AU2021225272A1 - Use of PAX4 inhibitor in preparation of drug for inhibiting fibrosis - Google Patents

Use of PAX4 inhibitor in preparation of drug for inhibiting fibrosis Download PDF

Info

Publication number
AU2021225272A1
AU2021225272A1 AU2021225272A AU2021225272A AU2021225272A1 AU 2021225272 A1 AU2021225272 A1 AU 2021225272A1 AU 2021225272 A AU2021225272 A AU 2021225272A AU 2021225272 A AU2021225272 A AU 2021225272A AU 2021225272 A1 AU2021225272 A1 AU 2021225272A1
Authority
AU
Australia
Prior art keywords
pax4
fibrosis
cardiac
inhibiting
protein
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Granted
Application number
AU2021225272A
Other versions
AU2021225272B2 (en
Inventor
Guomin Hu
Mingzhe Li
Han XIAO
Youyi Zhang
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Peking University Third Hospital
Original Assignee
Peking University Third Hospital
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 Peking University Third Hospital filed Critical Peking University Third Hospital
Publication of AU2021225272A1 publication Critical patent/AU2021225272A1/en
Application granted granted Critical
Publication of AU2021225272B2 publication Critical patent/AU2021225272B2/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7088Compounds having three or more nucleosides or nucleotides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/18Drugs for disorders of the alimentary tract or the digestive system for pancreatic disorders, e.g. pancreatic enzymes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Public Health (AREA)
  • Animal Behavior & Ethology (AREA)
  • Veterinary Medicine (AREA)
  • Chemical & Material Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Medicinal Chemistry (AREA)
  • Engineering & Computer Science (AREA)
  • Organic Chemistry (AREA)
  • General Chemical & Material Sciences (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Epidemiology (AREA)
  • Cardiology (AREA)
  • Molecular Biology (AREA)
  • Pulmonology (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Investigating Or Analysing Biological Materials (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)

Abstract

Disclosed is the use of a PAX4 inhibitor in the preparation of a drug for inhibiting fibrosis. The inhibiting effect of a transcription factor PAX4 interferes by means of small interfering RNA so as to block the inhibiting effect thereof on a downstream gene, such that multiple fibrosis inhibiting factors thereof in the downstream execute a function. By means of small interfering RNA, both the expression of PAX4 and the expression level of fibrosis promoting factors are reduced and the fibrosis promoting effect thereof is inhibited, thereby inhibiting cardiac fibrosis. Therefore, PAX4 is a potential brand-new important target for treating cardiac fibrosis and further preventing heart failure in the future.

Description

Description
USE OF PAX4 INHIBITOR IN THE MANUFACTURE OF A MEDICAMENT FOR INHIBITING FIBROSIS
CROSS-REFERENCE TO RELATED APPLICATIONS The present application claims the priority of Chinese invention patent application No.
CN202010115236.5, filed on February 25, 2020, which is hereby incorporated by
reference in its entirety.
FIELD OF THE INVENTION The present invention relates to the field of genes, and in particular to the use of a
PAX4 inhibitor in the manufacture of a medicament for inhibiting fibrosis.
BACKGROUND OF THE INVENTION Cardiac fibrosis is an important constituent part of most cardiac pathological
conditions. Cardiac fibrosis is manifested by excessive deposition of an extracellular
matrix in a cardiac tissue, which leads to the destruction of a structure of a
physiological cardiac tissue and finally leads to heart failure, seriously threatening the
health and life of human beings. The transdifferentiation of fibroblasts into
myofibroblasts is the key to the initiation and maintenance of cardiac fibrosis. The
myofibroblasts have an important contraction and secretion functions and are
characterized by the expression of a-smooth actin (aSMA), fibronectin, and collagen I
(Col I). Angiotensin II is a polypeptide formulation recognized in the current research
that regulates vasoconstriction, affects cardiac functions, and can induce cardiac
fibrosis. This formulation can usually be used for treating mice or fibroblasts to
simulate the state of cardiac fibrosis. Up to now, although researchers from various countries have done a lot of research on the mechanism of cardiac fibrosis, its specific molecular mechanism remains unclear. Cardiac fibrosis is still an important target in the current clinical treatment of heart diseases. Because the treatment of cardiac fibrosis can delay the occurrence and development of heart failure, it is an important problem to be solved to fill the gap still existing in the research mechanism of cardiac fibrosis.
A transcription factor PAX4 is a member of a subfamily IV of a Paired box (PAX)
family. In the human genome, a PAX4 gene is located in subband 1 of band 2 of
region 3 on a long arm of chromosome 7 and consists of 12 exons and 11 introns.
PAX4 is also known as KPD and MODY9. A PAX4 protein is in nucleus and can form
a transcription factor complex, which binds with the 5'-terminal specific sequence
of the target genes, and regulates the expression of downstream target genes. Current
research believed that those members of the PAX family play an important role in
many stages of embryonic development and organ formation, and also play a role in
every aspect of the body in adulthood. The members of the PAX family, from insects,
amphibians, birds, and mammals, have a considerably conservative sequence
evolution process. The full-length PAX4 includes 349 amino acids, and its protein
structure includes one bipartite paired domain (PD) of 128 amino acids and one
homeodomain (HD). Its C-terminal not only has a common transcriptional activation
domain of the PAX family, but also has a unique negative regulatory domain. Current
research believed that those members of the PAX family are important regulatory
factors of tissue development and cell differentiation. However, the research on PAX4
mainly focuses on research related to a pancreas islet, cancer, and retina. Current
research has shown that PAX4 is involved in the differentiation of islet beta cells and
delta cells during the period of embryonic development and insulin secretion under
normal conditions. Deletion of PAX4 induces type 1 and type 2 diabetes. In tumor-related research, PAX4 is a potent tumor-inhibiting factor of human insulinoma and melanoma. PAX4 can facilitate the migration and invasion of human epithelial carcinoma. Some researchers have reported that PAX4 is highly expressed in a rat retinal photoreceptor, suggesting that PAX4 may play a role in the retina. However, the research on the PAX4 function in the heart is still unknown.
SUMMARY An objective of the present invention is to provide a novel drug for inhibiting fibrosis.
To realize the objective of the present invention, the following technical solution is
intended to be employed: one of the objectives of the present invention is to provide
use of a PAX4 inhibitor in the manufacture of a medicament for inhibiting fibrosis.
In another aspect of the present invention, the present invention further relates to a
drug for inhibiting fibrosis, including a PAX4 inhibitor.
In another aspect of the present invention, the present invention further relates to a
method for inhibiting fibrosis, which is characterized by including a step of inhibiting
the expression of a PAX4 gene.
The inventor of the present application has found through researches by means of
biochemistry, molecular biology and cytology that PAX4 is highly expressed in a
pathological mouse models such as a mouse fibrosis model, a myocardial infarction
model and the like. Subsequently, a technical means of small interfering RNA is
utilized to knock down and reduce the protein level of PAX4 in a cell or cardiac tissue
by the small interfering RNA, respectively by transfecting fibroblasts with the small
interfering RNA or injection of the small interfering RNA via mouse tail vein, so as
to clarify the effect of PAX4 on the heart function of the mouse. Thereafter, a bioinformatics means is utilized to analyze and predict a target gene involved in cardiac fibrosis that can be bound and regulated by PAX4. Thereafter, the regulation effect of PAX4 on downstream genes is clarified by a method of knocking down
PAX4 with the small interfering RNA, thereby clarifying the mechanism of action.
This discovery is made by the inventor of the present application for the first time and
is unexpected.
In one preferred embodiment of the present invention, the PAX4 inhibitor comprises
siRNA and a PAX4 gene knockout reagent.
In one preferred embodiment of the present invention, the PAX4 gene knockout
reagent is siRNA, i.e., small interfering RNA.
In one preferred embodiment of the present invention, the fibrosis refers to cardiac
fibrosis, pancreatic fibrosis, or pulmonary fibrosis.
Although the findings of the present invention are preferably applied to cell fibrosis of
human or animal bodies, the present invention further relates to use of a PAX4 gene or
an expression product thereof in promoting the proliferation of cardiac fibroblasts in
vitro.
Although the findings of the present invention are preferably applied to cell fibrosis of
human or animal bodies, the present invention further relates to use of a PAX4 gene
knockout reagent in inhibiting the proliferation of cardiac fibroblasts in vitro.
Although the findings of the present invention are preferably applied to cell fibrosis of
human or animal bodies, the present invention further relates to use of a PAX4 gene knockout reagent in inhibiting or blocking the expression of a fibrosis promoting factor TGF Pand promoting the expression of fibrosis inhibiting factors IL1R2 and
TGIF2 in cardiac fibroblasts in vitro.
Although the findings of the present invention are preferably applied to cell fibrosis of
human or animal bodies, the present invention further relates to use of a PAX4 gene
knockout agent in inhibiting cell fibrosis by inhibiting a fibrosis promoting factor
TGF Pand promoting fibrosis inhibiting factors IL1R2 and TGIF2. The experimental
results of the present application have confirmed that after PAX4 is knocked down in
cardiac fibroblasts, the level of TGFP protein is decreased, while the levels of IL1R2
and TGIF2 proteins are increased, which suggests that PAX4 plays a
multi-dimensional role in promoting fibrosis by increasing the fibrosis promoting
factor TGFP and inhibiting the fibrosis suppressing factors IL1R2 and TGIF2.
The present invention provides a novel application of PAX4 as a new important target
for treating cardiac fibrosis for the first time. In particular, in the present invention, it
has been found by using biochemical, molecular biological and cytological method
that PAX4 is highly expressed in pathological mouse model such as a mouse
fibroblast fibrosis model, a myocardial infarction model and the like. Subsequently, a
technical means of small interfering RNA is utilized to knock down and reduce the
protein level of PAX4 in a cell or cardiac tissue by the small interfering RNA,
respectively by transfecting fibroblasts with the small interfering RNA or injection of
the small interfering RNA via mouse tail vein, so as to clarify the effect of PAX4 on
the heart function of the mouse. Thereafter, a bioinformatics means is utilized to
analyze and predict a target gene involved in cardiac fibrosis that can be bound and
regulated by PAX4. Thereafter, the regulation effect of PAX4 on downstream genes is
clarified by a method of knocking down PAX4 with the small interfering RNA, thereby clarifying the mechanism of action. Therefore, PAX4 is a potential brand-new important target for treating cardiac fibrosis and thus preventing heart failure.
BRIEF DESCRIPTION OF THE DRAWINGS Fig. 1: Immunohistochemical assay analysis confirms that PAX4 expression increased
under the pathological stimulation of fibroblasts. Fig. 1A: The protein level and
localization of PAX4 labeled with a PAX4 antibody immunohistochemically. Fig. IB:
Immunohistochemical results of quantitative IOD analysis of PAX4 labeled with the
PAX4 antibody.
Fig. 2: Western blot confirms that the expression level of a transcription factor PAX4
is increased in a mouse fibrosis model. Fig. 2A: Comparison of the protein level of
PAX4 in cardiac tissues of a cardiac fibrosis model and its control group by utilizing a
PAX4 antibody through Western blot, with GAPDH as an internal reference. Fig. 2B:
Quantitative and statistical analysis results of the protein content detected by Western
blot of PAX4.
Fig. 3: Detection of the protein levels of cardiac fibrosis markers fibronectin, aSMA
and Col I in a mouse fibrosis model by Western blot. Fig. 3A: Comparison of the
protein levels of fibronectin, aSMA and Col I in cardiac tissues of a cardiac fibrosis
model and its control group by utilizing antibodies against fibronectin, aSMA and Col
I through Western blot, with GAPDH as an internal reference. Fig. 3B: Quantitative
and statistical analysis results of the protein content detected by Western blot of
fibronectin, aSMA and Col I.
Fig. 4: Detection of the protein level of PAX4 in cardiac tissues of an infarct region, a
border region, and a remote region from the mouse myocardial infarction model at different time points after infarction by Western blot.
Fig. 5: Detection of the changes in fluorescence intensities of transcription factor
PAX4 and myofibroblast markers fibronectin, aSMA and Col I after cardiac
fibroblasts are stimulated with 1 M of angiotensin II for three days by an
immunofluorescence assay. Fig. 5A: The fluorescence intensities of the transcription
factor PAX4 and the myofibroblast markers fibronectin, aSMA and Col I, wherein
circled fluorescence indicates nuclei, and the rest of fluorescence indicates detected
genes of interest. Fig. 5B: Relative quantitative and statistical analysis results of
immunofluorescence.
Fig. 6: Western blot confirms that the expression level of transcription factor PAX4 is
increased under the stimulation of angiotensin II in fibroblasts. Fig. 6A: Detection of
the protein level of PAX4 after stimulation with angiotensin II by a Western blot assay.
Fig. 6B: Quantitative and statistical analysis results of the protein content of PAX4
detected by Western blot.
Fig. 7: Western blot confirms that the protein levels of myofibroblast markers
fibronectin, aSMA and Col I are increased under the stimulation of angiotensin II in
fibroblasts. Fig. 7A: Detection of the protein levels of fibronectin, aSMA and Col I
after stimulation with angiotensin II by a Western blot assay. Fig. 7B: Quantitative
and statistical analysis results of the protein contents of fibronectin, aSMA and Col I
detected by Western blot.
Fig. 8: The protein level of PAX4 in a cardiac tissue is detected by Western blot after
PAX4 is knocked down in mice transfected with small interfering RNA by tail vein
injection to verify the PAX4 knock-down efficiency. Fig. 8A: Detection of the protein level of PAX4 after injection of the small interfering RNA into the tail vein of the mouse by a Western blot assay. Fig. 8B: Quantitative and statistical analysis results of the protein content of PAX4 detected by Western blot.
Fig. 9: Small interfering RNA of PAX4 was injected into mice via tail vein and then
fibrosis model was established by implanting a micro-osmotic pump infused with
AnglI. And it is found that knocking down PAX4 inhibits fibrosis by staining with
Picrosirius red. Fig. 9A: Results of tissue sections stained with Picrosirius red. Fig. 9B:
Quantitative and statistical analysis results of cardiac fibrosis area.
Fig. 10: Small interfering RNA of PAX4 was injected into mice via tail vein and then
fibrosis model was established by implanting a micro-osmotic pump infused with
AnglI. the fluorescence intensity of Col I is detected by an immunofluorescence
staining. The upper figure shows the fluorescence intensity of Col I, and the lower
figure shows co-staining of Col I with a nuclear dye Hoechst.
Fig. 11: Small interfering RNA of PAX4 was injected into mice via tail vein and then
fibrosis model was established by implanting a micro-osmotic pump infused with
AnglI. and it is found that knocking down PAX4 has a protective effect on the heart
function of the mice by echocardiography. Fig. 11A: left ventricular posterior wall
thickness, Fig. 1IB: ejection fraction (EF) value, Fig. 1IC: fraction shortening (FS)
value, and Fig. I1D: the ratio of the peak flow velocities during early diastole and the
peak early diastolic velocities of the mitral valve ring E/E'.
Fig. 12: The protein level of PAX4 in cardiac fibroblasts transfected with small
interfering RNA is detected by Western blot to verify the knock-down efficiency in
knocking down PAX4.
Fig. 13: The protein expression levels of myofibroblast markers fibronectin (Fig. 13A),
aSMA (Fig. 13B) and Col I (Fig. 13C) are detected by immunofluorescence staining
after PAX4 is knocked down infibroblasts transfected with small interfering RNA to
verify the inhibitory effect of knocking down PAX4 on fibrosis.
Fig. 14: The protein expression levels of myofibroblast markers fibronectin, aSMA,
and Col I are detected by Western blot after PAX4 is overexpressed infibroblasts
infected with adenovirus to verify the promoting effect of PAX4 onfibrosis.
Fig. 15: Detection of the effect on the protein expression levels of possible
downstream genes TGFP, ILR2 and TGIF2 after PAX4 is knocked down in
fibroblasts transfected with small interfering RNA by a Western blot assay.
DETAILED DESCRIPTION The present invention will be described in further detail below with reference to
specific examples, which are explanations rather than limitations of the present
invention. The methods and related reagents used in the present invention can have
other optional and alternative solutions, as long as the same technical results can be
achieved.
The experimental methods in the following examples which are not specified with
specific conditions shall be carried out according to conventional operations in the art
to which the present invention belongs or the conditions suggested by a manufacturer.
Example 1 Assay of animal pathological model: a mouse cardiac fibrosis model was
established, a cardiac tissue was taken, and the expression location and content of
PAX4 protein, as well as the content of myofibroblast markers fibronectin and aSMA,
and the content of an extracellular matrix Col I were detected by utilizing
immunohistochemistry and a Western blot assay.
Preparation of angiotensin II-induced mouse cardiac fibrosis model: 10-week-old
male C57BL/6 mice were randomly divided into two groups, an operation group, and
a sham operation group. The mice were implanted with a micro-osmotic pump (Alzet
MODEL 1007D, DURECT, Cupertino, CA) injected with angiotensin (3 mg kg-I day-1)
for 7 days to establish the fibrosis model. Preparation of the micro-osmotic pump: 1
day before operation, angiotensin II (dissolved in a sterile PBS buffer) was injected
into the miniosmotic pump with a 1 mL syringe, and the micro-osmotic pump was
soaked in the sterile PBS buffer and equilibrated at 37°C overnight. During the
operation, the mice were anesthetized with 2%-3% isoflurane, and a transverse
incision of about 0.7 cm in length was cut in a back neck of the mouse. A tweezer was
inserted into the skin for blunt dissection of a subcutaneous tissue. The miniosmotic
pump was implanted, the wound was sutured, and neomycin ointment was applied
onto the wound to prevent infection. The operation group received continuous
infusion of angiotensin IIat a concentration of 3 mg kg-1 day-' for 7 days.
Assay method of immunohistochemistry of PAX4: a sample was fixed with
paraformaldehyde, embedded in paraffin, and then sliced. (1) Dewaxing: tissue
sections were treated with xylene for 15 min 3 times, with 100% ethanol for 5 min
twice, with 95% ethanol for 5 min twice, and with 80% ethanol for 5 min once.
Finally, the tissue sections were washed with distilled water for 2 min. (2) Removal of
endogenous catalase: the sections were placed in a 3% hydrogen peroxide solution
(formulated with 100% methanol) for 10-15 min to remove endogenous catalase.
Then the sections were washed with PBS for 3 times, with 5 minutes each time. (3)
Antigen thermal repair: the sections were placed in a citrate (pH 6.0) antigen repair
solution, and subjected to thermal repair in a pressure cooker, such that an antigen
epitope was fully unfolded (timing for 2 minutes after the pressure cooker
continuously emitted gas). After the thermal repair was completed, the sections were
placed at room temperature, and when the temperature of the sections was reduced to
room temperature, the sections were washed with PBS for 3 times, with 5 minutes
each time. (4) Serum blocking: the sections were placed in a wet box and blocked
with 10% goat serum at room temperature for 30 min. (5) Incubation with primary
antibody: the serum was discarded, and then a formulated primary antibody was
added onto the sections, which was kept overnight in a refrigerator at 4°C (or
incubated in an incubator at 37°C for 2 hours). (6) Incubation with secondary
antibody: the wet box was taken out from the refrigerator at 4°C, and naturally raised
to room temperature, and then the sections were washed with PBS for 3 times, with 5
minutes each time. Then, the second antibody labeled with horseradish peroxidase
(Beijing Zhong Shan-Golden Bridge Biological Technology Co., Ltd., a rabbit
two-step kit) was added, and incubation was carried out at room temperature for 30
min. (7) DAB color development: the sections were washed with PBS for 3 times and
subjected to color development with a DAB color developing solution. Formulation
method of a DAB working solution: 1 mL of a DAB diluent + 50 L of the DAB color
developing solution. (8) Re-staining of nucleus: the sections were first soaked in
distilled water for 2 min, then placed in a hematoxylin solution for 30 s, and rinsed
with tap water for 3 times. Differentiation was performed in 70% hydrochloric acid in
alcohol for 5-6 s, and the sections were washed with water for 30 s. The sections were
performed in 1% ammonia water for 60 s, and unfixed dyes were washed off in
distilled water. (It could be observed under a microscope whether the nucleus was
blued). (9) Dehydrating, permeabilizing, and mounting with a neutral resin mounting
medium. The sections were treated with 95% ethanol twice for 2 min, with absolute ethanol twice for 2 min, and finally in a xylene substitute twice for 5 min. (10) After dried in the air, the sections were scanned to conduct statistics of a positive area by software.
Extraction of total protein from myocardial tissue: a myocardial tissue stored in liquid
nitrogen was taken, put into a mortar and grounded with liquid nitrogen, and two
thirds (the other third was used for extracting RNAs) of the grounded myocardial
tissue was added into a tissue lysis buffer (20 mmol/L Tris-HCl pH 7.4, 150 mmol/L
NaCl, 2.5 mmol/L EDTA, 50 mmol/L NaF, 0.1 mmol/L Na4 P20 7 , 1 mmol/L Na3VO 4
, 1% Triton X-100, 10% Glycerol, 0.1% SDS, 1% deoxycholic acid, 1 mmol/L PMSF,
and lg/mL aprotinin), mixed uniformly, and allowed to stand on ice for 15 minutes,
wherein 800 microliters of the lysis buffer was added per 50 mg of the myocardial
tissue. The resulting homogenate was collected, ultrasonically crushed (45%, 5 s on, 5
s off for 4 cycles), and then centrifuged at 4°C and 12,000 rpm for 15 minutes. A part
of supernatant was transferred into a new EP tube and cryopreserved at -80°C after
protein quantification, and a quarter of the volume of a 5x loading buffer was added to
another part of supernatant, mixed evenly, boiled at 100°C for 5 minutes, and
cryopreserved for later detection of related proteins by Western blot.
Western blot assay: after electrophoresis with a 10% SDS-PAGE gel, protein was
transferred onto a nitrocellulose membrane, blocked with 5% fat free milk at room
temperature for 1 hour, and incubated with primary antibodies in a cold room at 4°C
overnight. The catalogue number of the primary antibodies were as follows:
Fibronectin (ab2413, abeam, Cambridge, MA, USA), aSMA (ab32575, abeam,
Cambridge, MA, USA), Col 1 (203002, MD Biosciences), PAX4 (ab101721, abeam,
Cambridge, MA, USA), TGFP (10804-MM33, Sino biological, Beijing, China),
IL1R2 (sc-376247, Santa Cruz Biotech, CA, USA), TGIF2 (ab190152, abeam,
Cambridge, MA, USA), and GAPDH (2118S, CST). The membrane was washed with
TBST for three times, and then was incubated with secondary antibodies of
corresponding species at room temperature for 1 hour, and the membrane was washed
with TBST again, and then put into a Chemiluminescent HRP substrate (Millipore
Corporation). Then the membrane was put into a luminescence detection machine for
exposure. The band intensity was quantified by using NIH ImageJ software.
A mouse cardiac fibrosis model was established with 10-week-old male C57BL/6
mice in the manner of implanting a micro-osmotic pump infused with angiotensin II.
The expression level of PAX4 in the cardiac tissue of the cardiac fibrosis model was
detected, and meanwhile the levels of fibrosis markers were detected to correspond to
the degree of fibrosis in the cardiac tissue.
Firstly, cardiac tissues were taken from the cardiac fibrosis model established by
implanting the micro-osmotic pump infused with angiotensin II and the sham
operation group and subjected to an immunohistochemical assay with a PAX4
antibody. The assay results were shown in Fig. 1A, and in the tissue in which cardiac
fibrosis occurred, the protein level of PAX4 was increased in both cardiac myocytes
and cardiac fibroblasts. The statistical results were shown in Fig. IB, and the
statistical analysis of the quantitative results showed that the protein level of PAX4 in
the fibrotic cardiac tissue was significantly increased compared with that in a healthy
cardiac tissue.
Thereafter, a Western blot assay was carried out by using the total proteins of the
cardiac tissues from the cardiac fibrosis model and the sham operation group. The
assay results of detecting the protein level of PAX4 were shown in Fig. 2. The
obtained results were consistent with those of the immunohistochemical assay, and the protein level of the transcription factor PAX4 was significantly increased in the fibrotic cardiac tissue (Fig. 2A). The statistical analysis of the quantitative results (Fig.
2B) showed that, the protein level of PAX4 in the fibrotic cardiac tissue was
significantly increased compared with that in a healthy cardiac tissue.
Meanwhile, the protein levels of the fibrosis markers fibronectin, aSMA and Col I in
the cardiac tissue were also detected by the Western blot assay. The assay results were
shown in Fig. 3A, the protein levels of thefibrosis markersfibronectin, aSMA and
Col I in the fibrotic cardiac tissue were significantly increased than those in a normal
cardiac tissue. The statistical analysis of the quantitative results (Fig. 3B) showed that
the protein levels of fibronectin, aSMA and Col I in the fibrotic cardiac tissue was
significantly increased. However, whether there was a causal relationship between the
increase of PAX4 and the increase of the fibrosis markers fibronectin, aSMA and Col
I still needed to be verified by subsequent experiments.
Example 2 Detection of the protein levels of PAX4 in an infarct region and a remote
region of a cardiac tissue of a mouse myocardial infarction model at different time
points after operation.
-week-old male C57BL/6 mice were used for establishing a myocardial infarction
model. The mice were randomly divided into a myocardial infarction group and a
sham operation control group. The myocardial infarction group was subjected to left
coronary artery stenosis to induce the occurrence of myocardial infarction. The
control group was subjected to sham operation. The operation was performed under
gas anesthesia, and the mice were subjected to gas anesthesia by inhaling 2%
isoflurane. In the assay group, an infarct region, a border region, and a remote region
of cardiac tissues of the mice were taken to collect the cardiac tissues on days 1, 4 and
7 after the operation, respectively.
The protein level of an endogenous transcription factor PAX4 in the cardiac tissue in
the infarct region, the border region, and the remote region on days 1, 4 and 7 after the
operation of the mouse myocardial infarction model was detected by Western blot. A
left side of Fig. 4 showed the specific locations of the infarct region, the border region,
and the remote region of heart infarction. The assay results showed that, compared
with the sham operation group, the protein levels of PAX4 in all areas of myocardial
infarction operation were increased in different degrees. The results of the myocardial
infarction model in combination with the results of the pathological conditions of
fibrosis in Example 1, suggested that the protein level of PAX4 in the cardiac tissue
was increased when pathological changes occurred in the heart, and the specific
function of this increase was verified in the following subsequent assays.
Example 3 Detection of the protein levels of PAX4, and fibrosis markers fibronectin,
aSMA and Col I by immunofluorescence and a Western blot assay after cardiac
fibroblasts were treated with 1 M angiotensin for three days.
Isolation and culture of cardiac fibroblasts from adult mice: male 8 weeks old
C57/BL6 micewere sacrificed by cervical dislocation, and quickly soaked in 75%
alcohol for about half a minute, and immediately chests of the mice were opened to
take out hearts on an ultra-clean workbench. The hearts were put into a PBS buffer at
4°C to be washed twice, then blood vessels in the atrium and the heart bottom were
cut off, and then ventricles were chopped into small pieces and washed with PBS once
to wash away some residual blood. 0.1% typeII collagenase (330U, Worthington,
Columbia, NJ, USA/Sigma, St. Louis, MO, USA) formulated in a PBS balanced salt
solution was added for digestion. The whole digestion process was carried out at a constant temperature of 36-37°C under stirring. A supernatant of the resulting digestion solution was taken every 8 minutes of the digestion and added into the same amount of a DMEM culture medium containing 10% FBS, followed by uniform mixing. This process was repeated for about 7-8 times until the tissue pieces were completely digested. Several tubes of collected cells were centrifuged at room temperature at 1,000 rpm for 5 minutes, the supernatant was discarded, and the cells were resuspended with a DMEM culture medium containing 10% FBS. The suspension of cardiac myocytes obtained each time was mixed, seeded in a culture dish with a diameter of 100 mm, and placed in a 37°C incubator with 5% C02 for 2 hours to make the fibroblasts basically adherent. The culture medium in the culture dish was pipetted and discarded, and a fresh DMEM culture medium containing 10%
FBS was added to continue the culture. After 3 days, the cells were fully grown and
passaged for subsequent assays.
A method of extracting protein from cardiac fibroblasts: the cells were first digested
from a base gel with pancreatin and centrifuged, and then the supernatant was taken,
washed with cold PBS for three times, subsequently subjected to cell lysis with a cell
lysis buffer (20 mmol/L Tris-HCl pH 7.4, 150 mmol/L NaCl, 2.5 mmol/L EDTA, 50
mmol/L NaF, 0.1 mmol/L Na4 P20 7 , 1 mmol/L Na3VO 4 , 1% Triton X-100, 10%
glycerol, 0.1% SDS, 1% deoxycholic acid, 1 mmol/L PMSF, and 1 mg/mL aprotinin),
subjected to ultrasonic crushing, and then centrifuged at 12,000 g under the condition
of 4°C for 15 minutes. The supernatant was collected. 5 microliters of the supernatant
were taken for protein quantification, and a 5x gel loading buffer was added into the
rest of the supernatant to be kept at 100°C for 5 minutes to ensure protein
denaturation.
Immunofluorescence staining assay: the cells were fixed with warm 4% paraformaldehyde of 37°C under the condition of 37°C for 15 minutes, then washed with warm PBS for 3 times, and subsequently permeabilized with 0.2% Triton X-100 for 20-30 minutes. The cells were washed with warm PBS for 3 times, and then a blocking solution (5% BSA) was added for blocking for 30 minutes. Thereafter, the cells were incubated with primary antibodies against aSMA (ab32575, abeam,
Cambridge, MA, USA), fibronectin (ab2413, abeam, Cambridge, MA, USA) and
PAX4 (ab101721, abeam, Cambridge, MA, USA) under a condition of 4°C overnight.
After the primary antibodies were recovered and stored, the cells were washed with
PBS for 3 times, and then incubated with a secondary antibody Alexa Fluor 488 at
room temperature for 1 hour. The cells were subjected to nuclear staining with
Hoechst (Invitrogen, Carlsbad, CA, USA) under the condition of room temperature
for 8 minutes. Statistics and analysis of the fluorescence intensity were conducted by
using a Morphology Explorer BioApplication module of a high-content screening
imaging system Cellomics ArrayScan VTI HCS Reader (Thermo Fisher Scientific,
Rockford, IL, USA).
At the cellular level, primary mouse cardiac fibroblasts were extracted, cultured in a
12-well plate to a P2 generation, and stimulated with angiotensin II at a concentration
of 1 M for three days, and then samples were collected. Firstly, the samples were
fixed, and detected for the protein levels of endogenous PAX4 and fibrosis markers
fibronectin, aSMA and Col I by immunofluorescence. The fluorescence circled in Fig.
A were positions of nuclei, and the rest of fluorescence indicated the locations and
contents of specific proteins of interest (PAX4, fibronectin, Col I and aSMA)
identified by specific antibodies, respectively. It could be seen from the assay results
that the transcription factor PAX4 was mainly expressed in the nucleus. The assay
results indicated that under the stimulation of angiotensin II, the fluorescence
intensities of PAX4, fibronectin, aSMA and Col I were enhanced in different degrees.
The quantitative results in Fig. 5B showed that the contents of the transcription factor
PAX4, and common cardiac fibrosis markers fibronectin, aSMA and Col I were
increased significantly after three days of stimulation with angiotensin II.
Thereafter, a western blot assay was conducted with the total protein of cardiac
fibroblasts cultured under the same conditions, and the protein level of PAX4 was
detected first. The assay results were shown in Fig. 6. The assay obtained results close
to those of the immunofluorescence assay, and the protein level of the transcription
factor PAX4 was increased after the stimulation with angiotensin II (Fig. 6A). The
statistical analysis of the quantitative results showed that the increase degree of the
protein level was significant. Meanwhile, the protein levels of the myofibroblast
markers fibronectin, aSMA and Col I were also detected (Fig. 7). The assay results
showed that the stimulation of the cardiac fibroblasts with angiotensin II for three
days could promote the protein levels of the fibrosis markers fibronectin, aSMA and
Col I (Fig. 7A). The statistical analysis of the quantitative results showed that after
stimulation with angiotensin II, the protein levels of the fibrosis markers were
significantly increased compared with those of the control group.
Example 4 The level of PAX4 in mice was knocked down in the manner of injecting
small interfering RNA into tail veins of mice, and then a fibrosis model was
established. The effects of PAX4 on the heart function and on the protein levels of
myofibroblast markers fibronectin and aSMA, and an extracellular matrix Col I were
detected by echocardiography, comparison of a heart weight/body weight ratio,
staining with Picrosirius red (PSR), a Western blot assay and an immunofluorescence
assay.
Tail vein injection of small interfering RNA into mice: wild C57BL/6 adult mice (11 weeks old, male, about 27 g) were selected, and administrated with 10 nmol of small interfering RNA sequence for knock down of PAX4 (a chemically modified sequence, including a knock-down sequence and a nonsense control sequence) (Ribobio Co., Ltd
(Guangzhou, China)) by tail vein injection every day, at an injection volume of 0.12
mL (dissolved in normal saline) each time for 3 days. On day 4, the infusion of
angiotensin II was started with the aforementioned pump implantation method. From
day 5, the control sequence and the small interfering RNA sequence for knock down
were injected into tail veins of the mice every other day until angiotensin II had been
continuously infused for 7 days. The ultrasonic indexes, body weights and heart
weights of the mice were measured, and myocardial tissues were collected for
subsequent detection.
Echocardiography: the mice were put into an anesthesia box and given isoflurane
(2.5% isoflurane, 0.8 L/min) for anesthesia. After anesthesia was completed, the mice
were taken out of the anesthesia box, quickly put on a heating plate in a supine
position and worn a nasal mask connected with an anesthetic. The limbs of the mice
were fixed with an adhesive tape, and the isoflurane concentration was adjusted to 1%
to maintain anesthesia. Hair removal was performed on the chest with depilatory
cream (Nail, Canada). An echocardiogram of the mice was detected by a Vevo 2100
ultrasonic instrument (Fujifilm Visual sonics, Canada).
Weighing of the heart: after blood sampling was completed, the chest of each mouse
was quickly cut open, and the heart was lavaged with cold PBS (0.8% NaCl, 0.02%
KCl, 0.02% KH 2 PO 4 , and 0.4% Na2HPO 4) through an indwelling needle. After that, a
whole heart was taken out, tissues such as blood vessels, fat and the like on the heart
were cut off in cold PBS, the heart was dried with filter paper, and a whole heart
weight (HW) was weighed. After weighing, the auricle on the heart was cut off, leaving a whole ventricle. A transection part of the papillary muscle in the middle of the heart was taken with a surgical blade and fixed in 4% paraformaldehyde (W/V%, formulated in PBS) for preparation of tissue sections. The rest of the myocardial tissues was cryopreserved in liquid nitrogen in a cryopreservation tube.
Tissue sections and staining: a horizontal cross section of the papillary muscle of the
heart was fixed in a 4% paraformaldehyde solution (W/V%, formulated in PBS) for
6-8 hours, then paraformaldehyde was discarded, and the horizontal cross section was
added into a 20% sucrose solution (W/V%, formulated in PBS) for dehydration. Then
the dehydrated horizontal cross section was sequentially put into 70% (3 hours) and
% (3 hours) ethanol solutions for gradient dehydration, and finally put into a
solution of 90% ethanol plus n-butanol (a volume ratio of 1:1) overnight. On the next
day, the dehydrated horizontal cross section was sequentially put into a solution of
% ethanol plus n-butanol (45 minutes twice), n-butanol (30 minutes) and butanol
(20 minutes). The surface liquid of the dehydrated horizontal cross section was
absorbed by filter paper, and a tissue mass was embedded with paraffin. After that,
paraffin heart sections with a thickness of 5 m were made with a microtome by
transecting horizontally at the papillary muscle, and the sections were stained with
Picrosirius red to detect collagen deposition. First, dewaxing was conducted in xylene
for 10 minutes for 3 times, in 100% ethanol for 3 minutes twice, in 95% ethanol for 3
minutes twice, in 80% ethanol for 3 minutes once, and in 70% ethanol for 3 minutes
once. Finally, the sections were washed with distilled water for 3 times. Thereafter
staining with Picrosirius red was conducted, wherein water was removed by dipping
first, and the sections were put into a Picrosirius red solution for staining for 1 minute,
washed with distilled water to remove unfixed dyes (for 3 times), quickly washed
once with 95% ethanol, then put into 100% ethanol to be washed for 1 minute twice
(be careful not to wash off yellow stained color), and finally, subjected to permeabilization treatment with 80% xylene (for 10 minutes twice), and covered with neutral resin on the surface thereof, followed by covering with a cover slip for mounting. Finally, observation and quantitative analysis of the tissue sections were conducted. A collagen fibrosis area (stained with Picrosirius red) was analyzed and quantified with a NanoZoomer-SQ (Hamamatsu, Japan) image analysis system. The sections stained with Picrosirius red were observed, and each specimen was measured for the collagen fibrosis area (red stained parts) and a cross-sectional area of the whole heart, and the percentage of fibrosis was obtained by dividing the fibrosis area by a total heart area.
After the indexes such as echocardiography were detected, the body weight was
weighed, and the cardiac tissue was collected and weighed. Part of the cardiac tissues
was taken to conduct tissue embedding of frozen sections and paraffin sections for
preparation for the subsequent immunohistochemical assay, and a protein sample of
the cardiac tissue was collected for a Western blot assay.
Firstly, the protein knock-down efficiency of PAX4 was detected (Fig. 8), and
meanwhile the effect of angiotensin II stimulation on the protein level of PAX4 was
detected. The assay results showed that the protein knock-down efficiency of PAX4
was good, and the expression of PAX4 protein in the myocardium of the mice injected
with the nonsense sequence was increased after administration of angiotensin II,
which was consistent with the assay results in the Example 1 above. The statistical
analysis of the quantitative results showed that angiotensin II significantly promoted
the PAX4 protein level, and the protein level after knocking down was one third of
that before knocking down (Fig. 8B).
Secondly, the results of staining with Picrosirius red were shown in Fig. 9. In the mice injected with the nonsense sequence, after administration of angiotensin II, a collagen area representing cardiac fibrosis was increased. There was no significant change in the cardiac fibrosis area between the mice injected with the small interfering RNA of
PAX4 and the mice injected with the nonsense sequence. The cardiac fibrosis area of
the mice injected with the small interfering RNA of PAX4 and then administrated
with angiotensin II was increased compared with that of the mice injected with the
small interfering RNA of PAX4 and not administrated with angiotensin II; and was
decreased compared with that of the mice injected with the nonsense small interfering
RNA and then administrated with angiotensin II (Fig. 9A). The statistical analysis of
the quantitative results of the cardiac fibrosis area of the mice showed that knocking
down PAX4 could significantly inhibit cardiac fibrosis induced by angiotensin II (Fig.
9B).
An immunofluorescence assay of Col I was carried out with frozen sections of the
cardiac tissues (Fig. 10), and the assay results were consistent with those of staining
with Picrosirius red. The red fluorescence intensity of Col I in the cardiac tissues of
mice injected with the nonsense sequence was increased after administration of
angiotensin II, and the fluorescence intensity of Col I in the cardiac tissues of the mice
injected with the small interfering RNA of PAX4 and then administrated with
angiotensin II was decreased significantly.
The results of echocardiography were shown in Fig. 11. Knocking down PAX4 could
significantly reduce the increase of the left ventricular posterior wall thickness
(LVPWD; d) induced by angiotensin II (Fig. 11A), could significantly improve the
decrease of EF caused by angiotensin II (Fig. 11B), had the trend of improving the
decrease of FS caused by angiotensin (Fig. 11C), and could significantly improve the
increase of E/E' caused by angiotensin II (Fig. 11D). The aforementioned echocardiographic results suggested that knocking down PAX4 could have a protection effect on both the systolic and diastolic functions of the heart of the mouse.
In summary of the aforementioned assay results, it was suggested that knocking down
PAX4 could have the effects of relieving the increase of the extracellular matrix,
inhibiting the occurrence of fibrosis and improving the heart function.
Example 5 Interfering the protein level of PAX4 at the level of cardiac fibroblasts.
The effect on the protein levels of myofibroblast markers fibronectin, aSMA and Col I
was detected by an immunofluorescence assay or a Western blot assay.
Transfection of small interfering RNA into cardiac fibroblasts: the cardiac fibroblasts
of P2 generation were passaged from a P1 generation into a 6-well plate on the night
before the assay of transfection of the small interfering RNA and cultured overnight in
an environment of 37°C and 5% C02 to ensure that the morphology of the fibroblasts
was spread and any extracellular matrix that affected the transfection efficiency had
not been produced. On the day of transfection, the fibroblasts in the 6-well plate were
washed with warm PBS at 37°C gently and repeatedly for three times to ensure that
the medium was thoroughly washed away. Then, 500 L of OPTI-MEM (Opti-MEM I
Reduced Serum Medium, 31985070, Life) was added into each well, and then 80
nmol/L of small interfering RNA of PAX4 (sc-152040, Santa Cruz Biotech, CA, USA)
or control small interfering RNA (sc-37007, Santa Cruz Biotech, CA, USA) and 3 L
of a HiPerFect transfection reagent (301705, QIAGEN, Beijing, China) were added
respectively. 6 hours after the transfection, a DMEM medium containing 10% fetal
bovine serum was added for culture. The assay of protein detection used cell samples
that had been transfected with the small interfering RNA for three days.
PAX4 of cardiac fibroblasts infected with adenoviruses: the cardiac fibroblasts of P2
generation were passaged from a P1 generation into a 6-well plate on the night before
the assay of adding virus for infection, and cultured overnight in an environment of
37°C and 5% C02 to ensure that the morphology of the fibroblasts was spread and
any extracellular matrix that affected the transfection efficiency had not been
produced. When the cells were grown to a density of 60%, cells were infected with
2MOI PAX4 or control adenoviruses under a serum-free condition. After 6 hours, the
medium was replaced by DMEM containing 10% fetal bovine serum, and culture was
conducted for three days for subsequent assays.
To further determine the role of PAX4 in the cardiac fibroblasts, firstly, an assay was
designed to knock down PAX4 by transfecting the small interfering RNA of PAX4
into the cardiac fibroblasts, and the Western blot assay had confirmed that knock
down of PAX4 in the cardiac fibroblasts was effective (Fig. 12). Thereafter, the
fluorescence intensities of the myofibroblast markers fibronectin, aSMA and Col I
after knocking down of PAX4 were detected by an immunofluorescence assay. The
assay results showed that the fluorescence of the myofibroblast markers fibronectin
(Fig. 13A), aSMA (Fig. 13B) and Col I (Fig. 13C) was decreased after knocking
down of PAX4.
Three days after over-expression of PAX4 in the fibroblasts, a Western blot assay was
carried out. The assay results were shown in Fig. 14. After the over-expression of
PAX4, all the protein levels of the myofibroblast markers fibronectin, aSMA and Col
I were increased in different degrees.
All the aforementioned assays suggested that PAX4 could promote the occurrence of
cardiac fibrosis, and knocking down PAX4 could reduce the contents of the fibrosis markers fibronectin, aSMA and Col I.
Example 6 In the cardiac fibroblasts, the protein level of PAX4 in the cells was
knocked down, and thus the expression level of its downstream regulatory gene was
detected by means of a Western blot assay.
Both the animal experiment and the cell assay suggested that knocking down PAX4
could have a protection effect on the heart function. However, the mechanism of
action was still unclear. Therefore, we used bioinformatics analysis to find three
possible effect targets that affected cardiac fibrosis and might be regulated by PAX4:
TGFP (promoting fibrosis), and IL1R2 and TGIF2 (fibrosis inhibiting factors).
TRANSFAC predicted that PAX4 could bind to the promoters of TGFP, IL1R2 and
TGIF2, so we detected the protein levels of these three genes upon knock down of
PAX4 by the method of a Western blot assay.
As shown in Fig. 15, after PAX4 was knocked down in the cardiac fibroblasts, the
protein level of TGFP was decreased, while the protein levels of IL1R2 and TGIF2
were increased, which suggested that PAX4 played a multi role in promoting fibrosis
by promoting the fibrosis promoting factor TGFP and inhibiting the fibrosis inhibiting
factors IL1R2 and TGIF2.
The aforementioned specific embodiments are only preferred examples of the present
invention, rather than limiting the present invention, and any modification, equivalent
substitution and improvement within the spirit and principle of the present invention
should be included in the protection scope of the present invention. The above
descriptions are preferred examples of the present invention. It should be noted that a
person of ordinary skill in the art may further make several improvements and modifications without departing from the principle of the present invention, but such improvements and modifications shall also be deemed as the protection scope of the present invention.

Claims (8)

Claims
1. Use of a PAX4 inhibitor in the manufacture of a medicament for inhibiting fibrosis.
2. The use according to claim 1, wherein the PAX4 inhibitor comprises siRNA and a
PAX4 gene knockout reagent.
3. The use according to claim 2, wherein the PAX4 gene knockout reagent is siRNA.
4. The use according to any one of claims 1-3, wherein thefibrosis refers to cardiac
fibrosis, pancreatic fibrosis or pulmonary fibrosis.
5. Use of a PAX4 gene or an expression product thereof in promoting the proliferation
of cardiac fibroblasts in vitro.
6. Use of a PAX4 gene knockout reagent in inhibiting the proliferation of cardiac
fibroblasts in vitro.
7. Use of a PAX4 gene knockout reagent in inhibiting or blocking the expression of a
fibrosis promoting factor TGF Pand promoting the expression of fibrosis inhibiting
factors IL1R2 and TGIF2 in cardiac fibroblasts in vitro.
8. Use of a PAX4 gene knockout agent in inhibiting cell fibrosis by inhibiting a
fibrosis promoting factor TGF Pand promoting fibrosis inhibiting factors IL1R2 and
TGIF2.
AU2021225272A 2020-02-25 2021-02-09 Use of PAX4 inhibitor in preparation of drug for inhibiting fibrosis Active AU2021225272B2 (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
CN202010115236.5A CN111214660B (en) 2020-02-25 2020-02-25 Application of PAX4 gene expression inhibitor in preparation of medicine for inhibiting fibrosis
CN202010115236.5 2020-02-25
PCT/CN2021/076352 WO2021169812A1 (en) 2020-02-25 2021-02-09 Use of pax4 inhibitor in preparation of drug for inhibiting fibrosis

Publications (2)

Publication Number Publication Date
AU2021225272A1 true AU2021225272A1 (en) 2022-09-29
AU2021225272B2 AU2021225272B2 (en) 2024-07-11

Family

ID=

Also Published As

Publication number Publication date
WO2021169812A1 (en) 2021-09-02
CN111214660A (en) 2020-06-02
CN111214660B (en) 2022-01-18

Similar Documents

Publication Publication Date Title
KR102100163B1 (en) Compositions of Prevention or Treatment of Keloid or Hypertrophic scar
Du et al. Pentamethylquercetin protects against cardiac remodeling via activation of Sestrin2
CN112251421B (en) EZH2 variable shear body and application thereof
CN115245567B (en) Application of FGL1 inhibitor in preparation of medicines for preventing and treating myocardial ischemia injury
WO2021169812A1 (en) Use of pax4 inhibitor in preparation of drug for inhibiting fibrosis
CN105457015B (en) Recombinant CREG protein inhibits the medical usage of hypertensive vascular remodeling
CN113372435A (en) Polypeptide for promoting angiogenesis and pharmaceutical application thereof
JP2018512456A (en) Epicardial-derived paracrine factor for repairing heart tissue
AU2021225272B2 (en) Use of PAX4 inhibitor in preparation of drug for inhibiting fibrosis
Jin et al. Effect of miR-9 on myocardial fibrosis in rats via TGF-β1/Smads signaling pathway.
Wang et al. Effects of recombinant human relaxin upon proliferation of cardiac fibroblast and synthesis of collagen under high glucose condition
CN111202847B (en) Application of PAX6 gene or its expression product in preparing medicine for inhibiting fibrosis
KR101829209B1 (en) Cosmetic composition with the extract of Cornus officinalis for the improvement of skin redness or face redness
Yang et al. Vasoactive intestinal peptide in rats with focal cerebral ischemia enhances angiogenesis
CN102008718A (en) Application of SIRT (Silent Mating Type Information Regulation 2 Homolog 1) to prepare medicine used for regulating down the expression of cyclin D1
CN107699616B (en) Application of Fibulin-3 as target in preparation of medicine for preventing and treating psoriasis
CN109295015B (en) Application of E3 ubiquitin ligase TRIM7 in liver cancer
CN114533726B (en) Small molecule drug for inhibiting fibrosis and application thereof
CN109464443A (en) The new opplication of Linifanib
CN114560817B (en) Small molecule drug for inhibiting fibrosis and application thereof
CN113616792B (en) Application of reagent for increasing expression level of smurf1 protein in preparation of medicine for preventing and treating calcified aortic valve diseases
CN109528749A (en) Purposes of the long-chain non-coding RNA-H19 in preparation treatment hypophysis tumor medicine
CN116555411A (en) Application of LncRNA KCND1 in preparation of medicament for preventing and treating pathological myocardial hypertrophy
CN117646064A (en) Application of FAF2 protein in preparation of medicines for treating myocardial hypertrophy
CN116042624A (en) Medical application of knockdown or inhibition of cyclic RNA circ0030586