首页 | 本学科首页   官方微博 | 高级检索  
相似文献
 共查询到20条相似文献,搜索用时 0 毫秒
1.
JGP study shows that a mechanosensitive complex containing Piezo1 and Pannexin1 couples osmotic pressure to ATP secretion in bile duct cholangiocytes.

Cholangiocytes are epithelial cells that line the bile ducts within the liver and modify the composition of hepatocyte-derived bile. In this issue of JGP, Desplat et al. identify a mechanosensory complex that may help cholangiocytes respond to changes in osmotic pressure (1).Angélique Desplat (left), Patrick Delmas (center), and colleagues identify a mechanosensitive pathway that couples hypotonic stress to calcium influx and ATP release in cholangiocytes. Cell swelling induces calcium influx through the stretch-activated ion channel Piezo, triggering ATP release by Pannexin1 channels. This leads to the activation of P2X4 receptors and further calcium influx. Piezo1 (red) and Pannexin1 (green) colocalize in cells and may interact to form a mechanosensory complex that facilitates the hypotonic stress response.The activity of cholangiocytes can be regulated not only by chemical signals, such as hormones and bile acids, but also by mechanical cues arising from changes in bile composition and flow. “Abnormal mechanical tension is also an aggravating factor in many biliary diseases, including primary sclerosing cholangitis,” explains Patrick Delmas, a Research Director at Centre National de la Recherche Scientifique/Aix-Marseille-Université. “So, identifying the molecular players in cholangiocyte force sensing could provide a step forward for better management of biliary diseases.”Current models suggest that mechanical cues trigger an influx of calcium into cholangiocytes, leading to the release of ATP, which, by stimulating purinergic receptors at the cell surface, promotes further calcium influx and induces the secretion of anions, water, and HCO3 to modify the tonicity and pH of hepatic bile (2, 3). To identify mechanosensitive proteins that might regulate this pathway, Delmas and colleagues, including first author Angélique Desplat, purified mouse cholangiocytes from intrahepatic bile ducts and subjected them to hypotonic stress (1). The subsequent cell swelling activates calcium influx and ATP release.Desplat et al. found that depleting or inhibiting the stretch-activated ion channel Piezo1 significantly reduced this response to hypotonic stress. This mechanosensitive channel mediates the initial calcium influx into cholangiocytes when activated by cell swelling.The subsequent release of ATP is mediated by a different channel, however. Desplat et al. found that cholangiocytes express high levels of the gap junction family protein Pannexin1, and that pharmacologically inhibiting Pannexin1 channels reduced the amount of ATP released in response to hypotonic stress and Piezo1 activation.Delmas and colleagues suspect that the increase in intracellular calcium mediated by Piezo1 may activate Pannexin1 channels to release ATP, and this activation may be facilitated by a physical association between the two proteins: the researchers found that recombinant versions of the two channel proteins colocalize within the plasma membrane of cholangiocytes and can be coimmunoprecipitated.Finally, the researchers determined that the ATP released through Pannexin1 channels amplifies the signal initiated by hypotonic stress by activating purinergic P2X4 receptors, leading to further increases in intracellular calcium levels. Transfecting Piezo1-deficient HEK293 cells, which usually don’t respond to hypotonic stress, with cDNAs encoding Piezo1, Pannexin1, and P2X4R was sufficient to reconstitute the entire pathway of calcium influx and ATP release.Cholangiocytes express other mechanosensitive channels, including TRPV4, which has previously been implicated in the cells’ response to hypotonic stress (4). The functions of TRPV4 and Piezo1 may therefore be partially redundant, providing some robustness to cholangiocytes mechanical signaling pathways. However, it is also possible that, in vivo, the two channels respond to different stimuli and elicit distinct downstream effects. “Further investigation is warranted to better understand the respective roles of these two molecular players,” says Delmas. “To continue our work, we would like to challenge our model in vivo by testing whether Piezo1 agonists are able to regulate bile acid secretion.”  相似文献   

2.
Piezo1 is a eukaryotic cation-selective mechanosensitive ion channel. To understand channel function in vivo, we first need to analyze and compare the response in the whole cell and the patch. In patches, Piezo1 inactivates and the current is fit well by a 3-state model with a single pressure-dependent rate. However, repeated stimulation led to an irreversible loss of inactivation. Remarkably, the loss of inactivation did not occur on a channel-by-channel basis but on all channels at the same time. Thus, the channels are in common mechanical domain. Divalent ions decreased the unitary conductance from ~68 pS to ~37 pS, irrespective of the cation species. Mg and Ca did not affect inactivation rates, but Zn caused a 3-fold slowing. CytochalasinD (cytoD) does not alter inactivation rates or the transition to the non-inactivating mode but does reduce the steady-state response. Whole-cell currents were similar to patch currents but also had significant differences. In contrast to the patch, cytoD inhibited the current suggesting that the activating forces were transmitted through the actin cytoskeleton. Hypotonic swelling that prestressed the cytoskeleton and the bilayer greatly increased the sensitivity of both control and cytoD cells so there are two pathways to transmit force to the channels. In contrast to patch, removing divalent ions decreased the whole-cell current. The difference between whole cell and patch properties provide new insights into our understanding of the Piezo1 gating mechanisms and cautions against generalization to in situ behavior.  相似文献   

3.
Piezo1是哺乳动物中新发现的一种机械敏感(mechanosensitive,MS)离子通道,在不同组织和器官中发挥着重要功能,包括骨骼、泌尿道、眼球和动脉等。然而,异常的Piezo1机械传导会造成多种疾病的发生并促进病程的发展。纤维化疾病几乎可以发生在任何一个组织和器官中,其主要特征是胶原蛋白和其他细胞外基质(extracellular matrix,ECM)成分的过度交联与累积,最终导致组织器官刚度增加,生理功能受到影响。目前,越来越多的研究表明,Piezo1在纤维化疾病的发生和发展中扮演着重要的调控作用,与其基质力学状态变化有着密切联系。本文叙述了Piezo1的结构和激活机理,并且系统地总结了Piezo1在心、肾、胰和肝等多种器官纤维化疾病中的研究进展,以期为纤维化疾病的治疗提供新的视角和策略。  相似文献   

4.
Ryanodine receptors (RyRs), located in the sarcoplasmic/endoplasmic reticulum (SR/ER) membrane, are required for intracellular Ca2+ release that is involved in a wide range of cellular functions. In addition to Ca2+-induced Ca2+ release in cardiac cells and voltage-induced Ca2+ release in skeletal muscle cells, we recently identified another mode of intracellular Ca2+ mobilization mediated by RyR, i.e., nitric oxide-induced Ca2+ release (NICR), in cerebellar Purkinje cells. NICR is evoked by neuronal activity, is dependent on S-nitrosylation of type 1 RyR (RyR1) and is involved in the induction of long-term potentiation (LTP) of cerebellar synapses. In this addendum, we examined whether peroxynitrite, which is produced by the reaction of nitric oxide with superoxide, may also have an effect on the Ca2+ release via RyR1 and the cerebellar LTP. We found that scavengers of peroxynitrite have no significant effect either on the Ca2+ release via RyR1 or on the cerebellar LTP. We also found that an application of a high concentration of peroxynitrite does not reproduce neuronal activity-dependent Ca2+ release in Purkinje cells. These results support that NICR is induced by endogenous nitric oxide produced by neuronal activity through S-nitrosylation of RyR1.  相似文献   

5.
6.
《Developmental cell》2021,56(20):2856-2870.e7
  1. Download : Download high-res image (194KB)
  2. Download : Download full-size image
  相似文献   

7.
8.
Bae C  Sachs F  Gottlieb PA 《Biochemistry》2011,50(29):6295-6300
Cells can respond to mechanical stress by gating mechanosensitive ion channels (MSCs). The cloning of Piezo1, a eukaryotic cation selective MSC, defines a new system for studying mechanical transduction at the cellular level. Because Piezo1 has electrophysiological properties similar to those of endogenous cationic MSCs that are selectively inhibited by the peptide GsMTx4, we tested whether the peptide targets Piezo1 activity. Extracellular GsMTx4 at micromolar concentrations reversibly inhibited ~80% of the mechanically induced current of outside-out patches from transfected HEK293 cells. The inhibition was voltage insensitive, and as seen with endogenous MSCs, the mirror image d enantiomer inhibited like the l. The rate constants for binding and unbinding based on Piezo1 current kinetics provided association and dissociation rates of 7.0 × 10(5) M(-1) s(-1) and 0.11 s(-1), respectively, and a K(D) of ~155 nM, similar to values previously reported for endogenous MSCs. Consistent with predicted gating modifier behavior, GsMTx4 produced an ~30 mmHg rightward shift in the pressure-gating curve and was active on closed channels. In contrast, streptomycin, a nonspecific inhibitor of cationic MSCs, showed the use-dependent inhibition characteristic of open channel block. The peptide did not block currents of the mechanical channel TREK-1 on outside-out patches. Whole-cell Piezo1 currents were also reversibly inhibited by GsMTx4, and although the off rate was nearly identical to that of outside-out patches, differences were observed for the on rate. The ability of GsMTx4 to target the mechanosensitivity of Piezo1 supports the use of this channel in high-throughput screens for pharmacological agents and diagnostic assays.  相似文献   

9.
Osteocytes function as critical regulators of bone homeostasis by coordinating the functions of osteoblasts and osteoclasts, and are constantly exposed to mechanical force. However, the molecular mechanism underlying the mechanical signal transduction in osteocytes is not well understood. Here, we found that Yoda1, a selective Piezo1 agonist, increased intracellular calcium mobilization and dose-dependently decreased the expression of Sost (encoding Sclerostin) in the osteocytic cell line IDG-SW3. We also demonstrated that mechanical stretch of IDG-SW3 suppressed Sost expression, a result which was abrogated by treatment with the Piezo1 inhibitor GsMTx4, and the deficiency of Piezo1. Furthermore, the suppression of Sost expression was abolished by treatment with an Akt inhibitor. Taken together, these results indicate that the activation of the Piezo1-Akt pathway in osteocytes is required for mechanical stretch-induced downregulation of Sost expression.  相似文献   

10.
《Current biology : CB》2022,32(16):3584-3592.e4
  1. Download : Download high-res image (224KB)
  2. Download : Download full-size image
  相似文献   

11.
《Cell reports》2020,30(3):870-880.e2
  1. Download : Download high-res image (144KB)
  2. Download : Download full-size image
  相似文献   

12.
Mechanical forces associated with fluid flow and/or circumferential stretch are sensed by renal epithelial cells and contribute to both adaptive or disease states. Non‐selective stretch‐activated ion channels (SACs), characterized by a lack of inactivation and a remarkably slow deactivation, are active at the basolateral side of renal proximal convoluted tubules. Knockdown of Piezo1 strongly reduces SAC activity in proximal convoluted tubule epithelial cells. Similarly, overexpression of Polycystin‐2 (PC2) or, to a greater extent its pathogenic mutant PC2‐740X, impairs native SACs. Moreover, PC2 inhibits exogenous Piezo1 SAC activity. PC2 coimmunoprecipitates with Piezo1 and deletion of its N‐terminal domain prevents both this interaction and inhibition of SAC activity. These findings indicate that renal SACs depend on Piezo1, but are critically conditioned by PC2.  相似文献   

13.
14.
15.
Piezo1 is a eukaryotic cation-selective mechanosensitive ion channel. To understand channel function in vivo, we first need to analyze and compare the response in the whole cell and the patch. In patches, Piezo1 inactivates and the current is fit well by a 3-state model with a single pressure-dependent rate. However, repeated stimulation led to an irreversible loss of inactivation. Remarkably, the loss of inactivation did not occur on a channel-by-channel basis but on all channels at the same time. Thus, the channels are in common mechanical domain. Divalent ions decreased the unitary conductance from ~68 pS to ~37 pS, irrespective of the cation species. Mg and Ca did not affect inactivation rates, but Zn caused a 3-fold slowing. CytochalasinD (cytoD) does not alter inactivation rates or the transition to the non-inactivating mode but does reduce the steady-state response. Whole-cell currents were similar to patch currents but also had significant differences. In contrast to the patch, cytoD inhibited the current suggesting that the activating forces were transmitted through the actin cytoskeleton. Hypotonic swelling that prestressed the cytoskeleton and the bilayer greatly increased the sensitivity of both control and cytoD cells so there are two pathways to transmit force to the channels. In contrast to patch, removing divalent ions decreased the whole-cell current. The difference between whole cell and patch properties provide new insights into our understanding of the Piezo1 gating mechanisms and cautions against generalization to in situ behavior.  相似文献   

16.
Piezo1 forms a mechanically activated calcium-permeable nonselective cation channel that is functionally important in many cell types. Structural data exist for C-terminal regions, but we lack information about N-terminal regions and how the entire channel interacts with the lipid bilayer. Here, we use computational approaches to predict the three-dimensional structure of the full-length Piezo1 and simulate it in an asymmetric membrane. A number of novel insights are suggested by the model: 1) Piezo1 creates a trilobed dome in the membrane that extends beyond the radius of the protein, 2) Piezo1 changes the lipid environment in its vicinity via preferential interactions with cholesterol and phosphatidylinositol 4,5-bisphosphate (PIP2) molecules, and 3) cholesterol changes the depth of the dome and PIP2 binding preference. In vitro alteration of cholesterol concentration inhibits Piezo1 activity in a manner complementing some of our computational findings. The data suggest the importance of N-terminal regions of Piezo1 for dome structure and membrane cholesterol and PIP2 interactions.  相似文献   

17.
18.
19.
Interleukin-1 (IL-1) is an inflammatory mediator that increases Cl- secretion in intestinal epithelial cells. To identify the signal transduction pathway(s) involved in IL-1's action, cells were treated with IL-1 and the levels of cyclooxygenase (COX) enzymes, prostaglandin E2 (PGE2) and phospholipase A2-activating protein (PLAP), and the activity of phospholipase A2 (PLA2) were measured. IL-1 caused concentration- and time-dependent increases in the levels of PLA2 activity, and/or in the levels of PLAP, COX-2 and PGE2. The IL-induced increase in PGE2 levels was biphasic, with the first peak due to the increase in PLAP levels, and the second peak due to the increase in COX-2 levels. This increase in PGE2 levels may provide a mechanism for acute and chronic inflammation in the intestine.  相似文献   

20.
Androgens induce rat prostate induction from the urogenital sinus epithelium at embryonic day 17.5. Subsequent morphogenesis, including epithelial cord growth, branching, and canalization, results from concerted paracrine interactions with the stroma. A significant number of paracrine factors bind heparan sulfate (HS). We hypothesized that interfering with overall sulfation could disrupt the signaling mediated by HS-binding factors and that the undersulfated environment would allow investigation of individual exogenous morphogens. First, we investigated whether acinar morphogenesis involved HS-proteoglycan expression and found that syndecans 1 and 3 were upregulated in RWPE1 cells in the transition from two- to three-dimensional (3D) Matrigel, capable of promoting spheroid formation. We then investigated whether sodium chlorate, a general sulfation inhibitor, interfered with spheroid formation by RWPE1 cells and acinar morphogenesis in ex vivo ventral prostate (VP) organ culture. As expected, treatment with sodium chlorate inhibited spheroid formation by RWPE1 cells in 3D culture. Chlorate also inhibited ex vivo VP epithelial branching and canalization, resulting in long branchless epithelial structures. We then investigated whether the HS-binding factors, FGF10, TGFβ1, and SDF1, could reverse the effect of sodium chlorate. Although no effect was seen in the FGF10- and TGFβ1-treated samples, SDF1 promoted epithelial canalization in the low sulfated environment, highlighting its specific role in lumen formation. Altogether, the results show that sodium chlorate perturbed prostate morphogenesis and allowed investigation of factors involved in branching and/or canalization, implicating SDF1 signaling in epithelial canalization.  相似文献   

设为首页 | 免责声明 | 关于勤云 | 加入收藏

Copyright©北京勤云科技发展有限公司  京ICP备09084417号