Summary

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

Published: January 26, 2024
doi:

Summary

This protocol involves transfecting cAMP sensors and bPAC-nLuc, an optogenetic protein, to accurately track its cellular distribution and response to light stimulation. The innovative approach of creating a cAMP response map using a point scanning system holds the potential for advancing research with optogenetic proteins in different fields.

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.

Introduction

Optogenetics, born as a tool that revolutionized neurosciences, is now a growing research field and a rising technology routinely used by many laboratories worldwide and across various research areas in biology. We developed bPAC-nLuc, a versatile optogenetic protein, by fusing a light-sensitive adenylyl cyclase (AC) from Beggiatoa sp. (bacterial photoactivatable adenylyl cyclase; bPAC) to nanoluciferase (nLuc)1,2,3. When stimulated with blue light, bPAC produces the second messenger 3',5'-cyclic adenosine monophosphate (cAMP). nLuc is a recently developed small luciferase that, in the presence of one of its substrates, can generate bioluminescence and activate cAMP production4. Thus, this optogenetic protein can be activated transiently by using brief light pulses or steadily with Furimazine or other luciferase substrates, allowing us to mimic different cAMP signaling patterns and assess cellular responses (activation of transcription factors, gene expression, cell proliferation, migration, etc.). Recent advances in 2nd messenger signaling have emphasized the significance of events occurring in very restricted cytosolic regions (e.g., endosomal cAMP production for cAMP response element-binding protein (CREB) phosphorylation or Ca2+ microdomains for nuclear factor of activated T-cells (NFAT) translocation to the nucleus)5,6. Therefore, developing consistent and systematic strategies to evaluate, mimic, and block signaling from these compartments in live cells is important. To show the ability of bPAC-nLuc to be specifically activated in different cell compartments, we co-transfected a hepatoma-derived cell line (HC-1) with the nuclear-targeted bPAC-nLuc and H208, a Förster resonance energy transfer (FRET) cAMP sensor (NLS-bPAC-nLuc; NLS-H208). HC-1 cells that derive from the HTC line are devoid of assayable AC activity, which results in very low basal cAMP levels, making it ideal to measure putative cAMP production while taking advantage of the full dynamic range of FRET sensors7,8. Using a solid-state laser (445 nm, LDI-7, 89 North) connected to a point scanning system (UGA-42 Geo, Rapp OptoElectronic), we describe a protocol to systematically stimulate very small circular areas or spots (~1 µm2) within individual cells. The point scanning system was connected to one of the backports of a two-deck microscope, which allowed us to stimulate cells and perform FRET measurements simultaneously via an independent lightpath. We present a method in this protocol where the SysCon Geo software, supplied with the stimulation system by Rapp OptoElectronic, is employed to perform a comprehensive scan of the cytoplasm of cells. The approach involves generating a cAMP response map by setting up a sequence of illuminations that stimulate cells in a grid pattern (Figure 1).

Protocol

1. HC-1 cell culture and preparation for imaging Maintain HC-1 cells in DMEM supplemented with 10% fetal bovine serum (FBS), penicillin (100 IU/L), streptomycin (100 mg/L), and L-glutamine in 10 cm dishes and incubated at 37 °C, 5% CO2, in 95% humidified air. Passage the cells every 2-4 days once cells are ~90% confluent using 1:5 or 1:10 dilutions. Seed cells for transfection on glass coverslips 2 days before the experiment. Working under sterile condit…

Representative Results

The results presented in Figure 1 show that only stimulations directed to the cell nucleus were able to generate measurable cAMP elevations. This confirms that NLS-bPAC-nLuc is expressed exclusively in the nuclear compartment of HC-1 cells. It is possible to precisely stimulate an optogenetic protein using this grid/matrix pattern to map its intracellular distribution. Additionally, the higher cAMP elevations towards the nuclear center reflect the higher mass…

Discussion

The objective of this study was to precisely monitor the intracellular distribution of an optogenetic protein and assess its performance within a particular cytoplasmic compartment. We also showed the precise stimulation capabilities of a point scanning system on cells expressing an optogenetic protein. To achieve this, we employed a nuclear-targeted bPAC-nLuc with high expression levels but a very confined distribution limited to the nucleus. The results showed that stimulation spots separated by only ~1 µm can eit…

Disclosures

The authors have nothing to disclose.

Acknowledgements

Funding was provided by the National Institutes of Health (NIH) grants R01 GM099775 and GM130612 to D.L.A.

Materials

13 W Amber compact fluorescence bulb – Low Blue Lights Photonic Developments 
3-Isobutyl-1-methylxanthine (IBMX) Sigma I7018
6-line multi-LED Lumencor Spectra X Lumencor  6-line multi-LED light engine 
Corning – DMEM  Thermo Fischer MT10013CMEA
Corning – Regular fetal bovine serum Thermo Fischer MT35011CV
Cover glasses: circles Thermo Fischer 12545102P
GBX-2 dark red safelight filter 5.5"  Kodak 1416827 Red safelight lamp  
Hanks' balanced salt solution (HBSS) 10x Thermo Fischer 14185052 Diluted to 1x, adjusted pH
LDI-7 89 North
L-Glutamine Thermo Fischer BW17605E
Lipofectamine 3000  Thermo Fischer L3000001 Transfection kit 
Olympus IX83 motorized two-deck microscope  Olympus Motorized two-deck microscope 
Opti-MEM, no phenol red Thermo Fischer 11058021
ORCA-fusion digital CMOS camera  Hamamatsu C14440-20UP
Penicillin-streptomycin (10,000 U/mL) Thermo Fischer 15140122
Phosphate buffered solution (1x) Lonza 17516F
Prior emission filter wheel and filter sets Prior Scientific, Inc. Emission filter wheel 
Prior Proscan XY stage Prior Scientific, Inc. XY stage 
Slidebook 6 Intelligent Imaging Innovations Digital microscopy software
SysCon software SysCon Software Software provided by the stimulation system 
UGA-42 Geo  Rapp OptoElectronic
UPlanSApo 100x Olympus 100x/1.4 NA oil objective (∞/0.17/FN26.5)
ZT458rdc dichroic  Chroma Technology Corp BS, Wavelength (CWL): 498 nm

References

  1. Naim, N., et al. Luminescence-activated nucleotide cyclase regulates spatial and temporal cAMP synthesis. The Journal of Biological Chemistry. 294 (4), 1095-1103 (2019).
  2. Naim, N., Reece, J. M., Zhang, X., Altschuler, D. L. Dual activation of cAMP production through photostimulation or chemical stimulation. Methods in Molecular Biology (Clifton, N.J). 2173, 201-216 (2020).
  3. Stierl, M., et al. Light modulation of cellular cAMP by a small bacterial photoactivated adenylyl cyclase, bPAC, of the soil bacterium Beggiatoa. The Journal of Biological Chemistry. 286 (2), 1181-1188 (2011).
  4. Hall, M. P., et al. Engineered luciferase reporter from a deep sea shrimp utilizing a novel imidazopyrazinone substrate. ACS Chemical Biology. 7 (11), 1848-1857 (2012).
  5. Liccardo, F., Luini, A., Di Martino, R. Endomembrane-based signaling by GPCRs and G-proteins. Cells. 11 (3), 528 (2022).
  6. Groschner, K., Graier, W. F., Romanin, C. . Store-operated ca(2)+ entry (SOCE) pathways. , (2017).
  7. Ross, E. M., Howlett, A. C., Ferguson, K. M., Gilman, A. G. Reconstitution of hormone-sensitive adenylate cyclase activity with resolved components of the enzyme. The Journal of Biological Chemistry. 253 (18), 6401-6412 (1978).
  8. Insel, P. A., Maguire, M. E., Gilman, A. G., Bourne, H. R., Coffino, P., Melmon, K. L. Beta adrenergic receptors and adenylate cyclase: products of separate genes. Molecular Pharmacology. 12, 1062-1069 (1976).
  9. Pizzoni, A., Zhang, X., Naim, N., Altschuler, D. L. Soluble cyclase-mediated nuclear cAMP synthesis is sufficient for cell proliferation. Proceedings of the National Academy of Sciences of the United States of America. 120 (4), e2208749120 (2023).
  10. Keravis, T., Lugnier, C. Cyclic nucleotide phosphodiesterase (PDE) isozymes as targets of the intracellular signalling network: benefits of PDE inhibitors in various diseases and perspectives for future therapeutic developments. British Journal of Pharmacology. 165 (5), 1288-1305 (2012).
This article has been published
Video Coming Soon
Keep me updated:

.

Cite This Article
Pizzoni, A., Naim, N., Zhang, X., Altschuler, D. L. 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. J. Vis. Exp. (203), e65471, doi:10.3791/65471 (2024).

View Video