Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 116
Filter
Add more filters

Publication year range
1.
Cell ; 174(3): 636-648.e18, 2018 07 26.
Article in English | MEDLINE | ID: mdl-30017246

ABSTRACT

The ex vivo generation of platelets from human-induced pluripotent cells (hiPSCs) is expected to compensate donor-dependent transfusion systems. However, manufacturing the clinically required number of platelets remains unachieved due to the low platelet release from hiPSC-derived megakaryocytes (hiPSC-MKs). Here, we report turbulence as a physical regulator in thrombopoiesis in vivo and its application to turbulence-controllable bioreactors. The identification of turbulent energy as a determinant parameter allowed scale-up to 8 L for the generation of 100 billion-order platelets from hiPSC-MKs, which satisfies clinical requirements. Turbulent flow promoted the release from megakaryocytes of IGFBP2, MIF, and Nardilysin to facilitate platelet shedding. hiPSC-platelets showed properties of bona fide human platelets, including circulation and hemostasis capacities upon transfusion in two animal models. This study provides a concept in which a coordinated physico-chemical mechanism promotes platelet biogenesis and an innovative strategy for ex vivo platelet manufacturing.


Subject(s)
Blood Platelets/metabolism , Cell Culture Techniques/methods , Thrombopoiesis/physiology , Bioreactors , Cell Culture Techniques/instrumentation , Humans , Hydrodynamics , Induced Pluripotent Stem Cells/metabolism , Megakaryocytes/metabolism , Megakaryocytes/physiology
2.
Blood ; 141(18): 2261-2274, 2023 05 04.
Article in English | MEDLINE | ID: mdl-36790527

ABSTRACT

Pathogenic missense variants in SLFN14, which encode an RNA endoribonuclease protein that regulates ribosomal RNA (rRNA) degradation, are known to cause inherited thrombocytopenia (TP) with impaired platelet aggregation and adenosine triphosphate secretion. Despite mild laboratory defects, the patients displayed an obvious bleeding phenotype. However, the function of SLFN14 in megakaryocyte (MK) and platelet biology remains unknown. This study aimed to model the disease in an immortalized MK cell line (imMKCL) and to characterize the platelet transcriptome in patients with the SLFN14 K219N variant. MK derived from heterozygous and homozygous SLFN14 K219N imMKCL and stem cells of blood from patients mainly presented with a defect in proplatelet formation and mitochondrial organization. SLFN14-defective platelets and mature MK showed signs of rRNA degradation; however, this was absent in undifferentiated imMKCL cells and granulocytes. Total platelet RNA was sequenced in 2 patients and 19 healthy controls. Differential gene expression analysis yielded 2999 and 2888 significantly (|log2 fold change| >1, false discovery rate <0.05) up- and downregulated genes, respectively. Remarkably, these downregulated genes were not enriched in any biological pathway, whereas upregulated genes were enriched in pathways involved in (mitochondrial) translation and transcription, with a significant upregulation of 134 ribosomal protein genes (RPGs). The upregulation of mitochondrial RPGs through increased mammalian target of rapamycin complex 1 (mTORC1) signaling in SLFN14 K219N MK seems to be a compensatory response to rRNA degradation. mTORC1 inhibition with rapamycin resulted in further enhanced rRNA degradation in SLFN14 K219N MK. Taken together, our study indicates dysregulation of mTORC1 coordinated ribosomal biogenesis is the disease mechanism for SLFN14-related TP.


Subject(s)
Thrombocytopenia , Humans , Thrombocytopenia/pathology , Blood Platelets/metabolism , Ribosomes/metabolism , Megakaryocytes/pathology , Ribosomal Proteins/genetics , Ribosomal Proteins/metabolism , Mechanistic Target of Rapamycin Complex 1/genetics , Mechanistic Target of Rapamycin Complex 1/metabolism , RNA/metabolism
3.
Biochem Biophys Res Commun ; 693: 149355, 2024 Jan 22.
Article in English | MEDLINE | ID: mdl-38096617

ABSTRACT

Nardilysin (NRDC) is a multifunctional protein required for maintaining homeostasis in various cellular and tissue contexts. However, its role in hematopoietic stem cells (HSCs) remains unclear. Here, through the conditional deletion of NRDC in hematopoietic cells, we demonstrate that NRDC is required for HSCs expansion in vitro and the reconstitution of hematopoiesis in vivo after transplantation. We found NRDC-deficient HSCs lose their self-renewal ability and display a preferential bias to myeloid differentiation in response to replication stress. Transcriptome data analysis revealed the upregulation of heat shock response-related genes in NRDC-deficient HSCs. Additionally, we observed increased protein synthesis in cultured NRDC-deficient HSCs. Thus, loss of NRDC may cause the inability to control protein synthesis in response to replication induced protein stress, leading to the impaired HSC self-renewal ability. This highlights a novel model of action of NRDC specifically in HSCs.


Subject(s)
Hematopoietic Stem Cells , Metalloendopeptidases , Hematopoietic Stem Cells/metabolism , Metalloendopeptidases/metabolism , Hematopoiesis/physiology , Up-Regulation , Cell Differentiation/genetics
4.
Biochem Biophys Res Commun ; 662: 76-83, 2023 06 25.
Article in English | MEDLINE | ID: mdl-37099813

ABSTRACT

Human induced pluripotent stem cells (hiPSCs) genetically depleted of human leucocyte antigen (HLA) class I expression can bypass T cell alloimmunity and thus serve as a one-for-all source for cell therapies. However, these same therapies may elicit rejection by natural killer (NK) cells, since HLA class I molecules serve as inhibitory ligands of NK cells. Here, we focused on testing the capacity of endogenously developed human NK cells in humanized mice (hu-mice) using MTSRG and NSG-SGM3 strains to assay the tolerance of HLA-edited iPSC-derived cells. High NK cell reconstitution was achieved with the engraftment of cord blood-derived human hematopoietic stem cells (hHSCs) followed by the administration of human interleukin-15 (hIL-15) and IL-15 receptor alpha (hIL-15Rα). Such "hu-NK mice" rejected HLA class I-null hiPSC-derived hematopoietic progenitor cells (HPCs), megakaryocytes and T cells, but not HLA-A/B-knockout, HLA-C expressing HPCs. To our knowledge, this study is the first to recapitulate the potent endogenous NK cell response to non-tumor HLA class I-downregulated cells in vivo. Our hu-NK mouse models are suitable for the non-clinical evaluation of HLA-edited cells and will contribute to the development of universal off-the-shelf regenerative medicine.


Subject(s)
Induced Pluripotent Stem Cells , Humans , Animals , Mice , Killer Cells, Natural , Histocompatibility Antigens Class I/metabolism , T-Lymphocytes , HLA Antigens/metabolism
5.
Blood ; 136(6): 715-725, 2020 08 06.
Article in English | MEDLINE | ID: mdl-32384141

ABSTRACT

Studies of inherited platelet disorders have provided many insights into platelet development and function. Loss of function of neurobeachin-like 2 (NBEAL2) causes gray platelet syndrome (GPS), where the absence of platelet α-granules indicates NBEAL2 is required for their production by precursor megakaryocytes. The endoplasmic reticulum is a dynamic network that interacts with numerous intracellular vesicles and organelles and plays key roles in their development. The megakaryocyte endoplasmic reticulum is extensive, and in this study we investigated its role in the biogenesis of α-granules by focusing on the membrane-resident trafficking protein SEC22B. Coimmunoprecipitation (co-IP) experiments using tagged proteins expressed in human HEK293 and megakaryocytic immortalized megakaryocyte progenitor (imMKCL) cells established binding of NBEAL2 with SEC22B, and demonstrated that NBEAL2 can simultaneously bind SEC22B and P-selectin. NBEAL2-SEC22B binding was also observed for endogenous proteins in human megakaryocytes using co-IP, and immunofluorescence microscopy detected substantial overlap. SEC22B binding was localized to a region of NBEAL2 spanning amino acids 1798 to 1903, where 2 GPS-associated missense variants have been reported: E1833K and R1839C. NBEAL2 containing either variant did not bind SEC22B coexpressed in HEK293 cells. CRISPR/Cas9-mediated knockout of SEC22B in imMKCL cells resulted in decreased NBEAL2, but not vice versa. Loss of either SEC22B or NBEAL2 expression resulted in failure of α-granule production and reduced granule proteins in imMKCL cells. We conclude that SEC22B is required for α-granule biogenesis in megakaryocytes, and that interactions with SEC22B and P-selectin facilitate the essential role of NBEAL2 in granule development and cargo stability.


Subject(s)
Blood Proteins/physiology , Cytoplasmic Granules/physiology , Endoplasmic Reticulum/physiology , Megakaryocytes/ultrastructure , Organelle Biogenesis , R-SNARE Proteins/physiology , Binding Sites , Blood Proteins/deficiency , Blood Proteins/genetics , Cells, Cultured , Gene Knockout Techniques , Gray Platelet Syndrome/genetics , HEK293 Cells , Humans , Immunoprecipitation , Megakaryocyte Progenitor Cells , Megakaryocytes/metabolism , Microscopy, Confocal , Microscopy, Fluorescence , Mutation, Missense , P-Selectin/physiology , Protein Interaction Mapping , Recombinant Proteins/metabolism
6.
Cell Mol Life Sci ; 78(7): 3385-3401, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33439272

ABSTRACT

The discovery of iPSCs has led to the ex vivo production of differentiated cells for regenerative medicine. In the case of transfusion products, the derivation of platelets from iPSCs is expected to complement our current blood-donor supplied transfusion system through donor-independent production with complete pathogen-free assurance. This derivation can also overcome alloimmune platelet transfusion refractoriness by resulting in autologous, HLA-homologous or HLA-deficient products. Several developments were necessary to produce a massive number of platelets required for a single transfusion. First, expandable megakaryocytes were established from iPSCs through transgene expression. Second, a turbulent-type bioreactor with improved platelet yield and quality was developed. Third, novel drugs that enabled efficient feeder cell-free conditions were developed. Fourth, the platelet-containing suspension was purified and resuspended in an appropriate buffer. Finally, the platelet product needed to be assured for competency and safety including non-tumorigenicity through in vitro and in vivo preclinical tests. Based on these advancements, a clinical trial has started. The generation of human iPSC-derived platelets could evolve transfusion medicine to the next stage and assure a ubiquitous, safe supply of platelet products. Further, considering the feasibility of gene manipulations in iPSCs, other platelet products may bring forth novel therapeutic measures.


Subject(s)
Blood Platelets/cytology , Blood Transfusion/methods , Induced Pluripotent Stem Cells/cytology , Megakaryocytes/cytology , Regenerative Medicine , Animals , Cell Differentiation , Humans
7.
Proc Natl Acad Sci U S A ; 116(11): 4983-4988, 2019 03 12.
Article in English | MEDLINE | ID: mdl-30804189

ABSTRACT

Platelets mediate primary hemostasis, and recent work has emphasized platelet participation in immunity and inflammation. The function of the platelet-specific integrin αIIbß3 as a fibrinogen receptor in hemostasis is well defined, but the roles of αIIbß3 or integrin-associated proteins in nonhemostatic platelet functions are poorly understood. Here we show that human platelets express the integrin-associated protein SHARPIN with functional consequences. In leukocytes, SHARPIN interacts with integrin α cytoplasmic tails, and it is also an obligate member of the linear ubiquitin chain assembly complex (LUBAC), which mediates Met1 linear ubiquitination of proteins leading to canonical NF-κB activation. SHARPIN interacted with αIIb in pull-down and coimmunoprecipitation assays. SHARPIN was partially localized, as was αIIbß3, at platelet edges, and thrombin stimulation induced more central SHARPIN localization. SHARPIN also coimmunoprecipitated from platelets with the two other proteins comprising LUBAC, the E3 ligase HOIP and HOIL-1. Platelet stimulation with thrombin or inflammatory agonists, including lipopolysaccharide or soluble CD40 ligand (sCD40L), induced Met1 linear ubiquitination of the NF-κB pathway protein NEMO and serine-536 phosphorylation of the p65 RelA subunit of NF-κB. In human megakaryocytes and/or platelets derived from induced pluripotent stem (iPS) cells, SHARPIN knockdown caused increased basal and agonist-induced fibrinogen binding to αIIbß3 as well as reduced Met1 ubiquitination and RelA phosphorylation. Moreover, these SHARPIN knockdown cells exhibited increased surface expression of MHC class I molecules and increased release of sCD40L. These results establish that SHARPIN functions in the human megakaryocyte/platelet lineage through protein interactions at the nexus of integrin and immune/inflammatory signaling.


Subject(s)
Blood Platelets/metabolism , Signal Transduction , Ubiquitins/metabolism , Cell Lineage , Gene Knockdown Techniques , Homeostasis , Humans , I-kappa B Kinase/metabolism , Induced Pluripotent Stem Cells/metabolism , Inflammation/pathology , Megakaryocytes/metabolism , Models, Biological , NF-kappa B/metabolism , Platelet Membrane Glycoprotein IIb/metabolism , Protein Binding , Ubiquitin/metabolism , Ubiquitin-Protein Ligases/metabolism , Ubiquitination
8.
Rinsho Ketsueki ; 63(10): 1430-1439, 2022.
Article in Japanese | MEDLINE | ID: mdl-36351652

ABSTRACT

The COVID-19 pandemic has cast a shadow over transfusion medicine based on the blood donation system. However, managing alloimmune platelet transfusion refractoriness (allo-PTR) has already been difficult. As a first step toward resolving this issue using induced pluripotent stem cell-derived platelet products (iPSC-PLTs), a clinical trial of autologous products (iPLAT1) was conducted in a patient with allo-PTR caused by anti-HPA-1a antibodies who had no compatible donor, and safety was confirmed. To produce iPSC-PLTs, a master cell bank (MCB) of expandable megakaryocyte lines (imMKCLs) is established from iPSCs. From this MCB, iPSC-PLTs are manufactured using a newly developed turbulent-type bioreactor and various compounds. Their quality, safety, and efficacy are confirmed by extensive preclinical studies. Based on the findings of the iPLAT1 study, a clinical trial of allo-transfusion of HLA homozygous iPSC-PLTs is currently ongoing and HLA class I-deficient O-type universal iPSC-PLTs are also being developed. iPSC-PLTs are expected to solve various problems, including allo-PTR in platelet transfusion, and greatly contribute to the advancement of transfusion medicine.


Subject(s)
COVID-19 , Induced Pluripotent Stem Cells , Thrombocytopenia , Humans , Blood Platelets/metabolism , Pandemics , Platelet Transfusion
9.
Am J Hum Genet ; 103(3): 440-447, 2018 09 06.
Article in English | MEDLINE | ID: mdl-30146126

ABSTRACT

Inherited bone-marrow-failure syndromes (IBMFSs) include heterogeneous genetic disorders characterized by bone-marrow failure, congenital anomalies, and an increased risk of malignancy. Many lines of evidence have suggested that p53 activation might be central to the pathogenesis of IBMFSs, including Diamond-Blackfan anemia (DBA) and dyskeratosis congenita (DC). However, the exact role of p53 activation in each clinical feature remains unknown. Here, we report unique de novo TP53 germline variants found in two individuals with an IBMFS accompanied by hypogammaglobulinemia, growth retardation, and microcephaly mimicking DBA and DC. TP53 is a tumor-suppressor gene most frequently mutated in human cancers, and occasional germline variants occur in Li-Fraumeni cancer-predisposition syndrome. Most of these mutations affect the core DNA-binding domain, leading to compromised transcriptional activities. In contrast, the variants found in the two individuals studied here caused the same truncation of the protein, resulting in the loss of 32 residues from the C-terminal domain (CTD). Unexpectedly, the p53 mutant had augmented transcriptional activities, an observation not previously described in humans. When we expressed this mutant in zebrafish and human-induced pluripotent stem cells, we observed impaired erythrocyte production. These findings together with close similarities to published knock-in mouse models of TP53 lacking the CTD demonstrate that the CTD-truncation mutations of TP53 cause IBMFS, providing important insights into the previously postulated connection between p53 and IBMFSs.


Subject(s)
Bone Marrow Diseases/genetics , Bone Marrow/pathology , Germ Cells/pathology , Mutation/genetics , Tumor Suppressor Protein p53/genetics , Adolescent , Adult , Agammaglobulinemia/genetics , Anemia, Diamond-Blackfan/genetics , Animals , Child, Preschool , Erythrocytes/pathology , Female , Growth Disorders/genetics , Humans , Induced Pluripotent Stem Cells/pathology , Infant , Infant, Newborn , Male , Mice , Middle Aged , Young Adult , Zebrafish
10.
Dev Growth Differ ; 63(2): 178-186, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33507533

ABSTRACT

In the body, platelets mainly work as a hemostatic agent, and the lack of platelets can cause serious bleeding. Induced pluripotent stem (iPS) cells potentially allow for a stable supply of platelets that are independent of donors and eliminate the risk of infection. However, a major challenge in iPS cell-based systems is producing the number of platelets required for a single transfusion (more than 200 billion in Japan). Thus, development in large-scale culturing technology is required. In previous studies, we generated a self-renewable, immortalized megakaryocyte cell line by transfecting iPS cell-derived hematopoietic progenitor cells with c-MYC, BMI1, and BCL-XL genes. Optimization of the culture conditions, including the discovery of a novel fluid-physical factor, turbulence, in the production of platelets in vivo, and the development of bioreactors that apply turbulence have enabled us to generate platelets of clinical quality and quantity. We have further generated platelets deleted of HLA class I expression by using genetic modification technology for patients suffering from alloimmune transfusion refractoriness, since these patients are underserved by current blood donation systems. In this review, we highlight current research and our recent work on iPS cell-derived platelet induction.


Subject(s)
Blood Platelets/cytology , Induced Pluripotent Stem Cells/cytology , Cell Culture Techniques , Cell Differentiation , Humans
11.
Proc Natl Acad Sci U S A ; 115(35): E8228-E8235, 2018 08 28.
Article in English | MEDLINE | ID: mdl-30104364

ABSTRACT

New mechanisms behind blood cell formation continue to be uncovered, with therapeutic approaches for hematological diseases being of great interest. Here we report an enzyme in protein synthesis, known for cell-based activities beyond translation, is a factor inducing megakaryocyte-biased hematopoiesis, most likely under stress conditions. We show an activated form of tyrosyl-tRNA synthetase (YRSACT), prepared either by rationally designed mutagenesis or alternative splicing, induces expansion of a previously unrecognized high-ploidy Sca-1+ megakaryocyte population capable of accelerating platelet replenishment after depletion. Moreover, YRSACT targets monocytic cells to induce secretion of transacting cytokines that enhance megakaryocyte expansion stimulating the Toll-like receptor/MyD88 pathway. Platelet replenishment by YRSACT is independent of thrombopoietin (TPO), as evidenced by expansion of the megakaryocytes from induced pluripotent stem cell-derived hematopoietic stem cells from a patient deficient in TPO signaling. We suggest megakaryocyte-biased hematopoiesis induced by YRSACT offers new approaches for treating thrombocytopenia, boosting yields from cell-culture production of platelet concentrates for transfusion, and bridging therapy for hematopoietic stem cell transplantation.


Subject(s)
Blood Platelets/metabolism , Hematopoiesis , Megakaryocytes/metabolism , Polyploidy , Thrombocytopenia/metabolism , Tyrosine-tRNA Ligase/metabolism , Blood Platelets/pathology , Cell Culture Techniques , Cells, Cultured , Female , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/pathology , Humans , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/pathology , Male , Megakaryocytes/pathology , Signal Transduction , Thrombocytopenia/pathology , Thrombopoietin/metabolism
12.
Rinsho Ketsueki ; 61(6): 628-633, 2020.
Article in Japanese | MEDLINE | ID: mdl-32624536

ABSTRACT

Since induced pluripotent stem (iPS) cell-derived blood products can be produced from any individual, they are expected to complement current transfusion products. However, a main problem is how to produce 10 U platelet preparations. Therefore, we established an immortalized megakaryocyte cell line (imMKCL) from iPS cells. We also found that turbulent flow was an essential physical factor for platelet generation in vivo. This knowledge enabled us to obtain 100 billion functional platelets from imMKCL using an 8 L bioreactor. We propose that the enhanced platelet production in the bioreactor occurs due to the turbulent flow that promoted the release of stress-induced cytokines.


Subject(s)
Blood Platelets , Bioreactors , Induced Pluripotent Stem Cells , Megakaryocytes , Thrombopoiesis
13.
Development ; 143(12): 2039-43, 2016 06 15.
Article in English | MEDLINE | ID: mdl-27302394

ABSTRACT

Ten years ago, the discovery that mature somatic cells could be reprogrammed into induced pluripotent stem cells (iPSCs) redefined the stem cell field and brought about a wealth of opportunities for both basic research and clinical applications. To celebrate the tenth anniversary of the discovery, the International Society for Stem Cell Research (ISSCR) and Center for iPS Cell Research and Application (CiRA), Kyoto University, together held the symposium 'Pluripotency: From Basic Science to Therapeutic Applications' in Kyoto, Japan. The three days of lectures examined both the mechanisms and therapeutic applications of iPSC reprogramming. Here we summarize the main findings reported, which are testament to how far the field has come in only a decade, as well as the enormous potential that iPSCs hold for the future.


Subject(s)
Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/transplantation , Stem Cell Transplantation , Animals , Cell Differentiation , Disease , Humans
15.
Rinsho Ketsueki ; 60(9): 1046-1055, 2019.
Article in Japanese | MEDLINE | ID: mdl-31597826

ABSTRACT

Human iPS cells are somatic cells reprogrammed to the pluripotent state. Because of their pluripotent nature, iPS cells are now commonly used to model several developmental processes including hematopoiesis in vitro. The in vitro models can be used to study the mechanisms regulating not only normal hematopoiesis but also hematological diseases ranging from monogenic congenital disorders to genetically multifactorial malignancies. Those disease models can also be used to investigate novel treatments through procedures including high throughput drug screening. The possible clinical applications of iPS cell-derived hematopoietic cells include immunotherapy with T lymphocytes, NK cells and macrophages, and transfusion therapy with platelets and red blood cells. Platelets have now been produced from iPS cells in quantities sufficient for clinical use. By developing expandable immortalized megakaryocyte cell lines (imMKCLs), several novel drugs and turbulence-incorporated bioreactors, efficient and scalable generation of platelets was achieved. This review summarizes the current status of iPS cell research in hematopoiesis with details on iPS cell-derived platelets.


Subject(s)
Blood Platelets/cytology , Cell- and Tissue-Based Therapy , Induced Pluripotent Stem Cells/cytology , Cell Differentiation , Erythrocytes , Hematopoiesis , Humans , Immunotherapy , Killer Cells, Natural , Macrophages , Megakaryocytes , T-Lymphocytes
16.
Br J Haematol ; 181(6): 791-802, 2018 06.
Article in English | MEDLINE | ID: mdl-29741776

ABSTRACT

Somatic mutations in the calreticulin (CALR) gene have been found in most patients with JAK2- and MPL-unmutated Philadelphia chromosome-negative myeloproliferative neoplasms (MPNs). It has recently been shown that mutant CALR constitutively activates the thrombopoietin receptor MPL and, thus, plays a causal role in the development of MPNs. However, the roles of mutant CALR in human haematopoietic cell differentiation remain predominantly elusive. To examine the impact of the 5-base insertion mutant CALR gene (Ins5) on haematopoietic cell differentiation, we generated induced pluripotent stem cells from an essential thrombocythaemia (ET) patient harbouring a CALR-Ins5 mutation and from a healthy individual (WT). Megakaryopoiesis was more prominent in Ins5-haematopoietic progenitor cells (Ins5-HPCs) than in WT-HPCs, implying that the system recapitulates megakaryocytosis observed in the bone marrow of CALR-mutant ET patients. Ins5-HPCs exhibited elevated expression levels of GATA1 and GATA2, suggesting a premature commitment to megakaryocytic differentiation in progenitor cells. We also demonstrated that 3-hydroxy anagrelide markedly perturbed megakaryopoiesis, but not erythropoiesis. Collectively, we established an in vitro model system that recapitulates megakaryopoiesis caused by mutant CALR. This system can be used to validate therapeutic compounds for MPN patients harbouring CALR mutations and in detailed studies on mutant CALR in human haematological cell differentiation.


Subject(s)
Calreticulin/metabolism , Cell Differentiation , Hematopoietic Stem Cells/metabolism , Induced Pluripotent Stem Cells/metabolism , Megakaryocytes/metabolism , Mutation , Myelopoiesis , Calreticulin/genetics , Female , GATA1 Transcription Factor/genetics , GATA1 Transcription Factor/metabolism , GATA2 Transcription Factor/genetics , GATA2 Transcription Factor/metabolism , Hematopoietic Stem Cells/cytology , Humans , Induced Pluripotent Stem Cells/cytology , Male , Megakaryocytes/cytology
17.
Blood ; 127(10): 1234-41, 2016 Mar 10.
Article in English | MEDLINE | ID: mdl-26787737

ABSTRACT

Thrombocytopenia is defined as a status in which platelet numbers are reduced. Imbalance between the homeostatic regulation of platelet generation and destruction is 1 potential cause of thrombocytopenia. In adults, platelet generation is a 2-stage process entailing the differentiation of hematopoietic stem cells into mature megakaryocytes (MKs; known as megakaryopoiesis) and release of platelets from MKs (known as thrombopoiesis or platelet biogenesis). Until recently, information about the genetic defects responsible for congenital thrombocytopenia was only available for a few forms of the disease. However, investigations over the past 15 years have identified mutations in genes encoding >20 different proteins that are responsible for these disorders, which has advanced our understanding of megakaryopoiesis and thrombopoiesis. The underlying pathogenic mechanisms can be categorized as (1) defects in MK lineage commitment and differentiation, (2) defects in MK maturation, and (3) defect in platelet release. Using these developmental stage categories, we here update recently described mechanisms underlying megakaryopoiesis and thrombopoiesis and discuss the association between platelet generation systems and thrombocytopenia.


Subject(s)
Genetic Diseases, Inborn , Hematopoietic Stem Cells , Megakaryocytes , Mutation , Thrombocytopenia , Thrombopoiesis/genetics , Adult , Animals , Genetic Diseases, Inborn/genetics , Genetic Diseases, Inborn/metabolism , Genetic Diseases, Inborn/pathology , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/pathology , Humans , Megakaryocytes/metabolism , Megakaryocytes/pathology , Thrombocytopenia/genetics , Thrombocytopenia/metabolism , Thrombocytopenia/pathology
18.
Proc Natl Acad Sci U S A ; 112(41): 12800-5, 2015 Oct 13.
Article in English | MEDLINE | ID: mdl-26417084

ABSTRACT

Phosphatidylserine (PtdSer) exposure on the surface of activated platelets requires the action of a phospholipid scramblase(s), and serves as a scaffold for the assembly of the tenase and prothrombinase complexes involved in blood coagulation. Here, we found that the activation of mouse platelets with thrombin/collagen or Ca(2+) ionophore at 20 °C induces PtdSer exposure without compromising plasma membrane integrity. Among five transmembrane protein 16 (TMEM16) members that support Ca(2+)-dependent phospholipid scrambling, TMEM16F was the only one that showed high expression in mouse platelets. Platelets from platelet-specific TMEM16F-deficient mice exhibited defects in activation-induced PtdSer exposure and microparticle shedding, although α-granule and dense granule release remained intact. The rate of tissue factor-induced thrombin generation by TMEM16F-deficient platelets was severely reduced, whereas thrombin-induced clot retraction was unaffected. The imaging of laser-induced thrombus formation in whole animals showed that PtdSer exposure on aggregated platelets was TMEM16F-dependent in vivo. The phenotypes of the platelet-specific TMEM16F-null mice resemble those of patients with Scott syndrome, a mild bleeding disorder, indicating that these mice may provide a useful model for human Scott syndrome.


Subject(s)
Blood Platelets/metabolism , Cell-Derived Microparticles/metabolism , Phosphatidylserines/metabolism , Phospholipid Transfer Proteins/metabolism , Platelet Activation , Animals , Anoctamins , Blood Coagulation/genetics , Blood Coagulation Disorders/genetics , Blood Coagulation Disorders/metabolism , Blood Coagulation Disorders/pathology , Blood Platelets/pathology , Calcium/metabolism , Cell-Derived Microparticles/genetics , Disease Models, Animal , Humans , Mice , Mice, Mutant Strains , Phosphatidylserines/genetics , Phospholipid Transfer Proteins/genetics , Thrombin/genetics , Thrombin/metabolism
19.
Rinsho Ketsueki ; 59(10): 1905-1913, 2018.
Article in Japanese | MEDLINE | ID: mdl-30305491

ABSTRACT

Platelet transfusion products derived from induced pluripotent stem cells (iPSCs) have been pursued as a blood donor-independent and genetically manipulative measure to complement or as an alternative to current platelet products. Platelets are enucleate blood cells indispensable for hemostasis. Thus, platelet transfusions have been clinically established to treat patients with severe thrombocytopenia. However, current blood products face issues in the balance of supply and demand, alloimmune responses, and infections and are expected to meet the shortage of donors in aging societies. iPSc-derived platelet products are qualitatively and quantitatively approaching a clinically applicable level, owing to advances and novel findings in expandable megakaryocyte cell lines, turbulence-incorporating bioreactors, and reagents that enable feeder cell-free production and improve platelet quality. Currently, the establishment of guidelines to assure the quality of iPSC-derived blood products for clinical application is in process. Considering the low risk of tumorigenicity and the large demand, ex vivo production of iPSC-derived platelets could lead to iPSC-based regenerative medicine becoming a common clinical practice and the development of a future system in which anyone can safely receive a platelet transfusion in their time of need.


Subject(s)
Blood Platelets/cytology , Cell Culture Techniques , Induced Pluripotent Stem Cells/cytology , Thrombopoiesis , Humans , Megakaryocytes/cytology , Platelet Transfusion
20.
Transfusion ; 57(8): 2035-2044, 2017 08.
Article in English | MEDLINE | ID: mdl-28681458

ABSTRACT

BACKGROUND: To bridge the gap between in vitro function and clinical efficacy of platelet (PLT) transfusion products, reliable in vivo PLT functional assays for hemostasis and survival in animal models are required. However, there are no standardized methods for assessing the in vivo quality of transfused human PLTs. STUDY DESIGN AND METHODS: Plasma-depleted human PLT concentrates (PCs; Day 3, Day 5, Day 7, Day 10, and damaged) were transfused into busulfan-induced rabbits with thrombocytopenia with prolonged bleeding times 1 day after treatment with ethyl palmitate (EP) to block their reticuloendothelial systems. The hemostatic effect of PC transfusion was evaluated by the ear fine vein bleeding time. For the in vivo survival assay, splenectomized EP-treated rabbits were transfused with human PCs, and viability of the human PLTs in the rabbits was determined by flow cytometry using human PLT-specific antibodies and Trucount tubes. RESULTS: The hemostatic effect of PCs was slightly reduced with increasing storage periods for early time points, but more dramatically reduced for later time points. PLT survival was similar after 3 and 7 days of storage, but PLTs stored for 10 days showed significantly poorer survival than those stored only 3 days. CONCLUSION: Our new and improved protocol for in vivo assessment of transfused PLTs is sufficiently sensitive to detect subtle changes in hemostatic function and viability of human PLTs transfused into rabbit models. This protocol could contribute to preclinical in vivo functional assessment and clinical quality assurance of emerging novel PLT products such as cultured cell-derived human PLTs.


Subject(s)
Blood Platelets/cytology , Cell Survival , Hemostasis , Platelet Function Tests/methods , Platelet Transfusion , Animals , Blood Preservation/methods , Flow Cytometry/methods , Humans , Methods , Models, Animal , Rabbits , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL