1
|
Guo L, Fu Z, Li H, Wei R, Guo J, Wang H, Qi J. Smart hydrogel: A new platform for cancer therapy. Adv Colloid Interface Sci 2025; 340:103470. [PMID: 40086017 DOI: 10.1016/j.cis.2025.103470] [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: 07/25/2024] [Revised: 01/17/2025] [Accepted: 03/03/2025] [Indexed: 03/16/2025]
Abstract
Cancer is a significant contributor to mortality worldwide, posing a significant threat to human life and health. The unique bioactivity, ability to precisely control drug release, and minimally invasive properties of hydrogels are indispensable attributes that facilitate optimal performance in cancer therapy. However, conventional hydrogels lack the ability to dynamically respond to changes in the surrounding environment, withstand drastic changes in the microenvironment, and trigger drug release on demand. Therefore, this review focuses on smart-responsive hydrogels that are capable of adapting and responding to external stimuli. We comprehensively summarize the raw materials, preparation, and cross-linking mechanisms of smart hydrogels derived from natural and synthetic materials, elucidate the response principles of various smart-responsive hydrogels according to different stimulation sources. Further, we systematically illustrate the important role played by hydrogels in modern cancer therapies within the context of therapeutic principles. Meanwhile, the smart hydrogel that uses machine learning to design precise drug delivery has shown great prospects in cancer therapy. Finally, we present the outlook on future developments and make suggestions for future related work. It is anticipated that this review will promote the practical application of smart hydrogels in cancer therapy and contribute to the advancement of medical treatment.
Collapse
Affiliation(s)
- Li Guo
- Key Laboratory of Dielectric and Electrolyte Functional Material Hebei Province, Northeastern University at Qinhuangdao, Qinhuangdao 066004, China
| | - Ziming Fu
- Key Laboratory of Dielectric and Electrolyte Functional Material Hebei Province, Northeastern University at Qinhuangdao, Qinhuangdao 066004, China
| | - Haoran Li
- Key Laboratory of Dielectric and Electrolyte Functional Material Hebei Province, Northeastern University at Qinhuangdao, Qinhuangdao 066004, China
| | - Ruibo Wei
- Key Laboratory of Dielectric and Electrolyte Functional Material Hebei Province, Northeastern University at Qinhuangdao, Qinhuangdao 066004, China
| | - Jing Guo
- Key Laboratory of Dielectric and Electrolyte Functional Material Hebei Province, Northeastern University at Qinhuangdao, Qinhuangdao 066004, China.
| | - Haiwang Wang
- Key Laboratory of Dielectric and Electrolyte Functional Material Hebei Province, Northeastern University at Qinhuangdao, Qinhuangdao 066004, China.
| | - Jian Qi
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China; School of Chemical Engineering, University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
2
|
Petit N, Chang YYJ, Lobianco FA, Hodgkinson T, Browne S. Hyaluronic acid as a versatile building block for the development of biofunctional hydrogels: In vitro models and preclinical innovations. Mater Today Bio 2025; 31:101596. [PMID: 40083836 PMCID: PMC11903855 DOI: 10.1016/j.mtbio.2025.101596] [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: 12/12/2024] [Revised: 02/07/2025] [Accepted: 02/17/2025] [Indexed: 03/16/2025] Open
Abstract
Hyaluronic acid (HyA) is a non-sulphated linear polysaccharide found abundantly in the extracellular matrix, known for its biocompatibility and versatility in tissue engineering. Chemical modifications of HyA, including methacrylate, acrylate, click chemistry, norbornene, or host-guest chemistry, are necessary for the formation of stable hydrogels with tuneable biophysical characteristics. These modifications enable precise control over stiffness, swelling, degradation, and advanced functionalities such as shear-thinning, self-healing, and injectability. Functionalisation further enhances hydrogel bioactivity, enabling controlled cell adhesion, modulation of cell behaviour, hydrogel degradation, and release profiles, as well as inflammation modulation or bacterial growth inhibition. These are achieved by conjugating proteins, peptides, antibodies, or reactive chemical groups. HyA hydrogels find broad applications both in vitro and in vivo. In vitro, HyA-based hydrogels can support the development of models to understand fundamental processes in health and mechanisms behind disease progression, serving as highly tuneable extracellular matrix mimetics. As therapeutic interventions, injectable or implantable HyA-based hydrogels have been developed to repair a range of tissues, including cartilage, bone, muscle, and skin defects. However, issues remain to be addressed before widespread adoption of HyA-based hydrogels as clinical options. Future innovations for HyA hydrogels include its establishment as an enabling technology for the delivery of novel therapeutics, with a particular focus on immunomodulatory molecules, and the development of more dynamic, tissue-mimetic HyA-based hydrogels.
Collapse
Affiliation(s)
- Noémie Petit
- Tissue Engineering Research Group, Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, 123, St Stephen's Green, Dublin 2, Ireland
- CÚRAM, Research Ireland Centre for Medical Devices, University of Galway, Galway, H91 W2TY, Ireland
| | - Yu-yin Joanne Chang
- Tissue Engineering Research Group, Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, 123, St Stephen's Green, Dublin 2, Ireland
- CÚRAM, Research Ireland Centre for Medical Devices, University of Galway, Galway, H91 W2TY, Ireland
| | - Franz Acker Lobianco
- Tissue Engineering Research Group, Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, 123, St Stephen's Green, Dublin 2, Ireland
| | - Tom Hodgkinson
- Tissue Engineering Research Group, Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, 123, St Stephen's Green, Dublin 2, Ireland
| | - Shane Browne
- Tissue Engineering Research Group, Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, 123, St Stephen's Green, Dublin 2, Ireland
- CÚRAM, Research Ireland Centre for Medical Devices, University of Galway, Galway, H91 W2TY, Ireland
- Trinity Centre for Biomedical Engineering, Trinity College Dublin, Dublin 2, Ireland
| |
Collapse
|
3
|
Wu H, Feng E, Yin H, Zhang Y, Chen G, Zhu B, Yue X, Zhang H, Liu Q, Xiong L. Biomaterials for neuroengineering: applications and challenges. Regen Biomater 2025; 12:rbae137. [PMID: 40007617 PMCID: PMC11855295 DOI: 10.1093/rb/rbae137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2024] [Revised: 10/19/2024] [Accepted: 11/03/2024] [Indexed: 02/27/2025] Open
Abstract
Neurological injuries and diseases are a leading cause of disability worldwide, underscoring the urgent need for effective therapies. Neural regaining and enhancement therapies are seen as the most promising strategies for restoring neural function, offering hope for individuals affected by these conditions. Despite their promise, the path from animal research to clinical application is fraught with challenges. Neuroengineering, particularly through the use of biomaterials, has emerged as a key field that is paving the way for innovative solutions to these challenges. It seeks to understand and treat neurological disorders, unravel the nature of consciousness, and explore the mechanisms of memory and the brain's relationship with behavior, offering solutions for neural tissue engineering, neural interfaces and targeted drug delivery systems. These biomaterials, including both natural and synthetic types, are designed to replicate the cellular environment of the brain, thereby facilitating neural repair. This review aims to provide a comprehensive overview for biomaterials in neuroengineering, highlighting their application in neural functional regaining and enhancement across both basic research and clinical practice. It covers recent developments in biomaterial-based products, including 2D to 3D bioprinted scaffolds for cell and organoid culture, brain-on-a-chip systems, biomimetic electrodes and brain-computer interfaces. It also explores artificial synapses and neural networks, discussing their applications in modeling neural microenvironments for repair and regeneration, neural modulation and manipulation and the integration of traditional Chinese medicine. This review serves as a comprehensive guide to the role of biomaterials in advancing neuroengineering solutions, providing insights into the ongoing efforts to bridge the gap between innovation and clinical application.
Collapse
Affiliation(s)
- Huanghui Wu
- Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Department of Anesthesiology and Perioperative Medicine, Shanghai Fourth People’s Hospital, School of Medicine, Tongji University, Shanghai 200434, China
| | - Enduo Feng
- Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Department of Anesthesiology and Perioperative Medicine, Shanghai Fourth People’s Hospital, School of Medicine, Tongji University, Shanghai 200434, China
| | - Huanxin Yin
- Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Department of Anesthesiology and Perioperative Medicine, Shanghai Fourth People’s Hospital, School of Medicine, Tongji University, Shanghai 200434, China
| | - Yuxin Zhang
- Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Department of Anesthesiology and Perioperative Medicine, Shanghai Fourth People’s Hospital, School of Medicine, Tongji University, Shanghai 200434, China
| | - Guozhong Chen
- Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Department of Anesthesiology and Perioperative Medicine, Shanghai Fourth People’s Hospital, School of Medicine, Tongji University, Shanghai 200434, China
| | - Beier Zhu
- Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Department of Anesthesiology and Perioperative Medicine, Shanghai Fourth People’s Hospital, School of Medicine, Tongji University, Shanghai 200434, China
| | - Xuezheng Yue
- School of Materials and Chemistry, University of Shanghai for Science and Technology, Shanghai 200093, China
| | - Haiguang Zhang
- Rapid Manufacturing Engineering Center, School of Mechatronical Engineering and Automation, Shanghai University, Shanghai 200444, China
- Shanghai Key Laboratory of Intelligent Manufacturing and Robotics, Shanghai University, Shanghai 200072, China
| | - Qiong Liu
- Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Department of Anesthesiology and Perioperative Medicine, Shanghai Fourth People’s Hospital, School of Medicine, Tongji University, Shanghai 200434, China
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Fudan University, Shanghai 200438, China
| | - Lize Xiong
- Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Department of Anesthesiology and Perioperative Medicine, Shanghai Fourth People’s Hospital, School of Medicine, Tongji University, Shanghai 200434, China
| |
Collapse
|
4
|
Simińska-Stanny J, Podstawczyk D, Delporte C, Nie L, Shavandi A. Hyaluronic Acid Role in Biomaterials Prevascularization. Adv Healthc Mater 2024; 13:e2402045. [PMID: 39254277 DOI: 10.1002/adhm.202402045] [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: 06/21/2024] [Indexed: 09/11/2024]
Abstract
Tissue vascularization is a major bottleneck in tissue engineering. In this review, the state of the art on the intricate role of hyaluronic acid (HA) in angiogenesis is explored. HA plays a twofold role in angiogenesis. First, when released as a free polymer in the extracellular matrix (ECM), HA acts as a signaling molecule triggering multiple cascades that foster smooth muscle cell differentiation, migration, and proliferation thereby contributing to vessel wall thickening. Simultaneously, HA bound to the plasma membrane in the pericellular space functions as a polymer block, participating in vessel formation. Starting with the HA origins in native vascular tissues, the approaches aimed at achieving vascularization in vivo are reviewed. The significance of HA molecular weight (MW) in angiogenesis and the challenges associated with utilizing HA in vascular tissue engineering (VTE) are conscientiously addressed. The review finally focuses on a thorough examination and comparison of the diverse strategies adopted to harness the benefits of HA in the vascularization of bioengineered materials. By providing a nuanced perspective on the multifaceted role of HA in angiogenesis, this review contributes to the ongoing discourse in tissue engineering and advances the collective understanding of optimizing vascularization processes assisted by functional biomaterials.
Collapse
Affiliation(s)
- Julia Simińska-Stanny
- 3BIO-BioMatter, Faculty of Engineering, Université libre de Bruxelles (ULB), École polytechnique de Bruxelles, Avenue F.D. Roosevelt, 50 - CP 165/61, Brussels, 1050, Belgium
| | - Daria Podstawczyk
- Department of Process Engineering and Technology of Polymer and Carbon Materials, Faculty of Chemistry, Wroclaw University of Science and Technology, Norwida 4/6, Wroclaw, 50-373, Poland
| | - Christine Delporte
- Laboratoire de Biochimie physiopathologique et nutritionnelle (LBNP), Faculté de Médecine, Université libre de Bruxelles (ULB), Campus Erasme - CP 611, Route de Lennik 808, Bruxelles, 1070, Belgium
| | - Lei Nie
- College of Life Science, Xinyang Normal University, Xinyang, 464031, China
| | - Armin Shavandi
- 3BIO-BioMatter, Faculty of Engineering, Université libre de Bruxelles (ULB), École polytechnique de Bruxelles, Avenue F.D. Roosevelt, 50 - CP 165/61, Brussels, 1050, Belgium
| |
Collapse
|
5
|
Sun Y, Sun X, Wang R, Xing Y, Ma X, Yue J, Zhang M, Wang Y, Tian W, Jing G. Oxidized sodium alginate hydrogel-mouse nerve growth factor sustained release system promotes repair of peripheral nerve injury. JOURNAL OF BIOMATERIALS SCIENCE. POLYMER EDITION 2024; 35:1550-1570. [PMID: 38630632 DOI: 10.1080/09205063.2024.2339636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Accepted: 04/02/2024] [Indexed: 04/19/2024]
Abstract
In recent years, mouse nerve growth factor (mNGF) has emerged as an important biological regulator to repair peripheral nerve injury, but its systemic application is restricted by low efficiency and large dosage requirement. These limitations prompted us to search for biomaterials that can be locally loaded. Oxidized sodium alginate hydrogel (OSA) exhibits good biocompatibility and physicochemical properties, and can be loaded with drugs to construct a sustained-release system that can act locally on nerve injury. Here, we constructed a sustained-release system of OSA-mouse nerve growth factor (mNGF), and investigated the loading and release of the drug through Fourier transform infrared spectroscopy and drug release curves. In vitro and in vivo experiments showed that OSA-mNGF significantly promoted the biological activities of RSC-96 cells and facilitated the recovery from sciatic nerve crush injury in rats. This observation may be attributed to the additive effect of OSA on promoting Schwann cell biological activities or its synergistic effect of cross-activating phosphoinositide 3-kinase (PI3K) through extracellular signal regulated kinase (ERK) signaling. Although the specific mechanism of OSA action needs to be explored in the future, the current results provide a valuable preliminary research basis for the clinical application of the OSA-mNGF sustained-release system for nerve repair.
Collapse
Affiliation(s)
- Yuming Sun
- Medical College, First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xiangyu Sun
- Medical College, First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Ruiqi Wang
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, China
| | - Yuhang Xing
- Medical College, First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xiang Ma
- Medical College, First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jie Yue
- Medical College, First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Min Zhang
- Medical College, First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yuezhu Wang
- Medical College, First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Weiming Tian
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, China
| | - Guangping Jing
- Medical College, First Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
6
|
Wasnik K, Gupta PS, Singh G, Maity S, Patra S, Pareek D, Kumar S, Rai V, Prakash R, Acharya A, Maiti P, Mukherjee S, Mastai Y, Paik P. Neurogenic and angiogenic poly( N-acryloylglycine)- co-(acrylamide)- co-( N-acryloyl-glutamate) hydrogel: preconditioning effect under oxidative stress and use in neuroregeneration. J Mater Chem B 2024; 12:6221-6241. [PMID: 38835196 DOI: 10.1039/d4tb00243a] [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: 06/06/2024]
Abstract
Traumatic injuries, neurodegenerative diseases and oxidative stress serve as the early biomarkers for neuronal damage and impede angiogenesis and subsequently neuronal growth. Considering this, the present work aimed to develop a poly(N-acryloylglycine)-co-(acrylamide)-co-(N-acryloylglutamate) hydrogel [p(NAG-Ac-NAE)] with angiogenesis/neurogenesis properties. As constituents of this polymer modulate their vital role in biological functions, inhibitory neurotransmitter glycine regulates neuronal homeostasis, and glutamatergic signalling regulates angiogenesis. The p(NAG-Ac-NAE) hydrogel is a highly branched, biodegradable and pH-responsive polymer with a very high swelling behavior of 6188%. The mechanical stability (G', 2.3-2.7 kPa) of this polymeric hydrogel is commendable in the differentiation of mature neurons. This hydrogel is biocompatible (as tested in HUVEC cells) and helps to proliferate PC12 cells (152.7 ± 13.7%), whereas it is cytotoxic towards aggressive cancers such as glioblastoma (LN229 cells) and triple negative breast cancer (TNBC; MDA-MB-231 cells) and helps to maintain the healthy cytoskeleton framework structure of primary cortical neurons by facilitating the elongation of the axonal pathway. Furthermore, FACS results revealed that the synthesized hydrogel potentiates neurogenesis by inducing the cell cycle (G0/G1) and arresting the sub-G1 phase by limiting apoptosis. Additionally, RT-PCR results revealed that this hydrogel induced an increased level of HIF-1α expression, providing preconditioning effects towards neuronal cells under oxidative stress by scavenging ROS and initiating neurogenic and angiogenic signalling. This hydrogel further exhibits more pro-angiogenic activities by increasing the expression of VEGF isoforms compared to previously reported hydrogels. In conclusion, the newly synthesized p(NAG-Ac-NAE) hydrogel can be one of the potential neuroregenerative materials for vasculogenesis-assisted neurogenic applications and paramount for the management of neurodegenerative diseases.
Collapse
Affiliation(s)
- Kirti Wasnik
- School of Biomedical Engineering, Indian Institute of Technology, Banaras Hindu University (BHU), Varanasi, Uttar Pradesh 221 005, India.
| | - Prem Shankar Gupta
- School of Biomedical Engineering, Indian Institute of Technology, Banaras Hindu University (BHU), Varanasi, Uttar Pradesh 221 005, India.
| | - Gurmeet Singh
- School of Biomedical Engineering, Indian Institute of Technology, Banaras Hindu University (BHU), Varanasi, Uttar Pradesh 221 005, India.
| | - Somedutta Maity
- School of Engineering Sciences and Technology, University of Hyderabad, Telangana State 500 046, India
| | - Sukanya Patra
- School of Biomedical Engineering, Indian Institute of Technology, Banaras Hindu University (BHU), Varanasi, Uttar Pradesh 221 005, India.
| | - Divya Pareek
- School of Biomedical Engineering, Indian Institute of Technology, Banaras Hindu University (BHU), Varanasi, Uttar Pradesh 221 005, India.
| | - Sandeep Kumar
- Department of Zoology, Banaras Hindu University (BHU), Varanasi, Uttar Pradesh 221005, India
| | - Vipin Rai
- Department of Biochemistry, Institute of Sciences, Banaras Hindu University (BHU), Varanasi, Uttar Pradesh 221005, India
| | - Ravi Prakash
- School of Material Science, Indian Institute of Technology, Banaras Hindu University (BHU), Varanasi, Uttar Pradesh 221 005, India
| | - Arbind Acharya
- Department of Zoology, Banaras Hindu University (BHU), Varanasi, Uttar Pradesh 221005, India
| | - Pralay Maiti
- School of Material Science, Indian Institute of Technology, Banaras Hindu University (BHU), Varanasi, Uttar Pradesh 221 005, India
| | - Sudip Mukherjee
- School of Biomedical Engineering, Indian Institute of Technology, Banaras Hindu University (BHU), Varanasi, Uttar Pradesh 221 005, India.
| | - Yitzhak Mastai
- Department of Chemistry and the Institute of Nanotechnology, Bar-Ilan University, Ramat-Gan, 52900, Israel
| | - Pradip Paik
- School of Biomedical Engineering, Indian Institute of Technology, Banaras Hindu University (BHU), Varanasi, Uttar Pradesh 221 005, India.
| |
Collapse
|
7
|
Zhou G, Cao Y, Yan Y, Xu H, Zhang X, Yan T, Wan H. Injectable Hydrogels Based on Hyaluronic Acid and Gelatin Combined with Salvianolic Acid B and Vascular Endothelial Growth Factor for Treatment of Traumatic Brain Injury in Mice. Molecules 2024; 29:1705. [PMID: 38675525 PMCID: PMC11052029 DOI: 10.3390/molecules29081705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Revised: 04/01/2024] [Accepted: 04/08/2024] [Indexed: 04/28/2024] Open
Abstract
Traumatic brain injury (TBI) leads to structural damage in the brain, and is one of the major causes of disability and death in the world. Herein, we developed a composite injectable hydrogel (HA/Gel) composed of hyaluronic acid (HA) and gelatin (Gel), loaded with vascular endothelial growth factor (VEGF) and salvianolic acid B (SAB) for treatment of TBI. The HA/Gel hydrogels were formed by the coupling of phenol-rich tyramine-modified HA (HA-TA) and tyramine-modified Gel (Gel-TA) catalyzed by horseradish peroxidase (HRP) in the presence of hydrogen peroxide (H2O2). SEM results showed that HA/Gel hydrogel had a porous structure. Rheological test results showed that the hydrogel possessed appropriate rheological properties, and UV spectrophotometry results showed that the hydrogel exhibited excellent SAB release performance. The results of LIVE/DEAD staining, CCK-8 and Phalloidin/DAPI fluorescence staining showed that the HA/Gel hydrogel possessed good cell biocompatibility. Moreover, the hydrogels loaded with SAB and VEGF (HA/Gel/SAB/VEGF) could effectively promote the proliferation of bone marrow mesenchymal stem cells (BMSCs). In addition, the results of H&E staining, CD31 and α-SMA immunofluorescence staining showed that the HA/Gel/SAB/VEGF hydrogel possessed good in vivo biocompatibility and pro-angiogenic ability. Furthermore, immunohistochemical results showed that the injection of HA/Gel/SAB/VEGF hydrogel to the injury site could effectively reduce the volume of defective tissues in traumatic brain injured mice. Our results suggest that the injection of HA/Gel hydrogel loaded with SAB and VEGF might provide a new approach for therapeutic brain tissue repair after traumatic brain injury.
Collapse
Affiliation(s)
- Guoying Zhou
- College of Life Science, Zhejiang Chinese Medical University, Hangzhou 310053, China; (G.Z.); (Y.C.); (Y.Y.); (H.X.); (X.Z.)
| | - Yajie Cao
- College of Life Science, Zhejiang Chinese Medical University, Hangzhou 310053, China; (G.Z.); (Y.C.); (Y.Y.); (H.X.); (X.Z.)
| | - Yujia Yan
- College of Life Science, Zhejiang Chinese Medical University, Hangzhou 310053, China; (G.Z.); (Y.C.); (Y.Y.); (H.X.); (X.Z.)
| | - Haibo Xu
- College of Life Science, Zhejiang Chinese Medical University, Hangzhou 310053, China; (G.Z.); (Y.C.); (Y.Y.); (H.X.); (X.Z.)
| | - Xiao Zhang
- College of Life Science, Zhejiang Chinese Medical University, Hangzhou 310053, China; (G.Z.); (Y.C.); (Y.Y.); (H.X.); (X.Z.)
| | - Tingzi Yan
- College of Material, Chemistry and Chemical Engineering, Hangzhou Normal University, Hangzhou 311121, China
| | - Haitong Wan
- Institute of Cardio-Cerebrovascular Disease, Zhejiang Chinese Medical University, Hangzhou 310053, China
| |
Collapse
|
8
|
Dong Z, Zhao J, Xu J, Deng W, Sun P. Strongly Adhesive, Self-Healing, Hemostatic Hydrogel for the Repair of Traumatic Brain Injury. Biomacromolecules 2024; 25:2462-2475. [PMID: 38533630 DOI: 10.1021/acs.biomac.3c01406] [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: 03/28/2024]
Abstract
With wide clinical demands, therapies for traumatic brain injury (TBI) are a major problem in surgical procedures and after major trauma. Due to the difficulty in regeneration of neurons or axons after injury, as well as the inhibition of blood vessel growth by the formation of neural scars, existing treatment measures have limited effectiveness in repairing brain tissue. Herein, the biomultifunctional hydrogels are developed for TBI treatment based on the Schiff base reaction of calcium ion (Ca2+)-cross-linked oxidized sodium alginate (OSA) and carboxymethyl chitosan (CMCS). The obtained COCS hydrogel exhibits excellent adhesion to wet tissues, self-repair capability, and antimicrobial properties. What's particularly interesting is that the addition of Ca2+ increases the hydrogel's extensibility, enhancing its hemostatic capabilities. Biological assessments indicate that the COCS hydrogel demonstrates excellent biocompatibility, hemostatic properties, and the ability to promote arterial vessel repair. Importantly, the COCS hydrogel promotes the growth of cerebral microvessels by upregulating CD31, accelerates the proliferation of astrocytes, enhances the expression of GFAP, and stimulates the expression of neuron-specific markers such as NEUN and β-tubulin. All of these findings highlight that the strongly adhesive, self-healing, hemostatic hydrogel shows great potential for the repair of traumatic brain injury and other tissue repair therapy.
Collapse
Affiliation(s)
- Zuoxiang Dong
- Department of Neurosurgery, The Affiliated Hospital of Qingdao University, Wutaishan Road 1677, Qingdao, Shandong 266000, China
| | - Jihu Zhao
- Department of Neurosurgery, The Affiliated Hospital of Qingdao University, Wutaishan Road 1677, Qingdao, Shandong 266000, China
| | - Jian Xu
- Department of Neurosurgery, The Affiliated Hospital of Qingdao University, Wutaishan Road 1677, Qingdao, Shandong 266000, China
| | - Wenshuai Deng
- Department of Neurosurgery, The Affiliated Hospital of Qingdao University, Wutaishan Road 1677, Qingdao, Shandong 266000, China
| | - Peng Sun
- Department of Neurosurgery, The Affiliated Hospital of Qingdao University, Wutaishan Road 1677, Qingdao, Shandong 266000, China
| |
Collapse
|
9
|
Mahmoudi N, Mohamed E, Dehnavi SS, Aguilar LMC, Harvey AR, Parish CL, Williams RJ, Nisbet DR. Calming the Nerves via the Immune Instructive Physiochemical Properties of Self-Assembling Peptide Hydrogels. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2303707. [PMID: 38030559 PMCID: PMC10837390 DOI: 10.1002/advs.202303707] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 10/22/2023] [Indexed: 12/01/2023]
Abstract
Current therapies for the devastating damage caused by traumatic brain injuries (TBI) are limited. This is in part due to poor drug efficacy to modulate neuroinflammation, angiogenesis and/or promoting neuroprotection and is the combined result of challenges in getting drugs across the blood brain barrier, in a targeted approach. The negative impact of the injured extracellular matrix (ECM) has been identified as a factor in restricting post-injury plasticity of residual neurons and is shown to reduce the functional integration of grafted cells. Therefore, new strategies are needed to manipulate the extracellular environment at the subacute phase to enhance brain regeneration. In this review, potential strategies are to be discussed for the treatment of TBI by using self-assembling peptide (SAP) hydrogels, fabricated via the rational design of supramolecular peptide scaffolds, as an artificial ECM which under the appropriate conditions yields a supramolecular hydrogel. Sequence selection of the peptides allows the tuning of these hydrogels' physical and biochemical properties such as charge, hydrophobicity, cell adhesiveness, stiffness, factor presentation, degradation profile and responsiveness to (external) stimuli. This review aims to facilitate the development of more intelligent biomaterials in the future to satisfy the parameters, requirements, and opportunities for the effective treatment of TBI.
Collapse
Affiliation(s)
- Negar Mahmoudi
- Laboratory of Advanced Biomaterialsthe John Curtin School of Medical ResearchAustralian National UniversityCanberraACT2601Australia
- ANU College of Engineering & Computer ScienceAustralian National UniversityCanberraACT2601Australia
- The Graeme Clark InstituteThe University of MelbourneMelbourneVIC3010Australia
- Department of Biomedical EngineeringFaculty of Engineering and Information TechnologyThe University of MelbourneMelbourneVIC3010Australia
| | - Elmira Mohamed
- Laboratory of Advanced Biomaterialsthe John Curtin School of Medical ResearchAustralian National UniversityCanberraACT2601Australia
| | - Shiva Soltani Dehnavi
- Laboratory of Advanced Biomaterialsthe John Curtin School of Medical ResearchAustralian National UniversityCanberraACT2601Australia
- ANU College of Engineering & Computer ScienceAustralian National UniversityCanberraACT2601Australia
| | - Lilith M. Caballero Aguilar
- Laboratory of Advanced Biomaterialsthe John Curtin School of Medical ResearchAustralian National UniversityCanberraACT2601Australia
- The Graeme Clark InstituteThe University of MelbourneMelbourneVIC3010Australia
- Department of Biomedical EngineeringFaculty of Engineering and Information TechnologyThe University of MelbourneMelbourneVIC3010Australia
| | - Alan R. Harvey
- School of Human SciencesThe University of Western Australiaand Perron Institute for Neurological and Translational SciencePerthWA6009Australia
| | - Clare L. Parish
- The Florey Institute of Neuroscience and Mental HealthThe University of MelbourneParkvilleMelbourneVIC3010Australia
| | | | - David R. Nisbet
- Laboratory of Advanced Biomaterialsthe John Curtin School of Medical ResearchAustralian National UniversityCanberraACT2601Australia
- The Graeme Clark InstituteThe University of MelbourneMelbourneVIC3010Australia
- Department of Biomedical EngineeringFaculty of Engineering and Information TechnologyThe University of MelbourneMelbourneVIC3010Australia
- Melbourne Medical SchoolFaculty of MedicineDentistry and Health ScienceThe University of MelbourneMelbourneVIC3010Australia
| |
Collapse
|
10
|
Pereira M, Pinto J, Arteaga B, Guerra A, Jorge RN, Monteiro FJ, Salgado CL. A Comprehensive Look at In Vitro Angiogenesis Image Analysis Software. Int J Mol Sci 2023; 24:17625. [PMID: 38139453 PMCID: PMC10743557 DOI: 10.3390/ijms242417625] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 12/11/2023] [Accepted: 12/14/2023] [Indexed: 12/24/2023] Open
Abstract
One of the complex challenges faced presently by tissue engineering (TE) is the development of vascularized constructs that accurately mimic the extracellular matrix (ECM) of native tissue in which they are inserted to promote vessel growth and, consequently, wound healing and tissue regeneration. TE technique is characterized by several stages, starting from the choice of cell culture and the more appropriate scaffold material that can adequately support and supply them with the necessary biological cues for microvessel development. The next step is to analyze the attained microvasculature, which is reliant on the available labeling and microscopy techniques to visualize the network, as well as metrics employed to characterize it. These are usually attained with the use of software, which has been cited in several works, although no clear standard procedure has been observed to promote the reproduction of the cell response analysis. The present review analyzes not only the various steps previously described in terms of the current standards for evaluation, but also surveys some of the available metrics and software used to quantify networks, along with the detection of analysis limitations and future improvements that could lead to considerable progress for angiogenesis evaluation and application in TE research.
Collapse
Affiliation(s)
- Mariana Pereira
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (M.P.); (J.P.); (B.A.); (F.J.M.)
- INEB—Instituto de Engenharia Biomédica, Universidade do Porto, 4200-135 Porto, Portugal
| | - Jéssica Pinto
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (M.P.); (J.P.); (B.A.); (F.J.M.)
- INEB—Instituto de Engenharia Biomédica, Universidade do Porto, 4200-135 Porto, Portugal
| | - Belén Arteaga
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (M.P.); (J.P.); (B.A.); (F.J.M.)
- INEB—Instituto de Engenharia Biomédica, Universidade do Porto, 4200-135 Porto, Portugal
- Faculty of Medicine, University of Granada, Parque Tecnológico de la Salud, Av. de la Investigación 11, 18016 Granada, Spain
| | - Ana Guerra
- INEGI—Instituto de Ciência e Inovação em Engenharia Mecânica e Engenharia Industrial, 4200-465 Porto, Portugal; (A.G.); (R.N.J.)
| | - Renato Natal Jorge
- INEGI—Instituto de Ciência e Inovação em Engenharia Mecânica e Engenharia Industrial, 4200-465 Porto, Portugal; (A.G.); (R.N.J.)
- LAETA—Laboratório Associado de Energia, Transportes e Aeronáutica, Universidade do Porto, 4200-165 Porto, Portugal
- FEUP—Faculdade de Engenharia, Departamento de Engenharia Metalúrgica e de Materiais, Universidade do Porto, 4200-165 Porto, Portugal
| | - Fernando Jorge Monteiro
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (M.P.); (J.P.); (B.A.); (F.J.M.)
- INEB—Instituto de Engenharia Biomédica, Universidade do Porto, 4200-135 Porto, Portugal
- FEUP—Faculdade de Engenharia, Departamento de Engenharia Metalúrgica e de Materiais, Universidade do Porto, 4200-165 Porto, Portugal
- PCCC—Porto Comprehensive Cancer Center, 4200-072 Porto, Portugal
| | - Christiane Laranjo Salgado
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (M.P.); (J.P.); (B.A.); (F.J.M.)
- INEB—Instituto de Engenharia Biomédica, Universidade do Porto, 4200-135 Porto, Portugal
| |
Collapse
|
11
|
Evans D, Barcons AM, Basit RH, Adams C, Chari DM. Evaluating the Feasibility of Hydrogel-Based Neural Cell Sprays. J Funct Biomater 2023; 14:527. [PMID: 37888192 PMCID: PMC10607175 DOI: 10.3390/jfb14100527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2023] [Revised: 10/05/2023] [Accepted: 10/11/2023] [Indexed: 10/28/2023] Open
Abstract
Neurological injuries have poor prognoses with serious clinical sequelae. Stem cell transplantation enhances neural repair but is hampered by low graft survival (ca. 80%) and marker expression/proliferative potential of hydrogel-sprayed astrocytes was retained. Combining a cell spray format with polymer encapsulation technologies could form the basis of a non-invasive graft delivery method, offering potential advantages over current cell delivery approaches.
Collapse
Affiliation(s)
- Daisy Evans
- Keele University School of Medicine, Keele University, Staffordshire ST5 5BG, UK;
| | - Aina Mogas Barcons
- Department of Physiology, Anatomy and Genetics, Oxford Parkinson’s Disease Centre, University of Oxford, Oxford OX1 3AZ, UK;
| | - Raja Haseeb Basit
- Department of General Surgery, Queen Elizabeth Hospital, Birmingham B15 2GW, UK;
| | - Christopher Adams
- Neural Tissue Engineering, School of Life Sciences, Keele University, Staffordshire ST5 5BG, UK;
| | - Divya Maitreyi Chari
- Neural Tissue Engineering, School of Life Sciences, Keele University, Staffordshire ST5 5BG, UK;
| |
Collapse
|
12
|
Karam J, Singer BJ, Miwa H, Chen LH, Maran K, Hasani M, Garza S, Onyekwere B, Yeh HC, Li S, Carlo DD, Seidlits SK. Molecular weight of hyaluronic acid crosslinked into biomaterial scaffolds affects angiogenic potential. Acta Biomater 2023; 169:228-242. [PMID: 37572983 PMCID: PMC11729822 DOI: 10.1016/j.actbio.2023.08.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 07/29/2023] [Accepted: 08/01/2023] [Indexed: 08/14/2023]
Abstract
While hyaluronic acid (HA)-based hydrogels have been used clinically for decades, the mechanisms by which HA exerts molecular weight-dependent bioactivity and how chemical modification and crosslinking may affect molecular weight-dependent bioactivity remain poorly understood. This knowledge gap presents a significant barrier to designing HA hydrogels with predictable bioactivities. As HA has been widely reported to have molecular weight-dependent effects on endothelial cells (ECs), we investigated how the molecular weight of HA in either soluble or crosslinked forms affects angiogenesis and interrogated CD44 clustering on the surface of endothelial cells as a candidate mechanism for these affects. Using soluble HA, our results show high molecular weight (HMW) HA, but not low molecular weight (LMW) HA, increased viability and tube formation in cultured human cerebral microvascular ECs (HCMVECs). No size of HA affected proliferation. When HCMVECs were cultured with crosslinked HA of varying molecular weights in the form of HA-based microporous annealed particle scaffold (HMAPS), the cell response was comparable to when cultured with soluble HA. Similarly, when implanted subcutaneously, HMAPS with HMW HA were more vascularized than those with LMW HA. We also show that antibody-mediated CD44 clustering resulted in HCMVECs with increased viability and tube-like structure formation in a manner comparable to exposure to HMW HA, suggesting that HMW acts through CD44 clustering. STATEMENT OF SIGNIFICANCE: Biomaterials based on hyaluronic acid (HA), a bioactive extracellular matrix polysaccharide, have been used in clinical products for several years. Despite the knowledge that HA molecular weight heavily influences its bioactivity, molecular weight has been largely ignored in the development of HA-based biomaterials. Given the high viscosity of high molecular weight HA typically found in native tissues, lower molecular weight polysaccharides have been used most commonly for biomaterial fabrication. By comparing the ability of injectable, microporous annealed particle scaffolds (MAPS) fabricated from variably sized HA to promote angiogenesis, this study demonstrates that MAPS with high molecular weight HA better support vascularization, likely through an unique ability to induce clustering of CD44 receptors on endothelial cells.
Collapse
Affiliation(s)
- Josh Karam
- Department of Bioengineering, University of California, Los Angeles, CA 90095, USA
| | - Breahna J Singer
- Department of Bioengineering, University of California, Los Angeles, CA 90095, USA; Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX 78712, USA
| | - Hiromi Miwa
- Department of Bioengineering, University of California, Los Angeles, CA 90095, USA
| | - Limin H Chen
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX 78712, USA
| | - Kajal Maran
- Department of Bioengineering, University of California, Los Angeles, CA 90095, USA
| | - Mahdi Hasani
- Department of Bioengineering, University of California, Los Angeles, CA 90095, USA
| | - Sarahi Garza
- Department of Bioengineering, University of California, Los Angeles, CA 90095, USA
| | - Bianca Onyekwere
- Department of Bioengineering, University of California, Los Angeles, CA 90095, USA
| | - Hsin-Chih Yeh
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX 78712, USA; Texas Materials Institute, The University of Texas at Austin, Austin, TX 78712, USA
| | - Song Li
- Department of Bioengineering, University of California, Los Angeles, CA 90095, USA
| | - Dino Di Carlo
- Department of Bioengineering, University of California, Los Angeles, CA 90095, USA
| | - Stephanie K Seidlits
- Department of Bioengineering, University of California, Los Angeles, CA 90095, USA; Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX 78712, USA.
| |
Collapse
|
13
|
Zhao ZA, Yan L, Wen J, Satyanarayanan SK, Yu F, Lu J, Liu YU, Su H. Cellular and molecular mechanisms in vascular repair after traumatic brain injury: a narrative review. BURNS & TRAUMA 2023; 11:tkad033. [PMID: 37675267 PMCID: PMC10478165 DOI: 10.1093/burnst/tkad033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Revised: 02/01/2023] [Accepted: 05/26/2023] [Indexed: 09/08/2023]
Abstract
Traumatic brain injury (TBI) disrupts normal brain function and is associated with high morbidity and fatality rates. TBI is characterized as mild, moderate or severe depending on its severity. The damage may be transient and limited to the dura matter, with only subtle changes in cerebral parenchyma, or life-threatening with obvious focal contusions, hematomas and edema. Blood vessels are often injured in TBI. Even in mild TBI, dysfunctional cerebral vascular repair may result in prolonged symptoms and poor outcomes. Various distinct types of cells participate in vascular repair after TBI. A better understanding of the cellular response and function in vascular repair can facilitate the development of new therapeutic strategies. In this review, we analyzed the mechanism of cerebrovascular impairment and the repercussions following various forms of TBI. We then discussed the role of distinct cell types in the repair of meningeal and parenchyma vasculature following TBI, including endothelial cells, endothelial progenitor cells, pericytes, glial cells (astrocytes and microglia), neurons, myeloid cells (macrophages and monocytes) and meningeal lymphatic endothelial cells. Finally, possible treatment techniques targeting these unique cell types for vascular repair after TBI are discussed.
Collapse
Affiliation(s)
- Zi-Ai Zhao
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Avenida da Universidade, Taipa, Macau 999078, China
- Department of Neurology, General Hospital of Northern Theater Command, 83# Wen-Hua Road, Shenyang 110840, China
| | - Lingli Yan
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Avenida da Universidade, Taipa, Macau 999078, China
| | - Jing Wen
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Avenida da Universidade, Taipa, Macau 999078, China
| | - Senthil Kumaran Satyanarayanan
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Avenida da Universidade, Taipa, Macau 999078, China
| | - Feng Yu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Avenida da Universidade, Taipa, Macau 999078, China
| | - Jiahong Lu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Avenida da Universidade, Taipa, Macau 999078, China
| | - Yong U Liu
- Laboratory of Neuroimmunology in Health and Disease Institute, Guangzhou First People’s Hospital School of Medicine, South China University of Technology, Guangzhou Higher Education Mega Center, Panyu District, Guangzhou 511400, China
| | - Huanxing Su
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Avenida da Universidade, Taipa, Macau 999078, China
| |
Collapse
|
14
|
Nicosia A, Salamone M, Costa S, Ragusa MA, Ghersi G. Mimicking Molecular Pathways in the Design of Smart Hydrogels for the Design of Vascularized Engineered Tissues. Int J Mol Sci 2023; 24:12314. [PMID: 37569691 PMCID: PMC10418696 DOI: 10.3390/ijms241512314] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 07/21/2023] [Accepted: 07/31/2023] [Indexed: 08/13/2023] Open
Abstract
Biomaterials are pivotal in supporting and guiding vascularization for therapeutic applications. To design effective, bioactive biomaterials, understanding the cellular and molecular processes involved in angiogenesis and vasculogenesis is crucial. Biomaterial platforms can replicate the interactions between cells, the ECM, and the signaling molecules that trigger blood vessel formation. Hydrogels, with their soft and hydrated properties resembling natural tissues, are widely utilized; particularly synthetic hydrogels, known for their bio-inertness and precise control over cell-material interactions, are utilized. Naturally derived and synthetic hydrogel bases are tailored with specific mechanical properties, controlled for biodegradation, and enhanced for cell adhesion, appropriate biochemical signaling, and architectural features that facilitate the assembly and tubulogenesis of vascular cells. This comprehensive review showcases the latest advancements in hydrogel materials and innovative design modifications aimed at effectively guiding and supporting vascularization processes. Furthermore, by leveraging this knowledge, researchers can advance biomaterial design, which will enable precise support and guidance of vascularization processes and ultimately enhance tissue functionality and therapeutic outcomes.
Collapse
Affiliation(s)
- Aldo Nicosia
- Institute for Biomedical Research and Innovation-National Research Council (IRIB-CNR), Via Ugo la Malfa 153, 90146 Palermo, Italy;
| | - Monica Salamone
- Institute for Biomedical Research and Innovation-National Research Council (IRIB-CNR), Via Ugo la Malfa 153, 90146 Palermo, Italy;
| | - Salvatore Costa
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Viale delle Scienze, Ed. 16, 90128 Palermo, Italy; (S.C.); (M.A.R.); (G.G.)
| | - Maria Antonietta Ragusa
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Viale delle Scienze, Ed. 16, 90128 Palermo, Italy; (S.C.); (M.A.R.); (G.G.)
| | - Giulio Ghersi
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Viale delle Scienze, Ed. 16, 90128 Palermo, Italy; (S.C.); (M.A.R.); (G.G.)
| |
Collapse
|
15
|
Hwang HS, Lee CS. Recent Progress in Hyaluronic-Acid-Based Hydrogels for Bone Tissue Engineering. Gels 2023; 9:588. [PMID: 37504467 PMCID: PMC10379028 DOI: 10.3390/gels9070588] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 07/17/2023] [Accepted: 07/19/2023] [Indexed: 07/29/2023] Open
Abstract
Hydrogel-based bone tissue engineering is a potential strategy for treating bone abnormalities and fractures. Hyaluronic acid (HA) is a natural polymer that is widely distributed in the human body and plays a significant role in numerous physiological processes such as cell migration, tissue hydration, and wound healing. Hydrogels based on HA and its derivatives have gained popularity as potential treatments for bone-related diseases. HA-based hydrogels have been extensively studied for their ability to mimic the natural extracellular matrix of bone tissue and provide a suitable microenvironment for cell support and tissue regeneration. The physical and chemical properties of HA can be modified to improve its mechanical strength, biocompatibility, and osteogenic potential. Moreover, HA-based hydrogels combined with other biomaterials in the presence or absence of bioactive agents have been investigated as a means of improving the mechanical properties and bioactivity of the hydrogel scaffold. Therefore, HA-based hydrogels have shown great promise in bone tissue engineering due to their biocompatibility, osteogenic activity, and ability to mimic the natural extracellular matrix of bone tissue. Overall, this review provides a comprehensive overview of the current state of the art in HA-based hydrogels for bone tissue engineering, highlighting the key advances, challenges, and future directions in this rapidly evolving field.
Collapse
Affiliation(s)
- Hee Sook Hwang
- Department of Pharmaceutical Engineering, Dankook University, Cheonan 31116, Republic of Korea
| | - Chung-Sung Lee
- Department of Pharmaceutical Engineering, Soonchunhyang University, Asan 31538, Republic of Korea
| |
Collapse
|
16
|
Villanueva-Flores F, Garcia-Atutxa I, Santos A, Armendariz-Borunda J. Toward a New Generation of Bio-Scaffolds for Neural Tissue Engineering: Challenges and Perspectives. Pharmaceutics 2023; 15:1750. [PMID: 37376198 DOI: 10.3390/pharmaceutics15061750] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 06/04/2023] [Accepted: 06/13/2023] [Indexed: 06/29/2023] Open
Abstract
Neural tissue engineering presents a compelling technological breakthrough in restoring brain function, holding immense promise. However, the quest to develop implantable scaffolds for neural culture that fulfill all necessary criteria poses a remarkable challenge for material science. These materials must possess a host of desirable characteristics, including support for cellular survival, proliferation, and neuronal migration and the minimization of inflammatory responses. Moreover, they should facilitate electrochemical cell communication, display mechanical properties akin to the brain, emulate the intricate architecture of the extracellular matrix, and ideally allow the controlled release of substances. This comprehensive review delves into the primary requisites, limitations, and prospective avenues for scaffold design in brain tissue engineering. By offering a panoramic overview, our work aims to serve as an essential resource, guiding the creation of materials endowed with bio-mimetic properties, ultimately revolutionizing the treatment of neurological disorders by developing brain-implantable scaffolds.
Collapse
Affiliation(s)
- Francisca Villanueva-Flores
- Escuela de Medicina y Ciencias de la Salud, Tecnologico de Monterrey, Campus Chihuahua, Av. Heroico Colegio Militar 4700, Nombre de Dios, Chihuahua 31300, Chihuahua, Mexico
| | - Igor Garcia-Atutxa
- Máster en Bioinformática y Bioestadística, Universitat Oberta de Catalunya, Rambla del Poblenou, 156, 08018 Barcelona, Spain
| | - Arturo Santos
- Escuela de Medicina y Ciencias de la Salud, Tecnologico de Monterrey, Campus Guadalajara, Av. Gral Ramón Corona No 2514, Colonia Nuevo México, Zapopan 45201, Jalisco, Mexico
| | - Juan Armendariz-Borunda
- Escuela de Medicina y Ciencias de la Salud, Tecnologico de Monterrey, Campus Guadalajara, Av. Gral Ramón Corona No 2514, Colonia Nuevo México, Zapopan 45201, Jalisco, Mexico
- Instituto de Biología Molecular en Medicina y Terapia Génica, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Sierra Mojada 950, Independencia Oriente, Guadalajara 44340, Jalisco, Mexico
| |
Collapse
|
17
|
Injectable hydrogel encapsulated with VEGF-mimetic peptide-loaded nanoliposomes promotes peripheral nerve repair in vivo. Acta Biomater 2023; 160:225-238. [PMID: 36774975 DOI: 10.1016/j.actbio.2023.02.004] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2022] [Revised: 01/18/2023] [Accepted: 02/02/2023] [Indexed: 02/12/2023]
Abstract
Repair of peripheral nerve crush injury remains a major clinical challenge. Currently, oral or intravenous neurotrophic drugs are the main treatment for peripheral nerve crush injury; however, this repair process is slow, and the final effect may be uncertain. The current study aimed at developing an injectable hydrogel with vascular endothelial growth factor (VEGF)-mimetic peptide (QK)-encapsulated nanoliposomes (QK-NLs@Gel) for sustainable drug release that creates an appropriate microenvironment for nerve regeneration. The QK-encapsulated nanoliposomes (QK-NLs) could facilitate the proliferation, migration, and tube formation capacities of human umbilical vein endothelial cells through the VEGF signaling pathway. The QK-NLs@Gel hydrogel encapsulated with QK-NLs showed enhanced physical properties and appropriate biocompatibility in vitro. Thereafter, the QK-NLs@Gel hydrogel was directly injected into the site of peripheral nerve crush injury in a rat model, where it enhanced revascularization and promoted the M2-polarization of the macrophages, thus providing an optimized microenvironment for nerve regeneration. At four weeks post-surgery, the QK-NLs@Gel injected rats exhibited enhanced axon regeneration, remyelination, and better functional recovery in comparison with other groups in vivo. Overall, these findings demonstrate that the composite hydrogel could promote a multicellular pro-regenerative microenvironment at the peripheral nerve injury site, thus revealing great potential for peripheral nerve restoration. STATEMENT OF SIGNIFICANCE: Peripheral nerve injury (PNI) is a leading public health issue, and how to delivery beneficial drugs to injured sites efficiently is still a big challenge. In the current study, an injectable hydrogel with VEGF-mimetic peptide (QK)-encapsulated nanoliposomes (QK-NLs@Gel) was first developed and used to repair a rat crush injury model. Our results showed that QK-NLs promoted the proliferation, migration, and angiogenesis of HUVEC via VEGF signaling pathway in vitro. Furthermore, when injected to the crushed sites in vivo, the QK-NLs@Gel hydrogel could accelerate nerve repair through enhanced revascularization and M2-polarization of macrophages. These results collectively demonstrate that injection of QK-NLs@Gel hydrogel could create an appropriate microenvironment for peripheral nerve regeneration. This strategy is effective, economical, and convenient for clinical applications.
Collapse
|
18
|
Lee H, Kim SH, Lee JS, Lee YJ, Lee OJ, Ajiteru O, Sultan MT, Lee SW, Park CH. Functional Skeletal Muscle Regeneration Using Muscle Mimetic Tissue Fabricated by Microvalve-Assisted Coaxial 3D Bioprinting. Adv Healthc Mater 2023; 12:e2202664. [PMID: 36469728 DOI: 10.1002/adhm.202202664] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 11/23/2022] [Indexed: 12/12/2022]
Abstract
3D-printed artificial skeletal muscle, which mimics the structural and functional characteristics of native skeletal muscle, is a promising treatment method for muscle reconstruction. Although various fabrication techniques for skeletal muscle using 3D bio-printers are studied, it is still challenging to build a functional muscle structure. A strategy using microvalve-assisted coaxial 3D bioprinting in consideration of functional skeletal muscle fabrication is reported. The unit (artificial muscle fascicle: AMF) of muscle mimetic tissue is composed of a core filled with medium-based C2C12 myoblast aggregates as a role of muscle fibers and a photo cross-linkable hydrogel-based shell as a role of connective tissue in muscles that enhances printability and cell adhesion and proliferation. Especially, a microvalve system is applied for the core part with even cell distribution and strong cell-cell interaction. This system enhances myotube formation and consequently shows spontaneous contraction. A multi-printed AMF (artificial muscle tissue: AMT) as a piece of muscle is implanted into the anterior tibia (TA) muscle defect site of immunocompromised rats. As a result, the TA-implanted AMT responds to electrical stimulation and represents histologically regenerated muscle tissue. This microvalve-assisted coaxial 3D bioprinting shows a significant step forward to mimicking native skeletal muscle tissue.
Collapse
Affiliation(s)
- Hanna Lee
- Nano-Bio Regenerative Medical Institute, College of Medicine, Hallym University, 1 Hallymdaehak-gil, Chuncheon, Gangwon-do, 24252, Republic of Korea
| | - Soon Hee Kim
- Nano-Bio Regenerative Medical Institute, College of Medicine, Hallym University, 1 Hallymdaehak-gil, Chuncheon, Gangwon-do, 24252, Republic of Korea
| | - Ji Seung Lee
- Nano-Bio Regenerative Medical Institute, College of Medicine, Hallym University, 1 Hallymdaehak-gil, Chuncheon, Gangwon-do, 24252, Republic of Korea
| | - Young Jin Lee
- Nano-Bio Regenerative Medical Institute, College of Medicine, Hallym University, 1 Hallymdaehak-gil, Chuncheon, Gangwon-do, 24252, Republic of Korea
| | - Ok Joo Lee
- Nano-Bio Regenerative Medical Institute, College of Medicine, Hallym University, 1 Hallymdaehak-gil, Chuncheon, Gangwon-do, 24252, Republic of Korea
| | - Olatunji Ajiteru
- Nano-Bio Regenerative Medical Institute, College of Medicine, Hallym University, 1 Hallymdaehak-gil, Chuncheon, Gangwon-do, 24252, Republic of Korea
| | - Md Tipu Sultan
- Nano-Bio Regenerative Medical Institute, College of Medicine, Hallym University, 1 Hallymdaehak-gil, Chuncheon, Gangwon-do, 24252, Republic of Korea
| | - Suk Woo Lee
- Department of Obstetrics and Gynecology, Hallym University Sacred Heart Hospital, Anyang, 14068, Republic of Korea
| | - Chan Hum Park
- Nano-Bio Regenerative Medical Institute, College of Medicine, Hallym University, 1 Hallymdaehak-gil, Chuncheon, Gangwon-do, 24252, Republic of Korea.,Department of Otorhinolaryngology-Head and Neck Surgery, Chuncheon Sacred Heart Hospital, School of Medicine, Hallym University, Chuncheon, 24252, Republic of Korea
| |
Collapse
|
19
|
Lee SH, Kang MS, Jeon S, Jo HJ, Hong SW, Kim B, Han DW. 3D bioprinting of human mesenchymal stem cells-laden hydrogels incorporating MXene for spontaneous osteodifferentiation. Heliyon 2023; 9:e14490. [PMID: 36994406 PMCID: PMC10040522 DOI: 10.1016/j.heliyon.2023.e14490] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 03/07/2023] [Accepted: 03/08/2023] [Indexed: 03/16/2023] Open
Abstract
Contemporary advances in three-dimensional (3D) bioprinting technologies have enabled the fabrication of tailored live 3D tissue mimetics. Furthermore, the development of advanced bioink materials has been highlighted to accurately reproduce the composition of a native extracellular matrix and mimic the intrinsic properties of laden cells. Recent research has shown that MXene is one of promising nanobiomaterials with osteogenic activity for bone grafts and scaffolds due to its unique atomic structure of three titanium layers between two carbon layers. In this study, the MXene-incorporated gelatin methacryloyl (GelMA) and hyaluronic acid methacryloyl (HAMA) (i.e., GelMA/HAMA-MXene) bioinks were prepared to explore if they have the potential to enable the spontaneous osteodifferentiation of human mesenchymal stem cells (hMSCs) when the hMSCs-laden GelMA/HAMA-MXene bioinks were 3D printed. The physicochemical and rheological characteristics of the GelMA/HAMA-MXene hydrogels were proven to be unprecedentedly favorable supportive matrices suited for the growth and survival of hMSCs. Furthermore, hMSCs were shown to spontaneously differentiate into osteoblasts within GelMA-HAMA/MXene composites to provide favorable microenvironments for osteogenesis. Therefore, our results suggest that the remarkable biofunctional advantages of the MXene-incorporated GelMA/HAMA bioink can be utilized in a wide range of strategies for the development of effective scaffolds in bone tissue regeneration.
Collapse
Affiliation(s)
- Seok Hyun Lee
- Department of Cogno-Mechatronics Engineering, College of Nanoscience and Nanotechnology, Pusan National University, Busan, 46241, Republic of Korea
| | - Moon Sung Kang
- Department of Cogno-Mechatronics Engineering, College of Nanoscience and Nanotechnology, Pusan National University, Busan, 46241, Republic of Korea
| | - Sangheon Jeon
- Department of Cogno-Mechatronics Engineering, College of Nanoscience and Nanotechnology, Pusan National University, Busan, 46241, Republic of Korea
| | - Hyo Jung Jo
- Department of Cogno-Mechatronics Engineering, College of Nanoscience and Nanotechnology, Pusan National University, Busan, 46241, Republic of Korea
| | - Suck Won Hong
- Department of Cogno-Mechatronics Engineering, College of Nanoscience and Nanotechnology, Pusan National University, Busan, 46241, Republic of Korea
- Engineering Research Center for Color-Modulated Extra-Sensory Perception Technology, Pusan National University, Busan, 46241, Republic of Korea
| | - Bongju Kim
- Dental Life Science Research Institute / Innovation Research & Support Center for Dental Science, Seoul 8 National University Dental Hospital, Seoul, 03080, Republic of Korea
- Corresponding author.
| | - Dong-Wook Han
- Department of Cogno-Mechatronics Engineering, College of Nanoscience and Nanotechnology, Pusan National University, Busan, 46241, Republic of Korea
- BIO-IT Fusion Technology Research Institute, Pusan National University, Busan, 46241, Republic of Korea
- Corresponding author. Department of Cogno-Mechatronics Engineering, College of Nanoscience and Nanotechnology, Pusan National University, Busan 46241, Republic of Korea.
| |
Collapse
|
20
|
Hwang SH, Kim J, Heo C, Yoon J, Kim H, Lee SH, Park HW, Heo MS, Moon HE, Kim C, Paek SH, Jang J. 3D printed multi-growth factor delivery patches fabricated using dual-crosslinked decellularized extracellular matrix-based hybrid inks to promote cerebral angiogenesis. Acta Biomater 2023; 157:137-148. [PMID: 36460287 DOI: 10.1016/j.actbio.2022.11.050] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 11/04/2022] [Accepted: 11/23/2022] [Indexed: 12/05/2022]
Abstract
Generally, brain angiogenesis is a tightly regulated process, which scarcely occurred in the absence of specific pathological conditions. Delivery of exogenous angiogenic factors enables the induction of desired angiogenesis by stimulating neovasculature formation. However, effective strategies of mimicking the angiogenesis process with exogenous factors have not yet been fully explored. Herein, we develop a 3D printed spatiotemporally compartmentalized cerebral angiogenesis inducing (SCAI) hydrogel patch, releasing dual angiogenic growth factors (GFs), using extracellular matrix-based hybrid inks. We introduce a new hybrid biomaterial-based ink for printing patches through dual crosslinking mechanisms: Chemical crosslinking with aza-Michael addition reaction with combining methacrylated hyaluronic acid (HAMA) and vascular-tissue-derived decellularized extracellular matrix (VdECM), and thermal crosslinking of VdECM. 3D printing technology, a useful approach with fabrication versatility with customizable systems and multiple biomaterials, is adopted to print three-layered hydrogel patch with spatially separated dual GFs as outer- and inner-layers that provide tunable release profiles of multiple GFs and fabrication versatility. Consequently, these layers of the patch spatiotemporally separated with dual GFs induce excellent neovascularization in the brain area, monitored by label-free photoacoustic microscopy in vivo. The developed multi-GFs releasing patch may offer a promising therapeutic approach of spatiotemporal drugs releasing such as cerebral ischemia, ischemic heart diseases, diabetes, and even use as vaccines. STATEMENT OF SIGNIFICANCE: Effective strategies of mimicking the angiogenesis process with exogenous factors have not yet been fully explored. In this study, we develop a 3D printed spatiotemporally compartmentalized cerebral angiogenesis inducing (SCAI) hydrogel patch, releasing dual angiogenic growth factors (GFs) using extracellular matrix-based hybrid inks. We introduce a new hybrid biomaterial-based ink through dual crosslinking mechanisms: Chemical crosslinking with aza-Michael addition, and thermal crosslinking. 3D printing technology is adopted to print three-layered hydrogel patch with spatially separated dual GFs as outer- and inner-layers that provide tunable release profiles of multiple GFs and fabrication versatility. Consequently, these layers of the patch spatiotemporally separated with dual GFs induce excellent neovascularization in the brain area, monitored by photoacoustic microscopy in vivo.
Collapse
Affiliation(s)
- Seung Hyeon Hwang
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), 77 Cheongam-Ro, Nam-Gu, Pohang 37673, Republic of Korea
| | - Jongbeom Kim
- Department of Convergence IT Engineering, Pohang University of Science and Technology (POSTECH), 77 Cheongam-Ro, Nam-Gu, Pohang 37673, Republic of Korea
| | - Chaejeong Heo
- Center for Integrated Nanostructure Physics (CINAP), Institute for Basic Science (IBS), Suwon 16419, Republic of Korea
| | - Jungbin Yoon
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), 77 Cheongam-Ro, Nam-Gu, Pohang 37673, Republic of Korea
| | - Hyeonji Kim
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), 77 Cheongam-Ro, Nam-Gu, Pohang 37673, Republic of Korea
| | - Se-Hwan Lee
- Department of Convergence IT Engineering, Pohang University of Science and Technology (POSTECH), 77 Cheongam-Ro, Nam-Gu, Pohang 37673, Republic of Korea
| | - Hyung Woo Park
- Department of Neurosurgery, Cancer Research Institute, Ischemia/Hypoxia Disease Institute, Seoul National University, College of Medicine, Seoul 03080, Republic of Korea; Advanced Institute of Convergence Technology, Seoul National University, Suwon 16229, Republic of Korea
| | - Man Seung Heo
- Department of Neurosurgery, Cancer Research Institute, Ischemia/Hypoxia Disease Institute, Seoul National University, College of Medicine, Seoul 03080, Republic of Korea; Advanced Institute of Convergence Technology, Seoul National University, Suwon 16229, Republic of Korea
| | - Hyo Eun Moon
- Department of Neurosurgery, Cancer Research Institute, Ischemia/Hypoxia Disease Institute, Seoul National University, College of Medicine, Seoul 03080, Republic of Korea; Advanced Institute of Convergence Technology, Seoul National University, Suwon 16229, Republic of Korea
| | - Chulhong Kim
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), 77 Cheongam-Ro, Nam-Gu, Pohang 37673, Republic of Korea; Department of Convergence IT Engineering, Pohang University of Science and Technology (POSTECH), 77 Cheongam-Ro, Nam-Gu, Pohang 37673, Republic of Korea; Departments of Electrical Engineering, and Medical Device Innovation Center, Pohang University of Science and Technology (POSTECH), 77 Cheongam-ro, Nam-gu, Pohang 37673, Republic of Korea; School of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science and Technology (POSTECH), 77 Cheongam-Ro, Nam-Gu, Pohang 37673, Republic of Korea; Institute for Convergence Research and Education in Advanced Technology, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea.
| | - Sun Ha Paek
- Department of Neurosurgery, Cancer Research Institute, Ischemia/Hypoxia Disease Institute, Seoul National University, College of Medicine, Seoul 03080, Republic of Korea; Advanced Institute of Convergence Technology, Seoul National University, Suwon 16229, Republic of Korea.
| | - Jinah Jang
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), 77 Cheongam-Ro, Nam-Gu, Pohang 37673, Republic of Korea; Department of Convergence IT Engineering, Pohang University of Science and Technology (POSTECH), 77 Cheongam-Ro, Nam-Gu, Pohang 37673, Republic of Korea; School of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science and Technology (POSTECH), 77 Cheongam-Ro, Nam-Gu, Pohang 37673, Republic of Korea; Institute for Convergence Research and Education in Advanced Technology, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea.
| |
Collapse
|
21
|
Hydrogel-Based Tissue-Mimics for Vascular Regeneration and Tumor Angiogenesis. Regen Med 2023. [DOI: 10.1007/978-981-19-6008-6_8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
|
22
|
Chen Z, Wang L, Guo C, Qiu M, Cheng L, Chen K, Qi J, Deng L, He C, Li X, Yan Y. Vascularized polypeptide hydrogel modulates macrophage polarization for wound healing. Acta Biomater 2023; 155:218-234. [PMID: 36396041 DOI: 10.1016/j.actbio.2022.11.002] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Revised: 10/18/2022] [Accepted: 11/01/2022] [Indexed: 11/16/2022]
Abstract
Wound repair involves a sophisticated process that includes angiogenesis, immunoregulation and collagen deposition. However, weak revascularization performance and the lack of biochemical cues to trigger immunomodulatory function currently limit biomaterial applications for skin regeneration and tissue engineering. Herein, we fabricate a new bioactive polypeptide hydrogel (QK-SF) constituted by silk fibroin (SF) and a vascular endothelial growth factor mimetic peptide KLTWQELYQLKYKGI (QK) for tissue regeneration by simultaneously promoting vascularization and macrophage polarization. Our results showed that this QK-SF hydrogel can be prepared via an easy manufacturing process, and exhibited good gel stability and low cytotoxicity to cultured human umbilical vein endothelial cells (HUVECs) via both live/dead and cell counting kit-8 assays. Importantly, this QK-SF hydrogel triggered macrophage polarization from M1 into M2, as exemplified by the enhanced expression of the M2 marker and decreased expression of the M1 marker in RAW264.7 cells. Furthermore, the QK-SF hydrogel showed high capacity for inducing endothelial growth, migration and angiogenesis, which were proved by increased expression of angiogenesis-related genes in HUVECs. Consistent with in vitro findings, in vivo data show that the QK-SF hydrogel promoted M2 polarization, keratinocyte differentiation, and collagen deposition in the mouse skin wound model in immunohistochemistry assay. Furthermore, this QK-SF hydrogel can reduce inflammation, induce angiogenesis and promote wound healing as exemplified by the increased vessel formation and decreased wound area in the mouse skin wound model. Altogether, these results indicate that the bioactive QK-SF hydrogel plays dual functional roles in promoting angiogenesis and immunoregulation for tissue regeneration. STATEMENT OF SIGNIFICANCE: The QK-SF hydrogel plays dual functional roles in promoting angiogenesis and immunoregulation for tissue repair and wound healing. The QK-SF hydrogel can be prepared via an easy manufacturing process, and exhibited good gel stability and low cytotoxicity to cultured HUVECs. The QK-SF hydrogel triggered macrophage polarization from M1 into M2. The QK-SF hydrogel showed high capacity for inducing endothelial growth, migration and angiogenesis. The QK-SF hydrogel promoted M2 polarization, keratinocyte differentiation, and collagen deposition.
Collapse
Affiliation(s)
- Zhijie Chen
- Department of Orthopaedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, PR China; Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai 200025, PR China
| | - Lianlian Wang
- College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou 215123, PR China
| | - Changjun Guo
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai 200025, PR China
| | - Minglong Qiu
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai 200025, PR China
| | - Liang Cheng
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai 200025, PR China
| | - Kaizhe Chen
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai 200025, PR China
| | - Jin Qi
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai 200025, PR China
| | - Lianfu Deng
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai 200025, PR China
| | - Chuan He
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai 200025, PR China.
| | - Xinming Li
- College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou 215123, PR China.
| | - Yufei Yan
- Department of Orthopaedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, PR China; Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai 200025, PR China.
| |
Collapse
|
23
|
Kutlehria S, D'Souza A, Bleier BS, Amiji MM. Role of 3D Printing in the Development of Biodegradable Implants for Central Nervous System Drug Delivery. Mol Pharm 2022; 19:4411-4427. [PMID: 36154128 DOI: 10.1021/acs.molpharmaceut.2c00344] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Increased life expectancy has led to a rise in age-related disorders including neurological diseases such as Alzheimer's disease and Parkinson's disease. Limited progress has been made in the development of clinically translatable therapies for these central nervous system (CNS) diseases. Challenges including the blood-brain barrier, brain complexity, and comorbidities in the elderly population are some of the contributing factors toward lower success rates. Various invasive and noninvasive ways are being employed to deliver small and large molecules across the brain. Biodegradable, implantable drug-delivery systems have gained lot of interest due to advantages such as sustained and targeted delivery, lower side effects, and higher patient compliance. 3D printing is a novel additive manufacturing technique where various materials and printing techniques can be used to fabricate implants with the desired complexity in terms of mechanical properties, shapes, or release profiles. This review discusses an overview of various types of 3D-printing techniques and illustrative examples of the existing literature on 3D-printed systems for CNS drug delivery. Currently, there are various technical and regulatory impediments that need to be addressed for successful translation from the bench to the clinical stage. Overall, 3D printing is a transformative technology with great potential in advancing customizable drug treatment in a high-throughput manner.
Collapse
Affiliation(s)
- Shallu Kutlehria
- Department of Pharmaceutical Sciences, School of Pharmacy, Northeastern University, Boston, Massachusetts 02115, United States
| | - Anisha D'Souza
- Department of Pharmaceutical Sciences, School of Pharmacy, Northeastern University, Boston, Massachusetts 02115, United States.,Department of Otolaryngology, Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Benjamin S Bleier
- Department of Otolaryngology, Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Mansoor M Amiji
- Department of Pharmaceutical Sciences, School of Pharmacy, Northeastern University, Boston, Massachusetts 02115, United States.,Department of Chemical Engineering, College of Engineering, Northeastern University, Boston, Massachusetts 02115, United States
| |
Collapse
|
24
|
Garkal A, Bangar P, Rajput A, Pingale P, Dhas N, Sami A, Mathur K, Joshi S, Dhuri S, Parikh D, Mutalik S, Mehta T. Long-acting formulation strategies for protein and peptide delivery in the treatment of PSED. J Control Release 2022; 350:538-568. [PMID: 36030993 DOI: 10.1016/j.jconrel.2022.08.040] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 08/18/2022] [Accepted: 08/21/2022] [Indexed: 12/17/2022]
Abstract
The invigoration of protein and peptides in serious eye disease includes age-related macular degeneration, choroidal neovascularization, retinal neovascularization, and diabetic retinopathy. The transportation of macromolecules like aptamers, recombinant proteins, and monoclonal antibodies to the posterior segment of the eye is challenging due to their high molecular weight, rapid degradation, and low solubility. Moreover, it requires frequent administration for prolonged therapy. The long-acting novel formulation strategies are helpful to overcome these issues and provide superior therapy. It avoids frequent administration, improves stability, high retention time, and avoids burst release. This review briefly enlightens posterior segments of eye diseases with their diagnosis techniques and treatments. This article mainly focuses on recent advanced approaches like intravitreal implants and injectables, electrospun injectables, 3D printed drug-loaded implants, nanostructure thin-film polymer devices encapsulated cell technology-based intravitreal implants, injectable and depots, microneedles, PDS with ranibizumab, polymer nanoparticles, inorganic nanoparticles, hydrogels and microparticles for delivering macromolecules in the eye for intended therapy. Furthermore, novel techniques like aptamer, small Interference RNA, and stem cell therapy were also discussed. It is predicted that these systems will make revolutionary changes in treating posterior segment eye diseases in future.
Collapse
Affiliation(s)
- Atul Garkal
- Department of Pharmaceutics, Institute of Pharmacy, Nirma University, Ahmedabad, Gujarat 382481, India
| | - Priyanka Bangar
- Department of Pharmaceutics, Institute of Pharmacy, Nirma University, Ahmedabad, Gujarat 382481, India
| | - Amarjitsing Rajput
- Department of Pharmaceutics, Bharti Vidyapeeth Deemed University, Poona College of Pharmacy, Pune, Maharashtra 411038, India
| | - Prashant Pingale
- Department of Pharmaceutics, GES's Sir Dr. M.S. Gosavi College of Pharmaceutical Education and Research, Nashik, Maharashtra 422005, India
| | - Namdev Dhas
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education (MAHE), Manipal, Karnataka 576104, India
| | - Anam Sami
- Department of Pharmaceutics, Institute of Pharmacy, Nirma University, Ahmedabad, Gujarat 382481, India
| | - Khushboo Mathur
- Department of Pharmaceutics, Institute of Pharmacy, Nirma University, Ahmedabad, Gujarat 382481, India
| | - Shubham Joshi
- Department of Pharmaceutics, Institute of Pharmacy, Nirma University, Ahmedabad, Gujarat 382481, India
| | - Sonika Dhuri
- Department of Pharmaceutics, Institute of Pharmacy, Nirma University, Ahmedabad, Gujarat 382481, India
| | - Dhaivat Parikh
- Department of Pharmaceutics, Institute of Pharmacy, Nirma University, Ahmedabad, Gujarat 382481, India
| | - Srinivas Mutalik
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education (MAHE), Manipal, Karnataka 576104, India
| | - Tejal Mehta
- Department of Pharmaceutics, Institute of Pharmacy, Nirma University, Ahmedabad, Gujarat 382481, India.
| |
Collapse
|
25
|
Mishchenko TA, Klimenko MO, Kuznetsova AI, Yarkov RS, Savelyev AG, Sochilina AV, Mariyanats AO, Popov VK, Khaydukov EV, Zvyagin AV, Vedunova MV. 3D-printed hyaluronic acid hydrogel scaffolds impregnated with neurotrophic factors (BDNF, GDNF) for post-traumatic brain tissue reconstruction. Front Bioeng Biotechnol 2022; 10:895406. [PMID: 36091441 PMCID: PMC9453866 DOI: 10.3389/fbioe.2022.895406] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2022] [Accepted: 07/07/2022] [Indexed: 11/13/2022] Open
Abstract
Brain tissue reconstruction posttraumatic injury remains a long-standing challenge in neurotransplantology, where a tissue-engineering construct (scaffold, SC) with specific biochemical properties is deemed the most essential building block. Such three-dimensional (3D) hydrogel scaffolds can be formed using brain-abundant endogenous hyaluronic acid modified with glycidyl methacrylate by employing our proprietary photopolymerisation technique. Herein, we produced 3D hyaluronic scaffolds impregnated with neurotrophic factors (BDNF, GDNF) possessing 600 kPa Young’s moduli and 336% swelling ratios. Stringent in vitro testing of fabricated scaffolds using primary hippocampal cultures revealed lack of significant cytotoxicity: the number of viable cells in the SC+BDNF (91.67 ± 1.08%) and SC+GDNF (88.69 ± 1.2%) groups was comparable to the sham values (p > 0.05). Interestingly, BDNF-loaded scaffolds promoted the stimulation of neuronal process outgrowth during the first 3 days of cultures development (day 1: 23.34 ± 1.46 µm; day 3: 37.26 ± 1.98 µm, p < 0.05, vs. sham), whereas GDNF-loaded scaffolds increased the functional activity of neuron-glial networks of cultures at later stages of cultivation (day 14) manifested in a 1.3-fold decrease in the duration coupled with a 2.4-fold increase in the frequency of Ca2+ oscillations (p < 0.05, vs. sham). In vivo studies were carried out using C57BL/6 mice with induced traumatic brain injury, followed by surgery augmented with scaffold implantation. We found positive dynamics of the morphological changes in the treated nerve tissue in the post-traumatic period, where the GDNF-loaded scaffolds indicated more favorable regenerative potential. In comparison with controls, the physiological state of the treated mice was improved manifested by the absence of severe neurological deficit, significant changes in motor and orienting-exploratory activity, and preservation of the ability to learn and retain long-term memory. Our results suggest in favor of biocompatibility of GDNF-loaded scaffolds, which provide a platform for personalized brain implants stimulating effective morphological and functional recovery of nerve tissue after traumatic brain injury.
Collapse
Affiliation(s)
- Tatiana A. Mishchenko
- Institute of Biology and Biomedicine, National Research Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod, Russia
| | - Maria O. Klimenko
- Institute of Biology and Biomedicine, National Research Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod, Russia
| | - Alisa I. Kuznetsova
- Institute of Biology and Biomedicine, National Research Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod, Russia
| | - Roman S. Yarkov
- Institute of Biology and Biomedicine, National Research Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod, Russia
| | - Alexander G. Savelyev
- Federal Scientific Research Centre “Crystallography and Photonics”, Russian Academy of Sciences, Troitsk-Moscow, Russia
- Sechenov First Moscow State Medical University, Moscow, Russia
| | - Anastasia V. Sochilina
- Federal Scientific Research Centre “Crystallography and Photonics”, Russian Academy of Sciences, Troitsk-Moscow, Russia
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry RAS, Moscow, Russia
| | - Alexandra O. Mariyanats
- Federal Scientific Research Centre “Crystallography and Photonics”, Russian Academy of Sciences, Troitsk-Moscow, Russia
| | - Vladimir K. Popov
- Federal Scientific Research Centre “Crystallography and Photonics”, Russian Academy of Sciences, Troitsk-Moscow, Russia
| | - Evgeny V. Khaydukov
- Federal Scientific Research Centre “Crystallography and Photonics”, Russian Academy of Sciences, Troitsk-Moscow, Russia
- Sechenov First Moscow State Medical University, Moscow, Russia
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry RAS, Moscow, Russia
| | - Andrei V. Zvyagin
- Institute of Biology and Biomedicine, National Research Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod, Russia
- Sechenov First Moscow State Medical University, Moscow, Russia
- MQ Photonics Centre, Macquarie University, Sydney, NSW, Australia
| | - Maria V. Vedunova
- Institute of Biology and Biomedicine, National Research Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod, Russia
- *Correspondence: Maria V. Vedunova,
| |
Collapse
|
26
|
Wang G, Yuan N, Li N, Wei Q, Qian Y, Zhang J, Qin M, Wang Y, Dong S. Vascular Endothelial Growth Factor Mimetic Peptide and Parathyroid Hormone (1-34) Delivered via a Blue-Light-Curable Hydrogel Synergistically Accelerate Bone Regeneration. ACS APPLIED MATERIALS & INTERFACES 2022; 14:35319-35332. [PMID: 35881151 DOI: 10.1021/acsami.2c06159] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Safe and effective biomaterials are in urgent clinical need for tissue regeneration and bone repair. While numerous advances have been made on hydrogels promoting osteogenesis in bone formation, co-stimulation of the angiogenic pathways in this process remains to be exploited. Here, we have developed a gelatin-based blue-light-curable hydrogel system, functionalized with an angiogenic vascular endothelial growth factor (VEGF) mimetic peptide, KLTWQELYQLKYKGI (KLT), and an osteoanabolic peptide, parathyroid hormone (PTH) 1-34. We have discovered that the covalent modification of gelatin scaffold with peptides can modulate the physical properties and biological activities of the produced hydrogels. Furthermore, we have demonstrated that those two peptides orchestrate synergistically and promote bone regeneration in a rat cranial bone defect model with remarkable efficacy. This dual-peptide-functionalized hydrogel system may serve as a promising lead to functional biomaterials in bone repair and tissue engineering.
Collapse
Affiliation(s)
- Guiyan Wang
- National Engineering Laboratory for Digital and Material Technology of Stomatology, National Clinical Research Center for Oral Diseases, Beijing Key Laboratory of Digital Stomatology, and Department of Pediatric Dentistry, Peking University School and Hospital of Stomatology, Beijing 100081, China
| | - Ning Yuan
- State Key Laboratory of Natural and Biomimetic Drugs, Chemical Biology Center, and School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Ningyu Li
- Department of Oral Comprehensive Treatment, Jilin University School and Hospital of Stomatology, Changchun 130021, China
| | - Qijia Wei
- State Key Laboratory of Natural and Biomimetic Drugs, Chemical Biology Center, and School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Yuping Qian
- Department of Prosthodontics, Jilin University School and Hospital of Stomatology, Changchun 130021, China
| | - Jun Zhang
- State Key Laboratory of Natural and Biomimetic Drugs, Chemical Biology Center, and School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Man Qin
- National Engineering Laboratory for Digital and Material Technology of Stomatology, National Clinical Research Center for Oral Diseases, Beijing Key Laboratory of Digital Stomatology, and Department of Pediatric Dentistry, Peking University School and Hospital of Stomatology, Beijing 100081, China
| | - Yuguang Wang
- National Engineering Laboratory for Digital and Material Technology of Stomatology, National Clinical Research Center for Oral Diseases, Beijing Key Laboratory of Digital Stomatology, and Department of Pediatric Dentistry, Peking University School and Hospital of Stomatology, Beijing 100081, China
| | - Suwei Dong
- State Key Laboratory of Natural and Biomimetic Drugs, Chemical Biology Center, and School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| |
Collapse
|
27
|
Applications and Mechanisms of Stimuli-Responsive Hydrogels in Traumatic Brain Injury. Gels 2022; 8:gels8080482. [PMID: 36005083 PMCID: PMC9407546 DOI: 10.3390/gels8080482] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 07/26/2022] [Accepted: 07/28/2022] [Indexed: 02/01/2023] Open
Abstract
Traumatic brain injury (TBI) is a global neurotrauma with high morbidity and mortality that seriously threatens the life quality of patients and causes heavy burdens to families, healthcare institutions, and society. Neuroinflammation and oxidative stress can further aggravate neuronal cell death, hinder functional recovery, and lead to secondary brain injury. In addition, the blood–brain barrier prevents drugs from entering the brain tissue, which is not conducive to the recovery of TBI. Due to their high water content, biodegradability, and similarity to the natural extracellular matrix (ECM), hydrogels are widely used for the delivery and release of various therapeutic agents (drugs, natural extracts, and cells, etc.) that exhibit beneficial therapeutic efficacy in tissue repair, such as TBI. Stimuli-responsive hydrogels can undergo reversible or irreversible changes in properties, structures, and functions in response to internal/external stimuli or physiological/pathological environmental stimuli, and further improve the therapeutic effects on diseases. In this paper, we reviewed the common types of stimuli-responsive hydrogels and their applications in TBI, and further analyzed the therapeutic effects of hydrogels in TBI, such as pro-neurogenesis, anti-inflammatory, anti-apoptosis, anti-oxidation, and pro-angiogenesis. Our study may provide strategies for the treatment of TBI by using stimuli-responsive hydrogels.
Collapse
|
28
|
A dual crosslinked hydrogel-mediated integrated peptides and BMSC therapy for myocardial regeneration. J Control Release 2022; 347:127-142. [PMID: 35460706 DOI: 10.1016/j.jconrel.2022.04.010] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 03/07/2022] [Accepted: 04/08/2022] [Indexed: 12/27/2022]
Abstract
The efficacy of myocardial regeneration strategies for myocardial infarction (MI) is significantly compromised by the complex structure and microenvironment of the myocardium. Although tissue engineering strategies based on cell therapy and/or pro-angiogenesis can somewhat improve cardiac function, the lack of proper myocardial materials that can withstand sustained deformability and adaptable mechanical properties severely affects myocardial wall integrity, systolic-diastolic cycles, and regeneration. Herein, we developed an integrated single "all-in-one" in situ dual crosslinking conductive hydrogel with favorable treatment properties termed as MaHA/B-G-SH/Fe3+ by ionic interactions and chemical covalency based on modified hyaluronic acid (HA), gelatin (G), and Fe3+. The resulting dual crosslinking dynamic hydrogel not only provides self-healing and mechanical properties adapted to the myocardial systolic-diastolic cycle with simultaneous electrical signal transmission to fibrous islands and normal tissue, but also leads to significant increase of the myocardial wall thickness very close to that of normal myocardium upon one single injection with complete degradation within 28 days. Notably, the hydrogel covalently conjugated with a tailored peptide sequence of GGR-KLT and encapsulated with bone mesenchymal stem cells (BMSCs) was further used for in situ injection in a rat MI model, which exhibited (i) efficient inhibition of excessive matrix degradation dependent on early MMP-2 expression, (ii) triggered on-demand release of KLT for at least 14 days and significant promotion of angiogenesis, and (iii) synergistic BMSCs considerably enhanced myocardial regeneration within 28 days. Taken together, the dual crosslinking conductive hydrogel-mediated synergistic peptide and cell therapy provides comprehensive recovery and regeneration of the structure and function of the injured myocardium, thus demonstrating great potential for clinical translations.
Collapse
|
29
|
Shokrani H, Shokrani A, Sajadi SM, Seidi F, Mashhadzadeh AH, Rabiee N, Saeb MR, Aminabhavi T, Webster TJ. Cell-Seeded Biomaterial Scaffolds: The Urgent Need for Unanswered Accelerated Angiogenesis. Int J Nanomedicine 2022; 17:1035-1068. [PMID: 35309965 PMCID: PMC8927652 DOI: 10.2147/ijn.s353062] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 02/22/2022] [Indexed: 12/12/2022] Open
Abstract
One of the most arduous challenges in tissue engineering is neovascularization, without which there is a lack of nutrients delivered to a target tissue. Angiogenesis should be completed at an optimal density and within an appropriate period of time to prevent cell necrosis. Failure to meet this challenge brings about poor functionality for the tissue in comparison with the native tissue, extensively reducing cell viability. Prior studies devoted to angiogenesis have provided researchers with some biomaterial scaffolds and cell choices for angiogenesis. For example, while most current angiogenesis approaches require a variety of stimulatory factors ranging from biomechanical to biomolecular to cellular, some other promising stimulatory factors have been underdeveloped (such as electrical, topographical, and magnetic). When it comes to choosing biomaterial scaffolds in tissue engineering for angiogenesis, key traits rush to mind including biocompatibility, appropriate physical and mechanical properties (adhesion strength, shear stress, and malleability), as well as identifying the appropriate biomaterial in terms of stability and degradation profile, all of which may leave essential trace materials behind adversely influencing angiogenesis. Nevertheless, the selection of the best biomaterial and cells still remains an area of hot dispute as such previous studies have not sufficiently classified, integrated, or compared approaches. To address the aforementioned need, this review article summarizes a variety of natural and synthetic scaffolds including hydrogels that support angiogenesis. Furthermore, we review a variety of cell sources utilized for cell seeding and influential factors used for angiogenesis with a concentrated focus on biomechanical factors, with unique stimulatory factors. Lastly, we provide a bottom-to-up overview of angiogenic biomaterials and cell selection, highlighting parameters that need to be addressed in future studies.
Collapse
Affiliation(s)
- Hanieh Shokrani
- Department of Chemical Engineering, Sharif University of Technology, Tehran, Iran
| | - Amirhossein Shokrani
- Department of Mechanical Engineering, Sharif University of Technology, Tehran, Iran
| | - S Mohammad Sajadi
- Department of Nutrition, Cihan University-Erbil, Erbil, 625, Iraq
- Department of Phytochemistry, SRC, Soran University, Soran, KRG, 624, Iraq
- Correspondence: S Mohammad Sajadi; Navid Rabiee, Email ; ;
| | - Farzad Seidi
- Jiangsu Co–Innovation Center for Efficient Processing and Utilization of Forest Resources and International Innovation Center for Forest Chemicals and Materials, Nanjing Forestry University, Nanjing, 210037, People’s Republic of China
| | - Amin Hamed Mashhadzadeh
- Mechanical and Aerospace Engineering, School of Engineering and Digital Sciences, Nazarbayev University, Nur-Sultan, 010000, Kazakhstan
| | - Navid Rabiee
- Department of Physics, Sharif University of Technology, Tehran, Iran
- School of Engineering, Macquarie University, Sydney, New South Wales, 2109, Australia
| | - Mohammad Reza Saeb
- Department of Polymer Technology, Faculty of Chemistry, Gdańsk University of Technology, Gdańsk, Poland
| | - Tejraj Aminabhavi
- School of Advanced Sciences, KLE Technological University, Hubballi, Karnataka, 580 031, India
- Department of Chemistry, Karnatak University, Dharwad, 580 003, India
| | - Thomas J Webster
- School of Health Sciences and Biomedical Engineering, Hebei University, Tianjin, People’s Republic of China
- Center for Biomaterials, Vellore Institute of Technology, Vellore, India
| |
Collapse
|
30
|
Liu C, Peng Z, Xu H, gao H, Li J, jin Y, wang Y, Wang C, liu Y, hu Y, jiang C, Guo J, Zhu L. 3D print Platelet-rich plasma loaded scaffold with sustained cytokine release for bone defect repair. Tissue Eng Part A 2022; 28:700-711. [PMID: 35152730 DOI: 10.1089/ten.tea.2021.0211] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Chun Liu
- Department of Spinal Surgery, Orthopedic Medical Center, Zhujiang Hospital, Southern Medical University, Guangzhou, China,
| | - Ziyue Peng
- Department of Spinal Surgery, Orthopedic Medical Center, Zhujiang Hospital, Southern Medical University, Guangzhou, China,
| | - Haixia Xu
- Department of Spinal Surgery, Orthopedic Medical Center, Zhujiang Hospital, Southern Medical University, Guangzhou, China,
| | - Huiling gao
- Department of Spinal Surgery, Orthopedic Medical Center, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China, Guangzhou, Guangdong, China,
| | - Jianjun Li
- Department of Spinal Surgery, Orthopedic Medical Center, Zhujiang Hospital, Southern Medical University, Guangzhou, China,
| | - yanglei jin
- The Fourth Affiliated Hospital Zhejiang University School of Medicine, 593059, Yiwu, Zhejiang, China,
| | - yihan wang
- Department of Spinal Surgery, Orthopedic Medical Center, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China, guangzhou, China,
| | - Chengqiang Wang
- Department of Spinal Surgery, Orthopedic Medical Center, Zhujiang Hospital, Southern Medical University, Department of Spinal Surgery, Orthopedic Medical Center, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China, No. 253 Industrial Avenue, Guangzhou, China, 510280,
| | - yang liu
- Xiang Yang Central Hospital, Affiliated Hospital of Hubei University of Art and Science, xiangyang, China,
| | - yunteng hu
- Department of Spinal Surgery, Orthopedic Medical Center, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China, guangzhou, China,
| | - cong jiang
- Department of Spinal Surgery, Orthopedic Medical Center, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China, guangzhou, China,
| | - Jiasong Guo
- Department of Spinal Surgery, Orthopedic Medical Center, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- Department of Histology and Embryology, Southern Medical University, Guangzhou, China
- Key Laboratory of Tissue Construction and Detection of Guangdong Province, Guangzhou, China
- Institute of Bone Biology, Academy of Orthopaedics, Guangdong Province, Guangzhou, China,
| | - Lixin Zhu
- Department of Spinal Surgery, Orthopedic Medical Center, Zhujiang Hospital, Southern Medical University, No. 253, Middle Gongye Avenue, Guangzhou, China, 510280,
| |
Collapse
|
31
|
Park SH, Ju HJ, Ji YB, Shah M, Min BH, Choi HS, Choi S, Kim MS. Endogenous Stem Cell-Based In Situ Tissue Regeneration Using Electrostatically Interactive Hydrogel with a Newly Discovered Substance P Analog and VEGF-Mimicking Peptide. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2021; 17:e2103244. [PMID: 34480409 DOI: 10.1002/smll.202103244] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 07/22/2021] [Indexed: 06/13/2023]
Abstract
The use of chemoattractants to promote endogenous stem cell-based in situ tissue regeneration has recently garnered much attention. This study is the first to assess the endogenous stem cell migration using a newly discovered substance P (SP) analog (SP1) by molecular dynamics simulations as an efficient chemoattractant. Further, a novel strategy based on electrostatic interaction using cationic chitosan (Ch) and anionic hyaluronic acid (HA) to prepare an SP1-loaded injectable C/H formulation without SP1 loss is developed. The formulation quickly forms an SP1-loaded C/H hydrogel in situ through in vivo injection. The newly discovered SP1 is found to possess human mesenchymal stromal cells (hMSCs) migration-inducing ability that is approximately two to three times higher than that of the existing SP. The designed VEGF-mimicking peptide (VP) chemically reacts with the hydrogel (C/H-VP) to sustain the release of VP, thus inducing vasculogenic differentiation of the hMSCs that migrate toward the C/H-VP hydrogel. Similarly, in animal experiments, SP1 attracts a large number of hMSCs toward the C/H-VP hydrogel, after which VP induces vasculogenic differentiation. Collectively, these findings indicate that SP1-loaded C/H-VP hydrogels are a promising strategy to facilitate endogenous stem cell-based in situ tissue regeneration.
Collapse
Affiliation(s)
- Seung Hun Park
- Department of Molecular Science and Technology, Ajou University, Suwon, 16499, Korea
| | - Hyeon Jin Ju
- Department of Molecular Science and Technology, Ajou University, Suwon, 16499, Korea
| | - Yun Bae Ji
- Department of Molecular Science and Technology, Ajou University, Suwon, 16499, Korea
| | - Masaud Shah
- Department of Molecular Science and Technology, Ajou University, Suwon, 16499, Korea
| | - Byoung Hyun Min
- Department of Molecular Science and Technology, Ajou University, Suwon, 16499, Korea
| | - Hak Soo Choi
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA
| | - Sangdun Choi
- Department of Molecular Science and Technology, Ajou University, Suwon, 16499, Korea
| | - Moon Suk Kim
- Department of Molecular Science and Technology, Ajou University, Suwon, 16499, Korea
- Medipolymers, Research Institute, Woncheon Dong 332-2, Suwon, 16522, Korea
| |
Collapse
|
32
|
Thomas JM, Louca I, Bolan F, Sava O, Allan SM, Lawrence CB, Pinteaux E. Regenerative Potential of Hydrogels for Intracerebral Hemorrhage: Lessons from Ischemic Stroke and Traumatic Brain Injury Research. Adv Healthc Mater 2021; 10:e2100455. [PMID: 34197036 PMCID: PMC11468990 DOI: 10.1002/adhm.202100455] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 06/15/2021] [Indexed: 01/02/2023]
Abstract
Intracerebral hemorrhage (ICH) is a deadly and debilitating type of stroke, caused by the rupture of cerebral blood vessels. To date, there are no restorative interventions approved for use in ICH patients, highlighting a critical unmet need. ICH shares some pathological features with other acute brain injuries such as ischemic stroke (IS) and traumatic brain injury (TBI), including the loss of brain tissue, disruption of the blood-brain barrier, and activation of a potent inflammatory response. New biomaterials such as hydrogels have been recently investigated for their therapeutic benefit in both experimental IS and TBI, owing to their provision of architectural support for damaged brain tissue and ability to deliver cellular and molecular therapies. Conversely, research on the use of hydrogels for ICH therapy is still in its infancy, with very few published reports investigating their therapeutic potential. Here, the published use of hydrogels in experimental ICH is commented upon and how approaches reported in the IS and TBI fields may be applied to ICH research to inform the design of future therapies is described. Unique aspects of ICH that are distinct from IS and TBI that should be considered when translating biomaterial-based therapies between disease models are also highlighted.
Collapse
Affiliation(s)
- Josephine M. Thomas
- Geoffrey Jefferson Brain Research CentreThe Manchester Academic Health Science CentreNorthern Care Alliance NHS GroupThe University of ManchesterManchesterM13 9PTUK
- Division of Neuroscience and Experimental PsychologyFaculty of BiologyMedicine and HealthThe University of ManchesterManchesterM13 9PTUK
| | - Irene Louca
- Geoffrey Jefferson Brain Research CentreThe Manchester Academic Health Science CentreNorthern Care Alliance NHS GroupThe University of ManchesterManchesterM13 9PTUK
- Division of Neuroscience and Experimental PsychologyFaculty of BiologyMedicine and HealthThe University of ManchesterManchesterM13 9PTUK
| | - Faye Bolan
- Geoffrey Jefferson Brain Research CentreThe Manchester Academic Health Science CentreNorthern Care Alliance NHS GroupThe University of ManchesterManchesterM13 9PTUK
- Division of Neuroscience and Experimental PsychologyFaculty of BiologyMedicine and HealthThe University of ManchesterManchesterM13 9PTUK
| | - Oana‐Roxana Sava
- Geoffrey Jefferson Brain Research CentreThe Manchester Academic Health Science CentreNorthern Care Alliance NHS GroupThe University of ManchesterManchesterM13 9PTUK
- Division of Neuroscience and Experimental PsychologyFaculty of BiologyMedicine and HealthThe University of ManchesterManchesterM13 9PTUK
| | - Stuart M. Allan
- Geoffrey Jefferson Brain Research CentreThe Manchester Academic Health Science CentreNorthern Care Alliance NHS GroupThe University of ManchesterManchesterM13 9PTUK
- Division of Neuroscience and Experimental PsychologyFaculty of BiologyMedicine and HealthThe University of ManchesterManchesterM13 9PTUK
| | - Catherine B. Lawrence
- Geoffrey Jefferson Brain Research CentreThe Manchester Academic Health Science CentreNorthern Care Alliance NHS GroupThe University of ManchesterManchesterM13 9PTUK
- Division of Neuroscience and Experimental PsychologyFaculty of BiologyMedicine and HealthThe University of ManchesterManchesterM13 9PTUK
| | - Emmanuel Pinteaux
- Geoffrey Jefferson Brain Research CentreThe Manchester Academic Health Science CentreNorthern Care Alliance NHS GroupThe University of ManchesterManchesterM13 9PTUK
- Division of Neuroscience and Experimental PsychologyFaculty of BiologyMedicine and HealthThe University of ManchesterManchesterM13 9PTUK
| |
Collapse
|
33
|
Shi J, Yu L, Ding J. PEG-based thermosensitive and biodegradable hydrogels. Acta Biomater 2021; 128:42-59. [PMID: 33857694 DOI: 10.1016/j.actbio.2021.04.009] [Citation(s) in RCA: 150] [Impact Index Per Article: 37.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 03/09/2021] [Accepted: 04/01/2021] [Indexed: 02/07/2023]
Abstract
Injectable thermosensitive hydrogels are free-flowing polymer solutions at low or room temperature, making them easy to encapsulate the therapeutic payload or cells via simply mixing. Upon injection into the body, in situ forming hydrogels triggered by body temperature can act as drug-releasing reservoirs or cell-growing scaffolds. Finally, the hydrogels are eliminated from the administration sites after they accomplish their missions as depots or scaffolds. This review outlines the recent progress of poly(ethylene glycol) (PEG)-based biodegradable thermosensitive hydrogels, especially those composed of PEG-polyester copolymers, PEG-polypeptide copolymers and poly(organophosphazene)s. The material design, performance regulation, thermogelation and degradation mechanisms, and corresponding applications in the biomedical field are summarized and discussed. A perspective on the future thermosensitive hydrogels is also highlighted. STATEMENT OF SIGNIFICANCE: Thermosensitive hydrogels undergoing reversible sol-to-gel phase transitions in response to temperature variations are a class of promising biomaterials that can serve as minimally invasive injectable systems for various biomedical applications. Hydrophilic PEG is a main component in the design and fabrication of thermoresponsive hydrogels due to its excellent biocompatibility. By incorporating hydrophobic segments, such as polyesters and polypeptides, into PEG-based systems, biodegradable and thermosensitive hydrogels with adjustable properties in vitro and in vivo have been developed and have recently become a research hotspot of biomaterials. The summary and discussion on molecular design, performance regulation, thermogelation and degradation mechanisms, and biomedical applications of PEG-based thermosensitive hydrogels may offer a demonstration of blueprint for designing new thermogelling systems and expanding their application scope.
Collapse
|
34
|
Ucar B. Natural biomaterials in brain repair: A focus on collagen. Neurochem Int 2021; 146:105033. [PMID: 33785419 DOI: 10.1016/j.neuint.2021.105033] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Revised: 03/07/2021] [Accepted: 03/22/2021] [Indexed: 12/16/2022]
Abstract
Biomaterials derived from natural resources have increasingly been used for versatile applications in the central nervous system (CNS). Thanks to their biocompatibility and biodegradability, natural biomaterials offer vast possibilities for future clinical repair strategies for the CNS. These materials can be used for diverse applications such as hydrogels to fill the tissue cavities, microparticles to deliver drugs across the blood-brain barrier, and scaffolds to transplant stem cells. In this review, various uses of prominent protein and polysaccharide biomaterials, with a special focus on collagen, in repair and regenerative applications for the brain are summarized together with their individual advantages and disadvantages.
Collapse
Affiliation(s)
- Buket Ucar
- Laboratory of Psychiatry and Experimental Alzheimer's Research, Medical University of Innsbruck, Austria.
| |
Collapse
|
35
|
Masson-Meyers DS, Tayebi L. Vascularization strategies in tissue engineering approaches for soft tissue repair. J Tissue Eng Regen Med 2021; 15:747-762. [PMID: 34058083 DOI: 10.1002/term.3225] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 05/08/2021] [Accepted: 05/17/2021] [Indexed: 12/21/2022]
Abstract
Insufficient vascularization during tissue repair is often associated with poor clinical outcomes. This is a concern especially when patients have critical-sized injuries, where the size of the defect restricts vascularity, or even in small defects that have to be treated under special conditions, such as after radiation therapy (relevant to tumor resection) that hinders vascularity. In fact, poor vascularization is one of the major obstacles for clinical application of tissue engineering methods in soft tissue repair. As a key issue, lack of graft integration, caused by inadequate vascularization after implantation, can lead to graft failure. Moreover, poor vascularization compromises the viability of cells seeded in deep portions of scaffolds/graft materials, due to hypoxia and insufficient nutrient supply. In this article we aim to review vascularization strategies employed in tissue engineering techniques to repair soft tissues. For this purpose, we start by providing a brief overview of the main events during the physiological wound healing process in soft tissues. Then, we discuss how tissue repair can be achieved through tissue engineering, and considerations with regards to the choice of scaffold materials, culture conditions, and vascularization techniques. Next, we highlight the importance of vascularization, along with strategies and methods of prevascularization of soft tissue equivalents, particularly cell-based prevascularization. Lastly, we present a summary of commonly used in vitro methods during the vascularization of tissue-engineered soft tissue constructs.
Collapse
Affiliation(s)
| | - Lobat Tayebi
- Marquette University School of Dentistry, Milwaukee, WI, USA
| |
Collapse
|
36
|
Biotherapeutic-loaded injectable hydrogels as a synergistic strategy to support myocardial repair after myocardial infarction. J Control Release 2021; 335:216-236. [PMID: 34022323 DOI: 10.1016/j.jconrel.2021.05.023] [Citation(s) in RCA: 71] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 05/16/2021] [Accepted: 05/18/2021] [Indexed: 12/18/2022]
Abstract
Myocardial infarction (MI) has been considered as the leading cause of cardiovascular-related deaths worldwide. Although traditional therapeutic agents including various bioactive species such as growth factors, stem cells, and nucleic acids have demonstrated somewhat usefulness for the restoration of cardiac functions, the therapeutic efficiency remains unsatisfactory most likely due to the off-target-associated side effects and low localized retention of the used therapeutic agents in the infarcted myocardium, which constitutes a substantial barrier for the effective treatment of MI. Injectable hydrogels are regarded as a minimally invasive technology that can overcome the clinical and surgical limitations of traditional stenting by a modulated sol-gel transition and localized transport of a variety of encapsulated cargoes, leading to enhanced therapeutic efficiency and improved patient comfort and compliance. However, the design of injectable hydrogels for myocardial repair and the mechanism of action of bioactive substance-loaded hydrogels for MI repair remain unclear. To elucidate these points, we summarized the recent progresses made on the use of injectable hydrogels for encapsulation of various therapeutic substances for MI treatment with an emphasis on the mechanism of action of hydrogel systems for myocardial repair. Specifically, the pathogenesis of MI and the rational design of injectable hydrogels for myocardial repair were presented. Next, the mechanisms of various biotherapeutic substance-loaded injectable hydrogels for myocardial repair was discussed. Finally, the potential challenges and future prospects for the use of injectable hydrogels for MI treatment were proposed for the purpose of drawing theoretical guidance on the development of novel therapeutic strategies for efficient treatment of MI.
Collapse
|
37
|
Carvalho IC, Mansur HS, Leonel AG, Mansur AAP, Lobato ZIP. Soft matter polysaccharide-based hydrogels as versatile bioengineered platforms for brain tissue repair and regeneration. Int J Biol Macromol 2021; 182:1091-1111. [PMID: 33892028 DOI: 10.1016/j.ijbiomac.2021.04.116] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 04/12/2021] [Accepted: 04/17/2021] [Indexed: 01/08/2023]
Abstract
Acute or chronic brain injuries promote deaths and the life-long debilitating neurological status where, despite advances in therapeutic strategies, clinical outcome hardly achieves total patient recovery. In recent decades, brain tissue engineering emerged as an encouraging area of research for helping in damaged central nervous system (CNS) recovery. Polysaccharides are abundant naturally occurring biomacromolecules with a great potential enhancement of advanced technologies in brain tissue repair and regeneration (BTRR). Besides carrying rich biological information, polysaccharides can interact and communicate with biomolecules, including glycosaminoglycans present in cell membranes and many signaling moieties, growth factors, chemokines, and axon guidance molecules. This review includes a comprehensive investigation of the current progress on designing and developing polysaccharide-based soft matter biomaterials for BTRR. Although few interesting reviews concerning BTRR have been reported, this is the first report specifically focusing on covering multiple polysaccharides and polysaccharide-based functionalized biomacromolecules in this emerging and intriguing field of multidisciplinary knowledge. This review aims to cover the state of art challenges and prospects of this fascinating field while presenting the richness of possibilities of using these natural biomacromolecules for advanced biomaterials in prospective neural tissue engineering applications.
Collapse
Affiliation(s)
- Isadora C Carvalho
- Center of Nanoscience, Nanotechnology and Innovation - CeNano(2)I, Department of Metallurgical and Materials Engineering, Federal University of Minas Gerais - UFMG, Av. Antônio Carlos, 6627 Belo Horizonte/M.G., Brazil
| | - Herman S Mansur
- Center of Nanoscience, Nanotechnology and Innovation - CeNano(2)I, Department of Metallurgical and Materials Engineering, Federal University of Minas Gerais - UFMG, Av. Antônio Carlos, 6627 Belo Horizonte/M.G., Brazil.
| | - Alice G Leonel
- Center of Nanoscience, Nanotechnology and Innovation - CeNano(2)I, Department of Metallurgical and Materials Engineering, Federal University of Minas Gerais - UFMG, Av. Antônio Carlos, 6627 Belo Horizonte/M.G., Brazil
| | - Alexandra A P Mansur
- Center of Nanoscience, Nanotechnology and Innovation - CeNano(2)I, Department of Metallurgical and Materials Engineering, Federal University of Minas Gerais - UFMG, Av. Antônio Carlos, 6627 Belo Horizonte/M.G., Brazil
| | - Zelia I P Lobato
- Department of Preventive Veterinary Medicine, Veterinary School, Federal University of Minas Gerais - UFMG, Brazil
| |
Collapse
|
38
|
Chen M, Liu Q, Xu Y, Wang Y, Han X, Wang Z, Liang J, Sun Y, Fan Y, Zhang X. The effect of LyPRP/collagen composite hydrogel on osteogenic differentiation of rBMSCs. Regen Biomater 2020; 8:rbaa053. [PMID: 33732498 PMCID: PMC7947583 DOI: 10.1093/rb/rbaa053] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 10/22/2020] [Accepted: 11/15/2020] [Indexed: 12/12/2022] Open
Abstract
Although platelet-rich plasma (PRP) plays a significant role in the orthopedic clinical application, it still faces two major problems, namely, uncontrollable factors release, frequent preparation and extraction processes as well as the inconvenient form of usage. To overcome these shortcomings, freeze-dried PRP (LyPRP) was encapsulated into bioactive Col I hydrogel to induce osteogenic differentiation of rabbit bone marrow mesenchymal stem cells (rBMSCs). And PRP/Col І composite hydrogel was prepared as a control. Compared with Col І hydrogel, the introduction of platelets significantly improved the mechanical properties of hydrogels. Meanwhile, platelets were evenly distributed in the composite hydrogels network. The sustainable release of related factors in the composite hydrogels could last for more than 14 days to maintain its long-term biological activity. Further cell experiments confirmed that PRP and LyPRP could effectively alleviate the contraction of collagen hydrogel in vitro, and promote the adhesion, proliferation and osteogenesis differentiation of rBMSCs. The results of osteogenic gene expression indicated that the 10% LyPRP/Col І composite hydrogel could facilitate the early expression of BMP-2 and late osteogenic associated protein formation with higher expression of alkaline phosphatase and Osteocalcin (OCN). These results might provide new insights for the clinical application of 10% LyPRP/Col І composite hydrogel as practical bone repair injection.
Collapse
Affiliation(s)
- Manyu Chen
- National Engineering Research Center for Biomaterials, Sichuan University, 29 Wangjiang Road, Chengdu, Sichuan 610064, P. R. China
| | - Quanying Liu
- National Engineering Research Center for Biomaterials, Sichuan University, 29 Wangjiang Road, Chengdu, Sichuan 610064, P. R. China
| | - Yang Xu
- National Engineering Research Center for Biomaterials, Sichuan University, 29 Wangjiang Road, Chengdu, Sichuan 610064, P. R. China
| | - Yuxiang Wang
- National Engineering Research Center for Biomaterials, Sichuan University, 29 Wangjiang Road, Chengdu, Sichuan 610064, P. R. China
| | - Xiaowen Han
- National Engineering Research Center for Biomaterials, Sichuan University, 29 Wangjiang Road, Chengdu, Sichuan 610064, P. R. China
| | - Zhe Wang
- Department of Medical Genetics, Zunyi Medical University, No. 6 West Xuefu Road, Zunyi, Guizhou 563000, P. R. China
| | - Jie Liang
- National Engineering Research Center for Biomaterials, Sichuan University, 29 Wangjiang Road, Chengdu, Sichuan 610064, P. R. China.,Sichuan Testing Center for Biomaterials and Medical Devices, Sichuan University, 29 Wangjiang Road, Chengdu 610064, P. R. China
| | - Yong Sun
- National Engineering Research Center for Biomaterials, Sichuan University, 29 Wangjiang Road, Chengdu, Sichuan 610064, P. R. China
| | - Yujiang Fan
- National Engineering Research Center for Biomaterials, Sichuan University, 29 Wangjiang Road, Chengdu, Sichuan 610064, P. R. China
| | - Xingdong Zhang
- National Engineering Research Center for Biomaterials, Sichuan University, 29 Wangjiang Road, Chengdu, Sichuan 610064, P. R. China
| |
Collapse
|
39
|
Narayanan N, Jia Z, Kim KH, Kuang L, Lengemann P, Shafer G, Bernal-Crespo V, Kuang S, Deng M. Biomimetic glycosaminoglycan-based scaffolds improve skeletal muscle regeneration in a Murine volumetric muscle loss model. Bioact Mater 2020; 6:1201-1213. [PMID: 33163701 PMCID: PMC7599371 DOI: 10.1016/j.bioactmat.2020.10.012] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 10/17/2020] [Accepted: 10/18/2020] [Indexed: 12/20/2022] Open
Abstract
Volumetric muscle loss (VML) injuries characterized by critical loss of skeletal muscle tissues result in severe functional impairment. Current treatments involving use of muscle grafts are limited by tissue availability and donor site morbidity. In this study, we designed and synthesized an implantable glycosaminoglycan-based hydrogel system consisting of thiolated hyaluronic acid (HA) and thiolated chondroitin sulfate (CS) cross-linked with poly(ethylene glycol) diacrylate to promote skeletal muscle regeneration of VML injuries in mice. The HA-CS hydrogels were optimized with suitable biophysical properties by fine-tuning degree of thiol group substitution to support C2C12 myoblast proliferation, myogenic differentiation and expression of myogenic markers MyoD, MyoG and MYH8. Furthermore, in vivo studies using a murine quadriceps VML model demonstrated that the HA-CS hydrogels supported integration of implants with the surrounding host tissue and facilitated migration of Pax7+ satellite cells, de novo myofiber formation, angiogenesis, and innervation with minimized scar tissue formation during 4-week implantation. The hydrogel-treated and autograft-treated mice showed similar functional improvements in treadmill performance as early as 1-week post-implantation compared to the untreated groups. Taken together, our results demonstrate the promise of HA-CS hydrogels as regenerative engineering matrices to accelerate healing of skeletal muscle injuries.
Collapse
Key Words
- AChR, Acetyl choline receptors
- CS, Chondroitin Sulfate
- Chondroitin sulfate
- ECM, Extracellular matrix
- EDC, 1-ethyl-3-(3-dimethylaminopropyl) carbodiimide
- GAG, Glycosaminoglycan
- HA, Hyaluronic acid
- Hyaluronic acid
- Hydrogels
- MES, 2-(N-morpholino) ethanesulfonic acid
- MHC, Myosin heavy chain
- Myoblasts
- NHS, N-hydroxysuccinimide
- PEGDA, Poly(ethylene glycol) diacrylate
- Skeletal muscle tissue engineering
- VML, Volumetric muscle loss
- Volumetric muscle loss
- eMHC, embryonic myosin heavy chain
Collapse
Affiliation(s)
- Naagarajan Narayanan
- Department of Agricultural and Biological Engineering, Purdue University, West Lafayette, IN, 47906, United States.,Bindley Bioscience Center, Purdue University, West Lafayette, IN, 47906, United States
| | - Zhihao Jia
- Department of Animal Science, Purdue University, West Lafayette, IN, 47906, United States
| | - Kun Ho Kim
- Department of Animal Science, Purdue University, West Lafayette, IN, 47906, United States
| | - Liangju Kuang
- Department of Agricultural and Biological Engineering, Purdue University, West Lafayette, IN, 47906, United States.,Bindley Bioscience Center, Purdue University, West Lafayette, IN, 47906, United States
| | - Paul Lengemann
- Department of Agricultural and Biological Engineering, Purdue University, West Lafayette, IN, 47906, United States.,Bindley Bioscience Center, Purdue University, West Lafayette, IN, 47906, United States
| | - Gabrielle Shafer
- Center for Comparative Translational Research, Purdue University, West Lafayette, IN, 47906, United States
| | - Victor Bernal-Crespo
- Center for Comparative Translational Research, Purdue University, West Lafayette, IN, 47906, United States
| | - Shihuan Kuang
- Department of Animal Science, Purdue University, West Lafayette, IN, 47906, United States
| | - Meng Deng
- Department of Agricultural and Biological Engineering, Purdue University, West Lafayette, IN, 47906, United States.,Bindley Bioscience Center, Purdue University, West Lafayette, IN, 47906, United States.,School of Materials Engineering, Purdue University, West Lafayette, IN, 47906, United States.,Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, 47906, United States
| |
Collapse
|
40
|
Jensen G, Holloway JL, Stabenfeldt SE. Hyaluronic Acid Biomaterials for Central Nervous System Regenerative Medicine. Cells 2020; 9:E2113. [PMID: 32957463 PMCID: PMC7565873 DOI: 10.3390/cells9092113] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 09/09/2020] [Accepted: 09/11/2020] [Indexed: 12/16/2022] Open
Abstract
Hyaluronic acid (HA) is a primary component of the brain extracellular matrix and functions through cellular receptors to regulate cell behavior within the central nervous system (CNS). These behaviors, such as migration, proliferation, differentiation, and inflammation contribute to maintenance and homeostasis of the CNS. However, such equilibrium is disrupted following injury or disease leading to significantly altered extracellular matrix milieu and cell functions. This imbalance thereby inhibits inherent homeostatic processes that support critical tissue health and functionality in the CNS. To mitigate the damage sustained by injury/disease, HA-based tissue engineering constructs have been investigated for CNS regenerative medicine applications. HA's effectiveness in tissue healing and regeneration is primarily attributed to its impact on cell signaling and the ease of customizing chemical and mechanical properties. This review focuses on recent findings to highlight the applications of HA-based materials in CNS regenerative medicine.
Collapse
Affiliation(s)
- Gregory Jensen
- Chemical Engineering, School for Engineering of Matter, Transport and Energy, Arizona State University, Tempe, AZ 85224, USA;
| | - Julianne L. Holloway
- Chemical Engineering, School for Engineering of Matter, Transport and Energy, Arizona State University, Tempe, AZ 85224, USA;
| | - Sarah E. Stabenfeldt
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ 85287, USA
| |
Collapse
|
41
|
Xing F, Xiang Z, Rommens PM, Ritz U. 3D Bioprinting for Vascularized Tissue-Engineered Bone Fabrication. MATERIALS (BASEL, SWITZERLAND) 2020; 13:E2278. [PMID: 32429135 PMCID: PMC7287611 DOI: 10.3390/ma13102278] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 03/26/2020] [Accepted: 04/08/2020] [Indexed: 02/05/2023]
Abstract
Vascularization in bone tissues is essential for the distribution of nutrients and oxygen, as well as the removal of waste products. Fabrication of tissue-engineered bone constructs with functional vascular networks has great potential for biomimicking nature bone tissue in vitro and enhancing bone regeneration in vivo. Over the past decades, many approaches have been applied to fabricate biomimetic vascularized tissue-engineered bone constructs. However, traditional tissue-engineered methods based on seeding cells into scaffolds are unable to control the spatial architecture and the encapsulated cell distribution precisely, which posed a significant challenge in constructing complex vascularized bone tissues with precise biomimetic properties. In recent years, as a pioneering technology, three-dimensional (3D) bioprinting technology has been applied to fabricate multiscale, biomimetic, multi-cellular tissues with a highly complex tissue microenvironment through layer-by-layer printing. This review discussed the application of 3D bioprinting technology in the vascularized tissue-engineered bone fabrication, where the current status and unique challenges were critically reviewed. Furthermore, the mechanisms of vascular formation, the process of 3D bioprinting, and the current development of bioink properties were also discussed.
Collapse
Affiliation(s)
- Fei Xing
- Department of Orthopaedics and Traumatology, Biomatics Group, University Medical Center of the Johannes Gutenberg University, Mainz 55131, Germany; (F.X.); (P.M.R.)
- Department of Orthopaedics, West China Hospital, Sichuan University, No. 37 Guoxue Lane, Chengdu 610041, China;
- Trauma Medical Center of West China Hospital, Sichuan University, No. 37 Guoxue Lane, Chengdu 610041, China
| | - Zhou Xiang
- Department of Orthopaedics, West China Hospital, Sichuan University, No. 37 Guoxue Lane, Chengdu 610041, China;
- Trauma Medical Center of West China Hospital, Sichuan University, No. 37 Guoxue Lane, Chengdu 610041, China
| | - Pol Maria Rommens
- Department of Orthopaedics and Traumatology, Biomatics Group, University Medical Center of the Johannes Gutenberg University, Mainz 55131, Germany; (F.X.); (P.M.R.)
| | - Ulrike Ritz
- Department of Orthopaedics and Traumatology, Biomatics Group, University Medical Center of the Johannes Gutenberg University, Mainz 55131, Germany; (F.X.); (P.M.R.)
| |
Collapse
|
42
|
Swetha S, Lavanya K, Sruthi R, Selvamurugan N. An insight into cell-laden 3D-printed constructs for bone tissue engineering. J Mater Chem B 2020; 8:9836-9862. [DOI: 10.1039/d0tb02019b] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
In this review, we have spotlighted various combinations of bioinks to optimize the biofabrication of 3D bone constructs.
Collapse
Affiliation(s)
- S. Swetha
- Department of Biotechnology, College of Engineering and Technology
- SRM Institute of Science and Technology
- Kattankulathur 603 203
- India
| | - K. Lavanya
- Department of Biotechnology, College of Engineering and Technology
- SRM Institute of Science and Technology
- Kattankulathur 603 203
- India
| | - R. Sruthi
- Department of Biotechnology, College of Engineering and Technology
- SRM Institute of Science and Technology
- Kattankulathur 603 203
- India
| | - N. Selvamurugan
- Department of Biotechnology, College of Engineering and Technology
- SRM Institute of Science and Technology
- Kattankulathur 603 203
- India
| |
Collapse
|