Skip to main content
International Journal of Clinical and Experimental Medicine logoLink to International Journal of Clinical and Experimental Medicine
. 2010 Sep 7;3(4):248–269.

Bone marrow and umbilical cord blood human mesenchymal stem cells: state of the art

Arianna Malgieri 1, Eugenia Kantzari 2, Maria Patrizia Patrizi 3, Stefano Gambardella 1,4
PMCID: PMC2971538  PMID: 21072260

Abstract

Mesenchymal stem cells (MSCs) are multipotent adult stem cells present in all tissues, as part of the perivascular population. As multipotent cells, MSCs can differentiate into different tissues originating from mesoderm ranging from bone and cartilage, to cardiac muscle. MSCs are an excellent candidate for cell therapy because they are easily accessible, their isolation is straightforward, they can be bio-preserved with minimal loss of potency, and they have shown no adverse reactions to allogeneic versus autologous MSCs transplants. Therefore, MSCs are being explored to regenerate damaged tissue and treat inflammation, resulting from cardiovascular disease and myo-cardial infarction (MI), brain and spinal cord injury, stroke, diabetes, cartilage and bone injury, Crohn's disease and graft versus host disease (GvHD). Most of the application and clinical trials involve MSCs from bone marrow (BMMSCs). Transplantation of MSCs from bone marrow is considered safe and has been widely tested in clinical trials of cardiovascular, neurological, and immunological disease with encouraging results. There are examples of MSCs utilization in the repair of kidney, muscle and lung. The cells were also found to promote angiogenesis, and were used in chronic skin wound treatment. Recent studies involve also mesenchymal stem cell transplant from umbilical cord (UCMSCt). One of these demonstrate that UCMSCt may improve symptoms and biochemical values in patients with severe refractory systemic lupus erythematosus (SLE), and therefore this source of MSCs need deeper studies and require more attention. However, also if there are 79 registered clinical trial sites for evaluating MSC therapy throughout the world, it is still a long way to go before using these cells as a routinely applied therapy in clinics.

Keywords: Umbilical cord blood, mesenchymal stem cells, regenerative medicine, cell therapy, umbilical cord blood banking

1. Mesenchymal Stem Cells (MSCs)

Introduction to MSCs

Mesenchymal stem cells (MSCs), also known as marrow stromal cells [1] or mesenchymal progenitor cells [2], are definied as self-renewal, multipotent progenitor cells with the capacity to differentiate into several distinct mesenchymal lineages [3]. To date, MSCs of multiple adult vertebrate species have been demonstrated to differentiate into connective skeletal tissue, bone, cartilage, marrow stroma and adipocytes [4,5]. In addition, controversial data suggest that MSCs may give rise to sarcomeric muscle (skeletal and cardiac) [6-8], endothelial cells [9] and even cells of non-mesodermal origin, such as hepatocytes [10], neural cells [11] and epithelial cells [12,13]. Hence, the descriptive terms pluripotent or multipotent are reciprocally used to describe the capacity of MSCs to differentiate into a wide arrange of mammalian tissues [14].

MSCs were initially identified in bone marrow (BM) and later in muscle, adipose and connective tissue of human adults [15-18]. However, because the frequency and differentiating capacity of MSCs decrease with age [19], alternative sources of MSCs have been sought. MSCs have been identified in human amniotic fluid, placenta, umbilical cord blood (UCB) and veins [20-22] as well as in several fetal tissues including bone marrow, liver, blood, lung and spleen [23-25]. MSCs isolated from the synovium as an adherent cell population were capable of differentiation into chondrocytes, osteocytes and adipocytes [26]. They also showed that these cells were capable of contributing to skeletal muscle regeneration in a nude mouse model and restored expression of dystrophin in the sarcolemma in dystrophic muscle of immunosuppressed mdx mice [27].

Stem cells from adipose tissue, variously referred to as processed lipoaspirate (PLA) cells [28] and adipose-derived adult stem (ADAS) cells [29], have been shown to have similar differentiation potential. De Ugarte et al. [30] suggest that there is little difference between cells from marrow and fat in terms of yield, growth kinetics, cell senescence, multi-lineage differentiation capacity, and gene transduction efficiency. The utility of these cells in therapeutic applications may then depend on the availability of tissue specimens and the ease of in vitro expansion. Kaviani et al. [31] first described the presence of a sub-population of amniotic fluid cells with mesenchymal features, able to proliferate in vitro more rapidly than comparable fetal and adult cells. In ‘t Anker et al. [20] demonstrated that the amniotic fluid can be an abundant source of fetal cells that exhibit a pheno-type and a multilineage differentiation potential similar to that of bone marrow-derived MSCs; these cells were named amniotic fluid mesenchymal stem cells (AFMSC). These characteristics, together with the absence of ethical issues concerning their employment, suggest that stem cells present in amniotic fluid might be promising candidates for tissue engineering and stem cell therapy of several human disorders.

Also if some confusion exists regarding the muscle-derived stem cells (MDSCs) and their potential application in regenerative medicine and gene therapy, more recent evidence supports the existence of a population of multipotential MDSCs able to differentiate into other mesodermal cell types. Muscle-derived cells have been shown to differentiate into mesenchymal tissues, functionally regenerating bone and muscle, as well as play a role in cartilage healing [32-35].

The considerable therapeutic potential of MSCs has generated markedly increasing interest in a wide variety of biomedical disciplines. In different papers investigators report studies of MSCs using different methods of isolation and expansion, and different approaches to characterizing the cells. Therefore, Mesenchymal and Tissue Stem Cell Committee of the International Society for Cellular Therapy proposes minimal criteria to define human MSCs. First, MSCs must be plastic-adherent when maintained in standard culture conditions. Second, MSCs must express CD105, CD73 and CD90, and lack expression of CD45, CD34, CD14 or CD11b, CD79a or CD19 and HLA-DR surface molecules. Third, MSCs must differentiate to osteoblasts, adipocytes and chondroblasts in vitro [36,37].

MSCs and bone marrow

BM is a complex tissue containing hematopoi-etic progenitor cells and a connective-tissue network of stromal cells. Blood and the system that forms it, known as the hematopoietic system, consist of many cell types with specialized functions. Red blood cells (erythrocytes) carry oxygen to the tissues. Platelets (derived from megakaryocytes) help prevent bleeding. Granulocytes (neutrophils, basophils and eosinophils) and macrophages (collectively known as myeloid cells) fight infections from bacteria, fungi, and other parasites such as nematodes (ubiquitous small worms). Some of these cells are also involved in tissue and bone remodeling and removal of dead cells. B lymphocytes produce antibodies, while T lymphocytes can directly kill or isolate by inflammation cells recognized as foreign to the body, including many virus-infected cells and cancer cells. Many blood cells are short-lived and need to be replenished continuously; the average human requires approximately one hundred billion new hematopoietic cells each day. The continued production of these cells depends directly on the presence of Hematopoietic Stem Cells (HSCs), the ultimate, and only, source of all these cells [38].

Marrow stroma includes a subpopulation of undifferentiated cells that are capable of becoming one of a number of phenotypes, including bone and cartilage, tendon, muscle, fat, and marrow stromal connective tissue which supports hematopoietic cell differentiation [39,40]. MSCs represent a very small fraction, 0.001–0.01% of the total population of nucleated cells in marrow [5]. Many studies have defined conditions for isolation, expansion, and in vitro and in vivo differentiation of the stromal cells. These cells are referred to as MSCs, since they are known to have capacity of proliferation and differentiation into the mesenchymal lineage. Their mesenchymal differentiation potential is retained even after repeated subcultivation in vitro [41,42].

Though BM has been represented as the main available source of MSCs [5,43], the use of BM-derived cells is not always acceptable owing to the high degree of viral exposure and the significant decrease in the cell number and the proliferative/differentiation capacity along with age. In addition, it requires a painful invasive procedure to obtain a BM sample. Therefore, the identification of alternative sources of MSCs may provide significant clinical benefits with respect to ease of accessibility and reduced morbidity [44-47].

MSCs and umbilical cord blood

The umbilical cord (UC) contains two arteries and one vein, which are surrounded by mucoid connective tissue, and this is called the Wharton's jelly. The cord is covered by an epithelium derived from the enveloping amnion. The network of glycoprotein microfibrils and collagen fibrils in the Wharton's jelly has been previously studied [48]. The UCB has been used as an alternative source since 1988 [49]. The blood remaining in the umbilical vein following birth contains a rich source of hematopoietic stem and progenitor cells, has been used successfully as an alternative allogeneic donor source to treat a variety of pediatric genetic, hematologic, immunologic, and oncologic disorders [50-53]. Fresh cord blood is also a promising source of non-hematopoietic stem cells. Among others, it contains endothelial cells, MSCs and unrestricted somatic stem cells (USSC) [54-57].

Primitive stromal cells can be isolated from umbilical cord Wharton's jelly and can be differentiated into different cells, like osteoblasts, chondrocytes, adipocyte, cardiomyocyte and neurocyte [58,59]. Most of the time UCB is still regarded as medical waste in the delivery rooms, whereas, in contrast to BM aspiration, it is obtained by a simple, safe and painless procedure when the baby is delivered.

There are many advantages of UCB as a source of Human Stem Cells (HSCs) as compared to BM and Peripheral Blood (PB). First, the collection of cord blood units is easy and non-invasive for the donor and therefore the number of potential donors is higher than for bone marrow. Umbilical cord can be easily obtained without causing pain, and the procedure avoids ethical and technical issues [58]. Then, cord blood units are stored in advance and are therefore rapidly available when needed while bone marrow has to be collected from the donor just before transplantation and there is always a risk of last minute consent refusal. Moreover, MSCs from UCB are more primitive than MSCs isolated from some other tissue sources [60-63]. Despite, reports to date have focused on obtaining those cells after culture expansion from a segment of the umbilical cord [64-66]. Culture expansion has a disadvantage, the cells cannot be frozen on the same day as UCB cells arrive in the laboratory, and there is the increased risk of contamination with any culture manipulation. Moreover, MSCs from UCB have lower success rate of isolating if compared with MSCs from BM (63% vs 100%) [67].

Finally, the human leukocyte antigen type (HLA) does not need to be a perfect match in case of allogeneic cord blood cell transplantation because these cells are less likely to induce immunological reactions than bone marrow cells. UCB cells are good substitutes for BM-derived hematopoietic progenitors due to the immaturity of newborn cells [68]. The immaturity of cells is associated with lower immunogenicity, therefore, UCB reduces graft-versus-host reactivity when compared with adult-derived marrow grafts. Furthermore, UCB raises no ethical issues for basic studies and clinical applications.

2. Technical characteristics of human cord mes-enchimal stem cells (hUCMSCs)

Isolation and characterization of MSCs from UCB: state of the art

Although the isolation of hematopoietic stem cells from UCB has been well established, the isolation and characterization of MSCs from UCB still need to be evaluated and are controversial. Erices et al. [21] reported that UCB-derived mononuclear cells gave rise to two adherent cell types, with only one of them expressing MSC-related surface antigens. Mareschi et al. [69] reported that under given conditions, it was possible to isolate MSCs from BM, but not from UCB; Goodwin et al. [70] have reported the multi-lineage differentiation ability of UCB-isolated cells. Neither of these reports provided sufficient evidence to fulfill the qualifying criteria for MSCs because relatively heterogeneous cells were reported by both groups. Wexler et al. [71] have recently reported that UCB is not a rich source of human MSCs, while Musina et al. [72] found that a specific feature of human umbilical cord blood mesenchymal stem cells (hUCBMSCs) is their low count per volume of the initial material and very low proliferative activity. On the other hand, Romanov et al. [73] suggested that umbilical cord contains a high number of MSC-like elements forming colonies of fibroblastoid cells that may be successfully expanded in culture. These MSC-like cells contain no endothelium- or leukocyte specific antigens but express alpha-smooth muscle actin (α-SMA) and several mesenchymal cell markers. Therefore, umbilical cord/placenta stroma could be regarded as an alternative source of MSCs for experimental and clinical needs.

Consistent findings within the literature include the extent of patient variability between each donor blood sample and the contamination of a large number of cells such as fibroblastic cells, dendritic cells, adherent monocytes, macrophages, and osteoclastic cells arising within the cultures [69,70,74].

Methods of isolation and hUCMSCs culture

There are four methods for isolation of MSCs from UC: density gradient centrifugation, flow cytometer isolation, attachment screening and two step, enzymatic digestion [75]. In 2003, Romanov et al. using enzymatic digestion and centrifugation methods isolated well developed colonies of fibroblast like cells and further characterization revealed that these cells expressed MSC markers [73]. Lu et al. [66] attempted to isolate MSC according to the protocol described in the report and obtained MSCs from three of ten UCs. Wang et al. [76] centrifuged the mesenchymal tissue scraped from the Wharton's jelly, treated it with collagenase and 2.5% trypsin and the cells were cultured in Dulbecco's modified Eagle's medium (DMEM) supplemented with 10% fetal bovine serum (FBS). Finally, they obtained 25 ×103 cells per centimetre of umbilical cord. In 2006, Weiss et al. [65] reported a more efficient method of starting the cultures via enzymatic degradation of the extracellular matrix to release the cells from the WJ.

hUCMSCs were isolated from 78% of the cords; including from one of the two cords refrigerate for 24 hours prior. Using this improved procedure, UCMSCs were isolated from every cord and up to 1.5 ×106 cells per cm of UC.

In Fu's [77] study, approximately 1×106 hUCMSCs were collected from 20 cm of UC and the number of hUCMSCs doubled (2×106) in 10% FBS DMEM in 3 days. They found that hUCMSCs in Wharton's jelly of the UC can be easily obtained and processed compared with embryonic and bone marrow stem cells. Lu et al. [66] established a simple, two step enzymatic digestion, to isolate and culture hUCMSCs from each of 36 UCs, which may be the most efficient way to isolate MSCs from UC.

A new and simple method of obtain and cryopre-serving hUCMSCs extracted from a small piece of UC was described by Friedman et al. [78]. This method is followed by mincing the tissue and cryopreserving it in autologous cord plasma to prevent exposure to allogeneic or animal serum, thus showing that UCMSCs are a reliable, easily accessible, noncontroversial source of MSCs.

Immunophenotype and comparison of hUCMSCs and hBMMSCs

MSCs express numerous receptors important for cell adhesion with hematopoietic cells. Much valuable information can also be gained from a systematic analysis of cell surface molecules on MSCs. Majumdar et al. [79] determined that MSCs express a large spectrum of cell adhesion molecules of potential importance in cell binding and homing interactions. MSCs exhibit high expression of integrins that could also play a role in homing to sites of injury and binding to specific matrix molecules in the manner suggested by Bogenrieder and Herlyn [80].

The morphology of hUCMSCs in the culture show a typical MSCs immunophenotypic markers and fibroblastoid morphology. The absence of endothelial CD31 and leukocyte surface markers support classifying hUCMSCs as mesenchymal progenitors. Very important, hUCMSCs are negative for CD14, CD28, CD31, CD33, CD34, CD45, CD56, CD133, HLA-DR, and for graft versus host disease CD80, CD86, CD40, which shows that they are appropriate for transplantation.

Recent studies show that hUCMSCs share most of their immunophenotype with Bone Marrow Mesenchymal Stem Cells (BMMSCs), including a cluster of differentiating markers, neural markers and extracellular adhesion molecules. Surface markers of hUCMSCs and BMMSCs are shown in Table 1. They share the expression of CD13, CD29, CD44, CD73, CD90, CD105, CD146. Moreover, they share the cell cycle status, the adipogenic and osteogenic differentiation capacity and finally the cytokines as well as haematopoietic supportive function.

Table 1.

Surface markers expressed by hUCMSCs and BMMSCs [186]

Surface markers hUC BM
CD13 ++++ ++++
CD14 - -
CD29 ++++ ++++
CD31 - -
CD34 - -
CD38 - -
CD44 ++++ ++++
CD45 - -
CD73 ++++ ++++
CD90 ++++ ++++
CD105 ++++ ++++
CD106 + +++
CD146 ++++ +++
HLA-ABC +++ ++++
HLA-DR - -

Despite this, recent studies show that there are still several differences between them [81]. Firstly, the fibroblast colony-forming units (CFUF) frequency was significantly higher in UC derived nucleated cells than in BM derived nucleated cells. Since CFUF represents the mesenchymal progenitor cell, this suggested a higher frequency of MSCs in the nucleated cells of UC than in those of BM. Secondly, the proliferation analysis revealed that hUCMSCs have a faster population doubling time, that not change after 30 passages. In contrast, BMMSC showed significantly slower population doubling time which became even longer after Passage 6. hUCMSCs had a higher proliferative capacity of in comparison with BM-MSCs indicating that UCMSCs may be a novel alternative source of human MSCs for clinical application.

In addition, hUCMSCs showed lower expression of CD106 and HLA-DR in comparison with hBMMSC. The different expression of CD106 in hUCMSCs and BMMSCs may represent a specific indicator for identifying peripheral MSCs from BMMSCs because low expression of CD106 has also been identified in adipocyte derived MSCs. Furthermore, Lu et al. [66] noted low expression of HLA-ABC on hUCMSCs and the absence of HLA-DR expression. Because HLA-ABC could be a hurdle for allogeneic cell therapies, the lower expression of HLA-ABC may favour the use of hUCMSCs for allogeneic cell therapy.

Collection strategies

Collection strategy is the first step for collecting good-quality UCB units. There are many techniques of UCB sample collection. The key difference regards whether samples are taken while the placenta is still in the uterus (in utero), or after the delivery of the placenta (ex utero). The in utero collections are usually performed by obstetricians and midwives in the delivery room, while ex utero UCB collection is performed after delivery of the placenta, in an adjacent room, by trained personnel of UCB bank.

Many authors have investigated the impact of in utero and ex utero collection strategy on the quality of UCB units. They compared both strategies in vaginal deliveries and concluded that in utero collection yielded a significantly higher volume, total mononuclear cells (tMNCs) number, CD34 cells and total CFU [82,83]. In one of this study, Surbek et al. [82] has confirmed that larger UCB volume and the tMNCs were obtained in the samples collected before placental delivery for Caesarean deliveries also. Wong et al. [84] reported collection of UCB from the same cord before and after the placenta was delivered, and they observed that the concentration of nuclear cells was higher when the UCB was in utero than after it was delivered. Solves et al. [83] showed that the proportion of excluded UCB units (discard rate) after collection was significantly higher for ex utero group. The main reasons for discarding these units were low volume and total cell count. All these authors concluded that it is beneficial to collect UCB while the placenta is still in utero. Nevertheless, Lasky et al. [85] in a large retrospective multicentre study did not find differences in volume, total cell count and discard rate between in utero and ex utero strategies. Several other methods of UCB collection have been previously described [86,87].

However, Bertolini et al. [88] showed that UCB collection using an open system was associated with significant risk of bacterial and maternal cell contamination compared to closed collection system. According to these data, open systems have been replaced by closed systems. Other means to increase UCB volume include multiple punctures or placenta manipulation, but these should increase the risk of bacterial or maternal cell contamination [89]. All of mentioned studies conclude that the in utero collection strategy, performed with single puncture of umbilical vein and into closed system is the best approach for collecting good-quality UCB units.

In order to improve in utero technique of UCB collection, more recently Skoric et al. [90] designed closed blood-collection set for active Syringe/Flush/Syringe method. The samples were collected before placental expulsion by trained personnel. They compared that method with the usual in utero method performed by obstetric staff. In all cases, the umbilical cord was clamped within 30 s after delivery and single umbilical vein puncture was used. Results from this study showed that the median volume, total number of nuclear and mononuclear cells were significantly higher and proportion of excluded UCB units was significantly lower in group A (active Syringe/Flush/Syringe method) compared with group B (by gravity method). The reasons for discarding these units were low volume and total cell count. Anywhere they did not have bacterial contaminations in any case.

Cryopreservation of UCB

The cryopreservation is the accepted method for cell preservation before or after cell expansion for clinical use, and it is important to determine the optimized cryopreservation conditions of cell derived UCB. Many studies have reported the cryopreservation strategies to prevent cell damage during the freezing and thawing processes [91-94]. To reduce the damage caused by the formation of ice crystals, a cryo-medium containing cryoprotectants (CPAs) was used. The standard CPAs is dimethyl sulfoxide (DMSO), which is usually used at a concentration of 10% and combined with normal saline and serum albumin. DMSO is an intracellular cryoprotectant as it can move across the cell membrane by displacing the water within the cell, thus preventing the formation of ice crystals in the cell and protecting the cell from rupture [91,95]. However, DMSO has toxic effects on cells depending on the temperature and the exposure time for both pre-freeze and post-thaw periods [91,95,96]. Therefore, many studies to avoid the toxic effects on the cells have been performed new protocol to reduce concentration of DMSO (from 3.5% to 7%) for bone marrow cells and peripheral blood stem cells , as well as for HSC from UCB [97-100].

Several studies compared the effect of cooling rate and cryoprotectant concentration on UCB recovery. Donaldson et al. [101] cryopreserved UCB samples with different concentrations of DMSO and hydroxyethylstarch in combination with a variety of cooling rates. They concluded that good recovery of UCB can be achieved with 5-10% DMSO at a controlled-rate freezing of 1° C/min. Recently, Hunt et al. [102] reported a statistically significant difference in recovery between cooling at 1°C and 5°C/min in favour of −1°C/min. In 2007 Skoric et al. [90] confirmed an evident effect of cooling rate on the recovery of UCB cells. This study highlights the comparative cryo-investigation of BM cells have shown that the post-thaw recovery and clonogeneic ability of immature pluripotent and mature committed progenitors were the highest when controlled-rate freezing and 5% DMSO was used [103]. Ethylene glycol (EG) in a high concentration (about 50%) is frequently used as a permeating CPA for vitrification of mammalian embryos and oocytes because it has a lower toxicity compared to other cryoprotectants, such as DMSO [104,105]. However, there are no studies on whether a low concentration of EG (10%) could be used for cryopreservation of cord blood cells.

The purpose of a recent study [106] is to optimize the cryopreservation conditions of HSC from UCB. Improved cryopreservation conditions of HSC were established with a new cryo-medium including EG, which is the least toxic and most stable cryoprotectants, compared to the control cryo-medium. The results presented in this study provided optimal cryopreservation conditions for HSC from UCB using EG combined with the controlled-rate freezing method and thus contributes to improvements in cryopreservation of cells from UCB for research purposes and could be applied to UCB banking in the near future.

3. Umbilical cord blood banking

Public and private UCB banks

Since the first successful umbilical cord blood transplants in children with Fanconi anemia, the collection and therapeutic use of these cells has grown quickly [107]. In order to have cord blood cells available for transplantation a number of banks were created worldwide. These banks are run by either hospitals or non-profit organizations that collect the samples from donors and provide them when the cells are needed for transplantation [108]. The New York Blood Center's Placenta l Blood Program, supported by NIH, is the largest U.S. public umbilical cord blood bank and now has 13,000 donations available for transplantation into small patients who need HSCs [http://stemcells.nih.gov/info/scireport/chapter5.asp].

It is agreed by most ethical review boards that blood donated to a public bank cannot be permanently linked to the donor. The larger obstacle facing public banks is that the high costs required to maintain them has prevented more than a handful from opening. Because public banks do not charge storage fees, many medical centers do not have the funds required to establish and maintain them [109]. Then, a number of private for-profit companies have been established encouraging parents to bank their children's cord blood for their own autolo-gous use or for directed donor allogeneic use for a family member should the need arise. Parents have been encouraged to bank their infants' cord blood as a form of “biological insurance” [110].

There are different opinions regarding cord blood use. A prospective by Sullivan [111], states that the information on cord blood banking that is provided for parents needs to be scientifically accurate, and he cites 16 publications that give negative opinions about the value of private cord blood banking. He states that the probability that a privately stored CB unit will be used for an autologous transplant is extremely low. Nietfield et al. [112] performed the first calculation of lifetime probability that a person has to undergo a hematopoietic stem cell transplant (HSCT). The result of almost 1:200 Is much higher than commonly appreciated, and therefore the opinions regarding CB use need to be considered with attention and need an update. The 2007 annual report of the European Group for Blood and Bone Marrow Transplantation (EBMT) describes the current status of HSCT activity in Europe. It highlights the increasing role of allogeneic HSCT and gives the first quantitative information on novel cellular therapies. In 2007, there were 25 563 first HSCTs, 10 072 allogeneic (39%), 15 491 autologous (61%) and 3606 additional transplants reported from 613 centers in 42 countries.

Regulation and accreditation

Unlike other blood banks, that have had specific guidelines and regulations in place for many years, cord blood banks have only recently become regulated. This is because cord blood storage is a fairly new area, therefore the usual authoritative bodies have not been as quick to established the appropriate regulations on the industry.

In the United States, the Food and Drug Administration (FDA) finally set out in 2005 a full body of regulations outlining the particular procedural methods that banks and laboratories must follow. These regulations cover the appropriate collection, processing, packaging, labelling and distribution of cord blood cells. The FDA regulates cord blood under the category of “Human Cells, Tissues, and Cellular and Tissue Based-Products.” The Code of Federal Regulations under which the FDA regulates public and private cord blood banks is Title 21 Section 1271 [113]. In addition, private cord blood banks can apply for voluntary accreditation with either the American Association of Blood Banks (AABB) or the Foundation for the Accreditation of Cellular Therapy (FACT) [http://www.aabb.org].

The FDA governs the collection, processing, storage, labeling, packaging, and distribution of cord blood stem cells. There are two different standards which can apply: cGTP (current Good Tissue Practices) and cGMP (current Good Manufacturing Practices). cGTP standards apply to the collection, processing and storage of human cells, tissues, and cellular/tissue-based products (HCT/Ps) and are regulated by the Center for Biologics Evaluation and Research. All US cord blood banks must be compliant with cGTP standards. cGMP standards apply to the manufacture of a product that is considered a drug [114].

In Europe, The European Union Group on Ethics (EGE) has issued Opinion No.19 titled Ethical Aspects of Umbilical Cord Blood Banking. The EGE concluded that “the legitimacy of commercial cord blood banks for autologous use should be questioned as they sell a service, which has presently, no real use regarding therapeutic options [115].

So, the article 6 of directive 2004/23/ec of the European parliament and of the council of 31 March 2004 (on setting standards of quality and safety for the donation, procurement, testing, processing, preservation, storage and distribution of human tissues and cells) declare: “Member States shall ensure that all tissue establishments where activities of testing, processing, preservation, storage or distribution of human tissues and cells intended for human applications are undertaken have been accredited, designated, authorised or licensed by a competent authority for the purpose of those activities” [http://www.irishstatutebook.ie/2007/en/si/0598.html]. In UK, Since 5 July 2008, Cord blood banks, both public and private, must be licensed by the Human Tissue Authority in order to release transplants to hospitals in the National Health Service.

The Human Tissue Authority (HTA) is an independent watchdog that supports public confidence by making sure human tissue is used safely and ethically, and with proper consent [http://www.hta.gov.uk]. They license and inspect organisations that store and use human tissue for purposes including teaching about the human body, carrying out post-mortem examinations, using human tissue to treat patients, carrying out research on human tissue and displaying human bodies or tissue in public.

4. Mesenchymal stem cell stherapy

Tissue engineering and regenerative medicine are the terms that are nowadays used to describe the approach to generate complex tissues and organs from simpler pieces. The main goal is to create new therapies for patients with severe injuries or chronic diseases in which the body's own responses do not suffice to restore functional tissue. Both are multidisciplinary, young and emerging fields in biotechnology and medicine, which are expected to change patient treatment profoundly, generating and regenerating tissues and organs instead of just repairing them. Whereas tissue engineering is a more technical concept of tissue and organ reconstruction by the use of cells, scaffolds, and bio-molecules, the term regenerative medicine is more focused on the support of self healing capabilities and the use of stem cells. Stem cell therapy utilizing MSCs are the focus of a multitude of clinical studies currently underway.

Widely described, MSCs are an excellent candidate for cell therapy because (a) are easily accessible; (b) the isolation is straightforward and cells can expand to clinical scales in a relatively short period of time [116,117]; (c) can be bio-preserved with minimal loss of potency [118,119]; and (d) human trials of MSCs thus far have shown no adverse reactions to alloge-neic versus autologous MSC transplants. This last, was proved that culture-expanded MSCs did not have MHC class II cell surface markers, but rather only MHC class I and no co-stimulator molecules [120]. Thus, human MSCs could not be antigen-presenting cells and would be invisible to the host's immune system [121,122]. These observations were used to suggest that MSCs could be used as allogeneic cells thera-peutically.

Transplantation of BMMSCs is carried out in two rather different settings, autologous and allogeneic. Autologous transplantations employ a patient's own bone marrow tissue and thus present no tissue incompatibility between the donor and the host. Allogeneic transplantations occur between two individuals who are not genetically identical (with the rare exceptions of transplantations between identical twins, often referred to as syngeneic transplantations). Non-identical individuals differ in their human leukocyte antigens (HLAs), proteins that are expressed by their white blood cells. The immune system uses these HLAs to distinguish between “self” and “nonself.". For successful transplantation, allogeneic grafts must match most, if not all, of the six to ten major HLA antigens between host and donor. Even if they do, however, enough differences remain in mostly uncharac-terized minor antigens to enable immune cells from the donor and the host to recognize the other as “nonself".

Other proprieties of MSCs including their wide-ranging differentiation potential, their possibility of engraftment [123], their immunosuppressive effects [124] and their expansion through culture have led to increasing clinical interest in the use of MSCs in numerous pathologic situations.

The reason for which the MSCs can repair damaged tissue may be due to different mechanisms, for example, differentiation towards tissue-specific pathways, repair of the microenvironment with paracrine/juxtacrine effects of growth factors and cytokines produced by the cells [125] or extracellular matrix reorganization [126].

Bone marrow derived- mesenchymal stem cells (BMMSCs)

Transplantation of MSCs from BM is considered safe and has been widely tested in clinical trials of cardiovascular [127,128], neurological [129,130], and immunological disease [131,132] with encouraging results. More recently, groups around the world have investigated MSCs transplantation for the treatment of myriad diseases based on a newfound appreciation for MSCs' pleiotropic functions that enhance endogenous repair and attenuate immunological dysfunction. Currently, there are 79 registered clinical trial sites for evaluating MSC therapy throughout the world (http://clinicaltrials.gov/).

Today there is a strong international interest in MSCs as a potential therapy, under this the United States has 28 registered trial sites, while the rest of the world accounts for more than half of the total number (19 in Europe, 16 in China, 5 in the Middle East, 4 in India, 3 in Canada, 2 each in Africa and Japan, and 1 in Australia). Within the past year, several of the pivotal lead trials either have undergone early termination or have failed to meet primary end-points, but on the other hand, the list of reports indicating that MSCs contribute to tissue repair in vivo enlarges. There are examples of MSCs utilization in the repair of kidney [133], muscle [27] and lung [134]. The cells were also found to promote angiogenesis [135], and were used in chronic skin wound treatment [136].

Here we report the most important application with MSC from bone marrow.

Vessel and heart

Myocardial tissue had been considered incapable of regeneration. However, in 2001, Orlic et al. [137] showed that injections of bone marrow cells into infarcted mice hearts resulted in improved cardiac function. Two years later, Beltrami et al. [138] demonstrated that cardiac progenitor cells are able to differentiate to car-diomyocytes, endothelial, and smooth muscle cells. This group also demonstrated that injection of these cells into damaged hearts improved cardiac function through regeneration of the myocardium itself. When MSCs are exposed to the DNA demethylating agent (5-azacytidine), they express specific cardiac genes, adopt myo-tube morphology, produce intercalated disks, and have other functions associated with myo-cytes [7,139,140]. Once injected in an infarcted heart, they contributes to the processes of myocardial remodeling, reduction of infarctsize, scar formation, vascular repair, angiogenesis, recruitment of other regenerative factors, and ultimately homing of stem cells, thus possibly facilitating myocyte regeneration. According to this experimental work, some clinical trials have been performed, mainly to treat heart damage in patients.

Some studies used freshly isolated autologous bone marrow-derived mononuclear cells, delivered to the myocardium anytime from less than 3-day post-infarct to late stages of congestive heart failure, and showed improvements in common indicators of cardiac function [141-144]. A recently completed phase I trial, using a single infusion of allogeneic BMMSCs in patients within 10 days of acute Myocardial Infarction (MI) corroborates these findings [145]. In the randomized trial, patients receiving MSC experienced a 4-fold decrease in arrhythmias and premature ventricular contractions (PVCs), and showed improved overall health compared to patients receiving placebo. After 1-year patients revealed a significant increase in left ventricular ejection fraction (LVEF). Importantly, there were no significant adverse events in this trials and so it is validated the safety of alloge-neic MSCs. Despite this, these results should be considered with cautious optimism because the viability of MSCs post-treatment and the role of MSCs in the recovery of cardiac function remain to be elucidated.

Two group performed independently intracoro-nary short injection of autologous cultured bone marrow MSCs cells after acute myocardial infarction and for chronic ischemic cardiomyopathy [13,146,147]. In Chen study 65 of the 68 patients tolerated the injections of cells and three of them showed a transient episode of pulmonary edema, which was controlled by intravenous injection of diuretics. In these two randomized trials, the authors found improved cardiac function in patients receiving the cells. Another group in Greece [147] performed a similar study in 11 patients and found the procedure to be safe and possibly contributing to regional regeneration of myocardium. Numerous clinical trials are going to begin or will begin next years (see http://www.clinicaltrial.gov), and results may be available in the next 3 years.

Orthopedic applications and treatment of cartilage lesions

Because the MSCs were able to differentiate into osteoblasts, many researcher embarked on clinical efforts to cure gene defects by alloge-neic transplantations, using normal cells that did not exhibit the genetic defect [148,149]. One current stem cell-based orthopedic therapy includes bone marrow-derived MSC transplantation for osteogenesis imperfecta (OI), a genetic disorder in which osteoblasts synthesize defective collagen type I, which leads to a variety of skeletal pathologies.

Researchers conducted innovative studies that leveraged the therapeutic potential of allogeneic MSC transplants to treat six children with a OI [8]. The therapeutic outcome was successful (1.5%-2% of engraftment), showing donor-derived MSCs located in the bone marrow of the recipient. Bone marrow MSCs were able to give rise to properly functioning osteoblasts, resulting in the increase in bone mineral content, as well as the improvement in growth velocity and the reduction of bone fracture frequencies . Encouraged by the results, the authors performed next trials [150]. Bone marrow was obtained from allogenic, HLA-compatible, sibling donors and was given twice to each patient. Among the five children enrolled in this study, three appeared chimeric and showed donor osteoblast engraftment. As a result, those children gained significant increase in total body length measured 6 months after transplantation, in comparison to 1.25 cm for control patients. Moreover, the bone mineral content improved by 45% to 77% of the baseline values. The number of fractures, visualised by radiography, declined from an average of 10 during 6 months before treatment, to 2. Unfortunately, the follow-up study demonstrated that the growth ratio either decreased or remained unchanged. In contrast, bone mineralization continued to increase.

Another great challenge for tissue engineering using MSCs is the treatment of cartilage lesions in orthopedic medicine there are also many examples of applications involving local delivery of marrow stem cells. These include spine fusion [151], the repair of segmental bone defects [152] and craniotomy defects [153]. Similar approaches have also been described for the repair of focal defects in articular cartilage and tendon [154].

Skin defects

BMMSCs have shown potential in improving the healing of skin defects in animal models [155-157] and in humans [158]. Autologous and al-logeneic MSCs seem to be equally effective for wound repair [159]. In vivo, cultivated bone marrow and adipose-tissue MSCs have been effective in repairing the cornea after alkali burn in a rat model [13]. This repair could be due to transdifferentiation of MSCs into cornea cells [160,161] or paracrine effects and decreased inflammatory / immune reaction [161]. However, the experience with use of MSCs for wound healing is encouraging, although many questions remain about optimal culture conditions, dosage, route of application, combination with scaffolds and type of MSCs.

Kidney disorders

Many kidney disorders involve both ischemic/inflammatory and immunologic injury. Therefore cell-based therapies such as those using MSCs which function through multiple mechanisms and have the potential to target the inflammatory and immunologic pathways have been considered a clinically relevant solution in contrast to pharmacologic agents that target only a single event or pathway in the pathophysiology of a given disease.

Several preclinical studies demonstrated that ex vivo expanded MSCs can ameliorate renal injury and accelerate repair. Effects have been demonstrated in models of acute ischemia and reperfusion, acute tubular epithelial injury and experimental glomerulonephritis [162,163]. MSCs can home to sites of injury, where they modulate the repair process. They may improve functional and structural recovery of both glomerular and tubular compartments. Morigi et al. [163] esplored human BMMSCs treatment could prevent acute kidney injury (AKI) induced by cisplatin and prolong survival in an immuno-deficient mouse model. Results showed that human BMMSC infusion decreased proximal tubular epithelial cell injury and ameliorated the deficit in renal function, resulting in reduced recipient mortality. These findings indicate that human MSCs of bone marrow origin hold potential to prolong survival in AKI and should be considered for testing in a clinical trial.

Neuronal disorders

When used in vivo for neurodegenerative disorders, MSCs do not normally seem to pass through the liquoral barrier. However, MSCs can survive, migrate and differentiate into neuralglial cells after in utero intraventricular injection inside fetal rat brains [164]. Neurological recovery has been shown in animal models of Parkinson's disease, hypoxic-ischemic neural damage and retinal injury following in vivo transplantation of these cells inside the lesion [165].

A few researchers investigated cell-based treatment modalities for Huntington's disease (HD). Rossignol et al. [166] investigated the potential treatment of BMSCs in the 3-nitropropionic acid rat model of HD. They demonstrated that intrastriatal injection of BMMSCs improved motor dysfunction modestly, and those transplanted cells were still viable and metabolically active 71 days post-transplantation. They suggested that the observed recovery of function was attributed to the release of trophic factors from the MSCs because very few MSCs showed evidence of trans-differentiation.

Recently, Venkataramana et al [167] reported a first open label clinical pilot study with autologous bone-marrow-derived stem cells transplanted into the striatum of patients with advanced Parkinson's disease (PD). This disorder is a progressive neurodegenerative disease for which stem cell research has created hope in the last few years. Seven PD patients aged 22 to 62 years were enrolled to participate in the prospective, uncontrolled, pilot study of single-dose, unilateral transplantation of autologous BMMSCs. The BMMSCs were transplanted into the sublateral ventricular zone by stereotaxic surgery. Patients were followed up for a period that ranged from 10 to 36 months. These results indicate that this protocol seems to be safe, and no serious adverse events occurred after stem-cell transplantation in PD patients. The number of patients recruited and the uncontrolled nature of the trial did not permit demonstration of effectiveness of the treatment involved. However, the results encourage future trials with more patients to demonstrate efficacy [167,168]. Although this was trial carefully designed with a favourable outcome in respect to adverse effects, many questions remain and preclinical studies need to demonstrate whether such cells either can differentiate into exogenous functional neurons or provide trophic support for endogenous cells.

Human umbilical cord blood derived mesenchymal stem cells (hUCBMSCs)

Spinal cord and brain injury applications

hUCMSCs have an higher capability of differentiating into nerve like cells and a hold great promise as tools for understanding development and as therapeutic agents for brain injury and spinal cord injury. Transplantation of hUCMSCs into the injured spinal cord may have the following functions: compensation for demyelination; removal of inhibition; promotion of axonal regeneration; direction of axons to appropriate targets and replacement of lost cells. Weiss et al. [65] has treated rat models affected by Parkinson's disease with hUCMSCs. The results demonstrated that the hUCMSCs produce significant amounts of glial cell line-derived neurotrophic factor (GDNF), one of the most potent trophic factors for dopaminergic neurons and fibroblast growth factor, the animals with transplanted cells showed a significant recovery in behaviour. Although the above data from hUCMSCs indicate that these cells may be therapeutically useful in treating CNS disorders, transplantation of hUCMSCs for treatment of spinal cord injury is just the beginning.

In ischemia studies, most data show that cell therapy is performed using hUCB. The first evidence of a therapeutic effect of hUCB came from a study where rat was used to induce focal ischemia. Intravenous administration of hUCB reduced behavioral deficits after stroke in rats [176]. In an other recently study the scientist examined the effects of hUCBMSCs in canine thromboembolic brain model [177]. Cerebral ischemia was induced through occlusion of the middle cerebral artery by injecting thrombus emboli into 10 beagles. hUCBMSCs were transplanted through the basilar artery 1 day after ischemic induction using an endovascular inter-ventional approach. Infarct volume was reduced after intra arterial delivery of hUCBMSCs in canine cerebral ischemia whereas infarct volume was increased in the control groups. hUCBMSCs expressed neuroprotective factors, such as brain-derived neurotrophic factor (BDNF) and vascular endothelial growth factor (VEGF), at 4 weeks after the transplantation. Jeong et al. [178] reported that transplantation of hUCBMSCs into contralateral regions of injured rat brain at 7 d after injury resulted in significant behavioral improvement. These results suggested that transplantation of hUCBMSCs showed their efficacy by reducing the infarction lesion volume and through earlier recovery from the neurological deficit. Therefore intra-arterial transplantation of hUCBMSCs could be useful in clinical treatment of cerebral ischemia.

UCB stem cells have shown promise in the treatment of Cerebral palsy (CP) in both animal models and early human trials. CP is a devastating brain disorder that affects many children worldwide, with 10 000 infants diagnosed annually, and stem cells ultimately have the capacity to generate new cells to replace those lost through injury or disease.

Recently, considerable excitement has been generated by anecdotal reports of improvement after umbilical CB stem cell infusions in children treated in a clinical study at Duke University. Although not a randomized trial, this treatment has been used to treat more than 50 children with cerebral palsy. Preliminary observations have been encouraging (see <http://www.msnbc.msn.com/id/> 23572206/), and many additional patients are being enrolled. It should be noted that not all children have benefited to the same extent, and it appears that the younger the patient the more significant the benefits that have been observed. However, the optimal therapeutic regime and the mechanism (s) behind any beneficial effects have yet to be determined.

Lung disease

Progression of acute respiratory distress syndrome is demonstrated by loss of lung tissue as a result of inflammation and fibrosis. Recently, the exogenous administration of BMMSCs significantly attenuated the bleomycin-induced lung injury by downmodulatingthe inflammatory responses and ameliorating their fibrotic effects [134]. However, it is not known if hUCBMSCs can differentiate into lung specific cell types in vivo, and whether these cells are suitable exogenous stem cell sources in lung injuries of experimental or clinical settings. A new study [179] examined whether intratracheal or intraperitoneal transplantation of hUCBMSCs can attenuate hyperoxia-induced lung injury in im-munocompetent newborn rats. Wild-type rats were randomly exposed to 95% oxygen or air from birth. hUCBMSCs was administered either intratracheally or intraperitoneally at postnatal day (P) 5 and after 10 days the harvested lungs were examined. The transplantation of hUCBMSCs to the wild-type newborn rat pups significantly attenuated the hyperoxia-induced lung injury in the surviving animals. This protective effect was associated and probably mediated by the down-modulation of the pulmonary inflammatory and the ensuing fibrotic responses. These findings suggest that the administration of hUCBMSCs might be a possible candidate for the new therapeutic modality for the hyperoxia-induced neonatal lung diseases, such as clinical Bronchopulmonary dysplasia (BPD).

Kidney injury

The application of hUCBMSCs in treating acute renal failure (ARF) has not been reported in a lot of studies. However a recently issue [180] showed the transplantation of hUCMSCs via the left carotid artery into ARF rats. Serum creatinine and urea nitrogen decreased compared to control groups. In addition, the transplanted hUCBMSCs could reside in local injury sites, leading to the relief of hyperemia and inflammation, but no obvious trans-differentiation into renal-like cells. The results lay the foundation for further study on the potential application of hUCMSC in human disease.

For acute Kidney Injury (AKI) a recently study showed the potentiality of human hUCBMSCs to cure mice with this disease. Infusion of hUCBMSCs in immunodeficient mice with cis-platin-induced AKI ameliorated both renal function and tubular cell injury, and prolonged survival. Transplanted hUCBMSCs are able to reduce the apoptosis and increased the cell proliferation by the capacity of the stem cell to produce growth factors. Altogether these results highlight the potential of human hUCBMSCs as future cell therapy for testing in human AKI [181].

Juvenile diabetes

Approximately 15 000 youth in the US are newly diagnosed with Types 1 Diabetes (T1D) annually (http://www.diabetes.niddk.nih.gov/dm pubsstatis-tics/#youngpeople) and 5–10% of all adults living with diabetes display the T1D phenotype (<http://diabetes.niddk.nih.gov). At present, autologous hUCBMSCs are being evaluated in a clinical trial to treat T1D in children [182]. To date, 23 children have been treated, and the first child treated under the study protocol showed significant improvement in glucose versus control and was able to produce insulin much longer than children with a similar prognosis [183]. Most of the treated children have reported enhanced blood glucose control and management. In addition, it appeared that there was retention of endogenous insulin production as assessed by stimulated C-peptide secretion.

Autoimmune Applications

Such diseases, which can affect either specific organs or the entire system, include multiple sclerosis, rheumatoid arthritis, and systemic lupus erythematosus (SLE). Chang et al. [184] investigated whether hUCBMSCs transplantation is useful in alleviating lupus nephritis in a murine model. It was found that hUCBMSCs transplantation significantly delayed the development of proteinuria, decreased anti-dsDNA, alleviated renal injury, and prolonged the life span. Together, these findings indicated that human MSCs were effective in decreasing renal inflammation and alleviating experimental lupus nephritis by inhibiting lymphocytes, inducing the cytokines and inhibition of pro-inflammatory cytokines production rather than direct engraftment and differentiating into renal tissue. Therapeutic effects demonstrated in this pre-clinical study support further exploration of the possibility to use hUCBMSCs from mismatched donors in lupus nephritis treatment.

In recent times, a new study showed a single-arm trial that involve 16 SLE patients whose disease was refractory to standard treatment for who had life-threatening visceral involvement. Sun and colleagues [185] explored if hUCBMSCs transplant may improve symptoms and biochemical values in this patients with SLE. All 16 patients received umbilical mesenchymal stem cell transplants. 10 patients completed at least 6 months of follow-up and 2 patients were followed for more than 2 years. There was no treatment-related mortality or other adverse event during or after hUCMSCt.

In conclusion, this study shows very clearly that UC-MSCT exerts a profound therapeutic effect in severe and refractory SLE patients. All the patients achieved at least 3 months of clinical and serologic improvement, and for 2 patients this was achieved without any immunosuppressive drugs. This study shows for the first time that allogenic hCBMSCs transplanatation is safe and effective, at least short term, in treating patients with severe SLE. Further clinical trials with more patients included and longer periods of follow-up compared to standard treatment will be needed to determine the efficacy and safety of this novel approach to the treatment of lupus.

5. Conclusion

MSCs are multipotent, non-haematopoietic progenitor cells that are being explored as a promising new treatment for tissue regeneration. It seems well-founded that MSCs constitute a superb potential tool in regenerative medicine approaches. They possess an extensive proliferative potential and are able to differentiate into various cell lineages. Due to these important features, the use of MSCs in clinical trials increases. It has been documented that these cells engraft successfully in patients and cause beneficial effects.

Developing new therapies that affect multiple disease pathways is of growing importance for patients care. MSCs transplantation represents an exciting approach that could potentially treat complex diseases by providing combinatorial therapy. Furthermore, the continued use of MSCs therapy can be recast to better our understanding of the natural role of these cells during health and disease in vivo.

After learning more about their properties, it will be possible to start new, more advanced and better treatment strategies for various diseases, even those, which seem to be incurable at present. Moreover, knowing that each patient is genetically different and may give different response to a treatment, and carry variable predisposition to different diseases, specifically targeted strategies using autologous MSCs, may be designed. However, it is still a long way to go before using these cells as a routinely applied therapy in clinics.

References

  • 1.Prockop DJ. Marrow stromal cells as stem cells for nonhematopoietic tissues. Science. 1997;276:71–74. doi: 10.1126/science.276.5309.71. [DOI] [PubMed] [Google Scholar]
  • 2.Conget PA, JJ Minguell. Phenotypical and functional properties of human bone marrow mesenchymal progenitor cells. J Cell Physiol. 1999;181:67–73. doi: 10.1002/(SICI)1097-4652(199910)181:1<67::AID-JCP7>3.0.CO;2-C. [DOI] [PubMed] [Google Scholar]
  • 3.Caplan AI. Why are MSCs therapeutic?. New data: new insight. J Pathol. 2009;217:318–24. doi: 10.1002/path.2469. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Alhadlaq A, Mao JJ. Tissue-engineered neo-genesis of human-shaped mandibular condyle from rat mesenchymal stem cells. J Dent Res. 2003;82:951–956. doi: 10.1177/154405910308201203. [DOI] [PubMed] [Google Scholar]
  • 5.Pittenger MF, Mackay AM, Beck SC, Jaiswal RK, Douglas R, Mosca J, Moorman M, Simonetti D, Craig S, Marshak DR. Multilineage potential of mesenchymal cells. Science. 1999;284:143–147. doi: 10.1126/science.284.5411.143. [DOI] [PubMed] [Google Scholar]
  • 6.Wakitani S, Saito T, Caplan AI. Myogenic cells derived from rat bone marrow mesenchymal stem cells exposed to 5-azacytidine. Muscle Nerve. 1995;18:1417–1426. doi: 10.1002/mus.880181212. [DOI] [PubMed] [Google Scholar]
  • 7.Makino S, Fukuda K, Miyoshi S, Konishi F, Ko-dama H, Pan J, Sano M, Takahashi T, Hori S, Abe H, Hata J, Umezawa A, Ogawa S. Cardio-myocytes can be generated from marrow stromal cells in vitro. J Clin Invest. 1999;103:697–705. doi: 10.1172/JCI5298. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Planat-Bénard V, Menard C, Andre M, Puceat M, Perez A, Garcia-Verdugo JM, Penicaud L, Casteilla L. Spontaneous cardiomyocyte differentiation from adipose tissue stroma cells. Circ Res. 2004;94:223–229. doi: 10.1161/01.RES.0000109792.43271.47. [DOI] [PubMed] [Google Scholar]
  • 9.Oswald J, Boxberger S, Jørgensen B, Feldmann S, Ehninger G, Bornhäuser M, Werner C. Mesenchymal stem cells can be differentiated into endothelial cells in vitro. Stem Cells. 2004;22:377–384. doi: 10.1634/stemcells.22-3-377. [DOI] [PubMed] [Google Scholar]
  • 10.Chagraoui J, Lepage-Noll A, Anjo A, Uzan G, Charbord P. Fetal liver stroma consists of cells in epithelial-to-mesenchymal transition. Blood. 2003;101:2973–2982. doi: 10.1182/blood-2002-05-1341. [DOI] [PubMed] [Google Scholar]
  • 11.Woodbury D, Schwarz EJ, Prockop DJ, Black IB. Adult rat and human bone marrow stromal cells differentiate into neurons. J Neurosci Res. 2000;61:364–370. doi: 10.1002/1097-4547(20000815)61:4<364::AID-JNR2>3.0.CO;2-C. [DOI] [PubMed] [Google Scholar]
  • 12.Spees JL, Olson SD, Ylostalo J, Lynch PJ, Smith J, Perry A, Peister A, Wang MY, Prockop DJ. Differentiation, cell fusion, and nuclear fusion during ex vivo repair of epithelium by human adult stem cells for bone marrow stroma. Proc Natl Acad Sci USA. 2003;100:2397–2402. doi: 10.1073/pnas.0437997100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Ma Y, Xu Y, Xiao Z, Yang W, Zhang C, Song E, Du Y, Li L. Reconstruction of chemically burned rat corneal surface by bone marrow-derived human mesenchymal stem cells. Stem Cells. 2006;24:315–321. doi: 10.1634/stemcells.2005-0046. [DOI] [PubMed] [Google Scholar]
  • 14.Jiang Y, BN Jahagirdar, RL Reinhardt, RE Schwartz, CD Keene, XR Ortiz-Gonzalez, M Reyes, T Lenvik, T Lund, M Blackstad, J Du, S Aldrich, A Lisberg, WC Low, DA Largaespada, CM Verfaillie. Pluripotency of mesenchymal stem cells derived from adult marrow. Nature. 2002;418:41–49. doi: 10.1038/nature00870. [DOI] [PubMed] [Google Scholar]
  • 15.Friedenstein AJ, Deriglasova UF, Kulagina NN, Panasuk AF, Rudakowa SF, Luria EA, et al. Precursors for fibroblasts in different populations of hematopoietic cells as detected by the in vitro colony assay method. Exp Hematol. 1974;2:83–92. [PubMed] [Google Scholar]
  • 16.Zuk PA, Zhu M, Mizuno H, Huang J, Futrell JW, Katz AJ, et al. Multilineage cells from human adipose tissue: implications for cell-based therapies. Tissue Eng. 2001;7:211–28. doi: 10.1089/107632701300062859. [DOI] [PubMed] [Google Scholar]
  • 17.Nakahara H, Dennis JE, Bruder SP, Haynesworth SE, Lennon DP, Caplan AI. In vitro differentiation of bone and hypertrophic cartilage from periostealderived cells. Exp Cell Res. 1991;195:492–503. doi: 10.1016/0014-4827(91)90401-f. [DOI] [PubMed] [Google Scholar]
  • 18.Nathanson MA. Bone matrix-directed chondro-genesis of muscle in vitro. Clin Orthop Relat Res. 1985:142–58. [PubMed] [Google Scholar]
  • 19.D'Ippolito G, Schiller PC, Ricordi C, Roos BA, Howard GA. Age-related osteogenic potential of esenchymal stromal stem cells from human vertebral bone marrow. J Bone Miner Res. 1999;14:1115–22. doi: 10.1359/jbmr.1999.14.7.1115. [DOI] [PubMed] [Google Scholar]
  • 20.In't Anker PS, Scherjon SA, Kleijburgvan der Keur C, de Groot-Swings GM, Claas FH, Fibbe WE, et al. Isolation of mesenchymal stem cells of fetal or maternal origin from human placenta. Stem Cells. 2004;22:1338–45. doi: 10.1634/stemcells.2004-0058. [DOI] [PubMed] [Google Scholar]
  • 21.Erices A, Conget P, Minguell JJ. Mesenchymal progenitor cells in human umbilical cord blood. Br J Haematol. 2000;109:235–42. doi: 10.1046/j.1365-2141.2000.01986.x. [DOI] [PubMed] [Google Scholar]
  • 22.Panepucci RA, Siufi JL, Silva WA, Jr, Proto-Siquiera R, Neder L, Orellana M, et al. Comparison of gene expression of umbilical cord vein and bone marrowderived mesenchymal stem cells. Stem Cells. 2004;22:1263–78. doi: 10.1634/stemcells.2004-0024. [DOI] [PubMed] [Google Scholar]
  • 23.Campagnoli C, Roberts IA, Kumar S, Bennett PR, Bellantuono I, Fisk NM. Identification of mesenchymal stem/progenitor cells in human first-trimester fetal blood, liver, and bone marrow. Blood. 2001;98:2396–402. doi: 10.1182/blood.v98.8.2396. [DOI] [PubMed] [Google Scholar]
  • 24.Gotherstrom C, Ringden O, Westgren M, Tam-mik C, Le Blanc K. Immunomodulatory effects of human foetal liver-derived mesenchymal stem cells. Bone Marrow Transplant. 2003;32:265–72. doi: 10.1038/sj.bmt.1704111. [DOI] [PubMed] [Google Scholar]
  • 25.in't Anker PS, Noort WA, Scherjon SA, Kleijburgvan der Keur C, Kruisselbrink AB, van Bezooijen RL, Beekhuizen W, Willemze R, Kanhai HH, Fibbe WE. Mesenchymal stem cells in human second trimester bone marrow, liver, lung, and spleen exhibit a similar immunophenotype but a heterogeneous multilineage differentiation potential. Haematologica. 2003;88:845–52. [PubMed] [Google Scholar]
  • 26.De Bari C, Dell'Accio F, Tylzanowski P, Luyten FP. Multipotent mesenchymal stem cells from adult human synovial membrane. Arthritis and Rheumatism. 2001;44:1928–1942. doi: 10.1002/1529-0131(200108)44:8<1928::AID-ART331>3.0.CO;2-P. [DOI] [PubMed] [Google Scholar]
  • 27.De Bari C, Dell'Accio F, Vandenabeele F, Ver-meesch J R, Raymackers J M, Luyten F P. Skeletal muscle repair by adult human mesenchymal stem cells from synovial membrane. The Journal of Cell Biology. 2003;160:909–918. doi: 10.1083/jcb.200212064. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Dragoo J L, Samimi B, Zhu M, Hame S L, Thomas B J, Lieberman J R, Hedrick M H, Benhaim P. Tissue-engineered cartilage and bone using stem cells from human infra patellar fat pads. The Journal of Bone and Joint Surgery. 2003;85:740–747. British. [PubMed] [Google Scholar]
  • 29.Gimble J M. Adipose tissue-derived therapeutics. Expert Opinion on Biological Therapy. 2003;3:705–713. doi: 10.1517/14712598.3.5.705. [DOI] [PubMed] [Google Scholar]
  • 30.De Ugarte D A, Morizono K, Elbarbary A, Alfonso Z, Zuk P A, Zhu M, Dragoo J L, Ashjian P, Thomas B, Benhaim P, Chen I, Fraser J, Hedrick M H. Comparison of multi-lineage cells from human adipose tissue and bone marrow. Cells Tissues Organs. 2003;174:101–109. doi: 10.1159/000071150. [DOI] [PubMed] [Google Scholar]
  • 31.Kaviani A, Perry TE, Dzakovic A, Jennings RW, Ziegler MM, Fauza DO. The amniotic fluid as a source of cells for fetal tissue engineering. Journal of Pediatric Surgery. 36:1662–1665. doi: 10.1053/jpsu.2001.27945. [DOI] [PubMed] [Google Scholar]
  • 32.Pate DW, et al. Isolation and differentiation of mesenchymal stem cells from rabbit muscle. Clin Res. 1993;41:374A. [Google Scholar]
  • 33.Young HE, Ceballos EM, Smith JC, Mancini ML, Wright RP, Ragan BL, Bushell I, Lucas PA. Pluri-potent mesenchymal stem cells reside within avian connective tissue matrices. In Vitro Cell Dev Biol Anim. 1993;29A:723–736. doi: 10.1007/BF02631429. [DOI] [PubMed] [Google Scholar]
  • 34.Rogers JJ, Young HE, Adkison LR, Lucas PA, Black AC., Jr Differentiation factors induce expression of muscle, fat, cartilage, and bone in a clone of mouse pluripotent mesenchymal stem cells. Am Surg. 1995;61:231–236. [PubMed] [Google Scholar]
  • 35.Williams JT, Southerland SS, Souza J, Calcutt AF, Cartledge RG. Cells isolated from adult human skeletal muscle capable of differentiating into multiple mesodermal phenotypes. Am Surg. 1999;65:22–26. [PubMed] [Google Scholar]
  • 36.Dominici M, Le Blanc K, Mueller I, Slaper-Cortenbach I, Marini F, Krause D, Deans R, Keating A, Prockop Dj, Horwitz E. Minimal criteria for defining multipotent mesenchymal stromal cells. The International Society for Cellular Therapy position statement. Cytotherapy. 2006;8:315–7. doi: 10.1080/14653240600855905. [DOI] [PubMed] [Google Scholar]
  • 37.Sensebé L, Krampera M, Schrezenmeier H, Bourin P, Giordano R. Mesenchymal stem cells for clinical application. Vox Sang. 2010;98:93–107. doi: 10.1111/j.1423-0410.2009.01227.x. [DOI] [PubMed] [Google Scholar]
  • 38.Chen BJ, Cui X, Sempowski GD, Domen J, Chao NJ. Hematopoietic stem cell dose correlates with the speed of immune reconstitution after stem cell transplantation. Blood. 2004;103:4344–52. doi: 10.1182/blood-2003-07-2534. [DOI] [PubMed] [Google Scholar]
  • 39.Dexter TM, Testa NG. Differentiation and proliferation of hemopoietic cells in culture. Methods Cell Biol. 1976;14:387–405. doi: 10.1016/s0091-679x(08)60498-7. [DOI] [PubMed] [Google Scholar]
  • 40.Friedrich C, Zausch E, Sugrue SP, Gutierrez-Ramos JC. Hematopoietic supportive functions of mouse bone marrow and fetal liver microen-vironment: dissection of granulocyte, B-lymphocyte, and hematopoietic progenitor support at the stroma cell clone level. Blood. 1996;87:4596–4606. [PubMed] [Google Scholar]
  • 41.Bruder SP, Jaiswal N, Haynesworth SE. Growth kinetics, selfrenewal, and the osteogenic potential of purified human mesenchymal stem cells during extensive subcultivation and following cryopreservation. J Cell Biochem. 1997;64:278–294. doi: 10.1002/(sici)1097-4644(199702)64:2<278::aid-jcb11>3.0.co;2-f. [DOI] [PubMed] [Google Scholar]
  • 42.Sekiya I, Larson BL, Smith JR, Pochampally R, Cui JG, Prockop DJ. Expansion of human adult stem cells from bone marrow stroma: conditions that maximize the yields of early progenitors and evaluate their quality. Stem Cells. 2002;20:530–541. doi: 10.1634/stemcells.20-6-530. [DOI] [PubMed] [Google Scholar]
  • 43.Haynesworth SE, Goshima J, Goldberg VM, Caplan AI. Characterization of cells with osteogenic potential from human marrow. Bone. 1992;13:81–88. doi: 10.1016/8756-3282(92)90364-3. [DOI] [PubMed] [Google Scholar]
  • 44.Stenderup K, Justesen J, Clausen C, Kassem M. Aging is associated with decreased maximal life span and accelerated senescence of bone marrow stromal cells. Bone. 200(333):919. doi: 10.1016/j.bone.2003.07.005. [DOI] [PubMed] [Google Scholar]
  • 45.Nishida S, Endo N, Yamagiwa H, Tanizawa T, Takahashi HE. Number of osteoprogenitor cells in human bone marrow markedly decreases after skeletal maturation. J Bone Miner Metab. 1999;17:171. doi: 10.1007/s007740050081. [DOI] [PubMed] [Google Scholar]
  • 46.Mueller SM, Glowacki J. Age-related decline in the osteogenic potential of human bone marrow cells cultured in three-dimensional collagen sponges. J Cell Biochem. 2001;82:583. doi: 10.1002/jcb.1174. [DOI] [PubMed] [Google Scholar]
  • 47.Zhang H, Fazel S, Tian H, Mickle DA, Weisel RD, Fujii T, Li RK. Increasing donor age adversely impacts beneficial effects of bone marrow but not smooth muscle myocardial cell therapy. Am J Physiol Heart Circ Physiol. 2005;289:H2089. doi: 10.1152/ajpheart.00019.2005. [DOI] [PubMed] [Google Scholar]
  • 48.Meyer FA, Laver-Rudich Z, Tanenbaum R. Evidence for a mechanical coupling of glycoprotein microfibrils with collagen fibrils in Wharton's jelly. Biochim Biophys Acta. 1983;755:376–387. doi: 10.1016/0304-4165(83)90241-6. [DOI] [PubMed] [Google Scholar]
  • 49.Gluckman E, HA Broxmeyer, AD Auerbach, HS Friedman, GW Douglas, A Devergie, H Esperou, D Thierry, G Socie, P Lehn, S Cooper, D English, J Kurtzberg, J Bard, EA Boyse. Hematopoietic reconstitution in a patient with Fanconi's anemia by means of umbilical-cord blood from an HLA-identical sibling. N Engl J Med. 1989;321:1174–1178. doi: 10.1056/NEJM198910263211707. [DOI] [PubMed] [Google Scholar]
  • 50.Broxmeyer HE, GW Douglas, G Hangoc, S Cooper, J Bard, D English, M Arny, L Thomas, EA Boyse. Human umbilical cord blood as a potential source of transplantable hematopoietic stem/progenitor cells. Proc Natl Acad Sci USA. 1989;86:3828–3832. doi: 10.1073/pnas.86.10.3828. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Gluckman E, V Rocha, A Boyer-Chammard, F Locatelli, W Arcese, R Pasquini, J Ortega, G Souillet, E Ferreira, JP Laporte, M Fernandez, C Chastang. Outcome of cord blood transplantation from related and unrelated donors. Eurocord Transplant Group and the European Blood and Marrow Transplantation Group. N Engl J Med. 1997;337:373–381. doi: 10.1056/NEJM199708073370602. [DOI] [PubMed] [Google Scholar]
  • 52.Han IS, JS Ra, MW Kim, EA Lee, HY Jun, SK Park, BS Kwon. Differentiation of CD34+ cells from human cord blood and murine bone marrow is suppressed by C6 betachemokines. Mol Cells. 2003;15:176–180. [PubMed] [Google Scholar]
  • 53.Kim SK, SK Koh, SU Song, SH Shin, GS Choi, WC Kim, MH Lee, JY Seoh, SK Park, JK Fraser. Ex vivo expansion and clonality of CD34+ selected cells from bone marrow and cord blood in a serum-free media. Mol Cells. 2002;14:367–373. [PubMed] [Google Scholar]
  • 54.Kögler G, Sensken S, Airey JA, Trapp T, Müschen M, Feldhahn N, Liedtke S, Sorg RV, Fischer J, Rosenbaum C, Greschat S, Knipper A, Bender J, Degistirici O, Gao J, Caplan AI, Colletti EJ, Almeida-Porada G, Müller HW, Zanjani E, Wernet P. A new human somatic stem cell from placental cord blood with intrinsic pluripotent differentiation potential. J Exp Med. 2004;200:123–35. doi: 10.1084/jem.20040440. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Kogler G, Sensken S, Wernet P. Comparative generation and characterization of pluripotent unrestricted somatic stem cells with mesenchymal stem cells from human cord blood. Exp Hematol. 2006;34:1589–95. doi: 10.1016/j.exphem.2006.07.011. [DOI] [PubMed] [Google Scholar]
  • 56.Sensken S, Waclawczyk S, Knaupp AS, Trapp T, Enczmann J, Wernet P, Kogler G. In vitro differentiation of human cord blood-derived unrestricted somatic stem cells towards an endodermal pathway. Cytotherapy. 2007;9:362–78. doi: 10.1080/14653240701320254. [DOI] [PubMed] [Google Scholar]
  • 57.Greschat S, Schira J, Kury P, Rosenbaum C, Souza Silva MA, Kogler G, Wernet P, Müller HW. Unrestricted somatic stem cells from human umbilical cord blood can be differentiated into neurons with a dopaminergic phenotype. Stem Cells Dev. 2008;17:221–32. doi: 10.1089/scd.2007.0118. [DOI] [PubMed] [Google Scholar]
  • 58.Wu LF, Wang NN, Liu YS, 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. 2009;15(10):2865–73. doi: 10.1089/ten.TEA.2008.0579. Oct. [DOI] [PubMed] [Google Scholar]
  • 59.Baksh D, Yao R, Tuan R. Comparison of prolif-erative and multilineage differentiation potential of human mesenchymal stem cells derived from umbilical cord and bone marrow. Stem Cells. 2007;25:1384. doi: 10.1634/stemcells.2006-0709. [DOI] [PubMed] [Google Scholar]
  • 60.Lu LL, Liu YJ, Yang SG, Zhao QJ, Wang X, Gong W, Han ZB, Xu ZS, Lu YX, Liu D, Chen ZZ, Han ZC. Isolation and characterization of human umbilical cord mesenchymal stem cells with hematopoiesis-supportive function and other potentials. Haematologica. 2006;91:1017. [PubMed] [Google Scholar]
  • 61.Can A, Karahuseyinoglu S. Concise review: human umbilical cord stroma with regard to the source of fetusderived stem cells. Stem Cells. 2007;25:2886. doi: 10.1634/stemcells.2007-0417. [DOI] [PubMed] [Google Scholar]
  • 62.Sarugaser R, Lickorish D, Baksh D, Hosseini MM, Davies JE. Human umbilical cord perivascular (HUCPV) cells: a source of mesenchymal progenitors. Stem Cells. 2005;23:220. doi: 10.1634/stemcells.2004-0166. [DOI] [PubMed] [Google Scholar]
  • 63.Wu KH, Zhou B, Lu SH, Feng B, Yang SG, Du WT, Gu DS, Han ZC, Liu YL. In vitro and in vivo differentiation of human umbilical cord derived stem cells into endothelial cells. J Cell Biochem. 2007;100:608. doi: 10.1002/jcb.21078. [DOI] [PubMed] [Google Scholar]
  • 64.Mitchell KE, Weiss ML, Mitchell BM, Martin P, Davis D, Morales L, Helwig B, Beerenstrauch M, Abou-Easa K, Hildreth T, Troyer D, Medicetty S. Matrix cells from Wharton's jelly form neurons and glia. Stem Cells. 2003;21:50–60. doi: 10.1634/stemcells.21-1-50. [DOI] [PubMed] [Google Scholar]
  • 65.Weiss ML, Medicetty S, Bledsoe AR, Rachakatla RS, Choi M, Merchav S, Luo Y, Rao MS, Velagaleti G, Troyer D. Human umbilical cord matrix stem cells: preliminary characterization and effect of transplantation in a rodent model of Parkinson's disease. Stem Cells. 2006;24:781–792. doi: 10.1634/stemcells.2005-0330. [DOI] [PubMed] [Google Scholar]
  • 66.Lu LL, Liu YJ, Yang SG, Zhao QJ, Wang X, Gong W, Han ZB, Xu ZS, Lu YX, Liu D, Chen ZZ, Han ZC. Isolation and characterization of human umbilical cord mesenchymal stem cells with hematopoiesis: supportive function and other potentials. Haematologica. 2006;91:1017–1026. [PubMed] [Google Scholar]
  • 67.Kern S, Eichler H, Stoeve J, Klüter H, Bieback K. Comparative analysis of mesenchymal stem cells from bone marrow, umbilical cord blood, or adipose tissue. Stem Cells. 2006;24:1294–301. doi: 10.1634/stemcells.2005-0342. [DOI] [PubMed] [Google Scholar]
  • 68.Park KS, YS Lee, KS Kang. In vitro neuronal and osteogenic differentiation of mesenchymal stem cells from human umbilical cord blood. J Vet Sci. 2006;7:343–348. doi: 10.4142/jvs.2006.7.4.343. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Mareschi K, E Biasin, W Piacibello, M Aglietta, E Madon, F Fagioli. Isolation of human mesenchymal stem cells:bone marrow versus umbilical cord blood. Haematologica. 2001;86:1099–1100. [PubMed] [Google Scholar]
  • 70.Goodwin HS, AR Bicknese, SN Chien, BD Bogucki, CO Quinn, DA Wall. Multilineage differentiation activity by cells isolated from umbilical cord blood: expression of bone, fat, and neural markers. Biol Blood Marrow Transplant. 2001;7:581–588. doi: 10.1053/bbmt.2001.v7.pm11760145. [DOI] [PubMed] [Google Scholar]
  • 71.Wexler SA, C Donaldson, P Denning-Kendall, C Rice, B Bradley, JM Hows. Adult bone marrow is a rich source of human mesenchymal ‘stem’ cells but umbilical cord and mobilized adult blood are not. Br J Haematol. 2003;121:368–374. doi: 10.1046/j.1365-2141.2003.04284.x. [DOI] [PubMed] [Google Scholar]
  • 72.Musina RA, Bekchanova ES, Belyavskii AV, Grinenko TS, Sukhikh GT. Umbilical cord blood mesenchymal stem cells. Bull Exp Biol Med. 2007;143:127–131. doi: 10.1007/s10517-007-0032-z. [DOI] [PubMed] [Google Scholar]
  • 73.Romanov YA, Svintsitskaya VA, Smirnov VN. Searching for alternative sources of postnatal human mesenchymal stem cells: candidate MSC-like cells from umbilical cord. Stem Cells. 2003;21:105–10. doi: 10.1634/stemcells.21-1-105. [DOI] [PubMed] [Google Scholar]
  • 74.Gutierrez-Rodriguez M, E Reyes-Maldonado, H Mayani. Characterization of the adherent cells developed in Dextertype long-term cultures from human umbilical cord blood. Stem Cells. 2000;18:46–52. doi: 10.1634/stemcells.18-1-46. [DOI] [PubMed] [Google Scholar]
  • 75.Zhang LH, Liu YJ, Lu LL, Wang AP, Xu ZS, Zhu XP. Mesenchymal stem cells derived from human umbilical cord inhibit activation and proliferation of allogeneic umbilical cord blood T lymphocytes. Chin J Cancer Biother (Chin) 2006;13:191–195. [Google Scholar]
  • 76.Wang HS, Hung SC, Peng ST, Huang CC, Wei HM, Guo YJ, Fu YS, Lai MC1, Chen CC. Mesenchymal stem cells in the Wharton’ s jelly of the human umbilical cord. Stem Cells. 2004;22:1330–1337. doi: 10.1634/stemcells.2004-0013. [DOI] [PubMed] [Google Scholar]
  • 77.Fu YS, Cheng YC, Lin MY, Cheng H, Chu PM, Chou SC, Shih HY, Ko MH, Sung MS. Conversion of human umbilical cord mesenchymal stem cells in Wharton's jelly to dopaminergic neurons in vitro: potential therapeutic application for Parkinsonism. Stem Cells. 2006;24:115–124. doi: 10.1634/stemcells.2005-0053. [DOI] [PubMed] [Google Scholar]
  • 78.Friedman R, Betancur M, Boissel L, Tuncer H, Cetrulo C, Klingemann H. Umbilical cord mesenchymal stem cells: adjuvants for human cell transplantation. Biol Blood Marrow Transplant. 2007;13:1477–1486. doi: 10.1016/j.bbmt.2007.08.048. [DOI] [PubMed] [Google Scholar]
  • 79.Majumdar MK, Keane-Moore M, Buyaner D, Hardy WB, Moorman MA, McIntosh KR, Mosca JD. Characterization and functionality of cell surface molecules on human mesenchymal stem cells. Journal of Biomedical Science. 2003;10:228–241. doi: 10.1007/BF02256058. [DOI] [PubMed] [Google Scholar]
  • 80.BogenriederT, Herlyn M. Axis of evil: molecular mechanisms of cancer metastasis. Oncogene. 2003;22:6524–6536. doi: 10.1038/sj.onc.1206757. [DOI] [PubMed] [Google Scholar]
  • 81.Baksh D, Yao R, Tuan RS. Comparison of proliferative and multilineage differentiation potential of human mesenchymal stem cells derived from umbilical cord and bone marrow. Stem cells. 2007;25:1384–1392. doi: 10.1634/stemcells.2006-0709. [DOI] [PubMed] [Google Scholar]
  • 82.Surbek DV, Visca E, Steinmann C, Tichelli A, Schatt S, Hahn S, Gratwohl A, Holzgreve W. Umbilical cord blood collection before placental delivery during cesarean delivery increases cord blood volume and nucleated cell number available for transplantation. American Journal of Obstetrics and Gynecology. 2000;183:218–221. doi: 10.1067/mob.2000.105744. [DOI] [PubMed] [Google Scholar]
  • 83.Solves P, Mirabet V, Larrea L, Moraga R, Planelles D, Saucedo E, Uberos FC, Planells T, Guillen M, Andres A, Monleon J, Soler MA, Franco E. Comparison between two cord blood collection strategies. Acta Obstetricia et Gyne-cologica Scandinavica. 2003;82:439–442. doi: 10.1034/j.1600-0412.2003.00129.x. [DOI] [PubMed] [Google Scholar]
  • 84.Wong A, Yuen PMP, Li K, Yu LM, Tsoi WC. Cord blood collection before and after placental delivery: levels of nucleated cells, hematopoietic progenitor cells, leukocyte subpopulations and macroscopic clots. Bone Marrow Transplantation. 2001;27:133–138. doi: 10.1038/sj.bmt.1702757. [DOI] [PubMed] [Google Scholar]
  • 85.Lasky LC, Lane TA, Miller JP, Lindgren B, Patterson HA, Haley NR, Ballen K. In utero or ex utero cord blood collection: which is better? Transfusion. 2001;42:1261–1267. doi: 10.1046/j.1537-2995.2002.t01-1-00177.x. [DOI] [PubMed] [Google Scholar]
  • 86.Broxmeyer HE, Kurtzburg J, Gluckman E. Umbilical cord blood hematopoietic stemand re-populating cells in human clinical transplantation: an expanded role for cord blood transplantation. Blood Cells. 1991;17:330–337. [PubMed] [Google Scholar]
  • 87.Harris DT, Schumacher MJ, Rychlik S, Booth A, Acevedo A, Rubinstein P, Bard J, Boyse EA. Collection, separation and cryopreservation of umbilical cord blood in transplantation. Bone Marrow Transplantation. 1994;13:135–143. [PubMed] [Google Scholar]
  • 88.Bertolini F, Lazzari L, Lauri E, Corsini C, Castelli C, Gorini F, Sirchia G. Comparative study of different procedures for the collection and banking of umbilical cord blood. Journal of Hematotherapy. 1995;4:29–36. doi: 10.1089/scd.1.1995.4.29. [DOI] [PubMed] [Google Scholar]
  • 89.Turner CW, Luzins J, Hutcheson CA. Modified harvest technique for cord blood hematopoietic stem cells. Bone Marrow Transplantation. 1992;10:89–91. [PubMed] [Google Scholar]
  • 90.Skoric D, Balint B, Petakov M, Sindjic M, Rodic P. Collection strategies and cryopreservation of umbilical cord blood. Transfus Med. 2007;17:107–13. doi: 10.1111/j.1365-3148.2007.00728.x. [DOI] [PubMed] [Google Scholar]
  • 91.Berz D, McCormack EM, Winer ES, Colvin GA, Quesenberry PG. Cryopreservation of hematopoietic stem cells. Am J Hematol. 2007;82:1–8. doi: 10.1002/ajh.20707. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Katayamaa Y, Yano T, Bessho A, Deguchi S, Sunami K, Mahmut N, Shinagawa K, Omoto E, Makino S, Miyamoto T, Mizuno S, Funkuda T, Eto T, Fujisaki T, Ohno Y, Inaba S, Niho Y, Harada M. The effect of a simplified method for cryopreservation and thawing procedures on peripheral blood stem cells. Bone Marrow Transplant. 1997;19:283–287. doi: 10.1038/sj.bmt.1700644. [DOI] [PubMed] [Google Scholar]
  • 93.Koliakos A, Alamdari DH, Tsagias N, Kouzi-Koliakos K, Michaloudi E, Karagiannis V. A novel high-yield volume-reduction method for the cryopreservation of UC blood units. Cyto-therapy. 2007;9:654–659. doi: 10.1080/14653240701508445. [DOI] [PubMed] [Google Scholar]
  • 94.Meyer TPH, Hofmann B, Zaisserer J, Jacobs VR, Fuchs B, Rapp S, Weinauer F, Burkhart J. Analysis and cryopreservation of hematopoietic stem and progenitor cells from umbilical cord blood. Cytotherapy. 2006;8:265–276. doi: 10.1080/14653240600735685. [DOI] [PubMed] [Google Scholar]
  • 95.Choi CW, Kim BS, Seo JH, Shin SW, Kim YH, Kim JS. Long-term engraftment stability of peripheral blood stem cells cryopreserved using the dump-freezing method in a −80 °C mechanical freezer with 10% dimethyl sulfoxide. Int J Hematol. 2007;3:245–250. doi: 10.1007/BF02981945. [DOI] [PubMed] [Google Scholar]
  • 96.Shlebak AA, Marley SB, Roberts IA, Davidson RJ, Goldman JM, Gordon MY. Optimal timing for processing and cryopreservation of umbilical cord haematopoietic stem cells for clinical transplantation. Bone Marrow Transplant. 1999;23:131–136. doi: 10.1038/sj.bmt.1701551. [DOI] [PubMed] [Google Scholar]
  • 97.Abrahamsen JF, Bakken AM, Bruserud O. Cryo-preserving human peripheral blood progenitor cells with 5-percent rather than 10-percent DMSO results in less apoptosis and necrosis in CD34+ cells. Transfusion. 2002;42:1537–1580. doi: 10.1046/j.1537-2995.2002.00242.x. [DOI] [PubMed] [Google Scholar]
  • 98.Galmés A, Besalduch J, Bargay J, Novo A, Morey M, Guerra JM, Duran MA. Long term storage at 80 C of hematopoietic progenitor cells with 5% dimethyl sulfoxide as the sole cryoprotectant. Transfusion. 1999;39:70–73. doi: 10.1046/j.1537-2995.1999.39199116897.x. [DOI] [PubMed] [Google Scholar]
  • 99.Halle P, Tournilhac O, Knopinska-Posluszny W, Kanold J, Gembara P, Boiret N, Rapatel C, Berger M, Travade P, Angielski S, Bonhomme J, Deméocq F. Uncontrolled-rate freezing and storage at −80 °C with only 3.5% DMSO in cryo-protective solution for 109 autologous peripheral blood progenitor cell transplantations. Transfusion. 2001;41:667–673. doi: 10.1046/j.1537-2995.2001.41050667.x. [DOI] [PubMed] [Google Scholar]
  • 100.Heo YJ, Son CH, Chung JS, Park YS, Son JH. The cryopreservation of high concentrated PBMC for dendritic cell (DC)-based cancer immunotherapy. Cryobiology. 2009;59:203–209. doi: 10.1016/j.cryobiol.2008.12.006. [DOI] [PubMed] [Google Scholar]
  • 101.Donaldson C, Armitage WJ, Denning-Kendall PA, Nicol AJ, Bradley BA, Hows JM. Optimal cryopreservation of human umbilical cord blood. Bone Marrow Transplantation. 1996;18:725–731. [PubMed] [Google Scholar]
  • 102.Hunt CJ, Armitage SE, Pegg DE. Cryopreservation of umbilical cord blood: 2. Tolerance of CD341 cells to multimolar dimethyl sulphoxide and the effect of cooling rate on recovery after freezing and thawing. Cryobiology. 2003;46:76–87. doi: 10.1016/s0011-2240(02)00181-5. [DOI] [PubMed] [Google Scholar]
  • 103.Balint B, Ivanovic Z, Petakov M, Taseski J, Jovcic G, Stojanovic N, Milenkovic P. The cryopreservation protocol optimal for progenitor recovery is not optimal for preservation of marrow repopulating ability. Bone Marrow Transplantation. 1999;23:613–619. doi: 10.1038/sj.bmt.1701623. [DOI] [PubMed] [Google Scholar]
  • 104.Gardner DK, Sheehan CB, Rienzi L, Katz-Jaffe M, Larman MG. Analysis of oocyte physiology to improve cryopreservation procedures. Theriogenology. 2007;67:64–72. doi: 10.1016/j.theriogenology.2006.09.012. [DOI] [PubMed] [Google Scholar]
  • 105.Yamada C, Caetano HV, Simões R, Nicacio AC, Feitosa WB, Assumpção ME, Visintin JA. Immature bovine oocyte cryopreservation: comparison of different associations with ethylene gly-col, glycerol and dimethylsulfoxide. Anim. Reprod. Sci. 2007;99:384–388. doi: 10.1016/j.anireprosci.2006.07.001. [DOI] [PubMed] [Google Scholar]
  • 106.Son JH, Heo YJ, Park MY, Kim HH, Lee KS. Optimization of cryopreservation condition for hematopoietic stem cells from umbilical cord blood. Cryobiology. 2010;60:287–92. doi: 10.1016/j.cryobiol.2010.01.007. [DOI] [PubMed] [Google Scholar]
  • 107.Armson BA. Maternal/Fetal Medicine Committee, Society of Obstetricians and Gynaecologists of Canada. Umbilical cord blood banking: implications for perinatal care providers. J Obstet Gynaecol Can. 2005;27:673. doi: 10.1016/s1701-2163(16)30520-5. [DOI] [PubMed] [Google Scholar]
  • 108. Opinion of the european group on ethics in science and new technologies to the european commission No 19 16th March 2004.
  • 109.Bellomo M. The Stem Cell Divide: The Facts, the Fiction, and the Fear Driving the Greatest Scientific, Political, and Religious Debate of Our Time. Edited by AMACOM 2006, ISBN: 0814408818.
  • 110.American Academy of Pediatrics Policy Statement, editor. PEDIATRICS Vol. 119 No. 1 January 2007, pp. 165–170.
  • 111.Sullivan MJ. Banking on cord blood stem cells. Nat Rev Cancer. 2008;8:555–63. doi: 10.1038/nrc2418. [DOI] [PubMed] [Google Scholar]
  • 112.Nietfeld JJ, Pasquini MC, Logan BR, Verter F, Horowitz MM. Lifetime probabilities of hematopoietic stem cell transplantation in the U.S. Biol Blood Marrow Transplant. 2008;14:316–22. doi: 10.1016/j.bbmt.2007.12.493. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113. The complete FDA 1271. American regulations for human reproductive tissue banks.
  • 114.U.S. Department of Health and Human Services Food and Drug Administration Center for Biologics Evaluation and Research October 2009, editor. Guidance for Industry Minimally Manipulated, Unrelated Allogeneic Placental/Umbilical Cord Blood Intended for Hematopoietic Reconstitution for Specified Indications.
  • 115. Opinion of the european group on ethics in science and new technologies to the european commission, 16th March 2004 Ethical aspects of umbilical cord blood banking.
  • 116.Colter DC, Class R, DiGirolamo CM, Prockop DJ. Rapid expansion of recycling stem cells in cultures of plastic-adherent cells from human bone marrow. Proc. Natl. Acad. Sci. USA. 2000;97:3213–18. doi: 10.1073/pnas.070034097. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Sekiya I, Larson BL, Smith JR, Pochampally R, Cui JG, Prockop DJ. Expansion of human adult stem cells from bone marrow stroma: conditions that maximize the yields of early progenitors and evaluate their quality. Stem Cells. 2002;20:530–41. doi: 10.1634/stemcells.20-6-530. [DOI] [PubMed] [Google Scholar]
  • 118.Lee MW, Yang MS, Park JS, Kim HC, Kim YJ, Choi J. Isolation of mesenchymal stem cells from cryopreserved human umbilical cord blood. Int J Hematol. 2005;81:126–30. doi: 10.1532/ijh97.a10404. [DOI] [PubMed] [Google Scholar]
  • 119.Kotobuki N, Hirose M, Takakura Y, Ohgushi H. Cultured autologous human cells for hard tissue regeneration: preparation and characterization of mesenchymal stem cells from bone marrow. Artif Organs. 2004;28:33–39. doi: 10.1111/j.1525-1594.2004.07320.x. [DOI] [PubMed] [Google Scholar]
  • 120.Le Blanc K, Tammik C, Rosendahl K, Zetterberg E, Ringden O. HLA expression and immunologic properties of differentiated and undifferentiated mesenchymal stem cells. Exp Hematol. 2003;31:890. doi: 10.1016/s0301-472x(03)00110-3. [DOI] [PubMed] [Google Scholar]
  • 121.Tse WT, Pendleton JD, Beyer WM, Egalka MC, Guinan EC. Suppression of allogeneic T cell proliferation by human marrow stromal cells: implications in transplantation. Transplantation. 2003;75:389. doi: 10.1097/01.TP.0000045055.63901.A9. [DOI] [PubMed] [Google Scholar]
  • 122.Krampera M, Glennie S, Dyson J, Scott D, Laylor R, Simpson E, et al. Bone marrow mesenchymal stem cells inhibit the response of naive and memory antigen-specific T cells to their cognate peptide. Blood. 2003;101:3722. doi: 10.1182/blood-2002-07-2104. 20,21. [DOI] [PubMed] [Google Scholar]
  • 123.Horwitz EM, Prockop DJ, Fitzpatrick LA, Koo WW, Gordon PL, Neel M, Sussman M, Orchard P, Marx JC, Pyeritz RE, Brenner MK. Transplantability and therapeutic effects of bone marrowderived mesenchymal cells in children with osteogenesis imperfecta. Nat Med. 1999;5:309–313. doi: 10.1038/6529. [DOI] [PubMed] [Google Scholar]
  • 124.Di Nicola M, Carlo-Stella C, Magni M, Milanesi M, Longoni PD, Matteucci P, Grisanti S, Gianni AM. Human bone marrow stromal cells suppress T-lymphocyte proliferation induced by cellular or nonspecific mitogenic stimuli. Blood. 2002;99:3838–3843. doi: 10.1182/blood.v99.10.3838. [DOI] [PubMed] [Google Scholar]
  • 125.Brittan M, Chance V, Elia G, Poulsom R, Alison MR, MacDonald TT, Wright NA. A regenerative role for bone marrow following experimental colitis: contribution to neovasculogenesis and myofibroblasts. Gastroenterology. 2005;128:1984–1995. doi: 10.1053/j.gastro.2005.03.028. [DOI] [PubMed] [Google Scholar]
  • 126.Schneider RK, Neuss S, Stainforth R, Laddach N, Bovi M, Knuechel R, Perez-Bouza A. Three-dimensional epidermis-like growth of human mesenchymal stem cells on derma equivalents: contribution to tissue organization by adaptation of myofibroblastic phenotype and function. Differentiation. 2008;76:156–16725. doi: 10.1111/j.1432-0436.2007.00204.x. [DOI] [PubMed] [Google Scholar]
  • 127.Ripa RS, Haack-Sørensen M, Wang Y, Jørgen-sen E, Mortensen S, Bindslev L, Friis T, Kastrup J. Bone marrow derived mesenchymal cell mobilization by granulocyte-colony stimulating factor after acute myocardial infarction: results from the Stem Cells in Myocardial Infarction (STEMMI) trial. Circulation. 2007;116:24–30. doi: 10.1161/CIRCULATIONAHA.106.678649. [DOI] [PubMed] [Google Scholar]
  • 128.Chen SL, Fang WW, Ye F, Liu YH, Qian J, Shan SJ, Zhang JJ, Chunhua RZ, Liao LM, Lin S, Sun JP. Effect on left ventricular function of intra-coronary transplantation of autologous bone marrow mesenchymal stem cell in patients with acute myocardial infarction. Am J Cardiol. 2004;94:92–95. doi: 10.1016/j.amjcard.2004.03.034. [DOI] [PubMed] [Google Scholar]
  • 129.Lee PH, Kim JW, Bang OY, Ahn YH, Joo IS, Huh K. Autologous mesenchymal stem cell therapy delays the progression of neurological deficits in patients with multiple system atrophy. Clin Pharmacol Ther. 2008;83:723–30. doi: 10.1038/sj.clpt.6100386. [DOI] [PubMed] [Google Scholar]
  • 130.Bang OY, Lee JS, Lee PH, Lee G. Autologous mesenchymal stem cell transplantation in stroke patients. Ann Neurol. 2005;57:874–82. doi: 10.1002/ana.20501. [DOI] [PubMed] [Google Scholar]
  • 131.Lazarus HM, Koc ON, Devine SM, Curtin P, Maziarz RT, Holland HK, Shpall EJ, McCarthy P, Atkinson K, Cooper BW, Gerson SL, Laughlin MJ, Loberiza FR, Jr, Moseley AB, Bacigalupo A. Cotransplantation of HLA identical sibling culture-expanded mesenchymal stem cells and hematopoietic stem cells in hematologic malignancy patients. Biol Blood Marrow Transplant. 2005;11:389–98. doi: 10.1016/j.bbmt.2005.02.001. [DOI] [PubMed] [Google Scholar]
  • 132.Ringdén O, Uzunel M, Rasmusson I, Remberger M, Sundberg B, Lönnies H, Marschall HU, Dlugosz A, Szakos A, Hassan Z, Omazic B, As-chan J, Barkholt L, Le Blanc K. Mesenchymal stem cells for treatment of therapy-resistant graft-versus-host disease. Transplantation. 2006;81:1390–7. doi: 10.1097/01.tp.0000214462.63943.14. [DOI] [PubMed] [Google Scholar]
  • 133.Herrera MB, Bussolati B, Bruno S, Fonsato V, Romanazzi GM, Camussi G. Mesenchymal stem cells contribute to the renal repair of acute tubular epithelial injury. Int J Mol Med. 2004;14:1035–1041. [PubMed] [Google Scholar]
  • 134.Ortiz LA, Gambelli F, McBride C, Gaupp D, Bad-doo M, Kaminski N, Phinney DG. Mesenchymal stem cell engraftment in lung is enchanced in response to bleomycin exposure and ameliorates its fibrotic effects. Proc Natl Acad Sci USA. 2003;100:8407–8411. doi: 10.1073/pnas.1432929100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Hernigou P, Beaujean F. Treatment of osteonecrosis with autologous bone marrow grafting. Clin Orthop Relat Res. 2002;405:14–23. doi: 10.1097/00003086-200212000-00003. [DOI] [PubMed] [Google Scholar]
  • 136.Badiavas EV, Falanga V. Treatment of chronic wounds with bone marrow derived cells. Arch Dermatol. 2003;139:510–516. doi: 10.1001/archderm.139.4.510. [DOI] [PubMed] [Google Scholar]
  • 137.Orlic D, Kajstura J, Chimenti S, et al. Bone marrow cells regenerate infracted myocardium. Nature. 2001;410:701–5. doi: 10.1038/35070587. [DOI] [PubMed] [Google Scholar]
  • 138.Beltrami AP, Barlucchi L, Torella D, et al. Adult cardiac stem cells are multipotent and support myocardial regeneration. Cell. 2003;114:763–76.12. doi: 10.1016/s0092-8674(03)00687-1. [DOI] [PubMed] [Google Scholar]
  • 139.Tomita S, Li RK, Weisel RD, Mickle DAG, Kim EJ, Sakai T, Jia ZQ. Autologous transplantation of bone marrow cells improves damaged heart function. Circulation. 1999;100:II247–256. doi: 10.1161/01.cir.100.suppl_2.ii-247. [DOI] [PubMed] [Google Scholar]
  • 140.Toma C, Pittenger MF, Cahill KS, et al. Human mesenchymal stem cells differentiate to a car-diomyocyte phenotype in the adult murine heart. Circulation. 2002;105:93–8. doi: 10.1161/hc0102.101442. [DOI] [PubMed] [Google Scholar]
  • 141.Assmus B, Schächinger V, Teupe C, Britten M, Lehmann R, Döbert N, Grünwald F, Aicher A, Urbich C, Martin H, Hoelzer D, Dimmeler S, Zeiher AM. Transplantation of progenitor cells and regeneration enhancement in acute myocardial infarction (topcare-ami) Circulation. 2002;106:3009–3017. doi: 10.1161/01.cir.0000043246.74879.cd. [DOI] [PubMed] [Google Scholar]
  • 142.Strauer BE, Brehm M, Zeus T, Kostering M, Hernandez A, Sorg RV, Kogler G, Wernet P. Repair of infarcted myocardium by autologous intracoronary mononuclear bone marrow cell transplantation in humans. Circulation. 2002;106:1913–1918. doi: 10.1161/01.cir.0000034046.87607.1c. [DOI] [PubMed] [Google Scholar]
  • 143.Perin EC, Dohmann HF, Borojevic R, Silva SA, Sousa AL, Mesquita CT, Rossi MI, Carvalho AC, Dutra HS, Dohmann HJ, Silva GV, Belém L, Vivacqua R, Rangel FO, Esporcatte R, Geng YJ, Vaughn WK, Assad JA, Mesquita ET, Willerson JT. Transendocardial, autologous bone marrow cell transplantation for severe, chronic ischemic heart failure. Circulation. 2003;107:2294–2302. doi: 10.1161/01.CIR.0000070596.30552.8B. [DOI] [PubMed] [Google Scholar]
  • 144.Tse HF, Kwong YL, Chan JK, Lo G, Ho CL, Lau CP. Angiogenesis in ischaemic myocardium by intra myocardial autologous bone marrow mononuclear cell implantation. Lancet. 2003;361:47–49. doi: 10.1016/S0140-6736(03)12111-3. [DOI] [PubMed] [Google Scholar]
  • 145.Hare JM, Traverse JH, Henry TD, Dib N, Strumpf RK, Schulman SP, Gerstenblith G, DeMaria AN, Denktas AE, Gammon RS, Hermiller JB, Jr, Reis-man MA, Schaer GL, Sherman W. A randomized, double-blind, placebo-controlled, dose-escalation study of intravenous adult human mesenchymal stem cells (prochymal) after acute myocardial infarction. J. Am Coll Cardiol. 2009;54:2277–2286. doi: 10.1016/j.jacc.2009.06.055. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146.Chen S, Liu Z, Tian N, Zhang J, Yei F, Duan B, Zhu Z, Lin S, Kwan TW. Intracoronary transplantation of autologous bone marrow mesenchymal stem cells for ischemic cardiomyopathy due to isolated chronic occluded left anterior descending artery. J Invasive Cardiol. 2006;18:552–556. [PubMed] [Google Scholar]
  • 147.Katritsis DG, Sotiropoulou PA, Karvouni E, Karabinos I, Korovesis S, Perez SA, Voridis EM, Papamichail M. Transcoronary transplantation of autologous mesenchymal stem cells and endothelial progenitors into infarcted human myocardium. Catheter Cardiovasc Interv. 2005;65:321–329. doi: 10.1002/ccd.20406. [DOI] [PubMed] [Google Scholar]
  • 148.Koc ON, Peters C, Aubourg P, Raghavan S, Dyhouse S, DeGasperi R, et al. Bone marrow derived mesenchymal stem cells remain host-derived despite successful hematopoietic engraftment after allogeneic transplantation in patients with lysosomal and peroxisomal storage diseases. Exp Hemtaol. 1999;27:1675–1681. doi: 10.1016/s0301-472x(99)00101-0. [DOI] [PubMed] [Google Scholar]
  • 149.Koc ON, Day J, Nieder M, Gerson SL, Lazarus HM, Krivit W. Allogeneic mesenchymal stem cell infusion for treatment of metachromatic leukodystrophy (MLD) and Hurler syndrome (MPS-IH) Bone Marrow Transpl. 2002;30:215–22. doi: 10.1038/sj.bmt.1703650. [DOI] [PubMed] [Google Scholar]
  • 150.Horwitz EM, Gordon PL, Koo WK, Marx JC, Neel MD, McNall RY, Muul L, Hofmann T. Isolated allogeneic bone marrow-derived mesenchymal cells engraft and stimulate growth in children with osteogenesis imperfecta: implications for cell therapy of bone. Proc Natl Acad Sci USA. 2002;99:8932–37. doi: 10.1073/pnas.132252399. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151.Muschler GF, Nitto H, Matsukura Y, Boehm C, Valdevit A, Kambic H, Davros W, Powell K, Easley K. Spine fusion using cell matrix composites enriched in bone marrow-derived cells. Clinical Orthopaedics. 2003;407:102–118. doi: 10.1097/00003086-200302000-00018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152.Quarto R, Mastrogiacomo M, Cancedda R, Ku-tepov SM, Mukhachev V, Lavroukov A, Kon E, Marcacci M. Repair of large bone defects with the use of autologous bone marrow stromal cells. The New England Journal of Medicine. 2001;344:385–386. doi: 10.1056/NEJM200102013440516. [DOI] [PubMed] [Google Scholar]
  • 153.Krebsbach P H, Mankani MH, Satomura K, Kuznetsov SA, Robey PG. Repair of craniotomy defects using bone marrow stromal cells. Transplantation. 1998;66:1272–1278. doi: 10.1097/00007890-199811270-00002. [DOI] [PubMed] [Google Scholar]
  • 154.Ponticiello MS, Schinagl RM, Kadiyala S, Barry FP. Gelatin-based resorbable sponge as a carrier matrix for human mesenchymal stem cells in cartilage regeneration therapy. Journal of Biomedical Materials Research. 2000;52:246–255. doi: 10.1002/1097-4636(200011)52:2<246::aid-jbm2>3.0.co;2-w. [DOI] [PubMed] [Google Scholar]
  • 155.Satoh H, Kishi K, Tanaka T, Kubota Y, Nakajima T, Akasaka Y, Ishii T. Transplanted mesenchymal stem cells are effective for skin regeneration in acute cutaneous wounds. Cell Transplant. 2004;13:405–412. doi: 10.3727/000000004783983765. 100. [DOI] [PubMed] [Google Scholar]
  • 156.Wu Y, Chen L, Scott PG, Tredget EE. Mesenchymal stem cells enhance wound healing through differentiation and angiogenesis. Stem Cells. 2007;25:2648–2659. doi: 10.1634/stemcells.2007-0226. [DOI] [PubMed] [Google Scholar]
  • 157.Chen L, Tredget EE, Wu PY, Wu Y. Paracrine factors of mesenchymal stem cells recruit macrophages and endothelial lineage cells and enhance wound healing. PLoS ONE. 2008;3:e1886. doi: 10.1371/journal.pone.0001886. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158.Falanga V, Iwamoto S, Chartier M, Yufit T, Butmarc J, Kouttab N, Shrayer D, Carson P. Autologous bone marrow-derived cultured mesenchymal stem cells delivered in a fibrin spray accelerate healing in murine and human cutaneous wounds. Tissue Eng. 2007;13:1299–1312. doi: 10.1089/ten.2006.0278. [DOI] [PubMed] [Google Scholar]
  • 159.Kim SS, Song CK, Shon SK, Lee KY, Kim CH, Lee MJ, Wang L. Effects of human amniotic membrane grafts combined with marrow mesenchymal stem cells on healing of full-thickness skin defects in rabbits. Cell Tissue Res. 2009;336:59–66. doi: 10.1007/s00441-009-0766-1. [DOI] [PubMed] [Google Scholar]
  • 160.Arnalich-Montiel F, Pastor S, Blazquez-Martinez A, Fernandez-Delgado J, Nistal M, Alio L, De Miguel MP. Adiposederived stem cells are a source for cell therapy of the corneal stroma. Stem Cells. 2008;26:570–579. doi: 10.1634/stemcells.2007-0653. [DOI] [PubMed] [Google Scholar]
  • 161.Oh JY, Kim MK, Shin MS, Lee HJ, Ko JH, Wee WR, Lee JH. The anti-inflammatory and anti-angiogenic role of mesenchymal stem cells in corneal wound healing following chemical injury. Stem Cells. 2008;26:1047–1055. doi: 10.1634/stemcells.2007-0737. [DOI] [PubMed] [Google Scholar]
  • 162.Qian H, Yang H, Xu W, Yan Y, Chen Q, Zhu W, Cao H, Yin Q, Zhou H, Mao F, Chen Y. Bone marrow mesenchymal stem cells ameliorate rat acute renal failure by differentiation into renal tubular epithelial-like cells. Int J Mol Med. 2008;22:325–332. [PubMed] [Google Scholar]
  • 163.Morigi M, Introna M, Imberti B, Corna D, Abbate M, Rota C, Rottoli D, Benigni A, Perico N, Zoja C, Rambaldi A, Remuzzi A, Remuzzi G. Human bone marrow mesenchymal stem cells accelerate recovery of acute renal injury and prolong survival in mice. Stem Cells. 2008;26:2075–2082. doi: 10.1634/stemcells.2007-0795. [DOI] [PubMed] [Google Scholar]
  • 164.Munoz-Elias G, Woodbury D, Black IB. Marrow stromal cells, mitosis, and neuronal differentiation: stem cell and precursor functions. Stem Cells. 2003;21:437–448. doi: 10.1634/stemcells.21-4-437. [DOI] [PubMed] [Google Scholar]
  • 165.Bouchez G, Sensebe' L, Vourc'h P, Garreau L, Bodard S, Rico A, Guilloteau D, Charbord P, Besnard JC, Chalon S. Partial recovery of dopaminergic pathway after graft of adult mesenchymal stem cells in a rat model of Parkinson's disease. Neurochem Int. 2008;52:1332–1342. doi: 10.1016/j.neuint.2008.02.003. [DOI] [PubMed] [Google Scholar]
  • 166.Rossignol J, Boyer C, Lévêque X, Dunbar GL, Lescaudron L. Mesenchymal stem cell transplants reduce behavioral deficits in the 3-nitropropionic rat model of Huntington's disease. Cell Transplant. 2009;18:233. [Google Scholar]
  • 167.Venkataramana NK, Kumar SK, Balaraju S, et al. Open-labeled study of unilateral autologous bone-marrow-derived mesenchymal stem cell transplantation in Parkinson's disease. Transl Res. 2010;155:62–70. doi: 10.1016/j.trsl.2009.07.006. [DOI] [PubMed] [Google Scholar]
  • 168.Schwarz J, Storch A. Transplantation in Parkinson's disease: will mesenchymal stem cells help to re-enter the clinical arena? Transl Res. 2010;155:55–6. doi: 10.1016/j.trsl.2009.08.008. [DOI] [PubMed] [Google Scholar]
  • 169.McGuckin CP, Forraz N, Allouard Q, Pettengell R. Umbilical cord blood stem cells can expand hematopoietic and neuroglial progenitors in vitro. Experimental Cell Research. 2004;295:350–359. doi: 10.1016/j.yexcr.2003.12.028. [DOI] [PubMed] [Google Scholar]
  • 170.McGuckin C, Forraz N, Baradez MO, Navran S, Zhao J, Urban R, Tilton R, Denner L. Production of stem cells with embryonic characteristics from human umbilical cord blood. Cell Proliferation. 2005:245–255. doi: 10.1111/j.1365-2184.2005.00346.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 171.Rogers I, Yamanaka N, Bielecki R, Wong CJ, Chua S, Yuen S, Casper RF. Identification and analysis of in vitro cultured CD45-positive cells capable of multi-lineage differentiation. Experimental Cell Research. 2007;313:1839–1852. doi: 10.1016/j.yexcr.2007.02.029. [DOI] [PubMed] [Google Scholar]
  • 172.Kucia M, Halasa M, Wysoczynski M, Baskiewicz-Masiuk M, oldenhawer S, Zuba-Surma E, Czajka R, Wojakowski W, achalinski B, Ratajczak MZ. Morphological and mlecular characterization of novel population of CXCR4+ SEA-4+ Oct-4+ very small embryonic-like cells purified from human umbilical cord blood-preliminary report. Leukemia. 2007;21:297–303. doi: 10.1038/sj.leu.2404470. [DOI] [PubMed] [Google Scholar]
  • 173.Harris DT, He X, Badowski M, Nichols JC. Regenerative medicine of the eye: a short review. Stem Cell Repair & Regeneration. 2008;3 [Google Scholar]
  • 174.Sunkomat JNE, Goldman S, Harris DT. Cord bloodderived MNCs delivered intracoronary contribute differently to vascularization compared to CD34+ cells in the rat model of acute ischemia. Journal of Molecular and Cellular Cardiology. 2007;42(Suppl. 1):S97. [Google Scholar]
  • 175.Harris DT, Rogers I. Umbilical cord blood: a unique source of pluripotent stem cells for regenerative medicine. Current Stem Cell Research & Therapy. 2007;2:301–309. doi: 10.2174/157488807782793790. [DOI] [PubMed] [Google Scholar]
  • 176.Sanberg PR, Willing AE, Garbuzova-Davis S, Saporta S, Liu G, Sanberg CD, Bickford PC, Klasko SK, El-Badri NS. Umbilical cord blood-derived stem cells and brain repair. Ann N Y Acad Sci. 2005;1049:67–83. doi: 10.1196/annals.1334.008. [DOI] [PubMed] [Google Scholar]
  • 177.Chung DJ, Choi CB, Lee SH, Kang EH, Lee JH, Hwang SH, Han H, Lee JH, Choe BY, Lee SY, Kim HY. Intraarterially delivered human umbilical cord blood-derived mesenchymal stem cells in canine cerebral ischemia. J Neurosci Res. 2009;87:3554–3567. doi: 10.1002/jnr.22162. [DOI] [PubMed] [Google Scholar]
  • 178.Jeong CH, Lim JY, Park SI, Kim SM, Jun JA, Oh W, Lee JW, Jeun SS. Human umbilical cord blood-derived mesencymal stem cell therapy for stroke in rat. Tissue Eng. and Reg. Med. 2006;3:445–450. [Google Scholar]
  • 179.Chang YS, Oh W, Choi SJ, Sung DK, Kim SY, Choi EY, Kang S, Jin HJ, Yang YS, Park WS. Human umbilical cord blood-derived mesenchymal stem cells attenuate hyperoxia-induced lung injury in neonatal rats. Cell Transplant. 2009;18(8):869–86. doi: 10.3727/096368909X471189. [DOI] [PubMed] [Google Scholar]
  • 180.Cao H, Qian H, Xu W, Zhu W, Zhang X, Chen Y, Wang M, Yan Y, Xie Y. Mesenchymal stem cells derived from human umbilical cord ameliorate ischemia/reperfusion-induced acute renal failure in rats. Biotechnol Lett. 2010;32(5):725–32. doi: 10.1007/s10529-010-0207-y. May. [DOI] [PubMed] [Google Scholar]
  • 181.Morigi M, Rota C, Montemurro T, Montelatici E, Lo Cicero V, Imberti B, Abbate M, Zoja C, Cassis P, Longaretti L, Rebulla P, Introna M, Capelli C, Benigni A, Remuzzi G, Lazzari L. Life-sparing effect of human cord blood-mesenchymal stem cells in experimental acute kidney injury. Stem Cells. 2010;28(3):513–22. doi: 10.1002/stem.293. Mar 31. [DOI] [PubMed] [Google Scholar]
  • 182.Haller MJ, Viener HL, Wasserfall C, Brusko T, Atkinson MA, Schatz DA. Autologous umbilical cord blood transfusion for type 1 diabetes. Experimental Hematology. 2008;36:710–715. doi: 10.1016/j.exphem.2008.01.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 183.Willing AE, Lixian J, Milliken M, Poulos S, Zigova T, Song S, Hart C, Sanchez-Ramos J, Sanberg PR. Intravenous versus intrastriatal cord blood administration in a rodent model of stroke. Journal of Neuroscience Research. 2003;73:296–307. doi: 10.1002/jnr.10659. [DOI] [PubMed] [Google Scholar]
  • 184.Chang JW, Hung SP, Wu HH, Wu WM, Yang AH, Tsai HL, Yang LY, Lee OK. Therapeutic Effects of Umbilical Cord Blood-Derived Mesenchymal Stem Cell Transplantation in Experimental Lupus Nephritis. Cell Transplant. 2010 doi: 10.3727/096368910X520056. [Epub ahead of print] [DOI] [PubMed] [Google Scholar]
  • 185.Sun L, Wang D, Liang J, Zhang H, Feng X, Wang H, Hua B, Liu B, Ye S, Hu X, Xu W, Zeng X, Hou Y, Gilkeson GS, Silver RM, Lu L, Shi S. Umbilical cord mesenchymal stem cell transplantation in severe and refractory systemic lupus erythema-tosus. Arthritis & Rheumatism Arthritis Rheum. 2010;62:2467–75. doi: 10.1002/art.27548. [DOI] [PubMed] [Google Scholar]
  • 186.Cao FJ, Feng SQ. Human umbilical cord mesenchymal stem cells and the treatment of spinal cord injury. Chin Med J (Engl) 2009;122(2):225–31. Jan 20. [PubMed] [Google Scholar]

Articles from International Journal of Clinical and Experimental Medicine are provided here courtesy of e-Century Publishing Corporation

RESOURCES