Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 57
Filter
1.
Cell ; 182(6): 1531-1544.e15, 2020 09 17.
Article in English | MEDLINE | ID: mdl-32846158

ABSTRACT

The fidelity of intracellular signaling hinges on the organization of dynamic activity architectures. Spatial compartmentation was first proposed over 30 years ago to explain how diverse G protein-coupled receptors achieve specificity despite converging on a ubiquitous messenger, cyclic adenosine monophosphate (cAMP). However, the mechanisms responsible for spatially constraining this diffusible messenger remain elusive. Here, we reveal that the type I regulatory subunit of cAMP-dependent protein kinase (PKA), RIα, undergoes liquid-liquid phase separation (LLPS) as a function of cAMP signaling to form biomolecular condensates enriched in cAMP and PKA activity, critical for effective cAMP compartmentation. We further show that a PKA fusion oncoprotein associated with an atypical liver cancer potently blocks RIα LLPS and induces aberrant cAMP signaling. Loss of RIα LLPS in normal cells increases cell proliferation and induces cell transformation. Our work reveals LLPS as a principal organizer of signaling compartments and highlights the pathological consequences of dysregulating this activity architecture.


Subject(s)
Carcinogenesis/metabolism , Carcinoma, Hepatocellular/genetics , Cell Compartmentation/genetics , Cyclic AMP-Dependent Protein Kinase RIalpha Subunit/metabolism , Cyclic AMP/metabolism , HSP40 Heat-Shock Proteins/genetics , Liver Neoplasms/genetics , Signal Transduction , Animals , Carcinogenesis/drug effects , Carcinogenesis/genetics , Carcinoma, Hepatocellular/metabolism , Cell Compartmentation/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/genetics , Cyclic AMP/pharmacology , Cyclic AMP-Dependent Protein Kinase RIalpha Subunit/genetics , Cyclic AMP-Dependent Protein Kinases/genetics , Cyclic AMP-Dependent Protein Kinases/metabolism , Cytoplasm/metabolism , Humans , Liver Neoplasms/metabolism , Mice , Oncogenes/genetics , Protein Domains , Rats , Rats, Sprague-Dawley , Recombinant Fusion Proteins , Spectroscopy, Fourier Transform Infrared , Time-Lapse Imaging/methods
2.
Mol Cell ; 84(8): 1570-1584.e7, 2024 Apr 18.
Article in English | MEDLINE | ID: mdl-38537638

ABSTRACT

Spatiotemporal regulation of intracellular signaling molecules, such as the 3',5'-cyclic adenosine monophosphate (cAMP)-dependent protein kinase (PKA), ensures proper cellular function. Liquid-liquid phase separation (LLPS) of the ubiquitous PKA regulatory subunit RIα promotes cAMP compartmentation and signaling specificity. However, the molecular determinants of RIα LLPS remain unclear. Here, we reveal that two separate dimerization interfaces, combined with the cAMP-induced unleashing of the PKA catalytic subunit (PKA-C) from the pseudosubstrate inhibitory sequence, drive RIα condensate formation in the cytosol of mammalian cells, which is antagonized by docking to A-kinase anchoring proteins. Strikingly, we find that the RIα pseudosubstrate region is critically involved in forming a non-canonical R:C complex, which recruits active PKA-C to RIα condensates to maintain low basal PKA activity in the cytosol. Our results suggest that RIα LLPS not only facilitates cAMP compartmentation but also spatially restrains active PKA-C, thus highlighting the functional versatility of biomolecular condensates in driving signaling specificity.


Subject(s)
Cyclic AMP-Dependent Protein Kinase RIalpha Subunit , Phase Separation , Animals , Cyclic AMP-Dependent Protein Kinase RIalpha Subunit/genetics , Cyclic AMP-Dependent Protein Kinase RIalpha Subunit/chemistry , Cyclic AMP-Dependent Protein Kinase RIalpha Subunit/metabolism , Signal Transduction , Cyclic AMP/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Mammals/metabolism
3.
Mol Cell ; 81(20): 4137-4146, 2021 10 21.
Article in English | MEDLINE | ID: mdl-34619090

ABSTRACT

Cell signaling is a complex process. The faithful transduction of information into specific cellular actions depends on the synergistic effects of many regulatory molecules, nurtured by their strict spatiotemporal regulation. Over the years, we have gained copious insights into the subcellular architecture supporting this spatiotemporal control, including the roles of membrane-bound organelles and various signaling nanodomains. Recently, liquid-liquid phase separation (LLPS) has been recognized as another potentially ubiquitous framework for organizing signaling molecules with high specificity and precise spatiotemporal control in cells. Here, we review the pervasive role of LLPS in signal transduction, highlighting several key pathways that intersect with LLPS, including examples in which LLPS is controlled by signaling events. We also examine how LLPS orchestrates signaling by compartmentalizing signaling molecules, amplifying signals non-linearly, and moderating signaling dynamics. We focus on the specific molecules that drive LLPS and highlight the known functional and pathological consequences of LLPS in each pathway.


Subject(s)
Cell Compartmentation , Intracellular Signaling Peptides and Proteins/metabolism , Intracellular Space/metabolism , Intrinsically Disordered Proteins/metabolism , Organelles/metabolism , Signal Transduction , Animals , Humans , Intracellular Signaling Peptides and Proteins/chemistry , Intrinsically Disordered Proteins/chemistry , Time Factors
4.
Nature ; 611(7934): 173-179, 2022 11.
Article in English | MEDLINE | ID: mdl-36289326

ABSTRACT

G-protein-coupled receptors (GPCRs), the largest family of signalling receptors, as well as important drug targets, are known to activate extracellular-signal-regulated kinase (ERK)-a master regulator of cell proliferation and survival1. However, the precise mechanisms that underlie GPCR-mediated ERK activation are not clearly understood2-4. Here we investigated how spatially organized ß2-adrenergic receptor (ß2AR) signalling controls ERK. Using subcellularly targeted ERK activity biosensors5, we show that ß2AR signalling induces ERK activity at endosomes, but not at the plasma membrane. This pool of ERK activity depends on active, endosome-localized Gαs and requires ligand-stimulated ß2AR endocytosis. We further identify an endosomally localized non-canonical signalling axis comprising Gαs, RAF and mitogen-activated protein kinase kinase, resulting in endosomal ERK activity that propagates into the nucleus. Selective inhibition of endosomal ß2AR and Gαs signalling blunted nuclear ERK activity, MYC gene expression and cell proliferation. These results reveal a non-canonical mechanism for the spatial regulation of ERK through GPCR signalling and identify a functionally important endosomal signalling axis.


Subject(s)
Adrenergic Agents , Endosomes , Extracellular Signal-Regulated MAP Kinases , Receptors, Adrenergic, beta-2 , Adrenergic Agents/metabolism , Adrenergic Agents/pharmacology , Cell Proliferation , Endosomes/drug effects , Endosomes/enzymology , Endosomes/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Genes, myc , GTP-Binding Protein alpha Subunits, Gs/metabolism , Mitogen-Activated Protein Kinase Kinases/metabolism , Phosphorylation/drug effects , Receptors, Adrenergic, beta-2/metabolism , Signal Transduction/drug effects , Signal Transduction/physiology
5.
Annu Rev Biochem ; 80: 375-401, 2011.
Article in English | MEDLINE | ID: mdl-21495849

ABSTRACT

Real-time visualization of a wide range of biochemical processes in living systems is being made possible through the development and application of genetically encoded fluorescent reporters. These versatile biosensors have proven themselves tailor-made to the study of signal transduction, and in this review, we discuss some of the unique insights that they continue to provide regarding the spatial organization and dynamic regulation of intracellular signaling networks. In addition, we explore the more recent push to expand the scope of biological phenomena that can be monitored using these reporters, while also considering the potential to integrate this highly adaptable technology with a number of emerging techniques that may significantly broaden our view of how networks of biochemical processes shape larger biological phenomena.


Subject(s)
Biosensing Techniques/methods , Fluorescence Resonance Energy Transfer/methods , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Signal Transduction/physiology , Animals , Calcium/metabolism , Fluorescent Dyes/chemistry , Fluorescent Dyes/metabolism , Green Fluorescent Proteins/chemistry , Intracellular Membranes/chemistry , Models, Molecular
6.
Trends Biochem Sci ; 45(10): 889-905, 2020 10.
Article in English | MEDLINE | ID: mdl-32660810

ABSTRACT

Cell signaling networks are intricately regulated in time and space to determine the responses and fates of cells to different cues. Genetically encodable fluorescent and bioluminescent biosensors enable the direct visualization of these spatiotemporal signaling dynamics within the native biological context, and have therefore become powerful molecular tools whose unique benefits are being used to address challenging biological questions. We first review the basis of biosensor design and remark on recent technologies that are accelerating biosensor development. We then discuss a few of the latest advances in the development and application of genetically encodable fluorescent and bioluminescent biosensors that have led to scientific or technological breakthroughs.


Subject(s)
Luminescence , Signal Transduction , Biosensing Techniques/methods
7.
Annu Rev Pharmacol Toxicol ; 61: 587-608, 2021 01 06.
Article in English | MEDLINE | ID: mdl-33411579

ABSTRACT

How cells muster a network of interlinking signaling pathways to faithfully convert diverse external cues to specific functional outcomes remains a central question in biology. Through their ability to convert dynamic biochemical activities to rapid and precise optical readouts, genetically encoded fluorescent biosensors have become instrumental in unraveling the molecular logic controlling the specificity of intracellular signaling. In this review, we discuss how the use of genetically encoded fluorescent biosensors to visualize dynamic signaling events within their native cellular context is elucidating the different strategies employed by cells to organize signaling activities into discrete compartments, or signaling microdomains, to ensure functional specificity.


Subject(s)
Biosensing Techniques , Fluorescence Resonance Energy Transfer , Humans , Signal Transduction
8.
Biochem J ; 480(20): 1693-1717, 2023 10 31.
Article in English | MEDLINE | ID: mdl-37903110

ABSTRACT

As cell signaling research has advanced, it has become clearer that signal transduction has complex spatiotemporal regulation that goes beyond foundational linear transduction models. Several technologies have enabled these discoveries, including fluorescent biosensors designed to report live biochemical signaling events. As genetically encoded and live-cell compatible tools, fluorescent biosensors are well suited to address diverse cell signaling questions across different spatial scales of regulation. In this review, methods of examining spatial signaling regulation and the design of fluorescent biosensors are introduced. Then, recent biosensor developments that illuminate the importance of spatial regulation in cell signaling are highlighted at several scales, including membranes and organelles, molecular assemblies, and cell/tissue heterogeneity. In closing, perspectives on how fluorescent biosensors will continue enhancing cell signaling research are discussed.


Subject(s)
Biosensing Techniques , Fluorescence Resonance Energy Transfer , Signal Transduction , Fluorescent Dyes
9.
J Physiol ; 601(19): 4227-4241, 2023 10.
Article in English | MEDLINE | ID: mdl-37747358

ABSTRACT

Cells execute specific responses to diverse environmental cues by encoding information in distinctly compartmentalized biochemical signalling reactions. Genetically encoded fluorescent biosensors enable the spatial and temporal monitoring of signalling events in live cells. Temporal and spatiotemporal computational models can be used to interpret biosensor experiments in complex biochemical networks and to explore hypotheses that are difficult to test experimentally. In this review, we first provide brief discussions of the experimental toolkit of fluorescent biosensors as well as computational basics with a focus on temporal and spatiotemporal deterministic models. We then describe how we used this combined approach to identify and investigate a protein kinase A (PKA) - cAMP - Ca2+ oscillatory circuit in MIN6 ß cells, a mouse pancreatic ß cell system. We describe the application of this combined approach to interrogate how this oscillatory circuit is differentially regulated in a nano-compartment formed at the plasma membrane by the scaffolding protein A kinase anchoring protein 79/150. We leveraged both temporal and spatiotemporal deterministic models to identify the key regulators of this oscillatory circuit, which we confirmed with further experiments. The powerful approach of combining live-cell biosensor imaging with quantitative modelling, as discussed here, should find widespread use in the investigation of spatiotemporal regulation of cell signalling.


Subject(s)
Biosensing Techniques , Signal Transduction , Animals , Mice , Cyclic AMP-Dependent Protein Kinases/metabolism , Diagnostic Imaging , Cell Membrane/metabolism , Biosensing Techniques/methods , Fluorescence Resonance Energy Transfer/methods
10.
Nat Chem Biol ; 17(1): 39-46, 2021 01.
Article in English | MEDLINE | ID: mdl-32989297

ABSTRACT

Protein kinases control nearly every facet of cellular function. These key signaling nodes integrate diverse pathway inputs to regulate complex physiological processes, and aberrant kinase signaling is linked to numerous pathologies. While fluorescent protein-based biosensors have revolutionized the study of kinase signaling by allowing direct, spatiotemporally precise kinase activity measurements in living cells, powerful new molecular tools capable of robustly tracking kinase activity dynamics across diverse experimental contexts are needed to fully dissect the role of kinase signaling in physiology and disease. Here, we report the development of an ultrasensitive, second-generation excitation-ratiometric protein kinase A (PKA) activity reporter (ExRai-AKAR2), obtained via high-throughput linker library screening, that enables sensitive and rapid monitoring of live-cell PKA activity across multiple fluorescence detection modalities, including plate reading, cell sorting and one- or two-photon imaging. Notably, in vivo visual cortex imaging in awake mice reveals highly dynamic neuronal PKA activity rapidly recruited by forced locomotion.


Subject(s)
Biosensing Techniques , Cyclic AMP-Dependent Protein Kinases/genetics , Myocytes, Cardiac/enzymology , Neurons/enzymology , Optical Imaging/methods , Alprostadil/pharmacology , Animals , Cyclic AMP-Dependent Protein Kinases/metabolism , Dihydroxyphenylalanine/pharmacology , Dinoprostone/pharmacology , Fluorescent Dyes/chemistry , Gene Expression , Gene Library , Genes, Reporter , Glucagon-Like Peptide 1/pharmacology , HEK293 Cells , HeLa Cells , High-Throughput Screening Assays , Hippocampus/cytology , Hippocampus/drug effects , Hippocampus/enzymology , Humans , Mice , Microscopy, Fluorescence, Multiphoton , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/ultrastructure , Neurons/drug effects , Neurons/ultrastructure , Primary Cell Culture , Signal Transduction
11.
Nat Methods ; 16(2): 171-174, 2019 02.
Article in English | MEDLINE | ID: mdl-30664778

ABSTRACT

We report an intensiometric, near-infrared fluorescent, genetically encoded calcium ion (Ca2+) indicator (GECI) with excitation and emission maxima at 678 and 704 nm, respectively. This GECI, designated NIR-GECO1, enables imaging of Ca2+ transients in cultured mammalian cells and brain tissue with sensitivity comparable to that of currently available visible-wavelength GECIs. We demonstrate that NIR-GECO1 opens up new vistas for multicolor Ca2+ imaging in combination with other optogenetic indicators and actuators.


Subject(s)
Calcium/chemistry , Fluorescent Dyes/chemistry , Microscopy, Fluorescence/methods , Spectroscopy, Near-Infrared/methods , Animals , Biliverdine/chemistry , DNA/analysis , Escherichia coli/chemistry , Female , Fluorescence Resonance Energy Transfer , Genetic Vectors , HeLa Cells , Hippocampus/chemistry , Humans , Ions , Male , Mice , Mice, Inbred C57BL , Microscopy, Confocal , Neurons/chemistry , Optogenetics , Protein Domains
12.
Acc Chem Res ; 54(10): 2409-2420, 2021 05 18.
Article in English | MEDLINE | ID: mdl-33949851

ABSTRACT

All biological processes arise through the coordinated actions of biochemical pathways. How such functional diversity is achieved by a finite cast of molecular players remains a central mystery in biology. Spatial compartmentation-the idea that biochemical activities are organized around discrete spatial domains within cells-was first proposed nearly 40 years ago and has become firmly rooted in our understanding of how biochemical pathways are regulated to ensure specificity. However, directly interrogating spatial compartmentation and its mechanistic origins has only really become possible in the last 20 or so years, following technological advances such as the development of genetically encoded fluorescent biosensors. These powerful molecular tools permit a direct, real-time visualization of dynamic biochemical processes in native biological contexts, and they are essential for probing the spatial regulation of biochemical activities. In this Account, we review our lab's efforts in developing and using biosensors to map the spatial compartmentation of intracellular signaling pathways and illuminate key mechanisms that establish the boundaries of an intricate biochemical activity architecture. We first discuss the role of regulatory fences, wherein the dynamic activation and deactivation of diffusible messengers produce diverse signaling compartments. For example, we used biosensors for the Ca2+ effector calmodulin and its downstream target calcineurin to reveal a spatial gradient of calmodulin that controls the temporal dynamics of calcineurin signaling. Our studies using cyclic adenosine monophosphate (cAMP) biosensors have similarly elucidated fenced cAMP domains generated by competing production and degradation pathways, ranging in size from cell-spanning gradients to nanoscale hotspots. Second, we describe the role played by intracellular membranes in creating unique signaling platforms with distinctive pathway regulation, as revealed through studies using subcellularly targeted fluorescent biosensors. Using biosensors to visualize subcellular extracellular response kinase (ERK) pathway activity, for example, led us to discover a local signaling circuit that mediates distinct plasma membrane ERK dynamics versus global ERK signaling. Similarly, our work developing biosensors to monitor the subcellular mechanistic target of rapamycin complex 1 (mTORC1) signaling allowed us to not only clarify the presence of mTORC1 activity in the nucleus but also identify a novel mechanism governing the activation of mTORC1 in this location. Finally, we detail how molecular assemblies enable the precise spatial tuning of biochemical activity, through investigations enabled by cutting-edge advances in biosensor design. We recently identified liquid-liquid phase separation as a major factor in cAMP compartmentation aided by a new strategy for targeting biosensors to endogenously expressed proteins via genome editing, for instance, and have also been able to directly visualize nanometer-scale protein kinase signalosomes using an entirely new class of biosensors specifically developed for the dynamic super-resolution imaging of live-cell biochemical activities. Our work provides key insights into the molecular logic of spatially regulated signaling and lays the foundation for a broader exploration of biochemical activity architectures across multiple spatial scales.


Subject(s)
Biosensing Techniques , Calcineurin/analysis , Calmodulin/analysis , Fluorescence , Mechanistic Target of Rapamycin Complex 1/analysis , Humans , Mechanistic Target of Rapamycin Complex 1/metabolism
13.
J Am Chem Soc ; 143(37): 14951-14955, 2021 09 22.
Article in English | MEDLINE | ID: mdl-34516108

ABSTRACT

Super-resolution activity imaging maps the biochemical architecture of living cells yet currently overlooks the locations of collaborating regulators/effectors. Building on the fluorescence fluctuation increase by contact (FLINC) principle, here we devise Dronpa-chromophore-removed FLINC (DrFLINC), where the nonfluorescent Dronpa can nevertheless enhance TagRFP-T fluorescence fluctuations. Exploiting DrFLINC, we develop a superior red label and a next-generation activity sensor for context-rich super-resolution biosensing.


Subject(s)
Fluorescent Dyes/chemistry , Green Fluorescent Proteins/chemistry , Fluorescence , HeLa Cells , Humans
14.
J Biol Chem ; 294(40): 14814-14822, 2019 10 04.
Article in English | MEDLINE | ID: mdl-31434714

ABSTRACT

Protein kinase signaling networks stringently regulate cellular processes, such as proliferation, motility, and cell survival. These networks are also central to the evolution and progression of cancer. Accordingly, genetically encoded fluorescent biosensors capable of directly illuminating the spatiotemporal dynamics of kinase signaling in live cells are being increasingly used to investigate kinase signaling in cancer cells and tumor tissue sections. These biosensors enable visualization of biological processes and events directly in situ, preserving the native biological context and providing detailed insight into their localization and dynamics in cells. Herein, we first review common design strategies for kinase activity biosensors, including signaling targets, biosensor components, and fluorescent proteins involved. Subsequently, we discuss applications of biosensors to study the biology and management of cancer. These versatile molecular tools have been deployed to study oncogenic kinase signaling in living cells and image kinase activities in tumors or to decipher the mechanisms of anticancer drugs. We anticipate that the diversity and precision of genetically encoded biosensors will expand their use to further unravel the dysregulation of kinase signaling in cancer and the modes of actions of cancer-targeting drugs.


Subject(s)
Biosensing Techniques , Neoplasms/genetics , Phosphotransferases/genetics , Protein Kinases/genetics , Fluorescent Dyes/chemistry , Green Fluorescent Proteins/genetics , Humans , Neoplasms/enzymology , Phosphorylation , Phosphotransferases/isolation & purification , Protein Kinases/isolation & purification , Signal Transduction/genetics
15.
Chem Rev ; 118(24): 11707-11794, 2018 12 26.
Article in English | MEDLINE | ID: mdl-30550275

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.


Subject(s)
Biosensing Techniques , Luminescent Proteins/genetics , Signal Transduction/genetics , Animals , Humans , Luminescent Proteins/chemistry , Luminescent Proteins/metabolism
16.
Nat Chem Biol ; 13(4): 425-431, 2017 04.
Article in English | MEDLINE | ID: mdl-28192412

ABSTRACT

Cyclic AMP (cAMP) and protein kinase A (PKA), classical examples of spatially compartmentalized signaling molecules, are critical axon determinants that regulate neuronal polarity and axon formation, yet little is known about micro-compartmentalization of cAMP and PKA signaling and its role in developing neurons. Here, we revealed that cAMP forms a gradient in developing hippocampal neurons, with higher cAMP levels in more distal regions of the axon compared to other regions of the cell. Interestingly, this cAMP gradient changed according to the developmental stage and depended on proper anchoring of PKA by A-kinase anchoring proteins (AKAPs). Disrupting PKA anchoring to AKAPs increased the cAMP gradient in early-stage neurons and led to enhanced axon elongation. Our results provide new evidence for a local negative-feedback loop, assembled by AKAPs, for the precise control of a growth-stage-dependent cAMP gradient to ensure proper axon growth.


Subject(s)
A Kinase Anchor Proteins/metabolism , Cyclic AMP/metabolism , Feedback, Physiological , Hippocampus/cytology , Neurons/metabolism , Animals , Cells, Cultured , Rats , Rats, Sprague-Dawley
17.
Biochemistry ; 56(39): 5210-5213, 2017 10 03.
Article in English | MEDLINE | ID: mdl-28718621

ABSTRACT

All cellular behaviors arise through the coordinated actions of numerous intracellular biochemical pathways. Over the past 20 years, efforts to probe intracellular biochemical processes have undergone a fundamental transformation brought about by advances in fluorescence imaging, such as the development of genetically encoded fluorescent reporters and new imaging technologies; the impact of these approaches on our understanding of the molecular underpinnings of biological function cannot be understated. In particular, the ability to obtain information on the spatiotemporal regulation of biochemical processes unfolding in real time in the native context of a living cell has crystallized the view, long a matter of speculation, that cells achieve specific biological outcomes through the imposition of spatial control over the distribution of various biomolecules, and their associated biochemical activities, within the cellular environment. Indeed, the compartmentalization of biochemical activities by cells is now known to be pervasive and to span a multitude of spatial scales, from the length of a cell to just a few enzymes. In this Perspective, part of this special issue on "Seeing into cells", we highlight several recent imaging studies that provide detailed insights into not just where molecules are but where molecules are active within cells, offering a glimpse into the emerging view of biochemical activity architecture as a complement to the physical architecture of a cell.


Subject(s)
Cells/metabolism , Molecular Imaging/methods , Animals
18.
J Biol Chem ; 290(11): 6681-8, 2015 Mar 13.
Article in English | MEDLINE | ID: mdl-25605723

ABSTRACT

The growing use of fluorescent biosensors to directly probe the spatiotemporal dynamics of biochemical processes in living cells has revolutionized the study of intracellular signaling. In this review, we summarize recent developments in the use of biosensors to illuminate the molecular details of G-protein-coupled receptor (GPCR) signaling pathways, which have long served as the model for our understanding of signal transduction, while also offering our perspectives on the future of this exciting field. Specifically, we highlight several ways in which biosensor-based single-cell analyses are being used to unravel many of the enduring mysteries that surround these diverse signaling pathways.


Subject(s)
Biosensing Techniques/methods , GTP-Binding Proteins/metabolism , Signal Transduction , Single-Cell Analysis/methods , Animals , Biosensing Techniques/instrumentation , Equipment Design , Fluorescence Resonance Energy Transfer/instrumentation , Fluorescence Resonance Energy Transfer/methods , Humans , Receptors, G-Protein-Coupled/metabolism , Single-Cell Analysis/instrumentation
19.
J Cell Sci ; 127(Pt 6): 1151-60, 2014 Mar 15.
Article in English | MEDLINE | ID: mdl-24634506

ABSTRACT

In this Commentary, we discuss two sets of genetically encoded molecular tools that have significantly enhanced our ability to observe and manipulate complex biochemical processes in their native context and that have been essential in deepening our molecular understanding of how intracellular signaling networks function. In particular, genetically encoded biosensors are widely used to directly visualize signaling events in living cells, and we highlight several examples of basic biosensor designs that have enabled researchers to capture the spatial and temporal dynamics of numerous signaling molecules, including second messengers and signaling enzymes, with remarkable detail. Similarly, we discuss a number of genetically encoded biochemical perturbation techniques that are being used to manipulate the activity of various signaling molecules with far greater spatial and temporal selectivity than can be achieved using standard pharmacological or genetic techniques, focusing specifically on examples of chemically driven and light-inducible perturbation strategies. We then describe recent efforts to combine these diverse and powerful molecular tools into a unified platform that can be used to elucidate the molecular details of biological processes that may potentially extend well beyond the realm of signal transduction.


Subject(s)
Fluorescent Dyes/metabolism , Green Fluorescent Proteins/genetics , Signal Transduction , Animals , Biosensing Techniques , Fluorescence Resonance Energy Transfer , Fluorescent Dyes/chemistry , Green Fluorescent Proteins/biosynthesis , Humans , Single-Cell Analysis
20.
RSC Chem Biol ; 5(4): 312-320, 2024 Apr 03.
Article in English | MEDLINE | ID: mdl-38576721

ABSTRACT

Genetically encoded Ras biosensors have been instrumental in illuminating the spatiotemporal dynamics of Ras activity since the beginning of the imaging revolution of the early 21st century. In general, these sensors employ Ras sensing units coupled with fluorescent proteins. These biosensors have not only helped elucidate Ras signalling dynamics at the plasma membrane but also revealed novel roles for Ras signalling within subcellular compartments such as the Golgi apparatus. In this review, we discuss the different classes of biosensors used to measure Ras activity and discuss their importance in uncovering new roles for Ras activity in cellular signalling and behavior.

SELECTION OF CITATIONS
SEARCH DETAIL