1
|
Gest AM, Sahan AZ, Zhong Y, Lin W, Mehta S, Zhang J. Molecular Spies in Action: Genetically Encoded Fluorescent Biosensors Light up Cellular Signals. Chem Rev 2024; 124:12573-12660. [PMID: 39535501 PMCID: PMC11613326 DOI: 10.1021/acs.chemrev.4c00293] [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: 04/17/2024] [Revised: 09/07/2024] [Accepted: 09/20/2024] [Indexed: 11/16/2024]
Abstract
Cellular function is controlled through intricate networks of signals, which lead to the myriad pathways governing cell fate. Fluorescent biosensors have enabled the study of these signaling pathways in living systems across temporal and spatial scales. Over the years there has been an explosion in the number of fluorescent biosensors, as they have become available for numerous targets, utilized across spectral space, and suited for various imaging techniques. To guide users through this extensive biosensor landscape, we discuss critical aspects of fluorescent proteins for consideration in biosensor development, smart tagging strategies, and the historical and recent biosensors of various types, grouped by target, and with a focus on the design and recent applications of these sensors in living systems.
Collapse
Affiliation(s)
- Anneliese
M. M. Gest
- Department
of Pharmacology, University of California,
San Diego, La Jolla, California 92093, United States
| | - Ayse Z. Sahan
- Department
of Pharmacology, University of California,
San Diego, La Jolla, California 92093, United States
- Biomedical
Sciences Graduate Program, University of
California, San Diego, La Jolla, California 92093, United States
| | - Yanghao Zhong
- Department
of Pharmacology, University of California,
San Diego, La Jolla, California 92093, United States
| | - Wei Lin
- Department
of Pharmacology, University of California,
San Diego, La Jolla, California 92093, United States
| | - Sohum Mehta
- Department
of Pharmacology, University of California,
San Diego, La Jolla, California 92093, United States
| | - Jin Zhang
- Department
of Pharmacology, University of California,
San Diego, La Jolla, California 92093, United States
- Shu
Chien-Gene Lay Department of Bioengineering, University of California, San Diego, La Jolla, California 92093, United States
- Department
of Chemistry and Biochemistry, University
of California, San Diego, La Jolla, California 92093, United States
| |
Collapse
|
2
|
Nakamori T, Komatsuzawa I, Iwata U, Makita A, Kagiya G, Fujitani K, Kitaguchi T, Tsuboi T, Ohki-Hamazaki H. The role of osteocrin in memory formation during early learning, as revealed by visual imprinting in chicks. iScience 2024; 27:111195. [PMID: 39600306 PMCID: PMC11591550 DOI: 10.1016/j.isci.2024.111195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 02/17/2024] [Accepted: 10/15/2024] [Indexed: 11/29/2024] Open
Abstract
Osteocrin (OSTN) is structurally associated with natriuretic peptides. Its expression in the brain, which has only been recognized in anthropoid primates, is induced by sensory stimuli and regulates the activity-dependent dendritic growth of neurons. However, details on the signaling mechanisms of OSTN and its function in plastic changes during learning and memory have yet to be elucidated. We found that OSTN was expressed in the cortical region of the chicken brain. The injection of chicken OSTN (chOSTN) after imprinting training prolonged the memory retention for the imprinting stimulus. Conversely, a reduction in the OSTN receptor chNPR3 inhibited memory retention. The memory retention was positively correlated with a high level of chOSTN and fewer neurites in the cortical region. In conclusion, OSTN-NPR3 signaling promoted memory consolidation and/or retention by regulating neurite branching during childhood.
Collapse
Affiliation(s)
- Tomoharu Nakamori
- College of Liberal Arts and Sciences, Kitasato University, Kitasato, Minami-ku, Sagamihara, Kanagawa 252-0373, Japan
| | - Izumi Komatsuzawa
- College of Liberal Arts and Sciences, Kitasato University, Kitasato, Minami-ku, Sagamihara, Kanagawa 252-0373, Japan
| | - Umi Iwata
- College of Liberal Arts and Sciences, Kitasato University, Kitasato, Minami-ku, Sagamihara, Kanagawa 252-0373, Japan
| | - Ami Makita
- College of Liberal Arts and Sciences, Kitasato University, Kitasato, Minami-ku, Sagamihara, Kanagawa 252-0373, Japan
| | - Go Kagiya
- School of Allied Health Sciences, and Regenerative Medicine and Cell Design Research Facility, Kitasato University, Kitasato, Minami-ku, Sagamihara, Kanagawa 252-0374, Japan
| | - Kazuko Fujitani
- Gene Analysis Center, School of Medicine, Kitasato University, Kitasato, Minami-ku, Sagamihara, Kanagawa 252-0374, Japan
| | - Tetsuya Kitaguchi
- Laboratory for Chemistry and Life Science, Institute of Innovative Research, Tokyo Institute of Technology, Nagatsuta-cho, Midori-ku, Yokohama, Kanagawa 226-8503, Japan
| | - Takashi Tsuboi
- Department of Life Sciences, Graduate School of Arts and Sciences, The University of Tokyo, Komaba, Meguro-ku, Tokyo 153-8902, Japan
| | - Hiroko Ohki-Hamazaki
- College of Liberal Arts and Sciences, Kitasato University, Kitasato, Minami-ku, Sagamihara, Kanagawa 252-0373, Japan
| |
Collapse
|
3
|
Yong JJM, Gao X, Prakash P, Ang JW, Lai SK, Chen MW, Neo JJL, Lescar J, Li HY, Preiser PR. Red blood cell signaling is functionally conserved in Plasmodium invasion. iScience 2024; 27:111052. [PMID: 39635131 PMCID: PMC11615254 DOI: 10.1016/j.isci.2024.111052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 06/20/2024] [Accepted: 09/24/2024] [Indexed: 12/07/2024] Open
Abstract
It is widely recognized that Plasmodium merozoites secrete ligands that interact with RBC receptors. Meanwhile the question on whether these interactions trigger RBC signals essential for invasion remains unresolved. There is evidence that Plasmodium falciparum parasites manipulate native RBC Ca2+ signaling to facilitate invasion. Here, we demonstrate a key role of RBC Ca2+ influx that is conserved across different Plasmodium species during invasion. RH5-basigin interaction triggers RBC cAMP increase to promote Ca2+ influx. The RBC signaling pathways can be blocked by a range of inhibitors during Plasmodium invasion, providing the evidence of a functionally conserved host cAMP-Ca2+ signaling that drives invasion and junction formation. Furthermore, RH5-basigin binding induces a pre-existing multimeric RBC membrane complex to undergo increased protein association containing the cAMP-inducing β-adrenergic receptor. Our work presents evidence of a conserved host cell signaling cascade necessary for Plasmodium invasion and will create opportunities to therapeutically target merozoite invasion.
Collapse
Affiliation(s)
- James Jia Ming Yong
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Singapore
| | - Xiaohong Gao
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Singapore
| | - Prem Prakash
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Singapore
| | - Jing Wen Ang
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Singapore
| | - Soak Kuan Lai
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Singapore
| | - Ming Wei Chen
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Singapore
| | - Jason Jun Long Neo
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Singapore
| | - Julien Lescar
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Singapore
| | - Hoi Yeung Li
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Singapore
| | - Peter R. Preiser
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Singapore
| |
Collapse
|
4
|
Roychaudhuri R, West T, Bhattacharya S, Saavedra HG, Lee H, Albacarys L, Gadalla MM, Amzel M, Yang P, Snyder SH. Mammalian D-Cysteine controls insulin secretion in the pancreas. Mol Metab 2024; 90:102043. [PMID: 39368613 PMCID: PMC11536007 DOI: 10.1016/j.molmet.2024.102043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 09/10/2024] [Accepted: 09/27/2024] [Indexed: 10/07/2024] Open
Abstract
BACKGROUND D-amino acids are being recognized as important molecules in mammals with function. This is a first identification of endogenous D-cysteine in mammalian pancreas. METHODS Using a novel stereospecific bioluminescent assay, chiral chromatography, enzyme kinetics and a transgenic mouse model we identify endogenous D-cysteine. We elucidate its function in two mice models of type 1 diabetes (STZ and NOD), and in tests of Glucose Stimulated Insulin Secretion in isolated mouse and human islets and INS-1 832/13 cell line. RESULTS AND DISCUSSION D-cysteine is synthesized by serine racemase (SR) and SR-/- mice produce 6-10 fold higher levels of insulin in the pancreas and plasma including higher glycogen and ketone bodies in the liver. The excess insulin is stored as amyloid in secretory vesicles and exosomes. In glucose stimulated insulin secretion in mouse and human islets, equimolar amount of D-cysteine showed higher inhibition of insulin secretion compared to D-serine, another closely related stereoisomer synthesized by SR. In mouse models of diabetes (Streptozotocin (STZ) and Non Obese Diabetes (NOD) and human pancreas, the diabetic state showed increased expression of D-cysteine compared to D-serine followed by increased expression of SR. SR-/- mice show decreased cAMP in the pancreas, lower DNA methyltransferase enzymatic and promoter activities followed by reduced phosphorylation of CREB (S133), resulting in decreased methylation of the Ins1 promoter. D-cysteine is efficiently metabolized by D-amino acid oxidase and transported by ASCT2 and Asc1. Dietary supplementation with methyl donors restored the high insulin levels and low DNMT enzymatic activity in SR-/- mice. CONCLUSIONS Our data show that endogenous D-cysteine in the mammalian pancreas is a regulator of insulin secretion.
Collapse
Affiliation(s)
- Robin Roychaudhuri
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Obstetrics, Gynecology and Reproductive Sciences, Center for Birth Defects, University of Maryland School of Medicine, Baltimore, MD 21201, USA.
| | - Timothy West
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Soumyaroop Bhattacharya
- Department of Neonatology, University of Rochester Medical Center, Rochester, New York, NY 14642, USA
| | - Harry G Saavedra
- Centro de Investigacion en Bioingenieria, Universidad de Ingenieria y Tecnologia (UTEC), 15063 Lima, Peru
| | - Hangnoh Lee
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Lauren Albacarys
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Moataz M Gadalla
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Mario Amzel
- Department of Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Peixin Yang
- Department of Obstetrics, Gynecology and Reproductive Sciences, Center for Birth Defects, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Solomon H Snyder
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| |
Collapse
|
5
|
Hakata T, Yamauchi I, Kosugi D, Sugawa T, Fujita H, Okamoto K, Ueda Y, Fujii T, Taura D, Inagaki N. High-throughput Screening for Cushing Disease: Therapeutic Potential of Thiostrepton via Cell Cycle Regulation. Endocrinology 2024; 165:bqae089. [PMID: 39058910 DOI: 10.1210/endocr/bqae089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Revised: 07/03/2024] [Accepted: 07/17/2024] [Indexed: 07/28/2024]
Abstract
Cushing disease is a life-threatening disorder caused by autonomous secretion of ACTH from pituitary neuroendocrine tumors (PitNETs). Few drugs are indicated for inoperative Cushing disease, in particular that due to aggressive PitNETs. To explore agents that regulate ACTH-secreting PitNETs, we conducted high-throughput screening (HTS) using AtT-20, a murine pituitary tumor cell line characterized by ACTH secretion. For the HTS, we constructed a live cell-based ACTH reporter assay for high-throughput evaluation of ACTH changes. This assay was based on HEK293T cells overexpressing components of the ACTH receptor and a fluorescent cAMP biosensor, with high-throughput acquisition of fluorescence images. We treated AtT-20 cells with compounds and assessed ACTH concentrations in the conditioned media using the reporter assay. Of 2480 screened bioactive compounds, over 50% inhibition of ACTH secreted from AtT-20 cells was seen with 84 compounds at 10 μM and 20 compounds at 1 μM. Among these hit compounds, we focused on thiostrepton (TS) and determined its antitumor effects in both in vitro and in vivo xenograft models of Cushing disease. Transcriptome and flow cytometry analyses revealed that TS administration induced AtT-20 cell cycle arrest at the G2/M phase, which was mediated by FOXM1-independent mechanisms including downregulation of cyclins. Simultaneous TS administration with a cyclin-dependent kinase 4/6 inhibitor that affected the cell cycle at the G0/1 phase showed cooperative antitumor effects. Thus, TS is a promising therapeutic agent for Cushing disease. Our list of hit compounds and new mechanistic insights into TS effects serve as a valuable foundation for future research.
Collapse
Affiliation(s)
- Takuro Hakata
- Department of Diabetes, Endocrinology and Nutrition, Kyoto University Graduate School of Medicine, Sakyo-ku, Kyoto 606-8507, Japan
| | - Ichiro Yamauchi
- Department of Diabetes, Endocrinology and Nutrition, Kyoto University Graduate School of Medicine, Sakyo-ku, Kyoto 606-8507, Japan
| | - Daisuke Kosugi
- Department of Diabetes, Endocrinology and Nutrition, Kyoto University Graduate School of Medicine, Sakyo-ku, Kyoto 606-8507, Japan
| | - Taku Sugawa
- Department of Diabetes, Endocrinology and Nutrition, Kyoto University Graduate School of Medicine, Sakyo-ku, Kyoto 606-8507, Japan
| | - Haruka Fujita
- Department of Diabetes, Endocrinology and Nutrition, Kyoto University Graduate School of Medicine, Sakyo-ku, Kyoto 606-8507, Japan
| | - Kentaro Okamoto
- Department of Diabetes, Endocrinology and Nutrition, Kyoto University Graduate School of Medicine, Sakyo-ku, Kyoto 606-8507, Japan
| | - Yohei Ueda
- Department of Diabetes, Endocrinology and Nutrition, Kyoto University Graduate School of Medicine, Sakyo-ku, Kyoto 606-8507, Japan
| | - Toshihito Fujii
- Department of Diabetes, Endocrinology and Nutrition, Kyoto University Graduate School of Medicine, Sakyo-ku, Kyoto 606-8507, Japan
| | - Daisuke Taura
- Department of Diabetes, Endocrinology and Nutrition, Kyoto University Graduate School of Medicine, Sakyo-ku, Kyoto 606-8507, Japan
| | - Nobuya Inagaki
- Medical Research Institute, Kitano Hospital, PIIF Tazuke-kofukai, Kita-ku, Osaka 530-8480, Japan
| |
Collapse
|
6
|
Boto T, Tomchik SM. Functional Imaging of Learning-Induced Plasticity in the Central Nervous System with Genetically Encoded Reporters in Drosophila. Cold Spring Harb Protoc 2024; 2024:pdb.top107799. [PMID: 37197830 DOI: 10.1101/pdb.top107799] [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] [Indexed: 05/19/2023]
Abstract
Learning and memory allow animals to adjust their behavior based on the predictive value of their past experiences. Memories often exist in complex representations, spread across numerous cells and synapses in the brain. Studying relatively simple forms of memory provides insights into the fundamental processes that underlie multiple forms of memory. Associative learning occurs when an animal learns the relationship between two previously unrelated sensory stimuli, such as when a hungry animal learns that a particular odor is followed by a tasty reward. Drosophila is a particularly powerful model to study how this type of memory works. The fundamental principles are widely shared among animals, and there is a wide range of genetic tools available to study circuit function in flies. In addition, the olfactory structures that mediate associative learning in flies, such as the mushroom body and its associated neurons, are anatomically organized, relatively well-characterized, and readily accessible to imaging. Here, we review the olfactory anatomy and physiology of the olfactory system, describe how plasticity in the olfactory pathway mediates learning and memory, and explain the general principles underlying calcium imaging approaches.
Collapse
Affiliation(s)
- Tamara Boto
- Department of Physiology, Trinity College Dublin, Dublin 2, Ireland
- Trinity College Institute of Neuroscience, Trinity College Dublin, Dublin 2, Ireland
| | - Seth M Tomchik
- Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, Iowa City, Iowa 52242, USA
- Stead Family Department of Pediatrics, University of Iowa Carver College of Medicine, Iowa City, Iowa 52242, USA
- Iowa Neuroscience Institute, University of Iowa Carver College of Medicine, Iowa City, Iowa 52242, USA
| |
Collapse
|
7
|
Yokoyama T, Manita S, Uwamori H, Tajiri M, Imayoshi I, Yagishita S, Murayama M, Kitamura K, Sakamoto M. A multicolor suite for deciphering population coding of calcium and cAMP in vivo. Nat Methods 2024; 21:897-907. [PMID: 38514778 PMCID: PMC11093745 DOI: 10.1038/s41592-024-02222-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Accepted: 02/21/2024] [Indexed: 03/23/2024]
Abstract
cAMP is a universal second messenger regulated by various upstream pathways including Ca2+ and G-protein-coupled receptors (GPCRs). To decipher in vivo cAMP dynamics, we rationally designed cAMPinG1, a sensitive genetically encoded green cAMP indicator that outperformed its predecessors in both dynamic range and cAMP affinity. Two-photon cAMPinG1 imaging detected cAMP transients in the somata and dendritic spines of neurons in the mouse visual cortex on the order of tens of seconds. In addition, multicolor imaging with a sensitive red Ca2+ indicator RCaMP3 allowed simultaneous measurement of population patterns in Ca2+ and cAMP in hundreds of neurons. We found Ca2+-related cAMP responses that represented specific information, such as direction selectivity in vision and locomotion, as well as GPCR-related cAMP responses. Overall, our multicolor suite will facilitate analysis of the interaction between the Ca2+, GPCR and cAMP signaling at single-cell resolution both in vitro and in vivo.
Collapse
Affiliation(s)
- Tatsushi Yokoyama
- Department of Optical Neural and Molecular Physiology, Graduate School of Biostudies, Kyoto University, Kyoto, Japan.
- Center for Living Systems Information Science, Graduate School of Biostudies, Kyoto University, Kyoto, Japan.
- Department of Brain Development and Regeneration, Graduate School of Biostudies, Kyoto University, Kyoto, Japan.
- Laboratory of Deconstruction of Stem Cells, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, Japan.
| | - Satoshi Manita
- Department of Neurophysiology, Graduate School of Medicine, University of Yamanashi, Chuo, Yamanashi, Japan
| | - Hiroyuki Uwamori
- Laboratory for Haptic Perception and Cognitive Physiology, Center for Brain Science, RIKEN, Wako, Saitama, Japan
| | - Mio Tajiri
- Department of Structural Physiology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Itaru Imayoshi
- Center for Living Systems Information Science, Graduate School of Biostudies, Kyoto University, Kyoto, Japan
- Department of Brain Development and Regeneration, Graduate School of Biostudies, Kyoto University, Kyoto, Japan
- Laboratory of Deconstruction of Stem Cells, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Sho Yagishita
- Department of Structural Physiology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Masanori Murayama
- Laboratory for Haptic Perception and Cognitive Physiology, Center for Brain Science, RIKEN, Wako, Saitama, Japan
| | - Kazuo Kitamura
- Department of Neurophysiology, Graduate School of Medicine, University of Yamanashi, Chuo, Yamanashi, Japan
| | - Masayuki Sakamoto
- Department of Optical Neural and Molecular Physiology, Graduate School of Biostudies, Kyoto University, Kyoto, Japan.
- Center for Living Systems Information Science, Graduate School of Biostudies, Kyoto University, Kyoto, Japan.
- Department of Brain Development and Regeneration, Graduate School of Biostudies, Kyoto University, Kyoto, Japan.
- Laboratory of Deconstruction of Stem Cells, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, Japan.
- Precursory Research for Embryonic Science and Technology (PRESTO), Japan Science and Technology Agency, Kyoto, Japan.
| |
Collapse
|
8
|
Cahill MK, Collard M, Tse V, Reitman ME, Etchenique R, Kirst C, Poskanzer KE. Network-level encoding of local neurotransmitters in cortical astrocytes. Nature 2024; 629:146-153. [PMID: 38632406 PMCID: PMC11062919 DOI: 10.1038/s41586-024-07311-5] [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: 04/03/2023] [Accepted: 03/13/2024] [Indexed: 04/19/2024]
Abstract
Astrocytes, the most abundant non-neuronal cell type in the mammalian brain, are crucial circuit components that respond to and modulate neuronal activity through calcium (Ca2+) signalling1-7. Astrocyte Ca2+ activity is highly heterogeneous and occurs across multiple spatiotemporal scales-from fast, subcellular activity3,4 to slow, synchronized activity across connected astrocyte networks8-10-to influence many processes5,7,11. However, the inputs that drive astrocyte network dynamics remain unclear. Here we used ex vivo and in vivo two-photon astrocyte imaging while mimicking neuronal neurotransmitter inputs at multiple spatiotemporal scales. We find that brief, subcellular inputs of GABA and glutamate lead to widespread, long-lasting astrocyte Ca2+ responses beyond an individual stimulated cell. Further, we find that a key subset of Ca2+ activity-propagative activity-differentiates astrocyte network responses to these two main neurotransmitters, and may influence responses to future inputs. Together, our results demonstrate that local, transient neurotransmitter inputs are encoded by broad cortical astrocyte networks over a minutes-long time course, contributing to accumulating evidence that substantial astrocyte-neuron communication occurs across slow, network-level spatiotemporal scales12-14. These findings will enable future studies to investigate the link between specific astrocyte Ca2+ activity and specific functional outputs, which could build a consistent framework for astrocytic modulation of neuronal activity.
Collapse
Affiliation(s)
- Michelle K Cahill
- Department of Biochemistry & Biophysics, University of California, San Francisco, CA, USA
- Neuroscience Graduate Program, University of California, San Francisco, CA, USA
| | - Max Collard
- Department of Biochemistry & Biophysics, University of California, San Francisco, CA, USA
- Neuroscience Graduate Program, University of California, San Francisco, CA, USA
| | - Vincent Tse
- Department of Biochemistry & Biophysics, University of California, San Francisco, CA, USA
| | - Michael E Reitman
- Department of Biochemistry & Biophysics, University of California, San Francisco, CA, USA
- Neuroscience Graduate Program, University of California, San Francisco, CA, USA
| | - Roberto Etchenique
- Departamento de Química Inorgánica, Analítica y Química Física, INQUIMAE, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, CONICET, Buenos Aires, Argentina
| | - Christoph Kirst
- Neuroscience Graduate Program, University of California, San Francisco, CA, USA
- Department of Anatomy, University of California, San Francisco, CA, USA
- Kavli Institute for Fundamental Neuroscience, San Francisco, CA, USA
- Lawrence Berkeley National Laboratory, Berkeley, CA, USA
| | - Kira E Poskanzer
- Department of Biochemistry & Biophysics, University of California, San Francisco, CA, USA.
- Neuroscience Graduate Program, University of California, San Francisco, CA, USA.
- Kavli Institute for Fundamental Neuroscience, San Francisco, CA, USA.
| |
Collapse
|
9
|
Casiraghi M, Wang H, Brennan P, Habrian C, Hubner H, Schmidt MF, Maul L, Pani B, Bahriz SM, Xu B, White E, Sunahara RK, Xiang YK, Lefkowitz RJ, Isacoff EY, Nucci N, Gmeiner P, Lerch M, Kobilka BK. Structure and dynamics determine G protein coupling specificity at a class A GPCR. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.28.587240. [PMID: 38586060 PMCID: PMC10996611 DOI: 10.1101/2024.03.28.587240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2024]
Abstract
G protein coupled receptors (GPCRs) exhibit varying degrees of selectivity for different G protein isoforms. Despite the abundant structures of GPCR-G protein complexes, little is known about the mechanism of G protein coupling specificity. The β2-adrenergic receptor is an example of GPCR with high selectivity for Gαs, the stimulatory G protein for adenylyl cyclase, and much weaker for the Gαi family of G proteins inhibiting adenylyl cyclase. By developing a new Gαi-biased agonist (LM189), we provide structural and biophysical evidence supporting that distinct conformations at ICL2 and TM6 are required for coupling of the different G protein subtypes Gαs and Gαi. These results deepen our understanding of G protein specificity and bias and can accelerate the design of ligands that select for preferred signaling pathways.
Collapse
|
10
|
Lee HC, Oliveira NMM, Hastings C, Baillie-Benson P, Moverley AA, Lu HC, Zheng Y, Wilby EL, Weil TT, Page KM, Fu J, Moris N, Stern CD. Regulation of long-range BMP gradients and embryonic polarity by propagation of local calcium-firing activity. Nat Commun 2024; 15:1463. [PMID: 38368410 PMCID: PMC10874436 DOI: 10.1038/s41467-024-45772-4] [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: 06/01/2023] [Accepted: 02/02/2024] [Indexed: 02/19/2024] Open
Abstract
Many amniote vertebrate species including humans can form identical twins from a single embryo, but this only occurs rarely. It has been suggested that the primitive-streak-forming embryonic region emits signals that inhibit streak formation elsewhere but the signals involved, how they are transmitted and how they act has not been elucidated. Here we show that short tracks of calcium firing activity propagate through extraembryonic tissue via gap junctions and prevent ectopic primitive streak formation in chick embryos. Cross-regulation of calcium activity and an inhibitor of primitive streak formation (Bone Morphogenetic Protein, BMP) via NF-κB and NFAT establishes a long-range BMP gradient spanning the embryo. This mechanism explains how embryos of widely different sizes can maintain positional information that determines embryo polarity. We provide evidence for similar mechanisms in two different human embryo models and in Drosophila, suggesting an ancient evolutionary origin.
Collapse
Affiliation(s)
- Hyung Chul Lee
- Department of Cell and Developmental Biology, University College London, Gower Street, London, WC1E 6BT, UK.
- School of Biological Sciences and Technology, College of Natural Sciences, Chonnam National University, 77 Yongbong-ro, Gwangju, 61186, Korea.
| | - Nidia M M Oliveira
- Department of Cell and Developmental Biology, University College London, Gower Street, London, WC1E 6BT, UK
- College of Professional Services, Murdoch University, 90 South St, Murdoch, WA, 6150, Australia
| | - Cato Hastings
- Department of Cell and Developmental Biology, University College London, Gower Street, London, WC1E 6BT, UK
| | | | - Adam A Moverley
- Department of Cell and Developmental Biology, University College London, Gower Street, London, WC1E 6BT, UK
- Department of Cell and Developmental Biology, Institute for Regenerative Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Hui-Chun Lu
- Department of Cell and Developmental Biology, University College London, Gower Street, London, WC1E 6BT, UK
- Centre for Craniofacial & Regenerative Biology, Faculty of Dentistry, Oral and Craniofacial Sciences, King's College London, Guy's Tower, London, SE1 9RT, UK
| | - Yi Zheng
- Departments of Mechanical Engineering, Biomedical Engineering, and Cell & Developmental Biology, University of Michigan, Ann Arbor, MI, USA
- Department of Biomedical and Chemical Engineering, Syracuse University, Syracuse, NY, USA
- BioInspired Syracuse Institute for Materials and Living Systems, Syracuse University, Syracuse, NY, USA
| | - Elise L Wilby
- Department of Zoology, University of Cambridge, Downing Street, Cambridge, CB2 3EJ, UK
| | - Timothy T Weil
- Department of Zoology, University of Cambridge, Downing Street, Cambridge, CB2 3EJ, UK
| | - Karen M Page
- Department of Mathematics, University College London, Gower Street, London, WC1E 6BT, UK
| | - Jianping Fu
- Departments of Mechanical Engineering, Biomedical Engineering, and Cell & Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| | - Naomi Moris
- The Francis Crick Institute, 1 Midland Road, London, NW1 1AT, UK
| | - Claudio D Stern
- Department of Cell and Developmental Biology, University College London, Gower Street, London, WC1E 6BT, UK.
| |
Collapse
|
11
|
Wang S, DeLeon C, Sun W, Quake SR, Roth BL, Südhof TC. Alternative splicing of latrophilin-3 controls synapse formation. Nature 2024; 626:128-135. [PMID: 38233523 PMCID: PMC10830413 DOI: 10.1038/s41586-023-06913-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 11/29/2023] [Indexed: 01/19/2024]
Abstract
The assembly and specification of synapses in the brain is incompletely understood1-3. Latrophilin-3 (encoded by Adgrl3, also known as Lphn3)-a postsynaptic adhesion G-protein-coupled receptor-mediates synapse formation in the hippocampus4 but the mechanisms involved remain unclear. Here we show in mice that LPHN3 organizes synapses through a convergent dual-pathway mechanism: activation of Gαs signalling and recruitment of phase-separated postsynaptic protein scaffolds. We found that cell-type-specific alternative splicing of Lphn3 controls the LPHN3 G-protein-coupling mode, resulting in LPHN3 variants that predominantly signal through Gαs or Gα12/13. CRISPR-mediated manipulation of Lphn3 alternative splicing that shifts LPHN3 from a Gαs- to a Gα12/13-coupled mode impaired synaptic connectivity as severely as the overall deletion of Lphn3, suggesting that Gαs signalling by LPHN3 splice variants mediates synapse formation. Notably, Gαs-coupled, but not Gα12/13-coupled, splice variants of LPHN3 also recruit phase-transitioned postsynaptic protein scaffold condensates, such that these condensates are clustered by binding of presynaptic teneurin and FLRT ligands to LPHN3. Moreover, neuronal activity promotes alternative splicing of the synaptogenic Gαs-coupled variant of LPHN3. Together, these data suggest that activity-dependent alternative splicing of a key synaptic adhesion molecule controls synapse formation by parallel activation of two convergent pathways: Gαs signalling and clustered phase separation of postsynaptic protein scaffolds.
Collapse
Affiliation(s)
- Shuai Wang
- Department of Molecular and Cellular Physiology, Stanford University, Stanford, CA, USA.
- Howard Hughes Medical Institute, Stanford University, Stanford, CA, USA.
| | - Chelsea DeLeon
- Department of Pharmacology, UNC Chapel Hill School of Medicine, Chapel Hill, NC, USA
| | - Wenfei Sun
- Department of Molecular and Cellular Physiology, Stanford University, Stanford, CA, USA
- Department of Applied Physics, Stanford University, Stanford, CA, USA
- Department of Bioengineering, Stanford University, Stanford, CA, USA
| | - Stephen R Quake
- Department of Applied Physics, Stanford University, Stanford, CA, USA
- Department of Bioengineering, Stanford University, Stanford, CA, USA
- The Chan Zuckerberg Initiative, Redwood City, CA, USA
| | - Bryan L Roth
- Department of Pharmacology, UNC Chapel Hill School of Medicine, Chapel Hill, NC, USA
| | - Thomas C Südhof
- Department of Molecular and Cellular Physiology, Stanford University, Stanford, CA, USA.
- Howard Hughes Medical Institute, Stanford University, Stanford, CA, USA.
| |
Collapse
|
12
|
Zhang Y, Gao J, Li N, Xu P, Qu S, Cheng J, Wang M, Li X, Song Y, Xiao F, Yang X, Liu J, Hong H, Mu R, Li X, Wang Y, Xu H, Xie Y, Gao T, Wang G, Aa J. Targeting cAMP in D1-MSNs in the nucleus accumbens, a new rapid antidepressant strategy. Acta Pharm Sin B 2024; 14:667-681. [PMID: 38322327 PMCID: PMC10840425 DOI: 10.1016/j.apsb.2023.12.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 09/11/2023] [Accepted: 11/14/2023] [Indexed: 02/08/2024] Open
Abstract
Studies have suggested that the nucleus accumbens (NAc) is implicated in the pathophysiology of major depression; however, the regulatory strategy that targets the NAc to achieve an exclusive and outstanding anti-depression benefit has not been elucidated. Here, we identified a specific reduction of cyclic adenosine monophosphate (cAMP) in the subset of dopamine D1 receptor medium spiny neurons (D1-MSNs) in the NAc that promoted stress susceptibility, while the stimulation of cAMP production in NAc D1-MSNs efficiently rescued depression-like behaviors. Ketamine treatment enhanced cAMP both in D1-MSNs and dopamine D2 receptor medium spiny neurons (D2-MSNs) of depressed mice, however, the rapid antidepressant effect of ketamine solely depended on elevating cAMP in NAc D1-MSNs. We discovered that a higher dose of crocin markedly increased cAMP in the NAc and consistently relieved depression 24 h after oral administration, but not a lower dose. The fast onset property of crocin was verified through multicenter studies. Moreover, crocin specifically targeted at D1-MSN cAMP signaling in the NAc to relieve depression and had no effect on D2-MSN. These findings characterize a new strategy to achieve an exclusive and outstanding anti-depression benefit by elevating cAMP in D1-MSNs in the NAc, and provide a potential rapid antidepressant drug candidate, crocin.
Collapse
Affiliation(s)
- Yue Zhang
- Jiangsu Provincial Key Laboratory of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, Research Unit of PK–PD Based Bioactive Components and Pharmacodynamic Target Discovery of Natural Medicine of Chinese Academy of Medical Sciences, China Pharmaceutical University, Nanjing 210009, China
| | - Jingwen Gao
- Jiangsu Provincial Key Laboratory of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, Research Unit of PK–PD Based Bioactive Components and Pharmacodynamic Target Discovery of Natural Medicine of Chinese Academy of Medical Sciences, China Pharmaceutical University, Nanjing 210009, China
| | - Na Li
- Jiangsu Provincial Key Laboratory of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, Research Unit of PK–PD Based Bioactive Components and Pharmacodynamic Target Discovery of Natural Medicine of Chinese Academy of Medical Sciences, China Pharmaceutical University, Nanjing 210009, China
| | - Peng Xu
- Key Laboratory of Drug Monitoring and Control, Drug Intelligence and Forensic Center, Ministry of Public Security, Beijing 100193, China
| | - Shimeng Qu
- Jiangsu Provincial Key Laboratory of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, Research Unit of PK–PD Based Bioactive Components and Pharmacodynamic Target Discovery of Natural Medicine of Chinese Academy of Medical Sciences, China Pharmaceutical University, Nanjing 210009, China
| | - Jinqian Cheng
- Jiangsu Provincial Key Laboratory of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, Research Unit of PK–PD Based Bioactive Components and Pharmacodynamic Target Discovery of Natural Medicine of Chinese Academy of Medical Sciences, China Pharmaceutical University, Nanjing 210009, China
| | - Mingrui Wang
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Xueru Li
- School of Foreign Languages, China Pharmaceutical University, Nanjing 211198, China
| | - Yaheng Song
- Jiangsu Provincial Key Laboratory of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, Research Unit of PK–PD Based Bioactive Components and Pharmacodynamic Target Discovery of Natural Medicine of Chinese Academy of Medical Sciences, China Pharmaceutical University, Nanjing 210009, China
| | - Fan Xiao
- Jiangsu Provincial Key Laboratory of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, Research Unit of PK–PD Based Bioactive Components and Pharmacodynamic Target Discovery of Natural Medicine of Chinese Academy of Medical Sciences, China Pharmaceutical University, Nanjing 210009, China
| | - Xinyu Yang
- Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Jihong Liu
- Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Hao Hong
- Department of Pharmacology, China Pharmaceutical University, Nanjing 211198, China
| | - Ronghao Mu
- Department of Pharmacology, China Pharmaceutical University, Nanjing 211198, China
| | - Xiaotian Li
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Youmei Wang
- Key Laboratory of Drug Monitoring and Control, Drug Intelligence and Forensic Center, Ministry of Public Security, Beijing 100193, China
| | - Hui Xu
- Jiangsu Provincial Key Laboratory of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, Research Unit of PK–PD Based Bioactive Components and Pharmacodynamic Target Discovery of Natural Medicine of Chinese Academy of Medical Sciences, China Pharmaceutical University, Nanjing 210009, China
| | - Yuan Xie
- Jiangsu Provincial Key Laboratory of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, Research Unit of PK–PD Based Bioactive Components and Pharmacodynamic Target Discovery of Natural Medicine of Chinese Academy of Medical Sciences, China Pharmaceutical University, Nanjing 210009, China
| | - Tianming Gao
- Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Guangji Wang
- Jiangsu Provincial Key Laboratory of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, Research Unit of PK–PD Based Bioactive Components and Pharmacodynamic Target Discovery of Natural Medicine of Chinese Academy of Medical Sciences, China Pharmaceutical University, Nanjing 210009, China
| | - Jiye Aa
- Jiangsu Provincial Key Laboratory of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, Research Unit of PK–PD Based Bioactive Components and Pharmacodynamic Target Discovery of Natural Medicine of Chinese Academy of Medical Sciences, China Pharmaceutical University, Nanjing 210009, China
| |
Collapse
|
13
|
Khramova YV, Katrukha VA, Chebanenko VV, Kostyuk AI, Gorbunov NP, Panasenko OM, Sokolov AV, Bilan DS. Reactive Halogen Species: Role in Living Systems and Current Research Approaches. BIOCHEMISTRY. BIOKHIMIIA 2024; 89:S90-S111. [PMID: 38621746 DOI: 10.1134/s0006297924140062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 09/21/2023] [Accepted: 10/04/2023] [Indexed: 04/17/2024]
Abstract
Reactive halogen species (RHS) are highly reactive compounds that are normally required for regulation of immune response, inflammatory reactions, enzyme function, etc. At the same time, hyperproduction of highly reactive compounds leads to the development of various socially significant diseases - asthma, pulmonary hypertension, oncological and neurodegenerative diseases, retinopathy, and many others. The main sources of (pseudo)hypohalous acids are enzymes from the family of heme peroxidases - myeloperoxidase, lactoperoxidase, eosinophil peroxidase, and thyroid peroxidase. Main targets of these compounds are proteins and peptides, primarily methionine and cysteine residues. Due to the short lifetime, detection of RHS can be difficult. The most common approach is detection of myeloperoxidase, which is thought to reflect the amount of RHS produced, but these methods are indirect, and the results are often contradictory. The most promising approaches seem to be those that provide direct registration of highly reactive compounds themselves or products of their interaction with components of living cells, such as fluorescent dyes. However, even such methods have a number of limitations and can often be applied mainly for in vitro studies with cell culture. Detection of reactive halogen species in living organisms in real time is a particularly acute issue. The present review is devoted to RHS, their characteristics, chemical properties, peculiarities of interaction with components of living cells, and methods of their detection in living systems. Special attention is paid to the genetically encoded tools, which have been introduced recently and allow avoiding a number of difficulties when working with living systems.
Collapse
Affiliation(s)
- Yuliya V Khramova
- Faculty of Biology, Lomonosov Moscow State University, Moscow, 119234, Russia.
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, 117997, Russia
| | - Veronika A Katrukha
- Faculty of Biology, Lomonosov Moscow State University, Moscow, 119234, Russia
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, 117997, Russia
| | - Victoria V Chebanenko
- Faculty of Biology, Lomonosov Moscow State University, Moscow, 119234, Russia
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, 117997, Russia
| | - Alexander I Kostyuk
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, 117997, Russia
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, Moscow, 117997, Russia
| | | | - Oleg M Panasenko
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine, Federal Medical Biological Agency, Moscow, 119435, Russia
| | - Alexey V Sokolov
- Institute of Experimental Medicine, Saint-Petersburg, 197022, Russia.
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine, Federal Medical Biological Agency, Moscow, 119435, Russia
| | - Dmitry S Bilan
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, 117997, Russia.
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, Moscow, 117997, Russia
| |
Collapse
|
14
|
Qian Y, Celiker OT, Wang Z, Guner-Ataman B, Boyden ES. Temporally multiplexed imaging of dynamic signaling networks in living cells. Cell 2023; 186:5656-5672.e21. [PMID: 38029746 PMCID: PMC10843875 DOI: 10.1016/j.cell.2023.11.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 07/30/2023] [Accepted: 11/05/2023] [Indexed: 12/01/2023]
Abstract
Molecular signals interact in networks to mediate biological processes. To analyze these networks, it would be useful to image many signals at once, in the same living cell, using standard microscopes and genetically encoded fluorescent reporters. Here, we report temporally multiplexed imaging (TMI), which uses genetically encoded fluorescent proteins with different clocklike properties-such as reversibly photoswitchable fluorescent proteins with different switching kinetics-to represent different cellular signals. We linearly decompose a brief (few-second-long) trace of the fluorescence fluctuations, at each point in a cell, into a weighted sum of the traces exhibited by each fluorophore expressed in the cell. The weights then represent the signal amplitudes. We use TMI to analyze relationships between different kinase activities in individual cells, as well as between different cell-cycle signals, pointing toward broad utility throughout biology in the analysis of signal transduction cascades in living systems.
Collapse
Affiliation(s)
- Yong Qian
- McGovern Institute for Brain Research, Massachusetts Institute of Technology (MIT), Cambridge, MA 01239, USA
| | - Orhan T Celiker
- McGovern Institute for Brain Research, Massachusetts Institute of Technology (MIT), Cambridge, MA 01239, USA; Department of Electrical Engineering and Computer Science, MIT, Cambridge, MA 01239, USA
| | - Zeguan Wang
- McGovern Institute for Brain Research, Massachusetts Institute of Technology (MIT), Cambridge, MA 01239, USA; Department of Media Arts and Sciences, MIT, Cambridge, MA 01239, USA
| | - Burcu Guner-Ataman
- McGovern Institute for Brain Research, Massachusetts Institute of Technology (MIT), Cambridge, MA 01239, USA
| | - Edward S Boyden
- McGovern Institute for Brain Research, Massachusetts Institute of Technology (MIT), Cambridge, MA 01239, USA; Department of Media Arts and Sciences, MIT, Cambridge, MA 01239, USA; Department of Brain and Cognitive Sciences, MIT, Cambridge, MA 01239, USA; Department of Biological Engineering, MIT, Cambridge, MA 01239, USA; Koch Institute, MIT, Cambridge, MA 01239, USA; Howard Hughes Medical Institute, Cambridge, MA 01239, USA; Center for Neurobiological Engineering and K. Lisa Yang Center for Bionics at MIT, Cambridge, MA 01239, USA.
| |
Collapse
|
15
|
Cahill MK, Collard M, Tse V, Reitman ME, Etchenique R, Kirst C, Poskanzer KE. Network-level encoding of local neurotransmitters in cortical astrocytes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.01.568932. [PMID: 38106119 PMCID: PMC10723263 DOI: 10.1101/2023.12.01.568932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2023]
Abstract
Astrocytes-the most abundant non-neuronal cell type in the mammalian brain-are crucial circuit components that respond to and modulate neuronal activity via calcium (Ca 2+ ) signaling 1-8 . Astrocyte Ca 2+ activity is highly heterogeneous and occurs across multiple spatiotemporal scales: from fast, subcellular activity 3,4 to slow, synchronized activity that travels across connected astrocyte networks 9-11 . Furthermore, astrocyte network activity has been shown to influence a wide range of processes 5,8,12 . While astrocyte network activity has important implications for neuronal circuit function, the inputs that drive astrocyte network dynamics remain unclear. Here we used ex vivo and in vivo two-photon Ca 2+ imaging of astrocytes while mimicking neuronal neurotransmitter inputs at multiple spatiotemporal scales. We find that brief, subcellular inputs of GABA and glutamate lead to widespread, long-lasting astrocyte Ca 2+ responses beyond an individual stimulated cell. Further, we find that a key subset of Ca 2+ activity-propagative events-differentiates astrocyte network responses to these two major neurotransmitters, and gates responses to future inputs. Together, our results demonstrate that local, transient neurotransmitter inputs are encoded by broad cortical astrocyte networks over the course of minutes, contributing to accumulating evidence across multiple model organisms that significant astrocyte-neuron communication occurs across slow, network-level spatiotemporal scales 13-15 . We anticipate that this study will be a starting point for future studies investigating the link between specific astrocyte Ca 2+ activity and specific astrocyte functional outputs, which could build a consistent framework for astrocytic modulation of neuronal activity.
Collapse
|
16
|
Chen Y, Pang S, Li J, Lu Y, Gao C, Xiao Y, Chen M, Wang M, Ren X. Genetically encoded protein sensors for metal ion detection in biological systems: a review and bibliometric analysis. Analyst 2023; 148:5564-5581. [PMID: 37872814 DOI: 10.1039/d3an01412f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2023]
Abstract
Metal ions are indispensable elements in living organisms and are associated with regulating various biological processes. An imbalance in metal ion content can lead to disorders in normal physiological functions of the human body and cause various diseases. Genetically encoded fluorescent protein sensors have the advantages of low biotoxicity, high specificity, and a long imaging time in vivo and have become a powerful tool to visualize or quantify the concentration level of biomolecules in vivo and in vitro, temporal and spatial distribution, and life activity process. This review analyzes the development status and current research hotspots in the field of genetically encoded fluorescent protein sensors by bibliometric analysis. Based on the results of bibliometric analysis, the research progress of genetically encoded fluorescent protein sensors for metal ion detection is reviewed, and the construction strategies, physicochemical properties, and applications of such sensors in biological imaging are summarized.
Collapse
Affiliation(s)
- Yuxueyuan Chen
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China
- College of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - ShuChao Pang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China
| | - Jingya Li
- College of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Yun Lu
- College of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Chenxia Gao
- College of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Yanyu Xiao
- College of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Meiling Chen
- College of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
- Tianjin Key Laboratory of Therapeutic Substance of Traditional Chinese Medicine, Tianjin 301617, China.
| | - Meng Wang
- State Key Laboratory of Component-based Chinese Medicine, Tianjin 301617, China
| | - Xiaoliang Ren
- College of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
- Tianjin Key Laboratory of Therapeutic Substance of Traditional Chinese Medicine, Tianjin 301617, China.
| |
Collapse
|
17
|
Bosquez-Berger T, Gudorf JA, Kuntz CP, Desmond JA, Schlebach JP, VanNieuwenhze MS, Straiker A. Structure-Activity Relationship Study of Cannabidiol-Based Analogs as Negative Allosteric Modulators of the μ-Opioid Receptor. J Med Chem 2023; 66:9466-9494. [PMID: 37437224 PMCID: PMC11299522 DOI: 10.1021/acs.jmedchem.3c00061] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/14/2023]
Abstract
The US faces an unprecedented surge in fatal drug overdoses. Naloxone, the only antidote for opiate overdose, competes at the mu opioid receptor (μOR) orthosteric site. Naloxone struggles against fentanyl-class synthetic opioids that now cause ∼80% of deaths. Negative allosteric modulators (NAMs) targeting secondary sites may noncompetitively downregulate μOR activation. (-)-Cannabidiol ((-)-CBD) is a candidate μOR NAM. To explore its therapeutic potential, we evaluated the structure-activity relationships among CBD analogs to identify NAMs with increased potency. Using a cyclic AMP assay, we characterize reversal of μOR activation by 15 CBD analogs, several of which proved more potent than (-)-CBD. Comparative docking investigations suggest that potent compounds interact with a putative allosteric pocket to stabilize the inactive μOR conformation. Finally, these compounds enhance naloxone displacement of fentanyl from the orthosteric site. Our results suggest that CBD analogs offer considerable potential for the development of next-generation antidotes for opioid overdose.
Collapse
Affiliation(s)
- Taryn Bosquez-Berger
- Gill Center for Biomolecular Science, Department of Psychological and Brain Sciences, Program in Neuroscience, Indiana University, Bloomington, Indiana 47405, United States
| | - Jessica A Gudorf
- Department of Chemistry, Indiana University, Bloomington, Indiana 47405, United States
| | - Charles P Kuntz
- Department of Chemistry, Indiana University, Bloomington, Indiana 47405, United States
| | - Jacob A Desmond
- Department of Chemistry, Indiana University, Bloomington, Indiana 47405, United States
| | - Jonathan P Schlebach
- Department of Chemistry, Indiana University, Bloomington, Indiana 47405, United States
| | | | - Alex Straiker
- Gill Center for Biomolecular Science, Department of Psychological and Brain Sciences, Program in Neuroscience, Indiana University, Bloomington, Indiana 47405, United States
| |
Collapse
|
18
|
Straiker A, Dvorakova M, Bosquez-Berger T, Blahos J, Mackie K. A collection of cannabinoid-related negative findings from autaptic hippocampal neurons. Sci Rep 2023; 13:9610. [PMID: 37311900 PMCID: PMC10264370 DOI: 10.1038/s41598-023-36710-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Accepted: 06/08/2023] [Indexed: 06/15/2023] Open
Abstract
Autaptic hippocampal neurons are an architecturally simple model of neurotransmission that express several forms of cannabinoid signaling. Over the past twenty years this model has proven valuable for studies ranging from enzymatic control of endocannabinoid production and breakdown, to CB1 receptor structure/function, to CB2 signaling, understanding 'spice' (synthetic cannabinoid) pharmacology, and more. However, while studying cannabinoid signaling in these neurons, we have occasionally encountered what one might call 'interesting negatives', valid and informative findings in the context of our experimental design that, given the nature of scientific publishing, may not otherwise find their way into the scientific literature. In autaptic hippocampal neurons we have found that: (1) The fatty acid binding protein (FABP) blocker SBFI-26 does not alter CB1-mediated neuroplasticity. (2) 1-AG signals poorly relative to 2-AG in autaptic neurons. (3) Indomethacin is not a CB1 PAM in autaptic neurons. (4) The CB1-associated protein SGIP1a is not necessary for CB1 desensitization. We are presenting these negative or perplexing findings in the hope that they will prove beneficial to other laboratories and elicit fruitful discussions regarding their relevance and significance.
Collapse
Affiliation(s)
- Alex Straiker
- Department of Psychological and Brain Sciences, Gill Center for Biomolecular Science, Program in Neuroscience, Indiana University, Bloomington, IN, 47405, USA.
| | - Michaela Dvorakova
- Department of Psychological and Brain Sciences, Gill Center for Biomolecular Science, Program in Neuroscience, Indiana University, Bloomington, IN, 47405, USA
| | - Taryn Bosquez-Berger
- Department of Psychological and Brain Sciences, Gill Center for Biomolecular Science, Program in Neuroscience, Indiana University, Bloomington, IN, 47405, USA
| | - Jaroslav Blahos
- Department of Molecular Pharmacology, Institute of Molecular Genetics of the Czech Academy of Sciences, Videnska 1083, 142 20, Prague 4, Czech Republic
| | - Ken Mackie
- Department of Psychological and Brain Sciences, Gill Center for Biomolecular Science, Program in Neuroscience, Indiana University, Bloomington, IN, 47405, USA
| |
Collapse
|
19
|
Hiasa S, Fujimori T, Aiki S, Ueda H, Tsuboi T, Kitaguchi T. Development of green fluorescent protein-based cAMP indicators for covering a wide range of cAMP concentrations. RSC Adv 2023; 13:15514-15520. [PMID: 37223420 PMCID: PMC10201545 DOI: 10.1039/d3ra01390a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Accepted: 05/12/2023] [Indexed: 05/25/2023] Open
Abstract
There is a wide range in the concentration of intracellular cyclic adenosine 3',5'-monophosphate (cAMP), which mediates specific effects as a second messenger in pathways affecting many physiological processes. Here, we developed green fluorescent cAMP indicators, named Green Falcan (Green fluorescent protein-based indicator visualizing cAMP dynamics) with various EC50 values (0.3, 1, 3, 10 μM) for covering the wide range of intracellular cAMP concentrations. The fluorescence intensity of Green Falcans increased in a cAMP dose-dependent manner, with a dynamic range of over 3-fold. Green Falcans showed a high specificity for cAMP over its structural analogues. When we expressed Green Falcans in HeLa cells, these indicators were applicable for visualization of cAMP dynamics in the low concentration range compared to the previously developed cAMP indicators, and visualized distinct kinetics of cAMP in various pathways with high spatiotemporal resolution in living cells. Furthermore, we demonstrated that Green Falcans are applicable to dual-color imaging with R-GECO, a red fluorescent Ca2+ indicator, in the cytoplasm and the nucleus. This study shows that Green Falcans open up a new avenue for understanding hierarchal and cooperative interactions with other molecules in various cAMP signaling pathways by multi-color imaging.
Collapse
Affiliation(s)
- Sohei Hiasa
- School of Life Science and Technology, Department of Life Science and Technology, Tokyo Institute of Technology 4259 Nagatsuta-cho, Midori-ku Yokohama-shi Kanagawa 226-8501 Japan
| | - Takeru Fujimori
- School of Life Science and Technology, Department of Life Science and Technology, Tokyo Institute of Technology 4259 Nagatsuta-cho, Midori-ku Yokohama-shi Kanagawa 226-8501 Japan
| | - Saki Aiki
- Department of Life Sciences, Graduate School of Arts and Sciences, The University of Tokyo 3-8-1 Komaba, Meguro-ku Tokyo 153-8902 Japan
| | - Hiroshi Ueda
- Laboratory for Chemistry and Life Science, Institute of Innovative Research, Tokyo Institute of Technology 4259 Nagatsuta-cho, Midori-ku Yokohama-shi Kanagawa 226-8503 Japan
| | - Takashi Tsuboi
- Department of Life Sciences, Graduate School of Arts and Sciences, The University of Tokyo 3-8-1 Komaba, Meguro-ku Tokyo 153-8902 Japan
| | - Tetsuya Kitaguchi
- Laboratory for Chemistry and Life Science, Institute of Innovative Research, Tokyo Institute of Technology 4259 Nagatsuta-cho, Midori-ku Yokohama-shi Kanagawa 226-8503 Japan
| |
Collapse
|
20
|
Paolocci E, Zaccolo M. Compartmentalised cAMP signalling in the primary cilium. Front Physiol 2023; 14:1187134. [PMID: 37256063 PMCID: PMC10226274 DOI: 10.3389/fphys.2023.1187134] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Accepted: 04/25/2023] [Indexed: 06/01/2023] Open
Abstract
cAMP is a universal second messenger that relies on precise spatio-temporal regulation to control varied, and often opposing, cellular functions. This is achieved via selective activation of effectors embedded in multiprotein complexes, or signalosomes, that reside at distinct subcellular locations. cAMP is also one of many pathways known to operate within the primary cilium. Dysfunction of ciliary signaling leads to a class of diseases known as ciliopathies. In Autosomal Dominant Polycystic Kidney Disease (ADPKD), a ciliopathy characterized by the formation of fluid-filled kidney cysts, upregulation of cAMP signaling is known to drive cystogenesis. For decades it has been debated whether the primary cilium is an independent cAMP sub-compartment, or whether it shares a diffusible pool of cAMP with the cell body. Recent studies now suggest it is a specific pool of cAMP generated in the cilium that propels cyst formation in ADPKD, supporting the notion that this antenna-like organelle is a compartment within which cAMP signaling occurs independently from cAMP signaling in the bulk cytosol. Here we present examples of cAMP function in the cilium which suggest this mysterious organelle is home to more than one cAMP signalosome. We review evidence that ciliary membrane localization of G-Protein Coupled Receptors (GPCRs) determines their downstream function and discuss how optogenetic tools have contributed to establish that cAMP generated in the primary cilium can drive cystogenesis.
Collapse
|
21
|
Xu X, Shonberg J, Kaindl J, Clark MJ, Stößel A, Maul L, Mayer D, Hübner H, Hirata K, Venkatakrishnan AJ, Dror RO, Kobilka BK, Sunahara RK, Liu X, Gmeiner P. Constrained catecholamines gain β 2AR selectivity through allosteric effects on pocket dynamics. Nat Commun 2023; 14:2138. [PMID: 37059717 PMCID: PMC10104803 DOI: 10.1038/s41467-023-37808-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 03/30/2023] [Indexed: 04/16/2023] Open
Abstract
G protein-coupled receptors (GPCRs) within the same subfamily often share high homology in their orthosteric pocket and therefore pose challenges to drug development. The amino acids that form the orthosteric binding pocket for epinephrine and norepinephrine in the β1 and β2 adrenergic receptors (β1AR and β2AR) are identical. Here, to examine the effect of conformational restriction on ligand binding kinetics, we synthesized a constrained form of epinephrine. Surprisingly, the constrained epinephrine exhibits over 100-fold selectivity for the β2AR over the β1AR. We provide evidence that the selectivity may be due to reduced ligand flexibility that enhances the association rate for the β2AR, as well as a less stable binding pocket for constrained epinephrine in the β1AR. The differences in the amino acid sequence of the extracellular vestibule of the β1AR allosterically alter the shape and stability of the binding pocket, resulting in a marked difference in affinity compared to the β2AR. These studies suggest that for receptors containing identical binding pocket residues, the binding selectivity may be influenced in an allosteric manner by surrounding residues, like those of the extracellular loops (ECLs) that form the vestibule. Exploiting these allosteric influences may facilitate the development of more subtype-selective ligands for GPCRs.
Collapse
Affiliation(s)
- Xinyu Xu
- State Key laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, School of Pharmaceutical Sciences, Tsinghua University, Beijing, 100084, China
- Beijing Frontier Research Center for Biological Structure, Beijing Advanced Innovation Center for Structural Biology, Tsinghua University, Beijing, 100084, China
| | - Jeremy Shonberg
- Department of Chemistry and Pharmacy, Medicinal Chemistry, Friedrich-Alexander University Erlangen-Nurnberg, Nikolaus-Fiebiger-Straße 10, 91058, Erlangen, Germany
| | - Jonas Kaindl
- Department of Chemistry and Pharmacy, Medicinal Chemistry, Friedrich-Alexander University Erlangen-Nurnberg, Nikolaus-Fiebiger-Straße 10, 91058, Erlangen, Germany
| | - Mary J Clark
- Department of Pharmacology, University of California San Diego School of Medicine, 9500 Gilman Drive, La Jolla, California, 92093, USA
| | - Anne Stößel
- Department of Chemistry and Pharmacy, Medicinal Chemistry, Friedrich-Alexander University Erlangen-Nurnberg, Nikolaus-Fiebiger-Straße 10, 91058, Erlangen, Germany
| | - Luis Maul
- Department of Chemistry and Pharmacy, Medicinal Chemistry, Friedrich-Alexander University Erlangen-Nurnberg, Nikolaus-Fiebiger-Straße 10, 91058, Erlangen, Germany
| | - Daniel Mayer
- Department of Pharmacology, University of California San Diego School of Medicine, 9500 Gilman Drive, La Jolla, California, 92093, USA
| | - Harald Hübner
- Department of Chemistry and Pharmacy, Medicinal Chemistry, Friedrich-Alexander University Erlangen-Nurnberg, Nikolaus-Fiebiger-Straße 10, 91058, Erlangen, Germany
| | - Kunio Hirata
- Advanced Photon Technology Division, Research Infrastructure Group, SR Life Science Instrumentation Unit, RIKEN/SPring-8 Center, 1-1-1 Kouto Sayo-cho Sayo-gun, Hyogo, 679-5148, Japan
- Precursory Research for Embryonic Science and Technology (PRESTO), Japan Science and Technology Agency, 4-1-8 Honcho, Kawaguchi, Saitama, 332-0012, Japan
| | - A J Venkatakrishnan
- Department of Computer Science, Stanford University, Stanford, CA, 94305, USA
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA, 94305, USA
- Department of Structural Biology, Stanford University School of Medicine, Stanford, CA, 94305, USA
- Institute for Computational and Mathematical Engineering, Stanford University, Stanford, CA, 94305, USA
| | - Ron O Dror
- Department of Computer Science, Stanford University, Stanford, CA, 94305, USA
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA, 94305, USA
- Department of Structural Biology, Stanford University School of Medicine, Stanford, CA, 94305, USA
- Institute for Computational and Mathematical Engineering, Stanford University, Stanford, CA, 94305, USA
| | - Brian K Kobilka
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA, 94305, USA.
| | - Roger K Sunahara
- Department of Pharmacology, University of California San Diego School of Medicine, 9500 Gilman Drive, La Jolla, California, 92093, USA.
| | - Xiangyu Liu
- State Key laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, School of Pharmaceutical Sciences, Tsinghua University, Beijing, 100084, China.
- Beijing Frontier Research Center for Biological Structure, Beijing Advanced Innovation Center for Structural Biology, Tsinghua University, Beijing, 100084, China.
- Beijing Key Laboratory of Cardiovascular Receptors Research, Peking University, Beijing, China.
| | - Peter Gmeiner
- Department of Chemistry and Pharmacy, Medicinal Chemistry, Friedrich-Alexander University Erlangen-Nurnberg, Nikolaus-Fiebiger-Straße 10, 91058, Erlangen, Germany.
| |
Collapse
|
22
|
Formozov A, Dieter A, Wiegert JS. A flexible and versatile system for multi-color fiber photometry and optogenetic manipulation. CELL REPORTS METHODS 2023; 3:100418. [PMID: 37056369 PMCID: PMC10088095 DOI: 10.1016/j.crmeth.2023.100418] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 12/20/2022] [Accepted: 02/08/2023] [Indexed: 03/09/2023]
Abstract
Here, we present simultaneous fiber photometry recordings and optogenetic stimulation based on a multimode fused fiber coupler for both light delivery and collection without the need for dichroic beam splitters. In combination with a multi-color light source and appropriate optical filters, our approach offers remarkable flexibility in experimental design and facilitates the exploration of new molecular tools in vivo at minimal cost. We demonstrate straightforward re-configuration of the setup to operate with green, red, and near-infrared calcium indicators with or without simultaneous optogenetic stimulation and further explore the multi-color photometry capabilities of the system. The ease of assembly, operation, characterization, and customization of this platform holds the potential to foster the development of experimental strategies for multi-color fused fiber photometry combined with optogenetics far beyond its current state.
Collapse
Affiliation(s)
- Andrey Formozov
- Research Group Synaptic Wiring and Information Processing, Center for Molecular Neurobiology Hamburg, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
- Department of Neurophysiology, MCTN, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
| | - Alexander Dieter
- Research Group Synaptic Wiring and Information Processing, Center for Molecular Neurobiology Hamburg, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
- Department of Neurophysiology, MCTN, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
| | - J. Simon Wiegert
- Research Group Synaptic Wiring and Information Processing, Center for Molecular Neurobiology Hamburg, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
- Department of Neurophysiology, MCTN, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
| |
Collapse
|
23
|
Astrocyte heterogeneity and interactions with local neural circuits. Essays Biochem 2023; 67:93-106. [PMID: 36748397 PMCID: PMC10011406 DOI: 10.1042/ebc20220136] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 01/09/2023] [Accepted: 01/09/2023] [Indexed: 02/08/2023]
Abstract
Astrocytes are ubiquitous within the central nervous system (CNS). These cells possess many individual processes which extend out into the neuropil, where they interact with a variety of other cell types, including neurons at synapses. Astrocytes are now known to be active players in all aspects of the synaptic life cycle, including synapse formation and elimination, synapse maturation, maintenance of synaptic homeostasis and modulation of synaptic transmission. Traditionally, astrocytes have been studied as a homogeneous group of cells. However, recent studies have uncovered a surprising degree of heterogeneity in their development and function, suggesting that astrocytes may be matched to neurons to support local circuits. Hence, a better understanding of astrocyte heterogeneity and its implications are needed to understand brain function.
Collapse
|
24
|
Harada K, Takashima M, Kitaguchi T, Tsuboi T. F-actin determines the time-dependent shift in docking dynamics of glucagon-like peptide-1 granules upon stimulation of secretion. FEBS Lett 2023; 597:657-671. [PMID: 36694275 DOI: 10.1002/1873-3468.14580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 01/09/2023] [Accepted: 01/09/2023] [Indexed: 01/26/2023]
Abstract
Although exocytosis can be categorized into several forms based on docking dynamics, temporal regulatory mechanisms of the exocytotic forms are unclear. We explored the dynamics of glucagon-like peptide-1 (GLP-1) exocytosis in murine GLUTag cells (GLP-1-secreting enteroendocrine L-cells) upon stimulation with deoxycholic acid (DCA) or high K+ to elucidate the mechanisms regulating the balance between the different types of exocytotic forms (pre-docked with the plasma membrane before stimulation; docked after stimulation and subsequently fused; or rapidly recruited and fused after stimulation, without stable docking). GLP-1 exocytosis showed a biphasic pattern, and we found that most exocytosis was from the pre-docked granules with the plasma membrane before stimulation, or granules rapidly fused to the plasma membrane without docking after stimulation. In contrast, granules docked with the plasma membrane after stimuli and eventually fused were predominant thereafter. Inhibition of actin polymerization suppressed exocytosis of the pre-docked granules. These results suggest that the docking dynamics of GLP-1 granules shows a time-dependent biphasic shift, which is determined by interaction with F-actin.
Collapse
Affiliation(s)
- Kazuki Harada
- Department of Life Sciences, Graduate School of Arts and Sciences, The University of Tokyo, Japan
| | - Maoko Takashima
- Department of Life Sciences, Graduate School of Arts and Sciences, The University of Tokyo, Japan
| | - Tetsuya Kitaguchi
- Laboratory for Chemistry and Life Science, Institute of Innovative Research, Tokyo Institute of Technology, Yokohama, Japan
| | - Takashi Tsuboi
- Department of Life Sciences, Graduate School of Arts and Sciences, The University of Tokyo, Japan
| |
Collapse
|
25
|
Shannonhouse J, Gomez R, Son H, Zhang Y, Kim YS. In Vivo Calcium Imaging of Neuronal Ensembles in Networks of Primary Sensory Neurons in Intact Dorsal Root Ganglia. J Vis Exp 2023:10.3791/64826. [PMID: 36847407 PMCID: PMC10785773 DOI: 10.3791/64826] [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] [Indexed: 02/12/2023] Open
Abstract
Ca2+ imaging can be used as a proxy for cellular activity, including action potentials and various signaling mechanisms involving Ca2+ entry into the cytoplasm or the release of intracellular Ca2+ stores. Pirt-GCaMP3-based Ca2+ imaging of primary sensory neurons of the dorsal root ganglion (DRG) in mice offers the advantage of simultaneous measurement of a large number of cells. Up to 1,800 neurons can be monitored, allowing neuronal networks and somatosensory processes to be studied as an ensemble in their normal physiological context at a populational level in vivo. The large number of neurons monitored allows the detection of activity patterns that would be challenging to detect using other methods. Stimuli can be applied to the mouse hindpaw, allowing the direct effects of stimuli on the DRG neuron ensemble to be studied. The number of neurons producing Ca2+ transients as well as the amplitude of Ca2+ transients indicates sensitivity to specific sensory modalities. The diameter of neurons provides evidence of activated fiber types (non-noxious mechano vs. noxious pain fibers, Aβ, Aδ, and C fibers). Neurons expressing specific receptors can be genetically labeled with td-Tomato and specific Cre recombinases together with Pirt-GCaMP. Therefore, Pirt-GCaMP3 Ca2+ imaging of DRG provides a powerful tool and model for the analysis of specific sensory modalities and neuron subtypes acting as an ensemble at the populational level to study pain, itch, touch, and other somatosensory signals.
Collapse
Affiliation(s)
- John Shannonhouse
- Department of Oral & Maxillofacial Surgery, School of Dentistry, University of Texas Health Science Center at San Antonio
| | - Ruben Gomez
- Department of Oral & Maxillofacial Surgery, School of Dentistry, University of Texas Health Science Center at San Antonio
| | - Hyeonwi Son
- Department of Oral & Maxillofacial Surgery, School of Dentistry, University of Texas Health Science Center at San Antonio
| | - Yan Zhang
- Department of Oral & Maxillofacial Surgery, School of Dentistry, University of Texas Health Science Center at San Antonio
| | - Yu Shin Kim
- Department of Oral & Maxillofacial Surgery, School of Dentistry, University of Texas Health Science Center at San Antonio; Programs in Integrated Biomedical Sciences, Translational Sciences, Biomedical Engineering, Radiological Sciences, University of Texas Health Science Center at San Antonio;
| |
Collapse
|
26
|
Day-Cooney J, Dalangin R, Zhong H, Mao T. Genetically encoded fluorescent sensors for imaging neuronal dynamics in vivo. J Neurochem 2023; 164:284-308. [PMID: 35285522 PMCID: PMC11322610 DOI: 10.1111/jnc.15608] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 02/14/2022] [Accepted: 02/25/2022] [Indexed: 11/29/2022]
Abstract
The brain relies on many forms of dynamic activities in individual neurons, from synaptic transmission to electrical activity and intracellular signaling events. Monitoring these neuronal activities with high spatiotemporal resolution in the context of animal behavior is a necessary step to achieve a mechanistic understanding of brain function. With the rapid development and dissemination of highly optimized genetically encoded fluorescent sensors, a growing number of brain activities can now be visualized in vivo. To date, cellular calcium imaging, which has been largely used as a proxy for electrical activity, has become a mainstay in systems neuroscience. While challenges remain, voltage imaging of neural populations is now possible. In addition, it is becoming increasingly practical to image over half a dozen neurotransmitters, as well as certain intracellular signaling and metabolic activities. These new capabilities enable neuroscientists to test previously unattainable hypotheses and questions. This review summarizes recent progress in the development and delivery of genetically encoded fluorescent sensors, and highlights example applications in the context of in vivo imaging.
Collapse
Affiliation(s)
- Julian Day-Cooney
- Vollum Institute, Oregon Health and Science University, Portland, Oregon, USA
| | - Rochelin Dalangin
- Department of Biochemistry and Molecular Medicine, University of California, Davis, Davis, California, USA
| | - Haining Zhong
- Vollum Institute, Oregon Health and Science University, Portland, Oregon, USA
| | - Tianyi Mao
- Vollum Institute, Oregon Health and Science University, Portland, Oregon, USA
| |
Collapse
|
27
|
Natsubori A, Hirai S, Kwon S, Ono D, Deng F, Wan J, Miyazawa M, Kojima T, Okado H, Karashima A, Li Y, Tanaka KF, Honda M. Serotonergic neurons control cortical neuronal intracellular energy dynamics by modulating astrocyte-neuron lactate shuttle. iScience 2023; 26:105830. [PMID: 36713262 PMCID: PMC9881222 DOI: 10.1016/j.isci.2022.105830] [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: 02/13/2022] [Revised: 08/15/2022] [Accepted: 12/12/2022] [Indexed: 01/07/2023] Open
Abstract
The central serotonergic system has multiple roles in animal physiology and behavior, including sleep-wake control. However, its function in controlling brain energy metabolism according to the state of animals remains undetermined. Through in vivo monitoring of energy metabolites and signaling, we demonstrated that optogenetic activation of raphe serotonergic neurons increased cortical neuronal intracellular concentration of ATP, an indispensable cellular energy molecule, which was suppressed by inhibiting neuronal uptake of lactate derived from astrocytes. Raphe serotonergic neuronal activation induced cortical astrocytic Ca2+ and cAMP surges and increased extracellular lactate concentrations, suggesting the facilitation of lactate release from astrocytes. Furthermore, chemogenetic inhibition of raphe serotonergic neurons partly attenuated the increase in cortical neuronal intracellular ATP levels as arousal increased in mice. Serotonergic neuronal activation promoted an increase in cortical neuronal intracellular ATP levels, partly mediated by the facilitation of the astrocyte-neuron lactate shuttle, contributing to state-dependent optimization of neuronal intracellular energy levels.
Collapse
Affiliation(s)
- Akiyo Natsubori
- Sleep Disorders Project, Tokyo Metropolitan Institute of Medical Science, Setagaya-ku, Tokyo 156-8506, Japan,Corresponding author
| | - Shinobu Hirai
- Sleep Disorders Project, Tokyo Metropolitan Institute of Medical Science, Setagaya-ku, Tokyo 156-8506, Japan
| | - Soojin Kwon
- Sleep Disorders Project, Tokyo Metropolitan Institute of Medical Science, Setagaya-ku, Tokyo 156-8506, Japan
| | - Daisuke Ono
- Department of Neuroscience Ⅱ, Research Institute of Environmental Medicine, Nagoya University, Nagoya 464-8601, Japan,Department of Neural Regulation, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Fei Deng
- State Key Laboratory of Membrane Biology, Peking University School of Life Sciences, Beijing 100871, China,PKU-IDG/McGovern Institute for Brain Research, Beijing 100871, China
| | - Jinxia Wan
- State Key Laboratory of Membrane Biology, Peking University School of Life Sciences, Beijing 100871, China,PKU-IDG/McGovern Institute for Brain Research, Beijing 100871, China
| | - Momoka Miyazawa
- Sleep Disorders Project, Tokyo Metropolitan Institute of Medical Science, Setagaya-ku, Tokyo 156-8506, Japan,Faculty of Science Division Ⅱ, Tokyo University of Science, Shinjuku-ku, Tokyo 162-8601, Japan
| | - Takashi Kojima
- Sleep Disorders Project, Tokyo Metropolitan Institute of Medical Science, Setagaya-ku, Tokyo 156-8506, Japan
| | - Haruo Okado
- Sleep Disorders Project, Tokyo Metropolitan Institute of Medical Science, Setagaya-ku, Tokyo 156-8506, Japan
| | - Akihiro Karashima
- Department of Electronics, Graduate School of Engineering, Tohoku Institute of Technology, Sendai 982-8577, Japan
| | - Yulong Li
- State Key Laboratory of Membrane Biology, Peking University School of Life Sciences, Beijing 100871, China,PKU-IDG/McGovern Institute for Brain Research, Beijing 100871, China
| | - Kenji F. Tanaka
- Division of Brain Sciences, Institute for Advanced Medical Research, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Makoto Honda
- Sleep Disorders Project, Tokyo Metropolitan Institute of Medical Science, Setagaya-ku, Tokyo 156-8506, Japan
| |
Collapse
|
28
|
Rozenfeld M, Azoulay IS, Ben Kasus Nissim T, Stavsky A, Melamed M, Stutzmann G, Hershfinkel M, Kofman O, Sekler I. Essential role of the mitochondrial Na +/Ca 2+ exchanger NCLX in mediating PDE2-dependent neuronal survival and learning. Cell Rep 2022; 41:111772. [PMID: 36476859 PMCID: PMC10521900 DOI: 10.1016/j.celrep.2022.111772] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 07/06/2022] [Accepted: 11/10/2022] [Indexed: 12/12/2022] Open
Abstract
Impaired phosphodiesterase (PDE) function and mitochondrial Ca2+ (i.e., [Ca2+]m) lead to multiple health syndromes by an unknown pathway. Here, we fluorescently monitor robust [Ca2+]m efflux mediated by the mitochondrial Na+/Ca2+ exchanger NCLX in hippocampal neurons sequentially evoked by caffeine and depolarization. Surprisingly, neuronal depolarization-induced Ca2+ transients alone fail to evoke strong [Ca2+]m efflux in wild-type (WT) neurons. However, pre-treatment with the selective PDE2 inhibitor Bay 60-7550 effectively rescues [Ca2+]m efflux similarly to caffeine. Moreover, PDE2 acts by diminishing mitochondrial cAMP, thus promoting NCLX phosphorylation at its PKA site. We find that the protection of neurons against excitotoxic insults, conferred by PDE2 inhibition in WT neurons, is NCLX dependent. Finally, the administration of Bay 60-7550 enhances new object recognition in WT, but not in NCLX knockout (KO), mice. Our results identify a link between PDE and [Ca2+]m signaling that may provide effective therapy for cognitive and ischemic syndromes.
Collapse
Affiliation(s)
- Maya Rozenfeld
- Department of Physiology and Cell Biology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Ivana Savic Azoulay
- Department of Physiology and Cell Biology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Tsipi Ben Kasus Nissim
- Department of Physiology and Cell Biology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Alexandra Stavsky
- Department of Physiology and Cell Biology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Moran Melamed
- Department of Physiology and Cell Biology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Grace Stutzmann
- Rosalind Franklin University of Medicine and Science, Chicago Medical School, Center for Neurodegenerative Disease and Therapeutics, Chicago, IL, USA
| | - Michal Hershfinkel
- Department of Physiology and Cell Biology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Ora Kofman
- Department of Psychology, Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Israel Sekler
- Department of Physiology and Cell Biology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel.
| |
Collapse
|
29
|
Bosquez-Berger T, Wilson S, Iliopoulos-Tsoutsouvas C, Jiang S, Wager-Miller J, Nikas SP, Mackie KP, Makriyannis A, Straiker A. Differential Enantiomer-Specific Signaling of Cannabidiol at CB 1 Receptors. Mol Pharmacol 2022; 102:259-268. [PMID: 36153039 PMCID: PMC11033957 DOI: 10.1124/molpharm.121.000305] [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: 04/19/2021] [Accepted: 09/05/2022] [Indexed: 11/22/2022] Open
Abstract
The two main constituents of cannabis are Δ9-tetrahydrocannabinol (THC) and cannabidiol (CBD). While Δ9-THC pharmacology has been studied extensively, CBD-long considered inactive-is now the subject of vigorous research related to epilepsy, pain, and inflammation and is popularly embraced as a virtual cure-all. However, our understanding of CBD pharmacology remains limited, although CBD inhibits cannabinoid CB1 receptor signaling, likely as a negative allosteric modulator. Cannabis synthesizes (-)-CBD, but CBD can also exist as an enantiomer, (+)-CBD. We enantioselectively synthesized both CBD enantiomers using established conditions and describe here a new, practical, and reliable, NMR-based method for confirming the enantiomeric purity of two CBD enantiomers. We also investigated the pharmacology of (+)-CBD in autaptic hippocampal neurons, a well-characterized neuronal model of endogenous cannabinoid signaling, and in CHO-K1 cells. We report the inhibition constant for displacing CP55,940 at CB1 by (+)-CBD, is 5-fold lower than (-)-CBD. We find that (+)-CBD is ∼10 times more potent at inhibiting depolarization-induced suppression of excitation (DSE), a form of endogenous cannabinoid-mediated retrograde synaptic plasticity. (+)-CBD also inhibits CB1 suppression of cAMP accumulation but with less potency, indicating that the signaling profiles of the enantiomers differ in a pathway-specific manner. In addition, we report that (+)-CBD stereoselectively and potently activates the sphingosine-1 phosphate (S1P) receptors, S1P1 and S1P3 These results provide an attractive method for synthesizing and distinguishing enantiomers of CBD and related phytocannabinoids and provide further evidence that these enantiomers have their own unique and interesting signaling properties. SIGNIFICANCE STATEMENT: Cannabidiol (CBD) is the subject of considerable scientific and popular interest, but we know little of the enantiomers of CBD. We find that the enantiomer (+)-CBD is substantially more potent inhibitor of cannabinoid CB1 receptors and that it activates sphingosine-1-phosphate receptors in an enantiomer-specific manner; we have additionally developed an improved method for the synthesis of enantiomers of CBD and related compounds.
Collapse
Affiliation(s)
- Taryn Bosquez-Berger
- Gill Center for Biomolecular Science, Department of Psychological and Brain Sciences, Program in Neuroscience, Indiana University, Bloomington, Indiana (T.B., S.W., J.W.M., K.M., A.S.); and Center for Drug Discovery and Department of Pharmaceutical Sciences (C.I.T., S.P.N., A.M.) and Center for Drug Discovery and Department of Chemistry and Chemical Biology (S.J., A.M.), Northeastern University, Boston, Massachusetts
| | - Sierra Wilson
- Gill Center for Biomolecular Science, Department of Psychological and Brain Sciences, Program in Neuroscience, Indiana University, Bloomington, Indiana (T.B., S.W., J.W.M., K.M., A.S.); and Center for Drug Discovery and Department of Pharmaceutical Sciences (C.I.T., S.P.N., A.M.) and Center for Drug Discovery and Department of Chemistry and Chemical Biology (S.J., A.M.), Northeastern University, Boston, Massachusetts
| | - Christos Iliopoulos-Tsoutsouvas
- Gill Center for Biomolecular Science, Department of Psychological and Brain Sciences, Program in Neuroscience, Indiana University, Bloomington, Indiana (T.B., S.W., J.W.M., K.M., A.S.); and Center for Drug Discovery and Department of Pharmaceutical Sciences (C.I.T., S.P.N., A.M.) and Center for Drug Discovery and Department of Chemistry and Chemical Biology (S.J., A.M.), Northeastern University, Boston, Massachusetts
| | - Shan Jiang
- Gill Center for Biomolecular Science, Department of Psychological and Brain Sciences, Program in Neuroscience, Indiana University, Bloomington, Indiana (T.B., S.W., J.W.M., K.M., A.S.); and Center for Drug Discovery and Department of Pharmaceutical Sciences (C.I.T., S.P.N., A.M.) and Center for Drug Discovery and Department of Chemistry and Chemical Biology (S.J., A.M.), Northeastern University, Boston, Massachusetts
| | - Jim Wager-Miller
- Gill Center for Biomolecular Science, Department of Psychological and Brain Sciences, Program in Neuroscience, Indiana University, Bloomington, Indiana (T.B., S.W., J.W.M., K.M., A.S.); and Center for Drug Discovery and Department of Pharmaceutical Sciences (C.I.T., S.P.N., A.M.) and Center for Drug Discovery and Department of Chemistry and Chemical Biology (S.J., A.M.), Northeastern University, Boston, Massachusetts
| | - Spyros P Nikas
- Gill Center for Biomolecular Science, Department of Psychological and Brain Sciences, Program in Neuroscience, Indiana University, Bloomington, Indiana (T.B., S.W., J.W.M., K.M., A.S.); and Center for Drug Discovery and Department of Pharmaceutical Sciences (C.I.T., S.P.N., A.M.) and Center for Drug Discovery and Department of Chemistry and Chemical Biology (S.J., A.M.), Northeastern University, Boston, Massachusetts
| | - Ken P Mackie
- Gill Center for Biomolecular Science, Department of Psychological and Brain Sciences, Program in Neuroscience, Indiana University, Bloomington, Indiana (T.B., S.W., J.W.M., K.M., A.S.); and Center for Drug Discovery and Department of Pharmaceutical Sciences (C.I.T., S.P.N., A.M.) and Center for Drug Discovery and Department of Chemistry and Chemical Biology (S.J., A.M.), Northeastern University, Boston, Massachusetts
| | - Alexandros Makriyannis
- Gill Center for Biomolecular Science, Department of Psychological and Brain Sciences, Program in Neuroscience, Indiana University, Bloomington, Indiana (T.B., S.W., J.W.M., K.M., A.S.); and Center for Drug Discovery and Department of Pharmaceutical Sciences (C.I.T., S.P.N., A.M.) and Center for Drug Discovery and Department of Chemistry and Chemical Biology (S.J., A.M.), Northeastern University, Boston, Massachusetts
| | - Alex Straiker
- Gill Center for Biomolecular Science, Department of Psychological and Brain Sciences, Program in Neuroscience, Indiana University, Bloomington, Indiana (T.B., S.W., J.W.M., K.M., A.S.); and Center for Drug Discovery and Department of Pharmaceutical Sciences (C.I.T., S.P.N., A.M.) and Center for Drug Discovery and Department of Chemistry and Chemical Biology (S.J., A.M.), Northeastern University, Boston, Massachusetts
| |
Collapse
|
30
|
Sensitive genetically encoded sensors for population and subcellular imaging of cAMP in vivo. Nat Methods 2022; 19:1461-1471. [PMID: 36303019 PMCID: PMC10171401 DOI: 10.1038/s41592-022-01646-5] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2021] [Accepted: 09/09/2022] [Indexed: 01/07/2023]
Abstract
Cyclic adenosine monophosphate (cAMP) signaling integrates information from diverse G-protein-coupled receptors, such as neuromodulator receptors, to regulate pivotal biological processes in a cellular-specific and subcellular-specific manner. However, in vivo cellular-resolution imaging of cAMP dynamics remains challenging. Here, we screen existing genetically encoded cAMP sensors and further develop the best performer to derive three improved variants, called cAMPFIREs. Compared with their parental sensor, these sensors exhibit up to 10-fold increased sensitivity to cAMP and a cytosolic distribution. cAMPFIREs are compatible with both ratiometric and fluorescence lifetime imaging and can detect cAMP dynamics elicited by norepinephrine at physiologically relevant, nanomolar concentrations. Imaging of cAMPFIREs in awake mice reveals tonic levels of cAMP in cortical neurons that are associated with wakefulness, modulated by opioids, and differentially regulated across subcellular compartments. Furthermore, enforced locomotion elicits neuron-specific, bidirectional cAMP dynamics. cAMPFIREs also function in Drosophila. Overall, cAMPFIREs may have broad applicability for studying intracellular signaling in vivo.
Collapse
|
31
|
Hashizume R, Fujii H, Mehta S, Ota K, Qian Y, Zhu W, Drobizhev M, Nasu Y, Zhang J, Bito H, Campbell RE. A genetically encoded far-red fluorescent calcium ion biosensor derived from a biliverdin-binding protein. Protein Sci 2022; 31:e4440. [PMID: 36173169 PMCID: PMC9518226 DOI: 10.1002/pro.4440] [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: 07/04/2022] [Revised: 09/02/2022] [Accepted: 09/05/2022] [Indexed: 11/09/2022]
Abstract
Far-red and near-infrared (NIR) genetically encoded calcium ion (Ca2+ ) indicators (GECIs) are powerful tools for in vivo and multiplexed imaging of neural activity and cell signaling. Inspired by a previous report to engineer a far-red fluorescent protein (FP) from a biliverdin (BV)-binding NIR FP, we have developed a far-red fluorescent GECI, designated iBB-GECO1, from a previously reported NIR GECI. iBB-GECO1 exhibits a relatively high molecular brightness, an inverse response to Ca2+ with ΔF/Fmin = -13, and a near-optimal dissociation constant (Kd ) for Ca2+ of 105 nM. We demonstrate the utility of iBB-GECO1 for four-color multiplexed imaging in MIN6 cells and five-color imaging in HEK293T cells. Like other BV-binding GECIs, iBB-GECO1 did not give robust signals during in vivo imaging of neural activity in mice, but did provide promising results that will guide future engineering efforts. SIGNIFICANCE: Genetically encoded calcium ion (Ca2+ ) indicators (GECIs) compatible with common far-red laser lines (~630-640 nm) on commercial microscopes are of critical importance for their widespread application to deep-tissue multiplexed imaging of neural activity. In this study, we engineered a far-red excitable fluorescent GECI, designated iBB-GECO1, that exhibits a range of preferable specifications such as high brightness, large fluorescence response to Ca2+ , and compatibility with multiplexed imaging in mammalian cells.
Collapse
Affiliation(s)
- Rina Hashizume
- Department of Chemistry, School of ScienceThe University of Tokyo, Bunkyo‐kuTokyoJapan
| | - Hajime Fujii
- Department of Neurochemistry, Graduate School of MedicineThe University of Tokyo, Bunkyo‐kuTokyoJapan
| | - Sohum Mehta
- Department of PharmacologyUniversity of California San DiegoLa JollaCaliforniaUSA
| | - Keisuke Ota
- Department of Neurochemistry, Graduate School of MedicineThe University of Tokyo, Bunkyo‐kuTokyoJapan
| | - Yong Qian
- Department of ChemistryUniversity of AlbertaEdmontonAlbertaCanada
- McGovern Institute for Brain Research, MITCambridgeMassachusettsUSA
| | - Wenchao Zhu
- Department of Chemistry, School of ScienceThe University of Tokyo, Bunkyo‐kuTokyoJapan
| | - Mikhail Drobizhev
- Department of Microbiology and Cell BiologyMontana State UniversityBozemanMontanaUSA
| | - Yusuke Nasu
- Department of Chemistry, School of ScienceThe University of Tokyo, Bunkyo‐kuTokyoJapan
| | - Jin Zhang
- Department of PharmacologyUniversity of California San DiegoLa JollaCaliforniaUSA
| | - Haruhiko Bito
- Department of Neurochemistry, Graduate School of MedicineThe University of Tokyo, Bunkyo‐kuTokyoJapan
| | - Robert E. Campbell
- Department of Chemistry, School of ScienceThe University of Tokyo, Bunkyo‐kuTokyoJapan
- Department of ChemistryUniversity of AlbertaEdmontonAlbertaCanada
| |
Collapse
|
32
|
Pittolo S, Yokoyama S, Willoughby DD, Taylor CR, Reitman ME, Tse V, Wu Z, Etchenique R, Li Y, Poskanzer KE. Dopamine activates astrocytes in prefrontal cortex via α1-adrenergic receptors. Cell Rep 2022; 40:111426. [PMID: 36170823 PMCID: PMC9555850 DOI: 10.1016/j.celrep.2022.111426] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 07/19/2022] [Accepted: 09/08/2022] [Indexed: 12/31/2022] Open
Abstract
The prefrontal cortex (PFC) is a hub for cognitive control, and dopamine profoundly influences its functions. In other brain regions, astrocytes sense diverse neurotransmitters and neuromodulators and, in turn, orchestrate regulation of neuroactive substances. However, basic physiology of PFC astrocytes, including which neuromodulatory signals they respond to and how they contribute to PFC function, is unclear. Here, we characterize divergent signaling signatures in mouse astrocytes of the PFC and primary sensory cortex, which show differential responsiveness to locomotion. We find that PFC astrocytes express receptors for dopamine but are unresponsive through the Gs/Gi-cAMP pathway. Instead, fast calcium signals in PFC astrocytes are time locked to dopamine release and are mediated by α1-adrenergic receptors both ex vivo and in vivo. Further, we describe dopamine-triggered regulation of extracellular ATP at PFC astrocyte territories. Thus, we identify astrocytes as active players in dopaminergic signaling in the PFC, contributing to PFC function though neuromodulator receptor crosstalk. Pittolo et al. demonstrate that the neuromodulator dopamine targets astrocytes, a type of brain cell, via receptors specific to another neuromodulator—norepinephrine. This study provides groundwork on how dopamine affects non-neuronal brain cells and suggests that crosstalk between neuromodulatory pathways occurs in vivo, with possible clinical implications.
Collapse
Affiliation(s)
- Silvia Pittolo
- Department of Biochemistry & Biophysics, University of California, San Francisco, San Francisco, CA, USA
| | - Sae Yokoyama
- Department of Biochemistry & Biophysics, University of California, San Francisco, San Francisco, CA, USA
| | - Drew D Willoughby
- Department of Biochemistry & Biophysics, University of California, San Francisco, San Francisco, CA, USA; Neuroscience Graduate Program, University of California, San Francisco, San Francisco, CA, USA
| | - Charlotte R Taylor
- Department of Biochemistry & Biophysics, University of California, San Francisco, San Francisco, CA, USA; Neuroscience Graduate Program, University of California, San Francisco, San Francisco, CA, USA
| | - Michael E Reitman
- Department of Biochemistry & Biophysics, University of California, San Francisco, San Francisco, CA, USA; Neuroscience Graduate Program, University of California, San Francisco, San Francisco, CA, USA
| | - Vincent Tse
- Department of Biochemistry & Biophysics, University of California, San Francisco, San Francisco, CA, USA
| | - Zhaofa Wu
- State Key Laboratory of Membrane Biology, Peking University School of Life Sciences, Beijing 100871, China; Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100871, China
| | - Roberto Etchenique
- Departamento de Química Inorgánica, Analítica y Química Física, INQUIMAE, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, CONICET, Intendente Güiraldes 2160, Ciudad Universitaria, Pabellón 2, C1428EGA, Buenos Aires, Argentina
| | - Yulong Li
- State Key Laboratory of Membrane Biology, Peking University School of Life Sciences, Beijing 100871, China; Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100871, China
| | - Kira E Poskanzer
- Department of Biochemistry & Biophysics, University of California, San Francisco, San Francisco, CA, USA; Neuroscience Graduate Program, University of California, San Francisco, San Francisco, CA, USA; Kavli Institute for Fundamental Neuroscience, San Francisco, CA, USA.
| |
Collapse
|
33
|
Wang L, Wu C, Peng W, Zhou Z, Zeng J, Li X, Yang Y, Yu S, Zou Y, Huang M, Liu C, Chen Y, Li Y, Ti P, Liu W, Gao Y, Zheng W, Zhong H, Gao S, Lu Z, Ren PG, Ng HL, He J, Chen S, Xu M, Li Y, Chu J. A high-performance genetically encoded fluorescent indicator for in vivo cAMP imaging. Nat Commun 2022; 13:5363. [PMID: 36097007 PMCID: PMC9468011 DOI: 10.1038/s41467-022-32994-7] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Accepted: 08/24/2022] [Indexed: 11/25/2022] Open
Abstract
cAMP is a key second messenger that regulates diverse cellular functions including neural plasticity. However, the spatiotemporal dynamics of intracellular cAMP in intact organisms are largely unknown due to low sensitivity and/or brightness of current genetically encoded fluorescent cAMP indicators. Here, we report the development of the new circularly permuted GFP (cpGFP)-based cAMP indicator G-Flamp1, which exhibits a large fluorescence increase (a maximum ΔF/F0 of 1100% in HEK293T cells), decent brightness, appropriate affinity (a Kd of 2.17 μM) and fast response kinetics (an association and dissociation half-time of 0.20 and 0.087 s, respectively). Furthermore, the crystal structure of the cAMP-bound G-Flamp1 reveals one linker connecting the cAMP-binding domain to cpGFP adopts a distorted β-strand conformation that may serve as a fluorescence modulation switch. We demonstrate that G-Flamp1 enables sensitive monitoring of endogenous cAMP signals in brain regions that are implicated in learning and motor control in living organisms such as fruit flies and mice.
Collapse
Affiliation(s)
- Liang Wang
- Research Center for Biomedical Optics and Molecular Imaging, Shenzhen Key Laboratory for Molecular Imaging, Guangdong Provincial Key Laboratory of Biomedical Optical Imaging Technology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Chunling Wu
- PKU-IDG-McGovern Institute for Brain Research, Beijing, 100871, China
- State Key Laboratory of Brain and Cognitive Science, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Wanling Peng
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Ziliang Zhou
- Molecular Imaging Center, Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, 519000, China
- Department of Oral Emergency and General Dentistry, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou, 510182, Guangdong, China
| | - Jianzhi Zeng
- PKU-IDG-McGovern Institute for Brain Research, Beijing, 100871, China
| | - Xuelin Li
- PKU-IDG-McGovern Institute for Brain Research, Beijing, 100871, China
| | - Yini Yang
- PKU-IDG-McGovern Institute for Brain Research, Beijing, 100871, China
| | - Shuguang Yu
- State Key Laboratory of Neuroscience, Institute of Neuroscience, Shanghai Institutes for Biological Sciences, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Ye Zou
- Department of Biochemistry and Molecular Biophysics, Kansas State University, Manhattan, 66506, KS, USA
| | - Mian Huang
- Department of Biochemistry and Molecular Biophysics, Kansas State University, Manhattan, 66506, KS, USA
| | - Chang Liu
- Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Yefei Chen
- Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Yi Li
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Panpan Ti
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Wenfeng Liu
- Research Center for Biomedical Optics and Molecular Imaging, Shenzhen Key Laboratory for Molecular Imaging, Guangdong Provincial Key Laboratory of Biomedical Optical Imaging Technology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Yufeng Gao
- Research Center for Biomedical Optics and Molecular Imaging, Shenzhen Key Laboratory for Molecular Imaging, Guangdong Provincial Key Laboratory of Biomedical Optical Imaging Technology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Wei Zheng
- Research Center for Biomedical Optics and Molecular Imaging, Shenzhen Key Laboratory for Molecular Imaging, Guangdong Provincial Key Laboratory of Biomedical Optical Imaging Technology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Haining Zhong
- Vollum Institute, Oregon Health and Science University, Portland, 97239, OR, USA
| | - Shangbang Gao
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Zhonghua Lu
- Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Pei-Gen Ren
- Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Ho Leung Ng
- Department of Biochemistry and Molecular Biophysics, Kansas State University, Manhattan, 66506, KS, USA
| | - Jie He
- State Key Laboratory of Neuroscience, Institute of Neuroscience, Shanghai Institutes for Biological Sciences, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Shoudeng Chen
- Molecular Imaging Center, Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, 519000, China
- Department of Experimental Medicine, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, 519000, China
| | - Min Xu
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Yulong Li
- PKU-IDG-McGovern Institute for Brain Research, Beijing, 100871, China
| | - Jun Chu
- Research Center for Biomedical Optics and Molecular Imaging, Shenzhen Key Laboratory for Molecular Imaging, Guangdong Provincial Key Laboratory of Biomedical Optical Imaging Technology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China.
- Shenzhen-Hong Kong Institute of Brain Science, and Shenzhen Institute of Synthetic Biology, Shenzhen, 518055, China.
- CAS Key Laboratory of Health Informatics, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China.
| |
Collapse
|
34
|
Development of a red fluorescent protein-based cGMP indicator applicable for live-cell imaging. Commun Biol 2022; 5:833. [PMID: 36064581 PMCID: PMC9445041 DOI: 10.1038/s42003-022-03790-2] [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: 10/05/2021] [Accepted: 08/02/2022] [Indexed: 11/08/2022] Open
Abstract
Cyclic guanosine 3', 5'-monophosphate (cGMP) is a second messenger that regulates a variety of physiological processes. Here, we develop a red fluorescent protein-based cGMP indicator, "Red cGull". The fluorescence intensity of Red cGull increase more than sixfold in response to cGMP. The features of this indicator include an EC50 of 0.33 μM for cGMP, an excitation and emission peak at 567 nm and 591 nm, respectively. Live-cell imaging analysis reveal the utility of Red cGull for dual-colour imaging and its ability to be used in conjunction with optogenetics tools. Using enteroendocrine cell lines, Red cGull detects an increase in cGMP following the application of L-arginine. An increase in intracellular cGMP is found to be inhibited by Ca2+, and L-arginine-mediated hormone secretion is not potentiated. We propose that Red cGull will facilitate future research in cell signalling in relation to cGMP and its interplay with other signalling molecules.
Collapse
|
35
|
Uefune F, Aonishi T, Kitaguchi T, Takahashi H, Seino S, Sakano D, Kume S. Dopamine Negatively Regulates Insulin Secretion Through Activation of D1-D2 Receptor Heteromer. Diabetes 2022; 71:1946-1961. [PMID: 35728809 DOI: 10.2337/db21-0644] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Accepted: 05/09/2022] [Indexed: 11/13/2022]
Abstract
There is increasing evidence that dopamine (DA) functions as a negative regulator of glucose-stimulated insulin secretion; however, the underlying molecular mechanism remains unknown. Using total internal reflection fluorescence microscopy, we monitored insulin granule exocytosis in primary islet cells to dissect the effect of DA. We found that D1 receptor antagonists rescued the DA-mediated inhibition of glucose-stimulated calcium (Ca2+) flux, thereby suggesting a role of D1 in the DA-mediated inhibition of insulin secretion. Overexpression of D2, but not D1, alone exerted an inhibitory and toxic effect that abolished the glucose-stimulated Ca2+ influx and insulin secretion in β-cells. Proximity ligation and Western blot assays revealed that D1 and D2 form heteromers in β-cells. Treatment with a D1-D2 heteromer agonist, SKF83959, transiently inhibited glucose-induced Ca2+ influx and insulin granule exocytosis. Coexpression of D1 and D2 enabled β-cells to bypass the toxic effect of D2 overexpression. DA transiently inhibited glucose-stimulated Ca2+ flux and insulin exocytosis by activating the D1-D2 heteromer. We conclude that D1 protects β-cells from the harmful effects of DA by modulating D2 signaling. The finding will contribute to our understanding of the DA signaling in regulating insulin secretion and improve methods for preventing and treating diabetes.
Collapse
Affiliation(s)
- Fumiya Uefune
- School of Life Science and Technology, Tokyo Institute of Technology, Yokohama, Kanagawa, Japan
| | - Toru Aonishi
- School of Computing, Tokyo Institute of Technology, Yokohama, Kanagawa, Japan
| | - Tetsuya Kitaguchi
- Institute of Innovative Research, Tokyo Institute of Technology, Yokohama, Kanagawa, Japan
| | - Harumi Takahashi
- Molecular and Metabolic Medicine, Kobe University Graduate School of Medicine, Chuo-ku, Kobe, Japan
| | - Susumu Seino
- Molecular and Metabolic Medicine, Kobe University Graduate School of Medicine, Chuo-ku, Kobe, Japan
| | - Daisuke Sakano
- School of Life Science and Technology, Tokyo Institute of Technology, Yokohama, Kanagawa, Japan
| | - Shoen Kume
- School of Life Science and Technology, Tokyo Institute of Technology, Yokohama, Kanagawa, Japan
| |
Collapse
|
36
|
Sidoli M, Chen LC, Lu AJ, Wandless TJ, Talbot WS. A cAMP Sensor Based on Ligand-Dependent Protein Stabilization. ACS Chem Biol 2022; 17:2024-2030. [PMID: 35839076 PMCID: PMC9396618 DOI: 10.1021/acschembio.2c00333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Accepted: 07/12/2022] [Indexed: 11/28/2022]
Abstract
cAMP is a ubiquitous second messenger with many functions in diverse organisms. Current cAMP sensors, including Föster resonance energy transfer (FRET)-based and single-wavelength-based sensors, allow for real time visualization of this small molecule in cultured cells and in some cases in vivo. Nonetheless the observation of cAMP in living animals is still difficult, typically requiring specialized microscopes and ex vivo tissue processing. Here we used ligand-dependent protein stabilization to create a new cAMP sensor. This sensor allows specific and sensitive detection of cAMP in living zebrafish embryos, which may enable new understanding of the functions of cAMP in living vertebrates.
Collapse
Affiliation(s)
- Mariapaola Sidoli
- Department
of Developmental Biology, School of Medicine, Stanford University, Stanford, California 94305, United States
| | - Ling-chun Chen
- Department
of Chemical and Systems Biology, School of Medicine, Stanford University, Stanford, California 94305, United States
| | - Alexander J. Lu
- Department
of Chemical and Systems Biology, School of Medicine, Stanford University, Stanford, California 94305, United States
| | - Thomas J. Wandless
- Department
of Chemical and Systems Biology, School of Medicine, Stanford University, Stanford, California 94305, United States
| | - William S. Talbot
- Department
of Developmental Biology, School of Medicine, Stanford University, Stanford, California 94305, United States
| |
Collapse
|
37
|
Hansen JN, Kaiser F, Leyendecker P, Stüven B, Krause J, Derakhshandeh F, Irfan J, Sroka TJ, Preval KM, Desai PB, Kraut M, Theis H, Drews A, De‐Domenico E, Händler K, Pazour GJ, Henderson DJP, Mick DU, Wachten D. A cAMP signalosome in primary cilia drives gene expression and kidney cyst formation. EMBO Rep 2022; 23:e54315. [PMID: 35695071 PMCID: PMC9346484 DOI: 10.15252/embr.202154315] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 05/11/2022] [Accepted: 05/19/2022] [Indexed: 12/22/2022] Open
Abstract
The primary cilium constitutes an organelle that orchestrates signal transduction independently from the cell body. Dysregulation of this intricate molecular architecture leads to severe human diseases, commonly referred to as ciliopathies. However, the molecular underpinnings how ciliary signaling orchestrates a specific cellular output remain elusive. By combining spatially resolved optogenetics with RNA sequencing and imaging, we reveal a novel cAMP signalosome that is functionally distinct from the cytoplasm. We identify the genes and pathways targeted by the ciliary cAMP signalosome and shed light on the underlying mechanisms and downstream signaling. We reveal that chronic stimulation of the ciliary cAMP signalosome transforms kidney epithelia from tubules into cysts. Counteracting this chronic cAMP elevation in the cilium by small molecules targeting activation of phosphodiesterase-4 long isoforms inhibits cyst growth. Thereby, we identify a novel concept of how the primary cilium controls cellular functions and maintains tissue integrity in a specific and spatially distinct manner and reveal novel molecular components that might be involved in the development of one of the most common genetic diseases, polycystic kidney disease.
Collapse
Affiliation(s)
- Jan N Hansen
- Institute of Innate ImmunityMedical FacultyUniversity of BonnBonnGermany
| | - Fabian Kaiser
- Institute of Innate ImmunityMedical FacultyUniversity of BonnBonnGermany
| | | | - Birthe Stüven
- Institute of Innate ImmunityMedical FacultyUniversity of BonnBonnGermany
| | | | | | | | - Tommy J Sroka
- Center for Molecular Signaling (PZMS)Center of Human and Molecular Biology (ZHMB)Saarland University, School of MedicineHomburgGermany
| | - Kenley M Preval
- Program in Molecular MedicineUniversity of Massachusetts Chan Medical School, Biotech IIWorcesterMAUSA
| | - Paurav B Desai
- Program in Molecular MedicineUniversity of Massachusetts Chan Medical School, Biotech IIWorcesterMAUSA
| | - Michael Kraut
- Precise Platform for Single Cell Genomics and EpigenomicsDepartment of Systems MedicineGerman Center for Neurogenerative DiseasesBonnGermany
| | - Heidi Theis
- Precise Platform for Single Cell Genomics and EpigenomicsDepartment of Systems MedicineGerman Center for Neurogenerative DiseasesBonnGermany
| | - Anna‐Dorothee Drews
- Precise Platform for Single Cell Genomics and EpigenomicsDepartment of Systems MedicineGerman Center for Neurogenerative DiseasesBonnGermany
| | - Elena De‐Domenico
- Precise Platform for Single Cell Genomics and EpigenomicsDepartment of Systems MedicineGerman Center for Neurogenerative DiseasesBonnGermany
| | - Kristian Händler
- Precise Platform for Single Cell Genomics and EpigenomicsDepartment of Systems MedicineGerman Center for Neurogenerative DiseasesBonnGermany
| | - Gregory J Pazour
- Program in Molecular MedicineUniversity of Massachusetts Chan Medical School, Biotech IIWorcesterMAUSA
| | | | - David U Mick
- Center for Molecular Signaling (PZMS)Center of Human and Molecular Biology (ZHMB)Saarland University, School of MedicineHomburgGermany
| | - Dagmar Wachten
- Institute of Innate ImmunityMedical FacultyUniversity of BonnBonnGermany
| |
Collapse
|
38
|
Hirrlinger J, Nimmerjahn A. A perspective on astrocyte regulation of neural circuit function and animal behavior. Glia 2022; 70:1554-1580. [PMID: 35297525 PMCID: PMC9291267 DOI: 10.1002/glia.24168] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 01/19/2022] [Accepted: 02/27/2022] [Indexed: 12/16/2022]
Abstract
Studies over the past two decades have demonstrated that astrocytes are tightly associated with neurons and play pivotal roles in neural circuit development, operation, and adaptation in health and disease. Nevertheless, precisely how astrocytes integrate diverse neuronal signals, modulate neural circuit structure and function at multiple temporal and spatial scales, and influence animal behavior or disease through aberrant excitation and molecular output remains unclear. This Perspective discusses how new and state-of-the-art approaches, including fluorescence indicators, opto- and chemogenetic actuators, genetic targeting tools, quantitative behavioral assays, and computational methods, might help resolve these longstanding questions. It also addresses complicating factors in interpreting astrocytes' role in neural circuit regulation and animal behavior, such as their heterogeneity, metabolism, and inter-glial communication. Research on these questions should provide a deeper mechanistic understanding of astrocyte-neuron assemblies' role in neural circuit function, complex behaviors, and disease.
Collapse
Affiliation(s)
- Johannes Hirrlinger
- Carl-Ludwig-Institute for Physiology, Medical Faculty,
University of Leipzig, Leipzig, Germany
- Department of Neurogenetics, Max-Planck-Institute for
Multidisciplinary Sciences, Göttingen, Germany
| | - Axel Nimmerjahn
- Waitt Advanced Biophotonics Center, The Salk Institute for
Biological Studies, La Jolla, California
| |
Collapse
|
39
|
Kawata S, Mukai Y, Nishimura Y, Takahashi T, Saitoh N. Green fluorescent cAMP indicator of high speed and specificity suitable for neuronal live-cell imaging. Proc Natl Acad Sci U S A 2022; 119:e2122618119. [PMID: 35867738 PMCID: PMC9282276 DOI: 10.1073/pnas.2122618119] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 04/29/2022] [Indexed: 11/18/2022] Open
Abstract
Cyclic adenosine monophosphate (cAMP) is a canonical intracellular messenger playing diverse roles in cell functions. In neurons, cAMP promotes axonal growth during early development, and mediates sensory transduction and synaptic plasticity after maturation. The molecular cascades of cAMP are well documented, but its spatiotemporal profiles associated with neuronal functions remain hidden. Hence, we developed a genetically encoded cAMP indicator based on a bacterial cAMP-binding protein. This indicator "gCarvi" monitors [cAMP]i at 0.2 to 20 µM with a subsecond time resolution and a high specificity over cyclic guanosine monophosphate (cGMP). gCarvi can be converted to a ratiometric probe for [cAMP]i quantification and its expression can be specifically targeted to various subcellular compartments. Monomeric gCarvi also enables simultaneous multisignal monitoring in combination with other indicators. As a proof of concept, simultaneous cAMP/Ca2+ imaging in hippocampal neurons revealed a tight linkage of cAMP to Ca2+ signals. In cerebellar presynaptic boutons, forskolin induced nonuniform cAMP elevations among boutons, which positively correlated with subsequent increases in the size of the recycling pool of synaptic vesicles assayed using FM dye. Thus, the cAMP domain in presynaptic boutons is an important determinant of the synaptic strength.
Collapse
Affiliation(s)
- Seiko Kawata
- Department of Neurophysiology, Graduate School of Life and Medical Sciences, Doshisha University, Kyoto, 610-0394, Japan
| | - Yuki Mukai
- Department of Neurophysiology, Graduate School of Life and Medical Sciences, Doshisha University, Kyoto, 610-0394, Japan
| | - Yumi Nishimura
- Department of Neurophysiology, Graduate School of Life and Medical Sciences, Doshisha University, Kyoto, 610-0394, Japan
| | - Tomoyuki Takahashi
- Cellular and Molecular Synaptic Function Unit, Okinawa Institute of Science and Technology Graduate University, Okinawa, 904-0495, Japan
| | - Naoto Saitoh
- Department of Neurophysiology, Graduate School of Life and Medical Sciences, Doshisha University, Kyoto, 610-0394, Japan
| |
Collapse
|
40
|
Papaioannou S, Medini P. Advantages, Pitfalls, and Developments of All Optical Interrogation Strategies of Microcircuits in vivo. Front Neurosci 2022; 16:859803. [PMID: 35837124 PMCID: PMC9274136 DOI: 10.3389/fnins.2022.859803] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 05/30/2022] [Indexed: 12/03/2022] Open
Abstract
The holy grail for every neurophysiologist is to conclude a causal relationship between an elementary behaviour and the function of a specific brain area or circuit. Our effort to map elementary behaviours to specific brain loci and to further manipulate neural activity while observing the alterations in behaviour is in essence the goal for neuroscientists. Recent advancements in the area of experimental brain imaging in the form of longer wavelength near infrared (NIR) pulsed lasers with the development of highly efficient optogenetic actuators and reporters of neural activity, has endowed us with unprecedented resolution in spatiotemporal precision both in imaging neural activity as well as manipulating it with multiphoton microscopy. This readily available toolbox has introduced a so called all-optical physiology and interrogation of circuits and has opened new horizons when it comes to precisely, fast and non-invasively map and manipulate anatomically, molecularly or functionally identified mesoscopic brain circuits. The purpose of this review is to describe the advantages and possible pitfalls of all-optical approaches in system neuroscience, where by all-optical we mean use of multiphoton microscopy to image the functional response of neuron(s) in the network so to attain flexible choice of the cells to be also optogenetically photostimulated by holography, in absence of electrophysiology. Spatio-temporal constraints will be compared toward the classical reference of electrophysiology methods. When appropriate, in relation to current limitations of current optical approaches, we will make reference to latest works aimed to overcome these limitations, in order to highlight the most recent developments. We will also provide examples of types of experiments uniquely approachable all-optically. Finally, although mechanically non-invasive, all-optical electrophysiology exhibits potential off-target effects which can ambiguate and complicate the interpretation of the results. In summary, this review is an effort to exemplify how an all-optical experiment can be designed, conducted and interpreted from the point of view of the integrative neurophysiologist.
Collapse
|
41
|
Mitgau J, Franke J, Schinner C, Stephan G, Berndt S, Placantonakis DG, Kalwa H, Spindler V, Wilde C, Liebscher I. The N Terminus of Adhesion G Protein–Coupled Receptor GPR126/ADGRG6 as Allosteric Force Integrator. Front Cell Dev Biol 2022; 10:873278. [PMID: 35813217 PMCID: PMC9259995 DOI: 10.3389/fcell.2022.873278] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Accepted: 05/12/2022] [Indexed: 12/15/2022] Open
Abstract
The adhesion G protein–coupled receptor (aGPCR) GPR126/ADGRG6 plays an important role in several physiological functions, such as myelination or peripheral nerve repair. This renders the receptor an attractive pharmacological target. GPR126 is a mechano-sensor that translates the binding of extracellular matrix (ECM) molecules to its N terminus into a metabotropic intracellular signal. To date, the structural requirements and the character of the forces needed for this ECM-mediated receptor activation are largely unknown. In this study, we provide this information by combining classic second-messenger detection with single-cell atomic force microscopy. We established a monoclonal antibody targeting the N terminus to stimulate GPR126 and compared it to the activation through its known ECM ligands, collagen IV and laminin 211. As each ligand uses a distinct mode of action, the N terminus can be regarded as an allosteric module that can fine-tune receptor activation in a context-specific manner.
Collapse
Affiliation(s)
- Jakob Mitgau
- Rudolf Schönheimer Institute for Biochemistry, Molecular Biochemistry, University of Leipzig, Leipzig, Germany
| | - Julius Franke
- Rudolf Schönheimer Institute for Biochemistry, Molecular Biochemistry, University of Leipzig, Leipzig, Germany
| | - Camilla Schinner
- Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Gabriele Stephan
- Department of Neurosurgery, Kimmel Center for Stem Cell Biology, Laura and Isaac Perlmutter Cancer Center, NYU Grossman School of Medicine, New York, NY, United States
| | - Sandra Berndt
- Rudolf Schönheimer Institute for Biochemistry, Molecular Biochemistry, University of Leipzig, Leipzig, Germany
| | - Dimitris G. Placantonakis
- Department of Neurosurgery, Kimmel Center for Stem Cell Biology, Laura and Isaac Perlmutter Cancer Center, NYU Grossman School of Medicine, New York, NY, United States
| | - Hermann Kalwa
- Rudolf-Boehm-Institute for Pharmacology and Toxicology, University of Leipzig, Leipzig, Germany
| | - Volker Spindler
- Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Caroline Wilde
- Rudolf Schönheimer Institute for Biochemistry, Molecular Biochemistry, University of Leipzig, Leipzig, Germany
| | - Ines Liebscher
- Rudolf Schönheimer Institute for Biochemistry, Molecular Biochemistry, University of Leipzig, Leipzig, Germany
- *Correspondence: Ines Liebscher,
| |
Collapse
|
42
|
Liu W, Liu C, Ren PG, Chu J, Wang L. An Improved Genetically Encoded Fluorescent cAMP Indicator for Sensitive cAMP Imaging and Fast Drug Screening. Front Pharmacol 2022; 13:902290. [PMID: 35694242 PMCID: PMC9175130 DOI: 10.3389/fphar.2022.902290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Accepted: 04/18/2022] [Indexed: 11/24/2022] Open
Abstract
Cyclic adenosine 3',5'-monophosphate (cAMP) is an important intracellular second messenger molecule downstream of many G protein-coupled receptors (GPCRs). Fluorescence imaging with bright and sensitive cAMP indicators allows not only dissecting the spatiotemporal dynamics of intracellular cAMP, but also high-content screening of compounds against GPCRs. We previously reported the high-performance circularly permuted GFP (cpGFP)-based cAMP indicator G-Flamp1. Here, we developed improved G-Flamp1 variants G-Flamp2 and G-Flamp2b. Compared to G-Flamp1, G-Flamp2 exhibited increased baseline fluorescence (1.6-fold) and larger fluorescence change (ΔF/F0) (1,300% vs. 1,100%) in HEK293T cells, while G-Flamp2b showed increased baseline fluorescence (3.1-fold) and smaller ΔF/F0 (400% vs. 1,100%). Furthermore, live cell imaging of mitochondrial matrix-targeted G-Flamp2 confirmed cytosolic cAMP was able to enter the mitochondrial matrix. G-Flamp2 imaging also showed that adipose tissue extract activated the Gi protein-coupled orphan GPCR GPR50 in HEK293T cells. Taken together, our results showed that the high-performance of G-Flamp2 would facilitate sensitive intracellular cAMP imaging and activity measurement of compounds targeting GPCR-cAMP signaling pathway during early drug development.
Collapse
Affiliation(s)
- Wenfeng Liu
- Guangdong Provincial Key Laboratory of Biomedical Optical Imaging Technology & Center for Biomedical Optics and Molecular Imaging & CAS Key Laboratory of Health Informatics, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Chang Liu
- Center for Energy Metabolism and Reproduction, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- Shenzhen College of Advanced Technology, University of Chinese Academy of Sciences, Shenzhen, China
| | - Pei-Gen Ren
- Center for Energy Metabolism and Reproduction, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- Shenzhen College of Advanced Technology, University of Chinese Academy of Sciences, Shenzhen, China
| | - Jun Chu
- Guangdong Provincial Key Laboratory of Biomedical Optical Imaging Technology & Center for Biomedical Optics and Molecular Imaging & CAS Key Laboratory of Health Informatics, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen, China
| | - Liang Wang
- Guangdong Provincial Key Laboratory of Biomedical Optical Imaging Technology & Center for Biomedical Optics and Molecular Imaging & CAS Key Laboratory of Health Informatics, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| |
Collapse
|
43
|
Cherkashin AP, Rogachevskaja OA, Kabanova NV, Kotova PD, Bystrova MF, Kolesnikov SS. Taste Cells of the Type III Employ CASR to Maintain Steady Serotonin Exocytosis at Variable Ca 2+ in the Extracellular Medium. Cells 2022; 11:1369. [PMID: 35456048 PMCID: PMC9030112 DOI: 10.3390/cells11081369] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 04/07/2022] [Accepted: 04/11/2022] [Indexed: 12/17/2022] Open
Abstract
Type III taste cells are the only taste bud cells which express voltage-gated (VG) Ca2+ channels and employ Ca2+-dependent exocytosis to release neurotransmitters, particularly serotonin. The taste bud is a tightly packed cell population, wherein extracellular Ca2+ is expected to fluctuate markedly due to the electrical activity of taste cells. It is currently unclear whether the Ca2+ entry-driven synapse in type III cells could be reliable enough at unsteady extracellular Ca2. Here we assayed depolarization-induced Ca2+ signals and associated serotonin release in isolated type III cells at varied extracellular Ca2+. It turned out that the same depolarizing stimulus elicited invariant Ca2+ signals in type III cells irrespective of bath Ca2+ varied within 0.5-5 mM. The serotonin release from type III cells was assayed with the biosensor approach by using HEK-293 cells co-expressing the recombinant 5-HT4 receptor and genetically encoded cAMP sensor Pink Flamindo. Consistently with the weak Ca2+ dependence of intracellular Ca2+ transients produced by VG Ca2+ entry, depolarization-triggered serotonin secretion varied negligibly with bath Ca2+. The evidence implicated the extracellular Ca2+-sensing receptor in mediating the negative feedback mechanism that regulates VG Ca2+ entry and levels off serotonin release in type III cells at deviating Ca2+ in the extracellular medium.
Collapse
Affiliation(s)
| | | | | | | | | | - Stanislav S. Kolesnikov
- Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences, Institute of Cell Biophysics of the Russian Academy of Sciences, Pushchino 142290, Russia; (A.P.C.); (O.A.R.); (N.V.K.); (P.D.K.); (M.F.B.)
| |
Collapse
|
44
|
Shahoha M, Cohen R, Ben-Simon Y, Ashery U. cAMP-Dependent Synaptic Plasticity at the Hippocampal Mossy Fiber Terminal. Front Synaptic Neurosci 2022; 14:861215. [PMID: 35444523 PMCID: PMC9013808 DOI: 10.3389/fnsyn.2022.861215] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Accepted: 02/23/2022] [Indexed: 11/24/2022] Open
Abstract
Cyclic adenosine monophosphate (cAMP) is a crucial second messenger involved in both pre- and postsynaptic plasticity in many neuronal types across species. In the hippocampal mossy fiber (MF) synapse, cAMP mediates presynaptic long-term potentiation and depression. The main cAMP-dependent signaling pathway linked to MF synaptic plasticity acts via the activation of the protein kinase A (PKA) molecular cascade. Accordingly, various downstream putative synaptic PKA target proteins have been linked to cAMP-dependent MF synaptic plasticity, such as synapsin, rabphilin, synaptotagmin-12, RIM1a, tomosyn, and P/Q-type calcium channels. Regulating the expression of some of these proteins alters synaptic release probability and calcium channel clustering, resulting in short- and long-term changes to synaptic efficacy. However, despite decades of research, the exact molecular mechanisms by which cAMP and PKA exert their influences in MF terminals remain largely unknown. Here, we review current knowledge of different cAMP catalysts and potential downstream PKA-dependent molecular cascades, in addition to non-canonical cAMP-dependent but PKA-independent cascades, which might serve as alternative, compensatory or competing pathways to the canonical PKA cascade. Since several other central synapses share a similar form of presynaptic plasticity with the MF, a better description of the molecular mechanisms governing MF plasticity could be key to understanding the relationship between the transcriptional and computational levels across brain regions.
Collapse
Affiliation(s)
- Meishar Shahoha
- Faculty of Life Sciences, School of Neurobiology, Biochemistry and Biophysics, Tel Aviv University, Tel Aviv, Israel
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - Ronni Cohen
- Faculty of Life Sciences, School of Neurobiology, Biochemistry and Biophysics, Tel Aviv University, Tel Aviv, Israel
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - Yoav Ben-Simon
- Department of Neurophysiology, Vienna Medical University, Vienna, Austria
- *Correspondence: Yoav Ben-Simon,
| | - Uri Ashery
- Faculty of Life Sciences, School of Neurobiology, Biochemistry and Biophysics, Tel Aviv University, Tel Aviv, Israel
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
- Uri Ashery,
| |
Collapse
|
45
|
Miete C, Solis GP, Koval A, Brückner M, Katanaev VL, Behrens J, Bernkopf DB. Gαi2-induced conductin/axin2 condensates inhibit Wnt/β-catenin signaling and suppress cancer growth. Nat Commun 2022; 13:674. [PMID: 35115535 PMCID: PMC8814139 DOI: 10.1038/s41467-022-28286-9] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Accepted: 01/14/2022] [Indexed: 12/25/2022] Open
Abstract
Conductin/axin2 is a scaffold protein negatively regulating the pro-proliferative Wnt/β-catenin signaling pathway. Accumulation of scaffold proteins in condensates frequently increases their activity, but whether condensation contributes to Wnt pathway inhibition by conductin remains unclear. Here, we show that the Gαi2 subunit of trimeric G-proteins induces conductin condensation by targeting a polymerization-inhibiting aggregon in its RGS domain, thereby promoting conductin-mediated β-catenin degradation. Consistently, transient Gαi2 expression inhibited, whereas knockdown activated Wnt signaling via conductin. Colorectal cancers appear to evade Gαi2-induced Wnt pathway suppression by decreased Gαi2 expression and inactivating mutations, associated with shorter patient survival. Notably, the Gαi2-activating drug guanabenz inhibited Wnt signaling via conductin, consequently reducing colorectal cancer growth in vitro and in mouse models. In summary, we demonstrate Wnt pathway inhibition via Gαi2-triggered conductin condensation, suggesting a tumor suppressor function for Gαi2 in colorectal cancer, and pointing to the FDA-approved drug guanabenz for targeted cancer therapy.
Collapse
Affiliation(s)
- Cezanne Miete
- Experimental Medicine II, Nikolaus-Fiebiger-Center, Friedrich-Alexander University Erlangen-Nürnberg, 91054, Erlangen, Germany
| | - Gonzalo P Solis
- Department of Cell Physiology and Metabolism, Centre Médical Universitaire, University of Geneva, 1211, Geneva 4, Geneva, Switzerland
| | - Alexey Koval
- Department of Cell Physiology and Metabolism, Centre Médical Universitaire, University of Geneva, 1211, Geneva 4, Geneva, Switzerland
| | - Martina Brückner
- Experimental Medicine II, Nikolaus-Fiebiger-Center, Friedrich-Alexander University Erlangen-Nürnberg, 91054, Erlangen, Germany
| | - Vladimir L Katanaev
- Department of Cell Physiology and Metabolism, Centre Médical Universitaire, University of Geneva, 1211, Geneva 4, Geneva, Switzerland
- School of Biomedicine, Far Eastern Federal University, 690922, Vladivostok, Russia
| | - Jürgen Behrens
- Experimental Medicine II, Nikolaus-Fiebiger-Center, Friedrich-Alexander University Erlangen-Nürnberg, 91054, Erlangen, Germany
| | - Dominic B Bernkopf
- Experimental Medicine II, Nikolaus-Fiebiger-Center, Friedrich-Alexander University Erlangen-Nürnberg, 91054, Erlangen, Germany.
| |
Collapse
|
46
|
Ernstsen CV, Login FH, Schelde AB, Therkildsen J, Møller‐Jensen J, Nørregaard R, Prætorius H, Nejsum LN. Acute pyelonephritis: Increased plasma membrane targeting of renal aquaporin-2. Acta Physiol (Oxf) 2022; 234:e13760. [PMID: 34978750 DOI: 10.1111/apha.13760] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 11/22/2021] [Accepted: 01/01/2022] [Indexed: 12/16/2022]
Abstract
AIM Aquaporin-2 (AQP2) shuttling between intracellular vesicles and the apical plasma membrane is pivotal in arginine vasopressin-mediated urine concentration and is dysregulated in multiple diseases associated with water balance disorders. Children and adults with acute pyelonephritis have a urinary concentration defect and studies in children revealed increased AQP2 excretion in the urine. This study aimed to analyse AQP2 trafficking in response to acute pyelonephritis. METHODS Immunofluorescence analysis was used to evaluate subcellular localization of AQP2 and AQP2-S256A (mimicking non-phosphorylated AQP2 on serine 256) in cells stimulated with bacterial lysates and of AQP2 and pS256-AQP2 in a mouse model at day 5 of acute pyelonephritis. Western blotting was used to evaluate AQP2 levels and AQP2 phosphorylation on S256 upon incubation with bacterial lysates. Time-lapse imaging was used to measure intracellular cAMP levels in response to incubation with bacterial lysates. RESULTS In cell cultures, lysates from both uropathogenic and nonpathogenic bacteria-mediated AQP2 plasma membrane targeting and increased AQP2 phosphorylation on serine 256 (pS256) without increasing cAMP levels. Both bacterial lysates induced plasma membrane targeting of AQP2-S256A. Immunofluorescence analysis of renal sections from mice after 5 days of acute pyelonephritis revealed apical plasma membrane targeting of AQP2 and pS256-AQP2 in inner medullary collecting ducts. CONCLUSION Uropathogenic bacteria induce AQP2 plasma membrane targeting in vitro and in vivo. cAMP levels were not elevated by the bacterial lysates and AQP2 plasma membrane targeting could occur without S256 phosphorylation. This may explain increased AQP2 excretion in the urine during acute pyelonephritis.
Collapse
Affiliation(s)
- Christina V. Ernstsen
- Department of Clinical Medicine Aarhus University Aarhus Denmark
- Department of Molecular Biology and Genetics Aarhus University Aarhus Denmark
| | | | | | | | - Jakob Møller‐Jensen
- Department of Biochemistry and Molecular Biology University of Southern Denmark Odense Denmark
| | - Rikke Nørregaard
- Department of Clinical Medicine Aarhus University Aarhus Denmark
| | | | - Lene N. Nejsum
- Department of Clinical Medicine Aarhus University Aarhus Denmark
| |
Collapse
|
47
|
Aktar A, Wodz KM, Heit B. Monitoring Cellular Responses to Infection with Fluorescent Biosensors. Methods Mol Biol 2022; 2440:99-114. [PMID: 35218535 DOI: 10.1007/978-1-0716-2051-9_6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Fluorescent biosensors are chemically or genetically encoded reporters of cellular processes, signaling pathways, or biomolecule concentration, whose output is quantified using fluorescence microscopy or fluorescence spectrometry. These biosensors can detect the target activity or metabolites via mechanisms including conversion between nonfluorescent and fluorescent forms, changes in reporter intensity, changes in the intensity ratio across fluorescence channels, alterations to the subcellular localization of the bioreporter, and by fluorescence resonance energy transfer. Here, we describe the use of a chemical photoconverting biosensor, and genetically encoded localization and ratiometric biosensors, for monitoring the cellular and signaling processes involved in pathogen-induced apoptosis and the resulting destruction of the pathogen. While this study uses biosensors to monitor responses to infection, these approaches can be readily translated to other cellular systems and other fluorescent biosensors.
Collapse
Affiliation(s)
- Amena Aktar
- Department of Microbiology and Immunology, Center for Human Immunology, The University of Western Ontario, London, ON, Canada
| | - Kasia M Wodz
- Department of Microbiology and Immunology, Center for Human Immunology, The University of Western Ontario, London, ON, Canada
| | - Bryan Heit
- Department of Microbiology and Immunology, Center for Human Immunology, The University of Western Ontario, London, ON, Canada.
- Robarts Research Institute, London, ON, Canada.
| |
Collapse
|
48
|
Sanjel B, Kim BH, Song MH, Carstens E, Shim WS. Glucosylsphingosine evokes pruritus via activation of 5-HT 2A receptor and TRPV4 in sensory neurons. Br J Pharmacol 2021; 179:2193-2207. [PMID: 34766332 DOI: 10.1111/bph.15733] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2021] [Revised: 10/25/2021] [Accepted: 10/27/2021] [Indexed: 11/02/2022] Open
Abstract
BACKGROUND AND PURPOSE Glucosylsphingosine (GS), an endogenous sphingolipid, is highly accumulated in the epidermis of patients with atopic dermatitis (AD) due to abnormal ceramide metabolism. More importantly, GS can evoke scratching behaviors. However, the precise molecular mechanism by which GS induces pruritus has been elusive. Thus, the present study aimed to elucidate the molecular signaling pathway of GS, especially at the peripheral sensory neuronal levels. EXPERIMENTAL APPROACH Calcium imaging was used to investigate the responses of HEK293T cells or mouse dorsal root ganglion (DRG) neurons to application of GS. Scratching behavior tests were also performed with wild-type and Trpv4 knockout mice. KEY RESULTS GS activated DRG neurons in a manner involving both the 5-HT2A receptor and TRPV4. Furthermore, GS-induced responses were significantly suppressed by various inhibitors, including ketanserin (5-HT2A receptor antagonist), YM254890 (Gαq/11 inhibitor), gallein (Gβγ complex inhibitor), U73122 (phospholipase C inhibitor), bisindolylmaleimide I (PKC inhibitor), and HC067047 (TRPV4 antagonist). Moreover, DRG neurons from Trpv4 knockout mice exhibited significantly reduced responses to GS. Additionally, GS-evoked scratching behaviors were greatly decreased by pretreatment with inhibitors of either 5-HT2A receptor or TRPV4. As expected, GS-evoked scratching behavior was also significantly decreased in Trpv4 knockout mice. CONCLUSION AND IMPLICATIONS Overall, the present study provides evidence for a novel molecular signaling pathway for GS-evoked pruritus, which utilizes both 5-HT2A receptor and TRPV4 in mouse sensory neurons. Considering the high accumulation of GS in the epidermis of patients with AD, GS could be another pruritogen in patients with AD.
Collapse
Affiliation(s)
- Babina Sanjel
- College of Pharmacy, Gachon University, Incheon, Republic of Korea.,Gachon Institute of Pharmaceutical Sciences, Incheon, Republic of Korea
| | - Bo-Hyun Kim
- College of Pharmacy, Gachon University, Incheon, Republic of Korea.,Gachon Institute of Pharmaceutical Sciences, Incheon, Republic of Korea
| | - Myung-Hyun Song
- College of Pharmacy, Gachon University, Incheon, Republic of Korea
| | - Earl Carstens
- Department of Neurobiology, Physiology and Behavior, University of California, Davis, California, USA
| | - Won-Sik Shim
- College of Pharmacy, Gachon University, Incheon, Republic of Korea.,Gachon Institute of Pharmaceutical Sciences, Incheon, Republic of Korea
| |
Collapse
|
49
|
Labouesse MA, Patriarchi T. A versatile GPCR toolkit to track in vivo neuromodulation: not a one-size-fits-all sensor. Neuropsychopharmacology 2021; 46:2043-2047. [PMID: 33603136 PMCID: PMC8505436 DOI: 10.1038/s41386-021-00982-y] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 01/12/2021] [Accepted: 01/23/2021] [Indexed: 12/23/2022]
Affiliation(s)
- Marie A Labouesse
- Department of Psychiatry, College of Physicians and Surgeons, Columbia University, Columbia, NY, USA
- Division of Molecular Therapeutics, New York State Psychiatric Institute, New York, NY, USA
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
| | - Tommaso Patriarchi
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland.
- Neuroscience Center Zurich, University and ETH Zurich, Zurich, Switzerland.
| |
Collapse
|
50
|
Saydmohammed M, Jha A, Mahajan V, Gavlock D, Shun TY, DeBiasio R, Lefever D, Li X, Reese C, Kershaw EE, Yechoor V, Behari J, Soto-Gutierrez A, Vernetti L, Stern A, Gough A, Miedel MT, Lansing Taylor D. Quantifying the progression of non-alcoholic fatty liver disease in human biomimetic liver microphysiology systems with fluorescent protein biosensors. Exp Biol Med (Maywood) 2021; 246:2420-2441. [PMID: 33957803 PMCID: PMC8606957 DOI: 10.1177/15353702211009228] [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: 12/30/2020] [Accepted: 03/23/2021] [Indexed: 12/12/2022] Open
Abstract
Metabolic syndrome is a complex disease that involves multiple organ systems including a critical role for the liver. Non-alcoholic fatty liver disease (NAFLD) is a key component of the metabolic syndrome and fatty liver is linked to a range of metabolic dysfunctions that occur in approximately 25% of the population. A panel of experts recently agreed that the acronym, NAFLD, did not properly characterize this heterogeneous disease given the associated metabolic abnormalities such as type 2 diabetes mellitus (T2D), obesity, and hypertension. Therefore, metabolic dysfunction-associated fatty liver disease (MAFLD) has been proposed as the new term to cover the heterogeneity identified in the NAFLD patient population. Although many rodent models of NAFLD/NASH have been developed, they do not recapitulate the full disease spectrum in patients. Therefore, a platform has evolved initially focused on human biomimetic liver microphysiology systems that integrates fluorescent protein biosensors along with other key metrics, the microphysiology systems database, and quantitative systems pharmacology. Quantitative systems pharmacology is being applied to investigate the mechanisms of NAFLD/MAFLD progression to select molecular targets for fluorescent protein biosensors, to integrate computational and experimental methods to predict drugs for repurposing, and to facilitate novel drug development. Fluorescent protein biosensors are critical components of the platform since they enable monitoring of the pathophysiology of disease progression by defining and quantifying the temporal and spatial dynamics of protein functions in the biosensor cells, and serve as minimally invasive biomarkers of the physiological state of the microphysiology system experimental disease models. Here, we summarize the progress in developing human microphysiology system disease models of NAFLD/MAFLD from several laboratories, developing fluorescent protein biosensors to monitor and to measure NAFLD/MAFLD disease progression and implementation of quantitative systems pharmacology with the goal of repurposing drugs and guiding the creation of novel therapeutics.
Collapse
Affiliation(s)
- Manush Saydmohammed
- University of Pittsburgh Drug Discovery Institute, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Anupma Jha
- University of Pittsburgh Drug Discovery Institute, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Vineet Mahajan
- University of Pittsburgh Drug Discovery Institute, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Dillon Gavlock
- University of Pittsburgh Drug Discovery Institute, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Tong Ying Shun
- University of Pittsburgh Drug Discovery Institute, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Richard DeBiasio
- University of Pittsburgh Drug Discovery Institute, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Daniel Lefever
- University of Pittsburgh Drug Discovery Institute, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Xiang Li
- University of Pittsburgh Drug Discovery Institute, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Celeste Reese
- University of Pittsburgh Drug Discovery Institute, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Erin E Kershaw
- Department of Medicine, Division of Endocrinology and Metabolism, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Vijay Yechoor
- Department of Medicine, Division of Endocrinology and Metabolism, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Jaideep Behari
- Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, Pittsburgh, PA 15261, USA
- UPMC Liver Clinic, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Alejandro Soto-Gutierrez
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA 15261, USA
- Pittsburgh Liver Research Center, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Larry Vernetti
- University of Pittsburgh Drug Discovery Institute, University of Pittsburgh, Pittsburgh, PA 15261, USA
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Andrew Stern
- University of Pittsburgh Drug Discovery Institute, University of Pittsburgh, Pittsburgh, PA 15261, USA
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Albert Gough
- University of Pittsburgh Drug Discovery Institute, University of Pittsburgh, Pittsburgh, PA 15261, USA
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Mark T Miedel
- University of Pittsburgh Drug Discovery Institute, University of Pittsburgh, Pittsburgh, PA 15261, USA
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - D Lansing Taylor
- University of Pittsburgh Drug Discovery Institute, University of Pittsburgh, Pittsburgh, PA 15261, USA
- Pittsburgh Liver Research Center, University of Pittsburgh, Pittsburgh, PA 15261, USA
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, PA 15261, USA
| |
Collapse
|