1
|
Mohanty S, Roy S. Bioactive Hydrogels Inspired by Laminin: An Emerging Biomaterial for Tissue Engineering Applications. Macromol Biosci 2024; 24:e2400207. [PMID: 39172212 DOI: 10.1002/mabi.202400207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 08/01/2024] [Indexed: 08/23/2024]
Abstract
Tissue or organ damage due to severe injuries or chronic diseases can adversely affect the quality of life. Current treatments rely on organ or tissue transplantation which has limitations including unavailability of donors, ethical issues, or immune rejection after transplantations. These limitations can be addressed by tissue regeneration which involves the development of bioactive scaffolds closely mimicking the extracellular matrix (ECM). One of the major components of ECM is the laminin protein which supports several tissues associated with important organs. In this direction, peptide-based hydrogels can effectively mimic the essential characteristics of laminin. While several reports have discussed the structure of laminin, the potential of laminin-derived peptide hydrogels as effective biomaterial for tissue engineering applications is yet to be discussed. In this context, the current review focuses on the structure of laminin and its role as an essential ECM protein. Further, the potential of short peptide hydrogels in mimicking the crucial properties of laminin is proposed. The review further highlights the significance of bioactive hydrogels inspired by laminin - in addressing numerous tissue engineering applications including angiogenesis, neural, skeletal muscle, liver, and adipose tissue regeneration along with a brief outlook on the future applications of these laminin-based hydrogels.
Collapse
Affiliation(s)
- Sweta Mohanty
- Institute of Nano Science and Technology (INST), Sector 81, Knowledge City, Mohali, Punjab, 140306, India
| | - Sangita Roy
- Institute of Nano Science and Technology (INST), Sector 81, Knowledge City, Mohali, Punjab, 140306, India
| |
Collapse
|
2
|
Ren H, Wu W, Chen J, Li Q, Wang H, Qian D, Guo S, Duan JA. Integrated serum metabolomics and network pharmacology analysis on the bioactive metabolites and mechanism exploration of Bufei huoxue capsule on chronic obstructive pulmonary disease rats. JOURNAL OF ETHNOPHARMACOLOGY 2024; 324:117816. [PMID: 38286154 DOI: 10.1016/j.jep.2024.117816] [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: 11/17/2023] [Revised: 01/09/2024] [Accepted: 01/21/2024] [Indexed: 01/31/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Bufei Huoxue capsule (BHC) as a classic Chinese patent medicine formula, has the efficacy of tonifying the lungs and activating the blood. It has been extensively used in China for the treatment of chronic obstructive pulmonary disease (COPD) clinically. However, its mechanism is still unclear, which hampers the applications of BHC in treating COPD. AIM OF THE STUDY The purpose of the present study was to demonstrate the protective efficacy and mechanism of BHC on COPD model rats by integrating serum metabolomics analysis and network pharmacology study. MATERIALS AND METHODS A COPD rat model was established by cigarette fumigation combined with lipopolysaccharide (LPS) airway drip for 90 consecutive days. After oral administration for 30 days, the rats were placed in the body tracing box of the EMKA Small Animal Noninvasive Lung Function Test System to determine lung function related indexes. Histopathological alteration was observed by H&E staining and Masson staining. The serum levels of inflammatory cytokine, matrix metalloprotein 9, and laminin were determined by ELISA kits. Oxidative stress levels were tested by biochemical methods. UHPLC-Q-TOF/MS analysis of serum metabolomics and network pharmacology were performed to reveal the bioactive metabolites, key components and pathways for BHC treating COPD. WB and ELISA kits were used to verify the effects of BHC on key pathway. RESULTS BHC could improve lung function, immunity, lung histopathological changes and collagen deposition in COPD model rats. It also could significantly reduce inflammatory response in vivo, regulate oxidative stress level, reduce laminin content, and regulate protease-antiprotease balance. Metabolomics analysis found 46 biomarkers of COPD, of which BHC significantly improved the levels of 23 differential metabolites including arachidonic acid, leukotriene B4 and prostaglandin E2. Combined with the results of network pharmacology, the components of BHC, such as calycosin, oxypaeoniflora, (S)-bavachin and neobavaisoflavone could play therapeutic roles through the arachidonic acid pathway. In addition, the results of WB and ELISA indicated that BHC could suppress the expressions of COX2 and 5-LOX in lung tissues and inhibit the generation of AA and its metabolites in serum samples. Regulation of arachidonic acid metabolic pathway may be the crucial mechanism for BHC treating COPD. CONCLUSIONS In summary, the studies indicated that BHC exhibited the protective effect on COPD model rats by anti-inflammatory and anti-oxidative properties through arachidonic acid metabolism pathway. This study provided beneficial support for the applications of BHC in treating COPD.
Collapse
Affiliation(s)
- Hui Ren
- National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization and Jiangsu Key Laboratory for High Technology Research of Traditional Chinese Medicine Formulae, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Wenxing Wu
- National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization and Jiangsu Key Laboratory for High Technology Research of Traditional Chinese Medicine Formulae, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Jiangyan Chen
- National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization and Jiangsu Key Laboratory for High Technology Research of Traditional Chinese Medicine Formulae, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Quan Li
- Leiyunshang Pharmaceutical Co. Limited, Suzhou, 215003, China
| | - Hengbin Wang
- Leiyunshang Pharmaceutical Co. Limited, Suzhou, 215003, China
| | - Dawei Qian
- National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization and Jiangsu Key Laboratory for High Technology Research of Traditional Chinese Medicine Formulae, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Sheng Guo
- National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization and Jiangsu Key Laboratory for High Technology Research of Traditional Chinese Medicine Formulae, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| | - Jin-Ao Duan
- National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization and Jiangsu Key Laboratory for High Technology Research of Traditional Chinese Medicine Formulae, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| |
Collapse
|
3
|
Mohammed Butt A, Rupareliya V, Hariharan A, Kumar H. Building a pathway to recovery: Targeting ECM remodeling in CNS injuries. Brain Res 2023; 1819:148533. [PMID: 37586675 DOI: 10.1016/j.brainres.2023.148533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 08/07/2023] [Accepted: 08/09/2023] [Indexed: 08/18/2023]
Abstract
Extracellular matrix (ECM) is a complex and dynamic network of proteoglycans, proteins, and other macromolecules that surrounds cells in tissues. The ECM provides structural support to cells and plays a critical role in regulating various cellular functions. ECM remodeling is a dynamic process involving the breakdown and reconstruction of the ECM. This process occurs naturally during tissue growth, wound healing, and tissue repair. However, in the context of central nervous system (CNS) injuries, dysregulated ECM remodeling can lead to the formation of fibrotic and glial scars. CNS injuries encompass various traumatic events, including concussions and fractures. Following CNS trauma, the formation of glial and fibrotic scars becomes prominent. Glial scars primarily consist of reactive astrocytes, while fibrotic scars are characterized by an abundance of ECM proteins. ECM remodeling plays a pivotal and tightly regulated role in the development of these scars after spinal cord and brain injuries. Various factors like ECM components, ECM remodeling enzymes, cell surface receptors of ECM molecules, and downstream pathways of ECM molecules are responsible for the remodeling of the ECM. The aim of this review article is to explore the changes in ECM during normal physiological conditions and following CNS injuries. Additionally, we discuss various approaches that target various factors responsible for ECM remodeling, with a focus on promoting axon regeneration and functional recovery after CNS injuries. By targeting ECM remodeling, it may be possible to enhance axonal regeneration and facilitate functional recovery after CNS injuries.
Collapse
Affiliation(s)
- Ayub Mohammed Butt
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, Gandhinagar, Gujarat, India
| | - Vimal Rupareliya
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, Gandhinagar, Gujarat, India
| | - A Hariharan
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, Gandhinagar, Gujarat, India
| | - Hemant Kumar
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, Gandhinagar, Gujarat, India.
| |
Collapse
|
4
|
Bakhshandeh B, Ranjbar N, Abbasi A, Amiri E, Abedi A, Mehrabi M, Dehghani Z, Pennisi CP. Recent progress in the manipulation of biochemical and biophysical cues for engineering functional tissues. Bioeng Transl Med 2023; 8:e10383. [PMID: 36925674 PMCID: PMC10013802 DOI: 10.1002/btm2.10383] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 06/28/2022] [Accepted: 07/16/2022] [Indexed: 11/11/2022] Open
Abstract
Tissue engineering (TE) is currently considered a cutting-edge discipline that offers the potential for developing treatments for health conditions that negatively affect the quality of life. This interdisciplinary field typically involves the combination of cells, scaffolds, and appropriate induction factors for the regeneration and repair of damaged tissue. Cell fate decisions, such as survival, proliferation, or differentiation, critically depend on various biochemical and biophysical factors provided by the extracellular environment during developmental, physiological, and pathological processes. Therefore, understanding the mechanisms of action of these factors is critical to accurately mimic the complex architecture of the extracellular environment of living tissues and improve the efficiency of TE approaches. In this review, we recapitulate the effects that biochemical and biophysical induction factors have on various aspects of cell fate. While the role of biochemical factors, such as growth factors, small molecules, extracellular matrix (ECM) components, and cytokines, has been extensively studied in the context of TE applications, it is only recently that we have begun to understand the effects of biophysical signals such as surface topography, mechanical, and electrical signals. These biophysical cues could provide a more robust set of stimuli to manipulate cell signaling pathways during the formation of the engineered tissue. Furthermore, the simultaneous application of different types of signals appears to elicit synergistic responses that are likely to improve functional outcomes, which could help translate results into successful clinical therapies in the future.
Collapse
Affiliation(s)
- Behnaz Bakhshandeh
- Department of Biotechnology, College of ScienceUniversity of TehranTehranIran
| | - Nika Ranjbar
- Department of Biotechnology, College of ScienceUniversity of TehranTehranIran
| | - Ardeshir Abbasi
- Department of Immunology, Faculty of Medical SciencesTarbiat Modares UniversityTehranIran
| | - Elahe Amiri
- Department of Life Science Engineering, Faculty of New Sciences and TechnologyUniversity of TehranTehranIran
| | - Ali Abedi
- Department of Life Science Engineering, Faculty of New Sciences and TechnologyUniversity of TehranTehranIran
| | - Mohammad‐Reza Mehrabi
- Department of Microbial Biotechnology, School of Biology, College of ScienceUniversity of TehranTehranIran
| | - Zahra Dehghani
- Department of Biotechnology, College of ScienceUniversity of TehranTehranIran
| | - Cristian Pablo Pennisi
- Regenerative Medicine Group, Department of Health Science and TechnologyAalborg UniversityAalborgDenmark
| |
Collapse
|
5
|
Zhang Q, Liu J, Chen L, Zhang M. Promoting Endogenous Neurogenesis as a Treatment for Alzheimer's Disease. Mol Neurobiol 2023; 60:1353-1368. [PMID: 36445633 DOI: 10.1007/s12035-022-03145-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Accepted: 11/19/2022] [Indexed: 11/30/2022]
Abstract
Alzheimer's disease (AD) is the most universal neurodegenerative disorder characterized by memory loss and cognitive impairment. AD is biologically defined by production and aggregation of misfolded protein including extracellular amyloid β (Aβ) peptide and intracellular microtubule-associated protein tau tangles in neurons, leading to irreversible neuronal loss. At present, regulation of endogenous neurogenesis to supplement lost neurons has been proposed as a promising strategy for treatment of AD. However, the exact underlying mechanisms of impaired neurogenesis in AD have not been fully explained and effective treatments targeting neurogenesis for AD are limited. In this review, we mainly focus on the latest research of impaired neurogenesis in AD. Then we discuss the factors affecting stages of neurogenesis and the interplay between neural stem cells (NSCs) and neurogenic niche under AD pathological conditions. This review aims to explore potential therapeutic strategies that promote endogenous neurogenesis for AD treatments.
Collapse
Affiliation(s)
- Qiang Zhang
- Department of Pharmacology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin Province, China
| | - Jingyue Liu
- Department of Pharmacology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin Province, China
| | - Li Chen
- Department of Pharmacology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin Province, China. .,School of Nursing, Jilin University, Changchun, China.
| | - Ming Zhang
- Department of Pharmacology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin Province, China.
| |
Collapse
|
6
|
Ghandy N, Ebrahimzadeh-Bideskan A, Gorji A, Negah SS. Co-transplantation of novel Nano-SDF scaffold with human neural stem cells attenuates inflammatory responses and apoptosis in traumatic brain injury. Int Immunopharmacol 2023; 115:109709. [PMID: 36638659 DOI: 10.1016/j.intimp.2023.109709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 01/02/2023] [Accepted: 01/05/2023] [Indexed: 01/12/2023]
Abstract
Traumatic brain injury (TBI) causes long-term disability and mortality worldwide. The prime pathological players in TBI are neuroinflammation and apoptosis. These pathological changes lead to a limited capacity of regeneration after TBI. To alleviate inflammatory responses and apoptosis triggered by TBI, developing bioactive scaffolds conjoined with stem cells is a decisive approach in neural tissue engineering. The aim of this study was to fabricate a novel nano-scaffold made of RADA-16 with a bioactive motif of stromal cell-derived factor-1 α (SDF-1α) and evaluate its effects with stem cell transplantation on inflammatory pathways, reactive gliosis, and apoptosis after TBI. Co-transplantation of Nano-SDF and human neural stem cells (hNSCs) derived from fetus brain in adult rats subjected to TBI led to the improvement of motor activitycompared with the control group. The treated animals with hNSCs + Nano-SDF had a significantly lower expression of toll-like receptor 4 and nuclear factor-kappa B at the injury site than the control animals. A significant reduction in the number of reactive astrocytes was also observed in rats that received hNSCs + Nano-SDF compared with the vehicle and Nano-SDF groups. Furthermore, the TUNEL assay indicated a significant reduction in TUNEL positive cells in the hNSCs + Nano-SDF group compared with the TBI, vehicle, and Nano-SDF groups. These data demonstrated co-transplantation of hNSCs with Nano-SDF can reduce inflammatory responses and cell death after TBI via creating a more supportive microenvironment. Further research is required to establish the therapeutic efficacy of Nano-SDF with stem cells for TBI.
Collapse
Affiliation(s)
- Nasibeh Ghandy
- Department of Anatomy and Cell Biology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Alireza Ebrahimzadeh-Bideskan
- Department of Anatomy and Cell Biology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Ali Gorji
- Neuroscience Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Shefa Neuroscience Research Center, Khatam Alanbia Hospital, Tehran, Iran; Department of Neuroscience, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Epilepsy Research Center, Westfälische Wilhelms-Universität Münster, Münster, Germany.
| | - Sajad Sahab Negah
- Neuroscience Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Shefa Neuroscience Research Center, Khatam Alanbia Hospital, Tehran, Iran; Department of Neuroscience, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
7
|
Hajinejad M, Ebrahimzadeh MH, Ebrahimzadeh-Bideskan A, Rajabian A, Gorji A, Sahab Negah S. Exosomes and Nano-SDF Scaffold as a Cell-Free-Based Treatment Strategy Improve Traumatic Brain Injury Mechanisms by Decreasing Oxidative Stress, Neuroinflammation, and Increasing Neurogenesis. Stem Cell Rev Rep 2023; 19:1001-1018. [PMID: 36652144 DOI: 10.1007/s12015-022-10483-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/13/2022] [Indexed: 01/19/2023]
Abstract
Traumatic brain injury (TBI) causes a variety of complex pathological changes in brain parenchymal tissue by increasing neuroinflammatory and apoptosis responses. Currently, there is no treatment to resolve the consequences related to TBI. Recently, an extensive literature has grown up around the theme of bystander effects of stem cells, a mechanism of stem cells without the need for cell transplantation, which is called cell-free therapy. The purpose of this investigation was to determine the efficacy of a cell-free-based therapy strategy using exosomes derived from human neural stem cells (hNSCs) and a novel nano-scaffold in rats subjected to TBI. In this study, a series of in vitro and in vivo experiments from behavior tests to gene expression was performed to define the effect of exosomes in combination with a three-dimensional (3D) nano-scaffold containing a bio-motif of SDF1α (Nano-SDF). Application of exosomes with Nano-SDF significantly decreased oxidative stress in serum and brain samples. Moreover, treatment with exosomes and Nano-SDF significantly reduced the expression of Toll-like receptor 4 and its downstream signaling pathway, including NF-kβ and interleukin-1β. We also found that the cell-free-based therapy strategy could decrease reactive gliosis at the injury site. Interestingly, we showed that exosomes with Nano-SDF increased neurogenesis in the sub-ventricular zone of the lateral ventricle, indicating a bio-bridge mechanism. To sum up, the most obvious finding to emerge from this study is that a cell-free-based therapy strategy can be an effective option for future practice in the course of TBI.
Collapse
Affiliation(s)
- Mehrdad Hajinejad
- Department of Anatomy and Cell Biology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.,Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Alireza Ebrahimzadeh-Bideskan
- Department of Anatomy and Cell Biology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran. .,Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Arezoo Rajabian
- Department of Internal Medicine, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ali Gorji
- Shefa Neuroscience Research Center, Khatam Alanbia Hospital, Tehran, Iran.,Epilepsy Research Center, Westfälische Wilhelms-Universität Münster, 48149, Munster, Germany
| | - Sajad Sahab Negah
- Neuroscience Research Center, Mashhad University of Medical Sciences, Mashhad, Iran. .,Department of Neuroscience, Faculty of Medicine, Mashhad University of Medical Sciences, Pardis Campus, Azadi Square, Kalantari Blvd, Mashhad, Iran.
| |
Collapse
|
8
|
Ran L, Peng SY, Wang W, Wu Q, Li YC, Wang RP. In vitro and in vivo Evaluation of the Bioactive Nanofibers-Encapsulated Benzalkonium Bromide for Accelerating Wound Repair with MRSA Skin Infection. Int J Nanomedicine 2022; 17:4419-4432. [PMID: 36172005 PMCID: PMC9510697 DOI: 10.2147/ijn.s380786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 09/07/2022] [Indexed: 11/23/2022] Open
Abstract
Purpose Developing the ideal drug or dressing is a serious challenge to controlling the occurrence of antibacterial infection during wound healing. Thus, it is important to prepare novel nanofibers for a wound dressing that can control bacterial infections. In our study, the novel self-assembled nanofibers of benzalkonium bromide with bioactive peptide materials of IKVAV and RGD were designed and fabricated. Methods Different drug concentration effects of encapsulation efficacy, swelling ratio and strength were determined. Its release profile in simulated wound fluid and its cytotoxicity were studied in vitro. Importantly, the antibacterial efficacy, inhibition of biofilm formation effect and wound healing against MRSA infections in vitro and in vivo were performed after observing the tissue toxicity in vivo. Results It was found that the optimized drug load (0.8%) was affected by the encapsulation efficacy, swelling ratio, and strength. In addition, the novel nanofibers with average diameter (222.0 nm) and stabile zeta potential (−11.2 mV) have good morphology and characteristics. It has a delayed released profile in the simulated wound fluid and good biocompatibility with L929 cells and most tissues. Importantly, the nanofibers were shown to improve antibacterial efficacy, inhibit biofilm formation, and lead to accelerated wound healing following infection with methicillin-resistant Staphylococcus aureus. Conclusion These data suggest that novel nanofibers could effectively shorten the wound-healing time by inhibiting biofilm formation, which make it promising candidates for treatment of MRSA-induced wound infections. ![]()
Point your SmartPhone at the code above. If you have a QR code reader the video abstract will appear. Or use: https://youtu.be/wBXjQQOPzyc
Collapse
Affiliation(s)
- Lei Ran
- Department of Rheumatology and Dermatology, Xinqiao Hospital, Third Military Medical University of Chinese PLA, Chongqing, 430037, People's Republic of China
| | - Shi-Ya Peng
- Department of Rheumatology and Dermatology, Xinqiao Hospital, Third Military Medical University of Chinese PLA, Chongqing, 430037, People's Republic of China
| | - Wei Wang
- Department of Rheumatology and Dermatology, Xinqiao Hospital, Third Military Medical University of Chinese PLA, Chongqing, 430037, People's Republic of China
| | - Qian Wu
- Department of Rheumatology and Dermatology, Xinqiao Hospital, Third Military Medical University of Chinese PLA, Chongqing, 430037, People's Republic of China
| | - Yuan-Chao Li
- Department of Rheumatology and Dermatology, Xinqiao Hospital, Third Military Medical University of Chinese PLA, Chongqing, 430037, People's Republic of China
| | - Ru-Peng Wang
- Department of Rheumatology and Dermatology, Xinqiao Hospital, Third Military Medical University of Chinese PLA, Chongqing, 430037, People's Republic of China
| |
Collapse
|
9
|
Impresari E, Bossi A, Lumina EM, Ortenzi MA, Kothuis JM, Cappelletti G, Maggioni D, Christodoulou MS, Bucci R, Pellegrino S. Fatty Acids/Tetraphenylethylene Conjugates: Hybrid AIEgens for the Preparation of Peptide-Based Supramolecular Gels. Front Chem 2022; 10:927563. [PMID: 36003614 PMCID: PMC9393247 DOI: 10.3389/fchem.2022.927563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Accepted: 06/22/2022] [Indexed: 11/13/2022] Open
Abstract
Aggregation-induced emissive materials are gaining particular attention in the last decades due to their wide application in different fields, from optical devices to biomedicine. In this work, compounds having these kinds of properties, composed of tetraphenylethylene scaffold combined with fatty acids of different lengths, were synthesized and characterized. These molecules were found able to self-assemble into different supramolecular emissive structures depending on the chemical composition and water content. Furthermore, they were used as N-terminus capping agents in the development of peptide-based materials. The functionalization of a 5-mer laminin-derived peptide led to the obtainment of luminescent fibrillary materials that were not cytotoxic and were able to form supramolecular gels in aqueous environment.
Collapse
Affiliation(s)
- Elisa Impresari
- DISFARM, Dipartimento Di Scienze Farmaceutiche, Sezione Chimica Generale e Organica “A. Marchesini”, Università degli Studi di Milano, Milan, Italy
| | - Alberto Bossi
- Istituto di Scienze e Tecnologie Chimiche “G.Natta”, Consiglio Nazionale delle Ricerche (CNR-SCITEC), Milan, Italy
- SmartMatLab Center, Milan, Italy
| | - Edoardo Mario Lumina
- DISFARM, Dipartimento Di Scienze Farmaceutiche, Sezione Chimica Generale e Organica “A. Marchesini”, Università degli Studi di Milano, Milan, Italy
| | - Marco Aldo Ortenzi
- CRC Materiali Polimerici “LaMPo”, Dipartimento di Chimica, Università degli Studi di Milano, Milan, Italy
| | | | | | - Daniela Maggioni
- Dipartimento di Chimica, Università degli Studi di Milano, Milan, Italy
| | - Michael S. Christodoulou
- Departiment of Food, Environmental and Nutritional Sciences (DeFENS), Università degli Studi di Milano, Milan, Italy
| | - Raffaella Bucci
- DISFARM, Dipartimento Di Scienze Farmaceutiche, Sezione Chimica Generale e Organica “A. Marchesini”, Università degli Studi di Milano, Milan, Italy
| | - Sara Pellegrino
- DISFARM, Dipartimento Di Scienze Farmaceutiche, Sezione Chimica Generale e Organica “A. Marchesini”, Università degli Studi di Milano, Milan, Italy
| |
Collapse
|
10
|
Self-Assembled Peptide Nanostructures for ECM Biomimicry. NANOMATERIALS 2022; 12:nano12132147. [PMID: 35807982 PMCID: PMC9268130 DOI: 10.3390/nano12132147] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Revised: 06/18/2022] [Accepted: 06/21/2022] [Indexed: 02/04/2023]
Abstract
Proteins are functional building blocks of living organisms that exert a wide variety of functions, but their synthesis and industrial production can be cumbersome and expensive. By contrast, short peptides are very convenient to prepare at a low cost on a large scale, and their self-assembly into nanostructures and gels is a popular avenue for protein biomimicry. In this Review, we will analyze the last 5-year progress on the incorporation of bioactive motifs into self-assembling peptides to mimic functional proteins of the extracellular matrix (ECM) and guide cell fate inside hydrogel scaffolds.
Collapse
|
11
|
Hao Z, Li H, Wang Y, Hu Y, Chen T, Zhang S, Guo X, Cai L, Li J. Supramolecular Peptide Nanofiber Hydrogels for Bone Tissue Engineering: From Multihierarchical Fabrications to Comprehensive Applications. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2103820. [PMID: 35128831 PMCID: PMC9008438 DOI: 10.1002/advs.202103820] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 01/02/2022] [Indexed: 05/03/2023]
Abstract
Bone tissue engineering is becoming an ideal strategy to replace autologous bone grafts for surgical bone repair, but the multihierarchical complexity of natural bone is still difficult to emulate due to the lack of suitable biomaterials. Supramolecular peptide nanofiber hydrogels (SPNHs) are emerging biomaterials because of their inherent biocompatibility, satisfied biodegradability, high purity, facile functionalization, and tunable mechanical properties. This review initially focuses on the multihierarchical fabrications by SPNHs to emulate natural bony extracellular matrix. Structurally, supramolecular peptides based on distinctive building blocks can assemble into nanofiber hydrogels, which can be used as nanomorphology-mimetic scaffolds for tissue engineering. Biochemically, bioactive motifs and bioactive factors can be covalently tethered or physically absorbed to SPNHs to endow various functions depending on physiological and pharmacological requirements. Mechanically, four strategies are summarized to optimize the biophysical microenvironment of SPNHs for bone regeneration. Furthermore, comprehensive applications about SPNHs for bone tissue engineering are reviewed. The biomaterials can be directly used in the form of injectable hydrogels or composite nanoscaffolds, or they can be used to construct engineered bone grafts by bioprinting or bioreactors. Finally, continuing challenges and outlook are discussed.
Collapse
Affiliation(s)
- Zhuowen Hao
- Department of OrthopedicsZhongnan Hospital of Wuhan UniversityDonghu Road 169Wuhan430071China
| | - Hanke Li
- Department of OrthopedicsZhongnan Hospital of Wuhan UniversityDonghu Road 169Wuhan430071China
| | - Yi Wang
- Department of OrthopedicsZhongnan Hospital of Wuhan UniversityDonghu Road 169Wuhan430071China
| | - Yingkun Hu
- Department of OrthopedicsZhongnan Hospital of Wuhan UniversityDonghu Road 169Wuhan430071China
| | - Tianhong Chen
- Department of OrthopedicsZhongnan Hospital of Wuhan UniversityDonghu Road 169Wuhan430071China
| | - Shuwei Zhang
- Department of OrthopedicsZhongnan Hospital of Wuhan UniversityDonghu Road 169Wuhan430071China
| | - Xiaodong Guo
- Department of OrthopedicsUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyJiefang Road 1277Wuhan430022China
| | - Lin Cai
- Department of OrthopedicsZhongnan Hospital of Wuhan UniversityDonghu Road 169Wuhan430071China
| | - Jingfeng Li
- Department of OrthopedicsZhongnan Hospital of Wuhan UniversityDonghu Road 169Wuhan430071China
| |
Collapse
|
12
|
Kalirajan C, Dukle A, Nathanael AJ, Oh TH, Manivasagam G. A Critical Review on Polymeric Biomaterials for Biomedical Applications. Polymers (Basel) 2021; 13:3015. [PMID: 34503054 PMCID: PMC8433665 DOI: 10.3390/polym13173015] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 09/01/2021] [Accepted: 09/02/2021] [Indexed: 12/18/2022] Open
Abstract
Natural and synthetic polymers have been explored for many years in the field of tissue engineering and regeneration. Researchers have developed many new strategies to design successful advanced polymeric biomaterials. In this review, we summarized the recent notable advancements in the preparation of smart polymeric biomaterials with self-healing and shape memory properties. We also discussed novel approaches used to develop different forms of polymeric biomaterials such as films, hydrogels and 3D printable biomaterials. In each part, the applications of the biomaterials in soft and hard tissue engineering with their in vitro and in vivo effects are underlined. The future direction of the polymeric biomaterials that could pave a path towards successful clinical implications is also underlined in this review.
Collapse
Affiliation(s)
- Cheirmadurai Kalirajan
- Centre for Biomaterials, Cellular and Molecular Theranostics (CBCMT), Vellore Institute of Technology (VIT), Vellore 632014, Tamil Nadu, India; (C.K.); (A.D.); (G.M.)
| | - Amey Dukle
- Centre for Biomaterials, Cellular and Molecular Theranostics (CBCMT), Vellore Institute of Technology (VIT), Vellore 632014, Tamil Nadu, India; (C.K.); (A.D.); (G.M.)
| | - Arputharaj Joseph Nathanael
- Centre for Biomaterials, Cellular and Molecular Theranostics (CBCMT), Vellore Institute of Technology (VIT), Vellore 632014, Tamil Nadu, India; (C.K.); (A.D.); (G.M.)
| | - Tae-Hwan Oh
- School of Chemical Engineering, Yeungnam University, Gyeongsan 38541, Korea
| | - Geetha Manivasagam
- Centre for Biomaterials, Cellular and Molecular Theranostics (CBCMT), Vellore Institute of Technology (VIT), Vellore 632014, Tamil Nadu, India; (C.K.); (A.D.); (G.M.)
| |
Collapse
|
13
|
Kim D, Park D, Kim TH, Chung JJ, Jung Y, Kim SH. Substance P/Heparin-Conjugated PLCL Mitigate Acute Gliosis on Neural Implants and Improve Neuronal Regeneration via Recruitment of Neural Stem Cells. Adv Healthc Mater 2021; 10:e2100107. [PMID: 34227258 DOI: 10.1002/adhm.202100107] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 05/03/2021] [Indexed: 12/15/2022]
Abstract
The inflammatory host tissue response, characterized by gliosis and neuronal death at the neural interface, limits signal transmission and longevity of the neural probe. Substance P induces an anti-inflammatory response and neuronal regeneration and recruits endogenous stem cells. Heparin prevents nonspecific protein adsorption, suppresses the inflammatory response, and is beneficial to neuronal behavior. Poly(l-lactide-co-ε-caprolactone) (PLCL) is a soft and flexible polymer, and PLCL covalently conjugated with biomolecules has been widely used in tissue engineering. Coatings of heparin-conjugated PLCL (Hep-PLCL), substance P-conjugated PLCL (SP-PLCL), and heparin/substance P-conjugated PLCL (Hep/SP-PLCL) reduced the adhesion of astrocytes and fibroblasts and improved neuronal adhesion and neurite development compared to bare glass. The effects of these coatings are evaluated using immunohistochemistry analysis after implantation of coated stainless steel probes in rat brain for 1 week. In particular, Hep/SP-PLCL coating reduced the activation of microglia and astrocytes, the neuronal degeneration caused by inflammation, and indicated a potential for neuronal regeneration at the tissue-device interface. Suppression of the acute host tissue response by coating Hep/SP-PLCL could lead to improved functionality of the neural prosthesis.
Collapse
Affiliation(s)
- Donghak Kim
- KU‐KIST Graduate School of Converging Science and Technology Korea University 145 Anam‐ro, Seongbuk‐gu Seoul 02841 Republic of Korea
- Biomaterials Research Center Korea Institute of Science and Technology (KIST) 5, Hwarang‐ro 14‐gil, Seongbuk‐gu Seoul 02792 Republic of Korea
| | - DoYeun Park
- Biomaterials Research Center Korea Institute of Science and Technology (KIST) 5, Hwarang‐ro 14‐gil, Seongbuk‐gu Seoul 02792 Republic of Korea
| | - Tae Hee Kim
- Biomaterials Research Center Korea Institute of Science and Technology (KIST) 5, Hwarang‐ro 14‐gil, Seongbuk‐gu Seoul 02792 Republic of Korea
| | - Justin J. Chung
- Biomaterials Research Center Korea Institute of Science and Technology (KIST) 5, Hwarang‐ro 14‐gil, Seongbuk‐gu Seoul 02792 Republic of Korea
| | - Youngmee Jung
- Biomaterials Research Center Korea Institute of Science and Technology (KIST) 5, Hwarang‐ro 14‐gil, Seongbuk‐gu Seoul 02792 Republic of Korea
| | - Soo Hyun Kim
- KU‐KIST Graduate School of Converging Science and Technology Korea University 145 Anam‐ro, Seongbuk‐gu Seoul 02841 Republic of Korea
- Biomaterials Research Center Korea Institute of Science and Technology (KIST) 5, Hwarang‐ro 14‐gil, Seongbuk‐gu Seoul 02792 Republic of Korea
| |
Collapse
|
14
|
Sharma P, Pal VK, Roy S. An overview of latest advances in exploring bioactive peptide hydrogels for neural tissue engineering. Biomater Sci 2021; 9:3911-3938. [PMID: 33973582 DOI: 10.1039/d0bm02049d] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Neural tissue engineering holds great potential in addressing current challenges faced by medical therapies employed for the functional recovery of the brain. In this context, self-assembling peptides have gained considerable interest owing to their diverse physicochemical properties, which enable them to closely mimic the biophysical characteristics of the native ECM. Additionally, in contrast to synthetic polymers, which lack inherent biological signaling, peptide-based nanomaterials could be easily designed to present essential biological cues to the cells to promote cellular adhesion. Moreover, injectability of these biomaterials further widens their scope in biomedicine. In this context, hydrogels obtained from short bioactive peptide sequences are of particular interest owing to their facile synthesis and highly tunable properties. In spite of their well-known advantages, the exploration of short peptides for neural tissue engineering is still in its infancy and thus detailed discussion is required to evoke interest in this direction. This review provides a general overview of various bioactive hydrogels derived from short peptide sequences explored for neural tissue engineering. The review also discusses the current challenges in translating the benefits of these hydrogels to clinical practices and presents future perspectives regarding the utilization of these hydrogels for advanced biomedical applications.
Collapse
Affiliation(s)
- Pooja Sharma
- Institute of Nano Science and Technology, Sector 81, Knowledge city, Mohali, 140306, Punjab, India.
| | - Vijay Kumar Pal
- Institute of Nano Science and Technology, Sector 81, Knowledge city, Mohali, 140306, Punjab, India.
| | - Sangita Roy
- Institute of Nano Science and Technology, Sector 81, Knowledge city, Mohali, 140306, Punjab, India.
| |
Collapse
|
15
|
Vargas-Sanchez K, Losada-Barragán M, Mogilevskaya M, Novoa-Herrán S, Medina Y, Buendía-Atencio C, Lorett-Velásquez V, Martínez-Bernal J, Gonzalez-Reyes RE, Ramírez D, Petry KG. Screening for Interacting Proteins with Peptide Biomarker of Blood-Brain Barrier Alteration under Inflammatory Conditions. Int J Mol Sci 2021; 22:ijms22094725. [PMID: 33946948 PMCID: PMC8124558 DOI: 10.3390/ijms22094725] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 04/25/2021] [Accepted: 04/26/2021] [Indexed: 02/06/2023] Open
Abstract
Neurodegenerative diseases are characterized by increased permeability of the blood-brain barrier (BBB) due to alterations in cellular and structural components of the neurovascular unit, particularly in association with neuroinflammation. A previous screening study of peptide ligands to identify molecular alterations of the BBB in neuroinflammation by phage-display, revealed that phage clone 88 presented specific binding affinity to endothelial cells under inflammatory conditions in vivo and in vitro. Here, we aimed to identify the possible target receptor of the peptide ligand 88 expressed under inflammatory conditions. A cross-link test between phage-peptide-88 with IL-1β-stimulated human hCMEC cells, followed by mass spectrometry analysis, was used to identify the target of peptide-88. We modeled the epitope-receptor molecular interaction between peptide-88 and its target by using docking simulations. Three proteins were selected as potential target candidates and tested in enzyme-linked immunosorbent assays with peptide-88: fibronectin, laminin subunit α5 and laminin subunit β-1. Among them, only laminin subunit β-1 presented measurable interaction with peptide-88. Peptide-88 showed specific interaction with laminin subunit β-1, highlighting its importance as a potential biomarker of the laminin changes that may occur at the BBB endothelial cells under pathological inflammation conditions.
Collapse
Affiliation(s)
- Karina Vargas-Sanchez
- Grupo de Neurociencia Translacional, Facultad de Medicina, Universidad de los Andes, Bogotá 111711, Colombia
- Correspondence: ; Tel.: +57-13102405706
| | - Monica Losada-Barragán
- Grupo de Biología Celular y Funcional e Ingeniería de Moléculas, Departamento de Biología, Universidad Antonio Nariño, Bogotá 110231, Colombia; (M.L.-B.); (Y.M.)
| | - Maria Mogilevskaya
- Grupo de Investigación GINIC-HUS, Universidad ECCI, Bogotá 111311, Colombia;
| | - Susana Novoa-Herrán
- Grupo de Investigación en Hormonas (Hormone Research Laboratory), Departamento de Química, Universidad Nacional de Colombia, Bogotá 111321, Colombia; or
- Grupo de Fisiología Molecular, Subdirección de Investigación Científica y Tecnológica, Instituto Nacional de Salud, Bogotá 111321, Colombia
| | - Yehidi Medina
- Grupo de Biología Celular y Funcional e Ingeniería de Moléculas, Departamento de Biología, Universidad Antonio Nariño, Bogotá 110231, Colombia; (M.L.-B.); (Y.M.)
| | - Cristian Buendía-Atencio
- Grupo de Investigación en Modelado y Computación Científica, Departamento de Química, Universidad Antonio Nariño, Bogotá 110231, Colombia;
| | - Vaneza Lorett-Velásquez
- Facultad de Medicina y Ciencias de la Salud, Universidad Militar Nueva Granada, Bogotá 110231, Colombia; (V.L.-V.); (J.M.-B.)
| | - Jessica Martínez-Bernal
- Facultad de Medicina y Ciencias de la Salud, Universidad Militar Nueva Granada, Bogotá 110231, Colombia; (V.L.-V.); (J.M.-B.)
| | - Rodrigo E. Gonzalez-Reyes
- Grupo de Investigación en Neurociencias (NeURos), Centro de Neurociencia Neurovitae-UR, Escuela de Medicina y Ciencias de la Salud, Universidad del Rosario, Bogotá 111711, Colombia;
| | - David Ramírez
- Instituto de Ciencias Biomédicas, Facultad de Ciencias de la Salud, Universidad Autónoma de Chile, El llano Subercaseaux 2801, Santiago 8900000, Chile;
| | - Klaus G. Petry
- INSERM U1049 and U1029 Neuroinflammation and Angiogenesis Group, Bordeaux University, F33000 Bordeaux, France;
| |
Collapse
|
16
|
Jiang J, Dai C, Liu X, Dai L, Li R, Ma K, Xu H, Zhao F, Zhang Z, He T, Niu X, Chen X, Zhang S. Implantation of regenerative complexes in traumatic brain injury canine models enhances the reconstruction of neural networks and motor function recovery. Am J Cancer Res 2021; 11:768-788. [PMID: 33391504 PMCID: PMC7738861 DOI: 10.7150/thno.50540] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Accepted: 10/13/2020] [Indexed: 02/05/2023] Open
Abstract
Rationale: The combination of medical and tissue engineering in neural regeneration studies is a promising field. Collagen, silk fibroin and seed cells are suitable options and have been widely used in the repair of spinal cord injury. In this study, we aimed to determine whether the implantation of a complex fabricated with collagen/silk fibroin (SF) and the human umbilical cord mesenchymal stem cells (hUCMSCs) can promote cerebral cortex repair and motor functional recovery in a canine model of traumatic brain injury (TBI). Methods: A porous scaffold was fabricated with cross-linked collagen and SF. Its physical properties and degeneration rate were measured. The scaffolds were co-cultured with hUCMSCs after which an implantable complex was formed. After complex implantation to a canine model of TBI, the motor evoked potential (MEP) and magnetic resonance imaging (MRI) were used to evaluate the integrity of the cerebral cortex. The neurologic score, motion capture, surface electromyography (sEMG), and vertical ground reaction force (vGRF) were measured in the analysis of motor functions. In vitro analysis of inflammation levels was performed by Elisa while immunohistochemistry was used in track the fate of hUCMSCs. In situ hybridization, transmission electron microscope, and immunofluorescence were used to assess neural and vascular regeneration. Results: Favorable physical properties, suitable degradation rate, and biocompatibility were observed in the collagen/SF scaffolds. The group with complex implantation exhibited the best cerebral cortex integrity and motor functions. The implantation also led to the regeneration of more blood vessels and nerve fibers, less glial fibers, and inflammatory factors. Conclusion: Implantation of this complex enhanced therapy in traumatic brain injury (TBI) through structural repair and functional recovery. These effects exhibit the translational prospects for the clinical application of this complex.
Collapse
|
17
|
Sahab Negah S, Shirzad MM, Biglari G, Naseri F, Hosseini Ravandi H, Hassani Dooghabadi A, Gorji A. Transplantation of R-GSIK scaffold with mesenchymal stem cells improves neuroinflammation in a traumatic brain injury model. Cell Tissue Res 2020; 382:575-583. [PMID: 32715374 PMCID: PMC7683465 DOI: 10.1007/s00441-020-03247-0] [Citation(s) in RCA: 6] [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: 02/04/2020] [Accepted: 06/18/2020] [Indexed: 01/20/2023]
Abstract
Neural tissue engineering has been introduced as a novel therapeutic strategy for traumatic brain injury (TBI). Transplantation of mesenchymal stem cells (MSCs) has been demonstrated to improve functional outcome of brain injury, and RADA4GGSIKVAV (R-GSIK), a self-assembling nano-peptide scaffold, has been suggested to promote the behavior of stem cells. This study was designed to determine the ability of the R-GSIK scaffold in supporting the effects of MSCs on motor function activity and inflammatory responses in an experimental TBI model. A significant recovery of motor function was observed in rats that received MSCs+R-GSIK compared with the control groups. Further analysis showed a reduction in the number of reactive astrocytes and microglial cells in the MSCs and MSCs+R-GSIK groups compared with the control groups. Furthermore, western blot analysis indicated a significant reduction in pro-inflammatory cytokines, such as TLR4, TNF, and IL6, in the MSCs and MSCs+R-GSIK groups compared with the TBI, vehicle, and R-GSIK groups. Overall, this study strengthens the idea that the co-transplantation of MSCs with R-GSIK can increase functional outcomes by preparing a beneficial environment. This improvement may be explained by the immunomodulatory effects of MSCs and the self-assembling nano-scaffold peptide.
Collapse
Affiliation(s)
- Sajad Sahab Negah
- Neuroscience Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Neuroscience, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Shefa Neuroscience Research Center, Khatam Alanbia Hospital, Tehran, Iran
| | | | - Ghazale Biglari
- Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Farzin Naseri
- Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | | | - Ali Gorji
- Neuroscience Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
- Department of Neuroscience, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
- Shefa Neuroscience Research Center, Khatam Alanbia Hospital, Tehran, Iran.
- Department of Neurosurgery and Department of Neurology, Westfälische Wilhelms-Universität, Münster, Germany.
- Epilepsy Research Center, Westfälische Wilhelms-Universität Münster, Münster, Germany.
| |
Collapse
|
18
|
Ding X, Zhao H, Li Y, Lee AL, Li Z, Fu M, Li C, Yang YY, Yuan P. Synthetic peptide hydrogels as 3D scaffolds for tissue engineering. Adv Drug Deliv Rev 2020; 160:78-104. [PMID: 33091503 DOI: 10.1016/j.addr.2020.10.005] [Citation(s) in RCA: 71] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2020] [Revised: 09/25/2020] [Accepted: 10/13/2020] [Indexed: 12/13/2022]
Abstract
The regeneration of tissues and organs poses an immense challenge due to the extreme complexity in the research work involved. Despite the tissue engineering approach being considered as a promising strategy for more than two decades, a key issue impeding its progress is the lack of ideal scaffold materials. Nature-inspired synthetic peptide hydrogels are inherently biocompatible, and its high resemblance to extracellular matrix makes peptide hydrogels suitable 3D scaffold materials. This review covers the important aspects of peptide hydrogels as 3D scaffolds, including mechanical properties, biodegradability and bioactivity, and the current approaches in creating matrices with optimized features. Many of these scaffolds contain peptide sequences that are widely reported for tissue repair and regeneration and these peptide sequences will also be discussed. Furthermore, 3D biofabrication strategies of synthetic peptide hydrogels and the recent advances of peptide hydrogels in tissue engineering will also be described to reflect the current trend in the field. In the final section, we will present the future outlook in the design and development of peptide-based hydrogels for translational tissue engineering applications.
Collapse
Affiliation(s)
- Xin Ding
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen 518107, China.
| | - Huimin Zhao
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen 518107, China
| | - Yuzhen Li
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen 518107, China
| | - Ashlynn Lingzhi Lee
- Institute of Bioengineering and Nanotechnology, 31 Biopolis Way, The Nanos, Singapore 138669, Singapore
| | - Zongshao Li
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen 518107, China
| | - Mengjing Fu
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen 518107, China
| | - Chengnan Li
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen 518107, China
| | - Yi Yan Yang
- Institute of Bioengineering and Nanotechnology, 31 Biopolis Way, The Nanos, Singapore 138669, Singapore.
| | - Peiyan Yuan
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen 518107, China.
| |
Collapse
|
19
|
Granz CL, Gorji A. Dental stem cells: The role of biomaterials and scaffolds in developing novel therapeutic strategies. World J Stem Cells 2020; 12:897-921. [PMID: 33033554 PMCID: PMC7524692 DOI: 10.4252/wjsc.v12.i9.897] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 06/05/2020] [Accepted: 08/16/2020] [Indexed: 02/06/2023] Open
Abstract
Dental stem cells (DSCs) are self-renewable cells that can be obtained easily from dental tissues, and are a desirable source of autologous stem cells. The use of DSCs for stem cell transplantation therapeutic approaches is attractive due to their simple isolation, high plasticity, immunomodulatory properties, and multipotential abilities. Using appropriate scaffolds loaded with favorable biomolecules, such as growth factors, and cytokines, can improve the proliferation, differentiation, migration, and functional capacity of DSCs and can optimize the cellular morphology to build tissue constructs for specific purposes. An enormous variety of scaffolds have been used for tissue engineering with DSCs. Of these, the scaffolds that particularly mimic tissue-specific micromilieu and loaded with biomolecules favorably regulate angiogenesis, cell-matrix interactions, degradation of extracellular matrix, organized matrix formation, and the mineralization abilities of DSCs in both in vitro and in vivo conditions. DSCs represent a promising cell source for tissue engineering, especially for tooth, bone, and neural tissue restoration. The purpose of the present review is to summarize the current developments in the major scaffolding approaches as crucial guidelines for tissue engineering using DSCs and compare their effects in tissue and organ regeneration.
Collapse
Affiliation(s)
- Cornelia Larissa Granz
- Epilepsy Research Center, Westfälische Wilhelms-Universität Münster, Münster 48149, Germany
| | - Ali Gorji
- Epilepsy Research Center, Westfälische Wilhelms-Universität Münster, Münster 48149, Germany
| |
Collapse
|
20
|
Hosseinzadeh S, Lindsay SL, Gallagher AG, Wellings DA, Riehle MO, Riddell JS, Barnett SC. A novel poly-ε-lysine based implant, Proliferate®, for promotion of CNS repair following spinal cord injury. Biomater Sci 2020; 8:3611-3627. [PMID: 32515439 DOI: 10.1039/d0bm00097c] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
The limited regenerative capacity of the CNS poses formidable challenges to the repair of spinal cord injury (SCI). Two key barriers to repair are (i) the physical gap left by the injury, and (ii) the inhibitory milieu surrounding the injury, the glial scar. Biomaterial implantation into the injury site can fill the cavity, provide a substrate for cell migration, and potentially attenuate the glial scar. We investigated the biological viability of a biocompatible and biodegradable poly-ε-lysine based biomaterial, Proliferate®, in low and high cross-linked forms and when coated with IKVAV peptide, for SCI implantation. We demonstrate altered astrocyte morphology and nestin expression on Proliferate® compared to conventional glass cell coverslips suggesting a less reactive phenotype. Moreover Proliferate® supported myelination in vitro, with myelination observed sooner on IKVAV-coated constructs compared with uncoated Proliferate®, and delayed overall compared with maintenance on glass coverslips. For in vivo implantation, parallel-aligned channels were fabricated into Proliferate® to provide cell guidance cues. Extensive vascularisation and cellular infiltration were observed in constructs implanted in vivo, along with an astrocyte border and microglial response. Axonal ingrowth was observed at the construct border and inside implants in intact channels. We conclude that Proliferate® is a promising biomaterial for implantation following SCI.
Collapse
Affiliation(s)
- Sara Hosseinzadeh
- Institute of Infection, Immunity and Inflammation, Sir Graeme Davies Building, 120 University Place, University of Glasgow, Glasgow G12 8TA, UK.
| | | | | | | | | | | | | |
Collapse
|
21
|
|
22
|
Klimek K, Ginalska G. Proteins and Peptides as Important Modifiers of the Polymer Scaffolds for Tissue Engineering Applications-A Review. Polymers (Basel) 2020; 12:E844. [PMID: 32268607 PMCID: PMC7240665 DOI: 10.3390/polym12040844] [Citation(s) in RCA: 95] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 03/31/2020] [Accepted: 04/02/2020] [Indexed: 12/21/2022] Open
Abstract
Polymer scaffolds constitute a very interesting strategy for tissue engineering. Even though they are generally non-toxic, in some cases, they may not provide suitable support for cell adhesion, proliferation, and differentiation, which decelerates tissue regeneration. To improve biological properties, scaffolds are frequently enriched with bioactive molecules, inter alia extracellular matrix proteins, adhesive peptides, growth factors, hormones, and cytokines. Although there are many papers describing synthesis and properties of polymer scaffolds enriched with proteins or peptides, few reviews comprehensively summarize these bioactive molecules. Thus, this review presents the current knowledge about the most important proteins and peptides used for modification of polymer scaffolds for tissue engineering. This paper also describes the influence of addition of proteins and peptides on physicochemical, mechanical, and biological properties of polymer scaffolds. Moreover, this article sums up the major applications of some biodegradable natural and synthetic polymer scaffolds modified with proteins and peptides, which have been developed within the past five years.
Collapse
Affiliation(s)
- Katarzyna Klimek
- Chair and Department of Biochemistry and Biotechnology, Medical University of Lublin, Chodzki 1 Street, 20-093 Lublin, Poland;
| | | |
Collapse
|
23
|
Sahab-Negah S, Hajali V, Moradi HR, Gorji A. The Impact of Estradiol on Neurogenesis and Cognitive Functions in Alzheimer's Disease. Cell Mol Neurobiol 2020; 40:283-299. [PMID: 31502112 DOI: 10.1007/s10571-019-00733-0] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Accepted: 08/31/2019] [Indexed: 12/18/2022]
Abstract
Alzheimer's disease (AD) is described as cognitive and memory impairments with a sex-related epidemiological profile, affecting two times more women than men. There is emerging evidence that alternations in the hippocampal neurogenesis occur at the early stage of AD. Therapies that may effectively slow, stop, or regenerate the dying neurons in AD are being extensively investigated in the last few decades, but none has yet been found to be effective. The regulation of endogenous neurogenesis is one of the main therapeutic targets for AD. Mounting evidence indicates that the neurosteroid estradiol (17β-estradiol) plays a supporting role in neurogenesis, neuronal activity, and synaptic plasticity of AD. This effect may provide preventive and/or therapeutic approaches for AD. In this article, we discuss the molecular mechanism of potential estradiol modulatory action on endogenous neurogenesis, synaptic plasticity, and cognitive function in AD.
Collapse
Affiliation(s)
- Sajad Sahab-Negah
- Neuroscience Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Neuroscience, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Shefa Neuroscience Research Center, Khatam Alanbia Hospital, Tehran, Iran
| | - Vahid Hajali
- Neuroscience Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Neuroscience, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hamid Reza Moradi
- Histology and Embryology Group, Basic Science Department, Faculty of Veterinary Medicine, Shiraz University, Shiraz, Iran
| | - Ali Gorji
- Neuroscience Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
- Department of Neuroscience, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
- Shefa Neuroscience Research Center, Khatam Alanbia Hospital, Tehran, Iran.
- Department of Neurosurgery and Department of Neurology, Westfälische Wilhelms-Universität Münster, Münster, Germany.
- Epilepsy Research Center, Westfälische Wilhelms-Universität Münster, Domagkstr. 11, Münster, Germany.
| |
Collapse
|
24
|
Perera TH, Howell SM, Smith Callahan LA. Manipulation of Extracellular Matrix Remodeling and Neurite Extension by Mouse Embryonic Stem Cells Using IKVAV and LRE Peptide Tethering in Hyaluronic Acid Matrices. Biomacromolecules 2019; 20:3009-3020. [PMID: 31306008 DOI: 10.1021/acs.biomac.9b00578] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Cellular remodeling of the matrix has recently emerged as a key factor in promoting neural differentiation. Most strategies to manipulate matrix remodeling focus on proteolytically cleavable cross-linkers, leading to changes in tethered biochemical signaling and matrix properties. Using peptides that are not the direct target of enzymatic degradation will likely reduce changes in the matrix and improve control of biological behavior. In this study, laminin-derived peptides, IKVAV and LRE, tethered to independent sites in hyaluronic acid matrices using Michael addition and strain-promoted azide-alkyne cycloaddition are sufficient to manipulate hyaluronic acid degradation, gelatinase expression, and protease expression, while promoting neurite extension through matrix metalloprotease-dependent mechanisms in mouse embryonic stem cells encapsulated in hyaluronic acid matrices using an oxidation-reduction reaction initiated gelation. This study provides the foundation for a new strategy to stimulate matrix remodeling that is not dependent on enzymatic cleavage targets.
Collapse
Affiliation(s)
- T Hiran Perera
- Vivian L. Smith Department of Neurosurgery , McGovern Medical School at the University of Texas Health Science Center at Houston McGovern Medical School , Houston , Texas 77030 , United States.,Center for Stem Cell and Regenerative Medicine, Brown Foundation Institute of Molecular Medicine , McGovern Medical School at the University of Texas Health Science Center at Houston , Houston , Texas 77030 , United States
| | - Skyler M Howell
- Vivian L. Smith Department of Neurosurgery , McGovern Medical School at the University of Texas Health Science Center at Houston McGovern Medical School , Houston , Texas 77030 , United States.,Center for Stem Cell and Regenerative Medicine, Brown Foundation Institute of Molecular Medicine , McGovern Medical School at the University of Texas Health Science Center at Houston , Houston , Texas 77030 , United States
| | - Laura A Smith Callahan
- Vivian L. Smith Department of Neurosurgery , McGovern Medical School at the University of Texas Health Science Center at Houston McGovern Medical School , Houston , Texas 77030 , United States.,Center for Stem Cell and Regenerative Medicine, Brown Foundation Institute of Molecular Medicine , McGovern Medical School at the University of Texas Health Science Center at Houston , Houston , Texas 77030 , United States.,Graduate School of Biomedical Sciences , MD Anderson Cancer Center UTHealth , Houston , Texas 77030 , United States
| |
Collapse
|
25
|
Cheong H, Kim J, Kim BJ, Kim E, Park HY, Choi BH, Joo KI, Cho ML, Rhie JW, Lee JI, Cha HJ. Multi-dimensional bioinspired tactics using an engineered mussel protein glue-based nanofiber conduit for accelerated functional nerve regeneration. Acta Biomater 2019; 90:87-99. [PMID: 30978510 DOI: 10.1016/j.actbio.2019.04.018] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2018] [Revised: 04/03/2019] [Accepted: 04/04/2019] [Indexed: 12/16/2022]
Abstract
Limited regenerative capacity of the nervous system makes treating traumatic nerve injuries with conventional polymer-based nerve grafting a challenging task. Consequently, utilizing natural polymers and biomimetic topologies became obvious strategies for nerve conduit designs. As a bioinspired natural polymer from a marine organism, mussel adhesive proteins (MAPs) fused with biofunctional peptides from extracellular matrix (ECM) were engineered for accelerated nerve regeneration by enhancing cell adhesion, proliferation, neural differentiation, and neurite formation. To physically promote contact guidance of neural and Schwann cells and to achieve guided nerve regeneration, MAP was fabricated into an electrospun aligned nanofiber conduit by introducing synthetic polymer poly(lactic-co-glycolic acid) (PLGA) to control solubility and mechanical property. In vitro and in vivo experiments demonstrated that the multi-dimensional tactics of combining adhesiveness from MAP, integrin-mediated interaction from ECM peptides (in particular, IKVAV derived from laminin α1 chain), and contact guidance from aligned nanofibers synergistically accelerated functional nerve regeneration. Thus, MAP-based multi-dimensional approach provides new opportunities for neural regenerative applications including nerve grafting. STATEMENT OF SIGNIFICANCE: Findings in neural regeneration indicate that a bioinspired polymer-based nerve conduit design should harmoniously constitute various factors, such as biocompatibility, neurotrophic molecule, biodegradability, and contact guidance. Here, we engineered three fusion proteins of mussel-derived adhesive protein with ECM-derived biofunctional peptides to simultaneously provide biocompatibility and integrin-based interactions. In addition, a fabrication of robust aligned nanofiber conduits containing the fusion proteins realized suitable biodegradability and contact guidance. Thus, our multi-dimensional strategy on conduit design provided outstanding biocompatibility, biodegradability, integrin-interaction, and contact guidance to achieve an accelerated functional nerve regeneration. We believe that our bioengineered mussel adhesive protein-based multi-dimensional strategy would offer new insights into the design of nerve tissue engineering biomaterials.
Collapse
|
26
|
Seidlits SK, Liang J, Bierman RD, Sohrabi A, Karam J, Holley SM, Cepeda C, Walthers CM. Peptide-modified, hyaluronic acid-based hydrogels as a 3D culture platform for neural stem/progenitor cell engineering. J Biomed Mater Res A 2019; 107:704-718. [PMID: 30615255 PMCID: PMC8862560 DOI: 10.1002/jbm.a.36603] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Revised: 12/23/2018] [Accepted: 01/03/2019] [Indexed: 07/26/2023]
Abstract
Neural stem/progenitor cell (NS/PC)-based therapies have shown exciting potential for regeneration of the central nervous system (CNS) and NS/PC cultures represent an important resource for disease modeling and drug screening. However, significant challenges limiting clinical translation remain, such as generating large numbers of cells required for model cultures or transplantation, maintaining physiologically representative phenotypes ex vivo and directing NS/PC differentiation into specific fates. Here, we report that culture of human NS/PCs in 3D, hyaluronic acid (HA)-rich biomaterial microenvironments increased differentiation toward oligodendrocytes and neurons over 2D cultures on laminin-coated glass. Moreover, NS/PCs in 3D culture exhibited a significant reduction in differentiation into reactive astrocytes. Many NS/PC-derived neurons in 3D, HA-based hydrogels expressed synaptophysin, indicating synapse formation, and displayed electrophysiological characteristics of immature neurons. While inclusion of integrin-binding, RGD peptides into hydrogels resulted in a modest increase in numbers of viable NS/PCs, no combination of laminin-derived, adhesive peptides affected differentiation outcomes. Notably, 3D cultures of differentiating NS/PCs were maintained for at least 70 days in medium with minimal growth factor supplementation. In sum, results demonstrate the use of 3D, HA-based biomaterials for long-term expansion and differentiation of NS/PCs toward oligodendroglial and neuronal fates, while inhibiting astroglial fates. © 2019 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 107A: 704-718, 2019.
Collapse
Affiliation(s)
- Stephanie K. Seidlits
- Department of Bioengineering, UCLA, Los Angels, California
- Board Stem Cell Research Center, UCLA, Los Angels, California
- Brain Research Institute, UCLA, Los Angels, California
- Jonsson Comprehensive Cancer Center, UCLA, Los Angels, California
- Center for Minimally Invasive Therapeutics, UCLA, Los Angels, California
| | - Jesse Liang
- Department of Bioengineering, UCLA, Los Angels, California
| | | | | | - Joshua Karam
- Department of Bioengineering, UCLA, Los Angels, California
| | - Sandra M. Holley
- Intellectual and Developmental Disabilities Research Center, Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine, UCLA, Los Angeles, California
| | - Carlos Cepeda
- Intellectual and Developmental Disabilities Research Center, Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine, UCLA, Los Angeles, California
| | | |
Collapse
|
27
|
D’Amato AR, Puhl DL, Ziemba AM, Johnson CDL, Doedee J, Bao J, Gilbert RJ. Exploring the effects of electrospun fiber surface nanotopography on neurite outgrowth and branching in neuron cultures. PLoS One 2019; 14:e0211731. [PMID: 30716106 PMCID: PMC6361509 DOI: 10.1371/journal.pone.0211731] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Accepted: 01/18/2019] [Indexed: 12/19/2022] Open
Abstract
Three aligned, electrospun fiber scaffolds with unique surface features were created from poly-L-lactic acid (PLLA). Fibers without surface nanotopography (smooth fibers), fibers with surface divots (shallow pits), and fibers with surface pits (deeper pits) were fabricated, and fiber alignment, diameter, and density were characterized using scanning electron microscopy (SEM). Whole dorsal root ganglia (DRG) were isolated from rats and placed onto uncoated fibers or fibers coated with laminin. On uncoated fibers, neurite outgrowth was restricted by fibers displaying divoted or pitted nanotopography when compared to neurite outgrowth on smooth fibers. However, neurites extending from whole DRG cultured on laminin-coated fibers were not restricted by divoted or pitted surface nanotopography. Thus, neurites extending on laminin-coated fibers were able to extend long neurites even in the presence of surface divots or pits. To further explore this result, individual neurons isolated from dissociated DRG were seeded onto laminin-coated smooth, pitted, or divoted fibers. Interestingly, neurons on pitted or divoted fibers exhibited a 1.5-fold increase in total neurite length, and a 2.3 or 2.7-fold increase in neurite branching compared to neurons on smooth fibers, respectively. Based on these findings, we conclude that fiber roughness in the form of pits or divots can promote extension and branching of long neurites along aligned electrospun fibers in the presence of an extracellular matrix protein coating. Thus, aligned, electrospun fibers can be crafted to not only direct the extension of axons but to induce unique branching morphologies.
Collapse
Affiliation(s)
- Anthony R. D’Amato
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, New York, United States of America
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, New York,United States of America
| | - Devan L. Puhl
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, New York, United States of America
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, New York,United States of America
| | - Alexis M. Ziemba
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, New York, United States of America
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, New York,United States of America
| | - Christopher D. L. Johnson
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, New York, United States of America
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, New York,United States of America
| | - Janneke Doedee
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, New York, United States of America
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, New York,United States of America
| | - Jonathan Bao
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, New York, United States of America
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, New York,United States of America
| | - Ryan J. Gilbert
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, New York, United States of America
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, New York,United States of America
| |
Collapse
|
28
|
Jahanbazi Jahan-Abad A, Karima S, Sahab Negah S, Noorbakhsh F, Borhani-Haghighi M, Gorji A. Therapeutic potential of conditioned medium derived from oligodendrocytes cultured in a self-assembling peptide nanoscaffold in experimental autoimmune encephalomyelitis. Brain Res 2019; 1711:226-235. [PMID: 30703369 DOI: 10.1016/j.brainres.2019.01.035] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Accepted: 01/26/2019] [Indexed: 01/12/2023]
Abstract
The use of neurotrophic factors is considered to be a novel therapeutic approach for restoring and/or maintaining neurological function in neurodegenerative disorders, such as multiple sclerosis (MS). Various studies have shown that conditioned medium produced by oligodendrocyte (OL-CM) contain a variety of neurotrophic factors. Here, we investigated the restorative effects of OL-CM, collected from oligodendrocytes cultured in a self-assembling peptide hydrogels scaffold (PuraMatrix), in experimental autoimmune encephalomyelitis (EAE) mouse model. Neural stem/progenitor cells, isolated from the embryonic mouse brain, were cultured and differentiated into oligodendrocyte. Cell viability and proliferation of oligodendrocytes were assessed by live/dead and MTT assays. Motor functions, myelination, cell infiltration, gliosis, and inflammatory process were assessed in EAE mice after intracranial injection of OL-CM at different concentrations. Application of OL-CM improved clinical score and neurological function in EAE mice and reduced the inflammatory cell infiltration and demyelination. Furthermore, administration of OL-CM reduced the expression of pro-inflammatory cytokines and suppressed the activation of NLRP3-inflammasome complex in EAE mice. These data suggest the potential therapeutic effect of OL-CM for MS treatment.
Collapse
Affiliation(s)
- Ali Jahanbazi Jahan-Abad
- Department of Clinical Biochemistry, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Saeed Karima
- Department of Clinical Biochemistry, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Sajad Sahab Negah
- Department of Neuroscience, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Shefa Neuroscience Research Center, Khatam Alanbia Hospital, Tehran, Iran
| | - Farshid Noorbakhsh
- Department of Immunology, Building No. 7, School of Medicine, Tehran University of Medical Sciences, Poursina Avenue, Tehran, Iran; Shefa Neuroscience Research Center, Khatam Alanbia Hospital, Tehran, Iran
| | - Maryam Borhani-Haghighi
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran; Shefa Neuroscience Research Center, Khatam Alanbia Hospital, Tehran, Iran
| | - Ali Gorji
- Department of Neuroscience, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Shefa Neuroscience Research Center, Khatam Alanbia Hospital, Tehran, Iran; Department of Neurosurgery, Department of Neurology, Westfälische Wilhelms-Universität Münster, Münster, Germany; Epilepsy Research Center, Westfälische Wilhelms-Universität Münster, Münster, Germany.
| |
Collapse
|
29
|
Jahanbazi Jahan-Abad A, Sahab Negah S, Hosseini Ravandi H, Ghasemi S, Borhani-Haghighi M, Stummer W, Gorji A, Khaleghi Ghadiri M. Human Neural Stem/Progenitor Cells Derived From Epileptic Human Brain in a Self-Assembling Peptide Nanoscaffold Improve Traumatic Brain Injury in Rats. Mol Neurobiol 2018; 55:9122-9138. [PMID: 29651746 DOI: 10.1007/s12035-018-1050-8] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2018] [Accepted: 03/27/2018] [Indexed: 01/09/2023]
Abstract
Traumatic brain injury (TBI) is a disruption in the brain functions following a head trauma. Cell therapy may provide a promising treatment for TBI. Among different cell types, human neural stem cells cultured in self-assembling peptide scaffolds have been suggested as a potential novel method for cell replacement treatment after TBI. In the present study, we accessed the effects of human neural stem/progenitor cells (hNS/PCs) derived from epileptic human brain and human adipose-derived stromal/stem cells (hADSCs) seeded in PuraMatrix hydrogel (PM) on brain function after TBI in an animal model of brain injury. hNS/PCs were isolated from patients with medically intractable epilepsy undergone epilepsy surgery. hNS/PCs and hADSCs have the potential for proliferation and differentiation into both neuronal and glial lineages. Assessment of the growth characteristics of hNS/PCs and hADSCs revealed that the hNS/PCs doubling time was significantly longer and the growth rate was lower than hADSCs. Transplantation of hNS/PCs and hADSCs seeded in PM improved functional recovery, decreased lesion volume, inhibited neuroinflammation, and reduced the reactive gliosis at the injury site. The data suggest the transplantation of hNS/PCs or hADSCs cultured in PM as a promising treatment option for cell replacement therapy in TBI.
Collapse
Affiliation(s)
- Ali Jahanbazi Jahan-Abad
- Shefa Neuroscience Research Center, Khatam Alanbia Hospital, Tehran, Iran.,Department of Clinical Biochemistry, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sajad Sahab Negah
- Shefa Neuroscience Research Center, Khatam Alanbia Hospital, Tehran, Iran.,Department of Neuroscience, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Sedigheh Ghasemi
- Shefa Neuroscience Research Center, Khatam Alanbia Hospital, Tehran, Iran
| | | | - Walter Stummer
- Department of Neurosurgery, Westfälische Wilhelms-Universität Münster, Münster, Germany
| | - Ali Gorji
- Shefa Neuroscience Research Center, Khatam Alanbia Hospital, Tehran, Iran. .,Department of Neuroscience, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran. .,Department of Neurosurgery, Westfälische Wilhelms-Universität Münster, Münster, Germany. .,Department of Neurology, Westfälische Wilhelms-Universität Münster, Münster, Germany. .,Epilepsy Research Center, Westfälische Wilhelms-Universität Münster, Robert-Koch-Strasse 45, 48149, Münster, Germany.
| | | |
Collapse
|