Heart infarct in NOD‐SCID mice: Therapeutic vasculogenesis by transplantation of human CD34 + cells and low dose CD34 + KDR + cells

2004 ◽  
Vol 18 (12) ◽  
pp. 1392-1394 ◽  
Author(s):  
Rosanna Botta ◽  
Erhe Gao ◽  
Giorgio Stassi ◽  
Desirée Bonci ◽  
Elvira Pelosi ◽  
...  
Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 1259-1259
Author(s):  
Abraham Avigdor ◽  
Yaron Vagima ◽  
Polina Goichberg ◽  
Shoham Shivtiel ◽  
Melania Tesio ◽  
...  

Abstract Hematopoietic progenitor cell release to the circulation is the outcome of signals provided by cytokines, chemokines, adhesion molecules, and proteolytic enzymes. Clinical recruitment of immature CD34+ cells to the peripheral blood (PB) is achieved by repeated G-CSF stimulations. Yet, the mechanisms governing progenitor cell egress during steady state homeostasis and clinical mobilization are not fully understood. Membrane type-1 metalloproteinase (MT1-MMP) and its endogenous inhibitor, RECK, are established key regulators of tumor and endothelial cell motility. We detected higher MT1-MMP and lower RECK expression on circulating human CD34+ progenitors and maturing leukocytes as compared to immature bone-marrow (BM) cells. MT1-MMP expression was even more prominent on CD34+ cells obtained from PB of G-CSF-treated healthy donors whereas RECK labeling was barely detected. In addition, five daily injections of G-CSF to NOD/SCID mice, previously engrafted with human cells, increased MT1-MMP and decreased RECK expression on human CD45+ leukocytes, immature CD34+ and primitive CD34+/CD38−/low cells, in a PI3K/Akt1-dependent manner, resulting in elevated MT1-MMP activity. Inverse regulation of MT1-MMP and RECK by G-CSF mobilization was confirmed by in situ immuno-labeling of BM sections, as well as by human MT1-MMP and RECK mRNA expression analysis of leukocytes repopulating the BM of chimeric mice. Blocking MT1-MMP function impaired mobilization, while RECK neutralization promoted egress of human CD34+ progenitors in the functional pre-clinical model of NOD/SCID chimeric mice. Targeting MT1-MMP expression by SiRNA or blocking its function reduced the in-vitro chemotactic response to SDF-1 of human CD34+ progenitors via matrigel and impaired to a similar extent the BM homing capacity of transplanted human CD34+ cells in NOD/SCID mice. In accordance, neutralization of RECK function, thus abrogating RECK-mediated inhibition of MT1-MMP, facilitated SDF-1-induced migration of steady state human BM CD34+ cells in vitro. Furthermore, following G-CSF mobilization, we also observed a reduction in CD44 expression on human leukocytes and, specifically, on immature CD34+ progenitor cells in the BM of chimeric mice. This was accompanied by accumulation of CD44 cleaved products of molecular weights, expected for MT1-MMP activity, in the BM supernatants. In chimeric mice co-injected with MT1-MMP-neutralizing Ab, less cleavage of CD44 was detected upon G-CSF mobilization, whereas in the absence of a mobilizing signal, increasing MT1-MMP activity by anti RECK Ab injection facilitated CD44 proteolysis on the BM cells. Finally, MT1-MMP expression correlated with the number of CD34+ cells, collected on the first apheresis day in 29 consecutive patients with lymphoid malignancies and in 21 healthy donors treated with G-CSF. In conclusion, our results indicate that G-CSF inversely regulates MT1-MMP and RECK expression on CD34+ progenitors, resulting in net increase in MT1-MMP activity. MT1-MMP proteolysis of CD44 diminishes progenitor adhesion to BM components, leading to cell egress. These cell autonomous changes provide a previously undefined mechanism for G-CSF recruitment of CD34+ progenitors and might serve as target for new approaches to improve clinical stem cell mobilization.


1999 ◽  
Vol 189 (10) ◽  
pp. 1601-1610 ◽  
Author(s):  
Catherine Robin ◽  
Françoise Pflumio ◽  
William Vainchenker ◽  
Laure Coulombel

Transplantation of genetically marked donor cells in mice have unambiguously identified individual clones with full differentiative potential in all lymphoid and myeloid pathways. Such evidence has been lacking in humans because of limitations inherent to clonal stem cell assays. In this work, we used single cell cultures to show that human cord blood (CB) contains totipotent CD34+ cells capable of T, B, natural killer, and granulocytic cell differentiation. Single CD34+ CD19−Thy1+ (or CD38−) cells from fresh CB were first induced to proliferate and their progeny separately studied in mouse fetal thymic organotypic cultures (FTOCs) and cocultures on murine stromal feeder layers. 10% of the clones individually analyzed produced CD19+, CD56+, and CD15+ cells in stromal cocultures and CD4+CD8+ T cells in FTOCs, identifying totipotent progenitor cells. Furthermore, we showed that totipotent clones with similar lymphomyeloid potential are detected in the bone marrow of nonobese diabetic severe combined immunodeficient (NOD-SCID) mice transplanted 4 mo earlier with human CB CD34+ cells. These results provide the first direct demonstration that human CB contains totipotent lymphomyeloid progenitors and transplantable CD34+ cells with the ability to reconstitute, in the marrow of recipient mice, the hierarchy of hematopoietic compartments, including a compartment of functional totipotent cells. These experimental approaches can now be exploited to analyze mechanisms controlling the decisions of such primitive human progenitors and to design conditions for their ampification that can be helpful for therapeutic purposes.


Science ◽  
1999 ◽  
Vol 283 (5402) ◽  
pp. 682-686 ◽  
Author(s):  
Hiroyuki Miyoshi ◽  
Kent A. Smith ◽  
Donald E. Mosier ◽  
Inder M. Verma ◽  
Bruce E. Torbett
Keyword(s):  

Blood ◽  
2000 ◽  
Vol 95 (11) ◽  
pp. 3289-3296 ◽  
Author(s):  
Amnon Peled ◽  
Orit Kollet ◽  
Tanya Ponomaryov ◽  
Isabelle Petit ◽  
Suzanna Franitza ◽  
...  

Abstract Hematopoietic stem cell homing and engraftment require several adhesion interactions, which are not fully understood. Engraftment of nonobese/severe combined immunodeficiency (NOD/SCID) mice by human stem cells is dependent on the major integrins very late activation antigen–4 (VLA-4); VLA-5; and to a lesser degree, lymphocyte function associated antigen–1 (LFA-1). Treatment of human CD34+cells with antibodies to either VLA-4 or VLA-5 prevented engraftment, and treatment with anti–LFA-1 antibodies significantly reduced the levels of engraftment. Activation of CD34+ cells, which bear the chemokine receptor CXCR4, with stromal derived factor 1 (SDF-1) led to firm adhesion and transendothelial migration, which was dependent on LFA-1/ICAM-1 (intracellular adhesion molecule–1) and VLA-4/VCAM-1 (vascular adhesion molecule–1). Furthermore, SDF-1–induced polarization and extravasation of CD34+/CXCR4+ cells through the extracellular matrix underlining the endothelium was dependent on both VLA-4 and VLA-5. Our results demonstrate that repopulating human stem cells functionally express LFA-1, VLA-4, and VLA-5. Furthermore, this study implies a novel approach to further advance clinical transplantation.


Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 3214-3214 ◽  
Author(s):  
David A. Hess ◽  
Phillip E. Herrbrich ◽  
Louisa Wirthlin ◽  
Timothy P. Craft ◽  
Jan A. Nolta

Abstract The successful development of stem cell-based therapies requires a thorough understanding of human hematopoietic stem cell (HSC) populations. Human CD34− cells engraft NOD/SCID mice with low efficiency by intravenous (IV) transplant. However, intra-femoral injection into immune deficient mice has identified potent human repopulating cells from CD34+ and CD34− subfractions. We recently described a novel strategy to purify reconstituting HSC from human umbilical cord blood (UCB) by lineage depletion (Lin−) followed by selection of cells with high aldehyde dehydrogenase (ALDH) activity. Hematopoietic progenitor function and in vivo reconstituting ability were exclusively maintained within the ALDHhiLin− population, which demonstrated variable expression of CD34. Here, we compared the repopulating ability of purified CD34+ALDHhiLin− and CD34−ALDHhiLin− populations to traditionally isolated CD34+Lin− and CD34−Lin− cells. Sorting of Lin− cells from human UCB isolated CD34−ALDHhi and CD34+ALDHhi cells (>96% purity) at an overall frequency of 4.4±1.3% or 29.1±3.5%, respectively. In contrast to CD34−Lin− cells, ALDHhiCD34−Lin− cells demonstrated robust clonogenic progenitor function in vitro (1 CFU in 9 cells, n=3), and total colony production was further increased in ALDHhiCD34+Lin− cells (1 CFU in 4.5 cells, n=4) (p<0.05). Human hematopoietic repopulation was consistently observed in the bone marrow (17.2±4.2%), spleen (0.8±0.2%), and peripheral blood (0.7±0.3%) of NOD/SCID β2M null mice 6–8 weeks after IV transplant with 103–104 purified ALDHhiCD34+Lin− cells (n=14). Similarly, intra-femoral injection (IF) of ALDHhiCD34+Lin− cells resulted in robust human repopulation (n=5). IV injection of equivalent doses of either ALDHhiCD34+Lin− or CD34+Lin− cells showed similar levels and frequencies of human hematopoietic engraftment. Repopulating ALDHhiCD34+Lin− cells also differentiated into cells expressing markers for mature myeloid (CD33, CD14), B-lymphoid (CD19, CD20) cells and primitive repopulating cells (CD34+CD38−) at similar frequencies as CD34+Lin− cells (n=5). IV injection of 2x104–1x105 ALDHhiCD34−Lin− cells engrafted at 0.2–0.3% in the BM of 3 of 4 NOD/SCID β2M null mice, whereas IV injection of up to 4x105 CD34−Lin− cells produced no detectable human engraftment (n=6). IF-injected ALDHhiCD34−Lin− cells engrafted the injected bone in 2 of 3 NOD/SCID mice at low levels and did not efficiently migrate to the non-injected femur or tibiae. In summary, the human UCB ALDHhiLin− population includes both CD34+ and CD34− cells capable of bone marrow homing and hematopoietic reconstitution. Therefore, isolation of CD34− cells based on high ALDH activity may reveal a novel population of hematopoietic stem and progenitor cells.


Blood ◽  
1999 ◽  
Vol 93 (11) ◽  
pp. 3736-3749 ◽  
Author(s):  
Wanda Piacibello ◽  
Fiorella Sanavio ◽  
Antonella Severino ◽  
Alessandra Danè ◽  
Loretta Gammaitoni ◽  
...  

Understanding the repopulating characteristics of human hematopoietic stem/progenitor cells is crucial for predicting their performance after transplant into patients receiving high-dose radiochemotherapy. We have previously reported that CD34+cord blood (CB) cells can be expanded in vitro for several months in serum containing culture conditions. The use of combinations of recombinant early acting growth factors and the absence of stroma was essential in determining this phenomenon. However, the effect of these manipulations on in vivo repopulating hematopoietic cells is not known. Recently, a new approach has been developed to establish an in vivo model for human primitive hematopoietic precursors by transplanting human hematopoietic cells into sublethally irradiated nonobese diabetic severe combined immunodeficient (NOD/SCID) mice. We have examined here the expansion of cells, CD34+ and CD34+38− subpopulations, colony-forming cells (CFC), long-term culture initiating cells (LTC-IC) and the maintenance or the expansion of SCID-repopulating cells (SRC) during stroma-free suspension cultures of human CD34+ CB cells for up to 12 weeks. Groups of sublethally irradiated NOD/SCID mice were injected with either 35,000, 20,000, and 10,000 unmanipulated CD34+ CB cells, which were cryopreserved at the start of cultures, or the cryopreserved cells expanded from 35,000, 20,000, or 10,000 CD34+ cells for 4, 8, and 12 weeks in the presence of a combination of early acting recombinant growth factors (flt 3/flk2 ligand [FL] + megakaryocyte growth and development factor [MGDF] ± stem cell factor [SCF] ± interleukin-6 [IL-6]). Mice that had been injected with ≥20,000 fresh or cryopreserved uncultured CD34+ cells did not show any sign or showed little engraftment in a limited number of animals. Conversely, cells that had been generated by the same number of initial CD34+ CB cells in 4 to 10 weeks of expansion cultures engrafted the vast majority of NOD/SCID mice. The level of engraftment, well above that usually observed when the same numbers of uncultured cells were injected in the same recipients (even in the presence of irradiated CD34− cells) suggested that primitive hematopoietic cells were maintained for up to 10 weeks of cultures. In addition, dilution experiments suggest that SRC are expanded more than 70-fold after 9 to 10 weeks of expansion. These results support and extend our previous findings that CD34+ CB stem cells (identified as LTC-IC) could indeed be grown and expanded in vitro for an extremely long period of time. Such information may be essential to design efficient stem cell expansion procedures for clinical use.


Blood ◽  
1998 ◽  
Vol 91 (7) ◽  
pp. 2406-2414 ◽  
Author(s):  
J.C.Y. Wang ◽  
T. Lapidot ◽  
J.D. Cashman ◽  
M. Doedens ◽  
L. Addy ◽  
...  

Abstract We have previously shown that intravenously injected peripheral blood (PB) or bone marrow (BM) cells from newly diagnosed chronic myeloid leukemia (CML) patients can engraft the BM of sublethally irradiated severe combined immunodeficient (SCID) mice. We now report engraftment results for chronic phase CML cells in nonobese diabetic (NOD)/SCID recipients which show the superiority of this latter model. Transplantation of NOD/SCID mice with 7 to 10 × 107 patient PB or BM cells resulted in the continuing presence of human cells in the BM of the mice for up to 7 months, and primitive human CD34+ cells, including those detectable as colony-forming cells (CFC), as long-term culture-initiating cells, or by their coexpression of Thy-1, were found in a higher proportion of the NOD/SCID recipients analyzed, and at higher levels than were seen previously in SCID recipients. The human CFC and total human cells present in the BM of the NOD/SCID mice transplanted with CML cells also contained higher proportions of leukemic cells than were obtained in the SCID model, and NOD/SCID mice could be repopulated with transplants of enriched CD34+ cells from patients with CML. These results suggest that the NOD/SCID mouse may allow greater engraftment and amplification of both normal and leukemic (Ph+) cells sufficient for the quantitation and characterization of the normal and leukemic stem cells present in patients with CML. In addition, this model should make practical the investigation of mechanisms underlying progression of the disease and the development of more effective in vivo therapies.


Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 1672-1672
Author(s):  
Kiyoshi Ando ◽  
Yoshihiko Nakamura ◽  
Takashi Yahata ◽  
Yukari Muguruma ◽  
Tadayuki Sato ◽  
...  

Abstract CD34 negative hematopoietic stem cells (CD34− HSCs) were identified in mice and humans. Human HSCs are evaluated as severe combined immunodeficient mouse (SCID)-repopulating cells (SRCs), originally identified by the ability to reconstitute hematopoiesis in nonobese diabetic (NOD)/SCID mouse. CD34− cord blood (CB) cells have been hard to engraft in NOD/SCID mice until recent report of successlul engraftment by intra-bone marrow transplantation (iBMT). However, CD34− bone marrow (BM) cells have not been analyzed precisely. We prepared lineage negative CD34 negative (Lin-CD34−) cells by negative selection using CD2,3,7,14,16,19,20,33,34,36,41,56,127, and GlyA antibody. Lin-CD34− BM cells did not engraft in NOD/SCID mice even by using iBMT (0/6). In the previous study, we reported that Lin-CD34− BM cells were able to differentiate into CD34+ cells accompanied by the emergence of colony forming activity after 7 days of stroma-dependent culture, while SRC activity was not detected. (BMT 28, 587–595, 2001) Here we cultured Lin-CD34− BM cells on stroma cells transfected with human angiopoietin-1 cDNA (AHESS-5), since we detected Tie-2 expression on Lin-CD34− BM cells. AHESS-5 supported induction of CD34 much better than HESS-5 cells or empty vector transfected control cells (EVHESS-5), and the effect was blocked by anti-Tie-2 antibody (Fig.1). Furtheremore, CD34+ cells produced from CD34− BM cells engrafted in NOD/SCID mice (11/12). As previously reported, CD34− CB cells differentiate CD34+ cells and acquire SRC activity by stroma-dependent culture without angiopoietin-1. These results highlighted the characteristic differences of CD34− HSCs of BM from CB and the unique role of BM niche for CD34− HSCs. Fig. 1 CD34 expression on Lin − CD34 − BM cells after 7 days of culture Fig. 1. CD34 expression on Lin−CD34− BM cells after 7 days of culture


Sign in / Sign up

Export Citation Format

Share Document