Improved Human Mesenchymal Stem Cells Isolation Tzu

advertisement
Improved Human Mesenchymal Stem Cells Isolation
Tzu-Min Chan1,2, Horng-Jyh Harn3,4, Hui-Ping Lin5, Pei-Wen Chou2,6, Julia Yi-Ru
Chen6, Hong-Meng Chuang2,4, Tzyy-Wen Chiou7, Bing-Chiang Liang8,9, Shinn-Zong
Lin1,10,11,12*
1
2
3
Neuropsychiatry Center, China Medical University Hospital, Taichung, Taiwan, ROC
Everfront Biotech Inc., New Taipei City, Chinese Taipei, Taiwan, ROC
Department of Pathology, China Medical University Hospital, Taichung, Taiwan,
ROC
4
Department of Medicine, China Medical University, Taichung, Taiwan, ROC
5
Institute of Cellular and System Medicine, National Health Research Institutes,
Miaoli ,Taiwan, ROC
6
Guang Li Biomedicine, Inc., New Taipei City, Chinese Taipei, Taiwan, ROC
7
Department of Life Science and Graduate Institute of Biotechnology, National Dong
Hwa University, Hualien, Taiwan, ROC
8
Department of Emergency Medicine, Taipei Veterans General Hospital, Taiwan,
ROC
9
National Yang-Ming University School of Medicine, Taiwan, ROC
10
Department of Neurosurgery, China Medical University Beigan Hospital, Yunlin,
Taiwan, ROC
11
Department of Neurosurgery, Tainan Municipal An-Nan Hospital-China Medical
University, Tainan, Taiwan, ROC
12
Graduate Institute of Immunology, China Medical University, Taichung, Taiwan,
ROC
Address correspondence to Prof. Shinn-Zong Lin, MD., PhD.
*
E-mail: shinnzong@yahoo.com.tw
key word: Human mesenchymal stem cells (hMSCs); improved isolation of hMSCs;
Vascular perfusion for liver progenitors; Mechanical dissociation lipoaspirate;
Isolation adipose-derived cell from blood/saline phase; Novel marrow filter device;
Clot Spots method; Stimulation BMMSCs mobilized into peripheral blood
ABSTRACT
Human mesenchymal stem cells (hMSCs) are currently available range of
applications and show much benefits to become a good material for regenerative
medicine, tissue engineering and disease therapy. Before ex vivo expansion, isolating
and characterizing primary hMSCs from peripheral tissues are the key steps for
obtaining adequate materials to clinical application. The proportion of peripheral stem
cells is very low in surrounding tissues or organs, thus, recovery ratio and quality will
be limiting factors. In this review, we summarized current common methods used to
isolate peripheral stem cells, and what new insights revealed to improve amount of
stem cells and their stemness. These strategies offered alternative ways to acquire
hMSCs in convenient and/or effective manners, which are important for commercial
purposes. Later mass-amplify primary hMSCs procedures, ensuring their stemness
until differentiate into tissue types cell for clinical usage. Enlarged qualified hMSCs
were more clinical applicability for therapeutic transplants, which may help people
live longer and better.
INTRODUCTION
Mesenchyme are derived from embryonic mesodermal progenitors, and we could
isolate hMSCs from tissues, such as bone marrow (BM), umbilical cord blood,
adipose, muscle, corneal stroma, tooth bud and so on (15,20,30,35,48,58,62,83). The
advantages of hMSCs, extracted from the individual, compared to general human
embryonic stem cells (hES) lines, having lower of immunorejection, pathogen
transmission and tumorigenesis (61,79,100). Besides, hMSCs can be handed easily in
their proliferation potential, and permitting differentiation into kinds of cell types
(74,86). MSCs are retained multipotent ability, which can proliferate into osteoblasts,
chondrocytes, adipocytes, neuron and hepatocyte like cells, and they have self-renew
capacity (50,58,64,68,97).
Expanding knowledge on salient features of hMSCs in regenerative and
immunosuppressive properties, which provided new strategies into clinical
applications (2,12,60,83). hMSCs have been largely used as potential therapeutic
strategies for diseases, involving hepatology, cardiology, neurology, orthopedics,
pancreatic, rheumatology and so on (14,27,51,60,67,79,86,105,108). hMSCs mainly
function as supplier to repair degenerative or defective organs (11,34). Combining
cures have been applied by treating with small molecular drugs, for example, valproic
acid (VPA) that stimulates stem cell proliferation and self-renewal (8,24,77). For
targeting deliver system, hMSCs have been considered to be the attractive vehicles to
carry therapeutic agents and effectively release them toward various tumor diseases
(1,36,57,84). In the neuron degenerative disease or injury issues, especially in
Parkinson's disease, amyotrophic lateral sclerosis, stroke and Alzheimer's disease,
stem cell-mediated transfer were widely used for gene therapy (5,28,47,56).
Regarding to transplant medicine, there are basic technologies in developing
separation from tissues and operating above these cells, but these are the key
procedures to success (6,102). hMSCs content are only 0.01 to 0.001% nucleated cells,
thus, isolation technologies become the important steps for clinical applications
(66,73). It would be an limited element about inadequate amounts and stemness of
hMSCs for clinical therapy; therefore, improved isolation of the stem cells is desired
(28,106). Clinical applications expected to be able to find a suitable in vitro isolation
conditions, to resolve the problem of limited cells and stemness maintaining. In this
review, we focused on issues that have been considered when manipulating hMSCs
techniques in varying isolation procedures. Further, we summarized improved
methods and new insights, theses may help people to improve hMSCs isolation and
provide more materials for transplant medicine.
ESTABLISHMENT OF hMSCs
hMSCs can be harvested in a relatively less invasive manner and easily isolated
from peripheral organs, and maintained their multipotentiality during passages
(15,72,88). It was worth noting the efficiency and quality of isolated hMSCs have
variable effects in different mediums, procedures, temperatures, and oxygen tension
(13,107). The simple method has been established for different kinds of hMSCs,
which is mainly relayed on their adherence properties to achieve isolation (21,45).
After plating of multipotent stem cells, extracted from umbilical cord and BM, most
of non-hMSCs could be separated through continuous cultures (81). For tissue types,
adipose and liver, of hMSC isolation were additional digested with collagenase
(38,54).
Following immunophenotypic analysis are necessary for identification above
adherent cells, however, to further separate non-multipotent or non-conformance cells
through their specific surface markers (59,76). It would be utmost importance to
identify hMSC lineage, and can easily obtainable to verify them through flow
cytometry (66,95). Although hMSCs were no confidence markers have been
consensus, the data showed that they still have some common cell surface markers. A
minimal phenotypic pattern requires expression of CD73, CD90, and CD105, but
shown negatively in CD34, CD45, HLA-DR and other molecules (43,66,76). For
specific immunophenotypic patterns, the variety tissues source of peripheral stem cell
could isolated by their lineage specific surface markers were summarized in Table 1
(68,109).
IMPROVED ISOLATION
Separating methods from tissues have been established, however, exist challenges
faced by investigators are that the primary hMSCs were decreased clinical
applicability (43,55). Obtaining primary hMSCs completely and rapidly, to explore a
optimized method for isolation and purification, have become a prerequisite for ex
vivo expansion (41). Improvement sections include lower segregation ratio of hMSCs,
and shorter times to maintain stem cell properties during subculture the cells (103).
Besides, the methods must be continuous developed to obtain sufficient qualities,
such as unified cell type and their multipotency (38,62). In current, improved isolation
methods have been concerned, and showed many new insights to acquire better
primary hMSCs for subsequent medical applications (25,62,97).
Vascular perfusion for liver progenitors
Traditional techniques, isolated from peripheral organs and tissues, yield only a
fewer available hMSCs and cause cell injuries with collagenase usage. This will be an
important subject to improve the quality of hMSCs, if investigators can reduce
collagenase exposure time to prevent cell from enzymatic damage (29,71).
Experimental fetal liver donated from therapeutic abortion, an alternative resource of
adult liver, have been used for isolating mesenchymal progenitor cells (49,78,93).
Further, a improved method proposed that vascular perfusion technique reduces the
exposure of the tissue to collagenase (26). The method mainly based on procurement
for perfusion via the portal vein with the adaptation of a 5-step portal vein in situ
perfusion method (26). They designed suitable solutions A to D and optimized other
conditions, to separate mesenchymal progenitor cells from liver, and cultured
hepatocytes in Williams’ E medium–based Heparmed Vito 143 (26). In contrast to the
static collagenase digestion, vascular perfusion obtained high viabilities of
mesenchymal progenitors and more cell numbers, and prolonged their stemness
(26,80).
Mechanical dissociation lipoaspirate
Methods to isolate and culture adipose-derived stem cells (ADSCs) were develop
extensively, however, little has been done to improve yields and multipotency
(3,23,38). An alternative method, mechanical dissociation, was used to isolate a
population of hMSCs from lipoaspirate without collagenase treatments. The aim of
previous study was to increase yields and cryopreservation them before ex vivo
expansion, maintaining multipotency, for therapeutic application. Adipose tissue
samples were incubate with ACK buffer solution, and then shake for preliminary red
blood cell lysis (3). Following subsequently culture to select adherent cells, and
isolate ADSCs through flow cytometry analysis. They further evidenced that treated
lipoaspirate samples can be stored without damage to ADSCs during mechanical
dissociation procedure at 4 ℃ (3). Mechanical dissociation offer a convenient
isolation method and allow large volume of adipose separation, which are more
suitable for commercial purposes (3,4,46,103).
Isolation adipose-derived cell from blood/saline phase
Compared with original isolation of ADSCs from adipose, spending 8-10 hour of
continuous intense effort in usually, another rapid separation method revealed in less
than 30 minutes (19,110). The basic principle relies on obtaining ADSCs from
blood/saline phase, containing rich adipose-derived cell due to perivascular origin,
easily than oil phase of adipose extracts (10,85). Defined simple 5-step process could
isolate ADSCs from more buoyant adipose tissue, and show a mesenchymal
morphology and immunophenotype (19). The method provides time-saving and
simple technique for ADSCs preparing, is critical for advancing transplant medicine
therapeutics.
Novel marrow filter device
A novel BM filter device has been explored to improve isolation methods, which
collects nucleated cells without continuous gradient centrifugation (70). The mainly
procedures are filtered BM deliver into the device and connect with a designed
nonwoven fabric filter in a closed system. Most of nucleated cells could be separated
through saline flow washed, removing red blood cells (RBCs) and platelets (PLTs),
and harvested in defined collection medium (70). Such closed workflow offered a
rapid purification duration times, and prevented a risk of contamination from
exposure operation. Besides, it permits decreased operator factors, influencing the cell
yields and qualities, and establish a standard protocol for clinical cell therapy trials
(31-33,69). It would be represent a major advance in BM by increasing the recovery
ratio, allowing for less ex vivo expansion, from freshly harvested peripheral stem cell
tissues (31,70).
Clot Spots method
Wharton's jelly mesenchymal stem cells (WJ-MSCs) within human umbilical cord,
is a noncontroversial source of hematopoietic stem cells (HSCs), which frequently
used in transplant medicine (18,65). The properties of WJ-MSCs considered
comparable with fetal rather than adult-derived hMSCs, thus, showed more
proliferative and immunosuppressive for therapeutically active (46). For more suitable
as a clinical application, a new approach displayed a improvement in isolating
WJ-MSCs and recognized it was a good source (39). Different to Rosset Sep method,
Clot Spot method use mesencult complete medium to culture primary human
umbilical cord blood (HUCB) (39,52). Semi-solid cord blood clots were explanted on
medium without disturbing the blood clots. Further, sub-cultured for adherent cells
selection, and then morphology identification and immunophenotyping. Compared to
Rosset Sep method, Clot Spot method demonstrated 3-fold increase of hMSCs from
WJ-MSCs (39).
Stimulation BMMSCs mobilized into peripheral blood
Since bone marrow mesenchymal stem cells (BMMSCs) found in the interior of
bones, are the most common source of hMSCs, the limitations are painful harvest and
surgical risks (18,104). To prevent invasive surgery, the in vivo mobilization of
BMMSCs into bloods seems a alternative method to harvest stem cells for
transplantation (44). Fibrin microbeads (FMB) could bind mononuclear cells and
isolate hMSCs from human peripheral blood that were mobilized with a granulocyte
colony-stimulating factor (G-CSF). G-CSF would alter homing niches in BM through
reduced VCAM-1, SDF-1, and SCF expression, caused marked down-regulation of
adhesion and released HSCs from BM into peripheral blood (53,99). This
mobilization procedure separates efficiently in HSCs isolation, and show significantly
lower contamination by other cell types (44,89). Advanced method were performed in
combinational treatment of G-CSF with plerixafor, reversibly blocks SDF-1 binding
to CXCR4, which can improve the collection of HSCs compared with G-CSF alone
(53,92).
CONCLUSION
To hMSCs used in regenerative medicine, it must be established the basis of core
technologies for preparing large amounts of suitable stem cells (87). With these key
technologies capabilities in order to provide an endless supply of stem cells, to
actually applied in cell therapy (9,74). The technologies involve acquiring source
from the peripheral tissues, isolation multipotent cells and further ex vivo expansion
(Fig 1) (74). However, enhancing isolation methods may be a critical issue, due to
inadequate source of peripheral stem cells, to acquire higher qualities of hMSCs
(3,43). In summary, improved separation technologies have following features: (1)
prevent potential contaminations during manipulation. (2) enhance recovery ratio of
hMSCs from limited source. (3) improve maintaining time of stemness during
passages. (4) make the procedures become more convenient and less cost, which are
important for commercialized purpose. Taking together, isolation strategies of hMSCs
were toward to increase the clinical applicability by improving primary stem cells to
reduce ex vivo expansion (Fig 1).
ACKNOWLEDGMENTS
Stem Cell and Regeneration Medicine Foundation, NSC 99-2320-B-039-008-MY3
and Everfront Biotech Inc.
REFERENCE
1.
2.
3.
4.
5.
6.
7.
8.
9.
Ahmed, A. U.; Tyler, M. A.; Thaci, B.; Alexiades, N. G.; Han, Y.; Ulasov, I. V.;
Lesniak, M. S. A comparative study of neural and mesenchymal stem
cell-based carriers for oncolytic adenovirus in a model of malignant glioma.
Mol Pharm 8(5):1559-1572; 2011.
Ayatollahi, M.; Salmani, M. K.; Geramizadeh, B.; Tabei, S. Z.; Soleimani, M.;
Sanati, M. H. Conditions to improve expansion of human mesenchymal stem
cells based on rat samples. World J Stem Cells 4(1):1-8; 2012.
Baptista, L. S.; do Amaral, R. J.; Carias, R. B.; Aniceto, M.; Claudio-da-Silva, C.;
Borojevic, R. An alternative method for the isolation of mesenchymal stromal
cells derived from lipoaspirate samples. Cytotherapy 11(6):706-715; 2009.
Bertolo, A.; Mehr, M.; Aebli, N.; Baur, M.; Ferguson, S. J.; Stoyanov, J. V.
Influence of different commercial scaffolds on the in vitro differentiation of
human mesenchymal stem cells to nucleus pulposus-like cells. Eur Spine J 21
Suppl 6:S826-838; 2012.
Borlongan, C. V. Recent preclinical evidence advancing cell therapy for
Alzheimer's disease. Exp Neurol 237(1):142-146; 2012.
Boulter, L.; Lu, W. Y.; Forbes, S. J. Differentiation of progenitors in the liver: a
matter of local choice. J Clin Invest 123(5):1867-1873; 2013.
Boxall, S. A.; Jones, E. Markers for characterization of bone marrow
multipotential stromal cells. Stem Cells Int 2012:975871; 2012.
Bug, G.; Gul, H.; Schwarz, K.; Pfeifer, H.; Kampfmann, M.; Zheng, X.; Beissert,
T.; Boehrer, S.; Hoelzer, D.; Ottmann, O. G. and others. Valproic acid stimulates
proliferation and self-renewal of hematopoietic stem cells. Cancer Res
65(7):2537-2541; 2005.
Capelli, C.; Domenghini, M.; Borleri, G.; Bellavita, P.; Poma, R.; Carobbio, A.;
Mico, C.; Rambaldi, A.; Golay, J.; Introna, M. Human platelet lysate allows
expansion and clinical grade production of mesenchymal stromal cells from
small samples of bone marrow aspirates or marrow filter washouts. Bone
Marrow Transplant 40(8):785-791; 2007.
10.
11.
12.
13.
14.
15.
16.
17.
18.
19.
20.
Crisan, M.; Yap, S.; Casteilla, L.; Chen, C. W.; Corselli, M.; Park, T. S.; Andriolo,
G.; Sun, B.; Zheng, B.; Zhang, L. and others. A perivascular origin for
mesenchymal stem cells in multiple human organs. Cell Stem Cell
3(3):301-313; 2008.
Daadi, M. M.; Hu, S.; Klausner, J.; Li, Z.; Sofilos, M.; Sun, G.; Wu, J. C.;
Steinberg, G. K. Imaging Neural Stem Cell Graft-Induced Structural Repair in
Stroke. Cell Transplant; 2012.
de Lima, M.; McNiece, I.; Robinson, S. N.; Munsell, M.; Eapen, M.; Horowitz,
M.; Alousi, A.; Saliba, R.; McMannis, J. D.; Kaur, I. and others. Cord-blood
engraftment with ex vivo mesenchymal-cell coculture. N Engl J Med
367(24):2305-2315; 2012.
Delalat, B.; Pourfathollah, A. A.; Soleimani, M.; Mozdarani, H.; Ghaemi, S. R.;
Movassaghpour, A. A.; Kaviani, S. Isolation and ex vivo expansion of human
umbilical cord blood-derived CD34+ stem cells and their cotransplantation
with or without mesenchymal stem cells. Hematology 14(3):125-132; 2009.
Deng, J.; Petersen, B. E.; Steindler, D. A.; Jorgensen, M. L.; Laywell, E. D.
Mesenchymal stem cells spontaneously express neural proteins in culture and
are neurogenic after transplantation. Stem Cells 24(4):1054-1064; 2006.
Ding, D. C.; Shyu, W. C.; Lin, S. Z. Mesenchymal stem cells. Cell Transplant
20(1):5-14; 2011.
Djouad, F.; Bony, C.; Haupl, T.; Uze, G.; Lahlou, N.; Louis-Plence, P.; Apparailly,
F.; Canovas, F.; Reme, T.; Sany, J. and others. Transcriptional profiles
discriminate bone marrow-derived and synovium-derived mesenchymal stem
cells. Arthritis Res Ther 7(6):R1304-1315; 2005.
Dominici, M.; Le Blanc, K.; Mueller, I.; Slaper-Cortenbach, I.; Marini, F.; Krause,
D.; Deans, R.; Keating, A.; Prockop, D.; Horwitz, E. Minimal criteria for defining
multipotent mesenchymal stromal cells. The International Society for Cellular
Therapy position statement. Cytotherapy 8(4):315-317; 2006.
Fong, C. Y.; Gauthaman, K.; Cheyyatraivendran, S.; Lin, H. D.; Biswas, A.;
Bongso, A. Human umbilical cord Wharton's jelly stem cells and its
conditioned medium support hematopoietic stem cell expansion ex vivo. J
Cell Biochem 113(2):658-668; 2012.
Francis, M. P.; Sachs, P. C.; Elmore, L. W.; Holt, S. E. Isolating adipose-derived
mesenchymal stem cells from lipoaspirate blood and saline fraction.
Organogenesis 6(1):11-14; 2010.
Friedenstein, A. J.; Chailakhjan, R. K.; Lalykina, K. S. The development of
fibroblast colonies in monolayer cultures of guinea-pig bone marrow and
spleen cells. Cell Tissue Kinet 3(4):393-403; 1970.
21.
22.
23.
24.
25.
26.
27.
28.
29.
30.
31.
Friedenstein, A. J.; Gorskaja, J. F.; Kulagina, N. N. Fibroblast precursors in
normal and irradiated mouse hematopoietic organs. Exp Hematol
4(5):267-274; 1976.
Gauthaman, K.; Fong, C. Y.; Subramanian, A.; Biswas, A.; Bongso, A. ROCK
inhibitor Y-27632 increases thaw-survival rates and preserves stemness and
differentiation potential of human Wharton's jelly stem cells after
cryopreservation. Stem Cell Rev 6(4):665-676; 2010.
Gimble, J.; Guilak, F. Adipose-derived adult stem cells: isolation,
characterization, and differentiation potential. Cytotherapy 5(5):362-369;
2003.
Gottlicher, M.; Minucci, S.; Zhu, P.; Kramer, O. H.; Schimpf, A.; Giavara, S.;
Sleeman, J. P.; Lo Coco, F.; Nervi, C.; Pelicci, P. G. and others. Valproic acid
defines a novel class of HDAC inhibitors inducing differentiation of
transformed cells. EMBO J 20(24):6969-6978; 2001.
Greinix, H. T.; Worel, N. New agents for mobilizing peripheral blood stem cells.
Transfus Apher Sci 41(1):67-71; 2009.
Gridelli, B.; Vizzini, G.; Pietrosi, G.; Luca, A.; Spada, M.; Gruttadauria, S.;
Cintorino, D.; Amico, G.; Chinnici, C.; Miki, T. and others. Efficient human fetal
liver cell isolation protocol based on vascular perfusion for liver cell-based
therapy and case report on cell transplantation. Liver Transpl 18(2):226-237;
2012.
Harn, H. J.; Lin, S. Z.; Hung, S. H.; Subeq, Y. M.; Li, Y. S.; Syu, W. S.; Ding, D. C.;
Lee, R. P.; Hsieh, D. K.; Lin, P. C. and others. Adipose-derived stem cells can
abrogate chemical-induced liver fibrosis and facilitate recovery of liver
function. Cell Transplant 21(12):2753-2764; 2012.
Hefferan, M. P.; Galik, J.; Kakinohana, O.; Sekerkova, G.; Santucci, C.; Marsala,
S.; Navarro, R.; Hruska-Plochan, M.; Johe, K.; Feldman, E. and others. Human
neural stem cell replacement therapy for amyotrophic lateral sclerosis by
spinal transplantation. PLoS One 7(8):e42614; 2012.
Herrera, M. B.; Bruno, S.; Buttiglieri, S.; Tetta, C.; Gatti, S.; Deregibus, M. C.;
Bussolati, B.; Camussi, G. Isolation and characterization of a stem cell
population from adult human liver. Stem Cells 24(12):2840-2850; 2006.
Ho, J. H.; Ma, W. H.; Tseng, T. C.; Chen, Y. F.; Chen, M. H.; Lee, O. K. Isolation
and characterization of multi-potent stem cells from human orbital fat tissues.
Tissue Eng Part A 17(1-2):255-266; 2011.
Horwitz, E. M.; Gordon, P. L.; Koo, W. K.; Marx, J. C.; Neel, M. D.; McNall, R. Y.;
Muul, L.; Hofmann, T. Isolated allogeneic bone marrow-derived mesenchymal
cells engraft and stimulate growth in children with osteogenesis imperfecta:
32.
33.
34.
35.
36.
37.
38.
39.
40.
41.
Implications for cell therapy of bone. Proc Natl Acad Sci U S A
99(13):8932-8937; 2002.
Horwitz, E. M.; Prockop, D. J.; Fitzpatrick, L. A.; Koo, W. W.; Gordon, P. L.; Neel,
M.; Sussman, M.; Orchard, P.; Marx, J. C.; Pyeritz, R. E. and others.
Transplantability and therapeutic effects of bone marrow-derived
mesenchymal cells in children with osteogenesis imperfecta. Nat Med
5(3):309-313; 1999.
Horwitz, E. M.; Prockop, D. J.; Gordon, P. L.; Koo, W. W.; Fitzpatrick, L. A.; Neel,
M. D.; McCarville, M. E.; Orchard, P. J.; Pyeritz, R. E.; Brenner, M. K. Clinical
responses to bone marrow transplantation in children with severe
osteogenesis imperfecta. Blood 97(5):1227-1231; 2001.
Hsiao, L. C.; Carr, C.; Chang, K. C.; Lin, S. Z.; Clarke, K. Stem cell-based therapy
for ischemic heart disease. Cell Transplant 22(4):663-675; 2013.
Hu, W.; Ye, Y.; Wang, J.; Zhang, W.; Chen, A.; Guo, F. Bone morphogenetic
proteins induce rabbit bone marrow-derived mesenchyme stem cells to
differentiate into osteoblasts via BMP signals pathway. Artif Cells Nanomed
Biotechnol; 2013.
Hu, Y. L.; Huang, B.; Zhang, T. Y.; Miao, P. H.; Tang, G. P.; Tabata, Y.; Gao, J. Q.
Mesenchymal stem cells as a novel carrier for targeted delivery of gene in
cancer therapy based on nonviral transfection. Mol Pharm 9(9):2698-2709;
2012.
Huang, G. T.; Gronthos, S.; Shi, S. Mesenchymal stem cells derived from dental
tissues vs. those from other sources: their biology and role in regenerative
medicine. J Dent Res 88(9):792-806; 2009.
Huang, S. J.; Fu, R. H.; Shyu, W. C.; Liu, S. P.; Jong, G. P.; Chiu, Y. W.; Wu, H. S.;
Tsou, Y. A.; Cheng, C. W.; Lin, S. Z. Adipose-derived stem cells: isolation,
characterization, and differentiation potential. Cell Transplant 22(4):701-709;
2013.
Hussain, I.; Magd, S. A.; Eremin, O.; El-Sheemy, M. New approach to isolate
mesenchymal stem cell (MSC) from human umbilical cord blood. Cell Biol Int
36(7):595-600; 2012.
Jankowski, R. J.; Deasy, B. M.; Huard, J. Muscle-derived stem cells. Gene Ther
9(10):642-647; 2002.
Jia, G. Q.; Zhang, M. M.; Yang, P.; Cheng, J. Q.; Lu, Y. R.; Wu, X. T. [Effects of the
different culture and isolation methods on the growth, proliferation and
biology characteristics of rat bone marrow mesenchymal stem cells]. Sichuan
Da Xue Xue Bao Yi Xue Ban 40(4):719-723; 2009.
42.
43.
44.
45.
Johnstone, B.; Hering, T. M.; Caplan, A. I.; Goldberg, V. M.; Yoo, J. U. In vitro
chondrogenesis of bone marrow-derived mesenchymal progenitor cells. Exp
Cell Res 238(1):265-272; 1998.
Jung, S.; Panchalingam, K. M.; Rosenberg, L.; Behie, L. A. Ex vivo expansion of
human mesenchymal stem cells in defined serum-free media. Stem Cells Int
2012:123030; 2012.
Kassis, I.; Zangi, L.; Rivkin, R.; Levdansky, L.; Samuel, S.; Marx, G.; Gorodetsky,
R. Isolation of mesenchymal stem cells from G-CSF-mobilized human
peripheral blood using fibrin microbeads. Bone Marrow Transplant
37(10):967-976; 2006.
Kastrinaki, M. C.; Andreakou, I.; Charbord, P.; Papadaki, H. A. Isolation of
human bone marrow mesenchymal stem cells using different membrane
markers: comparison of colony/cloning efficiency, differentiation potential,
and molecular profile. Tissue Eng Part C Methods 14(4):333-339; 2008.
46.
47.
48.
49.
50.
51.
52.
53.
Kim, D. W.; Staples, M.; Shinozuka, K.; Pantcheva, P.; Kang, S. D.; Borlongan, C.
V. Wharton's Jelly-Derived Mesenchymal Stem Cells: Phenotypic
Characterization and Optimizing Their Therapeutic Potential for Clinical
Applications. Int J Mol Sci 14(6):11692-11712; 2013.
Kim, S. U.; Lee, H. J.; Kim, Y. B. Neural stem cell-based treatment for
neurodegenerative diseases. Neuropathology; 2013.
Koga, H.; Muneta, T.; Nagase, T.; Nimura, A.; Ju, Y. J.; Mochizuki, T.; Sekiya, I.
Comparison of mesenchymal tissues-derived stem cells for in vivo
chondrogenesis: suitable conditions for cell therapy of cartilage defects in
rabbit. Cell Tissue Res 333(2):207-215; 2008.
Krishna, K. A.; Krishna, K. S.; Berrocal, R.; Tummala, A.; Rao, K. S.; Rao, K. R. A
review on the therapeutic potential of embryonic and induced pluripotent
stem cells in hepatic repair. J Nat Sci Biol Med 2(2):141-144; 2011.
Lee, K. D. Applications of mesenchymal stem cells: an updated review. Chang
Gung Med J 31(3):228-236; 2008.
Lee, K. D.; Kuo, T. K.; Whang-Peng, J.; Chung, Y. F.; Lin, C. T.; Chou, S. H.; Chen,
J. R.; Chen, Y. P.; Lee, O. K. In vitro hepatic differentiation of human
mesenchymal stem cells. Hepatology 40(6):1275-1284; 2004.
Lee, O. K.; Kuo, T. K.; Chen, W. M.; Lee, K. D.; Hsieh, S. L.; Chen, T. H. Isolation
of multipotent mesenchymal stem cells from umbilical cord blood. Blood
103(5):1669-1675; 2004.
Levesque, J. P.; Takamatsu, Y.; Nilsson, S. K.; Haylock, D. N.; Simmons, P. J.
Vascular cell adhesion molecule-1 (CD106) is cleaved by neutrophil proteases
in the bone marrow following hematopoietic progenitor cell mobilization by
granulocyte colony-stimulating factor. Blood 98(5):1289-1297; 2001.
54.
55.
56.
57.
58.
59.
60.
61.
62.
63.
64.
65.
Li, D. R.; Cai, J. H. Methods of isolation, expansion, differentiating induction
and preservation of human umbilical cord mesenchymal stem cells. Chin Med
J (Engl) 125(24):4504-4510; 2012.
Li, X.; Zhang, Y.; Qi, G. Evaluation of isolation methods and culture conditions
for rat bone marrow mesenchymal stem cells. Cytotechnology 65(3):323-334;
2013.
Mackay-Sim, A. Concise review: Patient-derived olfactory stem cells: new
models for brain diseases. Stem Cells 30(11):2361-2365; 2012.
Mader, E. K.; Butler, G.; Dowdy, S. C.; Mariani, A.; Knutson, K. L.; Federspiel, M.
J.; Russell, S. J.; Galanis, E.; Dietz, A. B.; Peng, K. W. Optimizing patient derived
mesenchymal stem cells as virus carriers for a phase I clinical trial in ovarian
cancer. J Transl Med 11:20; 2013.
Mafi, R.; Hindocha, S.; Mafi, P.; Griffin, M.; Khan, W. S. Sources of adult
mesenchymal stem cells applicable for musculoskeletal applications - a
systematic review of the literature. Open Orthop J 5 Suppl 2:242-248; 2011.
Majore, I.; Moretti, P.; Hass, R.; Kasper, C. Identification of subpopulations in
mesenchymal stem cell-like cultures from human umbilical cord. Cell
Commun Signal 7:6; 2009.
Maumus, M.; Guerit, D.; Toupet, K.; Jorgensen, C.; Noel, D. Mesenchymal
stem cell-based therapies in regenerative medicine: applications in
rheumatology. Stem Cell Res Ther 2(2):14; 2011.
Mendelson, A.; Frank, E.; Allred, C.; Jones, E.; Chen, M.; Zhao, W.; Mao, J. J.
Chondrogenesis by chemotactic homing of synovium, bone marrow, and
adipose stem cells in vitro. Faseb J 25(10):3496-3504; 2011.
Mendez-Ferrer, S.; Michurina, T. V.; Ferraro, F.; Mazloom, A. R.; Macarthur, B.
D.; Lira, S. A.; Scadden, D. T.; Ma'ayan, A.; Enikolopov, G. N.; Frenette, P. S.
Mesenchymal and haematopoietic stem cells form a unique bone marrow
niche. Nature 466(7308):829-834; 2010.
Michel, M.; Torok, N.; Godbout, M. J.; Lussier, M.; Gaudreau, P.; Royal, A.;
Germain, L. Keratin 19 as a biochemical marker of skin stem cells in vivo and
in vitro: keratin 19 expressing cells are differentially localized in function of
anatomic sites, and their number varies with donor age and culture stage. J
Cell Sci 109 ( Pt 5):1017-1028; 1996.
Minteer, D.; Marra, K. G.; Rubin, J. P. Adipose-derived mesenchymal stem cells:
biology and potential applications. Adv Biochem Eng Biotechnol 129:59-71;
2013.
Moon, J. H.; Kwak, S. S.; Park, G.; Jung, H. Y.; Yoon, B. S.; Park, J.; Ryu, K. S.;
Choi, S. C.; Maeng, I.; Kim, B. and others. Isolation and characterization of
66.
67.
multipotent human keloid-derived mesenchymal-like stem cells. Stem Cells
Dev 17(4):713-724; 2008.
Nery, A. A.; Nascimento, I. C.; Glaser, T.; Bassaneze, V.; Krieger, J. E.; Ulrich, H.
Human mesenchymal stem cells: from immunophenotyping by flow
cytometry to clinical applications. Cytometry A 83(1):48-61; 2013.
Noiseux, N.; Gnecchi, M.; Lopez-Ilasaca, M.; Zhang, L.; Solomon, S. D.; Deb, A.;
Dzau, V. J.; Pratt, R. E. Mesenchymal stem cells overexpressing Akt
dramatically repair infarcted myocardium and improve cardiac function
despite infrequent cellular fusion or differentiation. Mol Ther 14(6):840-850;
2006.
68.
Orbay, H.; Tobita, M.; Mizuno, H. Mesenchymal stem cells isolated from
adipose and other tissues: basic biological properties and clinical applications.
Stem Cells Int 2012:461718; 2012.
69.
Otsuru, S.; Gordon, P. L.; Shimono, K.; Jethva, R.; Marino, R.; Phillips, C. L.;
Hofmann, T. J.; Veronesi, E.; Dominici, M.; Iwamoto, M. and others.
Transplanted bone marrow mononuclear cells and MSCs impart clinical
benefit to children with osteogenesis imperfecta through different
mechanisms. Blood 120(9):1933-1941; 2012.
Otsuru, S.; Hofmann, T. J.; Olson, T. S.; Dominici, M.; Horwitz, E. M. Improved
isolation and expansion of bone marrow mesenchymal stromal cells using a
70.
71.
72.
73.
74.
75.
novel marrow filter device. Cytotherapy 15(2):146-153; 2013.
Park, C. H.; Bae, S. H.; Kim, H. Y.; Kim, J. K.; Jung, E. S.; Chun, H. J.; Song, M. J.;
Lee, S. E.; Cho, S. G.; Lee, J. W. and others. A pilot study of autologous
CD34-depleted bone marrow mononuclear cell transplantation via the
hepatic artery in five patients with liver failure. Cytotherapy; 2013.
Pendleton, C.; Li, Q.; Chesler, D. A.; Yuan, K.; Guerrero-Cazares, H.;
Quinones-Hinojosa, A. Mesenchymal stem cells derived from adipose tissue
vs bone marrow: in vitro comparison of their tropism towards gliomas. PLoS
One 8(3):e58198; 2013.
Pittenger, M. F.; Martin, B. J. Mesenchymal stem cells and their potential as
cardiac therapeutics. Circ Res 95(1):9-20; 2004.
Pountos, I.; Corscadden, D.; Emery, P.; Giannoudis, P. V. Mesenchymal stem
cell tissue engineering: techniques for isolation, expansion and application.
Injury 38 Suppl 4:S23-33; 2007.
Qu-Petersen, Z.; Deasy, B.; Jankowski, R.; Ikezawa, M.; Cummins, J.; Pruchnic,
R.; Mytinger, J.; Cao, B.; Gates, C.; Wernig, A. and others. Identification of a
novel population of muscle stem cells in mice: potential for muscle
regeneration. J Cell Biol 157(5):851-864; 2002.
76.
77.
78.
79.
Reyes, M.; Lund, T.; Lenvik, T.; Aguiar, D.; Koodie, L.; Verfaillie, C. M.
Purification and ex vivo expansion of postnatal human marrow mesodermal
progenitor cells. Blood 98(9):2615-2625; 2001.
Romanski, A.; Bacic, B.; Bug, G.; Pfeifer, H.; Gul, H.; Remiszewski, S.; Hoelzer,
D.; Atadja, P.; Ruthardt, M.; Ottmann, O. G. Use of a novel histone deacetylase
inhibitor to induce apoptosis in cell lines of acute lymphoblastic leukemia.
Haematologica 89(4):419-426; 2004.
Roskams, T. A.; Libbrecht, L.; Desmet, V. J. Progenitor cells in diseased human
liver. Semin Liver Dis 23(4):385-396; 2003.
Sato, Y.; Araki, H.; Kato, J.; Nakamura, K.; Kawano, Y.; Kobune, M.; Sato, T.;
Miyanishi, K.; Takayama, T.; Takahashi, M. and others. Human mesenchymal
stem cells xenografted directly to rat liver are differentiated into human
hepatocytes without fusion. Blood 106(2):756-763; 2005.
80.
81.
82.
83.
84.
85.
86.
Schmelzer, E.; Triolo, F.; Turner, M. E.; Thompson, R. L.; Zeilinger, K.; Reid, L.
M.; Gridelli, B.; Gerlach, J. C. Three-dimensional perfusion bioreactor culture
supports differentiation of human fetal liver cells. Tissue Eng Part A
16(6):2007-2016; 2010.
Secco, M.; Zucconi, E.; Vieira, N. M.; Fogaca, L. L.; Cerqueira, A.; Carvalho, M.
D.; Jazedje, T.; Okamoto, O. K.; Muotri, A. R.; Zatz, M. Multipotent stem cells
from umbilical cord: cord is richer than blood! Stem Cells 26(1):146-150;
2008.
Shih, D. T.; Lee, D. C.; Chen, S. C.; Tsai, R. Y.; Huang, C. T.; Tsai, C. C.; Shen, E. Y.;
Chiu, W. T. Isolation and characterization of neurogenic mesenchymal stem
cells in human scalp tissue. Stem Cells 23(7):1012-1020; 2005.
Sneddon, J. B.; Borowiak, M.; Melton, D. A. Self-renewal of
embryonic-stem-cell-derived progenitors by organ-matched mesenchyme.
Nature 491(7426):765-768; 2012.
Sonabend, A. M.; Ulasov, I. V.; Tyler, M. A.; Rivera, A. A.; Mathis, J. M.; Lesniak,
M. S. Mesenchymal stem cells effectively deliver an oncolytic adenovirus to
intracranial glioma. Stem Cells 26(3):831-841; 2008.
Sterodimas, A.; de Faria, J.; Nicaretta, B.; Pitanguy, I. Tissue engineering with
adipose-derived stem cells (ADSCs): current and future applications. J Plast
Reconstr Aesthet Surg 63(11):1886-1892; 2010.
Sykova, E.; Homola, A.; Mazanec, R.; Lachmann, H.; Konradova, S. L.; Kobylka,
P.; Padr, R.; Neuwirth, J.; Komrska, V.; Vavra, V. and others. Autologous bone
marrow transplantation in patients with subacute and chronic spinal cord
injury. Cell Transplant 15(8-9):675-687; 2006.
87.
88.
89.
90.
91.
Tarte, K.; Gaillard, J.; Lataillade, J. J.; Fouillard, L.; Becker, M.; Mossafa, H.;
Tchirkov, A.; Rouard, H.; Henry, C.; Splingard, M. and others. Clinical-grade
production of human mesenchymal stromal cells: occurrence of aneuploidy
without transformation. Blood 115(8):1549-1553; 2010.
Thirumala, S.; Goebel, W. S.; Woods, E. J. Clinical grade adult stem cell
banking. Organogenesis 5(3):143-154; 2009.
To, L. B.; Levesque, J. P.; Herbert, K. E. How I treat patients who mobilize
hematopoietic stem cells poorly. Blood 118(17):4530-4540; 2011.
Tosh, D.; Strain, A. Liver stem cells--prospects for clinical use. J Hepatol 42
Suppl(1):S75-84; 2005.
Tuli, R.; Tuli, S.; Nandi, S.; Wang, M. L.; Alexander, P. G.; Haleem-Smith, H.;
Hozack, W. J.; Manner, P. A.; Danielson, K. G.; Tuan, R. S. Characterization of
multipotential mesenchymal progenitor cells derived from human trabecular
bone. Stem Cells 21(6):681-693; 2003.
92.
93.
94.
95.
96.
97.
98.
99.
Uy, G. L.; Rettig, M. P.; Cashen, A. F. Plerixafor, a CXCR4 antagonist for the
mobilization of hematopoietic stem cells. Expert Opin Biol Ther
8(11):1797-1804; 2008.
Vessey, C. J.; de la Hall, P. M. Hepatic stem cells: a review. Pathology
33(2):130-141; 2001.
Vishnubalaji, R.; Manikandan, M.; Al-Nbaheen, M.; Kadalmani, B.; Aldahmash,
A.; Alajez, N. M. In vitro differentiation of human skin-derived multipotent
stromal cells into putative endothelial-like cells. BMC Dev Biol 12:7; 2012.
Wagner, W.; Horn, P.; Castoldi, M.; Diehlmann, A.; Bork, S.; Saffrich, R.; Benes,
V.; Blake, J.; Pfister, S.; Eckstein, V. and others. Replicative senescence of
mesenchymal stem cells: a continuous and organized process. PLoS One
3(5):e2213; 2008.
Wang, H. S.; Hung, S. C.; Peng, S. T.; Huang, C. C.; Wei, H. M.; Guo, Y. J.; Fu, Y.
S.; Lai, M. C.; Chen, C. C. Mesenchymal stem cells in the Wharton's jelly of the
human umbilical cord. Stem Cells 22(7):1330-1337; 2004.
Wang, S.; Qu, X.; Zhao, R. C. Clinical applications of mesenchymal stem cells. J
Hematol Oncol 5:19; 2012.
Wexler, S. A.; Donaldson, C.; Denning-Kendall, P.; Rice, C.; Bradley, B.; Hows, J.
M. Adult bone marrow is a rich source of human mesenchymal 'stem' cells
but umbilical cord and mobilized adult blood are not. Br J Haematol
121(2):368-374; 2003.
Winkler, I. G.; Sims, N. A.; Pettit, A. R.; Barbier, V.; Nowlan, B.; Helwani, F.;
Poulton, I. J.; van Rooijen, N.; Alexander, K. A.; Raggatt, L. J. and others. Bone
marrow macrophages maintain hematopoietic stem cell (HSC) niches and
their depletion mobilizes HSCs. Blood 116(23):4815-4828; 2010.
100.
101.
102.
103.
104.
Wu, K. H.; Wu, H. P.; Chan, C. K.; Hwang, S. M.; Peng, C. T.; Chao, Y. H. The role
of mesenchymal stem cells in hematopoietic stem cell transplantation: from
bench to bedsides. Cell Transplant 22(4):723-729; 2013.
Wu, L. F.; Wang, N. N.; Liu, Y. S.; Wei, X. Differentiation of Wharton's jelly
primitive stromal cells into insulin-producing cells in comparison with bone
marrow mesenchymal stem cells. Tissue Eng Part A 15(10):2865-2873; 2009.
Wu, X. B.; Tao, R. Hepatocyte differentiation of mesenchymal stem cells.
Hepatobiliary Pancreat Dis Int 11(4):360-371; 2012.
Xu, S.; De Becker, A.; Van Camp, B.; Vanderkerken, K.; Van Riet, I. An improved
harvest and in vitro expansion protocol for murine bone marrow-derived
mesenchymal stem cells. J Biomed Biotechnol 2010:105940; 2010.
Xu, S.; Menu, E.; De Becker, A.; Van Camp, B.; Vanderkerken, K.; Van Riet, I.
Bone marrow-derived mesenchymal stromal cells are attracted by multiple
myeloma cell-produced chemokine CCL25 and favor myeloma cell growth in
vitro and in vivo. Stem Cells 30(2):266-279; 2012.
105. Yang, J.; Zhou, W.; Zheng, W.; Ma, Y.; Lin, L.; Tang, T.; Liu, J.; Yu, J.; Zhou, X.; Hu,
J. Effects of myocardial transplantation of marrow mesenchymal stem cells
transfected with vascular endothelial growth factor for the improvement of
heart function and angiogenesis after myocardial infarction. Cardiology
107(1):17-29; 2007.
106.
107.
108.
109.
110.
Yang, J.; Zhu, Z.; Wang, H.; Li, F.; Du, X.; Ma, R. Z. Trop2 regulates the
proliferation and differentiation of murine compact-bone derived MSCs. Int J
Oncol; 2013.
Zachar, V.; Rasmussen, J. G.; Fink, T. Isolation and growth of adipose
tissue-derived stem cells. Methods Mol Biol 698:37-49; 2011.
Zhou, G.; Liu, W.; Cui, L.; Wang, X.; Liu, T.; Cao, Y. Repair of porcine articular
osteochondral defects in non-weightbearing areas with autologous bone
marrow stromal cells. Tissue Eng 12(11):3209-3221; 2006.
Zuk, P. A. The adipose-derived stem cell: looking back and looking ahead. Mol
Biol Cell 21(11):1783-1787; 2010.
Zuk, P. A.; Zhu, M.; Mizuno, H.; Huang, J.; Futrell, J. W.; Katz, A. J.; Benhaim, P.;
Lorenz, H. P.; Hedrick, M. H. Multilineage cells from human adipose tissue:
implications for cell-based therapies. Tissue Eng 7(2):211-228; 2001.
Table 1: Surface marker expression profiles of main MSCs types (updated from review article, Hakan
Orbay, 2012) (68).
MSCs
CD marker expression
CD9+ , CD10+, CD13+, CD29+, CD44+, CD49d+, CD49e+, CD54+, CD55+, CD71+,
ADSCs
CD73+, CD90+, CD105+, CD106+, CD146+, CD166+ and STRO-1+ (38,109).
CD13+, CD33+, CD44+, CD73+, CD90+,CD105+, CD166+, CD28+, HLA class I+
BMMSCs
(7,17,98)
PDLSCs
STRO-1+, CD13+, CD29+, CD44+, CD59+, CD90+, CD105+ (37)
TBMSCs
CD73+, STRO-1+, CD105+ (91)
SMMSCs
CD44+, CD73+, CD90+, CD105 + (16)
PMSCs
CD90+ (42)
MMSCs
CD34+, CD117+, Sca1+ (40,75)
CD105+, CD90+, CD73+, CD29+, CD13+, CD44+ CD59+, VCAM-1+, ICAM-1+,
SSCs
CD49+, CD166+, SH2+, SH4+, EGFR+, PDGFRa+, CD271+, Stro-1+, CD71+, CD133+,
CD166+, Keratin-19+ (63,82,94)
WJ-MSCs
CD13+, CD29+, CD44+, CD51+, CD73+, CD90+, CD105+, SH2, SH3 (22,96,101)
HSC
EpCAM+, E-cadherin+, CD133+, CD29 (90)+
Abbreviation: adipose-derived stem cells (ADSCs), bone marrow-derived-stem cells (BMMSCs),
periodontal ligament-derived stem cells (PDLSCs), trabecular bone-derived-stem cells (TBMSCs),
synovial membrane-derived stem cells (SMMSCs), periosteum-derived stem cells (PMSCs),
muscle-derived stem cells and satellite cells (MMSCs), skin stem cells (SSCs), wharton’s jelly
stem cells (WJ-MSCs), hepatic stem cells (HSC).
Figure 1. The relationship of hMSCs separation technology with potential clinical
application. Isolation methods would be influenced operating times, and the hMSCs
amounts and qualities that are important for transplant medicine.
Download