Application of PRMT3 protein and method for regulating HIV transcription

文档序号:56823 发布日期:2021-10-01 浏览:12次 中文

阅读说明:本技术 Prmt3蛋白的用途和调控hiv转录的方法 (Application of PRMT3 protein and method for regulating HIV transcription ) 是由 于丹 何玉先 姚开虎 于 2021-07-01 设计创作,主要内容包括:本发明的第一个目的在于提供一种具有调控HIV-1转录的宿主蛋白信息。本发明的另一个目的在于提供一种调控HIV-1转录的方法。本发明采用shRNA和抑制剂分别检测抑制PRMT3对于HIV-1转录的调控作用,并证明两种方式均可有效抑制HIV-1转录,并且发现PRMT3可与HIV-1LTR互作,影响H4R3Me2a水平调控HIV-1转录,为研发干扰HIV-1繁殖的药物提供了多种思路和手段。(The first purpose of the invention is to provide a host protein message for regulating HIV-1 transcription. Another object of the present invention is to provide a method for regulating HIV-1 transcription. The invention adopts shRNA and an inhibitor to respectively detect the regulation and control effect of PRMT3 on HIV-1 transcription, proves that both modes can effectively inhibit HIV-1 transcription, and discovers that PRMT3 can interact with HIV-1LTR to influence the H4R3Me2a level to regulate HIV-1 transcription, thereby providing a plurality of ideas and means for developing medicines for interfering HIV-1 reproduction.)

1. A complex, wherein the complex is formed by binding PRMT3 and HIV-LTR.

2. The complex of claim 1, wherein the HIV-LTR is the LTR region of HIV-1.

3. A drug for inhibiting HIV transcription, which comprises a PRMT3 inhibitor and/or a disruptor comprising a PRMT3 complex with HIV-LTR.

4. The medicament of claim 3, wherein the HIV is HIV-1.

Use of a modulator of PRMT3, or a modulator of the complex of PRMT3 and HIV-LTR, for the preparation of (a) or (b):

(a) agents that modulate HIV transcription;

(b) a formulation that modulates the methylation level of H4R3Me2 a.

6. The use according to claim 5, wherein the HIV is HIV-1.

7. The use of claim 5, wherein the modulation is promotion or inhibition.

8. The use of claim 5, the PRMT3 modulator comprising a PRMT3 inhibitor and a PRMT3 promoter.

9. The use of claim 8, the modulator of the PRMT3 and HIV-LTR complex comprises promoting the binding of PRMT3 to HIV-LTR and/or disrupting the binding of PRMT3 to HIV-LTR.

Use of a modulator of the methylation level of H4R3Me2a for the preparation of a formulation for modulating HIV transcription.

Technical Field

The invention relates to the field of biomedicine, in particular to application of PRMT3 protein and a method for regulating HIV transcription.

Background

Aids is a major infectious disease worldwide, and the Human Immunodeficiency Virus (HIV) has latent properties that once infected can remain latent for life by integrating the HIV genome into the Human body. The HIV transcription regulation mechanism is deeply known and understood, and the discovery of a novel anti-AIDS drug target is helpful for breaking through the bottleneck problem and exploring a potential way for discovering and curing HIV infection. Although scientists have revealed HIV infection of the host and signaling pathways for host immune defense over the years, a number of potential anti-aids targets have been discovered. However, the existing medicines can only kill HIV in active reproduction, and no clinical scheme for controlling latent HIV exists at present, so that the existing medicines are a main obstacle for curing patients. The discovery of the novel drug target is an important prerequisite for the research and development of new anti-AIDS drugs, and a need for discovering a novel potential anti-AIDS drug target from the perspective of basic scientific research is urgent, so that a potential innovative idea is provided for the clinical treatment of AIDS.

Protein arginine methyltransferases (PRMTs) mediate the methylation of a series of protein substrates containing arginine residues, and play a key role in the processes of cancer occurrence, development, invasion and the like. PRMT3 is related to tumors, although PRMT3 belongs to the type I PRMT family, most of the biological functions of PRMT3 and the relationship between the biological functions and tumorigenesis are not clear. Gemcitabine is the primary chemotherapeutic agent for mid-and advanced pancreatic cancer. The abnormal expression of PRMT3 in gemcitabine-resistant pancreatic cancer cells, and the inhibition of PRMT3 may be one of the treatment strategies for improving the gemcitabine sensitivity of pancreatic cancer cells (the research progress of the relationship between arginine methyltransferase and tumor in Chinese clinical medicine, No. 28, No. 2 of 4 months, No. 28 of 2021)

Disclosure of Invention

An object of the present invention is to provide a complex formed by combining PRMT3 with HIV-LTR.

The HIV-LTR is the LTR region of HIV-1.

In another aspect of the invention, a method of modulating HIV transcription is provided.

The modulation includes positive modulation and/or negative modulation. Further comprising, promoting and/or inhibiting regulation of HIV transcription. The modulation is achieved by modulating PRMT3 and/or modulating PRMT3 with the HIV-LTR (Long terminal repeats) complex. The regulation comprises positive regulation and/or negative regulation.

The up-regulation is promoting expression of PRMT3 or increasing PRMT3 activity. The down-regulation is inhibition of expression of PRMT3 or inhibition of activity of PRMT 3.

The aforementioned HIV may be HIV-1.

The invention also provides a medicine for inhibiting HIV transcription or treating AIDS, which comprises a PRMT3 inhibitor. Further, the aforementioned HIV is HIV-1.

Further, the aforementioned PRMT3 inhibitors include, but are not limited to, any agent capable of inhibiting the expression of PRMT3 or inhibiting PRMT3 activity, and further, include, but are not limited to, SGC707 and/or shRNA. Further, the aforementioned drugs are achieved by disrupting PRMT3 and HIV-LTR complex.

Further, the aforementioned medicament may comprise a disrupting and/or hindering agent for the complex of PRMT3 and HIV-LTR.

Further, agents that inhibit PRMT3 expression or PRMT3 activity may be included.

Further, this can be achieved by disrupting the structure of the HIV-LTR, specifically including disrupting the molecular structure, or the nucleic acid sequence.

Further, the disruption and/or inhibition includes disruption of primary or secondary structure, or protein conformation, or nucleic acid sequence, as well as any means capable of disrupting the interaction of the two and agents for effecting the same.

Further, the HIV-LTR is an LTR region of HIV-1, and the LTR region is an HIV-1 promoter region LTR.

Further, the complex is disrupted to inhibit transcription of HIV or to treat AIDS.

The invention also provides the use of a PRMT3 modulator for the preparation of a formulation for modulating HIV transcription.

Further, the aforementioned HIV is HIV-1.

Further, the aforementioned regulation includes positive regulation and/or negative regulation. Further comprising, promoting and/or inhibiting regulation of HIV transcription. The modulation is achieved by modulating PRMT3 and/or modulating PRMT3 and HIV-LTR complex. The regulation comprises positive regulation and/or negative regulation.

Further, the aforementioned transcription includes, but is not limited to, Tat-activated HIV-1 transcription and/or JQ-activated HIV-1 transcription.

Further, the aforementioned transcription does not include Prostratin-activated HIV-1.

Further, the aforementioned PRMT3 modulators include PRMT3 inhibitors and PRMT3 promoters.

Further, the aforementioned PRMT3 inhibitor may be any drug or agent capable of inhibiting the expression or activity of PRMT3, and may be, but is not limited to, SGC707 or shRNA.

Further, the aforementioned regulation can be in any cell, specifically, NH1 cell or Jurkat2D10 cell.

Further, the aforementioned regulation is effected by modulating the binding of PRMT3 to the LTR region of HIV-1.

The modulation is in particular inhibition or destruction and/or hindrance.

Further, any agent that inhibits PRMT3 expression or PRMT3 activity may be included.

Further, the disrupting agent may be achieved by disrupting the structure of the HIV-LTR, including specifically disrupting the molecular structure, or the nucleic acid sequence.

Further, the disruption and/or inhibition includes disruption of primary or secondary structure, or protein conformation, or nucleic acid sequence, or any means capable of disrupting the interaction of the two.

The invention also relates to the use of a regulator of the methylation level of H4R3Me2a for the preparation of a formulation for modulating HIV transcription.

Further, the HIV transcript is that of HIV-1.

Further, modulators of H4R3Me2a methylation level include PRMT3 modulators. Further comprises PRMT3 inhibitor and PRMT3 promoter. The aforementioned PRMT3 inhibitor may be, but is not limited to, any drug or agent capable of inhibiting the expression or activity of PRMT3, and may be, but is not limited to, SGC707 or shRNA.

The invention also comprises the regulation effect of shRNA of PRMT3 on HIV-1 transcription, the effect of the combination of PRMT3 and HIV-1 promoter region LTR on controlling HIV-1 transcription, and the effect of H4R3Me2a level on regulating HIV-1 transcription.

The invention adopts shRNA and an inhibitor to respectively detect the regulation and control effect of PRMT3 on HIV-1 transcription, proves that both modes can effectively inhibit HIV-1 transcription, and discovers that PRMT3 can interact with HIV-1LTR to influence the H4R3Me2a level regulation and control of HIV-1 transcription, thereby providing a plurality of ideas and means for developing medicines for interfering HIV-1 reproduction in the future.

Drawings

FIG. 1 ChIP-q-PCR detects the binding of PRMT3 in HIV-1 LTR.

FIG. 2 detection of Tat-activated transcription levels of HIV-1 by shRNA downregulation of PRMT3 levels in NH1 cells.

FIG. 3 detection of Tat-activated HIV-1 transcript levels by inhibition of PRMT3 activity by inhibitor SGC707 in NH1 cells.

FIG. 4 shows that the inhibitor SGC707 inhibits PRMT3 activity in NH1 cells, and JQ 1-activated transcription level of HIV-1 is detected.

FIG. 5 Prostratin-activated HIV-1 transcript levels were measured by treating cells with the PRMT3 inhibitor SGC707 in conjunction with Prostratin treatment.

FIG. 6 detection of the expression level of PRMT3 in the seed cell line.

Figure 7 treatment of SGC707, a PRMT3 inhibitor, in Jurkat2D10 cells, detected endogenous H4R3Me2a levels.

FIG. 8 Jurkat2D10 cells treated with SGC707, a PRMT3 inhibitor, and examined JQ-1 activated HIV-1 transcription by FACS.

FIG. 9 statistical analysis of the FACS results of FIG. 8.

Detailed Description

The present invention will be described in further detail with reference to the accompanying drawings and embodiments. The following examples are only exemplary and are intended to illustrate the technical solutions of the present invention in further detail, and it should be understood by those skilled in the art that modifications or substitutions to the technical solutions without departing from the spirit and scope of the technical solutions of the present invention should be covered by the claims of the present invention.

The present invention will be described in further detail with reference to the accompanying drawings and embodiments. The following examples are only exemplary and are intended to illustrate the technical solutions of the present invention in further detail, and it should be understood by those skilled in the art that modifications or substitutions to the technical solutions without departing from the spirit and scope of the technical solutions of the present invention should be covered by the claims of the present invention.

Example 1: PRMT3 interacts with the LTR.

Materials (I) and (II)

The beads required for the chromosomal co-immunoprecipitation were purchased from Invitrogen corporation, and the IgG antibody, PRMT3 antibody, were purchased from Cell Signaling Technology corporation.

Second, method results

1) Chromosome co-immunoprecipitation

Transfecting HIV-1LTR plasmid in NH1 cell, transferring to Tat or empty vector as negative control group, fixing cell, breaking DNA by ultrasonic wave to obtain DNA of 500-1000 bp; adding PRMT3 antibody into 600 μ l of supernatant, incubating overnight, adding IgG antibody into 600 μ l of supernatant, and incubating overnight to obtain control group; adding Protein A Dynabeads on the next day and incubating for 3 hours; washing the beads with the DNA product by a Wash buffer, and eluting by an Elution buffer; after overnight decrosslinking, RNA enzyme and protease treatment is carried out, and DNA is purified by using a PCR fragment recovery kit; two pairs of primers (sequences are shown below) were designed to amplify the LTR region and the binding of PRMT3 in the HIV-1LTR region was detected by q-PCR.

Nascent:

F:GTTAGACCAGATCTGAGCCT;(SEQ ID NO:1)

R:GTGGGTTCCCTAGTTAGCCA(SEQ ID NO:2)

Interior:

F:CTCTTTCGAAAGAAGTCGGGG;(SEQ ID NO:3)

R:GAACAACTTTACCGACCGCG(SEQ ID NO:4)

As shown in FIG. 1, PRMT3 was able to bind efficiently to the HIV-1LTR region when HIV-1 transcription was activated after Tat was added. The results show that: HIV-1 transcription can be interfered with by disrupting PRMT3 interaction with LTRs.

Example 2: shRNA construction that down-regulates PRMT3 expression.

Materials (I) and (II)

The primers were synthesized by Beijing Yihuiyuan; the buffer used for annealing is NEB buffer 2; ligase was supplied from NEB T4 ligase; sequencing was done by Tianyihui Inc.; other relevant reagents are commercially available.

Second, method results

1. The shRNA sequence was designed as follows:

shRNA-1F:

CCGGCCTTGTGGTATTAAGCATATACTCGAGTATATGCTTAATACCACAAGG TTTTTG(SEQ ID NO:5)

shRNA-1R:

AATTCAAAAACCTTGTGGTATTAAGCATATACTCGAGTATATGCTTAATACCAC AAGG(SEQ ID NO:6)

shRNA-2F:

CCGGGCAGTGAGTGATGTGAATAAACTCGAGTTTATTCACATCACTCACTGCTT TTTG(SEQ ID NO:7)

shRNA-2R:

AATTCAAAAAGCAGTGAGTGATGTGAATAAACTCGAGTTTATTCACATCACTCA CTGC(SEQ ID NO:8)

2. annealing of primers

1) Primer dilution:

the primer tube was centrifuged at 12000rpm for 2mins, and the mixture was dispensed as 100. mu.M liquid and stored at-20 ℃.

2) Respectively taking 4.5 mu l of each forward primer and reverse primer, adding 1 mu l of NEB buffer2, mixing uniformly, boiling to 100 ℃, and putting the mixture into 100 ℃ boiled water until the temperature is reduced to room temperature.

3. Enzyme digestion

The enzyme digestion system is as follows:

mu.g of plasmid was added with 2ml of digestion buffer, 1. mu.l of restriction enzyme, and sterile water was added to a total of 20. mu.l, and the mixture was digested in a 37 ℃ water bath for 2 hrs.

4. DNA gel electrophoresis purification of fragments

1) Prepare 1% agarose DNA gel.

2) Adding the enzyme digestion product into a 6 Xloading buffer, loading the mixture into a DNA glue hole, and running glue at 120V for 1 h.

3) DNA fragment gel recovery was performed using an Omega gel recovery kit.

5. Connection of

DNA containing shRNA sequences was ligated to the PLKO.1 plasmid by 16 ℃ ligase overnight. The connection system is as follows, plasmid 1 mug; mu.l buffer and 0.5. mu.l ligase were added and sterile water was supplemented to 10. mu.l.

6. Transformation of

Adding 5 μ l of the ligation product into 50 μ l of Escherichia coli competent cells, standing on ice for 30mins, thermally stimulating at 42 ℃ for 45s, standing on ice for 2mins, adding LB culture medium, recovering at 37 ℃ for 1h, and coating LB plate containing ampicillin. After overnight growth, the monoclonals are picked and sequenced to verify that the shRNA sequence is correctly connected to the plasmid.

Example 3: HIV-1 transcription can be inhibited by down-regulating PRMT3 level by shRNA.

Materials (I) and (II)

PEI from Polysciences was used as a transfection reagent; for cell culture, 12-well plates from Corning Inc. were used; DMEM from Sijiu was used as the medium, and serum was purchased from Sijiu; the Luciferase assay kit was purchased from Promega.

Second, method results

1) Transfection

Taking 2 1.5ml EP tubes, respectively adding 50 mu l of serum-free DMEM, adding 1 mu l of plasmid into 1 tube, uniformly mixing, adding 2.5 mu l of PEI into the other tube, uniformly mixing, mixing two EP tube seed liquids, standing at room temperature for 30mins, and dropwise adding into a cell culture plate.

2) Luciferase assay

And (3) collecting cells after 48 hours of transfection, resuspending the cells in each well by using 100 mu l of lysis solution, placing the cells in a refrigerator at minus 80 ℃ for at least 2hrs, taking out the cells, thawing the cells at room temperature, shaking the resuspended cells, taking out 10 mu l of the resuspended cells, placing the resuspended cells in a detection white board, adding 20 mu l of Luciferase substrate, detecting Luciferase signals by using a microplate reader, and counting data.

NH1 cells integrated the HIV-1LTR as the promoter of Luciferase in Hela cell line, and thus the expression of Luciferase was examined to evaluate the transcriptional activity of HIV-1. Two shRNAs targeting different regions of PRMT3 were ligated to PLKO.1 plasmid to give shPRMT3_1 and shPRMT3_ 2. Posttransient transfection of shPRMT3_1 and shPRMT3_2 to NH1 cells reduced PRMT3 level, and determination of HIV-1 transcription level by Luciferase. The experiment was repeated three times, with standard deviations shown using error bars and statistical analysis using a two-tailed test,. p < 0.05; p < 0.05; p < 0.001.

The results are shown in fig. 2, with shscarmble as a negative control; the left 3 columns are background gene transcripts and the right 3 columns are Tat-activated HIV-1 transcripts. The results show that: downregulation of PRMT3 by shRNA significantly inhibited Tat-activated HIV-1 transcription, but did not significantly affect host background gene transcription.

Example 4: inhibition of PRMT3 activity inhibits Tat-activated HIV-1 transcription.

Materials (I) and (II)

PRMT3 inhibitor SGC707 was purchased from TargetMOI.

Second, method results

1) Inhibitor treatment

The cells were collected after treating the cells for 16h with the inhibitor SGC707 PRMT3 at concentrations of 0.0625mM, 0.125mM, 0.25mM and 0.5mM, resuspending the cells in 100 μ l of lysate per well in a freezer at-80 ℃ for at least 2hrs, removing the cells after thawing at room temperature, shaking the resuspension, removing 10 μ l and placing in a test whiteboard, adding 20 μ l Luciferase substrate, measuring the Luciferase signal using a microplate reader, and counting the data. The experiment was repeated three times, with standard deviations shown using error bars and statistical analysis using a two-tailed test,. p < 0.05; p < 0.05; p < 0.001.

The results are shown in FIG. 3, where HIV-1 transcript levels were measured using Luciferase before and after Tat activation in NH1 cells treated with PRMT3 inhibitor. DMSO was negative control.

The results show that: it was found that inhibition of PRMT3 activity by inhibitor SGC707 significantly inhibited Tat-activated HIV-1 transcription.

Example 5: inhibition of PRMT3 activity inhibits JQ-1 activated transcription of HIV-1.

Materials (I) and (II)

JQ-1 was purchased from Sigma.

Second, method results

1) Inhibitor treatment

JQ-1 is a BRD4 inhibitor, which is also an HIV-1 activator, and activates HIV-1 through a Tat-dependent signaling pathway. Cells are treated by using a PRMT3 inhibitor SGC707 for transcription, simultaneously, the cells are treated by using JQ-1 for 16h and then collected, 100 mu l of lysis solution is used for each hole to re-suspend the cells and put into a refrigerator at minus 80 ℃ for at least 2hrs, the cells are taken out and thawed at room temperature, 10 mu l of the re-suspension is taken out and put into a detection white board after shaking, 20 mu l of Luciferase substrate is added, Luciferase signals are detected by using a microplate reader, and data are counted. The experiment was repeated three times, with standard deviations shown using error bars and statistical analysis using a two-tailed test,. p < 0.05; p < 0.05; p < 0.001.

As shown in FIG. 4, in NH1 cells, HIV-1 transcript levels were measured using Luciferase before and after JQ-1 activation by treatment with PRMT3 inhibitor. DMSO was negative control.

The results show that: treatment of cells with the PRMT3 inhibitor SGC707 for 16h to inhibit PRMT3 activity was effective in inhibiting JQ-1 activated HIV-1 transcription.

Example 6: inhibition of PRMT3 activity did not inhibit Prostratin-activated HIV-1 transcription.

Materials (I) and (II)

Prostratin was purchased from Sigma.

Second, method results

1) Inhibitor treatment

Prostratin activates HIV-1 transcription through the protein kinase C pathway. Treating cells with PRMT3 inhibitor SGC707, treating the cells with Prostratin for 16h, collecting the cells, resuspending the cells in 100 ul of lysis solution per well, placing the cells in a refrigerator at-80 ℃ for at least 2hrs, taking out the cells, thawing the cells at room temperature, shaking the resuspended cells, taking out 10 ul of the resuspended cells, placing the cells in a detection whiteboard, adding 20 ul of Luciferase substrate, detecting Luciferase signals by using a microplate reader, and counting data. The experiment was repeated three times, with standard deviations shown using error bars and statistical analysis using a two-tailed test,. p < 0.05; p < 0.05; p < 0.001.

The results are shown in FIG. 5, where HIV-1 transcript levels were measured using Luciferase before and after Prostratin activation in NH1 cells treated with PRMT3 inhibitor.

The results show that: PRMT3 inhibitors were found not to inhibit Prostratin-activated HIV-1 transcription. The above results suggest that PRMT3 specifically inhibits Tat-activated HIV-1 transcription. It is shown that SGC707 specifically inhibits Tat-activated HIV-1 transcriptional activation and does not affect the basic functional pathways of the host.

Example 7: PRMT3 was expressed in comparable amounts in various cell lines.

Materials (I) and (II)

Western-blot related experimental consumables were purchased from Lamborelide; PRMT3 antibody was purchased from Cell Signaling Technology; the H4R3Me2a antibody was purchased from Invitrogen.

Second, method results

1) Cell culture

Inoculation 105Cells were plated in 6-well plates using DMEM medium supplemented with 10% FBS and 5% CO at 37 ℃2The incubator of (1) is subjected to static culture.

2)Western-blot

a) Preparing 10% SDS polyacrylamide protein gel;

b) after the cells are collected and resuspended by using 1 xSDS loading buffer, the cells are boiled at 100 ℃ for 10 mins; centrifuging at 12000rpm to obtain supernatant;

c) after 30mins of 80V glue running, 140V glue running is carried out for 1 h.

d) The film was turned on ice for 2h at 300mA current.

e) 5% skimmed milk was sealed at room temperature for 1 h.

f) Primary antibody was added and incubated overnight at 4 ℃.

g) Secondary antibodies were added 3 times for 15mins each with TBST wash.

h) TBST wash was used 3 times, and the developing enhancing solution was added and the X film was developed in a darkroom.

Results the expression of PRMT3 was detected by WB in various cell lines as shown in fig. 6.

The results show that: PRMT3 was found to be highly expressed in a variety of cells as shown, including HIV-1 naturally infected Jurkat CD4+ T cells.

Example 8: inhibition of PRMT3 activity is effective in down-regulating the methylation levels of endogenous H4R3Me2 a.

H4R3Me2a levels are markers for open chromatin, gene transcriptional activation. According to literature, inhibition of PRMT3 was suggested to down-regulate endogenous H4R3Me2a methylation levels. HIV-1 latent model Jurkat2D10 cells are pseudoviral fusion expressing D2EGFP with an HIV-1Nef protein deletion and expressing a cell line with a Tat protein that attenuates interaction with P-TEFb. Therefore, we treated Jurkat2D10 cells with the PRMT3 inhibitor SGC707 to inhibit PRMT3 activity.

As shown in FIG. 7, it was found that the methylation level of H4R3Me2a endogenous to Jurkat2D10 cells was indeed reduced.

The results show that: PRMT3 may inhibit HIV-1 transcription by down-regulating endogenous H4R3Me2a methylation levels.

Example 9: inhibition of PRMT3 activity in the Jurkat2D10 cell model inhibited HIV-1 transcription.

Materials (I) and (II)

Flow cytometry was purchased from Invitrogen using the required tubes.

Second, method results

1) Cells were cultured using 24-well plates and seeded 104Jurkat2D10 cells (the Jurkat T cell line has HIV-1LTR integrated as GFP promoter, so it has the effect of evaluating HIV-1 transcriptional activity by detecting GFP expression) were cultured overnight with RMPI16410 added with 10% FBS per well.

2) Flow cytometry detection of GFP signals

Cells were harvested by centrifugation at 800g for 5mins 16h after treatment with the PRMT3 inhibitor SGC707, washed once with PBS, loaded onto a flow cytometer and data analyzed using software.

HIV-1 latent model Jurkat2D10 cells are pseudoviral cells that incorporate a deletion in HIV-1Nef protein and express an unstably enhanced green fluorescent protein, and express a cell line that has a protein that attenuates the interaction with P-TEFb. Therefore, we treated Jurkat2D10 cells at a concentration of inhibitor that reduces the level of H4R3Me2a after JQ-1 activation of latent HIV-1 transcription, and treated with PRMT3 inhibitor in Jurkat2D10 cells, FACS detected the level of HIV-1 transcription before and after JQ-1 activation, and found that inhibition of PRMT3 activity inhibited JQ-1 activated HIV-1 transcription (FIG. 8). Figure 8 three independent replicates were statistically analyzed (figure 9). The experiment was repeated three times, with standard deviations shown using error bars and statistical analysis using a two-tailed test, p < 0.05.

As shown in FIG. 9, HIV-1 transcript levels were reduced after JQ-1 activation in Jurkat2D10 cells by treatment with SGC707, a PRMT3 inhibitor.

The results show that: in Jurkat2D10 cells, treatment with PRMT3 inhibitor found that inhibition of PRMT3 activity inhibited JQ-1 activated HIV-1 transcription.

14页详细技术资料下载
上一篇:一种医用注射器针头装配设备
下一篇:OXPHOS抑制剂作为抗肿瘤药物Alisertib增效剂的应用

网友询问留言

已有0条留言

还没有人留言评论。精彩留言会获得点赞!

精彩留言,会给你点赞!