1
|
Juliá-Burchés C, Martínez-Varea A, Morales-Roselló J, Diago-Almela V. Impact of Maternal Smoking on Obstetric and Neonatal Outcomes in Twin Pregnancies: A Narrative Review. J Clin Med 2024; 13:7329. [PMID: 39685788 DOI: 10.3390/jcm13237329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 11/23/2024] [Accepted: 11/30/2024] [Indexed: 12/18/2024] Open
Abstract
Maternal smoking, including both traditional cigarettes and electronic ones, is a significant modifiable risk factor associated with adverse perinatal outcomes, especially in twin pregnancies. This narrative review aims to explore the impact of maternal smoking on obstetric and neonatal outcomes in twin pregnancies, which inherently carry a higher risk of complications. A literature search was conducted using the PubMed and EMBASE databases, selecting studies published between January 1994 and October 2024. The findings demonstrate a clear association between smoking and increased risks of preterm birth and fetal growth restriction (FGR) in twin pregnancies. These risks are exacerbated when smoking is combined with other factors, such as preeclampsia and elevated body mass index (BMI). Smoking was also associated with long-term post-natal complications, including respiratory problems like asthma, as well as cognitive and behavioral disorders. However, an association with preeclampsia was not found, and further studies are needed to clarify the relationship in the fields of preterm premature rupture of membranes (PPROM) and fetal death. The adverse effects of smoking are primarily due to reduced oxygen supply to the fetus, caused by nicotine-induced vasoconstriction and carbon monoxide exposure, leading to placental insufficiency and fetal hypoxia. These effects are amplified in twin pregnancies due to the increased physiological demands. The review highlights that smoking cessation interventions during pregnancy are crucial to mitigate these risks and improve maternal and neonatal health outcomes.
Collapse
Affiliation(s)
- Cristina Juliá-Burchés
- Department of Obstetrics and Gynecology, La Fe University and Polytechnic Hospital, 46026 Valencia, Spain
| | - Alicia Martínez-Varea
- Department of Obstetrics and Gynecology, La Fe University and Polytechnic Hospital, 46026 Valencia, Spain
- Department of Medicine, CEU Cardenal Herrera University, 12006 Castellón de la Plana, Spain
- Faculty of Health Sciences, Universidad Internacional de Valencia, 46026 Valencia, Spain
| | - José Morales-Roselló
- Department of Obstetrics and Gynecology, La Fe University and Polytechnic Hospital, 46026 Valencia, Spain
- Department of Pediatrics, Obstetrics and Gynecology, Faculty of Medicine, University of Valencia, 12006 Valencia, Spain
| | - Vicente Diago-Almela
- Department of Obstetrics and Gynecology, La Fe University and Polytechnic Hospital, 46026 Valencia, Spain
- Department of Pediatrics, Obstetrics and Gynecology, Faculty of Medicine, University of Valencia, 12006 Valencia, Spain
| |
Collapse
|
2
|
Park SY, Im JA, Kim JY. Exploring the Effect of Deep-Sea Water on the Therapeutic Potential of the Anti-Inflammatory Response in an Indomethacin-Induced Gastric Ulcer Rat Model. Int J Mol Sci 2023; 24:17430. [PMID: 38139257 PMCID: PMC10743565 DOI: 10.3390/ijms242417430] [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: 11/14/2023] [Revised: 12/05/2023] [Accepted: 12/08/2023] [Indexed: 12/24/2023] Open
Abstract
Gastric ulcers are often exacerbated by factors such as nonsteroidal anti-inflammatory drugs (NSAIDs) and inflammation, and they have a substantial impact on a significant portion of the population. Notably, indomethacin is recognized as a prominent contributor to ulcers. This study investigated this potential method, with normalization to the anti-inflammatory and antiulcer properties of deep-sea water (DSW)-derived mineral water, using an indomethacin-induced gastric ulcer model in rats. The study involved four groups (n = 6 rats/group): normal control group (CON), indomethacin-only group (IND), indomethacin with trace mineral water group (TM), and indomethacin with high magnesium low sodium water group (HMLS). For three weeks, the CON and IND groups consumed tap water, while the TM and HMLS groups had access to mineral water. Gastric ulcers were induced on the final day using indomethacin, for all groups except the CON group. The results demonstrated that HMLS intake significantly improved gastric mucosal damage, preserved mucin stability, and increased gastric thickness, indicating its potential to prevent and alleviate indomethacin-induced gastric ulcers. Furthermore, HMLS consumption led to the upregulation of key genes associated with inflammation and a reduction in inflammatory cytokines. These findings suggest that DSW-derived mineral water, and particularly its high Mg2+ content, may offer promising health benefits including anti-inflammatory and anti-ulcer properties.
Collapse
Affiliation(s)
- Soo-yeon Park
- Department of Food Science and Technology, Seoul National University of Science and Technology, Seoul 01811, Republic of Korea; (S.-y.P.); (J.A.I.)
| | - Jin A Im
- Department of Food Science and Technology, Seoul National University of Science and Technology, Seoul 01811, Republic of Korea; (S.-y.P.); (J.A.I.)
| | - Ji Yeon Kim
- Department of Food Science and Technology, Seoul National University of Science and Technology, Seoul 01811, Republic of Korea; (S.-y.P.); (J.A.I.)
- Department of Nano Bio Engineering, Seoul National University of Science and Technology, Seoul 01811, Republic of Korea
| |
Collapse
|
3
|
Srnovršnik T, Virant-Klun I, Pinter B. Heavy Metals and Essential Elements in Association with Oxidative Stress in Women with Polycystic Ovary Syndrome-A Systematic Review. Antioxidants (Basel) 2023; 12:1398. [PMID: 37507937 PMCID: PMC10376316 DOI: 10.3390/antiox12071398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 07/04/2023] [Accepted: 07/06/2023] [Indexed: 07/30/2023] Open
Abstract
Altered levels of heavy metals and essential elements have been associated with oxidative stress (OS) and metabolic and hormonal changes in women with polycystic ovary syndrome (PCOS). We aimed to summarize the knowledge on the association of heavy metals and essential elements with OS in PCOS. An electronic literature search using PubMed for studies published between January 2008 and April 2023 was conducted. We evaluated heavy metals and essential elements in relation to OS in PCOS in 15 articles. PCOS women had increased antimonium (Sb), cadmium (Cd), lead (Pb), mercury (Hg), arsenic (As), tellurium (Te), thallium (Tl) and osmium (Os) blood levels and decreased zinc (Zn) blood levels; the results of copper (Cu) blood levels were conflicting. Some studies showed a significant correlation between heavy metals (Sb, Cd, Pb, Hg, As, Te and Tl) and essential elements (Se, Zn, Cr, Ca, Mg and Cu) and markers of OS and chronic inflammation. Heavy metals (Sb, Cd, Pb and Hg) and essential elements (Zn, Cr, Se, Ca, Mg and Cu) were associated with metabolic and hormonal characteristics in PCOS. There might be a possible benefit from supplementation therapy in reducing OS and endocrinological problems related to PCOS. Our review confirmed an association between heavy metals and essential elements with OS in PCOS women. This systematic review is registered in PROSPERO under number CRD42023418453.
Collapse
Affiliation(s)
- Tinkara Srnovršnik
- Division for Women's Healthcare-Šiška Unit, Community Health Centre Ljubljana, Metelkova Ulica 9, 1000 Ljubljana, Slovenia
- Faculty of Medicine, University of Ljubljana, Vrazov Trg 2, 1000 Ljubljana, Slovenia
| | - Irma Virant-Klun
- Clinical Research Centre, University Medical Centre Ljubljana, Vrazov Trg 1, 1000 Ljubljana, Slovenia
| | - Bojana Pinter
- Faculty of Medicine, University of Ljubljana, Vrazov Trg 2, 1000 Ljubljana, Slovenia
- Division of Obstetrics and Gynecology, University Medical Centre Ljubljana, Šlajmerjeva 3, 1000 Ljubljana, Slovenia
| |
Collapse
|
4
|
Valenzuela I, Kinoshita M, van der Merwe J, Maršál K, Deprest J. Prenatal interventions for fetal growth restriction in animal models: A systematic review. Placenta 2022; 126:90-113. [PMID: 35796064 DOI: 10.1016/j.placenta.2022.06.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 05/20/2022] [Accepted: 06/17/2022] [Indexed: 12/09/2022]
Abstract
Fetal growth restriction (FGR) in human pregnancy is associated with perinatal mortality, short- and long-term morbidities. No prenatal therapy is currently established despite decades of research. We aimed to review interventions in animal models for prenatal FGR treatment, and to seek the next steps for an effective clinical therapy. We registered our protocol and searched MEDLINE, Embase, and The Cochrane Library with no language restrictions, in accordance with the PRISMA guideline. We included all studies that reported the effects of any prenatal intervention in animal models of induced FGR. From 3257 screened studies, 202 describing 237 interventions were included for the final synthesis. Mice and rats were the most used animals (79%) followed by sheep (16%). Antioxidants (23%), followed by vasodilators (18%), nutrients (14%), and immunomodulators (12%) were the most tested therapy. Two-thirds of studies only reported delivery or immediate neonatal outcomes. Adverse effects were rarely reported (11%). Most studies (73%), independent of the intervention, showed a benefit in fetal survival or birthweight. The risk of bias was high, mostly due to the lack of randomization, allocation concealment, and blinding. Future research should aim to describe both short- and long-term outcomes across various organ systems in well-characterized models. Further efforts must be made to reduce selection, performance, and detection bias.
Collapse
|
5
|
Zarate MA, De Dios RK, Balasubramaniyan D, Zheng L, Sherlock LG, Rozance PJ, Wright CJ. The Acute Hepatic NF-κB-Mediated Proinflammatory Response to Endotoxemia Is Attenuated in Intrauterine Growth-Restricted Newborn Mice. Front Immunol 2021; 12:706774. [PMID: 34539638 PMCID: PMC8440955 DOI: 10.3389/fimmu.2021.706774] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2021] [Accepted: 08/13/2021] [Indexed: 11/13/2022] Open
Abstract
Intrauterine growth restriction (IUGR) is a relevant predictor for higher rates of neonatal sepsis worldwide and is associated with an impaired neonatal immunity and lower immune cell counts. During the perinatal period, the liver is a key immunological organ responsible for the nuclear factor kappa B (NF-κB)-mediated innate immune response to inflammatory stimuli, but whether this role is affected by IUGR is unknown. Herein, we hypothesized that the newborn liver adapts to calorie-restriction IUGR by inducing changes in the NF-κB signaling transcriptome, leading to an attenuated acute proinflammatory response to intraperitoneal lipopolysaccharide (LPS). We first assessed the hepatic gene expression of key NF-κB factors in the IUGR and normally grown (NG) newborn mice. Real-time quantitative PCR (RT-qPCR) analysis revealed an upregulation of both IκB proteins genes (Nfkbia and Nfkbib) and the NF-κB subunit Nfkb1 in IUGR vs. NG. We next measured the LPS-induced hepatic expression of acute proinflammatory genes (Ccl3, Cxcl1, Il1b, Il6, and Tnf) and observed that the IUGR liver produced an attenuated acute proinflammatory cytokine gene response (Il1b and Tnf) to LPS in IUGR vs. unexposed (CTR). Consistent with these results, LPS-exposed hepatic tumor necrosis factor alpha (TNF-α) protein concentrations were lower in IUGR vs. LPS-exposed NG and did not differ from IUGR CTR. Sex differences at the transcriptome level were observed in the IUGR male vs. female. Our results demonstrate that IUGR induces key modifications in the NF-κB transcriptomic machinery in the newborn that compromised the acute proinflammatory cytokine gene and protein response to LPS. Our results bring novel insights in understanding how the IUGR newborn is immunocompromised due to fundamental changes in NF-κB key factors.
Collapse
Affiliation(s)
- Miguel A Zarate
- Section of Neonatology, Department of Pediatrics, Children's Hospital Colorado, University of Colorado School of Medicine, Aurora, CO, United States
| | - Robyn K De Dios
- Section of Neonatology, Department of Pediatrics, Children's Hospital Colorado, University of Colorado School of Medicine, Aurora, CO, United States
| | - Durganili Balasubramaniyan
- Section of Neonatology, Department of Pediatrics, Children's Hospital Colorado, University of Colorado School of Medicine, Aurora, CO, United States
| | - Lijun Zheng
- Section of Neonatology, Department of Pediatrics, Children's Hospital Colorado, University of Colorado School of Medicine, Aurora, CO, United States
| | - Laura G Sherlock
- Section of Neonatology, Department of Pediatrics, Children's Hospital Colorado, University of Colorado School of Medicine, Aurora, CO, United States
| | - Paul J Rozance
- Section of Neonatology, Department of Pediatrics, Children's Hospital Colorado, University of Colorado School of Medicine, Aurora, CO, United States
| | - Clyde J Wright
- Section of Neonatology, Department of Pediatrics, Children's Hospital Colorado, University of Colorado School of Medicine, Aurora, CO, United States
| |
Collapse
|
6
|
Golden TN, Simmons RA. Immune dysfunction in developmental programming of type 2 diabetes mellitus. Nat Rev Endocrinol 2021; 17:235-245. [PMID: 33526907 PMCID: PMC7969450 DOI: 10.1038/s41574-020-00464-z] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/17/2020] [Indexed: 01/30/2023]
Abstract
Intrauterine growth restriction (IUGR) is a common complication of pregnancy and increases the risk of the offspring developing type 2 diabetes mellitus (T2DM) later in life. Alterations in the immune system are implicated in the pathogenesis of IUGR-induced T2DM. The development of the fetal immune system is a delicate balance as it must remain tolerant of maternal antigens whilst also preparing for the post-birth environment. In addition, the fetal immune system is susceptible to an altered intrauterine milieu caused by maternal and placental inflammatory mediators or secondary to nutrient and oxygen deprivation. Pancreatic-resident macrophages populate the pancreas during fetal development, and their phenotype is dynamic through the neonatal period. Furthermore, macrophages in the islets are instrumental in islet development as they influence β-cell proliferation and islet neogenesis. In addition, cytokines, derived from β-cells and macrophages, are important to islet homeostasis in the fetus and adult and, when perturbed, can cause islet dysfunction. Several activated immune pathways have been identified in the islets of people who experienced IUGR, with alternations in the levels of IL-1β and IL-4 as well as changes in TGFβ signalling. Leptin levels are also altered. Immunomodulation has shown therapeutic benefit in T2DM and might be particularly useful in IUGR-induced T2DM.
Collapse
Affiliation(s)
- Thea N Golden
- Center for Research on Reproduction and Women's Health, Perelman School of Medicine University of Pennsylvania, Philadelphia, PA, USA
- Center of Excellence in Environmental Toxicology, Perelman School of Medicine University of Pennsylvania, Philadelphia, PA, USA
| | - Rebecca A Simmons
- Center for Research on Reproduction and Women's Health, Perelman School of Medicine University of Pennsylvania, Philadelphia, PA, USA.
- Center of Excellence in Environmental Toxicology, Perelman School of Medicine University of Pennsylvania, Philadelphia, PA, USA.
- Division of Neonatology, Children's Hospital of Philadelphia, Philadelphia, PA, USA.
| |
Collapse
|
7
|
Commentary on Zhao et al Manuscript Entitled: "Quantitative Association Between Serum/Dietary Magnesium and Cardiovascular Disease/Coronary Heart Disease Risk: A Dose-Response Meta-analysis of Prospective Cohort Studies": Magnesium and Cardiovascular Disease. J Cardiovasc Pharmacol 2020; 74:508-510. [PMID: 31815865 DOI: 10.1097/fjc.0000000000000768] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
8
|
Karlı P, Özdemir AZ, Ayan D. Maternal Serum and Fetal Cord Blood C-Reactive Protein Levels but not Procalcitonin Levels Are Increased in Idiopathic Intrauterine Growth Restriction. Med Sci Monit 2019; 25:6512-6517. [PMID: 31469126 PMCID: PMC6738000 DOI: 10.12659/msm.917397] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND The aim of the present study was to compare procalcitonin and CRP levels in maternal serum and fetal cord blood samples of patients with idiopathic intrauterine growth restriction (IUGR) vs. a control group of appropriate for gestational age (AGA) infants. MATERIAL AND METHODS The present prospective study included 43 patients: 27 patients with idiopathic IUGR (IUGR group) and 26 AGA infants at similar gestational ages (control group). Maternal serum and fetal cord blood samples were collected from the control group and IUGR group at time of delivery. Procalcitonin and CRP levels were analyzed in maternal blood. Procalcitonin and CRP levels were analyzed in fetal cord blood. RESULTS The median value of CRP levels in maternal blood was 47.5 mg/dl in the IUGR group and 15.255 mg/dl in the AGA group (p=0.001). The mdian CRP level in cord blood was 36.4 mg/dl (range, 17.3-47.2) in the IUGR group and 10.1 mg/dl (range, 4.07-16.5) in the control group, and the difference was statistically significant (p=0.001). The median maternal serum procalcitonin level was 0.05 µg/l in the IUGR group and 0.04 µg/l in the AGA group, and the difference was not statistically significant (p=0.435). The median procalcitonin value in fetal cord blood was 0.06 µg/l in the IUGR group and 0.04 µg/l in the AGA group, and the difference was not statistically significant (p=0.741). CONCLUSIONS Maternal serum and fetal cord CRP levels were higher in the IUGR group; however, there was no difference in procalcitonin, which is another inflammatory indicator, between the groups.
Collapse
Affiliation(s)
- Pervin Karlı
- Department of Obstetrics and Gynecology, Amasya University, Amasya, Turkey
| | - Ayşe Z Özdemir
- Department of Obstetrics and Gynecology, Ondokuz Mayis University IVF Center, Ondokuz Mayis University Hospital, Samsun, Turkey
| | - Durmuş Ayan
- Department of Biochemistry, Amasya Central Public Health Laboratory, Amasya, Turkey
| |
Collapse
|
9
|
Diz-Chaves Y, Toba L, Fandiño J, González-Matías LC, Garcia-Segura LM, Mallo F. The GLP-1 analog, liraglutide prevents the increase of proinflammatory mediators in the hippocampus of male rat pups submitted to maternal perinatal food restriction. J Neuroinflammation 2018; 15:337. [PMID: 30518432 PMCID: PMC6282252 DOI: 10.1186/s12974-018-1370-7] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Accepted: 11/18/2018] [Indexed: 12/17/2022] Open
Abstract
Background Perinatal maternal malnutrition is related to altered growth of tissues and organs. The nervous system development is very sensitive to environmental insults, being the hippocampus a vulnerable structure, in which altered number of neurons and granular cells has been observed. Moreover, glial cells are also affected, and increased expression of proinflammatory mediators has been observed. We studied the effect of Glucagon-like peptide-1 receptor (GLP-1R) agonists, liraglutide, which have very potent metabolic and neuroprotective effects, in order to ameliorate/prevent the glial alterations present in the hippocampus of the pups from mothers with food restriction during pregnancy and lactation (maternal perinatal food restriction—MPFR). Methods Pregnant Sprague-Dawley rats were randomly assigned to 50% food restriction (FR; n = 12) or ad libitum controls (CT, n = 12) groups at day of pregnancy 12 (GD12). From GD14 to parturition, pregnant FR and CT rats were treated with liraglutide (100 μg/kg) or vehicle. At postnatal day 21 and before weaning, 48 males and 45 females (CT and MPFR) were sacrificed. mRNA expression levels of interleukin-1β (IL1β), interleukin-6 (IL-6), nuclear factor-κβ, major histocompatibility complex-II (MHCII), interleukin 10 (IL10), arginase 1 (Arg1), and transforming growth factor (TGFβ) were assessed in the hippocampus by quantitative real-time polymerase chain reaction. Iba1 and GFAP-immunoreactivity were assessed by immunocytochemistry. Results The mRNA expression IL1β, IL6, NF-κB, and MHCII increased in the hippocampus of male but not in female pups from MPFR. In addition, there was an increase in the percentage of GFAP and Iba1-immupositive cells in the dentate gyrus compared to controls, indicating an inflammatory response in the brain. On the other hand, liraglutide treatment prevented the neuroinflammatory process, promoting the production of anti-inflammatory molecules such as IL10, TGFβ, and arginase 1, and decreasing the number and reactivity of microglial cells and astrocytes in the hippocampus of male pups. Conclusion Therefore, the GLP-1 analog, liraglutide, emerges as neuroprotective drug that minimizes the harmful effects of maternal food restriction, decreasing neuroinflammation in the hippocampus in a very early stage.
Collapse
Affiliation(s)
- Y Diz-Chaves
- Laboratory of Endocrinology, Biomedical Research Center (CINBIO), University of Vigo, Campus As Lagoas-Marcosende, E-36310, Vigo (Pontevedra), Spain.
| | - L Toba
- Laboratory of Endocrinology, Biomedical Research Center (CINBIO), University of Vigo, Campus As Lagoas-Marcosende, E-36310, Vigo (Pontevedra), Spain
| | - J Fandiño
- Laboratory of Endocrinology, Biomedical Research Center (CINBIO), University of Vigo, Campus As Lagoas-Marcosende, E-36310, Vigo (Pontevedra), Spain
| | - L C González-Matías
- Laboratory of Endocrinology, Biomedical Research Center (CINBIO), University of Vigo, Campus As Lagoas-Marcosende, E-36310, Vigo (Pontevedra), Spain
| | - L M Garcia-Segura
- Instituto Cajal, Consejo Superior de Investigaciones Científicas (CSIC), Avenida Doctor Arce 37, E-28002, Madrid, Spain.,Centro de Investigación en Red Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, Madrid, Spain
| | - F Mallo
- Laboratory of Endocrinology, Biomedical Research Center (CINBIO), University of Vigo, Campus As Lagoas-Marcosende, E-36310, Vigo (Pontevedra), Spain
| |
Collapse
|
10
|
Kreepala C, Kitporntheranunt M, Sangwipasnapaporn W, Rungsrithananon W, Wattanavaekin K. Assessment of preeclampsia risk by use of serum ionized magnesium-based equation. Ren Fail 2018; 40:99-106. [PMID: 29318926 PMCID: PMC6014514 DOI: 10.1080/0886022x.2017.1422518] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Background: Preeclampsia is a common medical complication in pregnancy. It has been reported to be associated with decreased serum magnesium levels. However, there has not been evidence demonstrating utilization of change in magnesium for prediction of preeclampsia. The purpose of this study was to develop magnesium fraction-based equations which took other significant clinical risk factors into consideration for prediction of preeclampsia. Methods: We collected serum total and ionized magnesium ionized magnesium levels from 84 pregnant women diagnosed with preeclampsia after week 20 of pregnancy. The ionized magnesium fraction was then calculated by the percentage ratio of ionized and total magnesium level. Results: Sixty-four (76.19%) women had normal pregnancy and 20 (23.81%) developed preeclampsia. The ionized magnesium fraction was significantly lower in preeclampsia group (23.95 ± 4.7% vs. 26.28 ± 2.3%, p = .04). Additionally, lower ionized magnesium fraction (24.67%), teenage and elderly primigravida were significantly associated with preeclampsia (OR = 4.41, 95% CI: 1.46–13.40, OR = 5.47, 95% CI: 1.85–35.42 and OR = 11.11, 95% CI: 1.09–113.78, respectively). Consequently, we attempted to develop ionized magnesium fraction-based equations calculate risk scores for preeclampsia. The area of ROC for predictive accuracy of the model was 0.77 (p < .001) and ROC suggested that the score of 0.27 would be a threshold for screening preeclampsia with 70% sensitivity and 81% specificity. Conclusions: Ionized magnesium fraction may have been appropriate for screening of preeclampsia. We suggested blood testing on total and ionized magnesium concentrations as well as calculation of ionized magnesium fraction in addition to routine antenatal care for better screening of the disease.
Collapse
Affiliation(s)
- Chatchai Kreepala
- a Department of Internal Medicine, Faculty of Medicine , Srinakharinwirot University , Ongkharak , Thailand
| | - Maethaphan Kitporntheranunt
- b Department of Obstetrics and Gynecology, Faculty of Medicine , Srinakharinwirot University , Ongkharak , Thailand
| | | | | | | |
Collapse
|
11
|
Wixey JA, Chand KK, Pham L, Colditz PB, Bjorkman ST. Therapeutic potential to reduce brain injury in growth restricted newborns. J Physiol 2018; 596:5675-5686. [PMID: 29700828 DOI: 10.1113/jp275428] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Accepted: 03/28/2018] [Indexed: 12/27/2022] Open
Abstract
Brain injury in intrauterine growth restricted (IUGR) infants is a major contributing factor to morbidity and mortality worldwide. Adverse outcomes range from mild learning difficulties, to attention difficulties, neurobehavioral issues, cerebral palsy, epilepsy, and other cognitive and psychiatric disorders. While the use of medication to ameliorate neurological deficits in IUGR neonates has been identified as warranting urgent research for several years, few trials have been reported. This review summarises clinical trials focusing on brain protection in the IUGR newborn as well as therapeutic interventions trialled in animal models of IUGR. Therapeutically targeting mechanisms of brain injury in the IUGR neonate is fundamental to improving long-term neurodevelopmental outcomes. Inflammation is a key mechanism in neonatal brain injury; and therefore an appealing target. Ibuprofen, an anti-inflammatory drug currently used in the preterm neonate, may be a potential therapeutic candidate to treat brain injury in the IUGR neonate. To better understand the potential of ibuprofen and other therapeutic agents to be neuroprotective in the IUGR neonate, long-term follow-up information of neurodevelopmental outcomes must be studied. Where agents such as ibuprofen are shown to be effective, have a good safety profile and are relatively inexpensive, they can be widely adopted and lead to improved outcomes.
Collapse
Affiliation(s)
- Julie A Wixey
- UQ Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Herston, Queensland, 4029, Australia
| | - Kirat K Chand
- UQ Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Herston, Queensland, 4029, Australia
| | - Lily Pham
- UQ Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Herston, Queensland, 4029, Australia
| | - Paul B Colditz
- UQ Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Herston, Queensland, 4029, Australia
| | - S Tracey Bjorkman
- UQ Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Herston, Queensland, 4029, Australia
| |
Collapse
|
12
|
Antenatal glucocorticoids, magnesium sulfate, and mode of birth in preterm fetal small for gestational age. Am J Obstet Gynecol 2018; 218:S818-S828. [PMID: 29422213 DOI: 10.1016/j.ajog.2017.12.227] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2017] [Revised: 12/09/2017] [Accepted: 12/21/2017] [Indexed: 11/20/2022]
Abstract
A diagnosis of fetal growth restriction and subsequent preterm birth is associated with increased risks of adverse perinatal and neurodevelopmental outcomes and potentially long-lasting effects to adulthood. Most such cases are associated with placental insufficiency and the fetal response to chronic intrauterine hypoxemia and nutrient deprivation leads to substantial physiological and metabolic adaptations. The management of such pregnancies, especially with respect to perinatal interventions and birth mode, remains an unresolved dilemma. The benefits from standard interventions for threatened preterm birth may not be necessarily translated to pregnancies with small-for-gestational-age fetuses. Clinical trials or retrospective studies on outcomes following administration of antenatal glucocorticoids and magnesium sulfate for neuroprotection when preterm birth is imminent either have yielded conflicting results for small-for-gestational-age fetuses, or did not include this subgroup of patients. Experimental models highlight potential harmful effects of administration of antenatal glucocorticoids and magnesium sulfate in the pregnancies with fetal small for gestational age although clinical data do not substantiate these concerns. In addition, heterogeneity in definitions of fetal small for gestational age, variations in the inclusion criteria, and the glucocorticoid regime contribute to inconsistent results. In this review, we discuss the physiologic adaptions of the small-for-gestational-age fetus to its abnormal in utero environment in relation to antenatal glucocorticoids; the impact of antenatal glucocorticoids and intrapartum magnesium sulfate in pregnancies with fetal small for gestational age; the current literature on birth mode for pregnancies with fetal small for gestational age; and the knowledge gaps in the existing literature.
Collapse
|
13
|
Lee DK, Sengupta A, Nevo O. The effect of magnesium sulfate on gene expression in maternal microvascular endothelial cells. Hypertens Pregnancy 2017; 37:30-36. [DOI: 10.1080/10641955.2017.1402924] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Affiliation(s)
- Dennis K. Lee
- Department of Obstetrics and Gynecology, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, Ontario, Canada
| | - Anindita Sengupta
- Department of Obstetrics and Gynecology, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, Ontario, Canada
| | - Ori Nevo
- Department of Obstetrics and Gynecology, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
14
|
Kreepala C, Luangphiphat W, Villarroel A, Kitporntheranunt M, Wattanavaekin K, Piyajarawong T. Effect of Magnesium on Glomerular Filtration Rate and Recovery of Hypertension in Women with Severe Preeclampsia. Nephron Clin Pract 2017; 138:35-41. [PMID: 29176311 DOI: 10.1159/000481463] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Accepted: 09/06/2017] [Indexed: 11/19/2022] Open
Abstract
INTRODUCTION Magnesium sulfate is used for preventing seizures in patients with severe preeclampsia. Previous studies have demonstrated that magnesium plays a significant role in the endothelial function and might have clinically beneficial vasodilating properties. OBJECTIVES This study is aimed at evaluating the effect of magnesium sulfate on the glomerular filtration rate (GFR) during the first 24 h after delivery and during the duration of recovery from hypertension in preeclampsia. METHODS Severe preeclamptic patients who had normal serum creatinine levels (0.4-0.8 mg/dL) were included in the study. Twenty-three women with severe preeclampsia were divided into groups of 9, 8, and 6, and given 1.0, 1.5, and 2.0 g/h of magnesium sulfate, respectively. Magnesium sulfate infusion was used as seizure prophylaxis for 24 h after delivery. The cystatin C-based GFR was monitored for 24 h, and the blood pressure was recorded for 12 weeks postpartum. RESULTS Despite the minimal improvement of GFR 24-h after treatment initiation, survival analysis demonstrated a statistically significant relationship (log rank, p = 0.04) between magnesium dosage and recovery period from hypertension. The group receiving 2.0 g/h of magnesium experienced the shortest recovery period from hypertension (6.5 ± 1.8 days). Meanwhile, the other groups required 66.0 ± 26.9 and 48.3 ± 15.6 days to recover after 1.0 and 1.5 g/h of magnesium infusion, respectively. CONCLUSION Magnesium sulfate has no impact on GFR improvement during the first 24 h after delivery. However, magnesium maintenance infusion at 2.0 g/h is capable of preventing seizure by optimizing the therapeutic magnesium level (4.8-8.4 mg/dL) and shortening the hypertensive episode in preeclampsia.
Collapse
Affiliation(s)
- Chatchai Kreepala
- Department of Internal Medicine, Faculty of Medicine, Srinakharinwirot University, Nakornnayok, Thailand
| | - Wongsakorn Luangphiphat
- Department of Internal Medicine, Faculty of Medicine, Srinakharinwirot University, Nakornnayok, Thailand
| | - Alfredo Villarroel
- Department of Preventive and Social Medicine, Faculty of Medicine, Srinakharinwirot University, Nakornnayok, Thailand
| | - Maethaphan Kitporntheranunt
- Department of Obstetrics and Gynecology, Faculty of Medicine, Srinakharinwirot University, Nakornnayok, Thailand
| | | | | |
Collapse
|
15
|
Wang C, Zhang R, Zhou L, He J, Huang Q, Siyal FA, Zhang L, Zhong X, Wang T. Intrauterine growth retardation promotes fetal intestinal autophagy in rats via the mechanistic target of rapamycin pathway. J Reprod Dev 2017; 63:547-554. [PMID: 28855439 PMCID: PMC5735265 DOI: 10.1262/jrd.2017-050] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Intrauterine growth retardation (IUGR) impairs fetal intestinal development, and is associated with high perinatal morbidity and mortality. However, the mechanism underlying this intestinal injury is largely unknown. We aimed to
investigate this mechanism through analysis of intestinal autophagy and related signaling pathways in a rat model of IUGR. Normal weight (NW) and IUGR fetuses were obtained from primiparous rats via ad libitum
food intake and 50% food restriction, respectively. Maternal serum parameters, fetal body weight, organ weights, and fetal blood glucose were determined. Intestinal apoptosis, autophagy, and the mechanistic target of rapamycin
(mTOR) signaling pathway were analyzed. The results indicated that maternal 50% food restriction reduced maternal serum glucose, bilirubin, and total cholesterol and produced IUGR fetuses, which had decreased body weight; blood
glucose; and weights of the small intestine, stomach, spleen, pancreas, and kidney. Decreased Bcl-2 and increased Casp9 mRNA expression was observed in IUGR fetal intestines. Analysis of
intestinal autophagy showed that the mRNA expression of WIPI1, MAP1LC3B, Atg5, and Atg14 was also increased, while the protein levels of p62 were decreased in
IUGR fetuses. Compared to NW fetuses, IUGR fetuses showed decreased mTOR protein levels and enhanced mRNA expression of ULK1 and Beclin1 in the small intestine. In summary, the results indicated
that maternal 50% food restriction on gestational days 10–21 reduced maternal serum glucose, bilirubin, and total cholesterol contents, and produced IUGR fetuses that had low blood glucose and reduced small intestine weight.
Intestinal injury of IUGR fetuses caused by maternal food restriction might be due to enhanced apoptosis and autophagy via the mTOR signaling pathway.
Collapse
Affiliation(s)
- Chao Wang
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, People's Republic of China
| | - Ruiming Zhang
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, People's Republic of China
| | - Le Zhou
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, People's Republic of China
| | - Jintian He
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, People's Republic of China
| | - Qiang Huang
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, People's Republic of China
| | - Farman A Siyal
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, People's Republic of China
| | - Lili Zhang
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, People's Republic of China
| | - Xiang Zhong
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, People's Republic of China
| | - Tian Wang
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, People's Republic of China
| |
Collapse
|
16
|
Abstract
BACKGROUND Magnesium (Mg) is an essential mineral required to regulate body temperature, nucleic acid, and protein synthesis with an important role in maintaining nerve and muscle cell electrical potentials. It may reduce fetal growth restriction and preeclampsia as well as increase birth weight. This study aimed to assess the effects of consuming Mg supplementation during pregnancy on pregnancy outcomes. MATERIALS AND METHODS This is a randomized controlled trial with three sixty populated groups of pregnant women. Participants were randomized to treatment or control groups through random table numbers. Participants with Mg serum levels more than 1.9 mg/dl considered as control group A randomly. They just received one multimineral tablet once a day until the end of pregnancy participants with hypomagnesemia consider as Group B and C. Participants in Group B received one multimineral tablet daily until the end of pregnancy. Participants in Group C received 200 mg effervescent Mg tablet from Vitafit Company once daily for 1 month, and also they consumed one multimineral tablet from Alhavi Company, which contains 100 mg Mg, once a day until the end of pregnancy. Intrauterine growth retardation, preterm labor, maternal body mass index, neonatal weight, pregnancy-induced hypertension, preeclampsia, gestational diabetes mellitus, cramps of the leg Apgar score were compared between three groups. RESULTS In all pregnancy outcomes, Group C that received effervescent Mg tablet plus multimineral showed a better result than other groups, and frequency of complications of pregnancy was fewer than the other two groups and showed a significant difference. CONCLUSION Mg supplement during pregnancy likely decrease probability occurrence of many complications of pregnancy.
Collapse
Affiliation(s)
- Elaheh Zarean
- Department of Obstetrics and Gynecology, Alzahra Hospital, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Amal Tarjan
- Department of Obstetrics and Gynecology, Alzahra Hospital, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
17
|
Wixey JA, Chand KK, Colditz PB, Bjorkman ST. Review: Neuroinflammation in intrauterine growth restriction. Placenta 2016; 54:117-124. [PMID: 27916232 DOI: 10.1016/j.placenta.2016.11.012] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2016] [Revised: 11/21/2016] [Accepted: 11/22/2016] [Indexed: 12/21/2022]
Abstract
Disruption to the maternal environment during pregnancy from events such as hypoxia, stress, toxins, inflammation, and reduced placental blood flow can affect fetal development. Intrauterine growth restriction (IUGR) is commonly caused by chronic placental insufficiency, interrupting supply of oxygen and nutrients to the fetus resulting in abnormal fetal growth. IUGR is a major cause of perinatal morbidity and mortality, occurring in approximately 5-10% of pregnancies. The fetal brain is particularly vulnerable in IUGR and there is an increased risk of long-term neurological disorders including cerebral palsy, epilepsy, learning difficulties, behavioural difficulties and psychiatric diagnoses. Few studies have focused on how growth restriction interferes with normal brain development in the IUGR neonate but recent studies in growth restricted animal models demonstrate increased neuroinflammation. This review describes the role of neuroinflammation in the progression of brain injury in growth restricted neonates. Identifying the mediators responsible for alterations in brain development in the IUGR infant is key to prevention and treatment of brain injury in these infants.
Collapse
Affiliation(s)
- Julie A Wixey
- The University of Queensland, Perinatal Research Centre, UQ Centre for Clinical Research, Herston, Queensland 4029, Australia.
| | - Kirat K Chand
- The University of Queensland, Perinatal Research Centre, UQ Centre for Clinical Research, Herston, Queensland 4029, Australia
| | - Paul B Colditz
- The University of Queensland, Perinatal Research Centre, UQ Centre for Clinical Research, Herston, Queensland 4029, Australia
| | - S Tracey Bjorkman
- The University of Queensland, Perinatal Research Centre, UQ Centre for Clinical Research, Herston, Queensland 4029, Australia
| |
Collapse
|
18
|
The effects of prenatal metformin on obesogenic diet-induced alterations in maternal and fetal fatty acid metabolism. Nutr Metab (Lond) 2016; 13:55. [PMID: 27555877 PMCID: PMC4994240 DOI: 10.1186/s12986-016-0115-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Accepted: 08/13/2016] [Indexed: 12/21/2022] Open
Abstract
Background Maternal obesity may program the fetus and increase the susceptibility of the offspring to adult diseases. Metformin crosses the placenta and has been associated with decreased inflammation and reversal of fatty liver in obese leptin-deficient mice. We investigated the effects of metformin on maternal and fetal lipid metabolism and hepatic inflammation using a rat model of diet-induced obesity during pregnancy. Methods Female Wistar rats (6–7 weeks old) were fed normal or high calorie diets for 5 weeks. After mating with normal-diet fed males, half of the high calorie-fed dams received metformin (300 mg/kg, daily); dams (8 per group) continued diets through gestational day 19. Maternal and fetal livers and fetal brains were analyzed for fatty acids and for fatty acid metabolism-related gene expression. Data were analyzed by ANOVA followed by Dunnett’s post hoc testing. Results When compared to control-lean maternal livers, obesogenic-diet-exposed maternal livers showed significantly higher saturated fatty acids (14:0 and 16:0) and monounsaturated fatty acids (16:1n7 and 18:1n9) and lower polyunsaturated (18:2n6 and 20:4n6 [arachidonic acid]) and anti-inflammatory n3 polyunsaturated fatty acids (18:3n3 and 22:6n3 [docosahexaenoic acid]) (p < 0.05). Metformin did not affect diet-induced changes in maternal livers. Fetal livers exposed to the high calorie diet showed significantly increased saturated fatty acids (18:0) and monounsaturated fatty acids (18:1n9 and 18:1n7) and decreased polyunsaturated fatty acids (18:2n6, 20:4n6 and 22:6n3) and anti-inflammatory n3 polyunsaturated fatty acids, along with increased gene expression of fatty acid metabolism markers (Fasn, D5d, D6d, Scd1, Lxrα). Metformin significantly attenuated diet-induced inflammation and 18:1n9 and 22:6n3 in fetal livers, as well as n3 fatty acids (p < 0.05). Prenatal obesogenic diet exposure significantly increased fetal liver IFNγ levels (p < 0.05), which was reversed by maternal metformin treatment (p < 0.05). Conclusions Consumption of a high calorie diet significantly affected maternal and fetal fatty acid metabolism. It reduced anti-inflammatory polyunsaturated fatty acids in maternal and fetal livers, altered gene expression of fatty acid metabolism markers, and induced inflammation in the fetal livers. Prenatal metformin attenuated some diet-induced fatty acid changes and inflammation in the fetal livers without affecting maternal livers, suggesting that maternal metformin may impact fetal/neonatal fatty acid/lipid metabolism. Electronic supplementary material The online version of this article (doi:10.1186/s12986-016-0115-9) contains supplementary material, which is available to authorized users.
Collapse
|
19
|
Das UN. Beneficial actions of magnesium in metabolic syndrome: Why and how? Nutrition 2016; 32:1308-10. [PMID: 27236634 DOI: 10.1016/j.nut.2016.03.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Revised: 03/20/2016] [Accepted: 03/23/2016] [Indexed: 10/22/2022]
Affiliation(s)
- Undurti N Das
- UND Life Sciences, Federal Way, Washington, USA; BioScience Research Centre GVP College of Engineering Campus and Department of Medicine, GVP Hospital Visakhapatnam, India
| |
Collapse
|
20
|
El-Tanbouly DM, Abdelsalam RM, Attia AS, Abdel-Aziz MT. Pretreatment with magnesium ameliorates lipopolysaccharide-induced liver injury in mice. Pharmacol Rep 2015; 67:914-20. [PMID: 26398385 DOI: 10.1016/j.pharep.2015.02.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2014] [Revised: 02/11/2015] [Accepted: 02/12/2015] [Indexed: 12/18/2022]
Abstract
BACKGROUND Lipopolysaccharide (LPS), a component of the outer membrane of Gram-negative bacteria, is involved in the pathogenesis of sepsis. LPS administration induces systemic inflammation that mimics many of the initial clinical features of sepsis and has deleterious effects on several organs including the liver and eventually leading to septic shock and death. The present study aimed to investigate the protective effect of magnesium (Mg), a well known cofactor in many enzymatic reactions and a critical component of the antioxidant system, on hepatic damage associated with LPS-induced endotoxima in mice. METHODS Mg (20 and 40mg/kg, po) was administered for 7 consecutive days. Systemic inflammation was induced 1h after the last dose of Mg by a single dose of LPS (2mg/kg, ip) and 3h thereafter plasma was separated, animals were sacrificed and their livers were isolated. RESULTS LPS-treated mice suffered from hepatic dysfunction revealed by histological observation, elevation in plasma transaminases activities, C-reactive protein content and caspase-3, a critical marker of apoptosis. Liver inflammation was evident by elevation in liver cytokines contents (TNF-α and IL-10) and MPO activity. Additionally, oxidative stress was manifested by increased liver lipoperoxidation, glutathione depletion, elevated total nitrate/nitrite (NOx) content and glutathione peroxidase (GPx) activity. Pretreatment with Mg largely mitigated these alternations. CONCLUSION Pretreatment with Mg protects the liver from the acute injury which occurs shortly after septicemia.
Collapse
Affiliation(s)
- Dalia M El-Tanbouly
- Pharmacology & Toxicology Department, Faculty of Pharmacy, Cairo University, Cairo, Egypt.
| | - Rania M Abdelsalam
- Pharmacology & Toxicology Department, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Amina S Attia
- Pharmacology & Toxicology Department, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Mohamed T Abdel-Aziz
- Pharmacology & Toxicology Department, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| |
Collapse
|
21
|
Acar N, Soylu H, Edizer I, Ozbey O, Er H, Akkoyunlu G, Gemici B, Ustunel I. Expression of nuclear factor erythroid 2-related factor 2 (Nrf2) and peroxiredoxin 6 (Prdx6) proteins in healthy and pathologic placentas of human and rat. Acta Histochem 2014; 116:1289-300. [PMID: 25171874 DOI: 10.1016/j.acthis.2014.07.012] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2014] [Revised: 07/22/2014] [Accepted: 07/23/2014] [Indexed: 12/31/2022]
Abstract
A relationship has been shown between preeclampsia (PE) and intrauterine growth restriction (IUGR) and oxidative stress (OS). Since such pregnancies experience OS, we aimed to detect the distribution pattern and expression levels of a transcription factor, Nuclear factor erythroid 2-related factor-2 (Nrf2) that has a role in the regulation of antioxidant enzymes, and peroxiredoxin 6 (Prdx6) an antioxidant enzyme, in human healthy, IUGR, PE and in groups of rat healthy and IUGR placentas using immunohistochemistry and Western blotting. Both Nrf2 and Prdx6 immunoreactivities were weaker in human and rat IUGR group placentas compared to human and rat control group placentas, respectively. Nrf2 and Prdx6 were immunostained in labyrinth trophoblasts, decidua, giant, glycogen and fetal vessel endothelial cells in rat control and IUGR group placentas. Nrf2 and Prdx6 immunoreactivities were seen in the decidua, syncytiotrophoblasts, villous stromal cells, and vascular endothelium in human control, IUGR and PE group placentas. Results of Nrf2 and Prdx6 Western blotting applied for rat and human placentas were compatible with the results of Nrf2 and Prdx6 immunohistochemical observations with regard to rat and human placentas. Down-regulation of Nrf2 and Prdx6 proteins in human and rat IUGR group placentas may have led to the formation of OS which may have impaired proliferation and invasion of cytotrophoblasts.
Collapse
Affiliation(s)
- Nuray Acar
- Department of Histology and Embryology, Faculty of Medicine, Akdeniz University, Antalya, Turkey
| | - Hakan Soylu
- Department of Histology and Embryology, Faculty of Medicine, Akdeniz University, Antalya, Turkey
| | - Imren Edizer
- Department of Histology and Embryology, Faculty of Medicine, Akdeniz University, Antalya, Turkey
| | - Ozlem Ozbey
- Department of Histology and Embryology, Faculty of Medicine, Akdeniz University, Antalya, Turkey
| | - Hakan Er
- Department of Biophysics, Faculty of Medicine, Akdeniz University, Antalya, Turkey
| | - Gokhan Akkoyunlu
- Department of Histology and Embryology, Faculty of Medicine, Akdeniz University, Antalya, Turkey
| | - Burcu Gemici
- Department of Physiology, Faculty of Medicine, Near East University, Nicosia, Mersin 10, Turkey
| | - Ismail Ustunel
- Department of Histology and Embryology, Faculty of Medicine, Akdeniz University, Antalya, Turkey.
| |
Collapse
|
22
|
Das UN. Magnesium Supplementation Reduces Metabolic Syndrome—How and Why? Arch Med Res 2014; 45:604-6. [DOI: 10.1016/j.arcmed.2014.10.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2014] [Accepted: 10/01/2014] [Indexed: 12/15/2022]
|
23
|
Gupta M, Solanki MH, Chatterjee PK, Xue X, Roman A, Desai N, Rochelson B, Metz CN. Maternal magnesium deficiency in mice leads to maternal metabolic dysfunction and altered lipid metabolism with fetal growth restriction. Mol Med 2014; 20:332-40. [PMID: 25025397 DOI: 10.2119/molmed.2014.00137] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2014] [Accepted: 07/11/2014] [Indexed: 01/10/2023] Open
Abstract
Inadequate magnesium (Mg) intake is a widespread problem, with over 50% of women of reproductive age consuming less than the Recommended Dietary Allowance (RDA). Because pregnancy increases the requirement for Mg and the beneficial effects of magnesium sulfate for preeclampsia/eclampsia and fetal neuroprotection are well described, we examined the outcomes of Mg deficiency during pregnancy. Briefly, pregnant Swiss Webster mice were fed either control or Mg-deficient diets starting on gestational day (GD) 6 through euthanasia on GD17. Mg-deficient dams had significantly reduced weight gain and higher plasma adipokines, in the absence of inflammation. Livers of Mg-deficient dams had significantly higher saturated fatty acids (SFAs) and monounsaturated fatty acids (MUFAs) and lower polyunsaturated fatty acids (PUFAs), including docosahexaenoic acid (DHA) (P < 0.0001) and arachidonic acid (AA) (P < 0.0001). Mechanistically, Mg deficiency was accompanied by enhanced desaturase and elongase mRNA expression in maternal livers along with higher circulating insulin and glucose concentrations (P < 0.05) and increased mRNA expression of Srebf1 and Chrebp, regulators of fatty acid synthesis (P < 0.05). Fetal pups exposed to Mg deficiency were growth-restricted and exhibited reduced survival. Mg-deficient fetal livers showed lower MUFAs and higher PUFAs, with lower desaturase and elongase mRNA expression than controls. In addition, DHA concentrations were lower in Mg-deficient fetal brains (P < 0.05). These results indicate that Mg deficiency during pregnancy influences both maternal and fetal fatty acid metabolism, fetal growth and fetal survival, and support better understanding maternal Mg status before and during pregnancy.
Collapse
Affiliation(s)
- Madhu Gupta
- Elmezzi Graduate School of Molecular Medicine, Manhasset, New York, United States of America The Feinstein Institute for Medical Research, The Center for Immunology and Inflammation, Manhasset, New York, United States of America
| | - Malvika H Solanki
- Elmezzi Graduate School of Molecular Medicine, Manhasset, New York, United States of America The Feinstein Institute for Medical Research, The Center for Immunology and Inflammation, Manhasset, New York, United States of America
| | - Prodyot K Chatterjee
- The Feinstein Institute for Medical Research, The Center for Immunology and Inflammation, Manhasset, New York, United States of America
| | - Xiangying Xue
- The Feinstein Institute for Medical Research, The Center for Immunology and Inflammation, Manhasset, New York, United States of America
| | - Amanda Roman
- Hofstra North Shore-LIJ School of Medicine, Division of Maternal-Fetal Medicine, Manhasset, New York, United States of America
| | - Neeraj Desai
- Hofstra North Shore-LIJ School of Medicine, Division of Maternal-Fetal Medicine, Manhasset, New York, United States of America
| | - Burton Rochelson
- Hofstra North Shore-LIJ School of Medicine, Division of Maternal-Fetal Medicine, Manhasset, New York, United States of America
| | - Christine N Metz
- Elmezzi Graduate School of Molecular Medicine, Manhasset, New York, United States of America The Feinstein Institute for Medical Research, The Center for Immunology and Inflammation, Manhasset, New York, United States of America Hofstra North Shore-LIJ School of Medicine, Division of Maternal-Fetal Medicine, Manhasset, New York, United States of America
| |
Collapse
|
24
|
Islet inflammation, hemosiderosis, and fibrosis in intrauterine growth-restricted and high fat-fed Sprague-Dawley rats. THE AMERICAN JOURNAL OF PATHOLOGY 2014; 184:1446-57. [PMID: 24631026 DOI: 10.1016/j.ajpath.2014.01.024] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/19/2013] [Revised: 01/03/2014] [Accepted: 01/28/2014] [Indexed: 12/27/2022]
Abstract
Prenatal and postnatal factors such as intrauterine growth restriction (IUGR) and high-fat (HF) diet contribute to type 2 diabetes. Our aim was to determine whether IUGR and HF diets interact in type 2 diabetes pathogenesis, with particular attention focused on pancreatic islet morphology including assessment for inflammation. A surgical model of IUGR (bilateral uterine artery ligation) in Sprague-Dawley rats with sham controls was used. Pups were fed either HF or chow diets after weaning. Serial measures of body weight and glucose tolerance were performed. At 25 weeks of age, rat pancreases were harvested for histologic assessment. The birth weight of IUGR pups was 13% lower than that of sham pups. HF diet caused excess weight gain, dyslipidemia, hyperinsulinemia, and mild glucose intolerance, however, this was not aggravated further by IUGR. Markedly abnormal islet morphology was evident in 0 of 6 sham-chow, 5 of 8 sham-HF, 4 of 8 IUGR-chow, and 8 of 9 IUGR-HF rats (chi-square, P = 0.007). Abnormal islets were characterized by larger size, irregular shape, inflammation with CD68-positive cells, marked fibrosis, and hemosiderosis. β-Cell mass was not altered by IUGR. In conclusion, HF and IUGR independently contribute to islet injury characterized by inflammation, hemosiderosis, and fibrosis. This suggests that both HF and IUGR can induce islet injury via converging pathways. The potential pathogenic or permissive role of iron in this process of islet inflammation warrants further investigation.
Collapse
|
25
|
Doll E, Wilkes J, Cook LJ, Korgenski EK, Faix RG, Yoder BA, Srivastava R, Sherwin CMT, Spigarelli MG, Clark EAS, Bonkowsky JL. Neonatal magnesium levels correlate with motor outcomes in premature infants: a long-term retrospective cohort study. Front Pediatr 2014; 2:120. [PMID: 25414842 PMCID: PMC4220726 DOI: 10.3389/fped.2014.00120] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2014] [Accepted: 10/22/2014] [Indexed: 02/03/2023] Open
Abstract
OBJECTIVE Chronic neurological deficits are a significant complication of preterm birth. Magnesium supplementation has been suggested to have neuroprotective function in the developing brain. Our objective was to determine whether higher neonatal serum magnesium levels were associated with better long-term neurodevelopmental outcomes in very-low birth weight infants. STUDY DESIGN A retrospective cohort of 75 preterm infants (<1500 g, gestational age <27 weeks) had follow-up for the outcomes of abnormal motor exam and for epilepsy. Average total serum magnesium level in the neonate during the period of prematurity was the main independent variable assessed, tested using a Wilcoxon rank-sum test. RESULTS Higher average serum magnesium level was associated with a statistically significant decreased risk for abnormal motor exam (p = 0.037). A lower risk for epilepsy in the group with higher magnesium level did not reach statistical significance (p = 0.06). CONCLUSION This study demonstrates a correlation between higher neonatal magnesium levels and decreased risk for long-term abnormal motor exam. Larger studies are needed to evaluate the hypothesis that higher neonatal magnesium levels can improve long-term neurodevelopmental outcomes.
Collapse
Affiliation(s)
- Elizabeth Doll
- Division of Pediatric Neurology, School of Medicine, University of Utah , Salt Lake City, UT , USA
| | - Jacob Wilkes
- Intermountain Healthcare , Salt Lake City, UT , USA
| | - Lawrence J Cook
- Division of Critical Care Medicine, School of Medicine, University of Utah , Salt Lake City, UT , USA
| | | | - Roger G Faix
- Division of Neonatology, School of Medicine, University of Utah , Salt Lake City, UT , USA
| | - Bradley A Yoder
- Division of Neonatology, School of Medicine, University of Utah , Salt Lake City, UT , USA
| | - Rajendu Srivastava
- Intermountain Healthcare , Salt Lake City, UT , USA ; Division of Inpatient Medicine, School of Medicine, University of Utah , Salt Lake City, UT , USA
| | - Catherine M T Sherwin
- Division of Clinical Pharmacology, Department of Pediatrics, School of Medicine, University of Utah , Salt Lake City, UT , USA
| | - Michael G Spigarelli
- Division of Clinical Pharmacology, Department of Pediatrics, School of Medicine, University of Utah , Salt Lake City, UT , USA
| | - Erin A S Clark
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, School of Medicine, University of Utah , Salt Lake City, UT , USA
| | - Joshua L Bonkowsky
- Division of Pediatric Neurology, School of Medicine, University of Utah , Salt Lake City, UT , USA
| |
Collapse
|
26
|
Das UN. Magnesium supplementation reduces intrauterine growth restriction and inflammation: how and why? Am J Obstet Gynecol 2013; 209:497-8. [PMID: 23770476 DOI: 10.1016/j.ajog.2013.06.014] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2013] [Accepted: 06/10/2013] [Indexed: 10/26/2022]
|
27
|
Metz C, Roman A, Gupta M, Chatterjee PK. Reply: To PMID 23474429. Am J Obstet Gynecol 2013; 209:498-9. [PMID: 23770469 DOI: 10.1016/j.ajog.2013.06.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2013] [Accepted: 06/10/2013] [Indexed: 11/15/2022]
Affiliation(s)
- Christine Metz
- Center for Immunology and Inflammation, Feinstein Institute for Medical Research, 350 Community Dr., Manhasset, NY 11030.
| | | | | | | |
Collapse
|