CN114515337A - Application of NADPH oxidase 2 as therapeutic target in preparation of medicine for treating vascular dysfunction - Google Patents

Application of NADPH oxidase 2 as therapeutic target in preparation of medicine for treating vascular dysfunction Download PDF

Info

Publication number
CN114515337A
CN114515337A CN202210124897.3A CN202210124897A CN114515337A CN 114515337 A CN114515337 A CN 114515337A CN 202210124897 A CN202210124897 A CN 202210124897A CN 114515337 A CN114515337 A CN 114515337A
Authority
CN
China
Prior art keywords
nadph oxidase
inhibiting
substance
vascular dysfunction
activity
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
CN202210124897.3A
Other languages
Chinese (zh)
Inventor
孙崇秀
张代民
肖平喜
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Nanjing Medical University
Original Assignee
Nanjing Medical University
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Nanjing Medical University filed Critical Nanjing Medical University
Priority to CN202210124897.3A priority Critical patent/CN114515337A/en
Publication of CN114515337A publication Critical patent/CN114515337A/en
Pending legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/14Vasoprotectives; Antihaemorrhoidals; Drugs for varicose therapy; Capillary stabilisers

Abstract

In order to solve the problem that the endothelial dependent arterial vasodilation function is damaged due to rapamycin drugs, the invention provides the application of NADPH oxidase 2 as a therapeutic target in the preparation of drugs for treating vascular dysfunction. And provides the application of the substance inhibiting the gene expression of NADPH oxidase 2 or the substance inhibiting the activity of NADPH oxidase 2 in the preparation of the drugs for treating vascular dysfunction. By inhibiting NADPH oxidase 2 activity or Nox2 gene expression or inhibiting the activities of EC cell p38 kinase and JNK1/2 kinase to inhibit Nox2 gene expression and activity, the method can reduce the level of active oxygen in blood, improve NO level and improve the function of mTORC2 in inhibiting the damaged arterial endothelium-dependent vasodilation, thereby reducing the side effect of rapamycin medicaments and enhancing the treatment effect, or can be applied to design new mTOR inhibiting medicaments with better curative effect and lower side effect. The invention also provides the use of a substance that reduces the amount of ROS in the blood and increases the amount of NO in the blood for the manufacture of a medicament for the treatment of vascular dysfunction.

Description

Application of NADPH oxidase 2 as therapeutic target in preparation of medicine for treating vascular dysfunction
Technical Field
The invention belongs to the technical field of biological medicines, and particularly relates to application of NADPH oxidase 2 as a therapeutic target in preparation of a medicine for treating vascular dysfunction.
Background
In China all around, the morbidity and mortality of cardiovascular diseases are the first, higher than those of tumors and other diseases, and seriously harm human health. The most common cardiovascular disease is coronary heart disease, which is a disease caused by myocardial ischemia, hypoxia and even necrosis due to the formation of atheromatous plaques in the blood vessels of the coronary arteries (coronary arteries) of the heart, resulting in narrowing or obstruction of the coronary lumens. The coronary stent implantation developed at the end of the last century is to implant a metal stent in a narrow coronary segment and to open blood vessels to restore blood circulation, and is the most effective means for treating coronary heart disease at present. However, when the traditional metal bare stent is implanted, the proliferation and migration of vascular smooth muscle cells can be caused, so that the incidence of restenosis of a vascular cavity after the stent is implanted is high. Thus, drug-coated stents (DES) have evolved as the primary means of treating coronary heart disease since the beginning of this century. Compared with the traditional metal bare stent, the DES can inhibit the proliferation and migration of vascular smooth muscle cells by locally releasing anti-cell proliferation drugs, and can obviously reduce the incidence of vascular restenosis after stent implantation. Currently, the coating drugs of DES are mainly mTOR inhibitor rapamycin (rapamycin, also known as sirolimus) and its derivative everolimus (everolimus), etc. mTOR is involved in the formation of two complexes in mammals, complex 1(mTORC1) and complex 2(mTORC 2). Wherein mTORC1 is formed by mTOR, Raptor and the like, and regulates protein translation and other processes by phosphorylating downstream substrate protein S6 kinase (S6K) and eukaryotic translation initiation factor 4E-binding protein 1(4E-BP 1). mTORC2 is formed by mTOR and Rictor, etc., and influences cytoskeletal recombination and cell survival by regulating the activities of downstream AKT and PKC α. Rapamycin can rapidly inhibit mTORC1, and prolonged (e.g., 24-hour) treatment also significantly inhibits mTORC 2. In addition to being used as a stent coating drug, rapamycin is also widely used in clinical antitumor therapy and in resistance to rejection after organ transplantation. Although widely used in clinics, many international large clinical studies have shown that the use of rapamycin results in impaired endothelial-dependent vasodilation (EDD) function.
Therefore, there is a need to find a drug capable of treating and/or preventing vascular dysfunction.
Disclosure of Invention
In order to solve the technical problems in the prior art, the application provides the application of NADPH oxidase 2(Nox2) as a therapeutic target in the preparation of drugs for treating and/or preventing vascular dysfunction.
The first object of the present application is to provide the use of a substance inhibiting the expression of NADPH oxidase 2 gene for the preparation of a medicament for the treatment and/or prevention of vascular dysfunction.
Further, the substance can reduce the content of Reactive Oxygen Species (ROS) in blood, especially in arterial blood vessels, and inhibit eNOS uncoupling, thereby increasing the content of Nitric Oxide (NO) in blood, especially in arterial blood vessels, by inhibiting NADPH oxidase 2 gene expression, thereby treating and/or preventing vascular dysfunction.
Further, the vascular dysfunction is caused by mTOR inhibitor drugs.
Further, the mTOR inhibitor drug is rapamycin.
A second object of the present application is to provide the use of a substance inhibiting NADPH oxidase 2 activity for the preparation of a medicament for the treatment and/or prevention of vascular dysfunctions.
Further, the agent inhibits NADPH oxidase 2 activity by decreasing p38 kinase and/or JNK1/2 kinase activity of endothelial cells.
Further, the substance can reduce the content of Reactive Oxygen Species (ROS) in blood, especially in arterial blood vessels, by inhibiting NADPH oxidase 2 activity, and inhibit eNOS uncoupling, thereby increasing the content of Nitric Oxide (NO) in blood, especially in arterial blood vessels, to thereby treat and/or prevent vascular dysfunction.
In a particular embodiment, the substance inhibiting NADPH oxidase 2 activity is the inhibitor SB203580 or SP 600125.
Further, the vascular dysfunction is caused by mTOR inhibitor drugs.
Further, the mTOR inhibitor drug is rapamycin.
The third purpose of the invention is to provide the application of the substance for reducing the content of active oxygen in blood in preparing the medicine for treating and/or preventing vascular dysfunction.
The substance can reduce active oxygen content in blood, inhibit eNOS uncoupling, increase nitric oxide content in blood, and treat vascular dysfunction.
Further, the vascular dysfunction is caused by mTOR inhibitor drugs.
Further, the mTOR inhibitor drug is rapamycin.
The fourth purpose of the invention is to provide the application of NADPH oxidase 2 as a therapeutic target in the preparation of medicines for treating and/or preventing vascular dysfunction.
Further, the vascular dysfunction is caused by mTOR inhibitor drugs.
Further, the mTOR inhibitor drug is rapamycin.
It is a fifth object of the present invention to provide a medicament for treating and/or preventing vascular dysfunction, comprising a substance inhibiting the expression of NADPH oxidase 2 gene, or a substance inhibiting the activity of NADPH oxidase 2.
Further, the vascular dysfunction is caused by mTOR inhibitor drugs.
Further, the mTOR inhibitor drug is rapamycin.
Compared with the prior art, the technical scheme of the application has the following beneficial effects:
the application discovers for the first time that the kinases of MAPK family members p38 and JNK1/22 are activated in Endothelial Cells (EC) when mTORC2 is inhibited due to rapamycin drugs or other factors. These MAPK family members up-regulate NADPH oxidase 2(Nox2) expression, thereby promoting the uncoupling of ROS accumulation in EC from eNOS, reducing the NO levels that EC generates and releases into the blood vessels, and ultimately leading to impaired arterial endothelium-dependent vasodilatory EDD function. Therefore, the application firstly proposes that the Nox2 can be used as a therapeutic target point in the preparation of medicines for treating vascular dysfunction.
The substance can reduce the level of ROS in blood and inhibit the uncoupling of eNOS by inhibiting the expression of Nox2 gene or inhibiting the activities of EC cell p38 kinase and JNK1/2 kinase to inhibit the activity of Nox2, thereby improving the level of NO in blood, especially in arterial blood vessels, and improving the inhibition of the damaged arterial endothelial dependent vasodilation function (EDD) by mTORC2, thereby reducing the side effect and enhancing the treatment effect of mTOR inhibitor medicines such as rapamycin, or being applied to design new mTOR inhibitor medicines such as rapamycin with better curative effect and lower side effect.
Drawings
FIG. 1 aortic EDD and EID function in mice after intraperitoneal injection of rapamycin. Wherein FIG. 1A is a percutaneous ultrasound detection vasomotor function image; figure 1B is a histogram of mouse EDD function; FIG. 1C is a histogram of mouse EID function; *: p <0.05 between the two groups.
FIG. 2 shows the EDD function of mouse aorta after knockout of Mtor gene in EC and of Rictor gene in EC. Wherein FIG. 2A is MtorEC-/-The EDD function and the percutaneous ultrasonic detection of the mice are combined to obtain an image of the vasomotor function; FIG. 2B shows RictorEC-/-The EDD function and the percutaneous ultrasonic detection of the mice are combined to obtain an image of the vasomotor function; *: p between two groups<0.05; **: p between two groups<0.01。
Figure 3 EDD function of mouse ex vivo vascular ring after inhibition of mTOR or mTORC 2. Wherein FIG. 3A is MtorEC-/-Group mouse EDD function; FIG. 3B shows RictorEC-/-Group mice EDD function. (ii) a *: p between two groups<0.05; **: p between two groups<0.01; ***: p between two groups<0.001。
Figure 4 mouse serum NO levels after mTOR or mTORC2 inhibition. Wherein FIG. 4A is a schematic diagram of a color development method for detecting a wild-type control group and an MtorEC-/-Group mice serum NO level histogram; FIGS. 4B-C are EPR assays for detection of wild-type control and MtorEC-/-Histograms and representative graphs of serum NO levels in the group of mice; FIG. 4D is a color development method for detecting wild type control group and RictorEC-/-Group mice serum NO level histogram; FIGS. 4E-F are EPR assays for wild-type control and RictorEC-/-Histograms and representative graphs of serum NO levels in the group of mice; (ii) a *: p between two groups<0.05; **: p between two groups<0.01. A and D, color development; B-C and E-F, EPR method.
Fig. 5 effect on ROS after inhibition of mTORC 2. Wherein FIG. 5A is a flow cytometry measurement of ROS levels after 1 hour rapamycin treatment with human aortic EC; FIG. 5B shows ROS levels measured after treating human aortic EC for toren 1 hours FIG. 5C shows ROS levels measured after treating human aortic EC for 48 hours with rapamycin; FIG. 5D is a graph showing the measurement of ROS levels in serum of mice intraperitoneally injected with rapamycin by chemiluminescence; FIG. 5E is MtoreC-/-group mice serum ROS levels; FIG. 5F is serum ROS levels in Rictor EC-/-group mice; FIG. 5G shows the detection of BH4 levels by ELISA after treating human aortic EC with torin; *: p <0.05 from control; **: p <0.01 with the control group.
Figure 6, effect on ROS accumulation and NO production barrier due to mTORC2 inhibition after Nox2 inhibition. Wherein FIG. 6A is the mRNA level of Nox2 detected after 10nM torein treatment of human aortic EC (1 hour); FIG. 6B shows protein levels of Nox2 detected after tropin treatment of human aortic EC; FIG. 6C shows ROS scavenging reagent acetylcysteine (N-acetylcysteine, NAC, 2mM) or Nox inhibitor Apocynin (Apo, 20M) pre-treated human aortic EC, treated with torin, and then flow cytometric ROS levels were measured; FIG. 6D shows NAC or Apo pretreatment of human aortic EC followed by torin treatment and chromogenic NO level detection; *: p <0.05 from control.
Figure 7 inhibition of mTORC2 activation of MAPK, up-regulating Nox2 expression and ROS production. Wherein FIG. 7A is a graph showing the phosphorylation of p38 and JNK1/2 after 1 hour of rapamycin or torein treatment of human aortic EC; FIGS. 7B-C show the pretreatment of human aortic EC with p38 or JNK1/2 inhibitors SB203580(SB) or SP600125(SP), respectively, followed by torin treatment to detect mRNA (B) or protein (C) expression of Nox 2; FIG. 7D shows Apo pretreatment of human aortic EC followed by torin treatment for phosphorylation of p38 and JNK 1/2; FIG. 7E shows pretreatment of human aortic EC with SB or SP followed by torren treatment followed by flow cytometry for ROS levels; FIG. 7F shows pretreatment of human aorta EC with SB or SP, followed by torin treatment and then chromogenic NO level detection; *: p <0.05 from control.
Figure 8, effect of knock-down mouse EC Nox2 gene on ROS accumulation and NO production dysfunction due to mTORC2 inhibition. Wherein FIG. 8A shows that AAV-shNox2 significantly knockdown the expression level of Nox2 in mouse primary EC in comparison with AAV empty vector (AAV-GFP); FIG. 8B shows the ROS content in serum of endothelial Rictor knockout mice injected with AAV-GFP or AAV-shNox 2; FIG. 8C shows the NO content in serum of endothelial Rictor knockout mice injected with AAV-GFP or AAV-shNox 2; *: p <0.05 between the two groups.
Figure 9, effect of knock-down mouse EC Nox2 gene on EDD function impairment due to mTORC2 inhibition. Wherein FIG. 9A is a histogram of EDD function in mice injected with AAV-GFP or the same dose of AAV-shNox2 vector; FIG. 9B is a histogram of EID function in mice injected with AAV-GFP or AAV-shNox2 vector at the same dose; **: p <0.01 between the two groups.
Detailed Description
Example 1 intraperitoneal injection of rapamycin to impair aortic EDD function in mice
The C57BL/6 background mice were randomly divided into control and treatment groups. The control mice were intraperitoneally injected with corn oil and the treatment mice were intraperitoneally injected with rapamycin at a dose of 2mg/kg body weight, which was similar to the blood levels achieved with clinical rapamycin. After the mice were anesthetized with 1% pentobarbital sodium (60mg/kg) by intraperitoneal injection, they were supine, shaved on the abdomen, smeared with a coupling agent, and examined for the end-diastolic inner diameter of the abdominal aorta with an ultrasonic instrument (Vevo 770). During the measurement, the ultrasound probe is kept at the same position, and the measurement average values of 3 cardiac cycles are taken respectively. First, the internal diameter of the abdominal aorta in the basal state (denoted as D) is measured0) Then, the right lower limb of the mouse was cuffed with a cuff, and after 2 minutes of pressurization of the sphygmomanometer to 200mmHg, rapid cuff release resulted in blood flow changes, thereby inducing vasodilation, which is dependent on EC. Abdominal aortic inner diameter (D) was recorded 1 minute after cuff release1) Calculating Endothelial (EC) -dependent vasodilation (EDD) ═ D1-D0)/D0X 100%. After resting for 15 minutes, the mice were sublingually administered 500. mu.g nitroglycerin, and after 3 minutes the abdominal aorta internal diameter (D) was recorded2). Since vasodilation by nitroglycerin is not dependent on EC, EC independent vasodilation (EID) ═ D (D) can be calculated2-D0)/D0X 100%. The results (fig. 1) show that intraperitoneal rapamycin can reduce EDD by 31.9%, while having no effect on EID. This is consistent with the finding that clinical use of rapamycin drugs leads to EDD dysfunction.
Example 2 knock-out of Mtor gene in EC to inhibit mTOR, or Rictor gene to inhibit mTORC2, all impaired aortic EDD function in mice
Mtorflox/floxAnd Cdh5CreERT2After the mice are hybridized, an EC-specific Mtor gene knockout mouse (Mtor) is finally obtainedEC-/-) And its control (WT) mice. Also, Rictorflox/floxAnd Cdh5CreERT2After the mice are hybridized, the EC-specific Rictor gene knockout mice (Rictor) are finally obtainedEC-/-) And its control mice. Selecting Mtor for about 6 weeksEC-/-And RictorEC-/-And wild-type mice of the same age and sex, were subjected to intraperitoneal injection for one week for tamoxifen (tamoxifen) to induce EC-specific gene knockout. The vasomotor function of the percutaneous ultrasonic detection is the same as above.
The results (fig. 2) show that knockout of the Mtor gene in EC to inhibit Mtor, or knockout of Rictor in EC to inhibit mTORC2, both impaired aortic EDD function in mice, i.e. deletion of Mtor or Rictor in EC both significantly reduced EDD by 33.9% and 41.1%, respectively. This suggests that mTORC2 is involved in maintaining normal arterial EDD function, and that the impairment of EDD function by rapamycin drugs is due, at least in part, to the inhibition of mTORC2 in EC.
Example 3 inhibition of both mTOR or mTORC2 significantly reduced EDD function of the ex vivo vascular ring
Mtor treated from example 2EC-/-Or RictorEC-/-And its control mice isolated aortic vascular rings at 10-9~10-5Separately, EDD was induced by acetylcholine (Ach) at a ten-fold gradient in mol/L concentration and detected by an isolated angiotensiometer (Myogaph). The results (fig. 3) show that both mTOR and Rictor deletion in EC significantly reduced the EDD level of aortic vascular rings, which is consistent with the above in vivo experimental results, further supporting the conclusion that the above mTOR inhibiting drugs impair EDD function by inhibiting mTORC 2.
Example 4 inhibition of mTOR or mTORC2 significantly reduced serum NO levels in mice
MtorEC-/-And RictorEC-/-The mouse and its control wild-type mouse are bred as described in example 2, Mtorflox/floxAnd Cdh5CreERT2After the mice are hybridized, an EC specific Mtor gene knockout mouse is obtained(MtorEC-/-) And its control (WT) mice. Also, Rictorflox/floxAnd Cdh5CreERT2After mouse hybridization, EC-specific Rictor knockout mice (Rictor) were obtainedEC-/-) And wild type control mice of the same age and sex. Selecting Mtor for about 6 weeksEC-/-And RictorEC-/-EC-specific gene knockout was induced by intraperitoneal injection of tamoxifen for one week in mice and wild-type mice of the same age and sex. Separating serum after heart blood sampling, using kit (A012-1, Nanjing) to colorimetry (figure 4) to determine NO in mouse serum, and result shows MtorEC-/-And RictorEC-/-The NO in the mouse serum is reduced by 28.8 percent and 45.8 percent respectively. The EPR signal of cPTIO was measured with an EPR meter (A300-10/12) and also showed 21.6% and 38.1% reduction in NO. This suggests that mTORC2 is involved in maintaining NO levels and mTOR inhibitor drugs inhibit NO production by inhibiting mTORC2 in EC.
Example 5 inhibition of mTORC2 results in increased ROS production decoupled from eNOS
Flow cytometry detection of ROS levels in human aortic EC after DHE staining showed: rapamycin treatment for a short time (1nM, 1 hour) (conditions known to inhibit mTORC1 only) did not affect ROS levels in human aortic EC (fig. 5A), but another mTOR inhibitor, toren (10nM,1 hour), inhibited mTORC2, promoting ROS accumulation (fig. 5B) rapamycin treatment for a long time (. gtoreq.24 hours) was also known to inhibit mTORC 2. Treatment with pactamycin (1nM) for 48 hours also increased ROS levels in EC (fig. 5C). Similarly, ROS in serum were detected with the chemiluminescence probe lucigen (lucigenin), and it was found that mice intraperitoneally injected with rapamycin (FIG. 5D), or knockout of Mtor or Rictor genes in EC all resulted in elevated ROS in serum (FIGS. 5E-F). The kit (JL15170, Jianglai, Shanghai) is used for detecting the cell BH4 level by an ELISA method, and the result shows that the short-time treatment of rapamycin does not affect the BH4 level, and the torein treatment increases the BH4 level. These results indicate that inhibition of mTORC2 results in increased ROS production and eNOS decoupling.
Example 6 upregulation of expression of Nox2 and increased ROS production following inhibition of mTORC2
The Nox family is an important enzyme that catalyzes the generation of ROS. Among all Nox family members, the Nox2 enzyme complex is expressed at the highest level in EC. Results of fluorescent quantitative PCR measurements after 1 hour of torin treatment of human aortic EC (purchased from ATCC in usa) showed an increase in mRNA of the core subunit of the Nox2 enzyme complex (also named Nox2), Western blots also showed an increase in Nox2 protein expression (fig. 6A-B), while short time (1 hour) of rapamycin treatment of human aortic EC did not affect Nox2 expression (results not shown). Human aorta EC was pretreated with 2mM nonspecific ROS scavenging reagent acetylcysteine (N-acetylcysteine, NAC) or 20M Nox enzyme inhibitor apocynin (apo) for 1 hours, followed by flow cytometry for ROS and colorimetric detection of NO, showing that both pretreatments were effective in reversing ROS accumulation caused by mTORC2 inhibition (fig. 6C) and improving NO levels (fig. 6D). These results indicate that inhibition of Nox2 or direct scavenging of ROS can improve inhibition of ROS accumulation and NO production disorders caused by mTOR-like drugs.
Example 7 inhibition of ROS accumulation and NO production disorders caused by mTOR drugs by Nox2 Activity inhibitors
Western blot analysis after treatment of human aortic EC1 hours with rapamycin (1nM) or orin (10nM) showed: rapamycin treatment did not significantly affect phosphorylation of p38 and JNK1/2, whereas torein treatment inhibited phosphorylation of mTORC2 significantly (fig. 7A). Pretreatment of human aorta EC1 with 20M Nox2 enzyme inhibitor apocynin (apo) inhibited Nox2 and also p38 and JNK1/2 phosphorylation (fig. 7C).
Fluorescence quantitative PCR and Western blot detection of human aortas pretreated with 20 μ M p38 inhibitor SB203580 or 20 μ M JNK1/2 inhibitor SP600125 for 1 hours followed by 10nM of torin for 1 hour showed: these pretreatments significantly reduced mTORC2 inhibition induced mRNA and protein level expression of the Nox2 subunit (fig. 7B-C); flow cytometry showed that these pretreatments effectively reversed ROS accumulation resulting from mTORC2 inhibition (fig. 7D), improving NO levels (fig. 7E). These results indicate that after mTORC2 is inhibited, MAPK is activated, and inhibition of MAPK members p38 or JNK1/2 can inhibit Nox2 subunit expression and Nox2 enzyme complex activity, thereby improving inhibition of ROS accumulation and NO production disorder caused by mTOR-like drugs.
Example 8 knock-down of the Nox2 Gene in mouse EC significantly reverses ROS accumulation and NO production disorders
An adeno-associated virus AAV vector (AAV-shNox2) was constructed by GmbH, Shanghai, to knock down the mouse Nox2 gene (also known as Cybb gene, ID: 13058). In the vector, two sequences (5'-GCUGCCAGUGUGUCGAAAUTT-3', 5'-CCUAUGUUCCUGUACCUUUTT-3') aiming at mouse Nox2 gene are fused with an Enhanced Green Fluorescent Protein (EGFP) and a tag protein Flag coding sequence. An EC-specific ICAM2 promoter sequence is inserted upstream of the sequences so as to achieve the aim of specifically knocking out the Nox2 in EC. The AAV vector was demonstrated to efficiently knock down Nox2 gene expression in mouse primary EC (fig. 8A).
The endothelial Rictor knockout mice obtained in example 2 were randomly divided into control and treatment groups. Control group was injected intravenously 3X 10 per mouse tail11The treatment group was injected with the same dose of AAV-shNox2 vector at vg doses of AAV empty vector (AAV-GFP). ROS were detected in serum with lucigenin, and the results showed a significant reduction in ROS in serum of mice injected with AAV-shNox2 (FIG. 8B). The amount of NO was measured by a color development method, and the results showed that the level of NO in serum of mice injected with AAV-shinox 2 was significantly increased (fig. 8C), confirming that inhibition of expression of the Nox2 gene was effective in improving the level of NO decreased by mTORC2 inhibition by reducing ROS production.
Example 9 knock-down of the Nox2 Gene in mouse EC alleviates EDD functional impairment
The experiment 9 shows that the mice injected with AAV-shNox2 have obviously improved blood flow-mediated EDD function (figure 9A) by using percutaneous ultrasonic detection of vasomotor function of the control and treatment group mice, and proves that the inhibition of the expression of the Nox2 gene can improve the inhibition of the injured EDD function of mTORC 2. EID of mice injected with AAV-shNox2 is also increased in FIG. 9B), and further, it is confirmed that inhibition of Nox2 can treat and/or prevent vascular dysfunction and improve vasodilation function.
Sequence listing
<110> Nanjing university of medical science
Application of <120> NADPH oxidase 2 as treatment target in preparation of medicine for treating vascular dysfunction
<160> 2
<170> SIPOSequenceListing 1.0
<210> 1
<211> 21
<212> DNA/RNA
<213> Artificial Sequence (Artificial Sequence)
<400> 1
gcugccagug ugucgaaaut t 21
<210> 2
<211> 21
<212> DNA/RNA
<213> Artificial Sequence (Artificial Sequence)
<400> 2
ccuauguucc uguaccuuut t 21

Claims (9)

1. Use of a substance inhibiting the expression of the NADPH oxidase-2 gene for the preparation of a medicament for the treatment and/or prevention of vascular dysfunctions.
2. Use of a substance inhibiting the activity of NADPH oxidase 2 for the preparation of a medicament for the treatment and/or prevention of vascular dysfunctions.
3. Use according to claim 2, characterized in that the substance inhibits NADPH oxidase 2 activity by decreasing the p38 kinase and/or JNK1/2 kinase activity of endothelial cells.
4. The use according to claim 1 or 2, wherein the substance is capable of treating vascular dysfunction by inhibiting NADPH oxidase 2 gene expression or inhibiting NADPH oxidase 2 activity, thereby decreasing the level of reactive oxygen species in the blood, inhibiting eNOS uncoupling, increasing the level of nitric oxide in the blood.
5. The use of a substance which reduces the active oxygen content in blood for the manufacture of a medicament for the treatment of vascular dysfunction.
Application of NADPH oxidase 2 as a therapeutic target in preparation of drugs for treating and/or preventing vascular dysfunction.
7. A therapeutic and/or prophylactic agent for vascular dysfunction, characterized by comprising a substance that inhibits the expression of NADPH oxidase 2 gene, or a substance that inhibits the activity of NADPH oxidase 2.
8. The use according to any one of claims 1, 2, 5, 6 or 7, wherein the vascular dysfunction is caused by an mTOR inhibitor drug.
9. The use of claim 7, wherein the mTOR inhibitor drug is rapamycin.
CN202210124897.3A 2022-02-10 2022-02-10 Application of NADPH oxidase 2 as therapeutic target in preparation of medicine for treating vascular dysfunction Pending CN114515337A (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202210124897.3A CN114515337A (en) 2022-02-10 2022-02-10 Application of NADPH oxidase 2 as therapeutic target in preparation of medicine for treating vascular dysfunction

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202210124897.3A CN114515337A (en) 2022-02-10 2022-02-10 Application of NADPH oxidase 2 as therapeutic target in preparation of medicine for treating vascular dysfunction

Publications (1)

Publication Number Publication Date
CN114515337A true CN114515337A (en) 2022-05-20

Family

ID=81596478

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202210124897.3A Pending CN114515337A (en) 2022-02-10 2022-02-10 Application of NADPH oxidase 2 as therapeutic target in preparation of medicine for treating vascular dysfunction

Country Status (1)

Country Link
CN (1) CN114515337A (en)

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20120316100A1 (en) * 2011-05-17 2012-12-13 University Of Pittsburgh - Of The Commonwealth System Of Higher Education Therapeutic Use of Peptide Inhibitors of NADPH Oxidase; Aerosolization as a Delivery Mechanism
US20140194422A1 (en) * 2011-06-13 2014-07-10 Emory University Piperazine derivatives, compositions, and uses related thereto
CN112336723A (en) * 2020-12-11 2021-02-09 江南大学 Method for preparing medicament for reducing coupling degree of TRPV4 and NOX2

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20120316100A1 (en) * 2011-05-17 2012-12-13 University Of Pittsburgh - Of The Commonwealth System Of Higher Education Therapeutic Use of Peptide Inhibitors of NADPH Oxidase; Aerosolization as a Delivery Mechanism
US20140194422A1 (en) * 2011-06-13 2014-07-10 Emory University Piperazine derivatives, compositions, and uses related thereto
CN112336723A (en) * 2020-12-11 2021-02-09 江南大学 Method for preparing medicament for reducing coupling degree of TRPV4 and NOX2

Non-Patent Citations (7)

* Cited by examiner, † Cited by third party
Title
C.RIPPE,ET AL: "Short-term calorie restriction reverses vascular endothelial dysfunction in old mice by increasing nitric oxide and reducing oxidative stress", 《AGING CELL》 *
C.RIPPE,ET AL: "Short-term calorie restriction reverses vascular endothelial dysfunction in old mice by increasing nitric oxide and reducing oxidative stress", 《AGING CELL》, vol. 9, 31 December 2010 (2010-12-31), pages 304 - 312 *
JABS ET AL: "Sirolimus-Induced Vascular Dysfunction Increased Mitochondrial and Nicotinamide Adenosine Dinucleotide Phosphate Oxidase-Dependent Superoxide Production and Decreased Vascular Nitric Oxide Formation", JACC, vol. 51, no. 22, pages 2130 - 2138, XP029652126, DOI: 10.1016/j.jacc.2008.01.058 *
吕明: "探讨NADPH 氧化酶NOX家族与高血压的关系", 《医学食疗与健康》 *
吕明: "探讨NADPH 氧化酶NOX家族与高血压的关系", 《医学食疗与健康》, 31 December 2019 (2019-12-31), pages 195 - 198 *
李琛等: "视黄酸通过TLR4/NF-κB通路对脂多糖诱导血管炎症与氧化应激的影响", 《中国优秀硕士学位论文全文数据库 医药卫生科技辑》 *
李琛等: "视黄酸通过TLR4/NF-κB通路对脂多糖诱导血管炎症与氧化应激的影响", 《中国优秀硕士学位论文全文数据库 医药卫生科技辑》, no. 5, 15 May 2021 (2021-05-15), pages 057 - 302 *

Similar Documents

Publication Publication Date Title
Green et al. The Nox4 inhibitor GKT137831 attenuates hypoxia-induced pulmonary vascular cell proliferation
Shiojima et al. Regulation of cardiac growth and coronary angiogenesis by the Akt/PKB signaling pathway
Lin et al. Expression, distribution and regulation of phosphodiesterase 5
Goureau et al. Increased nitric oxide production in endotoxin-induced uveitis. Reduction of uveitis by an inhibitor of nitric oxide synthase.
Gonçalves et al. Clenbuterol suppresses proteasomal and lysosomal proteolysis and atrophy-related genes in denervated rat soleus muscles independently of Akt
Wei et al. Pioglitazone protected against cardiac hypertrophy via inhibiting AKT/GSK3β and MAPK signaling pathways
Zhou et al. Regulation of cell cycle regulators by SIRT1 contributes to resveratrol-mediated prevention of pulmonary arterial hypertension
US11278549B2 (en) Method of treating obesity
Zhang et al. Triptolide protects rat heart against pressure overload-induced cardiac fibrosis
US20150164901A1 (en) Compounds, compositions and methods for treating or preventing neurodegenerative disorders
Sharma et al. The adiponectin signalling pathway-A therapeutic target for the cardiac complications of type 2 diabetes?
JP2010509258A (en) Use of adenosine antagonists
Wang et al. Cordycepin ameliorates cardiac hypertrophy via activating the AMPKα pathway
US7906308B2 (en) Phosphorylation and regulation of Akt/PKB by the rictor-mTOR complex
US20100173928A1 (en) Phosphorylation and regulation of AKT/PKB by the rictor-mTOR complex
Zhang et al. Beta3-Adrenergic receptor activation alleviates cardiac dysfunction in cardiac hypertrophy by regulating oxidative stress
Merkus et al. Phosphodiesterase 5 inhibition-induced coronary vasodilation is reduced after myocardial infarction
Kralova et al. l‐Arginine Attenuates Cardiac Dysfunction, But Further Down‐Regulates α‐Myosin Heavy Chain Expression in Isoproterenol‐Induced Cardiomyopathy
CN114515337A (en) Application of NADPH oxidase 2 as therapeutic target in preparation of medicine for treating vascular dysfunction
Xiao et al. Sanggenon C protects against pressure overload‑induced cardiac hypertrophy via the calcineurin/NFAT2 pathway
CN109550051B (en) Application of histone demethylase KDM6A inhibitor in preparation of obesity treatment drug
US11147807B2 (en) Pharmaceutical composition containing DUSP1 inhibitor
EP4279489A1 (en) Application of pyrido[1,2-a]pyrimidinone analog
Kuropatkina et al. Ubiquinol ameliorates endothelial dysfunction and increases expression of miRNA-34a in a rat model of pulmonary hypertension
CN111920802A (en) Application of andrographolide in preparing medicine for preventing and treating adult T cell leukemia

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
SE01 Entry into force of request for substantive examination
SE01 Entry into force of request for substantive examination