WO2016050203A1 - Use of inhibiting activity of casein kinase 2 for enhancing expression of type i interferon - Google Patents

Use of inhibiting activity of casein kinase 2 for enhancing expression of type i interferon Download PDF

Info

Publication number
WO2016050203A1
WO2016050203A1 PCT/CN2015/091111 CN2015091111W WO2016050203A1 WO 2016050203 A1 WO2016050203 A1 WO 2016050203A1 CN 2015091111 W CN2015091111 W CN 2015091111W WO 2016050203 A1 WO2016050203 A1 WO 2016050203A1
Authority
WO
WIPO (PCT)
Prior art keywords
interferon
type
virus
expression
cells
Prior art date
Application number
PCT/CN2015/091111
Other languages
French (fr)
Chinese (zh)
Inventor
肖晖
杜旻
刘晶华
谢亚栋
钟劲
向禹
Original Assignee
中国科学院上海巴斯德研究所
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 中国科学院上海巴斯德研究所 filed Critical 中国科学院上海巴斯德研究所
Publication of WO2016050203A1 publication Critical patent/WO2016050203A1/en

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • 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
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12PFERMENTATION OR ENZYME-USING PROCESSES TO SYNTHESISE A DESIRED CHEMICAL COMPOUND OR COMPOSITION OR TO SEPARATE OPTICAL ISOMERS FROM A RACEMIC MIXTURE
    • C12P21/00Preparation of peptides or proteins
    • C12P21/02Preparation of peptides or proteins having a known sequence of two or more amino acids, e.g. glutathione
    • 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
    • G01N33/68Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids

Definitions

  • the invention relates to the field of immunotherapy.
  • the invention relates to novel uses of casein kinase 2 inhibitors in promoting the expression of type I interferons and their downstream genes.
  • Type I interferons include IFN ⁇ and IFN ⁇ , which bind to interferon receptors on the cell surface to activate protein kinases Jak1 and Tyk2, and then activate the transcription factors STAT1 or/and STAT2 to induce expression of hundreds of genes (interferon induction) Genes, ISGs), including MX1, MX2, Rsad2/Viperine, CXCL10/IP-10, etc.
  • type I interferons is induced by pattern recognition receptors (PRRs, Pattern Recognition Receptors) and their mediated signaling pathways.
  • PRRs pattern recognition receptors
  • pattern recognition receptors include Toll-like receptors (TLRs), NOD-like receptors (NLRs), RIG-I-like receptors (RLRs) and cells.
  • Cytosolic DNA sensors (CDSs) activate complex signaling pathways (including protein kinase TBK1) by identifying pathway-associated Molecular Patterns (PAMPs) secreted by dead cells in tissues.
  • PAMPs pathway-associated Molecular Patterns
  • IKK ⁇ / ⁇ and MAP kinases activate key transcription factors IRF3, NF ⁇ B and AP-1, induce the expression of type I interferons, cytokines and chemokines, activate natural immune and adaptive immune responses, and implement hosts.
  • Physiological functions such as defense and tissue repair.
  • type I interferon may also cause diseases, especially autoimmune diseases such as systemic lupus erythematosus. Therefore, a negative regulation mechanism regulating type I interferon is often present in the body to avoid abnormal expression of interferon. These regulatory mechanisms often lead to the inability of type I interferons to be expressed efficiently under pathological conditions, leading to various diseases with insufficient immune response. Therefore, how to effectively control the immune-related diseases in the pathological state and break through the mechanism of the body's regulation of type I interferon expression is an important subject in the field of basic and clinical medicine.
  • the invention provides a use of a CK2 inhibitor for promoting expression of a type I interferon.
  • CK2 casein kinase 2
  • IFN ⁇ / ⁇ type I interferon
  • type I interferon induction A pharmaceutical composition for expression of a gene or protein thereof; and/or (ii) a pharmaceutical composition for the preparation of a disease associated with the treatment and/or prevention of type I interferon deficiency.
  • the CK2 is derived from a mammal, preferably from a human, a mouse, or a rat.
  • the type I interferon deficiency refers to a decrease in the amount and/or activity of type I interferon expression before and/or after the onset of the disease.
  • the type I interferon deficiency-associated disease means that the lack of type I interferon causes the occurrence and/or aggravation of the disease, and increases the expression level and/or activity of the type I interferon.
  • the disease has a therapeutic and/or preventive effect.
  • the inhibitor comprises an antisense nucleic acid, an inhibitory microRNA, an antibody or a small molecule compound of CK2.
  • the antibody comprises a small molecule polypeptide.
  • the inhibitor is an siRNA of the CK2 gene, shRNA.
  • the shRNA of the CK2 gene is as shown in (SEQ ID NO.: 1-2)
  • the amino acid sequence of the CK2 protein has Genbank accession numbers NP_031814.2 (mouse) and NP_808227.1 (human).
  • Genbank accession number of the nucleotide sequence encoding the CK2 protein is shown as NM_007788.3 (mouse) and NM_177559 (human).
  • the type I interferon deficiency-related diseases include: viral infectious diseases, malignant tumors, multiple sclerosis.
  • the viral infectious disease includes a disease infected with the following viruses: herpes simplex virus (HSV), vesicular virus (VSV), Kaposi's tumor herpesvirus (KSHV), respiratory syncytial disease Toxic (RSV), Hand, Foot and Mouth Disease Virus (Enterovirus EV71 and CA16) Hepatitis B Virus (HBV), Hepatitis C Virus (HCV), Human Acquired Immunodeficiency Virus/HIV (HIV); and/or
  • the malignant tumor includes the following tumors: hairy cell leukaemia, chronic myelogenous leukemia (CML), lymphoma, myeloma, melanoma, renal cell carcinoma and bladder cancer. (renal cell and bladder cell carcinoma), Kaposi's sarcoma.
  • the virus is a virus capable of activating a pattern recognition receptor after infecting a cell.
  • the inhibitor is also useful for inhibiting immune escape of viral and/or tumor cells.
  • the CK2 inhibitor is also used to inhibit viral replication and assembly.
  • the expression of the type I interferon and/or type I interferon-inducing gene or protein thereof comprises inducing and/or increasing a type I interferon and/or type I interferon-inducing gene or The amount and/or activity of the protein.
  • the inhibitor is a small molecule compound, preferably including tetrabromobenzotriazole (TBB), CX-4945 (Silmitasertib), DMAT (2-dimethylamino-4, 5, 6,7-tetrabromo-1H-benzimidazole), kaempferol, tyrosine phosphorylation inhibitor AG114 (Tyrphostin AG114), tetrabromocinnamic acid, 3-[[5-(4-tolyl)thiophene [2 ,3-d]pyrimidin-4-yl]thio]propionic acid (TTP 22), CX-5011, ellagic acid (Ellagic Acid), 3-methyl-1,6,8-trihydroanthracene (emodin) ), 4',5,7-trihydroxyflavone (apigenin).
  • TTBB tetrabromobenzotriazole
  • CX-4945 Siliconmitasertib
  • DMAT
  • the type I interferon-inducing gene or protein thereof comprises CXCL10/IP-10, STAT1, MX1, MX2, Rsad2/Viperine, phosphokinase TBK1, interferon regulatory factor 3 (IRF3).
  • the type I IFN comprises IFN- ⁇ , IFN- ⁇ .
  • the pharmaceutical composition comprises as an active ingredient a CK2 inhibitor, and a pharmaceutically acceptable carrier.
  • the effective concentration of the CK2 inhibitor is from 1 to 1000 ⁇ M, preferably from 20 to 100 ⁇ M.
  • the CK2 inhibitor is dose-dependently promoting the type I interferon and/or the type I interferon-inducing gene or its protein.
  • a non-therapeutic promoting cell expressing a type I interferon and/or a type I interferon-inducing gene or a protein thereof and/or a non-therapeutic inhibitor of a type I interferon deficiency-associated virus in vitro.
  • a method for growing cells and/or tumor cells comprising the steps of: culturing cells in the presence of a CK2 inhibitor, thereby promoting expression of a type I interferon and/or a type I interferon-inducing gene or a protein thereof inhibiting type I interferon Lack of associated viral cells and/or tumor cell growth.
  • the cell comprises a tumor cell, or a cell infected with a DNA or RNA virus.
  • the method can also be used to prepare a type I interferon or an induced protein thereof, for example, to collect and purify a type I interferon obtained in a cell culture system or an induced protein thereof.
  • a method of screening for a compound capable of inhibiting CK2 comprising the steps of:
  • test compound was added to the cell culture system, and the expression amount and/or activity of the type I interferon was determined; in the control group, the test compound was not added to the cell culture system, and the type I was determined. The amount and/or activity of interferon expression;
  • the candidate compound is a compound capable of inhibiting CK2 and/or
  • test compound In the test group, the test compound is added to the virus-infected cell culture system, and the titer of the virus in the cell culture system is determined; in the control group, the test compound is added to the virus-infected cell culture system, And measuring the virus titer in the cell culture system;
  • the candidate compound is a compound capable of inhibiting CK2.
  • the method further includes the steps of:
  • a method for preparing a type I interferon comprising the steps of:
  • a CK2 inhibitor is added to L929 cells, cultured for 12-48 hours under non-infective conditions or 12-24 hours under viral infection conditions. The supernatant was collected and the type I interferon was purified by HPLC.
  • the cells further comprise fibroblasts, or macrophages.
  • a method of treating a viral infection wherein a CK2 inhibitor is administered to a subject in need of treatment to treat a viral infection.
  • the subject in need of treatment is a mammal, preferably a human.
  • a method for determining whether a CK2 inhibitor is effective against a tumor or a virus comprising the steps of:
  • the expression level and/or activity I1 of type I interferon increased significantly compared with the expression level and/or activity I0 before the addition of the CK2 inhibitor, indicating that the CK2 inhibitor has an inhibitory effect on the tumor or virus.
  • Figure 1 shows that CK2 inhibits the expression of the type I interferon and/or type I interferon-inducible gene or its protein induced by Toll-like receptor signaling, but does not affect the expression of the inflammatory factor TNF ⁇ .
  • Figure 2 shows that CK2 inhibits activation of TBK1 and IRF3 induced by Toll-like receptor signaling, but does not affect activation of MAP kinases.
  • Figure 3 shows that CK2 inhibits the signaling pathway induced by pattern recognition receptors that recognize RNA and DNA in the cytosol and the expression of type I interferon and/or type I interferon-inducible genes or proteins thereof.
  • Figure 4 shows that CK2 inhibits the expression of type I interferon and/or type I interferon-inducible genes or their proteins in mouse macrophage cell line by HSV infection, and is highly expressed with type I interferon and its target gene product.
  • the replication, assembly and release of HSV virus were significantly inhibited, and the HSV virus titer detected in the CK2 ⁇ knockdown Raw264.7 cell culture was nearly 10 times lower than that of the control group.
  • Figure 5 shows that CK2 inhibits the expression of type I interferon and/or type I interferon-inducible genes or proteins thereof in the mouse fibroblast cell line L929 when infected with Sendai virus.
  • Figure 6 shows that CK2 inhibits the expression of type I interferon and/or type I interferon-inducible genes or proteins thereof in the mouse fibroblast cell line L929 when VSV virus is infected.
  • Figure 7 shows that the kinase activity of CK2 plays a key role in inhibiting virus-induced expression of type I interferons.
  • Figure 8 shows that CK2 inhibitor TBB induces TBK1 activation in mouse fibroblast cell line L929 and I Type of interferon and type I interferon-inducible gene or its protein expression.
  • Figure 9 shows that the CK2 inhibitor TBB inhibits infection and replication of VSV virus in the fibroblast cell line L929.
  • Figure 10 shows that the CK2 inhibitor TBB induces activation of TBK1 and expression of type I interferons and type I interferon-inducible genes or proteins thereof in human embryonic kidney cell line 293, and inhibits infection of VSV.
  • Figure 11 shows that prior to HCV infection of the human hepatocyte line Hu7.5, pretreatment with the CK2 inhibitor TBB induces type I interferon and/or type I interferon-inducible genes or their protein expression and prevents HCV infection.
  • Figure 12 shows that the CK2 inhibitor TBB is effective in inhibiting viral replication following HCV infection of the human hepatocyte cell line Hu7.5.
  • Figure 13 shows that the CK2 inhibitor TBB induces the expression of type I interferon and type I interferon-inducible genes or proteins thereof in human promyelocytic leukemia cells HL-60.
  • Figure 14 shows that the CK2 inhibitor TBB induces the expression of type I interferons and type I interferon-inducible genes or proteins thereof in human renal cancer cell line OS-RC-2.
  • Figures 15A-15B show the prophylactic and therapeutic effects of CK2 inhibitor TBB on EV71 virus infection, and both are more effective at high concentrations.
  • Figures 16A-16B show that the CK2 inhibitor TBB effectively activates the key kinase TBK1 that induces type I interferon expression in mouse tissues and inhibits the replication, assembly and release of HSV virus in the blood and spleen of TBB-treated mice.
  • the detected HSV virus titer was significantly lower than the control group.
  • CK2 protein is involved in regulating multiple pattern recognition receptor signaling pathways, controlling the activation of TBK1 and IRF3, and limiting the expression of type I interferon.
  • inhibition of CK2 can effectively induce and promote the production of type I interferon, and activate the expression of various type I interferon-inducible genes or their proteins, thereby improving the body's immune defense system and effectively preventing type I interferon-related Diseases, such as viruses associated with type I interferons, tumors, or the treatment of multiple sclerosis, have become a new strategy to improve human immunity, treat viral infectious diseases, and some non-infectious diseases. On the basis of this, the present invention has been completed.
  • type I interferon deficiency refers to before and/or after the onset of a disease, A decrease in the amount and/or activity of type I interferon.
  • a decrease in the expression level of type I interferon may lead to the occurrence of diseases such as multiple sclerosis and tumors, or the immune escape of tumors or viruses inhibits the expression of normal type I interferons, thereby further aggravating the occurrence of diseases such as tumors or viruses.
  • type I interferon deficiency-associated disease means that the absence of a type I interferon causes the occurrence and/or exacerbation of the disease, and increases the amount and/or activity of the type I interferon expression.
  • a disease in which the disease acts as a therapeutic and/or preventive agent refers to a type I interferon deficiency-associated viral infectious disease, multiple sclerosis or malignant tumor, and it is understood that it is not related to "type I interferon deficiency”. Viral infections or malignancies are not within the scope of this term.
  • CK2 (also known as casein kinase 2) is a serine and threonine kinase ubiquitous in eukaryotic cells, mainly tetramers - containing two catalytic subunits ( ⁇ and / or ⁇ ') and the form of two regulatory subunits ⁇ function.
  • the CK2 of the present invention is derived from a mammal and is usually derived from human, mouse or rat.
  • the preferred amino acid sequence of the CK2 protein is shown in Genbank accession numbers NP_808227.1 (mouse) and NP_031814.2 (human), and the nucleotide sequence encoded by Genbank accession number is NM_007788.3 (mouse) and NM_177559 (person) is shown.
  • Genbank accession number is NP_808227.1 (mouse) and NP_031814.2 (human)
  • the CK2 gene sequence is shown in Genbank ID No.: 12995 (mouse) and Genbank ID No.: 1457 (human).
  • CK2 plays an important role in regulating cell growth and tumorigenesis.
  • the expression and activity of CK2 tend to increase abnormally. Therefore, it has been reported that inhibition of the expression or activity of CK2 can effectively inhibit the growth of tumor cells and induce necrosis of tumor cells.
  • CX-4945 an inhibitor of CK2
  • CK2 inhibitors also have a selective effect on the therapeutic effects of tumors. Because of the many substrates and mechanisms of CK2, the specific mechanism of action of CK2 inhibitors in each type of tumor treatment is not known. Therefore, the solution to this selective performance is not currently available.
  • the present invention firstly found through experiments that CK2 is involved in the regulation of multiple pattern recognition receptor signaling pathways, and controls the activation of TBK1 and IRF3, thereby limiting the expression of type I interferon. Experiments have shown that inhibition of CK2 can increase the expression of type I interferon and its induced genes, and has the effect of inhibiting viral infection, multiple sclerosis and tumors. good effect.
  • CK2 is a key molecule that negatively regulates the expression of TBK1 and type I interferon, but also experimentally demonstrates that inhibition of CK2 kinase activity by small molecule compounds can effectively induce type I interferon production, and Thereby inhibiting and killing viral infection, multiple sclerosis and tumor cell growth.
  • PRRs Pattern Recognition Receptors activate complex signaling pathways to induce type I interferons and cells by recognizing Pathogen-Associated Molecular Patterns (PAMPs, RNA and DNA secreted by dead cells or viruses).
  • PAMPs Pathogen-Associated Molecular Patterns
  • RNA and DNA secreted by dead cells or viruses The expression of factors and chemokines, and the implementation of physiological functions such as host defense and tissue repair.
  • the Toll-like receptor TLR4 located on the cell membrane recognizes the glycoprotein on the surface of the virus and transports it into the endosome; TLR3 in the endosome recognizes the viral double-stranded DNA; TLR3/TLR4 passes through the linker molecule TRIF and in the endosome TRAM, which subsequently activates the phosphokinase TBK1 (TANK-binding kinase 1), causes activation of the downstream transcription factor IRF3, thereby inducing expression of type I interferon.
  • TLR4 located on the cell membrane recognizes the glycoprotein on the surface of the virus and transports it into the endosome
  • TLR3 in the endosome recognizes the viral double-stranded DNA
  • TLR3/TLR4 passes through the linker molecule TRIF and in the endosome TRAM, which subsequently activates the phosphokinase TBK1 (TANK-binding kinase 1), causes activation of the downstream transcription factor IRF3, thereby inducing
  • RNA released by the virus or the RNA released by the damaged cells of the body can be recognized by the RIG-I-like receptor RIG-I (retinoic acid-ducible gene-I) and MDA5 (melanoma differentiation-associated gene 5) in the cytoplasm, and the linker molecule MAVS /VISA activates phosphokinase TBK1 and the transcription factor IRF3 on mitochondria and also induces type I interferon expression.
  • Double-stranded DNA (dsDNA) released by viruses or damaged cells is recognized by various DNA receptors in the cytoplasm, such as cGAS, DDX41, and IFI16, and TBK1 and IRF3 are activated by the linker molecule STING to induce type I interferon. expression.
  • dsDNA Double-stranded DNA
  • dsDNA Double-stranded DNA released by viruses or damaged cells is recognized by various DNA receptors in the cytoplasm, such as cGAS, DDX41, and IFI16, and TBK
  • partial viral infection or death of the body's cells can cause activation of the pattern recognition receptor signaling pathway, thereby inducing expression of type I interferon IFN ⁇ / ⁇ .
  • Binding of IFN ⁇ / ⁇ to interferon receptor activates Jak1 and Tyk2, activates the transcription factor STAT1, and induces expression of hundreds of interferon- and/or type I interferon-inducible genes or proteins thereof, including MX1, MX2, Rsad2/ Viperine, CXCL10/IP-10, etc., implement host defense functions, remove invading viruses and infected cells, or repair tissue damage to remove diseased or cancerous cells.
  • the virus also acquires the ability to antagonize the pattern recognition receptor signaling pathway and inhibit the expression of type I interferon to escape the host's defense function.
  • the hepatitis C virus HCV-encoded protease NS3/4A cleaves the linker molecule of TLR3/4, the linker molecule Trif and RIG-I/MDA5.
  • MAVS/VISA causing them to not activate downstream TBK1 and IRF3.
  • Enterovirus EV71 can inhibit the expression of type I interferon by cleavage of MAVS by protease 2A (pro) and by cleavage of Trif by 3C protein.
  • the F protein of vesicular virus VSV and the NS1 of influenza virus can also interfere with host expression of type I interferon by inhibiting the function of RIG-I.
  • Human acquired immunodeficiency virus/HIV HIV
  • HIV Human acquired immunodeficiency virus/HIV
  • Trex1 and SamHD1 can inhibit the production of type I interferon by using cells such as Trex1 and SamHD1 to degrade or inhibit reverse transcription of double-stranded DNA.
  • the immune response of the virus to the host produces an escape phenomenon, resulting in an acute or chronic infection of the virus in the host cell, causing the host to develop severe lesions.
  • the mechanism of inhibition of type I interferon expression may also cause type I interferon not to be expressed, leading to disease progression.
  • active ingredient of the present invention As used herein, the terms "active ingredient of the present invention”, “inhibitor of the present invention”, and “inhibitor of CK2 of the present invention” are used interchangeably and mean a substance capable of reducing the amount and/or activity of expression of a CK2 gene or protein.
  • the experiments of the present invention prove that the CK2 inhibitor can effectively block the expression amount and/or activity of CK2, thereby inducing or promoting the expression of type I interferon, TBK1, and IRF3, in order to achieve treatment of viral infection, multiple sclerosis and tumor. purpose.
  • the CK2 inhibitor which can be used in the present invention is not particularly limited and may be any substance which reduces the expression amount and/or activity of the CK2 gene or protein.
  • Representative examples include antisense nucleic acids of the CK2 gene, microRNAs (siRNA) that inhibit CK2 expression, antibodies against the CK2 protein, polypeptides that inhibit the action of CK2 on TBK1, or small molecule compounds that have an inhibitory effect on the amount and/or activity of CK2 expression.
  • CK2 gene sequence one skilled in the art can design and synthesize an antisense nucleic acid or miRNA having a CK2 gene inhibitory effect by conventional techniques.
  • Small molecule compounds having CK2 inhibitory properties can be obtained commercially or synthetically.
  • Preferred examples are tetrabromobenzotriazole (TBB, available from Tocris), DMAT (2-dimethylamino-4, 5,6,7-tetrabromo-1H-benzimidazol, available from MedChem Express), kaempferol, tyrosine phosphate Inhibitor AG114 (Tyrphostin AG114, available from Alexis Biochemicals), CX-4945 (Silmitasertib, available from Selleck), tetrabromocinnamic acid, 3-[[5-(4-methylphenyl)thiophene [2,3-d] Pyrimidine-4-yl]thio]propionic acid (TTP 22, available from Tocris), CX-5011, ellagic acid (Ellagic Acid, available from MedChem Express), 3-methyl-1,6,8-trihydrogen Emo (emodin, purchased from Sigma), 4',5,7-trihydroxyflavone (apigenin, purchased from Shanghai Chemical Industry Development Co
  • the CK2 inhibitor of the present invention further comprises an expression vector comprising shRNA having CK2 inhibitory activity, It can be obtained by a conventional method.
  • a preferred shRNA sequence having CK2 inhibitory activity is as follows:
  • the effective therapeutic concentration range of the CK2 inhibitor useful in the present invention can be screened according to an effective concentration screening method commonly used in the art, thereby obtaining a safe and effective administration dose.
  • the effective concentration of the CK2 inhibitor is 1-1000 ⁇ M, preferably It is 20-100 ⁇ M.
  • Type I interferons include IFN ⁇ and IFN ⁇ , which bind to interferon receptors on the cell surface to activate protein kinases Jak1 and Tyk2, and then activate the transcription factors STAT1 or/and STAT2 to induce expression of hundreds of genes (interferon induction) Genes, ISGs), including MX1, MX2, Rsad2/Viperine, CXCL10/IP-10, etc. These genes induced by interferon play an important physiological role in antiviral infection, regulation of cell proliferation and apoptosis, angiogenesis, and T cell immune response. Therefore, the use of type I interferons in the treatment of viral infections, immune-related diseases and tumors has been extensively studied.
  • the indications that are effective after receiving type I interferon therapy include a variety of viral infections, such as hepatitis B and C infection, multiple sclerosis and more than a dozen tumors, including leukemia, lymphoma, myeloma, Melanoma, renal cell tumor, bladder tumor, and Kaposi's sarcoma.
  • the mechanism by which interferon inhibits multiple sclerosis may involve its inhibition of NLRP3-mediated inflammatory stimuli, and the mechanism by which interferon inhibits tumors involves activating anti-tumor immunity, inhibiting tumor cell proliferation, and inducing tumor cell dying. Dead and so on.
  • CK2 inhibitor of the present invention to promote the expression of type I interferon can treat various diseases which are deficient in type I interferon, for example, the above-mentioned viral infection, multiple sclerosis and tumor and many more.
  • type I interferon-inducible gene or protein thereof refers to a gene that is associated with type I interferon expression on a type I interferon or downstream, when type I interferon expression is altered. Its protein, usually an infection, an immune-related inflammatory factor or an antiviral factor.
  • the type I interferon-inducing gene or protein thereof comprises various antibodies such as activated phosphokinase (TBK1), interferon regulatory factor 3 (IRF3), MX1, MX2, Rsad2/Viperine, or CXCL10/IP-10. Regulatory factors and inflammatory chemokines.
  • viral infection As used herein, the terms “viral infection”, “viral infectious disease” are used interchangeably and refer to a viral infection-associated disease capable of activating a pattern recognition receptor upon infection of a cell.
  • the inhibitors of the invention are directed against viral infections caused by various viruses, including a variety of DNA or RNA viruses.
  • viruses including a variety of DNA or RNA viruses.
  • HSV herpes simplex virus
  • VSV vesicular virus
  • KSHV Kaposi's tumor virus
  • RSV respiratory syncytial virus
  • enter virus EV71 and CA16 hepatitis B virus
  • HCV hepatitis C virus
  • HAV human acquired immunodeficiency virus/HIV
  • type I interferons can directly cause apoptosis, inflammatory viral replication, assembly, and release of infected cells.
  • killer NK and NKT cells and CD8 T cells play various roles in antiviral effects.
  • the virus which can be used in the present invention is a plurality of viruses capable of inhibiting the expression of type I interferon in an infectious state, for example, viruses including hepatitis B and C viruses, enterovirus EV71, human acquired immunodeficiency virus/HIV ( HIV), herpes simplex virus (HSV), vesicular virus (VSV), Kaposi's tumor herpesvirus (KSHV), respiratory syncytial virus (RSV), and the like.
  • viruses including hepatitis B and C viruses, enterovirus EV71, human acquired immunodeficiency virus/HIV ( HIV), herpes simplex virus (HSV), vesicular virus (VSV), Kaposi's tumor herpesvirus (KSHV), respiratory syncytial virus (RSV), and the like.
  • viruses including hepatitis B and C viruses, enterovirus EV71, human acquired immunodeficiency virus/HIV ( HIV), herpes simplex virus (HSV), vesicular virus
  • the present invention utilizes corresponding small molecule compounds to gradually induce an appropriate amount of interferon expression in vivo, and will be an ideal solution for treating these viral diseases which antagonize the expression of interferon.
  • these viruses are often prone to mutations in viral genes during replication. Therefore, some antiviral drugs designed for viral gene products are temporarily effective, but they can quickly lead to the emergence of resistant strains after extensive application. Small molecule drugs designed for host proteins will not encounter similar problems. Therefore, the strategy for inducing expression of type I interferon against host proteins is more desirable than currently designed for HCV and HIV.
  • CK2 inhibitors to promote type I interferon can be applied to a variety of therapeutic or non-therapeutic viral infections, multiple sclerosis or tumors associated with type I interferon deficiency.
  • the present invention can utilize a CK2 inhibitor to induce or promote the expression of type I interferon in vitro, collect or purify the type I interferon obtained in the cell culture system, thereby providing a method for preparing type I interferon.
  • Method for collecting and purifying type I interferon after type I interferon is usually produced in a cell culture system All are routine experimental methods known to those skilled in the art.
  • CK2 inhibitors can also be used to screen for and identify candidate drugs for type I interferon deficiency-related diseases; the present invention can also use the relationship between CK2 inhibitors and type I interferons to determine a specific tumor, multiple sclerosis or Whether the virus has an inhibitory effect (sensitivity assay).
  • CK2 inhibitors can be administered to a desired subject to reduce replication and assembly of the infected virus, thereby reducing infection with type I interferon-associated virus, or reducing type I interferon-related multiple sclerosis and tumorigenesis. And development.
  • the term "effective amount” or “effective amount” refers to an amount that can produce a function or activity on a human and/or animal and that can be accepted by a human and/or animal.
  • the term "pharmaceutically acceptable” ingredient is a substance that is suitable for use in humans and/or mammals without excessive adverse side effects (eg, toxicity, irritation, and allergies), ie, a substance having a reasonable benefit/risk ratio.
  • pharmaceutically acceptable carrier refers to a carrier for the administration of a therapeutic agent, including various excipients and diluents.
  • compositions of the present invention comprise a safe and effective amount of the active ingredient of the present invention together with a pharmaceutically acceptable carrier.
  • a pharmaceutically acceptable carrier include, but are not limited to, saline, buffer, dextrose, water, glycerol, ethanol, and combinations thereof.
  • the pharmaceutical preparation should be matched with the administration mode, and the pharmaceutical composition of the present invention is in the form of an injection, an oral preparation (tablet, capsule, oral liquid), a transdermal agent, and a sustained release agent.
  • it is prepared by a conventional method using physiological saline or an aqueous solution containing glucose and other adjuvants.
  • the pharmaceutical composition is preferably manufactured under sterile conditions.
  • the effective amount of the active ingredient of the present invention may vary depending on the mode of administration and the severity of the disease to be treated and the like. The selection of a preferred effective amount can be determined by one of ordinary skill in the art based on various factors (e.g., by clinical trials). The factors include, but are not limited to, pharmacokinetic parameters of the active ingredient such as bioavailability, metabolism, half-life, etc.; severity of the disease to be treated by the patient, body weight of the patient, immune status of the patient, administration Ways, etc.
  • pharmacokinetic parameters of the active ingredient such as bioavailability, metabolism, half-life, etc.
  • severity of the disease to be treated by the patient body weight of the patient, immune status of the patient, administration Ways, etc.
  • a satisfactory effect can be obtained.
  • several separate doses may be administered per day, or the dose may be proportionally reduced, as is critical to the condition of the treatment.
  • Pharmaceutically acceptable carriers of the invention include, but are not limited to, water, saline, liposomes, lipids, proteins, protein-antibody conjugates, peptide materials, cellulose, nanogels, or Its combination.
  • the choice of carrier should be compatible with the mode of administration, which are well known to those of ordinary skill in the art.
  • CK2 is involved in the regulation of multiple pattern recognition receptor signaling pathways and controls the activation of TBK1 and IRF3, thereby limiting the expression of type I interferons.
  • inhibition of CK2 kinase activity by small molecule compounds such as TBB can effectively induce type I interferon production, thereby preventing the occurrence and development of type I interferon-related diseases, and providing new immune sclerosis for multiple sclerosis, virus or tumor.
  • the treatment The method of endogenously increasing type I interferon against the host can overcome the defect that the exogenous injection of interferon is difficult to control the amount of use and cause serious side effects, making the treatment method more effective and safer.
  • Example 1 Protein kinase CK2 is involved in the regulation of type I interferon expression induced by multiple pattern recognition receptors
  • Lenti-viral vector pLKO.1 was used to construct recombinant viral vectors expressing control shRNA or shRNA against CK2 ⁇ , and then transfected into macrophage cell line (Raw264.7) and fibroblast cell line (L929), respectively. A stably transfected cell line with low expression of CK2 ⁇ .
  • shRNA oligonucleotide sequence against CK2 ⁇ was designed based on the CK2 gene sequence in the NCBI gene bank:
  • Plasmid pLKO.1-shRNA expressing the control shRNA or shRNA against CK2 ⁇ was constructed using Addgene's commercialized pLKO.1 - TRC Cloning Vector vector (Addgene Plasmid No. 10878) and its standard protocol. 10 ⁇ g/ml control shRNA or shRNA plasmid against CK2 ⁇ pLKO.1-shRNA was transformed into HEK293 cells by calcium phosphate (125 mM Cacl2/1x HEPES) transformation with 2.5 ⁇ g/ml pMD2.G (Addgene plasmid product number 12259) and 7.5 ⁇ g/ml psPAX2 (Addgene plasmid product number 12260), respectively.
  • the cell supernatant was collected 48 hours later to obtain a control shRNA or a recombinant viral vector against shRNA of CK2 ⁇ (HEK293 cell culture medium: DMEM complete medium containing 10% FBS and 1% penicillin/streptomycin).
  • a 70% abundance macrophage cell line (Raw264.7 purchased from the US ATCC) or a fibroblast cell line (L929 purchased from the US ATCC) was transfected with a control shRNA or a recombinant viral vector directed against shRNA of CK2 ⁇ at 50%.
  • Raw264.7 and L929 cell culture media are DMEM complete medium.
  • the recombinant viral vector was removed after 4 hours of transfection and culture continued until the cell abundance reached 95%. Cell passage was performed at a ratio of 1:5. Since the control shRNA or shRNA against CK2 ⁇ had a puromycin resistance site, resistant cells were screened after passage using DMEM complete medium containing 4 ⁇ g/ml puromycin.
  • a positive stable cell line and a control cell line of CK2 ⁇ gene knockdown can be obtained by screening 3-4 generations (about 7-10 days).
  • the Raw 264.7 stable cell line knocked down by control and CK2 ⁇ was inoculated into 6-well plates at 10 6 /ml, and 2 ml was inoculated per well.
  • Cells were treated with LPS (100 nM, purchased from Invivogen), polyI:C (50 ⁇ g/ml, purchased from Invivogen), respectively, to activate the TLR4 or TLR3 signaling pathways ( Figures 1 and 2). After 4 hours of stimulation, cells were harvested and RNA was extracted (Fig. 1); or after 15 to 4 hours of stimulation, cells were collected at different time points to prepare a protein extract (Fig. 2).
  • Control and CK2 ⁇ knockdown L929 stable cell lines were inoculated into 6-well plates at 10 6 /ml, and 2 ml was inoculated per well.
  • PolydA:dT (1 ⁇ g/ml, purchased from Invivogen) and polyI:C (1 ⁇ g/ml, purchased from Invivogen) cells were transfected with lipofect lipofectamine 2000 (purchased from Invitrogen) to activate RIG-I/Mda5 and cGAS, respectively. /STING signal path ( Figure 3).
  • cells were directly treated with DMXAA (100 ⁇ g/ml, purchased from Selleck) to activate the STING signaling pathway ( Figure 3). The cells were harvested 6 hours after stimulation, and RNA was extracted (Fig. 3A); or after 3 hours of stimulation, the cells were collected to prepare a protein extract or a cytoplasmic, nuclear protein extract (Figs. 3B and 3C).
  • the protocol for preparing cytosolic and nuclear protein extracts is as follows: first with hypotonic buffer (10 mM HEPES (pH 7.6), 1.5 mM MgCl2, 10 mM KCl, 1 mM EDTA, protease inhibitor complete (Roche), 1 mM PMSF, 1 mM Cells were lysed with NaF, 1 mM Na3VO4) and treated back and forth 10-15 times with a homogenizer Douncer and then incubated on ice for 20 minutes. After centrifugation at a low temperature of 4 ° C (3000 rpm, 5 minutes), the supernatant was taken out, and after centrifugation at high speed to remove insoluble matter, it was a cytosolic protein extract.
  • hypotonic buffer 10 mM HEPES (pH 7.6), 1.5 mM MgCl2, 10 mM KCl, 1 mM EDTA, protease inhibitor complete (Roche), 1 mM PMSF, 1 mM Cell
  • the pellet after low-speed centrifugation was lysed by high-salt buffer (20 mM HEPES (pH 7.6), 500 mM NaCl, 1.5 mM MgCl2, 1 mM EDTA, protease inhibitor complete (Roche), 1 mM PMSF, 1 mM NaF, 1 mM Na3VO4), and centrifuged at high speed. After removing the insoluble matter, it is a nuclear protein extract. Phosphorylation of proteins such as TBK1, IRF3, I ⁇ B ⁇ , STAT1, JNK, ERK, and p38 was detected by Western blot. Tubulin, GAPDH, Lamin B1, etc. are reference molecules for total protein amount of total protein, cytosolic protein or nuclear protein extract.
  • type I interferon-activated p-STAT1 was significantly more potent than CK2 knockdown L929 cells.
  • the cells are taller (Fig. 3B).
  • CK2 knockdown L929 nuclei not only did p-TBK1 rise, but nuclear transfer of IRF3 was also significantly upregulated (Fig. 3C).
  • nuclear transfer of NF ⁇ B p65 did not increase significantly (Fig. 3C). It was further demonstrated that CK2-specific inhibition of TBK1 and IRF3 activation regulates the expression of multiple pattern recognition receptor-mediated type I interferons and their type I interferon-inducible genes.
  • Example 2 Protein Kinase CK2 is involved in the regulation of type I interferon expression induced by various viral infections
  • Control and CK2 ⁇ knockdown stable cell lines were inoculated into 6-well plates at 10 6 /ml, and 2 ml were inoculated per well.
  • DNA virus HSV-1 (1 ⁇ 10 7 pfu/ml)
  • RNA virus vesicular virus VSV 2.5 ⁇ 10 5 TCID 50 /ml
  • Sendai virus SeV (1 ⁇ 10 8 HA/ml) were infected, respectively.
  • Six hours after infection with the virus cells were harvested for RNA extraction preparation.
  • Six hours after infection with HSV virus the cells were exchanged for 66 hours and the cell supernatant was collected for HSV virus titer determination.
  • the cell supernatants with different concentration gradients are added separately (stock solution, 1:10 1 , 1:10 2 , 1:10 3 , 1:10 4 , 1:10 5 , 1:10 6 , 1 ) :10 7 , 1:10 8 , 1:10 9 , 1:10 10 ) and the blank control for 4 hours.
  • the supernatant was removed, and 400 ⁇ l of a 0.8% agar-MEM culture solution was added to each well, and allowed to stand at room temperature for 1 hour. After the medium was solidified, the culture was further inverted for 67 hours. After the completion of the culture, 500 ⁇ l of a crystal violet solution was added to each well, and the mixture was allowed to stand at room temperature for 2 hours. The agar was removed, the number of plaques was counted, and the virus titer (pfu/ml, plaque forming unit/ml) was calculated.
  • VSV and SeV-induced genes such as IFN ⁇ , IFN ⁇ , Cxcl10, Mx1, and Mx2 was also significantly up-regulated in CK2 knockdown L929 cells compared to control cells (Fig. 5 and Fig. 6). Therefore, down-regulation of CK2 expression in a variety of viral infections can significantly enhance the ability of different types of cells to express type I interferons. These results indicate that CK2 is a key molecule for the negative regulation of type I interferon expression.
  • CK2 ⁇ wild-type and kinase-inactivated mutant K68M was cloned on the lenti-viral vector pCDH, and then transferred into L929 cell line.
  • the control cell lines were screened by puromycin and expressed wild.
  • RNA in the cells was extracted by TRIZOL (Invitrogen) method. 1 ⁇ g RNA first strand cDNA synthesis with SuperScript TM III Reverse Transcriptase Kit (Invitrogen ) reverse transcriptase, respectively, for real-time quantitative PCR detection of gene expression levels of type I interferon, as measured using ⁇ ct method to calculate the relative expression of the gene relative to GAPDH .
  • Example 4 Inhibition of CK2 Kinase Activity Promotes Cellular Expression of Type I Interferon and Prevents Vesicular Virus Infection
  • Mouse fibroblast L929 was inoculated into 6-well plates at 10 6 /ml, 2 ml per well, and when the cell abundance reached 70%, different concentrations of CK2 kinase inhibitor - small molecule compound TBB were used.
  • DMSO control after treatment of the cells for 6 hours or 12 hours, using lysis buffer (50 mM Tris-HCl (pH 7.4), 150 mM NaCl, 1 mM EDTA, 1% Triton X-100, protease inhibitor Complete (Roche), The protein lysate of the cells was extracted and prepared by 1 mM PMSF, 1 mM NaF, 1 mM Na3VO4), and phosphorylation of NF ⁇ B p65 and TBK1 was detected by Western hybridization.
  • lysis buffer 50 mM Tris-HCl (pH 7.4), 150 mM NaCl, 1 mM EDTA, 1% Triton X-100, protease inhibitor Complete (Roche)
  • the protein lysate of the cells was extracted and prepared by 1 mM PMSF, 1 mM NaF, 1 mM Na3VO4), and phosphorylation of NF ⁇ B p65 and TBK1 was detected by Western hybridization.
  • L929 cells were treated with TBB (100 ⁇ M) or DMSO (control) for 12 hours in the same manner, and RNA in the cells was extracted by TRIZOL (Invitrogen) method. 1 ⁇ g RNA first strand cDNA synthesis with SuperScript TM III Reverse Transcriptase Kit (Invitrogen ) reverse transcriptase, respectively, for real-time quantitative PCR detection of gene expression levels of type I interferon, as measured using ⁇ ct method to calculate the relative expression of the gene relative to GAPDH .
  • HEK293T Human embryonic kidney cells HEK293T were inoculated into 6-well plates at 10 6 cells/ml, and 2 ml were inoculated per well. When the cell abundance reached 70%, cells were treated with TBB or DMSO (control). The protein lysate of the same method was used for Western detection, RNA was extracted for the detection of type I interferon-related gene expression, and the efficiency of VSV infection was detected by fluorescence microscopy.
  • RESULTS It was found that in human embryonic kidney cell HEK293T, TBK1 was also significantly activated after 12 or 24 hours of TBB treatment (Fig. 10A), and promoted the expression of genes such as IFN ⁇ , IFN ⁇ , Mx1 and Mx2 (Fig. 10B). . More importantly, the ability of TKB-treated HEK 293 cells to resist VSV infection was also greatly enhanced (Fig. 10C). These results indicate that TBB can induce the expression of type I interferon in both mouse and human cells, establish a defense state against viral infection, and effectively resist the infection of VSV virus.
  • Example 5 CK2 inhibitor TBB prevents HCV infection
  • Hu7.5 cells were inoculated into 6-well plates at 5 ⁇ 10 4 cells/ml, and each well was inoculated with 2 ml.
  • the medium used was DMEM complete medium.
  • MOI multipleplicity of infection
  • MOI 0.1
  • Cells were harvested for efficiency of RNA analysis and HCV infection, respectively, and cell supernatants were collected for HCV virus titer determination.
  • the virus titer of HCV in the cell supernatant was detected by classical virus titer assay.
  • the specific protocol was: inoculate Hu7.5 cells into 96-well plates at 5 ⁇ 10 4 /ml, and inoculate 200 ⁇ l per well. When the abundance reaches 30%, the cell supernatants with different concentration gradients are added separately (stock solution, 1:10 1 , 1:10 2 , 1:10 3 , 1:10 4 , 1:10 5 , 1:10 6 , 7 1:10, 1: 108, 1: 109, 1: 1010), cultured for 72 hours, the cells were collected, labeled antibodies specific for HCV paraformaldehyde after immunofluorescence, fluorescence microscopy has been HCV The number of Hu7.5 cells infected with fluorescently labeled cells was calculated and the virus titer (ffu/ml, fluorescence forming foci/ml) was calculated.
  • RNA and type I interferon-related genes in cells were extracted by the same method.
  • the results of quantitative PCR showed that the expression of type I interferon-related genes such as IFN ⁇ , IFN ⁇ , Mx1 and Mx2 in TBB pretreated cells.
  • TBB pretreatment also increased the expression of the inflammatory factor IL-6 in the cells (Fig. 11A).
  • the expression level of the Hcv gene was very small (Fig. 11A).
  • the cells were fixed in 4% paraformaldehyde for 30 minutes, washed with PBS phosphate buffer, labeled with HCV-specific antibodies by immunofluorescence, and the efficiency of HCV infection was detected by fluorescence microscopy.
  • Hu7.5 cells did not induce type I interferon expression after HCV infection.
  • TBB type I interferon-inducible genes MX1 and MX2, and some pro-inflammatory factors such as IL-6 and TNF ⁇ (Fig. 12A).
  • type I interferons and their target gene products, the replication, assembly and release of HCV virus were significantly inhibited, and the HCV virus titer detected in TBC-treated cell culture was lower than that of untreated. -100 times (Fig. 12B).
  • Example 7 regulates the expression of type I interferon in tumor cells
  • HL-60 Human promyelocytic leukemia cells HL-60 (purchased from ATCC) and human renal cancer cell line OS-RC-2 were cultured in RPMI-1640 medium supplemented with 10% fetal bovine serum. 2 mM glutamine and antibiotics. HL-60 or OS-RC-2 cells were inoculated into 6-well plates at 5 ⁇ 10 4 cells/ml, and each well was inoculated with 2 ml. When the cell abundance reached 30%, 100 ⁇ M TBB or DMSO (control) was added. . After 12 hours, cells were harvested and RNA was prepared using Trizol (Sigma).
  • RNA first strand cDNA synthesis with SuperScript TM III Reverse Transcriptase Kit (Invitrogen ) reverse transcriptase respectively, for real-time quantitative PCR detection of gene expression levels of type I interferon, as measured using ⁇ ct method to calculate the relative expression of the gene relative to GAPDH .
  • Example 8 CK2 inhibitor TBB inhibits EV71 infection
  • RESULTS Vero cells were inoculated into a 24-well plate at 3 ⁇ 10 4 cells/ml, and 0.5 ml per well was inoculated. When the abundance of Vero cells reached 70%, Vero cells were treated according to the TBB prevention group and the TBB treatment group, respectively.
  • TBB prevention group 20, 50, and 100 ⁇ M TBB or DMSO (control) were treated with cells for 2 hours, respectively, and infected with enterovirus EV712*10 2 TCID 50 /ml for 4 hours, then change the solution and continue to add 20, 50 and The medium of 100 ⁇ M TBB or DMSO (control) was cultured for 24 hours.
  • TBB treatment group Infected enterovirus EV712*10 2 TCID 50 /ml 4 hours later, change medium, culture with medium supplemented with 20, 50 and 100 ⁇ M TBB or DMSO (control) for 72 hours, collect cell supernatant
  • the EV71 virus titer was determined.
  • the viral titer of EV71 in the cell supernatant was determined by the classical virus titer assay TCID 50 (half the number of tissue cell infections).
  • the specific protocol was: inoculate Vero cells into 96-well plates at 3 ⁇ 10 4 /ml, respectively. Each well was inoculated with 0.1 ml. When the cell abundance reached 70%, cell supernatants with different concentration gradients were added (stock solution, 1:10 1 , 1:10 2 , 1:10 3 , 1:10 4 , 1: 10 5 , 1:10 6 , 1:10 7 , 1:10 8 , 1:10 9 , 1:10 10 ) and the blank control for 4 hours. After 4 hours, the supernatant was removed, the solution was changed, and incubation was continued for 68 hours.
  • the degree of infection of the cells was observed by an optical microscope, and the number of well plates exceeding half of the infection was counted, and the virus titer (TCID 50 /ml, tissue culture infective dose/ml) was calculated.
  • Vero cells were treated in different doses of TBB prevention group. After EV71 infection, the EV71 virus titer detected in the cell culture medium treated by TBB prevention group was lower than that in the untreated group, and showed a certain concentration correlation. In the prevention group, the higher the concentration of TBB used, the lower the EV71 virus titer detected in the cell culture solution (Fig. 15A). The EV71 virus titer detected in the cell culture medium of the treatment group infected with EV71 was 10-100 times lower than that of the untreated group (Fig. 15B). The high concentration of TBC (100 ⁇ M) can significantly inhibit the EV71 virus. Replication, assembly and release resulted in a 100-fold reduction in the final detected EV71 virus titer (Fig. 15B).
  • TBB activates the key kinase TBK1 that induces type I interferon expression in mice
  • mice of 6-8 weeks old, male and female were intraperitoneally injected with 2.5, 10, 25 mg/kg TBB or sunflower oil (control group) according to the body weight of the mice. After 24 hours, the mice were sacrificed according to the animal welfare method. Liver and spleen were taken to determine the degree of activation of the cytokine TBK1 associated with type I interferon synthesis in tissue samples.
  • lysis buffer 50 mM Tris-HCl (pH 7.4), 150 mM NaCl, 1 mM EDTA, 1% Triton X-100, Protease Inhibitor Complete (Roche), 1 mM PMSF, 1 mM NaF, 1 mM. Na3VO4
  • lysis buffer 50 mM Tris-HCl (pH 7.4), 150 mM NaCl, 1 mM EDTA, 1% Triton X-100, Protease Inhibitor Complete (Roche), 1 mM PMSF, 1 mM NaF, 1 mM. Na3VO4
  • mice of 6-8 weeks old, male and female were intraperitoneally injected with 10 mg/kg TBB or sunflower oil (control group) according to the body weight of the mice. After 6 hours, the mice were injected with 5*10 7 according to the tail weight of the mice. Pfu/g HSV. Twelve hours after the injection of HSV, the second TBB was given by the same method. After 24 hours of HSV injection, the mice were sacrificed according to the animal welfare method, and the blood and spleen were taken to determine the virus titer of HSV in the tissue samples. The virus titer of HSV in mouse tissue samples was detected by classical virus titer assay.
  • tissue sample homogenate (stock solution, 1 to 5 ⁇ 10 4 cells / ml Vero cells were seeded into 24-well plates, each well was inoculated 0.5ml, abundance of cells to be 30%, were added different concentration gradient: 101 , 1:10 2 , 1:10 3 , 1:10 4 , 1:10 5 , 1:10 6 , 1:10 7 , 1:10 8 ) and the blank control (PBS) for 4 hours.
  • the homogenate was removed, and 400 ⁇ l of a 0.8% agar-MEM culture solution was added to each well, and allowed to stand at room temperature for 1 hour. After the medium was solidified, the culture was further inverted for 67 hours. After the completion of the culture, 500 ⁇ l of a crystal violet solution was added to each well, and the mixture was allowed to stand at room temperature for 2 hours. The agar was removed, the number of plaques was counted, and the virus titer (pfu/g, plaque forming unit/g) was calculated.
  • RESULTS The intraperitoneal injection of 10 mg/kg TBB was effective in activating the key kinase TBK1 that induces type I interferon expression in mouse tissues (Fig. 16A), thereby significantly inhibiting the replication, assembly and release of HSV virus. After 24 hours of HSV infection, the HSV virus titer detected in the blood and spleen of the TBB-treated group was significantly lower than that of the control group (Fig. 16B).

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Molecular Biology (AREA)
  • Microbiology (AREA)
  • Animal Behavior & Ethology (AREA)
  • Organic Chemistry (AREA)
  • Veterinary Medicine (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Biotechnology (AREA)
  • Public Health (AREA)
  • Epidemiology (AREA)
  • Immunology (AREA)
  • Zoology (AREA)
  • Biochemistry (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Biomedical Technology (AREA)
  • Urology & Nephrology (AREA)
  • Genetics & Genomics (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Hematology (AREA)
  • Wood Science & Technology (AREA)
  • Physics & Mathematics (AREA)
  • General Chemical & Material Sciences (AREA)
  • General Engineering & Computer Science (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Pathology (AREA)
  • General Physics & Mathematics (AREA)
  • Analytical Chemistry (AREA)
  • Food Science & Technology (AREA)
  • Cell Biology (AREA)
  • Mycology (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Abstract

A use of a casein kinase 2 inhibitor in preparing a pharmaceutical composition for enhancing the expression of type I interferon and/or a type I interferon-induced gene or a protein thereof; and/or in preparing a pharmaceutical composition for treating and/or preventing the diseases related to the lack of type I interferon.

Description

抑制酪蛋白激酶2活性在促进I型干扰素表达的应用Application of inhibiting casein kinase 2 activity in promoting expression of type I interferon 技术领域Technical field
本发明涉及免疫学治疗领域。具体地,本发明涉及酪蛋白激酶2抑制剂在促进I型干扰素及其下游基因表达中的新用途。The invention relates to the field of immunotherapy. In particular, the invention relates to novel uses of casein kinase 2 inhibitors in promoting the expression of type I interferons and their downstream genes.
背景技术Background technique
I型干扰素包括IFNα和IFNβ两种,它们与细胞表面的干扰素受体结合后激活蛋白激酶Jak1和Tyk2,随后活化转录因子STAT1或/和STAT2,诱导上百种基因的表达(干扰素诱导基因,ISGs),包括MX1、MX2、Rsad2/Viperine、CXCL10/IP-10等。Type I interferons include IFNα and IFNβ, which bind to interferon receptors on the cell surface to activate protein kinases Jak1 and Tyk2, and then activate the transcription factors STAT1 or/and STAT2 to induce expression of hundreds of genes (interferon induction) Genes, ISGs), including MX1, MX2, Rsad2/Viperine, CXCL10/IP-10, etc.
干扰素诱导的这些基因在细胞的繁殖和凋亡、血管生成、T细胞免疫反应等过程中起着重要的生理作用。因此I型干扰素在免疫相关性疾病中的应用得到了广泛的研究。These genes induced by interferon play an important physiological role in cell proliferation and apoptosis, angiogenesis, and T cell immune response. Therefore, the application of type I interferon in immune-related diseases has been extensively studied.
I型干扰素的表达是受模式识别受体(PRRs,Pattern Recognition Receptors)及其介导的信号通路所诱导的。这些模式识别受体包括Toll样受体(Toll-like receptors/TLRs)、NOD样受体(NOD-like receptors/NLRs)、RIG-I样受体(RIG-I-like receptors/RLRs)和胞浆内DNA受体(cytosolic DNA sensors,CDSs)通过识别组织中死亡的细胞所分泌的RNA和DNA等“模式分子”(PAMPs,Pathogen-Associated Molecular Patterns),激活复杂的信号通路(包括蛋白激酶TBK1、IKKα/β和MAP kinases),活化关键性的转录因子IRF3、NFκB和AP-1等,诱导I型干扰素、细胞因子和趋化因子的表达,激活天然免疫和适应性免疫反应,实施宿主防御和组织修复等生理功能。The expression of type I interferons is induced by pattern recognition receptors (PRRs, Pattern Recognition Receptors) and their mediated signaling pathways. These pattern recognition receptors include Toll-like receptors (TLRs), NOD-like receptors (NLRs), RIG-I-like receptors (RLRs) and cells. Cytosolic DNA sensors (CDSs) activate complex signaling pathways (including protein kinase TBK1) by identifying pathway-associated Molecular Patterns (PAMPs) secreted by dead cells in tissues. , IKKα/β and MAP kinases), activate key transcription factors IRF3, NFκB and AP-1, induce the expression of type I interferons, cytokines and chemokines, activate natural immune and adaptive immune responses, and implement hosts. Physiological functions such as defense and tissue repair.
但是I型干扰素表达过量或异常也会引起疾病,特别是系统性红斑狼疮等自身免疫性疾病,因此体内往往存在调控I型干扰素的负调节机制,以避免干扰素的异常表达。这些调控机制往往导致在病理状态下,I型干扰素也不能有效的表达,从而导致各种免疫反应不足的相关性疾病。因此如何在病理状态下,突破机体调控I型干扰素表达的机制,从而有效地治疗免疫相关性疾病,是基础和临床医学领域研究的一个重要课题。However, excessive or abnormal expression of type I interferon may also cause diseases, especially autoimmune diseases such as systemic lupus erythematosus. Therefore, a negative regulation mechanism regulating type I interferon is often present in the body to avoid abnormal expression of interferon. These regulatory mechanisms often lead to the inability of type I interferons to be expressed efficiently under pathological conditions, leading to various diseases with insufficient immune response. Therefore, how to effectively control the immune-related diseases in the pathological state and break through the mechanism of the body's regulation of type I interferon expression is an important subject in the field of basic and clinical medicine.
因此,本领域迫切需要开发一种提高人体自身I型干扰素表达,提高人体免疫功能的有效方法,以治疗多种感染性和非感染性的人类疾病。 Therefore, there is an urgent need in the art to develop an effective method for improving the expression of human type I interferon and improving the immune function of the human body to treat various infectious and non-infectious human diseases.
发明内容Summary of the invention
本发明提供了一种CK2抑制剂在促进I型干扰素表达中的应用。The invention provides a use of a CK2 inhibitor for promoting expression of a type I interferon.
本发明第一方面,提供了一种酪蛋白激酶2(Casein kinase 2,CK2)抑制剂的用途,(i)用于制备促进I型干扰素(IFNα/β)和/或I型干扰素诱导基因或其蛋白表达的药物组合物;和/或(ii)用于制备治疗和/或预防I型干扰素缺乏相关疾病的药物组合物。In a first aspect of the invention, there is provided a use of a casein kinase 2 (CK2) inhibitor, (i) for the preparation of a type I interferon (IFNα/β) and/or type I interferon induction A pharmaceutical composition for expression of a gene or protein thereof; and/or (ii) a pharmaceutical composition for the preparation of a disease associated with the treatment and/or prevention of type I interferon deficiency.
在另一优选例中,所述的CK2来源于哺乳动物,较佳地,来源于人、小鼠、大鼠。In another preferred embodiment, the CK2 is derived from a mammal, preferably from a human, a mouse, or a rat.
在另一优选例中,所述的I型干扰素缺乏是指在疾病发生前和或/疾病发生后,I型干扰素表达量和/或活性的降低。In another preferred embodiment, the type I interferon deficiency refers to a decrease in the amount and/or activity of type I interferon expression before and/or after the onset of the disease.
在另一优选例中,所述的I型干扰素缺乏相关疾病是指缺乏I型干扰素会导致疾病的发生和/或加重、且提高I型干扰素的表达量和/或活性后能够对所述疾病起到治疗和/或预防作用的疾病。In another preferred embodiment, the type I interferon deficiency-associated disease means that the lack of type I interferon causes the occurrence and/or aggravation of the disease, and increases the expression level and/or activity of the type I interferon. The disease has a therapeutic and/or preventive effect.
在另一优选例中,所述的抑制剂包括CK2的反义核酸、抑制性microRNA、抗体或小分子化合物。In another preferred embodiment, the inhibitor comprises an antisense nucleic acid, an inhibitory microRNA, an antibody or a small molecule compound of CK2.
在另一优选例中,所述的抗体包括小分子多肽。In another preferred embodiment, the antibody comprises a small molecule polypeptide.
在另一优选例中,所述的抑制剂为CK2基因的siRNA、shRNA。In another preferred embodiment, the inhibitor is an siRNA of the CK2 gene, shRNA.
在另一优选例中,所述的CK2基因的shRNA如(SEQ ID NO.:1-2所示)In another preferred embodiment, the shRNA of the CK2 gene is as shown in (SEQ ID NO.: 1-2)
Forward(SEQ ID NO.:1):Forward (SEQ ID NO.: 1):
5’CCGGCCGAGTTGCTTCTCGATATTTCTCGAGAAATATCGAGAAGCAACTCGGTTTTTG 3’;5'CCGGCCGAGTTGCTTCTCGATATTTCTCGAGAAATATCGAGAAGCAACTCGGTTTTTG 3';
Reverse(SEQ ID NO.:2):Reverse (SEQ ID NO.: 2):
5’AATTCAAAAACCGAGTTGCTTCTCGATATTTCTCGAGAAATATCGAGAAGCAACTCGG 3’。5' AATTCAAAAACCGAGTTGCTTCTCGATATTTCTCGAGAAATATCGAGAAGCAACTCGG 3'.
在另一优选例中,所述的CK2蛋白的氨基酸序列的Genbank登录号为NP_031814.2(小鼠)和NP_808227.1(人)。In another preferred embodiment, the amino acid sequence of the CK2 protein has Genbank accession numbers NP_031814.2 (mouse) and NP_808227.1 (human).
在另一优选例中,所述的CK2蛋白的编码核苷酸序列的Genbank登录号如NM_007788.3(小鼠)和NM_177559(人)所示。In another preferred embodiment, the Genbank accession number of the nucleotide sequence encoding the CK2 protein is shown as NM_007788.3 (mouse) and NM_177559 (human).
在另一优选例中,所述的I型干扰素缺乏相关疾病包括:病毒感染性疾病、恶性肿瘤、多发性硬化。In another preferred embodiment, the type I interferon deficiency-related diseases include: viral infectious diseases, malignant tumors, multiple sclerosis.
在另一优选例中,所述的病毒感染性疾病包括感染了以下的病毒的疾病:单纯疱疹病毒(HSV)、水泡病毒(VSV)、卡波希氏瘤疱疹病毒(KSHV)、呼吸道合胞病 毒(RSV)、手足口病病毒(肠道病毒EV71和CA16)乙型肝炎病毒(HBV)、丙型肝炎病毒(HCV)、人类获得性免疫缺陷病毒/艾滋病病毒(HIV);和/或In another preferred embodiment, the viral infectious disease includes a disease infected with the following viruses: herpes simplex virus (HSV), vesicular virus (VSV), Kaposi's tumor herpesvirus (KSHV), respiratory syncytial disease Toxic (RSV), Hand, Foot and Mouth Disease Virus (Enterovirus EV71 and CA16) Hepatitis B Virus (HBV), Hepatitis C Virus (HCV), Human Acquired Immunodeficiency Virus/HIV (HIV); and/or
所述的恶性肿瘤包括以下肿瘤:多毛细胞白血病(hairy cell leukaemia)、慢性髓性白血病(CML)、淋巴瘤(lymphoma)、骨髓瘤(myeloma)、黑色素瘤(melanoma)、肾细胞癌与膀胱癌(renal cell and bladder cell carcinoma)、卡波希氏肉瘤(Kaposi’s sarcoma)。The malignant tumor includes the following tumors: hairy cell leukaemia, chronic myelogenous leukemia (CML), lymphoma, myeloma, melanoma, renal cell carcinoma and bladder cancer. (renal cell and bladder cell carcinoma), Kaposi's sarcoma.
在另一优选例中,所述的病毒为感染细胞后能够激活模式识别受体的病毒。In another preferred embodiment, the virus is a virus capable of activating a pattern recognition receptor after infecting a cell.
在另一优选例中,所述的抑制剂还用于抑制病毒和/或肿瘤细胞的免疫逃逸。In another preferred embodiment, the inhibitor is also useful for inhibiting immune escape of viral and/or tumor cells.
在另一优选例中,所述CK2抑制剂还用于抑制病毒的复制和组装。In another preferred embodiment, the CK2 inhibitor is also used to inhibit viral replication and assembly.
在另一优选例中,所述的I型干扰素和/或I型干扰素诱导基因或其蛋白的表达包括诱导产生和/或提高I型干扰素和/或I型干扰素诱导基因或其蛋白的表达量和/或活性。In another preferred embodiment, the expression of the type I interferon and/or type I interferon-inducing gene or protein thereof comprises inducing and/or increasing a type I interferon and/or type I interferon-inducing gene or The amount and/or activity of the protein.
在另一优选例中,所述的抑制剂为小分子化合物,较佳地,包括四溴苯三唑(TBB)、CX-4945(Silmitasertib)、DMAT(2-二甲氨基-4,5,6,7-四溴-1H-苯并咪唑)、山奈酚、酪氨酸磷酸化抑制剂AG114(Tyrphostin AG114)、四溴肉桂酸、3-[[5-(4-甲苯基)噻吩[2,3-d]嘧啶-4-yl]硫基]丙酸(TTP 22)、CX-5011、鞣花酸(Ellagic Acid)、3-甲基-1,6,8-三氢蒽醌(emodin)、4',5,7-三羟基黄酮(apigenin)。In another preferred embodiment, the inhibitor is a small molecule compound, preferably including tetrabromobenzotriazole (TBB), CX-4945 (Silmitasertib), DMAT (2-dimethylamino-4, 5, 6,7-tetrabromo-1H-benzimidazole), kaempferol, tyrosine phosphorylation inhibitor AG114 (Tyrphostin AG114), tetrabromocinnamic acid, 3-[[5-(4-tolyl)thiophene [2 ,3-d]pyrimidin-4-yl]thio]propionic acid (TTP 22), CX-5011, ellagic acid (Ellagic Acid), 3-methyl-1,6,8-trihydroanthracene (emodin) ), 4',5,7-trihydroxyflavone (apigenin).
在另一优选例中,所述的I型干扰素诱导基因或其蛋白包括CXCL10/IP-10、STAT1、MX1、MX2、Rsad2/Viperine、磷酸激酶TBK1、干扰素调节因子3(IRF3)。In another preferred embodiment, the type I interferon-inducing gene or protein thereof comprises CXCL10/IP-10, STAT1, MX1, MX2, Rsad2/Viperine, phosphokinase TBK1, interferon regulatory factor 3 (IRF3).
在另一优选例中,所述的I型IFN包括IFN-α、IFN-β。In another preferred embodiment, the type I IFN comprises IFN-α, IFN-β.
在另一优选例中,所述的药物组合物包括作为活性成分的CK2抑制剂,和药学上可接受的载体。In another preferred embodiment, the pharmaceutical composition comprises as an active ingredient a CK2 inhibitor, and a pharmaceutically acceptable carrier.
在另一优选例中,所述的CK2抑制剂的有效浓度为1-1000μM,较佳地,为20-100μM。In another preferred embodiment, the effective concentration of the CK2 inhibitor is from 1 to 1000 μM, preferably from 20 to 100 μM.
在另一优选例中,所述的CK2抑制剂对I型干扰素和/或I型干扰素诱导基因或其蛋白的促进作用呈剂量依赖性。In another preferred embodiment, the CK2 inhibitor is dose-dependently promoting the type I interferon and/or the type I interferon-inducing gene or its protein.
本发明第二方面,提供了一种体外非治疗性的促进细胞表达I型干扰素和/或I型干扰素诱导基因或其蛋白和/或体外非治疗性的抑制I型干扰素缺乏相关病毒细胞和/或肿瘤细胞生长的方法,包括步骤:在CK2抑制剂的存在下,培养细胞,从而促进细胞表达I型干扰素和/或I型干扰素诱导基因或其蛋白抑制I型干扰素 缺乏相关病毒细胞和/或肿瘤细胞生长。According to a second aspect of the present invention, there is provided a non-therapeutic promoting cell expressing a type I interferon and/or a type I interferon-inducing gene or a protein thereof and/or a non-therapeutic inhibitor of a type I interferon deficiency-associated virus in vitro. A method for growing cells and/or tumor cells, comprising the steps of: culturing cells in the presence of a CK2 inhibitor, thereby promoting expression of a type I interferon and/or a type I interferon-inducing gene or a protein thereof inhibiting type I interferon Lack of associated viral cells and/or tumor cell growth.
在另一优选例中,所述的细胞包括肿瘤细胞、或为DNA或RNA病毒感染的细胞。In another preferred embodiment, the cell comprises a tumor cell, or a cell infected with a DNA or RNA virus.
在另一优选例中,所述的方法还可用于制备I型干扰素或其诱导蛋白,例如收集并纯化细胞培养体系中获得的I型干扰素或其诱导蛋白。In another preferred embodiment, the method can also be used to prepare a type I interferon or an induced protein thereof, for example, to collect and purify a type I interferon obtained in a cell culture system or an induced protein thereof.
本发明第三方面,提供了一种筛选能够抑制CK2的化合物的方法,包括步骤:In a third aspect of the invention, there is provided a method of screening for a compound capable of inhibiting CK2, comprising the steps of:
(a)在试验组中,向细胞培养体系加入待测化合物,并测定I型干扰素的表达量和/或活性;在对照组中,向细胞培养体系不加入待测化合物,并测定I型干扰素的表达量和/或活性;(a) In the test group, the test compound was added to the cell culture system, and the expression amount and/or activity of the type I interferon was determined; in the control group, the test compound was not added to the cell culture system, and the type I was determined. The amount and/or activity of interferon expression;
其中,若试验组中I型干扰素的表达量和/或活性显著高于对照组,则说明所述的候选化合物为能够抑制CK2的化合物和/或Wherein, if the expression level and/or activity of the type I interferon in the test group is significantly higher than that of the control group, the candidate compound is a compound capable of inhibiting CK2 and/or
(a2)在试验组中,向病毒感染的细胞培养体系中加入待测化合物,并测定细胞培养体系中病毒的滴度;在对照组中,向病毒感染的细胞培养体系中加入待测化合物,并测定细胞培养体系中的病毒滴度;(a2) In the test group, the test compound is added to the virus-infected cell culture system, and the titer of the virus in the cell culture system is determined; in the control group, the test compound is added to the virus-infected cell culture system, And measuring the virus titer in the cell culture system;
其中,若试验组病毒的滴度显著低于对照组,则说明所述的候选化合物为能够抑制CK2的化合物。Wherein, if the titer of the test group virus is significantly lower than that of the control group, the candidate compound is a compound capable of inhibiting CK2.
在另一优选例中,所述的方法还包括步骤:In another preferred embodiment, the method further includes the steps of:
(b)对(a)中的化合物进一步测试其对酪蛋白激酶2或I型干扰素诱导基因或其蛋白的促进作用。(b) The compound in (a) is further tested for its promotion of the casein kinase 2 or type I interferon-inducible gene or its protein.
本发明第四方面,提供了一种制备I型干扰素的方法,包括步骤:According to a fourth aspect of the invention, a method for preparing a type I interferon is provided, comprising the steps of:
(i)在CK2抑制剂的存在下培养细胞;(i) culturing the cells in the presence of a CK2 inhibitor;
(ii)收集、分离并纯化细胞培养液中的I型干扰素。(ii) collecting, isolating and purifying type I interferons in the cell culture medium.
在另一优选例中,在L929细胞中加入CK2抑制剂,在非感染条件下培养12-48小时或者在病毒感染条件下培养12-24小时。收集上清,利用HPLC纯化I型干扰素。In another preferred embodiment, a CK2 inhibitor is added to L929 cells, cultured for 12-48 hours under non-infective conditions or 12-24 hours under viral infection conditions. The supernatant was collected and the type I interferon was purified by HPLC.
在另一优选例中,所述的细胞还包括成纤维细胞、或巨噬细胞。In another preferred embodiment, the cells further comprise fibroblasts, or macrophages.
本发明第五方面,提供了一种CK2基因或其蛋白的用途,用于制备降低I型干扰素和/或I型干扰素诱导基因或其蛋白表达的药物组合物。In a fifth aspect of the invention, there is provided a use of a CK2 gene or a protein thereof for the preparation of a pharmaceutical composition for reducing the expression of a type I interferon and/or a type I interferon-inducing gene or a protein thereof.
本发明第六方面,提供了一种治疗病毒性感染的方法,向需要治疗的对象施用CK2抑制剂,从而治疗病毒性感染。In a sixth aspect of the invention, a method of treating a viral infection is provided, wherein a CK2 inhibitor is administered to a subject in need of treatment to treat a viral infection.
在另一优选例中,所述需要治疗的对象为哺乳动物,较佳地,为人。In another preferred embodiment, the subject in need of treatment is a mammal, preferably a human.
本发明第七方面,提供了一种(i)治疗I型干扰素缺乏相关疾病;和/或(ii) 促进I型干扰素和/或I型干扰素诱导基因或其蛋白表达的药物组合物,所述的药物组合物包括作为活性成分的CK2抑制剂,和药学上可以接受的载体。According to a seventh aspect of the present invention, there is provided a (i) treatment of a type I interferon deficiency-related disease; and/or (ii) A pharmaceutical composition for promoting the expression of a type I interferon and/or a type I interferon-inducing gene or a protein thereof, the pharmaceutical composition comprising a CK2 inhibitor as an active ingredient, and a pharmaceutically acceptable carrier.
本发明第八方面,提供了一种判断CK2抑制剂是否对肿瘤或病毒有效的方法,包括步骤:According to an eighth aspect of the present invention, a method for determining whether a CK2 inhibitor is effective against a tumor or a virus is provided, comprising the steps of:
(a)向肿瘤细胞或病毒感染细胞培养体系中加入CK2抑制剂;(a) adding a CK2 inhibitor to a tumor cell or virus-infected cell culture system;
(b)观察加入CK2抑制剂前后所述培养体系中I型干扰素的表达量和/或活性;(b) observing the expression level and/or activity of type I interferon in the culture system before and after the addition of the CK2 inhibitor;
其中,当加入CK2抑制剂后I型干扰素的表达量和/或活性I1较加入之前的表达量和/或活性I0有显著升高,则说明CK2抑制剂对该肿瘤或病毒具有抑制作用。Among them, the expression level and/or activity I1 of type I interferon increased significantly compared with the expression level and/or activity I0 before the addition of the CK2 inhibitor, indicating that the CK2 inhibitor has an inhibitory effect on the tumor or virus.
应理解,在本发明范围内中,本发明的上述各技术特征和在下文(如实施例)中具体描述的各技术特征之间都可以互相组合,从而构成新的或优选的技术方案。限于篇幅,在此不再一一累述。It is to be understood that within the scope of the present invention, the various technical features of the present invention and the various technical features specifically described hereinafter (as in the embodiments) may be combined with each other to constitute a new or preferred technical solution. Due to space limitations, we will not repeat them here.
附图说明DRAWINGS
图1显示了CK2抑制Toll样受体信号所诱导的I型干扰素和/或I型干扰素诱导基因或其蛋白,但不影响炎症因子TNFα的表达。Figure 1 shows that CK2 inhibits the expression of the type I interferon and/or type I interferon-inducible gene or its protein induced by Toll-like receptor signaling, but does not affect the expression of the inflammatory factor TNFα.
图2显示了CK2抑制Toll样受体信号所诱导的TBK1和IRF3活化,但不影响MAP kinases的激活。Figure 2 shows that CK2 inhibits activation of TBK1 and IRF3 induced by Toll-like receptor signaling, but does not affect activation of MAP kinases.
图3显示了CK2抑制胞浆内识别RNA和DNA的模式识别受体所诱导的信号通路以及I型干扰素和/或I型干扰素诱导基因或其蛋白的表达。Figure 3 shows that CK2 inhibits the signaling pathway induced by pattern recognition receptors that recognize RNA and DNA in the cytosol and the expression of type I interferon and/or type I interferon-inducible genes or proteins thereof.
图4显示了HSV感染时,CK2抑制小鼠巨噬细胞系Raw表达I型干扰素和/或I型干扰素诱导基因或其蛋白,而随着I型干扰素及其靶基因产物的大量表达,HSV病毒的复制、组装和释放受到显著的抑制,在CK2α敲降的Raw264.7细胞培养液中检测到的HSV病毒滴度比对照组的要低接近10倍。Figure 4 shows that CK2 inhibits the expression of type I interferon and/or type I interferon-inducible genes or their proteins in mouse macrophage cell line by HSV infection, and is highly expressed with type I interferon and its target gene product. The replication, assembly and release of HSV virus were significantly inhibited, and the HSV virus titer detected in the CK2α knockdown Raw264.7 cell culture was nearly 10 times lower than that of the control group.
图5显示了仙台病毒感染时,CK2抑制小鼠成纤维细胞系L929表达I型干扰素和/或I型干扰素诱导基因或其蛋白。Figure 5 shows that CK2 inhibits the expression of type I interferon and/or type I interferon-inducible genes or proteins thereof in the mouse fibroblast cell line L929 when infected with Sendai virus.
图6显示了VSV病毒感染时,CK2抑制小鼠成纤维细胞系L929表达I型干扰素和/或I型干扰素诱导基因或其蛋白。Figure 6 shows that CK2 inhibits the expression of type I interferon and/or type I interferon-inducible genes or proteins thereof in the mouse fibroblast cell line L929 when VSV virus is infected.
图7显示了CK2的激酶活性在抑制病毒诱导的I型干扰素表达中起关键性的作用。Figure 7 shows that the kinase activity of CK2 plays a key role in inhibiting virus-induced expression of type I interferons.
图8显示了CK2抑制剂TBB在小鼠成纤维细胞系L929中诱导TBK1的活化和I 型干扰素以及I型干扰素诱导基因或其蛋白的表达。Figure 8 shows that CK2 inhibitor TBB induces TBK1 activation in mouse fibroblast cell line L929 and I Type of interferon and type I interferon-inducible gene or its protein expression.
图9显示了CK2抑制剂TBB在成纤维细胞系L929中抑制VSV病毒的感染和复制。Figure 9 shows that the CK2 inhibitor TBB inhibits infection and replication of VSV virus in the fibroblast cell line L929.
图10显示了CK2抑制剂TBB在人胚胎肾细胞系293中诱导TBK1的活化和I型干扰素以及I型干扰素诱导基因或其蛋白的表达,并抑制VSV的感染。Figure 10 shows that the CK2 inhibitor TBB induces activation of TBK1 and expression of type I interferons and type I interferon-inducible genes or proteins thereof in human embryonic kidney cell line 293, and inhibits infection of VSV.
图11显示了HCV感染人肝细胞系Hu7.5前,CK2抑制剂TBB预处理诱导I型干扰素和/或I型干扰素诱导基因或其蛋白表达并阻止HCV感染。Figure 11 shows that prior to HCV infection of the human hepatocyte line Hu7.5, pretreatment with the CK2 inhibitor TBB induces type I interferon and/or type I interferon-inducible genes or their protein expression and prevents HCV infection.
图12显示了HCV感染人肝细胞系Hu7.5后,CK2抑制剂TBB可以有效地抑制病毒复制。Figure 12 shows that the CK2 inhibitor TBB is effective in inhibiting viral replication following HCV infection of the human hepatocyte cell line Hu7.5.
图13显示了CK2抑制剂TBB在人原髓细胞白血病细胞(Human promyelocytic leukemia cells)HL-60中诱导I型干扰素以及I型干扰素诱导基因或其蛋白的表达。Figure 13 shows that the CK2 inhibitor TBB induces the expression of type I interferon and type I interferon-inducible genes or proteins thereof in human promyelocytic leukemia cells HL-60.
图14显示了CK2抑制剂TBB在人肾癌细胞OS-RC-2中诱导I型干扰素以及I型干扰素诱导基因或其蛋白的表达。Figure 14 shows that the CK2 inhibitor TBB induces the expression of type I interferons and type I interferon-inducible genes or proteins thereof in human renal cancer cell line OS-RC-2.
图15A-15B显示了经CK2抑制剂TBB对EV71病毒感染具有预防和治疗作用,且二者在高浓度下的效果更显著。Figures 15A-15B show the prophylactic and therapeutic effects of CK2 inhibitor TBB on EV71 virus infection, and both are more effective at high concentrations.
图16A-16B显示CK2抑制剂TBB有效地激活小鼠组织中诱导I型干扰素表达的关键性激酶TBK1,并抑制了HSV病毒的复制、组装和释放,而TBB处理组小鼠血液、脾脏中检测到的HSV病毒滴度显著低于对照组。Figures 16A-16B show that the CK2 inhibitor TBB effectively activates the key kinase TBK1 that induces type I interferon expression in mouse tissues and inhibits the replication, assembly and release of HSV virus in the blood and spleen of TBB-treated mice. The detected HSV virus titer was significantly lower than the control group.
具体实施方式detailed description
本发明人经过广泛而深入的研究,首次意外地发现,在复杂的人体免疫机制中,CK2蛋白参与调节多种模式识别受体信号通路,控制TBK1和IRF3的活化,限制I型干扰素的表达。实验证明,CK2的抑制能够有效诱导、促进I型干扰素的产生,并激活多种I型干扰素诱导基因或其蛋白的表达,从而改善人体的免疫防卫体系,能够有效阻止I型干扰素相关疾病,例如与I型干扰素相关的病毒、肿瘤,或治疗多发性硬化等疾病,成为一种新的提高人体免疫、治疗病毒感染性疾病和一些非感染性疾病的策略。在此基础上,完成了本发明。After extensive and intensive research, the present inventors discovered for the first time that in a complex human immune mechanism, CK2 protein is involved in regulating multiple pattern recognition receptor signaling pathways, controlling the activation of TBK1 and IRF3, and limiting the expression of type I interferon. . Experiments have shown that inhibition of CK2 can effectively induce and promote the production of type I interferon, and activate the expression of various type I interferon-inducible genes or their proteins, thereby improving the body's immune defense system and effectively preventing type I interferon-related Diseases, such as viruses associated with type I interferons, tumors, or the treatment of multiple sclerosis, have become a new strategy to improve human immunity, treat viral infectious diseases, and some non-infectious diseases. On the basis of this, the present invention has been completed.
术语the term
如本文所用,术语“I型干扰素缺乏”指的是在疾病发生前和/或疾病发生后, I型干扰素的表达量和/或活性的降低。例如,I型干扰素的表达量降低会导致多发性硬化和肿瘤等疾病的发生,或肿瘤或病毒的免疫逃逸抑制了正常I型干扰素的表达,从而进一步加重肿瘤或病毒等疾病的发生。As used herein, the term "type I interferon deficiency" refers to before and/or after the onset of a disease, A decrease in the amount and/or activity of type I interferon. For example, a decrease in the expression level of type I interferon may lead to the occurrence of diseases such as multiple sclerosis and tumors, or the immune escape of tumors or viruses inhibits the expression of normal type I interferons, thereby further aggravating the occurrence of diseases such as tumors or viruses.
如本文所用,术语“I型干扰素缺乏相关疾病”指的是缺乏I型干扰素会导致疾病的发生和/或加重、且提高I型干扰素的表达量和/或活性后能够对所述疾病起到治疗和/或预防作用的疾病。通常,本发明所指的“I型干扰素缺乏相关疾病”指的是I型干扰素缺乏相关的病毒感染性疾病、多发性硬化或恶性肿瘤,应理解,与“I型干扰素缺乏”无关的病毒性感染疾病或恶性肿瘤并不在本术语范围内。通常,判断该疾病(如肿瘤或病毒)是否与I型干扰素相关可以采用本领域常规技术,例如敲除或抑制I型干扰素后观察该疾病的预后(包括细胞生长)是否与I型干扰素的表达相关。As used herein, the term "type I interferon deficiency-associated disease" means that the absence of a type I interferon causes the occurrence and/or exacerbation of the disease, and increases the amount and/or activity of the type I interferon expression. A disease in which the disease acts as a therapeutic and/or preventive agent. Generally, the "type I interferon deficiency-associated disease" referred to in the present invention refers to a type I interferon deficiency-associated viral infectious disease, multiple sclerosis or malignant tumor, and it is understood that it is not related to "type I interferon deficiency". Viral infections or malignancies are not within the scope of this term. In general, it is known in the art to determine whether the disease (such as a tumor or virus) is associated with type I interferon, such as whether the prognosis (including cell growth) of the disease is interfered with type I interference after knocking or inhibiting type I interferon. The expression of the prime is related.
酪蛋白激酶2(CK2)Casein kinase 2 (CK2)
CK2(又称酪蛋白激酶2/Casein kinase 2)是普遍存在于真核细胞内的一种丝氨酸和苏氨酸激酶,主要以四聚体——包含两个催化亚基(α和/或α′)和两个调节亚基β的形式发挥功能。本发明所述的CK2来源于哺乳动物,通常来源于人、小鼠或大鼠。CK2 (also known as casein kinase 2) is a serine and threonine kinase ubiquitous in eukaryotic cells, mainly tetramers - containing two catalytic subunits (α and / or α ') and the form of two regulatory subunits β function. The CK2 of the present invention is derived from a mammal and is usually derived from human, mouse or rat.
一种优选的CK2蛋白的氨基酸序列的Genbank登录号为NP_808227.1(小鼠)和NP_031814.2(人)所示,其编码的核苷酸序列Genbank登录号为NM_007788.3(小鼠)和NM_177559(人)所示。CK2基因序列Genbank ID No.:12995(小鼠)和Genbank ID No.:1457(人)所示。The preferred amino acid sequence of the CK2 protein is shown in Genbank accession numbers NP_808227.1 (mouse) and NP_031814.2 (human), and the nucleotide sequence encoded by Genbank accession number is NM_007788.3 (mouse) and NM_177559 (person) is shown. The CK2 gene sequence is shown in Genbank ID No.: 12995 (mouse) and Genbank ID No.: 1457 (human).
目前,各种研究成果表明,CK2在调节细胞生长、肿瘤发生发展中具有重要作用。在肿瘤中,CK2的表达和活性往往异常增高。因此,有报道认为而抑制CK2的表达或活性能有效地抑制肿瘤细胞的生长、并诱导肿瘤细胞的坏死。CK2的抑制剂CX-4945已经进入临床II期,并显示了较好的抗肿瘤效果。然而,CK2抑制剂对肿瘤的治疗作用也具有一定的选择性表现。由于CK2的作用底物和机制比较多,目前尚无法获知CK2抑制剂在每一种肿瘤治疗中的具体作用机制,因此,目前也无法获得该选择性表现的解决方法。At present, various research results show that CK2 plays an important role in regulating cell growth and tumorigenesis. In tumors, the expression and activity of CK2 tend to increase abnormally. Therefore, it has been reported that inhibition of the expression or activity of CK2 can effectively inhibit the growth of tumor cells and induce necrosis of tumor cells. CX-4945, an inhibitor of CK2, has entered clinical phase II and shows a better anti-tumor effect. However, CK2 inhibitors also have a selective effect on the therapeutic effects of tumors. Because of the many substrates and mechanisms of CK2, the specific mechanism of action of CK2 inhibitors in each type of tumor treatment is not known. Therefore, the solution to this selective performance is not currently available.
本发明通过实验首次发现,CK2参与调节多种模式识别受体信号通路,控制TBK1和IRF3的活化,以此限制I型干扰素的表达。实验证明,抑制CK2能够提高I型干扰素及其诱导基因的表达,并对抑制病毒感染、多发性硬化和肿瘤具有 良好的效果。The present invention firstly found through experiments that CK2 is involved in the regulation of multiple pattern recognition receptor signaling pathways, and controls the activation of TBK1 and IRF3, thereby limiting the expression of type I interferon. Experiments have shown that inhibition of CK2 can increase the expression of type I interferon and its induced genes, and has the effect of inhibiting viral infection, multiple sclerosis and tumors. good effect.
具体地,本发明不仅从理论上揭示CK2是负调节TBK1和I型干扰素表达的关键分子,而且通过实验证明,利用小分子化合物抑制CK2的激酶活性可以有效地诱导I型干扰素产生,并由此对病毒感染、多发性硬化和肿瘤细胞的生长起到抑制和杀伤作用。Specifically, the present invention not only theoretically reveals that CK2 is a key molecule that negatively regulates the expression of TBK1 and type I interferon, but also experimentally demonstrates that inhibition of CK2 kinase activity by small molecule compounds can effectively induce type I interferon production, and Thereby inhibiting and killing viral infection, multiple sclerosis and tumor cell growth.
模式识别受体与I型干扰素表达的相关性Correlation between pattern recognition receptor and type I interferon expression
模式识别受体(PRRs,Pattern Recognition Receptors)通过识别死亡细胞或病毒分泌的RNA和DNA等“模式分子”(PAMPs,Pathogen-Associated Molecular Patterns),激活复杂的信号通路,诱导I型干扰素和细胞因子、趋化因子的表达,实施宿主防御和组织修复等生理功能。Pattern Recognition Receptors (PRRs) activate complex signaling pathways to induce type I interferons and cells by recognizing Pathogen-Associated Molecular Patterns (PAMPs, RNA and DNA secreted by dead cells or viruses). The expression of factors and chemokines, and the implementation of physiological functions such as host defense and tissue repair.
位于细胞膜上的Toll样受体TLR4识别病毒表面的糖蛋白,转运入内吞体(endosome);位于内吞体中的TLR3识别病毒双链DNA;TLR3/TLR4在内吞体中通过接头分子TRIF和TRAM,随后活化磷酸激酶TBK1(TANK-binding kinase 1),引起下游的转录因子IRF3的活化,从而诱导I型干扰素的表达。The Toll-like receptor TLR4 located on the cell membrane recognizes the glycoprotein on the surface of the virus and transports it into the endosome; TLR3 in the endosome recognizes the viral double-stranded DNA; TLR3/TLR4 passes through the linker molecule TRIF and in the endosome TRAM, which subsequently activates the phosphokinase TBK1 (TANK-binding kinase 1), causes activation of the downstream transcription factor IRF3, thereby inducing expression of type I interferon.
病毒产生的RNA或者机体的损伤细胞所释放的RNA能被细胞质中的RIG-I样受体RIG-I(retinoic acidinducible gene-I)和MDA5(melanoma differentiation-associated gene 5)识别,通过接头分子MAVS/VISA在线粒体上活化磷酸激酶TBK1和转录因子IRF3,也能诱导I型干扰素的表达。病毒或者损伤细胞释放的双链DNA(dsDNA)被细胞质中的多种DNA感受因子(DNA sensors),如cGAS、DDX41和IFI16等识别,通过接头分子STING激活TBK1和IRF3,诱导I型干扰素的表达。这些天然免疫受体也通过激活转录因子NFκB和AP1,诱导IL-6、TNFα和IL-1等促炎症因子和趋化因子的表达。The RNA released by the virus or the RNA released by the damaged cells of the body can be recognized by the RIG-I-like receptor RIG-I ( retinoic acid-ducible gene-I) and MDA5 (melanoma differentiation-associated gene 5) in the cytoplasm, and the linker molecule MAVS /VISA activates phosphokinase TBK1 and the transcription factor IRF3 on mitochondria and also induces type I interferon expression. Double-stranded DNA (dsDNA) released by viruses or damaged cells is recognized by various DNA receptors in the cytoplasm, such as cGAS, DDX41, and IFI16, and TBK1 and IRF3 are activated by the linker molecule STING to induce type I interferon. expression. These innate immune receptors also induce the expression of pro-inflammatory and chemokines such as IL-6, TNFα and IL-1 by activating the transcription factors NFκB and AP1.
例如,部分病毒感染或者机体细胞的死亡能引起模式识别受体信号通路激活,从而诱导I型干扰素IFNα/β表达。IFNα/β与干扰素受体结合后激活Jak1和Tyk2,活化转录因子STAT1,诱导上百种I型干扰素和/或I型干扰素诱导基因或其蛋白的表达,包括MX1、MX2、Rsad2/Viperine、CXCL10/IP-10等,实施宿主防御的功能,清除入侵的病毒和被感染的细胞,或者对组织损伤进行修复,清除病变或癌变的细胞。但是在长期进化的过程中,病毒也获得拮抗模式识别受体信号通路,抑制I型干扰素表达的能力,以逃避宿主的防御功能。例如,丙肝病毒HCV编码的蛋白酶NS3/4A能切割TLR3/4的接头分子Trif和RIG-I/MDA5的接头分子 MAVS/VISA,导致它们不能激活下游的TBK1和IRF3。肠道病毒EV71既能通过蛋白酶2A(pro)切割MAVS,又能通过3C蛋白切割Trif,从而抑制I型干扰素的表达。水泡病毒VSV的F蛋白和流感病毒的NS1通过抑制RIG-I的功能,也能干扰宿主表达I型干扰素。人类获得性免疫缺陷病毒/艾滋病病毒(HIV)在复制过程中,可以利用细胞的Trex1和SamHD1等分子降解或抑制反转录的双链DNA,阻碍I型干扰素的产生。通过这些机制,病毒对宿主的免疫反应产生逃逸现象,导致了病毒在宿主细胞中的急性或慢性感染,从而使宿主产生严重病变。此外,在多发性硬化和肿瘤形成过程中,机体抑制I型干扰素表达的机制也可能造成I型干扰素不表达,从而导致疾病的恶化。For example, partial viral infection or death of the body's cells can cause activation of the pattern recognition receptor signaling pathway, thereby inducing expression of type I interferon IFNα/β. Binding of IFNα/β to interferon receptor activates Jak1 and Tyk2, activates the transcription factor STAT1, and induces expression of hundreds of interferon- and/or type I interferon-inducible genes or proteins thereof, including MX1, MX2, Rsad2/ Viperine, CXCL10/IP-10, etc., implement host defense functions, remove invading viruses and infected cells, or repair tissue damage to remove diseased or cancerous cells. However, in the long-term evolution process, the virus also acquires the ability to antagonize the pattern recognition receptor signaling pathway and inhibit the expression of type I interferon to escape the host's defense function. For example, the hepatitis C virus HCV-encoded protease NS3/4A cleaves the linker molecule of TLR3/4, the linker molecule Trif and RIG-I/MDA5. MAVS/VISA, causing them to not activate downstream TBK1 and IRF3. Enterovirus EV71 can inhibit the expression of type I interferon by cleavage of MAVS by protease 2A (pro) and by cleavage of Trif by 3C protein. The F protein of vesicular virus VSV and the NS1 of influenza virus can also interfere with host expression of type I interferon by inhibiting the function of RIG-I. Human acquired immunodeficiency virus/HIV (HIV) can inhibit the production of type I interferon by using cells such as Trex1 and SamHD1 to degrade or inhibit reverse transcription of double-stranded DNA. Through these mechanisms, the immune response of the virus to the host produces an escape phenomenon, resulting in an acute or chronic infection of the virus in the host cell, causing the host to develop severe lesions. In addition, in the process of multiple sclerosis and tumor formation, the mechanism of inhibition of type I interferon expression may also cause type I interferon not to be expressed, leading to disease progression.
CK2抑制剂CK2 inhibitor
如本文所用,术语“本发明活性成分”、“本发明抑制剂”、“本发明CK2抑制剂”可互换使用,均指能够降低CK2基因或蛋白表达量和/或活性的物质。As used herein, the terms "active ingredient of the present invention", "inhibitor of the present invention", and "inhibitor of CK2 of the present invention" are used interchangeably and mean a substance capable of reducing the amount and/or activity of expression of a CK2 gene or protein.
本发明实验证明,CK2抑制剂能够有效地阻断CK2的表达量和/或活性,从而诱导或促进I型干扰素、TBK1、IRF3的表达,以达到治疗病毒性感染、多发性硬化和肿瘤的目的。The experiments of the present invention prove that the CK2 inhibitor can effectively block the expression amount and/or activity of CK2, thereby inducing or promoting the expression of type I interferon, TBK1, and IRF3, in order to achieve treatment of viral infection, multiple sclerosis and tumor. purpose.
可用于本发明的CK2抑制剂没有特殊限制,可以为任何降低CK2基因或蛋白表达量和/或活性的物质。代表性的例子包括CK2基因的反义核酸、抑制CK2表达的microRNA(siRNA)、CK2蛋白的抗体、抑制CK2对TBK1作用的多肽或对CK2表达量和/或活性具有抑制作用的小分子化合物。The CK2 inhibitor which can be used in the present invention is not particularly limited and may be any substance which reduces the expression amount and/or activity of the CK2 gene or protein. Representative examples include antisense nucleic acids of the CK2 gene, microRNAs (siRNA) that inhibit CK2 expression, antibodies against the CK2 protein, polypeptides that inhibit the action of CK2 on TBK1, or small molecule compounds that have an inhibitory effect on the amount and/or activity of CK2 expression.
通常,根据CK2基因序列,本领域技术人员可以通过常规技术手段设计并合成具有CK2基因抑制作用的反义核酸或miRNA。而具有CK2抑制功能的小分子化合物则可从市售或人工合成制备获得。优选的例子有四溴苯三唑(TBB,购自Tocris)、DMAT(2-dimethylamino-4,5,6,7-tetrabromo-1H-benzimidazol,购自MedChem Express)、山奈酚、酪氨酸磷酸化抑制剂AG114(Tyrphostin AG114,购自Alexis Biochemicals)、CX-4945(Silmitasertib,购自Selleck)、四溴肉桂酸、3-[[5-(4-甲苯基)噻吩[2,3-d]嘧啶-4-yl]硫基]丙酸(TTP 22,购自Tocris)、CX-5011、鞣花酸(Ellagic Acid,购自MedChem Express)、3-甲基-1,6,8-三氢蒽醌(emodin,购自Sigma)、4',5,7-三羟基黄酮(apigenin,购自上海化成工业发展有限公司)。Generally, according to the CK2 gene sequence, one skilled in the art can design and synthesize an antisense nucleic acid or miRNA having a CK2 gene inhibitory effect by conventional techniques. Small molecule compounds having CK2 inhibitory properties can be obtained commercially or synthetically. Preferred examples are tetrabromobenzotriazole (TBB, available from Tocris), DMAT (2-dimethylamino-4, 5,6,7-tetrabromo-1H-benzimidazol, available from MedChem Express), kaempferol, tyrosine phosphate Inhibitor AG114 (Tyrphostin AG114, available from Alexis Biochemicals), CX-4945 (Silmitasertib, available from Selleck), tetrabromocinnamic acid, 3-[[5-(4-methylphenyl)thiophene [2,3-d] Pyrimidine-4-yl]thio]propionic acid (TTP 22, available from Tocris), CX-5011, ellagic acid (Ellagic Acid, available from MedChem Express), 3-methyl-1,6,8-trihydrogen Emo (emodin, purchased from Sigma), 4',5,7-trihydroxyflavone (apigenin, purchased from Shanghai Chemical Industry Development Co., Ltd.).
优选地,本发明CK2抑制剂还包括含有CK2抑制活性的shRNA的表达载体,其 可通过常规方法制备获得。一种优选的具有CK2抑制活性的shRNA序列如下:Preferably, the CK2 inhibitor of the present invention further comprises an expression vector comprising shRNA having CK2 inhibitory activity, It can be obtained by a conventional method. A preferred shRNA sequence having CK2 inhibitory activity is as follows:
Forward(SEQ ID NO.:1):Forward (SEQ ID NO.: 1):
5’CCGGCCGAGTTGCTTCTCGATATTTCTCGAGAAATATCGAGAAGCAACTCGGTTTTTG 3’5'CCGGCCGAGTTGCTTCTCGATATTTCTCGAGAAATATCGAGAAGCAACTCGGTTTTTG 3’
Reverse(SEQ ID NO.:2):Reverse (SEQ ID NO.: 2):
5’AATTCAAAAACCGAGTTGCTTCTCGATATTTCTCGAGAAATATCGAGAAGCAACTCGG 3’5’AATTCAAAAACCGAGTTGCTTCTCGATATTTCTCGAGAAATATCGAGAAGCAACTCGG 3’
可用于本发明的CK2抑制剂的有效治疗浓度范围可以根据本领域常用的有效浓度筛选方法进行筛选,从而获得安全有效的施用剂量,通常,CK2抑制剂的有效浓度为1-1000μM,较佳地为20-100μM。The effective therapeutic concentration range of the CK2 inhibitor useful in the present invention can be screened according to an effective concentration screening method commonly used in the art, thereby obtaining a safe and effective administration dose. Usually, the effective concentration of the CK2 inhibitor is 1-1000 μM, preferably It is 20-100 μM.
I型干扰素Type I interferon
I型干扰素包括IFNα和IFNβ两种,它们与细胞表面的干扰素受体结合后激活蛋白激酶Jak1和Tyk2,随后活化转录因子STAT1或/和STAT2,诱导上百种基因的表达(干扰素诱导基因,ISGs),包括MX1、MX2、Rsad2/Viperine、CXCL10/IP-10等。干扰素诱导的这些基因在抗病毒感染、调节细胞的繁殖和凋亡、血管生成、T细胞免疫反应等过程中起着重要的生理作用。因此I型干扰素在治疗病毒性感染、免疫相关性疾病和肿瘤中的应用得到了广泛的研究。Type I interferons include IFNα and IFNβ, which bind to interferon receptors on the cell surface to activate protein kinases Jak1 and Tyk2, and then activate the transcription factors STAT1 or/and STAT2 to induce expression of hundreds of genes (interferon induction) Genes, ISGs), including MX1, MX2, Rsad2/Viperine, CXCL10/IP-10, etc. These genes induced by interferon play an important physiological role in antiviral infection, regulation of cell proliferation and apoptosis, angiogenesis, and T cell immune response. Therefore, the use of type I interferons in the treatment of viral infections, immune-related diseases and tumors has been extensively studied.
目前,明确在接受I型干扰素治疗后有效的适应症包括多种病毒感染,例如乙型和丙型肝炎病毒的感染,多发性硬化和十几种肿瘤,包括白血病、淋巴瘤、骨髓瘤、黑色素瘤、肾细胞瘤、膀胱瘤和卡波西氏肉瘤等。干扰素抑制多发性硬化的机制可能涉及到其对NLRP3介导的炎症小体的活性的抑制,而干扰素抑制肿瘤的机制涉及到激活抗肿瘤免疫、抑制肿瘤细胞的繁殖,诱导肿瘤细胞的凋亡等。因此,本领域技术人员可以合理预期,利用本发明CK2抑制剂促进I型干扰素的表达后,可以治疗各种与I型干扰素缺乏的疾病,例如,上述的病毒感染、多发性硬化和肿瘤等等。At present, the indications that are effective after receiving type I interferon therapy include a variety of viral infections, such as hepatitis B and C infection, multiple sclerosis and more than a dozen tumors, including leukemia, lymphoma, myeloma, Melanoma, renal cell tumor, bladder tumor, and Kaposi's sarcoma. The mechanism by which interferon inhibits multiple sclerosis may involve its inhibition of NLRP3-mediated inflammatory stimuli, and the mechanism by which interferon inhibits tumors involves activating anti-tumor immunity, inhibiting tumor cell proliferation, and inducing tumor cell dying. Dead and so on. Therefore, those skilled in the art can reasonably expect that the use of the CK2 inhibitor of the present invention to promote the expression of type I interferon can treat various diseases which are deficient in type I interferon, for example, the above-mentioned viral infection, multiple sclerosis and tumor and many more.
I型干扰素诱导基因或其蛋白Type I interferon-inducible gene or protein thereof
如本文所用,术语“I型干扰素诱导基因或其蛋白”指的是当I型干扰素表达改变时,在I型干扰素上、下游中与I型干扰素表达量成相关性的基因或其蛋白,通常为感染、免疫相关的炎症因子或抗病毒因子。优选地,所述的I型干扰素诱导基因或其蛋白包括活化磷酸激酶(TBK1)、干扰素调节因子3(IRF3)、MX1、MX2、Rsad2/Viperine、或CXCL10/IP-10等各种免疫调节因子和炎症趋化因子。 As used herein, the term "type I interferon-inducible gene or protein thereof" refers to a gene that is associated with type I interferon expression on a type I interferon or downstream, when type I interferon expression is altered. Its protein, usually an infection, an immune-related inflammatory factor or an antiviral factor. Preferably, the type I interferon-inducing gene or protein thereof comprises various antibodies such as activated phosphokinase (TBK1), interferon regulatory factor 3 (IRF3), MX1, MX2, Rsad2/Viperine, or CXCL10/IP-10. Regulatory factors and inflammatory chemokines.
病毒性感染Viral infection
如本文所用,术语“病毒性感染”、“病毒感染性疾病”可互换使用,均指在感染细胞后能够激活模式识别受体的病毒感染相关疾病。As used herein, the terms "viral infection", "viral infectious disease" are used interchangeably and refer to a viral infection-associated disease capable of activating a pattern recognition receptor upon infection of a cell.
本发明抑制剂可针对各种病毒引起的病毒性感染,包括多种DNA或RNA病毒。例如,单纯疱疹病毒(HSV)、水泡病毒(VSV)、卡波希氏瘤疱疹病毒(KSHV)、呼吸道合胞病毒(RSV)、手足口病病毒(肠道病毒EV71和CA16)乙型肝炎病毒(HBV)、丙型肝炎病毒(HCV)、人类获得性免疫缺陷病毒/艾滋病病毒(HIV)。The inhibitors of the invention are directed against viral infections caused by various viruses, including a variety of DNA or RNA viruses. For example, herpes simplex virus (HSV), vesicular virus (VSV), Kaposi's tumor virus (KSHV), respiratory syncytial virus (RSV), hand, foot and mouth disease virus (enteric virus EV71 and CA16) hepatitis B virus (HBV), hepatitis C virus (HCV), human acquired immunodeficiency virus/HIV (HIV).
病毒感染机体后,通过激活多种模式识别受体,启动复杂的信号转导通路(包括蛋白激酶TBK1、IKKα/β和MAP kinases),活化关键性的转录因子IRF3、NFκB和AP-1等,诱导I型干扰素、细胞因子和趋化因子的表达,激活天然免疫和适应性免疫反应,实施宿主防御和组织修复等生理功能。例如,I型干扰素能直接引起被感染细胞的凋亡、炎症病毒复制、组装和释放。此外,杀伤性的NK和NKT细胞、CD8T细胞,均在抗病毒效果中起到了各种不同的作用。After the virus infects the body, it activates complex signal transduction pathways (including protein kinases TBK1, IKKα/β and MAP kinases) by activating multiple pattern recognition receptors, and activates key transcription factors IRF3, NFκB and AP-1. Induces the expression of type I interferons, cytokines and chemokines, activates innate and adaptive immune responses, and performs physiological functions such as host defense and tissue repair. For example, type I interferons can directly cause apoptosis, inflammatory viral replication, assembly, and release of infected cells. In addition, killer NK and NKT cells and CD8 T cells play various roles in antiviral effects.
可用于本发明的病毒为多种能够在感染状态下抑制I型干扰素表达的病毒,例如,病毒包括乙型和丙型肝炎病毒、肠道病毒EV71、人类获得性免疫缺陷病毒/艾滋病病毒(HIV)、单纯疱疹病毒(HSV)、水泡病毒(VSV)、卡波希氏瘤疱疹病毒(KSHV)、呼吸道合胞病毒(RSV)等。The virus which can be used in the present invention is a plurality of viruses capable of inhibiting the expression of type I interferon in an infectious state, for example, viruses including hepatitis B and C viruses, enterovirus EV71, human acquired immunodeficiency virus/HIV ( HIV), herpes simplex virus (HSV), vesicular virus (VSV), Kaposi's tumor herpesvirus (KSHV), respiratory syncytial virus (RSV), and the like.
应用application
目前,本发明利用相应的小分子化合物在体内逐步诱导适量的干扰素表达,将成为治疗这些拮抗干扰素表达的病毒性疾病的理想方案。还有这些病毒在复制过程中往往很容易产生病毒基因突变,因此一些针对病毒基因产物设计的抗病毒药物虽然暂时有效,但广泛应用后会很快导致抗性毒株的出现。而针对宿主蛋白设计的小分子药物则不会遇到类似的问题。因此同目前一些针对HCV和HIV病毒设计的药物相比,针对宿主蛋白设计的诱导I型干扰素表达的策略更为理想。At present, the present invention utilizes corresponding small molecule compounds to gradually induce an appropriate amount of interferon expression in vivo, and will be an ideal solution for treating these viral diseases which antagonize the expression of interferon. Moreover, these viruses are often prone to mutations in viral genes during replication. Therefore, some antiviral drugs designed for viral gene products are temporarily effective, but they can quickly lead to the emergence of resistant strains after extensive application. Small molecule drugs designed for host proteins will not encounter similar problems. Therefore, the strategy for inducing expression of type I interferon against host proteins is more desirable than currently designed for HCV and HIV.
因此,利用CK2抑制剂对I型干扰素的促进作用,可将CK2抑制剂应用于各种治疗或非治疗性的与I型干扰素缺乏相关的病毒感染、多发性硬化或肿瘤中。Thus, the use of CK2 inhibitors to promote type I interferon can be applied to a variety of therapeutic or non-therapeutic viral infections, multiple sclerosis or tumors associated with type I interferon deficiency.
本发明可以利用CK2抑制剂在体外来诱导或促进I型干扰素的表达,收集或纯化细胞培养体系中所获得的I型干扰素,从而提供一种制备I型干扰素的方法。通常在细胞培养体系中产生了I型干扰素后,对I型干扰素的收集、纯化的方法 均为本领域技术人员已知的常规实验方法。The present invention can utilize a CK2 inhibitor to induce or promote the expression of type I interferon in vitro, collect or purify the type I interferon obtained in the cell culture system, thereby providing a method for preparing type I interferon. Method for collecting and purifying type I interferon after type I interferon is usually produced in a cell culture system All are routine experimental methods known to those skilled in the art.
此外,还可以利用CK2抑制剂来筛选、识别I型干扰素缺乏相关疾病的候选药物;本发明还可以采用CK2抑制剂与I型干扰素之间的关系来判断特定的肿瘤、多发性硬化或病毒是否具有抑制作用(敏感性测定)。In addition, CK2 inhibitors can also be used to screen for and identify candidate drugs for type I interferon deficiency-related diseases; the present invention can also use the relationship between CK2 inhibitors and type I interferons to determine a specific tumor, multiple sclerosis or Whether the virus has an inhibitory effect (sensitivity assay).
在体内,可向所需的对象施用CK2抑制剂来减少所感染病毒的复制、组装从而降低I型干扰素相关性病毒的感染,或减少I型干扰素相关性的多发性硬化和肿瘤的发生和发展。In vivo, CK2 inhibitors can be administered to a desired subject to reduce replication and assembly of the infected virus, thereby reducing infection with type I interferon-associated virus, or reducing type I interferon-related multiple sclerosis and tumorigenesis. And development.
当然,利用本发明发现的CK2与I型干扰素的相关性,还可以利用CK2本身建立抑制I型干扰素的动物模型等。Of course, by using the correlation between CK2 and type I interferon discovered by the present invention, it is also possible to establish an animal model for inhibiting type I interferon by using CK2 itself.
药物组合物及施用方法Pharmaceutical composition and method of administration
如本文所用,术语“有效量”或“有效剂量”是指可对人和/或动物产生功能或活性的且可被人和/或动物所接受的量。As used herein, the term "effective amount" or "effective amount" refers to an amount that can produce a function or activity on a human and/or animal and that can be accepted by a human and/or animal.
如本文所用,术语“药学上可接受的”的成分是适用于人和/或哺乳动物而无过度不良副反应(如毒性、刺激和变态反应)的,即具有合理的效益/风险比的物质。术语“药学上可接受的载体”指用于治疗剂给药的载体,包括各种赋形剂和稀释剂。As used herein, the term "pharmaceutically acceptable" ingredient is a substance that is suitable for use in humans and/or mammals without excessive adverse side effects (eg, toxicity, irritation, and allergies), ie, a substance having a reasonable benefit/risk ratio. . The term "pharmaceutically acceptable carrier" refers to a carrier for the administration of a therapeutic agent, including various excipients and diluents.
本发明的药物组合物含有安全有效量的本发明的活性成分以及药学上可接受的载体。这类载体包括(但并不限于):盐水、缓冲液、葡萄糖、水、甘油、乙醇、及其组合。通常药物制剂应与给药方式相匹配,本发明的药物组合物的剂型为注射剂、口服制剂(片剂、胶囊、口服液)、透皮剂、缓释剂。例如用生理盐水或含有葡萄糖和其他辅剂的水溶液通过常规方法进行制备。所述的药物组合物宜在无菌条件下制造。The pharmaceutical compositions of the present invention comprise a safe and effective amount of the active ingredient of the present invention together with a pharmaceutically acceptable carrier. Such carriers include, but are not limited to, saline, buffer, dextrose, water, glycerol, ethanol, and combinations thereof. Generally, the pharmaceutical preparation should be matched with the administration mode, and the pharmaceutical composition of the present invention is in the form of an injection, an oral preparation (tablet, capsule, oral liquid), a transdermal agent, and a sustained release agent. For example, it is prepared by a conventional method using physiological saline or an aqueous solution containing glucose and other adjuvants. The pharmaceutical composition is preferably manufactured under sterile conditions.
本发明所述的活性成分的有效量可随给药的模式和待治疗的疾病的严重程度等而变化。优选的有效量的选择可以由本领域普通技术人员根据各种因素来确定(例如通过临床试验)。所述的因素包括但不限于:所述的活性成分的药代动力学参数例如生物利用率、代谢、半衰期等;患者所要治疗的疾病的严重程度、患者的体重、患者的免疫状况、给药的途径等。通常,当本发明的活性成分每天以约0.00001mg-50mg/kg动物体重(较佳的0.0001mg-10mg/kg动物体重)的剂量给予,能得到令人满意的效果。例如,由治疗状况的迫切要求,可每天给予若干次分开的剂量,或将剂量按比例地减少。 The effective amount of the active ingredient of the present invention may vary depending on the mode of administration and the severity of the disease to be treated and the like. The selection of a preferred effective amount can be determined by one of ordinary skill in the art based on various factors (e.g., by clinical trials). The factors include, but are not limited to, pharmacokinetic parameters of the active ingredient such as bioavailability, metabolism, half-life, etc.; severity of the disease to be treated by the patient, body weight of the patient, immune status of the patient, administration Ways, etc. In general, when the active ingredient of the present invention is administered at a dose of about 0.00001 mg to 50 mg/kg of animal body weight per day (preferably 0.0001 mg to 10 mg/kg of animal body weight), a satisfactory effect can be obtained. For example, several separate doses may be administered per day, or the dose may be proportionally reduced, as is critical to the condition of the treatment.
本发明所述的药学上可接受的载体包括(但不限于):水、盐水、脂质体、脂质、蛋白、蛋白-抗体缀合物、肽类物质、纤维素、纳米凝胶、或其组合。载体的选择应与给药方式相匹配,这些都是本领域的普通技术人员所熟知的。Pharmaceutically acceptable carriers of the invention include, but are not limited to, water, saline, liposomes, lipids, proteins, protein-antibody conjugates, peptide materials, cellulose, nanogels, or Its combination. The choice of carrier should be compatible with the mode of administration, which are well known to those of ordinary skill in the art.
本发明有益效果Advantageous effects of the invention
本发明发现,CK2参与调节多种模式识别受体信号通路,控制TBK1和IRF3的活化,以此限制I型干扰素的表达。实验证明过TBB等小分子化合物抑制CK2的激酶活性可以有效地诱导I型干扰素产生,从而阻止I型干扰素相关性疾病的发生和发展,为多发性硬化、病毒或肿瘤免疫逃逸提供了新的治疗方法。而针对宿主的内源性提高I型干扰素的方法,能够克服外源注射干扰素难以控制使用量而造成严重毒副作用的缺陷,使该治疗方法更有效、更安全。The present inventors have found that CK2 is involved in the regulation of multiple pattern recognition receptor signaling pathways and controls the activation of TBK1 and IRF3, thereby limiting the expression of type I interferons. Experiments have shown that inhibition of CK2 kinase activity by small molecule compounds such as TBB can effectively induce type I interferon production, thereby preventing the occurrence and development of type I interferon-related diseases, and providing new immune sclerosis for multiple sclerosis, virus or tumor. The treatment. The method of endogenously increasing type I interferon against the host can overcome the defect that the exogenous injection of interferon is difficult to control the amount of use and cause serious side effects, making the treatment method more effective and safer.
下面结合具体实施例,进一步阐述本发明。应理解,这些实施例仅用于说明本发明而不用于限制本发明的范围。下列实施例中未注明具体条件的实验方法,通常按照常规条件,例如Sambrook等人,分子克隆:实验室手册(New York:Cold Spring Harbor Laboratory Press,1989)中所述的条件,或按照制造厂商所建议的条件。除非另外说明,否则百分比和份数是重量百分比和重量份数。The invention is further illustrated below in conjunction with specific embodiments. It is to be understood that the examples are not intended to limit the scope of the invention. Experimental methods in which the specific conditions are not indicated in the following examples are generally carried out according to the conditions described in conventional conditions, for example, Sambrook et al., Molecular Cloning: Laboratory Manual (New York: Cold Spring Harbor Laboratory Press, 1989), or according to the manufacturing conditions. The conditions recommended by the manufacturer. Unless otherwise stated, percentages and parts are by weight and parts by weight.
实施例1蛋白激酶CK2参与调控多种模式识别受体诱导的I型干扰素表达Example 1 Protein kinase CK2 is involved in the regulation of type I interferon expression induced by multiple pattern recognition receptors
方法:利用Lenti-viral载体pLKO.1构建表达对照shRNA或针对CK2α的shRNA的重组病毒载体,然后将它们分别转染巨噬细胞系(Raw264.7)和成纤维细胞系(L929),建立了CK2α低表达的稳转细胞系。Methods: Lenti-viral vector pLKO.1 was used to construct recombinant viral vectors expressing control shRNA or shRNA against CK2α, and then transfected into macrophage cell line (Raw264.7) and fibroblast cell line (L929), respectively. A stably transfected cell line with low expression of CK2α.
首先根据NCBI gene bank中的CK2基因序列设计针对CK2α的shRNA寡核苷酸序列:First, the shRNA oligonucleotide sequence against CK2α was designed based on the CK2 gene sequence in the NCBI gene bank:
Forward(SEQ ID NO.:1):Forward (SEQ ID NO.: 1):
CCGGCCGAGTTGCTTCTCGATATTTCTCGAGAAATATCGAGAAGCAACTCGGTTTTTG,CCGGCCGAGTTGCTTCTCGATATTTCTCGAGAAATATCGAGAAGCAACTCGGTTTTTG,
Reverse(SEQ ID NO.:2):Reverse (SEQ ID NO.: 2):
AATTCAAAAACCGAGTTGCTTCTCGATATTTCTCGAGA AATATCGAGAAGCAACTCGGAATTCAAAAACCGAGTTGCTTCTCGATATTTCTCGAGA AATATCGAGAAGCAACTCGG
利用Addgene公司的商业化pLKO.1–TRC Cloning Vector载体(Addgene质粒商品号10878)及其标准操作流程,构建表达对照shRNA或针对CK2α的shRNA的质粒pLKO.1-shRNA。将10μg/ml对照shRNA或针对CK2α的shRNA质粒 pLKO.1-shRNA分别与2.5μg/ml pMD2.G(Addgene质粒商品号12259)、7.5μg/ml psPAX2(Addgene质粒商品号12260)一起通过磷酸钙(125mM Cacl2/1x HEPES)转化法在HEK293细胞中包装,48小时后收集细胞上清液,得到对照shRNA或针对CK2α的shRNA的重组病毒载体(HEK293细胞培养基为:DMEM完全培养基,含10%FBS和1%青霉素/链霉素)。Plasmid pLKO.1-shRNA expressing the control shRNA or shRNA against CK2α was constructed using Addgene's commercialized pLKO.1 - TRC Cloning Vector vector (Addgene Plasmid No. 10878) and its standard protocol. 10 μg/ml control shRNA or shRNA plasmid against CK2α pLKO.1-shRNA was transformed into HEK293 cells by calcium phosphate (125 mM Cacl2/1x HEPES) transformation with 2.5 μg/ml pMD2.G (Addgene plasmid product number 12259) and 7.5 μg/ml psPAX2 (Addgene plasmid product number 12260), respectively. In the middle package, the cell supernatant was collected 48 hours later to obtain a control shRNA or a recombinant viral vector against shRNA of CK2α (HEK293 cell culture medium: DMEM complete medium containing 10% FBS and 1% penicillin/streptomycin).
使用对照shRNA或针对CK2α的shRNA的重组病毒载体,以50%的浓度分别转染70%丰度的巨噬细胞系(Raw264.7购自美国ATCC)或者成纤维细胞系(L929购自美国ATCC),Raw264.7和L929细胞培养基为DMEM完全培养基。转染4小时后去除重组病毒载体,继续培养直至细胞丰度达到95%。以1:5的比例进行细胞传代,由于对照shRNA或针对CK2α的shRNA均带有puromycin抗性位点,传代后使用含有4μg/ml puromycin的DMEM完全培养基进行抗性细胞筛选。筛选3-4代(约7-10天)即可获得CK2α基因敲降的阳性稳转细胞系和对照细胞系。A 70% abundance macrophage cell line (Raw264.7 purchased from the US ATCC) or a fibroblast cell line (L929 purchased from the US ATCC) was transfected with a control shRNA or a recombinant viral vector directed against shRNA of CK2α at 50%. ), Raw264.7 and L929 cell culture media are DMEM complete medium. The recombinant viral vector was removed after 4 hours of transfection and culture continued until the cell abundance reached 95%. Cell passage was performed at a ratio of 1:5. Since the control shRNA or shRNA against CK2α had a puromycin resistance site, resistant cells were screened after passage using DMEM complete medium containing 4 μg/ml puromycin. A positive stable cell line and a control cell line of CK2α gene knockdown can be obtained by screening 3-4 generations (about 7-10 days).
将对照和CK2α敲降的Raw 264.7稳转细胞系以106个/ml分别接种到6孔板中,每孔接种2ml。分别用LPS(100nM,购自Invivogen)、polyI:C(50μg/ml,购自Invivogen)处理细胞,分别激活TLR4或TLR3信号通路(图1和图2)。刺激4小时后收集细胞,提取RNA(图1);或刺激15分钟至4小时后,于不同的时间点收集细胞,制备蛋白抽提液(图2)。The Raw 264.7 stable cell line knocked down by control and CK2α was inoculated into 6-well plates at 10 6 /ml, and 2 ml was inoculated per well. Cells were treated with LPS (100 nM, purchased from Invivogen), polyI:C (50 μg/ml, purchased from Invivogen), respectively, to activate the TLR4 or TLR3 signaling pathways (Figures 1 and 2). After 4 hours of stimulation, cells were harvested and RNA was extracted (Fig. 1); or after 15 to 4 hours of stimulation, cells were collected at different time points to prepare a protein extract (Fig. 2).
将对照和CK2α敲降的L929稳转细胞系以106个/ml分别接种到6孔板中,每孔接种2ml。分别用脂质体lipofectamine 2000(购自Invitrogen)转染polydA:dT(1μg/ml,购自Invivogen)、polyI:C(1μg/ml,购自Invivogen)细胞,分别激活RIG-I/Mda5和cGAS/STING信号通路(图3)。或者用DMXAA(100μg/ml,购自Selleck)直接处理细胞激活STING信号通路(图3)。刺激6小时后收集细胞,提取RNA(图3A);或刺激3小时后,收集细胞,制备蛋白抽提液或胞浆、胞核蛋白抽提液(图3B和3C)。Control and CK2α knockdown L929 stable cell lines were inoculated into 6-well plates at 10 6 /ml, and 2 ml was inoculated per well. PolydA:dT (1 μg/ml, purchased from Invivogen) and polyI:C (1 μg/ml, purchased from Invivogen) cells were transfected with lipofect lipofectamine 2000 (purchased from Invitrogen) to activate RIG-I/Mda5 and cGAS, respectively. /STING signal path (Figure 3). Alternatively, cells were directly treated with DMXAA (100 μg/ml, purchased from Selleck) to activate the STING signaling pathway (Figure 3). The cells were harvested 6 hours after stimulation, and RNA was extracted (Fig. 3A); or after 3 hours of stimulation, the cells were collected to prepare a protein extract or a cytoplasmic, nuclear protein extract (Figs. 3B and 3C).
利用TRIZOL(Invitrogen)方法提取Raw264.7和L929的RNA,并用SuperScriptTM III Reverse Transcriptase Kit(Invitrogen)分别反转录1μg RNA合成第一链cDNA,用于实时定量PCR检测基因表达水平。实时定量PCR引物详见表1。运用ΔΔct方法计算所测基因相对于GAPDH的相对表达量。利用裂解缓冲液(50mM Tris-HCl(pH 7.4)、150mM NaCl、1mM EDTA、1%Triton X-100、蛋白酶抑制剂complete(Roche)、1mM PMSF、1mM NaF、1mM Na3VO4)于冰上裂解细胞20分钟后,4℃离心(13000rpm,15分钟)去除不溶物质,制备细胞总蛋白抽提液。制备胞浆和胞核蛋白抽提液的方案如下:首先用低渗缓冲液(10mM HEPES(pH 7.6)、1.5mM  MgCl2、10mM KCl、1mM EDTA,蛋白酶抑制剂complete(Roche)、1mM PMSF、1mM NaF、1mM Na3VO4)裂解细胞,用匀浆器Douncer来回处理10-15次,然后于冰上孵育20分钟。4℃低速离心(3000rpm,5分钟)后,将上清取出,再经过高速离心去除不溶物质后即为胞浆蛋白抽提液。低速离心后的沉淀经过高盐缓冲液(20mM HEPES(pH7.6)、500mM NaCl、1.5mM MgCl2、1mM EDTA、蛋白酶抑制剂complete(Roche)、1mM PMSF、1mM NaF、1mM Na3VO4)裂解,高速离心去除不溶物质后,即为细胞核蛋白抽提液。通过Western杂交检测TBK1、IRF3、IκBα、STAT1、JNK、ERK、p38等蛋白的磷酸化。Tubulin、GAPDH、Lamin B1等作为总蛋白、胞浆蛋白或核蛋白抽提液的总蛋白量的参照分子。Using TRIZOL (Invitrogen) and L929 Raw264.7 method of extracting RNA, and anti 1μg RNA are transcribed first strand cDNA synthesis with SuperScript TM III Reverse Transcriptase Kit (Invitrogen ), for real-time quantitative PCR detection of gene expression levels. Real-time quantitative PCR primers are shown in Table 1. The relative expression level of the measured gene relative to GAPDH was calculated using the ΔΔct method. Lysis of cells on ice using lysis buffer (50 mM Tris-HCl (pH 7.4), 150 mM NaCl, 1 mM EDTA, 1% Triton X-100, protease inhibitor complete (Roche), 1 mM PMSF, 1 mM NaF, 1 mM Na3VO4) After a minute, the insoluble matter was removed by centrifugation (13,000 rpm, 15 minutes) at 4 ° C to prepare a total cell protein extract. The protocol for preparing cytosolic and nuclear protein extracts is as follows: first with hypotonic buffer (10 mM HEPES (pH 7.6), 1.5 mM MgCl2, 10 mM KCl, 1 mM EDTA, protease inhibitor complete (Roche), 1 mM PMSF, 1 mM Cells were lysed with NaF, 1 mM Na3VO4) and treated back and forth 10-15 times with a homogenizer Douncer and then incubated on ice for 20 minutes. After centrifugation at a low temperature of 4 ° C (3000 rpm, 5 minutes), the supernatant was taken out, and after centrifugation at high speed to remove insoluble matter, it was a cytosolic protein extract. The pellet after low-speed centrifugation was lysed by high-salt buffer (20 mM HEPES (pH 7.6), 500 mM NaCl, 1.5 mM MgCl2, 1 mM EDTA, protease inhibitor complete (Roche), 1 mM PMSF, 1 mM NaF, 1 mM Na3VO4), and centrifuged at high speed. After removing the insoluble matter, it is a nuclear protein extract. Phosphorylation of proteins such as TBK1, IRF3, IκBα, STAT1, JNK, ERK, and p38 was detected by Western blot. Tubulin, GAPDH, Lamin B1, etc. are reference molecules for total protein amount of total protein, cytosolic protein or nuclear protein extract.
表1、实时定量PCR引物序列Table 1. Real-time quantitative PCR primer sequences
Figure PCTCN2015091111-appb-000001
Figure PCTCN2015091111-appb-000001
结果:实时定量PCR结果显示,在CK2α敲降的Raw264.7细胞中,LPS和polyI:C诱导的IFNβ和Cxcl10的表达明显比对照细胞中高(图1)。相应的,在LPS和polyI:C处理的CK2α敲降的Raw264.7细胞中,IFNβ诱导的靶基因Mx1和Mx2的表达量也大大增高了(图1)。相反,炎症因子TNFα和趋化因子CXCL1的表达与对照组相比没有显著性的差别(图1),表明CK2选择性地抑制I型干扰素及其I型干扰素诱导的基因表达。通过特异性识别蛋白磷酸化的抗体,利用Western杂交检测LPS诱导的TLR4信号通路中信号分子的活化,发现在CK2α敲降的Raw264.7细胞中,参与I型干扰素诱导的关键性信号分子TBK1和IRF3的磷酸化明显增高,但是MAP Kinases JNK、ERK、p38的磷酸化则没有明显的变化(图2)。相反,IκBα的磷酸化反而下降了(图2)。这些数据显示CK2一方面抑制 TBK1和IRF3的活化,另一方面正调节IKKα/β和NFκB的活化,因此CK2在TLR信号通路中针对不同的信号分子,具有不同的调节作用。RESULTS: Real-time quantitative PCR showed that LPS and polyI:C induced IFNβ and Cxcl10 expression were significantly higher in CK2α knockdown Raw264.7 cells than in control cells (Fig. 1). Correspondingly, the expression levels of IFNβ-induced target genes Mx1 and Mx2 were also significantly increased in LPS and polyI:C-treated CK2α knockdown Raw264.7 cells (Fig. 1). In contrast, the expression of the inflammatory factor TNFα and the chemokine CXCL1 was not significantly different from the control group (Fig. 1), indicating that CK2 selectively inhibits type I interferon and its type I interferon-induced gene expression. Western blotting was used to detect the activation of signaling molecules in the TLR4-induced TLR4 signaling pathway by Western blotting. It was found that the key signaling molecule TBK1 involved in type I interferon induction was found in CK2α knockdown Raw264.7 cells. Phosphorylation of IRF3 was significantly increased, but there was no significant change in phosphorylation of MAP Kinases JNK, ERK, and p38 (Fig. 2). In contrast, phosphorylation of IκBα decreased (Fig. 2). These data show that CK2 inhibits on the one hand Activation of TBK1 and IRF3, on the other hand, positively regulates the activation of IKKα/β and NFκB, and thus CK2 has different regulatory roles in different TLR signaling pathways for different signaling molecules.
同样,在CK2敲降的L929细胞中,转化入胞浆的polyI:C、polydA:dT和DMXAA通过激活RIG-I/MDA5或者cGAS/STING信号通路,诱导更多的IFNα、IFNβ、Cxcl10、Mx1和Mx2(图3A)。表明CK2也参与调节胞浆内的RNA和DNA等模式识别受体所诱导的I型干扰素及其I型干扰素诱导的基因表达。同样通过Western杂交,发现转化的polyI:C在CK2敲降的L929细胞内激活更多的TBK1和IRF3,相应地,I型干扰素激活的p-STAT1在CK2敲降的L929细胞也明显比对照细胞中要高(图3B)。在DMXAA刺激的CK2敲降的L929细胞核中,不仅p-TBK1上升,而且IRF3的核转移也明显上调(图3C)。相反,NFκB p65的核转移并没有显著增加(图3C)。进一步证明CK2特异性的抑制TBK1和IRF3的活化,以此调节多种模式识别受体介导的I型干扰素及其I型干扰素诱导基因的表达。Similarly, in CK2 knockdown L929 cells, polyI:C, polydA:dT and DMXAA transformed into cytoplasm induced more IFNα, IFNβ, Cxcl10, Mx1 by activating RIG-I/MDA5 or cGAS/STING signaling pathways. And Mx2 (Figure 3A). It is suggested that CK2 is also involved in the regulation of type I interferon and its type I interferon-induced gene expression induced by pattern recognition receptors such as RNA and DNA in the cytoplasm. Similarly, by Western blotting, it was found that transformed polyI:C activated more TBK1 and IRF3 in CK2 knockdown L929 cells. Accordingly, type I interferon-activated p-STAT1 was significantly more potent than CK2 knockdown L929 cells. The cells are taller (Fig. 3B). In DMXAA-stimulated CK2 knockdown L929 nuclei, not only did p-TBK1 rise, but nuclear transfer of IRF3 was also significantly upregulated (Fig. 3C). In contrast, nuclear transfer of NFκB p65 did not increase significantly (Fig. 3C). It was further demonstrated that CK2-specific inhibition of TBK1 and IRF3 activation regulates the expression of multiple pattern recognition receptor-mediated type I interferons and their type I interferon-inducible genes.
实施例2蛋白激酶CK2参与调控多种病毒感染诱导的I型干扰素表达Example 2 Protein Kinase CK2 is involved in the regulation of type I interferon expression induced by various viral infections
方法:将对照和CK2α敲降的稳转细胞系以106个/ml分别接种到6孔板中,每孔接种2ml。分别感染DNA病毒HSV-1(1×107pfu/ml)、RNA病毒水泡病毒VSV(2.5×105TCID50/ml)和仙台病毒SeV(1×108HA/ml)。感染病毒6小时后,收集细胞用于RNA抽提制备。感染HSV病毒6小时后,换液、继续培养66小时收集细胞上清液用于HSV病毒滴度测定。利用TRIZOL(Invitrogen)方法提取Raw264.7和L929的RNA,并用SuperScriptTM III Reverse Transcriptase Kit(Invitrogen)分别反转录1μg RNA合成第一链cDNA,用于实时定量PCR检测基因表达水平。实时定量PCR引物详见附件表格1。运用ΔΔct方法计算所测基因相对于GAPDH的相对表达量。用经典病毒滴度测定法检测细胞上清液中HSV的病毒滴度,具体方案为:以5×104个/ml分别接种Vero细胞到24孔板中,每孔接种0.5ml,待细胞丰度达到30%时,分别加入不同浓度梯度的细胞上清液(原液、1:101、1:102、1:103、1:104、1:105、1:106、1:107、1:108、1:109、1:1010)和空白对照培养4小时。4小时后,去除上清液,每孔加入0.8%的琼脂-MEM培养液400μl,常温静置1小时待培养基凝固后,继续倒置培养67小时。培养结束后,每孔加入500μl结晶紫溶液,常温静置2小时。抽掉琼脂,计数空斑的数目,计算病毒滴度(pfu/ml,plaque forming unit/ml)。Methods: Control and CK2α knockdown stable cell lines were inoculated into 6-well plates at 10 6 /ml, and 2 ml were inoculated per well. DNA virus HSV-1 (1 × 10 7 pfu/ml), RNA virus vesicular virus VSV (2.5 × 10 5 TCID 50 /ml) and Sendai virus SeV (1 × 10 8 HA/ml) were infected, respectively. Six hours after infection with the virus, cells were harvested for RNA extraction preparation. Six hours after infection with HSV virus, the cells were exchanged for 66 hours and the cell supernatant was collected for HSV virus titer determination. Using TRIZOL (Invitrogen) and L929 Raw264.7 method of extracting RNA, and anti 1μg RNA are transcribed first strand cDNA synthesis with SuperScript TM III Reverse Transcriptase Kit (Invitrogen ), for real-time quantitative PCR detection of gene expression levels. Real-time quantitative PCR primers are shown in Annex Table 1. The relative expression level of the measured gene relative to GAPDH was calculated using the ΔΔct method. The virus titer of HSV in the cell supernatant was detected by classical virus titer assay. The specific protocol was: inoculate Vero cells into 24 well plates at 5×10 4 /ml, and inoculate 0.5 ml per well. When the degree reaches 30%, the cell supernatants with different concentration gradients are added separately (stock solution, 1:10 1 , 1:10 2 , 1:10 3 , 1:10 4 , 1:10 5 , 1:10 6 , 1 ) :10 7 , 1:10 8 , 1:10 9 , 1:10 10 ) and the blank control for 4 hours. After 4 hours, the supernatant was removed, and 400 μl of a 0.8% agar-MEM culture solution was added to each well, and allowed to stand at room temperature for 1 hour. After the medium was solidified, the culture was further inverted for 67 hours. After the completion of the culture, 500 μl of a crystal violet solution was added to each well, and the mixture was allowed to stand at room temperature for 2 hours. The agar was removed, the number of plaques was counted, and the virus titer (pfu/ml, plaque forming unit/ml) was calculated.
结果:实时定量PCR结果显示,在CK2α敲降的Raw264.7细胞中,HSV诱导 的IFNα、IFNβ和Cxcl10的表达明显比对照细胞中高(图4)。相应的,IFNα/β的靶基因Mx1和Mx2的表达也在CK2α敲降的Raw264.7细胞中大大增高了(图4)。随着I型干扰素及其靶基因产物的大量表达,HSV病毒的复制、组装和释放受到显著的抑制,在CK2α敲降的Raw264.7细胞培养液中检测到的HSV病毒滴度比对照组的要低接近10倍(图4)。同样,与对照细胞相比,在CK2敲降的L929细胞中,VSV和SeV诱导的IFNα、IFNβ、Cxcl10、Mx1和Mx2等基因的表达也极其显著地上调(图5和图6)。因此在多种病毒感染状态下,下调CK2的表达均能显著增强不同类别细胞表达I型干扰素的能力。这些结果表明CK2是负调控I型干扰素表达的关键性分子。RESULTS: Real-time quantitative PCR results showed that HSV induction in Raw264.7 cells knocked down by CK2α The expression of IFNα, IFNβ and Cxcl10 was significantly higher than in control cells (Fig. 4). Correspondingly, the expression of the target genes Mx1 and Mx2 of IFNα/β was also greatly increased in CK2α knockdown Raw264.7 cells (Fig. 4). With the large expression of type I interferon and its target gene products, the replication, assembly and release of HSV virus were significantly inhibited, and the HSV virus titer detected in the CK2α knockdown Raw264.7 cell culture medium was higher than that of the control group. It is close to 10 times lower (Figure 4). Similarly, expression of VSV and SeV-induced genes such as IFNα, IFNβ, Cxcl10, Mx1, and Mx2 was also significantly up-regulated in CK2 knockdown L929 cells compared to control cells (Fig. 5 and Fig. 6). Therefore, down-regulation of CK2 expression in a variety of viral infections can significantly enhance the ability of different types of cells to express type I interferons. These results indicate that CK2 is a key molecule for the negative regulation of type I interferon expression.
实施例3 CK2的激酶活性在调控I型干扰素表达中起着关键性的作用Example 3 Kinase activity of CK2 plays a key role in the regulation of type I interferon expression
方法:在lenti-viral载体pCDH上分别克隆编码小鼠CK2α野生型和激酶失活的突变体(K68M)的cDNA,然后将它们转导入L929细胞系,通过puromycin筛选得到对照细胞株,分别表达野生型和突变型CK2α的细胞株。通过Western杂交验证了野生型和突变型CK2α的表达(图7A)。METHODS: The cDNA encoding mouse CK2α wild-type and kinase-inactivated mutant (K68M) was cloned on the lenti-viral vector pCDH, and then transferred into L929 cell line. The control cell lines were screened by puromycin and expressed wild. Type and mutant CK2α cell lines. Expression of wild-type and mutant CK2α was verified by Western blotting (Fig. 7A).
将表达野生型和突变型CK2α的细胞株以106个/ml分别接种到6孔板中,每孔接种2ml。VSV(2.5×105TCID50/ml)感染6小时后,利用TRIZOL(Invitrogen)法抽提制备细胞中的RNA。用SuperScriptTM III Reverse Transcriptase Kit(Invitrogen)分别反转录1μg RNA合成第一链cDNA,用于实时定量PCR检测I型干扰素基因表达水平,运用ΔΔct方法计算所测基因相对于GAPDH的相对表达量。Cell lines expressing wild type and mutant CK2α were inoculated into 6-well plates at 10 6 /ml, and 2 ml was inoculated per well. After 6 hours of infection with VSV (2.5×10 5 TCID 50 /ml), RNA in the cells was extracted by TRIZOL (Invitrogen) method. 1μg RNA first strand cDNA synthesis with SuperScript TM III Reverse Transcriptase Kit (Invitrogen ) reverse transcriptase, respectively, for real-time quantitative PCR detection of gene expression levels of type I interferon, as measured using ΔΔct method to calculate the relative expression of the gene relative to GAPDH .
结果:实验表明,在高表达野生型CK2α的细胞中,IFNα、IFNβ和Cxcl10等基因的表达量比对照细胞低(图7B)。相反,在高表达无激酶活性的CK2α突变体细胞中,这些基因的表达量与对照细胞比没有明显差别(图7B)。这些结果表明上调CK2的表达能降低细胞诱导I型干扰素的能力,而且CK2的激酶活性在调节I型干扰素表达中是必不可少的。Results: The experiments showed that the expression levels of IFNα, IFNβ and Cxcl10 were lower in the cells expressing wild-type CK2α than in the control cells (Fig. 7B). In contrast, in the high expression of kinase-free CK2α mutant cells, the expression levels of these genes were not significantly different from those of the control cells (Fig. 7B). These results indicate that upregulation of CK2 expression reduces the ability of cells to induce type I interferons, and that kinase activity of CK2 is essential for the regulation of type I interferon expression.
实施例4抑制CK2激酶活性促进细胞表达I型干扰素,阻止水泡病毒感染Example 4 Inhibition of CK2 Kinase Activity Promotes Cellular Expression of Type I Interferon and Prevents Vesicular Virus Infection
4.1方法:将小鼠成纤维细胞L929以106个/ml分别接种到6孔板中,每孔接种2ml,待细胞丰度达到70%时,使用不同浓度CK2激酶抑制剂—小分子化合物TBB或DMSO(对照)处理该细胞6小时或12小时后,利用裂解缓冲液(50mM  Tris-HCl(pH 7.4)、150mM NaCl、1mM EDTA,1%Triton X-100,蛋白酶抑制剂Complete(Roche)、1mM PMSF、1mM NaF、1mM Na3VO4)提取并制备细胞的蛋白裂解液,通过Western杂交检测NFκB p65和TBK1的磷酸化。4.1 Method: Mouse fibroblast L929 was inoculated into 6-well plates at 10 6 /ml, 2 ml per well, and when the cell abundance reached 70%, different concentrations of CK2 kinase inhibitor - small molecule compound TBB were used. Or DMSO (control) after treatment of the cells for 6 hours or 12 hours, using lysis buffer (50 mM Tris-HCl (pH 7.4), 150 mM NaCl, 1 mM EDTA, 1% Triton X-100, protease inhibitor Complete (Roche), The protein lysate of the cells was extracted and prepared by 1 mM PMSF, 1 mM NaF, 1 mM Na3VO4), and phosphorylation of NFκB p65 and TBK1 was detected by Western hybridization.
结果:图8A的结果表明20μM TBB处理L929细胞6小时后,CK2的激酶活性被抑制,它所介导的p65磷酸化明显降低。但总体而言,100μM TBB处理12小时能更有效地抑制CK2的活性,而且能最有效地激活诱导I型干扰素表达的关键性激酶TBK1(图8A)。Results: The results in Figure 8A indicate that after 20 hours of treatment of L929 cells with 20 μM TBB, the kinase activity of CK2 is inhibited, and its mediated phosphorylation of p65 is significantly reduced. Overall, however, treatment with 100 μM TBB for 12 hours was more effective at inhibiting CK2 activity and was the most potent activation of the key kinase TBK1 that induces type I interferon expression (Fig. 8A).
4.2方法:同法使用TBB(100μM)或DMSO(对照)处理L929细胞12小时后,利用TRIZOL(Invitrogen)法抽提制备细胞中的RNA。用SuperScriptTM III Reverse Transcriptase Kit(Invitrogen)分别反转录1μg RNA合成第一链cDNA,用于实时定量PCR检测I型干扰素基因表达水平,运用ΔΔct方法计算所测基因相对于GAPDH的相对表达量。4.2 Method: L929 cells were treated with TBB (100 μM) or DMSO (control) for 12 hours in the same manner, and RNA in the cells was extracted by TRIZOL (Invitrogen) method. 1μg RNA first strand cDNA synthesis with SuperScript TM III Reverse Transcriptase Kit (Invitrogen ) reverse transcriptase, respectively, for real-time quantitative PCR detection of gene expression levels of type I interferon, as measured using ΔΔct method to calculate the relative expression of the gene relative to GAPDH .
结果:发现TBB处理促进细胞合成IFNα、IFNβ、Cxcl10、Mx1和Mx2等基因的表达(图8B),因此可能建立了一个抗病毒感染的状态。果然,用0、20、50、100μM的TBB分别处理L929细胞12小时后,再接种VSV病毒(2.5×105TCID50/ml,表达GFP,因此可以利用荧光显微镜检测被感染的细胞);24小时后,利用荧光显微镜检测VSV感染的效率。发现TBB预处理的细胞呈现出对VSV感染的抗性,而且随着TBB浓度的增加,抗VSV感染的能力也变强。需要特别指出的是,100μM TBB处理后的L929细胞几乎完全不被VSV感染,具备了抵御病毒感染的能力(图9)。Results: It was found that TBB treatment promoted the synthesis of genes such as IFNα, IFNβ, Cxcl10, Mx1 and Mx2 (Fig. 8B), and thus it was possible to establish an anti-viral infection state. Sure enough, L929 cells were treated with 0, 20, 50, 100 μM TBB for 12 hours, then VSV virus (2.5 × 10 5 TCID 50 /ml, expressing GFP, so fluorescent cells can be used to detect infected cells); After an hour, the efficiency of VSV infection was examined using a fluorescence microscope. Cells pretreated with TBB were found to exhibit resistance to VSV infection, and as the concentration of TBB increased, the ability to resist VSV infection also became stronger. In particular, L929 cells treated with 100 μM TBB were almost completely devoid of VSV infection and were resistant to viral infection (Figure 9).
4.3在人胚胎肾细胞HEK293T中,验证了抑制CK2的激酶活性对细胞表达I型干扰素的影响以及在阻止水泡病毒感染中的作用。4.3 In human embryonic kidney cell HEK293T, the effect of inhibition of CK2 kinase activity on cell expression of type I interferon and its role in preventing vesicular virus infection was examined.
方法:将人胚胎肾细胞HEK293T以106个/ml分别接种到6孔板中,每孔接种2ml,待细胞丰度达到70%时,使用TBB或DMSO(对照)处理细胞。同法提取细胞的蛋白裂解液用于Western检测,提取RNA用于I型干扰素相关基因表达水平的检测,荧光显微镜检测VSV感染的效率。METHODS: Human embryonic kidney cells HEK293T were inoculated into 6-well plates at 10 6 cells/ml, and 2 ml were inoculated per well. When the cell abundance reached 70%, cells were treated with TBB or DMSO (control). The protein lysate of the same method was used for Western detection, RNA was extracted for the detection of type I interferon-related gene expression, and the efficiency of VSV infection was detected by fluorescence microscopy.
结果:实验发现,在人胚胎肾细胞HEK293T中,TBB处理12或24小时后,TBK1也被明显地激活(图10A),并促进了IFNα、IFNβ、Mx1和Mx2等基因的表达(图10B)。更重要的是,TBB处理后的HEK 293细胞抵抗VSV感染的能力也极大地增强(图10C)。这些结果说明TBB在小鼠和人细胞中均能诱导I型干扰素的表达,建立抗病毒感染的防御状态,有效地抵抗VSV病毒的感染。 RESULTS: It was found that in human embryonic kidney cell HEK293T, TBK1 was also significantly activated after 12 or 24 hours of TBB treatment (Fig. 10A), and promoted the expression of genes such as IFNα, IFNβ, Mx1 and Mx2 (Fig. 10B). . More importantly, the ability of TKB-treated HEK 293 cells to resist VSV infection was also greatly enhanced (Fig. 10C). These results indicate that TBB can induce the expression of type I interferon in both mouse and human cells, establish a defense state against viral infection, and effectively resist the infection of VSV virus.
实施例5 CK2抑制剂TBB阻止HCV感染Example 5 CK2 inhibitor TBB prevents HCV infection
HCV感染导致的慢性肝炎在中国和世界范围内都呈增加的趋势。在人肝癌细胞Hu7.5中,HCV病毒可以非常有效地感染和复制,但不能诱导I型干扰素的表达。因此,外源给予干扰素治疗对一部分HCV慢性感染病人有较好的效果,但干扰素价格昂贵,且副作用明显。所以,探究能否利用TBB来诱导宿主自体产生I型干扰素,以达到治疗HCV感染的效果。Chronic hepatitis caused by HCV infection is increasing in China and around the world. In human hepatoma cell Hu7.5, HCV virus can infect and replicate very efficiently, but does not induce expression of type I interferon. Therefore, exogenous administration of interferon therapy has a good effect on some patients with chronic HCV infection, but interferon is expensive and has obvious side effects. Therefore, it is explored whether TBB can be used to induce the host to produce type I interferon autologously in order to achieve the effect of treating HCV infection.
方法:将Hu7.5细胞以5×104个/ml分别接种到6孔板中,每孔接种2ml,使用的培养基是DMEM完全培养基。待细胞丰度达到30%时,100μM TBB或DMSO(对照)处理24小时后,MOI(multiplicity of infection)=0.01或者MOI=0.1的HCV感染8小时后,换液、继续培养64小时。收集细胞分别用于RNA分析和HCV感染的效率,收集细胞上清液用于HCV病毒滴度测定。用经典病毒滴度测定法检测细胞上清液中HCV的病毒滴度,具体方案为:以5×104个/ml分别接种Hu7.5细胞到96孔板中,每孔接种200μl,待细胞丰度达到30%时,分别加入不同浓度梯度的细胞上清液(原液、1:101、1:102、1:103、1:104、1:105、1:106、1:107、1:108、1:109、1:1010),继续培养72小时,收集细胞,多聚甲醛固定后免疫荧光化学法标记HCV特异性抗体,荧光显微镜检测已被HCV感染并带有荧光标记的Hu7.5细胞个数,计算病毒滴度(ffu/ml,fluorescence forming foci/ml)。Methods: Hu7.5 cells were inoculated into 6-well plates at 5×10 4 cells/ml, and each well was inoculated with 2 ml. The medium used was DMEM complete medium. When the cell abundance reached 30%, after treatment with 100 μM TBB or DMSO (control) for 24 hours, HCV infection with MOI (multiplicity of infection) = 0.01 or MOI = 0.1 was carried out for 8 hours, and the medium was changed and the culture was continued for 64 hours. Cells were harvested for efficiency of RNA analysis and HCV infection, respectively, and cell supernatants were collected for HCV virus titer determination. The virus titer of HCV in the cell supernatant was detected by classical virus titer assay. The specific protocol was: inoculate Hu7.5 cells into 96-well plates at 5×10 4 /ml, and inoculate 200 μl per well. When the abundance reaches 30%, the cell supernatants with different concentration gradients are added separately (stock solution, 1:10 1 , 1:10 2 , 1:10 3 , 1:10 4 , 1:10 5 , 1:10 6 , 7 1:10, 1: 108, 1: 109, 1: 1010), cultured for 72 hours, the cells were collected, labeled antibodies specific for HCV paraformaldehyde after immunofluorescence, fluorescence microscopy has been HCV The number of Hu7.5 cells infected with fluorescently labeled cells was calculated and the virus titer (ffu/ml, fluorescence forming foci/ml) was calculated.
结果:同法提取分析细胞中的RNA与I型干扰素相关基因的表达水平,定量PCR结果表明,在TBB预处理的细胞中,IFNα、IFNβ、Mx1和Mx2等I型干扰素相关基因的表达较高;同时,TBB预处理也提升了细胞中炎症因子IL-6的表达(图11A)。相反,Hcv基因的表达量则非常少(图11A)。细胞经4%多聚甲醛固定30分钟,PBS磷酸缓冲液洗涤,应用免疫荧光化学法标记HCV特异性抗体,荧光显微镜检测HCV感染效率。发现未受处理的细胞大量感染HCV,但TBB处理后仅仅有极少的细胞被HCV感染(图11B)。HCV的病毒滴度检测结果显示,TBB预处理后,细胞上清液中HCV病毒的滴度明显降低(图11C)。以上研究结果综合表明TBB预处理能促进肝脏细胞Hu7.5产生I型干扰素,阻止HCV的感染和复制。RESULTS: The expression levels of RNA and type I interferon-related genes in cells were extracted by the same method. The results of quantitative PCR showed that the expression of type I interferon-related genes such as IFNα, IFNβ, Mx1 and Mx2 in TBB pretreated cells. At the same time, TBB pretreatment also increased the expression of the inflammatory factor IL-6 in the cells (Fig. 11A). In contrast, the expression level of the Hcv gene was very small (Fig. 11A). The cells were fixed in 4% paraformaldehyde for 30 minutes, washed with PBS phosphate buffer, labeled with HCV-specific antibodies by immunofluorescence, and the efficiency of HCV infection was detected by fluorescence microscopy. Untreated cells were found to be heavily infected with HCV, but only very few cells were infected with HCV after TBB treatment (Fig. 11B). The virus titer test results of HCV showed that the titer of HCV virus in the cell supernatant was significantly reduced after TBB pretreatment (Fig. 11C). The above results show that TBB pretreatment can promote liver cell Hu7.5 to produce type I interferon and prevent HCV infection and replication.
实施例6 TBB抑制HCV复制、清除HCV感染Example 6 TBB inhibits HCV replication and clears HCV infection
方法:将Hu7.5细胞以5×104个/ml分别接种到6孔板中,每孔接种2ml,待细胞丰度达到30%时,首先用MOI(multiplicity of infection)=0.01或者MOI=0.1 的HCV感染细胞,24小时后,HCV已经进入细胞并开始复制,再使用100μM TBB或DMSO(对照)处理感染后的Hu7.5细胞。48小时后,同法收获细胞并制备RNA,通过定量PCR检测细胞和病毒基因的表达量。METHODS: Hu7.5 cells were inoculated into 6-well plates at 5×10 4 cells/ml, and each well was inoculated with 2 ml. When the cell abundance reached 30%, firstly, MOI (multiplicity of infection)=0.01 or MOI= After 0.1% of HCV-infected cells, 24 hours later, HCV had entered the cells and began to replicate, and the infected Hu7.5 cells were treated with 100 μM TBB or DMSO (control). After 48 hours, cells were harvested by the same method and RNA was prepared, and the expression levels of the cells and viral genes were detected by quantitative PCR.
结果:HCV感染后,Hu7.5细胞并不能诱导I型干扰素的表达。但加入TBB后,细胞开始大量表达I型干扰素、I型干扰素诱导的基因MX1和MX2,以及一些促炎症因子,像IL-6和TNFα(图12A)。随着I型干扰素及其靶基因产物的大量表达,HCV病毒的复制、组装和释放受到显著的抑制,在TBB处理的细胞培养液中检测到的HCV病毒滴度比未处理的要低10-100倍(图12B)。这些结果表明TBB可以用来治疗慢性HCV感染的病人。RESULTS: Hu7.5 cells did not induce type I interferon expression after HCV infection. However, after the addition of TBB, the cells began to express large amounts of type I interferons, type I interferon-inducible genes MX1 and MX2, and some pro-inflammatory factors such as IL-6 and TNFα (Fig. 12A). With the large expression of type I interferons and their target gene products, the replication, assembly and release of HCV virus were significantly inhibited, and the HCV virus titer detected in TBC-treated cell culture was lower than that of untreated. -100 times (Fig. 12B). These results indicate that TBB can be used to treat patients with chronic HCV infection.
实施例7 TBB调节肿瘤细胞中I型干扰素的表达Example 7 TBB regulates the expression of type I interferon in tumor cells
方法:人原髓细胞白血病细胞(Human promyelocytic leukemia cells)HL-60(购自ATCC)和人肾癌细胞OS-RC-2在RPMI-1640培养基中培养,并补加10%的胎牛血清、2mM谷氨酰胺和抗生素。将HL-60或OS-RC-2细胞以5×104个/ml分别接种到6孔板中,每孔接种2ml,待细胞丰度达到30%时,加100μM TBB或DMSO(对照)处理。12小时后,收集细胞,用Trizol(Sigma)制备RNA。用SuperScriptTM III Reverse Transcriptase Kit(Invitrogen)分别反转录1μg RNA合成第一链cDNA,用于实时定量PCR检测I型干扰素基因表达水平,运用ΔΔct方法计算所测基因相对于GAPDH的相对表达量。METHODS: Human promyelocytic leukemia cells HL-60 (purchased from ATCC) and human renal cancer cell line OS-RC-2 were cultured in RPMI-1640 medium supplemented with 10% fetal bovine serum. 2 mM glutamine and antibiotics. HL-60 or OS-RC-2 cells were inoculated into 6-well plates at 5×10 4 cells/ml, and each well was inoculated with 2 ml. When the cell abundance reached 30%, 100 μM TBB or DMSO (control) was added. . After 12 hours, cells were harvested and RNA was prepared using Trizol (Sigma). 1μg RNA first strand cDNA synthesis with SuperScript TM III Reverse Transcriptase Kit (Invitrogen ) reverse transcriptase, respectively, for real-time quantitative PCR detection of gene expression levels of type I interferon, as measured using ΔΔct method to calculate the relative expression of the gene relative to GAPDH .
结果:定量PCR结果表明,在TBB预处理的人原髓细胞白血病细胞HL-60和人肾癌细胞OS-RC-2细胞中,IFNα、IFNβ、Cxcl10、Mx1和Mx2等I型干扰素相关基因的表达比未处理的对照细胞明显增高(图13和图14),表明TBB也能在肿瘤细胞中诱导I型干扰素的表达。RESULTS: Quantitative PCR results showed that type I interferon-related genes such as IFNα, IFNβ, Cxcl10, Mx1 and Mx2 were expressed in human promyelocytic leukemia cell line HL-60 and human kidney cancer cell line OS-RC-2 pretreated with TBB. The expression was significantly higher than that of untreated control cells (Figures 13 and 14), indicating that TBB also induced type I interferon expression in tumor cells.
实施例8 CK2抑制剂TBB抑制EV71感染Example 8 CK2 inhibitor TBB inhibits EV71 infection
方法:将Vero细胞以3×104个/ml分别接种到24孔板中,每孔接种0.5ml,待Vero细胞丰度达到70%时,按照TBB预防组与TBB治疗组分别处理Vero细胞。TBB预防组:20、50、和100μM TBB或DMSO(对照)分别处理细胞2小时后,分别感染肠道病毒EV712*102TCID50/ml 4小时后,换液、继续用添加20、50和100μM TBB或DMSO(对照)的培养基分别培养24小时。换液,用未添加TBB或DMSO的培养基继续培养48小时,收集细胞上清液用于EV71病毒滴度测定。TBB 治疗组:感染肠道病毒EV712*102TCID50/ml 4小时后,换液、用添加20、50和100μM TBB或DMSO(对照)的培养基分别培养72小时,收集细胞上清液用于EV71病毒滴度测定。用经典病毒滴度测定法TCID50(半数组织细胞感染量)检测细胞上清液中EV71的病毒滴度,具体方案为:以3×104个/ml分别接种Vero细胞到96孔板中,每孔接种0.1ml,待细胞丰度达到70%时,分别加入不同浓度梯度的细胞上清液(原液、1:101、1:102、1:103、1:104、1:105、1:106、1:107、1:108、1:109、1:1010)和空白对照培养4小时。4小时后,去除上清液,换液、继续培养68小时。培养结束后,光学显微镜观察细胞受感染程度,计数超过半数感染的孔板的数目,计算病毒滴度(TCID50/ml,tissue culture infective dose/ml)。METHODS: Vero cells were inoculated into a 24-well plate at 3×10 4 cells/ml, and 0.5 ml per well was inoculated. When the abundance of Vero cells reached 70%, Vero cells were treated according to the TBB prevention group and the TBB treatment group, respectively. TBB prevention group: 20, 50, and 100 μM TBB or DMSO (control) were treated with cells for 2 hours, respectively, and infected with enterovirus EV712*10 2 TCID 50 /ml for 4 hours, then change the solution and continue to add 20, 50 and The medium of 100 μM TBB or DMSO (control) was cultured for 24 hours. The medium was changed, culture was continued for 48 hours with medium without addition of TBB or DMSO, and the cell supernatant was collected for EV71 virus titer determination. TBB treatment group: Infected enterovirus EV712*10 2 TCID 50 /ml 4 hours later, change medium, culture with medium supplemented with 20, 50 and 100 μM TBB or DMSO (control) for 72 hours, collect cell supernatant The EV71 virus titer was determined. The viral titer of EV71 in the cell supernatant was determined by the classical virus titer assay TCID 50 (half the number of tissue cell infections). The specific protocol was: inoculate Vero cells into 96-well plates at 3×10 4 /ml, respectively. Each well was inoculated with 0.1 ml. When the cell abundance reached 70%, cell supernatants with different concentration gradients were added (stock solution, 1:10 1 , 1:10 2 , 1:10 3 , 1:10 4 , 1: 10 5 , 1:10 6 , 1:10 7 , 1:10 8 , 1:10 9 , 1:10 10 ) and the blank control for 4 hours. After 4 hours, the supernatant was removed, the solution was changed, and incubation was continued for 68 hours. After the completion of the culture, the degree of infection of the cells was observed by an optical microscope, and the number of well plates exceeding half of the infection was counted, and the virus titer (TCID 50 /ml, tissue culture infective dose/ml) was calculated.
结果:不同剂量TBB预防组处理Vero细胞,EV71感染后,TBB预防组处理的细胞培养液中检测到的EV71病毒滴度低于未处理组,并呈现一定的浓度相关性。预防组中,使用TBB的浓度越高,细胞培养液中检测到的EV71病毒滴度越低(图15A)。先感染EV71,再使用TBB的治疗组细胞培养液中检测到的EV71病毒滴度比未处理组要低10-100倍(图15B),高浓度使用TBB(100μM)能够显著地抑制EV71病毒的复制、组装和释放,最终检测到的EV71病毒滴度降低达到百倍(图15B)。RESULTS: Vero cells were treated in different doses of TBB prevention group. After EV71 infection, the EV71 virus titer detected in the cell culture medium treated by TBB prevention group was lower than that in the untreated group, and showed a certain concentration correlation. In the prevention group, the higher the concentration of TBB used, the lower the EV71 virus titer detected in the cell culture solution (Fig. 15A). The EV71 virus titer detected in the cell culture medium of the treatment group infected with EV71 was 10-100 times lower than that of the untreated group (Fig. 15B). The high concentration of TBC (100 μM) can significantly inhibit the EV71 virus. Replication, assembly and release resulted in a 100-fold reduction in the final detected EV71 virus titer (Fig. 15B).
实施例9动物感染模型中TBB抑制疱疹病毒HSV的感染Example 9 Inhibition of herpesvirus HSV infection by TBB in an animal infection model
9.1 TBB激活小鼠体内诱导I型干扰素表达的关键性激酶TBK19.1 TBB activates the key kinase TBK1 that induces type I interferon expression in mice
方法:6-8周龄B6小鼠,雌雄各半,分别依照小鼠体重腹腔注射2.5、10、25mg/kg TBB或葵花籽油(对照组),24小时后按照动物福利法处死小鼠,取肝脏与脾脏测定组织样本中与I型干扰素合成相关的细胞因子TBK1的激活程度。称取单位重量的组织样本,加入裂解缓冲液(50mM Tris-HCl(pH 7.4)、150mM NaCl、1mM EDTA,1%Triton X-100,蛋白酶抑制剂Complete(Roche)、1mM PMSF、1mM NaF、1mM Na3VO4)匀浆,提取并制备组织样本的蛋白裂解液,通过Western杂交检测TBK1的磷酸化。METHODS: B6 mice of 6-8 weeks old, male and female, were intraperitoneally injected with 2.5, 10, 25 mg/kg TBB or sunflower oil (control group) according to the body weight of the mice. After 24 hours, the mice were sacrificed according to the animal welfare method. Liver and spleen were taken to determine the degree of activation of the cytokine TBK1 associated with type I interferon synthesis in tissue samples. Weigh a tissue sample per unit weight and add lysis buffer (50 mM Tris-HCl (pH 7.4), 150 mM NaCl, 1 mM EDTA, 1% Triton X-100, Protease Inhibitor Complete (Roche), 1 mM PMSF, 1 mM NaF, 1 mM. Na3VO4) homogenate, extract and prepare protein lysate of tissue samples, and detect the phosphorylation of TBK1 by Western blot.
结果:图16A的结果表明,腹腔注射10mg/kg TBB 24小时可以有效地激活肝脏中诱导I型干扰素表达的关键性激酶TBK1,腹腔注射2.5或者10mg/kg TBB24小时均可以有效地激活脾脏中诱导I型干扰素表达的关键性激酶TBK1。同时,根据Western检测的结果,我们采用10mg/kg的TBB作为后续小鼠动物感染模型腹腔注射实施例的剂量。 RESULTS: The results in Figure 16A indicate that intraperitoneal injection of 10 mg/kg TBB for 24 hours can effectively activate the key kinase TBK1 that induces type I interferon expression in the liver. Both intraperitoneal injection of 2.5 or 10 mg/kg TBB for 24 hours can effectively activate the spleen. The key kinase TBK1 that induces type I interferon expression. At the same time, according to the results of the Western test, we used 10 mg/kg of TBB as the dose of the intraperitoneal injection example of the subsequent mouse animal infection model.
9.2 TBB抑制小鼠体内HSV的感染9.2 TBB inhibits HSV infection in mice
方法:6-8周龄B6小鼠,雌雄各半,分别依照小鼠体重腹腔注射10mg/kg TBB或葵花籽油(对照组),6小时后分别依照小鼠体重尾静脉注射5*107pfu/g HSV。注射HSV 12小时后同法给予第二次TBB,注射HSV 24小时后按照动物福利法处死小鼠,取血液与脾脏测定组织样本中HSV的病毒滴度。用经典病毒滴度测定法检测小鼠组织样本中HSV的病毒滴度,具体方案为:称取1g组织样本,加入1ml PBS,匀浆得到组织匀浆液。以5×104个/ml分别接种Vero细胞到24孔板中,每孔接种0.5ml,待细胞丰度达到30%时,分别加入不同浓度梯度的组织样本匀浆液(原液、1:101、1:102、1:103、1:104、1:105、1:106、1:107、1:108)和空白对照(PBS)培养4小时。4小时后,去除匀浆液,每孔加入0.8%的琼脂-MEM培养液400μl,常温静置1小时待培养基凝固后,继续倒置培养67小时。培养结束后,每孔加入500μl结晶紫溶液,常温静置2小时。抽掉琼脂,计数空斑的数目,计算病毒滴度(pfu/g,plaque forming unit/g)。METHODS: B6 mice of 6-8 weeks old, male and female, were intraperitoneally injected with 10 mg/kg TBB or sunflower oil (control group) according to the body weight of the mice. After 6 hours, the mice were injected with 5*10 7 according to the tail weight of the mice. Pfu/g HSV. Twelve hours after the injection of HSV, the second TBB was given by the same method. After 24 hours of HSV injection, the mice were sacrificed according to the animal welfare method, and the blood and spleen were taken to determine the virus titer of HSV in the tissue samples. The virus titer of HSV in mouse tissue samples was detected by classical virus titer assay. The specific protocol was as follows: 1 g tissue sample was weighed, 1 ml PBS was added, and homogenate was obtained to obtain a tissue homogenate. Tissue sample homogenate (stock solution, 1 to 5 × 10 4 cells / ml Vero cells were seeded into 24-well plates, each well was inoculated 0.5ml, abundance of cells to be 30%, were added different concentration gradient: 101 , 1:10 2 , 1:10 3 , 1:10 4 , 1:10 5 , 1:10 6 , 1:10 7 , 1:10 8 ) and the blank control (PBS) for 4 hours. After 4 hours, the homogenate was removed, and 400 μl of a 0.8% agar-MEM culture solution was added to each well, and allowed to stand at room temperature for 1 hour. After the medium was solidified, the culture was further inverted for 67 hours. After the completion of the culture, 500 μl of a crystal violet solution was added to each well, and the mixture was allowed to stand at room temperature for 2 hours. The agar was removed, the number of plaques was counted, and the virus titer (pfu/g, plaque forming unit/g) was calculated.
结果:由于腹腔注射10mg/kg TBB可以有效激活小鼠组织中诱导I型干扰素表达的关键性激酶TBK1(图16A),从而使HSV病毒的复制、组装和释放受到显著的抑制。HSV感染24小时后,TBB处理组小鼠血液、脾脏中检测到的HSV病毒滴度显著低于对照组(图16B)。RESULTS: The intraperitoneal injection of 10 mg/kg TBB was effective in activating the key kinase TBK1 that induces type I interferon expression in mouse tissues (Fig. 16A), thereby significantly inhibiting the replication, assembly and release of HSV virus. After 24 hours of HSV infection, the HSV virus titer detected in the blood and spleen of the TBB-treated group was significantly lower than that of the control group (Fig. 16B).
在本发明提及的所有文献都在本申请中引用作为参考,就如同每一篇文献被单独引用作为参考那样。此外应理解,在阅读了本发明的上述讲授内容之后,本领域技术人员可以对本发明作各种改动或修改,这些等价形式同样落于本申请所附权利要求书所限定的范围。 All documents mentioned in the present application are hereby incorporated by reference in their entirety in their entireties in the the the the the the the the In addition, it should be understood that various modifications and changes may be made by those skilled in the art in the form of the appended claims.

Claims (10)

  1. 酪蛋白激酶2(Casein kinase 2,CK2)抑制剂的用途,其特征在于,(i)用于制备促进I型干扰素(I-IFN)和/或I型干扰素诱导基因或其蛋白表达的药物组合物;和/或(ii)用于制备治疗和/或预防I型干扰素缺乏相关疾病的药物组合物。Use of a casein kinase 2 (CK2) inhibitor, characterized in that (i) is used for the preparation of a type I interferon (I-IFN) and/or a type I interferon-inducible gene or protein thereof. a pharmaceutical composition; and/or (ii) a pharmaceutical composition for the preparation of a disease associated with the treatment and/or prevention of type I interferon deficiency.
  2. 如权利要求1所述的用途,其特征在于,所述的抑制剂包括CK2的反义核酸、抑制性microRNA、抗体或小分子化合物。The use according to claim 1, wherein the inhibitor comprises an antisense nucleic acid, an inhibitory microRNA, an antibody or a small molecule compound of CK2.
  3. 如权利要求1所述的用途,其特征在于,所述的I型干扰素缺乏相关疾病包括:病毒感染性疾病、恶性肿瘤、多发性硬化(Multiple Sclerosis)。The use according to claim 1, wherein the type I interferon deficiency-related diseases include: viral infectious diseases, malignant tumors, multiple sclerosis.
  4. 如权利要求3所述的用途,其特征在于,所述的病毒感染性疾病包括感染了以下的病毒的疾病:单纯疱疹病毒(HSV)、水泡病毒(VSV)、卡波希氏瘤疱疹病毒(KSHV)、呼吸道合胞病毒(RSV)、手足口病病毒(肠道病毒EV71和CA16)、乙型肝炎病毒(HBV)、丙型肝炎病毒(HCV)、或人类获得性免疫缺陷病毒/艾滋病病毒(HIV);和/或The use according to claim 3, wherein the viral infectious disease comprises a disease infected with the following viruses: herpes simplex virus (HSV), vesicular virus (VSV), Kaposi's tumor virus ( KSHV), respiratory syncytial virus (RSV), hand, foot and mouth disease virus (enteric virus EV71 and CA16), hepatitis B virus (HBV), hepatitis C virus (HCV), or human acquired immunodeficiency virus/HIV (HIV); and/or
    所述的恶性肿瘤包括以下肿瘤:多毛细胞白血病(hairy cell leukaemia)、慢性髓性白血病(CML)、淋巴瘤(lymphoma)、骨髓瘤(myeloma)、黑色素瘤(melanoma)、肾细胞癌(renal cell carcinoma)、膀胱癌(bladder cell carcinoma)、或卡波希氏肉瘤(Kaposi’s sarcoma)。The malignant tumor includes the following tumors: hairy cell leukaemia, chronic myelogenous leukemia (CML), lymphoma, myeloma, melanoma, renal cell carcinoma (renal cell) Carcinoma, bladder cell carcinoma, or Kaposi's sarcoma.
  5. 如权利要求2所述的用途,其特征在于,所述的抑制剂为抑制I型干扰素表达和/或活性的小分子化合物,较佳地,包括四溴苯三唑(TBB)、CX-4945(Silmitasertib)、DMAT(2-二甲氨基-4,5,6,7-四溴-1H-苯并咪唑)、山奈酚、酪氨酸磷酸化抑制剂AG114(Tyrphostin AG114)、四溴肉桂酸、3-[[5-(4-甲苯基)噻吩[2,3-d]嘧啶-4-yl]硫基]丙酸(TTP 22)、CX-5011、鞣花酸(Ellagic Acid)、3-甲基-1,6,8-三氢蒽醌(emodin)、4',5,7-三羟基黄酮(apigenin)。The use according to claim 2, wherein the inhibitor is a small molecule compound that inhibits type I interferon expression and/or activity, preferably including tetrabromobenzotriazole (TBB), CX-. 4945 (Silmitasertib), DMAT (2-dimethylamino-4,5,6,7-tetrabromo-1H-benzimidazole), kaempferol, tyrosine phosphorylation inhibitor AG114 (Tyrphostin AG114), tetrabromocinnamyl Acid, 3-[[5-(4-methylphenyl)thiophene [2,3-d]pyrimidin-4-yl]thio]propionic acid (TTP 22), CX-5011, Ellagic Acid, 3-methyl-1,6,8-trihydropurine (emodin), 4',5,7-trihydroxyflavone (apigenin).
  6. 如权利要求1所述的用途,其特征在于,所述的I型干扰素诱导基因或其蛋白包括CXCL10/IP-10、STAT1、MX1、MX2、Rsad2/Viperine、磷酸激酶TBK1、干扰素调节因子3(IRF3)。The use according to claim 1, wherein the type I interferon-inducing gene or protein thereof comprises CXCL10/IP-10, STAT1, MX1, MX2, Rsad2/Viperine, phosphokinase TBK1, interferon regulatory factor 3 (IRF3).
  7. 一种体外非治疗性的促进细胞表达I型干扰素和/或I型干扰素诱导基因或其蛋白和/或体外非治疗性的抑制I型干扰素缺乏相关病毒细胞和/或肿瘤细胞生长的方法,其特征在于,包括步骤:在CK2抑制剂的存在下,培养细胞,从而促进细胞表达I型干扰素和/或I型干扰素诱导基因或其蛋白和/或抑制I型干扰素缺乏相关病毒细胞和/或肿瘤细胞生长。 An in vitro non-therapeutic promoting cell expressing a type I interferon and/or a type I interferon-inducible gene or protein thereof and/or in vitro non-therapeutic inhibition of type I interferon deficiency-associated viral cells and/or tumor cell growth The method comprises the steps of: culturing a cell in the presence of a CK2 inhibitor, thereby promoting expression of a type I interferon and/or a type I interferon-inducing gene or a protein thereof and/or inhibiting a type I interferon deficiency Viral cells and/or tumor cells grow.
  8. 一种筛选能够抑制CK2的化合物的方法,其特征在于,包括步骤:A method for screening a compound capable of inhibiting CK2, comprising the steps of:
    (a1)在试验组中,向细胞培养体系加入待测化合物,并测定I型干扰素的表达量和/或活性;在对照组中,向细胞培养体系不加入待测化合物,并测定I型干扰素的表达量和/或活性;(a1) In the test group, a test compound was added to the cell culture system, and the expression amount and/or activity of the type I interferon was measured; in the control group, the test compound was not added to the cell culture system, and the type I was determined. The amount and/or activity of interferon expression;
    其中,若试验组中I型干扰素的表达量和/或活性显著高于对照组,则说明所述的候选化合物为能够抑制CK2的化合物;和/或Wherein, if the expression level and/or activity of the type I interferon in the test group is significantly higher than that of the control group, the candidate compound is a compound capable of inhibiting CK2; and/or
    (a2)在试验组中,向病毒感染的细胞培养体系中加入待测化合物,并测定细胞培养体系中病毒的滴度;在对照组中,向病毒感染的细胞培养体系中加入待测化合物,并测定细胞培养体系中的病毒滴度;(a2) In the test group, the test compound is added to the virus-infected cell culture system, and the titer of the virus in the cell culture system is determined; in the control group, the test compound is added to the virus-infected cell culture system, And measuring the virus titer in the cell culture system;
    其中,若试验组病毒的滴度显著低于对照组,则说明所述的候选化合物为能够抑制CK2的化合物。Wherein, if the titer of the test group virus is significantly lower than that of the control group, the candidate compound is a compound capable of inhibiting CK2.
  9. 如权利要求8所述的方法,其特征在于,还包括步骤:The method of claim 8 further comprising the step of:
    (b)对(a)中的化合物进一步测试其对酪蛋白激酶2基因或其蛋白的促进作用。(b) Further testing the compound of (a) for its promotion of the casein kinase 2 gene or its protein.
  10. 一种制备I型干扰素的方法,其特征在于,包括步骤:A method for preparing a type I interferon, comprising the steps of:
    (i)在CK2抑制剂的存在下培养细胞;(i) culturing the cells in the presence of a CK2 inhibitor;
    (ii)收集、分离并纯化细胞培养液中的I型干扰素。 (ii) collecting, isolating and purifying type I interferons in the cell culture medium.
PCT/CN2015/091111 2014-09-29 2015-09-29 Use of inhibiting activity of casein kinase 2 for enhancing expression of type i interferon WO2016050203A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN201410514107 2014-09-29
CN201410514107.8 2014-09-29

Publications (1)

Publication Number Publication Date
WO2016050203A1 true WO2016050203A1 (en) 2016-04-07

Family

ID=55595471

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2015/091111 WO2016050203A1 (en) 2014-09-29 2015-09-29 Use of inhibiting activity of casein kinase 2 for enhancing expression of type i interferon

Country Status (2)

Country Link
CN (1) CN105457029A (en)
WO (1) WO2016050203A1 (en)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2021236578A1 (en) * 2020-05-18 2021-11-25 Salk Institute For Biological Studies Chromen-4-one derivatives, such as e.g. flavones, for use as ck2 inhibitors for the treatment of neuroinflammation
EP4126845A4 (en) * 2020-03-30 2024-05-01 Senhwa Biosciences Inc Antiviral compounds and method for treating rna viral infection, particularly covid-19

Families Citing this family (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN107714711A (en) * 2017-09-15 2018-02-23 中国科学院成都生物研究所 The new application of the O glucopyranosides of 6 hydroxyl, 3 O methyl Kaempferol 6
CN109646678A (en) * 2017-10-12 2019-04-19 中国科学院上海生命科学研究院 SUN2 albumen, its pharmaceutical applications and drug
CN108379584B (en) * 2018-04-12 2020-08-11 上海交复生物医药科技有限公司 RSAD2 influences tumor cell temozolomide resistance through wnt pathway
CN108498798A (en) * 2018-04-17 2018-09-07 上海市第六人民医院 It is a kind of prevention skeletal muscle atrophy medicine target spot and its application
CN114948955A (en) * 2022-06-06 2022-08-30 中国科学技术大学 Small molecule covalent inhibitor of targeted cyclic guanylic acid-adenylic acid synthetase and application thereof

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN1857259A (en) * 2005-04-30 2006-11-08 韩国生命工学研究院 Medicinal composition for preventing and/or treating liver disease

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
RU2011146544A (en) * 2009-04-17 2013-05-27 Силин Фармасьютикалз, Инк. METHODS FOR TREATING DISEASES RELATED TO THE PROTEINKINASE CK2 ACTIVITY
AU2010326268B2 (en) * 2009-12-04 2016-11-03 Senhwa Biosciences, Inc. Pyrazolopyrimidines and related heterocycles as CK2 inhibitors

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN1857259A (en) * 2005-04-30 2006-11-08 韩国生命工学研究院 Medicinal composition for preventing and/or treating liver disease

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
LIU, CHAO ET AL.: "Effect of Tetrabromophthalic Triazole on Cell Apoptosis of Thyroid Gland Squamous Carcinoma SW579", CHINESE JOURNAL OF GERONTOLOGY, vol. 33, no. 1, 31 January 2013 (2013-01-31), ISSN: 1005-9202 *
LIU, M. ET AL.: "Discovery of Flavonoid Derivatives as anti-HCV Agents via Pharmacophore Search Combining Molecular Docking Strategy", EUROPEAN JOURNAL OF MEDICINAL CHEMISTRY, vol. 52, 30 June 2012 (2012-06-30), ISSN: 0223-5234 *
SMITH, M.C. ET AL.: "CK2 Inhibitors Increase the Sensitivity of HSV-1 to Interferon-p", ANTIVIRAL RESEARCH, vol. 91, no. 3, 30 September 2011 (2011-09-30), XP028267508, ISSN: 0166-3542, DOI: doi:10.1016/j.antiviral.2011.06.009 *
YAO, KAI ET AL.: "Tetrabromocinnamic Acid Reduces Cell Proliferation and Causes Cell Cycle Arrest in Prostate Cancer Cells", JOURNAL OF MODERN UROLOGY, vol. 18, no. 6, 30 November 2013 (2013-11-30), ISSN: 1009-8291 *

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP4126845A4 (en) * 2020-03-30 2024-05-01 Senhwa Biosciences Inc Antiviral compounds and method for treating rna viral infection, particularly covid-19
WO2021236578A1 (en) * 2020-05-18 2021-11-25 Salk Institute For Biological Studies Chromen-4-one derivatives, such as e.g. flavones, for use as ck2 inhibitors for the treatment of neuroinflammation

Also Published As

Publication number Publication date
CN105457029A (en) 2016-04-06

Similar Documents

Publication Publication Date Title
WO2016050203A1 (en) Use of inhibiting activity of casein kinase 2 for enhancing expression of type i interferon
Liu et al. Distinct and orchestrated functions of RNA sensors in innate immunity
Hiscott et al. Manipulation of the nuclear factor-κB pathway and the innate immune response by viruses
Onomoto et al. Regulation of RIG-I-like receptor-mediated signaling: interaction between host and viral factors
Perng et al. ISG15 in antiviral immunity and beyond
O'Neill et al. Sensing and signaling in antiviral innate immunity
Mukherjee et al. Activation of the innate signaling molecule MAVS by bunyavirus infection upregulates the adaptor protein SARM1, leading to neuronal death
Zhang et al. IL-17A contributes to brain ischemia reperfusion injury through calpain-TRPC6 pathway in mice
Nomi et al. Toll-like receptor 3 signaling induces apoptosis in human head and neck cancer via survivin associated pathway
Peltier et al. Human neuronal cells possess functional cytoplasmic and TLR-mediated innate immune pathways influenced by phosphatidylinositol-3 kinase signaling
JP7017012B2 (en) Hepatitis B virus-derived polypeptide and its antiviral use
Zhang et al. FOXO3a/p27kip1 expression and essential role after acute spinal cord injury in adult rat
Yang et al. Mechanisms and consequences of Newcastle disease virus W protein subcellular localization in the nucleus or mitochondria
Jing et al. TRIM59 inhibits porcine reproductive and respiratory syndrome virus (PRRSV)-2 replication in vitro
Schlee et al. Beyond double-stranded RNA-type I IFN induction by 3pRNA and other viral nucleic acids
Paun et al. The innate antiviral response: new insights into a continuing story
Michaelis et al. Increased replication of human cytomegalovirus in retinal pigment epithelial cells by valproic acid depends on histone deacetylase inhibition
Gao et al. Role of autophagy in inhibiting the proliferation of A549 cells by type III interferon
Zhang et al. Grouper USP12 exerts antiviral activity against nodavirus infection
Katayama et al. Proteolytic disassembly of viral outer capsid proteins is crucial for reovirus-mediated type-I interferon induction in both reovirus-susceptible and reovirus-refractory tumor cells
Chen et al. Influence of silencing TRAF6 with shRNA on LPS/TLR4 signaling in vitro
Cinatl et al. West Nile virus infection induces interferon signalling in human retinal pigment epithelial cells
IL297228A (en) Cell-derived vesicles comprising wild-type p53 protein for antiviral therapy
Fan et al. RIG-I-dependent antiviral immunity is effective against an RNA virus encoding a potent suppressor of RNAi
CN117442732B (en) New application of lncRNA SCARNA2 in antiviral

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 15845961

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 15845961

Country of ref document: EP

Kind code of ref document: A1