CN111575365A - ES marker and application thereof - Google Patents

ES marker and application thereof Download PDF

Info

Publication number
CN111575365A
CN111575365A CN202010393259.2A CN202010393259A CN111575365A CN 111575365 A CN111575365 A CN 111575365A CN 202010393259 A CN202010393259 A CN 202010393259A CN 111575365 A CN111575365 A CN 111575365A
Authority
CN
China
Prior art keywords
marker
relative abundance
sample
preset threshold
motif
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.)
Pending
Application number
CN202010393259.2A
Other languages
Chinese (zh)
Inventor
戴勇
汤冬娥
邱晓芬
胡芷洋
周俊
廖秋燕
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.)
Shenzhen Peoples Hospital
Original Assignee
Shenzhen Peoples 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 Shenzhen Peoples Hospital filed Critical Shenzhen Peoples Hospital
Priority to CN202010393259.2A priority Critical patent/CN111575365A/en
Publication of CN111575365A publication Critical patent/CN111575365A/en
Pending legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6876Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes
    • C12Q1/6883Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/68Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids
    • G01N33/6893Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids related to diseases not provided for elsewhere
    • GPHYSICS
    • G16INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR SPECIFIC APPLICATION FIELDS
    • G16BBIOINFORMATICS, i.e. INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR GENETIC OR PROTEIN-RELATED DATA PROCESSING IN COMPUTATIONAL MOLECULAR BIOLOGY
    • G16B25/00ICT specially adapted for hybridisation; ICT specially adapted for gene or protein expression
    • G16B25/10Gene or protein expression profiling; Expression-ratio estimation or normalisation
    • GPHYSICS
    • G16INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR SPECIFIC APPLICATION FIELDS
    • G16BBIOINFORMATICS, i.e. INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR GENETIC OR PROTEIN-RELATED DATA PROCESSING IN COMPUTATIONAL MOLECULAR BIOLOGY
    • G16B25/00ICT specially adapted for hybridisation; ICT specially adapted for gene or protein expression
    • G16B25/20Polymerase chain reaction [PCR]; Primer or probe design; Probe optimisation
    • GPHYSICS
    • G16INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR SPECIFIC APPLICATION FIELDS
    • G16BBIOINFORMATICS, i.e. INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR GENETIC OR PROTEIN-RELATED DATA PROCESSING IN COMPUTATIONAL MOLECULAR BIOLOGY
    • G16B30/00ICT specially adapted for sequence analysis involving nucleotides or amino acids
    • G16B30/10Sequence alignment; Homology search
    • GPHYSICS
    • G16INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR SPECIFIC APPLICATION FIELDS
    • G16HHEALTHCARE INFORMATICS, i.e. INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR THE HANDLING OR PROCESSING OF MEDICAL OR HEALTHCARE DATA
    • G16H20/00ICT specially adapted for therapies or health-improving plans, e.g. for handling prescriptions, for steering therapy or for monitoring patient compliance
    • G16H20/10ICT specially adapted for therapies or health-improving plans, e.g. for handling prescriptions, for steering therapy or for monitoring patient compliance relating to drugs or medications, e.g. for ensuring correct administration to patients
    • GPHYSICS
    • G16INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR SPECIFIC APPLICATION FIELDS
    • G16HHEALTHCARE INFORMATICS, i.e. INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR THE HANDLING OR PROCESSING OF MEDICAL OR HEALTHCARE DATA
    • G16H50/00ICT specially adapted for medical diagnosis, medical simulation or medical data mining; ICT specially adapted for detecting, monitoring or modelling epidemics or pandemics
    • G16H50/20ICT specially adapted for medical diagnosis, medical simulation or medical data mining; ICT specially adapted for detecting, monitoring or modelling epidemics or pandemics for computer-aided diagnosis, e.g. based on medical expert systems
    • GPHYSICS
    • G16INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR SPECIFIC APPLICATION FIELDS
    • G16HHEALTHCARE INFORMATICS, i.e. INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR THE HANDLING OR PROCESSING OF MEDICAL OR HEALTHCARE DATA
    • G16H50/00ICT specially adapted for medical diagnosis, medical simulation or medical data mining; ICT specially adapted for detecting, monitoring or modelling epidemics or pandemics
    • G16H50/30ICT specially adapted for medical diagnosis, medical simulation or medical data mining; ICT specially adapted for detecting, monitoring or modelling epidemics or pandemics for calculating health indices; for individual health risk assessment
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/118Prognosis of disease development
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/136Screening for pharmacological compounds
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/158Expression markers
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2500/00Screening for compounds of potential therapeutic value
    • G01N2500/04Screening involving studying the effect of compounds C directly on molecule A (e.g. C are potential ligands for a receptor A, or potential substrates for an enzyme A)
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2800/00Detection or diagnosis of diseases
    • G01N2800/38Pediatrics
    • G01N2800/385Congenital anomalies
    • G01N2800/387Down syndrome; Trisomy 18; Trisomy 13
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2800/00Detection or diagnosis of diseases
    • G01N2800/50Determining the risk of developing a disease
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2800/00Detection or diagnosis of diseases
    • G01N2800/52Predicting or monitoring the response to treatment, e.g. for selection of therapy based on assay results in personalised medicine; Prognosis

Landscapes

  • Health & Medical Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Physics & Mathematics (AREA)
  • Medical Informatics (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Biotechnology (AREA)
  • Public Health (AREA)
  • Molecular Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Biomedical Technology (AREA)
  • Genetics & Genomics (AREA)
  • Pathology (AREA)
  • Analytical Chemistry (AREA)
  • Biophysics (AREA)
  • Primary Health Care (AREA)
  • Immunology (AREA)
  • Bioinformatics & Computational Biology (AREA)
  • Epidemiology (AREA)
  • Evolutionary Biology (AREA)
  • Spectroscopy & Molecular Physics (AREA)
  • Theoretical Computer Science (AREA)
  • Organic Chemistry (AREA)
  • Hematology (AREA)
  • Medicinal Chemistry (AREA)
  • Zoology (AREA)
  • Wood Science & Technology (AREA)
  • Databases & Information Systems (AREA)
  • Microbiology (AREA)
  • Urology & Nephrology (AREA)
  • Data Mining & Analysis (AREA)
  • Biochemistry (AREA)
  • General Physics & Mathematics (AREA)
  • Food Science & Technology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Cell Biology (AREA)
  • General Engineering & Computer Science (AREA)
  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)

Abstract

The invention provides an ES marker and application thereof. The ES marker comprises at least one of: ESRRA, MYBL1, PBX1, TEAD1, TEAD2, TEAD4, FOS, JUNB, FOSL2, JUN, HOXC12, SREBF1, Twist2, ZNF410, TBX20, SREBF 2. The inventor finds that the expression levels of a plurality of transcription factors in samples of ES patients and healthy control group population have significant difference by means of the scATAC-seq technology, so that the transcription factors with different expression can be used as markers for ES diagnosis, prognosis evaluation or drug screening.

Description

ES marker and application thereof
Technical Field
The invention relates to the technical field of trisomy syndrome, in particular to an ES marker and application thereof.
Background
Trisomy 18, also known as Edwards Syndrome (ES), is one of the common diseases of chromosomal abnormalities. Specifically, ES is a disease in which a series of different organ and system abnormalities are caused by the presence of extra chromosome 18, and is classified into episomal (typical symptoms account for 90% or more), chimeric, and translocation types according to the karyotype of patients. ES is a chromosome abnormality disease, and no radical treatment method is available, and the ES can be prevented only by prenatal screening. Screening factors include age of pregnant women, ultrasound index, free fetal nucleic acid, etc. However, the high risk results of these techniques described above all eventually require a definitive diagnosis by karyotyping, with a certain risk of infection. The results of the research on trisomy syndrome by Fitz Patrick et al based on a cDNA array analysis technology show that the average transcription level of the gene of the trisomy is only improved by 1.1 times compared with that of the normal cells, and the difference of the expression level among chromosomes can be detected, which suggests that the increase of one chromosome may influence the transcription level of other chromosome genes. The data from this study support a model: slight upregulation of genes on the trisomy leads to secondary, extensive, and more extreme transcriptional dysregulation, the extent of which may determine the severity of the phenotype. These results suggest that ES can be further studied from an epigenetic perspective.
ATAC-seq (Assay for Transposase-Access chromosome with high-throughput put sequencing) is an emerging scientific research technology for studying Chromatin openness (or Chromatin accessibility) from an epigenetic point of view based on high-throughput sequencing, and the principle is to utilize the characteristic that Transposase Tn5 is easy to specifically bind with open Chromatin, sequence DNA sequence fragments captured by Tn5 enzyme, compare the sequencing result with human genome information, analyze open regions of DNA, and predict Transcription Factor Binding Sites (TFBS) in the whole genome range through further motif analysis. Compared with the traditional DNase-Seq, FAIRE-Seq and other methods for researching open chromatin, the ATAC-Seq has the advantages of less cell quantity requirement, low cost, short period, large data, no need of antibody enrichment, capability of detecting the open state of chromatin in the whole genome range and the like. At present, the single-cell ATAC-Seq is widely applied to the fields of chromosome open map analysis, apparent modification difference research, embryonic development epigenetic modification, tumorigenesis epigenetic mechanism research, tumor heterogeneity and typing research, disease potential marker prediction and the like. Therefore, it is necessary to discover specific biomarkers for ES by ATAC-seq technology.
Disclosure of Invention
The present invention is directed to solving at least one of the problems of the prior art. Therefore, the invention provides an ES marker and application thereof.
In a first aspect, an embodiment of the invention provides an ES marker comprising at least one of: ESRRA, MYBL1, PBX1, TEAD1, TEAD2, TEAD4, FOS, JUNB, FOSL2, JUN, HOXC12, SREBF1, Twist2, ZNF410, TBX20, SREBF 2.
The intestinal flora marker provided by the embodiment of the invention at least has the following beneficial effects:
the inventor finds that the expression levels of a plurality of transcription factors in samples of ES patients and healthy control group population have significant difference by means of the scATAC-seq technology, so that the transcription factors with different expression can be used as markers for ES diagnosis, prognosis evaluation or drug screening.
In a second aspect, one embodiment of the present invention provides the use of an ES marker as described above in the preparation of a diagnostic and/or prognostic reagent for ES. The ES markers are differentially expressed in the population of ES patients and healthy controls, and therefore, diagnosis of ES or assessment of prognosis can be achieved by quantitatively detecting the ES markers in a sample of a subject.
In a third aspect, one embodiment of the present invention provides the use of the ES marker described above for screening a drug for treating ES. The ES marker has differential expression in ES patients and healthy control groups, so that the evaluation of the drug effect of the candidate drug can be realized by quantitatively detecting the ES marker in a sample after drug administration, thereby achieving the purpose of drug screening.
In a fourth aspect, one embodiment of the present invention provides a kit comprising reagents for quantitatively detecting the above-described ES marker. The reagent realizes the quantitative detection of the ES marker in a sample of a subject, and the obtained result can be effectively used for diagnosing ES, evaluating the prognosis of the ES or screening ES medicaments after being compared with a preset threshold value.
In a fifth aspect, one embodiment of the present invention provides the use of a reagent for quantitatively detecting the above-described ES marker in the preparation of a diagnostic kit for ES. The ES diagnostic product obtained by the reagent can realize quantitative detection on the ES marker in a sample of a subject, and the detection result can be accurately used for diagnosing ES, or evaluating the prognosis of ES, or screening ES medicaments after being compared with a preset threshold value.
According to some embodiments of the invention, the reagent quantitatively detects the ES marker at a gene level and/or a protein level. For example, the amount of nucleic acid fragments of the ES marker in the sample is quantitatively determined by PCR, or the amount of expression of proteins of the ES marker in the sample is quantitatively determined by enzyme-linked immunoassay, radioimmunoassay, Western blotting, protein chip method, or the like.
In a sixth aspect, an embodiment of the present invention provides a method of assessing risk of ES, the method comprising the steps of:
(1) obtaining the relative abundance of the ES marker in the subject sample;
(2) comparing the relative abundance with a preset threshold value, and judging the ES risk according to the comparison result;
this method is not suitable for the diagnosis and/or treatment of diseases.
Wherein ES risk refers to the risk of the subject having ES, or to the prognosis of the subject; or to the effect of a candidate drug on ES. The preset threshold is a critical value of the ES marker of the normal subject, and specifically may be a relative abundance value of the ES marker measured from a sample of a healthy control group, or a normalized relative abundance value.
According to the method of some embodiments of the present invention, when directly judging the risk of ES based on the relative abundance result of the markers, at least one of the following conditions is satisfied, and it can be judged that the subject has ES, or the prognosis of the ES patient is poor, or the candidate drug does not meet the screening condition:
(1) the relative abundance of the ESRRA is lower than a preset threshold;
(2) the relative abundance of MYBL1 is lower than a preset threshold;
(3) the relative abundance of PBX1 is lower than a preset threshold;
(4) the relative abundance of TEAD1 is below a preset threshold;
(5) the relative abundance of TEAD2 is below a preset threshold;
(6) the relative abundance of TEAD4 is below a preset threshold;
(7) FOS is that the relative abundance ratio of JUNB is lower than a preset threshold value;
(8) FOSL2, the relative abundance of JUN is lower than the preset threshold;
(9) the relative abundance of HOXC12 is higher than a preset threshold;
(10) the relative abundance of SREBF1 is lower than a preset threshold;
(11) the relative abundance of Twist2 is lower than a preset threshold;
(12) the relative abundance of ZNF410 is lower than a preset threshold value;
(13) the relative abundance of TBX20 is higher than a preset threshold;
(14) the relative abundance of SREBF2 is below a preset threshold.
In a seventh aspect, an embodiment of the present invention provides a computer-readable storage medium storing computer-executable instructions for performing the above-mentioned method. By performing the above method, it is possible to accurately and efficiently determine whether a subject has ES, or the prognosis of an ES patient, or the efficacy of ES on a drug candidate.
In an eighth aspect, an embodiment of the present invention provides a system for assessing risk of ES, the system comprising:
a detection means for determining the relative abundance of the ES marker in the subject sample;
and the comparison device is used for comparing the relative abundance with a preset threshold value and judging the ES risk according to the comparison result.
Drawings
FIG. 1 is the result of differential transcription factor motif identification of a B cell subset of UBMC of ES disease patients according to example 1 of the present invention.
FIG. 2 is the result of differential transcription factor motif identification of CD8+ T cell subpopulation of UBMC of ES disease patient according to example 1 of the present invention.
FIG. 3 is the result of differential transcription factor motif identification of DC cell subpopulation of UBMC of ES disease patient according to example 1 of the present invention.
FIG. 4 is the result of differential transcription factor motif identification of monocyte subpopulation of UBMC of ES disease patient according to example 1 of the present invention.
FIG. 5 is the result of identifying the differential transcription factor motif of NK cell subsets of UBMC of ES disease patients according to example 1 of the present invention.
FIG. 6 is the result of differential transcription factor motif identification of T cell subpopulation of UBMC of ES disease patient according to example 1 of the present invention.
Fig. 7 is a schematic composition diagram of a system for evaluating ES risk according to embodiment 2 of the present invention.
Detailed Description
The concept and technical effects of the present invention will be clearly and completely described below in conjunction with the embodiments to fully understand the objects, features and effects of the present invention. It is obvious that the described embodiments are only a part of the embodiments of the present invention, and not all embodiments, and those skilled in the art can obtain other embodiments without inventive effort based on the embodiments of the present invention, and all embodiments are within the protection scope of the present invention.
Example 1
1. Selection of the subject
Based on the results of the G-banding karyotype analysis, 1 patient with trisomy 18 was selected, and 1 control with age-and sex-matched G-banding karyotype analysis results as normal was selected. Both ES patients and healthy control volunteers received informed consent and signed the consent form.
2. Obtaining a sample of umbilical cord blood mononuclear cells (UBMC)
(1) At the end of delivery of the pregnant woman and within 2min after birth of the infant, the umbilical cord is clamped with hemostatic forceps to prevent blood flow and cut the umbilical cord, the cutting position being selected to be 10cm to 15cm from the neonate's end. And (5) disinfecting the umbilical cord by using iodine gauze, and quickly disinfecting the umbilical cord to the placenta position along the umbilical cord broken position for three times. To prevent contamination of cord blood with bacteria, disinfection is critical to cord blood collection. If the disinfection is not in place, the single cell sequencing result can be interfered, and the result accuracy is influenced. The syringe needle of blood taking needle inserts from the umbilical vein, and EDTA anti-coagulation heparin tube is connected to the other end, treats that blood reaches two-thirds time in the heparin tube, extracts the blood taking needle and stops the blood sampling. The anticoagulation tube is turned upside down and shaken for 5 times, so that the EDTA anticoagulant and the cord blood are fully mixed to prevent the blood from coagulating.
(2) Respectively adding physiological saline with the same volume at room temperature, and uniformly mixing to dilute the umbilical cord blood; sucking 6mL of lymphocyte separation solution (GE Healthcare, 17-1440-03) and injecting into a 15mL sterile centrifuge tube, slowly adding 6mL of diluted peripheral blood along the tube wall, and centrifuging (2000rpm, 30min, 20 ℃) by using a centrifuge; dividing the tube from the bottom to the liquid level into four layers after centrifugation, namely a red blood cell layer, a lymph liquid layer, a mononuclear cell layer and a plasma layer, collecting the mononuclear cell layer by using a dropper, and transferring the mononuclear cell layer to a sterile centrifuge tube; PBS was added to 12mL, mixed well and centrifuged (2000rpm, 15min, 20 ℃), the supernatant removed, the UBMC washed and the procedure repeated twice; adding 1mL of erythrocyte lysate (Solarbio, cat # R1010), mixing and transferring to a 1.5mL sterile EP tube, standing in a refrigerator at 4 ℃ for 40min, centrifuging (1600rpm, 5min, 20 ℃) using a Centrifuge (Centrifuge 5418, Eppendorf, Germany), removing the supernatant to obtain a cell pellet; adding 1mL PBS, resuspending the cell pellet, centrifuging (1600rpm, 5min, 20 ℃), removing the supernatant to obtain cell pellet, and repeating the operation twice; adding PBS containing 0.04% BSA, mixing UBMC of ES patient and healthy control group, and concentrating to 1 mL; the cells were counted using a hemocytometer plate and the samples were stored at 4 ℃ until use.
Scata-seq UBMC suspension treatment and sequencing on machine
(1) 20 μ L of UBMC suspension was mixed with 20 μ L of 2% Trypan blue and 20 μ L of the mixture was pipetted from the mixture and added to the blood cell counting plate for counting viability, the UBMC was diluted with 0.04% BSA in PBS to 2 × 106one/mL and filtered using a 40 μm Flowmi cell filter. The cell activity was greater than 85% using trypan blue calculation for live filtered UBMC for further experiments.
(2) Transfer 1 × 106The UBMC were placed in a 2mL sterile centrifuge tube and centrifuged (3000g, 5min, 20 ℃) and the supernatant discarded. mu.L of cold lysis buffer (10mM Tris-HCl (pH 7.4), 10mM NaCl, 3mM MgCl) was added20.1% Tween-20, 0.1% Nonidet P40 substitee, 0.01% Digitonin, 1% BSA), mixing and ice-bath for 3 min; 1mL of cold wash (10mM Tris-HCl (pH 7.4), 10mM NaCl, 3mM MgCl) was added20.1% Tween-20, 1% BSA), centrifugation (500g, 20 ℃, 5min), discarding the supernatant, adding cold 10-fold diluted cell nucleus buffer (10 × Genomics, PN-2000153), and storing in ice bath for later use.
(3) Nuclei were counted using a blood cell counting plate and a microscope. 10 μ L of 30% ATAC transposase (10 XGenomics, PN-2000123/2000138) and 5 μ L of diluted cell nucleus solution were mixed slowly and uniformly, incubated at 37 deg.C for 60min, and stored in a refrigerator at 4 deg.C. Mixing 15 μ L of the above-mentioned Cell nucleus solution containing transposase processing with 65 μ L of a reaction mixture (Barcoding Reagent, PN-2000124; Reducing Agent B, PN-2000087; Barcoding enzyme, PN-2000125/2000139) according to the operational flow instructions of a kit (chromosome Single Cell ATAC Reagent kit, 10 × Genomics, PN-1000083), loading 75 μ L of the above-mentioned mixed sample and Gel beads (Gel beads, 10 × Genomics, PN-2000132) to 10 × Genomics chromium chips (10 × Genomics, PN-1000086), and forming oil drop-encapsulated GEMs by using microfluidic technology; mu.L of GEMs were pipetted from the recovery wells into a PCR-8 calandria, and the gel beads in the GEMs were dissolved using a thermal cycler to generate 10 XBarcode-labeled single stranded DNA (72 ℃, 5 min; 98 ℃, 30 s; 98 ℃, 10 s; 59 ℃, 30 s; 72 ℃, 1 min; 12 cycles; 15 ℃ storage).
(4) Add 125. mu.L of recovery reagent (10 XGenomics, PN-220016) to each sample, mix well and centrifuge, remove the bottom 125. mu.L of red oil layer. 200 μ L of the cleaning mixture (91.0% Cleanup buffer, 10 XGenomics, PN-2000088; 4.0% Dynabeads Myone SILANE, 10 XGenomics, PN-2000048; 2.5% Reducing agent B, 10 XGenomics, N-2000087; nucleic-free Watermo Fisher scientific AM9937) was added, mixed by shaking, allowed to stand at room temperature for 10min, and the supernatant removed. The precipitate was washed with 300. mu.L of 80% ethanol, and after standing for 30s, the ethanol was removed. Add 80% ethanol 200. mu.L, let stand for 30s, remove ethanol by centrifugation, add 5. mu.L of eluent I (98% Buffer EB, Qiagen, 9086; 1% Tween 20, Bio-Rad, 1662404; 1% Reducing Agent B, 10 Xgenomics, PN-2000087), mix well, let stand for 1min at room temperature, transfer 40. mu.L of sample to a new calandria after the solution is clarified. The sample was purified using the SPRISELECT Reagent Kit (Beckman Coulter, B23317), the purified sample was mixed with 48. mu.L of the LSPRIselect Reagent, reacted at room temperature for 5min, and the supernatant was removed after centrifugation. Adding 80% ethanol 200 μ L, standing for 30s, removing ethanol, and washing repeatedly. Adding 40.5 mu L of Buffer EB, uniformly mixing, standing for 2min at room temperature, clarifying the solution, and transferring 40 mu L of sample to a new calandria; storage at-20 deg.C (2 weeks).
(5) Sample Index amplification reagents (50. mu.L of Amp Mix, 10 XGenomics, PN-2000047; 7.5. mu.LSI-PCR Primer B, 10 XGenomics, PN-2000128; 2.5. mu.L of Chromium i7 Sample Index N, 10 XGenomics, PN-3000262) and 40. mu.L of Sample were mixed and the Sample was amplified using a thermal cycler (98 ℃, 45 s; 98 ℃, 20s, 67 ℃, 30s, 72 ℃, 20s, 12 cycles; 72 ℃, 1 min; 4 ℃ for up to 3 days). The sample was mixed with 40. mu.L of SPRISELECTReagent and after 5min of reaction 130. mu.L of supernatant was transferred to a new calandria. The sample was mixed with 74. mu.L of SPRISELECTReagent, reacted at room temperature for 5min and the supernatant removed. Add 80% ethanol 200. mu.L, after standing for 30s remove ethanol and repeat the washing once. Adding 20.5 μ L Buffer EB, mixing, standing for 2min, transferring 20 μ L clarified sample to new calandria, and storing in refrigerator at-20 deg.C. The distribution of the library was evaluated by measuring the concentration of the library sample using Qubit3.0 (reference concentration for DNA concentration detection interval: 0.2-100 ng/. mu.L), measuring 1. mu.L of the sample on Agilent 2100 chip, and determining the size of the DNA fragment. Using a Library Quantification Kit (KAPA Library Quantification Kit, KAPA Biosystems, KK4824) to detect DNA concentrations ranging from 150-1000kb, samples qualified for quality testing were screened for subsequent sequencing on the Illumina platform.
Bioinformatics analysis of scATAC-seq data
Preprocessing raw data: firstly, filtering a sequence with a connector by using Trimmomatic; removing sequences containing > 5% of indeterminate base information; low quality sequences are filtered (the number of bases with a quality value of < 10 is more than 20% of the total sequence).
And (3) data comparison: the effective sequences screened after the above treatment were aligned using bowtie with reference to human genome GRCh38 to analyze the distribution of DNA sequences on the genome.
Analyzing data:
peak identification (peak calling) using MACS software to find open areas of chromatin; counting peak number and indel insert segment length distribution; analyzing the enrichment condition of TSS (transcription Start site), the value of FRiP (Fractionof Reads in captured Peak) to evaluate the enrichment effect of the sample, and calculating the value of IDR (Irreproducibility discovery Rate) to evaluate the consistency of peak among repeated samples; annotating peaks, revealing the distribution of peaks on genome, chromosomes and functional elements, and finding genes that open chromatin regions; finding and annotating motif, exploring sequence preference of open chromatin; and analyzing the difference peaks and related genes among the samples, searching for a difference open chromatin region, and performing GO and KEGG enrichment analysis on the related genes.
RNA-seq UBMC suspension treatment and sequencing on machine
(1) The cells obtained in step 3 (approx. 1 × 10) were centrifuged (1600rpm, 5min, 20 ℃ C.)6Tube/tube), discard the supernatant, add 1mL PBS (RNAase free, room temperature), gently suspend the cell pellet, and transfer to a 2mL spin-top conical centrifuge tube. After removing PBS by centrifugation, 1mL of cell lysate (TRIzol, Invitrogen, Carlsbad, USA) was added, repeatedly blown to a clear and non-viscous liquid, and allowed to stand at room temperature for 5min and stored at-80 ℃ in a refrigerator.
(2) Thawing the above liquid at room temperature, adding chloroform 200 μ L, mixing, standing on ice for 3min, and centrifuging (12000rpm, 5min, 4 deg.C). Transferring the upper water phase into a new EP tube, adding equal amount of isopropanol, mixing, ice-cooling for 10min, centrifuging (12000rpm, 10min, 4 deg.C), removing supernatant, adding 600 μ L75% ethanol, mixing, centrifuging (7500rpm, 5min, 4 deg.C), removing supernatant, drying at room temperature for 10min, and adding 50 μ L pure water without RNase to dissolve precipitate. The quality of the extracted RNA was assessed spectrophotometrically (A260nm/A280nm >1.8, BioSpec-Nano, Shimadzu) and by capillary electrophoresis in combination with the RNA 6000Nano kit (RIN >9, 2100Bioanalyzer, Agilent Technologies, Santa Clara, Calif.).
(3) mRNA was enriched and purified using magnetic beads (with polyA tail). The treated mRNA was fragmented by dephosphorylation and phosphate addition treatment, so that a phosphate group was added to the 5 'end of the mRNA and a hydroxyl group was added to the 3' end. T4RNA ligase 2 was used to ligate an adenylated single stranded DNA3 'linker and a 5' linker to each end of the mRNA. cDNA was synthesized by reverse transcription. The RT products were screened for 350-450 base length using brads. The data library was constructed after 15 cycles of PCR amplification. The Illumina Hiseq 2500 was used for sequencing with a sequencing read length of 2 × 150bp paired end.
Bioinformatic analysis of RNA-seq data
And (3) screening data: firstly, filtering a sequence with a joint; sequences containing > 5% of indeterminate base information were removed and low quality sequences were filtered.
Using human genome GRCh38 as reference, using TopHat2 program to perform genome comparison with gene reference, and respectively counting sequence number, region distribution summary and chromosome density distribution of the comparison between sequencing data and reference genome according to gtf-specified gene position information;
analyzing the gene expression level, and measuring the abundance value of gene expression by using FPKM (fragments Per Kibase of exon model Million mapped reads);
analyzing gene differential expression, and performing differential analysis on a newly constructed gene by using a cuffdiff command of cufflinks software (the differential threshold value is P < 0.05);
differential gene function was analyzed, using GOseq for GO (Gene enrichment) enrichment and KOBAS for KEGG (Kyoto Encyclopedia of Genes and genomes) signal pathway enrichment.
As shown in fig. 1 to 6, fig. 1 to 6 are volcano charts showing the results of differential transcription factor motif identification of B cell subset, CD8+ T cell subset, DC cell subset, monocyte subset, NK cell subset and T cell subset of UBMC of the ES disease patient according to example 1 of the present invention, respectively, in which the left dotted line is down-regulated expression to the left and the right dotted line is up-regulated expression to the right.
As can be seen from fig. 1 to 6, in contrast to the healthy control group, in the ES environment:
(1) in the B cell subgroup, ESRRA motif, TEAD 1motif, TEAD2 motif, TEAD4 motif, PBX1motif, ZNF410 motif and FOS comprise JUNB motif and FOSL2, JUN motif, SREBF 1motif and Twist2 motif are expressed in a down-regulation mode, and HOXC12 motif is expressed in an up-regulation mode;
(2) TEAD 1motif down-regulated expression in the CD8+ T cell subset;
(3) HOXC12 motif upregulated expression in DC cell subsets;
(4) in the monocyte subgroup, PBX1motif, TEAD 1motif, TEAD2 motif, ZNF410 motif and ESRRA motif are expressed in a down-regulated manner, and HOXC12 motif and TBX20 motif are expressed in an up-regulated manner;
(5) in NK cell subset, SREBF2 motif down-regulates expression;
(6) in the T cell subsets, expression was down-regulated by ESRRA motif, TEAD 1motif, TEAD2 motif, TEAD4 motif, PBX1motif, MYBL1 motif.
Because of the specificity of the binding site sequences of the transcription factors when the transcription factors are combined with DNA sequences, the differential expression of the transcription factors motif can reflect that the transcription factors aiming at the binding site sequences also have obvious differential expression in ES patients. Thus, the differential transcription factor identified above in a sample can be quantitatively detected at the gene or protein level for diagnosis, prognosis or drug screening of ES.
Example 2
The present embodiments provide a system for assessing risk of ES. Fig. 7 is a schematic composition diagram of a system for evaluating ES risk according to embodiment 2 of the present invention. Referring to fig. 7, the system includes: a nucleic acid isolation apparatus 100 for isolating a nucleic acid sample from a stool sample of a subject; and the sequencing device 110 is used for detecting the ES markers in the nucleic acid sample, comparing the relative abundance of the markers with a preset threshold value, and judging the ES risk according to the relative sizes of the markers and the preset threshold value.
Example 3
The present embodiments provide a computer-readable storage medium having stored thereon computer-executable instructions for performing a method comprising:
(1) obtaining the relative abundance of the ES marker in the subject sample;
(2) and comparing the relative abundance with a preset threshold, judging whether the patients have ES or ES prognosis or screening the drugs according to the comparison result.
Example 4
The embodiment provides a protein chip, which is coated with antibodies of ESRRA, TEAD1, TEAD2, TEAD4 and PBX1, the protein chip and corresponding detection reagents can be used for detecting the contents of the markers in a serum sample of a subject, and the detection result is standardized and then compared with a preset threshold value, so that whether the sample is suffering from ES can be effectively judged.
The embodiments of the present invention have been described in detail with reference to the accompanying drawings, but the present invention is not limited to the above embodiments, and various changes can be made within the knowledge of those skilled in the art without departing from the gist of the present invention. Furthermore, the embodiments of the present invention and the features of the embodiments may be combined with each other without conflict.

Claims (9)

1. An ES marker comprising at least one of: ESRRA, MYBL1, PBX1, TEAD1, TEAD2, TEAD4, FOS, JUNB, FOSL2, JUN, HOXC12, SREBF1, Twist2, ZNF410, TBX20, SREBF 2.
2. Use of an ES marker according to claim 1 in the preparation of a diagnostic and/or prognostic reagent for ES.
3. Use of the ES marker of claim 1 for screening a medicament for treating ES.
4. A kit comprising reagents for quantitatively detecting the ES marker of claim 1.
5. Use of a reagent for quantitatively detecting an ES marker as claimed in claim 1 in the preparation of a diagnostic kit for ES.
6. The use according to claim 5, wherein the reagent quantitatively detects the ES marker of claim 1 at the gene level and/or protein level.
7. A method of assessing risk of ES, comprising the steps of:
(1) obtaining the relative abundance of the ES marker of claim 1 in a sample from a subject;
(2) comparing the relative abundance with a preset threshold value, and judging the ES risk according to a comparison result;
the method is not suitable for the diagnosis or treatment of diseases.
8. A computer-readable storage medium having stored thereon computer-executable instructions for performing the method of claim 7.
9. A system for assessing risk of ES, comprising:
a detection device for determining the relative abundance of the ES marker of claim 1 in a sample from a subject;
and the comparison device is used for comparing the relative abundance with a preset threshold value and judging the ES risk according to a comparison result.
CN202010393259.2A 2020-05-11 2020-05-11 ES marker and application thereof Pending CN111575365A (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202010393259.2A CN111575365A (en) 2020-05-11 2020-05-11 ES marker and application thereof

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202010393259.2A CN111575365A (en) 2020-05-11 2020-05-11 ES marker and application thereof

Publications (1)

Publication Number Publication Date
CN111575365A true CN111575365A (en) 2020-08-25

Family

ID=72126500

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202010393259.2A Pending CN111575365A (en) 2020-05-11 2020-05-11 ES marker and application thereof

Country Status (1)

Country Link
CN (1) CN111575365A (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2024077856A1 (en) * 2022-10-13 2024-04-18 中山大学中山眼科中心 Use of tbx family transcription factor as target in preparation of drug for treating ophthalmic disease

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR20070062476A (en) * 2007-05-28 2007-06-15 주식회사 마크로젠 Microarray and kit for diagnosing edwards syndrome
CN108548780A (en) * 2018-03-30 2018-09-18 中国人民解放军第二军医大学 The method of transcription factor chip agent box and high flux screening target gene transcription factor
CN110544509A (en) * 2019-08-20 2019-12-06 广州基迪奥生物科技有限公司 single-cell ATAC-seq data analysis method

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR20070062476A (en) * 2007-05-28 2007-06-15 주식회사 마크로젠 Microarray and kit for diagnosing edwards syndrome
CN108548780A (en) * 2018-03-30 2018-09-18 中国人民解放军第二军医大学 The method of transcription factor chip agent box and high flux screening target gene transcription factor
CN110544509A (en) * 2019-08-20 2019-12-06 广州基迪奥生物科技有限公司 single-cell ATAC-seq data analysis method

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
IGOR ALBIZUA等: "Analysis of the genomic expression profile in trisomy 18: insight into possible genes involved in the associated phenotypes", 《HUM MOL GENET》 *
李晓洲等: "分子生物学技术产前诊断18三体综合征的研究进展", 《国外医学(计划生育/生殖健康分册)》 *
邹耀霜等: "环状RNA在孕妇18-三体综合征的鉴定与分析", 《中国产前诊断杂志(电子版)》 *
黄滔滔等: "心脏病遗传因素对母胎的影响", 《妇产与遗传(电子版)》 *

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2024077856A1 (en) * 2022-10-13 2024-04-18 中山大学中山眼科中心 Use of tbx family transcription factor as target in preparation of drug for treating ophthalmic disease

Similar Documents

Publication Publication Date Title
Shah et al. A recurrent germline PAX5 mutation confers susceptibility to pre-B cell acute lymphoblastic leukemia
Gasparetto et al. Transcription and DNA methylation patterns of blood-derived CD8+ T cells are associated with age and inflammatory bowel disease but do not predict prognosis
Skibsted et al. Bench-to-bedside review: future novel diagnostics for sepsis-a systems biology approach
Sharp et al. Polymorphisms in protein tyrosine phosphatase non-receptor type 2 and 22 (PTPN2/22) are linked to hyper-proliferative T-Cells and susceptibility to mycobacteria in rheumatoid arthritis
CN109477145A (en) The biomarker of inflammatory bowel disease
EP3494235A1 (en) Swarm intelligence-enhanced diagnosis and therapy selection for cancer using tumor- educated platelets
CN112119166A (en) Diagnostic use of cell-free DNA chromatin immunoprecipitation
EP3693742B1 (en) Methods of detecting prostate cancer
JP2020511933A (en) Methods for cancer detection
Reggiardo et al. LncRNA biomarkers of inflammation and cancer
Jiang et al. circPTPN22 as a novel biomarker and ceRNA in peripheral blood mononuclear cells of rheumatoid arthritis
Rao et al. Transcription factor–nucleosome dynamics from plasma cfDNA identifies ER-driven states in breast cancer
Mohanty et al. Liquid Biopsy, the hype vs. hope in molecular and clinical oncology
CN110880356A (en) Method and apparatus for screening, diagnosing or risk stratification for ovarian cancer
Wang et al. Single-cell dissection of remodeled inflammatory ecosystem in primary and metastatic gallbladder carcinoma
Wang et al. Serum extracellular vesicle MicroRNAs as candidate biomarkers for acute rejection in patients subjected to liver transplant
CN111575365A (en) ES marker and application thereof
WO2021041299A1 (en) Method and kit for determining benefit of chemotherapy
CN103911439A (en) Analyzing method and application of differential expression gene of systemic lupus erythematosus hydroxymethylation status
Xuan et al. Do lncRNAs and circRNAs expression profiles influence discoid lupus erythematosus progression?—a comprehensive analysis
CN111518893A (en) Uremia marker and application thereof
WO2020028456A1 (en) Compositions and methods for detecting prostate cancer
CN115873936A (en) Biomarker CXCL8 and application thereof
WO2023044117A1 (en) Drain fluid for diagnostics
JP2022551488A (en) Using Concurrent Marker Detection to Assess Diffuse Glioma and Responsiveness to Therapy

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
SE01 Entry into force of request for substantive examination
SE01 Entry into force of request for substantive examination