Merck
CN
HomeHematologyRecommended Standard Method for Isolating Mononuclear Cells

Recommended Standard Method for Isolating Mononuclear Cells

Cell separation using Ficoll-Paque products can be carried out over a wide range of blood sample volumes. With its high yield, this method can be adapted to the processing of very small amounts of blood, such as may be obtained from children. For maximum reproducibility of separation it is recommended that a standardized procedure be used. The following procedure has been evaluated in our laboratories with Ficoll-Paque PLUS and is recommended for separation of normal blood samples. Simple changes can easily be made to suit a particular centrifugation system. The same procedure is recommended when separating cells using Ficoll-Paque PREMIUM, Ficoll-Paque PREMIUM 1.084, and Ficoll-Paque PREMIUM 1.073.

To standardize the technique, blood volume and diameter of the centrifuge tube should be chosen first. These factors determine the height of the blood sample in the tube and consequently the centrifugation time. Increasing the height of the blood sample in the tube increases red cell contamination. The separation is, however, not appreciably affected by changing the diameter of the tube. Hence a larger volume can be separated with the same degree of purification in a tube of larger diameter if the height of the blood sample in the tube and the separation time are kept constant.

The yield and degree of purity of the mononuclear cells depend to a considerable extent on the efficiency of red cell removal.

When erythrocytes in whole blood are aggregated, some mononuclear cells are trapped in the clumps and therefore sediment with the erythrocytes. This tendency to trap mononuclear cells is reduced by diluting the blood. Dilution gives a better yield of mononuclear cells and reduces the size of the red cell clumps. Aggregation of erythrocytes is enhanced at higher temperatures (37 °C), which consequently decreases the yield of mononuclear cells. At lower temperatures (4 °C); however, the rate of aggregation is decreased but the time of separation is increased, which also decreases the yield of mononuclear cells. A compromise temperature of 18 ºC to 20 °C gives optimal results.

Equipment and solutions required but not provided

  1. Sterile balanced salt solution or other standard salt solutions (see Preparation of reagents).
  2. Centrifuge with swing-out rotor (brake should be off).
  3. Sterile centrifuge tubes and pipettes.
  4. Sterile needles and syringes.
  5. Red blood cell lysis solution of choice (if isolating granulocytes).

Preparation of reagents

Diluent and washing solution
The balanced salt solution for dilution of the blood and cell washing can be made according to the instruction below. Other diluents and washing fluids such as isotonic Ca2+/Mg2+ free phosphate buffered saline (e.g. Dulbecco’s PBS), salt solutions (e.g., Hank’s) or cell culture media (e.g., RPMI 1640) may also be used.

Balanced salt solution
To prepare the balanced salt solution, mix one volume stock solution A with nine volumes stock solution B and sterilize. At least 20 mL for each sample should be processed. Other sterile standard salt solutions may also be used.

Dissolve in approximately 950 mL of distilled water and add concentrated HCl until the pH is 7.6 before adjusting the volume to 1L.

To prepare the balanced salt solution, mix 1 volume of solution A with 9 volumes of solution B. Prepare the solution freshly each week. Other standard salt solutions may be used.

Ficoll-Paque product
Warm the Ficoll-Paque density gradient media to 18 °C to 20 °C before use. For samples larger than 3 mL, see Notes.

Preparation of the sample
Fresh blood should be used to ensure high viability of isolated mononuclear cells. Prepare the sample at 18 ºC to 20 °C.

  1. To a 10 mL centrifuge tube, add 2 mL of defibrinated- or anticoagulant-treated blood and an equal volume of balanced salt solution (final volume 4 mL).
  2. Mix the blood and buffer by inverting the tube several times or by drawing the mixture in and out of a pipette.

Procedure for isolation of mononuclear cells

  1. Invert the Ficoll-Paque media bottle several times to ensure thorough mixing.
    For withdrawal of Ficoll-Paque media by syringe: Snap-off the polypropylene cap and insert the syringe needle through the septum (Figure 1).
    For withdrawal of Ficoll-Paque media by pipette: Remove the snap-off polypropylene cap. Lift the aluminum ring. Pull off the metal seal. Remove the silver ring.
    Remove the rubber closure. Using aseptic techniques, withdraw the required volume of Ficoll-Paque media (Figure 2).
  2. Add Ficoll-Paque media (3 mL) to the centrifuge tube.
  3. Carefully layer the diluted blood sample (4 mL) onto the Ficoll-Paque media solution (Figure 3).
    Important: When layering the sample do not mix the Ficoll-Paque media solution and the diluted blood sample.
  4. Centrifuge at 400 g for 30 to 40 min at 18 ºC to 20 °C (brake should be turned off).
  5. Draw off the upper layer containing plasma and platelets using a sterile pipette, leaving the mononuclear cell layer undisturbed at the interface (Figure 4 and Figure 5). The upper layer, which contains the plasma, may be saved for later use.
  6. Transfer the layer of mononuclear cells to a sterile centrifuge tube using a sterile pipette.
Polypropylene cap and silver ring removal

Figure 1.Polypropylene cap and silver ring removal

Withdrawal of Ficoll-Paque media

Figure 2. Withdrawal of Ficoll-Paque media

Blood sample layered onto Ficoll-Paque media

Figure 3.Blood sample layered onto Ficoll-Paque media

Prior to removal of the upper layer.

Figure 4.Prior to removal of the upper layer.

After the upper layer (Plasma) removed.

Figure 5.After the upper layer (Plasma) removed.

Washing the cell isolate

  1. Estimate the volume of the transferred mononuclear cells. Add at least 3 volumes (~ 6 mL) of balanced salt solution to the mononuclear cells in the centrifuge tube.
  2. Suspend the cells by gently drawing them in and out of a pipette.
  3. Centrifuge at 400 to 500 × g for 10 to 15 min at 18 °C to 20 °C.

Note: A centrifugation at high speed increases the mononuclear cell recovery. However, if it is important to also get rid of platelets a lower centrifugation speed is recommended (60 to 100 × g).

  1. Remove the supernatant.
  2. Resuspend the mononuclear cells in 6 to 8 ml balanced salt solution.
  3. Centrifuge at 400 to 500 × g (or 60 to 100 × g for removal of platelets) for 10 min at 18 °C to 20 °C.
  4. Remove the supernatant.
  5. Resuspend the cell pellet in media appropriate for the application.

Procedure for isolation of granulocytes

  1. Perform density gradient centrifugation using Ficoll-Paque media as described above in Procedure for isolation of mononuclear cells, steps 1 to 6.
  2. Draw off the upper layer of Ficoll-Paque media using a sterile pipette, leaving the white cell layer of granulocytes above the red blood cell layer undisturbed.
  3. Collect the thin white cell layer of granulocytes with a pipette and transfer to a sterile centrifuge tube.
  4. Resuspend the cells in at least five volumes of balanced salt solution and centrifuge at 400 × g for 15 min.
  5. Lyse remaining red blood cells with any red blood cell lysis solution of choice.
  6. Centrifuge the granulocytes at 400 to 500 × g for 10 to 15 min at 18 °C to 20 °C.
  7. Remove the supernatant.
  8. Resuspend the granulocytes in 6 to 8 ml balanced salt solution.
  9. Centrifuge at 400 to 500 × g for 10 min at 18 °C to 20 °C.
  10. Remove the supernatant.
  11. Resuspend the cell pellet in media appropriate for the application.

Notes
Anticoagulants:
Heparin, EDTA, citrate, acid citrate dextrose (ACD), and citrate phosphate dextrose (CPD) may be used as anticoagulants for the blood sample. Defibrinated blood requires no anticoagulant. Defibrination, however, results in a lower mononuclear cell yield and may cause increased contamination by red cells3. It also causes selective loss of monocytes.

Bøyum has found that a slightly purer mononuclear cell preparation is obtained using EDTA instead of heparin as anticoagulant3. It has also been noted in the purification of mononuclear cells from sources other than peripheral blood that addition of heparin may cause gelling of cell suspensions4. Citrate-stabilized blood may result in better quality RNA and DNA than other anticoagulants, and produce a higher yield of mononuclear cells. Heparin affects T-cell proliferation and binds to many proteins. EDTA is good for DNA based assays, but influences Mg2+ concentration and causes problems for cytogenetic analysis5. It has also been shown that the RNA yield is higher in EDTA-blood than in heparin-blood6.

Blood volume: Larger volumes of blood may be processed with the same efficiency of separation by using centrifuge tubes of increased diameter while maintaining approximately the same heights of Ficoll-Paque media (2.4 cm) and blood sample (3.0 cm) as in the standard method described above. Increasing the tube diameter does not affect the separation time required.

Blood sample storage: Blood samples should be free of clots and processed as soon as possible after collection to ensure optimal results. Delays in processing the blood can result in loss of viability, lower cell recoveries and more contaminating granulocytes and/or erythrocytes. Indeed, storage for 24 h at room temperature has been reported to result in reduced lymphocyte yield, altered expression of surface markers, and reduced response to mitogenic stimulation 5,7,8. Dead cell removal: Dead cells are removed during cell isolation using Ficoll-Paque PLUS9,10,58.

Density and temperature: The temperature at which density gradient separations are carried out are naturally affected by room temperature, centrifuge temperature, temperature of the density gradient media, and temperature of the liquid sample. There can, at times, also be confusion regarding what is meant by room temperature (e.g., in Europe it can be 20 ºC and in North America > 20 ºC). It is known that the densities of Ficoll-Paque products decrease with increase in temperature. For instance Ficoll-Paque PREMIUM (1.077 g/ml) exhibits a density of 1.0772, 1.0767, and 1.0758 at 20 ºC, 22 ºC, and 25 ºC, respectively.

Pathological blood samples: The standard method described above has been developed for the purification of mononuclear cells from peripheral blood of normal, healthy, human donors. Different results may be obtained with samples taken from donors with infections or other pathological conditions, such as cancer (see Further applications of Ficoll-Paque PLUS and Ficoll-Paque PREMIUM, page 12).

Platelet removal: If it is important to remove all platelets from the mononuclear cell fraction a second centrifugation in a 4% to 20% sucrose gradient layered over Ficoll-Paque PLUS can be applied. This procedure will effectively remove any platelet contamination11. Platelets will remain at the top of the sucrose gradient and mononuclear cells will sediment through the sucrose gradient to the top of the Ficoll-Paque PLUS layer. Alternatively, the platelets may be removed by aggregation with adenosine-5’-diphoshate (ADP) before separating the mononuclear cells12.

Troubleshooting inadequate performance
If used according to the recommended standard procedure, Ficoll-Paque PLUS and Ficoll-Paque PREMIUM may be expected to give trouble-free isolation of human peripheral blood mononuclear cells with results as shown on page 2. As mentioned earlier, Ficoll-Paque PREMIUM 1.073 and Ficoll-Paque PREMIUM 1.084 will lead to isolation of cell preparations having slightly different density subsets of mononuclear cells. However, deviations in certain experimental parameters may lead to poor results and the troubleshooting chart given here is intended to assist in the rapid identification and correction of the problem causing reduced performance.

Materials
Loading

References

1.
Böyum A. 1968. Isolation of mononuclear cells and granulocytes from human blood. Isolation of monuclear cells by one centrifugation, and of granulocytes by combining centrifugation and sedimentation at 1 g. Scand J Clin Lab Invest Suppl. 9777-89.
2.
Böyum A. 1968. Isolation of leucocytes from human blood – further observations. Scand. J. Clin. Lab. Invest. Suppl. 9731–50.
3.
BØYUM A. 1976. Isolation of Lymphocytes, Granulocytes and Macrophages. 59-15. https://doi.org/10.1111/j.1365-3083.1976.tb03851.x
4.
Almeida AP, A. Beaven M. 1980. Gel formation with leucocytes and heparin. Life Sciences. 26(7):549-555. https://doi.org/10.1016/0024-3205(80)90318-5
5.
Holland NT, Smith MT, Eskenazi B, Bastaki M. 2003. Biological sample collection and processing for molecular epidemiological studies. Mutation Research/Reviews in Mutation Research. 543(3):217-234. https://doi.org/10.1016/s1383-5742(02)00090-x
6.
Marteau J, Mohr S, Pfister M, Visvikis-Siest S. 2005. Collection and Storage of Human Blood Cells for mRNA Expression Profiling: A 15-Month Stability Study. 51(7):1250-1252. https://doi.org/10.1373/clinchem.2005.048546
7.
Kaplan J, Nolan D, Reed A. 1982. Altered lymphocyte markers and blastogenic responses associated with 24 hour delay in processing of blood samples. Journal of Immunological Methods. 50(2):187-191. https://doi.org/10.1016/0022-1759(82)90224-1
8.
Imeri F, Herklotz R, Risch L, Arbetsleitner C, Zerlauth M, Risch GM, Huber AR. 2008. Stability of hematological analytes depends on the hematology analyser used: A stability study with Bayer Advia 120, Beckman Coulter LH 750 and Sysmex XE 2100. Clinica Chimica Acta. 397(1-2):68-71. https://doi.org/10.1016/j.cca.2008.07.018
9.
Paietta E. 2003. How to optimize multiparameter flow cytometry for leukaemia/lymphoma diagnosis. Best Practice & Research Clinical Haematology. 16(4):671-683. https://doi.org/10.1016/s1521-6926(03)00070-7
10.
Dolan BP, Gibbs KD, Ostrand-Rosenberg S. 2006. Dendritic Cells Cross-Dressed with Peptide MHC Class I Complexes Prime CD8+ T Cells. J Immunol. 177(9):6018-6024. https://doi.org/10.4049/jimmunol.177.9.6018
11.
Perper RJ, Zee TW, Mickelson MM. 1968. Purification of lymphocytes and platelets by gradient centrifugation. J Lab Clin Med. 72(5):842-8.
12.
VIVES J, PARRA M, CASTILLO R. Platelet Aggregation Technique Used in the Preparation of Lymphocyte Suspensions. 1(6):276-278. https://doi.org/10.1111/j.1399-0039.1971.tb00106.x
13.
Alexander EL, Titus JA, Segal DM. 1978. Quantitation of Fc receptors and surface immunoglobulin is affected by cell isolation procedures using plasmagel and Ficoll-Hypaque. Journal of Immunological Methods. 22(3-4):263-272. https://doi.org/10.1016/0022-1759(78)90034-0
14.
Hokland P, Heron I. 1980. Analysis of the lymphocyte distribution during isopaque-ficoll isolation of mononuclear cells from human peropheral blood. Journal of Immunological Methods. 32(1):31-39. https://doi.org/10.1016/0022-1759(80)90114-3
15.
HOKLAND P, HERON I. 1980. The Isopaque-Ficoll Method Re-evaluated: Selective Loss of Autologous Rosette-forming Lymphocytes during Isolation of Mononuclear Cells from Human Peripheral Blood. Scand J Immunol. 11(3):353-356. https://doi.org/10.1111/j.1365-3083.1980.tb00245.x
16.
AssumpcióRomeu M, Mestre M, González L, Valls A, Verdaguer J, Corominas M, Bas J, Massip E, Buendia E. 1992. Lymphocyte immunophenotyping by flow cytometry in normal adults. Journal of Immunological Methods. 154(1):7-10. https://doi.org/10.1016/0022-1759(92)90206-9
17.
Lin S, Chao H, Yan D, Huang Y. 2002. Expression of adhesion molecules on T lymphocytes in young children and infants - a comparative study using whole blood lysis or density gradient separation. Clin Lab Haematol. 24(6):353-359. https://doi.org/10.1046/j.1365-2257.2002.00462.x
18.
Bain B, Pshyk K. 1973. Reactivity in mixed cultures of mononuclear leucocytes separated on Ficoll-Hypaque. Proceedings 7th Leucocyte Culture Conference; New York Academic Press. p. 29–37.
19.
Wu Y, Lu H, Cai J, He X, Hu Y, Zhao H, Wang X. 2009. Membrane Surface Nanostructures and Adhesion Property of T Lymphocytes Exploited by AFM. Nanoscale Res Lett. 4(8):942-947. https://doi.org/10.1007/s11671-009-9340-8
20.
Guia S, Cognet C, de Beaucoudrey L, Tessmer MS, Jouanguy E, Berger C, Filipe-Santos O, Feinberg J, Camcioglu Y, Levy J, et al. 2008. A role for interleukin-12/23 in the maturation of human natural killer and CD56+ T cells in vivo. 111(10):5008-5016. https://doi.org/10.1182/blood-2007-11-122259
21.
Frelin C. 2005. Targeting NF- B activation via pharmacologic inhibition of IKK2-induced apoptosis of human acute myeloid leukemia cells. Blood. 105(2):804-811. https://doi.org/10.1182/blood-2004-04-1463
22.
van den Akker ELT, Baan CC, van den Berg B, Russcher H, Joosten K, Hokken-Koelega ACS, Lamberts SWJ, Koper JW. 2008. Ficoll-separated mononuclear cells from sepsis patients are contaminated with granulocytes. Intensive Care Med. 34(5):912-916. https://doi.org/10.1007/s00134-007-0989-0
23.
Chan JK, Hamilton CA, Anderson EM, Cheung MK, Baker J, Husain A, Teng NN, Kong CS, Negrin RS. 2007. A novel technique for the enrichment of primary ovarian cancer cells. American Journal of Obstetrics and Gynecology. 197(5):507.e1-507.e5. https://doi.org/10.1016/j.ajog.2007.05.006
24.
Kluin-Nelemans JC, van-Helden HP. 1980. Non-lymphoid cells obtained by the Böyum technique and their significance in cancer patients. J Clin Lab Immunol. 4(2):99-102.
25.
Minami R, Yokota S, Teplitz RL. 1978. Gradient separation of normal and malignant cells. II. Application to in vivo tumor diagnosis. Acta Cytol. 22(6):584-8.
26.
Chang H, Jones OW, Bradshaw C, Sarkar S, Porreco RP. 1981. Enhancement of human amniotic cell growth by ficoll-paque gradient fractionation. In Vitro. 17(1):81-90. https://doi.org/10.1007/bf02618035
27.
Kekarainen T, Mannelin S, Laine J, Jaatinen T. 2006. BMC Cell Biol. 7(1):30. https://doi.org/10.1186/1471-2121-7-30
28.
Briquet A, Dubois S, Bekaert S, Dolhet M, Beguin Y, Gothot A. 2010. Prolonged ex vivo culture of human bone marrow mesenchymal stem cells influences their supportive activity toward NOD/SCID-repopulating cells and committed progenitor cells of B lymphoid and myeloid lineages. Haematologica. 95(1):47-56. https://doi.org/10.3324/haematol.2009.008524
29.
Malanga D, Barba P, Harris PE, Maffei A, Del Pozzo G. 2007. The active translation of MHCII mRNA during dendritic cells maturation supplies new molecules to the cell surface pool. Cellular Immunology. 246(2):75-80. https://doi.org/10.1016/j.cellimm.2007.06.003
30.
Ciccocioppo R, Ricci G, Rovati B, Pesce I, Mazzocchi S, Piancatelli D, Cagnoni A, Millimaggi D, Danova M, Corazza GR. Reduced number and function of peripheral dendritic cells in coeliac disease. 149(3):487-496. https://doi.org/10.1111/j.1365-2249.2007.03431.x
31.
Hattar K, van Bürck S, Bickenbach A, Grandel U, Maus U, Lohmeyer J, Csernok E, Hartung T, Seeger W, Grimminger F, et al. 2005. Anti-proteinase 3 antibodies (c-ANCA) prime CD14-dependent leukocyte activation. Journal of Leukocyte Biology. 78(4):992-1000. https://doi.org/10.1189/jlb.0902442
32.
Lubin I, Faktorowich Y, Lapidot T, Gan Y, Eshhar Z, Gazit E, Levite M, Reisner Y. 1991. Engraftment and development of human T and B cells in mice after bone marrow transplantation. Science. 252(5004):427-431. https://doi.org/10.1126/science.1826797
33.
Flaherty MP, Abdel-Latif A, Li Q, Hunt G, Ranjan S, Ou Q, Tang X, Johnson RK, Bolli R, Dawn B. 2008. Noncanonical Wnt11 Signaling Is Sufficient to Induce Cardiomyogenic Differentiation in Unfractionated Bone Marrow Mononuclear Cells. Circulation. 117(17):2241-2252. https://doi.org/10.1161/circulationaha.107.741066
34.
Kawka DW, Ouellet M, Hétu P, Singer II, Riendeau D. 2007. Double-label expression studies of prostacyclin synthase, thromboxane synthase and COX isoforms in normal aortic endothelium. Biochimica et Biophysica Acta (BBA) - Molecular and Cell Biology of Lipids. 1771(1):45-54. https://doi.org/10.1016/j.bbalip.2006.09.015
35.
Zhang Y, Lin H, Frimberger D, Epstein RB, Kropp BP. 2005. Growth of bone marrow stromal cells on small intestinal submucosa: an alternative cell source for tissue engineered bladder. BJU Int. 96(7):1120-1125. https://doi.org/10.1111/j.1464-410x.2005.05741.x
36.
Yang G, Qiu C, Zhao H, Liu Q, Shao Y. 2006. Expression of mRNA for multiple serotonin (5-HT) receptor types/subtypes by the peripheral blood mononuclear cells of rhesus macaques. Journal of Neuroimmunology. 178(1-2):24-29. https://doi.org/10.1016/j.jneuroim.2006.05.016
37.
Xu R, Jiang X, Guo Z, Chen J, Zou Y, Ke Y, Zhang S, Li Z, Cai Y, Du M, et al. 2008. Functional Analysis of Neuron-like Cells Differentiated from Neural Stem Cells Derived from Bone Marrow Stroma Cells in vitro. Cell Mol Neurobiol. 28(4):545-558. https://doi.org/10.1007/s10571-007-9174-9
38.
Pearson TW, Roelants GE, Lundin LB, Mayor-Withey KS. 1979. The bovine lymphoid system: Binding and stimulation of peripheral blood lymphocytes by lectins. Journal of Immunological Methods. 26(3):271-282. https://doi.org/10.1016/0022-1759(79)90252-7
39.
Yang TJ, Jantzen PA, Williams LF. 1979. Acid alpha-naphthyl acetate esterase: presence of activity in bovine and human T and B lymphocytes. Immunology. 38(1):85–93.
40.
Di Cesare S, Maloney S, Fernandes BF, Martins C, Marshall J, Antecka E, Odashiro AN, Dawson WW, Burnier MN. 2009. The effect of blue light exposure in an ocular melanoma animal model. J Exp Clin Cancer Res. 28(1): https://doi.org/10.1186/1756-9966-28-48
41.
Hueso P, Rocha M. 1978. Comparative study of six methods for lymphocyte isolation from several mammalian sources and determination of their carbohydrate composition. Rev Esp Fisiol. 34(3):339-44.
42.
Li X, Zhong Z, Liang S, Wang X, Zhong F. 2009. Effect of cryopreservation on IL-4, IFN? and IL-6 production of porcine peripheral blood lymphocytes. Cryobiology. 59(3):322-326. https://doi.org/10.1016/j.cryobiol.2009.09.004
43.
Blaxhall PC. 1981. A comparison of methods used for the separation of fish lymphocytes. J Fish Biology. 18(2):177-181. https://doi.org/10.1111/j.1095-8649.1981.tb02812.x
44.
Brandslund I, Rasmussen JM, Fisker D, Svehag S. 1982. Separation of human peripheral blood monocytes on continuous density gradients of polyvinylpyrrolidone-coated silica gel (Percoll®). Journal of Immunological Methods. 48(2):199-211. https://doi.org/10.1016/0022-1759(82)90194-6
45.
Feucht H, Hadam M, Frank F, Riethmüller G. 1980. Efficient separation of human T lymphocytes from venous blood using PVP-coated colloidal silica particles (Percoll). Journal of Immunological Methods. 38(1-2):43-51. https://doi.org/10.1016/0022-1759(80)90329-4
46.
Mizobe F, Martial E, Colby-Germinario S, Livett BG. 1982. An improved technique for the isolation of lymphocytes from small volumes of peripheral mouse blood. Journal of Immunological Methods. 48(3):269-279. https://doi.org/10.1016/0022-1759(82)90327-1
47.
Sato J, Kawano Y, Takaue Y, Hirao A, Makimoto A, Okamoto Y, Abe T, Kuroda Y, Shimokawa T, Iwai A. 1995. Quantitative and qualitative comparative analysis of gradient‐separated hematopoietic cells from cord blood and chemotherapy‐mobilized peripheral blood. Stem Cells. 13(5):548-555. https://doi.org/10.1002/stem.5530130513
48.
EU GMP Annex 1: Manufacture of Sterile Medicinal Products. [Internet].[updated 10 Jan 2008]. Available from: https://www.gmp-compliance.org/guidemgr/files/annex%2001[2008].pdf
49.
<1043> ANCILLARY MATERIALS FOR CELL, GENE, AND TISSUE-ENGINEERED PRODUCTS. [Internet]. Available from: https://www.drugfuture.com/Pharmacopoeia/USP32/pub/data/v32270/usp32nf27s0_c1043
50.
Leene W, Roholl PJM, Hoeben KA. 1982. Lymphocyte Differentiation in the Rabbit Thymus.319-325. https://doi.org/10.1007/978-1-4684-9066-4_44
51.
BOYUM A, LOVHAUG D, TRESLAND L, NORDLIE EM. 1991. Separation of Leucocytes: Improved Cell Purity by Fine Adjustments of Gradient Medium Density and Osmolality. Scand J Immunol. 34(6):697-712. https://doi.org/10.1111/j.1365-3083.1991.tb01594.x
52.
Archambault D, Morin G, Elazhary M. 1988. Isolation of bovine colostral lymphocytes: in vitro blastogenic responsiveness to concanavalin A and bovine rotavirus. Ann Rech Vet. 19(3):169-74.
53.
Van Riper G, Siciliano S, Fischer PA, Meurer R, Springer MS, Rosen H. 1993. Characterization and species distribution of high affinity GTP-coupled receptors for human rantes and monocyte chemoattractant protein 1.. 177(3):851-856. https://doi.org/10.1084/jem.177.3.851
54.
Chin S, Poey AC, Wong C, Chang S, Teh W, Mohr TJ, Cheong S. 2010. Cryopreserved mesenchymal stromal cell treatment is safe and feasible for severe dilated ischemic cardiomyopathy. Cytotherapy. 12(1):31-37. https://doi.org/10.3109/14653240903313966
55.
Garayoa M, Garcia JL, Santamaria C, Garcia-Gomez A, Blanco JF, Pandiella A, Hernández JM, Sanchez-Guijo FM, del Cañizo M, Gutiérrez NC, et al. 2009. Mesenchymal stem cells from multiple myeloma patients display distinct genomic profile as compared with those from normal donors. Leukemia. 23(8):1515-1527. https://doi.org/10.1038/leu.2009.65
56.
Grisendi G, Annerén C, Cafarelli L, Sternieri R, Veronesi E, Cervo GL, Luminari S, Maur M, Frassoldati A, Palazzi G, et al. 2010. GMP-manufactured density gradient media for optimized mesenchymal stromal/stem cell isolation and expansion. Cytotherapy. 12(4):466-477. https://doi.org/10.3109/14653241003649510
57.
Brooke G, Rossetti T, Pelekanos R, Ilic N, Murray P, Hancock S, Antonenas V, Huang G, Gottlieb D, Bradstock K, et al. 2009. Manufacturing of human placenta-derived mesenchymal stem cells for clinical trials. 144(4):571-579. https://doi.org/10.1111/j.1365-2141.2008.07492.x
58.
Miltenyi S, Müller W, Weichel W, Radbruch A. 1990. High gradient magnetic cell separation with MACS. Cytometry. 11(2):231-238. https://doi.org/10.1002/cyto.990110203
59.
Mauldin JP, Nagelin MH, Wojcik AJ, Srinivasan S, Skaflen MD, Ayers CR, McNamara CA, Hedrick CC. 2008. Reduced Expression of ATP-Binding Cassette Transporter G1 Increases Cholesterol Accumulation in Macrophages of Patients With Type 2 Diabetes Mellitus. Circulation. 117(21):2785-2792. https://doi.org/10.1161/circulationaha.107.741314
60.
Ali H, Jurga M, Kurgonaite K, Forraz N, McGuckin C. 2009. Defined serum-free culturing conditions for neural tissue engineering of human cord blood stem cells. Acta Neurobiol Exp (Wars). 69(1):12-23.
61.
Figueroa-Tentori D, Querol S, Dodi IA, Madrigal A, Duggleby R. 2008. High purity and yield of natural Tregs from cord blood using a single step selection method. Journal of Immunological Methods. 339(2):228-235. https://doi.org/10.1016/j.jim.2008.09.019
Sign In To Continue

To continue reading please sign in or create an account.

Don't Have An Account?