1
|
Friend NE, Zhang IW, Hu MM, McCoy AJ, Kent RN, DePalma SJ, Baker BM, Lesher-Pérez SC, Stegemann JP, Putnam AJ. Biofabrication and Characterization of Vascularizing PEG-Norbornene Microgels. J Biomed Mater Res A 2025; 113:e37900. [PMID: 40135515 PMCID: PMC11956422 DOI: 10.1002/jbm.a.37900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Revised: 03/04/2025] [Accepted: 03/08/2025] [Indexed: 03/27/2025]
Abstract
Establishing a robust, functional microvascular network remains a critical challenge for both the revascularization of damaged or diseased tissues and the development of engineered biological materials. Vascularizing microgels may aid in efforts to develop complex, multiphasic tissues by providing discrete, vascularized tissue modules that can be distributed throughout engineered constructs to vascularize large volumes. Here, we fabricated poly(ethylene glycol)-norbornene (PEGNB) microgels containing endothelial and stromal cells via flow-focusing microfluidic droplet generation. When embedded in bulk fibrin hydrogels, these cell-laden microgels initiated the formation and development of robust microvascular networks. Furthermore, extended preculture of cell-laden PEGNB microgels enabled the formation of vessel-like structures supported by basement membrane within the matrix without aggregation. Our findings highlight the suitability of PEG-based matrices for the development of vascularizing microgels capable of forming well-distributed, robust microvascular networks.
Collapse
Affiliation(s)
- Nicole E. Friend
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI
| | - Irene W. Zhang
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI
| | - Michael M. Hu
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI
| | - Atticus J. McCoy
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI
| | - Robert N. Kent
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI
| | - Samuel J. DePalma
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI
| | - Brendon M. Baker
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI
| | - Sasha Cai Lesher-Pérez
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI
| | - Jan P. Stegemann
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI
| | - Andrew J. Putnam
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI
| |
Collapse
|
2
|
Wallace EJ, O'Dwyer J, Dolan EB, Burke LP, Wylie R, Bellavia G, Straino S, Cianfarani F, Ciotti G, Serini S, Calviello G, Roche ET, Mitra T, Duffy GP. Actuation-Mediated Compression of a Mechanoresponsive Hydrogel by Soft Robotics to Control Release of Therapeutic Proteins. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2401744. [PMID: 39692747 PMCID: PMC11831469 DOI: 10.1002/advs.202401744] [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: 02/26/2024] [Revised: 07/31/2024] [Indexed: 12/19/2024]
Abstract
Therapeutic proteins, the fastest growing class of pharmaceuticals, are subject to rapid proteolytic degradation in vivo, rendering them inactive. Sophisticated drug delivery systems that maintain protein stability, prolong therapeutic effects, and reduce administration frequency are urgently required. Herein, a mechanoresponsive hydrogel is developed contained within a soft robotic drug delivery (SRDD) device. In a step-change from previously reported systems, pneumatic actuation of this system releases the cationic therapeutic protein Vascular Endothelial Growth Factor (VEGF) in a bioactive form which is required for therapeutic angiogenesis, the growth of new blood vessels, in numerous clinical conditions. The ability of the SRDD device to release bioactive VEGF in a spatiotemporal manner from the hydrogel is tested in diabetic rats - a model in which angiogenesis is difficult to stimulate. Daily actuation of the SRDD device in the diabetic rat model significantly increased cluster of differentiation 31+ (CD31+) blood vessel number (p = 0.0335) and the diameter of alpha-smooth muscle actin+ (α-SMA+) blood vessels (p = 0.0025) compared to passive release of VEGF from non-actuated devices. The SRDD device combined with the mechanoresponsive hydrogel offers the potential to deliver an array of bioactive therapeutics in a spatiotemporal manner to mimic their natural release in vivo.
Collapse
Affiliation(s)
- Eimear J. Wallace
- Anatomy and Regenerative Medicine Institute (REMEDI)School of MedicineCollege of Medicine Nursing and Health SciencesUniversity of GalwayGalwayH91 W2TYIreland
- Explora‐Bioscience SrlG. Peroni 386Rome00131Italy
| | - Joanne O'Dwyer
- Pharmacology and TherapeuticsSchool of MedicineCollege of Medicine Nursing and Health SciencesUniversity of GalwayGalwayH91 W2TYIreland
| | - Eimear B. Dolan
- Anatomy and Regenerative Medicine Institute (REMEDI)School of MedicineCollege of Medicine Nursing and Health SciencesUniversity of GalwayGalwayH91 W2TYIreland
- CÚRAMSFI Research Centre for Medical DevicesUniversity of GalwayGalwayH91 W2TYIreland
- Biomedical EngineeringSchool of EngineeringUniversity of GalwayGalwayH91 HX31Ireland
| | - Liam P. Burke
- Anatomy and Regenerative Medicine Institute (REMEDI)School of MedicineCollege of Medicine Nursing and Health SciencesUniversity of GalwayGalwayH91 W2TYIreland
- Antimicrobial Resistance and Microbial Ecology GroupSchool of Medicine, College of Medicine Nursing and Health SciencesUniversity of GalwayGalwayH91 DK59Ireland
- Centre for One HealthRyan InstituteUniversity of GalwayGalwayH91 DK59Ireland
| | - Robert Wylie
- Anatomy and Regenerative Medicine Institute (REMEDI)School of MedicineCollege of Medicine Nursing and Health SciencesUniversity of GalwayGalwayH91 W2TYIreland
| | | | | | | | | | - Simona Serini
- Department of Translational Medicine and SurgerySection of General Pathology, Faculty of Medicine and SurgeryUniversità Cattolica del Sacro CuoreLargo F. VitoRome1‐00168Italy
| | - Gabriella Calviello
- Department of Translational Medicine and SurgerySection of General Pathology, Faculty of Medicine and SurgeryUniversità Cattolica del Sacro CuoreLargo F. VitoRome1‐00168Italy
| | - Ellen T. Roche
- Institute for Medical Engineering and ScienceMassachusetts Institute of TechnologyCambridgeMA 01239USA
- Harvard‐MIT Program in Health Sciences and TechnologyCambridgeMA02139USA
- Department of Mechanical EngineeringMassachusetts Institute of TechnologyCambridgeMA02139USA
| | - Tapas Mitra
- Anatomy and Regenerative Medicine Institute (REMEDI)School of MedicineCollege of Medicine Nursing and Health SciencesUniversity of GalwayGalwayH91 W2TYIreland
- CÚRAMSFI Research Centre for Medical DevicesUniversity of GalwayGalwayH91 W2TYIreland
| | - Garry P. Duffy
- Anatomy and Regenerative Medicine Institute (REMEDI)School of MedicineCollege of Medicine Nursing and Health SciencesUniversity of GalwayGalwayH91 W2TYIreland
- CÚRAMSFI Research Centre for Medical DevicesUniversity of GalwayGalwayH91 W2TYIreland
- SFI Centre for Advanced Materials and BioEngineering Research Centre (AMBER)Trinity College DublinDublinD02 W9K7Ireland
| |
Collapse
|
3
|
Gupta VK, Vaishnavi VV, Arrieta-Ortiz ML, Abhirami P, Jyothsna K, Jeyasankar S, Raghunathan V, Baliga NS, Agarwal R. 3D Hydrogel Culture System Recapitulates Key Tuberculosis Phenotypes and Demonstrates Pyrazinamide Efficacy. Adv Healthc Mater 2025; 14:e2304299. [PMID: 38655817 PMCID: PMC7616495 DOI: 10.1002/adhm.202304299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 03/29/2024] [Indexed: 04/26/2024]
Abstract
The mortality caused by tuberculosis (TB) infections is a global concern, and there is a need to improve understanding of the disease. Current in vitro infection models to study the disease have limitations such as short investigation durations and divergent transcriptional signatures. This study aims to overcome these limitations by developing a 3D collagen culture system that mimics the biomechanical and extracellular matrix (ECM) of lung microenvironment (collagen fibers, stiffness comparable to in vivo conditions) as the infection primarily manifests in the lungs. The system incorporates Mycobacterium tuberculosis (Mtb) infected human THP-1 or primary monocytes/macrophages. Dual RNA sequencing reveals higher mammalian gene expression similarity with patient samples than 2D macrophage infections. Similarly, bacterial gene expression more accurately recapitulates in vivo gene expression patterns compared to bacteria in 2D infection models. Key phenotypes observed in humans, such as foamy macrophages and mycobacterial cords, are reproduced in the model. This biomaterial system overcomes challenges associated with traditional platforms by modulating immune cells and closely mimicking in vivo infection conditions, including showing efficacy with clinically relevant concentrations of anti-TB drug pyrazinamide, not seen in any other in vitro infection model, making it reliable and readily adoptable for tuberculosis studies and drug screening.
Collapse
Affiliation(s)
- Vishal K. Gupta
- Department of Bioengineering, Indian Institute of Science, Bengaluru, India
| | | | | | - P.S. Abhirami
- Department of Bioengineering, Indian Institute of Science, Bengaluru, India
| | - K.M. Jyothsna
- Department of Electrical Communication Engineering, Indian Institute of Science, Bengaluru, India
| | | | - Varun Raghunathan
- Department of Electrical Communication Engineering, Indian Institute of Science, Bengaluru, India
| | | | - Rachit Agarwal
- Department of Bioengineering, Indian Institute of Science, Bengaluru, India
| |
Collapse
|
4
|
Lin CH, Srioudom JR, Sun W, Xing M, Yan S, Yu L, Yang J. The use of hydrogel microspheres as cell and drug delivery carriers for bone, cartilage, and soft tissue regeneration. BIOMATERIALS TRANSLATIONAL 2024; 5:236-256. [PMID: 39734701 PMCID: PMC11681182 DOI: 10.12336/biomatertransl.2024.03.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 08/22/2024] [Accepted: 09/12/2024] [Indexed: 12/31/2024]
Abstract
Bone, cartilage, and soft tissue regeneration is a complex process involving many cellular activities across various cell types. Autografts remain the "gold standard" for the regeneration of these tissues. However, the use of autografts is associated with many disadvantages, including donor scarcity, the requirement of multiple surgeries, and the risk of infection. The development of tissue engineering techniques opens new avenues for enhanced tissue regeneration. Nowadays, the expectations of tissue engineering scaffolds have gone beyond merely providing physical support for cell attachment. Ideal scaffolds should also provide biological cues to actively boost tissue regeneration. As a new type of injectable biomaterial, hydrogel microspheres have been increasingly recognised as promising therapeutic carriers for the local delivery of cells and drugs to enhance tissue regeneration. Compared to traditional tissue engineering scaffolds and bulk hydrogel, hydrogel microspheres possess distinct advantages, including less invasive delivery, larger surface area, higher transparency for visualisation, and greater flexibility for functionalisation. Herein, we review the materials characteristics of hydrogel microspheres and compare their fabrication approaches, including microfluidics, batch emulsion, electrohydrodynamic spraying, lithography, and mechanical fragmentation. Additionally, based on the different requirements for bone, cartilage, nerve, skin, and muscle tissue regeneration, we summarize the applications of hydrogel microspheres as cell and drug delivery carriers for the regeneration of these tissues. Overall, hydrogel microspheres are regarded as effective therapeutic delivery carriers to enhance tissue regeneration in regenerative medicine. However, significant effort is required before hydrogel microspheres become widely accepted as commercial products for clinical use.
Collapse
Affiliation(s)
- Chung-Hsun Lin
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA, USA
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Jesse R. Srioudom
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA, USA
| | - Wei Sun
- Leicester International Institute, Dalian University of Technology, Panjin, Liaoning Province, China
| | - Malcolm Xing
- Department of Mechanical Engineering, University of Manitoba, Winnipeg, Canada
| | - Su Yan
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA, USA
| | - Le Yu
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA, USA
- Division of Biological and Biomedical Systems, University of Missouri-Kansas City, Kansas City, MO, USA
| | - Jian Yang
- Biomedical Engineering Program, Westlake University, Hangzhou, Zhejiang Province, China
- Research Centre for Industries of the Future, Westlake University, Hangzhou, Zhejiang Province, China
| |
Collapse
|
5
|
Mora-Boza A, Ahmedin Z, García AJ. Controlled release of therapeutic antibody using hydrolytically degradable microgels. J Biomed Mater Res A 2024; 112:1265-1275. [PMID: 37927169 PMCID: PMC11069594 DOI: 10.1002/jbm.a.37637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 10/17/2023] [Accepted: 10/19/2023] [Indexed: 11/07/2023]
Abstract
Monoclonal antibodies have gained significant interest as potential therapeutics for treating various diseases. However, these therapies are not always effective due to poor treatment compliance associated with multiple administrations and drug resistance. Thus, there is a growing interest in developing advanced monoclonal antibody delivery systems that can customize pharmacokinetics to enhance therapeutic outcomes. This work aimed to engineer hydrolytic 4-arm PEG maleimide (PEG-4MAL) microgels for the controlled delivery of therapeutic antibodies, specifically anti-angiogenic bevacizumab, to overcome the limitations of current monoclonal antibody therapies. Through a PEGylation reaction with a thiol-terminated PEG linker, the antibody was covalently conjugated to the macromer backbone before microgel synthesis. The PEGylation reaction was simple, effective, and did not affect antibody bioactivity. Antibody release kinetics was tuned by changing the concentration of the hydrolytic linker (0-2 mM) and/or PEG-4MAL:protein molar ratio (1000:1, 2000:1, and 5000:1) in the macromer precursor solution during microgel fabrication. The bioactivity of the released antibody was assessed on human umbilical endothelial vascular cells (HUVEC), demonstrating that extracts from hydrolytic microgels reduced cell proliferation over time. Collectively, this study demonstrates the development of highly tunable delivery platform based on degradable PEG-4MAL microgels that can be adapted for therapeutic antibody-controlled release.
Collapse
Affiliation(s)
- Ana Mora-Boza
- Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, Georgia, USA
- Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, Georgia, USA
| | - Zakir Ahmedin
- Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, Georgia, USA
| | - Andrés J García
- Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, Georgia, USA
- Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, Georgia, USA
| |
Collapse
|
6
|
Bulondo F, Babensee JE. Optimization of Interleukin-10 incorporation for dendritic cells embedded in Poly(ethylene glycol) hydrogels. J Biomed Mater Res A 2024; 112:1317-1336. [PMID: 38562052 DOI: 10.1002/jbm.a.37714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 03/19/2024] [Accepted: 03/22/2024] [Indexed: 04/04/2024]
Abstract
Translational research in biomaterials and immunoengineering is leading to the development of novel advanced therapeutics to treat diseases such as cancer, autoimmunity, and viral infections. Dendritic cells (DCs) are at the center of these therapeutics given that they bridge innate and adaptive immunity. The biomaterial system developed herein uses a hydrogel carrier to deliver immunomodulatory DCs for amelioration of autoimmunity. This biomaterial vehicle is comprised of a poly (ethylene glycol)-4 arm maleimide (PEG-4MAL) hydrogels, conjugated with the immunosuppressive cytokine, interleukin-10, IL-10, and cross-linked with a collagenase-degradable peptide sequence for the injectable delivery of immunosuppressive DCs to an anatomical disease-relevant site of the cervical lymph nodes, for intended application to treat multiple sclerosis. The amount of IL-10 incorporated in the hydrogel was optimized to be 500 ng in vitro, based on immunological endpoints. At this concentration, DCs exhibited the best viability, most immunosuppressive phenotype, and protection against proinflammatory insult as compared with hydrogel-incorporated DCs with lower IL-10 loading amounts. Additionally, the effect of the degradability of the PEG-4MAL hydrogel on the release rate of incorporated IL-10 was assessed by varying the ratio of degradable peptides: VPM (degradable) and DTT (nondegradable) and measuring the IL-10 release rates. This IL-10-conjugated hydrogel delivery system for immunosuppressive DCs is set to be assessed for in vivo functionality as the immunosuppressive cytokine provides a tolerogenic environment that keeps DCs in their immature phenotype, which consequently enhances cell viability and optimizes the system's immunomodulatory functionality.
Collapse
Affiliation(s)
- Fredrick Bulondo
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, Georgia, USA
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia, USA
- Department of Biomedical Sciences and Engineering, Mbarara University of Science and Technology, Mbarara, Uganda
| | - Julia E Babensee
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, Georgia, USA
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia, USA
| |
Collapse
|
7
|
Quizon MJ, Deppen JN, Barber GF, Kalelkar PP, Coronel MM, Levit RD, García AJ. VEGF-delivering PEG hydrogels promote vascularization in the porcine subcutaneous space. J Biomed Mater Res A 2024; 112:866-880. [PMID: 38189109 PMCID: PMC10984793 DOI: 10.1002/jbm.a.37666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 12/21/2023] [Accepted: 12/24/2023] [Indexed: 01/09/2024]
Abstract
For cell therapies, the subcutaneous space is an attractive transplant site due to its large surface area and accessibility for implantation, monitoring, biopsy, and retrieval. However, its poor vascularization has catalyzed research to induce blood vessel formation within the site to enhance cell revascularization and survival. Most studies focus on the subcutaneous space of rodents, which does not recapitulate important anatomical features and vascularization responses of humans. Herein, we evaluate biomaterial-driven vascularization in the porcine subcutaneous space. Additionally, we report the first use of cost-effective fluorescent microspheres to quantify perfusion in the porcine subcutaneous space. We investigate the vascularization-inducing efficacy of vascular endothelial growth factor (VEGF)-delivering synthetic hydrogels based on 4-arm poly(ethylene) glycol macromers with terminal maleimides (PEG-4MAL). We compare three groups: a non-degradable hydrogel with a VEGF-releasing PEG-4MAL gel coating (Core+VEGF gel); an uncoated, non-degradable hydrogel (Core-only); and naïve tissue. After 2 weeks, Core+VEGF gel has significantly higher tissue perfusion, blood vessel area, blood vessel density, and number of vessels compared to both Core-only and naïve tissue. Furthermore, healthy vital signs during surgery and post-procedure metrics demonstrate the safety of hydrogel delivery. We demonstrate that VEGF-delivering synthetic hydrogels induce robust vascularization and perfusion in the porcine subcutaneous space.
Collapse
Affiliation(s)
- Michelle J. Quizon
- Woodruff School of Mechanical Engineering and Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, 315 Ferst Dr. NW, Atlanta, GA 30332, USA
| | - Juline N. Deppen
- Division of Cardiology, Emory University School of Medicine, 1440 Clifton Rd, Atlanta, GA 30322, USA
| | - Graham F. Barber
- Woodruff School of Mechanical Engineering and Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, 315 Ferst Dr. NW, Atlanta, GA 30332, USA
| | - Pranav P. Kalelkar
- Woodruff School of Mechanical Engineering and Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, 315 Ferst Dr. NW, Atlanta, GA 30332, USA
| | - María M. Coronel
- Woodruff School of Mechanical Engineering and Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, 315 Ferst Dr. NW, Atlanta, GA 30332, USA
| | - Rebecca D. Levit
- Division of Cardiology, Emory University School of Medicine, 1440 Clifton Rd, Atlanta, GA 30322, USA
| | - Andrés J. García
- Woodruff School of Mechanical Engineering and Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, 315 Ferst Dr. NW, Atlanta, GA 30332, USA
| |
Collapse
|
8
|
Slaby EM, Hansen N, Sharma R, Pirrotte P, Weaver JD. Engineered 3D Hydrogel Matrices to Modulate Trophoblast Stem Cell-Derived Placental Organoid Phenotype. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.13.594007. [PMID: 38798435 PMCID: PMC11118344 DOI: 10.1101/2024.05.13.594007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Placental organoid models are a promising platform to study human placental development and function. Organoid systems typically use naturally derived hydrogel extracellular matrices (ECM), resulting in batch-to-batch variability that limits experimental reproducibility. As an alternative, synthetic ECM-mimicking hydrogel matrices offer greater consistency and control over environmental cues. Here, we generated trophoblast stem cell-derived placental organoids using poly(ethylene glycol) (PEG) hydrogels with tunable degradability and placenta-derived ECM cues to evaluate trophoblast differentiation relative to Matrigel and two-dimensional (2D) culture controls. Our data demonstrate that PEG hydrogels support trophoblast viability and metabolic function comparable to gold standard Matrigel. Additionally, phenotypic characterization via proteomic analysis revealed that PEG and Matrigel matrices drive syncytiotrophoblast and extravillous trophoblast-dominant placental organoid phenotypes, respectively. Further, three-dimensional (3D) environments promoted greater integrin expression and ECM production than 2D culture. This study demonstrates that engineered 3D culture environments can be used to reliably generate placental organoids and guide trophoblast differentiation.
Collapse
|
9
|
Abrahamsson A, Boroojeni FR, Naeimipour S, Reustle N, Selegård R, Aili D, Dabrosin C. Increased matrix stiffness enhances pro-tumorigenic traits in a physiologically relevant breast tissue- monocyte 3D model. Acta Biomater 2024; 178:160-169. [PMID: 38382828 DOI: 10.1016/j.actbio.2024.02.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 02/06/2024] [Accepted: 02/13/2024] [Indexed: 02/23/2024]
Abstract
High mammographic density, associated with increased tissue stiffness, is a strong risk factor for breast cancer per se. In postmenopausal women there is no differences in the occurrence of ductal carcinoma in situ (DCIS) depending on breast density. Preliminary data suggest that dense breast tissue is associated with a pro-inflammatory microenvironment including infiltrating monocytes. However, the underlying mechanism(s) remains largely unknown. A major roadblock to understanding this risk factor is the lack of relevant in vitro models. A biologically relevant 3D model with tunable stiffness was developed by cross-linking hyaluronic acid. Breast cancer cells were cultured with and without freshly isolated human monocytes. In a unique clinical setting, extracellular proteins were sampled using microdialysis in situ from women with various breast densities. We show that tissue stiffness resembling high mammographic density increases the attachment of monocytes to the cancer cells, increase the expression of adhesion molecules and epithelia-mesenchymal-transition proteins in estrogen receptor (ER) positive breast cancer. Increased tissue stiffness results in increased secretion of similar pro-tumorigenic proteins as those found in human dense breast tissue including inflammatory cytokines, proteases, and growth factors. ER negative breast cancer cells were mostly unaffected suggesting that diverse cancer cell phenotypes may respond differently to tissue stiffness. We introduce a biological relevant model with tunable stiffness that resembles the densities found in normal breast tissue in women. The model will be key for further mechanistic studies. Additionally, our data revealed several pro-tumorigenic pathways that may be exploited for prevention and therapy against breast cancer. STATEMENT OF SIGNIFICANCE: Women with mammographic high-density breasts have a 4-6-fold higher risk of breast cancer than low-density breasts. Biological mechanisms behind this increase are not fully understood and no preventive therapeutics are available. One major reason being a lack of suitable experimental models. Having such models available would greatly enhance the discovery of relevant targets for breast cancer prevention. We present a biologically relevant 3D-model for studies of human dense breasts, providing a platform for investigating both biophysical and biochemical properties that may affect cancer progression. This model will have a major scientific impact on studies for identification of novel targets for breast cancer prevention.
Collapse
Affiliation(s)
- Annelie Abrahamsson
- Department of Oncology and Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Fatemeh Rasti Boroojeni
- Laboratory of Molecular Materials, Division of Biophysics and Bioengineering, Department of Physics, Chemistry and Biology, Linköping University, 581 83 Linköping, Sweden
| | - Sajjad Naeimipour
- Laboratory of Molecular Materials, Division of Biophysics and Bioengineering, Department of Physics, Chemistry and Biology, Linköping University, 581 83 Linköping, Sweden
| | - Nina Reustle
- Laboratory of Molecular Materials, Division of Biophysics and Bioengineering, Department of Physics, Chemistry and Biology, Linköping University, 581 83 Linköping, Sweden
| | - Robert Selegård
- Laboratory of Molecular Materials, Division of Biophysics and Bioengineering, Department of Physics, Chemistry and Biology, Linköping University, 581 83 Linköping, Sweden
| | - Daniel Aili
- Laboratory of Molecular Materials, Division of Biophysics and Bioengineering, Department of Physics, Chemistry and Biology, Linköping University, 581 83 Linköping, Sweden.
| | - Charlotta Dabrosin
- Department of Oncology and Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden.
| |
Collapse
|
10
|
D’Elia A, Jones OL, Canziani G, Sarkar B, Chaiken I, Rodell CB. Injectable Granular Hydrogels Enable Avidity-Controlled Biotherapeutic Delivery. ACS Biomater Sci Eng 2024; 10:1577-1588. [PMID: 38357739 PMCID: PMC10934254 DOI: 10.1021/acsbiomaterials.3c01906] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 01/29/2024] [Accepted: 01/31/2024] [Indexed: 02/16/2024]
Abstract
Protein therapeutics represent a rapidly growing class of pharmaceutical agents that hold great promise for the treatment of various diseases such as cancer and autoimmune dysfunction. Conventional systemic delivery approaches, however, result in off-target drug exposure and a short therapeutic half-life, highlighting the need for more localized and controlled delivery. We have developed an affinity-based protein delivery system that uses guest-host complexation between β-cyclodextrin (CD, host) and adamantane (Ad, guest) to enable sustained localized biomolecule presentation. Hydrogels were formed by the copolymerization of methacrylated CD and methacrylated dextran. Extrusion fragmentation of bulk hydrogels yielded shear-thinning and self-healing granular hydrogels (particle diameter = 32.4 ± 16.4 μm) suitable for minimally invasive delivery and with a high host capacity for the retention of guest-modified proteins. Bovine serum albumin (BSA) was controllably conjugated to Ad via EDC chemistry without affecting the affinity of the Ad moiety for CD (KD = 12.0 ± 1.81 μM; isothermal titration calorimetry). The avidity of Ad-BSA conjugates was directly tunable through the number of guest groups attached, resulting in a fourfold increase in the complex half-life (t1/2 = 5.07 ± 1.23 h, surface plasmon resonance) that enabled a fivefold reduction in protein release at 28 days. Furthermore, we demonstrated that the conjugation of Ad to immunomodulatory cytokines (IL-4, IL-10, and IFNγ) did not detrimentally affect cytokine bioactivity and enabled their sustained release. Our strategy of avidity-controlled delivery of protein-based therapeutics is a promising approach for the sustained local presentation of protein therapeutics and can be applied to numerous biomedical applications.
Collapse
Affiliation(s)
- Arielle
M. D’Elia
- School
of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, Pennsylvania 19104, United States
| | - Olivia L. Jones
- School
of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, Pennsylvania 19104, United States
| | - Gabriela Canziani
- Department
of Biochemistry and Molecular Biology, Drexel
University College of Medicine, Philadelphia, Pennsylvania 19102, United States
| | - Biplab Sarkar
- School
of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, Pennsylvania 19104, United States
| | - Irwin Chaiken
- Department
of Biochemistry and Molecular Biology, Drexel
University College of Medicine, Philadelphia, Pennsylvania 19102, United States
| | - Christopher B. Rodell
- School
of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, Pennsylvania 19104, United States
| |
Collapse
|
11
|
Palmese LL, LeValley PJ, Pradhan L, Parsons AL, Oakey JS, Abraham M, D'Addio SM, Kloxin AM, Liang Y, Kiick KL. Injectable liposome-containing click hydrogel microparticles for release of macromolecular cargos. SOFT MATTER 2024; 20:1736-1745. [PMID: 38288734 PMCID: PMC10880143 DOI: 10.1039/d3sm01009k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Accepted: 12/16/2023] [Indexed: 02/22/2024]
Abstract
Hydrogel microparticles ranging from 0.1-100 μm, referred to as microgels, are attractive for biological applications afforded by their injectability and modularity, which allows facile delivery of mixed populations for tailored combinations of therapeutics. Significant efforts have been made to broaden methods for microgel production including via the materials and chemistries by which they are made. Via droplet-based-microfluidics we have established a method for producing click poly-(ethylene)-glycol (PEG)-based microgels with or without chemically crosslinked liposomes (lipo-microgels) through the Michael-type addition reaction between thiol and either vinyl-sulfone or maleimide groups. Unifom spherical microgels and lipo-microgels were generated with sizes of 74 ± 16 μm and 82 ± 25 μm, respectively, suggesting injectability that was further supported by rheological analyses. Super-resolution confocal microscopy was used to further verify the presence of liposomes within the lipo-microgels and determine their distribution. Atomic force microscopy (AFM) was conducted to compare the mechanical properties and network architecture of bulk hydrogels, microgels, and lipo-microgels. Further, encapsulation and release of model cargo (FITC-Dextran 5 kDa) and protein (equine myoglobin) showed sustained release for up to 3 weeks and retention of protein composition and secondary structure, indicating their ability to both protect and release cargos of interest.
Collapse
Affiliation(s)
- Luisa L Palmese
- Materials Science and Engineering, University of Delaware, Newark, DE, USA.
| | - Paige J LeValley
- Chemical and Biomolecular Engineering, University of Delaware, Newark, DE, USA
| | - Lina Pradhan
- Chemical and Biomolecular Engineering, University of Delaware, Newark, DE, USA
| | - Amanda L Parsons
- Chemical and Biomedical Engineering, University of Wyoming, Laramie, WY, USA
| | - John S Oakey
- Chemical and Biomedical Engineering, University of Wyoming, Laramie, WY, USA
| | - Mathew Abraham
- Translational Imaging, Merck & Co., Inc., West Point, PA, USA
| | - Suzanne M D'Addio
- Discovery Pharmaceutical Sciences, Merck & Co., Inc., West Point, PA, USA.
| | - April M Kloxin
- Materials Science and Engineering, University of Delaware, Newark, DE, USA.
- Chemical and Biomolecular Engineering, University of Delaware, Newark, DE, USA
| | - Yingkai Liang
- Discovery Pharmaceutical Sciences, Merck & Co., Inc., West Point, PA, USA.
| | - Kristi L Kiick
- Materials Science and Engineering, University of Delaware, Newark, DE, USA.
- Biomedical Engineering, University of Delaware, Newark, DE, USA
| |
Collapse
|
12
|
Nguyen TTK, Pham KY, Yook S. Engineered therapeutic proteins for sustained-release drug delivery systems. Acta Biomater 2023; 171:131-154. [PMID: 37717712 DOI: 10.1016/j.actbio.2023.09.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 09/04/2023] [Accepted: 09/12/2023] [Indexed: 09/19/2023]
Abstract
Proteins play a vital role in diverse biological processes in the human body, and protein therapeutics have been applied to treat different diseases such as cancers, genetic disorders, autoimmunity, and inflammation. Protein therapeutics have demonstrated their advantages, such as specific pharmaceutical effects, low toxicity, and strong solubility. However, several disadvantages arise in clinical applications, including short half-life, immunogenicity, and low permeation, leading to reduced drug effectiveness. The structure of protein therapeutics can be modified to increase molecular size, leading to prolonged stability and increased plasma half-life. Notably, the controlled-release delivery systems for the sustained release of protein drugs and preserving the stability of cargo proteins are envisioned as a potential approach to overcome these challenges. In this review, we summarize recent research progress related to structural modifications (PEGylation, glycosylation, poly amino acid modification, and molecular biology-based strategies) and promising long-term delivery systems, such as polymer-based systems (injectable gel/implants, microparticles, nanoparticles, micro/nanogels, functional polymers), lipid-based systems (liposomes, solid lipid nanoparticles, nanostructured lipid carriers), and inorganic nanoparticles exploited for protein therapeutics. STATEMENT OF SIGNIFICANCE: In this review, we highlight recent advances concerning modifying proteins directly to enhance their stability and functionality and discuss state-of-the-art methods for the delivery and controlled long-term release of active protein therapeutics to their target site. In terms of drug modifications, four widely used strategies, including PEGylation, poly amino acid modification, glycosylation, and genetic, are discussed. As for drug delivery systems, we emphasize recent progress relating to polymer-based systems, lipid-based systems developed, and inorganic nanoparticles for protein sustained-release delivery. This review points out the areas requiring focused research attention before the full potential of protein therapeutics for human health and disease can be realized.
Collapse
Affiliation(s)
- Thoa Thi Kim Nguyen
- College of Pharmacy, Keimyung University, 1095 Dalgubeol-daero, Dalseo-Gu, Daegu 42601, Republic of Korea
| | - Khang-Yen Pham
- College of Pharmacy, Keimyung University, 1095 Dalgubeol-daero, Dalseo-Gu, Daegu 42601, Republic of Korea.
| | - Simmyung Yook
- College of Pharmacy, Keimyung University, 1095 Dalgubeol-daero, Dalseo-Gu, Daegu 42601, Republic of Korea; School of Pharmacy, Sungkyunkwan University, Suwon 16419, Republic of Korea; Department of Biopharmaceutical Convergence, Sungkyunkwan University, Suwon 16419, Republic of Korea.
| |
Collapse
|
13
|
Mora-Boza A, Ghebrezadik SG, Leisen JE, García AJ. Rapid and Facile Light-Based Approach to Fabricate Protease-Degradable Poly(ethylene glycol)-norbornene Microgels for Cell Encapsulation. Adv Healthc Mater 2023; 12:e2300942. [PMID: 37235850 PMCID: PMC10592588 DOI: 10.1002/adhm.202300942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 05/15/2023] [Indexed: 05/28/2023]
Abstract
Thiol-norbornene photoclickable poly (ethylene glycol) (PEG)-based (PEG-NB) hydrogels are attractive biomaterials for cell encapsulation, drug delivery, and regenerative medicine applications. Although many crosslinking strategies and chemistries have been developed for PEG-NB bulk hydrogels, fabrication approaches of PEG-NB microgels have not been extensively explored. Here, a fabrication strategy for 4-arm amide-linked PEG-NB (PEG-4aNB) microgels using flow-focusing microfluidics for human mesenchymal stem/stromal cell (hMSCs) encapsulation is presented. PEG-4aNB photochemistry allows high-throughput, ultrafast generation, and cost-effective synthesis of monodispersed microgels (diameter 340 ± 18, 380 ± 24, and 420 ± 15 µm, for 6, 8, and 10 wt% of PEG-4aNB, respectively) using an in situ crosslinking methodology in a microfluidic device. PEG-4aNB microgels show in vitro degradability due to the incorporation of a protease-degradable peptide during photocrosslinking and encapsulated cells show excellent viability and metabolic activity for at least 13 days of culture. Furthermore, the secretory profile (i.e., MMP-13, ICAM-1, PD-L1, CXCL9, CCL3/MIP-1, IL-6, IL-12, IL-17E, TNF-α, CCL2/MCP-1) of encapsulated hMSCs shows increased expression in response to IFN-γ stimulation. Collectively, this work shows a versatile and facile approach for the fabrication of protease-degradable PEG-4aNB microgels for cell encapsulation.
Collapse
Affiliation(s)
- Ana Mora-Boza
- Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, 30332-0363, USA
- Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, 30332-0363, USA
- CÚRAM, University of Galway, Galway, H91 W2TY, Ireland
| | - Saron G Ghebrezadik
- Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, 30332-0363, USA
- Agnes Scott College, Decatur, GA, 30030, USA
| | - Johannes E Leisen
- Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, 30332-0363, USA
| | - Andrés J García
- Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, 30332-0363, USA
- Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, 30332-0363, USA
| |
Collapse
|
14
|
Sahiner M, Yilmaz AS, Ayyala RS, Sahiner N. Carboxymethyl Chitosan Microgels for Sustained Delivery of Vancomycin and Long-Lasting Antibacterial Effects. Gels 2023; 9:708. [PMID: 37754390 PMCID: PMC10529976 DOI: 10.3390/gels9090708] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 08/20/2023] [Accepted: 08/28/2023] [Indexed: 09/28/2023] Open
Abstract
Carboxymethyl chitosan (CMCh) is a unique polysaccharide with functional groups that can develop positive and negative charges due to the abundant numbers of amine and carboxylic acid groups. CMCh is widely used in different areas due to its excellent biocompatibility, biodegradability, water solubility, and chelating ability. CMCh microgels were synthesized in a microemulsion environment using divinyl sulfone (DVS) as a crosslinking agent. CMCh microgel with tailored size and zeta potential values were obtained in a single stem by crosslinking CMCh in a water-in-oil environment. The spherical microgel structure is confirmed by SEM analysis. The sizes of CMCh microgels varied from one micrometer to tens of micrometers. The isoelectric point of CMCh microgels was determined as pH 4.4. Biocompatibility of CMCh microgels was verified on L929 fibroblasts with 96.5 ± 1.5% cell viability at 1 mg/mL concentration. The drug-carrying abilities of CMCh microgels were evaluated by loading Vancomycin (Van) antibiotic as a model drug. Furthermore, the antibacterial activity efficiency of Van-loaded CMCh microgels (Van@CMCh) was investigated. The MIC values of the released drug from Van@CMCh microgels were found to be 68.6 and 7.95 µg/mL against E. coli and S. aureus, respectively, at 24 h contact time. Disk diffusion tests confirmed that Van@CMCh microgels, especially for Gram-positive (S. aureus) bacteria, revealed long-lasting inhibitory effects on bacteria growth up to 72 h.
Collapse
Affiliation(s)
- Mehtap Sahiner
- Department of Bioengineering, Faculty of Engineering, Canakkale, Onsekiz Mart University Terzioglu Campus, Canakkale 17100, Turkey;
- Department of Chemical, Biological and Materials Engineering, University of South Florida, Tampa, FL 33620, USA;
| | - Aynur S. Yilmaz
- Department of Chemical, Biological and Materials Engineering, University of South Florida, Tampa, FL 33620, USA;
- Department of Chemistry, Faculty of Sciences, and Nanoscience and Technology Research and Application Center (NANORAC), Canakkale Onsekiz Mart University Terzioglu Campus, Canakkale 17100, Turkey
| | - Ramesh S. Ayyala
- Department of Ophthalmology, Morsani College of Medicine, University of South Florida Eye Institute, 12901 Bruce B Down Blvd., MDC 21, Tampa, FL 33612, USA;
| | - Nurettin Sahiner
- Department of Chemical, Biological and Materials Engineering, University of South Florida, Tampa, FL 33620, USA;
- Department of Chemistry, Faculty of Sciences, and Nanoscience and Technology Research and Application Center (NANORAC), Canakkale Onsekiz Mart University Terzioglu Campus, Canakkale 17100, Turkey
- Department of Ophthalmology, Morsani College of Medicine, University of South Florida Eye Institute, 12901 Bruce B Down Blvd., MDC 21, Tampa, FL 33612, USA;
| |
Collapse
|
15
|
Luo Z, Zhang H, Chen R, Li H, Cheng F, Zhang L, Liu J, Kong T, Zhang Y, Wang H. Digital light processing 3D printing for microfluidic chips with enhanced resolution via dosing- and zoning-controlled vat photopolymerization. MICROSYSTEMS & NANOENGINEERING 2023; 9:103. [PMID: 37593440 PMCID: PMC10427687 DOI: 10.1038/s41378-023-00542-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Revised: 04/07/2023] [Accepted: 04/10/2023] [Indexed: 08/19/2023]
Abstract
Conventional manufacturing techniques to fabricate microfluidic chips, such as soft lithography and hot embossing process, have limitations that include difficulty in preparing multiple-layered structures, cost- and labor-consuming fabrication process, and low productivity. Digital light processing (DLP) technology has recently emerged as a cost-efficient microfabrication approach for the 3D printing of microfluidic chips; however, the fabrication resolution for microchannels is still limited to sub-100 microns at best. Here, we developed an innovative DLP printing strategy for high resolution and scalable microchannel fabrication by dosing- and zoning-controlled vat photopolymerization (DZC-VPP). Specifically, we proposed a modified mathematical model to precisely predict the accumulated UV irradiance for resin photopolymerization, thereby providing guidance for the fabrication of microchannels with enhanced resolution. By fine-tuning the printing parameters, including optical irradiance, exposure time, projection region, and step distance, we can precisely tailor the penetration irradiance stemming from the photopolymerization of the neighboring resin layers, thereby preventing channel blockage due to UV overexposure or compromised bonding stability owing to insufficient resin curing. Remarkably, this strategy can allow the preparation of microchannels with cross-sectional dimensions of 20 μm × 20 μm using a commercial printer with a pixel size of 10 μm × 10 μm; this is significantly higher resolution than previous reports. In addition, this method can enable the scalable and biocompatible fabrication of microfluidic drop-maker units that can be used for cell encapsulation. In general, the current DZC-VPP method can enable major advances in precise and scalable microchannel fabrication and represents a significant step forward for widespread applications of microfluidics-based techniques in biomedical fields.
Collapse
Affiliation(s)
- Zhiming Luo
- School of Biomedical Engineering, Shenzhen University Health Science Center, Shenzhen, 518000 P. R. China
| | - Haoyue Zhang
- State Key Laboratory of Fine Chemicals, Frontiers Science Center for Smart Materials Oriented Chemical Engineering, School of Bioengineering, Dalian University of Technology, Dalian, 116024 P. R. China
| | - Runze Chen
- State Key Laboratory of Fine Chemicals, Frontiers Science Center for Smart Materials Oriented Chemical Engineering, School of Bioengineering, Dalian University of Technology, Dalian, 116024 P. R. China
| | - Hanting Li
- State Key Laboratory of Fine Chemicals, Frontiers Science Center for Smart Materials Oriented Chemical Engineering, School of Bioengineering, Dalian University of Technology, Dalian, 116024 P. R. China
| | - Fang Cheng
- State Key Laboratory of Fine Chemicals, Frontiers Science Center for Smart Materials Oriented Chemical Engineering, School of Bioengineering, Dalian University of Technology, Dalian, 116024 P. R. China
| | - Lijun Zhang
- Third People’s Hospital of Dalian, Dalian Eye Hospital, Dalian, 116024 P. R. China
| | - Jia Liu
- Central Laboratory, The Second Affiliated Hospital of The, Chinese University of Hong Kong, Shenzhen, 518172 P. R. China
| | - Tiantian Kong
- School of Biomedical Engineering, Shenzhen University Health Science Center, Shenzhen, 518000 P. R. China
| | - Yang Zhang
- School of Biomedical Engineering, Shenzhen University Health Science Center, Shenzhen, 518000 P. R. China
| | - Huanan Wang
- School of Biomedical Engineering, Shenzhen University Health Science Center, Shenzhen, 518000 P. R. China
- State Key Laboratory of Fine Chemicals, Frontiers Science Center for Smart Materials Oriented Chemical Engineering, School of Bioengineering, Dalian University of Technology, Dalian, 116024 P. R. China
| |
Collapse
|
16
|
Nicosia A, Salamone M, Costa S, Ragusa MA, Ghersi G. Mimicking Molecular Pathways in the Design of Smart Hydrogels for the Design of Vascularized Engineered Tissues. Int J Mol Sci 2023; 24:12314. [PMID: 37569691 PMCID: PMC10418696 DOI: 10.3390/ijms241512314] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 07/21/2023] [Accepted: 07/31/2023] [Indexed: 08/13/2023] Open
Abstract
Biomaterials are pivotal in supporting and guiding vascularization for therapeutic applications. To design effective, bioactive biomaterials, understanding the cellular and molecular processes involved in angiogenesis and vasculogenesis is crucial. Biomaterial platforms can replicate the interactions between cells, the ECM, and the signaling molecules that trigger blood vessel formation. Hydrogels, with their soft and hydrated properties resembling natural tissues, are widely utilized; particularly synthetic hydrogels, known for their bio-inertness and precise control over cell-material interactions, are utilized. Naturally derived and synthetic hydrogel bases are tailored with specific mechanical properties, controlled for biodegradation, and enhanced for cell adhesion, appropriate biochemical signaling, and architectural features that facilitate the assembly and tubulogenesis of vascular cells. This comprehensive review showcases the latest advancements in hydrogel materials and innovative design modifications aimed at effectively guiding and supporting vascularization processes. Furthermore, by leveraging this knowledge, researchers can advance biomaterial design, which will enable precise support and guidance of vascularization processes and ultimately enhance tissue functionality and therapeutic outcomes.
Collapse
Affiliation(s)
- Aldo Nicosia
- Institute for Biomedical Research and Innovation-National Research Council (IRIB-CNR), Via Ugo la Malfa 153, 90146 Palermo, Italy;
| | - Monica Salamone
- Institute for Biomedical Research and Innovation-National Research Council (IRIB-CNR), Via Ugo la Malfa 153, 90146 Palermo, Italy;
| | - Salvatore Costa
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Viale delle Scienze, Ed. 16, 90128 Palermo, Italy; (S.C.); (M.A.R.); (G.G.)
| | - Maria Antonietta Ragusa
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Viale delle Scienze, Ed. 16, 90128 Palermo, Italy; (S.C.); (M.A.R.); (G.G.)
| | - Giulio Ghersi
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Viale delle Scienze, Ed. 16, 90128 Palermo, Italy; (S.C.); (M.A.R.); (G.G.)
| |
Collapse
|
17
|
Jia F, Gao Y, Wang H. Recent Advances in Drug Delivery System Fabricated by Microfluidics for Disease Therapy. Bioengineering (Basel) 2022; 9:625. [PMID: 36354536 PMCID: PMC9687342 DOI: 10.3390/bioengineering9110625] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 10/16/2022] [Accepted: 10/26/2022] [Indexed: 09/08/2024] Open
Abstract
Traditional drug therapy faces challenges such as drug distribution throughout the body, rapid degradation and excretion, and extensive adverse reactions. In contrast, micro/nanoparticles can controllably deliver drugs to target sites to improve drug efficacy. Unlike traditional large-scale synthetic systems, microfluidics allows manipulation of fluids at the microscale and shows great potential in drug delivery and precision medicine. Well-designed microfluidic devices have been used to fabricate multifunctional drug carriers using stimuli-responsive materials. In this review, we first introduce the selection of materials and processing techniques for microfluidic devices. Then, various well-designed microfluidic chips are shown for the fabrication of multifunctional micro/nanoparticles as drug delivery vehicles. Finally, we describe the interaction of drugs with lymphatic vessels that are neglected in organs-on-chips. Overall, the accelerated development of microfluidics holds great potential for the clinical translation of micro/nanoparticle drug delivery systems for disease treatment.
Collapse
Affiliation(s)
- Fuhao Jia
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China
- School of Nanoscience and Technology, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yanbing Gao
- Troop 96901 of the Chinese People’s Liberation Army, Beijing 100094, China
| | - Hai Wang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China
- School of Nanoscience and Technology, University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
18
|
Monferrer E, Dobre O, Trujillo S, González Oliva MA, Trubert-Paneli A, Acevedo-León D, Noguera R, Salmeron-Sanchez M. Vitronectin-based hydrogels recapitulate neuroblastoma growth conditions. Front Cell Dev Biol 2022; 10:988699. [PMID: 36425532 PMCID: PMC9679952 DOI: 10.3389/fcell.2022.988699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Accepted: 09/20/2022] [Indexed: 11/10/2022] Open
Abstract
The tumor microenvironment plays an important role in cancer development and the use of 3D in vitro systems that decouple different elements of this microenvironment is critical for the study of cancer progression. In neuroblastoma (NB), vitronectin (VN), an extracellular matrix protein, has been linked to poor prognosis and appears as a promising therapeutic target. Here, we developed hydrogels that incorporate VN into 3D polyethylene glycol (PEG) hydrogel networks to recapitulate the native NB microenvironment. The stiffness of the VN/PEG hydrogels was modulated to be comparable to the in vivo values reported for NB tissue samples. We used SK-N-BE (2) NB cells to demonstrate that PEGylated VN promotes cell adhesion as the native protein does. Furthermore, the PEGylation of VN allows its crosslinking into the hydrogel network, providing VN retention within the hydrogels that support viable cells in 3D. Confocal imaging and ELISA assays indicate that cells secrete VN also in the hydrogels and continue to reorganize their 3D environment. Overall, the 3D VN-based PEG hydrogels recapitulate the complexity of the native tumor extracellular matrix, showing that VN-cell interaction plays a key role in NB aggressiveness, and that VN could potentially be targeted in preclinical drug studies performed on the presented hydrogels.
Collapse
Affiliation(s)
- Ezequiel Monferrer
- Department of Pathology Medical School, University of Valencia-INCLIVA Biomedical Health Research Institute, Valencia, Spain
- Low Prevalence Tumors, Centro de Investigación Biomédica En Red de Cáncer (CIBERONC), Instituto de Salud Carlos III, Madrid, Spain
| | - Oana Dobre
- Centre for the Cellular Microenvironment, Advanced Research Centre, University of Glasgow, Glasgow, United Kingdom
| | - Sara Trujillo
- INM—Leibniz Institute for New Materials, Saarbrücken, Germany
| | | | - Alexandre Trubert-Paneli
- Centre for the Cellular Microenvironment, Advanced Research Centre, University of Glasgow, Glasgow, United Kingdom
| | - Delia Acevedo-León
- Clinical Analysis Service, Hospital Universitario Dr. Peset, Valencia, Spain
| | - Rosa Noguera
- Department of Pathology Medical School, University of Valencia-INCLIVA Biomedical Health Research Institute, Valencia, Spain
- Low Prevalence Tumors, Centro de Investigación Biomédica En Red de Cáncer (CIBERONC), Instituto de Salud Carlos III, Madrid, Spain
- *Correspondence: Rosa Noguera, ; Manuel Salmeron-Sanchez,
| | - Manuel Salmeron-Sanchez
- Centre for the Cellular Microenvironment, Advanced Research Centre, University of Glasgow, Glasgow, United Kingdom
- *Correspondence: Rosa Noguera, ; Manuel Salmeron-Sanchez,
| |
Collapse
|
19
|
Petaroudi M, Rodrigo‐Navarro A, Dobre O, Dalby MJ, Salmeron‐Sanchez M. Living Biomaterials to Engineer Hematopoietic Stem Cell Niches. Adv Healthc Mater 2022; 11:e2200964. [PMID: 35933595 PMCID: PMC11469072 DOI: 10.1002/adhm.202200964] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 08/01/2022] [Indexed: 01/28/2023]
Abstract
Living biointerfaces are a new class of biomaterials combining living cells and polymeric matrices that can act as biologically active and instructive materials that host and provide signals to surrounding cells. Here, living biomaterials based on Lactococcus lactis to control hematopoietic stem cells in 2D surfaces and 3D hydrogels are introduced. L. lactis is modified to express C-X-C motif chemokine ligand 12 (CXCL12), thrombopoietin (TPO), vascular cell adhesion protein 1 (VCAM1), and the 7th-10th type III domains of human plasma fibronectin (FN III7-10 ), in an attempt to mimic ex vivo the conditions of the human bone marrow. These results suggest that living biomaterials that incorporate bacteria expressing recombinant CXCL12, TPO, VCAM1, and FN in both 2D systems direct hematopoietic stem and progenitor cells (HSPCs)-bacteria interaction, and in 3D using hydrogels functionalized with full-length human plasma fibronectin allow for a notable expansion of the CD34+ /CD38- /CD90+ HSPC population compared to the initial population. These results provide a strong evidence based on data that suggest the possibility of using living materials based on genetically engineered bacteria for the ex-vivo expansion of HSPC with eventual practical clinical applications in HSPCs transplantation for hematological disorders.
Collapse
Affiliation(s)
- Michaela Petaroudi
- Centre for the Cellular MicroenvironmentUniversity of GlasgowGlasgowG12 8LTUK
| | | | - Oana Dobre
- Centre for the Cellular MicroenvironmentUniversity of GlasgowGlasgowG12 8LTUK
| | - Matthew J. Dalby
- Centre for the Cellular MicroenvironmentUniversity of GlasgowGlasgowG12 8LTUK
| | | |
Collapse
|
20
|
Coronel MM, Martin KE, Hunckler MD, Kalelkar P, Shah RM, García AJ. Hydrolytically Degradable Microgels with Tunable Mechanical Properties Modulate the Host Immune Response. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2022; 18:e2106896. [PMID: 35274457 PMCID: PMC10288386 DOI: 10.1002/smll.202106896] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 02/27/2022] [Indexed: 06/14/2023]
Abstract
Hydrogel microparticles (microgels) are an attractive approach for therapeutic delivery because of their modularity, injectability, and enhanced integration with the host tissue. Multiple microgel fabrication strategies and chemistries have been implemented, yet manipulation of microgel degradability and its effect on in vivo tissue responses remains underexplored. Here, the authors report a facile method to synthesize microgels crosslinked with ester-containing junctions to afford tunable degradation kinetics. Monodisperse microgels of maleimide-functionalized poly(ethylene-glycol) are generated using droplet microfluidics crosslinked with thiol-terminated, ester-containing molecules. Tunable mechanics are achievable based on the ratio of degradable to nondegradable crosslinkers in the continuous phase. Degradation in an aqueous medium leads to microgel deformation based on swelling and a decrease in elastic modulus. Furthermore, degradation byproducts are cytocompatible and do not cause monocytic cell activation under noninflammatory conditions. These injectable microgels possess time-dependent degradation on the order of weeks in vivo. Lastly, the evaluation of tissue responses in a subcutaneous dorsal pocket shows a dynamic type-1 like immune response to the synthetic microgels, driven by interferon gamma (IFN-γ ) expression, which can be moderated by tuning the degradation properties. Collectively, this study demonstrates the development of a hydrolytic microgel platform that can be adapted to desired host tissue immune responses.
Collapse
Affiliation(s)
- María M Coronel
- Woodruff School of Mechanical Engineering and Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA
| | - Karen E Martin
- Woodruff School of Mechanical Engineering and Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA
| | - Michael D Hunckler
- Woodruff School of Mechanical Engineering and Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA
| | - Pranav Kalelkar
- Woodruff School of Mechanical Engineering and Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA
| | - Rahul M Shah
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Andrés J García
- Woodruff School of Mechanical Engineering and Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA
| |
Collapse
|
21
|
Inflammation-triggered dual release of nitroxide radical and growth factor from heparin mimicking hydrogel-tissue composite as cardiovascular implants for anti-coagulation, endothelialization, anti-inflammation, and anti-calcification. Biomaterials 2022; 289:121761. [DOI: 10.1016/j.biomaterials.2022.121761] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 08/15/2022] [Accepted: 08/21/2022] [Indexed: 11/20/2022]
|
22
|
Wei Z, Wang S, Hirvonen J, Santos HA, Li W. Microfluidics Fabrication of Micrometer-Sized Hydrogels with Precisely Controlled Geometries for Biomedical Applications. Adv Healthc Mater 2022; 11:e2200846. [PMID: 35678152 PMCID: PMC11468590 DOI: 10.1002/adhm.202200846] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Indexed: 01/24/2023]
Abstract
Micrometer-sized hydrogels are cross-linked three-dimensional network matrices with high-water contents and dimensions ranging from several to hundreds of micrometers. Due to their excellent biocompatibility and capability to mimic physiological microenvironments in vivo, micrometer-sized hydrogels have attracted much attention in the biomedical engineering field. Their biological properties and applications are primarily influenced by their chemical compositions and geometries. However, inhomogeneous morphologies and uncontrollable geometries limit traditional micrometer-sized hydrogels obtained by bulk mixing. In contrast, microfluidic technology holds great potential for the fabrication of micrometer-sized hydrogels since their geometries, sizes, structures, compositions, and physicochemical properties can be precisely manipulated on demand based on the excellent control over fluids. Therefore, micrometer-sized hydrogels fabricated by microfluidic technology have been applied in the biomedical field, including drug encapsulation, cell encapsulation, and tissue engineering. This review introduces micrometer-sized hydrogels with various geometries synthesized by different microfluidic devices, highlighting their advantages in various biomedical applications over those from traditional approaches. Overall, emerging microfluidic technologies enrich the geometries and morphologies of hydrogels and accelerate translation for industrial production and clinical applications.
Collapse
Affiliation(s)
- Zhenyang Wei
- Drug Research ProgramDivision of Pharmaceutical Chemistry and TechnologyFaculty of PharmacyUniversity of HelsinkiHelsinki00014Finland
| | - Shiqi Wang
- Drug Research ProgramDivision of Pharmaceutical Chemistry and TechnologyFaculty of PharmacyUniversity of HelsinkiHelsinki00014Finland
| | - Jouni Hirvonen
- Drug Research ProgramDivision of Pharmaceutical Chemistry and TechnologyFaculty of PharmacyUniversity of HelsinkiHelsinki00014Finland
| | - Hélder A. Santos
- Drug Research ProgramDivision of Pharmaceutical Chemistry and TechnologyFaculty of PharmacyUniversity of HelsinkiHelsinki00014Finland
- Department of Biomedical EngineeringW.J. Kolff Institute for Biomedical Engineering and Materials ScienceUniversity Medical Center Groningen/University of GroningenAnt. Deusinglaan 1Groningen9713 AVThe Netherlands
| | - Wei Li
- Drug Research ProgramDivision of Pharmaceutical Chemistry and TechnologyFaculty of PharmacyUniversity of HelsinkiHelsinki00014Finland
| |
Collapse
|
23
|
Development of a nanocapsule-loaded hydrogel for drug delivery for intraperitoneal administration. Int J Pharm 2022; 622:121828. [PMID: 35595041 DOI: 10.1016/j.ijpharm.2022.121828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 05/01/2022] [Accepted: 05/09/2022] [Indexed: 11/22/2022]
Abstract
Intraperitoneal (IP) drug delivery of chemotherapeutic agents, administered through hyperthermal intraperitoneal chemotherapy (HIPEC) and pressurized intraperitoneal aerosolized chemotherapy (PIPAC), is effective for the treatment of peritoneal malignancies. However, these therapeutic interventions are cumbersome in terms of surgical practice and are often associated with the formation of peritoneal adhesions, due to the catheters inserted into the peritoneal cavity during these procedures. Hence, there is a need for the development of drug delivery systems that can be administered into the peritoneal cavity. In this study, we have developed a nanocapsule (NCs)-loaded hydrogel for drug delivery in the peritoneal cavity. The hydrogel has been developed using poly(ethylene glycol) (PEG) and thiol-maleimide chemistry. NCs-loaded hydrogels were characterized by rheology and their resistance to dilution and drug release were determined in vitro. Using IVIS® to measure individual organ and recovered gel fluorescence intensity, an in vivo imaging study was performed and demonstrated that NCs incorporated in the PEG gel were retained in the IP cavity for 24 h after IP administration. NCs-loaded PEG gels could find potential applications as biodegradable, drug delivery systems that could be implanted in the IP cavity, for example at a the tumour resection site to prevent recurrence of microscopic tumours.
Collapse
|
24
|
Qazi TH, Wu J, Muir VG, Weintraub S, Gullbrand SE, Lee D, Issadore D, Burdick JA. Anisotropic Rod-Shaped Particles Influence Injectable Granular Hydrogel Properties and Cell Invasion. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2022; 34:e2109194. [PMID: 34932833 PMCID: PMC8957565 DOI: 10.1002/adma.202109194] [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: 11/13/2021] [Revised: 12/15/2021] [Indexed: 05/08/2023]
Abstract
Granular hydrogels have emerged as a new class of injectable and porous biomaterials that improve integration with host tissue when compared to solid hydrogels. Granular hydrogels are typically prepared using spherical particles and this study considers whether particle shape (i.e., isotropic spheres vs anisotropic rods) influences granular hydrogel properties and cellular invasion. Simulations predict that anisotropic rods influence pore shape and interconnectivity, as well as bead transport through granular assemblies. Photo-cross-linkable norbornene-modified hyaluronic acid is used to produce spherical and rod-shaped particles using microfluidic droplet generators and formed into shear-thinning and self-healing granular hydrogels, with particle shape influencing mechanics and injectability. Rod-shaped particles form granular hydrogels that have anisotropic and interconnected pores, with pore size and number influenced by particle shape and degree of packing. Robust in vitro sprouting of endothelial cells from embedded cellular spheroids is observed with rod-shaped particles, including higher sprouting densities and sprout lengths when compared to hydrogels with spherical particles. Cell and vessel invasion into granular hydrogels when injected subcutaneously in vivo are significantly greater with rod-shaped particles, whereas a gradient of cellularity is observed with spherical particles. Overall, this work demonstrates potentially superior functional properties of granular hydrogels with rod-shaped particles for tissue repair.
Collapse
Affiliation(s)
- Taimoor H Qazi
- Department of Bioengineering, School of Engineering and Applied Sciences, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Jingyu Wu
- Department of Chemical and Biomolecular Engineering, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Victoria G Muir
- Department of Bioengineering, School of Engineering and Applied Sciences, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Shoshana Weintraub
- Department of Bioengineering, School of Engineering and Applied Sciences, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Sarah E Gullbrand
- Translational Musculoskeletal Research Center, Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA, 19104, USA
- Department of Orthopaedic Surgery, McKay Orthopaedic Research Laboratory, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Daeyeon Lee
- Department of Chemical and Biomolecular Engineering, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - David Issadore
- Department of Bioengineering, School of Engineering and Applied Sciences, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Department of Electrical and Systems Engineering, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Jason A Burdick
- Department of Bioengineering, School of Engineering and Applied Sciences, University of Pennsylvania, Philadelphia, PA, 19104, USA
| |
Collapse
|
25
|
Hauck N, Neuendorf TA, Männel MJ, Vogel L, Liu P, Stündel E, Zhang Y, Thiele J. Processing of fast-gelling hydrogel precursors in microfluidics by electrocoalescence of reactive species. SOFT MATTER 2021; 17:10312-10321. [PMID: 34664052 PMCID: PMC8612358 DOI: 10.1039/d1sm01176f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Accepted: 10/12/2021] [Indexed: 06/13/2023]
Abstract
Microscopic hydrogels, also referred to as microgels, find broad application in life and materials science. A well-established technique for fabricating uniform microgels is droplet microfluidics. Here, optimal mixing of hydrogel precursor components is crucial to yield homogeneous microgels with respect to their morphology, mechanics, and distribution of functional moieties. However, when processing premixed polymer precursors that are highly reactive, fast or even instantaneous gelation inside fluid reservoirs or the microchannels of the flow cell commonly occurs, leading to an increase of fluid viscosity over time, and thus exacerbating the intrinsic control over fluid flow rates, droplet and microgel uniformity, which are key selling points of microfluidics in material design. To address these challenges, we utilize microflow cells with integrated electrodes, which enable fast addition and mixing of hydrogel precursors on demand by means of emulsion droplet coalescence. Here, two populations of surfactant-stabilized aqueous droplets - the first containing the material basis of the microgel, and the second containing another gel-forming component (e.g., a crosslinker) are formed at two consecutive microchannel junctions and merged via temporary thin-film instability. Our approach provides the ability to process such hydrogel systems that are otherwise challenging to process into uniform droplets and microgels by conventional droplet microfluidics. To demonstrate its versatility, we fabricate microgels with uniform shape and composition using fast hydrogelation via thiol-Michael addition reaction or non-covalent self-assembly. Furthermore, we elucidate the limitations of electrocoalescence of reactive hydrogel precursors by processing sodium alginate, crosslinked by calcium-induced ionic interactions. For this instantaneous type of hydrogelation, electrocoalescence of alginate and calcium ions does not result in the formation of morphologically isotropic microgels. Instead, it enables the creation of anisotropic microgel morphologies with tunable shape, which have previously only been achieved by selective crosslinking of elaborate higher-order emulsions or by aqueous two-phase systems as microgel templates.
Collapse
Affiliation(s)
- Nicolas Hauck
- Leibniz-Institut für Polymerforschung Dresden e.V., Hohe Str. 6, 01069 Dresden, Germany.
| | - Talika A Neuendorf
- Leibniz-Institut für Polymerforschung Dresden e.V., Hohe Str. 6, 01069 Dresden, Germany.
| | - Max J Männel
- Leibniz-Institut für Polymerforschung Dresden e.V., Hohe Str. 6, 01069 Dresden, Germany.
| | - Lucas Vogel
- Leibniz-Institut für Polymerforschung Dresden e.V., Hohe Str. 6, 01069 Dresden, Germany.
| | - Ping Liu
- B CUBE Center for Molecular Bioengineering, Technische Universität Dresden, Tatzberg 41, 01307 Dresden, Germany
| | - Enno Stündel
- Leibniz-Institut für Polymerforschung Dresden e.V., Hohe Str. 6, 01069 Dresden, Germany.
| | - Yixin Zhang
- B CUBE Center for Molecular Bioengineering, Technische Universität Dresden, Tatzberg 41, 01307 Dresden, Germany
- Cluster of Excellence Physics of Life, TU Dresden, Dresden, Germany
| | - Julian Thiele
- Leibniz-Institut für Polymerforschung Dresden e.V., Hohe Str. 6, 01069 Dresden, Germany.
| |
Collapse
|
26
|
Nazeer MA, Karaoglu IC, Ozer O, Albayrak C, Kizilel S. Neovascularization of engineered tissues for clinical translation: Where we are, where we should be? APL Bioeng 2021; 5:021503. [PMID: 33834155 PMCID: PMC8024034 DOI: 10.1063/5.0044027] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Accepted: 03/10/2021] [Indexed: 12/11/2022] Open
Abstract
One of the key challenges in engineering three-dimensional tissue constructs is the development of a mature microvascular network capable of supplying sufficient oxygen and nutrients to the tissue. Recent angiogenic therapeutic strategies have focused on vascularization of the constructed tissue, and its integration in vitro; these strategies typically combine regenerative cells, growth factors (GFs) with custom-designed biomaterials. However, the field needs to progress in the clinical translation of tissue engineering strategies. The article first presents a detailed description of the steps in neovascularization and the roles of extracellular matrix elements such as GFs in angiogenesis. It then delves into decellularization, cell, and GF-based strategies employed thus far for therapeutic angiogenesis, with a particularly detailed examination of different methods by which GFs are delivered in biomaterial scaffolds. Finally, interdisciplinary approaches involving advancement in biomaterials science and current state of technological development in fabrication techniques are critically evaluated, and a list of remaining challenges is presented that need to be solved for successful translation to the clinics.
Collapse
Affiliation(s)
| | | | - Onur Ozer
- Biomedical Sciences and Engineering, Koç University, Istanbul 34450, Turkey
| | - Cem Albayrak
- Authors to whom correspondence should be addressed: and
| | - Seda Kizilel
- Authors to whom correspondence should be addressed: and
| |
Collapse
|
27
|
Sustained Release Systems for Delivery of Therapeutic Peptide/Protein. Biomacromolecules 2021; 22:2299-2324. [PMID: 33957752 DOI: 10.1021/acs.biomac.1c00160] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Peptide/protein therapeutics have been significantly applied in the clinical treatment of various diseases such as cancer, diabetes, etc. owing to their high biocompatibility, specificity, and therapeutic efficacy. However, due to their immunogenicity, instability stemming from its complex tertiary and quaternary structure, vulnerability to enzyme degradation, and rapid renal clearance, the clinical application of protein/peptide therapeutics is significantly confined. Though nanotechnology has been demonstrated to prevent enzyme degradation of the protein therapeutics and thus enhance the half-life, issues such as initial burst release and uncontrollable release kinetics are still unsolved. Moreover, the traditional administration method results in poor patient compliance, limiting the clinical application of protein/peptide therapeutics. Exploiting the sustained-release formulations for more controllable delivery of protein/peptide therapeutics to decrease the frequency of injection and enhance patient compliance is thus greatly meaningful. In this review, we comprehensively summarize the substantial advancements of protein/peptide sustained-release systems in the past decades. In addition, the advantages and disadvantages of all these sustained-release systems in clinical application together with their future challenges are also discussed in this review.
Collapse
|
28
|
Rivero Berti I, Islan GA, Castro GR. Enzymes and biopolymers. The opportunity for the smart design of molecular delivery systems. BIORESOURCE TECHNOLOGY 2021; 322:124546. [PMID: 33360273 DOI: 10.1016/j.biortech.2020.124546] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 12/09/2020] [Accepted: 12/11/2020] [Indexed: 06/12/2023]
Abstract
Enzymes exhibit a tremendous potential due to the catalytic activity in response to physiological conditions and specific microenvironments. Exploiting these properties in combination with the versatility of biopolymers, a fascinating field for the rational development of a new class of "smart" delivery systems for therapeutic molecules is proposed. Many strategies have been recently developed to produce matrices with the desirable properties of molecular release, and enzymes could be playing a relevant role in modify the chemical composition of the polymers, the porosity and surface area of the matrices and modulate the kinetic of controlled release. Enzyme based computational systems have appeared as a relevant complementary tool to design novel smart bioactive matrices for programmable drug delivery. The present review is reporting the recent advances and projections of smart biopolymeric matrices activated by enzymes for sustained release of therapeutic molecules, highlighting various applications in the area of advanced drug delivery.
Collapse
Affiliation(s)
- Ignacio Rivero Berti
- Laboratorio de Nanobiomateriales, CINDEFI, Departamento de Química, Facultad de Ciencias Exactas, Universidad Nacional de La Plata (UNLP) - CONICET (CCT La Plata), Calle 47 y 115, (B1900AJI), La Plata, Buenos Aires, Argentina
| | - German A Islan
- Laboratorio de Nanobiomateriales, CINDEFI, Departamento de Química, Facultad de Ciencias Exactas, Universidad Nacional de La Plata (UNLP) - CONICET (CCT La Plata), Calle 47 y 115, (B1900AJI), La Plata, Buenos Aires, Argentina
| | - Guillermo R Castro
- Laboratorio de Nanobiomateriales, CINDEFI, Departamento de Química, Facultad de Ciencias Exactas, Universidad Nacional de La Plata (UNLP) - CONICET (CCT La Plata), Calle 47 y 115, (B1900AJI), La Plata, Buenos Aires, Argentina; Max Planck Laboratory for Structural Biology, Chemistry and Molecular Biophysics of Rosario (MPLbioR, UNR-MPIbpC), Partner Laboratory of the Max Planck Institute for Biophysical Chemistry (MPIbpC, MPG), Centro de Estudios Interdisciplinarios (CEI), Universidad Nacional de Rosario, Maipú 1065, S2000 Rosario, Santa Fe, Argentina.
| |
Collapse
|
29
|
Bacteriophage-Delivering Hydrogels: Current Progress in Combating Antibiotic Resistant Bacterial Infection. Antibiotics (Basel) 2021; 10:antibiotics10020130. [PMID: 33572929 PMCID: PMC7911734 DOI: 10.3390/antibiotics10020130] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 01/22/2021] [Accepted: 01/26/2021] [Indexed: 12/12/2022] Open
Abstract
Antibiotic resistance remains as an unresolved global challenge in the health care system, posing serious threats to global health. As an alternative to antibiotics, bacteriophage (phage) therapy is rising as a key to combating antibiotic-resistant bacterial infections. In order to deliver a phage to the site of infection, hydrogels have been formulated to incorporate phages, owing to its favorable characteristics in delivering biological molecules. This paper reviews the formulation of phage-delivering hydrogels for orthopedic implant-associated bone infection, catheter-associated urinary tract infection and trauma-associated wound infection, with a focus on the preparation methods, stability, efficacy and safety of hydrogels as phage carriers.
Collapse
|
30
|
Shieh H, Saadatmand M, Eskandari M, Bastani D. Microfluidic on-chip production of microgels using combined geometries. Sci Rep 2021; 11:1565. [PMID: 33452407 PMCID: PMC7810975 DOI: 10.1038/s41598-021-81214-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2020] [Accepted: 01/05/2021] [Indexed: 02/07/2023] Open
Abstract
Microfluidic on-chip production of microgels using external gelation can serve numerous applications that involve encapsulation of sensitive cargos. Nevertheless, on-chip production of microgels in microfluidic devices can be challenging due to problems induced by the rapid increase in precursor solution viscosity like clogging. Here, a novel design incorporating a step, which includes a sudden increase in cross-sectional area, before a flow-focusing nozzle was proposed for microfluidic droplet generators. Besides, a shielding oil phase was utilized to avoid the occurrence of emulsification and gelation stages simultaneously. The step which was located before the flow-focusing nozzle facilitated the full shielding of the dispersed phase due to 3-dimensional fluid flow in this geometry. The results showed that the microfluidic device was capable of generating highly monodispersed spherical droplets (CV < 2% for step and CV < 5% for flow-focusing nozzle) with an average diameter in the range of 90-190 μm, both in step and flow-focusing nozzle. Moreover, it was proved that the device could adequately create a shelter for the dispersed phase regardless of the droplet formation locus. The ability of this microfluidic device in the production of microgels was validated by creating alginate microgels (with an average diameter of ~ 100 μm) through an external gelation process with on-chip calcium chloride emulsion in mineral oil.
Collapse
Affiliation(s)
- Hamed Shieh
- Department of Chemical and Petroleum Engineering, Sharif University of Technology, Tehran, Iran
| | - Maryam Saadatmand
- Department of Chemical and Petroleum Engineering, Sharif University of Technology, Tehran, Iran.
| | - Mahnaz Eskandari
- Department of Biomedical Engineering, Amirkabir University of Technology, Tehran, Iran
| | - Dariush Bastani
- Department of Chemical and Petroleum Engineering, Sharif University of Technology, Tehran, Iran
| |
Collapse
|
31
|
Mora-Boza A, Mancipe Castro LM, Schneider RS, Han WM, García AJ, Vázquez-Lasa B, San Román J. Microfluidics generation of chitosan microgels containing glycerylphytate crosslinker for in situ human mesenchymal stem cells encapsulation. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 120:111716. [PMID: 33545868 PMCID: PMC8237249 DOI: 10.1016/j.msec.2020.111716] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 11/04/2020] [Accepted: 11/08/2020] [Indexed: 12/15/2022]
Abstract
Human mesenchymal stem cells (hMSCs) are an attractive source for cell therapies because of their multiple beneficial properties, i.e. via immunomodulation and secretory factors. Microfluidics is particularly attractive for cell encapsulation since it provides a rapid and reproducible methodology for microgel generation of controlled size and simultaneous cell encapsulation. Here, we report the fabrication of hMSC-laden microcarriers based on in situ ionotropic gelation of water-soluble chitosan in a microfluidic device using a combination of an antioxidant glycerylphytate (G1Phy) compound and tripolyphosphate (TPP) as ionic crosslinkers (G1Phy:TPP-microgels). These microgels showed homogeneous size distributions providing an average diameter of 104 ± 12 μm, somewhat lower than that of control (127 ± 16 μm, TPP-microgels). The presence of G1Phy in microgels maintained cell viability over time and upregulated paracrine factor secretion under adverse conditions compared to control TPP-microgels. Encapsulated hMSCs in G1Phy:TPP-microgels were delivered to the subcutaneous space of immunocompromised mice via injection, and the delivery process was as simple as the injection of unencapsulated cells. Immediately post-injection, equivalent signal intensities were observed between luciferase-expressing microgel-encapsulated and unencapsulated hMSCs, demonstrating no adverse effects of the microcarrier on initial cell survival. Cell persistence, inferred by bioluminescence signal, decreased exponentially over time showing relatively higher half-life values for G1Phy:TPP-microgels compared to TPP-microgels and unencapsulated cells. In overall, results position the microfluidics generated G1Phy:TPP-microgels as a promising microcarrier for supporting hMSC survival and reparative activities.
Collapse
Affiliation(s)
- Ana Mora-Boza
- Institute of Polymer Science and Technology (ICTP-CSIC), Madrid, Spain; CIBER-BBN, Health Institute Carlos III, Madrid, Spain
| | - Lina M Mancipe Castro
- Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, USA; Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA
| | - Rebecca S Schneider
- Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA; School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Woojin M Han
- Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, USA; Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA
| | - Andrés J García
- Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, USA; Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA
| | - Blanca Vázquez-Lasa
- Institute of Polymer Science and Technology (ICTP-CSIC), Madrid, Spain; CIBER-BBN, Health Institute Carlos III, Madrid, Spain.
| | - Julio San Román
- Institute of Polymer Science and Technology (ICTP-CSIC), Madrid, Spain; CIBER-BBN, Health Institute Carlos III, Madrid, Spain
| |
Collapse
|
32
|
Bizeau J, Mertz D. Design and applications of protein delivery systems in nanomedicine and tissue engineering. Adv Colloid Interface Sci 2021; 287:102334. [PMID: 33341459 DOI: 10.1016/j.cis.2020.102334] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 11/24/2020] [Accepted: 11/26/2020] [Indexed: 02/06/2023]
Abstract
Proteins are biological macromolecules involved in a wide range of biological functions, which makes them very appealing as therapeutics agents. Indeed, compared to small molecule drugs, their endogenous nature ensures their biocompatibility and biodegradability, they can be used in a large range of applications and present a higher specificity and activity. However, they suffer from unfolding, enzymatic degradation, short half-life and poor membrane permeability. To overcome such drawbacks, the development of protein delivery systems to protect, carry and deliver them in a controlled way have emerged importantly these last years. In this review, the formulation of a wide panel of protein delivery systems either in the form of polymer or inorganic nanoengineered colloids and scaffolds are presented and the protein loading and release mechanisms are addressed. A section is also dedicated to the detection of proteins and the characterization methods of their release. Then, the main protein delivery systems developed these last three years for anticancer, tissue engineering or diabetes applications are presented, as well as the major in vivo models used to test them. The last part of this review aims at presenting the perspectives of the field such as the use of protein-rich material or the sequestration of proteins. This part will also deal with less common applications and gene therapy as an indirect method to deliver protein.
Collapse
|
33
|
de Lima CSA, Balogh TS, Varca JPRO, Varca GHC, Lugão AB, A. Camacho-Cruz L, Bucio E, Kadlubowski SS. An Updated Review of Macro, Micro, and Nanostructured Hydrogels for Biomedical and Pharmaceutical Applications. Pharmaceutics 2020; 12:E970. [PMID: 33076231 PMCID: PMC7602430 DOI: 10.3390/pharmaceutics12100970] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2020] [Revised: 09/10/2020] [Accepted: 09/29/2020] [Indexed: 12/14/2022] Open
Abstract
Hydrogels are materials with wide applications in several fields, including the biomedical and pharmaceutical industries. Their properties such as the capacity of absorbing great amounts of aqueous solutions without losing shape and mechanical properties, as well as loading drugs of different nature, including hydrophobic ones and biomolecules, give an idea of their versatility and promising demand. As they have been explored in a great number of studies for years, many routes of synthesis have been developed, especially for chemical/permanent hydrogels. In the same way, stimuli-responsive hydrogels, also known as intelligent materials, have been explored too, enhancing the regulation of properties such as targeting and drug release. By controlling the particle size, hydrogel on the micro- and nanoscale have been studied likewise and have increased, even more, the possibilities for applications of the so-called XXI century materials. In this paper, we aimed to produce an overview of the recent studies concerning methods of synthesis, biomedical, and pharmaceutical applications of macro-, micro, and nanogels.
Collapse
Affiliation(s)
- Caroline S. A. de Lima
- Nuclear and Energy Research Institute, IPEN-CNEN/SP, Av. Prof. Lineu Prestes, No. 2242, Cidade Universitária, São Paulo 05508-000, Brazil; (C.S.A.d.L.); (T.S.B.); (J.P.R.O.V.); (A.B.L.)
| | - Tatiana S. Balogh
- Nuclear and Energy Research Institute, IPEN-CNEN/SP, Av. Prof. Lineu Prestes, No. 2242, Cidade Universitária, São Paulo 05508-000, Brazil; (C.S.A.d.L.); (T.S.B.); (J.P.R.O.V.); (A.B.L.)
| | - Justine P. R. O. Varca
- Nuclear and Energy Research Institute, IPEN-CNEN/SP, Av. Prof. Lineu Prestes, No. 2242, Cidade Universitária, São Paulo 05508-000, Brazil; (C.S.A.d.L.); (T.S.B.); (J.P.R.O.V.); (A.B.L.)
| | - Gustavo H. C. Varca
- Nuclear and Energy Research Institute, IPEN-CNEN/SP, Av. Prof. Lineu Prestes, No. 2242, Cidade Universitária, São Paulo 05508-000, Brazil; (C.S.A.d.L.); (T.S.B.); (J.P.R.O.V.); (A.B.L.)
| | - Ademar B. Lugão
- Nuclear and Energy Research Institute, IPEN-CNEN/SP, Av. Prof. Lineu Prestes, No. 2242, Cidade Universitária, São Paulo 05508-000, Brazil; (C.S.A.d.L.); (T.S.B.); (J.P.R.O.V.); (A.B.L.)
| | - Luis A. Camacho-Cruz
- Departamento de Química de Radiaciones y Radioquímica, Instituto de Ciencias Nucleares, Universidad Nacional Autónoma de México, Circuito Exterior, Ciudad Universitaria, México CDMX 04510, Mexico; (L.A.C.-C.); (E.B.)
| | - Emilio Bucio
- Departamento de Química de Radiaciones y Radioquímica, Instituto de Ciencias Nucleares, Universidad Nacional Autónoma de México, Circuito Exterior, Ciudad Universitaria, México CDMX 04510, Mexico; (L.A.C.-C.); (E.B.)
| | - Slawomir S. Kadlubowski
- Institute of Applied Radiation Chemistry (IARC), Lodz University of Technology, Wroblewskiego No. 15, 93-590 Lodz, Poland;
| |
Collapse
|
34
|
Ngo MT, Harley BAC. Angiogenic biomaterials to promote therapeutic regeneration and investigate disease progression. Biomaterials 2020; 255:120207. [PMID: 32569868 PMCID: PMC7396313 DOI: 10.1016/j.biomaterials.2020.120207] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 06/08/2020] [Accepted: 06/10/2020] [Indexed: 02/06/2023]
Abstract
The vasculature is a key component of the tissue microenvironment. Traditionally known for its role in providing nutrients and oxygen to surrounding cells, the vasculature is now also acknowledged to provide signaling cues that influence biological outcomes in regeneration and disease. These cues come from the cells that comprise vasculature, as well as the dynamic biophysical and biochemical properties of the surrounding extracellular matrix that accompany vascular development and remodeling. In this review, we illustrate the larger role of the vasculature in the context of regenerative biology and cancer progression. We describe cellular, biophysical, biochemical, and metabolic components of vascularized microenvironments. Moreover, we provide an overview of multidimensional angiogenic biomaterials that have been developed to promote therapeutic vascularization and regeneration, as well as to mimic elements of vascularized microenvironments as a means to uncover mechanisms by which vasculature influences cancer progression and therapy.
Collapse
Affiliation(s)
- Mai T Ngo
- Dept. Chemical and Biomolecular Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Brendan A C Harley
- Dept. Chemical and Biomolecular Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA; Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA; Cancer Center at Illinois, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA.
| |
Collapse
|
35
|
Mancipe Castro LM, Sequeira A, García AJ, Guldberg RE. Articular Cartilage- and Synoviocyte-Binding Poly(ethylene glycol) Nanocomposite Microgels as Intra-Articular Drug Delivery Vehicles for the Treatment of Osteoarthritis. ACS Biomater Sci Eng 2020; 6:5084-5095. [PMID: 33455260 PMCID: PMC8221079 DOI: 10.1021/acsbiomaterials.0c00960] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Intra-articular (IA) injection is an attractive route of administration for the treatment of osteoarthritis (OA). However, free drugs injected into the joint space are rapidly cleared and many of them can induce adverse off-target effects on different IA tissues. To overcome these limitations, we designed nanocomposite 4-arm-poly(ethylene glycol)-maleimide (PEG-4MAL) microgels, presenting cartilage- or synoviocyte-binding peptides, containing poly(lactic-co-glycolic) acid (PLGA) nanoparticles (NPs) as an IA small molecule drug delivery system. Microgels containing rhodamine B (model drug)-loaded PLGA NPs were synthesized using microfluidics technology and exhibited a sustained, near zero-order release of the fluorophore over 16 days in vitro. PEG-4MAL microgels presenting synoviocyte- or cartilage-targeting peptides specifically bound to rabbit and human synoviocytes or to bovine articular cartilage in vitro, respectively. Finally, using a rat model of post-traumatic knee OA, PEG-4MAL microgels were shown to be retained in the joint space for at least 3 weeks without inducing any joint degenerative changes as measured by EPIC-μCT and histology. Additionally, all microgel formulations were found trapped in the synovial membrane and significantly increased the IA retention time of a model small molecule near-infrared (NIR) dye compared to that of the free dye. These results suggest that peptide-functionalized nanocomposite PEG-4MAL microgels represent a promising intra-articular vehicle for tissue-localized drug delivery and prolonged IA drug retention for the treatment of OA.
Collapse
Affiliation(s)
- Lina María Mancipe Castro
- Parker H. Petit Institute for Bioengineering and
Biosciences, Georgia Institute of Technology, 315 Ferst Dr NW, Atlanta, GA 30332,
U.S.A
- George W. Woodruff School of Mechanical Engineering,
Georgia Institute of Technology, 315 Ferst Dr NW, Atlanta, GA 30332, U.S.A
| | - Abigail Sequeira
- School of Chemical and Biomolecular Engineering, Georgia
Institute of Technology, 311 Ferst Drive NW, Atlanta, GA 30332, U.S.A
| | - Andrés J. García
- Parker H. Petit Institute for Bioengineering and
Biosciences, Georgia Institute of Technology, 315 Ferst Dr NW, Atlanta, GA 30332,
U.S.A
- George W. Woodruff School of Mechanical Engineering,
Georgia Institute of Technology, 315 Ferst Dr NW, Atlanta, GA 30332, U.S.A
| | - Robert E. Guldberg
- Phil and Penny Knight Campus for Accelerating Scientific
Impact, University of Oregon, 6231 University of Oregon, Eugene, OR 97403-6231
| |
Collapse
|
36
|
Caldwell AS, Aguado BA, Anseth KS. Designing Microgels for Cell Culture and Controlled Assembly of Tissue Microenvironments. ADVANCED FUNCTIONAL MATERIALS 2020; 30:1907670. [PMID: 33841061 PMCID: PMC8026140 DOI: 10.1002/adfm.201907670] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Indexed: 05/04/2023]
Abstract
Micron-sized hydrogels, termed microgels, are emerging as multifunctional platforms that can recapitulate tissue heterogeneity in engineered cell microenvironments. The microgels can function as either individual cell culture units or can be assembled into larger scaffolds. In this manner, individual microgels can be customized for single or multi-cell co-culture applications, or heterogeneous populations can be used as building blocks to create microporous assembled scaffolds that more closely mimic tissue heterogeneities. The inherent versatility of these materials allows user-defined control of the microenvironments, from the order of singly encapsulated cells to entire three-dimensional cell scaffolds. These hydrogel scaffolds are promising for moving towards personalized medicine approaches and recapitulating the multifaceted microenvironments that exist in vivo.
Collapse
Affiliation(s)
- Alexander S. Caldwell
- Department of Chemical and Biological Engineering, University of Colorado – Boulder, USA, 80303
- BioFrontiers Institute, University of Colorado – Boulder, USA, 80303
| | - Brian A. Aguado
- Department of Chemical and Biological Engineering, University of Colorado – Boulder, USA, 80303
- BioFrontiers Institute, University of Colorado – Boulder, USA, 80303
| | - Kristi S. Anseth
- Department of Chemical and Biological Engineering, University of Colorado – Boulder, USA, 80303
- BioFrontiers Institute, University of Colorado – Boulder, USA, 80303
| |
Collapse
|
37
|
Shodeinde AB, Murphy AC, Oldenkamp HF, Potdar AS, Ludolph CM, Peppas NA. Recent Advances in Smart Biomaterials for the Detection and Treatment of Autoimmune Diseases. ADVANCED FUNCTIONAL MATERIALS 2020; 30:1909556. [PMID: 33071713 PMCID: PMC7566744 DOI: 10.1002/adfm.201909556] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Accepted: 01/15/2020] [Indexed: 05/07/2023]
Abstract
Autoimmune diseases are a group of debilitating illnesses that are often idiopathic in nature. The steady rise in the prevalence of these conditions warrants new approaches for diagnosis and treatment. Stimuli-responsive biomaterials also known as "smart", "intelligent" or "recognitive" biomaterials are widely studied for their applications in drug delivery, biosensing and tissue engineering due to their ability to produce thermal, optical, chemical, or structural changes upon interacting with the biological environment. This critical analysis highlights studies within the last decade that harness the recognitive capabilities of these biomaterials towards the development of novel detection and treatment options for autoimmune diseases.
Collapse
Affiliation(s)
- Aaliyah B. Shodeinde
- McKetta Department of Chemical Engineering, 200 E. Dean Keeton St. Stop C0400, Austin, TX, USA, 78712
- Institute for Biomaterials, Drug Delivery, and Regenerative Medicine, The University of Texas at Austin, 107 W Dean Keeton Street Stop C0800, Austin, TX, USA, 78712
| | - Andrew C. Murphy
- McKetta Department of Chemical Engineering, 200 E. Dean Keeton St. Stop C0400, Austin, TX, USA, 78712
- Institute for Biomaterials, Drug Delivery, and Regenerative Medicine, The University of Texas at Austin, 107 W Dean Keeton Street Stop C0800, Austin, TX, USA, 78712
| | - Heidi F. Oldenkamp
- McKetta Department of Chemical Engineering, 200 E. Dean Keeton St. Stop C0400, Austin, TX, USA, 78712
- Institute for Biomaterials, Drug Delivery, and Regenerative Medicine, The University of Texas at Austin, 107 W Dean Keeton Street Stop C0800, Austin, TX, USA, 78712
| | - Abhishek S. Potdar
- Department of Biomedical Engineering, The University of Texas at Austin, 107 W Dean Keeton Street Stop C0800, Austin, TX, USA, 78712
| | - Catherine M. Ludolph
- McKetta Department of Chemical Engineering, 200 E. Dean Keeton St. Stop C0400, Austin, TX, USA, 78712
| | - Nicholas A. Peppas
- McKetta Department of Chemical Engineering, 200 E. Dean Keeton St. Stop C0400, Austin, TX, USA, 78712
- Institute for Biomaterials, Drug Delivery, and Regenerative Medicine, The University of Texas at Austin, 107 W Dean Keeton Street Stop C0800, Austin, TX, USA, 78712
- Department of Biomedical Engineering, The University of Texas at Austin, 107 W Dean Keeton Street Stop C0800, Austin, TX, USA, 78712
- Division of Molecular Pharmaceutics and Drug Delivery, College of Pharmacy, The University of Texas at Austin, 2409 University Ave. Stop A1900, Austin, TX, USA, 78712
- Department of Surgery and Perioperative Care, Dell Medical School, 1601 Trinity St., Bldg. B, Stop Z0800, Austin, TX, USA, 78712
- Department of Pediatrics, Dell Medical School, 1400 Barbara Jordan Blvd., Austin, TX, USA, 78723
| |
Collapse
|
38
|
Martin JR, Patil P, Yu F, Gupta MK, Duvall CL. Enhanced stem cell retention and antioxidative protection with injectable, ROS-degradable PEG hydrogels. Biomaterials 2020; 263:120377. [PMID: 32947094 DOI: 10.1016/j.biomaterials.2020.120377] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 09/04/2020] [Accepted: 09/06/2020] [Indexed: 12/15/2022]
Abstract
Poly(ethylene glycol) (PEG) hydrogels crosslinked with enzyme-cleavable peptides are promising biodegradable vehicles for therapeutic cell delivery. However, peptide synthesis at the level required for bulk biomaterial manufacturing is costly, and fabrication of hydrogels from scalable, low-cost synthetic precursors while supporting cell-specific degradation remains a challenge. Reactive oxygen species (ROS) are cell-generated signaling molecules that can also be used as a trigger to mediate specific in vivo degradation of biomaterials. Here, PEG-based hydrogels crosslinked with ROS-degradable poly(thioketal) (PTK) polymers were successfully synthesized via thiol-maleimide chemistry and employed as a cell-degradable, antioxidative stem cell delivery platform. PTK hydrogels were mechanically robust and underwent ROS-mediated, dose-dependent degradation in vitro, while promoting robust cellular infiltration, tissue regeneration, and bioresorption in vivo. Moreover, these ROS-sensitive materials successfully encapsulated mesenchymal stem cells (MSCs) and maintained over 40% more viable cells than gold-standard hydrogels crosslinked with enzymatically-degradable peptides. The higher cellular survival in PTK-based gels was associated with the antioxidative function of the ROS-sensitive crosslinker, which scavenged free radicals and protected encapsulated MSCs from cytotoxic doses of ROS. Improved MSC viability was also observed in vivo as MSCs delivered within injectable PTK hydrogels maintained significantly more viability over 11 days compared against cells delivered within gels crosslinked with either a PEG-only control polymer or a gold-standard enzymatically-degradable peptide. Together, this study establishes a new paradigm for scalable creation and application of cell-degradable hydrogels, particularly for cell delivery applications.
Collapse
Affiliation(s)
- John R Martin
- Biomedical Engineering, Vanderbilt University, 5824 Stevenson Center, PMB 351631, Nashville, TN, 37235-1631, USA
| | - Prarthana Patil
- Biomedical Engineering, Vanderbilt University, 5824 Stevenson Center, PMB 351631, Nashville, TN, 37235-1631, USA
| | - Fang Yu
- Biomedical Engineering, Vanderbilt University, 5824 Stevenson Center, PMB 351631, Nashville, TN, 37235-1631, USA
| | - Mukesh K Gupta
- Biomedical Engineering, Vanderbilt University, 5824 Stevenson Center, PMB 351631, Nashville, TN, 37235-1631, USA.
| | - Craig L Duvall
- Biomedical Engineering, Vanderbilt University, 5824 Stevenson Center, PMB 351631, Nashville, TN, 37235-1631, USA.
| |
Collapse
|
39
|
Trujillo S, Gonzalez-Garcia C, Rico P, Reid A, Windmill J, Dalby MJ, Salmeron-Sanchez M. Engineered 3D hydrogels with full-length fibronectin that sequester and present growth factors. Biomaterials 2020; 252:120104. [DOI: 10.1016/j.biomaterials.2020.120104] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Revised: 05/04/2020] [Accepted: 05/05/2020] [Indexed: 12/22/2022]
|
40
|
|
41
|
Lutzweiler G, Ndreu Halili A, Engin Vrana N. The Overview of Porous, Bioactive Scaffolds as Instructive Biomaterials for Tissue Regeneration and Their Clinical Translation. Pharmaceutics 2020; 12:E602. [PMID: 32610440 PMCID: PMC7407612 DOI: 10.3390/pharmaceutics12070602] [Citation(s) in RCA: 103] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Revised: 06/08/2020] [Accepted: 06/23/2020] [Indexed: 12/20/2022] Open
Abstract
Porous scaffolds have been employed for decades in the biomedical field where researchers have been seeking to produce an environment which could approach one of the extracellular matrixes supporting cells in natural tissues. Such three-dimensional systems offer many degrees of freedom to modulate cell activity, ranging from the chemistry of the structure and the architectural properties such as the porosity, the pore, and interconnection size. All these features can be exploited synergistically to tailor the cell-material interactions, and further, the tissue growth within the voids of the scaffold. Herein, an overview of the materials employed to generate porous scaffolds as well as the various techniques that are used to process them is supplied. Furthermore, scaffold parameters which modulate cell behavior are identified under distinct aspects: the architecture of inert scaffolds (i.e., pore and interconnection size, porosity, mechanical properties, etc.) alone on cell functions followed by comparison with bioactive scaffolds to grasp the most relevant features driving tissue regeneration. Finally, in vivo outcomes are highlighted comparing the accordance between in vitro and in vivo results in order to tackle the future translational challenges in tissue repair and regeneration.
Collapse
Affiliation(s)
- Gaëtan Lutzweiler
- Institut National de la Santé et de la Recherche Medicale, UMR_S 1121, 11 rue Humann, 67085 Strasbourg CEDEX, France
| | - Albana Ndreu Halili
- Department of Information Technology, Aleksander Moisiu University, 2001 Durres, Albania;
| | | |
Collapse
|
42
|
Chen X, Qian H, Qiao H, Dong B, Chen E, Huang D, Wang T, Chen W. Tumor-Adhesive and pH-Degradable Microgels by Microfluidics and Photo-Cross-Linking for Efficient Antiangiogenesis and Enhanced Cancer Chemotherapy. Biomacromolecules 2020; 21:1285-1294. [DOI: 10.1021/acs.biomac.0c00049] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Xingmei Chen
- Department of Pharmaceutical Engineering, School of Engineering, China Pharmaceutical University, Nanjing 211198, P. R. China
| | - Hongliang Qian
- Department of Pharmaceutical Engineering, School of Engineering, China Pharmaceutical University, Nanjing 211198, P. R. China
| | - Haishi Qiao
- Department of Pharmaceutical Engineering, School of Engineering, China Pharmaceutical University, Nanjing 211198, P. R. China
| | - Bin Dong
- Department of Pharmaceutical Engineering, School of Engineering, China Pharmaceutical University, Nanjing 211198, P. R. China
| | - Enping Chen
- Department of Pharmaceutical Engineering, School of Engineering, China Pharmaceutical University, Nanjing 211198, P. R. China
| | - Dechun Huang
- Department of Pharmaceutical Engineering, School of Engineering, China Pharmaceutical University, Nanjing 211198, P. R. China
| | - Ting Wang
- Department of Pharmaceutical Engineering, School of Engineering, China Pharmaceutical University, Nanjing 211198, P. R. China
| | - Wei Chen
- Department of Pharmaceutical Engineering, School of Engineering, China Pharmaceutical University, Nanjing 211198, P. R. China
| |
Collapse
|
43
|
Birgul Akolpoglu M, Inceoglu Y, Kizilel S. An all-aqueous approach for physical immobilization of PEG-lipid microgels on organoid surfaces. Colloids Surf B Biointerfaces 2020; 186:110708. [DOI: 10.1016/j.colsurfb.2019.110708] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Revised: 11/22/2019] [Accepted: 12/03/2019] [Indexed: 12/15/2022]
|
44
|
Daly AC, Riley L, Segura T, Burdick JA. Hydrogel microparticles for biomedical applications. NATURE REVIEWS. MATERIALS 2020; 5:20-43. [PMID: 34123409 PMCID: PMC8191408 DOI: 10.1038/s41578-019-0148-6] [Citation(s) in RCA: 622] [Impact Index Per Article: 124.4] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 09/18/2019] [Indexed: 05/13/2023]
Abstract
Hydrogel microparticles (HMPs) are promising for biomedical applications, ranging from the therapeutic delivery of cells and drugs to the production of scaffolds for tissue repair and bioinks for 3D printing. Biologics (cells and drugs) can be encapsulated into HMPs of predefined shapes and sizes using a variety of fabrication techniques (batch emulsion, microfluidics, lithography, electrohydrodynamic (EHD) spraying and mechanical fragmentation). HMPs can be formulated in suspensions to deliver therapeutics, as aggregates of particles (granular hydrogels) to form microporous scaffolds that promote cell infiltration or embedded within a bulk hydrogel to obtain multiscale behaviours. HMP suspensions and granular hydrogels can be injected for minimally invasive delivery of biologics, and they exhibit modular properties when comprised of mixtures of distinct HMP populations. In this Review, we discuss the fabrication techniques that are available for fabricating HMPs, as well as the multiscale behaviours of HMP systems and their functional properties, highlighting their advantages over traditional bulk hydrogels. Furthermore, we discuss applications of HMPs in the fields of cell delivery, drug delivery, scaffold design and biofabrication.
Collapse
Affiliation(s)
- Andrew C. Daly
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
- These authors contributed equally: Andrew C. Daly, Lindsay Riley
| | - Lindsay Riley
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
- These authors contributed equally: Andrew C. Daly, Lindsay Riley
| | - Tatiana Segura
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
- Departments of Dermatology and Neurology, Duke University, Durham, NC, USA
| | - Jason A. Burdick
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
45
|
Sun P, Huang T, Wang X, Wang G, Liu Z, Chen G, Fan Q. Dynamic-Covalent Hydrogel with NIR-Triggered Drug Delivery for Localized Chemo-Photothermal Combination Therapy. Biomacromolecules 2019; 21:556-565. [PMID: 31804804 DOI: 10.1021/acs.biomac.9b01290] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Near-infrared (NIR) light-responsive, injectable hydrogels are among the most promising drug delivery systems for localized anticancer therapy owing to its minimally invasive administration and remote-controlled manner. However, most currently reported NIR-responsive hydrogels were usually generated through physical mixing of thermosensitive polymers and photothermal conversion agents. In this study, a novel type of dynamic-covalent hydrogel (GelPV-DOX-DBNP) with NIR light-triggered drug release behavior was rationally designed for chemo-photothermal combination treatment of tumors. Concretely, this NIR-responsive hydrogel was formed by specific benzoxaborole-carbohydrate interactions between benzoxaborole (BOB)-modified hyaluronic acid (BOB-HA) and fructose-based glycopolymer (PolyFru), where photosensitizer perylene diimide zwitterionic polymer (PDS), reductant ascorbic acid (Vc), anticancer drug doxorubicin (DOX) as well as photothermal nanoparticles (DB-NPs) were encapsulated, simultaneously. Upon 660 nm light irradiation, both PDS and Vc within the designed hydrogel can convert oxygen into hydrogen peroxide, which could make hydrogel be degraded through the breakage of dynamic covalent bonds based on benzoxaborole-carbohydrate interactions, leading to NIR light-activatable release of DOX and DB-NPs from GelPV-DOX-DBNP. Furthermore, the released DB-NPs can convert 915 nm light irradiation into heat, enabling the application of GelPV-DOX-DBNP as a NIR-responsive drug delivery platform for both chemotherapy and photothermal therapy (PTT). In vivo results prove that GelPV-DOX-DBNP exhibited a markedly enhanced chemo-photothermal synergistic therapy for 4T1 tumor model mice, compared to chemotherapy alone or PTT. This work presents a new strategy to construct NIR light-responsive hydrogel as one alternative drug delivery system for anticancer applications.
Collapse
Affiliation(s)
- Pengfei Sun
- Key Laboratory for Organic Electronics and Information Displays & Institute of Advanced Materials (IAM), Jiangsu National Synergetic Innovation Center for Advanced Materials (SICAM) , Nanjing University of Posts & Telecommunications , Nanjing 210023 , China
| | - Ting Huang
- Key Laboratory for Organic Electronics and Information Displays & Institute of Advanced Materials (IAM), Jiangsu National Synergetic Innovation Center for Advanced Materials (SICAM) , Nanjing University of Posts & Telecommunications , Nanjing 210023 , China
| | - Xiaoxiao Wang
- Key Laboratory for Organic Electronics and Information Displays & Institute of Advanced Materials (IAM), Jiangsu National Synergetic Innovation Center for Advanced Materials (SICAM) , Nanjing University of Posts & Telecommunications , Nanjing 210023 , China
| | - Gaina Wang
- Key Laboratory for Organic Electronics and Information Displays & Institute of Advanced Materials (IAM), Jiangsu National Synergetic Innovation Center for Advanced Materials (SICAM) , Nanjing University of Posts & Telecommunications , Nanjing 210023 , China
| | - Zhijia Liu
- School of Materials Science and Engineering, GD Research Center for Functional Biomaterials Engineering and Technology , Sun Yat-sen University , Guangzhou 510275 , China.,Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM) , Nanjing University of Posts & Telecommunications , Nanjing 210023 , China
| | - Guosong Chen
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science , Fudan University , Shanghai 200433 , China
| | - Quli Fan
- Key Laboratory for Organic Electronics and Information Displays & Institute of Advanced Materials (IAM), Jiangsu National Synergetic Innovation Center for Advanced Materials (SICAM) , Nanjing University of Posts & Telecommunications , Nanjing 210023 , China
| |
Collapse
|
46
|
Mejías JC, Roy K. In-vitro and in-vivo characterization of a multi-stage enzyme-responsive nanoparticle-in-microgel pulmonary drug delivery system. J Control Release 2019; 316:393-403. [PMID: 31715279 DOI: 10.1016/j.jconrel.2019.09.012] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Accepted: 09/29/2019] [Indexed: 12/19/2022]
Abstract
Although the lung is an obvious target for site-specific delivery of many therapeutics for respiratory airway diseases such as asthma, COPD, and cystic fibrosis, novel strategies are needed to avoid key physiologic barriers for efficient delivery and controlled release of therapeutics to the lungs. Specifically, deposition into the deep lung requires particles with a 1-5μm aerodynamic diameter; however, particles with a geometric diameter less than 6μm are rapidly cleared by alveolar macrophages. Additionally, epithelial, endothelial, and fibroblast cells prefer smaller (< 300nm) nanoparticles for efficient endocytosis. Here we address these contradictory design requirements by using a nanoparticle-inside-microgel system (Nano-in-Microgel). Using an improved maleimide-thiol based Michael Addition during (water-in-oil) Emulsion (MADE) method, we fabricated both trypsin-responsive and neutrophil elastase-responsive polymeric Nano-in-Microgel to show the versatility of the system in easily exchanging enzyme-responsive crosslinkers for disease-specific proteases. By varying the initial macromer concentration, from 20 to 50% w/v, the size distribution means ranged from 4-8μm, enzymatic degradation of the microgels is within 30min, and in vitro macrophage phagocytosis is lower for the higher % w/v. We further demonstrated that in vivo lung delivery of the multi-stage carriers through the pulmonary route yields particle retention up to several hours and followed by clearance within in naïve mice. Our results provide a further understanding of how enzymatically-degradable multi-stage polymeric carriers can be used for pulmonary drug delivery.
Collapse
Affiliation(s)
- Joscelyn C Mejías
- The Wallace H. Coulter Department of Biomedical Engineering at Georgia Institute of Technology and Emory University, Atlanta, GA, 30332, USA
| | - Krishnendu Roy
- The Wallace H. Coulter Department of Biomedical Engineering at Georgia Institute of Technology and Emory University, Atlanta, GA, 30332, USA.
| |
Collapse
|
47
|
Yang D, Xiao J, Wang B, Li L, Kong X, Liao J. The immune reaction and degradation fate of scaffold in cartilage/bone tissue engineering. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2019; 104:109927. [DOI: 10.1016/j.msec.2019.109927] [Citation(s) in RCA: 115] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Revised: 06/17/2019] [Accepted: 06/26/2019] [Indexed: 01/05/2023]
|
48
|
Karg M, Pich A, Hellweg T, Hoare T, Lyon LA, Crassous JJ, Suzuki D, Gumerov RA, Schneider S, Potemkin II, Richtering W. Nanogels and Microgels: From Model Colloids to Applications, Recent Developments, and Future Trends. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2019; 35:6231-6255. [PMID: 30998365 DOI: 10.1021/acs.langmuir.8b04304] [Citation(s) in RCA: 374] [Impact Index Per Article: 62.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
Nanogels and microgels are soft, deformable, and penetrable objects with an internal gel-like structure that is swollen by the dispersing solvent. Their softness and the potential to respond to external stimuli like temperature, pressure, pH, ionic strength, and different analytes make them interesting as soft model systems in fundamental research as well as for a broad range of applications, in particular in the field of biological applications. Recent tremendous developments in their synthesis open access to systems with complex architectures and compositions allowing for tailoring microgels with specific properties. At the same time state-of-the-art theoretical and simulation approaches offer deeper understanding of the behavior and structure of nano- and microgels under external influences and confinement at interfaces or at high volume fractions. Developments in the experimental analysis of nano- and microgels have become particularly important for structural investigations covering a broad range of length scales relevant to the internal structure, the overall size and shape, and interparticle interactions in concentrated samples. Here we provide an overview of the state-of-the-art, recent developments as well as emerging trends in the field of nano- and microgels. The following aspects build the focus of our discussion: tailoring (multi)functionality through synthesis; the role in biological and biomedical applications; the structure and properties as a model system, e.g., for densely packed arrangements in bulk and at interfaces; as well as the theory and computer simulation.
Collapse
Affiliation(s)
- Matthias Karg
- Physical Chemistry I , Heinrich-Heine-University Duesseldorf , 40204 Duesseldorf , Germany
| | - Andrij Pich
- DWI-Leibnitz-Institute for Interactive Materials e.V. , 52056 Aachen , Germany
- Functional and Interactive Polymers, Institute for Technical and Macromolecular Chemistry , RWTH Aachen University , 52056 Aachen , Germany
| | - Thomas Hellweg
- Physical and Biophysical Chemistry , Bielefeld University , 33615 Bielefeld , Germany
| | - Todd Hoare
- Department of Chemical Engineering , McMaster University , Hamilton , Ontario L8S 4L8 , Canada
| | - L Andrew Lyon
- Schmid College of Science and Technology , Chapman University , Orange , California 92866 , United States
| | - J J Crassous
- Institute of Physical Chemistry , RWTH Aachen University , 52056 Aachen , Germany
| | | | - Rustam A Gumerov
- DWI-Leibnitz-Institute for Interactive Materials e.V. , 52056 Aachen , Germany
- Physics Department , Lomonosov Moscow State University , Moscow 119991 , Russian Federation
| | - Stefanie Schneider
- Institute of Physical Chemistry , RWTH Aachen University , 52056 Aachen , Germany
| | - Igor I Potemkin
- DWI-Leibnitz-Institute for Interactive Materials e.V. , 52056 Aachen , Germany
- Physics Department , Lomonosov Moscow State University , Moscow 119991 , Russian Federation
- National Research South Ural State University , Chelyabinsk 454080 , Russian Federation
| | - Walter Richtering
- Institute of Physical Chemistry , RWTH Aachen University , 52056 Aachen , Germany
| |
Collapse
|
49
|
Camerin F, Fernández-Rodríguez MÁ, Rovigatti L, Antonopoulou MN, Gnan N, Ninarello A, Isa L, Zaccarelli E. Microgels Adsorbed at Liquid-Liquid Interfaces: A Joint Numerical and Experimental Study. ACS NANO 2019; 13:4548-4559. [PMID: 30865829 DOI: 10.1021/acsnano.9b00390] [Citation(s) in RCA: 84] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/19/2023]
Abstract
Soft particles display highly versatile properties with respect to hard colloids and even more so at fluid-fluid interfaces. In particular, microgels, consisting of a cross-linked polymer network, are able to deform and flatten upon adsorption at the interface due to the balance between surface tension and internal elasticity. Despite the existence of experimental results, a detailed theoretical understanding of this phenomenon is still lacking due to the absence of appropriate microscopic models. In this work, we propose an advanced modeling of microgels at a flat water/oil interface. The model builds on a realistic description of the internal polymeric architecture and single-particle properties of the microgel and is able to reproduce its experimentally observed shape at the interface. Complementing molecular dynamics simulations with in situ cryo-electron microscopy experiments and atomic force microscopy imaging after Langmuir-Blodgett deposition, we compare the morphology of the microgels for different values of the cross-linking ratios. Our model allows for a systematic microscopic investigation of soft particles at fluid interfaces, which is essential to develop predictive power for the use of microgels in a broad range of applications, including the stabilization of smart emulsions and the versatile patterning of surfaces.
Collapse
Affiliation(s)
- Fabrizio Camerin
- CNR Institute for Complex Systems, Uos Sapienza , Piazzale Aldo Moro 2 , 00185 Roma , Italy
- Department of Basic and Applied Sciences for Engineering , Sapienza University of Rome , Via Antonio Scarpa 14 , 00161 Roma , Italy
| | - Miguel Ángel Fernández-Rodríguez
- Laboratory for Interfaces, Soft Matter and Assembly, Department of Materials , ETH Zürich , Vladimir-Prelog-Weg 5 , 8093 Zürich , Switzerland
| | - Lorenzo Rovigatti
- CNR Institute for Complex Systems, Uos Sapienza , Piazzale Aldo Moro 2 , 00185 Roma , Italy
- Department of Physics , Sapienza University of Rome , Piazzale Aldo Moro 2 , 00185 Roma , Italy
| | - Maria-Nefeli Antonopoulou
- Laboratory for Interfaces, Soft Matter and Assembly, Department of Materials , ETH Zürich , Vladimir-Prelog-Weg 5 , 8093 Zürich , Switzerland
| | - Nicoletta Gnan
- CNR Institute for Complex Systems, Uos Sapienza , Piazzale Aldo Moro 2 , 00185 Roma , Italy
- Department of Physics , Sapienza University of Rome , Piazzale Aldo Moro 2 , 00185 Roma , Italy
| | - Andrea Ninarello
- CNR Institute for Complex Systems, Uos Sapienza , Piazzale Aldo Moro 2 , 00185 Roma , Italy
- Department of Physics , Sapienza University of Rome , Piazzale Aldo Moro 2 , 00185 Roma , Italy
| | - Lucio Isa
- Laboratory for Interfaces, Soft Matter and Assembly, Department of Materials , ETH Zürich , Vladimir-Prelog-Weg 5 , 8093 Zürich , Switzerland
| | - Emanuela Zaccarelli
- CNR Institute for Complex Systems, Uos Sapienza , Piazzale Aldo Moro 2 , 00185 Roma , Italy
- Department of Physics , Sapienza University of Rome , Piazzale Aldo Moro 2 , 00185 Roma , Italy
| |
Collapse
|
50
|
Ahmad N, Colak B, Zhang DW, Gibbs MJ, Watkinson M, Becer CR, Gautrot JE, Krause S. Peptide Cross-Linked Poly (Ethylene Glycol) Hydrogel Films as Biosensor Coatings for the Detection of Collagenase. SENSORS (BASEL, SWITZERLAND) 2019; 19:E1677. [PMID: 30965649 PMCID: PMC6479908 DOI: 10.3390/s19071677] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Revised: 03/28/2019] [Accepted: 04/02/2019] [Indexed: 12/20/2022]
Abstract
Peptide cross-linked poly(ethylene glycol) hydrogel has been widely used for drug delivery and tissue engineering. However, the use of this material as a biosensor for the detection of collagenase has not been explored. Proteases play a key role in the pathology of diseases such as rheumatoid arthritis and osteoarthritis. The detection of this class of enzyme using the degradable hydrogel film format is promising as a point-of-care device for disease monitoring. In this study, a protease biosensor was developed based on the degradation of a peptide cross-linked poly(ethylene glycol) hydrogel film and demonstrated for the detection of collagenase. The hydrogel was deposited on gold-coated quartz crystals, and their degradation in the presence of collagenase was monitored using a quartz crystal microbalance (QCM). The biosensor was shown to respond to concentrations between 2 and 2000 nM in less than 10 min with a lower detection limit of 2 nM.
Collapse
Affiliation(s)
- Norlaily Ahmad
- School of Engineering and Material Science, Queen Mary University of London, Mile End Road, London, E1 4NS, UK.
- Centre of Foundation Studies, Universiti Teknologi MARA, Cawangan Selangor, Kampus Dengkil, Dengkil, Selangor 43800, Malaysia.
| | - Burcu Colak
- School of Engineering and Material Science, Queen Mary University of London, Mile End Road, London, E1 4NS, UK.
| | - De-Wen Zhang
- Institute of Medical Engineering, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an 710061, China.
| | - Martin John Gibbs
- School of Engineering and Material Science, Queen Mary University of London, Mile End Road, London, E1 4NS, UK.
| | - Michael Watkinson
- The Lennard-Jones Laboratories, School of Chemical and Physical Sciences, Keele University, Staffordshire, ST5 5BG, UK.
| | - C Remzi Becer
- Department of Chemistry, University of Warwick, Coventry, CV4 7AL, UK.
| | - Julien E Gautrot
- School of Engineering and Material Science, Queen Mary University of London, Mile End Road, London, E1 4NS, UK.
| | - Steffi Krause
- School of Engineering and Material Science, Queen Mary University of London, Mile End Road, London, E1 4NS, UK.
| |
Collapse
|