1
|
Ritter P, Oliveto S, Cordiglieri C, Fasciani A, Di Buduo CA, della Volpe L, Bocconi A, Conci C, Miguel CP, Di Micco R, Balduini A, Raimondi MT, Biffo S. A millifluidic bioreactor allows the long term culture of primary lymphocytes or CD34 + hematopoietic cells while allowing the detection of tumorigenic expansion. Front Bioeng Biotechnol 2024; 12:1388312. [PMID: 39416278 PMCID: PMC11479935 DOI: 10.3389/fbioe.2024.1388312] [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: 02/19/2024] [Accepted: 09/19/2024] [Indexed: 10/19/2024] Open
Abstract
Long-term culture of primary lymphocytes and hematopoietic stem and progenitor cells (HSPCs) is pivotal to their expansion and study. Furthermore, genetic engineering of the above-mentioned primary human cells has several safety needs, including the requirement of efficient in vitro assays for unwanted tumorigenic events. In this work, we tested and optimized the Miniaturized Optically Accessible Bioreactor (MOAB) platform. The MOAB consists of a millifluidic cell culture device with three optically-accessible culture chambers. Inside the MOAB, we inserted a silk-based framework that resembles some properties of the bone marrow environment and cultivated in this device either CD4+ T lymphocytes isolated from healthy donor buffy coat or cord blood-derived hematopoietic CD34+ cells. A fraction of these cells is viable for up to 3 months. Next, we tested the capability of the MOAB to detect tumorigenic events. Serial dilutions of engineered fluorescent tumor cells were mixed with either CD4+ or CD34+ primary cells, and their growth was followed. By this approach, we successfully detected as little as 100 tumorigenic cells mixed with 100,000 primary cells. We found that non-tumorigenic primary cells colonized the silk environment, whereas tumor cells, after an adaptation phase, expanded and entered the circulation. We conclude that the millifluidic platform allows the detection of rare tumorigenic events in the long-term culture of human cells.
Collapse
Affiliation(s)
- Paolo Ritter
- National Institute of Molecular Genetics, Fondazione Romeo ed Enrica Invernizzi, INGM, Milan, Italy
- Department of Chemistry, Materials and Chemical Engineering “Giulio Natta”, Politecnico di Milano, Milan, Italy
| | - Stefania Oliveto
- National Institute of Molecular Genetics, Fondazione Romeo ed Enrica Invernizzi, INGM, Milan, Italy
- Department of Biosciences, University of Milan, Milan, Italy
| | - Chiara Cordiglieri
- National Institute of Molecular Genetics, Fondazione Romeo ed Enrica Invernizzi, INGM, Milan, Italy
| | - Alessandra Fasciani
- National Institute of Molecular Genetics, Fondazione Romeo ed Enrica Invernizzi, INGM, Milan, Italy
| | | | - Lucrezia della Volpe
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Alberto Bocconi
- Department of Chemistry, Materials and Chemical Engineering “Giulio Natta”, Politecnico di Milano, Milan, Italy
| | - Claudio Conci
- Department of Chemistry, Materials and Chemical Engineering “Giulio Natta”, Politecnico di Milano, Milan, Italy
| | | | - Raffaella Di Micco
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy
- University School for Advanced Studies IUSS, Pavia, Italy
| | | | - Manuela Teresa Raimondi
- Department of Chemistry, Materials and Chemical Engineering “Giulio Natta”, Politecnico di Milano, Milan, Italy
| | - Stefano Biffo
- National Institute of Molecular Genetics, Fondazione Romeo ed Enrica Invernizzi, INGM, Milan, Italy
- Department of Biosciences, University of Milan, Milan, Italy
| |
Collapse
|
2
|
Di Buduo CA, Lunghi M, Kuzmenko V, Laurent P, Della Rosa G, Del Fante C, Dalle Nogare DE, Jug F, Perotti C, Eto K, Pecci A, Redwan IN, Balduini A. Bioprinting Soft 3D Models of Hematopoiesis using Natural Silk Fibroin-Based Bioink Efficiently Supports Platelet Differentiation. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2308276. [PMID: 38514919 PMCID: PMC11095152 DOI: 10.1002/advs.202308276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 02/09/2024] [Indexed: 03/23/2024]
Abstract
Hematopoietic stem and progenitor cells (HSPCs) continuously generate platelets throughout one's life. Inherited Platelet Disorders affect ≈ 3 million individuals worldwide and are characterized by defects in platelet formation or function. A critical challenge in the identification of these diseases lies in the absence of models that facilitate the study of hematopoiesis ex vivo. Here, a silk fibroin-based bioink is developed and designed for 3D bioprinting. This bioink replicates a soft and biomimetic environment, enabling the controlled differentiation of HSPCs into platelets. The formulation consisting of silk fibroin, gelatin, and alginate is fine-tuned to obtain a viscoelastic, shear-thinning, thixotropic bioink with the remarkable ability to rapidly recover after bioprinting and provide structural integrity and mechanical stability over long-term culture. Optical transparency allowed for high-resolution imaging of platelet generation, while the incorporation of enzymatic sensors allowed quantitative analysis of glycolytic metabolism during differentiation that is represented through measurable color changes. Bioprinting patient samples revealed a decrease in metabolic activity and platelet production in Inherited Platelet Disorders. These discoveries are instrumental in establishing reference ranges for classification and automating the assessment of treatment responses. This model has far-reaching implications for application in the research of blood-related diseases, prioritizing drug development strategies, and tailoring personalized therapies.
Collapse
Affiliation(s)
| | - Marco Lunghi
- Department of Molecular MedicineUniversity of PaviaPavia27100Italy
| | | | | | | | - Claudia Del Fante
- Immunohaematology and Transfusion ServiceI.R.C.C.S. Policlinico S. Matteo FoundationPavia27100Italy
| | | | | | - Cesare Perotti
- Immunohaematology and Transfusion ServiceI.R.C.C.S. Policlinico S. Matteo FoundationPavia27100Italy
| | - Koji Eto
- Department of Clinical ApplicationCenter for iPS Cell Research and Application (CiRA)Kyoto UniversityKyoto606‐8507Japan
- Department of Regenerative MedicineGraduate School of MedicineChiba UniversityChiba260‐8670Japan
| | - Alessandro Pecci
- Department of Internal MedicineI.R.C.C.S. Policlinico S. Matteo Foundation and University of PaviaPavia27100Italy
| | | | - Alessandra Balduini
- Department of Molecular MedicineUniversity of PaviaPavia27100Italy
- Department of Biomedical EngineeringTufts UniversityMedfordMA02155USA
| |
Collapse
|
3
|
Toni R, Barbaro F, Di Conza G, Zini N, Remaggi G, Elviri L, Spaletta G, Quarantini E, Quarantini M, Mosca S, Caravelli S, Mosca M, Ravanetti F, Sprio S, Tampieri A. A bioartificial and vasculomorphic bone matrix-based organoid mimicking microanatomy of flat and short bones. J Biomed Mater Res B Appl Biomater 2024; 112:e35329. [PMID: 37898921 DOI: 10.1002/jbm.b.35329] [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: 04/03/2023] [Revised: 08/08/2023] [Accepted: 09/07/2023] [Indexed: 10/31/2023]
Abstract
We engineered an in vitro model of bioartificial 3D bone organoid consistent with an anatomical and vascular microenvironment common to mammalian flat and short bones. To achieve this, we chose the decellularized-decalcified matrix of the adult male rat scapula, implemented with the reconstruction of its intrinsic vessels, obtained through an original intravascular perfusion with polylevolactic (PLLA), followed by coating of the PLLA-fabricated vascularization with rat tail collagen. As a result, the 3D bone and vascular geometry of the native bone cortical and cancellous compartments was reproduced, and the rat tail collagen-PLLA biomaterial could in vitro act as a surrogate of the perivascular extracellular matrix (ECM) around the wall of the biomaterial-reconstituted cancellous vessels. As a proof-of-concept of cell compatibility and site-dependent osteoinductive properties of this bioartificial 3D construct, we show that it in vitro leads to a time-dependent microtopographic positioning of rat mesenchymal stromal cells (MSCs), initiating an osteogenic fate in relation to the bone compartment. In addition, coating of PLLA-reconstructed vessels with rat tail collagen favored perivascular attachment and survival of MSC-like cells (mouse embryonic fibroblasts), confirming its potentiality as a perivascular stroma for triggering competence of seeded MSCs. Finally, in vivo radiographic topography of bone lesions in the human jaw and foot tarsus of subjects with primary osteoporosis revealed selective bone cortical versus cancellous involvement, suggesting usefulness of a human 3D bone organoid engineered with the same principles of our rat organoid, to in vitro investigate compartment-dependent activities of human MSC in flat and short bones under experimental osteoporotic challenge. We conclude that our 3D bioartificial construct offers a reliable replica of flat and short bones microanatomy, and promises to help in building a compartment-dependent mechanistic perspective of bone remodeling, including the microtopographic dysregulation of osteoporosis.
Collapse
Affiliation(s)
- Roberto Toni
- ISSMC, CNR, Faenza, Italy
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, Tufts Medical Center-Tufts University School of Medicine, Boston, Massachusetts, USA
- Academy of Sciences of the Institute of Bologna, Section IV-Medical Sciences, Bologna, Italy
- Endocrinology, Diabetes, and Nutrition Disorders Outpatient Clinic-OSTEONET (Osteoporosis, Nutrition, Endocrinology, and Innovative Therapies) and Odontostomatology Units, Galliera Medical Center, San Venanzio di Galliera (BO), Italy
| | - Fulvio Barbaro
- Department of Medicine and Surgery-DIMEC, Unit of Biomedical, Biotechnological and Translational Sciences (S.BI.BI.T.), Laboratory of Regenerative Morphology and Bioartificial Structures (Re.Mo.Bio.S.), and Museum and Historical Library of Biomedicine-BIOMED, University of Parma, Parma, Italy
| | - Giusy Di Conza
- Department of Medicine and Surgery-DIMEC, Unit of Biomedical, Biotechnological and Translational Sciences (S.BI.BI.T.), Laboratory of Regenerative Morphology and Bioartificial Structures (Re.Mo.Bio.S.), and Museum and Historical Library of Biomedicine-BIOMED, University of Parma, Parma, Italy
| | - Nicoletta Zini
- CNR Institute of Molecular Genetics "Luigi Luca Cavalli-Sforza", Unit of Bologna, Bologna, Italy
- IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Giulia Remaggi
- Food and Drug Department, University of Parma, Parma, Italy
| | - Lisa Elviri
- Food and Drug Department, University of Parma, Parma, Italy
| | - Giulia Spaletta
- Department of Statistical Sciences, University of Bologna, Bologna, Italy
| | - Enrico Quarantini
- Endocrinology, Diabetes, and Nutrition Disorders Outpatient Clinic-OSTEONET (Osteoporosis, Nutrition, Endocrinology, and Innovative Therapies) and Odontostomatology Units, Galliera Medical Center, San Venanzio di Galliera (BO), Italy
| | - Marco Quarantini
- Endocrinology, Diabetes, and Nutrition Disorders Outpatient Clinic-OSTEONET (Osteoporosis, Nutrition, Endocrinology, and Innovative Therapies) and Odontostomatology Units, Galliera Medical Center, San Venanzio di Galliera (BO), Italy
| | - Salvatore Mosca
- Course on Disorders of the Locomotor System, Fellow Program in Orthopaedics and Traumatology, University Vita-Salute San Raffaele, Milan, Italy
| | - Silvio Caravelli
- II Clinic of Orthopedic and Traumatology, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Massimiliano Mosca
- II Clinic of Orthopedic and Traumatology, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Francesca Ravanetti
- Department of Veterinary Medical Sciences, Section of Anatomy, University of Parma, Parma, Italy
| | | | | |
Collapse
|
4
|
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
|
5
|
Lau K, Waterhouse A, Akhavan B, Gao L, Kim HN, Tang F, Whitelock JM, Bilek MM, Lord MS, Rnjak-Kovacina J. Biomimetic silk biomaterials: Perlecan-functionalized silk fibroin for use in blood-contacting devices. Acta Biomater 2021; 132:162-175. [PMID: 33588126 DOI: 10.1016/j.actbio.2021.02.014] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 01/22/2021] [Accepted: 02/08/2021] [Indexed: 12/16/2022]
Abstract
Blood compatible materials are required for the development of therapeutic and diagnostic blood contacting devices as blood-material interactions are a key factor dictating device functionality. In this work, we explored biofunctionalization of silk biomaterials with a recombinantly expressed domain V of the human basement membrane proteoglycan perlecan (rDV) towards the development of blood compatible surfaces. Perlecan and rDV are of interest in vascular device development as they uniquely support endothelial cell, while inhibiting smooth muscle cell and platelet interactions. rDV was covalently immobilized on silk biomaterials using plasma immersion ion implantation (PIII), a new method of immobilizing proteins on silk biomaterials that does not rely on modification of specific amino acids in the silk protein chain, and compared to physisorbed and carbodiimide immobilized rDV. Untreated and treated silk biomaterials were examined for interactions with blood components with varying degrees of complexity, including isolated platelets, platelet rich plasma, blood plasma, and whole blood, both under agitated and flow conditions. rDV-biofunctionalized silk biomaterials were shown to be blood compatible in terms of platelet and whole blood interactions and the PIII treatment was shown to be an effective and efficient means of covalently immobilizing rDV in its bioactive form. These biomimetic silk biomaterials are a promising platform toward development of silk-based blood-contacting devices for therapeutic, diagnostic, and research applications. STATEMENT OF SIGNIFICANCE: Blood compatible materials are required for the development of therapeutic and diagnostic blood contacting devices as blood-material interactions are a key factor dictating device functionality. In this work, we explored biofunctionalization of silk biomaterials with a recombinantly expressed domain V (rDV) of the human basement membrane proteoglycan perlecan towards the development of blood compatible surfaces. Perlecan and rDV are of interest in vascular device development as they uniquely support endothelial cell, while inhibiting smooth muscle cell and platelet interactions. rDV was covalently immobilized on silk biomaterials using plasma immersion ion implantation (PIII), a new method of immobilizing proteins on silk biomaterials that does not rely on modification of specific amino acids in the silk protein chain. These biomimetic silk biomaterials are a promising platform toward development of silk-based blood-contacting devices for therapeutic, diagnostic, and research applications.
Collapse
|
6
|
Rebuilding the hematopoietic stem cell niche: Recent developments and future prospects. Acta Biomater 2021; 132:129-148. [PMID: 33813090 DOI: 10.1016/j.actbio.2021.03.061] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 03/25/2021] [Accepted: 03/25/2021] [Indexed: 12/20/2022]
Abstract
Hematopoietic stem cells (HSCs) have proven their clinical relevance in stem cell transplantation to cure patients with hematological disorders. Key to their regenerative potential is their natural microenvironment - their niche - in the bone marrow (BM). Developments in the field of biomaterials enable the recreation of such environments with increasing preciseness in the laboratory. Such artificial niches help to gain a fundamental understanding of the biophysical and biochemical processes underlying the interaction of HSCs with the materials in their environment and the disturbance of this interplay during diseases affecting the BM. Artificial niches also have the potential to multiply HSCs in vitro, to enable the targeted differentiation of HSCs into mature blood cells or to serve as drug-testing platforms. In this review, we will introduce the importance of artificial niches followed by the biology and biophysics of the natural archetype. We will outline how 2D biomaterials can be used to dissect the complexity of the natural niche into individual parameters for fundamental research and how 3D systems evolved from them. We will present commonly used biomaterials for HSC research and their applications. Finally, we will highlight two areas in the field of HSC research, which just started to unlock the possibilities provided by novel biomaterials, in vitro blood production and studying the pathophysiology of the niche in vitro. With these contents, the review aims to give a broad overview of the different biomaterials applied for HSC research and to discuss their potentials, challenges and future directions in the field. STATEMENT OF SIGNIFICANCE: Hematopoietic stem cells (HSCs) are multipotent cells responsible for maintaining the turnover of all blood cells. They are routinely applied to treat patients with hematological diseases. This high clinical relevance explains the necessity of multiplication or differentiation of HSCs in the laboratory, which is hampered by the missing natural microenvironment - the so called niche. Biomaterials offer the possibility to mimic the niche and thus overcome this hurdle. The review introduces the HSC niche in the bone marrow and discusses the utility of biomaterials in creating artificial niches. It outlines how 2D systems evolved into sophisticated 3D platforms, which opened the gateway to applications such as, expansion of clinically relevant HSCs, in vitro blood production, studying niche pathologies and drug testing.
Collapse
|
7
|
Di Buduo CA, Laurent PA, Zaninetti C, Lordier L, Soprano PM, Ntai A, Barozzi S, La Spada A, Biunno I, Raslova H, Bussel JB, Kaplan DL, Balduini CL, Pecci A, Balduini A. Miniaturized 3D bone marrow tissue model to assess response to Thrombopoietin-receptor agonists in patients. eLife 2021; 10:58775. [PMID: 34059198 PMCID: PMC8169123 DOI: 10.7554/elife.58775] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2020] [Accepted: 04/18/2021] [Indexed: 01/09/2023] Open
Abstract
Thrombocytopenic disorders have been treated with the Thrombopoietin-receptor agonist Eltrombopag. Patients with the same apparent form of thrombocytopenia may respond differently to the treatment. We describe a miniaturized bone marrow tissue model that provides a screening bioreactor for personalized, pre-treatment response prediction to Eltrombopag for individual patients. Using silk fibroin, a 3D bone marrow niche was developed that reproduces platelet biogenesis. Hematopoietic progenitors were isolated from a small amount of peripheral blood of patients with mutations in ANKRD26 and MYH9 genes, who had previously received Eltrombopag. The ex vivo response was strongly correlated with the in vivo platelet response. Induced Pluripotent Stem Cells (iPSCs) from one patient with mutated MYH9 differentiated into functional megakaryocytes that responded to Eltrombopag. Combining patient-derived cells and iPSCs with the 3D bone marrow model technology allows having a reproducible system for studying drug mechanisms and for individualized, pre-treatment selection of effective therapy in Inherited Thrombocytopenias. Platelets are tiny cell fragments essential for blood to clot. They are created and released into the bloodstream by megakaryocytes, giant cells that live in the bone marrow. In certain genetic diseases, such as Inherited Thrombocytopenia, the bone marrow fails to produce enough platelets: this leaves patients extremely susceptible to bruising, bleeding, and poor clotting after an injury or surgery. Certain patients with Inherited Thrombocytopenia respond well to treatments designed to boost platelet production, but others do not. Why these differences exist could be investigated by designing new test systems that recreate the form and function of bone marrow in the laboratory. However, it is challenging to build the complex and poorly understood bone marrow environment outside of the body. Here, Di Buduo et al. have developed an artificial three-dimensional miniature organ bioreactor system that recreates the key features of bone marrow. In this system, megakaryocytes were grown from patient blood samples, and hooked up to a tissue scaffold made of silk. The cells were able to grow as if they were in their normal environment, and they could shed platelets into an artificial bloodstream. After treating megakaryocytes with drugs to stimulate platelet production, Di Buduo et al. found that the number of platelets recovered from the bioreactor could accurately predict which patients would respond to these drugs in the clinic. This new test system enables researchers to predict how a patient will respond to treatment, and to tailor therapy options to each individual. This technology could also be used to test new drugs for Inherited Thrombocytopenias and other blood-related diseases; if scaled-up, it could also, one day, generate large quantities of lab-grown blood cells for transfusion.
Collapse
Affiliation(s)
| | | | - Carlo Zaninetti
- Department of Internal Medicine, I.R.C.C.S. San Matteo Foundation and the University of Pavia, Pavia, Italy
| | - Larissa Lordier
- UMR 1170, Institut National de la Santé et de la Recherche Médicale, Univ. Paris-Sud, Université Paris-Saclay, Gustave Roussy Cancer Campus, Equipe Labellisée Ligue Nationale Contre le Cancer, Villejuif, France
| | - Paolo M Soprano
- Department of Molecular Medicine, University of Pavia, Pavia, Italy
| | - Aikaterini Ntai
- Integrated Systems Engineering, Milano, Italy.,Isenet Biobanking, Milano, Italy
| | - Serena Barozzi
- Department of Internal Medicine, I.R.C.C.S. San Matteo Foundation and the University of Pavia, Pavia, Italy
| | - Alberto La Spada
- Integrated Systems Engineering, Milano, Italy.,Isenet Biobanking, Milano, Italy
| | - Ida Biunno
- Isenet Biobanking, Milano, Italy.,Institute for Genetic and Biomedical Research-CNR, Milano, Italy
| | - Hana Raslova
- UMR 1170, Institut National de la Santé et de la Recherche Médicale, Univ. Paris-Sud, Université Paris-Saclay, Gustave Roussy Cancer Campus, Equipe Labellisée Ligue Nationale Contre le Cancer, Villejuif, France
| | - James B Bussel
- Department of Pediatrics, Weill Cornell Medicine, New York, United States
| | - David L Kaplan
- Department of Biomedical Engineering, Tufts University, Medford, United States
| | - Carlo L Balduini
- Department of Internal Medicine, I.R.C.C.S. San Matteo Foundation and the University of Pavia, Pavia, Italy
| | - Alessandro Pecci
- Department of Internal Medicine, I.R.C.C.S. San Matteo Foundation and the University of Pavia, Pavia, Italy
| | - Alessandra Balduini
- Department of Molecular Medicine, University of Pavia, Pavia, Italy.,Department of Biomedical Engineering, Tufts University, Medford, United States
| |
Collapse
|
8
|
Balduini A, Fava C, Di Buduo CA, Abbonante V, Meneguzzi A, Soprano PM, Taus F, Castelli M, Giontella A, Dovizio M, Tacconelli S, Patrignani P, Minuz P. Expression and functional characterization of the large-conductance calcium and voltage-activated potassium channel K ca 1.1 in megakaryocytes and platelets. J Thromb Haemost 2021; 19:1558-1571. [PMID: 33590615 DOI: 10.1111/jth.15269] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 02/11/2021] [Accepted: 02/12/2021] [Indexed: 01/08/2023]
Abstract
BACKGROUND Ion channels are transmembrane proteins that play important roles in cell function regulation modulating ionic cell permeability. In megakaryocytes and platelets, regulated ion flows have been demonstrated to modulate platelet production and function. However, a relatively limited characterization of ion channel expression and function is available in the human megakaryocyte-platelet lineage. OBJECTIVE We analyzed the expression and function of the large-conductance calcium and voltage-activated potassium channel Kca 1.1 (also known as Maxi-K, BK, slo1) in human megakaryocytes and platelets. METHODS To investigate the functionality of Kca 1.1, we exploited different agonists (BMS-191011, NS1619, NS11021, epoxyeicosatrienoic acid isoforms) and inhibitors (iberiotoxin, penitrem A) of the channel. RESULTS In megakaryocytes, Kca 1.1 agonists determined a decreased proplatelet formation and altered interaction with the extracellular matrix. Analysis of the actin cytoskeleton demonstrated a significant decrease in megakaryocyte spreading and adhesion to collagen. In platelets, the opening of the channel Kca 1.1 led to a reduced sensitivity to agonists with blunted aggregation in response to ADP, with an inhibitory capacity additive to that of aspirin. The Kca 1.1 agonists, but not the inhibitors, determined a reduction of platelet adhesion and aggregation onto immobilized collagen underflow to an extent similar to that of aspirin and ticagrelor. The opening of the Kca 1.1 resulted in cell hyperpolarization impairing free intracellular calcium in ADP-stimulated platelets and megakaryocytes. CONCLUSIONS The present study reveals new mechanisms in platelet formation and activation, suggesting that targeting Kca 1.1 channels might be of potential pharmacological interest in hemostasis and thrombosis.
Collapse
Affiliation(s)
- Alessandra Balduini
- Department of Molecular Medicine, University of Pavia, Pavia, Italy
- Laboratory of Biochemistry, Biotechnology and Advanced Diagnosis, Istituto di Ricovero e Cura a Carattere Scientifico (I.R.C.C.S.) Policlinico San Matteo Foundation, Pavia, Italy
- Department of Biomedical Engineering, Tufts University, Medford, MA, USA
| | - Cristiano Fava
- Section of Internal Medicine C, Department of Medicine, University of Verona, Verona, Italy
| | - Christian A Di Buduo
- Department of Molecular Medicine, University of Pavia, Pavia, Italy
- Laboratory of Biochemistry, Biotechnology and Advanced Diagnosis, Istituto di Ricovero e Cura a Carattere Scientifico (I.R.C.C.S.) Policlinico San Matteo Foundation, Pavia, Italy
| | - Vittorio Abbonante
- Department of Molecular Medicine, University of Pavia, Pavia, Italy
- Laboratory of Biochemistry, Biotechnology and Advanced Diagnosis, Istituto di Ricovero e Cura a Carattere Scientifico (I.R.C.C.S.) Policlinico San Matteo Foundation, Pavia, Italy
| | - Alessandra Meneguzzi
- Section of Internal Medicine C, Department of Medicine, University of Verona, Verona, Italy
| | - Paolo M Soprano
- Department of Molecular Medicine, University of Pavia, Pavia, Italy
- Laboratory of Biochemistry, Biotechnology and Advanced Diagnosis, Istituto di Ricovero e Cura a Carattere Scientifico (I.R.C.C.S.) Policlinico San Matteo Foundation, Pavia, Italy
| | - Francesco Taus
- Section of Internal Medicine C, Department of Medicine, University of Verona, Verona, Italy
| | - Marco Castelli
- Section of Internal Medicine C, Department of Medicine, University of Verona, Verona, Italy
| | - Alice Giontella
- Section of Internal Medicine C, Department of Medicine, University of Verona, Verona, Italy
| | - Melania Dovizio
- Department of Neuroscience, Imaging and Clinical Sciences and Center for Advanced Studies and Technology (CAST, School of Medicine, "G. d'Annunzio" University, Chieti, Italy
| | - Stefania Tacconelli
- Department of Neuroscience, Imaging and Clinical Sciences and Center for Advanced Studies and Technology (CAST, School of Medicine, "G. d'Annunzio" University, Chieti, Italy
| | - Paola Patrignani
- Department of Neuroscience, Imaging and Clinical Sciences and Center for Advanced Studies and Technology (CAST, School of Medicine, "G. d'Annunzio" University, Chieti, Italy
| | - Pietro Minuz
- Section of Internal Medicine C, Department of Medicine, University of Verona, Verona, Italy
| |
Collapse
|
9
|
Process analysis of pluripotent stem cell differentiation to megakaryocytes to make platelets applying European GMP. NPJ Regen Med 2021; 6:27. [PMID: 34040001 PMCID: PMC8155004 DOI: 10.1038/s41536-021-00138-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Accepted: 04/20/2021] [Indexed: 12/27/2022] Open
Abstract
Quality, traceability and reproducibility are crucial factors in the reliable manufacture of cellular therapeutics, as part of the overall framework of Good Manufacturing Practice (GMP). As more and more cellular therapeutics progress towards the clinic and research protocols are adapted to comply with GMP standards, guidelines for safe and efficient adaptation have become increasingly relevant. In this paper, we describe the process analysis of megakaryocyte manufacture from induced pluripotent stem cells with a view to manufacturing in vitro platelets to European GMP for transfusion. This process analysis has allowed us an overview of the entire manufacturing process, enabling us to pinpoint the cause and severity of critical risks. Risk mitigations were then proposed for each risk, designed to be GMP compliant. These mitigations will be key in advancing this iPS-derived therapy towards the clinic and have broad applicability to other iPS-derived cellular therapeutics, many of which are currently advancing towards GMP-compliance. Taking these factors into account during protocol design could potentially save time and money, expediting the advent of safe, novel therapeutics from stem cells.
Collapse
|
10
|
Increased B4GALT1 expression is associated with platelet surface galactosylation and thrombopoietin plasma levels in MPNs. Blood 2021; 137:2085-2089. [PMID: 33238000 DOI: 10.1182/blood.2020007265] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Accepted: 11/09/2020] [Indexed: 12/31/2022] Open
Abstract
Aberrant megakaryopoiesis is a hallmark of the myeloproliferative neoplasms (MPNs), a group of clonal hematological malignancies originating from hematopoietic stem cells, leading to an increase in mature blood cells in the peripheral blood. Sialylated derivatives of the glycan structure β4-N-acetyllactosamine (Galβ1,4GlcNAc or type-2 LacNAc, hereafter referred to as LacNAc) regulate platelet life span, hepatic thrombopoietin (TPO) production, and thrombopoiesis. We found increased TPO plasma levels in MPNs with high allele burden of the mutated clones. Remarkably, platelets isolated from MPNs had a significant increase in LacNAc expression that correlated with the high allele burden regardless of the underlying identified mutation. Megakaryocytes derived in vitro from these patients showed an increased expression of the B4GALT1 gene encoding β-1,4-galactosyltransferase 1 (β4GalT1). Consistently, megakaryocytes from MPN showed increased LacNAc expression relative to healthy controls, which was counteracted by the treatment with a Janus kinase 1/2 inhibitor. Altered expression of B4GALT1 in mutant megakaryocytes can lead to the production of platelets with aberrant galactosylation, which in turn promote hepatic TPO synthesis regardless of platelet mass. Our findings provide a new paradigm for understanding aberrant megakaryopoiesis in MPNs and identify β4GalT1 as a potential actionable target for therapy.
Collapse
|
11
|
Di Buduo CA, Aguilar A, Soprano PM, Bocconi A, Miguel CP, Mantica G, Balduini A. Latest culture techniques: cracking the secrets of bone marrow to mass-produce erythrocytes and platelets ex vivo. Haematologica 2021; 106:947-957. [PMID: 33472355 PMCID: PMC8017859 DOI: 10.3324/haematol.2020.262485] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Indexed: 12/13/2022] Open
Abstract
Since the dawn of medicine, scientists have carefully observed, modeled and interpreted the human body to improve healthcare. At the beginning there were drawings and paintings, now there is three-dimensional modeling. Moving from two-dimensional cultures and towards complex and relevant biomaterials, tissue-engineering approaches have been developed in order to create three-dimensional functional mimics of native organs. The bone marrow represents a challenging organ to reproduce because of its structure and composition that confer it unique biochemical and mechanical features to control hematopoiesis. Reproducing the human bone marrow niche is instrumental to answer the growing demand for human erythrocytes and platelets for fundamental studies and clinical applications in transfusion medicine. In this review, we discuss the latest culture techniques and technological approaches to obtain functional platelets and erythrocytes ex vivo. This is a rapidly evolving field that will define the future of targeted therapies for thrombocytopenia and anemia, but also a long-term promise for new approaches to the understanding and cure of hematologic diseases.
Collapse
Affiliation(s)
| | - Alicia Aguilar
- Department of Molecular Medicine, University of Pavia, Pavia
| | - Paolo M Soprano
- Department of Molecular Medicine, University of Pavia, Pavia
| | - Alberto Bocconi
- Department of Molecular Medicine, University of Pavia, Pavia, Italy; Department of Chemistry, Materials and Chemical Engineering G. Natta, Politecnico di Milano, Milano
| | | | | | - Alessandra Balduini
- Department of Molecular Medicine, University of Pavia, Pavia, Italy; Department of Biomedical Engineering, Tufts University, Medford, MA
| |
Collapse
|
12
|
Di Buduo CA, Soprano PM, Miguel CP, Perotti C, Del Fante C, Balduini A. A Gold Standard Protocol for Human Megakaryocyte Culture Based on the Analysis of 1,500 Umbilical Cord Blood Samples. Thromb Haemost 2020; 121:538-542. [PMID: 33160288 DOI: 10.1055/s-0040-1719028] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Affiliation(s)
- Christian A Di Buduo
- Department of Molecular Medicine, University of Pavia, Pavia, Italy.,Laboratory of Biochemistry, Biotechnology and Advanced Diagnosis, Istituto di Ricovero e Cura a Carattere Scientifico (I.R.C.C.S.) Policlinico San Matteo Foundation, Pavia, Italy
| | - Paolo M Soprano
- Department of Molecular Medicine, University of Pavia, Pavia, Italy.,Laboratory of Biochemistry, Biotechnology and Advanced Diagnosis, Istituto di Ricovero e Cura a Carattere Scientifico (I.R.C.C.S.) Policlinico San Matteo Foundation, Pavia, Italy
| | - Carolina P Miguel
- Department of Molecular Medicine, University of Pavia, Pavia, Italy.,Laboratory of Biochemistry, Biotechnology and Advanced Diagnosis, Istituto di Ricovero e Cura a Carattere Scientifico (I.R.C.C.S.) Policlinico San Matteo Foundation, Pavia, Italy
| | - Cesare Perotti
- Immunohematology and Transfusion Service and Cell Therapy Unit, Istituto di Ricovero e Cura a Carattere Scientifico (I.R.C.C.S.) Policlinico San Matteo Foundation, Pavia, Italy
| | - Claudia Del Fante
- Immunohematology and Transfusion Service and Cell Therapy Unit, Istituto di Ricovero e Cura a Carattere Scientifico (I.R.C.C.S.) Policlinico San Matteo Foundation, Pavia, Italy
| | - Alessandra Balduini
- Department of Molecular Medicine, University of Pavia, Pavia, Italy.,Laboratory of Biochemistry, Biotechnology and Advanced Diagnosis, Istituto di Ricovero e Cura a Carattere Scientifico (I.R.C.C.S.) Policlinico San Matteo Foundation, Pavia, Italy.,Department of Biomedical Engineering, Tufts University, Medford, Massachusetts, United States
| |
Collapse
|
13
|
Abbonante V, Di Buduo CA, Malara A, Laurent PA, Balduini A. Mechanisms of platelet release: in vivo studies and in vitro modeling. Platelets 2020; 31:717-723. [PMID: 32522064 DOI: 10.1080/09537104.2020.1774532] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Mechanisms related to platelet release in the context of the bone marrow niche are not completely known. In this review we discuss what has been discovered about four critical aspects of this process: 1) the bone marrow niche organization, 2) the role of the extracellular matrix components, 3) the mechanisms by which megakaryocytes release platelets and 4) the novel approaches to mimic the bone marrow environment and produce platelets ex vivo.
Collapse
Affiliation(s)
| | | | - Alessandro Malara
- Department of Molecular Medicine, University of Pavia , Pavia, Italy
| | | | | |
Collapse
|
14
|
Ren P, Yang C, Lofchy LA, Wang G, Chen F, Simberg D. Establishing In Situ Closed Circuit Perfusion of Lower Abdominal Organs and Hind Limbs in Mice. J Vis Exp 2020. [PMID: 32865531 DOI: 10.3791/60847] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
Ex vivo perfusion is an important physiological tool to study the function of isolated organs (e.g. liver, kidneys). At the same time, due to the small size of mouse organs, ex vivo perfusion of bone, bladder, skin, prostate, and reproductive organs is challenging or not feasible. Here, we report for the first time an in situ lower body perfusion circuit in mice that includes the above tissues, but bypasses the main clearance organs (kidney, liver, and spleen). The circuit is established by cannulating the abdominal aorta and inferior vena cava above the iliac artery and vein and cauterizing peripheral blood vessels. Perfusion is performed via a peristaltic pump with perfusate flow maintained for up to 2 h. In situ staining with fluorescent lectin and Hoechst solution confirmed that the microvasculature was successfully perfused. This mouse model can be a very useful tool for studying pathological processes as well as mechanisms of drug delivery, migration/metastasis of circulating tumor cells into/from the tumor, and interactions of immune system with perfused organs and tissues.
Collapse
Affiliation(s)
- Ping Ren
- Department of Thoracic Surgery, The First Hospital of Jilin University; The Skaggs School of Pharmacy and Pharmaceutical Sciences, Department of Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus; Key Laboratory of Zoonoses Research, Ministry of Education, Jilin University
| | - Chunyan Yang
- The Skaggs School of Pharmacy and Pharmaceutical Sciences, Department of Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus; Department of Cardiology, China-Japan Union Hospital of Jilin University
| | - Laren A Lofchy
- The Skaggs School of Pharmacy and Pharmaceutical Sciences, Department of Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus
| | - Guankui Wang
- The Skaggs School of Pharmacy and Pharmaceutical Sciences, Department of Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus
| | - Fangfang Chen
- The Skaggs School of Pharmacy and Pharmaceutical Sciences, Department of Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus; Department of Cardiology, China-Japan Union Hospital of Jilin University; Department of Gastrointestinal, Colorectal and Anal Surgery, China-Japan Union Hospital of Jilin University;
| | - Dmitri Simberg
- The Skaggs School of Pharmacy and Pharmaceutical Sciences, Department of Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus
| |
Collapse
|
15
|
Kim S, Shah SB, Graney PL, Singh A. Multiscale engineering of immune cells and lymphoid organs. NATURE REVIEWS. MATERIALS 2019; 4:355-378. [PMID: 31903226 PMCID: PMC6941786 DOI: 10.1038/s41578-019-0100-9] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/06/2023]
Abstract
Immunoengineering applies quantitative and materials-based approaches for the investigation of the immune system and for the development of therapeutic solutions for various diseases, such as infection, cancer, inflammatory diseases and age-related malfunctions. The design of immunomodulatory and cell therapies requires the precise understanding of immune cell formation and activation in primary, secondary and ectopic tertiary immune organs. However, the study of the immune system has long been limited to in vivo approaches, which often do not allow multidimensional control of intracellular and extracellular processes, and to 2D in vitro models, which lack physiological relevance. 3D models built with synthetic and natural materials enable the structural and functional recreation of immune tissues. These models are being explored for the investigation of immune function and dysfunction at the cell, tissue and organ levels. In this Review, we discuss 2D and 3D approaches for the engineering of primary, secondary and tertiary immune structures at multiple scales. We highlight important insights gained using these models and examine multiscale engineering strategies for the design and development of immunotherapies. Finally, dynamic 4D materials are investigated for their potential to provide stimuli-dependent and context-dependent scaffolds for the generation of immune organ models.
Collapse
Affiliation(s)
- Sungwoong Kim
- Department of Materials Science and Engineering, Cornell University, Ithaca, NY, USA
- These authors contributed equally: Sungwoong Kim, Shivem B. Shah, Pamela L. Graney
| | - Shivem B. Shah
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
- These authors contributed equally: Sungwoong Kim, Shivem B. Shah, Pamela L. Graney
| | - Pamela L. Graney
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
- Sibley School of Mechanical and Aerospace Engineering, Cornell University, Ithaca, NY, USA
- These authors contributed equally: Sungwoong Kim, Shivem B. Shah, Pamela L. Graney
| | - Ankur Singh
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
- Sibley School of Mechanical and Aerospace Engineering, Cornell University, Ithaca, NY, USA
- Englander Institute for Precision Medicine, Weill Cornell Medical College, New York, NY, USA
| |
Collapse
|
16
|
Yaghoobi M, Hashemi-Najafabadi S, Soleimani M, Vasheghani-Farahani E. Osteogenic induction of human mesenchymal stem cells in multilayered electrospun scaffolds at different flow rates and configurations in a perfusion bioreactor. J Biosci Bioeng 2019; 128:495-503. [PMID: 31085079 DOI: 10.1016/j.jbiosc.2019.03.015] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Revised: 03/05/2019] [Accepted: 03/20/2019] [Indexed: 01/06/2023]
Abstract
Electrospun scaffolds are potentially interesting in bone tissue engineering due to a strong structural similarity to the natural bone matrix. To investigate the osteogenic behavior of cells on the scaffolds, dynamic culture of cells is essential to simulate the biological environment. In the present study, human mesenchymal stem cells (hMSCs) were cultured on multilayer nanohydroxyapatite-polycaprolactone electrospun scaffolds at different configurations (horizontal with or without pressure and parallel with the medium flow) and flow rates in a perfusion bioreactor. Alkaline phosphatase (ALP) activity, cell viability, Ca deposition and RUNX2 expression were determined in three different dynamic states, and compared with static culture after 1, 3, 7, and 14 days. Among dynamic groups, RUNX2 gene expression upregulated more in a horizontal state at a low flow rate without mechanical pressure (LF) and parallel flow (PF), than static group on day 7. At a high flow rate with mechanical pressure, Ca deposition and ALP activity increased 2.34 and 1.7 folds more than in static culture over 7 days, respectively. Furthermore, ALP activity, Ca deposition and RUNX2 gene expression increased in PF samples. PF provided longer culture time with higher cell differentiation. Therefore, high flow rate with mechanical pressure and PF are suggested for producing differentiated cell structure for bone tissue engineering.
Collapse
Affiliation(s)
- Maliheh Yaghoobi
- Engineering Department, Faculty of Chemical Engineering, University of Zanjan, P.O. Box 45371-38791, Zanjan, Iran; Biomedical Engineering Department, Faculty of Chemical Engineering, Tarbiat Modares University, P.O. Box 14115-114, Tehran, Iran
| | - Sameereh Hashemi-Najafabadi
- Biomedical Engineering Department, Faculty of Chemical Engineering, Tarbiat Modares University, P.O. Box 14115-114, Tehran, Iran.
| | - Masoud Soleimani
- Hematology Department, Faculty of Medical Sciences, Tarbiat Modares University, P.O. Box 14115- 331, Tehran, Iran
| | - Ebrahim Vasheghani-Farahani
- Biomedical Engineering Department, Faculty of Chemical Engineering, Tarbiat Modares University, P.O. Box 14115-114, Tehran, Iran
| |
Collapse
|
17
|
Using genome editing to engineer universal platelets. Emerg Top Life Sci 2019; 3:301-311. [PMID: 33523140 PMCID: PMC7289015 DOI: 10.1042/etls20180153] [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: 01/10/2019] [Revised: 03/19/2019] [Accepted: 03/20/2019] [Indexed: 12/29/2022]
Abstract
Genome editing technologies such as zinc finger nucleases, TALENs and CRISPR/Cas9 have recently emerged as tools with the potential to revolutionise cellular therapy. This is particularly exciting for the field of regenerative medicine, where the large-scale, quality-controlled editing of large numbers of cells could generate essential cellular products ready to move towards the clinic. This review details recent progress towards generating HLA Class I null platelets using genome editing technologies for β2-microglobulin deletion, generating a universally transfusable cellular product. In addition, we discuss various methods for megakaryocyte (MK) production from human pluripotent stem cells and subsequent platelet production from the MKs. As well as simply producing platelets, differentiating MK cultures can enable us to understand megakaryopoiesis in vivo and take steps towards ameliorating bleeding disorders or deficiencies in MK maturation in patients. Thus by intersecting both these areas of research, we can produce optimised differentiation systems for the production of universal platelets, thus offering a stable supply of platelets for difficult-to-match patients and providing areas with transmissible disease concerns or an unpredictable supply of platelets with a steady supply of quality-controlled platelet units.
Collapse
|
18
|
Megakaryocytes from fat: a new recipe for platelets. Blood 2019; 133:623-625. [PMID: 30765492 DOI: 10.1182/blood-2018-12-889766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|
19
|
Chramiec A, Vunjak-Novakovic G. Tissue engineered models of healthy and malignant human bone marrow. Adv Drug Deliv Rev 2019; 140:78-92. [PMID: 31002835 PMCID: PMC6663611 DOI: 10.1016/j.addr.2019.04.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2018] [Revised: 02/14/2019] [Accepted: 04/14/2019] [Indexed: 12/20/2022]
Abstract
Tissue engineering is becoming increasingly successful in providing in vitro models of human tissues that can be used for ex vivo recapitulation of functional tissues as well as predictive testing of drug efficacy and safety. From simple tissue models to microphysiological platforms comprising multiple tissue types connected by vascular perfusion, these "tissues on a chip" are emerging as a fast track application for tissue engineering, with great potential for modeling diseases and supporting the development of new drugs and therapeutic targets. We focus here on tissue engineering of the hematopoietic stem and progenitor cell compartment and the malignancies that can develop in the human bone marrow. Our overall goal is to demonstrate the utility and interconnectedness of improvements in bioengineering methods developed in one area of bone marrow studies for the remaining, seemingly disparate, bone marrow fields.
Collapse
|
20
|
Shepherd JH, Howard D, Waller AK, Foster HR, Mueller A, Moreau T, Evans AL, Arumugam M, Bouët Chalon G, Vriend E, Davidenko N, Ghevaert C, Best SM, Cameron RE. Structurally graduated collagen scaffolds applied to the ex vivo generation of platelets from human pluripotent stem cell-derived megakaryocytes: Enhancing production and purity. Biomaterials 2018; 182:135-144. [PMID: 30118981 DOI: 10.1016/j.biomaterials.2018.08.019] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Revised: 08/03/2018] [Accepted: 08/06/2018] [Indexed: 01/05/2023]
Abstract
Platelet transfusions are a key treatment option for a range of life threatening conditions including cancer, chemotherapy and surgery. Efficient ex vivo systems to generate donor independent platelets in clinically relevant numbers could provide a useful substitute. Large quantities of megakaryocytes (MKs) can be produced from human pluripotent stem cells, but in 2D culture the ratio of platelets harvested from MK cells has been limited and restricts production rate. The development of biomaterial cell supports that replicate vital hematopoietic micro-environment cues are one strategy that may increase in vitro platelet production rates from iPS derived Megakaryocyte cells. In this paper, we present the results obtained generating, simulating and using a novel structurally-graded collagen scaffold within a flow bioreactor system seeded with programmed stem cells. Theoretical analysis of porosity using micro-computed tomography analysis and synthetic micro-particle filtration provided a predictive tool to tailor cell distribution throughout the material. When used with MK programmed stem cells the graded scaffolds influenced cell location while maintaining the ability to continuously release metabolically active CD41 + CD42 + functional platelets. This scaffold design and novel fabrication technique offers a significant advance in understanding the influence of scaffold architectures on cell seeding, retention and platelet production.
Collapse
Affiliation(s)
- Jennifer H Shepherd
- Cambridge Centre for Medical Materials, Department of Materials Science and Metallurgy, 27 Charles Babbage Road, Cambridge CB3 0FS, UK.
| | - Daniel Howard
- Department of Haematology, University of Cambridge, National Health Blood Service Centre, Long Road, Cambridge CB2 0PT, UK
| | - Amie K Waller
- Department of Haematology, University of Cambridge, National Health Blood Service Centre, Long Road, Cambridge CB2 0PT, UK
| | - Holly Rebecca Foster
- Department of Haematology, University of Cambridge, National Health Blood Service Centre, Long Road, Cambridge CB2 0PT, UK
| | - Annett Mueller
- Department of Haematology, University of Cambridge, National Health Blood Service Centre, Long Road, Cambridge CB2 0PT, UK
| | - Thomas Moreau
- Department of Haematology, University of Cambridge, National Health Blood Service Centre, Long Road, Cambridge CB2 0PT, UK
| | - Amanda L Evans
- Department of Haematology, University of Cambridge, National Health Blood Service Centre, Long Road, Cambridge CB2 0PT, UK
| | - Meera Arumugam
- Department of Haematology, University of Cambridge, National Health Blood Service Centre, Long Road, Cambridge CB2 0PT, UK
| | - Guénaëlle Bouët Chalon
- Department of Haematology, University of Cambridge, National Health Blood Service Centre, Long Road, Cambridge CB2 0PT, UK
| | - Eleonora Vriend
- Cambridge Centre for Medical Materials, Department of Materials Science and Metallurgy, 27 Charles Babbage Road, Cambridge CB3 0FS, UK
| | - Natalia Davidenko
- Cambridge Centre for Medical Materials, Department of Materials Science and Metallurgy, 27 Charles Babbage Road, Cambridge CB3 0FS, UK
| | - Cedric Ghevaert
- Department of Haematology, University of Cambridge, National Health Blood Service Centre, Long Road, Cambridge CB2 0PT, UK.
| | - Serena M Best
- Cambridge Centre for Medical Materials, Department of Materials Science and Metallurgy, 27 Charles Babbage Road, Cambridge CB3 0FS, UK
| | - Ruth E Cameron
- Cambridge Centre for Medical Materials, Department of Materials Science and Metallurgy, 27 Charles Babbage Road, Cambridge CB3 0FS, UK
| |
Collapse
|
21
|
Vijey P, Posorske B, Machlus KR. In vitro culture of murine megakaryocytes from fetal liver-derived hematopoietic stem cells. Platelets 2018; 29:583-588. [PMID: 30047825 DOI: 10.1080/09537104.2018.1492107] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Megakaryocytes (MKs) are specialized precursor cells committed to producing and proliferating platelets. In a cytoskeletal-driven process, mature MKs generate platelets by releasing thin cytoplasmic extensions, named proplatelets, into the sinusoids. Due to knowledge gaps in this process and mounting clinical demand for non-donor-based platelet sources, investigators are successfully developing artificial culture systems to recreate the environment of platelet biogenesis. Nevertheless, drawbacks in current methods entail elaborate procedures for stem cell enrichment, extensive growth periods, low MK yield, and poor proplatelet production. We propose a simple, robust method of primary MK culture that utilizes fetal livers from pregnant mice. Our technique reduces expansion time to 4 days, and generates ~15,000-20,000 MKs per liver. Approximately, 20-50% of these MKs produce structurally dense, high-quality proplatelets. In this review, we outline our method of MK culture and isolation.
Collapse
Affiliation(s)
- Prakrith Vijey
- a Division of Hematology , Brigham and Women's Hospital , Boston , MA , USA
| | - Benjamin Posorske
- a Division of Hematology , Brigham and Women's Hospital , Boston , MA , USA
| | - Kellie R Machlus
- a Division of Hematology , Brigham and Women's Hospital , Boston , MA , USA.,b Department of Medicine , Harvard Medical School , Boston , MA , USA
| |
Collapse
|
22
|
Multi-channel silk sponge mimicking bone marrow vascular niche for platelet production. Biomaterials 2018; 178:122-133. [PMID: 29920404 DOI: 10.1016/j.biomaterials.2018.06.018] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Accepted: 06/12/2018] [Indexed: 01/09/2023]
Abstract
In the bone marrow, the interaction of progenitor cells with the vasculature is fundamental for the release of blood cells into circulation. Silk fibroin, derived from Bombyx mori silkworm cocoons, is a promising protein biomaterial for bone marrow tissue engineering, because of its tunable architecture and mechanical properties, the capacity to incorporate labile compounds without loss of bioactivity and the demonstrated ability to support blood cell formation without premature activation. In this study, we fabricated a custom perfusion chamber to contain a multi-channel lyophilized silk sponge mimicking the vascular network in the bone marrow niche. The perfusion system consisted in an inlet and an outlet and 2 splitters that allowed funneling flow in each single channel of the silk sponge. Computational Fluid Dynamic analysis demonstrated that this design permitted confined flow inside the vascular channels. The silk channeled sponge supported efficient platelet release from megakaryocytes (Mks). After seeding, the Mks localized along SDF-1α functionalized vascular channels in the sponge. Perfusion of the channels allowed the recovery of functional platelets as demonstrated by increased PAC-1 binding upon thrombin stimulation. Further, increasing the number of channels in the silk sponge resulted in a proportional increase in the numbers of platelets recovered, suggesting applicability to scale-up for platelet production. In conclusion, we have developed a scalable system consisting of a multi-channeled silk sponge incorporated in a perfusion chamber that can provide useful technology for functional platelet production ex vivo.
Collapse
|
23
|
Balduini A, Raslova H, Di Buduo CA, Donada A, Ballmaier M, Germeshausen M, Balduini CL. Clinic, pathogenic mechanisms and drug testing of two inherited thrombocytopenias, ANKRD26-related Thrombocytopenia and MYH9-related diseases. Eur J Med Genet 2018; 61:715-722. [PMID: 29545013 DOI: 10.1016/j.ejmg.2018.01.014] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Revised: 01/08/2018] [Accepted: 01/27/2018] [Indexed: 12/21/2022]
Abstract
Inherited thrombocytopenias (ITs) are a heterogeneous group of disorders characterized by low platelet count resulting in impaired hemostasis. Patients can have spontaneous hemorrhages and/or excessive bleedings provoked by hemostatic challenges as trauma or surgery. To date, ITs encompass 32 different rare monogenic disorders caused by mutations of 30 genes. This review will focus on the major discoveries that have been made in the last years on the diagnosis, treatment and molecular mechanisms of ANKRD26-Related Thrombocytopenia and MYH9-Related Diseases. Furthermore, we will discuss the use a Thrombopoietin mimetic as a novel approach to treat the thrombocytopenia in these patients. We will propose the use of a new 3D bone marrow model to study the mechanisms of action of these drugs and to test their efficacy and safety in patients. The overall purpose of this review is to point out that important progresses have been made in understanding the pathogenesis of ANKRD26-Related Thrombocytopenia and MYH9-Related Diseases and new therapeutic approaches have been proposed and tested. Future advancement in this research will rely in the development of more physiological models to study the regulation of human platelet biogenesis, disease mechanisms and specific pharmacologic targets.
Collapse
Affiliation(s)
- Alessandra Balduini
- University of Pavia, Pavia, Italy; IRCCS Policlinico San Matteo Foundation, Pavia, Italy.
| | - Hana Raslova
- INSERM UMR 1170, Gustave Roussy Cancer Campus, Université Paris-Saclay, Equipe Labellisée par la Ligue Nationale Contre le Cancer, Villejuif, France
| | - Christian A Di Buduo
- University of Pavia, Pavia, Italy; IRCCS Policlinico San Matteo Foundation, Pavia, Italy
| | - Alessandro Donada
- INSERM UMR 1170, Gustave Roussy Cancer Campus, Université Paris-Saclay, Equipe Labellisée par la Ligue Nationale Contre le Cancer, Villejuif, France
| | | | | | - Carlo L Balduini
- University of Pavia, Pavia, Italy; IRCCS Policlinico San Matteo Foundation, Pavia, Italy.
| |
Collapse
|
24
|
Di Buduo CA, Abbonante V, Tozzi L, Kaplan DL, Balduini A. Three-Dimensional Tissue Models for Studying Ex Vivo Megakaryocytopoiesis and Platelet Production. Methods Mol Biol 2018; 1812:177-193. [PMID: 30171579 DOI: 10.1007/978-1-4939-8585-2_11] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Three-dimensional (3D) tissue cultures in vitro enable a more physiological reconstruction of native tissues and organs. The bone marrow environment, structure and composition regulate megakaryocyte function and platelet production. Here, we describe the use of silk fibroin protein biomaterials to assemble 3D scaffolds mimicking the bone marrow niche architecture and extracellular matrix composition to support platelet release from human megakaryocytes. Additionally, we also propose the use of hyaluronan hydrogels, functionalized with extracellular matrix components, to reproduce the 3D matrix structure of the bone marrow environment for studying human megakaryocyte function.
Collapse
Affiliation(s)
| | | | - Lorenzo Tozzi
- Department of Biomedical Engineering, Tufts University, Medford, MA, USA
| | - David L Kaplan
- Department of Biomedical Engineering, Tufts University, Medford, MA, USA
| | - Alessandra Balduini
- Department of Molecular Medicine, University of Pavia, Pavia, Italy.
- Department of Biomedical Engineering, Tufts University, Medford, MA, USA.
| |
Collapse
|