Abstract

For the tissues and organs of our bodies to work properly, the cells within them need to communicate with each other. One important part of cellular communication is the movement of signals – usually small molecules or ions – directly from one cell to another. This happens via structures called gap junctions, a type of sealed ‘channel’ that connects two cells. Gap junctions are found throughout the body, but investigating their precise roles in health and disease has been difficult. This is due to problems with the tools available to detect and monitor gap junctions. Some are simply harmful to cells, while others cannot be restricted to specific cell populations within a tissue. This lack of specificity makes it difficult to study gap junctions in the brain, where it is important to understand the connectivity patterns between distinct types of nerve cells. Wu et al. wanted to develop a new, non-harmful method to track gap junctions in distinct groups of cells within living tissues. To do this, Wu et al. devised PARIS, a two-part, genetically encoded system. The first part comprises a light-sensitive molecular ‘pump’, which can only be turned on by shining a laser onto the cell of interest. When the pump is active, it transports hydrogen ions out of the cell. The second part of the system is a fluorescent sensor, present inside ‘receiving’ cells, which responds to the outcoming hydrogen ions (small enough to pass through gap junctions). If an illuminated ‘signaling’ cell is connected via gap junctions to cells containing the fluorescent sensor, they will light up within seconds, but other cells not connected through gap junctions will not. The researchers first tested PARIS in cultured human and rat cells that had been genetically engineered to produce both components of the system. The experiments confirmed that PARIS could both detect networks of gap junctions in healthy cells and reveal when these networks had been disrupted, for instance by drugs or genetic mutations. Experiments using fruit flies demonstrated that PARIS was stable in living tissue and could also map the gap junctions connecting specific groups of nerve cells. PARIS is a valuable addition to the toolbox available to study cell communication. In the future, it could help increase our understanding of diseases characterized by defective gap junctions, such as seizures, cardiac irregularities, and even some cancers.

Highlights

  • Gap junctions are intercellular channels that are expressed in virtually all tissue types in both vertebrates and invertebrates

  • We tested several pairs of optical actuators/receivers based on generating/detecting small molecules that can readily diffuse across gap junctions, such as cGMP, Ca2+ and proton (H+)

  • We describe the development of PARIS, a new all-optical approach for detecting gap junction coupling (GJC) in specific cells

Read more

Summary

Introduction

Gap junctions are intercellular channels that are expressed in virtually all tissue types in both vertebrates and invertebrates. Hybrid approaches have been developed in which genetically encoded hydrolytic enzymes or promiscuous transporters are used to introduce small molecule substrates or peptides tagged with a fluorescent label in specific cells (Qiao and Sanes, 2015; Tian et al, 2012) These methods might provide cell-type-specific investigation of GJC, the requirement of exogenous substrate and the background generated by endogenous enzymes or transporters still make them difficult to apply in vivo. To overcome these limitations, we developed an all-optical approach named PARIS (pairing actuators and receivers to optically isolate gap junctions) in which we express an optically controlled actuator in one cell, to generate an electrochemical gradient of specific molecules between two connected cells, and a fluorescent receiver in the adjacent cell, to detect the movement of the molecules across the gap junctions. GJC between the actuator cell (i.e. expressing actuators) and the receiver cell (i.e. expressing receivers) is detected by a fluorescence increase in the receiver following the optical activation of the actuator (Figure 1A)

Results
Discussion
Materials and methods
Funding Funder
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call