CN115612745B - Application of IPO7 in colorectal cancer treatment and prognosis prediction - Google Patents

Application of IPO7 in colorectal cancer treatment and prognosis prediction Download PDF

Info

Publication number
CN115612745B
CN115612745B CN202211629209.5A CN202211629209A CN115612745B CN 115612745 B CN115612745 B CN 115612745B CN 202211629209 A CN202211629209 A CN 202211629209A CN 115612745 B CN115612745 B CN 115612745B
Authority
CN
China
Prior art keywords
ipo7
colorectal cancer
cells
inhibitor
prognosis
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.)
Active
Application number
CN202211629209.5A
Other languages
Chinese (zh)
Other versions
CN115612745A (en
Inventor
申占龙
杨长江
叶颖江
王杉
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 Peoples Hospital
Original Assignee
Peking University 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 Peking University Peoples Hospital filed Critical Peking University Peoples Hospital
Priority to CN202211629209.5A priority Critical patent/CN115612745B/en
Publication of CN115612745A publication Critical patent/CN115612745A/en
Application granted granted Critical
Publication of CN115612745B publication Critical patent/CN115612745B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

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
    • C12Q1/6886Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material for cancer
    • 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
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • 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
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/574Immunoassay; Biospecific binding assay; Materials therefor for cancer
    • G01N33/57407Specifically defined cancers
    • G01N33/57419Specifically defined cancers of colon
    • 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
    • C12Q2600/158Expression markers

Abstract

The invention discloses application of IPO7 in colorectal cancer treatment and prognosis prediction, provides a product for diagnosing colorectal cancer/diagnosing colorectal cancer metastasis/predicting colorectal cancer prognosis, and also provides a pharmaceutical composition for treating colorectal cancer. According to the invention, extensive and in-depth experiments prove that IPO7 can effectively diagnose colorectal cancer and the metastasis of the colorectal cancer and can effectively predict the prognosis of the colorectal cancer, and proliferation, migration, invasion, apoptosis and division experiments prove that the effect of treating the colorectal cancer can be achieved by inhibiting the expression of IPO7.

Description

Application of IPO7 in colorectal cancer treatment and prognosis prediction
Technical Field
The invention belongs to the field of biomedicine, and particularly relates to application of IPO7 in colorectal cancer treatment and prognosis prediction.
Background
Colorectal Cancer (CRC) has become the third largest malignancy worldwide, second only to lung and female breast Cancer, and has become the second leading cause of Cancer-related death in 2020. Because colorectal cancer is hidden, specific symptoms and signs are not always present in the early stage, most patients are in the middle and late stage when visiting the doctor, even have multiple metastases of the whole body, the chance of surgical treatment is lost, and the colorectal cancer is the main reason for death. The patient prognosis varies significantly even after surgical treatment. Therefore, in order to develop a comprehensive and individual treatment scheme for patients, improve the prognosis level of patients, and find effective molecular markers and specific drug targets for colorectal cancer diagnosis and prognosis.
Disclosure of Invention
In order to make up for the defects of the prior art, the invention provides the molecular marker IPO7 which can realize the diagnosis, the metastasis diagnosis, the treatment and the prognosis prediction of colorectal cancer.
In order to achieve the purpose, the invention adopts the following technical scheme:
a first aspect of the invention provides a product for diagnosing colorectal cancer/diagnosing colorectal cancer metastasis/predicting colorectal cancer prognosis, the product comprising an agent capable of detecting the expression level of IPO7.
Further, the reagent is selected from an oligonucleotide probe that specifically recognizes the IPO7 gene, a primer that specifically amplifies the IPO7 gene, or a binding agent that specifically binds to a protein encoded by the IPO7 gene.
Further, the product comprises a chip, a kit or a nucleic acid membrane strip.
A second aspect of the invention provides a pharmaceutical composition for the treatment of colorectal cancer, the pharmaceutical composition comprising an inhibitor of IPO7.
Further, the inhibitor inhibits proliferation, migration, invasion, apoptosis and/or division of colorectal cancer.
Further, the inhibitor comprises a nucleic acid inhibitor and a protein inhibitor.
A third aspect of the present invention provides a method of screening for a candidate drug for the treatment of colorectal cancer, the method comprising: treating the culture system expressing or containing the IPO7 gene or its encoded protein with a substance to be screened; and detecting expression or activity of the IPO7 gene or protein encoded thereby in said system; wherein, when the substance to be screened inhibits the expression level or activity of the IPO7 gene or the protein encoded by the gene, the substance to be screened is a candidate drug for treating colorectal cancer.
A fourth aspect of the present invention provides a system/apparatus for diagnosing colorectal cancer/diagnosing colorectal cancer metastasis/predicting colorectal cancer prognosis, the system/apparatus comprising:
an acquisition unit: for obtaining the expression level of IPO7 in the sample;
a processing unit: and obtaining a diagnosis/diagnosis metastasis/prognosis prediction result of the colorectal cancer according to the expression condition of the IPO7.
A fifth aspect of the present invention provides a computer readable storage medium having stored thereon a computer program which, when executed by a processor, implements the system/apparatus of the fourth aspect of the present invention.
A sixth aspect of the invention provides the use of any one of:
(1) Use of an inhibitor of IPO7 or a pharmaceutical composition according to the second aspect of the invention in the manufacture of a product for the treatment of colorectal cancer;
(2) Use of a reagent for detecting the expression level of IPO7 or a product according to the first aspect of the invention for the manufacture of a tool for diagnosing colorectal cancer/diagnosing colorectal cancer metastasis/predicting colorectal cancer prognosis;
(3) Use of IPO7 for screening a candidate drug for the treatment of colorectal cancer;
(4) Use of IPO7 for promoting macrophage differentiation;
(5) Use of IPO7 in the construction of a system/device for diagnosing colorectal cancer/diagnosing colorectal cancer metastasis/predicting colorectal cancer prognosis;
(6) Use of IPO7 in the construction of a computer readable storage medium for diagnosing colorectal cancer/diagnosing colorectal cancer metastasis/predicting colorectal cancer prognosis.
The invention has the advantages and beneficial effects that:
the molecular marker IPO7 provided by the invention can effectively diagnose colorectal cancer and metastasis of the colorectal cancer, and can achieve the purpose of treating the colorectal cancer by inhibiting proliferation, migration, invasion, apoptosis and division of colorectal cancer cells, and in addition, the IPO7 can well predict the prognosis of the colorectal cancer.
Drawings
FIG. 1 is a protein expression difference volcano diagram, wherein 1A is a protein expression difference diagram of a colorectal cancer primary focus and a normal tissue, and 1B is a protein expression difference diagram of a colorectal cancer liver metastasis and a primary focus;
FIG. 2 is a Wien diagram of groups of co-up/down regulated proteins;
FIG. 3 is a graph of the difference in protein expression of IPO7 in normal colorectal tissue, primary foci, and liver metastases, wherein 3A is a graph of immunohistochemical staining results and 3B is a statistical graph of immunohistochemical scores;
FIG. 4 is a graph of the difference in protein expression of IPO7 in colorectal cancer and paracarcinoma, wherein 4A is a graph of immunohistochemical staining results and 4B is a statistical graph of immunohistochemical scores;
FIG. 5 is a graph of the relationship between IPO7 and overall survival of colorectal cancer patients;
FIG. 6 is a diagram of the detection of the knockdown efficiency of IPO7 and the construction of stable transgenic cell lines, wherein 6A is a diagram of the knockdown efficiency of siRNA to IPO7 in DLD1, 6B is a diagram of the mRNA expression level of IPO7 in stable transgenic cell lines, 6C is a Western immunoblot diagram of IPO7 in stable transgenic cell lines, and 6D is a diagram of lentivirus-transfected stable transgenic cell lines with knockdown of IPO 7;
FIG. 7 is a graph of the proliferative effect of knocking down IPO7 on DLD1 cells;
FIG. 8 is a graph showing the clonality effect of knocking down IPO7 on DLD1 cells, where 8A is a clonality map and 8B is a clonality statistic map;
FIG. 9 is a graph of the effect of knockdown of IPO7 on the migratory capacity of DLD1 cells, where 9A is a score healing profile, 9B is a 24h wound healing rate statistic, and 9C is a 48h wound healing rate statistic;
fig. 10 is a graph showing the effect of knocking down IPO7 on the migration and invasion abilities of DLD1 cells, wherein 10A is a DLD1 cell migration ability map, 10B is a DLD1 cell migration ability statistical map, 10C is an invasion ability map of DLD1 cells, and 10D is an invasion ability statistical map of DLD1 cells;
fig. 11 is a graph showing the effect of knocking-down IPO7 on DLD1 cell cycle, in which 11A is a graph showing the distribution ratio of the cells in different stages of the shmc, 11B is a graph showing the distribution ratio of the cells in different stages of the shmpo 7-1, 11C is a graph showing the distribution ratio of the cells in different stages of the shmpo 7-2, 11D is a graph showing the distribution statistics of the cells in G1 stage, 11E is a graph showing the distribution statistics of the cells in G2 stage, and 11F is a graph showing the distribution statistics of the cells in S stage;
fig. 12 is a graph of the apoptotic effect of knocking down IPO7 on DLD1 cells, in which 12A is a graph of the distribution ratio of the shmc in different apoptotic stages, 12B is a graph of the distribution ratio of the ship 7-1 in different apoptotic stages, 12C is a graph of the distribution ratio of the ship 7-2 in different apoptotic stages, 12D is a graph of the distribution statistics of early apoptotic cells, and 12E is a graph of the distribution statistics of late apoptotic cells;
FIG. 13 is a graph showing the correlation between IPO7 and macrophage infiltration, in which 13A is a graph showing the correlation between low expression of IPO7 and macrophage infiltration, and 13B is a graph showing the correlation between high expression of IPO7 and macrophage infiltration.
Detailed Description
The following provides definitions of some terms used in this specification. Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs.
The invention provides a product for diagnosing colorectal cancer/diagnosing colorectal cancer metastasis/predicting colorectal cancer prognosis, the product comprising an agent capable of detecting the expression level of IPO7.
IPO7 includes wild type, mutant or fragments thereof. The term encompasses full-length, unprocessed IPO7, as well as any form of IPO7 that results from processing in a cell. The term encompasses naturally occurring variants (e.g., splice variants or allelic variants) of IPO7. The term encompasses for example human IPO7 as well as IPO7 from any other vertebrate source, including mammalian, such as primate and rodent (e.g. mouse and rat) IPO7, gene ID:10527.
In the present invention, diagnosis and variations of the term thereof refer to the discovery, judgment or cognition of an individual's health state or condition based on one or more signs, symptoms, data or other information associated with the individual. The health status of an individual may be diagnosed as healthy/normal (i.e., absence of a disease or condition), or may be diagnosed as unhealthy/abnormal (i.e., presence of an assessment of a disease or condition or characteristic). The above terms diagnosis and variants of the terms include early detection of a disease in association with a particular disease or condition; the nature or classification of the disease; discovery of progression, cure or recurrence of disease; discovery of response to disease after treatment or therapy of an individual.
In the present invention, metastasis refers to a process in which cancer cells originating from one organ or part of the body migrate to another part of the body with or without transport of body fluids and continue to repeat. The metastasized cells may subsequently form tumors that may further metastasize. Metastasis is therefore called the spread of cancer, from the part of the body where it originally occurred to other parts of the body.
In an embodiment of the invention, metastasis refers to hepatic metastasis of colorectal cancer.
In the present invention, prognosis refers to the expectation regarding medical development (e.g., long-term survival likelihood, disease-free survival rate, etc.), including positive prognosis including disease progression such as relapse, colorectal cancer growth, metastasis, and drug-resistant mortality, or negative prognosis including disease remission such as disease-free state, disease improvement such as colorectal cancer regression or stabilization.
In the present invention, expression level or level of expression generally refers to the amount of a biomarker in a biological sample. Expression generally refers to the process by which information (e.g., gene coding and/or epigenetic information) is converted into structures present and operating in a cell. Thus, as used herein, expression may refer to transcription into a polynucleotide, translation into a polypeptide, or even polynucleotide and/or polypeptide modification (e.g., post-translational modification of a polypeptide). Transcribed polynucleotide, translated polypeptide or polynucleotide and/or polypeptide modified (e.g., post-translational modification of a polypeptide) fragments should also be considered expressed, whether they are derived from a transcript produced by alternative splicing or a degraded transcript, or from post-translational processing of a polypeptide (e.g., by proteolysis). Expressed genes include genes that are transcribed into a polynucleotide (e.g., mRNA) and then translated into a polypeptide, as well as genes that are transcribed into RNA but not translated into a polypeptide (e.g., transport and ribosomal RNA).
Further, the reagent for detecting the expression level of IPO7 is selected from an oligonucleotide probe specifically recognizing the IPO7 gene, a primer specifically amplifying the IPO7 gene or a binding agent specifically binding to a protein encoded by the IPO7 gene.
In the present invention, a probe refers to a molecule capable of binding to a specific sequence or subsequence or other portion of another molecule. Unless otherwise indicated, a probe generally refers to a polynucleotide probe that is capable of binding to another polynucleotide (often referred to as a target polynucleotide) by complementary base pairing. Depending on the stringency of the hybridization conditions, a probe can bind to a target polynucleotide that lacks complete sequence complementarity to the probe. The probe may be labeled directly or indirectly, and includes within its scope a primer. Hybridization modes include, but are not limited to: solution phase, solid phase, mixed phase or in situ hybridization assays.
In the present invention, a primer refers to a short nucleic acid sequence, which can form a base pair (basepair) with a complementary template (template) and serves as an origin of replication template, as a nucleic acid sequence having a short free 3 'terminal hydroxyl group (free 3' hydroxyl group).
In the present invention, a binding agent refers to a naturally occurring or non-naturally occurring molecule that specifically binds to a target. Examples of specific binding agents include, but are not limited to, proteins, peptides, nucleic acids, carbohydrates, and lipids.
In the present invention, the binding agent that specifically binds to the protein encoded by the IPO7 gene is, for example, a receptor for the protein IPO7, a lectin that binds to the protein IPO7, an antibody against the protein IPO7, a peptide antibody (peptidebody) against the protein IPO7, a bispecific dual binding agent or a bispecific antibody format. Specific examples of specific binding agents are peptides, peptidomimetics, aptamers, spiegelmers, dappin, ankyrin repeat proteins, kunitz-type domains, antibodies, single domain antibodies and monovalent antibody fragments.
Further, the product includes a chip, a kit or a nucleic acid membrane strip.
In the invention, the chip comprises a gene chip and a protein chip, wherein the gene chip comprises an oligonucleotide probe aiming at IPO7 gene and used for detecting the transcription level of the IPO7 gene, and the protein chip comprises a specific binding agent of IPO7 protein; the kit comprises a gene detection kit and a protein detection kit, wherein the gene detection kit comprises a reagent or a chip for detecting the IPO7 gene transcription level, and the protein detection kit comprises a reagent or a chip for detecting the IPO7 protein expression level.
In the present invention, a kit refers to a set of components provided in the context of a system for sequencing nucleotides and/or isolating nucleotide sequences and/or diagnosing a subject with a disease or infection based on the presence, absence and/or amount of expressed nucleotide sequences from a sample or cell.
The kit comprises a gene detection kit and a protein detection kit, wherein the gene detection kit comprises a reagent or a chip for detecting the IPO7 gene transcription level, and the protein detection kit comprises a reagent or a chip for detecting the IPO7 protein expression level.
The kit of the invention comprises one or more substances of the following group: container, instructions for use, positive control, negative control, buffer, adjuvant or solvent.
The kit can be also attached with an instruction book of the kit, wherein the instruction book describes how to adopt the kit for detection, how to judge the tumor development by using the detection result and how to select a treatment scheme.
The components of the kit may be packaged in the form of an aqueous medium or in lyophilized form. Suitable containers in the kit generally include at least one vial, test tube, flask, pet bottle, syringe, or other container in which a component may be placed and, preferably, suitably aliquoted. Where more than one component is present in the kit, the kit will also typically comprise a second, third or other additional container in which the additional components are separately disposed. However, different combinations of components may be contained in one vial. The kits of the invention also typically include a container for holding the reactants, sealed for commercial sale. Such containers may include injection molded or blow molded plastic containers in which the desired vials may be retained.
The chip, kit or membrane strip of the present invention can be used for detecting the expression level of a plurality of genes or proteins including IPO7 gene or protein and expression products thereof (e.g., a plurality of genes or proteins associated with colorectal cancer). The multiple markers of the colorectal cancer are detected at the same time, so that the accuracy of colorectal cancer diagnosis or prognosis prediction can be greatly improved.
The present invention provides a pharmaceutical composition for the treatment of colorectal cancer comprising an inhibitor of IPO7.
In the present invention, the inhibitor refers to any substance that can reduce the activity of the IPO7 protein, reduce the stability of the IPO7 gene or protein, down-regulate the expression of the IPO7 protein, reduce the effective acting time of the IPO7 protein, or inhibit the transcription and translation of the IPO7 gene, and these substances can be used in the present invention as a substance useful for down-regulating IPO7, and thus can be used for preventing or treating colorectal cancer.
In the present invention, the inhibitor includes a nucleic acid inhibitor and a protein inhibitor. Wherein the nucleic acid inhibitor is selected from: an interfering molecule that targets IPO7 or its transcript and is capable of inhibiting IPO7 gene expression or gene transcription, comprising: shRNA (small hairpin RNA), small interfering RNA (siRNA), dsRNA, microrna, antisense nucleic acid, or a construct capable of expressing or forming said shRNA, small interfering RNA, dsRNA, microrna, antisense nucleic acid. The protein inhibitor is selected from substances that specifically bind to the IPO7 protein, such as antibodies or ligands that are capable of inhibiting the activity of the IPO7 protein.
In an embodiment of the invention, the inhibitor is selected from nucleic acid inhibitors.
In a particular embodiment of the invention, the nucleic acid inhibitor is selected from the group consisting of shRNA (small hairpin RNA) and small interfering RNA (siRNA).
The pharmaceutical compositions of the present invention may be administered orally, parenterally, by inhalation spray, topically, rectally, nasally, buccally, vaginally or via an implanted reservoir. Oral administration or injection administration is preferred. The pharmaceutical composition of the present invention may contain any of the usual non-toxic pharmaceutically acceptable carriers, adjuvants or excipients.
The medicaments of the invention can also be combined with other medicaments for the treatment of colorectal cancer, and the other therapeutic compounds can be administered simultaneously with the main active ingredient (e.g. an inhibitor of IPO 7), even in the same composition. The other therapeutic compounds may also be administered separately, in a separate composition or in a different dosage form than the primary active ingredient. A partial dose of the main ingredient (e.g. an inhibitor of IPO 7) may be administered simultaneously with the other therapeutic compound, while the other dose may be administered separately.
The pharmaceutical compositions of the present invention also include a pharmaceutically acceptable carrier.
Pharmaceutically acceptable carriers include, but are not limited to, diluents, binders, surfactants, humectants, adsorbent carriers, lubricants, fillers, disintegrants.
Wherein the diluent is lactose, sodium chloride, glucose, urea, starch, water, etc.; binders such as starch, pregelatinized starch, dextrin, maltodextrin, sucrose, acacia, gelatin, methyl cellulose, carboxymethyl cellulose, ethyl cellulose, polyvinyl alcohol, polyethylene glycol, polyvinyl pyrrolidone, alginic acid and alginates, xanthan gum, hydroxypropyl cellulose, hydroxypropyl methyl cellulose, and the like; surfactants such as polyoxyethylene sorbitan fatty acid esters, sodium lauryl sulfate, glyceryl monostearate, cetyl alcohol, etc.; humectants such as glycerin, starch, etc.; adsorption carriers such as starch, lactose, bentonite, silica gel, kaolin, and bentonite; lubricants such as zinc stearate, glyceryl monostearate, polyethylene glycol, talc, calcium stearate and magnesium stearate, polyethylene glycol, boric acid powder, hydrogenated vegetable oil, sodium stearyl fumarate, polyoxyethylene monostearate, monolaure Gui Zhe sugar acid ester, sodium lauryl sulfate, magnesium lauryl sulfate, etc.; fillers such as mannitol (granular or powder), xylitol, sorbitol, maltose, erythrose, microcrystalline cellulose, polymeric sugar, coupling sugar, glucose, lactose, sucrose, dextrin, starch, sodium alginate, laminarin powder, agar powder, calcium carbonate, sodium bicarbonate, etc.; disintegrating agent such as crosslinked vinylpyrrolidone, sodium carboxymethyl starch, low-substituted hydroxypropyl methyl, crosslinked sodium carboxymethyl cellulose, and soybean polysaccharide.
The present invention provides a system/apparatus for diagnosing colorectal cancer/diagnosing colorectal cancer metastasis/predicting colorectal cancer prognosis, the system/apparatus comprising:
an acquisition unit: for obtaining the expression level of IPO7 in a sample;
a processing unit: and obtaining a diagnosis/diagnosis metastasis/prognosis prediction result of the colorectal cancer according to the expression condition of the IPO7.
If the expression level of the IPO7 is significantly up-regulated compared with a normal sample, the diagnosis result is colorectal cancer;
compared with the primary focus of colorectal cancer, if the expression level of IPO7 is remarkably up-regulated, the diagnosis result is colorectal cancer liver metastasis;
a high protein expression level of IPO7 indicates a poor prognosis for colorectal cancer.
The invention is further illustrated below with reference to specific examples. It should be understood that the particular embodiments described herein are presented by way of example and not as limitations of the invention. The principal features of this invention can be employed in various embodiments without departing from the scope of the invention.
Example 1 expression of IPO7 in colorectal cancer
1.1 Experimental materials
Tissue specimen information for proteomics: collecting paired paracancer normal tissues (more than 10cm away from the edge of a tumor), a primary colorectal cancer focus and a liver metastasis focus of colorectal cancer patients who are subjected to gastrointestinal surgical operation excision in Beijing university Hospital and do not receive preoperative neoadjuvant radiotherapy and chemotherapy and are combined with simultaneous liver metastasis in 2019-2020-7 months (3 patients have obvious clinical symptoms such as combined bleeding and obstruction, and the primary colorectal cancer focus and the liver metastasis focus can be excised, so that right hemicolectomy and liver partial excision are adopted, and preoperative neoadjuvant therapy is not received). Dividing all the obtained tissue specimens into two parts, quickly freezing one part in liquid nitrogen 30 minutes after the tissue specimens are separated, and then storing in an ultra-low temperature refrigerator at minus 80 ℃; the other part was fixed in 4% tissue cell fixative and then paraffin embedded. The final proteomics study was included in a total of 3 tissue specimens from colorectal cancer patients, the detailed clinical pathology data of which are shown in table 1. Extracting protein in corresponding tissues, and performing TMT labeled quantitative proteomics research after quality control is qualified.
Figure 889939DEST_PATH_IMAGE001
Organizing a chip: 10 cases of paraffin specimens of colorectal cancer tissues, liver metastasis tissues and paracancer normal tissues of patients subjected to gastrointestinal surgery simultaneous colorectal cancer resection and liver resection in Beijing university Hospital, from 1 month to 2021 month in 2019 are selected, and all the patients do not receive chemoradiotherapy before operation.
Large cohort paraffin specimens of colorectal cancer and paired paracancerous normal tissues: collecting colorectal cancer specimens which are surgically removed in Beijing university Hospital gastrointestinal surgery 2014 12-2016 12 months, and taking the specimens into a standard that the specimens are pathologically diagnosed as colorectal cancer; before operation, new auxiliary radiotherapy and chemotherapy and targeted therapy are not received; including complete clinicopathological data (including sex, age, tumor size, histological type, degree of differentiation, vascular cancer emboli, TNM staging, AJCC staging, etc.) and follow-up information (time to live and status of survival). Patients with combined other tumors were excluded when diagnosing colorectal cancer. Together, 128 pairs of normal tissue specimens were included in the colorectal cancer and its paired paracarcinoma. All specimens were prepared into tissue chips and stored in the surgery tumor research laboratory of people's hospital of Beijing university.
1.2 Experimental methods
1.2.1 proteomics analysis
The sample is subjected to standardization treatment, and omics analysis work is completed by Hangzhou Jing Jie Biotechnology Limited.
1.2.2 immunohistochemical staining
(1) Dewaxing and hydrating: baking the paraffin sections at 60 to 70 ℃ for 2 hours, putting the paraffin sections into fresh xylene, and soaking the paraffin sections for 10 minutes and multiplying the times by 3; after removing the redundant liquid, placing the mixture in absolute ethyl alcohol, and soaking for 3 minutes and 3 times; removing redundant liquid, placing in 95% ethanol, soaking for 3 minutes and 2 times; removing redundant liquid, placing in 75% ethanol, soaking for 3 minutes and multiplying 2 times; washing with distilled water for 1 min, and placing in PBS buffer solution;
(2) Antigen retrieval: preheating an antigen repairing solution (1 x) to 95-100 ℃ before use, soaking the slices in the antigen repairing solution, heating at 95 ℃ for 15 minutes, cooling to room temperature within about 20-30 minutes, washing 1~2 times with PBS buffer solution, and 3~5 minutes each time;
(3) Blocking endogenous peroxidase: adding proper amount of endogenous peroxidase blocker (covering all tissues) according to the tissue size, incubating for 10 minutes at room temperature, and washing for 3 minutes by 3 times by using PBS buffer;
(4) Dropping primary antibody: diluting the primary antibody with an antibody diluent according to a proper proportion according to a primary antibody specification, dropwise adding a proper amount of the primary antibody according to the tissue size, and standing overnight at 4 ℃; washing with PBS buffer for 3 min × 3 times;
(5) Dropwise adding a reaction enhancing solution: dripping a proper amount of reaction enhancing solution (only covering the tissue) and incubating for 20 minutes at room temperature; washing with PBS buffer for 3 min × 3 times;
(6) Dropwise adding an enhanced enzyme-labeled goat anti-rabbit IgG polymer: dripping a proper amount of an enhanced enzyme-labeled goat anti-rabbit IgG polymer, and incubating for 20 minutes at room temperature; washing with PBS buffer for 3 min × 3 times;
(7) Preparing a DAB color developing solution: adding 1 drop (about 50 microliter) of reagent 1 (DAB concentrated solution) into 1ml of reagent 2 (DAB substrate solution), and uniformly mixing to prepare DAB working solution;
(8) Counterdyeing: washing with tap water, incubating with hematoxylin staining solution for 20 s, washing with tap water to remove loose color, differentiating with 0.5% hydrochloric acid ethanol, and washing with tap water to obtain blue;
(9) And (3) dehydrating and transparency: soaking in 75% ethanol for 3 min × 2 times; after removing the redundant liquid, placing the mixture in 95 percent ethanol, and soaking for 3 minutes and 2 times; after removing the redundant liquid, placing the mixture in absolute ethyl alcohol, and soaking for 3 minutes and 3 times; after removing the redundant liquid, placing the mixture in fresh dimethylbenzene, and soaking the mixture for 10 minutes and 3 times;
(10) And (3) sealing: after neutral gum is dripped, cover glass is covered and the glass is sliced;
(11) Judging the result: the tissue staining proportion and intensity are observed, two pathologists with abundant experience assist and double-blind film reading is carried out, and the experimental result interpretation is evaluated by adopting a semi-quantitative scoring standard: according to the proportion of the positive staining cells of the corresponding index (< 10% to be judged as 0 point, 10% -40% to be judged as 1 point, 40% -70% to be judged as 2 points and more than 70% to be judged as 3 points) and the positive staining intensity of the corresponding index (0 point represents no staining: 1 point represents staining yellow: 2 point represents staining yellow: 3 point represents staining yellow), the staining indexes are obtained by adding the scores of the positive staining cells and the positive staining intensity of the corresponding index, wherein 0-3 points represent weak expression, and 4-6 points represent strong expression.
2.2.2 statistical analysis
Statistical analysis and mapping were performed using SPSS22.0 and GraphPad Prism 8. And (4) after the homogeneity of variance test conforms to normal distribution, performing difference comparison between groups by adopting an independent sample t test.
1.3 results of the experiment
Proteomics results show that in the samples of the primary focus of colorectal cancer and normal tissues beside the cancer, the expression of 171 proteins is obviously up-regulated, and the expression of 100 proteins is obviously down-regulated; in liver metastasis tissues and primary foci of colorectal cancer, 92 proteins were significantly up-regulated, and 99 proteins were significantly down-regulated (fig. 1).
And screening the differential expression protein of the normal tissue from the proteomic result into tumor-associated protein, and further analyzing the expression of the tumor-associated protein in the liver metastasis of the colorectal cancer. It was found by plotting a wien graph for two groups of differentially expressed proteins that there were 1 co-upregulated protein (IPO 7) in the two groups of differentially expressed proteins, which may be a key tumor-associated protein affecting liver metastasis in colorectal cancer (fig. 2).
In 10 paired colorectal cancer primary foci, liver metastases, and normal colorectal epithelial tissues, IPO7 was expressed higher in colorectal cancer primary foci and liver metastases than in normal tissues, and in liver metastases than in tumor tissues, consistent with proprotein results (fig. 3).
128 colorectal cancer patients matched the expression of IPO7 in the tumor tissue of colorectal cancer patients and paracarcinoma normal tissue in paraffin samples is obviously higher than that in normal tissue (p < 0.001), and the preliminary experimental results are further verified (figure 4).
Example 2 use of IPO7 in prognosis prediction of colorectal cancer
2.1 materials of the experiment
Patients and tissues: the same experimental material as in example 1,1.1, colorectal cancer, and paracarcinoma normal tissue paired large-array paraffin specimens.
2.2 Experimental methods
2.2.1 statistical analysis
The Kaplan-Meier method is adopted to draw a survival curve of the colorectal cancer patient, the significance test is carried out through a log-rank test, the relation of the clinical pathological indexes of the protein expression colorectal cancer patient is analyzed through a chi-square test, and the difference P <0.05 has statistical significance.
2.3 results of the experiment
Expression of IPO7 was clearly correlated with lymph node metastasis, distant metastasis and tumor stage in colorectal cancer patients (Table 2, p-straw 0.01). Simultaneous survival analysis showed that the overall survival rate of colorectal cancer patients with high IPO7 expression was significantly lower than that of low-expression patients (FIG. 5, p-straw 0.05)
Figure 490685DEST_PATH_IMAGE002
Figure 688448DEST_PATH_IMAGE003
Example 3 use of IPO7 in the treatment of colorectal cancer
3.1 Experimental materials
Cell line: the human colon cancer cell line DLD1 and the human embryonic kidney cell HEK293T are purchased from a national experimental cell resource sharing service platform.
3.2 Experimental methods
3.2.1 siRNA transfection
(1) Designing small interfering RNA aiming at the sequence of IPO7, wherein the sequence is shown in a table 3;
Figure 970525DEST_PATH_IMAGE004
(2) Placing the siRNA dry powder in a refrigerator at the temperature of 20 ℃ below zero for storage, centrifuging for 5 minutes at 5000rpm before use, centrifuging RNA powder on the tube wall to the tube bottom, carefully opening the tube in an ultra-clean workbench, adding a proper amount of enzyme-free water to prepare a 10 mu M solution, and subpackaging for storage;
(3) Inoculating cells to be transfected into a 6-well plate one day before transfection, wherein the density is based on the cell fusion rate of 50% in the next day of transfection;
(4) Preparing a transfection mixed solution in an ultra-clean workbench, preparing two sterile enzyme-free centrifuge tubes (a tube and a tube B) for each group of siRNA, adding 5 mu l of siRNA and 200 mu l of Opti-MEM into the tube A, adding 9 mu l of RNAiMAX and 200 mu l of Opti-MEM into the tube B, then slowly adding the liquid in the tube B into the tube A, gently mixing uniformly, and standing for 10 to 15 minutes at room temperature;
(5) And (3) replacing a fresh culture medium in a 6-hole plate, slowly adding the transfection mixed solution, shaking uniformly, placing in an incubator for continuous culture, replacing the fresh culture medium after 24 hours, and carrying out subsequent experiments after 48 to 72 hours.
3.2.2 Lentiviral packaging and transfection
(1) Inoculating HEK293T cells into a 10cm cell culture dish, wherein the density is based on the cell fusion degree of 70-80% on the next day;
(2) Preparing a transfection mixed solution in an ultra-clean workbench, preparing two sterile enzyme-free centrifuge tubes (a tube and a tube B) for each plasmid to be transfected, adding 2.5 mu g shRNA plasmid and 5.0 mu l Lenti-Pac-HIV mixture into the tube A, adding 200 mu l Opti-MEM, uniformly mixing, adding 15 mu l Endofetin and 200 mu l OptiMEM into the tube B, slowly adding the liquid in the tube B into the tube A, slightly mixing, and standing at room temperature for 10-15 minutes;
(3) Replacing the fresh culture medium for the cells, adding the transfection mixed solution into the corresponding cells for continuous culture, and replacing the fresh culture medium after 8 hours;
(4) 48 hours after transfection, the virus-containing medium was collected in a 15ml centrifuge tube, centrifuged at 4 ℃ at 2000g for 10 minutes, and after centrifugation, the supernatant was filtered through a 0.45 μm filter to remove cell debris;
(5) According to the slow virus liquid: concentrated reagent =5:1 volume lentivirus supernatant and concentrated reagent were mixed and incubated overnight at 4 ℃;
(6) 3500g of centrifugal virus liquid at 4 ℃ for 25 minutes, centrifuging, removing supernatant, adding 1ml of DMEM into the precipitate to culture and resuspend the slow virus precipitate, subpackaging the slow virus liquid and storing at-80 ℃;
(7) Before the slow virus transfects the cells, the cells are paved in a culture dish of 6cm, and the density is based on 70-80% of the fusion degree in the next day of virus transfection;
(8) On the day of infection, replacing the fresh culture medium, adding a proper amount of virus solution into the culture solution, adding Polybrene according to the proportion of 1/1000, continuously culturing for 24 hours, and then replacing the fresh culture medium for continuous culture;
(9) And observing the fluorescence expression intensity of the infected cells after 48 to 72 hours of infection, and screening the cells by using puromycin with proper concentration to obtain a stably transfected cell strain.
3.2.3 cell proliferation assay
(1) Digesting the cells of the experimental group and the control group, and adjusting the cell density to 10 4 /ml;
(2) Each group of cells of the experimental group and the control group are provided with 6 repeated holes, 100 mul of single cell suspension is respectively added into each hole of a 96-hole cell culture plate, the cells are required to be uniformly mixed before the single cell suspension is sucked each time, the same amount of PBS buffer solution is added into blank holes around the cells to reduce the influence of the evaporation of the culture medium on the experimental result, 5 96-hole plates are continuously added, and all 96-hole plates are placed in a cell culture box for continuous culture;
(3) After the cells are adhered to the wall by paving the plate for 8 to 10 hours, replacing the culture medium in the culture holes with a fresh culture medium, adding 10 mu l of CCK8 reagent into each hole, uniformly mixing the CCK8 reagent and the culture medium according to the proportion of 1:9 in advance, adding 100 mu l of CCK8 reagent into each hole in the culture holes, shaking the culture plate added with the CCK8 reagent uniformly, and then continuously putting the culture plate back to the culture box for incubation;
(4) Taking out the culture plate after 2 hours, and detecting the absorbance of the culture hole at the wavelength of 450nm by using an enzyme-labeling instrument;
(5) The above operations were repeated after 24 hours, 48 hours, 72 hours, and 96 hours from the first measurement, and the absorbance of the cells of the experimental group and the control group was obtained at different time points.
3.2.4 clone formation experiments
(1) Digesting the cells of the experimental group and the control group, and adjusting the cell density to 10 3 Per ml, 500. Mu.l of cell suspension is added to each 6-well plate, and then the culture medium is replenished for continuous culture;
(2) Changing a fresh culture medium every 3 days, continuously culturing the cells for 8 to 10 days, observing the cells, and finishing culturing when the number of cells of each cell colony is 80 to 100;
(3) Discarding the culture medium, carefully washing the cells 1~2 times with room temperature PBS, and adding 2ml of 4% tissue cell fixing solution into each hole to fix the cells for 15 minutes;
(4) Discarding the fixative, carefully washing 1~2 times with PBS, adding 2ml of crystal violet per well for dyeing for 15 minutes, slowly washing out the crystal violet dye solution with ultrapure water, taking a picture after the culture plate is air-dried, and analyzing the result with ImageJ.
3.2.5 scratch repair experiment
(1) Carrying out a scratch repair test by using a wound healing insert produced by Ibidi company, and placing the wound healing insert and forceps required by the test under ultraviolet light for disinfection 1~2 hours for later use before the test;
(2) Placing the wound healing inserts into 12-hole plates, one for each hole, and checking the adhesion degree of the inserts and the culture plate after the inserts are placed, so that the inserts are prevented from loosening in the cell culture process;
(3) Digesting and centrifuging the cells of the experimental group and the control group, and adjusting the cell density to 3~5X 10 5 Shaking uniformly, adding 80 mu l of cell suspension into each hole of the plug, and slightly shaking the culture plate to uniformly distribute the cells;
(4) Removing the plug-in unit after the cells are completely attached to the wall, carefully washing the cells which are not attached to the wall by using PBS, observing under an inverted microscope, photographing and recording, and recording the result as 0 hour;
(5) Continuously culturing the cells by using a culture medium containing 3% fetal calf serum, observing the healing condition of the scratch after 24 hours and 48 hours, and photographing and recording;
(6) The results were analyzed using ImageJ and the wound healing rate was calculated, wound healing rate = (24 or 48 hour blank area-0 hour area partial area)/0 hour blank area.
3.2.6 Transwell experiment
The Transwell experiment is divided into a migration experiment and an invasion experiment.
Migration experiment:
(1) Hydrated basement membrane: add 70. Mu.l RPMI1640 serum-free medium to each chamber, place in cell culture box and incubate for 30 minutes at 37 ℃ and carefully aspirate the residual medium;
(2) Paving a plate: the cells of the experimental group and the control group were digested, and the cell density was adjusted to 5X 10 using serum-free RPMI1640 medium 5 Adding 200 mu l of uniformly mixed single cell suspension into the upper layer of a Transwell chamber, adding 750 mu l of fresh culture medium containing 10% serum into the lower layer of the chamber, carefully shaking the culture plate to ensure that the cells in the upper chamber are uniformly distributed, and placing the culture plate in an incubator to continue culturing for 24-48 hours;
(3) Cell fixation and staining: culturing for a proper time, taking out the chamber, washing the chamber with normal-temperature PBS 1~2 times, placing the chamber in 4% tissue cell fixing solution for fixing for 15 minutes, washing 1~2 times with PBS to wash out residual fixing solution, placing the chamber in crystal violet staining solution for dyeing for 15 minutes, washing out residual crystal violet with PBS after dyeing is finished, carefully wiping the upper layer of a basement membrane with a cotton swab, and wiping out cells which do not migrate;
(4) Recording: after the liquid in the chamber is dried, the chamber is placed under a microscope for observation, 3 visual fields are randomly selected for photographing and recording, and the number of the migrated cells is counted by using ImageJ.
Invasion test:
(1) Spreading glue: melting Matrigel in a refrigerator at 4 ℃ one day before the experiment, placing a gun head and a centrifuge tube which are needed to be used in the experiment in a refrigerator at-20 ℃ for pre-cooling, and using serum-free RPMI1640 to perform the steps of 1:10, adding 70 mu l of matrigel into the upper layer of each chamber, and placing the culture plate in an incubator at 37 ℃ for 5~6 hours to solidify the matrigel;
(2) Invasion assay the rest of the procedure was identical to the migration assay.
3.2.7 cell cycle assays
(1) Collecting cells: digesting and centrifuging the cells of the experimental group and the control group, and collecting 10 6 Centrifuging the cells, removing the supernatant, re-suspending the cells by using PBS, centrifuging and removing the supernatant;
(2) Fixing: adding 1ml of PBS at room temperature for resuspending the cells again, slowly adding the cells into 3ml of precooled absolute ethanol at the temperature of-20 ℃ for overnight at the temperature of-20 ℃;
(3) Hydration: centrifuging the cells, removing ethanol, adding 5ml of PBS (phosphate buffer solution) at room temperature, standing for 15 minutes to hydrate the cells, centrifuging and removing supernatant;
(4) Dyeing: adding 1ml DNA staining reagent, mixing, incubating at 37 deg.C in dark for 30 min, and detecting on flow meter.
3.2.8 apoptosis assay
(1) Digesting and centrifuging the cells of the experimental group and the control group, counting and collecting 10 6 Cells were washed by centrifugation with pre-chilled PBS 1~2 times and 500. Mu.l of 1 XBinding Buffer resuspended cells were added to the cell pellet;
(2) Add 5. Mu.l Annexin V-FITC and 10. Mu. lPI into each tube, shake well and incubate for 5 minutes at room temperature in the dark;
(3) Annexin V-FITC detection by a FITC channel and PI detection by a PI channel on a cell flow instrument.
3.2.9 statistical analysis
Data analysis and mapping were performed using GraphPad Prism 8 and SPSS22.0, with results expressed as mean ± SD, and differences between the two groups were compared using independent sample t-test, with P <0.05 being statistically significant.
3.3 results of the experiment
4 different siRNAs (siRNA-1, siRNA-2, siRNA-3 and siRNA-4) can obviously down-regulate the expression of IPO7 mRNA, wherein the effect of low knock-out of siRNA-1 and siRNA-2 is most obvious; selecting sequences of siRNA-1 and siRNA-2 to construct shRNA, and further packaging lentivirus-transformed DLD1 cells to construct stable transgenic cell strains for knocking down IPO 7; after the stable transgenic cell strain expressing green fluorescence is obtained by puromycin screening, the knocking efficiency of IPO7 in the stable transgenic cell strain at the mRNA level and the protein level is detected again, and the result shows that the expression of IPO7 can be obviously reduced by two different interference sequences, so that the construction success of the stable transgenic cell strain with the knocked-down IPO7 is proved, and the stable transgenic cell strain can be used for the next experiment (figure 6).
The results of the clonogenic experiments showed that the proliferative capacity of DLD1 cells was significantly inhibited after downregulation of IPO7 expression compared to control cells (FIG. 7, p-woven 0.05). Meanwhile, the results of colony formation experiments showed that inhibition of IPO7 expression significantly decreased the clonogenic capacity of DLD1 cells (number of cell clones: shNC: 121.7. + -. 13.43, shIPO7-1: 87.67. + -. 11.02, shIPO7-2: 89.67. + -. 13.32, shIPO7-1vs. shNC, P < <0.05.
Scratch repair experiments show that the wound healing rate of the knocked-down IPO7 group is significantly lower than that of the control group (24-hour wound healing rate: shNC:17.4% + -0.9045%; shIPO 7-1.
Migration experiment results showed that the migration ability of DLD1 cells was significantly inhibited after interfering with IPO7 expression (number of cells reaching the lower surface of chamber: shNC: 452.3. + -. 22.01, shIPO7-1: 157.0. + -. 1.73, shIPO7-2: 161.7. + -. 10.50, shIPO7-1vs. shNC, P < -0.01, shIPO7-2vs. ShNC, p <0.05; FIG. 10A, 10B); the results of the invasion experiments showed that interfering with IPO7 expression significantly reduced the invasive capacity of DLD1 cells (number of cells reaching the lower surface of the chamber: shNC:490.0 + -25.24, shIPO7-1:206.7 + -4.04 shIPO7-2:197.7 + -24.50 shIPO7-1: vs. ShNC, P <0.01, shIPO7-2: vs. ShNC, P <0.01; FIG. 10C, 10D).
Cell cycle test results show that after down-regulating IPO7 expression, the number of cells in S phase of colon cancer cells is significantly increased (shNC: 31.16% ± 0.6929%; shIPO 7-1.
The apoptosis test results show that the IPO7 knockdown group and the control group have obvious difference in early apoptosis (the percentage of early apoptotic cells: shNC:3.687% + -0.4283%; shIPO 7-1. These results indicate that interfering IPO7 significantly promotes apoptosis of colorectal cancer, and it can be seen that IPO7 promotes tumor cell survival by inhibiting apoptosis of tumor cells (fig. 12).
Example 4 relationship between IPO7 and colorectal cancer immune cell infiltration
4.1 Experimental materials
Rabbit anti-IPO 7 monoclonal antibody, abcam; rabbit anti-CD 68 polyclonal antibody, CST; rabbit anti-CD 163 polyclonal antibody, CST; rabbit HLA-DR polyclonal antibody, CST; multi-marker immunohistochemical kit, baino biotech, beijing.
4.2 Experimental methods
4.2.1 tissue chip immunohistochemistry
Immunohistochemical staining was performed as in example 1, 1.2 of the experimental procedure 1.2.
4.2.2 Paraffin section Multi-target immunofluorescence
(1) In the multi-target immunofluorescence dyeing, a horseradish peroxidase (HRP) -labeled secondary antibody is selected to activate different fluorescent dyes, and the multi-target dyeing is realized through multiple cycles of dyeing by means of labeling of the different fluorescent dyes;
(2) After the primary antibody incubation is finished, using a washing solution to soak and wash the glass slide for 3 times, each time for 5 minutes, removing residual liquid on the glass slide, dropwise adding an HRP secondary antibody working solution, immersing the sample area, and incubating for 15 minutes at room temperature;
(3) Using washing liquid to dip and wash the glass slide for 3 times, each time for 5 minutes, removing residual liquid, then dropwise adding 100 mu l of 1 Xdye working solution on the glass slide, immersing the sample area, and incubating for 15 minutes at room temperature;
(4) Soaking and washing the glass slide for 3 times and 5 minutes each time by using a washing solution, performing microwave repair after washing is finished, naturally cooling the room temperature to the room temperature, and soaking and washing the glass slide for 3 times and 5 minutes each time by using the washing solution;
(5) Staining the next target according to the steps;
(6) DAPI was used to counterstain nuclei and incubated for 15 minutes at room temperature in the dark;
(9) And (3) sealing: mounting by using an anti-fluorescence quenching mounting agent, observing under a fluorescence microscope and collecting images.
4.2.3 statistical analysis
Download abundance data of different tumor infiltrating immune cells in the TIMER database, including B cells, CD4+ T cells, CD8+ T cells, neutrophils, macrophages and dendritic cells, and assess the correlation between IPO7 expression and six types of immune cell infiltration in colorectal cancer.
Data analysis and mapping were performed using software GraphPad Prism 8 and SPSS22.0, differences between the two groups were compared using t-test, relation between IPO7 expression and macrophage infiltration in paraffin specimens was analyzed using chi-square test, P <0.05 is statistically significant for differences.
4.3 results of the experiment
The results showed a strong correlation between IPO7 expression and macrophage infiltration in either colon or rectal cancer (Table 4)
Figure 734082DEST_PATH_IMAGE005
Expression of IPO7 was positively correlated with macrophage infiltration in colorectal cancer, with M1-type macrophage infiltration being down-regulated and M2-type macrophage infiltration being up-regulated in colorectal cancer tissues with high expression of IPO7, and M2-type macrophage infiltration being down-regulated and M1-type macrophage infiltration being up-regulated in colorectal cancer tissues with low expression of IPO7 (fig. 13).
The above description of the embodiments is only intended to illustrate the method of the invention and its core idea. It should be noted that it would be apparent to those skilled in the art that various modifications and variations can be made in the present invention without departing from the spirit of the invention, and these modifications and variations also fall within the scope of the claims of the present invention.

Claims (7)

1. Any one of the following applications:
(1) Use of an inhibitor of IPO7 or a pharmaceutical composition comprising the inhibitor for the manufacture of a product for the treatment of colorectal cancer;
(2) Use of a reagent for detecting the expression level of IPO7 or a product comprising the reagent for the manufacture of a tool for diagnosing colorectal cancer metastasis/predicting colorectal cancer prognosis;
(3) Use of IPO7 in the construction of a system/device for diagnosing colorectal cancer metastasis/predicting colorectal cancer prognosis;
(4) Use of IPO7 in the construction of a computer readable storage medium for diagnosing colorectal cancer metastasis/predicting colorectal cancer prognosis.
2. Use according to claim 1, wherein the reagent is selected from an oligonucleotide probe specifically recognizing the IPO7 gene, a primer specifically amplifying the IPO7 gene or a binding agent specifically binding to a protein encoded by the IPO7 gene.
3. The use of claim 1, wherein the product comprises a chip, a kit, or a nucleic acid membrane strip.
4. The use according to claim 1, wherein the inhibitor inhibits the proliferation, migration, invasion and/or division of colorectal cancer.
5. The use of claim 4, wherein the inhibitor comprises a nucleic acid inhibitor, a protein inhibitor.
6. The application according to claim 1, wherein the system/apparatus comprises:
an acquisition unit: for obtaining the expression level of IPO7 in the sample;
a processing unit: obtaining the colorectal cancer diagnosis metastasis/prognosis prediction result according to the expression condition of IPO7.
7. The application according to claim 1, wherein the computer readable storage medium has stored thereon a computer program which, when executed by a processor, implements the system/apparatus of claim 1 or 6.
CN202211629209.5A 2022-12-19 2022-12-19 Application of IPO7 in colorectal cancer treatment and prognosis prediction Active CN115612745B (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202211629209.5A CN115612745B (en) 2022-12-19 2022-12-19 Application of IPO7 in colorectal cancer treatment and prognosis prediction

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202211629209.5A CN115612745B (en) 2022-12-19 2022-12-19 Application of IPO7 in colorectal cancer treatment and prognosis prediction

Publications (2)

Publication Number Publication Date
CN115612745A CN115612745A (en) 2023-01-17
CN115612745B true CN115612745B (en) 2023-03-21

Family

ID=84879946

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202211629209.5A Active CN115612745B (en) 2022-12-19 2022-12-19 Application of IPO7 in colorectal cancer treatment and prognosis prediction

Country Status (1)

Country Link
CN (1) CN115612745B (en)

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2356446A4 (en) * 2008-11-14 2014-03-19 Brigham & Womens Hospital Therapeutic and diagnostic methods relating to cancer stem cells
WO2010125566A2 (en) * 2009-04-27 2010-11-04 Technion Research And Development Foundation Ltd. Markers for cancer detection
CR20180040A (en) * 2015-06-19 2018-06-18 Immatics Biotechnologies Gmbh NEW PEPTIDES AND NEW COMBINATIONS OF PEPTIDES FOR USE IN IMMUNOTHERAPY AND METHODS TO CREATE SUPPORTS FOR USE AGAINST CANCER OF PANCREAS AND OTHER TYPES OF CANCER.

Also Published As

Publication number Publication date
CN115612745A (en) 2023-01-17

Similar Documents

Publication Publication Date Title
Xia et al. Nonmuscle myosin IIA is associated with poor prognosis of esophageal squamous cancer
He et al. FOXM1 promotes tumor cell invasion and correlates with poor prognosis in early-stage cervical cancer
JP5767116B2 (en) Predicting response to platinum-based therapy
Ambrosio et al. Glucose impairs tamoxifen responsiveness modulating connective tissue growth factor in breast cancer cells
Ren et al. NFE2L3 promotes malignant behavior and EMT of human hepatocellular carcinoma (HepG2) cells via Wnt/β‑catenin pathway
Li et al. Expression and clinical implication of S100A12 in gastric carcinoma
Godlewski et al. The immunoexpression of YAP1 and LATS1 proteins in clear cell renal cell carcinoma: impact on patients’ survival
Wu et al. B-cell lymphoma/leukemia 10 promotes oral cancer progression through STAT1/ATF4/S100P signaling pathway
Mukherjee et al. Identification of EpCAM as a molecular target of prostate cancer stroma
JP2012501440A (en) ANLN protein as a predictor of endocrine treatment
CN110702923B (en) Application of GPR115 gene in preparation of anti-lung cancer drugs and diagnostic kit thereof
CN115612745B (en) Application of IPO7 in colorectal cancer treatment and prognosis prediction
US20120252748A1 (en) Methods and compositions for determining the responsiveness of cancer therapeutics
Amsterdam et al. Nuclear localization of phosphorylated ERK1 and ERK2 as markers for the progression of ovarian cancer
EP2288721A1 (en) Treatment prediction involving hmgcr protei
EP2746768A1 (en) Podxl in bladder cancer
EP2243032B1 (en) Rbm3 as a marker for breast cancer prognosis
Zhu et al. Association of TGF-β1 and WIF1 expression with 36 paired primary/recurrent nonfunctioning pituitary adenomas: A high-throughput tissue microarrays immunohistochemical study
Aoyama et al. Identification of Coiled-Coil Domain–Containing Protein 180 and Leucine-Rich Repeat–Containing Protein 4 as Potential Immunohistochemical Markers for Liposarcoma Based on Proteomic Analysis Using Formalin-Fixed, Paraffin-Embedded Tissue
US8945832B2 (en) Treatment prediction involving HMGCR
CN110564769A (en) Method for inhibiting ovarian cancer cell proliferation
EP2787350A1 (en) ASRGL1 in endometrial cancer
Hwang et al. PPIB/Cyclophilin B expression associates with tumor progression and unfavorable survival in patients with pulmonary adenocarcinoma
WO2010086400A1 (en) Combination treatment of breast cancer
EP2259059A1 (en) Means and methods for ovarian cancer prognosis

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
GR01 Patent grant
GR01 Patent grant