1
|
Alewel DI, Kodavanti UP. Neuroendocrine contribution to sex-related variations in adverse air pollution health effects. JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH. PART B, CRITICAL REVIEWS 2024; 27:287-314. [PMID: 39075643 DOI: 10.1080/10937404.2024.2383637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/31/2024]
Abstract
Air pollution exposure is ranked as a leading environmental risk factor for not only cardiopulmonary diseases but also for systemic health ailments including diabetes, reproductive abnormalities, and neuropsychiatric disorders, likely mediated by central neural stress mechanisms. Current experimental evidence links many air pollution health outcomes with activation of neuroendocrine sympathetic-adrenal-medullary and hypothalamic-pituitary-adrenal (HPA) stress axes associated with resultant increases in adrenal-derived hormone levels acting as circulating mediators of multi-organ stress reactions. Epidemiological and experimental investigations also demonstrated sex-specific responses to air pollutant inhalation, which may be attributed to hormonal interactions within the stress and reproductive axes. Sex hormones (androgens and estrogens) interact with neuroendocrine functions to influence hypothalamic responses, subsequently augmenting stress-mediated metabolic and immune changes. These neurohormonal interactions may contribute to innate sex-specific responses to inhaled irritants, inducing differing individual susceptibility. The aim of this review was to: (1) examine neuroendocrine co-regulation of the HPA axis by gonadal hormones, (2) provide experimental evidence demonstrating sex-specific respiratory and systemic effects attributed to air pollutant inhalation exposure, and (3) postulate proposed mechanisms of stress and sex hormone interactions during air pollution-related stress.
Collapse
Affiliation(s)
- Devin I Alewel
- Oak Ridge Institute for Science and Education Research Participation Program, U.S. Environmental Protection Agency, Research Triangle Park, NC, USA
| | - Urmila P Kodavanti
- Public Health and Integrated Toxicology Division, Center for Public Health and Environmental Assessment, U.S. Environmental Protection Agency, Research Triangle Park, NC, USA
| |
Collapse
|
2
|
Li YM, Shen CY, Jiang JG. Sedative and hypnotic effects of the saponins from a traditional edible plant Liriope spicata Lour. in PCPA-induced insomnia mice. JOURNAL OF ETHNOPHARMACOLOGY 2024; 327:118049. [PMID: 38484954 DOI: 10.1016/j.jep.2024.118049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 03/07/2024] [Accepted: 03/11/2024] [Indexed: 03/21/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Liriope spicata Lour., a species listed in the catalogue of 'Medicinal and Edible Homologous Species', is traditionally used for the treatment of fatigue, restlessness, insomnia and constipation. AIM OF THE STUDY This study is aimed to evaluate the sedative and hypnotic effect of the saponins from a natural plant L. spicata Lour. in vivo. MATERIALS AND METHODS The total saponin (LSTS) and purified saponin (LSPS) were extracted from L. spicata, followed by a thorough analysis of their major components using the HPLC-MS. Subsequently, the therapeutic efficacy of LSTS and LSPS was evaluated by the improvement of anxiety and depression behaviors of the PCPA-induced mice. RESULTS LSTS and LSPS exhibited similar saponin compositions but differ in their composition ratios, with liriopesides-type saponins accounting for a larger proportion in LSTS. Studies demonstrated that both LSTS and LSPS can extend sleep duration and immobility time, while reducing sleep latency in PCPA-induced mice. However, there was no significant difference in weight change among the various mice groups. Elisa results indicated that the LSTS and LSPS could decrease levels of NE, DA, IL-6, and elevate the levels of 5-HT, NO, PGD2 and TNF-α in mice plasma. LSTS enhanced the expression of neurotransmitter receptors, while LSPS exhibited a more pronounced effect in regulating the expression of inflammatory factors. In conclusion, the saponins derived from L. spicata might hold promise as ingredients for developing health foods with sedative and hypnotic effects, potentially related to the modulation of serotonergic and GABAAergic neuron expression, as well as immunomodulatory process.
Collapse
Affiliation(s)
- Yi-Meng Li
- School of Food Science and Engineering, South China University of Technology, Guangzhou, 510640, China
| | - Chun-Yan Shen
- School of Food Science and Engineering, South China University of Technology, Guangzhou, 510640, China; Southern Medical University, School of Traditional Chinese Medicine, Guangzhou, 510515, China
| | - Jian-Guo Jiang
- School of Food Science and Engineering, South China University of Technology, Guangzhou, 510640, China.
| |
Collapse
|
3
|
Czeisler MÉ, Shan Y, Schalek R, Berger DR, Suissa-Peleg A, Takahashi JS, Lichtman JW. Extensive soma-soma plate-like contact sites (ephapses) connect suprachiasmatic nucleus neurons. J Comp Neurol 2024; 532:e25624. [PMID: 38896499 PMCID: PMC11419332 DOI: 10.1002/cne.25624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2022] [Revised: 03/30/2024] [Accepted: 04/29/2024] [Indexed: 06/21/2024]
Abstract
The hypothalamic suprachiasmatic nucleus (SCN) is the central pacemaker for mammalian circadian rhythms. As such, this ensemble of cell-autonomous neuronal oscillators with divergent periods must maintain coordinated oscillations. To investigate ultrastructural features enabling such synchronization, 805 coronal ultrathin sections of mouse SCN tissue were imaged with electron microscopy and aligned into a volumetric stack, from which selected neurons within the SCN core were reconstructed in silico. We found that clustered SCN core neurons were physically connected to each other via multiple large soma-to-soma plate-like contacts. In some cases, a sliver of a glial process was interleaved. These contacts were large, covering on average ∼21% of apposing neuronal somata. It is possible that contacts may be the electrophysiological substrate for synchronization between SCN neurons. Such plate-like contacts may explain why the synchronization of SCN neurons is maintained even when chemical synaptic transmission or electrical synaptic transmission via gap junctions is blocked. Such ephaptic contact-mediated synchronization among nearby neurons may therefore contribute to the wave-like oscillations of circadian core clock genes and calcium signals observed in the SCN.
Collapse
Affiliation(s)
- Mark É. Czeisler
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA, 02138, USA
| | - Yongli Shan
- Department of Neuroscience, Peter O’Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390-9111, USA
| | - Richard Schalek
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA, 02138, USA
| | - Daniel R. Berger
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA, 02138, USA
| | - Adi Suissa-Peleg
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA, 02138, USA
| | - Joseph S. Takahashi
- Department of Neuroscience, Peter O’Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390-9111, USA
- Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390-9111, USA
| | - Jeff W. Lichtman
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA, 02138, USA
| |
Collapse
|
4
|
Branca C, Bortolato M. The role of neuroactive steroids in tic disorders. Neurosci Biobehav Rev 2024; 160:105637. [PMID: 38519023 PMCID: PMC11121756 DOI: 10.1016/j.neubiorev.2024.105637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2023] [Revised: 03/03/2024] [Accepted: 03/19/2024] [Indexed: 03/24/2024]
Abstract
Tics are sudden, repetitive movements or vocalizations. Tic disorders, such as Tourette syndrome (TS), are contributed by the interplay of genetic risk factors and environmental variables, leading to abnormalities in the functioning of the cortico-striatal-thalamo-cortical (CSTC) circuitry. Various neurotransmitter systems, such as gamma-aminobutyric acid (GABA) and dopamine, are implicated in the pathophysiology of these disorders. Building on the evidence that tic disorders are predominant in males and exacerbated by stress, emerging research is focusing on the involvement of neuroactive steroids, including dehydroepiandrosterone sulfate (DHEAS) and allopregnanolone, in the ontogeny of tics and other phenotypes associated with TS. Emerging evidence indicates that DHEAS levels are significantly elevated in the plasma of TS-affected boys, and the clinical onset of this disorder coincides with the period of adrenarche, the developmental stage characterized by a surge in DHEAS synthesis. On the other hand, allopregnanolone has garnered particular attention for its potential to mediate the adverse effects of acute stress on the exacerbation of tic severity and frequency. Notably, both neurosteroids act as key modulators of GABA-A receptors, suggesting a pivotal role of these targets in the pathophysiology of various clinical manifestations of tic disorders. This review explores the potential mechanisms by which these and other neuroactive steroids may influence tic disorders and discusses the emerging therapeutic strategies that target neuroactive steroids for the management of tic disorders.
Collapse
Affiliation(s)
- Caterina Branca
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Utah, Salt Lake City, UT, USA
| | - Marco Bortolato
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Utah, Salt Lake City, UT, USA.
| |
Collapse
|
5
|
Balan I, Boero G, Chéry SL, McFarland MH, Lopez AG, Morrow AL. Neuroactive Steroids, Toll-like Receptors, and Neuroimmune Regulation: Insights into Their Impact on Neuropsychiatric Disorders. Life (Basel) 2024; 14:582. [PMID: 38792602 PMCID: PMC11122352 DOI: 10.3390/life14050582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 04/18/2024] [Accepted: 04/28/2024] [Indexed: 05/26/2024] Open
Abstract
Pregnane neuroactive steroids, notably allopregnanolone and pregnenolone, exhibit efficacy in mitigating inflammatory signals triggered by toll-like receptor (TLR) activation, thus attenuating the production of inflammatory factors. Clinical studies highlight their therapeutic potential, particularly in conditions like postpartum depression (PPD), where the FDA-approved compound brexanolone, an intravenous formulation of allopregnanolone, effectively suppresses TLR-mediated inflammatory pathways, predicting symptom improvement. Additionally, pregnane neurosteroids exhibit trophic and anti-inflammatory properties, stimulating the production of vital trophic proteins and anti-inflammatory factors. Androstane neuroactive steroids, including estrogens and androgens, along with dehydroepiandrosterone (DHEA), display diverse effects on TLR expression and activation. Notably, androstenediol (ADIOL), an androstane neurosteroid, emerges as a potent anti-inflammatory agent, promising for therapeutic interventions. The dysregulation of immune responses via TLR signaling alongside reduced levels of endogenous neurosteroids significantly contributes to symptom severity across various neuropsychiatric disorders. Neuroactive steroids, such as allopregnanolone, demonstrate efficacy in alleviating symptoms of various neuropsychiatric disorders and modulating neuroimmune responses, offering potential intervention avenues. This review emphasizes the significant therapeutic potential of neuroactive steroids in modulating TLR signaling pathways, particularly in addressing inflammatory processes associated with neuropsychiatric disorders. It advances our understanding of the complex interplay between neuroactive steroids and immune responses, paving the way for personalized treatment strategies tailored to individual needs and providing insights for future research aimed at unraveling the intricacies of neuropsychiatric disorders.
Collapse
Affiliation(s)
- Irina Balan
- Bowles Center for Alcohol Studies, School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (I.B.); (S.L.C.); (M.H.M.); (A.G.L.)
- Department of Psychiatry, School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Giorgia Boero
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, NC 27710, USA;
| | - Samantha Lucenell Chéry
- Bowles Center for Alcohol Studies, School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (I.B.); (S.L.C.); (M.H.M.); (A.G.L.)
- Neuroscience Curriculum, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Minna H. McFarland
- Bowles Center for Alcohol Studies, School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (I.B.); (S.L.C.); (M.H.M.); (A.G.L.)
- Neuroscience Curriculum, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Alejandro G. Lopez
- Bowles Center for Alcohol Studies, School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (I.B.); (S.L.C.); (M.H.M.); (A.G.L.)
- Department of Biochemistry and Biophysics, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - A. Leslie Morrow
- Bowles Center for Alcohol Studies, School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (I.B.); (S.L.C.); (M.H.M.); (A.G.L.)
- Department of Psychiatry, School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Pharmacology, School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| |
Collapse
|
6
|
Yamaguchi J, Andrade MA, Truong TT, Toney GM. Glutamate Spillover Dynamically Strengthens Gabaergic Synaptic Inhibition of the Hypothalamic Paraventricular Nucleus. J Neurosci 2024; 44:e1851222023. [PMID: 38154957 PMCID: PMC10869154 DOI: 10.1523/jneurosci.1851-22.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 12/16/2023] [Accepted: 12/20/2023] [Indexed: 12/30/2023] Open
Abstract
The hypothalamic paraventricular nucleus (PVN) is strongly inhibited by γ-aminobutyric acid (GABA) from the surrounding peri-nuclear zone (PNZ). Because glutamate mediates fast excitatory transmission and is substrate for GABA synthesis, we tested its capacity to dynamically strengthen GABA inhibition. In PVN slices from male mice, bath glutamate applied during ionotropic glutamate receptor blockade increased PNZ-evoked inhibitory postsynaptic currents (eIPSCs) without affecting GABA-A receptor agonist currents or single-channel conductance, implicating a presynaptic mechanism(s). Consistent with this interpretation, bath glutamate failed to strengthen IPSCs during pharmacological saturation of GABA-A receptors. Presynaptic analyses revealed that glutamate did not affect paired-pulse ratio, peak eIPSC variability, GABA vesicle recycling speed, or readily releasable pool (RRP) size. Notably, glutamate-GABA strengthening (GGS) was unaffected by metabotropic glutamate receptor blockade and graded external Ca2+ when normalized to baseline amplitude. GGS was prevented by pan- but not glial-specific inhibition of glutamate uptake and by inhibition of glutamic acid decarboxylase (GAD), indicating reliance on glutamate uptake by neuronal excitatory amino acid transporter 3 (EAAT3) and enzymatic conversion of glutamate to GABA. EAAT3 immunoreactivity was strongly localized to presumptive PVN GABA terminals. High bath K+ also induced GGS, which was prevented by glutamate vesicle depletion, indicating that synaptic glutamate release strengthens PVN GABA inhibition. GGS suppressed PVN cell firing, indicating its functional significance. In sum, PVN GGS buffers neuronal excitation by apparent "over-filling" of vesicles with GABA synthesized from synaptically released glutamate. We posit that GGS protects against sustained PVN excitation and excitotoxicity while potentially aiding stress adaptation and habituation.
Collapse
Affiliation(s)
- Junya Yamaguchi
- Department of Cellular & Integrative Physiology, University of Texas Health San Antonio, San Antonio 78229-3900, Texas
| | - Mary Ann Andrade
- Department of Cellular & Integrative Physiology, University of Texas Health San Antonio, San Antonio 78229-3900, Texas
| | - Tamara T Truong
- Department of Cellular & Integrative Physiology, University of Texas Health San Antonio, San Antonio 78229-3900, Texas
| | - Glenn M Toney
- Department of Cellular & Integrative Physiology, University of Texas Health San Antonio, San Antonio 78229-3900, Texas
- Center for Biomedical Neuroscience, University of Texas Health San Antonio, San Antonio 78229-3900, Texas
| |
Collapse
|
7
|
Sato T, Sugaya T, Talukder AH, Tsushima Y, Sasaki S, Uchida K, Sato T, Ikoma Y, Sakimura K, Fukuda A, Matsui K, Itoi K. Dual action of serotonin on local excitatory and inhibitory neural circuits regulating the corticotropin-releasing factor neurons in the paraventricular nucleus of the hypothalamus. J Neuroendocrinol 2023; 35:e13351. [PMID: 37901949 DOI: 10.1111/jne.13351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 10/06/2023] [Accepted: 10/10/2023] [Indexed: 10/31/2023]
Abstract
Serotonergic neurons originating from the raphe nuclei have been proposed to regulate corticotropin-releasing factor (CRF) neurons in the paraventricular nucleus of the hypothalamus (PVH). Since glutamate- and γ-aminobutyric acid (GABA)-containing neurons, constituting the hypothalamic local circuits, innervate PVH CRF neurons, we examined whether they mediate the actions of serotonin (5-hydroxytryptamine [5-HT]) on CRF neurons. Spontaneous excitatory postsynaptic currents (sEPSCs) or spontaneous inhibitory postsynaptic currents (sIPSCs) were recorded in PVH CRF neurons, under whole cell patch-clamp, using the CRF-modified yellow fluorescent protein (Venus) ΔNeo mouse. Serotonin elicited an increase in the frequency of sEPSCs in 77% of the cells and a decrease in the frequency of sIPSCs in 71% of the cells, tested in normal medium. Neither the amplitude nor decay time of sEPSC and sIPSC was affected, thus the site(s) of action of serotonin may be presynaptic. In the presence of tetrodotoxin (TTX), serotonin had no significant effects on either parameter of sEPSC or sIPSC, indicating that the effects of serotonin are action potential-dependent, and that the presynaptic interneurons are largely intact within the slice; distant neurons may exist, though, since some 20%-30% of neurons did not respond to serotonin without TTX. We next examined through what receptor subtype(s) serotonin exerts its effects on presynaptic interneurons. DOI (5-HT2A/2C agonist) mimicked the action of serotonin on the sIPSCs, and the serotonin-induced decrease in sIPSC frequency was inhibited by a selective 5-HT2C antagonist RS102221. 8-OH-DPAT (5-HT1A/7 agonist) mimicked the action of serotonin on the sEPSCs, and the serotonin-induced increase in sEPSC frequency was inhibited by a selective 5-HT7 antagonist SB269970. Thus, serotonin showed a dual action on PVH CRF neurons, by upregulating glutamatergic- and downregulating GABAergic interneurons; the former may partly be mediated by 5-HT7 receptors, whereas the latter by 5-HT2C receptors. The CRF-Venus ΔNeo mouse was useful for the electrophysiological examination.
Collapse
Affiliation(s)
- Takayuki Sato
- Laboratory of Information Biology, Graduate School of Information Sciences, Tohoku University, Sendai, Japan
| | - Takuma Sugaya
- Laboratory of Information Biology, Graduate School of Information Sciences, Tohoku University, Sendai, Japan
| | - Ashraf Hossain Talukder
- Laboratory of Information Biology, Graduate School of Information Sciences, Tohoku University, Sendai, Japan
| | - Yuki Tsushima
- Laboratory of Information Biology, Graduate School of Information Sciences, Tohoku University, Sendai, Japan
| | - Shotaro Sasaki
- Laboratory of Information Biology, Graduate School of Information Sciences, Tohoku University, Sendai, Japan
| | - Katsuya Uchida
- Laboratory of Information Biology, Graduate School of Information Sciences, Tohoku University, Sendai, Japan
| | - Tatsuya Sato
- Laboratory of Information Biology, Graduate School of Information Sciences, Tohoku University, Sendai, Japan
| | - Yoko Ikoma
- Super-Network Brain Physiology, Graduate School of Life Sciences, Tohoku University, Sendai, Japan
| | - Kenji Sakimura
- Department of Animal Model Development, Brain Research Institute, Niigata University, Niigata, Japan
| | - Atsuo Fukuda
- Department of Physiology, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Ko Matsui
- Super-Network Brain Physiology, Graduate School of Life Sciences, Tohoku University, Sendai, Japan
| | - Keiichi Itoi
- Laboratory of Information Biology, Graduate School of Information Sciences, Tohoku University, Sendai, Japan
- Super-Network Brain Physiology, Graduate School of Life Sciences, Tohoku University, Sendai, Japan
- Department of Neuroendocrinology, Graduate School of Medicine, Tohoku University, Sendai, Japan
- Department of Nursing, Tohoku Fukushi University, Sendai, Japan
| |
Collapse
|
8
|
Romanov RA, Harkany T. Grabbing neuropeptide signals in the brain. Science 2023; 382:764-765. [PMID: 37972194 DOI: 10.1126/science.adl1788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2023]
Abstract
Bioengineered sensors resolve the dynamics of neuropeptide action.
Collapse
Affiliation(s)
- Roman A Romanov
- Department of Molecular Neurosciences, Center for Brain Research, Medical University of Vienna, Vienna, Austria
| | - Tibor Harkany
- Department of Molecular Neurosciences, Center for Brain Research, Medical University of Vienna, Vienna, Austria
- Department of Neuroscience, Biomedicum 7D, Karolinska Institutet, Solna, Sweden
| |
Collapse
|
9
|
Valtcheva S, Issa HA, Bair-Marshall CJ, Martin KA, Jung K, Zhang Y, Kwon HB, Froemke RC. Neural circuitry for maternal oxytocin release induced by infant cries. Nature 2023; 621:788-795. [PMID: 37730989 PMCID: PMC10639004 DOI: 10.1038/s41586-023-06540-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Accepted: 08/15/2023] [Indexed: 09/22/2023]
Abstract
Oxytocin is a neuropeptide that is important for maternal physiology and childcare, including parturition and milk ejection during nursing1-6. Suckling triggers the release of oxytocin, but other sensory cues-specifically, infant cries-can increase the levels of oxytocin in new human mothers7, which indicates that cries can activate hypothalamic oxytocin neurons. Here we describe a neural circuit that routes auditory information about infant vocalizations to mouse oxytocin neurons. We performed in vivo electrophysiological recordings and photometry from identified oxytocin neurons in awake maternal mice that were presented with pup calls. We found that oxytocin neurons responded to pup vocalizations, but not to pure tones, through input from the posterior intralaminar thalamus, and that repetitive thalamic stimulation induced lasting disinhibition of oxytocin neurons. This circuit gates central oxytocin release and maternal behaviour in response to calls, providing a mechanism for the integration of sensory cues from the offspring in maternal endocrine networks to ensure modulation of brain state for efficient parenting.
Collapse
Affiliation(s)
- Silvana Valtcheva
- Skirball Institute for Biomolecular Medicine, New York University School of Medicine, New York, NY, USA.
- Neuroscience Institute, New York University School of Medicine, New York, NY, USA.
- Department of Otolaryngology, New York University School of Medicine, New York, NY, USA.
- Department of Neuroscience and Physiology, New York University School of Medicine, New York, NY, USA.
- Center for Neural Science, New York University, New York, NY, USA.
| | - Habon A Issa
- Skirball Institute for Biomolecular Medicine, New York University School of Medicine, New York, NY, USA
- Neuroscience Institute, New York University School of Medicine, New York, NY, USA
- Department of Otolaryngology, New York University School of Medicine, New York, NY, USA
- Department of Neuroscience and Physiology, New York University School of Medicine, New York, NY, USA
- Center for Neural Science, New York University, New York, NY, USA
| | - Chloe J Bair-Marshall
- Skirball Institute for Biomolecular Medicine, New York University School of Medicine, New York, NY, USA
- Neuroscience Institute, New York University School of Medicine, New York, NY, USA
- Department of Otolaryngology, New York University School of Medicine, New York, NY, USA
- Department of Neuroscience and Physiology, New York University School of Medicine, New York, NY, USA
- Center for Neural Science, New York University, New York, NY, USA
| | - Kathleen A Martin
- Skirball Institute for Biomolecular Medicine, New York University School of Medicine, New York, NY, USA
- Neuroscience Institute, New York University School of Medicine, New York, NY, USA
- Department of Otolaryngology, New York University School of Medicine, New York, NY, USA
- Department of Neuroscience and Physiology, New York University School of Medicine, New York, NY, USA
- Center for Neural Science, New York University, New York, NY, USA
| | - Kanghoon Jung
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Yiyao Zhang
- Neuroscience Institute, New York University School of Medicine, New York, NY, USA
| | - Hyung-Bae Kwon
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Robert C Froemke
- Skirball Institute for Biomolecular Medicine, New York University School of Medicine, New York, NY, USA.
- Neuroscience Institute, New York University School of Medicine, New York, NY, USA.
- Department of Otolaryngology, New York University School of Medicine, New York, NY, USA.
- Department of Neuroscience and Physiology, New York University School of Medicine, New York, NY, USA.
- Center for Neural Science, New York University, New York, NY, USA.
| |
Collapse
|
10
|
Brivio E, Kos A, Ulivi AF, Karamihalev S, Ressle A, Stoffel R, Hirsch D, Stelzer G, Schmidt MV, Lopez JP, Chen A. Sex shapes cell-type-specific transcriptional signatures of stress exposure in the mouse hypothalamus. Cell Rep 2023; 42:112874. [PMID: 37516966 DOI: 10.1016/j.celrep.2023.112874] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 05/19/2023] [Accepted: 07/11/2023] [Indexed: 08/01/2023] Open
Abstract
Stress-related psychiatric disorders and the stress system show prominent differences between males and females, as well as strongly divergent transcriptional changes. Despite several proposed mechanisms, we still lack the understanding of the molecular processes at play. Here, we explore the contribution of cell types to transcriptional sex dimorphism using single-cell RNA sequencing. We identify cell-type-specific signatures of acute restraint stress in the paraventricular nucleus of the hypothalamus, a central hub of the stress response, in male and female mice. Further, we show that a history of chronic mild stress alters these signatures in a sex-specific way, and we identify oligodendrocytes as a major target for these sex-specific effects. This dataset, which we provide as an online interactive app, offers the transcriptomes of thousands of individual cells as a molecular resource for an in-depth dissection of the interplay between cell types and sex on the mechanisms of the stress response.
Collapse
Affiliation(s)
- Elena Brivio
- Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, 80804 Munich, Germany; International Max Planck Research School for Translational Psychiatry (IMPRS-TP), 80804 Munich, Germany; Department of Molecular Neuroscience, Weizmann Institute of Science, Rehovot 76100, Israel; Department of Brain Sciences, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Aron Kos
- Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, 80804 Munich, Germany
| | | | - Stoyo Karamihalev
- Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, 80804 Munich, Germany; International Max Planck Research School for Translational Psychiatry (IMPRS-TP), 80804 Munich, Germany
| | - Andrea Ressle
- Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, 80804 Munich, Germany
| | - Rainer Stoffel
- Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, 80804 Munich, Germany
| | - Dana Hirsch
- Department of Veterinary Resources, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Gil Stelzer
- Bioinformatics Unit, Department of Life Sciences Core Facilities, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Mathias V Schmidt
- Research Group Neurobiology of Stress Resilience, Max Planck Institute of Psychiatry, 80804 Munich, Germany
| | - Juan Pablo Lopez
- Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, 80804 Munich, Germany; Department of Neuroscience, Karolinska Institutet, 171 77 Stockholm, Sweden.
| | - Alon Chen
- Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, 80804 Munich, Germany; Department of Molecular Neuroscience, Weizmann Institute of Science, Rehovot 76100, Israel; Department of Brain Sciences, Weizmann Institute of Science, Rehovot 76100, Israel.
| |
Collapse
|
11
|
Norberto S, Assalin HB, Guadagnini D, Tobar N, Boer PA, Kang MC, Saad MJA, Kim YB, Prada PO. CLK2 in GABAergic neurons is critical in regulating energy balance and anxiety-like behavior in a gender-specific fashion. Front Endocrinol (Lausanne) 2023; 14:1172835. [PMID: 37635967 PMCID: PMC10449579 DOI: 10.3389/fendo.2023.1172835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Accepted: 07/24/2023] [Indexed: 08/29/2023] Open
Abstract
Introduction Cdc2-like kinase (CLK2) is a member of CLK kinases expressed in hypothalamic neurons and is activated in response to refeeding, leptin, or insulin. Diet-induced obesity and leptin receptor-deficient db/db mice lack CLK2 signal in the hypothalamic neurons. The neurotransmiter gamma-aminobutyric acid (GABA) is among the most prevalent in the central nervous system (CNS), particularly in the hypothalamus. Given the abundance of GABA-expressing neurons and their potential influence on regulating energy and behavioral homeostasis, we aimed to explore whether the deletion of CLK2 in GABAergic neurons alters energy homeostasis and behavioral and cognitive functions in both genders of mice lacking CLK2 in Vgat-expressing neurons (Vgat-Cre; Clk2loxP/loxP) on chow diet. Methods We generated mice lacking Clk2 in Vgat-expressing neurons (Vgat-Cre; Clk2loxP/loxP) by mating Clk2loxP/loxP mice with Vgat-IRES-Cre transgenic mice and employed behavior, and physiological tests, and molecular approaches to investigate energy metabolism and behavior phenotype of both genders. Results and discussion We showed that deletion of CLK2 in GABAergic neurons increased adiposity and food intake in females. The mechanisms behind these effects were likely due, at least in part, to hypothalamic insulin resistance and upregulation of hypothalamic Npy and Agrp expression. Besides normal insulin and pyruvate sensitivity, Vgat-Cre; Clk2loxP/loxP females were glucose intolerant. Male Vgat-Cre; Clk2loxP/loxP mice showed an increased energy expenditure (EE). Risen EE may account for avoiding weight and fat mass gain in male Vgat-Cre; Clk2loxP/loxP mice. Vgat-Cre; Clk2loxP/loxP mice had no alteration in cognition or memory functions in both genders. Interestingly, deleting CLK2 in GABAergic neurons changed anxiety-like behavior only in females, not males. These findings suggest that CLK2 in GABAergic neurons is critical in regulating energy balance and anxiety-like behavior in a gender-specific fashion and could be a molecular therapeutic target for combating obesity associated with psychological disorders in females.
Collapse
Affiliation(s)
- Sónia Norberto
- Department of Internal Medicine, School of Medical Science, University of Campinas (UNICAMP), Campinas, SP, Brazil
| | - Heloisa Balan Assalin
- Department of Internal Medicine, School of Medical Science, University of Campinas (UNICAMP), Campinas, SP, Brazil
| | - Dioze Guadagnini
- Department of Internal Medicine, School of Medical Science, University of Campinas (UNICAMP), Campinas, SP, Brazil
| | - Natália Tobar
- Department of Radiology, University of Campinas (UNICAMP), Campinas, SP, Brazil
| | - Patrícia Aline Boer
- Department of Internal Medicine, Fetal Programming Laboratory, School of Medical Science, University of Campinas (UNICAMP), Campinas, SP, Brazil
| | - Min-Cheol Kang
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
- Research Group of Food Processing, Korea Food Research Instute, Jeollabuk-do, Wanju, Republic of Korea
| | - Mario Jose Abdalla Saad
- Department of Internal Medicine, School of Medical Science, University of Campinas (UNICAMP), Campinas, SP, Brazil
| | - Young-Bum Kim
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| | - Patricia Oliveira Prada
- Department of Internal Medicine, School of Medical Science, University of Campinas (UNICAMP), Campinas, SP, Brazil
- School of Applied Sciences, University of Campinas (UNICAMP), Limeira, SP, Brazil
- Max-Planck Institute for Metabolism Research, Köln, Germany
| |
Collapse
|
12
|
Ruggiero-Ruff RE, Villa PA, Hijleh SA, Avalos B, DiPatrizio NV, Haga-Yamanaka S, Coss D. Increased body weight in mice with fragile X messenger ribonucleoprotein 1 (Fmr1) gene mutation is associated with hypothalamic dysfunction. Sci Rep 2023; 13:12666. [PMID: 37542065 PMCID: PMC10403586 DOI: 10.1038/s41598-023-39643-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Accepted: 07/28/2023] [Indexed: 08/06/2023] Open
Abstract
Mutations in the Fragile X Messenger Ribonucleoprotein 1 (FMR1) gene are linked to Fragile X Syndrome, the most common monogenic cause of intellectual disability and autism. People affected with mutations in FMR1 have higher incidence of obesity, but the mechanisms are largely unknown. In the current study, we determined that male Fmr1 knockout mice (KO, Fmr1-/y), but not female Fmr1-/-, exhibit increased weight when compared to wild-type controls, similarly to humans with FMR1 mutations. No differences in food or water intake were found between groups; however, male Fmr1-/y display lower locomotor activity, especially during their active phase. Moreover, Fmr1-/y have olfactory dysfunction determined by buried food test, although they exhibit increased compulsive behavior, determined by marble burying test. Since olfactory brain regions communicate with hypothalamic regions that regulate food intake, including POMC neurons that also regulate locomotion, we examined POMC neuron innervation and numbers in Fmr1-/y mice. POMC neurons express Fmrp, and POMC neurons in Fmr1-/y have higher inhibitory GABAergic synaptic inputs. Consistent with increased inhibitory innervation, POMC neurons in the Fmr1-/y mice exhibit lower activity, based on cFOS expression. Notably, Fmr1-/y mice have fewer POMC neurons than controls, specifically in the rostral arcuate nucleus, which could contribute to decreased locomotion and increased body weight. These results suggest a role for Fmr1 in the regulation of POMC neuron function and the etiology of Fmr1-linked obesity.
Collapse
Affiliation(s)
- Rebecca E Ruggiero-Ruff
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, CA, 92521, USA
| | - Pedro A Villa
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, CA, 92521, USA
| | - Sarah Abu Hijleh
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, CA, 92521, USA
| | - Bryant Avalos
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, CA, 92521, USA
| | - Nicholas V DiPatrizio
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, CA, 92521, USA
| | - Sachiko Haga-Yamanaka
- Department of Molecular, Cell, and Systems Biology, College of Natural and Agricultural Sciences, University of California, Riverside, Riverside, USA
| | - Djurdjica Coss
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, CA, 92521, USA.
| |
Collapse
|
13
|
Mota CMD, Siler DA, Burchiel KJ, Madden CJ. Acute deep brain stimulation of the paraventricular nucleus of the hypothalamus increases brown adipose tissue thermogenesis in rats. Neurosci Lett 2023; 799:137130. [PMID: 36792026 PMCID: PMC10069326 DOI: 10.1016/j.neulet.2023.137130] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 02/10/2023] [Accepted: 02/11/2023] [Indexed: 02/15/2023]
Abstract
Brown adipose tissue (BAT) activity is controlled by the sympathetic nervous system. Activation of BAT has shown significant promise in preclinical studies to elicit weight loss. Since the hypothalamic paraventricular nucleus (PVN) contributes to the regulation of BAT thermogenic activity, we sought to determine the effects of electrical stimulation of the PVN as a model of deep brain stimulation (DBS) for increasing BAT sympathetic nerve activity (SNA). The rostral raphe pallidus area (rRPa) was also chosen as a target for DBS since it contains the sympathetic premotor neurons for BAT. Electrical stimulation (100 µA, 100 µs, 100 Hz, for 5 min at a 50 % duty cycle) of the PVN increased BAT SNA and BAT thermogenesis. These effects were prevented by a local nanoinjection of bicuculline, a GABAA receptor antagonist. We suggest that electrical stimulation of the PVN elicited local release of GABA, which inhibited BAT sympathoinhibitory neurons in PVN, thereby releasing a restraint on BAT SNA. Electrical stimulation of the rRPa inhibited BAT thermogenesis and this was prevented by a local nanoinjection of bicuculline, suggesting that local release of GABA suppressed BAT SNA. Electrical stimulation of the PVN activates BAT metabolism via a mechanism that may include activation of local GABAA receptors. These findings contribute to our understanding of the mechanisms underlying the effects of DBS in the regulation of fat metabolism and provide a foundation for further DBS studies targeting hypothalamic circuits regulating BAT thermogenesis as a therapy for obesity.
Collapse
Affiliation(s)
- Clarissa M D Mota
- Department of Neurological Surgery, Oregon Health and Science University, Portland, OR, United States
| | - Dominic A Siler
- Department of Neurological Surgery, Oregon Health and Science University, Portland, OR, United States
| | - Kim J Burchiel
- Department of Neurological Surgery, Oregon Health and Science University, Portland, OR, United States
| | - Christopher J Madden
- Department of Neurological Surgery, Oregon Health and Science University, Portland, OR, United States.
| |
Collapse
|
14
|
Rastegarmanesh A, Rostami B, Nasimi A, Hatam M. In the parvocellular part of paraventricular nucleus, glutamatergic and GABAergic neurons mediate cardiovascular responses to AngII. Synapse 2023; 77:e22259. [PMID: 36271777 DOI: 10.1002/syn.22259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 09/03/2022] [Accepted: 09/18/2022] [Indexed: 01/29/2023]
Abstract
Angiotensinergic, GABAergic, and glutamatergic neurons are present in the parvocellular region of the paraventricular nucleus (PVNp). It has been shown that microinjection of AngII into the PVNp increases arterial pressure (AP) and heart rate (HR). The presence of synapses between the angiotensinergic, GABAergic, and glutamatergic neurons has been shown in the PVNp. In this study, we investigated the possible interaction between these three systems of the PVNp for control of AP and HR. All drugs were bilaterally (100 nl/side) microinjected into the PVNp of urethane-anesthetized rats, and AP and HR were recorded continuously. Microinjection of AngII into the PVNp produced pressor and tachycardia responses. Pretreatment of PVNp with AP5 or CNQX, glutamatergic NMDA and AMPA receptors antagonists, attenuated the responses to AngII. Pretreatment of PVNp with bicuculline greatly attenuated the pressor and tachycardia responses to AngII. In conclusion, this study provides the first evidence that pressor and tachycardia responses to microinjection of AngII into the PVNp are partly mediated by both NMDA and non-NMDA receptors of glutamate. Activation of glutamatergic neurons by AngII stimulates the sympathoexcitatory neurons. We also showed that the responses to AngII were strongly mediated by GABAA receptors, probably through activation of GABAergic neurons, which in turn inhibit sympathoinhibitory neurons.
Collapse
Affiliation(s)
- Ali Rastegarmanesh
- Department of Physiology, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Bahar Rostami
- Department of Physiology, Shiraz University of Medical Sciences, Shiraz, Iran.,Nanomedicine and Nanobiology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Ali Nasimi
- Department of Physiology, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Masoumeh Hatam
- Department of Physiology, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
15
|
GABAergic and Glutamatergic Phenotypes of Neurons Expressing Calcium-Binding Proteins in the Preoptic Area of the Guinea Pig. Int J Mol Sci 2022; 23:ijms23147963. [PMID: 35887305 PMCID: PMC9320123 DOI: 10.3390/ijms23147963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 07/18/2022] [Accepted: 07/18/2022] [Indexed: 11/17/2022] Open
Abstract
The mammalian preoptic area (POA) has large populations of calbindin (CB), calretinin (CR) and parvalbumin (PV) neurons, but phenotypes of these cells are unknown. Therefore, the question is whether neurons expressing CB, CR, and/or PV are GABAergic or glutamatergic. Double-immunofluorescence staining followed by epifluorescence and confocal microscopy was used to determine the coexpression patterns of CB, CR and PV expressing neurons with vesicular GABA transporters (VGAT) as specific markers of GABAergic neurons and vesicular glutamate transporters (VGLUT 2) as specific markers of glutamatergic neurons. The guinea pig was adopted as, like humans, it has a reproductive cycle with a true luteal phase and a long gestation period. The results demonstrated that in the guinea pig POA of both sexes, ~80% of CB+ and ~90% of CR+ neurons coexpress VGAT; however, one-fifth of CB+ neurons and one-third of CR+ cells coexpress VGLUT. About two-thirds of PV+ neurons express VGAT, and similar proportion of them coexpress VGLUT. Thus, many CB+, CR+ and PV+ neurons may be exclusively GABAergic (VGAT-expressing cells) or glutamatergic (VGLUT-expressing cells); however, at least a small fraction of CR+ cells and at least one-third of PV+ cells are likely neurons with a dual GABA/glutamate phenotype that may coexpress both transporters.
Collapse
|
16
|
Investigating the Role of GABA in Neural Development and Disease Using Mice Lacking GAD67 or VGAT Genes. Int J Mol Sci 2022; 23:ijms23147965. [PMID: 35887307 PMCID: PMC9318753 DOI: 10.3390/ijms23147965] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 07/12/2022] [Accepted: 07/16/2022] [Indexed: 11/18/2022] Open
Abstract
Normal development and function of the central nervous system involves a balance between excitatory and inhibitory neurotransmission. Activity of both excitatory and inhibitory neurons is modulated by inhibitory signalling of the GABAergic and glycinergic systems. Mechanisms that regulate formation, maturation, refinement, and maintenance of inhibitory synapses are established in early life. Deviations from ideal excitatory and inhibitory balance, such as down-regulated inhibition, are linked with many neurological diseases, including epilepsy, schizophrenia, anxiety, and autism spectrum disorders. In the mammalian forebrain, GABA is the primary inhibitory neurotransmitter, binding to GABA receptors, opening chloride channels and hyperpolarizing the cell. We review the involvement of down-regulated inhibitory signalling in neurological disorders, possible mechanisms for disease progression, and targets for therapeutic intervention. We conclude that transgenic models of disrupted inhibitory signalling—in GAD67+/− and VGAT−/− mice—are useful for investigating the effects of down-regulated inhibitory signalling in a range of neurological diseases.
Collapse
|
17
|
Wang H, Cheng L, Han Y. Effect of oral administration of GABA on thermoregulation in athletes during exercise in cold environments: A preliminary study. Front Nutr 2022; 9:883571. [PMID: 35911099 PMCID: PMC9335056 DOI: 10.3389/fnut.2022.883571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 06/27/2022] [Indexed: 11/21/2022] Open
Abstract
Background γ-aminobutyric acid (GABA), a common ingredient in sports supplements and other health products, regulates body temperature in the preoptic area and anterior hypothalamus (PO/AH). To date, no study has examined the effect of GABA on thermoregulation during exercise in humans in a cold temperature environment (11 ± 0.3°C, 45% ± 2% relative humidity). Methods We performed a randomized, double-blind study. Ten trained male athletes consumed either a drink (3 ml/kg weight) containing GABA (1,000 mg, trial G) or an equivalent amount of placebo drink (trial C) before exercise. They rested for 20 min and then cycled at 60% of maximum output power for 40 min, pedaling at 60 rpm, and recovered for 20 min. Core temperature (Tc), skin temperature (upper arm, chest, thigh, calf), and heart rate (HR) were monitored at rest (T0), exercise begins (T20), 20 min of exercise (T40), the exercise ends (T60), and at recovery (T80). Results Compared to T0, Tc decreased significantly at T20 and increased significantly at T40, T60 and T80 (p < 0.01). From 35–80 min, the Tc was higher in trial G (peaked at 37.96 ± 0.25°C) than in trial C (37.89 ± 0.37°C), but it failed to reach significant difference (p > 0.05); Tsk continued to increase during exercise and was significantly higher than T0 at T40 (p < 0.05), T60 and T80 (p < 0.01). There was no significant difference in Tsk between the two trials (p > 0.05). Conclusion Our findings provide initial evidence that oral administration of GABA does not affect thermoregulation and has no adverse effects on the body as an ergogenic exercise supplement during exercise in cold environments.
Collapse
|
18
|
Klusmann H, Schulze L, Engel S, Bücklein E, Daehn D, Lozza-Fiacco S, Geiling A, Meyer C, Andersen E, Knaevelsrud C, Schumacher S. HPA axis activity across the menstrual cycle - a systematic review and meta-analysis of longitudinal studies. Front Neuroendocrinol 2022; 66:100998. [PMID: 35597328 DOI: 10.1016/j.yfrne.2022.100998] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 04/09/2022] [Accepted: 05/07/2022] [Indexed: 12/29/2022]
Abstract
Differential HPA axis function has been proposed to underlie sex-differences in mental disorders; however, the impact of fluctuating sex hormones across the menstrual cycle on HPA axis activity is still unclear. This meta-analysis investigated basal cortisol concentrations as a marker for HPA axis activity across the menstrual cycle. Through a systematic literature search of five databases, 121 longitudinal studies were included, summarizing data of 2641 healthy, cycling participants between the ages of 18 and 45. The meta-analysis showed higher cortisol concentrations in the follicular vs. luteal phase (dSMC = 0.12, p =.004, [0.04 - 0.20]). Comparisons between more precise cycle phases were mostly insignificant, aside from higher concentrations in the menstrual vs. premenstrual phase (dSMC = 0.17, [0.02 - 0.33], p =.03). In all included studies, nine samples used established cortisol parameters to indicate HPA axis function, specifically diurnal profiles (k = 4) and the cortisol awakening response (CAR) (k = 5). Therefore, the meta-analysis highlights the need for more rigorous investigation of HPA axis activity and menstrual cycle phase.
Collapse
Affiliation(s)
- Hannah Klusmann
- Division of Clinical Psychological Intervention, Department of Education and Psychology, Freie Universität Berlin, Schwendenerstraße 27, 14195 Berlin, Germany.
| | - Lars Schulze
- Clinical Psychology and Psychotherapy, Department of Education and Psychology, Freie Universität Berlin, Habelschwerdter Allee 45, 14195 Berlin, Germany.
| | - Sinha Engel
- Division of Clinical Psychological Intervention, Department of Education and Psychology, Freie Universität Berlin, Schwendenerstraße 27, 14195 Berlin, Germany.
| | - Elise Bücklein
- Department of Clinical Psychology and Psychotherapy, Institute of Psychology and Education, Universität Ulm, Lise-Meitner-Str. 16, 89081 Ulm, Germany.
| | - Daria Daehn
- Clinical Psychology and Psychotherapy, Department of Education and Psychology, Freie Universität Berlin, Habelschwerdter Allee 45, 14195 Berlin, Germany.
| | - Serena Lozza-Fiacco
- School of Medicine, Department of Psychiatry, University of North Carolina at Chapel Hill, Carolina Crossings Building B, 2218 Nelson Highway, 27517 Chapel Hill, USA.
| | - Angelika Geiling
- Division of Clinical Psychological Intervention, Department of Education and Psychology, Freie Universität Berlin, Schwendenerstraße 27, 14195 Berlin, Germany.
| | - Caroline Meyer
- Division of Clinical Psychological Intervention, Department of Education and Psychology, Freie Universität Berlin, Schwendenerstraße 27, 14195 Berlin, Germany.
| | - Elizabeth Andersen
- School of Medicine, Department of Psychiatry, University of North Carolina at Chapel Hill, Carolina Crossings Building B, 2218 Nelson Highway, 27517 Chapel Hill, USA.
| | - Christine Knaevelsrud
- Division of Clinical Psychological Intervention, Department of Education and Psychology, Freie Universität Berlin, Schwendenerstraße 27, 14195 Berlin, Germany.
| | - Sarah Schumacher
- Division of Clinical Psychological Intervention, Department of Education and Psychology, Freie Universität Berlin, Schwendenerstraße 27, 14195 Berlin, Germany; Clinical Psychology and Psychotherapy, Department of Psychology, Faculty of Health, HMU Health and Medical University, Olympischer Weg 1, 14471 Potsdam, Germany.
| |
Collapse
|
19
|
Lee R, McGee A, Fernandez FX. Systematic review of drugs that modify the circadian system's phase-shifting responses to light exposure. Neuropsychopharmacology 2022; 47:866-879. [PMID: 34961774 PMCID: PMC8882192 DOI: 10.1038/s41386-021-01251-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Revised: 11/08/2021] [Accepted: 11/30/2021] [Indexed: 11/09/2022]
Abstract
We searched PubMed for primary research quantifying drug modification of light-induced circadian phase-shifting in rodents. This search, conducted for work published between 1960 and 2018, yielded a total of 146 papers reporting results from 901 studies. Relevant articles were those with any extractable data on phase resetting in wildtype (non-trait selected) rodents administered a drug, alongside a vehicle/control group, near or at the time of exposure. Most circadian pharmacology experiments were done using drugs thought to act directly on either the brain's central pacemaker, the suprachiasmatic nucleus (SCN), the SCN's primary relay, the retinohypothalamic tract, secondary pathways originating from the medial/dorsal raphe nuclei and intergeniculate leaflet, or the brain's sleep-arousal centers. While the neurotransmitter systems underlying these circuits were of particular interest, including those involving glutamate, gamma-aminobutyric acid, serotonin, and acetylcholine, other signaling modalities have also been assessed, including agonists and antagonists of receptors linked to dopamine, histamine, endocannabinoids, adenosine, opioids, and second-messenger pathways downstream of glutamate receptor activation. In an effort to identify drugs that unduly influence circadian responses to light, we quantified the net effects of each drug class by ratioing the size of the phase-shift observed after administration to that observed with vehicle in a given experiment. This allowed us to organize data across the literature, compare the relative efficacy of one mechanism versus another, and clarify which drugs might best suppress or potentiate phase resetting. Aggregation of the available data in this manner suggested that several candidates might be clinically relevant as auxiliary treatments to suppress ectopic light responses during shiftwork or amplify the circadian effects of timed bright light therapy. Future empirical research will be necessary to validate these possibilities.
Collapse
Affiliation(s)
- Robert Lee
- Department of Psychology, University of Arizona, Tucson, AZ, USA
| | - Austin McGee
- Department of Psychology, University of Arizona, Tucson, AZ, USA
| | - Fabian-Xosé Fernandez
- Department of Psychology, University of Arizona, Tucson, AZ, USA.
- Department of Neurology, University of Arizona, Tucson, AZ, USA.
- BIO5 and McKnight Brain Research Institutes, Tucson, AZ, USA.
| |
Collapse
|
20
|
Andersen ME, Guerrero T. Assessing Modes of Action, Measures of Tissue Dose and Human Relevance of Rodent Toxicity Endpoints with Octamethylcyclotetrasiloxane (D4). Toxicol Lett 2022; 357:57-72. [PMID: 34995712 DOI: 10.1016/j.toxlet.2021.12.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 12/07/2021] [Accepted: 12/30/2021] [Indexed: 10/19/2022]
Abstract
Octamethylcyclotetrasiloxane (D4), a highly lipophilic, volatile compound with low water solubility, is metabolized to lower molecular weight, linear silanols. Toxicity has been documented in several tissues in animals following mixed vapor/aerosol exposures by inhalation at near saturating vapor concentrations or with gavage dosing in vegetable oil vehicles. These results, together with more mechanism-based studies and detailed pharmacokinetic information, were used to assess likely modes of action (MOAs) and the tissue dose measures of D4 and metabolites that would serve as key events leading to these biological responses. This MOA analysis indicates that pulmonary effects arise from direct epithelial contact with mixed vapor/aerosol atmospheres of D4; liver hypertrophy and hepatocyte proliferation arise from adaptive, rodent-specific actions of D4 with nuclear receptor signaling pathways; and, nephropathy results from silanol metabolites binding with alpha-2μ globulin (a rat specific protein). At this time, the MOAs of other liver effects - pigment accumulation and bile duct hyperplasia (BDH) preferentially observed in Sprague-Dawley (SD) rats- are not known. Hypothalamic actions of D4 delaying the rat mid-cycle gonadotrophin releasing hormone (GnRH) surge that result in reproductive effects and subsequent vaginal/uterine/ovarian tissue responses, including small increases in incidence of benign endometrial adenomas, are associated with prolongation of endogenous estrogen exposures due to delays in ovulation. Human reproduction is not controlled by a mid-cycle GnRH surge. Since the rodent-specific reproductive and the vaginal/uterine/ovarian tissue responses are not relevant for risk assessments in human populations, D4 should neither be classified as a CMR (i.e., carcinogenic, mutagenic, or toxic for reproduction) substance nor be regarded as an endocrine disruptor. Bile duct hyperplasia (BDH) and pigment accumulation in liver seen in SD rats are endpoints that could serve to define a Benchmark Dose or No-Observed-Effect-Level (NOEL) for D4.
Collapse
Affiliation(s)
- Melvin E Andersen
- Andersen ToxConsulting LLC, 424 Granite Lake Ct., Denver, NC 28037, United States.
| | - Tracy Guerrero
- American Chemistry Council Director, Silicones, Environmental, Health, and Safety Center, 700 2nd Street, NE, Washington, DC, 20002, United States.
| |
Collapse
|
21
|
Coupling of GABA Metabolism to Mitochondrial Glucose Phosphorylation. Neurochem Res 2021; 47:470-480. [PMID: 34623563 DOI: 10.1007/s11064-021-03463-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Revised: 09/15/2021] [Accepted: 10/04/2021] [Indexed: 10/20/2022]
Abstract
Glucose and oxygen (O2) are vital to the brain. Glucose metabolism and mitochondria play a pivotal role in this process, culminating in the increase of reactive O2 species. Hexokinase (HK) is a key enzyme on glucose metabolism and is coupled to the brain mitochondrial redox modulation by recycling ADP for oxidative phosphorylation (OXPHOS). GABA shunt is an alternative pathway to GABA metabolism that increases succinate levels, a Krebs cycle intermediate. Although glucose and GABA metabolisms are intrinsically connected, their interplay coordinating mitochondrial function is poorly understood. Here, we hypothesize that the HK and the GABA shunt interact to control mitochondrial metabolism differently in the cortex and the hypothalamus. The GABA shunt stimulated mitochondrial O2 consumption and H2O2 production higher in hypothalamic synaptosomes (HSy) than cortical synaptosomes (CSy). The GABA shunt increased the HK coupled to OXPHOS activity in both population of synaptosomes, but the rate of activation was higher in HSy than CSy. Significantly, malonate and vigabatrin blocked the effects of the GABA shunt in the HK activity coupled to OXPHOS. It indicates that the glucose phosphorylation is linked to GABA and Krebs cycle reactions. Together, these data shed light on the HK and SDH role on the metabolism of each region fed by GABA turnover, which depends on the neurons' metabolic route.
Collapse
|
22
|
Thorsdottir D, Einwag Z, Erdos B. BDNF shifts excitatory-inhibitory balance in the paraventricular nucleus of the hypothalamus to elevate blood pressure. J Neurophysiol 2021; 126:1209-1220. [PMID: 34406887 DOI: 10.1152/jn.00247.2021] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Presympathetic neurons in the paraventricular nucleus of the hypothalamus (PVN) play a key role in cardiovascular regulation. We have previously shown that brain-derived neurotrophic factor (BDNF), acting in the PVN, increases sympathetic activity and blood pressure and serves as a key regulator of stress-induced hypertensive responses. BDNF is known to alter glutamatergic and GABA-ergic signaling broadly in the central nervous system, but whether BDNF has similar actions in the PVN remains to be investigated. Here, we tested the hypothesis that increased BDNF expression in the PVN elevates blood pressure by enhancing N-methyl-d-aspartate (NMDA) receptor (NMDAR)- and inhibiting GABAA receptor (GABAAR)-mediated signaling. Sprague-Dawley rats received bilateral PVN injections of AAV2 viral vectors expressing green fluorescent protein (GFP) or BDNF. Three weeks later, cardiovascular responses to PVN injections of NMDAR and GABAAR agonists and antagonists were recorded under α-chloralose-urethane anesthesia. In addition, expressions of excitatory and inhibitory signaling components in the PVN were assessed using immunofluorescence. Our results showed that NMDAR inhibition led to a greater decrease in blood pressure in the BDNF vs. GFP group, while GABAAR inhibition led to greater increases in blood pressure in the GFP group compared to BDNF. Conversely, GABAAR activation decreased blood pressure significantly more in GFP vs. BDNF rats. In addition, immunoreactivity of NMDAR1 was upregulated, while GABAAR-α1 and K+/Cl- cotransporter 2 were downregulated by BDNF overexpression in the PVN. In summary, our findings indicate that hypertensive actions of BDNF within the PVN are mediated, at least in part, by augmented NMDAR and reduced GABAAR signaling.NEW & NOTEWORTHY We have shown that BDNF, acting in the PVN, elevates blood pressure in part by augmenting NMDA receptor-mediated excitatory input and by diminishing GABAA receptor-mediated inhibitory input to PVN neurons. In addition, we demonstrate that elevated BDNF expression in the PVN upregulates NMDA receptor immunoreactivity and downregulates GABAA receptor as well as KCC2 transporter immunoreactivity.
Collapse
Affiliation(s)
| | - Zachary Einwag
- Department of Pharmacology, University of Vermont, Burlington, Vermont
| | - Benedek Erdos
- Department of Pharmacology, University of Vermont, Burlington, Vermont
| |
Collapse
|
23
|
Stradtman SC, Freeman JL. Mechanisms of Neurotoxicity Associated with Exposure to the Herbicide Atrazine. TOXICS 2021; 9:207. [PMID: 34564358 PMCID: PMC8473009 DOI: 10.3390/toxics9090207] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 08/20/2021] [Accepted: 08/29/2021] [Indexed: 12/19/2022]
Abstract
Atrazine is an herbicide commonly used on crops to prevent broadleaf weeds. Atrazine is an endocrine-disrupting chemical mainly targeting the neuroendocrine system and associated axes, especially as a reproductive toxicant through attenuation of the luteinizing hormone (LH). Current regulatory levels for chronic exposure are based on no observed adverse effect levels (NOAELs) of these LH alterations in rodent studies. Atrazine has also been studied for its effects on the central nervous system and neurotransmission. The European Union (EU) recognized the health risks of atrazine exposure as a public health concern with no way to contain contamination of drinking water. As such, the EU banned atrazine use in 2003. The United States recently reapproved atrazine's use in the fall of 2020. Research has shown that there is a wide array of adverse health effects that are seen across multiple models, exposure times, and exposure periods leading to dysfunction in many different systems in the body with most pointing to a neuroendocrine target of toxicity. There is evidence of crosstalk between systems that can be affected by atrazine exposure, causing widespread dysfunction and leading to changes in behavior even with no direct link to the hypothalamus. The hypothetical mechanism of toxicity of atrazine endocrine disruption and neurotoxicity can therefore be described as a web of pathways that are influenced through changes occurring in each and their multiple feedback loops with further research needed to refine NOAELs for neurotoxic outcomes.
Collapse
Affiliation(s)
| | - Jennifer L. Freeman
- School of Health Sciences, Purdue University, 550 Stadium Mall Drive, West Lafayette, IN 47907, USA;
| |
Collapse
|
24
|
Rijal S, Jang SH, Park SJ, Han SK. Lithium Enhances the GABAergic Synaptic Activities on the Hypothalamic Preoptic Area (hPOA) Neurons. Int J Mol Sci 2021; 22:3908. [PMID: 33918982 PMCID: PMC8069239 DOI: 10.3390/ijms22083908] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 04/06/2021] [Accepted: 04/08/2021] [Indexed: 12/12/2022] Open
Abstract
Lithium (Li+) salt is widely used as a therapeutic agent for treating neurological and psychiatric disorders. Despite its therapeutic effects on neurological and psychiatric disorders, it can also disturb the neuroendocrine axis in patients under lithium therapy. The hypothalamic area contains GABAergic and glutamatergic neurons and their receptors, which regulate various hypothalamic functions such as the release of neurohormones, control circadian activities. At the neuronal level, several neurotransmitter systems are modulated by lithium exposure. However, the effect of Li+ on hypothalamic neuron excitability and the precise action mechanism involved in such an effect have not been fully understood yet. Therefore, Li+ action on hypothalamic neurons was investigated using a whole-cell patch-clamp technique. In hypothalamic neurons, Li+ increased the GABAergic synaptic activities via action potential independent presynaptic mechanisms. Next, concentration-dependent replacement of Na+ by Li+ in artificial cerebrospinal fluid increased frequencies of GABAergic miniature inhibitory postsynaptic currents without altering their amplitudes. Li+ perfusion induced inward currents in the majority of hypothalamic neurons independent of amino-acids receptor activation. These results suggests that Li+ treatment can directly affect the hypothalamic region of the brain and regulate the release of various neurohormones involved in synchronizing the neuroendocrine axis.
Collapse
Affiliation(s)
| | | | | | - Seong Kyu Han
- Department of Oral Physiology, School of Dentistry & Institute of Oral Bioscience, Jeonbuk National University, Jeonju 54896, Korea; (S.R.); (S.H.J.); (S.J.P.)
| |
Collapse
|
25
|
Huang ST, Song ZJ, Liu Y, Luo WC, Yin Q, Zhang YM. BNST AV GABA-PVN CRF Circuit Regulates Visceral Hypersensitivity Induced by Maternal Separation in Vgat-Cre Mice. Front Pharmacol 2021; 12:615202. [PMID: 33815103 PMCID: PMC8017215 DOI: 10.3389/fphar.2021.615202] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Accepted: 02/04/2021] [Indexed: 12/12/2022] Open
Abstract
Visceral hypersensitivity as a common clinical manifestation of irritable bowel syndrome (IBS) may contribute to the development of chronic visceral pain. Our prior studies authenticated that the activation of the corticotropin-releasing factor (CRF) neurons in paraventricular nucleus (PVN) contributed to visceral hypersensitivity in mice, but puzzles still remain with respect to the underlying hyperactivation of corticotropin-releasing factor neurons. Herein, we employed maternal separation (MS) to establish mouse model of visceral hypersensitivity. The neuronal circuits associated with nociceptive hypersensitivity involved paraventricular nucleus CRF neurons by means of techniques such as behavioral test, pharmacology, molecular biology, retrograde neuronal circuit tracers, electrophysiology, chemogenetics and optogenetics. MS could predispose the elevated firing frequency of CRF neurons in PVN in murine adulthood, which could be annulled via the injection of exogenous GABA (0.3mM, 0.2µl) into PVN. The PVN-projecting GABAergic neurons were mainly distributed in the anterior ventral (AV) region in the bed nucleus of stria terminalis (BNST), wherein the excitability of these GABAergic neurons was reduced. Casp3 virus was utilized to induce apoptosis of GABA neurons in BNST-AV region, resulting in the activation of CRF neurons in PVN and visceral hyperalgesia. In parallel, chemogenetic and optogenetic approaches to activate GABAergic BNSTAV-PVN circuit in MS mice abated the spontaneous firing frequency of PVN CRF neurons and prevented the development of visceral hypersensitivity. A priori, PVNCRF-projecting GABAergic neurons in BNST-AV region participated in the occurrence of visceral hypersensitivity induced by MS. Our research may provide a new insight into the neural circuit mechanism of chronic visceral pain.
Collapse
Affiliation(s)
- Si-Ting Huang
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, China
| | - Zhi-Jing Song
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, China.,Department of Anesthesiology, Xuzhou Municipal Hospital Affiliated with Xuzhou Medical University, Xuzhou, China
| | - Yu Liu
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, China
| | - Wen-Chen Luo
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, China
| | - Qian Yin
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, China
| | - Yong-Mei Zhang
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, China
| |
Collapse
|
26
|
Dissociation of endocrine responses to the Trier Social Stress Test in Virtual Reality (VR-TSST) by the benzodiazepine alprazolam and the translocator protein 18 kDa (TSPO) ligand etifoxine. Psychoneuroendocrinology 2021; 124:105100. [PMID: 33338971 DOI: 10.1016/j.psyneuen.2020.105100] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Revised: 10/29/2020] [Accepted: 12/01/2020] [Indexed: 01/25/2023]
Abstract
BACKGROUND Activity of the two major stress systems, the hypothalamic-pituitary-adrenal (HPA) and the sympathetic-adrenal-medullary (SAM) axis, has already been shown to be modulated by different compounds that bind to the central benzodiazepine receptor. Less is known about ligands that modulate the peripheral benzodiazepine receptor - meanwhile known as the translocator protein 18 kDa (TSPO) - which constitute promising candidates in the search of novel anxiolytics. To close this gap, the present study compared the effects of the benzodiazepine alprazolam and the TSPO ligand etifoxine on responses of the HPA and SAM axes to the Trier Social Stress Test, a standardized paradigm to induce acute psychosocial stress in humans, performed in Virtual Reality (VR-TSST). METHODS Sixty healthy males, aged between 18 and 55 years, were randomly assigned to receive either a daily dose of 1.5 mg alprazolam, 150 mg etifoxine, or placebo over five days. On the last day of intake, they were exposed to the VR-TSST. We assessed changes of salivary cortisol, allopregnanolone, (nor-) epinephrine in serum, TSPO expression in platelets as well as heart rate (HR), skin conductance level (SCL) and self-reports in response to the stress task. Repeated measures ANOVAs were conducted to examine treatment effects on these stress response variables during the course of VR-TSST. RESULTS The response of salivary cortisol to the VR-TSST was significantly blunted in participants pre-treated with alprazolam but was not affected by etifoxine. While levels of allopregnanolone, epinephrine and norepinephrine increased in response to stress, TSPO expression decreased. None of those endocrine stress markers was affected by the active treatments, whereas TSPO expression increased after etifoxine administration over all study days. There were no effects of the two anxiolytics on HR, SCL or any self-report measurement. CONCLUSION The current study confirmed the attenuating effects of benzodiazepines on stress-induced HPA axis activity but did not reveal a comparable effect of the TSPO ligand etifoxine. The long-term consequences of a pharmacologically blunted response of the HPA axis to an acute stressor should be further elucidated. Due to the missing effects of etifoxine on stress-related parameters in our sample of healthy subjects, it might be concluded that the therapeutic effects of this TSPO ligand are restricted to stronger or pathological stress responses, respectively.
Collapse
|
27
|
Iovino M, Messana T, Tortora A, Giusti C, Lisco G, Giagulli VA, Guastamacchia E, De Pergola G, Triggiani V. Oxytocin Signaling Pathway: From Cell Biology to Clinical Implications. Endocr Metab Immune Disord Drug Targets 2021; 21:91-110. [PMID: 32433011 DOI: 10.2174/1871530320666200520093730] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Revised: 04/04/2020] [Accepted: 04/16/2020] [Indexed: 11/22/2022]
Abstract
BACKGROUND In addition to the well-known role played in lactation and parturition, Oxytocin (OT) and OT receptor (OTR) are involved in many other aspects such as the control of maternal and social behavior, the regulation of the growth of the neocortex, the maintenance of blood supply to the cortex, the stimulation of limbic olfactory area to mother-infant recognition bond, and the modulation of the autonomic nervous system via the vagal pathway. Moreover, OT and OTR show antiinflammatory, anti-oxidant, anti-pain, anti-diabetic, anti-dyslipidemic and anti-atherogenic effects. OBJECTIVE The aim of this narrative review is to summarize the main data coming from the literature dealing with the role of OT and OTR in physiology and pathologic conditions focusing on the most relevant aspects. METHODS Appropriate keywords and MeSH terms were identified and searched in Pubmed. Finally, references of original articles and reviews were examined. RESULTS We report the most significant and updated data on the role played by OT and OTR in physiology and different clinical contexts. CONCLUSION Emerging evidence indicates the involvement of OT system in several pathophysiological mechanisms influencing brain anatomy, cognition, language, sense of safety and trust and maternal behavior, with the possible use of exogenous administered OT in the treatment of specific neuropsychiatric conditions. Furthermore, it modulates pancreatic β-cell responsiveness and lipid metabolism leading to possible therapeutic use in diabetic and dyslipidemic patients and for limiting and even reversing atherosclerotic lesions.
Collapse
Affiliation(s)
- Michele Iovino
- Interdisciplinary Department of Medicine-Section of Internal Medicine, Geriatrics, Endocrinology and Rare Diseases. University of Bari "Aldo Moro", School of Medicine, Policlinico, Piazza Giulio Cesare 11, 70124, Bari, Italy
| | - Tullio Messana
- Infantile Neuropsychiatry, IRCCS - Institute of Neurological Sciences, Bologna, Italy
| | - Anna Tortora
- Interdisciplinary Department of Medicine-Section of Internal Medicine, Geriatrics, Endocrinology and Rare Diseases. University of Bari "Aldo Moro", School of Medicine, Policlinico, Piazza Giulio Cesare 11, 70124, Bari, Italy
| | - Consuelo Giusti
- Interdisciplinary Department of Medicine-Section of Internal Medicine, Geriatrics, Endocrinology and Rare Diseases. University of Bari "Aldo Moro", School of Medicine, Policlinico, Piazza Giulio Cesare 11, 70124, Bari, Italy
| | - Giuseppe Lisco
- Hospital Unit of Endocrinology, Perrino Hospital, Brindisi, Italy
| | - Vito Angelo Giagulli
- Interdisciplinary Department of Medicine-Section of Internal Medicine, Geriatrics, Endocrinology and Rare Diseases. University of Bari "Aldo Moro", School of Medicine, Policlinico, Piazza Giulio Cesare 11, 70124, Bari, Italy
| | - Edoardo Guastamacchia
- Interdisciplinary Department of Medicine-Section of Internal Medicine, Geriatrics, Endocrinology and Rare Diseases. University of Bari "Aldo Moro", School of Medicine, Policlinico, Piazza Giulio Cesare 11, 70124, Bari, Italy
| | - Giovanni De Pergola
- Clinical Nutrition Unit, Medical Oncology, Department of Internal Medicine and Clinical Oncology, University of Bari, School of Medicine, Policlinico, Piazza Giulio Cesare 11, 70124, Bari, Italy
| | - Vincenzo Triggiani
- Interdisciplinary Department of Medicine-Section of Internal Medicine, Geriatrics, Endocrinology and Rare Diseases. University of Bari "Aldo Moro", School of Medicine, Policlinico, Piazza Giulio Cesare 11, 70124, Bari, Italy
| |
Collapse
|
28
|
Abstract
Neuroanatomic and functional studies show the paraventricular (PVN) of the hypothalamus to have a central role in the autonomic control that supports cardiovascular regulation. Direct and indirect projections from the PVN preautonomic neurons to the sympathetic preganglionic neurons in the spinal cord modulate sympathetic activity. The preautonomic neurons of the PVN adjust their level of activation in response to afferent signals arising from peripheral viscerosensory receptors relayed through the nucleus tractus solitarius. The prevailing sympathetic tone is a balance between excitatory and inhibitory influences that arises from the preautonomic PVN neurons. Under physiologic conditions, tonic sympathetic inhibition driven by a nitric oxide-γ-aminobutyric acid-mediated mechanism is dominant, but in pathologic situation such as heart failure there is a switch from inhibition to sympathoexcitation driven by glutamate and angiotensin II. Angiotensin II, reactive oxygen species, and hypoxia as a result of myocardial infarction/ischemia alter the tightly regulated posttranslational protein-protein interaction of CAPON (carboxy-terminal postsynaptic density protein ligand of neuronal nitric oxide synthase (NOS1)) and PIN (protein inhibitor of NOS1) signaling mechanism. Within the preautonomic neurons of the PVN, the disruption of CAPON and PIN signaling leads to a downregulation of NOS1 expression and reduced NO bioavailability. These data support the notion that CAPON-PIN dysregulation of NO bioavailability is a major contributor to the pathogenesis of sympathoexcitation in heart failure.
Collapse
Affiliation(s)
- Susan Pyner
- Department of Biosciences, Durham University, Durham, United Kingdom.
| |
Collapse
|
29
|
Kim YB, Jung WW, Lee SW, Jin X, Kang HK, Hong EH, Min SS, Kim YS, Han HC, Colwell CS, Kim YI. Excessive maternal salt intake gives rise to vasopressin-dependent salt sensitivity of blood pressure in male offspring. J Mol Cell Cardiol 2021; 150:12-22. [PMID: 33011158 DOI: 10.1016/j.yjmcc.2020.09.013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 08/28/2020] [Accepted: 09/26/2020] [Indexed: 01/11/2023]
Abstract
Salt sensitivity of blood pressure (SSBP) is a trait carrying strong prognostic implications for various cardiovascular diseases. To test the hypothesis that excessive maternal salt intake causes SSBP in offspring through a mechanism dependent upon arginine-vasopressin (AVP), we performed a series of experiments using offspring of the rat dams salt-loaded during pregnancy and lactation with 1.5% saline drink ("experimental offspring") and those with normal perinatal salt exposure ("control offspring"). Salt challenge, given at 7-8 weeks of age with either 2% saline drink (3 days) or 8% NaCl-containing chow (4 weeks), had little or no effect on systolic blood pressure (SBP) in female offspring, whereas the salt challenge significantly raised SBP in male offspring, with the magnitude of increase being greater in experimental, than control, rats. Furthermore, the salt challenge not only raised plasma AVP level more and caused greater depressor responses to V1a and V2 AVP receptor antagonists to occur in experimental, than control, males, but it also made GABA excitatory in a significant proportion of magnocellular AVP neurons of experimental males by depolarizing GABA equilibrium potential. The effect of the maternal salt loading on the salt challenge-elicited SBP response in male offspring was precluded by maternal conivaptan treatment (non-selective AVP receptor antagonist) during the salt-loading period, whereas it was mimicked by neonatal AVP treatment. These results suggest that the excessive maternal salt intake brings about SSBP in male offspring, both the programming and the expression of which depend on increased AVP secretion that may partly result from excitatory GABAergic action.
Collapse
Affiliation(s)
- Young-Beom Kim
- Department of Physiology and Neuroscience Research Institute, Korea University College of Medicine, Seoul 02841, Republic of Korea
| | - Won Woo Jung
- Department of Physiology and Neuroscience Research Institute, Korea University College of Medicine, Seoul 02841, Republic of Korea
| | - Seung Won Lee
- Department of Physiology and Neuroscience Research Institute, Korea University College of Medicine, Seoul 02841, Republic of Korea
| | - Xiangyan Jin
- Department of Physiology and Neuroscience Research Institute, Korea University College of Medicine, Seoul 02841, Republic of Korea
| | - Hyung Kyung Kang
- Department of Physiology and Neuroscience Research Institute, Korea University College of Medicine, Seoul 02841, Republic of Korea
| | - Eun-Hwa Hong
- Department of Physiology and Neuroscience Research Institute, Korea University College of Medicine, Seoul 02841, Republic of Korea
| | - Sun Seek Min
- Department of Physiology and Biophysics, Eulji University School of Medicine, Daejeon 34824, Republic of Korea
| | - Yoon-Sik Kim
- Department of Physiology and Neuroscience Research Institute, Korea University College of Medicine, Seoul 02841, Republic of Korea
| | - Hee Chul Han
- Department of Physiology and Neuroscience Research Institute, Korea University College of Medicine, Seoul 02841, Republic of Korea
| | - Christopher S Colwell
- Department of Psychiatry and Biobehavioral Sciences, University of California School of Medicine, Los Angeles, CA, United States of America.
| | - Yang In Kim
- Department of Physiology and Neuroscience Research Institute, Korea University College of Medicine, Seoul 02841, Republic of Korea.
| |
Collapse
|
30
|
Si Y, Wang L, Lan J, Li H, Guo T, Chen X, Dong C, Ouyang Z, Chen SQ. Lilium davidii extract alleviates p‑chlorophenylalanine‑induced insomnia in rats through modification of the hypothalamic-related neurotransmitters, melatonin and homeostasis of the hypothalamic-pituitary-adrenal axis. PHARMACEUTICAL BIOLOGY 2020; 58:915-924. [PMID: 32924742 PMCID: PMC7534321 DOI: 10.1080/13880209.2020.1812674] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 07/30/2020] [Accepted: 08/15/2020] [Indexed: 06/11/2023]
Abstract
CONTEXT Lilium davidii var. unicolour Cotton (Lilium genus, Liliaceae) is an edible plant and a herb used in China to alleviate insomnia. OBJECTIVE To investigate the alleviating insomnia mechanism of L. davidii (LD). MATERIALS AND METHODS Wistar rats were intraperitoneally injected with p-chlorophenylalanine (PCPA) to establish an insomnia model. Rats were divided into six groups (n = 8): Control, PCPA, Estazolam (0.5 mg/kg), LD extract in low, medium and high doses (185.22, 370.44, 740.88 mg/kg). Serum hormone levels of the HPA axis, levels of 5-HT, NE and MT, and the expression of GABAA and 5-HT1A receptors in hypothalamus were determined. Moreover, behavioural and pathological changes in the hypothalamus were evaluated. RESULTS After LD administration, body weight and brain coefficient increased by 2.74% and 8.22%, respectively, and the adrenal coefficient decreased by 25%, compared with PCPA group. Elevation of the serum hypothalamic-pituitary-adrenal (HPA) axis hormone CRH (11.24 ± 3.16 ng/mL), ACTH (565.87 ± 103.44 pg/mL) and CORT (44.28 ± 8.73 ng/mL) in the PCPA group was reversed after LD treatment. Furthermore, abnormal excitatory behaviour [5 min movement distance (2096.34 ± 259.51 cm), central exercise time (5.28 ± 1.08 s)] of insomnia rats in the PCPA group was also relieved. LD extract increased 5-HT and MT levels, reduced NE level in the hypothalamus, and upregulated the expression of GABAA R and 5-HT1A. Moreover, LD extract may improve the pathology of neurons in the hypothalamus. CONCLUSIONS LD can be considered to develop health-care food or novel drugs to cope with the increasing number of insomniacs.
Collapse
Affiliation(s)
- Yanpo Si
- Department of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, China
| | - Lili Wang
- Department of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, China
| | - Jinxu Lan
- Department of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, China
| | - Hanwei Li
- Department of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, China
| | - Tao Guo
- Department of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, China
| | - Xiaohui Chen
- Academy of Chinese medicine, Henan University of Chinese Medicine, Zhengzhou, China
| | - Chunhong Dong
- Academy of Chinese medicine, Henan University of Chinese Medicine, Zhengzhou, China
| | - Zhen Ouyang
- School of Pharmacy, Jiangsu University, Zhenjiang, China
| | - Sui-qing Chen
- Department of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, China
| |
Collapse
|
31
|
Ammari R, Broberger C. Pre- and post-synaptic modulation by GABA B receptors of rat neuroendocrine dopamine neurones. J Neuroendocrinol 2020; 32:e12881. [PMID: 32803906 DOI: 10.1111/jne.12881] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 05/22/2020] [Accepted: 06/05/2020] [Indexed: 12/17/2022]
Abstract
The secretion of prolactin from the pituitary is negatively controlled by tuberoinfundibular dopamine (TIDA) neurones. The electrical properties of TIDA cells have recently been identified as a modulatory target of neurotransmitters and hormones in the lactotrophic axis. The role of the GABAB receptor in this control has received little attention, yet is of particular interest because it may act as a TIDA neurone autoreceptor. Here, this issue was explored in a spontaneously active rat TIDA in vitro slice preparation using whole-cell recordings. Application of the GABAB receptor agonist, baclofen, dose-dependently slowed down or abolished the network oscillations typical of this preparation. Pharmacological manipulations identify the underlying mechanism as an outward current mediated by G-protein-coupled inwardly rectifying K+ -like channels. In addition to this postsynaptic modulation, we describe a presynaptic modulation where GABAB receptors restrain the release of glutamate and GABA onto TIDA neurones. Our data identify both pre- and postsynaptic modulation of TIDA neurones by GABAB receptors that may play a role in the neuronal network control of pituitary prolactin secretion and lactation.
Collapse
Affiliation(s)
- Rachida Ammari
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Christian Broberger
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
- Department of Biochemistry and Biophysics, Stockholm University, Stockholm, Sweden
| |
Collapse
|
32
|
Herman JP, Nawreen N, Smail MA, Cotella EM. Brain mechanisms of HPA axis regulation: neurocircuitry and feedback in context Richard Kvetnansky lecture. Stress 2020; 23:617-632. [PMID: 33345670 PMCID: PMC8034599 DOI: 10.1080/10253890.2020.1859475] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Accepted: 11/28/2020] [Indexed: 12/11/2022] Open
Abstract
Regulation of stress reactivity is a fundamental priority of all organisms. Stress responses are critical for survival, yet can also cause physical and psychological damage. This review provides a synopsis of brain mechanisms designed to control physiological responses to stress, focusing primarily on glucocorticoid secretion via the hypothalamo-pituitary-adrenocortical (HPA) axis. The literature provides strong support for multi-faceted control of HPA axis responses, involving both direct and indirect actions at paraventricular nucleus (PVN) corticotropin releasing hormone neurons driving the secretory cascade. The PVN is directly excited by afferents from brainstem and hypothalamic circuits, likely relaying information on homeostatic challenge. Amygdala subnuclei drive HPA axis responses indirectly via disinhibition, mediated by GABAergic relays onto PVN-projecting neurons in the hypothalamus and bed nucleus of the stria terminalis (BST). Inhibition of stressor-evoked HPA axis responses is mediated by an elaborate network of glucocorticoid receptor (GR)-containing circuits, providing a distributed negative feedback signal that inhibits PVN neurons. Prefrontal and hippocampal neurons play a major role in HPA axis inhibition, again mediated by hypothalamic and BST GABAergic relays to the PVN. The complexity of the regulatory process suggests that information on stressors is integrated across functional disparate brain circuits prior to accessing the PVN, with regions such as the BST in prime position to relay contextual information provided by these sources into appropriate HPA activation. Dysregulation of the HPA in disease is likely a product of inappropriate checks and balances between excitatory and inhibitory inputs ultimately impacting PVN output.
Collapse
Affiliation(s)
- James P Herman
- Department of Pharmacology & Systems Physiology, University of Cincinnati, Cincinnati, OH, USA
- Department of Neurology and Rehabilitation Medicine, University of Cincinnati, Cincinnati, OH, USA
- Cincinnati Veterans Administration Medical Center, Cincinnati, OH, USA
| | - Nawshaba Nawreen
- Department of Pharmacology & Systems Physiology, University of Cincinnati, Cincinnati, OH, USA
| | - Marissa A Smail
- Department of Pharmacology & Systems Physiology, University of Cincinnati, Cincinnati, OH, USA
| | - Evelin M Cotella
- Department of Pharmacology & Systems Physiology, University of Cincinnati, Cincinnati, OH, USA
- Cincinnati Veterans Administration Medical Center, Cincinnati, OH, USA
| |
Collapse
|
33
|
Hieta JP, Kopra J, Räikkönen H, Kauppila TJ, Kostiainen R. Sub-100 μm Spatial Resolution Ambient Mass Spectrometry Imaging of Rodent Brain with Laser Ablation Atmospheric Pressure Photoionization (LAAPPI) and Laser Ablation Electrospray Ionization (LAESI). Anal Chem 2020; 92:13734-13741. [PMID: 32930596 DOI: 10.1021/acs.analchem.0c01597] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
In this study, we applied a new IR laser-beam-focusing technique to enable sub-100 μm spatial resolution in laser ablation atmospheric pressure photoionization (LAAPPI) and laser ablation electrospray ionization (LAESI) mass spectrometry imaging (MSI). After optimization of operational parameters, both LAAPPI- and LAESI-MSI with a spatial resolution of 70 μm produced high-quality MS images, which allowed accurate localization of metabolites and lipids in the mouse and rat brain. Negative and positive ion LAAPPI- and LAESI-MS detected many of the same metabolites and lipids in the brain. Many compounds were also detected either by LAAPPI- or LAESI-MS, indicating that LAAPPI and LAESI are more complementary than alternative methods.
Collapse
Affiliation(s)
- Juha-Pekka Hieta
- Drug Research Program and Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, P.O. Box 56, FI-00014 Helsinki, Finland
| | - Jaakko Kopra
- Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, P.O. Box 56, FI-00014 Helsinki, Finland
| | - Heikki Räikkönen
- Drug Research Program and Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, P.O. Box 56, FI-00014 Helsinki, Finland
| | - Tiina J Kauppila
- Finnish Institute for the Verification of the Chemical Weapons Convention (VERIFIN), Department of Chemistry, University of Helsinki, P.O. Box 55, FI-00014 Helsinki, Finland
| | - Risto Kostiainen
- Drug Research Program and Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, P.O. Box 56, FI-00014 Helsinki, Finland
| |
Collapse
|
34
|
Ferreira-Junior NC, Ruggeri A, Silva SD, Zampieri TT, Ceroni A, Michelini LC. Exercise training increases GAD65 expression, restores the depressed GABA A receptor function within the PVN and reduces sympathetic modulation in hypertension. Physiol Rep 2020; 7:e14107. [PMID: 31264387 PMCID: PMC6603325 DOI: 10.14814/phy2.14107] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Accepted: 05/09/2019] [Indexed: 11/24/2022] Open
Abstract
GABAergic inhibitory input within the paraventricular hypothalamic nucleus (PVN) plays a key role in restraining sympathetic outflow. Although experimental evidence has shown depressed GABAA receptor function plus sympathoexcitation in hypertension and augmented GABA levels with reduced sympathetic activity after exercise training (T), the mechanisms underlying T‐induced effects remain unclear. Here we investigated in T and sedentary (S) SHR and WKY: (1) time‐course changes of hemodynamic parameters and PVN glutamic acid decarboxylase (GAD) isoforms’ expression; (2) arterial pressure (AP) and heart rate (HR) responses, sympathetic/parasympathetic modulation of heart and vessels and baroreflex sensitivity to GABAA receptor blockade within the PVN. SHR‐S versus WKY‐S exhibited higher AP and HR, increased sympathetic reduced parasympathetic modulation, smaller baroreflex sensitivity, and reduced PVN GAD65 immunoreactivity. SHR‐T and WKY‐T showed prompt maintained increase (2–8 weeks) in GAD65 expression (responsible for GABA vesicular pool synthesis), which occurred simultaneously with HR reduction in SHR‐T and preceded MAP fall in SHR‐T and resting bradycardia in WKY‐T. There was no change in GAD67 expression (mainly involved with GABA metabolic pool). Resting HR in both groups and basal MAP in SHR were negatively correlated with PVN GAD65 expression. Normalized baroreflex sensitivity and autonomic control observed only in SHR‐T were due to recovery of GABAA receptor function into the PVN since bicuculline administration abolished these effects. Data indicated that training augments in both groups the expression/activity of GABAergic neurotransmission within presympathetic PVN neurons and restores GABAA receptors′ function specifically in the SHR, therefore strengthening GABAergic modulation of sympathetic outflow in hypertension.
Collapse
Affiliation(s)
- Nilson C Ferreira-Junior
- Department of Physiology & Biophysics, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, SP, Brazil
| | - Adriana Ruggeri
- Department of Physiology & Biophysics, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, SP, Brazil
| | - Sebastião D Silva
- Department of Physiology & Biophysics, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, SP, Brazil
| | - Thais T Zampieri
- Department of Physiology & Biophysics, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, SP, Brazil
| | - Alexandre Ceroni
- Department of Physiology & Biophysics, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, SP, Brazil
| | - Lisete C Michelini
- Department of Physiology & Biophysics, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, SP, Brazil
| |
Collapse
|
35
|
Meltzer-Brody S, Kanes SJ. Allopregnanolone in postpartum depression: Role in pathophysiology and treatment. Neurobiol Stress 2020; 12:100212. [PMID: 32435663 PMCID: PMC7231991 DOI: 10.1016/j.ynstr.2020.100212] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Revised: 01/22/2020] [Accepted: 01/30/2020] [Indexed: 02/06/2023] Open
Abstract
Postpartum depression (PPD) is a unique subtype of major depressive disorder and a substantial contributor to maternal morbidity and mortality. In addition to affecting the mother, PPD can have short- and long-term consequences for the infant and partner. The precise etiology of PPD is unknown, but proposed mechanisms include altered regulation of stress response pathways, such as the hypothalamic-pituitary-adrenal axis, and dysfunctional gamma-aminobutyric acid (GABA) signaling, and functional linkages exist between these pathways. Current PPD pharmacotherapies are not directly related to these proposed pathophysiologies. In this review, we focus on the potential role of GABAergic signaling and the GABAA receptor positive allosteric modulator allopregnanolone in PPD. Data implicating GABAergic signaling and allopregnanolone in PPD are discussed in the context of the development of brexanolone injection, an intravenous formulation of allopregnanolone recently approved by the United States Food and Drug Administration for the treatment of adult women with PPD.
Collapse
Affiliation(s)
- Samantha Meltzer-Brody
- Department of Psychiatry, School of Medicine, The University of North Carolina at Chapel Hill, USA
- Corresponding author.
| | | |
Collapse
|
36
|
Melnick I, Krishtal OA, Colmers WF. Integration of energy homeostasis and stress by parvocellular neurons in rat hypothalamic paraventricular nucleus. J Physiol 2020; 598:1073-1092. [PMID: 31952096 DOI: 10.1113/jp279387] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Accepted: 01/16/2020] [Indexed: 11/08/2022] Open
Abstract
KEY POINTS Central regulation of energy homeostasis and stress are believed to be reciprocally regulated, i.e. excessive food intake suppresses, while prolonged hunger exacerbates, stress responses in vivo. This relationship may be mediated by neuroendocrine parvocellular corticotropin-releasing hormone (CRH) neurons in the hypothalamic paraventricular nucleus that receive both stress- and feeding-related input. We find that hunger strongly and selectively potentiates, while re-feeding suppresses, a cellular analogue of a stress response induced by acute glucopenia in CRH neurons in rat hypothalamic slices. Neuronal activation in response to glucopenia was mediated synaptically, via the relative enhancement of glutamate over GABA input. These results illustrate how acute stress responses may be initiated in vivo and show that it is reciprocally integrated with energy balance via local hypothalamic mechanisms acting at the level of CRH neurons and their afferent terminals. ABSTRACT Increased food intake is a common response to help cope with stress, implying the existence of a previously postulated but imperfectly understood, inverse relationship between the regulation of feeding and stress. We have identified components of the neural circuitry that can integrate these homeostatic responses. Prior fasting (∼24 h) potentiates, and re-feeding suppresses, excitatory responses to acute glucopenia in about half of the corticotropin releasing hormone (CRH)-expressing, putatively neurosecretory, stress-related neurons in the paraventricular nucleus of the hypothalamus studied. Glucoprivation stress ex vivo resulted from a preferential relative increase in excitatory (glutamatergic) over inhibitory (GABAergic) inputs. Putative preautonomic cells were less sensitive to fasting, and showed a predominant inhibition to acute glucopenia. We conclude that hunger may sensitize hypothalamic stress responses by acting via local mechanisms, at the level of CRH neurons and their presynaptic inputs. Those mechanisms involve neither presynaptic ATP-sensitive potassium channels nor postsynaptic ATP levels.
Collapse
Affiliation(s)
- Igor Melnick
- Bogomoletz Institute of Physiology, Bogomoletz str 4, Kiev, 01024, Ukraine
| | - Oleg A Krishtal
- Bogomoletz Institute of Physiology, Bogomoletz str 4, Kiev, 01024, Ukraine
| | - William F Colmers
- Department of Pharmacology, University of Alberta, Edmonton, Alberta, Canada, T6G 2H7
| |
Collapse
|
37
|
Misztal T, Czauderna MR, Młotkowska P, Misztal A, Marciniak E. Temporal changes in the cerebrospinal fluid allopregnanolone concentration and hypothalamic-pituitary-adrenal axis activity in sheep during pregnancy and early lactation. Livest Sci 2020. [DOI: 10.1016/j.livsci.2019.103871] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
|
38
|
Auer T, Schreppel P, Erker T, Schwarzer C. Impaired chloride homeostasis in epilepsy: Molecular basis, impact on treatment, and current treatment approaches. Pharmacol Ther 2020; 205:107422. [DOI: 10.1016/j.pharmthera.2019.107422] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Accepted: 10/07/2019] [Indexed: 12/14/2022]
|
39
|
Guo S, Tang W, Hu Y, Chen Y, Gordon A, Li B, Li P. Enhancement of On-tissue Chemical Derivatization by Laser-Assisted Tissue Transfer for MALDI MS Imaging. Anal Chem 2019; 92:1431-1438. [PMID: 31800227 DOI: 10.1021/acs.analchem.9b04618] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The development of on-tissue chemical derivatization methods for matrix-assisted laser desorption/ionization (MALDI) mass spectrometry imaging (MSI) of small endogenous metabolites in tissues has attracted great attention for their advantages in improving detection sensitivity and ionization efficiency of poorly ionized and low abundant metabolites. Herein, a laser-assisted tissue transfer (LATT) technique was developed to enhance on-tissue derivatization of small molecules. Using a focused blue laser, a thin-layer tissue film (∼1 μm) was transferred to an acceptor slide from a 6 μm dry tissue section preliminarily coated with derivatization and matrix reagents. The acceptor slide with its ablated constituents was then imaged by MALDI MS. On-tissue chemical derivatization with amino-specific derivatization reagent 4-hydroxy-3-methoxycinnamaldehyde (CA) was carried out on LATT system. 20-235 folds increase in signal intensity for CA derivatized metabolites such as amino acids, neurotransmitters, and dipeptides were observed from rat brain tissues in comparison with conventional incubation-based derivatization. This technique was further extended to derivatize steroids with Girard reagent T (GirT). The remarkable derivatization efficiency can mainly be attributed to the minimization of ion suppression effects due to the reduced thickness of tissue section and endogenous components. Additionally, shorter derivatization time with no obvious metabolite delocalization was achieved using LATT method. These results demonstrate the advantages of LATT in the enhancement of on-tissue derivatization for the more specific and sensitive imaging of small metabolites in tissues with MALDI MS.
Collapse
Affiliation(s)
- Shuai Guo
- State Key Laboratory of Natural Medicines , China Pharmaceutical University , Nanjing , 210009 , China.,School of Basic Medicine and Clinical Pharmacy , China Pharmaceutical University , Nanjing , 211198 , China
| | - Weiwei Tang
- State Key Laboratory of Natural Medicines , China Pharmaceutical University , Nanjing , 210009 , China.,School of Traditional Chinese Pharmacy , China Pharmaceutical University , Nanjing , 211198 , China
| | - Yu Hu
- State Key Laboratory of Natural Medicines , China Pharmaceutical University , Nanjing , 210009 , China.,School of Traditional Chinese Pharmacy , China Pharmaceutical University , Nanjing , 211198 , China
| | - Yanwen Chen
- State Key Laboratory of Natural Medicines , China Pharmaceutical University , Nanjing , 210009 , China.,School of Traditional Chinese Pharmacy , China Pharmaceutical University , Nanjing , 211198 , China
| | - Andrew Gordon
- State Key Laboratory of Natural Medicines , China Pharmaceutical University , Nanjing , 210009 , China.,School of Traditional Chinese Pharmacy , China Pharmaceutical University , Nanjing , 211198 , China
| | - Bin Li
- State Key Laboratory of Natural Medicines , China Pharmaceutical University , Nanjing , 210009 , China.,School of Traditional Chinese Pharmacy , China Pharmaceutical University , Nanjing , 211198 , China
| | - Ping Li
- State Key Laboratory of Natural Medicines , China Pharmaceutical University , Nanjing , 210009 , China.,School of Traditional Chinese Pharmacy , China Pharmaceutical University , Nanjing , 211198 , China
| |
Collapse
|
40
|
Allopregnanolone-based treatments for postpartum depression: Why/how do they work? Neurobiol Stress 2019; 11:100198. [PMID: 31709278 PMCID: PMC6838978 DOI: 10.1016/j.ynstr.2019.100198] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Revised: 10/12/2019] [Accepted: 10/16/2019] [Indexed: 01/01/2023] Open
Abstract
Recent FDA approval of an allopregnanolone-based treatment specifically for postpartum depression, brexanolone, now commercially called Zulresso®, is an exciting development for patients and families impacted by postpartum depression and allows us to start asking questions about why and how this compound is so effective. Allopregnanolone is a neuroactive steroid, or neurosteroid, which can be synthesized from steroid hormone precursors, such as progesterone, or synthesized de novo from cholesterol. Neurosteroids are positive allosteric modulators at GABAA receptors (GABAARs), a property which is thought to mediate the therapeutic effects of these compounds. However, the durability of effect of brexanolone in clinical trials questions the mechanism of action mediating the remarkable antidepressant effects, leading us to ask why and how does this drug work. Asking why this drug is effective may provide insight into the underlying neurobiology of postpartum depression. Exploring how this drug works will potentially elucidate a novel antidepressant mechanism of action and may provide useful information for next generation drug development. In this review, we examine the clinical and preclinical evidence supporting a role for allopregnanolone in the underlying neurobiology of postpartum depression as well as foundational evidence supporting the therapeutic effects of allopregnanolone for treatment of postpartum depression.
Collapse
|
41
|
Induction of hypothalamic GABA synthetic enzymes mRNA (Gad 1 and Gad 2) expression by negative energy balance in broiler and layer chicks. Neurosci Lett 2019; 712:134498. [PMID: 31526832 DOI: 10.1016/j.neulet.2019.134498] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 09/04/2019] [Accepted: 09/12/2019] [Indexed: 12/20/2022]
Abstract
Broiler and layer chicks have been selected for higher and lower food intake and body weight gain, respectively. It has recently been reported that glutamate decarboxylase (Gad1) mRNA, a gamma-aminobutyric acid (GABA) synthetic enzyme gene, is a reliable proxy for GABA release. Previous studies have revealed that GABAergic system has a stimulatory role on food intake in both mammals and birds. Over the recent years, evidence has identified the presence of GABAergic neurons as either the first- or second-order neurons within the various feeding nuclei of hypothalamus of laboratory rodents. They respond to the negative energy balance representing a critical role for GABA in the regulation of food intake. In the current study, the mRNA abundance of Gad 1 and Gad 2 was measured within the hypothalamus of both broiler and layer free fed, 12 h-fasted and 12 h-fasted / 3 h refed chicks. Furthermore, the effect of intracerebroventricular (ICV) injection of GABA was studied on food intake of chicks. The results indicated an increase in both Gad 1 and 2 expressions during fasting which tended to return to the baseline after refeeding. However, this increase was greater in broilers than in layers. The results also showed that ICV injection of GABA had no effect on food intake with the exception of an increase in free fed broilers. This study suggests a role for hypothalamic GABAergic system in birds that respond to negative energy balance, which seems to be more considerable in broilers than in layers.
Collapse
|
42
|
Kim YB, Colwell CS, Kim YI. Long-term ionic plasticity of GABAergic signalling in the hypothalamus. J Neuroendocrinol 2019; 31:e12753. [PMID: 31166034 DOI: 10.1111/jne.12753] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Revised: 05/24/2019] [Accepted: 06/03/2019] [Indexed: 12/23/2022]
Abstract
The hypothalamus contains a number of nuclei that subserve a variety of functions, including generation of circadian rhythms, regulation of hormone secretion and maintenance of homeostatic levels for a variety of physiological parameters. Within the hypothalamus, γ-amino-butyric acid (GABA) is one of the major neurotransmitters responsible for cellular communication. Although GABA most commonly serves as an inhibitory neurotransmitter, a growing body of evidence indicates that it can evoke post-synaptic excitation as a result of the active regulation of intracellular chloride concentration. In this review, we consider the evidence for this ionic plasticity of GABAergic synaptic transmission in five distinct cases in hypothalamic cell populations. We argue that this plasticity serves as part of the functional response to or is at least associated with dehydration, lactation, hypertension and stress. As such, GABA excitation should be considered as part of the core homeostatic mechanisms of the hypothalamus.
Collapse
Affiliation(s)
- Young-Beom Kim
- Department of Physiology, Korea University College of Medicine, Seoul, Korea
- Neuroscience Research Institute, Korea University, Seoul, Korea
| | - Christopher S Colwell
- Department of Psychiatry and Biobehavioral Sciences, UCLA School of Medicine, Los Angeles, CA, USA
| | - Yang In Kim
- Department of Physiology, Korea University College of Medicine, Seoul, Korea
- Neuroscience Research Institute, Korea University, Seoul, Korea
| |
Collapse
|
43
|
Seifikalhor M, Aliniaeifard S, Hassani B, Niknam V, Lastochkina O. Diverse role of γ-aminobutyric acid in dynamic plant cell responses. PLANT CELL REPORTS 2019; 38:847-867. [PMID: 30739138 DOI: 10.1007/s00299-019-02396-z] [Citation(s) in RCA: 92] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Accepted: 02/02/2019] [Indexed: 05/05/2023]
Abstract
Gamma-aminobutyric acid (GABA), a four-carbon non-protein amino acid, is found in most prokaryotic and eukaryotic organisms. Although, ample research into GABA has occurred in mammals as it is a major inhibitory neurotransmitter; in plants, a role for GABA has often been suggested as a metabolite that changes under stress rather than as a signal, as no receptor or motif for GABA binding was identified until recently and many aspects of its biological function (ranging from perception to function) remain to be answered. In this review, flexible properties of GABA in regulation of plant responses to various environmental biotic and abiotic stresses and its integration in plant growth and development either as a metabolite or a signaling molecule are discussed. We have elaborated on the role of GABA in stress adaptation (i.e., salinity, hypoxia/anoxia, drought, temperature, heavy metals, plant-insect interplay and ROS-related responses) and its contribution in non-stress-related biological pathways (i.e., involvement in plant-microbe interaction, contribution to the carbon and nitrogen metabolism and governing of signal transduction pathways). This review aims to represent the multifunctional contribution of GABA in various biological and physiological mechanisms under stress conditions; the objective is to review the current state of knowledge about GABA role beyond stress-related responses. Our effort is to place findings about GABA in an organized and broader context to highlight its shared metabolic and biologic functions in plants under variable conditions. This will provide potential modes of GABA crosstalk in dynamic plant cell responses.
Collapse
Affiliation(s)
- Maryam Seifikalhor
- Department of Plant Biology, Center of Excellence in Phylogeny of Living Organisms in Iran, School of Biology, College of Science, University of Tehran, Tehran, 14155, Iran
| | - Sasan Aliniaeifard
- Department of Horticulture, College of Aburaihan, University of Tehran, Tehran, Iran.
| | - Batool Hassani
- Department of Plant Sciences, Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, Iran
| | - Vahid Niknam
- Department of Plant Biology, Center of Excellence in Phylogeny of Living Organisms in Iran, School of Biology, College of Science, University of Tehran, Tehran, 14155, Iran
| | - Oksana Lastochkina
- Bashkir Research Institute of Agriculture, Russian Academy of Sciences, Ufa, Russia
- Institute of Biochemistry and Genetics, Russian Academy of Sciences, Ufa, Russia
| |
Collapse
|
44
|
El-Naggar K, El-Kassas S, Abdo SE, Kirrella AAK, Al Wakeel RA. Role of gamma-aminobutyric acid in regulating feed intake in commercial broilers reared under normal and heat stress conditions. J Therm Biol 2019; 84:164-175. [PMID: 31466750 DOI: 10.1016/j.jtherbio.2019.07.004] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Revised: 06/22/2019] [Accepted: 07/01/2019] [Indexed: 12/17/2022]
Abstract
This study was conducted to investigate the effects of dietary GABA supplementation on blood biochemical parameters, the overall growth performance, and the relative mRNA expression of some FI- regulating genes in broiler chickens. A total of 192, three-day old chicks of mixed sex from two commercial broiler strains (Ross 308 and Cobb 500) were distributed into 2 groups; a control group and GABA-supplemented group (100 mg/kg diet). When the chicks reached 21 days of age, each group of each strain was randomly subdivided into two subgroups: one was exposed to HS (33 ± 2 °C for 5 h/day for 2 weeks), while the other remained at thermoneutral temperature (24 °C). GABA significantly improved bird growth performance under normal and HS conditions, by increasing body weight (BW), weight gain (WG), and FI and significantly reduced the elevated body temperature of birds under HS. GABA supplementation increased FI by reducing the mRNA expression levels of FI-inhibiting neuropeptides, such as POMC, leptin, Ghrelin, and CCK, during HS and by increasing the expression of FI-stimulating neuropeptides such as AgRP and NPY. Moreover, GABA significantly altered FAS and ACC gene expression, resulting in significant increases in abdominal fat content in birds reared normally. In contrast, GABA lowered fat content in Cobb birds and increased it in Ross birds under HS. Therefore, GABA (100 mg/kg diet) is a strong FI-stimulating neurotransmitter and its regulatory effects depend on broiler strain and housing temperature.
Collapse
Affiliation(s)
- Karima El-Naggar
- Department of Nutrition and Veterinary Clinical Nutrition, Faculty of Veterinary Medicine, Alexandria University, 22758, Egypt.
| | - Seham El-Kassas
- Animal, Poultry and Fish Breeding and Production, Department of Animal Wealth Development, Faculty of Veterinary Medicine, Kafrelsheikh University, Egypt
| | - Safaa E Abdo
- Genetics and Genetic Engineering, Department of Animal Wealth Development, Faculty of Veterinary Medicine, Kafrelsheikh University, Egypt
| | - Abeer A K Kirrella
- Poultry Physiology, Poultry Production Department, Faculty of Agriculture, Kafrelsheikh University, Egypt
| | - Rasha A Al Wakeel
- Department of Physiology, Faculty of Veterinary Medicine, Kafrelsheikh University, Egypt
| |
Collapse
|
45
|
Zhang BB, Jin H, Bing YH, Zhang XY, Chu CP, Li YZ, Qiu DL. A Nitric Oxide-Dependent Presynaptic LTP at Glutamatergic Synapses of the PVN Magnocellular Neurosecretory Cells in vitro in Rats. Front Cell Neurosci 2019; 13:283. [PMID: 31316353 PMCID: PMC6610542 DOI: 10.3389/fncel.2019.00283] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Accepted: 06/11/2019] [Indexed: 11/13/2022] Open
Abstract
The magnocellular neurosecretory cells (MNCs) of the hypothalamic paraventricular nucleus (PVN) integrate incoming signals to secrete oxytocin (OT), and vasopressin (VP) from their nerve terminals in the posterior pituitary gland. In the absence of gamma-aminobutyric acid A (GABAA) and cannabinoids 1 (CB1) receptor activity, we used whole-cell patch-clamp recording, single-cell reverse transcription-multiplex polymerase chain reaction (SC-RT-mPCR), biocytin histochemistry and pharmacological methods to examine the mechanism of high frequency stimulus (HFS, 100 Hz)-induced long-term potentiation (LTP) at glutamatergic synapses in the PVN MNCs of juvenile male rats. Our results showed that HFS-induced LTP at glutamatergic synapses was accompanied by a decrease in the paired-pulse ratio (PPR) of the PVN MNCs. In these MNCs, HFS-induced LTP persisted in the presence of a group 1 metabotropic glutamate receptor (mGluR1) antagonist; however, it was abolished by an N-methyl-D-aspartic acid (NMDA) receptor blocker. Notably, HFS-induced LTP in the PVN MNCs was completely prevented by a nitric oxide synthase (NOS) inhibitor. The application of an NO donor not only induced the LTP of excitatory glutamatergic inputs in the PVN MNCs, but also occluded the HFS-induced LTP in these MNCs. Moreover, HFS-induced LTP in the PVN MNCs was also abolished by a specific protein kinase A (PKA) inhibitor, KT5720. SC-RT-mPCR analysis revealed that 64.5% (62/96) of MNCs expressed OT mRNA. Our results indicate that a HFS can induce an NMDA receptor and NO cascades dependent on presynaptic glutamatergic LTP in the PVN MNCs via a PKA signaling pathway.
Collapse
Affiliation(s)
- Bin-Bin Zhang
- Key Laboratory of Cellular Function and Pharmacology of Jilin Province, Yanbian University, Yanji, China.,Department of Physiology and Pathophysiology, College of Medicine, Yanbian University, Yanji, China
| | - Hua Jin
- Key Laboratory of Cellular Function and Pharmacology of Jilin Province, Yanbian University, Yanji, China.,Department of Nephrology, Affiliated Hospital of Yanbian University, Yanji, China
| | - Yan-Hua Bing
- Key Laboratory of Cellular Function and Pharmacology of Jilin Province, Yanbian University, Yanji, China.,Department of Physiology and Pathophysiology, College of Medicine, Yanbian University, Yanji, China
| | - Xin-Yuan Zhang
- Department of Physiology and Pathophysiology, College of Medicine, Yanbian University, Yanji, China
| | - Chun-Ping Chu
- Key Laboratory of Cellular Function and Pharmacology of Jilin Province, Yanbian University, Yanji, China
| | - Yu-Zi Li
- Key Laboratory of Cellular Function and Pharmacology of Jilin Province, Yanbian University, Yanji, China.,Department of Cardiology, Affiliated Hospital of Yanbian University, Yanji, China
| | - De-Lai Qiu
- Key Laboratory of Cellular Function and Pharmacology of Jilin Province, Yanbian University, Yanji, China.,Department of Physiology and Pathophysiology, College of Medicine, Yanbian University, Yanji, China
| |
Collapse
|
46
|
Gomes-de-Souza L, Benini R, Costa-Ferreira W, Crestani CC. GABA A but not GABA B receptors in the lateral hypothalamus modulate the tachycardic response to emotional stress in rats. Eur Neuropsychopharmacol 2019; 29:672-680. [PMID: 30878320 DOI: 10.1016/j.euroneuro.2019.03.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2018] [Revised: 02/05/2019] [Accepted: 03/02/2019] [Indexed: 01/26/2023]
Abstract
The lateral hypothalamus (LH) has been described as one of the hypothalamic areas involved in the behavioral and physiological responses triggered by aversive stimuli. Previous studies indicated involvement of the LH in cardiovascular responses to stress. Despite this evidence, the local neurochemical mechanisms involved in LH control of stress responses is still poorly understood. Therefore, in the present study, we investigated the role of GABAergic neurotransmission within the LH in cardiovascular responses induced by an acute session of restraint stress in rats. For this, we evaluated the effect of bilateral microinjection of selective antagonists of either GABAA or GABAB receptors into the LH on arterial pressure increase, heart rate (HR) increase and reduction in tail skin temperature induced by restraint stress. We found that microinjection of the selective GABAA receptor antagonist SR95531 into the LH decreased the increase in HR caused by restraint stress, but without affecting the increase in arterial pressure increase or the reduction in tail skin temperature. Conversely, LH treatment with the selective GABAB receptor antagonist CGP35348 did not affect the restraint-evoked cardiovascular changes. These findings indicate that GABAergic neurotransmission in the LH, acting through activation of local GABAA receptors, plays a facilitatory role in the tachycardic response observed during aversive threats.
Collapse
Affiliation(s)
- Lucas Gomes-de-Souza
- Laboratory of Pharmacology, School of Pharmaceutical Sciences, São Paulo State University (UNESP), Rodovia Araraquara-Jau Km 01 (Campus Universitário), Campus Ville, 14800-903 Araraquara, SP, Brazil; Joint UFSCar-UNESP Graduate Program in Physiological Sciences, São Carlos, SP, Brazil
| | - Ricardo Benini
- Laboratory of Pharmacology, School of Pharmaceutical Sciences, São Paulo State University (UNESP), Rodovia Araraquara-Jau Km 01 (Campus Universitário), Campus Ville, 14800-903 Araraquara, SP, Brazil; Joint UFSCar-UNESP Graduate Program in Physiological Sciences, São Carlos, SP, Brazil
| | - Willian Costa-Ferreira
- Laboratory of Pharmacology, School of Pharmaceutical Sciences, São Paulo State University (UNESP), Rodovia Araraquara-Jau Km 01 (Campus Universitário), Campus Ville, 14800-903 Araraquara, SP, Brazil; Joint UFSCar-UNESP Graduate Program in Physiological Sciences, São Carlos, SP, Brazil
| | - Carlos C Crestani
- Laboratory of Pharmacology, School of Pharmaceutical Sciences, São Paulo State University (UNESP), Rodovia Araraquara-Jau Km 01 (Campus Universitário), Campus Ville, 14800-903 Araraquara, SP, Brazil; Joint UFSCar-UNESP Graduate Program in Physiological Sciences, São Carlos, SP, Brazil.
| |
Collapse
|
47
|
Maguire J. Neuroactive Steroids and GABAergic Involvement in the Neuroendocrine Dysfunction Associated With Major Depressive Disorder and Postpartum Depression. Front Cell Neurosci 2019; 13:83. [PMID: 30906252 PMCID: PMC6418819 DOI: 10.3389/fncel.2019.00083] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Accepted: 02/19/2019] [Indexed: 12/21/2022] Open
Abstract
Stress and previous adverse life events are well-established risk factors for depression. Further, neuroendocrine disruptions are associated with both major depressive disorder (MDD) and postpartum depression (PPD). However, the mechanisms whereby stress contributes to the underlying neurobiology of depression remains poorly understood. The hypothalamic-pituitary-adrenal (HPA) axis, which mediates the body's neuroendocrine response to stress, is tightly controlled by GABAergic signaling and there is accumulating evidence that GABAergic dysfunction contributes to the impact of stress on depression. GABAergic signaling plays a critical role in the neurobiological effects of stress, not only by tightly controlling the activity of the HPA axis, but also mediating stress effects in stress-related brain regions. Deficits in neuroactive steroids and neurosteroids, some of which are positive allosteric modulators of GABAA receptors (GABAARs), such as allopregnanolone and THDOC, have also been implicated in MDD and PPD, further supporting a role for GABAergic signaling in depression. Alterations in neurosteroid levels and GABAergic signaling are implicated as potential contributing factors to neuroendocrine dysfunction and vulnerability to MDD and PPD. Further, potential novel treatment strategies targeting these proposed underlying neurobiological mechanisms are discussed. The evidence summarized in the current review supports the notion that MDD and PPD are stress-related psychiatric disorders involving neurosteroids and GABAergic dysfunction.
Collapse
Affiliation(s)
- Jamie Maguire
- Neuroscience Department, Tufts University School of Medicine, Boston, MA, United States
| |
Collapse
|
48
|
He Y, Ouyang J, Hu Z, Yang J, Chu Y, Huang S, Yang Y, Liu C. Intervention mechanism of repeated oral GABA administration on anxiety-like behaviors induced by emotional stress in rats. Psychiatry Res 2019; 271:649-657. [PMID: 30791338 DOI: 10.1016/j.psychres.2018.12.025] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2018] [Revised: 12/03/2018] [Accepted: 12/05/2018] [Indexed: 02/07/2023]
Abstract
The purpose of this study was to investigate the effects and mechanism of repeated oral administration of gamma-aminobutyric acid (GABA) on anxiety-like behaviors induced by emotional stress. Male Sprague-Dawley rats were randomly divided into five groups (8 rats each): control, emotional stress model, three emotional stress + GABA-treated groups (0.5, 1, 2 mg/kg). The rats were given empty water bottles after the training of drinking water to induce emotional stress. Each group was treated with saline or different doses of GABA respectively for 21 consecutive days. Then open field and elevated plus maze were used to assess anxiety-like behaviors. Both frontal cortex and plasma NO metabolites nitrate and nitrite (NOx) levels were determined spectrophotometrically. Results showed that oral administration of GABA significantly reversed the stress-induced anxiety-like negative responses dose-dependently. The frontal cortex NOx levels were lower in stressed rats than in control group (P < 0.05), but higher in 2 mg/kg GABA-treated group than stress model group (P < 0.05). On the other hand, NOx levels in plasma showed a gradual decline trend. Collectively, these results suggest that short repeated oral administration of GABA has an anxiolytic-like effect possibly via preventing NO reduction caused by stress and improving availability of NO in the frontal cortex.
Collapse
Affiliation(s)
- Yongjian He
- College of Food Science, South China Agricultural University, Guangzhou, GZ, China; Guangdong Provincial Key Laboratory of Food Quality and Safety, South China Agricultural University, Guangzhou 510642, China
| | - Junyan Ouyang
- College of Food Science, South China Agricultural University, Guangzhou, GZ, China; Guangdong Provincial Key Laboratory of Food Quality and Safety, South China Agricultural University, Guangzhou 510642, China
| | - Zhuoyan Hu
- College of Food Science, South China Agricultural University, Guangzhou, GZ, China
| | - Jie Yang
- College of Food Science, South China Agricultural University, Guangzhou, GZ, China; Guangdong Provincial Key Laboratory of Food Quality and Safety, South China Agricultural University, Guangzhou 510642, China
| | - Yue Chu
- College of Food Science, South China Agricultural University, Guangzhou, GZ, China; Guangdong Provincial Key Laboratory of Food Quality and Safety, South China Agricultural University, Guangzhou 510642, China
| | - Shaowen Huang
- College of Food Science, South China Agricultural University, Guangzhou, GZ, China; Guangdong Provincial Key Laboratory of Food Quality and Safety, South China Agricultural University, Guangzhou 510642, China
| | - Yichao Yang
- School of Public Health, Guangzhou Medical University, Guangzhou, GZ, China
| | - Chunhong Liu
- College of Food Science, South China Agricultural University, Guangzhou, GZ, China; Guangdong Provincial Key Laboratory of Food Quality and Safety, South China Agricultural University, Guangzhou 510642, China.
| |
Collapse
|
49
|
Sunstrum JK, Inoue W. Heterosynaptic modulation in the paraventricular nucleus of the hypothalamus. Neuropharmacology 2018; 154:87-95. [PMID: 30408488 DOI: 10.1016/j.neuropharm.2018.11.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Revised: 10/18/2018] [Accepted: 11/03/2018] [Indexed: 12/21/2022]
Abstract
The stress response-originally described by Hans Selye as "the nonspecific response of the body to any demand made upon it"-is chiefly mediated by the hypothalamic-pituitary-adrenal (HPA) axis and is activated by diverse sensory stimuli that inform threats to homeostasis. The diversity of signals regulating the HPA axis is partly achieved by the complexity of afferent inputs that converge at the apex of the HPA axis: this apex is formed by a group of neurosecretory neurons that synthesize corticotropin-releasing hormone (CRH) in the paraventricular nucleus of the hypothalamus (PVN). The afferent synaptic inputs onto these PVN-CRH neurons originate from a number of brain areas, and PVN-CRH neurons respond to a long list of neurotransmitters/neuropeptides. Considering this complexity, an important question is how these diverse afferent signals independently and/or in concert influence the excitability of PVN-CRH neurons. While many of these inputs directly act on the postsynaptic PVN-CRH neurons for the summation of signals, accumulating data indicates that they also modulate each other's transmission in the PVN. This mode of transmission, termed heterosynaptic modulation, points to mechanisms through which the activity of a specific modulatory input (conveying a specific sensory signal) can up- or down-regulate the efficacy of other afferent synapses (mediating other stress modalities) depending on receptor expression for and spatial proximity to the heterosynaptic signals. Here, we review examples of heterosynaptic modulation in the PVN and discuss its potential role in the regulation of PVN-CRH neurons' excitability and resulting HPA axis activity. This article is part of the Special Issue entitled 'Hypothalamic Control of Homeostasis'.
Collapse
Affiliation(s)
- Julia K Sunstrum
- Neuroscience Program, Schulich School of Medicine & Dentistry, University of Western Ontario, London, Ontario, Canada
| | - Wataru Inoue
- Neuroscience Program, Schulich School of Medicine & Dentistry, University of Western Ontario, London, Ontario, Canada; Robarts Research Institute, Schulich School of Medicine & Dentistry, University of Western Ontario, London, Ontario, Canada; Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada.
| |
Collapse
|
50
|
Le Tissier P, Fiordelisio Coll T, Mollard P. The Processes of Anterior Pituitary Hormone Pulse Generation. Endocrinology 2018; 159:3524-3535. [PMID: 30020429 DOI: 10.1210/en.2018-00508] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Accepted: 07/11/2018] [Indexed: 12/16/2022]
Abstract
More than 60 years ago, Geoffrey Harris described his "neurohumoral theory," in which the regulation of pituitary hormone secretion was a "simple" hierarchal relationship, with the hypothalamus as the controller. In models based on this theory, the electrical activity of hypothalamic neurons determines the release of hypophysiotropic hormones into the portal circulation, and the pituitary simply responds with secretion of a pulse of hormone into the bloodstream. The development of methodologies allowing the monitoring of the activities of members of the hypothalamic-vascular-pituitary unit is increasingly allowing dissection of the mechanisms generating hypothalamic and pituitary pulses. These have revealed that whereas hypothalamic input is required, its role as a driver of pulsatile pituitary hormone secretion varies between pituitary axes. The organization of pituitary cells has a key role in the modification of their response to hypophysiotropic factors that can lead to a memory of previous demand and enhanced function. Feedback can lead to oscillatory hormone output that is independent of pulses of hypophysiotropic factors and instead, results from the temporal relationship between pituitary output and target organ response. Thus, the mechanisms underlying the generation of pulses cannot be generalized, and the circularity of feedforward and feedback interactions must be considered to understand both normal physiological function and pathology. We describe some examples of the clinical implications of recognizing the importance of the pituitary and target organs in pulse generation and suggest avenues for future research in both the short and long term.
Collapse
Affiliation(s)
- Paul Le Tissier
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Tatiana Fiordelisio Coll
- Institut de Génomique Fonctionnelle, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, University of Montpellier, Montpellier, France
- Laboratorio de Neuroendocrinología Comparada, Departamento de Ecología y Recursos Naturales, Biología, Facultad de Ciencias, Universidad Nacional Autónoma de México, Ciudad Universitaria, México City, Distrito Federal, México
| | - Patrice Mollard
- Institut de Génomique Fonctionnelle, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, University of Montpellier, Montpellier, France
| |
Collapse
|