Archives

  • 2018-07
  • 2019-08
  • 2019-09
  • 2019-10
  • 2019-11
  • 2019-12
  • 2020-01
  • 2020-02
  • 2020-03
  • 2020-04
  • 2020-05
  • 2020-06
  • 2020-07
  • 2020-08
  • 2020-09
  • 2020-10
  • 2020-11
  • 2020-12
  • 2021-01
  • 2021-02
  • 2021-03
  • 2021-04
  • 2021-05
  • 2021-06
  • 2021-07
  • 2021-08
  • 2021-09
  • 2021-10
  • 2021-11
  • 2021-12
  • 2022-01
  • 2022-02
  • 2022-03
  • 2022-04
  • 2022-05
  • 2022-06
  • 2022-07
  • 2022-08
  • 2022-09
  • 2022-10
  • 2022-11
  • 2022-12
  • 2023-01
  • 2023-02
  • 2023-03
  • 2023-04
  • 2023-05
  • 2023-06
  • 2023-08
  • 2023-09
  • 2023-10
  • 2023-11
  • 2023-12
  • 2024-01
  • 2024-02
  • 2024-03
  • br Conclusions In summary aromatase mRNA

    2023-01-16


    Conclusions In summary, aromatase mRNA expression in the NVP-BEP800 of A. leptorhynchus shows a similar distribution pattern as seen in other teleosts with expression detected in the forebrain and the pituitary gland. The lack of aromatase mRNA expression in the midbrain and hindbrain of A. leptorhynchus indicates that aromatase activity is unlikely to be involved in non-breeding electric signaling behavior in this species. Future studies should explore potential seasonal changes in the amount and distribution of aromatase mRNA expression in male and female A. leptorhynchus, as well as to extend the study of aromatase expression to other electrogenic species. To the best of our knowledge, this is the first experiment conducted to assess the neural expression pattern of aromatase mRNA in the brain of an electric fish and we hope that its findings will help to inspire further study of its potential role in electric signaling behavior.
    Conflicts of interest
    Ethics statement
    Acknowledgments The authors would like to thank Brenda Toscano-Márquez and Erik Harvey-Girard for technical advice. We would also like to thank Dr. Michael Hendricks for the use of equipment for sequencing the aromatase gene. This work was supported by a Discovery grant from the Natural Sciences and Engineering Research Council of Canada (NSERC) to R.K. (#2014-05364) and by a NSERC Canada Graduate Scholarship- Master’s Program to K.S.
    Introduction Stroke is a leading cause of mortality and morbidity in the industrialized world, with an incidence of approximately 350 cases per 100,000 individuals [1]. At present, thrombolysis using recombinant tissue plasminogen activator (tPA) is the only therapy for acute Ischemic stroke approved by the U.S. food and drug Administration. Nevertheless, since the therapeutic window is limited to 3h post onset of stroke, only a minority of patients with acute ischemic stroke benefit from therapy with thrombolytics [2], [3]. Therefore, the search for efficacious therapies remains a priority. Estrogen was proposed as a neuroprotective agent in a variety of experimental models, including global and focal ischemia, affording histological protection in young and middle-aged female and male rodents. Outcome from experimental and human stroke and cerebral ischemia is strongly dependent on sex, such that young women and female animals are protected from ischemic brain damage relative to their age-matched males. This protection is thought to be mediated by estradiol [4], [5], [6], [7], [8], [9], [10]. Aromatase is the last and obligatory enzyme catalyzing estrogen biosynthesis from androgenic precursors and the primary source of brain estrogen in men and post-menopausal women. The expression of aromatase is reportedly increased in the rat brain after neurotoxic damage and experimental stroke, and accumulating evidence suggests that aromatase exerts neuroprotection through local (extragonadal) synthesis of estrogens. The majority of these studies, including the NVP-BEP800 only study documenting the time course and topography of aromatase following experimental stroke, were performed in females [11], [12], [13], [14], [15]. Neuroinflammation is believed to play an important role in the neuropathological processes after the onset of cerebral ischemia. Neuroinflammation involves the activation of glia, including microglia and astrocytes [16]. It was recently suggested that neuroinflammation, even in the absence of mechanical or ischemic injury, can increase glial aromatase expression in the bird brain [17]. Conversely, exogenous estrogen was shown to reduce neuroinflammation in experimental stroke in male rats [8], but the relationship between aromatase expression and neuroinflammation following experimental stroke in male rats has not been elaborated to date. The regional intensity of the neuroinflammatory response can be examined by autoradiography with [3H]PK11195, which was found to be an excellent quantitative marker for neuroinflammation and microgliosis in the rodent and human brain [18], [19]. The ligand binds to the translocator protein 18kDa (TSPO, previously named the peripheral benzodiazepine receptors), which is only sparsely expressed in the healthy brain parenchyma. However, this protein is drastically upregulated under neuroinflammatory conditions and this upregulation correlates with activation, hypertrophy, and proliferation of glial cells [20], [21]. We and others have shown that upregulation of TSPO in stroke models lasts more than 30days in females [22] and at least 2 weeks in males [23], [24].