1
|
Baskaran D, Liu Y, Zhou J, Wang Y, Nguyen D, Wang H. In vitro and in vivo metabolic tagging and modulation of platelets. Mater Today Bio 2025; 32:101719. [PMID: 40236816 PMCID: PMC11999579 DOI: 10.1016/j.mtbio.2025.101719] [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: 12/31/2024] [Revised: 03/23/2025] [Accepted: 03/28/2025] [Indexed: 04/17/2025] Open
Abstract
Platelets play a critical role in hemostasis at sites of injury and are capable of interacting with various types of cells in the bloodstream. The promise of utilizing platelets for diagnostic and therapeutic applications has motivated the development of facile strategies to functionalize platelets. However, platelets with a small size, lack of nucleus and efficient protein machinery, and low tolerance to chemicals and transfection agents have posed significant challenges for chemical or genetic engineering. Here, for the first time, we report successful metabolic glycan labeling of platelets to introduce chemical tags (e.g., azido groups) onto the membrane of platelets. We demonstrate that azido-sugars can metabolically label platelets in a concentration dependent manner, with cell-surface azido groups detectable at as early as 4 hours. The cell-surface azido groups enable the conjugation of various macromolecular cargos including proteins and polymers onto platelets via efficient click chemistry. Small-molecule drugs such as doxorubicin can also be conjugated onto azido-labeled platelets and become subsequently released to kill surrounding cancer cells, demonstrating the feasibility of utilizing platelets as a drug delivery vehicle. We further show that azido-sugars, upon intraperitoneal injection, can metabolically label platelets with azido groups in vivo, which persist for up to 4 days in mice (nearly the life-span of murine platelets). This in vitro and in vivo platelet labeling and targeting technology opens a new avenue to platelet-based diagnostics and therapeutics.
Collapse
Affiliation(s)
- Dhyanesh Baskaran
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Yusheng Liu
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Jiadiao Zhou
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Yueji Wang
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Daniel Nguyen
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Hua Wang
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
- Cancer Center at Illinois (CCIL), Urbana, IL, 61801, USA
- Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
- Carle College of Medicine, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
- Materials Research Laboratory, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
- Grainger College of Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| |
Collapse
|
2
|
Ceron-Hernandez J, Martinez-Navajas G, Sanchez-Manas JM, Molina MP, Xie J, Aznar-Peralta I, Garcia-Diaz A, Perales S, Torres C, Serrano MJ, Real PJ. Oncogenic KRAS G12D Transfer from Platelet-like Particles Enhances Proliferation and Survival in Non-Small Cell Lung Cancer Cells. Int J Mol Sci 2025; 26:3264. [PMID: 40244100 PMCID: PMC11990068 DOI: 10.3390/ijms26073264] [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: 01/28/2025] [Revised: 03/11/2025] [Accepted: 03/26/2025] [Indexed: 04/18/2025] Open
Abstract
In the tumor context, platelets play a significant role in primary tumor progression, dissemination and metastasis. Analysis of this interaction in various cancers, such as non-small cell lung cancer (NSCLC), demonstrate that platelets can both transfer and receive biomolecules (e.g. RNA and proteins) to and from the tumor at different stages, becoming tumor-educated platelets. To investigate how platelets are able to transfer oncogenic material, we developed in vitro platelet-like particles (PLPs), from a differentiated MEG-01 cell line, that stably carry RNA and protein of the KRASG12D oncogene in fusion with GFP. We co-cultured these PLPs with NSCLC H1975 tumor cells to assess their ability to transfer this material. We observed that the generated platelets were capable of stably expressing the oncogene and transferring both its RNA and protein forms to tumor cells using qPCR and imaging techniques. Additionally, we found that coculturing PLPs loaded with GFP-KRASG12D with tumor cells increased their proliferative capacity at specific PLP concentrations. In conclusion, our study successfully engineered an MEG-01 cell line to produce PLPs carrying oncogenic GFP-KRASG12D simulating the tumor microenvironment, demonstrating the efficient transfer of this oncogene to tumor cells and its significant impact on enhancing proliferation.
Collapse
Affiliation(s)
- Jorge Ceron-Hernandez
- Gene Regulation, Stem Cells and Development Group, GENyO, Pfizer-University of Granada-Andalusian Regional Government Centre for Genomics and Oncological Research, Avenida de la Ilustración 114, 18016 Granada, Spain; (J.C.-H.); (G.M.-N.); (J.M.S.-M.); (J.X.); (S.P.); (C.T.)
- Liquid Biopsies and Cancer Interception Group, PTS, Granada GENyO, Pfizer-University of Granada-Andalusian Regional Government Centre for Genomics and Oncological Research, Avenida de la Ilustración 114, 18016 Granada, Spain; (M.P.M.); (I.A.-P.); (A.G.-D.)
- Department of Biochemistry and Molecular Biology I, Faculty of Science, University of Granada, Avenida Fuentenueva s/n, 18071 Granada, Spain
| | - Gonzalo Martinez-Navajas
- Gene Regulation, Stem Cells and Development Group, GENyO, Pfizer-University of Granada-Andalusian Regional Government Centre for Genomics and Oncological Research, Avenida de la Ilustración 114, 18016 Granada, Spain; (J.C.-H.); (G.M.-N.); (J.M.S.-M.); (J.X.); (S.P.); (C.T.)
- Department of Biochemistry and Molecular Biology I, Faculty of Science, University of Granada, Avenida Fuentenueva s/n, 18071 Granada, Spain
| | - Jose Manuel Sanchez-Manas
- Gene Regulation, Stem Cells and Development Group, GENyO, Pfizer-University of Granada-Andalusian Regional Government Centre for Genomics and Oncological Research, Avenida de la Ilustración 114, 18016 Granada, Spain; (J.C.-H.); (G.M.-N.); (J.M.S.-M.); (J.X.); (S.P.); (C.T.)
- Department of Biochemistry and Molecular Biology I, Faculty of Science, University of Granada, Avenida Fuentenueva s/n, 18071 Granada, Spain
| | - María Pilar Molina
- Liquid Biopsies and Cancer Interception Group, PTS, Granada GENyO, Pfizer-University of Granada-Andalusian Regional Government Centre for Genomics and Oncological Research, Avenida de la Ilustración 114, 18016 Granada, Spain; (M.P.M.); (I.A.-P.); (A.G.-D.)
| | - Jiajun Xie
- Gene Regulation, Stem Cells and Development Group, GENyO, Pfizer-University of Granada-Andalusian Regional Government Centre for Genomics and Oncological Research, Avenida de la Ilustración 114, 18016 Granada, Spain; (J.C.-H.); (G.M.-N.); (J.M.S.-M.); (J.X.); (S.P.); (C.T.)
- Department of Biochemistry and Molecular Biology I, Faculty of Science, University of Granada, Avenida Fuentenueva s/n, 18071 Granada, Spain
| | - Inés Aznar-Peralta
- Liquid Biopsies and Cancer Interception Group, PTS, Granada GENyO, Pfizer-University of Granada-Andalusian Regional Government Centre for Genomics and Oncological Research, Avenida de la Ilustración 114, 18016 Granada, Spain; (M.P.M.); (I.A.-P.); (A.G.-D.)
| | - Abel Garcia-Diaz
- Liquid Biopsies and Cancer Interception Group, PTS, Granada GENyO, Pfizer-University of Granada-Andalusian Regional Government Centre for Genomics and Oncological Research, Avenida de la Ilustración 114, 18016 Granada, Spain; (M.P.M.); (I.A.-P.); (A.G.-D.)
| | - Sonia Perales
- Gene Regulation, Stem Cells and Development Group, GENyO, Pfizer-University of Granada-Andalusian Regional Government Centre for Genomics and Oncological Research, Avenida de la Ilustración 114, 18016 Granada, Spain; (J.C.-H.); (G.M.-N.); (J.M.S.-M.); (J.X.); (S.P.); (C.T.)
- Department of Biochemistry and Molecular Biology I, Faculty of Science, University of Granada, Avenida Fuentenueva s/n, 18071 Granada, Spain
- Instituto de Investigación Biosanitaria ibs.GRANADA, 18012 Granada, Spain
| | - Carolina Torres
- Gene Regulation, Stem Cells and Development Group, GENyO, Pfizer-University of Granada-Andalusian Regional Government Centre for Genomics and Oncological Research, Avenida de la Ilustración 114, 18016 Granada, Spain; (J.C.-H.); (G.M.-N.); (J.M.S.-M.); (J.X.); (S.P.); (C.T.)
- Instituto de Investigación Biosanitaria ibs.GRANADA, 18012 Granada, Spain
- Department of Biochemistry and Molecular Biology III and Immunology, Faculty of Medicine, University of Granada, Avenida de la Investigación 11, 18016 Granada, Spain
| | - Maria J. Serrano
- Liquid Biopsies and Cancer Interception Group, PTS, Granada GENyO, Pfizer-University of Granada-Andalusian Regional Government Centre for Genomics and Oncological Research, Avenida de la Ilustración 114, 18016 Granada, Spain; (M.P.M.); (I.A.-P.); (A.G.-D.)
- Instituto de Investigación Biosanitaria ibs.GRANADA, 18012 Granada, Spain
- Molecular Pathology Lab. Intercenter Anatomical Pathology Unit, San Cecilio and Virgen de las Nieves University Hospitals, 18016 Granada, Spain
| | - Pedro J. Real
- Gene Regulation, Stem Cells and Development Group, GENyO, Pfizer-University of Granada-Andalusian Regional Government Centre for Genomics and Oncological Research, Avenida de la Ilustración 114, 18016 Granada, Spain; (J.C.-H.); (G.M.-N.); (J.M.S.-M.); (J.X.); (S.P.); (C.T.)
- Liquid Biopsies and Cancer Interception Group, PTS, Granada GENyO, Pfizer-University of Granada-Andalusian Regional Government Centre for Genomics and Oncological Research, Avenida de la Ilustración 114, 18016 Granada, Spain; (M.P.M.); (I.A.-P.); (A.G.-D.)
- Department of Biochemistry and Molecular Biology I, Faculty of Science, University of Granada, Avenida Fuentenueva s/n, 18071 Granada, Spain
| |
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 2025; 23:306-313. [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] [MESH Headings] [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
|
Strong C, Leung J, Kang E, Badior KE, Robertson M, Pereyra N, Rowe EM, Wietrzny A, Ma B, Noronha Z, Arnold D, Ciufolini MA, Devine DV, Jan E, Cullis PR, Kastrup CJ. Genetic engineering of transfusable platelets with mRNA-lipid nanoparticles is compatible with blood banking practices. Blood 2024; 144:2223-2236. [PMID: 39190426 DOI: 10.1182/blood.2024024405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Revised: 07/30/2024] [Accepted: 08/13/2024] [Indexed: 08/28/2024] Open
Abstract
ABSTRACT Platelets contribute to a variety of physiological processes, including inflammation, sepsis, and cancer. However, because of their primary role in hemostasis, platelet transfusions are largely restricted to managing thrombocytopenia and bleeding. One way to expand the utility of platelet transfusions would be to genetically engineer donor platelets with new or enhanced functions. We have previously shown that lipid nanoparticles containing mRNA (mRNA-LNP) can be used to genetically modify authentic platelets in a nonclinical crystalloid solution. Currently, platelets collected for transfusion are stored in plasma or in plasma supplemented with platelet additive solution (PAS) at supraphysiological concentrations at room temperature, or at 4°C if intended for use in acute hemorrhage. Here, we describe a new plasma-optimized mRNA-LNP for transfecting platelets directly in plasma and plasma supplemented with PAS that is scalable to physiological and supraphysiological platelet concentrations. Transfecting platelets in clinical solutions with mRNA-LNP does not affect aspects of in vitro physiology, and transfected platelets are storable. The compatibility of this transfection system with current clinical practices could enable future mRNA-LNP-based platelet products and cell therapies.
Collapse
Affiliation(s)
- Colton Strong
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC, Canada
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, Canada
- Centre for Blood Research, University of British Columbia, Vancouver, BC, Canada
| | - Jerry Leung
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC, Canada
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, Canada
- Centre for Blood Research, University of British Columbia, Vancouver, BC, Canada
- NanoMedicines Research Group, University of British Columbia, Vancouver, BC, Canada
| | - Emma Kang
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC, Canada
- Centre for Blood Research, University of British Columbia, Vancouver, BC, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | | | - Madelaine Robertson
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC, Canada
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, Canada
- Centre for Blood Research, University of British Columbia, Vancouver, BC, Canada
- NanoMedicines Research Group, University of British Columbia, Vancouver, BC, Canada
| | - Nicolas Pereyra
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, Canada
- Centre for Blood Research, University of British Columbia, Vancouver, BC, Canada
| | - Elyn M Rowe
- Centre for Blood Research, University of British Columbia, Vancouver, BC, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | | | - Brenda Ma
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, Canada
| | - Zechariah Noronha
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, Canada
| | | | - Marco A Ciufolini
- NanoVation Therapeutics, Vancouver, BC, Canada
- Department of Chemistry, University of British Columbia, Vancouver, BC, Canada
| | - Dana V Devine
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, Canada
- Centre for Blood Research, University of British Columbia, Vancouver, BC, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Eric Jan
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, Canada
| | - Pieter R Cullis
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, Canada
- NanoMedicines Research Group, University of British Columbia, Vancouver, BC, Canada
| | - Christian J Kastrup
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC, Canada
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, Canada
- Centre for Blood Research, University of British Columbia, Vancouver, BC, Canada
- Versiti Blood Research Institute, Milwaukee, WI
- Departments of Surgery, Biochemistry, Biomedical Engineering, and Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI
| |
Collapse
|
5
|
Kong YX, Perdomo JS, Passam FH. A new track for KDELivery of designer cargo by platelets. J Thromb Haemost 2024; 22:3007-3009. [PMID: 39461809 DOI: 10.1016/j.jtha.2024.09.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 09/03/2024] [Accepted: 09/05/2024] [Indexed: 10/29/2024]
Affiliation(s)
- Yvonne X Kong
- Haematology Research Group, Charles Perkins Centre, Central Clinical School, Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia
| | - Jose S Perdomo
- Haematology Research Group, Charles Perkins Centre, Central Clinical School, Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia
| | - Freda H Passam
- Haematology Research Group, Charles Perkins Centre, Central Clinical School, Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia.
| |
Collapse
|
6
|
Asquith NL, Becker IC, Scimone MT, Boccia T, Camacho V, Barrachina MN, Guo S, Freire D, Machlus K, Schulman S, Flaumenhaft R, Italiano JE. Targeting cargo to an unconventional secretory system within megakaryocytes allows the release of transgenic proteins from platelets. J Thromb Haemost 2024; 22:3235-3248. [PMID: 39122192 DOI: 10.1016/j.jtha.2024.07.021] [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: 01/16/2024] [Revised: 07/21/2024] [Accepted: 07/23/2024] [Indexed: 08/12/2024]
Abstract
BACKGROUND Platelets are essential for hemostasis and thrombosis and play vital roles during metastatic cancer progression and infection. Hallmarks of platelet function are activation, cytoskeletal rearrangements, and the degranulation of their cellular contents upon stimulation. While α-granules and dense granules are the most studied platelet secretory granules, the dense tubular system (DTS) also functions as a secretory system for vascular thiol isomerases. However, how DTS cargo is packaged and transported from megakaryocytes (MKs) to platelets is poorly understood. OBJECTIVES To underpin the mechanisms responsible for DTS cargo transport and leverage those for therapeutic protein packaging into platelets. METHODS A retroviral expression system combined with immunofluorescence confocal microscopy was employed to track protein DTS cargo protein disulfide isomerase fused to enhanced green fluorescent protein (eGFP-PDI) during platelet production. Murine bone marrow transplantation models were used to determine the release of therapeutic proteins from platelets. RESULTS We demonstrated that the endoplasmic reticulum retrieval motif Lys-Asp-Glu-Leu (KDEL) located at the C-terminus of protein disulfide isomerase was essential for the regular transport of eGFP-PDI-containing granules. eGFP-PDIΔKDEL, in which the retrieval signal was deleted, was aberrantly packaged, and its expression was upregulated within clathrin-coated endosomes. Finally, we found that ectopic transgenic proteins, such as tissue factor pathway inhibitor and interleukin 2, can be packaged into MKs and proplatelets by adding a KDEL retrieval sequence. CONCLUSION Our data corroborate the DTS as a noncanonical secretory system in platelets and demonstrate that in vitro-generated MKs and platelets may be used as a delivery system for transgenic proteins during cellular therapy.
Collapse
Affiliation(s)
- Nathan L Asquith
- Vascular Biology Program, Boston Children's Hospital, Boston, Massachusetts, USA; Harvard Medical School, Boston, Massachusetts, USA. https://twitter.com/NathanAsquith1
| | - Isabelle C Becker
- Vascular Biology Program, Boston Children's Hospital, Boston, Massachusetts, USA; Harvard Medical School, Boston, Massachusetts, USA
| | - Mark T Scimone
- Cellular Imaging Core, Neurobiology, Boston Children's Hospital, Boston, Massachusetts, USA; Life Sciences, Biotechnology, University of New Hampshire, Manchester, New Hampshire, USA
| | - Thais Boccia
- Harvard Medical School, Boston, Massachusetts, USA; Department of Surgery, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Virginia Camacho
- Vascular Biology Program, Boston Children's Hospital, Boston, Massachusetts, USA; Harvard Medical School, Boston, Massachusetts, USA
| | - María N Barrachina
- Vascular Biology Program, Boston Children's Hospital, Boston, Massachusetts, USA; Harvard Medical School, Boston, Massachusetts, USA
| | - Shihui Guo
- Harvard Medical School, Division of Hemostasis and Thrombosis, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| | - Daniela Freire
- Vascular Biology Program, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Kellie Machlus
- Vascular Biology Program, Boston Children's Hospital, Boston, Massachusetts, USA; Harvard Medical School, Boston, Massachusetts, USA
| | - Sol Schulman
- Harvard Medical School, Division of Hemostasis and Thrombosis, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| | - Robert Flaumenhaft
- Harvard Medical School, Division of Hemostasis and Thrombosis, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| | - Joseph E Italiano
- Vascular Biology Program, Boston Children's Hospital, Boston, Massachusetts, USA; Harvard Medical School, Boston, Massachusetts, USA.
| |
Collapse
|
7
|
Ferraresso F, Leung J, Kastrup CJ. RNA therapeutics to control fibrinolysis: review on applications in biology and medicine. J Thromb Haemost 2024; 22:2103-2114. [PMID: 38663489 PMCID: PMC11269028 DOI: 10.1016/j.jtha.2024.04.006] [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: 02/13/2024] [Revised: 04/11/2024] [Accepted: 04/11/2024] [Indexed: 05/26/2024]
Abstract
Regulation of fibrinolysis, the process that degrades blood clots, is pivotal in maintaining hemostasis. Dysregulation leads to thrombosis or excessive bleeding. Proteins in the fibrinolysis system include fibrinogen, coagulation factor XIII, plasminogen, tissue plasminogen activator, urokinase plasminogen activator, α2-antiplasmin, thrombin-activatable fibrinolysis inhibitor, plasminogen activator inhibitor-1, α2-macroglobulin, and others. While each of these is a potential therapeutic target for diseases, they lack effective or long-acting inhibitors. Rapid advances in RNA-based technologies are creating powerful tools to control the expression of proteins. RNA agents can be long-acting and tailored to either decrease or increase production of a specific protein. Advances in nucleic acid delivery, such as by lipid nanoparticles, have enabled the delivery of RNA to the liver, where most proteins of coagulation and fibrinolysis are produced. This review will summarize the classes of RNA that induce 1) inhibition of protein synthesis, including small interfering RNA and antisense oligonucleotides; 2) protein expression, including messenger RNA and self-amplifying RNA; and 3) gene editing for gene knockdown and precise editing. It will review specific examples of RNA therapies targeting proteins in the coagulation and fibrinolysis systems and comment on the wide range of opportunities for controlling fibrinolysis for biological applications and future therapeutics using state-of-the-art RNA therapies.
Collapse
Affiliation(s)
- Francesca Ferraresso
- Blood Research Institute, Versiti Wisconsin, Milwaukee, Wisconsin, USA; 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
| | - 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
| | - Christian J Kastrup
- Blood Research Institute, Versiti Wisconsin, Milwaukee, Wisconsin, USA; 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; Departments of Surgery, Biochemistry, Biomedical Engineering, and Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA.
| |
Collapse
|
8
|
Wu M, Shi Y, Zhao J, Kong M. Engineering unactivated platelets for targeted drug delivery. Biomater Sci 2024; 12:2244-2258. [PMID: 38482903 DOI: 10.1039/d4bm00029c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/01/2024]
Abstract
As a vital component of blood, platelets play crucial roles in hemostasis and maintaining vascular integrity, and actively participate in inflammation and immune regulation. The unique biological properties of natural platelets have enabled their utilization as drug delivery vehicles. The advancement and integration of various techniques, including biological, chemical, and physicochemical methods, have enabled the preparation of engineered platelets. Platelets can serve as drug delivery platforms combined with immunotherapy and chemokine therapy to enhance their therapeutic impact. This review focuses on the recent advancements in the application of unactivated platelets for drug delivery. The construction strategies of engineered platelets are comprehensively summarized, encompassing internal loading, surface modification, and genetic engineering techniques. Engineered platelets hold vast potential for treating cardiovascular diseases, cancers, and infectious diseases. Furthermore, the challenges and potential considerations in creating engineered platelets with natural activity are discussed.
Collapse
Affiliation(s)
- Meng Wu
- College of Marine Life Sciences, Ocean University of China, Qingdao, Shandong Province, 266003, China.
| | - Yan Shi
- College of Marine Life Sciences, Ocean University of China, Qingdao, Shandong Province, 266003, China.
| | - Jiaxuan Zhao
- College of Marine Life Sciences, Ocean University of China, Qingdao, Shandong Province, 266003, China.
| | - Ming Kong
- College of Marine Life Sciences, Ocean University of China, Qingdao, Shandong Province, 266003, China.
| |
Collapse
|
9
|
Wu L, Li X, Qian X, Wang S, Liu J, Yan J. Lipid Nanoparticle (LNP) Delivery Carrier-Assisted Targeted Controlled Release mRNA Vaccines in Tumor Immunity. Vaccines (Basel) 2024; 12:186. [PMID: 38400169 PMCID: PMC10891594 DOI: 10.3390/vaccines12020186] [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: 01/16/2024] [Revised: 02/02/2024] [Accepted: 02/06/2024] [Indexed: 02/25/2024] Open
Abstract
In recent years, lipid nanoparticles (LNPs) have attracted extensive attention in tumor immunotherapy. Targeting immune cells in cancer therapy has become a strategy of great research interest. mRNA vaccines are a potential choice for tumor immunotherapy, due to their ability to directly encode antigen proteins and stimulate a strong immune response. However, the mode of delivery and lack of stability of mRNA are key issues limiting its application. LNPs are an excellent mRNA delivery carrier, and their structural stability and biocompatibility make them an effective means for delivering mRNA to specific targets. This study summarizes the research progress in LNP delivery carrier-assisted targeted controlled release mRNA vaccines in tumor immunity. The role of LNPs in improving mRNA stability, immunogenicity, and targeting is discussed. This review aims to systematically summarize the latest research progress in LNP delivery carrier-assisted targeted controlled release mRNA vaccines in tumor immunity to provide new ideas and strategies for tumor immunotherapy, as well as to provide more effective treatment plans for patients.
Collapse
Affiliation(s)
- Liusheng Wu
- Center of Hepatobiliary Pancreatic Disease, Beijing Tsinghua Changgung Hospital, School of Medicine, Tsinghua University, Beijing 100084, China; (L.W.); (X.Q.); (S.W.)
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119077, Singapore
| | - Xiaoqiang Li
- Department of Thoracic Surgery, Peking University Shenzhen Hospital, Shenzhen 518036, China;
| | - Xinye Qian
- Center of Hepatobiliary Pancreatic Disease, Beijing Tsinghua Changgung Hospital, School of Medicine, Tsinghua University, Beijing 100084, China; (L.W.); (X.Q.); (S.W.)
| | - Shuang Wang
- Center of Hepatobiliary Pancreatic Disease, Beijing Tsinghua Changgung Hospital, School of Medicine, Tsinghua University, Beijing 100084, China; (L.W.); (X.Q.); (S.W.)
| | - Jixian Liu
- Department of Thoracic Surgery, Peking University Shenzhen Hospital, Shenzhen 518036, China;
| | - Jun Yan
- Center of Hepatobiliary Pancreatic Disease, Beijing Tsinghua Changgung Hospital, School of Medicine, Tsinghua University, Beijing 100084, China; (L.W.); (X.Q.); (S.W.)
| |
Collapse
|