WO2016050203A1 - Utilisation de l'inhibition de l'activité de la caséine kinase 2 pour améliorer l'expression de l'interféron de type i - Google Patents

Utilisation de l'inhibition de l'activité de la caséine kinase 2 pour améliorer l'expression de l'interféron de type i 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
English (en)
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/fr

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

L'invention concerne l'utilisation d'un inhibiteur de la caséine kinase 2 dans la préparation d'une composition pharmaceutique destinée à améliorer l'expression de l'interféron de type I et/ou d'un gène induit par l'interféron de type I ou d'une protéine de celui-ci ; et/ou dans la préparation d'une composition pharmaceutique destinée à traiter et/ou prévenir les maladies liées à absence d'interféron de type I.
PCT/CN2015/091111 2014-09-29 2015-09-29 Utilisation de l'inhibition de l'activité de la caséine kinase 2 pour améliorer l'expression de l'interféron de type i WO2016050203A1 (fr)

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 (fr) 2016-04-07

Family

ID=55595471

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2015/091111 WO2016050203A1 (fr) 2014-09-29 2015-09-29 Utilisation de l'inhibition de l'activité de la caséine kinase 2 pour améliorer l'expression de l'interféron de type i

Country Status (2)

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

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2021236578A1 (fr) * 2020-05-18 2021-11-25 Salk Institute For Biological Studies Dérivés de chromèn-4-one, tels que par exemple des flavones, destinés à être utilisés en tant qu'inhibiteurs de ck2 pour le traitement de la neuro-inflammation
EP4126845A4 (fr) * 2020-03-30 2024-05-01 Senhwa Biosciences Inc Composés antiviraux et méthode de traitement d'une infection virale à arn, en particulier de la covid-19

Families Citing this family (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN107714711A (zh) * 2017-09-15 2018-02-23 中国科学院成都生物研究所 6‑羟基‑3‑o‑甲基山奈酚6‑o‑吡喃葡萄糖苷的新用途
CN109646678A (zh) * 2017-10-12 2019-04-19 中国科学院上海生命科学研究院 Sun2蛋白、其制药用途及药物
CN108379584B (zh) * 2018-04-12 2020-08-11 上海交复生物医药科技有限公司 RSAD2通过wnt通路影响肿瘤细胞替莫唑胺耐药性
CN108498798A (zh) * 2018-04-17 2018-09-07 上海市第六人民医院 一种防治骨骼肌萎缩的治疗药物靶点及其应用
CN114948955A (zh) * 2022-06-06 2022-08-30 中国科学技术大学 靶向环鸟苷酸-腺苷酸合成酶的小分子共价抑制剂及其用途

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN1857259A (zh) * 2005-04-30 2006-11-08 韩国生命工学研究院 用于预防和/或治疗肝脏疾病的药物组合物

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
RU2011146544A (ru) * 2009-04-17 2013-05-27 Силин Фармасьютикалз, Инк. Способы лечения болезней, связанных с активностью протеинкиназы ck2
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 (zh) * 2005-04-30 2006-11-08 韩国生命工学研究院 用于预防和/或治疗肝脏疾病的药物组合物

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 (fr) * 2020-03-30 2024-05-01 Senhwa Biosciences Inc Composés antiviraux et méthode de traitement d'une infection virale à arn, en particulier de la covid-19
WO2021236578A1 (fr) * 2020-05-18 2021-11-25 Salk Institute For Biological Studies Dérivés de chromèn-4-one, tels que par exemple des flavones, destinés à être utilisés en tant qu'inhibiteurs de ck2 pour le traitement de la neuro-inflammation

Also Published As

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

Similar Documents

Publication Publication Date Title
WO2016050203A1 (fr) Utilisation de l'inhibition de l'activité de la caséine kinase 2 pour améliorer l'expression de l'interféron de type i
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
JP7017012B2 (ja) B型肝炎ウイルス由来ポリペプチド及びその抗ウイルス用途
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
Hemmi et al. The Early Activation of T Cells Is Dependent on Type I IFN Signaling following Intramuscular Vaccination of Adenovirus Vector
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
US20230135456A1 (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 (zh) lncRNA SCARNA2在抗病毒中的新用途

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