Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Cell ; 174(3): 636-648.e18, 2018 07 26.
Artigo em Inglês | MEDLINE | ID: mdl-30017246

RESUMO

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.


Assuntos
Plaquetas/metabolismo , Técnicas de Cultura de Células/métodos , Trombopoese/fisiologia , Reatores Biológicos , Técnicas de Cultura de Células/instrumentação , Humanos , Hidrodinâmica , Células-Tronco Pluripotentes Induzidas/metabolismo , Megacariócitos/metabolismo , Megacariócitos/fisiologia
2.
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
3.
Immunity ; 39(1): 123-35, 2013 Jul 25.
Artigo em Inglês | MEDLINE | ID: mdl-23871209

RESUMO

The NLRP3 inflammasome plays a major role in innate immune responses by activating caspase-1, resulting in secretion of interleukin-18 (IL-18) and IL-1ß. Although cytosolic double-stranded RNA (dsRNA) and bacterial RNA are known to activate the NLRP3 inflammasome, the upstream sensor is unknown. We investigated the potential function of DExD/H-box RNA helicase family members (previously shown to sense cytosolic DNA and RNA to induce type 1 interferon responses) in RNA-induced NLRP3 inflammasome activation. Among the helicase family members tested, we found that targeting of DHX33 expression by short hairpin RNA efficiently blocked the activation of caspase-1 and secretion of IL-18 and IL-1ß in human macrophages that were activated by cytosolic poly I:C, reoviral RNA, or bacterial RNA. DHX33 bound dsRNA via the helicase C domain. DHX33 interacted with NLRP3 and formed the inflammasome complex following stimulation with RNA. We therefore identified DHX33 as a cytosolic RNA sensor that activates the NLRP3 inflammasome.


Assuntos
Proteínas de Transporte/imunologia , RNA Helicases DEAD-box/imunologia , Inflamassomos/imunologia , Macrófagos/imunologia , RNA/imunologia , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Caspase 1/imunologia , Caspase 1/metabolismo , Linhagem Celular , Citosol/imunologia , Citosol/metabolismo , RNA Helicases DEAD-box/genética , RNA Helicases DEAD-box/metabolismo , Expressão Gênica/imunologia , Células HEK293 , Humanos , Immunoblotting , Inflamassomos/genética , Inflamassomos/metabolismo , Interleucina-18/imunologia , Interleucina-18/metabolismo , Interleucina-1beta/imunologia , Interleucina-1beta/metabolismo , Ativação de Macrófagos/imunologia , Macrófagos/metabolismo , Microscopia Confocal , Proteína 3 que Contém Domínio de Pirina da Família NLR , Poli I-C/imunologia , Ligação Proteica/imunologia , RNA/genética , RNA/metabolismo , Interferência de RNA , RNA Bacteriano/imunologia , RNA de Cadeia Dupla/genética , RNA de Cadeia Dupla/imunologia , RNA de Cadeia Dupla/metabolismo , RNA Viral/imunologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa
4.
Mol Ther ; 29(2): 762-774, 2021 02 03.
Artigo em Inglês | MEDLINE | ID: mdl-33038943

RESUMO

Oncolytic herpes simplex virus type 1 (HSV-1) has been investigated to expand its application to various malignancies. Because hematopoietic cells are resistant to HSV-1, its application to hematological malignancies has been rare. Here, we show that the third generation oncolytic HSV-1, T-01, infected and killed 18 of 26 human cell lines and 8 of 15 primary cells derived from various lineages of hematological malignancies. T-01 replicated at low levels in the cell lines. Viral entry and the oncolytic effect were positively correlated with the expression level of nectin-1 and to a lesser extent 3-O-sulfated heparan sulfate, receptors for glycoprotein D of HSV-1, on tumor cells. Transfection of nectin-1 into nectin-1-negative tumor cells made them susceptible to T-01. The oncolytic effects did not appear to correlate with the expression or phosphorylation of antiviral molecules in the cyclic GMP-AMP (cGAS)-stimulator of interferon genes (STING) and PKR-eIF2α pathways. In an immunocompetent mouse model, intratumoral injection of T-01 into lymphoma induced regression of injected, as well as non-injected, contralateral tumors accompanied by abundant infiltration of antigen-specific CD8+ T cells. These data suggest that intratumoral injection of oncolytic HSV-1 may be applicable to systemic hematological malignancies. Nectin-1 expression may be the most useful biomarker for optimal efficacy.


Assuntos
Terapia Genética , Vetores Genéticos/genética , Neoplasias Hematológicas/genética , Neoplasias Hematológicas/terapia , Herpesvirus Humano 1/genética , Terapia Viral Oncolítica , Vírus Oncolíticos/genética , Animais , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Células Cultivadas , Terapia Genética/métodos , Vetores Genéticos/administração & dosagem , Humanos , Terapia Viral Oncolítica/métodos , Transgenes
5.
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
6.
Rinsho Ketsueki ; 63(10): 1430-1439, 2022.
Artigo em Japonês | MEDLINE | ID: mdl-36351652

RESUMO

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.


Assuntos
COVID-19 , Células-Tronco Pluripotentes Induzidas , Trombocitopenia , Humanos , Plaquetas/metabolismo , Pandemias , Transfusão de Plaquetas
7.
Br J Haematol ; 192(2): 343-353, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33216988

RESUMO

Oncolytic viruses exert an anti-tumour effect through two mechanisms: direct oncolytic and indirect immune-mediated mechanisms. Although oncolytic herpes simplex virus type 1 (HSV-1) has been approved for melanoma treatment and is being examined for its applicability to a broad spectrum of malignancies, it is not known whether it has an anti-myeloma effect. In the present study, we show that the third-generation oncolytic HSV-1, T-01, had a direct oncolytic effect on five of six human myeloma cell lines in vitro. The anti-tumour effect was enhanced in the presence of peripheral blood mononuclear cells (PBMCs) from healthy individuals and, to a lesser extent, from patients with myeloma. The enhancing effect of PBMCs was abrogated by blocking type I interferons (IFNs) or by depleting plasmacytoid dendritic cells (pDCs) or natural killer (NK) cells, suggesting that pDC-derived type I IFNs and NK cells dominated the anti-tumour effect. Furthermore, the combination of T-01 and lenalidomide exhibited enhanced cytotoxicity, and the triple combination of T-01, lenalidomide and IFN-α had a maximal effect. These data indicate that oncolytic HSV-1 represents a viable therapy for plasma cell neoplasms through direct oncolysis and immune activation governed by pDCs and NK cells. Lenalidomide is likely to augment the anti-myeloma effect of HSV-1.


Assuntos
Herpesvirus Humano 1/imunologia , Fatores Imunológicos/uso terapêutico , Lenalidomida/uso terapêutico , Neoplasias de Plasmócitos/terapia , Terapia Viral Oncolítica , Vírus Oncolíticos/imunologia , Idoso , Animais , Linhagem Celular Tumoral , Feminino , Humanos , Interferon-alfa/uso terapêutico , Masculino , Camundongos SCID , Neoplasias de Plasmócitos/imunologia , Neoplasias de Plasmócitos/patologia
8.
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
10.
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
11.
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
12.
Proc Natl Acad Sci U S A ; 111(21): 7747-52, 2014 May 27.
Artigo em Inglês | MEDLINE | ID: mdl-24821782

RESUMO

The recognition of cytoplasmic nucleic acid is critical for innate immune responses against microbial infection and is responsible for autoimmunity induced by dead cells. Here, we report the identification of a unique cytosolic nucleic acid cosensor in human airway epithelial cells and fibroblasts: DEAH (Asp-Glu-Ala-His) box polypeptide 29 (DHX29), a member of the DExD/H (Asp-Glu-x-Asp/His)-box helicase family. Knocking down DHX29 by siRNA attenuated the ability of cells to mount type I IFN and IL-6 in response to cytosolic nucleic acids and various viruses by blocking the activation of interferon regulatory factor 3 and NF-κB-p65. The cytosolic nucleic acid sensing by DHX29 in human epithelial cells and fibroblasts is independent of stimulator of interferon genes but is dependent on retinoic acid-inducible gene 1 (RIG-I) and mitochondrial antiviral signaling protein (MAVS). DHX29 binds directly to nucleic acids and interacts with RIG-I and MAVS through its helicase 1 domain, activating the RIG-I-MAVS-dependent cytosolic nucleic acid response. These results suggest that DHX29 is a cytosolic nucleic acid cosensor that triggers RIG-I/MAVS-dependent signaling pathways. This study will have important implications in drug and vaccine design for control of viral infections and viral-induced pathology in the airway.


Assuntos
Citosol/metabolismo , RNA Helicases DEAD-box/metabolismo , Imunidade Inata/imunologia , Ácidos Nucleicos/metabolismo , RNA Helicases/metabolismo , Mucosa Respiratória/metabolismo , Viroses/imunologia , Proteína DEAD-box 58 , DNA Complementar , Humanos , Ácidos Nucleicos/genética , Plasmídeos/genética , Interferência de RNA , Reação em Cadeia da Polimerase em Tempo Real , Receptores Imunológicos , Análise de Sequência de DNA , Vírus/genética
13.
Proc Natl Acad Sci U S A ; 111(45): 16059-64, 2014 Nov 11.
Artigo em Inglês | MEDLINE | ID: mdl-25355909

RESUMO

Inflammasomes are multiprotein platforms that activate caspase-1, which leads to the processing and secretion of the proinflammatory cytokines IL-1ß and IL-18. Previous studies demonstrated that bacterial RNAs activate the nucleotide-binding domain, leucine-rich-repeat-containing family, pyrin domain-containing 3 (NLRP3) inflammasome in both human and murine macrophages. Interestingly, only mRNA, but neither tRNA nor rRNAs, derived from bacteria could activate the murine Nlrp3 inflammasome. Here, we report that all three types of bacterially derived RNA (mRNA, tRNA, and rRNAs) were capable of activating the NLRP3 inflammasome in human macrophages. Bacterial RNA's 5'-end triphosphate moieties, secondary structure, and double-stranded structure were dispensable; small fragments of bacterial RNA were sufficient to activate the inflammasome. In addition, we also found that 20-guanosine ssRNA can activate the NLRP3 inflammasome in human macrophages but not in murine macrophages. Therefore, human and murine macrophages may have evolved to recognize bacterial cytosolic RNA differently during bacterial infections.


Assuntos
Proteínas de Transporte/imunologia , Inflamassomos/imunologia , Macrófagos/imunologia , RNA Bacteriano/imunologia , RNA Mensageiro/imunologia , Animais , Linhagem Celular Tumoral , Humanos , Interleucina-18/imunologia , Interleucina-1beta/imunologia , Macrófagos/citologia , Camundongos , Proteína 3 que Contém Domínio de Pirina da Família NLR , Especificidade da Espécie
14.
Rinsho Ketsueki ; 58(10): 2150-2159, 2017.
Artigo em Japonês | MEDLINE | ID: mdl-28978860

RESUMO

Blood products derived from iPS cells have been pursued as a blood donor-independent and genetically manipulative measure to complement or alternate current transfusion products. Erythrocytes and platelets are anucleate blood cells that are indispensable for oxygen delivery and hemostasis, respectively. Consequently, blood transfusions have been clinically established to treat severe anemia and thrombocytopenia. However, current blood products exhibit issues with regard to supply-demand imbalance and alloimmune responses and infections, and they also face a future shortage of donors in aging societies. While the production of erythrocytes from iPS cells has challenges to overcome, such as their differentiation into an adult-type phenotype and scalable production, platelet products are qualitatively and quantitatively approaching a clinically applicable level owing to advances in expandable megakaryocyte (MK) lines, platelet-producing bioreactors, and novel reagents. Currently, the establishment of guidelines that assure the quality of iPSC-derived blood products for clinical application is in progress. Considering the minimal risk of tumorigenicity and the expected significant demand of such products, the ex vivo production of iPSC-derived blood cells can be expected to lead iPSC-based regenerative medicine to become common clinical practice.


Assuntos
Células-Tronco Pluripotentes Induzidas , Guias de Prática Clínica como Assunto , Plaquetas/efeitos dos fármacos , Desenho de Fármacos , Humanos , Inibidores da Agregação Plaquetária/uso terapêutico , Trombopoese
16.
J Immunol ; 193(2): 627-34, 2014 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-24928999

RESUMO

Accumulating evidence suggests elements within tumors induce exhaustion of effector T cells and infiltration of immunosuppressive regulatory T cells (Tregs), thus preventing the development of durable antitumor immunity. Therefore, the discovery of agents that simultaneously block Treg suppressive function and reinvigorate effector function of lymphocytes is key to the development of effective cancer immunotherapy. Previous studies have shown that TLR ligands (TLRLs) could modulate the function of these T cell targets; however, those studies relied on cell-free or accessory cell-based assay systems that do not accurately reflect in vivo responses. In contrast, we used a human PBMC-based proliferation assay system to simultaneously monitor the effect of TLRLs on T cells (CD4(+), CD8(+), Tregs), B cells, and NK cells, which gave different and even conflicting results. We found that the TLR7/8L:CL097 could simultaneously activate CD8(+) T cells, B cells, and NK cells plus block Treg suppression of T cells and B cells. The TLRLs TLR1/2L:Pam3CSK4, TLR5L:flagellin, TLR4L:LPS, and TLR8/7L:CL075 also blocked Treg suppression of CD4(+) or CD8(+) T cell proliferation, but not B cell proliferation. Besides CL097, TLR2L:PGN, CL075, and TLR9L:CpG-A, CpG-B, and CpG-C) were strong activators of NK cells. Importantly, we found that Pam3CSK4 could: 1) activate CD4(+) T cell proliferation, 2) inhibit the expansion of IL-10(+) naturally occurring FOXP3(+) Tregs and induction of IL-10(+) CD4(+) Tregs (IL-10-producing type 1 Treg), and 3) block naturally occurring FOXP3(+) Tregs suppressive function. Our results suggest these agents could serve as adjuvants to enhance the efficacy of current immunotherapeutic strategies in cancer patients.


Assuntos
Leucócitos Mononucleares/imunologia , Ativação Linfocitária/imunologia , Linfócitos T Reguladores/imunologia , Receptores Toll-Like/imunologia , Adulto , Análise de Variância , Linfócitos B/efeitos dos fármacos , Linfócitos B/imunologia , Linfócitos B/metabolismo , Linfócitos T CD4-Positivos/efeitos dos fármacos , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/metabolismo , Linfócitos T CD8-Positivos/efeitos dos fármacos , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Flagelina/farmacologia , Fatores de Transcrição Forkhead/imunologia , Fatores de Transcrição Forkhead/metabolismo , Humanos , Imidazóis/farmacologia , Interferon gama/imunologia , Interferon gama/metabolismo , Interleucina-10/imunologia , Interleucina-10/metabolismo , Interleucina-6/imunologia , Interleucina-6/metabolismo , Células Matadoras Naturais/efeitos dos fármacos , Células Matadoras Naturais/imunologia , Células Matadoras Naturais/metabolismo , Leucócitos Mononucleares/efeitos dos fármacos , Leucócitos Mononucleares/metabolismo , Lipopeptídeos/farmacologia , Lipopolissacarídeos/farmacologia , Ativação Linfocitária/efeitos dos fármacos , Quinolinas/farmacologia , Linfócitos T Reguladores/efeitos dos fármacos , Linfócitos T Reguladores/metabolismo , Tiazóis/farmacologia , Receptores Toll-Like/agonistas , Receptores Toll-Like/metabolismo , Fator de Necrose Tumoral alfa/imunologia , Fator de Necrose Tumoral alfa/metabolismo
17.
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
18.
Int J Hematol ; 117(3): 349-355, 2023 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-36574167

RESUMO

Platelet transfusion is a common clinical approach to providing platelets to patients suffering from thrombocytopenia or other ailments that require an additional platelet source. However, a stable supply of platelet products is challenged by aging societies, pandemics, and other factors. Many groups have made extensive efforts toward the in vitro generation of platelets for clinical application. We established immortalized megakaryocyte progenitor cell lines (imMKCLs) from human induced pluripotent stem cells (iPSCs) and achieved clinical-scale manufacturing of iPSC-derived platelets (iPSC-PLTs) from them by identifying turbulent flow as a key physical condition. We later completed the iPLAT1 study, the first-in-human clinical trial using autologous iPSC-PLTs. This review summarizes current findings on the ex vivo generation of iPSC-PLTs that led to the iPLAT1 study and beyond. We also discuss new insights regarding the heterogeneity of megakaryocytes and the implications for the ex vivo generation of iPSC-PLTs.


Assuntos
Plaquetas , Células-Tronco Pluripotentes Induzidas , Humanos , Plaquetas/metabolismo , Células-Tronco Pluripotentes Induzidas/metabolismo , Técnicas de Cultura de Células , Megacariócitos/metabolismo , Transfusão de Plaquetas
19.
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
20.
Clin Case Rep ; 9(3): 1383-1386, 2021 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-33768850

RESUMO

A 44-year-old male Japanese was admitted for further post-remission treatments for acute myeloid leukemia. He developed a right orbital abscess. An isolate of Lomentospora prolificans was obtained from the lesion, and orbital biopsy also revealed the presence of Aspergillus fumigatus. This fatal case involved a concurrent dual fungal infection.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA