Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 51
Filter
1.
Leukemia ; 32(7): 1670, 2018 07.
Article in English | MEDLINE | ID: mdl-29891936

ABSTRACT

At the time of publication the funding information was omitted from the article - this has now been corrected in both the HTML and the PDF.

2.
Leukemia ; 32(7): 1575-1586, 2018 07.
Article in English | MEDLINE | ID: mdl-29467489

ABSTRACT

The presence, within the human bone marrow, of cells with both endothelial and hemogenic potential has been controversial. Herein, we identify, within the human fetal bone marrow, prior to establishment of hematopoiesis, a unique APLNR+, Stro-1+ cell population, co-expressing markers of early mesodermal precursors and/or hemogenic endothelium. In adult marrow, cells expressing similar markers are also found, but at very low frequency. These adult-derived cells can be extensively culture expanded in vitro without loss of potential, they preserve a biased hemogenic transcriptional profile, and, upon in vitro induction with OCT4, assume a hematopoietic phenotype. In vivo, these cells, upon transplantation into a fetal microenvironment, contribute to the vasculature, and generate hematopoietic cells that provide multilineage repopulation upon serial transplantation. The identification of this human somatic cell population provides novel insights into human ontogenetic hematovascular potential, which could lead to a better understanding of, and new target therapies for, malignant and nonmalignant hematologic disorders.

3.
Stem Cell Reports ; 6(6): 957-969, 2016 06 14.
Article in English | MEDLINE | ID: mdl-27304918

ABSTRACT

In utero hematopoietic stem/progenitor cell transplantation (IUHSCT) has only been fully successful in the treatment of congenital immunodeficiency diseases. Using sheep as a large animal model of IUHSCT, we demonstrate that administration of CD146(+)CXCL12(+)VEGFR2(+) or CD146(+)CXCL12(+)VEGFR2(-) cells prior to, or in combination with, hematopoietic stem/progenitor cells (HSC), results in robust CXCL12 production within the fetal marrow environment, and significantly increases the levels of hematopoietic engraftment. While in the fetal recipient, donor-derived HSC were found to reside within the trabecular bone, the increased expression of VEGFR2 in the microvasculature of CD146(+)CXCL12(+)VEGFR2(+) transplanted animals enhanced levels of donor-derived hematopoietic cells in circulation. These studies provide important insights into IUHSCT biology, and demonstrate the feasibility of enhancing HSC engraftment to levels that would likely be therapeutic in many candidate diseases for IUHSCT.


Subject(s)
Blood Transfusion, Intrauterine , Bone Marrow Transplantation , Endothelial Cells/transplantation , Graft Survival , Hematopoietic Stem Cell Transplantation , Animals , Biomarkers/metabolism , CD146 Antigen/genetics , CD146 Antigen/immunology , Chemokine CXCL12/genetics , Chemokine CXCL12/immunology , Endothelial Cells/cytology , Endothelial Cells/immunology , Endovascular Procedures , Female , Fetus , Gene Expression , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/immunology , Pregnancy , Sheep, Domestic , Transplantation, Homologous , Treatment Outcome , Vascular Endothelial Growth Factor Receptor-2/genetics , Vascular Endothelial Growth Factor Receptor-2/immunology
4.
Cytotherapy ; 16(9): 1280-93, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25108653

ABSTRACT

BACKGROUND AIMS: Inadequate engraftment of hematopoietic stem cells (HSCs) after in utero HSC transplantation (IUHSCT) remains a major obstacle for the prenatal correction of numerous hereditary disorders. HSCs express CXCR4 receptors that allow homing and engraftment in response to stromal-derived factor 1 (SDF-1) ligand present in the bone marrow stromal niche. Plerixafor, a mobilization drug, works through the interruption of the CXCR4-SDF-1 axis. METHODS: We used the fetal sheep large-animal model to test our hypotheses that (i) by administering plerixafor in utero before performing IUHSCT to release fetal HSCs and thus vacating recipient HSC niches, (ii) by using human mesenchymal stromal/stem cells (MSCs) to immunomodulate and humanize the fetal BM niches and (iii) by increasing the CXCR4(+) fraction of CD34(+) HSCs, we could improve engraftment. Human cord blood-derived CD34(+) cells and human bone marrow-derived MSCs were used for these studies. RESULTS: When MSCs were transplanted 1 week before CD34(+) cells with plerixafor treatment, we observed 2.80% donor hematopoietic engraftment. Combination of this regimen with additional CD34(+) cells at the time of MSC infusion increased engraftment levels to 8.77%. Next, increasing the fraction of CXCR4(+) cells in the CD34(+) population albeit transplanting at a late gestation age was not beneficial. Our results show engraftment of both lymphoid and myeloid lineages. CONCLUSIONS: Prior MSC and HSC cotransplantation followed by manipulation of the CXCR4-SDF-1 axis in IUHSCT provides an innovative conceptual approach for conferring competitive advantage to donor HSCs. Our novel approach could provide a clinically relevant approach for enhancing engraftment early in the fetus.


Subject(s)
Genetic Diseases, Inborn/therapy , Hematopoietic Stem Cell Transplantation , Heterocyclic Compounds/administration & dosage , Mesenchymal Stem Cells/physiology , Receptors, CXCR4/administration & dosage , Animals , Antigens, CD34/metabolism , Benzylamines , Cell Differentiation , Cell Lineage , Cells, Cultured , Chemokine CXCL12/metabolism , Clinical Protocols , Cyclams , Disease Models, Animal , Female , Fetus , Graft Survival , Humans , Immunomodulation , Pregnancy , Sheep
5.
Br J Haematol ; 166(2): 268-78, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24673111

ABSTRACT

The fetal sheep model has served as a biologically relevant and translational model to study in utero haematopoietic stem cell transplantation (IUHSCT), yet little is known about the ontogeny of the bone marrow (BM) niches in this model. Because the BMmicroenvironment plays a critical role in the outcome of haematopoietic engraftment, we have established the correlation between the fetal-sheep and fetal-human BM niche ontogeny, so that studies addressing the role of niche development at the time of IUHSCT could be accurately performed. Immunofluorescence confocal microscopic analysis of sheep fetal bone from gestational days (gd) 25-68 showed that the BM microenvironment commences development with formation of the vascular niche between 25 and 36 gd in sheep; correlating with the events at 10-11 gestational weeks (gw) in humans. Subsequently, between 45 and 51 gd in sheep (c. 14 gw in humans), the osteoblastic/endosteal niche started developing, the presence of CD34(+)  CD45(+) cells were promptly detected, and their number increased with gestational age. IUHSCT, performed in sheep at 45 and 65 gd, showed significant haematopoietic engraftment only at the later time point, indicating that a fully functional BM microenvironment improved engraftment. These studies show that sheep niche ontogeny closely parallels human, validating this model for investigating niche influence/manipulation in IUHSCT engraftment.


Subject(s)
Hematopoietic Stem Cell Transplantation , Models, Animal , Stem Cell Niche/physiology , Animals , Antigens, CD34/metabolism , Bone Marrow/embryology , Bone Marrow Cells/cytology , Female , Fetal Development/physiology , Fetus/cytology , Gestational Age , Graft Survival/physiology , Heterografts , Humans , Osteoblasts/physiology , Pregnancy , Sheep
6.
World J Stem Cells ; 5(2): 43-52, 2013 Apr 26.
Article in English | MEDLINE | ID: mdl-23671718

ABSTRACT

In utero stem cell transplantation, which promises treatment for a host of genetic disorders early in gestation before disease effect stems from Ray Owen's seminal observation that self-tolerance, is acquired during gestation. To date, in utero transplantation (IUT) has proved useful in characterizing the hematopoietic stem cell. Recent observations support its use as an in vivo method to further understanding of self-tolerance. Preclinical development continues for its application as a treatment for childhood hematolymphoid diseases. In addition, IUT may offer therapeutic options in the treatment of diabetes among other diseases. Thus IUT serves as a technique or system important in both a basic and applied format. This review summarizes these findings.

7.
Exp Hematol ; 41(8): 749-758.e5, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23612043

ABSTRACT

Until now, ex vivo generation of CD34(+) hematopoietic stem cells (HSCs) from human embryonic stem cells (hESCs) mostly involved use of feeder cells of nonhuman origin. Although they provided invaluable models to study hematopoiesis, in vivo engraftment of hESC-derived HSCs remains a challenging task. In this study, we used a novel coculture system composed of human bone marrow-derived mesenchymal stromal/stem cells (MSCs) and peripheral blood CD14(+) monocyte-derived macrophages to generate CD34(+) cells from hESCs in vitro. Human ESC-derived CD34(+) cells generated using this method expressed surface makers associated with adult human HSCs and upregulated hematopoietic stem cell genes comparable to human bone marrow-derived CD34(+) cells. Finally, transplantation of purified hESC-derived CD34(+) cells into the preimmune fetal sheep, primed with transplantation of MSCs derived from the same hESC line, demonstrated multilineage hematopoietic activity with graft presence up to 16 weeks after transplantation. This in vivo demonstration of engraftment and robust multilineage hematopoietic activity by hESC-derived CD34(+) cells lends credence to the translational value and potential clinical utility of this novel differentiation and transplantation protocol.


Subject(s)
Antigens, CD34/immunology , Embryonic Stem Cells/cytology , Sheep/embryology , Animals , Base Sequence , Coculture Techniques , DNA Primers , Hematopoiesis/genetics , Humans , Macrophages/cytology , Models, Animal , Polymerase Chain Reaction
8.
PLoS One ; 8(3): e60461, 2013.
Article in English | MEDLINE | ID: mdl-23555976

ABSTRACT

Mesenchymal stem cells (MSC) preferentially migrate to damaged tissues and, due to their immunomodulatory and trophic properties, contribute to tissue repair. Although MSC express molecules, such as membrane cofactor protein (CD46), complement decay-accelerating factor (CD55), and protectin (CD59), which confer protection from complement-mediated lysis, MSC are recruited and activated by anaphylatoxins after transplantation, potentially causing MSC death and limiting therapeutic benefit. We have previously demonstrated that transduction of MSC with a retrovirus encoding HCMV-US proteins resulted in higher levels of MSC engraftment due to decreased HLA-I expression. Here, we investigate whether engineering MSC to express US2 (MSC-US2), US3 (MSC-US3), US6 (MSC-US6), or US11 (MSC-US11) HCMV proteins can alter complement recognition, thereby better protecting MSC from complement attack and lysis. HCMV-US proteins increased MSC CD59 expression at different levels as determined by flow cytometric evaluation of the median fluorescence intensity ratio (MFI). A significant increase in CD59 expression was seen in MSC-US2, MSC-US3, and MSC-US6, but not in MSC-US11. Only MSC-US2 displayed increased expression of CD46, while US2 and US3 proteins were both able to augment the percentage of MSC expressing this molecule. Regardless of the HCMV protein expressed, none changed CD55 MFI; however, expression of US6, US11, and US2 each increased the percentage of MSC that were positive for this molecule. Because US2 protein was the most efficient in up-regulating all three complement regulatory proteins, we used a functional complement-mediated cytotoxicity assay to investigate whether MSC-US2 were protected from complement-mediated lysis. We demonstrated that over-expression of the US2 protein reduced complement lysis by 59.10±12.89% when compared to untransduced MSC. This is the first report, to our knowledge, describing a role of HCMV-US proteins in complement evasion, and our data shows that over-expression of US2 protein on MSC could serve as a strategy to protect these cells from complement lysis.


Subject(s)
Complement System Proteins/immunology , Cytomegalovirus/genetics , Glycoproteins/genetics , Immediate-Early Proteins/genetics , Membrane Proteins/genetics , Mesenchymal Stem Cells/immunology , RNA-Binding Proteins/genetics , Viral Envelope Proteins/genetics , Viral Proteins/genetics , CD55 Antigens/analysis , CD55 Antigens/immunology , CD59 Antigens/analysis , CD59 Antigens/immunology , Cell Engineering/methods , Cells, Cultured , Genetic Vectors/genetics , Humans , Membrane Cofactor Protein/analysis , Membrane Cofactor Protein/immunology , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Transduction, Genetic
9.
FASEB J ; 27(6): 2111-21, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23413357

ABSTRACT

To identify human bone marrow stromal cell (BMSC) subsets with enhanced ability to engraft/contribute to the resident intestinal cellular pool, we transplanted clonally derived BMSCs into fetal sheep. Analysis at 75 d post-transplantation showed 2 of the 6 clones engrafting the intestine at 4- to 5-fold higher levels (5.03±0.089 and 5.04±0.15%, respectively) than the other clones (P<0.01), correlating with the percentage of donor-derived Musashi-1(+) (12.01-14.17 vs. 1.2-3.8%; P<0.01) or leucine-rich repeat-containing G-protein coupled receptor 5 (Lgr5)(+) cells within the intestinal stem cell (ISC) region. Phenotypic and transcriptome analysis determined that the clones with enhanced intestinal contribution expressed high levels of Ephrin type B receptor 2 (EphB2). Intestinal explants demonstrated proliferation of the engrafted cells and ability to generate crypt-like structures in vitro still expressing EphB2. Additional transplants based on BMSC EphB2 expression demonstrated that, at 7 d post-transplant, the EphB2(high) BMSCs engrafted in the ISC region at levels of 2.1 ± 0.2%, while control EphB2(low) BMSCs engrafted at 0.3 ± 0.1% (P<0.01). Therefore we identified a marker for isolating and culturing an expandable subpopulation of BMSCs with enhanced intestinal homing and contribution to the ISC region.


Subject(s)
Intestinal Mucosa/cytology , Intestinal Mucosa/metabolism , Mesenchymal Stem Cells/classification , Mesenchymal Stem Cells/metabolism , Receptor, EphB2/metabolism , Animals , Biomarkers/metabolism , Cell Lineage , Female , Fetus , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Intestinal Mucosa/embryology , Intestine, Small/cytology , Intestine, Small/embryology , Intestine, Small/metabolism , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/cytology , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Pregnancy , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Receptor, EphB2/genetics , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Sheep , Stem Cell Niche , Transcriptome , Transplantation, Heterologous
10.
J Cell Physiol ; 228(5): 1010-6, 2013 May.
Article in English | MEDLINE | ID: mdl-23042590

ABSTRACT

Besides the liver, it has been difficult to identify which organ(s) and/or cellular component(s) contribute significantly to the production of human FVIII:c (FVIII). Thus far, only endothelial cells have been shown to constitute a robust extrahepatic source of FVIII, possibly explaining both the diverse presence of FVIII mRNA in the body, and the observed increase in FVIII levels during liver failure. Here, we investigate whether human mesenchymal stem cells (MSC), ubiquitously present in different organs, could also contribute to FVIII production. MSC isolated from human lung, liver, brain, and bone marrow expressed FVIII message as determined by quantitative-RT-PCR. Using an antibody specific for FVIII, confocal microscopy, and umbilical cord-derived endothelial cells (HUVEC) as a negative control, we demonstrated that, in MSC, FVIII protein was not stored in granules; rather, it localized to the perinuclear region. Furthermore, functional FVIII was detected in MSC supernatants and cell lysates by aPTT and chromogenic assays. These results demonstrate that MSC can contribute at low levels to the functional FVIII pool, and advance the understanding of the physiology of FVIII production and secretion.


Subject(s)
Factor VIII/biosynthesis , Factor VIII/metabolism , Mesenchymal Stem Cells , Cell Differentiation , Endothelial Cells/cytology , Endothelial Cells/metabolism , Human Umbilical Vein Endothelial Cells , Humans , Mesenchymal Stem Cells/metabolism , Microscopy, Confocal , RNA, Messenger/metabolism , Secretory Vesicles/metabolism , Tissue Distribution
11.
PLoS One ; 7(5): e36163, 2012.
Article in English | MEDLINE | ID: mdl-22666319

ABSTRACT

BACKGROUND: Mesenchymal stem cells (MSC) are promising candidates for cell therapy, as they migrate to areas of injury, differentiate into a broad range of specialized cells, and have immunomodulatory properties. However, MSC are not invisible to the recipient's immune system, and upon in vivo administration, allogeneic MSC are able to trigger immune responses, resulting in rejection of the transplanted cells, precluding their full therapeutic potential. Human cytomegalovirus (HCMV) has developed several strategies to evade cytotoxic T lymphocyte (CTL) and Natural Killer (NK) cell recognition. Our goal is to exploit HCMV immunological evasion strategies to reduce MSC immunogenicity. METHODOLOGY/PRINCIPAL FINDINGS: We genetically engineered human MSC to express HCMV proteins known to downregulate HLA-I expression, and investigated whether modified MSC were protected from CTL and NK attack. Flow cytometric analysis showed that amongst the US proteins tested, US6 and US11 efficiently reduced MSC HLA-I expression, and mixed lymphocyte reaction demonstrated a corresponding decrease in human and sheep mononuclear cell proliferation. NK killing assays showed that the decrease in HLA-I expression did not result in increased NK cytotoxicity, and that at certain NK∶MSC ratios, US11 conferred protection from NK cytotoxic effects. Transplantation of MSC-US6 or MSC-US11 into pre-immune fetal sheep resulted in increased liver engraftment when compared to control MSC, as demonstrated by qPCR and immunofluorescence analyses. CONCLUSIONS AND SIGNIFICANCE: These data demonstrate that engineering MSC to express US6 and US11 can be used as a means of decreasing recognition of MSC by the immune system, allowing higher levels of engraftment in an allogeneic transplantation setting. Since one of the major factors responsible for the failure of allogeneic-donor MSC to engraft is the mismatch of HLA-I molecules between the donor and the recipient, MSC-US6 and MSC-US11 could constitute an off-the-shelf product to overcome donor-recipient HLA-I mismatch.


Subject(s)
Cytomegalovirus/genetics , Genetic Engineering , Mesenchymal Stem Cells/immunology , Mesenchymal Stem Cells/metabolism , Viral Proteins/genetics , Animals , Cell Differentiation , Cell Line , Cell Proliferation , Down-Regulation/immunology , Gene Expression , HLA Antigens/metabolism , Humans , Kanamycin Kinase/immunology , Killer Cells, Natural/immunology , Liver/cytology , Liver/surgery , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/cytology , Sheep , T-Lymphocytes, Cytotoxic/cytology , T-Lymphocytes, Cytotoxic/immunology
12.
Hepatology ; 56(3): 1086-96, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22488442

ABSTRACT

UNLABELLED: Although the vasculogenic potential of circulating and cord blood (CB)-derived endothelial colony-forming cells (ECFC) has been demonstrated in vitro and in vivo, little is known about the inherent biologic ability of these cells to home to different organs and contribute to tissue-specific cell populations. Here we used a fetal sheep model of in utero transplantation to investigate and compare the intrinsic ability of human CB-derived ECFC to migrate to the liver and to the intestine, and to define ECFC's intrinsic ability to integrate and contribute to the cytoarchitecture of these same organs. ECFCs were transplanted by an intraperitoneal or intrahepatic route (IH) into fetal sheep at concentrations ranging from 1.1-2.6 × 10(6) cells/fetus. Recipients were evaluated at 85 days posttransplant for donor (human) cells using flow cytometry and confocal microscopy. We found that, regardless of the route of injection, and despite the IH delivery of ECFC, the overall liver engraftment was low, but a significant percentage of cells were located in the perivascular regions and retained the expression of hallmark endothelial makers. By contrast, ECFC migrated preferentially to the intestinal crypt region and contributed significantly to the myofibroblast population. Furthermore, ECFC expressing CD133 and CD117 lodged in areas where endogenous cells expressed those same phenotypes. CONCLUSION: ECFC inherently constitute a potential source of cells for the treatment of intestinal diseases, but strategies to increase the numbers of ECFC persisting within the hepatic parenchyma are needed in order to enhance ECFC therapeutic potential for this organ.


Subject(s)
Cell Movement , Endothelial Cells/physiology , Fetal Blood , Intestines/cytology , Liver/cytology , Animals , Endothelial Cells/transplantation , Humans , Sheep
13.
Exp Hematol ; 39(12): 1124-1135.e4, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21906573

ABSTRACT

We recently re-established a line of sheep that accurately mimics the clinical symptoms and genetics of severe hemophilia A (HA). Here, we tested a novel, nonablative transplantation therapy in two pediatric HA animals. Paternal mesenchymal stem cells (MSC) were transduced with a porcine FVIII-encoding lentivector and transplanted via the intraperitoneal route without preconditioning. At the time of transplantation, these animals had received multiple human FVIII treatments for various spontaneous bleeds and had developed debilitating hemarthroses, which produced severe defects in posture and gait. Transplantation of transduced MSC resolved all existent hemarthroses, and spontaneous bleeds ceased. Damaged joints recovered fully; the animals regained normal posture and gait and resumed normal activity. Despite achieving factor-independence, a sharp rise in pre-existent Bethesda titers occurred following transplantation, decreasing the effectiveness and duration of therapy. Postmortem examination revealed widespread engraftment, with MSC present within the lung, liver, intestine, and thymus, but particularly within joints affected at the time of transplantation, suggesting MSC homed to sites of ongoing injury/inflammation to release FVIII, explaining the dramatic improvement in hemarthrotic joints. In summary, this novel, nonablative MSC transplantation was straightforward, safe, and converted life-threatening, debilitating HA to a moderate phenotype in a large animal model.


Subject(s)
Disease Models, Animal , Factor VIII/genetics , Hemophilia A/surgery , Mesenchymal Stem Cell Transplantation , Sheep/genetics , Animals , Cell Lineage , Cell Movement , Factor VIII/immunology , Female , Genetic Vectors/genetics , Graft Survival , Hemarthrosis/etiology , Hemarthrosis/pathology , Hemophilia A/complications , Hemophilia A/drug therapy , Hemorrhage/etiology , Humans , Injections, Intraperitoneal , Isoantibodies/biosynthesis , Isoantibodies/immunology , Male , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/virology , Phenotype , Recombinant Proteins/therapeutic use , Remission Induction , Sheep/blood , Sus scrofa/genetics , Tissue Distribution
14.
Exp Hematol ; 38(12): 1125, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20965227
15.
Exp Hematol ; 38(7): 574-80, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20417684

ABSTRACT

Development of new approaches to treat patients with hepatic diseases that can eliminate the need for liver transplantation is imperative. Use of cell therapy as a means of repopulating the liver has several advantages over whole-organ transplantation because it would be less invasive, less immunogenic, and would allow the use, in some instances, of autologous-derived cells. Stem/progenitor cells that would be ideal for liver repopulation would need to have characteristics such as availability and ease of isolation, the ability to be expanded in vitro, ensuring adequate numbers of cells, susceptibility to modification by viral vector transduction/genetic recombination, to correct any underlying genetic defects, and the ability of restoring liver function following transplantation. Bone marrow-derived stem cells, such as hematopoietic, mesenchymal and endothelial progenitor cells possess some or most of these characteristics, making them ideal candidates for liver regenerative therapies. Here, we will summarize the ability of each of these stem cell populations to give rise to functional hepatic elements that could mediate repair in patients with liver damage/disease.


Subject(s)
Bone Marrow Cells/cytology , Liver Diseases/therapy , Liver Regeneration , Stem Cell Transplantation , Stem Cells/cytology , Humans
16.
Exp Hematol ; 38(6): 516-525.e4, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20227460

ABSTRACT

OBJECTIVE: CD34(+) cells, present within the bone marrow, have previously been shown to possess pancreatic endocrine potential. Based on this observation, we explored the capacity of CD34(+) cells derived in culture from the differentiation of human embryonic stem cells (hESC), for their in vivo pancreatic endocrine capacity. MATERIALS AND METHODS: Sheep were transplanted with hESC-derived CD34(+) cells, as well as nonsorted differentiated cultures. Transplantations were carried out with in utero intraperitoneal injections prior to development of the immune system in the fetus so that tolerance toward foreign antigens was acquired during gestation and persisted in the adult. RESULTS: All cell populations that were tested demonstrated human cellular activity and long-term presence up to 5 years. However, the in vivo beta-cell-like activity achieved from the transplantation of the sorted CD34(+) cell population was not augmented by transplanting the entire cell population from which the CD34(+) cells were isolated. Human DNA and insulin messenger RNA were detected in sheep pancreases. An average of 1.51 ng/mL human C-peptide was detected in serum from eight animals transplanted with differentiated cell populations and assayed up to 55 months posttransplantation. Transplantation of as few as 23,500 cells resulted in long-term sustainable beta-cell-like activity. Teratomas were absent in the transplanted animals. CONCLUSION: Our data suggest that hESC-derived CD34(+) cells have a potential for long-term in vivo endocrine cellular activity that could prove useful in regenerative medicine. Because the same cell population has previously been shown to contain hematopoietic potential, it could be used for the induction of immunological tolerance and bone marrow chimerism prior to cellular therapy for diabetes.


Subject(s)
Antigens, CD34/immunology , Cell Differentiation , Embryonic Stem Cells/immunology , Islets of Langerhans/cytology , Animals , Base Sequence , Blood Glucose/analysis , DNA/analysis , DNA Primers , Embryonic Stem Cells/cytology , Enzyme-Linked Immunosorbent Assay , Humans , Immunohistochemistry , Insulin/genetics , Islets of Langerhans/immunology , RNA, Messenger/analysis , Reverse Transcriptase Polymerase Chain Reaction , Sheep
17.
Exp Hematol ; 38(4): 311-20, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20170708

ABSTRACT

OBJECTIVE: To determine if mesenchymal stem cells (MSC) derived from human fetal pancreatic tissue (pMSC) would engraft and differentiate in sheep pancreas following transplantation in utero. MATERIALS AND METHODS: A three-step culture system was established for generating human fetal pMSC. Sheep fetuses were transplanted during the fetal transplant receptivity period with human pMSC and evaluated for in situ and functional engraftment in their pancreas, liver, and bone marrow. RESULTS: Isolation and expansion of adherent cells from the human fetal pancreas yielded a cell population with morphologic and phenotypic characteristics similar to MSC derived from bone marrow. This putative stem cell population could undergo multilineage differentiation in vitro. Three to 27 months after fetal transplantation, the pancreatic engraftment frequency (chimeric index) was 79%, while functional engraftment was noted in 50% of transplanted sheep. Hepatic and marrow engraftment and expression was noted as well. CONCLUSION: We have established a procedure for isolation of human fetal pMSC that display characteristics similar to bone marrow-derived MSC. In vivo results suggest the pMSC engraft, differentiate, and secrete human insulin from the sheep pancreas.


Subject(s)
Insulin/blood , Islets of Langerhans/metabolism , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/cytology , Animals , Cell Differentiation , Cells, Cultured , Female , Flow Cytometry , Humans , Immunohistochemistry , Insulin/metabolism , Pregnancy , Reference Standards , Reverse Transcriptase Polymerase Chain Reaction , Sheep
18.
Stem Cell Res ; 2(2): 125-38, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19383418

ABSTRACT

Human mesenchymal stem cells (MSC) hold great promise for cellular replacement therapies. Despite their contributing to phenotypically distinct cells in multiple tissues, controversy remains regarding whether the phenotype switch results from a true differentiation process. Here, we studied the events occurring during the first 120 h after human MSC transplantation into a large animal model. We demonstrate that MSC, shortly after engrafting different tissues, undergo proliferation and rapidly initiate the differentiative process, changing their phenotype into tissue-specific cells. Thus, the final level of tissue-specific cell contribution is not determined solely by the initial level of engraftment of the MSC within that organ, but rather by the proliferative capability of the ensuing tissue-specific cells into which the MSC rapidly differentiate. Furthermore, we show that true differentiation, and not cell fusion or transfer of mitochondria or membrane-derived vesicles between transplanted and resident cells, is the primary mechanism contributing to the change of phenotype of MSC upon transplantation.


Subject(s)
Cell Differentiation , Graft Survival , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/cytology , Animals , Cell Fusion , Cell Proliferation , Cell-Derived Microparticles , Humans , Mitochondria , Sheep , Transplantation, Heterologous
19.
J Cell Physiol ; 220(1): 102-11, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19277981

ABSTRACT

Human hematopoietic stem/progenitor cells (HSC) isolated based upon specific patterns of CD34 and CD38 expression, despite phenotypically identical, were found to be functionally heterogeneous, raising the possibility that reversible expression of these antigens may occur during cellular activation and/or proliferation. In these studies, we combined PKH67 tracking with CD34/CD38 immunostaining to compare cell division kinetics between human bone marrow (BM) and cord blood (CB)-derived HSC expanded in a serum-free/stromal-based system for 14 days (d), and correlated CD34 and CD38 expression with the cell divisional history. CB cells began dividing 24 h earlier than BM cells, and significantly higher numbers underwent mitosis during the time in culture. By d10, over 55% of the CB-cells reached the ninth generation, whereas BM-cells were mostly distributed between the fifth and seventh generation. By d14, all CB cells had undergone multiple cell divisions, while 0.7-3.8% of BM CD34(+) cells remained quiescent. Furthermore, the percentage of BM cells expressing CD34 decreased from 60.8 +/- 6.3% to 30.6 +/- 6.7% prior to initiating division, suggesting that downmodulation of this antigen occurred before commencement of proliferation. Moreover, with BM, all primitive CD34(+)CD38(-) cells present at the end of culture arose from proliferating CD34(+)CD38(+) cells that downregulated CD38 expression, while in CB, a CD34(+)CD38(-) population was maintained throughout culture. These studies show that BM and CB cells differ significantly in cell division kinetics and expression of CD34 and CD38, and that the inherent modulation of these antigens during ex vivo expansion may lead to erroneous quantification of the stem cell content of the expanded graft.


Subject(s)
Bone Marrow Cells/physiology , Cell Proliferation , Fetal Blood/cytology , Hematopoietic Stem Cells/physiology , Mesenchymal Stem Cells/physiology , ADP-ribosyl Cyclase 1/metabolism , Antigens, CD34/metabolism , Biomarkers/metabolism , Bone Marrow Cells/immunology , Cell Separation/methods , Cells, Cultured , Flow Cytometry , Fluorescent Dyes , Hematopoietic Stem Cells/immunology , Humans , Immunophenotyping , Kinetics , Membrane Glycoproteins/metabolism , Mesenchymal Stem Cells/immunology , Mitosis , Organic Chemicals , Phenotype , Time Factors
20.
Cancer Biol Ther ; 8(4): 331-7, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19197149

ABSTRACT

Acute myeloid leukemia (AML) is the most frequently diagnosed adulthood leukemia, yet current therapies offer a cure rate of less than 30%. This may be due in part to the fact that the leukemia-initiating cells in AML reside within the rare and highly primitive CD34(+)CD38(-) hematopoietic stem/progenitor cell (HSC) population that are often resistant to chemotherapy. Docosahexanoic acid (DHA), a major component of fish oil, has previously been shown to inhibit the induction and progression of breast, prostate and colon cancer, and increase the therapeutic effects of numerous chemotherapeutics, often by enhancing apoptosis. In the present studies, we investigated DHA's effect on the primitive and undifferentiated AML cell line KG1a, to explore the potential of this fatty acid to serve as adjuvant therapy for AML. Treatment of KG1a cells with DHA for 96 hours did not lead to maturation or cell cycle modification when compared to an untreated KG1a control (n = 4). However, DHA treatment of KG1a cells resulted in a progressive loss of viability, DNA fragmentation, and an increase in Annexin V expression, demonstrating DHA-induced apoptosis (n = 4). Moreover, expression of the pro-apoptotic protein Bax was increased, with resultant skewing in the Bax/bcl-2 ratio, providing a mechanistic explanation for the observed DHA-induced increase in apoptosis. Since we also show that DHA does not have a detrimental effect on normal hematopoiesis our results suggest that DHA could potentially serve as an well-tolerated adjuvant in the treatment of AML patients.


Subject(s)
Apoptosis/drug effects , Docosahexaenoic Acids/administration & dosage , Leukemia, Myeloid, Acute/drug therapy , Annexin A5/biosynthesis , Caspase 3/biosynthesis , Cell Differentiation/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Chemotherapy, Adjuvant , DNA Fragmentation/drug effects , Dose-Response Relationship, Drug , Hematopoiesis/drug effects , Humans , bcl-2-Associated X Protein/biosynthesis
SELECTION OF CITATIONS
SEARCH DETAIL
...