1
|
Bühler L, de Moura AC, Giovenardi M, Goffin V, Rasia-Filho AA. Sex-related gene expression in the posterodorsal medial amygdala of cycling female rats along with prolactin modulation of lordosis behavior. Brain Res 2025; 1857:149602. [PMID: 40147695 DOI: 10.1016/j.brainres.2025.149602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 03/17/2025] [Accepted: 03/24/2025] [Indexed: 03/29/2025]
Abstract
The rat posterodorsal medial amygdala (MePD) is sexually dimorphic, has a high concentration of receptors for gonadal hormones and prolactin (PRL), and modulates reproduction. To unravel genetic and functional data for this relevant node of the social behavior network, we studied the expression of ERα, ERβ, GPER1, Kiss1, Kiss1R, PRGR, PRL, PRLR, EGR1, JAK2, STAT5A, and STAT5B in the MePD of males and females along the estrous cycle using the RT-qPCR technique. We also investigated whether PRL in the MePD would affect the sexual behavior display of proestrus females by microinjecting saline, the PRL receptor antagonist Del1-9-G129R-hPRL (1 µM and 10 µM), or PRL (1 nM) and Del1-9-G129R-hPRL (10 µM) 3 h before the onset of the dark-cycle period. The estrogen-dependent lordosis behavior, indicative of sexual receptivity of proestrus females, was recorded and compared before (control) and after (test) microinjections in these groups. Sex differences were found in the right and left MePD gene expression. ERα and Kiss1R, as well as PRL, Short PRLR, and STAT5B expression is higher in cycling females than males. Kiss1 expression is higher in males than females, and GPER1 is higher during diestrus than proestrus. Furthermore, Del1-9-G129R-hPRL in the MePD significantly reduced the full display and quotient of lordosis in proestrus females, an effect restored by the co-microinjection of PRL. In conjunction, the expression of studied genes showed specific sex and estrous cycle phase features, and PRL action in the MePD plays an essential role in the display of lordosis during the ovulatory period.
Collapse
Affiliation(s)
- Letícia Bühler
- Graduate Program in Neurosciences, Federal University of Rio Grande do Sul (UFRGS), Porto Alegre, Rio Grande do Sul 9035-003, Brazil
| | - Ana Carolina de Moura
- Graduate Program in Biosciences, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre, Rio Grande do Sul 90050-170, Brazil
| | - Márcia Giovenardi
- Graduate Program in Biosciences, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre, Rio Grande do Sul 90050-170, Brazil; Graduate Program in Health Sciences, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre, Rio Grande do Sul 90050-170, Brazil; Department of Basic Sciences/Physiology, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre, Rio Grande do Sul 90050-170, Brazil.
| | - Vincent Goffin
- Université Paris Cité, INSERM U1151, CNRS UMR8253, Institut Necker Enfants Malades, F-75015 Paris, France.
| | - Alberto A Rasia-Filho
- Graduate Program in Neurosciences, Federal University of Rio Grande do Sul (UFRGS), Porto Alegre, Rio Grande do Sul 9035-003, Brazil; Graduate Program in Biosciences, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre, Rio Grande do Sul 90050-170, Brazil; Department of Basic Sciences/Physiology, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre, Rio Grande do Sul 90050-170, Brazil.
| |
Collapse
|
2
|
Ignacio B, Baeza J, Ruiz B, Romero JP, Yañez P, Ramírez C, Caprile T, Farkas C, Recabal-Beyer A. The medial amygdala's neural circuitry: Insights into social processing and sex differences. Front Neuroendocrinol 2025; 77:101190. [PMID: 40294707 DOI: 10.1016/j.yfrne.2025.101190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2025] [Revised: 03/26/2025] [Accepted: 04/22/2025] [Indexed: 04/30/2025]
Abstract
The amygdala, a critical part of the limbic system, is essential for processing social stimuli and regulating stress responses. Among its various neuronal nuclei, the medial amygdala (MeA) remains one of the least studied in humans. The MeA plays a key role in receiving inputs from the olfactory system through pheromones, as well as from crucial areas such as the hypothalamus, hippocampus, and reward system. This allows the MeA to integrate external stimuli with the organism's internal state, finetuning social interactions, endocrine responses, and innate behaviors. Recent advances in neuroscience have highlighted the sex differences of the MeA and how they influence behavior and environmental perception. Understanding these sexspecific variations in brain structures, like the MeA in rodents, is vital for applying this knowledge to humans and could help bridge gaps in our understanding and treatment of mental health disorders, which often differ between sexes in both prevalence and presentation.
Collapse
Affiliation(s)
| | - Janina Baeza
- Faculty of Medicine, Universidad de Concepción, Chile
| | - Bastián Ruiz
- Faculty of Medicine, Universidad de Concepción, Chile
| | | | - Paulina Yañez
- Department of Cellular Biology, Faculty of Biological Sciences, Universidad de Concepción, Chile
| | - Camila Ramírez
- Department of Cellular Biology, Faculty of Biological Sciences, Universidad de Concepción, Chile
| | - Teresa Caprile
- Department of Cellular Biology, Faculty of Biological Sciences, Universidad de Concepción, Chile
| | - Carlos Farkas
- Department of Basic Sciences, Faculty of Medicine, Universidad Católica de la Santísima Concepción, Chile
| | - Antonia Recabal-Beyer
- Department of Cellular Biology, Faculty of Biological Sciences, Universidad de Concepción, Chile.
| |
Collapse
|
3
|
Yang MA, Kang S, Hong SI, Lee J, Bormann NL, Lee SW, Choi DS. Astrocytes in the External Globus Pallidus Selectively Represent Routine Formation During Repeated Reward-Seeking in Mice. eNeuro 2025; 12:ENEURO.0552-24.2025. [PMID: 40032533 PMCID: PMC11913404 DOI: 10.1523/eneuro.0552-24.2025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 02/03/2025] [Accepted: 02/24/2025] [Indexed: 03/05/2025] Open
Abstract
The external globus pallidus (GPe) is a central part of the basal ganglia indirect pathway implicated in movement and decision-making. As a hub connecting the dorsal striatum and subthalamic nucleus (STN), the GPe guides repetitive and routine behaviors. However, it remains unknown how diverse GPe cells engage in routine formation while learning action sequences in repetitive reward-seeking conditioning. Here, in male mice, we investigated the Ca2+ dynamics of two GPe cell types, astrocytes and parvalbumin-expressing neurons, during routine formation. Our findings show that the dynamics of GPe astrocytes may be involved in action sequence refinement, a characteristic potentially contributing to more efficient reward-seeking behavior.
Collapse
Affiliation(s)
- Minsu Abel Yang
- Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea
- Program of Brain and Cognitive Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea
| | - Shinwoo Kang
- Department of Clinical Pharmacology, College of Medicine, Soonchunhyang University, Cheonan 31151, Republic of Korea
| | - Sa-Ik Hong
- Department of Pharmacy, Pohang SM Christianity Hospital, Pohang 37816, Republic of Korea
| | - Jeyeon Lee
- Departments of Radiology, Mayo Clinic College of Medicine and Science, Rochester, Minnesota 55905
| | - Nicholas L Bormann
- Psychiatry and Psychology, Mayo Clinic College of Medicine and Science, Rochester, Minnesota 55905
| | - Sang Wan Lee
- Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea
- Program of Brain and Cognitive Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea
- Department of Brain & Cognitive Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea
- Kim Jaechul Graduate School of AI, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea
| | - Doo-Sup Choi
- Psychiatry and Psychology, Mayo Clinic College of Medicine and Science, Rochester, Minnesota 55905
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic College of Medicine and Science, Rochester, Minnesota 55905
- Neuroscience Program, Mayo Clinic College of Medicine and Science, Rochester, Minnesota 55905
| |
Collapse
|
4
|
Farhadi Z, Khaksari M, Alivirdiloo V, Mehrjerdi FZ, Alborzi N, Baktash KB, Rezvani ME. Review on the role of hypothalamic astrocytes in the neuroendocrine control of metabolism. J Diabetes Metab Disord 2024; 23:1635-1643. [PMID: 39610541 PMCID: PMC11599663 DOI: 10.1007/s40200-024-01465-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Accepted: 06/26/2024] [Indexed: 11/30/2024]
Abstract
Astrocytes are the most numerous type of glial cells found in the nervous system. They regulate energy homeostasis in collaboration with the neuronal circuits involved in energy balance. These glial cells are equipped with sensors and receptors for nutrients and metabolic hormones in order to control energy homeostasis. Astrocytes, like hypothalamic appetite-regulating neurons, are vulnerable to the negative consequences of a high-fat diet (HFD) feeding, which is associated with an inflammatory response and transforms them into a reactive astrocyte state, consequently leading to the disruption of energy balance. Additionally, these cells have sexually dimorphic characteristics, which will lead to different metabolic outcomes in males and females. In this review, we will discuss the various physiological and pathophysiological roles of astrocytes in regulating energy balance. Finally, we will discuss the sexual dimorphism in astrocytes and the impact of estrogen on eliciting distinct responses.
Collapse
Affiliation(s)
- Zeinab Farhadi
- Yazd Neuroendocrine Research Center, School of Medicine, Shahid Sadoughi University of Medical Sciences and Health Services, Yazd, Iran
| | - Mohammad Khaksari
- Department of Physiology, Faculty of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Vahid Alivirdiloo
- Medical Doctor Ramsar Campus, Mazandaran University of Medical Sciences, Ramsar, Iran
| | - Fatemeh Zare Mehrjerdi
- Yazd Neuroendocrine Research Center, School of Medicine, Shahid Sadoughi University of Medical Sciences and Health Services, Yazd, Iran
| | - Nasrin Alborzi
- Yazd Neuroendocrine Research Center, School of Medicine, Shahid Sadoughi University of Medical Sciences and Health Services, Yazd, Iran
| | | | - Mohammad Ebrahim Rezvani
- Yazd Neuroendocrine Research Center, School of Medicine, Shahid Sadoughi University of Medical Sciences and Health Services, Yazd, Iran
| |
Collapse
|
5
|
Gozlan E, Lewit-Cohen Y, Frenkel D. Sex Differences in Astrocyte Activity. Cells 2024; 13:1724. [PMID: 39451242 PMCID: PMC11506538 DOI: 10.3390/cells13201724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 09/27/2024] [Accepted: 10/09/2024] [Indexed: 10/26/2024] Open
Abstract
Astrocytes are essential for maintaining brain homeostasis. Alterations in their activity have been associated with various brain pathologies. Sex differences were reported to affect astrocyte development and activity, and even susceptibility to different neurodegenerative diseases. This review aims to summarize the current knowledge on the effects of sex on astrocyte activity in health and disease.
Collapse
Affiliation(s)
- Elisa Gozlan
- Department of Neurobiology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 6997801, Israel; (E.G.); (Y.L.-C.)
| | - Yarden Lewit-Cohen
- Department of Neurobiology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 6997801, Israel; (E.G.); (Y.L.-C.)
| | - Dan Frenkel
- Department of Neurobiology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 6997801, Israel; (E.G.); (Y.L.-C.)
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 6997801, Israel
| |
Collapse
|
6
|
Emvalomenos GM, Kang JWM, Jupp B, Mychasiuk R, Keay KA, Henderson LA. Recent developments and challenges in positron emission tomography imaging of gliosis in chronic neuropathic pain. Pain 2024; 165:2184-2199. [PMID: 38713812 DOI: 10.1097/j.pain.0000000000003247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 03/05/2024] [Indexed: 05/09/2024]
Abstract
ABSTRACT Understanding the mechanisms that underpin the transition from acute to chronic pain is critical for the development of more effective and targeted treatments. There is growing interest in the contribution of glial cells to this process, with cross-sectional preclinical studies demonstrating specific changes in these cell types capturing targeted timepoints from the acute phase and the chronic phase. In vivo longitudinal assessment of the development and evolution of these changes in experimental animals and humans has presented a significant challenge. Recent technological advances in preclinical and clinical positron emission tomography, including the development of specific radiotracers for gliosis, offer great promise for the field. These advances now permit tracking of glial changes over time and provide the ability to relate these changes to pain-relevant symptomology, comorbid psychiatric conditions, and treatment outcomes at both a group and an individual level. In this article, we summarize evidence for gliosis in the transition from acute to chronic pain and provide an overview of the specific radiotracers available to measure this process, highlighting their potential, particularly when combined with ex vivo / in vitro techniques, to understand the pathophysiology of chronic neuropathic pain. These complementary investigations can be used to bridge the existing gap in the field concerning the contribution of gliosis to neuropathic pain and identify potential targets for interventions.
Collapse
Affiliation(s)
- Gaelle M Emvalomenos
- School of Medical Sciences [Neuroscience], and the Brain and Mind Centre, The University of Sydney, Sydney, Australia
| | - James W M Kang
- School of Medical Sciences [Neuroscience], and the Brain and Mind Centre, The University of Sydney, Sydney, Australia
| | - Bianca Jupp
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Australia
| | - Richelle Mychasiuk
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Australia
| | - Kevin A Keay
- School of Medical Sciences [Neuroscience], and the Brain and Mind Centre, The University of Sydney, Sydney, Australia
| | - Luke A Henderson
- School of Medical Sciences [Neuroscience], and the Brain and Mind Centre, The University of Sydney, Sydney, Australia
| |
Collapse
|
7
|
Chan AE, Anderson JQ, Grigsby KB, Jensen BE, Ryabinin AE, Ozburn AR. Sex differences in nucleus accumbens core circuitry engaged by binge-like ethanol drinking. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.15.608144. [PMID: 39229134 PMCID: PMC11370393 DOI: 10.1101/2024.08.15.608144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
Growing parity in Alcohol Use Disorder (AUD) diagnoses in men and women necessitates consideration of sex as a biological variable. In humans and rodents, the nucleus accumbens core (NAcc) regulates alcohol binge drinking, a risk factor for developing AUD. We labeled NAcc inputs with a viral retrograde tracer and quantified whole-brain c-Fos to determine the regions and NAcc inputs differentially engaged in male and female mice during binge-like ethanol drinking. We found that binge-like ethanol drinking females had 129 brain areas with greater c-Fos than males. Moreover, ethanol engaged more NAcc inputs in binge-like ethanol drinking females (as compared with males), including GABAergic and glutamatergic inputs. Relative to water controls, ethanol increased network modularity and decreased connectivity in both sexes and did so more dramatically in males. These results demonstrate that early-stage binge-like ethanol drinking engages brain regions and NAcc-inputs and alters network dynamics in a sex-specific manner.
Collapse
Affiliation(s)
- Amy E Chan
- Oregon Health and Science University, Dept. of Behavioral Neuroscience, Portland Alcohol Research Center, Portland, OR, 97239, USA
- Veterans Affairs Portland Health Care System, Research and Development Service, Portland, OR, 97239, USA
| | - Justin Q Anderson
- Oregon Health and Science University, Dept. of Behavioral Neuroscience, Portland Alcohol Research Center, Portland, OR, 97239, USA
- Veterans Affairs Portland Health Care System, Research and Development Service, Portland, OR, 97239, USA
| | - Kolter B Grigsby
- Oregon Health and Science University, Dept. of Behavioral Neuroscience, Portland Alcohol Research Center, Portland, OR, 97239, USA
- Veterans Affairs Portland Health Care System, Research and Development Service, Portland, OR, 97239, USA
| | - Bryan E Jensen
- Veterans Affairs Portland Health Care System, Research and Development Service, Portland, OR, 97239, USA
| | - Andrey E Ryabinin
- Oregon Health and Science University, Dept. of Behavioral Neuroscience, Portland Alcohol Research Center, Portland, OR, 97239, USA
| | - Angela R Ozburn
- Oregon Health and Science University, Dept. of Behavioral Neuroscience, Portland Alcohol Research Center, Portland, OR, 97239, USA
- Veterans Affairs Portland Health Care System, Research and Development Service, Portland, OR, 97239, USA
| |
Collapse
|
8
|
Arenaza‐Urquijo EM, Boyle R, Casaletto K, Anstey KJ, Vila‐Castelar C, Colverson A, Palpatzis E, Eissman JM, Kheng Siang Ng T, Raghavan S, Akinci M, Vonk JMJ, Machado LS, Zanwar PP, Shrestha HL, Wagner M, Tamburin S, Sohrabi HR, Loi S, Bartrés‐Faz D, Dubal DB, Vemuri P, Okonkwo O, Hohman TJ, Ewers M, Buckley RF, for the Reserve, Resilience and Protective Factors Professional Interest Area, Sex and Gender Professional Interest area and the ADDRESS! Special Interest Group. Sex and gender differences in cognitive resilience to aging and Alzheimer's disease. Alzheimers Dement 2024; 20:5695-5719. [PMID: 38967222 PMCID: PMC11350140 DOI: 10.1002/alz.13844] [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: 11/08/2023] [Revised: 03/08/2024] [Accepted: 03/21/2024] [Indexed: 07/06/2024]
Abstract
Sex and gender-biological and social constructs-significantly impact the prevalence of protective and risk factors, influencing the burden of Alzheimer's disease (AD; amyloid beta and tau) and other pathologies (e.g., cerebrovascular disease) which ultimately shape cognitive trajectories. Understanding the interplay of these factors is central to understanding resilience and resistance mechanisms explaining maintained cognitive function and reduced pathology accumulation in aging and AD. In this narrative review, the ADDRESS! Special Interest Group (Alzheimer's Association) adopted a multidisciplinary approach to provide the foundations and recommendations for future research into sex- and gender-specific drivers of resilience, including a sex/gender-oriented review of risk factors, genetics, AD and non-AD pathologies, brain structure and function, and animal research. We urge the field to adopt a sex/gender-aware approach to resilience to advance our understanding of the intricate interplay of biological and social determinants and consider sex/gender-specific resilience throughout disease stages. HIGHLIGHTS: Sex differences in resilience to cognitive decline vary by age and cognitive status. Initial evidence supports sex-specific distinctions in brain pathology. Findings suggest sex differences in the impact of pathology on cognition. There is a sex-specific change in resilience in the transition to clinical stages. Gender and sex factors warrant study: modifiable, immune, inflammatory, and vascular.
Collapse
Affiliation(s)
- Eider M. Arenaza‐Urquijo
- Environment and Health Over the Life Course Programme, Climate, Air Pollution, Nature and Urban Health ProgrammeBarcelona Institute for Global Health (ISGlobal)BarcelonaSpain
- University of Pompeu FabraBarcelonaBarcelonaSpain
| | - Rory Boyle
- Massachusetts General HospitalHarvard Medical SchoolBostonMassachusettsUSA
| | - Kaitlin Casaletto
- Department of NeurologyMemory and Aging CenterUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | - Kaarin J. Anstey
- University of New South Wales Ageing Futures InstituteSydneyNew South WalesAustralia
- Neuroscience Research AustraliaSydneyNew South WalesAustralia
- School of Psychology, University of New South WalesSidneyNew South WalesAustralia
| | | | - Aaron Colverson
- University of Florida Center for Arts in Medicine Interdisciplinary Research LabUniversity of Florida, Center of Arts in MedicineGainesvilleFloridaUSA
| | - Eleni Palpatzis
- Environment and Health Over the Life Course Programme, Climate, Air Pollution, Nature and Urban Health ProgrammeBarcelona Institute for Global Health (ISGlobal)BarcelonaSpain
- University of Pompeu FabraBarcelonaBarcelonaSpain
| | - Jaclyn M. Eissman
- Vanderbilt Memory and Alzheimer's Center, Department of NeurologyVanderbilt University Medical CenterNashvilleTennesseeUSA
- Vanderbilt Genetics InstituteVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Ted Kheng Siang Ng
- Rush Institute for Healthy Aging and Department of Internal MedicineRush University Medical CenterChicagoIllinoisUSA
| | | | - Muge Akinci
- Environment and Health Over the Life Course Programme, Climate, Air Pollution, Nature and Urban Health ProgrammeBarcelona Institute for Global Health (ISGlobal)BarcelonaSpain
- University of Pompeu FabraBarcelonaBarcelonaSpain
| | - Jet M. J. Vonk
- Department of NeurologyMemory and Aging CenterUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | - Luiza S. Machado
- Graduate Program in Biological Sciences: Biochemistry, Universidade Federal Do Rio Grande Do Sul, FarroupilhaPorto AlegreBrazil
| | - Preeti P. Zanwar
- Jefferson College of Population Health, Thomas Jefferson UniversityPhiladelphiaPennsylvaniaUSA
- The Network on Life Course and Health Dynamics and Disparities, University of Southern CaliforniaLos AngelesCaliforniaUSA
| | | | - Maude Wagner
- Rush Alzheimer's Disease Center, Rush University Medical CenterChicagoIllinoisUSA
| | - Stefano Tamburin
- Department of Neurosciences, Biomedicine and Movement SciencesUniversity of VeronaVeronaItaly
| | - Hamid R. Sohrabi
- Centre for Healthy AgeingHealth Future InstituteMurdoch UniversityMurdochWestern AustraliaAustralia
- School of Psychology, Murdoch UniversityMurdochWestern AustraliaAustralia
| | - Samantha Loi
- Neuropsychiatry Centre, Royal Melbourne HospitalParkvilleVictoriaAustralia
- Department of PsychiatryUniversity of MelbourneParkvilleVictoriaAustralia
| | - David Bartrés‐Faz
- Department of MedicineFaculty of Medicine and Health Sciences & Institut de NeurociènciesUniversity of BarcelonaBarcelonaBarcelonaSpain
- Institut d'Investigacions Biomèdiques (IDIBAPS)BarcelonaBarcelonaSpain
- Institut Guttmann, Institut Universitari de Neurorehabilitació adscrit a la Universitat Autónoma de BarcelonaBadalonaBarcelonaSpain
| | - Dena B. Dubal
- Department of Neurology and Weill Institute of NeurosciencesUniversity of California, San FranciscoSan FranciscoCaliforniaUSA
- Biomedical and Neurosciences Graduate ProgramsUniversity of California, San FranciscoSan FranciscoCaliforniaUSA
| | | | - Ozioma Okonkwo
- Alzheimer's Disease Research Center and Department of MedicineUniversity of Wisconsin School of Medicine and Public HealthMadisonWisconsinUSA
| | - Timothy J. Hohman
- Vanderbilt Memory and Alzheimer's Center, Department of NeurologyVanderbilt University Medical CenterNashvilleTennesseeUSA
- Vanderbilt Genetics InstituteVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Michael Ewers
- Institute for Stroke and Dementia ResearchKlinikum der Universität MünchenLudwig Maximilians Universität (LMU)MunichGermany
- German Center for Neurodegenerative Diseases (DZNE, Munich)MunichGermany
| | - Rachel F. Buckley
- Massachusetts General HospitalHarvard Medical SchoolBostonMassachusettsUSA
| | | |
Collapse
|
9
|
Lazzarini G, Gatta A, Miragliotta V, Vaglini F, Viaggi C, Pirone A. Glial cells are affected more than interneurons by the loss of Engrailed 2 gene in the mouse cerebellum. J Anat 2024; 244:667-675. [PMID: 38009365 PMCID: PMC10941518 DOI: 10.1111/joa.13982] [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: 09/22/2023] [Revised: 11/10/2023] [Accepted: 11/14/2023] [Indexed: 11/28/2023] Open
Abstract
Glial cells play a pivotal role in the inflammatory processes, which are common features of several neurodevelopmental and neurodegenerative disorders. Their major role in modulating neuroinflammation underscores their significance in these conditions. Engrailed-2 knockout mice (En2-/- ) are considered a valuable model for autism spectrum disorder (ASD) due to their distinctive neuroanatomical and behavioral traits. Given the higher prevalence of ASD in males, our objective was to investigate glial and interneuron alterations in the cerebellum of En2-/- mice compared with wild-type (WT) mice in both sexes. We employed immunohistochemical analysis to assess cell density for all cell types studied and analyzed the area (A) and shape factor (SF) of microglia cell bodies. Our findings revealed the following: (a) In WT mice, the density of microglia and astrocytes was higher in females than in males, while interneuron density was lower in females. Notably, in En2-mutant mice, these differences between males and females were not present. (b) In both male and female En2-/- mice, astrocyte density exceeded that in WT mice, with microglia density being greater only in females. (c) In WT females, microglia cell bodies exhibited a larger area and a lower shape factor compared to WT males. Remarkably, the En2 mutation did not appear to influence these sex-related differences. (d) In both male and female En2-/- mice, we observed a consistent pattern: microglia cell bodies displayed a larger area and a smaller shape factor. Given the ongoing debate surrounding the roles of glia and sex-related factors in ASD, our observations provide valuable insights into understanding how an ASD-associated gene En2 affects specific cell types in the cerebellum.
Collapse
Affiliation(s)
| | | | | | - Francesca Vaglini
- Department of Translational Research and of New Surgical and Medical TechnologiesUniversity of PisaPisaItaly
| | - Cristina Viaggi
- Department of Translational Research and of New Surgical and Medical TechnologiesUniversity of PisaPisaItaly
| | - Andrea Pirone
- Department of Veterinary SciencesUniversity of PisaPisaItaly
| |
Collapse
|
10
|
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
|
11
|
Guebel DV. Human hippocampal astrocytes: Computational dissection of their transcriptome, sexual differences and exosomes across ageing and mild-cognitive impairment. Eur J Neurosci 2023; 58:2677-2707. [PMID: 37427765 DOI: 10.1111/ejn.16081] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Revised: 02/20/2023] [Accepted: 06/16/2023] [Indexed: 07/11/2023]
Abstract
The role of astrocytes in Alzheimer's disease is often disregarded. Hence, characterization of astrocytes along their early evolution toward Alzheimer would be greatly beneficial. However, due to their exquisite responsiveness, in vivo studies are difficult. So public microarray data of hippocampal homogenates from (healthy) young, (healthy) elder and elder with mild cognitive impairment (MCI) were subjected to re-analysis by a multi-step computational pipeline. Ontologies and pathway analyses were compared after determining the differential genes that, belonging to astrocytes, have splice forms. Likewise, the subset of molecules exportable to exosomes was also determined. The results showed that astrocyte's phenotypes changed significantly. While already 'activated' astrocytes were found in the younger group, major changes occurred during ageing (increased vascular remodelling and response to mechanical stimulus, diminished long-term potentiation and increased long-term depression). MCI's astrocytes showed some 'rejuvenated' features, but their sensitivity to shear stress was markedly lost. Importantly, most of the changes showed to be sex biassed. Men's astrocytes are enriched in a type 'endfeet-astrocytome', whereas women's astrocytes appear close to the 'scar-forming' type (prone to endothelial dysfunction, hypercholesterolemia, loss of glutamatergic synapses, Ca+2 dysregulation, hypoxia, oxidative stress and 'pro-coagulant' phenotype). In conclusion, the computational dissection of the networks based on the hippocampal gene isoforms provides a relevant proxy to in vivo astrocytes, also revealing the occurrence of sexual differences. Analyses of the astrocytic exosomes did not provide an acceptable approximation to the overall functioning of astrocytes in the hippocampus, probably due to the selective cellular mechanisms which charge the cargo molecules.
Collapse
|
12
|
Wright CJ, Milosavljevic S, Pocivavsek A. The stress of losing sleep: Sex-specific neurobiological outcomes. Neurobiol Stress 2023; 24:100543. [PMID: 37252645 PMCID: PMC10209346 DOI: 10.1016/j.ynstr.2023.100543] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 03/20/2023] [Accepted: 05/06/2023] [Indexed: 05/31/2023] Open
Abstract
Sleep is a vital and evolutionarily conserved process, critical to daily functioning and homeostatic balance. Losing sleep is inherently stressful and leads to numerous detrimental physiological outcomes. Despite sleep disturbances affecting everyone, women and female rodents are often excluded or underrepresented in clinical and pre-clinical studies. Advancing our understanding of the role of biological sex in the responses to sleep loss stands to greatly improve our ability to understand and treat health consequences of insufficient sleep. As such, this review discusses sex differences in response to sleep deprivation, with a focus on the sympathetic nervous system stress response and activation of the hypothalamic-pituitary-adrenal (HPA) axis. We review sex differences in several stress-related consequences of sleep loss, including inflammation, learning and memory deficits, and mood related changes. Focusing on women's health, we discuss the effects of sleep deprivation during the peripartum period. In closing, we present neurobiological mechanisms, including the contribution of sex hormones, orexins, circadian timing systems, and astrocytic neuromodulation, that may underlie potential sex differences in sleep deprivation responses.
Collapse
Affiliation(s)
- Courtney J. Wright
- Department of Pharmacology, Physiology, and Neuroscience, University of South Carolina School of Medicine, Columbia, SC, USA
| | - Snezana Milosavljevic
- Department of Pharmacology, Physiology, and Neuroscience, University of South Carolina School of Medicine, Columbia, SC, USA
| | - Ana Pocivavsek
- Department of Pharmacology, Physiology, and Neuroscience, University of South Carolina School of Medicine, Columbia, SC, USA
| |
Collapse
|
13
|
Rasia-Filho AA, Calcagnotto ME, von Bohlen Und Halbach O. Glial Cell Modulation of Dendritic Spine Structure and Synaptic Function. ADVANCES IN NEUROBIOLOGY 2023; 34:255-310. [PMID: 37962798 DOI: 10.1007/978-3-031-36159-3_6] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Glia comprise a heterogeneous group of cells involved in the structure and function of the central and peripheral nervous system. Glial cells are found from invertebrates to humans with morphological specializations related to the neural circuits in which they are embedded. Glial cells modulate neuronal functions, brain wiring and myelination, and information processing. For example, astrocytes send processes to the synaptic cleft, actively participate in the metabolism of neurotransmitters, and release gliotransmitters, whose multiple effects depend on the targeting cells. Human astrocytes are larger and more complex than their mice and rats counterparts. Astrocytes and microglia participate in the development and plasticity of neural circuits by modulating dendritic spines. Spines enhance neuronal connectivity, integrate most postsynaptic excitatory potentials, and balance the strength of each input. Not all central synapses are engulfed by astrocytic processes. When that relationship occurs, a different pattern for thin and large spines reflects an activity-dependent remodeling of motile astrocytic processes around presynaptic and postsynaptic elements. Microglia are equally relevant for synaptic processing, and both glial cells modulate the switch of neuroendocrine secretion and behavioral display needed for reproduction. In this chapter, we provide an overview of the structure, function, and plasticity of glial cells and relate them to synaptic maturation and modulation, also involving neurotrophic factors. Together, neurons and glia coordinate synaptic transmission in both normal and abnormal conditions. Neglected over decades, this exciting research field can unravel the complexity of species-specific neural cytoarchitecture as well as the dynamic region-specific functional interactions between diverse neurons and glial subtypes.
Collapse
Affiliation(s)
- Alberto A Rasia-Filho
- Department of Basic Sciences/Physiology and Graduate Program in Biosciences, Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre, RS, Brazil
- Graduate Program in Neuroscience, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Maria Elisa Calcagnotto
- Graduate Program in Neuroscience, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
- Department of Biochemistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
- Graduate Program in Biochemistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
- Graduate Program in Psychiatry and Behavioral Science, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | | |
Collapse
|
14
|
Manion MTC, Glasper ER, Wang KH. A sex difference in mouse dopaminergic projections from the midbrain to basolateral amygdala. Biol Sex Differ 2022; 13:75. [PMID: 36585727 PMCID: PMC9801632 DOI: 10.1186/s13293-022-00486-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Accepted: 12/20/2022] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Dopaminergic circuits play important roles in the motivational control of behavior and dysfunction in dopaminergic circuits have been implicated in several psychiatric disorders, such as schizophrenia and depression. While these disorders exhibit different incidence rates in men and women, the potential sex differences in the underlying neural circuits are not well-understood. Previous anatomical tracing studies in mammalian species have revealed a prominent circuit projection connecting the dopaminergic midbrain ventral tegmental area (VTA) to the basolateral amygdala (BLA), which is involved in emotional processing and associative learning. However, whether there is any sex difference in this anatomical circuit remains unknown. METHODS To study the potential sex differences in the VTA-to-BLA dopaminergic circuit, we injected two viral vectors encoding fluorescent reporters of axons and synaptic boutons (AAV-FLEX-tdTomato and AAV-FLEX-SynaptophysinGFP, respectively) into the VTA of a mouse transgenic driver line (tyrosine hydroxylase promoter-driven Cre, or TH-Cre), which restricts the reporter expression to dopaminergic neurons. We then used confocal fluorescent microscopy to image the distribution and density of dopaminergic axons and synaptic boutons in serial sections of both male and female mouse brain. RESULTS We found that the overall labeling intensity of VTA-to-BLA dopaminergic projections is intermediate among forebrain dopaminergic pathways, significantly higher than the projections to the prefrontal cortex, but lower than the projections to the nucleus accumbens. Within the amygdala areas, dopaminergic axons are concentrated in BLA. Although the size of BLA and the density of dopaminergic axons within BLA are similar between male and female mice, the density of dopaminergic synaptic boutons in BLA is significantly higher in male brain than female brain. CONCLUSIONS Our results demonstrate an anatomical sex difference in mouse dopaminergic innervations from the VTA to BLA. This finding may provide a structural foundation to study neural circuit mechanisms underlying sex differences in motivational and emotional behaviors and related psychiatric dysfunctions.
Collapse
Affiliation(s)
- Matthew T. C. Manion
- grid.416868.50000 0004 0464 0574Unit on Neural Circuits and Adaptive Behaviors, National Institute of Mental Health, National Institutes of Health, Bethesda, MD 20892 USA ,grid.164295.d0000 0001 0941 7177Department of Psychology, University of Maryland, College Park, MD 20742 USA ,grid.164295.d0000 0001 0941 7177Program in Neuroscience and Cognitive Science, University of Maryland, College Park, MD 20742 USA
| | - Erica R. Glasper
- grid.164295.d0000 0001 0941 7177Department of Psychology, University of Maryland, College Park, MD 20742 USA ,grid.164295.d0000 0001 0941 7177Program in Neuroscience and Cognitive Science, University of Maryland, College Park, MD 20742 USA ,grid.261331.40000 0001 2285 7943Department of Neuroscience and Institute for Behavioral Medicine Research, The Ohio State University, Columbus, OH 43235 USA
| | - Kuan Hong Wang
- grid.416868.50000 0004 0464 0574Unit on Neural Circuits and Adaptive Behaviors, National Institute of Mental Health, National Institutes of Health, Bethesda, MD 20892 USA ,grid.412750.50000 0004 1936 9166Department of Neuroscience, Del Monte Institute for Neuroscience, University of Rochester Medical Center, Rochester, NY 14642 USA
| |
Collapse
|
15
|
Alia AO, Jeon S, Popovic J, Salvo MA, Sadleir KR, Vassar R, Cuddy LK. Aberrant glial activation and synaptic defects in CaMKIIα-iCre and nestin-Cre transgenic mouse models. Sci Rep 2022; 12:22099. [PMID: 36543864 PMCID: PMC9772212 DOI: 10.1038/s41598-022-26671-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Accepted: 12/19/2022] [Indexed: 12/24/2022] Open
Abstract
Current scientific research is driven by the ability to manipulate gene expression by utilizing the Cre/loxP system in transgenic mouse models. However, artifacts in Cre-driver mouse lines that introduce undesired effects and confound results are increasingly being reported. Here, we show aberrant neuroinflammation and synaptic changes in two widely used Cre-driver mouse models. Neuroinflammation in CaMKIIα-iCre mice was characterized by the activation and proliferation of microglia and astrocytes in synaptic layers of the hippocampus. Increased GFAP and Iba1 levels were observed in hippocampal brain regions of 4-, 8- and 22-month-old CaMKIIα-iCre mice compared to WT littermates. Synaptic changes in NMDAR, AMPAR, PSD95 and phosphorylated CaMKIIα became apparent in 8-month-old CaMKIIα-iCre mice but were not observed in 4-month-old CaMKIIα-iCre mice. Synaptophysin and synaptoporin were unchanged in CaMKIIα-iCre compared to WT mice, suggesting that synaptic alterations may occur in excitatory postsynaptic regions in which iCre is predominantly expressed. Finally, hippocampal volume was reduced in 22-month-old CaMKIIα-iCre mice compared to WT mice. We tested the brains of mice of additional common Cre-driver mouse models for neuroinflammation; the nestin-Cre mouse model showed synaptic changes and astrocytosis marked by increased GFAP+ astrocytes in cortical and hippocampal regions, while the original CaMKIIα-Cre T29-1 strain was comparable to WT mice. The mechanisms underlying abnormal neuroinflammation in nestin-Cre and CaMKIIα-iCre are unknown but may be associated with high levels of Cre expression. Our findings are critical to the scientific community and demonstrate that the correct Cre-driver controls must be included in all studies using these mice.
Collapse
Affiliation(s)
- Alia O. Alia
- grid.16753.360000 0001 2299 3507The Ken and Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611 USA
| | - Sohee Jeon
- grid.16753.360000 0001 2299 3507The Ken and Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611 USA
| | - Jelena Popovic
- grid.16753.360000 0001 2299 3507The Ken and Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611 USA
| | - Miranda A. Salvo
- grid.16753.360000 0001 2299 3507The Ken and Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611 USA
| | - Katherine R. Sadleir
- grid.16753.360000 0001 2299 3507The Ken and Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611 USA
| | - Robert Vassar
- grid.16753.360000 0001 2299 3507The Ken and Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611 USA ,grid.16753.360000 0001 2299 3507Mesulam Center for Cognitive Neurology and Alzheimer’s Disease, Northwestern University Feinberg School of Medicine, Chicago, IL 60611 USA
| | - Leah K. Cuddy
- grid.16753.360000 0001 2299 3507The Ken and Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611 USA
| |
Collapse
|
16
|
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
|
17
|
Saleki K, Banazadeh M, Saghazadeh A, Rezaei N. Aging, testosterone, and neuroplasticity: friend or foe? Rev Neurosci 2022; 34:247-273. [PMID: 36017670 DOI: 10.1515/revneuro-2022-0033] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Accepted: 07/03/2022] [Indexed: 11/15/2022]
Abstract
Neuroplasticity or neural plasticity implicates the adaptive potential of the brain in response to extrinsic and intrinsic stimuli. The concept has been utilized in different contexts such as injury and neurological disease. Neuroplasticity mechanisms have been classified into neuroregenerative and function-restoring processes. In the context of injury, neuroplasticity has been defined in three post-injury epochs. Testosterone plays a key yet double-edged role in the regulation of several neuroplasticity alterations. Research has shown that testosterone levels are affected by numerous factors such as age, stress, surgical procedures on gonads, and pharmacological treatments. There is an ongoing debate for testosterone replacement therapy (TRT) in aging men; however, TRT is more useful in young individuals with testosterone deficit and more specific subgroups with cognitive dysfunction. Therefore, it is important to pay early attention to testosterone profile and precisely uncover its harms and benefits. In the present review, we discuss the influence of environmental factors, aging, and gender on testosterone-associated alterations in neuroplasticity, as well as the two-sided actions of testosterone in the nervous system. Finally, we provide practical insights for further study of pharmacological treatments for hormonal disorders focusing on restoring neuroplasticity.
Collapse
Affiliation(s)
- Kiarash Saleki
- Student Research Committee, Babol University of Medical Sciences, 47176 47745 Babol, Iran.,USERN Office, Babol University of Medical Sciences, 47176 47745 Babol, Iran.,Systematic Review and Meta-analysis Expert Group (SRMEG), Universal Scientific Education and Research Network (USERN), 14197 33151 Tehran, Iran
| | - Mohammad Banazadeh
- Systematic Review and Meta-analysis Expert Group (SRMEG), Universal Scientific Education and Research Network (USERN), 14197 33151 Tehran, Iran.,Pharmaceutical Sciences and Cosmetic Products Research Center, Kerman University of Medical Sciences, 76169 13555 Kerman, Iran
| | - Amene Saghazadeh
- Systematic Review and Meta-analysis Expert Group (SRMEG), Universal Scientific Education and Research Network (USERN), 14197 33151 Tehran, Iran.,Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, 14197 33151 Tehran, Iran
| | - Nima Rezaei
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, 14197 33151 Tehran, Iran.,Department of Immunology, School of Medicine, Tehran University of Medical Sciences, 14176 13151 Tehran, Iran.,Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), 14197 33151 Tehran, Iran
| |
Collapse
|
18
|
Shanmugan S, Seidlitz J, Cui Z, Adebimpe A, Bassett DS, Bertolero MA, Davatzikos C, Fair DA, Gur RE, Gur RC, Larsen B, Li H, Pines A, Raznahan A, Roalf DR, Shinohara RT, Vogel J, Wolf DH, Fan Y, Alexander-Bloch A, Satterthwaite TD. Sex differences in the functional topography of association networks in youth. Proc Natl Acad Sci U S A 2022; 119:e2110416119. [PMID: 35939696 PMCID: PMC9388107 DOI: 10.1073/pnas.2110416119] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Accepted: 07/15/2022] [Indexed: 01/16/2023] Open
Abstract
Prior work has shown that there is substantial interindividual variation in the spatial distribution of functional networks across the cerebral cortex, or functional topography. However, it remains unknown whether there are sex differences in the topography of individualized networks in youth. Here, we leveraged an advanced machine learning method (sparsity-regularized non-negative matrix factorization) to define individualized functional networks in 693 youth (ages 8 to 23 y) who underwent functional MRI as part of the Philadelphia Neurodevelopmental Cohort. Multivariate pattern analysis using support vector machines classified participant sex based on functional topography with 82.9% accuracy (P < 0.0001). Brain regions most effective in classifying participant sex belonged to association networks, including the ventral attention, default mode, and frontoparietal networks. Mass univariate analyses using generalized additive models with penalized splines provided convergent results. Furthermore, transcriptomic data from the Allen Human Brain Atlas revealed that sex differences in multivariate patterns of functional topography were spatially correlated with the expression of genes on the X chromosome. These results highlight the role of sex as a biological variable in shaping functional topography.
Collapse
Affiliation(s)
- Sheila Shanmugan
- Penn Lifespan Informatics and Neuroimaging Center, University of Pennsylvania, Philadelphia, PA 19104
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA 19104
- Penn-Children's Hospital of Philadelphia Lifespan Brain Institute, University of Pennsylvania, Philadelphia, PA 19104
| | - Jakob Seidlitz
- Penn Lifespan Informatics and Neuroimaging Center, University of Pennsylvania, Philadelphia, PA 19104
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA 19104
- Penn-Children's Hospital of Philadelphia Lifespan Brain Institute, University of Pennsylvania, Philadelphia, PA 19104
| | - Zaixu Cui
- Penn Lifespan Informatics and Neuroimaging Center, University of Pennsylvania, Philadelphia, PA 19104
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA 19104
- Penn-Children's Hospital of Philadelphia Lifespan Brain Institute, University of Pennsylvania, Philadelphia, PA 19104
- Chinese Institute for Brain Research, Beijing,102206, China
| | - Azeez Adebimpe
- Penn Lifespan Informatics and Neuroimaging Center, University of Pennsylvania, Philadelphia, PA 19104
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA 19104
- Penn-Children's Hospital of Philadelphia Lifespan Brain Institute, University of Pennsylvania, Philadelphia, PA 19104
| | - Danielle S. Bassett
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA 19104
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104
- Department of Neurology, University of Pennsylvania, Philadelphia, PA 19104
- Department of Electrical and Systems Engineering, University of Pennsylvania, Philadelphia, PA 19104
- Department of Physics and Astronomy, University of Pennsylvania, Philadelphia, PA 19104
- Santa Fe Institute, Santa Fe, NM 87501
| | - Maxwell A. Bertolero
- Penn Lifespan Informatics and Neuroimaging Center, University of Pennsylvania, Philadelphia, PA 19104
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA 19104
- Penn-Children's Hospital of Philadelphia Lifespan Brain Institute, University of Pennsylvania, Philadelphia, PA 19104
| | - Christos Davatzikos
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104
- Department of Electrical and Systems Engineering, University of Pennsylvania, Philadelphia, PA 19104
- Department of Radiology, University of Pennsylvania, Philadelphia, PA 19104
- Center for Biomedical Image Computation and Analytics, University of Pennsylvania, Philadelphia, PA 19104
| | - Damien A. Fair
- Department of Behavioral Neuroscience, Department of Psychiatry, Advanced Imaging Research Center, Oregon Health and Science University, Portland, OR 97239
| | - Raquel E. Gur
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA 19104
- Penn-Children's Hospital of Philadelphia Lifespan Brain Institute, University of Pennsylvania, Philadelphia, PA 19104
- Department of Neurology, University of Pennsylvania, Philadelphia, PA 19104
- Department of Radiology, University of Pennsylvania, Philadelphia, PA 19104
- Center for Biomedical Image Computation and Analytics, University of Pennsylvania, Philadelphia, PA 19104
| | - Ruben C. Gur
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA 19104
- Penn-Children's Hospital of Philadelphia Lifespan Brain Institute, University of Pennsylvania, Philadelphia, PA 19104
- Department of Neurology, University of Pennsylvania, Philadelphia, PA 19104
- Center for Biomedical Image Computation and Analytics, University of Pennsylvania, Philadelphia, PA 19104
| | - Bart Larsen
- Penn Lifespan Informatics and Neuroimaging Center, University of Pennsylvania, Philadelphia, PA 19104
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA 19104
- Penn-Children's Hospital of Philadelphia Lifespan Brain Institute, University of Pennsylvania, Philadelphia, PA 19104
| | - Hongming Li
- Department of Radiology, University of Pennsylvania, Philadelphia, PA 19104
- Center for Biomedical Image Computation and Analytics, University of Pennsylvania, Philadelphia, PA 19104
| | - Adam Pines
- Penn Lifespan Informatics and Neuroimaging Center, University of Pennsylvania, Philadelphia, PA 19104
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA 19104
- Penn-Children's Hospital of Philadelphia Lifespan Brain Institute, University of Pennsylvania, Philadelphia, PA 19104
| | - Armin Raznahan
- Section on Developmental Neurogenomics Unit, Intramural Research Program, National Institutes of Mental Health, Bethesda, MD 20892
| | - David R. Roalf
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA 19104
- Penn-Children's Hospital of Philadelphia Lifespan Brain Institute, University of Pennsylvania, Philadelphia, PA 19104
| | - Russell T. Shinohara
- Center for Biomedical Image Computation and Analytics, University of Pennsylvania, Philadelphia, PA 19104
- Penn Statistics in Imaging and Visualization Center, Department of Biostatistics, Epidemiology, and Informatics, University of Pennsylvania, Philadelphia, PA 19104
| | - Jacob Vogel
- Penn Lifespan Informatics and Neuroimaging Center, University of Pennsylvania, Philadelphia, PA 19104
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA 19104
- Penn-Children's Hospital of Philadelphia Lifespan Brain Institute, University of Pennsylvania, Philadelphia, PA 19104
| | - Daniel H. Wolf
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA 19104
- Penn-Children's Hospital of Philadelphia Lifespan Brain Institute, University of Pennsylvania, Philadelphia, PA 19104
- Center for Biomedical Image Computation and Analytics, University of Pennsylvania, Philadelphia, PA 19104
| | - Yong Fan
- Department of Radiology, University of Pennsylvania, Philadelphia, PA 19104
- Center for Biomedical Image Computation and Analytics, University of Pennsylvania, Philadelphia, PA 19104
| | - Aaron Alexander-Bloch
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA 19104
- Penn-Children's Hospital of Philadelphia Lifespan Brain Institute, University of Pennsylvania, Philadelphia, PA 19104
| | - Theodore D. Satterthwaite
- Penn Lifespan Informatics and Neuroimaging Center, University of Pennsylvania, Philadelphia, PA 19104
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA 19104
- Penn-Children's Hospital of Philadelphia Lifespan Brain Institute, University of Pennsylvania, Philadelphia, PA 19104
- Center for Biomedical Image Computation and Analytics, University of Pennsylvania, Philadelphia, PA 19104
| |
Collapse
|
19
|
Boorman DC, Keay KA. Sex differences in morphine sensitivity are associated with differential glial expression in the brainstem of rats with neuropathic pain. J Neurosci Res 2022; 100:1890-1907. [PMID: 35853016 PMCID: PMC9543783 DOI: 10.1002/jnr.25103] [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: 03/15/2022] [Revised: 06/20/2022] [Accepted: 06/22/2022] [Indexed: 11/11/2022]
Abstract
Chronic pain is more prevalent and reported to be more severe in women. Opioid analgesics are less effective in women and result in stronger nauseant effects. The neurobiological mechanisms underlying these sex differences have yet to be clearly defined, though recent research has suggested neuronal–glial interactions are likely involved. We have previously shown that similar to people, morphine is less effective at reducing pain behaviors in female rats. In this study, we used the immunohistochemical detection of glial fibrillary acidic protein (GFAP) expression to investigate sex differences in astrocyte density and morphology in six medullary regions known to be modulated by pain and/or opioids. Morphine administration had small sex‐dependent effects on overall GFAP expression, but not on astrocyte morphology, in the rostral ventromedial medulla, the subnucleus reticularis dorsalis, and the area postrema. Significant sex differences in the density and morphology of GFAP immunopositive astrocytes were detected in all six regions. In general, GFAP‐positive cells in females showed smaller volumes and reduced complexity than those observed in males. Furthermore, females showed lower overall GFAP expression in all regions except for the area postrema, the critical medullary region responsible for opioid‐induced nausea and emesis. These data support the possibility that differences in astrocyte activity might underlie the sex differences seen in the processing of opioids in the context of chronic neuropathic pain.
Collapse
Affiliation(s)
- Damien C. Boorman
- School of Medical Sciences and the Brain and Mind Centre The University of Sydney Camperdown New South Wales Australia
| | - Kevin A. Keay
- School of Medical Sciences and the Brain and Mind Centre The University of Sydney Camperdown New South Wales Australia
| |
Collapse
|
20
|
Lee RX, Stephens GJ, Kuhn B. Social Relationship as a Factor for the Development of Stress Incubation in Adult Mice. Front Behav Neurosci 2022; 16:854486. [PMID: 35685272 PMCID: PMC9172995 DOI: 10.3389/fnbeh.2022.854486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 04/12/2022] [Indexed: 11/13/2022] Open
Abstract
While stress reactions can emerge long after the triggering event, it remains elusive how they emerge after a protracted, seemingly stress-free period during which stress incubates. Here, we study the behavioral development in mice isolated after observing an aggressive encounter inflicted upon their pair-housed partners. We developed a spatially resolved fine-scale behavioral analysis and applied it to standard behavioral tests. It reveals that the seemingly sudden behavioral changes developed gradually. These behavioral changes were not observed if the aggressive encounter happened to a stranger mouse, suggesting that social bonding is a prerequisite for stress incubation in this paradigm. This finding was corroborated by hemisphere-specific morphological changes in cortex regions centering at the anterior cingulate cortex, a cognitive and emotional center. Our non-invasive analytical methods to capture informative behavioral details may have applications beyond laboratory animals.
Collapse
Affiliation(s)
- Ray X. Lee
- Optical Neuroimaging Unit, Okinawa Institute of Science and Technology (OIST) Graduate University, Okinawa, Japan
- Biological Physics Theory Unit, Okinawa Institute of Science and Technology (OIST) Graduate University, Okinawa, Japan
- *Correspondence: Ray X. Lee,
| | - Greg J. Stephens
- Biological Physics Theory Unit, Okinawa Institute of Science and Technology (OIST) Graduate University, Okinawa, Japan
- Department of Physics and Astronomy, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Bernd Kuhn
- Optical Neuroimaging Unit, Okinawa Institute of Science and Technology (OIST) Graduate University, Okinawa, Japan
| |
Collapse
|
21
|
Presto P, Mazzitelli M, Junell R, Griffin Z, Neugebauer V. Sex differences in pain along the neuraxis. Neuropharmacology 2022; 210:109030. [DOI: 10.1016/j.neuropharm.2022.109030] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 02/24/2022] [Accepted: 03/12/2022] [Indexed: 12/30/2022]
|
22
|
Krawczyk MC, Haney JR, Pan L, Caneda C, Khankan RR, Reyes SD, Chang JW, Morselli M, Vinters HV, Wang AC, Cobos I, Gandal MJ, Bergsneider M, Kim W, Liau LM, Yong W, Jalali A, Deneen B, Grant GA, Mathern GW, Fallah A, Zhang Y. Human Astrocytes Exhibit Tumor Microenvironment-, Age-, and Sex-Related Transcriptomic Signatures. J Neurosci 2022; 42:1587-1603. [PMID: 34987109 PMCID: PMC8883850 DOI: 10.1523/jneurosci.0407-21.2021] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 08/23/2021] [Accepted: 09/28/2021] [Indexed: 11/21/2022] Open
Abstract
Astrocytes are critical for the development and function of synapses. There are notable species differences between human astrocytes and commonly used animal models. Yet, it is unclear whether astrocytic genes involved in synaptic function are stable or exhibit dynamic changes associated with disease states and age in humans, which is a barrier in understanding human astrocyte biology and its potential involvement in neurologic diseases. To better understand the properties of human astrocytes, we acutely purified astrocytes from the cerebral cortices of over 40 humans across various ages, sexes, and disease states. We performed RNA sequencing to generate transcriptomic profiles of these astrocytes and identified genes associated with these biological variables. We found that human astrocytes in tumor-surrounding regions downregulate genes involved in synaptic function and sensing of signals in the microenvironment, suggesting involvement of peritumor astrocytes in tumor-associated neural circuit dysfunction. In aging, we also found downregulation of synaptic regulators and upregulation of markers of cytokine signaling, while in maturation we identified changes in ionic transport with implications for calcium signaling. In addition, we identified subtle sexual dimorphism in human cortical astrocytes, which has implications for observed sex differences across many neurologic disorders. Overall, genes involved in synaptic function exhibit dynamic changes in the peritumor microenvironment and aging. These data provide powerful new insights into human astrocyte biology in several biologically relevant states that will aid in generating novel testable hypotheses about homeostatic and reactive astrocytes in humans.SIGNIFICANCE STATEMENT Astrocytes are an abundant class of cells playing integral roles at synapses. Astrocyte dysfunction is implicated in a variety of human neurologic diseases. Yet our knowledge of astrocytes is largely based on mouse studies. Direct knowledge of human astrocyte biology remains limited. Here, we present transcriptomic profiles of human cortical astrocytes, and we identified molecular differences associated with age, sex, and disease state. We found that peritumor and aging astrocytes downregulate genes involved in astrocyte-synapse interactions. These data provide necessary insight into human astrocyte biology that will improve our understanding of human disease.
Collapse
Affiliation(s)
- Mitchell C Krawczyk
- Department of Psychiatry and Biobehavioral Sciences, Intellectual and Developmental Disabilities Research Center, Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine, University of California, Los Angeles, California, 90024
| | - Jillian R Haney
- Department of Psychiatry and Biobehavioral Sciences, Intellectual and Developmental Disabilities Research Center, Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine, University of California, Los Angeles, California, 90024
| | - Lin Pan
- Department of Psychiatry and Biobehavioral Sciences, Intellectual and Developmental Disabilities Research Center, Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine, University of California, Los Angeles, California, 90024
| | - Christine Caneda
- Department of Psychiatry and Biobehavioral Sciences, Intellectual and Developmental Disabilities Research Center, Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine, University of California, Los Angeles, California, 90024
| | - Rana R Khankan
- Department of Psychiatry and Biobehavioral Sciences, Intellectual and Developmental Disabilities Research Center, Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine, University of California, Los Angeles, California, 90024
| | - Samuel D Reyes
- Department of Neurosurgery, David Geffen School of Medicine, University of California, Los Angeles, California, 90024
| | - Julia W Chang
- Department of Psychiatry and Biobehavioral Sciences, Intellectual and Developmental Disabilities Research Center, Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine, University of California, Los Angeles, California, 90024
- Department of Neurosurgery, David Geffen School of Medicine, University of California, Los Angeles, California, 90024
| | - Marco Morselli
- Department of Molecular, Cell and Developmental Biology, UCLA-DOE Institute for Genomics and Proteomics, Institute for Quantitative and Computational Biosciences - The Collaboratory at University of California, Los Angeles, California, 90024
| | - Harry V Vinters
- Department of Pathology and Lab Medicine (Neuropathology) and Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, California, 90024
- Ronald Reagan UCLA Medical Center, Los Angeles, California, 90024
| | - Anthony C Wang
- Department of Neurosurgery, David Geffen School of Medicine, University of California, Los Angeles, California, 90024
| | - Inma Cobos
- Department of Pathology, Stanford University, Stanford, California, 94305
| | - Michael J Gandal
- Department of Psychiatry and Biobehavioral Sciences, Intellectual and Developmental Disabilities Research Center, Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine, University of California, Los Angeles, California, 90024
- Program in Neurobehavioral Genetics, Semel Institute, David Geffen School of Medicine; Department of Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles, California, 90024
| | - Marvin Bergsneider
- Department of Neurosurgery, David Geffen School of Medicine, University of California, Los Angeles, California, 90024
| | - Won Kim
- Department of Neurosurgery, David Geffen School of Medicine, University of California, Los Angeles, California, 90024
| | - Linda M Liau
- Department of Neurosurgery, David Geffen School of Medicine, University of California, Los Angeles, California, 90024
- Jonsson Comprehensive Cancer Center at UCLA, Los Angeles, California, 90024
| | - William Yong
- Department of Pathology, University of California, Irvine, California, 90095
| | - Ali Jalali
- Department of Neurosurgery, Baylor College of Medicine, Houston, Texas, 77030
| | - Benjamin Deneen
- Department of Neurosurgery, Baylor College of Medicine, Houston, Texas, 77030
- Center for Cell and Gene Therapy, Department of Neuroscience, Department of Neurosurgery, Baylor College of Medicine, Houston, Texas, 77030
| | - Gerald A Grant
- Department of Neurosurgery, Stanford University, Stanford, California, 94305
| | - Gary W Mathern
- Department of Psychiatry and Biobehavioral Sciences, Intellectual and Developmental Disabilities Research Center, Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine, University of California, Los Angeles, California, 90024
- Department of Neurosurgery, David Geffen School of Medicine, University of California, Los Angeles, California, 90024
| | - Aria Fallah
- Department of Neurosurgery, David Geffen School of Medicine, University of California, Los Angeles, California, 90024
| | - Ye Zhang
- Department of Psychiatry and Biobehavioral Sciences, Intellectual and Developmental Disabilities Research Center, Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine, University of California, Los Angeles, California, 90024
- Brain Research Institute at UCLA, Los Angeles, California, 90095
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research at UCLA, Los Angeles, California, 90095
- Molecular Biology Institute at UCLA, Los Angeles, California, 90095
| |
Collapse
|
23
|
Kuwahara N, Nicholson K, Isaacs L, MacLusky NJ. Androgen Effects on Neural Plasticity. ANDROGENS: CLINICAL RESEARCH AND THERAPEUTICS 2021; 2:216-230. [PMID: 35024693 PMCID: PMC8744448 DOI: 10.1089/andro.2021.0022] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Accepted: 10/24/2021] [Indexed: 12/20/2022]
Abstract
Androgens are synthesized in the brain, gonads, and adrenal glands, in both sexes, exerting physiologically important effects on the structure and function of the central nervous system. These effects may contribute to the incidence and progression of neurological disorders such as autism spectrum disorder, schizophrenia, and Alzheimer's disease, which occur at different rates in males and females. This review briefly summarizes the current state of knowledge with respect to the neuroplastic effects of androgens, with particular emphasis on the hippocampus, which has been the focus of much of the research in this field.
Collapse
Affiliation(s)
- Nariko Kuwahara
- Department of Biomedical Sciences, University of Guelph, Guelph, Ontario N1G 2W1, Canada
| | - Kate Nicholson
- Department of Biomedical Sciences, University of Guelph, Guelph, Ontario N1G 2W1, Canada
| | - Lauren Isaacs
- Department of Biomedical Sciences, University of Guelph, Guelph, Ontario N1G 2W1, Canada
| | - Neil J. MacLusky
- Department of Biomedical Sciences, University of Guelph, Guelph, Ontario N1G 2W1, Canada
| |
Collapse
|
24
|
Blank N, Mayer M, Mass E. The development and physiological and pathophysiological functions of resident macrophages and glial cells. Adv Immunol 2021; 151:1-47. [PMID: 34656287 DOI: 10.1016/bs.ai.2021.08.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
In the past, brain function and the onset and progression of neurological diseases have been studied in a neuron-centric manner. However, in recent years the focus of many neuroscientists has shifted to other cell types that promote neurodevelopment and contribute to the functionality of neuronal networks in health and disease. Particularly microglia and astrocytes have been implicated in actively contributing to and controlling neuronal development, neuroinflammation, and neurodegeneration. Here, we summarize the development of brain-resident macrophages and astrocytes and their core functions in the developing brain. We discuss their contribution and intercellular crosstalk during tissue homeostasis and pathophysiology. We argue that in-depth knowledge of non-neuronal cells in the brain could provide novel therapeutic targets to reverse or contain neurological diseases.
Collapse
Affiliation(s)
- Nelli Blank
- Developmental Biology of the Immune System, Life & Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany.
| | - Marina Mayer
- Developmental Biology of the Immune System, Life & Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany
| | - Elvira Mass
- Developmental Biology of the Immune System, Life & Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany.
| |
Collapse
|
25
|
Spichak S, Donoso F, Moloney GM, Gunnigle E, Brown JM, Codagnone M, Dinan TG, Cryan JF. Microbially-derived short-chain fatty acids impact astrocyte gene expression in a sex-specific manner. Brain Behav Immun Health 2021; 16:100318. [PMID: 34589808 PMCID: PMC8474187 DOI: 10.1016/j.bbih.2021.100318] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 07/28/2021] [Accepted: 08/03/2021] [Indexed: 01/02/2023] Open
Abstract
Recent investigations in neuroscience implicate the role of microbial-derived metabolites, such as short-chain fatty acids (SCFAs) in brain health and disease. The SCFAs acetate, propionate and butyrate have pleiotropic effects within the nervous system. They are crucial for the maturation of the brain's innate immune cells, the microglia, and modulate other glial cells through the aryl-hydrocarbon receptor. Investigations in preclinical and clinical models find that SCFAs exert neuroprotective and antidepressant affects, while also modulating the stress response and satiety. However, many investigations thus far have not assessed the impact of sex on SCFA activity. Our novel investigation tested the impact of physiologically relevant doses of SCFAs on male and female primary cortical astrocytes. We find that butyrate (0–25 μM) correlates with increased Bdnf and Pgc1-α expression, implicating histone-deacetylase inhibitor pathways. Intriguingly, this effect is only seen in females. We also find that acetate (0–1500 μM) correlates with increased Ahr and Gfap expression in males only, suggesting immune modulatory pathways. In males, propionate (0–35 μM) correlates with increased Il-22 expression, further suggesting immunomodulatory actions. These findings show a novel sex-dependent impact of acetate and butyrate, but not propionate on astrocyte gene expression.
Collapse
Affiliation(s)
- Simon Spichak
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland.,APC Microbiome Institute, University College Cork, Cork, Ireland
| | - Francisco Donoso
- APC Microbiome Institute, University College Cork, Cork, Ireland.,Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland
| | - Gerard M Moloney
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland.,APC Microbiome Institute, University College Cork, Cork, Ireland
| | - Eoin Gunnigle
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland.,APC Microbiome Institute, University College Cork, Cork, Ireland
| | - Jillian M Brown
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
| | - Martin Codagnone
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland.,APC Microbiome Institute, University College Cork, Cork, Ireland
| | - Timothy G Dinan
- APC Microbiome Institute, University College Cork, Cork, Ireland.,Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland
| | - John F Cryan
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland.,APC Microbiome Institute, University College Cork, Cork, Ireland
| |
Collapse
|
26
|
Codeluppi SA, Chatterjee D, Prevot TD, Bansal Y, Misquitta KA, Sibille E, Banasr M. Chronic Stress Alters Astrocyte Morphology in Mouse Prefrontal Cortex. Int J Neuropsychopharmacol 2021; 24:842-853. [PMID: 34346493 PMCID: PMC8538896 DOI: 10.1093/ijnp/pyab052] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 06/25/2021] [Accepted: 08/03/2021] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Neuromorphological changes are consistently reported in the prefrontal cortex of patients with stress-related disorders and in rodent stress models, but the effects of stress on astrocyte morphology and the potential link to behavioral deficits are relatively unknown. METHODS To answer these questions, transgenic mice expressing green fluorescent protein (GFP) under the glial fibrillary acid protein (GFAP) promotor were subjected to 7, 21, or 35 days of chronic restraint stress (CRS). CRS-induced behavioral effects on anhedonia- and anxiety-like behaviors were measured using the sucrose intake and the PhenoTyper tests, respectively. Prefrontal cortex GFP+ or GFAP+ cell morphology was assessed using Sholl analysis, and associations with behavior were determined using correlation analysis. RESULTS CRS-exposed male and female mice displayed anxiety-like behavior at 7, 21, and 35 days and anhedonia-like behavior at 35 days. Analysis of GFAP+ cell morphology revealed significant atrophy of distal processes following 21 and 35 days of CRS. CRS induced similar decreases in intersections at distal radii for GFP+ cells accompanied by increased proximal processes. In males, the number of intersections at the most distal radius step significantly correlated with anhedonia-like behavior (r = 0.622, P < .05) for GFP+ cells and with behavioral emotionality calculated by z-scoring all behavioral measured deficits (r = -0.667, P < .05). Similar but not significant correlations were observed in females. No correlation between GFP+ cell atrophy with anxiety-like behavior was found. CONCLUSION Chronic stress exposure induces a progressive atrophy of cortical astroglial cells, potentially contributing to maladaptive neuroplastic and behavioral changes associated with stress-related disorders.
Collapse
Affiliation(s)
- Sierra A Codeluppi
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health (CAMH), Toronto, Canada,Department of Pharmacology and Toxicology, University of Toronto, Toronto, Canada
| | - Dipashree Chatterjee
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health (CAMH), Toronto, Canada,Department of Pharmacology and Toxicology, University of Toronto, Toronto, Canada
| | - Thomas D Prevot
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health (CAMH), Toronto, Canada,Department of Psychiatry, University of Toronto, Toronto, Canada
| | - Yashika Bansal
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health (CAMH), Toronto, Canada
| | - Keith A Misquitta
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health (CAMH), Toronto, Canada,Department of Pharmacology and Toxicology, University of Toronto, Toronto, Canada
| | - Etienne Sibille
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health (CAMH), Toronto, Canada,Department of Pharmacology and Toxicology, University of Toronto, Toronto, Canada,Department of Psychiatry, University of Toronto, Toronto, Canada
| | - Mounira Banasr
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health (CAMH), Toronto, Canada,Department of Pharmacology and Toxicology, University of Toronto, Toronto, Canada,Department of Psychiatry, University of Toronto, Toronto, Canada,Correspondence: Mounira Banasr, PhD, CAMH, 250 College Street, Toronto, ON M5T 1R8, Canada ()
| |
Collapse
|
27
|
Zaman V, Shields DC, Shams R, Drasites KP, Matzelle D, Haque A, Banik NL. Cellular and molecular pathophysiology in the progression of Parkinson's disease. Metab Brain Dis 2021; 36:815-827. [PMID: 33599945 PMCID: PMC8170715 DOI: 10.1007/s11011-021-00689-5] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Accepted: 02/10/2021] [Indexed: 12/12/2022]
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder etiologically linked to the loss of substantia nigra (SN) dopaminergic neurons in the mid-brain. The etiopathology of sporadic PD is still unclear; however, the interaction of extrinsic and intrinsic factors may play a critical role in the onset and progression of the disease. Studies in animal models and human post-mortem tissue have identified distinct cellular and molecular changes in the diseased brain, suggesting complex interactions between different glial cell types and various molecular pathways. Small changes in the expression of specific genes in a single pathway or cell type possibly influence others at the cellular and system levels. These molecular and cellular signatures like neuroinflammation, oxidative stress, and autophagy have been observed in PD patients' brain tissue. While the etiopathology of PD is still poorly understood, the interplay between glial cells and molecular events may play a crucial role in disease onset and progression.
Collapse
Affiliation(s)
- Vandana Zaman
- Ralph H. Johnson Veterans Administration Medical Center, 109 Bee St, Charleston, SC, 29401, USA
- Department of Neurosurgery, Medical University of South Carolina, 96 Jonathan Lucas St., Suite 301, Charleston, SC, 29425, USA
| | - Donald C Shields
- Department of Neurosurgery, Medical University of South Carolina, 96 Jonathan Lucas St., Suite 301, Charleston, SC, 29425, USA
| | - Ramsha Shams
- Department of Neurosurgery, Medical University of South Carolina, 96 Jonathan Lucas St., Suite 301, Charleston, SC, 29425, USA
- Department of Health and Human Performance, The Citadel, 171 Moultrie St, Charleston, SC, 29409, USA
- Department of Microbiology and Immunology, Medical University of South Carolina, 173 Ashley Avenue, Charleston, SC, 29425, USA
| | - Kelsey P Drasites
- Department of Neurosurgery, Medical University of South Carolina, 96 Jonathan Lucas St., Suite 301, Charleston, SC, 29425, USA
- Department of Health and Human Performance, The Citadel, 171 Moultrie St, Charleston, SC, 29409, USA
- Department of Microbiology and Immunology, Medical University of South Carolina, 173 Ashley Avenue, Charleston, SC, 29425, USA
| | - Denise Matzelle
- Ralph H. Johnson Veterans Administration Medical Center, 109 Bee St, Charleston, SC, 29401, USA
- Department of Microbiology and Immunology, Medical University of South Carolina, 173 Ashley Avenue, Charleston, SC, 29425, USA
| | - Azizul Haque
- Department of Microbiology and Immunology, Medical University of South Carolina, 173 Ashley Avenue, Charleston, SC, 29425, USA.
| | - Narendra L Banik
- Ralph H. Johnson Veterans Administration Medical Center, 109 Bee St, Charleston, SC, 29401, USA.
- Department of Neurosurgery, Medical University of South Carolina, 96 Jonathan Lucas St., Suite 301, Charleston, SC, 29425, USA.
- Department of Microbiology and Immunology, Medical University of South Carolina, 173 Ashley Avenue, Charleston, SC, 29425, USA.
| |
Collapse
|
28
|
Reddy DS, Thompson W, Calderara G. Molecular mechanisms of sex differences in epilepsy and seizure susceptibility in chemical, genetic and acquired epileptogenesis. Neurosci Lett 2021; 750:135753. [PMID: 33610673 PMCID: PMC7994197 DOI: 10.1016/j.neulet.2021.135753] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Revised: 02/03/2021] [Accepted: 02/14/2021] [Indexed: 02/07/2023]
Abstract
This article provides a succinct overview of sex differences in epilepsy and putative molecular mechanisms underlying sex differences in seizure susceptibility in chemical, genetic, and acquired epileptogenesis. The susceptibility to excitability episodes and occurrence of epileptic seizures are generally higher in men than women. The precise molecular mechanisms remain unclear, but differences in regional morphology and neural circuits in men and women may explain differential vulnerability to seizures and epileptogenic cascades. Changes in seizure sensitivity can be attributed to steroid hormones, including fluctuations in neurosteroids as well as neuroplasticity in their receptor signaling systems. Other potential neurobiological bases for sex differences in epilepsies include differences in brain development, neurogenesis, neuronal chloride homeostasis, and neurotrophic and glial responses. In catamenial epilepsy, a gender-specific neuroendocrine condition, epileptic seizures are most often clustered around a specific menstrual period in adult women. A deeper understanding of the molecular and neural network basis of sex differences in seizures and response to antiepileptic drugs is highly warranted for designing effective, sex-specific therapies for epilepsy, epileptogenesis, and seizure disorders.
Collapse
Affiliation(s)
- Doodipala Samba Reddy
- Department of Neuroscience and Experimental Therapeutics, Texas A&M University College of Medicine, Bryan, TX, United States.
| | - Wesley Thompson
- Department of Neuroscience and Experimental Therapeutics, Texas A&M University College of Medicine, Bryan, TX, United States
| | - Gianmarco Calderara
- Department of Neuroscience and Experimental Therapeutics, Texas A&M University College of Medicine, Bryan, TX, United States
| |
Collapse
|
29
|
Nwachukwu KN, Evans WA, Sides TR, Trevisani CP, Davis A, Marshall SA. Chemogenetic manipulation of astrocytic signaling in the basolateral amygdala reduces binge-like alcohol consumption in male mice. J Neurosci Res 2021; 99:1957-1972. [PMID: 33844860 DOI: 10.1002/jnr.24841] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Accepted: 03/21/2021] [Indexed: 12/18/2022]
Abstract
Binge drinking is a common occurrence in the United States, but a high concentration of alcohol in the blood has been shown to have reinforcing and reciprocal effects on the neuroimmune system in both dependent and non-dependent scenarios. The first part of this study examined alcohol's effects on the astrocytic response in the central amygdala and basolateral amygdala (BLA) in a non-dependent model. C57BL/6J mice were given access to either ethanol, water, or sucrose during a "drinking in the dark" paradigm, and astrocyte number and astrogliosis were measured using immunohistochemistry. Results indicate that non-dependent consumption increased glial fibrillary acidic protein (GFAP) density but not the number of GFAP+ cells, suggesting that non-dependent ethanol is sufficient to elicit astrocyte activation. The second part of this study examined how astrocytes impacted behaviors and the neurochemistry related to alcohol using the chemogenetic tool, DREADDs (designer receptors exclusively activated by designer drugs). Transgenic GFAP-hM3Dq mice were administered clozapine N-oxide both peripherally, affecting the entire central nervous system (CNS), or directly into the BLA. In both instances, GFAP-Gq-signaling activation significantly reduced ethanol consumption and correlating blood ethanol concentrations. However, GFAP-Gq-DREADD activation throughout the CNS had more broad effects resulting in decreased locomotor activity and sucrose consumption. More targeted GFAP-Gq-signaling activation in the BLA only impacted ethanol consumption. Finally, a glutamate assay revealed that after GFAP-Gq-signaling activation glutamate concentrations in the amygdala were partially normalized to control levels. Altogether, these studies support the theory that astrocytes represent a viable target for alcohol use disorder therapies.
Collapse
Affiliation(s)
- Kala N Nwachukwu
- Department of Biological & Biomedical Sciences, North Carolina Central University, Durham, NC, USA
| | - William A Evans
- Department of Basic Pharmaceutical Sciences, Fred P. Wilson School of Pharmacy, High Point University, High Point, NC, USA
| | - Tori R Sides
- Department of Biological & Biomedical Sciences, North Carolina Central University, Durham, NC, USA
| | - Christopher P Trevisani
- Department of Basic Pharmaceutical Sciences, Fred P. Wilson School of Pharmacy, High Point University, High Point, NC, USA
| | - Ambryia Davis
- Department of Basic Pharmaceutical Sciences, Fred P. Wilson School of Pharmacy, High Point University, High Point, NC, USA
| | - S Alex Marshall
- Department of Biological & Biomedical Sciences, North Carolina Central University, Durham, NC, USA.,Department of Basic Pharmaceutical Sciences, Fred P. Wilson School of Pharmacy, High Point University, High Point, NC, USA.,Department of Psychology & Neuroscience, University of North Carolina-Chapel Hill, Chapel Hill, NC, USA
| |
Collapse
|
30
|
Taylor WW, Imhoff BR, Sathi ZS, Liu WY, Garza KM, Dias BG. Contributions of glucocorticoid receptors in cortical astrocytes to memory recall. Learn Mem 2021; 28:126-133. [PMID: 33723032 PMCID: PMC7970741 DOI: 10.1101/lm.053041.120] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Accepted: 01/14/2021] [Indexed: 01/15/2023]
Abstract
Dysfunctions in memory recall lead to pathological fear; a hallmark of trauma-related disorders, like posttraumatic stress disorder (PTSD). Both, heightened recall of an association between a cue and trauma, as well as impoverished recall that a previously trauma-related cue is no longer a threat, result in a debilitating fear toward the cue. Glucocorticoid-mediated action via the glucocorticoid receptor (GR) influences memory recall. This literature has primarily focused on GRs expressed in neurons or ignored cell-type specific contributions. To ask how GR action in nonneuronal cells influences memory recall, we combined auditory fear conditioning in mice and the knockout of GRs in astrocytes in the prefrontal cortex (PFC), a brain region implicated in memory recall. We found that knocking out GRs in astrocytes of the PFC disrupted memory recall. Specifically, we found that knocking out GRs in astrocytes in the PFC (AstroGRKO) after fear conditioning resulted in higher levels of freezing to the CS+ tone when compared with controls (AstroGRintact). While we did not find any differences in extinction of fear toward the CS+ between these groups, AstroGRKO female but not male mice showed impaired recall of extinction training. These results suggest that GRs in cortical astrocytes contribute to memory recall. These data demonstrate the need to examine GR action in cortical astrocytes to elucidate the basic neurobiology underlying memory recall and potential mechanisms that underlie female-specific biases in the incidence of PTSD.
Collapse
Affiliation(s)
- William W Taylor
- Neuroscience Graduate Program, Emory University, Atlanta, Georgia 30322, USA
- Neuroscience Graduate Program, University of Southern California, Los Angeles, California 90007, USA
- Developmental Neuroscience and Neurogenetics Program, Division of Research on Children, Youth, and Families, The Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, California 90027, USA
| | - Barry R Imhoff
- Division of Behavioral Neuroscience and Psychiatric Disorders, Yerkes National Primate Research Center, Atlanta, Georgia 30322, USA
| | - Zakia Sultana Sathi
- Division of Behavioral Neuroscience and Psychiatric Disorders, Yerkes National Primate Research Center, Atlanta, Georgia 30322, USA
| | - Wei Y Liu
- Developmental Neuroscience and Neurogenetics Program, Division of Research on Children, Youth, and Families, The Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, California 90027, USA
| | - Kristie M Garza
- Neuroscience Graduate Program, Emory University, Atlanta, Georgia 30322, USA
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia 30322, USA
| | - Brian G Dias
- Neuroscience Graduate Program, Emory University, Atlanta, Georgia 30322, USA
- Neuroscience Graduate Program, University of Southern California, Los Angeles, California 90007, USA
- Developmental Neuroscience and Neurogenetics Program, Division of Research on Children, Youth, and Families, The Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, California 90027, USA
- Division of Behavioral Neuroscience and Psychiatric Disorders, Yerkes National Primate Research Center, Atlanta, Georgia 30322, USA
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, Georgia 30322, USA
- Department of Pediatrics, Keck School of Medicine, University of Southern California, Los Angeles, California 90027, USA
| |
Collapse
|
31
|
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
|
32
|
Gobbo DR, Pereira LDS, Ferreira JGP, de Castro Horta-Junior JA, Bittencourt JC, Sá SI. Effects of ovariectomy on the inputs from the medial nucleus of the amygdala to the ventromedial nucleus of the hypothalamus in young adult rats. Neurosci Lett 2021; 746:135657. [PMID: 33482312 DOI: 10.1016/j.neulet.2021.135657] [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: 12/09/2020] [Accepted: 01/12/2021] [Indexed: 10/22/2022]
Abstract
During puberty, sexual hormones induce crucial changes in neural circuit organization, leading to significant sexual dimorphism in adult behaviours. The ventrolateral division of the ventromedial nucleus of the hypothalamus (VMHvl) is the major neural site controlling the receptive component of female sexual behaviour, which is dependent on ovarian hormones. The inputs to the VMHvl, originating from the medial nucleus of the amygdala (MeA), transmit essential information to trigger such behaviour. In this study, we investigated the projection pattern of the MeA to the VMHvl in ovariectomized rats at early puberty. Six-week-old Sprague-Dawley rats were ovariectomized (OVX) and, upon reaching 90 days of age, were subjected to iontophoretic injections of the neuronal anterograde tracer Phaseolus vulgaris leucoagglutinin into the MeA. Projections from the MeA to the VMHvl and to other structures included in the neural circuit responsible for female sexual behaviour were analysed in the Control and OVX groups. The results of the semi-quantitative analysis showed that peripubertal ovariectomy reduced the density of intra-amygdalar fibres. The stereological estimates, however, failed to find changes in the organization of the terminal fields of nerve fibres from the MeA to the VMHvl in the adult. The present data show that ovariectomized rats during the peripubertal phase did not undergo significant changes in MeA fibres reaching the VMHvl; however, they suggest a possible effect of ovariectomy on MeA connectivity under amygdalar subnuclei.
Collapse
Affiliation(s)
- Denise Ribeiro Gobbo
- Universidade de Sao Paulo, Instituto de Ciencias Biomedicas, Laboratorio de Neuroanatomia Quimica, Departamento de Anatomia, Sao Paulo, SP, Brazil
| | - Lais da Silva Pereira
- Universidade de Sao Paulo, Instituto de Ciencias Biomedicas, Laboratorio de Neuroanatomia Quimica, Departamento de Anatomia, Sao Paulo, SP, Brazil
| | - Jozélia Gomes Pacheco Ferreira
- Universidade de Sao Paulo, Instituto de Ciencias Biomedicas, Laboratorio de Neuroanatomia Quimica, Departamento de Anatomia, Sao Paulo, SP, Brazil
| | | | - Jackson Cioni Bittencourt
- Universidade de Sao Paulo, Instituto de Ciencias Biomedicas, Laboratorio de Neuroanatomia Quimica, Departamento de Anatomia, Sao Paulo, SP, Brazil; Universidade de Sao Paulo, Instituto de Psicologia, Nucleo de Neurociências e Comportamento, São Paulo, SP, Brazil.
| | - Susana Isabel Sá
- Department of Biomedicine, Unit of Anatomy, Faculty of Medicine, University of Porto, Porto, Portugal; CINTESIS, Center for Health Technology and Services Research, Faculty of Medicine, University of Porto, Porto, Portugal.
| |
Collapse
|
33
|
Dudek KA, Dion‐Albert L, Kaufmann FN, Tuck E, Lebel M, Menard C. Neurobiology of resilience in depression: immune and vascular insights from human and animal studies. Eur J Neurosci 2021; 53:183-221. [PMID: 31421056 PMCID: PMC7891571 DOI: 10.1111/ejn.14547] [Citation(s) in RCA: 79] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Revised: 07/22/2019] [Accepted: 08/12/2019] [Indexed: 12/12/2022]
Abstract
Major depressive disorder (MDD) is a chronic and recurrent psychiatric condition characterized by depressed mood, social isolation and anhedonia. It will affect 20% of individuals with considerable economic impacts. Unfortunately, 30-50% of depressed individuals are resistant to current antidepressant treatments. MDD is twice as prevalent in women and associated symptoms are different. Depression's main environmental risk factor is chronic stress, and women report higher levels of stress in daily life. However, not every stressed individual becomes depressed, highlighting the need to identify biological determinants of stress vulnerability but also resilience. Based on a reverse translational approach, rodent models of depression were developed to study the mechanisms underlying susceptibility vs resilience. Indeed, a subpopulation of animals can display coping mechanisms and a set of biological alterations leading to stress resilience. The aetiology of MDD is multifactorial and involves several physiological systems. Exacerbation of endocrine and immune responses from both innate and adaptive systems are observed in depressed individuals and mice exhibiting depression-like behaviours. Increasing attention has been given to neurovascular health since higher prevalence of cardiovascular diseases is found in MDD patients and inflammatory conditions are associated with depression, treatment resistance and relapse. Here, we provide an overview of endocrine, immune and vascular changes associated with stress vulnerability vs. resilience in rodents and when available, in humans. Lack of treatment efficacy suggests that neuron-centric treatments do not address important causal biological factors and better understanding of stress-induced adaptations, including sex differences, could contribute to develop novel therapeutic strategies including personalized medicine approaches.
Collapse
Affiliation(s)
- Katarzyna A. Dudek
- Department of Psychiatry and NeuroscienceFaculty of Medicine and CERVO Brain Research CenterUniversité LavalQuebec CityQCCanada
| | - Laurence Dion‐Albert
- Department of Psychiatry and NeuroscienceFaculty of Medicine and CERVO Brain Research CenterUniversité LavalQuebec CityQCCanada
| | - Fernanda Neutzling Kaufmann
- Department of Psychiatry and NeuroscienceFaculty of Medicine and CERVO Brain Research CenterUniversité LavalQuebec CityQCCanada
| | - Ellen Tuck
- Smurfit Institute of GeneticsTrinity CollegeDublinIreland
| | - Manon Lebel
- Department of Psychiatry and NeuroscienceFaculty of Medicine and CERVO Brain Research CenterUniversité LavalQuebec CityQCCanada
| | - Caroline Menard
- Department of Psychiatry and NeuroscienceFaculty of Medicine and CERVO Brain Research CenterUniversité LavalQuebec CityQCCanada
| |
Collapse
|
34
|
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
|
35
|
Christian CA, Reddy DS, Maguire J, Forcelli PA. Sex Differences in the Epilepsies and Associated Comorbidities: Implications for Use and Development of Pharmacotherapies. Pharmacol Rev 2020; 72:767-800. [PMID: 32817274 PMCID: PMC7495340 DOI: 10.1124/pr.119.017392] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The epilepsies are common neurologic disorders characterized by spontaneous recurrent seizures. Boys, girls, men, and women of all ages are affected by epilepsy and, in many cases, by associated comorbidities as well. The primary courses of treatment are pharmacological, dietary, and/or surgical, depending on several factors, including the areas of the brain affected and the severity of the epilepsy. There is a growing appreciation that sex differences in underlying brain function and in the neurobiology of epilepsy are important factors that should be accounted for in the design and development of new therapies. In this review, we discuss the current knowledge on sex differences in epilepsy and associated comorbidities, with emphasis on those aspects most informative for the development of new pharmacotherapies. Particular focus is placed on sex differences in the prevalence and presentation of various focal and generalized epilepsies; psychiatric, cognitive, and physiologic comorbidities; catamenial epilepsy in women; sex differences in brain development; the neural actions of sex and stress hormones and their metabolites; and cellular mechanisms, including brain-derived neurotrophic factor signaling and neuronal-glial interactions. Further attention placed on potential sex differences in epilepsies, comorbidities, and drug effects will enhance therapeutic options and efficacy for all patients with epilepsy. SIGNIFICANCE STATEMENT: Epilepsy is a common neurological disorder that often presents together with various comorbidities. The features of epilepsy and seizure activity as well as comorbid afflictions can vary between men and women. In this review, we discuss sex differences in types of epilepsies, associated comorbidities, pathophysiological mechanisms, and antiepileptic drug efficacy in both clinical patient populations and preclinical animal models.
Collapse
Affiliation(s)
- Catherine A Christian
- Department of Molecular and Integrative Physiology, Neuroscience Program, and Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, Illinois (C.A.C.); Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M University Health Science Center, Bryan, Texas (D.S.R.); Neuroscience Department, Tufts University School of Medicine, Boston, Massachusetts (J.M.); and Departments of Pharmacology and Physiology and Neuroscience, Georgetown University, Washington, D.C. (P.A.F.)
| | - Doodipala Samba Reddy
- Department of Molecular and Integrative Physiology, Neuroscience Program, and Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, Illinois (C.A.C.); Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M University Health Science Center, Bryan, Texas (D.S.R.); Neuroscience Department, Tufts University School of Medicine, Boston, Massachusetts (J.M.); and Departments of Pharmacology and Physiology and Neuroscience, Georgetown University, Washington, D.C. (P.A.F.)
| | - Jamie Maguire
- Department of Molecular and Integrative Physiology, Neuroscience Program, and Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, Illinois (C.A.C.); Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M University Health Science Center, Bryan, Texas (D.S.R.); Neuroscience Department, Tufts University School of Medicine, Boston, Massachusetts (J.M.); and Departments of Pharmacology and Physiology and Neuroscience, Georgetown University, Washington, D.C. (P.A.F.)
| | - Patrick A Forcelli
- Department of Molecular and Integrative Physiology, Neuroscience Program, and Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, Illinois (C.A.C.); Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M University Health Science Center, Bryan, Texas (D.S.R.); Neuroscience Department, Tufts University School of Medicine, Boston, Massachusetts (J.M.); and Departments of Pharmacology and Physiology and Neuroscience, Georgetown University, Washington, D.C. (P.A.F.)
| |
Collapse
|
36
|
Corain L, Grisan E, Graïc JM, Carvajal-Schiaffino R, Cozzi B, Peruffo A. Multi-aspect testing and ranking inference to quantify dimorphism in the cytoarchitecture of cerebellum of male, female and intersex individuals: a model applied to bovine brains. Brain Struct Funct 2020; 225:2669-2688. [PMID: 32989472 PMCID: PMC7674367 DOI: 10.1007/s00429-020-02147-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Accepted: 09/08/2020] [Indexed: 11/28/2022]
Abstract
The dimorphism among male, female and freemartin intersex bovines, focusing on the vermal lobules VIII and IX, was analyzed using a novel data analytics approach to quantify morphometric differences in the cytoarchitecture of digitalized sections of the cerebellum. This methodology consists of multivariate and multi-aspect testing for cytoarchitecture-ranking, based on neuronal cell complexity among populations defined by factors, such as sex, age or pathology. In this context, we computed a set of shape descriptors of the neural cell morphology, categorized them into three domains named size, regularity and density, respectively. The output and results of our methodology are multivariate in nature, allowing an in-depth analysis of the cytoarchitectonic organization and morphology of cells. Interestingly, the Purkinje neurons and the underlying granule cells revealed the same morphological pattern: female possessed larger, denser and more irregular neurons than males. In the Freemartin, Purkinje neurons showed an intermediate setting between males and females, while the granule cells were the largest, most regular and dense. This methodology could be a powerful instrument to carry out morphometric analysis providing robust bases for objective tissue screening, especially in the field of neurodegenerative pathologies.
Collapse
Affiliation(s)
- L Corain
- Department of Management and Engineering, University of Padova, 36100, Vicenza, VI, Italy
| | - E Grisan
- Department of Information Engineering, University of Padova, 35131, Padua, PD, Italy
- School of Engineering, London South Bank University, London, SE1 0AA, UK
| | - J-M Graïc
- Department of Comparative Biomedicine and Food Science, University of Padova, Viale dell'Università 16, 35020, Legnaro, PD, Italy.
| | - R Carvajal-Schiaffino
- Department of Mathematics and Computer Science, University of Santiago de Chile, Santiago, Chile
| | - B Cozzi
- Department of Comparative Biomedicine and Food Science, University of Padova, Viale dell'Università 16, 35020, Legnaro, PD, Italy
| | - A Peruffo
- Department of Comparative Biomedicine and Food Science, University of Padova, Viale dell'Università 16, 35020, Legnaro, PD, Italy
| |
Collapse
|
37
|
O'Leary LA, Davoli MA, Belliveau C, Tanti A, Ma JC, Farmer WT, Turecki G, Murai KK, Mechawar N. Characterization of Vimentin-Immunoreactive Astrocytes in the Human Brain. Front Neuroanat 2020; 14:31. [PMID: 32848635 PMCID: PMC7406576 DOI: 10.3389/fnana.2020.00031] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Accepted: 05/26/2020] [Indexed: 12/19/2022] Open
Abstract
Astrocytes are commonly identified by their expression of the intermediate filament protein glial fibrillary acidic protein (GFAP). GFAP-immunoreactive (GFAP-IR) astrocytes exhibit regional heterogeneity in density and morphology in the mouse brain as well as morphological diversity in the human cortex. However, regional variations in astrocyte distribution and morphology remain to be assessed comprehensively. This was the overarching objective of this postmortem study, which mainly exploited the immunolabeling of vimentin (VIM), an intermediate filament protein expressed by astrocytes and endothelial cells which presents the advantage of more extensively labeling cell structures. We compared the densities of vimentin-immunoreactive (VIM-IR) and GFAP-IR astrocytes in various brain regions (prefrontal and primary visual cortex, caudate nucleus, mediodorsal thalamus) from male individuals having died suddenly in the absence of neurological or psychiatric conditions. The morphometric properties of VIM-IR in these brain regions were also assessed. We found that VIM-IR astrocytes generally express the canonical astrocytic markers Aldh1L1 and GFAP but that VIM-IR astrocytes are less abundant than GFAP-IR astrocytes in all human brain regions, particularly in the thalamus, where VIM-IR cells were nearly absent. About 20% of all VIM-IR astrocytes presented a twin cell morphology, a phenomenon rarely observed for GFAP-IR astrocytes. Furthermore VIM-IR astrocytes in the striatum were often seen to extend numerous parallel processes which seemed to give rise to large VIM-IR fiber bundles projecting over long distances. Moreover, morphometric analyses revealed that VIM-IR astrocytes were more complex than their mouse counterparts in functionally homologous brain regions, as has been previously reported for GFAP-IR astrocytes. Lastly, the density of GFAP-IR astrocytes in gray and white matter were inversely correlated with vascular density, but for VIM-IR astrocytes this was only the case in gray matter, suggesting that gliovascular interactions may especially influence the regional heterogeneity of GFAP-IR astrocytes. Taken together, these findings reveal special features displayed uniquely by human VIM-IR astrocytes and illustrate that astrocytes display important region- and marker-specific differences in the healthy human brain.
Collapse
Affiliation(s)
- Liam Anuj O'Leary
- McGill Group for Suicide Studies, Douglas Mental Health University Institute, Verdun, QC, Canada.,Integrated Program in Neuroscience, McGill University, Montreal, QC, Canada
| | - Maria Antonietta Davoli
- McGill Group for Suicide Studies, Douglas Mental Health University Institute, Verdun, QC, Canada
| | - Claudia Belliveau
- McGill Group for Suicide Studies, Douglas Mental Health University Institute, Verdun, QC, Canada.,Integrated Program in Neuroscience, McGill University, Montreal, QC, Canada
| | - Arnaud Tanti
- McGill Group for Suicide Studies, Douglas Mental Health University Institute, Verdun, QC, Canada
| | - Jie Christopher Ma
- McGill Group for Suicide Studies, Douglas Mental Health University Institute, Verdun, QC, Canada
| | - William Todd Farmer
- Centre for Research in Neuroscience, Department of Neurology and Neurosurgery, Brain Repair and Integrative Neuroscience Program, The Research Institute of the McGill University Health Center, Montreal General Hospital, Montreal, QC, Canada
| | - Gustavo Turecki
- McGill Group for Suicide Studies, Douglas Mental Health University Institute, Verdun, QC, Canada.,Integrated Program in Neuroscience, McGill University, Montreal, QC, Canada.,Department of Psychiatry, McGill University, Montreal, QC, Canada
| | - Keith Kazuo Murai
- Centre for Research in Neuroscience, Department of Neurology and Neurosurgery, Brain Repair and Integrative Neuroscience Program, The Research Institute of the McGill University Health Center, Montreal General Hospital, Montreal, QC, Canada
| | - Naguib Mechawar
- McGill Group for Suicide Studies, Douglas Mental Health University Institute, Verdun, QC, Canada.,Integrated Program in Neuroscience, McGill University, Montreal, QC, Canada.,Department of Psychiatry, McGill University, Montreal, QC, Canada
| |
Collapse
|
38
|
Allen HN, Bobnar HJ, Kolber BJ. Left and right hemispheric lateralization of the amygdala in pain. Prog Neurobiol 2020; 196:101891. [PMID: 32730859 DOI: 10.1016/j.pneurobio.2020.101891] [Citation(s) in RCA: 86] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 05/29/2020] [Accepted: 07/22/2020] [Indexed: 02/04/2023]
Abstract
Hemispheric asymmetries within the brain have been identified across taxa and have been extensively studied since the early 19th century. Here, we discuss lateralization of a brain structure, the amygdala, and how this lateralization is reshaping how we understand the role of the amygdala in pain processing. The amygdala is an almond-shaped, bilateral brain structure located within the limbic system. Historically, the amygdala was known to have a role in the processing of emotions and attaching emotional valence to memories and other experiences. The amygdala has been extensively studied in fear conditioning and affect but recently has been shown to have an important role in processing noxious information and impacting pain. The amygdala is composed of multiple nuclei; of special interest is the central nucleus of the amygdala (CeA). The CeA receives direct nociceptive inputs from the parabrachial nucleus (PBN) through the spino-parabrachio-amygdaloid pathway as well as more highly processed cortical and thalamic input via the lateral and basolateral amygdala. Although the amygdala is a bilateral brain region, most data investigating the amygdala's role in pain have been generated from the right CeA, which has an overwhelmingly pro-nociceptive function across pain models. The left CeA has often been characterized to have no effect on pain modulation, a dampened pro-nociceptive function, or most recently an anti-nociceptive function. This review explores the current literature on CeA lateralization and the hemispheres' respective roles in the processing and modulation of different forms of pain.
Collapse
Affiliation(s)
- Heather N Allen
- Department of Biological Sciences and Chronic Pain Research Consortium, Duquesne University, Pittsburgh, PA, 15282, United States
| | - Harley J Bobnar
- Department of Biological Sciences and Chronic Pain Research Consortium, Duquesne University, Pittsburgh, PA, 15282, United States
| | - Benedict J Kolber
- Department of Biological Sciences and Chronic Pain Research Consortium, Duquesne University, Pittsburgh, PA, 15282, United States; Department of Neuroscience and Center for Advanced Pain Studies, The University of Texas at Dallas, Richardson, TX, 75080, United States.
| |
Collapse
|
39
|
Sex Differences in Biophysical Signatures across Molecularly Defined Medial Amygdala Neuronal Subpopulations. eNeuro 2020; 7:ENEURO.0035-20.2020. [PMID: 32493755 PMCID: PMC7333980 DOI: 10.1523/eneuro.0035-20.2020] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Accepted: 04/20/2020] [Indexed: 12/29/2022] Open
Abstract
The medial amygdala (MeA) is essential for processing innate social and non-social behaviors, such as territorial aggression and mating, which display in a sex-specific manner. While sex differences in cell numbers and neuronal morphology in the MeA are well established, if and how these differences extend to the biophysical level remain unknown. Our previous studies revealed that expression of the transcription factors, Dbx1 and Foxp2, during embryogenesis defines separate progenitor pools destined to generate different subclasses of MEA inhibitory output neurons. We have also previously shown that Dbx1-lineage and Foxp2-lineage neurons display different responses to innate olfactory cues and in a sex-specific manner. To examine whether these neurons also possess sex-specific biophysical signatures, we conducted a multidimensional analysis of the intrinsic electrophysiological profiles of these transcription factor defined neurons in the male and female MeA. We observed striking differences in the action potential (AP) spiking patterns across lineages, and across sex within each lineage, properties known to be modified by different voltage-gated ion channels. To identify the potential mechanism underlying the observed lineage-specific and sex-specific differences in spiking adaptation, we conducted a phase plot analysis to narrow down putative ion channel candidates. Of these candidates, we found a subset expressed in a lineage-biased and/or sex-biased manner. Thus, our results uncover neuronal subpopulation and sex differences in the biophysical signatures of developmentally defined MeA output neurons, providing a potential physiological substrate for how the male and female MeA may process social and non-social cues that trigger innate behavioral responses.
Collapse
|
40
|
Microglial and Astrocytic Function in Physiological and Pathological Conditions: Estrogenic Modulation. Int J Mol Sci 2020; 21:ijms21093219. [PMID: 32370112 PMCID: PMC7247358 DOI: 10.3390/ijms21093219] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 04/24/2020] [Accepted: 04/30/2020] [Indexed: 12/20/2022] Open
Abstract
There are sexual differences in the onset, prevalence, and outcome of numerous neurological diseases. Thus, in Alzheimer’s disease, multiple sclerosis, and major depression disorder, the incidence in women is higher than in men. In contrast, men are more likely to present other pathologies, such as amyotrophic lateral sclerosis, Parkinson’s disease, and autism spectrum. Although the neurological contribution to these diseases has classically always been studied, the truth is that neurons are not the only cells to be affected, and there are other cells, such as glial cells, that are also involved and could be key to understanding the development of these pathologies. Sexual differences exist not only in pathology but also in physiological processes, which shows how cells are differentially regulated in males and females. One of the reasons these sexual differences may occur could be due to the different action of sex hormones. Many studies have shown an increase in aromatase levels in the brain, which could indicate the main role of estrogens in modulating proinflammatory processes. This review will highlight data about sex differences in glial physiology and how estrogenic compounds, such as estradiol and tibolone, could be used as treatment in neurological diseases due to their anti-inflammatory effects and the ability to modulate glial cell functions.
Collapse
|
41
|
Vegeto E, Villa A, Della Torre S, Crippa V, Rusmini P, Cristofani R, Galbiati M, Maggi A, Poletti A. The Role of Sex and Sex Hormones in Neurodegenerative Diseases. Endocr Rev 2020; 41:5572525. [PMID: 31544208 PMCID: PMC7156855 DOI: 10.1210/endrev/bnz005] [Citation(s) in RCA: 134] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Accepted: 09/20/2019] [Indexed: 12/11/2022]
Abstract
Neurodegenerative diseases (NDs) are a wide class of disorders of the central nervous system (CNS) with unknown etiology. Several factors were hypothesized to be involved in the pathogenesis of these diseases, including genetic and environmental factors. Many of these diseases show a sex prevalence and sex steroids were shown to have a role in the progression of specific forms of neurodegeneration. Estrogens were reported to be neuroprotective through their action on cognate nuclear and membrane receptors, while adverse effects of male hormones have been described on neuronal cells, although some data also suggest neuroprotective activities. The response of the CNS to sex steroids is a complex and integrated process that depends on (i) the type and amount of the cognate steroid receptor and (ii) the target cell type-either neurons, glia, or microglia. Moreover, the levels of sex steroids in the CNS fluctuate due to gonadal activities and to local metabolism and synthesis. Importantly, biochemical processes involved in the pathogenesis of NDs are increasingly being recognized as different between the two sexes and as influenced by sex steroids. The aim of this review is to present current state-of-the-art understanding on the potential role of sex steroids and their receptors on the onset and progression of major neurodegenerative disorders, namely, Alzheimer's disease, Parkinson's diseases, amyotrophic lateral sclerosis, and the peculiar motoneuron disease spinal and bulbar muscular atrophy, in which hormonal therapy is potentially useful as disease modifier.
Collapse
Affiliation(s)
- Elisabetta Vegeto
- Center of Excellence on Neurodegenerative Diseases, Università degli Studi di Milano, Italy.,Dipartimento di Scienze Farmaceutiche (DiSFarm), Università degli Studi di Milano, Italy
| | - Alessandro Villa
- Center of Excellence on Neurodegenerative Diseases, Università degli Studi di Milano, Italy.,Dipartimento di Scienze della Salute (DiSS), Università degli Studi di Milano, Italy
| | - Sara Della Torre
- Center of Excellence on Neurodegenerative Diseases, Università degli Studi di Milano, Italy.,Dipartimento di Scienze Farmaceutiche (DiSFarm), Università degli Studi di Milano, Italy
| | - Valeria Crippa
- Center of Excellence on Neurodegenerative Diseases, Università degli Studi di Milano, Italy.,Dipartimento di Eccellenza di Scienze Farmacologiche e Biomolecolari (DiSFeB), Università degli Studi di Milano, Italy
| | - Paola Rusmini
- Center of Excellence on Neurodegenerative Diseases, Università degli Studi di Milano, Italy.,Dipartimento di Eccellenza di Scienze Farmacologiche e Biomolecolari (DiSFeB), Università degli Studi di Milano, Italy
| | - Riccardo Cristofani
- Center of Excellence on Neurodegenerative Diseases, Università degli Studi di Milano, Italy.,Dipartimento di Eccellenza di Scienze Farmacologiche e Biomolecolari (DiSFeB), Università degli Studi di Milano, Italy
| | - Mariarita Galbiati
- Center of Excellence on Neurodegenerative Diseases, Università degli Studi di Milano, Italy.,Dipartimento di Eccellenza di Scienze Farmacologiche e Biomolecolari (DiSFeB), Università degli Studi di Milano, Italy
| | - Adriana Maggi
- Center of Excellence on Neurodegenerative Diseases, Università degli Studi di Milano, Italy.,Dipartimento di Scienze Farmaceutiche (DiSFarm), Università degli Studi di Milano, Italy
| | - Angelo Poletti
- Center of Excellence on Neurodegenerative Diseases, Università degli Studi di Milano, Italy.,Dipartimento di Eccellenza di Scienze Farmacologiche e Biomolecolari (DiSFeB), Università degli Studi di Milano, Italy
| |
Collapse
|
42
|
Kim GS, Uddin M. Sex-specific and shared expression profiles of vulnerability and resilience to trauma in brain and blood. Biol Sex Differ 2020; 11:13. [PMID: 32228684 PMCID: PMC7106761 DOI: 10.1186/s13293-020-00288-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Accepted: 03/03/2020] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND While post-traumatic stress disorder (PTSD) is defined by behavioral/cognitive symptoms most directly relevant to brain function, it can be considered a systemic disorder characterized by a distinct inability to reinstate homeostasis after trauma. METHODS In this study, we conducted a secondary analysis of gene expression profiles in key PTSD-relevant tissues, namely blood, amygdala, and hippocampus, from a rat model of PTSD, to identify sex-specific and shared processes associated with individual differences in response to recent trauma exposure. RESULTS Our findings suggest both shared and sex-specific mechanisms underlying individual differences associated with vulnerability and resilience to trauma in hippocampus, amygdala, and blood. By disentangling cell composition from transcriptional changes, we found higher proportions of hippocampal oligodendrocytes in the PTSD-like, extreme behavioral response (EBR) group for both sexes and also identified modules for transcriptional activity associated with group differences (i.e., response to trauma) in the hippocampus that appeared to be sex-specific. By contrast, we found prominent sex differences, but no group differences, in amygdalar cell composition, and both shared and sex-specific modules representing PTSD-relevant transcriptional activity in the amygdala. Across amygdala and hippocampus, both sex-specific and shared processes were relevant to an overarching framework for EBR implicating disrupted TNFα/NFκΒ signaling and excitatory/inhibitory imbalance in dysregulated synaptic/structural plasticity with important implications for fear learning and memory. Our main finding in peripheral blood was consistent with the human literature and identified wound healing processes and hemostasis to be upregulated in the resilient, minimal behavioral response (MBR) group across sexes, but disrupted in a sexually dimorphic manner in the EBR group. CONCLUSION In contrast to the varied characterization of the PTSD-like EBR group, characterization of MBR across blood, amygdala, and hippocampus suggests a common theme of upregulated wound healing and extracellular matrix (ECM) remodeling shared between sexes. In all, we identified differential oligodendrocyte proportions in hippocampus between PTSD-like EBR and resilient MBR, and identified processes and pathways that characterize the EBR and MBR-associated transcriptional changes across hippocampus, amygdala, and blood. The sex-specific mechanisms involved in EBR may contribute to the pronounced disparity in risk for PTSD, with women much more likely to develop PTSD.
Collapse
Affiliation(s)
- Grace S Kim
- Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, IL, USA.,Medical Scholars Program, University of Illinois College of Medicine at Urbana-Champaign, Urbana, IL, USA
| | - Monica Uddin
- Genomics Program, Center for Global Health and Infectious Disease Research, College of Public Health, University of South Florida, 3720 Spectrum Blvd., Ste. 304, Tampa, FL, 33612, USA.
| |
Collapse
|
43
|
Contu L, Nizari S, Heath CJ, Hawkes CA. Pre- and Post-natal High Fat Feeding Differentially Affects the Structure and Integrity of the Neurovascular Unit of 16-Month Old Male and Female Mice. Front Neurosci 2019; 13:1045. [PMID: 31632236 PMCID: PMC6783577 DOI: 10.3389/fnins.2019.01045] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Accepted: 09/17/2019] [Indexed: 01/20/2023] Open
Abstract
Compelling experimental and clinical evidence supports a role for maternal obesity in offspring health. Adult children of obese mothers are at greater risk of obesity, diabetes, coronary heart disease and stroke. These offspring may also be at greater risk of age-related neurodegenerative diseases for which mid-life obesity is a risk factor. Rodent diet-induced obesity models have shown that high fat (HF) diet consumption damages the integrity of the blood–brain barrier (BBB) in the adult brain. However, there is currently little information about the effect of chronic HF feeding on the BBB of aged animals. Moreover, the long-term consequences of maternal obesity on the cerebrovasculature of aged offspring are not known. This study determined the impact of pre- and post-natal HF diet on the structure and integrity of cerebral blood vessels in aged male and female mice. Female C57Bl/6 mice were fed either a 10% fat control (C) or 45% HF diet before mating and during gestation and lactation. At weaning, male and female offspring were fed the C or HF diet until sacrifice at 16-months of age. Both dams and offspring fed the HF diet weighed significantly more than mice fed the C diet. Post-natal HF diet exposure increased hippocampal BBB leakiness in female offspring, in association with loss of astrocyte endfoot coverage of arteries. Markers of tight junctions, pericytes or smooth muscle cells were not altered by pre- or post-natal HF diet. Male offspring born to HF-fed mothers showed decreased parenchymal GFAP expression compared to offspring of mothers fed C diet, while microglial and macrophage markers were higher in the same female diet group. In addition, female offspring exposed to the HF diet for their entire lifespan showed more significant changes in vessel structure, BBB permeability and inflammation compared to male animals. These results suggest that the long-term impact of prenatal HF diet on the integrity of cerebral blood vessels differs between male and female offspring depending on the post-natal diet. This may have implications for the prevention and management of age- and obesity-related cerebrovascular diseases that differentially affect men and women.
Collapse
Affiliation(s)
- Laura Contu
- School of Life, Health and Chemical Sciences, Faculty of Science, Technology, Engineering and Mathematics, The Open University, Milton Keynes, United Kingdom
| | - Shereen Nizari
- School of Life, Health and Chemical Sciences, Faculty of Science, Technology, Engineering and Mathematics, The Open University, Milton Keynes, United Kingdom
| | - Christopher J Heath
- School of Life, Health and Chemical Sciences, Faculty of Science, Technology, Engineering and Mathematics, The Open University, Milton Keynes, United Kingdom
| | - Cheryl A Hawkes
- School of Life, Health and Chemical Sciences, Faculty of Science, Technology, Engineering and Mathematics, The Open University, Milton Keynes, United Kingdom
| |
Collapse
|
44
|
Jia C, Brown RW, Malone HM, Burgess KC, Gill DW, Keasey MP, Hagg T. Ciliary neurotrophic factor is a key sex-specific regulator of depressive-like behavior in mice. Psychoneuroendocrinology 2019; 100:96-105. [PMID: 30299260 PMCID: PMC6333501 DOI: 10.1016/j.psyneuen.2018.09.038] [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: 05/22/2018] [Revised: 08/23/2018] [Accepted: 09/26/2018] [Indexed: 10/28/2022]
Abstract
Ciliary neurotrophic factor (CNTF) is produced by astrocytes and promotes neurogenesis and neuroprotection. Little is known about the role of CNTF in affective behavior. We investigated whether CNTF affects depressive- and anxiety-like behavior in adult mice as tested in the forced swim, sucrose preference and elevated-T maze tests. Female wild type CNTF+/+ mice more readily developed behavioral despair with increased immobility time and decreased latency to immobility in the forced swim test than male CNTF+/+ littermates. The lack of CNTF in CNTF-/- mice had an opposite effect on depressive-like behavior in female mice (reduced immobility time and increased sucrose preference) vs. male mice (increased immobility time). Female wildtype mice expressed more CNTF in the amygdala than male mice. Ovariectomy increased CNTF expression, as well as immobility time, which was significantly reduced in CNTF-/- mice, suggesting that CNTF mediates overiectomy-induced immobility time, possibly in the amygdala. Progesterone but not 17-β estradiol inhibited CNTF expression in cultured C6 astroglioma cells. Progesterone treatment also reduced CNTF expression in the amygdala and decreased immobility time in female CNTF+/+ but not in CNTF-/- mice. Castration did not alter CNTF expression in males nor their behavior. Lastly, there were no effects of CNTF on the elevated T-maze, a behavioral test of anxiety, suggesting that a different mechanism may underlie anxiety-like behavior. This study reveals a novel CNTF-mediated mechanism in stress-induced depressive-like behavior and points to opportunities for sex-specific treatments for depression, e.g. progesterone in females and CNTF-stimulating drugs in males.
Collapse
Affiliation(s)
- Cuihong Jia
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, 37614, USA.
| | | | | | | | | | | | | |
Collapse
|
45
|
Villa A, Della Torre S, Maggi A. Sexual differentiation of microglia. Front Neuroendocrinol 2019; 52:156-164. [PMID: 30481522 DOI: 10.1016/j.yfrne.2018.11.003] [Citation(s) in RCA: 76] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Revised: 11/07/2018] [Accepted: 11/24/2018] [Indexed: 12/28/2022]
Abstract
Sex plays a role in the incidence and outcome of neurological illnesses, also influencing the response to treatments. Despite sexual differentiation of the brain has been extensively investigated, the study of sex differences in microglia, the brain's resident immune cells, has been largely neglected until recently. To fulfill this gap, our laboratory developed several tools, including cellular and animal models, which bolstered in-depth studies on sexual differentiation of microglia and its impact on brain physiology, as well as on the onset and progression of neurological disorders. Here, we summarize the current status of knowledge on the sex-dependent function of microglia, and report recent evidence linking these cells to the sexual bias in the susceptibility to neurological brain diseases.
Collapse
Affiliation(s)
- Alessandro Villa
- Center of Excellence on Neurodegenerative Diseases and Dept of Pharmacological and Biomolecular Sciences, University of Milan, via Balzaretti, 9, Milan, Italy
| | - Sara Della Torre
- Center of Excellence on Neurodegenerative Diseases and Dept of Pharmacological and Biomolecular Sciences, University of Milan, via Balzaretti, 9, Milan, Italy
| | - Adriana Maggi
- Center of Excellence on Neurodegenerative Diseases and Dept of Pharmacological and Biomolecular Sciences, University of Milan, via Balzaretti, 9, Milan, Italy.
| |
Collapse
|
46
|
Abstract
Astrocytes have historically been considered structural supporting cells for neurons. Thanks to new molecular tools, allowing specific cell ablation or over-expression of genes, new unexpected astrocytic functions have recently been unveiled. This review focus on emerging groundbreaking findings showing that hypothalamic astrocytes are pivotal for the regulation of whole body energy homeostasis. Hypothalamic astrocytes sense glucose and fatty acids, and express receptors for several peripheral hormones such as leptin and insulin. Furthermore, they display striking sexual dimorphism which may account, at least partially, for gender specific differences in energy homeostasis. Metabolic alterations have been shown to influence the initiation and progression of many neurodegenerative disorders. A better understanding of the roles and interplay between the different brain cells in regulating energy homeostasis could help develop new therapeutic strategies to prevent or cure neurodegenerative disorders.
Collapse
Affiliation(s)
- Simonetta Camandola
- Laboratory of Neurosciences, National Institute on Aging, Intramural Research Program, Baltimore, MD, USA
| |
Collapse
|
47
|
Rainville JR, Tsyglakova M, Hodes GE. Deciphering sex differences in the immune system and depression. Front Neuroendocrinol 2018; 50:67-90. [PMID: 29288680 DOI: 10.1016/j.yfrne.2017.12.004] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Revised: 12/21/2017] [Accepted: 12/22/2017] [Indexed: 02/07/2023]
Abstract
Certain mood disorders and autoimmune diseases are predominately female diseases but we do not know why. Here, we explore the relationship between depression and the immune system from a sex-based perspective. This review characterizes sex differences in the immune system in health and disease. We explore the contribution of gonadal and stress hormones to immune function at the cellular and molecular level in the brain and body. We propose hormonal and genetic sex specific immune mechanisms that may contribute to the etiology of mood disorders.
Collapse
Affiliation(s)
- Jennifer R Rainville
- Department of Neuroscience, Virginia Polytechnic Institute and State University, 1981 Kraft Drive, Blacksburg, VA 24060, USA
| | - Mariya Tsyglakova
- Department of Neuroscience, Virginia Polytechnic Institute and State University, 1981 Kraft Drive, Blacksburg, VA 24060, USA; Translational Biology, Medicine, and Health, Virginia Polytechnic Institute and State University, 1 Riverside Circle, Roanoke, VA 24016, USA
| | - Georgia E Hodes
- Department of Neuroscience, Virginia Polytechnic Institute and State University, 1981 Kraft Drive, Blacksburg, VA 24060, USA.
| |
Collapse
|
48
|
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
|
49
|
Hirsch MM, Brusco J, Vaccaro T, Margis R, Moreira JE, Gottfried C, Rasia-Filho AA. Sex Differences and Estrous Cycle Changes in Synaptic Plasticity-related microRNA in the Rat Medial Amygdala. Neuroscience 2018; 379:405-414. [DOI: 10.1016/j.neuroscience.2018.03.035] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2017] [Revised: 02/20/2018] [Accepted: 03/21/2018] [Indexed: 01/05/2023]
|
50
|
Abstract
Astrocytes are neural cells of ectodermal, neuroepithelial origin that provide for homeostasis and defense of the central nervous system (CNS). Astrocytes are highly heterogeneous in morphological appearance; they express a multitude of receptors, channels, and membrane transporters. This complement underlies their remarkable adaptive plasticity that defines the functional maintenance of the CNS in development and aging. Astrocytes are tightly integrated into neural networks and act within the context of neural tissue; astrocytes control homeostasis of the CNS at all levels of organization from molecular to the whole organ.
Collapse
Affiliation(s)
- Alexei Verkhratsky
- The University of Manchester , Manchester , United Kingdom ; Achúcarro Basque Center for Neuroscience, IKERBASQUE, Basque Foundation for Science , Bilbao , Spain ; Department of Neuroscience, University of the Basque Country UPV/EHU and CIBERNED, Leioa, Spain ; Center for Basic and Translational Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen , Copenhagen , Denmark ; and Center for Translational Neuromedicine, University of Rochester Medical Center , Rochester, New York
| | - Maiken Nedergaard
- The University of Manchester , Manchester , United Kingdom ; Achúcarro Basque Center for Neuroscience, IKERBASQUE, Basque Foundation for Science , Bilbao , Spain ; Department of Neuroscience, University of the Basque Country UPV/EHU and CIBERNED, Leioa, Spain ; Center for Basic and Translational Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen , Copenhagen , Denmark ; and Center for Translational Neuromedicine, University of Rochester Medical Center , Rochester, New York
| |
Collapse
|