Reduced Intensity BCNU/O6-BG Selection of MGMT-P140K Transduced Hematopoietic Stem Cells Is Efficient at Reduced Toxicity and Leads to an Unrestricted Oligoclonal Differentiation of Modified Murine Repopulating Stem Cells.

Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 2109-2109
Author(s):  
C. Ball ◽  
M. Schmidt ◽  
I. Pilz ◽  
S. Fessler ◽  
C. von Kalle ◽  
...  

Abstract Successful selection of gene modified hematopoietic stem cells against the majority of non-modified cells may increase the efficiency and safety of clinical gene therapy. Especially if a reduction of the intensity of the myelotoxic pre-transplant conditioning treatment is sought, selection of the initially low percentage of retrovirally gene modified stem cells is required. In addition to the decreased drug related toxicity, such a procedure reduces the risk of genotoxicity caused by insertional mutagenesis simply by diminishing the likelihood of transplanting stem cells that carry unwanted insertion sides compared to strategies that attempt to increase efficiency by increasing vector dose and/or the number of engrafted gene-modified cells. To date, the mutant O6-methylguanine-DNA methyltransferase (MGMT) enzyme that confers resistance to nitrosoureas such as BCNU is the drug-resistance gene that allows most efficient selection at the stem cell level. In the murine model, MGMT selection by BCNU and O6-BG has mostly been performed at LD 50 dose levels of the selecting agents, that is associated with very considerable toxicity. We now established minimal dosage requirements of transplanted cells as well as of BCNU and O6-BG that allow an efficient selection of murine long-term hematopoiesis. Bone marrow cells from 5-FU treated C57 BL/6J mice were transduced with an MGMT/IRES/eGFP encoding retroviral vector and transplanted at two dilutions (1x105 and 4x105) into 84 lethally irradiated syngeneic recipient mice. Starting 4 weeks post-transplant, the mice were treated monthly with two reduced dosages of O6-BG and BCNU (either 10 mg/kg O6-BG and 2,5 mg/kg BCNU or 20 mg/kg O6-BG and 5 mg/kg BCNU) as compared to the commonly used regimens. Kinetics, differentiation and clonality of the transduced hematopoiesis were monitored by FACS and LAM-PCR analysis of serial peripheral blood samples over a total of 5 rounds of selection. 3 of 48 mice died during the selection procedure. Although in 97% of all mice GFP+ blood cells were detectable 4 weeks after transplantation, an efficient selection (>twofold increase in transduced cells) was only seen in mice with >3% GFP+ initial blood cells. The percentage of transduced hematopoiesis in 9 of 24 mice treated with the higher BCNU and O6-BG dosage increased at least twofold as compared to 5 of 24 treated with the lower dosage and 1 of 24 mice without selection. Using Poisson statistics, the frequency of selectable hematopoietic units was calculated to be as low as 1 per 5x105 cells transplanted. Nevertheless, using LAM-PCR we detected stable oligoclonal hematopoiesis after transplantation of 4x105 cells for more than 6 months, suggesting that a subfraction of repopulating cell clones present were not selectable, or that the contribution of individual clones was small. No differences in lineage differentiation of the GFP+ hematopoiesis as determined in lineage marker expression by FACS could be observed after selection as compared to unselected control mice. In summary, our results demonstrate that a reduced dosage of BCNU and O6-BG with lower toxicity allows selection of MGMT expressing murine hematopoietic stem cells with unperturbed multilineage differentiation potential. Moreover, under these conditions, a subfraction of transduced repopulating cell clones was selectable.

Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 1412-1412
Author(s):  
Yuka Nakamura ◽  
Fumio Arai ◽  
Yumiko Gomei ◽  
Kentarou Hosokawa ◽  
Hiroki Yoshihara ◽  
...  

Abstract Currently, the niche for long-term hematopoietic stem cells (HSCs) is thought to consist conceptually of two parts: the endosteal surface (the osteoblastic niche) and a sinusoidal endothelium (the vascular niche), and a subset of osteoblasts functions as a key component of the hematopoietic stem cell niche. However, it is still unclear that the precise cellular and molecular contribution of osteoblastic cells on the HSC supportive microenvironment. In this study, we try to characterize the osteoblastic cells and investigate the property of osteoblastic niche cells. For isolation of osteoblastic cells, we treated the bone fragments of femur and tibiae with collagenase following flush-out of the bone marrow (BM). Non-hematopoietic and non-endothelial cells were then enriched by magnetic cell sorting of the CD45-CD31-Ter119- population, and expression of Sca-1 and platelet derived growth factor receptor α (PDGFRα) was analyzed. FACS analysis showed that CD45-CD31-TER119- cells were subdivided into three fractions: Sca-1+PDGFRα+, Sca-1-PDGFRα-, Sca-1-PDGFRα+. First we examined the multilineage differentiation potential of three populations. Although Sca-1- fractions efficiently differentiated into the osteoblastic lineage and showed calcium deposition, these cells hardly differentiated into adipocytes. In contrast to the Sca-1- cells, we found that Sca-1+PDGFRα+ cells can differentiate into osteoblastic and adipocytic lineages, suggesting that Sca-1+ cells have multi-potency. Next we examined the expression of osteoblastic marker expression by quantitative RT-PCR analysis, and found that Sca-1- populations expressed Runx2 and OB-cadherin. Alkaline phosphatase (ALP) staining of freshly isolated cells showed that Sca-1- fractions expressed ALP, while Sca-1+ cells did not express ALP. These data suggest that Sca-1- populations were the cell fractions, which were already committed to osteoblastic lineage. In addition, osteocalcin was expressed in PDGFRα+ fraction in Sca-1- cells, indicating that Sca-1-PDGFRα+ cells are more mature osteoblastic cells than Sca-1-PDGFRα-cells. Furthermore, N-cadherin was specifically expressed in Sca-1-PDGFRα+ cells, suggesting that N-cadherin was up-regulated with the maturation of osteoblastic cells. In addition, N-cadherin expression was up-regulated in Sca-1-PDGFRα+ cells with the postnatal development of BM. Interestingly, in the freshly isolated cells, we found that Sca-1+PDGFRα+ cells showed higher expression of Angiopoietin-1 (Ang-1), compared to Sca-1- fractions. Ang-1 expression was up-regulated in Sca-1-PDGFRα+ cells after over night incubation. Next we investigated the ability of these fractionated cells to support hematopoiesis. We examined the capacity of these fractionated cells on maintenance of colony formation ability of BM linage-Sca-1+c-Kit+ cells after 5 days of co-culture. Although CFU-C formation was supported Sca-1+PDGFRα+ cells, Sca-1-PDGFRα+ cells maintained CFU-Mix formation compared to the Sca-1+PDGFRα+ and Sca-1-PDGFRα-cells. From these data above, we hypothesize that multiple osteoblastic populations form a “niche complex” and collaborate with other supporting cells, such as CXCL12-abundant reticular (CAR) cells, to support HSCs, and that N-cadherin+ osteoblastic cells provide a foothold for anchoring of quiescent HSCs. Now we are investigating the gene expression profiles of these three populations and are tying to clarify the changes of characteristics of osteoblastic cells during postnatal BM development.


Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 1719-1719
Author(s):  
Edward F. Srour ◽  
Tamara L. Horvath

Abstract Murine bone marrow-derived cells expressing Sca-1+c-kit+lin− (KSL), as well as subfractions of these cells, represent an enriched population of hematopoietic stem cells (HSC) capable of long-term reconstitution of lethally irradiated recipients. Commitment to the hematopoietic lineage is invariably associated with expression of the pan-leukocyte marker CD45 which is also expressed on KSL cells. Whether KSL cells are the most primitive population of HSC present in the bone marrow (BM) is not fully resolved. We hypothesized that putative HSC that are more primitive than KSL cells may not express CD45 or genetic elements that mark early hematopoietic specification and commitment, but may mature under appropriate conditions into CD45+ cells capable of hematopoietic differentiation in conditioned hosts. BM cells from 8 to 10-week old BoyJ mice were collected by flushing and erythrocytes were lysed. The remaining cells were stained and sorted to yield CD45+ Sca-1+ c-kit+ (CD45+HSC) and CD45− Sca-1+ c-kit− (CD45−) cells which represented approximately 0.02% of total cells analyzed. PCR analysis of both cell populations revealed that CD45+HSC expressed CD45 and SCL but not PU.1 while CD45− cells did not express any of these genes. Directly after sorting, CD45+HSC, but not CD45− cells contained clonogenic cells that gave rise to hematopoietic colonies in progenitor cell assays. Similarly, while fresh CD45+HSC were able to respond to exogenous hematopoietic cytokines including SCF, TPO, and FL in liquid suspension cultures as evidenced by expansion and differentiation, their CD45− counterparts failed to proliferate under these conditions and none survived beyond 7 days of culture. When transplanted competitively into lethally irradiated congenic recipients, only freshly isolated CD45+HSC sustained donor-derived hematopoiesis, whereas hematopoiesis in mice injected with freshly isolated CD45− cells was sustained long term by competitor cells and endogenous host-derived stem cells. Both groups of CD45+HSC and CD45− cells could be expanded on irradiated M210B4 stromal cells when supplemented with SCF, TPO, and FL, with CD45− cells giving rise to cobblestone foci of small, round translucent cells beginning on day 7 of culture. Cultured CD45+HSC continued to express CD45 and SCL and, depending on the length of culture, also expressed PU.1. Interestingly, after 15 days in culture, CD45− cells expressed CD45 by RT-PCR and FACS (in addition to Sca-1) and also expressed mRNA for SCL. Given the ability of CD45− cells to expand under these conditions and to acquire CD45 expression, we next compared the repopulating potential of fresh and cultured CD45+HSC and CD45− cells using lethally irradiated C57Bl/6 recipients. As expected, fresh CD45+HSC sustained donor-derived engraftment and culture of these cells over M210B4 for 15 days reduced their repopulating potential more than 7-fold. In contrast, CD45− cells maintained on M210B4 (the expansion equivalent of 750 cells seeded) contributed to hematopoietic engraftment, albeit at low levels (under 5% chimerism). These data demonstrate that CD45− Sca-1+ c-kit− cells may be marrow resident precursors of hematopoietic stem cells and suggest that early stages of the HSC hierarchy may include CD45− cells. Whether these CD45− cells also posses endothelial differentiation potential and can give rise to CD45+HSC in vivo is now under investigation.


Blood ◽  
1992 ◽  
Vol 80 (1) ◽  
pp. 21-24 ◽  
Author(s):  
OM Smith ◽  
SA Dolan ◽  
JA Dvorak ◽  
TE Wellems ◽  
F Sieber

The purpose of this study was to evaluate the photosensitizing dye merocyanine 540 (MC540) as a means for extracorporeal purging of Plasmodium falciparum-infected erythrocytes from human blood. Parasitized red blood cells bound more dye than nonparasitized cells, and exposure to MC540 and light under conditions that are relatively well tolerated by normal erythrocytes and normal pluripotent hematopoietic stem cells reduced the concentration of parasitized cells by as much as 1,000-fold. Cells parasitized by the chloroquine- sensitive HB3 clone and the chloroquine-resistant Dd2 clone of P falciparum were equally susceptible to MC540-sensitized photolysis. These data suggest the potential usefulness of MC540 in the purging of P falciparum-infected blood.


Blood ◽  
1994 ◽  
Vol 83 (12) ◽  
pp. 3758-3779 ◽  
Author(s):  
N Uchida ◽  
HL Aguila ◽  
WH Fleming ◽  
L Jerabek ◽  
IL Weissman

Abstract Hematopoietic stem cells (HSCs) are believed to play a critical role in the sustained repopulation of all blood cells after bone marrow transplantation (BMT). However, understanding the role of HSCs versus other hematopoietic cells in the quantitative reconstitution of various blood cell types has awaited methods to isolate HSCs. A candidate population of mouse HSCs, Thy-1.1lo Lin-Sca-1+ cells, was isolated several years ago and, recently, this population has been shown to be the only population of BM cells that contains HSCs in C57BL/Ka-Thy-1.1 mice. As few as 100 of these cells can radioprotect 95% to 100% of irradiated mice, resulting long-term multilineage reconstitution. In this study, we examined the reconstitution potential of irradiated mice transplanted with purified Thy-1.1lo Lin-Sca-1+ BM cells. Donor-derived peripheral blood (PB) white blood cells were detected as early as day 9 or 10 when 100 to 1,000 Thy-1.1lo Lin-Sca-1+ cells were used, with minor dose-dependent differences. The reappearance of platelets by day 14 and thereafter was also seen at all HSC doses (100 to 1,000 cells), with a slight dose-dependence. All studied HSC doses also allowed RBC levels to recover, although at the 100 cell dose a delay in hematocrit recovery was observed at day 14. When irradiated mice were transplanted with 500 Thy-1.1lo Lin-Sca-1+ cells compared with 1 x 10(6) BM cells (the equivalent amount of cells that contain 500 Thy-1.1lo Lin-Sca-1+ cells as well as progenitor and mature cells), very little difference in the kinetics of recovery of PB, white blood cells, platelets, and hematocrit was observed. Surprisingly, even when 200 Thy1.1lo Lin-Sca- 1+ cells were mixed with 4 x 10(5) Sca-1- BM cells in a competitive repopulation assay, most of the early (days 11 and 14) PB myeloid cells were derived from the HSC genotype, indicating the superiority of the Thy-1.1lo Lin-Sca-1+ cells over Sca-1- cells even in the early phases of myeloid reconstitution. Within the Thy-1.1lo Lin-Sca-1+ population, the Rhodamine 123 (Rh123)hi subset dominates in PB myeloid reconstitution at 10 to 14 days, only to be overtaken by the Rh123lo subset at 3 weeks and thereafter. These findings indicate that HSCs can account for the early phase of hematopoietic recovery, as well as sustained hematopoiesis, and raise questions about the role of non-HSC BM populations in the setting of BMT.


Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 2563-2563
Author(s):  
Fernando Fierro ◽  
Thomas Illmer ◽  
Duhoui Jing ◽  
Philip Le Coutre ◽  
Gerhard Ehninger ◽  
...  

Abstract Recent data show that the tyrosine kinase inhibitor Imatinib mesylate (IM) also affects normal hematopoietic stem cells (HSC), T lymphocyte activation and dendritic cell function not relying on the specific inhibition of bcr-abl activity. Mesenchymal stem cells (MSC) have been identified in the bone marrow (BM) as multipotent non-hematopoietic progenitor cells that differentiate into osteoblasts, adipocytes, chondrocytes, tenocytes, skeletal myocytes, and cells of visceral mesoderm. MSC interact with HSC, influencing their homing and differentiation through cell-cell contact and the production of factors including chemokines We evaluated possible effects of IM in vitro on human bone marrow-derived MSC. Screening the activity of fourty-two receptor tyrosine kinases by a phospho-receptor tyrosine kinase (RTK)-array revealed an exclusive inhibition of platelet-derived growth factor receptor (PDGFRβ) by IM which consequently affects downstream targets of PDGFRβ as Akt and Erk1/2 signalling pathways in a concentration and time dependent manner. Furthermore, perinuclear multivesicular bodies harbouring PDGFRβ were found within 18–20 hours culture of MSC in the presence of 5 μM IM. Cell proliferation and clonogenicity (evaluated as the capability to form colony forming units - fibroblasts (CFU-F)) of MSC were significantly inhibited by IM in a concentration dependent fashion. IM inhibits significantly the differentiation process of MSC into osteoblasts as evaluated by decreased alkaline phosphatase activity and reduced calcium phosphate precipitates. In contrary, differentiation of MSC into adipocytes was strongly favoured in presence of IM. All these functional deficits described, probably contribute to an observed 50% reduction in the support of clonogenic hematopoietic stem cells, as evaluated by a long term culture-initiating cells (LTC-IC)-based assay. In summary our experiments show that IM inhibits the capacity of human MSC to proliferate and to differentiate into the osteogenic lineage, favouring adipogenesis. This effect is mainly mediated by an inhibition of PDGFRβ autophosphorylation leading to a more pronounced inhibition of PI3K/Akt compared to Erk1/2 signalling. This work confirms the role of PDGFRβ recently described for the proliferation and differentiation potential of MSC and provides a first possible explanation for the altered bone metabolism found in certain patients treated with IM.


Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 802-802 ◽  
Author(s):  
Sten Eirik W. Jacobsen ◽  
Robert Mansson ◽  
Anne Hultquist ◽  
Mikael Sigvardsson ◽  
Natalija Buza-Vidas ◽  
...  

Abstract We recently identified a novel Lin−Sca-1+c-kithiCD34+Flt3hi (LSKCD34+Flt3hi) lymphoid-primed multipotent progenitor (LMPP) in adult mouse bone marrow which, although possessing a combined lymphoid (B and T cell) and myeloid (granulocyte-monocyte; GM) differentiation potential, have little or no ability to adopt erythroid (E) and megakaryocyte (MK) lineage fates (Adolfsson et al, Cell121:295, 2005). The identification of this lineage restricted lymphomyeloid progenitor implicates the existence of alternative roadmaps for lineage commitment of pluripotent hematopoietic stem cells (HSCs), distinct from the classical model suggesting that the first HSC commitment step results in a strict separation into common lymphoid and myeloid progenitors. Herein we provide further, genetic evidence for such a model. Affymetrix global gene profiling, quantitative PCR, and multiplex single cell PCR analysis of LSKCD34−Flt3− long-term (LT)-HSCs, LSKCD34+Flt3− short-term (ST)-HSCs and LSKCD34+Flt3hi LMPPs, demonstrate that LMPPs in contrast to LT-HSCs and ST-HSCs down-regulate or turn off a number of genes critically involved in MkE lineage development, including GATA-1 and the receptors for erythropoietin and thrombopoietin. In contrast, a number of genes specific for early lymphoid development, including Rag-1, sterile Ig and IL-7 receptor are upregulated in LMPPs but absent in LT-HSCs and ST-HSCs. Importantly, within the LMPP, these lymphoid genes are typically co-expressed with a number of GM associated genes such as G-CSF receptor and MPO, but virtually never co-expressed with MkE associated genes. Investigating fetal liver day 14.5 we also provide evidence for existence of the LSKCD34+Flt3hi LMPPs at this early stage of development, and using a single cell clonal assay promoting combined B, T and myeloid lineage development, we demonstrate that a large fraction of fetal LMPPs lacking MkE potential possess a combined GM, B and T cell potential. Thus, evaluation at the single cell level of combined lineage potentials and multilineage gene expression provide compelling evidence for lymphoid-priming within the HSC compartment being preceeded by a loss of MkE potential, but occurring prior to loss of GM potential.


Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 89-89 ◽  
Author(s):  
Laura M. Calvi ◽  
Benjamin J. Frisch ◽  
Benjamin J. Gigliotti ◽  
Christina A. Christianson ◽  
Jonathan M. Weber ◽  
...  

Abstract Parathyroid Hormone (PTH) targets osteoblastic cells (OBs) in the bone marrow microenvironment and expands hematopoietic stem cells (HSC) through Notch activation. Since PTH stimulates the Notch ligand Jagged1 (J1) in OBs, we have focused on the signaling pathways involved in this PTH effect in order to identify novel activators of the HSC niche. Osteoblastic Protein Kinase A (PKA) activation is required for the PTH-dependent J1 increase in OBs. Therefore, we hypothesized that alternative PKA activators could also regulate osteoblastic J1, alter the HSC niche, and provide additional pharmacologic tools to expand HSC in vivo. Consistent with this hypothesis, direct PKA agonists 8-bromo-cAMP and dibutyryl-cAMP stimulated J1 in osteoblastic UMR106 cells. In addition, PGE2, a member of the prostaglandin family known to stimulate PKA in OBs, was studied in vivo and in vitro. By real-time RT-PCR analysis, J1 mRNA was increased up to 5 fold at 2 hours in UMR106 cells when treated with PGE2 (10−7 M) compared to vehicle. J1 protein was also increased after treatment with PGE2. The PGE2-dependent J1 increase was blocked in the presence of the specific PKA inhibitors H89 and myristoylated PKA Inhibitory Peptide (14–22)(PKI) (200ug/ml), demonstrating that PKA is necessary for osteoblastic J1 stimulation by PGE2. Since systemic PGE2 is known to have bone anabolic effects in both humans and animal models, adult wild-type FVB/N male mice were treated with PGE2 (6mg/kg/day i.p.) for 12 days. This regimen has previously been shown to have bone anabolic effects in rats. At day 12, histologic analysis demonstrated an anabolic effect mainly on cortical bone, as was evident in the femurs and tibiae of PGE2-treated mice compared to control. This histologic finding was confirmed by histomorphometry (trabecular bone area means 41% vs 12%,p=0.0916, n=3 in both groups; cortical thickness means 138 vs 85 μm, p=0.0071, n=3 in both groups). Frequency of hematopoietic stem cells (c-Kit+, Sca1+, lin−) was increased in bone marrow from PGE2-treated vs control mice by over 20% (p=0.0018, n=8 in both groups). In summary, PGE2 stimulates J1 in osteoblastic cells through PKA activation and increases mainly cortical bone in vivo. Ongoing studies will confirm whether in vivo PGE2 treatment expands HSC, and whether osteoblastic J1 regulates this process. This study identifies PGE2 as a novel regulator of osteoblastic J1, and as a potential new microenvironmental modulator of HSC, which could be used for in vivo therapeutic HSC niche manipulation.


Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 3650-3650
Author(s):  
Kent W. Christopherson ◽  
Tiki Bakhshi ◽  
Shamanique Bodie ◽  
Shannon Kidd ◽  
Ryan Zabriskie ◽  
...  

Abstract Hematopoietic Stem Cells (HSC) are routinely obtained from bone marrow, mobilized peripheral blood, and umbilical Cord Blood. Traditionally, adult bone marrow has been utilized as a source of Mesenchymal Stem Cells (MSC). Bone marrow derived MSC (BM-MSC) have previously been shown to maintain the growth of HSC obtained from cord blood and have been utilized for cord blood expansion purposes. However, the use of a mismatched BM-MSC feeder stromal layer to support the long term culture of cord blood HSC is not ideal for transplant purposes. The isolation of MSC from a novel source, the Wharton’s Jelly of Umbilical Cord segments, was recently reported (Romanov Y, et al. Stem Cells.2003; 21: 105–110) (Lee O, et al. Blood.2004; 103: 1669–1675). We therefore hypothesized that Umbilical Cord derived MSC (UC-MSC) have the ability to support the long term growth of cord blood derived HSC similar to that previously reported for BM-MSC. To test this hypothesis, MSC were isolated from the Wharton’s Jelly of Umbilical Cord segments and defined morphologically and by cell surface markers. UC-MSC were then tested for their ability to support the growth of pooled CD34+ cord blood cells in long term culture - initiating cell (LTC-IC) assays as compared to BM-MSC. We observed that like BM-MSC, CB-MSC express a defined set of cell surface markers. By flow cytometry we determined that that both UC-MSC and BM-MSC are positive for CD29, CD44, CD73, CD90, CD105, CD166, HLA-A and negative for CD45, CD34, CD38, CD117, HLA-DR expression. Utilizing Mitomycin C treated (200 μM, 15 min.) UC-MSC from multiple donors as a feeder layer we observed that UC-MSC have the ability to support the maintenance of long term hematopoiesis during the LTC-IC assay. Specifically, UC-MSC isolated from separate umbilical cord donors support the growth of 69.6±11.9 (1A), 31.7±3.9 (2B), 67.0±13.5 (3A), and 38.5±13.7 (3B) colony forming cells (CFC) per 1×104 CD34+ cord blood cells as compared to 64.0±4.2 CFC per 1×104 CD34+ cord blood cells supported by BM-MSC (Mean±SEM, N=4 separate segments from three different donors). Thus, Umbilical Cord derived Mesenchymal Stem Cells, a recently described novel source of MSC, have the ability to support long term maintenance of Hematopoietic Stem Cells, as defined by the LTC-IC assay. These results may have potential therapeutic application with respect to ex vivo stem cell expansion of Cord Blood Hematopoietic Stem Cells utilizing a Mesenchymal Stem Cell stromal layer. In addition, these data suggest the possibility of co-transplantation of matched Mesenchymal and Hematopoietic Stem Cells from the same umbilical cord and cord blood donor respectively. Lastly, these results describe a novel model system for the future study of the interaction between Cord Blood Hematopoietic Stem Cells and the appropriate supportive microenvironment represented by the Umbilical Cord - Mesenchymal Stem Cells.


Sign in / Sign up

Export Citation Format

Share Document