1
|
Weston KP, Gunelson AM, Maloney SE, Ge X, Stelzer JA, Kim KS, Collier S, Mindt MM, Agajanian MJ, Major MB, Goldfarb D, Noguchi KK, Yi JJ. The gain-of-function UBE3A Q588E variant causes Angelman-like neurodevelopmental phenotypes in mice. Sci Rep 2025; 15:9152. [PMID: 40097479 PMCID: PMC11914044 DOI: 10.1038/s41598-025-92511-w] [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: 09/03/2024] [Accepted: 02/27/2025] [Indexed: 03/19/2025] Open
Abstract
Mutations in the E3 ubiquitin ligase UBE3A that cause enzymatic gain-of-function result in disease phenotypes which differ from classic Angelman syndrome. However, these phenotypes are highly heterogeneous raising questions about the mechanistic basis of such phenotypic diversity. Here, we characterize a mouse model harboring a Ube3aQ606E gain of function variant (UBE3AQ588E in humans). Extensive behavioral phenotyping showed that animals possessing a maternally inherited mutation (Ube3amQ606E) paradoxically show many behavioral deficits indicative of overall UBE3A loss-of-function. These included pronounced motor deficits, hypoactivity, and reduced stereotypic behaviors. Moreover, brain weights and MRI analysis revealed global microcephaly with a postnatal onset, consistent with phenotypes described in Angelman syndrome model mice. Additional biochemical analyses demonstrated an increased abundance of UBE3A substrates in brain tissue and immunofluorescence analyses showed that microcephaly is not caused by increased apoptotic cell death. Together, our results strongly suggest a novel mechanism by which the Ube3amQ606E mutation leads to enhanced self-targeted degradation of UBE3A, leading to an overall loss of enzyme activity, resulting in Angelman-like phenotypes in vivo.
Collapse
Affiliation(s)
- Kellan P Weston
- Department of Neuroscience, Washington University School of Medicine, St. Louis, MO, 63110, USA
- COMBINEDBrain, Brentwood, TN, 37027, USA
| | - Anna M Gunelson
- Department of Neuroscience, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Susan E Maloney
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, 63110, USA
- Intellectual and Developmental Disabilities Research Center, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Xia Ge
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Jalin A Stelzer
- Department of Neuroscience, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Kwang-Soo Kim
- Department of Neuroscience, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Shylyn Collier
- Department of Neuroscience, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Marissa M Mindt
- Department of Neuroscience, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Megan J Agajanian
- Department of Cell Biology, Washington University School of Medicine, St. Louis, MO, 63110, USA
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Michael B Major
- Department of Cell Biology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Dennis Goldfarb
- Department of Cell Biology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Kevin K Noguchi
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Jason J Yi
- Department of Neuroscience, Washington University School of Medicine, St. Louis, MO, 63110, USA.
- Intellectual and Developmental Disabilities Research Center, Washington University School of Medicine, St. Louis, MO, 63110, USA.
| |
Collapse
|
2
|
Kaplan AA, Öztürk G, Bay S, Keskin İ. Investigating Neotenic and Metamorphic Axolotl Brain Complexity: A Stereological and Immunohistochemical Perspective. J Comp Neurol 2025; 533:e70031. [PMID: 40109227 PMCID: PMC11923732 DOI: 10.1002/cne.70031] [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: 08/22/2024] [Revised: 12/31/2024] [Accepted: 02/10/2025] [Indexed: 03/22/2025]
Abstract
The ability of certain tetrapods, such as amphibians, to regenerate complex structures, such as organs or limbs, is well-established, though this capacity varies significantly across species, with humans exhibiting limited regenerative potential. Ependymoglia cells in the ventricular region of the brain are known to exhibit proliferative properties during homeostasis and damage and to perform stem cell functions. This study investigated changes occurring in neurons and glia in the central nervous system following metamorphosis in axolotls. Morphological alterations in brain tissue, newly formed neurons, and cellular organizations in different brain regions were assessed using stereological and immunohistochemical methods, as well as light and electron microscopy. Interestingly, we observe no statistically significant difference in total neuron numbers in the telencephalon region between neotenic and metamorphic axolotls. However, the proliferation index and the numbers of cells expressing NeuN were significantly higher in metamorphic axolotls. Furthermore, structural changes in neuronal nuclei and myelin sheath organization were determined at the light and electron microscopic levels post-metamorphosis. Ultrastructural analyses revealed a change in chromatin organization from euchromatic to heterochromatic in neurons after metamorphosis, and morphological changes were also demonstrated in myelinated nerve fibers in the telencephalon. Additionally, mucopolysaccharide-containing secretory sacs were also identified on the apical surfaces of a subgroup of ependymoglia cells located in the lateral ventricle wall. Overall, this study sheds useful light on the intricate changes occurring in the central nervous system during metamorphosis in axolotls and provides valuable insights into the mechanisms underlying these processes.
Collapse
Affiliation(s)
- Arife Ahsen Kaplan
- Department of Histology and Embryology, The School of Medicine, Istanbul Medipol University, Istanbul, Turkey
- Regenerative and Restorative Medicine Research Center (REMER), Institute for Health Sciences and Technologies (SABITA), İstanbul Medipol University, Istanbul, Turkey
| | - Gürkan Öztürk
- Regenerative and Restorative Medicine Research Center (REMER), Institute for Health Sciences and Technologies (SABITA), İstanbul Medipol University, Istanbul, Turkey
- Department of Physiology, School of Medicine, Bolu Abant İzzet Baysal Üniversitesi, Bolu, Turkey
| | - Sadık Bay
- Regenerative and Restorative Medicine Research Center (REMER), Institute for Health Sciences and Technologies (SABITA), İstanbul Medipol University, Istanbul, Turkey
| | - İlknur Keskin
- Department of Histology and Embryology, The School of Medicine, Istanbul Medipol University, Istanbul, Turkey
- Regenerative and Restorative Medicine Research Center (REMER), Institute for Health Sciences and Technologies (SABITA), İstanbul Medipol University, Istanbul, Turkey
| |
Collapse
|
3
|
Gray JM, Major K, Castillo-Ruiz A, Shipley M, Gangappa S, Forger NG. The inflammatory response to birth requires MyD88 and is driven by both mother and offspring. Brain Behav Immun 2024; 115:617-630. [PMID: 37967662 PMCID: PMC11913035 DOI: 10.1016/j.bbi.2023.11.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 10/15/2023] [Accepted: 11/11/2023] [Indexed: 11/17/2023] Open
Abstract
Birth is an inflammatory event for the newborn, characterized by elevations in interleukin (IL)-6, IL-10, and tumor necrosis factor (TNF)-α peripherally and/or centrally, as well as changes in brain microglia. However, the mechanism(s) underlying these responses is unknown. Toll-like receptors (TLRs) play crucial roles in innate immunity and initiate inflammatory cascades upon recognition of endogenous or exogenous antigens. Most TLR signaling depends on the adaptor molecule myeloid differentiation primary response 88 (MyD88). We independently varied MyD88 gene status in mouse dams and their offspring to determine whether the inflammatory response to birth depends on MyD88 signaling and, if so, whether that signaling occurs in the offspring, the mother, or both. We find that the perinatal surges in plasma IL-6 and brain expression of TNF-α depend solely on MyD88 gene status of the offspring, whereas postnatal increases in plasma IL-10 and TNF-α depend on MyD88 in both the pup and dam. Interestingly, MyD88 genotype of the dam primarily drives differences in offspring brain microglial density and has robust effects on developmental neuronal cell death. Milk cytokines were evaluated as a possible source of postnatal maternal influence; although we found high levels of CXCL1/GROα and several other cytokines in ingested post-partum milk, their presence did not require MyD88. Thus, the inflammatory response previously described in the late-term fetus and newborn depends on MyD88 (and, by extension, TLRs), with signaling in both the dam and offspring contributing. Unexpectedly, naturally-occuring neuronal cell death in the newborn is modulated primarily by maternal MyD88 gene status.
Collapse
Affiliation(s)
- Jennifer M Gray
- Neuroscience Institute, Georgia State University, Atlanta, GA 30303, USA
| | - Kharli Major
- Neuroscience Institute, Georgia State University, Atlanta, GA 30303, USA
| | | | - Michael Shipley
- Influenza Division, Centers for Disease Control and Prevention, Atlanta, GA 30329, USA
| | - Shivaprakash Gangappa
- Influenza Division, Centers for Disease Control and Prevention, Atlanta, GA 30329, USA
| | - Nancy G Forger
- Neuroscience Institute, Georgia State University, Atlanta, GA 30303, USA.
| |
Collapse
|
4
|
Morrison V, Houpert M, Trapani J, Brockman A, Kingsley P, Katdare K, Layden H, Nguena-Jones G, Trevisan A, Maguire-Zeiss K, Marnett L, Bix G, Ihrie R, Carter B. Jedi-1/MEGF12-mediated phagocytosis controls the pro-neurogenic properties of microglia in the ventricular-subventricular zone. Cell Rep 2023; 42:113423. [PMID: 37952151 PMCID: PMC10842823 DOI: 10.1016/j.celrep.2023.113423] [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: 03/08/2023] [Revised: 10/03/2023] [Accepted: 10/25/2023] [Indexed: 11/14/2023] Open
Abstract
Microglia are the primary phagocytes in the central nervous system and clear dead cells generated during development or disease. The phagocytic process shapes the microglia phenotype, which affects the local environment. A unique population of microglia resides in the ventricular-subventricular zone (V-SVZ) of neonatal mice, but how they influence the neurogenic niche is not well understood. Here, we demonstrate that phagocytosis contributes to a pro-neurogenic microglial phenotype in the V-SVZ and that these microglia phagocytose apoptotic cells via the engulfment receptor Jedi-1. Deletion of Jedi-1 decreases apoptotic cell clearance, triggering a neuroinflammatory microglia phenotype that resembles dysfunctional microglia in neurodegeneration and aging and that reduces neural precursor proliferation via elevated interleukin-1β signaling; interleukin-1 receptor inhibition rescues precursor proliferation in vivo. Together, these results reveal a critical role for Jedi-1 in connecting microglial phagocytic activity to the maintenance of a pro-neurogenic phenotype in the developing V-SVZ.
Collapse
Affiliation(s)
- Vivianne Morrison
- Department of Biochemistry, Vanderbilt University, Nashville, TN 37232, USA; Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN 37235, USA; Department of Cell and Molecular Biology, Tulane University, New Orleans, LA 70118, USA
| | - Matthew Houpert
- Department of Biochemistry, Vanderbilt University, Nashville, TN 37232, USA; Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN 37235, USA
| | - Jonathan Trapani
- Department of Biochemistry, Vanderbilt University, Nashville, TN 37232, USA; Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN 37235, USA
| | - Asa Brockman
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN 37235, USA; Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN 37235, USA
| | - Philip Kingsley
- Department of Biochemistry, Vanderbilt University, Nashville, TN 37232, USA
| | - Ketaki Katdare
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN 37235, USA
| | - Hillary Layden
- Department of Biochemistry, Vanderbilt University, Nashville, TN 37232, USA
| | - Gabriela Nguena-Jones
- Department of Biochemistry, Vanderbilt University, Nashville, TN 37232, USA; Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN 37235, USA
| | - Alexandra Trevisan
- Department of Biochemistry, Vanderbilt University, Nashville, TN 37232, USA; Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | | | - Lawrence Marnett
- Department of Biochemistry, Vanderbilt University, Nashville, TN 37232, USA; Department of Chemistry, Vanderbilt University, Nashville, TN 37235, USA; Department of Pharmacology, Vanderbilt University, Nashville, TN 37235, USA; A.B. Hancock Jr. Memorial Laboratory for Cancer Research, Vanderbilt-Ingram Cancer Center, Nashville, TN 37232, USA
| | - Gregory Bix
- Center for Clinical Neuroscience Research, Tulane University, New Orleans, LA 70118, USA
| | - Rebecca Ihrie
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN 37235, USA; Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN 37235, USA
| | - Bruce Carter
- Department of Biochemistry, Vanderbilt University, Nashville, TN 37232, USA; Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN 37235, USA.
| |
Collapse
|
5
|
Rurak GM, Gahelrasoul A, Aguilar-Valles A, Salmaso N. Neonatal estrogen induces male-like expression of astroglial markers of maturation and plasticity in the neocortex of female mice. Brain Res 2023; 1818:148499. [PMID: 37499732 DOI: 10.1016/j.brainres.2023.148499] [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: 04/18/2023] [Revised: 06/29/2023] [Accepted: 07/21/2023] [Indexed: 07/29/2023]
Abstract
Astroglia play a crucial role in various aspects of neurodevelopment including building, maintaining, and modulating neuronal circuits that underly complex behaviours in the neocortex. Telencephalic regions exhibit sex differences in neuronal networks that arise early in development. Astroglia express receptors for gonadal hormones responsible for the organization of sex differences, such as estrogen, placing them in a key position to modulate sex differences in the development of neuronal networks. Astroglial cells express specific proteins related to their morphology, function, and maturation. We have previously shown that P7-P14 is a key transition period for neocortical astroglial maturation and that males reach a mature phenotype earlier than females, at P7. In this study, we investigated whether administration of perinatal estradiol to female mice is sufficient to masculinize astroglial protein and gene expression related to maturation that we previously observed at P7. We found that canonical astroglial markers like glial fibrillary acidic protein and glutamine synthetase are not affected by perinatal estrogen, but markers of astroglial maturation, Vimentin, Aldh1a1, Dio2, and the number of actively dividing astroglia are masculinized by perinatal estradiol administration. These findings suggest that sex differences in neocortical astroglial maturation are at least in-part due to the role of perinatal estrogen. Given the higher prevalence of neurodevelopmental disorders in males compared to females and the involvement of astroglia in virtually all neurodevelopmental disorders, further research is needed to determine other contributions to sex differences in neocortical astroglial cells.
Collapse
Affiliation(s)
- G M Rurak
- Department of Neuroscience, Carleton University, Ottawa, Ontario, Canada.
| | - A Gahelrasoul
- Department of Neuroscience, Carleton University, Ottawa, Ontario, Canada.
| | - A Aguilar-Valles
- Department of Neuroscience, Carleton University, Ottawa, Ontario, Canada.
| | - N Salmaso
- Department of Neuroscience, Carleton University, Ottawa, Ontario, Canada.
| |
Collapse
|
6
|
Trofimova I. Anticipatory attractors, functional neurochemistry and "Throw & Catch" mechanisms as illustrations of constructivism. Rev Neurosci 2023; 34:737-762. [PMID: 36584323 DOI: 10.1515/revneuro-2022-0120] [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: 10/01/2022] [Accepted: 12/07/2022] [Indexed: 12/31/2022]
Abstract
This review explores several rarely discussed examples illustrating constructivism principles, generative and selective features of neuronal regulation of behaviour. First, the review highlights Walter Freeman's experiments and mathematical analysis that uncovered the existence of anticipatory attractors, i.e. non-random dynamical patterns in neurodynamics. Since Freeman's work did not extend to neurochemistry, this paper then points to the proposed earlier neurochemical framework summarizing the managerial roles of monoaminergic, cholinergic and opioid receptor systems likely contributing to anticipatory attractors in line with functional constructivism. As a third example, neurochemistry's evidence points to the "Throw & Catch" (T&C) principle in neurodynamics. This principle refers to the pro-active, neurochemically expensive, massive but topical increase of potentials ("Throw") within electrodynamics and neurotransmission in the brain whenever there is an uncertainty in selection of degrees of freedom (DFs). The T&C also underlines the relay-like processes during the selection of DFs. The "Throw" works as an internally generated "flashlight" that, contrarily to the expectations of entropy reduction, increases entropy and variance observed in processes related to orientation and action-formation. The discussed examples highlight the deficiency of structures-oriented projects and excitation-inhibition concepts in neuroscience. The neural regulation of behaviour appears to be a fluid, constructive process, constantly upgrading the choice of behavioural DFs, to ensure the compatibility between the environmental and individual's individuals' needs and capacities.
Collapse
Affiliation(s)
- Irina Trofimova
- Laboratory of Collective Intelligence, Department of Psychiatry and Behavioural Neurosciences, McMaster University, Hamilton L8S 2T6, ON, Canada
| |
Collapse
|
7
|
Carmona-Alcocer V, Brown LS, Anchan A, Rohr KE, Evans JA. Developmental patterning of peptide transcription in the central circadian clock in both sexes. Front Neurosci 2023; 17:1177458. [PMID: 37274219 PMCID: PMC10235759 DOI: 10.3389/fnins.2023.1177458] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Accepted: 04/21/2023] [Indexed: 06/06/2023] Open
Abstract
Introduction Neuropeptide signaling modulates the function of central clock neurons in the suprachiasmatic nucleus (SCN) during development and adulthood. Arginine vasopressin (AVP) and vasoactive intestinal peptide (VIP) are expressed early in SCN development, but the precise timing of transcriptional onset has been difficult to establish due to age-related changes in the rhythmic expression of each peptide. Methods To provide insight into spatial patterning of peptide transcription during SCN development, we used a transgenic approach to define the onset of Avp and Vip transcription. Avp-Cre or Vip-Cre males were crossed to Ai9+/+ females, producing offspring in which the fluorescent protein tdTomato (tdT) is expressed at the onset of Avp or Vip transcription. Spatial patterning of Avp-tdT and Vip-tdT expression was examined at critical developmental time points spanning mid-embryonic age to adulthood in both sexes. Results We find that Avp-tdT and Vip-tdT expression is initiated at different developmental time points in spatial subclusters of SCN neurons, with developmental patterning that differs by sex. Conclusions These data suggest that SCN neurons can be distinguished into further subtypes based on the developmental patterning of neuropeptide expression, which may contribute to regional and/or sex differences in cellular function in adulthood.
Collapse
Affiliation(s)
- Vania Carmona-Alcocer
- Department of Biomedical Science, Marquette University, Milwaukee, WI, United States
| | - Lindsey S. Brown
- Harvard John A. Paulson School of Engineering and Applied Sciences, Harvard University, Allston, MA, United States
| | - Aiesha Anchan
- Department of Biomedical Science, Marquette University, Milwaukee, WI, United States
| | - Kayla E. Rohr
- Department of Biomedical Science, Marquette University, Milwaukee, WI, United States
| | - Jennifer A. Evans
- Department of Biomedical Science, Marquette University, Milwaukee, WI, United States
| |
Collapse
|
8
|
Cortes LR, Forger NG. DNA methylation and demethylation shape sexual differentiation of neurochemical phenotype. Horm Behav 2023; 151:105349. [PMID: 37001316 PMCID: PMC10133097 DOI: 10.1016/j.yhbeh.2023.105349] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 02/24/2023] [Accepted: 03/13/2023] [Indexed: 04/28/2023]
Abstract
Some of the best-studied neural sex differences depend on differential cell death in males and females, but other sex differences persist even if cell death is prevented. These include sex differences in neurochemical phenotype (i.e., stable patterns of gene expression). Work in our laboratory over the last several years has tested the hypothesis that sex differences in DNA methylation early in life underlie sexual differentiation of neuronal phenotype. We have shown that 1) expression of enzymes that place or remove DNA methylation marks is greatest during the first week of life in the mouse brain and overlaps with the perinatal critical period of sexual differentiation; 2) a transient inhibition of DNA methylation during neonatal life abolishes several sex differences in cell phenotype in the mouse hypothalamus; 3) both DNA methylation and de-methylation contribute to the development of neural sex differences; and 4) the effects of DNA methylation and de-methylation are brain region- and cell type-specific.
Collapse
Affiliation(s)
- L R Cortes
- Department of Integrative Biology & Physiology, University of California Los Angeles, Los Angeles, CA, USA
| | - N G Forger
- Neuroscience Institute, Georgia State University, Atlanta, GA, USA.
| |
Collapse
|
9
|
Morrison VE, Houpert MG, Trapani JB, Brockman AA, Kingsley PJ, Katdare KA, Layden HM, Nguena-Jones G, Trevisan AJ, Maguire-Zeiss KA, Marnett LJ, Bix GJ, Ihrie RA, Carter BD. Jedi-1/MEGF12-mediated phagocytosis controls the pro-neurogenic properties of microglia in the ventricular-subventricular zone. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.03.531012. [PMID: 36945622 PMCID: PMC10028845 DOI: 10.1101/2023.03.03.531012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/09/2023]
Abstract
Microglia are the primary phagocytes in the central nervous system and are responsible for clearing dead cells generated during development or disease. The phagocytic process shapes the phenotype of the microglia, which affects the local environment. A unique population of microglia reside in the ventricular-subventricular zone (V-SVZ) of neonatal mice, but how they influence this neurogenic niche is not well-understood. Here, we demonstrate that phagocytosis creates a pro-neurogenic microglial phenotype in the V-SVZ and that these microglia phagocytose apoptotic cells via the engulfment receptor Jedi-1. Deletion of Jedi-1 decreases apoptotic cell clearance, triggering the development of a neuroinflammatory phenotype, reminiscent of neurodegenerative and-age-associated microglia, that reduces neural precursor proliferation via elevated interleukin (IL)-1β signaling; inhibition of IL-1 receptor rescues precursor proliferation in vivo. Together, these results reveal a critical role for Jedi-1 in connecting microglial phagocytic activity to a phenotype that promotes neurogenesis in the developing V-SVZ.
Collapse
Affiliation(s)
- Vivianne E Morrison
- Vanderbilt University Department of Biochemistry
- Vanderbilt Brain Institute
- Tulane University Center for Clinical Neuroscience Research
| | - Matthew G Houpert
- Vanderbilt University Department of Biochemistry
- Vanderbilt Brain Institute
| | - Jonathan B Trapani
- Vanderbilt University Department of Biochemistry
- Vanderbilt Brain Institute
| | - Asa A Brockman
- Vanderbilt University Department of Cell and Developmental Biology
- Vanderbilt Brain Institute
| | | | | | | | | | - Alexandra J Trevisan
- Vanderbilt University Department of Biochemistry
- St. Jude Children's Research Hospital
| | | | - Lawrence J Marnett
- Vanderbilt University Department of Biochemistry
- Vanderbilt University Department of Chemistry
- Vanderbilt University Department of Pharmacology
- A.B. Hancock Jr. Memorial Laboratory for Cancer Research
| | - Gregory J Bix
- Tulane University Center for Clinical Neuroscience Research
| | - Rebecca A Ihrie
- Vanderbilt University Department of Cell and Developmental Biology
- Vanderbilt Brain Institute
| | - Bruce D Carter
- Vanderbilt University Department of Biochemistry
- Vanderbilt Brain Institute
| |
Collapse
|
10
|
Halladay LR, Herron SM. Lasting impact of postnatal maternal separation on the developing BNST: Lifelong socioemotional consequences. Neuropharmacology 2023; 225:109404. [PMID: 36572178 PMCID: PMC9926961 DOI: 10.1016/j.neuropharm.2022.109404] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Revised: 12/19/2022] [Accepted: 12/22/2022] [Indexed: 12/24/2022]
Abstract
Nearly one percent of children in the US experience childhood neglect or abuse, which can incite lifelong emotional and behavioral disorders. Many studies investigating the neural underpinnings of maleffects inflicted by early life stress have largely focused on dysregulation of the hypothalamic-pituitary-adrenal (HPA) axis. Newer veins of evidence suggest that exposure to early life stressors can interrupt neural development in extrahypothalamic areas as well, including the bed nucleus of the stria terminalis (BNST). One widely used approach in this area is rodent maternal separation (MS), which typically consists of separating pups from the dam for extended periods of time, over several days during the first weeks of postnatal life - a time when pups are highly dependent on maternal care for survival. MS has been shown to incite myriad lasting effects not limited to increased anxiety-like behavior, hyper-responsiveness to stressors, and social behavior deficits. The behavioral effects of MS are widespread and thus unlikely to be limited to hypothalamic mechanisms. Recent work has highlighted the BNST as a critical arbiter of some of the consequences of MS, especially socioemotional behavioral deficits. The BNST is a well-documented modulator of anxiety, reward, and social behavior by way of its connections with hypothalamic and extra-hypothalamic systems. Moreover, during the postnatal period when MS is typically administered, the BNST undergoes critical neural developmental events. This review highlights evidence that MS interferes with neural development to permanently alter BNST circuitry, which may account for a variety of behavioral deficits seen following early life stress. This article is part of the Special Issue on 'Fear, Anxiety and PTSD'.
Collapse
Affiliation(s)
- Lindsay R Halladay
- Department of Psychology, Santa Clara University, 500 El Camino Real, Santa Clara, CA, 95053, USA.
| | - Steven M Herron
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, 300 Pasteur Drive, Stanford, CA, 94305, USA
| |
Collapse
|
11
|
Moreno-Juan V, Aníbal-Martínez M, Herrero-Navarro Á, Valdeolmillos M, Martini FJ, López-Bendito G. Spontaneous Thalamic Activity Modulates the Cortical Innervation of the Primary Visual Nucleus of the Thalamus. Neuroscience 2023; 508:87-97. [PMID: 35878717 DOI: 10.1016/j.neuroscience.2022.07.022] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 07/15/2022] [Accepted: 07/18/2022] [Indexed: 01/17/2023]
Abstract
Sensory processing relies on the correct development of thalamocortical loops. Visual corticothalamic axons (CTAs) invade the dorsolateral geniculate nucleus (dLGN) of the thalamus in early postnatal mice according to a regulated program that includes activity-dependent mechanisms. Spontaneous retinal activity influences the thalamic incursion of CTAs, yet the perinatal thalamus also generates intrinsic patterns of spontaneous activity whose role in modulating afferent connectivity remains unknown. Here, we found that patterned spontaneous activity in the dLGN contributes to proper spatial and temporal innervation of CTAs. Disrupting patterned spontaneous activity in the dLGN delays corticogeniculate innervation under normal conditions and upon eye enucleation. The delayed innervation was evident throughout the first two postnatal weeks but resumes after eye-opening, suggesting that visual experience is necessary for the homeostatic recovery of corticogeniculate innervation.
Collapse
Affiliation(s)
- Verónica Moreno-Juan
- Instituto de Neurociencias de Alicante, Universidad Miguel Hernández-Consejo Superior de Investigaciones Científicas (UMH-CSIC), Sant Joan d'Alacant, Spain
| | - Mar Aníbal-Martínez
- Instituto de Neurociencias de Alicante, Universidad Miguel Hernández-Consejo Superior de Investigaciones Científicas (UMH-CSIC), Sant Joan d'Alacant, Spain
| | - Álvaro Herrero-Navarro
- Instituto de Neurociencias de Alicante, Universidad Miguel Hernández-Consejo Superior de Investigaciones Científicas (UMH-CSIC), Sant Joan d'Alacant, Spain
| | - Miguel Valdeolmillos
- Instituto de Neurociencias de Alicante, Universidad Miguel Hernández-Consejo Superior de Investigaciones Científicas (UMH-CSIC), Sant Joan d'Alacant, Spain
| | - Francisco J Martini
- Instituto de Neurociencias de Alicante, Universidad Miguel Hernández-Consejo Superior de Investigaciones Científicas (UMH-CSIC), Sant Joan d'Alacant, Spain.
| | - Guillermina López-Bendito
- Instituto de Neurociencias de Alicante, Universidad Miguel Hernández-Consejo Superior de Investigaciones Científicas (UMH-CSIC), Sant Joan d'Alacant, Spain.
| |
Collapse
|
12
|
Smiley JF, Bleiwas C, Marino BM, Vaddi P, Canals-Baker S, Wilson DA, Saito M. Estimates of total neuron number show that neonatal ethanol causes immediate and lasting neuron loss in cortical and subcortical areas. Front Neurosci 2023; 17:1186529. [PMID: 37205048 PMCID: PMC10185770 DOI: 10.3389/fnins.2023.1186529] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Accepted: 04/13/2023] [Indexed: 05/21/2023] Open
Abstract
In neonatal brain development there is a period of normal apoptotic cell death that regulates adult neuron number. At approximately the same period, ethanol exposure can cause a dramatic spike in apoptotic cell death. While ethanol-induced apoptosis has been shown to reduce adult neuron number, questions remain about the regional selectivity of the ethanol effect, and whether the brain might have some capacity to overcome the initial neuron loss. The present study used stereological cell counting to compare cumulative neuron loss 8 h after postnatal day 7 (P7) ethanol treatment to that of animals left to mature to adulthood (P70). Across several brain regions we found that the reduction of total neuron number after 8 h was as large as that of adult animals. Comparison between regions revealed that some areas are more vulnerable, with neuron loss in the anterior thalamic nuclei > the medial septum/vertical diagonal band, dorsal subiculum, and dorsal lateral geniculate nucleus > the mammillary bodies and cingulate cortex > whole neocortex. In contrast to estimates of total neuron number, estimates of apoptotic cell number in Nissl-stained sections at 8 h after ethanol treatment provided a less reliable predictor of adult neuron loss. The findings show that ethanol-induced neonatal apoptosis often causes immediate neuron deficits that persist in adulthood, and furthermore suggests that the brain may have limited capacity to compensate for ethanol-induced neuron loss.
Collapse
Affiliation(s)
- John F. Smiley
- Nathan Kline Institute for Psychiatric Research, Orangeburg, NY, United States
- Department of Psychiatry, School of Medicine, New York University, New York, NY, United States
- *Correspondence: John F. Smiley,
| | - Cynthia Bleiwas
- Nathan Kline Institute for Psychiatric Research, Orangeburg, NY, United States
| | - Brandon M. Marino
- Nathan Kline Institute for Psychiatric Research, Orangeburg, NY, United States
| | - Prerana Vaddi
- Nathan Kline Institute for Psychiatric Research, Orangeburg, NY, United States
| | | | - Donald A. Wilson
- Nathan Kline Institute for Psychiatric Research, Orangeburg, NY, United States
- Department of Child and Adolescent Psychiatry, School of Medicine, New York University, New York, NY, United States
- Department of Neuroscience and Physiology, School of Medicine, New York University, New York, NY, United States
| | - Mariko Saito
- Nathan Kline Institute for Psychiatric Research, Orangeburg, NY, United States
- Department of Psychiatry, School of Medicine, New York University, New York, NY, United States
| |
Collapse
|
13
|
Castillo-Ruiz A, Gars A, Sturgeon H, Ronczkowski NM, Pyaram DN, Dauriat CJG, Chassaing B, Forger NG. Brain effects of gestating germ-free persist in mouse neonates despite acquisition of a microbiota at birth. Front Neurosci 2023; 17:1130347. [PMID: 37207179 PMCID: PMC10188942 DOI: 10.3389/fnins.2023.1130347] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Accepted: 04/10/2023] [Indexed: 05/21/2023] Open
Abstract
At birth, mammals experience a massive colonization by microorganisms. We previously reported that newborn mice gestated and born germ-free (GF) have increased microglial labeling and alterations in developmental neuronal cell death in the hippocampus and hypothalamus, as well as greater forebrain volume and body weight when compared to conventionally colonized (CC) mice. To test whether these effects are solely due to differences in postnatal microbial exposure, or instead may be programmed in utero, we cross-fostered GF newborns immediately after birth to CC dams (GF→CC) and compared them to offspring fostered within the same microbiota status (CC→CC, GF→GF). Because key developmental events (including microglial colonization and neuronal cell death) shape the brain during the first postnatal week, we collected brains on postnatal day (P) 7. To track gut bacterial colonization, colonic content was also collected and subjected to 16S rRNA qPCR and Illumina sequencing. In the brains of GF→GF mice, we replicated most of the effects seen previously in GF mice. Interestingly, the GF brain phenotype persisted in GF→CC offspring for almost all measures. In contrast, total bacterial load did not differ between the CC→CC and GF→CC groups on P7, and bacterial community composition was also very similar, with a few exceptions. Thus, GF→CC offspring had altered brain development during at least the first 7 days after birth despite a largely normal microbiota. This suggests that prenatal influences of gestating in an altered microbial environment programs neonatal brain development.
Collapse
Affiliation(s)
- Alexandra Castillo-Ruiz
- Neuroscience Institute, Georgia State University, Atlanta, GA, United States
- *Correspondence: Alexandra Castillo-Ruiz,
| | - Aviva Gars
- Neuroscience Institute, Georgia State University, Atlanta, GA, United States
| | - Hannah Sturgeon
- Neuroscience Institute, Georgia State University, Atlanta, GA, United States
| | | | - Dhanya N. Pyaram
- Neuroscience Institute, Georgia State University, Atlanta, GA, United States
| | - Charlène J. G. Dauriat
- INSERM U1016, Team “Mucosal Microbiota in Chronic Inflammatory Diseases,” Université Paris Cité, Paris, France
| | - Benoit Chassaing
- INSERM U1016, Team “Mucosal Microbiota in Chronic Inflammatory Diseases,” Université Paris Cité, Paris, France
| | - Nancy G. Forger
- Neuroscience Institute, Georgia State University, Atlanta, GA, United States
| |
Collapse
|
14
|
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: 0] [Impact Index Per Article: 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
|
15
|
Shiu FH, Wong JC, Yamamoto T, Lala T, Purcell RH, Owino S, Zhu D, Van Meir EG, Hall RA, Escayg A. Mice lacking full length Adgrb1 (Bai1) exhibit social deficits, increased seizure susceptibility, and altered brain development. Exp Neurol 2022; 351:113994. [PMID: 35114205 PMCID: PMC9817291 DOI: 10.1016/j.expneurol.2022.113994] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 12/20/2021] [Accepted: 01/24/2022] [Indexed: 01/11/2023]
Abstract
The adhesion G protein-coupled receptor BAI1/ADGRB1 plays an important role in suppressing angiogenesis, mediating phagocytosis, and acting as a brain tumor suppressor. BAI1 is also a critical regulator of dendritic spine and excitatory synapse development and interacts with several autism-relevant proteins. However, little is known about the relationship between altered BAI1 function and clinically relevant phenotypes. Therefore, we studied the effect of reduced expression of full length Bai1 on behavior, seizure susceptibility, and brain morphology in Adgrb1 mutant mice. We compared homozygous (Adgrb1-/-), heterozygous (Adgrb1+/-), and wild-type (WT) littermates using a battery of tests to assess social behavior, anxiety, repetitive behavior, locomotor function, and seizure susceptibility. We found that Adgrb1-/- mice showed significant social behavior deficits and increased vulnerability to seizures. Adgrb1-/- mice also showed delayed growth and reduced brain weight. Furthermore, reduced neuron density and increased apoptosis during brain development were observed in the hippocampus of Adgrb1-/- mice, while levels of astrogliosis and microgliosis were comparable to WT littermates. These results show that reduced levels of full length Bai1 is associated with a broader range of clinically relevant phenotypes than previously reported.
Collapse
Affiliation(s)
- Fu Hung Shiu
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, USA; Neuroscience Graduate Program, Graduate Division of Biological and Biomedical Sciences, Laney Graduate School, Emory University, Atlanta, GA, USA
| | - Jennifer C Wong
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, USA
| | - Takahiro Yamamoto
- Department of Neurosurgery, School of Medicine, University of Alabama at Birmingham (UAB), Birmingham, AL, USA
| | - Trisha Lala
- Neuroscience Graduate Program, Graduate Division of Biological and Biomedical Sciences, Laney Graduate School, Emory University, Atlanta, GA, USA; Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA, USA
| | - Ryan H Purcell
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA, USA
| | - Sharon Owino
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA, USA
| | - Dan Zhu
- Department of Neurosurgery, Emory University School of Medicine, Atlanta, GA, USA
| | - Erwin G Van Meir
- Department of Neurosurgery, School of Medicine, University of Alabama at Birmingham (UAB), Birmingham, AL, USA; O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham (UAB), Birmingham, AL, USA
| | - Randy A Hall
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA, USA
| | - Andrew Escayg
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, USA.
| |
Collapse
|
16
|
Markert F, Storch A. Hyperoxygenation During Mid-Neurogenesis Accelerates Cortical Development in the Fetal Mouse Brain. Front Cell Dev Biol 2022; 10:732682. [PMID: 35372333 PMCID: PMC8969024 DOI: 10.3389/fcell.2022.732682] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Accepted: 01/20/2022] [Indexed: 11/13/2022] Open
Abstract
Oxygen tension is well-known to affect cortical development. Fetal brain hyperoxygenation during mid-neurogenesis in mice (embryonic stage E14.5. to E16.5) increases brain size evoked through an increase of neuroprecursor cells. Nevertheless, it is unknown whether these effects can lead to persistent morphological changes within the highly orchestrated brain development. To shed light on this, we used our model of controlled fetal brain hyperoxygenation in time-pregnant C57BL/6J mice housed in a chamber with 75% atmospheric oxygen from E14.5 to E16.5 and analyzed the brains from E14.5, E16.5, P0.5, and P3.5 mouse embryos and pups via immunofluorescence staining. Mid-neurogenesis hyperoxygenation led to an acceleration of cortical development by temporal expansion of the cortical plate with increased NeuN+ neuron counts in hyperoxic brains only until birth. More specifically, the number of Ctip2+ cortical layer 5 (L5) neurons was increased at E16.5 and at birth in hyperoxic brains but normalized in the early postnatal stage (P3.5). The absence of cleaved caspase 3 within the extended Ctip2+ L5 cell population largely excluded apoptosis as a major compensatory mechanism. Timed BrdU/EdU analyses likewise rule out a feedback mechanism. The normalization was, on the contrary, accompanied by an increase of active microglia within L5 targeting Ctip2+ neurons without any signs of apoptosis. Together, hyperoxygenation during mid-neurogenesis phase of fetal brain development provoked a specific transient overshoot of cortical L5 neurons leading to an accelerated cortical development without detectable persistent changes. These observations provide insight into cortical and L5 brain development.
Collapse
Affiliation(s)
- Franz Markert
- Department of Neurology, University of Rostock, Rostock, Germany
| | - Alexander Storch
- Department of Neurology, University of Rostock, Rostock, Germany
- German Center for Neurodegenerative Diseases (DZNE) Rostock/Greifswald, Rostock, Germany
- *Correspondence: Alexander Storch,
| |
Collapse
|
17
|
Cheng AH, Fung SW, Hegazi S, Abdalla OHMH, Cheng HYM. SOX2 Regulates Neuronal Differentiation of the Suprachiasmatic Nucleus. Int J Mol Sci 2021; 23:ijms23010229. [PMID: 35008655 PMCID: PMC8745319 DOI: 10.3390/ijms23010229] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 12/22/2021] [Accepted: 12/23/2021] [Indexed: 12/24/2022] Open
Abstract
In mammals, the hypothalamic suprachiasmatic nucleus (SCN) functions as the central circadian pacemaker, orchestrating behavioral and physiological rhythms in alignment to the environmental light/dark cycle. The neurons that comprise the SCN are anatomically and functionally heterogeneous, but despite their physiological importance, little is known about the pathways that guide their specification and differentiation. Here, we report that the stem/progenitor cell transcription factor, Sex determining region Y-box 2 (Sox2), is required in the embryonic SCN to control the expression of SCN-enriched neuropeptides and transcription factors. Ablation of Sox2 in the developing SCN leads to downregulation of circadian neuropeptides as early as embryonic day (E) 15.5, followed by a decrease in the expression of two transcription factors involved in SCN development, Lhx1 and Six6, in neonates. Thymidine analog-retention assays revealed that Sox2 deficiency contributed to reduced survival of SCN neurons during the postnatal period of cell clearance, but did not affect progenitor cell proliferation or SCN specification. Our results identify SOX2 as an essential transcription factor for the proper differentiation and survival of neurons within the developing SCN.
Collapse
Affiliation(s)
- Arthur H. Cheng
- Department of Biology, University of Toronto Mississauga, Mississauga, ON L5L 1C6, Canada; (A.H.C.); (S.W.F.); (S.H.); (O.H.M.H.A.)
- Department of Cell & Systems Biology, University of Toronto, Toronto, ON M5S 3G5, Canada
| | - Samuel W. Fung
- Department of Biology, University of Toronto Mississauga, Mississauga, ON L5L 1C6, Canada; (A.H.C.); (S.W.F.); (S.H.); (O.H.M.H.A.)
| | - Sara Hegazi
- Department of Biology, University of Toronto Mississauga, Mississauga, ON L5L 1C6, Canada; (A.H.C.); (S.W.F.); (S.H.); (O.H.M.H.A.)
- Department of Cell & Systems Biology, University of Toronto, Toronto, ON M5S 3G5, Canada
| | - Osama Hasan Mustafa Hasan Abdalla
- Department of Biology, University of Toronto Mississauga, Mississauga, ON L5L 1C6, Canada; (A.H.C.); (S.W.F.); (S.H.); (O.H.M.H.A.)
- Department of Cell & Systems Biology, University of Toronto, Toronto, ON M5S 3G5, Canada
| | - Hai-Ying Mary Cheng
- Department of Biology, University of Toronto Mississauga, Mississauga, ON L5L 1C6, Canada; (A.H.C.); (S.W.F.); (S.H.); (O.H.M.H.A.)
- Department of Cell & Systems Biology, University of Toronto, Toronto, ON M5S 3G5, Canada
- Correspondence:
| |
Collapse
|
18
|
Smiley JF, Bleiwas C, Canals-Baker S, Williams SZ, Sears R, Teixeira CM, Wilson DA, Saito M. Neonatal ethanol causes profound reduction of cholinergic cell number in the basal forebrain of adult animals. Alcohol 2021; 97:1-11. [PMID: 34464696 DOI: 10.1016/j.alcohol.2021.08.005] [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: 11/10/2020] [Revised: 05/24/2021] [Accepted: 08/12/2021] [Indexed: 11/26/2022]
Abstract
In animal models that mimic human third-trimester fetal development, ethanol causes substantial cellular apoptosis in the brain, but for most brain structures, the extent of permanent neuron loss that persists into adulthood is unknown. We injected ethanol into C57BL/6J mouse pups at postnatal day 7 (P7) to model human late-gestation ethanol toxicity, and then used stereological methods to investigate adult cell numbers in several subcortical neurotransmitter systems that project extensively in the forebrain to regulate arousal states. Ethanol treatment caused especially large reductions (34-42%) in the cholinergic cells of the basal forebrain, including cholinergic cells in the medial septal/vertical diagonal band nuclei (Ch1/Ch2) and in the horizontal diagonal band/substantia innominata/nucleus basalis nuclei (Ch3/Ch4). Cell loss was also present in non-cholinergic basal forebrain cells, as demonstrated by 34% reduction of parvalbumin-immunolabeled GABA cells and 25% reduction of total Nissl-stained neurons in the Ch1/Ch2 region. In contrast, cholinergic cells in the striatum were reduced only 12% by ethanol, and those of the brainstem pedunculopontine/lateral dorsal tegmental nuclei (Ch5/Ch6) were not significantly reduced. Similarly, ethanol did not significantly reduce dopamine cells of the ventral tegmental area/substantia nigra or serotonin cells in the dorsal raphe nucleus. Orexin (hypocretin) cells in the hypothalamus showed a modest reduction (14%). Our findings indicate that the basal forebrain is especially vulnerable to alcohol exposure in the late gestational period. Reduction of cholinergic and GABAergic projection neurons from the basal forebrain that regulate forebrain arousal may contribute to the behavioral and cognitive deficits associated with neonatal ethanol exposure.
Collapse
|
19
|
Cortes LR, Cisternas CD, Cabahug INKV, Mason D, Ramlall EK, Castillo-Ruiz A, Forger NG. DNA Methylation and Demethylation Underlie the Sex Difference in Estrogen Receptor Alpha in the Arcuate Nucleus. Neuroendocrinology 2021; 112:636-648. [PMID: 34547753 PMCID: PMC8934748 DOI: 10.1159/000519671] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Accepted: 09/15/2021] [Indexed: 01/03/2023]
Abstract
INTRODUCTION Neurons expressing estrogen receptor (ER) ɑ in the arcuate (ARC) and ventromedial (VMH) nuclei of the hypothalamus sex-specifically control energy homeostasis, sexual behavior, and bone density. Females have more ERɑ neurons in the VMH and ARC than males, and the sex difference in the VMH is eliminated by neonatal treatment with testosterone or a DNA methylation inhibitor. OBJECTIVE Here, we tested the roles of testosterone and DNA methylation/demethylation in development of ERɑ in the ARC. METHODS ERɑ was examined at birth and weaning in mice that received vehicle or testosterone subcutaneously, and vehicle or DNA methyltransferase inhibitor intracerebroventricularly, as neonates. To examine effects of DNA demethylation on the ERɑ cell number in the ARC, mice were treated neonatally with small interfering RNAs against ten-eleven translocase enzymes. The methylation status of the ERɑ gene (Esr1) was determined in the ARC and VMH using pyrosequencing of bisulfite-converted DNA. RESULTS A sex difference in ERɑ in the ARC, favoring females, developed between birth and weaning and was due to programming effects of testosterone. Neonatal inhibition of DNA methylation decreased ERɑ in the ARC of females, and an inhibition of demethylation increased ERɑ in the ARC of males. The promoter region of Esr1 exhibited a small sex difference in percent of total methylation in the ARC (females > males) that was opposite to that in the VMH (males > females). CONCLUSION DNA methylation and demethylation regulate ERɑ cell number in the ARC, and methylation correlates with activation of Esr1 in this region.
Collapse
Affiliation(s)
- Laura R Cortes
- Neuroscience Institute, Georgia State University, Atlanta, Georgia, USA
| | - Carla D Cisternas
- Instituto de Investigación Médica Mercedes y Martín Ferrreyra INIMEC-CONICET-UNC, Córdoba, Argentina
| | | | - Damian Mason
- Neuroscience Institute, Georgia State University, Atlanta, Georgia, USA
| | - Emma K Ramlall
- Neuroscience Institute, Georgia State University, Atlanta, Georgia, USA
| | | | - Nancy G Forger
- Neuroscience Institute, Georgia State University, Atlanta, Georgia, USA
| |
Collapse
|
20
|
Chiesa M, Rabiei H, Riffault B, Ferrari DC, Ben-Ari Y. Brain Volumes in Mice are Smaller at Birth After Term or Preterm Cesarean Section Delivery. Cereb Cortex 2021; 31:3579-3591. [PMID: 33754629 DOI: 10.1093/cercor/bhab033] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 01/26/2021] [Accepted: 01/29/2021] [Indexed: 12/31/2022] Open
Abstract
The rate of cesarean section (CS) delivery has steadily increased over the past decades despite epidemiological studies reporting higher risks of neonatal morbidity and neurodevelopmental disorders. Yet, little is known about the immediate impact of CS birth on the brain, hence the need of experimental studies to evaluate brain parameters following this mode of delivery. Using the solvent clearing method iDISCO and 3D imaging technique, we report that on the day of birth, whole-brain, hippocampus, and striatum volumes are reduced in CS-delivered as compared to vaginally-born mice, with a stronger effect observed in preterm CS pups. These results stress the impact of CS delivery, at term or preterm, during parturition and at birth. In contrast, cellular activity and apoptosis are reduced in mice born by CS preterm but not term, suggesting that these early-life processes are only impacted by the combination of preterm birth and CS delivery.
Collapse
Affiliation(s)
- Morgane Chiesa
- Fundamental Research Department, Neurochlore, Ben-Ari Institute of Neuroarcheology (IBEN), Marseille cedex 09, 13288, France
| | - Hamed Rabiei
- Fundamental Research Department, Neurochlore, Ben-Ari Institute of Neuroarcheology (IBEN), Marseille cedex 09, 13288, France
| | - Baptiste Riffault
- Fundamental Research Department, Neurochlore, Ben-Ari Institute of Neuroarcheology (IBEN), Marseille cedex 09, 13288, France
| | - Diana Carolina Ferrari
- Fundamental Research Department, Neurochlore, Ben-Ari Institute of Neuroarcheology (IBEN), Marseille cedex 09, 13288, France
| | - Yehezkel Ben-Ari
- Fundamental Research Department, Neurochlore, Ben-Ari Institute of Neuroarcheology (IBEN), Marseille cedex 09, 13288, France
| |
Collapse
|
21
|
Gars A, Ronczkowski NM, Chassaing B, Castillo-Ruiz A, Forger NG. First Encounters: Effects of the Microbiota on Neonatal Brain Development. Front Cell Neurosci 2021; 15:682505. [PMID: 34168540 PMCID: PMC8217657 DOI: 10.3389/fncel.2021.682505] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Accepted: 05/11/2021] [Indexed: 12/20/2022] Open
Abstract
The microbiota plays important roles in host metabolism and immunity, and its disruption affects adult brain physiology and behavior. Although such findings have been attributed to altered neurodevelopment, few studies have actually examined microbiota effects on the developing brain. This review focuses on developmental effects of the earliest exposure to microbes. At birth, the mammalian fetus enters a world teeming with microbes which colonize all body sites in contact with the environment. Bacteria reach the gut within a few hours of birth and cause a measurable response in the intestinal epithelium. In adults, the gut microbiota signals to the brain via the vagus nerve, bacterial metabolites, hormones, and immune signaling, and work in perinatal rodents is beginning to elucidate which of these signaling pathways herald the very first encounter with gut microbes in the neonate. Neural effects of the microbiota during the first few days of life include changes in neuronal cell death, microglia, and brain cytokine levels. In addition to these effects of direct exposure of the newborn to microbes, accumulating evidence points to a role for the maternal microbiota in affecting brain development via bacterial molecules and metabolites while the offspring is still in utero. Hence, perturbations to microbial exposure perinatally, such as through C-section delivery or antibiotic treatment, alter microbiota colonization and may have long-term neural consequences. The perinatal period is critical for brain development and a close look at microbiota effects during this time promises to reveal the earliest, most primary effects of the microbiota on neurodevelopment.
Collapse
Affiliation(s)
- Aviva Gars
- Neuroscience Institute, Georgia State University, Atlanta, GA, United States
| | | | - Benoit Chassaing
- INSERM U1016, Team "Mucosal Microbiota in Chronic Inflammatory Diseases", CNRS UMR 8104, Université de Paris, Paris, France
| | | | - Nancy G Forger
- Neuroscience Institute, Georgia State University, Atlanta, GA, United States
| |
Collapse
|
22
|
Nemes-Baran AD, White DR, DeSilva TM. Fractalkine-Dependent Microglial Pruning of Viable Oligodendrocyte Progenitor Cells Regulates Myelination. Cell Rep 2021; 32:108047. [PMID: 32814050 PMCID: PMC7478853 DOI: 10.1016/j.celrep.2020.108047] [Citation(s) in RCA: 105] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Revised: 05/22/2020] [Accepted: 07/28/2020] [Indexed: 12/14/2022] Open
Abstract
Oligodendrogenesis occurs during early postnatal development, coincident with neurogenesis and synaptogenesis, raising the possibility that microglia-dependent pruning mechanisms that modulate neurons regulate myelin sheath formation. Here we show a population of ameboid microglia migrating from the ventricular zone into the corpus callosum during early postnatal development, termed “the fountain of microglia,” phagocytosing viable oligodendrocyte progenitor cells (OPCs) before onset of myelination. Fractalkine receptor-deficient mice exhibit a reduction in microglial engulfment of viable OPCs, increased numbers of oligodendrocytes, and reduced myelin thickness but no change in axon number. These data provide evidence that microglia phagocytose OPCs as a homeostatic mechanism for proper myelination. A hallmark of hypomyelinating developmental disorders such as periventricular leukomalacia and of adult demyelinating diseases such as multiple sclerosis is increased numbers of oligodendrocytes but failure to myelinate, suggesting that microglial pruning of OPCs may be impaired in pathological states and hinder myelination. Nemes-Baran et al. show that ameboid microglia engulf living oligodendrocyte progenitor cells (OPCs) during brain development. Fractalkine receptor-deficient microglia exhibit a reduction in engulfment of OPCs, resulting in a surplus of oligodendrocytes and impaired myelination. These data provide evidence that microglia phagocytose OPCs as a homeostatic mechanism required for normal myelination.
Collapse
Affiliation(s)
- Ashley D Nemes-Baran
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Donovan R White
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Tara M DeSilva
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH, USA.
| |
Collapse
|
23
|
Cheng AH, Cheng HYM. Genesis of the Master Circadian Pacemaker in Mice. Front Neurosci 2021; 15:659974. [PMID: 33833665 PMCID: PMC8021851 DOI: 10.3389/fnins.2021.659974] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Accepted: 03/04/2021] [Indexed: 12/13/2022] Open
Abstract
The suprachiasmatic nucleus (SCN) of the hypothalamus is the central circadian clock of mammals. It is responsible for communicating temporal information to peripheral oscillators via humoral and endocrine signaling, ultimately controlling overt rhythms such as sleep-wake cycles, body temperature, and locomotor activity. Given the heterogeneity and complexity of the SCN, its genesis is tightly regulated by countless intrinsic and extrinsic factors. Here, we provide a brief overview of the development of the SCN, with special emphasis on the murine system.
Collapse
Affiliation(s)
- Arthur H. Cheng
- Department of Biology, University of Toronto Mississauga, Mississauga, ON, Canada
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON, Canada
| | - Hai-Ying Mary Cheng
- Department of Biology, University of Toronto Mississauga, Mississauga, ON, Canada
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
24
|
Drastichova Z, Rudajev V, Pallag G, Novotny J. Proteome profiling of different rat brain regions reveals the modulatory effect of prolonged maternal separation on proteins involved in cell death-related processes. Biol Res 2021; 54:4. [PMID: 33557947 PMCID: PMC7871601 DOI: 10.1186/s40659-021-00327-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Accepted: 01/25/2021] [Indexed: 01/08/2023] Open
Abstract
Background Early-life stress in the form of maternal separation can be associated with alterations in offspring neurodevelopment and brain functioning. Here, we aimed to investigate the potential impact of prolonged maternal separation on proteomic profiling of prefrontal cortex, hippocampus and cerebellum of juvenile and young adult rats. A special attention was devoted to proteins involved in the process of cell death and redox state maintenance. Methods Long-Evans pups were separated from their mothers for 3 h daily over the first 3 weeks of life (during days 2–21 of age). Brain tissue samples collected from juvenile (22-day-old) and young adult (90-day-old) rats were used for label-free quantitative (LFQ) proteomic analysis. In parallel, selected oxidative stress markers and apoptosis-related proteins were assessed biochemically and by Western blot, respectively. Results In total, 5526 proteins were detected in our proteomic analysis of rat brain tissue. Approximately one tenth of them (586 proteins) represented those involved in cell death processes or regulation of oxidative stress balance. Prolonged maternal separation caused changes in less than half of these proteins (271). The observed alterations in protein expression levels were age-, sex- and brain region-dependent. Interestingly, the proteins detected by mass spectrometry that are known to be involved in the maintenance of redox state were not markedly altered. Accordingly, we did not observe any significant differences between selected oxidative stress markers, such as the levels of hydrogen peroxide, reduced glutathione, protein carbonylation and lipid peroxidation in brain samples from rats that underwent maternal separation and from the corresponding controls. On the other hand, a number of changes were found in cell death-associated proteins, mainly in those involved in the apoptotic and autophagic pathways. However, there were no detectable alterations in the levels of cleaved products of caspases or Bcl-2 family members. Taken together, these data indicate that the apoptotic and autophagic cell death pathways were not activated by maternal separation either in adolescent or young adult rats. Conclusion Prolonged maternal separation can distinctly modulate expression profiles of proteins associated with cell death pathways in prefrontal cortex, hippocampus and cerebellum of juvenile rats and the consequences of early-life stress may last into adulthood and likely participate in variations in stress reactivity. Supplementary Information The online version contains supplementary material available at 10.1186/s40659-021-00327-5.
Collapse
Affiliation(s)
- Zdenka Drastichova
- Department of Physiology, Faculty of Science, Charles University, Prague, Czech Republic
| | - Vladimir Rudajev
- Department of Physiology, Faculty of Science, Charles University, Prague, Czech Republic
| | - Gergely Pallag
- Department of Physiology, Faculty of Science, Charles University, Prague, Czech Republic
| | - Jiri Novotny
- Department of Physiology, Faculty of Science, Charles University, Prague, Czech Republic.
| |
Collapse
|
25
|
Hoffiz YC, Castillo-Ruiz A, Hall MAL, Hite TA, Gray JM, Cisternas CD, Cortes LR, Jacobs AJ, Forger NG. Birth elicits a conserved neuroendocrine response with implications for perinatal osmoregulation and neuronal cell death. Sci Rep 2021; 11:2335. [PMID: 33504846 PMCID: PMC7840942 DOI: 10.1038/s41598-021-81511-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Accepted: 01/05/2021] [Indexed: 12/17/2022] Open
Abstract
Long-standing clinical findings report a dramatic surge of vasopressin in umbilical cord blood of the human neonate, but the neural underpinnings and function(s) of this phenomenon remain obscure. We studied neural activation in perinatal mice and rats, and found that birth triggers activation of the suprachiasmatic, supraoptic, and paraventricular nuclei of the hypothalamus. This was seen whether mice were born vaginally or via Cesarean section (C-section), and when birth timing was experimentally manipulated. Neuronal phenotyping showed that the activated neurons were predominantly vasopressinergic, and vasopressin mRNA increased fivefold in the hypothalamus during the 2–3 days before birth. Copeptin, a surrogate marker of vasopressin, was elevated 30-to 50-fold in plasma of perinatal mice, with higher levels after a vaginal than a C-section birth. We also found an acute decrease in plasma osmolality after a vaginal, but not C-section birth, suggesting that the difference in vasopressin release between birth modes is functionally meaningful. When vasopressin was administered centrally to newborns, we found an ~ 50% reduction in neuronal cell death in specific brain areas. Collectively, our results identify a conserved neuroendocrine response to birth that is sensitive to birth mode, and influences peripheral physiology and neurodevelopment.
Collapse
Affiliation(s)
- Yarely C Hoffiz
- Neuroscience Institute, Georgia State University, Atlanta, GA, 30302, USA
| | | | - Megan A L Hall
- Neuroscience Institute, Georgia State University, Atlanta, GA, 30302, USA
| | - Taylor A Hite
- Neuroscience Institute, Georgia State University, Atlanta, GA, 30302, USA
| | - Jennifer M Gray
- Neuroscience Institute, Georgia State University, Atlanta, GA, 30302, USA
| | - Carla D Cisternas
- Neuroscience Institute, Georgia State University, Atlanta, GA, 30302, USA.,Instituto de Investigación Médica M Y M Ferreyra, INIMEC-CONICET-UNC, Córdoba, Argentina
| | - Laura R Cortes
- Neuroscience Institute, Georgia State University, Atlanta, GA, 30302, USA
| | - Andrew J Jacobs
- Neuroscience Institute, Georgia State University, Atlanta, GA, 30302, USA
| | - Nancy G Forger
- Neuroscience Institute, Georgia State University, Atlanta, GA, 30302, USA.
| |
Collapse
|
26
|
Yoshida A, Kawata D, Shinotsuka N, Yoshida M, Yamaguchi Y, Miura M. Evidence for the involvement of caspases in establishing proper cerebrospinal fluid hydrodynamics. Neurosci Res 2021; 170:145-153. [PMID: 33417971 DOI: 10.1016/j.neures.2020.12.006] [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: 10/13/2020] [Revised: 12/26/2020] [Accepted: 12/27/2020] [Indexed: 11/19/2022]
Abstract
A large number of cells undergo apoptosis via caspase activation during and after neural tube closure (NTC) in mammals. Apoptosis is executed by either intrinsic or extrinsic apoptotic pathways, and inhibition of each pathway causes developmental defects around NTC stages, which hampers the physiological roles of apoptosis and caspases after NTC. We generated transgenic mice in which a broad spectrum of caspases could be suppressed in a spatiotemporal manner by pan-caspase inhibitor protein p35 originating from baculovirus. Mice with nervous system-specific expression of p35 (Nestin-Cre (NCre);p35V mice) exhibited postnatal lethality within 1 month after birth. They were born at the expected Mendelian ratio, but demonstrated severe postnatal growth retardation and hydrocephalus. The flow of cerebrospinal fluid (CSF) between the third and fourth ventricles was disturbed, whereas neither stenosis nor abnormality in ciliary morphology was observed in the pathway of CSF flow. Hydrocephalus and growth retardation of NCre;p35V mice were not rescued by the deletion of RIPK3, an essential factor for necroptosis which occurs in the absence of caspase-8 activation during development. The CSF of NCre;p35V mice contained a larger amount of secreted proteins than that of the controls. These findings suggest that the establishment of proper CSF dynamics requires caspase activity during brain development after NTC.
Collapse
Affiliation(s)
- Ayako Yoshida
- Department of Genetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Daisuke Kawata
- Department of Genetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Naomi Shinotsuka
- Department of Genetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Mariko Yoshida
- Department of Genetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Yoshifumi Yamaguchi
- Department of Genetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Japan; Hibernation Metabolism, Physiology, and Development Group, Institute of Low Temperature Science, Hokkaido University, Sapporo, Hokkaido 060-0819, Japan; Global Station for Biosurfaces and Drug Discovery, Hokkaido University, Sapporo, Hokkaido 060-0812, Japan.
| | - Masayuki Miura
- Department of Genetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Japan.
| |
Collapse
|
27
|
Does Birth Trigger Cell Death in the Developing Brain? eNeuro 2020; 7:ENEURO.0517-19.2020. [PMID: 32015098 PMCID: PMC7031855 DOI: 10.1523/eneuro.0517-19.2020] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2019] [Revised: 01/14/2020] [Accepted: 01/15/2020] [Indexed: 12/27/2022] Open
Abstract
Developmental cell death eliminates half of the neurons initially generated in the mammalian brain, and occurs perinatally in many species. It is possible that the timing of neuronal cell death is developmentally programmed, and only coincidentally associated with birth. Alternatively, birth may play a role in shaping cell death. To test these competing hypotheses, we experimentally advanced or delayed birth by 1 d in mice (within the normal range of gestation for the species) and examined effects on the temporal pattern and magnitude (amount) of neuronal cell death, using immunohistochemical detection of activated caspase-3 as a cell death marker. In order to detect effects of subtle changes in birth timing, we focused on brain areas that exhibit sharp postnatal peaks in cell death. We find that advancing birth advances peak cell death, supporting the hypothesis that birth triggers cell death. However, a delay of birth does not delay cell death. Thus, birth can advance cell death, but if postponed, a developmental program governs. Advancing or delaying birth also caused region-specific changes in the overall magnitude of cell death. Our findings shed light on the long-standing question of what controls the timing and magnitude of developmental neuronal cell death, and position birth as an orchestrator of brain development. Because humans across the world now routinely alter birth timing, these findings may have implications for current obstetric practices.
Collapse
|
28
|
Cisternas CD, Cortes LR, Golynker I, Castillo-Ruiz A, Forger NG. Neonatal Inhibition of DNA Methylation Disrupts Testosterone-Dependent Masculinization of Neurochemical Phenotype. Endocrinology 2020; 161:5631853. [PMID: 31742329 DOI: 10.1210/endocr/bqz022] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Accepted: 11/18/2019] [Indexed: 11/19/2022]
Abstract
Many neural sex differences are differences in the number of neurons of a particular phenotype. For example, male rodents have more calbindin-expressing neurons in the medial preoptic area (mPOA) and bed nucleus of the stria terminalis (BNST), and females have more neurons expressing estrogen receptor alpha (ERα) and kisspeptin in the ventromedial nucleus of the hypothalamus (VMH) and the anteroventral periventricular nucleus (AVPV), respectively. These sex differences depend on neonatal exposure to testosterone, but the underlying molecular mechanisms are unknown. DNA methylation is important for cell phenotype differentiation throughout the developing organism. We hypothesized that testosterone causes sex differences in neurochemical phenotype via changes in DNA methylation, and tested this by inhibiting DNA methylation neonatally in male and female mice, and in females given a masculinizing dose of testosterone. Neonatal testosterone treatment masculinized calbindin, ERα and kisspeptin cell number of females at weaning. Inhibiting DNA methylation with zebularine increased calbindin cell number only in control females, thus eliminating sex differences in calbindin in the mPOA and BNST. Zebularine also reduced the sex difference in ERα cell number in the VMH, in this case by increasing ERα neuron number in males and testosterone-treated females. In contrast, the neonatal inhibition of DNA methylation had no effect on kisspeptin cell number. We conclude that testosterone normally increases the number of calbindin cells and reduces ERα cells in males through orchestrated changes in DNA methylation, contributing to, or causing, the sex differences in both cell types.
Collapse
Affiliation(s)
| | - Laura R Cortes
- Neuroscience Institute, Georgia State University, Atlanta, GA
| | - Ilona Golynker
- Neuroscience Institute, Georgia State University, Atlanta, GA
| | | | - Nancy G Forger
- Neuroscience Institute, Georgia State University, Atlanta, GA
| |
Collapse
|
29
|
Hassani OK, Rymar VV, Nguyen KQ, Huo L, Cloutier JF, Miller FD, Sadikot AF. The noradrenergic system is necessary for survival of vulnerable midbrain dopaminergic neurons: implications for development and Parkinson's disease. Neurobiol Aging 2019; 85:22-37. [PMID: 31734438 DOI: 10.1016/j.neurobiolaging.2019.09.014] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2013] [Revised: 09/17/2019] [Accepted: 09/19/2019] [Indexed: 12/22/2022]
Abstract
The cause of midbrain dopaminergic (mDA) neuron loss in sporadic Parkinson's disease (PD) is multifactorial, involving cell autonomous factors, cell-cell interactions, and the effects of environmental toxins. Early loss of neurons in the locus coeruleus (LC), the main source of ascending noradrenergic (NA) projections, is an important feature of PD and other neurodegenerative disorders. We hypothesized that NA afferents provide trophic support for vulnerable mDA neurons. We demonstrate that depriving mDA neurons of NA input increases postnatal apoptosis and decreases cell survival in young adult rodents, with relative sparing of calbindin-positive subpopulations known to be resistant to degeneration in PD. As a mechanism, we propose that the neurotrophin brain-derived neurotrophic factor (BDNF) modulates anterograde survival effects of LC inputs to mDA neurons. We demonstrate that the LC is rich in BDNF mRNA in postnatal and young adult brains. Early postnatal NA denervation reduces both BDNF protein and activation of TrkB receptors in the ventral midbrain. Furthermore, overexpression of BDNF in NA afferents in transgenic mice increases mDA neuronal survival. Finally, increasing NA activity in primary cultures of mDA neurons improves survival, an effect that is additive or synergistic in the presence of different concentrations of BDNF. Taken together, our results point to a novel mechanism whereby LC afferents couple BDNF effects and NA activity to provide anterograde trophic support for vulnerable mDA neurons. Early loss of NA activity and anterograde neurotrophin support may contribute to degeneration of vulnerable neurons in PD and other neurodegenerative disorders.
Collapse
Affiliation(s)
- Oum Kaltoum Hassani
- Department of Neurology & Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
| | - Vladimir V Rymar
- Department of Neurology & Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
| | - Khanh Q Nguyen
- Department of Neurology & Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
| | - Lia Huo
- Department of Neurology & Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
| | - Jean-François Cloutier
- Department of Neurology & Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
| | - Freda D Miller
- Departments of Medical Genetics, Microbiology & Physiology, The Hospital for Sick Children Research Institute, University of Toronto, Toronto, Ontario, Canada
| | - Abbas F Sadikot
- Department of Neurology & Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada.
| |
Collapse
|
30
|
Jacobs AJ, Castillo‐Ruiz A, Cisternas CD, Forger NG. Microglial Depletion Causes Region‐Specific Changes to Developmental Neuronal Cell Death in the Mouse Brain. Dev Neurobiol 2019; 79:769-779. [DOI: 10.1002/dneu.22706] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Revised: 05/17/2019] [Accepted: 06/12/2019] [Indexed: 12/21/2022]
Affiliation(s)
- Andrew J. Jacobs
- Neuroscience Institute Georgia State University P.O. Box 5030 Atlanta Georgia30302‐5030
| | | | - Carla D. Cisternas
- Neuroscience Institute Georgia State University P.O. Box 5030 Atlanta Georgia30302‐5030
| | - Nancy G. Forger
- Neuroscience Institute Georgia State University P.O. Box 5030 Atlanta Georgia30302‐5030
| |
Collapse
|
31
|
Ono D, Honma KI, Yanagawa Y, Yamanaka A, Honma S. GABA in the suprachiasmatic nucleus refines circadian output rhythms in mice. Commun Biol 2019; 2:232. [PMID: 31263776 PMCID: PMC6588595 DOI: 10.1038/s42003-019-0483-6] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Accepted: 05/29/2019] [Indexed: 01/10/2023] Open
Abstract
In mammals, the circadian rhythms are regulated by the central clock located in the hypothalamic suprachiasmatic nucleus (SCN), which is composed of heterogeneous neurons with various neurotransmitters. Among them an inhibitory neurotransmitter, γ-Amino-Butyric-Acid (GABA), is expressed in almost all SCN neurons, however, its role in the circadian physiology is still unclear. Here, we show that the SCN of fetal mice lacking vesicular GABA transporter (VGAT-/-) or GABA synthesizing enzyme, glutamate decarboxylase (GAD65-/-/67-/-), shows burst firings associated with large Ca2+ spikes throughout 24 hours, which spread over the entire SCN slice in synchrony. By contrast, circadian PER2 rhythms in VGAT-/- and GAD65-/-/67-/- SCN remain intact. SCN-specific VGAT deletion in adult mice dampens circadian behavior rhythm. These findings indicate that GABA in the fetal SCN is necessary for refinement of the circadian firing rhythm and, possibly, for stabilizing the output signals, but not for circadian integration of multiple cellular oscillations.
Collapse
Affiliation(s)
- Daisuke Ono
- Department of Neuroscience II, Research Institute of Environmental Medicine, Nagoya University, Furo-cho, Chikusa-ku, Nagoya, 464-8601 Japan
- Department of Neural Regulation, Nagoya University Graduate School of Medicine, Nagoya, 466-8550 Japan
| | - Ken-ichi Honma
- Research and Education Center for Brain Science, Hokkaido University Graduate School of Medicine, Sapporo, 060-8638 Japan
| | - Yuchio Yanagawa
- Department of Genetic and Behavioral Neuroscience, Gunma University Graduate School of Medicine, Maebashi, Gunma 371-8511 Japan
| | - Akihiro Yamanaka
- Department of Neuroscience II, Research Institute of Environmental Medicine, Nagoya University, Furo-cho, Chikusa-ku, Nagoya, 464-8601 Japan
- Department of Neural Regulation, Nagoya University Graduate School of Medicine, Nagoya, 466-8550 Japan
| | - Sato Honma
- Research and Education Center for Brain Science, Hokkaido University Graduate School of Medicine, Sapporo, 060-8638 Japan
| |
Collapse
|
32
|
Carmona-Alcocer V, Rohr KE, Joye DAM, Evans JA. Circuit development in the master clock network of mammals. Eur J Neurosci 2018; 51:82-108. [PMID: 30402923 DOI: 10.1111/ejn.14259] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Revised: 10/08/2018] [Accepted: 10/31/2018] [Indexed: 12/24/2022]
Abstract
Daily rhythms are generated by the circadian timekeeping system, which is orchestrated by the master circadian clock in the suprachiasmatic nucleus (SCN) of mammals. Circadian timekeeping is endogenous and does not require exposure to external cues during development. Nevertheless, the circadian system is not fully formed at birth in many mammalian species and it is important to understand how SCN development can affect the function of the circadian system in adulthood. The purpose of the current review is to discuss the ontogeny of cellular and circuit function in the SCN, with a focus on work performed in model rodent species (i.e., mouse, rat, and hamster). Particular emphasis is placed on the spatial and temporal patterns of SCN development that may contribute to the function of the master clock during adulthood. Additional work aimed at decoding the mechanisms that guide circadian development is expected to provide a solid foundation upon which to better understand the sources and factors contributing to aberrant maturation of clock function.
Collapse
Affiliation(s)
| | - Kayla E Rohr
- Department of Biomedical Sciences, Marquette University, Milwaukee, Wisconsin
| | - Deborah A M Joye
- Department of Biomedical Sciences, Marquette University, Milwaukee, Wisconsin
| | - Jennifer A Evans
- Department of Biomedical Sciences, Marquette University, Milwaukee, Wisconsin
| |
Collapse
|
33
|
Birth delivery mode alters perinatal cell death in the mouse brain. Proc Natl Acad Sci U S A 2018; 115:11826-11831. [PMID: 30322936 DOI: 10.1073/pnas.1811962115] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Labor and a vaginal delivery trigger changes in peripheral organs that prepare the mammalian fetus to survive ex utero. Surprisingly little attention has been given to whether birth also influences the brain, and to how alterations in birth mode affect neonatal brain development. These are important questions, given the high rates of cesarean section (C-section) delivery worldwide, many of which are elective. We examined the effect of birth mode on neuronal cell death, a widespread developmental process that occurs primarily during the first postnatal week in mice. Timed-pregnant dams were randomly assigned to C-section deliveries that were yoked to vaginal births to carefully match gestation length and circadian time of parturition. Compared with rates of cell death just before birth, vaginally-born offspring had an abrupt, transient decrease in cell death in many brain regions, suggesting that a vaginal delivery is neuroprotective. In contrast, cell death was either unchanged or increased in C-section-born mice. Effects of delivery mode on cell death were greatest for the paraventricular nucleus of the hypothalamus (PVN), which is central to the stress response and brain-immune interactions. The greater cell death in the PVN of C-section-delivered newborns was associated with a reduction in the number of PVN neurons expressing vasopressin at weaning. C-section-delivered mice also showed altered vocalizations in a maternal separation test and greater body mass at weaning. Our results suggest that vaginal birth acutely impacts brain development, and that alterations in birth mode may have lasting consequences.
Collapse
|
34
|
The Neural Mechanisms of Sexually Dimorphic Aggressive Behaviors. Trends Genet 2018; 34:755-776. [PMID: 30173869 DOI: 10.1016/j.tig.2018.07.001] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Revised: 05/16/2018] [Accepted: 07/05/2018] [Indexed: 10/28/2022]
Abstract
Aggression is a fundamental social behavior that is essential for competing for resources and protecting oneself and families in both males and females. As a result of natural selection, aggression is often displayed differentially between the sexes, typically at a higher level in males than females. Here, we highlight the behavioral differences between male and female aggression in rodents. We further outline the aggression circuits in males and females, and compare their differences at each circuit node. Lastly, we summarize our current understanding regarding the generation of sexually dimorphic aggression circuits during development and their maintenance during adulthood. In both cases, gonadal steroid hormones appear to play crucial roles in differentiating the circuits by impacting on the survival, morphology, and intrinsic properties of relevant cells. Many other factors, such as environment and experience, may also contribute to sex differences in aggression and remain to be investigated in future studies.
Collapse
|
35
|
The microbiota influences cell death and microglial colonization in the perinatal mouse brain. Brain Behav Immun 2018; 67:218-229. [PMID: 28890156 PMCID: PMC5696094 DOI: 10.1016/j.bbi.2017.08.027] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Revised: 08/18/2017] [Accepted: 08/23/2017] [Indexed: 12/21/2022] Open
Abstract
The mammalian fetus develops in a largely sterile environment, and direct exposure to a complex microbiota does not occur until birth. We took advantage of this to examine the effect of the microbiota on brain development during the first few days of life. The expression of anti- and pro-inflammatory cytokines, developmental cell death, and microglial colonization in the brain were compared between newborn conventionally colonized mice and mice born in sterile, germ-free (GF) conditions. Expression of the pro-inflammatory cytokines interleukin 1β and tumor necrosis factor α was markedly suppressed in GF newborns. GF mice also had altered cell death, with some regions exhibiting higher rates (paraventricular nucleus of the hypothalamus and the CA1 oriens layer of the hippocampus) and other regions exhibiting no change or lower rates (arcuate nucleus of the hypothalamus) of cell death. Microglial labeling was elevated in GF mice, due to an increase in both microglial cell size and number. The changes in cytokine expression, cell death and microglial labeling were evident on the day of birth, but were absent on embryonic day 18.5, approximately one-half day prior to expected delivery. Taken together, our results suggest that direct exposure to the microbiota at birth influences key neurodevelopmental events and does so within hours. These findings may help to explain some of the behavioral and neurochemical alterations previously seen in adult GF mice.
Collapse
|
36
|
Morishita M, Maejima S, Tsukahara S. Gonadal Hormone-Dependent Sexual Differentiation of a Female-Biased Sexually Dimorphic Cell Group in the Principal Nucleus of the Bed Nucleus of the Stria Terminalis in Mice. Endocrinology 2017; 158:3512-3525. [PMID: 28977609 DOI: 10.1210/en.2017-00240] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Accepted: 07/27/2017] [Indexed: 11/19/2022]
Abstract
We recently reported a female-biased sexually dimorphic area in the mouse brain in the boundary region between the preoptic area and the bed nucleus of the stria terminalis (BNST). We reexamined this area and found that it is a ventral part of the principal nucleus of the BNST (BNSTp). The BNSTp is a male-biased sexually dimorphic nucleus, but the ventral part of the BNSTp (BNSTpv) exhibits female-biased sex differences in volume and neuron number. The volume and neuron number of the BNSTpv were increased in males by neonatal orchiectomy and decreased in females by treatment with testosterone, dihydrotestosterone, or estradiol within 5 days after birth. Sex differences in the volume and neuron number of the BNSTpv emerged before puberty. These sex differences became prominent in adulthood with increasing volume in females and loss of neurons in males during the pubertal/adolescent period. Prepubertal orchiectomy did not affect the BNSTpv, although prepubertal ovariectomy reduced the volume increase and induced loss of neurons in the female BNSTpv. In contrast, the volume and neuron number of male-biased sexually dimorphic nuclei that are composed of mainly calbindin neurons and are located in the preoptic area and BNST were decreased by prepubertal orchiectomy but not affected by prepubertal ovariectomy. Testicular testosterone during the postnatal period may defeminize the BNSTpv via binding directly to the androgen receptor and indirectly to the estrogen receptor after aromatization, although defeminization may proceed independently of testicular hormones in the pubertal/adolescent period. Ovarian hormones may act to feminize the BNSTpv during the pubertal/adolescent period.
Collapse
Affiliation(s)
- Masahiro Morishita
- Division of Life Science, Graduate School of Science and Engineering, Saitama University, Saitama 338-8570, Japan
| | - Sho Maejima
- Division of Life Science, Graduate School of Science and Engineering, Saitama University, Saitama 338-8570, Japan
| | - Shinji Tsukahara
- Division of Life Science, Graduate School of Science and Engineering, Saitama University, Saitama 338-8570, Japan
| |
Collapse
|
37
|
Blanquie O, Yang JW, Kilb W, Sharopov S, Sinning A, Luhmann HJ. Electrical activity controls area-specific expression of neuronal apoptosis in the mouse developing cerebral cortex. eLife 2017; 6:27696. [PMID: 28826501 PMCID: PMC5582867 DOI: 10.7554/elife.27696] [Citation(s) in RCA: 73] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Accepted: 08/18/2017] [Indexed: 12/22/2022] Open
Abstract
Programmed cell death widely but heterogeneously affects the developing brain, causing the loss of up to 50% of neurons in rodents. However, whether this heterogeneity originates from neuronal identity and/or network-dependent processes is unknown. Here, we report that the primary motor cortex (M1) and primary somatosensory cortex (S1), two adjacent but functionally distinct areas, display striking differences in density of apoptotic neurons during the early postnatal period. These differences in rate of apoptosis negatively correlate with region-dependent levels of activity. Disrupting this activity either pharmacologically or by electrical stimulation alters the spatial pattern of apoptosis and sensory deprivation leads to exacerbated amounts of apoptotic neurons in the corresponding functional area of the neocortex. Thus, our data demonstrate that spontaneous and periphery-driven activity patterns are important for the structural and functional maturation of the neocortex by refining the final number of cortical neurons in a region-dependent manner.
Collapse
Affiliation(s)
- Oriane Blanquie
- Institute of Physiology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Jenq-Wei Yang
- Institute of Physiology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Werner Kilb
- Institute of Physiology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Salim Sharopov
- Institute of Physiology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Anne Sinning
- Institute of Physiology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Heiko J Luhmann
- Institute of Physiology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| |
Collapse
|
38
|
Nelson LH, Warden S, Lenz KM. Sex differences in microglial phagocytosis in the neonatal hippocampus. Brain Behav Immun 2017; 64:11-22. [PMID: 28341582 PMCID: PMC5512447 DOI: 10.1016/j.bbi.2017.03.010] [Citation(s) in RCA: 125] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Revised: 03/07/2017] [Accepted: 03/20/2017] [Indexed: 12/12/2022] Open
Abstract
Microglia regulate brain development through many processes, such as promoting neurogenesis, supporting cell survival, and phagocytizing progenitor, newly-born, and dying cells. Many of these same developmental processes show robust sex differences, yet very few studies have assessed sex differences in microglia function during development. Hormonally-induced sexual differentiation of the brain occurs during the perinatal period, thus we examined sex differences in microglial morphology, phagocytosis, and proliferation in the hippocampus during the early postnatal period. We found that the neonatal female hippocampus had significantly more microglia with phagocytic cups than the male hippocampus. We subsequently found that female microglia phagocytized more neural progenitor cells and healthy cells compared to males, but there were no sex differences in the number of newly-born or dying cells targeted by microglial phagocytosis. We found that the number of phagocytic microglia in females was reduced to male-typical levels by treatment with estradiol, the hormone responsible for masculinizing the rodent brain. Females also had higher expression of several phagocytic pathway genes in the hippocampus compared to males. In contrast to robust sex differences in phagocytic microglia, we found no sex differences in the number of microglia with amoeboid, transitioning, or ramified morphologies or differences in three-dimensional reconstructions of microglial morphology. While we did not find a baseline sex difference in microglial proliferation during or following the prenatal gonadal hormone surge in males, we found that estradiol treatment increased microglia proliferation in females. Overall, these data show that there are important sex differences in microglia function in the hippocampus during the early neonatal period.
Collapse
Affiliation(s)
- Lars H Nelson
- Department of Neuroscience, The Ohio State University, 333 W. 10th Ave., Columbus, OH 43210, USA; Group in Behavioral Neuroendocrinology, The Ohio State University, Columbus OH, USA.
| | - Spencer Warden
- Department of Psychology, The Ohio State University, 1835 Neil Ave, Columbus, OH 43210,Group in Behavioral Neuroendocrinology, The Ohio State University, Columbus OH
| | - Kathryn M Lenz
- Department of Neuroscience, The Ohio State University, 333 W. 10th Ave., Columbus, OH 43210, USA; Department of Psychology, The Ohio State University, 1835 Neil Ave, Columbus, OH 43210, USA; Group in Behavioral Neuroendocrinology, The Ohio State University, Columbus OH, USA.
| |
Collapse
|
39
|
Mosley M, Weathington J, Cortes LR, Bruggeman E, Castillo-Ruiz A, Xue B, Forger NG. Neonatal Inhibition of DNA Methylation Alters Cell Phenotype in Sexually Dimorphic Regions of the Mouse Brain. Endocrinology 2017; 158:1838-1848. [PMID: 28398586 PMCID: PMC5460944 DOI: 10.1210/en.2017-00205] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2017] [Accepted: 03/24/2017] [Indexed: 02/06/2023]
Abstract
Many of the best-studied neural sex differences relate to differences in cell number and are due to the hormonal control of developmental cell death. However, several prominent neural sex differences persist even if cell death is eliminated. We hypothesized that these may reflect cell phenotype "decisions" that depend on epigenetic mechanisms, such as DNA methylation. To test this, we treated newborn mice with the DNA methyltransferase (DNMT) inhibitor zebularine, or vehicle, and examined two sexually dimorphic markers at weaning. As expected, control males had more cells immunoreactive for calbindin-D28k (CALB) in the medial preoptic area (mPOA) and fewer cells immunoreactive for estrogen receptor α (ERα) in the ventrolateral portion of the ventromedial nucleus of the hypothalamus (VMHvl) and the mPOA than did females. Neonatal DNMT inhibition markedly increased CALB cell number in both sexes and ERα cell density in males; as a result, the sex differences in ERα in the VMHvl and mPOA were completely eliminated in zebularine-treated animals. Zebularine treatment did not affect developmental cell death or the total density of Nissl-stained cells at weaning. Thus, a neonatal disruption of DNA methylation apparently has long-term effects on the proportion of cells expressing CALB and ERα, and some of these effects are sex specific. We also found that sex differences in CALB in the mPOA and ERα in the VMHvl persist in mice with a neuron-specific depletion of either Dnmt1 or Dnmt3b, indicating that neither DNMT alone is likely to be required for the sexually dimorphic expression of these markers.
Collapse
Affiliation(s)
- Morgan Mosley
- Neuroscience Institute and Center for Behavioral Neuroscience, Georgia State University, Atlanta, Georgia 30302
| | - Jill Weathington
- Neuroscience Institute and Center for Behavioral Neuroscience, Georgia State University, Atlanta, Georgia 30302
| | - Laura R. Cortes
- Neuroscience Institute and Center for Behavioral Neuroscience, Georgia State University, Atlanta, Georgia 30302
| | - Emily Bruggeman
- Department of Biology, Neuroscience Institute and Center for Obesity Reversal, Georgia State University, Atlanta, Georgia 30302
| | - Alexandra Castillo-Ruiz
- Neuroscience Institute and Center for Behavioral Neuroscience, Georgia State University, Atlanta, Georgia 30302
| | - Bingzhong Xue
- Department of Biology, Neuroscience Institute and Center for Obesity Reversal, Georgia State University, Atlanta, Georgia 30302
| | - Nancy G. Forger
- Neuroscience Institute and Center for Behavioral Neuroscience, Georgia State University, Atlanta, Georgia 30302
| |
Collapse
|
40
|
Huang WC, Ferris E, Cheng T, Hörndli CS, Gleason K, Tamminga C, Wagner JD, Boucher KM, Christian JL, Gregg C. Diverse Non-genetic, Allele-Specific Expression Effects Shape Genetic Architecture at the Cellular Level in the Mammalian Brain. Neuron 2017; 93:1094-1109.e7. [PMID: 28238550 PMCID: PMC5774018 DOI: 10.1016/j.neuron.2017.01.033] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2016] [Revised: 11/27/2016] [Accepted: 01/30/2017] [Indexed: 01/19/2023]
Abstract
Interactions between genetic and epigenetic effects shape brain function, behavior, and the risk for mental illness. Random X inactivation and genomic imprinting are epigenetic allelic effects that are well known to influence genetic architecture and disease risk. Less is known about the nature, prevalence, and conservation of other potential epigenetic allelic effects in vivo in the mouse and primate brain. Here we devise genomics, in situ hybridization, and mouse genetics strategies to uncover diverse allelic effects in the brain that are not caused by imprinting or genetic variation. We found allelic effects that are developmental stage and cell type specific, that are prevalent in the neonatal brain, and that cause mosaics of monoallelic brain cells that differentially express wild-type and mutant alleles for heterozygous mutations. Finally, we show that diverse non-genetic allelic effects that impact mental illness risk genes exist in the macaque and human brain. Our findings have potential implications for mammalian brain genetics. VIDEO ABSTRACT.
Collapse
Affiliation(s)
- Wei-Chao Huang
- Departments of Neurobiology & Anatomy, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
| | - Elliott Ferris
- Departments of Neurobiology & Anatomy, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
| | - Tong Cheng
- Departments of Neurobiology & Anatomy, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
| | - Cornelia Stacher Hörndli
- Departments of Neurobiology & Anatomy, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
| | - Kelly Gleason
- Department of Psychiatry, UT Southwestern, Dallas, TX 75390-9127, USA
| | - Carol Tamminga
- Department of Psychiatry, UT Southwestern, Dallas, TX 75390-9127, USA
| | - Janice D Wagner
- Department of Pathology, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| | - Kenneth M Boucher
- Department of Internal Medicine, University of Utah School of Medicine, Salt Lake City, UT 84112, USA; Cancer Biostatistics Shared Resource, University of Utah School of Medicine, Salt Lake City, UT 84112, USA; Huntsman Cancer Institute, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
| | - Jan L Christian
- Departments of Neurobiology & Anatomy, University of Utah School of Medicine, Salt Lake City, UT 84112, USA; Department of Internal Medicine, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
| | - Christopher Gregg
- Robertson Neuroscience Investigator, New York Stem Cell Foundation, University of Utah School of Medicine, Salt Lake City, UT 84112, USA; Departments of Neurobiology & Anatomy, University of Utah School of Medicine, Salt Lake City, UT 84112, USA; Department of Human Genetics, University of Utah School of Medicine, Salt Lake City, UT 84112, USA.
| |
Collapse
|
41
|
Evonuk KS, Prabhu SD, Young ME, DeSilva TM. Myocardial ischemia/reperfusion impairs neurogenesis and hippocampal-dependent learning and memory. Brain Behav Immun 2017; 61:266-273. [PMID: 27600185 PMCID: PMC5511033 DOI: 10.1016/j.bbi.2016.09.001] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Revised: 08/25/2016] [Accepted: 09/02/2016] [Indexed: 11/16/2022] Open
Abstract
The incidence of cognitive impairment in cardiovascular disease (CVD) patients has increased, adversely impacting quality of life and imposing a significant economic burden. Brain imaging of CVD patients has detected changes in the hippocampus, a brain region critical for normal learning and memory. However, it is not clear whether adverse cardiac events or other associated co-morbidities impair cognition. Here, using a murine model of acute myocardial ischemia/reperfusion (I/R), where the coronary artery was occluded for 30min followed by reperfusion, we tested the hypothesis that acute myocardial infarction triggers impairment in cognitive function. Two months following cardiac I/R, behavioral assessments specific for hippocampal cognitive function were performed. Mice subjected to cardiac I/R performed worse in the fear-conditioning paradigm as well as the object location memory behavioral test compared to sham-operated mice. Reactive gliosis was apparent in the hippocampal subregions CA1, CA3, and dentate gyrus 72h post-cardiac I/R as compared with sham, which was sustained two months post-cardiac I/R. Consistent with the inflammatory response, the abundance of doublecortin positive newborn neurons was decreased in the dentate gyrus 72h and 2months post-cardiac I/R as compared with sham. Therefore, we conclude that following acute myocardial infarction, rapid inflammatory responses negatively affect neurogenesis, which may underlie long-term changes in learning and memory.
Collapse
Affiliation(s)
- Kirsten S Evonuk
- Center for Glial Biology in Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, United States; Department of Physical Medicine Rehabilitation, University of Alabama at Birmingham, Birmingham, AL 35294, United States
| | - Sumanth D Prabhu
- Department of Medicine, Division of Cardiovascular Disease, University of Alabama at Birmingham, Birmingham, AL 35294, United States
| | - Martin E Young
- Department of Medicine, Division of Cardiovascular Disease, University of Alabama at Birmingham, Birmingham, AL 35294, United States
| | - Tara M DeSilva
- Center for Glial Biology in Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, United States; Department of Physical Medicine Rehabilitation, University of Alabama at Birmingham, Birmingham, AL 35294, United States; Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294, United States.
| |
Collapse
|
42
|
Mosley M, Shah C, Morse KA, Miloro SA, Holmes MM, Ahern TH, Forger NG. Patterns of cell death in the perinatal mouse forebrain. J Comp Neurol 2017; 525:47-64. [PMID: 27199256 PMCID: PMC5116296 DOI: 10.1002/cne.24041] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Revised: 05/16/2016] [Accepted: 05/17/2016] [Indexed: 12/15/2022]
Abstract
The importance of cell death in brain development has long been appreciated, but many basic questions remain, such as what initiates or terminates the cell death period. One obstacle has been the lack of quantitative data defining exactly when cell death occurs. We recently created a "cell death atlas," using the detection of activated caspase-3 (AC3) to quantify apoptosis in the postnatal mouse ventral forebrain and hypothalamus, and found that the highest rates of cell death were seen at the earliest postnatal ages in most regions. Here we have extended these analyses to prenatal ages and additional brain regions. We quantified cell death in 16 forebrain regions across nine perinatal ages from embryonic day (E) 17 to postnatal day (P) 11 and found that cell death peaks just after birth in most regions. We found greater cell death in several regions in offspring delivered vaginally on the day of parturition compared with those of the same postconception age but still in utero at the time of collection. We also found massive cell death in the oriens layer of the hippocampus on P1 and in regions surrounding the anterior crossing of the corpus callosum on E18 as well as the persistence of large numbers of cells in those regions in adult mice lacking the pro-death Bax gene. Together these findings suggest that birth may be an important trigger of neuronal cell death and identify transient cell groups that may undergo wholesale elimination perinatally. J. Comp. Neurol. 525:47-64, 2017. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Morgan Mosley
- Neuroscience Institute, Georgia State University, Atlanta, Georgia, 30302
| | - Charisma Shah
- Neuroscience Institute, Georgia State University, Atlanta, Georgia, 30302
| | - Kiriana A Morse
- Department of Psychology, Center for Behavioral Neuroscience, Quinnipiac University, Hamden, Connecticut, 06518
| | - Stephen A Miloro
- Department of Psychology, Center for Behavioral Neuroscience, Quinnipiac University, Hamden, Connecticut, 06518
| | - Melissa M Holmes
- Department of Psychology, University of Toronto Mississauga, Mississauga, Ontario, L5L 1C6, Canada
| | - Todd H Ahern
- Department of Psychology, Center for Behavioral Neuroscience, Quinnipiac University, Hamden, Connecticut, 06518
| | - Nancy G Forger
- Neuroscience Institute, Georgia State University, Atlanta, Georgia, 30302
| |
Collapse
|
43
|
Strahan JA, Walker WH, Montgomery TR, Forger NG. Minocycline causes widespread cell death and increases microglial labeling in the neonatal mouse brain. Dev Neurobiol 2016; 77:753-766. [PMID: 27706925 DOI: 10.1002/dneu.22457] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2016] [Revised: 09/09/2016] [Accepted: 09/28/2016] [Indexed: 11/09/2022]
Abstract
Minocycline, an antibiotic of the tetracycline family, inhibits microglia in many paradigms and is among the most commonly used tools for examining the role of microglia in physiological processes. Microglia may play an active role in triggering developmental neuronal cell death, although findings have been contradictory. To determine whether microglia influence developmental cell death, we treated perinatal mice with minocycline (45 mg/kg) and quantified effects on dying cells and microglial labeling using immunohistochemistry for activated caspase-3 (AC3) and ionized calcium-binding adapter molecule 1 (Iba1), respectively. Contrary to our expectations, minocycline treatment from embryonic day 18 to postnatal day (P)1 caused a > tenfold increase in cell death 8 h after the last injection in all brain regions examined, including the primary sensory cortex, septum, hippocampus and hypothalamus. Iba1 labeling was also increased in most regions. Similar effects, although of smaller magnitude, were seen when treatment was delayed to P3-P5. Minocycline treatment from P3 to P5 also decreased overall cell number in the septum at weaning, suggesting lasting effects of the neonatal exposure. When administered at lower doses (4.5 or 22.5 mg/kg), or at the same dose 1 week later (P10-P12), minocycline no longer increased microglial markers or cell death. Taken together, the most commonly used microglial "inhibitor" increases cell death and Iba1 labeling in the neonatal mouse brain. Minocycline is used clinically in infant and pediatric populations; caution is warrented when using minocycline in developing animals, or extrapolating the effects of this drug across ages. © 2016 Wiley Periodicals, Inc. Develop Neurobiol 77: 753-766, 2017.
Collapse
Affiliation(s)
- J Alex Strahan
- Neuroscience Institute and Center for Behavioral Neuroscience, Georgia State University, Atlanta, Georgia, 30302
| | - William H Walker
- Neuroscience Institute and Center for Behavioral Neuroscience, Georgia State University, Atlanta, Georgia, 30302
| | - Taylor R Montgomery
- Neuroscience Institute and Center for Behavioral Neuroscience, Georgia State University, Atlanta, Georgia, 30302
| | - Nancy G Forger
- Neuroscience Institute and Center for Behavioral Neuroscience, Georgia State University, Atlanta, Georgia, 30302
| |
Collapse
|
44
|
Stewart CE, Corella KM, Samberg BD, Jones PT, Linscott ML, Chung WCJ. Perinatal midline astrocyte development is impaired in fibroblast growth factor 8 hypomorphic mice. Brain Res 2016; 1646:287-296. [PMID: 27291295 DOI: 10.1016/j.brainres.2016.06.015] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2015] [Revised: 05/27/2016] [Accepted: 06/09/2016] [Indexed: 12/31/2022]
Abstract
Our previous studies showed that Fgf8 mutations can cause Kallmann syndrome (KS), a form of congenital hypogonadotropic hypogonadism, in which patients do not undergo puberty and are infertile. Interestingly, some KS patients also have agenesis of the corpus callosum (ACC) suggesting that KS pathology is not limited to reproductive function. Here, we asked whether FGF8 dysfunction is the underlying cause of ACC in some KS patients. Indeed, early studies in transgenic mice with Fgf8 mutations reported the presence of failed or incomplete corpus callosum formation. Additional studies in transgenic mice showed that FGF8 function most likely prevents the prenatal elimination of glial fibrillary acidic protein (GFAP)-immunoreactive (IR) glial cells in the indusium griseum (IG) and midline zipper (MZ), two anterior-dorsal midline regions required for corpus callosum formation (i.e., between embryonic days (E) 15.5-18.5). Here, we tested the hypothesis that FGF8 function is critical for the survival of the GFAP-IR midline glial cells. First, we measured the incidence of apoptosis in the anterior-dorsal midline region in Fgf8 hypomorphic mice during embryonic corpus callosum formation. Second, we quantified the GFAP expression in the anterior-dorsal midbrain region during pre- and postnatal development, in order to study: 1) how Fgf8 hypomorphy disrupts prenatal GFAP-IR midline glial cell development, and 2) whether Fgf8 hypomorphy continues to disrupt postnatal GFAP-IR midline glial cell development. Our results indicate that perinatal FGF8 signaling is important for the timing of the onset of anterior-dorsal Gfap expression in midline glial cells suggesting that FGF8 function regulates midline GFAP-IR glial cell development, which when disrupted by Fgf8 deficiency prevents the formation of the corpus callosum. These studies provide an experimentally-based mechanistic explanation as to why corpus callosum formation may fail in KS patients with deficits in FGF signaling.
Collapse
Affiliation(s)
- Courtney E Stewart
- School of Biomedical Sciences, Kent State University, Kent, OH 44242, USA
| | - Kristina M Corella
- School of Biomedical Sciences, Kent State University, Kent, OH 44242, USA
| | - Brittany D Samberg
- Department of Biological Sciences, Kent State University, Kent, OH 44242, USA
| | - Paula T Jones
- Department of Biological Sciences, Kent State University, Kent, OH 44242, USA
| | - Megan L Linscott
- Department of Biological Sciences, Kent State University, Kent, OH 44242, USA
| | - Wilson C J Chung
- School of Biomedical Sciences, Kent State University, Kent, OH 44242, USA; Department of Biological Sciences, Kent State University, Kent, OH 44242, USA.
| |
Collapse
|
45
|
Forger NG. Epigenetic mechanisms in sexual differentiation of the brain and behaviour. Philos Trans R Soc Lond B Biol Sci 2016; 371:20150114. [PMID: 26833835 DOI: 10.1098/rstb.2015.0114] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/12/2015] [Indexed: 11/12/2022] Open
Abstract
Circumstantial evidence alone argues that the establishment and maintenance of sex differences in the brain depend on epigenetic modifications of chromatin structure. More direct evidence has recently been obtained from two types of studies: those manipulating a particular epigenetic mechanism, and those examining the genome-wide distribution of specific epigenetic marks. The manipulation of histone acetylation or DNA methylation disrupts the development of several neural sex differences in rodents. Taken together, however, the evidence suggests there is unlikely to be a simple formula for masculine or feminine development of the brain and behaviour; instead, underlying epigenetic mechanisms may vary by brain region or even by dependent variable within a region. Whole-genome studies related to sex differences in the brain have only very recently been reported, but suggest that males and females may use different combinations of epigenetic modifications to control gene expression, even in cases where gene expression does not differ between the sexes. Finally, recent findings are discussed that are likely to direct future studies on the role of epigenetic mechanisms in sexual differentiation of the brain and behaviour.
Collapse
Affiliation(s)
- Nancy G Forger
- Neuroscience Institute, Georgia State University, Atlanta, GA 30307, USA
| |
Collapse
|
46
|
Forger NG, Strahan JA, Castillo-Ruiz A. Cellular and molecular mechanisms of sexual differentiation in the mammalian nervous system. Front Neuroendocrinol 2016; 40:67-86. [PMID: 26790970 PMCID: PMC4897775 DOI: 10.1016/j.yfrne.2016.01.001] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2015] [Revised: 12/31/2015] [Accepted: 01/09/2016] [Indexed: 01/16/2023]
Abstract
Neuroscientists are likely to discover new sex differences in the coming years, spurred by the National Institutes of Health initiative to include both sexes in preclinical studies. This review summarizes the current state of knowledge of the cellular and molecular mechanisms underlying sex differences in the mammalian nervous system, based primarily on work in rodents. Cellular mechanisms examined include neurogenesis, migration, the differentiation of neurochemical and morphological cell phenotype, and cell death. At the molecular level we discuss evolving roles for epigenetics, sex chromosome complement, the immune system, and newly identified cell signaling pathways. We review recent findings on the role of the environment, as well as genome-wide studies with some surprising results, causing us to re-think often-used models of sexual differentiation. We end by pointing to future directions, including an increased awareness of the important contributions of tissues outside of the nervous system to sexual differentiation of the brain.
Collapse
Affiliation(s)
- Nancy G Forger
- Neuroscience Institute, Georgia State University, Atlanta, GA 30303, United States.
| | - J Alex Strahan
- Neuroscience Institute, Georgia State University, Atlanta, GA 30303, United States.
| | | |
Collapse
|
47
|
Neonatal testosterone suppresses a neuroendocrine pulse generator required for reproduction. Nat Commun 2015; 5:3285. [PMID: 24518793 DOI: 10.1038/ncomms4285] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2013] [Accepted: 01/20/2014] [Indexed: 01/21/2023] Open
Abstract
The pituitary gland releases hormones in a pulsatile fashion guaranteeing signalling efficiency. The determinants of pulsatility are poorly circumscribed. Here we show in magnocellular hypothalamo-neurohypophyseal oxytocin (OT) neurons that the bursting activity underlying the neurohormonal pulses necessary for parturition and the milk-ejection reflex is entirely driven by a female-specific central pattern generator (CPG). Surprisingly, this CPG is active in both male and female neonates, but is inactivated in males after the first week of life. CPG activity can be restored in males by orchidectomy or silenced in females by exogenous testosterone. This steroid effect is aromatase and caspase dependent, and is mediated via oestrogen receptor-α. This indicates the apoptosis of the CPG network during hypothalamic sexual differentiation, explaining why OT neurons do not burst in adult males. This supports the view that stereotypic neuroendocrine pulsatility is governed by CPGs, some of which are subjected to gender-specific perinatal programming.
Collapse
|
48
|
Bedont JL, Blackshaw S. Constructing the suprachiasmatic nucleus: a watchmaker's perspective on the central clockworks. Front Syst Neurosci 2015; 9:74. [PMID: 26005407 PMCID: PMC4424844 DOI: 10.3389/fnsys.2015.00074] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2014] [Accepted: 04/23/2015] [Indexed: 11/13/2022] Open
Abstract
The circadian system constrains an organism's palette of behaviors to portions of the solar day appropriate to its ecological niche. The central light-entrained clock in the suprachiasmatic nucleus (SCN) of the mammalian circadian system has evolved a complex network of interdependent signaling mechanisms linking multiple distinct oscillators to serve this crucial function. However, studies of the mechanisms controlling SCN development have greatly lagged behind our understanding of its physiological functions. We review advances in the understanding of adult SCN function, what has been described about SCN development to date, and the potential of both current and future studies of SCN development to yield important insights into master clock function, dysfunction, and evolution.
Collapse
Affiliation(s)
- Joseph L Bedont
- Department of Neuroscience, Johns Hopkins University School of Medicine Baltimore, MD, USA
| | - Seth Blackshaw
- Department of Neuroscience, Johns Hopkins University School of Medicine Baltimore, MD, USA ; Department of Ophthalmology, Johns Hopkins University School of Medicine Baltimore, MD, USA ; Department of Physiology, Johns Hopkins University School of Medicine Baltimore, MD, USA ; Department of Neurology, Johns Hopkins University School of Medicine Baltimore, MD, USA ; Center for High-Throughput Biology, Johns Hopkins University School of Medicine Baltimore, MD, USA
| |
Collapse
|
49
|
Ghahramani NM, Ngun TC, Chen PY, Tian Y, Krishnan S, Muir S, Rubbi L, Arnold AP, de Vries GJ, Forger NG, Pellegrini M, Vilain E. The effects of perinatal testosterone exposure on the DNA methylome of the mouse brain are late-emerging. Biol Sex Differ 2014; 5:8. [PMID: 24976947 PMCID: PMC4074311 DOI: 10.1186/2042-6410-5-8] [Citation(s) in RCA: 83] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2014] [Accepted: 05/22/2014] [Indexed: 02/07/2023] Open
Abstract
Background The biological basis for sex differences in brain function and disease susceptibility is poorly understood. Examining the role of gonadal hormones in brain sexual differentiation may provide important information about sex differences in neural health and development. Permanent masculinization of brain structure, function, and disease is induced by testosterone prenatally in males, but the possible mediation of these effects by long-term changes in the epigenome is poorly understood. Methods We investigated the organizational effects of testosterone on the DNA methylome and transcriptome in two sexually dimorphic forebrain regions—the bed nucleus of the stria terminalis/preoptic area and the striatum. To study the contribution of testosterone to both the establishment and persistence of sex differences in DNA methylation, we performed genome-wide surveys in male, female, and female mice given testosterone on the day of birth. Methylation was assessed during the perinatal window for testosterone's organizational effects and in adulthood. Results The short-term effect of testosterone exposure was relatively modest. However, in adult animals the number of genes whose methylation was altered had increased by 20-fold. Furthermore, we found that in adulthood, methylation at a substantial number of sexually dimorphic CpG sites was masculinized in response to neonatal testosterone exposure. Consistent with this, testosterone's effect on gene expression in the striatum was more apparent in adulthood. Conclusion Taken together, our data imply that the organizational effects of testosterone on the brain methylome and transcriptome are dramatic and late-emerging. Our findings offer important insights into the long-term molecular effects of early-life hormonal exposure.
Collapse
Affiliation(s)
- Negar M Ghahramani
- Department of Human Genetics, David Geffen School of Medicine at University of California Los Angeles (UCLA), Los Angeles, CA 90095, USA.,Laboratory of Neuroendocrinology of the Brain Research Institute, UCLA, Los Angeles, CA 90095, USA
| | - Tuck C Ngun
- Department of Human Genetics, David Geffen School of Medicine at University of California Los Angeles (UCLA), Los Angeles, CA 90095, USA.,Laboratory of Neuroendocrinology of the Brain Research Institute, UCLA, Los Angeles, CA 90095, USA
| | - Pao-Yang Chen
- Institute of Plant and Microbial Biology, Academia Sinica, Taipei 11529, Taiwan
| | - Yuan Tian
- Interdepartmental PhD Program in Bioinformatics, UCLA, Los Angeles, CA 90095, USA
| | - Sangitha Krishnan
- Department of Human Genetics, David Geffen School of Medicine at University of California Los Angeles (UCLA), Los Angeles, CA 90095, USA.,Laboratory of Neuroendocrinology of the Brain Research Institute, UCLA, Los Angeles, CA 90095, USA
| | - Stephanie Muir
- Department of Human Genetics, David Geffen School of Medicine at University of California Los Angeles (UCLA), Los Angeles, CA 90095, USA.,Laboratory of Neuroendocrinology of the Brain Research Institute, UCLA, Los Angeles, CA 90095, USA
| | - Liudmilla Rubbi
- Department of Molecular, Cellular, and Developmental Biology, UCLA, Los Angeles, CA 90095, USA
| | - Arthur P Arnold
- Laboratory of Neuroendocrinology of the Brain Research Institute, UCLA, Los Angeles, CA 90095, USA.,Department of Integrative Biology and Physiology, UCLA, Los Angeles, CA 90095, USA
| | - Geert J de Vries
- Neuroscience Institute, Georgia State University, Atlanta, GA 30303, USA
| | - Nancy G Forger
- Neuroscience Institute, Georgia State University, Atlanta, GA 30303, USA
| | - Matteo Pellegrini
- Department of Molecular, Cellular, and Developmental Biology, UCLA, Los Angeles, CA 90095, USA
| | - Eric Vilain
- Department of Human Genetics, David Geffen School of Medicine at University of California Los Angeles (UCLA), Los Angeles, CA 90095, USA.,Laboratory of Neuroendocrinology of the Brain Research Institute, UCLA, Los Angeles, CA 90095, USA.,Department of Human Genetics, UCLA, 695 Charles Young Drive South, Gonda Room 5506, Los Angeles, CA 90095-7088, USA
| |
Collapse
|
50
|
Irles C, Nava-Kopp AT, Morán J, Zhang L. Neonatal maternal separation up-regulates protein signalling for cell survival in rat hypothalamus. Stress 2014; 17:275-84. [PMID: 24730533 DOI: 10.3109/10253890.2014.913017] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
We have previously reported that in response to early life stress, such as maternal hyperthyroidism and maternal separation (MS), the rat hypothalamic vasopressinergic system becomes up-regulated, showing enlarged nuclear volume and cell number, with stress hyperresponsivity and high anxiety during adulthood. The detailed signaling pathways involving cell death/survival, modified by adverse experiences in this developmental window remains unknown. Here, we report the effects of MS on cellular density and time-dependent fluctuations of the expression of pro- and anti-apoptotic factors during the development of the hypothalamus. Neonatal male rats were exposed to 3 h-daily MS from postnatal days 2 to 15 (PND 2-15). Cellular density was assessed in the hypothalamus at PND 21 using methylene blue staining, and neuronal nuclear specific protein and glial fibrillary acidic protein immunostaining at PND 36. Expression of factors related to apoptosis and cell survival in the hypothalamus was examined at PND 1, 3, 6, 9, 12, 15, 20 and 43 by Western blot. Rats subjected to MS exhibited greater cell-density and increased neuronal density in all hypothalamic regions assessed. The time course of protein expression in the postnatal brain showed: (1) decreased expression of active caspase 3; (2) increased Bcl-2/Bax ratio; (3) increased activation of ERK1/2, Akt and inactivation of Bad; PND 15 and PND 20 were the most prominent time-points. These data indicate that MS can induce hypothalamic structural reorganization by promoting survival, suppressing cell death pathways, increasing cellular density which may alter the contribution of these modified regions to homeostasis.
Collapse
|