1
|
Zanellati MC, Hsu CH, Cohen S. Imaging interorganelle contacts at a glance. J Cell Sci 2024; 137:jcs262020. [PMID: 39440475 PMCID: PMC11529887 DOI: 10.1242/jcs.262020] [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] [Indexed: 10/25/2024] Open
Abstract
Eukaryotic cells are compartmentalized into membrane-bound organelles that must coordinate their responses to stimuli. One way that organelles communicate is via membrane contact sites (MCSs), sites of close apposition between organelles used for the exchange of ions, lipids and information. In this Cell Science at a Glance article and the accompanying poster, we describe an explosion of new methods that have led to exciting progress in this area and discuss key examples of how these methods have advanced our understanding of MCSs. We discuss how diffraction-limited and super-resolution fluorescence imaging approaches have provided important insight into the biology of interorganelle communication. We also describe how the development of multiple proximity-based methods has enabled the detection of MCSs with high accuracy and precision. Finally, we assess how recent advances in electron microscopy (EM), considered the gold standard for detecting MCSs, have allowed the visualization of MCSs and associated proteins in 3D at ever greater resolution.
Collapse
Affiliation(s)
- Maria Clara Zanellati
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Chih-Hsuan Hsu
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Sarah Cohen
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| |
Collapse
|
2
|
Sanchez C, Ramirez A, Hodgson L. Unravelling molecular dynamics in living cells: Fluorescent protein biosensors for cell biology. J Microsc 2024. [PMID: 38357769 PMCID: PMC11324865 DOI: 10.1111/jmi.13270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 01/11/2024] [Accepted: 01/22/2024] [Indexed: 02/16/2024]
Abstract
Genetically encoded, fluorescent protein (FP)-based Förster resonance energy transfer (FRET) biosensors are microscopy imaging tools tailored for the precise monitoring and detection of molecular dynamics within subcellular microenvironments. They are characterised by their ability to provide an outstanding combination of spatial and temporal resolutions in live-cell microscopy. In this review, we begin by tracing back on the historical development of genetically encoded FP labelling for detection in live cells, which lead us to the development of early biosensors and finally to the engineering of single-chain FRET-based biosensors that have become the state-of-the-art today. Ultimately, this review delves into the fundamental principles of FRET and the design strategies underpinning FRET-based biosensors, discusses their diverse applications and addresses the distinct challenges associated with their implementation. We place particular emphasis on single-chain FRET biosensors for the Rho family of guanosine triphosphate hydrolases (GTPases), pointing to their historical role in driving our understanding of the molecular dynamics of this important class of signalling proteins and revealing the intricate relationships and regulatory mechanisms that comprise Rho GTPase biology in living cells.
Collapse
Affiliation(s)
- Colline Sanchez
- Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, New York, USA
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Andrea Ramirez
- Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, New York, USA
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Louis Hodgson
- Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, New York, USA
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, New York, USA
| |
Collapse
|
3
|
Chen Z, Chen W, Reheman Z, Jiang H, Wu J, Li X. Genetically encoded RNA-based sensors with Pepper fluorogenic aptamer. Nucleic Acids Res 2023; 51:8322-8336. [PMID: 37486780 PMCID: PMC10484673 DOI: 10.1093/nar/gkad620] [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] [Received: 01/24/2023] [Accepted: 07/21/2023] [Indexed: 07/26/2023] Open
Abstract
Sensors to measure the abundance and signaling of intracellular molecules are crucial for understanding their physiological functions. Although conventional fluorescent protein-based sensors have been designed, RNA-based sensors are promising imaging tools. Numerous RNA-based sensors have been developed. These sensors typically contain RNA G-quadruplex (RG4) motifs and thus may be suboptimal in living cells. Here we describe RNA-based sensors based on Pepper, a fluorogenic RNA without an RG4 motif. With Pepper, we engineered various sensors for metabolites, synthetic compounds, proteins and metal ions in vitro and in living cells. In addition, these sensors show high activation and selectivity, demonstrating their universality and robustness. In the case of sensors responding to S-adenosylmethionine (SAM), a metabolite produced by methionine adenosyltransferase (MATase), we showed that our sensors exhibited positively correlated fluorescence responding to different SAM levels. Importantly, we revealed the SAM biosynthesis pathway and monitored MATase activity and gene expression spatiotemporally in living individual human cells. Additionally, we constructed a ratiometric SAM sensor to determine the inhibition efficacy of a MATase inhibitor in living cells. Together, these sensors comprising Pepper provide a useful platform for imaging diverse cellular targets and their signaling pathway.
Collapse
Affiliation(s)
- Zhenyin Chen
- Beijing Institute of Life Sciences, Chinese Academy of Sciences, Beijing 100101, China
- Department of Pulmonary and Critical Care Medicine, Department of Inflammation and Clinical Allergology, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Wei Chen
- Beijing Institute of Life Sciences, Chinese Academy of Sciences, Beijing 100101, China
- Institute of Cytology and Genetics, the Hengyang Key Laboratory of Cellular Stress Biology, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Zhayila Reheman
- Beijing Institute of Life Sciences, Chinese Academy of Sciences, Beijing 100101, China
- School of Life Science, Hebei University, Baoding, Hebei 071000, China
| | - Haodong Jiang
- Beijing Institute of Life Sciences, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jiahui Wu
- Department of Chemistry, University of Massachusetts, Amherst, MA01003, USA
| | - Xing Li
- Beijing Institute of Life Sciences, Chinese Academy of Sciences, Beijing 100101, China
- Department of Pulmonary and Critical Care Medicine, Department of Inflammation and Clinical Allergology, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
- Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin 300308, China
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| |
Collapse
|
4
|
Miller MR, Lee YF, Kastanenka KV. Calcium sensor Yellow Cameleon 3.6 as a tool to support the calcium hypothesis of Alzheimer's disease. Alzheimers Dement 2023; 19:4196-4203. [PMID: 37154246 PMCID: PMC10524576 DOI: 10.1002/alz.13111] [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: 11/29/2022] [Revised: 03/30/2023] [Accepted: 04/03/2023] [Indexed: 05/10/2023]
Abstract
INTRODUCTION Alzheimer's disease (AD) is a neurodegenerative disease with increasing relevance as dementia cases rise. The etiology of AD is widely debated. The Calcium Hypothesis of Alzheimer's disease and brain aging states that the dysfunction of calcium signaling is the final common pathway leading to neurodegeneration. When the Calcium Hypothesis was originally coined, the technology did not exist to test it, but with the advent of Yellow Cameleon 3.6 (YC3.6) we are able to test its validity. METHODS Here we review use of YC3.6 in studying Alzheimer's disease using mouse models and discuss whether these studies support or refute the Calcium Hypothesis. RESULTS YC3.6 studies showed that amyloidosis preceded dysfunction in neuronal calcium signaling and changes in synapse structure. This evidence supports the Calcium Hypothesis. DISCUSSION In vivo YC3.6 studies point to calcium signaling as a promising therapeutic target; however, additional work is necessary to translate these findings to humans.
Collapse
Affiliation(s)
- Morgan R. Miller
- Department of Neurology, MassGeneral Institute of Neurodegenerative Diseases, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, 02129, USA
| | - Yee Fun Lee
- Department of Neurology, MassGeneral Institute of Neurodegenerative Diseases, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, 02129, USA
- Department of Anatomy and Neurobiology, Boston University School of Medicine, Boston, MA, 02118, USA
| | - Ksenia V. Kastanenka
- Department of Neurology, MassGeneral Institute of Neurodegenerative Diseases, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, 02129, USA
| |
Collapse
|
5
|
Chai F, Cheng D, Nasu Y, Terai T, Campbell RE. Maximizing the performance of protein-based fluorescent biosensors. Biochem Soc Trans 2023; 51:1585-1595. [PMID: 37431791 PMCID: PMC10586770 DOI: 10.1042/bst20221413] [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: 05/01/2023] [Revised: 06/22/2023] [Accepted: 06/26/2023] [Indexed: 07/12/2023]
Abstract
Fluorescent protein (FP)-based biosensors are genetically encoded tools that enable the imaging of biological processes in the context of cells, tissues, or live animals. Though widely used in biological research, practically all existing biosensors are far from ideal in terms of their performance, properties, and applicability for multiplexed imaging. These limitations have inspired researchers to explore an increasing number of innovative and creative ways to improve and maximize biosensor performance. Such strategies include new molecular biology methods to develop promising biosensor prototypes, high throughput microfluidics-based directed evolution screening strategies, and improved ways to perform multiplexed imaging. Yet another approach is to effectively replace components of biosensors with self-labeling proteins, such as HaloTag, that enable the biocompatible incorporation of synthetic fluorophores or other ligands in cells or tissues. This mini-review will summarize and highlight recent innovations and strategies for enhancing the performance of FP-based biosensors for multiplexed imaging to advance the frontiers of research.
Collapse
Affiliation(s)
- Fu Chai
- Department of Chemistry, Graduate School of Science, The University of Tokyo, Tokyo 113-0033, Japan
| | - Dazhou Cheng
- Department of Chemistry, Graduate School of Science, The University of Tokyo, Tokyo 113-0033, Japan
| | - Yusuke Nasu
- Department of Chemistry, Graduate School of Science, The University of Tokyo, Tokyo 113-0033, Japan
- PRESTO, Japan Science and Technology Agency, Chiyoda-ku, Tokyo 102-0075, Japan
| | - Takuya Terai
- Department of Chemistry, Graduate School of Science, The University of Tokyo, Tokyo 113-0033, Japan
| | - Robert E. Campbell
- Department of Chemistry, Graduate School of Science, The University of Tokyo, Tokyo 113-0033, Japan
- Department of Chemistry, University of Alberta, Edmonton, Alberta T6G 2G2, Canada
| |
Collapse
|
6
|
Ryan MB, Churchland AK, Gong Y, Baker C. Fastest-ever calcium sensors broaden the potential of neuronal imaging. Nature 2023; 615:804-805. [PMID: 36922656 DOI: 10.1038/d41586-023-00704-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/18/2023]
|
7
|
Bansal S, Lin S. Transcriptional Genetically Encoded Calcium Indicators in Drosophila. Cold Spring Harb Protoc 2023; 2023:8-18. [PMID: 36167674 DOI: 10.1101/pdb.top107797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Knowing which neurons are active during behavior is a crucial step toward understanding how nervous systems work. Neuronal activation is generally accompanied by an increase in intracellular calcium levels. Therefore, intracellular calcium levels are widely used as a proxy for neuronal activity. Many types of synthetic components and bioluminescent or fluorescent proteins that report transient and long-term changes in intracellular calcium levels have been developed over the past 60 years. Calcium indicators that enable imaging of the dynamic activity of a large ensemble of neurons in behaving animals have revolutionized the field of neuroscience. Among these, transcription-based genetically encoded calcium indicators (transcriptional GECIs) have proven easy to use and do not depend on sophisticated imaging systems, offering unique advantages over other types of calcium indicators. Here, we describe the two currently available fly transcriptional GECIs-calcium-dependent nuclear import of LexA (CaLexA) and transcriptional reporter of intracellular calcium (TRIC)-and review studies that have used them. In the accompanying protocol, we present step-by-step details for generating CaLexA- and TRIC-ready flies and for imaging CaLexA and TRIC signals in dissected brains after experimental manipulations of intact free-moving flies.
Collapse
Affiliation(s)
- Sonia Bansal
- Institute of Molecular Biology, Academia Sinica, Taipei 115, Taiwan
| | - Suewei Lin
- Institute of Molecular Biology, Academia Sinica, Taipei 115, Taiwan
| |
Collapse
|
8
|
Back V, Asgari A, Franczak A, Saito M, Castaneda Zaragoza D, Sandow SL, Plane F, Jurasz P. Inhibition of platelet aggregation by activation of platelet intermediate conductance Ca 2+ -activated potassium channels. J Thromb Haemost 2022; 20:2587-2600. [PMID: 35867883 DOI: 10.1111/jth.15827] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 07/05/2022] [Accepted: 07/11/2022] [Indexed: 11/28/2022]
Abstract
BACKGROUND Within the vasculature platelets and endothelial cells play crucial roles in hemostasis and thrombosis. Platelets, like endothelial cells, possess intermediate conductance Ca2+ -activated K+ (IKCa ) channels and generate nitric oxide (NO). Although NO limits platelet aggregation, the role of IKCa channels in platelet function and NO generation has not yet been explored. OBJECTIVES We investigated whether IKCa channel activation inhibits platelet aggregation, and per endothelial cells, enhances platelet NO production. METHODS Platelets were isolated from human volunteers. Aggregometry, confocal microscopy, and a novel flow chamber model, the Quartz Crystal Microbalance (QCM) were used to assess platelet function. Flow cytometry was used to measure platelet NO production, calcium signaling, membrane potential, integrin αIIb /β3 activation, granule release, and procoagulant platelet formation. RESULTS Platelet IKCa channel activation with SKA-31 inhibited aggregation in a concentration-dependent manner, an effect reversed by the selective IKCa channel blocker TRAM-34. The QCM model along with confocal microscopy demonstrated that SKA-31 inhibited platelet aggregation under flow conditions. Surprisingly, IKCa activation by SKA-31 inhibited platelet NO generation, but this could be explained by a concomitant reduction in platelet calcium signaling. IKCa activation by SKA-31 also inhibited dense and alpha-granule secretion and integrin αIIb /β3 activation, but maintained platelet phosphatidylserine surface exposure as a measure of procoagulant response. CONCLUSIONS Platelet IKCa channel activation inhibits aggregation by reducing calcium-signaling and granule secretion, but not by enhancing platelet NO generation. IKCa channels may be novel targets for the development of antiplatelet drugs that limit atherothrombosis, but not coagulation.
Collapse
Affiliation(s)
- Valentina Back
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada
| | - Amir Asgari
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada
| | - Aleksandra Franczak
- Department of Pharmacology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Max Saito
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada
| | - Diego Castaneda Zaragoza
- Department of Pharmacology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Shaun L Sandow
- Biomedical Sciences, University of the Sunshine Coast, Sydney, Queensland, Australia
- Department of Physiology, University of New South Wales, Sydney, Queensland, Australia
| | - Frances Plane
- Department of Pharmacology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
- Cardiovascular Research Centre, University of Alberta, Edmonton, Alberta, Canada
| | - Paul Jurasz
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada
- Department of Pharmacology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
- Cardiovascular Research Centre, University of Alberta, Edmonton, Alberta, Canada
- Mazankowski Alberta Heart Institute, Edmonton, Alberta, Canada
| |
Collapse
|
9
|
Fujii H, Bito H. Deciphering Ca2+-controlled biochemical computation governing neural circuit dynamics via multiplex imaging. Neurosci Res 2022; 179:79-90. [DOI: 10.1016/j.neures.2022.04.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 04/08/2022] [Accepted: 04/11/2022] [Indexed: 12/25/2022]
|
10
|
Rodrigo AP, Lopes A, Pereira R, Anjo SI, Manadas B, Grosso AR, Baptista PV, Fernandes AR, Costa PM. Endogenous Fluorescent Proteins in the Mucus of an Intertidal Polychaeta: Clues for Biotechnology. Mar Drugs 2022; 20:md20040224. [PMID: 35447897 PMCID: PMC9028460 DOI: 10.3390/md20040224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 03/23/2022] [Accepted: 03/23/2022] [Indexed: 02/04/2023] Open
Abstract
The vast ocean holds many unexplored organisms with unique adaptive features that enable them to thrive in their environment. The secretion of fluorescent proteins is one of them, with reports on the presence of such compounds in marine annelids being scarce. The intertidal Eulalia sp. is an example. The worm secretes copious amounts of mucus, that when purified and concentrated extracts, yield strong fluorescence under UV light. Emission has two main maxima, at 400 nm and at 500 nm, with the latter responsible for the blue–greenish fluorescence. Combining proteomics and transcriptomics techniques, we identified ubiquitin, peroxiredoxin, and 14-3-3 protein as key elements in the mucus. Fluorescence was found to be mainly modulated by redox status and pH, being consistently upheld in extracts prepared in Tris-HCl buffer with reducing agent at pH 7 and excited at 330 nm. One of the proteins associated with the fluorescent signal was localized in secretory cells in the pharynx. The results indicate that the secretion of fluorescent proteinaceous complexes can be an important defense against UV for this dweller. Additionally, the internalization of fluorescent complexes by ovarian cancer cells and modulation of fluorescence of redox status bears important considerations for biotechnological application of mucus components as markers.
Collapse
Affiliation(s)
- Ana P. Rodrigo
- UCIBIO—Applied Molecular Biosciences Unit, Department of Life Sciences, NOVA School of Science and Technology, Universidade NOVA de Lisboa, 2829-516 Caparica, Portugal; (A.L.); (R.P.); (A.R.G.); (P.V.B.); (A.R.F.)
- Associate Laboratory i4HB, Institute for Health and Bioeconomy, NOVA School of Science and Technology, Universidade NOVA de Lisboa, 2829-516 Caparica, Portugal
- Correspondence: (A.P.R.); (P.M.C.); Tel.: +351-212-948-300 (A.P.R. & P.M.C.)
| | - Ana Lopes
- UCIBIO—Applied Molecular Biosciences Unit, Department of Life Sciences, NOVA School of Science and Technology, Universidade NOVA de Lisboa, 2829-516 Caparica, Portugal; (A.L.); (R.P.); (A.R.G.); (P.V.B.); (A.R.F.)
| | - Ricardo Pereira
- UCIBIO—Applied Molecular Biosciences Unit, Department of Life Sciences, NOVA School of Science and Technology, Universidade NOVA de Lisboa, 2829-516 Caparica, Portugal; (A.L.); (R.P.); (A.R.G.); (P.V.B.); (A.R.F.)
| | - Sandra I. Anjo
- Center for Neuroscience and Cell Biology, University of Coimbra, Parque Tecnológico de Cantanhede, Núcleo 04, Lote 8, 3060-197 Cantanhede, Portugal; (S.I.A.); (B.M.)
| | - Bruno Manadas
- Center for Neuroscience and Cell Biology, University of Coimbra, Parque Tecnológico de Cantanhede, Núcleo 04, Lote 8, 3060-197 Cantanhede, Portugal; (S.I.A.); (B.M.)
| | - Ana R. Grosso
- UCIBIO—Applied Molecular Biosciences Unit, Department of Life Sciences, NOVA School of Science and Technology, Universidade NOVA de Lisboa, 2829-516 Caparica, Portugal; (A.L.); (R.P.); (A.R.G.); (P.V.B.); (A.R.F.)
- Associate Laboratory i4HB, Institute for Health and Bioeconomy, NOVA School of Science and Technology, Universidade NOVA de Lisboa, 2829-516 Caparica, Portugal
| | - Pedro V. Baptista
- UCIBIO—Applied Molecular Biosciences Unit, Department of Life Sciences, NOVA School of Science and Technology, Universidade NOVA de Lisboa, 2829-516 Caparica, Portugal; (A.L.); (R.P.); (A.R.G.); (P.V.B.); (A.R.F.)
- Associate Laboratory i4HB, Institute for Health and Bioeconomy, NOVA School of Science and Technology, Universidade NOVA de Lisboa, 2829-516 Caparica, Portugal
| | - Alexandra R. Fernandes
- UCIBIO—Applied Molecular Biosciences Unit, Department of Life Sciences, NOVA School of Science and Technology, Universidade NOVA de Lisboa, 2829-516 Caparica, Portugal; (A.L.); (R.P.); (A.R.G.); (P.V.B.); (A.R.F.)
- Associate Laboratory i4HB, Institute for Health and Bioeconomy, NOVA School of Science and Technology, Universidade NOVA de Lisboa, 2829-516 Caparica, Portugal
| | - Pedro M. Costa
- UCIBIO—Applied Molecular Biosciences Unit, Department of Life Sciences, NOVA School of Science and Technology, Universidade NOVA de Lisboa, 2829-516 Caparica, Portugal; (A.L.); (R.P.); (A.R.G.); (P.V.B.); (A.R.F.)
- Associate Laboratory i4HB, Institute for Health and Bioeconomy, NOVA School of Science and Technology, Universidade NOVA de Lisboa, 2829-516 Caparica, Portugal
- Correspondence: (A.P.R.); (P.M.C.); Tel.: +351-212-948-300 (A.P.R. & P.M.C.)
| |
Collapse
|
11
|
Serrat R, Oliveira-Pinto A, Marsicano G, Pouvreau S. Imaging mitochondrial calcium dynamics in the central nervous system. J Neurosci Methods 2022; 373:109560. [PMID: 35320763 DOI: 10.1016/j.jneumeth.2022.109560] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 03/04/2022] [Accepted: 03/06/2022] [Indexed: 12/28/2022]
Abstract
Mitochondrial calcium handling is a particularly active research area in the neuroscience field, as it plays key roles in the regulation of several functions of the central nervous system, such as synaptic transmission and plasticity, astrocyte calcium signaling, neuronal activity… In the last few decades, a panel of techniques have been developed to measure mitochondrial calcium dynamics, relying mostly on photonic microscopy, and including synthetic sensors, hybrid sensors and genetically encoded calcium sensors. The goal of this review is to endow the reader with a deep knowledge of the historical and latest tools to monitor mitochondrial calcium events in the brain, as well as a comprehensive overview of the current state of the art in brain mitochondrial calcium signaling. We will discuss the main calcium probes used in the field, their mitochondrial targeting strategies, their key properties and major drawbacks. In addition, we will detail the main roles of mitochondrial calcium handling in neuronal tissues through an extended report of the recent studies using mitochondrial targeted calcium sensors in neuronal and astroglial cells, in vitro and in vivo.
Collapse
Affiliation(s)
- Roman Serrat
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1215 NeuroCentre Magendie, France; University of Bordeaux, Bordeaux 33077, France
| | - Alexandre Oliveira-Pinto
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1215 NeuroCentre Magendie, France; University of Bordeaux, Bordeaux 33077, France
| | - Giovanni Marsicano
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1215 NeuroCentre Magendie, France; University of Bordeaux, Bordeaux 33077, France
| | - Sandrine Pouvreau
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1215 NeuroCentre Magendie, France; University of Bordeaux, Bordeaux 33077, France.
| |
Collapse
|
12
|
Kaster K, Patton J, Clayton S, Wauson E, Giles J, Tran QK. A novel assay to assess the effects of estrogen on the cardiac calmodulin binding equilibrium. Life Sci 2022; 290:120247. [PMID: 34954214 PMCID: PMC8779721 DOI: 10.1016/j.lfs.2021.120247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 12/08/2021] [Accepted: 12/14/2021] [Indexed: 02/03/2023]
Abstract
AIMS The Ca2+-binding protein calmodulin (CaM) modulates numerous target proteins but is produced insufficiently to bind all of them, generating a limiting CaM equilibrium. Menopause increases cardiac morbidity; however, it is unknown if the cardiac CaM equilibrium is affected by estrogen. We devised an assay to assess the effects of ovariectomy and estrogen treatment on the cardiac CaM equilibrium. MATERIALS AND METHODS Sprague-Dawley rats received sham surgery or ovariectomy, followed by 2-week treatment with vehicle or 17β-estradiol. Ca2+-saturated left ventricular (LV) lysates were processed through CaM sepharose columns, which retained CaM-binding proteins unoccupied by endogenous CaM. Eluants therefrom were subjected to a competitive binding assay against purified CaM and a CaM biosensor to assess the amounts of unoccupied CaM-binding sites. LV cellular composition was assessed by immunohistochemistry. KEY FINDINGS LV eluants processed from sham animals reduce biosensor response by ~32%, indicating baseline presence of unoccupied CaM-binding sites and a limiting CaM equilibrium. Ovariectomy exacerbates the limiting CaM equilibrium, reducing biosensor response by ~65%. 17β-estradiol treatment equalizes the difference between sham and ovariectomized animals. These changes reflect whole tissue responses and are not mirrored by changes in total surface areas of cardiomyocytes and fibroblasts. Consistently, Ca2+-dependent, but not Ca2+-independent, interaction between CaM and the cardiac inositol trisphosphate receptor (IP3R) is reduced following ovariectomy and is restored by subsequent 17β-estradiol treatment. SIGNIFICANCE Our assay provides a new parameter to assess tissue CaM equilibrium. The exacerbated limiting CaM equilibrium following estrogen loss may contribute to cardiac morbidity and is prevented by estrogen treatment.
Collapse
Affiliation(s)
- Kyle Kaster
- Department of Physiology & Pharmacology, Des Moines University Osteopathic Medical Center, IA 50312, United States
| | - John Patton
- Department of Physiology & Pharmacology, Des Moines University Osteopathic Medical Center, IA 50312, United States
| | - Sarah Clayton
- Department of Physiology & Pharmacology, Des Moines University Osteopathic Medical Center, IA 50312, United States
| | - Eric Wauson
- Department of Physiology & Pharmacology, Des Moines University Osteopathic Medical Center, IA 50312, United States
| | - Jennifer Giles
- Department of Physiology & Pharmacology, Des Moines University Osteopathic Medical Center, IA 50312, United States
| | - Quang-Kim Tran
- Department of Physiology & Pharmacology, Des Moines University Osteopathic Medical Center, IA 50312, United States.
| |
Collapse
|
13
|
Mahling R, Hovey L, Isbell HM, Marx DC, Miller MS, Kilpatrick AM, Weaver LD, Yoder JB, Kim EH, Andresen CNJ, Li S, Shea MA. Na V1.2 EFL domain allosterically enhances Ca 2+ binding to sites I and II of WT and pathogenic calmodulin mutants bound to the channel CTD. Structure 2021; 29:1339-1356.e7. [PMID: 33770503 PMCID: PMC8458505 DOI: 10.1016/j.str.2021.03.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Revised: 12/23/2020] [Accepted: 03/03/2021] [Indexed: 11/23/2022]
Abstract
Neuronal voltage-gated sodium channel NaV1.2 C-terminal domain (CTD) binds calmodulin (CaM) constitutively at its IQ motif. A solution structure (6BUT) and other NMR evidence showed that the CaM N domain (CaMN) is structurally independent of the C-domain (CaMC) whether CaM is bound to the NaV1.2IQp (1,901-1,927) or NaV1.2CTD (1,777-1,937) with or without calcium. However, in the CaM + NaV1.2CTD complex, the Ca2+ affinity of CaMN was more favorable than in free CaM, while Ca2+ affinity for CaMC was weaker than in the CaM + NaV1.2IQp complex. The CTD EF-like (EFL) domain allosterically widened the energetic gap between CaM domains. Cardiomyopathy-associated CaM mutants (N53I(N54I), D95V(D96V), A102V(A103V), E104A(E105A), D129G(D130G), and F141L(F142L)) all bound the NaV1.2 IQ motif favorably under resting (apo) conditions and bound calcium normally at CaMN sites. However, only N53I and A102V bound calcium at CaMC sites at [Ca2+] < 100 μM. Thus, they are expected to respond like wild-type CaM to Ca2+ spikes in excitable cells.
Collapse
Affiliation(s)
- Ryan Mahling
- Department of Biochemistry, Carver College of Medicine, University of Iowa, Iowa City, IA 52242-1109, USA
| | - Liam Hovey
- Department of Biochemistry, Carver College of Medicine, University of Iowa, Iowa City, IA 52242-1109, USA
| | - Holly M Isbell
- Department of Biochemistry, Carver College of Medicine, University of Iowa, Iowa City, IA 52242-1109, USA
| | - Dagan C Marx
- Department of Biochemistry, Carver College of Medicine, University of Iowa, Iowa City, IA 52242-1109, USA
| | - Mark S Miller
- Department of Biochemistry, Carver College of Medicine, University of Iowa, Iowa City, IA 52242-1109, USA
| | - Adina M Kilpatrick
- Department of Physics and Astronomy, Drake University, Des Moines, IA 50311-4516, USA
| | - Lisa D Weaver
- Department of Biochemistry, Carver College of Medicine, University of Iowa, Iowa City, IA 52242-1109, USA
| | - Jesse B Yoder
- Department of Biochemistry, Carver College of Medicine, University of Iowa, Iowa City, IA 52242-1109, USA
| | - Elaine H Kim
- Department of Biochemistry, Carver College of Medicine, University of Iowa, Iowa City, IA 52242-1109, USA
| | - Corinne N J Andresen
- Department of Biochemistry, Carver College of Medicine, University of Iowa, Iowa City, IA 52242-1109, USA
| | - Shuxiang Li
- Department of Biochemistry, Carver College of Medicine, University of Iowa, Iowa City, IA 52242-1109, USA
| | - Madeline A Shea
- Department of Biochemistry, Carver College of Medicine, University of Iowa, Iowa City, IA 52242-1109, USA.
| |
Collapse
|
14
|
Sadoine M, Ishikawa Y, Kleist TJ, Wudick MM, Nakamura M, Grossmann G, Frommer WB, Ho CH. Designs, applications, and limitations of genetically encoded fluorescent sensors to explore plant biology. PLANT PHYSIOLOGY 2021; 187:485-503. [PMID: 35237822 PMCID: PMC8491070 DOI: 10.1093/plphys/kiab353] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Accepted: 07/12/2021] [Indexed: 05/03/2023]
Abstract
The understanding of signaling and metabolic processes in multicellular organisms requires knowledge of the spatial dynamics of small molecules and the activities of enzymes, transporters, and other proteins in vivo, as well as biophysical parameters inside cells and across tissues. The cellular distribution of receptors, ligands, and activation state must be integrated with information about the cellular distribution of metabolites in relation to metabolic fluxes and signaling dynamics in order to achieve the promise of in vivo biochemistry. Genetically encoded sensors are engineered fluorescent proteins that have been developed for a wide range of small molecules, such as ions and metabolites, or to report biophysical processes, such as transmembrane voltage or tension. First steps have been taken to monitor the activity of transporters in vivo. Advancements in imaging technologies and specimen handling and stimulation have enabled researchers in plant sciences to implement sensor technologies in intact plants. Here, we provide a brief history of the development of genetically encoded sensors and an overview of the types of sensors available for quantifying and visualizing ion and metabolite distribution and dynamics. We further discuss the pros and cons of specific sensor designs, imaging systems, and sample manipulations, provide advice on the choice of technology, and give an outlook into future developments.
Collapse
Affiliation(s)
- Mayuri Sadoine
- Molecular Physiology, Heinrich-Heine-University Düsseldorf, Düsseldorf 40225, Germany
| | - Yuuma Ishikawa
- Molecular Physiology, Heinrich-Heine-University Düsseldorf, Düsseldorf 40225, Germany
- Institute of Transformative Bio-Molecules (WPI-ITbM), Nagoya University, Chikusa, Nagoya 464-8601, Japan
| | - Thomas J. Kleist
- Molecular Physiology, Heinrich-Heine-University Düsseldorf, Düsseldorf 40225, Germany
| | - Michael M. Wudick
- Molecular Physiology, Heinrich-Heine-University Düsseldorf, Düsseldorf 40225, Germany
- Cluster of Excellence on Plant Sciences, Heinrich-Heine-University Düsseldorf, Düsseldorf 40225, Germany
| | - Masayoshi Nakamura
- Institute of Transformative Bio-Molecules (WPI-ITbM), Nagoya University, Chikusa, Nagoya 464-8601, Japan
| | - Guido Grossmann
- Cluster of Excellence on Plant Sciences, Heinrich-Heine-University Düsseldorf, Düsseldorf 40225, Germany
- Institute for Cell and Interaction Biology, Heinrich-Heine-University Düsseldorf, Düsseldorf 40225, Germany
| | - Wolf B. Frommer
- Molecular Physiology, Heinrich-Heine-University Düsseldorf, Düsseldorf 40225, Germany
- Institute of Transformative Bio-Molecules (WPI-ITbM), Nagoya University, Chikusa, Nagoya 464-8601, Japan
- Cluster of Excellence on Plant Sciences, Heinrich-Heine-University Düsseldorf, Düsseldorf 40225, Germany
| | - Cheng-Hsun Ho
- Agricultural Biotechnology Research Center, Academia Sinica, Taipei 115, Taiwan
- Author for communication:
| |
Collapse
|
15
|
Sadoine M, Reger M, Wong KM, Frommer WB. Affinity Series of Genetically Encoded Förster Resonance Energy-Transfer Sensors for Sucrose. ACS Sens 2021; 6:1779-1784. [PMID: 33974799 DOI: 10.1021/acssensors.0c02495] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Genetically encoded fluorescent sugar sensors are valuable tools for the discovery of transporters and for quantitative monitoring of sugar steady-state levels in intact tissues. Genetically encoded Förster resonance energy-transfer sensors for glucose have been designed and optimized extensively, and a full series of affinity mutants is available for in vivo studies. However, to date, only a single improved sucrose sensor FLIPsuc-90μΔ1 with Km for sucrose of ∼90 μM was available. This sucrose sensor was engineered on the basis of an Agrobacterium tumefaciens sugar-binding protein. Here, we took a two-step approach to first improve the dynamic range of the FLIPsuc sensor and then expand the detection range from micro- to millimolar sucrose concentrations by mutating a key residue in the binding site. The resulting series of sucrose sensors may enable investigation of sucrose transporter candidates and comprehensive in vivo analyses of sucrose concentration in plants. Since FLIPsuc-90μ also detects trehalose in animal cells, the new series of sensors will likely be suitable for investigating trehalose transport and monitor trehalose steady-state levels in vivo.
Collapse
Affiliation(s)
- Mayuri Sadoine
- Institute for Molecular Physiology, Heinrich Heine University Düsseldorf, Düsseldorf 40225, Germany
| | - Mira Reger
- Department of Plant Biology, Carnegie Institution for Science, Stanford, California 94305, United States
| | - Ka Man Wong
- Department of Plant Biology, Carnegie Institution for Science, Stanford, California 94305, United States
| | - Wolf B. Frommer
- Institute for Molecular Physiology, Heinrich Heine University Düsseldorf, Düsseldorf 40225, Germany
- Department of Plant Biology, Carnegie Institution for Science, Stanford, California 94305, United States
- Institute for Transformative Biomolecules, ITbM, Nagoya University, Nagoya, Aichi 464-8601, Japan
| |
Collapse
|
16
|
Calvo-Rodriguez M, Kharitonova EK, Bacskai BJ. In vivo brain imaging of mitochondrial Ca 2+ in neurodegenerative diseases with multiphoton microscopy. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2021; 1868:118998. [PMID: 33684410 PMCID: PMC8057769 DOI: 10.1016/j.bbamcr.2021.118998] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Accepted: 02/22/2021] [Indexed: 10/22/2022]
Abstract
Mitochondria are involved in a large number of essential roles related to neuronal function. Ca2+ handling by mitochondria is critical for many of these functions, including energy production and cellular fate. Conversely, mitochondrial Ca2+ mishandling has been related to a variety of neurodegenerative diseases. Investigating mitochondrial Ca2+ dynamics is essential for advancing our understanding of the role of intracellular mitochondrial Ca2+ signals in physiology and pathology. Improved Ca2+ indicators, and the ability to target them to different cells and compartments, have emerged as useful tools for analysis of Ca2+ signals in living organisms. Combined with state-of-the-art techniques such as multiphoton microscopy, they allow for the study of mitochondrial Ca2+ dynamics in vivo in mouse models of the disease. Here, we provide an overview of the Ca2+ transporters/ion channels in mitochondrial membranes, and the involvement of mitochondrial Ca2+ in neurodegenerative diseases followed by a summary of the main tools available to evaluate mitochondrial Ca2+ dynamics in vivo using the aforementioned technique.
Collapse
Affiliation(s)
- Maria Calvo-Rodriguez
- Alzheimer Research Unit, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, 114, 16th St, Charlestown, MA, 02129, USA.
| | - Elizabeth K Kharitonova
- Alzheimer Research Unit, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, 114, 16th St, Charlestown, MA, 02129, USA
| | - Brian J Bacskai
- Alzheimer Research Unit, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, 114, 16th St, Charlestown, MA, 02129, USA
| |
Collapse
|
17
|
Ferkey DM, Sengupta P, L’Etoile ND. Chemosensory signal transduction in Caenorhabditis elegans. Genetics 2021; 217:iyab004. [PMID: 33693646 PMCID: PMC8045692 DOI: 10.1093/genetics/iyab004] [Citation(s) in RCA: 59] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Accepted: 01/05/2021] [Indexed: 12/16/2022] Open
Abstract
Chemosensory neurons translate perception of external chemical cues, including odorants, tastants, and pheromones, into information that drives attraction or avoidance motor programs. In the laboratory, robust behavioral assays, coupled with powerful genetic, molecular and optical tools, have made Caenorhabditis elegans an ideal experimental system in which to dissect the contributions of individual genes and neurons to ethologically relevant chemosensory behaviors. Here, we review current knowledge of the neurons, signal transduction molecules and regulatory mechanisms that underlie the response of C. elegans to chemicals, including pheromones. The majority of identified molecules and pathways share remarkable homology with sensory mechanisms in other organisms. With the development of new tools and technologies, we anticipate that continued study of chemosensory signal transduction and processing in C. elegans will yield additional new insights into the mechanisms by which this animal is able to detect and discriminate among thousands of chemical cues with a limited sensory neuron repertoire.
Collapse
Affiliation(s)
- Denise M Ferkey
- Department of Biological Sciences, University at Buffalo, The State University of New York, Buffalo, NY 14260, USA
| | - Piali Sengupta
- Department of Biology, Brandeis University, Waltham, MA 02454, USA
| | - Noelle D L’Etoile
- Department of Cell and Tissue Biology, University of California, San Francisco, CA 94143, USA
| |
Collapse
|
18
|
Heinrich R, Hussein W, Berlin S. Photo-transformable genetically-encoded optical probes for functional highlighting in vivo. J Neurosci Methods 2021; 355:109129. [PMID: 33711357 DOI: 10.1016/j.jneumeth.2021.109129] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2020] [Revised: 03/02/2021] [Accepted: 03/04/2021] [Indexed: 12/13/2022]
Abstract
Studying the brain requires knowledge about both structure (i.e., connectome) and function of its constituents (neurons and glia alike). This need has prompted the development of novel tools and techniques, in particular optical techniques to examine cells remotely. Early works (1900's) led to the development of novel cell-staining techniques that, when combined with the use of a very simple light microscope, visualized individual neurons and their subcellular compartments in fixed tissues. Today, highlighting of structure and function can be performed on live cells, notably in vivo, owing to discovery of GFP and subsequent development of genetically encoded fluorescent optical tools. In this review, we focus our attention on a subset of optical biosensors, namely probes whose emission can be modified by light. We designate them photo-transformable genetically encoded probes. The family of photo-transformable probes embraces current probes that undergo photoactivation (PA), photoconversion (PC) or photoswitching (PS). We argue that these are particularly suited for studying multiple features of neurons, such as structure, connectivity and function concomitantly, for functional highlighting of neurons in vivo.
Collapse
Affiliation(s)
- Ronit Heinrich
- Department of Neuroscience, The Ruth and Bruce Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, Israel
| | - Wessal Hussein
- Department of Neuroscience, The Ruth and Bruce Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, Israel
| | - Shai Berlin
- Department of Neuroscience, The Ruth and Bruce Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, Israel.
| |
Collapse
|
19
|
Ernst P, Chen K, Tang Y, Kim S, Guan J, He J, Xie M, Zhang JJ, Liu XM, Zhou L. Investigation into the difference in mitochondrial-cytosolic calcium coupling between adult cardiomyocyte and hiPSC-CM using a novel multifunctional genetic probe. Pflugers Arch 2021; 473:447-459. [PMID: 33587181 PMCID: PMC8100988 DOI: 10.1007/s00424-021-02524-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 01/24/2021] [Accepted: 01/26/2021] [Indexed: 10/22/2022]
Abstract
Ca2+ cycling plays a critical role in regulating cardiomyocyte (CM) function under both physiological and pathological conditions. Mitochondria have been implicated in Ca2+ handling in adult cardiomyocytes (ACMs). However, little is known about their role in the regulation of Ca2+ dynamics in human-induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs). In the present study, we developed a multifunctional genetically encoded Ca2+ probe capable of simultaneously measuring cytosolic and mitochondrial Ca2+ in real time. Using this novel probe, we determined and compared mitochondrial Ca2+ activity and the coupling with cytosolic Ca2+ dynamics in hiPSC-CMs and ACMs. Our data showed that while ACMs displayed a highly coordinated beat-by-beat response in mitochondrial Ca2+ in sync with cytosolic Ca2+, hiPSC-CMs showed high cell-wide variability in mitochondrial Ca2+ activity that is poorly coordinated with cytosolic Ca2+. We then revealed that mitochondrial-sarcoplasmic reticulum (SR) tethering, as well as the inter-mitochondrial network connection, is underdeveloped in hiPSC-CM compared to ACM, which may underlie the observed spatiotemporal decoupling between cytosolic and mitochondrial Ca2+ dynamics. Finally, we showed that knockdown of mitofusin-2 (Mfn2), a protein tethering mitochondria and SR, led to reduced cytosolic-mitochondrial Ca2+ coupling in ACMs, albeit to a lesser degree compared to hiPSC-CMs, suggesting that Mfn2 is a potential engineering target for improving mitochondrial-cytosolic Ca2+ coupling in hiPSC-CMs. Physiological relevance: The present study will advance our understanding of the role of mitochondria in Ca2+ handling and cycling in CMs, and guide the development of hiPSC-CMs for healing injured hearts.
Collapse
Affiliation(s)
- Patrick Ernst
- Department of Medicine, The University of Alabama at Birmingham, Birmingham, AL, 35294, USA
- Department of Biomedical Engineering, The University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Kai Chen
- Department of Medicine, The University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Yawen Tang
- Department of Biomedical Engineering, The University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Seulhee Kim
- Department of Medicine, The University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Jiashiung Guan
- Department of Medicine, The University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Jin He
- Department of Medicine, The University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Min Xie
- Department of Medicine, The University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Jianyi Jay Zhang
- Department of Biomedical Engineering, The University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Xiaoguang Margaret Liu
- Department of Biomedical Engineering, The University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Lufang Zhou
- Department of Medicine, The University of Alabama at Birmingham, Birmingham, AL, 35294, USA.
- Department of Biomedical Engineering, The University of Alabama at Birmingham, Birmingham, AL, 35294, USA.
| |
Collapse
|
20
|
Mahling R, Rahlf CR, Hansen SC, Hayden MR, Shea MA. Ca 2+-saturated calmodulin binds tightly to the N-terminal domain of A-type fibroblast growth factor homologous factors. J Biol Chem 2021; 296:100458. [PMID: 33639159 PMCID: PMC8059062 DOI: 10.1016/j.jbc.2021.100458] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 02/15/2021] [Accepted: 02/23/2021] [Indexed: 01/12/2023] Open
Abstract
Voltage-gated sodium channels (Navs) are tightly regulated by multiple conserved auxiliary proteins, including the four fibroblast growth factor homologous factors (FGFs), which bind the Nav EF-hand like domain (EFL), and calmodulin (CaM), a multifunctional messenger protein that binds the NaV IQ motif. The EFL domain and IQ motif are contiguous regions of NaV cytosolic C-terminal domains (CTD), placing CaM and FGF in close proximity. However, whether the FGFs and CaM act independently, directly associate, or operate through allosteric interactions to regulate channel function is unknown. Titrations monitored by steady-state fluorescence spectroscopy, structural studies with solution NMR, and computational modeling demonstrated for the first time that both domains of (Ca2+)4-CaM (but not apo CaM) directly bind two sites in the N-terminal domain (NTD) of A-type FGF splice variants (FGF11A, FGF12A, FGF13A, and FGF14A) with high affinity. The weaker of the (Ca2+)4-CaM-binding sites was known via electrophysiology to have a role in long-term inactivation of the channel but not known to bind CaM. FGF12A binding to a complex of CaM associated with a fragment of the NaV1.2 CTD increased the Ca2+-binding affinity of both CaM domains, consistent with (Ca2+)4-CaM interacting preferentially with its higher-affinity site in the FGF12A NTD. Thus, A-type FGFs can compete with NaV IQ motifs for (Ca2+)4-CaM. During spikes in the cytosolic Ca2+ concentration that accompany an action potential, CaM may translocate from the NaV IQ motif to the FGF NTD, or the A-type FGF NTD may recruit a second molecule of CaM to the channel.
Collapse
Affiliation(s)
- Ryan Mahling
- Department of Biochemistry, Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Cade R Rahlf
- Department of Biochemistry, Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Samuel C Hansen
- Department of Biochemistry, Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Matthew R Hayden
- Department of Biochemistry, Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Madeline A Shea
- Department of Biochemistry, Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA.
| |
Collapse
|
21
|
Filadi R, Greotti E. The yin and yang of mitochondrial Ca 2+ signaling in cell physiology and pathology. Cell Calcium 2020; 93:102321. [PMID: 33310302 DOI: 10.1016/j.ceca.2020.102321] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 11/16/2020] [Accepted: 11/16/2020] [Indexed: 12/26/2022]
Abstract
Mitochondria are autonomous and dynamic cellular organelles orchestrating a diverse range of cellular activities. Numerous cell-signaling pathways target these organelles and Ca2+ is one of the most significant. Mitochondria are able to rapidly and transiently take up Ca2+, thanks to the mitochondrial Ca2+ uniporter complex, as well as to extrude it through the Na+/Ca2+ and H+/Ca2+ exchangers. The transient accumulation of Ca2+ in the mitochondrial matrix impacts on mitochondrial functions and cell pathophysiology. Here we summarize the role of mitochondrial Ca2+ signaling in both physiological (yang) and pathological (yin) processes and the methods that can be used to investigate mitochondrial Ca2+ homeostasis. As an example of the pivotal role of mitochondria in pathology, we described the state of the art of mitochondrial Ca2+ alterations in different pathological conditions, with a special focus on Alzheimer's disease.
Collapse
Affiliation(s)
- Riccardo Filadi
- Neuroscience Institute, National Research Council (CNR), 35131 Padua, Italy; Department of Biomedical Sciences, University of Padua, 35131, Padua, Italy.
| | - Elisa Greotti
- Neuroscience Institute, National Research Council (CNR), 35131 Padua, Italy; Department of Biomedical Sciences, University of Padua, 35131, Padua, Italy.
| |
Collapse
|
22
|
Gammon ST, Liu TW, Piwnica-Worms D. Interrogating Cellular Communication in Cancer with Genetically Encoded Imaging Reporters. Radiol Imaging Cancer 2020; 2:e190053. [PMID: 32803164 PMCID: PMC7398120 DOI: 10.1148/rycan.2020190053] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Revised: 01/06/2020] [Accepted: 01/22/2020] [Indexed: 04/14/2023]
Abstract
Cells continuously communicate changes in their microenvironment, both locally and globally, with other cells in the organism. Integration of information arising from signaling networks impart continuous, time-dependent changes of cell function and phenotype. Use of genetically encoded reporters enable researchers to noninvasively monitor time-dependent changes in intercellular and intracellular signaling, which can be interrogated by macroscopic and microscopic optical imaging, nuclear medicine imaging, MRI, and even photoacoustic imaging techniques. Reporters enable noninvasive monitoring of changes in cell-to-cell proximity, transcription, translation, protein folding, protein association, protein degradation, drug action, and second messengers in real time. Because of their positive impact on preclinical research, attempts to improve the sensitivity and specificity of these reporters, and to develop new types and classes of reporters, remain an active area of investigation. A few reporters have migrated to proof-of-principle clinical demonstrations, and recent advances in genome editing technologies may enable the use of reporters in the context of genome-wide analysis and the imaging of complex genomic regulation in vivo that cannot be readily investigated through standard methodologies. The combination of genetically encoded imaging reporters with continuous improvements in other molecular biology techniques may enhance and expedite target discovery and drug development for cancer interventions and treatment. © RSNA, 2020.
Collapse
|
23
|
Chowdhury HH. Differences in cytosolic glucose dynamics in astrocytes and adipocytes measured by FRET-based nanosensors. Biophys Chem 2020; 261:106377. [PMID: 32302866 DOI: 10.1016/j.bpc.2020.106377] [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: 02/08/2020] [Revised: 04/08/2020] [Accepted: 04/08/2020] [Indexed: 11/17/2022]
Abstract
The cellular response to fluctuations in blood glucose levels consists of integrative regulation of cell glucose uptake and glucose utilization in the cytosol, resulting in altered levels of glucose in the cytosol. Cytosolic glucose is difficult to be measured in the intact tissue, however recently methods have become available that allow measurements of glucose in single living cells with fluorescence resonance energy transfer (FRET) based protein sensors. By studying the dynamics of cytosolic glucose levels in different experimental settings, we can gain insights into the properties of plasma membrane permeability to glucose and glucose utilization in the cytosol, and how these processes are modulated by different environmental conditions, agents and enzymes. In this review, we compare the cytosolic regulation of glucose in adipocytes and astrocytes - two important regulators of energy balance and glucose homeostasis in whole body and brain, respectively, with particular emphasis on the data obtained with FRET based protein sensors as well as other biochemical and molecular approaches.
Collapse
Affiliation(s)
- Helena H Chowdhury
- Laboratory of Neuroendocrinology - Molecular Cell Physiology, Institute of Pathophysiology, University of Ljubljana, Faculty of Medicine, 1000 Ljubljana, Slovenia; Laboratory of Cell Engineering, Celica Biomedical, 1000 Ljubljana, Slovenia.
| |
Collapse
|
24
|
Huang X, Jiang C, Yu L, Yang A. Current and Emerging Approaches for Studying Inter-Organelle Membrane Contact Sites. Front Cell Dev Biol 2020; 8:195. [PMID: 32292782 PMCID: PMC7118198 DOI: 10.3389/fcell.2020.00195] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Accepted: 03/09/2020] [Indexed: 12/24/2022] Open
Abstract
Inter-organelle membrane contact sites (MCSs) are classically defined as areas of close proximity between heterologous membranes and established by specific proteins (termed tethers). The interest on MCSs has rapidly increased in the last years, since MCSs play a crucial role in the transfer of cellular components between different organelles and have been involved in important cellular functions such as apoptosis, organelle division and biogenesis, and cell growth. Recently, an unprecedented depth and breadth in insights into the details of MCSs have been uncovered. On one hand, extensive MCSs (organelles interactome) are revealed by comprehensive analysis of organelle network with high temporal-spatial resolution at the system level. On the other hand, more and more tethers involving in MCSs are identified and further works are focusing on addressing the role of these tethers in regulating the function of MCSs at the molecular level. These enormous progresses largely depend on the powerful approaches, including several different types of microscopies and various biochemical techniques. These approaches have greatly accelerated recent advances in MCSs at the system and molecular level. In this review, we summarize the current and emerging approaches for studying MCSs, such as various microscopies, proximity-driven fluorescent signal generation and proximity-dependent biotinylation. In addition, we highlight the advantages and disadvantages of the techniques to provide a general guidance for the study of MCSs.
Collapse
Affiliation(s)
- Xue Huang
- School of Life Sciences, Chongqing University, Chongqing, China
| | - Chen Jiang
- School of Life Sciences, Chongqing University, Chongqing, China
| | - Lihua Yu
- School of Life Sciences, Chongqing University, Chongqing, China
| | - Aimin Yang
- School of Life Sciences, Chongqing University, Chongqing, China
| |
Collapse
|
25
|
Greotti E, Pozzan T. Live Mitochondrial or Cytosolic Calcium Imaging Using Genetically-encoded Cameleon Indicator in Mammalian Cells. Bio Protoc 2020; 10:e3504. [PMID: 33654731 DOI: 10.21769/bioprotoc.3504] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Revised: 12/16/2019] [Accepted: 12/17/2019] [Indexed: 02/04/2023] Open
Abstract
Calcium (Ca2+) imaging aims at investigating the dynamic changes in live cells of its concentration ([Ca2+]) in different pathophysiological conditions. Ca2+ is an ubiquitous and versatile intracellular signal that modulates a large variety of cellular functions thanks to a cell type-specific toolkit and a complex subcellular compartmentalization. Many Ca2+ sensors are presently available (chemical and genetically encoded) that can be specifically targeted to different cellular compartments. Using these probes, it is now possible to monitor Ca2+ dynamics of living cells not only in the cytosol but also within specific organelles. The choice of a specific sensor depends on the experimental design and the spatial and temporal resolution required. Here we describe the use of novel Förster resonance energy transfer (FRET)-based fluorescent Ca2+ probes to dynamically and quantitatively monitor the changes in cytosolic and mitochondrial [Ca2+] in a variety of cell types and experimental conditions. FRET-based sensors have the enormous advantage of being ratiometric, a feature that makes them particularly suitable for quantitative and in vivo applications.
Collapse
Affiliation(s)
- Elisa Greotti
- Neuroscience Institute, National Research Council (CNR), 35131 Padua, Italy.,Department of Biomedical Sciences, University of Padua, 35131 Padua, Italy
| | - Tullio Pozzan
- Neuroscience Institute, National Research Council (CNR), 35131 Padua, Italy.,Department of Biomedical Sciences, University of Padua, 35131 Padua, Italy.,Venetian Institute of Molecular Medicine (VIMM), 35129 Padua, Italy
| |
Collapse
|
26
|
Gökerküçük EB, Tramier M, Bertolin G. Imaging Mitochondrial Functions: from Fluorescent Dyes to Genetically-Encoded Sensors. Genes (Basel) 2020; 11:E125. [PMID: 31979408 PMCID: PMC7073610 DOI: 10.3390/genes11020125] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2019] [Revised: 01/20/2020] [Accepted: 01/21/2020] [Indexed: 12/18/2022] Open
Abstract
Mitochondria are multifunctional organelles that are crucial to cell homeostasis. They constitute the major site of energy production for the cell, they are key players in signalling pathways using secondary messengers such as calcium, and they are involved in cell death and redox balance paradigms. Mitochondria quickly adapt their dynamics and biogenesis rates to meet the varying energy demands of the cells, both in normal and in pathological conditions. Therefore, understanding simultaneous changes in mitochondrial functions is crucial in developing mitochondria-based therapy options for complex pathological conditions such as cancer, neurological disorders, and metabolic syndromes. To this end, fluorescence microscopy coupled to live imaging represents a promising strategy to track these changes in real time. In this review, we will first describe the commonly available tools to follow three key mitochondrial functions using fluorescence microscopy: Calcium signalling, mitochondrial dynamics, and mitophagy. Then, we will focus on how the development of genetically-encoded fluorescent sensors became a milestone for the understanding of these mitochondrial functions. In particular, we will show how these tools allowed researchers to address several biochemical activities in living cells, and with high spatiotemporal resolution. With the ultimate goal of tracking multiple mitochondrial functions simultaneously, we will conclude by presenting future perspectives for the development of novel genetically-encoded fluorescent biosensors.
Collapse
Affiliation(s)
| | | | - Giulia Bertolin
- Univ Rennes, CNRS, IGDR [Institut de génétique et développement de Rennes] UMR 6290, F-35000 Rennes, France
| |
Collapse
|
27
|
|
28
|
Fluorescent Protein-Based Indicators for Functional Super-Resolution Imaging of Biomolecular Activities in Living Cells. Int J Mol Sci 2019; 20:ijms20225784. [PMID: 31744242 PMCID: PMC6887983 DOI: 10.3390/ijms20225784] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Revised: 11/15/2019] [Accepted: 11/15/2019] [Indexed: 11/16/2022] Open
Abstract
Super-resolution light microscopy (SRM) offers a unique opportunity for diffraction-unlimited imaging of biomolecular activities in living cells. To realize such potential, genetically encoded indicators were developed recently from fluorescent proteins (FPs) that exhibit phototransformation behaviors including photoactivation, photoconversion, and photoswitching, etc. Super-resolution observations of biomolecule interactions and biochemical activities have been demonstrated by exploiting the principles of bimolecular fluorescence complementation (BiFC), points accumulation for imaging nanoscale topography (PAINT), and fluorescence fluctuation increase by contact (FLINC), etc. To improve functional nanoscopy with the technology of genetically encoded indicators, it is essential to fully decipher the photo-induced chemistry of FPs and opt for innovative indicator designs that utilize not only fluorescence intensity but also multi-parametric readouts such as phototransformation kinetics. In parallel, technical improvements to both the microscopy optics and image analysis pipeline are promising avenues to increase the sensitivity and versatility of functional SRM.
Collapse
|
29
|
Roopa, Kumar N, Kumar M, Bhalla V. Design and Applications of Small Molecular Probes for Calcium Detection. Chem Asian J 2019; 14:4493-4505. [PMID: 31549484 DOI: 10.1002/asia.201901149] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2019] [Indexed: 12/16/2022]
Abstract
The physiological significance of calcium ions such as the role in cellular signalling, cell growth, etc. have driven the development of methods to detect and monitor the level of Ca2+ ions, both in vivo and in vitro. Although various approaches for the detection of calcium ions have been reported, methods based on small molecular fluorescent probes have unique advantages including small probe size, easy monitoring of detection processes and applicability in biological systems. In this review article, we will discuss the progress in the development of Ca2+ -binding fluorescent probes by taking into account the types of chelating groups that have been employed for Ca2+ binding.
Collapse
Affiliation(s)
- Roopa
- Department of Chemical Sciences, IKG-Punjab Technical University, Kapurthala, 144603, Punjab, India
| | - Naresh Kumar
- Department of Chemistry, Kanya Maha Vidyalaya, Jalandhar, 144004, India
| | - Manoj Kumar
- Department of Chemistry, UGC Sponsored Centre for Advanced Studies-1, Guru Nanak Dev University, Amritsar-, 143005, Punjab, India
| | - Vandana Bhalla
- Department of Chemistry, UGC Sponsored Centre for Advanced Studies-1, Guru Nanak Dev University, Amritsar-, 143005, Punjab, India
| |
Collapse
|
30
|
Wu N, Nishioka WK, Derecki NC, Maher MP. High-throughput-compatible assays using a genetically-encoded calcium indicator. Sci Rep 2019; 9:12692. [PMID: 31481721 PMCID: PMC6722131 DOI: 10.1038/s41598-019-49070-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Accepted: 08/19/2019] [Indexed: 12/11/2022] Open
Abstract
Measurement of intracellular calcium in live cells is a key component of a wide range of basic life science research, and crucial for many high-throughput assays used in modern drug discovery. Synthetic calcium indicators have become the industry standard, due their ease of use, high reliability, wide dynamic range, and availability of a large variety of spectral and chemical properties. Genetically-encoded calcium indicators (GECIs) have been optimized to the point where their performance rivals that of synthetic calcium indicators in many applications. Stable expression of a GECI has distinct advantages over synthetic calcium indicators in terms of reagent cost and simplification of the assay process. We generated a clonal cell line constitutively expressing GCaMP6s; high expression of the GECI was driven by coupling to a blasticidin resistance gene with a self-cleaving cis-acting hydrolase element (CHYSEL) 2A peptide. Here, we compared the performance of the GECI GCaMP6s to the synthetic calcium indicator fluo-4 in a variety of assay formats. We demonstrate that the pharmacology of ion channel and GPCR ligands as determined using the two indicators is highly similar, and that GCaMP6s is viable as a direct replacement for a synthetic calcium indicator.
Collapse
Affiliation(s)
- Nyantsz Wu
- Janssen Research & Development, LLC, San Diego, CA, 92121, USA
| | | | - Noël C Derecki
- Janssen Research & Development, LLC, San Diego, CA, 92121, USA
| | - Michael P Maher
- Janssen Research & Development, LLC, San Diego, CA, 92121, USA.
| |
Collapse
|
31
|
Fernandez-Sanz C, De la Fuente S, Sheu SS. Mitochondrial Ca 2+ concentrations in live cells: quantification methods and discrepancies. FEBS Lett 2019; 593:1528-1541. [PMID: 31058316 DOI: 10.1002/1873-3468.13427] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Revised: 04/29/2019] [Accepted: 05/02/2019] [Indexed: 12/11/2022]
Abstract
Intracellular Ca2+ signaling controls numerous cellular functions. Mitochondria respond to cytosolic Ca2+ changes by adapting mitochondrial functions and, in some cell types, shaping the spatiotemporal properties of the cytosolic Ca2+ signal. Numerous methods have been developed to specifically and quantitatively measure the mitochondrial-free Ca2+ concentrations ([Ca2+ ]m ), but there are still significant discrepancies in the calculated absolute values of [Ca2+ ]m in stimulated live cells. These discrepancies may be due to the distinct properties of the methods used to measure [Ca2+ ]m , the calcium-free/bound ratio, and the cell-type and stimulus-dependent Ca2+ dynamics. Critical processes happening in the mitochondria, such as ATP generation, ROS homeostasis, and mitochondrial permeability transition opening, depend directly on the [Ca2+ ]m values. Thus, precise determination of absolute [Ca2+ ]m values is imperative for understanding Ca2+ signaling. This review summarizes the reported calibrated [Ca2+ ]m values in many cell types and discusses the discrepancies among these values. Areas for future research are also proposed.
Collapse
Affiliation(s)
- Celia Fernandez-Sanz
- Center for Translational Medicine, Department of Medicine, Thomas Jefferson University, Philadelphia, PA, USA
| | - Sergio De la Fuente
- Center for Translational Medicine, Department of Medicine, Thomas Jefferson University, Philadelphia, PA, USA
| | - Shey-Shing Sheu
- Center for Translational Medicine, Department of Medicine, Thomas Jefferson University, Philadelphia, PA, USA
| |
Collapse
|
32
|
Reuschlein AK, Jakobsen E, Mertz C, Bak LK. Aspects of astrocytic cAMP signaling with an emphasis on the putative power of compartmentalized signals in health and disease. Glia 2019; 67:1625-1636. [PMID: 31033018 DOI: 10.1002/glia.23622] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Revised: 03/29/2019] [Accepted: 03/29/2019] [Indexed: 12/17/2022]
Abstract
This review discusses aspects of known and putative compartmentalized 3',5'-cyclic adenosine monophosphate (cAMP) signaling in astrocytes, a cell type that has turned out to be a key player in brain physiology and pathology. cAMP has attracted less attention than Ca2+ in recent years, but could turn out to rival Ca2+ in its potential to drive cellular functions and responses to intra- and extracellular cues. Further, Ca2+ and cAMP are known to engage in extensive crosstalk and cAMP signals often take place within subcellular compartments revolving around multi-protein signaling complexes; however, we know surprisingly little about this in astrocytes. Here, we review aspects of astrocytic cAMP signaling, provide arguments for an increased interest in this subject, suggest possible future research directions within the field, and discuss putative drug targets.
Collapse
Affiliation(s)
- Ann-Kathrin Reuschlein
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Emil Jakobsen
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Christoffer Mertz
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Lasse K Bak
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
33
|
Sherpa RT, Pala R, Mohieldin AM, Nauli SM. Measurement of cytoplasmic and cilioplasmic calcium in a single living cell. Methods Cell Biol 2019; 153:25-42. [PMID: 31395382 DOI: 10.1016/bs.mcb.2019.03.015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Cellular signaling represents an evolution of biological systems to sense external stimuli and communicate extracellular microenvironment to the intracellular compartments. The processes underlying molecular signaling have been widely studied due to their important cellular functions. There are numerous techniques available to quantitate the different molecules involved in cellular processes. Among them, calcium is a ubiquitous signaling molecule involved in many biological pathways. Over time the methods to measure intracellular calcium have advanced to better understand its role as a second messenger. In this chapter, we introduce a method to study a single cilium, a mechanosensor that elicits a calcium signaling cascade. To successfully observe the calcium changes in this thin cylindrical-like projection from the cell surface, we utilize a genetically encoded sensor with a high spatial and temporal resolution. In addition, the probe must be localized to the ciliary compartment in order to observe the intraciliary calcium signaling dynamics. To this end, a cilium targeting genetically encoded indicator is used to observe calcium fluxes in both cytoplasm and cilioplasm.
Collapse
Affiliation(s)
- Rinzhin T Sherpa
- Department of Biomedical & Pharmaceutical Sciences, Chapman University School of Pharmacy (CUSP), Chapman University, Irvine, CA, United States
| | - Rajasekharreddy Pala
- Department of Biomedical & Pharmaceutical Sciences, Chapman University School of Pharmacy (CUSP), Chapman University, Irvine, CA, United States
| | - Ashraf M Mohieldin
- Department of Biomedical & Pharmaceutical Sciences, Chapman University School of Pharmacy (CUSP), Chapman University, Irvine, CA, United States
| | - Surya M Nauli
- Department of Biomedical & Pharmaceutical Sciences, Chapman University School of Pharmacy (CUSP), Chapman University, Irvine, CA, United States; Department of Medicine, University of California Irvine, Irvine, CA, United States.
| |
Collapse
|
34
|
Poli D, Magliaro C, Ahluwalia A. Experimental and Computational Methods for the Study of Cerebral Organoids: A Review. Front Neurosci 2019; 13:162. [PMID: 30890910 PMCID: PMC6411764 DOI: 10.3389/fnins.2019.00162] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Accepted: 02/12/2019] [Indexed: 01/04/2023] Open
Abstract
Cerebral (or brain) organoids derived from human cells have enormous potential as physiologically relevant downscaled in vitro models of the human brain. In fact, these stem cell-derived neural aggregates resemble the three-dimensional (3D) cytoarchitectural arrangement of the brain overcoming not only the unrealistic somatic flatness but also the planar neuritic outgrowth of the two-dimensional (2D) in vitro cultures. Despite the growing use of cerebral organoids in scientific research, a more critical evaluation of their reliability and reproducibility in terms of cellular diversity, mature traits, and neuronal dynamics is still required. Specifically, a quantitative framework for generating and investigating these in vitro models of the human brain is lacking. To this end, the aim of this review is to inspire new computational and technology driven ideas for methodological improvements and novel applications of brain organoids. After an overview of the organoid generation protocols described in the literature, we review the computational models employed to assess their formation, organization and resource uptake. The experimental approaches currently provided to structurally and functionally characterize brain organoid networks for studying single neuron morphology and their connections at cellular and sub-cellular resolution are also discussed. Well-established techniques based on current/voltage clamp, optogenetics, calcium imaging, and Micro-Electrode Arrays (MEAs) are proposed for monitoring intra- and extra-cellular responses underlying neuronal dynamics and functional connections. Finally, we consider critical aspects of the established procedures and the physiological limitations of these models, suggesting how a complement of engineering tools could improve the current approaches and their applications.
Collapse
Affiliation(s)
- Daniele Poli
- Research Center E. Piaggio, University of Pisa, Pisa, Italy
| | | | - Arti Ahluwalia
- Research Center E. Piaggio, University of Pisa, Pisa, Italy
- Department of Information Engineering, University of Pisa, Pisa, Italy
| |
Collapse
|
35
|
Kannan M, Vasan G, Pieribone VA. Optimizing Strategies for Developing Genetically Encoded Voltage Indicators. Front Cell Neurosci 2019; 13:53. [PMID: 30863283 PMCID: PMC6399427 DOI: 10.3389/fncel.2019.00053] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Accepted: 02/04/2019] [Indexed: 01/23/2023] Open
Abstract
Genetically encoded optical indicators of neuronal activity enable unambiguous recordings of input-output activity patterns from identified cells in intact circuits. Among them, genetically encoded voltage indicators (GEVIs) offer additional advantages over calcium indicators as they are direct sensors of membrane potential and can adeptly report subthreshold events and hyperpolarization. Here, we outline the major GEVI designs and give an account of properties that need to be carefully optimized during indicator engineering. While designing the ideal GEVI, one should keep in mind aspects such as membrane localization, signal size, signal-to-noise ratio, kinetics and voltage dependence of optical responses. Using ArcLight and derivatives as prototypes, we delineate how a probe should be optimized for the former properties and developed along other areas in a need-based manner. Finally, we present an overview of the GEVI engineering process and lend an insight into their discovery, delivery and diagnosis.
Collapse
Affiliation(s)
- Madhuvanthi Kannan
- The John B. Pierce Laboratory, New Haven, CT, United States.,Department of Cellular and Molecular Physiology, Yale School of Medicine, Yale University, New Haven, CT, United States
| | - Ganesh Vasan
- The John B. Pierce Laboratory, New Haven, CT, United States.,Department of Cellular and Molecular Physiology, Yale School of Medicine, Yale University, New Haven, CT, United States
| | - Vincent A Pieribone
- The John B. Pierce Laboratory, New Haven, CT, United States.,Department of Cellular and Molecular Physiology, Yale School of Medicine, Yale University, New Haven, CT, United States.,Department of Neuroscience, Yale School of Medicine, Yale University, New Haven, CT, United States
| |
Collapse
|
36
|
Vicario M, Calì T. Measuring Ca 2+ Levels in Subcellular Compartments with Genetically Encoded GFP-Based Indicators. Methods Mol Biol 2019; 1925:31-42. [PMID: 30674014 DOI: 10.1007/978-1-4939-9018-4_3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/30/2023]
Abstract
Ca2+ homeostasis is crucial for the entire life of eukaryotic cells from the beginning to the end. Mishandling in Ca2+ homeostasis is indeed linked with a large number of pathological conditions. Thus, the possibility to specifically monitor cellular calcium fluxes in different subcellular compartments represents a key tool to deeply understand the mechanisms involved in cellular dysfunctions. To cope with this need, several Ca2+ indicators have been developed allowing to accurately measure both basal Ca2+ concentration and agonist-induced Ca2+ signals in a wide spectrum of organelles. Among these, the genetically encoded GFP-based indicators are routinely used to measure Ca2+ transients thanks to their ability to change their spectral properties in response to Ca2+ binding. In this chapter, we will describe a protocol that utilizes the GCaMP6f probe targeted to mitochondria (4mtGCaMP) to measure mitochondrial calcium levels in resting conditions in HeLa cells. This method allows to easily and quickly register alterations of mitochondrial Ca2+ homeostasis in different cell populations and experimental settings, representing a precious tool to unravel the pathological pathways leading to pathogenic conditions.
Collapse
Affiliation(s)
- Mattia Vicario
- Department of Biomedical Sciences, University of Padua, Padua, Italy
| | - Tito Calì
- Department of Biomedical Sciences, University of Padua, Padua, Italy.
| |
Collapse
|
37
|
Friedmann KS, Bozem M, Hoth M. Calcium signal dynamics in T lymphocytes: Comparing in vivo and in vitro measurements. Semin Cell Dev Biol 2019; 94:84-93. [PMID: 30630031 DOI: 10.1016/j.semcdb.2019.01.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Revised: 12/18/2018] [Accepted: 01/05/2019] [Indexed: 02/06/2023]
Abstract
Amplitude and kinetics of intracellular Ca2+ signals ([Ca2+]int) determine many immune cell functions. To mimic in vivo changes of [Ca2+]int in human immune cells, two approaches may be best suited: 1) Analyze primary human immune cells taken from blood under conditions resembling best physiological or pathophysiological conditions. 2.) Analyze the immune system in vivo or ex vivo in explanted tissue from small vertebrate animals, such as mice. With the help of genetically encoded Ca2+ indicators and intravital microscopy, [Ca2+]int have been investigated in murine T lymphocytes (T cells) in vivo during the last five years and in explanted lymph node (LN) during the last 10 years. There are several important reasons to compare [Ca2+]int measured in primary murine T lymphocytes in vivo and in vitro with [Ca2+]int measured in primary human T lymphocytes in vitro. First, how do human and murine data compare? Second, how do in vivo and in vitro data compare? Third, can in vitro data predict in vivo data? The last point is particularly important considering the many technical challenges that limit in vivo measurements and to reduce the number of animals sacrificed. This review summarizes and compares the results of the available publications on in vivo and in vitro [Ca2+]int measurements in T lymphocytes stimulated focally by antigen-presenting cells (APC) after forming an immunological synapse.
Collapse
Affiliation(s)
- Kim S Friedmann
- Department of Biophysics, Center for Integrative Physiology and Molecular Medicine, Medical Faculty, Saarland University, Homburg, Germany
| | - Monika Bozem
- Department of Biophysics, Center for Integrative Physiology and Molecular Medicine, Medical Faculty, Saarland University, Homburg, Germany
| | - Markus Hoth
- Department of Biophysics, Center for Integrative Physiology and Molecular Medicine, Medical Faculty, Saarland University, Homburg, Germany.
| |
Collapse
|
38
|
Greenwald EC, Mehta S, Zhang J. Genetically Encoded Fluorescent Biosensors Illuminate the Spatiotemporal Regulation of Signaling Networks. Chem Rev 2018; 118:11707-11794. [PMID: 30550275 PMCID: PMC7462118 DOI: 10.1021/acs.chemrev.8b00333] [Citation(s) in RCA: 316] [Impact Index Per Article: 52.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Cellular signaling networks are the foundation which determines the fate and function of cells as they respond to various cues and stimuli. The discovery of fluorescent proteins over 25 years ago enabled the development of a diverse array of genetically encodable fluorescent biosensors that are capable of measuring the spatiotemporal dynamics of signal transduction pathways in live cells. In an effort to encapsulate the breadth over which fluorescent biosensors have expanded, we endeavored to assemble a comprehensive list of published engineered biosensors, and we discuss many of the molecular designs utilized in their development. Then, we review how the high temporal and spatial resolution afforded by fluorescent biosensors has aided our understanding of the spatiotemporal regulation of signaling networks at the cellular and subcellular level. Finally, we highlight some emerging areas of research in both biosensor design and applications that are on the forefront of biosensor development.
Collapse
Affiliation(s)
- Eric C Greenwald
- University of California , San Diego, 9500 Gilman Drive, BRFII , La Jolla , CA 92093-0702 , United States
| | - Sohum Mehta
- University of California , San Diego, 9500 Gilman Drive, BRFII , La Jolla , CA 92093-0702 , United States
| | - Jin Zhang
- University of California , San Diego, 9500 Gilman Drive, BRFII , La Jolla , CA 92093-0702 , United States
| |
Collapse
|
39
|
Luo F, Wei Y, Wang Z, Luo M, Hu J. Genetically Encoded Neural Activity Indicators. BRAIN SCIENCE ADVANCES 2018. [DOI: 10.26599/bsa.2018.9050007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Recent years have witnessed the fascinating development of imaging approaches to studying neural activities; this progress has been based on an influx of ideas and methods from molecular biology and optical engineering. Here we review the design and application of genetically encoded indicators for calcium ions, membrane potential and neurotransmitters. We also summarize common strategies for the design and optimization of genetically encoded neural activity indicators.
Collapse
Affiliation(s)
- Fang Luo
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Yin Wei
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Ziyue Wang
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Minmin Luo
- National Institute of Biological Sciences, Beijing 102206, China
| | - Ji Hu
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| |
Collapse
|
40
|
Diao M, Qu X, Huang S. Calcium imaging in Arabidopsis pollen cells using G-CaMP5. JOURNAL OF INTEGRATIVE PLANT BIOLOGY 2018; 60:897-906. [PMID: 29424471 DOI: 10.1111/jipb.12642] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Accepted: 02/07/2018] [Indexed: 05/18/2023]
Abstract
Calcium (Ca2+ ) signaling has been implicated in pollen germination and pollen tube growth. To date, however, we still know very little about how exactly Ca2+ signaling links to various physiological subcellular processes during pollen germination and pollen tube growth. Given that Ca2+ signaling is tightly related to the cytosolic concentration and dynamics of Ca2+ , it is vital to trace the dynamic changes in Ca2+ levels in order to decode Ca2+ signaling. Here, we demonstrate that G-CaMP5 serves well as an indicator for monitoring cytosolic Ca2+ dynamics in pollen cells. Using this probe, we show that cytosolic Ca2+ changes dramatically during pollen germination, and, as reported previously, Ca2+ forms a tip-focused gradient in the pollen tube and undergoes oscillation in the tip region during pollen tube growth. In particular, using G-CaMP5 allowed us to capture the dynamic changes in the cytosolic Ca2+ concentration ([Ca2+ ]cyt ) in pollen tubes in response to various exogenous treatments. Our data suggest that G-CaMP5 is a suitable probe for monitoring the dynamics of [Ca2+ ]cyt in pollen cells.
Collapse
Affiliation(s)
- Min Diao
- Key Laboratory of Plant Molecular Physiology, Institute of Botany, Chinese Academy of Sciences, Beijing 100093, China
- Center for Plant Biology, School of Life Sciences, Tsinghua University, Beijing 100084, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xiaolu Qu
- Center for Plant Biology, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Shanjin Huang
- Center for Plant Biology, School of Life Sciences, Tsinghua University, Beijing 100084, China
| |
Collapse
|
41
|
Dexter JP, Biddle JW, Gunawardena J. Model discrimination for Ca 2+ -dependent regulation of myosin light chain kinase in smooth muscle contraction. FEBS Lett 2018; 592:2811-2821. [PMID: 30066333 DOI: 10.1002/1873-3468.13207] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Revised: 07/05/2018] [Accepted: 07/11/2018] [Indexed: 11/11/2022]
Abstract
Excitation-contraction coupling in smooth muscle is mediated by the Ca2+ - and calmodulin-dependent regulation of myosin light chain kinase. The precise mechanism of this regulation remains controversial, and several mathematical models have been proposed for the interaction of the three species. These models have previously been analyzed at steady state primarily by numerical simulation of differential equations, for which parameter values must be estimated from data. Here, we use the linear framework for timescale separation to demonstrate that models of this general kind can be solved analytically for an equilibrium steady state, without having to determine parameter values. This analysis leads to parameter-independent methods for discriminating between the models, for which we propose experiments that could be performed with existing methods.
Collapse
Affiliation(s)
- Joseph P Dexter
- Department of Systems Biology, Harvard Medical School, Boston, MA, USA
| | - John W Biddle
- Department of Systems Biology, Harvard Medical School, Boston, MA, USA
| | | |
Collapse
|
42
|
Abstract
Technologies for measuring the transient Ca2+ spikes that accompany neural signaling have revolutionized our understanding of the brain. Nevertheless, microscopic visualization of Ca2+ spikes on the time scale of neural activity across large brain regions or in thick specimens remains a significant challenge. The recent development of stable integrators of Ca2+, instead of transient reporters, provides an avenue to investigate neural signaling in otherwise challenging systems. Here, we describe an engineered Ca2+-sensing enzyme consisting of a split Tobacco Etch Virus (TEV) protease with each half tethered to a calmodulin or M13 Ca2+ binding domain. This Split TEV, Ca2+ Activated Neuron Recorder (SCANR) remains separate and catalytically incompetent until a spike in cellular Ca2+ triggers its reconstitution and the subsequent turnover of a caged, genetically encoded reporter substrate. We report the identification of a successful Ca2+-sensing split TEV from a library of chimeras and deployment of the enzyme in primary rat hippocampal neurons.
Collapse
Affiliation(s)
- Brianna K. O’Neill
- Department of Chemistry, Stony Brook University, Stony Brook, New York 11794, United States
| | - Scott T. Laughlin
- Department of Chemistry, Stony Brook University, Stony Brook, New York 11794, United States
| |
Collapse
|
43
|
Dissection of Protein Kinase Pathways in Live Cells Using Photoluminescent Probes: Surveillance or Interrogation? CHEMOSENSORS 2018. [DOI: 10.3390/chemosensors6020019] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
44
|
Boss C, Bouche N, De Marchi U. Encapsulated Optically Responsive Cell Systems: Toward Smart Implants in Biomedicine. Adv Healthc Mater 2018; 7:e1701148. [PMID: 29283209 DOI: 10.1002/adhm.201701148] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Revised: 11/06/2017] [Indexed: 01/09/2023]
Abstract
Managing increasingly prevalent chronic diseases will require close continuous monitoring of patients. Cell-based biosensors may be used for implantable diagnostic systems to monitor health status. Cells are indeed natural sensors in the body. Functional cellular systems can be maintained in the body for long-term implantation using cell encapsulation technology. By taking advantage of recent progress in miniaturized optoelectronic systems, the genetic engineering of optically responsive cells may be combined with cell encapsulation to generate smart implantable cell-based sensing systems. In biomedical research, cell-based biosensors may be used to study cell signaling, therapeutic effects, and dosing of bioactive molecules in preclinical models. Today, a wide variety of genetically encoded fluorescent sensors have been developed for real-time imaging of living cells. Here, recent developments in genetically encoded sensors, cell encapsulation, and ultrasmall optical systems are highlighted. The integration of these components in a new generation of biosensors is creating innovative smart in vivo cell-based systems, bringing novel perspectives for biomedical research and ultimately allowing unique health monitoring applications.
Collapse
Affiliation(s)
- Christophe Boss
- Device EngineeringNestlé Institute of Health Sciences EPFL Innovation Park Lausanne CH‐1015 Switzerland
| | - Nicolas Bouche
- Device EngineeringNestlé Institute of Health Sciences EPFL Innovation Park Lausanne CH‐1015 Switzerland
| | - Umberto De Marchi
- Mitochondrial FunctionNestlé Institute of Health Sciences EPFL Innovation Park Lausanne CH‐1015 Switzerland
| |
Collapse
|
45
|
Abstract
Intracellular levels of the RNA-binding protein and pluripotency factor, Lin28a, are tightly controlled to govern cellular and organismal growth. Lin28a is extensively regulated at the posttranscriptional level, and can undergo mitogen-activated protein kinase (MAPK)-mediated elevation from low basal levels in differentiated cells by phosphorylation-dependent stabilizing interaction with the RNA-silencing factor HIV TAR RNA-binding protein (TRBP). However, molecular and spatiotemporal details of this critical control mechanism remained unknown. In this work, we dissect the interacting regions of Lin28a and TRBP proteins and develop biosensors to visualize this interaction. We identify truncated domains of Lin28a and of TRBP that are sufficient to support coassociation and mutual elevation of protein levels, and a requirement for MAPK-dependent phosphorylation of TRBP at putative Erk-target serine 152, as well as Lin28a serine 200 phosphorylation, in mediating the increase of Lin28a protein by TRBP. The phosphorylation-dependent association of Lin28a and TRBP truncated constructs is leveraged to develop fluorescence resonance energy transfer (FRET)-based sensors for dynamic monitoring of Lin28a and TRBP interaction. We demonstrate the response of bimolecular and unimolecular FRET sensors to growth factor stimulation in living cells, with coimaging of Erk activation to achieve further understanding of the role of MAPK signaling in Lin28a regulation.
Collapse
Affiliation(s)
- Laurel M Oldach
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21205
| | - Kirill Gorshkov
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21205.,Therapeutics for Rare and Neglected Diseases Program, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD 20850
| | - William T Mills
- Department of Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore, MD 21205
| | - Jin Zhang
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21205.,Department of Pharmacology, University of California, San Diego, La Jolla, CA 92093
| | - Mollie K Meffert
- Department of Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore, MD 21205.,Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205
| |
Collapse
|
46
|
Dong TX, Othy S, Jairaman A, Skupsky J, Zavala A, Parker I, Dynes JL, Cahalan MD. T-cell calcium dynamics visualized in a ratiometric tdTomato-GCaMP6f transgenic reporter mouse. eLife 2017; 6:32417. [PMID: 29239725 PMCID: PMC5747524 DOI: 10.7554/elife.32417] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Accepted: 12/11/2017] [Indexed: 11/13/2022] Open
Abstract
Calcium is an essential cellular messenger that regulates numerous functions in living organisms. Here, we describe development and characterization of ‘Salsa6f’, a fusion of GCaMP6f and tdTomato optimized for cell tracking while monitoring cytosolic Ca2+, and a transgenic Ca2+ reporter mouse with Salsa6f targeted to the Rosa26 locus for Cre-dependent expression in specific cell types. The development and function of T cells was unaffected in Cd4-Salsa6f mice. We describe Ca2+ signals reported by Salsa6f during T cell receptor activation in naive T cells, helper Th17 T cells and regulatory T cells, and Ca2+ signals mediated in T cells by an activator of mechanosensitive Piezo1 channels. Transgenic expression of Salsa6f enables ratiometric imaging of Ca2+ signals in complex tissue environments found in vivo. Two-photon imaging of migrating T cells in the steady-state lymph node revealed both cell-wide and localized sub-cellular Ca2+ transients (‘sparkles’) as cells migrate. To help protect the body from disease, small immune cells called T lymphocytes move rapidly, searching for signs of infection. These signs are antigens – processed pieces of proteins from invading bacteria and viruses – which are displayed on the surface of so-called antigen-presenting cells. To visit as many different antigen-presenting cells as possible, T cells move quickly from one to the next in an apparently random manner. How T cells are programmed to move in this way is largely unknown. The entry of calcium ions into cells, through channel proteins, triggers characteristic actions in many cells throughout the body. As such it is possible that the T cells’ movements are related to calcium signals too. However, it was technically challenging to directly measure the amount of calcium in moving cells within the body. To overcome this issue, Dong, Othy et al. genetically engineered mice to produce a new calcium-sensitive reporter protein in their T cells. The reporter, which was named Salsa6f, consisted of a red fluorescent protein fused to another protein that glows green when it binds to calcium ions. Measuring the ratio of red and green fluorescence gives a measure of the concentration of calcium ions inside the cell. In the absence of calcium signaling, the cells can still be tracked via the red fluorescence of Salsa6f. Importantly, the reporter did not affect the development or activity of the T cells in the mice. In a related study, Dong, Othy et al. then used their transgenic mice to ask whether calcium signals guide moving T cells as they search for antigens. Future studies could use these transgenic mice to track the calcium ion concentration in numerous cell types. This would enable new approaches to relate the inner workings of cells to their behaviors in many different organ systems throughout the body.
Collapse
Affiliation(s)
- Tobias X Dong
- Department of Physiology and Biophysics, University of California, Irvine, United States
| | - Shivashankar Othy
- Department of Physiology and Biophysics, University of California, Irvine, United States
| | - Amit Jairaman
- Department of Physiology and Biophysics, University of California, Irvine, United States
| | - Jonathan Skupsky
- Department of Physiology and Biophysics, University of California, Irvine, United States.,Department of Medicine, University of California, Irvine, United States
| | - Angel Zavala
- Department of Physiology and Biophysics, University of California, Irvine, United States
| | - Ian Parker
- Department of Physiology and Biophysics, University of California, Irvine, United States.,Department of Neurobiology & Behavior, University of California, Irvine, United States
| | - Joseph L Dynes
- Department of Physiology and Biophysics, University of California, Irvine, United States
| | - Michael D Cahalan
- Department of Physiology and Biophysics, University of California, Irvine, United States.,Institute for Immunology, University of California, Irvine, United States
| |
Collapse
|
47
|
Chemin J, Taiakina V, Monteil A, Piazza M, Guan W, Stephens RF, Kitmitto A, Pang ZP, Dolphin AC, Perez-Reyes E, Dieckmann T, Guillemette JG, Spafford JD. Calmodulin regulates Ca v3 T-type channels at their gating brake. J Biol Chem 2017; 292:20010-20031. [PMID: 28972185 PMCID: PMC5723990 DOI: 10.1074/jbc.m117.807925] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Revised: 09/19/2017] [Indexed: 01/10/2023] Open
Abstract
Calcium (Cav1 and Cav2) and sodium channels possess homologous CaM-binding motifs, known as IQ motifs in their C termini, which associate with calmodulin (CaM), a universal calcium sensor. Cav3 T-type channels, which serve as pacemakers of the mammalian brain and heart, lack a C-terminal IQ motif. We illustrate that T-type channels associate with CaM using co-immunoprecipitation experiments and single particle cryo-electron microscopy. We demonstrate that protostome invertebrate (LCav3) and human Cav3.1, Cav3.2, and Cav3.3 T-type channels specifically associate with CaM at helix 2 of the gating brake in the I-II linker of the channels. Isothermal titration calorimetry results revealed that the gating brake and CaM bind each other with high-nanomolar affinity. We show that the gating brake assumes a helical conformation upon binding CaM, with associated conformational changes to both CaM lobes as indicated by amide chemical shifts of the amino acids of CaM in 1H-15N HSQC NMR spectra. Intact Ca2+-binding sites on CaM and an intact gating brake sequence (first 39 amino acids of the I-II linker) were required in Cav3.2 channels to prevent the runaway gating phenotype, a hyperpolarizing shift in voltage sensitivities and faster gating kinetics. We conclude that the presence of high-nanomolar affinity binding sites for CaM at its universal gating brake and its unique form of regulation via the tuning of the voltage range of activity could influence the participation of Cav3 T-type channels in heart and brain rhythms. Our findings may have implications for arrhythmia disorders arising from mutations in the gating brake or CaM.
Collapse
Affiliation(s)
- Jean Chemin
- Institut de Génomique Fonctionnelle, CNRS, INSERM, Université de Montpellier, Montpellier F-34094, France
| | | | - Arnaud Monteil
- Institut de Génomique Fonctionnelle, CNRS, INSERM, Université de Montpellier, Montpellier F-34094, France
| | - Michael Piazza
- Departments of Chemistry, Waterloo, Ontario N2L 3G1, Canada
| | - Wendy Guan
- Biology, University of Waterloo, Waterloo, Ontario N2L 3G1, Canada
| | | | - Ashraf Kitmitto
- Division of Cardiovascular Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9NT, United Kingdom
| | - Zhiping P Pang
- Department of Neuroscience and Cell Biology, Rutgers Robert Wood Johnson Medical School, New Brunswick, New Jersey 08901
| | - Annette C Dolphin
- Department of Neuroscience, Physiology and Pharmacology, University College London, London WC1E 6BT, United Kingdom
| | - Edward Perez-Reyes
- Department of Pharmacology, University of Virginia, Charlottesville, Virginia 22908
| | | | | | - J David Spafford
- Biology, University of Waterloo, Waterloo, Ontario N2L 3G1, Canada.
| |
Collapse
|
48
|
Hovey L, Fowler CA, Mahling R, Lin Z, Miller MS, Marx DC, Yoder JB, Kim EH, Tefft KM, Waite BC, Feldkamp MD, Yu L, Shea MA. Calcium triggers reversal of calmodulin on nested anti-parallel sites in the IQ motif of the neuronal voltage-dependent sodium channel Na V1.2. Biophys Chem 2017; 224:1-19. [PMID: 28343066 PMCID: PMC5503752 DOI: 10.1016/j.bpc.2017.02.006] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2017] [Revised: 02/23/2017] [Accepted: 02/23/2017] [Indexed: 01/26/2023]
Abstract
Several members of the voltage-gated sodium channel family are regulated by calmodulin (CaM) and ionic calcium. The neuronal voltage-gated sodium channel NaV1.2 contains binding sites for both apo (calcium-depleted) and calcium-saturated CaM. We have determined equilibrium dissociation constants for rat NaV1.2 IQ motif [IQRAYRRYLLK] binding to apo CaM (~3nM) and (Ca2+)4-CaM (~85nM), showing that apo CaM binding is favored by 30-fold. For both apo and (Ca2+)4-CaM, NMR demonstrated that NaV1.2 IQ motif peptide (NaV1.2IQp) exclusively made contacts with C-domain residues of CaM (CaMC). To understand how calcium triggers conformational change at the CaM-IQ interface, we determined a solution structure (2M5E.pdb) of (Ca2+)2-CaMC bound to NaV1.2IQp. The polarity of (Ca2+)2-CaMC relative to the IQ motif was opposite to that seen in apo CaMC-Nav1.2IQp (2KXW), revealing that CaMC recognizes nested, anti-parallel sites in Nav1.2IQp. Reversal of CaM may require transient release from the IQ motif during calcium binding, and facilitate a re-orientation of CaMN allowing interactions with non-IQ NaV1.2 residues or auxiliary regulatory proteins interacting in the vicinity of the IQ motif.
Collapse
Affiliation(s)
- Liam Hovey
- Department of Biochemistry, University of Iowa, 52242-1109 Iowa City, United States
| | - C Andrew Fowler
- NMR Facility, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, 52242-1109 Iowa City, United States
| | - Ryan Mahling
- Department of Biochemistry, University of Iowa, 52242-1109 Iowa City, United States
| | - Zesen Lin
- Department of Biochemistry, University of Iowa, 52242-1109 Iowa City, United States
| | - Mark Stephen Miller
- Department of Biochemistry, University of Iowa, 52242-1109 Iowa City, United States
| | - Dagan C Marx
- Department of Biochemistry, University of Iowa, 52242-1109 Iowa City, United States
| | - Jesse B Yoder
- Department of Biochemistry, University of Iowa, 52242-1109 Iowa City, United States
| | - Elaine H Kim
- Department of Biochemistry, University of Iowa, 52242-1109 Iowa City, United States
| | - Kristin M Tefft
- Department of Biochemistry, University of Iowa, 52242-1109 Iowa City, United States
| | - Brett C Waite
- Department of Biochemistry, University of Iowa, 52242-1109 Iowa City, United States
| | - Michael D Feldkamp
- Department of Biochemistry, University of Iowa, 52242-1109 Iowa City, United States
| | - Liping Yu
- NMR Facility, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, 52242-1109 Iowa City, United States
| | - Madeline A Shea
- Department of Biochemistry, University of Iowa, 52242-1109 Iowa City, United States.
| |
Collapse
|
49
|
Chen J, Xia L, Bruchas MR, Solnica-Krezel L. Imaging early embryonic calcium activity with GCaMP6s transgenic zebrafish. Dev Biol 2017; 430:385-396. [PMID: 28322738 PMCID: PMC5835148 DOI: 10.1016/j.ydbio.2017.03.010] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2016] [Revised: 01/12/2017] [Accepted: 03/11/2017] [Indexed: 12/02/2022]
Abstract
Intracellular Ca2+ signaling regulates cellular activities during embryogenesis and in adult organisms. We generated stable Tg[βactin2:GCaMP6s]stl351 and Tg[ubi:GCaMP6s]stl352 transgenic lines that combine the ubiquitously-expressed Ca2+ indicator GCaMP6s with the transparent characteristics of zebrafish embryos to achieve superior in vivo Ca2+ imaging. Using the Tg[βactin2:GCaMP6s]stl351 line featuring strong GCaMP6s expression from cleavage through gastrula stages, we detected higher frequency of Ca2+ transients in the superficial blastomeres during the blastula stages preceding the midblastula transition. Additionally, GCaMP6s also revealed that dorsal-biased Ca2+ signaling that follows the midblastula transition persisted longer during gastrulation, compared with earlier studies. We observed that dorsal-biased Ca2+ signaling is diminished in ventralized ichabod/β-catenin2 mutant embryos and ectopically induced in embryos dorsalized by excess β-catenin. During gastrulation, we directly visualized Ca2+ signaling in the dorsal forerunner cells, which form in a Nodal signaling dependent manner and later give rise to the laterality organ. We found that excess Nodal increases the number and the duration of Ca2+ transients specifically in the dorsal forerunner cells. The GCaMP6s transgenic lines described here enable unprecedented visualization of dynamic Ca2+ events from embryogenesis through adulthood, augmenting the zebrafish toolbox.
Collapse
Affiliation(s)
- Jiakun Chen
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Li Xia
- Department of Anesthesiology, Division of Basic Research, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Biomedical Engineering, Washington University in St. Louis, St Louis, MO, 63105, USA
| | - Michael R Bruchas
- Department of Anesthesiology, Division of Basic Research, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Biomedical Engineering, Washington University in St. Louis, St Louis, MO, 63105, USA; Department of Neuroscience, Washington University in St. Louis, St. Louis, MO, USA
| | - Lilianna Solnica-Krezel
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA.
| |
Collapse
|
50
|
Ma Q, Ye L, Liu H, Shi Y, Zhou N. An overview of Ca 2+ mobilization assays in GPCR drug discovery. Expert Opin Drug Discov 2017; 12:511-523. [PMID: 28277837 DOI: 10.1080/17460441.2017.1303473] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
INTRODUCTION Calcium ions (Ca2+) serve as a second messenger or universal signal transducer implicated in the regulation of a wide range of physiological processes. A change in the concentration of intracellular Ca2+ is an important step in intracellular signal transduction. G protein-coupled receptors (GPCRs), the largest and most versatile group of cell surface receptors, transduce extracellular signals into intracellular responses via their coupling to heterotrimeric G proteins. Since Ca2+ plays a crucial role in GPCR-induced signaling, measurement of intracellular Ca2+ has attracted more and more attention in GPCR-targeted drug discovery. Areas covered: This review focuses on the most popular functional assays measuring GPCRs-induced intracellular Ca2+ signaling. These include photoprotein-based, synthetic fluorescent indicator-based and genetically encoded calcium indicator (GECI)-based Ca2+ mobilization assays. A brief discussion of the design strategy of fluorescent probes in GPCR studies is also presented. Expert opinion: GPCR-mediated intracellular signaling is multidimensional. There is an urgent need for the development of multiple-readout screening assays capable of simultaneous detection of biased signaling and screening of both agonists and antagonists in the same assay. It is also necessary to develop GECIs offering low cost and consistent assays suitable for investigating GPCR activation in vivo.
Collapse
Affiliation(s)
- Qiang Ma
- a College of Life Sciences, Zijingang Campus , Zhejiang University, Institute of Biochemistry and Molecular Biology , Hangzhou , Zhejiang , China
| | - Lingyan Ye
- a College of Life Sciences, Zijingang Campus , Zhejiang University, Institute of Biochemistry and Molecular Biology , Hangzhou , Zhejiang , China
| | - Hongxia Liu
- b Department of Internal Medicine , Edong Healthcare Group , Huangshi , Hubei , China
| | - Ying Shi
- a College of Life Sciences, Zijingang Campus , Zhejiang University, Institute of Biochemistry and Molecular Biology , Hangzhou , Zhejiang , China
| | - Naiming Zhou
- a College of Life Sciences, Zijingang Campus , Zhejiang University, Institute of Biochemistry and Molecular Biology , Hangzhou , Zhejiang , China
| |
Collapse
|