1
|
Ding J, Cheng Z, Ma Y, Zhang T, Du L, Jiang X, Zhu M, Li W, Xu B. Engineering Injectable and Highly Interconnected Porous Silk Fibroin Microspheres for Tissue Regeneration. Adv Healthc Mater 2024:e2402932. [PMID: 39498746 DOI: 10.1002/adhm.202402932] [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: 08/07/2024] [Revised: 10/08/2024] [Indexed: 11/07/2024]
Abstract
Injectable porous microspheres represent a promising therapeutic platform for cell delivery, drug delivery, and tissue regeneration. Yet, the engineering of silk fibroin microspheres with a highly interconnected porous structure remains an unsolved challenge. In this study, a simple and efficient method is developed that does not require the use of organic solvents to prepare silk fibroin microspheres with a predictable structure. Through extensive screening, the addition of glucose is found to direct the formation of a highly interconnected porous structure from the interior to the surface of silk fibroin microspheres. Compared to silk fibroin microspheres (SF microspheres) produced through a combination of electro-spray, cryopreservation, and freeze drying, silk fibroin-glucose microspheres (SF-Glu microspheres) demonstrates enhanced capabilities in promoting cell adhesion and proliferation in vitro. Both SF-Glu and SF microspheres exhibit the capacity to maintain the sustained release kinetics of the loaded model drug. Furthermore, SF-Glu microspheres facilitate the recruitment of endogenous cells, capillary migration, and macrophage phenotype switch following subcutaneous injection in the rats. This study opens a new avenue for the construction of porous silk fibroin microspheres, which could lead to a broader range of applications in regenerative medicine.
Collapse
Affiliation(s)
- Ji Ding
- College of Life Sciences, Key Laboratory of Bioactive Materials (Ministry of Education), Nankai University, Tianjin, 300071, China
- Department of Minimally Invasive Spine Surgery, Tianjin Hospital, Tianjin University, Tianjin, 300211, China
| | - Zhaojun Cheng
- Department of Spinal Surgery, The First Affiliated Hospital of Guangzhou University of Chinese Medicine and Guangdong Clinical Research Academy of Chinese Medicine Guangzhou, Guangzhou, Guangdong Province, 510260, China
- Postdoctoral Research Station of Guangzhou University of Chinese Medicine, Guangzhou, 510260, China
| | - Yulong Ma
- Department of Stomatology, Chifeng Municipal Hospital, Chifeng, Inner Mongolia, 024000, China
| | - Tongxing Zhang
- Department of Minimally Invasive Spine Surgery, Tianjin Hospital, Tianjin University, Tianjin, 300211, China
| | - Lilong Du
- Department of Minimally Invasive Spine Surgery, Tianjin Hospital, Tianjin University, Tianjin, 300211, China
| | - Xiaobing Jiang
- The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong Province, 510260, China
| | - Meifeng Zhu
- College of Life Sciences, Key Laboratory of Bioactive Materials (Ministry of Education), Nankai University, Tianjin, 300071, China
| | - Wen Li
- School of Disaster and Emergency Medicine, Tianjin University, Tianjin, 300072, China
| | - Baoshan Xu
- Department of Minimally Invasive Spine Surgery, Tianjin Hospital, Tianjin University, Tianjin, 300211, China
| |
Collapse
|
2
|
Hassan MA, Basha AA, Eraky M, Abbas E, El-Samad LM. Advancements in silk fibroin and silk sericin-based biomaterial applications for cancer therapy and wound dressing formulation: A comprehensive review. Int J Pharm 2024; 662:124494. [PMID: 39038721 DOI: 10.1016/j.ijpharm.2024.124494] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 06/25/2024] [Accepted: 07/18/2024] [Indexed: 07/24/2024]
Abstract
Silks are a class of proteins generated naturally by different arthropods, including silkworms, spiders, scorpions, mites, wasps, and bees. This review discusses the silk fibroin and silk sericin fabricated by Bombyx mori silkworm as versatile fibers. This silk fiber is predominantly composed of hydrophobic silk fibroin and hydrophilic silk sericin. Fibroin is defined as a structural protein that bestows silk with strength, while sericin is characterized as a gum-like protein, tying the two fibrous proteins together and endowing silk proteins with elasticity. Due to their versatile structures, biocompatibility, and biodegradability, they could be tailored into intricate structures to warrant particular demands. The intrinsic functional groups of both proteins enable their functionalization and cross-linking with various biomaterials to endow the matrix with favorable antioxidant and antibacterial properties. Depending on the target applications, they can be integrated with other materials to formulate nanofibrous, hydrogels, films, and micro-nanoparticles. Given the outstanding biological and controllable physicochemical features of fibroin and sericin, they could be exploited in pharmaceutical applications involving tissue engineering, wound repair, drug delivery, and cancer therapy. This review comprehensively discusses the advancements in the implementation of different formulations of silk fibroin and sericin in wound healing and drug delivery systems, particularly for cancer treatment.
Collapse
Affiliation(s)
- Mohamed A Hassan
- Protein Research Department, Genetic Engineering and Biotechnology Research Institute (GEBRI), City of Scientific Research and Technological Applications (SRTA-City), New Borg El-Arab City, P.O. Box: 21934 Alexandria, Egypt; University Medical Center Göttingen, Georg-August-University, 37073 Göttingen, Germany.
| | - Amal A Basha
- Zoology Department, Faculty of Science, Damanhour University, Egypt
| | - Mohamed Eraky
- College of Engineering, Huazhong Agricultural University, 430070 Wuhan, China
| | - Eman Abbas
- Zoology Department, Faculty of Science, Alexandria University, Egypt
| | - Lamia M El-Samad
- Zoology Department, Faculty of Science, Alexandria University, Egypt
| |
Collapse
|
3
|
Liu J, Zhang B, Cui Y, Song H, Shang D. In vitro co-culture models for studying organoids-macrophages interaction: the golden technology of cancer immunotherapy. Am J Cancer Res 2024; 14:3222-3240. [PMID: 39113861 PMCID: PMC11301299 DOI: 10.62347/bqfh7352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Accepted: 06/12/2024] [Indexed: 08/10/2024] Open
Abstract
Macrophages, as the largest immune cell group in tumour tissues, play a crucial role in influencing various malignant behaviours of tumour cells and tumour immune evasion. As the research on macrophages and cancer immunotherapy develops, the importance of appropriate research models becomes increasingly evident. The development of organoids has bridged the gap between traditional two-dimensional (2D) cultures and animal experiments. Recent studies have demonstrated that organoids exhibit similar physiological characteristics to the source tissue and closely resemble the in vivo genome and molecular markers of the source tissue or organ. However, organoids still lack an immune component. Developing a co-culture model of organoids and macrophages is crucial for studying the interaction and mechanisms between tumour cells and macrophages. This paper presents an overview of the establishment of co-culture models, the current research status of organoid macrophage interactions, and the current status of immunotherapy. In addition, the application prospects and shortcomings of the model are explained. Ultimately, it is hoped that the co-culture model will offer a preclinical testing platform for maximising a precise cancer immunotherapy strategy.
Collapse
Affiliation(s)
- Jinming Liu
- Department of General Surgery, Clinical Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical UniversityDalian, Liaoning, PR China
| | - Biao Zhang
- Department of General Surgery, Clinical Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical UniversityDalian, Liaoning, PR China
| | - Yuying Cui
- Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical UniversityDalian, Liaoning, PR China
| | - Huiyi Song
- Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical UniversityDalian, Liaoning, PR China
| | - Dong Shang
- Department of General Surgery, Clinical Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical UniversityDalian, Liaoning, PR China
- Institute (College) of Integrative Medicine, Dalian Medical UniversityDalian, Liaoning, PR China
| |
Collapse
|
4
|
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
|
5
|
Kozan NG, Caswell S, Patel M, Grasman JM. Aligned Collagen Sponges with Tunable Pore Size for Skeletal Muscle Tissue Regeneration. J Funct Biomater 2023; 14:533. [PMID: 37998102 PMCID: PMC10672557 DOI: 10.3390/jfb14110533] [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: 09/19/2023] [Revised: 10/20/2023] [Accepted: 10/22/2023] [Indexed: 11/25/2023] Open
Abstract
Volumetric muscle loss (VML) is a traumatic injury where at least 20% of the mass of a skeletal muscle has been destroyed and functionality is lost. The standard treatment for VML, autologous tissue transfer, is limited as approximately 1 in 10 grafts fail because of necrosis or infection. Tissue engineering strategies seek to develop scaffolds that can regenerate injured muscles and restore functionality. Many of these scaffolds, however, are limited in their ability to restore muscle functionality because of an inability to promote the alignment of regenerating myofibers. For aligned myofibers to form on a scaffold, myoblasts infiltrate the scaffold and receive topographical cues to direct targeted myofiber growth. We seek to determine the optimal pore size for myoblast infiltration and differentiation. We developed a method of tuning the pore size within collagen scaffolds while inducing longitudinal alignment of these pores. Significantly different pore sizes were generated by adjusting the freezing rate of the scaffolds. Scaffolds frozen at -20 °C contained the largest pores. These scaffolds promoted the greatest level of cell infiltration and orientation in the direction of pore alignment. Further research will be conducted to induce higher levels of myofiber formation, to ultimately create an off-the-shelf treatment for VML injuries.
Collapse
Affiliation(s)
| | | | | | - Jonathan M. Grasman
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ 07102, USA
| |
Collapse
|
6
|
Mu X, Amouzandeh R, Vogts H, Luallen E, Arzani M. A brief review on the mechanisms and approaches of silk spinning-inspired biofabrication. Front Bioeng Biotechnol 2023; 11:1252499. [PMID: 37744248 PMCID: PMC10512026 DOI: 10.3389/fbioe.2023.1252499] [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: 07/03/2023] [Accepted: 08/22/2023] [Indexed: 09/26/2023] Open
Abstract
Silk spinning, observed in spiders and insects, exhibits a remarkable biological source of inspiration for advanced polymer fabrications. Because of the systems design, silk spinning represents a holistic and circular approach to sustainable polymer fabrication, characterized by renewable resources, ambient and aqueous processing conditions, and fully recyclable "wastes." Also, silk spinning results in structures that are characterized by the combination of monolithic proteinaceous composition and mechanical strength, as well as demonstrate tunable degradation profiles and minimal immunogenicity, thus making it a viable alternative to most synthetic polymers for the development of advanced biomedical devices. However, the fundamental mechanisms of silk spinning remain incompletely understood, thus impeding the efforts to harness the advantageous properties of silk spinning. Here, we present a concise and timely review of several essential features of silk spinning, including the molecular designs of silk proteins and the solvent cues along the spinning apparatus. The solvent cues, including salt ions, pH, and water content, are suggested to direct the hierarchical assembly of silk proteins and thus play a central role in silk spinning. We also discuss several hypotheses on the roles of solvent cues to provide a relatively comprehensive analysis and to identify the current knowledge gap. We then review the state-of-the-art bioinspired fabrications with silk proteins, including fiber spinning and additive approaches/three-dimensional (3D) printing. An emphasis throughout the article is placed on the universal characteristics of silk spinning developed through millions of years of individual evolution pathways in spiders and silkworms. This review serves as a stepping stone for future research endeavors, facilitating the in vitro recapitulation of silk spinning and advancing the field of bioinspired polymer fabrication.
Collapse
Affiliation(s)
- Xuan Mu
- Roy J. Carver Department of Biomedical Engineering, College of Engineering, University of Iowa, Iowa City, IA, United States
| | | | | | | | | |
Collapse
|
7
|
Adelfio M, Bonzanni M, Callen GE, Paster BJ, Hasturk H, Ghezzi CE. A physiologically relevant culture platform for long-term studies of in vitro gingival tissue. Acta Biomater 2023; 167:321-334. [PMID: 37331612 PMCID: PMC10528240 DOI: 10.1016/j.actbio.2023.06.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 06/06/2023] [Accepted: 06/08/2023] [Indexed: 06/20/2023]
Abstract
There is a clinical need to understand the etiologies of periodontitis, considering the growing socio-economic impact of the disease. Despite recent advances in oral tissue engineering, experimental approaches have failed to develop a physiologically relevant gingival model that combines tissue organization with salivary flow dynamics and stimulation of the shedding and non-shedding oral surfaces. Herein, we develop a dynamic gingival tissue model composed of a silk scaffold, replicating the cyto-architecture and oxygen profile of the human gingiva, along with a saliva-mimicking medium that reflected the ionic composition, viscosity, and non-Newtonian behavior of human saliva. The construct was cultured in a custom designed bioreactor, in which force profiles on the gingival epithelium were modulated through analysis of inlet position, velocity and vorticity to replicate the physiological shear stress of salivary flow. The gingival bioreactor supported the long-term in vivo features of the gingiva and improved the integrity of the epithelial barrier, critical against the invasion of pathogenic bacteria. Furthermore, the challenge of the gingival tissue with P. gingivalis lipopolysaccharide, as an in vitro surrogate for microbial interactions, indicated a greater stability of the dynamic model in maintaining tissue homeostasis and, thus, its applicability in long-term studies. The model will be integrated into future studies with the human subgingival microbiome to investigate host-pathogen and host-commensal interactions. STATEMENT OF SIGNIFICANCE: The major societal impact of human microbiome had reverberated up to the establishment of the Common Fund's Human Microbiome Project, that has the intent of studying the role of microbial communities in human health and diseases, including periodontitis, atopic dermatitis, or asthma and inflammatory bowel disease. In addition, these chronic diseases are emergent drivers of global socioeconomic status. Not only common oral diseases have been shown to be directly correlated with several systemic conditions, but they are differentially impacting some racial/ethnic and socioeconomic groups. To address this growing social disparity, the development of in vitro gingival model would provide a time and cost-effective experimental platform, able to mimic the spectrum of periodontal disease presentation, for the identification of predictive biomarkers for early-stage diagnosis.
Collapse
Affiliation(s)
- M Adelfio
- Department of Biomedical Engineering, University of Massachusetts Lowell, 1 University Avenue, Lowell, MA 01854, USA
| | - M Bonzanni
- Department of Neuroscience, School of Medicine, Tufts University, 136 Harrison Avenue, Boston, MA 02111, USA
| | - G E Callen
- Department of Biomedical Engineering, University of Massachusetts Lowell, 1 University Avenue, Lowell, MA 01854, USA
| | - B J Paster
- The Forsyth Institute, 245 First St, Cambridge, MA 02142, USA
| | - H Hasturk
- The Forsyth Institute, 245 First St, Cambridge, MA 02142, USA
| | - C E Ghezzi
- Department of Biomedical Engineering, University of Massachusetts Lowell, 1 University Avenue, Lowell, MA 01854, USA.
| |
Collapse
|
8
|
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
|
9
|
Mu X, Gerhard-Herman MD, Zhang YS. Building Blood Vessel Chips with Enhanced Physiological Relevance. ADVANCED MATERIALS TECHNOLOGIES 2023; 8:2201778. [PMID: 37693798 PMCID: PMC10489284 DOI: 10.1002/admt.202201778] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Indexed: 09/12/2023]
Abstract
Blood vessel chips are bioengineered microdevices, consisting of biomaterials, human cells, and microstructures, which recapitulate essential vascular structure and physiology and allow a well-controlled microenvironment and spatial-temporal readouts. Blood vessel chips afford promising opportunities to understand molecular and cellular mechanisms underlying a range of vascular diseases. The physiological relevance is key to these blood vessel chips that rely on bioinspired strategies and bioengineering approaches to translate vascular physiology into artificial units. Here, we discuss several critical aspects of vascular physiology, including morphology, material composition, mechanical properties, flow dynamics, and mass transport, which provide essential guidelines and a valuable source of bioinspiration for the rational design of blood vessel chips. We also review state-of-art blood vessel chips that exhibit important physiological features of the vessel and reveal crucial insights into the biological processes and disease pathogenesis, including rare diseases, with notable implications for drug screening and clinical trials. We envision that the advances in biomaterials, biofabrication, and stem cells improve the physiological relevance of blood vessel chips, which, along with the close collaborations between clinicians and bioengineers, enable their widespread utility.
Collapse
Affiliation(s)
- Xuan Mu
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA 02139, USA; Roy J. Carver Department of Biomedical Engineering, College of Engineering, University of Iowa, Iowa City, IA 52242, USA
| | - Marie Denise Gerhard-Herman
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Yu Shrike Zhang
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA 02139, USA
| |
Collapse
|
10
|
Liu J, Xie X, Wang T, Chen H, Fu Y, Cheng X, Wu J, Li G, Liu C, Liimatainen H, Zheng Z, Wang X, Kaplan DL. Promotion of Wound Healing Using Nanoporous Silk Fibroin Sponges. ACS APPLIED MATERIALS & INTERFACES 2023; 15:12696-12707. [PMID: 36855948 DOI: 10.1021/acsami.2c20274] [Citation(s) in RCA: 22] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Wound dressings are important for wound repair. The morphology of the biomaterials used in these dressings, and in particular, the pore structure affects tissue regeneration by facilitating attachment and proliferation of cells due to the hierarchical multiscale, water absorbance, and nutrient transport. In the present study, silk fibroin (SF) sponges with walls containing nanopores (SFNS) were prepared from SF nanoparticles generated during the autoclaving of SF solutions, followed by leaching the SF nanoparticles from the freeze-dried sponges of SF. The nano/microporous structure, biofluid absorbance, and porosity of the SF sponges with and without nanopores were characterized. In vitro cell proliferation, in vivo biocompatibility, and wound healing were evaluated with the sponges. The results demonstrated that SFNS had significantly increased porosity and water permeability, as well as cell attachment and proliferation when compared with SF sponges without the nanopores (SFS). Wound dressings were assessed in a rat skin wound model, and SFNS was superior to SFS in accelerating wound healing, supported by vascularization, deposition of collagen, and increased epidermal thickness over 21 days. Hence, such a dressing material with a hierarchical multiscale pore structure could promote cell migration, vascularization, and tissue regeneration independently without adding any growth factor, which would offer a new strategy to design and engineer better-performed wound dressing.
Collapse
Affiliation(s)
- Jian Liu
- National Engineering Laboratory for Modern Silk, College of Textile and Clothing Engineering, Soochow University, Suzhou 215123, China
- China National Textile and Apparel Council Key Laboratory for Silk Functional Materials and Technology, Soochow University, Suzhou 215123, China
| | - Xusheng Xie
- National Engineering Laboratory for Modern Silk, College of Textile and Clothing Engineering, Soochow University, Suzhou 215123, China
| | - Tao Wang
- National Engineering Laboratory for Modern Silk, College of Textile and Clothing Engineering, Soochow University, Suzhou 215123, China
| | - Hao Chen
- National Engineering Laboratory for Modern Silk, College of Textile and Clothing Engineering, Soochow University, Suzhou 215123, China
| | - Yuhang Fu
- National Engineering Laboratory for Modern Silk, College of Textile and Clothing Engineering, Soochow University, Suzhou 215123, China
| | - Xinyu Cheng
- National Engineering Laboratory for Modern Silk, College of Textile and Clothing Engineering, Soochow University, Suzhou 215123, China
| | - Jianbing Wu
- College of Textile, Garment and Design, Changshu Institute of Technology, Suzhou 215500, China
| | - Gang Li
- National Engineering Laboratory for Modern Silk, College of Textile and Clothing Engineering, Soochow University, Suzhou 215123, China
| | - Chenming Liu
- Fiber and Particle Engineering Research Unit, University of Oulu, Oulu 90014, Finland
| | - Henrikki Liimatainen
- Fiber and Particle Engineering Research Unit, University of Oulu, Oulu 90014, Finland
| | - Zhaozhu Zheng
- National Engineering Laboratory for Modern Silk, College of Textile and Clothing Engineering, Soochow University, Suzhou 215123, China
- Simatech Incorporation, Suzhou 215123, China
| | - Xiaoqin Wang
- National Engineering Laboratory for Modern Silk, College of Textile and Clothing Engineering, Soochow University, Suzhou 215123, China
| | - David L Kaplan
- Department of Biomedical Engineering, Tufts University, 4 Colby Street, Medford, Massachusetts 02155, United States
| |
Collapse
|
11
|
Khan AO, Rodriguez-Romera A, Reyat JS, Olijnik AA, Colombo M, Wang G, Wen WX, Sousos N, Murphy LC, Grygielska B, Perrella G, Mahony CB, Ling RE, Elliott NE, Karali CS, Stone AP, Kemble S, Cutler EA, Fielding AK, Croft AP, Bassett D, Poologasundarampillai G, Roy A, Gooding S, Rayes J, Machlus KR, Psaila B. Human Bone Marrow Organoids for Disease Modeling, Discovery, and Validation of Therapeutic Targets in Hematologic Malignancies. Cancer Discov 2023; 13:364-385. [PMID: 36351055 PMCID: PMC9900323 DOI: 10.1158/2159-8290.cd-22-0199] [Citation(s) in RCA: 36] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 10/04/2022] [Accepted: 11/07/2022] [Indexed: 11/11/2022]
Abstract
A lack of models that recapitulate the complexity of human bone marrow has hampered mechanistic studies of normal and malignant hematopoiesis and the validation of novel therapies. Here, we describe a step-wise, directed-differentiation protocol in which organoids are generated from induced pluripotent stem cells committed to mesenchymal, endothelial, and hematopoietic lineages. These 3D structures capture key features of human bone marrow-stroma, lumen-forming sinusoids, and myeloid cells including proplatelet-forming megakaryocytes. The organoids supported the engraftment and survival of cells from patients with blood malignancies, including cancer types notoriously difficult to maintain ex vivo. Fibrosis of the organoid occurred following TGFβ stimulation and engraftment with myelofibrosis but not healthy donor-derived cells, validating this platform as a powerful tool for studies of malignant cells and their interactions within a human bone marrow-like milieu. This enabling technology is likely to accelerate the discovery and prioritization of novel targets for bone marrow disorders and blood cancers. SIGNIFICANCE We present a human bone marrow organoid that supports the growth of primary cells from patients with myeloid and lymphoid blood cancers. This model allows for mechanistic studies of blood cancers in the context of their microenvironment and provides a much-needed ex vivo tool for the prioritization of new therapeutics. See related commentary by Derecka and Crispino, p. 263. This article is highlighted in the In This Issue feature, p. 247.
Collapse
Affiliation(s)
- Abdullah O. Khan
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Vincent Drive, Birmingham, United Kingdom
- MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine and National Institute of Health Research (NIHR) Oxford Biomedical Research Centre, University of Oxford, Oxford, United Kingdom
| | - Antonio Rodriguez-Romera
- MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine and National Institute of Health Research (NIHR) Oxford Biomedical Research Centre, University of Oxford, Oxford, United Kingdom
| | - Jasmeet S. Reyat
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Vincent Drive, Birmingham, United Kingdom
| | - Aude-Anais Olijnik
- MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine and National Institute of Health Research (NIHR) Oxford Biomedical Research Centre, University of Oxford, Oxford, United Kingdom
| | - Michela Colombo
- MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine and National Institute of Health Research (NIHR) Oxford Biomedical Research Centre, University of Oxford, Oxford, United Kingdom
| | - Guanlin Wang
- MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine and National Institute of Health Research (NIHR) Oxford Biomedical Research Centre, University of Oxford, Oxford, United Kingdom
- Centre for Computational Biology, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom
| | - Wei Xiong Wen
- MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine and National Institute of Health Research (NIHR) Oxford Biomedical Research Centre, University of Oxford, Oxford, United Kingdom
- Centre for Computational Biology, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom
| | - Nikolaos Sousos
- MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine and National Institute of Health Research (NIHR) Oxford Biomedical Research Centre, University of Oxford, Oxford, United Kingdom
- Cancer and Haematology Centre, Churchill Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford, United Kingdom
| | - Lauren C. Murphy
- MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine and National Institute of Health Research (NIHR) Oxford Biomedical Research Centre, University of Oxford, Oxford, United Kingdom
| | - Beata Grygielska
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Vincent Drive, Birmingham, United Kingdom
| | - Gina Perrella
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Vincent Drive, Birmingham, United Kingdom
| | - Christopher B. Mahony
- Rheumatology Research Group, Institute of Inflammation and Ageing, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Rebecca E. Ling
- MRC Weatherall Institute of Molecular Medicine, Department of Paediatrics and National Institute of Health Research (NIHR) Oxford Biomedical Research Centre, University of Oxford, Oxford, United Kingdom
| | - Natalina E. Elliott
- MRC Weatherall Institute of Molecular Medicine, Department of Paediatrics and National Institute of Health Research (NIHR) Oxford Biomedical Research Centre, University of Oxford, Oxford, United Kingdom
| | - Christina Simoglou Karali
- MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine and National Institute of Health Research (NIHR) Oxford Biomedical Research Centre, University of Oxford, Oxford, United Kingdom
| | - Andrew P. Stone
- Vascular Biology Program, Boston Children's Hospital, Department of Surgery, Harvard Medical School, Boston, Massachusetts
| | - Samuel Kemble
- Rheumatology Research Group, Institute of Inflammation and Ageing, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Emily A. Cutler
- University College London Cancer Institute, London, United Kingdom
| | | | - Adam P. Croft
- Rheumatology Research Group, Institute of Inflammation and Ageing, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - David Bassett
- Healthcare Technologies Institute, School of Chemical Engineering, University of Birmingham, Birmingham, United Kingdom
| | | | - Anindita Roy
- MRC Weatherall Institute of Molecular Medicine, Department of Paediatrics and National Institute of Health Research (NIHR) Oxford Biomedical Research Centre, University of Oxford, Oxford, United Kingdom
| | - Sarah Gooding
- MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine and National Institute of Health Research (NIHR) Oxford Biomedical Research Centre, University of Oxford, Oxford, United Kingdom
- Cancer and Haematology Centre, Churchill Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford, United Kingdom
| | - Julie Rayes
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Vincent Drive, Birmingham, United Kingdom
| | - Kellie R. Machlus
- Vascular Biology Program, Boston Children's Hospital, Department of Surgery, Harvard Medical School, Boston, Massachusetts
| | - Bethan Psaila
- MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine and National Institute of Health Research (NIHR) Oxford Biomedical Research Centre, University of Oxford, Oxford, United Kingdom
- Cancer and Haematology Centre, Churchill Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford, United Kingdom
| |
Collapse
|
12
|
Forrestal DP, Allenby MC, Simpson B, Klein TJ, Woodruff MA. Personalized Volumetric Tissue Generation by Enhancing Multiscale Mass Transport through 3D Printed Scaffolds in Perfused Bioreactors. Adv Healthc Mater 2022; 11:e2200454. [PMID: 35765715 PMCID: PMC11468985 DOI: 10.1002/adhm.202200454] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 06/06/2022] [Indexed: 01/28/2023]
Abstract
Engineered tissues provide an alternative to graft material, circumventing the use of donor tissue such as autografts or allografts and non-physiological synthetic implants. However, their lack of vasculature limits the growth of volumetric tissue more than several millimeters thick which limits their success post-implantation. Perfused bioreactors enhance nutrient mass transport inside lab-grown tissue but remain poorly customizable to support the culture of personalized implants. Here, a multiscale framework of computational fluid dynamics (CFD), additive manufacturing, and a perfusion bioreactor system are presented to engineer personalized volumetric tissue in the laboratory. First, microscale 3D printed scaffold pore geometries are designed and 3D printed to characterize media perfusion through CFD and experimental fluid testing rigs. Then, perfusion bioreactors are custom-designed to combine 3D printed scaffolds with flow-focusing inserts in patient-specific shapes as simulated using macroscale CFD. Finally, these computationally optimized bioreactor-scaffold assemblies are additively manufactured and cultured with pre-osteoblast cells for 7, 20, and 24 days to achieve tissue growth in the shape of human calcaneus bones of 13 mL volume and 1 cm thickness. This framework enables an intelligent model-based design of 3D printed scaffolds and perfusion bioreactors which enhances nutrient transport for long-term volumetric tissue growth in personalized implant shapes. The novel methods described here are readily applicable for use with different cell types, biomaterials, and scaffold microstructures to research therapeutic solutions for a wide range of tissues.
Collapse
Affiliation(s)
- David P Forrestal
- Centre for Biomedical TechnologiesQueensland University of Technology60 Musk AvenueKelvin GroveQueensland4059Australia
- Herston Biofabrication InstituteMetro North Hospital and Health Service7 Butterfield StHerstonQueensland4029Australia
- School of Mechanical and Mining EngineeringThe University of QueenslandStaff House RdSt LuciaQueensland4072Australia
| | - Mark C Allenby
- Centre for Biomedical TechnologiesQueensland University of Technology60 Musk AvenueKelvin GroveQueensland4059Australia
- School of Chemical EngineeringUniversity of QueenslandStaff House RdSt LuciaQueensland4072Australia
| | - Benjamin Simpson
- School of Science and TechnologyNottingham Trent UniversityClifton Campus RdNottinghamNG11 8NFUK
| | - Travis J Klein
- Centre for Biomedical TechnologiesQueensland University of Technology60 Musk AvenueKelvin GroveQueensland4059Australia
| | - Maria A Woodruff
- Centre for Biomedical TechnologiesQueensland University of Technology60 Musk AvenueKelvin GroveQueensland4059Australia
| |
Collapse
|
13
|
Moses JC, Dey S, Bandyopadhyay A, Agarwala M, Mandal BB. Silk-Based Bioengineered Diaphyseal Cortical Bone Unit Enclosing an Implantable Bone Marrow toward Atrophic Nonunion Grafting. Adv Healthc Mater 2022; 11:e2102031. [PMID: 34881525 DOI: 10.1002/adhm.202102031] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 12/02/2021] [Indexed: 12/11/2022]
Abstract
Postnatal fracture healing of atrophic long bone diaphyseal nonunions remains a challenge for orthopedic surgeons. Paucity of autologous spongiosa has potentiated the use of tissue engineered bone grafts to improve success rates of bone marrow engraftment used in plate reosteosynthesis. Herein, the development and in vitro validation of a "sandwich-type" biofabricated diaphyseal cross-sectional unit, with an outer mechanically robust bioprinted cortical bone shell, encompassing an engineered bone marrow, are reported. Channelized silk fibroin blend sponges derived from Bombyx mori and Antheraea assama help in developing compartmentalized endosteum, exhibiting specialized osteoblasts (endosteal niche) and discontinuous endothelium (vascular niche). The cellular cross-talk between these two niches triggered via integrin-mediated cell adhesion, enables in preserving quiescence state of CD34+ /CD38- hematopoietic stem cells and their recycling in the engineered marrow. The outer cortical bone strut is developed through multimaterial microextrusion bioprinting strategy. Osteogenically primed mesenchymal stem cells-laden silk fibroin-nano-hydroxyapatite bioink is bioprinted alongside paramagnetic Fe-doped bioactive glass-polycaprolactone blend thermoplastic ink, reinforcing it for mechanical stability. Pulsed magnetic field actuation positively influences the osteogenic commitment and maturation of the bioprinted constructs via mechanotransductory route. Therefore, the assembled engineered marrow and bioprinted cortical shell hold promise as potential orthobiologic substitutes toward atrophic nonunion repairs.
Collapse
Affiliation(s)
- Joseph Christakiran Moses
- Biomaterials and Tissue Engineering Laboratory Department of Biosciences and Bioengineering Indian Institute of Technology Guwahati Guwahati Assam 781039 India
| | - Souradeep Dey
- Centre for Nanotechnology Indian Institute of Technology Guwahati Guwahati Assam 781039 India
| | - Ashutosh Bandyopadhyay
- Biomaterials and Tissue Engineering Laboratory Department of Biosciences and Bioengineering Indian Institute of Technology Guwahati Guwahati Assam 781039 India
| | - Manoj Agarwala
- GNRC Institute of Medical Sciences (formerly known as Guwahati Neurological Research Centre) Guwahati Assam 781039 India
| | - Biman B. Mandal
- Biomaterials and Tissue Engineering Laboratory Department of Biosciences and Bioengineering Indian Institute of Technology Guwahati Guwahati Assam 781039 India
- Centre for Nanotechnology Indian Institute of Technology Guwahati Guwahati Assam 781039 India
- School of Health Science and Technology Indian Institute of Technology Guwahati Guwahati Assam 781039 India
| |
Collapse
|
14
|
Luc NF, Rohner N, Girish A, Sekhon UDS, Neal MD, Gupta AS. Bioinspired artificial platelets: past, present and future. Platelets 2022; 33:35-47. [PMID: 34455908 PMCID: PMC8795470 DOI: 10.1080/09537104.2021.1967916] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Platelets are anucleate blood cells produced from megakaryocytes predominantly in the bone marrow and released into blood circulation at a healthy count of 150,000-400,00 per μL and circulation lifespan of 7-9 days. Platelets are the first responders at the site of vascular injury and bleeding, and participate in clot formation via injury site-specific primary mechanisms of adhesion, activation and aggregation to form a platelet plug, as well as secondary mechanisms of augmenting coagulation via thrombin amplification and fibrin generation. Platelets also secrete various granule contents that enhance these mechanisms for clot growth and stability. The resultant clot seals the injury site to stanch bleeding, a process termed as hemostasis. Due to this critical role, a reduction in platelet count or dysregulation in platelet function is associated with bleeding risks and hemorrhagic complications. These scenarios are often treated by prophylactic or emergency transfusion of platelets. However, platelet transfusions face significant challenges due to limited donor availability, difficult portability and storage, high bacterial contamination risks, and very short shelf life (~5-7 days). These are currently being addressed by a robust volume of research involving reduced temperature storage and pathogen reduction processes on donor platelets to improve shelf-life and reduce contamination, as well as bioreactor-based approaches to generate donor-independent platelets from stem cells in vitro. In parallel, a complementary research field has emerged that involves the design of artificial platelets utilizing biosynthetic particle constructs that functionally emulate various hemostatic mechanisms of platelets. Here, we provide a comprehensive review of the history and the current state-of-the-art artificial platelet approaches, along with discussing the translational opportunities and challenges.
Collapse
Affiliation(s)
- Norman F. Luc
- Case Western Reserve University, Department of Biomedical Engineering, Cleveland, OH 44106, USA
| | - Nathan Rohner
- Case Western Reserve University, Department of Biomedical Engineering, Cleveland, OH 44106, USA
| | - Aditya Girish
- Case Western Reserve University, Department of Biomedical Engineering, Cleveland, OH 44106, USA
| | | | - Matthew D. Neal
- University of Pittsburgh, Pittsburgh Trauma Research Center, Department of Surgery, Pittsburgh, PA 15123, USA
| | - Anirban Sen Gupta
- Case Western Reserve University, Department of Biomedical Engineering, Cleveland, OH 44106, USA
| |
Collapse
|
15
|
Jansen LE, Kim H, Hall CL, McCarthy TP, Lee MJ, Peyton SR. A poly(ethylene glycol) three-dimensional bone marrow hydrogel. Biomaterials 2022; 280:121270. [PMID: 34890973 PMCID: PMC8890749 DOI: 10.1016/j.biomaterials.2021.121270] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 11/19/2021] [Accepted: 11/22/2021] [Indexed: 01/03/2023]
Abstract
Three-dimensional (3D) hydrogels made from synthetic polymers have emerged as in vitro cell culture platforms capable of representing the extracellular geometry, modulus, and water content of tissues in a tunable fashion. Hydrogels made from these otherwise non-bioactive polymers can be decorated with short peptides derived from proteins naturally found in tissues to support cell viability and direct phenotype. We identified two key limitations that limit the ability of this class of materials to recapitulate real tissue. First, these environments typically display between 1 and 3 bioactive peptides, which vastly underrepresents the diversity of proteins found in the extracellular matrix (ECM) of real tissues. Second, peptides chosen are ubiquitous in ECM and not derived from proteins found in specific tissues, per se. To overcome this critical limitation in hydrogel design and functionality, we developed an approach to incorporate the complex and specific protein signature of bone marrow into a poly (ethylene glycol) (PEG) hydrogel. This bone marrow hydrogel mimics the elasticity of marrow and has 20 bone marrow-specific and cell-instructive peptides. We propose this tissue-centric approach as the next generation of 3D hydrogel design for applications in tissue engineering and beyond.
Collapse
Affiliation(s)
- Lauren E Jansen
- Department of Chemical Engineering, University of Massachusetts Amherst, USA
| | - Hyuna Kim
- Molecular and Cellular Biology Graduate Program, University of Massachusetts Amherst, USA
| | - Christopher L Hall
- Department of Chemical Engineering, University of Massachusetts Amherst, USA
| | - Thomas P McCarthy
- Department of Chemical Engineering, University of Massachusetts Amherst, USA
| | - Michael J Lee
- Department of Systems Biology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Shelly R Peyton
- Department of Chemical Engineering, University of Massachusetts Amherst, USA; Molecular and Cellular Biology Graduate Program, University of Massachusetts Amherst, USA; Institute for Applied Life Sciences, University of Massachusetts Amherst 240 Thatcher Way, Life Sciences Laboratory N531, Amherst, MA, 01003, USA.
| |
Collapse
|
16
|
Jaffredo T, Balduini A, Bigas A, Bernardi R, Bonnet D, Canque B, Charbord P, Cumano A, Delwel R, Durand C, Fibbe W, Forrester L, de Franceschi L, Ghevaert C, Gjertsen B, Gottgens B, Graf T, Heidenreich O, Hermine O, Higgs D, Kleanthous M, Klump H, Kouskoff V, Krause D, Lacaud G, Celso CL, Martens JH, Méndez-Ferrer S, Menendez P, Oostendorp R, Philipsen S, Porse B, Raaijmakers M, Robin C, Stunnenberg H, Theilgaard-Mönch K, Touw I, Vainchenker W, Corrons JLV, Yvernogeau L, Schuringa JJ. The EHA Research Roadmap: Normal Hematopoiesis. Hemasphere 2021; 5:e669. [PMID: 34853826 PMCID: PMC8615310 DOI: 10.1097/hs9.0000000000000669] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 11/02/2021] [Indexed: 01/01/2023] Open
Affiliation(s)
- Thierry Jaffredo
- Sorbonne Université, Institut de Biologie Paris Seine, Laboratoire de Biologie du Développement/UMR7622, Paris, France
| | | | - Anna Bigas
- Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain
- Josep Carreras Leukemia Research Institute (IJC), Barcelona, Spain
- Centro de Investigación Biomedica en Red-Oncología (CIBERONC), Instituto de Salud Carlos III, Madrid, Spain
| | - Rosa Bernardi
- IRCCS San Raffaele Scientific Institute, Milan, Italy
| | | | - Bruno Canque
- INSERM U976, Universite de Paris, Ecole Pratique des Hautes Etudes/PSL Research University, Institut de Recherche Saint Louis, France
| | - Pierre Charbord
- Sorbonne Université, Institut de Biologie Paris Seine, Laboratoire de Biologie du Développement/UMR7622, Paris, France
| | - Anna Cumano
- Unité Lymphopoïèse, Département d’Immunologie, INSERM U1223, Institut Pasteur, Cellule Pasteur, Université de Paris, France
| | - Ruud Delwel
- Department of Hematology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Charles Durand
- Sorbonne Université, Institut de Biologie Paris Seine, Laboratoire de Biologie du Développement/UMR7622, Paris, France
| | - Willem Fibbe
- Leiden University Medical Center, The Netherlands
| | - Lesley Forrester
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, University of Edinburgh, Scotland
| | | | | | - Bjørn Gjertsen
- Department of Medicine, Hematology Section, Haukeland University Hospital, Bergen, Norway
- Department of Clinical Science, Centre for Cancer Biomarkers CCBIO, University of Bergen, Norway
| | - Berthold Gottgens
- Wellcome - MRC Cambridge Stem Cell Institute and Department of Haematology, University of Cambridge, United Kingdom
| | - Thomas Graf
- Center for Genomic Regulation, Barcelona Institute for Science and Technology and Universitat Pompeu Fabra, Barcelona, Spain
| | - Olaf Heidenreich
- Prinses Máxima Centrum voor kinderoncologie, Utecht, The Netherlands
| | - Olivier Hermine
- Department of Hematology and Laboratory of Physiopathology and Treatment of Blood Disorders, Hôpital Necker, Imagine institute, University of Paris, France
| | - Douglas Higgs
- MRC Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, Oxford, United Kingdom
| | | | - Hannes Klump
- Institute for Transfusion Medicine, University Hospital Essen, Germany
| | | | - Daniela Krause
- Goethe University Frankfurt and Georg-Speyer-Haus, Frankfurt am Main, Germany
| | - George Lacaud
- Cancer Research UK Manchester Institute, The University of Manchester, United Kingdom
| | | | - Joost H.A. Martens
- Department of Molecular Biology, RIMLS, Radboud University, Nijmegen, The Netherlands
| | | | - Pablo Menendez
- Centro de Investigación Biomedica en Red-Oncología (CIBERONC), Instituto de Salud Carlos III, Madrid, Spain
- Josep Carreras Leukemia Research Institute, Barcelona, Spain
- RICORS-RETAV, Instituto de Salud Carlos III, Madrid, Spain
- Department of Biomedicine, School of Medicine, Universitat de Barcelona, Barcelona, Spain
- Institució Catalana de Recerca i Estudis Avancats (ICREA), Barcelona, Spain
| | - Robert Oostendorp
- Department of Internal Medicine III, Technical University of Munich, School of Medicine, Germany
| | - Sjaak Philipsen
- Department of Cell Biology, Erasmus University Medical Center Rotterdam, The Netherlands
| | - Bo Porse
- The Finsen Laboratory, Rigshospitalet, Faculty of Health Sciences, University of Copenhagen, Denmark
- Biotech Research and Innovation Center (BRIC), University of Copenhagen, Denmark
- Novo Nordisk Foundation Center for Stem Cell Biology, DanStem, Faculty of Health Sciences, University of Copenhagen, Denmark
| | - Marc Raaijmakers
- Department of Hematology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Catherine Robin
- Hubrecht Institute-KNAW and University Medical Center Utrecht, The Netherlands
- Regenerative medicine center, University Medical Center Utrecht, The Netherlands
| | - Henk Stunnenberg
- Prinses Máxima Centrum voor kinderoncologie, Utecht, The Netherlands
| | - Kim Theilgaard-Mönch
- The Finsen Laboratory, Rigshospitalet, Faculty of Health Sciences, University of Copenhagen, Denmark
- Biotech Research and Innovation Center (BRIC), University of Copenhagen, Denmark
- Novo Nordisk Foundation Center for Stem Cell Biology, DanStem, Faculty of Health Sciences, University of Copenhagen, Denmark
- Department of Hematology, Rigshospitalet/National University Hospital, University of Copenhagen, Denmark
| | - Ivo Touw
- Department of Hematology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | | | - Joan-Lluis Vives Corrons
- Red Blood Cell and Hematopoietic Disorders Research Unit, Institute for Leukaemia Research Josep Carreras, Badalona, Barcelona
| | - Laurent Yvernogeau
- Sorbonne Université, Institut de Biologie Paris Seine, Laboratoire de Biologie du Développement/UMR7622, Paris, France
| | - Jan Jacob Schuringa
- Department of Experimental Hematology, University Medical Center Groningen, The Netherlands
| |
Collapse
|
17
|
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
|
18
|
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
|
19
|
Leung J, Cau MF, Kastrup CJ. Emerging gene therapies for enhancing the hemostatic potential of platelets. Transfusion 2021; 61 Suppl 1:S275-S285. [PMID: 34269451 DOI: 10.1111/trf.16519] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 03/08/2021] [Accepted: 03/12/2021] [Indexed: 01/03/2023]
Abstract
Platelet transfusions are an integral component of balanced hemostatic resuscitation protocols used to manage severe hemorrhage following trauma. Enhancing the hemostatic potential of platelets could lead to further increases in the efficacy of transfusions, particularly for non-compressible torso hemorrhage or severe hemorrhage with coagulopathy, by decreasing blood loss and improving overall patient outcomes. Advances in gene therapies, including RNA therapies, are leading to new strategies to enhance platelets for better control of hemorrhage. This review will highlight three approaches for creating modified platelets using gene therapies: (i) direct transfection of transfusable platelets ex vivo, (ii) in vitro production of engineered platelets from platelet-precursor cells, and (iii) modifying the bone marrow for in vivo production of modified platelets. In summary, modifying platelets to enhance their hemostatic potential is an exciting new frontier in transfusion medicine, but more preclinical development as well as studies testing the safety and efficacy of these agents are needed.
Collapse
Affiliation(s)
- Jerry Leung
- Michael Smith Laboratories, University of British Columbia, Vancouver, British Columbia, Canada.,Centre for Blood Research, University of British Columbia, Vancouver, British Columbia, Canada.,Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Massimo F Cau
- Michael Smith Laboratories, University of British Columbia, Vancouver, British Columbia, Canada.,Centre for Blood Research, University of British Columbia, Vancouver, British Columbia, Canada.,School of Biomedical Engineering, University of British Columbia, Vancouver, British Columbia, Canada
| | - Christian J Kastrup
- Michael Smith Laboratories, University of British Columbia, Vancouver, British Columbia, Canada.,Centre for Blood Research, University of British Columbia, Vancouver, British Columbia, Canada.,Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
20
|
Allenby MC, Okutsu N, Brailey K, Guasch J, Zhang Q, Panoskaltsis N, Mantalaris A. A spatiotemporal microenvironment model to improve design of a 3D bioreactor for red cell production. Tissue Eng Part A 2021; 28:38-53. [PMID: 34130508 DOI: 10.1089/ten.tea.2021.0028] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Cellular microenvironments provide stimuli including paracrine and autocrine growth factors and physico-chemical cues, which support efficient in vivo cell production unmatched by current in vitro biomanufacturing platforms. While three-dimensional (3D) culture systems aim to recapitulate niche architecture and function of the target tissue/organ, they are limited in accessing spatiotemporal information to evaluate and optimize in situ cell/tissue process development. Herein, a mathematical modelling framework is parameterized by single-cell phenotypic imaging and multiplexed biochemical assays to simulate the non-uniform tissue distribution of nutrients/metabolites and growth factors in cell niche environments. This model is applied to a bone marrow mimicry 3D perfusion bioreactor containing dense stromal and hematopoietic tissue with limited red blood cell (RBC) egress. The model characterized an imbalance between endogenous cytokine production and nutrient starvation within the microenvironmental niches, and recommended increased cell inoculum density and enhanced medium exchange, guiding the development of a miniaturized prototype bioreactor. The second-generation prototype improved the distribution of nutrients and growth factors and supported a 50-fold increase in RBC production efficiency. This image-informed bioprocess modelling framework leverages spatiotemporal niche information to enhance biochemical factor utilization and improve cell manufacturing in 3D systems.
Collapse
Affiliation(s)
- Mark Colin Allenby
- Queensland University of Technology, 1969, Institute of Health and Biomedical Innovation (IHBI), Kelvin Grove, Queensland, Australia.,Imperial College London, 4615, Department of Chemical Engineering, London, London, United Kingdom of Great Britain and Northern Ireland;
| | - Naoki Okutsu
- Imperial College London, 4615, Department of Chemical Engineering, London, London, United Kingdom of Great Britain and Northern Ireland;
| | - Kate Brailey
- Imperial College London, 4615, Department of Chemical Engineering, London, London, United Kingdom of Great Britain and Northern Ireland;
| | - Joana Guasch
- Imperial College London, 4615, Department of Chemical Engineering, London, London, United Kingdom of Great Britain and Northern Ireland;
| | - Qiming Zhang
- Imperial College London, 4615, Department of Chemical Engineering, London, London, United Kingdom of Great Britain and Northern Ireland;
| | - Nicki Panoskaltsis
- Emory University, 1371, Winship Cancer Institute, Department of Hematology & Medical Oncology, Atlanta, Georgia, United States.,Imperial College London, 4615, Department of Haematology, London, London, United Kingdom of Great Britain and Northern Ireland;
| | - Athanasios Mantalaris
- Georgia Institute of Technology, 1372, BME, Atlanta, Georgia, United States.,Imperial College London, 4615, Department of Chemical Engineering, London, London, United Kingdom of Great Britain and Northern Ireland;
| |
Collapse
|
21
|
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
|
22
|
Jiang Z, Zhang K, Du L, Cheng Z, Zhang T, Ding J, Li W, Xu B, Zhu M. Construction of chitosan scaffolds with controllable microchannel for tissue engineering and regenerative medicine. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 126:112178. [PMID: 34082978 DOI: 10.1016/j.msec.2021.112178] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 04/16/2021] [Accepted: 05/04/2021] [Indexed: 12/16/2022]
Abstract
Microchannels are effective means of enabling the functional performance of tissue engineering scaffolds. Chitosan, a partial deacetylation derivative of chitin, exhibiting excellent biocompatibility, has been widely used in clinical practice. However, development of chitosan scaffolds with controllable microchannels architecture remains an engineering challenge. Here, we generated chitosan scaffolds with adjustable microchannel by combining a 3D printing microfiber templates-leaching method and a freeze-drying method. We can precisely control the arrangement, diameter and density of microchannel within chitosan scaffolds. Moreover, the integrated bilayer scaffolds with the desired structural parameters in each layer were fabricated and exhibited no delamination. The flow rate and volume of the simulated fluid can be modulated by diverse channels architecture. Additionally, the microchannel structure promoted cell survival, proliferation and distribution in vitro, and improved cell and tissue ingrowth and vascular formation in vivo. This study opens a new road for constructing chitosan scaffolds, and can further extend their application scope across tissue engineering and regenerative medicine.
Collapse
Affiliation(s)
- Zhuyan Jiang
- The Second Hospital of Tianjin Medical University, Tianjin Medical University, Tianjin 300070, China; Graduate School, Tianjin Medical University, Tianjin 300070, China
| | - Kaihui Zhang
- Graduate School, Tianjin Medical University, Tianjin 300070, China; Department of Minimally Invasive Spine Surgery, Tianjin Hospital, Tianjin 300211, China
| | - Lilong Du
- Department of Minimally Invasive Spine Surgery, Tianjin Hospital, Tianjin 300211, China.
| | - Zhaojun Cheng
- Department of Minimally Invasive Spine Surgery, Tianjin Hospital, Tianjin 300211, China
| | - Tongxing Zhang
- Department of Minimally Invasive Spine Surgery, Tianjin Hospital, Tianjin 300211, China
| | - Ji Ding
- Graduate School, Tianjin Medical University, Tianjin 300070, China; Department of Minimally Invasive Spine Surgery, Tianjin Hospital, Tianjin 300211, China
| | - Wen Li
- College of Life Sciences, Key Laboratory of Bioactive Materials (Ministry of Education), State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin 300071, China
| | - Baoshan Xu
- Graduate School, Tianjin Medical University, Tianjin 300070, China; Department of Minimally Invasive Spine Surgery, Tianjin Hospital, Tianjin 300211, China.
| | - Meifeng Zhu
- College of Life Sciences, Key Laboratory of Bioactive Materials (Ministry of Education), State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin 300071, China.
| |
Collapse
|
23
|
Mu X, Agostinacchio F, Xiang N, Pei Y, Khan Y, Guo C, Cebe P, Motta A, Kaplan DL. Recent Advances in 3D Printing with Protein-Based Inks. Prog Polym Sci 2021; 115:101375. [PMID: 33776158 PMCID: PMC7996313 DOI: 10.1016/j.progpolymsci.2021.101375] [Citation(s) in RCA: 55] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Three-dimensional (3D) printing is a transformative manufacturing strategy, allowing rapid prototyping, customization, and flexible manipulation of structure-property relationships. Proteins are particularly appealing to formulate inks for 3D printing as they serve as essential structural components of living systems, provide a support presence in and around cells and for tissue functions, and also provide the basis for many essential ex vivo secreted structures in nature. Protein-based inks are beneficial in vivo due to their mechanics, chemical and physical match to the specific tissue, and full degradability, while also to promoting implant-host integration and serving as an interface between technology and biology. Exploiting the biological, chemical, and physical features of protein-based inks can provide key opportunities to meet the needs of tissue engineering and regenerative medicine. Despite these benefits, protein-based inks impose nontrivial challenges to 3D printing such as concentration and rheological features and reconstitution of the structural hierarchy observed in nature that is a source of the robust mechanics and functions of these materials. This review introduces photo-crosslinking mechanisms and rheological principles that underpins a variety of 3D printing techniques. The review also highlights recent advances in the design, development, and biomedical utility of monolithic and composite inks from a range of proteins, including collagen, silk, fibrinogen, and others. One particular focus throughout the review is to introduce unique material characteristics of proteins, including amino acid sequences, molecular assembly, and secondary conformations, which are useful for designing printing inks and for controlling the printed structures. Future perspectives of 3D printing with protein-based inks are also provided to support the promising spectrum of biomedical research accessible to these materials.
Collapse
Affiliation(s)
- Xuan Mu
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155, USA
| | - Francesca Agostinacchio
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155, USA
- Department of Industrial Engineering, University of Trento, via Sommarive 9, Trento 38123, Italy
| | - Ning Xiang
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155, USA
| | - Ying Pei
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155, USA
- School of Materials Science and Engineering, Zhengzhou University, Zhengzhou 450001, China
| | - Yousef Khan
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155, USA
| | - Chengchen Guo
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155, USA
| | - Peggy Cebe
- Department of Physics and Astronomy, Tufts University, Medford, MA 02155, USA
| | - Antonella Motta
- Department of Industrial Engineering, University of Trento, via Sommarive 9, Trento 38123, Italy
| | - David L. Kaplan
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155, USA
| |
Collapse
|
24
|
Szymkowiak S, Sandler N, Kaplan DL. Aligned Silk Sponge Fabrication and Perfusion Culture for Scalable Proximal Tubule Tissue Engineering. ACS APPLIED MATERIALS & INTERFACES 2021; 13:10768-10777. [PMID: 33621042 DOI: 10.1021/acsami.1c00548] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Chronic kidney disease and kidney failure are on the rise globally, yet there has not been a corresponding improvement in available therapies. A key challenge in a biological approach to developing kidney tissue is to identify scaffolding materials that support cell growth both in vitro and in vivo to facilitate translational goals. Scaffolds composed of silk fibroin protein possess the biocompatibility, mechanical robustness, and stability required for tissue engineering. Here, we use a silk sponge system to support kidney cells in a perfused bioreactor system. Silk fibroin protein underwent directional freezing to form parallel porous structures that mimic the native kidney structure of aligned tubules and are able to support more cells than nonaligned silk sponges. Adult immortalized renal proximal tubule epithelial cells were seeded into the sponges and cultured under static conditions for 1 week, then grown statically or with perfusion with culture media flowing through the sponge to enhance cell alignment and maturation. The sponges were imaged with confocal and scanning electron microscopies to analyze and quantify cell attachment, alignment, and expression of proteins important to proximal tubule differentiation and function. The perfused tissue constructs showed higher number of cells that are more evenly distributed through the construct and increased gene expression of several key markers of proximal tubule epithelial cell function compared to sponges grown under static conditions. These perfused tissue constructs represent a step toward a scalable approach to engineering proximal tubule structures with the potential to be used as in vitro models or as in vivo implantable tissues to supplement or replace impaired kidney function.
Collapse
Affiliation(s)
- Sophia Szymkowiak
- Department of Biomedical Engineering, Tufts University, 4 Colby Street, Medford, Massachusetts 02155, United States
| | - Nathan Sandler
- Department of Biomedical Engineering, Tufts University, 4 Colby Street, Medford, Massachusetts 02155, United States
| | - David L Kaplan
- Department of Biomedical Engineering, Tufts University, 4 Colby Street, Medford, Massachusetts 02155, United States
| |
Collapse
|
25
|
Mu X, Sahoo JK, Cebe P, Kaplan DL. Photo-Crosslinked Silk Fibroin for 3D Printing. Polymers (Basel) 2020; 12:E2936. [PMID: 33316890 PMCID: PMC7763742 DOI: 10.3390/polym12122936] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 12/04/2020] [Accepted: 12/07/2020] [Indexed: 12/14/2022] Open
Abstract
Silk fibroin in material formats provides robust mechanical properties, and thus is a promising protein for 3D printing inks for a range of applications, including tissue engineering, bioelectronics, and bio-optics. Among the various crosslinking mechanisms, photo-crosslinking is particularly useful for 3D printing with silk fibroin inks due to the rapid kinetics, tunable crosslinking dynamics, light-assisted shape control, and the option to use visible light as a biocompatible processing condition. Multiple photo-crosslinking approaches have been applied to native or chemically modified silk fibroin, including photo-oxidation and free radical methacrylate polymerization. The molecular characteristics of silk fibroin, i.e., conformational polymorphism, provide a unique method for crosslinking and microfabrication via light. The molecular design features of silk fibroin inks and the exploitation of photo-crosslinking mechanisms suggest the exciting potential for meeting many biomedical needs in the future.
Collapse
Affiliation(s)
- Xuan Mu
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155, USA; (X.M.); (J.K.S.)
| | - Jugal Kishore Sahoo
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155, USA; (X.M.); (J.K.S.)
| | - Peggy Cebe
- Department of Physics and Astronomy, Tufts University, Medford, MA 02155, USA;
| | - David L. Kaplan
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155, USA; (X.M.); (J.K.S.)
| |
Collapse
|
26
|
Martínez-Botía P, Acebes-Huerta A, Seghatchian J, Gutiérrez L. On the Quest for In Vitro Platelet Production by Re-Tailoring the Concepts of Megakaryocyte Differentiation. ACTA ACUST UNITED AC 2020; 56:medicina56120671. [PMID: 33287459 PMCID: PMC7761839 DOI: 10.3390/medicina56120671] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Revised: 11/26/2020] [Accepted: 11/30/2020] [Indexed: 12/14/2022]
Abstract
The demand of platelet transfusions is steadily growing worldwide, inter-donor variation, donor dependency, or storability/viability being the main contributing factors to the current global, donor-dependent platelet concentrate shortage concern. In vitro platelet production has been proposed as a plausible alternative to cover, at least partially, the increasing demand. However, in practice, such a logical production strategy does not lack complexity, and hence, efforts are focused internationally on developing large scale industrial methods and technologies to provide efficient, viable, and functional platelet production. This would allow obtaining not only sufficient numbers of platelets but also functional ones fit for all clinical purposes and civil scenarios. In this review, we cover the evolution around the in vitro culture and differentiation of megakaryocytes into platelets, the progress made thus far to bring the culture concept from basic research towards good manufacturing practices certified production, and subsequent clinical trial studies. However, little is known about how these in vitro products should be stored or whether any safety measure should be implemented (e.g., pathogen reduction technology), as well as their quality assessment (how to isolate platelets from the rest of the culture cells, debris, microvesicles, or what their molecular and functional profile is). Importantly, we highlight how the scientific community has overcome the old dogmas and how the new perspectives influence the future of platelet-based therapy for transfusion purposes.
Collapse
Affiliation(s)
- Patricia Martínez-Botía
- Platelet Research Lab, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain; (P.M.-B.); (A.A.-H.)
- Department of Medicine, University of Oviedo, 33003 Oviedo, Spain
| | - Andrea Acebes-Huerta
- Platelet Research Lab, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain; (P.M.-B.); (A.A.-H.)
| | - Jerard Seghatchian
- International Consultancy in Strategic Safety/Quality Improvements of Blood-Derived Bioproducts and Suppliers Quality Audit/Inspection, London NW3 3AA, UK;
| | - Laura Gutiérrez
- Platelet Research Lab, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain; (P.M.-B.); (A.A.-H.)
- Department of Medicine, University of Oviedo, 33003 Oviedo, Spain
- Correspondence:
| |
Collapse
|
27
|
Bouet G, Mookerjee S, Foster H, Waller A, Ghevaert C. [From the bench to the clinic: The challenge of translating platelet production in vitro]. BULLETIN DE L'ACADEMIE NATIONALE DE MEDECINE 2020; 204:981-988. [PMID: 33078026 PMCID: PMC7553122 DOI: 10.1016/j.banm.2020.10.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Accepted: 06/02/2020] [Indexed: 11/22/2022]
Abstract
Platelet transfusions, which are currently totally dependent on altruistic donations, are absolutely necessary to the treatment of patients with thrombocytopenia following trauma, surgery or other pathologies (especially malignancies). Producing platelets in vitro represent a major technological and scientific breathrough that would address logistical issues (supply chain, stock holding…) and medical concerns (compatibility and biosafety). The translation of this innovation will need to be accompanied by rigorous quality control, harmonised between laboratory when it comes to functionality and biosafety for use in the clinic.
Collapse
Affiliation(s)
- G Bouet
- Mines Saint-Étienne, université Lyon, université Jean-Monnet, Inserm, U 1059 Sainbiose, Centre CIS, Saint-Étienne, France
| | - S Mookerjee
- Wellcome trust-medical research council Cambridge Stem Cell Institute and department of haematology, university of Cambridge, CB2 0PT Cambridge, UK
- National health service blood and transplant, Cambridge biomedical campus, CB2 0PT Cambridge, UK
| | - H Foster
- Wellcome trust-medical research council Cambridge Stem Cell Institute and department of haematology, university of Cambridge, CB2 0PT Cambridge, UK
- National health service blood and transplant, Cambridge biomedical campus, CB2 0PT Cambridge, UK
| | - A Waller
- Wellcome trust-medical research council Cambridge Stem Cell Institute and department of haematology, university of Cambridge, CB2 0PT Cambridge, UK
- National health service blood and transplant, Cambridge biomedical campus, CB2 0PT Cambridge, UK
| | - C Ghevaert
- Wellcome trust-medical research council Cambridge Stem Cell Institute and department of haematology, university of Cambridge, CB2 0PT Cambridge, UK
- National health service blood and transplant, Cambridge biomedical campus, CB2 0PT Cambridge, UK
| |
Collapse
|
28
|
Bilayer nicorandil-loaded small-diameter vascular grafts improve endothelial cell function via PI3K/AKT/eNOS pathway. Biodes Manuf 2020. [DOI: 10.1007/s42242-020-00107-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
29
|
Yang J, Luan J, Shen Y, Chen B. Developments in the production of platelets from stem cells (Review). Mol Med Rep 2020; 23:7. [PMID: 33179095 PMCID: PMC7673345 DOI: 10.3892/mmr.2020.11645] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 10/13/2020] [Indexed: 01/01/2023] Open
Abstract
Platelets are small pieces of cytoplasm that have become detached from the cytoplasm of mature megakaryocytes (MKs) in the bone marrow. Platelets modulate vascular system integrity and serve important role, particularly in hemostasis. With the rapid development of clinical medicine, the demand for platelet transfusion as a life‑saving intervention increases continuously. Stem cell technology appears to be highly promising for transfusion medicine, and the generation of platelets from stem cells would be of great value in the clinical setting. Furthermore, several studies have been undertaken to investigate the potential of producing platelets from stem cells. Initial success has been achieved in terms of the yields and function of platelets generated from stem cells. However, the requirements of clinical practice remain unmet. The aim of the present review was to focus on several sources of stem cells and factors that induce MK differentiation. Updated information on current research into the genetic regulation of megakaryocytopoiesis and platelet generation was summarized. Additionally, advanced strategies of platelet generation were reviewed and the progress made in this field was discussed.
Collapse
Affiliation(s)
- Jie Yang
- Department of Hematology and Oncology, School of Medicine, Zhongda Hospital, Southeast University, Nanjing, Jiangsu 210009, P.R. China
| | - Jianfeng Luan
- Jinling Hospital Department of Blood Transfusion, School of Medicine, Nanjing University, Nanjing, Jiangsu 210002, P.R. China
| | - Yanfei Shen
- Medical School, School of Chemistry and Chemical Engineering, Southeast University, Nanjing, Jiangsu 210009, P.R. China
| | - Baoan Chen
- Department of Hematology and Oncology, School of Medicine, Zhongda Hospital, Southeast University, Nanjing, Jiangsu 210009, P.R. China
| |
Collapse
|
30
|
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
|
31
|
Lin X, Tang F, Jiang S, Khamis H, Bongers A, Whitelock JM, Lord MS, Rnjak‐Kovacina J. A Biomimetic Approach toward Enhancing Angiogenesis: Recombinantly Expressed Domain V of Human Perlecan Is a Bioactive Molecule That Promotes Angiogenesis and Vascularization of Implanted Biomaterials. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2020; 7:2000900. [PMID: 32995122 PMCID: PMC7507460 DOI: 10.1002/advs.202000900] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Revised: 04/27/2020] [Accepted: 04/28/2020] [Indexed: 05/07/2023]
Abstract
Angiogenic therapy involving delivery of pro-angiogenic growth factors to stimulate new blood vessel formation in ischemic disease is promising but has seen limited clinical success due to issues associated with the need to deliver supra-physiological growth factor concentrations. Bio-inspired growth factor delivery utilizing the native growth factor signaling roles of the extracellular matrix proteoglycans has the potential to overcome many of the drawbacks of angiogenic therapy. In this study, the potential of the recombinantly expressed domain V (rDV) of human perlecan is investigated as a means of promoting growth factor signaling toward enhanced angiogenesis and vascularization of implanted biomaterials. rDV is found to promote angiogenesis in established in vitro and in vivo angiogenesis assays by potentiating endogenous growth factor signaling via its glycosaminoglycan chains. Further, rDV is found to potentiate fibroblast growth factor 2 (FGF2) signaling at low concentrations that in the absence of rDV are not biologically active. Finally, rDV immobilized on 3D porous silk fibroin biomaterials promotes enhanced vascular ingrowth and integration of the implanted scaffolds with the surrounding tissue. Together, these studies demonstrate the important role of this biologically active perlecan fragment and its potential in the treatment of ischemia in both native and bioengineered tissues.
Collapse
Affiliation(s)
- Xiaoting Lin
- Graduate School of Biomedical EngineerinUniversity of New South WalesSydneyNSW2052Australia
| | - Fengying Tang
- Graduate School of Biomedical EngineerinUniversity of New South WalesSydneyNSW2052Australia
- Comparative Pathology ProgramDepartment of Comparative MedicineUniversity of Washington School of MedicineSeattleWA98195USA
| | - Shouyuan Jiang
- Graduate School of Biomedical EngineerinUniversity of New South WalesSydneyNSW2052Australia
| | - Heba Khamis
- Graduate School of Biomedical EngineerinUniversity of New South WalesSydneyNSW2052Australia
| | - Andre Bongers
- Biological Resources Imaging LaboratoryUniversity of New South WalesSydneyNSW2052Australia
| | - John M. Whitelock
- Graduate School of Biomedical EngineerinUniversity of New South WalesSydneyNSW2052Australia
| | - Megan S. Lord
- Graduate School of Biomedical EngineerinUniversity of New South WalesSydneyNSW2052Australia
| | - Jelena Rnjak‐Kovacina
- Graduate School of Biomedical EngineerinUniversity of New South WalesSydneyNSW2052Australia
| |
Collapse
|
32
|
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
|
33
|
Martínez-Botía P, Acebes-Huerta A, Seghatchian J, Gutiérrez L. In vitro platelet production for transfusion purposes: Where are we now? Transfus Apher Sci 2020; 59:102864. [PMID: 32646795 DOI: 10.1016/j.transci.2020.102864] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Over the last decade there has been a worldwide increase in the demand of platelet concentrates (PCs) for transfusion. This is, to a great extent, due to a growing and aging population with the concomitant increase in the incidence of onco-hematological diseases, which require frequent platelet (PLT) transfusions. Currently, PLTs are sourced uniquely from donations, and their storage time is limited only to a few days. The necessity to store PCs at room temperature (to minimize loss of PLT functional integrity), poses a major risk for bacterial contamination. While the implementation of pathogen reduction treatments (PRTs) and new-generation PLT additive solutions have allowed the extension of the shelf life and a safer PLT transfusion product, the concern of PCs shortage still pressures the scientific community to find alternative solutions with the aim of meeting the PLT transfusion increasing demand. In this concise report, we will focus on the efforts made to produce, in in vitro culture, high yields of viable and functional PLTs for transfusion purposes in a cost-effective manner, meeting not only current Good Manufacturing Practices (cGMPs), but also transfusion safety standards.
Collapse
Affiliation(s)
- Patricia Martínez-Botía
- Platelet Research Lab, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain; Dept. of Medicine, University of Oviedo, Spain
| | - Andrea Acebes-Huerta
- Platelet Research Lab, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain
| | - Jerard Seghatchian
- International Consultancy in Strategic Advices on Safety Improvements of Blood-Derived Bioproducts and Suppliers Quality Audit / Inspection, London, England, UK
| | - Laura Gutiérrez
- Platelet Research Lab, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain; Dept. of Medicine, University of Oviedo, Spain.
| |
Collapse
|
34
|
Mookerjee S, Foster HR, Waller AK, Ghevaert CJ. In vitro-derived platelets: the challenges we will have to face to assess quality and safety. Platelets 2020; 31:724-730. [PMID: 32486997 DOI: 10.1080/09537104.2020.1769051] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Platelet transfusions are given to patients in hospital who have a low blood platelet count (thrombocytopenia) either because of major bleeding (following trauma or surgery) or because the bone marrow production of platelets is impaired often due to chemotherapy, infiltration with malignant cells, fibrosis or genetic disorders. We are currently entirely reliant on blood donors as a source of platelets in transfusion medicine. However, the demand for platelets continues to rise, driven by an aging population, advances in medical procedures and ever more aggressive cancer therapies, while the supply of blood donors continues to remain static. In recent years, several groups have made major advances toward the generation of platelets in vitro for human transfusion. Recent successes include results in both generating mature human megakaryocytes as well as in developing bioreactors for extracting platelets from these megakaryocytes. Platelets made in vitro could address several issues inherent to platelets derived from blood donors - the ability to scale up/down more flexibly according to demand and therefore less precarious supply line, reduction of the risk of exposure to infectious agents and finally the possibility of engineering stem cells to reduce immunogenicity. Here we define the quality control tools and suggest measures for implementation across the field for in vitro platelet genesis, to aid collaboration between laboratories and to aid production of the burdens of proof that will eventually be required by regulators for efficacy and biosafety. We will do this firstly, by addressing the quality control of the nucleated cells used to make the platelets with a particular emphasis to safety issues and secondly, we will look at how platelet function measurement are addressed particularly in the context of platelets derived in vitro.
Collapse
Affiliation(s)
- S Mookerjee
- Wellcome-MRC Cambridge Stem Cell Institute, University of Cambridge , Cambridge, UK
| | - H R Foster
- Wellcome-MRC Cambridge Stem Cell Institute, University of Cambridge , Cambridge, UK
| | - A K Waller
- Wellcome-MRC Cambridge Stem Cell Institute, University of Cambridge , Cambridge, UK
| | - C J Ghevaert
- Wellcome-MRC Cambridge Stem Cell Institute, University of Cambridge , Cambridge, UK
| |
Collapse
|
35
|
Siavashani AZ, Mohammadi J, Rottmar M, Senturk B, Nourmohammadi J, Sadeghi B, Huber L, Maniura-Weber K. Silk fibroin/sericin 3D sponges: The effect of sericin on structural and biological properties of fibroin. Int J Biol Macromol 2020; 153:317-326. [DOI: 10.1016/j.ijbiomac.2020.02.316] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Revised: 02/16/2020] [Accepted: 02/26/2020] [Indexed: 12/19/2022]
|
36
|
Li L, Wang X, Li D, Qin J, Zhang M, Wang K, Zhao J, Zhang L. LBL deposition of chitosan/heparin bilayers for improving biological ability and reducing infection of nanofibers. Int J Biol Macromol 2020; 154:999-1006. [PMID: 32198036 DOI: 10.1016/j.ijbiomac.2020.03.152] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 03/16/2020] [Accepted: 03/16/2020] [Indexed: 12/21/2022]
Abstract
The complexity of cardiovascular disease requires that the materials for preparing vascular grafts possess good biocompatibility, high mechanical property, and even some excellent additional properties. In this study, polycaprolactone (PCL) with good mechanical property and natural source silk fibroin (SF) were electrospun into PCL/SF nanofibers to obtain the nanofibrous substrate. With the addition of SF, the mechanical property of PCL/SF nanofibrous mats was maintained to a certain extent. While, the hydrophilicity of PCL/SF nanofibrous mats was greatly improved which is more suitable for immersive layer-by-layer assembly (LBL). The oppositely charged heparin (Hep) and chitosan (CS) were alternatively deposited on the surface of PCL/SF nanofibers via LBL. After implanting human umbilical vein endothelial cells (HUVEC) on the LBL-structured nanofibrous mats for 48 h, it was confirmed that the CS/Hep bilayers enhanced the biocompatibility of the nanofibers. Furthermore, the results of the antibacterial test showed that the antibacterial effects of the LBL-structured nanofibrous mats for Escherichia coli and Staphylococcus aureus were both achieved 95% when the number of Hep/CS bilayer was 10. It can be demonstrated that the LBL-structured nanofibrous mats with improved biocompatibility and reduced infectivity had been prepared successfully, and can be potentially used in vascular grafts.
Collapse
Affiliation(s)
- Long Li
- Department of Cardiothoracic Surgery, ZhongNan Hospital of Wuhan University, Wuhan, Hubei 430060, China
| | - Xianguo Wang
- Department of Cardiothoracic Surgery, ZhongNan Hospital of Wuhan University, Wuhan, Hubei 430060, China
| | - Dan Li
- Hubei International Scientific and Technological Cooperation Base of Sustainable Resource and Energy, School of Resource and Environmental Science, Wuhan University, Wuhan 430079, China
| | - Jinfa Qin
- Department of Cardiothoracic Surgery, ZhongNan Hospital of Wuhan University, Wuhan, Hubei 430060, China
| | - Ming Zhang
- Department of Cardiothoracic Surgery, ZhongNan Hospital of Wuhan University, Wuhan, Hubei 430060, China
| | - Kaijie Wang
- Department of Cardiothoracic Surgery, ZhongNan Hospital of Wuhan University, Wuhan, Hubei 430060, China
| | - Jinping Zhao
- Department of Cardiothoracic Surgery, ZhongNan Hospital of Wuhan University, Wuhan, Hubei 430060, China.
| | - Li Zhang
- Department of Cardiothoracic Surgery, ZhongNan Hospital of Wuhan University, Wuhan, Hubei 430060, China.
| |
Collapse
|
37
|
Tang F, Manz XD, Bongers A, Odell RA, Joukhdar H, Whitelock JM, Lord MS, Rnjak-Kovacina J. Microchannels Are an Architectural Cue That Promotes Integration and Vascularization of Silk Biomaterials in Vivo. ACS Biomater Sci Eng 2020; 6:1476-1486. [DOI: 10.1021/acsbiomaterials.9b01624] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Fengying Tang
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, New South Wales 2052, Australia
| | - Xue D. Manz
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, New South Wales 2052, Australia
- Department of Pulmonary Medicine, Amsterdam UMC, VU University Medical Center, Amsterdam Cardiovascular Sciences (ACS), Amsterdam 1081 HV, The Netherlands
| | - Andre Bongers
- Biological Resources Imaging Laboratory, University of New South Wales, Sydney, New South Wales 2052, Australia
| | - Ross A. Odell
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, New South Wales 2052, Australia
| | - Habib Joukhdar
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, New South Wales 2052, Australia
| | - John M. Whitelock
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, New South Wales 2052, Australia
| | - Megan S. Lord
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, New South Wales 2052, Australia
| | - Jelena Rnjak-Kovacina
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, New South Wales 2052, Australia
| |
Collapse
|
38
|
Kukla DA, Stoppel WL, Kaplan DL, Khetani SR. Assessing the compatibility of primary human hepatocyte culture within porous silk sponges. RSC Adv 2020; 10:37662-37674. [PMID: 35515172 PMCID: PMC9057238 DOI: 10.1039/d0ra04954a] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Accepted: 10/04/2020] [Indexed: 12/24/2022] Open
Abstract
Donor organ shortages have prompted the development of alternative implantable human liver tissues for patients suffering from end-stage liver failure. Purified silk proteins provide desirable features for generating implantable tissues, including sustainable sourcing from insects/arachnids, biocompatibility, tunable mechanical properties and degradation rates, and low immunogenicity upon implantation. While different cell types were previously cultured for weeks within silk-based scaffolds, it remains unclear whether such scaffolds can be used to culture primary human hepatocytes (PHH) isolated from livers. Therefore, here we assessed the compatibility of PHH culture within porous silk scaffolds that enable diffusion of oxygen/nutrients through the pores. We found that incorporation of type I collagen during the fabrication and/or autoclaving of porous silk scaffolds, as opposed to simple adsorption of collagen onto pre-fabricated silk scaffolds, was necessary to enable robust PHH attachment/function. Scaffolds with small pores (73 ± 25 μm) promoted larger PHH spheroids and consequently higher PHH functions than large pores (235 ± 84 μm) for at least 1 month in culture. Further incorporation of supportive fibroblasts into scaffolds enhanced PHH functions up to 5-fold relative to scaffolds with PHHs alone and 2D co-cultures on plastic. Lastly, encapsulating PHHs within protein hydrogels while housed in the silk scaffold led to higher functions than protein hydrogel-only or silk-only controls. In conclusion, porous silk scaffolds containing extracellular matrix proteins can be used for the culture of PHHs ± supportive non-parenchymal cells, which can be further built on in the future to create optimized silk-based liver tissue surrogates for cell-based therapy. Porous silk scaffolds hybridized with extracellular matrix proteins are useful for culture of primary human hepatocytes ± supportive non-parenchymal cells.![]()
Collapse
Affiliation(s)
- David A. Kukla
- Department of Bioengineering
- University of Illinois at Chicago
- Chicago
- USA
| | | | - David L. Kaplan
- Department of Biomedical Engineering
- Tufts University
- Medford
- USA
| | - Salman R. Khetani
- Department of Bioengineering
- University of Illinois at Chicago
- Chicago
- USA
| |
Collapse
|
39
|
Baptista M, Joukhdar H, Alcala-Orozco CR, Lau K, Jiang S, Cui X, He S, Tang F, Heu C, Woodfield TBF, Lim KS, Rnjak-Kovacina J. Silk fibroin photo-lyogels containing microchannels as a biomaterial platform for in situ tissue engineering. Biomater Sci 2020; 8:7093-7105. [DOI: 10.1039/d0bm01010c] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Silk photo-lyogels fabricated by di-tyrosine photo-crosslinking and ice-templating silk fibroin on 3D printed templates toward in situ tissue engineering applications.
Collapse
|
40
|
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
|
41
|
Hang Y, Ma J, Li S, Zhang X, Liu B, Ding Z, Lu Q, Chen H, Kaplan DL. Structure–Chemical Modification Relationships with Silk Materials. ACS Biomater Sci Eng 2019; 5:2762-2768. [DOI: 10.1021/acsbiomaterials.9b00369] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Yingjie Hang
- College of Chemistry, Chemical Engineering and Materials Science & Collaborative Innovation Center of Suzhou Nano Science and Technology, Soochow University, Suzhou 215123, People’s Republic of China
| | - Jie Ma
- Department of Burns, Gansu Provincial Hospital, Lanzhou 730000, People’s Republic of China
| | - Siyuan Li
- College of Chemistry, Chemical Engineering and Materials Science & Collaborative Innovation Center of Suzhou Nano Science and Technology, Soochow University, Suzhou 215123, People’s Republic of China
| | - Xiaoyi Zhang
- National Engineering Laboratory for Modern Silk, Soochow University, Suzhou 215123, People’s Republic of China
| | - Bing Liu
- College of Chemistry, Chemical Engineering and Materials Science & Collaborative Innovation Center of Suzhou Nano Science and Technology, Soochow University, Suzhou 215123, People’s Republic of China
| | - Zhaozhao Ding
- National Engineering Laboratory for Modern Silk, Soochow University, Suzhou 215123, People’s Republic of China
| | - Qiang Lu
- College of Chemistry, Chemical Engineering and Materials Science & Collaborative Innovation Center of Suzhou Nano Science and Technology, Soochow University, Suzhou 215123, People’s Republic of China
- National Engineering Laboratory for Modern Silk, Soochow University, Suzhou 215123, People’s Republic of China
| | - Hong Chen
- College of Chemistry, Chemical Engineering and Materials Science & Collaborative Innovation Center of Suzhou Nano Science and Technology, Soochow University, Suzhou 215123, People’s Republic of China
| | - David L Kaplan
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts 02155, United States
| |
Collapse
|
42
|
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
|
43
|
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
|