclusions These data suggest that a solution containing resveratrol plus carboxymethyl-β-glucan might have a positive impact on both clinical and socio-economic burden due to infant common cold.
Collapse
Affiliation(s)
- Maria Elisabetta Baldassarre
- Department of Biomedical Science and Human Oncology, Neonatology and Neonatal Intensive Care Unit, "Aldo Moro" University of Bari, Bari 70100, Italy
| | - Antonio Di Mauro
- Department of Biomedical Science and Human Oncology, Neonatology and Neonatal Intensive Care Unit, "Aldo Moro" University of Bari, Bari 70100, Italy
| | - Grazia Labellarte
- Department of Biomedical Science and Human Oncology, Neonatology and Neonatal Intensive Care Unit, "Aldo Moro" University of Bari, Bari 70100, Italy
| | - Mariacristina Pignatelli
- Department of Biomedical Science and Human Oncology, Neonatology and Neonatal Intensive Care Unit, "Aldo Moro" University of Bari, Bari 70100, Italy
| | - Margherita Fanelli
- Department of Interdisciplinary Medicine, "Aldo Moro" University of Bari, Bari 70100, Italy
| | - Elisa Schiavi
- Department of Public Health and Infectious Disease, "Sapienza" University of Rome, Rome 00100, Italy
| | - Paola Mastromarino
- Department of Public Health and Infectious Disease, "Sapienza" University of Rome, Rome 00100, Italy
| | - Manuela Capozza
- Department of Biomedical Science and Human Oncology, Neonatology and Neonatal Intensive Care Unit, "Aldo Moro" University of Bari, Bari 70100, Italy
| | - Raffaella Panza
- Department of Biomedical Science and Human Oncology, Neonatology and Neonatal Intensive Care Unit, "Aldo Moro" University of Bari, Bari 70100, Italy
| | - Nicola Laforgia
- Department of Biomedical Science and Human Oncology, Neonatology and Neonatal Intensive Care Unit, "Aldo Moro" University of Bari, Bari 70100, Italy
| |
Collapse
|
38
|
Low Expression of Sirtuin 1 in the Dairy Cows with Mild Fatty Liver Alters Hepatic Lipid Metabolism. Animals (Basel) 2020; 10:ani10040560. [PMID: 32230804 PMCID: PMC7222401 DOI: 10.3390/ani10040560] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2020] [Revised: 03/19/2020] [Accepted: 03/24/2020] [Indexed: 12/11/2022] Open
Abstract
Simple Summary Sirtuin 1 (SIRT1), a NAD-dependent histone deacetylase, is involved in oxidative stress and lipid metabolism regulation. Limited studies exist regarding the role of SIRT1 in lipid metabolism disorder in periparturient dairy cows. This study explores the effect of hepatic steatosis on the expression of the SIRT1 gene and protein and the proteins encoded by the genes downstream to it, all of which are involved in lipid metabolism in the liver. Control cows (n = 6, parity 3.0 ± 2.0, milk production 28 ± 47 kg/d) and mild fatty liver cows (n = 6, parity 2.3 ± 1.5, milk production 20 ± 6 kg/d) were retrospectively selected based on liver triglycerides (TG) content (% wet liver). The present study indicates that low SIRT1 expression caused by hepatic steatosis promotes hepatic fatty acid synthesis and inhibits fatty acid β-oxidation. We believe that our study makes a significant contribution to the literature because it demonstrates that hepatic steatosis is associated with increased hepatic fatty acid synthesis, inhibited fatty acid β-oxidation and reduced lipid transport. Abstract Dairy cows usually experience negative energy balance coupled with an increased incidence of fatty liver during the periparturient period. The purpose of this study was to investigate the effect of hepatic steatosis on the expression of the sirtuin 1 (SIRT1), along with the target mRNA and protein expressions and activities related to lipid metabolism in liver tissue. Control cows (n = 6, parity 3.0 ± 2.0, milk production 28 ± 7 kg/d) and mild fatty liver cows (n = 6, parity 2.3 ± 1.5, milk production 20 ± 6 kg/d) were retrospectively selected based on liver triglycerides (TG) content (% wet liver). Compared with the control group, fatty liver cows had greater concentrations of cholesterol and TG along with the typically vacuolated appearance and greater lipid droplets in the liver. Furthermore, fatty liver cows had greater mRNA and protein abundance related to hepatic lipid synthesis proteins sterol regulatory element binding proteins (SREBP-1c), long-chain acyl-CoA synthetase (ACSL), acyl-CoA carbrolase (ACC) and fatty acid synthase (FAS) and lipid transport proteins Liver fatty acid binding protein (L-FABP), apolipoprotein E (ApoE), low density lipoprotein receptor (LDLR) and microsomal TG transfer protein (MTTP) (p < 0.05). However, they had lower mRNA and protein abundance associated with fatty acid β-oxidation proteins SIRT1, peroxisome proliferator-activated receptor co-activator-1 (PGC-1α), peroxisome proliferator–activated receptor-α (PPARα), retinoid X receptor (RXRα), acyl-CoA 1 (ACO), carnitine palmitoyltransferase 1 (CPT1), carnitine palmitoyltransferase 2 (CPT2) and long- and medium-chain 3-hydroxyacyl-CoA dehydrogenases (LCAD) (p < 0.05). Additionally, mRNA abundance and enzyme activity of enzymes copper/zinc superoxide dismutase (Cu/Zn SOD), catalase (CAT), glutathione peroxidase (GSH-Px) and manganese superoxide dismutase (Mn SOD) decreased and mRNA and protein abundance of p45 nuclear factor-erythroid 2 (p45 NF-E2)-related factor 1 (Nrf1), mitochondrial transcription factor A (TFAM) decreased (p < 0.05). Lower enzyme activities of SIRT1, PGC-1α, Cu/Zn SOD, CAT, GSH-Px, SREBP-1c and Mn SOD (p < 0.05) and concentration of reactive oxygen species (ROS) were observed in dairy cows with fatty liver. These results demonstrate that decreased SIRT1 associated with hepatic steatosis promotes hepatic fatty acid synthesis and inhibits fatty acid β-oxidation. Hence, SIRT1 may represent a novel therapeutic target for the treatment of the fatty liver disease in dairy cows.
Collapse
|
39
|
Plasma Oxidative Status in Preterm Infants Receiving LCPUFA Supplementation: A Pilot Study. Nutrients 2020; 12:nu12010122. [PMID: 31906339 PMCID: PMC7019959 DOI: 10.3390/nu12010122] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Revised: 12/29/2019] [Accepted: 12/30/2019] [Indexed: 02/07/2023] Open
Abstract
After birth, preterm infants are deficient in arachidonic acid (ARA), docosahexaenoic acid (DHA), and antioxidants, increasing their risk of oxidative stress-related pathologies. The principal aim was to evaluate if supplementation with long-chain polyunsaturated fatty acids (LCPUFAs) improves antioxidant defenses. In total, 21 preterm infants were supplemented with ARA and DHA in a 2:1 ratio (ARA:DHA-S) or with medium-chain triglycerides (MCT-S). Plasma n-3 and n-6 LCPUFAs were measured at birth, postnatal day 28, and 36 weeks of postmenstrual age (36 WPA) by gas chromatography–mass spectroscopy. Plasma antioxidants (glutathione (GSH), catalase, and thiols) and oxidative damage biomarkers (malondialdehyde (MDA), carbonyls) were analyzed at the same time points by spectrophotometry, and scores of antioxidant status (Antiox-S) and oxidative damage (Proxy-S) were calculated. At 36 WPA, linoleic acid (LA) and dihomo-γ-linolenic acid (DGLA) were decreased in ARA:DHA-S compared to the MCT-S group (LA: ARA:DHA-S = 18.54 ± 1.68, MCT-S = 22.80 ± 1.41; p = 0.018; DGLA: ARA:DHA-S = 1.68 ± 0.38, MCT-S = 2.32 ± 0.58; p = 0.018). Furthermore, α-linolenic acid (ALA) was increased in ARA:DHA-S (ARA:DHA-S = 0.52 ± 0.33, MCT-S = 0.22 ± 0.10; p = 0.018). Additionally, LA:DHA ratio was decreased in the ARA:DHA-S compared to control group (ARA:DHA-S = 6.26 ± 2.35, MCT-S = 8.21 ± 2.65; p = 0.045). By the end of supplementation (36 WPA), catalase, thiol groups, and Antiox-S were significantly higher in neonates receiving ARA:DHA-S compared to those receiving MCT-S, with no differences in oxidative stress biomarkers. In conclusion, ARA:DHA supplementation in preterm neonates resulted in an overall improvement in antioxidant to oxidant balance and a decrease in early fatty acid precursors of the n-6 relative to the n-3 pathway. These effects may reduce oxidative stress and inflammation.
Collapse
|
40
|
Abstract
In the absence of effective interventions to prevent preterm births, improved survival of infants who are born at the biological limits of viability has relied on advances in perinatal care over the past 50 years. Except for extremely preterm infants with suboptimal perinatal care or major antenatal events that cause severe respiratory failure at birth, most extremely preterm infants now survive, but they often develop chronic lung dysfunction termed bronchopulmonary dysplasia (BPD; also known as chronic lung disease). Despite major efforts to minimize injurious but often life-saving postnatal interventions (such as oxygen, mechanical ventilation and corticosteroids), BPD remains the most frequent complication of extreme preterm birth. BPD is now recognized as the result of an aberrant reparative response to both antenatal injury and repetitive postnatal injury to the developing lungs. Consequently, lung development is markedly impaired, which leads to persistent airway and pulmonary vascular disease that can affect adult lung function. Greater insights into the pathobiology of BPD will provide a better understanding of disease mechanisms and lung repair and regeneration, which will enable the discovery of novel therapeutic targets. In parallel, clinical and translational studies that improve the classification of disease phenotypes and enable early identification of at-risk preterm infants should improve trial design and individualized care to enhance outcomes in preterm infants.
Collapse
|
41
|
Matyas M, Hasmasanu MG, Zaharie G. Antioxidant Capacity of Preterm Neonates Assessed by Hydrogen Donor Value. MEDICINA (KAUNAS, LITHUANIA) 2019; 55:E720. [PMID: 31671553 PMCID: PMC6915410 DOI: 10.3390/medicina55110720] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Revised: 10/21/2019] [Accepted: 10/22/2019] [Indexed: 12/28/2022]
Abstract
Background and objectives: Premature newborns have a number of oxidative stress-inducing disorders. Antioxidant defense is deficient in premature newborns. Hydrogen donors can be used to evaluate the non-enzymatic antioxidant defense. By measuring hydrogen donors, a group of antioxidants can be assessed: tocopherol, ascorbic acid, and glutathione. These represent the most relevant group of non-enzymatic antioxidants. The main aim of this study was to evaluate the non-enzymatic antioxidant defense capacity of premature newborns by measuring hydrogen donors. Materials and Methods: We evaluated the non-enzymatic antioxidant capacity by hydrogen donor measurement in 24 premature newborns with various oxidative stress-inducing disorders and in 14 premature newborns without oxidative stress-inducing conditions. Statistical analysis was performed using the Statistica program (v. 8, StatSoft, Round Rock, TX, USA). Differences between groups were tested with Wilcoxon matched test for quantitative paired data or Mann-Whitney test for quantitative independent data. The Z test for proportions was used to compare qualitative data among subgroups. Results: Hydrogen donors in the study group had a significantly lower value on the first day of life compared to the value of the control group. Also, the hydrogen donor value in the study group was significantly lower on the first day compared to the third day of life (p < 0.05). Neonates with mild respiratory distress (14 cases) had increased hydrogen donor values on their third day of life compared to the first day of life. Conclusions: The antioxidant capacity is influenced by oxidative stress-inducing disorders. Respiratory distress influenced the hydrogen donor value and antioxidant defense. Antioxidant defense gradually improves after birth according to gestational age.
Collapse
Affiliation(s)
- Melinda Matyas
- Neonatology Department, University of Medicine and Pharmacy, Cluj-Napoca, 400006, str. V. Babes no. 8, Romania.
- 1st Neonatology Department, County Emergency Hospital Cluj-Napoca, 400006, str. Clinicilor no. 3-5, Romania.
| | - Monica G Hasmasanu
- Neonatology Department, University of Medicine and Pharmacy, Cluj-Napoca, 400006, str. V. Babes no. 8, Romania.
- 1st Neonatology Department, County Emergency Hospital Cluj-Napoca, 400006, str. Clinicilor no. 3-5, Romania.
| | - Gabriela Zaharie
- Neonatology Department, University of Medicine and Pharmacy, Cluj-Napoca, 400006, str. V. Babes no. 8, Romania.
- 1st Neonatology Department, County Emergency Hospital Cluj-Napoca, 400006, str. Clinicilor no. 3-5, Romania.
| |
Collapse
|
42
|
Cai C, Zhang Z, Morales M, Wang Y, Khafipour E, Friel J. Feeding practice influences gut microbiome composition in very low birth weight preterm infants and the association with oxidative stress: A prospective cohort study. Free Radic Biol Med 2019; 142:146-154. [PMID: 30851363 DOI: 10.1016/j.freeradbiomed.2019.02.032] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Revised: 02/23/2019] [Accepted: 02/28/2019] [Indexed: 12/12/2022]
Abstract
Knowledge about the development of the preterm infant gut microbiota is emerging and is critical to their health. Very-low-birth-weight (VLBW; birth weight, <1500 g) infants usually have special dietary needs while showing increased oxidative stress related to intensive care. This prospective cohort study assessed the effect of feeding practice on gut microbiome development and oxidative stress in preterm infants. Fecal samples were collected from each infant in the early (1-2 weeks of enteral feeding) and late (2-4 weeks of enteral feeding) feeding stages. We performed high-throughput sequencing of V3-V4 regions of the 16S rRNA gene to analyze the fecal microbiome composition of 20 VLBW preterm infants and to determine the association of gut bacterial composition with feeding practice using an oxidative stress marker (urinary F2-isoprostane). Our results showed that feeding practices in the late stage significantly influenced the gut microbiome composition and oxidative stress in preterm infants. Preterm infants fed human milk + human milk fortifier and only formula diets showed a significant increase in F2-isoprostane levels (P < 0.05) compared with those fed human milk + formula diet. The gut microbiome of the infants fed the human milk + Human milk fortifier diet showed the lower relative abundance of Veillonella (P < 0.05) compared with that of the infants fed the human milk + formula diet. The gut microbiome of the infants fed the only formula diet showed the lowest microbial diversity and the highest relative abundance of Terrisporobacter (P < 0.05) compared with the gut microbiome of the infants fed the other diets. Correlation network analysis showed that urinary F2-isoprostane level was positively correlated with Terrisporobacter and Enterobacteriaceae abundance (P < 0.05) in the preterm infants. In conclusion, these data suggest that feeding practice affects the bacterial diversity and composition in the gut microbiome and is associated with oxidative stress in VLBW preterm infants.
Collapse
Affiliation(s)
- Chenxi Cai
- Department of Food and Human Nutritional Sciences, Faculty of Agricultural and Food Sciences, 208A Human Ecology Building, University of Manitoba, Winnipeg, MB, R3T 2N2, Canada.
| | - Zhengxiao Zhang
- Department of Animal Science, Faculty of Agricultural and Food Sciences, 208A Human Ecology Building, University of Manitoba, Winnipeg, MB, R3T 2N2, Canada.
| | - Maria Morales
- Department of Food and Human Nutritional Sciences, Faculty of Agricultural and Food Sciences, 208A Human Ecology Building, University of Manitoba, Winnipeg, MB, R3T 2N2, Canada.
| | - Yanan Wang
- Department of Animal Science, Faculty of Agricultural and Food Sciences, 208A Human Ecology Building, University of Manitoba, Winnipeg, MB, R3T 2N2, Canada.
| | - Ehsan Khafipour
- Department of Animal Science, Faculty of Agricultural and Food Sciences, 208A Human Ecology Building, University of Manitoba, Winnipeg, MB, R3T 2N2, Canada
| | - James Friel
- Department of Food and Human Nutritional Sciences, Faculty of Agricultural and Food Sciences, 208A Human Ecology Building, University of Manitoba, Winnipeg, MB, R3T 2N2, Canada.
| |
Collapse
|
43
|
Singh-Mallah G, Nair S, Sandberg M, Mallard C, Hagberg H. The Role of Mitochondrial and Endoplasmic Reticulum Reactive Oxygen Species Production in Models of Perinatal Brain Injury. Antioxid Redox Signal 2019; 31:643-663. [PMID: 30957515 PMCID: PMC6657303 DOI: 10.1089/ars.2019.7779] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Revised: 04/01/2019] [Accepted: 04/03/2019] [Indexed: 12/20/2022]
Abstract
Significance: Perinatal brain injury is caused by hypoxia-ischemia (HI) in term neonates, perinatal arterial stroke, and infection/inflammation leading to devastating long-term neurodevelopmental deficits. Therapeutic hypothermia is the only currently available treatment but is not successful in more than 50% of term neonates suffering from hypoxic-ischemic encephalopathy. Thus, there is an urgent unmet need for alternative or adjunct therapies. Reactive oxygen species (ROS) are important for physiological signaling, however, their overproduction/accumulation from mitochondria and endoplasmic reticulum (ER) during HI aggravate cell death. Recent Advances and Critical Issues: Mechanisms underlying ER stress-associated ROS production have been primarily elucidated using either non-neuronal cells or adult neurodegenerative experimental models. Findings from mature brain cannot be simply transferred to the immature brain. Therefore, age-specific studies investigating ER stress modulators may help investigate ER stress-associated ROS pathways in the immature brain. New therapeutics such as mitochondrial site-specific ROS inhibitors that selectively inhibit superoxide (O2•-)/hydrogen peroxide (H2O2) production are currently being developed. Future Directions: Because ER stress and oxidative stress accentuate each other, a combinatorial therapy utilizing both antioxidants and ER stress inhibitors may prove to be more protective against perinatal brain injury. Moreover, multiple relevant targets need to be identified for targeting ROS before they are formed. The role of organelle-specific ROS in brain repair needs investigation. Antioxid. Redox Signal. 31, 643-663.
Collapse
Affiliation(s)
- Gagandeep Singh-Mallah
- Institute of Biomedicine, Department of Medical Biochemistry, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Centre of Perinatal Medicine and Health, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Syam Nair
- Centre of Perinatal Medicine and Health, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Institute of Neuroscience and Physiology, Department of Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Institute of Clinical Sciences, Department of Obstetrics and Gynecology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Mats Sandberg
- Institute of Biomedicine, Department of Medical Biochemistry, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Centre of Perinatal Medicine and Health, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Carina Mallard
- Centre of Perinatal Medicine and Health, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Institute of Neuroscience and Physiology, Department of Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Henrik Hagberg
- Centre of Perinatal Medicine and Health, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Institute of Clinical Sciences, Department of Obstetrics and Gynecology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
44
|
Abstract
Fetal development occurs in a relatively hypoxemic environment, and birth represents significant oxidative stress. Premature infants are disadvantaged by a lack of maternal antioxidant transfer and impaired endogenous antioxidant responses. O2 metabolism is essential for life and its biochemical reactions are dynamic, compartmentalized, and difficult to characterize in vivo. There is a growing appreciation for the role of reactive oxygen species in nonpathologic processes, including regulation of cell signaling and mitochondrial function. There are several gaps in the knowledge about the role of reactive oxygen species in normal development and how oxidative stress alters normal signaling and subsequent development.
Collapse
Affiliation(s)
- Trent E Tipple
- Division of Neonatology, Department of Pediatrics, The University of Alabama at Birmingham, 176 F Suite 9380, 619 19th Street South, Birmingham, AL 35249-7335, USA.
| | - Namasivayam Ambalavanan
- Division of Neonatology, Department of Pediatrics, The University of Alabama at Birmingham, 176 F Suite 9380, 619 19th Street South, Birmingham, AL 35249-7335, USA
| |
Collapse
|
45
|
Kicinski P, Malachowska B, Wyka K, Gach A, Jakubowski L, Gulczynska E. The level of extracellular superoxide dismutase in the first week of life in very and extremely low birth weight infants and the risk of developing bronchopulmonary dysplasia. J Perinat Med 2019; 47:671-676. [PMID: 31365347 DOI: 10.1515/jpm-2018-0418] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Accepted: 03/27/2019] [Indexed: 11/15/2022]
Abstract
Background Antioxidant enzymes may play a significant role in the development of bronchopulmonary dysplasia (BPD). The aim of the study was to assess the relationship between the level of extracellular superoxide dismutase (SOD3) in the serum at days 1 and 7 of life and the risk of developing BPD. Methods The study comprised 103 neonates born before 32 weeks' gestation with a birth weight of ≤1500 g. Results In the investigated group, the median serum SOD3 level at day 1 of life was 4.01 ng/mL [interquartile range (IQR) 2.59-5.09 ng/mL] and at day 7 of life 3.13 ng/mL (IQR 2.49-4.34 ng/mL). A statistically significant decrease in the serum SOD3 level was found in the first week of life, P < 0.0001. No correlation was found between the serum SOD3 level at day 1 of life and gestational age R = 0.07, P = 0.4543 and birth weight R = 0.10, P = 0.3083. No statistically significant correlation was found between the dynamics of change in the SOD3 level in serum at days 1 and 7 of life and the risk of BPD development for the definition of BPD at day 28 of life, P = 0.8764 nor at 36 weeks' postmenstrual age, P = 0.6598. Conclusion The study revealed a statistically significant decrease in the serum SOD3 level in the first week of life in very and extremely low birth weight infants born before 32 weeks of gestation. In the clinical setting, no relationship was observed between the level of SOD3 in serum and the risk of developing BPD.
Collapse
Affiliation(s)
- Przemyslaw Kicinski
- Department of Neonatology, Polish Mother's Memorial Hospital - Research Institute, 281/289 Rzgowska Street, 93-338 Lodz, Poland.,Department of Angiology, Medical University of Lodz, Lodz, Poland, Tel.: +48 42 2711041
| | - Beata Malachowska
- Department of Biostatistics and Translational Medicine, Medical University of Lodz, Lodz, Poland
| | - Krystyna Wyka
- Department of Pediatrics, Oncology, Hematology and Diabetology, Medical University of Lodz, Lodz, Poland
| | - Agnieszka Gach
- Department of Medical Genetics, Polish Mother's Memorial Hospital - Research Institute, Lodz, Poland
| | - Lucjusz Jakubowski
- Department of Medical Genetics, Polish Mother's Memorial Hospital - Research Institute, Lodz, Poland
| | - Ewa Gulczynska
- Department of Neonatology, Polish Mother's Memorial Hospital - Research Institute, Lodz, Poland
| |
Collapse
|
46
|
Bhatia S, Drake DM, Miller L, Wells PG. Oxidative stress and DNA damage in the mechanism of fetal alcohol spectrum disorders. Birth Defects Res 2019; 111:714-748. [PMID: 31033255 DOI: 10.1002/bdr2.1509] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Revised: 03/07/2019] [Accepted: 03/14/2019] [Indexed: 12/18/2022]
Abstract
This review covers molecular mechanisms involving oxidative stress and DNA damage that may contribute to morphological and functional developmental disorders in animal models resulting from exposure to alcohol (ethanol, EtOH) in utero or in embryo culture. Components covered include: (a) a brief overview of EtOH metabolism and embryopathic mechanisms other than oxidative stress; (b) mechanisms within the embryo and fetal brain by which EtOH increases the formation of reactive oxygen species (ROS); (c) critical embryonic/fetal antioxidative enzymes and substrates that detoxify ROS; (d) mechanisms by which ROS can alter development, including ROS-mediated signal transduction and oxidative DNA damage, the latter of which leads to pathogenic genetic (mutations) and epigenetic changes; (e) pathways of DNA repair that mitigate the pathogenic effects of DNA damage; (f) related indirect mechanisms by which EtOH enhances risk, for example by enhancing the degradation of some DNA repair proteins; and, (g) embryonic/fetal pathways like NRF2 that regulate the levels of many of the above components. Particular attention is paid to studies in which chemical and/or genetic manipulation of the above mechanisms has been shown to alter the ability of EtOH to adversely affect development. Alterations in the above components are also discussed in terms of: (a) individual embryonic and fetal determinants of risk and (b) potential risk biomarkers and mitigating strategies. FASD risk is likely increased in progeny which/who are biochemically predisposed via genetic and/or environmental mechanisms, including enhanced pathways for ROS formation and/or deficient pathways for ROS detoxification or DNA repair.
Collapse
Affiliation(s)
- Shama Bhatia
- Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada.,Centre for Pharmaceutical Oncology, University of Toronto, Toronto, Ontario, Canada
| | - Danielle M Drake
- Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada.,Centre for Pharmaceutical Oncology, University of Toronto, Toronto, Ontario, Canada
| | | | - Peter G Wells
- Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada.,Centre for Pharmaceutical Oncology, University of Toronto, Toronto, Ontario, Canada.,Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
47
|
Vishala T, Pitchaiah G, Pravadha D, Annapurna A. Effect of plain and fortified amla fruit powder on aluminum-induced Alzheimer's Disease in Wistar Rats. Pharmacognosy Res 2019. [DOI: 10.4103/pr.pr_17_17] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
48
|
Laforgia N, Di Mauro A, Favia Guarnieri G, Varvara D, De Cosmo L, Panza R, Capozza M, Baldassarre ME, Resta N. The Role of Oxidative Stress in the Pathomechanism of Congenital Malformations. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:7404082. [PMID: 30693064 PMCID: PMC6332879 DOI: 10.1155/2018/7404082] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Revised: 08/20/2018] [Accepted: 10/30/2018] [Indexed: 02/07/2023]
Abstract
Congenital anomalies are significant causes of mortality and morbidity in infancy and childhood. Embryogenesis requires specific signaling pathways to regulate cell proliferation and differentiation. These signaling pathways are sensitive to endogenous and exogenous agents able to produce several structural changes of the developing fetus. Oxidative stress, due to an imbalance between the production of reactive oxygen species and antioxidant defenses, disrupts signaling pathways with a causative role in birth defects. This review provides a basis for understanding the role of oxidative stress in the pathomechanism of congenital malformations, discussing the mechanisms related to some congenital malformations. New insights in the knowledge of pathomechanism of oxidative stress-related congenital malformations, according to experimental and human studies, represent the basis of possible clinical applications in screening, prevention, and therapies.
Collapse
Affiliation(s)
- Nicola Laforgia
- Neonatology and Neonatal Intensive Care Unit, Department of Biomedical Science and Human Oncology, “Aldo Moro” University of Bari, Policlinico Hospital-Piazza Giulio Cesare n. 11, 70124 Bari, Italy
| | - Antonio Di Mauro
- Neonatology and Neonatal Intensive Care Unit, Department of Biomedical Science and Human Oncology, “Aldo Moro” University of Bari, Policlinico Hospital-Piazza Giulio Cesare n. 11, 70124 Bari, Italy
| | - Giovanna Favia Guarnieri
- Neonatology and Neonatal Intensive Care Unit, Department of Biomedical Science and Human Oncology, “Aldo Moro” University of Bari, Policlinico Hospital-Piazza Giulio Cesare n. 11, 70124 Bari, Italy
| | - Dora Varvara
- Medical Genetics Unit, Department of Biomedical Sciences and Human Oncology, “Aldo Moro” University of Bari, Policlinico Hospital-Piazza Giulio Cesare n. 11, 70124 Bari, Italy
| | - Lucrezia De Cosmo
- Neonatology and Neonatal Intensive Care Unit, Department of Biomedical Science and Human Oncology, “Aldo Moro” University of Bari, Policlinico Hospital-Piazza Giulio Cesare n. 11, 70124 Bari, Italy
| | - Raffaella Panza
- Neonatology and Neonatal Intensive Care Unit, Department of Biomedical Science and Human Oncology, “Aldo Moro” University of Bari, Policlinico Hospital-Piazza Giulio Cesare n. 11, 70124 Bari, Italy
| | - Manuela Capozza
- Neonatology and Neonatal Intensive Care Unit, Department of Biomedical Science and Human Oncology, “Aldo Moro” University of Bari, Policlinico Hospital-Piazza Giulio Cesare n. 11, 70124 Bari, Italy
| | - Maria Elisabetta Baldassarre
- Neonatology and Neonatal Intensive Care Unit, Department of Biomedical Science and Human Oncology, “Aldo Moro” University of Bari, Policlinico Hospital-Piazza Giulio Cesare n. 11, 70124 Bari, Italy
| | - Nicoletta Resta
- Medical Genetics Unit, Department of Biomedical Sciences and Human Oncology, “Aldo Moro” University of Bari, Policlinico Hospital-Piazza Giulio Cesare n. 11, 70124 Bari, Italy
| |
Collapse
|
49
|
Therapeutic and preventative effects of ankaferd blood stopper in an experimental necrotizing enterocolitis model. Biomed Pharmacother 2018; 110:105-110. [PMID: 30465999 DOI: 10.1016/j.biopha.2018.11.023] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Revised: 11/06/2018] [Accepted: 11/06/2018] [Indexed: 02/02/2023] Open
Abstract
Necrotizing enterocolitis (NEC) is a major neonatal health problem that especially affects preterm infants and causes severe morbidity and mortality. Although its pathogenesis is not fully understood, important risk factors include prematurity, oxidative stress, inflammation, and apoptosis. Ankaferd Blood Stopper® (ABS) has antioxidant, antiinflammatory, antimicrobial, antiapoptotic, and wound healing accelerant properties. In this study, we aimed to investigate whether treatment with ABS reduced the severity of NEC in rat pups in an experimental NEC model. Thirty-six newborn Wistar albino rat pups were randomly assigned to the control, NEC + saline, or NEC + ABS groups. NEC was induced by intraperitoneal injection of lipopolysaccharide, feeding with hyperosmolar enteral formula, and exposure to hypoxia/hyperoxia and cold stress. ABS was administered intraperitoneally to the pups in the NEC + ABS group daily starting on day 1 of the study at a dose of 2 ml/kg by diluting 2 ml with saline at a ratio of 1:3. All pups were sacrificed on day 4. The terminal ileum including the proximal colon was removed for histopathological and immunohistochemical examination and biochemical analysis. Macroscopic assessment and intestinal injury scores were lower in NEC + ABS group compared to the NEC + saline group (p < 0.05). Immunohistochemical evaluations of caspase-3, -8, and -9 revealed significantly reduced apoptosis in the NEC + ABS group compared to the NEC + saline group (p = 0.001). Total oxidant status, oxidative stress index, tumor necrosis factor α and interleukin-1β levels, and lipid, protein, and deoxyribonucleic acid oxidation products were significantly lower in the NEC + ABS group compared to NEC + saline group (p < 0.001 for all), while total antioxidant status, glutathione, and superoxide dismutase levels were higher in the NEC + ABS group (p < 0.001, p < 0.001, p = 0.01, respectively). ABS treatment has the potential to effectively reduce the severity of intestinal damage in NEC due to its antioxidant, antiinflammatory, and antiapoptotic properties. Therefore, NEC may be an alternative option for treatment.
Collapse
|
50
|
Cakir U, Tayman C, Serkant U, Yakut HI, Cakir E, Ates U, Koyuncu I, Karaogul E. Ginger (Zingiber officinale Roscoe) for the treatment and prevention of necrotizing enterocolitis. JOURNAL OF ETHNOPHARMACOLOGY 2018; 225:297-308. [PMID: 30005955 DOI: 10.1016/j.jep.2018.07.009] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/19/2018] [Revised: 07/02/2018] [Accepted: 07/09/2018] [Indexed: 06/08/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Necrotizing enterocolitis (NEC) is the most important gastrointestinal emergency affecting especially preterm infants and causes severe morbidities and mortality. However, there is no cure. Oxidant stress, inflammation, apoptosis, as well as prematurity are believed to responsible in the pathogenesis of the disease. Ginger and its compounds have anti-inflammatory, antimicrobial, anti-oxidant properties and immunomodulatory, cytoprotective/regenerative actions. AIM OF THE STUDY This study aimed to evaluate the beneficial effects of ginger on the intestinal damage in an experimental rat model of NEC. MATERIALS AND METHODS Thirty newborn Wistar rats were divided into three groups: NEC, NEC + ginger and control in this experimental study. NEC was induced by injection of intraperitoneal lipopolysaccharide, feeding with enteral formula, hypoxia-hyperoxia and cold stress exposure. The pups in the NEC + ginger group were orally administered ginger at a dose of 1000 mg/kg/day. Proximal colon and ileum were excised. Histopathological, immunohistochemical (TUNEL for apoptosis, caspase 3 and 8) and biochemical assays including xanthine oxidase (XO), superoxide dismutase (SOD), glutathione peroxidase (GSH-Px), malonaldehyde (MDA) and myeloperoxidase (MPO), tumor necrosis factor-α (TNF-α), interleukin1β (IL-1β), and interleukin 6 (IL-6) activity were evaluated. RESULTS Compared with the NEC group, the rat pups in the NEC + ginger group had better clinical disease scores and weight gain (p < 0.05). Macroscopic evaluation, Histopathologic and apoptosis assessment (TUNEL, caspase 3 and 8) releaved that severity of intestinal damage were significantly lower in the NEC + ginger group (p < 0.05). The levels of TNF-α, IL-1β and IL-6 in the ginger treated group were significantly decreased (P < 0.05). The GSH-Px and SOD levels of the ginger treated group were significantly preserved in the NEC + ginger group (p < 0.05). The tissue XO, MDA and MPO levels of the NEC + ginger group were significantly lower than those in the NEC group (P < 0.05). CONCLUSION Ginger therapy efficiently ameliorated the severity of intestinal damage in NEC and may be a promising treatment option.
Collapse
Affiliation(s)
- Ufuk Cakir
- Department of Neonatology, Health Sciences University, Zekai Tahir Burak Maternity Education and Research Hospital, Ankara, Turkey.
| | - Cuneyt Tayman
- Health Sciences University, Zekai Tahir Burak Maternity Education and Research Hospital, Ankara, Turkey.
| | - Utku Serkant
- Department of Biochemistry, Golbası Public Hospital, Ankara, Turkey.
| | - Halil Ibrahim Yakut
- Department of Pediatrics, Health Sciences University, Ankara Hematology Oncology Children Education and Research Hospital, Ankara, Turkey.
| | - Esra Cakir
- Health Sciences University, Anesthesiology and Clinical of Critical Care, Ankara Numune Education and Research Hospital, Ankara, Turkey.
| | - Ufuk Ates
- Department of Pediatric Surgery, Ankara University Faculty of Medicine, Ankara, Turkey.
| | - Ismail Koyuncu
- Harran University Faculty of Medicine Department of Biochemistry, Sanlıurfa, Turkey.
| | - Eyyup Karaogul
- Harran University Engineering Faculty Food Science and Technology, Sanlıurfa, Turkey.
| |
Collapse
|