1
|
Bollinger JL, Johnsamuel S, Vollmer LL, Kuhn AM, Wohleb ES. Stress-induced dysfunction of neurovascular astrocytes in the prefrontal cortex contributes to sex-dependent deficits in cognition and behavior. Mol Psychiatry 2025:10.1038/s41380-025-02993-3. [PMID: 40185903 DOI: 10.1038/s41380-025-02993-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 03/14/2025] [Accepted: 03/26/2025] [Indexed: 04/07/2025]
Abstract
Astrocytes form an integral component of the neurovascular unit, ensheathing brain blood vessels with endfeet high in aquaporin-4 (AQP4) expression. These AQP4-rich endfeet facilitate interaction between the vascular endothelium, astrocytes, and neurons, and help stabilize vascular morphology. Studies using preclinical models of psychological stress and post-mortem tissue from patients with major depressive disorder (MDD) have reported reductions in AQP4, loss of astrocytic structures, and vascular impairment in the prefrontal cortex (PFC). Though compelling, the role of AQP4 in mediating stress-induced alterations in neurovascular function and behavior remains unclear. Here, we address this, alongside potential sex differences in chronic unpredictable stress (CUS) effects on astrocyte phenotype, blood-brain barrier integrity, and behavior. CUS led to more pronounced shifts in stress-coping behavior and working memory deficits in male- as compared to female mice. Following behavioral testing, astrocytes from the frontal cortex were isolated for gene expression analyses. We found that CUS increased transcripts associated with blood vessel maintenance in males, but either had no effect on- or decreased- these transcripts in females. Furthermore, CUS caused a reduction in vascular-localized AQP4 and elevated extravasation of a small fluorescent reporter (Dextran) in the PFC in males but not females. Studies showed that knockdown of AQP4 in the PFC is sufficient to disrupt astrocyte phenotype and increase behavioral susceptibility to a sub-chronic stressor in males yet has little effect on stress susceptibility in females. Our findings provide evidence that sex-specific alterations in astrocyte phenotype and neurovascular integrity in the PFC contribute to cognitive-behavioral consequences following stress.
Collapse
Affiliation(s)
- Justin L Bollinger
- Department of Pharmacology, Physiology, & Neurobiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA.
| | - Shobha Johnsamuel
- Department of Pharmacology, Physiology, & Neurobiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Lauren L Vollmer
- Department of Pharmacology, Physiology, & Neurobiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Alexander M Kuhn
- Department of Pharmacology, Physiology, & Neurobiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Eric S Wohleb
- Department of Pharmacology, Physiology, & Neurobiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| |
Collapse
|
2
|
Bollinger JL, Johnsamuel S, Vollmer LL, Kuhn AM, Wohleb ES. Stress-induced dysfunction of neurovascular astrocytes contributes to sex-specific behavioral deficits. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.14.594147. [PMID: 38798398 PMCID: PMC11118421 DOI: 10.1101/2024.05.14.594147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Astrocytes form an integral component of the neurovascular unit, ensheathing brain blood vessels with projections high in aquaporin-4 (AQP4) expression. These AQP4-rich projections facilitate interaction between the vascular endothelium, astrocytes, and neurons, and help stabilize vascular morphology. Studies using preclinical models of psychological stress and post-mortem tissue from patients with major depressive disorder (MDD) have reported reductions in AQP4, loss of astrocytic structures, and vascular impairment in the prefrontal cortex (PFC). Though compelling, the role of AQP4 in mediating stress-induced alterations in blood vessel function and behavior remains unclear. Here, we address this, alongside potential sex differences in chronic unpredictable stress (CUS) effects on astrocyte phenotype, blood-brain barrier integrity, and behavior. CUS led to pronounced shifts in stress-coping behavior and working memory deficits in male -but not female- mice. Following behavioral testing, astrocytes from the frontal cortex were isolated for gene expression analyses. We found that CUS increased various transcripts associated with blood vessel maintenance in astrocytes from males, but either had no effect on- or decreased- these genes in females. Furthermore, CUS caused a reduction in vascular-localized AQP4 and elevated extravasation of a small molecule fluorescent reporter (Dextran) in the PFC in males but not females. Studies showed that knockdown of AQP4 in the PFC in males is sufficient to disrupt astrocyte phenotype and increase behavioral susceptibility to a sub-chronic stressor. Collectively, these findings provide initial evidence that sex-specific alterations in astrocyte phenotype and neurovascular integrity in the PFC contribute to behavioral and cognitive consequences following chronic stress.
Collapse
Affiliation(s)
- Justin L Bollinger
- Department of Pharmacology & Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH
| | - Shobha Johnsamuel
- Department of Pharmacology & Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH
| | - Lauren L Vollmer
- Department of Pharmacology & Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH
| | - Alexander M Kuhn
- Department of Pharmacology & Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH
| | - Eric S Wohleb
- Department of Pharmacology & Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH
| |
Collapse
|
3
|
Turniak-Kusy M, Studzian M, Szpakowski P, Kuchta P, Smietanka K, Mattern C, Pulaski L, Bielecki B. Testosterone Inhibits Secretion of the Pro-Inflammatory Chemokine CXCL1 from Astrocytes. Curr Issues Mol Biol 2024; 46:2105-2118. [PMID: 38534751 DOI: 10.3390/cimb46030135] [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: 07/21/2023] [Revised: 02/25/2024] [Accepted: 03/01/2024] [Indexed: 03/28/2024] Open
Abstract
Astrocytes play an important role in the regulation of the inflammatory response in the CNS, e.g., in demyelinating diseases. Since the chemokine CXCL1 is known to be secreted by astrocytes and to have a pro-inflammatory effect on immune cells in the CNS, we verified the effect of testosterone on its secretion in vitro (in the astrocytic cell line DI TNC1). Testosterone reduced the increase in CXCL1 production caused by the pro-inflammatory agent lysophosphatidylcholine and restored the basal production level of CXCL1. The androgen receptor (present and functional in the studied cell line) was strongly suggested to mediate this effect-its non-steroid ligand flutamide exerted an agonist-like effect, mimicking the activity of testosterone itself on CXCL1 secretion. This novel mechanism has important implications for the known immunomodulatory effect of testosterone and potentially other androgenic hormones. It provides a potential explanation on the molecular level and shows that astrocytes are important players in inflammatory homeostasis in the CNS and its hormonal regulation. Therefore, it suggests new directions for the development of the therapeutic intervention.
Collapse
Affiliation(s)
| | - Maciej Studzian
- Department of Oncobiology and Epigenetics, Faculty of Biology and Environmental Protection, University of Lodz, 90-237 Lodz, Poland
- Laboratory of Transcriptional Regulation, Institute of Medical Biology, Polish Academy of Sciences, 90-364 Lodz, Poland
| | - Piotr Szpakowski
- Department of Neurology and Stroke, Medical University of Lodz, 90-549 Lodz, Poland
| | - Piotr Kuchta
- Faculty of Medicine, Medical University of Lodz, 90-419 Lodz, Poland
| | - Kaja Smietanka
- Department of Neurology and Stroke, Medical University of Lodz, 90-549 Lodz, Poland
| | - Claudia Mattern
- Oceanographic Center, Nova Southeastern University, Fort Lauderdale, FL 33314, USA
- M&P Pharma AG, 6376 Emmetten, Switzerland
| | - Lukasz Pulaski
- Department of Oncobiology and Epigenetics, Faculty of Biology and Environmental Protection, University of Lodz, 90-237 Lodz, Poland
- Laboratory of Transcriptional Regulation, Institute of Medical Biology, Polish Academy of Sciences, 90-364 Lodz, Poland
| | - Bartosz Bielecki
- Department of Neurology, Laboratory of Neuroimmunology, Medical University of Lodz, 90-153 Lodz, Poland
| |
Collapse
|
4
|
Pfau DR, Baribeau S, Brown F, Khetarpal N, Marc Breedlove S, Jordan CL. Loss of TRPC2 function in mice alters sex differences in brain regions regulating social behaviors. J Comp Neurol 2023; 531:1550-1561. [PMID: 37496437 PMCID: PMC10642801 DOI: 10.1002/cne.25528] [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: 01/20/2023] [Revised: 05/15/2023] [Accepted: 06/29/2023] [Indexed: 07/28/2023]
Abstract
The transient receptor potential cation channel 2 (TRPC2) conveys pheromonal information from the vomeronasal organ (VNO) to the brain. Both male and female mice lacking this gene show altered sex-typical behavior as adults. We asked whether TRPC2, highly expressed in the VNO, normally participates in the development of VNO-recipient brain regions controlling mounting and aggression, two behaviors affected by TRPC2 loss. We now report significant effects of TRPC2 loss in both the posterodorsal aspect of the medial amygdala (MePD) and ventromedial nucleus of the hypothalamus (VMH) of male and female mice. In the MePD, a sex difference in neuron number was eliminated by the TRPC2 knockout (KO), but the effect was complex, with fewer neurons in the right MePD of females, and fewer neurons in the left MePD of males. In contrast, MePD astrocytes were unaffected by the KO. In the ventrolateral (vl) aspect of the VMH, KO females were like wildtype (WT) females, but TRPC2 loss had a dramatic effect in males, with fewer neurons than WT males and a smaller VMHvl overall. We also discovered a glial sex difference in VMHvl of WTs, with females having more astrocytes than males. Interestingly, TRPC2 loss increased astrocyte number in males in this region. We conclude that TRPC2 normally participates in the sexual differentiation of the mouse MePD and VMHvl. These changes in two key VNO-recipient regions may underlie the effects of the TRPC2 KO on behavior.
Collapse
Affiliation(s)
- Daniel R Pfau
- Neuroscience Program, Michigan State University, East Lansing, Michigan, USA
| | - Sarah Baribeau
- Neuroscience Program, Michigan State University, East Lansing, Michigan, USA
| | - Felix Brown
- Neuroscience Program, Michigan State University, East Lansing, Michigan, USA
| | - Niki Khetarpal
- Neuroscience Program, Michigan State University, East Lansing, Michigan, USA
| | - S Marc Breedlove
- Neuroscience Program, Michigan State University, East Lansing, Michigan, USA
| | - Cynthia L Jordan
- Neuroscience Program, Michigan State University, East Lansing, Michigan, USA
| |
Collapse
|
5
|
Mohr MA, Michael NS, DonCarlos LL, Sisk CL. Sex differences in proliferation and attrition of pubertally born cells in the rat posterior dorsal medial amygdala. Dev Cogn Neurosci 2022; 57:101141. [PMID: 35933923 PMCID: PMC9357828 DOI: 10.1016/j.dcn.2022.101141] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 07/08/2022] [Accepted: 07/21/2022] [Indexed: 11/26/2022] Open
Abstract
The rodent posterodorsal medial amygdala (MePD) evaluates and assigns valence to social sensory stimuli. The perception of social stimuli evolves during puberty, when the focus of social interactions shifts from kin to peers. Using the cell birthdate marker bromo-deoxyuridine (BrdU), we previously discovered that more pubertally born cells are added to the rat MePD in males than females. Here we addressed several questions that remained unanswered by our previous work. First, to determine whether there are sex differences in cell proliferation within the MePD, we examined BrdU-immunoreactive (-ir) cells at 2 and 4 h following BrdU administration on postnatal day 30 (P30). The density of BrdU-ir cells was greater in males than in females, indicating greater proliferation in males. Proliferation was substantiated by double-label immunohistochemistry showing that MePD BrdU-ir cells colocalize proliferating cell nuclear antigen, but not the cell death marker Caspase3. We next studied longer time points (2-21 days) following BrdU administration on P30 and found that the rate of cell attrition is higher in males. Finally, triple-label immunohistochemistry of P30-born MePD cells revealed that some of these cells differentiate into neurons or astrocytes within three weeks of cell birth, with no discernable sex differences. The demonstration of pubertal neuro- and glio-genesis in the MePD of male and female rats adds a new dimension to developmental plasticity of the MePD that may contribute to pubertal changes in the perception of social stimuli in both sexes.
Collapse
Affiliation(s)
- Margaret A Mohr
- Neuroscience Program, Michigan State University, East Lansing, MI 48824, USA
| | | | - Lydia L DonCarlos
- Department of Cell and Molecular Physiology, Loyola University Chicago, Stritch School of Medicine, Maywood, IL 60153, USA
| | - Cheryl L Sisk
- Neuroscience Program, Michigan State University, East Lansing, MI 48824, USA.
| |
Collapse
|
6
|
Immature excitatory neurons in the amygdala come of age during puberty. Dev Cogn Neurosci 2022; 56:101133. [PMID: 35841648 PMCID: PMC9289873 DOI: 10.1016/j.dcn.2022.101133] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 06/23/2022] [Accepted: 07/08/2022] [Indexed: 11/21/2022] Open
Abstract
The human amygdala is critical for emotional learning, valence coding, and complex social interactions, all of which mature throughout childhood, puberty, and adolescence. Across these ages, the amygdala paralaminar nucleus (PL) undergoes significant structural changes including increased numbers of mature neurons. The PL contains a large population of immature excitatory neurons at birth, some of which may continue to be born from local progenitors. These progenitors disappear rapidly in infancy, but the immature neurons persist throughout childhood and adolescent ages, indicating that they develop on a protracted timeline. Many of these late-maturing neurons settle locally within the PL, though a small subset appear to migrate into neighboring amygdala subnuclei. Despite its prominent growth during postnatal life and possible contributions to multiple amygdala circuits, the function of the PL remains unknown. PL maturation occurs predominately during late childhood and into puberty when sex hormone levels change. Sex hormones can promote developmental processes such as neuron migration, dendritic outgrowth, and synaptic plasticity, which appear to be ongoing in late-maturing PL neurons. Collectively, we describe how the growth of late-maturing neurons occurs in the right time and place to be relevant for amygdala functions and neuropsychiatric conditions.
Collapse
|
7
|
Chowen JA, Garcia-Segura LM. Role of glial cells in the generation of sex differences in neurodegenerative diseases and brain aging. Mech Ageing Dev 2021; 196:111473. [PMID: 33766745 DOI: 10.1016/j.mad.2021.111473] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 03/14/2021] [Accepted: 03/16/2021] [Indexed: 12/11/2022]
Abstract
Diseases and aging-associated alterations of the nervous system often show sex-specific characteristics. Glial cells play a major role in the endogenous homeostatic response of neural tissue, and sex differences in the glial transcriptome and function have been described. Therefore, the possible role of these cells in the generation of sex differences in pathological alterations of the nervous system is reviewed here. Studies have shown that glia react to pathological insults with sex-specific neuroprotective and regenerative effects. At least three factors determine this sex-specific response of glia: sex chromosome genes, gonadal hormones and neuroactive steroid hormone metabolites. The sex chromosome complement determines differences in the transcriptional responses in glia after brain injury, while gonadal hormones and their metabolites activate sex-specific neuroprotective mechanisms in these cells. Since the sex-specific neuroprotective and regenerative activity of glial cells causes sex differences in the pathological alterations of the nervous system, glia may represent a relevant target for sex-specific therapeutic interventions.
Collapse
Affiliation(s)
- Julie A Chowen
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación la Princesa, Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutriciόn (CIBEROBN), Instituto de Salud Carlos III, and IMDEA Food Institute, CEIUAM+CSIC, Madrid, Spain.
| | - Luis M Garcia-Segura
- Instituto Cajal, Consejo Superior de Investigaciones Científicas (CSIC) and Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, Madrid, Spain.
| |
Collapse
|
8
|
Meeh KL, Rickel CT, Sansano AJ, Shirangi TR. The development of sex differences in the nervous system and behavior of flies, worms, and rodents. Dev Biol 2021; 472:75-84. [PMID: 33484707 DOI: 10.1016/j.ydbio.2021.01.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 01/12/2021] [Accepted: 01/15/2021] [Indexed: 01/14/2023]
Abstract
Understanding how sex differences in innate animal behaviors arise has long fascinated biologists. As a general rule, the potential for sex differences in behavior is built by the developmental actions of sex-specific hormones or regulatory proteins that direct the sexual differentiation of the nervous system. In the last decade, studies in several animal systems have uncovered neural circuit mechanisms underlying discrete sexually dimorphic behaviors. Moreover, how certain hormones and regulatory proteins implement the sexual differentiation of these neural circuits has been illuminated in tremendous detail. Here, we discuss some of these mechanisms with three case-studies-mate recognition in flies, maturation of mating behavior in worms, and play-fighting behavior in young rodents. These studies illustrate general and unique developmental mechanisms to establish sex differences in neuroanatomy and behavior and highlight future challenges for the field.
Collapse
Affiliation(s)
- Kristen L Meeh
- Villanova University, Department of Biology, 800 Lancaster Ave, Villanova, PA, 19085, USA
| | - Clare T Rickel
- Villanova University, Department of Biology, 800 Lancaster Ave, Villanova, PA, 19085, USA
| | - Alexander J Sansano
- Villanova University, Department of Biology, 800 Lancaster Ave, Villanova, PA, 19085, USA
| | - Troy R Shirangi
- Villanova University, Department of Biology, 800 Lancaster Ave, Villanova, PA, 19085, USA.
| |
Collapse
|
9
|
Rurak GM, Woodside B, Aguilar-Valles A, Salmaso N. Astroglial cells as neuroendocrine targets in forebrain development: Implications for sex differences in psychiatric disease. Front Neuroendocrinol 2021; 60:100897. [PMID: 33359797 DOI: 10.1016/j.yfrne.2020.100897] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 12/05/2020] [Accepted: 12/15/2020] [Indexed: 12/23/2022]
Abstract
Astroglial cells are the most abundant cell type in the mammalian brain. They are implicated in almost every aspect of brain physiology, including maintaining homeostasis, building and maintaining the blood brain barrier, and the development and maturation of neuronal networks. Critically, astroglia also express receptors for gonadal sex hormones, respond rapidly to gonadal hormones, and are able to synthesize hormones. Thus, they are positioned to guide and mediate sexual differentiation of the brain, particularly neuronal networks in typical and pathological conditions. In this review, we describe astroglial involvement in the organization and development of the brain, and consider known sex differences in astroglial responses to understand how astroglial cell-mediated organization may play a role in forebrain sexual dimorphisms in human populations. Finally, we consider how sexually dimorphic astroglial responses and functions in development may lead to sex differences in vulnerability for neuropsychiatric disorders.
Collapse
Affiliation(s)
- Gareth M Rurak
- Department of Neuroscience, Carleton University, Ottawa, Ontario, Canada
| | - Barbara Woodside
- Department of Neuroscience, Carleton University, Ottawa, Ontario, Canada; Concordia University, Montreal, Quebec, Canada
| | | | - Natalina Salmaso
- Department of Neuroscience, Carleton University, Ottawa, Ontario, Canada.
| |
Collapse
|
10
|
Tabor J, Wright DK, Christensen J, Zamani A, Collins R, Shultz SR, Mychasiuk R. Examining the Effects of Anabolic-Androgenic Steroids on Repetitive Mild Traumatic Brain Injury (RmTBI) Outcomes in Adolescent Rats. Brain Sci 2020; 10:brainsci10050258. [PMID: 32354109 PMCID: PMC7288073 DOI: 10.3390/brainsci10050258] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 04/20/2020] [Accepted: 04/27/2020] [Indexed: 12/12/2022] Open
Abstract
Background: Repetitive mild traumatic brain injury (RmTBI) is increasingly common in adolescents. Anabolic–androgenic steroid (AAS) consumption among younger professional athletes is a significant risk factor for impaired neurodevelopment. Given the increased rates and overlapping symptomology of RmTBI and AAS use, we sought to investigate the behavioural and neuropathological outcomes associated with the AAS Metandienone (Met) and RmTBI on rats. Methods: Rats received either Met or placebo and were then administered RmTBIs or sham injuries, followed by a behavioural test battery. Post-mortem MRI was conducted to examine markers of brain integrity and qRT-PCR assessed mRNA expression of markers for neurodevelopment, neuroinflammation, stress responses, and repair processes. Results: Although AAS and RmTBI did not produce cumulative deficits, AAS use was associated with detrimental outcomes including changes to depression, aggression, and memory; prefrontal cortex (PFC) atrophy and amygdala (AMYG) enlargement; damaged white matter integrity in the corpus callosum; and altered mRNA expression in the PFC and AMYG. RmTBI affected general activity and contributed to PFC atrophy. Conclusions: Findings corroborate previous results indicating that RmTBI negatively impacts neurodevelopment but also demonstrates that AAS results in significant neuropathological insult to the developing brain.
Collapse
Affiliation(s)
- Jason Tabor
- Department of Psychology, Alberta Children’s Hospital Research Institute, Hotchkiss Brain Institute, University of Calgary, Calgary, AB T2N 1N4, Canada; (J.T.); (J.C.); (R.C.)
| | - David. K. Wright
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne 3004, Australia; (D.K.W.); (A.Z.); (S.R.S.)
| | - Jennaya Christensen
- Department of Psychology, Alberta Children’s Hospital Research Institute, Hotchkiss Brain Institute, University of Calgary, Calgary, AB T2N 1N4, Canada; (J.T.); (J.C.); (R.C.)
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne 3004, Australia; (D.K.W.); (A.Z.); (S.R.S.)
| | - Akram Zamani
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne 3004, Australia; (D.K.W.); (A.Z.); (S.R.S.)
| | - Reid Collins
- Department of Psychology, Alberta Children’s Hospital Research Institute, Hotchkiss Brain Institute, University of Calgary, Calgary, AB T2N 1N4, Canada; (J.T.); (J.C.); (R.C.)
| | - Sandy R. Shultz
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne 3004, Australia; (D.K.W.); (A.Z.); (S.R.S.)
| | - Richelle Mychasiuk
- Department of Psychology, Alberta Children’s Hospital Research Institute, Hotchkiss Brain Institute, University of Calgary, Calgary, AB T2N 1N4, Canada; (J.T.); (J.C.); (R.C.)
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne 3004, Australia; (D.K.W.); (A.Z.); (S.R.S.)
- Correspondence: ; Tel.: +61-3-9903-0897
| |
Collapse
|
11
|
Spielberg JM, Schwarz JM, Matyi MA. Anxiety in transition: Neuroendocrine mechanisms supporting the development of anxiety pathology in adolescence and young adulthood. Front Neuroendocrinol 2019; 55:100791. [PMID: 31542287 PMCID: PMC8265407 DOI: 10.1016/j.yfrne.2019.100791] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Revised: 09/04/2019] [Accepted: 09/18/2019] [Indexed: 11/29/2022]
Abstract
Adolescence marks a key developmental window during which emotion dysregulation increases, along with risk for the onset of anxiety and other affect-related pathologies. Although emotion dysregulation and related pathologies normatively decline during the transition into adulthood, this does not occur for a sizable minority of individuals. Finally, sex differences in anxiety emerge during adolescence, with females developing a 2-fold increase in risk relative to males. Unfortunately, a neurobiological model of the mechanisms that cause these changes during adolescence has yet to be proposed. In the present work, we first provide brief reviews of relevant literature. Next, we outline a dual-mechanism model focused on (i) the influence of pubertal testosterone on key emotion-regulation circuitry (i.e., orbitofrontal cortex-amygdala coupling) and (ii) myelination of the fiber bundles connecting such circuitry (i.e., uncinate fasciculus). The proposed model offers a set of specific, testable hypotheses that will hopefully spur much needed cross-disciplinary research.
Collapse
Affiliation(s)
- Jeffrey M Spielberg
- Department of Psychological and Brain Sciences, University of Delaware, 105 the Green, Newark, DE 19716, United States.
| | - Jaclyn M Schwarz
- Department of Psychological and Brain Sciences, University of Delaware, 105 the Green, Newark, DE 19716, United States.
| | - Melanie A Matyi
- Department of Psychological and Brain Sciences, University of Delaware, 105 the Green, Newark, DE 19716, United States.
| |
Collapse
|
12
|
Bollinger JL, Salinas I, Fender E, Sengelaub DR, Wellman CL. Gonadal hormones differentially regulate sex-specific stress effects on glia in the medial prefrontal cortex. J Neuroendocrinol 2019; 31:e12762. [PMID: 31228875 PMCID: PMC6715499 DOI: 10.1111/jne.12762] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Revised: 05/20/2019] [Accepted: 06/18/2019] [Indexed: 12/21/2022]
Abstract
Women are more susceptible to various stress-linked psychopathologies, including depression. Dysfunction of the medial prefrontal cortex (mPFC) has been implicated in depression, and studies indicate sex differences in stress effects on mPFC structure and function. For example, chronic stress induces dendritic atrophy in the mPFC in male rats, yet dendritic growth in females. Recent findings suggest glial pathways toward depression. Glia are highly responsive to neuronal activity and function as critical regulators of synaptic plasticity. Preclinical models demonstrate stress-induced microglial activation in mPFC in males, yet deactivation in females. By contrast, stress reduces astrocyte complexity in mPFC in male rats, whereas the effects in females are unknown. Glia possess receptors for most gonadal hormones and gonadal hormones are known to modulate neuronal activity. Thus, gonadal hormones represent a potential mechanism underlying sex differences in glia, as well as divergent stress effects. Therefore, we examined the role of gonadal hormones in sex-specific stress effects on neuronal activity (ie FosB/ ΔFosB induction) and glia in the mPFC. The findings obtained indicate greater microglial activation in mPFC in females and a greater astrocyte area in males. Basal astrocyte morphology is modulated by androgens, whereas androgens or oestrogens dampen the microglial state in males. Astrocyte morphology is associated with neuronal activity in both sexes, regardless of hormonal condition. Chronic stress induced astrocytic atrophy in males, yet hypertrophy in females, with gonadal hormones partly regulating this difference. Stress effects on microglia are oestradiol-dependent in females. Taken together, these data suggest sex-specific, gonadal hormone-dependent stress effects on astrocytes and microglia in the mPFC.
Collapse
Affiliation(s)
- Justin L Bollinger
- Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN, USA
- Program in Neuroscience, Indiana University, Bloomington, IN, USA
- Center for the Integrative Study of Animal Behavior, Indiana University, Bloomington, IN, USA
| | - Isabella Salinas
- Center for the Integrative Study of Animal Behavior, Indiana University, Bloomington, IN, USA
| | - Emily Fender
- Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN, USA
| | - Dale R Sengelaub
- Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN, USA
- Program in Neuroscience, Indiana University, Bloomington, IN, USA
- Center for the Integrative Study of Animal Behavior, Indiana University, Bloomington, IN, USA
| | - Cara L Wellman
- Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN, USA
- Program in Neuroscience, Indiana University, Bloomington, IN, USA
- Center for the Integrative Study of Animal Behavior, Indiana University, Bloomington, IN, USA
| |
Collapse
|
13
|
Sadrtdinova II, Khizmatullina ZR. [Reactive changes in morphological and morphometric parameters of immunopositive astrocytes of the amygdala in response to hormone level in absence epilepsy]. Zh Nevrol Psikhiatr Im S S Korsakova 2019; 118:61-66. [PMID: 30698546 DOI: 10.17116/jnevro201811810261] [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] [Indexed: 01/01/2023]
Abstract
AIM To study the changes in the morphological and morphometric parameters of immunopositive astrocytes of the amygdala in absence epilepsy depending on hormonal profile. MATERIAL AND METHODS Adult female WAG/Rij rats were used as experimental subjects. The astrocytes were detected on serial sections using a reaction to glial fibrillary acidic protein (GFAP) with pre-stained hematoxylin. Quantitative analysis was carried out for a 204.8´153.6 μm2 field of view. RESULTS In the control group, astrocytes had a relatively regular stellate form and GFAP was moderately expressed in their bodies and processes. The number of astrocytes was 18.20±2.87, and their area was 164±3.29 μm2. After ovariectomy, a high expression of the protein, both in the bodies and in the processes of astrocytes, increasing the cell size to 188.85±4.97 μm2 (p<0.05) was observed. The astrocytes increased to 34.55±3.03 (p<0.05). In addition, the deformation of the processes and their diffuse defibration were observed. After hormone replacement therapy, a decrease in GFAP expression was detected, the area of astrocytes became smaller in comparison with the group after ovariectomy: 173.54±5.48 μm2 (p<0.05). Morphological changes in glial cells were manifested as a decrease in the size of their bodies, the processes became smooth without diffuse sprouting and swelling, which is probably associated with neuroprotective functions of estradiol. CONCLUSION Thus, the results of our study demonstrated that the deficiency of female sex hormones led to the increase in both the amount and area of astrocytes in the anterior cortical nucleus of the amygdala, and hormone replacement therapy positively affected the structural and quantitative characteristics of astrocytes due to the endogenous protective role of estradiol.
Collapse
|
14
|
Kaufman MJ, Kanayama G, Hudson JI, Pope HG. Supraphysiologic-dose anabolic-androgenic steroid use: A risk factor for dementia? Neurosci Biobehav Rev 2019; 100:180-207. [PMID: 30817935 PMCID: PMC6451684 DOI: 10.1016/j.neubiorev.2019.02.014] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Revised: 02/13/2019] [Accepted: 02/17/2019] [Indexed: 02/06/2023]
Abstract
Supraphysiologic-dose anabolic-androgenic steroid (AAS) use is associated with physiologic, cognitive, and brain abnormalities similar to those found in people at risk for developing Alzheimer's Disease and its related dementias (AD/ADRD), which are associated with high brain β-amyloid (Aβ) and hyperphosphorylated tau (tau-P) protein levels. Supraphysiologic-dose AAS induces androgen abnormalities and excess oxidative stress, which have been linked to increased and decreased expression or activity of proteins that synthesize and eliminate, respectively, Aβ and tau-P. Aβ and tau-P accumulation may begin soon after initiating supraphysiologic-dose AAS use, which typically occurs in the early 20s, and their accumulation may be accelerated by other psychoactive substance use, which is common among non-medical AAS users. Accordingly, the widespread use of supraphysiologic-dose AAS may increase the numbers of people who develop dementia. Early diagnosis and correction of sex-steroid level abnormalities and excess oxidative stress could attenuate risk for developing AD/ADRD in supraphysiologic-dose AAS users, in people with other substance use disorders, and in people with low sex-steroid levels or excess oxidative stress associated with aging.
Collapse
Affiliation(s)
- Marc J Kaufman
- McLean Imaging Center, McLean Hospital, 115 Mill St., Belmont, MA 02478, USA; Department of Psychiatry, Harvard Medical School, Boston, MA 02115, USA.
| | - Gen Kanayama
- Biological Psychiatry Laboratory, McLean Hospital, 115 Mill St., Belmont, MA 02478, USA; Department of Psychiatry, Harvard Medical School, Boston, MA 02115, USA
| | - James I Hudson
- Biological Psychiatry Laboratory, McLean Hospital, 115 Mill St., Belmont, MA 02478, USA; Department of Psychiatry, Harvard Medical School, Boston, MA 02115, USA
| | - Harrison G Pope
- Biological Psychiatry Laboratory, McLean Hospital, 115 Mill St., Belmont, MA 02478, USA; Department of Psychiatry, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
15
|
Wellman CL, Bangasser DA, Bollinger JL, Coutellier L, Logrip ML, Moench KM, Urban KR. Sex Differences in Risk and Resilience: Stress Effects on the Neural Substrates of Emotion and Motivation. J Neurosci 2018; 38:9423-9432. [PMID: 30381434 PMCID: PMC6209838 DOI: 10.1523/jneurosci.1673-18.2018] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Revised: 09/20/2018] [Accepted: 09/22/2018] [Indexed: 02/06/2023] Open
Abstract
Risk for stress-sensitive psychopathologies differs in men and women, yet little is known about sex-dependent effects of stress on cellular structure and function in corticolimbic regions implicated in these disorders. Determining how stress influences these regions in males and females will deepen our understanding of the mechanisms underlying sex-biased psychopathology. Here, we discuss sex differences in CRF regulation of arousal and cognition, glucocorticoid modulation of amygdalar physiology and alcohol consumption, the age-dependent impact of social stress on prefrontal pyramidal cell excitability, stress effects on the prefrontal parvalbumin system in relation to emotional behaviors, contributions of stress and gonadal hormones to stress effects on prefrontal glia, and alterations in corticolimbic structure and function after cessation of chronic stress. These studies demonstrate that, while sex differences in stress effects may be nuanced, nonuniform, and nonlinear, investigations of these differences are nonetheless critical for developing effective, sex-specific treatments for psychological disorders.
Collapse
Affiliation(s)
- Cara L Wellman
- Department of Psychological and Brain Sciences, Program in Neuroscience, and Center for the Integrative Study of Animal Behavior, Indiana University, Bloomington, Indiana 47405,
| | - Debra A Bangasser
- Psychology Department and Neuroscience Program, Temple University, Philadelphia, Pennsylvania 19122
| | - Justin L Bollinger
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, Ohio 45237
| | - Laurence Coutellier
- Departments of Psychology and Neuroscience, Ohio State University, Columbus, Ohio 43210
| | - Marian L Logrip
- Department of Psychology, Indiana University-Purdue University Indianapolis, Indianapolis, Indiana 46202, and
| | - Kelly M Moench
- Department of Psychological and Brain Sciences, Program in Neuroscience, and Center for the Integrative Study of Animal Behavior, Indiana University, Bloomington, Indiana 47405
| | - Kimberly R Urban
- Children's Hospital of Philadelphia, Abramson Research Center, Philadelphia, Pennsylvania 19104
| |
Collapse
|
16
|
Keller D, Erö C, Markram H. Cell Densities in the Mouse Brain: A Systematic Review. Front Neuroanat 2018; 12:83. [PMID: 30405363 PMCID: PMC6205984 DOI: 10.3389/fnana.2018.00083] [Citation(s) in RCA: 238] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Accepted: 09/20/2018] [Indexed: 11/29/2022] Open
Abstract
The mouse brain is the most extensively studied brain of all species. We performed an exhaustive review of the literature to establish our current state of knowledge on cell numbers in mouse brain regions, arguably the most fundamental property to measure when attempting to understand a brain. The synthesized information, collected in one place, can be used by both theorists and experimentalists. Although for commonly-studied regions cell densities could be obtained for principal cell types, overall we know very little about how many cells are present in most brain regions and even less about cell-type specific densities. There is also substantial variation in cell density values obtained from different sources. This suggests that we need a new approach to obtain cell density datasets for the mouse brain.
Collapse
Affiliation(s)
- Daniel Keller
- Blue Brain Project, École Polytechnique Fédérale de Lausanne, Geneva, Switzerland
| | | | | |
Collapse
|
17
|
Schümann D, Sommer T. Dissociable contributions of the amygdala to the immediate and delayed effects of emotional arousal on memory. ACTA ACUST UNITED AC 2018; 25:283-293. [PMID: 29764974 PMCID: PMC5959227 DOI: 10.1101/lm.047282.117] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Accepted: 04/04/2018] [Indexed: 12/15/2022]
Abstract
Emotional arousal enhances memory encoding and consolidation leading to better immediate and delayed memory. Although the central noradrenergic system and the amygdala play critical roles in both effects of emotional arousal, we have recently shown that these effects are at least partly independent of each other, suggesting distinct underlying neural mechanisms. Here we aim to dissociate the neural substrates of both effects in 70 female participants using an emotional memory paradigm to investigate how neural activity, as measured by fMRI, and a polymorphism in the α2B-noradrenoceptor vary for these effects. To also test whether the immediate and delayed effects of emotional arousal on memory are stable traits, we invited back participants who were a part of a large-scale behavioral memory study ∼3.5 yr ago. We replicated the low correlation of the immediate and delayed emotional enhancement of memory across participants (r = 0.16) and observed, moreover, that only the delayed effect was, to some degree, stable over time (r = 0.23). Bilateral amygdala activity, as well as its coupling with the visual cortex and the fusiform gyrus, was related to the preferential encoding of emotional stimuli, which is consistent with affect-biased attention. Moreover, the adrenoceptor genotype modulated the bilateral amygdala activity associated with this effect. The left amygdala and its coupling with the hippocampus was specifically associated with the more efficient consolidation of emotional stimuli, which is consistent with amygdalar modulation of hippocampal consolidation.
Collapse
Affiliation(s)
- Dirk Schümann
- Institute for Systems Neuroscience, Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Tobias Sommer
- Institute for Systems Neuroscience, Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
18
|
Arambula SE, Jima D, Patisaul HB. Prenatal bisphenol A (BPA) exposure alters the transcriptome of the neonate rat amygdala in a sex-specific manner: a CLARITY-BPA consortium study. Neurotoxicology 2017; 65:207-220. [PMID: 29097150 DOI: 10.1016/j.neuro.2017.10.005] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Revised: 10/23/2017] [Accepted: 10/24/2017] [Indexed: 12/11/2022]
Abstract
Bisphenol A (BPA) is a widely recognized endocrine disruptor prevalent in many household items. Because experimental and epidemiological data suggest links between prenatal BPA exposure and altered affective behaviors in children, even at levels below the current US FDA No Observed Adverse Effect Level (NOAEL) of 5mg/kg body weight (bw)/day, there is concern that early life exposure may alter neurodevelopment. The current study was conducted as part of the CLARITY-BPA (Consortium Linking Academic and Regulatory Insights on BPA Toxicity) program and examined the full amygdalar transcriptome on postnatal day (PND) 1, with the hypothesis that prenatal BPA exposure would alter the expression of genes and pathways fundamental to sex-specific affective behaviors. NCTR Sprague-Dawley dams were gavaged from gestational day 6 until parturition with BPA (2.5, 25, 250, 2500, or 25000μg/kg bw/day), a reference estrogen (0.05 or 0.5μg ethinyl estradiol (EE2)/kg bw/day), or vehicle. PND 1 amygdalae were microdissected and gene expression was assessed with qRT-PCR (all exposure groups) and RNAseq (vehicle, 25 and 250μg BPA, and 0.5μg EE2 groups only). Our results demonstrate that that prenatal BPA exposure can disrupt the transcriptome of the neonate amygdala, at doses below the FDA NOAEL, in a sex-specific manner and indicate that the female amygdala may be more sensitive to BPA exposure during fetal development. We also provide additional evidence that developmental BPA exposure can interfere with estrogen, oxytocin, and vasopressin signaling pathways in the developing brain and alter signaling pathways critical for synaptic organization and transmission.
Collapse
Affiliation(s)
- Sheryl E Arambula
- Department of Biological Sciences, North Carolina State University, Raleigh, NC 27695, USA; WM Keck Center for Behavioral Biology, North Carolina State University, Raleigh, NC 27695, USA
| | - Dereje Jima
- Center for Human Health and the Environment, North Carolina State University, Raleigh, NC 27695, USA; Bioinformatics Research Center, North Carolina State University, Raleigh, NC 27695
| | - Heather B Patisaul
- Department of Biological Sciences, North Carolina State University, Raleigh, NC 27695, USA; WM Keck Center for Behavioral Biology, North Carolina State University, Raleigh, NC 27695, USA; Center for Human Health and the Environment, North Carolina State University, Raleigh, NC 27695, USA.
| |
Collapse
|
19
|
Low KL, Ma C, Soma KK. Tyramide Signal Amplification Permits Immunohistochemical Analyses of Androgen Receptors in the Rat Prefrontal Cortex. J Histochem Cytochem 2017; 65:295-308. [PMID: 28438093 PMCID: PMC5407533 DOI: 10.1369/0022155417694870] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2016] [Accepted: 01/24/2017] [Indexed: 11/22/2022] Open
Abstract
Research on neural androgen receptors (ARs) has traditionally focused on brain regions that regulate reproductive and aggressive behaviors, such as the hypothalamus and amygdala. Although many cells in the prefrontal cortex (PFC) also express ARs, the number of ARs per cell appears to be much lower, and thus, AR immunostaining is often hard to detect and quantify in the PFC. Here, we demonstrate that biotin tyramide signal amplification (TSA) dramatically increases AR immunoreactivity in the rat brain, including critical regions of the PFC such as the medial PFC (mPFC) and orbitofrontal cortex (OFC). We show that TSA is useful for AR detection with both chromogenic and immunofluorescent immunohistochemistry. Double-labeling studies reveal that AR+ cells in the PFC and hippocampus are NeuN+ but not GFAP+ and thus primarily neuronal. Finally, in gonadally intact rats, more AR+ cells are present in the mPFC and OFC of males than of females. Future studies can use TSA to further examine AR immunoreactivity across ages, sexes, strains, and different procedures (e.g., fixation methods). In light of emerging evidence for the androgen regulation of executive function and working memory, these results may help understand the distribution and roles of ARs in the PFC.
Collapse
Affiliation(s)
- Katelyn L. Low
- Department of Psychology and The Djavad Mowafaghian Centre for Brain Health (KLL, CM, KKS), The University of British Columbia, Vancouver, British Columbia, Canada
- Department of Zoology (KLL, KKS), The University of British Columbia, Vancouver, British Columbia, Canada
| | - Chunqi Ma
- Department of Psychology and The Djavad Mowafaghian Centre for Brain Health (KLL, CM, KKS), The University of British Columbia, Vancouver, British Columbia, Canada
- Department of Zoology (KLL, KKS), The University of British Columbia, Vancouver, British Columbia, Canada
| | - Kiran K. Soma
- Kiran K. Soma, Department of Psychology, University of British Columbia, 2136 West Mall, Vancouver, British Columbia, Canada V6T 1Z4.E-mail:
| |
Collapse
|
20
|
Diniz DG, de Oliveira MA, de Lima CM, Fôro CAR, Sosthenes MCK, Bento-Torres J, da Costa Vasconcelos PF, Anthony DC, Diniz CWP. Age, environment, object recognition and morphological diversity of GFAP-immunolabeled astrocytes. Behav Brain Funct 2016; 12:28. [PMID: 27719674 PMCID: PMC5056502 DOI: 10.1186/s12993-016-0111-2] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2016] [Accepted: 09/22/2016] [Indexed: 12/12/2022] Open
Abstract
Background Few studies have explored the glial response to a standard environment and how the response may be associated with age-related cognitive decline in learning and memory. Here we investigated aging and environmental influences on hippocampal-dependent tasks and on the morphology of an unbiased selected population of astrocytes from the molecular layer of dentate gyrus, which is the main target of perforant pathway. Results Six and twenty-month-old female, albino Swiss mice were housed, from weaning, in a standard or enriched environment, including running wheels for exercise and tested for object recognition and contextual memories. Young adult and aged subjects, independent of environment, were able to distinguish familiar from novel objects. All experimental groups, except aged mice from standard environment, distinguish stationary from displaced objects. Young adult but not aged mice, independent of environment, were able to distinguish older from recent objects. Only young mice from an enriched environment were able to distinguish novel from familiar contexts. Unbiased selected astrocytes from the molecular layer of the dentate gyrus were reconstructed in three-dimensions and classified using hierarchical cluster analysis of bimodal or multimodal morphological features. We found two morphological phenotypes of astrocytes and we designated type I the astrocytes that exhibited significantly higher values of morphological complexity as compared with type II. Complexity = [Sum of the terminal orders + Number of terminals] × [Total branch length/Number of primary branches]. On average, type I morphological complexity seems to be much more sensitive to age and environmental influences than that of type II. Indeed, aging and environmental impoverishment interact and reduce the morphological complexity of type I astrocytes at a point that they could not be distinguished anymore from type II. Conclusions We suggest these two types of astrocytes may have different physiological roles and that the detrimental effects of aging on memory in mice from a standard environment may be associated with a reduction of astrocytes morphological diversity. Electronic supplementary material The online version of this article (doi:10.1186/s12993-016-0111-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Daniel Guerreiro Diniz
- Laboratório de Investigações Em Neurodegeneração e Infecção, Instituto de Ciências Biológicas, Universidade Federal do Pará, Hospital Universitário João de Barros Barreto, Rua dos Mundurucus 4487, Guamá, Belém, Pará, CEP 66073-000, Brazil.,Laboratory of Experimental Neuropathology, Department of Pharmacology, University of Oxford, Oxford, England, UK
| | - Marcus Augusto de Oliveira
- Laboratório de Investigações Em Neurodegeneração e Infecção, Instituto de Ciências Biológicas, Universidade Federal do Pará, Hospital Universitário João de Barros Barreto, Rua dos Mundurucus 4487, Guamá, Belém, Pará, CEP 66073-000, Brazil
| | - Camila Mendes de Lima
- Laboratório de Investigações Em Neurodegeneração e Infecção, Instituto de Ciências Biológicas, Universidade Federal do Pará, Hospital Universitário João de Barros Barreto, Rua dos Mundurucus 4487, Guamá, Belém, Pará, CEP 66073-000, Brazil
| | - César Augusto Raiol Fôro
- Laboratório de Investigações Em Neurodegeneração e Infecção, Instituto de Ciências Biológicas, Universidade Federal do Pará, Hospital Universitário João de Barros Barreto, Rua dos Mundurucus 4487, Guamá, Belém, Pará, CEP 66073-000, Brazil
| | - Marcia Consentino Kronka Sosthenes
- Laboratório de Investigações Em Neurodegeneração e Infecção, Instituto de Ciências Biológicas, Universidade Federal do Pará, Hospital Universitário João de Barros Barreto, Rua dos Mundurucus 4487, Guamá, Belém, Pará, CEP 66073-000, Brazil
| | - João Bento-Torres
- Laboratório de Investigações Em Neurodegeneração e Infecção, Instituto de Ciências Biológicas, Universidade Federal do Pará, Hospital Universitário João de Barros Barreto, Rua dos Mundurucus 4487, Guamá, Belém, Pará, CEP 66073-000, Brazil
| | | | - Daniel Clive Anthony
- Laboratory of Experimental Neuropathology, Department of Pharmacology, University of Oxford, Oxford, England, UK
| | - Cristovam Wanderley Picanço Diniz
- Laboratório de Investigações Em Neurodegeneração e Infecção, Instituto de Ciências Biológicas, Universidade Federal do Pará, Hospital Universitário João de Barros Barreto, Rua dos Mundurucus 4487, Guamá, Belém, Pará, CEP 66073-000, Brazil. .,Laboratory of Experimental Neuropathology, Department of Pharmacology, University of Oxford, Oxford, England, UK.
| |
Collapse
|
21
|
Acaz-Fonseca E, Avila-Rodriguez M, Garcia-Segura LM, Barreto GE. Regulation of astroglia by gonadal steroid hormones under physiological and pathological conditions. Prog Neurobiol 2016; 144:5-26. [DOI: 10.1016/j.pneurobio.2016.06.002] [Citation(s) in RCA: 81] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Accepted: 06/05/2016] [Indexed: 01/07/2023]
|
22
|
Schulz KM, Sisk CL. The organizing actions of adolescent gonadal steroid hormones on brain and behavioral development. Neurosci Biobehav Rev 2016; 70:148-158. [PMID: 27497718 DOI: 10.1016/j.neubiorev.2016.07.036] [Citation(s) in RCA: 196] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Revised: 07/13/2016] [Accepted: 07/14/2016] [Indexed: 01/03/2023]
Abstract
Adolescence is a developmental period characterized by dramatic changes in cognition, risk-taking and social behavior. Although gonadal steroid hormones are well-known mediators of these behaviors in adulthood, the role gonadal steroid hormones play in shaping the adolescent brain and behavioral development has only come to light in recent years. Here we discuss the sex-specific impact of gonadal steroid hormones on the developing adolescent brain. Indeed, the effects of gonadal steroid hormones during adolescence on brain structure and behavioral outcomes differs markedly between the sexes. Research findings suggest that adolescence, like the perinatal period, is a sensitive period for the sex-specific effects of gonadal steroid hormones on brain and behavioral development. Furthermore, evidence from studies on male sexual behavior suggests that adolescence is part of a protracted postnatal sensitive period that begins perinatally and ends following adolescence. As such, the perinatal and peripubertal periods of brain and behavioral organization likely do not represent two discrete sensitive periods, but instead are the consequence of normative developmental timing of gonadal hormone secretions in males and females.
Collapse
Affiliation(s)
- Kalynn M Schulz
- Department of Psychology, University of Tennessee, Knoxville, TN 37996, United States.
| | - Cheryl L Sisk
- Neuroscience Program, Michigan State University, East Lansing, MI 48824, United States.
| |
Collapse
|
23
|
Pfau DR, Hobbs NJ, Breedlove SM, Jordan CL. Sex and laterality differences in medial amygdala neurons and astrocytes of adult mice. J Comp Neurol 2016; 524:2492-502. [PMID: 26780286 DOI: 10.1002/cne.23964] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2015] [Revised: 01/03/2016] [Accepted: 01/13/2016] [Indexed: 01/16/2023]
Abstract
The posterodorsal aspect of the medial amygdala (MePD) in rats is sexually dimorphic, being larger and containing more and larger neurons in males than in females. It is also highly lateralized, with the right MePD larger than the left in both sexes, but with the smaller left MePD actually containing more and larger neurons than the larger right. Astrocytes are also strikingly sexually differentiated, with male-biased numbers and lateralized favoring the right in the rat MePD. However, comparable information is scant for mice where genetic tools offer greater experimental power. Hence, we examined the MePD from adult male and female C57Bl/6(J) mice. We now report that the MePD is larger in males than in females, with the MePD in males containing more astrocytes and neurons than in females. However, we did not find sex differences in astrocyte complexity or overall glial number nor effects of laterality in either measure. While the mouse MePD is generally less lateralized than in rats, we did find that the sex difference in astrocyte number is only on the right because of a significant lateralization in females, with significantly fewer astrocytes on the right than the left but only in females. A sex difference in neuronal soma size favoring males was also evident, but only on the left. Sex differences in the number of neurons and astrocytes common to both rodent species may represent core morphological features that critically underlie the expression of sex-specific behaviors that depend on the MePD. J. Comp. Neurol. 524:2492-2502, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Daniel R Pfau
- Neuroscience Program, Michigan State University, East Lansing, Michigan
| | - Nicholas J Hobbs
- Neuroscience Program, Michigan State University, East Lansing, Michigan
| | - S Marc Breedlove
- Neuroscience Program, Michigan State University, East Lansing, Michigan
| | - Cynthia L Jordan
- Neuroscience Program, Michigan State University, East Lansing, Michigan
| |
Collapse
|
24
|
Kaufman MJ, Janes AC, Hudson JI, Brennan BP, Kanayama G, Kerrigan AR, Jensen JE, Pope HG. Brain and cognition abnormalities in long-term anabolic-androgenic steroid users. Drug Alcohol Depend 2015; 152:47-56. [PMID: 25986964 PMCID: PMC4458166 DOI: 10.1016/j.drugalcdep.2015.04.023] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Revised: 04/21/2015] [Accepted: 04/22/2015] [Indexed: 11/30/2022]
Abstract
BACKGROUND Anabolic-androgenic steroid (AAS) use is associated with psychiatric symptoms including increased aggression as well as with cognitive dysfunction. The brain effects of long-term AAS use have not been assessed in humans. METHODS This multimodal magnetic resonance imaging study of the brain compared 10 male weightlifters reporting long-term AAS use with 10 age-matched weightlifters reporting no AAS exposure. Participants were administered visuospatial memory tests and underwent neuroimaging. Brain volumetric analyses were performed; resting-state fMRI functional connectivity (rsFC) was evaluated using a region-of-interest analysis focused on the amygdala; and dorsal anterior cingulate cortex (dACC) metabolites were quantified by proton magnetic resonance spectroscopy (MRS). RESULTS AAS users had larger right amygdala volumes than nonusers (P=0.002) and reduced rsFC between right amygdala and frontal, striatal, limbic, hippocampal, and visual cortical areas. Left amygdala volumes were slightly larger in AAS users (P=0.061) but few group differences were detected in left amygdala rsFC. AAS users also had lower dACC scyllo-inositol levels (P=0.004) and higher glutamine/glutamate ratios (P=0.028), possibly reflecting increased glutamate turnover. On a visuospatial cognitive task, AAS users performed more poorly than nonusers, with the difference approaching significance (P=0.053). CONCLUSIONS Long-term AAS use is associated with right amygdala enlargement and reduced right amygdala rsFC with brain areas involved in cognitive control and spatial memory, which could contribute to the psychiatric effects and cognitive dysfunction associated with AAS use. The MRS abnormalities we detected could reflect enhanced glutamate turnover and increased vulnerability to neurotoxic or neurodegenerative processes, which could contribute to AAS-associated cognitive dysfunction.
Collapse
Affiliation(s)
- Marc J. Kaufman
- McLean Imaging Center, McLean Hospital, and the Department of Psychiatry, Harvard Medical School, 115 Mill St., Belmont, Massachusetts, 02478, USA
| | - Amy C. Janes
- McLean Imaging Center, McLean Hospital, and the Department of Psychiatry, Harvard Medical School, 115 Mill St., Belmont, Massachusetts, 02478, USA
| | - James I. Hudson
- Biological Psychiatry Laboratory, McLean Hospital, and the Department of Psychiatry, Harvard Medical School, 115 Mill St., Belmont, Massachusetts, 02478, USA
| | - Brian P. Brennan
- Biological Psychiatry Laboratory, McLean Hospital, and the Department of Psychiatry, Harvard Medical School, 115 Mill St., Belmont, Massachusetts, 02478, USA
| | - Gen Kanayama
- Biological Psychiatry Laboratory, McLean Hospital, and the Department of Psychiatry, Harvard Medical School, 115 Mill St., Belmont, Massachusetts, 02478, USA
| | - Andrew R. Kerrigan
- McLean Imaging Center, McLean Hospital, and the Department of Psychiatry, Harvard Medical School, 115 Mill St., Belmont, Massachusetts, 02478, USA
| | - J. Eric Jensen
- McLean Imaging Center, McLean Hospital, and the Department of Psychiatry, Harvard Medical School, 115 Mill St., Belmont, Massachusetts, 02478, USA
| | - Harrison G. Pope
- Biological Psychiatry Laboratory, McLean Hospital, and the Department of Psychiatry, Harvard Medical School, 115 Mill St., Belmont, Massachusetts, 02478, USA
| |
Collapse
|
25
|
Renier KJ, Troxell-Smith SM, Johansen JA, Katsuno M, Adachi H, Sobue G, Chua JP, Sun Kim H, Lieberman AP, Breedlove SM, Jordan CL. Antiandrogen flutamide protects male mice from androgen-dependent toxicity in three models of spinal bulbar muscular atrophy. Endocrinology 2014; 155:2624-34. [PMID: 24742193 PMCID: PMC4060177 DOI: 10.1210/en.2013-1756] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Spinal and bulbar muscular atrophy (SBMA) is a late-onset, progressive neurodegenerative disease linked to a polyglutamine (polyQ) expansion in the androgen receptor (AR). Men affected by SBMA show marked muscle weakness and atrophy, typically emerging midlife. Given the androgen-dependent nature of this disease, one might expect AR antagonists to have therapeutic value for treating SBMA. However, current work from animal models suggests otherwise, raising questions about whether polyQ-expanded AR exerts androgen-dependent toxicity through mechanisms distinct from normal AR function. In this study, we asked whether the nonsteroidal AR antagonist flutamide, delivered via a time-release pellet, could reverse or prevent androgen-dependent AR toxicity in three different mouse models of SBMA: the AR97Q transgenic (Tg) model, a knock-in (KI) model, and a myogenic Tg model. We find that flutamide protects mice from androgen-dependent AR toxicity in all three SBMA models, preventing or reversing motor dysfunction in the Tg models and significantly extending the life span in KI males. Given that flutamide effectively protects against androgen-dependent disease in three different mouse models of SBMA, our data are proof of principle that AR antagonists have therapeutic potential for treating SBMA in humans and support the notion that toxicity caused by polyQ-expanded AR uses at least some of the same mechanisms as normal AR before diverging to produce disease and muscle atrophy.
Collapse
Affiliation(s)
- Kayla J Renier
- Neuroscience Program (K.J.R., S.M.T.-S., S.M.B., C.L.J.), Michigan State University, E Lansing, Michigan 48824-1101; College of Medicine (J.A.J.), Central Michigan University, Mt Pleasant Michigan 48859; Department of Neurology (M.K., H.A., G.S.), Nagoya University Graduate School of Medicine, Nagoya, Japan 466-8550; and Department of Pathology (J.P.C., H.S.K., A.P.L.), University of Michigan, Ann Arbor, Michigan 48109
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
MeCP2 regulates GFAP expression within the developing brain. Brain Res 2013; 1543:151-8. [PMID: 24269336 DOI: 10.1016/j.brainres.2013.11.011] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2013] [Revised: 11/06/2013] [Accepted: 11/07/2013] [Indexed: 11/22/2022]
Abstract
Mutations in MECP2 cause Rett syndrome (RTT), an X-linked neurodevelopmental disorder that primarily affects females. Individuals with RTT have increased glial fibrillary acidic protein (GFAP) expression in the brain. GFAP is an intermediate filament protein that is expressed predominately within astrocytes in the CNS. MeCP2 binds to methylated regions of the GFAP promoter region and suppresses GFAP expression in vitro. Therefore, we wanted to determine if transiently reducing MeCP2 expression would increase GFAP expression in the developing rat brain. Male and female rats received infusions of either MeCP2 or control siRNA targeting the amygdala during the first 3 days of postnatal life. Brains were collected after 6h or 2 weeks following the last infusion. MeCP2 siRNA increased GFAP mRNA and protein within the female, but not the male, amygdala on postnatal day (PN) 2. Two weeks following the infusion, levels returned to normal. MeCP2 siRNA targeting the hypothalamus also increases GFAP mRNA within the female hypothalamus on PN2, suggesting that the regulation is not brain region-specific. It appears that MeCP2 does not regulate all astrocyte markers in the developing female brain, but specifically regulates GFAP expression, as levels of S100β and vimentin were not altered in the female amygdala at either time point. These data contribute to the idea that the role of MeCP2 differs in the developing male versus female brain. Further elucidating the regulation and function of GFAP can contribute to our understanding of MeCP2 function and perhaps RTT etiology.
Collapse
|
27
|
Johnson RT, Breedlove SM, Jordan CL. Androgen receptors mediate masculinization of astrocytes in the rat posterodorsal medial amygdala during puberty. J Comp Neurol 2013; 521:2298-309. [PMID: 23239016 DOI: 10.1002/cne.23286] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2012] [Revised: 12/03/2012] [Accepted: 12/11/2012] [Indexed: 11/07/2022]
Abstract
Astrocytes in the posterodorsal portion of the medial amygdala (MePD) are sexually dimorphic in adult rats: males have more astrocytes in the right MePD and more elaborate processes in the left MePD than do females. Functional androgen receptors (ARs) are required for masculinization of MePD astrocytes, as these measures are demasculinized in adult males carrying the testicular feminization mutation (Tfm) of the AR gene, which renders AR dysfunctional. We now report that the number of astrocytes is already sexually dimorphic in the right MePD of juvenile 25-day-old (P25) rats. Because Tfm males have as many astrocytes as wild-type males at this age, this prepubertal sexual dimorphism is independent of ARs. After P25, astrocyte number increases in the MePD of all groups, but activation of ARs augments this increase in the right MePD, where more astrocytes are added in males than in Tfm males. Consequently, by adulthood, females and Tfm males have equivalent numbers of astrocytes in the right MePD. Sexual dimorphism in astrocyte arbor complexity in the left MePD arises after P25, and is entirely AR-dependent. Thus, masculinization of MePD astrocytes is a result of both AR-independent processes before the juvenile period and AR-dependent processes afterward.
Collapse
Affiliation(s)
- Ryan T Johnson
- Neuroscience Program, Michigan State University, East Lansing, Michigan 48824-1101, USA.
| | | | | |
Collapse
|
28
|
Burke NN, Llorente R, Marco EM, Tong K, Finn DP, Viveros MP, Roche M. Maternal deprivation is associated with sex-dependent alterations in nociceptive behavior and neuroinflammatory mediators in the rat following peripheral nerve injury. THE JOURNAL OF PAIN 2013; 14:1173-84. [PMID: 23850096 DOI: 10.1016/j.jpain.2013.05.003] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 03/09/2013] [Revised: 04/24/2013] [Accepted: 05/02/2013] [Indexed: 12/30/2022]
Abstract
UNLABELLED Early-life stress is associated with an increased risk of developing affective disorders and chronic pain conditions. This study examined the effect of maternal deprivation (MD) on nociceptive responding prior to and following peripheral nerve injury (L5-L6 spinal nerve ligation [SNL]). Because neuroimmune signaling plays an important role in pain and affective disorders, associated alterations in glial and cytokine expression were assessed in key brain regions associated with emotional and nociceptive responding, the hippocampus and prefrontal cortex. MD female, but not male, rats exhibited thermal hypoalgesia and mechanical allodynia compared with control (non-MD) counterparts. SNL resulted in mechanical and cold allodynia in MD and control rats of both sexes. However, MD females exhibited enhanced SNL-induced allodynic responding compared with non-MD counterparts. Interleukin 6 (IL-6) expression was reduced in the prefrontal cortex of MD-SNL males when compared with non-SNL counterparts. Glial fibrillary acidic protein and IL-1β expression in the hippocampus of MD-SNL males was increased compared with non-MD controls. MD-SNL females exhibited reduced tumor necrosis factor alpha in the prefrontal cortex with a concomitant increase in IL-6 and tumor necrosis factor alpha expression in the hippocampus, compared with either MD or SNL alone. In conclusion, MD female, but not male, rats exhibit enhanced nociceptive responding following peripheral nerve injury, effects that may relate to the distinct neuroinflammatory profile observed in female versus male rats. PERSPECTIVE This study demonstrates that females rats exposed to early-life stress exhibit enhanced neuropathic pain responding, effects that are associated with alterations in neuroinflammatory mediators. Increased understanding of the interactions among early-life stress, gender, and pain may lead to the identification of novel therapeutic targets for the treatment of chronic pain disorders.
Collapse
Affiliation(s)
- Nikita N Burke
- Physiology, School of Medicine, NCBES Centre for Pain Research and Galway Neuroscience Centre, National University of Ireland, Galway, Ireland
| | | | | | | | | | | | | |
Collapse
|
29
|
Juraska JM, Sisk CL, DonCarlos LL. Sexual differentiation of the adolescent rodent brain: hormonal influences and developmental mechanisms. Horm Behav 2013; 64:203-10. [PMID: 23998664 DOI: 10.1016/j.yhbeh.2013.05.010] [Citation(s) in RCA: 149] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2013] [Revised: 05/03/2013] [Accepted: 05/28/2013] [Indexed: 11/24/2022]
Abstract
This article is part of a Special Issue "Puberty and Adolescence". Sexual differentiation is the process by which the nervous system becomes structurally and functionally dissimilar in females and males. In mammals, this process has been thought to occur during prenatal and early postnatal development, when a transient increase in testosterone secretion masculinizes and defeminizes the developing male nervous system. Decades of research have led to the views that structural sexual dimorphisms created during perinatal development are passively maintained throughout life, and that ovarian hormones do not play an active role in feminization of the nervous system. Furthermore, perinatal testosterone was thought to determine sex differences in neuron number by regulating cell death and cell survival, and not by regulating cell proliferation. As investigations of neural development during adolescence became more prominent in the late 20th century and revealed the extent of brain remodeling during this time, each of these tenets has been challenged and modified. Here we review evidence from the animal literature that 1) the brain is further sexually differentiated during puberty and adolescence; 2) ovarian hormones play an active role in the feminization of the brain during puberty; and 3) hormonally modulated, sex-specific addition of new neurons and glial cells, as well as loss of neurons, contribute to sexual differentiation of hypothalamic, limbic, and cortical regions during adolescence. This architectural remodeling during the adolescent phase of sexual differentiation of the brain may underlie the known sex differences in vulnerability to addiction and psychiatric disorders that emerge during this developmental period.
Collapse
Affiliation(s)
- Janice M Juraska
- Department of Psychology and Neuroscience Program, University of Illinois, 603 E Daniel St., Champaign, IL 61820, United States.
| | | | | |
Collapse
|
30
|
Bos PA, van Honk J, Ramsey NF, Stein DJ, Hermans EJ. Testosterone administration in women increases amygdala responses to fearful and happy faces. Psychoneuroendocrinology 2013; 38:808-17. [PMID: 22999654 DOI: 10.1016/j.psyneuen.2012.09.005] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2012] [Revised: 09/03/2012] [Accepted: 09/03/2012] [Indexed: 01/03/2023]
Abstract
Data from both rodents and humans show that testosterone reduces fear. This effect is hypothesized to result from testosterone's down regulating effects on the amygdala, a key region in the detection of threat and instigator of fight-or-flight behavior. However, neuroimaging studies employing testosterone administration in humans have consistently shown increased amygdala responsivity. Yet, no study to date has investigated specifically how testosterone affects the amygdala response to fearful emotional expressions. Such stimuli signal the presence of environmental threat and elicit robust amygdala responses that have consistently been associated with anxious traits. In the present study, we therefore used functional magnetic resonance imaging combined with a single administration of 0.5mg testosterone in 12 healthy women to assess testosterone's effects on amygdala responses to dynamic fearful (and happy control) faces. Our results show that both stimuli activate the amygdala. Notably, testosterone increased the amygdala response to both stimuli, and to an equal degree. Thus, testosterone appears not to reduce fear by attenuating the amygdala response toward signals of threat. Data further show that testosterone selectively increases activation of the superficial amygdala (SFA) and, to a lesser extent, the basolateral amygdala (BLA). No effect was found in the central nucleus, which is involved in the generation of autonomic fear responses. Both the SFA and BLA are considered input regions, and enhanced activation by testosterone might reflect the role of this hormone in adaptive responding to socially relevant stimuli. Furthermore, literature on the distinct roles of the SFA and BLA in fear processing show that increased activation of these subregions of the amygdala is consistent with a fear reducing effect of testosterone.
Collapse
Affiliation(s)
- Peter A Bos
- Utrecht University, Department of Experimental Psychology, Utrecht, The Netherlands.
| | | | | | | | | |
Collapse
|
31
|
Oki K, Wiseman RW, Breedlove SM, Jordan CL. Androgen receptors in muscle fibers induce rapid loss of force but not mass: implications for spinal bulbar muscular atrophy. Muscle Nerve 2013; 47:823-34. [PMID: 23629944 DOI: 10.1002/mus.23813] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/03/2013] [Indexed: 11/10/2022]
Abstract
INTRODUCTION Testosterone (T) induces motor dysfunction in transgenic (Tg) mice that overexpress wild-type androgen receptor (AR) in skeletal muscles. Because many genes implicated in motor neuron disease are expressed in skeletal muscle, mutant proteins may act in muscle to cause dysfunction in motor neuron disease. METHODS We examined contractile properties of the extensor digitorum longus (EDL) and soleus (SOL) muscles in vitro after 5 and 3 days of T treatment in motor-impaired Tg female mice. RESULTS Both muscles showed deficits in tetanic force after 5 days of T treatment, without losses in muscle mass, protein content, or fiber number. After 3 days of T treatment, only SOL showed a deficit in tetanic force comparable to that of 5 days of treatment. In both treatments, EDL showed slowed twitch kinetics, whereas SOL showed deficits in the twitch/tetanus ratio. CONCLUSIONS These results suggest calcium-handling mechanisms in muscle fibers are defective in motor-impaired mice.
Collapse
Affiliation(s)
- Kentaro Oki
- Neuroscience Program, Michigan State University, 108 Giltner Hall, 293 Farm Lane, East Lansing, Michigan 48824-1101, USA
| | | | | | | |
Collapse
|
32
|
Filová B, Ostatníková D, Celec P, Hodosy J. The effect of testosterone on the formation of brain structures. Cells Tissues Organs 2013; 197:169-77. [PMID: 23306974 DOI: 10.1159/000345567] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/01/2012] [Indexed: 11/19/2022] Open
Abstract
It has been confirmed in several studies that testosterone can significantly affect brain development. Following metabolism of this hormone by 5α-reductase to dihydrotestosterone, testosterone may act via androgen receptors, or after conversion by aromatase to estradiol, it may act via estrogen receptors. The parts of the brain which are changed under the influence of sex hormones are known as sexually dimorphic nuclei, especially in the preoptic area of the hypothalamus. Nevertheless, evidence suggests that testosterone also influences the structure of the hippocampus, specifically CA1 and CA3 areas of the hippocampus, as well as the amygdala. These brain areas are designed to convert information from short-term into long-term memory. In this review, we summarize the effects of testosterone on the organization of brain structures with respect to spatial cognitive abilities in small rodents.
Collapse
Affiliation(s)
- Barbora Filová
- Institute of Molecular Biomedicine, Comenius University, Bratislava, Slovakia
| | | | | | | |
Collapse
|