1
|
Sebastian A, Shanmuganathan MAA, Tripathy C, Chakravarty S, Ghosh S. Understanding Neurogenesis and Neuritogenesis via Molecular Insights, Gender Influence, and Therapeutic Implications: Intervention of Nanomaterials. ACS APPLIED BIO MATERIALS 2025; 8:12-41. [PMID: 39718903 DOI: 10.1021/acsabm.4c01079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2024]
Abstract
Neurological disorders impact global health by affecting both central and peripheral nervous systems. Understanding the neurogenic processes, i.e., neurogenesis and neuritogenesis, is of paramount importance in the context of nervous system development and regeneration as they hold promising therapeutic implications. Neurogenesis forms functional neurons from precursor cells, while neuritogenesis involves extending neurites for neuron connections. This review discusses how these processes are influenced by genetics, epigenetics, neurotrophic factors, environment, neuroinflammation, and neurotransmitters. It also covers gender-specific aspects of neurogenesis and neuritogenesis, their impact on brain plasticity, and susceptibility to neurological disorders. Alterations in these processes, under the influence of cytokines, growth factors, neurotransmitters, and aging, are linked to neurological disorders and potential therapeutic targets. Gender-specific effects of pharmacological interventions, like SSRIs, TCAs, atypical antipsychotics, and lithium, are explored in this review. Hormone-mediated effects of BDNF and PPAR-γ agonists, as well as variations in efficacy and tolerability of MAOIs, AEDs, NMDA receptor modulators, and ampakines, are detailed for accurate therapeutic design. The review also discusses nanotechnology's significant contribution to neural tissue regeneration for mending neurodegenerative disorders, enhancing neuronal connectivity, and stem cell differentiation. Gold nanoparticles support hippocampal neurogenesis, while other nanoparticles aid neuron growth and neurite outgrowth. Quantum dots and nanolayered double hydroxides assist neuroregeneration, which improves brain drug delivery. Gender-specific responses to nanomedicines designed to enhance neuroregeneration have not been extensively investigated. However, we have specified certain gender-related variables that should be taken into account during the development of nanomedicines in an aim to improve therapeutic efficacy. Further research on gender-specific responses to nanomedicines in neural processes could enhance personalized treatments for neurological disorders, paving the way for novel therapeutic approaches in neuroscience.
Collapse
Affiliation(s)
- Aishwarya Sebastian
- Polymers & Functional Materials Division, CSIR- Indian Institute of Chemical Technology, Hyderabad 500007, India
| | - Mohanraj Alias Ayyappan Shanmuganathan
- Polymers & Functional Materials Division, CSIR- Indian Institute of Chemical Technology, Hyderabad 500007, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Chinmayee Tripathy
- Applied Biology Division, CSIR- Indian Institute of Chemical Technology, Hyderabad 500007, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Sumana Chakravarty
- Applied Biology Division, CSIR- Indian Institute of Chemical Technology, Hyderabad 500007, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Sutapa Ghosh
- Polymers & Functional Materials Division, CSIR- Indian Institute of Chemical Technology, Hyderabad 500007, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| |
Collapse
|
2
|
Varghese SM, Patel S, Nandan A, Jose A, Ghosh S, Sah RK, Menon B, K V A, Chakravarty S. Unraveling the Role of the Blood-Brain Barrier in the Pathophysiology of Depression: Recent Advances and Future Perspectives. Mol Neurobiol 2024; 61:10398-10447. [PMID: 38730081 DOI: 10.1007/s12035-024-04205-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Accepted: 04/19/2024] [Indexed: 05/12/2024]
Abstract
Depression is a highly prevalent psychological disorder characterized by persistent dysphoria, psychomotor retardation, insomnia, anhedonia, suicidal ideation, and a remarkable decrease in overall well-being. Despite the prevalence of accessible antidepressant therapies, many individuals do not achieve substantial improvement. Understanding the multifactorial pathophysiology and the heterogeneous nature of the disorder could lead the way toward better outcomes. Recent findings have elucidated the substantial impact of compromised blood-brain barrier (BBB) integrity on the manifestation of depression. BBB functions as an indispensable defense mechanism, tightly overseeing the transport of molecules from the periphery to preserve the integrity of the brain parenchyma. The dysfunction of the BBB has been implicated in a multitude of neurological disorders, and its disruption and consequent brain alterations could potentially serve as important factors in the pathogenesis and progression of depression. In this review, we extensively examine the pathophysiological relevance of the BBB and delve into the specific modifications of its components that underlie the complexities of depression. A particular focus has been placed on examining the effects of peripheral inflammation on the BBB in depression and elucidating the intricate interactions between the gut, BBB, and brain. Furthermore, this review encompasses significant updates on the assessment of BBB integrity and permeability, providing a comprehensive overview of the topic. Finally, we outline the therapeutic relevance and strategies based on BBB in depression, including COVID-19-associated BBB disruption and neuropsychiatric implications. Understanding the comprehensive pathogenic cascade of depression is crucial for shaping the trajectory of future research endeavors.
Collapse
Affiliation(s)
- Shamili Mariya Varghese
- Department of Pharmacology, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Sciences Campus, Kochi, Kerala, 682 041, India
| | - Shashikant Patel
- Applied Biology Division, CSIR-Indian Institute of Chemical Technology, Tarnaka, Uppal Road, Hyderabad, Telangana, 500007, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Amritasree Nandan
- Department of Pharmacology, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Sciences Campus, Kochi, Kerala, 682 041, India
| | - Anju Jose
- Department of Pharmacology, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Sciences Campus, Kochi, Kerala, 682 041, India
| | - Soumya Ghosh
- Applied Biology Division, CSIR-Indian Institute of Chemical Technology, Tarnaka, Uppal Road, Hyderabad, Telangana, 500007, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Ranjay Kumar Sah
- Department of Pharmacology, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Sciences Campus, Kochi, Kerala, 682 041, India
| | - Bindu Menon
- Department of Psychiatry, Amrita School of Medicine, Amrita Vishwa Vidyapeetham, AIMS Health Sciences Campus, Kochi, Kerala, 682 041, India
| | - Athira K V
- Department of Pharmacology, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Sciences Campus, Kochi, Kerala, 682 041, India.
| | - Sumana Chakravarty
- Applied Biology Division, CSIR-Indian Institute of Chemical Technology, Tarnaka, Uppal Road, Hyderabad, Telangana, 500007, India.
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India.
| |
Collapse
|
3
|
Wakonigg Alonso C, McElhatton F, O'Mahony B, Campbell M, Pollak TA, Stokes PRA. The blood-brain barrier in bipolar disorders: A systematic review. J Affect Disord 2024; 361:434-444. [PMID: 38897301 DOI: 10.1016/j.jad.2024.06.032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 05/13/2024] [Accepted: 06/13/2024] [Indexed: 06/21/2024]
Abstract
BACKGROUND Bipolar disorders (BD) are chronic, debilitating disorders. The blood-brain barrier (BBB) has been increasingly investigated in BD. This systematic review aimed to assess the available evidence on the relationship between BD and markers of BBB dysfunction. METHODS A systematic search in PubMed, Embase, PsycINFO, CINAHL and Web of Science was run where the primary outcomes were BBB markers such as S100B, albumin ratio, matrix metalloproteinase (MMP), cell adhesion molecule (CAM), and tight junction proteins. Techniques included blood, cerebrospinal fluid (CSF), post-mortem, genetic and imaging methods in BD compared to healthy controls. RESULTS 55 studies were identified, 38 of which found an association between BD and markers of BBB dysfunction. 16/29 studies found increased blood/CSF albumin ratio, S100B, CAMs or MMP levels in BD participants compared to controls. 5/19 post-mortem studies found increased levels of chondroitin sulphate proteoglycans, intercellular CAM, neurexin or claudin-5 mRNA in distinct locations throughout the brain in BD compared to controls. One imaging study identified extensive BBB leakage in 30 % of BD participants, compared to 0 % in controls. LIMITATIONS The diversity in methodologies used in the included studies makes direct comparison of results challenging. Furthermore, imaging methods are the gold standard, but only one study used them. Other markers are only indicative of BBB permeability. CONCLUSIONS This review suggests an association between BD and BBB dysfunction. Further research is needed to provide definite answers considering the existing literature's limitations, and to clarify whether this association provides a pathogenic mechanism, or is an epiphenomenon of BD.
Collapse
Affiliation(s)
- Clara Wakonigg Alonso
- Institute of Psychiatry & Psychology and Neuroscience, King's College London,United Kingdom.
| | - Frances McElhatton
- Institute of Psychiatry & Psychology and Neuroscience, King's College London,United Kingdom
| | - Brian O'Mahony
- Institute of Psychiatry & Psychology and Neuroscience, King's College London,United Kingdom
| | - Matthew Campbell
- Smurfit Institute of Genetics, Trinity College Dublin, Lincoln Place Gate, Dublin 2, Ireland
| | - Thomas A Pollak
- Dept of Psychosis Studies, Institute of Psychiatry & Psychology and Neuroscience, King's College London, United Kingdom; South London and Maudsley NHS Foundation Trust,Bethlem Royal Hospital, Monks Orchard Road, Beckenham, Kent, BR3 3BX, United Kingdom
| | - Paul R A Stokes
- South London and Maudsley NHS Foundation Trust,Bethlem Royal Hospital, Monks Orchard Road, Beckenham, Kent, BR3 3BX, United Kingdom; Centre for Affective Disorders, Department of Psychological Medicine, Institute of Psychiatry & Psychology and Neuroscience, King's College London,United Kingdom
| |
Collapse
|
4
|
Liu X, Li F, Wen X, Zheng J, Pan W, Li Z. 18F-Labeled dihydropyridines via Hantzsch reaction for positron emission tomography of P-glycoprotein dysfunction. Bioorg Med Chem Lett 2024; 109:129818. [PMID: 38823726 DOI: 10.1016/j.bmcl.2024.129818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 05/13/2024] [Accepted: 05/27/2024] [Indexed: 06/03/2024]
Abstract
Despite the availability of various 11C-labeled positron emission tomography (PET) tracers for assessing P-glycoprotein (P-gp) function, there are still limitations related to complex metabolism, high lipophilicity, and low baseline uptake. This study aimed to address these issues by exploring a series of customized dihydropyridines (DHPs) with enhanced stability and reduced lipophilicity as alternative PET tracers for P-gp dysfunction. Compared with verapamil and the rest DHPs, dimethyl 4-(4-fluorophenyl)-2,6-dimethyl-1,4-dihydropyridine-3,5-dicarboxylate (1) exhibited superior cellular uptake differences between the human gastric cancer cell line SGC7901 and its drug-resistant counterpart. [18F]1 is successfully synthesized using a novel "hot-Hantzsch" approach in 22.1 ± 0.1 % radiochemical yields. MicroPET/CT imaging demonstrated that the uptake of [18F]1 in the brains of P-gp blocked mice increased by > 3 times compared to the control group. Additionally, [18F]1 displayed favorable lipophilicity (log D = 2.3) and excellent clearance characteristics, making it a promising tracer candidate with low background noise and high contrast.
Collapse
Affiliation(s)
- Xia Liu
- State Key Laboratory of Vaccines for Infectious Diseases, Center for Molecular Imaging and Translational Medicine, Xiang An Biomedicine Laboratory, School of Public Health, Xiamen University, Xiamen, Fujian 361102, China; State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, Xiamen University, Xiamen, Fujian 361102, China
| | - Feng Li
- Department of Radiology, Hainan Women and Children's Medical Center, Haikou, Hainan 570206, China
| | - Xing Wen
- Department of Ultrasound, Chengdu Fifth People's Hospital, Chengdu, Sichuan 611130, China
| | - Jiamei Zheng
- State Key Laboratory of Vaccines for Infectious Diseases, Center for Molecular Imaging and Translational Medicine, Xiang An Biomedicine Laboratory, School of Public Health, Xiamen University, Xiamen, Fujian 361102, China; State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, Xiamen University, Xiamen, Fujian 361102, China
| | - Weimin Pan
- Department of Nuclear Medicine, Xiang'an Hospital of Xiamen University, Xiamen, Fujian 361102, China; Department of Nuclear Medicine, Quanzhou First Hospital, Quanzhou, Fujian 362002, China.
| | - Zijing Li
- State Key Laboratory of Vaccines for Infectious Diseases, Center for Molecular Imaging and Translational Medicine, Xiang An Biomedicine Laboratory, School of Public Health, Xiamen University, Xiamen, Fujian 361102, China; State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, Xiamen University, Xiamen, Fujian 361102, China.
| |
Collapse
|
5
|
Cunha S, Bicker J, Sereno J, Falcão A, Fortuna A. Blood brain barrier dysfunction in healthy aging and dementia: Why, how, what for? Ageing Res Rev 2024; 99:102395. [PMID: 38950867 DOI: 10.1016/j.arr.2024.102395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 06/03/2024] [Accepted: 06/23/2024] [Indexed: 07/03/2024]
Abstract
The blood brain barrier (BBB) is an indispensable structure that maintains the central nervous system (CNS) microenvironment for a correct neuronal function. It is composed by highly specialized microvessels, surrounded by astrocytes, pericytes, neurons and microglia cells, which tightly control the influx and efflux of substances to the brain parenchyma. During aging, the BBB becomes impaired, and it may contribute to the development of neurodegenerative and neurological disorders including Alzheimer's disease and other dementias. Restoring the BBB can be a strategy to prevent disease onset and development, reducing the symptoms of these conditions. This work critically reviews the major mechanisms underlying BBB breakdown in healthy and unhealthy aging, as well as biomarkers and methodologies that accurately assess its impairment. Complementarily, potential therapeutic targets are discussed as new strategies to restore the normal function of the BBB in aging.
Collapse
Affiliation(s)
- Susana Cunha
- Faculty of Pharmacy, FFUC, University of Coimbra, Coimbra 3000-548, Portugal
| | - Joana Bicker
- Faculty of Pharmacy, FFUC, University of Coimbra, Coimbra 3000-548, Portugal; CIBIT - Coimbra Institute for Biomedical Imaging and Translational Research, University of Coimbra, Coimbra, Portugal
| | - José Sereno
- CIBIT - Coimbra Institute for Biomedical Imaging and Translational Research, University of Coimbra, Coimbra, Portugal
| | - Amílcar Falcão
- Faculty of Pharmacy, FFUC, University of Coimbra, Coimbra 3000-548, Portugal; CIBIT - Coimbra Institute for Biomedical Imaging and Translational Research, University of Coimbra, Coimbra, Portugal
| | - Ana Fortuna
- Faculty of Pharmacy, FFUC, University of Coimbra, Coimbra 3000-548, Portugal; CIBIT - Coimbra Institute for Biomedical Imaging and Translational Research, University of Coimbra, Coimbra, Portugal.
| |
Collapse
|
6
|
Chen T, Dai Y, Hu C, Lin Z, Wang S, Yang J, Zeng L, Li S, Li W. Cellular and molecular mechanisms of the blood-brain barrier dysfunction in neurodegenerative diseases. Fluids Barriers CNS 2024; 21:60. [PMID: 39030617 PMCID: PMC11264766 DOI: 10.1186/s12987-024-00557-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2024] [Accepted: 06/20/2024] [Indexed: 07/21/2024] Open
Abstract
BACKGROUND Maintaining the structural and functional integrity of the blood-brain barrier (BBB) is vital for neuronal equilibrium and optimal brain function. Disruptions to BBB performance are implicated in the pathology of neurodegenerative diseases. MAIN BODY Early indicators of multiple neurodegenerative disorders in humans and animal models include impaired BBB stability, regional cerebral blood flow shortfalls, and vascular inflammation associated with BBB dysfunction. Understanding the cellular and molecular mechanisms of BBB dysfunction in brain disorders is crucial for elucidating the sustenance of neural computations under pathological conditions and for developing treatments for these diseases. This paper initially explores the cellular and molecular definition of the BBB, along with the signaling pathways regulating BBB stability, cerebral blood flow, and vascular inflammation. Subsequently, we review current insights into BBB dynamics in Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, and multiple sclerosis. The paper concludes by proposing a unified mechanism whereby BBB dysfunction contributes to neurodegenerative disorders, highlights potential BBB-focused therapeutic strategies and targets, and outlines lessons learned and future research directions. CONCLUSIONS BBB breakdown significantly impacts the development and progression of neurodegenerative diseases, and unraveling the cellular and molecular mechanisms underlying BBB dysfunction is vital to elucidate how neural computations are sustained under pathological conditions and to devise therapeutic approaches.
Collapse
Affiliation(s)
- Tongli Chen
- School of Medicine, Hangzhou City University, Hangzhou, China
| | - Yan Dai
- School of Medicine, Hangzhou City University, Hangzhou, China
| | - Chenghao Hu
- School of Medicine, Hangzhou City University, Hangzhou, China
| | - Zihao Lin
- School of Medicine, Hangzhou City University, Hangzhou, China
| | - Shengzhe Wang
- School of Medicine, Hangzhou City University, Hangzhou, China
| | - Jing Yang
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, China.
- Institute of Brain and Cognitive Science, Hangzhou City University, Hangzhou, China.
| | - Linghui Zeng
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, China.
- Institute of Brain and Cognitive Science, Hangzhou City University, Hangzhou, China.
| | - Shanshan Li
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, China.
- Institute of Brain and Cognitive Science, Hangzhou City University, Hangzhou, China.
| | - Weiyun Li
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, China.
- Institute of Brain and Cognitive Science, Hangzhou City University, Hangzhou, China.
| |
Collapse
|
7
|
Wakid M, Almeida D, Aouabed Z, Rahimian R, Davoli MA, Yerko V, Leonova-Erko E, Richard V, Zahedi R, Borchers C, Turecki G, Mechawar N. Universal method for the isolation of microvessels from frozen brain tissue: A proof-of-concept multiomic investigation of the neurovasculature. Brain Behav Immun Health 2023; 34:100684. [PMID: 37822873 PMCID: PMC10562768 DOI: 10.1016/j.bbih.2023.100684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 08/29/2023] [Accepted: 09/06/2023] [Indexed: 10/13/2023] Open
Abstract
The neurovascular unit, comprised of vascular cell types that collectively regulate cerebral blood flow to meet the needs of coupled neurons, is paramount for the proper function of the central nervous system. The neurovascular unit gatekeeps blood-brain barrier properties, which experiences impairment in several central nervous system diseases associated with neuroinflammation and contributes to pathogenesis. To better understand function and dysfunction at the neurovascular unit and how it may confer inflammatory processes within the brain, isolation and characterization of the neurovascular unit is needed. Here, we describe a singular, standardized protocol to enrich and isolate microvessels from archived snap-frozen human and frozen mouse cerebral cortex using mechanical homogenization and centrifugation-separation that preserves the structural integrity and multicellular composition of microvessel fragments. For the first time, microvessels are isolated from postmortem ventromedial prefrontal cortex tissue and are comprehensively investigated as a structural unit using both RNA sequencing and Liquid Chromatography with tandem mass spectrometry (LC-MS/MS). Both the transcriptome and proteome are obtained and compared, demonstrating that the isolated brain microvessel is a robust model for the NVU and can be used to generate highly informative datasets in both physiological and disease contexts.
Collapse
Affiliation(s)
- Marina Wakid
- McGill Group for Suicide Studies, Douglas Research Centre, Montréal, Quebec, Canada
- Integrated Program in Neuroscience, McGill University, Montréal, Quebec, Canada
| | - Daniel Almeida
- McGill Group for Suicide Studies, Douglas Research Centre, Montréal, Quebec, Canada
- Integrated Program in Neuroscience, McGill University, Montréal, Quebec, Canada
| | - Zahia Aouabed
- McGill Group for Suicide Studies, Douglas Research Centre, Montréal, Quebec, Canada
| | - Reza Rahimian
- McGill Group for Suicide Studies, Douglas Research Centre, Montréal, Quebec, Canada
| | | | - Volodymyr Yerko
- McGill Group for Suicide Studies, Douglas Research Centre, Montréal, Quebec, Canada
| | - Elena Leonova-Erko
- McGill Group for Suicide Studies, Douglas Research Centre, Montréal, Quebec, Canada
| | - Vincent Richard
- Segal Cancer Proteomics Centre, Lady Davis Institute for Medical Research, Jewish General Hospital, McGill University, Montréal, Quebec, Canada
| | - René Zahedi
- Segal Cancer Proteomics Centre, Lady Davis Institute for Medical Research, Jewish General Hospital, McGill University, Montréal, Quebec, Canada
| | - Christoph Borchers
- Segal Cancer Proteomics Centre, Lady Davis Institute for Medical Research, Jewish General Hospital, McGill University, Montréal, Quebec, Canada
| | - Gustavo Turecki
- McGill Group for Suicide Studies, Douglas Research Centre, Montréal, Quebec, Canada
- Integrated Program in Neuroscience, McGill University, Montréal, Quebec, Canada
- Department of Psychiatry, McGill University, Montréal, Quebec, Canada
| | - Naguib Mechawar
- McGill Group for Suicide Studies, Douglas Research Centre, Montréal, Quebec, Canada
- Integrated Program in Neuroscience, McGill University, Montréal, Quebec, Canada
- Department of Psychiatry, McGill University, Montréal, Quebec, Canada
| |
Collapse
|
8
|
Axelrod S, Li X, Sun Y, Lincoln S, Terceros A, O’Neil J, Wang Z, Nguyen A, Vora A, Spicer C, Shapiro B, Young MW. The Drosophila blood-brain barrier regulates sleep via Moody G protein-coupled receptor signaling. Proc Natl Acad Sci U S A 2023; 120:e2309331120. [PMID: 37831742 PMCID: PMC10589661 DOI: 10.1073/pnas.2309331120] [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: 06/02/2023] [Accepted: 08/28/2023] [Indexed: 10/15/2023] Open
Abstract
Sleep is vital for most animals, yet its mechanism and function remain unclear. We found that permeability of the BBB (blood-brain barrier)-the organ required for the maintenance of homeostatic levels of nutrients, ions, and other molecules in the brain-is modulated by sleep deprivation (SD) and can cell-autonomously effect sleep changes. We observed increased BBB permeability in known sleep mutants as well as in acutely sleep-deprived animals. In addition to molecular tracers, SD-induced BBB changes also increased the penetration of drugs used in the treatment of brain pathologies. After chronic/genetic or acute SD, rebound sleep or administration of the sleeping aid gaboxadol normalized BBB permeability, showing that SD effects on the BBB are reversible. Along with BBB permeability, RNA levels of the BBB master regulator moody are modulated by sleep. Conversely, altering BBB permeability alone through glia-specific modulation of moody, gαo, loco, lachesin, or neuroglian-each a well-studied regulator of BBB function-was sufficient to induce robust sleep phenotypes. These studies demonstrate a tight link between BBB permeability and sleep and indicate a unique role for the BBB in the regulation of sleep.
Collapse
Affiliation(s)
- Sofia Axelrod
- Laboratory of Genetics, The Rockefeller University, New York, NY10065
| | - Xiaoling Li
- International Personalized Cancer Center, Tianjin Cancer Hospital Airport Hospital, Tianjin300308, China
| | - Yingwo Sun
- Laboratory of Genetics, The Rockefeller University, New York, NY10065
| | - Samantha Lincoln
- Laboratory of Genetics, The Rockefeller University, New York, NY10065
| | - Andrea Terceros
- Laboratory of Genetics, The Rockefeller University, New York, NY10065
| | - Jenna O’Neil
- Laboratory of Genetics, The Rockefeller University, New York, NY10065
| | - Zikun Wang
- Laboratory of Genetics, The Rockefeller University, New York, NY10065
| | - Andrew Nguyen
- Laboratory of Genetics, The Rockefeller University, New York, NY10065
| | - Aabha Vora
- Laboratory of Genetics, The Rockefeller University, New York, NY10065
| | - Carmen Spicer
- Laboratory of Genetics, The Rockefeller University, New York, NY10065
| | - Benjamin Shapiro
- Laboratory of Genetics, The Rockefeller University, New York, NY10065
| | - Michael W. Young
- Laboratory of Genetics, The Rockefeller University, New York, NY10065
| |
Collapse
|
9
|
Ju L, Glastad KM, Sheng L, Gospocic J, Kingwell CJ, Davidson SM, Kocher SD, Bonasio R, Berger SL. Hormonal gatekeeping via the blood-brain barrier governs caste-specific behavior in ants. Cell 2023; 186:4289-4309.e23. [PMID: 37683635 PMCID: PMC10807403 DOI: 10.1016/j.cell.2023.08.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 05/10/2023] [Accepted: 08/01/2023] [Indexed: 09/10/2023]
Abstract
Here, we reveal an unanticipated role of the blood-brain barrier (BBB) in regulating complex social behavior in ants. Using scRNA-seq, we find localization in the BBB of a key hormone-degrading enzyme called juvenile hormone esterase (Jhe), and we show that this localization governs the level of juvenile hormone (JH3) entering the brain. Manipulation of the Jhe level reprograms the brain transcriptome between ant castes. Although ant Jhe is retained and functions intracellularly within the BBB, we show that Drosophila Jhe is naturally extracellular. Heterologous expression of ant Jhe into the Drosophila BBB alters behavior in fly to mimic what is seen in ants. Most strikingly, manipulation of Jhe levels in ants reprograms complex behavior between worker castes. Our study thus uncovers a remarkable, potentially conserved role of the BBB serving as a molecular gatekeeper for a neurohormonal pathway that regulates social behavior.
Collapse
Affiliation(s)
- Linyang Ju
- Department of Biology, School of Arts and Sciences, University of Pennsylvania, Philadelphia, PA 19104, USA; Epigenetics Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA; Department of Cell and Developmental Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Karl M Glastad
- Epigenetics Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA; Department of Cell and Developmental Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA.
| | - Lihong Sheng
- Epigenetics Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA; Department of Cell and Developmental Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Janko Gospocic
- Epigenetics Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA; Department of Cell and Developmental Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA; Department of Urology and Institute of Neuropathology, Medical Center-University of Freiburg, Freiburg, Germany
| | - Callum J Kingwell
- Department of Ecology and Evolutionary Biology, Princeton University, Princeton, NJ 08544, USA
| | - Shawn M Davidson
- Lewis-Sigler Institute for Genomics, Princeton University, Princeton, NJ 08544, USA
| | - Sarah D Kocher
- Department of Ecology and Evolutionary Biology, Princeton University, Princeton, NJ 08544, USA; Lewis-Sigler Institute for Genomics, Princeton University, Princeton, NJ 08544, USA
| | - Roberto Bonasio
- Epigenetics Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA; Department of Cell and Developmental Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Shelley L Berger
- Department of Biology, School of Arts and Sciences, University of Pennsylvania, Philadelphia, PA 19104, USA; Epigenetics Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA; Department of Cell and Developmental Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA; Department of Genetics, Perelman School of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA.
| |
Collapse
|
10
|
Zola M, Bousquet E, Bourges JL, Azan F, Zhao M, Jaworski T, Pussard E, Behar-Cohen F. Ocular steroidome in human eyes and in eyes with complex central serous chorioretinopathy (CSCR). Sci Rep 2023; 13:14111. [PMID: 37644063 PMCID: PMC10465571 DOI: 10.1038/s41598-023-41126-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Accepted: 08/22/2023] [Indexed: 08/31/2023] Open
Abstract
The exact link between systemic and ocular endogenous corticoids (steroidome) is unclear and whether the ocular steroidome is altered in CSCR eyes is unknown. The aims of this study were to analyze the human steroidome in the aqueous humor as a function of age, sex and time of the day, to correlate systemic and ocular steroidome and to analyze the ocular steroidome in long lasting complex inactive CSCR. Based on our results, we present two CSCR cases treated by the combination of oral mineralocorticoid antagonist and glucocorticoids drops. In a cross-sectional study, aqueous humor (AH) was collected between 8am and 6 pm from 50 unaffected individuals (25 men and 25 women) and from 14 patients with chronic CSCR, during cataract surgery. In addition, simultaneous serum and AH were collected from 27 individuals undergoing cataract surgery and, simultaneous AH and vitreous were collected from 9 patients undergoing cataract and vitrectomy to estimate corticoids levels in the different compartments. The steroidome was determined using a LC-MS/MS method that quantifies 13 endogenous corticoids from the gluco, mineralocorticoid and androgen pathways. In AH and vitreous, the highest corticoid level is reached by cortisol (F), that represents less than 10% of F serum level. The cortisol levels in the serum did not correlate with ocular cortisol levels. Serum and ocular cortisone (E) levels correlate, although less than 5% of circulating E reaches the eye. The only mineralocorticoids measured in the AH were corticosterone (B) and its inactive form, the 11-desoxycorticosterone (A). There was no influence of circadian rhythm on cortisol ocular levels and there was no correlation between the age or the sex and the level of F, E, A, and B. In eyes with chronic inactive CSCR, the levels of the active glucocorticoid form F was lower than in control eyes and the F/E ratio was reduced by 50% but the B/A ratio was higher indicating imbalance towards active mineralocorticoids. Base on this observation, we propose to combine an antagonist of the mineralocorticoid receptor together with topical glucocorticoids in two CSCR patients, resistant to all other treatments, with favorable outcome. Our results indicate that the ocular psteroidome is highly regulated suggesting a local metabolism of ocular corticoids. In eyes with long-lasting complex inactive CSCR, the steroidome analysis shows lower active glucocorticoids and higher active mineralocorticoids.
Collapse
Affiliation(s)
- Marta Zola
- Department of Ophthalmology, Cochin Hospital, Assistance Publique-Hôpitaux de Paris, University of Paris Cité, Paris, France
- Centre de Recherche des Cordeliers, INSERM, Physiopathology of Ocular Diseases: Therapeutic Innovations, University Paris Cité, Paris, France
- Department of Ophthalmology, Hôpital Foch, Suresnes, France
| | - Elodie Bousquet
- Department of Ophthalmology, Cochin Hospital, Assistance Publique-Hôpitaux de Paris, University of Paris Cité, Paris, France
- Centre de Recherche des Cordeliers, INSERM, Physiopathology of Ocular Diseases: Therapeutic Innovations, University Paris Cité, Paris, France
| | - Jean-Louis Bourges
- Department of Ophthalmology, Cochin Hospital, Assistance Publique-Hôpitaux de Paris, University of Paris Cité, Paris, France
- Centre de Recherche des Cordeliers, INSERM, Physiopathology of Ocular Diseases: Therapeutic Innovations, University Paris Cité, Paris, France
| | - Fréderic Azan
- Department of Ophthalmology, Cochin Hospital, Assistance Publique-Hôpitaux de Paris, University of Paris Cité, Paris, France
| | - Min Zhao
- Centre de Recherche des Cordeliers, INSERM, Physiopathology of Ocular Diseases: Therapeutic Innovations, University Paris Cité, Paris, France
| | - Thara Jaworski
- Centre de Recherche des Cordeliers, INSERM, Physiopathology of Ocular Diseases: Therapeutic Innovations, University Paris Cité, Paris, France
| | - Eric Pussard
- Department of Genetic and Hormonology, Bicêtre Hospital, Assistance Publique-Hôpitaux de Paris, University of Paris-Saclay, Le Kremlin Bicêtre, France
| | - Francine Behar-Cohen
- Department of Ophthalmology, Cochin Hospital, Assistance Publique-Hôpitaux de Paris, University of Paris Cité, Paris, France.
- Centre de Recherche des Cordeliers, INSERM, Physiopathology of Ocular Diseases: Therapeutic Innovations, University Paris Cité, Paris, France.
- Department of Ophthalmology, Hôpital Foch, Suresnes, France.
| |
Collapse
|
11
|
Contreras EG, Klämbt C. The Drosophila blood-brain barrier emerges as a model for understanding human brain diseases. Neurobiol Dis 2023; 180:106071. [PMID: 36898613 DOI: 10.1016/j.nbd.2023.106071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 02/24/2023] [Accepted: 03/07/2023] [Indexed: 03/12/2023] Open
Abstract
The accurate regulation of the microenvironment within the nervous system is one of the key features characterizing complex organisms. To this end, neural tissue has to be physically separated from circulation, but at the same time, mechanisms must be in place to allow controlled transport of nutrients and macromolecules into and out of the brain. These roles are executed by cells of the blood-brain barrier (BBB) found at the interface of circulation and neural tissue. BBB dysfunction is observed in several neurological diseases in human. Although this can be considered as a consequence of diseases, strong evidence supports the notion that BBB dysfunction can promote the progression of brain disorders. In this review, we compile the recent evidence describing the contribution of the Drosophila BBB to the further understanding of brain disease features in human patients. We discuss the function of the Drosophila BBB during infection and inflammation, drug clearance and addictions, sleep, chronic neurodegenerative disorders and epilepsy. In summary, this evidence suggests that the fruit fly, Drosophila melanogaster, can be successfully employed as a model to disentangle mechanisms underlying human diseases.
Collapse
Affiliation(s)
- Esteban G Contreras
- University of Münster, Institute of Neuro- and Behavioral Biology, Badestr. 9, Münster, Germany.
| | - Christian Klämbt
- University of Münster, Institute of Neuro- and Behavioral Biology, Badestr. 9, Münster, Germany.
| |
Collapse
|
12
|
Nicolicht-Amorim P, Delgado-Garcia LM, Nakamura TKE, Courbassier NR, Mosini AC, Porcionatto MA. Simple and efficient protocol to isolate and culture brain microvascular endothelial cells from newborn mice. Front Cell Neurosci 2022; 16:949412. [PMID: 36313615 PMCID: PMC9606660 DOI: 10.3389/fncel.2022.949412] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Accepted: 09/06/2022] [Indexed: 10/09/2023] Open
Abstract
The neurovascular unit (NVU) is a multicellular structure comprising of neurons, glial cells, and non-neural cells, and it is supported by a specialized extracellular matrix, the basal lamina. Astrocytes, brain microvascular endothelial cells (BMECs), pericytes, and smooth muscle cells constitute the blood-brain barrier (BBB). BMECs have a mesodermal origin and invade the nervous system early in neural tube development, forming the BBB anatomical core. BMECs are connected by adherent junction complexes composed of integral membrane and cytoplasmic proteins. In vivo and in vitro studies have shown that, given the proximity and relationship with neural cells, BMECs acquire a unique gene expression profile, proteome, and specific mechanical and physical properties compared to endothelial cells from the general vasculature. BMECs are fundamental in maintaining brain homeostasis by regulating transcellular and paracellular transport of fluids, molecules, and cells. Therefore, it is essential to gain in-depth knowledge of the dynamic cellular structure of the cells in the NVU and their interactions with health and disease. Here we describe a significantly improved and simplified protocol using C57BL/6 newborn mice at postnatal day 1 (PND1) to isolate, purify, and culture BMECs monolayers in two different substrates (glass coverslips and transwell culture inserts). In vitro characterization and validation of the BMEC primary culture monolayers seeded on glass or insert included light microscopy, immunolabeling, and gene expression profile. Transendothelial electrical resistance (TEER) measurement and diffusion test were used as functional assays for adherent junction complexes and integrity and permeability of BMECs monolayers. The protocol presented here for the isolation and culture of BMECs is more straightforward than previously published protocols and yields a high number of purified cells. Finally, we tested BMECs function using the oxygen-glucose deprivation (OGD) model of hypoxia. This protocol may be suitable as a bioscaffold for secondary cell seeding allowing the study and better understanding of the NVU.
Collapse
Affiliation(s)
- Priscila Nicolicht-Amorim
- Laboratory of Molecular Neurobiology, Department of Biochemistry, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Lina M. Delgado-Garcia
- Laboratory of Molecular Neurobiology, Department of Biochemistry, Universidade Federal de São Paulo, São Paulo, Brazil
| | | | - Natália Rodrigues Courbassier
- Laboratory of Molecular Neurobiology, Department of Biochemistry, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Amanda Cristina Mosini
- Laboratory of Neurobiology, Department of Physiology, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Marimelia A. Porcionatto
- Laboratory of Molecular Neurobiology, Department of Biochemistry, Universidade Federal de São Paulo, São Paulo, Brazil
| |
Collapse
|
13
|
Ben-Zvi A, Liebner S. Developmental regulation of barrier- and non-barrier blood vessels in the CNS. J Intern Med 2022; 292:31-46. [PMID: 33665890 DOI: 10.1111/joim.13263] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Accepted: 02/01/2021] [Indexed: 12/22/2022]
Abstract
The blood-brain barrier (BBB) is essential for creating and maintaining tissue homeostasis in the central nervous system (CNS), which is key for proper neuronal function. In most vertebrates, the BBB is localized to microvascular endothelial cells that acquire barrier properties during angiogenesis of the neuroectoderm. Complex and continuous tight junctions, and the lack of fenestrae combined with low pinocytotic activity render the BBB endothelium a tight barrier for water-soluble molecules that may only enter the CNS via specific transporters. The differentiation of these unique endothelial properties during embryonic development is initiated by endothelial-specific flavours of the Wnt/β-catenin pathway in a precise spatiotemporal manner. In this review, we summarize the currently known cellular (neural precursor and endothelial cells) and molecular (VEGF and Wnt/β-catenin) mechanisms mediating brain angiogenesis and barrier formation. Moreover, we introduce more recently discovered crosstalk with cellular and acellular elements within the developing CNS such as the extracellular matrix. We discuss recent insights into the downstream molecular mechanisms of Wnt/β-catenin in particular, the recently identified target genes like Foxf2, Foxl2, Foxq1, Lef1, Ppard, Zfp551, Zic3, Sox17, Apcdd1 and Fgfbp1 that are involved in refining and maintaining barrier characteristics in the mature BBB endothelium. Additionally, we elute to recent insight into barrier heterogeneity and differential endothelial barrier properties within the CNS, focussing on the circumventricular organs as well as on the neurogenic niches in the subventricular zone and the hippocampus. Finally, open questions and future BBB research directions are highlighted in the context of taking benefit from understanding BBB development for strategies to modulate BBB function under pathological conditions.
Collapse
Affiliation(s)
- A Ben-Zvi
- From the, The Department of Developmental Biology and Cancer Research, Institute for Medical Research IMRIC, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - S Liebner
- Institute of Neurology (Edinger Institute), University Hospital, Goethe University Frankfurt, Frankfurt am Main, Germany.,Excellence Cluster Cardio-Pulmonary Systems (ECCPS), Partner Site Frankfurt, Frankfurt am Main, Germany.,German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Frankfurt am Main, Germany
| |
Collapse
|
14
|
Pogodalla N, Winkler B, Klämbt C. Glial Tiling in the Insect Nervous System. Front Cell Neurosci 2022; 16:825695. [PMID: 35250488 PMCID: PMC8891220 DOI: 10.3389/fncel.2022.825695] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 01/06/2022] [Indexed: 11/30/2022] Open
Abstract
The Drosophila nervous system comprises a small number of well characterized glial cell classes. The outer surface of the central nervous system (CNS) is protected by a glial derived blood-brain barrier generated by perineurial and subperineurial glia. All neural stem cells and all neurons are engulfed by cortex glial cells. The inner neuropil region, that harbors all synapses and dendrites, is covered by ensheathing glia and infiltrated by astrocyte-like glial cells. All these glial cells show a tiled organization with an often remarkable plasticity where glial cells of one cell type invade the territory of the neighboring glial cell type upon its ablation. Here, we summarize the different glial tiling patterns and based on the different modes of cell-cell contacts we hypothesize that different molecular mechanisms underlie tiling of the different glial cell types.
Collapse
|
15
|
The nuclear receptor Hr46/Hr3 is required in the blood brain barrier of mature males for courtship. PLoS Genet 2022; 18:e1009519. [PMID: 35077443 PMCID: PMC8815886 DOI: 10.1371/journal.pgen.1009519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 02/04/2022] [Accepted: 12/24/2021] [Indexed: 11/19/2022] Open
Abstract
The blood brain barrier (BBB) forms a stringent barrier that protects the brain from components in the circulation that could interfere with neuronal function. At the same time, the BBB enables selective transport of critical nutrients and other chemicals to the brain. Beyond these functions, another recently recognized function is even less characterized, specifically the role of the BBB in modulating behavior by affecting neuronal function in a sex-dependent manner. Notably, signaling in the adult Drosophila BBB is required for normal male courtship behavior. Courtship regulation also relies on male-specific molecules in the BBB. Our previous studies have demonstrated that adult feminization of these cells in males significantly lowered courtship. Here, we conducted microarray analysis of BBB cells isolated from males and females. Findings revealed that these cells contain male- and female-enriched transcripts, respectively. Among these transcripts, nuclear receptor Hr46/Hr3 was identified as a male-enriched BBB transcript. Hr46/Hr3 is best known for its essential roles in the ecdysone response during development and metamorphosis. In this study, we demonstrate that Hr46/Hr3 is specifically required in the BBB cells for courtship behavior in mature males. The protein is localized in the nuclei of sub-perineurial glial cells (SPG), indicating that it might act as a transcriptional regulator. These data provide a catalogue of sexually dimorphic BBB transcripts and demonstrate a physiological adult role for the nuclear receptor Hr46/Hr3 in the regulation of male courtship, a novel function that is independent of its developmental role.
Collapse
|
16
|
Contreras EG, Sierralta J. The Fly Blood-Brain Barrier Fights Against Nutritional Stress. Neurosci Insights 2022; 17:26331055221120252. [PMID: 36225749 PMCID: PMC9549514 DOI: 10.1177/26331055221120252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 08/01/2022] [Indexed: 11/17/2022] Open
Abstract
In the wild, animals face different challenges including multiple events of food
scarcity. How they overcome these conditions is essential for survival. Thus,
adaptation mechanisms evolved to allow the development and survival of an
organism during nutrient restriction periods. Given the high energy demand of
the nervous system, the molecular mechanisms of adaptation to malnutrition are
of great relevance to fuel the brain. The blood-brain barrier (BBB) is the
interface between the central nervous system (CNS) and the circulatory system.
The BBB mediates the transport of macromolecules in and out of the CNS, and
therefore, it can buffer changes in nutrient availability. In this review, we
collect the current evidence using the fruit fly, Drosophila
melanogaster, as a model of the role of the BBB in the adaptation
to starvation. We discuss the role of the Drosophila BBB during
nutrient deprivation as a potential sensor for circulating nutrients, and
transient nutrient storage as a regulator of the CNS neurogenic niche.
Collapse
Affiliation(s)
- Esteban G Contreras
- Institute of Neuro- and Behavioral Biology, University of Münster, Münster, Germany
| | - Jimena Sierralta
- Biomedical Neuroscience Institute and Department of Neuroscience, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| |
Collapse
|
17
|
Behar-Cohen F, Zhao M. Mineralocorticoid pathway in retinal health and diseases. Br J Pharmacol 2021; 179:3190-3204. [PMID: 34877649 DOI: 10.1111/bph.15770] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 10/29/2021] [Accepted: 11/22/2021] [Indexed: 11/30/2022] Open
Abstract
In the retina, the mineralocorticoid receptor (MR) is expressed in retinal and choroidal vessels and in cells from neural and glial origins. Like in the brain, the major ligand of the MR is cortisol and the MR/glucocorticoid receptor (GR) balance regulates the activation of the MR pathway. Experimental MR pathway activation using either pharmacological agents or transgenic manipulation favors retinal and choroidal pathology. In various models of retinal diseases, such as glaucomatous neuropathy, retinopathy of prematurity, ischemic retinopathies, diabetic retinopathy and choroidal neovascularization, MR antagonism exerts beneficial effects, demonstrating its potential in the treatment of major blinding retinal diseases. But specific formulations are required to optimize the bioavailability of MR antagonists in various compartments of the eye and molecular biomarkers of MR pathway activation remain to be identify in humans to select patients amenable to clinical trials.
Collapse
Affiliation(s)
- Francine Behar-Cohen
- Assistance Publique - Hôpitaux de Paris, Hôpital Cochin Ophtalmopole, Paris, France.,Centre de Recherche des Cordeliers, Sorbonne Université, Université de Paris, Inserm, From physiopathology of retinal diseases to clinical advances, Paris, France
| | - Min Zhao
- Centre de Recherche des Cordeliers, Sorbonne Université, Université de Paris, Inserm, From physiopathology of retinal diseases to clinical advances, Paris, France
| |
Collapse
|
18
|
Dunton AD, Göpel T, Ho DH, Burggren W. Form and Function of the Vertebrate and Invertebrate Blood-Brain Barriers. Int J Mol Sci 2021; 22:ijms222212111. [PMID: 34829989 PMCID: PMC8618301 DOI: 10.3390/ijms222212111] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 10/23/2021] [Accepted: 10/28/2021] [Indexed: 12/25/2022] Open
Abstract
The need to protect neural tissue from toxins or other substances is as old as neural tissue itself. Early recognition of this need has led to more than a century of investigation of the blood-brain barrier (BBB). Many aspects of this important neuroprotective barrier have now been well established, including its cellular architecture and barrier and transport functions. Unsurprisingly, most research has had a human orientation, using mammalian and other animal models to develop translational research findings. However, cell layers forming a barrier between vascular spaces and neural tissues are found broadly throughout the invertebrates as well as in all vertebrates. Unfortunately, previous scenarios for the evolution of the BBB typically adopt a classic, now discredited 'scala naturae' approach, which inaccurately describes a putative evolutionary progression of the mammalian BBB from simple invertebrates to mammals. In fact, BBB-like structures have evolved independently numerous times, complicating simplistic views of the evolution of the BBB as a linear process. Here, we review BBBs in their various forms in both invertebrates and vertebrates, with an emphasis on the function, evolution, and conditional relevance of popular animal models such as the fruit fly and the zebrafish to mammalian BBB research.
Collapse
Affiliation(s)
- Alicia D. Dunton
- Developmental Integrative Biology Group, Department of Biological Sciences, University of North Texas, Denton, TX 76203, USA; (T.G.); (W.B.)
- Correspondence:
| | - Torben Göpel
- Developmental Integrative Biology Group, Department of Biological Sciences, University of North Texas, Denton, TX 76203, USA; (T.G.); (W.B.)
| | - Dao H. Ho
- Department of Clinical Investigation, Tripler Army Medical Center, Honolulu, HI 96859, USA;
| | - Warren Burggren
- Developmental Integrative Biology Group, Department of Biological Sciences, University of North Texas, Denton, TX 76203, USA; (T.G.); (W.B.)
| |
Collapse
|
19
|
Drosophila ß Heavy-Spectrin is required in polarized ensheathing glia that form a diffusion-barrier around the neuropil. Nat Commun 2021; 12:6357. [PMID: 34737284 PMCID: PMC8569210 DOI: 10.1038/s41467-021-26462-x] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Accepted: 10/06/2021] [Indexed: 01/03/2023] Open
Abstract
In the central nervous system (CNS), functional tasks are often allocated to distinct compartments. This is also evident in the Drosophila CNS where synapses and dendrites are clustered in distinct neuropil regions. The neuropil is separated from neuronal cell bodies by ensheathing glia, which as we show using dye injection experiments, contribute to the formation of an internal diffusion barrier. We find that ensheathing glia are polarized with a basolateral plasma membrane rich in phosphatidylinositol-(3,4,5)-triphosphate (PIP3) and the Na+/K+-ATPase Nervana2 (Nrv2) that abuts an extracellular matrix formed at neuropil-cortex interface. The apical plasma membrane is facing the neuropil and is rich in phosphatidylinositol-(4,5)-bisphosphate (PIP2) that is supported by a sub-membranous ßHeavy-Spectrin cytoskeleton. ßHeavy-spectrin mutant larvae affect ensheathing glial cell polarity with delocalized PIP2 and Nrv2 and exhibit an abnormal locomotion which is similarly shown by ensheathing glia ablated larvae. Thus, polarized glia compartmentalizes the brain and is essential for proper nervous system function.
Collapse
|
20
|
Galea I. The blood-brain barrier in systemic infection and inflammation. Cell Mol Immunol 2021; 18:2489-2501. [PMID: 34594000 PMCID: PMC8481764 DOI: 10.1038/s41423-021-00757-x] [Citation(s) in RCA: 273] [Impact Index Per Article: 68.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 08/04/2021] [Indexed: 02/08/2023] Open
Abstract
The vascular blood-brain barrier is a highly regulated interface between the blood and brain. Its primary function is to protect central neurons while signaling the presence of systemic inflammation and infection to the brain to enable a protective sickness behavior response. With increasing degrees and duration of systemic inflammation, the vascular blood-brain barrier becomes more permeable to solutes, undergoes an increase in lymphocyte trafficking, and is infiltrated by innate immune cells; endothelial cell damage may occasionally occur. Perturbation of neuronal function results in the clinical features of encephalopathy. Here, the molecular and cellular anatomy of the vascular blood-brain barrier is reviewed, first in a healthy context and second in a systemic inflammatory context. Distinct from the molecular and cellular mediators of the blood-brain barrier's response to inflammation, several moderators influence the direction and magnitude at genetic, system, cellular and molecular levels. These include sex, genetic background, age, pre-existing brain pathology, systemic comorbidity, and gut dysbiosis. Further progress is required to define and measure mediators and moderators of the blood-brain barrier's response to systemic inflammation in order to explain the heterogeneity observed in animal and human studies.
Collapse
Affiliation(s)
- Ian Galea
- grid.5491.90000 0004 1936 9297Clinical Neurosciences, Clinical & Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, SO16 6YD UK
| |
Collapse
|
21
|
Winkler B, Funke D, Benmimoun B, Spéder P, Rey S, Logan MA, Klämbt C. Brain inflammation triggers macrophage invasion across the blood-brain barrier in Drosophila during pupal stages. SCIENCE ADVANCES 2021; 7:eabh0050. [PMID: 34705495 PMCID: PMC8550232 DOI: 10.1126/sciadv.abh0050] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/02/2023]
Abstract
The nervous system is shielded from circulating immune cells by the blood-brain barrier (BBB). During infections and autoimmune diseases, macrophages can enter the brain where they participate in pathogen elimination but can also cause tissue damage. Here, we establish a Drosophila model to study macrophage invasion into the inflamed brain. We show that the immune deficiency (Imd) pathway, but not the Toll pathway, is responsible for attraction and invasion of hemolymph-borne macrophages across the BBB during pupal stages. Macrophage recruitment is mediated by glial, but not neuronal, induction of the Imd pathway through expression of Pvf2. Within the brain, macrophages can phagocytose synaptic material and reduce locomotor abilities and longevity. Similarly, we show that central nervous system infection by group B Streptococcus elicits macrophage recruitment in an Imd-dependent manner. This suggests that evolutionarily conserved inflammatory responses require a delicate balance between beneficial and detrimental activities.
Collapse
Affiliation(s)
- Bente Winkler
- Institut für Neuro- und Verhaltensbiologie, Universität Münster, Badestr. 9, 48149 Münster, Germany
| | - Dominik Funke
- Institut für Neuro- und Verhaltensbiologie, Universität Münster, Badestr. 9, 48149 Münster, Germany
| | - Billel Benmimoun
- Brain Plasticity in response to the Environment, Institut Pasteur, UMR3738 CNRS, 75015 Paris, France
| | - Pauline Spéder
- Brain Plasticity in response to the Environment, Institut Pasteur, UMR3738 CNRS, 75015 Paris, France
| | - Simone Rey
- Institut für Neuro- und Verhaltensbiologie, Universität Münster, Badestr. 9, 48149 Münster, Germany
| | - Mary A. Logan
- Jungers Center for Neurosciences Research, Oregon Health and Science University, Portland, OR 97239, USA
| | - Christian Klämbt
- Institut für Neuro- und Verhaltensbiologie, Universität Münster, Badestr. 9, 48149 Münster, Germany
- Corresponding author.
| |
Collapse
|
22
|
The Serine Protease Homolog, Scarface, Is Sensitive to Nutrient Availability and Modulates the Development of the Drosophila Blood-Brain Barrier. J Neurosci 2021; 41:6430-6448. [PMID: 34210781 PMCID: PMC8318086 DOI: 10.1523/jneurosci.0452-20.2021] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2020] [Revised: 02/08/2021] [Accepted: 03/14/2021] [Indexed: 01/21/2023] Open
Abstract
The adaptable transcriptional response to changes in food availability not only ensures animal survival but also lets embryonic development progress. Interestingly, the CNS is preferentially protected from periods of malnutrition, a phenomenon known as “brain sparing.” However, the mechanisms that mediate this response remain poorly understood. To get a better understanding of this, we used Drosophila melanogaster as a model, analyzing the transcriptional response of neural stem cells (neuroblasts) and glia of the blood–brain barrier (BBB) from larvae of both sexes during nutrient restriction using targeted DamID. We found differentially expressed genes in both neuroblasts and glia of the BBB, although the effect of nutrient deficiency was primarily observed in the BBB. We characterized the function of a nutritional sensitive gene expressed in the BBB, the serine protease homolog, scarface (scaf). Scaf is expressed in subperineurial glia in the BBB in response to nutrition. Tissue-specific knockdown of scaf increases subperineurial glia endoreplication and proliferation of perineurial glia in the blood–brain barrier. Furthermore, neuroblast proliferation is diminished on scaf knockdown in subperineurial glia. Interestingly, reexpression of Scaf in subperineurial glia is able to enhance neuroblast proliferation and brain growth of animals in starvation. Finally, we show that loss of scaf in the blood–brain barrier increases sensitivity to drugs in adulthood, suggesting a physiological impairment. We propose that Scaf integrates the nutrient status to modulate the balance between neurogenesis and growth of the BBB, preserving the proper equilibrium between the size of the barrier and the brain. SIGNIFICANCE STATEMENT The Drosophila BBB separates the CNS from the open circulatory system. The BBB glia are not only acting as a physical segregation of tissues but participate in the regulation of the metabolism and neurogenesis during development. Here we analyze the transcriptional response of the BBB glia to nutrient deprivation during larval development, a condition in which protective mechanisms are switched on in the brain. Our findings show that the gene scarface reduces growth in the BBB while promoting the proliferation of neural stem, assuring the balanced growth of the larval brain. Thus, Scarface would link animal nutrition with brain development, coordinating neurogenesis with the growth of the BBB.
Collapse
|
23
|
Semyachkina-Glushkovskaya O, Mamedova A, Vinnik V, Klimova M, Saranceva E, Ageev V, Yu T, Zhu D, Penzel T, Kurths J. Brain Mechanisms of COVID-19-Sleep Disorders. Int J Mol Sci 2021; 22:6917. [PMID: 34203143 PMCID: PMC8268116 DOI: 10.3390/ijms22136917] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 06/15/2021] [Accepted: 06/17/2021] [Indexed: 02/06/2023] Open
Abstract
2020 and 2021 have been unprecedented years due to the rapid spread of the modified severe acute respiratory syndrome coronavirus around the world. The coronavirus disease 2019 (COVID-19) causes atypical infiltrated pneumonia with many neurological symptoms, and major sleep changes. The exposure of people to stress, such as social confinement and changes in daily routines, is accompanied by various sleep disturbances, known as 'coronasomnia' phenomenon. Sleep disorders induce neuroinflammation, which promotes the blood-brain barrier (BBB) disruption and entry of antigens and inflammatory factors into the brain. Here, we review findings and trends in sleep research in 2020-2021, demonstrating how COVID-19 and sleep disorders can induce BBB leakage via neuroinflammation, which might contribute to the 'coronasomnia' phenomenon. The new studies suggest that the control of sleep hygiene and quality should be incorporated into the rehabilitation of COVID-19 patients. We also discuss perspective strategies for the prevention of COVID-19-related BBB disorders. We demonstrate that sleep might be a novel biomarker of BBB leakage, and the analysis of sleep EEG patterns can be a breakthrough non-invasive technology for diagnosis of the COVID-19-caused BBB disruption.
Collapse
Affiliation(s)
- Oxana Semyachkina-Glushkovskaya
- Institute of Physics, Humboldt University, Newtonstrasse 15, 12489 Berlin, Germany;
- Department of Biology, Saratov State University, Atrakhanskaya Str. 83, 410012 Saratov, Russia; (A.M.); (V.V.); (M.K.); (E.S.); (V.A.)
| | - Aysel Mamedova
- Department of Biology, Saratov State University, Atrakhanskaya Str. 83, 410012 Saratov, Russia; (A.M.); (V.V.); (M.K.); (E.S.); (V.A.)
| | - Valeria Vinnik
- Department of Biology, Saratov State University, Atrakhanskaya Str. 83, 410012 Saratov, Russia; (A.M.); (V.V.); (M.K.); (E.S.); (V.A.)
| | - Maria Klimova
- Department of Biology, Saratov State University, Atrakhanskaya Str. 83, 410012 Saratov, Russia; (A.M.); (V.V.); (M.K.); (E.S.); (V.A.)
| | - Elena Saranceva
- Department of Biology, Saratov State University, Atrakhanskaya Str. 83, 410012 Saratov, Russia; (A.M.); (V.V.); (M.K.); (E.S.); (V.A.)
| | - Vasily Ageev
- Department of Biology, Saratov State University, Atrakhanskaya Str. 83, 410012 Saratov, Russia; (A.M.); (V.V.); (M.K.); (E.S.); (V.A.)
| | - Tingting Yu
- Wuhan National Laboratory for Optoelectronics, Britton Chance Center for Biomedical Photonics, Huazhong University of Science and Technology, Wuhan 430074, China; (T.Y.); (D.Z.)
- MoE Key Laboratory for Biomedical Photonics, Collaborative Innovation Center for Biomedical Engineering, School of Engineering Sciences, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Dan Zhu
- Wuhan National Laboratory for Optoelectronics, Britton Chance Center for Biomedical Photonics, Huazhong University of Science and Technology, Wuhan 430074, China; (T.Y.); (D.Z.)
- MoE Key Laboratory for Biomedical Photonics, Collaborative Innovation Center for Biomedical Engineering, School of Engineering Sciences, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Thomas Penzel
- Department of Biology, Saratov State University, Atrakhanskaya Str. 83, 410012 Saratov, Russia; (A.M.); (V.V.); (M.K.); (E.S.); (V.A.)
- Sleep Medicine Center, Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - Jürgen Kurths
- Institute of Physics, Humboldt University, Newtonstrasse 15, 12489 Berlin, Germany;
- Department of Biology, Saratov State University, Atrakhanskaya Str. 83, 410012 Saratov, Russia; (A.M.); (V.V.); (M.K.); (E.S.); (V.A.)
- Potsdam Institute for Climate Impact Research, Telegrafenberg A31, 14473 Potsdam, Germany
| |
Collapse
|
24
|
Vatansever S, Schlessinger A, Wacker D, Kaniskan HÜ, Jin J, Zhou M, Zhang B. Artificial intelligence and machine learning-aided drug discovery in central nervous system diseases: State-of-the-arts and future directions. Med Res Rev 2021; 41:1427-1473. [PMID: 33295676 PMCID: PMC8043990 DOI: 10.1002/med.21764] [Citation(s) in RCA: 131] [Impact Index Per Article: 32.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 10/30/2020] [Accepted: 11/20/2020] [Indexed: 01/11/2023]
Abstract
Neurological disorders significantly outnumber diseases in other therapeutic areas. However, developing drugs for central nervous system (CNS) disorders remains the most challenging area in drug discovery, accompanied with the long timelines and high attrition rates. With the rapid growth of biomedical data enabled by advanced experimental technologies, artificial intelligence (AI) and machine learning (ML) have emerged as an indispensable tool to draw meaningful insights and improve decision making in drug discovery. Thanks to the advancements in AI and ML algorithms, now the AI/ML-driven solutions have an unprecedented potential to accelerate the process of CNS drug discovery with better success rate. In this review, we comprehensively summarize AI/ML-powered pharmaceutical discovery efforts and their implementations in the CNS area. After introducing the AI/ML models as well as the conceptualization and data preparation, we outline the applications of AI/ML technologies to several key procedures in drug discovery, including target identification, compound screening, hit/lead generation and optimization, drug response and synergy prediction, de novo drug design, and drug repurposing. We review the current state-of-the-art of AI/ML-guided CNS drug discovery, focusing on blood-brain barrier permeability prediction and implementation into therapeutic discovery for neurological diseases. Finally, we discuss the major challenges and limitations of current approaches and possible future directions that may provide resolutions to these difficulties.
Collapse
Affiliation(s)
- Sezen Vatansever
- Department of Genetics and Genomic SciencesIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
- Mount Sinai Center for Transformative Disease ModelingIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
- Icahn Institute for Data Science and Genomic TechnologyIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Avner Schlessinger
- Department of Pharmacological SciencesIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
- Mount Sinai Center for Therapeutics DiscoveryIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Daniel Wacker
- Department of Pharmacological SciencesIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
- Mount Sinai Center for Therapeutics DiscoveryIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
- Department of NeuroscienceIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - H. Ümit Kaniskan
- Department of Pharmacological SciencesIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
- Mount Sinai Center for Therapeutics DiscoveryIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
- Department of Oncological Sciences, Tisch Cancer InstituteIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Jian Jin
- Department of Pharmacological SciencesIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
- Mount Sinai Center for Therapeutics DiscoveryIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
- Department of Oncological Sciences, Tisch Cancer InstituteIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Ming‐Ming Zhou
- Department of Pharmacological SciencesIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
- Department of Oncological Sciences, Tisch Cancer InstituteIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Bin Zhang
- Department of Genetics and Genomic SciencesIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
- Mount Sinai Center for Transformative Disease ModelingIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
- Icahn Institute for Data Science and Genomic TechnologyIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
- Department of Pharmacological SciencesIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| |
Collapse
|
25
|
Nicewicz Ł, Nicewicz AW, Kafel A, Nakonieczny M. Set of stress biomarkers as a practical tool in the assessment of multistress effect using honeybees from urban and rural areas as a model organism: a pilot study. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2021; 28:9084-9096. [PMID: 33128148 PMCID: PMC7884360 DOI: 10.1007/s11356-020-11338-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 10/20/2020] [Indexed: 06/11/2023]
Abstract
A decrease among honey bee populations (Apis mellifera) in the traditional apiaries has been observed in recent years. In light of this negative phenomenon, urban beekeeping seems to be an appropriate alternative solution for the bee population in reducing the toxic effects of a large number of pesticides that are commonly used in agricultural ecosystems. Despite the rapid development of urban beekeeping, there is little information regarding the different aspects of the defense effectiveness of bees from the urban and rural areas. The study was aimed to show whether honey bees from these two locations differ in the level of the valuable biomarkers of stress exposure helpful in establishing which bees, from urban or rural areas, are under greater environmental pressure. For this purpose, foragers from an urban rooftop apiary and a traditional rural apiary were collected. The chosen biomarkers were measured in various tissues of bees. The activity of glutathione S-transferase and acetylcholinesterase, the level of total antioxidant capacity, heat shock protein 70 (Hsp70), and defensin were selected for the analyses. In our opinion, the Hsp70 and defensin levels seemed to be important in the indication of urban multistress factors. The higher level of heat shock proteins and defensins in tissues/organs of bees from the urban apiary-in the gut (an increase, respectively, 92% and 7.3%) and fat body (an increase, respectively, 130% and 7.8%), known as targets of environmental toxins, pointed out the urban environment as highly stressful at both the individual and colony levels. In turn, high total antioxidant capacity was measured in the guts of honey bees from rural area (an increase 107%). Such a situation suggests a different mechanism of defense and specificity of rural and urban environmental stressors and also honey bees foraging activity.
Collapse
Affiliation(s)
- Łukasz Nicewicz
- Research Team of Animal Physiology and Ecotoxicology, Faculty of Natural Sciences, Institute of Biology, Biotechnology and Environmental Protection, University of Silesia, Bankowa 9, 40-007, Katowice, PL, Poland.
| | - Agata W Nicewicz
- Research Team of Animal Physiology and Ecotoxicology, Faculty of Natural Sciences, Institute of Biology, Biotechnology and Environmental Protection, University of Silesia, Bankowa 9, 40-007, Katowice, PL, Poland
| | - Alina Kafel
- Research Team of Animal Physiology and Ecotoxicology, Faculty of Natural Sciences, Institute of Biology, Biotechnology and Environmental Protection, University of Silesia, Bankowa 9, 40-007, Katowice, PL, Poland
| | - Mirosław Nakonieczny
- Research Team of Animal Physiology and Ecotoxicology, Faculty of Natural Sciences, Institute of Biology, Biotechnology and Environmental Protection, University of Silesia, Bankowa 9, 40-007, Katowice, PL, Poland
| |
Collapse
|
26
|
Benmimoun B, Papastefanaki F, Périchon B, Segklia K, Roby N, Miriagou V, Schmitt C, Dramsi S, Matsas R, Spéder P. An original infection model identifies host lipoprotein import as a route for blood-brain barrier crossing. Nat Commun 2020; 11:6106. [PMID: 33257684 PMCID: PMC7704634 DOI: 10.1038/s41467-020-19826-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Accepted: 11/02/2020] [Indexed: 02/06/2023] Open
Abstract
Pathogens able to cross the blood-brain barrier (BBB) induce long-term neurological sequelae and death. Understanding how neurotropic pathogens bypass this strong physiological barrier is a prerequisite to devise therapeutic strategies. Here we propose an innovative model of infection in the developing Drosophila brain, combining whole brain explants with in vivo systemic infection. We find that several mammalian pathogens are able to cross the Drosophila BBB, including Group B Streptococcus (GBS). Amongst GBS surface components, lipoproteins, and in particular the B leucine-rich Blr, are important for BBB crossing and virulence in Drosophila. Further, we identify (V)LDL receptor LpR2, expressed in the BBB, as a host receptor for Blr, allowing GBS translocation through endocytosis. Finally, we show that Blr is required for BBB crossing and pathogenicity in a murine model of infection. Our results demonstrate the potential of Drosophila for studying BBB crossing by pathogens and identify a new mechanism by which pathogens exploit the machinery of host barriers to generate brain infection.
Collapse
Affiliation(s)
- Billel Benmimoun
- Institut Pasteur, Brain Plasticity in Response to the Environment, CNRS, UMR3738, Paris, France
| | - Florentia Papastefanaki
- Laboratory of Cellular and Molecular Neurobiology-Stem Cells, Department of Neurobiology, Hellenic Pasteur Institute, Athens, Greece
| | - Bruno Périchon
- Unité de Biologie des Bactéries Pathogènes à Gram-positif, Institut Pasteur, CNRS, UMR 2001, Paris, France
| | - Katerina Segklia
- Laboratory of Cellular and Molecular Neurobiology-Stem Cells, Department of Neurobiology, Hellenic Pasteur Institute, Athens, Greece
| | - Nicolas Roby
- Institut Pasteur, Brain Plasticity in Response to the Environment, CNRS, UMR3738, Paris, France
| | - Vivi Miriagou
- Laboratory of Bacteriology, Department of Microbiology, Hellenic Pasteur Institute, Athens, Greece
| | - Christine Schmitt
- Ultrastructure UTechS Ultrastructural Bioimaging Platform, Institut Pasteur, Paris, France
| | - Shaynoor Dramsi
- Unité de Biologie des Bactéries Pathogènes à Gram-positif, Institut Pasteur, CNRS, UMR 2001, Paris, France
| | - Rebecca Matsas
- Laboratory of Cellular and Molecular Neurobiology-Stem Cells, Department of Neurobiology, Hellenic Pasteur Institute, Athens, Greece
| | - Pauline Spéder
- Institut Pasteur, Brain Plasticity in Response to the Environment, CNRS, UMR3738, Paris, France.
| |
Collapse
|
27
|
Profaci CP, Munji RN, Pulido RS, Daneman R. The blood-brain barrier in health and disease: Important unanswered questions. J Exp Med 2020; 217:151582. [PMID: 32211826 PMCID: PMC7144528 DOI: 10.1084/jem.20190062] [Citation(s) in RCA: 377] [Impact Index Per Article: 75.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Revised: 09/21/2019] [Accepted: 11/21/2019] [Indexed: 12/11/2022] Open
Abstract
The blood vessels vascularizing the central nervous system exhibit a series of distinct properties that tightly control the movement of ions, molecules, and cells between the blood and the parenchyma. This "blood-brain barrier" is initiated during angiogenesis via signals from the surrounding neural environment, and its integrity remains vital for homeostasis and neural protection throughout life. Blood-brain barrier dysfunction contributes to pathology in a range of neurological conditions including multiple sclerosis, stroke, and epilepsy, and has also been implicated in neurodegenerative diseases such as Alzheimer's disease. This review will discuss current knowledge and key unanswered questions regarding the blood-brain barrier in health and disease.
Collapse
Affiliation(s)
- Caterina P Profaci
- Department of Neurosciences, University of California, San Diego, San Diego, CA.,Department of Pharmacology, University of California, San Diego, San Diego, CA
| | - Roeben N Munji
- Department of Neurosciences, University of California, San Diego, San Diego, CA.,Department of Pharmacology, University of California, San Diego, San Diego, CA
| | - Robert S Pulido
- Department of Neurosciences, University of California, San Diego, San Diego, CA.,Department of Pharmacology, University of California, San Diego, San Diego, CA
| | - Richard Daneman
- Department of Neurosciences, University of California, San Diego, San Diego, CA.,Department of Pharmacology, University of California, San Diego, San Diego, CA
| |
Collapse
|
28
|
Neuronal Activity Regulates Blood-Brain Barrier Efflux Transport through Endothelial Circadian Genes. Neuron 2020; 108:937-952.e7. [PMID: 32979312 DOI: 10.1016/j.neuron.2020.09.002] [Citation(s) in RCA: 91] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Revised: 07/22/2020] [Accepted: 08/31/2020] [Indexed: 01/11/2023]
Abstract
The blood vessels in the central nervous system (CNS) have a series of unique properties, termed the blood-brain barrier (BBB), which stringently regulate the entry of molecules into the brain, thus maintaining proper brain homeostasis. We sought to understand whether neuronal activity could regulate BBB properties. Using both chemogenetics and a volitional behavior paradigm, we identified a core set of brain endothelial genes whose expression is regulated by neuronal activity. In particular, neuronal activity regulates BBB efflux transporter expression and function, which is critical for excluding many small lipophilic molecules from the brain parenchyma. Furthermore, we found that neuronal activity regulates the expression of circadian clock genes within brain endothelial cells, which in turn mediate the activity-dependent control of BBB efflux transport. These results have important clinical implications for CNS drug delivery and clearance of CNS waste products, including Aβ, and for understanding how neuronal activity can modulate diurnal processes.
Collapse
|
29
|
Kaiser K, Bryja V. Choroid Plexus: The Orchestrator of Long-Range Signalling Within the CNS. Int J Mol Sci 2020; 21:E4760. [PMID: 32635478 PMCID: PMC7369786 DOI: 10.3390/ijms21134760] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2020] [Revised: 06/26/2020] [Accepted: 07/02/2020] [Indexed: 01/24/2023] Open
Abstract
Cerebrospinal fluid (CSF) is the liquid that fills the brain ventricles. CSF represents not only a mechanical brain protection but also a rich source of signalling factors modulating diverse processes during brain development and adulthood. The choroid plexus (CP) is a major source of CSF and as such it has recently emerged as an important mediator of extracellular signalling within the brain. Growing interest in the CP revealed its capacity to release a broad variety of bioactive molecules that, via CSF, regulate processes across the whole central nervous system (CNS). Moreover, CP has been also recognized as a sensor, responding to altered composition of CSF associated with changes in the patterns of CNS activity. In this review, we summarize the recent advances in our understanding of the CP as a signalling centre that mediates long-range communication in the CNS. By providing a detailed account of the CP secretory repertoire, we describe how the CP contributes to the regulation of the extracellular environment-in the context of both the embryonal as well as the adult CNS. We highlight the role of the CP as an important regulator of CNS function that acts via CSF-mediated signalling. Further studies of CP-CSF signalling hold the potential to provide key insights into the biology of the CNS, with implications for better understanding and treatment of neuropathological conditions.
Collapse
Affiliation(s)
- Karol Kaiser
- Department of Experimental Biology, Faculty of Science, Masaryk University, 625 00 Brno, Czech Republic
| | - Vitezslav Bryja
- Department of Experimental Biology, Faculty of Science, Masaryk University, 625 00 Brno, Czech Republic
| |
Collapse
|
30
|
A Cell-Based High-Throughput Screening Identified Two Compounds that Enhance PINK1-Parkin Signaling. iScience 2020; 23:101048. [PMID: 32335362 PMCID: PMC7183160 DOI: 10.1016/j.isci.2020.101048] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2019] [Revised: 03/14/2020] [Accepted: 04/04/2020] [Indexed: 12/21/2022] Open
Abstract
Early-onset Parkinson's disease-associated PINK1-Parkin signaling maintains mitochondrial health. Therapeutic approaches for enhancing PINK1-Parkin signaling present a potential strategy for treating various diseases caused by mitochondrial dysfunction. We report two chemical enhancers of PINK1-Parkin signaling, identified using a robust cell-based high-throughput screening system. These small molecules, T0466 and T0467, activate Parkin mitochondrial translocation in dopaminergic neurons and myoblasts at low doses that do not induce mitochondrial accumulation of PINK1. Moreover, both compounds reduce unfolded mitochondrial protein levels, presumably through enhanced PINK1-Parkin signaling. These molecules also mitigate the locomotion defect, reduced ATP production, and disturbed mitochondrial Ca2+ response in the muscles along with the mitochondrial aggregation in dopaminergic neurons through reduced PINK1 activity in Drosophila. Our results suggested that T0466 and T0467 may hold promise as therapeutic reagents in Parkinson's disease and related disorders.
Collapse
|
31
|
Bittern J, Pogodalla N, Ohm H, Brüser L, Kottmeier R, Schirmeier S, Klämbt C. Neuron-glia interaction in the Drosophila nervous system. Dev Neurobiol 2020; 81:438-452. [PMID: 32096904 DOI: 10.1002/dneu.22737] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Revised: 02/11/2020] [Accepted: 02/24/2020] [Indexed: 12/14/2022]
Abstract
Animals are able to move and react in manifold ways to external stimuli. Thus, environmental stimuli need to be detected, information must be processed, and, finally, an output decision must be transmitted to the musculature to get the animal moving. All these processes depend on the nervous system which comprises an intricate neuronal network and many glial cells. Glial cells have an equally important contribution in nervous system function as their neuronal counterpart. Manifold roles are attributed to glia ranging from controlling neuronal cell number and axonal pathfinding to regulation of synapse formation, function, and plasticity. Glial cells metabolically support neurons and contribute to the blood-brain barrier. All of the aforementioned aspects require extensive cell-cell interactions between neurons and glial cells. Not surprisingly, many of these processes are found in all phyla executed by evolutionarily conserved molecules. Here, we review the recent advance in understanding neuron-glia interaction in Drosophila melanogaster to suggest that work in simple model organisms will shed light on the function of mammalian glial cells, too.
Collapse
Affiliation(s)
- Jonas Bittern
- Institut für Neuro- und Verhaltensbiologie, Universität Münster, Münster, Germany
| | - Nicole Pogodalla
- Institut für Neuro- und Verhaltensbiologie, Universität Münster, Münster, Germany
| | - Henrike Ohm
- Institut für Neuro- und Verhaltensbiologie, Universität Münster, Münster, Germany
| | - Lena Brüser
- Institut für Neuro- und Verhaltensbiologie, Universität Münster, Münster, Germany
| | - Rita Kottmeier
- Institut für Neuro- und Verhaltensbiologie, Universität Münster, Münster, Germany
| | - Stefanie Schirmeier
- Institut für Neuro- und Verhaltensbiologie, Universität Münster, Münster, Germany
| | - Christian Klämbt
- Institut für Neuro- und Verhaltensbiologie, Universität Münster, Münster, Germany
| |
Collapse
|
32
|
Fame RM, Cortés-Campos C, Sive HL. Brain Ventricular System and Cerebrospinal Fluid Development and Function: Light at the End of the Tube: A Primer with Latest Insights. Bioessays 2020; 42:e1900186. [PMID: 32078177 DOI: 10.1002/bies.201900186] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Revised: 01/02/2020] [Indexed: 12/12/2022]
Abstract
The brain ventricular system is a series of connected cavities, filled with cerebrospinal fluid (CSF), that forms within the vertebrate central nervous system (CNS). The hollow neural tube is a hallmark of the chordate CNS, and a closed neural tube is essential for normal development. Development and function of the ventricular system is examined, emphasizing three interdigitating components that form a functional system: ventricle walls, CSF fluid properties, and activity of CSF constituent factors. The cellular lining of the ventricle both can produce and is responsive to CSF. Fluid properties and conserved CSF components contribute to normal CNS development. Anomalies of the CSF/ventricular system serve as diagnostics and may cause CNS disorders, further highlighting their importance. This review focuses on the evolution and development of the brain ventricular system, associated function, and connected pathologies. It is geared as an introduction for scholars with little background in the field.
Collapse
Affiliation(s)
- Ryann M Fame
- Whitehead Institute for Biomedical Research, Cambridge, MA, 02142, USA
| | | | - Hazel L Sive
- Whitehead Institute for Biomedical Research, Cambridge, MA, 02142, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| |
Collapse
|
33
|
Robertson RM, Dawson-Scully KD, Andrew RD. Neural shutdown under stress: an evolutionary perspective on spreading depolarization. J Neurophysiol 2020; 123:885-895. [PMID: 32023142 PMCID: PMC7099469 DOI: 10.1152/jn.00724.2019] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 01/13/2020] [Accepted: 01/14/2020] [Indexed: 11/22/2022] Open
Abstract
Neural function depends on maintaining cellular membrane potentials as the basis for electrical signaling. Yet, in mammals and insects, neuronal and glial membrane potentials can reversibly depolarize to zero, shutting down neural function by the process of spreading depolarization (SD) that collapses the ion gradients across membranes. SD is not evident in all metazoan taxa with centralized nervous systems. We consider the occurrence and similarities of SD in different animals and suggest that it is an emergent property of nervous systems that have evolved to control complex behaviors requiring energetically expensive, rapid information processing in a tightly regulated extracellular environment. Whether SD is beneficial or not in mammals remains an open question. However, in insects, it is associated with the response to harsh environments and may provide an energetic advantage that improves the chances of survival. The remarkable similarity of SD in diverse taxa supports a model systems approach to understanding the mechanistic underpinning of human neuropathology associated with migraine, stroke, and traumatic brain injury.
Collapse
Affiliation(s)
- R Meldrum Robertson
- Department of Biology and Centre for Neuroscience Studies, Queen's University, Kingston, Ontario, Canada
| | - Ken D Dawson-Scully
- Department of Biological Sciences, Florida Atlantic University, Boca Raton, Florida
| | - R David Andrew
- Department of Biomedical and Molecular Sciences and Centre for Neuroscience Studies, Queen's University, Kingston, Ontario, Canada
| |
Collapse
|
34
|
Okamoto N, Yamanaka N. Steroid Hormone Entry into the Brain Requires a Membrane Transporter in Drosophila. Curr Biol 2020; 30:359-366.e3. [PMID: 31928869 DOI: 10.1016/j.cub.2019.11.085] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Revised: 11/14/2019] [Accepted: 11/28/2019] [Indexed: 02/08/2023]
Abstract
Steroid hormones control various aspects of brain development and behavior in metazoans, but how they enter the central nervous system (CNS) through the blood-brain barrier (BBB) remains poorly understood. It is generally believed that steroid hormones freely diffuse through the plasma membrane of the BBB cells to reach the brain [1], because of the predominant "simple diffusion" model of steroid hormone transport across cell membranes. Recently, however, we challenged the simple diffusion model by showing that a Drosophila organic anion-transporting polypeptide (OATP), which we named Ecdysone Importer (EcI), is required for cellular uptake of the primary insect steroid hormone ecdysone [2]. As ecdysone is first secreted into the hemolymph before reaching the CNS [3], our finding raised the question of how ecdysone enters the CNS through the BBB to exert its diverse role in Drosophila brain development. Here, we demonstrate in the Drosophila BBB that EcI is indispensable for ecdysone entry into the CNS to facilitate brain development. EcI is highly expressed in surface glial cells that form the BBB, and EcI knockdown in the BBB suppresses ecdysone signaling within the CNS and blocks ecdysone-mediated neuronal events during development. In an ex vivo culture system, the CNS requires EcI in the BBB to incorporate ecdysone from the culture medium. Our results suggest a transporter-mediated mechanism of steroid hormone entry into the CNS, which may provide important implications in controlling brain development and behavior by regulating steroid hormone permeability across the BBB.
Collapse
Affiliation(s)
- Naoki Okamoto
- Department of Entomology, Institute for Integrative Genome Biology, University of California, Riverside, 900 University Ave., Riverside, CA 92521, USA
| | - Naoki Yamanaka
- Department of Entomology, Institute for Integrative Genome Biology, University of California, Riverside, 900 University Ave., Riverside, CA 92521, USA.
| |
Collapse
|
35
|
Del Giudice M. Invisible Designers: Brain Evolution Through the Lens of Parasite Manipulation. QUARTERLY REVIEW OF BIOLOGY 2019. [DOI: 10.1086/705038] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
|
36
|
Effect of acute and chronic aldosterone exposure on the retinal pigment epithelium-choroid complex in rodents. Exp Eye Res 2019; 187:107747. [PMID: 31394103 DOI: 10.1016/j.exer.2019.107747] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Revised: 07/17/2019] [Accepted: 07/31/2019] [Indexed: 11/23/2022]
Abstract
Preclinical and clinical evidences show that aldosterone and/or mineralocorticoid receptor (MR) over-activation by glucocorticoids can be deleterious to the retina and to the retinal pigment epithelium (RPE)-choroid complex. However, the exact molecular mechanisms driving these effects remain poorly understood and pathological consequences of chronic exposure of the retina and RPE/choroid to aldosterone have not been completely explored. We aimed to decipher the transcriptomic regulation in the RPE-choroid complex in rats in response to acute intraocular aldosterone injection and to explore the consequences of systemic chronic aldosterone exposure on the morphology and the gene regulation in RPE/choroid in mice. High dose of aldosterone (100 nM) was intravitreously injected in Lewis rat eyes in order to yield an aldosterone dose able to induce a molecular response at the apical side of the RPE-choroid complex. The posterior segment morphology was evaluated in vivo using optical coherence tomography (OCT) before and 24 h after aldosterone injection. Rat RPE-choroid complexes were used for RNA sequencing and analysis. Uninephrectomy/aldosterone/salt (NAS) model was created in wild-type C57BL/6 mice. After 6 weeks, histology of mouse posterior segments were observed ex vivo. Gene expression in the RPE-choroid complex was analyzed using quantitative PCR. Acute intravitreous injection of aldosterone induced posterior segment inflammation observed on OCT. RNA sequencing of rat RPE-choroid complexes revealed up-regulation of pathways involved in inflammation, oxidative stress and RNA procession, and down-regulation of genes involved in synaptic activity, muscle contraction, cytoskeleton, cell junction and transporters. Chronic aldosterone/salt exposure in NAS model induces retinal edema, choroidal vasodilation and RPE cell dysfunction and migration. Quantitative PCR showed deregulation of genes involved in inflammatory response, oxidative stress, particularly the NOX pathway, angiogenesis and cell contractility. Both rodent models share some common phenotypes and molecular regulations in the RPE-choroid complex that could contribute to pachychoroid epitheliopathy in humans. The difference in inflammatory status relies on different intraocular or systemic route of aldosterone administration and on the different doses of aldosterone exposed to the RPE-choroid complex.
Collapse
|
37
|
Lee KM, Mathies LD, Grotewiel M. Alcohol sedation in adult Drosophila is regulated by Cysteine proteinase-1 in cortex glia. Commun Biol 2019; 2:252. [PMID: 31286069 PMCID: PMC6610072 DOI: 10.1038/s42003-019-0492-5] [Citation(s) in RCA: 10] [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: 02/19/2018] [Accepted: 05/30/2019] [Indexed: 02/06/2023] Open
Abstract
Although numerous studies have demonstrated that neuronal mechanisms regulate alcohol-related behaviors, very few have investigated the direct role of glia in behavioral responses to alcohol. The results described here begin to fill this gap in the alcohol behavior and gliobiology fields. Since Drosophila exhibit conserved behavioral responses to alcohol and their CNS glia are similar to mammalian CNS glia, we used Drosophila to begin exploring the role of glia in alcohol behavior. We found that knockdown of Cysteine proteinase-1 (Cp1) in glia increased Drosophila alcohol sedation and that this effect was specific to cortex glia and adulthood. These data implicate Cp1 and cortex glia in alcohol-related behaviors. Cortex glia are functionally homologous to mammalian astrocytes and Cp1 is orthologous to mammalian Cathepsin L. Our studies raise the possibility that cathepsins may influence behavioral responses to alcohol in mammals via roles in astrocytes.
Collapse
Affiliation(s)
- Kristen M. Lee
- Neuroscience Graduate Program, Virginia Commonwealth University, Richmond, VA 23298 USA
| | - Laura D. Mathies
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA 23298 USA
- Virginia Commonwealth University Alcohol Research Center, Virginia Commonwealth University, Richmond, VA 23298 USA
| | - Mike Grotewiel
- Neuroscience Graduate Program, Virginia Commonwealth University, Richmond, VA 23298 USA
- Virginia Commonwealth University Alcohol Research Center, Virginia Commonwealth University, Richmond, VA 23298 USA
- Department of Human and Molecular Genetics, Virginia Commonwealth University, Richmond, VA 23298 USA
| |
Collapse
|
38
|
Cuddapah VA, Zhang SL, Sehgal A. Regulation of the Blood-Brain Barrier by Circadian Rhythms and Sleep. Trends Neurosci 2019; 42:500-510. [PMID: 31253251 PMCID: PMC6602072 DOI: 10.1016/j.tins.2019.05.001] [Citation(s) in RCA: 121] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2019] [Revised: 04/27/2019] [Accepted: 05/01/2019] [Indexed: 01/09/2023]
Abstract
The blood-brain barrier (BBB) is an evolutionarily conserved, structural, and functional separation between circulating blood and the central nervous system (CNS). By controlling permeability into and out of the nervous system, the BBB has a critical role in the precise regulation of neural processes. Here, we review recent studies demonstrating that permeability at the BBB is dynamically controlled by circadian rhythms and sleep. An endogenous circadian rhythm in the BBB controls transporter function, regulating permeability across the BBB. In addition, sleep promotes the clearance of metabolites along the BBB, as well as endocytosis across the BBB. Finally, we highlight the implications of this regulation for diseases, including epilepsy.
Collapse
Affiliation(s)
- Vishnu Anand Cuddapah
- Division of Neurology, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; Center for Sleep and Circadian Neurobiology, Chronobiology Program, and Howard Hughes Medical Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Shirley L Zhang
- Center for Sleep and Circadian Neurobiology, Chronobiology Program, and Howard Hughes Medical Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Amita Sehgal
- Center for Sleep and Circadian Neurobiology, Chronobiology Program, and Howard Hughes Medical Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
39
|
Guerriero G, Parisi C, Abdel-Gawad FK, Hentati O, D'Errico G. Seasonal and pharmaceutical-induced changes in selenoprotein glutathione peroxidase 4 activity in the reproductive dynamics of the soil biosentinel Podarcis sicula (Chordata: Reptilia). Mol Reprod Dev 2019; 86:1378-1387. [PMID: 30957323 DOI: 10.1002/mrd.23143] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2019] [Revised: 03/11/2019] [Accepted: 03/14/2019] [Indexed: 01/08/2023]
Abstract
There is rising concern for the interaction of environmental contaminants with brain transcriptome and the potential effect on reproductive processes. The present study sought to determine selenoprotein glutathione peroxidase 4 (gpx4) transcriptional activity in the brain and testis of the soil biosentinel, Podarcis sicula, through the main phases of the reproductive cycle and whether pharmaceuticals exert an endocrine disruption. Based on gpx4 cloned amminoacids sequence (GenBank AEX09236.1.), we used a bioinformatic approach to assess the structural role. Specifically, we detected seasonally the reactive oxygen species (ROS) level using electron spin resonance spectroscopy and gpx4 transcriptional activity using quantitative real-time polymerase chain reaction. In addition, the impact of pharmaceuticals was assessed after 21-days of treatment with ICI 182,780 and human chorionic gonadotropin administration in mating and winter stasis, respectively. Bioinformatic data shows the gpx4 proteic activity and a phylogenetic profile. ROS contents in lizard brain are significantly less than in testis and display higher levels after treatments. Brain gpx4 expression gives statistically significant seasonal differences, opposite trends in testis and altered expression in both tissues, with evidence of testis morphological and DNA disruption. Taken together, these results provide direct evidence that gpx4 in P. sicula plays a seasonal regulatory role and may be a reliable biomarker for reproductive health toxicity screening.
Collapse
Affiliation(s)
- Giulia Guerriero
- Department of Biology, University of Naples Federico II, Complesso Universitario di Monte S. Angelo, Naples, Italy.,Interdepartmental Research Center for Environment, University of Naples Federico II, Naples, Italy
| | - Costantino Parisi
- Department of Biology, University of Naples Federico II, Complesso Universitario di Monte S. Angelo, Naples, Italy
| | - Fagr Kh Abdel-Gawad
- Department of Biology, University of Naples Federico II, Complesso Universitario di Monte S. Angelo, Naples, Italy.,Department of Water Pollution, Centre of Excellence for Advanced Sciences (CEAS), National Research Centre, Giza, Egypt
| | - Olfa Hentati
- Department of Biotechnology and Health, Institut Supérieur de Biotechnologie de Sfax, Sfax, Tunisia
| | - Gerardino D'Errico
- Department of Chemical Sciences, University of Naples Federico II, Complesso Universitario di Monte S. Angelo, Naples, Italy
| |
Collapse
|
40
|
Wu VM, Huynh E, Tang S, Uskoković V. Brain and bone cancer targeting by a ferrofluid composed of superparamagnetic iron-oxide/silica/carbon nanoparticles (earthicles). Acta Biomater 2019; 88:422-447. [PMID: 30711662 DOI: 10.1016/j.actbio.2019.01.064] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Revised: 01/11/2019] [Accepted: 01/30/2019] [Indexed: 01/02/2023]
Abstract
Despite the advances in molecularly targeted therapies, delivery across the blood-brain barrier (BBB) and the targeting of brain tumors remains a challenge. Like brain, bone is a common site of metastasis and requires therapies capable of discerning the tumor from its healthy cellular milieu. To tackle these challenges, we made a variation on the previously proposed concept of the earthicle and fabricated an aqueous, surfactant-free ferrofluid containing superparamagnetic iron oxide nanoparticles (SPIONs) coated with silicate mesolayers and carbon shells, having 13 nm in size on average. Nanoparticles were synthesized hydrothermally and characterized using a range of spectroscopic, diffractometric, hydrodynamic and electron microscopy techniques. The double coating on SPIONs affected a number of physicochemical and biological properties, including colloidal stability and cancer targeting efficacy. Nanoparticles decreased the viability of glioblastoma and osteosarcoma cells and tumors more than that of their primary and non-transformed analogues. They showed a greater preference for cancer cells because of a higher rate of uptake by these cells and a pronounced adherence to cancer cell membrane. Even in an ultralow alternate magnetic field, nanoparticles generated sufficient heat to cause tumor death. Nanoparticles in MDCK-MDR1 BBB model caused mislocalization of claudin-1 at the tight junctions, underexpression of ZO-1 and no effect on occludin-1 and transepithelial resistance. Nanoparticles were detected in the basolateral compartments and examination of LAMP1 demonstrated that nanoparticles escaped the lysosome, traversed the BBB transcellularly and localized to the optic lobes of the third instar larval brains of Drosophila melanogaster. The passage was noninvasive and caused no adverse systemic effects to the animals. In conclusion, these nanoparticulate ferrofluids preferentially bind to cancer cells and, hence, exhibit a greater toxicity in these cells compared to the primary cells. They are also effective against solid tumors in vitro, can cross the BBB in Drosophila, and are nontoxic based on the developmental studies of flies raised in ferrofluid-infused media. STATEMENT OF SIGNIFICANCE: We demonstrate that a novel, hydrothermally synthesized composite nanoparticle-based ferrofluid is effective in reducing the viability of osteosarcoma and glioblastoma cells in vitro, while having minimal effects on primary cell lines. In 3D tumor spheroids, nanoparticles greatly reduced the metastatic migration of cancer cells, while the tumor viability was reduced compared to the control group by applying magnetic hyperthermia to nanoparticle-treated spheroids. Both in vitro and in vivo models of the blood-brain barrier evidence the ability of nanoparticles to cross the barrier and localize to the brain tissue. These composite nanoparticles show great promise as an anticancer biomaterial for the treatment of different types of cancer and may serve as an alternative or addendum to traditional chemotherapies.
Collapse
Affiliation(s)
- Victoria M Wu
- Advanced Materials and Nanobiotechnology Laboratory, Center for Targeted Drug Delivery, Department of Biomedical and Pharmaceutical Sciences, Chapman University, Irvine, CA 92618-1908, USA
| | - Eric Huynh
- Advanced Materials and Nanobiotechnology Laboratory, Center for Targeted Drug Delivery, Department of Biomedical and Pharmaceutical Sciences, Chapman University, Irvine, CA 92618-1908, USA
| | - Sean Tang
- Advanced Materials and Nanobiotechnology Laboratory, Center for Targeted Drug Delivery, Department of Biomedical and Pharmaceutical Sciences, Chapman University, Irvine, CA 92618-1908, USA
| | - Vuk Uskoković
- Advanced Materials and Nanobiotechnology Laboratory, Center for Targeted Drug Delivery, Department of Biomedical and Pharmaceutical Sciences, Chapman University, Irvine, CA 92618-1908, USA; Advanced Materials and Nanobiotechnology Laboratory, Department of Bioengineering, University of Illinois, Chicago, IL 60607-7052, USA.
| |
Collapse
|
41
|
Artiushin G, Zhang SL, Tricoire H, Sehgal A. Endocytosis at the Drosophila blood-brain barrier as a function for sleep. eLife 2018; 7:e43326. [PMID: 30475209 PMCID: PMC6255390 DOI: 10.7554/elife.43326] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Accepted: 11/09/2018] [Indexed: 12/18/2022] Open
Abstract
Glia are important modulators of neural activity, yet few studies link glia to sleep regulation. We find that blocking activity of the endocytosis protein, dynamin, in adult Drosophila glia increases sleep and enhances sleep need, manifest as resistance to sleep deprivation. Surface glia comprising the fly equivalent of the blood-brain barrier (BBB) mediate the effect of dynamin on sleep. Blocking dynamin in the surface glia causes ultrastructural changes, albeit without compromising the integrity of the barrier. Supporting a role for endocytic trafficking in sleep, a screen of Rab GTPases identifies sleep-modulating effects of the recycling endosome Rab11 in surface glia. We also find that endocytosis is increased in BBB glia during sleep and reflects sleep need. We propose that endocytic trafficking through the BBB represents a function of sleep.
Collapse
Affiliation(s)
- Gregory Artiushin
- Neuroscience Graduate GroupPerelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
| | - Shirley L Zhang
- Howard Hughes Medical InstitutePerelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
| | - Hervé Tricoire
- Laboratory of Degenerative Processes, Stress and AgingUMR8251, Université Paris DiderotParisFrance
| | - Amita Sehgal
- Neuroscience Graduate GroupPerelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
- Howard Hughes Medical InstitutePerelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
| |
Collapse
|
42
|
Yildirim K, Petri J, Kottmeier R, Klämbt C. Drosophila glia: Few cell types and many conserved functions. Glia 2018; 67:5-26. [DOI: 10.1002/glia.23459] [Citation(s) in RCA: 75] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Revised: 04/25/2018] [Accepted: 05/04/2018] [Indexed: 12/20/2022]
Affiliation(s)
- Kerem Yildirim
- Institute for Neuro and Behavioral Biology; University of Münster; Badestraße 9, 48149 Münster Germany
| | - Johanna Petri
- Institute for Neuro and Behavioral Biology; University of Münster; Badestraße 9, 48149 Münster Germany
| | - Rita Kottmeier
- Institute for Neuro and Behavioral Biology; University of Münster; Badestraße 9, 48149 Münster Germany
| | - Christian Klämbt
- Institute for Neuro and Behavioral Biology; University of Münster; Badestraße 9, 48149 Münster Germany
| |
Collapse
|
43
|
Zubiaur P, Saiz-Rodríguez M, Koller D, Ovejero-Benito MC, Wojnicz A, Abad-Santos F. How to make P-glycoprotein (ABCB1, MDR1) harbor mutations and measure its expression and activity in cell cultures? Pharmacogenomics 2018; 19:1285-1297. [PMID: 30334473 DOI: 10.2217/pgs-2018-0101] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Several polymorphisms have been identified in ABCB1, the gene encoding for the P-glycoprotein. This transporter alters the pharmacokinetics or effectiveness of drugs by excreting them from cells where it is expressed (e.g., blood-brain barrier, intestine or tumors). No consensus has been reached regarding the functional consequences of these polymorphisms in the transporter's function. The aim of this review was to describe a methodology that allows the assessment of P-gp function when harboring polymorphisms. We describe how to obtain cell lines with high expression levels of the transporter with polymorphisms and several tactics to measure its expression and activity. This methodology may help elucidate the contribution of polymorphisms in ABCB1 to drug pharmacokinetics, effectiveness and safety or to cancer chemotherapy failure.
Collapse
Affiliation(s)
- Pablo Zubiaur
- Clinical Pharmacology Department, Hospital Universitario de la Princesa, Instituto Teófilo Hernando, Universidad Autónoma de Madrid (UAM), Instituto de Investigación Sanitaria la Princesa (IP), Madrid, Spain
| | - Miriam Saiz-Rodríguez
- Clinical Pharmacology Department, Hospital Universitario de la Princesa, Instituto Teófilo Hernando, Universidad Autónoma de Madrid (UAM), Instituto de Investigación Sanitaria la Princesa (IP), Madrid, Spain
| | - Dora Koller
- Clinical Pharmacology Department, Hospital Universitario de la Princesa, Instituto Teófilo Hernando, Universidad Autónoma de Madrid (UAM), Instituto de Investigación Sanitaria la Princesa (IP), Madrid, Spain
| | - María Carmen Ovejero-Benito
- Clinical Pharmacology Department, Hospital Universitario de la Princesa, Instituto Teófilo Hernando, Universidad Autónoma de Madrid (UAM), Instituto de Investigación Sanitaria la Princesa (IP), Madrid, Spain
| | - Aneta Wojnicz
- Clinical Pharmacology Department, Hospital Universitario de la Princesa, Instituto Teófilo Hernando, Universidad Autónoma de Madrid (UAM), Instituto de Investigación Sanitaria la Princesa (IP), Madrid, Spain
| | - Francisco Abad-Santos
- Clinical Pharmacology Department, Hospital Universitario de la Princesa, Instituto Teófilo Hernando, Universidad Autónoma de Madrid (UAM), Instituto de Investigación Sanitaria la Princesa (IP), Madrid, Spain.,UICEC Hospital Universitario de la Princesa, Plataforma SCReN (Spanish Clinical Reseach Network), Instituto de Investigación Sanitaria la Princesa (IP), Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
44
|
Abstract
This review by O'Brown et al. discusses the cellular nature of the blood–brain barrier (BBB) and the conservation and variation of BBB function across taxa. It compares the BBB across organisms in order to provide insight into the human BBB both under normal physiological conditions and in neurological diseases. The blood–brain barrier (BBB) restricts free access of molecules between the blood and the brain and is essential for regulating the neural microenvironment. Here, we describe how the BBB was initially characterized and how the current field evaluates barrier properties. We next detail the cellular nature of the BBB and discuss both the conservation and variation of BBB function across taxa. Finally, we examine our current understanding of mouse and zebrafish model systems, as we expect that comparison of the BBB across organisms will provide insight into the human BBB under normal physiological conditions and in neurological diseases.
Collapse
Affiliation(s)
- Natasha M O'Brown
- Department of Neurobiology, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Sarah J Pfau
- Department of Neurobiology, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Chenghua Gu
- Department of Neurobiology, Harvard Medical School, Boston, Massachusetts 02115, USA
| |
Collapse
|
45
|
Zhang SL, Yue Z, Arnold DM, Artiushin G, Sehgal A. A Circadian Clock in the Blood-Brain Barrier Regulates Xenobiotic Efflux. Cell 2018; 173:130-139.e10. [PMID: 29526461 PMCID: PMC5866247 DOI: 10.1016/j.cell.2018.02.017] [Citation(s) in RCA: 155] [Impact Index Per Article: 22.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2017] [Revised: 12/22/2017] [Accepted: 02/07/2018] [Indexed: 12/29/2022]
Abstract
Endogenous circadian rhythms are thought to modulate responses to external factors, but mechanisms that confer time-of-day differences in organismal responses to environmental insults/therapeutic treatments are poorly understood. Using a xenobiotic, we find that permeability of the Drosophila "blood"-brain barrier (BBB) is higher at night. The permeability rhythm is driven by circadian regulation of efflux and depends on a molecular clock in the perineurial glia of the BBB, although efflux transporters are restricted to subperineurial glia (SPG). We show that transmission of circadian signals across the layers requires cyclically expressed gap junctions. Specifically, during nighttime, gap junctions reduce intracellular magnesium ([Mg2+]i), a positive regulator of efflux, in SPG. Consistent with lower nighttime efflux, nighttime administration of the anti-epileptic phenytoin is more effective at treating a Drosophila seizure model. These findings identify a novel mechanism of circadian regulation and have therapeutic implications for drugs targeted to the central nervous system.
Collapse
Affiliation(s)
- Shirley L Zhang
- Center for Sleep and Circadian Neurobiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA; Chronobiology Program at Penn and Howard Hughes Medical Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Zhifeng Yue
- Chronobiology Program at Penn and Howard Hughes Medical Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Denice M Arnold
- Chronobiology Program at Penn and Howard Hughes Medical Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Gregory Artiushin
- Chronobiology Program at Penn and Howard Hughes Medical Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Amita Sehgal
- Center for Sleep and Circadian Neurobiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA; Chronobiology Program at Penn and Howard Hughes Medical Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|