Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 73
Filtrar
1.
Adv Sci (Weinh) ; : e2308276, 2024 Mar 21.
Artigo em Inglês | MEDLINE | ID: mdl-38514919

RESUMO

Hematopoietic stem and progenitor cells (HSPCs) continuously generate platelets throughout one's life. Inherited Platelet Disorders affect ≈ 3 million individuals worldwide and are characterized by defects in platelet formation or function. A critical challenge in the identification of these diseases lies in the absence of models that facilitate the study of hematopoiesis ex vivo. Here, a silk fibroin-based bioink is developed and designed for 3D bioprinting. This bioink replicates a soft and biomimetic environment, enabling the controlled differentiation of HSPCs into platelets. The formulation consisting of silk fibroin, gelatin, and alginate is fine-tuned to obtain a viscoelastic, shear-thinning, thixotropic bioink with the remarkable ability to rapidly recover after bioprinting and provide structural integrity and mechanical stability over long-term culture. Optical transparency allowed for high-resolution imaging of platelet generation, while the incorporation of enzymatic sensors allowed quantitative analysis of glycolytic metabolism during differentiation that is represented through measurable color changes. Bioprinting patient samples revealed a decrease in metabolic activity and platelet production in Inherited Platelet Disorders. These discoveries are instrumental in establishing reference ranges for classification and automating the assessment of treatment responses. This model has far-reaching implications for application in the research of blood-related diseases, prioritizing drug development strategies, and tailoring personalized therapies.

2.
Nat Commun ; 15(1): 2588, 2024 Mar 22.
Artigo em Inglês | MEDLINE | ID: mdl-38519457

RESUMO

We recently achieved the first-in-human transfusion of induced pluripotent stem cell-derived platelets (iPSC-PLTs) as an alternative to standard transfusions, which are dependent on donors and therefore variable in supply. However, heterogeneity characterized by thrombopoiesis-biased or immune-biased megakaryocytes (MKs) continues to pose a bottleneck against the standardization of iPSC-PLT manufacturing. To address this problem, here we employ microRNA (miRNA) switch biotechnology to distinguish subpopulations of imMKCLs, the MK cell lines producing iPSC-PLTs. Upon miRNA switch-based screening, we find imMKCLs with lower let-7 activity exhibit an immune-skewed transcriptional signature. Notably, the low activity of let-7a-5p results in the upregulation of RAS like proto-oncogene B (RALB) expression, which is crucial for the lineage determination of immune-biased imMKCL subpopulations and leads to the activation of interferon-dependent signaling. The dysregulation of immune properties/subpopulations, along with the secretion of inflammatory cytokines, contributes to a decline in the quality of the whole imMKCL population.


Assuntos
Células-Tronco Pluripotentes Induzidas , MicroRNAs , Humanos , Megacariócitos , Células-Tronco Pluripotentes Induzidas/metabolismo , Plaquetas/metabolismo , Trombopoese/genética , MicroRNAs/genética , MicroRNAs/metabolismo
3.
Biochem Biophys Res Commun ; 693: 149355, 2024 Jan 22.
Artigo em Inglês | MEDLINE | ID: mdl-38096617

RESUMO

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.


Assuntos
Células-Tronco Hematopoéticas , Metaloendopeptidases , Células-Tronco Hematopoéticas/metabolismo , Metaloendopeptidases/metabolismo , Hematopoese/fisiologia , Regulação para Cima , Diferenciação Celular/genética
4.
Biochem Biophys Res Commun ; 662: 76-83, 2023 06 25.
Artigo em Inglês | MEDLINE | ID: mdl-37099813

RESUMO

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.


Assuntos
Células-Tronco Pluripotentes Induzidas , Humanos , Animais , Camundongos , Células Matadoras Naturais , Antígenos de Histocompatibilidade Classe I/metabolismo , Linfócitos T , Antígenos HLA/metabolismo
5.
Blood ; 141(18): 2261-2274, 2023 05 04.
Artigo em Inglês | MEDLINE | ID: mdl-36790527

RESUMO

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.


Assuntos
Trombocitopenia , Humanos , Trombocitopenia/patologia , Plaquetas/metabolismo , Ribossomos/metabolismo , Megacariócitos/patologia , Proteínas Ribossômicas/genética , Proteínas Ribossômicas/metabolismo , Alvo Mecanístico do Complexo 1 de Rapamicina/genética , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , RNA/metabolismo
6.
Blood Adv ; 6(23): 6056-6069, 2022 12 13.
Artigo em Inglês | MEDLINE | ID: mdl-36149941

RESUMO

Donor-derived platelets are used to treat or prevent hemorrhage in patients with thrombocytopenia. However, ∼5% or more of these patients are complicated with alloimmune platelet transfusion refractoriness (allo-PTR) due to alloantibodies against HLA-I or human platelet antigens (HPA). In these cases, platelets from compatible donors are necessary, but it is difficult to find such donors for patients with rare HLA-I or HPA. To produce platelet products for patients with aplastic anemia with allo-PTR due to rare HPA-1 mismatch in Japan, we developed an ex vivo good manufacturing process (GMP)-based production system for an induced pluripotent stem cell-derived platelet product (iPSC-PLTs). Immortalized megakaryocyte progenitor cell lines (imMKCLs) were established from patient iPSCs, and a competent imMKCL clone was selected for the master cell bank (MCB) and confirmed for safety, including negativity of pathogens. From this MCB, iPSC-PLTs were produced using turbulent flow bioreactors and new drugs. In extensive nonclinical studies, iPSC-PLTs were confirmed for quality, safety, and efficacy, including hemostasis in a rabbit model. This report presents a complete system for the GMP-based production of iPSC-PLTs and the required nonclinical studies and thus supports the iPLAT1 study, the first-in-human clinical trial of iPSC-PLTs in a patient with allo-PTR and no compatible donor using the autologous product. It also serves as a comprehensive reference for the development of widely applicable allogeneic iPSC-PLTs and other cell products that use iPSC-derived progenitor cells as MCB.


Assuntos
Antígenos de Plaquetas Humanas , Transplante de Células-Tronco Hematopoéticas , Células-Tronco Pluripotentes Induzidas , Trombocitopenia , Animais , Humanos , Coelhos , Transfusão de Plaquetas/efeitos adversos , Células-Tronco Pluripotentes Induzidas/metabolismo , Plaquetas/metabolismo , Trombocitopenia/etiologia , Transplante de Células-Tronco Hematopoéticas/efeitos adversos
7.
J Clin Invest ; 132(19)2022 10 03.
Artigo em Inglês | MEDLINE | ID: mdl-35925681

RESUMO

Infantile (fetal and neonatal) megakaryocytes (Mks) have a distinct phenotype consisting of hyperproliferation, limited morphogenesis, and low platelet production capacity. These properties contribute to clinical problems that include thrombocytopenia in neonates, delayed platelet engraftment in recipients of cord blood stem cell transplants, and inefficient ex vivo platelet production from pluripotent stem cell-derived Mks. The infantile phenotype results from deficiency of the actin-regulated coactivator, MKL1, which programs cytoskeletal changes driving morphogenesis. As a strategy to complement this molecular defect, we screened pathways with the potential to affect MKL1 function and found that DYRK1A inhibition dramatically enhanced Mk morphogenesis in vitro and in vivo. Dyrk1 inhibitors rescued enlargement, polyploidization, and thrombopoiesis in human neonatal Mks. Mks derived from induced pluripotent stem cells responded in a similar manner. Progenitors undergoing Dyrk1 inhibition demonstrated filamentous actin assembly, MKL1 nuclear translocation, and modulation of MKL1 target genes. Loss-of-function studies confirmed MKL1 involvement in this morphogenetic pathway. Expression of Ablim2, a stabilizer of filamentous actin, increased with Dyrk1 inhibition, and Ablim2 knockdown abrogated the actin, MKL1, and morphogenetic responses to Dyrk1 inhibition. These results delineate a pharmacologically tractable morphogenetic pathway whose manipulation may alleviate clinical problems associated with the limited thrombopoietic capacity of infantile Mks.


Assuntos
Megacariócitos , Trombocitopenia , Actinas/metabolismo , Plaquetas/metabolismo , Humanos , Recém-Nascido , Megacariócitos/metabolismo , Fenótipo , Proteínas Serina-Treonina Quinases , Proteínas Tirosina Quinases , Trombocitopenia/genética , Trombopoese/genética , Quinases Dyrk
8.
Nat Commun ; 13(1): 2691, 2022 05 16.
Artigo em Inglês | MEDLINE | ID: mdl-35577813

RESUMO

Hematopoietic stem cells (HSCs) exhibit considerable cell-intrinsic changes with age. Here, we present an integrated analysis of transcriptome and chromatin accessibility of aged HSCs and downstream progenitors. Alterations in chromatin accessibility preferentially take place in HSCs with aging, which gradually resolve with differentiation. Differentially open accessible regions (open DARs) in aged HSCs are enriched for enhancers and show enrichment of binding motifs of the STAT, ATF, and CNC family transcription factors that are activated in response to external stresses. Genes linked to open DARs show significantly higher levels of basal expression and their expression reaches significantly higher peaks after cytokine stimulation in aged HSCs than in young HSCs, suggesting that open DARs contribute to augmented transcriptional responses under stress conditions. However, a short-term stress challenge that mimics infection is not sufficient to induce persistent chromatin accessibility changes in young HSCs. These results indicate that the ongoing and/or history of exposure to external stresses may be epigenetically inscribed in HSCs to augment their responses to external stimuli.


Assuntos
Cromatina , Células-Tronco Hematopoéticas , Diferenciação Celular/genética , Cromatina/genética , Cromatina/metabolismo , Epigênese Genética , Epigenômica , Células-Tronco Hematopoéticas/metabolismo
9.
Methods Mol Biol ; 2454: 411-422, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-34724185

RESUMO

The human hematopoietic differentiation in vitro of human pluripotent stem cells (hPSCs) has provided new tools to elucidate the mechanisms of related genetic abnormalities, such as congenital diseases and acquired hematopoietic malignancies, and to discover new treatments. The differentiation can also be applied to developing a stable source of blood products for transfusion with minimal risk of several blood-borne infections. We previously proposed a method for hematopoietic progenitor cell (HPC) differentiation, the "hPSC-sac method", in which hPSCs are cocultured with C3H10T1/2 mouse stromal cells and mixed with a single cytokine, VEGF. The hPSC-sac method can differentiate hPSCs to multiple blood lineages. Here we describe improvements in the method by adding bFGF, TGFß inhibitor and heparin to the culture, which increases the yield of CD34+CD43+ HPCs 50-fold compared with the original protocol. This revised hPSC-sac method is expected to contribute to the development of disease models and regenerative medicine using hematopoietic lineage cells.


Assuntos
Células-Tronco Pluripotentes , Animais , Diferenciação Celular/genética , Hematopoese , Células-Tronco Hematopoéticas , Humanos , Camundongos , Fator de Crescimento Transformador beta
10.
Stem Cell Reports ; 16(12): 2861-2870, 2021 12 14.
Artigo em Inglês | MEDLINE | ID: mdl-34861163

RESUMO

Platelet transfusions are critical for severe thrombocytopenia but depend on blood donors. The shortage of donors and the potential of universal HLA-null platelet products have stimulated research on the ex vivo differentiation of human pluripotent stem cells (hPSCs) to platelets. We recently established expandable immortalized megakaryocyte cell lines (imMKCLs) from hPSCs by transducing MYC, BMI1, and BCL-XL (MBX). imMKCLs can act as cryopreservable master cells to supply platelet concentrates. However, the proliferation rates of the imMKCLs vary with the starting hPSC clone. In this study, we reveal from the gene expression profiles of several MKCL clones that the proliferation arrest is correlated with the expression levels of specific cyclin-dependent kinase inhibitors. Silencing CDKN1A and p53 with the overexpression of MBX was effective at stably inducing imMKCLs that generate functional platelets irrespective of the hPSC clone. Collectively, this improvement in generating imMKCLs should contribute to platelet industrialization and platelet biology.


Assuntos
Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Inativação Gênica , Células-Tronco Pluripotentes Induzidas/metabolismo , Células Progenitoras de Megacariócitos/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Plaquetas/metabolismo , Linhagem Celular , Proliferação de Células , Células Clonais , Técnicas de Silenciamento de Genes , Células HEK293 , Humanos , Complexo Repressor Polycomb 1/metabolismo , Proteínas Proto-Oncogênicas c-myc/metabolismo , Regulação para Cima , Proteína bcl-X/metabolismo
11.
Spine Surg Relat Res ; 5(3): 196-204, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34179558

RESUMO

INTRODUCTION: Platelet-rich plasma (PRP) is drawing attention as a substance that can promote bone formation. The growth factors present in PRP are stable for a long time after freeze-drying. However, the effects of PRP are inconsistent, and its effects on bone union in spinal surgery remain controversial. The immortalized megakaryocyte cell lines (imMKCLs) derived from human-induced pluripotent stem cells (hiPSCs) have been developed to produce numerous stable and clinically functional platelets. In this study, growth factors present in freeze-dried hiPSC-derived imMKCLs and platelets (iPS-MK/Plts) were evaluated, and their ability to promote bone formation was examined using a rat lumbar artificial bone grafting model. METHODS: We prepared freeze-dried iPS-MK/Plts and quantified their growth factors by enzyme-linked immunosorbent assays. Surgical grafting of artificial bone to the lumbar transverse processes was performed in 8-week-old female rats, which were divided into two groups: artificial bone graft (control) and artificial bone graft plus freeze-dried iPS-MK/Plts (iPS group). Transplantation was performed only on the left side. Eight weeks after the surgery, we captured computed tomography images and compared bilateral differences in the bone volume of the graft site in each rat. We also compared the left side/right side bone volume ratio between the two groups. RESULTS: The freeze-dried iPS-MK/Plts contained numerous growth factors. While there was no significant increase in bone volume on the transplanted side than that on the non-grafted side in the control group, bone volume significantly increased on the transplanted side in the iPS group, as evidenced by augmented mean left/right bone volume ratio of the iPS group compared with that of the control group. But the new bone observed in the iPS group was histologically normal. CONCLUSIONS: Freeze-dried hiPSC-derived MKCLs and platelets contain several stable growth factors and have the potential for promoting new bone formation.

12.
Stem Cell Res ; 53: 102287, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33813173

RESUMO

Recombinant matrices have enabled feeder cell-free maintenance cultures of human pluripotent stem cells (hPSCs), with laminin 511-E8 fragment (LM511-E8) being widely used. However, we herein report that hPSCs maintained on LM511-E8 resist differentiating to multipotent hematopoietic progenitor cells (HPCs), unlike hPSCs maintained on LM421-E8 or LM121-E8. The latter two LM-E8s bound weakly to hPSCs compared with LM511-E8 and activated the canonical Wnt/ß-catenin signaling pathway. Moreover, the extracellular LM-E8-dependent preferential hematopoiesis was associated with a higher expression of integrin ß1 (ITGB1) and downstream integrin-linked protein kinase (ILK), ß-catenin and phosphorylated JUN. Accordingly, the lower coating concentration of LM511-E8 or addition of a Wnt/ß-catenin signaling activator, CHIR99021, facilitated higher HPC yield. In contrast, the inhibition of ILK, Wnt or JNK by inhibitors or mRNA knockdown suppressed the HPC yield. These findings suggest that extracellular laminin scaffolds modulate the hematopoietic differentiation potential of hPSCs by activating the ITGB1-ILK-ß-catenin-JUN axis at the undifferentiated stage. Finally, the combination of low-concentrated LM511-E8 and a revised hPSC-sac method, which adds bFGF, SB431542 and heparin to the conventional method, enabled a higher yield of HPCs and higher rate for definitive hematopoiesis, suggesting a useful protocol for obtaining differentiated hematopoietic cells from hPSCs in general.


Assuntos
Transplante de Células-Tronco Hematopoéticas , Células-Tronco Pluripotentes , Humanos , Integrina beta1 , Laminina , beta Catenina/genética
13.
Cell Mol Life Sci ; 78(7): 3385-3401, 2021 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-33439272

RESUMO

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.


Assuntos
Plaquetas/citologia , Transfusão de Sangue/métodos , Células-Tronco Pluripotentes Induzidas/citologia , Megacariócitos/citologia , Medicina Regenerativa , Animais , Diferenciação Celular , Humanos
14.
Dev Growth Differ ; 63(2): 178-186, 2021 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-33507533

RESUMO

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.


Assuntos
Plaquetas/citologia , Células-Tronco Pluripotentes Induzidas/citologia , Técnicas de Cultura de Células , Diferenciação Celular , Humanos
15.
Inflamm Regen ; 40(1): 30, 2020 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-33292717

RESUMO

Platelet products are used in treatments for thrombocytopenia caused by hematopoietic diseases, chemotherapy, massive hemorrhages, extracorporeal circulation, and others. Their manufacturing depends on volunteers who donate blood. However, it is becoming increasingly necessary to reinforce this blood donation system with other blood sources due to the increase in demand and shortage of supply accompanying aging societies. In addition, blood-borne infections and alloimmune platelet transfusion refractoriness are not completely resolved. Since human induced pluripotent stem cell (iPSC)-platelet products can be supplied independently from the donor, it is expected to complement current platelet products. One big hurdle with iPSC-based systems is the production of 10 units, which is equivalent to 200 billion platelets. To overcome this issue, we established immortalized megakaryocyte cell lines (imMKCLs) by introducing three transgenes, c-MYC, BMI1, and BCL-XL, sequentially into hematopoietic and megakaryocytic progenitor stage cells derived from iPSCs. The three transgenes are regulated in a Tet-ON manner, enabling the addition and depletion of doxycycline to expand and maturate the imMKCLs, respectively. In addition, we succeeded in discovering drug combinations that enable feeder-free culture conditions in the imMKCL cultivation. Furthermore, we discovered the importance of turbulence in thrombopoiesis through live bone marrow imaging and developed a bioreactor based on the concept of turbulent flow. Eventually, through the identification of two key fluid physic parameters, turbulent energy and shear stress, we succeeded in scaling up the bioreactor to qualitatively and quantitatively achieve clinically applicable levels. Interestingly, three soluble factors released from imMKCLs in the turbulent flow condition, macrophage migration inhibitory factor (MIF), insulin growth factor binding protein 2 (IGFBP2), and nardilysin (NRDC), enhanced platelet production. Based on these developments, we initiated the first-in-human clinical trial of iPSC-derived platelets to a patient with alloimmune platelet transfusion refractoriness (allo-PTR) using an autologous product. In this review, we detail current research in this field and our study about the ex vivo production of iPSC-derived platelets.

16.
Stem Cell Reports ; 14(1): 49-59, 2020 01 14.
Artigo em Inglês | MEDLINE | ID: mdl-31883921

RESUMO

The ex vivo production of platelets depleted of human leukocyte antigen class I (HLA-I) could serve as a universal measure to overcome platelet transfusion refractoriness caused by HLA-I incompatibility. Here, we developed human induced pluripotent cell-derived HLA-I-deficient platelets (HLA-KO iPLATs) in a clinically applicable imMKCL system by genetic manipulation and assessed their immunogenic properties including natural killer (NK) cells, which reject HLA-I downregulated cells. HLA-KO iPLATs were deficient for all HLA-I but did not elicit a cytotoxic response by NK cells in vitro and showed circulation equal to wild-type iPLATs upon transfusion in our newly established Hu-NK-MSTRG mice reconstituted with human NK cells. Additionally, HLA-KO iPLATs successfully circulated in an alloimmune platelet transfusion refractoriness model of Hu-NK-MISTRG mice. Mechanistically, the lack of NK cell-activating ligands on platelets may be responsible for evading the NK cell response. This study revealed the unique non-immunogenic property of platelets and provides a proof of concept for the clinical application of HLA-KO iPLATs.


Assuntos
Plaquetas/citologia , Plaquetas/metabolismo , Diferenciação Celular , Antígenos de Histocompatibilidade Classe I/imunologia , Células-Tronco Pluripotentes Induzidas/citologia , Células Matadoras Naturais/imunologia , Células Matadoras Naturais/metabolismo , Animais , Citotoxicidade Imunológica/genética , Citotoxicidade Imunológica/imunologia , Técnicas de Inativação de Genes , Antígenos de Histocompatibilidade Classe I/genética , Humanos , Ativação Linfocitária/genética , Ativação Linfocitária/imunologia , Camundongos , Camundongos Knockout , Microglobulina beta-2/deficiência , Microglobulina beta-2/genética
17.
Rinsho Ketsueki ; 60(9): 1046-1055, 2019.
Artigo em Japonês | MEDLINE | ID: mdl-31597826

RESUMO

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.


Assuntos
Plaquetas/citologia , Terapia Baseada em Transplante de Células e Tecidos , Células-Tronco Pluripotentes Induzidas/citologia , Diferenciação Celular , Eritrócitos , Hematopoese , Humanos , Imunoterapia , Células Matadoras Naturais , Macrófagos , Megacariócitos , Linfócitos T
18.
Rinsho Ketsueki ; 59(10): 1905-1913, 2018.
Artigo em Japonês | MEDLINE | ID: mdl-30305491

RESUMO

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.


Assuntos
Plaquetas/citologia , Técnicas de Cultura de Células , Células-Tronco Pluripotentes Induzidas/citologia , Trombopoese , Humanos , Megacariócitos/citologia , Transfusão de Plaquetas
19.
Proc Natl Acad Sci U S A ; 115(35): E8228-E8235, 2018 08 28.
Artigo em Inglês | MEDLINE | ID: mdl-30104364

RESUMO

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.


Assuntos
Plaquetas/metabolismo , Hematopoese , Megacariócitos/metabolismo , Poliploidia , Trombocitopenia/metabolismo , Tirosina-tRNA Ligase/metabolismo , Plaquetas/patologia , Técnicas de Cultura de Células , Células Cultivadas , Feminino , Células-Tronco Hematopoéticas/metabolismo , Células-Tronco Hematopoéticas/patologia , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Células-Tronco Pluripotentes Induzidas/patologia , Masculino , Megacariócitos/patologia , Transdução de Sinais , Trombocitopenia/patologia , Trombopoetina/metabolismo
20.
Am J Hum Genet ; 103(3): 440-447, 2018 09 06.
Artigo em Inglês | MEDLINE | ID: mdl-30146126

RESUMO

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.


Assuntos
Doenças da Medula Óssea/genética , Medula Óssea/patologia , Células Germinativas/patologia , Mutação/genética , Proteína Supressora de Tumor p53/genética , Adolescente , Adulto , Agamaglobulinemia/genética , Anemia de Diamond-Blackfan/genética , Animais , Pré-Escolar , Eritrócitos/patologia , Feminino , Transtornos do Crescimento/genética , Humanos , Células-Tronco Pluripotentes Induzidas/patologia , Lactente , Recém-Nascido , Masculino , Camundongos , Pessoa de Meia-Idade , Adulto Jovem , Peixe-Zebra
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA