Redistribution of NMDA Receptors in Estrogen-Receptor-β-Containing Paraventricular Hypothalamic Neurons following Slow-Pressor Angiotensin II Hypertension in Female Mice with Accelerated Ovarian Failure

Neuroendocrinology. 2017;104(3):239-256. doi: 10.1159/000446073. Epub 2016 Apr 15.

Abstract

Hypertension in male and aging female rodents is associated with glutamate-dependent plasticity in the hypothalamus, but existing models have failed to capture distinct transitional menopausal phases that could have a significant impact on the synaptic plasticity and emergent hypertension. In rodents, accelerated ovarian failure (AOF) induced by systemic injection of 4-vinylcyclohexane diepoxide mimics the estrogen fluctuations seen in human menopause including the perimenopause transition (peri-AOF) and postmenopause (post-AOF). Thus, we used the mouse AOF model to determine the impact of slow-pressor angiotensin II (AngII) administration on blood pressure and on the subcellular distribution of obligatory N-methyl-D-aspartate (NMDA) receptor GluN1 subunits in the paraventricular hypothalamic nucleus (PVN), a key estrogen-responsive cardiovascular regulatory area. Estrogen-sensitive neuronal profiles were identified in mice expressing enhanced green fluorescent protein under the promoter for estrogen receptor (ER) β, a major ER in the PVN. Slow-pressor AngII increased arterial blood pressure in mice at peri- and post-AOF time points. In control oil-injected (nonhypertensive) mice, AngII decreased the total number of GluN1 in ERβ-containing PVN dendrites. In contrast, AngII resulted in a reapportionment of GluN1 from the cytoplasm to the plasma membrane of ERβ-containing PVN dendrites in peri-AOF mice. Moreover, in post-AOF mice, AngII increased total GluN1, dendritic size and radical production in ERβ-containing neurons. These results indicate that unique patterns of hypothalamic glutamate receptor plasticity and dendritic structure accompany the elevated blood pressure in peri- and post-AOF time points. Our findings suggest the possibility that distinct neurobiological processes are associated with the increased blood pressure during perimenopausal and postmenopausal periods.

Publication types

  • Research Support, N.I.H., Extramural

MeSH terms

  • Angiotensin II / toxicity
  • Animals
  • Blood Pressure / drug effects
  • Cyclohexenes / toxicity
  • Disease Models, Animal
  • Estrous Cycle / drug effects
  • Estrous Cycle / genetics
  • Female
  • Green Fluorescent Proteins / genetics
  • Green Fluorescent Proteins / metabolism
  • Hypertension* / chemically induced
  • Hypertension* / complications
  • Hypertension* / pathology
  • Mice
  • Mice, Inbred C57BL
  • Mice, Transgenic
  • Microscopy, Immunoelectron
  • Nerve Tissue Proteins / metabolism*
  • Neurons / metabolism*
  • Neurons / ultrastructure
  • Ovarian Diseases / etiology*
  • Paraventricular Hypothalamic Nucleus / metabolism
  • Paraventricular Hypothalamic Nucleus / pathology*
  • Paraventricular Hypothalamic Nucleus / ultrastructure
  • Reactive Oxygen Species / metabolism
  • Receptors, Estrogen / genetics
  • Receptors, Estrogen / metabolism*
  • Receptors, N-Methyl-D-Aspartate / metabolism*
  • Vinyl Compounds / toxicity

Substances

  • Cyclohexenes
  • Gprin1 protein, mouse
  • Nerve Tissue Proteins
  • Reactive Oxygen Species
  • Receptors, Estrogen
  • Receptors, N-Methyl-D-Aspartate
  • Vinyl Compounds
  • estrogen receptor-related receptor beta
  • Angiotensin II
  • Green Fluorescent Proteins
  • 4-vinyl-1-cyclohexene dioxide