Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Arterioscler Thromb Vasc Biol ; 37(11): 2007-2013, 2017 11.
Article in English | MEDLINE | ID: mdl-28982668

ABSTRACT

Platelets, derived from megakaryocytes, are anucleate cytoplasmic discs that circulate in the blood stream and play major roles in hemostasis, inflammation, and vascular biology. Platelet transfusions are used in a variety of medical settings to prevent life-threatening thrombocytopenia because of cancer therapy, other causes of acquired or inherited thrombocytopenia, and trauma. Currently, platelets used for transfusion purposes are donor derived. However, there is a drive to generate nondonor sources of platelets to help supplement donor-derived platelets. Efforts have been made by many laboratories to generate in vitro platelets and optimize their production and quality. In vitro-derived platelets have the potential to be a safer, more uniform product, and genetic manipulation could allow for better treatment of patients who become refractory to donor-derived units. This review focuses on potential clinical applications of in vitro-derived megakaryocytes and platelets, current methods to generate and expand megakaryocytes from pluripotent stem cell sources, and the use of these cells for disease modeling.


Subject(s)
Blood Platelets/physiology , Induced Pluripotent Stem Cells/physiology , Megakaryocytes/physiology , Thrombopoiesis , Blood Platelets/metabolism , Cell Line , Gene Expression Regulation, Developmental , Genetic Predisposition to Disease , Hematologic Diseases/blood , Hematologic Diseases/genetics , Hematologic Diseases/therapy , Humans , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/transplantation , Megakaryocytes/metabolism , Megakaryocytes/transplantation , Phenotype , Platelet Transfusion
2.
Curr Opin Hematol ; 24(6): 565-571, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28985194

ABSTRACT

PURPOSE OF REVIEW: Donor-derived platelets have proven to be of hemostatic value in many clinical settings. There is a fear that the need for platelets may outgrow the donor pool in first-world countries. Moreover, there are other challenges with donor platelets that add to the impetus to find an alternative platelet source, especially after the megakaryocyte cytokine thrombopoietin was identified. Megakaryocytes have since been differentiated from numerous cell sources and the observed released platelet-like particles (PLPs) have led to calls to develop such products for clinical use. The development of megakaryocytes from embryonic stem cell also supported the concept of developing nondonor-based platelets. RECENT FINDINGS: Several groups have claimed that nondonor-based platelets derived from in-vitro grown megakaryocytes may soon become available to supplement or replace donor-derived products, but their number and quality has been wanting. A possible alternative of directly infusing megakaryocytes that release platelets in the lungs - similar to that recently shown for endogenous megakaryocytes - has been proposed. SUMMARY: This present review will describe the present state-of-the-art in generating and delivering nondonor-derived platelets. Progress has been slow, but advances in our ability to generate human megakaryocytes in culture, generate PLPs from these cells, and test the functionality of the resultant platelets in vitro and in vivo have identified important remaining challenges and raised alternative potential solutions.


Subject(s)
Blood Component Transfusion , Blood Platelets , Cell Culture Techniques/methods , Human Embryonic Stem Cells , Megakaryocytes , Blood Platelets/cytology , Blood Platelets/metabolism , Human Embryonic Stem Cells/cytology , Human Embryonic Stem Cells/metabolism , Humans , Megakaryocytes/cytology , Megakaryocytes/transplantation
3.
Blood ; 125(23): 3627-36, 2015 Jun 04.
Article in English | MEDLINE | ID: mdl-25852052

ABSTRACT

Thrombopoiesis is the process by which megakaryocytes release platelets that circulate as uniform small, disc-shaped anucleate cytoplasmic fragments with critical roles in hemostasis and related biology. The exact mechanism of thrombopoiesis and the maturation pathways of platelets released into the circulation remain incompletely understood. We showed that ex vivo-generated murine megakaryocytes infused into mice release platelets within the pulmonary vasculature. Here we now show that infused human megakaryocytes also release platelets within the lungs of recipient mice. In addition, we observed a population of platelet-like particles (PLPs) in the infusate, which include platelets released during ex vivo growth conditions. By comparing these 2 platelet populations to human donor platelets, we found marked differences: platelets derived from infused megakaryocytes closely resembled infused donor platelets in morphology, size, and function. On the other hand, the PLP was a mixture of nonplatelet cellular fragments and nonuniform-sized, preactivated platelets mostly lacking surface CD42b that were rapidly cleared by macrophages. These data raise a cautionary note for the clinical use of human platelets released under standard ex vivo conditions. In contrast, human platelets released by intrapulmonary-entrapped megakaryocytes appear more physiologic in nature and nearly comparable to donor platelets for clinical application.


Subject(s)
Blood Platelets , Macrophages , Megakaryocytes , Animals , Blood Platelets/metabolism , Blood Platelets/pathology , Cell Line , Humans , Macrophages/metabolism , Macrophages/pathology , Megakaryocytes/metabolism , Megakaryocytes/pathology , Megakaryocytes/transplantation , Mice , Platelet Glycoprotein GPIb-IX Complex/metabolism , Thrombopoiesis
5.
Cytotherapy ; 13(1): 114-28, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20230224

ABSTRACT

BACKGROUND AIMS: Ex vivo generation of megakaryocytes (MK) from hematopoietic stem cells (HSC) is important for both basic research, to understand the mechanism of platelet biogenesis, and clinical infusions, for rapid platelet recovery in thrombocytopenic patients. We investigated the role of two nutraceuticals, docosahexanoic acid (DHA) and arachidonic acid (AA), in the in vitro generation of MK. METHODS: Umbilical cord blood (UCB)-derived CD34+cells were cultured with stem cell factor (SCF) and thrombopoietin (TPO) in the presence (test) or absence (control) of the two additives. On day 10, MK and platelets generated were quantitated by morphologic, phenotypic and functional assays. RESULTS: The cell yield of MK and platelet numbers were significantly higher in test compared with control cells. Phenotypic analyzes and gene expression profiles confirmed these findings. Functional properties, such as colony-forming unit (CFU)-MK formation, chemotaxis and platelet activation, were found to be enhanced in cells cultured with nutraceuticals. The engraftment potential of ex vivo-expanded cells was studied in NOD/SCID mice. Mice that received MK cultured in the presence of DHA/AA engrafted better. There was a reduction in apoptosis and total reactive oxygen species (ROS) levels in the CD41(+) compartment of the test compared with control sets. The data suggest that these compounds probably exert their beneficial effect by modulating apoptotic and redox pathways. CONCLUSIONS: Use of nutraceuticals like DHA and AA may prove to be a useful strategy for efficient generation of MK and platelets from cord blood cells, for future use in clinics and basic research.


Subject(s)
Antigens, CD34/metabolism , Arachidonic Acid/pharmacology , Culture Media/chemistry , Cytokines/pharmacology , Docosahexaenoic Acids/pharmacology , Fetal Blood/cytology , Megakaryocytes/cytology , Animals , Apoptosis/drug effects , Blood Platelets/cytology , Blood Platelets/drug effects , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Chemotaxis/drug effects , Colony-Forming Units Assay , Dietary Supplements , Humans , Megakaryocytes/transplantation , Mice , Mice, SCID , Phenotype , Platelet Activation/drug effects , Ploidies , Reactive Oxygen Species/metabolism , Transcription Factors/metabolism
6.
Blood Adv ; 3(9): 1368-1378, 2019 05 14.
Article in English | MEDLINE | ID: mdl-31036722

ABSTRACT

B-domainless factor VIII (FVIII) ectopically expressed in megakaryocytes (MKs) is stored in α granules of platelets (pFVIII) and is capable of restoring hemostasis in FVIIInull mice, even in the presence of circulating inhibitors. However, our prior studies have shown that this ectopically expressed pFVIII can injure developing MKs. Moreover, the known risks of prolonged thrombocytopenia after bone marrow transplantation are significant challenges to the use of this strategy to treat individuals with severe hemophilia A and particularly those with intractable clinically relevant inhibitors. Because of these limitations, we now propose the alternative therapeutic pFVIII strategy of infusing pFVIII-expressing MKs or platelets derived from induced pluripotent stem cells (iPSCs). pFVIII-expressing iPSC-derived MKs, termed iMKs, release platelets that can contribute to improved hemostasis in problematic inhibitor patients with hemophilia A. As proof of principle, we demonstrate that hemostasis can be achieved in vitro and in vivo with pFVIII-expressing platelets and show prolonged efficacy. Notably, pFVIII-expressing platelets are also effective in the presence of inhibitors, and their effect was enhanced with recombinant FVIIa. Human pFVIII-expressing iMKs improved hemostasis in vitro, and derived platelets from infused human pFVIII-expressing iMKs improved hemostasis in FVIIInull mice. These studies indicate the potential therapeutic use of recurrent pFVIII-expressing MK or platelet infusions with prolonged hemostatic coverage that may be additive with bypassing agents in hemophilia A patients with neutralizing inhibitors.


Subject(s)
Factor VIII/genetics , Hemophilia A/therapy , Megakaryocytes/transplantation , Platelet Transfusion , Animals , Area Under Curve , Blood Platelets/cytology , Blood Platelets/metabolism , Factor VIII/analysis , Factor VIII/metabolism , Factor VIIa/therapeutic use , Hemophilia A/mortality , Humans , Male , Megakaryocytes/cytology , Megakaryocytes/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , ROC Curve , Survival Rate , Treatment Outcome
7.
J Control Release ; 228: 38-47, 2016 Apr 28.
Article in English | MEDLINE | ID: mdl-26921521

ABSTRACT

Mounting experimental evidence demonstrates that platelets support cancer metastasis. Within the circulatory system, platelets guard circulating tumor cells (CTCs) from immune elimination and promote their arrest at the endothelium, supporting CTC extravasation into secondary sites. Neutralization of CTCs in blood circulation can potentially attenuate metastases to distant organs. Therefore, extensive studies have explored the blockade of platelet-CTC interactions as an anti-metastatic strategy. Such an intervention approach, however, may cause bleeding disorders since the platelet-CTC interactions inherently rely on the blood coagulation cascade including platelet activation. On the other hand, platelets have been genetically engineered to correct inherited bleeding disorders in both animal models and human clinical trials. In this study, inspired by the physical association between platelets and CTCs, platelets were genetically modified to express surface-bound tumor necrosis factor-related apoptosis-inducing ligand (TRAIL), a cytokine known to induce apoptosis specifically in tumor cells. The TRAIL-expressing platelets were demonstrated to kill cancer cells in vitro and significantly reduce metastases in a mouse model of prostate cancer metastasis. Our results suggest that using platelets to produce and deliver cancer-specific therapeutics can provide a Trojan-horse strategy of neutralizing CTCs to attenuate metastasis.


Subject(s)
Blood Platelets/pathology , Genetic Engineering/methods , Neoplasm Metastasis/therapy , Neoplastic Cells, Circulating/pathology , Prostatic Neoplasms/therapy , TNF-Related Apoptosis-Inducing Ligand/genetics , Animals , Blood Platelets/cytology , Blood Platelets/metabolism , Cell Differentiation , Cell Line, Tumor , Cells, Cultured , Gene Expression , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Humans , Male , Megakaryocytes/cytology , Megakaryocytes/metabolism , Megakaryocytes/transplantation , Mice, Inbred C57BL , Neoplasm Metastasis/genetics , Neoplasm Metastasis/pathology , Prostate/pathology , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Transduction, Genetic/methods
8.
Exp Hematol ; 32(9): 877-80, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15345290

ABSTRACT

Assessment of donor chimerism is becoming increasingly important in patients undergoing reduced-intensity conditioning (RIC) allogeneic bone marrow transplants, due to the possibility of mixed chimeras. This regimen has been used successfully for patients with leukemia and genetic disorders with donor chimerism occurring in the myeloid, lymphoid, and/or erythroid lineages. Less toxic RIC expands the potential application of stem cell transplants to patients with nonmalignant disorders of hematopoiesis, such as the severe form of Glanzmann thrombasthenia, who previously were not considered suitable candidates based on risk-benefit analysis. To assess megakaryocyte/platelet chimerism after stem cell transplantation conducted with RIC, we used restriction fragment length polymorphism (RFLP) and sequence analyses of the HPA-3 polymorphism in the megakaryocyte/platelet-specific glycoprotein alphaIIb. In this study we show that at 23 weeks post-RIC, a leukemia patient acquired the HPA-3 donor phenotype at the DNA and platelet RNA levels.


Subject(s)
Antigens, Human Platelet/genetics , Hematopoietic Stem Cell Transplantation , Megakaryocytes/transplantation , Transplantation Chimera , Transplantation Conditioning , Cell Lineage , Humans , Male , Middle Aged , Polymorphism, Restriction Fragment Length , Transplantation, Homologous
9.
Exp Hematol ; 27(2): 293-301, 1999 Feb.
Article in English | MEDLINE | ID: mdl-10029169

ABSTRACT

Thrombocytopenia remains an important problem for patients post high-dose chemotherapy and hematopoietic stem cell transplantation. The study of megakaryocytes, the direct precursors of platelets, has been hampered by their relatively low frequency in hematopoietic tissues. In an attempt to obtain a large number of functional megakaryocytic cells, we established a serum-free culture system to grow megakaryocytic progenitor cells derived from normal human bone marrow (BM) and cord blood (CB). Highly purified (purity >95%) CD34+ cells were obtained using magnetic cell sorting (MACS) followed by fluorescence activated cell sorting (FACS). The cells were cultured in a serum-free culture system for 3 weeks in the presence of a single dose of MGDF (50 ng/mL). On days 0, 5, 8, 12, 14, 18, and 21 of culture, the cellularity and morphology were examined. Megakaryocytic cells were monitored by detecting the expression of GPIIIa (CD61), GPIIb/IIIa (CD41) and GPIb (CD42b), and the distribution of megakaryocyte (MK) ploidy was analyzed by two-color flow cytometry. MGDF alone induced maximal nucleated cell expansion at day 14, resulting in a 38.20+/-10.47-fold increase in cell number for CB and a 5.08+/-1.30-fold increase in cell number for BM. On day 14 of the culture, the percentage of CD41-/CD14- cells derived from CB reached 73.54%+/-6.01% giving an absolute number of CD41+/CD14- cells of 27.25+/-2.23 x 10(4)/mL (27,250-fold increase), whilst the percentage of CD41+/CD14- cells derived from BM was only 29.21%+/-5.63% with an absolute number of 1.36+/-0.26 x 10(4)/mL (680-fold increase). Increased expression of GPIIIa occurred the earliest in culture, followed by GPIIb/IIIa, and then GPIb. The majority (81.6%-92.6%) of megakaryocytes (CD41+ cells) on day 14 of culture were 2N, although we did detect some 4N, 8N and greater ploidy cells. In conclusion, CD34+ cells stimulated by MGDF alone generated highly enriched MK progenitor cells at day 14 of serum-free culture. CB stem and progenitor cells have a greater proliferative response to MGDF alone than those derived from BM and may, therefore, prove to be a better source of cells for MK expansion.


Subject(s)
Hematopoietic Stem Cell Mobilization , Hematopoietic Stem Cells/cytology , Megakaryocytes/cytology , Thrombocytopenia/therapy , Cell Differentiation , Female , Fetal Blood/cytology , Humans , Megakaryocytes/transplantation , Pregnancy
10.
Stem Cells Transl Med ; 4(4): 309-19, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25713465

ABSTRACT

Megakaryocytes (MKs) are rare hematopoietic cells in the adult bone marrow and produce platelets that are critical to vascular hemostasis and wound healing. Ex vivo generation of MKs from human induced pluripotent stem cells (hiPSCs) provides a renewable cell source of platelets for treating thrombocytopenic patients and allows a better understanding of MK/platelet biology. The key requirements in this approach include developing a robust and consistent method to produce functional progeny cells, such as MKs from hiPSCs, and minimizing the risk and variation from the animal-derived products in cell cultures. In this study, we developed an efficient system to generate MKs from hiPSCs under a feeder-free and xeno-free condition, in which all animal-derived products were eliminated. Several crucial reagents were evaluated and replaced with Food and Drug Administration-approved pharmacological reagents, including romiplostim (Nplate, a thrombopoietin analog), oprelvekin (recombinant interleukin-11), and Plasbumin (human albumin). We used this method to induce MK generation from hiPSCs derived from 23 individuals in two steps: generation of CD34(+)CD45(+) hematopoietic progenitor cells (HPCs) for 14 days; and generation and expansion of CD41(+)CD42a(+) MKs from HPCs for an additional 5 days. After 19 days, we observed abundant CD41(+)CD42a(+) MKs that also expressed the MK markers CD42b and CD61 and displayed polyploidy (≥16% of derived cells with DNA contents >4N). Transcriptome analysis by RNA sequencing revealed that megakaryocytic-related genes were highly expressed. Additional maturation and investigation of hiPSC-derived MKs should provide insights into MK biology and lead to the generation of large numbers of platelets ex vivo.


Subject(s)
Albumins/administration & dosage , Induced Pluripotent Stem Cells/drug effects , Megakaryocytes/drug effects , Receptors, Fc/administration & dosage , Recombinant Fusion Proteins/administration & dosage , Thrombocytopenia/therapy , Thrombopoietin/administration & dosage , Blood Platelets/drug effects , Cell Differentiation , Humans , Induced Pluripotent Stem Cells/transplantation , Megakaryocytes/transplantation , Thrombocytopenia/pathology , Transcriptome/genetics , United States , United States Food and Drug Administration , Wound Healing/drug effects
11.
Haematologica ; 89(5): 630-1, 2004 May.
Article in English | MEDLINE | ID: mdl-15136238

ABSTRACT

The additional transplantation of ex vivo generated hematopoietic (post)-progenitor cells represents a possible approach to ameliorate high-dose chemotherapy induced cytopenia. We investigated the feasibility of the large-scale expansion and transplantation of autologous megakaryocytic cells in four patients with advanced solid tumors. Up to 1,460x10(6) ex vivo generated cells were administered without adverse effects but no clear cut effect on platelet recovery was observed.


Subject(s)
Antineoplastic Agents/administration & dosage , Megakaryocytes/transplantation , Antigens, CD34/blood , Antineoplastic Agents/adverse effects , Dose-Response Relationship, Drug , Hematopoietic Stem Cell Mobilization , Hematopoietic Stem Cell Transplantation , Megakaryocytes/immunology , Pilot Projects , Transplantation, Autologous , Tumor Cells, Cultured
12.
Bone Marrow Transplant ; 34(12): 1089-93, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15489877

ABSTRACT

To evaluate the impact of ex vivo expanded megakaryocyte (MK) progenitors on high-dose chemotherapy-induced thrombocytopenia, we conducted a phase II study in 10 patients with relapsed lymphoma. Two fractions of peripheral blood progenitor cells (PBPC) were cryopreserved, one with enough cells for at least 2 x 10(6) CD34+ cells/kg and a second obtained after CD34+ selection. Ten days before autologous stem cell transplantation, the CD34+ fraction was cultured with MGDF+SCF for 10 days. After BEAM (BCNU, cyclophosphamide, cytarabine, and melphalan) chemotherapy, patients were reinfused with standard PBPC and ex vivo expanded cells. No toxicity was observed after reinfusion. The mean fold expansion was 9.27 for nucleated cells, 2 for CD34+ cells, 676 for CD41+ cells, and 627 for CD61+ cells. The median date of platelet transfusion independence was day 8 (range: 7-12). All patients received at least one platelet transfusion. In conclusion, ex vivo expansion of MK progenitors was feasible and safe, but this procedure did not prevent BEAM-induced thrombocytopenia. Future studies will determine if expansion of higher numbers of CD34+ cells towards the MK-differentiation pathway will translate into a functional effect in terms of shortening of BEAM-induced thrombocytopenia.


Subject(s)
Erythroid Precursor Cells/cytology , Megakaryocytes/cytology , Peripheral Blood Stem Cell Transplantation/methods , Antigens, CD34/analysis , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/toxicity , Carmustine/administration & dosage , Cell Culture Techniques/methods , Cells, Cultured , Cyclophosphamide/administration & dosage , Cytarabine/administration & dosage , Erythroid Precursor Cells/drug effects , Erythroid Precursor Cells/transplantation , Humans , Integrin beta3/analysis , Lymphoma/complications , Lymphoma/therapy , Megakaryocytes/transplantation , Melphalan/administration & dosage , Platelet Membrane Glycoprotein IIb/analysis , Platelet Transfusion , Salvage Therapy , Thrombocytopenia/chemically induced , Thrombocytopenia/prevention & control , Thrombopoietin/pharmacology , Transplantation, Autologous , Treatment Outcome
13.
Cell Transplant ; 9(1): 125-31, 2000.
Article in English | MEDLINE | ID: mdl-10784074

ABSTRACT

As an early acting growth factor, flt-3 ligand (FL) promotes the ex vivo expansion of hematopoietic stem and progenitor cells. The effect and mechanism of FL on the development of the megakaryocytic lineage remain unclear. In this study, we compared the effects of FL and stem cell factor (SCF) in combination with other megakaryocyte-promoting cytokines on the differentiation and proliferation of megakaryocytic progenitors and investigated the expression of flt-3 receptors on megakaryocytic cell lines. In liquid cultures of enriched CD34+ cells from human umbilical cord blood for 14 days, FL plus thrombopoietin (TPO), interleukin-3 (IL-3), and IL-6 promoted the expansion of nucleated cells, CD34+ cells, CD34+ CD38- cells, and megakaryocyte colony-forming units (CFU-MK) by 300 +/- 115-, 23.8 +/- 11.3-, 33.9 +/- 28.6-, and 584 +/- 220-fold, respectively. Replacing FL with SCF significantly decreased the yield of all cell types. Using murine bone marrow (BM) cells, we demonstrated that FL at a range of 0-100 ng/ml had no significant mitogenic effect on CFU-MK formation. TPO increased CFU-MK (p < 0.001) but the effect was not significantly modified by FL. While one human acute lymphoblastic leukemia sample expressed high levels of flt-3 receptor, the four megakaryocytic cell lines (Meg-01, CHRF-288-11, M-07e, and Dami) did not show any positive expression. Our data suggest that the present cytokine combination and expansion conditions provide an effective and potentially useful system for the clinical expansion of cord blood for bone marrow transplantation (BMT). FL alone did not stimulate megakaryocytopoiesis, possibly due to the lack of receptor expression on megakaryocytes. The effect of FL in augmenting the expansion of CFU-MK in liquid culture might be due to the early action of FL at the pluripotent stem cell stage.


Subject(s)
Adjuvants, Immunologic/pharmacology , Hematopoietic Stem Cell Transplantation , Megakaryocytes/transplantation , Membrane Proteins/pharmacology , Thrombopoietin/pharmacology , Antigens, CD34/analysis , Cell Survival/drug effects , Flow Cytometry , Graft Survival/drug effects , Humans , Interleukin-3/pharmacology , Interleukin-6/pharmacology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive , Megakaryocytes/chemistry , Megakaryocytes/cytology , Stem Cell Factor/pharmacology , Tumor Cells, Cultured
14.
Int J Hematol ; 71(3): 203-10, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10846824

ABSTRACT

The use of platelet transfusion to ensure the recovery of thrombopoiesis in patients constitutes high-cost support. The identification and cloning of recombinant human thrombopoietin (TPO) and the development of efficient methods of purification of hematopoietic stem cells and progenitor cells have ameliorated the development of strategies of ex vivo expansion of megakaryocyte (MK) progenitor cells and mature MKs. Synergistic combinations of cytokines including TPO, interleukin (IL)-1, IL-3, IL-11, stem cell factor, and FLT-3 ligand induce the ex vivo expansion of colony-forming unit-MK progenitors and MKs from cytokine-mobilized peripheral blood cells, bone marrow, and cord blood CD34+ cells. Depending on the various culture conditions, i.e., combinations of growth factors, initial concentration of CD34+, serum or serum-free cultures, and/or oxygen tensions, the expansion-fold of MKs and their progenitor cells vary greatly. The clinical applications of the reinfusion of ex vivo-generated MK cells have been investigated successfully in cancer patients following high-dose chemotherapy. This review reports the latest information concerning ex vivo expansion of MKs and the current status of clinical trials.


Subject(s)
Megakaryocytes/cytology , Cell Culture Techniques/methods , Cell Division , Clinical Trials as Topic , Humans , Megakaryocytes/transplantation , Review Literature as Topic , Tissue Transplantation/methods , Tissue Transplantation/standards
15.
PLoS One ; 8(2): e54941, 2013.
Article in English | MEDLINE | ID: mdl-23390507

ABSTRACT

BACKGROUND: Currently, a constant shortage in the supply of platelets has become an important medical and society challenge, especially in developing country, and the in vitro production of megakaryocytic progenitor cells (MPs) from cord blood could represent an effective platelet substitute. In the present study, our objective was to determine the safety and feasibility of ex vivo generated MPs in patients. METHODS AND FINDINGS: MPs were produced and characterized from cord blood mononuclear cells under a serum free medium with cytokines. We investigated the feasibility of expansion and infusion of cord blood-derived MPs in 24 patients with advanced hematological malignancies. The primary end point was the safety and tolerability of the infusion of cord blood-derived MPs. No adverse effects were observed in patients who received ex vivo-generated cells at concentrations of up to a median value of 5.45 Ɨ 10(6)cells/kg of body weight. With one year follow-up, acute and chronic GVHD had not been observed among patients who received MPs infusion, even without ABO blood group and HLA typing matching. CONCLUSIONS: These initial results in patients are very encouraging. They suggest that infusion of cord blood-derived MPs appears safe and feasible for treatment of thrombocytopenia.


Subject(s)
Fetal Blood/cytology , Graft vs Host Disease/prevention & control , Hematologic Neoplasms/therapy , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/cytology , Megakaryocytes/transplantation , Thrombocytopenia/therapy , Adolescent , Adult , Aged , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Cells, Cultured , Culture Media, Serum-Free , Cytokines/pharmacology , Female , Hematologic Neoplasms/immunology , Hematologic Neoplasms/pathology , Humans , Male , Megakaryocytes/cytology , Middle Aged , Thrombocytopenia/immunology , Thrombocytopenia/pathology , Treatment Outcome
16.
J Clin Invest ; 120(11): 3807-10, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20972327

ABSTRACT

Thrombocytopenia, an abnormally low number of circulating platelets, results from inadequate platelet production, splenic platelet sequestration, or accelerated platelet clearance. Platelet transfusions are now the cornerstone for treating thrombocytopenia. With an ever-expanding demand for platelets, and with many patients having an inadequate response to platelet transfusions, new strategies are needed to treat thrombocytopenia. In this issue of the JCI, Fuentes et al. present provocative data regarding the use of direct megakaryocyte infusions as a novel approach to manage this vexing clinical problem.


Subject(s)
Megakaryocytes/transplantation , Platelet Transfusion/methods , Thrombocytopenia/therapy , Animals , Blood Platelets/metabolism , Humans , Mice , Thrombopoietin/metabolism , Transplantation, Heterologous
17.
J Clin Invest ; 120(11): 3917-22, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20972336

ABSTRACT

Thrombopoiesis, the process by which circulating platelets arise from megakaryocytes, remains incompletely understood. Prior studies suggest that megakaryocytes shed platelets in the pulmonary vasculature. To better understand thrombopoiesis and to develop a potential platelet transfusion strategy that is not dependent upon donors, of which there remains a shortage, we examined whether megakaryocytes infused into mice shed platelets. Infused megakaryocytes led to clinically relevant increases in platelet numbers. The released platelets were normal in size, displayed appropriate surface markers, and had a near-normal circulating half-life. The functionality of the donor-derived platelets was also demonstrated in vivo. The infused megakaryocytes mostly localized to the pulmonary vasculature, where they appeared to shed platelets. These data suggest that it may be unnecessary to generate platelets from ex vivo grown megakaryocytes to achieve clinically relevant increases in platelet numbers.


Subject(s)
Blood Platelets/metabolism , Cell Transplantation/methods , Megakaryocytes/transplantation , Animals , Blood Platelets/ultrastructure , Disease Models, Animal , Half-Life , Mice , Mice, Inbred C57BL , Platelet Count , Thrombocytopenia/physiopathology , Thrombocytopenia/therapy , Thrombopoiesis/physiology , Thrombosis/pathology
20.
Exp Hematol ; 37(11): 1330-1339.e5, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19664680

ABSTRACT

OBJECTIVE: A complete process for mass generation of megakaryocytes from hematopoietic stem cells under serum-free conditions has great clinical potential for rapid platelet reconstruction in thrombocytopenia patients. We have previously reported on the generation of an optimized serum-free medium (serum-free hematopoietic stem cell medium) for ex vivo expansion of CD34(+) cells. Here, we further generated large amounts of functional megakaryocytes from serum-free expanded CD34(+) cells under a complete and optimal serum-free condition for complying with clinical regulations. MATERIALS AND METHODS: Serum substitutes and cytokines were screened and optimized for their concentration for megakaryocyte generation by systemically methods. Serum-free induced megakaryocytes were characterized by surface antigens, gene expression, ex vivo megakaryocyte activation ability, and ability of megakaryocyte and platelet recovery in nonobese diabetic/severe combined immunodeficient mice. RESULTS: The optimal serum-free megakaryocyte induction medium was Iscove's modified Dulbecco's medium containing serum substitutes (i.e., human serum albumin, human insulin, and human transferrin) and a cytokine cocktail (i.e., thrombopoietin, stem cell factor, Fms-like tyrosine kinase 3 ligand, interleukin-3, interleukin-6, interleukin-9, and granulocyte-macrophage colony-stimulating factor). After induction, induced megakaryocytes expressed CD41a and CD61 surface antigens, nuclear factor erythroid-derived 2 and GATA-1 transcription factors and megakaryocyte activation ability. Importantly, transplantation of induced megakaryocytes could accelerate megakaryocyte and platelet recovery in irradiated nonobese diabetic/severe combined immunodeficient mice. CONCLUSION: In conclusion, we have developed a serum-free megakaryocyte induction medium, and the combination of serum-free megakaryocyte and serum-free hematopoietic stem cell media can generate a large amount of functional megakaryocytes efficiently. Our method represents a promising source of megakaryocytes and platelets for future cell therapy.


Subject(s)
Blood Platelets/cytology , Cell Culture Techniques/methods , Culture Media, Serum-Free/pharmacology , Hematopoietic Stem Cells/cytology , Megakaryocytes/transplantation , Animals , Antigens, CD34/analysis , Blood Cells/cytology , Bone Marrow Cells/cytology , Culture Media/pharmacology , Cytokines/pharmacology , Fetal Blood/cytology , Graft Survival , Humans , Integrin beta3/biosynthesis , Megakaryocytes/cytology , Megakaryocytes/drug effects , Mice , Mice, Inbred NOD , Mice, SCID , Platelet Glycoprotein GPIb-IX Complex/biosynthesis , Radiation Chimera , Thrombopoiesis
SELECTION OF CITATIONS
SEARCH DETAIL