1
|
Yasuda R, Hashimoto H, Oka M, Mok JW, Waqar M, Dauwalder B, Kanca O, Mizuno T, Yamamoto S. Functional analysis of pathogenic variants in LAMB1-related leukoencephalopathy reveals genotype-phenotype correlations and suggests its role in glial cells. Hum Mol Genet 2025; 34:990-999. [PMID: 40237576 DOI: 10.1093/hmg/ddaf050] [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: 12/13/2024] [Revised: 03/13/2025] [Accepted: 04/02/2025] [Indexed: 04/18/2025] Open
Abstract
Laminin B1 (LAMB1) is one of the extracellular matrix (ECM) proteins that make up the basement membrane. Early frameshift, late frameshift, and missense variants in LAMB1 have been reported to cause rare monogenic neurological disorders that are collectively known as LAMB1-related leukoencephalopathy. Although there is some genotype-phenotype correlation, functional consequences of pathogenic LAMB1 variants are largely unknown. In this study, we aimed to elucidate function of the fly ortholog of this gene (LanB1) in the nervous system and to further study the functional consequences of the LAMB1 variants using Drosophila melanogaster. We found that the LanB1 gene is expressed on the surface of adult fly brains in a subset of glia cells. We further found that LanB1 protein localizes to the blood-brain barrier (BBB) in adult fly brains and knockdown of LanB1 in the BBB resulted in short life span and locomotor defects. Although human LAMB1 was not able to function in flies, in vivo overexpression and rescue experiments using analogous variants in fly LanB1 suggested that the frameshift variants behave as strong loss-of-function (LoF) alleles whereas a missense variant functions as a milder LoF allele. In vitro assay in HEK293T cells revealed that late-truncated LAMB1 is uniquely detected as a monomer in the culture medium, which might be the basis of dominant inheritance of these variants through a gain-of-function mechanism. Our data contributes to the understanding of the ECM component of the fly BBB and lays the foundation to unravel the molecular consequences of different pathogenic variants in LAMB1.
Collapse
Affiliation(s)
- Rei Yasuda
- Department of Molecular and Human Genetics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, United States
- Jan and Duncan Neurological Research Institute, Texas Children's Hospital, 1250 Moursund St, Houston, TX 77030, United States
- Department of Neurology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Hirokazu Hashimoto
- Department of Molecular and Human Genetics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, United States
- Jan and Duncan Neurological Research Institute, Texas Children's Hospital, 1250 Moursund St, Houston, TX 77030, United States
| | - Mikiko Oka
- Department of Molecular and Human Genetics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, United States
- Jan and Duncan Neurological Research Institute, Texas Children's Hospital, 1250 Moursund St, Houston, TX 77030, United States
| | - Jung-Wan Mok
- Department of Molecular and Human Genetics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, United States
- Jan and Duncan Neurological Research Institute, Texas Children's Hospital, 1250 Moursund St, Houston, TX 77030, United States
| | - Marium Waqar
- Department of Biology and Biochemistry, University of Houston, 3455 Cullen Blvd, Houston, TX 77204, United States
| | - Brigitte Dauwalder
- Department of Biology and Biochemistry, University of Houston, 3455 Cullen Blvd, Houston, TX 77204, United States
| | - Oguz Kanca
- Department of Molecular and Human Genetics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, United States
- Jan and Duncan Neurological Research Institute, Texas Children's Hospital, 1250 Moursund St, Houston, TX 77030, United States
| | - Toshiki Mizuno
- Department of Neurology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Shinya Yamamoto
- Department of Molecular and Human Genetics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, United States
- Jan and Duncan Neurological Research Institute, Texas Children's Hospital, 1250 Moursund St, Houston, TX 77030, United States
| |
Collapse
|
2
|
Epiney DG, Chaya GM, Dillon NR, Lai SL, Doe CQ. Single nuclei RNA-sequencing of adult brain neurons derived from type 2 neuroblasts reveals transcriptional complexity in the insect central complex. eLife 2025; 14:RP105896. [PMID: 40371710 PMCID: PMC12081001 DOI: 10.7554/elife.105896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/16/2025] Open
Abstract
In both Drosophila and mammals, the brain contains the most diverse population of cell types of any tissue. It is generally accepted that transcriptional diversity is an early step in generating neuronal and glial diversity, followed by the establishment of a unique gene expression profile that determines morphology, connectivity, and function. In Drosophila, there are two types of neural stem cells, called Type 1 (T1) and Type 2 (T2) neuroblasts. The diversity of T2-derived neurons contributes a large portion of the central complex (CX), a conserved brain region that plays a role in sensorimotor integration. Recent work has revealed much of the connectome of the CX, but how this connectome is assembled remains unclear. Mapping the transcriptional diversity of T2-derived neurons is a necessary step in linking transcriptional profile to the assembly of the adult brain. Here we perform single nuclei RNA sequencing of T2 neuroblast-derived adult neurons and glia. We identify clusters containing all known classes of glia, clusters that are male/female enriched, and 161 neuron-specific clusters. We map neurotransmitter and neuropeptide expression and identify unique transcription factor combinatorial codes for each cluster. This is a necessary step that directs functional studies to determine whether each transcription factor combinatorial code specifies a distinct neuron type within the CX. We map several columnar neuron subtypes to distinct clusters and identify two neuronal classes (NPF+ and AstA+) that both map to two closely related clusters. Our data support the hypothesis that each transcriptional cluster represents one or a few closely related neuron subtypes.
Collapse
Affiliation(s)
- Derek G Epiney
- Institute of Neuroscience, Howard Hughes Medical Institute, University of OregonEugeneUnited States
| | - Gonzalo Morales Chaya
- Institute of Neuroscience, Howard Hughes Medical Institute, University of OregonEugeneUnited States
| | - Noah R Dillon
- Institute of Neuroscience, Howard Hughes Medical Institute, University of OregonEugeneUnited States
| | - Sen-Lin Lai
- Institute of Neuroscience, Howard Hughes Medical Institute, University of OregonEugeneUnited States
| | - Chris Q Doe
- Institute of Neuroscience, Howard Hughes Medical Institute, University of OregonEugeneUnited States
| |
Collapse
|
3
|
González-Gutiérrez A, Gaete J, Esparza A, Ibacache A, Contreras EG, Sierralta J. Starvation Induces Upregulation of Monocarboxylate Transport in Glial Cells at the Drosophila Blood-Brain Barrier. Glia 2025. [PMID: 40241296 DOI: 10.1002/glia.70021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Revised: 03/11/2025] [Accepted: 03/31/2025] [Indexed: 04/18/2025]
Abstract
Living organisms can sense and adapt to constant changes in food availability. Maintaining a homeostatic supply of energy molecules is crucial for animal survival and normal organ functioning, particularly the brain, due to its high-energy demands. However, the mechanisms underlying brain adaptive responses to food availability have not been completely established. The nervous system is separated from the rest of the body by a physical barrier called the blood-brain barrier (BBB). In addition to its structural role, the BBB regulates the transport of metabolites and nutrients into the nervous system. This regulation is achieved through adaptive mechanisms that control the transport of nutrients, including glucose and monocarboxylates such as lactate, pyruvate, and ketone bodies. In Drosophila melanogaster, carbohydrate transporters increase their expression in glial cells of the BBB in response to starvation. However, changes in the expression or activity of Drosophila monocarboxylate transporters (dMCTs) at the BBB have not yet been reported. Here, we show that neuronal ATP levels remain unaffected despite reduced energy-related metabolites in the hemolymph of Drosophila larvae during starvation. Simultaneously, the transport of lactate and beta-hydroxybutyrate increases in the glial cells of the BBB. Using genetically encoded sensors, we identified Yarqay as a proton-coupled monocarboxylate transporter whose expression is upregulated in the subperineurial glia of the BBB during starvation. Our findings reveal a novel component of the adaptive response of the brain to starvation: the increase in the transport of monocarboxylates across the BBB, mediated by Yarqay, a novel dMCT enriched in the BBB.
Collapse
Affiliation(s)
- Andrés González-Gutiérrez
- Department of Neuroscience, School of Medicine, University of Chile, Independencia, Chile
- Institute of Biomedical Neurosciences (BNI), School of Medicine, University of Chile, Independencia, Chile
| | - Jorge Gaete
- Department of Neuroscience, School of Medicine, University of Chile, Independencia, Chile
| | - Andrés Esparza
- Department of Neuroscience, School of Medicine, University of Chile, Independencia, Chile
| | - Andrés Ibacache
- Department of Neuroscience, School of Medicine, University of Chile, Independencia, Chile
| | - Esteban G Contreras
- Department of Cell Biology, Faculty of Biological Sciences, University of Concepción, Concepción, Chile
| | - Jimena Sierralta
- Department of Neuroscience, School of Medicine, University of Chile, Independencia, Chile
- Institute of Biomedical Neurosciences (BNI), School of Medicine, University of Chile, Independencia, Chile
| |
Collapse
|
4
|
Epiney D, Morales Chaya GN, Dillon NR, Lai SL, Doe CQ. Transcriptional complexity in the insect central complex: single nuclei RNA-sequencing of adult brain neurons derived from type 2 neuroblasts. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2023.12.10.571022. [PMID: 40093129 PMCID: PMC11908175 DOI: 10.1101/2023.12.10.571022] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
In both invertebrates such as Drosophila and vertebrates such as mouse or human, the brain contains the most diverse population of cell types of any tissue. It is generally accepted that transcriptional diversity is an early step in generating neuronal and glial diversity, followed by the establishment of a unique gene expression profile that determines morphology, connectivity, and function. In Drosophila, there are two types of neural stem cells, called Type 1 (T1) and Type 2 (T2) neuroblasts. In contrast to T1 neuroblasts, T2 neuroblasts generate intermediate neural progenitors (INPs) that expand the number and diversity of cell types. The diversity of T2-derived neurons contributes a large portion of the central complex (CX), a conserved brain region that plays a role in sensorimotor integration. Recent work has revealed much of the connectome of the CX, but how this connectome is assembled remains unclear. Mapping the transcriptional diversity of neurons derived from T2 neuroblasts is a necessary step in linking transcriptional profile to the assembly of the adult brain. Here we perform single nuclei RNA sequencing of T2 neuroblast-derived adult neurons and glia. We identify clusters containing all known classes of glia, clusters that are male/female enriched, and 161 neuron-specific clusters. We map neurotransmitter and neuropeptide expression and identify unique transcription factor combinatorial codes for each cluster (presumptive neuron subtype). This is a necessary step that directs functional studies to determine whether each transcription factor combinatorial code specifies a distinct neuron type within the CX. We map several columnar neuron subtypes to distinct clusters and identify two neuronal classes (NPF+ and AstA+) that both map to two closely related clusters. Our data support the hypothesis that each transcriptional cluster represents one or a few closely related neuron subtypes.
Collapse
Affiliation(s)
| | | | | | - Sen-Lin Lai
- Institute of Neuroscience, Howard Hughes Medical Institute, University of Oregon, Eugene, OR 97403
| | - Chris Q. Doe
- Institute of Neuroscience, Howard Hughes Medical Institute, University of Oregon, Eugene, OR 97403
| |
Collapse
|
5
|
Avila A, Lewandowski AS, Li Y, Gui J, Lee KA, Yang Z, Kim M, Lyles JT, Man K, Sehgal A, Chandler JD, Zhang SL. A carnitine transporter at the blood-brain barrier modulates sleep via glial lipid metabolism in Drosophila. Proc Natl Acad Sci U S A 2025; 122:e2421178122. [PMID: 39847335 PMCID: PMC11789159 DOI: 10.1073/pnas.2421178122] [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: 10/14/2024] [Accepted: 12/13/2024] [Indexed: 01/24/2025] Open
Abstract
To regulate brain function, peripheral compounds must traverse the blood-brain barrier (BBB), an interface between the brain and the circulatory system. To determine whether specific transport mechanisms are relevant for sleep, we conducted a BBB-specific inducible RNAi knockdown (iKD) screen for genes affecting sleep in Drosophila. We observed reduced sleep with knockdown of solute carrier CG6126, a carnitine transporter, as determined by isotope flux. Our findings suggest that CG6126 regulation of sleep is through the role of the carnitine shuttle in regulating fatty acid metabolism as lipid droplets accumulate in the brains of CG6126 BBB iKD flies. Knocking down mitochondrial carnitine transferases in non-BBB glial cells mimicked the reduced sleep of the CG6126 BBB iKD flies, while bypassing the necessity of carnitine transport with dietary medium-chain fatty acids or palmitoylcarnitine rescued sleep. We propose that carnitine transport via CG6126 promotes brain fatty acid metabolism necessary for maintaining sleep.
Collapse
Affiliation(s)
- Ashley Avila
- Department of Cell Biology, Emory University, Atlanta, GA30322
| | | | - Yongjun Li
- HHMI, University of Pennsylvania, Philadelphia, PA19104
- Chronobiology and Sleep Institute, University of Pennsylvania, Philadelphia, PA19104
| | - Jesse Gui
- Department of Cell Biology, Emory University, Atlanta, GA30322
| | - Kaeun A. Lee
- Department of Cell Biology, Emory University, Atlanta, GA30322
| | - Zhenglang Yang
- Department of Cell Biology, Emory University, Atlanta, GA30322
| | - Mari Kim
- Department of Cell Biology, Emory University, Atlanta, GA30322
| | - James T. Lyles
- Department of Pediatrics, Emory University, Atlanta, GA30322
| | - Kai Man
- Department of Cell Biology, Emory University, Atlanta, GA30322
| | - Amita Sehgal
- HHMI, University of Pennsylvania, Philadelphia, PA19104
- Chronobiology and Sleep Institute, University of Pennsylvania, Philadelphia, PA19104
| | - Joshua D. Chandler
- Department of Pediatrics, Emory University, Atlanta, GA30322
- Children’s Healthcare of Atlanta, Emory University, Atlanta, GA30322
| | | |
Collapse
|
6
|
Hu R, Li M, Chen S, Wang M, Tao X, Zhu Y, Yan H, Liu Y. Sniffer restricts arboviral brain infections by regulating ROS levels and protecting blood-brain barrier integrity in Drosophila and mosquitoes. PLoS Pathog 2024; 20:e1012797. [PMID: 39680616 PMCID: PMC11684763 DOI: 10.1371/journal.ppat.1012797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 12/30/2024] [Accepted: 12/03/2024] [Indexed: 12/18/2024] Open
Abstract
Arthropod-borne viruses (arboviruses) are transmitted to humans by arthropod vectors and pose a serious threat to global public health. Neurotropic arboviruses including Sindbis virus (SINV) persistently infect the central nervous system (CNS) of vector insects without causing notable pathological changes or affecting their behavior or lifespan. However, the mechanisms by which vector insects evade these viral infections in the brains are poorly understood. In this study, we found that loss of the carbonyl reductase Sniffer (Sni) led to a significant increase in SINV infection in the Drosophila brain. Sni regulates reactive oxygen species (ROS) levels, and its depletion leads to elevated ROS, which in turn disrupts the septate junctions (SJs) between subperineurial glia (SPG) cells, compromising the integrity and barrier function of the blood-brain barrier (BBB). Genetic and pharmacological reduction of ROS restored BBB integrity and reduced viral load in the brains of Sni-depleted flies. Additionally, we identified Sni homologs and revealed that the antiviral function of Sni is highly conserved in mosquitoes, where it regulates ROS and protects BBB integrity. Our results revealed an evolutionarily conserved antiviral mechanism in which Sni acts as an antioxidant that protects BBB integrity and restricts viral infection in the vector insect brain.
Collapse
Affiliation(s)
- Rui Hu
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, China
| | - Mengzhu Li
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, China
| | - Shulin Chen
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, China
| | - Man Wang
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, China
| | - Xinjun Tao
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, China
| | - Yihan Zhu
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, China
| | - Huan Yan
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, China
| | - Yuan Liu
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, China
| |
Collapse
|
7
|
Mulay AR, Hwang J, Kim DH. Microphysiological Blood-Brain Barrier Systems for Disease Modeling and Drug Development. Adv Healthc Mater 2024; 13:e2303180. [PMID: 38430211 PMCID: PMC11338747 DOI: 10.1002/adhm.202303180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 02/22/2024] [Indexed: 03/03/2024]
Abstract
The blood-brain barrier (BBB) is a highly controlled microenvironment that regulates the interactions between cerebral blood and brain tissue. Due to its selectivity, many therapeutics targeting various neurological disorders are not able to penetrate into brain tissue. Pre-clinical studies using animals and other in vitro platforms have not shown the ability to fully replicate the human BBB leading to the failure of a majority of therapeutics in clinical trials. However, recent innovations in vitro and ex vivo modeling called organs-on-chips have shown the potential to create more accurate disease models for improved drug development. These microfluidic platforms induce physiological stressors on cultured cells and are able to generate more physiologically accurate BBBs compared to previous in vitro models. In this review, different approaches to create BBBs-on-chips are explored alongside their application in modeling various neurological disorders and potential therapeutic efficacy. Additionally, organs-on-chips use in BBB drug delivery studies is discussed, and advances in linking brain organs-on-chips onto multiorgan platforms to mimic organ crosstalk are reviewed.
Collapse
Affiliation(s)
- Atharva R. Mulay
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, Maryland 21218
| | - Jihyun Hwang
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, Maryland 21218
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, 21205
| | - Deok-Ho Kim
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, 21205
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, 21205
- Center for Microphysiological Systems, Johns Hopkins University School of Medicine, Baltimore, MD, 21205
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, MD, 21218
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, 21218
| |
Collapse
|
8
|
Lau K, Kotzur R, Richter F. Blood-brain barrier alterations and their impact on Parkinson's disease pathogenesis and therapy. Transl Neurodegener 2024; 13:37. [PMID: 39075566 PMCID: PMC11285262 DOI: 10.1186/s40035-024-00430-z] [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: 03/20/2024] [Accepted: 07/11/2024] [Indexed: 07/31/2024] Open
Abstract
There is increasing evidence for blood-brain barrier (BBB) alterations in Parkinson's disease (PD), the second most common neurodegenerative disorder with rapidly rising prevalence. Altered tight junction and transporter protein levels, accumulation of α-synuclein and increase in inflammatory processes lead to extravasation of blood molecules and vessel degeneration. This could result in a self-perpetuating pathophysiology of inflammation and BBB alteration, which contribute to neurodegeneration. Toxin exposure or α-synuclein over-expression in animal models has been shown to initiate similar pathologies, providing a platform to study underlying mechanisms and therapeutic interventions. Here we provide a comprehensive review of the current knowledge on BBB alterations in PD patients and how rodent models that replicate some of these changes can be used to study disease mechanisms. Specific challenges in assessing the BBB in patients and in healthy controls are discussed. Finally, a potential role of BBB alterations in disease pathogenesis and possible implications for therapy are explored. The interference of BBB alterations with current and novel therapeutic strategies requires more attention. Brain region-specific BBB alterations could also open up novel opportunities to target specifically vulnerable neuronal subpopulations.
Collapse
Affiliation(s)
- Kristina Lau
- Department of Pharmacology, Toxicology and Pharmacy, University of Veterinary Medicine Hannover, Foundation, Bünteweg 17, 30559, Hannover, Germany
- Center for Systems Neuroscience, Hannover, Germany
| | - Rebecca Kotzur
- Department of Pharmacology, Toxicology and Pharmacy, University of Veterinary Medicine Hannover, Foundation, Bünteweg 17, 30559, Hannover, Germany
| | - Franziska Richter
- Department of Pharmacology, Toxicology and Pharmacy, University of Veterinary Medicine Hannover, Foundation, Bünteweg 17, 30559, Hannover, Germany.
- Center for Systems Neuroscience, Hannover, Germany.
| |
Collapse
|
9
|
Tsuji A, Kotani E, Inoue YH. Sesamin Exerts an Antioxidative Effect by Activating the Nrf2 Transcription Factor in the Glial Cells of the Central Nervous System in Drosophila Larvae. Antioxidants (Basel) 2024; 13:787. [PMID: 39061856 PMCID: PMC11274309 DOI: 10.3390/antiox13070787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 06/27/2024] [Accepted: 06/27/2024] [Indexed: 07/28/2024] Open
Abstract
Sesame seeds are abundant in sesamin, which exerts health-promoting effects such as extending the lifespan of adult Drosophila and suppressing oxidative stress by activating the Nrf2 transcription factor. Here, we investigated whether sesamin activated Nrf2 in larval tissues and induced the expression of Nrf2 target genes. In the sesamin-fed larvae, Nrf2 was activated in the central nervous system (CNS), gut, and salivary glands. The ectopic expression of Keap1 in glial cells inhibited sesamin-induced Nrf2 activation in the whole CNS more than in the neurons, indicating that sesamin activates Nrf2 in glia efficiently. We labeled the astrocytes as well as cortex and surface glia with fluorescence to identify the glial cell types in which Nrf2 was activated; we observed their activation in both cell types. These data suggest that sesamin may stimulate the expression of antioxidative genes in glial cells. Among the 17 candidate Nrf2 targets, the mRNA levels of Cyp6a2 and Cyp6g1 in cytochrome P450 were elevated in the CNS, gut, and salivary glands of the sesamin-fed larvae. However, this elevation did not lead to resistance against imidacloprid, which is detoxified by these enzymes. Our results suggest that sesamin may exert similar health-promoting effects on the human CNS and digestive tissues.
Collapse
Affiliation(s)
| | | | - Yoshihiro H. Inoue
- Biomedical Research Center, Graduate School of Science and Technology, Kyoto Institute of Technology, Matsugasaki, Sakyo, Kyoto 606-0962, Japan; (A.T.); (E.K.)
| |
Collapse
|
10
|
Contreras EG, Kautzmann S, Klämbt C. The Drosophila blood-brain barrier invades the nervous system in a GPCR-dependent manner. Front Cell Neurosci 2024; 18:1397627. [PMID: 38846639 PMCID: PMC11153769 DOI: 10.3389/fncel.2024.1397627] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Accepted: 05/07/2024] [Indexed: 06/09/2024] Open
Abstract
The blood-brain barrier (BBB) represents a crucial interface between the circulatory system and the brain. In Drosophila melanogaster, the BBB is composed of perineurial and subperineurial glial cells. The perineurial glial cells are small mitotically active cells forming the outermost layer of the nervous system and are engaged in nutrient uptake. The subperineurial glial cells form occluding septate junctions to prevent paracellular diffusion of macromolecules into the nervous system. To address whether the subperineurial glia just form a simple barrier or whether they establish specific contacts with both the perineurial glial cells and inner central nervous system (CNS) cells, we undertook a detailed morphological analysis. Using genetically encoded markers alongside with high-resolution laser scanning confocal microscopy and transmission electron microscopy, we identified thin cell processes extending into the perineurial layer and into the CNS cortex. Interestingly, long cell processes were observed reaching the glia ensheathing the neuropil of the central brain. GFP reconstitution experiments highlighted multiple regions of membrane contacts between subperineurial and ensheathing glia. Furthermore, we identify the G-protein-coupled receptor (GPCR) Moody as negative regulator of the growth of subperineurial cell processes. Loss of moody triggered a massive overgrowth of subperineurial cell processes into the CNS cortex and, moreover, affected the polarized localization of the xenobiotic transporter Mdr65. Finally, we found that GPCR signaling, but not septate junction formation, is responsible for controlling membrane overgrowth. Our findings support the notion that the Drosophila BBB is able to bridge the communication gap between circulation and synaptic regions of the brain by long cell processes.
Collapse
Affiliation(s)
| | | | - Christian Klämbt
- Multiscale Imaging Center, Institute of Neuro- and Behavioral Biology, University of Münster, Münster, Germany
| |
Collapse
|
11
|
Avila A, Zhang SL. A circadian clock regulates the blood-brain barrier across phylogeny. VITAMINS AND HORMONES 2024; 126:241-287. [PMID: 39029975 DOI: 10.1016/bs.vh.2024.04.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/21/2024]
Abstract
As the central regulatory system of an organism, the brain is responsible for overseeing a wide variety of physiological processes essential for an organism's survival. To maintain the environment necessary for neurons to function, the brain requires highly selective uptake and elimination of specific molecules through the blood-brain barrier (BBB). As an organism's activities vary throughout the day, how does the BBB adapt to meet the changing needs of the brain? A mechanism is through temporal regulation of BBB permeability via its circadian clock, which will be the focal point of this chapter. To comprehend the circadian clock's role within the BBB, we will first examine the anatomy of the BBB and the transport mechanisms enabling it to fulfill its role as a restrictive barrier. Next, we will define the circadian clock, and the discussion will encompass an introduction to circadian rhythms, the Transcription-Translation Feedback Loop (TTFL) as the mechanistic basis of circadian timekeeping, and the organization of tissue clocks found in organisms. Then, we will cover the role of the circadian rhythms in regulating the cellular mechanisms and functions of the BBB. We discuss the implications of this regulation in influencing sleep behavior, the progression of neurodegenerative diseases, and finally drug delivery for treatment of neurological diseases.
Collapse
Affiliation(s)
- Ashley Avila
- Cell Biology Department, Emory University, Atlanta, GA, United States
| | - Shirley L Zhang
- Cell Biology Department, Emory University, Atlanta, GA, United States.
| |
Collapse
|
12
|
Abstract
The blood-brain barrier (BBB) is a critical interface separating the central nervous system from the peripheral circulation, ensuring brain homeostasis and function. Recent research has unveiled a profound connection between the BBB and circadian rhythms, the endogenous oscillations synchronizing biological processes with the 24-hour light-dark cycle. This review explores the significance of circadian rhythms in the context of BBB functions, with an emphasis on substrate passage through the BBB. Our discussion includes efflux transporters and the molecular timing mechanisms that regulate their activities. A significant focus of this review is the potential implications of chronotherapy, leveraging our knowledge of circadian rhythms for improving drug delivery to the brain. Understanding the temporal changes in BBB can lead to optimized timing of drug administration, to enhance therapeutic efficacy for neurological disorders while reducing side effects. By elucidating the interplay between circadian rhythms and drug transport across the BBB, this review offers insights into innovative therapeutic interventions.
Collapse
Affiliation(s)
- Mari Kim
- Cell Biology Department, Emory University, Atlanta, GA, USA
| | | | | |
Collapse
|
13
|
Feng M, Fei S, Zou J, Xia J, Lai W, Huang Y, Swevers L, Sun J. Single-Nucleus Sequencing of Silkworm Larval Brain Reveals the Key Role of Lysozyme in the Antiviral Immune Response in Brain Hemocytes. J Innate Immun 2024; 16:173-187. [PMID: 38387449 PMCID: PMC10965234 DOI: 10.1159/000537815] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2023] [Accepted: 02/01/2024] [Indexed: 02/24/2024] Open
Abstract
INTRODUCTION The brain is considered as an immune-privileged organ, yet innate immune reactions can occur in the central nervous system of vertebrates and invertebrates. Silkworm (Bombyx mori) is an economically important insect and a lepidopteran model species. The diversity of cell types in the silkworm brain, and how these cell subsets produce an immune response to virus infection, remains largely unknown. METHODS Single-nucleus RNA sequencing (snRNA-seq), bioinformatics analysis, RNAi, and other methods were mainly used to analyze the cell types and gene functions of the silkworm brain. RESULTS We used snRNA-seq to identify 19 distinct clusters representing Kenyon cell, glial cell, olfactory projection neuron, optic lobes neuron, hemocyte-like cell, and muscle cell types in the B. mori nucleopolyhedrovirus (BmNPV)-infected and BmNPV-uninfected silkworm larvae brain at the late stage of infection. Further, we found that the cell subset that exerts an antiviral function in the silkworm larvae brain corresponds to hemocytes. Specifically, antimicrobial peptides were significantly induced by BmNPV infection in the hemocytes, especially lysozyme, exerting antiviral effects. CONCLUSION Our single-cell dataset reveals the diversity of silkworm larvae brain cells, and the transcriptome analysis provides insights into the immune response following virus infection at the single-cell level.
Collapse
Affiliation(s)
- Min Feng
- Guangdong Provincial Key Laboratory of Agro-animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Shigang Fei
- Guangdong Provincial Key Laboratory of Agro-animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Jinglei Zou
- Guangdong Provincial Key Laboratory of Agro-animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Junming Xia
- Guangdong Provincial Key Laboratory of Agro-animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Wenxuan Lai
- Guangdong Provincial Key Laboratory of Agro-animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Yigui Huang
- Guangdong Provincial Key Laboratory of Agro-animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Luc Swevers
- Insect Molecular Genetics and Biotechnology, National Centre for Scientific Research Demokritos, Institute of Biosciences and Applications, Athens, Greece
| | - Jingchen Sun
- Guangdong Provincial Key Laboratory of Agro-animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, China
| |
Collapse
|
14
|
Gasque SN, Han Y, van der Ham I, van Leeuwen D, van Oers MM, Haverkamp A, Ros VID. Baculovirus entry into the central nervous system of Spodoptera exigua caterpillars is independent of the viral protein tyrosine phosphatase. Open Biol 2024; 14:230278. [PMID: 38378139 PMCID: PMC10878822 DOI: 10.1098/rsob.230278] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2023] [Accepted: 01/04/2024] [Indexed: 02/22/2024] Open
Abstract
Neuroparasitism concerns the hostile take-over of a host's nervous system by a foreign invader, in order to alter the behaviour of the host in favour of the parasite. One of the most remarkable cases of parasite-induced host behavioural manipulation comprises the changes baculoviruses induce in their caterpillar hosts. Baculoviruses may manipulate caterpillar behaviour in two ways: hyperactivity (increased movement in the horizontal plane) and/or tree-top disease (movement to elevated levels in the vertical plane). Those behavioural changes are followed by liquefaction and death of the caterpillar. In Autographa californica multiple nucleopolyhedrovirus (AcMNPV)-infected Spodoptera exigua caterpillars, an enzymatic active form of the virally encoded protein tyrosine phosphatase (PTP) is needed for the expression of hyperactivity from 3 days post infection (dpi). Using eGFP-expressing recombinant AcMNPV strains, we show that infection of the caterpillar's central nervous system (CNS) can be observed primarily from 3 dpi onwards. In addition, we demonstrate that the structural and enzymatic function of PTP does not play a role in infection of the CNS. Instead we show that the virus entered the CNS via the trachea, progressing caudally to frontally through the CNS and that the infection progressed from the outermost cell layers towards the inner cell layers of the CNS, in a PTP independent manner. These findings help to further understand parasitic manipulation and the mechanisms by which neuroparasites infect the host nervous system to manipulate host behaviour.
Collapse
Affiliation(s)
- Simone N. Gasque
- Laboratory of Virology, Wageningen University and Research, Droevendaalsesteeg 1, 6708 PB Wageningen, The Netherlands
| | - Yue Han
- Laboratory of Virology, Wageningen University and Research, Droevendaalsesteeg 1, 6708 PB Wageningen, The Netherlands
| | - Iris van der Ham
- Laboratory of Virology, Wageningen University and Research, Droevendaalsesteeg 1, 6708 PB Wageningen, The Netherlands
| | - Dorothy van Leeuwen
- Laboratory of Virology, Wageningen University and Research, Droevendaalsesteeg 1, 6708 PB Wageningen, The Netherlands
| | - Monique M. van Oers
- Laboratory of Virology, Wageningen University and Research, Droevendaalsesteeg 1, 6708 PB Wageningen, The Netherlands
| | - Alexander Haverkamp
- Laboratory of Entomology, Wageningen University and Research, Droevendaalsesteeg 1, 6708 PB Wageningen, The Netherlands
| | - Vera I. D. Ros
- Laboratory of Virology, Wageningen University and Research, Droevendaalsesteeg 1, 6708 PB Wageningen, The Netherlands
| |
Collapse
|
15
|
Valamparamban GF, Spéder P. Homemade: building the structure of the neurogenic niche. Front Cell Dev Biol 2023; 11:1275963. [PMID: 38107074 PMCID: PMC10722289 DOI: 10.3389/fcell.2023.1275963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Accepted: 11/16/2023] [Indexed: 12/19/2023] Open
Abstract
Neural stem/progenitor cells live in an intricate cellular environment, the neurogenic niche, which supports their function and enables neurogenesis. The niche is made of a diversity of cell types, including neurons, glia and the vasculature, which are able to signal to and are structurally organised around neural stem/progenitor cells. While the focus has been on how individual cell types signal to and influence the behaviour of neural stem/progenitor cells, very little is actually known on how the niche is assembled during development from multiple cellular origins, and on the role of the resulting topology on these cells. This review proposes to draw a state-of-the art picture of this emerging field of research, with the aim to expose our knowledge on niche architecture and formation from different animal models (mouse, zebrafish and fruit fly). We will span its multiple aspects, from the existence and importance of local, adhesive interactions to the potential emergence of larger-scale topological properties through the careful assembly of diverse cellular and acellular components.
Collapse
Affiliation(s)
| | - Pauline Spéder
- Institut Pasteur, Université Paris Cité, CNRS UMR3738, Structure and Signals in the Neurogenic Niche, Paris, France
| |
Collapse
|
16
|
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
|
17
|
Barooah N, Karmakar P, Sharanya MK, Mishra M, Bhasikuttan AC, Mohanty J. Spectroscopic features of a perylenediimide probe for sensing amyloid fibrils: in vivo imaging of Aβ-aggregates in a Drosophila model organism. J Mater Chem B 2023; 11:9545-9554. [PMID: 37753638 DOI: 10.1039/d3tb01233f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/28/2023]
Abstract
Customised perylenediimide (PDI) chromophores find diverse applications not only as chemosensors, inorganic-organic semiconductors, photovoltaics, photocatalysts, etc., but also in protein surface engineering, bio-sensors and drug delivery systems. This study focuses on the interaction of a custom synthesized phenylalanine derivatized perylenediimide (L-Phe-PDI) dye with a model protein, insulin, and its structurally distinct fibrils to develop fluorescence sensors for fibrillar aggregates and in vivo imaging applications. Detailed photophysical studies revealed that L-Phe-PDI gets aggregated in the presence of insulin and causes emission quenching at pH 7.4, which in the absence of insulin occurs only at pH ∼2. During in vitro incubation of insulin to its fibrils, the fluorescence intensity of the L-Phe-PDI probe is enhanced to ∼150 fold in a two-stage manner, manifesting the pathways of structural transformation to β-sheet rich mature fibrils. The in vivo sensing has further been validated in living models of the Aβ-mutant Drosophila fly, which is known to develop progressive neurodegeneration comparable to that of human brains with Alzheimer's disease (AD). Bioimaging of the L-Phe-PDI treated Aβ-mutant Drosophila documented the blood-brain/blood-retina-barrier cross-over ability of L-Phe-PDI with no toxic effects. Comparison of the fibrillar images from the brain and eye region with the reference thioflavin T (ThT) probe established the uptake of L-Phe-PDI by the aggregate/fibrillar moieties. The samples from L-Phe-PDI-treated flies apparently displayed reduced fibrillar spots, a possible case of L-Phe-PDI-induced disintegration of fibrillar aggregates at large, an observation substantiated by the improved phenotype activities as compared to the untreated flies. The findings reported both in vitro and in vivo with the L-Phe-PDI material for the first time open up avenues to explore the therapeutic potential of custom-designed PDI derivatives for amyloid fibril sensors and bioimaging.
Collapse
Affiliation(s)
- Nilotpal Barooah
- Radiation & Photochemistry Division, Bhabha Atomic Research Centre, Mumbai 400085, India.
- Homi Bhabha National Institute, Training School Complex, Anushaktinagar, Mumbai 400094, India
| | - Puja Karmakar
- Department of Life Science, National Institute of Technology Rourkela, Odisha 769008, India.
| | - M K Sharanya
- Radiation & Photochemistry Division, Bhabha Atomic Research Centre, Mumbai 400085, India.
| | - Monalisa Mishra
- Department of Life Science, National Institute of Technology Rourkela, Odisha 769008, India.
| | - Achikanath C Bhasikuttan
- Radiation & Photochemistry Division, Bhabha Atomic Research Centre, Mumbai 400085, India.
- Homi Bhabha National Institute, Training School Complex, Anushaktinagar, Mumbai 400094, India
| | - Jyotirmayee Mohanty
- Radiation & Photochemistry Division, Bhabha Atomic Research Centre, Mumbai 400085, India.
- Homi Bhabha National Institute, Training School Complex, Anushaktinagar, Mumbai 400094, India
| |
Collapse
|
18
|
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: 13] [Impact Index Per Article: 6.5] [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
|
19
|
Krupp S, Hubbard I, Tam O, Hammell GM, Dubnau J. TDP-43 pathology in Drosophila induces glial-cell type specific toxicity that can be ameliorated by knock-down of SF2/SRSF1. PLoS Genet 2023; 19:e1010973. [PMID: 37747929 PMCID: PMC10553832 DOI: 10.1371/journal.pgen.1010973] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 10/05/2023] [Accepted: 09/13/2023] [Indexed: 09/27/2023] Open
Abstract
Accumulation of cytoplasmic inclusions of TAR-DNA binding protein 43 (TDP-43) is seen in both neurons and glia in a range of neurodegenerative disorders, including amyotrophic lateral sclerosis (ALS), frontotemporal dementia (FTD) and Alzheimer's disease (AD). Disease progression involves non-cell autonomous interactions among multiple cell types, including neurons, microglia and astrocytes. We investigated the effects in Drosophila of inducible, glial cell type-specific TDP-43 overexpression, a model that causes TDP-43 protein pathology including loss of nuclear TDP-43 and accumulation of cytoplasmic inclusions. We report that TDP-43 pathology in Drosophila is sufficient to cause progressive loss of each of the 5 glial sub-types. But the effects on organismal survival were most pronounced when TDP-43 pathology was induced in the perineural glia (PNG) or astrocytes. In the case of PNG, this effect is not attributable to loss of the glial population, because ablation of these glia by expression of pro-apoptotic reaper expression has relatively little impact on survival. To uncover underlying mechanisms, we used cell-type-specific nuclear RNA sequencing to characterize the transcriptional changes induced by pathological TDP-43 expression. We identified numerous glial cell-type specific transcriptional changes. Notably, SF2/SRSF1 levels were found to be decreased in both PNG and in astrocytes. We found that further knockdown of SF2/SRSF1 in either PNG or astrocytes lessens the detrimental effects of TDP-43 pathology on lifespan, but extends survival of the glial cells. Thus TDP-43 pathology in astrocytes or PNG causes systemic effects that shorten lifespan and SF2/SRSF1 knockdown rescues the loss of these glia, and also reduces their systemic toxicity to the organism.
Collapse
Affiliation(s)
- Sarah Krupp
- Program in Neuroscience, Department of Neurobiology and Behavior, Stony Brook University, New York, United States of America
| | - Isabel Hubbard
- Program in Neuroscience, Department of Neurobiology and Behavior, Stony Brook University, New York, United States of America
| | - Oliver Tam
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, United States of America
| | - Gale M. Hammell
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, United States of America
| | - Josh Dubnau
- Program in Neuroscience, Department of Neurobiology and Behavior, Stony Brook University, New York, United States of America
- Department of Anesthesiology, Stony Brook School of Medicine, New York, United States of America
| |
Collapse
|
20
|
Santarelli S, Londero C, Soldano A, Candelaresi C, Todeschini L, Vernizzi L, Bellosta P. Drosophila melanogaster as a model to study autophagy in neurodegenerative diseases induced by proteinopathies. Front Neurosci 2023; 17:1082047. [PMID: 37274187 PMCID: PMC10232775 DOI: 10.3389/fnins.2023.1082047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Accepted: 04/14/2023] [Indexed: 06/06/2023] Open
Abstract
Proteinopathies are a large group of neurodegenerative diseases caused by both genetic and sporadic mutations in particular genes which can lead to alterations of the protein structure and to the formation of aggregates, especially toxic for neurons. Autophagy is a key mechanism for clearing those aggregates and its function has been strongly associated with the ubiquitin-proteasome system (UPS), hence mutations in both pathways have been associated with the onset of neurodegenerative diseases, particularly those induced by protein misfolding and accumulation of aggregates. Many crucial discoveries regarding the molecular and cellular events underlying the role of autophagy in these diseases have come from studies using Drosophila models. Indeed, despite the physiological and morphological differences between the fly and the human brain, most of the biochemical and molecular aspects regulating protein homeostasis, including autophagy, are conserved between the two species.In this review, we will provide an overview of the most common neurodegenerative proteinopathies, which include PolyQ diseases (Huntington's disease, Spinocerebellar ataxia 1, 2, and 3), Amyotrophic Lateral Sclerosis (C9orf72, SOD1, TDP-43, FUS), Alzheimer's disease (APP, Tau) Parkinson's disease (a-syn, parkin and PINK1, LRRK2) and prion diseases, highlighting the studies using Drosophila that have contributed to understanding the conserved mechanisms and elucidating the role of autophagy in these diseases.
Collapse
Affiliation(s)
- Stefania Santarelli
- Department of Cellular, Computational and Integrative Biology (CiBiO), University of Trento, Trento, Italy
| | - Chiara Londero
- Department of Cellular, Computational and Integrative Biology (CiBiO), University of Trento, Trento, Italy
| | - Alessia Soldano
- Department of Cellular, Computational and Integrative Biology (CiBiO), University of Trento, Trento, Italy
- Department of Neuroscience, Scuola Internazionale Superiore di Studi Avanzati (SISSA), Trieste, Italy
| | - Carlotta Candelaresi
- Department of Cellular, Computational and Integrative Biology (CiBiO), University of Trento, Trento, Italy
| | - Leonardo Todeschini
- Department of Cellular, Computational and Integrative Biology (CiBiO), University of Trento, Trento, Italy
| | - Luisa Vernizzi
- Institute of Molecular Life Sciences, University of Zurich, Zürich, Switzerland
| | - Paola Bellosta
- Department of Cellular, Computational and Integrative Biology (CiBiO), University of Trento, Trento, Italy
- Department of Medicine, NYU Langone Medical Center, New York, NY, United States
| |
Collapse
|
21
|
Elya C, Lavrentovich D, Lee E, Pasadyn C, Duval J, Basak M, Saykina V, de Bivort B. Neural mechanisms of parasite-induced summiting behavior in 'zombie' Drosophila. eLife 2023; 12:e85410. [PMID: 37184212 PMCID: PMC10259475 DOI: 10.7554/elife.85410] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Accepted: 05/14/2023] [Indexed: 05/16/2023] Open
Abstract
For at least two centuries, scientists have been enthralled by the "zombie" behaviors induced by mind-controlling parasites. Despite this interest, the mechanistic bases of these uncanny processes have remained mostly a mystery. Here, we leverage the Entomophthora muscae-Drosophila melanogaster "zombie fly" system to reveal the mechanistic underpinnings of summit disease, a manipulated behavior evoked by many fungal parasites. Using a high-throughput approach to measure summiting, we discovered that summiting behavior is characterized by a burst of locomotion and requires the host circadian and neurosecretory systems, specifically DN1p circadian neurons, pars intercerebralis to corpora allata projecting (PI-CA) neurons and corpora allata (CA), the latter being solely responsible for juvenile hormone (JH) synthesis and release. Using a machine learning classifier to identify summiting animals in real time, we observed that PI-CA neurons and CA appeared intact in summiting animals, despite invasion of adjacent regions of the "zombie fly" brain by E. muscae cells and extensive host tissue damage in the body cavity. The blood-brain barrier of flies late in their infection was significantly permeabilized, suggesting that factors in the hemolymph may have greater access to the central nervous system during summiting. Metabolomic analysis of hemolymph from summiting flies revealed differential abundance of several compounds compared to non-summiting flies. Transfusing the hemolymph of summiting flies into non-summiting recipients induced a burst of locomotion, demonstrating that factor(s) in the hemolymph likely cause summiting behavior. Altogether, our work reveals a neuro-mechanistic model for summiting wherein fungal cells perturb the fly's hemolymph, activating a neurohormonal pathway linking clock neurons to juvenile hormone production in the CA, ultimately inducing locomotor activity in their host.
Collapse
Affiliation(s)
- Carolyn Elya
- Department of Organismic and Evolutionary Biology, Harvard UniversityCambridgeUnited States
| | - Danylo Lavrentovich
- Department of Organismic and Evolutionary Biology, Harvard UniversityCambridgeUnited States
| | - Emily Lee
- Department of Organismic and Evolutionary Biology, Harvard UniversityCambridgeUnited States
| | - Cassandra Pasadyn
- Department of Organismic and Evolutionary Biology, Harvard UniversityCambridgeUnited States
| | - Jasper Duval
- Department of Organismic and Evolutionary Biology, Harvard UniversityCambridgeUnited States
| | - Maya Basak
- Department of Organismic and Evolutionary Biology, Harvard UniversityCambridgeUnited States
| | - Valerie Saykina
- Department of Organismic and Evolutionary Biology, Harvard UniversityCambridgeUnited States
| | - Benjamin de Bivort
- Department of Organismic and Evolutionary Biology, Harvard UniversityCambridgeUnited States
| |
Collapse
|
22
|
Krupp S, Tam O, Hammell MG, Dubnau J. TDP-43 pathology in Drosophila induces glial-cell type specific toxicity that can be ameliorated by knock-down of SF2/SRSF1. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.04.539439. [PMID: 37205372 PMCID: PMC10187300 DOI: 10.1101/2023.05.04.539439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
Accumulation of cytoplasmic inclusions of TAR-DNA binding protein 43 (TDP-43) is seen in both neurons and glia in a range of neurodegenerative disorders, including amyotrophic lateral sclerosis (ALS), frontotemporal dementia (FTD) and Alzheimer's disease (AD). Disease progression involves non-cell autonomous interactions among multiple cell types, including neurons, microglia and astrocytes. We investigated the effects in Drosophila of inducible, glial cell type-specific TDP-43 overexpression, a model that causes TDP-43 protein pathology including loss of nuclear TDP-43 and accumulation of cytoplasmic inclusions. We report that TDP-43 pathology in Drosophila is sufficient to cause progressive loss of each of the 5 glial sub-types. But the effects on organismal survival were most pronounced when TDP-43 pathology was induced in the perineural glia (PNG) or astrocytes. In the case of PNG, this effect is not attributable to loss of the glial population, because ablation of these glia by expression of pro-apoptotic reaper expression has relatively little impact on survival. To uncover underlying mechanisms, we used cell-type-specific nuclear RNA sequencing to characterize the transcriptional changes induced by pathological TDP-43 expression. We identified numerous glial cell-type specific transcriptional changes. Notably, SF2/SRSF1 levels were found to be decreased in both PNG and in astrocytes. We found that further knockdown of SF2/SRSF1 in either PNG or astrocytes lessens the detrimental effects of TDP-43 pathology on lifespan, but extends survival of the glial cells. Thus TDP-43 pathology in astrocytes or PNG causes systemic effects that shorten lifespan and SF2/SRSF1 knockdown rescues the loss of these glia, and also reduces their systemic toxicity to the organism.
Collapse
Affiliation(s)
- S. Krupp
- Program in Neuroscience, Department of Neurobiology and Behavior, Stony Brook University, NY 11794, USA
| | - O Tam
- Cold Spring Harbor Laboratory, 1 Bungtown road, Cold Spring Harbor, NY.,11794
| | - M Gale Hammell
- Cold Spring Harbor Laboratory, 1 Bungtown road, Cold Spring Harbor, NY.,11794
| | - J Dubnau
- Program in Neuroscience, Department of Neurobiology and Behavior, Stony Brook University, NY 11794, USA
- Department of Anesthesiology, Stony Brook School of Medicine, NY 11794, USA
| |
Collapse
|
23
|
Fischer ML, Fabian B, Pauchet Y, Wielsch N, Sachse S, Vilcinskas A, Vogel H. An Assassin's Secret: Multifunctional Cytotoxic Compounds in the Predation Venom of the Assassin Bug Psytalla horrida (Reduviidae, Hemiptera). Toxins (Basel) 2023; 15:toxins15040302. [PMID: 37104240 PMCID: PMC10144120 DOI: 10.3390/toxins15040302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 04/17/2023] [Accepted: 04/18/2023] [Indexed: 04/28/2023] Open
Abstract
Predatory assassin bugs produce venomous saliva that enables them to overwhelm, kill, and pre-digest large prey animals. Venom from the posterior main gland (PMG) of the African assassin bug Psytalla horrida has strong cytotoxic effects, but the responsible compounds are yet unknown. Using cation-exchange chromatography, we fractionated PMG extracts from P. horrida and screened the fractions for toxicity. Two venom fractions strongly affected insect cell viability, bacterial growth, erythrocyte integrity, and intracellular calcium levels in Drosophila melanogaster olfactory sensory neurons. LC-MS/MS analysis revealed that both fractions contained gelsolin, redulysins, S1 family peptidases, and proteins from the uncharacterized venom protein family 2. Synthetic peptides representing the putative lytic domain of redulysins had strong antimicrobial activity against Escherichia coli and/or Bacillus subtilis but only weak toxicity towards insect or mammalian cells, indicating a primary role in preventing the intake of microbial pathogens. In contrast, a recombinant venom protein family 2 protein significantly reduced insect cell viability but exhibited no antibacterial or hemolytic activity, suggesting that it plays a role in prey overwhelming and killing. The results of our study show that P. horrida secretes multiple cytotoxic compounds targeting different organisms to facilitate predation and antimicrobial defense.
Collapse
Affiliation(s)
- Maike Laura Fischer
- Department of Insect Symbiosis, Max Planck Institute for Chemical Ecology, 07745 Jena, Germany
| | - Benjamin Fabian
- Research Group Olfactory Coding, Max Planck Institute for Chemical Ecology, 07745 Jena, Germany
| | - Yannick Pauchet
- Department of Insect Symbiosis, Max Planck Institute for Chemical Ecology, 07745 Jena, Germany
| | - Natalie Wielsch
- Research Group Mass Spectrometry/Proteomics, Max Planck Institute for Chemical Ecology, 07745 Jena, Germany
| | - Silke Sachse
- Research Group Olfactory Coding, Max Planck Institute for Chemical Ecology, 07745 Jena, Germany
| | - Andreas Vilcinskas
- Institute for Insect Biotechnology, Justus Liebig University, 35392 Giessen, Germany
- Branch Bioresources of the Fraunhofer Institute for Molecular Biology and Applied Ecology, 35392 Giessen, Germany
| | - Heiko Vogel
- Department of Insect Symbiosis, Max Planck Institute for Chemical Ecology, 07745 Jena, Germany
| |
Collapse
|
24
|
Love CR, Gautam S, Lama C, Le NH, Dauwalder B. The Drosophila dopamine 2-like receptor D2R (Dop2R) is required in the blood brain barrier for male courtship. GENES, BRAIN, AND BEHAVIOR 2023; 22:e12836. [PMID: 36636829 PMCID: PMC9994173 DOI: 10.1111/gbb.12836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 12/11/2022] [Accepted: 12/22/2022] [Indexed: 01/14/2023]
Abstract
The blood brain barrier (BBB) has the essential function to protect the brain from potentially hazardous molecules while also enabling controlled selective uptake. How these processes and signaling inside BBB cells control neuronal function is an intense area of interest. Signaling in the adult Drosophila BBB is required for normal male courtship behavior and relies on male-specific molecules in the BBB. Here we show that the dopamine receptor D2R is expressed in the BBB and is required in mature males for normal mating behavior. Conditional adult male knockdown of D2R in BBB cells causes courtship defects. The courtship defects observed in genetic D2R mutants can be rescued by expression of normal D2R specifically in the BBB of adult males. Drosophila BBB cells are glial cells. Our findings thus identify a specific glial function for the DR2 receptor and dopamine signaling in the regulation of a complex behavior.
Collapse
Affiliation(s)
- Cameron R Love
- Department of Biology and Biochemistry, University of Houston, Houston, Texas, USA.,Center for Advanced Biotechnology and Medicine, Rutgers University, Piscataway, New Jersey, USA
| | - Sumit Gautam
- Department of Biology and Biochemistry, University of Houston, Houston, Texas, USA
| | - Chamala Lama
- Department of Biology and Biochemistry, University of Houston, Houston, Texas, USA
| | - Nhu Hoa Le
- Department of Biology and Biochemistry, University of Houston, Houston, Texas, USA
| | - Brigitte Dauwalder
- Department of Biology and Biochemistry, University of Houston, Houston, Texas, USA
| |
Collapse
|
25
|
Benmimoun B, Winkler B, Spéder P. Infection of the Developing Central Nervous System of Drosophila by Mammalian Eukaryotic and Prokaryotic Pathogens. Bio Protoc 2022; 12:e4563. [PMID: 36561113 PMCID: PMC9729854 DOI: 10.21769/bioprotoc.4563] [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: 08/02/2022] [Revised: 09/12/2022] [Accepted: 10/17/2022] [Indexed: 12/07/2022] Open
Abstract
Pathogen invasion of the central nervous system (CNS) is an important cause of infection-related mortality worldwide and can lead to severe neurological sequelae. To gain access to the CNS cells, pathogens have to overcome the blood-brain barrier (BBB), a protective fence from blood-borne factors. To study host-pathogen interactions, a number of cell culture and animal models were developed. However, in vitro models do not recapitulate the 3D architecture of the BBB and CNS tissue, and in vivo mammalian models present cellular and technical complexities as well as ethical issues, rendering systematic and genetic approaches difficult. Here, we present a two-pronged methodology allowing and validating the use of Drosophila larvae as a model system to decipher the mechanisms of infection in a developing CNS. First, an ex vivo protocol based on whole CNS explants serves as a fast and versatile screening platform, permitting the investigation of molecular and cellular mechanisms contributing to the crossing of the BBB and consequences of infection on the CNS. Then, an in vivo CNS infection protocol through direct pathogen microinjection into the fly circulatory system evaluates the impact of systemic parameters, including the contribution of circulating immune cells to CNS infection, and assesses infection pathogenicity at the whole host level. These combined complementary approaches identify mechanisms of BBB crossing and responses of a diversity of CNS cells contributing to infection, as well as novel virulence factors of the pathogen. This protocol was validated in: Nat Commun (2020), DOI: 10.1038/s41467-020-19826-2 Graphical abstract Procedures flowchart. Mammalian neurotropic pathogens could be tested in two Drosophila central nervous system (CNS) infection setups (ex vivo and in vivo) for their ability to: (1) invade the CNS (pathogen quantifications), (2) disturb blood-brain barrier permeability, (3) affect CNS host cell behaviour (gene expression), and (4) alter host viability.
Collapse
Affiliation(s)
| | - Bente Winkler
- Institut für Neuro- und Verhaltensbiologie, Universität Münster, Badestr. 9, 48149 Münster, Germany
| | - Pauline Spéder
- Institut Pasteur, UMR3738 CNRS, 75015 Paris, France
,
*For correspondence:
pauline.speder@pasteur
| |
Collapse
|
26
|
Delta/Notch signaling in glia maintains motor nerve barrier function and synaptic transmission by controlling matrix metalloproteinase expression. Proc Natl Acad Sci U S A 2022; 119:e2110097119. [PMID: 35969789 PMCID: PMC9407389 DOI: 10.1073/pnas.2110097119] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
We have made a surprising discovery linking Delta/Notch signaling in subperineurial glia (SPG) to the regulation of nerve ensheathment and neurotransmitter release at the Drosophila neuromuscular junction (NMJ). SPG, the counterpart of the endothelial layer in the vertebrate blood–brain barrier, form the key cellular layer that is critical for axonal ensheathment and the blood–brain barrier in Drosophila. Our findings demonstrate that Delta/Notch signaling exerts a constitutive negative inhibition on JNK signaling in SPG, thereby limiting the expression of Mmp1, a matrix metalloproteinase. SPG-specific and temporally regulated knockdown of Delta leads to breakdown of barrier function and compromises neurotransmitter release at the NMJ. Our results provide a mechanistic insight into the biology of barrier function and glia–neuron interactions. While the role of barrier function in establishing a protective, nutrient-rich, and ionically balanced environment for neurons has been appreciated for some time, little is known about how signaling cues originating in barrier-forming cells participate in maintaining barrier function and influence synaptic activity. We have identified Delta/Notch signaling in subperineurial glia (SPG), a crucial glial type for Drosophila motor axon ensheathment and the blood–brain barrier, to be essential for controlling the expression of matrix metalloproteinase 1 (Mmp1), a major regulator of the extracellular matrix (ECM). Our genetic analysis indicates that Delta/Notch signaling in SPG exerts an inhibitory control on Mmp1 expression. In the absence of this inhibition, abnormally enhanced Mmp1 activity disrupts septate junctions and glial ensheathment of peripheral motor nerves, compromising neurotransmitter release at the neuromuscular junction (NMJ). Temporally controlled and cell type–specific transgenic analysis shows that Delta/Notch signaling inhibits transcription of Mmp1 by inhibiting c-Jun N-terminal kinase (JNK) signaling in SPG. Our results provide a mechanistic insight into the regulation of neuronal health and function via glial-initiated signaling and open a framework for understanding the complex relationship between ECM regulation and the maintenance of barrier function.
Collapse
|
27
|
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
|
28
|
Avila L, Dunne E, Hofmann D, Brosi BJ. Upper-limit agricultural dietary exposure to streptomycin in the laboratory reduces learning and foraging in bumblebees. Proc Biol Sci 2022; 289:20212514. [PMID: 35135346 PMCID: PMC8826297 DOI: 10.1098/rspb.2021.2514] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Accepted: 01/13/2022] [Indexed: 11/12/2022] Open
Abstract
In the past decade, the broadcast-spray application of antibiotics in US crops has increased exponentially in response to bacterial crop pathogens, but little is known about the sublethal impacts on beneficial organisms in agroecosystems. This is concerning given the key roles that microbes play in modulating insect fitness. A growing body of evidence suggests that insect gut microbiomes may play a role in learning and behaviour, which are key for the survival of pollinators and for their pollination efficacy, and which in turn could be disrupted by dietary antibiotic exposure. In the laboratory, we tested the effects of an upper-limit dietary exposure to streptomycin (200 ppm)-an antibiotic widely used to treat bacterial pathogens in crops-on bumblebee (Bombus impatiens) associative learning, foraging and stimulus avoidance behaviour. We used two operant conditioning assays: a free movement proboscis extension reflex protocol focused on short-term memory formation, and an automated radio-frequency identification tracking system focused on foraging. We show that upper-limit dietary streptomycin exposure slowed training, decreased foraging choice accuracy, increased avoidance behaviour and was associated with reduced foraging on sucrose-rewarding artificial flowers flowers. This work underscores the need to further study the impacts of antibiotic use on beneficial insects in agricultural systems.
Collapse
Affiliation(s)
- Laura Avila
- Department of Biology, Emory University, Atlanta, GA 30322, USA
| | - Elizabeth Dunne
- Department of Biology, Emory University, Atlanta, GA 30322, USA
| | - David Hofmann
- Department of Physics, Emory University, Atlanta, GA 30322, USA
- Initiative in Theory and Modeling of Living Systems, Emory University, Atlanta, GA 30322, USA
| | - Berry J. Brosi
- Department of Biology, University of Washington, Seattle, WA 98195, USA
| |
Collapse
|
29
|
Ye X, Schreck KC, Ozer BH, Grossman SA. High-grade glioma therapy: adding flexibility in trial design to improve patient outcomes. Expert Rev Anticancer Ther 2022; 22:275-287. [PMID: 35130447 DOI: 10.1080/14737140.2022.2038138] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Outcomes for patients with high grade gliomas have changed little over the past thirty years. This realization prompted renewed efforts to increase flexibility in the design and conduct of clinical brain tumor trials. AREAS COVERED This manuscript reviews the development of clinical trial methods, challenges and considerations of flexible clinical trial designs, approaches to improve identification and testing of active agents for high grade gliomas, and evaluation of their delivery to the central nervous system. EXPERT OPINION Flexibility can be introduced in clinical trials in several ways. Flexible designs tout smaller sample sizes, adaptive modifications, fewer control arms, and inclusion of multiple arms in one study. Unfortunately, modifications in study designs cannot address two challenges that are largely responsible for the lack of progress in treating high grade gliomas: 1) the identification of active pharmaceutical agents and 2) the delivery of these agents to brain tumor tissue in therapeutic concentrations. To improve the outcomes of patients with high grade gliomas efforts must be focused on the pre-clinical screening of drugs for activity, the ability of these agents to achieve therapeutic concentrations in non-enhancing tumors, and a willingness to introduce novel compounds in minimally pre-treated patient populations.
Collapse
Affiliation(s)
- Xiaobu Ye
- The Johns Hopkins University School of Medicine and Sidney Kimmel Comprehensive Cancer Center, Baltimore MD, USA
| | - Karisa C Schreck
- The Johns Hopkins University School of Medicine and Sidney Kimmel Comprehensive Cancer Center, Baltimore MD, USA
| | - Byram H Ozer
- The Johns Hopkins University School of Medicine and Sidney Kimmel Comprehensive Cancer Center, Baltimore MD, USA
| | - Stuart A Grossman
- The Johns Hopkins University School of Medicine and Sidney Kimmel Comprehensive Cancer Center, Baltimore MD, USA
| |
Collapse
|
30
|
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
|
31
|
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
|
32
|
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: 25] [Impact Index Per Article: 6.3] [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
|
33
|
Kim J, Chuang HC, Wolf NK, Nicolai CJ, Raulet DH, Saijo K, Bilder D. Tumor-induced disruption of the blood-brain barrier promotes host death. Dev Cell 2021; 56:2712-2721.e4. [PMID: 34496290 DOI: 10.1016/j.devcel.2021.08.010] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 06/29/2021] [Accepted: 08/12/2021] [Indexed: 11/24/2022]
Abstract
Cancer patients often die from symptoms that manifest at a distance from any tumor. Mechanisms underlying these systemic physiological perturbations, called paraneoplastic syndromes, may benefit from investigation in non-mammalian systems. Using a non-metastatic Drosophila adult model, we find that malignant-tumor-produced cytokines drive widespread host activation of JAK-STAT signaling and cause premature lethality. STAT activity is particularly high in cells of the blood-brain barrier (BBB), where it induces aberrant BBB permeability. Remarkably, inhibiting STAT in the BBB not only rescues barrier function but also extends the lifespan of tumor-bearing hosts. We identify BBB damage in other pathological conditions that cause elevated inflammatory signaling, including obesity and infection, where BBB permeability also regulates host survival. IL-6-dependent BBB dysfunction is further seen in a mouse tumor model, and it again promotes host morbidity. Therefore, BBB alterations constitute a conserved lethal tumor-host interaction that also underlies other physiological morbidities.
Collapse
Affiliation(s)
- Jung Kim
- Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, CA 94720, USA
| | - Hsiu-Chun Chuang
- Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, CA 94720, USA
| | - Natalie K Wolf
- Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, CA 94720, USA
| | - Christopher J Nicolai
- Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, CA 94720, USA
| | - David H Raulet
- Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, CA 94720, USA
| | - Kaoru Saijo
- Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, CA 94720, USA; Helen Wills Neuroscience Institute, University of California Berkeley, Berkeley, CA 94720, USA
| | - David Bilder
- Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, CA 94720, USA.
| |
Collapse
|
34
|
Baker BM, Mokashi SS, Shankar V, Hatfield JS, Hannah RC, Mackay TFC, Anholt RRH. The Drosophila brain on cocaine at single-cell resolution. Genome Res 2021; 31:1927-1937. [PMID: 34035044 PMCID: PMC8494231 DOI: 10.1101/gr.268037.120] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 02/02/2021] [Indexed: 11/24/2022]
Abstract
Whereas the neurological effects of cocaine have been well documented, effects of acute cocaine consumption on genome-wide gene expression across the brain remain largely unexplored. This question cannot be readily addressed in humans but can be approached using the Drosophila melanogaster model, where gene expression in the entire brain can be surveyed at once. Flies exposed to cocaine show impaired locomotor activity, including climbing behavior and startle response (a measure of sensorimotor integration), and increased incidence of seizures and compulsive grooming. To identify specific cell populations that respond to acute cocaine exposure, we analyzed single-cell transcriptional responses in duplicate samples of flies that consumed fixed amounts of sucrose or sucrose supplemented with cocaine, in both sexes. Unsupervised clustering of the transcriptional profiles of a total of 86,224 cells yielded 36 distinct clusters. Annotation of clusters based on gene markers revealed that all major cell types (neuronal and glial) as well as neurotransmitter types from most brain regions were represented. The brain transcriptional responses to cocaine showed profound sexual dimorphism and were considerably more pronounced in males than females. Differential expression analysis within individual clusters indicated cluster-specific responses to cocaine. Clusters corresponding to Kenyon cells of the mushroom bodies and glia showed especially large transcriptional responses following cocaine exposure. Cluster specific coexpression networks and global interaction networks revealed a diverse array of cellular processes affected by acute cocaine exposure. These results provide an atlas of sexually dimorphic cocaine-modulated gene expression in a model brain.
Collapse
Affiliation(s)
- Brandon M Baker
- Center for Human Genetics, Department of Genetics and Biochemistry, Clemson University, Greenwood, South Carolina 29646, USA
| | - Sneha S Mokashi
- Center for Human Genetics, Department of Genetics and Biochemistry, Clemson University, Greenwood, South Carolina 29646, USA
| | - Vijay Shankar
- Center for Human Genetics, Department of Genetics and Biochemistry, Clemson University, Greenwood, South Carolina 29646, USA
| | - Jeffrey S Hatfield
- Center for Human Genetics, Department of Genetics and Biochemistry, Clemson University, Greenwood, South Carolina 29646, USA
| | - Rachel C Hannah
- Center for Human Genetics, Department of Genetics and Biochemistry, Clemson University, Greenwood, South Carolina 29646, USA
| | - Trudy F C Mackay
- Center for Human Genetics, Department of Genetics and Biochemistry, Clemson University, Greenwood, South Carolina 29646, USA
| | - Robert R H Anholt
- Center for Human Genetics, Department of Genetics and Biochemistry, Clemson University, Greenwood, South Carolina 29646, USA
| |
Collapse
|
35
|
Sanz FJ, Solana-Manrique C, Torres J, Masiá E, Vicent MJ, Paricio N. A High-Throughput Chemical Screen in DJ-1β Mutant Flies Identifies Zaprinast as a Potential Parkinson's Disease Treatment. Neurotherapeutics 2021; 18:2565-2578. [PMID: 34697772 PMCID: PMC8804136 DOI: 10.1007/s13311-021-01134-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/27/2021] [Indexed: 12/12/2022] Open
Abstract
Dopamine replacement represents the standard therapy for Parkinson's disease (PD), a common, chronic, and incurable neurological disorder; however, this approach only treats the symptoms of this devastating disease. In the search for novel disease-modifying therapies that target other relevant molecular and cellular mechanisms, Drosophila has emerged as a valuable tool to study neurodegenerative diseases due to the presence of a complex central nervous system, the blood-brain barrier, and a similar neurotransmitter profile to humans. Human PD-related genes also display conservation in flies; DJ-1β is the fly ortholog of DJ-1, a gene for which mutations prompt early-onset recessive PD. Interestingly, flies mutant for DJ-1β exhibit PD-related phenotypes, including motor defects, high oxidative stress (OS) levels and metabolic alterations. To identify novel therapies for PD, we performed an in vivo high-throughput screening assay using DJ-1β mutant flies and compounds from the Prestwick® chemical library. Drugs that improved motor performance in DJ-1ß mutant flies were validated in DJ-1-deficient human neural-like cells, revealing that zaprinast displayed the most significant ability to suppress OS-induced cell death. Zaprinast inhibits phosphodiesterases and activates GPR35, an orphan G-protein-coupled receptor not previously associated with PD. We found that zaprinast exerts its beneficial effect in both fly and human PD models through several disease-modifying mechanisms, including reduced OS levels, attenuated apoptosis, increased mitochondrial viability, and enhanced glycolysis. Therefore, our results support zaprinast as a potential therapeutic for PD in future clinical trials.
Collapse
Affiliation(s)
- Francisco José Sanz
- Departamento de Genética, Facultad CC Biológicas, Universidad de Valencia, 46100, Burjassot, Spain
- Instituto Universitario de Biotecnología Y Biomedicina (BIOTECMED), Universidad de Valencia, 46100, Burjassot, Spain
| | - Cristina Solana-Manrique
- Departamento de Genética, Facultad CC Biológicas, Universidad de Valencia, 46100, Burjassot, Spain
- Instituto Universitario de Biotecnología Y Biomedicina (BIOTECMED), Universidad de Valencia, 46100, Burjassot, Spain
| | - Josema Torres
- Departamento de Biología Celular, Biología Funcional Y Antropología Física, Facultad CC Biológicas, Universidad de Valencia, 46100, Burjassot, Spain
| | - Esther Masiá
- Polymer Therapeutics Lab and Screening Platform, Centro de Investigación Príncipe Felipe, 46012, Valencia, Spain
| | - María J Vicent
- Polymer Therapeutics Lab and Screening Platform, Centro de Investigación Príncipe Felipe, 46012, Valencia, Spain
| | - Nuria Paricio
- Departamento de Genética, Facultad CC Biológicas, Universidad de Valencia, 46100, Burjassot, Spain.
- Instituto Universitario de Biotecnología Y Biomedicina (BIOTECMED), Universidad de Valencia, 46100, Burjassot, Spain.
| |
Collapse
|
36
|
Li X, Fetter R, Schwabe T, Jung C, Liu L, Steller H, Gaul U. The cAMP effector PKA mediates Moody GPCR signaling in Drosophila blood-brain barrier formation and maturation. eLife 2021; 10:68275. [PMID: 34382936 PMCID: PMC8390003 DOI: 10.7554/elife.68275] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Accepted: 08/11/2021] [Indexed: 01/01/2023] Open
Abstract
The blood-brain barrier (BBB) of Drosophila comprises a thin epithelial layer of subperineural glia (SPG), which ensheath the nerve cord and insulate it against the potassium-rich hemolymph by forming intercellular septate junctions (SJs). Previously, we identified a novel Gi/Go protein-coupled receptor (GPCR), Moody, as a key factor in BBB formation at the embryonic stage. However, the molecular and cellular mechanisms of Moody signaling in BBB formation and maturation remain unclear. Here, we identify cAMP-dependent protein kinase A (PKA) as a crucial antagonistic Moody effector that is required for the formation, as well as for the continued SPG growth and BBB maintenance in the larva and adult stage. We show that PKA is enriched at the basal side of the SPG cell and that this polarized activity of the Moody/PKA pathway finely tunes the enormous cell growth and BBB integrity. Moody/PKA signaling precisely regulates the actomyosin contractility, vesicle trafficking, and the proper SJ organization in a highly coordinated spatiotemporal manner. These effects are mediated in part by PKA's molecular targets MLCK and Rho1. Moreover, 3D reconstruction of SJ ultrastructure demonstrates that the continuity of individual SJ segments, and not their total length, is crucial for generating a proper paracellular seal. Based on these findings, we propose that polarized Moody/PKA signaling plays a central role in controlling the cell growth and maintaining BBB integrity during the continuous morphogenesis of the SPG secondary epithelium, which is critical to maintain tissue size and brain homeostasis during organogenesis.
Collapse
Affiliation(s)
- Xiaoling Li
- Tianjin Cancer Hospital Airport Hospital, Tianjin Medical University Cancer Institute & Hospital, Tianjin, China.,Department of Biochemistry, Gene Center, Center of Integrated Protein Science (CIPSM), University of Munich, Munich, Germany.,Rockefeller University, New York, United States
| | - Richard Fetter
- Janelia Farm Research Campus, Howard Hughes Medical Institute, Ashburn, United States
| | - Tina Schwabe
- Department of Biochemistry, Gene Center, Center of Integrated Protein Science (CIPSM), University of Munich, Munich, Germany
| | - Christophe Jung
- Department of Biochemistry, Gene Center, Center of Integrated Protein Science (CIPSM), University of Munich, Munich, Germany
| | - Liren Liu
- Department of Gastrointestinal Cancer Biology, Tianjin Medical University Cancer Institute & Hospital; National Clinical Research Center for Cancer; Key Laboratory of Cancer Prevention and Therapy; Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | | | - Ulrike Gaul
- Department of Biochemistry, Gene Center, Center of Integrated Protein Science (CIPSM), University of Munich, Munich, Germany.,Rockefeller University, New York, United States
| |
Collapse
|
37
|
Giong HK, Subramanian M, Yu K, Lee JS. Non-Rodent Genetic Animal Models for Studying Tauopathy: Review of Drosophila, Zebrafish, and C. elegans Models. Int J Mol Sci 2021; 22:8465. [PMID: 34445171 PMCID: PMC8395099 DOI: 10.3390/ijms22168465] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 08/03/2021] [Accepted: 08/04/2021] [Indexed: 12/12/2022] Open
Abstract
Tauopathy refers to a group of progressive neurodegenerative diseases, including frontotemporal lobar degeneration and Alzheimer's disease, which correlate with the malfunction of microtubule-associated protein Tau (MAPT) due to abnormal hyperphosphorylation, leading to the formation of intracellular aggregates in the brain. Despite extensive efforts to understand tauopathy and develop an efficient therapy, our knowledge is still far from complete. To find a solution for this group of devastating diseases, several animal models that mimic diverse disease phenotypes of tauopathy have been developed. Rodents are the dominating tauopathy models because of their similarity to humans and established disease lines, as well as experimental approaches. However, powerful genetic animal models using Drosophila, zebrafish, and C. elegans have also been developed for modeling tauopathy and have contributed to understanding the pathophysiology of tauopathy. The success of these models stems from the short lifespans, versatile genetic tools, real-time in-vivo imaging, low maintenance costs, and the capability for high-throughput screening. In this review, we summarize the main findings on mechanisms of tauopathy and discuss the current tauopathy models of these non-rodent genetic animals, highlighting their key advantages and limitations in tauopathy research.
Collapse
Affiliation(s)
- Hoi-Khoanh Giong
- Disease Target Structure Research Center, KRIBB, 125 Gwahak-ro, Yuseong-gu, Daejeon 34141, Korea; (H.-K.G.); (M.S.)
- KRIBB School, University of Science and Technology, 125 Gwahak-ro, Yuseong-gu, Daejeon 34141, Korea
- Dementia DTC R&D Convergence Program, KIST, Hwarang-ro 14 gil 5, Seongbuk-gu, Seoul 02792, Korea
| | - Manivannan Subramanian
- Disease Target Structure Research Center, KRIBB, 125 Gwahak-ro, Yuseong-gu, Daejeon 34141, Korea; (H.-K.G.); (M.S.)
- Dementia DTC R&D Convergence Program, KIST, Hwarang-ro 14 gil 5, Seongbuk-gu, Seoul 02792, Korea
| | - Kweon Yu
- Disease Target Structure Research Center, KRIBB, 125 Gwahak-ro, Yuseong-gu, Daejeon 34141, Korea; (H.-K.G.); (M.S.)
- KRIBB School, University of Science and Technology, 125 Gwahak-ro, Yuseong-gu, Daejeon 34141, Korea
- Dementia DTC R&D Convergence Program, KIST, Hwarang-ro 14 gil 5, Seongbuk-gu, Seoul 02792, Korea
| | - Jeong-Soo Lee
- Disease Target Structure Research Center, KRIBB, 125 Gwahak-ro, Yuseong-gu, Daejeon 34141, Korea; (H.-K.G.); (M.S.)
- KRIBB School, University of Science and Technology, 125 Gwahak-ro, Yuseong-gu, Daejeon 34141, Korea
- Dementia DTC R&D Convergence Program, KIST, Hwarang-ro 14 gil 5, Seongbuk-gu, Seoul 02792, Korea
| |
Collapse
|
38
|
Benowitz KM, Coleman JM, Allan CW, Matzkin LM. Contributions of cis- and trans-Regulatory Evolution to Transcriptomic Divergence across Populations in the Drosophila mojavensis Larval Brain. Genome Biol Evol 2021; 12:1407-1418. [PMID: 32653899 PMCID: PMC7495911 DOI: 10.1093/gbe/evaa145] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/06/2020] [Indexed: 12/22/2022] Open
Abstract
Natural selection on gene expression was originally predicted to result primarily in cis- rather than trans-regulatory evolution, due to the expectation of reduced pleiotropy. Despite this, numerous studies have ascribed recent evolutionary divergence in gene expression predominantly to trans-regulation. Performing RNA-seq on single isofemale lines from genetically distinct populations of the cactophilic fly Drosophila mojavensis and their F1 hybrids, we recapitulated this pattern in both larval brains and whole bodies. However, we demonstrate that improving the measurement of brain expression divergence between populations by using seven additional genotypes considerably reduces the estimate of trans-regulatory contributions to expression evolution. We argue that the finding of trans-regulatory predominance can result from biases due to environmental variation in expression or other sources of noise, and that cis-regulation is likely a greater contributor to transcriptional evolution across D. mojavensis populations. Lastly, we merge these lines of data to identify several previously hypothesized and intriguing novel candidate genes, and suggest that the integration of regulatory and population-level transcriptomic data can provide useful filters for the identification of potentially adaptive genes.
Collapse
Affiliation(s)
| | - Joshua M Coleman
- Department of Entomology, University of Arizona.,Department of Biological Sciences, University of Alabama in Huntsville
| | | | - Luciano M Matzkin
- Department of Entomology, University of Arizona.,Department of Ecology and Evolutionary Biology, University of Arizona.,BIO5 Institute, University of Arizona
| |
Collapse
|
39
|
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: 10] [Impact Index Per Article: 2.5] [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
|
40
|
Le TD, Inoue YH. Sesamin Activates Nrf2/Cnc-Dependent Transcription in the Absence of Oxidative Stress in Drosophila Adult Brains. Antioxidants (Basel) 2021; 10:antiox10060924. [PMID: 34200419 PMCID: PMC8227698 DOI: 10.3390/antiox10060924] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2021] [Revised: 05/31/2021] [Accepted: 06/04/2021] [Indexed: 11/11/2022] Open
Abstract
Sesamin, a major lignin in sesame seeds, possesses health-promoting properties. Sesamin feeding suppresses several aging-related phenotypes such as age-dependent accumulation of damaged proteins in the muscles and neuronal loss in the brains of Drosophila adults with high levels of reactive oxygen species. Sesamin promotes the transcription of several genes that are responsible for oxidative stress, although the underlying mechanism remains unclear. Here, we aimed to demonstrate that sesamin mediates its action through activation of a transcription factor, Nrf2 (Cnc in Drosophila), essential for anti-aging oxidative stress response. Nrf2/Cnc activation was determined using the antioxidant response element, Green Fluorescence Protein reporter, that can monitor Nrf2/Cnc-dependent transcription. We observed strong fluorescence in the entire bodies, particularly in the abdomens and brains, of adult flies fed sesamin. Interestingly, Nrf2/Cnc was strongly activated in neuronal cells, especially in several neuron types, including glutamatergic and cholinergic, and some dopaminergic and/or serotonergic neurons but not in GABAergic neurons or the mushroom bodies of flies fed sesamin. These results indicate that the anti-aging effects of sesamin are exerted via activation of Nrf2/Cnc-dependent transcription to circumvent oxidative stress accumulation in several types of neurons of adult brains. Sesamin could be explored as a potential dietary supplement for preventing neurodegeneration associated with accumulation of oxidative stress.
Collapse
|
41
|
Hertenstein H, McMullen E, Weiler A, Volkenhoff A, Becker HM, Schirmeier S. Starvation-induced regulation of carbohydrate transport at the blood-brain barrier is TGF-β-signaling dependent. eLife 2021; 10:e62503. [PMID: 34032568 PMCID: PMC8149124 DOI: 10.7554/elife.62503] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Accepted: 04/13/2021] [Indexed: 12/15/2022] Open
Abstract
During hunger or malnutrition, animals prioritize alimentation of the brain over other organs to ensure its function and, thus, their survival. This protection, also-called brain sparing, is described from Drosophila to humans. However, little is known about the molecular mechanisms adapting carbohydrate transport. Here, we used Drosophila genetics to unravel the mechanisms operating at the blood-brain barrier (BBB) under nutrient restriction. During starvation, expression of the carbohydrate transporter Tret1-1 is increased to provide more efficient carbohydrate uptake. Two mechanisms are responsible for this increase. Similar to the regulation of mammalian GLUT4, Rab-dependent intracellular shuttling is needed for Tret1-1 integration into the plasma membrane; even though Tret1-1 regulation is independent of insulin signaling. In addition, starvation induces transcriptional upregulation that is controlled by TGF-β signaling. Considering TGF-β-dependent regulation of the glucose transporter GLUT1 in murine chondrocytes, our study reveals an evolutionarily conserved regulatory paradigm adapting the expression of sugar transporters at the BBB.
Collapse
Affiliation(s)
- Helen Hertenstein
- Department of Biology, Institute of Zoology, Technische Universität DresdenDresdenGermany
| | - Ellen McMullen
- Institut für Neuro- und Verhaltensbiologie, WWU MünsterMünsterGermany
| | - Astrid Weiler
- Department of Biology, Institute of Zoology, Technische Universität DresdenDresdenGermany
| | - Anne Volkenhoff
- Department of Biology, Institute of Zoology, Technische Universität DresdenDresdenGermany
| | - Holger M Becker
- Department of Biology, Institute of Zoology, Technische Universität DresdenDresdenGermany
- Division of General Zoology, Department of Biology, University of KaiserslauternKaiserslauternGermany
| | - Stefanie Schirmeier
- Department of Biology, Institute of Zoology, Technische Universität DresdenDresdenGermany
| |
Collapse
|
42
|
Morpho-Functional Consequences of Swiss Cheese Knockdown in Glia of Drosophila melanogaster. Cells 2021; 10:cells10030529. [PMID: 33801404 PMCID: PMC7998100 DOI: 10.3390/cells10030529] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 02/18/2021] [Accepted: 02/24/2021] [Indexed: 01/15/2023] Open
Abstract
Glia are crucial for the normal development and functioning of the nervous system in many animals. Insects are widely used for studies of glia genetics and physiology. Drosophila melanogaster surface glia (perineurial and subperineurial) form a blood–brain barrier in the central nervous system and blood–nerve barrier in the peripheral nervous system. Under the subperineurial glia layer, in the cortical region of the central nervous system, cortex glia encapsulate neuronal cell bodies, whilst in the peripheral nervous system, wrapping glia ensheath axons of peripheral nerves. Here, we show that the expression of the evolutionarily conserved swiss cheese gene is important in several types of glia. swiss cheese knockdown in subperineurial glia leads to morphological abnormalities of these cells. We found that the number of subperineurial glia nuclei is reduced under swiss cheese knockdown, possibly due to apoptosis. In addition, the downregulation of swiss cheese in wrapping glia causes a loss of its integrity. We reveal transcriptome changes under swiss cheese knockdown in subperineurial glia and in cortex + wrapping glia and show that the downregulation of swiss cheese in these types of glia provokes reactive oxygen species acceleration. These results are accompanied by a decline in animal mobility measured by the negative geotaxis performance assay.
Collapse
|
43
|
Okamoto N, Yamanaka N. Transporter-mediated ecdysteroid trafficking across cell membranes: A novel target for insect growth regulators. JOURNAL OF PESTICIDE SCIENCE 2021; 46:23-28. [PMID: 33746543 PMCID: PMC7953032 DOI: 10.1584/jpestics.d20-071] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Accepted: 12/16/2020] [Indexed: 06/12/2023]
Abstract
Ecdysteroids are a class of steroid hormones in arthropods that control molting and metamorphosis through interaction with intracellular nuclear receptors. In contrast to the extensive literature describing their biosynthetic pathways and signaling components, little has been known about how these hormones are traveling into and out of the cells through lipid bilayers of the cell membranes. Recently, a series of studies conducted in the fruit fly Drosophila melanogaster revealed that membrane transporters have critical functions in trafficking ecdysteroids across cell membranes, challenging the classical simple diffusion model of steroid hormone transport. Here we summarize recent advances in our understanding of membrane transporters involved in ecdysteroid signaling in Drosophila, with particular focus on Ecdysone Importer (EcI) that is involved in ecdysteroid uptake in peripheral tissues. We then discuss the potential advantage of EcI blockers as a novel pest management tool as compared to classical insect growth regulators.
Collapse
Affiliation(s)
- Naoki Okamoto
- Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance, University of Tsukuba, Tsukuba, Ibaraki 305–8577, Japan
| | - Naoki Yamanaka
- Department of Entomology, Institute for Integrative Genome Biology, University of California, Riverside, Riverside, CA 92521, USA
| |
Collapse
|
44
|
Shahid SS, Kerskens CM, Burrows M, Witney AG. Elucidating the complex organization of neural micro-domains in the locust Schistocerca gregaria using dMRI. Sci Rep 2021; 11:3418. [PMID: 33564031 PMCID: PMC7873062 DOI: 10.1038/s41598-021-82187-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Accepted: 01/13/2021] [Indexed: 01/30/2023] Open
Abstract
To understand brain function it is necessary to characterize both the underlying structural connectivity between neurons and the physiological integrity of these connections. Previous research exploring insect brain connectivity has typically used electron microscopy techniques, but this methodology cannot be applied to living animals and so cannot be used to understand dynamic physiological processes. The relatively large brain of the desert locust, Schistercera gregaria (Forksȧl) is ideal for exploring a novel methodology; micro diffusion magnetic resonance imaging (micro-dMRI) for the characterization of neuronal connectivity in an insect brain. The diffusion-weighted imaging (DWI) data were acquired on a preclinical system using a customised multi-shell diffusion MRI scheme optimized to image the locust brain. Endogenous imaging contrasts from the averaged DWIs and Diffusion Kurtosis Imaging (DKI) scheme were applied to classify various anatomical features and diffusion patterns in neuropils, respectively. The application of micro-dMRI modelling to the locust brain provides a novel means of identifying anatomical regions and inferring connectivity of large tracts in an insect brain. Furthermore, quantitative imaging indices derived from the kurtosis model that include fractional anisotropy (FA), mean diffusivity (MD) and kurtosis anisotropy (KA) can be extracted. These metrics could, in future, be used to quantify longitudinal structural changes in the nervous system of the locust brain that occur due to environmental stressors or ageing.
Collapse
Affiliation(s)
- Syed Salman Shahid
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Christian M Kerskens
- Trinity College Institute of Neuroscience, Trinity Centre for Biomedical Engineering, School of Medicine, Trinity College Dublin, Dublin 2, Ireland
| | - Malcolm Burrows
- Department of Zoology, University of Cambridge, Cambridge, UK
| | - Alice G Witney
- Department of Physiology, School of Medicine, Trinity Biomedical Sciences Institute, Trinity Centre for Biomedical Engineering, Trinity College Institute of Neuroscience, Trinity College Dublin, Dublin 2, Ireland.
| |
Collapse
|
45
|
McMullen E, Weiler A, Becker HM, Schirmeier S. Plasticity of Carbohydrate Transport at the Blood-Brain Barrier. Front Behav Neurosci 2021; 14:612430. [PMID: 33551766 PMCID: PMC7863721 DOI: 10.3389/fnbeh.2020.612430] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 12/17/2020] [Indexed: 12/21/2022] Open
Abstract
Neuronal function is highly energy demanding, requiring efficient transport of nutrients into the central nervous system (CNS). Simultaneously the brain must be protected from the influx of unwanted solutes. Most of the energy is supplied from dietary sugars, delivered from circulation via the blood-brain barrier (BBB). Therefore, selective transporters are required to shuttle metabolites into the nervous system where they can be utilized. The Drosophila BBB is formed by perineural and subperineurial glial cells, which effectively separate the brain from the surrounding hemolymph, maintaining a constant microenvironment. We identified two previously unknown BBB transporters, MFS3 (Major Facilitator Superfamily Transporter 3), located in the perineurial glial cells, and Pippin, found in both the perineurial and subperineurial glial cells. Both transporters facilitate uptake of circulating trehalose and glucose into the BBB-forming glial cells. RNA interference-mediated knockdown of these transporters leads to pupal lethality. However, null mutants reach adulthood, although they do show reduced lifespan and activity. Here, we report that both carbohydrate transport efficiency and resulting lethality found upon loss of MFS3 or Pippin are rescued via compensatory upregulation of Tret1-1, another BBB carbohydrate transporter, in Mfs3 and pippin null mutants, while RNAi-mediated knockdown is not compensated for. This means that the compensatory mechanisms in place upon mRNA degradation following RNA interference can be vastly different from those resulting from a null mutation.
Collapse
Affiliation(s)
- Ellen McMullen
- Department of Biology, Institute of Zoology, Technische Universität Dresden, Dresden, Germany
| | - Astrid Weiler
- Department of Biology, Institute of Zoology, Technische Universität Dresden, Dresden, Germany
| | - Holger M. Becker
- Division of General Zoology, Department of Biology, University of Kaiserslautern, Kaiserslautern, Germany
| | - Stefanie Schirmeier
- Department of Biology, Institute of Zoology, Technische Universität Dresden, Dresden, Germany
| |
Collapse
|
46
|
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: 23] [Impact Index Per Article: 4.6] [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
|
47
|
Banfill CR, Wilson ACC, Lu HL. Further evidence that mechanisms of host/symbiont integration are dissimilar in the maternal versus embryonic Acyrthosiphon pisum bacteriome. EvoDevo 2020; 11:23. [PMID: 33292476 PMCID: PMC7654044 DOI: 10.1186/s13227-020-00168-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Accepted: 10/29/2020] [Indexed: 03/29/2023] Open
Abstract
Background Host/symbiont integration is a signature of evolutionarily ancient, obligate endosymbioses. However, little is known about the cellular and developmental mechanisms of host/symbiont integration at the molecular level. Many insects possess obligate bacterial endosymbionts that provide essential nutrients. To advance understanding of the developmental and metabolic integration of hosts and endosymbionts, we track the localization of a non-essential amino acid transporter, ApNEAAT1, across asexual embryogenesis in the aphid, Acyrthosiphon pisum. Previous work in adult bacteriomes revealed that ApNEAAT1 functions to exchange non-essential amino acids at the A. pisum/Buchnera aphidicola symbiotic interface. Driven by amino acid concentration gradients, ApNEAAT1 moves proline, serine, and alanine from A. pisum to Buchnera and cysteine from Buchnera to A. pisum. Here, we test the hypothesis that ApNEAAT1 is localized to the symbiotic interface during asexual embryogenesis. Results During A. pisum asexual embryogenesis, ApNEAAT1 does not localize to the symbiotic interface. We observed ApNEAAT1 localization to the maternal follicular epithelium, the germline, and, in late-stage embryos, to anterior neural structures and insect immune cells (hemocytes). We predict that ApNEAAT1 provisions non-essential amino acids to developing oocytes and embryos, as well as to the brain and related neural structures. Additionally, ApNEAAT1 may perform roles related to host immunity. Conclusions Our work provides further evidence that the embryonic and adult bacteriomes of asexual A. pisum are not equivalent. Future research is needed to elucidate the developmental time point at which the bacteriome reaches maturity.
Collapse
Affiliation(s)
- Celeste R Banfill
- Department of Biology, University of Miami, Coral Gables, FL, 33146, USA
| | - Alex C C Wilson
- Department of Biology, University of Miami, Coral Gables, FL, 33146, USA.
| | - Hsiao-Ling Lu
- Department of Biotechnology, National Formosa University, Huwei, Taiwan.
| |
Collapse
|
48
|
Solomatina AI, Slobodina AD, Ryabova EV, Bolshakova OI, Chelushkin PS, Sarantseva SV, Tunik SP. Blood-Brain Barrier Penetrating Luminescent Conjugates Based on Cyclometalated Platinum(II) Complexes. Bioconjug Chem 2020; 31:2628-2637. [DOI: 10.1021/acs.bioconjchem.0c00542] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Affiliation(s)
| | - Aleksandra D. Slobodina
- Petersburg Nuclear Physics Institute named by B.P. Konstantinov of National Research Centre ≪Kurchatov Institute≫, Gatchina 188300, Russia
| | - Elena V. Ryabova
- Petersburg Nuclear Physics Institute named by B.P. Konstantinov of National Research Centre ≪Kurchatov Institute≫, Gatchina 188300, Russia
| | - Olga I. Bolshakova
- Petersburg Nuclear Physics Institute named by B.P. Konstantinov of National Research Centre ≪Kurchatov Institute≫, Gatchina 188300, Russia
| | - Pavel S. Chelushkin
- Institute of Chemistry, St. Petersburg State University, St. Petersburg 198504, Russia
| | - Svetlana V. Sarantseva
- Petersburg Nuclear Physics Institute named by B.P. Konstantinov of National Research Centre ≪Kurchatov Institute≫, Gatchina 188300, Russia
| | - Sergey P. Tunik
- Institute of Chemistry, St. Petersburg State University, St. Petersburg 198504, Russia
| |
Collapse
|
49
|
Saikumar J, Byrns CN, Hemphill M, Meaney DF, Bonini NM. Dynamic neural and glial responses of a head-specific model for traumatic brain injury in Drosophila. Proc Natl Acad Sci U S A 2020; 117:17269-17277. [PMID: 32611818 PMCID: PMC7382229 DOI: 10.1073/pnas.2003909117] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Traumatic brain injury (TBI) is the strongest environmental risk factor for the accelerated development of neurodegenerative diseases. There are currently no therapeutics to address this due to lack of insight into mechanisms of injury progression, which are challenging to study in mammalian models. Here, we have developed and extensively characterized a head-specific approach to TBI in Drosophila, a powerful genetic system that shares many conserved genes and pathways with humans. The Drosophila TBI (dTBI) device inflicts mild, moderate, or severe brain trauma by precise compression of the head using a piezoelectric actuator. Head-injured animals display features characteristic of mammalian TBI, including severity-dependent ataxia, life span reduction, and brain degeneration. Severe dTBI is associated with cognitive decline and transient glial dysfunction, and stimulates antioxidant, proteasome, and chaperone activity. Moreover, genetic or environmental augmentation of the stress response protects from severe dTBI-induced brain degeneration and life span deficits. Together, these findings present a tunable, head-specific approach for TBI in Drosophila that recapitulates mammalian injury phenotypes and underscores the ability of the stress response to mitigate TBI-induced brain degeneration.
Collapse
Affiliation(s)
- Janani Saikumar
- Department of Biology, University of Pennsylvania, Philadelphia, PA 19104
| | - China N Byrns
- Neuroscience Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Matthew Hemphill
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104
| | - David F Meaney
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104
- Department of Neurosurgery, University of Pennsylvania, Philadelphia, PA 19104
| | - Nancy M Bonini
- Department of Biology, University of Pennsylvania, Philadelphia, PA 19104;
- Neuroscience Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104
| |
Collapse
|
50
|
Abstract
While neurons and circuits are almost unequivocally considered to be the computational units and actuators of behavior, a complete understanding of the nervous system must incorporate glial cells. Far beyond a copious but passive substrate, glial influence is inextricable from neuronal physiology, whether during developmental guidance and synaptic shaping or through the trophic support, neurotransmitter and ion homeostasis, cytokine signaling and immune function, and debris engulfment contributions that this class provides throughout an organism's life. With such essential functions, among a growing literature of nuanced roles, it follows that glia are consequential to behavior in adult animals, with novel genetic tools allowing for the investigation of these phenomena in living organisms. We discuss here the relevance of glia for maintaining circadian rhythms and also for serving functions of sleep.
Collapse
Affiliation(s)
- Gregory Artiushin
- Chronobiology and Sleep Institute, Perelman School of Medicine, and Howard Hughes Medical Institute, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA;
| | - Amita Sehgal
- Chronobiology and Sleep Institute, Perelman School of Medicine, and Howard Hughes Medical Institute, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA;
| |
Collapse
|