CN104800243B - Application of recombinant mesenchymal stem cells in preparation of immunosuppressant - Google Patents

Application of recombinant mesenchymal stem cells in preparation of immunosuppressant Download PDF

Info

Publication number
CN104800243B
CN104800243B CN201410188453.1A CN201410188453A CN104800243B CN 104800243 B CN104800243 B CN 104800243B CN 201410188453 A CN201410188453 A CN 201410188453A CN 104800243 B CN104800243 B CN 104800243B
Authority
CN
China
Prior art keywords
mesenchymal stem
ccr7
mscs
egfp
cell
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Active
Application number
CN201410188453.1A
Other languages
Chinese (zh)
Other versions
CN104800243A (en
Inventor
李红
郭希民
毛宁
江小霞
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Institute of Basic Medical Sciences of AMMS
Original Assignee
Institute of Basic Medical Sciences of AMMS
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Institute of Basic Medical Sciences of AMMS filed Critical Institute of Basic Medical Sciences of AMMS
Priority to CN201410188453.1A priority Critical patent/CN104800243B/en
Publication of CN104800243A publication Critical patent/CN104800243A/en
Application granted granted Critical
Publication of CN104800243B publication Critical patent/CN104800243B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Images

Landscapes

  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)

Abstract

The invention discloses an application of a recombinant mesenchymal stem cell in preparation of an immunosuppressant. The recombinant mesenchymal stem cells can be subjected to targeted migration in vivo to a secondary lymphoid organ after being infused and can be massively accumulated in a T cell enrichment area in the secondary lymphoid organ, the immunoreaction of T cells can be effectively inhibited, and the cost, the cell dosage and the side effect can be reduced in the treatment of graft-versus-host disease after bone marrow transplantation, immune rejection reaction after organ transplantation and autoimmune diseases. More particularly, the effect of killing tumor cells of the organism is kept in the treatment, and a high-efficiency and beneficial new strategy is provided for clinically treating graft-versus-host disease after bone marrow transplantation, immune rejection after organ transplantation and autoimmune disease of high-risk tumor people.

Description

Application of recombinant mesenchymal stem cells in preparation of immunosuppressant
Technical Field
The invention relates to the field of stem cell therapy, in particular to application of a mesenchymal stem cell of which the cell membrane expresses a chemokine receptor CCR7 in preparation of an immunosuppressant.
Background
Mesenchymal stem cells are a population of adult stem cells with self-renewal and multipotent differentiation potential that were first isolated from bone marrow. Recent studies have shown that: mesenchymal stem cells have a wide range of immunomodulatory functions. The mesenchymal stem cells have the immunoregulation function on various immune cells including T cells, B cells, NK cells, DCs and the like. In effective animal experiments and clinical disease treatment, mesenchymal stem cells are found to play a role in vivo immunoregulation mainly through secretory cytokines.
Currently, the immunotherapy of clinical diseases such as hematopoietic stem cell transplantation or graft-versus-host disease, immune rejection after solid organ transplantation, crohn's disease, systemic lupus erythematosus and the like by mesenchymal stem cells has entered the clinical phase II-III in the united states. 5/17/2012, the health regulatory agency in Canada announced approval of a human mesenchymal stem cell drug, procymmalTMMarketed under the condition of allowing ProchymalTMBecomes the first stem cell medicine which is really approved to be used for treating the systemic diseases in the world, and marks that the door of the clinical application of the mesenchymal stem cells as the immunomodulator is opened. The problems still faced are: 1) phase III clinical results in multicenter indicate: the in vivo immunoregulation effect is not obvious; 2) large cell usage [ Tyndall a. vaccines and failures of stem cell transfer in autoimmune diseases. hematology Am Soc Hematol Educ program.2011; 2011: 280-284.][Martin I,Baldomero H,Tyndall A,Niederwieser D,Gratwohl A.A survey on cell and engineered tissue therapies in Europe in2008.Tissue Eng Part A2010;16:2419-2427.]
Many clinical treatment modalities of mesenchymal stem cells include in situ injection and intravenous injection [ http: // www.clinicaltrials.gov `]. However, intravenous injection is simple and easy, and systemic diseases such as graft-versus-host disease, Crohn's disease, systemic lupus erythematosus and the like are mainly intravenous injection. Therefore, a large amount of cells are needed for clinically treating the diseases, for example, the clinical mesenchymal stem cell immunotherapy dosage is 1-9 x 106MSCs/Kg, 2 times per week for four weeks, clearly increases the cost of treatment, and may also present risks such as embolism, tumor recurrence, etc. Thus, mesenchymal stem cells are efficientIn vivo migration is the key to achieving the two most efficient and economical goals of Cell therapy [ Karp JM and Leng Teo gs.mesenchyme Stem Cell hosting: the Devil Is in The details cell Stem cell.2009; 4(3): 206-216.Ankrum J, Karp JM.Mesenchyl stem cell therapy: two steps forward, one step back. trends Mol med.2010; 16(5): 203-9]。
Secondary lymphoid organs including spleen white marrow (SPW), Lymph Nodes (LN), Mesenteric Lymph Nodes (MLN), PP nodes, etc., are simple and similar in structure, and are composed of T, B lymphocytes, Antigen Presenting Cells (APC), stromal cells, and microtubule junctions. T cells receive antigen presented by APC in secondary lymphoid organs, and proliferate by activation, and thus, secondary lymphoid organs are the sites where immune responses are initiated in vivo. After intravenous infusion, mesenchymal stem cells are widely distributed in the whole body, such as lung, intestine, liver, bone marrow, and the like, but rarely exist in secondary lymphoid organs. Mesenchymal stem cells are "diluted" in almost all tissues and organs, making them simple and thin in the upper body of the main battlefield, so that they have strong immunoregulatory capacity in vitro experiments, but have unsatisfactory effects in vivo immunoregulatory therapy.
Clinical tumor patients may be at increased risk of tumor recurrence following hematopoietic stem cell transplantation, such as by massive mesenchymal stem cell infusion [ Ning H, the clinical study of metabolic stem cells and high recurrence rates in clinical malignity patents: outcome of a pilot clinical study. Leukemia.2008; 22: 593-599]. And a large number of research results show that Mesenchymal Stem Cells can promote the generation, growth, drug resistance, invasion and metastasis of tumors [ Eterno V, et al. adopise-derived sensory Stem Cells (ASCs) may facial nerve cancer cell VIF/c-Met signaling. oncotarget.2013; epub ahead of print ] [ Bergfeld SA, Blavier L, Declerck YA. bone Marrow-Derived Mesenchyl structural Cells project overview and Drug Resistance in Tumor Cells The.2014; epub ahead of print ] [ Wang D1, Wang S, Shi C.update on placed diseases of sensory Stem Cell-based therapeutics. Current Stem Cell Res. 2012; 7(5): 370-80.].
In clinical treatment of graft-versus-host disease after hematopoietic stem cell transplantation of tumor patients, immune rejection after organ transplantation in high risk groups with tumor occurrence and autoimmune disease treatment, for example, application of mesenchymal stem cell mass infusion with wide immune suppression function, except for the defects of high price and influence on blood homeostasis, the larger problem is that the mesenchymal stem cell mass infusion brings the risk of increasing tumor occurrence/recurrence. Therefore, common mesenchymal stem cells are cautious, even forbidden, in the treatment of these groups of people.
Based on the disadvantages of weak in-vivo immune regulation capability, large cell dosage and potential increase of tumor recurrence/occurrence risk of common mesenchymal stem cells in clinical treatment, the demand of more economical and efficient cell treatment strategies capable of overcoming the disadvantages is particularly urgent.
Disclosure of Invention
The invention provides an application of a recombinant mesenchymal stem cell in preparation of an immunosuppressant. By modifying the mesenchymal stem cells, the cell membranes of the mesenchymal stem cells express CCR7, the mesenchymal stem cells after infusion are guided to migrate in a targeting manner to enter a secondary lymphoid organ and can be contacted with T lymphocytes in a short distance in a T cell enrichment area gathered in the secondary lymphoid organ, and the T cell mediated immune reaction is effectively inhibited. More particularly, the recombinant mesenchymal stem cells of the invention simultaneously retain the anti-tumor capacity of the body.
In order to achieve the purpose, the invention comprises the following technical scheme:
the application of the recombinant mesenchymal stem cells in preparing the immunosuppressant is characterized in that the recombinant mesenchymal stem cells are obtained by expressing a chemokine receptor CCR7 on the cell membrane of the mesenchymal stem cells, and the immunosuppressant is applied to graft-versus-host disease after hematopoietic stem cell transplantation of high risk tumor groups, immune rejection reaction and/or autoimmune disease after organ transplantation.
For use as described above, preferably the mesenchymal stem cells are of autologous, allogeneic or commercial origin.
The use as described above, preferably, the method for expressing the chemokine receptor CCR7 on the cell membrane of the mesenchymal stem cell comprises: introducing a CCR7 gene into the mesenchymal stem cell by adopting a gene transfection method, so that the cell membrane of the mesenchymal stem cell expresses CCR 7; expressing CCR7 through a vector transfection system on a mesenchymal stem cell membrane; using Trolocytosis method, using antigen presenting cell to present CCR7 to the cell membrane of mesenchymal stem cell; or by modification of existing cell membrane proteins or by exoproteins.
The gene transfection method comprises a viral vector transfection system and a non-viral transfection system.
As mentioned above, preferably, the viral vector transfection system is any one or more of adeno-associated virus AA-V, lentivirus, adenovirus Ad-V, baculovirus, herpes virus and retrovirus transfection system.
As mentioned above, preferably, the non-viral transfection system is a plasmid vector transfection system, a chitosan nanoparticle transfection system, or a naked gene transfection system.
The use as described above, preferably, the high risk group of tumors comprises: 1) middle aged and elderly people over 45 years old; 2) high risk group caused by genetic factors: the history that the direct or collateral relatives within the third generation suffer from malignant tumor; 3) people with bad life habits: people who smoke abundantly for a long time, drink abuses for a long time, abuse of drugs, long-term overwork, stay up all night, have high pressure of life, high pressure of work, have bad mood for a long time, have depressed characters or use the mobile phone frequently; 4) people with poor eating habits: food preference, malnutrition, unclean diet, irregular diet, slow eating speed, frequent eating of overheated, hard and spicy food or frequent eating of pickled smoked fried food; 5) people with occupational factors exposed to toxic and harmful substances for a long time; 6) the people with polluted living environment: water source and/or air chemical pollution, heavy metal pollution, nuclear pollution, microbial pollution or people living in haze weather for a long time; 7) people with chronic or recurrent inflammation: chronic bronchitis, chronic hepatitis B virus infected person, AIDS virus infected person, chronic cervicitis, chronic gastritis, stomach polyp, gastric ulcer, mastitis, mammary gland chronic hyperplasia, intestinal polyp, gallbladder polyp, prostatitis, sinusitis or long-term functional uterine bleeding; 8) and treating benign and malignant tumor patients.
The use as described above, preferably, the autoimmune disease is systemic lupus erythematosus, xerostomia syndrome, rheumatoid arthritis, ankylosing spondylitis, scleroderma, polyarteritis nodosa, Wegener's granulomatosis, ulcerative colitis, autoimmune hemolytic anemia, idiopathic thrombocytopenic purpura, dermatomyositis, mixed connective tissue disease, myasthenia gravis, thyroid autoimmune disease, insulin-dependent diabetes mellitus, malignant anemia with chronic atrophic gastritis, goodpasture's syndrome, pemphigus vulgaris, pemphigoid, primary biliary cirrhosis, multiple sclerosis or acute idiopathic polyneuritis.
For use as described above, preferably, the recombinant mesenchymal stem cells are used in combination with a conventional immunosuppressive agent.
For the above applications, preferably, the dosage of the recombinant mesenchymal stem cells to human body is 1 × 104~1×106MSCs/kg。
The invention has the beneficial effects that:
1. the cell membrane of the mesenchymal stem cell expresses CCR7, after being infused, the mesenchymal stem cell can target the migration of a secondary lymph organ in vivo, and can be greatly accumulated in the local part of the secondary lymph organ, particularly the mesenchymal stem cell can be accumulated in a T cell enrichment area of the secondary lymph organ and can be in close contact with T lymphocytes, so that the T cell mediated immune reaction is effectively inhibited, the cell dosage can be reduced when the graft-versus-host disease after hematopoietic stem cell transplantation, the immune rejection reaction after solid organ transplantation and the autoimmune disease are clinically treated, the cost is reduced, and the side effects of embolism and the like are reduced.
2. The recombinant mesenchymal stem cells of the invention simultaneously reserve the anti-tumor effect of the organism. Therefore, the cell can reduce the risk of tumor recurrence when clinically treating the graft-versus-host disease after the hematopoietic stem cell transplantation of a tumor patient; the risk of tumorigenesis can also be reduced in the treatment of autoimmune diseases and immune rejection reactions after organ transplantation in the high risk group of tumorigenesis.
The immunosuppressant prepared by the recombinant mesenchymal stem cell overcomes the defects of weak in-vivo immunoregulation capability, large cell dosage and potential increase of tumor recurrence/occurrence danger of the common mesenchymal stem cell when treating high-incidence immune reaction of clinical patients, and is obviously superior to the common mesenchymal stem cell. The invention provides a novel efficient and beneficial strategy for clinically inhibiting the immune response of a patient.
Drawings
FIG. 1: CCR7 expression status of Mesenchymal Stem Cells (MSCs).
A: detecting the CCR7 expression conditions of Balb/C mice and C57BL/6 mouse bone marrow-derived MSCs by an RT-PCR method;
b: detecting the expression of eGFP and CCR7 of the three cells by an RT-PCR method;
c: detecting the mRNA expression level of the eGFP genes of the three cells by real-time quantitative PCR;
d: detecting the mRNA expression level of the CCR7 genes of the three cells by real-time quantitative PCR;
e: flow cytometry detected the membrane expression of CCR7 in three cells.
FIG. 2A: fluorescence microscopy images of three groups of mice Spleen (SP), Lymph Node (LN), Mesenteric Lymph Node (MLN), PP node (PP) sections with DAPI nuclear markers.
FIG. 2B: eGFP+Frequency of MSCs in various tissues and organs in vivo.
FIG. 3: MSCs/CCR7-eGFP post tail vein infusion in spleen and lymph nodes with CD3+T cells, CD11c+Dendritic cells and B220+Photomicrographs of B cells.
FIG. 4: experimental results of MSCs/CCR7-eGFP for highly inhibiting graft versus host disease (GvHD).
FIG. 5: in GvHD mouse model, MSCs/CCR7-eGFP was tested for the ability to resist tumors in the body.
FIG. 6: in a mouse xenogeneic skin transplantation model, the results of the immune regulation and control of the MSCs/CCR7-eGFP and the test of the influence on the growth capacity of the tumor of the organism are shown.
FIG. 7: the results of the immune regulation of MSCs/CCR7-eGFP and the test of the influence on the growth capacity of the tumors in the body in a mouse inflammatory bowel disease model.
Detailed Description
The expression of the chemokine receptor CCR7 on the cell membrane of the mesenchymal stem cell can be realized by the following method: introducing a CCR7 gene into the mesenchymal stem cell by adopting a gene transfection method, so that the cell membrane of the mesenchymal stem cell expresses CCR 7; expressing CCR7 through a vector transfection system on a mesenchymal stem cell membrane; using Trolocytosis method, using antigen presenting cell to present CCR7 to the cell membrane of mesenchymal stem cell; or by modification of existing cell membrane proteins or by exoproteins.
Wherein, the gene transfection method comprises a viral vector transfection system and a non-viral transfection system; the virus vector transfection system can be any one or more of adeno-associated virus AA-V, lentivirus, adenovirus Ad-V, baculovirus, herpes virus and retrovirus transfection systems; the non-viral transfection system can be a plasmid vector transfection system, a chitosan nanoparticle transfection system or a naked gene transfection system.
The present invention will be further described with reference to the following examples, but the present invention is not limited to the following examples.
In the following examples, unless otherwise specified, all methods are conventional.
Example 1 preparation of recombinant mesenchymal Stem cells expressing chemokine receptor CCR7
1. Mouse bone marrow MSCs separation culture
The sample is 2-3 weeks old C57BL/6 mouse (Experimental animal center of military medical science institute), the mouse is killed by breaking neck, the mouse is soaked in 75% alcohol and disinfected → the tibia and femur of the mouse are separated under aseptic condition, the mouse is placed in a flat dish → the adhered muscle is rubbed by using sterile gauze, the metaphysis is removed → the marrow cavity is repeatedly washed by PBS + 10% FCS (Hyclone), the marrow cells are washed out → inoculated in a culture bottle, the bone fragments are uniformly distributed on the bottom of the bottle, the culture system contains alpha t-MEM (Gibco) + 10% FCS (Hyclone) +100U/ml penicilliumSumin +100U/ml streptomycin, 37 deg.C, 5% CO2Culturing under saturated humidity conditions → changing the solution 1 time every 3 days → when the cells are approximately 70-80% confluent, digesting and passaging with 0.25% pancreatin (Hyclone), and the inoculation density of the passaged cells is 4000 cells/cm2
2. Adeno-associated virus infected mouse MSCs
eGFP-adeno-associated virus vector and CCR 7-eGFP-adeno-associated virus vector, both of which were prepared by Invitrogen, were transferred to mouse MSCs. The method comprises the following steps: spreading MSCs on 48-well plate (2 × 10)4A hole. The next day, the medium was changed to a new medium, to which virus (MOI ═ 20), polybrene (8 μ g/ml) was added, and the normal culture system was changed 6 hours after infection. The MSCs introduced with the eGFP are named MSCs/eGFP; MSCs introduced into the CCR7-eGFP vector were designated MSCs/CCR 7-eGFP.
RT-PCR detection of CCR7 expression of MSCs at mRNA level
MSCs isolated from BalB/C mice and C57BL/6 mice were collected, splenic mononuclear cells (SPMNC) of the same strain were used as positive controls, and transfected MSCs/eGFP, MSCs/CCR7-eGFP, were collected. The RT-PCR method is used for detecting the mRNA level of the cells expressing CCR 7. The primers are as follows:
TABLE 1
Figure BDA0000501481170000061
The results are shown in FIG. 1, FIG. 1A: RT-PCR results showed that neither BalB/C mice nor C57BL/6 mouse-derived MSCs expressed CCR7, whereas spleen cells of the same strain origin (SPMNC) were positive. The expression of housekeeping gene HPRT is used as an internal reference; FIG. 1B: normal MSCs were used as blank controls, MSC with empty vector as transfection control (MSCs/eGFP), and CCR7 gene was transferred into MSCs (MSCs/CCR 7-eGFP). The RT-PCR detection result shows that: MSC/eGFP and MSC/eGFP-CCR7 express eGFP; of the three cells, MSC/CCR7-eGFP successfully expressed CCR7 mRNA, and expression of the housekeeping gene HPRT served as an internal control. This result indicates that the CCR7 gene was successfully transferred into MSCs.
4. Real-time quantitative PCR method for detecting mRNA level of expression CCR7 of MSCs
And respectively collecting three groups of cells including MSCs, MSCs/eGFP and MSCs/eGFP-CCR 7. RNA was extracted by the conventional method and the mRNA level of CCR7 of the three cells was detected by real-time quantitative PCR. The primers are as follows:
TABLE 2
Figure BDA0000501481170000062
See fig. 1C, D: detecting mRNA expression levels of eGFP genes and CCR7 genes of three cells including MSCs, MSCs/eGFP and MSCs/eGFP-CCR7 by real-time quantitative PCR; the results of fig. 1B are further confirmed.
5. Flow cytometry detection of MSCs membrane expression CCR7
The three groups of cells, namely the MSCs, the MSCs/eGFP and the MSCs/CCR7-eGFP, are respectively blocked by a mouse CD16/32 antibody (ebioscience company), and then the cells are labeled by a CCR7-PE antibody (ebioscience company), and the detection result of a flow cytometer in FIG. 1E shows that: the MSCs and the MSCs/eGFP do not express the membrane molecule CCR7, and 93.0-98.4% of the MSCs/CCR7-eGFP expresses the membrane molecule CCR 7.
Example 2 detection of the ability of MSCs to migrate to Secondary lymphoid organs in vivo and to enrich the region
1. Detection of migration ability of MSCs to secondary lymphoid organs in vivo
Infusion of MSCs, MSCs/eGFP or MSCs/CCR7-eGFP (1X 10)6) Three groups of mice (4 per group); spleen (SP), Lymph Node (LN), Mesenteric Lymph Node (MLN), PP node were cryosectioned 5 days after infusion and nuclear labeling was performed with DAPI. The green fluorescent cells were observed under a fluorescence microscope. The results in FIG. 2A show that: MSCs/eGFP did not have the ability to migrate to secondary lymphoid organs, whereas MSCs/eGFP-CCR7 has a large distribution in SP, LN, MLN, PP. FIG. 2B shows the calculation of eGFP for each organ+The frequency of distribution of MSCs in each tissue organ in vivo (number of green fluorescences/high power field) quantitatively showed that infused MSCs/eGFP had no ability to migrate to secondary lymphoid organs in vivo, whereas infused MSCs/eGFP-CCR7 had the ability to target migration of secondary lymphoid organs in vivo.
MSCs/eGFP-CCR7 present in T cell-rich regions in secondary lymphoid organs
Mixing MSCsFrozen sections of SP, LN from mice infused with/eGFP-CCR 7 were immunofluorescent stained and incubated with phycoerythrin-labeled (PE) CD3 or B220 antibodies and then observed under a fluorescent microscope for MSCs/CCR7-eGFP (green) and CD3+T lymphocytes (Red, FIG. 3A), CD11c+Dendritic cells (Red, FIG. 3B) or B220+Relationship of B lymphocytes (red, fig. 3C), DAPI (blue) staining shows nuclei. There are many cells in the secondary lymphoid organs, MSCs/CCR7-eGFP are present in T cell-enriched regions in secondary lymphoid organs, which lays the foundation for MSCs/CCR7-eGFP having strong immunomodulatory capacity in vivo against T cell-mediated immune responses.
Example 3 detection of the Effect of MSCs/CCR7-eGFP on in vivo immunomodulation and Effect on tumor growth
Inhibition of GvHD by MSCs/CCR7-eGFP
GvHD is a typical T cell-mediated immune disease. Establishing a mouse GvHD model: collecting 20-24g BALB/c male mouse, collecting spleen cells and bone marrow cells, and counting; 2X 107Splenocytes +1 × 107Bone marrow cells/0.25 ml PBS tail vein injection of 22-24g C57BL/6 male mice irradiated with 9Gy to establish aGvHD model (GvHD group). On the basis of the above-mentioned two-step co-infusion, 1X 105Or 1X 106 1X 10 MSCs/eGFP of5 Or 1X 106MSCs/CCR7-eGFP。1×106Mice co-infused with MSCs/eGFP tail vein were referred to as GvHD + MSCs/eGFP mice; 1X 106Mice co-infused tail vein with MSCs/CCR7-eGFP were designated GvHD + MSCs/CCR7-eGFP mice.
After infusion, three groups of mice were observed daily for survival and status, survival curves were recorded to observe the in vivo immunomodulatory effects of MSCs, and serum cytokine levels were measured. As shown in FIG. 4A, the MSCs/CCR7-eGFP infusion significantly prolonged the survival time of GvHD mice, with effects significantly better than the same dose of MSCs/eGFP infusion group. After 2 weeks of modeling, GvHD mice showed weight loss, bows and backs, diarrhea, rough hair, and reduced mobility. The GvHD performance of mice infused with MSCs/CCR7-eGFP is obviously improved, and the effect of the GvHD is obviously better than that of the MSCs/eGFP co-infusion group with the same dose (figure 4B). ELISA results showed that co-infusion of MSCs/CCR7-eGFP significantly increased the level of anti-inflammatory factor IL-4 in the serum of mice (FIG. 4C) and significantly decreased the level of inflammatory factor IFN-. gamma. (FIG. 4D) compared to GvHD, GvHD + MSCs/eGFP mice. The above results demonstrate that: MSCs expressing CCR7 have greater in vivo immunomodulatory capacity than normal MSCs.
2. Detection of the influence of MSCs/eGFP-CCR7 on the anti-tumor capability of an organism in a mouse GvHD model
Mouse GvHD, GvHD + MSCs/eGFP (1X 10)6)、MSCs/CCR7-eGFP(1×106) On the day of model creation, co-infusion was 5 × 105EL4 leukemia cells to establish a tumor-bearing GvHD mouse model. Survival time of each group of mice was recorded, survival curves were plotted, and the effect of the two MSCs on tumor growth was observed. As a result, it was found that: MSCs/eGFP co-infusion did not prolong the survival time of mice compared to GvHD + EL4 tumor-bearing mice, whereas MSCs/eGFP-CCR7 co-infusion significantly prolonged the survival time of mice (fig. 5A); tumor infiltration of the liver of each group of mice was observed 2 weeks after modeling, and the results in fig. 5B show: syngeneic splenocytes and tumor cells co-infused group (syno-infusion + EL4) mice had obvious tumor nodules in their livers, demonstrating successful tumor loading in mice (FIG. 5 Ba); the liver of mice in GvHD + EL4 group had obvious hemorrhage and necrosis, indicating that the mice had obvious GvHD (figure 5 Bb); the GvHD + EL4+ MSCs/eGFP group mice have certain improvement on liver GvHD change and still have a plurality of tumor nodules (figure 5 Bc); the GvHD + EL4+ MSCs/CCR7-eGFP mice have improved hemorrhage, necrosis and tumor nodules, which indicates that the co-infusion of the MSCs/CCR7-eGFP reduces GvHD and simultaneously retains the anti-tumor capability of the body (figure 5 Bd);
3. xenograft skin graft model (alloSG) mice were loaded with melanoma (B16) and the effect of MSCs/eGFP-CCR7 on the ability of body tumor growth was examined.
Establishing a melanoma-loaded xenogeneic skin transplantation mouse model (alloSG + B16+ PBS): Balb/C mice skin (1 Gm)2) Transplanted to the back of C57 mouse, melanoma cells (B16) 5X 105Performing lateral abdominal subcutaneous injection, establishing a melanoma-loaded mouse xenogeneic skin transplantation model, and infusing 0.2ml PBS (phosphate buffer solution) into tail vein on the next day; tail vein infusion of 1X 10 on model basis6MSCs/eGFP, defined as alloSG + B16+ MSCs/eGFP set; or tail vein infusion of 1X 106MSCs/CCR7-eGFP, defined as alloSG + B16+ MSCs/CCR7-eGFP group.
The survival time of the xenogenic skin of the three groups of mice was observed and showed that the xenogenic skin survival time of the mice of the alloSG + B16+ MSCs/CCR7-eGFP group was significantly prolonged (×, p < 0.05), whereas the survival time of the xenogenic skin of the mice of the alloSG + B16+ MSCs/eGFP group was not different from that of the control group (fig. 6A). After 2 weeks of modeling, the melanoma of the alloSG + B16+ MSCs/eGFP group was significantly larger in volume and significantly increased in weight (p < 0.05) than the control group; however, the melanoma group of alloSG + B16+ MSCs/CCR7-eGFP was not significantly different from the control group in both volume and weight (n.s., no significant difference) (fig. 6B, C).
4. In an melanoma-loaded mouse Inflammatory Bowel Disease (IBD) model, the immune regulation of MSCs/CCR7-eGFP and the ability to grow tumors in the body were tested.
Mice were fed with 2% DSS for 7 consecutive days, with melanoma cells (B16) at 5X 10 on the first day of feeding water5Lateral abdominal subcutaneous injection, tail vein infusion of 0.2ml PBS induced a melanoma-loaded mouse model of inflammatory bowel disease (IBD + B16). Tail vein infusion of 1X 10 on model basis6MSCs/eGFP, defined as IBD + B16+ MSCs/eGFP group; or tail vein infusion of 1X 106MSCs/CCR7-eGFP, defined as the IBD + B16+ MSCs/CCR7-eGFP group.
The survival time of three groups of mice is observed, and the result shows that: IBD + B16+ MSCs/CCR7-eGFP mice survived significantly longer than controls (FIG. 7A). After 14 days of modeling, the melanoma of IBD + B16+ MSCs/eGFP group is obviously larger than that of the control group in volume and obviously increased in weight (p is less than 0.05); however, there was no significant difference in the volumes and weights of the IBD + B16+ MSCs/CCR7-eGFP group melanomas from the control group (n.s., no significant difference) (fig. 7B).

Claims (9)

1. The application of recombinant mesenchymal stem cells in preparing an immunosuppressant for treating immunosuppressive diseases and retaining the anti-tumor effect of an organism is disclosed, the recombinant mesenchymal stem cells are obtained by expressing a chemokine receptor CCR7 on cell membranes of the mesenchymal stem cells, and the immunosuppressive diseases are graft-versus-host diseases after hematopoietic stem cell transplantation, immune rejection reactions after organ transplantation and/or autoimmune diseases.
2. The use of claim 1, wherein the recombinant mesenchymal stem cells are autologous or allogeneic in origin.
3. The use of claim 1, wherein the method of expressing chemokine receptor CCR7 on the cell membrane of a mesenchymal stem cell comprises: introducing a CCR7 gene into the mesenchymal stem cell by adopting a gene transfection method, so that the cell membrane of the mesenchymal stem cell expresses CCR 7; expressing CCR7 through a vector transfection system on a mesenchymal stem cell membrane; using Trolocytosis method, using antigen presenting cell to present CCR7 to the cell membrane of mesenchymal stem cell; or by modification of existing cell membrane proteins or by exoproteins.
4. The use of claim 3, wherein the gene transfection method comprises a viral vector transfection system and a non-viral transfection system.
5. The use according to claim 4, wherein the viral vector transfection system is any one or more of adeno-associated virus AA-V, lentivirus, adenovirus Ad-V, baculovirus, herpes virus and retrovirus transfection systems.
6. The use of claim 4, wherein the non-viral transfection system is a plasmid vector transfection system, a chitosan nanoparticle transfection system, or a naked gene transfection system.
7. The use of claim 1, wherein the tumor is melanoma.
8. The use as claimed in claim 1, wherein the autoimmune disease is systemic lupus erythematosus, xerostomia syndrome, rheumatoid arthritis, ankylosing spondylitis, scleroderma, polyarteritis nodosa, Wegener's granulomatosis, ulcerative colitis, autoimmune hemolytic anemia, idiopathic thrombocytopenic purpura, dermatomyositis, mixed connective tissue disease, myasthenia gravis, thyroid autoimmune disease, insulin-dependent diabetes mellitus, pernicious anemia with chronic atrophic gastritis, goodpasture's syndrome, pemphigus vulgaris, pemphigoid, primary biliary cirrhosis, multiple sclerosis of the cerebrospinal fluid or acute idiopathic polyneuritis.
9. The use of any one of claims 1-8, wherein the recombinant mesenchymal stem cells are used in combination with a conventional immunosuppressive agent.
CN201410188453.1A 2014-01-28 2014-05-06 Application of recombinant mesenchymal stem cells in preparation of immunosuppressant Active CN104800243B (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN201410188453.1A CN104800243B (en) 2014-01-28 2014-05-06 Application of recombinant mesenchymal stem cells in preparation of immunosuppressant

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
CN2014100409564 2014-01-28
CN201410040956 2014-01-28
CN201410188453.1A CN104800243B (en) 2014-01-28 2014-05-06 Application of recombinant mesenchymal stem cells in preparation of immunosuppressant

Publications (2)

Publication Number Publication Date
CN104800243A CN104800243A (en) 2015-07-29
CN104800243B true CN104800243B (en) 2021-04-30

Family

ID=53685732

Family Applications (1)

Application Number Title Priority Date Filing Date
CN201410188453.1A Active CN104800243B (en) 2014-01-28 2014-05-06 Application of recombinant mesenchymal stem cells in preparation of immunosuppressant

Country Status (1)

Country Link
CN (1) CN104800243B (en)

Families Citing this family (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN105343894B (en) * 2015-11-26 2019-01-29 中国人民解放军军事医学科学院基础医学研究所 The recombination mescenchymal stem cell of efficient secretion prostaglandin E2 (PGE2) and its application
CN105316286B (en) * 2015-11-26 2018-12-14 中国人民解放军军事医学科学院基础医学研究所 A kind of method of preparation and reorganization mescenchymal stem cell and its obtained recombination mescenchymal stem cell
WO2017214939A1 (en) * 2016-06-16 2017-12-21 毛侃琅 Lentiviral expression vector for improving expression level of ccr7 gene, and applications thereof
KR101971323B1 (en) * 2016-10-17 2019-04-23 사회복지법인 삼성생명공익재단 Methods for Selecting Improved Stem Cell for Treating Immune Disease
WO2018077385A1 (en) * 2016-10-25 2018-05-03 Biontech Rna Pharmaceuticals Gmbh Dose determination for immunotherapeutic agents
CN111139222B (en) * 2019-05-30 2020-12-29 北京双因生物科技有限公司 Recombinant mesenchymal stem cell and preparation method and application thereof
CN110447601A (en) * 2019-08-09 2019-11-15 南京医科大学 A kind of targeting macrophage is overexpressed the preparation method of genetic mouse model

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN102031244A (en) * 2010-11-08 2011-04-27 中国人民解放军军事医学科学院基础医学研究所 Recombinant mesenchymal stem cell and preparation method and application thereof

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR20080096091A (en) * 2007-04-27 2008-10-30 재단법인서울대학교산학협력재단 Novel use of chemokine receptor 7 for controlling immune response

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN102031244A (en) * 2010-11-08 2011-04-27 中国人民解放军军事医学科学院基础医学研究所 Recombinant mesenchymal stem cell and preparation method and application thereof

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
CCR7 Guides Migration of Mesenchymal Stem Cell;HONG LI;《STEM CELL》;20140204;第32卷;1890-1903 *
Mesenchymal Stem Cells;Biju Parekkadan;《Annu. Rev. Biomed. Eng.》;20100420;第12卷;87–117 *

Also Published As

Publication number Publication date
CN104800243A (en) 2015-07-29

Similar Documents

Publication Publication Date Title
CN104800243B (en) Application of recombinant mesenchymal stem cells in preparation of immunosuppressant
US11464806B2 (en) Genetically modified mesenchymal stem cells expressing an immune response-stimulating cytokine to attract and/or activate immune cells
JP6899333B2 (en) General-purpose killer T cells
Wei et al. Adipose-derived stem cells promote tumor initiation and accelerate tumor growth by interleukin-6 production
JP6764912B2 (en) Cell population with immunomodulatory activity, its preparation method and use
KR101846464B1 (en) Natural killer cell comprising foreign mitochondria and pharmaceutical composition comprising same
Mao et al. MicroRNA-23a is involved in tumor necrosis factor-α induced apoptosis in mesenchymal stem cells and myocardial infarction
Koh et al. The pro‐metastatic role of bone marrow‐derived cells: a focus on MSCs and regulatory T cells
KR101507174B1 (en) Immune privileged and modulatory progenitor cells
CN102575231B (en) Process for production of cytokine-induced killer cells
Song et al. Mouse bone marrow-derived mesenchymal stem cells inhibit leukemia/lymphoma cell proliferation in vitro and in a mouse model of allogeneic bone marrow transplant
CN105296431A (en) Tumor binding specific gamma delta TCR gene modified alpha beta T cell and cancer suppression application thereof
US20210071143A1 (en) Active cxcr4+ immune cells and methods for their production and use
CN103396992A (en) Culture and application of oligoclonal tumor-infiltrating lymphocytes for liver cancer
CN113166224A (en) anti-LMP 2TCR-T cell therapy for treating EBV-associated cancer
CN114410686B (en) Engineering exosome in-situ nanometer vaccine and preparation method and application thereof
CN104434973A (en) Method for intensifying functions of cytokine-induced killer cells
CN105535953A (en) NKT cell preparation sensitized by tumor cells and stimulating factors and preparation method thereof
CN115944650B (en) Application of tumor infiltration cells in preparation of antitumor drugs and model construction method
CN117304343B (en) Preparation and application of GPC3 targeted CAR-NK cells
RU2777371C2 (en) Natural killer cell containing exogenous mitochondria, and pharmaceutical composition including such a cell
Xu Recruitment and Polarization of Tumor Associated Macrophages
Dabaja CD8+ T Cell Exhaustion and Immune Checkpoint Inhibitors in Mutant Isocitrate Dehydrogenase I Gliomas
CN118185877A (en) Gene editing CD8+T cell and application thereof in preparation of medicines for treating tumors
CN104436178A (en) Application of annexin A3 in preparation of drug for target killing liver cancer stem cells

Legal Events

Date Code Title Description
C06 Publication
PB01 Publication
EXSB Decision made by sipo to initiate substantive examination
SE01 Entry into force of request for substantive examination
GR01 Patent grant
GR01 Patent grant