Maternal pomegranate juice intake and brain structure and function in infants with intrauterine growth restriction: A randomized controlled pilot study

Lillian G Matthews, Christopher D Smyser, Sara Cherkerzian, Dimitrios Alexopoulos, Jeanette Kenley, Methodius G Tuuli, D Michael Nelson, Terrie E Inder, Lillian G Matthews, Christopher D Smyser, Sara Cherkerzian, Dimitrios Alexopoulos, Jeanette Kenley, Methodius G Tuuli, D Michael Nelson, Terrie E Inder

Abstract

Polyphenol-rich pomegranate juice has been shown to have benefit as a neuroprotectant in animal models of neonatal hypoxic-ischemia. No published studies have investigated maternal polyphenol administration as a potential neuroprotectant in at-risk newborns, such as those with intrauterine growth restriction (IUGR). This was a randomized, placebo-controlled, double-blind pilot study to investigate the impact of maternal pomegranate juice intake in pregnancies with IUGR, on newborn brain structure and function at term-equivalent age (TEA). Mothers with IUGR at 24-34 weeks' gestation were recruited from Barnes-Jewish Hospital obstetrical clinic. Consented mothers were randomized to treatment (8 oz. pomegranate juice) or placebo (8 oz. polyphenol-free juice) and continued to take juice daily from enrollment until delivery (mean 20.1 and 27.1 days, respectively). Infants underwent brain MRI at TEA (36-41 weeks' gestation). Brain measures were compared between groups including: brain injury score, brain metrics, brain volumes, diffusion tensor imaging and resting state functional connectivity. Statistical analyses were undertaken as modified intention-to-treat (including randomized participants who received their allocated intervention and whose infants received brain MRI) and per-protocol (including participants who strictly adhered to the protocol, based on metabolite status). Seventy-seven mothers were randomized to treatment (n = 40) or placebo (n = 37). Of these, 28 and 27 infants, respectively, underwent term-equivalent MRI. There were no group differences in brain injury, metrics or volumes. However, treatment subjects displayed reduced diffusivity within the anterior and posterior limbs of the internal capsule compared with placebo. Resting state functional connectivity demonstrated increased correlation and covariance within several networks in treatment subjects, with alterations most apparent in the visual network in per-protocol analyses. Direct effects on health were not found. In conclusion, maternal pomegranate juice intake in pregnancies with known IUGR was associated with altered white matter organization and functional connectivity in the infant brain, suggesting differences in brain structure and function following in utero pomegranate juice exposure, warranting continued investigation. Clinical trial registration. NCT00788866, registered November 11, 2008, initial participant enrollment August 21, 2012.

Conflict of interest statement

The authors have declared that no competing interests exist.

Figures

Fig 1. Participant flowchart.
Fig 1. Participant flowchart.
Fig 2. Relationships between maternal pomegranate juice…
Fig 2. Relationships between maternal pomegranate juice intake and infant DTI measures.
(Top) Treatment vs. placebo (modified intention-to-treat analysis). (Bottom) Metabolite-positive treatment vs. metabolite-negative placebo (per-protocol analysis). The point estimates represent the difference between groups in white matter DTI measures based on analyses run using generalized linear models (GLM) adjusted for postmenstrual age at scan. The error bars represent the 95% confidence intervals. ALIC–anterior limb of internal capsule; CC–corpus callosum; CNBD–cingulum bundle; CSOV–centrum semiovale; FA–fractional anisotropy; Frontal–frontal lobe; OR–optic radiation; MD–mean diffusivity; PLIC–posterior limb of internal capsule; RD–radial diffusivity. Left/Right indicate measures for bilateral white matter tracts.
Fig 3. Relationships between maternal pomegranate juice…
Fig 3. Relationships between maternal pomegranate juice intake and infant fcMRI measures (correlations).
Group mean Fisher’s z-transformed correlation coefficient matrices are shown representing all ROI pairs. The block structure along the diagonal seen in both groups corresponds to resting state networks. Warm hues within diagonal blocks reflect positively correlated ROIs within RSN, while cool hues reflect negatively correlated ROIs within RSNs. (Top, A-C) Treatment vs. placebo (modified intention-to-treat analysis). (A) Infants in treatment (pomegranate juice) group and (B) infants in placebo group at term-equivalent. (C) Shows the difference between groups (treatment minus placebo). (Bottom, D-F) Metabolite-positive treatment vs. metabolite-negative placebo (per-protocol analysis). (D) Infants in metabolite-positive treatment group and (E) infants in metabolite-negative placebo group at term-equivalent. (F) Shows the difference between groups (metabolite-positive treatment minus metabolite-negative placebo). Note metabolite-positive treatment > metabolite-negative placebo correlation in subcortical and visual network. CER—cerebellar; CO—cingulo-opercular; DAN–dorsal attention network; DMN–default mode network; FP–frontal parietal network; LAN–language area network; Metab–metabolite; Neg–negative; POM–pomegranate; Pos–positive; SAL–salience network; SMN–sensorimotor network; SUB–subcortical grey matter; VAN–ventral attention network; VIS–visual network.
Fig 4. Relationships between maternal pomegranate juice…
Fig 4. Relationships between maternal pomegranate juice intake and infant fcMRI measures (covariance).
Group mean covariance matrices are shown representing all ROI pairs. The block structure along the diagonal seen in both groups corresponds to resting state networks. Warm hues within diagonal blocks reflect positively correlated ROIs within RSN, while cool hues reflect negatively correlated ROIs within RSNs. (Top, A-C) Treatment vs. placebo (modified intention-to-treat analysis). (A) Infants in treatment (pomegranate juice) group and (B) infants in placebo group at term-equivalent. (C) Shows the difference between groups (treatment minus placebo). Note treatment > placebo covariance within subcortical and visual networks. (Bottom, D-F) Metabolite-positive treatment vs. metabolite-negative placebo (per-protocol analysis). (D) Infants in metabolite-positive treatment group and (E) infants in metabolite-negative placebo group at term-equivalent. (F) Shows the difference between groups (metabolite-positive treatment minus metabolite-negative placebo). Note metabolite-positive treatment > metabolite-negative placebo covariance within subcortical and visual networks. CER—cerebellar; CO—cingulo-opercular; DAN–dorsal attention network; DMN–default mode network; FP–frontal parietal network; LAN–language area network; Metab–metabolite; Neg–negative; POM–pomegranate; Pos–positive; SAL–salience network; SMN–sensorimotor network; SUB–subcortical grey matter; VAN–ventral attention network; VIS–visual network.

References

    1. Volpe JJ. Chapter 19. Hypoxic-Ischemic Injury in the Term Infant: Pathophysiology. Volpe's Neurology of the Newborn, 6th Edition. 5th ed ed Philadelphia: Elsevier; 2018. p. 423.
    1. Kurinczuk JJ, White-Koning M, Badawi N. Epidemiology of neonatal encephalopathy and hypoxic-ischaemic encephalopathy. Early human development. 2010;86(6):329–38. 10.1016/j.earlhumdev.2010.05.010
    1. Lawn JE, Cousens S, Zupan J. 4 million neonatal deaths: when? Where? Why? Lancet (London, England). 2005;365(9462):891–900.
    1. Groenendaal F, Casaer A, Dijkman KP, Gavilanes AWD, de Haan TR, ter Horst HJ, et al. Introduction of Hypothermia for Neonates with Perinatal Asphyxia in the Netherlands and Flanders. Neonatology. 2013;104(1):15–21. 10.1159/000348823
    1. Shah PS, Beyene J, To T, Ohlsson A, Perlman M. Postasphyxial hypoxic-ischemic encephalopathy in neonates: outcome prediction rule within 4 hours of birth. Archives of pediatrics & adolescent medicine. 2006;160(7):729–36.
    1. Tekgul H, Gauvreau K, Soul J, Murphy L, Robertson R, Stewart J, et al. The current etiologic profile and neurodevelopmental outcome of seizures in term newborn infants. Pediatrics. 2006;117(4):1270–80. 10.1542/peds.2005-1178
    1. Kumar A, Ramakrishna SV, Basu S, Rao GR. Oxidative stress in perinatal asphyxia. Pediatric neurology. 2008;38(3):181–5. 10.1016/j.pediatrneurol.2007.10.008
    1. Zhao M, Zhu P, Fujino M, Zhuang J, Guo H, Sheikh I, et al. Oxidative Stress in Hypoxic-Ischemic Encephalopathy: Molecular Mechanisms and Therapeutic Strategies. International Journal of Molecular Sciences. 2016;17(12):2078.
    1. Ferriero DM. The Vulnerable Newborn Brain: Imaging Patterns of Acquired Perinatal Injury. Neonatology. 2016;109(4):345–51. 10.1159/000444896
    1. Inder TE, Volpe JJ. Mechanisms of perinatal brain injury. Seminars in neonatology: SN. 2000;5(1):3–16. 10.1053/siny.1999.0112
    1. McLean C, Ferriero D. Mechanisms of hypoxic-ischemic injury in the term infant. Seminars in perinatology. 2004;28(6):425–32.
    1. Mokni M, Hamlaoui S, Karkouch I, Amri M, Marzouki L, Limam F, et al. Resveratrol Provides Cardioprotection after Ischemia/reperfusion Injury via Modulation of Antioxidant Enzyme Activities. Iranian Journal of Pharmaceutical Research: IJPR. 2013;12(4):867–75.
    1. Youdim KA, Shukitt-Hale B, Joseph JA. Flavonoids and the brain: interactions at the blood-brain barrier and their physiological effects on the central nervous system. Free radical biology & medicine. 2004;37(11):1683–93.
    1. Aquilano K, Baldelli S, Rotilio G, Ciriolo MR. Role of nitric oxide synthases in Parkinson's disease: a review on the antioxidant and anti-inflammatory activity of polyphenols. Neurochemical research. 2008;33(12):2416–26. 10.1007/s11064-008-9697-6
    1. Bastianetto S, Krantic S, Quirion R. Polyphenols as potential inhibitors of amyloid aggregation and toxicity: possible significance to Alzheimer's disease. Mini reviews in medicinal chemistry. 2008;8(5):429–35.
    1. Esmaillzadeh A, Tahbaz F, Gaieni I, Alavi-Majd H, Azadbakht L. Cholesterol-lowering effect of concentrated pomegranate juice consumption in type II diabetic patients with hyperlipidemia. International journal for vitamin and nutrition research Internationale Zeitschrift fur Vitamin- und Ernahrungsforschung Journal international de vitaminologie et de nutrition. 2006;76(3):147–51. 10.1024/0300-9831.76.3.147
    1. Hong MY, Seeram NP, Heber D. Pomegranate polyphenols down-regulate expression of androgen-synthesizing genes in human prostate cancer cells overexpressing the androgen receptor. The Journal of nutritional biochemistry. 2008;19(12):848–55. 10.1016/j.jnutbio.2007.11.006
    1. Shema-Didi L, Sela S, Ore L, Shapiro G, Geron R, Moshe G, et al. One year of pomegranate juice intake decreases oxidative stress, inflammation, and incidence of infections in hemodialysis patients: A randomized placebo-controlled trial. Free Radical Biology and Medicine. 2012;53(2):297–304. 10.1016/j.freeradbiomed.2012.05.013
    1. Heber D, Seeram NP, Wyatt H, Henning SM, Zhang Y, Ogden LG, et al. Safety and antioxidant activity of a pomegranate ellagitannin-enriched polyphenol dietary supplement in overweight individuals with increased waist size. Journal of agricultural and food chemistry. 2007;55(24):10050–4. 10.1021/jf071689v
    1. Seeram NP, Aviram M, Zhang Y, Henning SM, Feng L, Dreher M, et al. Comparison of antioxidant potency of commonly consumed polyphenol-rich beverages in the United States. Journal of agricultural and food chemistry. 2008;56(4):1415–22. 10.1021/jf073035s
    1. Loren DJ, Seeram NP, Schulman RN, Holtzman DM. Maternal dietary supplementation with pomegranate juice is neuroprotective in an animal model of neonatal hypoxic-ischemic brain injury. Pediatric research. 2005;57(6):858–64. 10.1203/01.PDR.0000157722.07810.15
    1. West T, Atzeva M, Holtzman DM. Pomegranate polyphenols and resveratrol protect the neonatal brain against hypoxic-ischemic injury. Developmental neuroscience. 2007;29(4–5):363–72. 10.1159/000105477
    1. Chen B, Longtine MS, Nelson DM. Punicalagin, a polyphenol in pomegranate juice, downregulates p53 and attenuates hypoxia-induced apoptosis in cultured human placental syncytiotrophoblasts. American journal of physiology Endocrinology and metabolism. 2013;305(10):E1274–80. 10.1152/ajpendo.00218.2013
    1. Chen B, Tuuli MG, Longtine MS, Shin JS, Lawrence R, Inder T, et al. Pomegranate juice and punicalagin attenuate oxidative stress and apoptosis in human placenta and in human placental trophoblasts. American journal of physiology Endocrinology and metabolism. 2012;302(9):E1142–52. 10.1152/ajpendo.00003.2012
    1. Kingdom J, Smith G. Diagnosis and management of IUGR In: Kingdom J, Baker P, editors. Intrauterine Growth Restriction Aetiology and Management. London: Springer; 2000. p. 257–73.
    1. Resnik R. Intrauterine growth restriction. Obstetrics and gynecology. 2002;99(3):490–6.
    1. Miller SL, Huppi PS, Mallard C. The consequences of fetal growth restriction on brain structure and neurodevelopmental outcome. The Journal of Physiology. 2016;594(4):807–23. 10.1113/JP271402
    1. Lodygensky GA, Seghier ML, Warfield SK, Tolsa CB, Sizonenko S, Lazeyras F, et al. Intrauterine growth restriction affects the preterm infant's hippocampus. Pediatric research. 2008;63(4):438–43. 10.1203/PDR.0b013e318165c005
    1. Tolsa CB, Zimine S, Warfield SK, Freschi M, Sancho Rossignol A, Lazeyras F, et al. Early alteration of structural and functional brain development in premature infants born with intrauterine growth restriction. Pediatric research. 2004;56(1):132–8. 10.1203/01.PDR.0000128983.54614.7E
    1. American College of Obstetricians and Gynecologists. ACOG Practice bulletin no. 134: fetal growth restriction. Obstetrics and gynecology. 2013;121(5):1122–33. 10.1097/01.AOG.0000429658.85846.f9
    1. Ben Nasr C, Ayed N, Metche M. Quantitative determination of the polyphenolic content of pomegranate peel. Zeitschrift fur Lebensmittel-Untersuchung und -Forschung. 1996;203(4):374–8.
    1. Singleton VL, Rossi JA. Colorimetry of Total Phenolics with Phosphomolybdic-Phosphotungstic Acid Reagents. 1965;16(3):144–58.
    1. Mathur AM, Neil JJ, McKinstry RC, Inder TE. Transport, monitoring, and successful brain MR imaging in unsedated neonates. Pediatr Radiol. 2008;38(3):260–4. 10.1007/s00247-007-0705-9
    1. Kidokoro H, Neil JJ, Inder TE. New MR imaging assessment tool to define brain abnormalities in very preterm infants at term. AJNR American journal of neuroradiology. 2013;34(11):2208–14. 10.3174/ajnr.A3521
    1. Nguyen The Tich S, Anderson PJ, Shimony JS, Hunt RW, Doyle LW, Inder TE. A novel quantitative simple brain metric using MR imaging for preterm infants. AJNR American journal of neuroradiology. 2009;30(1):125–31. 10.3174/ajnr.A1309
    1. Beare R, Chen J, Kelly C, Alexopoulos D, Smyser C, Rogers C, et al. Neonatal Brain Tissue Classification with Morphological Adaptation and Unified Segmentation. Frontiers in Neuroinformatics. 2016;10 10.3389/fninf.2016.00010
    1. Rogers CE, Smyser T, Smyser CD, Shimony J, Inder TE, Neil JJ. Regional white matter development in very preterm infants: perinatal predictors and early developmental outcomes. Pediatric research. 2016;79(1–1):87–95. 10.1038/pr.2015.172
    1. Smith SM, Jenkinson M, Woolrich MW, Beckmann CF, Behrens TE, Johansen-Berg H, et al. Advances in functional and structural MR image analysis and implementation as FSL. NeuroImage. 2004;23 Suppl 1:S208–19.
    1. Smyser CD, Inder TE, Shimony JS, Hill JE, Degnan AJ, Snyder AZ, et al. Longitudinal Analysis of Neural Network Development in Preterm Infants. Cerebral Cortex (New York, NY). 2010;20(12):2852–62.
    1. Herzmann C, Snyder A, Kenley J, Rogers C, Shimony J, Smyser C. Cerebellar Functional Connectivity in Term- and Very Preterm-Born Infants. Cerebral Cortex (New York, NY). 2018:10.1093/cercor/bhy023.
    1. Power JD, Barnes KA, Snyder AZ, Schlaggar BL, Petersen SE. Spurious but systematic correlations in functional connectivity MRI networks arise from subject motion. NeuroImage. 2012;59(3):2142–54. 10.1016/j.neuroimage.2011.10.018
    1. Smyser CD, Dosenbach NUF, Smyser TA, Snyder AZ, Rogers CE, Inder TE, et al. Prediction of brain maturity in infants using machine-learning algorithms. NeuroImage. 2016;136:1–9. 10.1016/j.neuroimage.2016.05.029
    1. Cohen AL, Fair DA, Dosenbach NUF, Miezin FM, Dierker D, Van Essen DC, et al. Defining Functional Areas in Individual Human Brains using Resting Functional Connectivity MRI. NeuroImage. 2008;41(1):45–57. 10.1016/j.neuroimage.2008.01.066
    1. Power JD, Cohen AL, Nelson SM, Wig GS, Barnes KA, Church JA, et al. Functional network organization of the human brain. Neuron. 2011;72(4):665–78. 10.1016/j.neuron.2011.09.006
    1. Smyser CD, Snyder AZ, Shimony JS, Mitra A, Inder TE, Neil JJ. Resting-State Network Complexity and Magnitude Are Reduced in Prematurely Born Infants. Cerebral cortex (New York, NY: 1991). 2016;26(1):322–33.
    1. Jenkins G, Watts D. Spectral Analysis and Its Applications. San Francisco, CA: Holden-Day; 1968.
    1. Smyser CD, Snyder AZ, Shimony JS, Blazey TM, Inder TE, Neil JJ. Effects of white matter injury on resting state fMRI measures in prematurely born infants. PloS one. 2013;8(7):e68098 10.1371/journal.pone.0068098
    1. Brier M, Thomas JB, Snyder AZ, Benzinger TL, Zhang D, Raichle ME, et al. Loss of Intra- and Inter-Network Resting State Functional Connections with Alzheimer’s Disease Progression. The Journal of neuroscience: the official journal of the Society for Neuroscience. 2012;32(26):8890–9.
    1. Papile LA, Burstein J, Burstein R, Koffler H. Incidence and evolution of subependymal and intraventricular hemorrhage: a study of infants with birth weights less than 1,500 gm. The Journal of pediatrics. 1978;92(4):529–34. 10.1016/s0022-3476(78)80282-0
    1. Inder TE, Warfield SK, Wang H, Huppi PS, Volpe JJ. Abnormal cerebral structure is present at term in premature infants. Pediatrics. 2005;115(2):286–94. 10.1542/peds.2004-0326
    1. Bender R, Lange S. Adjusting for multiple testing—when and how? Journal of clinical epidemiology. 2001;54(4):343–9.
    1. Rothman KJ. No adjustments are needed for multiple comparisons. Epidemiology. 1990;1(1):43–6.
    1. Olsen IE, Groveman SA, Lawson ML, Clark RH, Zemel BS. New intrauterine growth curves based on United States data. Pediatrics. 2010;125(2):e214–24. 10.1542/peds.2009-0913
    1. Ginsberg Y, Khatib N, Saadi N, Ross MG, Weiner Z, Beloosesky R. Maternal pomegranate juice attenuates maternal inflammation-induced fetal brain injury by inhibition of apoptosis, neuronal nitric oxide synthase, and NF-kappaB in a rat model. American journal of obstetrics and gynecology. 2018;219(1):113.e1–.e9.
    1. Chen B, Longtine MS, Riley JK, Nelson DM. Antenatal pomegranate juice rescues hypoxia-induced fetal growth restriction in pregnant mice while reducing placental cell stress and apoptosis. Placenta. 2018;66:1–7. 10.1016/j.placenta.2018.04.009
    1. Gluckman PD, Wyatt JS, Azzopardi D, Ballard R, Edwards AD, Ferriero DM, et al. Selective head cooling with mild systemic hypothermia after neonatal encephalopathy: multicentre randomised trial. Lancet (London, England). 2005;365(9460):663–70.
    1. Spittle AJ, Doyle LW, Anderson PJ, Inder TE, Lee KJ, Boyd RN, et al. Reduced cerebellar diameter in very preterm infants with abnormal general movements. Early Hum Dev. 2010;86(1):1–5. 10.1016/j.earlhumdev.2009.11.002
    1. Lawrence RK, Inder TE. Anatomic Changes and Imaging in Assessing Brain Injury in the Term Infant. Clinics in Perinatology. 2008;35(4):679–vi. 10.1016/j.clp.2008.07.013
    1. de Vries LS, Groenendaal F. Patterns of neonatal hypoxic–ischaemic brain injury. Neuroradiology. 2010;52(6):555–66. 10.1007/s00234-010-0674-9
    1. Panickar KS, Anderson RA. Effect of polyphenols on oxidative stress and mitochondrial dysfunction in neuronal death and brain edema in cerebral ischemia. Int J Mol Sci. 2011;12(11):8181–207. 10.3390/ijms12118181
    1. Lee H, Bae JH, Lee SR. Protective effect of green tea polyphenol EGCG against neuronal damage and brain edema after unilateral cerebral ischemia in gerbils. Journal of neuroscience research. 2004;77(6):892–900. 10.1002/jnr.20193
    1. Lee S, Suh S, Kim S. Protective effects of the green tea polyphenol (-)-epigallocatechin gallate against hippocampal neuronal damage after transient global ischemia in gerbils. Neuroscience letters. 2000;287(3):191–4. 10.1016/s0304-3940(00)01159-9
    1. Park JW, Jang YH, Kim JM, Lee H, Park WK, Lim MB, et al. Green tea polyphenol (-)-epigallocatechin gallate reduces neuronal cell damage and up-regulation of MMP-9 activity in hippocampal CA1 and CA2 areas following transient global cerebral ischemia. Journal of neuroscience research. 2009;87(2):567–75. 10.1002/jnr.21847
    1. Ritz MF, Ratajczak P, Curin Y, Cam E, Mendelowitsch A, Pinet F, et al. Chronic treatment with red wine polyphenol compounds mediates neuroprotection in a rat model of ischemic cerebral stroke. The Journal of nutrition. 2008;138(3):519–25. 10.1093/jn/138.3.519
    1. Shukla PK, Khanna VK, Ali MM, Khan MY, Srimal RC. Anti-ischemic effect of curcumin in rat brain. Neurochemical research. 2008;33(6):1036–43. 10.1007/s11064-007-9547-y
    1. Simao F, Matte A, Matte C, Soares FM, Wyse AT, Netto CA, et al. Resveratrol prevents oxidative stress and inhibition of Na(+)K(+)-ATPase activity induced by transient global cerebral ischemia in rats. The Journal of nutritional biochemistry. 2011;22(10):921–8. 10.1016/j.jnutbio.2010.07.013
    1. Della-Morte D, Dave KR, DeFazio RA, Bao YC, Raval AP, Perez-Pinzon MA. Resveratrol pretreatment protects rat brain from cerebral ischemic damage via a sirtuin 1-uncoupling protein 2 pathway. Neuroscience. 2009;159(3):993–1002. 10.1016/j.neuroscience.2009.01.017
    1. Zhao HF, Li N, Wang Q, Cheng XJ, Li XM, Liu TT. Resveratrol decreases the insoluble Aβ1–42 level in hippocampus and protects the integrity of the blood–brain barrier in AD rats. Neuroscience. 2015;310:641–9. 10.1016/j.neuroscience.2015.10.006
    1. Salberg S, Yamakawa G, Christensen J, Kolb B, Mychasiuk R. Assessment of a nutritional supplement containing resveratrol, prebiotic fiber, and omega-3 fatty acids for the prevention and treatment of mild traumatic brain injury in rats. Neuroscience. 2017;365:146–57. 10.1016/j.neuroscience.2017.09.053
    1. Chahboune H, Ment LR, Stewart WB, Rothman DL, Vaccarino FM, Hyder F, et al. Hypoxic injury during neonatal development in murine brain: correlation between in vivo DTI findings and behavioral assessment. Cerebral cortex (New York, NY: 1991). 2009;19(12):2891–901. 10.1093/cercor/bhp068
    1. Jones DK, Knösche TR, Turner R. White matter integrity, fiber count, and other fallacies: The do's and don'ts of diffusion MRI. NeuroImage. 2013;73:239–54. 10.1016/j.neuroimage.2012.06.081
    1. Wozniak JR, Lim KO. Advances in white matter imaging: a review of in vivo magnetic resonance methodologies and their applicability to the study of development and aging. Neurosci Biobehav Rev. 2006;30(6):762–74. 10.1016/j.neubiorev.2006.06.003
    1. Li K, Sun Z, Han Y, Gao L, Yuan L, Zeng D. Fractional anisotropy alterations in individuals born preterm: a diffusion tensor imaging meta-analysis. Developmental Medicine & Child Neurology. 2015;57(4):328–38.
    1. Taoka T, Kin T, Nakagawa H, Hirano M, Sakamoto M, Wada T, et al. Diffusivity and diffusion anisotropy of cerebellar peduncles in cases of spinocerebellar degenerative disease. NeuroImage. 2007;37(2):387–93. 10.1016/j.neuroimage.2007.05.028
    1. Mukherjee P, Miller J. Diffusion tensor MR imaging of gray and white matter development during normal human brain maturation. AJNR American journal of neuroradiology. 2002;23(9):1445–56.
    1. Smyser TA, Smyser CD, Rogers CE, Gillespie SK, Inder TE, Neil JJ. Cortical Gray and Adjacent White Matter Demonstrate Synchronous Maturation in Very Preterm Infants. Cereb Cortex. 2016;26(8):3370–8. 10.1093/cercor/bhv164
    1. Cascio CJ, Gerig G, Piven J. Diffusion Tensor Imaging: Application to the Study of the Developing Brain. Journal of the American Academy of Child & Adolescent Psychiatry. 2007;46(2):213–23.
    1. Gao W, Lin W, Chen Y, Gerig G, Smith JK, Jewells V, et al. Temporal and spatial development of axonal maturation and myelination of white matter in the developing brain. AJNR American journal of neuroradiology. 2009;30(2):290–6. 10.3174/ajnr.A1363
    1. Dulcich MS, Hartman RE. Pomegranate supplementation improves affective and motor behavior in mice after radiation exposure. Evidence-based complementary and alternative medicine: eCAM. 2013;2013:940830.
    1. Subash S, Braidy N, Essa MM, Zayana AB, Ragini V, Al-Adawi S, et al. Long-term (15 mo) dietary supplementation with pomegranates from Oman attenuates cognitive and behavioral deficits in a transgenic mice model of Alzheimer's disease. Nutrition (Burbank, Los Angeles County, Calif). 2015;31(1):223–9.
    1. Braidy N, Essa MM, Poljak A, Selvaraju S, Al-Adawi S, Manivasagm T, et al. Consumption of pomegranates improves synaptic function in a transgenic mice model of Alzheimer's disease. Oncotarget. 2016;7(40):64589–604. 10.18632/oncotarget.10905
    1. Fox MD, Raichle ME. Spontaneous fluctuations in brain activity observed with functional magnetic resonance imaging. Nature reviews Neuroscience. 2007;8(9):700–11. 10.1038/nrn2201
    1. Meyer A. A note on the postnatal development of the human cerebral cortex. Cerebral palsy bulletin. 1961;3:263–8.
    1. Conel J. The postnatal development of the human cerebral cortex. Cambridge (MA): Harvard University Press; 1939.
    1. Benjamini Y, Hochberg Y. Controlling the False Discovery Rate: A Practical and Powerful Approach to Multiple Testing. Journal of the Royal Statistical Society Series B (Methodological). 1995;57(1):289–300.
    1. Ruzafa N, Rey-Santano C, Mielgo V, Pereiro X, Vecino E. Effect of hypoxia on the retina and superior colliculus of neonatal pigs. PloS one. 2017;12(4):e0175301 10.1371/journal.pone.0175301
    1. Kaur C, Rathnasamy G, Ling EA. Roles of activated microglia in hypoxia induced neuroinflammation in the developing brain and the retina. Journal of neuroimmune pharmacology: the official journal of the Society on NeuroImmune Pharmacology. 2013;8(1):66–78.
    1. Rothman SM, Olney JW. Glutamate and the pathophysiology of hypoxic—ischemic brain damage. Annals of neurology. 1986;19(2):105–11. 10.1002/ana.410190202
    1. Murphy TH, Miyamoto M, Sastre A, Schnaar RL, Coyle JT. Glutamate toxicity in a neuronal cell line involves inhibition of cystine transport leading to oxidative stress. Neuron. 1989;2(6):1547–58.
    1. Hanneken A, Lin FF, Johnson J, Maher P. Flavonoids protect human retinal pigment epithelial cells from oxidative-stress-induced death. Investigative ophthalmology & visual science. 2006;47(7):3164–77.
    1. Maher P, Hanneken A. Flavonoids protect retinal ganglion cells from oxidative stress-induced death. Investigative ophthalmology & visual science. 2005;46(12):4796–803.
    1. Maher P, Hanneken A. The molecular basis of oxidative stress-induced cell death in an immortalized retinal ganglion cell line. Investigative ophthalmology & visual science. 2005;46(2):749–57.
    1. Nakayama M, Aihara M, Chen Y-N, Araie M, Tomita-Yokotani K, Iwashina T. Neuroprotective effects of flavonoids on hypoxia-, glutamate-, and oxidative stress–induced retinal ganglion cell death. Molecular Vision. 2011;17:1784–93.
    1. Andrews-Hanna JR, Snyder AZ, Vincent JL, Lustig C, Head D, Raichle ME, et al. Disruption of large-scale brain systems in advanced aging. Neuron. 2007;56(5):924–35. 10.1016/j.neuron.2007.10.038
    1. Fjell AM, Sneve MH, Storsve AB, Grydeland H, Yendiki A, Walhovd KB. Brain Events Underlying Episodic Memory Changes in Aging: A Longitudinal Investigation of Structural and Functional Connectivity. Cerebral cortex (New York, NY: 1991). 2016;26(3):1272–86.
    1. Hirsiger S, Koppelmans V, Merillat S, Liem F, Erdeniz B, Seidler RD, et al. Structural and functional connectivity in healthy aging: Associations for cognition and motor behavior. Human brain mapping. 2016;37(3):855–67. 10.1002/hbm.23067
    1. Tsang A, Lebel CA, Bray SL, Goodyear BG, Hafeez M, Sotero RC, et al. White Matter Structural Connectivity Is Not Correlated to Cortical Resting-State Functional Connectivity over the Healthy Adult Lifespan. Frontiers in Aging Neuroscience. 2017;9:144 10.3389/fnagi.2017.00144
    1. Honey CJ, Sporns O, Cammoun L, Gigandet X, Thiran JP, Meuli R, et al. Predicting human resting-state functional connectivity from structural connectivity. Proceedings of the National Academy of Sciences. 2009;106(6):2035–40.
    1. Espín JC, Larrosa M, García-Conesa MT, Tomás-Barberán F. Biological Significance of Urolithins, the Gut Microbial Ellagic Acid-Derived Metabolites: The Evidence So Far. Evidence-based complementary and alternative medicine: eCAM. 2013;2013:270418.
    1. Seeram NP, Henning SM, Zhang Y, Suchard M, Li Z, Heber D. Pomegranate juice ellagitannin metabolites are present in human plasma and some persist in urine for up to 48 hours. The Journal of nutrition. 2006;136(10):2481–5. 10.1093/jn/136.10.2481
    1. Cerda B, Espin JC, Parra S, Martinez P, Tomas-Barberan FA. The potent in vitro antioxidant ellagitannins from pomegranate juice are metabolised into bioavailable but poor antioxidant hydroxy-6H-dibenzopyran-6-one derivatives by the colonic microflora of healthy humans. European journal of nutrition. 2004;43(4):205–20. 10.1007/s00394-004-0461-7
    1. Mertens-Talcott SU, Jilma-Stohlawetz P, Rios J, Hingorani L, Derendorf H. Absorption, metabolism, and antioxidant effects of pomegranate (Punica granatum l.) polyphenols after ingestion of a standardized extract in healthy human volunteers. Journal of agricultural and food chemistry. 2006;54(23):8956–61. 10.1021/jf061674h
    1. Tomas-Barberan FA, Selma MV, Espin JC. Interactions of gut microbiota with dietary polyphenols and consequences to human health. Current opinion in clinical nutrition and metabolic care. 2016;19(6):471–6. 10.1097/MCO.0000000000000314
    1. Hammerstone JF, Lazarus SA, Schmitz HH. Procyanidin content and variation in some commonly consumed foods. The Journal of nutrition. 2000;130(8S Suppl):2086s–92s. 10.1093/jn/130.8.2086S
    1. Lacroix S, Klicic Badoux J, Scott-Boyer MP, Parolo S, Matone A, Priami C, et al. A computationally driven analysis of the polyphenol-protein interactome. Scientific reports. 2018;8(1):2232 10.1038/s41598-018-20625-5

Source: PubMed

3
Abonner