KR20110067407A - Treatment for lupus and lupus nephritis - Google Patents

Treatment for lupus and lupus nephritis Download PDF

Info

Publication number
KR20110067407A
KR20110067407A KR1020090123985A KR20090123985A KR20110067407A KR 20110067407 A KR20110067407 A KR 20110067407A KR 1020090123985 A KR1020090123985 A KR 1020090123985A KR 20090123985 A KR20090123985 A KR 20090123985A KR 20110067407 A KR20110067407 A KR 20110067407A
Authority
KR
South Korea
Prior art keywords
lupus
bee venom
cells
nephritis
regulatory
Prior art date
Application number
KR1020090123985A
Other languages
Korean (ko)
Inventor
이의준
Original Assignee
이의준
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 이의준 filed Critical 이의준
Priority to KR1020090123985A priority Critical patent/KR20110067407A/en
Publication of KR20110067407A publication Critical patent/KR20110067407A/en

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/56Materials from animals other than mammals
    • A61K35/63Arthropods

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Insects & Arthropods (AREA)
  • Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Epidemiology (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)

Abstract

PURPOSE: A pharmaceutical composition containing bee venom for treating lupus and lupus nephritis is provided to activate CD4+ CD25+ regulatory T cells. CONSTITUTION: A pharmaceutical composition for treating lupus and lupus nephritis contains bee venom as an active ingredient. The bee venom activates CD4+ CD25+ regulatory T cells and reduces IgG, IgG2, and anti-ds-DNA production. The bee venom also activates CD4+ CD25+ regulatory T cells and reduces TNF-alpha IL-6 production. A therapeutic agent contains the bee venom as an active ingredient.

Description

Pharmaceutical composition for treating lupus nephritis {Treatment for lupus and lupus nephritis}

The present invention relates to a pharmaceutical composition for treating lupus and lupus nephritis, and more particularly, to activate specific cells capable of inhibiting the development of lupus and lupus nephritis, and bee venom improves the therapeutic efficacy and proteinuria for lupus and lupus nephritis. It relates to a natural substance that is effective in.

Systemic lupus erythematosus (SLE) is a chronic autoimmune inflammatory disease with a complex clinical symptom called lupus, caused by excessive antibody production of hyper-activated B and T cells. It is an autoimmune disease considered to be pathogenic autoantibodies and immunocomplexes.

Lupus nephritis is a serious complication of systemic lupus erythematosus that occurs frequently in patients with systemic lupus erythematosus, which not only directly contributes to morbidity and mortality, but also indirectly leads to kidney failure (Agrawal). et al., 2006).

Although many other immune and non-immune factors contribute to the development of lupus nephritis, the production of autoantibodies against the nucleat and endogenous antigens and the formation of glomerular immune deposits Plays an important role in the development of lupus nephritis (Deocharan et al., 2002; Lefkowith and Gilkeson, 1996).

In many studies, anti-DNA antibodies are important diagnostic markers that bind cell surface antigens or directly cross-reactive antigen (s) or inflammatory processes in the kidneys. The ability to indirectly bind glomerular basal membrane components through starting chromatin materials has been reported to be associated with the development of lupus nephritis (Yung and Chan, 2008).

Furthermore, cytokines and chemoattractants induced by renal cells and invasive immune cells exacerbate immunocomplex mediated renal imjury (Aringer and Smolen, 2005; Kulkarni, 2005; Kulkarni). and Anders, 2008).

Lupus treatment is currently focused on immunosuppressants such as corticosteroids, cyclophosphamide, azathioprine, and mycophenolate mofetil (Waldman and Appel, 2006). . However, these drugs carry dangerous side effects that are susceptible to infectious diseases and cancer (Radis et al., 1995).

This raises interest in the development of molecules that directly counter inflammatory responses, as well as low-toxic drugs for controlling immune complex formation and deposition.

Early nephritis in systemic lupus erythematosus has been reported to be due to the generation of pathogenic autoantibodies, deposition of immune complexes, and activation of multistage complement (Cabral and Alarcon-Segovia, 1998; Lefkowith and Gilkeson, 1996). However, it is known that invasion of macrophages and lymphocytes (primarily T cells) in the kidney plays an important role in the pathogenesis of the kidneys (Kuroiwa and Lee, 1998). Therefore, it is important to reduce pathogenic autoantibody mediators and immune cell mediators that affect the treatment of lupus nephritis.

Bee venom (BV), on the other hand, is one of the oriental medicine therapies that have been widely used for the treatment of several immune-related diseases, especially rheumatoid arthritis (RA) (Suh et al., 2006).

In many studies bee venom has been shown to reduce rheumatoid arthritis in human and animal studies and to have an anti-inflammatory effect (Yoon et al., 2008). In addition, Zusanli acupoint injection of bee venom has been known to have anti-inflammatory and anti-pain effects on the Freund's adjuvant-induced arthritis in rats. Many studies have been conducted to demonstrate its effectiveness.

Bee venom is a bee stored by honey bees in their own sac for self-defense. Bee venom contains melittin, apamin, aolapin and mast cell degranulation peptides. It consists of a variety of peptides, including MCD-peptide, as well as various enzymes such as phospholipase A2 and non-peptide components such as histamines, carbonhydrates and lipids. kwon et al., 2001).

Among them, melittin is the main ingredient that accounts for about 50% of bee venom, and Hartman et al. (1991) reported that melittin injection reduces rat edema, and melittin is the main ingredient of bee venom. It has been reported to inhibit the enzymatic activity of phospholipase A2 (Saini et al., 1997).

Therefore, the present inventors have studied whether the bee venom is effective in the treatment of lupus and lupus glomerulonephritis, and found that bee venom is involved in the activation of specific cells that are effective in suppressing the development of lupus and lupus nephritis. The invention came to the following.

The present invention is to solve the above problems,

The purpose of the present invention is to confirm that bee venom is involved in activating specific cells capable of inhibiting the development of lupus and lupus nephritis, and to provide natural substances with therapeutic efficacy against lupus and lupus nephritis.

According to an aspect of the present invention,

It provides a pharmaceutical composition for treating lupus and lupus nephritis, comprising bee venom as an active ingredient.

In addition, the bee venom is characterized by inhibiting the expression of lupus or lupus nephritis by activating CD4 + CD25 + regulatory T cells.

The bee venom is characterized by activating CD4 + CD25 + regulatory T cells, reducing the production of IgG and IgG2, anti-ds-DNA, and inhibiting IgG1 and IgG2a deposition.

The bee venom is characterized by reducing the production of TNF-α and IL-6, which are inflammatory molecules of nephritis by activating CD4 + CD25 + regulatory T cells.

In addition, the present invention provides a therapeutic or adjuvant for treating lupus and lupus nephritis symptoms due to inactivation of CD4 + CD25 + regulatory T cells containing bee venom as an active ingredient.

As described above, the pharmaceutical composition for treating lupus and lupus nephritis using bee venom according to the present invention is an anti-DNA antibody and IL-6, which are the etiology of lupus and lupus nephritis by activating CD4 + CD25 + regulatory T cells. Inhibition of the etiology associated with lupus and lupus nephritis, such as inhibiting production, can be usefully developed and used as pharmaceuticals and proteinuria therapeutics for inhibiting the development and progression of lupus and lupus nephritis.

Hereinafter, the present invention will be described in more detail.

The present invention provides a new use of bee venom. Bee venom is known to have anti-inflammatory activity and is used to treat arthritis. Inflammation can be caused by a variety of causes and progresses through a complex mechanism in which a myriad of substances act in vivo.

The inventors have found that bee venom is effective in improving lupus and lupus nephritis and, to elucidate its mechanism, genetically engineered New Zealand Black / white (NZB / W) that inhibited expression to resemble human systemic lupus erythematosus. Experiments using the F1 mice confirmed that bee venom had significant effects and efficacy in improving lupus and lupus nephritis.

As a result of examining the effect of bee venom according to the present invention on the symptoms of proteinuria, which is a symptom of lupus and lupus nephritis, it showed an effect of reducing proteinuria production.

In addition, the anti-DNA antibody, which is the etiology of lupus and lupus nephritis, and serum levels circulating IgG, IgG1, IgG2a, and reduced deposition.

In addition, it showed an effect of reducing TNF-α, IL-6, which affects the production of anti-DNA antibodies, which is the cause of lupus and lupus nephritis.

In addition, it showed an effect of activating CD4 + CD25 + regulatory T cells known as immune cells suppressing autoimmune disease expression.

Therefore, the present invention directly confirmed the effect of bee venom on immune cells that suppress the etiology and expression of lupus and lupus nephritis.

Specifically, after treatment with bee venom in NZB / W F1 rats with systemic lupus erythematosus, it was measured whether the symptoms of proteinuria in the rat, the pathogenesis of lupus and lupus nephritis as described above, and the increase of immune suppressor immune cells.

CD4 + CD25 + regulatory T cells are known to play a pivotal role in maintaining tolerance in rodents such as mice, squirrels, and humans (Sakaguchi et al., 1995). In addition, many studies have shown that CD4 + CD25 + regulatory T cells can reestablish autoimmunity and prevent autoimmune diseases such as multiple sclerosis and colitis (Sakaguchi et al., 2003). The famous Wolf et al. Have provided evidence that CD4 + CD25 + regulatory T cells are important inhibitory immune cells of anti-glomerular membrane glomerulonephritis in mice. Activation of anti-dsDNA-producing autoreactive B cells in lupus rats leads to overcoming inhibition of CD4 + helper T cell populations and CD4 + CD25 + regulatory T cells Ask for help. Activated CD4 + helper T cell populations produce high amounts of cytokines and form immunocomplexes that can remain bound or trapped in tissues with subsequent inflammation and organ damage. Help B cells secrete autoantibodies (La Cava, 2009).

Recent studies have shown that the transcription factor gene Foxp3 is specifically expressed by CD4 + CD25 + regulatory T cells and programs their development and function (Fortenot et al., 2003.).

Therefore, the present invention verified that bee venom activates the expression of CD4 + CD25 + regulatory T cells, as shown in FIG. 9, through the pharmaceutical composition for treating lupus and lupus nephritis including bee venom as an active ingredient, lupus nephritis and lupus It was confirmed that the expression of can be suppressed or improved.

In addition, in many studies, cytokines not only produce mutations in immune regulation, but also cause local inflammation, which ultimately leads to tissue destruction in autoimmune diseases (Aringer and Smolen, 2005). Recent investigations suggest that TNF blocking treatment may improve lupus nephritis (Segal et al., 2001). TNF stimulates IL-6 production (Feldmann and Maini, 2001), and IL-6 production affects both anti-DNA antibody production and the pathogenesis of nephritis in NZB / W in mice (Finck et al., 1994). .

Infitrating monocytes / macrophages, expressed as proinflammtory cytokines, are known to be important mediators of injury in most forms of kidney disease (Wang et al., 2008). Recently, CD4 + CD25 + T cells have demonstrated that they can exert direct inhibitory effects on monocytes / macrophages (Taams et al., 2005).

Accordingly, the present invention is activated by the bee venom of the CD4 + CD25 + regulatory T cells, the production of IL-6 affecting the production of anti-DNA antibody and anti-DNA antibody affecting the pathogenesis of nephritis is inhibited in Table 2 and Proven through the results of FIGS. 3 to 8.

Based on the above results, the pharmaceutical composition for treating lupus and lupus nephritis using bee venom according to the present invention as an active ingredient is an anti-DNA antibody and IL-, which is the pathogenesis of lupus and lupus nephritis by activating CD4 + CD25 + regulatory T cells. Inhibition of the etiology associated with lupus and lupus nephritis, such as inhibiting the production of 6, and a pharmaceutical composition comprising such bee venom as an active ingredient has been developed and useful as a medicine for inhibiting the development and progression of lupus and lupus nephritis Can be.

Hereinafter, the present invention will be described in detail by examples. The following examples are provided to illustrate the present invention, but the content of the present invention is not limited thereto.

-Experiment Preparation-

NZB / WF1 rats (6 years old) purchased from SLC Inc., Japan (Shizuoka, Japan) were used to carry out the experiment, and the specific pathogen-free status was maintained during the experiment. In addition, experiments are conducted in accordance with the ethical laws and guidelines established by Kyung Hee University for the protection and use of laboratory animals. The NZB / WF1 rats developed autoimmune disease, which shows immune complex glomerulonephritis, proteinuria and renal failure progression, suggesting human systemic lupus erythematosus (SLB), and almost all mice die within one year (Foster, 1999).

The mice were initially included in two groups of 10 animals, one group was treated with saline, and the other group was treated with bee venom. Bee venom was provided by Sigma (St. Louis, Mo, USA), and the bee venom was diluted in 0.5 mg / ml of normal saline, and the bee venom solution (3 mg / kg body wt) was once a week Each rat in the treatment group (Bee venom) was allowed to be injected subcutaneously.

In addition, the rats in the control group (Saline) are treated with the same dosing method as in the bee venom treatment group.

Experimental Example 1 Rats Survival and Weight Change after 32 Weeks

Three mice in the control group and two mice in the bee venom group died during the experiment. In addition, as a result of measuring and comparing the average body weight of the rats of the control group and the bee venom treatment group, it was confirmed that the average body weight of the rats belonging to the bee venom treatment group and the control group is not significantly different.

Experimental Example 2 Proteinuria Measurement

After the bee venom treatment and saline treatment in the bee venom treatment group and control group, respectively, proteinuria was measured from 18 weeks. Proteinuria was measured semiquantitatively by infiltrating the urine of each group of rats with woolpaper test strips (URiSCAN; Youngdong Pharmaceutical Co., Seoul, Korea).

The color change of the strip penetrated by the urine sample is visually judged compared to the standard strip according to the manufacturer's criteria and scored from 0 to 4+. Urinary protein is graded according to the following score.

0 = <15 mg / dL, 1 + = 30mg / dL, 2 + = 100mg / dL, 3 + = 300mg / dL, 4 + = 1000mg / dL.

Table 1 shows the average level of proteinuria and Figure 2 shows the effect of bee venom treatment on proteinuria, before the saline and bee venom treatment from Table 1 and Figure 2 100% rats of the control group and the bee venom group Significant proteinuria (≥ 1+ = 30mg / dL) was detected, and the mean level of urinary protein was not significantly different between the two groups until 26 weeks. However, at 30 weeks, the control group developed severe proteinuria. In contrast, the average level of urine protein in the bee venom group began to decrease. Upon completion of the study, the rats in the control group continued to degrade kidney function, whereas the rats in the bee venom control group significantly reduced amplification of urinary protein production. It can be seen that bee venom is effective in improving proteinuria.

Weeks of age 18 22 26 30 32 Control group 2.1 ± 0.78 2.4 ± 0.74 3.0 ± 0.67 3.2 ± 0.83 3.4 ± 0.85 Bee Venom Treatment Group 2.6 ± 0.53 2.9 ± 0.35 2.9 ± 0.35 2.3 ± 0.64 * 1.9 ± 1.1 *

(The above values are expressed as mean ± SEM, and statistics were used by Student's t-test; * P <0.05)

Experimental Example 3 Bee Venom Effect in IgG, IgG1, IgG2a and Anti-DNA Antibodies

-Serum Collection and Autoantibody Measurement-

After 32 weeks, blood was collected from the retro-orbital sinus with isofluorane anesthesia inhaled in each group of rats. Serum immunoglobulin (Ig) class-specific anti-DNA antibody assays were performed using mouse whole IgG, IgG1, IgG2a enzyme-linked immunosorbent assay (ELISA) kit (Bethyl Laboratories Inc., Montgomery, Tx, USA). Serum IgG antibodies that react with anti-double-stranded DNA (anti-dsDNA) are determined by ELISA purchased from Alpha Diagnostic Internationa (San Antonio, TX, USA). All ELISAs are performed according to the manufacturer's instructions.

-Measurement result

Serum levels of circulating IgG, IgG1, IgG2a and anti-DNA antibodies were measured at 16 and 32 weeks and the results are shown in Table 2 and FIG. 3 below. Table 2 shows the standard levels of the anti-DNA antibodies, Figure 3 is a view showing the production effect of IgG, IgG1, IgG2a during bee venom treatment, respectively. It was confirmed that the levels of IgG and IgG2a antibodies in the bee venom group were significantly reduced in the bee venom group compared to the control group.

16 Weeks Total IgG IgG1 IgG2a Control group 820 ± 28 770 ± 85 1400 ± 50 Bee Venom Treatment Group 690 ± 30 940 ± 130 1400 ± 69 32 Weeks Total IgG IgG1 IgG2a Control group 1100 ± 54 1300 ± 260 5300 ± 410 Bee Venom Treatment Group 700 ± 38 1300 ± 130 2400 ± 260

(The above values are expressed as mean ± SEM.)

In addition, the serum level of anti-dsDNA was measured after 32 weeks, and the results are shown in FIG. 4. 4 is a diagram showing the effect of anti-double-stranded DNA production during bee venom treatment, as shown in the anti-dsDNA antibody formation generated at the same time as the anti-DNA antibody production is confirmed that the growth in the bee venom group significantly reduced Could.

These results suggest that bee venom can inhibit the formation of IgG, IgG2a, anti-ds-DNA associated with the development of lupus and lupus nephritis.

Experimental Example 4 Effect of Bee Venom in Kidney Pathology

-Immunofluorescence staining of autoantibodies

Kidneys are embedded in Tissue-Tek OCT compounds (Sakura, CA, USA), quickly frozen with Freezer Spray, and cut through cryostats in 20-μm-thick sections. The section was permeablized for 20 minutes in PBS containing 0.5% Triton X-100 and then blocked for non-specific reactions and the sample was rhodamine-conjugated goat anti-mouse IgG1 (Santa) at the optimal concentration (pretreatment, concentration). cruz, CA, USA) and FITC-hybrid goat anti-mouse IgG2a (Santa cruz, CA, USA) antibodies. Finally, the fluorescence intensity in the glomeruli was detected with a confocal laser scannig microscope system (LSM 5 PASCAL, Carl Zeiss, Jena, Germany). Glomerular sections were tested in rats of each group. At least 10 glomeruli / sections were analyzed using the manufacturer's software (LSM% PASCAL release version 4.0 SP2).

Organizational structure and morphometric evaluation

After sacrifice for pathological evaluation of 32 week old mice, a portion of the kidney is mixed in 4% paraformaldehyde solution and embedded in paraffin. The elongate block is cut into 5 μm sections and stained with periodic acid-Schiff (PAS) reagents.

-Results of Bee Venom Treatment in Renal Pathology-

5 is a diagram showing the effect of bee venom treatment for glomerulonephritis, kidney section in the control group was confirmed that the glomerulonephritis appeared in comparison with the bee venom group. It was also found that IgG1 and IgG2a deposition was detected by immunofluorescence.

6 and 7 show the results, FIG. 6 shows a confocal image of immunological deposition, and FIG. 7 compares immunodeposition. Here, A and C (red) of Figure 6 represents IgG1 in the renal glomeruli, B and D (green) represents the immunodeposition of IgG2a in the renal glomeruli, A and B in the kidney of the control group Glomeruli, C and D are rat kidney glomeruli of the bee venom group.

As can be seen, the symbolic deposition of IgG1 and IgG2a was observed in the renal glomeruli of the control group, while the IgG1 and IgG2a were less detected in the bee venom treated mice. In other words, bee venom inhibits IgG1 and IgG2a deposition and prevents the exacerbation of immune-mediated renal trauma.

Experimental Example 5 Effect of Bee Venom on Renal Cytokine

Renal Cytokine Analysis

Rat kidneys were measured using ELISA (BD Biosciences, San Jose, CA, USA) to test pre-inflammatory molecules caused by nephritis, TNF-α, IL-6, IL-10, and IFN-γ. In short, snap-frozen kidney pieces are homogenized in PRO-PREP protein extract (iNtRON Biotechnology Inc., Seoul, Korea), incubated for 20 minutes in ice, and centrifuged for 15 minutes at 13000 rpm, 4 ° C. Supernatants are used for kidney proinflammatory cytokine arrays. Protein concentration in each supernatant is measured in the BCATM Protein Assay Kit (Thermo Fisher Scientific Inc. Rockford, IL, USA). Protein levels of cytokines are being corrected by the total amount of protein. The result is expressed as pg / mg.

Effect of Bee Venom on Renal Cytokines

Rat kidneys were measured to test pre-inflammatory molecules caused by nephritis, TNF-α, IL-6, IL-10 and IFN-γ, and the results are shown in FIG. 8. FIG. 8A shows TNF-α cytokine production, FIG. 8B shows IL-6 cytokine analysis, in which mice in the bee venom group had significantly reduced increases in TNF-α and IL-6 compared to the control group. Confirmed.

FIG. 8C shows an IL-10 cytokine assay, and FIG. 8D shows an IFN-γ cytokine assay, showing no significant difference between IFN-γ and IL-10 in rats of the control group and the bee venom group. there was.

Experimental Example 6 Effect of Bee Venom in CD4 + CD25 + Regulatory T Cells

Flow cytometric

Using flow cytometry, antibodies include FITC-hybrid anti-mouse CD4, phycoerythrin (PE) -hybrid anti-mouse Foxp3, allophycocyanin (APC) -hybridized anti-mouse CD 25. Multiple-color immunofluorescence is performed by a Mouse Regulatory T cell Staining Kit (eBioscience) according to the manufacturer's instructions. All samples are analyzed in FACSCalibur and CellQuest (BD Bioscience).

Statistical analysis

Statistical analysis of the data was performed using Prism 4.02 software (GraphicPad Software Inc., San Die Sea, CA, USA). Statistics were performed using Student's t-test.

Measurement and Results

All rats in each group received bee venom (3.0 mg / kg bod y wt) or saline once weekly for 14 weeks starting at 18 weeks. T cell subset profiles in CD4 + CD25 + (lymphocyte gated) and Foxp3 (CD4 + T cell gated) in splenocytes of 32-week-old cancer NZB / W F1 mice were measured using FACS analysis, and the results are shown in FIG. 9. It was.

Figure 9A is a diagram showing the results of FACS analysis of splenic regulatory T cells, Figure 9B is a view showing the FACS analysis of Foxp3 expression, as shown in the CD4 + CD25 + regulatory T cell population of the bee venom group rat control group It can be seen that the increase considerably. In addition, Foxp3 expression in CD4 + CD25 + regulatory T cells was also improved in the bee venom group. In addition, the results are statistically significant with a p-value <0.05.

Through the above results, it can be seen that bee venom according to the present invention improves lupus and lupus nephritis by activating the expression of CD4 + CD25 + regulatory T cells, which are autoimmune diseases suppressing immune cells, and thereby enhancing the expression of Foxp3 expressed therein. Reduce the TNF-α, IL-6, IgG, IgG1 and IgG2a, anti-DNA antibodies, which are the pathogenesis of lupus and lupus nephritis, have the effect of improving proteinuria, and thus bee venom expresses lupus and lupus nephritis And progression can be suppressed.

1 is a view showing the average body weight of the control group (saline) and bee venom group (Bee venom).

2 is a view showing the bee venom treatment effect on proteinuria.

Figure 3a is a diagram showing the effect of the production of bee venom treatment.

Figure 3b is a view showing the production effect of IgG1 during bee venom treatment.

Figure 3c is a view showing the production effect of IgG2a during bee venom treatment.

Figure 4 shows the effect of anti-double-stranded DNA production during bee venom treatment.

Figure 5 shows the effect of bee venom treatment for glomerulonephritis.

6 shows a confosal image of immune deposition.

Figure 7 is a comparison of the immunodeposition of the control group and the bee venom group ( * P <0.01, *** P <0.001).

8A shows the production of TNF-α cytokines ( * P <0.05).

FIG. 8B shows IL-6 cytokine analysis ** P <0.01).

8C shows IL-10 cytokine analysis.

8D shows IFN-γ cytokine analysis.

Figure 9a is a diagram showing the results of FACS analysis of splenic regulatory T cells of each group of 32 weeks old ( *** P <0.001).

Figure 9b is a diagram showing the results of FACS analysis of Foxp3 expression in each group of 32 weeks old ( * P <0.05).

Claims (5)

A pharmaceutical composition for treating lupus and lupus nephritis, comprising bee venom as an active ingredient. The method according to claim 1, The bee venom is a pharmaceutical composition for treating lupus and lupus nephritis, characterized in that to activate the CD4 + CD25 + regulatory T cell to inhibit the expression of lupus or lupus nephritis. The method according to claim 2, The bee venom activates CD4 + CD25 + regulatory T cells to reduce the production of IgG and IgG2, anti-ds-DNA, and inhibits IgG1 and IgG2a deposition, the pharmaceutical composition for treating lupus and lupus nephritis. The method according to claim 2, The bee venom activates CD4 + CD25 + regulatory T cells to reduce the production of TNF-α and IL-6, which are pre-inflammatory molecules of nephritis, a pharmaceutical composition for treating lupus and lupus nephritis. A therapeutic or adjuvant for the treatment of lupus and lupus nephritis symptoms due to inactivation of CD4 + CD25 + regulatory T cells containing bee venom as an active ingredient.
KR1020090123985A 2009-12-14 2009-12-14 Treatment for lupus and lupus nephritis KR20110067407A (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
KR1020090123985A KR20110067407A (en) 2009-12-14 2009-12-14 Treatment for lupus and lupus nephritis

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
KR1020090123985A KR20110067407A (en) 2009-12-14 2009-12-14 Treatment for lupus and lupus nephritis

Publications (1)

Publication Number Publication Date
KR20110067407A true KR20110067407A (en) 2011-06-22

Family

ID=44399861

Family Applications (1)

Application Number Title Priority Date Filing Date
KR1020090123985A KR20110067407A (en) 2009-12-14 2009-12-14 Treatment for lupus and lupus nephritis

Country Status (1)

Country Link
KR (1) KR20110067407A (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR101469167B1 (en) * 2012-02-27 2014-12-04 경희대학교 산학협력단 Composition for preventing or treating diseases related to abnormal suppression of regulatory T cell activation comprising bee venom-derived PLA2

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR101469167B1 (en) * 2012-02-27 2014-12-04 경희대학교 산학협력단 Composition for preventing or treating diseases related to abnormal suppression of regulatory T cell activation comprising bee venom-derived PLA2
US9526767B2 (en) 2012-02-27 2016-12-27 University-Industry Cooperation Group Of Kyung Hee University Pharmaceutical composition comprising bee venom-phospholipase A2 (BV-PLA2) for treating or preventing diseases related to degradation of abnormal regulatory T cell activity
US9919035B2 (en) 2012-02-27 2018-03-20 Inist St Co., Ltd. Pharmaceutical composition comprising bee venom-phospholipase A2 (bv-PLA2) for treating or preventing diseases related to degradation of abnormal regulatory T cell activity
US9956270B2 (en) 2012-02-27 2018-05-01 Inist St Co., Ltd. Pharmaceutical composition comprising bee venom-phospholipase A2 (BV-PLA2) for treating or preventing diseases related to degradation of abnormal regulatory T cell activity

Similar Documents

Publication Publication Date Title
Ramirez et al. Eosinophils from physiology to disease: a comprehensive review
Lee et al. Neutrophil extracellular traps (NETs) in autoimmune diseases: a comprehensive review
Brea et al. Inflammatory and neuroimmunomodulatory changes in acute cerebral ischemia
De Sarno et al. Lithium prevents and ameliorates experimental autoimmune encephalomyelitis
Park et al. Eosinophil survival and apoptosis in health and disease
Bodhankar et al. Treatment of experimental stroke with IL-10-producing B-cells reduces infarct size and peripheral and CNS inflammation in wild-type B-cell-sufficient mice
Tsai et al. Antroquinonol differentially modulates T cell activity and reduces interleukin‐18 production, but enhances Nrf2 activation, in murine accelerated severe lupus nephritis
Holdgate et al. Recent advances in primary Sjogren's syndrome
Riedel et al. IL-17F promotes tissue injury in autoimmune kidney diseases
Lee et al. Curcumin attenuates lupus nephritis upon interaction with regulatory T cells in New Zealand Black/White mice
Kim et al. HM71224, a selective Bruton’s tyrosine kinase inhibitor, attenuates the development of murine lupus
Stracher Calpain inhibitors as therapeutic agents in nerve and muscle degeneration
Milanova et al. TLR2 elicits IL‐17‐mediated RANKL expression, IL‐17, and OPG production in neutrophils from arthritic mice
Kwan et al. IL-17 deficiency attenuates allograft injury and prolongs survival in a murine model of fully MHC-mismatched renal allograft transplantation
Wang et al. Losartan suppresses the inflammatory response in collagen-induced arthritis by inhibiting the MAPK and NF-κB pathways in B and T cells
Zhang et al. FTY720 attenuates lesional interleukin‐17+ cell accumulation in rat experimental autoimmune neuritis
Huang et al. CD200Fc attenuates retinal glial responses and RGCs apoptosis after optic nerve crush by modulating CD200/CD200R1 interaction
Paoliello-Paschoalato et al. Fcγ and complement receptors and complement proteins in neutrophil activation in rheumatoid arthritis: contribution to pathogenesis and progression and modulation by natural products
Dong et al. Low-dose IL-2 treatment affords protection against subarachnoid hemorrhage injury by expanding peripheral regulatory T cells
ES2704027T3 (en) Use of ginsenoside M1 for the treatment of lupus nephritis
Lee et al. Bee venom-associated Th1/Th2 immunoglobulin class switching results in immune tolerance of NZB/W F1 murine lupus nephritis
Byun et al. Rebamipide attenuates autoimmune arthritis severity in SKG mice via regulation of B cell and antibody production
Fang et al. Anti-CD45RB antibody therapy attenuates renal ischemia-reperfusion injury by inducing regulatory B cells
Ruocco et al. The role of C5a-C5aR1 axis in bone pathophysiology: A mini-review
McRae et al. Blockade of the G-CSF Receptor Is Protective in a Mouse Model of Renal Ischemia–Reperfusion Injury

Legal Events

Date Code Title Description
A201 Request for examination
E902 Notification of reason for refusal
E601 Decision to refuse application