Acylcarnitine Profiles in HIV-Exposed, Uninfected Neonates in the United States

Brian Kirmse, Tzy-Jyun Yao, Sean Hofherr, Deborah Kacanek, Paige L Williams, Charlotte V Hobbs, Rohan Hazra, William Borkowsky, Russell B Van Dyke, Marshall Summar, Brian Kirmse, Tzy-Jyun Yao, Sean Hofherr, Deborah Kacanek, Paige L Williams, Charlotte V Hobbs, Rohan Hazra, William Borkowsky, Russell B Van Dyke, Marshall Summar

Abstract

We sought to determine the prevalence of abnormal acylcarnitine profiles (ACP) in HIV-exposed uninfected (HEU) newborns and to explore the association of abnormal ACP with clinical laboratory outcomes and antiretroviral drug exposures. Clinically, ACP are used to assess for fatty acid oxidation (FAO) dysfunction and normal FAO is necessary for optimal fetal/neonatal growth and development. We analyzed serum ACP in 522 HEU neonates enrolled in the Surveillance Monitoring for ART Toxicities (SMARTT) study of the Pediatric HIV/AIDS Cohort Study (PHACS) and evaluated the associations of abnormal ACP with in utero exposure to combination antiretroviral therapy (cART) in logistic regression models, adjusting for maternal demographic, disease, and behavioral characteristics. We evaluated the associations of abnormal ACP with laboratory parameters and measures of neurodevelopment and growth. Of 522 neonates, 89 (17%) had abnormal ACP. In adjusted analyses, in utero exposure to a protease inhibitor (PI) was associated with higher odds of having an abnormal ACP [adjusted odds ratio (aOR) = 2.35, 95% CI: 0.96, 5.76, p = 0.06] with marginal significance while exposure to a nonnucleoside reverse transcriptase inhibitor (NNRTI) was associated with lower odds (aOR = 0.23, 95% CI: 0.07, 0.80, p = 0.02). Mean ALT levels were slightly higher in those with abnormal ACP, but no differences in lactate, glucose, or CPK were observed. ACP status was not associated with neurodevelopment at 1 year or growth at 2 and 3 years of age. Abnormal ACP in HEU neonates are associated with exposure to PI-containing as opposed to NNRTI-containing antiretroviral (ARV) regimens but are not associated with serious postnatal clinical problems. Further studies are needed to determine the long-term health implications of abnormal acylcarnitine metabolism at birth in HEU children.

Figures

FIG. 1.
FIG. 1.
Proportions and patterns of abnormal ACP in HEU newborns. The pattern of acylcarnitine elevations can point to the level of biochemical dysfunction in FAO. Long-chain acylcarnitine elevations (C12–C18, where the number corresponds to the number of carbons chain length of fatty acid) indicate dysfunction in the carnitine shuttle (CPT1, CACT, CPT2), which transports long-chain fatty acids into the mitochondria, or the first steps (TFP, VLCAD) of beta-oxidation, which convert long-chain fatty acids into medium-chain fatty acids. Elevations of medium-chain acylcarnitines (C6–C10) indicate dysfunction in MCAD. Short-chain acylcarnitine elevations can indicate dysfunction in SCAD (C4) or in pathways of organic acid catabolism (C3, C5, C5-OH). Generalized acylcarnitine elevations can be seen in ETF-ETF:QO dysfunction or in primary dysfunction of oxidative phosphorylation. ACP, acylcarnitine profiles; ATP, adenosine triphosphate; HEU, HIV-exposed uninfected; FAO, fatty acid oxidation; OXPHOS, oxidative phosphorylation; TCA, tricarboxylic acid cycle; CPT, carnitine palmitoyltransferase; CACT, carnitine-acylcarnitine translocase; TFP, trifunctional protein; VLCAD, very-long-chain acyl-CoA dehydrogenase; MCAD, medium-chain acyl-CoA dehydrogenase deficiency; SCAD, short-chain acyl-CoA dehydrogenase deficiency; ETF, electron transport flavoprotein; ETF:QO, electron transport flavoprotein oxidoreductase. Color images available online at www.liebertpub.com/aid

References

    1. Heidari S, Mofenson L, Cotton MF, et al. : Antiretroviral drugs for preventing mother-to-child transmission of HIV: A review of potential effects on HIV-exposed but uninfected children. J Acquir Immune Defic Syndr 2011;57(4):290–296
    1. Brogly SB, Dimauro S, Van Dyke RB, et al. : Transplacental nucleoside analogue exposure and mitochondrial parameters in HIV-uninfected children. AIDS Res Hum Retroviruses 2011;27:777–783
    1. Grinspoon S. and Carr A: Cardiovascular risk and body-fat abnormalities in HIV-infected adults. N Engl J Med 2005;352(1):48–62
    1. Gutierrez AD. and Balasubramanyam A: Dysregulation of glucose metabolism in HIV patients: Epidemiology, mechanisms, and management. Endocrine 2012;41(1):1–10
    1. Melvin AJ, Kang M, Hitti J, et al. : Cord blood lipids in infants born to HIV-1-infected women treated with combination antiretroviral therapy. Antivir Ther 2008;13(3):349–355
    1. Gregersen N. and Olsen RK: Disease mechanisms and protein structures in fatty acid oxidation defects. J Inherit Metab Dis 2010;33(5):547–553
    1. Oey NA, den Boer ME, Wijburg FA, et al. : Long-chain fatty acid oxidation during early human development. Pediatr Res 2005;57(6):755–759
    1. Wilcken B: Fatty acid oxidation disorders: Outcome and long-term prognosis. J Inherit Metab Dis 2010;33(5):501–506
    1. Adams SH, Hoppel CL, Lok KH, et al. : Plasma acylcarnitine profiles suggest incomplete long-chain fatty acid beta-oxidation and altered tricarboxylic acid cycle activity in type 2 diabetic African-American women. J Nutr 2009;139(6):1073–1081
    1. Alul FY, Cook DE, Shchelochkov OA, et al. : The heritability of metabolic profiles in newborn twins. Heredity (Edinb) 2013;110(3):253–258
    1. Shah SH, Sun JL, Stevens RD, et al. : Baseline metabolomic profiles predict cardiovascular events in patients at risk for coronary artery disease. Am Heart J 2012;163(5):844–850
    1. Wilcken B, Wiley V, Hammond J, and Carpenter K: Screening newborns for inborn errors of metabolism by tandem mass spectrometry. N Engl J Med 2003;348(23):2304–2312
    1. Rinaldo P, Cowan TM, and Matern D: Acylcarnitine profile analysis. Genet Med 2008;10(2):151–156
    1. Spiekerkoetter U, Bastin J, Gillingham M, et al. : Current issues regarding treatment of mitochondrial fatty acid oxidation disorders. J Inherit Metab Dis 2010;33(5):555–561
    1. Schooneman MG, Vaz FM, Houten SM, and Soeters MR: Acylcarnitines: Reflecting or inflicting insulin resistance? Diabetes 2013;62(1):1–8
    1. Sewell AC. and Bohles HJ: Acylcarnitines in intermediary metabolism. Eur J Pediatr 1995;154(11):871–877
    1. Kirmse B, Hobbs CV, Peter I, et al. : Abnormal newborn screening data and acylcarnitines in HIV/ARV-exposed infants. Pediatr Infect Dis J 2013;32(2):146–150
    1. Blau N, ed.: Laboratory Guide to the Methods in Biochemical Genetics (1st ed.). Springer-Verlag, Berlin, 2008
    1. Millington DS. and Stevens RD: Acylcarnitines: Analysis in plasma and whole blood using tandem mass spectrometry. Methods Mol Biol 2011;708:55–72
    1. Griner R, Williams PL, Read JS, et al. : In utero and postnatal exposure to antiretrovirals among HIV-exposed but uninfected children in the United States. AIDS Patient Care STDS 2011;25(7):385–394
    1. Tassiopoulos K, Read JS, Brogly S, et al. : Substance use in HIV-infected women during pregnancy: Self-report versus meconium analysis. AIDS Behav 2010;14(6):1269–1278
    1. Fenton TR. and Sauve RS: Using the LMS method to calculate z-scores for the Fenton preterm infant growth chart. Eur J Clin Nutr 2007;61(12):1380–1385
    1. Sirois PA, Huo Y, Williams PL, et al. : Safety of perinatal exposure to antiretroviral medications: Developmental outcomes in infants. Pediatr Infect Dis J 2013;32(6):648–655
    1. Rice ML, Zeldow B, Siberry GK, et al. : Evaluation of risk for late language emergence after in utero antiretroviral drug exposure in HIV-exposed uninfected infants. Pediatr Infect Dis J 2013;32(10):e406–413
    1. Crain MJ, Williams PL, Griner R, et al. : Point-of-care capillary blood lactate measurements in human immunodeficiency virus-uninfected children with in utero exposure to human immunodeficiency virus and antiretroviral medications. Pediatr Infect Dis J 2011;30(12):1069–1074
    1. Kirmse B, Baumgart S, and Rakhmanina N: Metabolic and mitochondrial effects of antiretroviral drug exposure in pregnancy and postpartum: Implications for fetal and future health. Semin Fetal Neonatal Med 2013;18(1):48–55
    1. Olpin SE: Pathophysiology of fatty acid oxidation disorders and resultant phenotypic variability. J Inherit Metab Dis 2013;36(4):645–658
    1. Haas RH, Parikh S, Falk MJ, et al. : The in-depth evaluation of suspected mitochondrial disease. Mol Genet Metab 2008;94(1):16–37
    1. Adler-Wailes DC, Liu H, Ahmad F, et al. : Effects of the human immunodeficiency virus-protease inhibitor, ritonavir, on basal and catecholamine-stimulated lipolysis. J Clin Endocrinol Metab 2005;90(6):3251–3261
    1. Richmond SR, Carper MJ, Lei X, et al. : HIV-protease inhibitors suppress skeletal muscle fatty acid oxidation by reducing CD36 and CPT1 fatty acid transporters. Biochim Biophys Acta 2010;1801(5):559–566
    1. Apostolova N, Blas-Garcia A, and Esplugues JV: Mitochondrial interference by anti-HIV drugs: Mechanisms beyond Pol-gamma inhibition. Trends Pharmacol Sci 2011;32(12):715–725
    1. Cassol E, Misra V, Holman A, et al. : Plasma metabolomics identifies lipid abnormalities linked to markers of inflammation, microbial translocation, and hepatic function in HIV patients receiving protease inhibitors. BMC Infect Dis 2013;13:203.
    1. Samuel VT. and Shulman G: Mechanisms for insulin resistance: Common threads and missing links. Cell 2012;148(5):852–871
    1. Penazzato M. and Giaquinto C: Role of non-nucleoside reverse transcriptase inhibitors in treating HIV-infected children. Drugs 2011;71(16):2131–2149
    1. Kao E, Shinohara M, Feng M, et al. : Human immunodeficiency virus protease inhibitors modulate Ca2+ homeostasis and potentiate alcoholic stress and injury in mice and primary mouse and human hepatocytes. Hepatology 2012;56(2):594–604
    1. Nunez M, Lana R, Mendoza JL, et al. : Risk factors for severe hepatic injury after introduction of highly active antiretroviral therapy. J Acquir Immune Defic Syndr 2001;27(5):426–431
    1. Feltes BC, de Faria Poloni J, Notari DL, and Bonatto D: Toxicological effects of the different substances in tobacco smoke on human embryonic development by a systems chemo-biology approach. PLoS One 2013;8(4):e61743.
    1. Clark RH, Kelleher AS, Chace DH, and Spitzer AR: Gestational age and age at sampling influence metabolic profiles in premature infants. Pediatrics 2014;134(1):e37–46
    1. Slaughter JL, Meinzen-Derr J, Rose SR, et al. : The effects of gestational age and birth weight on false-positive newborn-screening rates. Pediatrics 2010;126(5):910–916
    1. Tarini BA, Clark SJ, Pilli S, et al. : False-positive newborn screening result and future health care use in a state Medicaid cohort. Pediatrics 2011;128(4):715–722
    1. Yang Z, Zhao Y, Bennett MJ, et al. : Fetal genotypes and pregnancy outcomes in 35 families with mitochondrial trifunctional protein mutations. Am J Obstet Gynecol 2002;187(3):715–720
    1. Watts DH, Williams PL, Kacanek D, et al. : Combination antiretroviral use and preterm birth. J Infect Dis 2013;207(4):612–621
    1. Shneider BL, Rinaldo P, Emre S, et al. : Abnormal concentrations of esterified carnitine in bile: A feature of pediatric acute liver failure with poor prognosis. Hepatology 2005;41(4):717–721
    1. Warshaw JB: Cellular energy metabolism during fetal development. IV. Fatty acid activation, acyl transfer and fatty acid oxidation during development of the chick and rat. Dev Biol 1972;28(4):537–544
    1. Waisbren SE, Landau Y, Wilson J, and Vockley J: Neuropsychological outcomes in fatty acid oxidation disorders: 85 cases detected by newborn screening. Dev Disabil Res Rev 2013;17(3):260–268
    1. Rinaudo P. and Wang E: Fetal programming and metabolic syndrome. Annu Rev Physiol 2012;74:107–130
    1. Victora CG, Adair L, Fall C, et al. : Maternal and child undernutrition: Consequences for adult health and human capital. Lancet 2008;371(9609):340–357
    1. Ryckman KK, Smith CJ, Jelliffe-Pawlowski LL, et al. : Metabolic heritability at birth: Implications for chronic disease research. Hum Genet 2014;133(8):1049–1057
    1. El-Sadr WM, Holmes CB, Mugyenyi P, et al. : Scale-up of HIV treatment through PEPFAR: A historic public health achievement. J Acquir Immune Defic Syndr 2012;60(Suppl 3):S96–104
    1. Organization WH: Global action plan for the prevention and control of noncommunicable diseases 2013–2020, 2013

Source: PubMed

3
Suscribir