Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
1.
Cell ; 174(3): 636-648.e18, 2018 07 26.
Artículo en Inglés | MEDLINE | ID: mdl-30017246

RESUMEN

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.


Asunto(s)
Plaquetas/metabolismo , Técnicas de Cultivo de Célula/métodos , Trombopoyesis/fisiología , Reactores Biológicos , Técnicas de Cultivo de Célula/instrumentación , Humanos , Hidrodinámica , Células Madre Pluripotentes Inducidas/metabolismo , Megacariocitos/metabolismo , Megacariocitos/fisiología
2.
Biochem Biophys Res Commun ; 662: 76-83, 2023 06 25.
Artículo en Inglés | MEDLINE | ID: mdl-37099813

RESUMEN

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.


Asunto(s)
Células Madre Pluripotentes Inducidas , Humanos , Animales , Ratones , Células Asesinas Naturales , Antígenos de Histocompatibilidad Clase I/metabolismo , Linfocitos T , Antígenos HLA/metabolismo
3.
Am J Hum Genet ; 103(3): 440-447, 2018 09 06.
Artículo en Inglés | MEDLINE | ID: mdl-30146126

RESUMEN

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.


Asunto(s)
Enfermedades de la Médula Ósea/genética , Médula Ósea/patología , Células Germinativas/patología , Mutación/genética , Proteína p53 Supresora de Tumor/genética , Adolescente , Adulto , Agammaglobulinemia/genética , Anemia de Diamond-Blackfan/genética , Animales , Preescolar , Eritrocitos/patología , Femenino , Trastornos del Crecimiento/genética , Humanos , Células Madre Pluripotentes Inducidas/patología , Lactante , Recién Nacido , Masculino , Ratones , Persona de Mediana Edad , Adulto Joven , Pez Cebra
4.
Dev Growth Differ ; 63(2): 178-186, 2021 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-33507533

RESUMEN

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.


Asunto(s)
Plaquetas/citología , Células Madre Pluripotentes Inducidas/citología , Técnicas de Cultivo de Célula , Diferenciación Celular , Humanos
5.
Rinsho Ketsueki ; 61(6): 628-633, 2020.
Artículo en Japonés | MEDLINE | ID: mdl-32624536

RESUMEN

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.


Asunto(s)
Plaquetas , Reactores Biológicos , Células Madre Pluripotentes Inducidas , Megacariocitos , Trombopoyesis
7.
Br J Haematol ; 181(6): 791-802, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29741776

RESUMEN

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.


Asunto(s)
Calreticulina/metabolismo , Diferenciación Celular , Células Madre Hematopoyéticas/metabolismo , Células Madre Pluripotentes Inducidas/metabolismo , Megacariocitos/metabolismo , Mutación , Mielopoyesis , Calreticulina/genética , Femenino , Factor de Transcripción GATA1/genética , Factor de Transcripción GATA1/metabolismo , Factor de Transcripción GATA2/genética , Factor de Transcripción GATA2/metabolismo , Células Madre Hematopoyéticas/citología , Humanos , Células Madre Pluripotentes Inducidas/citología , Masculino , Megacariocitos/citología
9.
Nat Commun ; 15(1): 2588, 2024 Mar 22.
Artículo en Inglés | MEDLINE | ID: mdl-38519457

RESUMEN

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.


Asunto(s)
Células Madre Pluripotentes Inducidas , MicroARNs , Humanos , Megacariocitos , Células Madre Pluripotentes Inducidas/metabolismo , Plaquetas/metabolismo , Trombopoyesis/genética , MicroARNs/genética , MicroARNs/metabolismo
10.
ALTEX ; 40(2): 204-216, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-35229878

RESUMEN

Although several in vitro assays that predict the sensitizing potential of chemicals have been developed, none can distinguish between chemical respiratory and skin sensitizers. Recently, we established a new three-dimensional dendritic cell (DC) coculture system consisting of a human airway epithelial cell line, immature DCs derived from human peripheral monocytes, and a human lung fibroblast cell line. In this coculture system, compared to skin sensitizers, respiratory sensitizers showed enhanced mRNA expression in DCs of the key costimulatory molecule OX40 ligand (OX40L), which is important for T helper 2 (Th2) cell differentiation. Herein, we established a new two-step DC/T cell coculture system by adding peripheral allogeneic naïve CD4+ T cells to the DCs stimulated in the DC coculture system. In this DC/T cell coculture system, model respiratory sensitizers, but not skin sensitizers, enhanced mRNA expression of the predominant Th2 marker interleukin-4 (IL-4). To improve the versatility, in place of peripheral monocytes, monocyte-derived proliferating cells called CD14-ML were used in the DC coculture system. As in peripheral monocytes, enhanced mRNA expression of OX40L was induced in CD14-ML by respiratory sensitizers compared to skin sensitizers. When these cell lines were applied to the DC/T cell coculture system with peripheral allogeneic naïve CD4+ T cells, respiratory sensitizers but not skin sensitizers enhanced the mRNA expression of IL-4. Thus, this DC/T cell coculture system may be useful for discriminating between respiratory and skin sensitizers by differential mRNA upregulation of IL-4 in T cells.


Asunto(s)
Técnicas de Cocultivo , Interleucina-4 , Células Th2 , Humanos , Diferenciación Celular , Células Cultivadas , Células Dendríticas , Interleucina-4/metabolismo , Interleucina-4/farmacología , Monocitos , ARN Mensajero/metabolismo , Células Th2/metabolismo
11.
Blood Adv ; 6(23): 6056-6069, 2022 12 13.
Artículo en Inglés | MEDLINE | ID: mdl-36149941

RESUMEN

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.


Asunto(s)
Antígenos de Plaqueta Humana , Trasplante de Células Madre Hematopoyéticas , Células Madre Pluripotentes Inducidas , Trombocitopenia , Animales , Humanos , Conejos , Transfusión de Plaquetas/efectos adversos , Células Madre Pluripotentes Inducidas/metabolismo , Plaquetas/metabolismo , Trombocitopenia/etiología , Trasplante de Células Madre Hematopoyéticas/efectos adversos
12.
Micromachines (Basel) ; 12(10)2021 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-34683304

RESUMEN

We previously proposed a microfluidic bioreactor with glass-Si-glass layers to evaluate the effect of the fluid force on platelet (PLT) production and fabricated a three-dimensional (3D) microchannel by combining grayscale photolithography and deep reactive ion etching. However, a challenge remains in observing the detailed process of PLT production owing to the low visibility of the microfluidic bioreactor. In this paper, we present a transparent microfluidic bioreactor made of cyclo-olefin polymer (COP) with which to observe the process of platelet-like particle (PLP) production under a bright-field, which allows us to obtain image data at a high sampling rate. We succeeded in fabricating the COP microfluidic bioreactor with a 3D microchannel. We investigated the bonding strength of COP-COP layers and confirmed the effectiveness of the microfluidic bioreactor. Results of on-chip PLP production using immortalized megakaryocyte cell lines (imMKCLs) derived from human-induced pluripotent stem cells show that the average total number of produced PLPs per imMKCL was 17.6 PLPs/imMKCL, which is comparable to that of our previous glass-Si-glass microfluidic bioreactor (17.4 PLPs/imMKCL). We succeeded in observing PLP production under a bright-field using the presented microfluidic bioreactor and confirmed that PLP fragmented in a narrow area of proplatelet-like protrusions.

13.
Stem Cell Res ; 53: 102287, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33813173

RESUMEN

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.


Asunto(s)
Trasplante de Células Madre Hematopoyéticas , Células Madre Pluripotentes , Humanos , Integrina beta1 , Laminina , beta Catenina/genética
14.
Stem Cell Reports ; 16(12): 2861-2870, 2021 12 14.
Artículo en Inglés | MEDLINE | ID: mdl-34861163

RESUMEN

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.


Asunto(s)
Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Silenciador del Gen , Células Madre Pluripotentes Inducidas/metabolismo , Células Progenitoras de Megacariocitos/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Plaquetas/metabolismo , Línea Celular , Proliferación Celular , Células Clonales , Técnicas de Silenciamiento del Gen , Células HEK293 , Humanos , Complejo Represivo Polycomb 1/metabolismo , Proteínas Proto-Oncogénicas c-myc/metabolismo , Regulación hacia Arriba , Proteína bcl-X/metabolismo
15.
Inflamm Regen ; 40(1): 30, 2020 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-33292717

RESUMEN

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.
Artículo en Inglés | MEDLINE | ID: mdl-31883921

RESUMEN

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.


Asunto(s)
Plaquetas/citología , Plaquetas/metabolismo , Diferenciación Celular , Antígenos de Histocompatibilidad Clase I/inmunología , Células Madre Pluripotentes Inducidas/citología , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo , Animales , Citotoxicidad Inmunológica/genética , Citotoxicidad Inmunológica/inmunología , Técnicas de Inactivación de Genes , Antígenos de Histocompatibilidad Clase I/genética , Humanos , Activación de Linfocitos/genética , Activación de Linfocitos/inmunología , Ratones , Ratones Noqueados , Microglobulina beta-2/deficiencia , Microglobulina beta-2/genética
17.
Blood Adv ; 1(7): 468-476, 2017 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-29296963

RESUMEN

Signaling by thrombopoietin (TPO) in complex with its receptor, c-MPL, is critical for hematopoietic stem cell (HSC) homeostasis and platelet generation. Here we show that TA-316, a novel chemically synthesized c-MPL agonist (CMA), is useful for ex vivo platelet generation from human-induced pluripotent stem (iPS) cell-derived immortalized megakaryocyte progenitor cell lines (imMKCLs). Moreover, the generation is clinically applicable, because self-renewal expansion and platelet release is tightly controllable. TA-316 but not eltrombopag, another CMA, promoted both the self-renewal and maturation of imMKCLs, leading to more than a twofold higher platelet production than that achieved with recombinant human TPO (rhTPO). Interestingly, TA-316 seemed to favor MK-biased differentiation from bone marrow CD34+ HSC/progenitors and imMKCLs through the upregulation of vascular endothelial growth factor A and fibroblast growth factor 2. This result suggests TA-316 could facilitate the development of an efficient and useful system to expand platelets from imMKCLs.

18.
Stem Cells Transl Med ; 6(3): 720-730, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-28297575

RESUMEN

Donor-independent platelet concentrates for transfusion can be produced in vitro from induced pluripotent stem cells (iPSCs). However, culture at 37°C induces ectodomain shedding on platelets of glycoprotein Ibα (GPIbα), the von Willebrand factor receptor critical for adhesive function and platelet lifetime in vivo, through temperature-dependent activation of a disintegrin and metalloproteinase 17 (ADAM17). The shedding can be suppressed by using inhibitors of panmetalloproteinases and possibly of the upstream regulator p38 mitogen-activated protein kinase (p38 MAPK), but residues of these inhibitors in the final platelet products may be accompanied by harmful risks that prevent clinical application. Here, we optimized the culture conditions for generating human iPSC-derived GPIbα+ platelets, focusing on culture temperature and additives, by comparing a new and safe selective ADAM17 inhibitor, KP-457, with previous inhibitors. Because cultivation at 24°C (at which conventional platelet concentrates are stored) markedly diminished the yield of platelets with high expression of platelet receptors, 37°C was requisite for normal platelet production from iPSCs. KP-457 blocked GPIbα shedding from iPSC platelets at a lower half-maximal inhibitory concentration than panmetalloproteinase inhibitor GM-6001, whereas p38 MAPK inhibitors did not. iPSC platelets generated in the presence of KP-457 exhibited improved GPIbα-dependent aggregation not inferior to human fresh platelets. A thrombus formation model using immunodeficient mice after platelet transfusion revealed that iPSC platelets generated with KP-457 exerted better hemostatic function in vivo. Our findings suggest that KP-457, unlike GM-6001 or p38 MAPK inhibitors, effectively enhances the production of functional human iPSC-derived platelets at 37°C, which is an important step toward their clinical application. Stem Cells Translational Medicine 2017;6:720-730.


Asunto(s)
Proteína ADAM17/antagonistas & inhibidores , Plaquetas/metabolismo , Células Madre Pluripotentes Inducidas/citología , Complejo GPIb-IX de Glicoproteína Plaquetaria/metabolismo , Proteína ADAM17/metabolismo , Envejecimiento/metabolismo , Plaquetas/efectos de los fármacos , Plaquetas/ultraestructura , Carbonil Cianuro m-Clorofenil Hidrazona/farmacología , Células Cultivadas , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/efectos de los fármacos , Células Madre Hematopoyéticas/metabolismo , Hemostasis/efectos de los fármacos , Humanos , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Células Madre Pluripotentes Inducidas/ultraestructura , Megacariocitos/efectos de los fármacos , Megacariocitos/metabolismo , Temperatura , Trombopoyesis/efectos de los fármacos
19.
Reprod Med Biol ; 4(2): 169-178, 2005 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-29699220

RESUMEN

Aim: In most mammalian fertilization, the sperm introduces the centrosome, which acts as a microtubule organizing center (MTOC) and is essential for pronuclear movement. In rabbit fertilization, biparental centrosomal contribution in microtubule organization has been suggested. Methods: To reveal the function and inheritance of the centrosome during rabbit fertilization, we compared microtubule organization and early embryonal development following intracytoplasmic sperm injection (ICSI) with and without sperm centrosome. Sperm centrosome was removed by sonication, and the isolated sperm head was injected by a Piezo-driven micromanipulator. Samples were studied by light microscope after immunocytological stain. Results: The sperm aster formation was observed 2-3 h after ICSI with intact sperm. In contrast, microtubules were organized between the male and female pronucleus without a nucleation site in the eggs after ICSI with an isolated sperm head. In the late pronuclear stage following ICSI with an isolated sperm head, microtubule organization was the same as in late pronuclear stage eggs after intact sperm injection. The first mitotic spindle was organized in eggs following ICSI with an isolated sperm head, as observed in eggs following ICSI with an intact sperm. Conclusions: These results indicate that the MTOC is in oocyte cytoplasm during fertilization and fulfils the function when the sperm centrosome is absent. (Reprod Med Biol 2005; 4: 169-178).

20.
Nihon Hinyokika Gakkai Zasshi ; 94(7): 689-92, 2003 Nov.
Artículo en Japonés | MEDLINE | ID: mdl-14672000

RESUMEN

A case of haemangiopericytoma of right spermatic cord is reported. A 50-year old male presented with a month-lasting painless swelling of right scrotum. Ultrasonography revealed a soft tissue mass in right spermatic cord, which was about 3.5 cm in diameter. Right high inguinal orchiectomy was performed. The tumor was solid, smooth surfaced and well circumscribed. Histologically, the tumor had many capillary vessels and short spindle calls around the vessels. On the silver impregnation, argyrophil fibers surrounded the tumor cells. Immunohistologically, tumor cells were positive for CD34 antigen and negative for factor VIII antigen. The mitotic rate was 2 per highpower field. Accordingly, this tumor was diagnosed as benign haemangiopericytoma. The patient is doing well without any sign of recurrence, as of 30 months post-operatively. Haemangiopericytoma is a rare neoplasm of pericyte origin. It commonly occurs in retroperitoneum and lower extremities. To our knowledge, only two cases of malignant haemangiopericytoma of the spermatic cord was reported, and this case is the first benign case of the spermatic cord.


Asunto(s)
Neoplasias de los Genitales Masculinos/etiología , Hemangiopericitoma/etiología , Cordón Espermático , Neoplasias de los Genitales Masculinos/cirugía , Hemangiopericitoma/cirugía , Humanos , Masculino , Persona de Mediana Edad , Cordón Espermático/cirugía
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA