CN110755619A - Host marker for activation of human herpesvirus lysis replication and application thereof - Google Patents

Host marker for activation of human herpesvirus lysis replication and application thereof Download PDF

Info

Publication number
CN110755619A
CN110755619A CN201810855253.5A CN201810855253A CN110755619A CN 110755619 A CN110755619 A CN 110755619A CN 201810855253 A CN201810855253 A CN 201810855253A CN 110755619 A CN110755619 A CN 110755619A
Authority
CN
China
Prior art keywords
protein
stat6
activation
replication
rta
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
CN201810855253.5A
Other languages
Chinese (zh)
Inventor
蔡启良
顾峰
王冲
朱彩霞
王玉燕
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.)
Fudan University
Original Assignee
Fudan 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 Fudan University filed Critical Fudan University
Priority to CN201810855253.5A priority Critical patent/CN110755619A/en
Publication of CN110755619A publication Critical patent/CN110755619A/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
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/20Antivirals for DNA viruses
    • A61P31/22Antivirals for DNA viruses for herpes viruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/531Production of immunochemical test materials
    • G01N33/532Production of labelled immunochemicals
    • G01N33/533Production of labelled immunochemicals with fluorescent label

Abstract

The invention belongs to the technical field of biomedicine, relates to a human herpes virus lytic replication activation detection marker, and particularly relates to application of Signal transducer and activator of transcription6 (STAT 6) as a human herpes virus lytic replication activation detection marker. The STAT6 protein can be used as a marker for the activation and detection of the splitting and replication of various herpesviruses, can also be used as a common target for diagnosis and treatment of herpesvirus-related tumor diseases, and is further used for preparing products for prevention, detection and treatment of human herpesvirus infection.

Description

Host marker for activation of human herpesvirus lysis replication and application thereof
Technical Field
The invention belongs to the technical field of biomedicine, relates to a human herpes virus lytic replication activation detection marker, and particularly relates to application of Signal transducer and activator of transcription6 (STAT 6) as a human herpes virus lytic replication activation detection marker.
The invention also relates to the use of the STAT6 protein as a marker for antiviral applications, including the use in the preparation of preparations for the prevention, detection and treatment of human herpes virus infections.
Technical Field
According to the report of the world health organization, more than 90% of the world's population is infected by different types of herpes viruses(1)The herpes virus belongs to the herpes virus family, 8 kinds of herpes viruses are known to infect human beings at present and are classified into α, β and gamma three subfamilies according to different infection characteristics, wherein α -herpes viruses comprise herpes simplex virus 1(HSV-1 or HHV-1), herpes simplex virus 2(HSV-1 or HHV-1), varicella zoster virus (VZV or HHV-3), β -herpes viruses comprise human cytomegalovirus (HCMV or HHV-5) and human herpes virus 6(HHV-6), gamma-herpes viruses comprise Epstein Barr virus (EBV or HHV-4) and Kaposi sarcoma herpes virus (KSHV or HHV-8), HSV-1 is a virus which is highly contagious by contact and often causes uncomfortable primary infection symptoms, such as herpes labialis, herpes genitalis, herpes keratidis, eczema, herpes manus and meningitis and the like(2,3). In most cases, HCMV infection establishes a lifelong latent state, manifested by replication arrest, low expression of viral genes, and generally no apparent clinical symptoms(4,5). After the virus establishes latent infection, once the virus is stressed by the outside and the immunity of a host is low, the virus can be quickly activated to cause diseases and even death(6-8). The infection rate of EBV is more than 90%, and the EBV is involved in the occurrence and development of various tumors, including nasopharyngeal carcinoma, Burkitt lymphoma, Hodgkin lymphoma and the like(9,10). KSHV is associated with the development of a variety of tumors, such as Kaposi's sarcoma, primary effusion lymphoma, multicenter Castleman's disease, and the like(11)
Herpes virus infection is generally divided into three stages of primary infection, latent infection and lytic replication infection, and researches show that the virus is activated when being stimulated by the outside or the immune function of an organism is low, so that various diseases and even death are caused. The activation of herpesvirus lytic replication detection relies primarily on the detection of lytic phase viral gene expression, e.g., HSV-1 immediate early gene ICPO, HCMV lytic early gene IE1/2, EBV immediate early gene ZTA, and KSHV immediate early gene RTA. However, no unified index for detecting activation of herpesvirus lytic replication exists at present. Therefore, a common target molecule capable of indicating activation of splitting replication of the herpes virus is found, so that convenience is provided for detection of activation of splitting replication of the herpes virus, and a potential common target is provided for development of a therapeutic preparation for resisting herpes virus infection.
Based on the current state of the prior art, the inventor intends to provide a host-derived herpes virus lytic replication activation detection marker STAT6 protein and application of STAT6 protein as a marker in antiviral aspects, including application in preparation of preparations for prevention, detection and treatment of human herpes virus infection.
References relevant to the present invention are:
1.Kukhanova MK,Korovina AN,Kochetkov SN.2014.Human herpes simplexvirus:life cycle and development of inhibitors.Biochemistry.Biokhimiia 79:1635-1652.
2.Fatahzadeh M,Schwartz RA.2007.Human herpes simplex virusinfections:epidemiology,pathogenesis,symptomatology,diagnosis,andmanagement.Journal of the American Academy of Dermatology 57:737-763;quiz764-736.
3.Steiner I,Benninger F.2013.Update on herpes virus infections of thenervous system.Current neurology and neuroscience reports 13:414.
4.Reddehase MJ,Podlech J,Grzimek NK.2002.Mouse models ofcytomegalovirus latency:overview.Journal of clinical virology:the officialpublication of the Pan American Society for Clinical Virology 25 Suppl 2:S23-36.
5.Slobedman B,Cao JZ,Avdic S,Webster B,McAllery S,Cheung AK,Tan JC,Abendroth A.2010.Human cytomegalovirus latent infection and associated viralgene expression.Future microbiology 5:883-900.
6.Hummel M,Zhang Z,Yan S,DePlaen I,Golia P,Varghese T,Thomas G,Abecassis MI.2001.Allogeneic transplantation induces expression ofcytomegalovirus immediate-early genes in vivo:a model for reactivation fromlatency.J Virol 75:4814-4822.
7.de Maar EF,Verschuuren EA,Harmsen MC,The TH,van SonWJ.2003.Pulmonary involvement during cytomegalovirus infection inimmunosuppressed patients.Transplant infectious disease:an official journalof the Transplantation Society 5:112-120.
8.Balthesen M,Messerle M,Reddehase MJ.1993.Lungs are a major organsite of cytomegalovirus latency and recurrence.J Virol 67:5360-5366.
9.Hislop AD,Taylor GS,Sauce D,Rickinson AB.2007.Cellular responses toviral infection in humans:lessons from Epstein-Barr virus.Annu Rev Immunol25:587-617.
10.Kuppers R.2003.B cells under influence:transformation of B cellsby Epstein-Barr virus.Nature reviews.Immunology 3:801-812.
11.Dupin N,Fisher C,Kellam P,Ariad S,Tulliez M,Franck N,van Marck E,Salmon D,Gorin I,Escande JP,Weiss RA,Alitalo K,Boshoff C.1999.Distribution ofhuman herpesvirus-8latently infected cells in Kaposi’s sarcoma,multicentricCastleman’s disease,and primary effusion lymphoma.Proceedings of the NationalAcademy of Sciences of the United States of America 96:4546-4551.
disclosure of Invention
The present invention aims to provide a marker for detecting activation of human herpesvirus lytic replication based on the current state of the art, and more particularly, to the use of Signal transducer and activator transcription factor 6 (STAT 6) as a marker for detecting activation of human herpesvirus lytic replication.
A further object of the present invention provides the use of said STAT6 protein as a marker for antiviral applications, including the preparation of a preparation for the prevention, detection and treatment of human herpes virus infections.
The invention obtains a target protein STAT6 which is consistently regulated and controlled during herpesvirus lysis replication from the perspective of a host, and experimental results show that the STAT6 protein is obviously reduced in the infection process of Kaposi's Sarcoma Herpesvirus (KSHV), and the STAT6 protein can be used as a marker for the activation and detection of various herpesvirus lysis replication and can also be used as a common target for diagnosis and treatment of herpesvirus-related tumor diseases.
Compared with STAT3 protein belonging to STAT family, which is expressed and reduced in HSV-1 lytic replication infection process, and is up-regulated in HCMV lytic replication infection process, and has no inconsistency such as obvious change in EBV and KSHV lytic replication activation process, the STAT6 protein expression level is consistently regulated by the activation of various human herpesviruses, and can be used as a general marker for the activation detection of various human herpesviruses by the activation of various human herpesviruses;
furthermore, the STAT6 can be used as a basis for preparing anti-human herpesvirus and related tumor disease diagnosis and treatment preparations and small molecule drugs.
The invention has the advantages that:
the invention can overcome the defects that the usual detection of the activation of the splitting and the replication of the herpes virus depends on the detection of the expression of the splitting and the replication activation genes of the virus, the indexes and the methods of different virus detections are different, the cost is high, the operation is complicated, and the like, and the STAT6 which is a target protein consistently regulated and controlled during the splitting and the replication of the herpes virus is obtained from the perspective of host cells, and the STAT6 protein can be used as an effective detection marker of the splitting and the replication of the herpes virus and a target for the treatment of virus and related tumor diseases.
The present invention will be described in further detail with reference to the accompanying drawings.
Drawings
FIG. 1 shows that KSHV lytic replication activation down-regulates STAT6 protein expression levels
In the figure, BJAB is a KSHV negative cell line, and BCBL-1 and BC-3 are KSHV positive cell lines.
FIG. 2 shows the effect of RTA-induced expression on STAT6 and STAT3 protein levels
In the figure, the iSLK-RTA cell is a cell strain for artificially constructing a cell strain in which an RTA gene is subjected to tetracycline Doxycyline (Dox, 2ug/ml) inducible expression, and after the cell strain is treated by the Dox for 0, 6 and 12 hours, the RTA protein with gradually increased expression level and the STAT6 protein with gradually decreased expression level can be seen, but the STAT3 protein expression level has no obvious difference.
FIG. 3 shows that KSHV encodes RTA interacting with STAT6 protein, wherein,
a, picture A: co-immunoprecipitation detects the interaction result of the externally expressed RTA and STAT6,
and B, drawing: co-immunoprecipitation assay KSHV in KSHV positive cells BC-3 treated with TPA and sodium butyrate (T/NB) for 24 hours activated RTA expression and interaction results with STAT 6.
Fig. 4 shows that RTA degrades STAT6 protein via ubiquitin-proteasome pathway, wherein,
a, picture A: the proteasome inhibitor MGl32 can inhibit the influence of the induction of expression of RTA on the degradation of STAT6 protein by TPA and sodium butyrate (T/NB);
and B, drawing: the effect of the RTA ubiquitin ligase activity region RING domin on STAT6 protein levels was removed.
FIG. 5 shows the down-regulation of expression levels of STAT6 and STAT3 proteins upon activation of lytic replication of HSV-1 infected VERO cells.
Figure 6 shows that HCMV infection Mrc5 cell lysis activated down-regulation of STAT6 protein expression and up-regulation of STAT3 expression.
Figure 7 shows that downregulation of STAT6 protein expression after activation of EBV-positive cell lytic replication whereas STAT3 protein expression was unaffected, with DG75 being an EBV-negative cell line and B95.8 and LCL being EBV-positive cell lines.
Detailed Description
Example 1: activation of viral lytic replication in KSHV positive cells downregulating STAT6 protein expression
KSHV negative cells (BJAB), KSHV positive cells (BCBL-1 and BC-3) were all from ATCC deposit, treated with TPA (20ng/ml) and sodium butyrate NaB (3mM) at 37 deg.C with 5% CO2The culture was performed for 24 hours under wet conditions. After centrifugal collection, cells were lysed with RIPA lysate for 30min on ice, centrifuged and boiled for 5min with SDS loading buffer. After SDS-polyacrylamide gel electrophoresis separation, transfer to nitrocellulose membrane, 5% degreaser milk powder blocked at room temperature for 30min, and immunoblot detection of anti-STAT 6(D3H4, Cell Signaling Technology), STAT3(79D7, Cell Signaling Technology), KSHV poison RTA, and intracellular reference protein GADPH (G8140-01, US Biological), respectively. The results show that TPA and NaB treatment hardly affected STAT6 expression in BJAB cells, whereas TPA and NaB induced expression of the KSHV immediate early protein RTA in BCBL-1 and BC-3 cells, indicating that the KSHV virus entered the lytic replication phase and STAT6 protein levels were significantly down-regulated, with no significant change in STAT3 belonging to the STAT family. Indicating that KSHV activation specifically down-regulates expression of STAT6 protein levels.
The specific steps of western blotting are as follows:
1) the scraped cells were transferred to a 15ml centrifuge tube at 2000rpm for 5min, the supernatant was decanted, washed 2 times with PBS buffer, and centrifuged at 2000rpm for 5min at 4 ℃.
2) The cell sediment is cracked by RIPA lysate containing protease inhibitors Pepsatin, Aprotinin, PMSF and Leupepptin for 30min on ice.
3) After cell lysis, the cells were centrifuged at 14500rpm at 4 ℃ for 5min, and the supernatant was collected.
4) And performing SDS-polyacrylamide gel electrophoresis on the supernatant, setting the voltage to be 80V, operating for 30min, adjusting the voltage to be 100V until bromophenol blue reaches the bottom of the separation gel, and stopping the voltage.
5) Assembling a transfer printing interlayer, wherein the assembling sequence is as follows: negative pole, sponge, 2 layers of filter paper, gel, NC membrane, 2 layers of filter paper, sponge, positive pole. And (3) putting the assembled interlayer combination into a transfer printing groove containing a film transfer buffer solution, and performing low-temperature transfer printing for 60min at the voltage of 100V.
6) 5% skimmed milk was sealed and kept at room temperature for 30 min.
7) Washing with TBST lotion for three times (5 min/time), adding the prepared primary antibody into NC membrane, and incubating overnight at 4 deg.C with shaking slowly in shaking table
8) Washing with TBST lotion for three times (5 min/time), and incubating for 1h with prepared fluorescent secondary antibody at room temperature with slow shaking.
9) TBST is washed for three times, 5 min/time, and scanned and developed on a Li-cor double infrared laser color developing instrument.
Example 2: KSHV encodes an immediate early protein RTA degradation STAT6 protein
RTA is known to be a key protein for the transition of KSHV from latency to lytic replication phase, and has the function of E3 ligase, and can participate in the ubiquitination modification process. To investigate whether RTA is involved in STAT6 downregulation. iSLK-RTA cells were treated with 2ug/ml DOX to activate RTA expression, and protein samples were harvested at6 and 12 hours, respectively. The expression of STAT6, STAT3, RTA and an internal reference protein GADPH is detected by western blot, and experimental results show that the expression level of STAT6 protein is reduced along with the increase of the expression of the RTA protein, while the STAT3 protein is not obviously changed. Indicating that KSHV RTA is involved in the degradation of STAT6 protein.
Example 3: interaction of RTA with STAT6 protein
To further explore the relationship between RTA and STAT6 protein, we constructed expression plasmids RTA-myc and FLAG-STAT6 carrying different tags, respectively. RTA-myc and FLAG-STAT6 are transfected into 293 cells singly or together according to the volume ratio of 1: 3 of plasmid and PEI by adopting a PEI transfection reagent method, and the cells are harvested after transfection for 48 h. After cell centrifugation, RIPA lysate is added for ice lysis for 30min, 5% of supernatant is removed after centrifugation to serve as Input, and the remaining supernatant is added with ProteinA/G + IgG to remove non-specific binding. Centrifuging, collecting supernatant, adding anti-myc primary antibody, adding ProteinA/G, rotating and shaking for 2hr, centrifuging, washing beads with RIPA lysate for 4 times, adding SDS protein buffer solution, and boiling for 5 min. Western blot detects the expression of STAT6 with RTA. To further confirm the interaction of RTA with STAT6 protein, we used the KSHV positive wild-type cell strain BC-3 and co-induced activation of RTA expression using TPA and NaB, and STAT6 was detected by immunoblotting using anti-RTA antibodies for co-immunoprecipitation. The above results demonstrate that RTA can interact with STAT6 protein after activation of KSHV lytic replication.
The method comprises the following specific steps of co-immunoprecipitation:
1) the scraped cells were transferred to a 15ml centrifuge tube at 2000rpm for 5min, the supernatant was decanted, washed 2 times with PBS buffer at 4 ℃ at 2000rpm for 5 min.
2) The cell sediment is cracked by RIPA lysate containing protease inhibitors Pepsatin, Aprotinin, PMSF and Leupepptin for 30min on ice.
3) After cell lysis, the cells were centrifuged at 14500rpm at 4 ℃ for 5min, and the supernatant was collected.
4) After protein extraction, 5% of the total protein was taken as Input.
5) To the remaining Protein was added 10. mu.l Protein A/G beads, 1. mu.l IgG1 and incubated on a shaker at 4 ℃ for 1hr to exclude non-specific Protein binding.
6) After incubation of the Protein with Protein A/G and IgG antibodies, the cells were centrifuged at 4000rpm for 30s, and the supernatant was aspirated into a new centrifuge tube. The centrifuged bead pellet was washed three times with TBS, each wash consisting of resuspension in 600. mu.l of TBS and centrifugation at 4000rpm for 30 s. The washed beads were resuspended in 50. mu.l TBS.
7) Adding 1 μ g of antibody of the desired detection protein to the supernatant protein aspirated in (6), and incubating overnight in a shaker at 4 ℃ to allow the antibody to bind to the target protein sufficiently.
8) After the Protein was bound to the antibody overnight, 20. mu.l of Protein A/G was added, and the mixture was incubated on a shaker at 4 ℃ for 2hr to allow the Protein A/G to be sufficiently bound to the antibody.
9) After binding of the beads to the antibody and protein, the beads were centrifuged at 4000rpm for 30s, the supernatant was aspirated off, and the bead pellet was washed three times with TBS, each wash consisting of adding 600. mu.l of TBS for resuspension and centrifuging at 4000rpm for 30 s. The washed beads were resuspended in 50. mu.l of TBS.
10) Add 10. mu.l of 6 × loading Buffer to the resuspended beads of (6) and (9), mix by gentle shaking (to prevent the beads from sticking to the tube wall), cook in a metal bath at 100 ℃ for 5min, centrifuge briefly, and aspirate the supernatant for western blot identification.
Example 4: RTA relies on its RING domain to degrade STAT6 via the ubiquitin-proteasome pathway
Since RTA has E3 ligase activity, to explore whether RTA degrades STAT6 via the ubiquitin-proteasome pathway, we treated BCBL-1 cells that activated RTA induced by TPA/sodium NaB using the proteasome inhibitor MG-132 and immunoblotted to detect expression of RTA, STAT6, and the endoglin GADPH. The results show that the STAT6 protein expression is reduced after the TPA and NaB induce the RTA to be highly expressed. STAT6 protein degradation was reduced by inhibition of proteasome activity with MG-132. To further confirm whether RTA functions via E3 ligase, we constructed a truncation of RTA, i.e., deletion of the RING domain at amino acid 118-207 that functions as an E3 ligase, and then cotransfected the expression plasmids STAT6 with RTA- Δ RING in 293T cells. The results show that the expression level of STAT6 is not significantly changed with the increase of the expression level of RTA- Δ RING. The above results further demonstrate that RTA relies on its E3 ligase activity to degrade STAT6 via the ubiquitin-proteasome pathway.
Example 5: down-regulation of STAT6 protein expression by HSV-1 infected Vero cells
HSV-1 virus was infected with Vero cells at MOI ═ 0.1 and cell samples were collected at 0h, 6h, 12h, 24h and 48h of infection, respectively. A protein immunoblotting method is carried out to detect the changes of STAT6, STAT3 and immediate early protein ICP0 at different time points, and GADPH is used as an internal reference protein. The results show that the virus has obvious lytic replication activation at 12h of HSV-1 infection and leads to the significant reduction of STAT6 protein expression level.
Extraction and infection of HSV-1 virus:
HSV-1 Virus isolation and purification
1) The Vero cells are planted in a 75cm cell culture bottle until the confluency of the cells is 80 percent
2) PBS was washed twice, 10ml serum-free medium was added, HSV-1 virus of 0.01MOI was added, and cultured for 1h at 37 ℃ with 5% CO 2.
3) Washing twice with PBS, adding complete culture medium, and culturing until all cells are diseased.
4) Freeze thawing the pathological cells at-80 deg.C for 2-3 times, centrifuging at 2000rpm for 5min, filtering with 0.45um filter, packaging, and placing in-80 deg.C refrigerator.
HSV-1 Virus infection
1) And (4) spreading the Vero cells to a six-hole plate until the confluency of the cells is 80-90%.
2) The six-well plate culture medium is discarded, washed twice by PBS, added with lml serum-free culture medium premixed with virus, and cultured for 2h at 37 ℃.
3) The medium was discarded and 2ml of complete medium was added to continue the culture. Cell samples were collected at 0h, 6h, 12h, 24h and 48h post virus infection.
Example 6: HCMV infection of Mrc5 cells results in down-regulation of STAT6 protein expression
HCMV virus was infected Mrc5 cells at MOI ═ 3 and cell samples were collected at infection 0h, 24h, 48h and 72h, respectively. Western blotting examined changes in expression levels of STAT6 and STAT3 proteins at different time points, as well as the HCMV immediate early protein IE1/2, GADPH, as an internal control protein. The results show that IE1/2 expression is obviously up-regulated at 24h of HCMV infection, which indicates that the STAT6 protein expression is down-regulated at the same time of HCMV lytic replication activation, but STAT3 expression is obviously up-regulated.
HCMV virus extraction and infection steps:
1) human Foreskin Fibroblasts (HFF) grow to a confluence of 80-90%
2) HFF cells were infected with MOI ═ 0.01HCMV virus.
3) The incubator is continuously cultured for 12-14 days at 37 ℃ until 70% of cells are diseased.
4) The pathological cells were centrifuged at 4000rpm at 4 ℃ for 20min, and the supernatant was split-filled and placed in a-80 freezer for further use.
Mrc5 cells infected by HCMV virus
1) Mrc5 cells were plated in six well plates to a confluence of 80-90%.
2) Six well plates of medium were discarded and washed twice with PBS. Adding lml serum-free medium premixed with virus, and culturing at 37 ℃ for 2 h.
3) The medium was discarded and 2ml of normal medium was added to continue the culture. Cell samples were collected at 0h, 24h, 48h and 72h after virus addition.
Example 7: activation of EBV-positive cells leads to down-regulation of STAT6 protein expression
EBV positive cells B95.8 and LCL and negative cell DG75 were selected as controls, and TPA (20ng/ml) and NaB (3mM) were used to co-induce activation of latent EBV, allowing the virus to move from the latent phase to the lytic replication phase. Western blot detects the expression of STAT6, STAT3, and cleavage phase protein ZTA, GAPDH as a reference. The results show that compared with DG75 of a negative cell, the expression of ZTA (sc-53904, Santa cruz) for entering the lytic replication phase of EBV is up-regulated, and the expression level of STAT6 protein is down-regulated, but the STAT3 protein is not obviously influenced.
Test results show that STAT6 protein expression is down-regulated after activation of herpesviruses KSHV, HSV-1, HCMV and EBV lytic replication, but the influence on STAT3 which is taken as a signal transduction and transcriptional activation factor family is inconsistent, which shows that STAT6 plays an important role in activation of herpesviruses lytic replication and can be used as a uniform index for indicating activation of herpesviruses lytic replication; the invention takes KSHV as a typical representative as a research object, and finds that the key protein RTA for KSHV cleavage, replication and activation can degrade STAT6 by using E3 ligase activity and an ubiquitin-proteasome pathway; the experimental result also shows that the STAT6 can be used as a potential target for resisting herpes virus infection and relevant tumor diseases, and further used for preparing products for preventing, detecting and treating human herpes virus infection.

Claims (6)

  1. Use of the STAT6 protein as a marker for the preparation of a preparation for the prevention, detection and treatment of human herpes virus infections.
  2. 2. Use according to claim 1, characterized in that the STAT6 protein is used for the preparation of a diagnostic target for the detection of the activation of lytic replication of human herpes viruses.
  3. 3. Use according to claim 1, wherein the STAT6 protein is used for the preparation of an antiviral therapeutic agent.
  4. 4. Use according to claim 1 or 3, wherein the STAT6 protein is used for the preparation of small molecule drugs for the treatment of viruses.
  5. 5. The use according to claim 1, wherein said human herpesvirus comprises Kaposi's Sarcoma Herpesvirus (KSHV), herpes simplex virus type 1 (HSV-1), Human Cytomegalovirus (HCMV), Epstein-Barr virus (EBV).
  6. 6. Use according to claim 5, characterized in that the STAT6 protein is used as a universal marker for the detection of the lytic replication activation of human herpesvirus.
CN201810855253.5A 2018-07-27 2018-07-27 Host marker for activation of human herpesvirus lysis replication and application thereof Pending CN110755619A (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN201810855253.5A CN110755619A (en) 2018-07-27 2018-07-27 Host marker for activation of human herpesvirus lysis replication and application thereof

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN201810855253.5A CN110755619A (en) 2018-07-27 2018-07-27 Host marker for activation of human herpesvirus lysis replication and application thereof

Publications (1)

Publication Number Publication Date
CN110755619A true CN110755619A (en) 2020-02-07

Family

ID=69328853

Family Applications (1)

Application Number Title Priority Date Filing Date
CN201810855253.5A Pending CN110755619A (en) 2018-07-27 2018-07-27 Host marker for activation of human herpesvirus lysis replication and application thereof

Country Status (1)

Country Link
CN (1) CN110755619A (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN111041089A (en) * 2020-03-13 2020-04-21 广州微远基因科技有限公司 Application of host marker for COVID-19 infection

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2003026641A2 (en) * 2001-09-22 2003-04-03 University Of Nottingham Modulation of stat activity
CN102191224A (en) * 2010-03-12 2011-09-21 复旦大学 Heavy targeted modification method for herpes simplex viruses and application thereof
CN104427992A (en) * 2012-01-25 2015-03-18 德那翠丝有限公司 Biomarkers and combination therapies using oncolytic virus and immunomodulation
CN104447964A (en) * 2013-09-18 2015-03-25 复旦大学 Vaccine medicine target SIM resisting Kaposi's sarcoma-associated herpesvirus
CN106834543A (en) * 2017-03-01 2017-06-13 复旦大学 Each hypotype quick detection of herpes virus hominis and quantitative reagent and kit

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2003026641A2 (en) * 2001-09-22 2003-04-03 University Of Nottingham Modulation of stat activity
CN102191224A (en) * 2010-03-12 2011-09-21 复旦大学 Heavy targeted modification method for herpes simplex viruses and application thereof
CN104427992A (en) * 2012-01-25 2015-03-18 德那翠丝有限公司 Biomarkers and combination therapies using oncolytic virus and immunomodulation
CN104447964A (en) * 2013-09-18 2015-03-25 复旦大学 Vaccine medicine target SIM resisting Kaposi's sarcoma-associated herpesvirus
CN106834543A (en) * 2017-03-01 2017-06-13 复旦大学 Each hypotype quick detection of herpes virus hominis and quantitative reagent and kit

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
CHONG WANG ET AL: "Nuclear Localization and Cleavage of STAT6 Is Induced by Kaposi"s Sarcoma-Associated Herpesvirus for Viral Latency", 《PLOS PATHOGENS》 *
LISA S PORITZ ET AL: "Impaired IL-4 Phosphorylation of STAT6 in EBV Transformed B-Cells", 《JOURNAL OF SURGICAL RESEARCH》 *
QILIANG CAI ET AL: "Kaposi’s Sarcoma-Associated Herpesvirus Inhibits Interleukin-4-Mediated STAT6 Phosphorylation To Regulate Apoptosis and Maintain Latency", 《JOURNAL OF VIROLOGY》 *
王士杰: "人巨细胞病毒通过STAT3 上调胶质瘤中endocan 表达", 《微生物与感染》 *

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN111041089A (en) * 2020-03-13 2020-04-21 广州微远基因科技有限公司 Application of host marker for COVID-19 infection
CN111041089B (en) * 2020-03-13 2020-06-19 广州微远基因科技有限公司 Application of host marker for COVID-19 infection

Similar Documents

Publication Publication Date Title
Li et al. Epstein-Barr virus lytic reactivation regulation and its pathogenic role in carcinogenesis
Kalejta Tegument proteins of human cytomegalovirus
Flemington Herpesvirus lytic replication and the cell cycle: arresting new developments
Kuny et al. Cyclin-dependent kinase-like function is shared by the beta-and gamma-subset of the conserved herpesvirus protein kinases
Traylen et al. Virus reactivation: a panoramic view in human infections
Hermiston et al. Identification and characterization of the human cytomegalovirus immediate-early region 2 gene that stimulates gene expression from an inducible promoter
Kudoh et al. Reactivation of lytic replication from B cells latently infected with Epstein-Barr virus occurs with high S-phase cyclin-dependent kinase activity while inhibiting cellular DNA replication
Kato et al. Herpes simplex virus 1-encoded protein kinase UL13 phosphorylates viral Us3 protein kinase and regulates nuclear localization of viral envelopment factors UL34 and UL31
Wiebusch et al. Human cytomegalovirus 86-kilodalton IE2 protein blocks cell cycle progression in G1
Hayward Viral interactions with the Notch pathway
Perng et al. The human cytomegalovirus gene UL79 is required for the accumulation of late viral transcripts
Rodriguez et al. Genetic dissection of cell growth arrest functions mediated by the Epstein-Barr virus lytic gene product, Zta
Davido et al. The cyclin-dependent kinase inhibitor roscovitine inhibits the transactivating activity and alters the posttranslational modification of herpes simplex virus type 1 ICP0
Auerochs et al. A reporter system for Epstein-Barr virus (EBV) lytic replication: anti-EBV activity of the broad anti-herpesviral drug artesunate
Noris et al. Cell cycle arrest by human cytomegalovirus 86-kDa IE2 protein resembles premature senescence
Chattopadhyay et al. Immuno-metabolic changes in herpes virus infection
Akhova et al. The neuronal host cell factor-binding protein Zhangfei inhibits herpes simplex virus replication
Mostafa et al. N-terminal phosphorylation sites of herpes simplex virus 1 ICP0 differentially regulate its activities and enhance viral replication
Lutzky et al. Novel approach to the formulation of an Epstein-Barr virus antigen-based nasopharyngeal carcinoma vaccine
Gerada et al. Varicella-zoster virus ORF63 protects human neuronal and keratinocyte cell lines from apoptosis and changes its localization upon apoptosis induction
Ottinger et al. The interaction of the gammaherpesvirus 68 orf73 protein with cellular BET proteins affects the activation of cell cycle promoters
CN110755619A (en) Host marker for activation of human herpesvirus lysis replication and application thereof
Chua et al. p53 and Sp1 cooperate to regulate the expression of epstein–barr viral Zta protein
Shimada et al. Human herpesvirus 6 immediate‐early 2 protein interacts with heterogeneous ribonucleoprotein K and casein kinase 2
Munger et al. Infected cell protein No. 22 is subject to proteolytic cleavage by caspases activated by a mutant that induces apoptosis

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
WD01 Invention patent application deemed withdrawn after publication

Application publication date: 20200207

WD01 Invention patent application deemed withdrawn after publication