Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2013 Aug 1.
Published in final edited form as: Ann N Y Acad Sci. 2012 Aug;1266(1):151–160. doi: 10.1111/j.1749-6632.2012.06509.x

Enhancing engraftment of cord blood cells via insight into biology of stem/progenitor cell function

Hal E Broxmeyer 1
PMCID: PMC3428743  NIHMSID: NIHMS361421  PMID: 22901266

Abstract

Cord blood (CB) transplantation has been used over the last 24 years to treat patients with malignant and non-malignant disorders. CB has its advantages and disadvantages compared to other sources of hematopoietic stem (HSCs) and progenitor (HPCs) cells for transplantation. More knowledge of the cytokines, and intracellular signaling molecules regulating HSCs and HPCs could be used to modulate these regulators for clinical benefit. This review provides brief information on the field of CB transplantation and studies from the author’s laboratory that focus on regulation of HSCs and HPCs by CD26/DPPIV, SDF-1/CXCL12, the Rheb2-mTOR pathway, SIRT1, DEK, cyclin dependent kinase inhibitors, and cytokines/growth factors. It also briefly discusses cryopreservation of CB HSCs and HPCs.

Keywords: Cord blood, stem and progenitor cells, intracellular signals, cytokines

Introduction

Cord blood (CB) has served as a transplantable source of hematopoietic stem and progenitor cells to treat malignant and non-malignant disorders since our initial laboratory,(19) and clinical studies(1014) over 20 years ago. There have now been over 25,000 CB transplants performed worldwide,(15) since our initial clinical report in which a child with Fanconi anemia was successfully treated with CB from his HLA-matched sister.(10) The field has moved rapidly, and advances in CB banking(1622) and transplantation(2344) have been encouraging, but there is still much to be learned to make CB transplantation a more efficient and efficacious procedure.(15,45)

There are advantages to using CB as a source of transplantable cells over that of other sources of hematopoietic stem (HSCs) and progenitor (HPCs) cells. This includes that CB is a readily available source of HLA-typed cells being stored in CB banks, they have already been used to treat essentially all malignant and non-malignant diseases that can be treated by bone marrow (BM) transplantation, and CB transplantation induces a lowered level of acute and chronic graft versus host disease (GVHD) when used as a single minimally manipulated unit. This latter characteristic of CB cells allows for more flexibility in related and unrelated donors using partially HLA-mismatched CB, than that used in BM transplantation. There are also disadvantages to using CB as a source of transplantable HSCs and HPCs. CB transplantation is associated with a slower time to neutrophil and platelet engraftment, and to immune cell recovery than that of BM and mobilized peripheral blood (mPB), events at least in part due to the more limiting numbers of HSCs and HPCs in CB collections compared to that in BM and mPB. For BM and mPB one can collect many cells, but with CB what is collected at the birth of a baby is all that is available, although, it is possible that means to collect greater numbers of CB cells at the child’s birth may be feasible in the future.(46) However, at present, the lesser number of HSCs and HPCs collected in single CB units is somewhat problematic for single CB unit transplantation for adults and higher weight pediatric recipients, and CB transplantation has been associated with enhanced graft failure.

A number of investigators, including our own group, have worked on means to enhance the engraftment of limiting numbers of HSCs and HPCs, in a basic science laboratory setting, in pre-clinical animal studies, and in pilot, phase I, and other clinical studies. Examples of such attempts to foster a more potent CB transplant have been recently reviewed by experts in the field in very recent updates and include: double CB transplantation,(4043, 4750) ex vivo expansion of HSCs and HPCs,(5154) intrabone transplantation,(5557) and efforts to enhance the homing and engrafting of cells through ex vivo or in vivo inhibition of CD26, a Dipeptidylpeptidase (DPPIV),(5866) incubation of cells ex vivo with Prostaglandin E,(6770) or by fucosylation of donor cells ex vivo.(71,72)

Ultimately, a better understanding of the biology of HSCs and HPCs will allow for more innovative means to enhance the homing and engraftment of limiting numbers of HSCs and HPCs in CB. Our knowledge of the characteristics and functions of HSCs and HPCs is becoming clearer. We now know much more about the phenotypic,(73) and functional(74,75) characteristics of human HSCs and HPCs, the cytokines that regulate these cells,(76,77) and the BM microenvironmental cells and factors that influence HSCs and HPCs in vivo.(7880)

This review mainly focuses in on some of the more recent work from our laboratory that evaluates intracellular and extracellular factors that influence HSC/HPC function, with the goal to eventually utilize this information to enhance the engrafting capability of HSCs and HPCs for clinical advantage. Through all of this work and for eventual translation of these studies from the lab to the clinic the goal is: “The simpler, the better.” The topics of research from our laboratory to be discussed include: inhibition of CD26/DPPIV, intracellular molecule modulation of HSCs and HPCs, cytokine and growth factor effects on HSCs and HPCs, and cryopreservation of cord blood HSCs and HPCs.

CD26/DPPIV influence on SDF-1/CXCL12-CXCR4 axis and homing/engraftment of HSCs and HPCs

Stromal cell-derived factor (SDF)-1/CXCL12 is a well-known chemokine that acts as a chemotactic (directed cell movement) agent for HSCs and HPCs through its action on the receptor CXCR4. SDF-1/CXCL12 has been implicated in the homing, survival, and nurturing of HSCs and HPCs.(76,8082) CD26/DPPIV is an enzyme that cleaves dipeptides from the N-terminus of proteins after a proline or alanine. SDF-1/CXCL12 is an example of a molecule that has a DPPIV cleavage site. CD26/DPPIV cleavage results in a truncated SDF-1/CXCL12 molecule that is no longer chemotactic.(58) Moreover, the truncated SDF-1/CXCL12 blocks the chemotactic activity of full-length SDF-1/CXCL12.(58) Since SDF-1/CXCL12 acts as an in vivo homing molecule for HSCs and HPCs,(80,81) and a number of cell types including HSCs and HPCs express CD26/DPPIV on their cell surface,(59) we reasoned that inhibition of CD26/DPPIV by small peptide molecules such as diprotin A (ILE-PRO-ILE) or VAL-PYR on the donor cells would enhance the homing and engrafting capability of limiting numbers of donor mouse BM cells in a congenic mouse model of competitive and non-competitive HSC transplantation.(59) This it did, results confirmed and extended by others.(6062) These engraftment enhancing effects of CD26/DPPIV inhibition were extended to pretreatment of human CD34+ donor CB(63,64) and mPB(65) cells transplanted into mice with a NOD/SCID genotype. In vivo inhibition of CD/DPPIV in recipient mice(65,66) was also effective in enhancing engraftment, effects also likely due to effects of DPPIV truncation on non-chemokine growth factors. We recently found that a number of colony stimulating factors (CSFs), such as GM-CSF, G-CSF, IL-3, and EPO contain DPPIV truncation sites, and that DPPIV inhibitors used to pretreat target cells expressing CD26 enhances the detectable activity of the CSFs. Also, in vivo use of DPPIV inhibitors allows accelerated recovery of HSCs and HPCs after stress (e.g. chemotherapy and irradiation) (Broxmeyer, et al., unpublished observations; manuscript in revision). An FDA-approved CD26 inhibitor is currently being evaluated in a pilot clinical study at the Indiana University School of Medicine under the direction of Sherif Farag for its effects on enhancing the engrafting capability of single CB collections in adult patients with leukemia and lymphoma.(66)

Intracellular modulation of HSCs/HPCs

Understanding the intracellular signals involved in HSC and HPC function may lead to successful efforts to manipulate these signaling molecules for clinical benefit.

Effect of the Rheb2-mTOR pathway on HSC engraftment

Rheb is one member of the ras homologue enriched in the brain family of small ras-like GTPase molecules. Rheb cycles between active GTP and inactive GDP-bound forms. Both RHEB1 and RHEB2 are able to activate mammalian target of rapamycin (mTOR) signaling in mammalian cells. Since Rheb2 was found to be preferentially expressed in immature mouse HSCs compared to mature hematopoietic cells,(83) we evaluated effects of Rheb2 overexpression by means of a MIEG3 bicistronic retroviral vector, in mouse BM HSCs and HPCs on the functioning of these cells.(84) In this study, we identified Rheb2 as a pathway important in expansion of immature progenitor cells in vitro and in vivo. However, this expansion was accompanied by a loss of HSC activity. We felt that regulating the activity of the Rheb-mTOR pathway might allow for effective expansion of cells without their loss of HSC repopulating ability. Towards this goal, we treated human CD34+ CB cells ex vivo with a combination of HSC expansion cytokines (SCF, FL, and thrombopoietin (TPO)) in the presence and absence of rapamycin prior to assessing the engrafting capability of these ex vivo cultured cells in sublethally-irradiated NOD/SCID IL-2R gamma chain null (NSG) mice.(85) Ex vivo rapamycin treatment of these cells in the presence of SCF, FL, and TPO greatly enhanced the engrafting capability of the CD34+ CB cells. More mechanistic evaluation of these studies are warranted, as is further preclinical analysis for enhancing CB transplantation.

Tip110/p110nrb/SART3/p110 regulation of hematopoiesis

Tip110 is a Tat-interacting protein of 110 KDa that has been implicated in RNA metabolism and tumor-antigen presentation. Tip110 was found to regulate the transcription of HIV-1 and cellular genes.(86,87) It also functions as a general pre-mRNA splicing factor.(88) The various effects of Tip110 that impinged on transcription factors and cellular gene expression enticed us to evaluate a possible role for Tip110 in the regulation of hematopoiesis. Using Tip110-overexpressing transgenic mice, haploinsufficient (Tip110+/−) mice, and means to up- and downregulate Tip110 expression in human cells through lentiviral gene transduction, we were able to demonstrate that Tip110 transgenic expression increased the numbers, cell cycling status, and survival of HPCs, while Tip110+/− mice manifested opposite effects to the Tip110 transgenic mice in terms of HPC function.(89) Also, Tip110+/− BM HPCs responded better, and Tip110 transgenic BM HPCs worse than control mice to recovery from the cytotoxic effects of 5-flurouracil.(89) Mechanistically, Tip110 regulated expression of CMYC and GATA2 expression, with Tip110 and CMYC regulating the expression of each other, thus linking Tip110 hematopoietic regulation to Tip110 reciprocal regulation of CMYC.(89) We also found that Tip110 was expressed in human embryonic stem (hES) cells, and that it was important for maintenance of expression of pluripotency factors such as NANOG, Oct4, and SOX2, and pluripotency of hES cells.(90) How these Tip110 effects in hES cells are mediated, and its role in induced pluripotent stem (iPS) cells remains to be investigated.

SIRT1 effects on hematopoiesis

SIRT1 is a member of the sirtuin family encompassing seven proteins and histone deacytelases, that has been conserved from bacteria to humans.(91) Mammalian sirtuins have been implicated in numerous cell functions, some with disease relevance.(92) SIRT1 is a human homologue close to the Sir2 yeast protein. We had shown that SIRT1 is involved in regulating apoptosis and Nanog expression in mouse ES cells when in the presence of LIF, at least in part by controlling the subcellular localization of the tumor suppressor p53 protein.(93) We followed up on that study by demonstrating the need for SIRT1 for differentiation of mouse ESCs in the absence of LIF, but presence of 2 mercaptoethanol.(94) A mouse ES cell line deficient in SIRT1 formed few mature blast cell colonies, and the replated cells from these colonies were defective in hematopoietic potential. There was decreased primitive and definitive hematopoiesis associated with LIF removal-induced differentiation of the SIRT1−/− mouse ES cell line, and this corresponded to a delayed capacity to turn off expression of Oct4, nanog, and Fgr5, and to decrease expression of β-H1 globin, β-major globin, and Scl genes. SIRT1−/− mice had fewer yolk sac primitive erythroid precursors, and manifested decreased embryogenesis. In adult mice, both SIRT1−/− and SIRT1+/− BM cells had decreased numbers and cycling status of HPCs, an effect most apparent when cells were cultured in vitro under lowered (5%), compared to normal (~20%) oxygen tension. Thus, these results linked oxygen tension and SIRT1 activity. Most recently, others have suggested that cell-autonomous SIRT1 intracellular signaling may be dispensable for adult HSC functional maintenance in mice.(95) How this above information on adult HSCs and HPCs may be used to modulate and influence engrafting capability remains to be determined. Of interest in this context is that deficiency of SIRT1 in mouse ES cells causes a downregulation of the PTEN-JNK-FOXO1 pathway with a concomitant block in reactive oxygen species (ROS)-induction of apoptosis.(96) Thus, at least in mouse ES cells, SIRT1 appears to play an important role in adjusting the PTEN/JNK/FOXO1 pathway for response to cellular ROS. Mitochondria and mitochondrial-generated ROS are important for hematopoiesis,(97,98) and it is possible that SIRT1 may still play a role in asymmetric divisions of HSCs and self-renewal,(99) especially in cases of heavy stress-induced hematopoiesis. In this regard, energy metabolism is likely crucial to normal, and especially stress-induced hematopoiesis.

We very recently assessed whether cellular energy homeostasis took part in the maintenance of pluripotency and self-renewal in mouse ES cells.(100) AMP-activated protein kinase (AMPK), one regulator for the control of energy metabolism, is activated during stress that induces exhaustion of ATP. We used 5-aminoimidazole-4-carboxyamide ribonucleoside (AICAR) and AMPK activator to demonstrate: activation of the p53-p21cip1 pathway, decreased mouse ES cell proliferation, a G1/S-phase cell cycle block, and decreased expression of the pluripotency markers NANOG and SSEA-1, without effects on Oct4 expression. This was associated with enhanced differentiation of erythroid cells from mES cells.

DEK regulation of hematopoiesis

DEK is a relatively newly-defined molecule involved in a number of cellular activities, such as transcriptional repression and activation, processing of mRNA, and chromatin remodeling.(101,102) DEK is a unique molecule in that it can be found in the nuclear, yet it can also be secreted outside of the cell and influence other cell types.(101) It can act as a chemoattractant for specific mature blood cells (CD8+ T cells and natural killer cells) after being released from macrophages. (103) We identified a role for DEK in hematopoiesis.(104) There were increased numbers of HPCs in BM and spleen of DEK/ mice, and purified recombinant DEK protein suppressed in vitro colony formation by normal mouse BM and human CB HPC colony formation in vitro. This negative effect of DEK on HPC proliferation in vitro and in vivo was associated with decreased long-term competitive and secondary mouse HSC repopulating capacity, suggesting a positive role for DEK in HSC functions such as engraftment. What these hematopoietic effects of DEK are due to requires further investigation, but this information may be of potential clinical relevance.

Role for cyclin-dependent kinase inhibitors in hematopoiesis

A number of cyclin-dependent kinase inhibitors (CDKIs) have been implicated in HSC and HPC function.(76,105108) These include p21cip1/waf1 (p21), p27kip1 (p27), and p18INK4c (p18). However, other than a paper demonstrating that p18/ counters the exhaustion of p21/ HSCs after serial transplantation,(109) there is nothing else in the literature that links the different CDKIs and their networking interactions for HSC and HPC function. We noted that CDKIs had different effects on HPC proliferation, and that they differentially modulated the responsiveness of HPCs to synergistic combinations of cytokines such as a CSF plus SCF.(110) Deletion of p18 resulted in decreased numbers and proliferation of HPCs, effects similar to that previously reported by us for p21/ mice.(77) These positive effects of p18 dominated over the negative effects of p27 where p27/ was associated with enhanced HPC proliferation. The responsiveness of HPCs to suppression by certain chemokine family members was directly related to the ability of HPCs to respond to synergistic stimulation, and cycling HPCs. Deletion of the p18 gene rescued the loss of chemokine suppression of synergistic cell proliferation associated with deletion of the p21 gene. Thus, there is interplay of cell cycle regulators on HPC proliferation, and loss of one CDKI can sometimes be compensated for by another missing CDKI.

Cytokine/growth factor regulation of hematopoiesis

Numerous cytokines are known to regulate the proliferation and survival of HPCs.(76,77,108)

Role of immune cells in the regulation of hematopoiesis

T lymphocytes play a role in the proliferation of HPCs and their homeostasis,(111113) in part through the transcription factors STAT4, STAT6, BCL-6, and BAZF, and the growth factor oncostatin M. We recently reported a role for STAT3-dependent IL-21 production from helper T cells in maintenance of HPC homeostasis.(114) There was decreased activity of HPC proliferation in mice with a specific deficiency of STAT3 in T cells, and STAT3 expression was required for production of IL-21. Neutralization of IL-21, but not IL-22, resulted in a decrease in HPC number and cycling, similar to that seen in STAT3-deficient T cell mice. Moreover, exogenous administration of IL-21 was able to rescue suppressed HPC proliferation in mice with STAT3−/− T cells.

Angiopoietin-like molecules -2 and -3 enhance survival of HPCs in CB

Angiopoietin-lilke (ANGPLT) molecules have been implicated in regulation of mouse fetal liver and BM HSCs and human CB NOD/SCID repopulating cells,(115117) but no information was available on effects of ANGPTL molecules on HPCs. We identified the actions of ANGPTL-2 and -3 molecules on enhancement of the survival and replating ability of HPCs from human CB.(118) These activities of ANGPTL-2 and -3 were manifested through the coiled-coil domains of these proteins. We did not detect functional activities of ANGPTL-4, -5, -6, or -7 on HPC survival or replating capacity, and none of the ANGPTL molecules tested influenced the proliferation of CB HPCs. The survival and replating effects of ANGPTL-2 and -3 on HPCs(118) and that of such ANGPTL molecules on expanding NOD/SCID repopulating human HPCs(116) may be of future relevance to CB transplantation.

A role for neuronally-active molecules on hematopoiesis

The nervous system has been implicated in microenvironmental control of hematopoiesis.(79) This opened up the possibility that other neuronally-associated proteins and their receptors could play a role in hematopoiesis. We identified such a role for neurexophilin1.(119) Neurexophilins bind neurexin1α. neurexin1α, and dystroglycan are membrane receptors. They serve as mutual ligands within the neuronal system. We found that neurexophilin1 was able to suppress the proliferation of HPCs, and it acted through neurexin1α, an effect that could be counter-modulated by dystroglycan. The suppressive effect of neurexophilin1 on HPCs was direct-acting on the HPCs and inhibition was apparent both in vitro on human CB and mouse BM HPCs, and in vivo after injection of recombinant neurexophilin1 into mice. Thus, a signaling axis in the hematopoietic system centered on neurexin1α and its modulation by neurexophilin1 and dystroglycan. Additional information on links between the nervous and hematopoietic systems may offer the opportunity to modulate one for the benefit of the other in a transplant setting.

Potential for modulation of intracellular signals by cytokines and growth factors and other means for enhanced CB transplantation

There are many transcription factors and other intracellular signaling molecules that impinge on HSC and HPC numbers and functions.(76,108) How all these different intracellular factors may interact with each other if at all, will need to be better elucidated if they are to be modulated for clinical advantage with minimal side-effects. Interconnected with these intracellular molecules are numerous cytokines and growth factors that can trigger/activate these intracellular molecules.(76,77) While much is known regarding cytokines and growth factor effects on HPC function, we still know very little of how these cytokines/growth factors influence HSC functions such as self-renewal, survival, and engraftment. Future information in these areas will likely have a positive influence on how we might be able to enhance HSC transplantation, especially with CB cells.

Cryopreservation of CB HSCs, HPCs, and other cells

CB transplantation is critically-dependent on CB banking, which in turn is dependent on the capacity to adequately cryopreserve the HSCs and HPCs in CB, and maintain these cells in a frozen state. Many banks, both public and private, have been formed in order to supply cryopreserved CBs for transplantation purposes. However, how long such frozen CB units can be stored and then thawed for efficient recovery of HSCs and HPCs is critical information for the success of CB banking and CB transplantation. Over the last 23 plus years we have reported on the cryopreservation and subsequent recovery of thawed cells.(1,7,120,121) Most recently, we demonstrated that we could recover functionally-intact HPCs at high efficiency from cryopreserved CB after thawing of cells stored frozen for up to 21–23.5 years.(122) While there was a range of recoveries of HPCs from different CB units, 80–100% recovery was apparent for most samples, with maintenance of high proliferative and replating capability. Moreover, CB cryopreserved for up to 21 years could be thawed, the CD34+ cells isolated, and these cells could be used to long-term repopulate primary and secondary immune-deficient mice suggesting that long-term marrow repopulating and self-renewing HSCs had been adequately cryopreserved and could be retrieved after thawing with excellent intact functional capabilities. From the long-term cryopreserved cells, we were also able to retrieve functionally responsive CD4+ and CD8+ T cells, and high proliferative potential endothelial colony forming cells (=endothelial progenitor cells), and we were able to generate from the thawed and subsequently purified CD34+ cell population, iPS cells that could be differentiated into all three germ cell lineages in vitro and in vivo. (122) Subsequent studies with these iPS cells found that only a percentage of these cells were fully reprogrammed. Efforts are underway to enhance the full reprogramming capacity of a larger percent of these iPS cells by modulating expression of the micro RNA 302 cluster and its downstream targets. However, how effective these generated iPS cells may be for future regenerative medicine possibilities remains to be determined.

Concluding remarks

The intent of this article was to focus mainly on laboratory and translational research in the author’s laboratory. There are many outstanding laboratories working on means to gain a better mechanistic insight into optimizing the functional activities of HSCs and HPCs. It is hoped that together this work will translate into clinical utility for health benefit, with one such benefit being enhanced efficacy of cord blood HSC/HPC transplantation.

Acknowledgments

Most of the studies in the reference list reported from the author’s laboratory were funded through NIH R01 or NIH P01 grants. Dr. Broxmeyer is currently Principal Investigator (PI) on and supported by Public Health Service Grant R01 HL056416, R01 HL67384, R01 HL112669, and P01 DK090948 from the National Institutes of Health. He is also PI on NIH Training Grants: T32 DK07519 and T32 HL07910.

Footnotes

Conflicts of interest

The author is on the Medical Scientific Advisory Board of CordUse, a cord blood banking company.

References

  • 1.Broxmeyer HE, Douglas GW, Hangoc G, Cooper S, Bard J, English D, Arny M, Thomas L, Boyse EA. 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]
  • 2.Broxmeyer HE, Gluckman E, Auerbach A, Douglas GW, Friedman H, Cooper S, Hangoc G, Kurtzberg J, Bard J, Boyse EA. Human umbilical cord blood: A clinically useful source of transplantable hematopoietic stem/progenitor cells. Int J Cell Cloning. 1990;8(Suppl 1):76–91. doi: 10.1002/stem.5530080708. [DOI] [PubMed] [Google Scholar]
  • 3.Auerbach AD, Liu Q, Ghosh R, Pollack MS, Douglas GW, Broxmeyer HE. Prenatal identification of potential donors for umbilical cord blood transplantation for Fanconi Anemia. Transfusion. 1990;30:682–687. doi: 10.1046/j.1537-2995.1990.30891020324.x. [DOI] [PubMed] [Google Scholar]
  • 4.Pollack MS, Auerbach AD, Broxmeyer HE, Zaafran A, Erlich HA. The use of DNA amplification for DQ typing as an adjunct to serological prenatal HLA typing for the identification of potential donors for umbilical cord blood transplantation. Human Immunol. 1991;30:45–49. doi: 10.1016/0198-8859(91)90070-p. [DOI] [PubMed] [Google Scholar]
  • 5.Broxmeyer HE, Kurtzberg J, Gluckman E, Auerbach AD, Douglas G, Cooper S, Falkenburg JHF, Bard J, Boyse EA. Umbilical cord blood hematopoietic stem and repopulating cells in human clinical transplantation. Blood Cells. 1991;17:313–329. [PubMed] [Google Scholar]
  • 6.Carow C, Hangoc G, Cooper S, Williams DE, Broxmeyer HE. Mast cell growth factor (c-kit ligand) supports the growth of human multipotential (CFU-GEMM) progenitor cells with a high replating potential. Blood. 1991;78:2216–2221. [PubMed] [Google Scholar]
  • 7.Broxmeyer HE, Hangoc G, Cooper S, Ribeiro RC, Graves V, Yoder M, Wagner J, Vadhan-Raj S, Rubinstein P, Broun ER. Growth characteristics and expansion of human umbilical cord blood and estimation of its potential for transplantation of adults. Proc Natl Acad Sci USA. 1992;89:4109–4113. doi: 10.1073/pnas.89.9.4109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Carow CE, Hangoc G, Broxmeyer HE. Human multipotential progenitor cells (CFU-GEMM) have extensive replating capacity for secondary CFU-GEMM: An effect enhanced by cord blood plasma. Blood. 1993;81:942–949. [PubMed] [Google Scholar]
  • 9.Lu L, Xiao M, Shen RN, Grigsby S, Broxmeyer HE. Enrichment, characterization and responsiveness of single primitive CD34+++ human umbilical cord blood hematopoietic progenitor with high proliferative and replating potential. Blood. 1993;81:41–48. [PubMed] [Google Scholar]
  • 10.Gluckman E, Broxmeyer HE, Auerbach AD, Friedman H, Douglas GW, Devergie A, Esperou H, Thierry D, Socie G, Lehn P, Cooper S, English D, Kurtzberg J, Bard J, Boyse EA. Hematopoietic reconstitution in a patient with Fanconi anemia by means of umbilical-cord blood from an HLA-identical sibling. New Engl J Medicine. 1989;321:1174–1178. doi: 10.1056/NEJM198910263211707. [DOI] [PubMed] [Google Scholar]
  • 11.Wagner JE, Broxmeyer HE, Byrd RL, Zehnbauer B, Schmeckpeper B, Shah N, Griffin C, Emanuel P, Zuckerman K, Cooper S, Carow C, Bias W, Santos GW. Transplantation of umbilical cord blood after myeloblative therapy: analysis of engraftment. Blood. 1992;79:1874–1881. [PubMed] [Google Scholar]
  • 12.Kohli-Kumar M, Shahidi NT, Broxmeyer HE, Masterson M, Delaat C, Sambrano J, Morris C, Auerbach AD, Harris RE. Haematopoietic stem/progenitor cell transplant in Fanconi anemia using HLA-matched sibling umbilical cord blood cells. Brit J Haematol. 1993;85:419–422. doi: 10.1111/j.1365-2141.1993.tb03192.x. [DOI] [PubMed] [Google Scholar]
  • 13.Wagner JE, Kernan NA, Steinbuch M, Broxmeyer HE, Gluckman E. Allogeneic sibling umbilical cord blood transplantation in forty-four children with malignant and non-malignant disease. Lancet. 1995;346:214–219. doi: 10.1016/s0140-6736(95)91268-1. [DOI] [PubMed] [Google Scholar]
  • 14.Broxmeyer HE, Smith FO. Cord Blood Hematopoietic Cell Transplantation. In: Appelbaum FR, Forman SJ, Negrin RS, Blume KG, editors. Thomas’ Hematopoietic Cell Transplantation. 4. Section 4. Wiley-Blackwell, West Sussex; United Kingdom: 2009. pp. 559–576. Chapter 39. [Google Scholar]
  • 15.Rocha V, Broxmeyer HE. New approaches for improving engraftment after cord blood transplantation. Blood and Bone Marrow Transplantation. 2009;16:S126–S132. doi: 10.1016/j.bbmt.2009.11.001. [DOI] [PubMed] [Google Scholar]
  • 16.Rubinstein P, Rosenfield RE, Adamson JW, Stevens CE. Stored placental blood for unrelated bone marrow reconstitution. Blood. 1993;81:1679–1690. [PubMed] [Google Scholar]
  • 17.Rubinstein P, Dobrila L, Rosenfield RE, Adamson JW, Migliaccio G, Migliaccio AR, Taylor PE, Stevens CE. Processing and cryopreservation of placental/umbilical cord blood for unrelated bone marrow reconstitution. Proc Natl Acad Sci U S A. 1995;92:10119–10122. doi: 10.1073/pnas.92.22.10119. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Guindi ES, Moss TF, Ernst MT. Public and Private Cord Blood Banking. In: Broxmeyer HE, editor. Cord Blood Biology, Transplantation, Banking, and Regulation. Chapter 34. AABB Press; 2011. pp. 595–628. [Google Scholar]
  • 19.Regan DM. Cord Blood Banking: The Development and Application of Cord Blood Banking Processes, Standards, and Regulations. In: Broxmeyer HE, editor. Cord Blood Biology, Transplantation, Banking, and Regulation. Chapter 35. AABB Press; 2011. pp. 633–644. [Google Scholar]
  • 20.Sniecinski I. Cord Blood Banking in Developing Countries. In: Broxmeyer HE, editor. Cord Blood Biology, Transplantation, Banking, and Regulation. Chapter 36. AABB Press; 2011. pp. 647–661. [Google Scholar]
  • 21.Boo M, Welte K, Confer D. Accreditation and Regulation of Cord Blood Banking. In: Broxmeyer HE, editor. Cord Blood Biology, Transplantation, Banking, and Regulation. Chapter 37. AABB Press; 2011. pp. 663–671. [Google Scholar]
  • 22.Welte K, Boo M, Confer D. Worldwide Searching and Distribution of Cord Blood Units. In: Broxmeyer HE, editor. Cord Blood Biology, Transplantation, Banking, and Regulation. Chapter 38. AABB Press; 2011. pp. 673–684. [Google Scholar]
  • 23.Kurtzberg J, Laughlin M, Graham ML, Smith C, Olson JF, Halperin EC, Ciocci G, Carrier C, Stevens CE, Rubinstein P. Placental blood as a source of hematopoietic stem cells for transplantation into unrelated recipients. N Engl J Med. 1996;335:157–166. doi: 10.1056/NEJM199607183350303. [DOI] [PubMed] [Google Scholar]
  • 24.Wagner JE, Rosenthal J, Sweetman R, Shu XO, Davies SM, Ramsay NK, McGlave PB, Sener L, Cairo MS. Successful transplantation of HLA-matched and HLA-mismatched umbilical cord blood from unrelated donors: analysis of engraftment and acute graft-versus-host disease. Blood. 1996;88:795–802. [PubMed] [Google Scholar]
  • 25.Gluckman E, Rocha V, Boyer-Chammard A, Locatelli F, Arcese W, Pasquini R, Ortega J, Souillet G, Ferreira E, LaPorte JP, Fernandez M, Chastang C. 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]
  • 26.Rubinstein P, Carrier C, Scaradavou A, Kurtzberg J, Adamson J, Migliaccio AR, Berkowitz RL, Cabbad M, Dobrila NL, Taylor PE, Rosenfield RE, Stevens CE. Outcomes among 562 recipients of placental-blood transplants from unrelated donors. N Engl J Med. 1998;339:1565–1577. doi: 10.1056/NEJM199811263392201. [DOI] [PubMed] [Google Scholar]
  • 27.Rocha V, Wagner JE, Jr, Sobocinski KA, Klein JP, Zhang MJ, Horowitz MM, Gluckman E. Graft-versus-host disease in children who have received a cord-blood or bone marrow transplant from an HLA-identical sibling. Eurocord and International Bone Marrow Transplant Registry Working Committee on Alternative Donor and Stem Cell Sources. N Engl J Med. 2000;342:1846–1854. doi: 10.1056/NEJM200006223422501. [DOI] [PubMed] [Google Scholar]
  • 28.Laughlin MJ, Barker J, Bambach B, Koc ON, Rizzieri DA, Wagner JE, Gerson SL, Lazarus HM, Cairo M, Stevens CE, Rubinstein P, Kurtzberg J. Hematopoietic engraftment and survival in adult recipients of umbilical-cord blood from unrelated donors. N Engl J Med. 2001;344:1815–1822. doi: 10.1056/NEJM200106143442402. [DOI] [PubMed] [Google Scholar]
  • 29.Rocha V, Cornish J, Sievers EL, Filipovich A, Locatelli F, Peters C, Remberger M, Michel G, Arcese W, Dallorso S, Tiedemann K, Busca A, Chan KW, Kato S, Ortega J, Vowels M, Zander A, Souillet G, Oakill A, Woolfrey A, Pay AL, Green A, Garnier F, Ionescu I, Wernet P, Sirchia G, Rubinstein P, Chevret S, Gluckman E. Comparison of outcomes of unrelated bone marrow and umbilical cord blood transplants in children with acute leukemia. Blood. 2001;97:2962–2971. doi: 10.1182/blood.v97.10.2962. [DOI] [PubMed] [Google Scholar]
  • 30.Wagner JE, Barker JN, Defor TE, Baker KS, Blazar BR, Eide C, Goldman A, Kersey J, Krivit W, Macmillan ML, Orchard PJ, Peters C, Weisdorf DJ, Ramsay NK, Davies SM. Transplantation of unrelated donor umbilical cord blood in 102 patients with malignant and nonmalignant diseases: influence of CD34 cell dose and HLA disparity on treatment-related mortality and survival. Blood. 2002;100:1611–1618. doi: 10.1182/blood-2002-01-0294. [DOI] [PubMed] [Google Scholar]
  • 31.Barker JN, Weisdorf DJ, Defor TE, Blazar BR, Miller JS, Wagner JE. Rapid and complete donor chimerism in adult recipients of unrelated donor umbilical cord blood transplantation after reduced-intensity conditioning. Blood. 2003;102:1915–1919. doi: 10.1182/blood-2002-11-3337. [DOI] [PubMed] [Google Scholar]
  • 32.Rocha V, Labopin M, Sanz G, Arcese W, Schwerdtfeger R, Bosi A, Jacobsen N, Ruutu T, De LM, Finke J, Frassoni F, Gluckman E. Transplants of umbilical-cord blood or bone marrow from unrelated donors in adults with acute leukemia. N Engl J Med. 2004;351:2276–2285. doi: 10.1056/NEJMoa041469. [DOI] [PubMed] [Google Scholar]
  • 33.Laughlin MJ, Eapen M, Rubinstein P, Wagner JE, Zhang MJ, Champlin RE, Stevens C, Barker JN, Gale RP, Lazarus HM, Marks DI, Van Rood JJ, Scaradavou A, Horowitz MM. Outcomes after transplantation of cord blood or bone marrow from unrelated donors in adults with leukemia. N Engl J Med. 2004;351:2265–2275. doi: 10.1056/NEJMoa041276. [DOI] [PubMed] [Google Scholar]
  • 34.Brunstein CG, Barker JN, Weisdorf DJ, Defor TE, Miller JS, Blazar BR, Mcglave PB, Wagner JE. Umbilical cord blood transplantation after nonmyeloablative conditioning: impact on transplantation outcomes in 110 adults with hematologic disease. Blood. 2007;110:3064–3070. doi: 10.1182/blood-2007-04-067215. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Eapen M, Rubinstein P, Zhang MJ, Stevens C, Kurtzberg J, Scaradavou A, Loberiza FR, Champlin RE, Klein JP, Horowitz MM, Wagner JE. Outcomes of transplantation of unrelated donor umbilical cord blood and bone marrow in children with acute leukaemia: a comparison study. Lancet. 2007;369:1947–1954. doi: 10.1016/S0140-6736(07)60915-5. [DOI] [PubMed] [Google Scholar]
  • 36.Kurtzberg J, V, Prasad K, Carter SL, Wagner JE, Baxter-Lowe LA, Wall D, Kapoor N, Guinan EC, Feig SA, Wagner EL, Kernan NA. Results of the Cord Blood Transplantation Study (COBLT): clinical outcomes of unrelated donor umbilical cord blood transplantation in pediatric patients with hematologic malignancies. Blood. 2008;112:4318–4327. doi: 10.1182/blood-2007-06-098020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Eapen M, Rocha V, Sanz G, Scaradavou A, Zhang MJ, Arcese W, Sirvent A, Champlin RE, Chao N, Gee AP, Isola L, Laughlin MJ, Marks DI, Nabhan S, Ruggeri A, Soiffer R, Horowitz MM, Gluckman E, Wagner JE. Effect of graft source on unrelated donor haemopoietic stem-cell transplantation in adults with acute leukaemia: a retrospective analysis. Lancet Oncol. 2010;11:653–660. doi: 10.1016/S1470-2045(10)70127-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Eapen M. Cord Blood Transplantation for Leukemia in Children. In: Broxmeyer HE, editor. Cord Blood Biology, Transplantation, Banking, and Regulation. Chapter 26. AABB Press; 2011. pp. 467–474. [Google Scholar]
  • 39.Horwitz ME, Chao N. Umbilical Cord Blood Transplantation for Treatment of Nonmalignant Disorders. In: Broxmeyer HE, editor. Cord Blood Biology, Transplantation, Banking, and Regulation. Chapter 27. AABB Press; 2011. pp. 477–483. [Google Scholar]
  • 40.Gluckman E, Ruggeri A, Rocha V. Extending the Use of Cord Blood Cells to Adults. In: Broxmeyer HE, editor. Cord Blood Biology, Transplantation, Banking, and Regulation. Chapter 28. AABB Press; 2011. pp. 487–499. [Google Scholar]
  • 41.Brunstein CG, Wagner JE. Double Umbilical Cord Blood Transplantation for Malignant Disorders. In: Broxmeyer HE, editor. Cord Blood Biology, Transplantation, Banking, and Regulation. Chapter 29. AABB Press; 2011. pp. 503–524. [Google Scholar]
  • 42.Ponce DM, Barker JN. Determinants of Engraftment after Double-Unit Cord Blood Transplantation. In: Broxmeyer HE, editor. Cord Blood Biology, Transplantation, Banking, and Regulation. Chapter 30. AABB Press; 2011. pp. 529–541. [Google Scholar]
  • 43.Chen Y-B, Cutler C, Ballen K. Reduced-Intensity Conditioning and Double Umbilical Cord Blood Transplantation. In: Broxmeyer HE, editor. Cord Blood Biology, Transplantation, Banking, and Regulation. Chapter 31. AABB Press; 2011. pp. 545–553. [Google Scholar]
  • 44.McCullough J, McKenna D. Management of Umbilical Cord Blood at the Transplant Center. In: Broxmeyer HE, editor. Cord Blood Biology, Transplantation, Banking, and Regulation. Chapter 33. AABB Press; 2011. pp. 585–592. [Google Scholar]
  • 45.Petz LD, Spellman SR, Gragert L. The Underutilization of Cord Blood Transplantation: Extent of the Problem, Causes, and Methods of Improvement. In: Broxmeyer HE, editor. Cord Blood Biology, Transplantation, Banking, and Regulation. Chapter 32. AABB Press; 2011. pp. 557–581. [Google Scholar]
  • 46.Broxmeyer HE, Cooper S, Haas DM, Hathaway JK, Stehman FB, Hangoc G. Experimental Basis of Cord Blood Transplantation. Bone Marrow Transplantation (Special Issue) 2009;44:627–633. doi: 10.1038/bmt.2009.285. [DOI] [PubMed] [Google Scholar]
  • 47.Barker JN, Weisdorf DJ, Defor TE, Blazar BR, Mcglave PB, Miller JS, Verfaillie CM, Wagner JE. Transplantation of 2 partially HLA-matched umbilical cord blood units to enhance engraftment in adults with hematologic malignancy. Blood. 2005;105:1343–1347. doi: 10.1182/blood-2004-07-2717. [DOI] [PubMed] [Google Scholar]
  • 48.Ballen KK, Spitzer TR, Yeap BY, Mcafee S, Dey BR, Attar E, Haspel R, Kao G, Liney D, Alyea E, Lee S, Cutler C, Ho V, Soiffer R, Antin JH. Double unrelated reduced-intensity umbilical cord blood transplantation in adults. Biol Blood Marrow Transplant. 2007;13:82–89. doi: 10.1016/j.bbmt.2006.08.041. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Brunstein CG, Gutman JA, Weisdorf DJ, Woolfrey AE, Defor TE, Gooley TA, Verneris MR, Appelbaum FR, Wagner JE, Delaney C. Allogeneic hematopoietic cell transplantation for hematologic malignancy: relative risks and benefits of double umbilical cord blood. Blood. 2010;116:4693–4699. doi: 10.1182/blood-2010-05-285304. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Cutler C, Stevenson K, Kim HT, Brown J, Mcdonough S, Herrera M, Reynolds C, Liney D, Kao G, Ho V, Armand P, Koreth J, Alyea E, Dey BR, Attar E, Spitzer T, Boussiotis VA, Ritz J, Soiffer R, Antin JH, Ballen K. Double umbilical cord blood transplantation with reduced intensity conditioning and sirolimus-based GVHD prophylaxis. Bone Marrow Transplant. 2011;46:659–667. doi: 10.1038/bmt.2010.192. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Shpall EJ, Quinones R, Giller R, Zeng C, Baron AE, Jones RB, Bearman SI, Nieto Y, Freed B, Madinger N, Hogan CJ, Slat-Vasquez V, Russell P, Blunk B, Schissel D, Hild E, Malcolm J, Ward W, Mcniece IK. Transplantation of ex vivo expanded cord blood. Biol Blood Marrow Transplant. 2002;8:368–376. doi: 10.1053/bbmt.2002.v8.pm12171483. [DOI] [PubMed] [Google Scholar]
  • 52.Jaroscak J, Goltry K, Smith A, Waters-Pick B, Martin PL, Driscoll TA, Howrey R, Chao N, Douville J, Burhop S, Fu P, Kurtzberg J. Augmentation of umbilical cord blood (UCB) transplantation with ex vivo-expanded UCB cells: results of a phase 1 trial using the AastromReplicell System. Blood. 2003;101:5061–5067. doi: 10.1182/blood-2001-12-0290. [DOI] [PubMed] [Google Scholar]
  • 53.Delaney C, Heimfeld S, Brashem-Stein C, Voorhies H, Manger RL, Bernstein ID. Notch-mediated expansion of human cord blood progenitor cells capable of rapid myeloid reconstitution. Nat Med. 2010;16:232–236. doi: 10.1038/nm.2080. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Delaney C. Ex vivo Expansion of Cord Blood Stem and Progenitor Cells. In: Broxmeyer HE, editor. Cord Blood Biology, Transplantation, Banking, and Regulation. Chapter 12. AABB Press; 2011. pp. 215–224. [Google Scholar]
  • 55.Frassoni F, Gualandi F, Podesta M, Raiola AM, Ibatici A, Piaggio G, Sessarego M, Sessarego N, Gobbi M, Sacchi N, Labopin M, Bacigalupo A. Direct intrabone transplant of unrelated cord-blood cells in acute leukaemia: a phase I/II study. Lancet Oncol. 2008;9:831–839. doi: 10.1016/S1470-2045(08)70180-3. [DOI] [PubMed] [Google Scholar]
  • 56.Brunstein CG, Barker JN, Weisdorf DJ, Defor TE, Mckenna D, Chong SY, Miller JS, Mcglave PB, Wagner JE. Intra-BM injection to enhance engraftment after myeloablative umbilical cord blood transplantation with two partially HLA-matched units. Bone Marrow Transplant. 2009;43:935–940. doi: 10.1038/bmt.2008.417. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Frassoni F, Podesta M, Varaldo R, Magrassi L, Gualandi F, Bacigalupo A, Buschiazzo A, Fiz F, Uccelli A, Sacchi N, Marini C, Piana M, Sambuceti G. Intrabone Transplantation of Cord Blood Cells and the Journey of Hematopoietic Cells. In: Broxmeyer HE, editor. Cord Blood Biology, Transplantation, Banking, and Regulation. Chapter 14. AABB Press; 2011. pp. 239–250. [Google Scholar]
  • 58.Christopherson KW, Hangoc G, Broxmeyer HE. Cell surface peptidase CD26/DPPIV regulates CXCL12/SDF-1α mediated chemotaxis of human CD34+ progenitor cells. J Immunol. 2002;169:7000–7008. doi: 10.4049/jimmunol.169.12.7000. [DOI] [PubMed] [Google Scholar]
  • 59.Christopherson KW, II, Hangoc G, Mantel C, Broxmeyer HE. Modulation of hematopoietic stem cell homing and engraftment by CD26. Science. 2004;305:1000–1003. doi: 10.1126/science.1097071. [DOI] [PubMed] [Google Scholar]
  • 60.Tian C, Bagley J, Forman D, Iacomini J. Inhibition of CD26 peptidase activity significantly improves engraftment of retrovirally transduced hematopoietic progenitors. Gene Ther. 2006;13:652–658. doi: 10.1038/sj.gt.3302695. [DOI] [PubMed] [Google Scholar]
  • 61.Peranteau WH, Endo M, Adibe OO, Merchant A, Zoltick PW, Flake AW. CD26 inhibition enhances allogeneic donor-cell homing and engraftment after in utero hematopoietic-cell transplantation. Blood. 2006;108:4268–4274. doi: 10.1182/blood-2006-04-018986. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Wyss BK, Donnelly AFW, Zhou D, Sinn AL, Pollok KE, Goebel WS. Enhanced homing and engraftment of fresh but not ex vivo cultured murine marrow cells in submyeloablated hosts following CD26 inhibition by Diprotin A. Exp Hematol. 2009;37:814–823. doi: 10.1016/j.exphem.2009.03.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Campbell TB, Hangoc G, Liu Y, Pollok K, Broxmeyer HE. Inhibition of CD26 in Human Cord Blood CD34+ Cells Enhances their Engraftment of Nonobese Diabetic/Severe Combined Immunodeficiency Mice. Stem Cells and Development. 2007;16:347–354. doi: 10.1089/scd.2007.9995. [DOI] [PubMed] [Google Scholar]
  • 64.Christopherson KW, II, Paganessi LA, Napier S, Porecha NK. CD26 Inhibition on CD34+ or Lineage Human Umbilical Cord Blood Donor Hematopoietic Stem Cells/Hematopoietic Progenitor Cells Improves Long-Term Engraftment into NOD/SCID/Beta2null Immunodeficient Mice. Stem Cells and Develop. 2007;16:355–360. doi: 10.1089/scd.2007.9996. [DOI] [PubMed] [Google Scholar]
  • 65.Kawai T, Choi U, Liu PO, Whiting-Theobald NL, Linton GF, Malech HL. Diprotin A Infusion into Nonobese Diabetic/Severe Combined Immunodeficiency Mice Markedly Enhances Engraftment of Human Mobilized CD34+ Peripheral Blood Cells. Stem Cells and Develop. 2007;16:361–370. doi: 10.1089/scd.2007.9997. [DOI] [PubMed] [Google Scholar]
  • 66.Farag SS, Broxmeyer HE. CD26 Inhibition to Enhance Cord Blood Engraftment. In: Broxmeyer HE, editor. Cord Blood Biology, Transplantation, Banking, and Regulation. Chapter 11. AABB Press; 2011. pp. 203–210. [Google Scholar]
  • 67.North TE, Goessling W, Walkley CR, Lengerke C, Kopani KR, Lord AM, Weber GJ, Bowman TV, Jang IH, Grosser T, Fitzgerald GA, Daley GQ, Orkin SH, Zon LI. Prostaglandin E2 regulates vertebrate haematopoietic stem cell homeostasis. Nature. 2007;447:1007–1011. doi: 10.1038/nature05883. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Hoggatt J, Singh P, Sampath J, Pelus LM. Prostaglandin E2 enhances hematopoietic stem cell homing, survival, and proliferation. Blood. 2009;113:5444–5455. doi: 10.1182/blood-2009-01-201335. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Durand EM, Zon LI. Newly emerging roles for prostaglandin E2 regulation of hematopoiesis and hematopoietic stem cell engraftment. Curr Opin Hematol. 2010;17:308–312. doi: 10.1097/MOH.0b013e32833a888c. [DOI] [PubMed] [Google Scholar]
  • 70.Pelus LM, Speth J, Hoggatt J. Prostaglandin E2 and other Eicosanoid-Based Strategies to Enhance Engraftment. In: Broxmeyer HE, editor. Cord Blood Biology, Transplantation, Banking, and Regulation. Chapter 10. AABB Press; 2011. pp. 187–197. [Google Scholar]
  • 71.Xia L, Mcdaniel JM, Yago T, Doeden A, Mcever RP. Surface fucosylation of human cord blood cells augments binding to P-selectin and E-selectin and enhances engraftment in bone marrow. Blood. 2004;104:3091–3096. doi: 10.1182/blood-2004-02-0650. [DOI] [PubMed] [Google Scholar]
  • 72.Robinson SN, Simmons PJ, Zweidler-McKay PA, Shpall EJ. Fucosylation for Improved Engraftment. In: Broxmeyer HE, editor. Cord Blood Biology, Transplantation, Banking, and Regulation. Chapter 13. AABB Press; 2011. pp. 227–234. [Google Scholar]
  • 73.Chitteti BR, Kacena MA, Srour EF. Phenotypic Characterization of Hematopoietic Stem Cells. In: Broxmeyer HE, editor. Cord Blood Biology, Transplantation, Banking, and Regulation. Chapter 4. AABB Press; 2011. pp. 75–83. [Google Scholar]
  • 74.Cai S, Wang H, Pollok KE. Immunodeficient Mouse Models for Assessing Human Hematopoiesis. In: Broxmeyer HE, editor. Cord Blood Biology, Transplantation, Banking, and Regulation. Chapter 5. AABB Press; 2011. pp. 87–106. [Google Scholar]
  • 75.Notta F, Doulatov S, Laurenti E, Poeppl A, Jurisica I, Dick JE. Isolation of Single Human Hematopoietic Stem Cells Capable of Long-Term Multilineage Engraftment. Science. 2011;333:218–221. doi: 10.1126/science.1201219. [DOI] [PubMed] [Google Scholar]
  • 76.Shaheen M, Broxmeyer HE. The humoral regulation of hematopoiesis. In: Hoffman R, Benz EJ Jr, Shattil SJ, Furie B, Silberstein LE, McGlave P, Heslop H, Anastasi J, editors. Hematology: Basic Principles and Practice. 5. Part III. Chapter 24. Elsevier Churchill Livingston; Philadelphia, PA: 2009. pp. 253–275. [Google Scholar]
  • 77.Shaheen M, Broxmeyer HE. Hematopoietic Cytokines and Growth Factors. In: Broxmeyer HE, editor. Cord Blood Biology, Transplantation, Banking, and Regulation. Chapter 3. AABB Press; 2011. pp. 35–63. [Google Scholar]
  • 78.Chute J. Regulation of Hematopoiesis by the Bone Marrow Vascular Niche. In: Broxmeyer HE, editor. Cord Blood Biology, Transplantation, Banking, and Regulation. Chapter 7. AABB Press; 2011. pp. 133–147. [Google Scholar]
  • 79.Lucas D, Frenette PS. Cellular Components and Regulation of the Hematopoietic Stem Cell Niche. In: Broxmeyer HE, editor. Cord Blood Biology, Transplantation, Banking, and Regulation. Chapter 8. AABB Press; 2011. pp. 153–162. [Google Scholar]
  • 80.Kalinkovich A, Canaani J, Lapidot T. The Dynamic and Modifiable Nature of Hematopoietic Stem Cells and Their Niches. In: Broxmeyer HE, editor. Cord Blood Biology, Transplantation, Banking, and Regulation. Chapter 9. AABB Press; 2011. pp. 169–182. [Google Scholar]
  • 81.Peled A, Petit I, Kollet O, Magid M, Ponomaryov T, Byk T, Nagler A, Ben-Hur H, Many A, Shultz L, Lider O, Alon R, Zipori D, Lapidot T. Dependence of human stem cell engraftment and repopulation of NOD/SCID mice on CXCR4. Science. 1999;283:845–848. doi: 10.1126/science.283.5403.845. [DOI] [PubMed] [Google Scholar]
  • 82.Broxmeyer HE, Cooper S, Kohli L, Hangoc G, Lee Y, Mantel C, Clapp DW, Kim CH. Transgenic expression of stromal cell-derived factor-1/CXC chemokine ligand 12 enhances myeloid progenitor cell survival/antiapoptosis in vitro in response to growth factor withdrawal and enhances myelopoiesis in vivo. J Immunol. 2003;170:421–429. doi: 10.4049/jimmunol.170.1.421. [DOI] [PubMed] [Google Scholar]
  • 83.Ivanova NB, Dimos JT, Schaniel C, Hackney JA, Moore KA, Lemischka IR. A stem cell molecular signature. Science. 2002;298:601–604. doi: 10.1126/science.1073823. [DOI] [PubMed] [Google Scholar]
  • 84.Campbell TB, Basu S, Hangoc G, Tao W, Broxmeyer HE. Overexpression of Rheb2 enhances mouse hematopoietic progenitor cell growth while impairing stem cell repopulation. Blood. 2009;114:3392–3401. doi: 10.1182/blood-2008-12-195214. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Rohrabaugh SL, Campbell TB, Hangoc G, Broxmeyer HE. Ex vivo rapamycin treatment of human cord blood CD34+ cells enhances their engraftment of NSG mice. Blood Cells Mol Dis. 2011;46:318–20. doi: 10.1016/j.bcmd.2011.02.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Liu Y, Li J, Kim BO, Pace BS, He JJ. HIV-1 Tat protein-mediated transactivation of the HIV-1 long terminal repeat promoter is potentiated by a novel nuclear Tat-interacting protein of 110 kDa, Tip110. J Biol Chem. 2002;277:23854–23863. doi: 10.1074/jbc.M200773200. [DOI] [PubMed] [Google Scholar]
  • 87.Liu Y, Kim BO, Kao C, Jung C, Dalton JT, He JJ. Tip110, the human immunodeficiency virus type 1 (HIV-1) Tat-interacting protein of 110 kDa as a negative regulator of androgen receptor (AR) transcriptional activation. J Biol Chem. 2004;279:21766–21773. doi: 10.1074/jbc.M314321200. [DOI] [PubMed] [Google Scholar]
  • 88.Bell M, Schreiner S, Damianov A, Reddy R, Bindereif A. p110, a novel human U6 snRNP protein and U4/U6 snRNP recycling factor. EMBO J. 2002;21:2724–2735. doi: 10.1093/emboj/21.11.2724. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Liu Y, Timani K, Mantel C, Fan Y, Hangoc G, Cooper S, He JJ, Broxmeyer HE. Tip110/p110nrb/SART3/p110 regulation of hematopoiesis through c-Myc. Blood. 2011;117:5643–5651. doi: 10.1182/blood-2010-12-325332. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Liu Y, Lee M-R, Timani K, He JJ, Broxmeyer HE. Tip110 maintains expression of pluripotent factors in and pluripotency of human embryonic stem cells. Stem Cells and Development. 2011 Dec 1; doi: 10.1089/scd.2011.0512. In Press. [Epub ahead of print] [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Frye RA. Phylogenetic Classification of Prokaryotic and Eukaryotic Sir2-like Proteins. Biochem Biophys Res Comm. 2000;273:793–798. doi: 10.1006/bbrc.2000.3000. [DOI] [PubMed] [Google Scholar]
  • 92.Haigis MC, Sinclair DA. Mammalian sirtuins: Biological insights and disease relevance. Annual Rev Pathol. 2010;5:253–295. doi: 10.1146/annurev.pathol.4.110807.092250. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Han M-K, Song EK, Guo Y, Ou X, Mantel C, Broxmeyer HE. SIRT1 regulates apoptosis and Nanog expression in mouse embryonic stem cells by controlling p53 subcellular localization. Cell Stem Cell. 2008;2:241–251. doi: 10.1016/j.stem.2008.01.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Ou X, Chae HD, Wang RH, Shelley WC, Cooper S, Taylor T, Kim YJ, Deng CX, Yoder MC, Broxmeyer HE. SIRT1 deficiency compromises mouse embryonic stem cell hematopoietic differentiation, and embryonic and adult hematopoiesis in the mouse. Blood. 2011;117:440–450. doi: 10.1182/blood-2010-03-273011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Leko V, Varnum-Finney B, Li H, Gu Y, Flowers D, Nourigat C, Bernstein ID, Bedalov A. SIRT1 is dispensable for function of hematopoietic stem cells in adult mice. Blood. 2011 doi: 10.1182/blood-2011–09–377077. published ahead of print January 4, 2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Chae H-D, Broxmeyer HE. SIRT1 deficiency downregulates PTEN/JNK/FOXO1 pathway to block reactive oxygen species-induced apoptosis in mouse embryonic stem cells. Stem Cells Dev. 2011;20:1277–1285. doi: 10.1089/scd.2010.0465. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Mantel C, Messina-Graham S, Broxmeyer HE. Upregulation of nascent mitochondrial biogenesis in mouse hematopoietic stem cells parallels upregulation of CD34 and loss of pluripotency: A potential strategy for reducing oxidative risk in stem cells. Cell Cycle. 2010;9:1–10. doi: 10.4161/cc.9.10.11733. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Mantel C, Messina-Graham S, Broxmeyer HE. Superoxide Flashes, ROS, and The mPTP: Potential Implications for Hematopoietic Stem Cell Function. In: Broxmeyer HE, editor. Current Opinions in Hematopoiesis. Vol. 18. 2011. pp. 208–213. [DOI] [PubMed] [Google Scholar]
  • 99.Mantel CR, Wang RH, Deng C, Broxmeyer HE. Sirt1, Notch, and stem cell “age asymmetry”. Cell Cycle. 2008;7:2821–2825. doi: 10.4161/cc.7.18.6517. [DOI] [PubMed] [Google Scholar]
  • 100.Chae HD, Lee MR, Broxmeyer HE. AICAR induces G1/S arrest and enhances erythroid differentiation in mouse embryonic stem cells by modulating p53-Nanog pathways. Stem Cells. 2012;30:140–9. doi: 10.1002/stem.778. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Waldmann T, Scholten I, Kappes F, Hu HG, Knippers R. The DEK protein-an abundant and ubiquitous constituent of mammalian chromatin. Gene. 2004;343:1–9. doi: 10.1016/j.gene.2004.08.029. [DOI] [PubMed] [Google Scholar]
  • 102.Kappes F, Waldmann T, Mathew V, Yu J, Zhang L, Khodadoust MS, Chinnaiyan AM, Luger K, Erhardt S, Schneider R, Markovitz DM. The DEK oncoprotein is a Su(var) that is essential to heterochromatin integrity. Genes Dev. 2011;25:673–678. doi: 10.1101/gad.2036411. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Mor-Vaknin N, Punturieri A, Sitwala K, Faulkner N, Legendre M, Khodadoust MS, Kappes F, Ruth JH, Koch A, Glass D, Petruzzelli L, Adams BS, Markovitz DM. The DEK Nuclear Autoantigen Is a Secreted Chemotactic Factor. Mole Cell Biol. 2006;26:9484–9496. doi: 10.1128/MCB.01030-06. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Broxmeyer HE, Kappas F, Mor-Vaknin N, Legendre M, Kinzfogl J, Cooper S, Hangoc G, Markowitz DM. DEK regulates hematopoietic stem cell engraftment and progenitor cell proliferation. Stem Cells and Development. 2011 doi: 10.1089/scd.2011.0451. In Press. [Epub ahead of print] [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Mantel C, Luo Z, Canfield J, Braun S, Deng C, Broxmeyer HE. Involvement of p21cip-1 and p27kip-1 in the molecular mechanisms of steel factor induced proliferative synergy in vitro and of p21cip-1 in the maintenance of stem/progenitor cells in vivo. Blood. 1996;88:3710–3719. [PubMed] [Google Scholar]
  • 106.Cheng T, Rodrigues N, Shen H, Yang Y, Dombkowski D, Sykes M, Scadden DT. Hematopoietic stem cell quiescence maintained by p21cip1/waf1. Science. 2000;287:1804–1808. doi: 10.1126/science.287.5459.1804. [DOI] [PubMed] [Google Scholar]
  • 107.Stier S, Cheng T, Forkert R, Lutz C, Dombkowski DM, Zhang JL, Scadden DT. Ex vivo targeting of p21Cip1/Waf1 permits relative expansion of human hematopoietic stem cells. Blood. 2003;102:1260–1266. doi: 10.1182/blood-2002-10-3053. [DOI] [PubMed] [Google Scholar]
  • 108.Shaheen M, Broxmeyer HE. Principles of Cytokine Signaling. In: Hoffman R, Benz EJ Jr, Shattil SJ, Furie B, Silberstein LE, McGlave P, Heslop H, Anastasi J, editors. Hematology: Basic Principles and Practice. 6. Elsevier Churchill Livingston; Philadelphia, PA: 2012. In Press. [Google Scholar]
  • 109.Yuan Y, Shen H, Franklin DS, Scadden DT, Cheng T. In vivo self-renewing divisions of haematopoietic stem cells are increased in the absence of the early G1-phase inhibitor, p18INK4C. Nat Cell Biol. 2004;6:436–442. doi: 10.1038/ncb1126. [DOI] [PubMed] [Google Scholar]
  • 110.Broxmeyer HE, Franklin DS, Cooper S, Hangoc G, Mantel C. Hematopoietic progenitor cell proliferation and responsiveness to cytokines/chemokines vis p18INK4C, and its functional interactions with p21CIP1/WAF1 and p27KIP1. Stem Cells and Development. 2011 In Press. [Google Scholar]
  • 111.Broxmeyer HE, Bruns H, Zhang S, Cooper S, Hangoc G, McKenzie ANJ, Dent A, Schindler U, Naeger LK, Hoey T, Kaplan MK. Th1 cells regulate hematopoietic progenitor cell homeostasis by production of oncostatin M. Immunity. 2002;16:815–825. doi: 10.1016/s1074-7613(02)00319-9. [DOI] [PubMed] [Google Scholar]
  • 112.Kaplan MH, Chang CH, Cooper S, Lee YH, Broxmeyer HE. Distinct requirements for Stat4 and Stat6 in hematopoietic progenitor cell responses to growth factors and chemokines. J Hematotherapy & Stem Cell Res. 2003;12:401–408. doi: 10.1089/152581603322286033. [DOI] [PubMed] [Google Scholar]
  • 113.Broxmeyer HE, Sehra S, Cooper S, Toney LM, Kusam S, Aloor JJ, Marchal CC, Dinauer M, Dent AL. BAZF-deficient mice have unusual alterations in hematopoietic progenitor cell activity similar to that of BCL-6 deficient mice. Mole Cell Biol. 2007;27:5275–5285. doi: 10.1128/MCB.01967-05. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Kaplan MH, Glosson NL, Stritesky GL, Yeh N, Rohrabaugh S, Kinzfogl J, Goswami R, Levy DE, Brutkiewicz RR, Blum JS, Cooper S, Hangoc G, Broxmeyer HE. STAT3-dependent IL-21 production from T cells regulates hematopoietic progenitor cell homeostasis. Blood. 2011;117:6198–6201. doi: 10.1182/blood-2011-02-334367. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Zhang CC, Kaba M, Ge G, Xie K, Tong W, Hug C, Lodish HF. Angiopoietin-like proteins stimulate ex vivo expansion of hematopoietic stem cells. Nat Med. 2006;12:240–245. doi: 10.1038/nm1342. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Zhang CC, Kaba M, Iizuka S, Huynh H, Lodish HF. Angiopoietin-like 5 and IGFBP2 stimulate ex vivo expansion of human cord blood hematopoietic stem cells as assayed by NOD/SCID transplantation. Blood. 2008;111:3415–3423. doi: 10.1182/blood-2007-11-122119. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Zheng J, Huynh H, Umikawa M, Silvany R, Zhang CC. Angiopoietin-like protein 3 supports the activity of hematopoietic stem cells in the bone marrow niche. Blood. 2011;117:470–479. doi: 10.1182/blood-2010-06-291716. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Broxmeyer HE, Srour EF, Cooper S, Wallace CT, Hangoc G, Youn BS. Angiopoietin-like-2 and -3 act through their coiled-coil domains to enhance survival and replating capacity of human cord blood hematopoietic progenitors. Blood Cells Mol Dis. 2012;48:25–29. doi: 10.1016/j.bcmd.2011.09.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Kinzfogl J, Hangoc G, Broxmeyer HE. Neurexin1α/Neurexophilin 1 function as a myelosuppressive axis in human cord blood and murine bone marrow hematopoiesis. Blood. 2011;118:565–575. doi: 10.1182/blood-2010-12-325381. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Broxmeyer HE, Cooper S. High efficiency recovery of immature hematopoietic progenitor cells with extensive proliferative capacity from human cord blood cryopreserved for ten years. Clin and Exp Immunol. 1997;107:45–53. [PubMed] [Google Scholar]
  • 121.Broxmeyer HE, Srour EF, Hangoc G, Cooper S, Anderson JA, Bodine D. High efficiency recovery of hematopoietic progenitor cells with extensive proliferative and ex vivo expansion activity and of hematopoietic stem cells with NOD/SCID mouse repopulation ability from human cord blood stored frozen for 15 years. Proc Natl Acad Sci USA. 2003;100:645–650. doi: 10.1073/pnas.0237086100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Broxmeyer HE, Lee MR, Hangoc G, Cooper S, Prasain N, Kim YJ, Mallett C, Ye Z, Witting S, Cornetta K, Cheng L, Yoder MC. Hematopoietic stem/progenitor cells, generation of induced pluripotent stem cells, and isolation of endothelial progenitors from 21- to 23.5-year cryopreserved cord blood. Blood. 2011;117:4773–4777. doi: 10.1182/blood-2011-01-330514. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES