1
|
Castranova D, Kenton MI, Kraus A, Dell CW, Park JS, Venero Galanternik M, Park G, Lumbantobing DN, Dye L, Marvel M, Iben J, Taimatsu K, Pham V, Willms RJ, Blevens L, Robertson TF, Hou Y, Huttenlocher A, Foley E, Parenti LR, Frazer JK, Narayan K, Weinstein BM. The axillary lymphoid organ is an external, experimentally accessible immune organ in the zebrafish. J Exp Med 2025; 222:e20241435. [PMID: 40167600 PMCID: PMC11960710 DOI: 10.1084/jem.20241435] [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: 08/12/2024] [Revised: 01/13/2025] [Accepted: 03/03/2025] [Indexed: 04/02/2025] Open
Abstract
Lymph nodes and other secondary lymphoid organs play critical roles in immune surveillance and immune activation in mammals, but the deep internal locations of these organs make it challenging to image and study them in living animals. Here, we describe a previously uncharacterized external immune organ in the zebrafish ideally suited for studying immune cell dynamics in vivo, the axillary lymphoid organ (ALO). This small, translucent organ has an outer cortex teeming with immune cells, an inner medulla with a mesh-like network of fibroblastic reticular cells along which immune cells migrate, and a network of lymphatic vessels draining to a large adjacent lymph sac. Noninvasive high-resolution imaging of transgenically marked immune cells can be carried out in ALOs of living animals, which are readily accessible to external treatment. This newly discovered tissue provides a superb model for dynamic live imaging of immune cells and their interaction with pathogens and surrounding tissues, including blood and lymphatic vessels.
Collapse
Affiliation(s)
- Daniel Castranova
- Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Madeleine I. Kenton
- Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Aurora Kraus
- Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Christopher W. Dell
- Center for Molecular Microscopy, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
- Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Jong S. Park
- Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Marina Venero Galanternik
- Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Gilseung Park
- Section of Pediatric Hematology-Oncology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Daniel N. Lumbantobing
- Division of Fishes, Department of Vertebrate Zoology, National Museum of Natural History, Smithsonian Institution, Washington, DC, USA
| | - Louis Dye
- Microscopy and Imaging Core, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Miranda Marvel
- Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - James Iben
- Molecular Genomics Core, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Kiyohito Taimatsu
- Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Van Pham
- Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Reegan J. Willms
- Department of Medical Microbiology and Immunology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada
| | - Lucas Blevens
- Section of Pediatric Hematology-Oncology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Tanner F. Robertson
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, WI, USA
| | - Yiran Hou
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, WI, USA
| | - Anna Huttenlocher
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, WI, USA
| | - Edan Foley
- Department of Medical Microbiology and Immunology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada
| | - Lynne R. Parenti
- Division of Fishes, Department of Vertebrate Zoology, National Museum of Natural History, Smithsonian Institution, Washington, DC, USA
| | - J. Kimble Frazer
- Section of Pediatric Hematology-Oncology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Kedar Narayan
- Center for Molecular Microscopy, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
- Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Brant M. Weinstein
- Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
2
|
Wang Y, Pang X, Li R, Chen J, Wen C, Zhu H, Long T, Li J, Zheng L, Deng Y, Zheng J, Xu B. Mesenchymal Stem Cells Carrying Viral Fusogenic Protein p14 to Treat Solid Tumors by Inducing Cell-Cell Fusion and Immune Activation. RESEARCH (WASHINGTON, D.C.) 2025; 8:0594. [PMID: 39872128 PMCID: PMC11770199 DOI: 10.34133/research.0594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 12/30/2024] [Accepted: 01/02/2025] [Indexed: 01/29/2025]
Abstract
Background: Chimeric antigen receptor (CAR)-based immune cell therapies attack neighboring cancer cells after receptor recognition but are unable to directly affect distant tumor cells. This limitation may contribute to their inefficiency in treating solid tumors, given the restricted intratumoral infiltration and immunosuppressive tumor microenvironment. Therefore, cell-cell fusion as a cell-killing mechanism might develop a novel cytotherapy aimed at improving the efficacy against solid tumors. Methods: We constructed a fusogenic protein, fusion-associated small transmembrane (FAST) p14 of reptilian reovirus, into cancer cells and mesenchymal stem cells (MSCs), which cocultured with various colon cancer cells and melenoma cells to validate its ability to induce cell fusion and syncytia formation. RNA sequencing, quantitative reverse transcription polymerase chain reaction, and Western blot were performed to elucidate the mechanism of syncytia death. Cell viability assay was employed to assess the killing effects of MSCs carrying the p14 protein (MSCs-p14), which was also identified in the subcutaneous tumor models. Subsequently, the Tet-On system was introduced to enhance the controllability and safety of therapy. Results: Cancer cells incorporated with fusogenic protein p14 FAST from reovirus fused together to form syncytia and subsequently died through apoptosis and pyroptosis. MSCs-p14 cocultured with different cancer cells and effienctly induced cancer cell fusion and caused widespread cancer cell death in vitro. In mouse tumor models, mMSCs-p14 treatment markedly suppressed tumor growth and also enhanced the activity of natural killer cells and macrophages. Controllability and safety of MSCs-p14 therapy were further improved by introducing the tetracycline-controlled transcriptional system. Conclusion: MSC-based cytotherapy carrying viral fusogenic protein in this study kills cancer cells by inducing cell-cell fusion. It has demonstrated definite efficacy in treating solid tumors and is worth considering for clinical development.
Collapse
Affiliation(s)
- Yao Wang
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
- Center of Clinical Oncology,
The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221002, China
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy,
Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Xunlei Pang
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
- Department of Gastroenterology,
The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Ruirui Li
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
- Center of Clinical Oncology,
The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221002, China
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy,
Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Jiuzhou Chen
- Department of Radiation Oncology, the Affiliated Huaian No.1 People’s Hospital of Nanjing Medical University, Huaian, Jiangsu, China
| | - Chen Wen
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
- Center of Clinical Oncology,
The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221002, China
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy,
Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Huihuang Zhu
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
- Center of Clinical Oncology,
The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221002, China
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy,
Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Tingyu Long
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
- Center of Clinical Oncology,
The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221002, China
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy,
Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Jianjie Li
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
- Center of Clinical Oncology,
The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221002, China
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy,
Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Lijun Zheng
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
- Center of Clinical Oncology,
The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221002, China
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy,
Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Youcai Deng
- Department of Clinical Hematology,
College of Pharmacy and Laboratory Medicine Science, Army Medical University, Chongqing 400038, China
| | - Junnian Zheng
- Center of Clinical Oncology,
The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221002, China
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy,
Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Bo Xu
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
- Center of Clinical Oncology,
The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221002, China
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy,
Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| |
Collapse
|
3
|
Castranova D, Kenton MI, Kraus A, Dell CW, Park JS, Galanternik MV, Park G, Lumbantobing DN, Dye L, Marvel M, Iben J, Taimatsu K, Pham V, Willms RJ, Blevens L, Robertson TF, Hou Y, Huttenlocher A, Foley E, Parenti LR, Frazer JK, Narayan K, Weinstein BM. The axillary lymphoid organ - an external, experimentally accessible immune organ in the zebrafish. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.25.605139. [PMID: 39091802 PMCID: PMC11291151 DOI: 10.1101/2024.07.25.605139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/04/2024]
Abstract
Lymph nodes and other secondary lymphoid organs play critical roles in immune surveillance and immune activation in mammals, but the deep internal locations of these organs make it challenging to image and study them in living animals. Here, we describe a previously uncharacterized external immune organ in the zebrafish ideally suited for studying immune cell dynamics in vivo, the axillary lymphoid organ (ALO). This small, translucent organ has an outer cortex teeming with immune cells, an inner medulla with a mesh-like network of fibroblastic reticular cells along which immune cells migrate, and a network of lymphatic vessels draining to a large adjacent lymph sac. Noninvasive high-resolution imaging of transgenically marked immune cells can be carried out in the lobes of living animals, and the ALO is readily accessible to external treatment. This newly discovered tissue provides a superb model for dynamic live imaging of immune cells and their interaction with pathogens and surrounding tissues, including blood and lymphatic vessels.
Collapse
Affiliation(s)
- Daniel Castranova
- Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD 20814, USA
| | - Madeleine I. Kenton
- Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD 20814, USA
| | - Aurora Kraus
- Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD 20814, USA
| | - Christopher W. Dell
- Center for Molecular Microscopy, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA and Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA
| | - Jong S. Park
- Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD 20814, USA
| | - Marina Venero Galanternik
- Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD 20814, USA
| | - Gilseung Park
- Section of Pediatric Hematology-Oncology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Daniel N. Lumbantobing
- Division of Fishes, Department of Vertebrate Zoology, National Museum of Natural History, Smithsonian Institution, Washington, DC 20560, USA
| | - Louis Dye
- Microscopy and Imaging Core, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD 20814, USA
| | - Miranda Marvel
- Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD 20814, USA
| | - James Iben
- Molecular Genomics Core, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD 20814, USA
| | - Kiyohito Taimatsu
- Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD 20814, USA
| | - Van Pham
- Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD 20814, USA
| | - Reegan J. Willms
- Department of Medical Microbiology and Immunology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Lucas Blevens
- Section of Pediatric Hematology-Oncology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Tanner F. Robertson
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, WI 53706
| | - Yiran Hou
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, WI 53706
| | - Anna Huttenlocher
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, WI 53706
| | - Edan Foley
- Department of Medical Microbiology and Immunology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Lynne R. Parenti
- Division of Fishes, Department of Vertebrate Zoology, National Museum of Natural History, Smithsonian Institution, Washington, DC 20560, USA
| | - J. Kimble Frazer
- Section of Pediatric Hematology-Oncology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Kedar Narayan
- Center for Molecular Microscopy, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA and Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA
| | - Brant M. Weinstein
- Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD 20814, USA
| |
Collapse
|
4
|
Zhao Y, Huang CX, Gu Y, Zhao Y, Ren W, Wang Y, Chen J, Guan NN, Song J. Serotonergic modulation of vigilance states in zebrafish and mice. Nat Commun 2024; 15:2596. [PMID: 38519480 PMCID: PMC10959952 DOI: 10.1038/s41467-024-47021-0] [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: 10/22/2023] [Accepted: 03/12/2024] [Indexed: 03/25/2024] Open
Abstract
Vigilance refers to being alertly watchful or paying sustained attention to avoid potential threats. Animals in vigilance states reduce locomotion and have an enhanced sensitivity to aversive stimuli so as to react quickly to dangers. Here we report that an unconventional 5-HT driven mechanism operating at neural circuit level which shapes the internal state underlying vigilance behavior in zebrafish and male mice. The neural signature of internal vigilance state was characterized by persistent low-frequency high-amplitude neuronal synchrony in zebrafish dorsal pallium and mice prefrontal cortex. The neuronal synchronization underlying vigilance was dependent on intense release of 5-HT induced by persistent activation of either DRN 5-HT neuron or local 5-HT axon terminals in related brain regions via activation of 5-HTR7. Thus, we identify a mechanism of vigilance behavior across species that illustrates the interplay between neuromodulators and neural circuits necessary to shape behavior states.
Collapse
Affiliation(s)
- Yang Zhao
- Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, 200434, China
- Clinical Center for Brain and Spinal Cord Research, Tongji University, 200092, Shanghai, China
| | - Chun-Xiao Huang
- Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, 200434, China
- Clinical Center for Brain and Spinal Cord Research, Tongji University, 200092, Shanghai, China
| | - Yiming Gu
- Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, 200434, China
- Clinical Center for Brain and Spinal Cord Research, Tongji University, 200092, Shanghai, China
| | - Yacong Zhao
- Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, 200434, China
- Clinical Center for Brain and Spinal Cord Research, Tongji University, 200092, Shanghai, China
| | - Wenjie Ren
- Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, 200434, China
- Clinical Center for Brain and Spinal Cord Research, Tongji University, 200092, Shanghai, China
| | - Yutong Wang
- Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, 200434, China
- Clinical Center for Brain and Spinal Cord Research, Tongji University, 200092, Shanghai, China
| | - Jinjin Chen
- Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, 200434, China
- Clinical Center for Brain and Spinal Cord Research, Tongji University, 200092, Shanghai, China
| | - Na N Guan
- Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, 200434, China.
- Clinical Center for Brain and Spinal Cord Research, Tongji University, 200092, Shanghai, China.
- Frontiers Science Center for Intelligent Autonomous Systems, Tongji University, Shanghai, China.
- Department of Neuroscience, Karolinska Institutet, 171 77, Stockholm, Sweden.
| | - Jianren Song
- Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, 200434, China.
- Clinical Center for Brain and Spinal Cord Research, Tongji University, 200092, Shanghai, China.
- Frontiers Science Center for Intelligent Autonomous Systems, Tongji University, Shanghai, China.
- Department of Neuroscience, Karolinska Institutet, 171 77, Stockholm, Sweden.
| |
Collapse
|
5
|
Wisenden BD, Anderson CM, Hanson KA, Johnson MIM, Stockwell CA. Acquired predator recognition via epidermal alarm cues but not dietary alarm cues by isolated pupfish. ROYAL SOCIETY OPEN SCIENCE 2023; 10:230444. [PMID: 37711143 PMCID: PMC10498034 DOI: 10.1098/rsos.230444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Accepted: 08/21/2023] [Indexed: 09/16/2023]
Abstract
We tested whether Shoshone pupfish Cyprinodon nevadensis shoshone and Amargosa River pupfish C. n. amargosae respond behaviourally to conspecific chemical alarm cues released when epidermal tissue is damaged by a predator. We found that both subspecies reduced activity and vertical position in the water column in response to alarm cues. We then tested if pupfish can use alarm cue to acquire recognition of a novel predator. We trained pupfish with (1) water + odour of largemouth bass fed a diet of earthworms, (2) alarm cues from skin extract (epidermal alarm cues) + odour of bass fed a diet of earthworms, or (3) water + odour of bass fed a diet of pupfish (dietary alarm cues). Pupfish responded to epidermal alarm cues but not to dietary alarm cues. Pupfish were retested with the odour of bass that were fed an earthworm diet. Pupfish that had previously received epidermal alarm cues reduced vertical position and activity relative to the other two treatments. This is the first demonstration of acquired recognition of a novel predator by a pupfish, the first report of partial predator naiveté, and opens the possibility of predator-recognition training as a tool for management and conservation of endangered desert fishes.
Collapse
Affiliation(s)
- Brian D. Wisenden
- Biosciences Department, Minnesota State University Moorhead, Moorhead, MN 56563, USA
| | - Cody M. Anderson
- Environmental & Conservation Sciences Graduate Program, North Dakota State University, Fargo, ND 58108, USA
| | - Kathryn A. Hanson
- Biosciences Department, Minnesota State University Moorhead, Moorhead, MN 56563, USA
| | - Molly I. M. Johnson
- Biosciences Department, Minnesota State University Moorhead, Moorhead, MN 56563, USA
- Environmental & Conservation Sciences Graduate Program, North Dakota State University, Fargo, ND 58108, USA
| | - Craig A. Stockwell
- Biological Sciences Department, North Dakota State University, Fargo, ND 58108, USA
| |
Collapse
|
6
|
Kraus A, Garcia B, Ma J, Herrera KJ, Zwaka H, Harpaz R, Wong RY, Engert F, Salinas I. Olfactory detection of viruses shapes brain immunity and behavior in zebrafish. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.17.533129. [PMID: 37034630 PMCID: PMC10081220 DOI: 10.1101/2023.03.17.533129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/19/2023]
Abstract
Olfactory sensory neurons (OSNs) are constantly exposed to pathogens, including viruses. However, serious brain infection via the olfactory route rarely occurs. When OSNs detect a virus, they coordinate local antiviral immune responses to stop virus progression to the brain. Despite effective immune control in the olfactory periphery, pathogen-triggered neuronal signals reach the CNS via the olfactory bulb (OB). We hypothesized that neuronal detection of a virus by OSNs initiates neuroimmune responses in the OB that prevent pathogen invasion. Using zebrafish ( Danio rerio ) as a model, we demonstrate viral-specific neuronal activation of OSNs projecting into the OB, indicating that OSNs are electrically activated by viruses. Further, behavioral changes are seen in both adult and larval zebrafish after viral exposure. By profiling the transcription of single cells in the OB after OSNs are exposed to virus, we found that both microglia and neurons enter a protective state. Microglia and macrophage populations in the OB respond within minutes of nasal viral delivery followed decreased expression of neuronal differentiation factors and enrichment of genes in the neuropeptide signaling pathway in neuronal clusters. Pituitary adenylate-cyclase-activating polypeptide ( pacap ), a known antimicrobial, was especially enriched in a neuronal cluster. We confirm that PACAP is antiviral in vitro and that PACAP expression increases in the OB 1 day post-viral treatment. Our work reveals how encounters with viruses in the olfactory periphery shape the vertebrate brain by inducing antimicrobial programs in neurons and by altering host behavior.
Collapse
|
7
|
Shainer I, Kuehn E, Laurell E, Al Kassar M, Mokayes N, Sherman S, Larsch J, Kunst M, Baier H. A single-cell resolution gene expression atlas of the larval zebrafish brain. SCIENCE ADVANCES 2023; 9:eade9909. [PMID: 36812331 PMCID: PMC9946346 DOI: 10.1126/sciadv.ade9909] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Accepted: 12/22/2022] [Indexed: 06/18/2023]
Abstract
The advent of multimodal brain atlases promises to accelerate progress in neuroscience by allowing in silico queries of neuron morphology, connectivity, and gene expression. We used multiplexed fluorescent in situ RNA hybridization chain reaction (HCR) technology to generate expression maps across the larval zebrafish brain for a growing set of marker genes. The data were registered to the Max Planck Zebrafish Brain (mapzebrain) atlas, thus allowing covisualization of gene expression, single-neuron tracings, and expertly curated anatomical segmentations. Using post hoc HCR labeling of the immediate early gene cfos, we mapped responses to prey stimuli and food ingestion across the brain of freely swimming larvae. This unbiased approach revealed, in addition to previously described visual and motor areas, a cluster of neurons in the secondary gustatory nucleus, which express the marker calb2a, as well as a specific neuropeptide Y receptor, and project to the hypothalamus. This discovery exemplifies the power of this new atlas resource for zebrafish neurobiology.
Collapse
|
8
|
Neuromasts and Olfactory Organs of Zebrafish Larvae Represent Possible Sites of SARS-CoV-2 Pseudovirus Host Cell Entry. J Virol 2022; 96:e0141822. [PMID: 36448804 PMCID: PMC9769390 DOI: 10.1128/jvi.01418-22] [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] [Indexed: 12/02/2022] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the causative agent of the acute respiratory disease coronavirus disease 2019 (COVID-19), which has resulted in millions of deaths globally. Here, we explored the mechanism of host cell entry of a luciferase-ZsGreen spike (SARS-CoV-2)-pseudotyped lentivirus using zebrafish embryos/larvae as an in vivo model. Successful pseudovirus entry was demonstrated via the expression of the luciferase (luc) gene, which was validated by reverse transcription-PCR (RT-PCR). Treatment of larvae with chloroquine (a broad-spectrum viral inhibitor that blocks membrane fusion) or bafilomycin A1 (a specific inhibitor of vacuolar proton ATPases, which blocks endolysosomal trafficking) significantly reduced luc expression, indicating the possible involvement of the endolysosomal system in the viral entry mechanism. The pharmacological inhibition of two-pore channel (TPC) activity or use of the tpcn2dhkz1a mutant zebrafish line also led to diminished luc expression. The localized expression of ACE2 and TPC2 in the anterior neuromasts and the forming olfactory organs was demonstrated, and the occurrence of endocytosis in both locations was confirmed. Together, our data indicate that zebrafish embryos/larvae are a viable and tractable model to explore the mechanism of SARS-CoV-2 host cell entry, that the peripheral sense organs are a likely site for viral host cell entry, and that TPC2 plays a key role in the translocation of the virus through the endolysosomal system. IMPORTANCE Despite the development of effective vaccines to combat the COVID-19 pandemic, which help prevent the most life-threatening symptoms, full protection cannot be guaranteed, especially with the emergence of new viral variants. Moreover, some resistance to vaccination remains in certain age groups and cultures. As such, there is an urgent need for the development of new strategies and therapies to help combat this deadly disease. Here, we provide compelling evidence that the peripheral sensory organs of zebrafish possess several key components required for SARS-CoV-2 host cell entry. The nearly transparent larvae provide a most amenable complementary platform to investigate the key steps of viral entry into host cells, as well as its spread through the tissues and organs. This will help in the identification of key viral entry steps for therapeutic intervention, provide an inexpensive model for screening novel antiviral compounds, and assist in the development of new and more effective vaccines.
Collapse
|
9
|
Levraud JP, Rawls JF, Clatworthy AE. Using zebrafish to understand reciprocal interactions between the nervous and immune systems and the microbial world. J Neuroinflammation 2022; 19:170. [PMID: 35765004 PMCID: PMC9238045 DOI: 10.1186/s12974-022-02506-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Accepted: 06/01/2022] [Indexed: 11/10/2022] Open
Abstract
Animals rely heavily on their nervous and immune systems to perceive and survive within their environment. Despite the traditional view of the brain as an immunologically privileged organ, these two systems interact with major consequences. Furthermore, microorganisms within their environment are major sources of stimuli and can establish relationships with animal hosts that range from pathogenic to mutualistic. Research from a variety of human and experimental animal systems are revealing that reciprocal interactions between microbiota and the nervous and immune systems contribute significantly to normal development, homeostasis, and disease. The zebrafish has emerged as an outstanding model within which to interrogate these interactions due to facile genetic and microbial manipulation and optical transparency facilitating in vivo imaging. This review summarizes recent studies that have used the zebrafish for analysis of bidirectional control between the immune and nervous systems, the nervous system and the microbiota, and the microbiota and immune system in zebrafish during development that promotes homeostasis between these systems. We also describe how the zebrafish have contributed to our understanding of the interconnections between these systems during infection in fish and how perturbations may result in pathology.
Collapse
Affiliation(s)
- Jean-Pierre Levraud
- Université Paris-Saclay, CNRS, Institut Pasteur, Université Paris-Cité, Institut des Neurosciences Paris-Saclay, 91400, Saclay, France.
| | - John F. Rawls
- grid.26009.3d0000 0004 1936 7961Department of Molecular Genetics and Microbiology, Duke Microbiome Center, Duke University School of Medicine, 213 Research Drive, Durham, NC 27710 USA
| | - Anne E. Clatworthy
- grid.66859.340000 0004 0546 1623Broad Institute of MIT and Harvard, 415 Main Street, Cambridge, MA 02142 USA ,grid.32224.350000 0004 0386 9924Department of Molecular Biology and Center for Computational and Integrative Biology, Massachusetts General Hospital, 185 Cambridge Street, Boston, MA 02114 USA
| |
Collapse
|
10
|
Tan JXM, Ang RJW, Wee CL. Larval Zebrafish as a Model for Mechanistic Discovery in Mental Health. Front Mol Neurosci 2022; 15:900213. [PMID: 35813062 PMCID: PMC9263853 DOI: 10.3389/fnmol.2022.900213] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2022] [Accepted: 04/25/2022] [Indexed: 12/23/2022] Open
Abstract
Animal models are essential for the discovery of mechanisms and treatments for neuropsychiatric disorders. However, complex mental health disorders such as depression and anxiety are difficult to fully recapitulate in these models. Borrowing from the field of psychiatric genetics, we reiterate the framework of 'endophenotypes' - biological or behavioral markers with cellular, molecular or genetic underpinnings - to reduce complex disorders into measurable behaviors that can be compared across organisms. Zebrafish are popular disease models due to the conserved genetic, physiological and anatomical pathways between zebrafish and humans. Adult zebrafish, which display more sophisticated behaviors and cognition, have long been used to model psychiatric disorders. However, larvae (up to 1 month old) are more numerous and also optically transparent, and hence are particularly suited for high-throughput screening and brain-wide neural circuit imaging. A number of behavioral assays have been developed to quantify neuropsychiatric phenomena in larval zebrafish. Here, we will review these assays and the current knowledge regarding the underlying mechanisms of their behavioral readouts. We will also discuss the existing evidence linking larval zebrafish behavior to specific human behavioral traits and how the endophenotype framework can be applied. Importantly, many of the endophenotypes we review do not solely define a diseased state but could manifest as a spectrum across the general population. As such, we make the case for larval zebrafish as a promising model for extending our understanding of population mental health, and for identifying novel therapeutics and interventions with broad impact.
Collapse
Affiliation(s)
| | | | - Caroline Lei Wee
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| |
Collapse
|
11
|
Jesuthasan S, Krishnan S, Cheng RK, Mathuru A. Neural correlates of state transitions elicited by a chemosensory danger cue. Prog Neuropsychopharmacol Biol Psychiatry 2021; 111:110110. [PMID: 32950538 DOI: 10.1016/j.pnpbp.2020.110110] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 09/04/2020] [Accepted: 09/11/2020] [Indexed: 01/13/2023]
Abstract
BACKGROUND Detection of predator cues changes the brain state in prey species and helps them avoid danger. Dysfunctionality in changing the central state appropriately in stressful situations is proposed to be an underlying cause of multiple psychiatric disorders in humans. METHODS Here, we investigate the dynamics of neural circuits mediating response to a threat, to characterize these states and to identify potential control networks. We use resonant scanning 2-photon microscopy for in vivo brain-wide imaging and custom designed behavioral assays for the study. RESULTS We first show that 5-7 day old zebrafish larvae react to an alarm pheromone (Schreckstoff) with reduced mobility. They subsequently display heightened vigilance, as evidenced by increased dark avoidance. Calcium imaging indicates that exposure to Schreckstoff elicits stimulus-locked activity in olfactory sensory neurons innervating a lateral glomerulus and in telencephalic regions including the putative medial amygdala and entopeduncular nucleus. Sustained activity outlasting the stimulus delivery was detected in regions regulating neuromodulator release, including the lateral habenula, posterior tuberculum, superior raphe, and locus coeruleus. CONCLUSION We propose that these latter regions contribute to the network that defines the "threatened" state, while neurons with transient activity serve as the trigger. Our study highlights the utility of the zebrafish larval alarm response system to examine neural circuits during stress dependent brain state transitions and to discover potential therapeutic agents when such transitions are disrupted.
Collapse
Affiliation(s)
- Suresh Jesuthasan
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, Singapore; Institute of Molecular and Cell Biology, A*STAR, 61 Biopolis Drive, Singapore.
| | - Seetha Krishnan
- NUS Graduate School of Integrative Sciences and Engineering, National University of Singapore, Singapore
| | - Ruey-Kuang Cheng
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, Singapore
| | - Ajay Mathuru
- Institute of Molecular and Cell Biology, A*STAR, 61 Biopolis Drive, Singapore; Yale-NUS College, 12 College Avenue West, Singapore; Dept. of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.
| |
Collapse
|
12
|
The habenula clock influences response to a stressor. Neurobiol Stress 2021; 15:100403. [PMID: 34632007 PMCID: PMC8488752 DOI: 10.1016/j.ynstr.2021.100403] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 09/17/2021] [Accepted: 09/19/2021] [Indexed: 12/12/2022] Open
Abstract
The response of an animal to a sensory stimulus depends on the nature of the stimulus and on expectations, which are mediated by spontaneous activity. Here, we ask how circadian variation in the expectation of danger, and thus the response to a potential threat, is controlled. We focus on the habenula, a mediator of threat response that functions by regulating neuromodulator release, and use zebrafish as the experimental system. Single cell transcriptomics indicates that multiple clock genes are expressed throughout the habenula, while quantitative in situ hybridization confirms that the clock oscillates. Two-photon calcium imaging indicates a circadian change in spontaneous activity of habenula neurons. To assess the role of this clock, a truncated clocka gene was specifically expressed in the habenula. This partially inhibited the clock, as shown by changes in per3 expression as well as altered day-night variation in dopamine, serotonin and acetylcholine levels. Behaviourally, anxiety-like responses evoked by an alarm pheromone were reduced. Circadian effects of the pheromone were disrupted, such that responses in the day resembled those at night. Behaviours that are regulated by the pineal clock and not triggered by stressors were unaffected. We suggest that the habenula clock regulates the expectation of danger, thus providing one mechanism for circadian change in the response to a stressor.
Collapse
|
13
|
Microbiota-brain interactions: Moving toward mechanisms in model organisms. Neuron 2021; 109:3930-3953. [PMID: 34653349 DOI: 10.1016/j.neuron.2021.09.036] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 08/03/2021] [Accepted: 09/17/2021] [Indexed: 02/07/2023]
Abstract
Changes in the microbiota are associated with alterations in nervous system structure-function and behavior and have been implicated in the etiology of neuropsychiatric and neurodegenerative disorders. Most of these studies have centered on mammalian models due to their phylogenetic proximity to humans. Indeed, the germ-free mouse has been a particularly useful model organism for investigating microbiota-brain interactions. However, microbiota-brain axis research on simpler genetic model organisms with a vast and diverse scientific toolkit (zebrafish, Drosophila melanogaster, and Caenorhabditis elegans) is now also coming of age. In this review, we summarize the current state of microbiota-brain axis research in rodents and humans, and then we elaborate and discuss recent research on the neurobiological and behavioral effects of the microbiota in the model systems of fish, flies, and worms. We propose that a cross-species, holistic and mechanistic approach to unravel the microbiota-brain communication is an essential step toward rational microbiota-based therapeutics to combat brain disorders.
Collapse
|
14
|
Rong J, He Y, Tang J, Qiao R, Lin S. "Fishing" nano-bio interactions at the key biological barriers. NANOSCALE 2021; 13:5954-5964. [PMID: 33734277 DOI: 10.1039/d1nr00328c] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Understanding nano-bio interactions is pivotal to the safe implementation of nanotechnology for both biological and environmental applications. Zebrafish as a model organism provides unique opportunities to dissect nano-bio interactions occurring at different biological barriers. In this review, we focus on four key biological barriers, namely cell membrane, blood-brain barrier (BBB), skin and gill epithelia, and gastrointestinal tract (GIT), and highlight recent advancement achieved by using zebrafish to conduct both visualized observations and mechanistic investigations on a diversity of nano-bio interactions.
Collapse
Affiliation(s)
- Jinyu Rong
- College of Environmental Science and Engineering, Biomedical Multidisciplinary Innovation Research Institute, Shanghai East Hospital, Tongji University, Shanghai 200092, China.
| | | | | | | | | |
Collapse
|
15
|
Zolkipli-Cunningham Z, Naviaux JC, Nakayama T, Hirsch CM, Monk JM, Li K, Wang L, Le TP, Meinardi S, Blake DR, Naviaux RK. Metabolic and behavioral features of acute hyperpurinergia and the maternal immune activation mouse model of autism spectrum disorder. PLoS One 2021; 16:e0248771. [PMID: 33735311 PMCID: PMC7971557 DOI: 10.1371/journal.pone.0248771] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Accepted: 03/05/2021] [Indexed: 12/11/2022] Open
Abstract
Since 2012, studies in mice, rats, and humans have suggested that abnormalities in purinergic signaling may be a final common pathway for many genetic and environmental causes of autism spectrum disorder (ASD). The current study in mice was conducted to characterize the bioenergetic, metabolomic, breathomic, and behavioral features of acute hyperpurinergia triggered by systemic injection of the purinergic agonist and danger signal, extracellular ATP (eATP). Responses were studied in C57BL/6J mice in the maternal immune activation (MIA) model and controls. Basal metabolic rates and locomotor activity were measured in CLAMS cages. Plasma metabolomics measured 401 metabolites. Breathomics measured 98 volatile organic compounds. Intraperitoneal eATP dropped basal metabolic rate measured by whole body oxygen consumption by 74% ± 6% (mean ± SEM) and rectal temperature by 6.2˚ ± 0.3˚C in 30 minutes. Over 200 metabolites from 37 different biochemical pathways where changed. Breathomics showed an increase in exhaled carbon monoxide, dimethylsulfide, and isoprene. Metabolomics revealed an acute increase in lactate, citrate, purines, urea, dopamine, eicosanoids, microbiome metabolites, oxidized glutathione, thiamine, niacinamide, and pyridoxic acid, and decreased folate-methylation-1-carbon intermediates, amino acids, short and medium chain acyl-carnitines, phospholipids, ceramides, sphingomyelins, cholesterol, bile acids, and vitamin D similar to some children with ASD. MIA animals were hypersensitive to postnatal exposure to eATP or poly(IC), which produced a rebound increase in body temperature that lasted several weeks before returning to baseline. Acute hyperpurinergia produced metabolic and behavioral changes in mice. The behaviors and metabolic changes produced by ATP injection were associated with mitochondrial functional changes that were profound but reversible.
Collapse
Affiliation(s)
- Zarazuela Zolkipli-Cunningham
- The Mitochondrial and Metabolic Disease Center, University of California, San Diego School of Medicine, San Diego, CA, United States of America
- Department of Neurosciences, University of California, San Diego School of Medicine, San Diego, CA, United States of America
| | - Jane C. Naviaux
- The Mitochondrial and Metabolic Disease Center, University of California, San Diego School of Medicine, San Diego, CA, United States of America
- Department of Neurosciences, University of California, San Diego School of Medicine, San Diego, CA, United States of America
| | - Tomohiro Nakayama
- The Mitochondrial and Metabolic Disease Center, University of California, San Diego School of Medicine, San Diego, CA, United States of America
- Department of Neurosciences, University of California, San Diego School of Medicine, San Diego, CA, United States of America
| | - Charlotte M. Hirsch
- Department of Chemistry, University of California, Irvine (UCI), Irvine, CA, United States of America
| | - Jonathan M. Monk
- The Mitochondrial and Metabolic Disease Center, University of California, San Diego School of Medicine, San Diego, CA, United States of America
- Department of Medicine, University of California, San Diego School of Medicine, San Diego, CA, United States of America
| | - Kefeng Li
- The Mitochondrial and Metabolic Disease Center, University of California, San Diego School of Medicine, San Diego, CA, United States of America
- Department of Medicine, University of California, San Diego School of Medicine, San Diego, CA, United States of America
| | - Lin Wang
- The Mitochondrial and Metabolic Disease Center, University of California, San Diego School of Medicine, San Diego, CA, United States of America
- Department of Medicine, University of California, San Diego School of Medicine, San Diego, CA, United States of America
| | - Thuy P. Le
- The Mitochondrial and Metabolic Disease Center, University of California, San Diego School of Medicine, San Diego, CA, United States of America
- Department of Neurosciences, University of California, San Diego School of Medicine, San Diego, CA, United States of America
| | - Simone Meinardi
- Department of Chemistry, University of California, Irvine (UCI), Irvine, CA, United States of America
| | - Donald R. Blake
- Department of Chemistry, University of California, Irvine (UCI), Irvine, CA, United States of America
| | - Robert K. Naviaux
- The Mitochondrial and Metabolic Disease Center, University of California, San Diego School of Medicine, San Diego, CA, United States of America
- Department of Medicine, University of California, San Diego School of Medicine, San Diego, CA, United States of America
- Department of Pediatrics, University of California, San Diego School of Medicine, San Diego, CA, United States of America
- Department of Pathology, University of California, San Diego School of Medicine, San Diego, CA, United States of America
| |
Collapse
|
16
|
Cheung KY, Jesuthasan SJ, Baxendale S, van Hateren NJ, Marzo M, Hill CJ, Whitfield TT. Olfactory Rod Cells: A Rare Cell Type in the Larval Zebrafish Olfactory Epithelium With a Large Actin-Rich Apical Projection. Front Physiol 2021; 12:626080. [PMID: 33716772 PMCID: PMC7952648 DOI: 10.3389/fphys.2021.626080] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Accepted: 01/25/2021] [Indexed: 11/13/2022] Open
Abstract
We report the presence of a rare cell type, the olfactory rod cell, in the developing zebrafish olfactory epithelium. These cells each bear a single actin-rich rod-like apical projection extending 5–10 μm from the epithelial surface. Live imaging with a ubiquitous Lifeact-RFP label indicates that the olfactory rods can oscillate. Olfactory rods arise within a few hours of the olfactory pit opening, increase in numbers and size during larval stages, and can develop in the absence of olfactory cilia. Olfactory rod cells differ in morphology from the known classes of olfactory sensory neuron, but express reporters driven by neuronal promoters. A sub-population of olfactory rod cells expresses a Lifeact-mRFPruby transgene driven by the sox10 promoter. Mosaic expression of this transgene reveals that olfactory rod cells have rounded cell bodies located apically in the olfactory epithelium and have no detectable axon. We offer speculation on the possible function of these cells in the Discussion.
Collapse
Affiliation(s)
- King Yee Cheung
- Department of Biomedical Science, Bateson Centre and Neuroscience Institute, University of Sheffield, Sheffield, United Kingdom
| | - Suresh J Jesuthasan
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore.,Institute of Molecular and Cell Biology, Singapore, Singapore
| | - Sarah Baxendale
- Department of Biomedical Science, Bateson Centre and Neuroscience Institute, University of Sheffield, Sheffield, United Kingdom
| | - Nicholas J van Hateren
- Department of Biomedical Science, Bateson Centre and Neuroscience Institute, University of Sheffield, Sheffield, United Kingdom
| | - Mar Marzo
- Department of Biomedical Science, Bateson Centre and Neuroscience Institute, University of Sheffield, Sheffield, United Kingdom
| | - Christopher J Hill
- Department of Biomedical Science, Bateson Centre and Neuroscience Institute, University of Sheffield, Sheffield, United Kingdom
| | - Tanya T Whitfield
- Department of Biomedical Science, Bateson Centre and Neuroscience Institute, University of Sheffield, Sheffield, United Kingdom
| |
Collapse
|
17
|
Epidermal Club Cells in Fishes: A Case for Ecoimmunological Analysis. Int J Mol Sci 2021; 22:ijms22031440. [PMID: 33535506 PMCID: PMC7867084 DOI: 10.3390/ijms22031440] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2021] [Revised: 01/24/2021] [Accepted: 01/28/2021] [Indexed: 12/14/2022] Open
Abstract
Epidermal club cells (ECCs), along with mucus cells, are present in the skin of many fishes, particularly in the well-studied Ostariophysan family Cyprinidae. Most ECC-associated literature has focused on the potential role of ECCs as a component of chemical alarm cues released passively when a predator damages the skin of its prey, alerting nearby prey to the presence of an active predator. Because this warning system is maintained by receiver-side selection (senders are eaten), there is want of a mechanism to confer fitness benefits to the individual that invests in ECCs to explain their evolutionary origin and maintenance in this speciose group of fishes. In an attempt to understand the fitness benefits that accrue from investment in ECCs, we reviewed the phylogenetic distribution of ECCs and their histochemical properties. ECCs are found in various forms in all teleost superorders and in the chondrostei inferring either early or multiple independent origins over evolutionary time. We noted that ECCs respond to several environmental stressors/immunomodulators including parasites and pathogens, are suppressed by immunomodulators such as testosterone and cortisol, and their density covaries with food ration, demonstrating a dynamic metabolic cost to maintaining these cells. ECC density varies widely among and within fish populations, suggesting that ECCs may be a convenient tool with which to assay ecoimmunological tradeoffs between immune stress and foraging activity, reproductive state, and predator-prey interactions. Here, we review the case for ECC immune function, immune functions in fishes generally, and encourage future work describing the precise role of ECCs in the immune system and life history evolution in fishes.
Collapse
|
18
|
Kermen F, Mandairon N, Chalençon L. Odor hedonics coding in the vertebrate olfactory bulb. Cell Tissue Res 2021; 383:485-493. [PMID: 33515292 PMCID: PMC7873110 DOI: 10.1007/s00441-020-03372-w] [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] [Received: 09/04/2020] [Accepted: 11/29/2020] [Indexed: 12/21/2022]
Abstract
Whether an odorant is perceived as pleasant or unpleasant (hedonic value) governs a range of crucial behaviors: foraging, escaping danger, and social interaction. Despite its importance in olfactory perception, little is known regarding how odor hedonics is represented and encoded in the brain. Here, we review recent findings describing how odorant hedonic value is represented in the first olfaction processing center, the olfactory bulb. We discuss how olfactory bulb circuits might contribute to the coding of innate and learned odorant hedonics in addition to the odorant's physicochemical properties.
Collapse
Affiliation(s)
- Florence Kermen
- Department of Biology, Faculty of Natural Sciences, Norwegian University of Science and Technology, 7030, Trondheim, Norway.
| | - Nathalie Mandairon
- CNRS. UMR 5292: INSERM, U1028: Lyon Neuroscience Research Center Neuroplasticity and Neuropathology of Olfactory Perception Team, University Lyon, University Lyon1, F-69000, Villeurbanne, France
| | - Laura Chalençon
- CNRS. UMR 5292: INSERM, U1028: Lyon Neuroscience Research Center Neuroplasticity and Neuropathology of Olfactory Perception Team, University Lyon, University Lyon1, F-69000, Villeurbanne, France
| |
Collapse
|
19
|
de Luis B, Morellá-Aucejo Á, Llopis-Lorente A, Godoy-Reyes TM, Villalonga R, Aznar E, Sancenón F, Martínez-Máñez R. A chemical circular communication network at the nanoscale. Chem Sci 2020; 12:1551-1559. [PMID: 34163918 PMCID: PMC8179104 DOI: 10.1039/d0sc04743k] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Accepted: 11/29/2020] [Indexed: 12/18/2022] Open
Abstract
In nature, coordinated communication between different entities enables a group to accomplish sophisticated functionalities that go beyond those carried out by individual agents. The possibility of programming and developing coordinated communication networks at the nanoscale-based on the exchange of chemical messengers-may open new approaches in biomedical and communication areas. Here, a stimulus-responsive circular model of communication between three nanodevices based on enzyme-functionalized Janus Au-mesoporous silica capped nanoparticles is presented. The output in the community of nanoparticles is only observed after a hierarchically programmed flow of chemical information between the members.
Collapse
Affiliation(s)
- Beatriz de Luis
- Instituto Interuniversitario de Investigación de Reconocimiento Molecular y Desarrollo Tecnológico (IDM), Universitat Politècnica de València, Universitat de València Camino de Vera s/n 46022 Valencia Spain
- CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN) Spain
| | - Ángela Morellá-Aucejo
- Instituto Interuniversitario de Investigación de Reconocimiento Molecular y Desarrollo Tecnológico (IDM), Universitat Politècnica de València, Universitat de València Camino de Vera s/n 46022 Valencia Spain
- CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN) Spain
| | - Antoni Llopis-Lorente
- Instituto Interuniversitario de Investigación de Reconocimiento Molecular y Desarrollo Tecnológico (IDM), Universitat Politècnica de València, Universitat de València Camino de Vera s/n 46022 Valencia Spain
- CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN) Spain
| | - Tania M Godoy-Reyes
- Instituto Interuniversitario de Investigación de Reconocimiento Molecular y Desarrollo Tecnológico (IDM), Universitat Politècnica de València, Universitat de València Camino de Vera s/n 46022 Valencia Spain
- CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN) Spain
| | - Reynaldo Villalonga
- Nanosensors & Nanomachines Group, Department of Analytical Chemistry, Faculty of Chemistry, Complutense University of Madrid Madrid Spain
| | - Elena Aznar
- Instituto Interuniversitario de Investigación de Reconocimiento Molecular y Desarrollo Tecnológico (IDM), Universitat Politècnica de València, Universitat de València Camino de Vera s/n 46022 Valencia Spain
- CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN) Spain
| | - Félix Sancenón
- Instituto Interuniversitario de Investigación de Reconocimiento Molecular y Desarrollo Tecnológico (IDM), Universitat Politècnica de València, Universitat de València Camino de Vera s/n 46022 Valencia Spain
- CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN) Spain
| | - Ramón Martínez-Máñez
- Instituto Interuniversitario de Investigación de Reconocimiento Molecular y Desarrollo Tecnológico (IDM), Universitat Politècnica de València, Universitat de València Camino de Vera s/n 46022 Valencia Spain
- CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN) Spain
- Unidad Mixta UPV-CIPF de Investigación en Mecanismos de Enfermedades y Nanomedicina, Universitat Politècnica de València, Centro de Investigación Príncipe Felipe Valencia Spain
- Unidad Mixta de Investigación en Nanomedicina y Sensores, Universitat Politècnica de València, Instituto de Investigación Sanitaria La Fe Valencia Spain
| |
Collapse
|
20
|
Kermen F, Darnet L, Wiest C, Palumbo F, Bechert J, Uslu O, Yaksi E. Stimulus-specific behavioral responses of zebrafish to a large range of odors exhibit individual variability. BMC Biol 2020; 18:66. [PMID: 32539727 PMCID: PMC7296676 DOI: 10.1186/s12915-020-00801-8] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2019] [Accepted: 05/22/2020] [Indexed: 02/02/2023] Open
Abstract
BACKGROUND Odor-driven behaviors such as feeding, mating, and predator avoidance are crucial for animal survival. The neural pathways processing these behaviors have been well characterized in a number of species, and involve the activity of diverse brain regions following stimulation of the olfactory bulb by specific odors. However, while the zebrafish olfactory circuitry is well understood, a comprehensive characterization linking odor-driven behaviors to specific odors is needed to better relate olfactory computations to animal responses. RESULTS Here, we used a medium-throughput setup to measure the swimming trajectories of 10 zebrafish in response to 17 ecologically relevant odors. By selecting appropriate locomotor metrics, we constructed ethograms systematically describing odor-induced changes in the swimming trajectory. We found that adult zebrafish reacted to most odorants using different behavioral programs and that a combination of a few relevant behavioral metrics enabled us to capture most of the variance in these innate odor responses. We observed that individual components of natural food and alarm odors do not elicit the full behavioral response. Finally, we show that zebrafish blood elicits prominent defensive behaviors similar to those evoked by skin extract and activates spatially overlapping olfactory bulb domains. CONCLUSION Altogether, our results highlight a prominent intra- and inter-individual variability in zebrafish odor-driven behaviors and identify a small set of waterborne odors that elicit robust responses. Our behavioral setup and our results will be useful resources for future studies interested in characterizing innate olfactory behaviors in aquatic animals.
Collapse
Affiliation(s)
- Florence Kermen
- Kavli Institute for Systems Neuroscience and Centre for Neural Computation, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, 7030, Trondheim, Norway.
- Neuro-Electronics Research Flanders, Imec Campus, Kapeldreef 75, 3001, Leuven, Belgium.
- Department of Biology, Faculty of Natural Sciences, Norwegian University of Science and Technology, 7491, Trondheim, Norway.
- KU Leuven, 3000, Leuven, Belgium.
| | - Lea Darnet
- Neuro-Electronics Research Flanders, Imec Campus, Kapeldreef 75, 3001, Leuven, Belgium
- KU Leuven, 3000, Leuven, Belgium
| | - Christoph Wiest
- Kavli Institute for Systems Neuroscience and Centre for Neural Computation, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, 7030, Trondheim, Norway
- Medical Research Council Brain Network Dynamics Unit, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, OX3 9DU, UK
| | - Fabrizio Palumbo
- Kavli Institute for Systems Neuroscience and Centre for Neural Computation, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, 7030, Trondheim, Norway
| | - Jack Bechert
- Neuro-Electronics Research Flanders, Imec Campus, Kapeldreef 75, 3001, Leuven, Belgium
- KU Leuven, 3000, Leuven, Belgium
| | - Ozge Uslu
- Neuro-Electronics Research Flanders, Imec Campus, Kapeldreef 75, 3001, Leuven, Belgium
- KU Leuven, 3000, Leuven, Belgium
| | - Emre Yaksi
- Kavli Institute for Systems Neuroscience and Centre for Neural Computation, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, 7030, Trondheim, Norway.
- Neuro-Electronics Research Flanders, Imec Campus, Kapeldreef 75, 3001, Leuven, Belgium.
- KU Leuven, 3000, Leuven, Belgium.
| |
Collapse
|