1.Investigation of Pain Mechanisms by Calcium Imaging Approaches.
Michael ANDERSON ; Qin ZHENG ; Xinzhong DONG
Neuroscience Bulletin 2018;34(1):194-199
Due to the complex circuitry and plethora of cell types involved in somatosensation, it is becoming increasingly important to be able to observe cellular activity at the population level. In addition, since cells rely on an intricate variety of extracellular factors, it is important to strive to maintain the physiological environment. Many electrophysiological techniques require the implementation of artificially-produced physiological environments and it can be difficult to assess the activity of many cells simultaneously. Moreover, imaging Ca transients using Ca-sensitive dyes often requires in vitro preparations or in vivo injections, which can lead to variable expression levels. With the development of more sensitive genetically-encoded Ca indicators (GECIs) it is now possible to observe changes in Ca transients in large populations of cells at the same time. Recently, groups have used a GECI called GCaMP to address fundamental questions in somatosensation. Researchers can now induce GCaMP expression in the mouse genome using viral or gene knock-in approaches and observe the activity of populations of cells in the pain pathway such as dorsal root ganglia (DRG), spinal neurons, or glia. This approach can be used in vivo and thus maintains the organism's biological integrity. The implementation of GCaMP imaging has led to many advances in our understanding of somatosensation. Here, we review the current findings in pain research using GCaMP imaging as well as discussing potential methodological considerations.
Afferent Pathways
;
physiology
;
Animals
;
Calcium
;
metabolism
;
Calcium Signaling
;
drug effects
;
genetics
;
Ganglia, Spinal
;
metabolism
;
Humans
;
Pain
;
metabolism
;
pathology
2.Alteration of Expression of Ca(2+) Signaling Proteins and Adaptation of Ca(2+) Signaling in SERCA2(+/-) Mouse Parotid Acini.
Jong Hoon CHOI ; Hae JO ; Jeong Hee HONG ; Syng Ill LEE ; Dong Min SHIN
Yonsei Medical Journal 2008;49(2):311-321
PURPOSE: The sarco/endoplasmic reticulum Ca(2+)-ATPase (SERCA), encoded by ATP2A2, is an essential component for G-protein coupled receptor (GPCR)-dependent Ca(2+) signaling. However, whether the changes in Ca(2+) signaling and Ca(2+) signaling proteins in parotid acinar cells are affected by a partial loss of SERCA2 are not known. MATERIALS AND METHODS: In SERCA2(+/-) mouse parotid gland acinar cells, Ca(2+) signaling, expression levels of Ca(2+) signaling proteins, and amylase secretion were investigated. RESULTS: SERCA2(+/-) mice showed decreased SERCA2 expression and an upregulation of the plasma membrane Ca(2+) ATPase. A partial loss of SERCA2 changed the expression level of 1, 4, 5-tris-inositolphosphate receptors (IP(3)Rs), but the localization and activities of IP3Rs were not altered. In SERCA2(+/-) mice, muscarinic stimulation resulted in greater amylase release, and the expression of synaptotagmin was increased compared to wild type mice. CONCLUSION: These results suggest that a partial loss of SERCA2 affects the expression and activity of Ca(2+) signaling proteins in the parotid gland acini, however, overall Ca(2+) signaling is unchanged.
Amylases/metabolism
;
Animals
;
Blotting, Western
;
Calcium/metabolism
;
Calcium Signaling/drug effects/genetics/*physiology
;
Carbachol/pharmacology
;
Immunohistochemistry
;
Inositol 1,4,5-Trisphosphate Receptors/metabolism
;
Mice
;
Mice, Knockout
;
Parotid Gland/*metabolism
;
Sarcoplasmic Reticulum Calcium-Transporting ATPases/genetics/*metabolism
;
Signal Transduction/drug effects/genetics/physiology
3.The mechanism of calcium signal regulate preadipocyte differentiation and lipid accumulation in mice.
Li WANG ; Chao SUN ; Jingquan KANG
Chinese Journal of Biotechnology 2009;25(5):739-744
We stimulated preadipocyte of mice with calcium acetate, p38 mitogen-activated protein kinase (p38 MAPK) inhibitor SB203580, the paralysors and excitomotors of calcium channel. Then we detected expression level of preadipocyte differentiation's marker genes and calcium signal related acceptor genes by real-time PCR, and determined intracellular free Ca2+ concentration ([Ca2+]i]) with Fura-2/AM, intracellular lipid accumulation by oil red O staining. Our aim was to investigate the potential mechanism between calcium signal and preadipocyte differentiation. The results indicated that the paralysors and excitomotors of calcium channel changed the expression level of lipoprotein lipase (LPL), peroxisome proliferators-activated receptor gamma (PPARgamma), fatty acid synthetase (FAS), and the lipid accumulation, markedly. Compared with exocellular Ca2+'s decrease, inhibited intracellular Ca2+'s liberation can promoted preadipocyte differentiation (P < 0.01), and compared with intracellular Ca2+'s increase, promoted exocellular Ca2+'s ingest inhibited preadipocyte differentiation (P < 0.01). SB203580 degraded [Ca2+]i, promoted differentiation marker genes' expression and lipid accumulation in preadipocyte (P < 0.01). But calcium signal didn't have effects to vitamin D receptor (VDR) and extracellular Ca2+-sensing receptor (CaSR)'s expression. It indicated that calcium signal may effect preadipocyte different and lipid accumulation by p38 MAPK pathway.
Adipocytes
;
cytology
;
Animals
;
Calcium
;
metabolism
;
Calcium Signaling
;
drug effects
;
Cell Differentiation
;
physiology
;
Cells, Cultured
;
Imidazoles
;
pharmacology
;
Lipids
;
biosynthesis
;
Mice
;
Pyridines
;
pharmacology
;
p38 Mitogen-Activated Protein Kinases
;
antagonists & inhibitors
4.Autocrine Extracellular Signal-regulated Kinase Activation in Normal Human Keratinocytes is not Interrupted by Calcium Triggering and is Involved in the Control of Cell Cycle at the Early Stage of Calciuminduced Differentiation.
Geon Tae PARK ; Hyo Youn KIM ; Eun Kyoung KIM ; Jun Mo YANG
Journal of Korean Medical Science 2007;22(2):290-297
Normal human epidermal keratinocytes (NHEK) respond to the autocrine activated extracellular signal-regulated kinase (ERK) signaling pathway, which contributes to the survival of keratinocytes. However, during the condition of calcium-induced differentiation, how the autocrine ERK signaling is regulated and affected is poorly understood. The purpose of this study was to understand and to obtain clues to the possible function of the autocrine ERK activation during the calcium-induced differentiation of NHEK. We demonstrated that the autocrine activated ERK was not interrupted by calcium triggering and that it was sustained for at least one day after changing the medium. We also found that the autocrine ERK activation was associated with the expression of cyclin D1 and the cell cycle regulation at the early stage of calcium triggering by treating the cells with the mitogen-activated protein kinase inhibitor PD98059. However, the PD98059 treatment did not have a significant influence on the expression of involucrin and loricrin. In addition, we demonstrated that autocrine ERK activation was associated with protein kinase C and p38MAPK signaling. We suggest that the activation of autocrine ERK is not interrupted by calcium triggering and it might participate in cell growth during the early stage of calcium-induced differentiation in NHEK.
Keratinocytes/*cytology/drug effects/*physiology
;
Humans
;
Extracellular Signal-Regulated MAP Kinases/*metabolism
;
Enzyme Activation/drug effects
;
Dose-Response Relationship, Drug
;
Cells, Cultured
;
Cell Differentiation/drug effects
;
Cell Cycle/drug effects/*physiology
;
Calcium Signaling/drug effects/*physiology
;
Calcium/*administration & dosage
;
Autocrine Communication/drug effects/*physiology
5.Inotropic effects of MCI-154 on rat cardiac myocytes.
Huan-Zhen CHEN ; Xiang-Li CUI ; Hua-Chen ZHAO ; Lu-Ying ZHAO ; Ji-Yuan LU ; Bo-Wei WU
Acta Physiologica Sinica 2004;56(3):301-305
Calcium sensitizers exert positive inotropic effects without increasing intracellular Ca(2+). Thus, they avoid the undesired effects of Ca(2+) overload such as arrhythmias and cell injury, but most of them may impair myocyte relaxation. However, MCI-154, also a calcium sensitizer, has no impairment to cardiomyocyte relaxation. To clarify the underlying mechanisms, we examined the effects of MCI-154 on Ca(2+) transient and cell contraction using ion imaging system, and its influence on L-type Ca(2+) current and Na(+)/ Ca(2+) exchange current with patch clamp technique in rat ventricular myocytes as well. The results showed that: (1) MCI-154 (1-100 micromol/L) had no effect on L-type Ca(2+) current; (2) MCI-154 concentration-dependently increased cell shortening from 5.00+/-1.6 microm of control to 6.2+/-1.6 microm at 1 micromol/L, 8.7+/-1.6 microm at 10 micromol/L and 14.0+/-1.4 microm at 100 micromol/L, respectively, with a slight increase in Ca(2+) transient amplitude and an abbreviation of Ca(2+) transient restore kinetics assessed by time to 50% restore (TR(50)) and time to 90% restore (TR(90)); (3) MCI-154 dose-dependently increased the electrogenic Na(+)/ Ca(2+) exchange current both in the inward and the outward directions in rat ventricular myocytes. These results indicate that MCI-154 exerted a positive inotropic action without impairing myocyte relaxation. The stimulation of inward Na(+)/ Ca(2+) exchange current may accelerate the Ca(2+) efflux, leading to abbreviations of TR(50) and TR(90) in rat myocytes. The findings suggest that the improvement by MCI-154 of myocyte relaxation is attributed to the forward mode of Na(+)/ Ca(2+) exchange.
Animals
;
Calcium
;
physiology
;
Calcium Channels, L-Type
;
drug effects
;
Calcium Signaling
;
drug effects
;
Cardiotonic Agents
;
pharmacology
;
Cell Separation
;
Cells, Cultured
;
Dose-Response Relationship, Drug
;
Heart Ventricles
;
cytology
;
Myocardial Contraction
;
drug effects
;
Myocytes, Cardiac
;
cytology
;
metabolism
;
Patch-Clamp Techniques
;
Pyridazines
;
pharmacology
;
Rats
;
Rats, Wistar
;
Sodium-Calcium Exchanger
;
drug effects
;
physiology
6.Atorvastatin induces autophagy of mesenchymal stem cells under hypoxia and serum deprivation conditions by activating the mitogen-activated protein kinase/extracellular signal-regulated kinase pathway.
Na LI ; Qian ZHANG ; Haiyan QIAN ; Chen JIN ; Yuejin YANG ; Runlin GAO
Chinese Medical Journal 2014;127(6):1046-1051
BACKGROUNDThe survival ratio of implanted mesenchymal stem cells (MSCs) in the infarcted myocardium is low. Autophagy is a complex "self-eating" process and can be utilized for cell survival. We have found that atorvastatin (ATV) can effectively activate autophagy to enhance MSCs survival during hypoxia and serum deprivation (H/SD). The mitogen-activated protein kinase/extracellular signal-regulated kinase (MEK/ERK) pathway is a non-canonical autophagy pathway. We hypothesized that the MEK/ERK pathway mediated ATV-induced autophagy of MSCs under H/SD.
METHODSMSCs were pretreated with ATV (0.01-10 µmol/L) under H/SD for three hours. For inhibitor studies, the cells were pre-incubated with the MEK1/2 inhibitor U0126. Cell autophagy was assessed by acidic vesicular organelles (AVO)-positive cells using flow cytometry, autophagy related protein using Western blotting and autophagosome using transmission electron microscopy.
RESULTSAutophagy was elevated in the H/SD group compared with the normal group. ATV further enhanced the autophagic activity as well as the phosphorylation of ERK1/2 evidenced by more AVO-positive cells ((8.63 ± 0.63)% vs. (5.77 ± 0.44)%, P < 0.05), higher LC3-II/LC3-I ratio (4.36 ± 0.31 vs. 2.52 ± 0.18, P < 0.05) and more autophagosomes. And treatment with U0126 downregulated the phosphorylation of ERK1/2 and attenuated ATV-induced autophagy.
CONCLUSIONThe MEK/ERK pathway participates in ATV-induced autophagy in MSCs under H/SD, and modulation of the pathway could be a novel strategy to improve MSCs survival.
Animals ; Atorvastatin Calcium ; Autophagy ; drug effects ; Cell Hypoxia ; physiology ; Cells, Cultured ; Flow Cytometry ; Heptanoic Acids ; pharmacology ; MAP Kinase Signaling System ; drug effects ; Male ; Mesenchymal Stromal Cells ; cytology ; drug effects ; ultrastructure ; Microscopy, Electron, Transmission ; Pyrroles ; pharmacology ; Rats
7.ERp44 C160S/C212S mutants regulate IP3R1 channel activity.
Congyan PAN ; Ji ZHENG ; Yanyun WU ; Yingxiao CHEN ; Likun WANG ; Zhansong ZHOU ; Wenxuan YIN ; Guangju JI
Protein & Cell 2011;2(12):990-996
Previous studies have indicated that ERp44 inhibits inositol 1,4,5-trisphosphate (IP(3))-induced Ca(2+) release (IICR) via IP(3)R(1), but the mechanism remains largely unexplored. Using extracellular ATP to induce intracellular calcium transient as an IICR model, Ca(2+) image, pull down assay, and Western blotting experiments were carried out in the present study. We found that extracellular ATP induced calcium transient via IP(3)Rs (IICR) and the IICR were markedly decreased in ERp44 overexpressed Hela cells. The inhibitory effect of C160S/C212S but not C29S/T396A/ΔT(331-377) mutants of ERp44 on IICR were significantly decreased compared with ERp44. However, the binding capacity of ERp44 to L3V domain of IP(3)R(1) (1L3V) was enhanced by ERp44 C160S/C212S mutation. Taken together, these results suggest that the mutants of ERp44, C160/C212, can more tightly bind to IP(3)R(1) but exhibit a weak inhibition of IP(3)R(1) channel activity in Hela cells.
Adenosine Triphosphate
;
pharmacology
;
Amino Acid Substitution
;
Biological Transport
;
drug effects
;
physiology
;
Blotting, Western
;
Calcium
;
metabolism
;
Calcium Signaling
;
drug effects
;
physiology
;
HeLa Cells
;
Humans
;
Immunoprecipitation
;
Inositol 1,4,5-Trisphosphate
;
metabolism
;
Inositol 1,4,5-Trisphosphate Receptors
;
physiology
;
Membrane Potentials
;
drug effects
;
physiology
;
Membrane Proteins
;
genetics
;
metabolism
;
Microscopy, Confocal
;
Molecular Chaperones
;
genetics
;
metabolism
;
Mutation
;
Plasmids
;
Transfection
8.PAR-1 regulation of intracellular Ca²(+) mobilization in pulmonary giant cell carcinoma cell line PLA801D/PLA801C.
Yu-hong MENG ; Jin-qiang ZHANG ; Hao-yong NING ; Ping LU ; Liu HONG ; Xiao LIU ; Xiao-ling KANG ; Ji-yao YU ; Ying-lin LU
Chinese Journal of Pathology 2010;39(2):100-105
OBJECTIVESTo investigate molecular mechanisms of PAR-1 regulation on intracellular Ca²(+) mobilization in lung giant cell carcinoma cells in vitro and its involvement in tumor metastasis.
METHODSFree intracellular Ca²(+) ([Ca²(+)]i) was measured in lung giant cell carcinoma PLA801C and PLA801D cells by confocal microscopy. Sense and anti-sense PAR-1 expression vectors were transfected into PLA801C (C+)and PLA801D(D-) cells, respectively. The effects of PAR-1 expression were investigated by thrombin and TRAP-induced mobilization of [Ca²(+)]i in the C+ and D-cells.
RESULTSThere were significant differences of the mean values of [Ca²(+)]i between PLA801D (59.55) and PLA801C cells (35.46, P < 0.01). The mean [Ca²(+)]i of C+ cells (45.77) was significantly higher than that of its control CV cells (35.46, P < 0.05), and the mean [Ca²(+)]i of D-cells (48.42) was significantly lower than that of its control DV cells (59.55, P < 0.05). The peaks of [Ca²(+)]i of C+ and CV cells were 48.19 ± 9.84 and 45.64 ± 9.87 (P < 0.05) respectively at 80 s and 100 s after thrombin treatment, but were 111.31 ± 25.00 and 52.93 ± 11.21 (P < 0.05) respectively at 60 s after TRAP treatment. The peaks of [Ca²(+)]i of D- and DV cells were 40.71 ± 5.89 and 61.07 ± 21.36 (P < 0.05) respectively at 60 s after thrombin treatment, but were 84.98 ± 11.23 and 102.58 ± 21.48 (P < 0.05) respectively at 40 s after TRAP treatment.
CONCLUSIONSThe high metastatic potential of PLA801D and PLA801C may be related to [Ca²(+)]i of the tumor cells. PAR-1 may play an important role in the metastasis of lung giant cell carcinoma cells by up-regulating the intracellular Ca²(+).
Calcium ; metabolism ; Calcium Signaling ; drug effects ; Carcinoma, Giant Cell ; metabolism ; pathology ; Cell Line, Tumor ; DNA, Antisense ; genetics ; Humans ; Lung Neoplasms ; metabolism ; pathology ; RNA, Messenger ; metabolism ; Receptor, PAR-1 ; genetics ; metabolism ; physiology ; Receptors, Thrombin ; metabolism ; Thrombin ; pharmacology ; Transfection ; Up-Regulation
9.Effect of TRPV1 channel on proliferation and apoptosis of airway smooth muscle cells of rats.
Li-min ZHAO ; Hong-yan KUANG ; Luo-xian ZHANG ; Ji-zhen WU ; Xian-liang CHEN ; Xiao-yu ZHANG ; Li-jun MA
Journal of Huazhong University of Science and Technology (Medical Sciences) 2014;34(4):504-509
Airway remodeling is an important pathological feature of asthma and the basis of severe asthma. Proliferation of airway smooth muscle cells (ASMCs) is a major contributor to airway remodeling. As an important Ca(2+) channel, transient receptor potential vanilloid 1 (TRPV1) plays the key role in the cell pathological and physiological processes. This study investigated the expression and activity of TRPV1 channel, and further clarified the effect of TRPV1 channel on the ASMCs proliferation and apoptosis in order to provide the scientific basis to treat asthmatic airway remodeling in clinical practice. Immunofluorescence staining and reverse transcription polymerase chain reaction (RT-PCR) were used to detect the expression of TRPV1 in rat ASMCs. Intracellular Ca(2+) was detected using the single cell confocal fluorescence microscopy measurement loaded with Fluo-4/AM. The cell cycles were observed by flow cytometry. MTT assay and Hoechst 33258 staining were used to detect the proliferation and apoptosis of ASMCs in rats respectively. The data showed that: (1) TRPV1 channel was present in rat ASMCs. (2) TRPV1 channel agonist, capsaicin, increased the Ca(2+) influx in a concentration-dependent manner (EC50=284.3±58 nmol/L). TRPV1 channel antagonist, capsazepine, inhibited Ca(2+) influx in rat ASMCs. (3) Capsaicin significantly increased the percentage of S+G2M ASMCs and the absorbance of MTT assay. Capsazepine had the opposite effect. (4) Capsaicin significantly inhibited the apoptosis, whereas capsazepine had the opposite effect. These results suggest that TRPV1 is present and mediates Ca(2+) influx in rat ASMCs. TRPV1 activity stimulates proliferation of ASMCs in rats.
Animals
;
Antipruritics
;
pharmacology
;
Apoptosis
;
physiology
;
Bronchi
;
cytology
;
metabolism
;
Calcium Signaling
;
drug effects
;
physiology
;
Capsaicin
;
analogs & derivatives
;
pharmacology
;
Cell Proliferation
;
Myocytes, Smooth Muscle
;
cytology
;
metabolism
;
Rats
;
Rats, Sprague-Dawley
;
TRPV Cation Channels
;
antagonists & inhibitors
;
metabolism
10.Tacrolimus inhibits vasoconstriction by increasing Ca(2+) sparks in rat aorta.
Yu-fang CHEN ; Chen WANG ; Rui ZHANG ; Huan WANG ; Rong MA ; Si JIN ; Ji-zhou XIANG ; Qiang TANG
Journal of Huazhong University of Science and Technology (Medical Sciences) 2016;36(1):8-13
The present study attempted to test a novel hypothesis that Ca(2+) sparks play an important role in arterial relaxation induced by tacrolimus. Recorded with confocal laser scanning microscopy, tacrolimus (10 µmol/L) increased the frequency of Ca(2+) sparks, which could be reversed by ryanodine (10 µmol/L). Electrophysiological experiments revealed that tacrolimus (10 µmol/L) increased the large-conductance Ca(2+)-activated K(+) currents (BKCa) in rat aortic vascular smooth muscle cells (AVSMCs), which could be blocked by ryanodine (10 µmol/L). Furthermore, tacrolimus (10 and 50 µmol/L) reduced the contractile force induced by norepinephrine (NE) or KCl in aortic vascular smooth muscle in a concentration-dependent manner, which could be also significantly attenuated by iberiotoxin (100 nmol/L) and ryanodine (10 µmol/L) respectively. In conclusion, tacrolimus could indirectly activate BKCa currents by increasing Ca(2+) sparks released from ryanodine receptors, which inhibited the NE- or KCl-induced contraction in rat aorta.
Animals
;
Aorta
;
cytology
;
metabolism
;
physiology
;
Calcium Signaling
;
Cells, Cultured
;
Large-Conductance Calcium-Activated Potassium Channels
;
metabolism
;
Male
;
Muscle, Smooth, Vascular
;
drug effects
;
metabolism
;
physiology
;
Myocytes, Smooth Muscle
;
drug effects
;
metabolism
;
Norepinephrine
;
pharmacology
;
Rats
;
Rats, Sprague-Dawley
;
Ryanodine
;
pharmacology
;
Tacrolimus
;
pharmacology
;
Vasoconstriction