1
|
Li T, Xing HM, Qian HD, Gao Q, Xu SL, Ma H, Chi ZL. Small extracellular vesicles derived from human induced pluripotent stem cell-differentiated neural progenitor cells mitigate retinal ganglion cell degeneration in a mouse model of optic nerve injury. Neural Regen Res 2025; 20:587-597. [PMID: 38819069 PMCID: PMC11317950 DOI: 10.4103/nrr.nrr-d-23-01414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 11/14/2023] [Accepted: 12/29/2023] [Indexed: 06/01/2024] Open
Abstract
JOURNAL/nrgr/04.03/01300535-202502000-00034/figure1/v/2024-05-28T214302Z/r/image-tiff Several studies have found that transplantation of neural progenitor cells (NPCs) promotes the survival of injured neurons. However, a poor integration rate and high risk of tumorigenicity after cell transplantation limits their clinical application. Small extracellular vesicles (sEVs) contain bioactive molecules for neuronal protection and regeneration. Previous studies have shown that stem/progenitor cell-derived sEVs can promote neuronal survival and recovery of neurological function in neurodegenerative eye diseases and other eye diseases. In this study, we intravitreally transplanted sEVs derived from human induced pluripotent stem cells (hiPSCs) and hiPSCs-differentiated NPCs (hiPSC-NPC) in a mouse model of optic nerve crush. Our results show that these intravitreally injected sEVs were ingested by retinal cells, especially those localized in the ganglion cell layer. Treatment with hiPSC-NPC-derived sEVs mitigated optic nerve crush-induced retinal ganglion cell degeneration, and regulated the retinal microenvironment by inhibiting excessive activation of microglia. Component analysis further revealed that hiPSC-NPC derived sEVs transported neuroprotective and anti-inflammatory miRNA cargos to target cells, which had protective effects on RGCs after optic nerve injury. These findings suggest that sEVs derived from hiPSC-NPC are a promising cell-free therapeutic strategy for optic neuropathy.
Collapse
Affiliation(s)
- Tong Li
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Hui-Min Xing
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Hai-Dong Qian
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Qiao Gao
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Sheng-Lan Xu
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Hua Ma
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Zai-Long Chi
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| |
Collapse
|
2
|
Chen X, Zhao Y, Lv Y, Xie J. Immunological platelet transfusion refractoriness: current insights from mechanisms to therapeutics. Platelets 2024; 35:2306983. [PMID: 38314765 DOI: 10.1080/09537104.2024.2306983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Accepted: 01/14/2024] [Indexed: 02/07/2024]
Abstract
Although there have been tremendous improvements in the production and storage of platelets, platelet transfusion refractoriness (PTR) remains a serious clinical issue that may lead to various severe adverse events. The burden of supplying platelets is worsened by rising market demand and limited donor pools of compatible platelets. Antibodies against platelet antigens are known to activate platelets through FcγR-dependent or complement-activated channels, thereby rapidly eliminating foreign platelets. Recently, other mechanisms of platelet clearance have been reported. The current treatment strategy for PTR is to select appropriate and compatible platelets; however, this necessitates a sizable donor pool and technical assistance for costly testing. Consolidation of these mechanisms should be of critical significance in providing insight to establish novel therapeutics to target immunological platelet refractoriness. Therefore, the purposes of this review were to explore the modulation of the immune system over the activation and elimination of allogeneic platelets and to summarize the development of alternative approaches for treating and avoiding alloimmunization to human leukocyte antigen or human platelet antigen in PTR.
Collapse
Affiliation(s)
- Xiaoyu Chen
- Department of Blood Transfusion, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Yuhong Zhao
- Department of Blood Transfusion, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Yan Lv
- Department of Blood Transfusion, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Jue Xie
- Department of Blood Transfusion, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| |
Collapse
|
3
|
Leung J, Primbetova A, Strong C, Hay BN, Hsu HH, Hagner A, Foster LJ, Devine D, Cullis PR, Zandstra PW, Kastrup CJ. Genetic engineering of megakaryocytes from blood progenitor cells using messenger RNA lipid nanoparticles. J Thromb Haemost 2024:S1538-7836(24)00553-1. [PMID: 39341369 DOI: 10.1016/j.jtha.2024.09.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 09/09/2024] [Accepted: 09/13/2024] [Indexed: 10/01/2024]
Abstract
BACKGROUND Platelets are an essential component of hemorrhage control and management, and engineering platelets to express therapeutic proteins could expand their use as a cell therapy. Genetically engineered platelets can be achieved by modifying the platelet precursor cells, megakaryocytes (MKs). Current strategies include transfecting MK progenitors ex vivo with viral vectors harboring lineage-driven transgenes and inducing the production of in vitro modified platelets. The use of viruses, however, poses challenges in clinical implementation, and no methods currently exist to genetically modify MKs with nonviral techniques. Lipid nanoparticles (LNPs) are a nonviral delivery system that could enable a facile strategy to modify MKs with a variety of nucleic acid payloads. OBJECTIVES To investigate whether LNPs can transfect cultured hematopoietic stem/progenitor cell-derived MKs to express exogenous proteins and induce functional changes. METHODS MK and MK progenitors differentiated from cord blood-derived hematopoietic stem/progenitor cells were treated with LNP formulations containing messenger RNA and resembling the clinically approved LNP formulations. Transfection efficiency was assessed through flow cytometry by expression of enhanced green fluorescent protein. Functional changes to the MKs were assessed through rotational thromboelastometry by expression of exogenous coagulation factor (F)VII, a representative physiologically relevant protein. RESULTS LNPs enabled transfection efficiencies of 99% in MKs and did not impair MK maturation, viability, and morphology. MKs engineered to express exogenous FVII decreased clotting time in FVII-deficient plasma following clot initiation. CONCLUSION This approach provides an easy-to-use modular platform to genetically modify MK and MK progenitors, which can be potentially extended to producing genetically modified cultured platelets.
Collapse
Affiliation(s)
- Jerry Leung
- Michael Smith Laboratories, University of British Columbia, Vancouver, British Columbia, Canada; Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, British Columbia, Canada; Centre for Blood Research, University of British Columbia, Vancouver, British Columbia, Canada; NanoMedicines Research Group, Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Asel Primbetova
- School of Biomedical Engineering, University of British Columbia, Vancouver, British Columbia, Canada
| | - Colton Strong
- Michael Smith Laboratories, University of British Columbia, Vancouver, British Columbia, Canada; Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, British Columbia, Canada; Centre for Blood Research, University of British Columbia, Vancouver, British Columbia, Canada
| | - Brenna N Hay
- Michael Smith Laboratories, University of British Columbia, Vancouver, British Columbia, Canada; Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Han Hsuan Hsu
- School of Biomedical Engineering, University of British Columbia, Vancouver, British Columbia, Canada
| | - Andrew Hagner
- School of Biomedical Engineering, University of British Columbia, Vancouver, British Columbia, Canada
| | - Leonard J Foster
- Michael Smith Laboratories, University of British Columbia, Vancouver, British Columbia, Canada; Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, British Columbia, Canada; Centre for Blood Research, University of British Columbia, Vancouver, British Columbia, Canada
| | - Dana Devine
- Centre for Blood Research, University of British Columbia, Vancouver, British Columbia, Canada; Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Pieter R Cullis
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, British Columbia, Canada; NanoMedicines Research Group, Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Peter W Zandstra
- Michael Smith Laboratories, University of British Columbia, Vancouver, British Columbia, Canada; School of Biomedical Engineering, University of British Columbia, Vancouver, British Columbia, Canada.
| | - Christian J Kastrup
- Michael Smith Laboratories, University of British Columbia, Vancouver, British Columbia, Canada; Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, British Columbia, Canada; Centre for Blood Research, University of British Columbia, Vancouver, British Columbia, Canada; Versiti Blood Research Institute, Milwaukee, Wisconsin, USA; Departments of Surgery, Biochemistry, Biomedical Engineering, and Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA.
| |
Collapse
|
4
|
Poncz M, Zaitsev SV, Ahn H, Kowalska MA, Bdeir K, Dergilev KV, Ivanciu L, Camire RM, Cines DB, Stepanova V. Packaging of supplemented urokinase into alpha granules of in vitro-grown megakaryocytes for targeted nascent clot lysis. Blood Adv 2024; 8:3798-3809. [PMID: 38805575 PMCID: PMC11298819 DOI: 10.1182/bloodadvances.2024012835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 05/16/2024] [Accepted: 05/18/2024] [Indexed: 05/30/2024] Open
Abstract
ABSTRACT Fibrinolytics delivered into the general circulation lack selectivity for nascent thrombi, reducing efficacy and increasing the risk of bleeding. Urokinase-type plasminogen activator (uPA) transgenically expressed within murine platelets provided targeted thromboprophylaxis without causing bleeding but is not clinically feasible. Recent advances in generating megakaryocytes prompted us to develop a potentially clinically relevant means to produce "antithrombotic" platelets from CD34+ hematopoietic stem cell-derived in vitro-grown megakaryocytes. CD34+ megakaryocytes internalize and store in alpha granules (α-granules) single-chain uPA (scuPA) and a plasmin-resistant thrombin-activatable variant (uPAT). Both uPAs colocalized with internalized factor V (FV), fibrinogen and plasminogen, low-density lipoprotein receptor-related protein 1 (LRP1), and interferon-induced transmembrane protein 3, but not with endogenous von Willebrand factor (VWF). Endocytosis of uPA by CD34+ megakaryocytes was mediated, in part, via LRP1 and αIIbβ3. scuPA-containing megakaryocytes degraded endocytosed intragranular FV but not endogenous VWF in the presence of internalized plasminogen, whereas uPAT-megakaryocytes did not significantly degrade either protein. We used a carotid artery injury model in nonobese diabetic-severe combined immunodeficiency IL2rγnull (NSG) mice homozygous for VWFR1326H (a mutation switching binding VWF specificity from mouse to human glycoprotein Ibα) to test whether platelets derived from scuPA- or uPAT-megakaryocytes would prevent thrombus formation. NSG/VWFR1326H mice exhibited a lower thrombotic burden after carotid artery injury compared with NSG mice unless infused with human platelets or megakaryocytes, whereas intravenous injection of uPA-megakaryocytes generated sufficient uPA-containing human platelets to lyse nascent thrombi. These studies describe the use of in vitro-generated megakaryocytes as a potential platform for delivering uPA or other ectopic proteins within platelet α-granules to sites of vascular injury.
Collapse
Affiliation(s)
- Mortimer Poncz
- Department of Pediatrics, Children’s Hospital of Philadelphia, Philadelphia, PA
- Department of Pediatrics, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA
| | - Sergei V. Zaitsev
- Department of Pediatrics, Children’s Hospital of Philadelphia, Philadelphia, PA
| | - Hyunsook Ahn
- Department of Pediatrics, Children’s Hospital of Philadelphia, Philadelphia, PA
| | - M. Anna Kowalska
- Department of Pediatrics, Children’s Hospital of Philadelphia, Philadelphia, PA
- Institute of Medical Biology, Polish Academy of Sciences, Lodz, Poland
| | - Khalil Bdeir
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA
| | - Konstantin V. Dergilev
- Institute of Experimental Cardiology, National Medical Research Center of Cardiology named after Academician E.I. Chazov, Moscow, Russia
| | - Lacramioara Ivanciu
- Department of Pediatrics, Children’s Hospital of Philadelphia, Philadelphia, PA
- Department of Pediatrics, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA
| | - Rodney M. Camire
- Department of Pediatrics, Children’s Hospital of Philadelphia, Philadelphia, PA
- Department of Pediatrics, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA
| | - Douglas B. Cines
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA
| | - Victoria Stepanova
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA
| |
Collapse
|
5
|
Kim H, Jarocha D, Johnson I, Ahn H, Hlinka N, French DL, Rauova L, Lee K, Poncz M. Studies of infused megakaryocytes into mice support a "catch-and-release" model of pulmonary-centric thrombopoiesis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.04.597316. [PMID: 38895231 PMCID: PMC11185690 DOI: 10.1101/2024.06.04.597316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
Many aspects of thrombopoiesis, the release of platelets from megakaryocytes (Mks), remain under debate, including where this process occurs. Murine lung in situ -microscopy studies suggested that a significant fraction of circulating platelets were released from lung-entrapped, marrow-derived Mks. We now confirm these in situ studies that endogenous mMks are entrapped in the lungs and show that intravenously infused in vitro -differentiated, mature murine (m) and human (h) Mks are similarly entrapped followed by shedding of their cytoplasm over ∼30 minutes with a peak number of released platelets occurring 1.5-4 hours later. However, while infused Mks from both species shed large intrapulmonary cytoplasmic fragments that underwent further processing into platelet-sized fragments, the two differed: many mMks escaped from and then recycled back to the lungs, while most hMks were enucleated upon first intrapulmonary passage. Infused immature hMks, inflammatory hMks, umbilical cord-blood-derived hMks and immortalized Mk progenitor cell (imMKCL)-derived hMks were also entrapped in the lung of recipient mice, and released their cytoplasm, but did so to different degrees. Intraarterial infused hMks resulted in few Mks being entrapped in tissues other than the lungs and was accompanied by a blunted and delayed rise in circulating human platelets. These studies demonstrate that the lung entraps and processes both circulating Mks and released large cytoplasmic fragments consistent with a recent lung/heart murine study and support a pulmonary-centric "catch-and-release" model of thrombopoiesis. Thus, thrombopoiesis is a drawn-out process with the majority of cytoplasmic processing derived from Mks occurring in the pulmonary bed. Key Points Infused in vitro -differentiated megakaryocytes synchronously release cytoplasmic fragments highly selectively in the pulmonary bed. Large, released megakaryocyte fragments recycle to the lungs, undergo further fission, terminally form platelets.
Collapse
|
6
|
Arai T, Shiga Y, Mukai M, Takayama N, Tashiro S, Tajiri I, Kosaka K, Sato M, Nakamura S, Okamoto H, Kimura S, Inage K, Suzuki-Narita M, Eguchi Y, Orita S, Eto K, Ohtori S. Osteogenic effects and safety of human induced pluripotent stem cell-derived megakaryocytes and platelets produced on a clinical scale. Regen Ther 2024; 26:850-858. [PMID: 39430581 PMCID: PMC11488481 DOI: 10.1016/j.reth.2024.09.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 09/16/2024] [Accepted: 09/25/2024] [Indexed: 10/22/2024] Open
Abstract
Introduction Platelet-rich plasma obtained by centrifuging peripheral blood can promote osteogenesis owing to its abundant growth factors but has drawbacks, including rapid growth factor loss and inconsistent effects depending on donor factors. To overcome these issues, we were the first in the world to use freeze-dried human induced pluripotent stem cell-derived megakaryocytes and platelets (S-FD-iMPs) and found that they have osteogenesis-promoting effects. Since turbulence was found to activate platelet biogenesis and iPS cell-derived platelets can now be produced on a clinical scale by a device called VerMES, this study examined the osteogenesis-promoting effect and safety of clinical-scale FD-iMP (V-FD-iMPs) for future human clinical application. Method We administered either S-FD-iMPs, V-FD-iMPs, or saline along with artificial bone to the lumbar spine of 8-week-old male Sprague-Dawley rats (n = 4 each) and evaluated bone formation by computed tomography (CT) and pathology. Next, we administered V-FD-iMPs or saline along with artificial bone to the lumber spines of 5-week-old male New Zealand White rabbits (n = 4 each) and evaluated the bone formation by CT and pathology. Rats (n = 10) and rabbits (n = 6) that received artificial bone and V-FD-iMPs in the lumbar spine were also observed for 6 months for adverse events, including infection, tumor formation, and death. Results Both V-FD-iMPs and S-FD-iMPs significantly enhanced osteogenesis in the lumber spines of rats in comparison with the controls 8 weeks postoperatively, with no significant differences between them. Furthermore, V-FD-iMPs vigorously promoted osteogenesis in the lumber spines of rabbits 8 weeks postoperatively. In rats and rabbits, V-FD-iMPs showed no adverse effects, including infection, tumor formation, and death, over 6 months. Conclusion These results suggest that V-FD-iMPs promote safe osteogenesis.
Collapse
Affiliation(s)
- Takahito Arai
- Department of Orthopaedic Surgery, Chiba University Graduate School of Medicine, Chiba, 2608670, Japan
| | - Yasuhiro Shiga
- Department of Orthopaedic Surgery, Chiba University Graduate School of Medicine, Chiba, 2608670, Japan
| | - Michiaki Mukai
- Department of Orthopaedic Surgery, Chiba University Graduate School of Medicine, Chiba, 2608670, Japan
| | - Naoya Takayama
- Department of Regenerative Medicine, Chiba University Graduate School of Medicine, Chiba, 2608670, Japan
| | - Susumu Tashiro
- Department of Orthopaedic Surgery, Chiba University Graduate School of Medicine, Chiba, 2608670, Japan
| | - Ikuko Tajiri
- Department of Orthopaedic Surgery, Chiba University Graduate School of Medicine, Chiba, 2608670, Japan
| | - Kentaro Kosaka
- Department of Plastic, Reconstructive, and Aesthetic Surgery, Graduate School of Medicine, Chiba University, Chiba, 2608670, Japan
| | - Masashi Sato
- Department of Orthopaedic Surgery, Eastern Chiba Medical Center, Chiba, 2838686, Japan
| | - Sou Nakamura
- Department of Clinical Application, Center for IPS Cell Research and Application (CiRA), Kyoto University, Kyoto, 6068507, Japan
| | - Haruki Okamoto
- Department of Clinical Application, Center for IPS Cell Research and Application (CiRA), Kyoto University, Kyoto, 6068507, Japan
| | - Seiji Kimura
- Department of Orthopaedic Surgery, Chiba University Graduate School of Medicine, Chiba, 2608670, Japan
| | - Kazuhide Inage
- Department of Orthopaedic Surgery, Chiba University Graduate School of Medicine, Chiba, 2608670, Japan
| | - Miyako Suzuki-Narita
- Department of Bioenvironmental Medicine, Graduate School of Medicine, Chiba University, Chiba, 2608670, Japan
| | - Yawara Eguchi
- Department of Orthopaedic Surgery, Chiba University Graduate School of Medicine, Chiba, 2608670, Japan
| | - Sumihisa Orita
- Center for Frontier Medical Engineering, Chiba University, Chiba, 2638522, Japan
| | - Koji Eto
- Department of Regenerative Medicine, Chiba University Graduate School of Medicine, Chiba, 2608670, Japan
- Department of Clinical Application, Center for IPS Cell Research and Application (CiRA), Kyoto University, Kyoto, 6068507, Japan
| | - Seiji Ohtori
- Department of Orthopaedic Surgery, Chiba University Graduate School of Medicine, Chiba, 2608670, Japan
| |
Collapse
|
7
|
Nakamura Y, Niho S, Shimizu Y. Cell-Based Therapy for Fibrosing Interstitial Lung Diseases, Current Status, and Potential Applications of iPSC-Derived Cells. Cells 2024; 13:893. [PMID: 38891026 PMCID: PMC11172081 DOI: 10.3390/cells13110893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Revised: 05/09/2024] [Accepted: 05/17/2024] [Indexed: 06/20/2024] Open
Abstract
Fibrosing interstitial lung diseases (FILDs), e.g., due to idiopathic pulmonary fibrosis (IPF), are chronic progressive diseases with a poor prognosis. The management of these diseases is challenging and focuses mainly on the suppression of progression with anti-fibrotic drugs. Therefore, novel FILD treatments are needed. In recent years, cell-based therapy with various stem cells has been investigated for FILD, and the use of mesenchymal stem cells (MSCs) has been widely reported and clinical studies are also ongoing. Induced pluripotent stem cells (iPSCs) have also been reported to have an anti-fibrotic effect in FILD; however, these have not been as well studied as MSCs in terms of the mechanisms and side effects. While MSCs show a potent anti-fibrotic effect, the possibility of quality differences between donors and a stable supply in the case of donor shortage or reduced proliferative capacity after cell passaging needs to be considered. The application of iPSC-derived cells has the potential to overcome these problems and may lead to consistent quality of the cell product and stable product supply. This review provides an overview of iPSCs and FILD, followed by the current status of cell-based therapy for FILD, and then discusses the possibilities and perspectives of FILD therapy with iPSC-derived cells.
Collapse
Affiliation(s)
- Yusuke Nakamura
- Department of Pulmonary Medicine and Clinical Immunology, Dokkyo Medical University School of Medicine, Mibu 321-0293, Japan; (Y.N.); (S.N.)
- Center of Regenerative Medicine, Dokkyo Medical University Hospital, Mibu 321-0293, Japan
| | - Seiji Niho
- Department of Pulmonary Medicine and Clinical Immunology, Dokkyo Medical University School of Medicine, Mibu 321-0293, Japan; (Y.N.); (S.N.)
| | - Yasuo Shimizu
- Department of Pulmonary Medicine and Clinical Immunology, Dokkyo Medical University School of Medicine, Mibu 321-0293, Japan; (Y.N.); (S.N.)
- Center of Regenerative Medicine, Dokkyo Medical University Hospital, Mibu 321-0293, Japan
- Respiratory Endoscopy Center, Dokkyo Medical University Hospital, Mibu 321-0293, Japan
| |
Collapse
|
8
|
Luanpitpong S, Tangkiettrakul K, Kang X, Srisook P, Poohadsuan J, Samart P, Klaihmon P, Janan M, Lorthongpanich C, Laowtammathron C, Issaragrisil S. OGT and OGA gene-edited human induced pluripotent stem cells for dissecting the functional roles of O-GlcNAcylation in hematopoiesis. Front Cell Dev Biol 2024; 12:1361943. [PMID: 38752196 PMCID: PMC11094211 DOI: 10.3389/fcell.2024.1361943] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Accepted: 03/14/2024] [Indexed: 05/18/2024] Open
Abstract
Hematopoiesis continues throughout life to produce all types of blood cells from hematopoietic stem cells (HSCs). Metabolic state is a known regulator of HSC self-renewal and differentiation, but whether and how metabolic sensor O-GlcNAcylation, which can be modulated via an inhibition of its cycling enzymes O-GlcNAcase (OGA) and O-GlcNAc transferase (OGT), contributes to hematopoiesis remains largely unknown. Herein, isogenic, single-cell clones of OGA-depleted (OGAi) and OGT-depleted (OGTi) human induced pluripotent stem cells (hiPSCs) were successfully generated from the master hiPSC line MUSIi012-A, which were reprogrammed from CD34+ hematopoietic stem/progenitor cells (HSPCs) containing epigenetic memory. The established OGAi and OGTi hiPSCs exhibiting an increase or decrease in cellular O-GlcNAcylation concomitant with their loss of OGA and OGT, respectively, appeared normal in phenotype and karyotype, and retained pluripotency, although they may favor differentiation toward certain germ lineages. Upon hematopoietic differentiation through mesoderm induction and endothelial-to-hematopoietic transition, we found that OGA inhibition accelerates hiPSC commitment toward HSPCs and that disruption of O-GlcNAc homeostasis affects their commitment toward erythroid lineage. The differentiated HSPCs from all groups were capable of giving rise to all hematopoietic progenitors, thus confirming their functional characteristics. Altogether, the established single-cell clones of OGTi and OGAi hiPSCs represent a valuable platform for further dissecting the roles of O-GlcNAcylation in blood cell development at various stages and lineages of blood cells. The incomplete knockout of OGA and OGT in these hiPSCs makes them susceptible to additional manipulation, i.e., by small molecules, allowing the molecular dynamics studies of O-GlcNAcylation.
Collapse
Affiliation(s)
- Sudjit Luanpitpong
- Siriraj Center of Excellence for Stem Cell Research, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
- Blood Products and Cellular Immunotherapy Research Group, Research Division, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Kantpitchar Tangkiettrakul
- Siriraj Center of Excellence for Stem Cell Research, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
- Blood Products and Cellular Immunotherapy Research Group, Research Division, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Xing Kang
- Siriraj Center of Excellence for Stem Cell Research, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Pimonwan Srisook
- Siriraj Center of Excellence for Stem Cell Research, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Jirarat Poohadsuan
- Siriraj Center of Excellence for Stem Cell Research, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Parinya Samart
- Siriraj Center of Excellence for Stem Cell Research, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Phatchanat Klaihmon
- Siriraj Center of Excellence for Stem Cell Research, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Montira Janan
- Siriraj Center of Excellence for Stem Cell Research, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
- Blood Products and Cellular Immunotherapy Research Group, Research Division, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Chanchao Lorthongpanich
- Siriraj Center of Excellence for Stem Cell Research, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
- Blood Products and Cellular Immunotherapy Research Group, Research Division, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Chuti Laowtammathron
- Siriraj Center of Excellence for Stem Cell Research, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Surapol Issaragrisil
- Siriraj Center of Excellence for Stem Cell Research, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
- Division of Hematology, Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| |
Collapse
|
9
|
Bogomiakova ME, Bogomazova AN, Lagarkova MA. Dysregulation of Immune Tolerance to Autologous iPSCs and Their Differentiated Derivatives. BIOCHEMISTRY. BIOKHIMIIA 2024; 89:799-816. [PMID: 38880643 DOI: 10.1134/s0006297924050031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 12/21/2023] [Accepted: 02/13/2024] [Indexed: 06/18/2024]
Abstract
Induced pluripotent stem cells (iPSCs), capable of differentiating into any cell type, are a promising tool for solving the problem of donor organ shortage. In addition, reprogramming technology makes it possible to obtain a personalized, i.e., patient-specific, cell product transplantation of which should not cause problems related to histocompatibility of the transplanted tissues and organs. At the same time, inconsistent information about the main advantage of autologous iPSC-derivatives - lack of immunogenicity - still casts doubt on the possibility of using such cells beyond immunosuppressive therapy protocols. This review is devoted to immunogenic properties of the syngeneic and autologous iPSCs and their derivatives, as well as to the reasons for dysregulation of their immune tolerance.
Collapse
Affiliation(s)
- Margarita E Bogomiakova
- Federal Research and Clinical Center of Physical-Chemical Medicine, Federal Medical Biological Agency, Moscow, 119435, Russia.
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Federal Research and Clinical Center of Physical-Chemical Medicine, Federal Medical Biological Agency, Moscow, 119435, Russia
| | - Alexandra N Bogomazova
- Federal Research and Clinical Center of Physical-Chemical Medicine, Federal Medical Biological Agency, Moscow, 119435, Russia
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Federal Research and Clinical Center of Physical-Chemical Medicine, Federal Medical Biological Agency, Moscow, 119435, Russia
| | - Maria A Lagarkova
- Federal Research and Clinical Center of Physical-Chemical Medicine, Federal Medical Biological Agency, Moscow, 119435, Russia
- Lomonosov Moscow State University, Moscow, 119991, Russia
| |
Collapse
|
10
|
Chen Y, Li M, Wu Y. The occurrence and development of induced pluripotent stem cells. Front Genet 2024; 15:1389558. [PMID: 38699229 PMCID: PMC11063328 DOI: 10.3389/fgene.2024.1389558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Accepted: 04/08/2024] [Indexed: 05/05/2024] Open
Abstract
The ectopic expression of four transcription factors, Oct3/4, Sox2, Klf4, and c-Myc (OSKM), known as "Yamanaka factors," can reprogram or stimulate the production of induced pluripotent stem cells (iPSCs). Although OSKM is still the gold standard, there are multiple ways to reprogram cells into iPSCs. In recent years, significant progress has been made in improving the efficiency of this technology. Ten years after the first report was published, human pluripotent stem cells have gradually been applied in clinical settings, including disease modeling, cell therapy, new drug development, and cell derivation. Here, we provide a review of the discovery of iPSCs and their applications in disease and development.
Collapse
Affiliation(s)
| | - Meng Li
- Department of Cardiology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Yanqing Wu
- Department of Cardiology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| |
Collapse
|
11
|
Christiansen JR, Kirkeby A. Clinical translation of pluripotent stem cell-based therapies: successes and challenges. Development 2024; 151:dev202067. [PMID: 38564308 PMCID: PMC11057818 DOI: 10.1242/dev.202067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
The translational stem cell research field has progressed immensely in the past decade. Development and refinement of differentiation protocols now allows the generation of a range of cell types, such as pancreatic β-cells and dopaminergic neurons, from human pluripotent stem cells (hPSCs) in an efficient and good manufacturing practice-compliant fashion. This has led to the initiation of several clinical trials using hPSC-derived cells to replace lost or dysfunctional cells, demonstrating evidence of both safety and efficacy. Here, we highlight successes from some of the hPSC-based trials reporting early signs of efficacy and discuss common challenges in clinical translation of cell therapies.
Collapse
Affiliation(s)
- Josefine Rågård Christiansen
- Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), University of Copenhagen, 2200 Copenhagen N, Denmark
| | - Agnete Kirkeby
- Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), University of Copenhagen, 2200 Copenhagen N, Denmark
- Department of Neuroscience, University of Copenhagen, 2200 Copenhagen N, Denmark
- Wallenberg Center for Molecular Medicine, Department of Experimental Medical Science, Lund University, 221 84 Lund, Sweden
| |
Collapse
|
12
|
Chen SJ, Hashimoto K, Fujio K, Hayashi K, Paul SK, Yuzuriha A, Qiu WY, Nakamura E, Kanashiro MA, Kabata M, Nakamura S, Sugimoto N, Kaneda A, Yamamoto T, Saito H, Takayama N, Eto K. A let-7 microRNA-RALB axis links the immune properties of iPSC-derived megakaryocytes with platelet producibility. Nat Commun 2024; 15:2588. [PMID: 38519457 PMCID: PMC10960040 DOI: 10.1038/s41467-024-46605-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Accepted: 03/04/2024] [Indexed: 03/25/2024] Open
Abstract
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.
Collapse
Affiliation(s)
- Si Jing Chen
- Department of Clinical Application, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
- Department of Regenerative Medicine, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Kazuya Hashimoto
- Department of Clinical Application, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Kosuke Fujio
- Department of Clinical Application, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Karin Hayashi
- Department of Life Science Frontiers, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Sudip Kumar Paul
- Department of Regenerative Medicine, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Akinori Yuzuriha
- Department of Clinical Application, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Wei-Yin Qiu
- Department of Clinical Application, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Emiri Nakamura
- Department of Clinical Application, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | | | - Mio Kabata
- Department of Life Science Frontiers, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Sou Nakamura
- Department of Clinical Application, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Naoshi Sugimoto
- Department of Clinical Application, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Atsushi Kaneda
- Department of Molecular Oncology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Takuya Yamamoto
- Department of Life Science Frontiers, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
- Institute for the Advanced Study of Human Biology (WPI-ASHBi), Kyoto University, Kyoto, Japan
- Medical-risk Avoidance Based on iPS Cells Team, RIKEN Center for Advanced Intelligence Project (AIP), Kyoto, Japan
| | - Hirohide Saito
- Department of Life Science Frontiers, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan.
| | - Naoya Takayama
- Department of Regenerative Medicine, Graduate School of Medicine, Chiba University, Chiba, Japan.
| | - Koji Eto
- Department of Clinical Application, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan.
- Department of Regenerative Medicine, Graduate School of Medicine, Chiba University, Chiba, Japan.
| |
Collapse
|
13
|
Kayama A, Eto K. Mass production of iPSC-derived platelets toward the clinical application. Regen Ther 2024; 25:213-219. [PMID: 38260088 PMCID: PMC10801197 DOI: 10.1016/j.reth.2023.12.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 11/06/2023] [Accepted: 12/17/2023] [Indexed: 01/24/2024] Open
Abstract
The ex vivo production of platelets from induced pluripotent cells (iPSCs) may offer a safer and sustainable alternative for transfusions and drug delivery systems (DDS). However, the mass production of the clinically required number of iPSC-derived platelets (iPSC-PLTs) is challenging. Here, we introduce recent technologies for mass production and the first-in-human clinical trial using ex vivo iPSC-PLTs. To this end, we established immortalized megakaryocyte progenitor cell lines (imMKCLs) as an expandable master cell bank (MCB) through the overexpression of c-MYC, BMI1 and BCL-XL, which modulated megakaryopoiesis and thrombopoiesis. We also optimized a culture cocktail for maturation of the imMKCLs by mixing an aryl hydrocarbon receptor (AhR) antagonist, SR1/GNF-316; a Rho-associated protein kinase (ROCK) inhibitor, Y-27632/Y-39983; and a small-molecule compound replacing recombinant thrombopoietin (TPO), TA-316. Finally, we discovered the importance of turbulence on the manufacturing of intact iPSC-PLTs, allowing us to develop a turbulence-based bioreactor, VerMES. Combination of the MCB and VerMES enabled us to produce more than 100 billion iPSC-PLTs, leading to the first-in-human clinical trial. Despite these advancements, many challenges remain before expanding the clinical implementation of this strategy.
Collapse
|
14
|
Hayashi Y, Ohnishi H, Kitano M, Kishimoto Y, Takezawa T, Okuyama H, Yoshimatsu M, Kuwata F, Tada T, Mizuno K, Omori K. Comparative Study of Immunodeficient Rat Strains in Engraftment of Human-Induced Pluripotent Stem Cell-Derived Airway Epithelia. Tissue Eng Part A 2024; 30:144-153. [PMID: 37950719 DOI: 10.1089/ten.tea.2023.0214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2023] Open
Abstract
The airway epithelia (AE) play a role in the clearance of foreign substances through ciliary motility and mucus secreted. We developed an artificial trachea that is made of collagen sponges and polypropylene mesh for the regeneration of the tracheal defect, and it was used for a clinical study. Then, a model in which the luminal surface of an artificial trachea was covered with a human-induced pluripotent stem cell-derived AE (hiPSC-AE) was transplanted into the tracheal defect of nude rats to promote epithelialization. In the future, this model was expected to be applied to research on infectious diseases and drug discovery as a trachea-humanized rat model. However, at present, sufficient engraftment has not been achieved to evaluate functional recovery in transplanted cells. Therefore, this study focused on immunosuppression in recipient rats. Nude rats lack T cell function and are widely used for transplantation experiments; however, more severe immunosuppressed recipients are preferred for xenotransplantation. Several strains of immunodeficient rats were created as rats that exhibit more severe immunodeficiency until now. In this study, to establish a trachea-humanized rat model in which human AE function can be analyzed to improve engraftment efficiency, engraftment efficiency in nude rats and X-linked severe combined immunodeficiency (X-SCID) rats following hiPSC-AE transplantation was compared. In the analysis of the proportion of engrafted cells in total cells at the graft site, the engraftment efficiency of epithelial cells tended to be high in X-SCID rats, although no statistical difference was found between the two groups, whereas the engraftment efficiency of mesenchymal cells was higher in X-SCID rats. Furthermore, the number of immune cells that accumulated in the grafts showed that a pan T cell marker, that is, CD3-positive cells, did not differ between the two strains; however, CD45-positive cells and major histocompatibility complex (MHC) class II-positive cells significantly decreased in X-SCID rats. These results indicate that X-SCID rats are more useful for the transplantation of hiPSC-AE into the tracheae to generate trachea-humanized rat models.
Collapse
Affiliation(s)
- Yasuyuki Hayashi
- Department of Otolaryngology-Head and Neck Surgery, Graduate School of medicine, Kyoto University, Kyoto, Japan
| | - Hiroe Ohnishi
- Department of Otolaryngology-Head and Neck Surgery, Graduate School of medicine, Kyoto University, Kyoto, Japan
| | - Masayuki Kitano
- Department of Otolaryngology-Head and Neck Surgery, Graduate School of medicine, Kyoto University, Kyoto, Japan
| | - Yo Kishimoto
- Department of Otolaryngology-Head and Neck Surgery, Graduate School of medicine, Kyoto University, Kyoto, Japan
| | - Toshiaki Takezawa
- Faculty of Pharmacy, Chiba Institute of Science, Chiba, Japan
- Institute of Agrobiological Sciences, National Agriculture and Food Research Organization, Ibaraki, Japan
| | - Hideaki Okuyama
- Faculty of Medicine and Health Sciences, School of Communication Sciences and Disorders, McGill University, Montreal, Canada
| | - Masayoshi Yoshimatsu
- Department of Otolaryngology-Head and Neck Surgery, Graduate School of medicine, Kyoto University, Kyoto, Japan
- Department of Otolaryngology, Head and Neck Surgery, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan
| | - Fumihiko Kuwata
- Department of Otolaryngology-Head and Neck Surgery, Graduate School of medicine, Kyoto University, Kyoto, Japan
| | - Takeshi Tada
- Center for Inflammation, Immunity and Infection Institute for Biomedical Sciences, Georgia State University, Atlanta, Georgia, USA
| | - Keisuke Mizuno
- Department of Otolaryngology-Head and Neck Surgery, Graduate School of medicine, Kyoto University, Kyoto, Japan
| | - Koichi Omori
- Department of Otolaryngology-Head and Neck Surgery, Graduate School of medicine, Kyoto University, Kyoto, Japan
| |
Collapse
|
15
|
Yamamoto R, Sakakibara R, Kim MH, Fujinaga Y, Kino-Oka M. Growth prolongation of human induced pluripotent stem cell aggregate in three-dimensional suspension culture system by addition of botulinum hemagglutinin. J Biosci Bioeng 2024; 137:141-148. [PMID: 38110319 DOI: 10.1016/j.jbiosc.2023.11.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 10/11/2023] [Accepted: 11/28/2023] [Indexed: 12/20/2023]
Abstract
Human induced pluripotent stem cells (hiPSCs) can be used in regenerative therapy as an irresistible cell source, and so the development of scalable production of hiPSCs for three-dimensional (3D) suspension culture is required. In this study, we established a simple culture strategy for improving hiPSC aggregate growth using botulinum hemagglutinin (HA), which disrupts cell-cell adhesion mediated by E-cadherin. When HA was added to the suspension culture of hiPSC aggregates, E-cadherin-mediated cell-cell adhesion was temporarily disrupted within 24 h, but then recovered. Phosphorylated myosin light chain, a contractile force marker, was also recovered at the periphery of hiPSC aggregates. The cell aggregates were suppressed the formation of collagen type I shell-like structures at the periphery by HA and collagen type I was homogenously distributed within the cell aggregates. In addition, these cell aggregates retained the proliferation marker Ki-67 throughout the cell aggregates. The apparent specific growth rate with HA addition was maintained continuously throughout the culture, and the final cell density was 1.7-fold higher than that in the control culture. These cells retained high expression levels of pluripotency markers. These observations indicated that relaxation of cell-cell adhesions by HA addition induced rearrangement of the mechanical tensions generated by actomyosin in hiPSC aggregates and suppression of collagen type I shell-like structure formation. These results suggest that this simple and readily culture strategy is a potentially useful tool for improving the scalable production of hiPSCs for 3D suspension cultures.
Collapse
Affiliation(s)
- Riku Yamamoto
- Department of Biotechnology, Graduate School of Engineering, Osaka University, 2-1 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Ryo Sakakibara
- Department of Biotechnology, Graduate School of Engineering, Osaka University, 2-1 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Mee-Hae Kim
- Department of Biotechnology, Graduate School of Engineering, Osaka University, 2-1 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Yukako Fujinaga
- Department of Bacteriology, Graduate School of Medical Sciences, Kanazawa University, 13-1Takara-machi, Kanazawa, Ishikawa 920-8641, Japan
| | - Masahiro Kino-Oka
- Department of Biotechnology, Graduate School of Engineering, Osaka University, 2-1 Yamadaoka, Suita, Osaka 565-0871, Japan; Research Base for Cell Manufacturability, Graduate School of Engineering, Osaka University, 2-1 Yamadaoka, Suita, Osaka 565-0871, Japan.
| |
Collapse
|
16
|
Poncz M, Zaitsev SV, Ahn H, Kowalska MA, Bdeir K, Camire RM, Cines DB, Stepanova V. Packaging of supplemented urokinase into naked alpha-granules of in vitro -grown megakaryocytes for targeted therapeutic delivery. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.05.570278. [PMID: 38106191 PMCID: PMC10723305 DOI: 10.1101/2023.12.05.570278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2023]
Abstract
Our prior finding that uPA endogenously expressed and stored in the platelets of transgenic mice prevented thrombus formation without causing bleeding, prompted us to develop a potentially clinically relevant means of generating anti-thrombotic human platelets in vitro from CD34 + hematopoietic cell-derived megakaryocytes. CD34 + -megakaryocytes internalize and store in α-granules single-chain uPA (scuPA) and a uPA variant modified to be plasmin-resistant, but thrombin-activatable, (uPAT). Both uPAs co-localized with internalized factor V (FV), fibrinogen and plasminogen, low-density lipoprotein receptor-related protein 1 (LRP1), and interferon-induced transmembrane protein 3 (IFITM3), but not with endogenous von Willebrand factor (VWF). Endocytosis of uPA by CD34 + -\megakaryocytes was mediated in part via LRP1 and αIIbβ3. scuPA-containing megakaryocytes degraded endocytosed intragranular FV, but not endogenous VWF, in the presence of internalized plasminogen, whereas uPAT-megakaryocytes did not significantly degrade either protein. We used a carotid-artery injury model in NOD-scid IL2rγnull (NSG) mice homozygous for VWF R1326H (a mutation switching binding VWF specificity from mouse to human glycoprotein IbmlIX) to test whether platelets derived from scuPA-MKs or uPAT-Mks would prevent thrombus formation. NSG/VWF R1326H mice exhibited a lower thrombotic burden after carotid artery injury compared to NSG mice unless infused with human platelets or MKs, whereas intravenous injection of either uPA-containing megakaryocytes into NSG/VWF R1326H generated sufficient uPA-containing human platelets to lyse nascent thrombi. These studies suggest the potential to deliver uPA or potentially other ectopic proteins within platelet α-granules from in vitro- generated megakaryocytes. Key points Unlike platelets, in vitro-grown megakaryocytes can store exogenous uPA in its α-granules.uPA uptake involves LRP1 and αIIbβ3 receptors and is functionally available from activated platelets.
Collapse
|
17
|
Leung J, Strong C, Badior KE, Robertson M, Wu X, Meledeo MA, Kang E, Paul M, Sato Y, Harashima H, Cap AP, Devine DV, Jan E, Cullis PR, Kastrup CJ. Genetically engineered transfusable platelets using mRNA lipid nanoparticles. SCIENCE ADVANCES 2023; 9:eadi0508. [PMID: 38039367 PMCID: PMC10691771 DOI: 10.1126/sciadv.adi0508] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Accepted: 10/31/2023] [Indexed: 12/03/2023]
Abstract
Platelet transfusions are essential for managing bleeding and hemostatic dysfunction and could be expanded as a cell therapy due to the multifunctional role of platelets in various diseases. Creating these cell therapies will require modifying transfusable donor platelets to express therapeutic proteins. However, there are currently no appropriate methods for genetically modifying platelets collected from blood donors. Here, we describe an approach using platelet-optimized lipid nanoparticles containing mRNA (mRNA-LNP) to enable exogenous protein expression in human and rat platelets. Within the library of mRNA-LNP tested, exogenous protein expression did not require nor correlate with platelet activation. Transfected platelets retained hemostatic function and accumulated in regions of vascular damage after transfusion into rats with hemorrhagic shock. We expect this technology will expand the therapeutic potential of platelets.
Collapse
Affiliation(s)
- Jerry Leung
- Michael Smith Laboratories, University of British Columbia, Vancouver, V6T 1Z4, Canada
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, V6T 1Z3, Canada
- Centre for Blood Research, University of British Columbia, Vancouver, V6T 1Z3, Canada
- NanoMedicines Research Group, University of British Columbia, Vancouver, V6T 1Z3, Canada
| | - Colton Strong
- Michael Smith Laboratories, University of British Columbia, Vancouver, V6T 1Z4, Canada
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, V6T 1Z3, Canada
- Centre for Blood Research, University of British Columbia, Vancouver, V6T 1Z3, Canada
| | | | - Madelaine Robertson
- Michael Smith Laboratories, University of British Columbia, Vancouver, V6T 1Z4, Canada
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, V6T 1Z3, Canada
- Centre for Blood Research, University of British Columbia, Vancouver, V6T 1Z3, Canada
- NanoMedicines Research Group, University of British Columbia, Vancouver, V6T 1Z3, Canada
| | - Xiaowu Wu
- Blood and Shock Resuscitation Program, United States Army Institute of Surgical Research, JBSA-FT Sam Houston, San Antonio, TX 78234, USA
| | - Michael A. Meledeo
- Blood and Shock Resuscitation Program, United States Army Institute of Surgical Research, JBSA-FT Sam Houston, San Antonio, TX 78234, USA
| | - Emma Kang
- Centre for Blood Research, University of British Columbia, Vancouver, V6T 1Z3, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, V6T 2B5, Canada
| | - Manoj Paul
- Blood Research Institute, Versiti, Milwaukee,WI 53226, USA
| | - Yusuke Sato
- Laboratory for Molecular Design of Pharmaceutics, Faculty of Pharmaceutical Sciences, Hokkaido University, Hokkaido, 060-0812, Japan
| | - Hideyoshi Harashima
- Laboratory for Molecular Design of Pharmaceutics, Faculty of Pharmaceutical Sciences, Hokkaido University, Hokkaido, 060-0812, Japan
| | - Andrew P. Cap
- Blood and Shock Resuscitation Program, United States Army Institute of Surgical Research, JBSA-FT Sam Houston, San Antonio, TX 78234, USA
| | - Dana V. Devine
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, V6T 1Z3, Canada
- Centre for Blood Research, University of British Columbia, Vancouver, V6T 1Z3, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, V6T 2B5, Canada
- Centre for Innovation, Canadian Blood Services, Vancouver, V6T 1Z3, Canada
| | - Eric Jan
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, V6T 1Z3, Canada
| | - Pieter R. Cullis
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, V6T 1Z3, Canada
- NanoMedicines Research Group, University of British Columbia, Vancouver, V6T 1Z3, Canada
| | - Christian J. Kastrup
- Michael Smith Laboratories, University of British Columbia, Vancouver, V6T 1Z4, Canada
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, V6T 1Z3, Canada
- Centre for Blood Research, University of British Columbia, Vancouver, V6T 1Z3, Canada
- Blood Research Institute, Versiti, Milwaukee,WI 53226, USA
- Departments of Surgery, Biochemistry, Biomedical Engineering, and Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| |
Collapse
|
18
|
Neal MD, Hunt BJ. Precision in Transfusion Medicine. JAMA 2023; 330:1847-1848. [PMID: 37824169 DOI: 10.1001/jama.2023.16134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/13/2023]
Affiliation(s)
- Matthew D Neal
- Trauma and Transfusion Medicine Research Center, Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Beverley J Hunt
- Thrombosis and Haemophilia Centre, Guy's and St Thomas' NHS Foundation Trust, London, United Kingdom
| |
Collapse
|
19
|
Panch SR, Guo L, Vassallo R. Platelet transfusion refractoriness due to HLA alloimmunization: Evolving paradigms in mechanisms and management. Blood Rev 2023; 62:101135. [PMID: 37805287 DOI: 10.1016/j.blre.2023.101135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 09/28/2023] [Accepted: 09/29/2023] [Indexed: 10/09/2023]
Abstract
Platelet transfusion refractoriness due to HLA alloimmunization presents a significant medical problem, particularly among multiply transfused patients with hematologic malignancies and those undergoing hematopoietic stem cell transplants. HLA compatible platelet transfusions also impose significant financial burden on these patients. Recently, several novel mechanisms have been described in the development of HLA alloimmunization and platelet transfusion refractoriness. We review the history of platelet transfusions and mechanisms of HLA-sensitization and transfusion refractoriness. We also summarize advances in the diagnosis and treatment of platelet transfusion refractoriness due to HLA alloimmunization.
Collapse
Affiliation(s)
- Sandhya R Panch
- Clinical Research Division, Fred Hutchinson Cancer Center, United States of America; BloodWorks NorthWest, United States of America.
| | - Li Guo
- BloodWorks NorthWest, United States of America; Division of Hematology and Oncology, University of Washington School of Medicine, United States of America
| | | |
Collapse
|
20
|
He R, Weng Z, Liu Y, Li B, Wang W, Meng W, Li B, Li L. Application of Induced Pluripotent Stem Cells in Malignant Solid Tumors. Stem Cell Rev Rep 2023; 19:2557-2575. [PMID: 37755647 PMCID: PMC10661832 DOI: 10.1007/s12015-023-10633-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/18/2023] [Indexed: 09/28/2023]
Abstract
In the past decade, induced pluripotent stem cells (iPSCs) technology has significantly progressed in studying malignant solid tumors. This technically feasible reprogramming techniques can reawaken sequestered dormant regions that regulate the fate of differentiated cells. Despite the evolving therapeutic modalities for malignant solid tumors, treatment outcomes have not been satisfactory. Recently, scientists attempted to apply induced pluripotent stem cell technology to cancer research, from modeling to treatment. Induced pluripotent stem cells derived from somatic cells, cancer cell lines, primary tumors, and individuals with an inherited propensity to develop cancer have shown great potential in cancer modeling, cell therapy, immunotherapy, and understanding tumor progression. This review summarizes the evolution of induced pluripotent stem cells technology and its applications in malignant solid tumor. Additionally, we discuss potential obstacles to induced pluripotent stem cell technology.
Collapse
Affiliation(s)
- Rong He
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Head and Neck Oncology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Zhijie Weng
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Head and Neck Oncology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yunkun Liu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Head and Neck Oncology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Bingzhi Li
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Head and Neck Oncology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Wenxuan Wang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Head and Neck Oncology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Wanrong Meng
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Head and Neck Oncology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Bo Li
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China.
| | - Longjiang Li
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Head and Neck Oncology, West China Hospital of Stomatology, Sichuan University, Chengdu, China.
| |
Collapse
|
21
|
Kweon S, Kim S, Choi HS, Jo K, Park JM, Baek EJ. Current status of platelet manufacturing in 3D or bioreactors. Biotechnol Prog 2023; 39:e3364. [PMID: 37294031 DOI: 10.1002/btpr.3364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 05/09/2023] [Accepted: 05/09/2023] [Indexed: 06/10/2023]
Abstract
Blood shortages for transfusion are global issues of grave concern. As in vitro manufactured platelets are promising substitutes for blood donation, recent research has shown progresses including different cell sources, different bioreactors, and three-dimensional materials. The first-in-human clinical trial of cultured platelets using induced pluripotent stem cell-derived platelets began in Japan and demonstrated its quality, safety, and efficacy. A novel bioreactor with fluid motion for platelet production has been reported. Herein, we discuss various cell sources for blood cell production, recent advances in manufacturing processes, and clinical applications of cultured blood.
Collapse
Affiliation(s)
- Soonho Kweon
- Department of Research and Development, ArtBlood Inc, Seoul, Republic of Korea
| | - Suyeon Kim
- Department of Research and Development, ArtBlood Inc, Seoul, Republic of Korea
| | - Hye Sook Choi
- Department of Research and Development, ArtBlood Inc, Seoul, Republic of Korea
| | - Kyeongwon Jo
- Department of Research and Development, ArtBlood Inc, Seoul, Republic of Korea
| | - Ju Mi Park
- Department of Research and Development, ArtBlood Inc, Seoul, Republic of Korea
| | - Eun Jung Baek
- Department of Research and Development, ArtBlood Inc, Seoul, Republic of Korea
- Department of Translational Medicine, Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, Republic of Korea
- Department of Laboratory Medicine, Hanyang University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
22
|
Voloshin N, Tyurin-Kuzmin P, Karagyaur M, Akopyan Z, Kulebyakin K. Practical Use of Immortalized Cells in Medicine: Current Advances and Future Perspectives. Int J Mol Sci 2023; 24:12716. [PMID: 37628897 PMCID: PMC10454025 DOI: 10.3390/ijms241612716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 07/23/2023] [Accepted: 08/09/2023] [Indexed: 08/27/2023] Open
Abstract
In modern science, immortalized cells are not only a convenient tool in fundamental research, but they are also increasingly used in practical medicine. This happens due to their advantages compared to the primary cells, such as the possibility to produce larger amounts of cells and to use them for longer periods of time, the convenience of genetic modification, the absence of donor-to-donor variability when comparing the results of different experiments, etc. On the other hand, immortalization comes with drawbacks: possibilities of malignant transformation and/or major phenotype change due to genetic modification itself or upon long-term cultivation appear. At first glance, such issues are huge hurdles in the way of immortalized cells translation into medicine. However, there are certain ways to overcome such barriers that we describe in this review. We determined four major areas of usage of immortalized cells for practical medicinal purposes, and each has its own means to negate the drawbacks associated with immortalization. Moreover, here we describe specific fields of application of immortalized cells in which these problems are of much lesser concern, for example, in some cases where the possibility of malignant growth is not there at all. In general, we can conclude that immortalized cells have their niches in certain areas of practical medicine where they can successfully compete with other therapeutic approaches, and more preclinical and clinical trials with them should be expected.
Collapse
Affiliation(s)
- Nikita Voloshin
- Faculty of Medicine, Lomonosov Moscow State University, 119991 Moscow, Russia; (N.V.); (P.T.-K.); (M.K.)
| | - Pyotr Tyurin-Kuzmin
- Faculty of Medicine, Lomonosov Moscow State University, 119991 Moscow, Russia; (N.V.); (P.T.-K.); (M.K.)
| | - Maxim Karagyaur
- Faculty of Medicine, Lomonosov Moscow State University, 119991 Moscow, Russia; (N.V.); (P.T.-K.); (M.K.)
| | - Zhanna Akopyan
- Medical Research and Education Center, Lomonosov Moscow State University, 119234 Moscow, Russia;
| | - Konstantin Kulebyakin
- Faculty of Medicine, Lomonosov Moscow State University, 119991 Moscow, Russia; (N.V.); (P.T.-K.); (M.K.)
- Medical Research and Education Center, Lomonosov Moscow State University, 119234 Moscow, Russia;
| |
Collapse
|
23
|
Bargehr C, Knöfler R, Streif W. Treatment of Inherited Platelet Disorders: Current Status and Future Options. Hamostaseologie 2023; 43:261-270. [PMID: 37611608 DOI: 10.1055/a-2080-6602] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/25/2023] Open
Abstract
Inherited platelet disorders (IPDs) comprise a heterogeneous group of entities that manifest with variable bleeding tendencies. For successful treatment, the underlying platelet disorder, bleeding severity and location, age, and sex must be considered in the broader clinical context. Previous information from the AWMF S2K guideline #086-004 (www.awmf.org) is evaluated for validity and supplemented by information of new available and future treatment options and clinical scenarios that need specific measures. Special attention is given to the treatment of menorrhagia and risk management during pregnancy in women with IPDs. Established treatment options of IPDs include local hemostatic treatment, tranexamic acid, desmopressin, platelet concentrates, and recombinant activated factor VII. Hematopoietic stem cell therapy is a curative approach for selected patients. We also provide an outlook on promising new therapies. These include autologous hematopoietic stem cell gene therapy, artificial platelets and nanoparticles, and various other procoagulant treatments that are currently tested in clinical trials in the context of hemophilia.
Collapse
Affiliation(s)
- Caroline Bargehr
- Department of Paediatrics 1, Medical University of Innsbruck, Innsbruck, Austria
| | - Ralf Knöfler
- Department of Paediatric Haemostaseology, University Hospital Carl Gustav Carus Dresden, Dresden, Germany
| | - Werner Streif
- Department of Paediatrics 1, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
24
|
Sugimoto N, Eto K. Ex Vivo Production of Platelets From iPSCs: The iPLAT1 Study and Beyond. Hemasphere 2023; 7:e884. [PMID: 37213327 PMCID: PMC10194644 DOI: 10.1097/hs9.0000000000000884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Accepted: 03/30/2023] [Indexed: 05/23/2023] Open
Affiliation(s)
- Naoshi Sugimoto
- Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto University, Shogoin, Sakyo-ku, Kyoto, Japan
| | - Koji Eto
- Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto University, Shogoin, Sakyo-ku, Kyoto, Japan
- Department of Regenerative Medicine, Chiba University Graduate School of Medicine, Chiba, Japan
| |
Collapse
|
25
|
Di Buduo CA, Miguel CP, Balduini A. Inside-to-outside and back to the future of megakaryopoiesis. Res Pract Thromb Haemost 2023; 7:100197. [PMID: 37416054 PMCID: PMC10320384 DOI: 10.1016/j.rpth.2023.100197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 04/12/2023] [Accepted: 04/23/2023] [Indexed: 07/08/2023] Open
Abstract
A State of the Art lecture titled "Megakaryocytes and different thrombopoietic environments" was presented at the ISTH Congress in 2022. Circulating platelets are specialized cells produced by megakaryocytes. Leading studies point to the bone marrow niche as the core of hematopoietic stem cell differentiation, revealing interesting and complex environmental factors for consideration. Megakaryocytes take cues from the physiochemical bone marrow microenvironment, which includes cell-cell interactions, contact with extracellular matrix components, and flow generated by blood circulation in the sinusoidal lumen. Germinal and acquired mutations in hematopoietic stem cells may manifest in altered megakaryocyte maturation, proliferation, and platelet production. Diseased megakaryopoiesis may also cause modifications of the entire hematopoietic niche, highlighting the central role of megakaryocytes in the control of physiologic bone marrow homeostasis. Tissue-engineering approaches have been developed to translate knowledge from in vivo (inside) to functional mimics of native tissue ex vivo (outside). Reproducing the thrombopoietic environment is instrumental to gain new insight into its activity and answering the growing demand for human platelets for fundamental studies and clinical applications. In this review, we discuss the major achievements on this topic, and finally, we summarize relevant new data presented during the 2022 ISTH Congress that pave the road to the future of megakaryopoiesis.
Collapse
Affiliation(s)
| | | | - Alessandra Balduini
- Department of Molecular Medicine, University of Pavia, Pavia, Italy
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts, USA
| |
Collapse
|
26
|
Flahou C, Morishima T, Higashi N, Hayashi Y, Xu H, Wang B, Zhang C, Ninomiya A, Qiu WY, Yuzuriha A, Suzuki D, Nakamura S, Manz M, Kaneko S, Hotta A, Takizawa H, Eto K, Sugimoto N. Humanized mouse models with endogenously developed human natural killer cells for in vivo immunogenicity testing of HLA class I-edited iPSC-derived cells. Biochem Biophys Res Commun 2023; 662:76-83. [PMID: 37099813 DOI: 10.1016/j.bbrc.2023.04.067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Accepted: 04/19/2023] [Indexed: 04/28/2023]
Abstract
Human induced pluripotent stem cells (hiPSCs) genetically depleted of human leucocyte antigen (HLA) class I expression can bypass T cell alloimmunity and thus serve as a one-for-all source for cell therapies. However, these same therapies may elicit rejection by natural killer (NK) cells, since HLA class I molecules serve as inhibitory ligands of NK cells. Here, we focused on testing the capacity of endogenously developed human NK cells in humanized mice (hu-mice) using MTSRG and NSG-SGM3 strains to assay the tolerance of HLA-edited iPSC-derived cells. High NK cell reconstitution was achieved with the engraftment of cord blood-derived human hematopoietic stem cells (hHSCs) followed by the administration of human interleukin-15 (hIL-15) and IL-15 receptor alpha (hIL-15Rα). Such "hu-NK mice" rejected HLA class I-null hiPSC-derived hematopoietic progenitor cells (HPCs), megakaryocytes and T cells, but not HLA-A/B-knockout, HLA-C expressing HPCs. To our knowledge, this study is the first to recapitulate the potent endogenous NK cell response to non-tumor HLA class I-downregulated cells in vivo. Our hu-NK mouse models are suitable for the non-clinical evaluation of HLA-edited cells and will contribute to the development of universal off-the-shelf regenerative medicine.
Collapse
Affiliation(s)
- Charlotte Flahou
- Department of Clinical Application, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Tatsuya Morishima
- Laboratory of Stem Cell Stress, Kumamoto University, Kumamoto, Japan; Laboratory of Hematopoietic Stem Cell Engineering, International Research Center for Medical Sciences (IRCMS), Kumamoto University, Kumamoto, Japan
| | - Natsumi Higashi
- Department of Clinical Application, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Yoshikazu Hayashi
- Laboratory of Stem Cell Stress, Kumamoto University, Kumamoto, Japan; Laboratory of Hematopoietic Stem Cell Engineering, International Research Center for Medical Sciences (IRCMS), Kumamoto University, Kumamoto, Japan
| | - Huaigeng Xu
- Department of Clinical Application, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Bo Wang
- Department of Cell Growth and Differentiation, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Chaoqi Zhang
- Department of Cell Growth and Differentiation, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Atsushi Ninomiya
- Department of Clinical Application, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Wei-Yin Qiu
- Department of Clinical Application, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Akinori Yuzuriha
- Department of Clinical Application, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Daisuke Suzuki
- Department of Clinical Application, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Sou Nakamura
- Department of Clinical Application, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Markus Manz
- Department of Hematology, University and University Hospital Zurich, 8091, Switzerland
| | - Shin Kaneko
- Department of Cell Growth and Differentiation, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Akitsu Hotta
- Department of Clinical Application, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Hitoshi Takizawa
- Laboratory of Stem Cell Stress, Kumamoto University, Kumamoto, Japan; Center for Metabolic Regulation of Healthy Aging, Kumamoto University, Kumamoto, Japan
| | - Koji Eto
- Department of Clinical Application, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan; Department of Regenerative Medicine, Chiba University Graduate School of Medicine, Chiba, Japan.
| | - Naoshi Sugimoto
- Department of Clinical Application, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan.
| |
Collapse
|
27
|
Cleary SJ, Conrad C. Investigating and imaging platelets in inflammation. Int J Biochem Cell Biol 2023; 157:106373. [PMID: 36716816 DOI: 10.1016/j.biocel.2023.106373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 01/12/2023] [Accepted: 01/23/2023] [Indexed: 01/29/2023]
Abstract
Blood platelets are best known for their roles in hemostasis and thrombosis, but platelets also make important contributions to inflammation, immunity, and inflammatory resolution. Experiments involving depletion, genetic modification, and live imaging of platelets in animal models have increased our mechanistic understanding of platelet contributions to inflammation. In this minireview, we provide a critical overview of experimental techniques for manipulating and imaging platelets in inflammation models. We then highlight studies using innovative approaches to elucidate molecular mechanisms through which platelet subsets, platelet Fc gamma receptors, and pro-resolution platelet functions influence inflammatory responses. We also propose future technologies and research directions which might move us closer to harnessing of platelet functions for improved therapeutic modulation of inflammatory diseases.
Collapse
Affiliation(s)
- Simon J Cleary
- Department of Medicine, UCSF, Health Sciences East 1355A, 513 Parnassus Ave., San Francisco, CA 94143, USA.
| | - Catharina Conrad
- Department of Medicine, UCSF, Health Sciences East 1355A, 513 Parnassus Ave., San Francisco, CA 94143, USA
| |
Collapse
|
28
|
Couvidou A, Rojas-Jiménez G, Dupuis A, Maître B. Anti-HLA Class I alloantibodies in platelet transfusion refractoriness: From mechanisms and determinants to therapeutic prospects. Front Immunol 2023; 14:1125367. [PMID: 36845153 PMCID: PMC9947338 DOI: 10.3389/fimmu.2023.1125367] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Accepted: 01/31/2023] [Indexed: 02/11/2023] Open
Abstract
Patients with hematological disorders and severe thrombocytopenia require extensive and iterative platelet transfusion support. In these patients, platelet transfusion refractoriness represents a serious adverse transfusion event with major outcomes for patient care. Recipient alloantibodies against the donor HLA Class I antigens expressed at the cell surface of platelets result in a rapid removal of transfused platelets from the circulation and thus, therapeutic and prophylactic transfusion failure leading to a major bleeding risk. In this case, the only way to support the patient relies on the selection of HLA Class I compatible platelets, an approach restricted by the limited number of HLA-typed donors available and the difficulty of meeting the demand in an emergency. However, not all patients with anti-HLA Class I antibodies develop refractoriness to platelet transfusions, raising the question of the intrinsic characteristics of the antibodies and the immune-mediated mechanisms of platelet clearance associated with a refractory state. In this review, we examine the current challenges in platelet transfusion refractoriness and detail the key features of the antibodies involved that should be considered. Finally, we also provide an overview of future therapeutic strategies.
Collapse
Affiliation(s)
- Adèle Couvidou
- UMR_S1255, INSERM, Strasbourg, France
- Etablissement Français du Sang-Grand Est, Strasbourg, France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
- Université de Strasbourg, Strasbourg, France
| | - Gabriel Rojas-Jiménez
- UMR_S1255, INSERM, Strasbourg, France
- Etablissement Français du Sang-Grand Est, Strasbourg, France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
- Université de Strasbourg, Strasbourg, France
| | - Arnaud Dupuis
- UMR_S1255, INSERM, Strasbourg, France
- Etablissement Français du Sang-Grand Est, Strasbourg, France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
- Université de Strasbourg, Strasbourg, France
| | - Blandine Maître
- UMR_S1255, INSERM, Strasbourg, France
- Etablissement Français du Sang-Grand Est, Strasbourg, France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
- Université de Strasbourg, Strasbourg, France
| |
Collapse
|
29
|
Chen SJ, Sugimoto N, Eto K. Ex vivo manufacturing of platelets: beyond the first-in-human clinical trial using autologous iPSC-platelets. Int J Hematol 2023; 117:349-355. [PMID: 36574167 PMCID: PMC9792917 DOI: 10.1007/s12185-022-03512-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 12/05/2022] [Accepted: 12/06/2022] [Indexed: 12/28/2022]
Abstract
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.
Collapse
Affiliation(s)
- Si Jing Chen
- Department of Clinical Application, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan.
| | - Naoshi Sugimoto
- Department of Clinical Application, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Koji Eto
- Department of Clinical Application, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan. .,Department of Regenerative Medicine, Graduate School of Medicine, Chiba University, Chiba, Japan.
| |
Collapse
|
30
|
McOmish C. A monthly roundup of key articles in other journals. Transfusion 2023; 63:7-10. [PMID: 36565230 DOI: 10.1111/trf.17226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Accepted: 12/05/2022] [Indexed: 12/25/2022]
|
31
|
Progress in pathophysiological understanding and treatment of thrombocytopenia. Int J Hematol 2023; 117:314-315. [PMID: 36656456 PMCID: PMC9851102 DOI: 10.1007/s12185-023-03542-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 01/11/2023] [Accepted: 01/12/2023] [Indexed: 01/20/2023]
Abstract
Immune thrombocytopenia (ITP), thrombotic thrombocytopenic purpura (TTP), and vaccine-induced immune thrombotic thrombocytopenia (VITT) all have "thrombocytopenia" in their name, and all but congenital TTP are caused by immune mechanisms, but these conditions are quite different in their clinical features and pathophysiology. This review series covers recent progress in pathophysiology and treatment of these diseases, as well as a recent epoch-making clinical trial of induced pluripotent stem cells (iPSC)-derived platelets for patients with thrombocytopenia.
Collapse
|
32
|
Abstract
Human induced pluripotent stem cells (iPSCs), since their discovery in 2007, have rapidly become a starting cell type of choice for the differentiation of many mature cell types. Their flexibility, amenability to gene editing and functional equivalence to embryonic stem cells ensured their subsequent adoption by many manufacturing processes for cellular products. In this chapter, we will discuss the process whereby iPSCs are generated, key quality control steps which should be considered during manufacturing, the application of good manufacturing practice to production processes and iPSC-derived cellular products which are already undergoing clinical trials. iPSCs provide a new avenue for the next generation of cellular therapeutics and by combining new differentiation protocols, quality control and reproducible manufacturing, iPSC-derived cellular products could provide treatments for many currently untreatable diseases, allowing the large-scale manufacture of high-quality cell therapies.
Collapse
Affiliation(s)
- Moyra Lawrence
- Centre for iPS Cell Research and Application (CiRA) and Institute for the Advanced Study of Human Biology (ASHBi), Kyoto University, Kyoto, Japan.
| |
Collapse
|
33
|
Enosawa S. Clinical Trials of Stem Cell Therapy in Japan: The Decade of Progress under the National Program. J Clin Med 2022; 11:7030. [PMID: 36498605 PMCID: PMC9736364 DOI: 10.3390/jcm11237030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 11/24/2022] [Accepted: 11/25/2022] [Indexed: 11/29/2022] Open
Abstract
Stem cell therapy is a current world-wide topic in medical science. Various therapies have been approved based on their effectiveness and put into practical use. In Japan, research and development-related stem cell therapy, generally referred to as regenerative medicine, has been led by the government. The national scheme started in 2002, and support for the transition to clinical trials has been accelerating since 2011. Of the initial 18 projects that were accepted in the budget for preclinical research, 15 projects have begun clinical trials so far. These include the transplantation of retinal, cardiac, and dopamine-producing cells differentiated from human induced pluripotent stem (iPS) cells and hepatocyte-like cells differentiated from human embryonic stem (ES) cells. The distinctive feature of the stem cell research in Japan is the use of iPS cells. A national framework was also been set-up to attain the final goal: health insurance coverage. Now, insurance covers cell transplantation therapies for the repair and recovery of damaged skin, articular cartilage, and stroke as well as therapies introduced from abroad, such as allogeneic mesenchymal stem cells for graft-versus-host disease and chimeric antigen receptor-T (CAR-T) cell therapy. To prepare this review, original information was sought from Japanese authentic websites, which are reliable but a little hard to access due to the fact of multiple less-organized databases and the language barrier. Then, each fact was corroborated by citing its English version or publication in international journals as much as possible. This review provides a summary of progress over the past decade under the national program and a state-of-the-art factual view of research activities, government policy, and regulation in Japan for the realization of stem cell therapy.
Collapse
Affiliation(s)
- Shin Enosawa
- Division for Advanced Medical Sciences, National Center for Child Health and Development, Tokyo 157-8535, Japan
| |
Collapse
|