Home>>Signaling Pathways>> Neuroscience>> Gap Junction>>Gap19

Gap19 Sale

目录号 : GC17681

An inhibitor of Cx43 hemichannels

Gap19 Chemical Structure

Cas No.:1507930-57-5

规格 价格 库存 购买数量
1mg
¥1,050.00
现货
5mg
¥3,150.00
现货

电话:400-920-5774 Email: sales@glpbio.cn

Customer Reviews

Based on customer reviews.

Sample solution is provided at 25 µL, 10mM.

产品文档

Quality Control & SDS

View current batch:

实验参考方法

Cell experiment [1]:

Cell lines

Astrocyte cell

Preparation Method

Concentration-dependent inhibition by Gap19 (30 min pre-incubation) of ATP release in cultured cortical astrocytes triggered by glutamate.

Reaction Conditions

0-104uM Gap19 for 30 min

Applications

Gap19 (30 min treatment) inhibited glutamate-triggered ATP release (Figure 2A).Etd+ uptake was inhibited in the presence of Gap19 in a dose-dependent manner, with the peptide applied prior to (30 min) and during Etd+ uptake.Gap19 was without any effect on gap junctional communication in astrocytes which in culture express only Cx43.

Animal experiment [2]:

Animal models

Male ICR mice (25-30 g)

Preparation Method

Animals were randomly divided into different groups as follows: (I) vehicle group (sham); (II) Gap19 treated vehicle group (Gap19 group); (III) I/R group; (IV) Gap26 treated I/R group (I/R + Gap26 group); (V) Gap19 treated I/R group (I/R + Gap19 group).10 µg Gap19/Gap26 in 10 µl ddH2O was injected in lateral ventricle (300 µg/kg body weight, i.c.v.). TAT-Gap19 was administered at a dose of 25 mg/kg body weight (i.p.) in the post-treatment groups.

Dosage form

10 µg Gap19

Applications

When investigated the role of Gap19 on cerebral ischemia/reperfusion (I/R) injury in a mouse model of middle cerebral artery occlusion (MCAO). Ventricle-injected Gap19 significantly alleviated infarct volume, neuronal cell damage and neurological deficits after ischemia, the neuroprotective effect of Gap19 was significant stronger than Gap26. Post-treatment with TAT-Gap19 still provided neuroprotection when it was administered intraperitoneally at 4 h after reperfusion.

References:

[1]. Abudara V, Bechberger J, et,al. The connexin43 mimetic peptide Gap19 inhibits hemichannels without altering gap junctional communication in astrocytes. Front Cell Neurosci. 2014 Oct 21;8:306. doi: 10.3389/fncel.2014.00306. PMID: 25374505; PMCID: PMC4204617.
[2].Chen B, Yang L, et,al. Inhibition of Connexin43 hemichannels with Gap19 protects cerebral ischemia/reperfusion injury via the JAK2/STAT3 pathway in mice. Brain Res Bull. 2019 Mar;146:124-135. doi: 10.1016/j.brainresbull.2018.12.009. Epub 2018 Dec 26. PMID: 30593877.

产品描述

Gap19 (KQIEIKKFK) blocks HCs but not GJCs and is specific for Cx43. Gap19 may resist myocardial ischemic injury[2]. Gap19 has a bimodal effect on Cx43 HC gating, decreasing gating to the fully open state while increasing substate gating, suggesting that Gap19 acts like a gating modifier on Cx43 HC.Gap19 disrupts the CT-CL interaction, thus making hc difficult to open[7].

Gap19, as a nonapeptide derived from the cytoplasmic loop of Cx43, inhibits astroglial Cx43 hemichannels in a dose-dependent manner, without affecting gap junction channels. This peptide, which not only selectively inhibits hemichannels but is also specific for Cx43[5].

When investigated the role of Gap19 on cerebral ischemia/reperfusion (I/R) injury in a mouse model of middle cerebral artery occlusion (MCAO). Ventricle-injected Gap19 significantly alleviated infarct volume, neuronal cell damage and neurological deficits after ischemia, the neuroprotective effect of Gap19 was significant stronger than Gap26. Post-treatment with TAT-Gap19 still provided neuroprotection when it was administered intraperitoneally at 4 h after reperfusion[1]. Gap19 exerted a neuroprotective effect after stroke via inhibition of the TLR4-mediated signaling pathway[4]. Treatment with Gap19 prevented metabolic inhibition-enhanced hemichannel openings, protected cardiomyocytes against volume overload and cell death following ischemia/reperfusion in vitro and modestly decreased the infarct size after myocardial ischemia/reperfusion in mice in vivo[3]. Intra-TG injection of the mimetic peptide GAP19, which inhibits Cx43 hemichannel formation, greatly reduced TMJ-evoked MMemg activity in all CFA-inflamed groups, while activity in sham groups was not affected[6].

References:
[1]. Chen B, Yang L, et,al. Inhibition of Connexin43 hemichannels with Gap19 protects cerebral ischemia/reperfusion injury via the JAK2/STAT3 pathway in mice. Brain Res Bull. 2019 Mar;146:124-135. doi: 10.1016/j.brainresbull.2018.12.009. Epub 2018 Dec 26. PMID: 30593877.
[2]. Wang JP, Yang ZT, et,al. L-carnosine inhibits neuronal cell apoptosis through signal transducer and activator of transcription 3 signaling pathway after acute focal cerebral ischemia. Brain Res. 2013 Apr 24;1507:125-33. doi: 10.1016/j.brainres.2013.02.032. Epub 2013 Feb 27. PMID: 23454231.
[3]. Wang N, De Vuyst E, et,al. Selective inhibition of Cx43 hemichannels by Gap19 and its impact on myocardial ischemia/reperfusion injury. Basic Res Cardiol. 2013 Jan;108(1):309. doi: 10.1007/s00395-012-0309-x. Epub 2012 Nov 8. PMID: 23184389; PMCID: PMC3666173.
[4]. Chen Y, Wang L, et,al. Inhibition of Connexin 43 Hemichannels Alleviates Cerebral Ischemia/Reperfusion Injury via the TLR4 Signaling Pathway. Front Cell Neurosci. 2018 Oct 17;12:372. doi: 10.3389/fncel.2018.00372. PMID: 30386214; PMCID: PMC6199357.
[5]. Abudara V, Bechberger J, et,al. The connexin43 mimetic peptide Gap19 inhibits hemichannels without altering gap junctional communication in astrocytes. Front Cell Neurosci. 2014 Oct 21;8:306. doi: 10.3389/fncel.2014.00306. PMID: 25374505; PMCID: PMC4204617.
[6]. Ahmed F, Rahman M, et,al. Role of Connexin 43 in an Inflammatory Model for TMJ Hyperalgesia. Front Pain Res (Lausanne). 2021 Aug 3;2:715871. doi: 10.3389/fpain.2021.715871. PMID: 35295418; PMCID: PMC8915650.
[7]. Lissoni A, Wang N, et,al. Gap19, a Cx43 Hemichannel Inhibitor, Acts as a Gating Modifier That Decreases Main State Opening While Increasing Substate Gating. Int J Mol Sci. 2020 Oct 5;21(19):7340. doi: 10.3390/ijms21197340. PMID: 33027889; PMCID: PMC7583728.

Gap19 (KQIEIKKFK) 阻断 HC 但不阻断 GJC,并且特定于 Cx43。 Gap19可能抵抗心肌缺血损伤[2]。 Gap19 对 Cx43 HC 门控具有双峰效应,减少完全打开状态的门控,同时增加亚状态门控,表明 Gap19 就像 Cx43 HC 上的门控修饰符。Gap19 破坏 CT-CL 相互作用,从而使 hc 难以打开[7].

Gap19 作为一种来源于 Cx43 细胞质环的九肽,以剂量依赖性方式抑制星形胶质细胞 Cx43 半通道,而不影响间隙连接通道。该肽不仅选择性抑制半通道,而且对 Cx43[5] 具有特异性。

研究 Gap19 在脑缺血/再灌注 (I/R) 损伤中的作用时在大脑中动脉闭塞 (MCAO) 的小鼠模型中。脑室注射Gap19可显着减轻缺血后的梗死体积、神经元细胞损伤和神经功能缺损,Gap19的神经保护作用明显强于Gap26。再灌注后 4 小时腹膜内给药时,TAT-Gap19 后处理仍提供神经保护作用[1]。 Gap19 通过抑制 TLR4 介导的信号通路在卒中后发挥神经保护作用[4]。 Gap19 治疗可防止代谢抑制增强的半通道开放,保护心肌细胞免受体外缺血/再灌注后的体积超载和细胞死亡,并适度减少小鼠体内心肌缺血/再灌注后的梗塞面积[3] . TG 内注射抑制 Cx43 半通道形成的模拟肽 GAP19,大大降低了所有 CFA 炎症组中 TMJ 诱发的 MMemg 活性,而假手术组的活性未受影响[6]。< /p>

Chemical Properties

Cas No. 1507930-57-5 SDF
Canonical SMILES CC[C@]([C@@](/N=C(O)/[C@](/N=C(O)/[C@](/N=C(O)/[C@](/N=C(O)/[C@](N)([H])CCCCN)([H])CCC(O)=N)([H])[C@@](CC)([H])C)([H])CCC(O)=O)([H])/C(O)=N/[C@@](/C(O)=N/[C@@](/C(O)=N/[C@@](/C(O)=N/[C@@](C(O)=O)([H])CCCCN)([H])CC1=CC=CC=C1)([H])CCCCN)([H])CCCCN)([H])C
分子式 C55H96N14O13 分子量 1161.45
溶解度 ≥ 58.0725mg/mL in Water 储存条件 Store at -20°C
General tips 请根据产品在不同溶剂中的溶解度选择合适的溶剂配制储备液;一旦配成溶液,请分装保存,避免反复冻融造成的产品失效。
储备液的保存方式和期限:-80°C 储存时,请在 6 个月内使用,-20°C 储存时,请在 1 个月内使用。
为了提高溶解度,请将管子加热至37℃,然后在超声波浴中震荡一段时间。
Shipping Condition 评估样品解决方案:配备蓝冰进行发货。所有其他可用尺寸:配备RT,或根据请求配备蓝冰。

溶解性数据

制备储备液
1 mg 5 mg 10 mg
1 mM 0.861 mL 4.305 mL 8.6099 mL
5 mM 0.1722 mL 0.861 mL 1.722 mL
10 mM 0.0861 mL 0.4305 mL 0.861 mL
  • 摩尔浓度计算器

  • 稀释计算器

  • 分子量计算器

质量
=
浓度
x
体积
x
分子量
 
 
 
*在配置溶液时,请务必参考产品标签上、MSDS / COA(可在Glpbio的产品页面获得)批次特异的分子量使用本工具。

计算

动物体内配方计算器 (澄清溶液)

第一步:请输入基本实验信息(考虑到实验过程中的损耗,建议多配一只动物的药量)
给药剂量 mg/kg 动物平均体重 g 每只动物给药体积 ul 动物数量
第二步:请输入动物体内配方组成(配方适用于不溶于水的药物;不同批次药物配方比例不同,请联系GLPBIO为您提供正确的澄清溶液配方)
% DMSO % % Tween 80 % saline
计算重置

Research Update

Xentry-Gap19 inhibits Connexin43 hemichannel opening especially during hypoxic injury

Hypoxic injury results in cell death, tissue damage and activation of inflammatory pathways. This is mediated by pathological Connexin43 (Cx43) hemichannel (HC) opening resulting in osmotic and ionic imbalances as well as cytokine production perpetuating the inflammatory environment. Gap19 is an intracellularly acting Cx43 mimetic peptide that blocks HC opening and thus promotes cell survival. However, native Gap19, which must enter the cell in order to function, exhibits low cell permeability. In this study, Gap19 was conjugated to the cell-penetrating peptide, Xentry, to investigate if cellular uptake could be improved while maintaining peptide function. Cellular uptake of Xentry-Gap19 (XG19) was much greater than that of native Gap19 even under normal cell culture conditions. Peptide function was maintained post uptake as shown by reduced ethidium homodimer influx and ATP release due to Cx43 HC block. While XG19 blocked pathologic HC opening though, normal gap junction communication required for cell repair and survival mechanisms was not affected as shown in a dye scrape-load assay. Under hypoxic conditions, increased expression of Syndecan-4, a plasma membrane proteoglycan targeted by Xentry, enabled even greater XG19 uptake leading to higher inhibition of ATP release and greater cell survival. This suggests that XG19, which is targeted specifically to hypoxic cells, can efficiently and safely block Cx43 HC and could therefore be a novel treatment for hypoxic and inflammatory diseases. Graphical abstract.

Targeting connexin 43 provides anti-inflammatory effects after intracerebral hemorrhage injury by regulating YAP signaling

Background: In the central nervous system (CNS), connexin 43 (Cx43) is mainly expressed in astrocytes and regulates astrocytic network homeostasis. Similar to Cx43 overexpression, abnormal excessive opening of Cx43 hemichannels (Cx43Hcs) on reactive astrocytes aggravates the inflammatory response and cell death in CNS pathologies. However, the role of excessive Cx43Hc opening in intracerebral hemorrhage (ICH) injury is not clear.
Methods: Hemin stimulation in primary cells and collagenase IV injection in C57BL/6J (B6) mice were used as ICH models in vitro and in vivo. After ICH injury, the Cx43 mimetic peptide Gap19 was used for treatment. Ethidium bromide (EtBr) uptake assays were used to measure the opening of Cx43Hcs. Western blotting and immunofluorescence were used to measure protein expression. qRT-PCR and ELISA were used to determine the levels of cytokines. Coimmunoprecipitation (Co-IP) and the Duolink in situ proximity ligation assay (PLA) were applied to measure the association between proteins.
Results: In this study, Cx43 expression upregulation and excessive Cx43Hc opening was observed in mice after ICH injury. Delayed treatment with Gap19 significantly alleviated hematoma volume and neurological deficits after ICH injury. In addition, Gap19 decreased inflammatory cytokine levels in the tissue surrounding the hematoma and decreased reactive astrogliosis after ICH injury in vitro and in vivo. Intriguingly, Cx43 transcriptional activity and expression in astrocytes were significantly increased after hemin stimulation in culture. However, Gap19 treatment downregulated astrocytic Cx43 expression through the ubiquitin-proteasome pathway without affecting Cx43 transcription. Additionally, our data showed that Gap19 increased Yes-associated protein (YAP) nuclear translocation. This subsequently upregulated SOCS1 and SOCS3 expression and then inhibited the TLR4-NFκB and JAK2-STAT3 pathways in hemin-stimulated astrocytes. Finally, the YAP inhibitor, verteporfin (VP), reversed the anti-inflammatory effect of Gap19 in vitro and almost completely blocked its protective effects in vivo after ICH injury.
Conclusions: This study provides new insight into potential treatment strategies for ICH injury involving astroglial Cx43 and Cx43Hcs. Suppression of abnormal astroglial Cx43 expression and Cx43Hc opening by Gap19 has anti-inflammatory and neuroprotective effects after ICH injury.

MST1 Suppresses Disturbed Flow Induced Atherosclerosis

Background: Atherosclerosis occurs mainly at arterial branching points exposed to disturbed blood flow. How MST1 (mammalian sterile 20-like kinase 1), the primary kinase in the mechanosensitive Hippo pathway modulates disturbed flow induced endothelial cells (ECs) activation and atherosclerosis remains unclear.
Methods: To assess the role of MST1 in vivo, mice with EC-specific Mst1 deficiency on ApoE-/- background (Mst1iECKOApoE-/-) were used in an atherosclerosis model generated by carotid artery ligation. Mass spectrometry, immunoprecipitation, proximity ligation assay, and dye uptake assay were used to identify the functional substrate of MST1. Human umbilical vein endothelial cells and human aortic endothelial cells were subjected to oscillatory shear stress that mimic disturbed flow in experiments conducted in vitro.
Results: We found that the phosphorylation of endothelial MST1 was significantly inhibited in oscillatory shear stress-exposed regions of human and mouse arteries and ECs. Ectopic lenti-mediated overexpression of wild-type MST1, but not a kinase-deficient mutant of MST1, reversed disturbed flow-caused EC activation and atherosclerosis in EC-specific Mst1 deficiency on ApoE-/- background (Mst1iECKOApoE-/-). Inhibition of MST1 by oscillatory shear stress led to reduced phosphorylation of Cx43 (connexin 43) at Ser255, the Cx43 hemichannel open, EC activation, and atherosclerosis, which were blocked by TAT-GAP19, a Cx43 hemichannel inhibitory peptide. Mass spectrometry studies identified that Filamin B fueled the translocation of Cx43 to lipid rafts for further hemichannel open. Finally, lenti-mediated overexpression of the Cx43S255 mutant into glutamate to mimic phosphorylation blunted disturbed flow-induced EC activation, thereby inhibiting the atherogenesis in both ApoE-/- and Mst1 iECKOApoE-/- mice.
Conclusions: Our study reveals that inhibition of the MST1-Cx43 axis is an essential driver of oscillatory shear stress-induced endothelial dysfunction and atherosclerosis, which provides a new therapeutic target for the treatment of atherosclerosis.

Gap19, a Cx43 Hemichannel Inhibitor, Acts as a Gating Modifier That Decreases Main State Opening While Increasing Substate Gating

Cx43 hemichannels (HCs) are electrically and chemically gated transmembrane pores with low open probability and multiple conductance states, which makes kinetic studies of channel gating in large datasets challenging. Here, we developed open access software, named HemiGUI, to analyze HC gating transitions and investigated voltage-induced HC opening based on up to ≈4000 events recorded in HeLa-Cx43-overexpressing cells. We performed a detailed characterization of Cx43 HC gating profiles and specifically focused on the role of the C-terminal tail (CT) domain by recording the impact of adding an EGFP tag to the Cx43 CT end (Cx43-EGFP) or by supplying the Cx43 HC-inhibiting peptide Gap19 that interferes with CT interaction with the cytoplasmic loop (CL). We found that Gap19 not only decreased HC opening activity to the open state (≈217 pS) but also increased the propensity of subconductance (≈80 pS) transitions that additionally became slower as compared to the control. The work demonstrates that large sample transition analysis allows detailed investigations on Cx43 HC gating and shows that Gap19 acts as a HC gating modifier by interacting with the CT that forms a crucial gating element.

TAT-Gap19 and Carbenoxolone Alleviate Liver Fibrosis in Mice

Although a plethora of signaling pathways are known to drive the activation of hepatic stellate cells in liver fibrosis, the involvement of connexin-based communication in this process remains elusive. Connexin43 expression is enhanced in activated hepatic stellate cells and constitutes the molecular building stone of hemichannels and gap junctions. While gap junctions support intercellular communication, and hence the maintenance of liver homeostasis, hemichannels provide a circuit for extracellular communication and are typically opened by pathological stimuli, such as oxidative stress and inflammation. The present study was set up to investigate the effects of inhibition of connexin43-based hemichannels and gap junctions on liver fibrosis in mice. Liver fibrosis was induced by administration of thioacetamide to Balb/c mice for eight weeks. Thereafter, mice were treated for two weeks with TAT-Gap19, a specific connexin43 hemichannel inhibitor, or carbenoxolone, a general hemichannel and gap junction inhibitor. Subsequently, histopathological analysis was performed and markers of hepatic damage and functionality, oxidative stress, hepatic stellate cell activation and inflammation were evaluated. Connexin43 hemichannel specificity of TAT-Gap19 was confirmed in vitro by fluorescence recovery after photobleaching analysis and the measurement of extracellular release of adenosine-5'-triphosphate. Upon administration to animals, both TAT-Gap19 and carbenoxolone lowered the degree of liver fibrosis accompanied by superoxide dismutase overactivation and reduced production of inflammatory proteins, respectively. These results support a role of connexin-based signaling in the resolution of liver fibrosis, and simultaneously demonstrate the therapeutic potential of TAT-Gap19 and carbenoxolone in the treatment of this type of chronic liver disease.