1
|
Stea DM, D’Alessio A. Caveolae: Metabolic Platforms at the Crossroads of Health and Disease. Int J Mol Sci 2025; 26:2918. [PMID: 40243482 PMCID: PMC11988808 DOI: 10.3390/ijms26072918] [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: 02/27/2025] [Revised: 03/14/2025] [Accepted: 03/21/2025] [Indexed: 04/18/2025] Open
Abstract
Caveolae are small flask-shaped invaginations of the plasma membrane enriched in cholesterol and sphingolipids. They play a critical role in various cellular processes, including signal transduction, endocytosis, and mechanotransduction. Caveolin proteins, specifically Cav-1, Cav-2, and Cav-3, in addition to their role as structural components of caveolae, have been found to regulate the activity of signaling molecules. A growing body of research has highlighted the pivotal role of caveolae and caveolins in maintaining cellular metabolic homeostasis. Indeed, studies have demonstrated that caveolins interact with the key components of insulin signaling, glucose uptake, and lipid metabolism, thereby influencing energy production and storage. The dysfunction of caveolae or the altered expression of caveolins has been associated with metabolic disorders, including obesity, type 2 diabetes, and ocular diseases. Remarkably, mutations in caveolin genes can disrupt cellular energy balance, promote oxidative stress, and exacerbate metabolic dysregulation. This review examines current research on the molecular mechanisms through which caveolae and caveolins regulate cellular metabolism, explores their involvement in the pathogenesis of metabolic disorders, and discusses potential therapeutic strategies targeting caveolin function and the stabilization of caveolae to restore metabolic homeostasis.
Collapse
Affiliation(s)
- Dante Maria Stea
- Facoltà di Medicina e Chirurgia, Università Cattolica del Sacro Cuore, 00168 Rome, Italy;
| | - Alessio D’Alessio
- Sezione di Istologia ed Embriologia, Dipartimento di Scienze della Vita e Sanità Pubblica, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
- Fondazione Policlinico Universitario “Agostino Gemelli”, IRCCS, 00168 Rome, Italy
| |
Collapse
|
2
|
Pauss SN, Bates EA, Martinez GJ, Bates ZT, Kipp ZA, Gipson CD, Hinds TD. Steroid receptors and coregulators: Dissemination of sex differences and emerging technologies. J Biol Chem 2025; 301:108363. [PMID: 40023399 PMCID: PMC11986243 DOI: 10.1016/j.jbc.2025.108363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Revised: 02/19/2025] [Accepted: 02/21/2025] [Indexed: 03/04/2025] Open
Abstract
Steroid receptors are ligand-induced transcription factors that have broad functions among all living animal species, ranging from control of sex differences, body weight, stress responses, and many others. Their binding to coregulator proteins is regulated by corepressors and coactivators that interchange upon stimulation with a ligand. Coregulator proteins are an imperative and understudied aspect of steroid receptor signaling. Here, we discuss steroid receptor basics from protein domain structures that allow them to interact with coregulators and other proteins, their essential functions as transcription factors, and other elemental protein-protein interactions. We deliberate about the mechanisms that coregulators control in steroid receptor signaling, sex hormone signaling differences, sex hormone treatment in the opposite sex, and how these affect the coregulator and sex steroid receptor complexes. The steroid receptor-coregulator signaling mechanisms are essential built-in components of the mammalian DNA that mediate physiological and everyday functions. Targeting their crosstalk might be useful when imbalances lead to disease. We introduce novel technologies, such as the PamGene PamStation, which make investigating the heterogeneity of the steroid receptor-coregulator complexes and targeting their binding more feasible. This review provides an extensive understanding of steroid receptor-coregulator signaling and how these interactions are intrinsic to many physiological functions that may offer therapeutic advantages.
Collapse
Affiliation(s)
- Sally N Pauss
- Drug & Disease Discovery D3 Research Center, Department of Pharmacology and Nutritional Sciences, University of Kentucky College of Medicine, Lexington, Kentucky, USA
| | - Evelyn A Bates
- Drug & Disease Discovery D3 Research Center, Department of Pharmacology and Nutritional Sciences, University of Kentucky College of Medicine, Lexington, Kentucky, USA
| | - Genesee J Martinez
- Drug & Disease Discovery D3 Research Center, Department of Pharmacology and Nutritional Sciences, University of Kentucky College of Medicine, Lexington, Kentucky, USA
| | - Zane T Bates
- Department of Bioengineering, University of Toledo College of Engineering, Toledo, Ohio, USA
| | - Zachary A Kipp
- Drug & Disease Discovery D3 Research Center, Department of Pharmacology and Nutritional Sciences, University of Kentucky College of Medicine, Lexington, Kentucky, USA
| | - Cassandra D Gipson
- Drug & Disease Discovery D3 Research Center, Department of Pharmacology and Nutritional Sciences, University of Kentucky College of Medicine, Lexington, Kentucky, USA
| | - Terry D Hinds
- Drug & Disease Discovery D3 Research Center, Department of Pharmacology and Nutritional Sciences, University of Kentucky College of Medicine, Lexington, Kentucky, USA; Markey Cancer Center, University of Kentucky, Lexington, Kentucky, USA; Barnstable Brown Diabetes Center, University of Kentucky College of Medicine, Lexington, Kentucky, USA.
| |
Collapse
|
3
|
Valero-Ochando J, Cantó A, López-Pedrajas R, Almansa I, Miranda M. Role of Gonadal Steroid Hormones in the Eye: Therapeutic Implications. Biomolecules 2024; 14:1262. [PMID: 39456195 PMCID: PMC11506707 DOI: 10.3390/biom14101262] [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: 08/31/2024] [Revised: 10/01/2024] [Accepted: 10/03/2024] [Indexed: 10/28/2024] Open
Abstract
Gonadal steroid hormones are critical regulatory substances involved in various developmental and physiological processes from fetal development through adulthood. These hormones, derived from cholesterol, are synthesized primarily by the gonads, adrenal cortex, and placenta. The synthesis of these hormones involves a series of enzymatic steps starting in the mitochondria and includes enzymes such as cytochrome P450 and aromatase. Beyond their genomic actions, which involve altering gene transcription over hours, gonadal steroids also exhibit rapid, nongenomic effects through receptors located on the cell membrane. Additionally, recent research has highlighted the role of these hormones in the central nervous system (CNS). However, the interactions between gonadal steroid hormones and the retina have received limited attention, though it has been suggested that they may play a protective role in retinal diseases. This review explores the synthesis of gonadal hormones, their mechanisms of action, and their potential implications in various retinal and optic nerve diseases, such as glaucoma, age-related macular degeneration (AMD), diabetic retinopathy (DR), or retinitis pigmentosa (RP), discussing both protective and risk factors associated with hormone levels and their therapeutic potential.
Collapse
Affiliation(s)
| | | | | | | | - María Miranda
- Department of Biomedical Sciences, Faculty of Health Sciences, Institute of Biomedical Sciences, Cardenal Herrera-CEU University, CEU Universities, 46115 Valencia, Spain; (J.V.-O.); (A.C.); (R.L.-P.); (I.A.)
| |
Collapse
|
4
|
Davis D, Dovey J, Sagoshi S, Thaweepanyaporn K, Ogawa S, Vasudevan N. Steroid hormone-mediated regulation of sexual and aggressive behaviour by non-genomic signalling. Steroids 2023; 200:109324. [PMID: 37820890 DOI: 10.1016/j.steroids.2023.109324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 10/02/2023] [Accepted: 10/04/2023] [Indexed: 10/13/2023]
Abstract
Sex and aggression are well studied examples of social behaviours that are common to most animals and are mediated by an evolutionary conserved group of interconnected nuclei in the brain called the social behaviour network. Though glucocorticoids and in particular estrogen regulate these social behaviours, their effects in the brain are generally thought to be mediated by genomic signalling, a slow transcriptional regulation mediated by nuclear hormone receptors. In the last decade or so, there has been renewed interest in understanding the physiological significance of rapid, non-genomic signalling mediated by steroids. Though the identity of the membrane hormone receptors that mediate this signalling is not clearly understood and appears to be different in different cell types, such signalling contributes to physiologically relevant behaviours such as sex and aggression. In this short review, we summarise the evidence for this phenomenon in the rodent, by focusing on estrogen and to some extent, glucocorticoid signalling. The use of these signals, in relation to genomic signalling is manifold and ranges from potentiation of transcription to the possible transduction of environmental signals.
Collapse
Affiliation(s)
- DeAsia Davis
- School of Biological Sciences, University of Reading, United Kingdom
| | - Janine Dovey
- School of Biological Sciences, University of Reading, United Kingdom
| | - Shoko Sagoshi
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, United States; Laboratory of Behavioural Neuroendocrinology, University of Tsukuba, Tsukuba, Japan
| | | | - Sonoko Ogawa
- Laboratory of Behavioural Neuroendocrinology, University of Tsukuba, Tsukuba, Japan
| | - Nandini Vasudevan
- School of Biological Sciences, University of Reading, United Kingdom.
| |
Collapse
|
5
|
Samhan-Arias AK, Poejo J, Marques-da-Silva D, Martínez-Costa OH, Gutierrez-Merino C. Hexa-Histidine, a Peptide with Versatile Applications in the Study of Amyloid-β(1-42) Molecular Mechanisms of Action. Molecules 2023; 28:7138. [PMID: 37894616 PMCID: PMC10708093 DOI: 10.3390/molecules28237909] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 10/10/2023] [Accepted: 10/13/2023] [Indexed: 12/18/2023] Open
Abstract
Amyloid β (Aβ) oligomers are the most neurotoxic forms of Aβ, and Aβ(1-42) is the prevalent Aβ peptide found in the amyloid plaques of Alzheimer's disease patients. Aβ(25-35) is the shortest peptide that retains the toxicity of Aβ(1-42). Aβ oligomers bind to calmodulin (CaM) and calbindin-D28k with dissociation constants in the nanomolar Aβ(1-42) concentration range. Aβ and histidine-rich proteins have a high affinity for transition metal ions Cu2+, Fe3+ and Zn2+. In this work, we show that the fluorescence of Aβ(1-42) HiLyteTM-Fluor555 can be used to monitor hexa-histidine peptide (His6) interaction with Aβ(1-42). The formation of His6/Aβ(1-42) complexes is also supported by docking results yielded by the MDockPeP Server. Also, we found that micromolar concentrations of His6 block the increase in the fluorescence of Aβ(1-42) HiLyteTM-Fluor555 produced by its interaction with the proteins CaM and calbindin-D28k. In addition, we found that the His6-tag provides a high-affinity site for the binding of Aβ(1-42) and Aβ(25-35) peptides to the human recombinant cytochrome b5 reductase, and sensitizes this enzyme to inhibition by these peptides. In conclusion, our results suggest that a His6-tag could provide a valuable new tool to experimentally direct the action of neurotoxic Aβ peptides toward selected cellular targets.
Collapse
Affiliation(s)
- Alejandro K. Samhan-Arias
- Departamento de Bioquímica, Universidad Autónoma de Madrid (UAM), C/Arturo Duperier 4, 28029 Madrid, Spain;
- Instituto de Investigaciones Biomédicas ‘Sols-Morreale’ (CSIC-UAM), C/Arturo Duperier 4, 28029 Madrid, Spain
| | - Joana Poejo
- Instituto de Biomarcadores de Patologías Moleculares, Universidad de Extremadura, 06006 Badajoz, Spain;
| | - Dorinda Marques-da-Silva
- LSRE—Laboratory of Separation and Reaction Engineering and LCM—Laboratory of Catalysis and Materials, School of Management and Technology, Polytechnic Institute of Leiria, Morro do Lena-Alto do Vieiro, 2411-901 Leiria, Portugal;
- ALiCE—Associate Laboratory in Chemical Engineering, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal
- School of Technology and Management, Polytechnic Institute of Leiria, Morro do Lena-Alto do Vieiro, 2411-901 Leiria, Portugal
| | - Oscar H. Martínez-Costa
- Departamento de Bioquímica, Universidad Autónoma de Madrid (UAM), C/Arturo Duperier 4, 28029 Madrid, Spain;
- Instituto de Investigaciones Biomédicas ‘Sols-Morreale’ (CSIC-UAM), C/Arturo Duperier 4, 28029 Madrid, Spain
| | - Carlos Gutierrez-Merino
- Instituto de Biomarcadores de Patologías Moleculares, Universidad de Extremadura, 06006 Badajoz, Spain;
| |
Collapse
|
6
|
Anirudhan A, Mattethra GC, Alzahrani KJ, Banjer HJ, Alzahrani FM, Halawani IF, Patil S, Sharma A, Paramasivam P, Ahmed SSSJ. Eleven Crucial Pesticides Appear to Regulate Key Genes That Link MPTP Mechanism to Cause Parkinson's Disease through the Selective Degeneration of Dopamine Neurons. Brain Sci 2023; 13:1003. [PMID: 37508933 PMCID: PMC10377611 DOI: 10.3390/brainsci13071003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 06/23/2023] [Accepted: 06/26/2023] [Indexed: 07/30/2023] Open
Abstract
Pesticides kill neurons, but the mechanism leading to selective dopaminergic loss in Parkinson's disease (PD) is unknown. Understanding the pesticide's effect on dopaminergic neurons (DA) can help to screen and treat PD. The critical uptake of pesticides by the membrane receptors at DA is hypothesized to activate a signaling cascade and accelerate degeneration. Using MPTP as a reference, we demonstrate the mechanisms of eleven crucial pesticides through molecular docking, protein networks, regulatory pathways, and prioritization of key pesticide-regulating proteins. Participants were recruited and grouped into control and PD based on clinical characteristics as well as pesticide traces in their blood plasma. Then, qPCR was used to measure pesticide-associated gene expression in peripheral blood mononuclear cells between groups. As a result of molecular docking, all eleven pesticides and the MPTP showed high binding efficiency against 274 membrane receptor proteins of DA. Further, the protein interaction networks showed activation of multiple signaling cascades through these receptors. Subsequent analysis revealed 31 biological pathways shared by all 11pesticides and MPTP that were overrepresented by 46 crucial proteins. Among these, CTNNB1, NDUFS6, and CAV1 were prioritized to show a significant change in gene expression in pesticide-exposed PD which guides toward therapy.
Collapse
Affiliation(s)
- Athira Anirudhan
- Central Research Laboratory, Believers Church Medical College Hospital, Kuttapuzha, Thiruvalla 689103, Kerala, India
| | - George Chandy Mattethra
- Central Research Laboratory, Believers Church Medical College Hospital, Kuttapuzha, Thiruvalla 689103, Kerala, India
| | - Khalid J Alzahrani
- Department of Clinical Laboratories Sciences, College of Applied Medical Sciences, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia
| | - Hamsa Jameel Banjer
- Department of Clinical Laboratories Sciences, College of Applied Medical Sciences, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia
| | - Fuad M Alzahrani
- Department of Clinical Laboratories Sciences, College of Applied Medical Sciences, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia
| | - Ibrahim F Halawani
- Department of Clinical Laboratories Sciences, College of Applied Medical Sciences, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia
| | - Shankargouda Patil
- College of Dental Medicine, Roseman University of Health Sciences, South Jordan, UT 84095, USA
| | - Ashutosh Sharma
- Regional Department of Bioengineering, NatProLab-Plant Innovation Lab, Tecnologico de Monterrey, Queretaro 76130, Mexico
| | - Prabu Paramasivam
- School of Medicine, Department of Neurology, University of New Mexico Health Sciences Center, University of New Mexico, Albuquerque, NM 87131, USA
| | - Shiek S S J Ahmed
- Drug Discovery & Omics Lab, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Kelambakkam 603103, Tamil Nadu, India
| |
Collapse
|
7
|
Wnuk A, Przepiórska K, Pietrzak BA, Kajta M. Emerging Evidence on Membrane Estrogen Receptors as Novel Therapeutic Targets for Central Nervous System Pathologies. Int J Mol Sci 2023; 24:ijms24044043. [PMID: 36835454 PMCID: PMC9968034 DOI: 10.3390/ijms24044043] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 02/06/2023] [Accepted: 02/15/2023] [Indexed: 02/19/2023] Open
Abstract
Nuclear- and membrane-initiated estrogen signaling cooperate to orchestrate the pleiotropic effects of estrogens. Classical estrogen receptors (ERs) act transcriptionally and govern the vast majority of hormonal effects, whereas membrane ERs (mERs) enable acute modulation of estrogenic signaling and have recently been shown to exert strong neuroprotective capacity without the negative side effects associated with nuclear ER activity. In recent years, GPER1 was the most extensively characterized mER. Despite triggering neuroprotective effects, cognitive improvements, and vascular protective effects and maintaining metabolic homeostasis, GPER1 has become the subject of controversy, particularly due to its participation in tumorigenesis. This is why interest has recently turned toward non-GPER-dependent mERs, namely, mERα and mERβ. According to available data, non-GPER-dependent mERs elicit protective effects against brain damage, synaptic plasticity impairment, memory and cognitive dysfunctions, metabolic imbalance, and vascular insufficiency. We postulate that these properties are emerging platforms for designing new therapeutics that may be used in the treatment of stroke and neurodegenerative diseases. Since mERs have the ability to interfere with noncoding RNAs and to regulate the translational status of brain tissue by affecting histones, non-GPER-dependent mERs appear to be attractive targets for modern pharmacotherapy for nervous system diseases.
Collapse
Affiliation(s)
- Agnieszka Wnuk
- Correspondence: (A.W.); (M.K.); Tel.: +48-12-662-3339 (A.W.); +48-12-662-3235 (M.K.); Fax: +48-12-637-4500 (A.W. & M.K.)
| | | | | | - Małgorzata Kajta
- Correspondence: (A.W.); (M.K.); Tel.: +48-12-662-3339 (A.W.); +48-12-662-3235 (M.K.); Fax: +48-12-637-4500 (A.W. & M.K.)
| |
Collapse
|
8
|
Johnson CS, Mermelstein PG. The interaction of membrane estradiol receptors and metabotropic glutamate receptors in adaptive and maladaptive estradiol-mediated motivated behaviors in females. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2023; 168:33-91. [PMID: 36868633 DOI: 10.1016/bs.irn.2022.11.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Estrogen receptors were initially identified as intracellular, ligand-regulated transcription factors that result in genomic change upon ligand binding. However, rapid estrogen receptor signaling initiated outside of the nucleus was also known to occur via mechanisms that were less clear. Recent studies indicate that these traditional receptors, estrogen receptor α and estrogen receptor β, can also be trafficked to act at the surface membrane. Signaling cascades from these membrane-bound estrogen receptors (mERs) can rapidly alter cellular excitability and gene expression, particularly through the phosphorylation of CREB. A principal mechanism of neuronal mER action has been shown to occur through glutamate-independent transactivation of metabotropic glutamate receptors (mGlu), which elicits multiple signaling outcomes. The interaction of mERs with mGlu has been shown to be important in many diverse functions in females, including driving motivated behaviors. Experimental evidence suggests that a large part of estradiol-induced neuroplasticity and motivated behaviors, both adaptive and maladaptive, occurs through estradiol-dependent mER activation of mGlu. Herein we will review signaling through estrogen receptors, both "classical" nuclear receptors and membrane-bound receptors, as well as estradiol signaling through mGlu. We will focus on how the interactions of these receptors and their downstream signaling cascades are involved in driving motivated behaviors in females, discussing a representative adaptive motivated behavior (reproduction) and maladaptive motivated behavior (addiction).
Collapse
Affiliation(s)
- Caroline S Johnson
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, United States
| | - Paul G Mermelstein
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, United States.
| |
Collapse
|
9
|
Enyong EN, Gurley JM, De Ieso ML, Stamer WD, Elliott MH. Caveolar and non-Caveolar Caveolin-1 in ocular homeostasis and disease. Prog Retin Eye Res 2022; 91:101094. [PMID: 35729002 PMCID: PMC9669151 DOI: 10.1016/j.preteyeres.2022.101094] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 06/03/2022] [Accepted: 06/10/2022] [Indexed: 11/17/2022]
Abstract
Caveolae, specialized plasma membrane invaginations present in most cell types, play important roles in multiple cellular processes including cell signaling, lipid uptake and metabolism, endocytosis and mechanotransduction. They are found in almost all cell types but most abundant in endothelial cells, adipocytes and fibroblasts. Caveolin-1 (Cav1), the signature structural protein of caveolae was the first protein associated with caveolae, and in association with Cavin1/PTRF is required for caveolae formation. Genetic ablation of either Cav1 or Cavin1/PTRF downregulates expression of the other resulting in loss of caveolae. Studies using Cav1-deficient mouse models have implicated caveolae with human diseases such as cardiomyopathies, lipodystrophies, diabetes and muscular dystrophies. While caveolins and caveolae are extensively studied in extra-ocular settings, their contributions to ocular function and disease pathogenesis are just beginning to be appreciated. Several putative caveolin/caveolae functions are relevant to the eye and Cav1 is highly expressed in retinal vascular and choroidal endothelium, Müller glia, the retinal pigment epithelium (RPE), and the Schlemm's canal endothelium and trabecular meshwork cells. Variants at the CAV1/2 gene locus are associated with risk of primary open angle glaucoma and the high risk HTRA1 variant for age-related macular degeneration is thought to exert its effect through regulation of Cav1 expression. Caveolins also play important roles in modulating retinal neuroinflammation and blood retinal barrier permeability. In this article, we describe the current state of caveolin/caveolae research in the context of ocular function and pathophysiology. Finally, we discuss new evidence showing that retinal Cav1 exists and functions outside caveolae.
Collapse
Affiliation(s)
- Eric N Enyong
- Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Department of Ophthalmology, Dean A. McGee Eye Institute, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Jami M Gurley
- Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Department of Ophthalmology, Dean A. McGee Eye Institute, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Michael L De Ieso
- Department of Ophthalmology, Duke Eye Center, Duke University, Durham, NC, USA
| | - W Daniel Stamer
- Department of Ophthalmology, Duke Eye Center, Duke University, Durham, NC, USA
| | - Michael H Elliott
- Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Department of Ophthalmology, Dean A. McGee Eye Institute, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
| |
Collapse
|
10
|
Mauvais-Jarvis F, Lange CA, Levin ER. Membrane-Initiated Estrogen, Androgen, and Progesterone Receptor Signaling in Health and Disease. Endocr Rev 2022; 43:720-742. [PMID: 34791092 PMCID: PMC9277649 DOI: 10.1210/endrev/bnab041] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Indexed: 12/15/2022]
Abstract
Rapid effects of steroid hormones were discovered in the early 1950s, but the subject was dominated in the 1970s by discoveries of estradiol and progesterone stimulating protein synthesis. This led to the paradigm that steroid hormones regulate growth, differentiation, and metabolism via binding a receptor in the nucleus. It took 30 years to appreciate not only that some cellular functions arise solely from membrane-localized steroid receptor (SR) actions, but that rapid sex steroid signaling from membrane-localized SRs is a prerequisite for the phosphorylation, nuclear import, and potentiation of the transcriptional activity of nuclear SR counterparts. Here, we provide a review and update on the current state of knowledge of membrane-initiated estrogen (ER), androgen (AR) and progesterone (PR) receptor signaling, the mechanisms of membrane-associated SR potentiation of their nuclear SR homologues, and the importance of this membrane-nuclear SR collaboration in physiology and disease. We also highlight potential clinical implications of pathway-selective modulation of membrane-associated SR.
Collapse
Affiliation(s)
- Franck Mauvais-Jarvis
- Department of Medicine, Section of Endocrinology and Metabolism, Tulane University School of Medicine, New Orleans, LA, 70112, USA.,Tulane Center of Excellence in Sex-Based Biology & Medicine, New Orleans, LA, 70112, USA.,Southeast Louisiana Veterans Affairs Medical Center, New Orleans, LA, 70119, USA
| | - Carol A Lange
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA.,Department of Medicine (Division of Hematology, Oncology, and Transplantation), University of Minnesota, Minneapolis, MN 55455, USA.,Department of Pharmacology, University of Minnesota, Minneapolis, MN 55455, USA
| | - Ellis R Levin
- Division of Endocrinology, Department of Medicine, University of California, Irvine, Irvine, CA, 92697, USA.,Department of Veterans Affairs Medical Center, Long Beach, Long Beach, CA, 90822, USA
| |
Collapse
|
11
|
Estradiol and Estrogen-like Alternative Therapies in Use: The Importance of the Selective and Non-Classical Actions. Biomedicines 2022; 10:biomedicines10040861. [PMID: 35453610 PMCID: PMC9029610 DOI: 10.3390/biomedicines10040861] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 04/03/2022] [Accepted: 04/04/2022] [Indexed: 12/17/2022] Open
Abstract
Estrogen is one of the most important female sex hormones, and is indispensable for reproduction. However, its role is much wider. Among others, due to its neuroprotective effects, estrogen protects the brain against dementia and complications of traumatic injury. Previously, it was used mainly as a therapeutic option for influencing the menstrual cycle and treating menopausal symptoms. Unfortunately, hormone replacement therapy might be associated with detrimental side effects, such as increased risk of stroke and breast cancer, raising concerns about its safety. Thus, tissue-selective and non-classical estrogen analogues have become the focus of interest. Here, we review the current knowledge about estrogen effects in a broader sense, and the possibility of using selective estrogen-receptor modulators (SERMs), selective estrogen-receptor downregulators (SERDs), phytoestrogens, and activators of non-genomic estrogen-like signaling (ANGELS) molecules as treatment.
Collapse
|
12
|
Sepúlveda D, Cisternas-Olmedo M, Arcos J, Nassif M, Vidal RL. Contribution of Autophagy-Lysosomal Pathway in the Exosomal Secretion of Alpha-Synuclein and Its Impact in the Progression of Parkinson’s Disease. Front Mol Neurosci 2022; 15:805087. [PMID: 35250476 PMCID: PMC8891570 DOI: 10.3389/fnmol.2022.805087] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Accepted: 01/07/2022] [Indexed: 01/07/2023] Open
Abstract
Parkinson’s disease (PD) is caused by the degeneration of dopaminergic neurons due to an accumulation of intraneuronal abnormal alpha-synuclein (α-syn) protein aggregates. It has been reported that the levels of exosomal α-syn of neuronal origin in plasma correlate significantly with motor dysfunction, highlighting the exosomes containing α-syn as a potential biomarker of PD. In addition, it has been found that the selective autophagy-lysosomal pathway (ALP) contributes to the secretion of misfolded proteins involved in neurodegenerative diseases. In this review, we describe the evidence that supports the relationship between the ALP and α-syn exosomal secretion on the PD progression and its implications in the diagnosis and progression of this pathology.
Collapse
Affiliation(s)
- Denisse Sepúlveda
- Center for Integrative Biology, Facultad de Ciencias, Universidad Mayor, Santiago, Chile
- Biomedical Neuroscience Institute, University of Chile, Santiago, Chile
- Geroscience Center for Brain Health and Metabolism, Santiago, Chile
| | - Marisol Cisternas-Olmedo
- Center for Integrative Biology, Facultad de Ciencias, Universidad Mayor, Santiago, Chile
- Biomedical Neuroscience Institute, University of Chile, Santiago, Chile
- Geroscience Center for Brain Health and Metabolism, Santiago, Chile
| | - Javiera Arcos
- Center for Integrative Biology, Facultad de Ciencias, Universidad Mayor, Santiago, Chile
- Biomedical Neuroscience Institute, University of Chile, Santiago, Chile
- Geroscience Center for Brain Health and Metabolism, Santiago, Chile
| | - Melissa Nassif
- Center for Integrative Biology, Facultad de Ciencias, Universidad Mayor, Santiago, Chile
- Escuela de Biotecnología, Facultad de Ciencias, Universidad Mayor, Santiago, Chile
| | - René L. Vidal
- Center for Integrative Biology, Facultad de Ciencias, Universidad Mayor, Santiago, Chile
- Biomedical Neuroscience Institute, University of Chile, Santiago, Chile
- Geroscience Center for Brain Health and Metabolism, Santiago, Chile
- *Correspondence: René L. Vidal,
| |
Collapse
|
13
|
Peart DR, Andrade AK, Logan CN, Knackstedt LA, Murray JE. Regulation of Cocaine-related Behaviors by Estrogen and Progesterone. Neurosci Biobehav Rev 2022; 135:104584. [DOI: 10.1016/j.neubiorev.2022.104584] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 01/30/2022] [Accepted: 02/12/2022] [Indexed: 10/19/2022]
|
14
|
Johnson CS, Micevych PE, Mermelstein PG. Membrane estrogen signaling in female reproduction and motivation. Front Endocrinol (Lausanne) 2022; 13:1009379. [PMID: 36246891 PMCID: PMC9557733 DOI: 10.3389/fendo.2022.1009379] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Accepted: 09/13/2022] [Indexed: 01/13/2023] Open
Abstract
Estrogen receptors were initially identified in the uterus, and later throughout the brain and body as intracellular, ligand-regulated transcription factors that affect genomic change upon ligand binding. However, rapid estrogen receptor signaling initiated outside of the nucleus was also known to occur via mechanisms that were less clear. Recent studies indicate that these traditional receptors, estrogen receptor-α and estrogen receptor-β, can also be trafficked to act at the surface membrane. Signaling cascades from these membrane-bound estrogen receptors (mERs) not only rapidly effect cellular excitability, but can and do ultimately affect gene expression, as seen through the phosphorylation of CREB. A principal mechanism of neuronal mER action is through glutamate-independent transactivation of metabotropic glutamate receptors (mGluRs), which elicits multiple signaling outcomes. The interaction of mERs with mGluRs has been shown to be important in many diverse functions in females, including, but not limited to, reproduction and motivation. Here we review membrane-initiated estrogen receptor signaling in females, with a focus on the interactions between these mERs and mGluRs.
Collapse
Affiliation(s)
- Caroline S. Johnson
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, United States
- *Correspondence: Caroline S. Johnson,
| | - Paul E Micevych
- Laboratory of Neuroendocrinology, Department of Neurobiology, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA, United States
| | - Paul G. Mermelstein
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, United States
| |
Collapse
|
15
|
Maher EE, Overby PF, Bull AH, Beckmann JS, Leyrer-Jackson JM, Koebele SV, Bimonte-Nelson HA, Gipson CD. Natural and synthetic estrogens specifically alter nicotine demand and cue-induced nicotine seeking in female rats. Neuropharmacology 2021; 198:108756. [PMID: 34416269 DOI: 10.1016/j.neuropharm.2021.108756] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 08/05/2021] [Accepted: 08/14/2021] [Indexed: 11/17/2022]
Abstract
Women have more difficulty maintaining smoking cessation than men, and experience greater withdrawal symptomatology as well as higher prevalence of relapse. Further, currently available treatments for smoking cessation, such as the nicotine patch and varenicline, have been shown to be less effective in women. Fluctuations in ovarian hormones across the menstrual cycle can affect craving and smoking relapse propensity. In addition, many women who smoke use some form of oral contraceptives, which most often contain ethinyl estradiol (EE), a synthetic, orally bio-available estrogen that is currently prescribed to women chronically and has been shown to alter smoking reward in women. The current study examined the impact of 17β-estradiol (E2), the prominent endogenous form of the steroid hormone estrogen, as well as EE, on nicotine self-administration, demand, and reinstatement following ovariectomy (OVX) or sham surgery. OVX vehicle-treated female rats consumed less nicotine, had lower intensity of demand, and reinstated less compared to sham vehicle-treated female rats. OVX-E2 and OVX-EE treatment groups showed a rebound of nicotine intake later in training, and Q0 levels of consumption were partially rescued in both groups. Further, E2 but not EE reversed the abolishment of reinstated nicotine seeking induced by OVX. Taken together, these results demonstrate that natural and synthetic estrogens play a critical role in mediating the neurobehavioral effects of nicotine, and future studies are essential for our understanding of how synthetic hormones contained within oral contraceptives interact with smoking.
Collapse
Affiliation(s)
- Erin E Maher
- Department of Family and Community Medicine, University of Kentucky, Lexington, KY, USA
| | - Paula F Overby
- Department of Psychology, Arizona State University, Tempe, AZ, USA
| | - Amanda H Bull
- Department of Psychology, Arizona State University, Tempe, AZ, USA
| | - Joshua S Beckmann
- Department of Psychology, University of Kentucky, Lexington, KY, USA
| | | | - Stephanie V Koebele
- Department of Psychology, Arizona State University, Tempe, AZ, USA; Arizonia Alzheimer's Consortium, Phoenix, AZ, USA
| | - Heather A Bimonte-Nelson
- Department of Psychology, Arizona State University, Tempe, AZ, USA; Arizonia Alzheimer's Consortium, Phoenix, AZ, USA
| | - Cassandra D Gipson
- Department of Family and Community Medicine, University of Kentucky, Lexington, KY, USA.
| |
Collapse
|
16
|
Joshua Cohen D, ElBaradie K, Boyan BD, Schwartz Z. Sex-specific effects of 17β-estradiol and dihydrotestosterone (DHT) on growth plate chondrocytes are dependent on both ERα and ERβ and require palmitoylation to translocate the receptors to the plasma membrane. Biochim Biophys Acta Mol Cell Biol Lipids 2021; 1866:159028. [PMID: 34416391 DOI: 10.1016/j.bbalip.2021.159028] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 07/19/2021] [Accepted: 08/13/2021] [Indexed: 11/24/2022]
Abstract
Rat costochondral cartilage growth plate chondrocytes exhibit cell sex-specific responses to 17β-estradiol (E2), testosterone, and dihydrotestosterone (DHT). Mechanistically, E2 and DHT stimulate proliferation and extracellular matrix synthesis in chondrocytes from female and male rats, respectively, by signaling through protein kinase C (PKC) and phospholipase C (PLC). Estrogen receptors (ERα; ERβ) and androgen receptors (ARs) are present in both male and female cells, but it is not known whether they interact to elicit sex-specific signaling. We used specific agonists and antagonists of these receptors to examine the relative contributions of ERs and ARs in membrane-mediated E2 signaling in female chondrocytes and DHT signaling in male chondrocytes. PKC activity in female chondrocytes was stimulated by agonists of ERα and ERβ and required intact caveolae; PKC activity was inhibited by the E2 enantiomer and by an inhibitor of ERβ. Western blots of cell lysates co-immunoprecipitated for ERα suggested the formation of a complex containing both ERα and ERß with E2 treatment. DHT and DHT agonists activated PKC in male cells, while AR inhibition blocked the stimulatory effect of DHT on PKC. Inhibition of ERα and ERβ also blocked PKC activation by DHT. Western blots of whole-cell lysates, plasma membranes, and caveolae indicated the translocation of AR to the plasma membrane and specifically to caveolae with DHT treatment. These results suggest that E2 and DHT promote chondrocyte differentiation via the ability of ARs and ERs to form a complex. The results also indicate that intact caveolae and palmitoylation of the membrane receptor(s) or membrane receptor complex containing ERα and ERβ is required for E2 and DHT membrane-associated PKC activity in costochondral cartilage cells.
Collapse
Affiliation(s)
- D Joshua Cohen
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA 23284, USA
| | - Khairat ElBaradie
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA 30033, USA
| | - Barbara D Boyan
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA 23284, USA; Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA 30033, USA.
| | - Zvi Schwartz
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA 23284, USA; Department of Periodontics, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| |
Collapse
|
17
|
Ha TY, Choi YR, Noh HR, Cha SH, Kim JB, Park SM. Age-related increase in caveolin-1 expression facilitates cell-to-cell transmission of α-synuclein in neurons. Mol Brain 2021; 14:122. [PMID: 34321069 PMCID: PMC8320051 DOI: 10.1186/s13041-021-00834-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 07/20/2021] [Indexed: 02/07/2023] Open
Abstract
Parkinson's disease (PD) is the second most prevalent neurodegenerative disease, with aging being considered the greatest risk factor for developing PD. Caveolin-1 (Cav-1) is known to participate in the aging process. Recent evidence indicates that prion-like propagation of misfolded α-synuclein (α-syn) released from neurons to neighboring neurons plays an important role in PD progression. In the present study, we demonstrated that cav-1 expression in the brain increased with age, and considerably increased in the brain of A53T α-syn transgenic mice. Cav-1 overexpression facilitated the uptake of α-syn into neurons and formation of additional Lewy body-like inclusion bodies, phosphorylation of cav-1 at tyrosine 14 was found to be crucial for this process. This study demonstrates the relationship between age and α-syn spread and will facilitate our understanding of the molecular mechanism of the cell-to-cell transmission of α-syn.
Collapse
Affiliation(s)
- Tae-Young Ha
- Department of Pharmacology, Ajou University School of Medicine, 164, Worldcup-ro, Yeongtong-gu, Suwon, 16499, Korea
- Center for Convergence Research of Neurological Disorders, Ajou University School of Medicine, Suwon, Korea
| | - Yu Ree Choi
- Department of Pharmacology, Ajou University School of Medicine, 164, Worldcup-ro, Yeongtong-gu, Suwon, 16499, Korea
- Center for Convergence Research of Neurological Disorders, Ajou University School of Medicine, Suwon, Korea
- Department of Biomedical Sciences, Ajou University School of Medicine, Suwon, Korea
| | - Hye Rin Noh
- Department of Pharmacology, Ajou University School of Medicine, 164, Worldcup-ro, Yeongtong-gu, Suwon, 16499, Korea
- Center for Convergence Research of Neurological Disorders, Ajou University School of Medicine, Suwon, Korea
- Department of Biomedical Sciences, Ajou University School of Medicine, Suwon, Korea
| | - Seon-Heui Cha
- Department of Pharmacology, Ajou University School of Medicine, 164, Worldcup-ro, Yeongtong-gu, Suwon, 16499, Korea
- Center for Convergence Research of Neurological Disorders, Ajou University School of Medicine, Suwon, Korea
- Department of Marine Biomedical Sciences, Hanseo University, Seosan, Chungcheongnam-do, Korea
| | - Jae-Bong Kim
- Department of Pharmacology, Ajou University School of Medicine, 164, Worldcup-ro, Yeongtong-gu, Suwon, 16499, Korea
- Center for Convergence Research of Neurological Disorders, Ajou University School of Medicine, Suwon, Korea
- Department of Biomedical Sciences, Ajou University School of Medicine, Suwon, Korea
| | - Sang Myun Park
- Department of Pharmacology, Ajou University School of Medicine, 164, Worldcup-ro, Yeongtong-gu, Suwon, 16499, Korea.
- Center for Convergence Research of Neurological Disorders, Ajou University School of Medicine, Suwon, Korea.
- Department of Biomedical Sciences, Ajou University School of Medicine, Suwon, Korea.
| |
Collapse
|
18
|
Camblor-Perujo S, Kononenko NL. Brain-specific functions of the endocytic machinery. FEBS J 2021; 289:2219-2246. [PMID: 33896112 DOI: 10.1111/febs.15897] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 03/29/2021] [Indexed: 12/12/2022]
Abstract
Endocytosis is an essential cellular process required for multiple physiological functions, including communication with the extracellular environment, nutrient uptake, and signaling by the cell surface receptors. In a broad sense, endocytosis is accomplished through either constitutive or ligand-induced invagination of the plasma membrane, which results in the formation of the plasma membrane-retrieved endocytic vesicles, which can either be sent for degradation to the lysosomes or recycled back to the PM. This additional function of endocytosis in membrane retrieval has been adopted by excitable cells, such as neurons, for membrane equilibrium maintenance at synapses. The last two decades were especially productive with respect to the identification of brain-specific functions of the endocytic machinery, which additionally include but not limited to regulation of neuronal differentiation and migration, maintenance of neuron morphology and synaptic plasticity, and prevention of neurotoxic aggregates spreading. In this review, we highlight the current knowledge of brain-specific functions of endocytic machinery with a specific focus on three brain cell types, neuronal progenitor cells, neurons, and glial cells.
Collapse
Affiliation(s)
| | - Natalia L Kononenko
- CECAD Cluster of Excellence, University of Cologne, Germany.,Center for Physiology & Pathophysiology, Medical Faculty, University of Cologne, Germany
| |
Collapse
|
19
|
Quigley JA, Logsdon MK, Turner CA, Gonzalez IL, Leonardo NB, Becker JB. Sex differences in vulnerability to addiction. Neuropharmacology 2021; 187:108491. [PMID: 33567305 PMCID: PMC7979496 DOI: 10.1016/j.neuropharm.2021.108491] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Revised: 01/27/2021] [Accepted: 02/04/2021] [Indexed: 02/07/2023]
Abstract
This article reviews evidence for sex differences in vulnerability to addiction with an emphasis on the neural mechanisms underlying these differences. Sex differences in the way that the gonadal hormone, estradiol, interacts with the ascending telencephalic dopamine system results in sex differences in motivated behaviors, including drug-seeking. In rodents, repeated psychostimulant exposure enhances incentive sensitization to a greater extent in females than males. Estradiol increases females' motivation to attain psychostimulants and enhances the value of drug related cues, which ultimately increases their susceptibility towards spontaneous relapse. This, along with females' dampened ability to alter decisions regarding risky behaviors, enhances their vulnerability for escalation of drug use. In males, recent evidence suggests that estradiol may be protective against susceptibility towards drug-preference. Sex differences in the actions of estradiol are reviewed to provide a foundation for understanding how future research might enhance understanding of the mechanisms of sex differences in addiction-related behaviors, which are dependent on estradiol receptor (ER) subtype and the region of the brain they are acting in. A comprehensive review of the distribution of ERα, ERβ, and GPER1 throughout the rodent brain are provided along with a discussion of the possible ways in which these patterns differentially regulate drug-taking between the sexes. The article concludes with a brief discussion of the actions of gonadal hormones on the circuitry of the stress system, including the hypothalamic pituitary adrenal axis and regulation of corticotropin-releasing factor. Sex differences in the stress system can also contribute to females' enhanced vulnerability towards addiction.
Collapse
Affiliation(s)
- Jacqueline A Quigley
- Psychology Department, Ann Arbor MI, 48109 USA; Michigan Neuroscience Institute, University of Michigan, Ann Arbor MI, 48109 USA
| | - Molly K Logsdon
- Michigan Neuroscience Institute, University of Michigan, Ann Arbor MI, 48109 USA
| | - Christopher A Turner
- Psychology Department, Ann Arbor MI, 48109 USA; Michigan Neuroscience Institute, University of Michigan, Ann Arbor MI, 48109 USA
| | - Ivette L Gonzalez
- Psychology Department, Ann Arbor MI, 48109 USA; Michigan Neuroscience Institute, University of Michigan, Ann Arbor MI, 48109 USA
| | - N B Leonardo
- Michigan Neuroscience Institute, University of Michigan, Ann Arbor MI, 48109 USA
| | - Jill B Becker
- Psychology Department, Ann Arbor MI, 48109 USA; Michigan Neuroscience Institute, University of Michigan, Ann Arbor MI, 48109 USA.
| |
Collapse
|
20
|
Zheng S, Wu L, Fan C, Lin J, Zhang Y, Simoncini T, Fu X. The role of Gα protein signaling in the membrane estrogen receptor-mediated signaling. Gynecol Endocrinol 2021; 37:2-9. [PMID: 33412963 DOI: 10.1080/09513590.2020.1851674] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Estrogens exert rapid, extranuclear effects by their action on the plasma membrane estrogen receptors (mERs). Gα protein associated with the cell membrane is involved in many important processes regulated by estrogens. However, the Gα's role in the mER-mediated signaling and the signaling pathways involved are poorly understood. This review aims to outline the Gα's role in the mER-mediated signaling. Immunoblotting, immunofluorescence, co-immunoprecipitation, and RNA interference were carried out using vascular endothelial cells (ECs) and human breast carcinoma cell lines as experimental models. Electrophysiology and immunocytochemistry were carried out using guinea pigs as animal models. Recent advances suggest that the signaling of mERα through Gα is required for vascular EC migration or endothelial H2S release, while Gα13 is involved in estrogen-induced breast cancer cell invasion. Besides, the Gαq-coupled PLC-PKC-PKA pathway is critical for the neural regulation of energy homeostasis. This review summarizes the contributions of Gα to mER-mediated signaling, including cardiovascular protection, breast cancer metastasis, neural regulation of homeostatic functions, and osteogenesis.
Collapse
Affiliation(s)
- Shuhui Zheng
- Research Center for Translational Medicine, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Lin Wu
- Department of Cardiology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Chao Fan
- Department of Gynecology and Obstetrics, The Sixth Affiliated Hospital, Key Laboratory of Cardiovascular Diseases, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
- Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Disease, the Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, P.R. China
| | - Jingxia Lin
- Department of Blood Transfusion, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Yaxing Zhang
- Department of Traditional Chinese Medicine, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Tommaso Simoncini
- Molecular and Cellular Gynecological Endocrinology Laboratory (MCGEL), Department of Reproductive Medicine and Child Development, University of Pisa, Pisa, Italy
| | - Xiaodong Fu
- Department of Gynecology and Obstetrics, The Sixth Affiliated Hospital, Key Laboratory of Cardiovascular Diseases, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
- Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Disease, the Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, P.R. China
| |
Collapse
|
21
|
Alsaab HO, Altowairqi E, Alzahrani N, Alzahrani R, Alshehri FS, Almalki AH, Alsanie WF, Gaber A, Alkhalifa T, Almalki A, Shah ZA, Althobaiti YS. Sex differences in pregabalin-seeking like behavior in a conditioned place preference paradigm. Saudi Pharm J 2020; 28:1749-1755. [PMID: 33424265 PMCID: PMC7783229 DOI: 10.1016/j.jsps.2020.11.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2020] [Accepted: 11/03/2020] [Indexed: 12/30/2022] Open
Abstract
Substance abuse is a chronic, relapsing disorder characterized by compulsive drug use regardless of negative consequences. Incremental increases in pregabalin abuse have been observed in Saudi Arabia and throughout the world. In previous studies, the potential for pregabalin abuse with escalating doses of the drug (30, 60, 90, and 120 mg/kg) were investigated in male mice. Notably, researchers have argued that women may exhibit a greater tendency to consume drugs without a prescription to alleviate stress and depression. Moreover, female subjects are more prone to impulsivity in drug intake or abuse than their male counterparts. Therefore, in the present study, we compared the potential for pregabalin abuse between male and female mice using a conditioned place preference paradigm. Male and female BALB/c mice were divided into four groups based on the pregabalin dose administered (30, 60, 90, or 120 mg/kg, intraperitoneal). Preference scores were then calculated and compared between male and female mice in each dosage group. Interestingly, preference scores were significantly higher in female mice than in male mice at dosages of 30 and 120 mg/kg. These findings indicate that female mice may be more prone to pregabalin abuse and tolerance than male mice. These results might be helpful to the healthcare providers and policymakers to consider these sex differences in choosing therapeutic plans and consider alternatives to the misused prescription medications.
Collapse
Affiliation(s)
- Hashem O. Alsaab
- Addiction and Neuroscience Research Unit, Taif University, Health Science Campus, Al Haweiah, Taif 21974, Saudi Arabia
- Department of Pharmaceutics and Pharmaceutical Technology, College of Pharmacy, Taif University, Health Science Campus, Al Haweiah, Taif 21974, Saudi Arabia
| | - Ebtehal Altowairqi
- Department of Pharmacology and Toxicology, College of Pharmacy, Taif University, Health Science Campus, Al Haweiah, Taif 21974, Saudi Arabia
| | - Nada Alzahrani
- Addiction and Neuroscience Research Unit, Taif University, Health Science Campus, Al Haweiah, Taif 21974, Saudi Arabia
| | - Reem Alzahrani
- Addiction and Neuroscience Research Unit, Taif University, Health Science Campus, Al Haweiah, Taif 21974, Saudi Arabia
| | - Fahad S. Alshehri
- Department of Pharmacology and Toxicology, College of Pharmacy, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Atiah H. Almalki
- Addiction and Neuroscience Research Unit, Taif University, Health Science Campus, Al Haweiah, Taif 21974, Saudi Arabia
- Department of Pharmaceutical Chemistry, College of Pharmacy, Taif University, Health Science Campus, Al Haweiah, Taif 21974, Saudi Arabia
| | - Walaa F. Alsanie
- Addiction and Neuroscience Research Unit, Taif University, Health Science Campus, Al Haweiah, Taif 21974, Saudi Arabia
- Faculty of Applied Medical Sciences, Department of Clinical Laboratories, Taif University, Taif 21974, Saudi Arabia
| | - Ahmed Gaber
- Addiction and Neuroscience Research Unit, Taif University, Health Science Campus, Al Haweiah, Taif 21974, Saudi Arabia
- Department of Biology, Faculty of Science, Taif University, Taif 21974, Saudi Arabia
| | - Turki Alkhalifa
- General Administration for Precursors and Laboratories, General Directorate of Narcotics Control, Ministry of Interior, Riyadh, Saudi Arabia
| | - Ahmad Almalki
- General Administration for Precursors and Laboratories, General Directorate of Narcotics Control, Ministry of Interior, Riyadh, Saudi Arabia
| | - Zahoor A. Shah
- Department of Medicinal and Biological Chemistry, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, Toledo, OH 43614, USA
| | - Yusuf S. Althobaiti
- Addiction and Neuroscience Research Unit, Taif University, Health Science Campus, Al Haweiah, Taif 21974, Saudi Arabia
- Department of Pharmacology and Toxicology, College of Pharmacy, Taif University, Health Science Campus, Al Haweiah, Taif 21974, Saudi Arabia
- General Administration for Precursors and Laboratories, General Directorate of Narcotics Control, Ministry of Interior, Riyadh, Saudi Arabia
| |
Collapse
|
22
|
Taxier LR, Gross KS, Frick KM. Oestradiol as a neuromodulator of learning and memory. Nat Rev Neurosci 2020; 21:535-550. [PMID: 32879508 PMCID: PMC8302223 DOI: 10.1038/s41583-020-0362-7] [Citation(s) in RCA: 123] [Impact Index Per Article: 24.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/31/2020] [Indexed: 12/24/2022]
Abstract
Although hormones such as glucocorticoids have been broadly accepted in recent decades as general neuromodulators of memory processes, sex steroid hormones such as the potent oestrogen 17β-oestradiol have been less well recognized by the scientific community in this capacity. The predominance of females in studies of oestradiol and memory and the general (but erroneous) perception that oestrogens are 'female' hormones have probably prevented oestradiol from being more widely considered as a key memory modulator in both sexes. Indeed, although considerable evidence supports a crucial role for oestradiol in regulating learning and memory in females, a growing body of literature indicates a similar role in males. This Review discusses the mechanisms of oestradiol signalling and provides an overview of the effects of oestradiol on spatial, object recognition, social and fear memories. Although the primary focus is on data collected in females, effects of oestradiol on memory in males will be discussed, as will sex differences in the molecular mechanisms that regulate oestrogenic modulation of memory, which may have important implications for the development of future cognitive therapeutics.
Collapse
Affiliation(s)
- Lisa R Taxier
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, WI, USA
| | - Kellie S Gross
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, WI, USA
| | - Karyn M Frick
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, WI, USA.
| |
Collapse
|
23
|
Caveolin-1 regulates medium spiny neuron structural and functional plasticity. Psychopharmacology (Berl) 2020; 237:2673-2684. [PMID: 32488350 PMCID: PMC7502476 DOI: 10.1007/s00213-020-05564-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Accepted: 05/18/2020] [Indexed: 12/14/2022]
Abstract
RATIONALE Caveolin-1 (CAV1) is a structural protein critical for spatial organization of neuronal signaling molecules. Whether CAV1 is required for long-lasting neuronal plasticity remains unknown. OBJECTIVE AND METHODS We sought to examine the effects of CAV1 knockout (KO) on functional plasticity and hypothesized that CAV1 deficiency would impact drug-induced long-term plasticity in the nucleus accumbens (NAc). We first examined cell morphology of NAc medium spiny neurons in a striatal/cortical co-culture system before moving in vivo to study effects of CAV1 KO on cocaine-induced plasticity. Whole-cell patch-clamp recordings were performed to determine effects of chronic cocaine (15 mg/kg) on medium spiny neuron excitability. To test for deficits in behavioral plasticity, we examined the effect of CAV1 KO on locomotor sensitization. RESULTS Disruption of CAV1 expression leads to baseline differences in medium spiny neuron (MSN) structural morphology, such that MSNs derived from CAV1 KO animals have increased dendritic arborization when cultured with cortical neurons. The effect was dependent on phospholipase C and cell-type intrinsic loss of CAV1. Slice recordings of nucleus accumbens shell MSNs revealed that CAV1 deficiency produces a loss of neuronal plasticity. Specifically, cocaine-induced firing rate depression was absent in CAV1 KO animals, whereas baseline electrophysiological properties were similar. This was reflected by a loss of cocaine-mediated behavioral sensitization in CAV1 KO animals, with unaffected baseline locomotor responsiveness. CONCLUSIONS This study highlights a critical role for nucleus accumbens CAV1 in plasticity related to the administration of drugs of abuse.
Collapse
|
24
|
Kotova A, Timonina K, Zoidl GR. Endocytosis of Connexin 36 is Mediated by Interaction with Caveolin-1. Int J Mol Sci 2020; 21:E5401. [PMID: 32751343 PMCID: PMC7432810 DOI: 10.3390/ijms21155401] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2020] [Revised: 07/25/2020] [Accepted: 07/28/2020] [Indexed: 12/22/2022] Open
Abstract
The gap junctional protein connexin 36 (Cx36) has been co-purified with the lipid raft protein caveolin-1 (Cav-1). The relevance of an interaction between the two proteins is unknown. In this study, we explored the significance of Cav-1 interaction in the context of intracellular and membrane transport of Cx36. Coimmunoprecipitation assays and Förster resonance energy transfer analysis (FRET) were used to confirm the interaction between the two proteins in the Neuro 2a cell line. We found that the Cx36 and Cav-1 interaction was dependent on the intracellular calcium levels. By employing different microscopy techniques, we demonstrated that Cav-1 enhances the vesicular transport of Cx36. Pharmacological interventions coupled with cell surface biotinylation assays and FRET analysis revealed that Cav-1 regulates membrane localization of Cx36. Our data indicate that the interaction between Cx36 and Cav-1 plays a role in the internalization of Cx36 by a caveolin-dependent pathway.
Collapse
Affiliation(s)
- Anna Kotova
- Department of Biology, York University, Toronto, ON M3J 1P3, Canada; (A.K.); (K.T.)
| | - Ksenia Timonina
- Department of Biology, York University, Toronto, ON M3J 1P3, Canada; (A.K.); (K.T.)
| | - Georg R. Zoidl
- Department of Biology, York University, Toronto, ON M3J 1P3, Canada; (A.K.); (K.T.)
- Department of Psychology, York University, Toronto, ON M3J 1P3, Canada
| |
Collapse
|
25
|
Gross KS, Mermelstein PG. Estrogen receptor signaling through metabotropic glutamate receptors. VITAMINS AND HORMONES 2020; 114:211-232. [PMID: 32723544 DOI: 10.1016/bs.vh.2020.06.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
As the non-nuclear initiated effects of steroid hormone signaling have become more widely accepted, there has been a need to define the novel mechanisms of hormone receptor action that account for these outcomes. One mechanism that has emerged is the coupling of classical estrogen receptors (ERα and ERβ) with metabotropic glutamate receptors (mGluRs) to initiate G protein signaling cascades that ultimately influence neuronal physiology, structure, and behavior. Since its initial discovery in hippocampal neurons, evidence of ER/mGluR associations have been found throughout the nervous system, and the heterogeneity of possible receptor pairings afforded by multiple ER and mGluR subtypes appears to drive diverse molecular outcomes that can impact processes like cognition, motivation, movement, and pain. Recent evidence also suggests that the role of mGluRs in steroid hormone signaling may not be unique to ERs, but rather a conserved mechanism of membrane-initiated hormone receptor action.
Collapse
Affiliation(s)
- Kellie S Gross
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, WI, United States
| | - Paul G Mermelstein
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, United States.
| |
Collapse
|
26
|
Vandegrift BJ, Hilderbrand ER, Satta R, Tai R, He D, You C, Chen H, Xu P, Coles C, Brodie MS, Lasek AW. Estrogen Receptor α Regulates Ethanol Excitation of Ventral Tegmental Area Neurons and Binge Drinking in Female Mice. J Neurosci 2020; 40:5196-5207. [PMID: 32482639 PMCID: PMC7329299 DOI: 10.1523/jneurosci.2364-19.2020] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Revised: 03/25/2020] [Accepted: 04/22/2020] [Indexed: 12/16/2022] Open
Abstract
Elevations in estrogen (17β-estradiol, E2) are associated with increased alcohol drinking by women and experimentally in rodents. E2 alters the activity of the dopamine system, including the VTA and its projection targets, which plays an important role in binge drinking. A previous study demonstrated that, during high E2 states, VTA neurons in female mice are more sensitive to ethanol excitation. However, the mechanisms responsible for the ability of E2 to enhance ethanol sensitivity of VTA neurons have not been investigated. In this study, we used selective agonists and antagonists to examine the role of ER subtypes (ERα and ERβ) in regulating the ethanol sensitivity of VTA neurons in female mice and found that ERα promotes the enhanced ethanol response of VTA neurons. We also demonstrated that enhancement of ethanol excitation requires the activity of the metabotropic glutamate receptor, mGluR1, which is known to couple with ERα at the plasma membrane. To investigate the behavioral relevance of these findings, we administered lentivirus-expressing short hairpin RNAs targeting either ERα or ERβ into the VTA and found that knockdown of each receptor in the VTA reduced binge-like ethanol drinking in female, but not male, mice. Reducing ERα in the VTA had a more dramatic effect on binge-like drinking than reducing ERβ, consistent with the ability of ERα to alter ethanol sensitivity of VTA neurons. These results provide important insight into sex-specific mechanisms that drive excessive alcohol drinking.SIGNIFICANCE STATEMENT Estrogen has potent effects on the dopamine system and increases the vulnerability of females to develop addiction to substances, such as alcohol. We investigated the mechanisms by which estrogen increases the response of neurons in the VTA to ethanol. We found that activation of the ERα increased the ethanol-induced excitation of VTA neurons. 17β-Estradiol-mediated enhancement of ethanol-induced excitation required the metabotropic glutamate receptor mGluR1. We also demonstrated that ERs in the VTA regulate binge-like alcohol drinking by female, but not male, mice. The influence of ERs on binge drinking in female mice suggests that treatments for alcohol use disorder in women may need to account for this sex difference.
Collapse
Affiliation(s)
- Bertha J Vandegrift
- Center for Alcohol Research in Epigenetics and Department of Psychiatry
- Department of Physiology and Biophysics
| | | | - Rosalba Satta
- Center for Alcohol Research in Epigenetics and Department of Psychiatry
| | - Rex Tai
- Center for Alcohol Research in Epigenetics and Department of Psychiatry
| | - Donghong He
- Center for Alcohol Research in Epigenetics and Department of Psychiatry
| | - Chang You
- Department of Physiology and Biophysics
| | - Hu Chen
- Center for Alcohol Research in Epigenetics and Department of Psychiatry
| | - Pingwen Xu
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, University of Illinois at Chicago, Chicago, Illinois 60612
| | - Cassandre Coles
- Center for Alcohol Research in Epigenetics and Department of Psychiatry
| | - Mark S Brodie
- Center for Alcohol Research in Epigenetics and Department of Psychiatry
- Department of Physiology and Biophysics
| | - Amy W Lasek
- Center for Alcohol Research in Epigenetics and Department of Psychiatry
| |
Collapse
|
27
|
Kokane SS, Perrotti LI. Sex Differences and the Role of Estradiol in Mesolimbic Reward Circuits and Vulnerability to Cocaine and Opiate Addiction. Front Behav Neurosci 2020; 14:74. [PMID: 32508605 PMCID: PMC7251038 DOI: 10.3389/fnbeh.2020.00074] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Accepted: 04/22/2020] [Indexed: 12/15/2022] Open
Abstract
Although both men and women become addicted to drugs of abuse, women transition to addiction faster, experience greater difficulties remaining abstinent, and relapse more often than men. In both humans and rodents, hormonal cycles are associated with females' faster progression to addiction. Higher concentrations and fluctuating levels of ovarian hormones in females modulate the mesolimbic reward system and influence reward-directed behavior. For example, in female rodents, estradiol (E2) influences dopamine activity within the mesolimbic reward system such that drug-directed behaviors that are normally rewarding and reinforcing become enhanced when circulating levels of E2 are high. Therefore, neuroendocrine interactions, in part, explain sex differences in behaviors motivated by drug reward. Here, we review sex differences in the physiology and function of the mesolimbic reward system in order to explore the notion that sex differences in response to drugs of abuse, specifically cocaine and opiates, are the result of molecular neuroadaptations that differentially develop depending upon the hormonal state of the animal. We also reconsider the notion that ovarian hormones, specifically estrogen/estradiol, sensitize target neurons thereby increasing responsivity when under the influence of either cocaine or opiates or in response to exposure to drug-associated cues. These adaptations may ultimately serve to guide the motivational behaviors that underlie the factors that cause women to be more vulnerable to cocaine and opiate addiction than men.
Collapse
Affiliation(s)
- Saurabh S Kokane
- Department of Psychology, University of Texas at Arlington, Arlington, TX, United States
| | - Linda I Perrotti
- Department of Psychology, University of Texas at Arlington, Arlington, TX, United States
| |
Collapse
|
28
|
Pol A, Morales-Paytuví F, Bosch M, Parton RG. Non-caveolar caveolins – duties outside the caves. J Cell Sci 2020; 133:133/9/jcs241562. [DOI: 10.1242/jcs.241562] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
ABSTRACT
Caveolae are invaginations of the plasma membrane that are remarkably abundant in adipocytes, endothelial cells and muscle. Caveolae provide cells with resources for mechanoprotection, can undergo fission from the plasma membrane and can regulate a variety of signaling pathways. Caveolins are fundamental components of caveolae, but many cells, such as hepatocytes and many neurons, express caveolins without forming distinguishable caveolae. Thus, the function of caveolins goes beyond their roles as caveolar components. The membrane-organizing and -sculpting capacities of caveolins, in combination with their complex intracellular trafficking, might contribute to these additional roles. Furthermore, non-caveolar caveolins can potentially interact with proteins normally excluded from caveolae. Here, we revisit the non-canonical roles of caveolins in a variety of cellular contexts including liver, brain, lymphocytes, cilia and cancer cells, as well as consider insights from invertebrate systems. Non-caveolar caveolins can determine the intracellular fluxes of active lipids, including cholesterol and sphingolipids. Accordingly, caveolins directly or remotely control a plethora of lipid-dependent processes such as the endocytosis of specific cargoes, sorting and transport in endocytic compartments, or different signaling pathways. Indeed, loss-of-function of non-caveolar caveolins might contribute to the common phenotypes and pathologies of caveolin-deficient cells and animals.
Collapse
Affiliation(s)
- Albert Pol
- Cell Compartments and Signaling Group, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, 08036, Barcelona, Spain
- Department of Biomedical Sciences, Faculty of Medicine, Universitat de Barcelona, 08036, Barcelona, Spain
- Institució Catalana de Recerca i Estudis Avançats (ICREA), 08010, Barcelona, Spain
| | - Frederic Morales-Paytuví
- Cell Compartments and Signaling Group, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, 08036, Barcelona, Spain
| | - Marta Bosch
- Cell Compartments and Signaling Group, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, 08036, Barcelona, Spain
- Department of Biomedical Sciences, Faculty of Medicine, Universitat de Barcelona, 08036, Barcelona, Spain
| | - Robert G. Parton
- Institute for Molecular Bioscience (IMB), The University of Queensland (UQ), Brisbane, Queensland 4072, Australia
- Centre for Microscopy and Microanalysis (CMM) IMB, The University of Queensland (UQ), Brisbane, Queensland 4072, Australia
| |
Collapse
|
29
|
Wong AM, Scott AK, Johnson CS, Mohr MA, Mittelman-Smith M, Micevych PE. ERαΔ4, an ERα splice variant missing exon4, interacts with caveolin-3 and mGluR2/3. J Neuroendocrinol 2019; 31:e12725. [PMID: 31050077 PMCID: PMC6591055 DOI: 10.1111/jne.12725] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Revised: 04/11/2019] [Accepted: 04/26/2019] [Indexed: 12/11/2022]
Abstract
The two isoforms of the nuclear estrogen receptor, ERα and ERβ are widely expressed in the central nervous system. Although they were first described as nuclear receptors, both isoforms have also been found at the cell membrane where they mediate cell signaling. Surface biotinylation studies using neuronal and glial primary cultures label an alternatively spliced form of ERα. The 52 kDa protein, ERαΔ4, is missing exon 4 and is highly expressed in membrane fractions derived from cultured cells. In vivo, both full-length (66 kDa) ERα and ERαΔ4 are present in membrane fractions. In response to estradiol, full-length ERα and ERαΔ4 are initially trafficked to the membrane, and then internalized in parallel. Previous studies determined that only the full-length ERα associates with metabotropic glutamate receptor-1a (mGluR1a), initiating cellular signaling. The role of ERαΔ4, remained to be elucidated. Here, we report ERαΔ4 trafficking, association with mGluR2/3, and downstream signaling in female rat arcuate nucleus (ARH). Caveolin (CAV) proteins are needed for ER transport to the cell membrane, and using co-immunoprecipitation CAV-3 was shown to associate with ERαΔ4. CAV-3 was necessary for ERαΔ4 trafficking to the membrane: in the ARH, microinjection of CAV-3 siRNA reduced CAV-3 and ERαΔ4a in membrane fractions by 50%, and 60%, respectively. Moreover, co-immunoprecipitation revealed that ERαΔ4 associated with inhibitory mGluRs, mGluR2/3. Estrogen benzoate (EB) treatment (5 μg; s.c.; every 4 days; three cycles) reduced levels of cAMP, an effect attenuated by antagonizing mGluR2/3. Following EB treatment, membrane levels of ERαΔ4 and mGluR2/3 were reduced implying ligand-induced internalization. These results implicate ERαΔ4 in an estradiol-induced inhibitory cell signaling in the ARH.
Collapse
Affiliation(s)
- Angela M Wong
- Department of Neurobiology, David Geffen School of Medicine at UCLA, Los Angeles, California
- Laboratory of Neuroendocrinology of the Brain Research Institute, University of California, Los Angeles, California
| | - Alexandra K Scott
- Department of Neurobiology, David Geffen School of Medicine at UCLA, Los Angeles, California
- Laboratory of Neuroendocrinology of the Brain Research Institute, University of California, Los Angeles, California
| | - Caroline S Johnson
- Department of Neurobiology, David Geffen School of Medicine at UCLA, Los Angeles, California
- Laboratory of Neuroendocrinology of the Brain Research Institute, University of California, Los Angeles, California
| | - Margaret A Mohr
- Department of Neurobiology, David Geffen School of Medicine at UCLA, Los Angeles, California
- Laboratory of Neuroendocrinology of the Brain Research Institute, University of California, Los Angeles, California
| | - Melinda Mittelman-Smith
- Department of Neurobiology, David Geffen School of Medicine at UCLA, Los Angeles, California
- Laboratory of Neuroendocrinology of the Brain Research Institute, University of California, Los Angeles, California
| | - Paul E Micevych
- Department of Neurobiology, David Geffen School of Medicine at UCLA, Los Angeles, California
- Laboratory of Neuroendocrinology of the Brain Research Institute, University of California, Los Angeles, California
| |
Collapse
|
30
|
Meitzen J, Britson KA, Tuomela K, Mermelstein PG. The expression of select genes necessary for membrane-associated estrogen receptor signaling differ by sex in adult rat hippocampus. Steroids 2019; 142:21-27. [PMID: 28962849 PMCID: PMC5874170 DOI: 10.1016/j.steroids.2017.09.012] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Revised: 09/14/2017] [Accepted: 09/22/2017] [Indexed: 12/21/2022]
Abstract
17β-estradiol can rapidly modulate neuron function via membrane estrogen receptors (ERs) in a sex-specific manner. For example, female rat hippocampal neurons express palmitoylated versions of ERα and ERβ that associate with the plasma membrane. These membrane-associated ERs are organized by caveolin proteins into functional signaling microdomains with metabotropic glutamate receptors (mGluRs). ER/mGluR signaling mediates several sex-specific estradiol actions on hippocampal neuron function. An important unanswered question regards the mechanism by which sex-specific membrane-associated ER signaling is generated, especially since it has been previously demonstrated that mGluR action is not sex-specific. One possibility is that the genes necessary for the ER membrane complex are differentially expressed between males and females, including genes that encode ERα and β, caveolin 1 and 3, and/or the palmitoylacyltransferases DHHC-7 and -21. Thus we used qPCR to test the hypothesis that these genes show sex differences in expression in neonatal and adult rat hippocampus. As an additional control we tested the expression of the 20 other DHHC palmitoylacyltransferases with no known connections to ER. In neonatal hippocampus, no sex differences were detected in gene expression. In adult hippocampus, the genes that encode caveolin 1 and DHHC-7 showed decreased expression in females compared to males. Thus, select genes differ by sex at specific developmental stages, arguing for a more nuanced model than simple widespread perinatal emergence of sex differences in all genes enabling sex-specific estradiol action. These findings enable the generation of new hypotheses regarding the mechanisms by which sex differences in membrane-associated ER signaling are programmed.
Collapse
Affiliation(s)
- John Meitzen
- Dept. of Biological Sciences, North Carolina State University, Raleigh, NC, United States; W.M. Keck Center for Behavioral Biology, North Carolina State University, Raleigh, NC, United States; Center for Human Health and the Environment, North Carolina State University, Raleigh, NC, United States; Comparative Medicine Institute, North Carolina State University, Raleigh, NC, United States.
| | - Kyla A Britson
- Cellular and Molecular Medicine Graduate Program, Johns Hopkins School of Medicine, Baltimore, MD, United States
| | - Krista Tuomela
- Medical College of Wisconsin, Milwaukee, WI, United States
| | - Paul G Mermelstein
- Dept. of Neuroscience, University of Minnesota, Minneapolis, MN, United States
| |
Collapse
|
31
|
Becker JB, Chartoff E. Sex differences in neural mechanisms mediating reward and addiction. Neuropsychopharmacology 2019; 44:166-183. [PMID: 29946108 PMCID: PMC6235836 DOI: 10.1038/s41386-018-0125-6] [Citation(s) in RCA: 298] [Impact Index Per Article: 49.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Revised: 04/27/2018] [Accepted: 06/11/2018] [Indexed: 12/18/2022]
Abstract
There is increasing evidence in humans and laboratory animals for biologically based sex differences in every phase of drug addiction: acute reinforcing effects, transition from occasional to compulsive use, withdrawal-associated negative affective states, craving, and relapse. There is also evidence that many qualitative aspects of the addiction phases do not differ significantly between males and females, but one sex may be more likely to exhibit a trait than the other, resulting in population differences. The conceptual framework of this review is to focus on hormonal, chromosomal, and epigenetic organizational and contingent, sex-dependent mechanisms of four neural systems that are known-primarily in males-to be key players in addiction: dopamine, mu-opioid receptors (MOR), kappa opioid receptors (KOR), and brain-derived neurotrophic factor (BDNF). We highlight data demonstrating sex differences in development, expression, and function of these neural systems as they relate-directly or indirectly-to processes of reward and addictive behavior, with a focus on psychostimulants and opioids. We identify gaps in knowledge about how these neural systems interact with sex to influence addictive behavior, emphasizing throughout that the impact of sex can be highly nuanced and male/female data should be reported regardless of the outcome.
Collapse
Affiliation(s)
- Jill B Becker
- Department of Psychology and the Molecular & Behavioral Neuroscience Institute, University of Michigan, Ann Arbor, MI, USA
| | - Elena Chartoff
- Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA, USA.
| |
Collapse
|
32
|
Zafer D, Aycan N, Ozaydin B, Kemanli P, Ferrazzano P, Levine JE, Cengiz P. Sex differences in Hippocampal Memory and Learning following Neonatal Brain Injury: Is There a Role for Estrogen Receptor-α? Neuroendocrinology 2019; 109:249-256. [PMID: 30884486 PMCID: PMC6893032 DOI: 10.1159/000499661] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2018] [Accepted: 03/17/2019] [Indexed: 01/11/2023]
Abstract
Neonatal encephalopathy due to hypoxia-ischemia (HI) leads to severe, life-long morbidities in thousands of neonates born in the USA and worldwide each year. Varying capacities of long-term episodic memory, verbal working memory, and learning can present without cerebral palsy and have been associated with the severity of neonatal encephalopathy sustained at birth. Among children who sustain a moderate degree of HI at birth, girls have larger hippocampal volumes compared to boys. Clinical studies indicate that female neonatal brains are more resistant to the effects of neonatal HI, resulting in better long-term cognitive outcomes compared to males with comparable brain injury. Our most recent mechanistic studies have addressed the origins and cellular basis of sex differences in hippocampal neuroprotection following neonatal HI-related brain injury and implicate estrogen receptor-α (ERα) in the neurotrophin receptor-mediated hippocampal neuroprotection in female mice. This review summarizes the recent findings on ERα-dependent, neurotrophin-mediated hippocampal neuroprotection and weighs the evidence that this mechanism plays an important role in preservation of long-term memory and learning following HI in females.
Collapse
Affiliation(s)
- Dila Zafer
- Waisman Center, University of Wisconsin, Madison, Wisconsin, USA
| | - Nur Aycan
- Waisman Center, University of Wisconsin, Madison, Wisconsin, USA
| | - Burak Ozaydin
- Waisman Center, University of Wisconsin, Madison, Wisconsin, USA
| | - Pinar Kemanli
- Waisman Center, University of Wisconsin, Madison, Wisconsin, USA
| | - Peter Ferrazzano
- Waisman Center, University of Wisconsin, Madison, Wisconsin, USA
- Division of Pediatric Critical Care Medicine, Department of Pediatrics, University of Wisconsin, Madison, Wisconsin, USA
| | - Jon E Levine
- Department of Neuroscience, University of Wisconsin, Madison, Wisconsin, USA
- Wisconsin National Primate Research Center, Madison, Wisconsin, USA
| | - Pelin Cengiz
- Waisman Center, University of Wisconsin, Madison, Wisconsin, USA,
- Division of Pediatric Critical Care Medicine, Department of Pediatrics, University of Wisconsin, Madison, Wisconsin, USA,
| |
Collapse
|
33
|
Santollo J, Daniels D. Anorexigenic effects of estradiol in the medial preoptic area occur through membrane-associated estrogen receptors and metabotropic glutamate receptors. Horm Behav 2019; 107:20-25. [PMID: 30462987 PMCID: PMC6348004 DOI: 10.1016/j.yhbeh.2018.11.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Revised: 11/01/2018] [Accepted: 11/03/2018] [Indexed: 11/22/2022]
Abstract
Activation of membrane-associated estrogen receptors (mER) decreases food and water intake in female rats. Additional studies suggest these effects are mediated, at least in part, by membrane-associated estrogen receptor alpha (ERα). Nevertheless, the critical site of action and the intracellular signaling required for the ingestive effects of ERα remain unclear. Estradiol given to the medial preoptic area (mPOA) decreases ingestive behaviors, and membrane-associated ERα has been shown to affect intracellular signaling through interactions with metabotropic glutamate receptor (mGluR) subtypes, but an involvement of this signaling pathway, in the mPOA, in ingestive behavior remains untested. To address these open questions, we first showed that activation of mER in the mPOA decreased both overnight food and water intake, and did so in a time course consistent with a genomic mechanism of action. Next, we tested the requirement of mGluR1a signaling in the mPOA for the anorexigenic and anti-dipsogenic effects of estradiol. As expected, estradiol in the mPOA decreased food intake, but only in the absence of an mGluR1a antagonist. The same was not true for estradiol effects on water intake, which were unaffected by an mGluR1a antagonist. These results suggest that estrogens require mGluR activation for at least some of their effects on ingestive behaviors, and indicate that the mPOA is a critical site of action. The results also reveal an interesting divergence in the estrogenic control of ingestive behavior by which mGluR signaling in the mPOA plays a role in the control of food intake, but not water intake.
Collapse
Affiliation(s)
- Jessica Santollo
- Department of Biology, University of Kentucky, Lexington, KY 40506, USA.
| | - Derek Daniels
- Behavioral Neuroscience Program, Department of Psychology, The State University of New York at Buffalo, Buffalo, New York 14260, USA
| |
Collapse
|
34
|
Gross KS, Moore KM, Meisel RL, Mermelstein PG. mGluR5 Mediates Dihydrotestosterone-Induced Nucleus Accumbens Structural Plasticity, but Not Conditioned Reward. Front Neurosci 2018; 12:855. [PMID: 30515075 PMCID: PMC6255826 DOI: 10.3389/fnins.2018.00855] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Accepted: 11/01/2018] [Indexed: 12/12/2022] Open
Abstract
Gonadal hormones play a vital role in driving motivated behavior. They not only modulate responses to naturally rewarding stimuli, but also influence responses to drugs of abuse. A commonality between gonadal hormones and drugs of abuse is that they both impact the neurocircuitry of reward, including the regulation of structural plasticity in the nucleus accumbens (NAc). Previous hormonal studies have focused on the mechanisms and behavioral correlates of estradiol-induced dendritic spine changes in the female NAc. Here we sought to determine the effects of androgens on medium spiny neuron (MSN) spine plasticity in the male NAc. Following treatment with the androgen receptor agonist dihydrotestosterone (DHT), MSNs in castrated male rats exhibited a significant decrease in dendritic spine density. This effect was isolated to the shell subregion of the NAc. The effect of DHT was dependent on mGluR5 activity, and local mGluR5 activation and subsequent endocannabinoid signaling produce an analogous NAc shell spine decrease. Somewhat surprisingly, DHT-induced conditioned place preference remained intact following systemic inhibition of mGluR5. These findings indicate that androgens can utilize mGluR signaling, similar to estrogens, to mediate changes in NAc dendritic structure. In addition, there are notable differences in the direction of spine changes, and site specificity of estrogen and androgen action, suggesting sex differences in the hormonal regulation of motivated behaviors.
Collapse
Affiliation(s)
- Kellie S Gross
- Graduate Program in Neuroscience, University of Minnesota, Minneapolis, MN, United States.,Department of Neuroscience, University of Minnesota, Minneapolis, MN, United States
| | - Kelsey M Moore
- Graduate Program in Neuroscience, University of Minnesota, Minneapolis, MN, United States.,Department of Neuroscience, University of Minnesota, Minneapolis, MN, United States
| | - Robert L Meisel
- Graduate Program in Neuroscience, University of Minnesota, Minneapolis, MN, United States.,Department of Neuroscience, University of Minnesota, Minneapolis, MN, United States
| | - Paul G Mermelstein
- Graduate Program in Neuroscience, University of Minnesota, Minneapolis, MN, United States.,Department of Neuroscience, University of Minnesota, Minneapolis, MN, United States
| |
Collapse
|
35
|
Ogawa S, Tsukahara S, Choleris E, Vasudevan N. Estrogenic regulation of social behavior and sexually dimorphic brain formation. Neurosci Biobehav Rev 2018; 110:46-59. [PMID: 30392880 DOI: 10.1016/j.neubiorev.2018.10.012] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Revised: 10/17/2018] [Accepted: 10/22/2018] [Indexed: 02/07/2023]
Abstract
It has long been known that the estrogen, 17β-estradiol (17β-E), plays a central role for female reproductive physiology and behavior. Numerous studies have established the neurochemical and molecular basis of estrogenic induction of female sexual behavior, i.e., lordosis, in animal models. In addition, 17β-E also regulates male-type sexual and aggressive behavior. In males, testosterone secreted from the testes is irreversibly aromatized to 17β-E in the brain. We discuss the contribution of two nuclear receptor isoforms, estrogen receptor (ER)α and ERβ to the estrogenic regulation of sexually dimorphic brain formation and sex-typical expression of these social behaviors. Furthermore, 17β-E is a key player for social behaviors such as social investigation, preference, recognition and memory as well as anxiety-related behaviors in social contexts. Recent studies also demonstrated that not only nuclear receptor-mediated genomic signaling but also membrane receptor-mediated non-genomic actions of 17β-E may underlie the regulation of these behaviors. Finally, we will discuss how rapidly developing research tools and ideas allow us to investigate estrogenic action by emphasizing behavioral neural networks.
Collapse
Affiliation(s)
- Sonoko Ogawa
- Laboratory of Behavioral Neuroendocrinology, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, 305-8577, Japan.
| | - Shinji Tsukahara
- Division of Life Science, Graduate School of Science and Engineering, Saitama University, 255 Shimo-Okubo, Sakura-ku, Saitama City, Saitama 338-8570, Japan
| | - Elena Choleris
- Department of Psychology and Neuroscience Program, University of Guelph, Guelph, ON, N1G 2W1, Canada
| | - Nandini Vasudevan
- School of Biological Sciences, University of Reading, WhiteKnights Campus, Reading, RG6 6AS, United Kingdom
| |
Collapse
|
36
|
Tonn Eisinger KR, Woolfrey KM, Swanson SP, Schnell SA, Meitzen J, Dell'Acqua M, Mermelstein PG. Palmitoylation of caveolin-1 is regulated by the same DHHC acyltransferases that modify steroid hormone receptors. J Biol Chem 2018; 293:15901-15911. [PMID: 30158247 PMCID: PMC6187622 DOI: 10.1074/jbc.ra118.004167] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Revised: 08/27/2018] [Indexed: 12/19/2022] Open
Abstract
Palmitoylation is a reversible post-translational addition of a 16-carbon lipid chain involved in trafficking and compartmentalizing target proteins. It is important for many cellular functions, including signaling via membrane-localized estrogen receptors (ERs). Within the nervous system, palmitoylation of ERα is necessary for membrane surface localization and mediation of downstream signaling through the activation of metabotropic glutamate receptors (mGluRs). Substitution of the single palmitoylation site on ERα prevents its physical association with the integral membrane protein caveolin-1 (CAV1), required for the formation of the ER/mGluR signaling complex. Interestingly, siRNA knockdown of either of two palmitoyl acyltransferases, zinc finger DHHC type-containing 7 (DHHC7) or DHHC21, also eliminates this signaling mechanism. Because ERα has only one palmitoylation site, we hypothesized that one of these DHHCs palmitoylates CAV1. We investigated this possibility by using an acyl-biotin exchange assay in HEK293 cells in conjunction with DHHC overexpression and found that DHHC7 increases CAV1 palmitoylation. Substitution of the palmitoylation sites on CAV1 eliminated this effect but did not disrupt the ability of the DHHC enzyme to associate with CAV1. In contrast, siRNA-mediated knockdown of DHHC7 alone was not sufficient to decrease CAV1 palmitoylation but rather required simultaneous knockdown of DHHC21. These findings provide additional information about the overall influence of palmitoylation on the membrane-initiated estrogen signaling pathway and highlight the importance of considering the influence of palmitoylation on other CAV1-dependent processes.
Collapse
Affiliation(s)
- Katherine R Tonn Eisinger
- From the Department of Neuroscience and
- the Graduate Program in Neuroscience, University of Minnesota, Minneapolis, Minnesota 55455
| | - Kevin M Woolfrey
- the Department of Pharmacology, University of Colorado Denver, Aurora, Colorado 80045, and
| | | | | | - John Meitzen
- the Department of Biological Sciences, North Carolina State University, Raleigh, North Carolina 27695
| | - Mark Dell'Acqua
- the Department of Pharmacology, University of Colorado Denver, Aurora, Colorado 80045, and
| | - Paul G Mermelstein
- From the Department of Neuroscience and
- the Graduate Program in Neuroscience, University of Minnesota, Minneapolis, Minnesota 55455
| |
Collapse
|
37
|
Memory-Related Synaptic Plasticity Is Sexually Dimorphic in Rodent Hippocampus. J Neurosci 2018; 38:7935-7951. [PMID: 30209204 DOI: 10.1523/jneurosci.0801-18.2018] [Citation(s) in RCA: 83] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Revised: 06/22/2018] [Accepted: 07/15/2018] [Indexed: 12/22/2022] Open
Abstract
Men are generally superior to women in remembering spatial relationships, whereas the reverse holds for semantic information, but the neurobiological bases for these differences are not understood. Here we describe striking sexual dimorphism in synaptic mechanisms of memory encoding in hippocampal field CA1, a region critical for spatial learning. Studies of acute hippocampal slices from adult rats and mice show that for excitatory Schaffer-commissural projections, the memory-related long-term potentiation (LTP) effect depends upon endogenous estrogen and membrane estrogen receptor α (ERα) in females but not in males; there was no evident involvement of nuclear ERα in females, or of ERβ or GPER1 (G-protein-coupled estrogen receptor 1) in either sex. Quantitative immunofluorescence showed that stimulation-induced activation of two LTP-related kinases (Src, ERK1/2), and of postsynaptic TrkB, required ERα in females only, and that postsynaptic ERα levels are higher in females than in males. Several downstream signaling events involved in LTP were comparable between the sexes. In contrast to endogenous estrogen effects, infused estradiol facilitated LTP and synaptic signaling in females via both ERα and ERβ. The estrogen dependence of LTP in females was associated with a higher threshold for both inducing potentiation and acquiring spatial information. These results indicate that the observed sexual dimorphism in hippocampal LTP reflects differences in synaptic kinase activation, including both a weaker association with NMDA receptors and a greater ERα-mediated kinase activation in response to locally produced estrogen in females. We propose that male/female differences in mechanisms and threshold for field CA1 LTP contribute to differences in encoding specific types of memories.SIGNIFICANCE STATEMENT There is good evidence for male/female differences in memory-related cognitive function, but the neurobiological basis for this sexual dimorphism is not understood. Here we describe sex differences in synaptic function in a brain area that is critical for learning spatial cues. Our results show that female rodents have higher synaptic levels of estrogen receptor α (ERα) and, in contrast to males, require membrane ERα for the activation of signaling kinases that support long-term potentiation (LTP), a form of synaptic plasticity thought to underlie learning. The additional requirement of estrogen signaling in females resulted in a higher threshold for both LTP and hippocampal field CA1-dependent spatial learning. These results describe a synaptic basis for sexual dimorphism in encoding spatial information.
Collapse
|
38
|
Tonn Eisinger KR, Gross KS, Head BP, Mermelstein PG. Interactions between estrogen receptors and metabotropic glutamate receptors and their impact on drug addiction in females. Horm Behav 2018; 104:130-137. [PMID: 29505763 PMCID: PMC6131090 DOI: 10.1016/j.yhbeh.2018.03.001] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Revised: 02/28/2018] [Accepted: 03/01/2018] [Indexed: 02/07/2023]
Abstract
Contribution to Special Issue on Fast effects of steroids. Estrogen receptors α and β (ERα and ERβ) have a unique relationship with metabotropic glutamate receptors (mGluRs) in the female rodent brain such that estradiol is able to recruit intracellular G-protein signaling cascades to influence neuronal physiology, structure, and ultimately behavior. While this association between ERs and mGluRs exists in many cell types and brain regions, its effects are perhaps most striking in the nucleus accumbens (NAc). This review will discuss the original characterization of ER/mGluR signaling and how estradiol activity in the NAc confers increased sensitivity to drugs of abuse in females through this mechanism.
Collapse
Affiliation(s)
- Katherine R Tonn Eisinger
- Department of Neuroscience and Graduate Program in Neuroscience, University of Minnesota, Minneapolis, MN 55455, USA
| | - Kellie S Gross
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, WI 53211, USA
| | - Brian P Head
- Department of Anesthesiology, University of California-San Diego, La Jolla, CA 92093, USA
| | - Paul G Mermelstein
- Department of Neuroscience and Graduate Program in Neuroscience, University of Minnesota, Minneapolis, MN 55455, USA.
| |
Collapse
|
39
|
Micevych PE, Sinchak K. Extranuclear signaling by ovarian steroids in the regulation of sexual receptivity. Horm Behav 2018; 104:4-14. [PMID: 29753716 PMCID: PMC6240501 DOI: 10.1016/j.yhbeh.2018.05.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Revised: 05/02/2018] [Accepted: 05/03/2018] [Indexed: 01/06/2023]
Affiliation(s)
- Paul E Micevych
- Dept of Neurobiology, David Geffen School of Medicine at UCLA, Laboratory of Neuroendocrinology of the UCLA Brain Research Institute, United States
| | - Kevin Sinchak
- Dept of Biological Sciences, California State University, Long Beach, United States.
| |
Collapse
|
40
|
Kow LM, Pfaff DW. Can distinctly different rapid estrogen actions share a common mechanistic step? Horm Behav 2018; 104:156-164. [PMID: 29476777 DOI: 10.1016/j.yhbeh.2018.02.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Revised: 02/15/2018] [Accepted: 02/19/2018] [Indexed: 12/23/2022]
Abstract
Contribution to Special Issue on Fast effects of steroids. This paper reviews early evidence for the existence of rapid, non-genomic effects of estrogens on neurons, and, further, proposes that these rapid effects are often synergistic with later, genomic effects. Finally, suggestions about potential molecular mechanisms underlying the rapid effects of estrogens are offered. A mechanistic step we propose to be common among rapid estrogenic actions includes membrane ER's binding to histamine, and NMDA receptors and subsequent dimerization, and clustering (respectively) in a manner that enhances histamine and NMDA actions.
Collapse
Affiliation(s)
- Lee-Ming Kow
- Laboratory of Neurobiology and Behavior, The Rockefeller University, New York, NY, United States.
| | - Donald W Pfaff
- Laboratory of Neurobiology and Behavior, The Rockefeller University, New York, NY, United States
| |
Collapse
|
41
|
Stincic TL, Rønnekleiv OK, Kelly MJ. Diverse actions of estradiol on anorexigenic and orexigenic hypothalamic arcuate neurons. Horm Behav 2018; 104:146-155. [PMID: 29626486 PMCID: PMC6196116 DOI: 10.1016/j.yhbeh.2018.04.001] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2018] [Revised: 03/29/2018] [Accepted: 04/02/2018] [Indexed: 12/13/2022]
Abstract
Contribution to Special Issue on Fast effects of steroids. There is now compelling evidence for membrane-associated estrogen receptors in hypothalamic neurons that are critical for the hypothalamic control of homeostatic functions. It has been known for some time that estradiol (E2) can rapidly alter hypothalamic neuronal activity within seconds, indicating that some cellular effects can occur via membrane initiated events. However, our understanding of how E2 signals via membrane-associated receptors and how these signals impact physiological functions is only just emerging. Thus, E2 can affect second messenger systems including calcium mobilization and a plethora of kinases to alter cell excitability and even gene transcription in hypothalamic neurons. One population of hypothalamic neurons, the anorexigenic proopiomelanocortin (POMC) neurons, has long been considered to be a target of E2's actions based on gene (Pomc) expression studies. However, we now know that E2 can rapidly alter POMC neuronal activity within seconds and activate several intracellular signaling cascades that ultimately affect gene expression, actions which are critical for maintaining sensitivity to insulin in metabolically stressed states. E2 also affects the orexigenic Neuropeptide Y/Agouti-related Peptide (NPY/AgRP) neurons in similarly rapid but antagonistic manner. Therefore, this review will summarize our current state of knowledge of how E2 signals via rapid membrane-initiated and intracellular signaling cascades in POMC and NPY/AgRP neurons to regulate energy homeostasis.
Collapse
Affiliation(s)
- Todd L Stincic
- Department of Physiology and Pharmacology, Oregon Health & Science University, Portland, OR 97239, USA
| | - Oline K Rønnekleiv
- Department of Physiology and Pharmacology, Oregon Health & Science University, Portland, OR 97239, USA; Department of Anesthesiology and Perioperative Medicine, Oregon Health & Science University, Portland, OR 97239, USA; Division of Neuroscience, Oregon Regional Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006, USA
| | - Martin J Kelly
- Department of Physiology and Pharmacology, Oregon Health & Science University, Portland, OR 97239, USA; Division of Neuroscience, Oregon Regional Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006, USA.
| |
Collapse
|
42
|
Barabás K, Godó S, Lengyel F, Ernszt D, Pál J, Ábrahám IM. Rapid non-classical effects of steroids on the membrane receptor dynamics and downstream signaling in neurons. Horm Behav 2018; 104:183-191. [PMID: 29775570 DOI: 10.1016/j.yhbeh.2018.05.008] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Revised: 05/08/2018] [Accepted: 05/09/2018] [Indexed: 12/26/2022]
Abstract
Contribution to Special Issue on Fast effects of steroids. Although rapid effects of steroid hormones on membrane receptors and intracellular signaling molecules have been extensively studied in neurons, we are only beginning to understand the molecular mechanisms behind these non-classical steroid actions. Single molecule tracking (SMT) studies on live cells demonstrated that surface trafficking of membrane receptors determines their ligand binding properties and downstream signaling events. Recent findings suggest that one of the underlying mechanisms of non-classical steroid actions is the alteration of receptor movements on the membrane surface. In order to highlight this novel aspect of steroid effects, we first address the types of receptor movements in the plasma membrane and the role of cortical actin dynamics in receptor movement. We then discuss how single molecules and the surface movements of receptors can be detected in live cells. Next, we review the fundamental processes, which determine the effect of steroids on the plasma membrane: steroid movement through the lipid bilayer and the role of steroid membrane receptors. Using glutamate and neurotrophin receptors (NTRs) as examples, we demonstrate the features of receptor dynamics in the membrane. In addition, we survey the available data of rapid steroid actions on membrane receptor trafficking: we discuss how glucocorticoids act on the surface diffusion of glutamate receptor molecules and how estradiol acts on NTRs and gamma-aminobutyric acid type A receptors (GABAARs) and their related signaling events as well as on cortical actin. Finally, we address the physiological relevance of rapid steroid action on membrane receptors dynamics.
Collapse
Affiliation(s)
- Klaudia Barabás
- MTA NAP-B Molecular Neuroendocrinology Research Group, Institute of Physiology, Medical School, Centre for Neuroscience, Szentágothai Research Institute, University of Pécs, Pécs, Hungary
| | - Soma Godó
- MTA NAP-B Molecular Neuroendocrinology Research Group, Institute of Physiology, Medical School, Centre for Neuroscience, Szentágothai Research Institute, University of Pécs, Pécs, Hungary
| | - Ferenc Lengyel
- MTA NAP-B Molecular Neuroendocrinology Research Group, Institute of Physiology, Medical School, Centre for Neuroscience, Szentágothai Research Institute, University of Pécs, Pécs, Hungary
| | - Dávid Ernszt
- MTA NAP-B Molecular Neuroendocrinology Research Group, Institute of Physiology, Medical School, Centre for Neuroscience, Szentágothai Research Institute, University of Pécs, Pécs, Hungary
| | - József Pál
- MTA NAP-B Molecular Neuroendocrinology Research Group, Institute of Physiology, Medical School, Centre for Neuroscience, Szentágothai Research Institute, University of Pécs, Pécs, Hungary
| | - István M Ábrahám
- MTA NAP-B Molecular Neuroendocrinology Research Group, Institute of Physiology, Medical School, Centre for Neuroscience, Szentágothai Research Institute, University of Pécs, Pécs, Hungary.
| |
Collapse
|
43
|
Makdissy N, Haddad K, AlBacha JD, Chaker D, Ismail B, Azar A, Oreibi G, Ayoub D, Achkar I, Quilliot D, Fajloun Z. Essential role of ATP6AP2 enrichment in caveolae/lipid raft microdomains for the induction of neuronal differentiation of stem cells. Stem Cell Res Ther 2018; 9:132. [PMID: 29751779 PMCID: PMC5948768 DOI: 10.1186/s13287-018-0862-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Revised: 03/19/2018] [Accepted: 04/04/2018] [Indexed: 01/19/2023] Open
Abstract
Background The subcellular distribution of prorenin receptor and adaptor protein ATP6AP2 may affect neurogenesis. In this study, we hypothesized that ATP6AP2 expression and subcellular relocalization from caveolae/lipid raft microdomains (CLR-Ms) to intracellular sites may correlate with neuronal differentiation (Neu-Dif) of adipose-derived mesenchymal stem cells (ADSCs). Methods Human ADSCs isolated from 24 healthy donors and 24 patients with neurological disorders (ND) were cultured and induced for Neu-Dif. The mechanism of action of ATP6AP2 and the impact of its localization within the plasma membrane (particularly CLR-Ms) and intracellular sites on several pathways (mitogen-activated protein kinase, Wnt(s) signaling and others) and intracellular calcium and exosome release were evaluated. The impact of CLR-Ms on ATP6AP2 or vice versa was determined by pharmacological disruption of CLR-Ms or siATP6AP2 assays. Results In patients with ND, loss of ATP6AP2 from CLR-Ms correlated with an inhibition of Neu-Dif and signaling. However, its relocalization in CLR-Ms was positively correlated to induction of Neu-Dif in healthy subjects. An apparent switch from canonical to noncanonical Wnt signaling as well as from caveolin to flotillin occurs concurrently with the increases of ATP6AP2 expression during neurogenesis. Stimulation by renin activates ERK/JNK/CREB/c-Jun but failed to induce β-catenin. Wnt5a enhanced the renin-induced JNK responsiveness. Gα proteins crosslink ATP6AP2 to caveolin where a switch from Gαi to Gαq is necessary for Neu-Dif. In ATP6AP2-enriched CLR-Ms, the release of exosomes was induced dependently from the intracellular Ca2+ and Gαq. Pharmacological disruption of CLR-M formation/stability impairs both ATP6AP2 localization and Neu-Dif in addition to reducing exosome release, indicating an essential role of ATP6AP2 enrichment in CLR-Ms for the induction of Neu-Dif. The mechanism is dependent on CLR-M dynamics, particularly the membrane fluidity. Knockdown of ATP6AP2 inhibited Neu-Dif but increased astrocytic-Dif, depleted ATP6AP2/flotillin/Gαq but accumulated caveolin/Gαi in CLR-Ms, and blocked the activation of JNK/ERK/c-Jun/CREB/exosome release. siATP6AP2 cells treated with sphingomyelinase/methyl-β-cyclodextrin reversed the levels of caveolin/flotillin in CLR-Ms but did not induce Neu-Dif, indicating the crucial relocalization of ATP6AP2 in CLR-Ms for neurogenesis. Treatment of ND-derived cells with nSMase showed reversibility in ATP6AP2 abundance in CLR-Ms and enhanced Neu-Dif. Conclusions This study gives evidence of the determinant role of CLR-M ATP6AP2 localization for neuronal and oligodendrocyte differentiation involving mechanisms of switches from Gαi/caveolin/canonical to Gαq/flotillin/PCP, the ERK/JNK pathway and Ca2+-dependent release of exosomes and as a potential target of drug therapy for neurodegenerative disorders. Electronic supplementary material The online version of this article (10.1186/s13287-018-0862-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Nehman Makdissy
- Department of Biology, Lebanese University, Faculty of Sciences III, Kobbe, Lebanon.
| | - Katia Haddad
- Department of Biology, Lebanese University, Faculty of Sciences III, Kobbe, Lebanon
| | - Jeanne D'arc AlBacha
- Doctoral School for Sciences and Technology, Azm Center for the Research in Biotechnology and its Applications, Lebanese University, Tripoli, Lebanon
| | - Diana Chaker
- Doctoral School for Sciences and Technology, Azm Center for the Research in Biotechnology and its Applications, Lebanese University, Tripoli, Lebanon
| | - Bassel Ismail
- Doctoral School for Sciences and Technology, Faculty of Sciences I, Lebanese University, Hadath, Lebanon
| | - Albert Azar
- Reviva Regenerative Medicine Center, Human Genetic Center, Middle East Institute of Health Hospital, Bsalim, Lebanon
| | - Ghada Oreibi
- Reviva Regenerative Medicine Center, Human Genetic Center, Middle East Institute of Health Hospital, Bsalim, Lebanon
| | - David Ayoub
- Ayoub Clinic Lebanon and Department of Neuroloradiology, Limoges University Hospital, EA3842, Limoges, Lebanon
| | | | - Didier Quilliot
- Diabetologia-Endocrinology & Nutrition, CHRU Nancy, INSERM 954, University Henri Poincaré, Faculty of Medicine, Nancy, France
| | - Ziad Fajloun
- Department of Biology, Lebanese University, Faculty of Sciences III, Kobbe, Lebanon.,Doctoral School for Sciences and Technology, Azm Center for the Research in Biotechnology and its Applications, Lebanese University, Tripoli, Lebanon
| |
Collapse
|
44
|
Tonn Eisinger KR, Larson EB, Boulware MI, Thomas MJ, Mermelstein PG. Membrane estrogen receptor signaling impacts the reward circuitry of the female brain to influence motivated behaviors. Steroids 2018; 133:53-59. [PMID: 29195840 PMCID: PMC5864533 DOI: 10.1016/j.steroids.2017.11.013] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Revised: 11/20/2017] [Accepted: 11/23/2017] [Indexed: 12/12/2022]
Abstract
Within the adult female, estrogen signaling is well-described as an integral component of the physiologically significant hypothalamic-pituitary-gonadal axis. In rodents, the timing of ovulation is intrinsically entwined with the display of sexual receptivity. For decades, the importance of estradiol activating intracellular estrogen receptors within the hypothalamus and midbrain/spinal cord lordosis circuits has been appreciated. These signaling pathways primarily account for the ability of the female to reproduce. Yet, often overlooked is that the desire to reproduce is also tightly regulated by estrogen receptor signaling. This lack of emphasis can be attributed to an absence of nuclear estrogen receptors in brain regions associated with reward, such as the nucleus accumbens, which are associated with motivated behaviors. This review outlines how membrane-localized estrogen receptors affect metabotropic glutamate receptor signaling within the rodent nucleus accumbens. In addition, we discuss how, as estrogens drive increased motivation for reproduction, they also produce the untoward side effect of heightening female vulnerability to drug addiction.
Collapse
Affiliation(s)
- Katherine R Tonn Eisinger
- Department of Neuroscience and Graduate Program in Neuroscience, University of Minnesota, Minneapolis, MN 55455, USA
| | - Erin B Larson
- Department of Neuroscience and Graduate Program in Neuroscience, University of Minnesota, Minneapolis, MN 55455, USA
| | - Marissa I Boulware
- Department of Obstetrics and Gynecology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Mark J Thomas
- Department of Neuroscience and Graduate Program in Neuroscience, University of Minnesota, Minneapolis, MN 55455, USA
| | - Paul G Mermelstein
- Department of Neuroscience and Graduate Program in Neuroscience, University of Minnesota, Minneapolis, MN 55455, USA.
| |
Collapse
|
45
|
Marin R, Diaz M. Estrogen Interactions With Lipid Rafts Related to Neuroprotection. Impact of Brain Ageing and Menopause. Front Neurosci 2018; 12:128. [PMID: 29559883 PMCID: PMC5845729 DOI: 10.3389/fnins.2018.00128] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Accepted: 02/16/2018] [Indexed: 12/22/2022] Open
Abstract
Estrogens (E2) exert a plethora of neuroprotective actions against aged-associated brain diseases, including Alzheimer's disease (AD). Part of these actions takes place through binding to estrogen receptors (ER) embedded in signalosomes, where numerous signaling proteins are clustered. Signalosomes are preferentially located in lipid rafts which are dynamic membrane microstructures characterized by a peculiar lipid composition enriched in gangliosides, saturated fatty acids, cholesterol, and sphingolipids. Rapid E2 interactions with ER-related signalosomes appear to trigger intracellular signaling ultimately leading to the activation of molecular mechanisms against AD. We have previously observed that the reduction of E2 blood levels occurring during menopause induced disruption of ER-signalosomes at frontal cortical brain areas. These molecular changes may reduce neuronal protection activities, as similar ER signalosome derangements were observed in AD brains. The molecular impairments may be associated with changes in the lipid composition of lipid rafts observed in neurons during menopause and AD. These evidences indicate that the changes in lipid raft structure during aging may be at the basis of alterations in the activity of ER and other neuroprotective proteins integrated in these membrane microstructures. Moreover, E2 is a homeostatic modulator of lipid rafts. Recent work has pointed to this relevant aspect of E2 activity to preserve brain integrity, through mechanisms affecting lipid uptake and local biosynthesis in the brain. Some evidences have demonstrated that estrogens and the docosahexaenoic acid (DHA) exert synergistic effects to stabilize brain lipid matrix. DHA is essential to enhance molecular fluidity at the plasma membrane, promoting functional macromolecular interactions in signaling platforms. In support of this, DHA detriment in neuronal lipid rafts has been associated with the most common age-associated neuropathologies, namely AD and Parkinson disease. Altogether, these findings indicate that E2 may participate in brain preservation through a dual membrane-related mechanism. On the one hand, E2 interacting with ER related signalosomes may protect against neurotoxic insults. On the other hand, E2 may exert lipostatic actions to preserve lipid balance in neuronal membrane microdomains. The different aspects of the emerging multifunctional role of estrogens in membrane-related signalosomes will be discussed in this review.
Collapse
Affiliation(s)
- Raquel Marin
- Laboratory of Cellular Neurobiology, Department of Basic Medical Sciences, Medicine, Faculty of Health Sciences, University of La Laguna, Tenerife, Spain.,Fisiología y Biofísica de la Membrana Celular en Patologías Neurodegenerativas y Tumorales, Consejo Superior de Investigaciones Cientificas, Unidad Asociada de Investigación, Universidad de La Laguna Tenerife, Tenerife, Spain
| | - Mario Diaz
- Fisiología y Biofísica de la Membrana Celular en Patologías Neurodegenerativas y Tumorales, Consejo Superior de Investigaciones Cientificas, Unidad Asociada de Investigación, Universidad de La Laguna Tenerife, Tenerife, Spain.,Laboratory of Membrane Physiology and Biophysics, Department of Animal Biology, Edaphology and Geology, University of La Laguna, Tenerife, Spain
| |
Collapse
|
46
|
Zárate S, Stevnsner T, Gredilla R. Role of Estrogen and Other Sex Hormones in Brain Aging. Neuroprotection and DNA Repair. Front Aging Neurosci 2018. [PMID: 29311911 DOI: 10.3389/fnagi.2017.00430/xml/nlm] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/07/2023] Open
Abstract
Aging is an inevitable biological process characterized by a progressive decline in physiological function and increased susceptibility to disease. The detrimental effects of aging are observed in all tissues, the brain being the most important one due to its main role in the homeostasis of the organism. As our knowledge about the underlying mechanisms of brain aging increases, potential approaches to preserve brain function rise significantly. Accumulating evidence suggests that loss of genomic maintenance may contribute to aging, especially in the central nervous system (CNS) owing to its low DNA repair capacity. Sex hormones, particularly estrogens, possess potent antioxidant properties and play important roles in maintaining normal reproductive and non-reproductive functions. They exert neuroprotective actions and their loss during aging and natural or surgical menopause is associated with mitochondrial dysfunction, neuroinflammation, synaptic decline, cognitive impairment and increased risk of age-related disorders. Moreover, loss of sex hormones has been suggested to promote an accelerated aging phenotype eventually leading to the development of brain hypometabolism, a feature often observed in menopausal women and prodromal Alzheimer's disease (AD). Although data on the relation between sex hormones and DNA repair mechanisms in the brain is still limited, various investigations have linked sex hormone levels with different DNA repair enzymes. Here, we review estrogen anti-aging and neuroprotective mechanisms, which are currently an area of intense study, together with the effect they may have on the DNA repair capacity in the brain.
Collapse
Affiliation(s)
- Sandra Zárate
- Instituto de Investigaciones Biomédicas (INBIOMED, UBA-CONICET), Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina.,Departamento de Histología, Embriología, Biología Celular y Genética, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Tinna Stevnsner
- Danish Center for Molecular Gerontology and Danish Aging Research Center, Department of Molecular Biology and Genetics, University of Aarhus, Aarhus, Denmark
| | - Ricardo Gredilla
- Department of Physiology, Faculty of Medicine, Complutense University, Madrid, Spain
| |
Collapse
|
47
|
MacDonald JL, Fame RM, Gillis-Buck EM, Macklis JD. Caveolin1 Identifies a Specific Subpopulation of Cerebral Cortex Callosal Projection Neurons (CPN) Including Dual Projecting Cortical Callosal/Frontal Projection Neurons (CPN/FPN). eNeuro 2018; 5:ENEURO.0234-17.2017. [PMID: 29379878 PMCID: PMC5780842 DOI: 10.1523/eneuro.0234-17.2017] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Revised: 12/11/2017] [Accepted: 12/19/2017] [Indexed: 12/27/2022] Open
Abstract
The neocortex is composed of many distinct subtypes of neurons that must form precise subtype-specific connections to enable the cortex to perform complex functions. Callosal projection neurons (CPN) are the broad population of commissural neurons that connect the cerebral hemispheres via the corpus callosum (CC). Currently, how the remarkable diversity of CPN subtypes and connectivity is specified, and how they differentiate to form highly precise and specific circuits, are largely unknown. We identify in mouse that the lipid-bound scaffolding domain protein Caveolin 1 (CAV1) is specifically expressed by a unique subpopulation of Layer V CPN that maintain dual ipsilateral frontal projections to premotor cortex. CAV1 is expressed by over 80% of these dual projecting callosal/frontal projection neurons (CPN/FPN), with expression peaking early postnatally as axonal and dendritic targets are being reached and refined. CAV1 is localized to the soma and dendrites of CPN/FPN, a unique population of neurons that shares information both between hemispheres and with premotor cortex, suggesting function during postmitotic development and refinement of these neurons, rather than in their specification. Consistent with this, we find that Cav1 function is not necessary for the early specification of CPN/FPN, or for projecting to their dual axonal targets. CPN subtype-specific expression of Cav1 identifies and characterizes a first molecular component that distinguishes this functionally unique projection neuron population, a population that expands in primates, and is prototypical of additional dual and higher-order projection neuron subtypes.
Collapse
Affiliation(s)
- Jessica L. MacDonald
- Department of Stem Cell and Regenerative Biology, Center for Brain Science, and Harvard Stem Cell Institute, Harvard University, Cambridge, MA 02138
- Department of Biology, Syracuse University, Syracuse, NY 13244
| | - Ryann M. Fame
- Department of Stem Cell and Regenerative Biology, Center for Brain Science, and Harvard Stem Cell Institute, Harvard University, Cambridge, MA 02138
| | - Eva M. Gillis-Buck
- Department of Stem Cell and Regenerative Biology, Center for Brain Science, and Harvard Stem Cell Institute, Harvard University, Cambridge, MA 02138
| | - Jeffrey D. Macklis
- Department of Stem Cell and Regenerative Biology, Center for Brain Science, and Harvard Stem Cell Institute, Harvard University, Cambridge, MA 02138
| |
Collapse
|
48
|
Zárate S, Stevnsner T, Gredilla R. Role of Estrogen and Other Sex Hormones in Brain Aging. Neuroprotection and DNA Repair. Front Aging Neurosci 2017; 9:430. [PMID: 29311911 PMCID: PMC5743731 DOI: 10.3389/fnagi.2017.00430] [Citation(s) in RCA: 188] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Accepted: 12/14/2017] [Indexed: 12/13/2022] Open
Abstract
Aging is an inevitable biological process characterized by a progressive decline in physiological function and increased susceptibility to disease. The detrimental effects of aging are observed in all tissues, the brain being the most important one due to its main role in the homeostasis of the organism. As our knowledge about the underlying mechanisms of brain aging increases, potential approaches to preserve brain function rise significantly. Accumulating evidence suggests that loss of genomic maintenance may contribute to aging, especially in the central nervous system (CNS) owing to its low DNA repair capacity. Sex hormones, particularly estrogens, possess potent antioxidant properties and play important roles in maintaining normal reproductive and non-reproductive functions. They exert neuroprotective actions and their loss during aging and natural or surgical menopause is associated with mitochondrial dysfunction, neuroinflammation, synaptic decline, cognitive impairment and increased risk of age-related disorders. Moreover, loss of sex hormones has been suggested to promote an accelerated aging phenotype eventually leading to the development of brain hypometabolism, a feature often observed in menopausal women and prodromal Alzheimer's disease (AD). Although data on the relation between sex hormones and DNA repair mechanisms in the brain is still limited, various investigations have linked sex hormone levels with different DNA repair enzymes. Here, we review estrogen anti-aging and neuroprotective mechanisms, which are currently an area of intense study, together with the effect they may have on the DNA repair capacity in the brain.
Collapse
Affiliation(s)
- Sandra Zárate
- Instituto de Investigaciones Biomédicas (INBIOMED, UBA-CONICET), Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
- Departamento de Histología, Embriología, Biología Celular y Genética, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Tinna Stevnsner
- Danish Center for Molecular Gerontology and Danish Aging Research Center, Department of Molecular Biology and Genetics, University of Aarhus, Aarhus, Denmark
| | - Ricardo Gredilla
- Department of Physiology, Faculty of Medicine, Complutense University, Madrid, Spain
| |
Collapse
|
49
|
Balsevich G, Petrie GN, Hill MN. Endocannabinoids: Effectors of glucocorticoid signaling. Front Neuroendocrinol 2017; 47:86-108. [PMID: 28739508 DOI: 10.1016/j.yfrne.2017.07.005] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Revised: 07/17/2017] [Accepted: 07/19/2017] [Indexed: 01/17/2023]
Abstract
For decades, there has been speculation regarding the interaction of cannabinoids with glucocorticoid systems. Given the functional redundancy between many of the physiological effects of glucocorticoids and cannabinoids, it was originally speculated that the biological mechanisms of cannabinoids were mediated by direct interactions with glucocorticoid systems. With the discovery of the endocannabinoid system, additional research demonstrated that it was actually the opposite; glucocorticoids recruit endocannabinoid signaling, and that the engagement of endocannabinoid signaling mediated many of the neurobiological and physiological effects of glucocorticoids. With the development of advances in pharmacology and genetics, significant advances in this area have been made, and it is now clear that functional interactions between these systems are critical for a wide array of physiological processes. The current review acts a comprehensive summary of the contemporary state of knowledge regarding the biological interactions between glucocorticoids and endocannabinoids, and their potential role in health and disease.
Collapse
Affiliation(s)
- Georgia Balsevich
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
| | - Gavin N Petrie
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
| | - Matthew N Hill
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada; Mathison Centre for Mental Health Research and Education, University of Calgary, Calgary, AB, Canada; Departments of Cell Biology and Anatomy and Psychiatry, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.
| |
Collapse
|
50
|
Micevych PE, Mermelstein PG, Sinchak K. Estradiol Membrane-Initiated Signaling in the Brain Mediates Reproduction. Trends Neurosci 2017; 40:654-666. [PMID: 28969926 DOI: 10.1016/j.tins.2017.09.001] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Revised: 08/28/2017] [Accepted: 09/10/2017] [Indexed: 12/21/2022]
Abstract
Over the past few years our understanding of estrogen signaling in the brain has expanded rapidly. Estrogens are synthesized in the periphery and in the brain, acting on multiple receptors to regulate gene transcription, neural function, and behavior. Various estrogen-sensitive signaling pathways often operate in concert within the same cell, increasing the complexity of the system. In females, estrogen concentrations fluctuate over the estrous/menstrual cycle, dynamically modulating estrogen receptor (ER) expression, activity, and trafficking. These dynamic changes influence multiple behaviors but are particularly important for reproduction. Using the female rodent model, we review our current understanding of estradiol signaling in the regulation of sexual receptivity.
Collapse
Affiliation(s)
- Paul E Micevych
- Department of Neurobiology, David Geffen School of Medicine at the University of California Los Angeles (UCLA), and Laboratory of Neuroendocrinology of the UCLA Brain Research Institute, Los Angeles, CA 90095, USA.
| | - Paul G Mermelstein
- Department of Neuroscience, University of Minnesota, Minneapolis, MN 55455, USA
| | - Kevin Sinchak
- Department of Biological Sciences, California State University, Long Beach, Long Beach, CA 90840, USA
| |
Collapse
|