WO2018137701A1 - Pharmaceutical composition targeting cxcr7 and method - Google Patents
Pharmaceutical composition targeting cxcr7 and method Download PDFInfo
- Publication number
- WO2018137701A1 WO2018137701A1 PCT/CN2018/074264 CN2018074264W WO2018137701A1 WO 2018137701 A1 WO2018137701 A1 WO 2018137701A1 CN 2018074264 W CN2018074264 W CN 2018074264W WO 2018137701 A1 WO2018137701 A1 WO 2018137701A1
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- cxcr7
- cxcr4
- protein
- expression
- drug
- Prior art date
Links
Images
Classifications
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K45/00—Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61L—METHODS OR APPARATUS FOR STERILISING MATERIALS OR OBJECTS IN GENERAL; DISINFECTION, STERILISATION OR DEODORISATION OF AIR; CHEMICAL ASPECTS OF BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES; MATERIALS FOR BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES
- A61L29/00—Materials for catheters, medical tubing, cannulae, or endoscopes or for coating catheters
- A61L29/14—Materials characterised by their function or physical properties, e.g. lubricating compositions
- A61L29/16—Biologically active materials, e.g. therapeutic substances
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61L—METHODS OR APPARATUS FOR STERILISING MATERIALS OR OBJECTS IN GENERAL; DISINFECTION, STERILISATION OR DEODORISATION OF AIR; CHEMICAL ASPECTS OF BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES; MATERIALS FOR BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES
- A61L31/00—Materials for other surgical articles, e.g. stents, stent-grafts, shunts, surgical drapes, guide wires, materials for adhesion prevention, occluding devices, surgical gloves, tissue fixation devices
- A61L31/14—Materials characterised by their function or physical properties, e.g. injectable or lubricating compositions, shape-memory materials, surface modified materials
- A61L31/16—Biologically active materials, e.g. therapeutic substances
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P35/00—Antineoplastic agents
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P7/00—Drugs for disorders of the blood or the extracellular fluid
- A61P7/02—Antithrombotic agents; Anticoagulants; Platelet aggregation inhibitors
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P9/00—Drugs for disorders of the cardiovascular system
-
- G—PHYSICS
- G01—MEASURING; TESTING
- G01N—INVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
- G01N33/00—Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
- G01N33/48—Biological material, e.g. blood, urine; Haemocytometers
- G01N33/50—Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
- G01N33/68—Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids
-
- G—PHYSICS
- G01—MEASURING; TESTING
- G01N—INVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
- G01N33/00—Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
- G01N33/48—Biological material, e.g. blood, urine; Haemocytometers
- G01N33/50—Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
- G01N33/68—Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids
- G01N33/6803—General methods of protein analysis not limited to specific proteins or families of proteins
- G01N33/6806—Determination of free amino acids
- G01N33/6812—Assays for specific amino acids
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61L—METHODS OR APPARATUS FOR STERILISING MATERIALS OR OBJECTS IN GENERAL; DISINFECTION, STERILISATION OR DEODORISATION OF AIR; CHEMICAL ASPECTS OF BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES; MATERIALS FOR BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES
- A61L2300/00—Biologically active materials used in bandages, wound dressings, absorbent pads or medical devices
- A61L2300/60—Biologically active materials used in bandages, wound dressings, absorbent pads or medical devices characterised by a special physical form
- A61L2300/606—Coatings
Definitions
- the present invention relates to pharmaceutical compositions and methods for treating cardiovascular diseases.
- the present invention relates to a method and a pharmaceutical composition for vascular endothelial injury disease in a subject to be treated with CXCR7 or to improve cardiac remodeling after myocardial infarction in a subject.
- GWA genome-wide association study
- CXCL12 locus which encodes the chemokine CXCL12, also known as stromal cell-derived factor-1, SDF1
- CAD coronary artery disease
- MI myocardial infarction
- Higher plasma CXCL12 is associated with myocardial infarction (MI) and death events in patients with chronic kidney disease (European heart journal. 2014; 35: 2115-2122), and a prospective study of the Framingham Heart Study, which is also associated with heart failure Associated with total mortality (Arteriosclerosis, thrombosis, and vascular biology. 2014; 34: 2100-2105).
- CXCL12 has two receptors: CXCR4, a classical G-protein coupled receptor (GPCR), and CXCR7, which was discovered in 2005 as the second receptor for CXCL12 (The Journal of biological chemistry .2005;280:35760-35766).
- CXCR4 is thought to be involved in vascular remodeling (Circulation. 2003; 108: 2491-2497; Circulation research. 2005; 96: 784-791; Arteriosclerosis, thrombosis, and vascular biology. 2014; 34: 1209-1220; Thrombosis and haemostasis. 2012 ; 107: 356-368), atherosclerosis (Circulation research.
- CXCR7 myocardial infarction
- a decrease in CXCR4 (rather than CXCR7) expression is associated with all-cause death and/or MI joint end points (Journal of thrombosis and haemostasis: JTH. 2015; 13: 719-728).
- CXCR7 is closely related to chemokine receptors in phylogeny, binds to CXCL12 with higher affinity than CXCR4, transmits signals through ⁇ -arrestin (rather than classical G-protein), but cannot be coupled with G protein to induce A typical chemokine receptor-mediated cellular response (J Exp Med. 2006; 203: 2201-2213).
- CXCR7 was previously thought to act as a scavenging receptor for CXCL12, mediating efficient endocytosis and degradation (PLoS One. 2010; 5: e9175; Cell. 2008; 132: 463-473; Proc Natl Acad Sci USA. 2010; 107: 628-632).
- CXCR7 also has signaling activity beyond ligand clearance, including signaling activity in tumor cell growth and organ regeneration (Proc Natl Acad Sci USA. 2007; 104: 15735-15740; J Biol Chem. 2008 ; 283: 4283-4294; Mol Cancer. 2014; 13: 198; Nature medicine. 2016; 22: 154-162).
- CXCR7 is expressed in brain, heart, kidney, endothelial and tumor cells (J Exp Med. 2006; 203: 2201-2213; PLoS One. 2011; 6: e20680). It is widely expressed in tumor vascular endothelium (Proc Natl Acad Sci USA. 2007; 104: 15735-15740) and is induced by hypoxia (PLoS One. 2013; 8: e55290). Platelets express both CXCR4 and CXCR7 (European heart journal. 2014; 35: 386-394), but CXCR7 protein is not expressed on leukocytes in human or mouse blood (Journal of Immunology. 2010; 185: 5130-5139).
- CXCR7-deficient mice die from abnormal heart valves before and after birth (Proc Natl Acad Sci USA. 2007; 104: 14759-14764). Weber et al. used hyperlipidemia Apoe -/- mice to confirm that the overall knockout of CXCR7 exacerbates atherosclerosis due to defects in cholesterol uptake in adipose tissue (Circulation. 2014; 129: 1244-1253) .
- the invention provides a method of treating or preventing a cardiovascular disease in a subject comprising administering to the subject an effective amount of a first drug that increases the expression and/or activity of a CXCR7 protein.
- the present invention provides a method of treating a vascular endothelial injury disease in a subject or ameliorating cardiac remodeling after myocardial infarction in a subject, comprising administering to the subject an effective amount of a first drug that increases expression and/or activity of a CXCR7 protein. .
- the method of the present invention for treating or preventing cardiovascular disease in a subject further comprises administering to the subject an effective amount of a second drug that reduces expression and/or activity of the CXCR4 protein.
- the methods of the invention further comprise administering to the subject a drug that inhibits platelet activation and/or aggregation.
- the method of the invention further comprises administering to the subject a drug that stabilizes the plaque.
- the invention also provides a pharmaceutical composition for treating or preventing a cardiovascular disease in a subject, comprising an effective amount of a first drug for increasing the expression and/or activity of a CXCR7 protein.
- a pharmaceutical composition for treating a vascular endothelial injury disease in a subject or ameliorating cardiac remodeling after myocardial infarction in a subject comprising an effective amount of an amount for increasing the expression and/or activity of the CXCR7 protein. a drug.
- the pharmaceutical composition further comprises an effective amount of a selective antagonist of CXCR4 or a nucleic acid molecule or expression vector that inhibits expression of the CXCR4 protein.
- the pharmaceutical composition is for use in combination with an effective amount of a selective antagonist comprising CXCR4 or a nucleic acid molecule or expression vector that inhibits expression of a CXCR4 protein.
- the pharmaceutical composition is for use in combination with a drug that inhibits platelet activation and/or aggregation.
- the pharmaceutical composition is for use in combination with a drug that stabilizes the plaque.
- the present invention also provides the use of a first medicament for increasing the expression and/or activity of a CXCR7 protein for the preparation of a pharmaceutical composition for treating or preventing a cardiovascular disease in a subject.
- the pharmaceutical composition further comprises an effective amount of a selective antagonist of CXCR4 or a nucleic acid molecule or expression vector that inhibits expression of the CXCR4 protein.
- the pharmaceutical composition is used in combination with an effective amount of a selective antagonist comprising CXCR4 or a nucleic acid molecule or expression vector that inhibits expression of a CXCR4 protein.
- the pharmaceutical composition is for use in combination with a medicament for inhibiting platelet activation and/or aggregation.
- the pharmaceutical composition is for use in combination with a drug that stabilizes the plaque.
- the disease is selected from the group consisting of thrombosis, thromboembolism, vascular wall injury, vascular stenosis after injury, vascular restenosis after PCI and Bypass, coronary heart disease, myocardial deficiency Blood, myocardial infarction, heart failure after myocardial infarction, arrhythmia after myocardial infarction, and any combination thereof.
- the invention also provides a vascular stent or catheter with a balloon, wherein the surface of the stent or balloon is coated with an effective amount of a first drug for increasing the expression and/or activity of the CXCR7 protein.
- the surface of the stent or balloon is further coated with an effective amount of a selective antagonist of CXCR4 or a nucleic acid molecule or expression that inhibits expression of the CXCR4 protein.
- Carrier In one embodiment of the vascular stent or balloon with a balloon of the invention, the surface of the stent or balloon is further coated with an effective amount of a selective antagonist of CXCR4 or a nucleic acid molecule or expression that inhibits expression of the CXCR4 protein. Carrier.
- the vascular stent or the balloon-equipped catheter of the present invention is for treating or preventing vascular injury and/or myocardial ischemia-related disease in a subject selected from the group consisting of thrombosis, thromboembolism, blood vessel wall damage, Vascular stenosis after injury, vascular restenosis after PCI and Bypass, coronary heart disease, myocardial ischemia, myocardial infarction, heart failure after myocardial infarction, arrhythmia after myocardial infarction, and any combination thereof.
- the present invention also provides a method of treating or preventing a thrombosis-related disease in a subject, comprising administering to the subject an effective amount of a drug that reduces the level or activity of circulating CXCL12, or a selective antagonist of CXCR4, or inhibiting CXCR4 protein expression.
- a drug that reduces the level or activity of circulating CXCL12, or a selective antagonist of CXCR4, or inhibiting CXCR4 protein expression Nucleic acid molecule or expression vector, or a combination thereof.
- the present invention also provides a pharmaceutical composition for treating or preventing a thrombosis-related disease in a subject, which comprises an effective amount of a drug which lowers the level or activity of CXCL12 in circulation, or a selective antagonist of CXCR4, or inhibits CXCR4 protein expression.
- a pharmaceutical composition for treating or preventing a thrombosis-related disease in a subject which comprises an effective amount of a drug which lowers the level or activity of CXCL12 in circulation, or a selective antagonist of CXCR4, or inhibits CXCR4 protein expression.
- Nucleic acid molecule or expression vector, or a combination thereof are examples of nucleic acid molecule or expression vector, or a combination thereof.
- the present invention also provides a medicament for reducing the level or activity of CXCL12 in circulation, or a selective antagonist of CXCR4, or a nucleic acid molecule or expression vector for inhibiting expression of CXCR4 protein, or a combination thereof for preparing thrombus for treating or preventing a subject Use in a pharmaceutical composition of a related disease.
- the invention also provides a pharmaceutical composition for treating cancer comprising a CXCR7 inhibitor, wherein the CXCR7 inhibitor does not increase blood CXCL12 levels when administered to a subject.
- the invention also provides a method of screening for a medicament for treating cancer with high cardiovascular safety, comprising:
- the activity of the CXCR7 protein in the tissue sample of the animal is decreased relative to the activity of the CXCR7 protein in the same tissue sample of the control animal to which the drug candidate is not administered, and the content of CXCL12 protein in the blood sample of the animal is not administered.
- the level of CXCL12 protein in the blood sample of the control animal of the drug candidate is comparable or lower, suggesting that the drug candidate is a drug for treating cancer with high cardiovascular safety.
- the invention also provides methods of screening for a medicament useful for treating or preventing a cardiovascular disease, comprising:
- the expression level and/or activity of the CXCR7 protein in the tissue sample of the animal relative to the expression level and/or activity of the CXCR7 protein in the same tissue sample of the control animal to which the drug candidate is not administered is indicative of the candidate drug Can treat or prevent cardiovascular disease.
- AD Proliferative femoral artery (B) 28 days after injury in the healthy femoral artery (A) and guidewire in mice, and CXCR7 in aortic sections (C, D) from patients with aortic dissection Red) and vWF (endothelial cell marker, green) were immunofluorescently stained and DAPI stained with nuclei (blue). At the far right is the combination of the three colors.
- the gray photo in C1 shows the staining position in Figure 1C at a lower magnification. The arrow points to a severe injury. Representative sections from three independent stains are shown.
- L lumen
- P plaque
- Bar 50 ⁇ m.
- A-F CXCR7 mRNA expression (A) in mouse lung endothelial cells (MLEC) isolated from endothelial CXCR7 conditional knockout mice (cKO) and littermate control (Ctl) was detected by RT-PCR.
- DAPI stained with nuclei blue.
- C neointimal formation
- D intima to media ratio
- E median thickness remained unchanged
- IL-1 ⁇ (10 ng/mL) treatment increased CXCR7 mRNA (A) and protein (B) levels in cultured mouse lung endothelial cells (MLEC).
- IL-1 ⁇ (10 ng/mL) promotes cell growth.
- Drug inhibition (CCX771) or gene deletion CXCR7 inhibits cell proliferation in MLEC (C) and mouse aortic endothelial cells (MAEC) (D).
- MAEC immunoblot analysis indicated that IL-1 ⁇ increased the ERK signaling pathway, which is inhibited by CXCR7 inhibition or deletion (E, F).
- CCX771 (G) or CXCR7 (H) knockdown reduces cell proliferation when stimulated with IL-1 ⁇ .
- Each experiment was conducted no less than 3 times. *, p ⁇ 0.05; **, p ⁇ 0.01.
- CXCR4 and CXCL12 in cultured mouse lung endothelial cells (MLEC) treated with IL-1 ⁇ .
- CXCR4 (A) and CXCL12 (B) mRNA were detected by RT-PCR before and 6 hours after stimulation with IL-1 ⁇ (10 ng/mL).
- CXCR4 protein levels (C) of cells incubated with IL-1 ⁇ (10 ng/mL) for 0, 6, 12 and 24 hours were detected by immunoblotting.
- J Effect of drug inhibition of CXCR7 on proliferation of unstimulated endothelial cells. Proliferation studies were performed in mouse lung endothelial cells (MLEC; A) and mouse aortic endothelial cells (MAEC; B) that were not stimulated by IL-1 ⁇ . No statistical significance.
- CXCR7 inhibition reduces TNF ⁇ -induced endothelial cell proliferation and angiogenesis.
- TNF ⁇ -induced CXCR7 protein expression A
- B endothelial cell proliferation
- D silencing
- Bar 200 ⁇ m.
- HUVEC HUVEC
- HAEC human aortic endothelial cells
- si-CXCR7 si-CXCR7 or negative si-RNA (si-Neg)
- Bar 500 ⁇ m.
- vascular density (H) in the muscle space (IS) of the gastrocnemius muscle was measured by immunostaining of vWF.
- vWF green
- DAPI blue
- EF ejection fraction
- FAC left ventricular sectional area change score
- E/A ratio of early mitral peak to end-diastolic filling rate
- LVAWd Left ventricular anterior wall thickness
- CXCR7 is expressed in Ctl mouse endothelial cells after MI, but not in cKO endothelial cells (G).
- MI increases CXCL12 plasma levels in Ctl and cKO mice. Compared to Ctl, cKO showed higher plasma CXCL12 levels before MI (defined as "0") and after MI. *, p ⁇ 0.05vs.Ctl; 0 in p ⁇ 0.05 vs. cKO; 0 in p ⁇ 0.05 vs. Ctl.
- A-F Expression of CXCR7 was confirmed in a 293T cell line transfected with adenovirus expressing CXCR7 (Ad-CXCR7) (A).
- EF ejection fraction
- LVAWd diastolic left ventricular anterior wall thickness
- D, F; n 10 Ad-Neg, 11 Ad-CXCR7.
- A-F Conditional knockout (cKO) of endothelial CXCR7 induced by tamoxifen treatment is described in detail in the method and is used to determine plasma CXCL12 and photochemically induced thrombosis.
- Wild type mice (C57BL/6) were treated by subcutaneous injection of 20 mg/kg CCX771 or vehicle, and plasma CXCL12 and photochemically induced thrombosis were examined two hours later. Representative carotid blood flow during induced thrombosis is shown in the accompanying drawings (A&C).
- A-F Intravenous infusion of CXCL12 increases circulating CXCL12 levels (A&B) and enhances collagen-induced ex vivo whole blood accumulation (C&D).
- CXCL12 infusion accelerates photochemically induced thrombosis (E&F) and AMD3100 (CXCR4-specific inhibitor) eliminates accelerated thrombosis.
- E&F photochemically induced thrombosis
- AMD3100 CXCR4-specific inhibitor
- A-B AMD3100 treatment abolishes the prothrombotic response caused by loss of endothelial CXCR7.
- A shows representative carotid blood flow during induced thrombosis.
- B represents the results of statistical analysis of occlusion time in each group.
- Platelets significantly contribute to the increase in circulating CXCL12 after endothelial CXCR7 removal
- A-H Platelet agonist U46619 (A) or collagen (B) triggers CXCL12 release when whole blood is treated ex vivo.
- Intravenous injection of U46619 (20 ⁇ g/mouse) reduced platelet counts in 3 minutes (Fig. 4C) and increased plasma CXCL12 (Fig. 4D).
- Platelet depletion by anti-CD41 antibodies reduced circulating CXCL12 in cKO and Ctl, but elevated CXCL12 levels were maintained in cKO mice (Fig. 4G).
- the proportion of CXCL12 decreased per million platelets in cKO was higher than that of control (H).
- Flow cytometric analysis of P-selectin (CD62P) expression in platelets showed higher CD62P expression and activated platelets (E&F) in cKO compared to Ctl.
- I Platelet depletion and recovery time.
- Figure 15A-B Correlation of CXCL12 levels in human blood circulation with platelet reactivity.
- CXCR7 plays a key role in maintaining endothelial integrity. Specifically, the present inventors have found that CXCR7 is expressed in damaged blood vessels, which can be induced by inflammatory factors released by vascular injury, and promotes proliferation of inflammation-associated endothelial cells; whereas deletion of endothelial CXCR7 promotes repair of endothelial cells Reduced blood vessels caused by stenosis. The inventors have also discovered that endothelial CXCR7 also plays a key role in ischemia-induced angiogenesis.
- Angiogenesis is a process of endothelial cell dependence, which results in the formation of new blood vessels, which is necessary for the regeneration of vascular regeneration after cardiomyocytes and the promotion of tissue regeneration after ischemia injury. More importantly, the inventors found that delivery of the CXCR7 gene to the myocardium by the adenovirus improves cardiac function after MI and reduces myocardial infarct size. Therefore, activation of CXCR7 will be able to treat vascular endothelial injury or improve cardiac remodeling after myocardial infarction.
- the invention provides a method of treating or preventing a cardiovascular disease in a subject comprising administering to the subject an effective amount of a first drug that increases the expression and/or activity of a CXCR7 protein.
- the present invention provides a method of treating a vascular endothelial injury disease in a subject or ameliorating cardiac remodeling after myocardial infarction in a subject, comprising administering to the subject an effective amount of a first drug that increases expression and/or activity of a CXCR7 protein. .
- the term "subject” refers to a mammal, preferably a primate, more preferably a human.
- the first medicament comprises a selective agonist of CXCR7, an expression vector comprising a polynucleotide encoding a CXCR7 protein or a functional fragment thereof, or a combination thereof.
- the selective agonist of CXCR7 is selected from the group consisting of an activated antibody of CXCR7, an activated ligand of CXCR7, TC14012 or a functional analog thereof, and combinations thereof.
- the method of treating or preventing cardiovascular disease in a subject of the present invention further comprises administering to the subject an effective amount of a second drug that reduces expression and/or activity of the CXCR4 protein.
- the second drug comprises a selective antagonist of CXCR4, a nucleic acid molecule or expression vector that inhibits expression of a CXCR4 protein, or a combination thereof.
- the selective antagonist of CXCR4 is selected from the group consisting of AMD3100 or a functional analog thereof, a blocking antibody to CXCR4 or an antigen binding fragment thereof, TC14012 or a functional analog thereof, and combinations thereof.
- the nucleic acid molecule that inhibits expression of a CXCR4 protein is an siRNA or a precursor thereof that targets a transcription product of the CXCR4 gene. In another specific embodiment, the nucleic acid molecule that inhibits expression of a CXCR4 protein is an antisense RNA that targets a transcription product of the CXCR4 gene.
- the methods of the invention further comprise administering to the subject a drug that inhibits platelet activation and/or aggregation. In another embodiment, the methods of the invention further comprise administering to the subject a drug that stabilizes the plaque, such as a statin.
- the subject is administered to the subject by oral administration, buccal administration, inhalation, intravenous injection, intraarterial injection, intramuscular injection, subcutaneous injection, intraperitoneal injection or topical administration. And / or a second drug.
- the administration is achieved by intracoronary administration or by coating the first and/or second medicament on a balloon stent or a balloon of a catheter with a balloon. Topical application.
- the invention also provides a pharmaceutical composition for treating or preventing a cardiovascular disease in a subject, comprising an effective amount of a first drug for increasing the expression and/or activity of a CXCR7 protein.
- a pharmaceutical composition for treating a vascular endothelial injury disease in a subject or ameliorating cardiac remodeling after myocardial infarction in a subject comprising an effective amount of an amount for increasing the expression and/or activity of the CXCR7 protein. a drug.
- the first drug comprises a selective agonist of CXCR7, an expression vector comprising a polynucleotide encoding a CXCR7 protein or a functional fragment thereof, or a combination thereof.
- the selective agonist of CXCR7 is selected from the group consisting of an activated antibody of CXCR7, an activated ligand of CXCR7, TC14012 or a functional analog thereof, and combinations thereof.
- the pharmaceutical composition further comprises an effective amount of a selective antagonist of CXCR4 or a nucleic acid molecule or expression vector that inhibits expression of the CXCR4 protein.
- the pharmaceutical composition is for use in combination with an effective amount of a selective antagonist comprising CXCR4 or a nucleic acid molecule or expression vector that inhibits expression of a CXCR4 protein.
- the selective antagonist of CXCR4 is selected from the group consisting of AMD3100 or a functional analog thereof, a blocking antibody to CXCR4 or an antigen binding fragment thereof, TC14012 or a functional analog thereof, and combinations thereof.
- the nucleic acid molecule that inhibits expression of a CXCR4 protein is an siRNA or a precursor thereof that targets a transcription product of the CXCR4 gene. In another specific embodiment, the nucleic acid molecule that inhibits expression of a CXCR4 protein is an antisense RNA that targets a transcription product of the CXCR4 gene.
- the pharmaceutical composition is for use in combination with a drug that inhibits platelet activation and/or aggregation. In another embodiment, the pharmaceutical composition is for use in combination with a drug that stabilizes the plaque, such as a statin.
- the pharmaceutical composition is for administration to a subject by oral, buccal, inhalation, intravenous, intraarterial, intramuscular, subcutaneous, intraperitoneal or topical administration.
- the pharmaceutical composition is for intracoronary administration.
- the present invention also provides the use of a first medicament for increasing the expression and/or activity of a CXCR7 protein for the preparation of a pharmaceutical composition for treating or preventing a cardiovascular disease in a subject.
- the first medicament comprises an effective amount of a selective agonist of CXCR7, an expression vector comprising a polynucleotide encoding a CXCR7 protein or a functional fragment thereof, or a combination thereof.
- the selective agonist of CXCR7 is selected from the group consisting of an activated antibody of CXCR7, an activated ligand of CXCR7, TC14012 or a functional analog thereof, and combinations thereof.
- the pharmaceutical composition further comprises an effective amount of a selective antagonist of CXCR4 or a nucleic acid molecule or expression vector that inhibits expression of the CXCR4 protein.
- the pharmaceutical composition is used in combination with an effective amount of a selective antagonist comprising CXCR4 or a nucleic acid molecule or expression vector that inhibits expression of a CXCR4 protein.
- the selective antagonist of CXCR4 is selected from the group consisting of AMD3100 or a functional analog thereof, a blocking antibody to CXCR4 or an antigen binding fragment thereof, TC14012 or a functional analog thereof, and combinations thereof.
- the nucleic acid molecule that inhibits expression of a CXCR4 protein is an siRNA or a precursor thereof that targets a transcription product of the CXCR4 gene. In another specific embodiment, the nucleic acid molecule that inhibits expression of a CXCR4 protein is an antisense RNA that targets a transcription product of the CXCR4 gene.
- the pharmaceutical composition is for use in combination with a medicament for inhibiting platelet activation and/or aggregation.
- the pharmaceutical composition is for use in combination with a drug that stabilizes the plaque, such as a statin.
- the pharmaceutical composition is for administration to a subject by oral, buccal, inhalation, intravenous, intraarterial, intramuscular, subcutaneous, intraperitoneal or topical administration.
- the pharmaceutical composition is formulated in a form for intracoronary administration.
- the disease is selected from the group consisting of thrombosis, thromboembolism, vascular wall injury, vascular stenosis after injury, vascular restenosis after PCI and Bypass, coronary heart disease, myocardial deficiency Blood, myocardial infarction, heart failure after myocardial infarction, arrhythmia after myocardial infarction, and any combination thereof.
- the invention also provides a vascular stent or catheter with a balloon, wherein the surface of the stent or balloon is coated with an effective amount of a first drug for increasing the expression and/or activity of the CXCR7 protein.
- the first drug comprises a selective agonist of CXCR7, an expression vector comprising a polynucleotide encoding a CXCR7 protein or a functional fragment thereof, or combination.
- the selective agonist of CXCR7 is selected from the group consisting of an activated antibody of CXCR7, an activated ligand of CXCR7, TC14012 or a functional analog thereof, and combinations thereof.
- the surface of the stent or balloon is further coated with an effective amount of a selective antagonist of CXCR4 or a nucleic acid molecule that inhibits expression of the CXCR4 protein or Expression vector.
- the selective antagonist of CXCR4 is selected from the group consisting of AMD3100 or a functional analog thereof, a blocking antibody to CXCR4 or an antigen binding fragment thereof, and combinations thereof.
- the nucleic acid molecule that inhibits expression of a CXCR4 protein is an siRNA or a precursor thereof that targets a transcription product of the CXCR4 gene.
- the nucleic acid molecule that inhibits expression of a CXCR4 protein is an antisense RNA that targets a transcription product of the CXCR4 gene.
- the vascular stent or the balloon-equipped catheter of the present invention is for treating or preventing vascular injury and/or myocardial ischemia-related disease in a subject selected from the group consisting of thrombosis, thromboembolism, blood vessel wall damage, Vascular stenosis after injury, vascular restenosis after PCI and Bypass, coronary heart disease, myocardial ischemia, myocardial infarction, heart failure after myocardial infarction, arrhythmia after myocardial infarction, and any combination thereof.
- the vascular injury-related disease is coronary plaque and stenosis.
- the myocardial ischemia related disease is myocardial infarction.
- CXCL12 causes a tendency to thrombosis at a concentration having pathophysiological significance.
- the inventors have also surprisingly found that the sensitizing effect of platelet activation is caused by circulating CXCL12, rather than the vessel wall CXCL12.
- the present inventors have further found that the trapped receptor endothelial CXCR7 of CXCL12 is essential for maintaining the physiological level of CXCL12, while inhibiting CXCR7 leads to an increase in CXCL12, which acts on platelet CXCR4, thereby promoting thrombus formation.
- the present invention also provides a method of treating or preventing a thrombosis-related disease in a subject, comprising administering to the subject an effective amount of a drug that reduces the level or activity of CXCL12 in circulation, or a selective antagonist of CXCR4, or inhibiting CXCR4 A nucleic acid molecule or expression vector, or a combination thereof, expressed by a protein.
- the drug that reduces the level of circulating CXCL12 is a drug that increases the expression and/or activity of a CXCR7 protein, such as an expression vector comprising a polynucleotide encoding a CXCR7 protein or a functional fragment thereof.
- the drug that reduces the level or activity of CXCL12 in the circulation is an anti-CXCL12 antibody.
- the selective antagonist of CXCR4 is selected from the group consisting of AMD3100 or a functional analog thereof, a blocking antibody to CXCR4 or an antigen binding fragment thereof, and combinations thereof.
- the nucleic acid molecule that inhibits expression of a CXCR4 protein is an siRNA or a precursor thereof that targets a transcription product of the CXCR4 gene.
- the nucleic acid molecule that inhibits expression of a CXCR4 protein is an antisense RNA that targets a transcription product of the CXCR4 gene.
- the present invention also provides a pharmaceutical composition for treating or preventing a thrombosis-related disease in a subject, which comprises an effective amount of a drug which lowers the level or activity of CXCL12 in circulation, or a selective antagonist of CXCR4, or inhibits CXCR4 protein expression.
- the drug that reduces the level of circulating CXCL12 is a drug that increases the expression and/or activity of a CXCR7 protein, such as an expression vector comprising a polynucleotide encoding a CXCR7 protein or a functional fragment thereof.
- the drug that reduces the level or activity of CXCL12 in the circulation is an anti-CXCL12 antibody.
- the selective antagonist of CXCR4 is selected from the group consisting of AMD3100 or a functional analog thereof, a blocking antibody to CXCR4 or an antigen binding fragment thereof, and combinations thereof.
- the nucleic acid molecule that inhibits expression of a CXCR4 protein is an siRNA or a precursor thereof that targets a transcription product of the CXCR4 gene.
- the nucleic acid molecule that inhibits expression of a CXCR4 protein is an antisense RNA that targets a transcription product of the CXCR4 gene.
- the present invention also provides a medicament for reducing the level or activity of CXCL12 in circulation, or a selective antagonist of CXCR4, or a nucleic acid molecule or expression vector for inhibiting expression of CXCR4 protein, or a combination thereof for preparing thrombus for treating or preventing a subject Use in a pharmaceutical composition of a related disease.
- the drug that reduces the level of circulating CXCL12 is a drug that increases the expression and/or activity of a CXCR7 protein, such as an expression vector comprising a polynucleotide encoding a CXCR7 protein or a functional fragment thereof.
- the drug that reduces the level or activity of CXCL12 in the circulation is an anti-CXCL12 antibody.
- the selective antagonist of CXCR4 is selected from the group consisting of AMD3100 or a functional analog thereof, a blocking antibody to CXCR4 or an antigen binding fragment thereof, and combinations thereof.
- the nucleic acid molecule that inhibits expression of a CXCR4 protein is an siRNA or a precursor thereof that targets a transcription product of the CXCR4 gene.
- the nucleic acid molecule that inhibits expression of a CXCR4 protein is an antisense RNA that targets a transcription product of the CXCR4 gene.
- CXCR7 inhibitors have been proposed for the treatment of diseases such as cancer.
- an inhibitor such as CCX771
- CCX771 increases the risk of cardiovascular thrombosis by increasing the level of CXCL12 in the blood. Therefore, in any treatment using a CXCR7 inhibitor such as cancer treatment, it is preferred to use a CXCR7 inhibitor that does not increase the level of CXCL12.
- the invention also provides a pharmaceutical composition for treating cancer comprising a CXCR7 inhibitor, wherein the CXCR7 inhibitor does not increase blood CXCL12 levels when administered to a subject.
- the cancer includes, but is not limited to, liver cancer, breast cancer, and the like.
- the invention also provides a method of screening for a medicament for treating cancer with high cardiovascular safety, comprising:
- the activity of the CXCR7 protein in the tissue sample of the animal is decreased relative to the activity of the CXCR7 protein in the same tissue sample of the control animal to which the drug candidate is not administered, and the content of CXCL12 protein in the blood sample of the animal is not administered.
- the level of CXCL12 protein in the blood sample of the control animal of the drug candidate is comparable or lower, suggesting that the drug candidate is a drug for treating cancer with high cardiovascular safety.
- the invention also provides methods of screening for a medicament useful for treating or preventing a cardiovascular disease, comprising:
- the expression level and/or activity of the CXCR7 protein in the tissue sample of the animal relative to the expression level and/or activity of the CXCR7 protein in the same tissue sample of the control animal to which the drug candidate is not administered is indicative of the candidate drug Can treat or prevent cardiovascular disease.
- the activity of the CXCR7 protein is selected from the group consisting of promoting vascular endothelial proliferation, promoting angiogenesis, promoting damage to blood vessel repair, and reducing Myocardial infarct size, improved myocardial remodeling after infarction, and improved cardiac function after infarction.
- the above pharmaceutical or pharmaceutical compositions of the present invention may further comprise a pharmaceutically acceptable carrier.
- pharmaceutically acceptable refers to molecular entities and compositions that do not produce an undesirable, allergic or other untoward reaction when administered to an animal, such as a human, as desired.
- the preparation of suitable pharmaceutical compositions is known to those skilled in the art in light of the present disclosure and is exemplified in "Remington: The Science and Practice of Pharmacy," 21st Edition, 2005, which is incorporated herein by reference.
- human administration it should be understood that the preparation should also meet the criteria for sterility, pyrogenicity, overall safety, and purity required by the drug approval authority.
- pharmaceutically acceptable carrier includes any and all solvents, dispersion media, antioxidants, salts, coatings, surfactants, preservatives (eg, methyl or propyl paraben, sorbic acid, antibacterial).
- Agent antifungal agent
- isotonic agent such as paraffin
- adsorbent for example, kaolin, bentonite
- drug stabilizer for example, sodium lauryl sulfate
- gel adhesive
- adhesive eg, syrup, gum arabic, gelatin, sorbitol, tragacanth, polyvinylpyrrolidone, carboxymethylcellulose, alginate
- excipients eg, lactose, polyethylene glycol
- disintegrants eg Agar, starch, lactose, calcium phosphate, calcium carbonate, alginic acid, sorbitol, glycine
- wetting agents eg, cetyl alcohol, glyceryl monostearate
- lubricants eg, quarters) Ammonium salt
- edible oil eg, almond oil, coconut oil, oily ester or propylene glycol
- sweeteners eg, flavoring agents, coloring agents, fillers (eg, starch, lac
- the composition can comprise a plurality of antioxidants to retard oxidation of one or more components.
- antioxidants include ascorbic acid, cysteine hydrochloride, sodium sulfite, sodium hydrogensulfite, sodium metabisulfite, ascorbyl palmitate, butylated hydroxytoluene, butylated hydroxyanisole, lecithin, propyl gallate, and tocopherol.
- microorganisms can be achieved by the use of preservatives such as various antibacterial and antifungal agents including, but not limited to, parabens (for example, methylparaben, p-hydroxyl Propyl benzoate), chlorobutanol, phenol, sorbic acid, thimerosal or a combination thereof.
- preservatives such as various antibacterial and antifungal agents including, but not limited to, parabens (for example, methylparaben, p-hydroxyl Propyl benzoate), chlorobutanol, phenol, sorbic acid, thimerosal or a combination thereof.
- the carrier can be a solvent or dispersion medium including, but not limited to, water, ethanol, polyol (eg, glycerol, propylene glycol, liquid polyethylene glycol, etc.), liquid ( For example, triglycerides, vegetable oils, liposomes, and combinations thereof.
- a coating such as lecithin
- a surfactant For example, hydroxypropyl cellulose
- prolonged absorption of the injectable compositions can be brought about by the use of agents that delay absorption (e.g., aluminum monostearate, gelatin, or a combination thereof) in the compositions.
- agents that delay absorption e.g., aluminum monostearate, gelatin, or a combination thereof
- an effective amount refers to an amount of a substance, compound, material, or composition comprising a compound that is at least sufficient to produce a therapeutic effect after administration to a subject. Thus, it is an amount necessary to prevent, cure, ameliorate, block or partially arrest the symptoms of a disease or condition.
- the actual dosage of a composition of the invention administered to a patient can be determined according to the following physical and physiological factors: body weight, sex, severity of symptoms, type of disease being treated, prior or current therapeutic intervention, unknown etiology of the patient, time of administration, The excretion rate of the specific compound and the route of administration. In any event, the concentration of the active ingredient in the composition and the appropriate dosage for the subject will be determined by the medical personnel responsible for administration.
- Endothelial CXCR7 deleted mice were constructed using the tamoxifen-CreERT2 strategy. Briefly, as described previously (Nature cell biology. 2015; 17: 123-136) mice bearing loxP-site flanking CXCR7 (CXCR7 f/f ) and Cdh5-promoter driven by Ralf Adams CreERT2 (Cdh5(PAC)-CreERT2+) mice (Nature. 2010; 465: 483-486) were crossed.
- Ctl The resulting progeny CXCR7 f/f Cdh5-CreERT2+ male mice were crossed with CXCR7 f/f Cdh5-CreERT2-male to produce endothelial CXCR7 conditional knockout animals (CXCR7 f/f Cdh5-CreERT2+, cKO for short) and littermate control (CXCR7 f/f Cdh5-CreERT2-, referred to as Ctl).
- mice and littermates were injected intraperitoneally with tamoxifen (Alfa Aesar, Heysham, England) (37.5 mg/ml in sunflower seed oil) at a dose of 150 mg/ Kg weight per day for three consecutive days. The mice were then allowed to rest for three days before another three days of injection.
- C57BL/6 mice were purchased from the China Food and Drug Administration and used to evaluate the effect of adenovirus overexpressing CXCR7 in MI. All animal programs were approved by the National Cardiovascular Disease Center, Fuwai Hospital, Laboratory Animal Center, and the Institutional Animal Care and Use Committee.
- Femoral artery injury models were prepared as previously described (Circulation. 2011; 123:631-639). Briefly, mice were anesthetized by intraperitoneal injection of sodium pentobarbital (70 mg/kg). A one-sided inguinal incision exposes the femoral artery and separates the accompanying nerves and veins. The proximal and distal ends of the femoral artery were tied using a 6-0 suture thread to temporarily control blood flow. A small section of the artery between the rectus femoris and the medial femoral muscle was isolated, and the proximal end was restrained with a 6-0 suture thread, and the artery was laterally cut in the section.
- a flexible guide wire (diameter 0.35 mm, Cook Inc., IN, USA) was then inserted from the segment into the femoral artery and inserted 5 mm or more into the radial artery.
- the guidewire was left there for 3 minutes to exfoliate and dilate the artery.
- the guidewire is then removed and the suture thread that binds the proximal end of the artery is tightened.
- the suture thread used to interrupt the blood flow is relaxed to restore blood flow to the femoral artery.
- the skin incision was closed using a 5-0 suture thread.
- Femoral arteries were collected, paraffin-embedded, and transverse sections of 10-13 layers of injured arteries were continuously made from the distal end of the femoral artery segment at 150 ⁇ m intervals for morphological or histological analysis.
- Arterial sections on day 28 of injury were subjected to H&E staining to observe the severity of hyperplasia, and the most severely propagated sections were used for comparison. Images were acquired using a CCD camera mounted on an inverted microscope (DM6000B; Leica), and images were measured using Image-Pro Plus 6.0 software (Media Cybernetics). The area of the official cavity, the inner area of the inner elastic layer and the inner area of the outer elastic layer were collected and analyzed. To assess endothelial regeneration and leukocyte infiltration/migration, immunostaining analysis was performed on the middle of the artery on day 7 of injury using the corresponding antibodies.
- LAD left anterior descending coronary artery
- a recombinant adenovirus expressing mouse CXCR7 (Ad-CXCR7) was injected into the left ventricle of each mouse 1 minute prior to coronary artery ligation (using 418-1506 of mouse CXCR7 mRNA (NM_001271607.1))
- the nucleotide sequence of the position is used to construct a recombinant adenovirus or an empty vector at a dose of 1 x 10 9 plaque forming units per mouse.
- the success of the ligation was confirmed by left ventricular color change and electrocardiogram (ECG) ST-segment elevation.
- cardiac function and left ventricular structure were measured by echocardiography (VisualSonics VeVo 2100 Imaging System), and the measured indicators were left ventricular ejection fraction (EF), left ventricular partial area change (FAC), and left ventricular diameter shortening score. (FS), mitral valve diastolic early/late blood flow peak velocity ratio (E/A), left ventricular anterior wall thickness (LVAW), left ventricular posterior wall thickness (LVPW), and left ventricular volume, and left ventricular size were measured.
- E/A mitral valve diastolic early/late blood flow peak velocity ratio
- LVAW left ventricular anterior wall thickness
- LVPW left ventricular posterior wall thickness
- left ventricular volume and left ventricular size
- Cardiac infarction was assessed by Masson staining (Circulation. 2015; 132: 47-58). Briefly, a series of parasternal short-axis slices (thickness 5 ⁇ m) were obtained at 200 ⁇ m intervals. Representative midsection sections were stained using Masson's trichrome reagent (Leagene Biotec. Co, Ltd) and photographed using a Zeiss optical microscope (AXI0; Zeiss). Infarct size was measured and calculated using Image-Pro Plus 6.0 software (Media Cybernetics).
- Hind limb ischemia is caused by ligation of the left femoral artery, and the ligation site is bifurcated at the distal end of the saphenous artery (Arteriosclerosis, thrombosis, and vascular biology. 2014; 34: 408-418). Blood flow to the hind limbs was measured before and after ligation using a laser Doppler flow meter (LDF; PeriCam PSI). Mice in which the blood flow of hind limbs was reduced by not less than 50% after ligation were included in the experiment. For mice that successfully ligated the artery, blood flow was measured three times on days 4, 7, and 14, respectively. Blood flow was measured and analyzed blindly.
- LDF laser Doppler flow meter
- mice were sacrificed by administration of an excess of sedative.
- the gastrocnemius muscle was divided, fixed and paraffin embedded for analysis of vascular density. Briefly, the largest transverse section of each gastrocnemius muscle was stained, and three muscle gaps were randomly photographed in each section. The blood vessels in the photo are then counted and analyzed.
- MLEC Mouse lung endothelial cells
- mice were perfused through the right ventricle with sterile PBS to remove blood cells.
- the lung lobes were isolated, minced, and digested with collagenase (180-200 U/mL; Worthington) at 37 ° C (40 minutes).
- collagenase 180-200 U/mL; Worthington
- the cells were incubated with Dynabeads (Dynal Biotech) coated with anti-mouse CD31 (BD Biosciences).
- the cells on the beads were separated using a magnetic separator (Dynal) and then cultured for 3 days in a collagen I (Worthington) coated Petri dish/culture flask containing 20% fetal bovine serum (FBS), 1% AA ( GIBCO), and DMEM of 100 mg/L endothelial cell growth additive (ECGS; ScienCell).
- FBS fetal bovine serum
- GIBCO fetal bovine serum
- DMEM 100 mg/L endothelial cell growth additive
- the cells were dissociated and selected with Dynabeads coated with rat anti-mouse CD102 (ICAM-2; Pharmingen).
- IAM-2 rat anti-mouse CD102
- Mouse aortic endothelial cells were isolated as previously described (Cell metabolism. 2011; 13: 592-600). Briefly, the aorta was collected and the fat and connective tissue surrounding the adventitia were removed and cut into small pieces of 1-2 mm 2 . The aortic fragments were cultured in medium for 5-7 days to grow endothelial cells. The endothelial cells are then passaged and cultured. To isolate human aortic endothelial cells (HAEC), human aortic samples from an external hospital were collected in DMEM. The aortic intimal layer was exfoliated and treated in the same manner as MAEC culture.
- HAEC human aortic endothelial cells
- Myocardial fibroblasts were isolated as previously described (Journal of cellular and molecular medicine. 2014; 18: 2266-2274). Briefly, the ventricles of newborn Wistar rats were isolated, washed and minced in PBS. The tissue was then digested in PBS containing 0.06% collagenase (Worthington) at 37 °C. The collected cell suspension was centrifuged and resuspended in 10% DMEM. The suspension was placed in a culture flask and incubated for 90 minutes. Fibroblasts tend to adhere to the bottom. Remove non-adherent cells. Adherent cardiac fibroblasts are cultured and subsequently passaged with trypsin.
- siRNA gene silencing was performed in order to knock down CXCR7, CXCR4, ⁇ -arrestin1 or ⁇ -arrestin2 proteins in endothelial cells. Briefly, endothelial cells were seeded in 12-well plates. Before transfection, 40pmol siRNA mixed with 2.0 ⁇ L Hieff Trans TM Liposomal Transfection Reagent ( Yeasen, China) in 200 ⁇ L DMEM for 20 minutes. The medium was changed to DMEM. After 20 minutes, the siRNA transfection reagent mixture was added to the wells (200 ⁇ L/well). Transfection lasted for 6 hours. The transfection medium was then discarded and the cells were cultured in medium containing 20% FBS for not less than 6 hours prior to further analysis.
- the siRNA used in this study is as follows:
- si-CXCR4 (Arteriosclerosis, thrombosis, and vascular biology. 2014; 34: 1716-1722):
- Cell proliferation studies Cell growth was measured using Cell Counting Kit-8 (CCK-8; Yeasen, Shanghai, China). Briefly, cells were planted in a 9-well flat bottom plate. After the cells were completely attached to the bottom, the cells were starved for 6-8 h in medium containing 3% FBS but no ECGS. The medium was then replaced with a medium-CCK-8 mixture (10:1 by volume). After 4 hours, the absorbance at 450 nm was measured as a background. The cells were then incubated with the indicated reagents for 48 hours. Finally, the medium was replaced again with the medium-CCK-8 mixture. After 4 hours, the absorbance at 450 nm was measured to show cell growth.
- CCK-8 Cell Counting Kit-8
- reagent concentrations used in the cell studies were as follows: 10 ng/mL IL-1 ⁇ , 1 ⁇ M CCX771 or CCX704 (both compounds were supplied by ChemoCentryx, Inc., Mountain View, CA, USA).
- Tubule formation assay hypoxic-conditioned media for endothelial tubule formation assays. Briefly, cardiac fibroblasts in DMEM containing 3% FBS received hypoxia for 12 hours by AnaeroPack-Anaero (MGC, Japan). Conditioned medium was collected. For tubelet formation assays, Matrigel (Corning, NY, USA) with reduced growth factors was dispersed in 96-well plates (40 ⁇ L/well) using a cold pipette. Matrigel was then polymerized at 37 ° C for 1 hour. Endothelial cells were then trypsinized, resuspended in conditioned medium, and plated in plates at a concentration of 2 x 10 4 cells per well.
- IL-1 ⁇ 10 ng/mL
- TNF ⁇ 25 ng/mL
- FBS medium containing 20% FBS
- Microtubule formation was observed and photographed every 2 hours using an inverted phase contrast imaging microscope (Leica, Germany) with a 5x objective. Results from the 6th hour were used for analysis. Record and analyze at least 5 images from different cells.
- Calcium ion response assay Cells with negative siRNA or si-CXCR7 were plated in 96-well plates and grown overnight in medium containing 20% FBS and 10 ng/mL IL-1 ⁇ .
- For Calcium Fluorescence staining cell-like solution (AAT Bioquest, Sunnyvale, CA) with a Dye Cal-520 TM incubated for 2 hours. Fluorescence was monitored using excitation and emission at wavelengths of 490 and 525 nm, respectively, using a FlexStation 3 Microplate Reader (Molecular Devices, USA). After a 17 second baseline measurement, 100 ng/mL CXCL12 was added and the resulting calcium ion response was additionally measured for 78 seconds. The CXCR4 inhibitor AMD3100 (1 ⁇ g/mL) was added 2 hours before CXCL12 stimulation.
- Imaging flow cytometric assay Cell surface expression of CXCR4 and CXCR7 was analyzed in HUVEC using ImageStreamX Mark II Imaging Flow Cytometer (Merck, Darmstadt, Germany). Briefly, HUVECs transfected with si-RNA or si-CXCR7 in 12-well plates were treated with IL-1 ⁇ (10 ng/mL) for 6-8 hours. It was then trypsinized, centrifuged and resuspended in 100 ⁇ L of FACS buffer (HBSS containing 0.6 mg/mL bovine serum albumin and 0.3 mM EDTA). The antibody was added to the cell suspension and placed on ice for 30 minutes for staining.
- FACS buffer HBSS containing 0.6 mg/mL bovine serum albumin and 0.3 mM EDTA
- CXCR7 was stained with anti-CXCR7 mAb (11G8, 1:100) and Alexa Fluor-594-conjugated secondary antibody.
- CXCR4 was labeled with CXCR4 antibody (polyclonal antibody, 1:200, Sigma) and Alexa Fluor-488-conjugated secondary antibody. Cells were then analyzed and photographed using the ImageStreamX Mark II Imaging Flow Cytometer.
- RNA isolation cells were lysed in TRIzol (Invitrogen). The solution was then mixed with chloroform (5:1, by volume) and centrifuged (12,000 g; 15 min; 4 °C). The aqueous phase was collected, mixed with isopropanol and centrifuged (12,000 g; 10 min; 4 ° C). After washing with 75% alcohol, the resulting RNA can be used. use Select Master Mix (Invitrogen) for quantitative RT-PCR.
- the primers used in this study are as follows:
- CXCR4 forward CTCTACAGCAGCGTTCTC;
- eNOS forward GGCATCACCAGGAAGAAG;
- Sections (5 ⁇ m) from paraffin-embedded tissues were dewaxed, rehydrated, and subjected to antigen retrieval by boiling in EDTA antigen-repairing water (pH 9.0; ZSGB-BIO, Beijing, China) for 2 minutes.
- HUVEC was plated on coverslips (NUNC) and fixed in 4% paraformaldehyde for 20 minutes. The samples were then incubated with goat serum containing 0.3% Triton X-100 for blocking and membrane disruption. After incubation, the antibody was incubated overnight at 4 °C with the primary antibody and the samples were incubated with Alexa Fluor-594 coupled and/or Alexa Fluor-488 conjugated secondary antibody for 3 hours at room temperature.
- the coverslips were covered with VectaShield medium containing DAPI to stain the nuclei.
- Sections were imaged using a Zeiss inverted fluorescence microscope (AXI0; Zeiss) equipped with Zen software or a laser scanning confocal microscope (SP8; Leica) equipped with a 20x water immersion objective. Images were analyzed using Image-Pro Plus 6.0 software (Media Cybernetics, Inc. Rockville, MD, USA).
- the CXCL12 concentration was determined using a human CXCL12 ELISA kit (R&D Systems, Minnesota, USA).
- PDGF-BB concentrations were determined using a human PDGF-BB Quantikine ELISA kit (R&D Systems, Minnesota, USA).
- Plasma glucose (GLU), total cholesterol (CHOL, triglyceride (TG), free fatty acid (FFA), low density lipoprotein cholesterol (LDL-C) using an automated biochemical analyzer (AU5421, Beckman Coulter, California, USA) ), high density lipoprotein cholesterol (HDL-C) and alanine aminotransferase (ALT) were measured.
- MAEC was pre-starved for 6-8 h in medium containing 3% FBS and added to the indicated reagent temperature for 12 hours.
- the cells were then lysed in RIPA buffer containing protease inhibitors (Roche, Basel, Switherland), centrifuged (15800 g, 10 min), cell lysates were mixed with loading buffer and separated by 10% SDS-PAGE, then transferred to On the PVDF membrane. Hybridization of the membrane with the designated antibody. Some membranes re-hybridize with actin antibodies after decolorization.
- mice were intravenously injected with rose bengal (50 mg/kg).
- the left common carotid artery was exposed to a 2.5-mW green laser (540 nm; Melles Griot Inc).
- Blood flow was continuously detected by pulsed Doppler (Transonic, Sidney, Australia) from the onset of injury until a stable blockage (defined as no blood flow within 2 minutes) or 90 minutes without occlusion occurred.
- the occlusion time is defined as the time between the onset of vascular injury and the occurrence of stable occlusion. To calculate the average occlusion time, the obstruction time for animals that did not occlude was classified as 90 minutes.
- ACD anticoagulated whole blood (ACD: blood volume ratio 1:9) was mixed with the same volume of pre-warmed physiological saline, and collagen (2 ⁇ g/mL) was added to induce aggregation.
- the sample was equilibrated at 37 ° C for 7 minutes before the measurement. Platelet aggregation was then carried out in a Chronolog 710 aggregometer (Chronolog, Havertown, PA, USA) under constant agitation (1200 rpm) at 37 °C. The results were recorded and analyzed using Aggro/Link5 software (Chronolog, Havertown, PA, USA).
- platelets were activated in vivo and in vitro, respectively.
- the platelet activation method is the same as that used to measure platelet aggregation. Briefly, ACD anticoagulated whole blood was mixed with the same volume of pre-warmed physiological saline. The samples were equilibrated at 37 °C for 7 minutes and then collagen (2 ⁇ g/mL) or U46619 (2 mM; Sigma) was added to activate platelets. After 30 minutes, centrifugation (6000 g, 5 min, 4 ° C) and plasma were collected for detection of CXCL12. In vivo, anesthetized mice were intravenously injected with U46619 (20 ug/mouse). EDTA anticoagulated whole blood was collected within 10 minutes.
- Platelets were depleted by intravenous injection of CD41 antibody (20 ug/mouse; Ebioscience, USA). EDTA anticoagulated whole blood samples were collected before injection, 2 hours and 5 days after injection, respectively.
- the patients in this study were 95 patients who were diagnosed with acute myocardial infarction (AMI) and who underwent PCI immediately from August 2014 to January 2015.
- AMI acute myocardial infarction
- Example 1 Endothelial expression of CXCR7 in injured arteries of mice and humans
- CXCR7 The vascular expression of CXCR7 in mice was first examined.
- CXCR7 is expressed at low levels in healthy mouse dispersed endothelial cells (Fig. 1A). However, in damaged arteries, CXCR7 expression is upregulated and is mainly found in endothelial cells of the neointimal, colocalizing with the endothelial cell marker vWF (Fig. 1B).
- Fig. 1C the atherosclerotic plaque shoulder
- Fig. 1D microvessels in the plaque
- mice The baseline arterial expression of CXCR7 in cKO mice and littermates was similar before tamoxifen induction (Fig. 1E).
- mice After tamoxifen treatment, mice underwent endothelium exfoliation injury by angioplasty in the femoral artery. This injury induces vascular hyperplasia and mimics clinical restenosis after percutaneous coronary intervention. All mice were not subjected to genetic manipulation related to lipid metabolism and fed a normal diet.
- endothelial CXCR7 The mRNA expression of endothelial CXCR7 in endothelial cells isolated from cKO mice was essentially zero (Fig. 2A), which was further confirmed by immunostaining of damaged arteries (Fig. 2B). Deletion of CXCR7 did not alter the expression of CXCR4 and CXCL12 (Fig. 2G). Endothelial exfoliation damage leads to neointimal hyperplasia. Deletion of endothelial CXCR7 significantly increased the ratio of the neointimal zone and neointimal to medial membrane, but did not alter the medial thickness (Fig. 2C-F). Deletion of endothelial CXCR7 did not alter the body weight or plasma lipids of these normal blood lipid mice.
- Example 3 CXCR7 increases IL-1 ⁇ -treated endothelial cell proliferation and promotes endothelial regeneration after endothelial exfoliation injury
- IL-1 ⁇ stimulation up-regulated CXCR7 (Figs. 4A and 4B), CXCR4 and CXCL12 expression (Fig. 4I).
- IL-1 ⁇ promotes proliferation of CXCR7-functioning endothelial cells but does not promote proliferation of CXCR7-deficient cells (Fig. 4C and 4D).
- endothelial cells were treated with the CXCR7-specific antagonist CCX771 (which has an IC50 of about 5.3 nM, which does not affect the binding of CXCL12 to CXCR4) (Journal of immunology. 2009; 183:3204-3211) and the control compound CCX704.
- CCX771 inhibits proliferation of endothelial cells from the lung source (Fig. 4C) and aortic source (Fig. 4D). CCX771 treatment reduced ERK phosphorylation but did not affect JNK or p38 phosphorylation ( Figures 4E and 4F). This is consistent with the role of CXCR7 in promoting endothelial cell proliferation. This growth-promoting effect was not significant in the absence of IL-1 ⁇ treatment (Fig. 4J). In addition, in HUVEC, CCX771 or siRNA knockdown CXCR7 inhibited cell proliferation in the presence of IL-1 ⁇ (Figs. 4G and 4H). Similar results were observed when TNF ⁇ was used (Fig. 4K).
- CXCR7 can promote endothelial regeneration of damaged blood vessels, promote endothelial cell repair, and reduce blood vessel stenosis caused by injury.
- CXCR4 siRNA or AMD3100 (a CXCR4 antagonist with an IC50 of about 44 nM (Journal of immunology. 2009; 183:3204-3211) does not affect the binding of CXCL2 to CXCR7 (J Exp Med. 2006; 203: 2201-2213) Or with the weak binding of CXCR7, there was no significant difference in the proliferation of endothelial cells treated with Ki of about 34.5 ⁇ M (Molecular pharmacology. 2009; 75: 1240-1247) (C and D of Fig. 4L). It can be seen that inhibition of CXCR4 does not affect endothelial cell proliferation.
- Tubule formation was used to examine the role of CXCR7 in endothelial cell neovascularization responses.
- Blocking CXCR7 by siRNA significantly inhibited angiogenesis in HUVEC and HAEC as well as mouse EC ( Figures AA-D and A and B of Figure 4L).
- endothelial CXCR7 deletion significantly reduced blood flow recovery after femoral artery ligation, which was detected by laser Doppler imaging (Fig. 5E-G). Further histochemical staining of the endothelium showed a decrease in the number of blood vessels in the ischemic gastrocnemius muscle ( Figures 5H and 5I).
- CXCR7 or CXCR4 Knockdown of CXCR7 or CXCR4 by small RNA (si-RNA) transfection.
- Transfection control si-Neg
- si-CXCR7 si-CXCR7 1, si-CXCR7 2, and si-CXCR7 3
- si-CXCR4 si-CXCR4 1, si
- MAEC mouse arterial endothelial cells
- CXCR7 (A) and CXCR4 (C) protein expression was determined.
- a (CXCR4 immunoblot picture) and B show that CXCR7 knockdown has no effect on CXCR4 expression. *, p ⁇ 0.05 vs. si-Neg.
- endothelial cell CXCR7 plays a key role in promoting ischemia-induced angiogenesis, whereas ischemia-induced angiogenesis was previously thought to be mediated only by the interaction of CXCL12 with CXCR4 (Trends Immunol. 2007; 28:299 -307).
- Example 5 Deletion of endothelial CXCR7 impairs cardiac function after MI and increases mortality and infarct size
- endothelial CXCR7 deletion significantly shortened survival and reduced cumulative survival within 30 days after MI compared to control mice (Ctl) (Fig. 6A).
- cKO showed significant disruption of cardiac function and remodeling after MI, including decreased EF, E/A, and LVAWd, although there was no change in baseline cardiac function of cKO ( Figure 6B-F, Tables 2 and 3).
- n 12 in both groups.
- n 7Ctl, 6cKO.
- Example 6 Gene delivery of CXCR7 to infarcted hearts improves cardiac function and reduces infarct size after MI
- Ad-CXCR7 a recombinant adenovirus expressing CXCR7 expressing CXCR7
- Left ventricular cavity see Materials and General Methods for details.
- the results showed that Ad-CXCR7 delivery improved cardiac function after MI and reduced infarct size compared to the control vehicle ( Figure 8, Table 4).
- n 10Ad-Neg.11Ad-CXCR7.
- Example 7 Activation of CXCR7 promotes endothelial cell proliferation and reduces infarct size
- Mouse aortic endothelial cells were stimulated with IL-1 ⁇ and treated with CXCR7 selective agonist TC14012 (https://www.rndsystems.com/cn/products/tc-14012_4300) (Cayman Chemical, Michigan, USA) Hours were then measured for cell growth using Cell Counting Kit-8 (CCK-8; Yeasen, Shanghai, China). The results showed that TC14012 significantly promoted the growth of mouse aortic endothelial cells at 100 ng/mL compared to the control (0 ng/mL) (p ⁇ 0.05) (Fig. 9).
- CXCR7 selective agonist such as TC14012 to activate CXCR7 can reduce the range of myocardial infarction caused by ischemia, and has a protective effect on ischemic myocardium.
- the reason and activation of CXCR7 directly promote vascular endothelial cells in myocardial tissue of infarcted area. Proliferation is associated with angiogenesis, which facilitates myocardial remodeling and recovery of cardiac function after MI.
- Example 8 Deletion of endothelial CXCR7 and pharmacological blockade of CXCR7 raises the level of CXCL12 in the circulation
- CXCR7 conditional knockout mice were constructed and photochemically induced in mice as described in the literature (The Journal of clinical investigation. 2006; 116: 1391-1399). Thrombosis assessment. The time to complete occlusion of cKO mice was significantly shorter compared to control mice born in littermates (Ctl) (Fig. 11A & B). Deletion of CXCR7 by endothelial cells leads to an increase in the level of CXCL12 in the circulation (Fig. 11E), which is consistent with the function of the ligand scavenger of CXCR7 (PLoS One. 2010; 5: e9175; Blood.
- Example 9 Elevated levels of CXCL12 in the circulation promote thrombosis through the CXCL12-CXCR4 signaling pathway
- CXCL12 promotes platelet activation via its cognate receptor CXCR4.
- CXCL12 receptor for CXCL12
- a venous access was established in mice to directly infuse CXCL12 (infusion of CXCL12 or physiological saline in saline at 0.25 or 0.5 ng/min) In the bloodstream (Figure 12A).
- CXCL12 was confirmed to be successfully delivered by measuring the plasma concentration of CXCL12 (Fig. 12B).
- Moderate elevation of CXCL12 in the post-infusion cycle promoted ex vivo platelet aggregation in a concentration dependent manner (Figure 12C&D).
- the CXCL12 infusion also accelerated thrombus formation in vivo (Figure 12E&F).
- This prothrombotic effect of CXCL12 was abolished by pretreatment with the CXCR4-specific antagonist AMD3100 (Fig. 12E & F).
- AMD3100 treatment also abolished the prothrombotic effect caused by endothelial cell CXCR7 knockout (Fig. 13A & B).
- a physiologically elevated CXCL12 in the circulation causes a tendency to thrombosis.
- Example 10 Platelet promotes and responds to elevated CXCL12 following endothelial cell CXCR7 deletion
- Platelets contain CXCL12 and release CXCL12 upon activation (The Journal of experimental medicine. 2006; 203: 1221-1233), but it is unclear whether platelets in vivo are the source of CXCL12 and whether platelets contribute to the elevation of CXCL12 in cKO.
- Fig. 14I platelets were depleted by injection of anti-CD41 antibodies to mice, which resulted in substantial depletion of platelets 2 hours after injection and recovered on day 5 (Fig. 14I).
- Platelet depletion resulted in a decrease in circulating CXCL12 in both cKO and Ctl, but cKO mice maintained higher CXCL12 levels compared to both (Fig. 14G).
- CXCL12 in the circulation also includes CXCL12 from other sources than platelets, and circulating CXCL12 levels are regulated by endothelial cell CXCR7. It is worth noting that platelets contribute more to circulating CXCL12 in cKO than Ctl ( Figure 14H).
- CD62P p-selectin
- Example 11 Correlation between CXCL12 levels and platelet reactivity in human blood circulation
- the inventors further investigated the association of circulating CXCL12 with platelet reactivity in patients with acute myocardial infarction.
- Peripheral venous blood from consecutively enrolled patients was collected for measurement of plasma CXCL12 and platelet reactivity was measured in situ using the VerifyNow method (see Materials and General Methods section for details).
- CXCL12 levels were positively correlated with ADP-induced platelet reactivity (Fig. 15A), which supports a direct effect of CXCL12 in the circulation on platelet activation.
- CXCL12 is not associated with VerifyNow-AA (Fig. 15B), which may be related to the use of aspirin in these patients (Table 1), as TxB 2 release triggered by CXCL12 mediates platelet aggregation in vitro (Blood. 2000; 96:50- 57).
Landscapes
- Health & Medical Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- Chemical & Material Sciences (AREA)
- Engineering & Computer Science (AREA)
- Medicinal Chemistry (AREA)
- General Health & Medical Sciences (AREA)
- Molecular Biology (AREA)
- Animal Behavior & Ethology (AREA)
- Public Health (AREA)
- Veterinary Medicine (AREA)
- Biomedical Technology (AREA)
- Epidemiology (AREA)
- Hematology (AREA)
- Pharmacology & Pharmacy (AREA)
- Immunology (AREA)
- Urology & Nephrology (AREA)
- Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
- Heart & Thoracic Surgery (AREA)
- Bioinformatics & Cheminformatics (AREA)
- Physics & Mathematics (AREA)
- Organic Chemistry (AREA)
- General Chemical & Material Sciences (AREA)
- Chemical Kinetics & Catalysis (AREA)
- Cell Biology (AREA)
- Vascular Medicine (AREA)
- Food Science & Technology (AREA)
- Biotechnology (AREA)
- Analytical Chemistry (AREA)
- Biochemistry (AREA)
- General Physics & Mathematics (AREA)
- Pathology (AREA)
- Proteomics, Peptides & Aminoacids (AREA)
- Microbiology (AREA)
- Surgery (AREA)
- Diabetes (AREA)
- Cardiology (AREA)
- Bioinformatics & Computational Biology (AREA)
- Biophysics (AREA)
- Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
- Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
Abstract
The present invention provides a pharmaceutical composition and a method for treating a cardiovascular disease. Particularly, the present invention provides a CXCR7-targeting method and pharmaceutical composition for treating vascular endothelial injury in a subject or for improving cardiac remodeling in a subject after a myocardial infarction.
Description
相关申请Related application
本申请要求2017年1月26日提交的中国专利申请No.201710061692.4的优先权,在此通过援引将其全部内容并入本申请。The present application claims priority to Chinese Patent Application No. JP-A No. No. No. No. No. No. No. No.
本发明涉及用于治疗心血管疾病的药物组合物和方法。具体而言,本发明涉及靶向CXCR7的治疗对象的血管内皮损伤性疾病或改善对象的心肌梗死后的心脏重塑的方法和药物组合物。The present invention relates to pharmaceutical compositions and methods for treating cardiovascular diseases. In particular, the present invention relates to a method and a pharmaceutical composition for vascular endothelial injury disease in a subject to be treated with CXCR7 or to improve cardiac remodeling after myocardial infarction in a subject.
基因组范围的关联研究(GWA)发现,CXCL12基因座(其编码趋化因子CXCL12,又名基质细胞衍生因子-1,SDF1)与冠状动脉疾病(CAD)和心肌梗死(MI)具有关联性(Nat Genet.2009;41:334-341;Nat Genet.2013;45:25-33),其中某些风险等位基因与血浆CXCL12水平升高相关(European heart journal.2011;32:963-971)。较高的血浆CXCL12与慢性肾病患者的心肌梗死(MI)和死亡事件相关(European heart journal.2014;35:2115-2122),而Framingham Heart Study的一项前瞻性研究表,其也与心衰和总死亡率相关联(Arteriosclerosis,thrombosis,and vascular biology.2014;34:2100-2105)。A genome-wide association study (GWA) found that the CXCL12 locus, which encodes the chemokine CXCL12, also known as stromal cell-derived factor-1, SDF1, is associated with coronary artery disease (CAD) and myocardial infarction (MI) (Nat Genet. 2009; 41: 334-341; Nat Genet. 2013; 45: 25-33), in which certain risk alleles are associated with elevated plasma CXCL12 levels (European heart journal. 2011; 32: 963-971). Higher plasma CXCL12 is associated with myocardial infarction (MI) and death events in patients with chronic kidney disease (European heart journal. 2014; 35: 2115-2122), and a prospective study of the Framingham Heart Study, which is also associated with heart failure Associated with total mortality (Arteriosclerosis, thrombosis, and vascular biology. 2014; 34: 2100-2105).
CXCL12有两种受体:CXCR4,其是一种经典的G-蛋白偶联受体(GPCR),和CXCR7,其在2005年才被发现是CXCL12的第二种受体(The Journal of biological chemistry.2005;280:35760-35766)。CXCR4被认为参与血管重塑(Circulation.2003;108:2491-2497;Circulation research.2005;96:784-791;Arteriosclerosis,thrombosis,and vascular biology.2014;34:1209-1220;Thrombosis and haemostasis.2012;107:356-368)、动脉粥样硬化(Circulation research.2008;102:209-217)和心肌梗死(Circulation.2007;116:654-663;Circulation.2008;117:2224-2231;J Am Coll Cardiol.2011;58:2415-2423),但CXCR7在心血管疾病中的功能并不清楚。在有症状的冠心病患者中,CXCR4(而非CXCR7)表达降低与全因死亡和/或MI联合终点相关(Journal of thrombosis and haemostasis:JTH.2015;13:719-728)。CXCL12 has two receptors: CXCR4, a classical G-protein coupled receptor (GPCR), and CXCR7, which was discovered in 2005 as the second receptor for CXCL12 (The Journal of biological chemistry .2005;280:35760-35766). CXCR4 is thought to be involved in vascular remodeling (Circulation. 2003; 108: 2491-2497; Circulation research. 2005; 96: 784-791; Arteriosclerosis, thrombosis, and vascular biology. 2014; 34: 1209-1220; Thrombosis and haemostasis. 2012 ; 107: 356-368), atherosclerosis (Circulation research. 2008; 102: 209-217) and myocardial infarction (Circulation. 2007; 116: 654-663; Circulation. 2008; 117: 2224-2231; J Am Coll Cardiol.2011; 58:2415-2423), but the function of CXCR7 in cardiovascular disease is not clear. In patients with symptomatic coronary heart disease, a decrease in CXCR4 (rather than CXCR7) expression is associated with all-cause death and/or MI joint end points (Journal of thrombosis and haemostasis: JTH. 2015; 13: 719-728).
CXCR7在系统发生上与趋化因子受体密切相关,以比CXCR4更高的亲和力与 CXCL12结合,通过β-arrestin(而非经典的G-蛋白)传递信号,但不能与G蛋白偶联从而诱导典型的趋化因子受体-介导的细胞应答(J Exp Med.2006;203:2201-2213)。CXCR7之前被认为作为CXCL12的清除受体,介导有效的配体内吞和降解(PLoS One.2010;5:e9175;Cell.2008;132:463-473;Proc Natl Acad Sci USA.2010;107:628-632)。然而,诸多证据表明,CXCR7还具有配体清除之外的信号活性,包括在肿瘤细胞生长和器官再生中的信号活性(Proc Natl Acad Sci USA.2007;104:15735-15740;J Biol Chem.2008;283:4283-4294;Mol Cancer.2014;13:198;Nature medicine.2016;22:154-162)。CXCR7 is closely related to chemokine receptors in phylogeny, binds to CXCL12 with higher affinity than CXCR4, transmits signals through β-arrestin (rather than classical G-protein), but cannot be coupled with G protein to induce A typical chemokine receptor-mediated cellular response (J Exp Med. 2006; 203: 2201-2213). CXCR7 was previously thought to act as a scavenging receptor for CXCL12, mediating efficient endocytosis and degradation (PLoS One. 2010; 5: e9175; Cell. 2008; 132: 463-473; Proc Natl Acad Sci USA. 2010; 107: 628-632). However, there is ample evidence that CXCR7 also has signaling activity beyond ligand clearance, including signaling activity in tumor cell growth and organ regeneration (Proc Natl Acad Sci USA. 2007; 104: 15735-15740; J Biol Chem. 2008 ; 283: 4283-4294; Mol Cancer. 2014; 13: 198; Nature medicine. 2016; 22: 154-162).
在人中,CXCR7在脑、心脏、肾脏、内皮和肿瘤细胞中表达(J Exp Med.2006;203:2201-2213;PLoS One.2011;6:e20680)。其广泛表达于肿瘤血管内皮中(Proc Natl Acad Sci USA.2007;104:15735-15740),并由缺氧诱导(PLoS One.2013;8:e55290)。血小板既表达CXCR4也表达CXCR7(European heart journal.2014;35:386-394),但人或小鼠血液中的白细胞上不表达CXCR7蛋白(Journal of immunology.2010;185:5130-5139)。CXCR7缺失的小鼠出生前后便因心脏瓣膜异常死亡(Proc Natl Acad Sci USA.2007;104:14759-14764)。Weber等人使用高脂血症性Apoe
-/-小鼠证实,CXCR7的整体敲除加剧动脉粥样硬化,这缘于脂肪组织在胆固醇摄取方面的缺陷(Circulation.2014;129:1244-1253)。
In humans, CXCR7 is expressed in brain, heart, kidney, endothelial and tumor cells (J Exp Med. 2006; 203: 2201-2213; PLoS One. 2011; 6: e20680). It is widely expressed in tumor vascular endothelium (Proc Natl Acad Sci USA. 2007; 104: 15735-15740) and is induced by hypoxia (PLoS One. 2013; 8: e55290). Platelets express both CXCR4 and CXCR7 (European heart journal. 2014; 35: 386-394), but CXCR7 protein is not expressed on leukocytes in human or mouse blood (Journal of Immunology. 2010; 185: 5130-5139). CXCR7-deficient mice die from abnormal heart valves before and after birth (Proc Natl Acad Sci USA. 2007; 104: 14759-14764). Weber et al. used hyperlipidemia Apoe -/- mice to confirm that the overall knockout of CXCR7 exacerbates atherosclerosis due to defects in cholesterol uptake in adipose tissue (Circulation. 2014; 129: 1244-1253) .
发明内容Summary of the invention
本发明提供治疗或预防对象的心血管疾病的方法,包括给所述对象施用有效量的增加CXCR7蛋白的表达和/或活性的第一药物。优选地,本发明提供治疗对象的血管内皮损伤性疾病或改善对象的心肌梗死后的心脏重塑的方法,包括给所述对象施用有效量的增加CXCR7蛋白的表达和/或活性的第一药物。The invention provides a method of treating or preventing a cardiovascular disease in a subject comprising administering to the subject an effective amount of a first drug that increases the expression and/or activity of a CXCR7 protein. Preferably, the present invention provides a method of treating a vascular endothelial injury disease in a subject or ameliorating cardiac remodeling after myocardial infarction in a subject, comprising administering to the subject an effective amount of a first drug that increases expression and/or activity of a CXCR7 protein. .
在本发明的方法的一个实施方式中,本发明的治疗或预防对象的心血管疾病的方法还包括给所述对象施用有效量的降低CXCR4蛋白的表达和/或活性的第二药物。在另一个实施方式中,本发明的方法还包括给所述对象施用抑制血小板活化和/或聚集的药物。在另一个实施方式中,本发明的方法还包括给所述对象施用稳定斑块的药物。In one embodiment of the method of the present invention, the method of the present invention for treating or preventing cardiovascular disease in a subject further comprises administering to the subject an effective amount of a second drug that reduces expression and/or activity of the CXCR4 protein. In another embodiment, the methods of the invention further comprise administering to the subject a drug that inhibits platelet activation and/or aggregation. In another embodiment, the method of the invention further comprises administering to the subject a drug that stabilizes the plaque.
本发明还提供用于治疗或预防对象的心血管疾病的药物组合物,其包含有效量的用于增加CXCR7蛋白的表达和/或活性的第一药物。优选地,本发明提供用于治疗对象的血管内皮损伤性疾病或改善对象的心肌梗死后的心脏重塑的药物组合物,其包含有效量的用于增加CXCR7蛋白的表达和/或活性的第一药物。The invention also provides a pharmaceutical composition for treating or preventing a cardiovascular disease in a subject, comprising an effective amount of a first drug for increasing the expression and/or activity of a CXCR7 protein. Preferably, the present invention provides a pharmaceutical composition for treating a vascular endothelial injury disease in a subject or ameliorating cardiac remodeling after myocardial infarction in a subject, comprising an effective amount of an amount for increasing the expression and/or activity of the CXCR7 protein. a drug.
在本发明的药物组合物的一个实施方式中,所述药物组合物还包含有效量的CXCR4的选择性拮抗剂或抑制CXCR4蛋白表达的核酸分子或表达载体。在另一个实施方式中,所述药物组合物用于与有效量的包含CXCR4的选择性拮抗剂或抑制CXCR4蛋白表达的核酸分子或表达载体的第二药物联合使用。在另一个实施方式中,所述药物组合物用于与抑制血小板活化和/或聚集的药物联合使用。在另一个实施方式中,所述药物组合物用于与稳定斑块的药物联合使用。In one embodiment of the pharmaceutical composition of the invention, the pharmaceutical composition further comprises an effective amount of a selective antagonist of CXCR4 or a nucleic acid molecule or expression vector that inhibits expression of the CXCR4 protein. In another embodiment, the pharmaceutical composition is for use in combination with an effective amount of a selective antagonist comprising CXCR4 or a nucleic acid molecule or expression vector that inhibits expression of a CXCR4 protein. In another embodiment, the pharmaceutical composition is for use in combination with a drug that inhibits platelet activation and/or aggregation. In another embodiment, the pharmaceutical composition is for use in combination with a drug that stabilizes the plaque.
本发明还提供用于增加CXCR7蛋白的表达和/或活性的第一药物在制备用于治疗或预防对象的心血管疾病的药物组合物中的用途。优选地,本发明提供用于增加CXCR7蛋白的表达和/或活性的第一药物在制备用于治疗对象的血管内皮损伤性疾病或改善对象的心肌梗死后的心脏重塑的药物组合物中的用途。The present invention also provides the use of a first medicament for increasing the expression and/or activity of a CXCR7 protein for the preparation of a pharmaceutical composition for treating or preventing a cardiovascular disease in a subject. Preferably, the present invention provides a first drug for increasing the expression and/or activity of a CXCR7 protein in a pharmaceutical composition for preparing a vascular endothelial injury disease for treating a subject or improving cardiac remodeling after myocardial infarction in a subject use.
在本发明的用途的一个实施方式中,所述药物组合物还包含有效量的CXCR4的选择性拮抗剂或抑制CXCR4蛋白表达的核酸分子或表达载体。在另一个实施方式中,所述药物组合物与有效量的包含CXCR4的选择性拮抗剂或抑制CXCR4蛋白表达的核酸分子或表达载体的第二药物联合使用。在本发明的用途的另一个实施方式中,所述药物组合物用于与抑制血小板活化和/或聚集的药物联合使用。在另一个实施方式中,所述药物组合物用于与稳定斑块的药物联合使用。In one embodiment of the use of the invention, the pharmaceutical composition further comprises an effective amount of a selective antagonist of CXCR4 or a nucleic acid molecule or expression vector that inhibits expression of the CXCR4 protein. In another embodiment, the pharmaceutical composition is used in combination with an effective amount of a selective antagonist comprising CXCR4 or a nucleic acid molecule or expression vector that inhibits expression of a CXCR4 protein. In another embodiment of the use of the invention, the pharmaceutical composition is for use in combination with a medicament for inhibiting platelet activation and/or aggregation. In another embodiment, the pharmaceutical composition is for use in combination with a drug that stabilizes the plaque.
根据本发明的方法、药物组合物或用途,所述疾病选自以下一组:血栓形成、血栓栓塞、血管壁损伤、损伤后血管狭窄、PCI和Bypass术后血管再狭窄、冠心病、心肌缺血、心肌梗死、心梗后心力衰竭、心梗后心律失常及其任意组合。According to the method, pharmaceutical composition or use of the present invention, the disease is selected from the group consisting of thrombosis, thromboembolism, vascular wall injury, vascular stenosis after injury, vascular restenosis after PCI and Bypass, coronary heart disease, myocardial deficiency Blood, myocardial infarction, heart failure after myocardial infarction, arrhythmia after myocardial infarction, and any combination thereof.
本发明还提供血管支架或带有球囊的导管,其中所述支架或球囊的表面涂覆有有效量的用于增加CXCR7蛋白的表达和/或活性的第一药物。The invention also provides a vascular stent or catheter with a balloon, wherein the surface of the stent or balloon is coated with an effective amount of a first drug for increasing the expression and/or activity of the CXCR7 protein.
在本发明的血管支架或带有球囊的导管的一个实施方式中,所述支架或球囊的表面还涂覆有有效量的CXCR4的选择性拮抗剂或抑制CXCR4蛋白表达的核酸分子或表达载体。In one embodiment of the vascular stent or balloon with a balloon of the invention, the surface of the stent or balloon is further coated with an effective amount of a selective antagonist of CXCR4 or a nucleic acid molecule or expression that inhibits expression of the CXCR4 protein. Carrier.
本发明的血管支架或带有球囊的导管用于治疗或预防对象的血管损伤和/或心肌缺血相关性疾病,所述疾病选自以下一组:血栓形成、血栓栓塞、血管壁损伤、损伤后血管狭窄、PCI和Bypass术后血管再狭窄、冠心病、心肌缺血、心肌梗死、心梗后心力衰竭、心梗后心律失常及其任意组合。The vascular stent or the balloon-equipped catheter of the present invention is for treating or preventing vascular injury and/or myocardial ischemia-related disease in a subject selected from the group consisting of thrombosis, thromboembolism, blood vessel wall damage, Vascular stenosis after injury, vascular restenosis after PCI and Bypass, coronary heart disease, myocardial ischemia, myocardial infarction, heart failure after myocardial infarction, arrhythmia after myocardial infarction, and any combination thereof.
本发明还提供治疗或预防对象的血栓形成相关疾病的方法,包括给所述对象施用有效量的降低循环中的CXCL12的水平或活性的药物、或CXCR4的选择性拮抗剂、或抑制CXCR4蛋白表达的核酸分子或表达载体、或其组合。The present invention also provides a method of treating or preventing a thrombosis-related disease in a subject, comprising administering to the subject an effective amount of a drug that reduces the level or activity of circulating CXCL12, or a selective antagonist of CXCR4, or inhibiting CXCR4 protein expression. Nucleic acid molecule or expression vector, or a combination thereof.
本发明还提供用于治疗或预防对象的血栓形成相关疾病的药物组合物,其包含有效量的降低循环中的CXCL12的水平或活性的药物、或CXCR4的选择性拮抗剂、或抑制CXCR4蛋白表达的核酸分子或表达载体、或其组合。The present invention also provides a pharmaceutical composition for treating or preventing a thrombosis-related disease in a subject, which comprises an effective amount of a drug which lowers the level or activity of CXCL12 in circulation, or a selective antagonist of CXCR4, or inhibits CXCR4 protein expression. Nucleic acid molecule or expression vector, or a combination thereof.
本发明还提供降低循环中的CXCL12的水平或活性的药物、或CXCR4的选择性拮抗剂、或抑制CXCR4蛋白表达的核酸分子或表达载体、或其组合在制备用于治疗或预防对象的血栓形成相关疾病的药物组合物中的用途。The present invention also provides a medicament for reducing the level or activity of CXCL12 in circulation, or a selective antagonist of CXCR4, or a nucleic acid molecule or expression vector for inhibiting expression of CXCR4 protein, or a combination thereof for preparing thrombus for treating or preventing a subject Use in a pharmaceutical composition of a related disease.
本发明还提供用于治疗癌症的药物组合物,其包含CXCR7抑制剂,其中所述CXCR7抑制剂在施用至对象体内时不增加血液CXCL12水平。The invention also provides a pharmaceutical composition for treating cancer comprising a CXCR7 inhibitor, wherein the CXCR7 inhibitor does not increase blood CXCL12 levels when administered to a subject.
本发明还提供筛选具有高心血管安全性的用于治疗癌症的药物的方法,包括:The invention also provides a method of screening for a medicament for treating cancer with high cardiovascular safety, comprising:
(i)给动物施用候选药物,(i) administering a drug candidate to the animal,
(ii)测定来自所述动物的组织样品中的CXCR7蛋白的活性,和(ii) determining the activity of the CXCR7 protein in a tissue sample from the animal, and
(iii)测定来自所述动物的血液样品中的CXCL12蛋白的水平,(iii) determining the level of CXCL12 protein in a blood sample from the animal,
其中所述动物的组织样品中CXCR7蛋白的活性相对于未施用所述候选药物的对照动物的相同组织样品中CXCR7蛋白的活性的降低,且所述动物的血液样品中CXCL12蛋白的含量与未施用所述候选药物的对照动物的血液样品中CXCL12蛋白的水平相当或更低,提示所述候选药物是具有高心血管安全性的治疗癌症的药物。Wherein the activity of the CXCR7 protein in the tissue sample of the animal is decreased relative to the activity of the CXCR7 protein in the same tissue sample of the control animal to which the drug candidate is not administered, and the content of CXCL12 protein in the blood sample of the animal is not administered. The level of CXCL12 protein in the blood sample of the control animal of the drug candidate is comparable or lower, suggesting that the drug candidate is a drug for treating cancer with high cardiovascular safety.
本发明还提供筛选能够用于治疗或预防心血管疾病的药物的方法,包括:The invention also provides methods of screening for a medicament useful for treating or preventing a cardiovascular disease, comprising:
(i)给动物施用候选药物,和(i) administering a drug candidate to the animal, and
(ii)测定来自所述动物的组织样品中的CXCR7蛋白的表达水平和/或活性,(ii) determining the expression level and/or activity of the CXCR7 protein in a tissue sample from the animal,
其中所述动物的组织样品中CXCR7蛋白的表达水平和/或活性相对于未施用所述候选药物的对照动物的相同组织样品中CXCR7蛋白的表达水平和/或活性的升高提示所述候选药物能够治疗或预防心血管疾病。Wherein the expression level and/or activity of the CXCR7 protein in the tissue sample of the animal relative to the expression level and/or activity of the CXCR7 protein in the same tissue sample of the control animal to which the drug candidate is not administered is indicative of the candidate drug Can treat or prevent cardiovascular disease.
图1.人和小鼠损伤动脉的内皮细胞表达CXCR7Figure 1. Human and mouse injured arterial endothelial cells express CXCR7
A-D:在小鼠健康股动脉(A)和导丝损伤后28天的增生股动脉(B),以及在来自患有主动脉夹层的患者的主动脉切片(C,D)中,对CXCR7(红色)和vWF(内皮细胞标志物,绿色)进行免疫荧光染色,DAPI染核(蓝色)。最右侧是三种颜色的合并。C1中的灰色照片显示较低放大倍数的图1C中染色位置。箭头指向重度损伤。示出来自三个独立染色的代表性切片。L=内腔,P=斑块,Bar=50μm。E:来自Ctl和他莫西芬诱导前的cKO小鼠的股动脉中CXCR7的表达。来自Ctl和他莫西芬诱导前的cKO 小鼠的股动脉用CXCR7(红色)和vWF(内皮细胞标志,绿色)进行染色。DAPI染核(蓝色)。Bar=50μm。AD: Proliferative femoral artery (B) 28 days after injury in the healthy femoral artery (A) and guidewire in mice, and CXCR7 in aortic sections (C, D) from patients with aortic dissection Red) and vWF (endothelial cell marker, green) were immunofluorescently stained and DAPI stained with nuclei (blue). At the far right is the combination of the three colors. The gray photo in C1 shows the staining position in Figure 1C at a lower magnification. The arrow points to a severe injury. Representative sections from three independent stains are shown. L = lumen, P = plaque, Bar = 50 μm. E: Expression of CXCR7 in the femoral artery of cKO mice before Ctl and tamoxifen induction. Femoral arteries from cKO mice before Ctl and tamoxifen induction were stained with CXCR7 (red) and vWF (endothelial cell marker, green). DAPI stained with nuclei (blue). Bar = 50 μm.
图2.内皮CXCR7的可诱导缺失增加内皮剥脱损伤后的新生内膜形成Figure 2. Inducible deletion of endothelial CXCR7 increases neointimal formation after endothelial exfoliation injury
A-F:通过RT-PCR检测分离自内皮CXCR7条件性敲除小鼠(cKO)和同窝对照(Ctl)的小鼠肺内皮细胞(MLEC)中的CXCR7 mRNA表达(A)。在导丝剥脱损伤后的cKO和Ctl动脉中进行CXCR7(红色)和vWF(绿色)免疫荧光染色(B)。DAPI染核(蓝色)。在cKO中,新生内膜形成(C)和内膜与中膜比率(D)增加,而中膜厚度不变(E)。来自cKO和Ctl的受损及未受损动脉的代表性的HE染色(F)。Bar=100μm;*,p<0.05;**,p<0.01;n=12cKO,15Ctl。A-F: CXCR7 mRNA expression (A) in mouse lung endothelial cells (MLEC) isolated from endothelial CXCR7 conditional knockout mice (cKO) and littermate control (Ctl) was detected by RT-PCR. CXCR7 (red) and vWF (green) immunofluorescence staining (B) were performed in cKO and Ctl arteries after wire stripping injury. DAPI stained with nuclei (blue). In cKO, neointimal formation (C) and intima to media ratio (D) increased while median thickness remained unchanged (E). Representative HE staining (F) of damaged and undamaged arteries from cKO and Ctl. Bar = 100 μm; *, p < 0.05; **, p < 0.01; n = 12 cKO, 15 Ctl.
G:来自Ctl和他莫西芬诱导后的cKO小鼠内皮细胞中CXCR7的表达。从CXCR7内皮条件性敲除的小鼠(cKO)和同窝对照(Ctl)中分离小鼠肺内皮细胞(MLEC)。通过RT-PCR检测CXCR4(A)和CXCL12(B)mRNA表达。G: Expression of CXCR7 in endothelial cells of cKO mice after induction of Ctl and tamoxifen. Mouse lung endothelial cells (MLEC) were isolated from CXCR7 endothelial conditional knockout mice (cKO) and littermate controls (Ctl). CXCR4 (A) and CXCL12 (B) mRNA expression was detected by RT-PCR.
图3.CXCR7的内皮缺失破坏通过导丝损伤造成的剥脱后的再内皮化Figure 3. Endothelial Destruction of CXCR7 De-endothelialization after exfoliation by guidewire injury
A-F:使用导丝损伤后第7天的动脉,针对内皮细胞(A)(vWF,绿色)、内膜巨噬细胞(C)(F4/80,红色)和PDGF-BB表达(E)(绿色)进行免疫荧光染色(A,C,E)以及对应的定量(B,D,F)。箭头表示内皮化。Bar=100μm;*,p<0.05;n=10cKO,9Ctl。G:导丝损伤后第7天cKO中PDGF-BB的血浆水平升高。(n=15,p<0.05)AF: Arteries on day 7 after injury with guidewire, against endothelial cells (A) (vWF, green), intimal macrophages (C) (F4/80, red) and PDGF-BB (E) (green) ) Immunofluorescence staining (A, C, E) and corresponding quantification (B, D, F). Arrows indicate endothelialization. Bar = 100 μm; *, p < 0.05; n = 10 cKO, 9 Ctl. G: Plasma levels of PDGF-BB in cKO increased on day 7 after guidewire injury. (n=15, p<0.05)
图4.阻断CXCR7影响体外内皮细胞增殖Figure 4. Blocking CXCR7 affects endothelial cell proliferation in vitro
A-H:IL-1β(10ng/mL)处理增加培养的小鼠肺内皮细胞(MLEC)中CXCR7 mRNA(A)和蛋白(B)水平。IL-1β(10ng/mL)促进细胞生长。药物抑制(CCX771)或基因缺失CXCR7抑制MLEC(C)和小鼠主动脉内皮细胞(MAEC)(D)中的细胞增殖。在MAEC中,免疫印迹分析表明IL-1β增加ERK信号通路,该促生长信号通路被CXCR7抑制或缺失所抑制(E,F)。在HUVEC中,当用IL-1β刺激时,CCX771(G)或CXCR7(H)敲低降低细胞增殖。每个实验进行不少于3次。*,p<0.05;**,p<0.01。A-H: IL-1β (10 ng/mL) treatment increased CXCR7 mRNA (A) and protein (B) levels in cultured mouse lung endothelial cells (MLEC). IL-1β (10 ng/mL) promotes cell growth. Drug inhibition (CCX771) or gene deletion CXCR7 inhibits cell proliferation in MLEC (C) and mouse aortic endothelial cells (MAEC) (D). In MAEC, immunoblot analysis indicated that IL-1β increased the ERK signaling pathway, which is inhibited by CXCR7 inhibition or deletion (E, F). In HUVEC, CCX771 (G) or CXCR7 (H) knockdown reduces cell proliferation when stimulated with IL-1β. Each experiment was conducted no less than 3 times. *, p < 0.05; **, p < 0.01.
I:用IL-1β处理的培养小鼠肺内皮细胞(MLEC)中CXCR4和CXCL12的表达。在IL-1β(10ng/mL)刺激前和刺激后6小时,通过RT-PCR检测CXCR4(A)和CXCL12(B)mRNA。通过免疫印迹检测用IL-1β(10ng/mL)孵育0,6,12和24小时的细胞的CXCR4蛋白水平(C)。*,p<0.05;**,p<0.01。I: Expression of CXCR4 and CXCL12 in cultured mouse lung endothelial cells (MLEC) treated with IL-1β. CXCR4 (A) and CXCL12 (B) mRNA were detected by RT-PCR before and 6 hours after stimulation with IL-1β (10 ng/mL). CXCR4 protein levels (C) of cells incubated with IL-1β (10 ng/mL) for 0, 6, 12 and 24 hours were detected by immunoblotting. *, p < 0.05; **, p < 0.01.
J:药物抑制CXCR7对未受刺激的内皮细胞增殖的影响。在未受IL-1β刺激的小鼠肺内皮细胞(MLEC;A)和小鼠主动脉内皮细胞(MAEC;B)中进行增殖研究。无统计学显著性。J: Effect of drug inhibition of CXCR7 on proliferation of unstimulated endothelial cells. Proliferation studies were performed in mouse lung endothelial cells (MLEC; A) and mouse aortic endothelial cells (MAEC; B) that were not stimulated by IL-1β. No statistical significance.
K:CXCR7抑制降低TNFα诱导的内皮细胞增殖和血管生成。K: CXCR7 inhibition reduces TNFα-induced endothelial cell proliferation and angiogenesis.
TNFα诱导的CXCR7蛋白表达(A)和内皮细胞增殖(B)。CXCR7拮抗剂(B)或通过si-RNA敲低(C)降低诱导的细胞增殖和血管生成反应(D).Bar=200μm。TNFα-induced CXCR7 protein expression (A) and endothelial cell proliferation (B). CXCR7 antagonist (B) or reduced cell proliferation and angiogenic response (D) by silencing (C) by si-RNA. Bar = 200 μm.
L:CXCR7抑制对CXCR4信号的影响。L: CXCR7 inhibits the effect on CXCR4 signaling.
在培养的小鼠动脉内皮细胞(MAEC)中比较抑制CXCR7和/或CXCR4对小管形成(A,B;Bar=500μm)、增殖(C,D)、迁移(E,F;Bar=200μm)和钙释放(G)的影响。HUVEC中CXCR4的内皮表面表达不受CXCR7抑制的影响(H)。*,p<0.05vs.si-Neg;N.S.,nonsense vs.si-Neg。Inhibition of CXCR7 and/or CXCR4 on tubule formation (A, B; Bar = 500 μm), proliferation (C, D), migration (E, F; Bar = 200 μm) and culture in cultured mouse arterial endothelial cells (MAEC) The effect of calcium release (G). Endothelial surface expression of CXCR4 in HUVEC was not affected by CXCR7 inhibition (H). *, p<0.05 vs. si-Neg; N.S., nonsense vs. si-Neg.
图5.CXCR7的内皮缺失使后肢缺血的小鼠中血管生成和血流功能性恢复降低Figure 5. Endothelial loss of CXCR7 reduces angiogenesis and functional recovery of blood flow in mice with hind limb ischemia
A-I:在用或不用IL-1β预处理6小时并用si-CXCR7或阴性si-RNA(si-Neg)转染的HUVEC(A,B)或人主动脉内皮细胞(HAEC,C,D)中分析小管形成。Bar=500μm。以盲试的方式,在预定时间点监测后肢缺血小鼠的血流(E)。CXCR7的内皮缺失降低后肢血流恢复(F;n=8)。被监测的后肢区域在两组之间相同(G)。缺血后第21天,通过vWF的免疫染色检测腓肠肌的肌间隙(IS)中的血管密度(H)。vWF(绿色)染出内皮细胞,并用于血管密度定量。DAPI(蓝色)染核。对肌间隙中的平均血管数目进行定量(I;n=3).Bar=50μm in H.*,p<0.05;
p=0.053。
AI: HUVEC (A, B) or human aortic endothelial cells (HAEC, C, D) transfected with or without IL-1β for 6 hours and transfected with si-CXCR7 or negative si-RNA (si-Neg) Analyze tubule formation. Bar = 500 μm. Blood flow (E) of hindlimb ischemia mice was monitored at a predetermined time point in a blinded manner. Endothelial loss of CXCR7 reduced hindlimb blood flow recovery (F; n=8). The hind limb area being monitored was the same between the two groups (G). On day 21 after ischemia, vascular density (H) in the muscle space (IS) of the gastrocnemius muscle was measured by immunostaining of vWF. vWF (green) stains endothelial cells and is used for quantification of vascular density. DAPI (blue) dyed. The number of mean blood vessels in the muscle space was quantified (I; n = 3). Bar = 50 μm in H.*, p <0.05; p=0.053.
图6.小鼠内皮CXCR7缺失破坏心脏功能,降低存活率,增加MI后的梗死面积Figure 6. Mouse endothelial CXCR7 deletion disrupts cardiac function, reduces survival, and increases infarct size after MI
A-K:存活曲线(A;Kaplan-Meier方法)表明cKO小鼠在MI后30天内具有降低的存活时间和更高的累积死亡率(n=18cKO,20Ctl)。由超声专业人员在MI手术后7天通过盲试评价小鼠心脏功能。在B示出来自Ctl和cKO小鼠的代表性超声心动图。在cKO小鼠中,射血分数(EF;C),左心室切面面积变化分数(FAC;D),二尖瓣峰值早期到舒张末期充盈速度的比值(E/A;E),以及舒张期左室前壁厚度(LVAWd;F)均降低(两组中,n=12)。CXCR7在MI后的Ctl小鼠内皮细胞中表达,但在cKO内皮细胞中不表达(G)。对MI手术后第28天分离的心脏进行Masson染色表明,cKO组中梗死面积增加(H,J;n=8cKO,9Ctl)。对缺血区域中的内皮细胞免疫染色(vWF
+,绿色,箭头表示代表性的脉管系统)表明cKO中血管密度显著降低(I,K;n=3).*,p<0.05.G和I中,Bar=50μm。
AK: Survival curve (A; Kaplan-Meier method) showed that cKO mice had reduced survival time and higher cumulative mortality within 30 days after MI (n=18 cKO, 20 Ctl). Cardiac function in mice was assessed by ultrasound specialists by blind test 7 days after MI surgery. Representative echocardiograms from Ctl and cKO mice are shown at B. In cKO mice, ejection fraction (EF; C), left ventricular sectional area change score (FAC; D), ratio of early mitral peak to end-diastolic filling rate (E/A; E), and diastolic phase Left ventricular anterior wall thickness (LVAWd; F) was reduced (n=12 in both groups). CXCR7 is expressed in Ctl mouse endothelial cells after MI, but not in cKO endothelial cells (G). Masson staining of hearts isolated on day 28 after MI surgery showed an increase in infarct size in the cKO group (H, J; n = 8 cKO, 9 Ctl). Immunostaining of endothelial cells in the ischemic area (vWF + , green, arrows indicate representative vasculature) showed a significant decrease in vascular density in cKO (I, K; n = 3).*, p<0.05.G and In I, Bar = 50 μm.
图7.MI后小鼠中血浆CXCL12升高Figure 7. Elevated plasma CXCL12 in mice after MI
MI增加Ctl和cKO小鼠中的CXCL12血浆水平。与Ctl相比,cKO显示在MI前(定义为“0”)和MI后更高的血浆CXCL12水平。*,p<0.05vs.Ctl;
p<0.05vs.cKO中的0;
p<0.05vs.Ctl中的0。
MI increases CXCL12 plasma levels in Ctl and cKO mice. Compared to Ctl, cKO showed higher plasma CXCL12 levels before MI (defined as "0") and after MI. *, p<0.05vs.Ctl; 0 in p<0.05 vs. cKO; 0 in p<0.05 vs. Ctl.
图8.MI后左心室注入表达CXCR7的重组腺病毒改善心脏功能并降低梗死面积Figure 8. Recombinant adenovirus expressing CXCR7 in the left ventricle after MI improves cardiac function and reduces infarct size
A-F:在用表达CXCR7的腺病毒(Ad-CXCR7)转染的293T细胞系中确证CXCR7的表达(A)。从注射CXCR7阴性的腺病毒(Ad-Neg)和Ad-CXCR7的小鼠收集心脏,进行CXCR7染色(B,bar=50μm)。在注射Ad-CXCR7的小鼠(n=10Ad-Neg,11Ad-CXCR7)中,射血分数(EF;C)和舒张期左室前壁厚度(LVAWd;E)均得到改善。在注射Ad-CXCR7的小鼠中梗死面积降低(D,F;n=10Ad-Neg,11Ad-CXCR7).*,p<0.05。A-F: Expression of CXCR7 was confirmed in a 293T cell line transfected with adenovirus expressing CXCR7 (Ad-CXCR7) (A). Hearts were collected from mice injected with CXCR7-negative adenovirus (Ad-Neg) and Ad-CXCR7, and subjected to CXCR7 staining (B, bar = 50 μm). In mice injected with Ad-CXCR7 (n=10Ad-Neg, 11Ad-CXCR7), both the ejection fraction (EF; C) and the diastolic left ventricular anterior wall thickness (LVAWd; E) were improved. Infarct size was reduced in mice injected with Ad-CXCR7 (D, F; n = 10 Ad-Neg, 11 Ad-CXCR7).*, p < 0.05.
图9.TC14012促进血管内皮细胞增殖Figure 9. TC14012 promotes vascular endothelial cell proliferation
图10.TC14012减少心肌梗死的面积Figure 10. TC14012 reduces the area of myocardial infarction
图11.CXCR7的内皮限制性缺失(A,B和E)和药理拮抗剂(C,D和F)加快了大鼠形成完全的血栓闭塞的时间(A-D)并增加循环中的CXCL12(E&F)。Figure 11. Endothelium-deficient deletions of CXCR7 (A, B and E) and pharmacological antagonists (C, D and F) accelerate the time to complete thromboembolism in rats (AD) and increase CXCL12 (E&F) in circulation .
A-F:方法中详细描述了他莫西芬处理诱导的内皮CXCR7的条件性敲除(cKO),并用于测定血浆CXCL12和光化学诱导的血栓形成。通过皮下注射20mg/kg CCX771或载体处理野生型小鼠(C57BL/6),两小时后检查血浆CXCL12和光化学诱导的血栓形成。在诱导的血栓形成期间的代表性颈动脉血流量参见附图(A&C)。A-F: Conditional knockout (cKO) of endothelial CXCR7 induced by tamoxifen treatment is described in detail in the method and is used to determine plasma CXCL12 and photochemically induced thrombosis. Wild type mice (C57BL/6) were treated by subcutaneous injection of 20 mg/kg CCX771 or vehicle, and plasma CXCL12 and photochemically induced thrombosis were examined two hours later. Representative carotid blood flow during induced thrombosis is shown in the accompanying drawings (A&C).
图12.CXCL12静脉输注增加CXCL12并通过CXCR4增强光化学诱导的血栓形成Figure 12. Intravenous infusion of CXCL12 increases CXCL12 and enhances photochemically induced thrombosis by CXCR4
A-F:静脉内输注CXCL12增加循环CXCL12水平(A&B),增强胶原诱导的离体全血聚集(C&D)。CXCL12输注加速光化学诱导的血栓形成(E&F),且AMD3100(CXCR4特异性抑制剂)消除加速的血栓形成。代表性的全血聚集曲线显示在图C中,代表性的颈动脉血流示于图E中。如所示,输注速率为2μl/min,剂量为25ng/min/Kg。A-F: Intravenous infusion of CXCL12 increases circulating CXCL12 levels (A&B) and enhances collagen-induced ex vivo whole blood accumulation (C&D). CXCL12 infusion accelerates photochemically induced thrombosis (E&F) and AMD3100 (CXCR4-specific inhibitor) eliminates accelerated thrombosis. A representative whole blood aggregation curve is shown in panel C, and representative carotid blood flow is shown in panel E. As shown, the infusion rate was 2 μl/min and the dose was 25 ng/min/Kg.
图13.cKO中增强的血栓形成反应依赖于CXCR4Figure 13. Enhanced thrombotic response in cKO depends on CXCR4
A-B:AMD3100治疗消除了由内皮CXCR7的损失导致的促血栓形成反应。A显示在诱导的血栓形成期间的代表性颈动脉血流量。B表示每组中的闭塞时间的统计分析的结果。A-B: AMD3100 treatment abolishes the prothrombotic response caused by loss of endothelial CXCR7. A shows representative carotid blood flow during induced thrombosis. B represents the results of statistical analysis of occlusion time in each group.
图14.血小板明显有助于内皮CXCR7去除后循环CXCL12的增加Figure 14. Platelets significantly contribute to the increase in circulating CXCL12 after endothelial CXCR7 removal
A-H:当离体处理全血时,血小板激动剂U46619(A)或胶原(B)触发CXCL12释放。静脉内注射U46619(20μg/小鼠)在3分钟内减少血小板数目(图4C),并增加血浆CXCL12(图4D)。抗CD41抗体导致的血小板消耗会减少cKO和Ctl中的循环CXCL12,但cKO小鼠中升高的CXCL12水平得到维持(图4G)。在cKO中每百万血 小板减少中CXCL12的比例降低高于对照(H)。血小板中P-选择素(CD62P)表达的流式细胞术分析显示相比于Ctl,cKO中具有更高的CD62P表达和活化的血小板(E&F)。I:血小板耗竭和恢复时间。A-H: Platelet agonist U46619 (A) or collagen (B) triggers CXCL12 release when whole blood is treated ex vivo. Intravenous injection of U46619 (20 μg/mouse) reduced platelet counts in 3 minutes (Fig. 4C) and increased plasma CXCL12 (Fig. 4D). Platelet depletion by anti-CD41 antibodies reduced circulating CXCL12 in cKO and Ctl, but elevated CXCL12 levels were maintained in cKO mice (Fig. 4G). The proportion of CXCL12 decreased per million platelets in cKO was higher than that of control (H). Flow cytometric analysis of P-selectin (CD62P) expression in platelets showed higher CD62P expression and activated platelets (E&F) in cKO compared to Ctl. I: Platelet depletion and recovery time.
图15A-B:人血液循环中的CXCL12水平与血小板反应性的关联性。Figure 15A-B: Correlation of CXCL12 levels in human blood circulation with platelet reactivity.
发明详述Detailed description of the invention
本发明人令人惊奇地发现,CXCR7在维持内皮完整性中起到关键作用。具体而言,本发明人发现CXCR7在受损的血管中表达,其可被血管损伤中释放的炎症因子刺激诱导,并促进炎症相关内皮细胞的增殖;而内皮CXCR7的缺失则会促进由于内皮修复降低而造成的血管狭窄。本发明人还发现,内皮CXCR7在缺血诱导的血管生成中也起到关键作用。血管生成是内皮细胞依赖的过程,由此形成新的血管,这对于MI后用于挽救心肌细胞的血管再生以及缺血损伤后促进组织再生是必需的。更重要的是,发明人发现,将CXCR7基因通过腺病毒递送至心肌改善MI后的心脏功能并减少心肌梗死面积。因此,活化CXCR7将能够治疗血管内皮损伤性疾病或改善心肌梗死后的心脏重塑。The inventors have surprisingly found that CXCR7 plays a key role in maintaining endothelial integrity. Specifically, the present inventors have found that CXCR7 is expressed in damaged blood vessels, which can be induced by inflammatory factors released by vascular injury, and promotes proliferation of inflammation-associated endothelial cells; whereas deletion of endothelial CXCR7 promotes repair of endothelial cells Reduced blood vessels caused by stenosis. The inventors have also discovered that endothelial CXCR7 also plays a key role in ischemia-induced angiogenesis. Angiogenesis is a process of endothelial cell dependence, which results in the formation of new blood vessels, which is necessary for the regeneration of vascular regeneration after cardiomyocytes and the promotion of tissue regeneration after ischemia injury. More importantly, the inventors found that delivery of the CXCR7 gene to the myocardium by the adenovirus improves cardiac function after MI and reduces myocardial infarct size. Therefore, activation of CXCR7 will be able to treat vascular endothelial injury or improve cardiac remodeling after myocardial infarction.
相应地,本发明提供治疗或预防对象的心血管疾病的方法,包括给所述对象施用有效量的增加CXCR7蛋白的表达和/或活性的第一药物。优选地,本发明提供治疗对象的血管内皮损伤性疾病或改善对象的心肌梗死后的心脏重塑的方法,包括给所述对象施用有效量的增加CXCR7蛋白的表达和/或活性的第一药物。Accordingly, the invention provides a method of treating or preventing a cardiovascular disease in a subject comprising administering to the subject an effective amount of a first drug that increases the expression and/or activity of a CXCR7 protein. Preferably, the present invention provides a method of treating a vascular endothelial injury disease in a subject or ameliorating cardiac remodeling after myocardial infarction in a subject, comprising administering to the subject an effective amount of a first drug that increases expression and/or activity of a CXCR7 protein. .
如本文所用,术语“对象”是指哺乳动物,优选灵长类动物,更优选人。As used herein, the term "subject" refers to a mammal, preferably a primate, more preferably a human.
在本发明的方法的一个实施方式中,所述第一药物包含CXCR7的选择性激动剂、包含编码CXCR7蛋白或其功能片段的多核苷酸的表达载体、或其组合。在一个具体实施方式中,所述CXCR7的选择性激动剂选自CXCR7的活化型抗体、CXCR7的活化型配体、TC14012或其功能性类似物、及其组合。In one embodiment of the methods of the invention, the first medicament comprises a selective agonist of CXCR7, an expression vector comprising a polynucleotide encoding a CXCR7 protein or a functional fragment thereof, or a combination thereof. In a specific embodiment, the selective agonist of CXCR7 is selected from the group consisting of an activated antibody of CXCR7, an activated ligand of CXCR7, TC14012 or a functional analog thereof, and combinations thereof.
在本发明的方法的另一个实施方式中,本发明的治疗或预防对象的心血管疾病的方法还包括给所述对象施用有效量的降低CXCR4蛋白的表达和/或活性的第二药物。在一个实施方式中,所述第二药物包含CXCR4的选择性拮抗剂、抑制CXCR4蛋白表达的核酸分子或表达载体、或其组合。在一个具体实施方式中,所述CXCR4的选择性拮抗剂选自AMD3100或其功能类似物、CXCR4的阻断性抗体或其抗原结合片段、TC14012或其功能类似物、及其组合。在另一个具体实施方式中,所述抑制CXCR4蛋白表达的核酸分子是靶向CXCR4基因转录产物的siRNA或其前体。在另一个具体 实施方式中,所述抑制CXCR4蛋白表达的核酸分子是靶向CXCR4基因转录产物的反义RNA。In another embodiment of the methods of the present invention, the method of treating or preventing cardiovascular disease in a subject of the present invention further comprises administering to the subject an effective amount of a second drug that reduces expression and/or activity of the CXCR4 protein. In one embodiment, the second drug comprises a selective antagonist of CXCR4, a nucleic acid molecule or expression vector that inhibits expression of a CXCR4 protein, or a combination thereof. In a specific embodiment, the selective antagonist of CXCR4 is selected from the group consisting of AMD3100 or a functional analog thereof, a blocking antibody to CXCR4 or an antigen binding fragment thereof, TC14012 or a functional analog thereof, and combinations thereof. In another specific embodiment, the nucleic acid molecule that inhibits expression of a CXCR4 protein is an siRNA or a precursor thereof that targets a transcription product of the CXCR4 gene. In another specific embodiment, the nucleic acid molecule that inhibits expression of a CXCR4 protein is an antisense RNA that targets a transcription product of the CXCR4 gene.
在另一个实施方式中,本发明的方法还包括给所述对象施用抑制血小板活化和/或聚集的药物。在另一个实施方式中,本发明的方法还包括给所述对象施用稳定斑块的药物,例如他汀类药物。In another embodiment, the methods of the invention further comprise administering to the subject a drug that inhibits platelet activation and/or aggregation. In another embodiment, the methods of the invention further comprise administering to the subject a drug that stabilizes the plaque, such as a statin.
在本发明的治疗或预防对象的心血管疾病的方法中,通过口服、含服、吸入、静脉注射、动脉注射、肌肉注射、皮下注射、腹腔注射或局部施用的方式给对象施用所述第一和/或第二药物。在本发明的方法的一个具体实施方式中,通过冠脉内给药或将所述第一和/或第二药物涂覆在血管支架上或带有球囊的导管的球囊上而实现所述局部施用。In the method of treating or preventing cardiovascular disease in a subject of the present invention, the subject is administered to the subject by oral administration, buccal administration, inhalation, intravenous injection, intraarterial injection, intramuscular injection, subcutaneous injection, intraperitoneal injection or topical administration. And / or a second drug. In a specific embodiment of the method of the invention, the administration is achieved by intracoronary administration or by coating the first and/or second medicament on a balloon stent or a balloon of a catheter with a balloon. Topical application.
本发明还提供用于治疗或预防对象的心血管疾病的药物组合物,其包含有效量的用于增加CXCR7蛋白的表达和/或活性的第一药物。优选地,本发明提供用于治疗对象的血管内皮损伤性疾病或改善对象的心肌梗死后的心脏重塑的药物组合物,其包含有效量的用于增加CXCR7蛋白的表达和/或活性的第一药物。The invention also provides a pharmaceutical composition for treating or preventing a cardiovascular disease in a subject, comprising an effective amount of a first drug for increasing the expression and/or activity of a CXCR7 protein. Preferably, the present invention provides a pharmaceutical composition for treating a vascular endothelial injury disease in a subject or ameliorating cardiac remodeling after myocardial infarction in a subject, comprising an effective amount of an amount for increasing the expression and/or activity of the CXCR7 protein. a drug.
在本发明的药物组合物的一个实施方式中,所述第一药物包含CXCR7的选择性激动剂、包含编码CXCR7蛋白或其功能片段的多核苷酸的表达载体、或其组合。在一个具体实施方式中,所述CXCR7的选择性激动剂选自CXCR7的活化型抗体、CXCR7的活化型配体、TC14012或其功能类似物、及其组合。In one embodiment of the pharmaceutical composition of the present invention, the first drug comprises a selective agonist of CXCR7, an expression vector comprising a polynucleotide encoding a CXCR7 protein or a functional fragment thereof, or a combination thereof. In a specific embodiment, the selective agonist of CXCR7 is selected from the group consisting of an activated antibody of CXCR7, an activated ligand of CXCR7, TC14012 or a functional analog thereof, and combinations thereof.
在本发明的药物组合物的一个实施方式中,所述药物组合物还包含有效量的CXCR4的选择性拮抗剂或抑制CXCR4蛋白表达的核酸分子或表达载体。在另一个实施方式中,所述药物组合物用于与有效量的包含CXCR4的选择性拮抗剂或抑制CXCR4蛋白表达的核酸分子或表达载体的第二药物联合使用。在一个具体实施方式中,所述CXCR4的选择性拮抗剂选自AMD3100或其功能类似物、CXCR4的阻断性抗体或其抗原结合片段、TC14012或其功能类似物、及其组合。在另一个具体实施方式中,所述抑制CXCR4蛋白表达的核酸分子是靶向CXCR4基因转录产物的siRNA或其前体。在另一个具体实施方式中,所述抑制CXCR4蛋白表达的核酸分子是靶向CXCR4基因转录产物的反义RNA。In one embodiment of the pharmaceutical composition of the invention, the pharmaceutical composition further comprises an effective amount of a selective antagonist of CXCR4 or a nucleic acid molecule or expression vector that inhibits expression of the CXCR4 protein. In another embodiment, the pharmaceutical composition is for use in combination with an effective amount of a selective antagonist comprising CXCR4 or a nucleic acid molecule or expression vector that inhibits expression of a CXCR4 protein. In a specific embodiment, the selective antagonist of CXCR4 is selected from the group consisting of AMD3100 or a functional analog thereof, a blocking antibody to CXCR4 or an antigen binding fragment thereof, TC14012 or a functional analog thereof, and combinations thereof. In another specific embodiment, the nucleic acid molecule that inhibits expression of a CXCR4 protein is an siRNA or a precursor thereof that targets a transcription product of the CXCR4 gene. In another specific embodiment, the nucleic acid molecule that inhibits expression of a CXCR4 protein is an antisense RNA that targets a transcription product of the CXCR4 gene.
在另一个实施方式中,所述药物组合物用于与抑制血小板活化和/或聚集的药物联合使用。在另一个实施方式中,所述药物组合物用于与稳定斑块的药物联合使用,例如他汀类药物。In another embodiment, the pharmaceutical composition is for use in combination with a drug that inhibits platelet activation and/or aggregation. In another embodiment, the pharmaceutical composition is for use in combination with a drug that stabilizes the plaque, such as a statin.
根据本发明,所述药物组合物用于通过口服、含服、吸入、静脉注射、动脉注射、 肌肉注射、皮下注射、腹腔注射或局部施用的方式给对象施用。在一个具体的实施方式中,所述药物组合物用于冠脉内施用。According to the invention, the pharmaceutical composition is for administration to a subject by oral, buccal, inhalation, intravenous, intraarterial, intramuscular, subcutaneous, intraperitoneal or topical administration. In a specific embodiment, the pharmaceutical composition is for intracoronary administration.
本发明还提供用于增加CXCR7蛋白的表达和/或活性的第一药物在制备用于治疗或预防对象的心血管疾病的药物组合物中的用途。优选地,本发明提供用于增加CXCR7蛋白的表达和/或活性的第一药物在制备用于治疗对象的血管内皮损伤性疾病或改善对象的心肌梗死后的心脏重塑的药物组合物中的用途。The present invention also provides the use of a first medicament for increasing the expression and/or activity of a CXCR7 protein for the preparation of a pharmaceutical composition for treating or preventing a cardiovascular disease in a subject. Preferably, the present invention provides a first drug for increasing the expression and/or activity of a CXCR7 protein in a pharmaceutical composition for preparing a vascular endothelial injury disease for treating a subject or improving cardiac remodeling after myocardial infarction in a subject use.
在本发明的用途的一个实施方式中,所述第一药物包含有效量的CXCR7的选择性激动剂、包含编码CXCR7蛋白或其功能片段的多核苷酸的表达载体、或其组合。在一个具体实施方式中,所述CXCR7的选择性激动剂选自CXCR7的活化型抗体、CXCR7的活化型配体、TC14012或其功能类似物、及其组合。In one embodiment of the use of the invention, the first medicament comprises an effective amount of a selective agonist of CXCR7, an expression vector comprising a polynucleotide encoding a CXCR7 protein or a functional fragment thereof, or a combination thereof. In a specific embodiment, the selective agonist of CXCR7 is selected from the group consisting of an activated antibody of CXCR7, an activated ligand of CXCR7, TC14012 or a functional analog thereof, and combinations thereof.
在本发明的用途的一个实施方式中,所述药物组合物还包含有效量的CXCR4的选择性拮抗剂或抑制CXCR4蛋白表达的核酸分子或表达载体。在另一个实施方式中,所述药物组合物与有效量的包含CXCR4的选择性拮抗剂或抑制CXCR4蛋白表达的核酸分子或表达载体的第二药物联合使用。在一个具体实施方式中,所述CXCR4的选择性拮抗剂选自AMD3100或其功能类似物、CXCR4的阻断性抗体或其抗原结合片段、TC14012或其功能类似物、及其组合。在另一个具体实施方式中,所述抑制CXCR4蛋白表达的核酸分子是靶向CXCR4基因转录产物的siRNA或其前体。在另一个具体实施方式中,所述抑制CXCR4蛋白表达的核酸分子是靶向CXCR4基因转录产物的反义RNA。In one embodiment of the use of the invention, the pharmaceutical composition further comprises an effective amount of a selective antagonist of CXCR4 or a nucleic acid molecule or expression vector that inhibits expression of the CXCR4 protein. In another embodiment, the pharmaceutical composition is used in combination with an effective amount of a selective antagonist comprising CXCR4 or a nucleic acid molecule or expression vector that inhibits expression of a CXCR4 protein. In a specific embodiment, the selective antagonist of CXCR4 is selected from the group consisting of AMD3100 or a functional analog thereof, a blocking antibody to CXCR4 or an antigen binding fragment thereof, TC14012 or a functional analog thereof, and combinations thereof. In another specific embodiment, the nucleic acid molecule that inhibits expression of a CXCR4 protein is an siRNA or a precursor thereof that targets a transcription product of the CXCR4 gene. In another specific embodiment, the nucleic acid molecule that inhibits expression of a CXCR4 protein is an antisense RNA that targets a transcription product of the CXCR4 gene.
在本发明的用途的另一个实施方式中,所述药物组合物用于与抑制血小板活化和/或聚集的药物联合使用。在另一个实施方式中,所述药物组合物用于与稳定斑块的药物联合使用,例如他汀类药物。In another embodiment of the use of the invention, the pharmaceutical composition is for use in combination with a medicament for inhibiting platelet activation and/or aggregation. In another embodiment, the pharmaceutical composition is for use in combination with a drug that stabilizes the plaque, such as a statin.
根据本发明的用途,所述药物组合物用于通过口服、含服、吸入、静脉注射、动脉注射、肌肉注射、皮下注射、腹腔注射或局部施用的方式给对象施用。在一个具体的实施方式中,所述药物组合物被配制为用于冠脉内施用的形式。According to the use of the present invention, the pharmaceutical composition is for administration to a subject by oral, buccal, inhalation, intravenous, intraarterial, intramuscular, subcutaneous, intraperitoneal or topical administration. In a specific embodiment, the pharmaceutical composition is formulated in a form for intracoronary administration.
根据本发明的方法、药物组合物或用途,所述疾病选自以下一组:血栓形成、血栓栓塞、血管壁损伤、损伤后血管狭窄、PCI和Bypass术后血管再狭窄、冠心病、心肌缺血、心肌梗死、心梗后心力衰竭、心梗后心律失常及其任意组合。According to the method, pharmaceutical composition or use of the present invention, the disease is selected from the group consisting of thrombosis, thromboembolism, vascular wall injury, vascular stenosis after injury, vascular restenosis after PCI and Bypass, coronary heart disease, myocardial deficiency Blood, myocardial infarction, heart failure after myocardial infarction, arrhythmia after myocardial infarction, and any combination thereof.
本发明还提供血管支架或带有球囊的导管,其中所述支架或球囊的表面涂覆有有效量的用于增加CXCR7蛋白的表达和/或活性的第一药物。The invention also provides a vascular stent or catheter with a balloon, wherein the surface of the stent or balloon is coated with an effective amount of a first drug for increasing the expression and/or activity of the CXCR7 protein.
在本发明的血管支架或带有球囊的导管的一个实施方式中,所述第一药物包含 CXCR7的选择性激动剂、包含编码CXCR7蛋白或其功能片段的多核苷酸的表达载体、或其组合。在一个具体实施方式中,所述CXCR7的选择性激动剂选自CXCR7的活化型抗体、CXCR7的活化型配体、TC14012或其功能类似物、及其组合。In one embodiment of the vascular stent or balloon with a balloon of the present invention, the first drug comprises a selective agonist of CXCR7, an expression vector comprising a polynucleotide encoding a CXCR7 protein or a functional fragment thereof, or combination. In a specific embodiment, the selective agonist of CXCR7 is selected from the group consisting of an activated antibody of CXCR7, an activated ligand of CXCR7, TC14012 or a functional analog thereof, and combinations thereof.
在本发明的血管支架或带有球囊的导管的另一个实施方式中,所述支架或球囊的表面还涂覆有有效量的CXCR4的选择性拮抗剂或抑制CXCR4蛋白表达的核酸分子或表达载体。在一个具体实施方式中,所述CXCR4的选择性拮抗剂选自AMD3100或其功能类似物、CXCR4的阻断性抗体或其抗原结合片段、及其组合。在另一个具体实施方式中,所述抑制CXCR4蛋白表达的核酸分子是靶向CXCR4基因转录产物的siRNA或其前体。在另一个具体实施方式中,所述抑制CXCR4蛋白表达的核酸分子是靶向CXCR4基因转录产物的反义RNA。In another embodiment of the vascular stent or balloon with a balloon of the present invention, the surface of the stent or balloon is further coated with an effective amount of a selective antagonist of CXCR4 or a nucleic acid molecule that inhibits expression of the CXCR4 protein or Expression vector. In a specific embodiment, the selective antagonist of CXCR4 is selected from the group consisting of AMD3100 or a functional analog thereof, a blocking antibody to CXCR4 or an antigen binding fragment thereof, and combinations thereof. In another specific embodiment, the nucleic acid molecule that inhibits expression of a CXCR4 protein is an siRNA or a precursor thereof that targets a transcription product of the CXCR4 gene. In another specific embodiment, the nucleic acid molecule that inhibits expression of a CXCR4 protein is an antisense RNA that targets a transcription product of the CXCR4 gene.
本发明的血管支架或带有球囊的导管用于治疗或预防对象的血管损伤和/或心肌缺血相关性疾病,所述疾病选自以下一组:血栓形成、血栓栓塞、血管壁损伤、损伤后血管狭窄、PCI和Bypass术后血管再狭窄、冠心病、心肌缺血、心肌梗死、心梗后心力衰竭、心梗后心律失常及其任意组合。优选地,所述血管损伤相关性疾病是冠状动脉斑块和狭窄。优选地,所述心肌缺血相关性疾病是心肌梗死。The vascular stent or the balloon-equipped catheter of the present invention is for treating or preventing vascular injury and/or myocardial ischemia-related disease in a subject selected from the group consisting of thrombosis, thromboembolism, blood vessel wall damage, Vascular stenosis after injury, vascular restenosis after PCI and Bypass, coronary heart disease, myocardial ischemia, myocardial infarction, heart failure after myocardial infarction, arrhythmia after myocardial infarction, and any combination thereof. Preferably, the vascular injury-related disease is coronary plaque and stenosis. Preferably, the myocardial ischemia related disease is myocardial infarction.
另外,本发明人通过体内实验令人惊奇地发现,在具有病理生理学意义的浓度,CXCL12导致血栓形成倾向。本发明人还令人惊奇地发现,血小板活化的增敏效应是由循环中的CXCL12引起的,而非血管壁CXCL12。本发明人进一步发现,CXCL12的诱捕型受体内皮CXCR7对于维持CXCL12的生理学水平是必需的,而抑制CXCR7到导致CXCL12升高,后者作用于血小板CXCR4,从而促进血栓形成。In addition, the inventors have surprisingly found through in vivo experiments that CXCL12 causes a tendency to thrombosis at a concentration having pathophysiological significance. The inventors have also surprisingly found that the sensitizing effect of platelet activation is caused by circulating CXCL12, rather than the vessel wall CXCL12. The present inventors have further found that the trapped receptor endothelial CXCR7 of CXCL12 is essential for maintaining the physiological level of CXCL12, while inhibiting CXCR7 leads to an increase in CXCL12, which acts on platelet CXCR4, thereby promoting thrombus formation.
因此,本发明还提供治疗或预防对象的血栓形成相关疾病的方法,包括给所述对象施用有效量的降低循环中的CXCL12的水平或活性的药物、或CXCR4的选择性拮抗剂、或抑制CXCR4蛋白表达的核酸分子或表达载体、或其组合。Accordingly, the present invention also provides a method of treating or preventing a thrombosis-related disease in a subject, comprising administering to the subject an effective amount of a drug that reduces the level or activity of CXCL12 in circulation, or a selective antagonist of CXCR4, or inhibiting CXCR4 A nucleic acid molecule or expression vector, or a combination thereof, expressed by a protein.
CXCR7是CXCL12的清除受体,其介导有效的CXCL12内吞和降解。因此,在一个实施方式中,所述降低循环中的CXCL12的水平的药物是增加CXCR7蛋白的表达和/或活性的药物,例如包含编码CXCR7蛋白或其功能片段的多核苷酸的表达载体。CXCR7 is a scavenging receptor for CXCL12 that mediates efficient endocytosis and degradation of CXCL12. Thus, in one embodiment, the drug that reduces the level of circulating CXCL12 is a drug that increases the expression and/or activity of a CXCR7 protein, such as an expression vector comprising a polynucleotide encoding a CXCR7 protein or a functional fragment thereof.
在另一个实施方式中,所述降低循环中的CXCL12的水平或活性的药物是抗CXCL12抗体。In another embodiment, the drug that reduces the level or activity of CXCL12 in the circulation is an anti-CXCL12 antibody.
在另一个实施方式中,所述CXCR4的选择性拮抗剂选自AMD3100或其功能类似物、CXCR4的阻断性抗体或其抗原结合片段、及其组合。在另一个实施方式中, 所述抑制CXCR4蛋白表达的核酸分子是靶向CXCR4基因转录产物的siRNA或其前体。在另一个具体实施方式中,所述抑制CXCR4蛋白表达的核酸分子是靶向CXCR4基因转录产物的反义RNA。In another embodiment, the selective antagonist of CXCR4 is selected from the group consisting of AMD3100 or a functional analog thereof, a blocking antibody to CXCR4 or an antigen binding fragment thereof, and combinations thereof. In another embodiment, the nucleic acid molecule that inhibits expression of a CXCR4 protein is an siRNA or a precursor thereof that targets a transcription product of the CXCR4 gene. In another specific embodiment, the nucleic acid molecule that inhibits expression of a CXCR4 protein is an antisense RNA that targets a transcription product of the CXCR4 gene.
本发明还提供用于治疗或预防对象的血栓形成相关疾病的药物组合物,其包含有效量的降低循环中的CXCL12的水平或活性的药物、或CXCR4的选择性拮抗剂、或抑制CXCR4蛋白表达的核酸分子或表达载体、或其组合。在一个实施方式中,所述降低循环中的CXCL12的水平的药物是增加CXCR7蛋白的表达和/或活性的药物,例如包含编码CXCR7蛋白或其功能片段的多核苷酸的表达载体。在另一个实施方式中,所述降低循环中的CXCL12的水平或活性的药物是抗CXCL12抗体。在另一个实施方式中,所述CXCR4的选择性拮抗剂选自AMD3100或其功能类似物、CXCR4的阻断性抗体或其抗原结合片段、及其组合。在另一个实施方式中,所述抑制CXCR4蛋白表达的核酸分子是靶向CXCR4基因转录产物的siRNA或其前体。在另一个具体实施方式中,所述抑制CXCR4蛋白表达的核酸分子是靶向CXCR4基因转录产物的反义RNA。The present invention also provides a pharmaceutical composition for treating or preventing a thrombosis-related disease in a subject, which comprises an effective amount of a drug which lowers the level or activity of CXCL12 in circulation, or a selective antagonist of CXCR4, or inhibits CXCR4 protein expression. Nucleic acid molecule or expression vector, or a combination thereof. In one embodiment, the drug that reduces the level of circulating CXCL12 is a drug that increases the expression and/or activity of a CXCR7 protein, such as an expression vector comprising a polynucleotide encoding a CXCR7 protein or a functional fragment thereof. In another embodiment, the drug that reduces the level or activity of CXCL12 in the circulation is an anti-CXCL12 antibody. In another embodiment, the selective antagonist of CXCR4 is selected from the group consisting of AMD3100 or a functional analog thereof, a blocking antibody to CXCR4 or an antigen binding fragment thereof, and combinations thereof. In another embodiment, the nucleic acid molecule that inhibits expression of a CXCR4 protein is an siRNA or a precursor thereof that targets a transcription product of the CXCR4 gene. In another specific embodiment, the nucleic acid molecule that inhibits expression of a CXCR4 protein is an antisense RNA that targets a transcription product of the CXCR4 gene.
本发明还提供降低循环中的CXCL12的水平或活性的药物、或CXCR4的选择性拮抗剂、或抑制CXCR4蛋白表达的核酸分子或表达载体、或其组合在制备用于治疗或预防对象的血栓形成相关疾病的药物组合物中的用途。在一个实施方式中,所述降低循环中的CXCL12的水平的药物是增加CXCR7蛋白的表达和/或活性的药物,例如包含编码CXCR7蛋白或其功能片段的多核苷酸的表达载体。在另一个实施方式中,所述降低循环中的CXCL12的水平或活性的药物是抗CXCL12抗体。在另一个实施方式中,所述CXCR4的选择性拮抗剂选自AMD3100或其功能类似物、CXCR4的阻断性抗体或其抗原结合片段、及其组合。在另一个实施方式中,所述抑制CXCR4蛋白表达的核酸分子是靶向CXCR4基因转录产物的siRNA或其前体。在另一个具体实施方式中,所述抑制CXCR4蛋白表达的核酸分子是靶向CXCR4基因转录产物的反义RNA。The present invention also provides a medicament for reducing the level or activity of CXCL12 in circulation, or a selective antagonist of CXCR4, or a nucleic acid molecule or expression vector for inhibiting expression of CXCR4 protein, or a combination thereof for preparing thrombus for treating or preventing a subject Use in a pharmaceutical composition of a related disease. In one embodiment, the drug that reduces the level of circulating CXCL12 is a drug that increases the expression and/or activity of a CXCR7 protein, such as an expression vector comprising a polynucleotide encoding a CXCR7 protein or a functional fragment thereof. In another embodiment, the drug that reduces the level or activity of CXCL12 in the circulation is an anti-CXCL12 antibody. In another embodiment, the selective antagonist of CXCR4 is selected from the group consisting of AMD3100 or a functional analog thereof, a blocking antibody to CXCR4 or an antigen binding fragment thereof, and combinations thereof. In another embodiment, the nucleic acid molecule that inhibits expression of a CXCR4 protein is an siRNA or a precursor thereof that targets a transcription product of the CXCR4 gene. In another specific embodiment, the nucleic acid molecule that inhibits expression of a CXCR4 protein is an antisense RNA that targets a transcription product of the CXCR4 gene.
此外,最近CXCR7抑制剂已被提议用于癌症等疾病的治疗。然而,如上所述,本发明人令人惊奇地发现,使用例如CCX771的抑制剂来抑制CXCR7活性会通过增加血液中CXCL12水平而增加心血管血栓风险。因此,在任何使用CXCR7抑制剂的治疗例如癌症治疗中,优选使用不增加CXCL12水平的CXCR7抑制剂。In addition, recent CXCR7 inhibitors have been proposed for the treatment of diseases such as cancer. However, as described above, the inventors have surprisingly found that inhibition of CXCR7 activity using an inhibitor such as CCX771 increases the risk of cardiovascular thrombosis by increasing the level of CXCL12 in the blood. Therefore, in any treatment using a CXCR7 inhibitor such as cancer treatment, it is preferred to use a CXCR7 inhibitor that does not increase the level of CXCL12.
本发明还提供用于治疗癌症的药物组合物,其包含CXCR7抑制剂,其中所述CXCR7抑制剂在施用至对象体内时不增加血液CXCL12水平。在一些实施方案中, 所述癌症包括但不限于肝癌、乳腺癌等。The invention also provides a pharmaceutical composition for treating cancer comprising a CXCR7 inhibitor, wherein the CXCR7 inhibitor does not increase blood CXCL12 levels when administered to a subject. In some embodiments, the cancer includes, but is not limited to, liver cancer, breast cancer, and the like.
本发明还提供筛选具有高心血管安全性的用于治疗癌症的药物的方法,包括:The invention also provides a method of screening for a medicament for treating cancer with high cardiovascular safety, comprising:
(i)给动物施用候选药物,(i) administering a drug candidate to the animal,
(ii)测定来自所述动物的组织样品中的CXCR7蛋白的活性,和(ii) determining the activity of the CXCR7 protein in a tissue sample from the animal, and
(iii)测定来自所述动物的血液样品中的CXCL12蛋白的水平,(iii) determining the level of CXCL12 protein in a blood sample from the animal,
其中所述动物的组织样品中CXCR7蛋白的活性相对于未施用所述候选药物的对照动物的相同组织样品中CXCR7蛋白的活性的降低,且所述动物的血液样品中CXCL12蛋白的含量与未施用所述候选药物的对照动物的血液样品中CXCL12蛋白的水平相当或更低,提示所述候选药物是具有高心血管安全性的治疗癌症的药物。Wherein the activity of the CXCR7 protein in the tissue sample of the animal is decreased relative to the activity of the CXCR7 protein in the same tissue sample of the control animal to which the drug candidate is not administered, and the content of CXCL12 protein in the blood sample of the animal is not administered. The level of CXCL12 protein in the blood sample of the control animal of the drug candidate is comparable or lower, suggesting that the drug candidate is a drug for treating cancer with high cardiovascular safety.
本发明还提供筛选能够用于治疗或预防心血管疾病的药物的方法,包括:The invention also provides methods of screening for a medicament useful for treating or preventing a cardiovascular disease, comprising:
(i)给动物施用候选药物,和(i) administering a drug candidate to the animal, and
(ii)测定来自所述动物的组织样品中的CXCR7蛋白的表达水平和/或活性,(ii) determining the expression level and/or activity of the CXCR7 protein in a tissue sample from the animal,
其中所述动物的组织样品中CXCR7蛋白的表达水平和/或活性相对于未施用所述候选药物的对照动物的相同组织样品中CXCR7蛋白的表达水平和/或活性的升高提示所述候选药物能够治疗或预防心血管疾病。Wherein the expression level and/or activity of the CXCR7 protein in the tissue sample of the animal relative to the expression level and/or activity of the CXCR7 protein in the same tissue sample of the control animal to which the drug candidate is not administered is indicative of the candidate drug Can treat or prevent cardiovascular disease.
在本发明的用于筛选能够用于治疗或预防心血管疾病的药物的方法的一个实施方式中,所述CXCR7蛋白的活性选自促进血管内皮增殖、促进血管新生、促进损伤血管修复、减小心肌梗死面积、改善梗死后心肌重塑、改善梗死后的心脏功能。In one embodiment of the method of the present invention for screening a drug capable of treating or preventing a cardiovascular disease, the activity of the CXCR7 protein is selected from the group consisting of promoting vascular endothelial proliferation, promoting angiogenesis, promoting damage to blood vessel repair, and reducing Myocardial infarct size, improved myocardial remodeling after infarction, and improved cardiac function after infarction.
如本领域技术人员所了解,本发明上述的药物或药物组合物还可以包含药学可接受的载体。短语“药学可接受的”是指当按照需要施用于动物(例如人)时不会产生不良的、变应性的或其它不利的反应的分子实体和组合物。合适的药物组合物的制备是本领域技术人员根据本公开内容已知的,并示例于“Remington:The Science and Practice of Pharmacy,”第21版,2005,其通过参考并入本文中。另外,对于人类施用来说,应当理解,制备还应满足药物审批机构所要求的对无菌性、热原性、整体安全性以及纯度的标准。As will be appreciated by those skilled in the art, the above pharmaceutical or pharmaceutical compositions of the present invention may further comprise a pharmaceutically acceptable carrier. The phrase "pharmaceutically acceptable" refers to molecular entities and compositions that do not produce an undesirable, allergic or other untoward reaction when administered to an animal, such as a human, as desired. The preparation of suitable pharmaceutical compositions is known to those skilled in the art in light of the present disclosure and is exemplified in "Remington: The Science and Practice of Pharmacy," 21st Edition, 2005, which is incorporated herein by reference. In addition, for human administration, it should be understood that the preparation should also meet the criteria for sterility, pyrogenicity, overall safety, and purity required by the drug approval authority.
本文使用的“药学可接受的载体”包括任何和所有的溶剂、分散介质、抗氧化剂、盐、包衣、表面活性剂、防腐剂(例如对羟基苯甲酸甲酯或丙酯、山梨酸、抗菌剂、抗真菌剂)、等渗剂、溶液阻滞剂(例如石蜡)、吸附剂(例如,高岭土、膨润土)、药物稳定剂(例如,十二烷基硫酸钠)、凝胶、粘合剂(例如,糖浆、阿拉伯胶、明胶、山梨醇、黄芪胶、聚乙烯吡咯烷酮、羧甲基纤维素、藻酸盐)、赋形剂(例如,乳糖、聚乙二醇)、崩解剂(例如琼脂、淀粉、乳糖、磷酸钙、碳酸钙、海藻酸、山梨醇、甘氨酸)、 润湿剂(例如,十六烷醇、单硬脂酸甘油酯)、润滑剂、吸收促进剂(例如,季铵盐)、可食用油(例如,杏仁油、椰油、油性酯或丙二醇)、甜味剂、调味剂、着色剂、填充剂(例如,淀粉、乳糖、蔗糖、葡萄糖、甘露醇、硅酸)、压片润滑剂(例如,硬脂酸镁、淀粉、葡萄糖、乳糖、白垩)、吸入用载体(例如,烃抛射剂)、缓冲剂或诸如此类的物质及其组合(参见例如,“Remington:The Science and Practice of Pharmacy,”第21版,2005)。将任何除了与所述活性成分不相容的常规载体以外的常规载体用于所述治疗性或药物组合物也在考虑范围内。As used herein, "pharmaceutically acceptable carrier" includes any and all solvents, dispersion media, antioxidants, salts, coatings, surfactants, preservatives (eg, methyl or propyl paraben, sorbic acid, antibacterial). Agent, antifungal agent), isotonic agent, solution blocker (such as paraffin), adsorbent (for example, kaolin, bentonite), drug stabilizer (for example, sodium lauryl sulfate), gel, adhesive (eg, syrup, gum arabic, gelatin, sorbitol, tragacanth, polyvinylpyrrolidone, carboxymethylcellulose, alginate), excipients (eg, lactose, polyethylene glycol), disintegrants (eg Agar, starch, lactose, calcium phosphate, calcium carbonate, alginic acid, sorbitol, glycine), wetting agents (eg, cetyl alcohol, glyceryl monostearate), lubricants, absorption enhancers (eg, quarters) Ammonium salt), edible oil (eg, almond oil, coconut oil, oily ester or propylene glycol), sweeteners, flavoring agents, coloring agents, fillers (eg, starch, lactose, sucrose, glucose, mannitol, silicic acid) ), tableting lubricant (for example, magnesium stearate, lake , Glucose, lactose, chalk), the suction carrier (e.g., a hydrocarbon propellant), buffers or the like materials and combinations thereof (see, e.g., "Remington: The Science and Practice of Pharmacy," 21st Edition, 2005). It is also contemplated that conventional carriers other than conventional carriers that are incompatible with the active ingredients are employed in the therapeutic or pharmaceutical compositions.
在任何情形中,所述组合物可包含多种抗氧化剂以阻滞一种或多种组分的氧化。抗氧化剂的实例包括抗坏血酸、盐酸半胱氨酸、亚硫酸钠、亚硫酸氢钠、焦亚硫酸钠、抗坏血酸棕榈酸酯、丁羟甲苯、丁羟茴醚、卵磷脂、没食子酸丙酯和生育酚。另外,防止微生物作用可通过使用防腐剂来实现,所述防腐剂例如多种抗菌剂和抗真菌剂,其包括但不限于对羟基苯甲酸酯类(例如,对羟基苯甲酸甲酯、对羟基苯甲酸丙酯)、氯丁醇、苯酚、山梨酸、硫柳汞或其组合。In any event, the composition can comprise a plurality of antioxidants to retard oxidation of one or more components. Examples of the antioxidant include ascorbic acid, cysteine hydrochloride, sodium sulfite, sodium hydrogensulfite, sodium metabisulfite, ascorbyl palmitate, butylated hydroxytoluene, butylated hydroxyanisole, lecithin, propyl gallate, and tocopherol. In addition, the prevention of the action of microorganisms can be achieved by the use of preservatives such as various antibacterial and antifungal agents including, but not limited to, parabens (for example, methylparaben, p-hydroxyl Propyl benzoate), chlorobutanol, phenol, sorbic acid, thimerosal or a combination thereof.
在所述组合物为液体形式的一些实施方案中,载体可以是溶剂或分散介质,其包括但不限于水、乙醇、多元醇(例如,甘油、丙二醇、液体聚乙二醇等)、液体(例如,甘油三酯、植物油、脂质体)及其组合。可维持适当的流动性,例如通过使用包衣(如卵磷脂)来实现;通过分散于载体(例如,液体多元醇或脂类)中而维持所需粒径来实现;通过使用表面活性剂(例如,羟丙基纤维素)来实现;或者这些方法的组合。在许多情形中,优选包含等渗剂(例如,糖、氯化钠或其组合)。In some embodiments where the composition is in liquid form, the carrier can be a solvent or dispersion medium including, but not limited to, water, ethanol, polyol (eg, glycerol, propylene glycol, liquid polyethylene glycol, etc.), liquid ( For example, triglycerides, vegetable oils, liposomes, and combinations thereof. Proper fluidity can be maintained, for example, by the use of a coating such as lecithin; by maintaining the desired particle size by dispersion in a carrier such as a liquid polyol or a lipid; by using a surfactant ( For example, hydroxypropyl cellulose); or a combination of these methods. In many cases, it will be preferred to include isotonic agents (for example, sugars, sodium chloride or combinations thereof).
在一些具体的实施方案中,可通过在所述组合物中使用延迟吸收的药剂(例如,单硬脂酸铝、明胶或其组合)来实现注射用组合物的延长吸收。In some specific embodiments, prolonged absorption of the injectable compositions can be brought about by the use of agents that delay absorption (e.g., aluminum monostearate, gelatin, or a combination thereof) in the compositions.
如本文所用,“有效量”或“治疗有效量”指施用于对象之后至少足以产生疗效的物质、化合物、材料或包含化合物的组合物的量。因此,其为防止、治愈、改善、阻滞或部分阻滞疾病或病症的症状所必需的量。As used herein, "effective amount" or "therapeutically effective amount" refers to an amount of a substance, compound, material, or composition comprising a compound that is at least sufficient to produce a therapeutic effect after administration to a subject. Thus, it is an amount necessary to prevent, cure, ameliorate, block or partially arrest the symptoms of a disease or condition.
对患者施用本发明组合物的实际剂量可根据以下身体和生理因素来确定:体重、性别、症状严重程度、所治疗疾病的类型、先前或当前的治疗干预、患者的未知病因疾病、施用时间、具体化合物的排泄率以及施用途径。在任何情况下,将由负责施用的医务人员确定组合物中活性成分的浓度以及用于个体对象的合适剂量。The actual dosage of a composition of the invention administered to a patient can be determined according to the following physical and physiological factors: body weight, sex, severity of symptoms, type of disease being treated, prior or current therapeutic intervention, unknown etiology of the patient, time of administration, The excretion rate of the specific compound and the route of administration. In any event, the concentration of the active ingredient in the composition and the appropriate dosage for the subject will be determined by the medical personnel responsible for administration.
材料和通用方法Materials and general methods
小鼠Mouse
使用他莫西芬-CreERT2策略构建内皮CXCR7缺失的小鼠。简言之,如前所述(Nature cell biology.2015;17:123-136)在CXCR7侧翼含有loxP-位点的小鼠(CXCR7
f/f)与由Ralf Adams提供的Cdh5-启动子驱动的CreERT2(Cdh5(PAC)-CreERT2+)小鼠(Nature.2010;465:483-486)杂交。产生的后代CXCR7
f/f Cdh5-CreERT2+雄鼠与CXCR7
f/f Cdh5-CreERT2-雌鼠互交产生内皮CXCR7条件性敲除的动物(CXCR7
f/fCdh5-CreERT2+,简称cKO)和同窝对照(CXCR7
f/fCdh5-CreERT2-,简称Ctl)。为了诱导Cre酶介导的CXCR7缺失,给实验小鼠和同窝对照腹腔内注射他莫西芬(Alfa Aesar,Heysham,England)(37.5mg/ml溶于向日葵种子油中),剂量为150mg/kg体重每天,连续三天。然后,让小鼠休息三天,之后再进行另外三天注射。C57BL/6小鼠购自中国食品药品检定研究院并用于评估在MI中腺病毒过表达CXCR7的效果。所有动物方案均获中国国家心血管疾病中心,阜外医院,实验动物中心,机构动物护理和使用委员会的批准。
Endothelial CXCR7 deleted mice were constructed using the tamoxifen-CreERT2 strategy. Briefly, as described previously (Nature cell biology. 2015; 17: 123-136) mice bearing loxP-site flanking CXCR7 (CXCR7 f/f ) and Cdh5-promoter driven by Ralf Adams CreERT2 (Cdh5(PAC)-CreERT2+) mice (Nature. 2010; 465: 483-486) were crossed. The resulting progeny CXCR7 f/f Cdh5-CreERT2+ male mice were crossed with CXCR7 f/f Cdh5-CreERT2-male to produce endothelial CXCR7 conditional knockout animals (CXCR7 f/f Cdh5-CreERT2+, cKO for short) and littermate control (CXCR7 f/f Cdh5-CreERT2-, referred to as Ctl). To induce Cre-mediated CXCR7 deletion, experimental mice and littermates were injected intraperitoneally with tamoxifen (Alfa Aesar, Heysham, England) (37.5 mg/ml in sunflower seed oil) at a dose of 150 mg/ Kg weight per day for three consecutive days. The mice were then allowed to rest for three days before another three days of injection. C57BL/6 mice were purchased from the China Food and Drug Administration and used to evaluate the effect of adenovirus overexpressing CXCR7 in MI. All animal programs were approved by the National Cardiovascular Disease Center, Fuwai Hospital, Laboratory Animal Center, and the Institutional Animal Care and Use Committee.
人主动脉样品Human aortic sample
人主动脉样品获自阜外医院收住的三名主动脉夹层患者。在主动脉修复手术期间,样品收集入生理盐水试管,在10%福尔马林缓冲液中固定过夜,并进一步进行石蜡包埋和组织切片。Human aortic samples were obtained from three aortic dissection patients admitted to the Fuwai Hospital. During the aortic repair procedure, samples were collected into saline tubes, fixed overnight in 10% formalin buffer, and further paraffin embedded and tissue sections.
股动脉损伤模型Femoral artery injury model
如前所述(Circulation.2011;123:631-639)制备股动脉损伤模型。简言之,通过腹腔注射戊巴比妥钠(70mg/kg)麻醉小鼠。一侧腹股沟切口暴露股动脉并使伴随的神经和静脉与之分开。使用6-0号缝合丝线束缚股动脉的近端和远端以便暂时控制血流。分离股直肌与股内侧肌之间的一小段动脉,以6-0号缝合丝线束缚其近端,并在该段横向切开动脉。然后将柔性导丝(直径0.35mm,Cook Inc.,IN,USA)从该段插入股动脉,并向髂动脉方向插入5mm以上。将导丝在该处留置3分钟以剥脱并扩张动脉。然后撤除导丝,将束缚该段动脉近端的缝合丝线扎紧。将用于中断血流的缝合丝线放松使得股动脉血流恢复。使用5-0号缝合丝线闭合皮肤切口。Femoral artery injury models were prepared as previously described (Circulation. 2011; 123:631-639). Briefly, mice were anesthetized by intraperitoneal injection of sodium pentobarbital (70 mg/kg). A one-sided inguinal incision exposes the femoral artery and separates the accompanying nerves and veins. The proximal and distal ends of the femoral artery were tied using a 6-0 suture thread to temporarily control blood flow. A small section of the artery between the rectus femoris and the medial femoral muscle was isolated, and the proximal end was restrained with a 6-0 suture thread, and the artery was laterally cut in the section. A flexible guide wire (diameter 0.35 mm, Cook Inc., IN, USA) was then inserted from the segment into the femoral artery and inserted 5 mm or more into the radial artery. The guidewire was left there for 3 minutes to exfoliate and dilate the artery. The guidewire is then removed and the suture thread that binds the proximal end of the artery is tightened. The suture thread used to interrupt the blood flow is relaxed to restore blood flow to the femoral artery. The skin incision was closed using a 5-0 suture thread.
收集股动脉,石蜡包埋,以150μm的间隔自股动脉节段远端起连续制作10-13层损伤动脉的横切片,用于形态学或组织学分析。对损伤第28天的动脉切片进行H&E染色以观察增生的严重程度,将增生最严重的切片用于比较。使用安装于倒置显微镜 (DM6000B;Leica)的CCD相机获取图像,然后使用Image-Pro Plus 6.0软件(Media Cybernetics)对图像进行测量。采集官腔面积、内弹性层内部面积和外弹性层内部面积并进行分析。为了评估内皮再生和白细胞浸润/迁移,使用相应抗体对损伤第7天的动脉中部进行免疫染色分析。Femoral arteries were collected, paraffin-embedded, and transverse sections of 10-13 layers of injured arteries were continuously made from the distal end of the femoral artery segment at 150 μm intervals for morphological or histological analysis. Arterial sections on day 28 of injury were subjected to H&E staining to observe the severity of hyperplasia, and the most severely propagated sections were used for comparison. Images were acquired using a CCD camera mounted on an inverted microscope (DM6000B; Leica), and images were measured using Image-Pro Plus 6.0 software (Media Cybernetics). The area of the official cavity, the inner area of the inner elastic layer and the inner area of the outer elastic layer were collected and analyzed. To assess endothelial regeneration and leukocyte infiltration/migration, immunostaining analysis was performed on the middle of the artery on day 7 of injury using the corresponding antibodies.
心肌梗死模型Myocardial infarction model
如前所述(Circulation research.2010;107:1445-1453)通过永久结扎左前降支(LAD)冠状动脉造成慢性心肌缺血。简言之,吸入麻醉野生型C57BL/6小鼠,开胸,以6-0号缝合丝线在距离起始处2-3mm的位置结扎LAD。为过表达CXCR7,在冠状动脉结扎之前1分钟给每只小鼠左心室内注射表达小鼠CXCR7的重组腺病毒(Ad-CXCR7)(使用小鼠CXCR7 mRNA(NM_001271607.1)的第418-1506位的核苷酸序列用于构建重组腺病毒)或空载体,注射剂量为每只小鼠1×10
9噬菌斑形成单位。通过左心室颜色改变和心电图(ECG)ST-段抬高确认结扎的成功。
Chronic myocardial ischemia was caused by permanent ligation of the left anterior descending (LAD) coronary artery as previously described (Circulation research. 2010; 107: 1445-1453). Briefly, wild-type C57BL/6 mice were inhaled and the thorax was opened, and LAD was ligated at a position 2-3 mm from the start with a 6-0 suture thread. To overexpress CXCR7, a recombinant adenovirus expressing mouse CXCR7 (Ad-CXCR7) was injected into the left ventricle of each mouse 1 minute prior to coronary artery ligation (using 418-1506 of mouse CXCR7 mRNA (NM_001271607.1)) The nucleotide sequence of the position is used to construct a recombinant adenovirus or an empty vector at a dose of 1 x 10 9 plaque forming units per mouse. The success of the ligation was confirmed by left ventricular color change and electrocardiogram (ECG) ST-segment elevation.
然后闭合胸腔并停止麻醉,继续呼吸机给入空气2-5分钟后让动物逐渐苏醒。在第7天通过超声心动图(VisualSonics VeVo 2100 Imaging System)测定心功能和左室结构,测量指标为左心室射血分数(EF)、左心室部分面积的变化(FAC)、左室内径缩短分数(FS)、二尖瓣舒张早/晚期血流峰值速度比(E/A)、左室前壁厚度(LVAW)、左室后壁厚度(LVPW)和左室容积,并测量左室大小。在实验结束时(第28天)灌注并福尔马林固定后采集心脏并石蜡包埋。通过Masson染色(Circulation.2015;132:47-58)评估心脏梗死情况。简言之,以200μm间隔获得一系列胸骨旁短轴切片(厚度5μm)。使用Masson三色试剂(Leagene Biotec.Co,Ltd)对代表性中层切片进行染色并使用Zeiss光学显微镜(AXI0;Zeiss)拍摄。使用Image-Pro Plus 6.0软件(Media Cybernetics)测量并计算梗死面积。Then close the chest and stop the anesthesia, continue to ventilate the air for 2-5 minutes and let the animal gradually wake up. On day 7, cardiac function and left ventricular structure were measured by echocardiography (VisualSonics VeVo 2100 Imaging System), and the measured indicators were left ventricular ejection fraction (EF), left ventricular partial area change (FAC), and left ventricular diameter shortening score. (FS), mitral valve diastolic early/late blood flow peak velocity ratio (E/A), left ventricular anterior wall thickness (LVAW), left ventricular posterior wall thickness (LVPW), and left ventricular volume, and left ventricular size were measured. At the end of the experiment (Day 28), the hearts were harvested and formalized with formalin and embedded in paraffin. Cardiac infarction was assessed by Masson staining (Circulation. 2015; 132: 47-58). Briefly, a series of parasternal short-axis slices (thickness 5 μm) were obtained at 200 μm intervals. Representative midsection sections were stained using Masson's trichrome reagent (Leagene Biotec. Co, Ltd) and photographed using a Zeiss optical microscope (AXI0; Zeiss). Infarct size was measured and calculated using Image-Pro Plus 6.0 software (Media Cybernetics).
小鼠后肢缺血模型Mouse hind limb ischemia model
通过结扎左侧股动脉造成后肢缺血(HLI),结扎部位在股动脉分叉出隐动脉处的远端(Arteriosclerosis,thrombosis,and vascular biology.2014;34:408-418)。使用激光多普勒血流仪(LDF;PeriCam PSI)在结扎之前以及结扎后立即测量后肢的血流。将结扎后做后肢血流下降不低于50%的小鼠纳入本实验。对于成功结扎动脉的小鼠,分别在第4、7和14天测量三次血流。均采用盲法测量并分析血流。在第21天,通过施用过量的镇静剂处死小鼠。分了腓肠肌,固定并石蜡包埋,用于分析血管密度。简言之, 取每一腓肠肌的最大横切面进行染色,病在每一切片中随机拍摄三个肌间隙。然后计数并分析照片中的血管。Hind limb ischemia (HLI) is caused by ligation of the left femoral artery, and the ligation site is bifurcated at the distal end of the saphenous artery (Arteriosclerosis, thrombosis, and vascular biology. 2014; 34: 408-418). Blood flow to the hind limbs was measured before and after ligation using a laser Doppler flow meter (LDF; PeriCam PSI). Mice in which the blood flow of hind limbs was reduced by not less than 50% after ligation were included in the experiment. For mice that successfully ligated the artery, blood flow was measured three times on days 4, 7, and 14, respectively. Blood flow was measured and analyzed blindly. On day 21, mice were sacrificed by administration of an excess of sedative. The gastrocnemius muscle was divided, fixed and paraffin embedded for analysis of vascular density. Briefly, the largest transverse section of each gastrocnemius muscle was stained, and three muscle gaps were randomly photographed in each section. The blood vessels in the photo are then counted and analyzed.
细胞实验Cell experiment
细胞分离:如前所述(Blood.2011;118:464-472)分离小鼠肺内皮细胞(MLEC)。简言之,用无菌PBS通过右心室灌注小鼠,移除血细胞。分离肺叶,切碎,并用胶原酶(180-200U/mL;Worthington)在37℃消化(40分钟)。在通过75μm细胞过滤器(BD Biosciences)过滤后,用包被抗-小鼠CD31(BD Biosciences)的Dynabeads(Dynal Biotech)孵育细胞。用磁分离器(Dynal)分离珠上的细胞,然后在胶原蛋白I(Worthington)包被的培养皿/培养瓶中培养3天,其中含有添加20%胎牛血清(FBS),1%AA(GIBCO),以及100mg/L内皮细胞生长添加剂(ECGS;ScienCell)的DMEM。解离细胞并用包被大鼠抗-小鼠CD102(ICAM-2;Pharmingen)的Dynabeads对其进行选择。培养分离的MLEC。Cell isolation: Mouse lung endothelial cells (MLEC) were isolated as previously described (Blood. 2011; 118: 464-472). Briefly, mice were perfused through the right ventricle with sterile PBS to remove blood cells. The lung lobes were isolated, minced, and digested with collagenase (180-200 U/mL; Worthington) at 37 ° C (40 minutes). After filtration through a 75 μm cell strainer (BD Biosciences), the cells were incubated with Dynabeads (Dynal Biotech) coated with anti-mouse CD31 (BD Biosciences). The cells on the beads were separated using a magnetic separator (Dynal) and then cultured for 3 days in a collagen I (Worthington) coated Petri dish/culture flask containing 20% fetal bovine serum (FBS), 1% AA ( GIBCO), and DMEM of 100 mg/L endothelial cell growth additive (ECGS; ScienCell). The cells were dissociated and selected with Dynabeads coated with rat anti-mouse CD102 (ICAM-2; Pharmingen). The isolated MLEC is cultured.
如前所述(Cell metabolism.2011;13:592-600)分离小鼠主动脉内皮细胞(MAEC)。简言之,收集主动脉并去除外膜周围脂肪和结缔组织,并剪成1-2mm
2的小段。在培养基中培养主动脉片段5-7天,使其长出内皮细胞。然后将内皮细胞传代并培养。为了分离人主动脉内皮细胞(HAEC),在DMEM中收集来自阜外医院的人主动脉样品。剥脱主动脉内膜层并以与MAEC培养相同的方式处理。
Mouse aortic endothelial cells (MAEC) were isolated as previously described (Cell metabolism. 2011; 13: 592-600). Briefly, the aorta was collected and the fat and connective tissue surrounding the adventitia were removed and cut into small pieces of 1-2 mm 2 . The aortic fragments were cultured in medium for 5-7 days to grow endothelial cells. The endothelial cells are then passaged and cultured. To isolate human aortic endothelial cells (HAEC), human aortic samples from an external hospital were collected in DMEM. The aortic intimal layer was exfoliated and treated in the same manner as MAEC culture.
如前所述(Journal of cellular and molecular medicine.2014;18:2266-2274)分离心肌成纤维细胞。简言之,分离新生Wistar大鼠的心室,清洗并于PBS中切碎。然后于37℃在包含0.06%胶原酶(Worthington)的PBS中消化组织。收集的细胞悬浮液经离心并在包含10%的DMEM中重悬。将重悬液至于培养瓶中并培养90分钟。成纤维细胞倾向于附着在底部。去除非粘附细胞。培养粘附的心肌成纤维细胞并随后用胰酶传代。Myocardial fibroblasts were isolated as previously described (Journal of cellular and molecular medicine. 2014; 18: 2266-2274). Briefly, the ventricles of newborn Wistar rats were isolated, washed and minced in PBS. The tissue was then digested in PBS containing 0.06% collagenase (Worthington) at 37 °C. The collected cell suspension was centrifuged and resuspended in 10% DMEM. The suspension was placed in a culture flask and incubated for 90 minutes. Fibroblasts tend to adhere to the bottom. Remove non-adherent cells. Adherent cardiac fibroblasts are cultured and subsequently passaged with trypsin.
通过RNA干扰进行基因沉默:为了敲低内皮细胞中的CXCR7、CXCR4、β-arrestin1或β-arrestin2蛋白,进行siRNA基因沉默。简言之,将内皮细胞植于12孔板中。在转染前,将40pmol siRNA与2.0μL Hieff Trans
TM Liposomal Transfection Reagent(Yeasen,China)在200μL DMEM中混合20分钟。将培养基换成DMEM。20分钟后,将siRNA转染试剂混合物加入孔中(200μL/孔)。转染持续6小时。之后弃 去转染培养基,在进一步分析之前,将细胞在含有20%FBS的培养基中培养不少于6小时。用于本研究的siRNA如下:
Gene silencing by RNA interference: siRNA gene silencing was performed in order to knock down CXCR7, CXCR4, β-arrestin1 or β-arrestin2 proteins in endothelial cells. Briefly, endothelial cells were seeded in 12-well plates. Before transfection, 40pmol siRNA mixed with 2.0μL Hieff Trans TM Liposomal Transfection Reagent ( Yeasen, China) in 200μL DMEM for 20 minutes. The medium was changed to DMEM. After 20 minutes, the siRNA transfection reagent mixture was added to the wells (200 μL/well). Transfection lasted for 6 hours. The transfection medium was then discarded and the cells were cultured in medium containing 20% FBS for not less than 6 hours prior to further analysis. The siRNA used in this study is as follows:
阴性对照:Negative control:
UUUUCCGAACGUGUCACGUTT;UUUUCCGAACGUGUCACGUTT;
si-CXCR7:si-CXCR7:
CCCUGGAACAGAACACCAATT,CCCUGGAACAGAACACCAATT,
GCAACUACUCUGACAUCAATT,GCAACUACUCUGACAUCAATT,
GCAAGAUCACACACCUCAUTT;GCAAGAUCACACACCUCAUTT;
si-CXCR4(Arteriosclerosis,thrombosis,and vascular biology.2014;34:1716-1722):si-CXCR4 (Arteriosclerosis, thrombosis, and vascular biology. 2014; 34: 1716-1722):
TGTCTCAACCGAGTCTGAATCTTCA,TGTCTCAACCGAGTCTGAATCTTCA,
TGGTACTTTGGGAAGTTCCTCTGCA,TGGTACTTTGGGAAGTTCCTCTGCA,
CAGTTATCCTCATCCTGACTTTCTT,CAGTTATCCTCATCCTGACTTTCTT,
GATCCGTATATTCACTTCCGATAAT;GATCCGTATATTCACTTCCGATAAT;
si-β-arrestin1(Proceedings of the National Academy of Sciences of the United States of America.2010;107:628-632):Si-β-arrestin1 (Proceedings of the National Academy of Sciences of the United States of America. 2010; 107: 628-632):
AGCCUUCUGUGCUGAGAAC;AGCCUUCUGUGCUGAGAAC;
si-β-arrestin2(Proceedings of the National Academy of Sciences of the United States of America.2010;107:628-632):Si-β-arrestin 2 (Proceedings of the National Academy of Sciences of the United States of America. 2010; 107: 628-632):
GGACCGCAAAGUGUUUGUG。GGACCGCAAAGUGUUUGUG.
细胞增殖研究:使用细胞计数试剂盒-8(CCK-8;Yeasen,Shanghai,China)测量细胞生长。简言之,将细胞植于9孔平底板中。在细胞完全附着于底部后,在含有3%FBS但不含ECGS的培养基中饥饿细胞6-8h。然后用培养基-CCK-8混合物(10∶1体积比)更换培养基。4小时后,测量450nm的吸光值作为本底。然后用指定试剂孵育细胞48小时。最后,再次用培养基-CCK-8混合物替换培养基。4小时后,测量450nm的吸光值以显示细胞生长。用于细胞研究中的试剂浓度如下:10ng/mL IL-1β,1μM CCX771或CCX704(两种化合物均由ChemoCentryx,Inc.,Mountain View,CA,USA提供)。Cell proliferation studies: Cell growth was measured using Cell Counting Kit-8 (CCK-8; Yeasen, Shanghai, China). Briefly, cells were planted in a 9-well flat bottom plate. After the cells were completely attached to the bottom, the cells were starved for 6-8 h in medium containing 3% FBS but no ECGS. The medium was then replaced with a medium-CCK-8 mixture (10:1 by volume). After 4 hours, the absorbance at 450 nm was measured as a background. The cells were then incubated with the indicated reagents for 48 hours. Finally, the medium was replaced again with the medium-CCK-8 mixture. After 4 hours, the absorbance at 450 nm was measured to show cell growth. The reagent concentrations used in the cell studies were as follows: 10 ng/mL IL-1 β, 1 μM CCX771 or CCX704 (both compounds were supplied by ChemoCentryx, Inc., Mountain View, CA, USA).
小管形成测定:低氧-条件性培养基用于内皮小管形成测定。简言之,在包含3%FBS的DMEM中的心脏成纤维细胞接受由AnaeroPack-Anaero(MGC,Japan)造成的低 氧12小时。收集条件性培养基。为了进行小管形成测定,使用冷吸管将生长因子减少的Matrigel(Corning,NY,USA)分散于96孔板中(40μL/well)。然后于37℃使Matrigel聚合1小时。然后用胰酶消化内皮细胞,重悬于条件性培养基中,并以2×10
4细胞每孔的浓度植于平板中。对于炎性刺激,细胞在含有20%FBS的培养基中用IL-1β(10ng/mL)或TNFα(25ng/mL)预处理6-8小时。每2小时使用具有5×物镜的倒置相差成像显微镜(莱卡,德国)观察微管形成并拍照。来自第6小时的结果用于分析。记录并分析至少5张来自不同细胞的图像。
Tubule formation assay: hypoxic-conditioned media for endothelial tubule formation assays. Briefly, cardiac fibroblasts in DMEM containing 3% FBS received hypoxia for 12 hours by AnaeroPack-Anaero (MGC, Japan). Conditioned medium was collected. For tubelet formation assays, Matrigel (Corning, NY, USA) with reduced growth factors was dispersed in 96-well plates (40 μL/well) using a cold pipette. Matrigel was then polymerized at 37 ° C for 1 hour. Endothelial cells were then trypsinized, resuspended in conditioned medium, and plated in plates at a concentration of 2 x 10 4 cells per well. For inflammatory stimulation, cells were pretreated with IL-1β (10 ng/mL) or TNFα (25 ng/mL) for 6-8 hours in medium containing 20% FBS. Microtubule formation was observed and photographed every 2 hours using an inverted phase contrast imaging microscope (Leica, Germany) with a 5x objective. Results from the 6th hour were used for analysis. Record and analyze at least 5 images from different cells.
损伤愈合测定:对于细胞迁移测定,将内皮细胞植入12孔板。当达到90%生长汇合时,用siRNA转染细胞。然后在包含3%FBS和10ng/mL IL-1β的培养基中培养细胞6-8小时。之后使用无菌移液器尖刮每个孔中的单层细胞以产生无细胞区。用PBS清洗去除脱落的细胞。使用具有5×物镜的成像显微镜(莱卡,德国)立即对损伤进行拍照和记录。24小时后,再次对损伤进行拍照和记录。每个孔拍摄5张连续的显微照片。使用Image-Pro Plus 6.0软件(Media Cybernetics)对迁移至损伤区域的细胞区域进行定量。Damage healing assay: For cell migration assays, endothelial cells were seeded into 12-well plates. When 90% growth confluence was reached, cells were transfected with siRNA. The cells were then cultured in medium containing 3% FBS and 10 ng/mL IL-1β for 6-8 hours. The monolayer cells in each well were then scraped with a sterile pipette tip to create a cell free zone. The detached cells were removed by washing with PBS. The lesion was immediately photographed and recorded using an imaging microscope (Leica, Germany) with a 5x objective. After 24 hours, the injury was photographed and recorded again. Five consecutive photomicrographs were taken for each well. Cell areas that migrated to the damaged area were quantified using Image-Pro Plus 6.0 software (Media Cybernetics).
钙离子响应测定:具有阴性siRNA或si-CXCR7的细胞植于96孔板中并在包含20%FBS和10ng/mL IL-1β的培养基中培养过夜。对于钙离子荧光染色,细胞用Cal-520
TM染料上样溶液(AAT Bioquest,Sunnyvale,CA)培养2小时。使用FlexStation3 Microplate Reader(Molecular Devices,USA),分别在波长490和525nm的激发光和发射光监测荧光。在17秒的基线测量后,添加100ng/mL CXCL12,并对产生的钙离子响应另外测量78秒钟。CXCR4抑制剂AMD3100(1μg/mL)在CXCL12刺激之前2小时添加。
Calcium ion response assay: Cells with negative siRNA or si-CXCR7 were plated in 96-well plates and grown overnight in medium containing 20% FBS and 10 ng/mL IL-1β. For Calcium Fluorescence staining, cell-like solution (AAT Bioquest, Sunnyvale, CA) with a Dye Cal-520 TM incubated for 2 hours. Fluorescence was monitored using excitation and emission at wavelengths of 490 and 525 nm, respectively, using a FlexStation 3 Microplate Reader (Molecular Devices, USA). After a 17 second baseline measurement, 100 ng/mL CXCL12 was added and the resulting calcium ion response was additionally measured for 78 seconds. The CXCR4 inhibitor AMD3100 (1 μg/mL) was added 2 hours before CXCL12 stimulation.
成像流式细胞分析测定:使用ImageStreamX Mark II Imaging Flow Cytometer(Merck,Darmstadt,Germany)在HUVEC中分析CXCR4和CXCR7的细胞表面表达。简言之,在12孔板中转染了si-RNA或si-CXCR7的HUVEC用IL-1β(10ng/mL)处理6-8小时。然后胰酶消化、离心并在100μL FACS缓冲液(包含0.6mg/mL牛血清白蛋白和0.3mM EDTA的HBSS)中重悬。将抗体添加到细胞悬液中置于冰上30分钟进行染色。用抗CXCR7mAb(11G8,1∶100)和Alexa Fluor-594-缀合的二抗对CXCR7进行染色。用CXCR4抗体(多抗,1∶200,Sigma)和Alexa Fluor-488-缀合的二抗对 CXCR4进行标记。然后使用ImageStreamX Mark II Imaging Flow Cytometer对细胞进行分析和拍照。Imaging flow cytometric assay: Cell surface expression of CXCR4 and CXCR7 was analyzed in HUVEC using ImageStreamX Mark II Imaging Flow Cytometer (Merck, Darmstadt, Germany). Briefly, HUVECs transfected with si-RNA or si-CXCR7 in 12-well plates were treated with IL-1β (10 ng/mL) for 6-8 hours. It was then trypsinized, centrifuged and resuspended in 100 μL of FACS buffer (HBSS containing 0.6 mg/mL bovine serum albumin and 0.3 mM EDTA). The antibody was added to the cell suspension and placed on ice for 30 minutes for staining. CXCR7 was stained with anti-CXCR7 mAb (11G8, 1:100) and Alexa Fluor-594-conjugated secondary antibody. CXCR4 was labeled with CXCR4 antibody (polyclonal antibody, 1:200, Sigma) and Alexa Fluor-488-conjugated secondary antibody. Cells were then analyzed and photographed using the ImageStreamX Mark II Imaging Flow Cytometer.
RNA分离和定量实时PCRRNA isolation and quantitative real-time PCR
为了进行RNA分离,在TRIzol(Invitrogen)中裂解细胞。然后将溶液与氯仿(5∶1,体积比)混合并离心(12,000g;15min;4℃)。收集水相,与异丙醇混合,离心(12,000g;10min;4℃)。用75%酒精清洗后,得到的RNA可备用。使用
Select Master Mix(Invitrogen)进行定量RT-PCR。本研究中所用引物如下:
For RNA isolation, cells were lysed in TRIzol (Invitrogen). The solution was then mixed with chloroform (5:1, by volume) and centrifuged (12,000 g; 15 min; 4 °C). The aqueous phase was collected, mixed with isopropanol and centrifuged (12,000 g; 10 min; 4 ° C). After washing with 75% alcohol, the resulting RNA can be used. use Select Master Mix (Invitrogen) for quantitative RT-PCR. The primers used in this study are as follows:
β-actin正向:ACCTTCTACAATGAGCTGCG;--actin forward: ACCTTCTACAATGAGCTGCG;
β-actin反向:CTGGATGGCTACGTACATGG;--actin reverse: CTGGATGGCTACGTACATGG;
CXCR7正向:TTCATCAACCGCAACTACA;CXCR7 forward: TTCATCAACCGCAACTACA;
CXCR7反向:TCTCCTCTTCATACCACTCA;CXCR7 reverse: TCTCCTCTTCATACCACTCA;
CXCR4正向:CTCTACAGCAGCGTTCTC;CXCR4 forward: CTCTACAGCAGCGTTCTC;
CXCR4反向:TCAGGTATAGTCAGGAGGAG;CXCR4 reverse: TCAGGTATAGTCAGGAGGAG;
CXCL12正向:ACGGCTGAAGAACAACAA;CXCL12 forward: ACGGCTGAAGAACAACAA;
CXCL12反向:GAAGATGAGGATGAGGAGAAT;CXCL12 reverse: GAAGATGAGGATGAGGAGAAT;
eNOS正向:GGCATCACCAGGAAGAAG;eNOS forward: GGCATCACCAGGAAGAAG;
eNOS反向:GGACACCACATCATACTCAT。eNOS reverse: GGACACCACATCATACTCAT.
免疫荧光Immunofluorescence
来自石蜡包埋的组织的切片(5μm)经脱蜡、再水化,并通过在EDTA抗原修复水(PH 9.0;ZSGB-BIO,北京,中国)中煮沸2分钟进行抗原修复。将HUVEC铺在盖玻片(NUNC)上,并在4%多聚甲醛中固定20分钟。然后将样品与含0.3%Triton X-100的山羊血清孵育,进行封闭和膜破裂。孵育后,与一抗在4℃孵育过夜,样品与Alexa Fluor-594偶联和/或Alexa Fluor-488偶联的二抗在室温孵育3小时。用含有DAPI的VectaShield培养基覆盖盖玻片以染核。使用配有Zen软件的蔡司倒置荧光显微镜(AXI0;Zeiss)或配有20×水浸物镜的激光扫描共聚焦显微镜(SP8;Leica)对切片进行成像。使用Image-Pro Plus 6.0软件(Media Cybernetics,Inc.Rockville,MD,USA)对图像进行分析。所用抗体包括单抗抗-CXCR7(11G8;1∶100;由ChemoCentryx,Inc.提供),多抗抗-vWF(1∶800;Sigma),多抗抗-PDGF-BB(1∶200;Abcam),单抗抗-F4/80(1∶50;BM8;Abcam),单抗抗-βArrestin2(1∶100;Abcam),多抗抗-p-ERK(1∶100;CST)。Sections (5 μm) from paraffin-embedded tissues were dewaxed, rehydrated, and subjected to antigen retrieval by boiling in EDTA antigen-repairing water (pH 9.0; ZSGB-BIO, Beijing, China) for 2 minutes. HUVEC was plated on coverslips (NUNC) and fixed in 4% paraformaldehyde for 20 minutes. The samples were then incubated with goat serum containing 0.3% Triton X-100 for blocking and membrane disruption. After incubation, the antibody was incubated overnight at 4 °C with the primary antibody and the samples were incubated with Alexa Fluor-594 coupled and/or Alexa Fluor-488 conjugated secondary antibody for 3 hours at room temperature. The coverslips were covered with VectaShield medium containing DAPI to stain the nuclei. Sections were imaged using a Zeiss inverted fluorescence microscope (AXI0; Zeiss) equipped with Zen software or a laser scanning confocal microscope (SP8; Leica) equipped with a 20x water immersion objective. Images were analyzed using Image-Pro Plus 6.0 software (Media Cybernetics, Inc. Rockville, MD, USA). Antibodies used included monoclonal antibody anti-CXCR7 (11G8; 1:100; supplied by ChemoCentryx, Inc.), polyclonal anti-vWF (1:800; Sigma), polyclonal anti-PDGF-BB (1:200; Abcam) , mAb anti-F4/80 (1:50; BM8; Abcam), mAb anti-beta Arrestin 2 (1:100; Abcam), polyclonal anti-p-ERK (1:100; CST).
血浆中的分析Analysis in plasma
血浆获自离心(6000g,5min,4℃)后的EDTA抗凝血血液。使用人CXCL12ELISA试剂盒(R&D Systems,Minnesota,USA)确定CXCL12浓度。使用人PDGF-BB Quantikine ELISA试剂盒(R&D Systems,Minnesota,USA)确定PDGF-BB浓度。使用自动生化分析仪(AU5421,Beckman Coulter,California,USA)对血浆葡萄糖(GLU)、总胆固醇(CHOL、甘油三酸酯(TG)、游离脂肪酸(FFA)、低密度脂蛋白胆固醇(LDL-C)、高密度脂蛋白胆固醇(HDL-C)和丙氨酸转氨酶(ALT)进行测量。Plasma was obtained from EDTA anticoagulant blood after centrifugation (6000 g, 5 min, 4 °C). The CXCL12 concentration was determined using a human CXCL12 ELISA kit (R&D Systems, Minnesota, USA). PDGF-BB concentrations were determined using a human PDGF-BB Quantikine ELISA kit (R&D Systems, Minnesota, USA). Plasma glucose (GLU), total cholesterol (CHOL, triglyceride (TG), free fatty acid (FFA), low density lipoprotein cholesterol (LDL-C) using an automated biochemical analyzer (AU5421, Beckman Coulter, California, USA) ), high density lipoprotein cholesterol (HDL-C) and alanine aminotransferase (ALT) were measured.
蛋白印迹分析Western blot analysis
为了阐明所涉及的信号通路,将MAEC在含有3%FBS的培养基中预饥饿6-8h,加入指定的试剂温预12小时。然后在含有蛋白酶抑制剂(Roche,Basel,Switherland)的RIPA缓冲液中裂解细胞,离心(15800g,10min)后将细胞裂解物与上样缓冲液混合并以10%SDS-PAGE分离,之后转移至PVDF膜上。用指定抗体与膜杂交。一些膜在脱色后与actin抗体重新杂交。使用了如下一抗:针对phospho-ERK的多克隆抗体(1∶1000;CST),针对ERK的多克隆抗体(1∶1000;CST),针对phospho-P38的多克隆抗体(1∶1000;CST),针对P38的多克隆抗体(1∶1000;CST),针对phospho-JNK的多克隆抗体(1∶1000;CST),针对JNK的多克隆抗体(1∶1000;CST),针对phospho-eNOS的多克隆抗体(1∶500;CST),针对CXCR4的多克隆抗体(1∶1000;Sigma),和针对CXCR7的多克隆抗体(1∶500;Proteintech,Rosemont,USA)。To elucidate the signaling pathway involved, MAEC was pre-starved for 6-8 h in medium containing 3% FBS and added to the indicated reagent temperature for 12 hours. The cells were then lysed in RIPA buffer containing protease inhibitors (Roche, Basel, Switherland), centrifuged (15800 g, 10 min), cell lysates were mixed with loading buffer and separated by 10% SDS-PAGE, then transferred to On the PVDF membrane. Hybridization of the membrane with the designated antibody. Some membranes re-hybridize with actin antibodies after decolorization. The following primary antibodies were used: polyclonal antibody against phospho-ERK (1:1000; CST), polyclonal antibody against ERK (1:1000; CST), polyclonal antibody against phospho-P38 (1:1000; CST) , polyclonal antibody against P38 (1:1000; CST), polyclonal antibody against phospho-JNK (1:1000; CST), polyclonal antibody against JNK (1:1000; CST), for phospho-eNOS Polyclonal antibody (1:500; CST), polyclonal antibody against CXCR4 (1:1000; Sigma), and polyclonal antibody against CXCR7 (1:500; Proteintech, Rosemont, USA).
颈动脉血栓模型Carotid thrombosis model
如前所述(Circulation research.2014;114:947-956;Blood.2000;95:577-580)制备小鼠颈动脉激光血栓模型。简言之,给麻醉的小鼠(戊巴比妥钠,70mg/kg)静脉注射玫瑰红(50mg/kg)。将左侧颈总动脉暴露于2.5-mW绿色激光(540nm;Melles Griot Inc)。从出现损伤开始以脉冲多普勒(Transonic,Sidney,Australia)连续检测血流直至出现稳定的阻塞(定义为2分钟内无血流),或在无阻塞发生的情况下连续检测90分钟。阻塞时间定义为血管损伤起始至出现稳定阻塞之间的时间。为了计算平均阻塞时间,对于无阻塞发生的动物,其阻塞时间归为90分钟。A mouse carotid laser thrombosis model was prepared as previously described (Circulation research. 2014; 114: 947-956; Blood. 2000; 95: 577-580). Briefly, anesthetized mice (sodium pentobarbital, 70 mg/kg) were intravenously injected with rose bengal (50 mg/kg). The left common carotid artery was exposed to a 2.5-mW green laser (540 nm; Melles Griot Inc). Blood flow was continuously detected by pulsed Doppler (Transonic, Sidney, Australia) from the onset of injury until a stable blockage (defined as no blood flow within 2 minutes) or 90 minutes without occlusion occurred. The occlusion time is defined as the time between the onset of vascular injury and the occurrence of stable occlusion. To calculate the average occlusion time, the obstruction time for animals that did not occlude was classified as 90 minutes.
血小板聚集测定Platelet aggregation assay
将ACD抗凝的全血(ACD∶血液体积比为1∶9)与相同体积的预加热的生理盐水混 合,加入胶原(2μg/mL)诱导聚集。测定前将样品在37℃平衡7分钟。然后在Chronolog710凝集计(Chronolog,Havertown,PA,USA)中在37℃恒定搅拌下(1200rpm)进行血小板聚集。记录结果并采用Aggro/Link5软件(Chronolog,Havertown,PA,USA)分析数据。ACD anticoagulated whole blood (ACD: blood volume ratio 1:9) was mixed with the same volume of pre-warmed physiological saline, and collagen (2 μg/mL) was added to induce aggregation. The sample was equilibrated at 37 ° C for 7 minutes before the measurement. Platelet aggregation was then carried out in a Chronolog 710 aggregometer (Chronolog, Havertown, PA, USA) under constant agitation (1200 rpm) at 37 °C. The results were recorded and analyzed using Aggro/Link5 software (Chronolog, Havertown, PA, USA).
血小板活化Platelet activation
为了测定CXCL12自血小板的释放,分别在体内和体外活化血小板。在体外,血小板活化方法与测定血小板聚集所用方法相同。简言之,将ACD抗凝的全血与相同体积的预加热的生理盐水混合。将样品在37℃平衡7分钟,然后加入胶原(2μg/mL)或U46619(2mM;Sigma)以活化血小板。30分钟后离心(6000g,5min,4℃)并收集血浆用于检测CXCL12。在体内,给麻醉的小鼠静脉注射U46619(20ug/小鼠)。在10分钟内收集EDTA抗凝全血。To determine the release of CXCL12 from platelets, platelets were activated in vivo and in vitro, respectively. In vitro, the platelet activation method is the same as that used to measure platelet aggregation. Briefly, ACD anticoagulated whole blood was mixed with the same volume of pre-warmed physiological saline. The samples were equilibrated at 37 °C for 7 minutes and then collagen (2 μg/mL) or U46619 (2 mM; Sigma) was added to activate platelets. After 30 minutes, centrifugation (6000 g, 5 min, 4 ° C) and plasma were collected for detection of CXCL12. In vivo, anesthetized mice were intravenously injected with U46619 (20 ug/mouse). EDTA anticoagulated whole blood was collected within 10 minutes.
血小板耗竭Platelet depletion
通过静脉注射CD41抗体(20ug/小鼠;Ebioscience,USA)耗竭血小板。分别在注射前、注射后2小时和5天收集EDTA抗凝全血样品。Platelets were depleted by intravenous injection of CD41 antibody (20 ug/mouse; Ebioscience, USA). EDTA anticoagulated whole blood samples were collected before injection, 2 hours and 5 days after injection, respectively.
研究人群Research population
本研究中的患者为自2014年8月至2015年1月连续收入阜外医院CCU的95名诊断为急性心肌梗死(AMI)并立即进行PCI的患者。The patients in this study were 95 patients who were diagnosed with acute myocardial infarction (AMI) and who underwent PCI immediately from August 2014 to January 2015.
人血收集和血小板反应性测定Human blood collection and platelet reactivity determination
在PCI后4天采集4毫升枸橼酸抗凝的肘静脉血,使用VerifyNow System(Accumetrics,San Diego,CA,USA)检测血小板功能。枸橼酸抗凝血在室温存放4小时内用于阿司匹林和P2Y12测定。结果以血小板的P2Y12反应单位(PRU)和阿司匹林反应单位(ARU)表示。所有进行PCI的患者在PCI之前均口服阿司匹林和负荷量的300mg的波立维,之后每天口服100mg的阿司匹林和75mg的波立维。STEMI的患者如果他们在收入之前没有服用过阿司匹林,则在PCI之前服用负荷量的300mg的阿司匹林。患者情况总结于表1。本研究的方案经北京协和医学院和中国医学科学院阜外医院医学伦理学委员会的批准,并获得每一患者的书面知情同意。Four milliliters of citrate anticoagulated venous blood was collected 4 days after PCI, and platelet function was measured using a VerifyNow System (Accumetrics, San Diego, CA, USA). Citric acid anticoagulation was used for aspirin and P2Y12 assays for 4 hours at room temperature. Results are expressed in platelet P2Y12 response units (PRU) and aspirin response units (ARU). All PCI patients received oral aspirin and a loading of 300 mg of Plavix prior to PCI, followed by oral administration of 100 mg of aspirin and 75 mg of Plavix daily. STEMI patients take 300 mg of aspirin before PCI if they have not taken aspirin before their income. Patient status is summarized in Table 1. The protocol of this study was approved by the Peking Union Medical College and the Medical Ethics Committee of the Fuwai Hospital of the Chinese Academy of Medical Sciences, and obtained written informed consent from each patient.
表1.患者的基线表征Table 1. Baseline characterization of patients
统计学分析Statistical analysis
在一个实验的不同时间点获得的数据使用重复测量的ANOVA进行分析。采用 Tukey多重检验校正进行ANOVA比较。当只比较2个平均值时,使用双尾Mann-Whitney t检验。对数(Mantel-Cox)检验用于比较两组之间的存活差异。数据以平均值±SEM表示。P<0.05时认为有统计学显著差异。Data obtained at different time points of one experiment were analyzed using repeated measures ANOVA. ANOVA comparisons were performed using Tukey multiple test calibration. When only 2 averages were compared, a two-tailed Mann-Whitney t test was used. The logarithmic (Mantel-Cox) test was used to compare the difference in survival between the two groups. Data are expressed as mean ± SEM. A statistically significant difference was considered at P < 0.05.
实施例1:小鼠和人损伤动脉中CXCR7的内皮表达Example 1: Endothelial expression of CXCR7 in injured arteries of mice and humans
首先检查了小鼠中CXCR7的血管表达。CXCR7在健康小鼠分散的内皮细胞中低水平表达(图1A)。然而在受损动脉中,CXCR7表达上调,并且主要见于新生内膜的内皮细胞中,与内皮细胞标志物vWF共定位(图1B)。来自进行主动脉夹层的患者的人主动脉的受损内皮细胞中有CXCR7表达,特别是在动脉粥样硬化斑块肩部(图1C)和斑块中的微血管中(图1D)。The vascular expression of CXCR7 in mice was first examined. CXCR7 is expressed at low levels in healthy mouse dispersed endothelial cells (Fig. 1A). However, in damaged arteries, CXCR7 expression is upregulated and is mainly found in endothelial cells of the neointimal, colocalizing with the endothelial cell marker vWF (Fig. 1B). There is CXCR7 expression in damaged endothelial cells from the human aorta of patients undergoing aortic dissection, particularly in the atherosclerotic plaque shoulder (Fig. 1C) and microvessels in the plaque (Fig. 1D).
实施例2:内皮CXCR7缺失加剧小鼠内皮剥脱损伤后的新生内膜形成Example 2: Endothelial CXCR7 deletion aggravates neointimal formation after endothelial exfoliation injury in mice
他莫西芬诱导之前,cKO小鼠和同窝对照的CXCR7基线动脉表达类似(图1E)。他莫西芬处理后,小鼠在股动脉通过血管成形术进行内皮剥脱损伤。该损伤诱导血管增生反应,模拟经皮冠状动脉介入术后的临床再狭窄。所有小鼠均未进行与脂代谢有关的基因操作,并饲喂正常饮食。The baseline arterial expression of CXCR7 in cKO mice and littermates was similar before tamoxifen induction (Fig. 1E). After tamoxifen treatment, mice underwent endothelium exfoliation injury by angioplasty in the femoral artery. This injury induces vascular hyperplasia and mimics clinical restenosis after percutaneous coronary intervention. All mice were not subjected to genetic manipulation related to lipid metabolism and fed a normal diet.
从cKO小鼠中分离得到的内皮细胞中内皮CXCR7的mRNA表达基本上为零(图2A),这得到了受损动脉免疫染色的进一步确认(图2B)。CXCR7的缺失并未改变CXCR4和CXCL12的表达(图2G)。内皮剥脱损伤导致新生内膜增生。内皮CXCR7的缺失显著增加新生内膜区以及新生内膜与中膜的比率,但不改变中膜厚度(图2C-F)。内皮CXCR7缺失不改变这些血脂正常的小鼠的体重或血浆脂质。The mRNA expression of endothelial CXCR7 in endothelial cells isolated from cKO mice was essentially zero (Fig. 2A), which was further confirmed by immunostaining of damaged arteries (Fig. 2B). Deletion of CXCR7 did not alter the expression of CXCR4 and CXCL12 (Fig. 2G). Endothelial exfoliation damage leads to neointimal hyperplasia. Deletion of endothelial CXCR7 significantly increased the ratio of the neointimal zone and neointimal to medial membrane, but did not alter the medial thickness (Fig. 2C-F). Deletion of endothelial CXCR7 did not alter the body weight or plasma lipids of these normal blood lipid mice.
实施例3:CXCR7增加IL-1β处理的内皮细胞增殖并促进内皮剥脱损伤后的内皮再生Example 3: CXCR7 increases IL-1β-treated endothelial cell proliferation and promotes endothelial regeneration after endothelial exfoliation injury
内皮损伤后第7天检查早期血管变化。内皮CXCR7缺失小鼠表现出内皮再生受损(图3A和3B)。还观察到浸润单核细胞的减少(图3C和3D)。有趣的是,PDGF-BB的表达在内膜层(图3E和3F)和血浆中提高(图3G)。Early vascular changes were examined on day 7 after endothelial injury. Endothelial CXCR7 deficient mice exhibited impaired endothelial regeneration (Figures 3A and 3B). A decrease in infiltrating monocytes was also observed (Figures 3C and 3D). Interestingly, expression of PDGF-BB was elevated in the inner membrane layer (Figs. 3E and 3F) and plasma (Fig. 3G).
在分离自小鼠的培养的内皮细胞中,IL-1β刺激使CXCR7(图4A和4B)、CXCR4和CXCL12表达上调(图4I)。IL-1β促进CXCR7功能正常的内皮细胞的增殖,但并不促进CXCR7缺失细胞的增殖(图4C和4D)。此外,用CXCR7特异性拮抗剂CCX771(其具有约5.3nM的IC50,不影响CXCL12与CXCR4的结合)(Journal of immunology.2009;183:3204-3211)和对照化合物CCX704处理内皮细胞。CCX771抑制肺来源(图 4C)和主动脉来源(图4D)的内皮细胞的增殖。CCX771处理降低ERK磷酸化,但不影响JNK或p38磷酸化(图4E和4F)。这与CXCR7促进内皮细胞增殖的作用一致。在无IL-1β处理时,该促生长效果不明显(图4J)。另外,在HUVEC中,CCX771或siRNA敲低CXCR7均抑制IL-1β存在时的细胞增殖(图4G和4H)。使用TNFα时也观察到类似结果(图4K)。血管损伤伴随炎症因子的释放,而CXCR7可被炎症刺激诱导,并在促进炎症相关内皮细胞的增殖。因此,CXCR7可以促进受损血管的内皮再生,促进内皮细胞修复,减轻损伤导致的血管狭窄。In cultured endothelial cells isolated from mice, IL-1β stimulation up-regulated CXCR7 (Figs. 4A and 4B), CXCR4 and CXCL12 expression (Fig. 4I). IL-1β promotes proliferation of CXCR7-functioning endothelial cells but does not promote proliferation of CXCR7-deficient cells (Fig. 4C and 4D). Furthermore, endothelial cells were treated with the CXCR7-specific antagonist CCX771 (which has an IC50 of about 5.3 nM, which does not affect the binding of CXCL12 to CXCR4) (Journal of immunology. 2009; 183:3204-3211) and the control compound CCX704. CCX771 inhibits proliferation of endothelial cells from the lung source (Fig. 4C) and aortic source (Fig. 4D). CCX771 treatment reduced ERK phosphorylation but did not affect JNK or p38 phosphorylation (Figures 4E and 4F). This is consistent with the role of CXCR7 in promoting endothelial cell proliferation. This growth-promoting effect was not significant in the absence of IL-1β treatment (Fig. 4J). In addition, in HUVEC, CCX771 or siRNA knockdown CXCR7 inhibited cell proliferation in the presence of IL-1β (Figs. 4G and 4H). Similar results were observed when TNFα was used (Fig. 4K). Vascular injury is accompanied by the release of inflammatory factors, whereas CXCR7 is induced by inflammatory stimuli and promotes proliferation of inflammation-associated endothelial cells. Therefore, CXCR7 can promote endothelial regeneration of damaged blood vessels, promote endothelial cell repair, and reduce blood vessel stenosis caused by injury.
CXCR4 siRNA或AMD3100(一种CXCR4拮抗剂,其具有约44nM的IC50(Journal of immunology.2009;183:3204-3211),不影响CXCL2与CXCR7的结合(J Exp Med.2006;203:2201-2213)或与CXCR7的微弱结合,Ki约为34.5μM(Molecular pharmacology.2009;75:1240-1247))处理的内皮细胞的增殖无显著差异(图4L的C和D)。可见,抑制CXCR4不影响内皮细胞增殖。CXCR4 siRNA or AMD3100 (a CXCR4 antagonist with an IC50 of about 44 nM (Journal of immunology. 2009; 183:3204-3211) does not affect the binding of CXCL2 to CXCR7 (J Exp Med. 2006; 203: 2201-2213) Or with the weak binding of CXCR7, there was no significant difference in the proliferation of endothelial cells treated with Ki of about 34.5 μM (Molecular pharmacology. 2009; 75: 1240-1247) (C and D of Fig. 4L). It can be seen that inhibition of CXCR4 does not affect endothelial cell proliferation.
实施例4:内皮CXCR7对血管生成至关重要Example 4: Endothelial CXCR7 is essential for angiogenesis
小管形成用于检查CXCR7在内皮细胞新血管生成反应中的作用。通过siRNA阻断CXCR7显著抑制HUVEC和HAEC以及小鼠EC中的血管生成(图5A-D和图4L的A和B)。在后肢缺血小鼠模型中,内皮CXCR7缺失显著降低股动脉结扎后的血流恢复,这可通过激光多普勒成像检测(图5E-G)。对内皮的进一步组化染色显示缺血性腓肠肌中血管数目降低(图5H和5I)。通过小RNA(si-RNA)转染敲低CXCR7或CXCR4。在小鼠动脉内皮细胞(MAEC)中转染对照(si-Neg),si-CXCR7(si-CXCR7 1,si-CXCR7 2,和si-CXCR7 3)或si-CXCR4(si-CXCR4 1,si-CXCR4 2,si-CXCR4 3,和si-CXCR4 4)12小时后,确定CXCR7(A)和CXCR4(C)蛋白表达。A(CXCR4免疫印迹图片)和B示出CXCR7敲低对CXCR4表达无影响。*,p<0.05vs.si-Neg。Tubule formation was used to examine the role of CXCR7 in endothelial cell neovascularization responses. Blocking CXCR7 by siRNA significantly inhibited angiogenesis in HUVEC and HAEC as well as mouse EC (Figures AA-D and A and B of Figure 4L). In a mouse model of hindlimb ischemia, endothelial CXCR7 deletion significantly reduced blood flow recovery after femoral artery ligation, which was detected by laser Doppler imaging (Fig. 5E-G). Further histochemical staining of the endothelium showed a decrease in the number of blood vessels in the ischemic gastrocnemius muscle (Figures 5H and 5I). Knockdown of CXCR7 or CXCR4 by small RNA (si-RNA) transfection. Transfection control (si-Neg), si-CXCR7 (si-CXCR7 1, si-CXCR7 2, and si-CXCR7 3) or si-CXCR4 (si-CXCR4 1, si) in mouse arterial endothelial cells (MAEC) -CXCR4 2, si-CXCR4 3, and si-CXCR4 4) After 12 hours, CXCR7 (A) and CXCR4 (C) protein expression was determined. A (CXCR4 immunoblot picture) and B show that CXCR7 knockdown has no effect on CXCR4 expression. *, p < 0.05 vs. si-Neg.
因此,内皮细胞的CXCR7在促进缺血诱导的血管生成中起到关键作用,而之前,缺血诱导的血管生成被认为仅由CXCL12与CXCR4的相互作用介导(Trends Immunol.2007;28:299-307)。Thus, endothelial cell CXCR7 plays a key role in promoting ischemia-induced angiogenesis, whereas ischemia-induced angiogenesis was previously thought to be mediated only by the interaction of CXCL12 with CXCR4 (Trends Immunol. 2007; 28:299 -307).
实施例5:内皮CXCR7缺失损害MI后心脏功能并增加死亡率和梗死面积Example 5: Deletion of endothelial CXCR7 impairs cardiac function after MI and increases mortality and infarct size
通过结扎前降支研究了内皮CXCR7在MI中的潜在功能。令人惊讶的是,与对照小鼠(Ctl)相比,内皮CXCR7缺失(cKO)显著缩短存活时间,并降低MI后30天内的累积存活率(图6A)。cKO显示MI后显著破坏的心脏功能和重塑,包括降低的EF、 E/A以及LVAWd,尽管cKO的基线心脏功能没有变化(图6B-F,表2和3)。The potential function of endothelial CXCR7 in MI was studied by ligation of the anterior descending branch. Surprisingly, endothelial CXCR7 deletion (cKO) significantly shortened survival and reduced cumulative survival within 30 days after MI compared to control mice (Ctl) (Fig. 6A). cKO showed significant disruption of cardiac function and remodeling after MI, including decreased EF, E/A, and LVAWd, although there was no change in baseline cardiac function of cKO (Figure 6B-F, Tables 2 and 3).
表2.在MI后第7天对cKO和Ctl进行心脏超声Table 2. Cardiac ultrasound of cKO and Ctl on day 7 after MI
两组中n=12。n=12 in both groups.
数据以平均值±SEM表示。Data are expressed as mean ± SEM.
表3.对未经处理的cKO和Ctl进行心脏超声Table 3. Cardiac ultrasound for untreated cKO and Ctl
n=7Ctl,6cKO。n=7Ctl, 6cKO.
数据以平均值±SEM表示。Data are expressed as mean ± SEM.
梗死心脏的免疫荧光染色显示对照小鼠的内皮表达CXCR7,但在CXCR7 cKO 小鼠中并无表达(图6G)。Masson染色显示cKO小鼠心脏中梗死面积增加(图6H和J),与此同时梗死区域血管密度降低(图6I和K)。这表明血管生成受损可造成功能缺陷和增加的心肌纤维化。值得注意的是,尽管之前推测CXCL12在MI后的心脏保护中起作用(J Am Coll Cardiol.2011;58:2415-2423;J Exp Med.2006;203:2201-2213;Circulation research.2013;112:816-825),且本实验也证实内皮CXCR7缺失动物中CXCL12水平是升高(图7),但并未观察到其对MI后心脏功能的保护作用。Immunofluorescence staining of the infarcted heart showed that the endothelial cells of the control mice expressed CXCR7 but were not expressed in CXCR7 cKO mice (Fig. 6G). Masson staining showed an increase in infarct size in the hearts of cKO mice (Fig. 6H and J), while vascular density in the infarct area decreased (Fig. 6I and K). This suggests that impaired angiogenesis can cause functional deficits and increased myocardial fibrosis. It is worth noting that although CXCL12 was previously suggested to play a role in cardioprotection after MI (J Am Coll Cardiol. 2011; 58:2415-2423; J Exp Med. 2006; 203:2201-2213; Circulation research.2013;112 :816-825), and this experiment also confirmed that CXCL12 levels were elevated in endothelial CXCR7-deficient animals (Fig. 7), but its protective effect on cardiac function after MI was not observed.
实施例6:向梗死心脏中基因递送CXCR7改善MI后心脏功能并降低梗死面积Example 6: Gene delivery of CXCR7 to infarcted hearts improves cardiac function and reduces infarct size after MI
为了验证CXCR7在体内对心梗的保护作用,我们进一步构建了表达CXCR7的重组腺病毒(Ad-CXCR7),并在结扎前降支之前1分钟将病毒颗粒注入C57BL/6小鼠MI模型动物的左心室腔(详见材料和通用方法)。结果显示,与对照载体相比,Ad-CXCR7递送改善了MI后的心脏功能,降低了梗死面积(图8,表4)。In order to verify the protective effect of CXCR7 on myocardial infarction in vivo, we further constructed a recombinant adenovirus expressing CXCR7 (Ad-CXCR7), and injected the virus particles into C57BL/6 mouse MI model animals 1 minute before the branching before the ligation. Left ventricular cavity (see Materials and General Methods for details). The results showed that Ad-CXCR7 delivery improved cardiac function after MI and reduced infarct size compared to the control vehicle (Figure 8, Table 4).
表4.对转染了腺病毒载体(Ad-Neg)或表达CXCR7的腺病毒(Ad-CXCR7)的小鼠进行心脏超声。Table 4. Heart ultrasound was performed on mice transfected with adenovirus vector (Ad-Neg) or adenovirus expressing CXCR7 (Ad-CXCR7).
n=10Ad-Neg.11Ad-CXCR7。n=10Ad-Neg.11Ad-CXCR7.
数据以平均值±SEM表示。Data are expressed as mean ± SEM.
实施例7:CXCR7的活化促进内皮细胞增殖并减小梗死面积Example 7: Activation of CXCR7 promotes endothelial cell proliferation and reduces infarct size
以IL-1β刺激小鼠主动脉内皮细胞,并以CXCR7的选择激动剂TC14012(https://www.rndsystems.com/cn/products/tc-14012_4300)(Cayman Chemical,Michigan,USA)处理细胞48小时,然后使用细胞计数试剂盒-8(CCK-8;Yeasen,Shanghai,China) 测量细胞生长。结果显示,与对照(0ng/mL)相比,TC14012在100ng/mL显著促进小鼠主动脉内皮细胞的生长(p<0.05)(图9)。Mouse aortic endothelial cells were stimulated with IL-1β and treated with CXCR7 selective agonist TC14012 (https://www.rndsystems.com/cn/products/tc-14012_4300) (Cayman Chemical, Michigan, USA) Hours were then measured for cell growth using Cell Counting Kit-8 (CCK-8; Yeasen, Shanghai, China). The results showed that TC14012 significantly promoted the growth of mouse aortic endothelial cells at 100 ng/mL compared to the control (0 ng/mL) (p<0.05) (Fig. 9).
通过结扎前降支研究了CXCR7的选择激动剂TC14012对心肌梗死的影响。结扎野生型C57BL/6小鼠冠状动脉前降支造成缺血,随后给小鼠腹腔注射TC14012,剂量为10mg/kg,溶于生理盐水,每6天注射一次,共4次,持续24天。结果显示,与对照(生理盐水)相比,TC14012显著缩小了前降支结扎造成的心肌梗死面积(图10)。The effect of CXCR7, the selective agonist TC14012, on myocardial infarction was studied by ligation of the anterior descending branch. The wild-type C57BL/6 mice were ligated to the left anterior descending coronary artery to cause ischemia. Subsequently, the mice were intraperitoneally injected with TC14012 at a dose of 10 mg/kg, dissolved in normal saline, and injected once every 6 days for 4 times for 24 days. The results showed that TC14012 significantly reduced the area of myocardial infarction caused by ligation of the anterior descending branch compared to the control (saline) (Figure 10).
上述结果提示,采用CXCR7的选择激动剂例如TC14012活化CXCR7能够减小缺血造成的心肌梗死的范围,对缺血心肌具有保护作用,其原因与CXCR7的活化直接促进梗死部位心肌组织中血管内皮细胞的增殖和血管形成相关联,这有利于MI后的心肌重塑和心脏功能的恢复。These results suggest that the use of CXCR7 selective agonist such as TC14012 to activate CXCR7 can reduce the range of myocardial infarction caused by ischemia, and has a protective effect on ischemic myocardium. The reason and activation of CXCR7 directly promote vascular endothelial cells in myocardial tissue of infarcted area. Proliferation is associated with angiogenesis, which facilitates myocardial remodeling and recovery of cardiac function after MI.
实施例8:内皮CXCR7缺失以及药理学阻断CXCR7升高循环中CXCL12的水平Example 8: Deletion of endothelial CXCR7 and pharmacological blockade of CXCR7 raises the level of CXCL12 in the circulation
为了确定CXCR7在血栓形成中的作用,构建了CXCR7条件性敲除小鼠(cKO),并按照文献所述(The Journal of clinical investigation.2006;116:1391-1399)对小鼠进行光化学诱导的血栓形成评估。与同窝出生的对照小鼠(Ctl)相比,cKO小鼠形成完全阻塞的时间显著缩短(图11A&B)。内皮细胞缺失CXCR7导致CXCL12在循环中的水平升高(图11E),这与CXCR7的配体清道夫的功能相一致(PLoS One.2010;5:e9175;Blood.2012;119:465-468)。类似地,以CXCR7特异性拮抗剂CCX771(Journal of immunology.2009;183:3204-3211)药理学抑制CXCR7也促进了血栓的形成(图11C&D)并使得血浆CXCL12升高(图11F)。而另一种可以被CXCR7清除的配体——CXCL11(PLoS One.2010;5:e9175)——在所有这些小鼠均未检测到。可见内皮细胞CXCR7的表达水平或活性降低导致循环中CXCL12的水平升高并加速血栓形成应答。To determine the role of CXCR7 in thrombosis, CXCR7 conditional knockout mice (cKO) were constructed and photochemically induced in mice as described in the literature (The Journal of clinical investigation. 2006; 116: 1391-1399). Thrombosis assessment. The time to complete occlusion of cKO mice was significantly shorter compared to control mice born in littermates (Ctl) (Fig. 11A & B). Deletion of CXCR7 by endothelial cells leads to an increase in the level of CXCL12 in the circulation (Fig. 11E), which is consistent with the function of the ligand scavenger of CXCR7 (PLoS One. 2010; 5: e9175; Blood. 2012; 119: 465-468) . Similarly, pharmacological inhibition of CXCR7 with the CXCR7-specific antagonist CCX771 (Journal of immunology. 2009; 183:3204-3211) also promoted thrombus formation (Fig. 11C&D) and elevated plasma CXCL12 (Fig. 11F). Another ligand that can be cleared by CXCR7, CXCL11 (PLoS One. 2010; 5: e9175), was not detected in all of these mice. It can be seen that a decrease in the expression level or activity of endothelial cell CXCR7 leads to an increase in the level of CXCL12 in the circulation and accelerates the thrombotic response.
实施例9:循环中CXCL12水平升高通过CXCL12-CXCR4信号途径促进血栓形成Example 9: Elevated levels of CXCL12 in the circulation promote thrombosis through the CXCL12-CXCR4 signaling pathway
在体外,CXCL12通过其同源受体CXCR4促进血小板活化。为了直接评估循环中的CXCL12在体内对血栓形成的影响,给小鼠建立静脉通路以直接将CXCL12输注(以0.25或0.5ng/min输注溶于生理盐水的CXCL12或输注生理盐水)至血流中(图12A)。通过测量CXCL12的血浆浓度,证实CXCL12可被成功递送(图12B)。输注后循环中CXCL12的中度升高以浓度依赖性方式促进离体血小板聚集(图12C&D)。CXCL12输注还在体内促使血栓形成加速(图12E&F)。通过使用CXCR4特异性拮抗 剂AMD3100进行预处理,可消除CXCL12的这种促血栓形成作用(图12E&F)。此外,AMD3100处理也消除了内皮细胞CXCR7敲除所造成的促血栓形成作用(图13A&B)。因此,通过CXCR4信号途径,循环中的CXCL12生理学意义上的升高造成血栓形成倾向。In vitro, CXCL12 promotes platelet activation via its cognate receptor CXCR4. To directly assess the effect of circulating CXCL12 on thrombosis in vivo, a venous access was established in mice to directly infuse CXCL12 (infusion of CXCL12 or physiological saline in saline at 0.25 or 0.5 ng/min) In the bloodstream (Figure 12A). CXCL12 was confirmed to be successfully delivered by measuring the plasma concentration of CXCL12 (Fig. 12B). Moderate elevation of CXCL12 in the post-infusion cycle promoted ex vivo platelet aggregation in a concentration dependent manner (Figure 12C&D). The CXCL12 infusion also accelerated thrombus formation in vivo (Figure 12E&F). This prothrombotic effect of CXCL12 was abolished by pretreatment with the CXCR4-specific antagonist AMD3100 (Fig. 12E & F). In addition, AMD3100 treatment also abolished the prothrombotic effect caused by endothelial cell CXCR7 knockout (Fig. 13A & B). Thus, through the CXCR4 signaling pathway, a physiologically elevated CXCL12 in the circulation causes a tendency to thrombosis.
实施例10:血小板促成并应答于内皮细胞CXCR7缺失之后的CXCL12升高Example 10: Platelet promotes and responds to elevated CXCL12 following endothelial cell CXCR7 deletion
血小板含有CXCL12且在活化后释放CXCL12(The Journal of experimental medicine.2006;203:1221-1233),但尚不清楚在体内血小板是否是CXCL12的来源以及在cKO中血小板是否促成CXCL12的升高。Platelets contain CXCL12 and release CXCL12 upon activation (The Journal of experimental medicine. 2006; 203: 1221-1233), but it is unclear whether platelets in vivo are the source of CXCL12 and whether platelets contribute to the elevation of CXCL12 in cKO.
首先,以血小板激动剂,即U46619或胶原,处理离体全血触发了血小板释放(图14A&B)。静脉注射血小板激活剂U46619使得血流中的血小板迅速聚集(图14C),且CXCL12水平大约成倍升高,这提示活化的血小板是体内循环CXCL12的重要来源(图14D)。First, treatment of ex vivo whole blood with a platelet agonist, U46619 or collagen, triggered platelet release (Fig. 14A & B). Intravenous platelet activating agent U46619 caused rapid aggregation of platelets in the bloodstream (Fig. 14C), and the CXCL12 level was approximately multiplied, suggesting that activated platelets are an important source of circulating CXCL12 in vivo (Fig. 14D).
随后,通过给小鼠注射抗CD41抗体耗竭血小板,该处理导致注射后2小时血小板基本上耗竭,第5天才恢复(图14I)。血小板耗竭在cKO和Ctl中均使得循环中的CXCL12下降,但两者相比,cKO小鼠维持了更高的CXCL12水平(图14G)。因此,循环中的CXCL12也包括来自除血小板之外的其他来源的CXCL12,且循环中的CXCL12水平受内皮细胞CXCR7的调节。值得注意的是,相比于Ctl,血小板在cKO中对循环中的CXCL12的贡献更大一些(图14H)。通过流式细胞术测定血小板的p-选择素(CD62P)的表达(Circulation.2015;132:47-58),相比于Ctl,发现cKO有更多CD62P阳性血小板。这提示,在cKO中,血小板活化导致循环CXCL12水平升高(图14H)。Subsequently, platelets were depleted by injection of anti-CD41 antibodies to mice, which resulted in substantial depletion of platelets 2 hours after injection and recovered on day 5 (Fig. 14I). Platelet depletion resulted in a decrease in circulating CXCL12 in both cKO and Ctl, but cKO mice maintained higher CXCL12 levels compared to both (Fig. 14G). Thus, CXCL12 in the circulation also includes CXCL12 from other sources than platelets, and circulating CXCL12 levels are regulated by endothelial cell CXCR7. It is worth noting that platelets contribute more to circulating CXCL12 in cKO than Ctl (Figure 14H). The expression of p-selectin (CD62P) in platelets was determined by flow cytometry (Circulation. 2015; 132: 47-58), and cKO was found to have more CD62P-positive platelets than Ctl. This suggests that in cKO, platelet activation leads to elevated levels of circulating CXCL12 (Fig. 14H).
实施例11:人血液循环中的CXCL12水平与血小板反应性的关联性Example 11: Correlation between CXCL12 levels and platelet reactivity in human blood circulation
发明人进一步在急性心肌梗死患者中研究了循环的CXCL12与血小板反应性的相关性。收集来自连续入选患者的外周静脉血,用于测量血浆CXCL12,并采用VerifyNow方法现场测量血小板反应性(详见材料和通用方法部分)。出乎预料的是,CXCL12水平与ADP诱导的血小板反应性正相关(图15A),这支持循环中的CXCL12对血小板活化具有直接的作用。CXCL12与VerifyNow-AA不相关(图15B),其原因或许与这些患者服用阿司匹林有关(表1),因为通过CXCL12触发的TxB
2释放在体外介导了血小板聚集(Blood.2000;96:50-57)。
The inventors further investigated the association of circulating CXCL12 with platelet reactivity in patients with acute myocardial infarction. Peripheral venous blood from consecutively enrolled patients was collected for measurement of plasma CXCL12 and platelet reactivity was measured in situ using the VerifyNow method (see Materials and General Methods section for details). Unexpectedly, CXCL12 levels were positively correlated with ADP-induced platelet reactivity (Fig. 15A), which supports a direct effect of CXCL12 in the circulation on platelet activation. CXCL12 is not associated with VerifyNow-AA (Fig. 15B), which may be related to the use of aspirin in these patients (Table 1), as TxB 2 release triggered by CXCL12 mediates platelet aggregation in vitro (Blood. 2000; 96:50- 57).
Claims (27)
- 增加CXCR7蛋白的表达和/或活性的第一药物在制备用于治疗或预防对象的心血管疾病的药物组合物中的用途。Use of a first medicament for increasing the expression and/or activity of a CXCR7 protein for the preparation of a pharmaceutical composition for treating or preventing a cardiovascular disease in a subject.
- 权利要求1的用途,其中增加CXCR7蛋白的表达和/或活性的第一药物选自CXCR7的选择性激动剂或包含编码CXCR7蛋白或其功能片段的多核苷酸的表达载体。The use of claim 1, wherein the first drug that increases the expression and/or activity of the CXCR7 protein is selected from a selective agonist of CXCR7 or an expression vector comprising a polynucleotide encoding a CXCR7 protein or a functional fragment thereof.
- 权利要求2的用途,其中所述CXCR7的选择性激动剂选自CXCR7的活化型抗体、CXCR7的活化型配体、TC14012或其功能类似物、及其组合。The use of claim 2, wherein the selective agonist of CXCR7 is selected from the group consisting of an activated antibody of CXCR7, an activated ligand of CXCR7, TC14012 or a functional analog thereof, and combinations thereof.
- 权利要求1-3任一项的用途,其中所述药物组合物还包含有效量的CXCR4的选择性拮抗剂或抑制CXCR4蛋白表达的核酸分子或表达载体。The use according to any one of claims 1 to 3, wherein the pharmaceutical composition further comprises an effective amount of a selective antagonist of CXCR4 or a nucleic acid molecule or expression vector which inhibits expression of CXCR4 protein.
- 权利要求1-3任一项的用途,其中药物组合物与有效量的包含CXCR4的选择性拮抗剂或抑制CXCR4蛋白表达的核酸分子或表达载体的第二药物联合使用。The use according to any one of claims 1 to 3, wherein the pharmaceutical composition is used in combination with an effective amount of a selective antagonist comprising CXCR4 or a nucleic acid molecule or expression vector for inhibiting expression of CXCR4 protein.
- 权利要求4或5的用途,其中所述CXCR4的选择性拮抗剂选自AMD3100或其功能类似物、CXCR4的阻断性抗体或其抗原结合片段、TC14012或其功能类似物、及其组合。The use according to claim 4 or 5, wherein the selective antagonist of CXCR4 is selected from the group consisting of AMD3100 or a functional analog thereof, a blocking antibody of CXCR4 or an antigen-binding fragment thereof, TC14012 or a functional analogue thereof, and combinations thereof.
- 权利要求4或5的用途,其中所述抑制CXCR4蛋白表达的核酸分子选自靶向CXCR4基因转录产物的siRNA或其前体和靶向CXCR4基因转录产物的反义RNA。The use according to claim 4 or 5, wherein the nucleic acid molecule which inhibits expression of the CXCR4 protein is selected from the group consisting of an siRNA targeting a CXCR4 gene transcription product or a precursor thereof and an antisense RNA targeting a CXCR4 gene transcription product.
- 权利要求1-7任一项的用途,其中所述药物组合物用于与选自抑制血小板活化和/或聚集的药物和稳定斑块的药物联合使用。The use according to any one of claims 1 to 7, wherein the pharmaceutical composition is for use in combination with a drug selected from the group consisting of a drug that inhibits platelet activation and/or aggregation and a stable plaque.
- 权利要求1-8任一项的用途,其中所述药物组合物用于通过口服、含服、吸入、静脉注射、动脉注射、肌肉注射、皮下注射、腹腔注射或局部施用的方式给对象施用。The use according to any one of claims 1 to 8, wherein the pharmaceutical composition is for administration to a subject by oral, buccal, inhalation, intravenous, intraarterial, intramuscular, subcutaneous, intraperitoneal or topical administration.
- 权利要求9的用途,其中所述所述药物组合物用于冠脉内施用。The use of claim 9, wherein the pharmaceutical composition is for intracoronary administration.
- 权利要求1-10任一项的用途,其中所述疾病选自以下一组:血栓形成、血栓栓塞、血管壁损伤、损伤后血管狭窄、PCI和Bypass术后血管再狭窄、冠心病、心肌缺血、心肌梗死、心梗后心力衰竭、心梗后心律失常及其任意组合。The use according to any one of claims 1 to 10, wherein the disease is selected from the group consisting of thrombosis, thromboembolism, vascular wall injury, vascular stenosis after injury, vascular restenosis after PCI and Bypass, coronary heart disease, myocardial deficiency Blood, myocardial infarction, heart failure after myocardial infarction, arrhythmia after myocardial infarction, and any combination thereof.
- 权利要求1-11的用途,其中所述药物组合物用于治疗对象的血管内皮损伤性或改善对象的心肌梗死后的心脏重塑。Use according to claims 1-11, wherein the pharmaceutical composition is for treating vascular endothelial damage in a subject or improving cardiac remodeling after myocardial infarction in a subject.
- 血管支架或带有球囊的导管,其中所述支架或球囊的表面涂覆有有效量的用于增加CXCR7蛋白的表达和/或活性的第一药物。A vascular stent or catheter with a balloon, wherein the surface of the stent or balloon is coated with an effective amount of a first drug for increasing the expression and/or activity of the CXCR7 protein.
- 权利要求13的血管支架或带有球囊的导管,其中所述第一药物选自CXCR7的选择性激动剂、包含编码CXCR7蛋白或其功能片段的多核苷酸的表达载体及其组合。The vascular stent or balloon catheter of claim 13 wherein said first drug is selected from the group consisting of a selective agonist of CXCR7, an expression vector comprising a polynucleotide encoding a CXCR7 protein or a functional fragment thereof, and combinations thereof.
- 权利要求14的血管支架或带有球囊的导管,其中所述CXCR7的选择性激动剂选自CXCR7的活化型抗体、CXCR7的活化型配体、TC14012或其功能类似物、及其组合。The vascular stent or balloon having a balloon of claim 14, wherein the selective agonist of CXCR7 is selected from the group consisting of an activated antibody of CXCR7, an activated ligand of CXCR7, TC14012 or a functional analog thereof, and combinations thereof.
- 权利要求13-15任一项的血管支架或带有球囊的导管,其中所述支架或球囊的表面还涂覆有有效量的CXCR4的选择性拮抗剂或抑制CXCR4蛋白表达的核酸分子或表达载体。The vascular stent or balloon with a balloon according to any one of claims 13 to 15, wherein the surface of the stent or balloon is further coated with an effective amount of a selective antagonist of CXCR4 or a nucleic acid molecule that inhibits CXCR4 protein expression or Expression vector.
- 权利要求16的血管支架或带有球囊的导管,其中所述CXCR4的选择性拮抗剂选自AMD3100或其功能类似物、CXCR4的阻断性抗体或其抗原结合片段、及其组合。The vascular stent or balloon having a balloon according to claim 16, wherein the selective antagonist of CXCR4 is selected from the group consisting of AMD3100 or a functional analog thereof, a blocking antibody of CXCR4 or an antigen-binding fragment thereof, and combinations thereof.
- 权利要求16的血管支架或带有球囊的导管,其中所述抑制CXCR4蛋白表达的核酸分子选自靶向CXCR4基因转录产物的siRNA或其前体或靶向CXCR4基因转录产物的反义RNA。The vascular stent or balloon having a balloon according to claim 16, wherein the nucleic acid molecule which inhibits expression of the CXCR4 protein is selected from the group consisting of an siRNA targeting a CXCR4 gene transcription product or a precursor thereof or an antisense RNA targeting a CXCR4 gene transcription product.
- 降低循环中的CXCL12的水平或活性的药物、或CXCR4的选择性拮抗剂、或抑制CXCR4蛋白表达的核酸分子或表达载体、或其组合在制备用于治疗或预防对象的血栓形成相关疾病的药物组合物中的用途。A drug for lowering the level or activity of CXCL12 in circulation, or a selective antagonist of CXCR4, or a nucleic acid molecule or expression vector for inhibiting expression of CXCR4 protein, or a combination thereof, for preparing a drug for treating or preventing thrombosis-related diseases in a subject Use in the composition.
- 权利要求19的用途,其中所述降低循环中的CXCL12的水平或活性的药物选自增加CXCR7蛋白的表达和/或活性的药物和抗CXCL12抗体。The use according to claim 19, wherein the drug which lowers the level or activity of CXCL12 in the circulation is selected from the group consisting of a drug which increases the expression and/or activity of the CXCR7 protein and an anti-CXCL12 antibody.
- 权利要求20的用途,其中所述增加CXCR7蛋白的表达和/或活性的药物是包含编码CXCR7蛋白或其功能片段的多核苷酸的表达载体。The use according to claim 20, wherein the drug which increases the expression and/or activity of the CXCR7 protein is an expression vector comprising a polynucleotide encoding a CXCR7 protein or a functional fragment thereof.
- 权利要求19的用途,其中所述CXCR4的选择性拮抗剂选自AMD3100或其功能类似物、CXCR4的阻断性抗体或其抗原结合片段、及其组合。The use of claim 19, wherein the selective antagonist of CXCR4 is selected from the group consisting of AMD3100 or a functional analog thereof, a blocking antibody of CXCR4 or an antigen-binding fragment thereof, and combinations thereof.
- 权利要求19的用途,其中所述抑制CXCR4蛋白表达的核酸分子选自靶向CXCR4基因转录产物的siRNA或其前体和靶向CXCR4基因转录产物的反义RNA。The use of claim 19, wherein the nucleic acid molecule that inhibits expression of a CXCR4 protein is selected from the group consisting of an siRNA or a precursor thereof that targets a CXCR4 gene transcription product and an antisense RNA that targets a CXCR4 gene transcription product.
- 用于治疗癌症的药物组合物,其包含CXCR7抑制剂,其中所述CXCR7抑制剂在施用至对象体内时不增加血液CXCL12水平。A pharmaceutical composition for treating cancer comprising a CXCR7 inhibitor, wherein the CXCR7 inhibitor does not increase blood CXCL12 levels when administered to a subject.
- 筛选具有高心血管安全性的用于治疗癌症的药物的方法,包括:Methods for screening for drugs for treating cancer with high cardiovascular safety, including:(i)给动物施用候选药物,(i) administering a drug candidate to the animal,(ii)测定来自所述动物的组织样品中的CXCR7蛋白的活性,和(ii) determining the activity of the CXCR7 protein in a tissue sample from the animal, and(iii)测定来自所述动物的血液样品中的CXCL12蛋白的水平,(iii) determining the level of CXCL12 protein in a blood sample from the animal,其中所述动物的组织样品中CXCR7蛋白的活性相对于未施用所述候选药物的对照动物的相同组织样品中CXCR7蛋白的活性的降低,且所述动物的血液样品中CXCL12蛋白的含量与未施用所述候选药物的对照动物的血液样品中CXCL12蛋白的水平相当或更低,提示所述候选药物是具有高心血管安全性的治疗癌症的药物。Wherein the activity of the CXCR7 protein in the tissue sample of the animal is decreased relative to the activity of the CXCR7 protein in the same tissue sample of the control animal to which the drug candidate is not administered, and the content of CXCL12 protein in the blood sample of the animal is not administered. The level of CXCL12 protein in the blood sample of the control animal of the drug candidate is comparable or lower, suggesting that the drug candidate is a drug for treating cancer with high cardiovascular safety.
- 筛选能够用于治疗或预防心血管疾病的药物的方法,包括:Methods of screening for drugs that can be used to treat or prevent cardiovascular disease, including:(i)给动物施用候选药物,和(i) administering a drug candidate to the animal, and(ii)测定来自所述动物的组织样品中的CXCR7蛋白的表达水平和/或活性,(ii) determining the expression level and/or activity of the CXCR7 protein in a tissue sample from the animal,其中所述动物的组织样品中CXCR7蛋白的表达水平和/或活性相对于未施用所述候选药物的对照动物的相同组织样品中CXCR7蛋白的表达水平和/或活性的升高表明所述候选药物能够治疗或预防心血管疾病。Wherein the expression level and/or activity of the CXCR7 protein in the tissue sample of the animal is increased relative to the expression level and/or activity of the CXCR7 protein in the same tissue sample of the control animal to which the drug candidate is not administered, indicating the candidate drug Can treat or prevent cardiovascular disease.
- 权利要求26的方法,其中所述CXCR7蛋白的活性选自促进血管内皮增殖、促进血管新生、促进损伤血管修复、减小心肌梗死面积、改善梗死后心肌重塑、改善梗死后的心脏功能。The method of claim 26, wherein the activity of said CXCR7 protein is selected from the group consisting of promoting vascular endothelial proliferation, promoting angiogenesis, promoting damaged vascular repair, reducing myocardial infarct size, improving myocardial remodeling after infarction, and improving cardiac function after infarction.
Priority Applications (1)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
CN201880008458.8A CN110225768A (en) | 2017-01-26 | 2018-01-26 | Pharmaceutical compositions and methods targeting CXCR7 |
Applications Claiming Priority (2)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
CN201710061692.4 | 2017-01-26 | ||
CN201710061692.4A CN108355133A (en) | 2017-01-26 | 2017-01-26 | Target the pharmaceutical composition and method of CXCR7 |
Publications (1)
Publication Number | Publication Date |
---|---|
WO2018137701A1 true WO2018137701A1 (en) | 2018-08-02 |
Family
ID=62979005
Family Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
PCT/CN2018/074264 WO2018137701A1 (en) | 2017-01-26 | 2018-01-26 | Pharmaceutical composition targeting cxcr7 and method |
Country Status (2)
Country | Link |
---|---|
CN (2) | CN108355133A (en) |
WO (1) | WO2018137701A1 (en) |
Cited By (1)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
WO2024243361A1 (en) * | 2023-05-23 | 2024-11-28 | The Regents Of The University Of California | Compositions and methods to treat muscle injury and disease |
Families Citing this family (1)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
CA3136396A1 (en) * | 2019-05-03 | 2020-11-12 | Luxembourg Institute Of Health (Lih) | Novel selective ackr3 modulators and uses thereof |
Citations (4)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
WO2009120186A1 (en) * | 2008-03-24 | 2009-10-01 | Carolus Therapeutics, Inc. | Methods and compositions for treating atherosclerosis and related conditions |
CN102088993A (en) * | 2008-03-20 | 2011-06-08 | 卡罗勒斯治疗公司 | Methods of treating inflammation |
WO2012047339A2 (en) * | 2010-06-28 | 2012-04-12 | The General Hospital Corporation | Anti-cxcr4 as a sensitizer to cancer therapeutics |
CN106065401A (en) * | 2015-04-20 | 2016-11-02 | 成都医学院 | Lentivirus mediated CXCR7 high expressed through engineering approaches endothelial progenitor cells treatment use in ischemic diseases |
Family Cites Families (1)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
CN103397052A (en) * | 2013-08-15 | 2013-11-20 | 温州医科大学 | Endothelial progenitor cell with highly-expressed human CXCR7 (C-X-C chemokine receptor 7) gene as well as preparation method and application of endothelial progenitor cell |
-
2017
- 2017-01-26 CN CN201710061692.4A patent/CN108355133A/en active Pending
-
2018
- 2018-01-26 CN CN201880008458.8A patent/CN110225768A/en active Pending
- 2018-01-26 WO PCT/CN2018/074264 patent/WO2018137701A1/en active Application Filing
Patent Citations (4)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
CN102088993A (en) * | 2008-03-20 | 2011-06-08 | 卡罗勒斯治疗公司 | Methods of treating inflammation |
WO2009120186A1 (en) * | 2008-03-24 | 2009-10-01 | Carolus Therapeutics, Inc. | Methods and compositions for treating atherosclerosis and related conditions |
WO2012047339A2 (en) * | 2010-06-28 | 2012-04-12 | The General Hospital Corporation | Anti-cxcr4 as a sensitizer to cancer therapeutics |
CN106065401A (en) * | 2015-04-20 | 2016-11-02 | 成都医学院 | Lentivirus mediated CXCR7 high expressed through engineering approaches endothelial progenitor cells treatment use in ischemic diseases |
Non-Patent Citations (1)
Title |
---|
TU, XIAOLI ET AL.: "Relevance of the Chemokine CXCL12/CXCR7 Axis in Cardiovascular Disease", JOURNAL OF CHONGQING MEDICAL UNIVERSITY, vol. 40, no. 4, 20 March 2015 (2015-03-20), pages 483 * |
Cited By (1)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
WO2024243361A1 (en) * | 2023-05-23 | 2024-11-28 | The Regents Of The University Of California | Compositions and methods to treat muscle injury and disease |
Also Published As
Publication number | Publication date |
---|---|
CN108355133A (en) | 2018-08-03 |
CN110225768A (en) | 2019-09-10 |
Similar Documents
Publication | Publication Date | Title |
---|---|---|
JP2008291040A (en) | Nicotine receptor agonists in the recruitment of stem and progenitor cells | |
JP2007084570A (en) | Prevention and treatment of pathogeny associated with abnormally proliferative smooth muscle cell | |
US7045534B2 (en) | Methods of reducing angiogenesis | |
Mohammadipoor et al. | Stanniocalcin-1 attenuates ischemic cardiac injury and response of differentiating monocytes/macrophages to inflammatory stimuli | |
Frommhold et al. | Protein C concentrate controls leukocyte recruitment during inflammation and improves survival during endotoxemia after efficient in vivo activation | |
US8828724B2 (en) | Method for preparing cells for engraftment | |
WO2018137701A1 (en) | Pharmaceutical composition targeting cxcr7 and method | |
US9084761B2 (en) | Use of interleukin-15 to treat cardiovascular diseases | |
US20160193290A1 (en) | Methods of Reducing Myocardial Injury Following Myocardial Infarction | |
Zhang et al. | The effects of parathyroid hormone‐related peptide on cardiac angiogenesis, apoptosis, and function in mice with myocardial infarction | |
TWI823085B (en) | neutrophil activation modulator | |
WO2020010958A1 (en) | Application of metrnl protein or gene in blocked blood vessel disease | |
US20210093599A1 (en) | N-acetylcysteine attenuates aortic stenosis progression by inhibiting shear-mediated tgf-beta activation and signaling | |
US10314862B2 (en) | Hypoxia-cultured mesenchymal stem cells for treating atherosclerotic lesions | |
WO2018196870A1 (en) | Drug targeting prostaglandin e2 and receptor thereof and application | |
P Sarapultsev et al. | Effect of a new class of compounds of the group of substituted 5R1, 6H2-1, 3, 4-thiadiazine-2-amines on the inflammatory and cytokine response in experimental myocardial infarction | |
JP7246094B2 (en) | activity regulator | |
RU2752089C1 (en) | Application of the compound for the preparation of a drug for the treatment of cerebral microangiopathy | |
US8343916B2 (en) | Use of heat-shock protein 27 for cardiovascular disease prevention and treatment | |
TWI876411B (en) | Use for treating a peripheral arterial disease | |
RU2817822C1 (en) | Method for creating translational model of diabetic phenotype of chronic heart failure | |
US20050191276A1 (en) | Treatment of inflammatory bowel disease through induction of indoleamine 2.3-dioxygenase | |
CN108685896B (en) | Use of Melaleuca A in the preparation of medicine for treating and/or preventing chronic peripheral vascular occlusive disease | |
Cremers | Cytoprotective mechanisms, palatogenesis, and wound repair | |
Wu | Heterodimer of erythropoietin receptor and β common receptor in renal ischemia-reperfusion injury and repair |
Legal Events
Date | Code | Title | Description |
---|---|---|---|
121 | Ep: the epo has been informed by wipo that ep was designated in this application |
Ref document number: 18744284 Country of ref document: EP Kind code of ref document: A1 |
|
NENP | Non-entry into the national phase |
Ref country code: DE |
|
122 | Ep: pct application non-entry in european phase |
Ref document number: 18744284 Country of ref document: EP Kind code of ref document: A1 |