Next Article in Journal
Making the Rounds: Exploring the Role of Circulating Tumor DNA (ctDNA) in Non-Small Cell Lung Cancer
Previous Article in Journal
Microfluidic Approaches for Affinity-Based Exosome Separation
Previous Article in Special Issue
Roles of Oxytocin in Stress Responses, Allostasis and Resilience
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Editorial

Molecular Mechanisms Underlying Stress Response and Resilience

by
Kazunori Kageyama
1,* and
Takahiro Nemoto
2
1
Department of Endocrinology and Metabolism, Hirosaki University Graduate School of Medicine, 5 Zaifu-cho, Hirosaki, Aomori 036-8562, Japan
2
Department of Bioregulatory Science (Physiology), Nippon Medical School, Tokyo 113-8602, Japan
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2022, 23(16), 9007; https://doi.org/10.3390/ijms23169007
Submission received: 22 July 2022 / Accepted: 11 August 2022 / Published: 12 August 2022
(This article belongs to the Special Issue Molecular Mechanisms Underlying Stress Response and Resilience)
A variety of stressors induce various physiological responses by modulating sympathetic, neuroendocrine, and behavioral systems. Stress activates the hypothalamic–pituitary–adrenal (HPA) axis, whereas it suppresses the hypothalamic–pituitary–gonadal axis. The stress response also includes a physiological system for stress adaptation. The stressed state may be relieved or reduced by an action, medicine, or therapy. This Special Issue focuses on the stress response and resilience, which are related to their molecular mechanisms. In this Special Issue, two studies report the importance of elucidating stress mechanisms and stress-related translational research. Otubo et al. (2021) reported that, in Japanese macaque monkeys, both arginine vasopressin (AVP)/corticotropin-releasing factor (CRF) and the vesicular glutamate transporter 2/glutamate were co-localized in the magnocellular endings of the posterior pituitary and the parvocellular endings in the external layer of the median eminence [1]. Therefore, magnocellular and parvocellular AVP neurons are glutamatergic in primates. Theodoridi et al. (2021) generated a 11β-hydroxysteroid dehydrogenase type 2 (Hsd11b2) zebrafish mutant line to evaluate the involvement of this gene in stress-response regulation [2]. This new zebrafish model may represent an excellent tool for studying the response to different stressors and identifying cortisol-induced mechanisms during the stress response.
CRF in the hypothalamus is a crucial hormone to control the stress response. CRF in the hypothalamic paraventricular nucleus (PVN) regulates the HPA axis [3]. CRF receptor type 1 (CRF1 receptor) is mainly involved in stress responses, depression, anorexia, and seizures. The CRF2 receptor mediates “stress coping” responses, such as anxiolysis in the brain [3]. Orexin A upregulates CRF1 receptor expression, and CRF induces Pomc transcription [4]. Orexin A also suppresses bone morphogenetic protein (BMP)-4-induced intracellular signaling molecules [4]. Thus, orexin A has a stimulatory effect on Pomc expression by upregulating and downregulating CRF and BMP signaling, respectively. The orexin system is involved in the control of stress response and resilience. Pituitary BMPs are involved in the secretory control of follicle-stimulating hormone produced by gonadotroph cells. Soejima et al. (2021) elucidated the effects of Clock gene suppression and BMPs on the expression of luteinizing hormone (Lh) and Clock genes induced by gonadotropin-releasing hormone in gonadotroph cells [5]. Control of Clock gene expression by regulating the extracellular signal-regulated kinase pathway in the early phase and the BMP-Smad signal in the chronic phase could be a future strategy for modulating the LH surge [5].
Glucocorticoids are essential for stress coping, stress resilience, and homeostasis. The FK506-binding immunophilins FKBP4 and FKBP5 differentially regulate dynein interaction and nuclear translocation of the glucocorticoid receptor, resulting in the modulation of the glucocorticoid action [6]. FKBP4 contributes to the negative feedback of glucocorticoids, whereas FKBP5 reduces the efficiency of the glucocorticoid effect on proopiomelanocortin (Pomc) gene expression in pituitary corticotroph cells [6]. Low body weight at birth is a risk factor for future metabolic disorders, stress response abnormalities, and depression. Nemoto and Kakinuma (2021) observed impaired glucocorticoid responsiveness in low-body-weight rats due to malnutrition during the embryonic period in a pituitary-specific manner [7]. The authors also described that the expression of growth arrest-specific 5 (Gas5) lncRNA, a decoy receptor for the glucocorticoid receptor, increased only in the pituitary, and the expression of the Fkbp5 against dexamethasone was induced in the liver, muscle, and adipose tissue. The dampened glucocorticoid response may be related to elevated Gas5 lncRNA expression levels. Prenatal or postnatal methyl-modulator intervention can attenuate Gas5 lncRNA expression in the pituitary.
Stress affects eating behavior. A complex network of appetite-related peptides controls appetite. Among various factors, ghrelin is a critical regulator of metabolism or energy homeostasis. Dexamethasone reportedly increased Ghrelin mRNA levels and its receptor mRNA and protein levels in hypothalamic cells [8]. Yamada (2021) demonstrated the relationship between the appetite-promoting peptide ghrelin and stress [9]. The orexigenic peptide, ghrelin, mediates stress-induced eating disorders by abnormal secretion and signal transduction. Disturbances in ghrelin signals are also influenced by aging and sex [9]. Sex differences regulate ghrelin responsiveness, and aging triggers a persistent stress response.
Oxytocin is also stimulated by various stressful stimuli. Oxytocin is related to anxiety suppression and stress resilience [10]. The oxytocin system directly and indirectly exhibits adaptive changes dependent on internal and external conditions. Oxytocin in the hypothalamus has been shown to facilitate affiliative social behaviors and induce anxiolytic actions. Post-weaning stroking procedures reportedly induced affiliative responses via the activation of oxytocin neurons in the caudal PVN [11]. The oxytocin receptor also shows plastic changes during adaptation. Effective activation of the endogenous oxytocin system may strengthen the ability of the individual to adapt and facilitate resilience. Therefore, modulation of the oxytocin system may be valuable for preventing lifestyle-related or early life trauma-related diseases and health promotion. Al Jowf et al. (2021) provided an overview of various molecular biological, biochemical, and physiological alterations in post-traumatic stress disorder (PTSD), focusing on changes at the genomic and epigenomic levels [12]. PTSD is characterized by structural and functional brain changes. The underlying metabolic effects of conventional pharmacotherapies for PTSD are linked to epigenetic changes, which also occur throughout the disease progression.
Various factors contribute to stress resilience. All these factors have differing roles in stress resilience. The stressed state may be relieved or reduced via the HPA axis and other hormones by therapy, medicines, or natural stimulation. Successful therapy results in normal regulation of the HPA axis. To adequately treat stress-related diseases, molecular mechanisms underlying the stress response and resilience must be further explored.

Funding

This research received no external funding.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Otubo, A.; Maejima, S.; Oti, T.; Satoh, K.; Ueda, Y.; Morris, J.F.; Sakamoto, T.; Sakamoto, H. Immunoelectron microscopic characterization of vasopressin-producing neurons in the hypothalamo-pituitary axis of non-human primates by use of formaldehyde-fixed tissues stored at −25 °C for several years. Int. J. Mol. Sci. 2021, 22, 9180. [Google Scholar] [CrossRef] [PubMed]
  2. Theodoridi, A.; Dinarello, A.; Badenetti, L.; Pavlidis, M.; Dalla Valle, L.; Tsalafouta, A. Knockout of the hsd11b2 gene extends the cortisol stress response in both zebrafish larvae and adults. Int. J. Mol. Sci. 2021, 22, 12525. [Google Scholar] [CrossRef] [PubMed]
  3. Kageyama, K.; Iwasaki, Y.; Daimon, M. Hypothalamic regulation of corticotropin-releasing factor under stress and stress resilience. Int. J. Mol. Sci. 2021, 22, 12242. [Google Scholar] [CrossRef] [PubMed]
  4. Fujisawa, S.; Komatsubara, M.; Tsukamoto-Yamauchi, N.; Iwata, N.; Nada, T.; Wada, J.; Otsuka, F. Orexin A enhances pro-opiomelanocortin transcription regulated by BMP-4 in mouse corticotrope AtT20 cells. Int. J. Mol. Sci. 2021, 22, 4553. [Google Scholar] [CrossRef] [PubMed]
  5. Soejima, Y.; Iwata, N.; Nakano, Y.; Yamamoto, K.; Suyama, A.; Nada, T.; Otsuka, F. Biphasic roles of clock genes and bone morphogenetic proteins in gonadotropin expression by mouse gonadotrope cells. Int. J. Mol. Sci. 2021, 22, 11186. [Google Scholar] [CrossRef] [PubMed]
  6. Kageyama, K.; Iwasaki, Y.; Watanuki, Y.; Niioka, K.; Daimon, M. Differential effects of Fkbp4 and Fkbp5 on regulation of the proopiomelanocortin gene in murine AtT-20 corticotroph cells. Int. J. Mol. Sci. 2021, 22, 5724. [Google Scholar] [CrossRef] [PubMed]
  7. Nemoto, T.; Kakinuma, Y. Prenatal and postnatal methyl-modulator intervention corrects the stress-induced glucocorticoid response in low-birthweight rats. Int. J. Mol. Sci. 2021, 22, 9767. [Google Scholar] [CrossRef] [PubMed]
  8. Kageyama, K.; Akimoto, K.; Yamagata, S.; Sugiyama, A.; Murasawa, S.; Watanuki, Y.; Tamasawa, N.; Suda, T. Dexamethasone stimulates the expression of ghrelin and its receptor in rat hypothalamic 4B cells. Regul. Pept. 2012, 174, 12–17. [Google Scholar] [CrossRef] [PubMed]
  9. Yamada, C. Involvement of ghrelin dynamics in stress-induced eating disorder: Effects of sex and aging. Int. J. Mol. Sci. 2021, 22, 11695. [Google Scholar] [CrossRef] [PubMed]
  10. Takayanagi, Y.; Onaka, T. Roles of oxytocin in stress responses, allostasis and resilience. Int. J. Mol. Sci. 2021, 23, 150. [Google Scholar] [CrossRef] [PubMed]
  11. Okabe, S.; Takayanagi, Y.; Yoshida, M.; Onaka, T. Gentle stroking stimuli induce affiliative responsiveness to humans in male rats. Sci. Rep. 2020, 10, 9135. [Google Scholar] [CrossRef] [PubMed]
  12. Al Jowf, G.I.; Snijders, C.; Rutten, B.P.F.; de Nijs, L.; Eijssen, L.M.T. The molecular biology of susceptibility to post-traumatic stress disorder: Highlights of epigenetics and Epigenomics. Int. J. Mol. Sci. 2021, 22, 10743. [Google Scholar] [CrossRef] [PubMed]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Kageyama, K.; Nemoto, T. Molecular Mechanisms Underlying Stress Response and Resilience. Int. J. Mol. Sci. 2022, 23, 9007. https://doi.org/10.3390/ijms23169007

AMA Style

Kageyama K, Nemoto T. Molecular Mechanisms Underlying Stress Response and Resilience. International Journal of Molecular Sciences. 2022; 23(16):9007. https://doi.org/10.3390/ijms23169007

Chicago/Turabian Style

Kageyama, Kazunori, and Takahiro Nemoto. 2022. "Molecular Mechanisms Underlying Stress Response and Resilience" International Journal of Molecular Sciences 23, no. 16: 9007. https://doi.org/10.3390/ijms23169007

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop