Am J Perinatol 2018; 35(11): 1100-1106
DOI: 10.1055/s-0038-1641169
Original Article
Thieme Medical Publishers 333 Seventh Avenue, New York, NY 10001, USA.

Acute Hypoxia Induces Enkephalin Production and Release in an Adrenergic Cell Line Model of Neonatal Chromaffin Cell Responses to Hypoxic Stress

Alexa Calero
1   Department of Pediatrics, Stony Brook Children's Hospital, Stony Brook, New York
2   Division of Neonatology, Stony Brook Children's Hospital, Stony Brook, New York
,
Hazel Villanueva
1   Department of Pediatrics, Stony Brook Children's Hospital, Stony Brook, New York
2   Division of Neonatology, Stony Brook Children's Hospital, Stony Brook, New York
,
Priyadarshani Giri
1   Department of Pediatrics, Stony Brook Children's Hospital, Stony Brook, New York
,
Arthur S. Tischler
3   Department of Pathology, Tufts University Medical Center, Boston, Massachusetts
,
James F. Powers
3   Department of Pathology, Tufts University Medical Center, Boston, Massachusetts
,
Marian Evinger
1   Department of Pediatrics, Stony Brook Children's Hospital, Stony Brook, New York
› Author Affiliations
Funding This study was funded by internal funds provided by the Department of Pediatrics at Stony Brook University Hospital.
Further Information

Publication History

30 October 2017

26 February 2018

Publication Date:
10 April 2018 (online)

Abstract

Objective Prior to maturation of the human sympathetic nervous system, the neonatal adrenal medulla senses and responds to hypoxia. In addition to catecholamine release, the adrenal medulla synthesizes and stores opioid peptides, notably enkephalin (ENK). However, it is not known whether acute hypoxia evokes adrenal ENK production and release, as seen in the central nervous system (CNS). We hypothesize that acute hypoxia stimulates synthesis and release of ENK in chromaffin cells.

Study Design Cultures of adrenergic mouse pheochromocytoma cells (MPC) 10/9/96CR were incubated in 10% oxygen (O2) at intervals of up to 60 minutes. ENK content and release were measured by Met-ENK enzyme-linked immunosorbent assay (ELISA). ENK messenger ribonucleic acid (mRNA) was analyzed by quantitative reverse-transcriptase polymerase chain reaction (PCR).

Results Incubation of MPC 10/9 cells in 10% O2 evoked rapid release of epinephrine and of Met-ENK which increased approximately twofold in 15 minutes. Reduced [O2] also induced an overall increase (14%) in cellular ENK peptide content within 60 minutes. Acute hypoxia-stimulated release of Met-ENK was accompanied by increased mRNAENK expression in MPC 10/9s, a cell culture model of adrenergic chromaffin cells.

Conclusion We speculate that the ability of reduced [O2] to evoke ENK release from chromaffin cells may influence blood pressure regulation and heart contractility, thereby providing an adaptive survival advantage during neonatal asphyxia.

 
  • References

  • 1 Oza S, Lawn JE, Hogan DR, Mathers C, Cousens SN. Neonatal cause-of-death estimates for the early and late neonatal periods for 194 countries: 2000-2013. Bull World Health Organ 2015; 93 (01) 19-28
  • 2 Martin RJ, Wang K, Köroğlu O, Di Fiore J, Kc P. Intermittent hypoxic episodes in preterm infants: do they matter?. Neonatology 2011; 100 (03) 303-310
  • 3 Michiels C. Physiological and pathological responses to hypoxia. Am J Pathol 2004; 164 (06) 1875-1882
  • 4 Prabhakar NR, Semenza GL. Adaptive and maladaptive cardiorespiratory responses to continuous and intermittent hypoxia mediated by hypoxia-inducible factors 1 and 2. Physiol Rev 2012; 92 (03) 967-1003
  • 5 Lagercrantz H, Marcus C. Sympathoadrenal Mechanisms during Development. In Polin R, Fox W. , eds., Fetal and Neonatal Physiology. Vol 1. Philadelphia: W.B. Saunders; 1992
  • 6 Rychkov GY, Adams MB, McMillen IC, Roberts ML. Oxygen-sensing mechanisms are present in the chromaffin cells of the sheep adrenal medulla before birth. J Physiol 1998; 509 (Pt 3): 887-893
  • 7 Duman JG, Chen L, Hille B. Calcium transport mechanisms of PC12 cells. J Gen Physiol 2008; 131 (04) 307-323
  • 8 García-Fernández M, Mejías R, López-Barneo J. Developmental changes of chromaffin cell secretory response to hypoxia studied in thin adrenal slices. Pflugers Arch 2007; 454 (01) 93-100
  • 9 Nurse CA, Buttigieg J, Brown S, Holloway AC. Regulation of oxygen sensitivity in adrenal chromaffin cells. Ann N Y Acad Sci 2009; 1177: 132-139
  • 10 Rico AJ, Prieto-Lloret J, Gonzalez C, Rigual R. Hypoxia and acidosis increase the secretion of catecholamines in the neonatal rat adrenal medulla: an in vitro study. Am J Physiol Cell Physiol 2005; 289 (06) C1417-C1425
  • 11 Schmitt P, Garcia C, Soulier V, Pujol JF, Pequignot JM. Influence of long-term hypoxia on tyrosine hydroxylase in the rat carotid body and adrenal gland. J Auton Nerv Syst 1992; 40 (01) 13-19
  • 12 Feinsilver SH, Wong R, Raybin DM. Adaptations of neurotransmitter synthesis to chronic hypoxia in cell culture. Biochim Biophys Acta 1987; 928 (01) 56-62
  • 13 Millhorn DE, Raymond R, Conforti L. , et al. Regulation of gene expression for tyrosine hydroxylase in oxygen sensitive cells by hypoxia. Kidney Int 1997; 51 (02) 527-535
  • 14 Millhorn DE, Czyzyk-Krzeska M, Bayliss DA, Lawson EE. Regulation of gene expression by hypoxia. Sleep 1993; 16 (8, Suppl) S44-S48
  • 15 DeCristofaro JD, LaGamma EF. Neonatal stress: effects of hypoglycemia and hypoxia on adrenal tyrosine hydroxylase gene expression. Pediatr Res 1994; 36 (06) 719-723
  • 16 Wickström HR, Mas C, Simonneau M, Holgert H, Hökfelt T, Lagercrantz H. Perinatal nicotine attenuates the hypoxia-induced up-regulation of tyrosine hydroxylase and galanin mRNA in locus ceruleus of the newborn mouse. Pediatr Res 2002; 52 (05) 763-769
  • 17 Adams MB, McMillen IC. Actions of hypoxia on catecholamine synthetic enzyme mRNA expression before and after development of adrenal innervation in the sheep fetus. J Physiol 2000; 529 (Pt 3): 519-531
  • 18 Evinger MJ, He R, Powers JF, Tischler AS. Hypoxia Stimulates PNMT Gene Expression. Paper presented at: 2000 Society for Neuroscience Planner; 2000; New Orleans, LA
  • 19 Chatow U, Davidson S, Reichman BL, Akselrod S. Development and maturation of the autonomic nervous system in premature and full-term infants using spectral analysis of heart rate fluctuations. Pediatr Res 1995; 37 (03) 294-302
  • 20 Borges R, Dominguez N, Smith CB. , et al. Granins and catecholamines: functional interaction in chromaffin cells and adipose tissue. Adv Pharmacol 2013; 68: 93-113
  • 21 Pellizzari EH, Barontini M, Figuerola MdeL, Cigorraga SB, Levin G. Possible autocrine enkephalin regulation of catecholamine release in human pheochromocytoma cells. Life Sci 2008; 83 (11-12): 413-420
  • 22 Livett BG, Dean DM, Whelan LG, Udenfriend S, Rossier J. Co-release of enkephalin and catecholamines from cultured adrenal chromaffin cells. Nature 1981; 289 (5795): 317-319
  • 23 Varndell IM, Tapia FJ, De Mey J, Rush RA, Bloom SR, Polak JM. Electron immunocytochemical localization of enkephalin-like material in catecholamine-containing cells of the carotid body, the adrenal medulla, and in pheochromocytomas of man and other mammals. J Histochem Cytochem 1982; 30 (07) 682-690
  • 24 Viveros OH, Wilson SP, Chang KJ. Regulation of synthesis and secretion of enkephalins and related peptides in adrenomedullary chromaffin cells and human pheochromocytoma. Adv Biochem Psychopharmacol 1982; 33: 217-224
  • 25 Wilson SP, Chang KJ, Viveros OH. Proportional secretion of opioid peptides and catecholamines from adrenal chromaffin cells in culture. J Neurosci 1982; 2 (08) 1150-1156
  • 26 Gingras JL, Long WA. Chronic maternal hypoxia. Effect of mid-gestational maternal hypoxia on methionine-enkephalin concentrations within pre- and postnatal rabbit brainstem regions. Dev Neurosci 1988; 10 (03) 180-189
  • 27 Kumar GK, Overholt JL, Bright GR. , et al. Release of dopamine and norepinephrine by hypoxia from PC-12 cells. Am J Physiol 1998; 274 (6 Pt 1): C1592-C1600
  • 28 Lagercrantz H, Slotkin TA. The “stress” of being born. Sci Am 1986; 254 (04) 100-107
  • 29 Powers JF, Evinger MJ, Tsokas P. , et al. Pheochromocytoma cell lines from heterozygous neurofibromatosis knockout mice. Cell Tissue Res 2000; 302 (03) 309-320
  • 30 Evinger MJ, Mathew E, Cikos S. , et al. Nicotine stimulates expression of the PNMT gene through a novel promoter sequence. J Mol Neurosci 2005; 26 (01) 39-55
  • 31 Tischler AS, Powers JF, Alroy J. Animal models of pheochromocytoma. Histol Histopathol 2004; 19 (03) 883-895
  • 32 Tischler AS, Shih TS, Williams BO, Jacks T. Characterization of pheochromocytomas in a mouse strain with a targeted disruptive mutation of the neurofibromatosis gene Nf1. Endocr Pathol 1995; 6 (04) 323-335
  • 33 Jacks T, Shih TS, Schmitt EM, Bronson RT, Bernards A, Weinberg RA. Tumour predisposition in mice heterozygous for a targeted mutation in Nf1. Nat Genet 1994; 7 (03) 353-361
  • 34 Taylor SC, Peers C. Chronic hypoxia enhances the secretory response of rat phaeochromocytoma cells to acute hypoxia. J Physiol 1999; 514 (Pt 2): 483-491
  • 35 Anton AH, Sayre DF. A study of the factors affecting the aluminum oxide-trihydroxyindole procedure for the analysis of catecholamines. J Pharmacol Exp Ther 1962; 138 (03) 360-375
  • 36 Tönshoff C, Hemmick L, Evinger MJ. Pituitary adenylate cyclase activating polypeptide (PACAP) regulates expression of catecholamine biosynthetic enzyme genes in bovine adrenal chromaffin cells. J Mol Neurosci 1997; 9 (02) 127-140
  • 37 Martinez AM, Padbury JF, Burnell EE, Thio SL, Humme J. The effects of hypoxia on (methionine) enkephalin peptide and catecholamine release in fetal sheep. Pediatr Res 1990; 27 (01) 52-55
  • 38 Evinger MJ, Cikos S, Nwafor-Anene V, Powers JF, Tischler AS. Hypoxia activates multiple transcriptional pathways in mouse pheochromocytoma cells. Ann N Y Acad Sci 2002; 971: 61-65
  • 39 Thompson RJ, Jackson A, Nurse CA. Developmental loss of hypoxic chemosensitivity in rat adrenomedullary chromaffin cells. J Physiol 1997; 498 (Pt 2): 503-510
  • 40 Mizobe F, Livett BG. Nicotine stimulates secretion of both catecholamines and acetylcholinesterase from cultured adrenal chromaffin cells. J Neurosci 1983; 3 (04) 871-876
  • 41 Edwards AV, Jones CT. Adrenal responses to splanchnic nerve stimulation in conscious calves given naloxone. J Physiol 1989; 418: 339-351
  • 42 Pagida MA, Konstantinidou AE, Korelidou A. , et al. The effect of perinatal hypoxic/ischemic injury on tyrosine hydroxylase expression in the locus coeruleus of the human neonate. Dev Neurosci 2016; 38 (01) 41-53
  • 43 Crivellato E, Nico B, Ribatti D. The chromaffin vesicle: advances in understanding the composition of a versatile, multifunctional secretory organelle. Anat Rec (Hoboken) 2008; 291 (12) 1587-1602
  • 44 La Gamma EF, Agarwal BL, DeCristofaro JD. Regulation of adrenomedullary preproenkephalin mRNA: effects of hypoglycemia during development. Brain Res Mol Brain Res 1992; 13 (03) 189-197
  • 45 Mole DR, Blancher C, Copley RR. , et al. Genome-wide association of hypoxia-inducible factor (HIF)-1alpha and HIF-2alpha DNA binding with expression profiling of hypoxia-inducible transcripts. J Biol Chem 2009; 284 (25) 16767-16775
  • 46 Paulding WR, Schnell PO, Bauer AL. , et al. Regulation of gene expression for neurotransmitters during adaptation to hypoxia in oxygen-sensitive neuroendocrine cells. Microsc Res Tech 2002; 59 (03) 178-187
  • 47 Eiden LE, Ruth JA. Enkephalins modulate the responsiveness of rat atria in vitro to norepinephrine. Peptides 1982; 3 (03) 475-478