WO2018236194A1 - Composition for prevention or treatment of fibrosis, comprising gas6 protein or receptor activator thereof - Google Patents

Composition for prevention or treatment of fibrosis, comprising gas6 protein or receptor activator thereof Download PDF

Info

Publication number
WO2018236194A1
WO2018236194A1 PCT/KR2018/007128 KR2018007128W WO2018236194A1 WO 2018236194 A1 WO2018236194 A1 WO 2018236194A1 KR 2018007128 W KR2018007128 W KR 2018007128W WO 2018236194 A1 WO2018236194 A1 WO 2018236194A1
Authority
WO
WIPO (PCT)
Prior art keywords
gas6
cells
hours
treatment
protein
Prior art date
Application number
PCT/KR2018/007128
Other languages
French (fr)
Korean (ko)
Inventor
이지희
Original Assignee
이화여자대학교 산학협력단
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 이화여자대학교 산학협력단 filed Critical 이화여자대학교 산학협력단
Publication of WO2018236194A1 publication Critical patent/WO2018236194A1/en

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/1703Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • A61K38/1709Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • A61K38/1732Lectins
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0053Mouth and digestive tract, i.e. intraoral and peroral administration

Definitions

  • the present invention relates to a composition for preventing or treating fibrosis comprising Gas6 protein or a receptor activator thereof.
  • Fibrosis is a disease in which abnormal formation, accumulation, and deposition of extracellular matrix by fibroblasts occurs and is caused by fibrosis of an organ or tissue. Fibrosis is a very fatal disease that causes organ damage. For example, IPF (idiopathic pulmonary fibrosis) is a consequence of recurrent alveolar epithelial cell injury associated with fibroblast accumulation and fibroblast differentiation, and is associated with the extracellular matrix (ECM) of the lung parenchyma ), which is a chronic, progressive, and lethal disease.
  • IPF idiopathic pulmonary fibrosis
  • ECM extracellular matrix
  • One object of the present invention is to provide a composition for preventing or treating fibrosis, which comprises a receptor activator of Gas6 protein or Gas6 protein.
  • Gas6 proteins induce secretion production of PGE 2, PGD 2 and HGF with its receptor in the epithelial cells and through those of the sub-transmission path self-inhibiting EMT developed with a secretion system, and carries the fibrosis prevention and can be treated in accordance with Therefore, the Gas6 protein and its receptor activator have excellent effects for the prevention or treatment of fibrosis.
  • FIG. 1 shows the results of confirming the shape of LA-4 cells by TGF- ⁇ and Gas6 treatment using a phase contrast microscope. After treatment with 400 ng / ml Gas6 for 20 hours, 10 ng / ml of TGF-? was treated for 48 hours or 72 hours.
  • FIG. 2 shows the results of confirming the expression level of EMT marker protein in LA-4 cells following treatment with TGF- ⁇ and Gas6. After treatment with 400 ng / ml Gas6 for 20 hours, 10 ng / ml of TGF-? was treated for 48 hours or 72 hours. * P ⁇ 0.05, compared to control group; + P ⁇ 0.05, comparison of displayed values.
  • FIG. 3 shows the results of confirming the expression level of EMT marker mRNA in LA-4 cells according to TGF- ⁇ and Gas6 treatment. After treatment with 400 ng / ml Gas6 for 20 hours, 10 ng / ml of TGF-? was treated for 48 hours or 72 hours. * P ⁇ 0.05, compared to control group; + P ⁇ 0.05, comparison of displayed values.
  • FIG. 4 shows the results of confirming the expression level of EMT marker protein in HEK-293 kidney epithelial cells following treatment with TGF- ⁇ and Gas6. After treatment with 400 ng / ml Gas6 for 20 hours, 10 ng / ml of TGF-beta was treated for 72 hours. * P ⁇ 0.05, compared to control group; + P ⁇ 0.05, comparison of displayed values.
  • FIG. 5 shows the results of confirming the expression levels of Snai1 / 2, Zeb1 / 2 and Twist1 mRNA in LA-4 cells according to TGF-beta and Gas6 treatment. After treatment with 400 ng / ml Gas6 for 20 hours, 10 ng / ml of TGF-? was treated for 48 hours or 72 hours.
  • FIG. 6 shows the results of confirming the expression levels of Snai1 / 2, Zeb1 / 2, and Twist1 mRNA in HEK-293 cells following treatment with TGF- ⁇ and Gas6. After treatment with 400 ng / ml Gas6 for 20 hours, 10 ng / ml of TGF-beta was treated for 72 hours.
  • FIG. 7 shows the results of confirming phosphorylation of Smad2 or Smad3 protein in LA-4 cells following treatment with TGF-beta and Gas6. After treatment with 400 ng / ml Gas6 for 20 hours, 10 ng / ml of TGF-beta was treated for 30 minutes or 1 hour. * P ⁇ 0.05 compared to control.
  • FIG. 8 shows the results of confirming phosphorylation of ERK1 / 2 protein in LA-4 cells following treatment with TGF-beta and Gas6. After treatment with 400 ng / ml Gas6 for 20 hours, 10 ng / ml of TGF- ⁇ was treated for 5 minutes, 30 minutes or 1 hour. * P ⁇ 0.05, compared to control group; + P ⁇ 0.05, comparison of displayed values.
  • FIG. 9 shows the results of confirming phosphorylation of AKT protein in LA-4 cells by TGF- ⁇ and Gas6 treatment. After treatment with 400 ng / ml Gas6 for 20 hours, 10 ng / ml of TGF-beta was treated for 1 hour, 3 hours or 8 hours. * P ⁇ 0.05, compared to control group; + P ⁇ 0.05, comparison of displayed values.
  • FIG. 10 shows the results of confirming phosphorylation of P38 protein in LA-4 cells following treatment with TGF- ⁇ and Gas6. After treatment with 400 ng / ml Gas6 for 20 hours, 10 ng / ml of TGF- ⁇ was treated for 15 minutes, 1 hour, or 6 hours. * P ⁇ 0.05 compared to control.
  • Fig. 11 shows the result of confirming the level of COX-2 or COX-1 mRNA in LA-4 cells according to Gas6 treatment. 400 ng / ml Gas6 were each treated with the indicated times. * P ⁇ 0.05 compared to control.
  • Fig. 12 shows the result of confirming the level of COX-2 or COX-1 protein in LA-4 cells according to Gas6 treatment. 400 ng / ml Gas6 were each treated with the indicated times. * P ⁇ 0.05 compared to control.
  • FIG. 13 shows the results of confirming the level of PGE 2 or PGD 2 protein in the LA-4 cell culture medium according to Gas6 treatment. 400 ng / ml Gas6 was treated for 8 hours or 20 hours. * P ⁇ 0.05 compared to control.
  • FIG. 14 shows the results of confirming the levels of COX-2 protein in LA-4 cells following treatment with COX-2 specific siRNA.
  • LA-4 cells were transfected with COX-2 specific siRNA or control siRNA for 6 hours. * P ⁇ 0.05 compared to control.
  • FIG. 15 shows the results of confirming the level of PGE 2 or PGD 2 protein in LA-4 cell culture medium according to treatment with COX-2 specific siRNA.
  • LA-4 cells were transfected with COX-2 specific siRNA or control siRNA for 6 hours and treated with 400 ng / ml Gas6 for 20 hours. * P ⁇ 0.05, compared to control group; + P ⁇ 0.05, comparison of displayed values
  • Fig. 16 shows the results of confirming morphological changes of LA-4 cells according to NS-398 treatment.
  • LA-4 cells were treated with 10 ⁇ M NS-398 for 1 hour, treated with 400 ng / ml Gas6 for 20 hours, and stimulated with 10 ng / ml TGF- ⁇ for 72 hours.
  • FIG. 17 shows the results of confirming mRNA levels of EMT markers in LA-4 cells according to NS-398 treatment.
  • LA-4 cells were treated with 10 ⁇ M NS-398 for 1 hour, treated with 400 ng / ml Gas6 for 20 hours, and stimulated with 10 ng / ml TGF- ⁇ for 72 hours.
  • Fig. 18 shows the results of confirming the protein level of EMT markers in LA-4 cells according to NS-398 treatment.
  • LA-4 cells were treated with 10 ⁇ M NS-398 for 1 hour, treated with 400 ng / ml Gas6 for 20 hours, and stimulated with 10 ng / ml TGF- ⁇ for 72 hours.
  • FIG. 19 shows the results of confirming mRNA levels of EMT markers in LA-4 cells according to COX-2-specific siRNA treatment.
  • LA-4 cells were transfected with COX-2 siRNA or control siRNA for 6 hours, treated with 400 ng / ml Gas6 for 20 hours, and stimulated with 10 ng / ml TGF- ⁇ for 72 hours.
  • FIG. 20 shows the results of confirming the protein level of EMT markers in LA-4 cells according to COX-2-specific siRNA treatment.
  • LA-4 cells were transfected with COX-2 siRNA or control siRNA for 6 hours, treated with 400 ng / ml Gas6 for 20 hours, and stimulated with 10 ng / ml TGF- ⁇ for 72 hours.
  • FIG. 21 shows the results of confirming mRNA levels of Snai1, Zeb1 and Twist1 in LA-4 cells according to NS-398 treatment.
  • LA-4 cells were treated with 10 ⁇ M NS-398 for 1 hour, treated with 400 ng / ml Gas6 for 20 hours, and stimulated with 10 ng / ml TGF- ⁇ for 72 hours.
  • FIG. 22 shows the results of confirming mRNA levels of Snai1, Zeb1 and Twist1 in LA-4 cells following treatment with COX-2 siRNA.
  • LA-4 cells were treated with COX-2 siRNA or control siRNA for 6 hours, treated with 400 ng / ml Gas6 for 20 hours, and stimulated with 10 ng / ml TGF- ⁇ for 72 hours.
  • FIG. 23 shows the results of confirming the phosphorylation level of ERK1 / 2 protein in LA-4 cells according to COX-2 siRNA treatment.
  • LA-4 cells were treated with COX-2 siRNA or control siRNA for 6 hours, treated with 400 ng / ml Gas6 for 20 hours, and stimulated with 10 ng / ml TGF- ⁇ for 5 minutes.
  • LA-4 cells were treated with COX-2 siRNA or control siRNA for 6 hours, treated with 400 ng / ml Gas6 for 20 hours, and stimulated with 10 ng / ml TGF- ⁇ for 8 hours.
  • LA-4 cells were treated with 400 ng / ml Gas6 for 20 hours and 10 mM of each receptor antagonist EP2 (AH-6809), EP4 (AH-23848), DP1 (BW-A868C) or DP2 (BAY-u3405)
  • EP2 AH-6809
  • EP4 AH-23848
  • DP1 BW-A868C
  • DP2 BAY-u3405
  • LA-4 cells were treated with 400 ng / ml Gas6 for 20 hours and treated with TGF- ⁇ for 48 hours in the absence or administration of 10 mM of each receptor antagonist EP2 (AH-6809) or EP4 (AH-23848) Respectively.
  • EP2 AH-6809
  • EP4 AH-23848
  • FIG. 27 shows the results of confirming mRNA levels of EMT markers in LA-4 cells following receptor antagonist treatment.
  • LA-4 cells were treated with 400 ng / ml Gas6 for 20 hours and treated with TGF-beta for 48 hours in the presence or absence of 10 mM DP2 antagonist (BAY-u3405). * P ⁇ 0.05, compared to control group; + P ⁇ 0.05, comparison of displayed values.
  • LA-4 cells were treated with 400 ng / ml Gas6 for 20 hours and treated with TGF-beta for 72 hours in the presence or absence of 10 mM DP1 antagonist (BW-A868C). * P ⁇ 0.05, compared to control group; + P ⁇ 0.05, comparison of displayed values.
  • FIG. 29 shows the results of confirming the protein level of EMT marker in LA-4 cells according to receptor antagonist treatment.
  • LA-4 cells were treated with 400 ng / ml Gas6 for 20 hours and treated with TGF- ⁇ for 48 hours in the absence or administration of 10 mM of each receptor antagonist EP2 (AH-6809) or EP4 (AH-23848) Respectively.
  • EP2 AH-6809
  • EP4 AH-23848
  • Figure 30 shows the results of confirming the protein level of EMT markers in LA-4 cells following receptor antagonist treatment.
  • LA-4 cells were treated with 400 ng / ml Gas6 for 20 hours and treated with TGF- ⁇ 1 for 48 hours in the presence or absence of 10 mM DP2 antagonist (BAY-u3405). * P ⁇ 0.05, compared to control group; + P ⁇ 0.05, comparison of displayed values.
  • LA-4 cells were treated with 400 ng / ml Gas6 for 20 hours and treated with TGF-beta for 72 hours in the presence or absence of 10 mM DP1 antagonist (BW-A868C). * P ⁇ 0.05, compared to control group; + P ⁇ 0.05, comparison of displayed values.
  • Figure 32 shows the results of confirming the levels of Snai1, Zeb1 and Twist1 mRNA in LA-4 cells following receptor antagonist treatment.
  • LA-4 cells were treated with 400 ng / ml Gas6 for 20 hours and treated with 10 mM of each receptor antagonist EP2 (AH-6809), EP4 (AH-23848) or DP2 (BAY-u3405) TGF-beta was treated for 48 hours.
  • EP2 AH-6809
  • EP4 AH-23848
  • DP2 BAY-u3405
  • Figure 33 shows the results of confirming the levels of Snai1, Zeb1 and Twist1 mRNA in LA-4 cells following receptor antagonist treatment.
  • LA-4 cells were treated with 400 ng / ml Gas6 for 20 hours and treated with TGF-beta for 72 hours in the presence or absence of 10 mM DP1 antagonist (BW-A868C). * P ⁇ 0.05, compared to control group; + P ⁇ 0.05, comparison of displayed values.
  • FIG. 34 shows the results of confirming the levels of HGF mRNA in LA-4 cells following Gas6 treatment.
  • LA-4 cells were treated with 400 ng / ml Gas6 for the indicated time. * P ⁇ 0.05 compared to control.
  • FIG. 35 shows the result of confirming the HGF protein level in the LA-4 cell culture medium according to the Gas6 treatment.
  • LA-4 cells were treated with 400 ng / ml Gas6 for 8 or 20 hours. * P ⁇ 0.05 compared to control.
  • Fig. 36 shows the result of confirming the level of HGF protein in LA-4 cells according to Gas6 treatment.
  • LA-4 cells were treated with 400 ng / ml Gas6 for 8 or 20 hours. * P ⁇ 0.05 compared to control.
  • FIG. 37 shows the results of confirming the level of RhoA protein in LA-4 cells according to RhoA-specific siRNA treatment.
  • LA-4 cells were treated with RhoA-specific siRNA or control siRNA for 24 hours. * P ⁇ 0.05 compared to control.
  • FIG. 38 shows the results of confirming the level of EMT marker mRNA in LA-4 cells according to RhoA-specific siRNA treatment.
  • LA-4 cells were treated with RhoA-specific siRNA or control siRNA for 24 hours, treated with 400 ng / ml Gas6 for 20 hours, and stimulated with 10 ng / ml TGF- ⁇ for 48 hours.
  • FIG. 39 shows the results of confirming the level of EMT marker protein in LA-4 cells according to RhoA-specific siRNA treatment.
  • LA-4 cells were treated with RhoA-specific siRNA or control siRNA for 24 hours, treated with 400 ng / ml Gas6 for 20 hours, and stimulated with 10 ng / ml TGF- ⁇ for 48 hours.
  • LA-4 cells were treated with 10 mM Y-27632 for 1 hour, treated with 400 ng / ml Gas6 for 20 hours, and stimulated with 10 ng / ml TGF- ⁇ for 48 hours. The following morphological changes were observed.
  • LA-4 cells were treated with 10 mM Y-27632 for 1 hour, treated with 400 ng / ml Gas6 for 20 hours, and stimulated with 10 ng / ml TGF- ⁇ for 48 hours.
  • Figure 42 shows the results of confirming the level of EMT marker protein in LA-4 cells according to Y-27632 treatment.
  • LA-4 cells were treated with 10 mM Y-27632 for 1 hour, treated with 400 ng / ml Gas6 for 20 hours, and stimulated with 10 ng / ml TGF- ⁇ for 48 hours.
  • FIG. 43 shows the results of confirming the levels of Snai1, Zeb1 and Twist1 mRNA in LA-4 cells according to RhoA-specific siRNA treatment.
  • LA-4 cells were transfected with RhoA siRNA or control siRNA for 24 hours, treated with 400 ng / ml Gas6 for 20 hours, and stimulated with 10 ng / ml TGF- ⁇ for 72 hours.
  • Figure 44 shows the results of confirming the levels of Snai1, Zeb1 and Twist1 mRNA in LA-4 cells according to Y-27632 treatment.
  • LA-4 cells were treated with 10 mM Y-27632 for 1 hour, treated with 400 ng / ml Gas6 for 20 hours, and stimulated with 10 ng / ml TGF- ⁇ for 48 hours.
  • Figure 45 shows the results of confirming the phosphorylation level of ERK1 / 2 protein in LA-4 cells according to RhoA-specific siRNA treatment.
  • LA-4 cells were transfected with RhoA siRNA or control siRNA for 24 hours, treated with 400 ng / ml Gas6 for 20 hours, and stimulated with 10 ng / ml TGF- ⁇ for 5 minutes.
  • Figure 46 shows the results of confirming the phosphorylation level of AKT protein in LA-4 cells according to RhoA-specific siRNA treatment.
  • LA-4 cells were transfected with RhoA siRNA or control siRNA for 24 hours, treated with 400 ng / ml Gas6 for 20 hours, and stimulated with 10 ng / ml TGF- ⁇ for 8 hours.
  • FIG. 47 shows the results of confirming the morphology of LA-4 cells according to c-Met antagonist treatment.
  • LA-4 cells were treated with 400 ng / ml Gas6 for 20 hours and treated with TGF- ⁇ for 72 hours in the presence or absence of 250 nM of c-Met antagonist PHA-665752.
  • LA-4 cells were treated with 400 ng / ml Gas6 for 20 hours and treated with TGF- ⁇ for 72 hours in the presence or absence of 250 nM of c-Met antagonist PHA-665752. * P ⁇ 0.05, compared to control group; + P ⁇ 0.05, comparison of displayed values.
  • LA-4 cells were treated with 400 ng / ml Gas6 for 20 hours and treated with TGF- ⁇ for 72 hours in the presence or absence of 250 nM of c-Met antagonist PHA-665752. * P ⁇ 0.05, compared to control group; + P ⁇ 0.05, comparison of displayed values.
  • Figure 50 shows the results of confirming mRNA levels of Snai1, Zeb1 and Twist1 in LA-4 cells following c-Met antagonist treatment.
  • LA-4 cells were treated with 400 ng / ml Gas6 for 20 hours and treated with TGF- ⁇ for 72 hours in the presence or absence of 250 nM of c-Met antagonist PHA-665752.
  • FIG. 51 shows the results of confirming the protein levels of EMT markers in LA-4 cells following PGE 2 , PGD 2 or HGF treatment.
  • LA-4 cells were treated with PGE 2 (35 or 118 pg / ml), PGD 2 (6 or 28 pg / ml) or HGF (169 or 194 pg / ml) with 10 ng / ml TGF- .
  • PGE 2 35 or 118 pg / ml
  • PGD 2 (6 or 28 pg / ml)
  • HGF 169 or 194 pg / ml
  • Figure 52 shows the results of confirming the protein level of EMT markers in LA-4 cells following HGF treatment.
  • LA-4 cells were treated with 400 ng / ml Gas6 for 20 h and then replaced with fresh medium.
  • HGF (169 or 194 pg / ml) was treated with 10 ng / ml TGF- ⁇ for 72 hours. * P ⁇ 0.05, compared to control group; + P ⁇ 0.05, comparison of displayed values.
  • FIG. 53 shows the results of confirming c-Met, EP2, EP4, DP1 or DP2 receptor protein levels in LA-4 cells following Gas6 treatment.
  • LA-4 cells were treated with 400 ng / ml Gas6 for 12 or 20 hours. * P ⁇ 0.05, compared to control group; + P ⁇ 0.05, comparison of displayed values.
  • Figure 54 shows the results of confirming the level of Axl or Mer expression in LA-4 cells following Gas6 treatment. 400 ng / ml Gas6 was treated for 0, 5, 15, 30, 60 and 120 minutes. * P ⁇ 0.05, compared to control group; + P ⁇ 0.05, comparison of displayed values.
  • Figure 55 shows the results of confirming Axl or Mer protein expression levels in LA-4 cells following Axl or Mer specific siRNA treatment.
  • LA-4 cells were treated with Axl or Mer siRNA or control siRNA for 48 hours and treated with 400 ng / ml Gas6.
  • FIG. 56 shows the results of confirming the levels of COX-2 mRNA, PGE 2 and PGD 2 expression in LA-4 cells following Axl or Mer specific siRNA treatment.
  • LA-4 cells were treated with Axl or Mer siRNA or control siRNA for 48 hours and treated with 400 ng / ml Gas6. * P ⁇ 0.05, compared to control group; + P ⁇ 0.05, comparison of displayed values.
  • FIG. 57 shows the results of confirming RhoA activity, HGF mRNA and protein expression level in LA-4 cells according to Axl or Mer specific siRNA treatment.
  • LA-4 cells were treated with Axl or Mer siRNA or control siRNA for 48 hours and treated with 400 ng / ml Gas6.
  • FIG. 58 shows the results of confirming the mRNA level of EMT marker in LA-4 cells according to Axl or Mer specific siRNA treatment.
  • LA-4 cells were treated with Axl or Mer siRNA or control siRNA for 48 hours, treated with 400 ng / ml Gas6 for 20 hours, and treated with TGF- ⁇ for 72 hours.
  • FIG. 59 shows the results of confirming the level of protein expression of EMT markers in LA-4 cells following Axl or Mer-specific siRNA treatment.
  • LA-4 cells were treated with Axl or Mer siRNA or control siRNA for 48 hours, treated with 400 ng / ml Gas6 for 20 hours, and treated with TGF- ⁇ for 72 hours.
  • Figure 60 shows mRNA levels of Snai1, Zeb1 and Twist1 in LA-4 cells following Axl or Mer specific siRNA treatment.
  • LA-4 cells were treated with Axl or Mer siRNA or control siRNA for 48 hours, treated with 400 ng / ml Gas6 for 20 hours, and treated with TGF- ⁇ for 72 hours.
  • FIG. 61 shows the results of confirming the phosphorylation of ERK1 / 2 protein in LA-4 cells following Axl or Mer specific siRNA treatment.
  • LA-4 cells were treated with Axl or Mer siRNA or control siRNA for 48 hours, treated with 400 ng / ml Gas6 for 20 hours, and treated with TGF- ⁇ for 72 hours.
  • Figure 62 shows the results of confirming phosphorylation of AKT protein in LA-4 cells following treatment with AxI or Mer specific siRNA.
  • LA-4 cells were treated with Axl or Mer siRNA or control siRNA for 48 hours, treated with 400 ng / ml Gas6 for 20 hours, and treated with TGF- ⁇ for 72 hours. * P ⁇ 0.05, compared to control group; + P ⁇ 0.05, comparison of displayed values.
  • TGF-beta shows the results of confirming the expression level of EMT marker mRNA in the mouse AT II cell according to Gas6 treatment. After treatment with 400 ng / ml Gas6 for 20 hours, TGF-beta was treated for 48 or 72 hours. * P ⁇ 0.05, compared to control group; + P ⁇ 0.05, comparison of displayed values.
  • Fig. 64 shows the result of confirming the expression level of EMT marker protein in the mouse AT II cell according to Gas6 treatment. After treatment with 400 ng / ml Gas6 for 20 hours, TGF-beta was treated for 72 hours. * P ⁇ 0.05, compared to control group; + P ⁇ 0.05, comparison of displayed values.
  • FIG. 65 shows the results of confirming the expression levels of Snai1 / 2, Zeb1 / 2 and Twist1 mRNA in the mouse AT II cells according to TGF- ⁇ and Gas6 treatment. After treatment with 400 ng / ml Gas6 for 20 hours, 10 ng / ml of TGF-? was treated for 48 hours or 72 hours. * P ⁇ 0.05, compared to control group; + P ⁇ 0.05, comparison of displayed values.
  • FIG. 66 shows the results of confirming the levels of COX-2 mRNA in premature mouse AT II cells following treatment with Gas6. 400 ng / ml Gas6 were each treated with the indicated times. * P ⁇ 0.05 compared to control.
  • 67 shows the results of confirming the expression levels of EMT marker mRNA in primary mouse AT II cells following treatment with NS-398 and Gas6.
  • 10 ⁇ M NS-398 was treated for 1 hour, treated with 400 ng / ml Gas6 for 20 hours, and treated with 10 ng / ml of TGF- ⁇ for 72 hours.
  • 68 shows the results of confirming the expression levels of Snai1 / 2, Zeb1 / 2 and Twist1 mRNA in the mouse AT II cells according to NS-398 and Gas6 treatment.
  • 10 ⁇ M NS-398 was treated for 1 hour, treated with 400 ng / ml Gas6 for 20 hours, and treated with 10 ng / ml of TGF- ⁇ for 72 hours.
  • FIG. 70 shows the results of confirming the expression levels of Snai1 / 2, Zeb1 / 2 and Twist1 mRNA in the mouse AT II cells according to treatment with AH6, BAY and Gas6.
  • treatment with 400 ng / ml Gas6 for 20 hours treatment with TGF- ⁇ alone, treatment with 10 ⁇ M of EP2 (AH-6809) or treatment with 10 ⁇ M of DP2 (BAY-u3405) were performed for 48 hours.
  • 71 shows the result of confirming the level of HGF mRNA in the mouse AT II cell according to Gas6 treatment.
  • LA-4 cells were treated with 400 ng / ml Gas6 for the indicated time. * P ⁇ 0.05 compared to control.
  • FIG. 72 shows the results of confirming the level of mature AT II cell EMT marker mRNA in the wild-type mouse according to Y-27632 treatment.
  • LA-4 cells were treated with 10 mM Y-27632 for 1 hour, treated with 400 ng / ml Gas6 for 20 hours, and stimulated with 10 ng / ml TGF- ⁇ for 48 hours.
  • LA-4 cells were treated with 10 mM Y-27632 for 1 hour, treated with 400 ng / ml Gas6 for 20 hours, and stimulated with 10 ng / ml TGF- ⁇ for 48 hours.
  • LA-4 cells were treated with 400 ng / ml Gas6 for 20 hours and treated with TGF- ⁇ for 72 hours in the presence or absence of 250 nM of c-Met antagonist PHA-665752. * P ⁇ 0.05, compared to control group; + P ⁇ 0.05, comparison of displayed values.
  • FIG. 75 shows the results of confirming the levels of Snai1, Zeb1 and Twist1 mRNA in the mouse AT II cells according to the c-Met antagonist treatment.
  • LA-4 cells were treated with 400 ng / ml Gas6 for 20 hours and treated with TGF- ⁇ for 72 hours in the presence or absence of 250 nM of c-Met antagonist PHA-665752.
  • 76 is a graph showing the effect of TGF-beta and Gas6 on human lung adenocarcinoma cells (549 cells) And the expression level of the EMT marker mRNA was confirmed. After treatment with 400 ng / ml Gas6 for 20 hours, 10 ng / ml of TGF-? was treated for 48 hours or 72 hours. * P ⁇ 0.05, compared to control group; + P ⁇ 0.05, comparison of displayed values.
  • FIG. 77 shows the results of confirming the level of EMT marker protein expression in human lung adenocarcinoma cells (549 cells) following TGF- ⁇ and Gas6 treatment. After treatment with 400 ng / ml Gas6 for 20 hours, 10 ng / ml of TGF-? was treated for 48 hours or 72 hours. * P ⁇ 0.05, compared to control group; + P ⁇ 0.05, comparison of displayed values.
  • FIG. 79 shows the results of in vivo blomycin and Gas6 treatment, in which the shape of primary mouse AT II cells was confirmed by a phase contrast microscope.
  • FIG. 82 shows the results of confirming COX-2 mRNA levels in the mouse AT II cells according to in vivo bleomycin and Gas6 treatment. * P ⁇ 0.05, compared to control group; + P ⁇ 0.05, comparison of displayed values.
  • FIG. 87 shows the results of confirming the expression levels of EMT markers and extracellular matrix proteins in the lung tissue at day 21 following in vivo treatment with bromomycin and Gas6. * P ⁇ 0.05, compared to control group; + P ⁇ 0.05, comparison of displayed values.
  • One aspect of the present invention for achieving the above object is a pharmaceutical composition for preventing or treating fibrosis, which comprises Gas6 (Growth arrest-specific 6) protein or Gas6 protein receptor activator as an active ingredient.
  • Gas6 Heating arrest-specific 6
  • Gas6 protein receptor activator as an active ingredient.
  • fibrosis in the present invention means that excessive fibrous connective tissue is formed in an organ or tissue. This can be distinguished from fibrous tissue as a normal component in the organ or tissue. Because of the excessive accumulation of extracellular metrix such as fibronectin and collagen by fibroblast, fibrosis can be understood as a fatal disease that eventually leads to organ damage.
  • epithelial to mesenchymal transition refers to a phenomenon in which epithelial cells are transformed into mesenchymal cells, and is associated with embryonic development, organ development, wound healing and stem cell behavior, (J Clin Invest, 2009, 119 (6): 1420). Fibroblasts are classified into (1) proliferation and differentiation of resident lung fibroblasts, (2) EMTs converted from (ave) epithelial cells to (my) fibroblasts, and (3) (Curr Rheumatol Rep, 2006, 8: 145).
  • the fibrotic syndrome may be selected from the group consisting of lung, kidney, liver, heart, brain, blood vessels, joints, bowel, skin, soft tissues, bone marrow, penis, peritoneum, lance, muscle, spine, testis, ovary, breast, thyroid, Gallbladder, bladder, or prostate.
  • fibrosis is a disease caused by fibrosis occurring in each tissue of the body, such as abnormal wound healing, alcohol-induced hepatic injury induced fibrosis, connective fibrosis, Crohn's disease (intestinal fibrosis), pancreatic and lung cystic Fibrosis, fibrosis caused by Graft-Versus-Host Disease (GVHD), fibrosis of the spleen, fibrosis of the spleen and retinal fibrosis, which may occur as complications of intermuscular injection, especially in children, endocardial myocardial fibrosis or cardiac fibrosis Fibrosis complications of surgery or injection fibrosis, glomerulonephritis, epileptic fibrosis, keloids and hypertrophic scarring (skin fibrosis), macular degeneration, mediastinal fibrosis (soft tissue fibrosis of the mediastinum), morph
  • Proliferative fibrosis pipestem fibrosis, postfiber fibrosis, progressive bundle fibrosis (pulmonary fibrosis type, complications of coal worker's pneumoconiosis), pleural fibrosis, fibrosis as a result of surgery (for example, (Fibrosis of the retroperitoneal soft tissue), post-surgical scarring, post-operative fibrosis, fibromyalgia, fibromyalgia, chronic fibrosis, chronic myelogenous leukemia, Scleroderma / systemic sclerosis (skin fibrosis), epithelial cells, uterine fibrosis, or viral hepatitis induced fibrosis.
  • surgery for example, (Fibrosis of the retroperitoneal soft tissue), post-surgical scarring, post-operative fibrosis, fibromyalgia, fibromyalgia, chronic fibrosis, chronic myelogenous leukemia, Scleroderma / systemic
  • pulmonary fibrosis is classified as idiopathic pulmonary fibrosis, nonspecific interstitial pneumonia, acute interstitial pneumonia, cryptogenic organizing pneumonia, respiratory bronchitis-related epilepsy But not limited to, Respiratory Bronchiolitisassociated Interstitial Lung, Desquamative Interstitial Pneumonia, Lymphoid Interstitial Pneumonia, Interstitial Pulmonary Fibrosis, and Diffuse Pulmonary Fibrosis. It is not.
  • treatment means any action that improves or alleviates the symptoms of fibrosis with the administration of the pharmaceutical composition
  • prevention means inhibiting the onset of fibrosis by administration of the pharmaceutical composition Or delaying any action that
  • the activator of Gas6 protein or Gas6 protein receptor can inhibit the EMT phenomenon, thereby confirming that the activator of Gas6 protein or Gas6 protein receptor can be used for prevention and treatment of fibrosis.
  • Growth arrest-specific protein 6 in the present invention refers to a secretable vitamin K-dependent protein, also called AXSF or AXLLG.
  • the Gas6 protein may have, for example, an amino acid sequence of the NCBI accession number (NP_062394.2) (SEQ ID NO: 35), but it is not limited thereto, and may have homology with known Gas6 protein, Deletion, addition or substitution of a part of the sequences as long as they have the same activity or Gas6 proteins derived from human or other animals can all be included in the scope of the present invention.
  • the sequence homology may be 70% or more, 80% or more, 90% or more, 95% or more, 98% or more or 99% or more of the sequence of the wild type Gas6 protein.
  • fragments obtained by cleaving some domains for activity in the wild-type Gas6 protein, and fragments having the same activity as the wild-type Gas6 protein in the form of a fusion protein in which known peptides are bound to increase the expression, isolation, purification, May be included within the Gas6 protein category of the present invention.
  • the Gas6 protein can be produced by a method known in the art, for example, through peptide synthesis or production of a protein using transformed cells, but is not limited thereto.
  • the murine Gas6 protein of SEQ ID NO: 35 was synthesized and used (R & D Systems; Minneapolis, MN, USA).
  • Gas6 contains a large C-terminal region that is homologous to the N-terminal gamma-carboxyglutamic acid (Gla) domain, four epidermal growth factor (EGF) -like domains, and sex hormone binding globulin (SHBG) Biol, 1993, 13: 4976; Blood Cells Mol Dis, 2006, 36: 373).
  • Gas6 is expressed in the lung, heart, kidney, intestine, fibroblasts, endothelial cells, bone marrow cells, vascular smooth muscle, white blood cells and neurons and is known as a common ligand of the TAM (Tyro3 / Axl / Mer) 1995, 82: 355; Nature Reviews Immunology, 2008, 8: 327).
  • the Gas6 protein receptor in the present invention includes, but is not limited to, AXL receptor tyrosine kinase, Mer tyrosine kinase, or TYRO3. These receptors share considerable domain similarity, including both extracellular N-terminal immunoglobulin-like domains and two fibronectin-III-like domains and the tyrosine kinase domain located at the following C-terminal cytoplasmic end of the receptor . TAM activity by Gas6 has been reported to induce signals that mediate cell survival, proliferation, predation, differentiation, platelet function and thrombolytic stabilization.
  • Activators of Gas6 protein receptor in the present invention is to enable the Gas6 protein receptor means a substance capable of activating the sub-transmission path, particularly in the purposes of PEG2, PGD 2 or HGF of the present invention to produce, secrete Quot; refers to the active material for the signal transduction pathway.
  • the activator of the Gas6 protein receptor may be, but is not limited to, Protein S (Pros1), Tubby and tubby-like protein 1, or Galectin3.
  • Gas6 acts on Axl or Mer receptor tyrosine kinase distributed in the cell membrane to block the TGF- ⁇ signal through COX-2-derived PGE 2 , PGD 2 secretion and RhoA-dependent HGF secretion, thus confirming anti-EMT effect (Figs. 5 to 62).
  • mouse invitation in alveolar epithelial cells was confirmed the effect of the anti--EMT natdeut receive from mouse alveolar epithelial cells, PGE 2, PGD 2 and HGF-dependent signal Gas6 (Fig. 63 to Fig. 75).
  • the anti-EMT effect was similarly observed when human Gas6 recombinant protein was administered to human lung carcinoma cells (A549 cells) (Figs. 76 to 78).
  • lung fibrosis was improved by administration of Gas6 in an animal model in which pulmonary fibrosis was induced (Figs. 79 to 88).
  • compositions of the invention produce PEG2, PGD 2 or HGF in the cell, and secretion can be suppressed EMT (epithelial to mesenchymal transition) by autocrine manner, because of the excellent effects in the prevention and treatment of fibrosis, the fibrosis Can be very useful for prevention or treatment.
  • compositions comprising a pharmaceutically acceptable carrier can be of various oral or parenteral formulations. In the case of formulation, it can be prepared using diluents or excipients such as fillers, extenders, binders, humectants, disintegrants, surfactants and the like which are usually used.
  • Solid formulations for oral administration may include tablet pills, powders, granules, capsules and the like, which may contain one or more excipients, such as starch, calcium carbonate, sucrose or lactose, lactose, gelatin, and the like.
  • Liquid preparations for oral administration include suspensions, solutions, emulsions, syrups and the like.
  • excipients such as wetting agents, sweeteners, fragrances, preservatives and the like may be included in addition to water and liquid paraffin, which are simple diluents commonly used. have.
  • Formulations for parenteral administration may include sterile aqueous solutions, non-aqueous solutions, suspensions, emulsions, freeze-dried preparations, and suppositories.
  • non-aqueous solvent and the suspending agent examples include propylene glycol, polyethylene glycol, vegetable oil such as olive oil, and injectable ester such as ethyl oleate.
  • injectable ester such as ethyl oleate.
  • the suppository base examples include witepsol, macrogol, tween 61, cacao paper, laurin, glycerogelatin and the like.
  • the pharmaceutical composition of the present invention may also be in the form of tablets, pills, powders, granules, capsules, suspensions, solutions, emulsions, syrups, sterilized aqueous solutions, nonaqueous solutions, suspensions, emulsions, A pharmaceutical preparation and a suppository.
  • the composition may be administered in a dose of 100 ⁇ ⁇ / kg to 500 ⁇ ⁇ / kg, but is not limited thereto.
  • the composition of the present invention can be administered at a different time or at the same time as the already known fibrosis treatment agent or can be applied together with known fibrosis treatment methods already known.
  • the composition may also be administered singly or multiply. It is important to take into account all of the above factors and to administer the amount in which the maximum effect can be obtained in a minimal amount without side effects, which can be easily determined by a person skilled in the art.
  • administration refers to the introduction of a pharmaceutical composition of the present invention to a subject by any suitable method, and the administration route can be administered through various routes of oral or parenteral administration as long as it can reach the target tissue .
  • the pharmaceutical composition may be appropriately administered to a subject according to the purpose or necessity, depending on the conventional method, route of administration and dosage used in the art.
  • routes of administration include oral, parenteral, subcutaneous, intraperitoneal, intrapulmonary, and intranasal routes
  • parenteral injection includes intramuscular, intravenous, intraarterial, intraperitoneal, or subcutaneous administration.
  • the appropriate dosage and the frequency of administration may be selected according to methods known in the art, and the amount and the frequency of administration of the pharmaceutical composition of the present invention to be actually administered depends on the type of symptom to be treated, route of administration, sex, , Diet, age and weight of the individual, and the severity of the disease.
  • pharmaceutically effective amount means an amount sufficient to inhibit or alleviate an increase in vascular permeability at a reasonable benefit / risk ratio applicable to medical use, and effective dosage levels will vary depending on the species and severity, Sex, the activity of the drug, the sensitivity to the drug, the time of administration, the route of administration and the rate of excretion, the duration of the treatment, factors including co-administered drugs, and other factors well known in the medical arts.
  • the term "individual" means all animals including humans, which have the fibrosis disease or the disease of the present invention.
  • administering the pharmaceutical composition of the present invention to an individual fibrosis can be prevented or treated.
  • Another aspect of the present invention is a method of preventing or treating fibrosis, comprising administering to a subject a receptor activator of Gas6 protein or Gas6 protein.
  • Gas6 protein its receptor activator
  • LA-4 and HEK-293 cells were purchased from ATCC.
  • LA-4 cells were cultured in F12K medium (Lonza, Switzerland) containing 15% FBS (fetal bovine serum) inactivated by heat treatment at 37 ° C and 5% CO 2 .
  • FBS fetal bovine serum
  • HEK-293 cells 37 °C 5% 10% in CO 2 conditions, FBS, 2 mM L- glutamine, 100 U / ml penicillin and 100 mg / ml streptomycin, DMEM containing (Dulbecco's modified Eagle's medium; US Media Tech, Inc.) Lt; / RTI >
  • the primary mouse AT II (alveolar type II) epithelial cells were isolated and purified from BALB / c mice.
  • Pulmonary artery was infused with 0.9% saline to remove pulmonary blood. After washing the lungs with 1 ml of saline, 100 units of dispase were injected into the mouse lungs and then incubated at room temperature for 45 minutes. Then, the lungs were separated from the large bronchus by mechanical means, and the separated lung tissues were cultured in DMEM medium containing 0.01% of DNase I at 37 ° C for 10 minutes. The cells were filtered, centrifuged and resuspended in a cell culture Petri dish coated with mouse IgG 0.75 mg / ml for 1 hour at 37 ° C to remove macrophages and fibroblasts, respectively, by sequential plating.
  • the purity of AT II cells evaluated by pro-SP-C immunofluorescence staining was over 90%.
  • LA-4 and HEK-293 cells were inoculated (2 x 10 5 cells / well) in 6-well culture dishes and incubated overnight in 200 ⁇ l of RPMI 1640 or DMEM containing 10% FBS.
  • the primary AT II cells were inoculated into type 1 collagen-coated culture dishes (1 x 10 6 cells / well) and cultured for 48 hours. Cells were treated with 400 ng / ml Gas6 for 20 hours in the presence or absence of 10 ng / ml TGF-beta (R & D Systems Inc).
  • 10 ⁇ M NS-398 was used for COX-2 inhibition and 30 mM Y-27632 was used for Rho kinase inhibition. Each inhibitor was added 1 hour before the treatment of Gas6.
  • AH-6809, AH-23848, BW-A868C, BAY-u3405 10 mM or PHA-665752 250 nM were used as antagonists for EP2, EP4, DP1, DP2 or c-Met, respectively.
  • Each receptor antagonist was added 1 hour before TGF-beta treatment.
  • siRNA transfection reagent Genlantis
  • 75 nM of Axl or Mer specific siRNA was used.
  • the siRNA sequences used are shown in Table 1.
  • the cells were lysed in lysis buffer containing 0.5% Triton X-100 and loaded onto 10% SDS-PAGE gels and transferred to nitrocellulose membranes.
  • the membrane was blocked with TBS (Tris-buffered saline) containing 3% BSA or 5% skim milk at room temperature and then incubated with each anti-mouse primary antibody at room temperature to attach anti-mouse HRP-conjugated secondary antibody . Protein bands were identified using enhanced chemiluminescence.
  • cDNA was normalized to the amount of HPRT (hypoxanthine-guanine phosphoribosyltransferase) and expressed as fold-change in the control.
  • RhoA activity was measured in LA-4 cell lysate using ELISA-based RhoA activation assay Biochem Kit (G-LISA; Cytoskeleton). Cell lysates were added to RhoA-GTP affinity plates coated with Rhotekin binding domain of RhoA for 30 min. The active GTP-binding form of RhoA was measured using indirect immunoassay and the chromaticity response was measured at 490 nm with a microplate spectrophotometer.
  • the content of hydroxyproline was measured using a hydroxyproline assay kit (Nanjing Jiancheng Bioengineering Co., Ltd.) according to the manufacturer's instructions.
  • mice Male C57BL / 6 mice (Orient Bio, Seongnam, Korea) were used for all experiments with 20-25 g of specific pathogen.
  • the inhibitors after the first bleomycin dosing were administered once a day and the mice were euthanized on days 14 and 21 of bleomycin administration.
  • TGF- ⁇ signaling has been shown to play an important role in EMT and fibrosis development (Am J Physiol Lung Cell Mol Physiol, 2007, 293: L525).
  • LA-4 cells were converted into spindle-like morphology when stimulated with TGF- ⁇ at 10 ng / ml for 48 hours or 72 hours.
  • TGF- ⁇ was stimulated with 400 ng / ml of Gas6 inhibited spindle-like morphological transformation of LA-4 cells and showed morphology of epithelial cells (Fig. 1).
  • the activated Smad or non-Smad signal mediates transcriptional regulation of the Snai, Zeb and Basic helix-loop-helix transcription factor families, inhibiting the expression of epithelial marker genes, (Sci Signal, 2014, 7: re8).
  • the present inventors sought to determine whether Gas6 inhibits the expression of the transcription factor in LA-4 cells stimulated with TGF- ?.
  • COX-2 / PGE 2 and PGD 2 pathways are known to inhibit EMT in lung and kidney epithelial cells (Sci Rep, 2016, 6: 20992). Therefore, the inventors sought to determine whether COX-2-derived PGE 2 and PGD 2 secreted from LA-4 in response to Gas6 mediate the anti-EMT effect in LA-4 cells by autocrine secretion.
  • the present inventors confirmed mRNA and protein expression of COX-1 and COX-2 by Gas6 and changes in production of PGE 2 and PGD 2 in LA-4 cells.
  • the level of COX-2 mRNA peaked at 1 hour after treatment with Gas6, returned to its original level at 20 hours, and the expression of COX-1 mRNA was not changed within 24 hours after treatment with Gas6 (FIG. COX-2 protein expression gradually increased until 24 hours after Gas6 treatment, but COX-1 expression did not change during this period (FIG. 12).
  • PGE 2 and PGD 2 secretion measured by EIA in LA-4 cells increased 3.3 and 2.5-fold at 20 hours after Gas6 treatment (Fig. 13).
  • LA-4 cells were transfected with a COX-2 specific siRNA or a negative-control siRNA in order to confirm that COX-2 induction by Gas6 stimulation enhanced PGE 2 and PGD 2 production in LA-4 cells And cultured for 6 hours.
  • the negative control siRNA did not affect the amount of intracellular COX-2 protein but the amount of COX-2 protein was ⁇ 60% or more higher than that of wild-type LA-4 cells in the cells transfected with COX-2 specific siRNA (Fig. 14).
  • 15 inhibited Gas6-induced PGE 2 and PGD 2 secretion when COX-2 siRNA was injected (FIG. 15), suggesting that increased production of Gas6-induced PGE 2 and PGD 2 in LA-4 cells resulted in COX-2 expression Induction < / RTI >
  • LA-4 cells were treated with the highly selective COX-2 inhibitor NS-398 for 1 hour before treatment with Gas6.
  • NS-398 reversed both the cell morphology changes induced by TGF- ⁇ and E-cadherin loss at gene and protein levels and the effect of Gas6 on the synthesis of ⁇ -SMA and N-cadherin (FIGS. 18).
  • knockdown of the COX-2 gene in LA-4 cells also reversed the effect of Gas6 (FIGS. 19 and 20).
  • both NS-398 and COX-2 siRNA reversed the effect of Gas6 on reducing TGF- ⁇ -induced Snai1, Zeb1 and Twist1 mRNA expression (FIGS. 21 and 22).
  • COX-2 siRNA reversed the effect of Gas6, which reduces phosphorylation of ERK1 / 2 and AKT induced by TGF-beta in LA-4 cells, but this result was not observed in negative control siRNA (Fig. 23 and Fig. 24 ).
  • LA-4 cells were pretreated with TGF- EP2 PGE 2, such as (E-prostanoid-2 receptor) antagonists (AH-6809), EP4 antagonists (AH-23848), DP1 antagonist (BW-A868C) or DP2 antagonists (BAY-u3405) - or PGD 2 - specific Receptor antagonist.
  • TGF- EP2 PGE 2 such as (E-prostanoid-2 receptor) antagonists (AH-6809), EP4 antagonists (AH-23848), DP1 antagonist (BW-A868C) or DP2 antagonists (BAY-u3405) - or PGD 2 - specific Receptor antagonist.
  • RhoA-specific siRNA was transfected into LA-4 cells before Gas6 stimulation for 24 hours or the Rho kinase inhibitor Y-27632 was treated for 1 hour.
  • RhoA knockdown and Rho kinase inhibition reversed the effect of Gas6 on TGF-beta induced EMT ( Figures 37-44).
  • RhoA siRNA completely reversed the effect of Gas6 on phosphorylation of TGF-beta induced ERK1 / 2 and AKT in LA-4 cells, but negative control did not show this effect (FIGS. 45 and 46).
  • PGE 2 , PGD 2 and HGF produced by the Gas6 treatment mainly cause anti-EMT signals through EP2, DP2 and c-MET, respectively.
  • the effects of the soluble mediator on LA-4 cells were evaluated at basal concentrations (35, 6 and 169 pg / ml, respectively) Stimulation concentrations (118, 28 and 194 pg / ml, respectively).
  • HGF did not show an anti-EMT effect at both stimulation (194 pg / ml) and basal (169 pg / ml) concentrations (Figure 51), but after 20 hours of pretreatment with Gas6,
  • 194 pg / ml was added, changes in EMT markers induced by TGF- ⁇ were inhibited (FIG. 52).
  • basal HGF did not show anti-EMT effect under the above experimental conditions.
  • the amount of c-MET protein was increased after 20 hours of Gas6 treatment (Fig. 53). This data suggests that despite the relatively low HGF production, HGF may have anti-EMT activity because it increases c-MET expression in the experimental conditions.
  • LA-4 cells were transfected with AxI or Mer specific siRNA or negative-control siRNA, cultured for 48 hours, and the level of AxI or Mer expression was measured (Fig. 55).
  • the negative control siRNA did not affect the amount of AxI or Mer protein, but the amount of AxI or Mer protein decreased by 70% or more in the cells transfected with AxI or Mer specific siRNA compared with wild type LA-4 cells .
  • LA-4 cells were transfected with AxI or Mer-specific siRNA or negative-control siRNA, cultured for 48 hours, and mRNA and protein expression levels of EMT markers were measured (FIGS. 58 and 59).
  • E-cadherin induced by Gas6 and ⁇ -SMA and N-cadherin decreased when Axl or Mer siRNA was transfected. This suggests that the increase in E-cadherin and decrease in? -SMA and N-cadherin induced in Gas-6 in LA-4 cells result from induction of Axl or Mer expression.
  • LA-4 cells were transfected with AxI or Mer specific siRNA or negative-control siRNA, cultured for 48 hours, and mRNA and protein expression levels of EMT regulated transcription factors were measured (Figure 60).
  • expression of mRNA of Snai1, Zeb1 and Twist1 inhibited by Gas6 was increased.
  • both ERK (extracellular signal-regulated kinase) suppressed by Gas6 and phosphorylation of Akt were both increased (Figs. 61 and 62). This suggests that the EMT transcription factor and ERK and Akt phosphorylation inhibition induced in Gas-6 in LA-4 cells are derived from induction of Axl or Mer expression.
  • the present inventors intend to confirm the regulation of COX-2 signal by Gas6 in early mouse AT II cells.
  • NS-398 a highly selective COX-2 inhibitor, for 1 hour before treatment with Gas6 in primary mouse AT II cells Lt; / RTI > As a result, NS-398 reversed the effects of Gas6 on E-cadherin loss, synthesis of ⁇ -SMA and N-cadherin, and reduction of Snai1, Zeb1 and Twist1 mRNA expression at the gene level induced by TGF- ⁇ 67 and 68).
  • TGF-beta-treated primary mouse AT II cells were treated with EP2 (E-prostanoid-2 receptor) antagonist (AH-6809) or DP2 antagonist (BAY- u3405) Antagonists of PGE 2 - or PGD 2 - specific receptors were treated.
  • EP2 E-prostanoid-2 receptor
  • DP2 antagonist BAY- u3405
  • Antagonists of PGE 2 - or PGD 2 - specific receptors were treated.
  • the anti-EMT effect of Gas6 was significantly reversed by antagonists of EP2 and DP2 (Fig. 69 and Fig. 70).
  • the present inventors have determined to modulate RhoA pathway-dependent HGF-signal by Gas6 in primary mouse AT II cells.
  • RhoA-specific siRNA was transfected into the invasive mouse AT II cells before Gas6 stimulation for 24 hours or the Rho kinase inhibitor Y-27632 was treated for 1 hour.
  • RhoA knockdown and Rho kinase inhibition reversed the effect of Gas6 on TGF-beta induced EMT (FIGS. 72 and 73).
  • treatment of PHA-665752, a c-Met antagonist, with AT II cells 1 hour before TGF-beta treatment reversed the anti-EMT effect of Gas6 (FIGS. 74 and 75).
  • RhoA / Rho kinase-dependent HGF production in AT II cells also mediates the anti-EMT effect of Gas6 via self-secretion of the c-Met signal.
  • A549 cells were treated with 400 ng / ml of Gas6, and after 20 hours, 10 ng / ml of TGF-beta was treated for 72 hours to confirm the level of EMT marker mRNA and protein expression. mRNA expression levels were measured on the basis of Hprt mRNA ( Figures 76 and 77). This data suggests that synthetic and secreted mediators after Gas6 treatment may block TGF- ⁇ signaling in a self-secreted manner in A549 cells.
  • TGF-beta treatment of A549 cells after 20 hours of treatment with Gas6 inhibited the expression of mRNA of Snai1 / 2, Zeb1 / 2 and Twist1 induced by TGF-beta 78).
  • GasE6 treatment reduces the transcriptional regulators to prevent EMT processes in A549 cells.
  • the present inventors sought to determine whether administration of Gas6 regulates the EMT process in lung epithelial cells in vivo.
  • C57BL / 6 mice were divided into five groups by Control, Gas6 group, Bleomycin group (BLM) and Gas6 + Bleomycin group (Gas6 + BLM). Each day, 50 ⁇ g / kg of Gas6 was administered according to each taxon before administration of bleomycin. At day 14 after bleomycin administration, primary mouse AT II cells were isolated from the lungs of mice.
  • the morphology of isolated mouse cells was determined. As a result, it was confirmed that the cells of the mice to which bromomycin was administered were converted into spindle-like morphology, but the cells of the mice to which Gas6 was administered before the bleomycin administration showed a decrease in the spindle-like morphology 79).
  • EMT markers E-cadherin, N-cadherin and a-SMA
  • EMT-activated transcription factors Snai1, Zeb1 and Twist1
  • each mRNA level was measured (FIGS. 80 and 81) .
  • E-cadherin expression and N-cadherin and ⁇ -SMA expression were increased in bleomycin-treated mice, but the level of EMT marker expression was inhibited by Gas6 treatment at the mRNA level.
  • gas6 treatment it was confirmed that expression of mRNA of Snai1, Zeb1, and Twist1 induced by bleomycin was all inhibited when Gas6 was treated.
  • the present inventors sought to determine whether administration of Gas6 in vivo regulates PGE 2 , PGD 2 and HGF pathways.
  • C57BL / 6 mice were divided into five groups by Control, Gas6 group, Bleomycin group (BLM) and Gas6 + Bleomycin group (Gas6 + BLM). Each day, 50 ⁇ g / kg of Gas6 was administered according to each taxon before administration of bleomycin. At day 14 after bleomycin administration, primary mouse AT II cells were isolated from the lungs of mice. The production of mRNA, PGE 2 , PGD 2 , HGF and TGF- ⁇ of COX-2 and HGF was confirmed in isolated mouse cells.
  • the present inventors sought to determine whether administration of Gas6 in vivo controls changes in EMT markers and collagen deposition.
  • C57BL / 6 mice were divided into five groups by Control, Gas6 group, Bleomycin group (BLM) and Gas6 + Bleomycin group (Gas6 + BLM). Each day, 50 ⁇ g / kg of Gas6 was administered according to each taxon before administration of bleomycin.
  • Early mouse AT II cells were isolated from the lungs of mice on days 14 and 21 after administration of bleomycin. Each separated mouse cell was Western blotted using anti-E-cadherin, anti-N-cadherin, anti- ⁇ -SMA, anti-type 1 collagen 1 or anti-fibronectin antibody. Relative density of each band was determined based on ⁇ -tubulin.
  • Gas6 has induced the secretory production of PGE 2, PGD 2 and HGF in epithelial cells and, through a signal transmission path through their receptors party by acting as a secretion system, induced by TGF- ⁇ EMT-inhibiting effect of EMT.
  • Gas6 in LA-4 cells secretes COX-2 signal pathway and RhoA pathway dependent HGF based on Axl or Mer signal pathway and has the same effect on LA-4 cell in primary mouse AT II cell and human lung adenocarcinoma cell .
  • Gas6 can be very usefully used for the prevention or treatment of fibrosis.

Abstract

The present invention relates to a composition for prevention or treatment of fibrosis, comprising Gas6 protein or a receptor activator thereof. Gas6 protein can induce the secretion and production of PGE2, PGD2, and HGF through a receptor thereof in epithelial cells and inhibit an EMT phenomenon in an auto-secretion manner through a downstream signaling pathway thereof, thereby preventing and treating fibrosis. Thus, Gas6 protein and a receptor activator thereof has an excellent effect for use in prevention or treatment of fibrosis.

Description

Gas6 단백질 또는 이의 수용체 활성화제를 포함하는 섬유증의 예방 또는 치료용 조성물Composition for preventing or treating fibrosis comprising a Gas6 protein or a receptor activator thereof
본 발명은 Gas6 단백질 또는 이의 수용체 활성화제를 포함하는 섬유증의 예방 또는 치료용 조성물에 관한 것이다.The present invention relates to a composition for preventing or treating fibrosis comprising Gas6 protein or a receptor activator thereof.
섬유증은 섬유아세포에 의한 세포 외 기질의 비정상적 생성, 축적 및 침착이 일어나는 질환으로, 기관 또는 조직의 섬유화에 의해 발병한다. 섬유증은 장기 손상을 유발하는 매우 치명적인 질환이다. 일례로 IPF (idiopathic pulmonary fibrosis)는 섬유아세포 축적 및 근섬유아세포 분화와 관련된 재발성 폐포 상피세포 손상의 결과로 나타나며, 폐 실질 (lung parenchyma) 조직의 비가역적 파괴와 함께 세포 외 기질 (extracellular matrix; ECM)의 과량 축적을 야기하는 만성, 진행성, 그리고 치사성 질환이다. 그러나 현재까지 섬유증에 대한 효과적인 치료법이 없는 실정이므로 (Fibrogenesis Tissue Repair, 2012, 5(1): 11; N Engl J Med, 2001, 345(7): 517), 섬유증을 효과적으로 예방 또는 치료할 수 있는 치료제 개발이 지속적으로 요구되고 있다.Fibrosis is a disease in which abnormal formation, accumulation, and deposition of extracellular matrix by fibroblasts occurs and is caused by fibrosis of an organ or tissue. Fibrosis is a very fatal disease that causes organ damage. For example, IPF (idiopathic pulmonary fibrosis) is a consequence of recurrent alveolar epithelial cell injury associated with fibroblast accumulation and fibroblast differentiation, and is associated with the extracellular matrix (ECM) of the lung parenchyma ), Which is a chronic, progressive, and lethal disease. However, there is no effective treatment for fibrosis until now (Fibrogenesis Tissue Repair, 2012, 5 (1): 11; N Engl J Med, 2001, 345 (7): 517) Development continues to be required.
본 발명자들은 섬유증의 예방 또는 치료용 조성물을 개발하기 위해 예의 노력한 결과, Gas6 단백질의 새로운 역할 및 폐포 제2형 상피세포의 EMT (epithelial to mesenchymal transition), 이동 및 침습에 대한 Gas6 단백질의 작용기전을 규명하고, 블레오마이신으로 유도된 폐 섬유증 마우스 모델에서 Gas6의 예방 및 치료 효과를 확인함으로써 본 발명을 완성하였다.As a result of intensive efforts to develop a composition for the prevention or treatment of fibrosis, the present inventors have found that the new role of Gas6 protein and the mechanism of action of Gas6 protein on epithelial to mesenchymal transition (EMT), migration and invasion of alveolar type II epithelial cells And confirmed the preventive and therapeutic effects of Gas6 in a bleomycin-induced pulmonary fibrosis mouse model, thereby completing the present invention.
본 발명의 하나의 목적은 Gas6 단백질 또는 Gas6 단백질의 수용체 활성화제를 포함하는, 섬유증의 예방 또는 치료용 조성물을 제공하는 것이다.One object of the present invention is to provide a composition for preventing or treating fibrosis, which comprises a receptor activator of Gas6 protein or Gas6 protein.
본 발명의 다른 하나의 목적은 Gas6 단백질 또는 Gas6 단백질의 수용체 활성화제를 개체에 투여하는 단계를 포함하는, 섬유증의 예방 또는 치료 방법을 제공하는 것이다.It is another object of the present invention to provide a method for preventing or treating fibrosis, which comprises administering to a subject a receptor activator of Gas6 protein or Gas6 protein.
Gas6 단백질은 상피세포에서 그 수용체를 통해 PGE2, PGD2 및 HGF의 분비 생산을 유도하고, 이들의 하위 신호전달 경로를 거쳐 자가분비 방식으로 EMT 현상을 억제하며, 이에 따라 섬유증을 예방 및 치료할 수 있으므로, Gas6 단백질 및 이의 수용체 활성화제는 섬유증의 예방 또는 치료 용도로 매우 우수한 효과를 가진다.Gas6 proteins induce secretion production of PGE 2, PGD 2 and HGF with its receptor in the epithelial cells and through those of the sub-transmission path self-inhibiting EMT developed with a secretion system, and carries the fibrosis prevention and can be treated in accordance with Therefore, the Gas6 protein and its receptor activator have excellent effects for the prevention or treatment of fibrosis.
도 1은 TGF-β및 Gas6 처리에 따른 LA-4 세포의 모양을 위상차 현미경으로 확인한 결과를 나타낸다. 400 ng/ml Gas6를 20 시간 처리한 뒤 10 ng/ml의 TGF-β를 48 시간 또는 72 시간 처리하였다. FIG. 1 shows the results of confirming the shape of LA-4 cells by TGF-β and Gas6 treatment using a phase contrast microscope. After treatment with 400 ng / ml Gas6 for 20 hours, 10 ng / ml of TGF-? Was treated for 48 hours or 72 hours.
도 2는 TGF-β및 Gas6 처리에 따른 LA-4 세포 내 EMT 마커 단백질의 발현 수준을 확인한 결과를 나타낸다. 400 ng/ml Gas6를 20 시간 처리한 뒤 10 ng/ml의 TGF-β를 48 시간 또는 72 시간 처리하였다. *P < 0.05, 대조군 대비; +P < 0.05, 표시된 값 비교. FIG. 2 shows the results of confirming the expression level of EMT marker protein in LA-4 cells following treatment with TGF-β and Gas6. After treatment with 400 ng / ml Gas6 for 20 hours, 10 ng / ml of TGF-? Was treated for 48 hours or 72 hours. * P < 0.05, compared to control group; + P < 0.05, comparison of displayed values.
도 3은 TGF-β및 Gas6 처리에 따른 LA-4 세포 내 EMT 마커 mRNA의 발현 수준을 확인한 결과를 나타낸다. 400 ng/ml Gas6를 20 시간 처리한 뒤 10 ng/ml의 TGF-β를 48 시간 또는 72 시간 처리하였다. *P < 0.05, 대조군 대비; +P < 0.05, 표시된 값 비교. FIG. 3 shows the results of confirming the expression level of EMT marker mRNA in LA-4 cells according to TGF-β and Gas6 treatment. After treatment with 400 ng / ml Gas6 for 20 hours, 10 ng / ml of TGF-? Was treated for 48 hours or 72 hours. * P < 0.05, compared to control group; + P < 0.05, comparison of displayed values.
도 4는 TGF-β및 Gas6 처리에 따른 HEK-293 신장 상피 세포 내 EMT 마커 단백질의 발현 수준을 확인한 결과를 나타낸다. 400 ng/ml Gas6를 20 시간 처리한 뒤 10 ng/ml의 TGF-β를 72 시간 처리하였다. *P < 0.05, 대조군 대비; +P < 0.05, 표시된 값 비교. FIG. 4 shows the results of confirming the expression level of EMT marker protein in HEK-293 kidney epithelial cells following treatment with TGF-β and Gas6. After treatment with 400 ng / ml Gas6 for 20 hours, 10 ng / ml of TGF-beta was treated for 72 hours. * P < 0.05, compared to control group; + P < 0.05, comparison of displayed values.
도 5은 TGF-β및 Gas6 처리에 따른 LA-4 세포 내 Snai1/2, Zeb1/2 및 Twist1 mRNA의 발현 수준을 확인한 결과를 나타낸다. 400 ng/ml Gas6를 20 시간 처리한 뒤 10 ng/ml의 TGF-β를 48 시간 또는 72 시간 처리하였다. FIG. 5 shows the results of confirming the expression levels of Snai1 / 2, Zeb1 / 2 and Twist1 mRNA in LA-4 cells according to TGF-beta and Gas6 treatment. After treatment with 400 ng / ml Gas6 for 20 hours, 10 ng / ml of TGF-? Was treated for 48 hours or 72 hours.
도 6은 TGF-β및 Gas6 처리에 따른 HEK-293 세포 내 Snai1/2, Zeb1/2, 및 Twist1 mRNA 발현 수준을 확인한 결과를 나타낸다. 400 ng/ml Gas6를 20 시간 처리한 뒤 10 ng/ml의 TGF-β를 72 시간 처리하였다.FIG. 6 shows the results of confirming the expression levels of Snai1 / 2, Zeb1 / 2, and Twist1 mRNA in HEK-293 cells following treatment with TGF-β and Gas6. After treatment with 400 ng / ml Gas6 for 20 hours, 10 ng / ml of TGF-beta was treated for 72 hours.
도 7은 TGF-β및 Gas6 처리에 따른 LA-4 세포 내 Smad2 또는 Smad3 단백질의 인산화를 확인한 결과를 나타낸다. 400 ng/ml Gas6를 20 시간 처리한 뒤 10 ng/ml의 TGF-β를 30 분 또는 1 시간 처리하였다. *P < 0.05, 대조군 대비.FIG. 7 shows the results of confirming phosphorylation of Smad2 or Smad3 protein in LA-4 cells following treatment with TGF-beta and Gas6. After treatment with 400 ng / ml Gas6 for 20 hours, 10 ng / ml of TGF-beta was treated for 30 minutes or 1 hour. * P <0.05 compared to control.
도 8는 TGF-β및 Gas6 처리에 따른 LA-4 세포 내 ERK1/2 단백질의 인산화를 확인한 결과를 나타낸다. 400 ng/ml Gas6를 20 시간 처리한 뒤 10 ng/ml의 TGF-β를 5 분, 30 분 또는 1 시간 처리하였다. *P < 0.05, 대조군 대비; +P < 0.05, 표시된 값 비교.FIG. 8 shows the results of confirming phosphorylation of ERK1 / 2 protein in LA-4 cells following treatment with TGF-beta and Gas6. After treatment with 400 ng / ml Gas6 for 20 hours, 10 ng / ml of TGF-β was treated for 5 minutes, 30 minutes or 1 hour. * P < 0.05, compared to control group; + P < 0.05, comparison of displayed values.
도 9은 TGF-β및 Gas6 처리에 따른 LA-4 세포 내 AKT 단백질의 인산화를 확인한 결과를 나타낸다. 400 ng/ml Gas6를 20 시간 처리한 뒤 10 ng/ml의 TGF-β를 1 시간, 3 시간 또는 8 시간 처리하였다. *P < 0.05, 대조군 대비; +P < 0.05, 표시된 값 비교.FIG. 9 shows the results of confirming phosphorylation of AKT protein in LA-4 cells by TGF-β and Gas6 treatment. After treatment with 400 ng / ml Gas6 for 20 hours, 10 ng / ml of TGF-beta was treated for 1 hour, 3 hours or 8 hours. * P < 0.05, compared to control group; + P < 0.05, comparison of displayed values.
도 10은 TGF-β및 Gas6 처리에 따른 LA-4 세포 내 P38 단백질의 인산화를 확인한 결과를 나타낸다. 400 ng/ml Gas6를 20 시간 처리한 뒤 10 ng/ml의 TGF-β를 15 분, 1 시간 또는 6 시간 처리하였다. *P < 0.05, 대조군 대비.FIG. 10 shows the results of confirming phosphorylation of P38 protein in LA-4 cells following treatment with TGF-β and Gas6. After treatment with 400 ng / ml Gas6 for 20 hours, 10 ng / ml of TGF-β was treated for 15 minutes, 1 hour, or 6 hours. * P <0.05 compared to control.
도 11는 Gas6 처리에 따른 LA-4 세포 내 COX-2 또는 COX-1 mRNA 수준을 확인한 결과를 나타낸다. 400 ng/ml Gas6를 각각 표시된 시간으로 처리하였다. *P < 0.05, 대조군 대비.Fig. 11 shows the result of confirming the level of COX-2 or COX-1 mRNA in LA-4 cells according to Gas6 treatment. 400 ng / ml Gas6 were each treated with the indicated times. * P <0.05 compared to control.
도 12은 Gas6 처리에 따른 LA-4 세포 내 COX-2 또는 COX-1 단백질 수준을 확인한 결과를 나타낸다. 400 ng/ml Gas6를 각각 표시된 시간으로 처리하였다. *P < 0.05, 대조군 대비.Fig. 12 shows the result of confirming the level of COX-2 or COX-1 protein in LA-4 cells according to Gas6 treatment. 400 ng / ml Gas6 were each treated with the indicated times. * P <0.05 compared to control.
도 13는 Gas6 처리에 따른 LA-4 세포 배양 배지 내 PGE2 또는 PGD2 단백질 수준을 확인한 결과를 나타낸다. 400 ng/ml Gas6를 8 시간 또는 20 시간 처리하였다. *P < 0.05, 대조군 대비.FIG. 13 shows the results of confirming the level of PGE 2 or PGD 2 protein in the LA-4 cell culture medium according to Gas6 treatment. 400 ng / ml Gas6 was treated for 8 hours or 20 hours. * P <0.05 compared to control.
도 14는 COX-2 특이적 siRNA 처리에 따른 LA-4 세포 내 COX-2 단백질 수준을 확인한 결과를 나타낸다. LA-4 세포를 COX-2 특이적 siRNA 또는 대조군 siRNA로 6 시간 동안 형질주입하였다. *P < 0.05, 대조군 대비.FIG. 14 shows the results of confirming the levels of COX-2 protein in LA-4 cells following treatment with COX-2 specific siRNA. LA-4 cells were transfected with COX-2 specific siRNA or control siRNA for 6 hours. * P <0.05 compared to control.
도 15은 COX-2 특이적 siRNA 처리에 따른 LA-4 세포 배양 배지 내 PGE2 또는 PGD2 단백질 수준을 확인한 결과를 나타낸다. LA-4 세포를 COX-2 특이적 siRNA 또는 대조군 siRNA로 6 시간 동안 형질주입한 다음, 400 ng/ml Gas6를 20 시간 처리하였다. *P < 0.05, 대조군 대비; +P < 0.05, 표시된 값 비교FIG. 15 shows the results of confirming the level of PGE 2 or PGD 2 protein in LA-4 cell culture medium according to treatment with COX-2 specific siRNA. LA-4 cells were transfected with COX-2 specific siRNA or control siRNA for 6 hours and treated with 400 ng / ml Gas6 for 20 hours. * P < 0.05, compared to control group; + P < 0.05, comparison of displayed values
도 16은 NS-398 처리에 따른 LA-4 세포의 형태 변화를 확인한 결과를 나타낸다. LA-4 세포에 10 μM NS-398을 1 시간 처리하고 400 ng/ml Gas6를 20 시간 처리한 뒤, 10 ng/ml TGF-β으로 72 시간 자극하였다. Fig. 16 shows the results of confirming morphological changes of LA-4 cells according to NS-398 treatment. LA-4 cells were treated with 10 μM NS-398 for 1 hour, treated with 400 ng / ml Gas6 for 20 hours, and stimulated with 10 ng / ml TGF-β for 72 hours.
도 17은 NS-398 처리에 따른 LA-4 세포 내 EMT 마커의 mRNA 수준을 확인한 결과를 나타낸다. LA-4 세포에 10 μM NS-398을 1 시간 처리하고 400 ng/ml Gas6를 20 시간 처리한 뒤, 10 ng/ml TGF-β으로 72 시간 자극하였다. *P < 0.05, 대조군 대비; +P < 0.05, 표시된 값 비교.FIG. 17 shows the results of confirming mRNA levels of EMT markers in LA-4 cells according to NS-398 treatment. LA-4 cells were treated with 10 μM NS-398 for 1 hour, treated with 400 ng / ml Gas6 for 20 hours, and stimulated with 10 ng / ml TGF-β for 72 hours. * P < 0.05, compared to control group; + P < 0.05, comparison of displayed values.
도 18는 NS-398 처리에 따른 LA-4 세포 내 EMT 마커의 단백질 수준을 확인한 결과를 나타낸다. LA-4 세포에 10 μM NS-398을 1 시간 처리하고 400 ng/ml Gas6를 20 시간 처리한 뒤, 10 ng/ml TGF-β으로 72 시간 자극하였다. *P < 0.05, 대조군 대비; +P < 0.05, 표시된 값 비교.Fig. 18 shows the results of confirming the protein level of EMT markers in LA-4 cells according to NS-398 treatment. LA-4 cells were treated with 10 μM NS-398 for 1 hour, treated with 400 ng / ml Gas6 for 20 hours, and stimulated with 10 ng / ml TGF-β for 72 hours. * P < 0.05, compared to control group; + P < 0.05, comparison of displayed values.
도 19은 COX-2 특이적 siRNA 처리에 따른 LA-4 세포 내 EMT 마커의 mRNA 수준을 확인한 결과를 나타낸다. LA-4 세포에 COX-2 siRNA 또는 대조군 siRNA를 6 시간 형질주입하고 400 ng/ml Gas6를 20 시간 처리한 뒤, 10 ng/ml TGF-β으로 72 시간 자극하였다. *P < 0.05, 대조군 대비; +P < 0.05, 표시된 값 비교.FIG. 19 shows the results of confirming mRNA levels of EMT markers in LA-4 cells according to COX-2-specific siRNA treatment. LA-4 cells were transfected with COX-2 siRNA or control siRNA for 6 hours, treated with 400 ng / ml Gas6 for 20 hours, and stimulated with 10 ng / ml TGF-β for 72 hours. * P < 0.05, compared to control group; + P < 0.05, comparison of displayed values.
도 20은 COX-2 특이적 siRNA 처리에 따른 LA-4 세포 내 EMT 마커의 단백질 수준을 확인한 결과를 나타낸다. LA-4 세포에 COX-2 siRNA 또는 대조군 siRNA를 6 시간 형질주입하고 400 ng/ml Gas6를 20 시간 처리한 뒤, 10 ng/ml TGF-β으로 72 시간 자극하였다. *P < 0.05, 대조군 대비; +P < 0.05, 표시된 값 비교.FIG. 20 shows the results of confirming the protein level of EMT markers in LA-4 cells according to COX-2-specific siRNA treatment. LA-4 cells were transfected with COX-2 siRNA or control siRNA for 6 hours, treated with 400 ng / ml Gas6 for 20 hours, and stimulated with 10 ng / ml TGF-β for 72 hours. * P < 0.05, compared to control group; + P < 0.05, comparison of displayed values.
도 21는 NS-398 처리에 따른 LA-4 세포 내 Snai1, Zeb1 및 Twist1의 mRNA 수준을 확인한 결과를 나타낸다. LA-4 세포에 10 μM NS-398을 1 시간 처리하고 400 ng/ml Gas6를 20 시간 처리한 뒤, 10 ng/ml TGF-β으로 72 시간 자극을 주었다. *P < 0.05, 대조군 대비; +P < 0.05, 표시된 값 비교.FIG. 21 shows the results of confirming mRNA levels of Snai1, Zeb1 and Twist1 in LA-4 cells according to NS-398 treatment. LA-4 cells were treated with 10 μM NS-398 for 1 hour, treated with 400 ng / ml Gas6 for 20 hours, and stimulated with 10 ng / ml TGF-β for 72 hours. * P < 0.05, compared to control group; + P < 0.05, comparison of displayed values.
도 22은 COX-2 siRNA 처리에 따른 LA-4 세포 내 Snai1, Zeb1 및 Twist1의 mRNA 수준을 확인한 결과를 나타낸다. LA-4 세포에 COX-2 siRNA 또는 대조군 siRNA를 6 시간 처리하고 400 ng/ml Gas6를 20 시간 처리한 뒤, 10 ng/ml TGF-β으로 72 시간 자극을 주었다. *P < 0.05, 대조군 대비; +P < 0.05, 표시된 값 비교.FIG. 22 shows the results of confirming mRNA levels of Snai1, Zeb1 and Twist1 in LA-4 cells following treatment with COX-2 siRNA. LA-4 cells were treated with COX-2 siRNA or control siRNA for 6 hours, treated with 400 ng / ml Gas6 for 20 hours, and stimulated with 10 ng / ml TGF-β for 72 hours. * P < 0.05, compared to control group; + P < 0.05, comparison of displayed values.
도 23는 COX-2 siRNA 처리에 따른 LA-4 세포 내 ERK1/2 단백질의 인산화 수준을 확인한 결과를 나타낸다. LA-4 세포에 COX-2 siRNA 또는 대조군 siRNA를 6 시간 처리하고 400 ng/ml Gas6를 20 시간 처리한 뒤, 10 ng/ml TGF-β으로 5 분 자극을 주었다. *P < 0.05, 대조군 대비; +P < 0.05, 표시된 값 비교.FIG. 23 shows the results of confirming the phosphorylation level of ERK1 / 2 protein in LA-4 cells according to COX-2 siRNA treatment. LA-4 cells were treated with COX-2 siRNA or control siRNA for 6 hours, treated with 400 ng / ml Gas6 for 20 hours, and stimulated with 10 ng / ml TGF-β for 5 minutes. * P < 0.05, compared to control group; + P < 0.05, comparison of displayed values.
도 24는 COX-2 siRNA 처리에 따른 LA-4 세포 내 AKT 단백질의 인산화 수준을 확인한 결과를 나타낸다. LA-4 세포에 COX-2 siRNA 또는 대조군 siRNA를 6 시간 처리하고 400 ng/ml Gas6를 20 시간 처리한 뒤, 10 ng/ml TGF-β으로 8 시간 자극을 주었다. *P < 0.05, 대조군 대비; +P < 0.05, 표시된 값 비교.24 shows the results of confirming the phosphorylation level of AKT protein in LA-4 cells according to COX-2 siRNA treatment. LA-4 cells were treated with COX-2 siRNA or control siRNA for 6 hours, treated with 400 ng / ml Gas6 for 20 hours, and stimulated with 10 ng / ml TGF-β for 8 hours. * P < 0.05, compared to control group; + P < 0.05, comparison of displayed values.
도 25은 수용체 길항제 처리에 따른 LA-4 세포의 형태 변화를 확인한 결과를 나타낸다. LA-4 세포에 400 ng/ml Gas6를 20 시간 처리하고, 각 수용체의 길항제 EP2 (AH-6809), EP4 (AH-23848), DP1 (BW-A868C) 또는 DP2 (BAY-u3405) 10 mM을 투여하거나 투여하지 않은 조건에서 TGF-β을 48 시간 또는 72 시간째에 형태 변화를 확인하였다.25 shows the result of confirming morphological changes of LA-4 cells according to receptor antagonist treatment. LA-4 cells were treated with 400 ng / ml Gas6 for 20 hours and 10 mM of each receptor antagonist EP2 (AH-6809), EP4 (AH-23848), DP1 (BW-A868C) or DP2 (BAY-u3405) The morphological changes of TGF-β were observed at 48 hours or 72 hours after administration or no administration.
도 26은 수용체 길항제 처리에 따른 LA-4 세포 내 EMT 마커의 mRNA 수준을 확인한 결과를 나타낸다. LA-4 세포에 400 ng/ml Gas6를 20 시간 처리하고, 각 수용체의 길항제 EP2 (AH-6809) 또는 EP4 (AH-23848) 10 mM을 투여하거나 투여하지 않은 조건에서 TGF-β을 48 시간 처리하였다. *P < 0.05, 대조군 대비; +P < 0.05, 표시된 값 비교.26 shows the results of confirming mRNA levels of EMT markers in LA-4 cells following receptor antagonist treatment. LA-4 cells were treated with 400 ng / ml Gas6 for 20 hours and treated with TGF-β for 48 hours in the absence or administration of 10 mM of each receptor antagonist EP2 (AH-6809) or EP4 (AH-23848) Respectively. * P < 0.05, compared to control group; + P < 0.05, comparison of displayed values.
도 27은 수용체 길항제 처리에 따른 LA-4 세포 내 EMT 마커의 mRNA 수준을 확인한 결과를 나타낸다. LA-4 세포에 400 ng/ml Gas6를 20 시간 처리하고, 10 mM DP2 길항제 (BAY-u3405)이 존재하거나 존재하지 않는 조건에서 TGF-β을 48 시간 처리하였다. *P < 0.05, 대조군 대비; +P < 0.05, 표시된 값 비교.FIG. 27 shows the results of confirming mRNA levels of EMT markers in LA-4 cells following receptor antagonist treatment. LA-4 cells were treated with 400 ng / ml Gas6 for 20 hours and treated with TGF-beta for 48 hours in the presence or absence of 10 mM DP2 antagonist (BAY-u3405). * P < 0.05, compared to control group; + P < 0.05, comparison of displayed values.
도 28는 수용체 길항제 처리에 따른 LA-4 세포 내 EMT 마커의 mRNA 수준을 확인한 결과를 나타낸다. LA-4 세포에 400 ng/ml Gas6를 20 시간 처리하고, 10 mM DP1 길항제 (BW-A868C)이 존재하거나 존재하지 않는 조건에서 TGF-β을 72 시간 처리하였다. *P < 0.05, 대조군 대비; +P < 0.05, 표시된 값 비교.28 shows the results of confirming the level of mRNA of EMT markers in LA-4 cells following receptor antagonist treatment. LA-4 cells were treated with 400 ng / ml Gas6 for 20 hours and treated with TGF-beta for 72 hours in the presence or absence of 10 mM DP1 antagonist (BW-A868C). * P < 0.05, compared to control group; + P < 0.05, comparison of displayed values.
도 29은 수용체 길항제 처리에 따른 LA-4 세포 내 EMT 마커의 단백질 수준을 확인한 결과를 나타낸다. LA-4 세포에 400 ng/ml Gas6를 20 시간 처리하고, 각 수용체의 길항제 EP2 (AH-6809) 또는 EP4 (AH-23848) 10 mM을 투여하거나 투여하지 않은 조건에서 TGF-β을 48 시간 처리하였다. *P < 0.05, 대조군 대비; +P < 0.05, 표시된 값 비교.FIG. 29 shows the results of confirming the protein level of EMT marker in LA-4 cells according to receptor antagonist treatment. LA-4 cells were treated with 400 ng / ml Gas6 for 20 hours and treated with TGF-β for 48 hours in the absence or administration of 10 mM of each receptor antagonist EP2 (AH-6809) or EP4 (AH-23848) Respectively. * P < 0.05, compared to control group; + P < 0.05, comparison of displayed values.
도 30은 수용체 길항제 처리에 따른 LA-4 세포 내 EMT 마커의 단백질 수준을 확인한 결과를 나타낸다. LA-4 세포에 400 ng/ml Gas6를 20 시간 처리하고, 10 mM DP2 길항제 (BAY-u3405)이 존재하거나 존재하지 않는 조건에서 TGF-β1을 48 시간 처리하였다. *P < 0.05, 대조군 대비; +P < 0.05, 표시된 값 비교.Figure 30 shows the results of confirming the protein level of EMT markers in LA-4 cells following receptor antagonist treatment. LA-4 cells were treated with 400 ng / ml Gas6 for 20 hours and treated with TGF-β1 for 48 hours in the presence or absence of 10 mM DP2 antagonist (BAY-u3405). * P < 0.05, compared to control group; + P < 0.05, comparison of displayed values.
도 31는 수용체 길항제 처리에 따른 LA-4 세포 내 EMT 마커의 단백질 수준을 확인한 결과를 나타낸다. LA-4 세포에 400 ng/ml Gas6를 20 시간 처리하고, 10 mM DP1 길항제 (BW-A868C)이 존재하거나 존재하지 않는 조건에서 TGF-β을 72 시간 처리하였다. *P < 0.05, 대조군 대비; +P < 0.05, 표시된 값 비교.31 shows the results of confirming the protein level of EMT markers in LA-4 cells according to receptor antagonist treatment. LA-4 cells were treated with 400 ng / ml Gas6 for 20 hours and treated with TGF-beta for 72 hours in the presence or absence of 10 mM DP1 antagonist (BW-A868C). * P < 0.05, compared to control group; + P < 0.05, comparison of displayed values.
도 32은 수용체 길항제 처리에 따른 LA-4 세포 내 Snai1, Zeb1 및 Twist1 mRNA 수준을 확인한 결과를 나타낸다. LA-4 세포에 400 ng/ml Gas6를 20 시간 처리하고, 각 수용체의 길항제 EP2 (AH-6809), EP4 (AH-23848) 또는 DP2 (BAY-u3405) 10 mM을 투여하거나 투여하지 않은 조건에서 TGF-β을 48 시간 처리하였다. *P < 0.05, 대조군 대비; +P < 0.05, 표시된 값 비교.Figure 32 shows the results of confirming the levels of Snai1, Zeb1 and Twist1 mRNA in LA-4 cells following receptor antagonist treatment. LA-4 cells were treated with 400 ng / ml Gas6 for 20 hours and treated with 10 mM of each receptor antagonist EP2 (AH-6809), EP4 (AH-23848) or DP2 (BAY-u3405) TGF-beta was treated for 48 hours. * P < 0.05, compared to control group; + P < 0.05, comparison of displayed values.
도 33는 수용체 길항제 처리에 따른 LA-4 세포 내 Snai1, Zeb1 및 Twist1 mRNA 수준을 확인한 결과를 나타낸다. LA-4 세포에 400 ng/ml Gas6를 20 시간 처리하고, 10 mM DP1 길항제 (BW-A868C)이 존재하거나 존재하지 않는 조건에서 TGF-β을 72 시간 처리하였다. *P < 0.05, 대조군 대비; +P < 0.05, 표시된 값 비교.Figure 33 shows the results of confirming the levels of Snai1, Zeb1 and Twist1 mRNA in LA-4 cells following receptor antagonist treatment. LA-4 cells were treated with 400 ng / ml Gas6 for 20 hours and treated with TGF-beta for 72 hours in the presence or absence of 10 mM DP1 antagonist (BW-A868C). * P < 0.05, compared to control group; + P < 0.05, comparison of displayed values.
도 34는 Gas6 처리에 따른 LA-4 세포 내 HGF mRNA 수준을 확인한 결과를 나타낸다. LA-4 세포에 400 ng/ml Gas6를 표시된 시간으로 처리하였다. *P < 0.05, 대조군 대비.FIG. 34 shows the results of confirming the levels of HGF mRNA in LA-4 cells following Gas6 treatment. LA-4 cells were treated with 400 ng / ml Gas6 for the indicated time. * P <0.05 compared to control.
도 35은 Gas6 처리에 따른 LA-4 세포 배양 배지 내 HGF 단백질 수준을 확인한 결과를 나타낸다. LA-4 세포에 400 ng/ml Gas6를 8 시간 또는 20 시간 처리하였다. *P < 0.05, 대조군 대비.FIG. 35 shows the result of confirming the HGF protein level in the LA-4 cell culture medium according to the Gas6 treatment. LA-4 cells were treated with 400 ng / ml Gas6 for 8 or 20 hours. * P <0.05 compared to control.
도 36은 Gas6 처리에 따른 LA-4 세포 내 HGF 단백질 수준을 확인한 결과를 나타낸다. LA-4 세포에 400 ng/ml Gas6를 8 시간 또는 20 시간 처리하였다. *P < 0.05, 대조군 대비.Fig. 36 shows the result of confirming the level of HGF protein in LA-4 cells according to Gas6 treatment. LA-4 cells were treated with 400 ng / ml Gas6 for 8 or 20 hours. * P <0.05 compared to control.
도 37은 RhoA 특이적 siRNA 처리에 따른 LA-4 세포 내 RhoA 단백질 수준을 확인한 결과를 나타낸다. LA-4 세포에 RhoA 특이적 siRNA 또는 대조군 siRNA를 24 시간 처리하였다. *P < 0.05, 대조군 대비.FIG. 37 shows the results of confirming the level of RhoA protein in LA-4 cells according to RhoA-specific siRNA treatment. LA-4 cells were treated with RhoA-specific siRNA or control siRNA for 24 hours. * P <0.05 compared to control.
도 38는 RhoA 특이적 siRNA 처리에 따른 LA-4 세포 내 EMT 마커 mRNA 수준을 확인한 결과를 나타낸다. LA-4 세포에 RhoA 특이적 siRNA 또는 대조군 siRNA를 24 시간 처리한 뒤 400 ng/ml Gas6를 20 시간 처리한 뒤, 10 ng/ml TGF-β으로 48 시간 자극하였다. *P < 0.05, 대조군 대비; +P < 0.05, 표시된 값 비교.FIG. 38 shows the results of confirming the level of EMT marker mRNA in LA-4 cells according to RhoA-specific siRNA treatment. LA-4 cells were treated with RhoA-specific siRNA or control siRNA for 24 hours, treated with 400 ng / ml Gas6 for 20 hours, and stimulated with 10 ng / ml TGF-β for 48 hours. * P < 0.05, compared to control group; + P < 0.05, comparison of displayed values.
도 39은 RhoA 특이적 siRNA 처리에 따른 LA-4 세포 내 EMT 마커 단백질 수준을 확인한 결과를 나타낸다. LA-4 세포에 RhoA 특이적 siRNA 또는 대조군 siRNA를 24 시간 처리한 뒤 400 ng/ml Gas6를 20 시간 처리한 뒤, 10 ng/ml TGF-β으로 48 시간 자극하였다. *P < 0.05, 대조군 대비; +P < 0.05, 표시된 값 비교.FIG. 39 shows the results of confirming the level of EMT marker protein in LA-4 cells according to RhoA-specific siRNA treatment. LA-4 cells were treated with RhoA-specific siRNA or control siRNA for 24 hours, treated with 400 ng / ml Gas6 for 20 hours, and stimulated with 10 ng / ml TGF-β for 48 hours. * P < 0.05, compared to control group; + P < 0.05, comparison of displayed values.
도 40은 Y-27632 처리에 따른 LA-4 세포의 형태 변화를 확인한 결과를 나타낸다. LA-4 세포에 10 mM Y-27632를 1 시간 처리한 뒤 400 ng/ml Gas6를 20 시간 처리한 처리한 뒤, 10 ng/ml TGF-β으로 48 시간 자극하였다. 다음 세포 형태 변화를 관찰하였다. 40 shows the results of confirming morphological changes of LA-4 cells according to Y-27632 treatment. LA-4 cells were treated with 10 mM Y-27632 for 1 hour, treated with 400 ng / ml Gas6 for 20 hours, and stimulated with 10 ng / ml TGF-β for 48 hours. The following morphological changes were observed.
도 41는 Y-27632 처리에 따른 LA-4 세포 내 EMT 마커 mRNA 수준을 확인한 결과를 나타낸다. LA-4 세포에 10 mM Y-27632를 1 시간 처리한 뒤 400 ng/ml Gas6를 20 시간 처리한 뒤, 10 ng/ml TGF-β으로 48 시간 자극하였다. *P < 0.05, 대조군 대비; +P < 0.05, 표시된 값 비교.41 shows the results of confirming the level of EMT marker mRNA in LA-4 cells according to Y-27632 treatment. LA-4 cells were treated with 10 mM Y-27632 for 1 hour, treated with 400 ng / ml Gas6 for 20 hours, and stimulated with 10 ng / ml TGF-β for 48 hours. * P < 0.05, compared to control group; + P < 0.05, comparison of displayed values.
도 42은 Y-27632 처리에 따른 LA-4 세포 내 EMT 마커 단백질 수준을 확인한 결과를 나타낸다. LA-4 세포에 10 mM Y-27632를 1 시간 처리한 뒤 400 ng/ml Gas6를 20 시간 처리하한 뒤, 10 ng/ml TGF-β으로 48 시간 자극하였다. *P < 0.05, 대조군 대비; +P < 0.05, 표시된 값 비교.Figure 42 shows the results of confirming the level of EMT marker protein in LA-4 cells according to Y-27632 treatment. LA-4 cells were treated with 10 mM Y-27632 for 1 hour, treated with 400 ng / ml Gas6 for 20 hours, and stimulated with 10 ng / ml TGF-β for 48 hours. * P < 0.05, compared to control group; + P < 0.05, comparison of displayed values.
도 43는 RhoA 특이적 siRNA 처리에 따른 LA-4 세포 내 Snai1, Zeb1 및 Twist1 mRNA 수준을 확인한 결과를 나타낸다. LA-4 세포에 RhoA siRNA 또는 대조군 siRNA를 24 시간 형질주입한 뒤, 400 ng/ml Gas6를 20 시간 처리한 뒤, 10 ng/ml TGF-β으로 72 시간 자극하였다. *P < 0.05, 대조군 대비; +P < 0.05, 표시된 값 비교.FIG. 43 shows the results of confirming the levels of Snai1, Zeb1 and Twist1 mRNA in LA-4 cells according to RhoA-specific siRNA treatment. LA-4 cells were transfected with RhoA siRNA or control siRNA for 24 hours, treated with 400 ng / ml Gas6 for 20 hours, and stimulated with 10 ng / ml TGF-β for 72 hours. * P < 0.05, compared to control group; + P < 0.05, comparison of displayed values.
도 44는 Y-27632 처리에 따른 LA-4 세포 내 Snai1, Zeb1 및 Twist1 mRNA 수준을 확인한 결과를 나타낸다. LA-4 세포에 10 mM Y-27632를 1 시간 처리한 뒤, 400 ng/ml Gas6를 20 시간 처리한 뒤, 10 ng/ml TGF-β으로 48 시간 자극하였다. *P < 0.05, 대조군 대비; +P < 0.05, 표시된 값 비교.Figure 44 shows the results of confirming the levels of Snai1, Zeb1 and Twist1 mRNA in LA-4 cells according to Y-27632 treatment. LA-4 cells were treated with 10 mM Y-27632 for 1 hour, treated with 400 ng / ml Gas6 for 20 hours, and stimulated with 10 ng / ml TGF-β for 48 hours. * P < 0.05, compared to control group; + P < 0.05, comparison of displayed values.
도 45은 RhoA 특이적 siRNA 처리에 따른 LA-4 세포 내 ERK1/2 단백질의 인산화 수준을 확인한 결과를 나타낸다. LA-4 세포에 RhoA siRNA 또는 대조군 siRNA를 24 시간 형질주입한 뒤, 400 ng/ml Gas6를 20 시간 처리한 뒤, 10 ng/ml TGF-β으로 5 분 자극하였다. *P < 0.05, 대조군 대비; +P < 0.05, 표시된 값 비교.Figure 45 shows the results of confirming the phosphorylation level of ERK1 / 2 protein in LA-4 cells according to RhoA-specific siRNA treatment. LA-4 cells were transfected with RhoA siRNA or control siRNA for 24 hours, treated with 400 ng / ml Gas6 for 20 hours, and stimulated with 10 ng / ml TGF-β for 5 minutes. * P < 0.05, compared to control group; + P < 0.05, comparison of displayed values.
도 46은 RhoA 특이적 siRNA 처리에 따른 LA-4 세포 내 AKT 단백질의 인산화 수준을 확인한 결과를 나타낸다. LA-4 세포에 RhoA siRNA 또는 대조군 siRNA를 24 시간 형질주입한 뒤, 400 ng/ml Gas6를 20 시간 처리한 뒤, 10 ng/ml TGF-β으로 8 시간 자극하였다. *P < 0.05, 대조군 대비; +P < 0.05, 표시된 값 비교.Figure 46 shows the results of confirming the phosphorylation level of AKT protein in LA-4 cells according to RhoA-specific siRNA treatment. LA-4 cells were transfected with RhoA siRNA or control siRNA for 24 hours, treated with 400 ng / ml Gas6 for 20 hours, and stimulated with 10 ng / ml TGF-β for 8 hours. * P < 0.05, compared to control group; + P < 0.05, comparison of displayed values.
도 47은 c-Met 길항제 처리에 따른 LA-4 세포의 형태를 확인한 결과를 나타낸다. LA-4 세포에 400 ng/ml Gas6를 20 시간 처리한 뒤, c-Met 길항제인 PHA-665752 250 nM이 존재하거나 존재하지 않는 조건에서 TGF-β을 72 시간 처리하고 형태 변화를 관찰하였다. FIG. 47 shows the results of confirming the morphology of LA-4 cells according to c-Met antagonist treatment. LA-4 cells were treated with 400 ng / ml Gas6 for 20 hours and treated with TGF-β for 72 hours in the presence or absence of 250 nM of c-Met antagonist PHA-665752.
도 48는 c-Met 길항제 처리에 따른 LA-4 세포 내 EMT 마커의 mRNA 수준을 확인한 결과를 나타낸다. LA-4 세포에 400 ng/ml Gas6를 20 시간 처리한 뒤, c-Met 길항제인 PHA-665752 250 nM이 존재하거나 존재하지 않는 조건에서 TGF-β을 72 시간 처리하였다. *P < 0.05, 대조군 대비; +P < 0.05, 표시된 값 비교.48 shows the result of confirming the mRNA level of EMT marker in LA-4 cells according to c-Met antagonist treatment. LA-4 cells were treated with 400 ng / ml Gas6 for 20 hours and treated with TGF-β for 72 hours in the presence or absence of 250 nM of c-Met antagonist PHA-665752. * P < 0.05, compared to control group; + P < 0.05, comparison of displayed values.
도 49은 c-Met 길항제 처리에 따른 LA-4 세포 내 EMT 마커의 단백질 수준을 확인한 결과를 나타낸다. LA-4 세포에 400 ng/ml Gas6를 20 시간 처리한 뒤, c-Met 길항제인 PHA-665752 250 nM이 존재하거나 존재하지 않는 조건에서 TGF-β을 72 시간 처리하였다. *P < 0.05, 대조군 대비; +P < 0.05, 표시된 값 비교.49 shows the results of confirming the protein level of the EMT marker in LA-4 cells according to the c-Met antagonist treatment. LA-4 cells were treated with 400 ng / ml Gas6 for 20 hours and treated with TGF-β for 72 hours in the presence or absence of 250 nM of c-Met antagonist PHA-665752. * P < 0.05, compared to control group; + P < 0.05, comparison of displayed values.
도 50은 c-Met 길항제 처리에 따른 LA-4 세포 내 Snai1, Zeb1 및 Twist1의 mRNA 수준을 확인한 결과를 나타낸다. LA-4 세포에 400 ng/ml Gas6를 20 시간 처리한 뒤, c-Met 길항제인 PHA-665752 250 nM이 존재하거나 존재하지 않는 조건에서 TGF-β을 72 시간 처리하였다. *P < 0.05, 대조군 대비; +P < 0.05, 표시된 값 비교.Figure 50 shows the results of confirming mRNA levels of Snai1, Zeb1 and Twist1 in LA-4 cells following c-Met antagonist treatment. LA-4 cells were treated with 400 ng / ml Gas6 for 20 hours and treated with TGF-β for 72 hours in the presence or absence of 250 nM of c-Met antagonist PHA-665752. * P < 0.05, compared to control group; + P < 0.05, comparison of displayed values.
도 51는 PGE2, PGD2 또는 HGF 처리에 따른 LA-4 세포 내 EMT 마커의 단백질 수준을 확인한 결과를 나타낸다. LA-4 세포에 PGE2 (35 또는 118 pg/ml), PGD2 (6 또는 28 pg/ml) 또는 HGF (169 또는 194 pg/ml)를 10 ng/ml TGF-β과 함께 72 시간 처리하였다. *P < 0.05, 대조군 대비; +P < 0.05, 표시된 값 비교.FIG. 51 shows the results of confirming the protein levels of EMT markers in LA-4 cells following PGE 2 , PGD 2 or HGF treatment. LA-4 cells were treated with PGE 2 (35 or 118 pg / ml), PGD 2 (6 or 28 pg / ml) or HGF (169 or 194 pg / ml) with 10 ng / ml TGF- . * P < 0.05, compared to control group; + P < 0.05, comparison of displayed values.
도 52은 HGF 처리에 따른 LA-4 세포 내 EMT 마커의 단백질 수준을 확인한 결과를 나타낸다. LA-4 세포에 400 ng/ml Gas6를 20 시간 처리한 다음 새로운 배지로 교체한 뒤, HGF (169 또는 194 pg/ml)를 10 ng/ml TGF-β과 함께 72 시간 처리하였다. *P < 0.05, 대조군 대비; +P < 0.05, 표시된 값 비교.Figure 52 shows the results of confirming the protein level of EMT markers in LA-4 cells following HGF treatment. LA-4 cells were treated with 400 ng / ml Gas6 for 20 h and then replaced with fresh medium. HGF (169 or 194 pg / ml) was treated with 10 ng / ml TGF-β for 72 hours. * P < 0.05, compared to control group; + P < 0.05, comparison of displayed values.
도 53는 Gas6 처리에 따른 LA-4 세포 내 c-Met, EP2, EP4, DP1 또는 DP2 수용체 단백질 수준을 확인한 결과를 나타낸다. LA-4 세포에 400 ng/ml Gas6를 12 시간 또는 20 시간 처리하였다. *P < 0.05, 대조군 대비; +P < 0.05, 표시된 값 비교.FIG. 53 shows the results of confirming c-Met, EP2, EP4, DP1 or DP2 receptor protein levels in LA-4 cells following Gas6 treatment. LA-4 cells were treated with 400 ng / ml Gas6 for 12 or 20 hours. * P < 0.05, compared to control group; + P < 0.05, comparison of displayed values.
도 54는 Gas6 처리에 따른 LA-4 세포 내 Axl 또는 Mer 발현 수준을 확인한 결과를 나타낸다. 400 ng/ml Gas6를 0, 5, 15, 30, 60 및 120 분 동안 처리하였다. *P < 0.05, 대조군 대비; +P < 0.05, 표시된 값 비교.Figure 54 shows the results of confirming the level of Axl or Mer expression in LA-4 cells following Gas6 treatment. 400 ng / ml Gas6 was treated for 0, 5, 15, 30, 60 and 120 minutes. * P < 0.05, compared to control group; + P < 0.05, comparison of displayed values.
도 55은 Axl 또는 Mer 특이적 siRNA 처리에 따른 LA-4 세포 내 Axl 또는 Mer 단백질 발현 수준을 확인한 결과를 나타낸다. LA-4 세포에 Axl 또는 Mer siRNA 또는 대조군 siRNA를 48시간 동안처리하고 400 ng/ml Gas6를 처리하였다. *P < 0.05, 대조군 대비; +P < 0.05, 표시된 값 비교.Figure 55 shows the results of confirming Axl or Mer protein expression levels in LA-4 cells following Axl or Mer specific siRNA treatment. LA-4 cells were treated with Axl or Mer siRNA or control siRNA for 48 hours and treated with 400 ng / ml Gas6. * P < 0.05, compared to control group; + P < 0.05, comparison of displayed values.
도 56은 Axl 또는 Mer 특이적 siRNA 처리에 따른 LA-4 세포 내 COX-2 mRNA, PGE2 및 PGD2 발현 수준을 확인한 결과를 나타낸다. LA-4 세포에 Axl 또는 Mer siRNA 또는 대조군 siRNA를 48시간 동안처리하고 400 ng/ml Gas6를 처리하였다. *P < 0.05, 대조군 대비; +P < 0.05, 표시된 값 비교.FIG. 56 shows the results of confirming the levels of COX-2 mRNA, PGE 2 and PGD 2 expression in LA-4 cells following Axl or Mer specific siRNA treatment. LA-4 cells were treated with Axl or Mer siRNA or control siRNA for 48 hours and treated with 400 ng / ml Gas6. * P < 0.05, compared to control group; + P < 0.05, comparison of displayed values.
도 57은 Axl 또는 Mer 특이적 siRNA 처리에 따른 LA-4 세포 내 RhoA 활성, HGF mRAN 및 단백질 발현 수준을 확인한 결과를 나타낸다. LA-4 세포에 Axl 또는 Mer siRNA 또는 대조군 siRNA를 48시간 동안 처리하고 400 ng/ml Gas6를 처리하였다. *P < 0.05, 대조군 대비; +P < 0.05, 표시된 값 비교.FIG. 57 shows the results of confirming RhoA activity, HGF mRNA and protein expression level in LA-4 cells according to Axl or Mer specific siRNA treatment. LA-4 cells were treated with Axl or Mer siRNA or control siRNA for 48 hours and treated with 400 ng / ml Gas6. * P < 0.05, compared to control group; + P < 0.05, comparison of displayed values.
도 58는 Axl 또는 Mer 특이적 siRNA 처리에 따른 LA-4 세포 내 EMT 마커의 mRNA 수준을 확인한 결과를 나타낸다. LA-4 세포에 Axl 또는 Mer siRNA 또는 대조군 siRNA를 48시간 동안 처리하고 400 ng/ml Gas6를 20 시간 처리한 뒤, TGF-β을 72 시간 처리하였다. *P < 0.05, 대조군 대비; +P < 0.05, 표시된 값 비교.FIG. 58 shows the results of confirming the mRNA level of EMT marker in LA-4 cells according to Axl or Mer specific siRNA treatment. LA-4 cells were treated with Axl or Mer siRNA or control siRNA for 48 hours, treated with 400 ng / ml Gas6 for 20 hours, and treated with TGF-β for 72 hours. * P < 0.05, compared to control group; + P < 0.05, comparison of displayed values.
도 59은 Axl 또는 Mer 특이적 siRNA 처리에 따른 LA-4 세포 내 EMT 마커의 단백질 발현 수준을 확인한 결과를 나타낸다. LA-4 세포에 Axl 또는 Mer siRNA 또는 대조군 siRNA를 48시간 동안 처리하고 400 ng/ml Gas6를 20 시간 처리한 뒤, TGF-β을 72 시간 처리하였다. *P < 0.05, 대조군 대비; +P < 0.05, 표시된 값 비교.FIG. 59 shows the results of confirming the level of protein expression of EMT markers in LA-4 cells following Axl or Mer-specific siRNA treatment. LA-4 cells were treated with Axl or Mer siRNA or control siRNA for 48 hours, treated with 400 ng / ml Gas6 for 20 hours, and treated with TGF-β for 72 hours. * P < 0.05, compared to control group; + P < 0.05, comparison of displayed values.
도 60은 Axl 또는 Mer 특이적 siRNA 처리에 따른 LA-4 세포 내 Snai1, Zeb1 및 Twist1의 mRNA 수준을 확인한 결과를 나타낸다. LA-4 세포에 Axl 또는 Mer siRNA 또는 대조군 siRNA를 48시간 동안 처리하고 400 ng/ml Gas6를 20 시간 처리한 뒤, TGF-β을 72 시간 처리하였다. *P < 0.05, 대조군 대비; +P < 0.05, 표시된 값 비교.Figure 60 shows mRNA levels of Snai1, Zeb1 and Twist1 in LA-4 cells following Axl or Mer specific siRNA treatment. LA-4 cells were treated with Axl or Mer siRNA or control siRNA for 48 hours, treated with 400 ng / ml Gas6 for 20 hours, and treated with TGF-β for 72 hours. * P < 0.05, compared to control group; + P < 0.05, comparison of displayed values.
도 61는 Axl 또는 Mer 특이적 siRNA 처리에 따른 LA-4 세포 내 ERK1/2 단백질의 인산화를 확인한 결과를 나타낸다. LA-4 세포에 Axl 또는 Mer siRNA 또는 대조군 siRNA를 48시간 동안처리하고 400 ng/ml Gas6를 20 시간 처리한 뒤, TGF-β을 72 시간 처리하였다. *P < 0.05, 대조군 대비; +P < 0.05, 표시된 값 비교.FIG. 61 shows the results of confirming the phosphorylation of ERK1 / 2 protein in LA-4 cells following Axl or Mer specific siRNA treatment. LA-4 cells were treated with Axl or Mer siRNA or control siRNA for 48 hours, treated with 400 ng / ml Gas6 for 20 hours, and treated with TGF-β for 72 hours. * P < 0.05, compared to control group; + P < 0.05, comparison of displayed values.
도 62은 Axl 또는 Mer 특이적 siRNA 처리에 따른 LA-4 세포 내 AKT 단백질의 인산화를 확인한 결과를 나타낸다. LA-4 세포에 Axl 또는 Mer siRNA 또는 대조군 siRNA를 48시간 동안처리하고 400 ng/ml Gas6를 20 시간 처리한 뒤, TGF-β을 72 시간 처리하였다. *P < 0.05, 대조군 대비; +P < 0.05, 표시된 값 비교.Figure 62 shows the results of confirming phosphorylation of AKT protein in LA-4 cells following treatment with AxI or Mer specific siRNA. LA-4 cells were treated with Axl or Mer siRNA or control siRNA for 48 hours, treated with 400 ng / ml Gas6 for 20 hours, and treated with TGF-β for 72 hours. * P < 0.05, compared to control group; + P < 0.05, comparison of displayed values.
도 63은 Gas6 처리에 따른 초대 마우스 AT II 세포 내 EMT 마커 mRNA의 발현 수준을 확인한 결과를 나타낸다. 400 ng/ml Gas6를 20 시간 처리한 뒤, TGF-β을 48 또는 72 시간 처리하였다. *P < 0.05, 대조군 대비; +P < 0.05, 표시된 값 비교.63 shows the results of confirming the expression level of EMT marker mRNA in the mouse AT II cell according to Gas6 treatment. After treatment with 400 ng / ml Gas6 for 20 hours, TGF-beta was treated for 48 or 72 hours. * P < 0.05, compared to control group; + P < 0.05, comparison of displayed values.
도 64은 Gas6 처리에 따른 초대 마우스 AT II 세포 내 EMT 마커 단백질의 발현 수준을 확인한 결과를 나타낸다. 400 ng/ml Gas6를 20 시간 처리한 뒤, TGF-β을 72 시간 처리하였다. *P < 0.05, 대조군 대비; +P < 0.05, 표시된 값 비교.Fig. 64 shows the result of confirming the expression level of EMT marker protein in the mouse AT II cell according to Gas6 treatment. After treatment with 400 ng / ml Gas6 for 20 hours, TGF-beta was treated for 72 hours. * P < 0.05, compared to control group; + P < 0.05, comparison of displayed values.
도 65은 TGF-β및 Gas6 처리에 따른 초대 마우스 AT II 세포 내 Snai1/2, Zeb1/2 및 Twist1 mRNA의 발현 수준을 확인한 결과를 나타낸다. 400 ng/ml Gas6를 20 시간 처리한 뒤 10 ng/ml의 TGF-β를 48 시간 또는 72 시간 처리하였다. *P < 0.05, 대조군 대비; +P < 0.05, 표시된 값 비교.FIG. 65 shows the results of confirming the expression levels of Snai1 / 2, Zeb1 / 2 and Twist1 mRNA in the mouse AT II cells according to TGF-β and Gas6 treatment. After treatment with 400 ng / ml Gas6 for 20 hours, 10 ng / ml of TGF-? Was treated for 48 hours or 72 hours. * P < 0.05, compared to control group; + P < 0.05, comparison of displayed values.
도 66은 Gas6 처리에 따른 초대 마우스 AT II 세포 내 COX-2 mRNA 수준을 확인한 결과를 나타낸다. 400 ng/ml Gas6를 각각 표시된 시간으로 처리하였다. *P < 0.05, 대조군 대비.FIG. 66 shows the results of confirming the levels of COX-2 mRNA in premature mouse AT II cells following treatment with Gas6. 400 ng / ml Gas6 were each treated with the indicated times. * P <0.05 compared to control.
도 67은 NS-398 및 Gas6 처리에 따른 초대 마우스 AT II 세포 내 EMT 마커 mRNA의 발현 수준을 확인한 결과를 나타낸다. 10 μM NS-398를 1시간 처리한 후, 400 ng/ml Gas6를 20 시간 처리한 뒤 10 ng/ml의 TGF-β를 72 시간 처리하였다. *P < 0.05, 대조군 대비; +P < 0.05, 표시된 값 비교.67 shows the results of confirming the expression levels of EMT marker mRNA in primary mouse AT II cells following treatment with NS-398 and Gas6. 10 μM NS-398 was treated for 1 hour, treated with 400 ng / ml Gas6 for 20 hours, and treated with 10 ng / ml of TGF-β for 72 hours. * P < 0.05, compared to control group; + P < 0.05, comparison of displayed values.
도 68는 NS-398 및 Gas6 처리에 따른 초대 마우스 AT II 세포 내 Snai1/2, Zeb1/2 및 Twist1 mRNA의 발현 수준을 확인한 결과를 나타낸다. 10 μM NS-398를 1시간 처리한 후, 400 ng/ml Gas6를 20 시간 처리한 뒤 10 ng/ml의 TGF-β를 72 시간 처리하였다. *P < 0.05, 대조군 대비; +P < 0.05, 표시된 값 비교.68 shows the results of confirming the expression levels of Snai1 / 2, Zeb1 / 2 and Twist1 mRNA in the mouse AT II cells according to NS-398 and Gas6 treatment. 10 μM NS-398 was treated for 1 hour, treated with 400 ng / ml Gas6 for 20 hours, and treated with 10 ng / ml of TGF-β for 72 hours. * P < 0.05, compared to control group; + P < 0.05, comparison of displayed values.
도 69은 AH6, BAY 및 Gas6 처리에 따른 초대 마우스 AT II 세포 내 EMT 마커 mRNA의 발현 수준을 확인한 결과를 나타낸다. 400 ng/ml Gas6를 20 시간 처리한 뒤 48 시간동안 TGF-β만 처리하거나, 10 μM의 EP2 (AH-6809)을 함께 처리하거나, 10 μM의 DP2 (BAY-u3405)을 함께 처리하였다. *P < 0.05, 대조군 대비; +P < 0.05, 표시된 값 비교.69 shows the results of confirming the expression level of EMT marker mRNA in the early mouse AT II cells according to the treatment with AH6, BAY and Gas6. After treatment with 400 ng / ml Gas6 for 20 hours, treatment with TGF-β alone, treatment with 10 μM of EP2 (AH-6809) or treatment with 10 μM of DP2 (BAY-u3405) were performed for 48 hours. * P < 0.05, compared to control group; + P < 0.05, comparison of displayed values.
도 70은 AH6, BAY 및 Gas6 처리에 따른 초대 마우스 AT II 세포 내 Snai1/2, Zeb1/2 및 Twist1 mRNA의 발현 수준을 확인한 결과를 나타낸다. 400 ng/ml Gas6를 20 시간 처리한 뒤 48 시간동안 TGF-β만 처리하거나, 10 μM의 EP2 (AH-6809)을 함께 처리하거나, 10 μM의 DP2 (BAY-u3405)을 함께 처리하였다. *P < 0.05, 대조군 대비; +P < 0.05, 표시된 값 비교.FIG. 70 shows the results of confirming the expression levels of Snai1 / 2, Zeb1 / 2 and Twist1 mRNA in the mouse AT II cells according to treatment with AH6, BAY and Gas6. After treatment with 400 ng / ml Gas6 for 20 hours, treatment with TGF-β alone, treatment with 10 μM of EP2 (AH-6809) or treatment with 10 μM of DP2 (BAY-u3405) were performed for 48 hours. * P < 0.05, compared to control group; + P < 0.05, comparison of displayed values.
도 71는 Gas6 처리에 따른 초대 마우스 AT II 세포 내 HGF mRNA 수준을 확인한 결과를 나타낸다. LA-4 세포에 400 ng/ml Gas6를 표시된 시간으로 처리하였다. *P < 0.05, 대조군 대비.71 shows the result of confirming the level of HGF mRNA in the mouse AT II cell according to Gas6 treatment. LA-4 cells were treated with 400 ng / ml Gas6 for the indicated time. * P <0.05 compared to control.
도 72은 Y-27632 처리에 따른 초대 마우스 AT II 세포 EMT 마커 mRNA 수준을 확인한 결과를 나타낸다. LA-4 세포에 10 mM Y-27632를 1 시간 처리한 뒤 400 ng/ml Gas6를 20 시간 처리한 뒤, 10 ng/ml TGF-β으로 48 시간 자극하였다. *P < 0.05, 대조군 대비; +P < 0.05, 표시된 값 비교.FIG. 72 shows the results of confirming the level of mature AT II cell EMT marker mRNA in the wild-type mouse according to Y-27632 treatment. LA-4 cells were treated with 10 mM Y-27632 for 1 hour, treated with 400 ng / ml Gas6 for 20 hours, and stimulated with 10 ng / ml TGF-β for 48 hours. * P < 0.05, compared to control group; + P < 0.05, comparison of displayed values.
도 73는 Y-27632 처리에 따른 초대 마우스 AT II 세포 Snai1, Zeb1 및 Twist1 mRNA 수준을 확인한 결과를 나타낸다. LA-4 세포에 10 mM Y-27632를 1 시간 처리한 뒤 400 ng/ml Gas6를 20 시간 처리한 뒤, 10 ng/ml TGF-β으로 48 시간 자극하였다. *P < 0.05, 대조군 대비; +P < 0.05, 표시된 값 비교.73 shows the results of confirming the levels of Snai1, Zeb1, and Twist1 mRNA of the premature mouse AT II cells according to Y-27632 treatment. LA-4 cells were treated with 10 mM Y-27632 for 1 hour, treated with 400 ng / ml Gas6 for 20 hours, and stimulated with 10 ng / ml TGF-β for 48 hours. * P < 0.05, compared to control group; + P < 0.05, comparison of displayed values.
도 74는 c-Met 길항제 처리에 따른 초대 마우스 AT II 세포 내 EMT 마커의 mRNA 수준을 확인한 결과를 나타낸다. LA-4 세포에 400 ng/ml Gas6를 20 시간 처리한 뒤, c-Met 길항제인 PHA-665752 250 nM이 존재하거나 존재하지 않는 조건에서 TGF-β을 72 시간 처리하였다. *P < 0.05, 대조군 대비; +P < 0.05, 표시된 값 비교.74 shows the results of confirming mRNA levels of EMT markers in primary mouse AT II cells following c-Met antagonist treatment. LA-4 cells were treated with 400 ng / ml Gas6 for 20 hours and treated with TGF-β for 72 hours in the presence or absence of 250 nM of c-Met antagonist PHA-665752. * P < 0.05, compared to control group; + P < 0.05, comparison of displayed values.
도 75은 c-Met 길항제 처리에 따른 초대 마우스 AT II 세포 내 Snai1, Zeb1 및 Twist1 mRNA 수준을 확인한 결과를 나타낸다. LA-4 세포에 400 ng/ml Gas6를 20 시간 처리한 뒤, c-Met 길항제인 PHA-665752 250 nM이 존재하거나 존재하지 않는 조건에서 TGF-β을 72 시간 처리하였다. *P < 0.05, 대조군 대비; +P < 0.05, 표시된 값 비교.FIG. 75 shows the results of confirming the levels of Snai1, Zeb1 and Twist1 mRNA in the mouse AT II cells according to the c-Met antagonist treatment. LA-4 cells were treated with 400 ng / ml Gas6 for 20 hours and treated with TGF-β for 72 hours in the presence or absence of 250 nM of c-Met antagonist PHA-665752. * P < 0.05, compared to control group; + P < 0.05, comparison of displayed values.
도 76은 TGF-β및 Gas6 처리에 따른 사람 폐 선암종 세포(549 세포) 내 EMT 마커 mRNA의 발현 수준을 확인한 결과를 나타낸다. 400 ng/ml Gas6를 20 시간 처리한 뒤 10 ng/ml의 TGF-β를 48 시간 또는 72 시간 처리하였다. *P < 0.05, 대조군 대비; +P < 0.05, 표시된 값 비교.76 is a graph showing the effect of TGF-beta and Gas6 on human lung adenocarcinoma cells (549 cells) And the expression level of the EMT marker mRNA was confirmed. After treatment with 400 ng / ml Gas6 for 20 hours, 10 ng / ml of TGF-? Was treated for 48 hours or 72 hours. * P < 0.05, compared to control group; + P < 0.05, comparison of displayed values.
도 77은 TGF-β및 Gas6 처리에 따른 사람 폐 선암종 세포(549 세포) 내 EMT 마커 단백질 발현 수준을 확인한 결과를 나타낸다. 400 ng/ml Gas6를 20 시간 처리한 뒤 10 ng/ml의 TGF-β를 48 시간 또는 72 시간 처리하였다. *P < 0.05, 대조군 대비; +P < 0.05, 표시된 값 비교.FIG. 77 shows the results of confirming the level of EMT marker protein expression in human lung adenocarcinoma cells (549 cells) following TGF-β and Gas6 treatment. After treatment with 400 ng / ml Gas6 for 20 hours, 10 ng / ml of TGF-? Was treated for 48 hours or 72 hours. * P < 0.05, compared to control group; + P < 0.05, comparison of displayed values.
도 78는 TGF-β및 Gas6 처리에 따른 사람 폐 선암종 세포(549 세포) 내 Snai1/2, Zeb1/2 및 Twist1 mRNA의 수준을 확인한 결과를 나타낸다. 400 ng/ml Gas6를 20 시간 처리한 뒤 10 ng/ml의 TGF-β를 48 시간 또는 72 시간 처리하였다. *P < 0.05, 대조군 대비; +P < 0.05, 표시된 값 비교.78 shows the results of confirming the levels of Snai1 / 2, Zeb1 / 2 and Twist1 mRNA in human lung adenocarcinoma cells (549 cells) following TGF-beta and Gas6 treatment. After treatment with 400 ng / ml Gas6 for 20 hours, 10 ng / ml of TGF-? Was treated for 48 hours or 72 hours. * P < 0.05, compared to control group; + P < 0.05, comparison of displayed values.
도 79은 생체 내 블레오마이신 및 Gas6 처리에 따른 초대 마우스 AT II 세포의 모양을 위상차 현미경으로 확인한 결과를 나타낸다. FIG. 79 shows the results of in vivo blomycin and Gas6 treatment, in which the shape of primary mouse AT II cells was confirmed by a phase contrast microscope.
도 80은 생체 내 블레오마이신 및 Gas6 처리에 따른 초대 마우스 AT II 세포의 EMT 마커 mRNA의 발현 수준을 확인한 결과를 나타낸다. *P < 0.05, 대조군 대비; +P < 0.05, 표시된 값 비교.80 shows the results of confirming the expression level of EMT marker mRNA in the first mouse AT II cells in vivo by treatment with bleomycin and Gas6. * P < 0.05, compared to control group; + P < 0.05, comparison of displayed values.
도 81는 생체 내 블레오마이신 및 Gas6 처리에 따른 초대 마우스 AT II 세포의 Snai1, Zeb1 및 Twist1 mRNA의 발현 수준을 확인한 결과를 나타낸다. *P < 0.05, 대조군 대비; +P < 0.05, 표시된 값 비교.81 shows the results of confirming the expression levels of Snai1, Zeb1, and Twist1 mRNA of invasive mouse AT II cells in vivo by treatment with bleomycin and Gas6. * P < 0.05, compared to control group; + P < 0.05, comparison of displayed values.
도 82은 생체 내 블레오마이신 및 Gas6 처리에 따른 초대 마우스 AT II 세포의 COX-2 mRNA 수준을 확인한 결과를 나타낸다. *P < 0.05, 대조군 대비; +P < 0.05, 표시된 값 비교.FIG. 82 shows the results of confirming COX-2 mRNA levels in the mouse AT II cells according to in vivo bleomycin and Gas6 treatment. * P < 0.05, compared to control group; + P < 0.05, comparison of displayed values.
도 83는 생체 내 블레오마이신 및 Gas6 처리에 따른 초대 마우스 AT II 세포의 PGE2 또는 PGD2 단백질 수준을 확인한 결과를 나타낸다. *P < 0.05, 대조군 대비; +P < 0.05, 표시된 값 비교.83 shows the results of confirming the level of PGE 2 or PGD 2 protein in primary mouse AT II cells in vivo by treatment with bleomycin and Gas6. * P < 0.05, compared to control group; + P < 0.05, comparison of displayed values.
도 84는 생체 내 블레오마이신 및 Gas6 처리에 따른 초대 마우스 AT II 세포의 HGF mRNA 수준을 확인한 결과를 나타낸다. *P < 0.05, 대조군 대비; +P < 0.05, 표시된 값 비교.84 shows the results of confirming the level of HGF mRNA in primary mouse AT II cells in vivo by treatment with bleomycin and Gas6. * P < 0.05, compared to control group; + P < 0.05, comparison of displayed values.
도 85은 생체 내 블레오마이신 및 Gas6 처리에 따른 초대 마우스 AT II 세포의 HGF 및 TGF-β단백질 수준을 확인한 결과를 나타낸다. *P < 0.05, 대조군 대비; +P < 0.05, 표시된 값 비교.85 shows the results of confirming levels of HGF and TGF-beta protein in primary mouse AT II cells in vivo by treatment with bleomycin and Gas6. * P < 0.05, compared to control group; + P < 0.05, comparison of displayed values.
도 86은 생체 내 블레오마이신 및 Gas6 처리에 따른 14일 째 폐조직에서EMT 마커 및 세포외기질 단백질의 발현 수준을 확인한 결과를 나타낸다. *P < 0.05, 대조군 대비; +P < 0.05, 표시된 값 비교.86 shows the results of confirming the expression levels of EMT markers and extracellular matrix proteins in the lung tissue at day 14 following treatment with in vivo bleomycin and Gas6. * P < 0.05, compared to control group; + P < 0.05, comparison of displayed values.
도 87은 생체 내 블레오마이신 및 Gas6 처리에 따른 21일 째 폐조직에서EMT 마커 및 세포외기질 단백질의 발현 수준을 확인한 결과를 나타낸다. *P < 0.05, 대조군 대비; +P < 0.05, 표시된 값 비교.FIG. 87 shows the results of confirming the expression levels of EMT markers and extracellular matrix proteins in the lung tissue at day 21 following in vivo treatment with bromomycin and Gas6. * P < 0.05, compared to control group; + P < 0.05, comparison of displayed values.
도 88는 생체 내 블레오마이신 및 Gas6 처리에 따른 21일 째 폐조직에서 하이드록시프롤린 수준을 확인한 결과를 나타낸다. *P < 0.05, 대조군 대비; +P < 0.05, 표시된 값 비교.88 shows the results of confirming the level of hydroxyproline in the lung tissue at day 21 following treatment with in vivo bleomycin and Gas6. * P < 0.05, compared to control group; + P < 0.05, comparison of displayed values.
이를 구체적으로 설명하면 다음과 같다. 한편, 본 발명에서 개시된 각각의 설명 및 실시형태는 각각의 다른 설명 및 실시 형태에도 적용될 수 있다. 즉, 본 발명에서 개시된 다양한 요소들의 모든 조합이 본 발명의 범주에 속한다. 또한, 하기 기술된 구체적인 서술에 의하여 본 발명의 범주가 제한된다고 볼 수 없다.This will be described in detail as follows. On the other hand, each description and embodiment disclosed in the present invention can be applied to each other description and embodiment. That is, all combinations of various elements disclosed in the present invention fall within the scope of the present invention. Further, the scope of the present invention is not limited by the detailed description described below.
상기 목적을 달성하기 위한 본 발명의 하나의 양태는, Gas6 (Growth arrest-specific 6) 단백질 또는 Gas6 단백질 수용체의 활성화제를 유효성분으로 포함하는, 섬유증의 예방 또는 치료용 약학적 조성물이다.One aspect of the present invention for achieving the above object is a pharmaceutical composition for preventing or treating fibrosis, which comprises Gas6 (Growth arrest-specific 6) protein or Gas6 protein receptor activator as an active ingredient.
본 발명에서 용어, "섬유증"은 장기 또는 조직에서 과량의 섬유성 결합 조직이 형성되는 것을 의미한다. 이는 장기 또는 조직에서 정상적 구성요소로서의 섬유성 조직과는 구별될 수 있다. 섬유아세포에 의한 피브로넥틴 (fibronectin), 콜라겐 (collagen) 등의 세포 외 기질 (extracellular metrix)의 과량 축적으로 인해, 섬유증은 종국적으로 장기 손상을 일으키는 치명적인 질병으로 이해될 수 있다.The term " fibrosis " in the present invention means that excessive fibrous connective tissue is formed in an organ or tissue. This can be distinguished from fibrous tissue as a normal component in the organ or tissue. Because of the excessive accumulation of extracellular metrix such as fibronectin and collagen by fibroblast, fibrosis can be understood as a fatal disease that eventually leads to organ damage.
본 발명에서 용어, "EMT (epithelial to mesenchymal transition)"는 상피 세포가 간엽 세포로 전환되는 현상을 말하며, 배아 발생, 기관 발달, 상처 치유 및 줄기세포 거동 등과 관련되고, 기관 섬유화 및 암 발달에 병리학적으로 기여하는 것으로 알려져 있다 (J Clin Invest, 2009, 119(6): 1420). 섬유아세포는 (1) 체류 폐 섬유아세포 (resident lung fibroblast)의 증식 및 분화, (2) AEC (alveolar epithelial cell)에서 (근)섬유아세포로 전환되는 EMT, 및 (3) 골수 전구세포의 분화와 같이 다양한 유래를 갖는 것으로 보고되었는데 (Curr Rheumatol Rep, 2006, 8: 145), 현재까지 보고된 결과들은 EMT가 IPF와 같은 섬유증 발병에서 주요 현상임을 시사한다 (Am J Pathol, 2009, 175: 3; J Exp Med, 2011, 208: 1339). 따라서 EMT 억제가 섬유증에 대한 예방 및 치료 효과를 가질 수 있음은 당업자에게 매우 자명한 사항으로 볼 수 있다.The term " epithelial to mesenchymal transition " (EMT) in the present invention refers to a phenomenon in which epithelial cells are transformed into mesenchymal cells, and is associated with embryonic development, organ development, wound healing and stem cell behavior, (J Clin Invest, 2009, 119 (6): 1420). Fibroblasts are classified into (1) proliferation and differentiation of resident lung fibroblasts, (2) EMTs converted from (ave) epithelial cells to (my) fibroblasts, and (3) (Curr Rheumatol Rep, 2006, 8: 145). The results reported so far suggest that EMT is a major event in the development of fibrosis such as IPF (Am J Pathol, 2009, 175: 3; J Exp Med, 2011,208: 1339). It is therefore obvious to those skilled in the art that EMT inhibition can have preventive and therapeutic effects on fibrosis.
본 발명에서 상기 섬유증은 폐, 신장, 간, 심장, 뇌, 혈관, 관절, 장, 피부, 연조직, 골수, 음경, 복막, 랜즈, 근육, 척추, 고환, 난소, 유방, 갑상선, 고막, 췌장, 담낭, 방광 또는 전립선 등 모든 조직에서 나타나는 섬유화 관련 질환을 포함할 수 있다. 구체적으로, 본 발명에서 섬유증은 신체 각 조직에서 발생하는 섬유화에 의해 야기되는 질환, 예컨대 이상 창상 치유, 알콜성 간 손상 유도 간 섬유증, 연결 섬유증, 크론병 (장의 섬유증), 췌장 및 폐의 낭포성 섬유증, 특히 아이들에서 근육간 주사의 합병증으로서 발생할 수 있는 주사 섬유증, 심내막심근 섬유증 또는 심장 섬유증, 이식편대숙주병 (Graft-Versus-Host Disease, GVHD)에 의한 섬유증, 비장의 섬유증, 망막 섬유증을 포함한 눈의 섬유증, 수술 또는 주사 섬유증의 섬유화 합병증, 사구체 신염, 간질 섬유증, 켈로이드 및 비대성 반흔 (피부의 섬유증), 황반 변성, 종격동 섬유증 (종격동의 연조직 섬유증), 국소피부경화증 (morphea), 다초점 섬유소증, 골수 섬유증, 신장성 전신 섬유증 (피부의 섬유증), 결절성 표피 섬유증 (예를 들면, 양성 섬유성 조직구종, 흉막 섬유증, 수술 (예를 들면, 수술 이식)의 결과로서의 섬유증, 증식성 섬유증, 파이프자루 섬유증 (pipestem fibrosis), 후섬유소 섬유증, 진행성 다발 섬유증 (폐의 섬유증 유형, 석탄 노동자 진폐증의 합병증), 오래된 심근 경색 (심장의 섬유증), 췌장 섬유증, 진행성 다발 섬유증, 방사선 섬유증, 신장 섬유증, 만성 신장 질환 관련 또는 이를 유발하는 신장 섬유증, 후복막 섬유증 (후복막의 연조직의 섬유증), 수술 후 반흔화, 경피증/전신 경화증 (피부의 섬유증), 상피세포증, 자궁섬유증, 또는 바이러스성 간염 유도 섬유증일 수 있으나, 이에 제한되지 않는다. In the present invention, the fibrotic syndrome may be selected from the group consisting of lung, kidney, liver, heart, brain, blood vessels, joints, bowel, skin, soft tissues, bone marrow, penis, peritoneum, lance, muscle, spine, testis, ovary, breast, thyroid, Gallbladder, bladder, or prostate. &Lt; / RTI &gt; Specifically, in the present invention, fibrosis is a disease caused by fibrosis occurring in each tissue of the body, such as abnormal wound healing, alcohol-induced hepatic injury induced fibrosis, connective fibrosis, Crohn's disease (intestinal fibrosis), pancreatic and lung cystic Fibrosis, fibrosis caused by Graft-Versus-Host Disease (GVHD), fibrosis of the spleen, fibrosis of the spleen and retinal fibrosis, which may occur as complications of intermuscular injection, especially in children, endocardial myocardial fibrosis or cardiac fibrosis Fibrosis complications of surgery or injection fibrosis, glomerulonephritis, epileptic fibrosis, keloids and hypertrophic scarring (skin fibrosis), macular degeneration, mediastinal fibrosis (soft tissue fibrosis of the mediastinum), morphea, Fibrosis, bone marrow fibrosis, renal systemic fibrosis (skin fibrosis), nodular epidermal fibrosis (e. G., Benign fibrous tissue &lt; Proliferative fibrosis, pipestem fibrosis, postfiber fibrosis, progressive bundle fibrosis (pulmonary fibrosis type, complications of coal worker's pneumoconiosis), pleural fibrosis, fibrosis as a result of surgery (for example, (Fibrosis of the retroperitoneal soft tissue), post-surgical scarring, post-operative fibrosis, fibromyalgia, fibromyalgia, chronic fibrosis, chronic myelogenous leukemia, Scleroderma / systemic sclerosis (skin fibrosis), epithelial cells, uterine fibrosis, or viral hepatitis induced fibrosis.
본 발명에서 폐 섬유증은 특발성 폐 섬유증 (Idiopathic Pulmonary Fibrosis), 비특이성 간질성 폐렴 (Nonspecific Interstitial Pneumonia), 급성간질성 폐렴 (Acute Interstitial Pneumonia), 특발성 기질화 폐렴 (Cryptogenic Organizing Pneumonia), 호흡계기관지염 관련성 간질성 폐질환 (Respiratory Bronchiolitisassociated Interstitial Lung), 박리성 간질성 폐렴 (Desquamative Interstitial Pneumonia), 임파구성 간질성 폐렴 (Lymphoid Interstitial Pneumonia), 간질성 폐 섬유증, 및 미만성 폐 섬유증 등을 포함할 수 있으나, 이에 제한되는 것은 아니다. In the present invention, pulmonary fibrosis is classified as idiopathic pulmonary fibrosis, nonspecific interstitial pneumonia, acute interstitial pneumonia, cryptogenic organizing pneumonia, respiratory bronchitis-related epilepsy But not limited to, Respiratory Bronchiolitisassociated Interstitial Lung, Desquamative Interstitial Pneumonia, Lymphoid Interstitial Pneumonia, Interstitial Pulmonary Fibrosis, and Diffuse Pulmonary Fibrosis. It is not.
본 발명에서 사용된 용어, "치료"는 상기 약학적 조성물의 투여로 섬유증의 증세를 호전시키거나 이롭게 변경하는 모든 행위를 의미하고, "예방"은 상기 약학적 조성물의 투여로 섬유증의 발병을 억제하거나 지연시키는 모든 행위를 의미한다.As used herein, the term " treatment " means any action that improves or alleviates the symptoms of fibrosis with the administration of the pharmaceutical composition, and " prevention " means inhibiting the onset of fibrosis by administration of the pharmaceutical composition Or delaying any action that
본 발명에서는 Gas6 단백질 또는 Gas6 단백질 수용체의 활성화제가 EMT 현상을 억제할 수 있음을 최초로 확인하여, Gas6 단백질 또는 Gas6 단백질 수용체의 활성화제를 섬유증에 대한 예방 및 치료 용도로 사용할 수 있음을 규명하였다.In the present invention, it has been confirmed for the first time that the activator of Gas6 protein or Gas6 protein receptor can inhibit the EMT phenomenon, thereby confirming that the activator of Gas6 protein or Gas6 protein receptor can be used for prevention and treatment of fibrosis.
본 발명에서 용어, "Gas6 (Growth arrest-specific protein 6)"는 분비성 비타민 K-의존성 단백질을 말하며, AXSF 또는 AXLLG 등으로도 명명된다. 본 발명에서 Gas6 단백질은, 예컨대 NCBI accession number (NP_062394.2)의 아미노산 서열 (서열번호 35)을 갖는 것일 수 있으나, 이에 제한되지 않고, 공지된 Gas6 단백질과 상동성을 가지고, 본 발명에서 확인한 것과 동일한 활성을 가지는 한 일부 서열이 결실, 부가, 치환된 것이거나, 혹은 인간 또는 다른 동물 유래의 Gas6 단백질이라 하더라도 모두 본 발명의 범주 내에 포함될 수 있다. 본 발명에서 서열 상동성은 야생형 Gas6 단백질과 70 % 이상, 80 % 이상, 90 % 이상, 95 % 이상, 98 % 이상 또는 99 % 이상의 서열이 일치하는 것일 수 있다. 또한, 야생형 Gas6 단백질에서 활성을 위해 일부 도메인을 절단한 단편, 그리고 단백질의 발현, 분리, 정제, 또는 활성을 증가시키기 위해 공지된 펩타이드를 결합한 융합 단백질 형태의 경우에도 야생형 Gas6 단백질과 동일한 활성을 갖는 한 본 발명의 Gas6 단백질 범주 내에 포함될 수 있다. 본 발명에서 Gas6 단백질은 당업계에 공지된 방법, 예를 들어 펩타이드 합성 또는 형질전환 세포를 이용한 단백질의 생산을 통해 제조될 수 있으나, 이에 제한되지 않는다. 본 발명의 실시예에서는 예시적으로 서열번호 35의 마우스 Gas6 단백질을 합성하여 사용하였다 (R&D Systems; Minneapolis, MN, USA).The term " Growth arrest-specific protein 6 (Gas6) " in the present invention refers to a secretable vitamin K-dependent protein, also called AXSF or AXLLG. In the present invention, the Gas6 protein may have, for example, an amino acid sequence of the NCBI accession number (NP_062394.2) (SEQ ID NO: 35), but it is not limited thereto, and may have homology with known Gas6 protein, Deletion, addition or substitution of a part of the sequences as long as they have the same activity or Gas6 proteins derived from human or other animals can all be included in the scope of the present invention. In the present invention, the sequence homology may be 70% or more, 80% or more, 90% or more, 95% or more, 98% or more or 99% or more of the sequence of the wild type Gas6 protein. In addition, fragments obtained by cleaving some domains for activity in the wild-type Gas6 protein, and fragments having the same activity as the wild-type Gas6 protein in the form of a fusion protein in which known peptides are bound to increase the expression, isolation, purification, May be included within the Gas6 protein category of the present invention. In the present invention, the Gas6 protein can be produced by a method known in the art, for example, through peptide synthesis or production of a protein using transformed cells, but is not limited thereto. In an exemplary embodiment of the present invention, the murine Gas6 protein of SEQ ID NO: 35 was synthesized and used (R & D Systems; Minneapolis, MN, USA).
Gas6는 N-말단 감마-카르복시글루탐산 (Gla) 도메인, 4 개의 EGF (epidermal growth factor)-유사 도메인, 및 SHBG (sex hormone binding globulin)에 상동성을 가지는 큰 C-말단 영역을 포함한다 (Mol Cell Biol, 1993, 13: 4976; Blood Cells Mol Dis, 2006, 36: 373). Gas6는 폐, 심장, 신장, 장, 섬유아세포, 내피세포, 골수세포, 혈관 평활근, 백혈구, 및 뉴런 등에서 발현되며, TAM (Tyro3/Axl/Mer) 수용체 서브패밀리의 공통 리간드로 알려져 있다 (Cell, 1995, 82: 355; Nature Reviews Immunology, 2008, 8: 327). 이에, 본 발명에서 Gas6 단백질 수용체는 이에 제한되는 것은 아니나, AXL 수용체 타이로신 키나아제, Mer 타이로신 키나아제 또는 TYRO3를 포함한다. 상기 수용체들은 모두 두 개의 세포 외 N-말단 면역글로불린-유사 도메인 및 두 개의 피브로넥틴-III-유사 도메인과 이에 뒤 따르는 수용체 C-말단 세포질 끝 부분에 위치한 타이로신 키나아제 도메인을 포함하여 상당한 도메인 유사성을 공유한다. Gas6에 의한 TAM 활성은 세포 생존, 증식, 포식 작용, 분화, 혈소판 기능 및 혈전 안정화를 매개하는 신호를 유도하는 것으로 보고되었다. 또한 최근 연구에서는 대식세포 및 수지상세포에서 Gas6 신호가 TLR에 의해 유도되는 사이토카인 분비를 조절하여 면역 반응을 조절하는 것으로 확인되었다. 나아가 최근, 본 발명자들은 대식세포가 Gas6에 의해 리프로그래밍되어 대식세포에서 Mer/RhoA/Akt/MAP 키나아제 신호전달경로에 의해 유도되는 HGF의 주변분비 (paracrine) 역할을 통해 상피세포 증식 및 상처 회복을 촉진할 수 있음을 확인하였으나, 상피세포 항상성에 대한 Gas6의 직접적인 역할은 여전히 알려지지 않았다.Gas6 contains a large C-terminal region that is homologous to the N-terminal gamma-carboxyglutamic acid (Gla) domain, four epidermal growth factor (EGF) -like domains, and sex hormone binding globulin (SHBG) Biol, 1993, 13: 4976; Blood Cells Mol Dis, 2006, 36: 373). Gas6 is expressed in the lung, heart, kidney, intestine, fibroblasts, endothelial cells, bone marrow cells, vascular smooth muscle, white blood cells and neurons and is known as a common ligand of the TAM (Tyro3 / Axl / Mer) 1995, 82: 355; Nature Reviews Immunology, 2008, 8: 327). Thus, the Gas6 protein receptor in the present invention includes, but is not limited to, AXL receptor tyrosine kinase, Mer tyrosine kinase, or TYRO3. These receptors share considerable domain similarity, including both extracellular N-terminal immunoglobulin-like domains and two fibronectin-III-like domains and the tyrosine kinase domain located at the following C-terminal cytoplasmic end of the receptor . TAM activity by Gas6 has been reported to induce signals that mediate cell survival, proliferation, predation, differentiation, platelet function and thrombolytic stabilization. Recent studies have also shown that Gas6 signaling in macrophages and dendritic cells regulates the immune response by regulating TLR-induced cytokine secretion. Furthermore, recently, we have found that macrophages are reprogrammed by Gas6 to play a role in the paracrine of HGF induced by Mer / RhoA / Akt / MAP kinase signaling pathway in macrophages, leading to epithelial cell proliferation and wound repair . However, the direct role of Gas6 on epithelial cell homeostasis remains unknown.
본 발명에서 Gas6 단백질 수용체의 활성화제는 Gas6 단백질 수용체를 활성화시켜 그 하위 신호전달경로를 활성화시킬 수 있는 물질을 의미하며, 특히 본 발명의 목적상 PEG2, PGD2 또는 HGF를 생산, 분비할 수 있는 신호전달경로에 대한 활성화 물질을 말한다. 예를 들어, 상기 Gas6 단백질 수용체의 활성화제는 Protein S (Pros1), Tubby and tubby-like protein 1, 또는 Galectin3일 수 있으나, 이에 제한되는 것은 아니다.Activators of Gas6 protein receptor in the present invention is to enable the Gas6 protein receptor means a substance capable of activating the sub-transmission path, particularly in the purposes of PEG2, PGD 2 or HGF of the present invention to produce, secrete Quot; refers to the active material for the signal transduction pathway. For example, the activator of the Gas6 protein receptor may be, but is not limited to, Protein S (Pros1), Tubby and tubby-like protein 1, or Galectin3.
본 발명의 구체적 일 실시예에서는 폐포 및 신장 상피세포에서 TGF-β으로 유발되는 EMT를 Gas6가 억제하는 효과를 세포의 형태와 EMT 마커의 mRNA 및 단백질 수준 변화를 통해 직접적으로 확인하였다 (도 1 내지 도 4). 다른 일 실시예에서는 이러한 Gas6의 항-EMT 활성은 ERK 및 Akt 경로를 포함하는 Smad 독립적 TGF-β신호를 차단함으로써 나타난다는 것을 확인하였다 (도 5 내지 도 10). 또한, Gas6 처리에 의해 PGE2, PGD2 및 HGF 분비가 유도되고, 주로 EP2, DP2 및 c-Met 수용체를 통해 항-EMT 효과를 나타낸다는 것을 확인하였다 (도 11 내지 도 53). Gas6는 세포막에 분포하는 Axl 또는 Mer 수용체 티로신 키네이즈와 작용하여 COX-2 유래 PGE2, PGD2 분비 와 RhoA 의존적인 HGF 분비를 통해 TGF-β신호를 차단함으로써 항-EMT 효과가 나타남을 확인하였다(도 5 내지 도 62). 특히 마우스 초대 폐포상피세포에서도, 마우스 폐포상피세포에서 나타났듯이, PGE2, PGD2 및 HGF 신호 의존적인 Gas6의 항-EMT 효과를 확인하였다(도 63 내지 도 75). 더욱이 인간의 폐선종세포(A549 세포)에서 인간 Gas6 재조합 단백질을 투여시 유사하게 항-EMT 효과가 확인되었다(도 76 내지 도 78). 다른 일 실시예에서는 폐 섬유증이 유발된 동물모델에서 Gas6 투여에 따라 폐 섬유증이 개선되는 것을 확인하였다(도 79 내지 88).In a specific example of the present invention, Gas6-inhibiting effect of EMT induced by TGF-beta in alveolar and renal epithelial cells was directly confirmed through changes in cell morphology and EMT marker mRNA and protein levels 4). In another embodiment this anti-EMT activity of Gas6 was shown to be blocked by blocking Smad-independent TGF-ss signals including ERK and Akt pathways (Figures 5 to 10). In addition, it was confirmed that PGE 2 , PGD 2 and HGF secretion were induced by treatment with Gas6, and showed an anti-EMT effect mainly through EP2, DP2 and c-Met receptors (FIGS. 11 to 53). Gas6 acts on Axl or Mer receptor tyrosine kinase distributed in the cell membrane to block the TGF-β signal through COX-2-derived PGE 2 , PGD 2 secretion and RhoA-dependent HGF secretion, thus confirming anti-EMT effect (Figs. 5 to 62). In particular, mouse invitation in alveolar epithelial cells was confirmed the effect of the anti--EMT natdeut receive from mouse alveolar epithelial cells, PGE 2, PGD 2 and HGF-dependent signal Gas6 (Fig. 63 to Fig. 75). Furthermore, the anti-EMT effect was similarly observed when human Gas6 recombinant protein was administered to human lung carcinoma cells (A549 cells) (Figs. 76 to 78). In another embodiment, lung fibrosis was improved by administration of Gas6 in an animal model in which pulmonary fibrosis was induced (Figs. 79 to 88).
따라서 본 발명의 조성물은 세포에서 PEG2, PGD2 또는 HGF를 생산, 분비하여 자가분비 방식으로 EMT (epithelial to mesenchymal transition)를 억제할 수 있어, 섬유증의 예방 및 치료에 우수한 효과를 가지므로, 섬유증의 예방 또는 치료 용도로 매우 유용하게 사용될 수 있다.Thus compositions of the invention produce PEG2, PGD 2 or HGF in the cell, and secretion can be suppressed EMT (epithelial to mesenchymal transition) by autocrine manner, because of the excellent effects in the prevention and treatment of fibrosis, the fibrosis Can be very useful for prevention or treatment.
또한, 본 발명의 약학적 조성물은 약학적 조성물의 제조에 통상적으로 사용하는 적절한 담체, 부형제 또는 희석제를 추가로 포함할 수 있다. 약학적으로 허용 가능한 담체를 포함하는 조성물은 경구 또는 비경구의 여러 가지 제형일 수 있다. 제제화할 경우에는 보통 사용하는 충진제, 증량제, 결합제, 습윤제, 붕해제, 계면활성제 등의 희석제 또는 부형제를 사용하여 조제될 수 있다. 경구 투여를 위한 고형제제에는 정제환제, 산제, 과립제, 캡슐제 등이 포함될 수 있으며, 이러한 고형제제는 하나 이상의 화합물에 적어도 하나 이상의 부형제, 예를 들면, 전분, 탄산칼슘, 수크로오스 (sucrose) 또는 락토오스 (lactose), 젤라틴 등을 섞어 조제될 수 있다. 또한, 단순한 부형제 이외에 스테아린산 마그네슘, 탈크 등과 같은 윤활제들도 사용될 수 있다. 경구 투여를 위한 액상제제로는 현탁제, 내용액제, 유제, 시럽제 등이 해당되는데 흔히 사용되는 단순 희석제인 물, 리퀴드 파라핀 이외에 여러 가지 부형제, 예를 들면 습윤제, 감미제, 방향제, 보존제 등이 포함될 수 있다. 비경구투여를 위한 제제에는 멸균된 수용액, 비수성용제, 현탁제, 유제, 동결건조제제, 좌제가 포함될 수 있다. 비수성용제, 현탁용제로는 프로필렌글리콜 (propylene glycol), 폴리에틸렌 글리콜, 올리브 오일과 같은 식물성 기름, 에틸올레이트와 같은 주사 가능한 에스테로 등이 사용될 수 있다. 좌제의 기제로는 위텝솔 (witepsol), 마크로골, 트윈 (tween) 61, 카카오지, 라우린지, 글리세로젤라틴 등이 사용될 수 있다.In addition, the pharmaceutical composition of the present invention may further comprise an appropriate carrier, excipient or diluent conventionally used in the production of a pharmaceutical composition. Compositions comprising a pharmaceutically acceptable carrier can be of various oral or parenteral formulations. In the case of formulation, it can be prepared using diluents or excipients such as fillers, extenders, binders, humectants, disintegrants, surfactants and the like which are usually used. Solid formulations for oral administration may include tablet pills, powders, granules, capsules and the like, which may contain one or more excipients, such as starch, calcium carbonate, sucrose or lactose, lactose, gelatin, and the like. In addition to simple excipients, lubricants such as magnesium stearate, talc, and the like may also be used. Liquid preparations for oral administration include suspensions, solutions, emulsions, syrups and the like. Various excipients such as wetting agents, sweeteners, fragrances, preservatives and the like may be included in addition to water and liquid paraffin, which are simple diluents commonly used. have. Formulations for parenteral administration may include sterile aqueous solutions, non-aqueous solutions, suspensions, emulsions, freeze-dried preparations, and suppositories. Examples of the non-aqueous solvent and the suspending agent include propylene glycol, polyethylene glycol, vegetable oil such as olive oil, and injectable ester such as ethyl oleate. Examples of the suppository base include witepsol, macrogol, tween 61, cacao paper, laurin, glycerogelatin and the like.
또한, 본 발명의 약학적 조성물은 이에 제한되지는 않으나, 정제, 환제, 산제, 과립제, 캡슐제, 현탁제, 내용액제, 유제, 시럽제, 멸균된 수용액, 비수성용제, 현탁제, 유제, 동결건조제제 및 좌제로 이루어진 군으로부터 선택되는 어느 하나의 제형을 가질 수 있다.The pharmaceutical composition of the present invention may also be in the form of tablets, pills, powders, granules, capsules, suspensions, solutions, emulsions, syrups, sterilized aqueous solutions, nonaqueous solutions, suspensions, emulsions, A pharmaceutical preparation and a suppository.
상기 조성물은 100 ㎍/kg 내지 500 ㎍/kg의 용량으로 투여되는 것일 수 있으나, 이에 제한되지 않는다. 본 발명의 조성물은 이미 알려진 섬유증 치료제와 다른 시간에 또는 동시에 병용하여 투여될 수 있고, 또는 이미 알려진 공지의 섬유증 치료 방법이 함께 적용될 수 있다. 또한 상기 조성물은 그리고 단일 또는 다중 투여될 수 있다. 상기 요소를 모두 고려하여 부작용 없이 최소한의 양으로 최대 효과를 얻을 수 있는 양을 투여하는 것이 중요하며, 이는 당업자에 의해 용이하게 결정될 수 있다.The composition may be administered in a dose of 100 占 퐂 / kg to 500 占 퐂 / kg, but is not limited thereto. The composition of the present invention can be administered at a different time or at the same time as the already known fibrosis treatment agent or can be applied together with known fibrosis treatment methods already known. The composition may also be administered singly or multiply. It is important to take into account all of the above factors and to administer the amount in which the maximum effect can be obtained in a minimal amount without side effects, which can be easily determined by a person skilled in the art.
본 발명에서 용어, "투여"는 어떠한 적절한 방법으로 대상에게 본 발명의 약학적 조성물을 도입하는 것을 말하며, 투여 경로는 목적 조직에 도달할 수 있는 한 경구 또는 비경구의 다양한 경로를 통하여 투여될 수 있다.The term " administration " as used herein refers to the introduction of a pharmaceutical composition of the present invention to a subject by any suitable method, and the administration route can be administered through various routes of oral or parenteral administration as long as it can reach the target tissue .
상기 약학적 조성물은 목적 또는 필요에 따라 당업계에서 사용되는 통상적인 방법, 투여 경로, 투여량에 따라 적절하게 개체에 투여될 수 있다. 투여 경로의 예로는 경구, 비경구, 피하, 복강 내, 폐 내, 및 비강 내로 투여될 수 있으며, 비경구 주입에는 근육 내, 정맥 내, 동맥 내, 복강 내 또는 피하투여가 포함된다. 또한 당업계에 공지된 방법에 따라 적절한 투여량 및 투여 횟수가 선택될 수 있으며, 실제로 투여되는 본 발명의 약학적 조성물의 양 및 투여 횟수는 치료하고자 하는 증상의 종류, 투여 경로, 성별, 건강 상태, 식이, 개체의 연령 및 체중, 및 질환의 중증도와 같은 다양한 인자에 의해 적절하게 결정될 수 있다.The pharmaceutical composition may be appropriately administered to a subject according to the purpose or necessity, depending on the conventional method, route of administration and dosage used in the art. Examples of routes of administration include oral, parenteral, subcutaneous, intraperitoneal, intrapulmonary, and intranasal routes, and parenteral injection includes intramuscular, intravenous, intraarterial, intraperitoneal, or subcutaneous administration. The appropriate dosage and the frequency of administration may be selected according to methods known in the art, and the amount and the frequency of administration of the pharmaceutical composition of the present invention to be actually administered depends on the type of symptom to be treated, route of administration, sex, , Diet, age and weight of the individual, and the severity of the disease.
본 발명에서의 용어 "약학적으로 유효한 양"은 의학적 용도에 적용 가능한 합리적인 수혜/위험 비율로 혈관 투과성 증가를 억제 또는 완화하기에 충분한 양을 의미하며, 유효 용량 수준은 개체 종류 및 중증도, 연령, 성별, 약물의 활성, 약물에 대한 민감도, 투여 시간, 투여 경로 및 배출 비율, 치료기간, 동시 사용되는 약물을 포함한 요소 및 기타 의학 분야에 잘 알려진 요소에 따라 결정될 수 있다. The term " pharmaceutically effective amount " as used herein means an amount sufficient to inhibit or alleviate an increase in vascular permeability at a reasonable benefit / risk ratio applicable to medical use, and effective dosage levels will vary depending on the species and severity, Sex, the activity of the drug, the sensitivity to the drug, the time of administration, the route of administration and the rate of excretion, the duration of the treatment, factors including co-administered drugs, and other factors well known in the medical arts.
본 발명에서 용어, "개체"는 본 발명의 섬유증 질환을 보유하거나 또는 질환이 발병한, 인간을 포함한 모든 동물을 의미한다. 본 발명의 약학적 조성물을 개체에 투여함으로써, 섬유증을 예방 또는 치료할 수 있다. In the present invention, the term " individual " means all animals including humans, which have the fibrosis disease or the disease of the present invention. By administering the pharmaceutical composition of the present invention to an individual, fibrosis can be prevented or treated.
본 발명의 다른 하나의 양태는, Gas6 단백질 또는 Gas6 단백질의 수용체 활성화제를 개체에 투여하는 단계를 포함하는, 섬유증의 예방 또는 치료 방법이다.Another aspect of the present invention is a method of preventing or treating fibrosis, comprising administering to a subject a receptor activator of Gas6 protein or Gas6 protein.
Gas6 단백질, 이의 수용체 활성화제의 섬유증 예방 또는 치료 용도는 상기 설명한 바와 같다.The use of Gas6 protein, its receptor activator, for the prevention or treatment of fibrosis has been described above.
이하 본 발명을 실시예를 통하여 보다 상세하게 설명한다. 그러나 이들 실시예는 본 발명을 예시적으로 설명하기 위한 것으로 본 발명의 범위가 이들 실시예에 국한되는 것은 아니다.Hereinafter, the present invention will be described in more detail with reference to examples. However, these examples are intended to illustrate the present invention, and the scope of the present invention is not limited to these examples.
실시예 1: 세포주 및 배양 Example 1: Cell line and culture
LA-4 및 HEK-293 세포는 ATCC로부터 구입하였다. LA-4 세포는 37 ℃5 % CO2 조건에서 열 처리로 불활성화된 15 % FBS (fetal bovine serum)를 함유하는 F12K 배지 (스위스 Lonza 사)에서 배양하였다. HEK-293 세포는 37 ℃5 % CO2 조건에서 10 % FBS, 2 mM L-글루타민, 100 U/ml 페니실린 및 100 mg/ml 스트렙토마이신을 함유하는 DMEM (Dulbecco's modified Eagle's medium; 미국 Media Tech 사)에서 배양하였다.LA-4 and HEK-293 cells were purchased from ATCC. LA-4 cells were cultured in F12K medium (Lonza, Switzerland) containing 15% FBS (fetal bovine serum) inactivated by heat treatment at 37 ° C and 5% CO 2 . HEK-293 cells 37 5% 10% in CO 2 conditions, FBS, 2 mM L- glutamine, 100 U / ml penicillin and 100 mg / ml streptomycin, DMEM containing (Dulbecco's modified Eagle's medium; US Media Tech, Inc.) Lt; / RTI &gt;
실시예 2: 초대 세포의 분리 및 배양Example 2: Isolation and culture of early cells
초대 마우스 AT II (alveolar type II) 상피 세포를 BALB/c 마우스로부터 분리, 정제하였다. The primary mouse AT II (alveolar type II) epithelial cells were isolated and purified from BALB / c mice.
폐 동맥에 0.9% 식염수를 주입하여 폐 혈액을 제거하였다. 1 ml의 식염수로 폐를 세척한 후, 디스파제(dispase) 100 unit을 마우스 폐에 주입한 다음, 실온에서 45분 동안 배양하였다. 그 다음, 기계적 수단으로 큰 기관지에서 폐를 분리하고, 분리된 폐 조직을 DNase I 0.01%를 함유한 DMEM 배지에 37 ℃에서 10 분간 배양하였다. 세포를 여과하고, 원심 분리한 후, 대식세포와 섬유아세포를 각각 제거하기 위해 37 ℃에서 1 시간 동안 마우스 IgG 0.75 mg/ml으로 코팅된 세포 배양 페트리 디쉬에 순차적 플레이팅으로 재현탁하였다. 최종 세포 분리물을 15 mM HEPES, 0.8 mM CaCl2, 0.25 % BSA, 5 mg/ml 인슐린, 5 mg/ml 트랜스페린, 5 ng/ml 소듐 셀레나이트 및 2 % 마우스 혈청을 함유하는 Ham 's F12 배지가 첨가된 제1형 콜라겐 코팅 35-mm 배양 접시에 접종하였다.Pulmonary artery was infused with 0.9% saline to remove pulmonary blood. After washing the lungs with 1 ml of saline, 100 units of dispase were injected into the mouse lungs and then incubated at room temperature for 45 minutes. Then, the lungs were separated from the large bronchus by mechanical means, and the separated lung tissues were cultured in DMEM medium containing 0.01% of DNase I at 37 ° C for 10 minutes. The cells were filtered, centrifuged and resuspended in a cell culture Petri dish coated with mouse IgG 0.75 mg / ml for 1 hour at 37 ° C to remove macrophages and fibroblasts, respectively, by sequential plating. The final cell isolates 15 mM HEPES, 0.8 mM CaCl 2 , 0.25% BSA, 5 mg / ml insulin, 5 mg / ml transferrin, Ham 's F12 medium containing 5 ng / ml sodium selenite and 2% mouse serum Lt; RTI ID = 0.0 &gt; 35-mm &lt; / RTI &gt;
pro-SP-C 면역형광 염색으로 평가한 AT II 세포의 순도는 90 % 이상이었다.The purity of AT II cells evaluated by pro-SP-C immunofluorescence staining was over 90%.
실시예 3: 상피세포의 인큐베이션Example 3: Incubation of epithelial cells
LA-4 및 HEK-293 세포는 6-웰 배양 접시에 접종 (2 x 105 개의 세포/웰)하고 10 % FBS를 함유하는 RPMI 1640 또는 DMEM 200 ㎕에서 밤새 배양하였다. 초대 AT II 세포는 제1형 콜라겐 코팅 배양 접시 (1 x 106 개의 세포/웰)에 접종하고 48 시간 동안 배양하였다. 10 ng/ml의 TGF-β(R&D Systems Inc)이 존재하거나 존재하지 않는 조건에서 400 ng/ml의 Gas6로 20 시간 동안 세포를 처리하였다. 일부 실험에서, COX-2 억제를 위해 10 μM NS-398, Rho 키나아제 억제를 위해 30 mM Y-27632를 사용하였다. 각 억제제는 Gas6를 처리하기 1 시간 전에 첨가되었다. 또한, EP2, EP4, DP1, DP2 또는 c-Met에 대한 길항제로 각각 AH-6809, AH-23848, BW-A868C, BAY-u3405 10 mM 또는 PHA-665752 250 nM을 사용하였다. 각 수용체 길항제는 TGF-β처리 1 시간 전에 첨가되었다. LA-4 and HEK-293 cells were inoculated (2 x 10 5 cells / well) in 6-well culture dishes and incubated overnight in 200 μl of RPMI 1640 or DMEM containing 10% FBS. The primary AT II cells were inoculated into type 1 collagen-coated culture dishes (1 x 10 6 cells / well) and cultured for 48 hours. Cells were treated with 400 ng / ml Gas6 for 20 hours in the presence or absence of 10 ng / ml TGF-beta (R & D Systems Inc). In some experiments, 10 μM NS-398 was used for COX-2 inhibition and 30 mM Y-27632 was used for Rho kinase inhibition. Each inhibitor was added 1 hour before the treatment of Gas6. AH-6809, AH-23848, BW-A868C, BAY-u3405 10 mM or PHA-665752 250 nM were used as antagonists for EP2, EP4, DP1, DP2 or c-Met, respectively. Each receptor antagonist was added 1 hour before TGF-beta treatment.
실시예 4: 일시적 형질주입 (transient transfection) Example 4: Transient transfection &lt; RTI ID = 0.0 &gt;
5 μl의 siRNA 형질주입 시약 (Genlantis 사)를 사용하여 COX-2 또는 RhoA 특이적 siRNA, 또는 대조군 siRNA 1 μg/ml를 LA-4 세포에 일시적으로 형질주입하였다. Axl 또는 Mer 특이적 siRNA는 75 nM을 사용하였다. 사용된 siRNA 서열을 표 1에 나타내었다.1 μg / ml of COX-2 or RhoA-specific siRNA or control siRNA was transiently transfected into LA-4 cells using 5 μl siRNA transfection reagent (Genlantis). 75 nM of Axl or Mer specific siRNA was used. The siRNA sequences used are shown in Table 1.
[표 1][Table 1]
Figure PCTKR2018007128-appb-I000001
Figure PCTKR2018007128-appb-I000001
실험 전에 세포를 COX-2 siRNA를 함유하는 무혈청 배지에서 6 시간, RhoA siRNA를 함유하는 무혈청 배지에서 24 시간, 또는 Axl 또는 Mer siRNA를 함유하는 무혈청 배지에서 48 시간 동안 배양하였다. 사용된 모든 siRNA에 대하여 세포 생존성에 유의적 효과는 관찰되지 않았다.Before the experiment, cells were cultured in serum-free medium containing COX-2 siRNA for 6 hours, in serum-free medium containing RhoA siRNA for 24 hours, or in serum-free medium containing Axl or Mer siRNA for 48 hours. No significant effect on cell survival was observed for all siRNA used.
실시예 5: 면역블랏 분석 Example 5: Immunoblot analysis
상피성 마커 및 간엽성 마커 발현을 확인하기 위해, 0.5 % 트리톤 X-100을 함유하는 용해 완충액에서 세포를 용해시키고 10 % SDS-PAGE 젤에 로딩한 뒤 니트로셀룰로스 멤브레인으로 옮겼다. 상기 멤브레인을 상온에서 3 % BSA 또는 5 % 탈지유 함유 TBS (Tris-buffered saline)로 블록킹한 뒤, 각각의 항-마우스 일차 항체와 함께 상온에서 인큐베이션하고, 항-마우스 HRP-결합 이차 항체를 부착시켰다. 단백질 밴드는 향상된 화학발광법 (enhanced chemiluminescence)을 이용하여 확인하였다. To confirm epithelial marker and hepatic lobe marker expression, the cells were lysed in lysis buffer containing 0.5% Triton X-100 and loaded onto 10% SDS-PAGE gels and transferred to nitrocellulose membranes. The membrane was blocked with TBS (Tris-buffered saline) containing 3% BSA or 5% skim milk at room temperature and then incubated with each anti-mouse primary antibody at room temperature to attach anti-mouse HRP-conjugated secondary antibody . Protein bands were identified using enhanced chemiluminescence.
실시예 6: ELISA (Enzyme-linked immunosorbent assay) 측정Example 6 Measurement of Enzyme-Linked Immunosorbent Assay (ELISA)
세포 배양 상층액을 회수하여 ELISA를 수행하였다. PGE2 PGD2, HGF 및 활성 형태의 TGF-β 농도 수준을 ELISA 키트 (R&D Systems, Minneapolis, MN)로 측정하였다.Cell culture supernatants were collected and subjected to ELISA. The levels of PGE 2 PGD 2 , HGF and TGF-β in active form were measured by ELISA kit (R & D Systems, Minneapolis, MN).
실시예 7: qPCR (Quantitative real-time PCR) Example 7: qPCR (Quantitative real-time PCR)
StepOnePlus 시스템 (Applied Biosystems, Life Technologies, Carlsbad, CA, USA)에서 리얼타임 qPCR로 유전자 발현을 분석하였다. 각각의 qPCR 분석에서, 총 50 ng의 cDNA를 사용하였다. Primer Express 소프트웨어를 사용하여 PCR-기반 증폭에 대한 프라이머 세트를 설계하였다. 본 발명에서 사용된 프라이머는 표 2에 나타내었다. Gene expression was analyzed by real-time qPCR in a StepOnePlus system (Applied Biosystems, Life Technologies, Carlsbad, Calif., USA). For each qPCR assay, a total of 50 ng of cDNA was used. Primer Express software was used to design primer sets for PCR-based amplification. The primers used in the present invention are shown in Table 2.
[표 2][Table 2]
Figure PCTKR2018007128-appb-I000002
Figure PCTKR2018007128-appb-I000002
cDNA는 HPRT (hypoxanthine-guanine phosphoribosyltransferase)의 양으로 표준화하였고, 대조군에 대비 배수 (fold-change)로 나타내었다.cDNA was normalized to the amount of HPRT (hypoxanthine-guanine phosphoribosyltransferase) and expressed as fold-change in the control.
실시예 8: RhoA 활성 분석Example 8: RhoA activity assay
ELISA-based RhoA activation assay Biochem Kit (G-LISA; Cytoskeleton)를 사용하여 LA-4 세포 용해물에서 RhoA 활성을 측정하였다. RhoA의 Rhotekin 결합 도메인으로 30 분간 코팅한 RhoA-GTP 친화성 플레이트에 세포 용해물을 첨가하였다. RhoA의 활성 GTP-결합 형태는 간접 면역 검출법을 사용하여 측정하였고 마이크로플레이트 분광 광도계로 490 nm에서 색도 반응을 측정하였다.RhoA activity was measured in LA-4 cell lysate using ELISA-based RhoA activation assay Biochem Kit (G-LISA; Cytoskeleton). Cell lysates were added to RhoA-GTP affinity plates coated with Rhotekin binding domain of RhoA for 30 min. The active GTP-binding form of RhoA was measured using indirect immunoassay and the chromaticity response was measured at 490 nm with a microplate spectrophotometer.
실시예 9: 히드록시프롤린 측정Example 9: Measurement of hydroxyproline
히드록시프롤린 함량은 히드록시프롤린 분석 키트(중국 난징 Jiancheng Bioengineering 사)를 제조사의 지시에 따라 사용하여 측정하였다.The content of hydroxyproline was measured using a hydroxyproline assay kit (Nanjing Jiancheng Bioengineering Co., Ltd.) according to the manufacturer's instructions.
실시예 10: 동물 실험 Example 10: Animal experiment
모든 실험에 20 - 25 g의 특정 병원체 부재 수컷 C57BL/6 마우스(Orient Bio, 성남, 한국)를 사용하였다. 마우스 인두 흡인으로 블레오마이신(5 U/kg 체중 30 μl)을 함유한 시험 용액을 투여하였다. 블레오마이신 투여 하루 전에 식염수 단독 또는 Gas6 (50 μg/kg, i.p.)를 투여하였다. 첫 블레오마이신 투약 후 억제제는 하루에 한번 투여 되었으며, 블레오마이신 투여 14일 및 21일 째에 마우스를 안락사시켰다.Male C57BL / 6 mice (Orient Bio, Seongnam, Korea) were used for all experiments with 20-25 g of specific pathogen. A test solution containing bleomycin (30 μl of 5 U / kg body weight) was administered by aspiration of the mouse pharynx. Saline alone or Gas6 (50 μg / kg, i.p.) was administered one day before the administration of bleomycin. The inhibitors after the first bleomycin dosing were administered once a day and the mice were euthanized on days 14 and 21 of bleomycin administration.
실시예 11: 통계적 분석Example 11: Statistical analysis
평균 값은 ± SEM 으로 표시하였다. 다양한 분석을 위해 분산 분석(ANOVA)을 적용하고, Tukey's post hoc test를 적용하였다. 두 샘플의 표본 평균을 비교하기 위해 Student's t-tests을 사용하였다. 0.05 이하의 p 값을 통계적으로 유의하다고 간주하였다. 모든 데이터는 JMP 소프트웨어 (SAS Institute, Cary, NC)를 사용하여 분석하였다.Mean values were expressed as ± SEM. Analysis of variance (ANOVA) was applied for various analyzes and Tukey's post hoc test was applied. Student's t-tests were used to compare the sample mean of the two samples. A p value of 0.05 or less was considered statistically significant. All data were analyzed using JMP software (SAS Institute, Cary, NC).
실험예 1: 폐 및 신장 상피세포에서 TGF-β유도 EMT에 대한 Gas6의 억제 효과Experimental Example 1: Inhibitory effect of Gas6 on TGF-beta induced EMT in lung and kidney epithelial cells
TGF-β신호는 EMT와 섬유증 발달에서 중요한 역할을 하는 것으로 알려져 있다 (Am J Physiol Lung Cell Mol Physiol, 2007, 293: L525). 본 발명자들은 Gas6 처리가 마우스 폐포 제2형 유사 폐 상피세포 (LA-4)에서 TGF-β유도 EMT 과정을 조절하는지 확인하고자 하였다. TGF-β signaling has been shown to play an important role in EMT and fibrosis development (Am J Physiol Lung Cell Mol Physiol, 2007, 293: L525). We sought to determine if Gas6 treatment modulates the TGF-beta induced EMT process in mouse alveolar type 2-like lung lung epithelial cells (LA-4).
먼저, 10 ng/ml의 TGF-β처리로 48 시간 또는 72 시간 자극을 준 경우 LA-4 세포가 방추-유사 형태 (spindle-like morphology)로 변환되는 것을 확인하였으나, TGF-β과 함께 400 ng/ml의 Gas6를 처리한 경우 LA-4 세포의 방추-유사 형태 변환을 막고 상피세포의 형태를 나타내는 것을 확인하였다 (도 1). 또한 10 ng/ml의 TGF-β으로 48 또는 72 시간 자극을 준 경우, 부착 접합점 (adherens junction) 단백질인 E-cadherin 발현이 감소되고, 근섬유아세포 분화 마커인 N-cadherin과 α-SMA (α-smooth muscle actin)의 발현이 증가되었으나, TGF-β과 함께 400 ng/ml의 Gas6를 처리한 경우 이러한 EMT 마커 발현 양상이 억제되는 것을 단백질 및 mRNA 수준에서 확인하였다 (도 2 및 도 3). 상기 데이터는 Gas6 처리 후 합성, 분비된 생활성 매개체들이 LA-4 세포에서 자가분비 (autocrine) 방식으로 TGF-β신호를 막을 수 있다는 가능성을 시사한다. 또한 본 HEK-293 인간 신장 상피세포 (도 4)에서도 Gas6 처리를 통해 TGF-β유도 EMT 마커 변화가 억제되는 것을 단백질 수준에서 확인하였다.First, it was confirmed that LA-4 cells were converted into spindle-like morphology when stimulated with TGF-β at 10 ng / ml for 48 hours or 72 hours. However, when TGF-β was stimulated with 400 ng / ml of Gas6 inhibited spindle-like morphological transformation of LA-4 cells and showed morphology of epithelial cells (Fig. 1). In addition, when stimulated with TGF-β at 10 ng / ml for 48 or 72 hours, expression of E-cadherin, an adherens junction protein, was decreased and N-cadherin and α-SMA (α- Smooth muscle actin expression was increased, but the expression of EMT markers was suppressed at the level of protein and mRNA when treated with 400 ng / ml of Gas6 together with TGF-β (FIGS. 2 and 3). This data suggests that synthetic and secreted mediators after Gas6 treatment can block TGF-β signaling in an autocrine manner in LA-4 cells. In addition, the present HEK-293 human kidney epithelial cell (FIG. 4) also confirmed the inhibition of TGF-β induced EMT marker change by Gas6 treatment at the protein level.
실험예 2: LA-4 세포에서 TGF-β에 의해 유도되는 Smad-독립적 신호 및 EMT 조절 전사인자 발현에 대한 Gas6 처리의 효과Experimental Example 2: Effect of Gas6 treatment on TGF-β-induced Smad-independent signal and EMT regulatory transcription factor expression in LA-4 cells
EMT 유도에서, 활성화된 Smad 또는 비-Smad 신호는 Snai, Zeb 및 Basic helix-loop-helix 전사인자 패밀리의 전사 조절을 매개하여, 상피성 마커 유전자 (epithelial marker gene) 발현을 억제하고, 간엽성 유전자 (mesenchymal gene) 발현을 활성화시킨다 (Sci Signal, 2014, 7: re8). 이에, 본 발명자들은 Gas6가 TGF-β으로 자극된 LA-4 세포에서 상기 전사인자 발현을 억제하는지 확인하고자 하였다. In EMT induction, the activated Smad or non-Smad signal mediates transcriptional regulation of the Snai, Zeb and Basic helix-loop-helix transcription factor families, inhibiting the expression of epithelial marker genes, (Sci Signal, 2014, 7: re8). Thus, the present inventors sought to determine whether Gas6 inhibits the expression of the transcription factor in LA-4 cells stimulated with TGF- ?.
LA-4 세포 또는 HEK 293 세포에 Gas6를 처리하고 20 시간 후 TGF-β을 처리한 결과, TGF-β에 의해 유도되는 Snai1/2, Zeb1/2 및 Twist1의 mRNA 발현이 모두 억제되는 것을 확인하였다 (각각 도 5 및 도 6). LA-4 세포에서 Gas6의 처리는 TGF-β에 의해 매개되는 Smad2 및 Smad3의 인산화에 영향을 미치지 않았으나 (도 7), TGF-β에 의해 유도되는 ERK (extracellular signal-regulated kinase) 및 Akt의 인산화를 부분적으로 억제하였다 (도 8 및 도 9). p38 MAP (mitogen-activated protein) 키나아제의 인산화에는 영향이 없었다 (도 10). 상기 데이터들은 Gas6 처리 후 합성되거나 분비되는 생활성 매개체들이 ERK 및 Akt 경로를 포함하는 Smad-독립적인 TGF-β신호를 실질적으로 차단함으로써, LA-4 세포에서 EMT 과정을 막기 위해 상기 전사 조절자들을 감소시킨다는 것을 시사한다.As a result of treatment of LA-4 cells or HEK 293 cells with TGF-β after 20 hours of treatment with Gas6, it was confirmed that mRNA expression of TGF-β-induced Snai1 / 2, Zeb1 / 2 and Twist1 was all suppressed (Figures 5 and 6, respectively). Gas6 treatment in LA-4 cells did not affect the phosphorylation of Smad2 and Smad3 mediated by TGF-β (FIG. 7), but the TGF-β induced extracellular signal-regulated kinase (ERK) (Figs. 8 and 9). There was no effect on the phosphorylation of p38 MAP (mitogen-activated protein) kinase (Fig. 10). The data show that the bioactive agents synthesized or secreted following Gas6 treatment substantially block Smad-independent TGF-β signals including ERK and Akt pathways, thereby preventing the EMG process in LA-4 cells .
실험예 3: LA-4 세포에서 Gas6 처리에 의한 COX-2 유래 PGE2 및 PGD2 분비, 및 이의 EMT 억제 효과 확인Experimental Example 3: Confirmation of COX-2-derived PGE 2 and PGD 2 secretion and their EMT inhibitory effects by Gas6 treatment in LA-4 cells
COX-2/PGE2 및 PGD2 경로는 폐와 신장 상피세포에서 EMT를 억제하는 것으로 알려져 있다 (Sci Rep, 2016, 6: 20992). 따라서, 본 발명자들은 Gas6에 반응하여 LA-4에서 분비되는 COX-2 유래 PGE2 및 PGD2가 자가분비로 LA-4 세포에서 항-EMT 효과를 매개하는 것인지 확인하고자 하였다. The COX-2 / PGE 2 and PGD 2 pathways are known to inhibit EMT in lung and kidney epithelial cells (Sci Rep, 2016, 6: 20992). Therefore, the inventors sought to determine whether COX-2-derived PGE 2 and PGD 2 secreted from LA-4 in response to Gas6 mediate the anti-EMT effect in LA-4 cells by autocrine secretion.
먼저, 본 발명자들은 LA-4 세포에서 Gas6에 의한 COX-1 및 COX-2의 mRNA 및 단백질 발현, 그리고 PGE2 및 PGD2의 생산 변화를 확인하였다. 그 결과, COX-2 mRNA 수준은 Gas6 처리 1 시간 후 최대치를 보이고, 20 시간째에 원래 수준으로 회복되었으며, COX-1 mRNA 발현은 Gas6 처리 후 24 시간 이내에 변화가 없었다 (도 11). COX-2 단백질 발현은 Gas6 처리 후 24 시간 까지 점진적으로 증가하였으나, COX-1 발현은 상기 기간 동안 변화가 없었다 (도 12). 또한 Gas6 처리 후 20 시간째에 LA-4 세포에서 EIA로 측정된 PGE2 및 PGD2 분비는 각각 3.3 및 2.5 배로 증가하였다 (도 13). First, the present inventors confirmed mRNA and protein expression of COX-1 and COX-2 by Gas6 and changes in production of PGE 2 and PGD 2 in LA-4 cells. As a result, the level of COX-2 mRNA peaked at 1 hour after treatment with Gas6, returned to its original level at 20 hours, and the expression of COX-1 mRNA was not changed within 24 hours after treatment with Gas6 (FIG. COX-2 protein expression gradually increased until 24 hours after Gas6 treatment, but COX-1 expression did not change during this period (FIG. 12). In addition, PGE 2 and PGD 2 secretion measured by EIA in LA-4 cells increased 3.3 and 2.5-fold at 20 hours after Gas6 treatment (Fig. 13).
다음으로, LA-4 세포에서 Gas6 자극에 의한 COX-2 유도가 PGE2 및 PGD2 생산을 향상시키는지 확인하기 위해, LA-4 세포를 COX-2 특이적 siRNA 또는 음성-대조군 siRNA로 형질주입하고, 6 시간 동안 배양하였다. 그 결과, 음성-대조군 siRNA는 세포 내 COX-2 단백질 양에 영향을 미치지 않았으나, COX-2 특이적 siRNA로 형질주입시킨 세포에서는 야생형 LA-4 세포에 비해 COX-2 단백질 양이 ~60 % 이상 감소되었다 (도 14). COX-2 siRNA를 형질주입한 경우 Gas6에 의해 유도되는 PGE2 및 PGD2 분비를 막았는데 (도 15), 이는 LA-4 세포에서 Gas6로 유도된 PGE2 및 PGD2 생산 증가가 COX-2 발현 유도로부터 유래된다는 것을 시사한다.Next, LA-4 cells were transfected with a COX-2 specific siRNA or a negative-control siRNA in order to confirm that COX-2 induction by Gas6 stimulation enhanced PGE 2 and PGD 2 production in LA-4 cells And cultured for 6 hours. As a result, the negative control siRNA did not affect the amount of intracellular COX-2 protein but the amount of COX-2 protein was ~ 60% or more higher than that of wild-type LA-4 cells in the cells transfected with COX-2 specific siRNA (Fig. 14). 15 inhibited Gas6-induced PGE 2 and PGD 2 secretion when COX-2 siRNA was injected (FIG. 15), suggesting that increased production of Gas6-induced PGE 2 and PGD 2 in LA-4 cells resulted in COX-2 expression Induction &lt; / RTI &gt;
Gas6의 항-EMT 효과에서 COX-2 신호의 기능적 연관성을 추가적으로 확인하기 위해, Gas6를 처리하기 전 1 시간 동안 고선택성 COX-2 억제제인 NS-398을 LA-4 세포에 처리하였다. 그 결과, NS-398은 TGF-β으로 유도되는 세포 형태 변화, 그리고 유전자 및 단백질 수준에서 E-cadherin 손실 및 α-SMA와 N-cadherin의 합성에 대한 Gas6의 효과를 모두 반전시켰다 (도 16 내지 도 18). 또한 LA-4 세포에서 COX-2 유전자를 넉다운 (knockdown)시킨 경우에도 마찬가지로 Gas6의 효과를 반전시켰다 (도 19 및 도 20). 나아가, NS-398과 COX-2 siRNA는 모두 TGF-β으로 유도되는 Snai1, Zeb1 및 Twist1 mRNA 발현을 감소시키는 Gas6의 효과를 반전시켰다 (도 21 및 도 22). 또한, COX-2 siRNA는 LA-4 세포에서 TGF-β으로 유도되는 ERK1/2 및 AKT의 인산화를 감소시키는 Gas6의 효과를 반전시켰으나, 음성 대조군 siRNA에서는 이러한 결과가 나타나지 않았다 (도 23 및 도 24).In order to further confirm the functional relevance of the COX-2 signal in the anti-EMT effect of Gas6, LA-4 cells were treated with the highly selective COX-2 inhibitor NS-398 for 1 hour before treatment with Gas6. As a result, NS-398 reversed both the cell morphology changes induced by TGF-β and E-cadherin loss at gene and protein levels and the effect of Gas6 on the synthesis of α-SMA and N-cadherin (FIGS. 18). In addition, knockdown of the COX-2 gene in LA-4 cells also reversed the effect of Gas6 (FIGS. 19 and 20). Furthermore, both NS-398 and COX-2 siRNA reversed the effect of Gas6 on reducing TGF-β-induced Snai1, Zeb1 and Twist1 mRNA expression (FIGS. 21 and 22). In addition, COX-2 siRNA reversed the effect of Gas6, which reduces phosphorylation of ERK1 / 2 and AKT induced by TGF-beta in LA-4 cells, but this result was not observed in negative control siRNA (Fig. 23 and Fig. 24 ).
다음으로, LA-4 세포에서 COX-2로부터 유래된 PGE2 및 PGD2 분비가 항-EMT 효과를 매개하는지 확인하기 위해, Gas6가 있거나 없는 조건에서 TGF-β처리 한 시간 전 LA-4 세포에 EP2 (E-prostanoid-2 receptor) 길항제 (AH-6809), EP4 길항제 (AH-23848), DP1 길항제 (BW-A868C) 또는 DP2 길항제 (BAY-u3405)와 같은 PGE2- 또는 PGD2- 특이적 수용체의 길항제를 처리하였다. 그 결과, Gas6의 항-EMT 효과는 EP2와 DP2의 길항제에 의해 현저하게 반전되었다 (도 25 내지 도 27, 도 29, 도 30 및 도 32). 그러나 EP4 길항제 및 DP1 길항제는 Gas6의 항-EMT 효과에 약하게 영향을 미치거나 거의 영향을 미치지 않았다 (도 25, 도 26, 도 28, 도 29, 도 31 및 도 33). 상기 결과들은 LA-4 세포에서 항-EMT 효과는 주로 EP2 및 DP2 수용체를 통한 COX-2 유래 PGE2 및 PGD2 신호 각각에 의해 매개된다는 것을 시사한다.Next, in order to confirm whether PGE 2 and PGD 2 secretion derived from COX-2 mediate the anti-EMT effect in LA-4 cells, LA-4 cells were pretreated with TGF- EP2 PGE 2, such as (E-prostanoid-2 receptor) antagonists (AH-6809), EP4 antagonists (AH-23848), DP1 antagonist (BW-A868C) or DP2 antagonists (BAY-u3405) - or PGD 2 - specific Receptor antagonist. As a result, the anti-EMT effect of Gas6 was significantly reversed by antagonists of EP2 and DP2 (Figs. 25-27, 29, 30 and 32). However, EP4 antagonist and DP1 antagonist had little or no effect on the anti-EMT effect of Gas6 (Figs. 25, 26, 28, 29, 31 and 33). These results suggest that the anti-EMT effect in LA-4 cells is mediated primarily by COX-2-derived PGE 2 and PGD 2 signals through EP2 and DP2 receptors, respectively.
실험예 4: LA-4 세포에서 Gas6 처리에 의한 RhoA 경로-의존적 HGF 분비 및 이의 EMT 억제 효과 확인Experimental Example 4: Confirmation of RhoA pathway-dependent HGF secretion and its EMT inhibitory effect by Gas6 treatment in LA-4 cells
본 발명자들은 종래 Gas6가 RhoA-의존적 경로를 통해 대식세포에서 HGF의 mRNA 및 단백질 생산을 유도한다고 보고한바 있다 (J Pharmacol Exp Ther, 2014, 350: 563). HGF는 LA-4 세포에서 TGF-β으로 유도되는 EMT를 매개한다 (Sci Rep, 2016, 6: 20992). 따라서 본 발명자들은 Gas6로 유도되는 항-EMT 효과에 있어서 LA-4 세포에서의 RhoA-의존적 HGF 분비가 가지는 역할을 확인하고자 하였다. We have previously reported that Gas6 induces mRNA and protein production of HGF in macrophages via the RhoA-dependent pathway (J Pharmacol Exp Ther, 2014, 350: 563). HGF mediates EMT induced by TGF-? In LA-4 cells (Sci. Rep., 2016, 6: 20992). Therefore, the inventors sought to determine the role of RhoA-dependent HGF secretion in LA-4 cells in the anti-EMT effect induced by Gas6.
LA-4 세포에 Gas6를 처리한 결과, Gas6로 유도되는 HGF mRNA 발현은 3 시간째에 가장 높은 수준을 나타낸 뒤, 6 시간째까지 절반 수준으로 감소되었다 (도 34). ELISA로 측정된 LA-4 세포의 HGF 분비는 기본 분비 수준과 비교하였을 때 Gas6 처리로 인해 다소 증가하였다 (도 35). 항-HGF α쇄 항체를 이용한 LA-4 세포 용해물에 대한 면역블랏 분석 결과를 통해 세포 내 HGF 단백질 수준 역시 Gas6 처리에 의해 증가된다는 것을 알 수 있었다 (도 36). As a result of treatment of LA-4 cells with Gas6, Gas6-induced HGF mRNA expression showed the highest level at 3 hours and then decreased to half level at 6 hours (Fig. 34). HGF secretion of LA-4 cells as measured by ELISA increased somewhat due to treatment with Gas6 as compared to the basal secretion level (Fig. 35). Immunoblot analysis of the LA-4 cell lysate using the anti-HGF? Chain antibody showed that the intracellular HGF protein level was also increased by Gas6 treatment (FIG. 36).
다음으로, RhoA/Rho 키나아제 신호를 억제하기 위해, Gas6 자극 전 LA-4 세포에 RhoA-특이적 siRNA를 24 시간 동안 형질주입하거나 Rho 키나아제 억제제인 Y-27632를 1 시간 동안 처리하였다. 그 결과 RhoA 넉다운 및 Rho 키나아제 억제는 모두 TGF-β유도 EMT에 대한 Gas6의 효과를 반전시켰다 (도 37 내지 도 44). 또한 RhoA siRNA 는 LA-4 세포에서 TGF-β유도 ERK1/2 및 AKT의 인산화 감소에 대한 Gas6의 효과를 완전히 반전시켰으나, 음성 대조군은 이러한 효과를 나타내지 않았다 (도 45 및 도 46). 또한 TGF-β처리 1 시간 전 LA-4 세포에 c-Met 길항제인 PHA-665752를 처리함으로써 c-Met 신호를 억제한 경우에도, Gas6의 항-EMT 효과를 반전시켰다 (도 47 내지 도 50). 상기 데이터들은 LA-4 세포에서 RhoA/Rho 키나아제-의존적 HGF 생산 역시 자가분비로 c-Met 신호를 통해 Gas6의 항-EMT 효과를 매개한다는 것을 강력하게 시사한다.Next, in order to suppress the RhoA / Rho kinase signal, RhoA-specific siRNA was transfected into LA-4 cells before Gas6 stimulation for 24 hours or the Rho kinase inhibitor Y-27632 was treated for 1 hour. As a result, both RhoA knockdown and Rho kinase inhibition reversed the effect of Gas6 on TGF-beta induced EMT (Figures 37-44). In addition, RhoA siRNA completely reversed the effect of Gas6 on phosphorylation of TGF-beta induced ERK1 / 2 and AKT in LA-4 cells, but negative control did not show this effect (FIGS. 45 and 46). In addition, the c-Met antagonist PHA-665752 was treated with LA-4 cells 1 hour before TGF-beta treatment to invert the anti-EMT effect of Gas6 (Figs. 47 to 50) . These data strongly suggest that RhoA / Rho kinase-dependent HGF production in LA-4 cells also mediates the anti-EMT effect of Gas6 through c-Met signaling by self-secretion.
실험예 5: Gas6의 EP2, DP2 및 c-Met 발현 향상 효과Experimental Example 5: Enhancement of EP2, DP2 and c-Met Expression of Gas6
앞선 실시예에서 확인한 바와 같이, Gas6 처리에 의해 생산된 PGE2, PGD2 및 HGF는 주로 각각 EP2, DP2 및 c-MET을 통해 항-EMT 신호를 야기한다. PGE2, PGD2 및 HGF가 EMT 억제제로서 자가분비 방식으로 작용하는지 평가하기 위해, LA-4 세포에서 상기 가용성 중재자 (soluble mediator)의 효과를 기저 농도 (각각 35, 6 및 169 pg/ml) 및 자극 농도 (각각 118, 28 및 194 pg/ml)에서 확인하였다. As confirmed in the previous examples, PGE 2 , PGD 2 and HGF produced by the Gas6 treatment mainly cause anti-EMT signals through EP2, DP2 and c-MET, respectively. In order to evaluate whether PGE 2 , PGD 2 and HGF acted in a self-secretion manner as an EMT inhibitor, the effects of the soluble mediator on LA-4 cells were evaluated at basal concentrations (35, 6 and 169 pg / ml, respectively) Stimulation concentrations (118, 28 and 194 pg / ml, respectively).
그 결과, 자극 농도의 PGE2 및 PGD2는 기저 농도와 비교하여 단백질 수준에서 TGF-β으로 유도되는 EMT 마커 변화를 억제하였다 (도 51). 상기 데이터는 LA-4 세포에서 Gas6로 유도되는 PGE2 및 PGD2 방출에 의해 EMT 억제가 현저히 유발된다는 것을 뒷받침하는 것이다. As a result, stimulation concentrations of PGE 2 and PGD 2 inhibited the change of EMT markers induced by TGF-β at the protein level as compared with the basal concentration (FIG. 51). The data support that EMT inhibition is significantly induced by PGE 2 and PGD 2 release induced in Gas-6 in LA-4 cells.
이와 달리, HGF는 자극 농도 (194 pg/ml) 및 기저 농도 (169 pg/ml) 모두에서 항-EMT 효과를 보이지 않았지만 (도 51), Gas6로 전처리하고 20 시간 후 새로운 배지로 교체한 뒤 HGF 194 pg/ml을 첨가한 경우, TGF-β으로 유도된 EMT 마커 변화가 억제되었다 (도 52). 그러나 기저 농도의 HGF는 상기 실험 조건에서 항-EMT 효과를 보이지 않았다. 나아가 c-MET 단백질 양은 Gas6 처리 20 시간 후 증가되었다 (도 53). 상기 데이터는 상대적으로 낮은 HGF 생산에도 불구하고, 상기 실험 조건에서 HGF가 c-MET 발현을 증가시키기 때문에 항-EMT 활성을 가지게 할 수 있다는 가능성을 시사한다. LA-4 세포에서 Gas6 처리는 EP2 및 DP2 단백질 양 역시 증가시켰으나, EP4 및 DP1 단백질 양을 증가시키지는 않았다 (도 53). 한편, LA-4 세포에서 상기 중재자들을 모두 자극 농도로 첨가한 경우 시너지 효과를 보이지 않았는데 (도 51), 이는 이들이 동일 표적 분자를 통해 TGF-β으로 유도되는 신호경로를 억제한다는 것을 시사한다.In contrast, HGF did not show an anti-EMT effect at both stimulation (194 pg / ml) and basal (169 pg / ml) concentrations (Figure 51), but after 20 hours of pretreatment with Gas6, When 194 pg / ml was added, changes in EMT markers induced by TGF-β were inhibited (FIG. 52). However, basal HGF did not show anti-EMT effect under the above experimental conditions. Furthermore, the amount of c-MET protein was increased after 20 hours of Gas6 treatment (Fig. 53). This data suggests that despite the relatively low HGF production, HGF may have anti-EMT activity because it increases c-MET expression in the experimental conditions. Gas6 treatment in LA-4 cells also increased the amount of EP2 and DP2 protein, but did not increase the amount of EP4 and DP1 protein (FIG. 53). On the other hand, when all of the mediators were added at the stimulation concentration in LA-4 cells, they did not show synergistic effect (Fig. 51), suggesting that they inhibit TGF-beta induced signal pathway through the same target molecule.
실험예 6: LA-4 세포에서 Gas6-유도 EMT 억제에 대한 Axl 또는 Mer 영향Experimental Example 6 Effect of Axl or Mer on Gas6-induced EMT Inhibition in LA-4 Cells
다음으로, 본 발명자들은 LA-4 세포에서 Gas6에 의해 Gas6/Axl 또는 Mer 신호가 조절되는지 확인하고자 하였다.Next, the present inventors tried to determine whether the Gas6 / Ax1 or Mer signal was regulated by Gas6 in LA-4 cells.
LA-4 세포에서 400 ng/ml의 Gas6를 시간 별로(0, 5, 15, 30, 60 및 120 분) 처리 하고, Axl 또는 Mer의 인산화 수준을 측정하였다(도 54). 그 결과, Axl 또는 Mer 인산화 수준은 모두 Gas6 처리 30분 후 최대치를 보이고, 점진적으로 감소하였다.In the LA-4 cells, 400 ng / ml of Gas6 was treated with time (0, 5, 15, 30, 60 and 120 minutes) and the level of phosphorylation of Axl or Mer was measured (Fig. 54). As a result, the levels of AxI or Mer phosphorylation all peaked at 30 minutes after treatment with Gas6, and gradually decreased.
다음으로, LA-4 세포를 Axl 또는 Mer 특이적 siRNA 또는 음성-대조군 siRNA로 형질주입하고, 48 시간 동안 배양하고, Axl 또는 Mer 발현 수준을 측정하였다(도 55). 그 결과, 음성-대조군 siRNA는 세포 내 Axl 또는 Mer 단백질 양에 영향을 미치지 않았으나, Axl 또는 Mer 특이적 siRNA로 형질주입시킨 세포에서는 야생형 LA-4 세포에 비해 Axl 또는 Mer 단백질 양이 70% 이상 감소되었다. Next, LA-4 cells were transfected with AxI or Mer specific siRNA or negative-control siRNA, cultured for 48 hours, and the level of AxI or Mer expression was measured (Fig. 55). As a result, the negative control siRNA did not affect the amount of AxI or Mer protein, but the amount of AxI or Mer protein decreased by 70% or more in the cells transfected with AxI or Mer specific siRNA compared with wild type LA-4 cells .
또한, Axl 또는 Mer siRNA를 형질주입한 경우 Gas6에 의해 유도되는 COX-2 mRNA 상승, PGE2 및 PGD2 분비를 막았는데(도 56), 이는 LA-4 세포에서 Gas6로 유도된 PGE2 및 PGD2 생산 증가가 Axl 또는 Mer 발현 유도로부터 유래된다는 것을 시사한다.In addition, when Axl or Mer siRNA was transfected, it inhibited Gas6-induced COX-2 mRNA elevation and PGE 2 and PGD 2 secretion (Fig. 56), indicating that Gas6-induced PGE 2 and PGD 2 production is derived from induction of Axl or Mer expression.
또한, Axl 또는 Mer siRNA를 형질주입한 경우 Gas6에 의해 유도되는 RhoA 활성 증가, HGF mRNA 및 단백질 수준 증가를 막았는데(도 57), 이는 LA-4 세포에서 Gas6로 유도된 RhoA활성 증가, HGF mRNA 및 단백질 수준 증가가 Axl 또는 Mer 발현 유도로부터 유래된다는 것을 시사한다.In addition, when Axl or Mer siRNA was transfected, it inhibited Gas6-induced increase of RhoA activity and increase of HGF mRNA and protein level (Fig. 57), indicating that Gas6-induced increase of RhoA activity and HGF mRNA And that increased protein levels are derived from inducing Axl or Mer expression.
또한, 본 발명자들은 LA-4 세포에서 Gas6에 의해 EMT 억제에 대한 Axl 또는 Mer의 영향을 확인하고자 하였다.In addition, the present inventors tried to confirm the effect of Axl or Mer on EMT inhibition by Gas6 in LA-4 cells.
LA-4 세포를 Axl 또는 Mer 특이적 siRNA 또는 음성-대조군 siRNA로 형질주입하고, 48 시간 동안 배양하고, EMT 마커의 mRNA 및 단백질 발현 수준을 측정하였다(도 58 및 도 59). 그 결과, Axl 또는 Mer siRNA를 형질주입한 경우 Gas6에 의해 유도되는 E-cadherin 증가 및 α-SMA와 N-cadherin 감소를 모두 반전시켰다. 이는 LA-4 세포에서 Gas6로 유도된 E-cadherin 증가 및 α-SMA와 N-cadherin 감소가 Axl 또는 Mer 발현 유도로부터 유래된다는 것을 시사한다.LA-4 cells were transfected with AxI or Mer-specific siRNA or negative-control siRNA, cultured for 48 hours, and mRNA and protein expression levels of EMT markers were measured (FIGS. 58 and 59). As a result, E-cadherin induced by Gas6 and α-SMA and N-cadherin decreased when Axl or Mer siRNA was transfected. This suggests that the increase in E-cadherin and decrease in? -SMA and N-cadherin induced in Gas-6 in LA-4 cells result from induction of Axl or Mer expression.
또한, 본 발명자들은 LA-4 세포에서 Gas6에 의해 EMT 조절 전사 인자 발현 및 비 Smad TGF-β1 신호에 대한 Axl 또는 Mer의 영향을 확인하고자 하였다.In addition, we sought to determine the effect of Axl or Mer on the expression of EMT regulatory transcription factors and non-Smad TGF-β1 signal by Gas6 in LA-4 cells.
LA-4 세포를 Axl 또는 Mer 특이적 siRNA 또는 음성-대조군 siRNA로 형질주입하고, 48 시간 동안 배양하고, EMT 조절 전사 인자의 mRNA 및 단백질 발현 수준을 측정하였다(도 60). 그 결과, Gas6에 의해 억제된 Snai1, Zeb1 및 Twist1의 mRNA 발현이 모두 증가하였다. 또한, Gas6에 의해 억제된 ERK (extracellular signal-regulated kinase) 및 Akt의 인산화를 모두 증가하였다(도 61 및 도 62). 이는 LA-4 세포에서 Gas6로 유도된 EMT 전사 인자 및 ERK 및 Akt 인산화 억제가 Axl 또는 Mer 발현 유도로부터 유래된다는 것을 시사한다.LA-4 cells were transfected with AxI or Mer specific siRNA or negative-control siRNA, cultured for 48 hours, and mRNA and protein expression levels of EMT regulated transcription factors were measured (Figure 60). As a result, expression of mRNA of Snai1, Zeb1 and Twist1 inhibited by Gas6 was increased. In addition, both ERK (extracellular signal-regulated kinase) suppressed by Gas6 and phosphorylation of Akt were both increased (Figs. 61 and 62). This suggests that the EMT transcription factor and ERK and Akt phosphorylation inhibition induced in Gas-6 in LA-4 cells are derived from induction of Axl or Mer expression.
실험예 7: 초대 마우스 AT II 세포에서 EMT에 대한 Gas6 영향Experimental Example 7: Effect of Gas6 on EMT in primary mouse AT II cells
7-1. 초대 마우스 AT II 세포에서 TGF-β1-유도 EMT에 대한 Gas6 전처리의 효과7-1. Effect of pretreatment of Gas6 on TGF-β1-induced EMT in invasive mouse AT II cells
본 발명자들은 Gas6 처리가 초대 마우스 AT II 세포에서 TGF-β유도 EMT 과정을 조절하는지 확인하고자 하였다. AT II 세포에 400 ng/ml의 Gas6를 처리하고 20 시간 후, 10 ng/ml의 TGF-β처리로 48 시간 또는 72 시간 처리하고, EMT 마커 mRNA 및 단백질 발현 수준을 확인하였다. mRNA 발현 수준은 Hprt mRNA을 기준으로 측정하였다(도 63 및 도 64). 상기 데이터는 Gas6 처리 후 합성, 분비된 생활성 매개체들이 AT II 세포에서 자가분비 (autocrine) 방식으로 TGF-β신호를 막을 수 있다는 가능성을 시사한다. 또한 본 발명자들은 AT II 세포에 Gas6를 처리하고 20 시간 후 TGF-β을 처리한 결과, TGF-β에 의해 유도되는 Snai1/2, Zeb1/2 및 Twist1의 mRNA 발현이 모두 억제되는 것을 확인하였다(도 65). 상기 데이터들은 Gas6 처리 후 AT II 세포에서 EMT 과정을 막기 위해 상기 전사 조절자들을 감소시킨다는 것을 시사한다.We sought to determine if Gas6 treatment modulates the TGF-beta induced EMT process in invasive mouse AT II cells. AT II cells were treated with 400 ng / ml of Gas6 and treated with TGF-β at 10 ng / ml for 48 hours or 72 hours after 20 hours, and the level of EMT marker mRNA and protein expression was confirmed. mRNA expression levels were measured on the basis of Hprt mRNA (Figures 63 and 64). This data suggests that synthetic and secreted mediators after Gas6 treatment can block TGF-beta signaling in autocrine manner in AT II cells. In addition, the present inventors confirmed that TGF-β treatment of AT II cells after 20 hours of treatment with Gas6 inhibited the expression of mRNA of Snai1 / 2, Zeb1 / 2, and Twist1 induced by TGF-β 65). These data suggest that the GasE6 treatment reduces the transcriptional regulators to block the EMT process in AT II cells.
7-2. 초대 마우스 AT II 세포에서 Gas6-유도 EMT 억제에 대한 COX-2-신호 확인7-2. COX-2 signaling for Gas6-induced EMT inhibition in invasive mouse AT II cells
본 발명자들은 초대 마우스 AT II 세포에서 Gas6에 의한 COX-2 신호를 조절하는 확인하고자 하였다.The present inventors intend to confirm the regulation of COX-2 signal by Gas6 in early mouse AT II cells.
본 발명자들은 초대 마우스 AT II 세포에 400 ng/ml의 Gas6를 시간 별로(0, 1, 2, 3, 및 6 시간) 처리 하고, COX-2 mRNA 발현 수준을 측정하였다(도 66). COX-2 mRNA 발현 수준은 Gas6 처리 1시간 후 최대치를 보이고, 점진적으로 감소하였다.We treated 400 ng / ml Gas6 with time (0, 1, 2, 3, and 6 hours) and measured levels of COX-2 mRNA expression in primary mouse AT II cells (Fig. 66). COX-2 mRNA expression levels were maximal after one hour of treatment with Gas6 and gradually decreased.
다음으로, 본 발명자들은 Gas6의 항-EMT 효과에서 COX-2 신호의 기능적 연관성을 추가적으로 확인하기 위해, Gas6를 처리하기 전 1 시간 동안 고선택성 COX-2 억제제인 NS-398을 초대 마우스 AT II 세포에 처리하였다. 그 결과, NS-398은 TGF-β으로 유도되는 유전자 수준에서 E-cadherin 손실 및 α-SMA와 N-cadherin의 합성, Snai1, Zeb1 및 Twist1 mRNA 발현 감소에 대한 Gas6의 효과를 모두 반전시켰다(도 67 및 도 68).Next, in order to further confirm the functional relevance of the COX-2 signal in the anti-EMT effect of Gas6, we used NS-398, a highly selective COX-2 inhibitor, for 1 hour before treatment with Gas6 in primary mouse AT II cells Lt; / RTI &gt; As a result, NS-398 reversed the effects of Gas6 on E-cadherin loss, synthesis of α-SMA and N-cadherin, and reduction of Snai1, Zeb1 and Twist1 mRNA expression at the gene level induced by TGF-β 67 and 68).
다음으로, 본 발명자들은 Gas6가 있거나 없는 조건에서 TGF-β처리 한 시간 전 초대 마우스 AT II 세포에 EP2 (E-prostanoid-2 receptor) 길항제 (AH-6809) 또는 DP2 길항제 (BAY-u3405)와 같은 PGE2- 또는 PGD2- 특이적 수용체의 길항제를 처리하였다. 그 결과, Gas6의 항-EMT 효과는 EP2와 DP2의 길항제에 의해 현저하게 반전되었다(도 69 및 도 70).Next, the present inventors examined whether or not TGF-beta-treated primary mouse AT II cells were treated with EP2 (E-prostanoid-2 receptor) antagonist (AH-6809) or DP2 antagonist (BAY- u3405) Antagonists of PGE 2 - or PGD 2 - specific receptors were treated. As a result, the anti-EMT effect of Gas6 was significantly reversed by antagonists of EP2 and DP2 (Fig. 69 and Fig. 70).
상기 결과들은 AT II 세포에서 항-EMT 효과는 주로 EP2 및 DP2 수용체를 통한 COX-2 유래 PGE2 및 PGD2 신호 각각에 의해 매개된다는 것을 시사한다.These results suggest that the anti-EMT effect in AT II cells is mediated primarily by COX-2-derived PGE 2 and PGD 2 signals through EP2 and DP2 receptors, respectively.
7-3. 초대 마우스 AT II 세포에서 Gas6-유도 EMT 억제에 대한 RhoA 경로-의존적 HGF-신호 확인7-3. Identification of RhoA pathway-dependent HGF-signaling for Gas6-induced EMT inhibition in invasive mouse AT II cells
본 발명자들은 초대 마우스 AT II 세포에서 Gas6에 의한 RhoA 경로-의존적 HGF-신호를 조절하는 확인하고자 하였다.The present inventors have determined to modulate RhoA pathway-dependent HGF-signal by Gas6 in primary mouse AT II cells.
초대 마우스 AT II 세포에 Gas6를 처리한 결과, Gas6로 유도되는 HGF mRNA 발현은 점진적으로 증가하였다(도 71).As a result of treatment of Inv6 mouse AT II cells with Gas6, expression of HGF mRNA induced by Gas6 gradually increased (FIG. 71).
다음으로, RhoA/Rho 키나아제 신호를 억제하기 위해, Gas6 자극 전 초대 마우스 AT II 세포에 RhoA-특이적 siRNA를 24 시간 동안 형질주입하거나 Rho 키나아제 억제제인 Y-27632를 1 시간 동안 처리하였다. 그 결과 RhoA 넉다운 및 Rho 키나아제 억제는 모두 TGF-β유도 EMT에 대한 Gas6의 효과를 반전시켰다(도 72 및 도 73). 또한 TGF-β처리 1 시간 전 AT II 세포에 c-Met 길항제인 PHA-665752를 처리함으로써 c-Met 신호를 억제한 경우에도, Gas6의 항-EMT 효과를 반전시켰다(도 74 및 도 75).Next, in order to suppress the RhoA / Rho kinase signal, RhoA-specific siRNA was transfected into the invasive mouse AT II cells before Gas6 stimulation for 24 hours or the Rho kinase inhibitor Y-27632 was treated for 1 hour. As a result, both RhoA knockdown and Rho kinase inhibition reversed the effect of Gas6 on TGF-beta induced EMT (FIGS. 72 and 73). In addition, treatment of PHA-665752, a c-Met antagonist, with AT II cells 1 hour before TGF-beta treatment reversed the anti-EMT effect of Gas6 (FIGS. 74 and 75).
상기 데이터들은 AT II 세포에서 RhoA/Rho 키나아제-의존적 HGF 생산 역시 자가분비로 c-Met 신호를 통해 Gas6의 항-EMT 효과를 매개한다는 것을 강력하게 시사한다.The data strongly suggest that RhoA / Rho kinase-dependent HGF production in AT II cells also mediates the anti-EMT effect of Gas6 via self-secretion of the c-Met signal.
실험예 8: 사람 폐 선암종 세포(A549 세포)에서 TGF-β1-유도 EMT에 대한 Gas6 전처리의 효과Experimental Example 8: Effect of pretreatment of Gas6 on TGF-β1-induced EMT in human lung adenocarcinoma cells (A549 cells)
본 발명자들은 A549 세포에서 Gas6 투여가 TGF-β유도 EMT 과정을 조절하는지 확인하고자 하였다. We sought to determine whether administration of Gas6 in A549 cells regulates the TGF-beta induced EMT process.
A549 세포에 400 ng/ml의 Gas6를 처리하고 20 시간 후, 10 ng/ml의 TGF-β를 72 시간 처리하고, EMT 마커 mRNA 및 단백질 발현 수준을 확인하였다. mRNA 발현 수준은 Hprt mRNA을 기준으로 측정하였다(도 76 및 도 77). 상기 데이터는 Gas6 처리 후 합성, 분비된 생활성 매개체들이 A549 세포에서 자가분비 방식으로 TGF-β신호를 막을 수 있다는 가능성을 시사한다. A549 cells were treated with 400 ng / ml of Gas6, and after 20 hours, 10 ng / ml of TGF-beta was treated for 72 hours to confirm the level of EMT marker mRNA and protein expression. mRNA expression levels were measured on the basis of Hprt mRNA (Figures 76 and 77). This data suggests that synthetic and secreted mediators after Gas6 treatment may block TGF-β signaling in a self-secreted manner in A549 cells.
또한 본 발명자들은 A549 세포에 Gas6를 처리하고 20 시간 후 TGF-β을 처리한 결과, TGF-β에 의해 유도되는 Snai1/2, Zeb1/2 및 Twist1의 mRNA 발현이 모두 억제되는 것을 확인하였다(도 78). 상기 데이터들은 Gas6 처리 후 A549 세포에서 EMT 과정을 막기 위해 상기 전사 조절자들을 감소시킨다는 것을 시사한다.In addition, the present inventors confirmed that TGF-beta treatment of A549 cells after 20 hours of treatment with Gas6 inhibited the expression of mRNA of Snai1 / 2, Zeb1 / 2 and Twist1 induced by TGF-beta 78). These data suggest that the GasE6 treatment reduces the transcriptional regulators to prevent EMT processes in A549 cells.
실험예 9: 폐 섬유증 동물모델에서 Gas6의 EMT 억제 효과Experimental Example 9: EMT inhibition effect of Gas6 in an animal model of pulmonary fibrosis
9-1. 폐 섬유증 동물모델에서 블레오마이신-유도 EMT에 대한 Gas6의 억제 효과9-1. Inhibitory effect of Gas6 on bleomycin-induced EMT in an animal model of pulmonary fibrosis
본 발명자들은 Gas6 투여가 생체 내 폐 상피세포에서 EMT 과정을 조절하는지 확인하고자 하였다. The present inventors sought to determine whether administration of Gas6 regulates the EMT process in lung epithelial cells in vivo.
C57BL/6 마우스를 대조군(Control), Gas6 투여군(Gas6), 블레오마이신 투여군(BLM), Gas6+블레오마이신 투여군(Gas6+BLM)으로 각 5마리씩 분류하였다. 매일 블레오마이신 투여 전에 50 μg/ kg의 Gas6을 각 분류군 에 따라 투여하였다. 블레오마이신 투여 후 14일 째에 마우스의 폐에서 초대 마우스 AT II 세포를 분리하였다. C57BL / 6 mice were divided into five groups by Control, Gas6 group, Bleomycin group (BLM) and Gas6 + Bleomycin group (Gas6 + BLM). Each day, 50 μg / kg of Gas6 was administered according to each taxon before administration of bleomycin. At day 14 after bleomycin administration, primary mouse AT II cells were isolated from the lungs of mice.
분리된 마우스 세포의 형태를 확인하하였다. 그 결과, 블레오마이신이 투여된 마우스의 세포는 방추-유사 형태 (spindle-like morphology)로 변환되는 것을 확인하였으나, 블레오마이신 투여 전에 Gas6이 투여된 마우스의 세포는 방추-유사 형태가 감소하였다(도 79). The morphology of isolated mouse cells was determined. As a result, it was confirmed that the cells of the mice to which bromomycin was administered were converted into spindle-like morphology, but the cells of the mice to which Gas6 was administered before the bleomycin administration showed a decrease in the spindle-like morphology 79).
또한, EMT 마커(E-cadherin, N-cadherin 및 α-SMA)와 EMT-활성화 전사인자(Snai1, Zeb1 및 Twist1)의 발현 양상을 확인하기 위해 각 mRNA 수준을 측정하였다(도 80 및 도 81). 블레오마이신을 투여한 마우스는 E-cadherin 발현이 감소되고, N-cadherin과 α-SMA의 발현이 증가되었으나, Gas6를 처리한 경우 이러한 EMT 마커 발현 양상이 억제되는 것을 mRNA 수준에서 확인하였다. 또한, 블레오마이신에 의해 유도되는 Snai1, Zeb1 및 Twist1의 mRNA 발현이 Gas6를 처리한 경우 모두 억제되는 것을 확인하였다. 상기 결과는 Gas6 처리를 통해 폐 섬유증 마우스 모델에서 블레오마이신으로 유도된 EMT를 억제할 수 있다는 가능성을 시사한다. In order to confirm the expression pattern of EMT markers (E-cadherin, N-cadherin and a-SMA) and EMT-activated transcription factors (Snai1, Zeb1 and Twist1), each mRNA level was measured (FIGS. 80 and 81) . E-cadherin expression and N-cadherin and α-SMA expression were increased in bleomycin-treated mice, but the level of EMT marker expression was inhibited by Gas6 treatment at the mRNA level. In addition, it was confirmed that expression of mRNA of Snai1, Zeb1, and Twist1 induced by bleomycin was all inhibited when Gas6 was treated. These results suggest that Gas6 treatment may inhibit bleomycin-induced EMT in a pulmonary fibrosis mouse model.
9-2. 폐 섬유증 동물모델에서 COX-2 유래 PGE2, PGD2 및 HGF 분비에 대한 Gas6 처리의 효과9-2. Effect of Gas6 Treatment on COX-2-Derived PGE 2 , PGD 2 and HGF Secretion in Pulmonary Fibrosis Animal Model
본 발명자들은 생체 내에서 Gas6 투여가 PGE2, PGD2 및 HGF 경로를 조절하는지 확인하고자 하였다.The present inventors sought to determine whether administration of Gas6 in vivo regulates PGE 2 , PGD 2 and HGF pathways.
C57BL/6 마우스를 대조군(Control), Gas6 투여군(Gas6), 블레오마이신 투여군(BLM), Gas6+블레오마이신 투여군(Gas6+BLM)으로 각 5마리씩 분류하였다. 매일 블레오마이신 투여 전에 50 μg/ kg의 Gas6을 각 분류군 에 따라 투여하였다. 블레오마이신 투여 후 14일 째에 마우스의 폐에서 초대 마우스 AT II 세포를 분리하였다. 분리된 마우스 세포에서 COX-2 및 HGF의 mRNA, PGE2, PGD2, HGF 및 TGF-β 생산량을 확인하였다.C57BL / 6 mice were divided into five groups by Control, Gas6 group, Bleomycin group (BLM) and Gas6 + Bleomycin group (Gas6 + BLM). Each day, 50 μg / kg of Gas6 was administered according to each taxon before administration of bleomycin. At day 14 after bleomycin administration, primary mouse AT II cells were isolated from the lungs of mice. The production of mRNA, PGE 2 , PGD 2 , HGF and TGF-β of COX-2 and HGF was confirmed in isolated mouse cells.
그 결과, COX-2의 mRNA 수준은 Gas6 투여로 인해 대조군에 비해 2배 이상 상승하였고, 대조군에서 분비되지 않던 PGE2는 4000 pg/ml 이상 분비되었고, PGD2 단백질 수준은 대조군의 비해 약 4배 정도 증가하였다(도 82 및 도 83). 상기 결과는 폐 섬유증 마우스 모델에서 Gas6로 인한 EMT 억제가 COX-2 발현, PGE2 및 PGD2 생산 증가로부터 유래된다는 것을 시사한다.As a result, the mRNA level of COX-2 was increased more than twice as much as that of the control group due to the administration of Gas6, the PGE 2 secreted by the control group was secreted by 4000 pg / ml or more, and the PGD 2 protein level was about 4 times (Figs. 82 and 83). These results suggest that EM6 suppression due to Gas6 in pulmonary fibrosis mouse models results from increased COX-2 expression, PGE 2 and PGD 2 production.
또한, HGF의 mRNA 수준은 Gas6 투여로 인해 대조군에 비해 3배 이상 상승하였고, HGF는 2500 pg/ml 이상 분비되었고, 블레오마이신에 의해 증가한 TGF-β 단백질 수준은 Gas6 투여로 인해 감소하였다(도 84 및 도 85). 상기 결과는 폐 섬유증 마우스 모델에서 Gas6로 인한 EM을 억제가 HGF 증가 및 TGF-β 억제로부터 유래된다는 것을 시사한다.In addition, the mRNA level of HGF was increased more than 3 times as compared to the control group due to the administration of Gas6, HGF was secreted by 2500 pg / ml or more, and the level of TGF-beta protein increased by bleomycin was decreased due to administration of Gas6 And Fig. 85). These results suggest that inhibition of EM due to Gas6 in the lung fibrosis mouse model results from HGF increase and TGF-beta inhibition.
9-3. 폐 섬유증 동물모델에서 블레오마이신에 의해 유도된 EMT 마커 및 콜라겐 침착 변화에 대한 Gas6 처리의 효과9-3. Effect of Gas6 treatment on bleomycin-induced EMT markers and collagen deposition changes in pulmonary fibrosis animal models
본 발명자들은 생체 내에서 Gas6 투여가 EMT 마커 및 콜라겐 침착 변화를 조절하는지 확인하고자 하였다.The present inventors sought to determine whether administration of Gas6 in vivo controls changes in EMT markers and collagen deposition.
C57BL/6 마우스를 대조군(Control), Gas6 투여군(Gas6), 블레오마이신 투여군(BLM), Gas6+블레오마이신 투여군(Gas6+BLM)으로 각 5마리씩 분류하였다. 매일 블레오마이신 투여 전에 50 μg/ kg의 Gas6을 각 분류군 에 따라 투여하였다. 블레오마이신 투여 후 14일 및 21일 째에 마우스의 폐에서 초대 마우스 AT II 세포를 분리하였다. 각 분리된 마우스 세포에서 항-E-cadherin, 항-N-cadherin, 항-α-SMA, 항-제1형 콜라겐(collagen 1) 또는 항-피브로넥틴(Fibronectin) 항체를 이용하여 웨스턴블럿하였다. Α-tubulin을 기준으로 각 밴드의 상대 밀도(Relative Density)를 결정하였다.C57BL / 6 mice were divided into five groups by Control, Gas6 group, Bleomycin group (BLM) and Gas6 + Bleomycin group (Gas6 + BLM). Each day, 50 μg / kg of Gas6 was administered according to each taxon before administration of bleomycin. Early mouse AT II cells were isolated from the lungs of mice on days 14 and 21 after administration of bleomycin. Each separated mouse cell was Western blotted using anti-E-cadherin, anti-N-cadherin, anti-α-SMA, anti-type 1 collagen 1 or anti-fibronectin antibody. Relative density of each band was determined based on Α-tubulin.
그 결과, 14일째 및 21일째의 마우스 폐 조직에서 블레오마이신에 의해 감소한 E-cadherin 수준은 Gas6 투여로 인해 상승하였고, 블레오마이신에 의해 증가한 N-cadherin, α-SMA, 제1형 콜라겐 및 피브로넥틴 수준은 Gas6 투여로 인해 감소하였다(도 86 및 도 87).As a result, E-cadherin levels decreased by bleomycin in mouse lung tissue on days 14 and 21 were elevated due to the administration of Gas6, and the levels of N-cadherin, a-SMA, type 1 collagen and fibronectin increased by bleomycin (Fig. 86 and Fig. 87).
또한, 전체 폐의 콜라겐 축적은 21일 째에 하이드록시프롤린 수준을 측정하였다(도 88). 그 결과 블레오마이신에 의해 증가한 하이드록시프롤린 수준은 Gas6 투여로 인해 대조군과 비슷한 수준으로 감소하였다.In addition, the collagen accumulation of the entire lung was measured at the 21th day of hydroxyproline level (Fig. 88). As a result, the level of hydroxyproline increased by bromomycin was reduced to a level similar to that of the control group due to administration of Gas6.
상기 결과는 생체 내 블레오마이신에 의해 유발되는 섬유증이 Gas6 투여로 개선된다는 것을 시사한다.These results suggest that fibromyxia induced by in vivo bleomycin is improved by administration of Gas6.
상기 결과를 모두 종합하면, Gas6는 상피세포에서 PGE2, PGD2 및 HGF의 분비 생산을 유도하고, 이들의 수용체를 통한 신호전달 경로를 거쳐 자가분비 방식으로 작용함으로써, TGF-β에 의해 유도되는 EMT를 억제하는 항-EMT 효과를 가짐을 알 수 있다.Taken together, all of the above results, Gas6 has induced the secretory production of PGE 2, PGD 2 and HGF in epithelial cells and, through a signal transmission path through their receptors party by acting as a secretion system, induced by TGF-β EMT-inhibiting effect of EMT.
또한, LA-4 세포에서 Gas6는 Axl 또는 Mer 신호 경로를 기반으로 COX-2 신호 경로 및 RhoA 경로 의존적 HGF 분비하며, 초대 마우스 AT II 세포 및 사람 폐 선암종 세포에서도 LA-4 세포와 동일한 효과를 가짐을 알 수 있다. In addition, Gas6 in LA-4 cells secretes COX-2 signal pathway and RhoA pathway dependent HGF based on Axl or Mer signal pathway and has the same effect on LA-4 cell in primary mouse AT II cell and human lung adenocarcinoma cell .
또한, 동물실험을 통해 Gas6 투여가 생체 내에서 블레오마이신에 의해 유도되는 EMT를 억제하여 섬유증에 대한 우수한 예방 및 치료 효과를 가짐을 확인하였다.In addition, it was confirmed through animal experiments that administration of Gas6 suppressed EMT induced by bromomycin in vivo, and thus had an excellent prophylactic and therapeutic effect on fibrosis.
따라서 Gas6는 섬유증의 예방 또는 치료 용도로 매우 유용하게 이용될 수 있다.Therefore, Gas6 can be very usefully used for the prevention or treatment of fibrosis.
이상의 설명으로부터, 본 발명이 속하는 기술분야의 당업자는 본 발명이 그 기술적 사상이나 필수적 특징을 변경하지 않고서 다른 구체적인 형태로 실시될 수 있다는 것을 이해할 수 있을 것이다. 이와 관련하여, 이상에서 기술한 실시예들은 모든 면에서 예시적인 것이며 한정적인 것이 아닌 것으로 이해해야만 한다. 본 발명의 범위는 상기 상세한 설명보다는 후술하는 특허 청구범위의 의미 및 범위 그리고 그 등가 개념으로부터 도출되는 모든 변경 또는 변형된 형태가 본 발명의 범위에 포함되는 것으로 해석되어야 한다.From the above description, it will be understood by those skilled in the art that the present invention may be embodied in other specific forms without departing from the spirit or essential characteristics thereof. In this regard, it should be understood that the embodiments described above are illustrative in all aspects and not restrictive. The scope of the present invention should be construed as being included in the scope of the present invention without departing from the scope of the present invention as defined by the appended claims.

Claims (10)

  1. Gas6 (Growth arrest-specific 6) 단백질 또는 Gas6 단백질 수용체의 활성화제를 유효성분으로 포함하는, 섬유증의 예방 또는 치료용 약학적 조성물.A pharmaceutical composition for preventing or treating fibrosis, comprising as an active ingredient an activator of Gas6 (Growth arrest-specific 6) protein or Gas6 protein receptor.
  2. 제1항에 있어서, 상기 Gas6 단백질 수용체는 AXL 수용체 타이로신 키나아제, Mer 타이로신 키나아제 및 TYRO3로 이루어진 군에서 선택된 어느 하나 이상인, 약학적 조성물.The pharmaceutical composition according to claim 1, wherein the Gas6 protein receptor is any one or more selected from the group consisting of AXL receptor tyrosine kinase, Mer tyrosine kinase, and TYRO3.
  3. 제1항에 있어서, 상기 Gas6 단백질 수용체의 활성화제는 Protein S (Pros1), Tubby and tubby-like protein 1, 또는 Galectin3로 이루어진 군에서 선택된 어느 하나 이상인, 약학적 조성물.The pharmaceutical composition according to claim 1, wherein the activator of the Gas6 protein receptor is at least one selected from the group consisting of Protein S (Pros1), Tubby and tubby-like protein 1, or Galectin3.
  4. 제1항에 있어서, 상기 섬유증은 폐, 신장, 간, 심장, 뇌, 혈관, 관절, 장, 피부, 연조직, 골수, 음경, 복막, 랜즈, 근육, 척추, 고환, 난소, 유방, 갑상선, 고막, 췌장, 담낭, 방광 또는 전립선에서 발병한 것인, 약학적 조성물.The method of claim 1, wherein the fibrosis is selected from the group consisting of lung, kidney, liver, heart, brain, blood vessels, joints, bowel, skin, soft tissue, bone marrow, penis, peritoneum, lens, muscle, vertebra, testes, , Pancreas, gall bladder, bladder or prostate.
  5. 제1항에 있어서, 상기 조성물은 약학적으로 허용가능한 담체를 추가로 포함하는 것인, 약학적 조성물.The pharmaceutical composition of claim 1, wherein the composition further comprises a pharmaceutically acceptable carrier.
  6. 제1항에 있어서, 상기 조성물은 100 ㎍/kg 내지 500 ㎍/kg의 용량으로 투여되는 것인, 약학적 조성물.2. The pharmaceutical composition according to claim 1, wherein the composition is administered in a dose of 100 [mu] g / kg to 500 [mu] g / kg.
  7. 제1항에 있어서, 상기 조성물은 경구 또는 비경구로 투여되는 것인, 약학적 조성물.The pharmaceutical composition according to claim 1, wherein the composition is administered orally or parenterally.
  8. 제1항에 있어서, 상기 조성물은 세포의 EMT (epithelial to mesenchymal transition)를 억제하는 것인, 약학적 조성물.The pharmaceutical composition according to claim 1, wherein the composition inhibits the epithelial to mesenchymal transition (EMT) of the cell.
  9. Gas6 단백질 또는 Gas6 단백질의 수용체 활성화제를 섬유증을 갖거나 섬유증이 발병할 위험이 있는 개체에 투여하는 단계를 포함하는, 섬유증의 예방 또는 치료 방법.A method for preventing or treating fibrosis, comprising administering a receptor activator of Gas6 protein or Gas6 protein to a subject having fibrosis or at risk of developing fibrosis.
  10. Gas6 단백질 또는 이의 수용체 활성화제의 섬유증 예방 또는 치료 용도.Use of a Gas6 protein or a receptor activator thereof for the prophylaxis or treatment of fibrosis.
PCT/KR2018/007128 2017-06-23 2018-06-22 Composition for prevention or treatment of fibrosis, comprising gas6 protein or receptor activator thereof WO2018236194A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
KR20170079974 2017-06-23
KR10-2017-0079974 2017-06-23

Publications (1)

Publication Number Publication Date
WO2018236194A1 true WO2018236194A1 (en) 2018-12-27

Family

ID=64735998

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/KR2018/007128 WO2018236194A1 (en) 2017-06-23 2018-06-22 Composition for prevention or treatment of fibrosis, comprising gas6 protein or receptor activator thereof

Country Status (2)

Country Link
KR (1) KR102076777B1 (en)
WO (1) WO2018236194A1 (en)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN114272382A (en) * 2021-12-31 2022-04-05 上海中医药大学 Application and pharmaceutical composition of Galectin-3 inhibitor
CN114592009A (en) * 2022-04-20 2022-06-07 东南大学 Preparation method and application of recombinant adeno-associated virus GAS6 for promoting myocardial regeneration

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR20130004568A (en) * 2010-01-22 2013-01-11 더 보드 어브 트러스티스 어브 더 리랜드 스탠포드 주니어 유니버시티 Inhibition of axl signaling in anti-metastatic therapy
US20130189254A1 (en) * 2011-12-15 2013-07-25 Ruga Corporation Inhibition of AXL/GAS6 Signaling in the Treatment of Disease
CN105194673A (en) * 2015-09-29 2015-12-30 武汉大学 Functions and application of growth arrest-specific proteins 6 (GAS6) to treating myocardial hypertrophy
WO2017059555A1 (en) * 2015-10-05 2017-04-13 Universidad Del Desarrollo Biopharmacological composition comprising components produced and/or secreted by mesenchymal stem cells (msc), which allows the recovery of tissue regenerative capacity in a situation of hepatosteatosis or when the endogenous regenerative capacity of hepatic tissue is inhibited

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR20130004568A (en) * 2010-01-22 2013-01-11 더 보드 어브 트러스티스 어브 더 리랜드 스탠포드 주니어 유니버시티 Inhibition of axl signaling in anti-metastatic therapy
US20130189254A1 (en) * 2011-12-15 2013-07-25 Ruga Corporation Inhibition of AXL/GAS6 Signaling in the Treatment of Disease
CN105194673A (en) * 2015-09-29 2015-12-30 武汉大学 Functions and application of growth arrest-specific proteins 6 (GAS6) to treating myocardial hypertrophy
WO2017059555A1 (en) * 2015-10-05 2017-04-13 Universidad Del Desarrollo Biopharmacological composition comprising components produced and/or secreted by mesenchymal stem cells (msc), which allows the recovery of tissue regenerative capacity in a situation of hepatosteatosis or when the endogenous regenerative capacity of hepatic tissue is inhibited

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
FOURCOT, AGNES ET AL.: "Gas6 Deficiency Prevents Liver Inflammation, Steatohepatitis, and Fibrosis in Mice", AMERICAN JOURNAL OF PHYSIOLOGY-GASTROINTESTINAL AND LIVER PHYSIOLOGY, vol. 300, 2011, pages G1043 - G1053, XP055486956 *
LANDOLT, LEA ET AL.: "Clear Cell Renal Cell Carcinoma Is Linked to Epithelial-to-mesenchymaJ Transition and to Fibrosis", PHYSIOLOGICAL REPORTS, vol. 5, no. 11, 8 June 2017 (2017-06-08), pages 1 - 18, XP055567278 *

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN114272382A (en) * 2021-12-31 2022-04-05 上海中医药大学 Application and pharmaceutical composition of Galectin-3 inhibitor
CN114592009A (en) * 2022-04-20 2022-06-07 东南大学 Preparation method and application of recombinant adeno-associated virus GAS6 for promoting myocardial regeneration
CN114592009B (en) * 2022-04-20 2023-10-03 东南大学 Preparation method and application of recombinant adeno-associated virus GAS6 for promoting myocardial regeneration

Also Published As

Publication number Publication date
KR20190000841A (en) 2019-01-03
KR102076777B1 (en) 2020-02-12

Similar Documents

Publication Publication Date Title
WO2013169077A1 (en) Composition for preventing or treating cachexia
WO2015076621A1 (en) Peptide having angiogenesis inhibitory activity and composition containing same
WO2012026712A2 (en) Pharmaceutical composition for preventing or treating immune diseases or inflammatory diseases, containing stem cells treated with nod2 agonist or cultured product thereof
WO2017022962A1 (en) Composition for preventing or treating immune disease, comprising ripk inhibitor as active ingredient
WO2015156649A1 (en) Peptide having fibrosis inhibitory activity and composition containing same
WO2015167067A1 (en) Composition for organ, tissue, or cell transplantation, kit, and transplantation method
WO2011142514A1 (en) Composition containing pias3 as an active ingredient for preventing or treating cancer or immune disease
WO2011052883A2 (en) Method for activating a natural killer cell by adjusting the expression of the socs2 gene
WO2018236194A1 (en) Composition for prevention or treatment of fibrosis, comprising gas6 protein or receptor activator thereof
WO2018208011A2 (en) BIOCOMPATIBLE PEPTIDE SUPPRESSIVE OF AGGREGATION OF β-AMYLOID PROTEIN
WO2021033990A1 (en) Composition comprising exosomes derived from induced pluripotent stem cell-derived mesenchymal stem cell precursor for prevention or treatment of non-alcoholic steatohepatitis
WO2018026212A2 (en) Method for producing fibrosis disease model, and use of fibrosis disease model
WO2019022482A2 (en) Composition for preventing or treating fibrotic diseases comprising dendropanax morbifera extract as active ingredient
WO2017171373A2 (en) Composition for suppressing resistance to egfr-targeting agent
WO2014157965A1 (en) Composition for treatment or metastasis suppression of cancers which includes p34 expression inhibitor or activity inhibitor as active ingredient
WO2022055334A1 (en) Composition for preventing or treating pulmonary fibrosis disease
WO2020071641A1 (en) Pharmaceutical composition for preventing or treating fabry disease, containing tsp1 protein inhibitor as active ingredient
WO2017164486A1 (en) Composition for inhibiting resistance to anticancer agents, containing tesk1 inhibitor, and method for screening for tesk1 inhibitor
WO2011155643A1 (en) Phosphatidylinositol 3-kinases activity regulator including the fifth zinc finger domain of fog2
WO2021033973A1 (en) Composition for inhibiting myeloid-derived suppressor cell comprising mitf inhibitor as active ingredient
WO2020122498A1 (en) Pharmaceutical composition for treating pancreatitis, comprising clonal stem cells
WO2016204545A1 (en) Peptide for promoting osteogenesis or inhibiting osteolysis, and use thereof
WO2019231262A1 (en) Novel biphenyl derivative compound and use thereof
WO2018135738A1 (en) COMPOSITION FOR INHIBITING MYOSTATIN ACTIVITY, CONTAINING MATRIX gla PROTEIN AS ACTIVE INGREDIENT
WO2021251602A1 (en) Composition for preventing or treating disease caused by muscle loss, comprising agent inhibiting phf20

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: 18820241

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: 18820241

Country of ref document: EP

Kind code of ref document: A1