1
|
Das S, Thompson W, Papoutsakis ET. Engineered and hybrid human megakaryocytic extracellular vesicles for targeted non-viral cargo delivery to hematopoietic (blood) stem and progenitor cells. Front Bioeng Biotechnol 2024; 12:1435228. [PMID: 39386042 PMCID: PMC11461334 DOI: 10.3389/fbioe.2024.1435228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2024] [Accepted: 09/11/2024] [Indexed: 10/12/2024] Open
Abstract
Native and engineered extracellular vesicles generated from human megakaryocytes (huMkEVs) or from the human megakaryocytic cell line CHRF (CHEVs) interact with tropism delivering their cargo to both human and murine hematopoietic stem and progenitor cells (HSPCs). To develop non-viral delivery vectors to HSPCs based on MkEVs, we first confirmed, using NOD-scid IL2Rγnull (NSG™) mice, the targeting potential of the large EVs, enriched in microparticles (huMkMPs), chosen for their large cargo capacity. 24 h post intravenous infusion into NSG mice, huMkEVs induced a nearly 50% increase in murine platelet counts. PKH26-labeled huMkEVs or CHEVs localized to the HSPC-rich bone marrow preferentially interacting with murine HSPCs, thus confirming their receptor-mediated tropism for NSG HSPCs, and their potential to treat thromobocytopenias. We explored this tropism to functionally deliver synthetic cargo, notably plasmid DNA coding for a fluorescent reporter, to NSG HSPCs both in vitro and in vivo. We loaded huMkEVs with plasmid DNA either through electroporation or by generating hybrid particles with preloaded liposomes. Both methods facilitated successful functional targeted delivery of pDNA, as tissue weight-normalized fluorescence intensity of the expressed fluorescent reporter was significantly higher in bone marrow than other tissues. Furthermore, the fraction of fluorescent CD117+ HSPCs was nearly 19-fold higher than other cell types within the bone marrow 72-h following administration of the hybrid particles, further supporting that HSPC tropism is retained when using hybrid particles. These data demonstrate the potential of these EVs as a non-viral, HSPC-specific cargo vehicle for gene therapy applications to treat hematological diseases.
Collapse
Affiliation(s)
- Samik Das
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, DE, United States
- Delaware Biotechnology Institute, University of Delaware, Newark, DE, United States
| | - Will Thompson
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, DE, United States
- Delaware Biotechnology Institute, University of Delaware, Newark, DE, United States
| | - Eleftherios Terry Papoutsakis
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, DE, United States
- Delaware Biotechnology Institute, University of Delaware, Newark, DE, United States
- Department of Biological Sciences, University of Delaware, Newark, DE, United States
| |
Collapse
|
2
|
Carminita E, Becker IC, Italiano JE. What It Takes To Be a Platelet: Evolving Concepts in Platelet Production. Circ Res 2024; 135:540-549. [PMID: 39088641 DOI: 10.1161/circresaha.124.323579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 08/03/2024]
Abstract
Platelets are among the most abundant cells within the circulation. Given that the platelet lifespan is 7 to 10 days in humans, a constant production of around 100 billion platelets per day is required. Platelet production from precursor cells called megakaryocytes is one of the most enigmatic processes in human biology. Although it has been studied for over a century, there is still controversy about the exact mechanisms leading to platelet release into circulation. The formation of proplatelet extensions from megakaryocytes into bone marrow sinusoids is the best-described mechanism explaining the origin of blood platelets. However, using powerful imaging techniques, several emerging studies have recently raised challenging questions in the field, suggesting that small platelet-sized structures called buds might also contribute to the circulating platelet pool. How and whether these structures differ from microvesicles or membrane blebs, which have previously been described to be released from megakaryocytes, is still a matter of discussion. In this review, we will summarize what the past and present have revealed about platelet production and whether mature blood platelets might emerge via different mechanisms.
Collapse
Affiliation(s)
- Estelle Carminita
- Vascular Biology Program, Boston Children's Hospital, Boston, MA (E.C., I.C.B., J.E.I.)
- Harvard Medical School, Boston, MA (E.C., I.C.B.)
| | - Isabelle C Becker
- Vascular Biology Program, Boston Children's Hospital, Boston, MA (E.C., I.C.B., J.E.I.)
- Harvard Medical School, Boston, MA (E.C., I.C.B.)
| | - Joseph E Italiano
- Vascular Biology Program, Boston Children's Hospital, Boston, MA (E.C., I.C.B., J.E.I.)
| |
Collapse
|
3
|
Thompson W, Papoutsakis ET. Similar but distinct: The impact of biomechanical forces and culture age on the production, cargo loading, and biological efficacy of human megakaryocytic extracellular vesicles for applications in cell and gene therapies. Bioeng Transl Med 2023; 8:e10563. [PMID: 37693047 PMCID: PMC10486331 DOI: 10.1002/btm2.10563] [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: 03/24/2023] [Revised: 05/18/2023] [Accepted: 06/01/2023] [Indexed: 09/12/2023] Open
Abstract
Megakaryocytic extracellular vesicles (MkEVs) promote the growth and megakaryopoiesis of hematopoietic stem and progenitor cells (HSPCs) largely through endogenous miR-486-5p and miR-22-3p cargo. Here, we examine the impact of biomechanical force and culture age/differentiation on the formation, properties, and biological efficacy of MkEVs. We applied biomechanical force to Mks using two methods: shake flask cultures and a syringe pump system. Force increased MkEV production in a magnitude-dependent manner, with similar trends emerging regardless of whether flow cytometry or nanoparticle tracking analysis was used for MkEV counting. Both methods produced MkEVs that were relatively depleted of miR-486-5p and miR-22-3p cargo. However, while the shake flask-derived MkEVs were correspondingly less effective in promoting megakaryocytic differentiation of HSPCs, the syringe pump-derived MkEVs were more effective in doing so, suggesting the presence of unique, unidentified miRNA cargo components. Higher numbers of MkEVs were also produced by "older" Mk cultures, though miRNA cargo levels and MkEV bioactivity were unaffected by culture age. A reduction in MkEV production by Mks derived from late-differentiating HSPCs was also noted. Taken together, our results demonstrate that biomechanical force has an underappreciated and deeply influential role in MkEV biology, though that role may vary significantly depending on the nature of the force. Given the ubiquity of biomechanical force in vivo and in biomanufacturing, this phenomenon must be grappled with before MkEVs can attain clinical relevance.
Collapse
Affiliation(s)
- Will Thompson
- Department of Chemical and Biomolecular EngineeringUniversity of DelawareNewarkDelawareUSA
| | | |
Collapse
|
4
|
Adamczyk AM, Leicaj ML, Fabiano MP, Cabrerizo G, Bannoud N, Croci DO, Witwer KW, Remes Lenicov F, Ostrowski M, Pérez PS. Extracellular vesicles from human plasma dampen inflammation and promote tissue repair functions in macrophages. J Extracell Vesicles 2023; 12:e12331. [PMID: 37272889 PMCID: PMC10241174 DOI: 10.1002/jev2.12331] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 04/14/2023] [Accepted: 05/04/2023] [Indexed: 06/06/2023] Open
Abstract
Although inflammation is a vital defence response to infection, if left uncontrolled, it can lead to pathology. Macrophages are critical players both in driving the inflammatory response and in the subsequent events required for restoring tissue homeostasis. Extracellular vesicles (EVs) are membrane-enclosed structures released by cells that mediate intercellular communication and are present in all biological fluids, including blood. Herein, we show that extracellular vesicles from plasma (pEVs) play a relevant role in the control of inflammation by counteracting PAMP-induced macrophage activation. Indeed, pEV-treatment of macrophages simultaneously with or prior to PAMP exposure reduced the secretion of pro-inflammatory IL-6 and TNF-α and increased IL-10 response. This anti-inflammatory activity was associated with the promotion of tissue-repair functions in macrophages, characterized by augmented efferocytosis and pro-angiogenic capacity, and increased expression of VEGFa, CD300e, RGS2 and CD93, genes involved in cell growth and tissue remodelling. We also show that simultaneous stimulation of macrophages with a PAMP and pEVs promoted COX2 expression and CREB phosphorylation as well as the accumulation of higher concentrations of PGE2 in cell culture supernatants. Remarkably, the anti-inflammatory activity of pEVs was abolished if cells were treated with a pharmacological inhibitor of COX2, indicating that pEV-mediated induction of COX2 is critical for the pEV-mediated inhibition of inflammation. Finally, we show that pEVs added to monocytes prior to their M-CSF-induced differentiation to macrophages increased efferocytosis and diminished pro-inflammatory cytokine responses to PAMP stimulation. In conclusion, our results suggest that pEVs are endogenous homeostatic modulators of macrophages, activating the PGE2/CREB pathway, decreasing the production of inflammatory cytokines and promoting tissue repair functions.
Collapse
Affiliation(s)
- Alan M. Adamczyk
- Instituto de Investigaciones Biomédicas en Retrovirus y SIDA (INBIRS)Universidad de Buenos Aires‐CONICETBuenos AiresArgentina
| | - María Luz Leicaj
- Instituto de Investigaciones Biomédicas en Retrovirus y SIDA (INBIRS)Universidad de Buenos Aires‐CONICETBuenos AiresArgentina
| | - Martina Paula Fabiano
- Instituto de Investigaciones Biomédicas en Retrovirus y SIDA (INBIRS)Universidad de Buenos Aires‐CONICETBuenos AiresArgentina
| | - Gonzalo Cabrerizo
- Instituto de Investigaciones Biomédicas en Retrovirus y SIDA (INBIRS)Universidad de Buenos Aires‐CONICETBuenos AiresArgentina
| | - Nadia Bannoud
- Laboratorio de Glicobiología y Biología VascularInstituto de Histología y Embriología de MendozaCONICET‐Universidad Nacional de CuyoMendozaArgentina
| | - Diego O. Croci
- Laboratorio de Glicobiología y Biología VascularInstituto de Histología y Embriología de MendozaCONICET‐Universidad Nacional de CuyoMendozaArgentina
| | - Kenneth W. Witwer
- Department of Molecular and Comparative PathobiologyThe Johns Hopkins University School of MedicineBaltimoreMarylandUSA
- Department of NeurologyThe Johns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Federico Remes Lenicov
- Instituto de Investigaciones Biomédicas en Retrovirus y SIDA (INBIRS)Universidad de Buenos Aires‐CONICETBuenos AiresArgentina
| | - Matías Ostrowski
- Instituto de Investigaciones Biomédicas en Retrovirus y SIDA (INBIRS)Universidad de Buenos Aires‐CONICETBuenos AiresArgentina
| | - Paula Soledad Pérez
- Instituto de Investigaciones Biomédicas en Retrovirus y SIDA (INBIRS)Universidad de Buenos Aires‐CONICETBuenos AiresArgentina
| |
Collapse
|
5
|
Das S, Harris JC, Winter EJ, Kao C, Day ES, Papoutsakis ET. Megakaryocyte membrane-wrapped nanoparticles for targeted cargo delivery to hematopoietic stem and progenitor cells. Bioeng Transl Med 2023; 8:e10456. [PMID: 37206243 PMCID: PMC10189472 DOI: 10.1002/btm2.10456] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 11/02/2022] [Accepted: 11/17/2022] [Indexed: 09/12/2023] Open
Abstract
Hematopoietic stem and progenitor cells (HSPCs) are desirable targets for gene therapy but are notoriously difficult to target and transfect. Existing viral vector-based delivery methods are not effective in HSPCs due to their cytotoxicity, limited HSPC uptake and lack of target specificity (tropism). Poly(lactic-co-glycolic acid) (PLGA) nanoparticles (NPs) are attractive, nontoxic carriers that can encapsulate various cargo and enable its controlled release. To engineer PLGA NP tropism for HSPCs, megakaryocyte (Mk) membranes, which possess HSPC-targeting moieties, were extracted and wrapped around PLGA NPs, producing MkNPs. In vitro, fluorophore-labeled MkNPs are internalized by HSPCs within 24 h and were selectively taken up by HSPCs versus other physiologically related cell types. Using membranes from megakaryoblastic CHRF-288 cells containing the same HSPC-targeting moieties as Mks, CHRF-wrapped NPs (CHNPs) loaded with small interfering RNA facilitated efficient RNA interference upon delivery to HSPCs in vitro. HSPC targeting was conserved in vivo, as poly(ethylene glycol)-PLGA NPs wrapped in CHRF membranes specifically targeted and were taken up by murine bone marrow HSPCs following intravenous administration. These findings suggest that MkNPs and CHNPs are effective and promising vehicles for targeted cargo delivery to HSPCs.
Collapse
Affiliation(s)
- Samik Das
- Department of Chemical and Biomolecular EngineeringUniversity of DelawareNewarkDelawareUSA
- Delaware Biotechnology InstituteUniversity of DelawareNewarkDelawareUSA
| | - Jenna C. Harris
- Department of Materials Science and EngineeringUniversity of DelawareNewarkDelawareUSA
| | - Erica J. Winter
- Delaware Biotechnology InstituteUniversity of DelawareNewarkDelawareUSA
- Department of Biological SciencesUniversity of DelawareNewarkDelawareUSA
| | - Chen‐Yuan Kao
- Department of Chemical and Biomolecular EngineeringUniversity of DelawareNewarkDelawareUSA
- Delaware Biotechnology InstituteUniversity of DelawareNewarkDelawareUSA
| | - Emily S. Day
- Department of Materials Science and EngineeringUniversity of DelawareNewarkDelawareUSA
- Department of Biomedical EngineeringUniversity of DelawareNewarkDelawareUSA
- Helen F. Graham Cancer Center and Research InstituteNewarkDelawareUSA
| | - Eleftherios Terry Papoutsakis
- Department of Chemical and Biomolecular EngineeringUniversity of DelawareNewarkDelawareUSA
- Delaware Biotechnology InstituteUniversity of DelawareNewarkDelawareUSA
- Department of Biological SciencesUniversity of DelawareNewarkDelawareUSA
| |
Collapse
|
6
|
Thompson W, Papoutsakis ET. The role of biomechanical stress in extracellular vesicle formation, composition and activity. Biotechnol Adv 2023; 66:108158. [PMID: 37105240 DOI: 10.1016/j.biotechadv.2023.108158] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 04/23/2023] [Accepted: 04/24/2023] [Indexed: 04/29/2023]
Abstract
Extracellular vesicles (EVs) are cornerstones of intercellular communication with exciting fundamental, clinical, and more broadly biotechnological applications. However, variability in EV composition, which results from the culture conditions used to generate the EVs, poses significant fundamental and applied challenges and a hurdle for scalable bioprocessing. Thus, an understanding of the relationship between EV production (and for clinical applications, manufacturing) and EV composition is increasingly recognized as important and necessary. While chemical stimulation and culture conditions such as cell density are known to influence EV biology, the impact of biomechanical forces on the generation, properties, and biological activity of EVs remains poorly understood. Given the omnipresence of these forces in EV preparation and in biomanufacturing, expanding the understanding of their impact on EV composition-and thus, activity-is vital. Although several publications have examined EV preparation and bioprocessing and briefly discussed biomechanical stresses as variables of interest, this review represents the first comprehensive evaluation of the impact of such stresses on EV production, composition and biological activity. We review how EV biogenesis, cargo, efficacy, and uptake are uniquely affected by various types, magnitudes, and durations of biomechanical forces, identifying trends that emerge both generically and for individual cell types. We also describe implications for scalable bioprocessing, evaluating processes inherent in common EV production and isolation methods, and propose a path forward for rigorous EV quality control.
Collapse
Affiliation(s)
- Will Thompson
- Department of Chemical and Biomolecular Engineering, University of Delaware, 590 Avenue 1743, Newark, DE 19713, USA
| | - Eleftherios Terry Papoutsakis
- Department of Chemical and Biomolecular Engineering, University of Delaware, 590 Avenue 1743, Newark, DE 19713, USA.
| |
Collapse
|
7
|
Allegra A, Cicero N, Mirabile G, Giorgianni CM, Gangemi S. Novel Biomarkers for Diagnosis and Monitoring of Immune Thrombocytopenia. Int J Mol Sci 2023; 24:ijms24054438. [PMID: 36901864 PMCID: PMC10003036 DOI: 10.3390/ijms24054438] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 02/12/2023] [Accepted: 02/21/2023] [Indexed: 03/12/2023] Open
Abstract
Lower-than-normal platelet counts are a hallmark of the acquired autoimmune illness known as immune thrombocytopenia, which can affect both adults and children. Immune thrombocytopenia patients' care has evolved significantly in recent years, but the disease's diagnosis has not, and it is still only clinically achievable with the elimination of other causes of thrombocytopenia. The lack of a valid biomarker or gold-standard diagnostic test, despite ongoing efforts to find one, adds to the high rate of disease misdiagnosis. However, in recent years, several studies have helped to elucidate a number of features of the disease's etiology, highlighting how the platelet loss is not only caused by an increase in peripheral platelet destruction but also involves a number of humoral and cellular immune system effectors. This made it possible to identify the role of immune-activating substances such cytokines and chemokines, complement, non-coding genetic material, the microbiome, and gene mutations. Furthermore, platelet and megakaryocyte immaturity indices have been emphasized as new disease markers, and prognostic signs and responses to particular types of therapy have been suggested. Our review's goal was to compile information from the literature on novel immune thrombocytopenia biomarkers, markers that will help us improve the management of these patients.
Collapse
Affiliation(s)
- Alessandro Allegra
- Division of Hematology, Department of Human Pathology in Adulthood and Childhood “Gaetano Barresi”, University of Messina, 98100 Messina, Italy
- Correspondence:
| | - Nicola Cicero
- Department of Biomedical, Dental, Morphological and Functional Imaging Sciences (BIOMORF), University of Messina, 98100 Messina, Italy
| | - Giuseppe Mirabile
- Division of Hematology, Department of Human Pathology in Adulthood and Childhood “Gaetano Barresi”, University of Messina, 98100 Messina, Italy
| | - Concetto Mario Giorgianni
- Department of Biomedical, Dental, Morphological and Functional Imaging Sciences (BIOMORF), University of Messina, 98100 Messina, Italy
| | - Sebastiano Gangemi
- Allergy and Clinical Immunology Unit, Department of Clinical and Experimental Medicine, University of Messina, 98100 Messina, Italy
| |
Collapse
|
8
|
Megakaryocyte- and Platelet-Derived Microparticles as Novel Diagnostic and Prognostic Biomarkers for Immune Thrombocytopenia. J Clin Med 2022; 11:jcm11226776. [PMID: 36431253 PMCID: PMC9698595 DOI: 10.3390/jcm11226776] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 11/04/2022] [Accepted: 11/14/2022] [Indexed: 11/19/2022] Open
Abstract
Altered cell-derived microparticles (MPs) have been reported in multiple autoimmune diseases. However, the roles of megakaryocyte- and platelet-derived MPs (MKMPs and PMPs) in immune thrombocytopenia (ITP) have not been investigated. In this study, we examined plasma MKMP and PMP levels in patients with ITP and evaluated their potential diagnostic values. Plasma MKMP and PMP levels were analyzed by flow cytometry in a discovery set of ITP patients (n = 78), non-immune thrombocytopenia (TP) patients (n = 69), and age- and gender-matched healthy controls (n = 88). Samples from a therapy set of ITP patients (n = 21) were used to assess the response to thrombopoietin receptor agonist (TPO-RA) treatment. Spearman correlation analysis was performed between MP levels and disease parameters. Receiver operator characteristic (ROC) curves were generated to evaluate the diagnostic values of the MPs. We found that plasma MKMP and PMP levels were significantly lower in ITP patients than those in healthy controls (p values < 0.0001) but higher than in those in TP patients (p < 0.002 and p < 0.0002, respectively). After normalization to platelet counts, PMP/Platelet ratios in ITP patients were higher than those in TP patients and healthy controls (p values < 0.001). PMP/Platelet ratios had a diagnostic value for ITP (area under the curve = 0.808, p < 0.0001) with 73.1% sensitivity and 77.3% specificity. MKMP levels can be used to discriminate ITP from TP with a cut-off value of 112.5 MPs/μL and a sensitivity of 74.4%. Moreover, both MKMP and PMP levels were elevated in ITP patients who responded to TPO-RA treatment. Plasma PMP levels positively correlated with platelet counts in the responders (r = 0.558, p < 0.01). Our results indicate that plasma MKMP and PMP levels are decreased in ITP patients and that plasma MKMP and PMP levels may serve as biomarkers for ITP diagnosis and prediction of TPO-RA treatment response.
Collapse
|
9
|
Harris JC, Sterin EH, Day ES. Membrane-Wrapped Nanoparticles for Enhanced Chemotherapy of Acute Myeloid Leukemia. ACS Biomater Sci Eng 2022; 8:4439-4448. [PMID: 36103274 PMCID: PMC9633094 DOI: 10.1021/acsbiomaterials.2c00832] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
This work reports the development of a biomimetic membrane-wrapped nanoparticle (MWNP) platform for targeted chemotherapy of acute myeloid leukemia (AML). Doxorubicin (DOX), a chemotherapeutic used to treat leukemias, lymphomas, and other cancers, was encapsulated in polymeric NPs that were coated with cytoplasmic membranes derived from human AML cells. The release rate of DOX from the MWNPs was characterized under both storage and physiological conditions, with faster release observed at pH 5.5 than pH 7.4. The system was then introduced to AML cell cultures to test the functionality of the released DOX cargo as compared to DOX delivered freely or via NPs coated with poly(ethylene glycol) (PEG). The MWNPs delivered DOX in an efficient and targeted manner, inducing up to 80% apoptosis in treated cells at a dose of 5 μM, compared to 15% for free DOX and 17% for DOX-loaded PEG-coated NPs at the same drug concentration. The mechanism of cell death was confirmed as DNA double-strand breaks through a γH2A.X assay, indicating that the released DOX retained its expected mechanism of action. These findings designate MWNPs as a robust drug delivery system with great potential for future development in treatments of AML and other blood cancers.
Collapse
Affiliation(s)
- Jenna C Harris
- Department of Materials Science and Engineering, University of Delaware, 201 DuPont Hall, Newark, Delaware 19716, United States
| | - Eric H Sterin
- Department of Biomedical Engineering, University of Delaware, 590 Avenue 1743, 4th Floor, Newark, Delaware 19713, United States
| | - Emily S Day
- Department of Materials Science and Engineering, University of Delaware, 201 DuPont Hall, Newark, Delaware 19716, United States
- Department of Biomedical Engineering, University of Delaware, 590 Avenue 1743, 4th Floor, Newark, Delaware 19713, United States
- Helen F. Graham Cancer Center and Research Institute, 4701 Ogletown-Stanton Road, Newark, Delaware 19713, United States
| |
Collapse
|
10
|
Desai C, Koupenova M, Machlus KR, Sen Gupta A. Beyond the thrombus: Platelet-inspired nanomedicine approaches in inflammation, immune response, and cancer. J Thromb Haemost 2022; 20:1523-1534. [PMID: 35441793 PMCID: PMC9321119 DOI: 10.1111/jth.15733] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 04/13/2022] [Accepted: 04/14/2022] [Indexed: 12/03/2022]
Abstract
The traditional role of platelets is in the formation of blood clots for physiologic (e.g., in hemostasis) or pathologic (e.g., in thrombosis) functions. The cellular and subcellular mechanisms and signaling in platelets involved in these functions have been extensively elucidated and new knowledge continues to emerge, resulting in various therapeutic developments in this area for the management of hemorrhagic or thrombotic events. Nanomedicine, a field involving design of nanoparticles with unique biointeractive surface modifications and payload encapsulation for disease-targeted drug delivery, has become an important component of such therapeutic development. Beyond their traditional role in blood clotting, platelets have been implicated to play crucial mechanistic roles in other diseases including inflammation, immune response, and cancer, via direct cellular interactions, as well as secretion of soluble factors that aid in the disease microenvironment. To date, the development of nanomedicine systems that leverage these broader roles of platelets has been limited. Additionally, another exciting area of research that has emerged in recent years is that of platelet-derived extracellular vesicles (PEVs) that can directly and indirectly influence physiological and pathological processes. This makes PEVs a unique paradigm for platelet-inspired therapeutic design. This review aims to provide mechanistic insight into the involvement of platelets and PEVs beyond hemostasis and thrombosis, and to discuss the current state of the art in the development of platelet-inspired therapeutic technologies in these areas, with an emphasis on future opportunities.
Collapse
Affiliation(s)
- Cian Desai
- Department of PharmacologyCase Western Reserve UniversityClevelandOhioUSA
| | - Milka Koupenova
- Division of Cardiovascular MedicineDepartment of MedicineUniversity of Massachusetts Chan Medical SchoolWorcesterMassachusettsUSA
| | - Kellie R. Machlus
- Department of SurgeryVascular Biology ProgramBoston Children's HospitalHarvard Medical SchoolBostonMassachusettsUSA
| | - Anirban Sen Gupta
- Department of PharmacologyCase Western Reserve UniversityClevelandOhioUSA
- Department of Biomedical EngineeringCase Western Reserve UniversityClevelandOhioUSA
| |
Collapse
|
11
|
Stone AP, Nikols E, Freire D, Machlus KR. The pathobiology of platelet and megakaryocyte extracellular vesicles: A (c)lot has changed. J Thromb Haemost 2022; 20:1550-1558. [PMID: 35506218 DOI: 10.1111/jth.15750] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 04/26/2022] [Accepted: 05/02/2022] [Indexed: 11/28/2022]
Abstract
Platelet-derived extracellular vesicles (PEVs) were originally studied for their potential as regulators of coagulation, a function redundant with that of their parent cells. However, as the understanding of the diverse roles of platelets in hemostasis and disease has developed, so has the understanding of PEVs. In addition, the more recent revelation of constitutively released megakaryocyte-derived extracellular vesicles (MKEVs) in circulation provides an interesting counterpoint and avenue for investigation. In this review, we highlight the historical link of PEVs to thrombosis and hemostasis and provide critical updates. We also expand our discussion to encompass the roles that distinguish PEVs and MKEVs from their parent cells. Furthermore, the role of extracellular vesicles in disease pathology, both as biomarkers and as exacerbators, has been of great interest in recent years. We highlight some of the key roles that PEVs and MKEVs play in autoimmune blood cell disorders, liver pathology, and cardiovascular disease. We then look at the future of PEVs and MKEVs as candidates for novel therapeutics.
Collapse
Affiliation(s)
- Andrew P Stone
- Vascular Biology Program, Boston Children's Hospital, Boston, Massachusetts, USA
- Department of Surgery, Harvard Medical School, Boston, Massachusetts, USA
| | - Emma Nikols
- Vascular Biology Program, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Daniela Freire
- Vascular Biology Program, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Kellie R Machlus
- Vascular Biology Program, Boston Children's Hospital, Boston, Massachusetts, USA
- Department of Surgery, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
12
|
Kao CY, Jiang J, Thompson W, Papoutsakis ET. miR-486-5p and miR-22-3p Enable Megakaryocytic Differentiation of Hematopoietic Stem and Progenitor Cells without Thrombopoietin. Int J Mol Sci 2022; 23:ijms23105355. [PMID: 35628168 PMCID: PMC9141330 DOI: 10.3390/ijms23105355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2022] [Accepted: 05/07/2022] [Indexed: 12/10/2022] Open
Abstract
Megakaryocytes release submicron size microparticles (MkMPs) in circulation. We have shown that MkMPs target CD34+ hematopoietic stem/progenitor cells (HSPCs) to induce megakaryocytic differentiation, and that small RNAs in MkMPs play an important role in the development of this phenotype. Here, using single-molecule real-time (SMRT) RNA sequencing (RNAseq), we identify the synergetic effect of two microRNAs (miRs), miR-486-5p and miR-22-3p (highly enriched in MkMPs), in driving the Mk differentiation of HSPCs in the absence of thrombopoietin (TPO). Separately, our data suggest that the MkMP-induced Mk differentiation of HSPCs is enabled through JNK and PI3K/Akt/mTOR signaling. The interaction between the two signaling pathways is likely mediated by a direct target of miR-486-5p and a negative regulator of PI3K/Akt signaling, the phosphatase and tensin homologue (PTEN) protein. Our data provide a possible mechanistic explanation of the biological effect of MkMPs in inducing megakaryocytic differentiation of HSPCs, a phenotype of potential physiological significance in stress megakaryopoiesis.
Collapse
Affiliation(s)
- Chen-Yuan Kao
- Department of Chemical and Biomolecular Engineering, University of Delaware, 590 Ave. 1743, Newark, DE 19713, USA; (C.-Y.K.); (J.J.); (W.T.)
| | - Jinlin Jiang
- Department of Chemical and Biomolecular Engineering, University of Delaware, 590 Ave. 1743, Newark, DE 19713, USA; (C.-Y.K.); (J.J.); (W.T.)
| | - Will Thompson
- Department of Chemical and Biomolecular Engineering, University of Delaware, 590 Ave. 1743, Newark, DE 19713, USA; (C.-Y.K.); (J.J.); (W.T.)
| | - Eleftherios T. Papoutsakis
- Department of Chemical and Biomolecular Engineering, University of Delaware, 590 Ave. 1743, Newark, DE 19713, USA; (C.-Y.K.); (J.J.); (W.T.)
- Department of Biological Sciences, University of Delaware, 590 Ave. 1743, Newark, DE 19713, USA
- Correspondence: ; Tel.: +1-302-831-8376
| |
Collapse
|
13
|
Belliveau J, Papoutsakis ET. Extracellular Vesicles Facilitate Large-Scale Dynamic Exchange of Proteins and RNA Among Cultured Chinese Hamster Ovary (CHO) and Human Cells. Biotechnol Bioeng 2022; 119:1222-1238. [PMID: 35120270 DOI: 10.1002/bit.28053] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Accepted: 01/31/2022] [Indexed: 11/11/2022]
Abstract
Cells in culture are viewed as unique individuals in a large population communicating through extracellular molecules and, more recently extracellular vesicles (EVs). Our data here paint a different picture: large-scale exchange of cellular material through EVs. To visualize the dynamic production and cellular uptake of EVs, we used correlative confocal microscopy and scanning electron microscopy, as well as flow cytometry to interrogate labeled cells. Using cells expressing fluorescent proteins (GFP, miRFP703) and cells tagged with protein and RNA dyes, we show that Chinese Hamster Ovary (CHO) cells dynamically produce and uptake EVs to exchange proteins and RNAs at a large scale. Applying a simple model to our data, we estimate, for the first time, the per cell specific rates of EV production (68 and 203 microparticles and exosomes, respectively, per day). This EV-mediated massive exchange of cellular material observed in CHO cultures was also observed in cultured human CHRF-288-11 and primary hematopoietic stem and progenitor cells. This study demonstrates an underappreciated massive protein and RNA exchange between cells mediated by EVs spanning cell type, suggesting that the proximity of cells in normal and tumor tissues may also result in prolific exchange of cellular material. This exchange would be expected to homogenize the cell-population cytosol and dynamically regulate cell proliferation and the cellular state. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Jessica Belliveau
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, DE, 19711.,Delaware Biotechnology Institute,, University of Delaware, Newark, DE, 19711
| | - Eleftherios T Papoutsakis
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, DE, 19711.,Delaware Biotechnology Institute,, University of Delaware, Newark, DE, 19711.,Department of Biological Sciences, University of Delaware, Newark, DE, 19711
| |
Collapse
|
14
|
Italiano JE, Bender M, Merrill-Skoloff G, Ghevaert C, Nieswandt B, Flaumenhaft R. Microvesicles, but not platelets, bud off from mouse bone marrow megakaryocytes. Blood 2021; 138:1998-2001. [PMID: 34324659 PMCID: PMC8602935 DOI: 10.1182/blood.2021012496] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Accepted: 07/02/2021] [Indexed: 11/20/2022] Open
Affiliation(s)
- Joseph E Italiano
- Vascular Biology Program, Boston Children's Hospital, Harvard Medical School, Boston, MA
| | - Markus Bender
- Institute of Experimental Biomedicine I, University Hospital, Würzburg, Germany
- Rudolf Virchow Center, University of Würzburg, Würzburg, Germany
| | - Glenn Merrill-Skoloff
- Division of Hemostasis and Thrombosis, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA; and
| | - Cedric Ghevaert
- Wellcome-Medical Research Council Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, Department of Haematology, University of Cambridge, Cambridge, United Kingdom
| | - Bernhard Nieswandt
- Institute of Experimental Biomedicine I, University Hospital, Würzburg, Germany
- Rudolf Virchow Center, University of Würzburg, Würzburg, Germany
| | - Robert Flaumenhaft
- Division of Hemostasis and Thrombosis, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA; and
| |
Collapse
|
15
|
Tang A, Mendelson A. Recent lessons learned for ex-vivo platelet production. Curr Opin Hematol 2021; 28:424-430. [PMID: 34232141 PMCID: PMC8490274 DOI: 10.1097/moh.0000000000000662] [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: 11/25/2022]
Abstract
PURPOSE OF REVIEW Platelet transfusion can be life-saving but carries a risk of infection or alloimmunization and is limited by insufficient donor sources and restricted unit shelf life. Generating sufficient platelets in vitro to replace a unit of collected blood remains a challenge. Here, we examine the latest advances in the regulation of megakaryocyte maturation and expansion along with platelet formation and survival. We also discuss alternative therapies investigated to induce platelet production. RECENT FINDINGS Recent studies examined candidate niche cells in the bone marrow microenvironment for promoting platelet formation and developed an explant-based bioreactor to enhance platelet production ex vivo. Chemical inhibitors were examined for their ability to promote megakaryocyte maturation and expansion. Microparticles from megakaryocytes or platelets were found to improve megakaryocyte maturation and platelet formation. Membrane budding was identified as a novel mode of platelet formation. Lastly, a chemical inhibitor to improve cold-stored platelets was identified. SUMMARY Recent advances in the regulation of megakaryocyte expansion and platelet production provide exciting promise for the development of improved approaches to generate platelets in vitro. These findings bring the field one step closer to achieving the ultimate goal of creating a unit of platelets without the need for donation.
Collapse
Affiliation(s)
- Alice Tang
- Lindsley F. Kimball Research Institute, New York Blood Center, New York, NY
| | - Avital Mendelson
- Lindsley F. Kimball Research Institute, New York Blood Center, New York, NY
| |
Collapse
|
16
|
Potential Diagnostic Approaches for Prediction of Therapeutic Responses in Immune Thrombocytopenia. J Clin Med 2021; 10:jcm10153403. [PMID: 34362187 PMCID: PMC8347743 DOI: 10.3390/jcm10153403] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 07/27/2021] [Accepted: 07/28/2021] [Indexed: 12/13/2022] Open
Abstract
Immune thrombocytopenia (ITP) is an autoimmune bleeding disorder in which, via unresolved mechanisms, platelets and megakaryocytes (MKs) are targeted by autoantibodies and/or T cells resulting in increased platelet destruction and impairment of MK function. Over the years, several therapeutic modalities have become available for ITP, however, therapeutic management has proven to be very challenging in several cases. Patients refractory to treatment can develop a clinically worsening disease course, treatment-induced toxicities and are predisposed to development of potentially life-endangering bleedings. It is therefore of critical importance to timely identify potential refractory patients, for which novel diagnostic approaches are urgently needed in order to monitor and predict specific therapeutic responses. In this paper, we propose promising diagnostic investigations into immune functions and characteristics in ITP, which may potentially be exploited to help predict platelet count responses and thereby distinguish therapeutic responders from non-responders. This importantly includes analysis of T cell homeostasis, which generally appears to be disturbed in ITP due to decreased and/or dysfunctional T regulatory cells (Tregs) leading to loss of immune tolerance and initiation/perpetuation of ITP, and this may be normalized by several therapeutic modalities. Additional avenues to explore in possible prediction of therapeutic responses include examination of platelet surface sialic acids, platelet apoptosis, monocyte surface markers, B regulatory cells and platelet microparticles. Initial studies have started evaluating these markers in relation to response to various treatments including glucocorticosteroids (GCs), intravenous immunoglobulins (IVIg) and/or thrombopoietin receptor agonists (TPO-RA), however, further studies are highly warranted. The systematic molecular analysis of a broad panel of immune functions may ultimately help guide and improve personalized therapeutic management in ITP.
Collapse
|
17
|
Abstract
[Figure: see text].
Collapse
|
18
|
Platelet-derived extracellular vesicles infiltrate and modify the bone marrow during inflammation. Blood Adv 2021; 4:3011-3023. [PMID: 32614966 DOI: 10.1182/bloodadvances.2020001758] [Citation(s) in RCA: 70] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Accepted: 05/26/2020] [Indexed: 12/18/2022] Open
Abstract
During inflammation, steady-state hematopoiesis switches to emergency hematopoiesis to repopulate myeloid cells, with a bias toward the megakaryocytic lineage. Soluble inflammatory cues are thought to be largely responsible for these alterations. However, how these plasma factors rapidly alter the bone marrow (BM) is not understood. Inflammation also drives platelet activation, causing the release of platelet-derived extracellular vesicles (PEVs), which package diverse cargo and reprogram target cells. We hypothesized that PEVs infiltrate the BM, providing a direct mode of communication between the plasma and BM environments. We transfused fluorescent, wild-type (MPL+) platelets into recipient cMpl-/-mice before triggering systemic inflammation. Twenty hours postinfusion, we observed significant infiltration of donor platelet-derived particles in the BM, which we tracked immunophenotypically (MPL+ immunohistochemistry staining) and quantified by flow cytometry. To determine if this phenomenon relates to humans, we extensively characterized both megakaryocyte-derived and PEVs generated in vitro and in vivo, and found enrichment of extracellular vesicles in bone marrow compared with autologous peripheral blood. Last, BM from cMpl-/- mice was cultured in the presence or absence of wild-type (MPL+) PEVs. After 72 hours, flow cytometry revealed increased megakaryocytes only in cultures with added PEVs. The majority of CD41+ cells were bound to PEVs, suggesting a PEV-mediated rescue of megakaryopoiesis. In conclusion, we report for the first time that plasma-residing PEVs infiltrate the BM. Further, PEVs interact with BM cells in vivo and in vitro, causing functional reprogramming that may represent a novel model of inflammation-induced hematopoiesis.
Collapse
|
19
|
Kapur R, Semple JW. Megakaryocytes listen for their progeny's progeny during inflammation. J Thromb Haemost 2021; 19:604-606. [PMID: 33346949 PMCID: PMC7986206 DOI: 10.1111/jth.15178] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 11/12/2020] [Accepted: 11/12/2020] [Indexed: 12/25/2022]
Affiliation(s)
- Rick Kapur
- Sanquin ResearchDepartment of Experimental ImmunohematologyAmsterdam and Landsteiner LaboratoryAmsterdam UMCUniversity of AmsterdamAmsterdamthe Netherlands
| | - John W. Semple
- Division of Hematology and Transfusion MedicineLund UniversityLundSweden
- Departments of Pharmacology and MedicineUniversity of TorontoTorontoONCanada
| |
Collapse
|
20
|
Bian W, Chen W, Jiang X, Qu H, Jiang J, Yang J, Liang X, Zhao B, Sun Y, Zhang C. Downregulation of Long Non-coding RNA Nuclear Paraspeckle Assembly Transcript 1 Inhibits MEG-01 Differentiation and Platelet-Like Particles Activity. Front Genet 2020; 11:571467. [PMID: 33193674 PMCID: PMC7596361 DOI: 10.3389/fgene.2020.571467] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Accepted: 09/22/2020] [Indexed: 01/22/2023] Open
Abstract
Platelets are derived from megakaryocytes and play an important role in blood coagulation. By using high throughput sequencing, we have found that the long non-coding RNA (lncRNA) nuclear paraspeckle assembly transcript 1 (NEAT1) is abundant in platelets (GEO ID: 200097348). However, little is known about its role in regulating megakaryocyte differentiation and platelet activity. This study aims to clarify the effect of NEAT1 on MEG-01 differentiation and platelet-like particle (PLP) activity. NEAT1 in MEG-01 cells was knocked down by siRNA transfection. The adhesion of MEG-01 and PLP to collagen-coated coverslips was observed under a fluorescence microscope. Flow cytometry was used to investigate cell apoptosis, cell cycle, the levels of D41/CD42b on MEG-01 cells and CD62P on PLPs. Quantitative real-time polymerase chain reaction was used to detect NEAT1 and IL-8 expression levels. Western blot was used to measure the protein levels of Bcl-2, Bax, cleaved caspase-3, and IL-8. RNA-binding protein immunoprecipitation was used to detect the interaction of NEAT1 and splicing factor proline/glutamine-rich (SFPQ). Results showed that NEAT1 knockdown decreased the adhesion ability of thrombin-stimulated MEG-01 and PLP. The expression of CD62P on PLPs and CD41/CD42b on MEG-01 cells was inhibited by NEAT1 knockdown. In addition, NEAT1 knockdown inhibited cell apoptosis with increased Bcl2/Bax ratio and decreased cleaved caspase-3, and reduced the percentage of cells in the G0/G1 phase. Meanwhile, NEAT1 knockdown inhibited the expression of IL-8. A strong interaction of NEAT1 and SFPQ, a transcriptional repressor of IL-8, was identified. NEAT1 knockdown reduced the interaction between SFPQ and NEAT1.The results suggest that lncRNA NEAT1 knockdown decreases MEG-01 differentiation, PLP activity, and IL-8 level. The results also indicate that the regulation of NEAT1 on IL-8 may be realized via a direct interaction between NEAT1 and SFPQ.
Collapse
Affiliation(s)
- Weihua Bian
- School of Pharmacy, Binzhou Medical University, Yantai, China
| | - Wangping Chen
- Department of Cardiovascular Surgery, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Xiaoli Jiang
- School of Pharmacy, Binzhou Medical University, Yantai, China
| | - Huiqing Qu
- Department of Blood Transfusion, Affiliated Hospital of Binzhou Medical University, Binzhou, China
| | - Jing Jiang
- School of Pharmacy, Binzhou Medical University, Yantai, China
| | - Jinfu Yang
- Department of Cardiovascular Surgery, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Xinyue Liang
- School of Pharmacy, Binzhou Medical University, Yantai, China
| | - Bingrui Zhao
- School of Pharmacy, Binzhou Medical University, Yantai, China
| | - Yeying Sun
- School of Pharmacy, Binzhou Medical University, Yantai, China
| | - Chunxiang Zhang
- School of Pharmacy, Binzhou Medical University, Yantai, China
| |
Collapse
|