1
|
Zhang R, Brooker C, Whitehouse LLE, Thomson NH, Wood D, Tronci G. Mechanical and suture-holding properties of a UV-cured atelocollagen membrane with varied crosslinked architecture. Biomed Mater 2024; 19:065036. [PMID: 39419110 DOI: 10.1088/1748-605x/ad8828] [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: 07/10/2024] [Accepted: 10/17/2024] [Indexed: 10/19/2024]
Abstract
The mechanical competence and suturing ability of collagen-based membranes are paramount in guided bone regeneration (GBR) therapy, to ensure damage-free implantation, fixation and space maintenancein vivo. However, contact with the biological medium can induce swelling of collagen molecules, yielding risks of membrane sinking into the bone defect, early loss of barrier function, and irreversibly compromised clinical outcomes. To address these challenges, this study investigates the effect of the crosslinked network architecture on both mechanical and suture-holding properties of a new atelocollagen (AC) membrane. UV-cured networks were obtained via either single functionalisation of AC with 4-vinylbenzyl chloride (4VBC) or sequential functionalisation of AC with both 4VBC and methacrylic anhydride. The wet-state compression modulus (Ec) and swelling ratio (SR) were significantly affected by the UV-cured network architecture, leading up to a three-fold reduction in SR and about two-fold increase inEcin the sequentially functionalised, compared to the single-functionalised, samples. Electron microscopy, dimensional analysis and compression testing revealed the direct impact of the ethanol series dehydration process on membrane microstructure, yielding densification of the freshly synthesised porous samples and a pore-free microstructure with increasedEc. Nanoindentation tests via spherical bead-probe atomic force microscopy (AFM) confirmed an approximately two-fold increase in median (interquartile range (IQR)) elastic modulus in the sequentially functionalised (EAFM= 40 (13) kPa), with respect to single-functionalised (EAFM= 15 (9) kPa), variants. Noteworthy, the single-functionalised, but not the sequentially functionalised, samples displayed higher suture retention strength (SRS = 28 ± 2-35 ± 10 N∙mm-1) in both the dry state and following 1 h in phosphate buffered saline (PBS), compared to Bio-Gide® (SRS: 6 ± 1-14 ± 2 N∙mm-1), while a significant decrease was measured after 24 h in PBS (SRS= 1 ± 1 N∙mm-1). These structure-property relationships confirm the key role played by the molecular architecture of covalently crosslinked collagen, aimed towards long-lasting resorbable membranes for predictable GBR therapy.
Collapse
Affiliation(s)
- Ruya Zhang
- School of Dentistry, St. James's University Hospital, University of Leeds, Leeds LS9 7TF, United Kingdom
| | - Charles Brooker
- School of Dentistry, St. James's University Hospital, University of Leeds, Leeds LS9 7TF, United Kingdom
- Clothworkers' Centre for Textile Materials Innovation for Healthcare (CCTMIH), School of Design, University of Leeds, Leeds LS2 9JT, United Kingdom
| | - Laura L E Whitehouse
- School of Dentistry, St. James's University Hospital, University of Leeds, Leeds LS9 7TF, United Kingdom
| | - Neil H Thomson
- School of Dentistry, St. James's University Hospital, University of Leeds, Leeds LS9 7TF, United Kingdom
- School of Physics and Astronomy, University of Leeds, Leeds LS2 9JT, United Kingdom
- Bragg Centre for Materials Research, University of Leeds, Leeds LS2 9JT, United Kingdom
| | - David Wood
- School of Dentistry, St. James's University Hospital, University of Leeds, Leeds LS9 7TF, United Kingdom
- Bragg Centre for Materials Research, University of Leeds, Leeds LS2 9JT, United Kingdom
| | - Giuseppe Tronci
- School of Dentistry, St. James's University Hospital, University of Leeds, Leeds LS9 7TF, United Kingdom
- Clothworkers' Centre for Textile Materials Innovation for Healthcare (CCTMIH), School of Design, University of Leeds, Leeds LS2 9JT, United Kingdom
| |
Collapse
|
2
|
Babiak PM, Battistoni CM, Cahya L, Athreya R, Minnich J, Panitch A, Liu JC. Tunable Blended Collagen I/II and Collagen I/III Hydrogels as Tissue Mimics. Macromol Biosci 2024:e2400280. [PMID: 39427345 DOI: 10.1002/mabi.202400280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 10/03/2024] [Indexed: 10/22/2024]
Abstract
Collagen (Col) is commonly used as a natural biomaterial for biomedical applications. Although Col I is the most prevalent col type employed, many collagen types work together in vivo to confer function and biological activity. Thus, blending collagen types can better recapitulate many native environments. This work investigates how hydrogel properties can be tuned through blending collagen types (col I/II and col I/III) and by varying polymerization temperatures. Col I/II results in poorly developed fibril networks, which softened the gels, especially at lower polymerization temperatures. Conversely, col I/III hydrogels exhibit well-connected fibril networks with localized areas of fine fibrils and result in stiffer hydrogels. A decreased molecular mass recovery rate is observed in blended hydrogels. The altered fibril morphologies, mechanical properties, and biological signals of the blended gels can be leveraged to alter cell responses and can be used as models for different tissue types (e.g., healthy vs fibrotic tissue). Furthermore, the biomimetic hydrogel properties are a tool that can be used to modulate the transport of drugs, nutrients, and wastes in tissue engineering applications.
Collapse
Affiliation(s)
- Paulina M Babiak
- Davidson School of Chemical Engineering, Purdue University, West Lafayette, IN, 47907, USA
| | - Carly M Battistoni
- Davidson School of Chemical Engineering, Purdue University, West Lafayette, IN, 47907, USA
| | - Leonard Cahya
- Davidson School of Chemical Engineering, Purdue University, West Lafayette, IN, 47907, USA
| | - Rithika Athreya
- Davidson School of Chemical Engineering, Purdue University, West Lafayette, IN, 47907, USA
| | - Jason Minnich
- Davidson School of Chemical Engineering, Purdue University, West Lafayette, IN, 47907, USA
| | - Alyssa Panitch
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, 30332, USA
| | - Julie C Liu
- Davidson School of Chemical Engineering, Purdue University, West Lafayette, IN, 47907, USA
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, 47907, USA
| |
Collapse
|
3
|
Ates B, Eroglu T, Sahsuvar S, Kirimli CE, Kocaturk O, Senay S, Gok O. Hydrogel-Integrated Heart-on-a-Chip Platform for Assessment of Myocardial Ischemia Markers. ACS OMEGA 2024; 9:42103-42115. [PMID: 39431078 PMCID: PMC11483411 DOI: 10.1021/acsomega.4c02121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 09/11/2024] [Accepted: 09/20/2024] [Indexed: 10/22/2024]
Abstract
Organ-on-a-chip platform scans offer a controllable environment and a physiological similarity to mimic human pathophysiology. In this study, a single-channel PDMS microchip was fabricated, characterized, and optimized to obtain a heart-on-a-chip platform, which is integrated with a hydrogel scaffold suitable for cardiomyocyte growth inside its channel. Single-channel chips with a size of 20 × 12 mm and a channel height ranging from 60 to 100 μm were produced using photolithography and soft lithography techniques. A gelatin-embedded alginate network-based hydrogel was further augmented with 3% (v/v) collagen type I. Pore sizes were in the range of 74-153 μm for H9C2 implantation and biomimicry. The hydrogels are characterized both on PDMS surfaces and in capillaries. The primary feature distinguishing this study from previous microchip studies is that it mimics the cell microenvironment much better using different hydrogel formulations instead of creating a 2D cell culture by passing fluids, such as fibronectin, for cell adhesion. Instead of using complex microchip designs, the chip system we created intends to provide a physiologically relevant copy by using a 3D cell culture to its advantage and a simple, single-channel architecture. The microchip study was combined with cardiomyocytes to create the heart-on-a-chip system and tested under normoxic and hypoxic conditions to create a myocardial ischemia model inside this channel. As a result, this heart-on-a-chip platform was shown to be utilized for the detection of several small-size biomarkers such as adenosine, ADP, lactic acid, l-isoleucine, l-glutamic acid, and oxidized glutathione via LC-MS/MS from control conditions and a myocardial ischemia model. Cell-embedded and hydrogel matrix-supported versions of this heart-on-a-chip system were successfully prepared and shown to provide powerful outputs with myocardial ischemia markers. In light of this research, these outputs aim to develop simple and biologically effective organ-on-a-chip systems for future research.
Collapse
Affiliation(s)
- Berna Ates
- Department
of Biomedical Engineering, Faculty of Engineering and Natural Sciences, Acibadem Mehmet Ali Aydinlar University, Istanbul 34752, Turkey
| | - Tolga Eroglu
- School
of Medicine, Acibadem Mehmet Ali Aydinlar
University, Istanbul 34752, Turkey
| | - Seray Sahsuvar
- Department
of Medical Biotechnology, Institute of Health Sciences, Acibadem Mehmet Ali Aydinlar University, Istanbul 34752, Turkey
| | - Ceyhun Ekrem Kirimli
- Department
of Biomedical Engineering, Faculty of Engineering and Natural Sciences, Acibadem Mehmet Ali Aydinlar University, Istanbul 34752, Turkey
| | - Ozgur Kocaturk
- Institute
of Biomedical Engineering, Bogazici University, Istanbul 34684, Turkey
| | - Sahin Senay
- Department
of Cardiovascular Surgery, School of Medicine, Acibadem Mehmet Ali Aydinlar University, Istanbul 34752, Turkey
| | - Ozgul Gok
- Department
of Biomedical Engineering, Faculty of Engineering and Natural Sciences, Acibadem Mehmet Ali Aydinlar University, Istanbul 34752, Turkey
| |
Collapse
|
4
|
Leon-Valdivieso CY, Bethry A, Pinese C, Dai M, Pompee C, Pernot JM, Garric X. Engineering Shape to Overcome Contraction: The Role of Polymer-Collagen Hybrids in Advanced Dermal Substitutes. J Biomed Mater Res A 2024. [PMID: 39381904 DOI: 10.1002/jbm.a.37805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 08/29/2024] [Accepted: 09/23/2024] [Indexed: 10/10/2024]
Abstract
Collagen gels are the standard dermal equivalents par excellence, however the problem of rapid cell-mediated contraction remains unresolved. Therefore, the development of hybrid constructs (HCs) based on collagen and polymeric scaffolds is proposed to address the mechanical instability that usually limits the formation of new, functional tissue. Equally important, these synthetic structures should be temporary (degradable) while ensuring that cells are well-adapted to the new extracellular environment. In this study, we screened a library of scaffolds made of various polymers, including homopolymers of polycaprolactone (PCL) and poly D,L-lactide (PLA50), their blends (PCL/PLA50), and copolymers (poly(D,L-lactide-co-caprolactone), PCLLA50) to prepare HCs in a layer-by-layer fashion. The properties of polymers and copolymers along with their processability by electrospinning and 3D-printing were evaluated. Then, we assessed the HCs resistance toward cell-mediated contraction as well as the degradation of the polymeric scaffolds. Our results indicate that scaffolds with higher PLA50 content (e.g., PLA50 100%, PCL/PLA50 or PCLLA50, both at 50/50 caprolactone-to-D,L-lactide molar ratio) presented more drawbacks in terms of handleability and processing, while those with greater PCL presence showed structural steadiness and ease to use. All the scaffolds integrated well with the collagen gel to form the corresponding HCs. With few exceptions, the HCs demonstrated good resistance to cell-derived contraction over 3 weeks. Notably, HCs based on PCLLA50 90/10 (both versions, electrospun or 3D-printed) performed best, showing only a 5%-17% area reduction compared to the 93% observed in collagen-only gels. This copolymer displayed hydrolytic degradation depending on its shape, with up to 45% and 65% loss of molecular weight for the electrospun and 3D-printed forms, respectively, correlating with their progressive change in mechanical features. HCs containing PCLLA50 90/10 also exhibited a better fibroblast distribution, enhanced myofibroblastic differentiation, and a three-fold increase in cell proliferation (when the electrospun type was used) compared to collagen controls. These findings were instrumental in selecting a potential HC that might be used for future experiments in vivo.
Collapse
Affiliation(s)
- Christopher Y Leon-Valdivieso
- Polymers for Health and Biomaterials, IBMM, University of Montpellier, CNRS, ENSCM, Montpellier, France
- CARTIGEN, University Hospital of Montpellier, Montpellier, France
| | - Audrey Bethry
- Polymers for Health and Biomaterials, IBMM, University of Montpellier, CNRS, ENSCM, Montpellier, France
| | - Coline Pinese
- Polymers for Health and Biomaterials, IBMM, University of Montpellier, CNRS, ENSCM, Montpellier, France
- Department of Pharmacy, University Hospital of Nimes, Nimes, France
| | - Michèle Dai
- URGO Recherche Innovation et Développement, Chenôve, France
| | - Christian Pompee
- Polymers for Health and Biomaterials, IBMM, University of Montpellier, CNRS, ENSCM, Montpellier, France
| | | | - Xavier Garric
- Polymers for Health and Biomaterials, IBMM, University of Montpellier, CNRS, ENSCM, Montpellier, France
- Department of Pharmacy, University Hospital of Nimes, Nimes, France
| |
Collapse
|
5
|
Zhang S, Zhong R, Younis MR, He H, Xu H, Li G, Yang R, Lui S, Wang Y, Wu M. Hydrogel Applications in the Diagnosis and Treatment of Glioblastoma. ACS APPLIED MATERIALS & INTERFACES 2024. [PMID: 39366948 DOI: 10.1021/acsami.4c11855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/06/2024]
Abstract
Glioblastoma multiforme (GBM), a common malignant neurological tumor, has boundaries indistinguishable from those of normal tissue, making complete surgical removal ineffective. The blood-brain barrier (BBB) further impedes the efficacy of radiotherapy and chemotherapy, leading to suboptimal treatment outcomes and a heightened probability of recurrence. Hydrogels offer multiple advantages for GBM diagnosis and treatment, including overcoming the BBB for improved drug delivery, controlled drug release for long-term efficacy, and enhanced relaxation properties of magnetic resonance imaging (MRI) contrast agents. Hydrogels, with their excellent biocompatibility and customizability, can mimic the in vivo microenvironment, support tumor cell culture, enable drug screening, and facilitate the study of tumor invasion and metastasis. This paper reviews the classification of hydrogels and recent research for the diagnosis and treatment of GBM, including their applications as cell culture platforms and drugs including imaging contrast agents carriers. The mechanisms of drug release from hydrogels and methods to monitor the activity of hydrogel-loaded drugs are also discussed. This review is intended to facilitate a more comprehensive understanding of the current state of GBM research. It offers insights into the design of integrated hydrogel-based GBM diagnosis and treatment with the objective of achieving the desired therapeutic effect and improving the prognosis of GBM.
Collapse
Affiliation(s)
- Shuaimei Zhang
- Department of Radiology and Huaxi MR Research Center (HMRRC), Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P. R. China
| | - Renming Zhong
- Radiotherapy Physics & Technology Center, Cancer Center, West China Hospital, Chengdu, Sichuan 610041, P. R. China
| | - Muhammad Rizwan Younis
- Department of Chemical and Biomolecular Engineering, Samueli School of Engineering, University of California at Los Angeles, Los Angeles, California 90095, United States
| | - Hualong He
- Department of Radiology and Huaxi MR Research Center (HMRRC), Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P. R. China
| | - Hong Xu
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan 610041, P. R. China
| | - Gaocan Li
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan 610041, P. R. China
| | - Ruiyan Yang
- Department of Biology, Macalester College, Saint Paul, Minnesota 55105, United States
| | - Su Lui
- Department of Radiology and Huaxi MR Research Center (HMRRC), Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P. R. China
| | - Yunbing Wang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan 610041, P. R. China
| | - Min Wu
- Department of Radiology and Huaxi MR Research Center (HMRRC), Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P. R. China
- Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu, Sichuan 610041, P. R. China
| |
Collapse
|
6
|
de Hilster RHJ, Reinders-Luinge MA, Schuil A, Borghuis T, Harmsen MC, Burgess JK, Hylkema MN. A 3D Epithelial-Mesenchymal Co-Culture Model of the Airway Wall Using Native Lung Extracellular Matrix. Bioengineering (Basel) 2024; 11:946. [PMID: 39329688 PMCID: PMC11428669 DOI: 10.3390/bioengineering11090946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2024] [Revised: 09/02/2024] [Accepted: 09/14/2024] [Indexed: 09/28/2024] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a chronic lung disease characterized by ongoing inflammation, impaired tissue repair, and aberrant interplay between airway epithelium and fibroblasts, resulting in an altered extracellular matrix (ECM) composition. The ECM is the three-dimensional (3D) scaffold that provides mechanical support and biochemical signals to cells, now recognized not only as a consequence but as a potential driver of disease progression. To elucidate how the ECM influences pathophysiological changes occurring in COPD, in vitro models are needed that incorporate the ECM. ECM hydrogels are a novel experimental tool for incorporating the ECM in experimental setups. We developed an airway wall model by combining lung-derived ECM hydrogels with a co-culture of primary human fibroblasts and epithelial cells at an air-liquid interface. Collagen IV and a mixture of collagen I, fibronectin, and bovine serum albumin were used as basement membrane-mimicking coatings. The model was initially assembled using porcine lung-derived ECM hydrogels and subsequently with COPD and non-COPD human lung-derived ECM hydrogels. The resulting 3D construct exhibited considerable contraction and supported co-culture, resulting in a differentiated epithelial layer. This multi-component 3D model allows the investigation of remodelling mechanisms, exploring ECM involvement in cellular crosstalk, and holds promise as a model for drug discovery studies exploring ECM involvement in cellular interactions.
Collapse
Affiliation(s)
- Roderick H. J. de Hilster
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, 9713 GZ Groningen, The Netherlands; (R.H.J.d.H.)
- Groningen Research Institute for Asthma and COPD (GRIAC), University of Groningen, University Medical Center Groningen, 9713 GZ Groningen, The Netherlands
| | - Marjan A. Reinders-Luinge
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, 9713 GZ Groningen, The Netherlands; (R.H.J.d.H.)
| | - Annemarie Schuil
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, 9713 GZ Groningen, The Netherlands; (R.H.J.d.H.)
| | - Theo Borghuis
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, 9713 GZ Groningen, The Netherlands; (R.H.J.d.H.)
- Groningen Research Institute for Asthma and COPD (GRIAC), University of Groningen, University Medical Center Groningen, 9713 GZ Groningen, The Netherlands
| | - Martin C. Harmsen
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, 9713 GZ Groningen, The Netherlands; (R.H.J.d.H.)
- KOLFF Institute—REGENERATE, University of Groningen, University Medical Center Groningen, FB41, 9713 AV Groningen, The Netherlands
| | - Janette K. Burgess
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, 9713 GZ Groningen, The Netherlands; (R.H.J.d.H.)
- Groningen Research Institute for Asthma and COPD (GRIAC), University of Groningen, University Medical Center Groningen, 9713 GZ Groningen, The Netherlands
- KOLFF Institute—REGENERATE, University of Groningen, University Medical Center Groningen, FB41, 9713 AV Groningen, The Netherlands
| | - Machteld N. Hylkema
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, 9713 GZ Groningen, The Netherlands; (R.H.J.d.H.)
- Groningen Research Institute for Asthma and COPD (GRIAC), University of Groningen, University Medical Center Groningen, 9713 GZ Groningen, The Netherlands
| |
Collapse
|
7
|
Brunel LG, Long CM, Christakopoulos F, Cai B, Johansson PK, Singhal D, Enejder A, Myung D, Heilshorn SC. Interpenetrating networks of fibrillar and amorphous collagen promote cell spreading and hydrogel stability. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.11.612534. [PMID: 39345483 PMCID: PMC11429934 DOI: 10.1101/2024.09.11.612534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Hydrogels composed of collagen, the most abundant protein in the human body, are widely used as scaffolds for tissue engineering due to their ability to support cellular activity. However, collagen hydrogels with encapsulated cells often experience bulk contraction due to cell-generated forces, and conventional strategies to mitigate this undesired deformation often compromise either the fibrillar microstructure or cytocompatibility of the collagen. To support the spreading of encapsulated cells while preserving the structural integrity of the gels, we present an interpenetrating network (IPN) of two distinct collagen networks with different crosslinking mechanisms and microstructures. First, a physically self-assembled collagen network preserves the fibrillar microstructure and enables the spreading of encapsulated human corneal mesenchymal stromal cells. Second, an amorphous collagen network covalently crosslinked with bioorthogonal chemistry fills the voids between fibrils and stabilizes the gel against cell-induced contraction. This collagen IPN balances the biofunctionality of natural collagen with the stability of covalently crosslinked, engineered polymers. Taken together, these data represent a new avenue for maintaining both the fiber-induced spreading of cells and the structural integrity of collagen hydrogels by leveraging an IPN of fibrillar and amorphous collagen networks. Statement of significance Collagen hydrogels are widely used as scaffolds for tissue engineering due to their support of cellular activity. However, collagen hydrogels often undergo undesired changes in size and shape due to cell-generated forces, and conventional strategies to mitigate this deformation typically compromise either the fibrillar microstructure or cytocompatibility of the collagen. In this study, we introduce an innovative interpenetrating network (IPN) that combines physically self-assembled, fibrillar collagen-ideal for promoting cell adhesion and spreading-with covalently crosslinked, amorphous collagen-ideal for enhancing bulk hydrogel stability. Our IPN design maintains the native fibrillar structure of collagen while significantly improving resistance against cell-induced contraction, providing a promising solution to enhance the performance and reliability of collagen hydrogels for tissue engineering applications. Graphical abstract
Collapse
|
8
|
Tan YH, Habing KM, Riesterer JL, Stempinski ES, Lewis SH, Pfeifer CS, Malhotra SV, Nakayama KH. Engineered nanofibrillar collagen with tunable biophysical properties for myogenic, endothelial, and osteogenic cell guidance. Acta Biomater 2024; 186:95-107. [PMID: 39117115 PMCID: PMC11407781 DOI: 10.1016/j.actbio.2024.08.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 07/10/2024] [Accepted: 08/01/2024] [Indexed: 08/10/2024]
Abstract
A goal of regenerative engineering is the rational design of materials to restore the structure-function relationships that drive reparative programs in damaged tissues. Despite the widespread use of extracellular matrices for engineering tissues, their application has been limited by a narrow range of tunable features. The primary objective of this study is to develop a versatile platform for evaluating tissue-specific cellular interactions using Type I collagen scaffolds with highly tunable biophysical properties. The kinetics of collagen fibrillogenesis were modulated through a combination of varied shear rate and pH during neutralization, to achieve a broad range of fibril anisotropy, porosity, diameter, and storage modulus. The role that each of these properties play in guiding muscle, bone, and vascular cell types was comprehensively identified, and informed the in vitro generation of three distinct musculoskeletal engineered constructs. Myogenesis was highly regulated by smaller fibrils and larger storage moduli, endothelial inflammatory phenotype was predominantly guided by fibril anisotropy, and osteogenesis was enhanced by highly porous collagen with larger fibrils. This study introduces a novel approach for dynamically modulating Type I collagen materials and provides a robust platform for investigating cell-material interactions, offering insights for the future rational design of tissue-specific regenerative biomaterials. STATEMENT OF SIGNIFICANCE: The biophysical properties of regenerative materials facilitate key cell-substrate interactions that can guide the morphology, phenotype, and biological response of cells. In this study, we describe the fabrication of an engineered collagen hydrogel that can be modified to exhibit control over a wide range of biophysical features, including fibril organization and size, nanoscale porosity, and mechanics. We identified the unique combination of collagen features that optimally promote regenerative muscle, bone, and vascular cell types while also delineating the properties that hinder these same cellular responses. This study presents a highly accessible method to control the biophysical properties of collagen hydrogels that can be adapted for a broad range of tissue engineering and regenerative applications.
Collapse
Affiliation(s)
- Yong How Tan
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, OR, USA
| | - Krista M Habing
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, OR, USA
| | - Jessica L Riesterer
- Cancer Early Detection Advanced Research Center, Oregon Health & Science University, Portland, OR, USA
| | - Erin S Stempinski
- Multiscale Microscopy Core, Oregon Health & Science University, Portland, OR, USA
| | - Steven H Lewis
- Biomaterial and Biomedical Sciences, Oregon Health & Science University, Portland, OR, USA
| | - Carmem S Pfeifer
- Biomaterial and Biomedical Sciences, Oregon Health & Science University, Portland, OR, USA
| | - Sanjay V Malhotra
- Department of Cell, Developmental and Cancer Biology, Oregon Health & Science University, Portland, OR, USA
| | - Karina H Nakayama
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, OR, USA; Department of Orthopaedics and Rehabilitation, Oregon Health & Science University, Portland, OR, USA.
| |
Collapse
|
9
|
Wang Y, Lv H, Ren S, Zhang J, Liu X, Chen S, Zhai J, Zhou Y. Biological Functions of Macromolecular Protein Hydrogels in Constructing Osteogenic Microenvironment. ACS Biomater Sci Eng 2024; 10:5513-5536. [PMID: 39173130 DOI: 10.1021/acsbiomaterials.4c00910] [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] [Indexed: 08/24/2024]
Abstract
Irreversible bone defects resulting from trauma, infection, and degenerative illnesses have emerged as a significant health concern. Structurally and functionally controllable hydrogels made by bone tissue engineering (BTE) have become promising biomaterials. Natural proteins are able to establish connections with autologous proteins through unique biologically active regions. Hydrogels based on proteins can simulate the bone microenvironment and regulate the biological behavior of stem cells in the tissue niche, making them candidates for research related to bone regeneration. This article reviews the biological functions of various natural macromolecular proteins (such as collagen, gelatin, fibrin, and silk fibroin) and highlights their special advantages as hydrogels. Then the latest research trends on cross-linking modified macromolecular protein hydrogels with improved mechanical properties and composite hydrogels loaded with exogenous micromolecular proteins have been discussed. Finally, the applications of protein hydrogels, such as 3D printed hydrogels, microspheres, and injectable hydrogels, were introduced, aiming to provide a reference for the repair of clinical bone defects.
Collapse
Affiliation(s)
- Yihan Wang
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun 130021, P. R. China
| | - Huixin Lv
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun 130021, P. R. China
| | - Sicong Ren
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun 130021, P. R. China
| | - Jiameng Zhang
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun 130021, P. R. China
| | - Xiuyu Liu
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun 130021, P. R. China
| | - Sheng Chen
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun 130021, P. R. China
| | - Jingjie Zhai
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun 130021, P. R. China
| | - Yanmin Zhou
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun 130021, P. R. China
| |
Collapse
|
10
|
Rodríguez-Cruz D, Boquet-Pujadas A, López-Muñoz E, Rincón-Heredia R, Paredes-Díaz R, Flores-Fortis M, Olivo-Marin JC, Guillén N, Aguilar-Rojas A. Three-dimensional cell culture conditions promoted the Mesenchymal-Amoeboid Transition in the Triple-Negative Breast Cancer cell line MDA-MB-231. Front Cell Dev Biol 2024; 12:1435708. [PMID: 39156975 PMCID: PMC11327030 DOI: 10.3389/fcell.2024.1435708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Accepted: 07/17/2024] [Indexed: 08/20/2024] Open
Abstract
Introduction Breast cancer (BC) is the leading cause of death among women, primarily due to its potential for metastasis. As BC progresses, the extracellular matrix (ECM) produces more type-I collagen, resulting in increased stiffness. This alteration influences cellular behaviors such as migration, invasion, and metastasis. Specifically, cancer cells undergo changes in gene expression that initially promote an epithelial-to-mesenchymal transition (EMT) and subsequently, a transition from a mesenchymal to an amoeboid (MAT) migration mode. In this way, cancer cells can migrate more easily through the stiffer microenvironment. Despite their importance, understanding MATs remains challenging due to the difficulty of replicating in vitro the conditions for cell migration that are observed in vivo. Methods To address this challenge, we developed a three-dimensional (3D) growth system that replicates the different matrix properties observed during the progression of a breast tumor. We used this model to study the migration and invasion of the Triple-Negative BC (TNBC) cell line MDA-MB-231, which is particularly subject to metastasis. Results Our results indicate that denser collagen matrices present a reduction in porosity, collagen fiber size, and collagen fiber orientation, which are associated with the transition of cells to a rounder morphology with bleb-like protrusions. We quantified how this transition is associated with a more persistent migration, an enhanced invasion capacity, and a reduced secretion of matrix metalloproteinases. Discussion Our findings suggest that the proposed 3D growth conditions (especially those with high collagen concentrations) mimic key features of MATs, providing a new platform to study the physiology of migratory transitions and their role in BC progression.
Collapse
Affiliation(s)
- Daniela Rodríguez-Cruz
- Medical Research Unit in Reproductive Medicine, High Specialty Medical Unit in Gynecology and Obstetrics No. 4 “Luis Castelazo Ayala”, Mexican Social Security Institute, Mexico City, Mexico
| | - Aleix Boquet-Pujadas
- École Polytechnique Fédérale de Lausanne, Biomedical Imaging Group, Lausanne, Switzerland
- Bioimage Analysis Unit, Pasteur Institute, Paris, France
- National Center for Scientific Research, CNRS UMR3691, Paris, France
| | - Eunice López-Muñoz
- Medical Research Unit in Reproductive Medicine, High Specialty Medical Unit in Gynecology and Obstetrics No. 4 “Luis Castelazo Ayala”, Mexican Social Security Institute, Mexico City, Mexico
| | - Ruth Rincón-Heredia
- Microscopy Core Unit, Institute of Cellular Physiology, National Autonomous University of Mexico, Mexico City, Mexico
| | - Rodolfo Paredes-Díaz
- Microscopy Core Unit, Institute of Cellular Physiology, National Autonomous University of Mexico, Mexico City, Mexico
| | - Mauricio Flores-Fortis
- Cuajimalpa Unit, Engineering and Natural Science Doctoral Program, Metropolitan Autonomous University, Mexico City, Mexico
- Cuajimalpa Unit, Department of Natural Science, Metropolitan Autonomous University, Mexico City, Mexico
| | - Jean-Christophe Olivo-Marin
- Bioimage Analysis Unit, Pasteur Institute, Paris, France
- National Center for Scientific Research, CNRS UMR3691, Paris, France
| | - Nancy Guillén
- Bioimage Analysis Unit, Pasteur Institute, Paris, France
- National Center for Scientific Research, CNRS ERL9195, Paris, France
| | - Arturo Aguilar-Rojas
- Medical Research Unit in Reproductive Medicine, High Specialty Medical Unit in Gynecology and Obstetrics No. 4 “Luis Castelazo Ayala”, Mexican Social Security Institute, Mexico City, Mexico
- Bioimage Analysis Unit, Pasteur Institute, Paris, France
| |
Collapse
|
11
|
Wierzbicka A, Bartniak M, Waśko J, Kolesińska B, Grabarczyk J, Bociaga D. The Impact of Gelatin and Fish Collagen on Alginate Hydrogel Properties: A Comparative Study. Gels 2024; 10:491. [PMID: 39195020 DOI: 10.3390/gels10080491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 07/19/2024] [Accepted: 07/22/2024] [Indexed: 08/29/2024] Open
Abstract
Hydrogel materials based on sodium alginate find versatile applications in regenerative medicine and tissue engineering due to their unique properties, such as biocompatibility and biodegradability, and the possibility of the customization of their mechanical properties, such as in terms of the individual requirements of separate clinical applications. These materials, however, have numerous limitations in the area of biological activity. In order to eliminate their limitations, sodium alginate is popularly applied in combination with added gelatin, which represents a product of collagen hydrolysis. Despite numerous beneficial biological properties, matrix materials based on gelatin have poor mechanical properties and are characterized by their ability for rapid degradation in an aqueous environment, particularly at the physiological temperature of the body, which significantly limits the independent application opportunities of this type of composition in the range of scaffolding production dedicated for tissue engineering. Collagen hydrogels, unlike gelatin, are characterized by higher bioactivity, dictated by a greater number of ligands that allow for cell adhesion, as well as better stability under physiological conditions. Fish-derived collagen provides a material that may be efficiently extracted without the risk of mammalian prion infection and can be used in all patients without religious restrictions. Considering the numerous advantages of collagen indicating its superiority over gelatin, within the framework of this study, the compositions of hydrogel materials based on sodium alginate and fish collagen in different concentrations were developed. Prepared hydrogel materials were compared with the properties of a typical composition of alginate with the addition of gelatin. The rheological, mechanical, and physicochemical properties of the developed polymer compositions were evaluated. The first trials of 3D printing by extrusion technique using the analyzed polymer solutions were also conducted. The results obtained indicate that replacing gelatin with fish collagen at an analogous concentration leads to obtaining materials with a lower swelling degree, better mechanical properties, higher stability, limited release kinetics of calcium ions cross-linking the alginate matrix, a slowed process of protein release under physiological conditions, and the possibility of extrusion 3D printing. The conducted analysis highlights that the optimization of the applied concentrations of fish collagen additives to composition based on sodium alginate creates the possibility of designing materials with appropriate mechanical and rheological properties and degradation kinetics adjusted to the requirements of specific applications, leading to the prospective opportunity to produce materials capable of mimicking the properties of relevant soft tissues. Thanks to its excellent bioactivity and lower-than-gelatin viscosity of the polymer solution, fish collagen also provides a prospective solution for applications in the field of 3D bioprinting.
Collapse
Affiliation(s)
- Adrianna Wierzbicka
- Institute of Materials Science and Engineering, Lodz University of Technology, 90-537 Lodz, Poland
| | - Mateusz Bartniak
- Institute of Materials Science and Engineering, Lodz University of Technology, 90-537 Lodz, Poland
| | - Joanna Waśko
- Institute of Organic Chemistry, Lodz University of Technology, 90-543 Lodz, Poland
| | - Beata Kolesińska
- Institute of Organic Chemistry, Lodz University of Technology, 90-543 Lodz, Poland
| | - Jacek Grabarczyk
- Institute of Materials Science and Engineering, Lodz University of Technology, 90-537 Lodz, Poland
| | - Dorota Bociaga
- Institute of Materials Science and Engineering, Lodz University of Technology, 90-537 Lodz, Poland
| |
Collapse
|
12
|
Pipis N, Stewart KA, Tabatabaei M, Williams LN, Allen JB. Exploring the Fibrous Nature of Single-Stranded DNA-Collagen Complexes: Nanostructural Observations and Physicochemical Insights. ACS OMEGA 2024; 9:32052-32058. [PMID: 39072094 PMCID: PMC11270544 DOI: 10.1021/acsomega.4c04104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 06/26/2024] [Accepted: 07/08/2024] [Indexed: 07/30/2024]
Abstract
Nucleic acid-collagen complexes (NACCs) are a self-assembled biomimetic fibrillary platform arising from the spontaneous complexation of single-stranded DNA (ssDNA) oligonucleotides and collagen. NACCs merge the extracellular matrix functionality of collagen with the tunable bioactivity of ssDNA as aptamers for broad biomedical applications. We hypothesize that NACCs offer a hierarchical architecture across multiple length scales that significantly varies compared to native collagen. We investigate this using atomic force microscopy and electron microscopy (transmission electron microscopy and cryogenic electron microscopy). Results demonstrate key topographical differences induced by adding ssDNA oligonucleotides to collagen type I. NACCs form a dense network of intertwined collagen fiber bundles in the microscale and nanoscale while retaining their characteristic D-band periodicities (∼67 nm). Additionally, our exploration of thermodynamic parameters governing the interaction indicates an entropically favorable NACC formation driven by ssDNA. Thermal analysis demonstrates the preservation of collagen's triple helical domains and a more stabilized polypeptide structure at higher temperatures than native collagen. These findings offer important insights into our understanding of the ssDNA-induced complexation of collagen toward the further establishment of structure-property relationships in NACCs and their future development into practical biomaterials. They also provide pathways for manipulating and enhancing collagenous matrices' properties without requiring complex chemical modifications or fabrication procedures.
Collapse
Affiliation(s)
- Nikolaos Pipis
- J.
Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, Florida 32611, United States
| | - Kevin A. Stewart
- George
& Josephine Butler Polymer Research Laboratory, Department of
Chemistry, Center for Macromolecular Science and Engineering, University of Florida, Gainesville, Florida 32611, United States
| | - Mohammad Tabatabaei
- J.
Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, Florida 32611, United States
| | - Lakiesha N. Williams
- J.
Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, Florida 32611, United States
| | - Josephine B. Allen
- J.
Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, Florida 32611, United States
- Department
of Materials Science & Engineering, University of Florida, Gainesville, Florida 32611, United States
| |
Collapse
|
13
|
Talaat S, Hashem AA, Abu-Seida A, Abdel Wahed A, Abdel Aziz TM. Regenerative potential of mesoporous silica nanoparticles scaffold on dental pulp and root maturation in immature dog's teeth: a histologic and radiographic study. BMC Oral Health 2024; 24:817. [PMID: 39026199 PMCID: PMC11264670 DOI: 10.1186/s12903-024-04368-6] [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: 11/23/2023] [Accepted: 05/13/2024] [Indexed: 07/20/2024] Open
Abstract
OBJECTIVE To evaluate histologically and radiographically the potential of dog's immature roots with apical periodontitis to regenerate after regenerative endodontic treatment using mesoporous silica nanoparticles (MSNs) with/without bone morphogenic protein (BMP-2) as scaffolds. METHODS In 4 mongrel dogs, 56 immature teeth with 96 roots were infected, resulting in necrotic pulps and periapical pathosis. According to the evaluation time (Group I = 30 days and Group II = 90 days), 90 roots were divided into two equal groups (45 roots each) and 6 roots used to replace any lost root during the procedure. The two main groups were further divided according to treatment protocol into 5 subgroups (9 roots each): blood clot (BC subgroup), mesoporous silica nanoparticles scaffold only (MSNs subgroup), mesoporous silica nanoparticles impregnated with BMP2 (MSNs + BMP2 subgroup), infected teeth without treatment (+ ve control subgroup) and normal untouched teeth (-ve control subgroup). All teeth surfaces were coated with Tincture iodine and calcium hydroxide was applied prior to treatment protocols. Then, teeth were restored with glass ionomer filling to seal the remaining part of the access cavity. Radiography evaluation of the increase in root length, root thickness and occurrence of apical closure were performed. Following the sacrifice of the two dogs at each time of evaluation, histopathological analysis was performed and included the inflammatory cells count, bone resorption, tissue ingrowth, deposition of hard tissue, and closure of the apical part. All data were statistically analyzed. RESULTS Compared to BC subgroup, MSNs and MSNs + BMP-2 subgroups exhibited significant higher increase in root length and thickness as well as higher vital tissue in-growth and new hard tissue formation in group II (P < 0.05). MSNs + BMP-2 subgroup had significant higher increase in root length and thickness as well as significant lower inflammatory cell count than MSNs subgroup in both groups (P < 0.05). There were no significant differences between MSNs and MSNs + BMP-2 subgroups regarding new hard tissue formation in both groups and apical closure in group I (P > 0.05). CONCLUSION MSNs with/without BMP-2 scaffolds enabled the continuing growth of roots in immature teeth with necrotic pulps and periapical pathosis. Addition of BMP-2 to MSNs scaffold improved its outcome in regenerative endodontics. CLINICAL RELEVANCE MSNs with/without BMP-2 scaffolds may alternate blood clot for regenerative endodontic treatment of immature teeth with necrotic pulps.
Collapse
Affiliation(s)
- Samar Talaat
- Endodontic Department, Faculty of Oral and Dental Medicine, Future University in Egypt, Cairo, Egypt.
| | - Ahmed A Hashem
- Department of Endodontic, Faculty of Dentistry, Ain Shams University, Cairo, Egypt.
| | - Ashraf Abu-Seida
- Department of Surgery, Anesthesiology and Radiology, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt
- Faculty of Dentistry, Galala University, New Galala City, Suez, Egypt
| | - Adel Abdel Wahed
- Endodontic Department, Faculty of Oral and Dental Medicine, Future University in Egypt, Cairo, Egypt
| | - Tarek M Abdel Aziz
- Department of Endodontic, Faculty of Dentistry, Ain Shams University, Cairo, Egypt
| |
Collapse
|
14
|
Mahmoudi C, Tahraoui Douma N, Mahmoudi H, Iurciuc (Tincu) CE, Popa M. Hydrogels Based on Proteins Cross-Linked with Carbonyl Derivatives of Polysaccharides, with Biomedical Applications. Int J Mol Sci 2024; 25:7839. [PMID: 39063081 PMCID: PMC11277554 DOI: 10.3390/ijms25147839] [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: 05/18/2024] [Revised: 07/08/2024] [Accepted: 07/10/2024] [Indexed: 07/28/2024] Open
Abstract
Adding carbonyl groups into the hydrogel matrix improves the stability and biocompatibility of the hydrogels, making them suitable for different biomedical applications. In this review article, we will discuss the use of hydrogels based on polysaccharides modified by oxidation, with particular attention paid to the introduction of carbonyl groups. These hydrogels have been developed for several applications in tissue engineering, drug delivery, and wound healing. The review article discusses the mechanism by which oxidized polysaccharides can introduce carbonyl groups, leading to the development of hydrogels through cross-linking with proteins. These hydrogels have tunable mechanical properties and improved biocompatibility. Hydrogels have dynamic properties that make them promising biomaterials for various biomedical applications. This paper comprehensively analyzes hydrogels based on cross-linked proteins with carbonyl groups derived from oxidized polysaccharides, including microparticles, nanoparticles, and films. The applications of these hydrogels in tissue engineering, drug delivery, and wound healing are also discussed.
Collapse
Affiliation(s)
- Chahrazed Mahmoudi
- Laboratory of Water and Environment, Faculty of Technology, University Hassiba Benbouali of Chlef, Chlef 02000, Algeria
- Department of Natural and Synthetic Polymers, Faculty of Chemical Engineering and Protection of the Environment, “Gheorghe Asachi” Technical University, 700050 Iasi, Romania
| | - Naïma Tahraoui Douma
- Laboratory of Water and Environment, Faculty of Technology, University Hassiba Benbouali of Chlef, Chlef 02000, Algeria
| | - Hacene Mahmoudi
- National Higher School of Nanosciences and Nanotechnologies, Algiers 16000, Algeria;
| | - Camelia Elena Iurciuc (Tincu)
- Department of Natural and Synthetic Polymers, Faculty of Chemical Engineering and Protection of the Environment, “Gheorghe Asachi” Technical University, 700050 Iasi, Romania
- Department of Pharmaceutical Technology, Faculty of Pharmacy, “Grigore T. Popa” University of Medicine and Pharmacy, University Street, No. 16, 700115 Iasi, Romania
| | - Marcel Popa
- Department of Natural and Synthetic Polymers, Faculty of Chemical Engineering and Protection of the Environment, “Gheorghe Asachi” Technical University, 700050 Iasi, Romania
- Academy of Romanian Scientists, 3 Ilfov, 050044 Bucharest, Romania
| |
Collapse
|
15
|
Kalairaj MS, Pradhan R, Saleem W, Smith MM, Gaharwar AK. Intra-Articular Injectable Biomaterials for Cartilage Repair and Regeneration. Adv Healthc Mater 2024; 13:e2303794. [PMID: 38324655 PMCID: PMC11468459 DOI: 10.1002/adhm.202303794] [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: 10/31/2023] [Revised: 12/29/2023] [Indexed: 02/09/2024]
Abstract
Osteoarthritis is a degenerative joint disease characterized by cartilage deterioration and subsequent inflammatory changes in the underlying bone. Injectable hydrogels have emerged as a promising approach for controlled drug delivery in cartilage therapies. This review focuses on the latest developments in utilizing injectable hydrogels as vehicles for targeted drug delivery to promote cartilage repair and regeneration. The pathogenesis of osteoarthritis is discussed to provide a comprehensive understanding of the disease progression. Subsequently, the various types of injectable hydrogels used for intra-articular delivery are discussed. Specifically, physically and chemically crosslinked injectable hydrogels are critically analyzed, with an emphasis on their fabrication strategies and their capacity to encapsulate and release therapeutic agents in a controlled manner. Furthermore, the potential of incorporating growth factors, anti-inflammatory drugs, and cells within these injectable hydrogels are discussed. Overall, this review offers a comprehensive guide to navigating the landscape of hydrogel-based therapeutics in osteoarthritis.
Collapse
Affiliation(s)
| | - Ridhi Pradhan
- Department of Biomedical EngineeringCollege of EngineeringTexas A&M UniversityCollege StationTX77843USA
| | - Waqas Saleem
- Department of Biomedical EngineeringCollege of EngineeringTexas A&M UniversityCollege StationTX77843USA
| | - Morgan M. Smith
- Department of Veterinary Integrative BiosciencesSchool of Veterinary Medicine and Biomedical SciencesTexas A&M UniversityCollege StationTX77843USA
| | - Akhilesh K. Gaharwar
- Department of Biomedical EngineeringCollege of EngineeringTexas A&M UniversityCollege StationTX77843USA
- Department of Material Science and EngineeringCollege of EngineeringTexas A&M UniversityCollege StationTX77843USA
- Genetics and Genomics Interdisciplinary ProgramTexas A&M UniversityCollege StationTX77843USA
| |
Collapse
|
16
|
Cheung BCH, Abbed RJ, Wu M, Leggett SE. 3D Traction Force Microscopy in Biological Gels: From Single Cells to Multicellular Spheroids. Annu Rev Biomed Eng 2024; 26:93-118. [PMID: 38316064 DOI: 10.1146/annurev-bioeng-103122-031130] [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] [Indexed: 02/07/2024]
Abstract
Cell traction force plays a critical role in directing cellular functions, such as proliferation, migration, and differentiation. Current understanding of cell traction force is largely derived from 2D measurements where cells are plated on 2D substrates. However, 2D measurements do not recapitulate a vital aspect of living systems; that is, cells actively remodel their surrounding extracellular matrix (ECM), and the remodeled ECM, in return, can have a profound impact on cell phenotype and traction force generation. This reciprocal adaptivity of living systems is encoded in the material properties of biological gels. In this review, we summarize recent progress in measuring cell traction force for cells embedded within 3D biological gels, with an emphasis on cell-ECM cross talk. We also provide perspectives on tools and techniques that could be adapted to measure cell traction force in complex biochemical and biophysical environments.
Collapse
Affiliation(s)
- Brian C H Cheung
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, New York, USA;
| | - Rana J Abbed
- Department of Bioengineering, University of Illinois Urbana-Champaign, Urbana, Illinois, USA;
| | - Mingming Wu
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, New York, USA;
| | - Susan E Leggett
- Department of Bioengineering, University of Illinois Urbana-Champaign, Urbana, Illinois, USA;
- Cancer Center at Illinois, University of Illinois Urbana-Champaign, Urbana, Illinois, USA
| |
Collapse
|
17
|
Patel R, Patel D. Injectable Hydrogels in Cardiovascular Tissue Engineering. Polymers (Basel) 2024; 16:1878. [PMID: 39000733 PMCID: PMC11244148 DOI: 10.3390/polym16131878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 06/28/2024] [Accepted: 06/29/2024] [Indexed: 07/17/2024] Open
Abstract
Heart problems are quite prevalent worldwide. Cardiomyocytes and stem cells are two examples of the cells and supporting matrix that are used in the integrated process of cardiac tissue regeneration. The objective is to create innovative materials that can effectively replace or repair damaged cardiac muscle. One of the most effective and appealing 3D/4D scaffolds for creating an appropriate milieu for damaged tissue growth and healing is hydrogel. In order to successfully regenerate heart tissue, bioactive and biocompatible hydrogels are required to preserve cells in the infarcted region and to bid support for the restoration of myocardial wall stress, cell survival and function. Heart tissue engineering uses a variety of hydrogels, such as natural or synthetic polymeric hydrogels. This article provides a quick overview of the various hydrogel types employed in cardiac tissue engineering. Their benefits and drawbacks are discussed. Hydrogel-based techniques for heart regeneration are also addressed, along with their clinical application and future in cardiac tissue engineering.
Collapse
Affiliation(s)
- Raj Patel
- Banas Medical College and Research Institute, Palanpur 385001, India;
| | - Dhruvi Patel
- School of Civil and Environmental Engineering, Cornell University, Ithaca, NY 14850, USA
| |
Collapse
|
18
|
Phuphanitcharoenkun S, Louis F, Sowa Y, Matsusaki M, Palaga T. Improving stability of human three dimensional skin equivalents using plasma surface treatment. Biotechnol Bioeng 2024; 121:1950-1960. [PMID: 38470332 DOI: 10.1002/bit.28690] [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: 10/21/2023] [Revised: 01/10/2024] [Accepted: 02/19/2024] [Indexed: 03/13/2024]
Abstract
In developing three-dimensional (3D) human skin equivalents (HSEs), preventing dermis and epidermis layer distortion due to the contraction of hydrogels by fibroblasts is a challenging issue. Previously, a fabrication method of HSEs was tested using a modified solid scaffold or a hydrogel matrix in combination with the natural polymer coated onto the tissue culture surface, but the obtained HSEs exhibited skin layer contraction and loss of the skin integrity and barrier functions. In this study, we investigated the method of HSE fabrication that enhances the stability of the skin model by using surface plasma treatment. The results showed that plasma treatment of the tissue culture surface prevented dermal layer shrinkage of HSEs, in contrast to the HSE fabrication using fibronectin coating. The HSEs from plasma-treated surface showed significantly higher transepithelial electrical resistance compared to the fibronectin-coated model. They also expressed markers of epidermal differentiation (keratin 10, keratin 14 and loricrin), epidermal tight junctions (claudin 1 and zonula occludens-1), and extracellular matrix proteins (collagen IV), and exhibited morphological characteristics of the primary human skins. Taken together, the use of plasma surface treatment significantly improves the stability of 3D HSEs with well-defined dermis and epidermis layers and enhanced skin integrity and the barrier functions.
Collapse
Affiliation(s)
- Suphanun Phuphanitcharoenkun
- Graduate Program in Biotechnology, Faculty of Science, Chulalongkorn University, Bangkok, Thailand
- Center of Excellence in Materials and Bio-Interfaces, Chulalongkorn University, Bangkok, Thailand
| | - Fiona Louis
- Joint Research Laboratory (TOPPAN) for Advanced Cell Regulatory Chemistry, Graduate School of Engineering, Osaka University, Osaka, Japan
| | - Yoshihiro Sowa
- Department of Plastic and Reconstructive Surgery, Graduate School of Medical Sciences, Kyoto Prefectural University of Medicine, Kyoto, Japan
- Department of Plastic Surgery, Jichi Medical University, Tochigi, Japan
| | - Michiya Matsusaki
- Joint Research Laboratory (TOPPAN) for Advanced Cell Regulatory Chemistry, Graduate School of Engineering, Osaka University, Osaka, Japan
- Department of Applied Chemistry, Graduate School of Engineering, Osaka University, Osaka, Japan
| | - Tanapat Palaga
- Center of Excellence in Materials and Bio-Interfaces, Chulalongkorn University, Bangkok, Thailand
- Department of Microbiology, Faculty of Science, Chulalongkorn University, Bangkok, Thailand
| |
Collapse
|
19
|
Siaghi M, Karimizade A, Mellati A, Saeedi M, Talebpour Amiri F, Kalhori S, Shahani S. Luteolin-incorporated fish collagen hydrogel scaffold: An effective drug delivery strategy for wound healing. Int J Pharm 2024; 657:124138. [PMID: 38642619 DOI: 10.1016/j.ijpharm.2024.124138] [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: 03/02/2024] [Revised: 04/17/2024] [Accepted: 04/17/2024] [Indexed: 04/22/2024]
Abstract
In clinical practice, wound care has always been challenging. Hydrogels play a key role in facilitating active wound recovery by absorbing exudates, maintaining moisture, and alleviating pain through cooling. In this study, type I collagen was isolated from the skin of crucian carp (Carassius carassius) and verified by amino acid analysis, FTIR, and SDS-PAGE. By adopting a new approach, luteolin was added to collagen hydrogels in situ after being dissolved in an alkaline solution. XRD and SEM confirmed the luteolin was incorporated and entirely distributed throughout the hydrogel. The plastic compression improved the young's modulus of hydrogel to 15.24 ± 0.59 kPa, which is adequate for wound protection. The drug loading efficiency was 98 ± 1.47 % in the selected formulation. The luteolin-incorporated hydrogel enabled regulated drug release. We assessed the cytotoxicity using MTT and live-dead assays, as well as examined the hemocompatibility to determine the biocompatibility of the hydrogel. In vivo experiments showed that the hydrogel with luteolin had the highest wound closure rate (94.01 ± 2.1 %) and improved wound healing with granular tissue formation, collagen deposition, and re-epithelialization. These findings indicate that this efficient drug delivery technology can accelerate the process of wound healing.
Collapse
Affiliation(s)
- Masoud Siaghi
- Department of Pharmacognosy and Biotechnology, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| | - Ayoob Karimizade
- Department of Tissue Engineering and Regenerative Medicine, School of Advanced Technologies in Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Amir Mellati
- Department of Tissue Engineering and Regenerative Medicine, School of Advanced Technologies in Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Majid Saeedi
- Department of Pharmaceutics, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| | - Fereshteh Talebpour Amiri
- Department of Anatomy, Molecular and Cell Biology Research Center, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Shakiba Kalhori
- Department of Pharmacognosy and Biotechnology, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| | - Somayeh Shahani
- Department of Pharmacognosy and Biotechnology, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran; Medicinal Plants Research Center, Mazandaran University of Medical Sciences, Sari, Iran.
| |
Collapse
|
20
|
Fuller AM, Pruitt HC, Liu Y, Irizarry-Negron VM, Pan H, Song H, DeVine A, Katti RS, Devalaraja S, Ciotti GE, Gonzalez MV, Williams EF, Murazzi I, Ntekoumes D, Skuli N, Hakonarson H, Zabransky DJ, Trevino JG, Weeraratna A, Weber K, Haldar M, Fraietta JA, Gerecht S, Eisinger-Mathason TSK. Oncogene-induced matrix reorganization controls CD8+ T cell function in the soft-tissue sarcoma microenvironment. J Clin Invest 2024; 134:e167826. [PMID: 38652549 PMCID: PMC11142734 DOI: 10.1172/jci167826] [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: 12/09/2022] [Accepted: 04/09/2024] [Indexed: 04/25/2024] Open
Abstract
CD8+ T cell dysfunction impedes antitumor immunity in solid cancers, but the underlying mechanisms are diverse and poorly understood. Extracellular matrix (ECM) composition has been linked to impaired T cell migration and enhanced tumor progression; however, impacts of individual ECM molecules on T cell function in the tumor microenvironment (TME) are only beginning to be elucidated. Upstream regulators of aberrant ECM deposition and organization in solid tumors are equally ill-defined. Therefore, we investigated how ECM composition modulates CD8+ T cell function in undifferentiated pleomorphic sarcoma (UPS), an immunologically active desmoplastic tumor. Using an autochthonous murine model of UPS and data from multiple human patient cohorts, we discovered a multifaceted mechanism wherein the transcriptional coactivator YAP1 promotes collagen VI (COLVI) deposition in the UPS TME. In turn, COLVI induces CD8+ T cell dysfunction and immune evasion by remodeling fibrillar collagen and inhibiting T cell autophagic flux. Unexpectedly, collagen I (COLI) opposed COLVI in this setting, promoting CD8+ T cell function and acting as a tumor suppressor. Thus, CD8+ T cell responses in sarcoma depend on oncogene-mediated ECM composition and remodeling.
Collapse
Affiliation(s)
- Ashley M Fuller
- Abramson Family Cancer Research Institute, Department of Pathology and Laboratory Medicine, Penn Sarcoma Program, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Hawley C Pruitt
- Department of Chemical and Biomolecular Engineering, Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, Maryland, USA
| | - Ying Liu
- Abramson Family Cancer Research Institute, Department of Pathology and Laboratory Medicine, Penn Sarcoma Program, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Valerie M Irizarry-Negron
- Abramson Family Cancer Research Institute, Department of Pathology and Laboratory Medicine, Penn Sarcoma Program, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Hehai Pan
- Abramson Family Cancer Research Institute, Department of Pathology and Laboratory Medicine, Penn Sarcoma Program, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Hoogeun Song
- Abramson Family Cancer Research Institute, Department of Pathology and Laboratory Medicine, Penn Sarcoma Program, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Ann DeVine
- Abramson Family Cancer Research Institute, Department of Pathology and Laboratory Medicine, Penn Sarcoma Program, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Rohan S Katti
- Abramson Family Cancer Research Institute, Department of Pathology and Laboratory Medicine, Penn Sarcoma Program, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Samir Devalaraja
- Abramson Family Cancer Research Institute, Department of Pathology and Laboratory Medicine, Penn Sarcoma Program, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Gabrielle E Ciotti
- Abramson Family Cancer Research Institute, Department of Pathology and Laboratory Medicine, Penn Sarcoma Program, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | | | - Erik F Williams
- Department of Microbiology, Center for Cellular Immunotherapies, Parker Institute for Cancer Immunotherapy, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Ileana Murazzi
- Abramson Family Cancer Research Institute, Department of Pathology and Laboratory Medicine, Penn Sarcoma Program, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Dimitris Ntekoumes
- Department of Chemical and Biomolecular Engineering, Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, Maryland, USA
- Department of Biomedical Engineering, Duke University, Durham, North Carolina, USA
| | - Nicolas Skuli
- Abramson Family Cancer Research Institute, Department of Pathology and Laboratory Medicine, Penn Sarcoma Program, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Hakon Hakonarson
- Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Daniel J Zabransky
- Department of Oncology, The Sidney Kimmel Cancer Center, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Jose G Trevino
- Division of Surgical Oncology, Department of Surgery, Virginia Commonwealth University School of Medicine, Richmond, Virginia, USA
| | - Ashani Weeraratna
- Department of Oncology, The Sidney Kimmel Cancer Center, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
- Department of Biochemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Kristy Weber
- Department of Orthopaedic Surgery, Penn Sarcoma Program, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Malay Haldar
- Abramson Family Cancer Research Institute, Department of Pathology and Laboratory Medicine, Penn Sarcoma Program, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Joseph A Fraietta
- Department of Microbiology, Center for Cellular Immunotherapies, Parker Institute for Cancer Immunotherapy, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Sharon Gerecht
- Department of Chemical and Biomolecular Engineering, Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, Maryland, USA
- Department of Biomedical Engineering, Duke University, Durham, North Carolina, USA
| | - T S Karin Eisinger-Mathason
- Abramson Family Cancer Research Institute, Department of Pathology and Laboratory Medicine, Penn Sarcoma Program, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| |
Collapse
|
21
|
Han Y, Xu J, Chopra H, Zhang Z, Dubey N, Dissanayaka W, Nör J, Bottino M. Injectable Tissue-Specific Hydrogel System for Pulp-Dentin Regeneration. J Dent Res 2024; 103:398-408. [PMID: 38410924 PMCID: PMC11457960 DOI: 10.1177/00220345241226649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/28/2024] Open
Abstract
The quest for finding a suitable scaffold system that supports cell survival and function and, ultimately, the regeneration of the pulp-dentin complex remains challenging. Herein, we hypothesized that dental pulp stem cells (DPSCs) encapsulated in a collagen-based hydrogel with varying stiffness would regenerate functional dental pulp and dentin when concentrically injected into the tooth slices. Collagen hydrogels with concentrations of 3 mg/mL (Col3) and 10 mg/mL (Col10) were prepared, and their stiffness and microstructure were assessed using a rheometer and scanning electron microscopy, respectively. DPSCs were then encapsulated in the hydrogels, and their viability and differentiation capacity toward endothelial and odontogenic lineages were evaluated using live/dead assay and quantitative real-time polymerase chain reaction. For in vivo experiments, DPSC-encapsulated collagen hydrogels with different stiffness, with or without growth factors, were injected into pulp chambers of dentin tooth slices and implanted subcutaneously in severe combined immunodeficient (SCID) mice. Specifically, vascular endothelial growth factor (VEGF [50 ng/mL]) was loaded into Col3 and bone morphogenetic protein (BMP2 [50 ng/mL]) into Col10. Pulp-dentin regeneration was evaluated by histological and immunofluorescence staining. Data were analyzed using 1-way or 2-way analysis of variance accordingly (α = 0.05). Rheology and microscopy data revealed that Col10 had a stiffness of 8,142 Pa with a more condensed and less porous structure, whereas Col3 had a stiffness of 735 Pa with a loose microstructure. Furthermore, both Col3 and Col10 supported DPSCs' survival. Quantitative polymerase chain reaction showed Col3 promoted significantly higher von Willebrand factor (VWF) and CD31 expression after 7 and 14 d under endothelial differentiation conditions (P < 0.05), whereas Col10 enhanced the expression of dentin sialophosphoprotein (DSPP), alkaline phosphatase (ALP), runt-related transcription factor 2 (Runx2), and collagen 1 (Col1) after 7, 14, and 21 d of odontogenic differentiation (P < 0.05). Hematoxylin and eosin and immunofluorescence (CD31 and vWF) staining revealed Col10+Col3+DPSCs+GFs enhanced pulp-dentin tissue regeneration. In conclusion, the collagen-based concentric construct modified by growth factors guided the specific lineage differentiation of DPSCs and promoted pulp-dentin tissue regeneration in vivo.
Collapse
Affiliation(s)
- Y. Han
- Department of Cariology, Restorative Sciences, and Endodontics, School of Dentistry, University of Michigan, Ann Arbor, MI, USA
- Applied Oral Sciences and Community Dental Care, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China
| | - J. Xu
- Department of Cariology, Restorative Sciences, and Endodontics, School of Dentistry, University of Michigan, Ann Arbor, MI, USA
| | - H. Chopra
- Department of Periodontics and Oral Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Z. Zhang
- Department of Cariology, Restorative Sciences, and Endodontics, School of Dentistry, University of Michigan, Ann Arbor, MI, USA
| | - N. Dubey
- Faculty of Dentistry, National University of Singapore, Singapore, Singapore
| | - W.L. Dissanayaka
- Applied Oral Sciences and Community Dental Care, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China
| | - J.E. Nör
- Department of Cariology, Restorative Sciences, and Endodontics, School of Dentistry, University of Michigan, Ann Arbor, MI, USA
- Department of Otolaryngology–Head and Neck Surgery, University of Michigan Health System, Ann Arbor, MI, USA
- Department of Biomedical Engineering, College of Engineering, University of Michigan, Ann Arbor, MI, USA
| | - M.C. Bottino
- Department of Cariology, Restorative Sciences, and Endodontics, School of Dentistry, University of Michigan, Ann Arbor, MI, USA
- Department of Biomedical Engineering, College of Engineering, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
22
|
Guo L, Li C, Gong W. Toward reproducible tumor organoid culture: focusing on primary liver cancer. Front Immunol 2024; 15:1290504. [PMID: 38571961 PMCID: PMC10987700 DOI: 10.3389/fimmu.2024.1290504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Accepted: 02/29/2024] [Indexed: 04/05/2024] Open
Abstract
Organoids present substantial potential for pushing forward preclinical research and personalized medicine by accurately recapitulating tissue and tumor heterogeneity in vitro. However, the lack of standardized protocols for cancer organoid culture has hindered reproducibility. This paper comprehensively reviews the current challenges associated with cancer organoid culture and highlights recent multidisciplinary advancements in the field with a specific focus on standardizing liver cancer organoid culture. We discuss the non-standardized aspects, including tissue sources, processing techniques, medium formulations, and matrix materials, that contribute to technical variability. Furthermore, we emphasize the need to establish reproducible platforms that accurately preserve the genetic, proteomic, morphological, and pharmacotypic features of the parent tumor. At the end of each section, our focus shifts to organoid culture standardization in primary liver cancer. By addressing these challenges, we can enhance the reproducibility and clinical translation of cancer organoid systems, enabling their potential applications in precision medicine, drug screening, and preclinical research.
Collapse
Affiliation(s)
| | | | - Weiqiang Gong
- Department of Hepatobiliary and Pancreatic Surgery, Weifang People’s Hospital, Weifang, Shandong, China
| |
Collapse
|
23
|
Sharma AD, Jarman EH, Kuppalli K, Murphy MJ, Longaker MT, Gurtner G, Fox PM. Successful topical treatment of human biofilms using multiple antibiotic elution from a collagen-rich hydrogel. Sci Rep 2024; 14:5621. [PMID: 38454046 PMCID: PMC10920629 DOI: 10.1038/s41598-024-54477-z] [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: 08/10/2023] [Accepted: 02/12/2024] [Indexed: 03/09/2024] Open
Abstract
Chronic non-healing wounds significantly strain modern healthcare systems, affecting 1-2% of the population in developed countries with costs ranging between $28.1 and $96.8 billion annually. Additionally, it has been established that chronic wounds resulting from comorbidities, such as peripheral vascular disease and diabetes mellitus, tend to be polymicrobial in nature. Treatment of polymicrobial chronic wounds with oral and IV antibiotics can result in antimicrobial resistance, leading to more difficult-to-treat wounds. Ideally, chronic ulcers would be topically treated with antibiotic combinations tailored to the microbiome of a patient's wound. We have previously shown that a topical collagen-rich hydrogel (cHG) can elute single antibiotics to inhibit bacterial growth in a manner that is nontoxic to mammalian cells. Here, we analyzed the microbiology of cultures taken from human patients diagnosed with diabetes mellitus suffering from chronic wounds present for more than 6 weeks. Additionally, we examined the safety of the elution of multiple antibiotics from collagen-rich hydrogel in mammalian cells in vivo. Finally, we aimed to create tailored combinations of antibiotics impregnated into cHG to successfully target and treat infections and eradicate biofilms cultured from human chronic diabetic wound tissue. We found that the majority of human chronic wounds in our study were polymicrobial in nature. The elution of multiple antibiotics from cHG was well-tolerated in mammalian cells, making it a potential topical treatment of the polymicrobial chronic wound. Finally, combinations of antibiotics tailored to each patient's microbiome eluted from a collagen-rich hydrogel successfully treated bacterial cultures isolated from patient samples via an in vitro assay.
Collapse
Affiliation(s)
- Ayushi D Sharma
- Division of Plastic & Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
- Division of Plastic & Reconstructive Surgery, Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, USA
- Division of Plastic & Reconstructive Surgery, Baylor Scott & White Medical Center, Temple, TX, USA
| | - Evan H Jarman
- Division of Plastic & Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
- Division of Plastic & Reconstructive Surgery, Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, USA
| | - Krutika Kuppalli
- Division of Plastic & Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Matthew J Murphy
- Division of Plastic & Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Michael T Longaker
- Division of Plastic & Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Geoffrey Gurtner
- Department of Surgery, The University of Arizona College of Medicine, Tuscon, AZ, USA
| | - Paige M Fox
- Division of Plastic & Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA.
- Division of Plastic & Reconstructive Surgery, Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, USA.
| |
Collapse
|
24
|
Cao L, Zhang Z, Yuan D, Yu M, Min J. Tissue engineering applications of recombinant human collagen: a review of recent progress. Front Bioeng Biotechnol 2024; 12:1358246. [PMID: 38419725 PMCID: PMC10900516 DOI: 10.3389/fbioe.2024.1358246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Accepted: 01/31/2024] [Indexed: 03/02/2024] Open
Abstract
With the rapid development of synthetic biology, recombinant human collagen has emerged as a cutting-edge biological material globally. Its innovative applications in the fields of material science and medicine have opened new horizons in biomedical research. Recombinant human collagen stands out as a highly promising biomaterial, playing a pivotal role in crucial areas such as wound healing, stroma regeneration, and orthopedics. However, realizing its full potential by efficiently delivering it for optimal therapeutic outcomes remains a formidable challenge. This review provides a comprehensive overview of the applications of recombinant human collagen in biomedical systems, focusing on resolving this crucial issue. Additionally, it encompasses the exploration of 3D printing technologies incorporating recombinant collagen to address some urgent clinical challenges in regenerative repair in the future. The primary aim of this review also is to spotlight the advancements in the realm of biomaterials utilizing recombinant collagen, with the intention of fostering additional innovation and making significant contributions to the enhancement of regenerative biomaterials, therapeutic methodologies, and overall patient outcomes.
Collapse
Affiliation(s)
- Lili Cao
- Department of Plastic Surgery, Zhejiang Rongjun Hospital, Jiaxing, Zhejiang, China
| | - Zhongfeng Zhang
- Department of Plastic Surgery, Zhejiang Rongjun Hospital, Jiaxing, Zhejiang, China
| | - Dan Yuan
- Department of Plastic Surgery, Zhejiang Rongjun Hospital, Jiaxing, Zhejiang, China
| | - Meiping Yu
- Department of Plastic Surgery, Zhejiang Rongjun Hospital, Jiaxing, Zhejiang, China
| | - Jie Min
- General Surgery Department, Jiaxing No.1 Hospital, Jiaxing, Zhejiang, China
| |
Collapse
|
25
|
Zhou Z, Wang J, Jiang C, Xu K, Xu T, Yu X, Fang J, Yang Y, Dai X. Advances in Hydrogels for Meniscus Tissue Engineering: A Focus on Biomaterials, Crosslinking, Therapeutic Additives. Gels 2024; 10:114. [PMID: 38391445 PMCID: PMC10887778 DOI: 10.3390/gels10020114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 01/23/2024] [Accepted: 01/26/2024] [Indexed: 02/24/2024] Open
Abstract
Meniscus tissue engineering (MTE) has emerged as a promising strategy for meniscus repair and regeneration. As versatile platforms, hydrogels have gained significant attention in this field, as they possess tunable properties that allow them to mimic native extracellular matrices and provide a suitable microenvironment. Additionally, hydrogels can be minimally invasively injected and can be adjusted to match the shape of the implant site. They can conveniently and effectively deliver bioactive additives and demonstrate good compatibility with other functional materials. These inherent qualities have made hydrogel a promising candidate for therapeutic approaches in meniscus repair and regeneration. This article provides a comprehensive review of the advancements made in the research on hydrogel application for meniscus tissue engineering. Firstly, the biomaterials and crosslinking strategies used in the formation of hydrogels are summarized and analyzed. Subsequently, the role of therapeutic additives, including cells, growth factors, and other active products, in facilitating meniscus repair and regeneration is thoroughly discussed. Furthermore, we summarize the key issues for designing hydrogels used in MTE. Finally, we conclude with the current challenges encountered by hydrogel applications and suggest potential solutions for addressing these challenges in the field of MTE. We hope this review provides a resource for researchers and practitioners interested in this field, thereby facilitating the exploration of new design possibilities.
Collapse
Affiliation(s)
- Zhuxing Zhou
- Department of Orthopedic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310000, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou 310000, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou 310000, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou 310000, China
| | - Jiajie Wang
- Department of Orthopedic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310000, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou 310000, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou 310000, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou 310000, China
| | - Chaoqian Jiang
- School of Materials and Engineering, Zhengzhou University, Zhengzhou 450001, China
| | - Kaiwang Xu
- Department of Orthopedic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310000, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou 310000, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou 310000, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou 310000, China
| | - Tengjing Xu
- Department of Orthopedic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310000, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou 310000, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou 310000, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou 310000, China
| | - Xinning Yu
- Department of Orthopedic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310000, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou 310000, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou 310000, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou 310000, China
| | - Jinghua Fang
- Department of Orthopedic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310000, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou 310000, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou 310000, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou 310000, China
| | - Yanyu Yang
- School of Materials and Engineering, Zhengzhou University, Zhengzhou 450001, China
| | - Xuesong Dai
- Department of Orthopedic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310000, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou 310000, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou 310000, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou 310000, China
| |
Collapse
|
26
|
Desai SU, Srinivasan SS, Kumbar SG, Moss IL. Hydrogel-Based Strategies for Intervertebral Disc Regeneration: Advances, Challenges and Clinical Prospects. Gels 2024; 10:62. [PMID: 38247785 PMCID: PMC10815657 DOI: 10.3390/gels10010062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 01/08/2024] [Accepted: 01/10/2024] [Indexed: 01/23/2024] Open
Abstract
Millions of people worldwide suffer from low back pain and disability associated with intervertebral disc (IVD) degeneration. IVD degeneration is highly correlated with aging, as the nucleus pulposus (NP) dehydrates and the annulus fibrosus (AF) fissures form, which often results in intervertebral disc herniation or disc space collapse and related clinical symptoms. Currently available options for treating intervertebral disc degeneration are symptoms control with therapy modalities, and/or medication, and/or surgical resection of the IVD with or without spinal fusion. As such, there is an urgent clinical demand for more effective disease-modifying treatments for this ubiquitous disorder, rather than the current paradigms focused only on symptom control. Hydrogels are unique biomaterials that have a variety of distinctive qualities, including (but not limited to) biocompatibility, highly adjustable mechanical characteristics, and most importantly, the capacity to absorb and retain water in a manner like that of native human nucleus pulposus tissue. In recent years, various hydrogels have been investigated in vitro and in vivo for the repair of intervertebral discs, some of which are ready for clinical testing. In this review, we summarize the latest findings and developments in the application of hydrogel technology for the repair and regeneration of intervertebral discs.
Collapse
Affiliation(s)
- Shivam U. Desai
- Department of Orthopedic Surgery, Central Michigan University, College of Medicine, Saginaw, MI 48602, USA
| | | | | | - Isaac L. Moss
- Department of Orthopedic Surgery, University of Connecticut, Storrs, CT 06269, USA
| |
Collapse
|
27
|
Khan MUA, Stojanović GM, Abdullah MFB, Dolatshahi-Pirouz A, Marei HE, Ashammakhi N, Hasan A. Fundamental properties of smart hydrogels for tissue engineering applications: A review. Int J Biol Macromol 2024; 254:127882. [PMID: 37951446 DOI: 10.1016/j.ijbiomac.2023.127882] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 10/22/2023] [Accepted: 11/02/2023] [Indexed: 11/14/2023]
Abstract
Tissue engineering is an advanced and potential biomedical approach to treat patients suffering from lost or failed an organ or tissue to repair and regenerate damaged tissues that increase life expectancy. The biopolymers have been used to fabricate smart hydrogels to repair damaged tissue as they imitate the extracellular matrix (ECM) with intricate structural and functional characteristics. These hydrogels offer desired and controllable qualities, such as tunable mechanical stiffness and strength, inherent adaptability and biocompatibility, swellability, and biodegradability, all crucial for tissue engineering. Smart hydrogels provide a superior cellular environment for tissue engineering, enabling the generation of cutting-edge synthetic tissues due to their special qualities, such as stimuli sensitivity and reactivity. Numerous review articles have presented the exceptional potential of hydrogels for various biomedical applications, including drug delivery, regenerative medicine, and tissue engineering. Still, it is essential to write a comprehensive review article on smart hydrogels that successfully addresses the essential challenging issues in tissue engineering. Hence, the recent development on smart hydrogel for state-of-the-art tissue engineering conferred progress, highlighting significant challenges and future perspectives. This review discusses recent advances in smart hydrogels fabricated from biological macromolecules and their use for advanced tissue engineering. It also provides critical insight, emphasizing future research directions and progress in tissue engineering.
Collapse
Affiliation(s)
- Muhammad Umar Aslam Khan
- Department of Mechanical and Industrial Engineering, Qatar University, Doha 2713, Qatar; Biomedical Research Center, Qatar University, Doha 2713, Qatar.
| | - Goran M Stojanović
- Department of Electronics, Faculty of Technical Sciences, University of Novi Sad, 21000 Novi Sad, Serbia
| | - Mohd Faizal Bin Abdullah
- Oral and Maxillofacial Surgery Unit, School of Dental Sciences, Universiti Sains Malaysia, Health Campus, 16150, Kubang Kerian, Kota Bharu, Kelantan, Malaysia; Oral and Maxillofacial Surgery Unit, Hospital Universiti Sains Malaysia, Universiti Sains Malaysia, Health Campus, 16150, Kubang Kerian, Kota Bharu, Kelantan, Malaysia.
| | | | - Hany E Marei
- Department of Cytology and Histology, Faculty of Veterinary Medicine, Mansoura University, Mansoura, Egypt
| | - Nureddin Ashammakhi
- Institute for Quantitative Health Science and Engineering (IQ), Department of Biomedical Engineering, College of Engineering and Human Medicine, Michigan State University, East Lansing, MI 48824, USA.
| | - Anwarul Hasan
- Department of Mechanical and Industrial Engineering, Qatar University, Doha 2713, Qatar; Biomedical Research Center, Qatar University, Doha 2713, Qatar
| |
Collapse
|
28
|
Jiang Z, Song Z, Cao C, Yan M, Liu Z, Cheng X, Wang H, Wang Q, Liu H, Chen S. Multiple Natural Polymers in Drug and Gene Delivery Systems. Curr Med Chem 2024; 31:1691-1715. [PMID: 36927424 DOI: 10.2174/0929867330666230316094540] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Revised: 01/29/2023] [Accepted: 02/10/2023] [Indexed: 03/18/2023]
Abstract
Natural polymers are organic compounds produced by living organisms. In nature, they exist in three main forms, including proteins, polysaccharides, and nucleic acids. In recent years, with the continuous research on drug and gene delivery systems, scholars have found that natural polymers have promising applications in drug and gene delivery systems due to their excellent properties such as biocompatibility, biodegradability, low immunogenicity, and easy modification. However, since the structure, physicochemical properties, pharmacological properties and biological characteristics of biopolymer molecules have not yet been entirely understood, further studies are required before large-scale clinical application. This review focuses on recent advances in the representative natural polymers such as proteins (albumin, collagen, elastin), polysaccharides (chitosan, alginate, cellulose) and nucleic acids. We introduce the characteristics of various types of natural polymers, and further outline the characterization methods and delivery forms of these natural polymers. Finally, we discuss possible challenges for natural polymers in subsequent experimental studies and clinical applications. It provides an important strategy for the clinical application of natural polymers in drug and gene delivery systems.
Collapse
Affiliation(s)
- Zhengfa Jiang
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, PR China
| | - Zongmian Song
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, PR China
| | - Chen Cao
- Department of Orthopedics, Zhengzhou University People's Hospital, Zhengzhou, 450003, PR China
- Department of Orthopedics, Henan Provincial People's Hospital, Zhengzhou, PR China
| | - Miaoheng Yan
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, PR China
| | - Zhendong Liu
- Department of Orthopedics, Zhengzhou University People's Hospital, Zhengzhou, 450003, PR China
- Department of Orthopedics, Henan Provincial People's Hospital, Zhengzhou, PR China
| | - Xingbo Cheng
- Department of Orthopedics, Zhengzhou University People's Hospital, Zhengzhou, 450003, PR China
- Department of Orthopedics, Henan Provincial People's Hospital, Zhengzhou, PR China
| | - Hongbo Wang
- Department of Orthopedics, Zhengzhou University People's Hospital, Zhengzhou, 450003, PR China
- Department of Orthopedics, Henan Provincial People's Hospital, Zhengzhou, PR China
| | - Qingnan Wang
- Department of Orthopedics, Zhengzhou University People's Hospital, Zhengzhou, 450003, PR China
- Department of Orthopedics, Henan Provincial People's Hospital, 450003, PR China
| | - Hongjian Liu
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, PR China
| | - Songfeng Chen
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, PR China
| |
Collapse
|
29
|
Patel DK, Jung E, Priya S, Won SY, Han SS. Recent advances in biopolymer-based hydrogels and their potential biomedical applications. Carbohydr Polym 2024; 323:121408. [PMID: 37940291 DOI: 10.1016/j.carbpol.2023.121408] [Citation(s) in RCA: 30] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 09/12/2023] [Accepted: 09/14/2023] [Indexed: 11/10/2023]
Abstract
Hydrogels are three-dimensional networks of polymer chains containing large amounts of water in their structure. Hydrogels have received significant attention in biomedical applications owing to their attractive physicochemical properties, including flexibility, softness, biodegradability, and biocompatibility. Different natural and synthetic polymers have been intensely explored in developing hydrogels for the desired applications. Biopolymers-based hydrogels have advantages over synthetic polymers regarding improved cellular activity and weak immune response. These properties can be further improved by grafting with other polymers or adding nanomaterials, and they structurally mimic the living tissue environments, which opens their broad applicability. The hydrogels can be physically or chemically cross-linked depending on the structure. The use of different biopolymers-based hydrogels in biomedical applications has been reviewed and discussed earlier. However, no report is still available to comprehensively introduce the synthesis, advantages, disadvantages, and biomedical applications of biopolymers-based hydrogels from the material point of view. Herein, we systematically overview different synthesis methods of hydrogels and provide a holistic approach to biopolymers-based hydrogels for biomedical applications, especially in bone regeneration, wound healing, drug delivery, bioimaging, and therapy. The current challenges and prospects of biopolymers-based hydrogels are highlighted rationally, giving an insight into the progress of these hydrogels and their practical applications.
Collapse
Affiliation(s)
- Dinesh K Patel
- School of Chemical Engineering, Yeungnam University, 280-Daehak-ro, Gyeongsan 38541, Republic of Korea
| | - Eunseo Jung
- School of Chemical Engineering, Yeungnam University, 280-Daehak-ro, Gyeongsan 38541, Republic of Korea
| | - Sahariya Priya
- School of Chemical Engineering, Yeungnam University, 280-Daehak-ro, Gyeongsan 38541, Republic of Korea
| | - So-Yeon Won
- School of Chemical Engineering, Yeungnam University, 280-Daehak-ro, Gyeongsan 38541, Republic of Korea
| | - Sung Soo Han
- School of Chemical Engineering, Yeungnam University, 280-Daehak-ro, Gyeongsan 38541, Republic of Korea.
| |
Collapse
|
30
|
Dobaj Štiglic A, Lackner F, Nagaraj C, Beaumont M, Bračič M, Duarte I, Kononenko V, Drobne D, Madhan B, Finšgar M, Kargl R, Stana Kleinschek K, Mohan T. 3D-Printed Collagen-Nanocellulose Hybrid Bioscaffolds with Tailored Properties for Tissue Engineering Applications. ACS APPLIED BIO MATERIALS 2023; 6:5596-5608. [PMID: 38050684 PMCID: PMC10731651 DOI: 10.1021/acsabm.3c00767] [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: 09/06/2023] [Revised: 11/16/2023] [Accepted: 11/19/2023] [Indexed: 12/06/2023]
Abstract
Hybrid collagen (Coll) bioscaffolds have emerged as a promising solution for tissue engineering (TE) and regenerative medicine. These innovative bioscaffolds combine the beneficial properties of Coll, an important structural protein of the extracellular matrix, with various other biomaterials to create platforms for long-term cell growth and tissue formation. The integration or cross-linking of Coll with other biomaterials increases mechanical strength and stability and introduces tailored biochemical and physical factors that mimic the natural tissue microenvironment. This work reports on the fabrication of chemically cross-linked hybrid bioscaffolds with enhanced properties from the combination of Coll, nanofibrillated cellulose (NFC), carboxymethylcellulose (CMC), and citric acid (CA). The bioscaffolds were prepared by 3D printing ink containing Coll-NFC-CMC-CA followed by freeze-drying, dehydrothermal treatment, and neutralization. Cross-linking through the formation of ester bonds between the polymers and CA in the bioscaffolds was achieved by exposing the bioscaffolds to elevated temperatures in the dry state. The morphology, pores/porosity, chemical composition, structure, thermal behavior, swelling, degradation, and mechanical properties of the bioscaffolds in the dry and wet states were investigated as a function of Coll concentration. The bioscaffolds showed no cytotoxicity to MG-63 human bone osteosarcoma cells as tested by different assays measuring different end points. Overall, the presented hybrid Coll bioscaffolds offer a unique combination of biocompatibility, stability, and structural support, making them valuable tools for TE.
Collapse
Affiliation(s)
- Andreja Dobaj Štiglic
- Faculty
of Mechanical Engineering, Laboratory for Characterization and Processing
of Polymers, University of Maribor, Smetanova ulica 17, 2000 Maribor, Slovenia
- Faculty
of Chemistry and Chemical Engineering, Laboratory for Analytical Chemistry
and Industrial Analysis, University of Maribor, Smetanova ulica 17, 2000 Maribor, Slovenia
| | - Florian Lackner
- Institute
of Chemistry and Technology of Biobased System (IBioSys), Graz University of Technology, Stremayrgasse 9, 8010 Graz, Austria
| | - Chandran Nagaraj
- Ludwig
Boltzmann Institute for Lung Vascular Research, Stiftingtalstrasse 24, 8010 Graz, Austria
| | - Marco Beaumont
- Department
of Chemistry, Institute of Chemistry o Renewable Resources, University of Natural Resources and Life Sciences
Vienna (BOKU), A-3430 Tulln, Austria
| | - Matej Bračič
- Faculty
of Mechanical Engineering, Laboratory for Characterization and Processing
of Polymers, University of Maribor, Smetanova ulica 17, 2000 Maribor, Slovenia
| | - Isabel Duarte
- Department
of Mechanical Engineering, Centre for Mechanical Technology and Automation
(TEMA), Intelligent Systems Associate Laboratory (LASI), University of Aveiro, 3810-193 Aveiro, Portugal
| | - Veno Kononenko
- Department
of Biology, Biotechnical Faculty, Večna pot 111, 1000 Ljubljana, Slovenia
| | - Damjana Drobne
- Department
of Biology, Biotechnical Faculty, Večna pot 111, 1000 Ljubljana, Slovenia
| | - Balaraman Madhan
- CSIR-Central
Leather Research Institute, Chennai 600 020, Tamil Nadu, India
| | - Matjaž Finšgar
- Faculty
of Chemistry and Chemical Engineering, Laboratory for Analytical Chemistry
and Industrial Analysis, University of Maribor, Smetanova ulica 17, 2000 Maribor, Slovenia
| | - Rupert Kargl
- Faculty
of Mechanical Engineering, Laboratory for Characterization and Processing
of Polymers, University of Maribor, Smetanova ulica 17, 2000 Maribor, Slovenia
- Institute
of Chemistry and Technology of Biobased System (IBioSys), Graz University of Technology, Stremayrgasse 9, 8010 Graz, Austria
| | - Karin Stana Kleinschek
- Institute
of Chemistry and Technology of Biobased System (IBioSys), Graz University of Technology, Stremayrgasse 9, 8010 Graz, Austria
- Institute
of Automation, Faculty of Electrical Engineering and Computer Science, University of Maribor, Koroska cesta 46, 2000 Maribor, Slovenia
| | - Tamilselvan Mohan
- Faculty
of Mechanical Engineering, Laboratory for Characterization and Processing
of Polymers, University of Maribor, Smetanova ulica 17, 2000 Maribor, Slovenia
- Institute
of Chemistry and Technology of Biobased System (IBioSys), Graz University of Technology, Stremayrgasse 9, 8010 Graz, Austria
| |
Collapse
|
31
|
Sinad KVG, Ebubechukwu RC, Chu CK. Recent advances in double network hydrogels based on naturally-derived polymers: synthesis, properties, and biological applications. J Mater Chem B 2023; 11:11460-11482. [PMID: 38047404 DOI: 10.1039/d3tb00773a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/05/2023]
Abstract
Hydrogels composed of naturally-derived biopolymers have garnered significant research interest due to the bioavailability and biocompatibility of starting materials. However, translating these advantages to practical use is challenged by limitations of mechanical properties and stability of the resulting materials. The development of double network (DN) hydrogels has led to greatly enhanced mechanical properties and shows promise toward broadening the applications of conventional synthetic or natural hydrogels. This review highlights recently developed protein-based and polysaccharide-based DN hydrogels. For each biopolymer, we focus on a subset of DN hydrogels centered around a theme related to synthetic design or applications. Network structures and crosslinking mechanisms that endow enhanced mechanical properties and performance to the materials are discussed. Important applications, including tissue engineering, drug delivery, bioadhesives, wound healing, and wearable sensors, that arise from the inherent properties of the natural polymer or its combination with other materials are also emphasized. Finally, we discuss ongoing challenges to stimulate the discovery of new design principles for the future of DN hydrogels based on naturally-derived polymers for biological applications.
Collapse
Affiliation(s)
| | - Ruth C Ebubechukwu
- Department of Chemistry, Lehigh University, Bethlehem, Pennsylvania, USA.
| | - Crystal K Chu
- Department of Chemistry, Lehigh University, Bethlehem, Pennsylvania, USA.
| |
Collapse
|
32
|
Nashchekina Y, Guryanov E, Lihachev A, Vaganov G, Popova E, Mikhailova N, Nashchekin A. Effect of Phytic Acid Addition on the Structure of Collagen-Hyaluronic Acid Composite Gel. Gels 2023; 9:963. [PMID: 38131949 PMCID: PMC10743047 DOI: 10.3390/gels9120963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 12/04/2023] [Accepted: 12/06/2023] [Indexed: 12/23/2023] Open
Abstract
Composite collagen gels with hyaluronic acid are developed tissue-engineered structures for filling and regeneration of defects in various organs and tissues. For the first time, phytic acid was used to increase the stability and improve the mechanical properties of collagen gels with hyaluronic acid. Phytic acid is a promising cross-linker for collagen hydrogels and is a plant-derived antioxidant found in rich sources of beans, grains, and oilseeds. Phytic acid has several benefits due to its antioxidant, anticancer, and antitumor properties. In this work, studies were carried out on the kinetics of the self-assembly of collagen molecules in the presence of phytic and hyaluronic acids. It was shown that both of these acids do not lead to collagen self-assembly. Scanning electron microscopy showed that in the presence of phytic and hyaluronic acids, the collagen fibrils had a native structure, and the FTIR method confirmed the chemical cross-links between the collagen fibrils. DSC and rheological studies demonstrated that adding the phytic acid improved the stability and modulus of elasticity of the collagen gel. The presence of hyaluronic acid in the collagen gel slightly reduced the effect of phytic acid. The presence of phytic acid in the collagen gel improved the stability of the scaffold, but, after 1 week of cultivation, slightly reduced the viability of mesenchymal stromal cells cultured in the gel. The collagen type I gel with hyaluronic and phytic acids can be used to replace tissue defects, especially after the removal of cancerous tumors.
Collapse
Affiliation(s)
- Yuliya Nashchekina
- Center of Cell Technologies, Institute of Cytology of the Russian Academy of Sciences, Tikhoretsky Pr. 4, 194064 St. Petersburg, Russia; (E.G.); (N.M.)
| | - Evgeny Guryanov
- Center of Cell Technologies, Institute of Cytology of the Russian Academy of Sciences, Tikhoretsky Pr. 4, 194064 St. Petersburg, Russia; (E.G.); (N.M.)
| | - Alexey Lihachev
- Laboratory «Characterization of Materials and Structures of Solid State Electronics», Ioffe Institute, Polytekhnicheskaya St. 26, 194021 St. Petersburg, Russia; (A.L.); (A.N.)
| | - Gleb Vaganov
- Institute of Macromolecular Compounds of Russian Academy of Sciences, V.O., Bol’shoy Pr. 31, 199004 St. Petersburg, Russia; (G.V.); (E.P.)
| | - Elena Popova
- Institute of Macromolecular Compounds of Russian Academy of Sciences, V.O., Bol’shoy Pr. 31, 199004 St. Petersburg, Russia; (G.V.); (E.P.)
| | - Natalya Mikhailova
- Center of Cell Technologies, Institute of Cytology of the Russian Academy of Sciences, Tikhoretsky Pr. 4, 194064 St. Petersburg, Russia; (E.G.); (N.M.)
| | - Alexey Nashchekin
- Laboratory «Characterization of Materials and Structures of Solid State Electronics», Ioffe Institute, Polytekhnicheskaya St. 26, 194021 St. Petersburg, Russia; (A.L.); (A.N.)
| |
Collapse
|
33
|
Ranjbar J, Yang Y, Harper AGS. Developing human tissue engineered arterial constructs to simulate human in vivo thrombus formation. Platelets 2023; 34:2153823. [PMID: 36550074 DOI: 10.1080/09537104.2022.2153823] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Thrombus formation is highly dependent upon the physico-chemical environment in which it is triggered. Our ability to understand how thrombus formation is initiated, regulated, and resolved in the human body is dependent upon our ability to replicate the mechanical and biological properties of the arterial wall. Current in vitro thrombosis models principally use reductionist approaches to model the complex biochemical and cellular milieu present in the arterial wall, and so researcher have favored the use of in vivo models. The field of vascular tissue engineering has developed a range of techniques for culturing artificial human arteries for use as vascular grafts. These techniques therefore provide a basis for developing more sophisticated 3D replicas of the arterial wall that can be used in in vitro thrombosis models. In this review, we consider how tissue engineering approaches can be used to generate 3D models of the arterial wall that improve upon current in vivo and in vitro approaches. We consider the current benefits and limitations of reported 3D tissue engineered models and consider what additional evidence is required to validate them as alternatives to current in vivo models.
Collapse
Affiliation(s)
| | - Ying Yang
- School of Pharmacy & Bioengineering, Keele University, Keele, UK
| | | |
Collapse
|
34
|
Kouthouridis S, Sotra A, Khan Z, Alvarado J, Raha S, Zhang B. Modeling the Progression of Placental Transport from Early- to Late-Stage Pregnancy by Tuning Trophoblast Differentiation and Vascularization. Adv Healthc Mater 2023; 12:e2301428. [PMID: 37830445 PMCID: PMC11468690 DOI: 10.1002/adhm.202301428] [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: 05/05/2023] [Revised: 09/18/2023] [Indexed: 10/14/2023]
Abstract
The early-stage placental barrier is characterized by a lack of fetal circulation and by a thick trophoblastic barrier, whereas the later-stage placenta consists of vascularized chorionic villi encased in a thin, differentiated trophoblast layer, ideal for nutrient transport. In this work, predictive models of early- and late-stage placental transport are created using blastocyst-derived placental stem cells (PSCs) by modulating PSC differentiation and model vascularization. PSC differentiation results in a thinner, fused trophoblast layer, as well as an increase in human chorionic gonadotropin secretion, barrier permeability, and secretion of certain inflammatory cytokines, which are consistent with in vivo findings. Further, gene expression confirms this shift toward a differentiated trophoblast subtype. Vascularization results in a molecule type- and size-dependent change in dextran and insulin permeability. These results demonstrate that trophoblast differentiation and vascularization have critical effects on placental barrier permeability and that this model can be used as a predictive measure to assess fetal toxicity of xenobiotic substances at different stages of pregnancy.
Collapse
Affiliation(s)
- Sonya Kouthouridis
- Department of Chemical EngineeringMcMaster UniversityHamiltonONL8S 4L8Canada
| | - Alexander Sotra
- School of Biomedical EngineeringMcMaster UniversityHamiltonONL8S 4L8Canada
| | - Zaim Khan
- Department of Biochemistry and Biomedical SciencesMcMaster UniversityHamiltonONL8S 4L8Canada
| | - Justin Alvarado
- Department of Biochemistry and Biomedical SciencesMcMaster UniversityHamiltonONL8S 4L8Canada
| | - Sandeep Raha
- Department of Pediatrics and the Graduate Programme in Medical SciencesMcMaster UniversityHamiltonONL8S 4L8Canada
| | - Boyang Zhang
- Department of Chemical EngineeringMcMaster UniversityHamiltonONL8S 4L8Canada
- School of Biomedical EngineeringMcMaster UniversityHamiltonONL8S 4L8Canada
| |
Collapse
|
35
|
Qian Y, Lu S, Meng J, Chen W, Li J. Thermo-Responsive Hydrogels Coupled with Photothermal Agents for Biomedical Applications. Macromol Biosci 2023; 23:e2300214. [PMID: 37526220 DOI: 10.1002/mabi.202300214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2023] [Revised: 07/04/2023] [Indexed: 08/02/2023]
Abstract
Intelligent hydrogels are materials with abilities to change their chemical nature or physical structure in response to external stimuli showing promising potential in multitudinous applications. Especially, photo-thermo coupled responsive hydrogels that are prepared by encapsulating photothermal agents into thermo-responsive hydrogel matrix exhibit more attractive advantages in biomedical applications owing to their spatiotemporal control and precise therapy. This work summarizes the latest progress of the photo-thermo coupled responsive hydrogel in biomedical applications. Three major elements of the photo-thermo coupled responsive hydrogel, i.e., thermo-responsive hydrogel matrix, photothermal agents, and construction methods are introduced. Furthermore, the recent developments of these hydrogels for biomedical applications are described with some selected examples. Finally, the challenges and future perspectives for photo-thermo coupled responsive hydrogels are outlined.
Collapse
Affiliation(s)
- Yafei Qian
- Hunan Provincial Key Laboratory of Micro & Nano Materials Interface Science, College of Chemistry and Chemical Engineering, Central South University, Changsha, 410083, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Central South University, Changsha, 410008, China
| | - Sha Lu
- Hunan Provincial Key Laboratory of Micro & Nano Materials Interface Science, College of Chemistry and Chemical Engineering, Central South University, Changsha, 410083, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Central South University, Changsha, 410008, China
| | - Jianqiang Meng
- Hunan Provincial Key Laboratory of Micro & Nano Materials Interface Science, College of Chemistry and Chemical Engineering, Central South University, Changsha, 410083, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Central South University, Changsha, 410008, China
| | - Wansong Chen
- Hunan Provincial Key Laboratory of Micro & Nano Materials Interface Science, College of Chemistry and Chemical Engineering, Central South University, Changsha, 410083, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Central South University, Changsha, 410008, China
| | - Juan Li
- Hunan Provincial Key Laboratory of Micro & Nano Materials Interface Science, College of Chemistry and Chemical Engineering, Central South University, Changsha, 410083, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Central South University, Changsha, 410008, China
| |
Collapse
|
36
|
Qiao Y, Yu L, Yang P, Chen M, Sun H, Wang L, Wu B, Oh C, Yang H, Bai J, Geng D. Spatiotemporal Immunomodulation and Biphasic Osteo-Vascular Aligned Electrospun Membrane for Diabetic Periosteum Regeneration. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2302874. [PMID: 37973554 PMCID: PMC10754081 DOI: 10.1002/advs.202302874] [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: 05/08/2023] [Revised: 07/10/2023] [Indexed: 11/19/2023]
Abstract
Under diabetic conditions, blood glucose fluctuations and exacerbated immunopathological inflammatory environments pose significant challenges to periosteal regenerative repair strategies. Responsive immune regulation in damaged tissues is critical for the immune microenvironment, osteogenesis, and angiogenesis stabilization. Considering the high-glucose microenvironment of such acute injury sites, a functional glucose-responsive immunomodulation-assisted periosteal regeneration composite material-PLA(Polylactic Acid)/COLI(Collagen I)/Lipo(Liposome)-APY29 (PCLA)-is constructed. Aside from stimulating osteogenic differentiation, owing to the presence of surface self-assembled type I collagen in the scaffolds, PCLA can directly respond to focal area high-glucose microenvironments. The PCLA scaffolds trigger the release of APY29-loaded liposomes, shifting the macrophages toward the M2 phenotype, inhibiting the release of inflammatory cytokines, improving the bone immune microenvironment, and promoting osteogenic differentiation and angiogenesis. Bioinformatics analyses show that PCLA enhances bone repair by inhibiting the inflammatory signal pathway regulating the polarization direction and promoting osteogenic and angiogenic gene expression. In the calvarial periosteal defect model of diabetic rats, PCLA scaffolds induce M2 macrophage polarization and improve the inflammatory microenvironment, significantly accelerating periosteal repair. Overall, the PCLA scaffold material regulates immunity in fluctuating high-glucose inflammatory microenvironments, achieves relatively stable and favorable osteogenic microenvironments, and facilitates the effective design of functionalized biomaterials for bone regeneration therapy in patients with diabetes.
Collapse
Affiliation(s)
- Yusen Qiao
- Department of OrthopedicsThe First Affiliated Hospital of Soochow University188 Shizi RoadSuzhouJiangsu215006China
- Department of Orthopedic SurgeryRush University Medical CenterChicagoIL60612USA
| | - Lei Yu
- Department of OrthopedicsThe First Affiliated Hospital of Soochow University188 Shizi RoadSuzhouJiangsu215006China
| | - Peng Yang
- Department of OrthopedicsThe First Affiliated Hospital of Soochow University188 Shizi RoadSuzhouJiangsu215006China
| | - Miao Chen
- Department of OrthopedicsThe First Affiliated Hospital of Soochow University188 Shizi RoadSuzhouJiangsu215006China
| | - Haifu Sun
- Department of OrthopedicsThe First Affiliated Hospital of Soochow University188 Shizi RoadSuzhouJiangsu215006China
| | - Lingjie Wang
- Department of OrthopedicsThe First Affiliated Hospital of Soochow University188 Shizi RoadSuzhouJiangsu215006China
| | - Bangzhao Wu
- Department of OrthopedicsThe First Affiliated Hospital of Soochow University188 Shizi RoadSuzhouJiangsu215006China
| | - Chun‐do Oh
- Department of Orthopedic SurgeryRush University Medical CenterChicagoIL60612USA
| | - Huilin Yang
- Department of OrthopedicsThe First Affiliated Hospital of Soochow University188 Shizi RoadSuzhouJiangsu215006China
| | - Jiaxiang Bai
- Department of OrthopedicsThe First Affiliated Hospital of Soochow University188 Shizi RoadSuzhouJiangsu215006China
- Department of Orthopedics, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefei230022China
- National Center for Translational Medicine (Shanghai) SHU BranchShanghai UniversityShanghaiChina
| | - Dechun Geng
- Department of OrthopedicsThe First Affiliated Hospital of Soochow University188 Shizi RoadSuzhouJiangsu215006China
| |
Collapse
|
37
|
Xu P, Kankala RK, Wang S, Chen A. Decellularized extracellular matrix-based composite scaffolds for tissue engineering and regenerative medicine. Regen Biomater 2023; 11:rbad107. [PMID: 38173774 PMCID: PMC10761212 DOI: 10.1093/rb/rbad107] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 10/17/2023] [Accepted: 11/28/2023] [Indexed: 01/05/2024] Open
Abstract
Despite the considerable advancements in fabricating polymeric-based scaffolds for tissue engineering, the clinical transformation of these scaffolds remained a big challenge because of the difficulty of simulating native organs/tissues' microenvironment. As a kind of natural tissue-derived biomaterials, decellularized extracellular matrix (dECM)-based scaffolds have gained attention due to their unique biomimetic properties, providing a specific microenvironment suitable for promoting cell proliferation, migration, attachment and regulating differentiation. The medical applications of dECM-based scaffolds have addressed critical challenges, including poor mechanical strength and insufficient stability. For promoting the reconstruction of damaged tissues or organs, different types of dECM-based composite platforms have been designed to mimic tissue microenvironment, including by integrating with natural polymer or/and syntenic polymer or adding bioactive factors. In this review, we summarized the research progress of dECM-based composite scaffolds in regenerative medicine, highlighting the critical challenges and future perspectives related to the medical application of these composite materials.
Collapse
Affiliation(s)
- Peiyao Xu
- Institute of Biomaterials and Tissue Engineering, Huaqiao University, Xiamen, Fujian 361021, PR China
- Fujian Provincial Key Laboratory of Biochemical Technology (Huaqiao University), Xiamen, Fujian 361021, PR China
| | - Ranjith Kumar Kankala
- Institute of Biomaterials and Tissue Engineering, Huaqiao University, Xiamen, Fujian 361021, PR China
- Fujian Provincial Key Laboratory of Biochemical Technology (Huaqiao University), Xiamen, Fujian 361021, PR China
| | - Shibin Wang
- Institute of Biomaterials and Tissue Engineering, Huaqiao University, Xiamen, Fujian 361021, PR China
- Fujian Provincial Key Laboratory of Biochemical Technology (Huaqiao University), Xiamen, Fujian 361021, PR China
| | - Aizheng Chen
- Institute of Biomaterials and Tissue Engineering, Huaqiao University, Xiamen, Fujian 361021, PR China
- Fujian Provincial Key Laboratory of Biochemical Technology (Huaqiao University), Xiamen, Fujian 361021, PR China
| |
Collapse
|
38
|
Zagoskin YD, Sergeeva YE, Fomina YS, Sukhinov DV, Malakhov SN, Osidak EO, Khramtsova EA, Gotovtsev PM, Chvalun SN, Grigoriev TE. Porous Polylactide Microparticles as Effective Fillers for Hydrogels. Biomimetics (Basel) 2023; 8:565. [PMID: 38132504 PMCID: PMC10741550 DOI: 10.3390/biomimetics8080565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 09/25/2023] [Accepted: 11/17/2023] [Indexed: 12/23/2023] Open
Abstract
High-strength composite hydrogels based on collagen or chitosan-genipin were obtained via mixing using highly porous polylactide (PLA) microparticles with diameters of 50-75 µm and porosity values of over 98%. The elastic modulus of hydrogels depended on the filler concentration. The modulus increased from 80 kPa to 400-600 kPa at a concentration of porous particles of 12-15 wt.% and up to 1.8 MPa at a filling of 20-25 wt.% for collagen hydrogels. The elastic modulus of the chitosan-genipin hydrogel increases from 75 kPa to 900 kPa at a fraction of particles of 20 wt.%. These elastic modulus values cover a range of strength properties from connective tissue to cartilage tissue. It is important to note that the increase in strength in this case is accompanied by a decrease in the density of the material, that is, an increase in porosity. PLA particles were loaded with C-phycocyanin and showed an advanced release profile up to 48 h. Thus, composite hydrogels mimic the structure, biomechanics and release of biomolecules in the tissues of a living organism.
Collapse
Affiliation(s)
- Yuriy D. Zagoskin
- National Research Centre “Kurchatov Institute”, 123182 Moscow, Russia; (Y.D.Z.); (Y.E.S.); (Y.S.F.); (D.V.S.); (S.N.M.); (S.N.C.); (T.E.G.)
| | - Yana E. Sergeeva
- National Research Centre “Kurchatov Institute”, 123182 Moscow, Russia; (Y.D.Z.); (Y.E.S.); (Y.S.F.); (D.V.S.); (S.N.M.); (S.N.C.); (T.E.G.)
- Moscow Institute of Physics and Technology, National Research University, 141700 Dolgoprudny, Russia
| | - Yuliya S. Fomina
- National Research Centre “Kurchatov Institute”, 123182 Moscow, Russia; (Y.D.Z.); (Y.E.S.); (Y.S.F.); (D.V.S.); (S.N.M.); (S.N.C.); (T.E.G.)
| | - Daniil V. Sukhinov
- National Research Centre “Kurchatov Institute”, 123182 Moscow, Russia; (Y.D.Z.); (Y.E.S.); (Y.S.F.); (D.V.S.); (S.N.M.); (S.N.C.); (T.E.G.)
| | - Sergey N. Malakhov
- National Research Centre “Kurchatov Institute”, 123182 Moscow, Russia; (Y.D.Z.); (Y.E.S.); (Y.S.F.); (D.V.S.); (S.N.M.); (S.N.C.); (T.E.G.)
| | - Egor O. Osidak
- Imtek Ltd., 121552 Moscow, Russia;
- Dmitry Rogachev National Medical Research Center of Paediatric Haematology, Oncology and Immunology, 117198 Moscow, Russia
| | - Elena A. Khramtsova
- N.M. Emanuel Institute of Biochemical Physics, Russian Academy of Sciences, 119334 Moscow, Russia;
| | - Pavel M. Gotovtsev
- National Research Centre “Kurchatov Institute”, 123182 Moscow, Russia; (Y.D.Z.); (Y.E.S.); (Y.S.F.); (D.V.S.); (S.N.M.); (S.N.C.); (T.E.G.)
- Moscow Institute of Physics and Technology, National Research University, 141700 Dolgoprudny, Russia
| | - Sergei N. Chvalun
- National Research Centre “Kurchatov Institute”, 123182 Moscow, Russia; (Y.D.Z.); (Y.E.S.); (Y.S.F.); (D.V.S.); (S.N.M.); (S.N.C.); (T.E.G.)
| | - Timofei E. Grigoriev
- National Research Centre “Kurchatov Institute”, 123182 Moscow, Russia; (Y.D.Z.); (Y.E.S.); (Y.S.F.); (D.V.S.); (S.N.M.); (S.N.C.); (T.E.G.)
- Moscow Institute of Physics and Technology, National Research University, 141700 Dolgoprudny, Russia
| |
Collapse
|
39
|
Orieshyna A, Puetzer JL, Amdursky N. Proton Transport Across Collagen Fibrils and Scaffolds: The Role of Hydroxyproline. Biomacromolecules 2023; 24:4653-4662. [PMID: 37656903 DOI: 10.1021/acs.biomac.3c00326] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/03/2023]
Abstract
Collagen is one of the most studied proteins due to its fundamental role in creating fibrillar structures and supporting tissues in our bodies. Accordingly, collagen is also one of the most used proteins for making tissue-engineered scaffolds for various types of tissues. To date, the high abundance of hydroxyproline (Hyp) within collagen is commonly ascribed to the structure and stability of collagen. Here, we hypothesize a new role for the presence of Hyp within collagen, which is to support proton transport (PT) across collagen fibrils. For this purpose, we explore here three different collagen-based hydrogels: the first is prepared by the self-assembly of natural collagen fibrils, and the second and third are based on covalently linking between collagen via either a self-coupling method or with an additional cross-linker. Following the formation of the hydrogel, we introduce here a two-step reaction, involving (1) attaching methanesulfonyl to the -OH group of Hyp, followed by (2) removing the methanesulfonyl, thus reverting Hyp to proline (Pro). We explore the PT efficiency at each step of the reaction using electrical measurements and show that adding the methanesulfonyl group vastly enhances PT, while reverting Hyp to Pro significantly reduces PT efficiency (compared with the initial point) with different efficiencies for the various collagen-based hydrogels. The role of Hyp in supporting the PT can assist in our understanding of the physiological roles of collagen. Furthermore, the capacity to modulate conductivity across collagen is very important to the use of collagen in regenerative medicine.
Collapse
Affiliation(s)
- Anna Orieshyna
- Schulich Faculty of Chemistry, Technion - Israel Institute of Technology, Haifa 3200003, Israel
| | - Jennifer L Puetzer
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, Virginia 23220, United States
| | - Nadav Amdursky
- Schulich Faculty of Chemistry, Technion - Israel Institute of Technology, Haifa 3200003, Israel
| |
Collapse
|
40
|
Zielinska D, Fisch P, Moehrlen U, Finkielsztein S, Linder T, Zenobi-Wong M, Biedermann T, Klar AS. Combining bioengineered human skin with bioprinted cartilage for ear reconstruction. SCIENCE ADVANCES 2023; 9:eadh1890. [PMID: 37792948 PMCID: PMC10550230 DOI: 10.1126/sciadv.adh1890] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 09/01/2023] [Indexed: 10/06/2023]
Abstract
Microtia is a congenital disorder that manifests as a malformation of the external ear leading to psychosocial problems in affected children. Here, we present a tissue-engineered treatment approach based on a bioprinted autologous auricular cartilage construct (EarCartilage) combined with a bioengineered human pigmented and prevascularized dermo-epidermal skin substitute (EarSkin) tested in immunocompromised rats. We confirmed that human-engineered blood capillaries of EarSkin connected to the recipient's vasculature within 1 week, enabling rapid blood perfusion and epidermal maturation. Bioengineered EarSkin displayed a stratified epidermis containing mature keratinocytes and melanocytes. The latter resided within the basal layer of the epidermis and efficiently restored the skin color. Further, in vivo tests demonstrated favorable mechanical stability of EarCartilage along with enhanced extracellular matrix deposition. In conclusion, EarCartilage combined with EarSkin represents a novel approach for the treatment of microtia with the potential to circumvent existing limitations and improve the aesthetic outcome of microtia reconstruction.
Collapse
Affiliation(s)
- Dominika Zielinska
- Tissue Biology Research Unit, University Children’s Hospital Zurich, University of Zurich, Zurich, Switzerland
- Children’s Research Center, University Children’s Hospital Zurich, Zurich, Switzerland
- University of Zurich, Zurich, Switzerland
| | - Philipp Fisch
- Tissue Engineering and Biofabrication Laboratory, Department of Health Sciences and Technology, ETH Zurich, Switzerland
| | - Ueli Moehrlen
- Tissue Biology Research Unit, University Children’s Hospital Zurich, University of Zurich, Zurich, Switzerland
- Children’s Research Center, University Children’s Hospital Zurich, Zurich, Switzerland
- University of Zurich, Zurich, Switzerland
| | | | - Thomas Linder
- Klinik für Hals-, Nasen-, Ohren- und Gesichtschirurgie, Luzerner Kantonsspital, Luzern, Switzerland
| | - Marcy Zenobi-Wong
- Tissue Engineering and Biofabrication Laboratory, Department of Health Sciences and Technology, ETH Zurich, Switzerland
| | - Thomas Biedermann
- Tissue Biology Research Unit, University Children’s Hospital Zurich, University of Zurich, Zurich, Switzerland
- Children’s Research Center, University Children’s Hospital Zurich, Zurich, Switzerland
- University of Zurich, Zurich, Switzerland
| | - Agnes S. Klar
- Tissue Biology Research Unit, University Children’s Hospital Zurich, University of Zurich, Zurich, Switzerland
- Children’s Research Center, University Children’s Hospital Zurich, Zurich, Switzerland
- University of Zurich, Zurich, Switzerland
| |
Collapse
|
41
|
Liu J, Wu W, Zhu Q, Zhu H. Hydrogel-Based Therapeutics for Pancreatic Ductal Adenocarcinoma Treatment. Pharmaceutics 2023; 15:2421. [PMID: 37896181 PMCID: PMC10610350 DOI: 10.3390/pharmaceutics15102421] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 09/20/2023] [Accepted: 09/28/2023] [Indexed: 10/29/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC), one of the deadliest malignancies worldwide, is characteristic of the tumor microenvironments (TME) comprising numerous fibroblasts and immunosuppressive cells. Conventional therapies for PDAC are often restricted by limited drug delivery efficiency, immunosuppressive TME, and adverse effects. Thus, effective and safe therapeutics are urgently required for PDAC treatment. In recent years, hydrogels, with their excellent biocompatibility, high drug load capacity, and sustainable release profiles, have been developed as effective drug-delivery systems, offering potential therapeutic options for PDAC. This review summarizes the distinctive features of the immunosuppressive TME of PDAC and discusses the application of hydrogel-based therapies in PDAC, with a focus on how these hydrogels remodel the TME and deliver different types of cargoes in a controlled manner. Furthermore, we also discuss potential drug candidates and the challenges and prospects for hydrogel-based therapeutics for PDAC. By providing a comprehensive overview of hydrogel-based therapeutics for PDAC treatment, this review seeks to serve as a reference for researchers and clinicians involved in developing therapeutic strategies targeting the PDAC microenvironment.
Collapse
Affiliation(s)
- Jinlu Liu
- Division of Abdominal Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China; (J.L.); (Q.Z.)
| | - Wenbi Wu
- Department of Biotherapy, Cancer Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China;
| | - Qing Zhu
- Division of Abdominal Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China; (J.L.); (Q.Z.)
| | - Hong Zhu
- Division of Abdominal Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China; (J.L.); (Q.Z.)
| |
Collapse
|
42
|
Cheng Q, Zhang L, Zhang J, Zhou X, Wu B, Wang D, Wei T, Shafiq M, Li S, Zhi D, Guan Y, Wang K, Kong D. Decellularized Scaffolds with Double-Layer Aligned Microchannels Induce the Oriented Growth of Bladder Smooth Muscle Cells: Toward Urethral and Ureteral Reconstruction. Adv Healthc Mater 2023; 12:e2300544. [PMID: 37638600 DOI: 10.1002/adhm.202300544] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 06/27/2023] [Indexed: 08/29/2023]
Abstract
There is a great clinical need for regenerating urinary tissue. Native urethras and ureters have bidirectional aligned smooth muscle cells (SMCs) layers, which plays a pivotal role in micturition and transporting urine and inhibiting reflux. Thus far, urinary scaffolds have not been designed to induce the native-mimicking aligned arrangement of SMCs. In this study, a tubular decellularized extracellular matrix (dECM) with an intact internal layer and bidirectional aligned microchannels in the tubular wall, which is realized by the subcutaneous implantation of a template, followed by the removal of the template, and decellularization, is engineered. The dense and intact internal layer effectively increases the leakage pressure of the tubular dECM scaffolds. Rat-derived dECM scaffolds with three different sizes of microchannels are fabricated by tailoring the fiber diameter of the templates. The rat-derived dECM scaffolds exhibiting microchannels of ≈65 µm show suitable mechanical properties, good ability to induce the bidirectional alignment and growth of human bladder SMCs, and elevated higher functional protein expression in vitro. These data indicate that rat-derived tubular dECM scaffolds manifesting double-layer aligned microchannels may be promising candidates to induce the native-mimicking regeneration of SMCs in urethra and ureter reconstruction.
Collapse
Affiliation(s)
- Quhan Cheng
- Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Linli Zhang
- Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Jingai Zhang
- Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Xin Zhou
- Department of Medical Imaging, Shanxi Medical University, Taiyuan, 030001, China
| | - Boyu Wu
- Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Dezheng Wang
- Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Tingting Wei
- Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Muhammad Shafiq
- Department of Chemical Engineering, Faculty of Engineering, Graduate School, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka, 819-0395, Japan
| | - Shengbin Li
- Department of Urology, Tianjin Children's Hospital/Tianjin University Children's Hospital, Tianjin, 300134, China
| | - Dengke Zhi
- Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Yong Guan
- Department of Urology, Tianjin Children's Hospital/Tianjin University Children's Hospital, Tianjin, 300134, China
| | - Kai Wang
- Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Deling Kong
- Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Sciences, Nankai University, Tianjin, 300071, China
| |
Collapse
|
43
|
Malandain N, Sanz-Fraile H, Farré R, Otero J, Roig A, Laromaine A. Cell-Laden 3D Hydrogels of Type I Collagen Incorporating Bacterial Nanocellulose Fibers. ACS APPLIED BIO MATERIALS 2023; 6:3638-3647. [PMID: 37669535 PMCID: PMC10521014 DOI: 10.1021/acsabm.3c00126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 08/08/2023] [Indexed: 09/07/2023]
Abstract
There is a growing interest in developing natural hydrogel-based scaffolds to culture cells in a three-dimensional (3D) millieu that better mimics the in vivo cells' microenvironment. A promising approach is to use hydrogels from animal tissues, such as decellularized extracellular matrices; however, they usually exhibit suboptimal mechanical properties compared to native tissue and their composition with hundreds of different protein complicates to elucidate which stimulus triggers cell's responses. As simpler scaffolds, type I collagen hydrogels are used to study cell behavior in mechanobiology even though they are also softer than native tissues. In this work, type I collagen is mixed with bacterial nanocellulose fibers (BCf) to develop reinforced scaffolds with mechanical properties suitable for 3D cell culture. BCf were produced from blended pellicles biosynthesized from Komagataeibacter xylinus. Then, BCf were mixed with concentrated collagen from rat-tail tendons to form composite hydrogels. Confocal laser scanning microscopy and scanning electron microscopy images confirmed the homogeneous macro- and microdistribution of both natural polymers. Porosity analysis confirmed that BCf do not disrupt the scaffold structure. Tensile strength and rheology measurements demonstrated the reinforcement action of BCf (43% increased stiffness) compared to the collagen hydrogel while maintaining the same viscoelastic response. Additionally, this reinforcement of collagen hydrogels with BCf offers the possibility to mix cells before gelation and then proceed to the culture of the 3D cell scaffolds. We obtained scaffolds with human bone marrow-derived mesenchymal stromal cells or human fibroblasts within the composite hydrogels, allowing a homogeneous 3D viable culture for at least 7 days. A smaller surface shrinkage in the reinforced hydrogels compared to type I collagen hydrogels confirmed the strengthening of the composite hydrogels. These collagen hydrogels reinforced with BCf might emerge as a promising platform for 3D in vitro organ modeling, tissue-engineering applications, and suitable to conduct fundamental mechanobiology studies.
Collapse
Affiliation(s)
- Nanthilde Malandain
- Institut
de Ciència de Materials de Barcelona (ICMAB-CSIC), Campus UAB, 08193 Bellaterra, Spain
- Unitat
de Biofísica i Bioenginyeria, Facultat de Medicina i Ciències
de la Salut, Universitat de Barcelona, 08036 Barcelona, Spain
| | - Hector Sanz-Fraile
- Unitat
de Biofísica i Bioenginyeria, Facultat de Medicina i Ciències
de la Salut, Universitat de Barcelona, 08036 Barcelona, Spain
| | - Ramon Farré
- Unitat
de Biofísica i Bioenginyeria, Facultat de Medicina i Ciències
de la Salut, Universitat de Barcelona, 08036 Barcelona, Spain
- CIBER
de Enfermedades Respiratorias, 28029 Madrid, Spain
- Institut
d’Investigacions Biomèdiques August Pi i Sunyer, 08036 Barcelona, Spain
| | - Jorge Otero
- Unitat
de Biofísica i Bioenginyeria, Facultat de Medicina i Ciències
de la Salut, Universitat de Barcelona, 08036 Barcelona, Spain
- CIBER
de Enfermedades Respiratorias, 28029 Madrid, Spain
- The
Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), 08028 Barcelona, Spain
| | - Anna Roig
- Institut
de Ciència de Materials de Barcelona (ICMAB-CSIC), Campus UAB, 08193 Bellaterra, Spain
| | - Anna Laromaine
- Institut
de Ciència de Materials de Barcelona (ICMAB-CSIC), Campus UAB, 08193 Bellaterra, Spain
| |
Collapse
|
44
|
Bokhari N, Yasmeen A, Ali A, Khalid H, Wang R, Bashir M, Sharif F. Silk Meshes Coated with Chitosan-Bioactive Phytochemicals Activate Wound Healing Genes In Vitro. Macromol Biosci 2023; 23:e2300039. [PMID: 37203244 DOI: 10.1002/mabi.202300039] [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: 02/03/2023] [Revised: 04/15/2023] [Indexed: 05/20/2023]
Abstract
Meshes from natural silk are hand knitted and surface functionalized to facilitate hernia repair and other load bearing, tissue applications. Purified organic silk is - hand knitted and then coated with chitosan (CH)/bacterial cellulose (BC) blend polymer using four phytochemicals such as pomegranate (PG) peel, Nigella sativa (NS) seed, Licorice root (LE), and Bearberry leaf extracts (BE) separately. Characterizations using GCMS analysis shows the presence of bioactive chemicals in the extracts. Scanning electron microcopy (SEM) shows that the surface is coated with the composite polymer t. Fourier transform infrared spectroscopy (FTIR) shows significant elements found in CH, BC, and phytochemicals in plant extracts with no chemical changes. Tensile strength of the coated meshes is higher to support tissue as implants. The release kinetics suggest sustained release of phytochemical extracts. In vitro studies confirmed the noncytotoxic, biocompatible, wound healing potential of the meshes. Furthermore, gene expression analysis of 3-wound healing genes shows marked increase in the in vitro cell cultures due to the presence of extracts. These results suggest that the composite meshes can efficiently support hernia closure while facilitating wound/tissue healing and combating bacterial infections. Therefore, these meshes can be good candidates for fistula and cleft palate repair.
Collapse
Affiliation(s)
- Natasha Bokhari
- Interdisciplinary Research Centre in Biomedical Materials, COMSATS University Islamabad, Lahore Campus, Lahore, 54000, Pakistan
- Department of Chemistry, Lahore College for Women University, Lahore, 54000, Pakistan
| | - Abida Yasmeen
- Department of Chemistry, Lahore College for Women University, Lahore, 54000, Pakistan
| | - Asif Ali
- Interdisciplinary Research Centre in Biomedical Materials, COMSATS University Islamabad, Lahore Campus, Lahore, 54000, Pakistan
- Research Unit Plasma Technology (RUPT), Department of Applied Physics, Faculty of Engineering and Architecture, Ghent University, Ghent, 9000, Belgium
| | - Hamad Khalid
- Interdisciplinary Research Centre in Biomedical Materials, COMSATS University Islamabad, Lahore Campus, Lahore, 54000, Pakistan
| | - Rong Wang
- Biomedical Polymer Research Group, Cixi Institute of, Biomedical Engineering, Ningbo Institute of Materials Technology & Engineering, Chinese Academy of Sciences, No. 99 Xuelin Road, Cixi, Ningbo, 315000, China
| | - Mustehsan Bashir
- Department of Plastic, Reconstructive Surgery and Burn Unit, King Edward Medical University, Lahore, 54000, Pakistan
| | - Faiza Sharif
- Interdisciplinary Research Centre in Biomedical Materials, COMSATS University Islamabad, Lahore Campus, Lahore, 54000, Pakistan
| |
Collapse
|
45
|
Rana D, Desai N, Salave S, Karunakaran B, Giri J, Benival D, Gorantla S, Kommineni N. Collagen-Based Hydrogels for the Eye: A Comprehensive Review. Gels 2023; 9:643. [PMID: 37623098 PMCID: PMC10454301 DOI: 10.3390/gels9080643] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 08/01/2023] [Accepted: 08/04/2023] [Indexed: 08/26/2023] Open
Abstract
Collagen-based hydrogels have emerged as a highly promising platform for diverse applications in ophthalmology, spanning from drug delivery systems to biomedical interventions. This review explores the diverse sources of collagen, which give rise to different types of collagen protein. The critical isolation and purification steps are discussed, emphasizing their pivotal role in preparing collagen for biomedical use. To ensure collagen quality and purity, and the suitability of collagen for targeted applications, a comprehensive characterization and quality control are essential, encompassing assessments of its physical, chemical, and biological properties. Also, various cross-linking collagen methods have been examined for providing insight into this crucial process. This comprehensive review delves into every facet of collagen and explores the wide-ranging applications of collagen-based hydrogels, with a particular emphasis on their use in drug delivery systems and their potential in diverse biomedical interventions. By consolidating current knowledge and advancements in the field, this review aims to provide a detailed overview of the utilization of engineered collagen-based hydrogels in ocular therapeutics.
Collapse
Affiliation(s)
- Dhwani Rana
- National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad 382355, Gujarat, India; (D.R.); (S.S.); (B.K.); (D.B.)
| | - Nimeet Desai
- Department of Biomedical Engineering, Indian Institute of Technology Hyderabad, Kandi 502285, Telangana, India; (N.D.); (J.G.)
| | - Sagar Salave
- National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad 382355, Gujarat, India; (D.R.); (S.S.); (B.K.); (D.B.)
| | - Bharathi Karunakaran
- National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad 382355, Gujarat, India; (D.R.); (S.S.); (B.K.); (D.B.)
| | - Jyotsnendu Giri
- Department of Biomedical Engineering, Indian Institute of Technology Hyderabad, Kandi 502285, Telangana, India; (N.D.); (J.G.)
| | - Derajram Benival
- National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad 382355, Gujarat, India; (D.R.); (S.S.); (B.K.); (D.B.)
| | - Srividya Gorantla
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155, USA;
| | | |
Collapse
|
46
|
Kutluk H, Bastounis EE, Constantinou I. Integration of Extracellular Matrices into Organ-on-Chip Systems. Adv Healthc Mater 2023; 12:e2203256. [PMID: 37018430 PMCID: PMC11468608 DOI: 10.1002/adhm.202203256] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 03/20/2023] [Indexed: 04/07/2023]
Abstract
The extracellular matrix (ECM) is a complex, dynamic network present within all tissues and organs that not only acts as a mechanical support and anchorage point but can also direct fundamental cell behavior, function, and characteristics. Although the importance of the ECM is well established, the integration of well-controlled ECMs into Organ-on-Chip (OoC) platforms remains challenging and the methods to modulate and assess ECM properties on OoCs remain underdeveloped. In this review, current state-of-the-art design and assessment of in vitro ECM environments is discussed with a focus on their integration into OoCs. Among other things, synthetic and natural hydrogels, as well as polydimethylsiloxane (PDMS) used as substrates, coatings, or cell culture membranes are reviewed in terms of their ability to mimic the native ECM and their accessibility for characterization. The intricate interplay among materials, OoC architecture, and ECM characterization is critically discussed as it significantly complicates the design of ECM-related studies, comparability between works, and reproducibility that can be achieved across research laboratories. Improving the biomimetic nature of OoCs by integrating properly considered ECMs would contribute to their further adoption as replacements for animal models, and precisely tailored ECM properties would promote the use of OoCs in mechanobiology.
Collapse
Affiliation(s)
- Hazal Kutluk
- Institute of Microtechnology (IMT)Technical University of BraunschweigAlte Salzdahlumer Str. 20338124BraunschweigGermany
- Center of Pharmaceutical Engineering (PVZ)Technical University of BraunschweigFranz‐Liszt‐Str. 35a38106BraunschweigGermany
| | - Effie E. Bastounis
- Institute of Microbiology and Infection Medicine (IMIT)Eberhard Karls University of TübingenAuf der Morgenstelle 28, E872076TübingenGermany
- Cluster of Excellence “Controlling Microbes to Fight Infections” EXC 2124Eberhard Karls University of TübingenAuf der Morgenstelle 2872076TübingenGermany
| | - Iordania Constantinou
- Institute of Microtechnology (IMT)Technical University of BraunschweigAlte Salzdahlumer Str. 20338124BraunschweigGermany
- Center of Pharmaceutical Engineering (PVZ)Technical University of BraunschweigFranz‐Liszt‐Str. 35a38106BraunschweigGermany
| |
Collapse
|
47
|
Sun Z, Zhao J, Leung E, Flandes-Iparraguirre M, Vernon M, Silberstein J, De-Juan-Pardo EM, Jansen S. Three-Dimensional Bioprinting in Cardiovascular Disease: Current Status and Future Directions. Biomolecules 2023; 13:1180. [PMID: 37627245 PMCID: PMC10452258 DOI: 10.3390/biom13081180] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 07/24/2023] [Accepted: 07/26/2023] [Indexed: 08/27/2023] Open
Abstract
Three-dimensional (3D) printing plays an important role in cardiovascular disease through the use of personalised models that replicate the normal anatomy and its pathology with high accuracy and reliability. While 3D printed heart and vascular models have been shown to improve medical education, preoperative planning and simulation of cardiac procedures, as well as to enhance communication with patients, 3D bioprinting represents a potential advancement of 3D printing technology by allowing the printing of cellular or biological components, functional tissues and organs that can be used in a variety of applications in cardiovascular disease. Recent advances in bioprinting technology have shown the ability to support vascularisation of large-scale constructs with enhanced biocompatibility and structural stability, thus creating opportunities to replace damaged tissues or organs. In this review, we provide an overview of the use of 3D bioprinting in cardiovascular disease with a focus on technologies and applications in cardiac tissues, vascular constructs and grafts, heart valves and myocardium. Limitations and future research directions are highlighted.
Collapse
Affiliation(s)
- Zhonghua Sun
- Discipline of Medical Radiation Science, Curtin Medical School, Curtin University, Perth, WA 6102, Australia;
- Curtin Health Innovation Research Institute (CHIRI), Curtin University, Perth, WA 6102, Australia
| | - Jack Zhao
- School of Medicine, Faculty of Health Sciences, The University of Western Australia, Perth, WA 6009, Australia; (J.Z.); (E.L.)
| | - Emily Leung
- School of Medicine, Faculty of Health Sciences, The University of Western Australia, Perth, WA 6009, Australia; (J.Z.); (E.L.)
| | - Maria Flandes-Iparraguirre
- Regenerative Medicine Program, Cima Universidad de Navarra, 31008 Pamplona, Spain;
- T3mPLATE, Harry Perkins Institute of Medical Research, QEII Medical Centre and UWA Centre for Medical Research, The University of Western Australia, Perth, WA 6009, Australia; (M.V.); (E.M.D.-J.-P.)
- School of Engineering, The University of Western Australia, Perth, WA 6009, Australia
| | - Michael Vernon
- T3mPLATE, Harry Perkins Institute of Medical Research, QEII Medical Centre and UWA Centre for Medical Research, The University of Western Australia, Perth, WA 6009, Australia; (M.V.); (E.M.D.-J.-P.)
- School of Engineering, The University of Western Australia, Perth, WA 6009, Australia
- Vascular Engineering Laboratory, Harry Perkins Institute of Medical Research, QEII Medical Centre and UWA Centre for Medical Research, The University of Western Australia, Perth, WA 6009, Australia
| | - Jenna Silberstein
- Discipline of Medical Radiation Science, Curtin Medical School, Curtin University, Perth, WA 6102, Australia;
| | - Elena M. De-Juan-Pardo
- T3mPLATE, Harry Perkins Institute of Medical Research, QEII Medical Centre and UWA Centre for Medical Research, The University of Western Australia, Perth, WA 6009, Australia; (M.V.); (E.M.D.-J.-P.)
- School of Engineering, The University of Western Australia, Perth, WA 6009, Australia
- Curtin Medical School, Curtin University, Perth, WA 6102, Australia;
| | - Shirley Jansen
- Curtin Medical School, Curtin University, Perth, WA 6102, Australia;
- Department of Vascular and Endovascular Surgery, Sir Charles Gairdner Hospital, Perth, WA 6009, Australia
- Heart and Vascular Research Institute, Harry Perkins Medical Research Institute, Perth, WA 6009, Australia
- School of Medicine, The University of Western Australia, Perth, WA 6009, Australia
| |
Collapse
|
48
|
Sanchez‐Rubio A, Jayawarna V, Maxwell E, Dalby MJ, Salmeron‐Sanchez M. Keeping It Organized: Multicompartment Constructs to Mimic Tissue Heterogeneity. Adv Healthc Mater 2023; 12:e2202110. [PMID: 36938891 PMCID: PMC11469230 DOI: 10.1002/adhm.202202110] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2022] [Revised: 02/17/2023] [Indexed: 03/21/2023]
Abstract
Tissue engineering aims at replicating tissues and organs to develop applications in vivo and in vitro. In vivo, by engineering artificial constructs using functional materials and cells to provide both physiological form and function. In vitro, by engineering three-dimensional (3D) models to support drug discovery and enable understanding of fundamental biology. 3D culture constructs mimic cell-cell and cell-matrix interactions and use biomaterials seeking to increase the resemblance of engineered tissues with its in vivo homologues. Native tissues, however, include complex architectures, with compartmentalized regions of different properties containing different types of cells that can be captured by multicompartment constructs. Recent advances in fabrication technologies, such as micropatterning, microfluidics or 3D bioprinting, have enabled compartmentalized structures with defined compositions and properties that are essential in creating 3D cell-laden multiphasic complex architectures. This review focuses on advances in engineered multicompartment constructs that mimic tissue heterogeneity. It includes multiphasic 3D implantable scaffolds and in vitro models, including systems that incorporate different regions emulating in vivo tissues, highlighting the emergence and relevance of 3D bioprinting in the future of biological research and medicine.
Collapse
Affiliation(s)
| | - Vineetha Jayawarna
- Centre for the Cellular MicroenvironmentUniversity of GlasgowGlasgowG11 6EWUK
| | - Emily Maxwell
- Centre for the Cellular MicroenvironmentUniversity of GlasgowGlasgowG11 6EWUK
| | - Matthew J. Dalby
- Centre for the Cellular MicroenvironmentUniversity of GlasgowGlasgowG11 6EWUK
| | | |
Collapse
|
49
|
Elango J, Zamora-Ledezma C, Alexis F, Wu W, Maté-Sánchez de Val JE. Protein Adsorption, Calcium-Binding Ability, and Biocompatibility of Silver Nanoparticle-Loaded Polyvinyl Alcohol (PVA) Hydrogels Using Bone Marrow-Derived Mesenchymal Stem Cells. Pharmaceutics 2023; 15:1843. [PMID: 37514030 PMCID: PMC10384843 DOI: 10.3390/pharmaceutics15071843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 06/19/2023] [Accepted: 06/24/2023] [Indexed: 07/30/2023] Open
Abstract
Several approaches have evolved to facilitate the exploration of hydrogel systems in biomedical research. In this sense, poly(vinyl alcohol) (PVA) has been widely used in hydrogel (HG) fabrication for several therapeutic applications. The biological properties of PVA hydrogels (PVA-HGs) are highly dependent on their interaction with protein receptors and extracellular matrix (mainly calcium) deposition, for which there is not enough evidence from existing research yet. Thus, for the first time, the functional properties, like protein and mineral interactions, related to the proliferation of mesenchymal stem cells (MSCs) by silver nanoparticle (AgNP)-loaded PVA hydrogels (AgNPs-PVA-HGs) were investigated in the present study. The UV absorption spectrum and TEM microscopic results showed a maximum absorbance of synthesized AgNPs at 409 nm, with an average particle size of 14.5 ± 2.5 nm, respectively. The functional properties, such as the calcium-binding and the protein adsorption of PVA-HG, were accelerated by incorporating AgNPs; however, the swelling properties of the HGs were reduced by AgNPs, which might be due to the masking of the free functional groups (hydroxyl groups of PVA) by AgNPs. SEM images showed the presence of AgNPs with a more porous structure in the HGs. The proliferative effect of MSCs increased over culture time from day 1 to day 7, and the cell proliferative effect was upregulated by HGs with more pronounced AgNPs-PVA-HG. In addition, both HGs did not produce any significant cytotoxicity in the MSCs. The histological (bright light and H&E staining) and fluorescence microscopic images showed the presence of a cytoskeleton and the fibrillar structure of the MSCs, and the cells adhered more firmly to all HGs. More fibrillar bipolar and dense fibrillar structures were seen in the day 1 and day 7 cultures, respectively. Interestingly, the MSCs cultured on AgNPs-PVA-HG produced extracellular matrix deposition on day 7. Accordingly, the present results proved the biocompatibility of AgNPs-PVA-HG as a suitable system for culturing mammalian stem cells for regenerative tissue applications.
Collapse
Affiliation(s)
- Jeevithan Elango
- Department of Biomaterials Engineering, Faculty of Health Sciences, UCAM-Universidad Católica San Antonio de Murcia, Campus de los Jerónimos 135, Guadalupe, 30107 Murcia, Spain
- Center of Molecular Medicine and Diagnostics (COMManD), Department of Biochemistry, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai 600077, India
- Department of Marine Bio-Pharmacology, College of Food Science and Technology, Shanghai Ocean University, Shanghai 201306, China
| | - Camilo Zamora-Ledezma
- Green and Innovative Technologies for Food, Environment and Bioengineering Research Group (FEnBeT), Faculty of Pharmacy and Nutrition, UCAM-Universidad Católica San Antonio de Murcia, Campus de los Jerónimos 135, Guadalupe, 30107 Murcia, Spain
| | - Frank Alexis
- Departmento de Ingenería Química, Colegio de Ciencias y Ingenierias, Universidad San Francisco de Quito (Ecuador), Campus Cumbayá, Diego de Robles s/n, Quito 170901, Ecuador
| | - Wenhui Wu
- Department of Marine Bio-Pharmacology, College of Food Science and Technology, Shanghai Ocean University, Shanghai 201306, China
| | - José Eduardo Maté-Sánchez de Val
- Department of Biomaterials Engineering, Faculty of Health Sciences, UCAM-Universidad Católica San Antonio de Murcia, Campus de los Jerónimos 135, Guadalupe, 30107 Murcia, Spain
| |
Collapse
|
50
|
Lee KZ, Jeon J, Jiang B, Subramani SV, Li J, Zhang F. Protein-Based Hydrogels and Their Biomedical Applications. Molecules 2023; 28:4988. [PMID: 37446650 DOI: 10.3390/molecules28134988] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 06/16/2023] [Accepted: 06/21/2023] [Indexed: 07/15/2023] Open
Abstract
Hydrogels made from proteins are attractive materials for diverse medical applications, as they are biocompatible, biodegradable, and amenable to chemical and biological modifications. Recent advances in protein engineering, synthetic biology, and material science have enabled the fine-tuning of protein sequences, hydrogel structures, and hydrogel mechanical properties, allowing for a broad range of biomedical applications using protein hydrogels. This article reviews recent progresses on protein hydrogels with special focus on those made of microbially produced proteins. We discuss different hydrogel formation strategies and their associated hydrogel properties. We also review various biomedical applications, categorized by the origin of protein sequences. Lastly, current challenges and future opportunities in engineering protein-based hydrogels are discussed. We hope this review will inspire new ideas in material innovation, leading to advanced protein hydrogels with desirable properties for a wide range of biomedical applications.
Collapse
Affiliation(s)
- Kok Zhi Lee
- Department of Energy, Environmental & Chemical Engineering, Washington University in St. Louis, One Brookings Drive, Saint Louis, MI 63130, USA
| | - Juya Jeon
- Department of Energy, Environmental & Chemical Engineering, Washington University in St. Louis, One Brookings Drive, Saint Louis, MI 63130, USA
| | - Bojing Jiang
- Department of Energy, Environmental & Chemical Engineering, Washington University in St. Louis, One Brookings Drive, Saint Louis, MI 63130, USA
| | - Shri Venkatesh Subramani
- Department of Energy, Environmental & Chemical Engineering, Washington University in St. Louis, One Brookings Drive, Saint Louis, MI 63130, USA
| | - Jingyao Li
- Department of Energy, Environmental & Chemical Engineering, Washington University in St. Louis, One Brookings Drive, Saint Louis, MI 63130, USA
| | - Fuzhong Zhang
- Department of Energy, Environmental & Chemical Engineering, Washington University in St. Louis, One Brookings Drive, Saint Louis, MI 63130, USA
- Institute of Materials Science and Engineering, Washington University in St. Louis, One Brookings Drive, Saint Louis, MI 63130, USA
- Division of Biological & Biomedical Sciences, Washington University in St. Louis, One Brookings Drive, Saint Louis, MI 63130, USA
| |
Collapse
|