Curated Optogenetic Publication Database

Search precisely and efficiently by using the advantage of the hand-assigned publication tags that allow you to search for papers involving a specific trait, e.g. a particular optogenetic switch or a host organism.

Showing 1 - 4 of 4 results
1.

Mapping the Cellular Distribution of an Optogenetic Protein Using a Light-Stimulation Grid Mapping the Cellular Distribution of an Optogenetic Protein Using a Light-Stimulation Grid.

blue bPAC (BlaC) HC-1
J Vis Exp, 26 Jan 2024 DOI: 10.3791/65471 Link to full text
Abstract: Our goal was to accurately track the cellular distribution of an optogenetic protein and evaluate its functionality within a specific cytoplasmic location. To achieve this, we co-transfected cells with nuclear-targeted cAMP sensors and our laboratory-developed optogenetic protein, bacterial photoactivatable adenylyl cyclase-nanoluciferase (bPAC-nLuc). bPAC-nLuc, when stimulated with 445 nm light or luciferase substrates, generates adenosine 3',5'-cyclic monophosphate (cAMP). We employed a solid-state laser illuminator connected to a point scanning system that allowed us to create a grid/matrix pattern of small illuminated spots (~1 µm2) throughout the cytoplasm of HC-1 cells. By doing so, we were able to effectively track the distribution of nuclear-targeted bPAC-nLuc and generate a comprehensive cAMP response map. This map accurately represented the cellular distribution of bPAC-nLuc, and its response to light stimulation varied according to the amount of protein in the illuminated spot. This innovative approach contributes to the expanding toolkit of techniques available for investigating cellular optogenetic proteins. The ability to map its distribution and response with high precision has far-reaching potential and could advance various fields of research.
2.

Soluble cyclase-mediated nuclear cAMP synthesis is sufficient for cell proliferation.

blue bPAC (BlaC) PCCL3 Signaling cascade control Immediate control of second messengers
bioRxiv, 19 May 2022 DOI: 10.1101/2022.05.18.492464 Link to full text
Abstract: cAMP is a key player in many physiological processes. Classically considered to originate solely from the plasma membrane, this view was recently challenged by observations showing that GPCRs can sustain cAMP signaling from intracellular compartments associated with nuclear PKA translocation and activation of transcriptional events. In this report we show that neither PKA translocation nor cAMP diffusion, but rather nuclear sAC activation represents the only source of nuclear cAMP accumulation, PKA activation, and CREB phosphorylation. Both pharmacological and genetic sAC inhibition, that did not affect the cytosolic cAMP levels, completed blunted nuclear cAMP accumulation, PKA activation and proliferation, while an increase in sAC nuclear expression significantly enhanced cell proliferation. Moreover, utilizing novel compartment-specific optogenetic actuators we showed that light-dependent nuclear cAMP synthesis can stimulate PKA, CREB and trigger cell proliferation. Thus, our results show that sAC-mediated nuclear accumulation is not only necessary but sufficient and rate-limiting for cAMP-dependent proliferation.
3.

Dual Activation of cAMP Production Through Photostimulation or Chemical Stimulation.

blue bPAC (BlaC) HC-1
Methods Mol Biol, 11 Jul 2020 DOI: 10.1007/978-1-0716-0755-8_14 Link to full text
Abstract: cAMP is a crucial mediator of multiple cell signaling pathways. This cyclic nucleotide requires strict spatiotemporal control for effective function. Light-activated proteins have become a powerful tool to study signaling kinetics due to having quick on/off rates and minimal off-target effects. The photoactivated adenylyl cyclase from Beggiatoa (bPAC) produces cAMP rapidly upon stimulation with blue light. However, light delivery is not always feasible, especially in vivo. Hence, we created a luminescence-activated cyclase by fusing bPAC with nanoluciferase (nLuc) to allow chemical activation of cAMP activity. This dual-activated adenylyl cyclase can be stimulated using short bursts of light or long-term chemical activation with furimazine and other related luciferins. Together these can be used to mimic transient, chronic, and oscillating patterns of cAMP signaling. Moreover, when coupled to compartment-specific targeting domains, these reagents provide a new powerful tool for cAMP spatiotemporal dynamic studies. Here, we describe detailed methods for working with bPAC-nLuc in mammalian cells, stimulating cAMP production with light and luciferins, and measuring total cAMP accumulation.
4.

Luminescence-activated nucleotide cyclase regulates spatial and temporal cAMP synthesis.

blue bPAC (BlaC) HC-1 HEK293 PCCL3 Cell cycle control Immediate control of second messengers
J Biol Chem, 17 Dec 2018 DOI: 10.1074/jbc.ac118.004905 Link to full text
Abstract: cAMP is a ubiquitous second messenger that regulates cellular proliferation, differentiation, attachment, migration, and several other processes. It has become increasingly evident that tight regulation of cAMP accumulation and localization confers divergent yet specific signaling to downstream pathways. Currently, few tools are available that have sufficient spatial and temporal resolution to study location-biased cAMP signaling. Here, we introduce a new fusion protein consisting of a light-activated adenylyl cyclase (bPAC) and luciferase (nLuc). This construct allows dual activation of cAMP production through temporally precise photostimulation or chronic chemical stimulation that can be fined-tuned to mimic physiological levels and duration of cAMP synthesis to trigger downstream events. By targeting this construct to different compartments, we show that cAMP produced in the cytosol and nucleus stimulates proliferation in thyroid cells. The bPAC-nLuc fusion construct adds a new reagent to the available toolkit to study cAMP-regulated processes in living cells.
Submit a new publication to our database