Optogenetic control of small GTPases reveals RhoA mediates intracellular calcium signaling

Autor: Hironori Inaba, Takao Nakata, Qianqian Miao
Jazyk: angličtina
Rok vydání: 2021
Předmět:
0301 basic medicine
rac1 GTP-Binding Protein
RHOA
Light
IP3
inositol-triphosphate

CDC42
GTPase
Biosensing Techniques
Biochemistry
IP3R
IP3 receptor

Calcium in biology
Madin Darby Canine Kidney Cells
Phosphoinositide Phospholipase C
Small GTPase
phospholipase C
cdc42 GTP-Binding Protein
Calcium signaling
LOV2
light-oxygen-voltage-sensing domain 2

PI(4
5)P2
phosphatidylinositol 4
5-bisphosphate

biology
Chemistry
Cell Differentiation
Cell biology
Organ Specificity
Guanine nucleotide exchange factor
Ras protein
signal transduction
Research Article
Light Signal Transduction
chemistry.chemical_element
inositol 1
4
5-triphosphate (IP3)

Calcium
iLID
improved light-inducible dimer system

ER
endoplasmic reticulum

GEF
guanine nucleotide exchange factor

03 medical and health sciences
NFAT transcription factor
PLC
phospholipase C

Dogs
Animals
Humans
Calcium Signaling
optogenetics
Molecular Biology
Cell Proliferation
calcium
030102 biochemistry & molecular biology
TGN
trans-Golgi network

TRP channel
transient receptor potential channel

PI(4)P
phosphatidylinositol 4-phosphate

Cell Biology
RA domain
Ras association domain

030104 developmental biology
HEK293 Cells
rap GTP-Binding Proteins
Gene Expression Regulation
Rho (Rho GTPase)
GAP
GTPase activating protein

RBD
Ras/Rho binding domain

biology.protein
ras Proteins
ral GTP-Binding Proteins
rhoA GTP-Binding Protein
DAG
diacylglycerol

MS
mechanosensitive

HeLa Cells
Zdroj: The Journal of Biological Chemistry
ISSN: 1083-351X
0021-9258
Popis: Rho/Ras family small GTPases are known to regulate numerous cellular processes, including cytoskeletal reorganization, cell proliferation, and cell differentiation. These processes are also controlled by Ca2+, and consequently, cross talk between these signals is considered likely. However, systematic quantitative evaluation has not yet been reported. To fill this gap, we constructed optogenetic tools to control the activity of small GTPases (RhoA, Rac1, Cdc42, Ras, Rap, and Ral) using an improved light-inducible dimer system (iLID). We characterized these optogenetic tools with genetically encoded red fluorescence intensity-based small GTPase biosensors and confirmed these optogenetic tools' specificities. Using these optogenetic tools, we investigated calcium mobilization immediately after small GTPase activation. Unexpectedly, we found that a transient intracellular calcium elevation was specifically induced by RhoA activation in RPE1 and HeLa cells. RhoA activation also induced transient intracellular calcium elevation in MDCK and HEK293T cells, suggesting that generally RhoA induces calcium signaling. Interestingly, the molecular mechanisms linking RhoA activation to calcium increases were shown to be different among the different cell types: In RPE1 and HeLa cells, RhoA activated phospholipase C epsilon (PLCe) at the plasma membrane, which in turn induced Ca2+ release from the endoplasmic reticulum (ER). The RhoA-PLCe axis induced calcium-dependent nuclear factor of activated T cells nuclear translocation, suggesting that it does activate intracellular calcium signaling. Conversely, in MDCK and HEK293T cells, RhoA-ROCK-myosin II axis induced the calcium transients. These data suggest universal coordination of RhoA and calcium signaling in cellular processes, such as cellular contraction and gene expression.
Databáze: OpenAIRE