1
|
Wang T, Li Z, Lei J, Zhang Y, Tong Y, Guan X, Wang S. RGD peptide-functionalized micelles loaded with crocetin ameliorate doxorubicin-induced cardiotoxicity. Int J Pharm X 2025; 9:100326. [PMID: 40103672 PMCID: PMC11914822 DOI: 10.1016/j.ijpx.2025.100326] [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: 12/06/2024] [Revised: 02/24/2025] [Accepted: 02/25/2025] [Indexed: 03/20/2025] Open
Abstract
Doxorubicin (Dox)-induced cardiotoxicity presents a significant challenge to fully harnessing its chemotherapeutic potential. Crocetin (Cro), a dicarboxylic acid found in the crocus flower and gardenia fruit, has shown remarkable antioxidant and anti-inflammatory activities. However, its poor aqueous solubility and limited cellular uptake severely constrain its further application in treating diseases. In this study, we developed Arg-Gly-Asp (RGD) peptide-decorated nanomicelles delivering Cro to alleviate Dox-induced cardiac injury. The RGD@M(Cro) nanomicelles exhibited excellent aqueous solubility and a drug-loading efficiency of 93.3 %. RGD-decorated micelles could enhance the cellular uptake of Cro in cardiomyocytes and inhibit approximately 60 % of HL-1 cell apoptosis through efficient reactive oxygen species (ROS) scavenging. In a cardiomyopathy mouse model, RGD@M(Cro) substantially reduced cardiac damage and improved cardiac indicators. This study highlights the great potential of RGD-decorated micelles in treating cardiac injury and other diseases.
Collapse
Affiliation(s)
- Ting Wang
- Department of Cardiology, The First People's Hospital of Wenling (Taizhou University Affiliated Wenling Hospital), School of Medicine, Taizhou University, Taizhou 317500, PR China
| | - Zhimin Li
- College of Medical Technology, Beihua University, Jilin 132013, PR China
| | - Jiawei Lei
- Department of Cardiology, The First People's Hospital of Wenling (Taizhou University Affiliated Wenling Hospital), School of Medicine, Taizhou University, Taizhou 317500, PR China
| | - Yuchen Zhang
- Department of Cardiology, The First People's Hospital of Wenling (Taizhou University Affiliated Wenling Hospital), School of Medicine, Taizhou University, Taizhou 317500, PR China
| | - Yingpeng Tong
- Department of Cardiology, The First People's Hospital of Wenling (Taizhou University Affiliated Wenling Hospital), School of Medicine, Taizhou University, Taizhou 317500, PR China
| | - Xingang Guan
- Department of Cardiology, The First People's Hospital of Wenling (Taizhou University Affiliated Wenling Hospital), School of Medicine, Taizhou University, Taizhou 317500, PR China
| | - Shuangshuang Wang
- Department of Cardiology, The First People's Hospital of Wenling (Taizhou University Affiliated Wenling Hospital), School of Medicine, Taizhou University, Taizhou 317500, PR China
- Key Laboratory of Precision Medicine for Atherosclerotic Diseases of Zhejiang Province, Affiliated First Hospital of Ningbo University, Ningbo 315010, China
| |
Collapse
|
2
|
Vithalani H, Dave H, Singh H, Sharma D, Navale A, Dhanka M. Mechanically robust, mouldable, dynamically crosslinked hydrogel flap with multiple functionalities for accelerated deep skin wound healing. BIOMATERIALS ADVANCES 2025; 169:214195. [PMID: 39862713 DOI: 10.1016/j.bioadv.2025.214195] [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: 10/26/2024] [Revised: 12/30/2024] [Accepted: 01/18/2025] [Indexed: 01/27/2025]
Abstract
Deep cutaneous wounds, which are difficult to heal and specifically occur on dynamic body surfaces, remain a substantial healthcare challenge in clinical practice because of multiple underlying factors, including excessive reactive oxygen species, potential bacterial infection, and extensive degradation of the extracellular matrix (ECM) which further leads to the progressive deterioration of the wound microenvironment. Any available individual wound therapy, such as antibiotic-loaded cotton gauze, cannot address all these issues. Engineering an advanced multifunctional wound dressing is the current need to promote the overall healing process of such wounds. Here, we report a multifunctional hydrogel flap primarily composed of biodegradable polymers gelatin (G) and poly-methyl vinyl ether-alt-maleic acid (MA) as the base material. The hydrogel physically incorporates tannic acid (TA) and vancomycin (V), for added functionality. The resulting hydrogel flap, gelatin- poly-methyl vinyl ether-alt-maleic acid-tannic acid-vancomycin (G-MA-TA-V/E-N), is formed through a chemical crosslinking process using EDC (E) and NHS (N). Thus, the hydrogel flap reveals multiple ideal properties that support its ease of application, including stretchability, porous microstructure (honey-comb structure), mouldability, and adhesiveness to multiple surfaces, including wet biological surfaces. The in vitro studies demonstrated strong antioxidant, antibacterial, and absorption properties essential for accelerated wound-healing applications. In vivo studies further reveal accelerated wound contraction and enhanced healing kinetics, promoting re-epithelialization, angiogenesis, and formation of apocrine glands. These findings underscore the efficacy and cost-effectiveness of fabricated hydrogel flaps as viable therapeutic options for treating deep skin wounds and make it worthwhile to integrate them with medical devices for tissue adhesion.
Collapse
Affiliation(s)
- Hitasha Vithalani
- Department of Biological Sciences and Engineering, Indian Institute of Technology Gandhinagar, Gujarat, India
| | - Harshil Dave
- Department of Biological Sciences and Engineering, Indian Institute of Technology Gandhinagar, Gujarat, India
| | - Hemant Singh
- Department of Biological Sciences and Engineering, Indian Institute of Technology Gandhinagar, Gujarat, India
| | - Dinesh Sharma
- Department of Pharmacy, Parul University, Vadodara, Gujarat, India
| | - Archana Navale
- Department of Pharmacy, Parul University, Vadodara, Gujarat, India
| | - Mukesh Dhanka
- Department of Biological Sciences and Engineering, Indian Institute of Technology Gandhinagar, Gujarat, India.
| |
Collapse
|
3
|
Jammes M, Tabasi A, Bach T, Ritter T. Healing the cornea: Exploring the therapeutic solutions offered by MSCs and MSC-derived EVs. Prog Retin Eye Res 2025; 105:101325. [PMID: 39709150 DOI: 10.1016/j.preteyeres.2024.101325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 12/16/2024] [Accepted: 12/17/2024] [Indexed: 12/23/2024]
Abstract
Affecting a large proportion of the population worldwide, corneal disorders constitute a concerning health hazard associated to compromised eyesight or blindness for most severe cases. In the last decades, mesenchymal stem/stromal cells (MSCs) demonstrated promising abilities in improving symptoms associated to corneal diseases or alleviating these affections, especially through their anti-inflammatory, immunomodulatory and pro-regenerative properties. More recently, MSC therapeutic potential was shown to be mediated by the molecules they release, and particularly by their extracellular vesicles (EVs; MSC-EVs). Consequently, using MSC-EVs emerged as a pioneering strategy to mitigate the risks related to cell therapy while providing MSC therapeutic benefits. Despite the promises given by MSC- and MSC-EV-based approaches, many improvements are considered to optimize the therapeutic significance of these therapies. This review aspires to provide a comprehensive and detailed overview of current knowledge on corneal therapies involving MSCs and MSC-EVs, the strategies currently under evaluation, and the gaps remaining to be addressed for clinical implementation. From encapsulating MSCs or their EVs into biomaterials to enhance the ocular retention time to loading MSC-EVs with therapeutic drugs, a wide range of ground-breaking strategies are currently contemplated to lead to the safest and most effective treatments. Promising research initiatives also include diverse gene therapies and the targeting of specific cell types through the modification of the EV surface, paving the way for future therapeutic innovations. As one of the most important challenges, MSC-EV large-scale production strategies are extensively investigated and offer a wide array of possibilities to meet the needs of clinical applications.
Collapse
Affiliation(s)
- Manon Jammes
- Regenerative Medicine Institute, School of Medicine, University of Galway, Galway, Ireland
| | - Abbas Tabasi
- Regenerative Medicine Institute, School of Medicine, University of Galway, Galway, Ireland
| | - Trung Bach
- Regenerative Medicine Institute, School of Medicine, University of Galway, Galway, Ireland
| | - Thomas Ritter
- Regenerative Medicine Institute, School of Medicine, University of Galway, Galway, Ireland; CURAM Centre for Research in Medical Devices, University of Galway, Galway, Ireland.
| |
Collapse
|
4
|
Ławkowska K, Bonowicz K, Jerka D, Bai Y, Gagat M. Integrins in Cardiovascular Health and Disease: Molecular Mechanisms and Therapeutic Opportunities. Biomolecules 2025; 15:233. [PMID: 40001536 PMCID: PMC11853560 DOI: 10.3390/biom15020233] [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: 12/18/2024] [Revised: 01/30/2025] [Accepted: 02/01/2025] [Indexed: 02/27/2025] Open
Abstract
Cardiovascular diseases, including atherosclerosis, hypertension, and heart failure, remain the leading cause of global mortality, with endothelial dysfunction and vascular remodeling as critical contributors. Integrins, as transmembrane adhesion proteins, are central regulators of cell adhesion, migration, and signaling, playing a pivotal role in maintaining vascular homeostasis and mediating pathological processes such as inflammation, angiogenesis, and extracellular matrix remodeling. This article comprehensively examines the role of integrins in the pathogenesis of cardiovascular diseases, focusing on their dysfunction in endothelial cells and interactions with inflammatory mediators, such as TNF-α. Molecular mechanisms of integrin action are discussed, including their involvement in mechanotransduction, leukocyte adhesion, and signaling pathways that regulate vascular integrity. The review also highlights experimental findings, such as the use of specific integrin-targeting plasmids and immunofluorescence to elucidate integrin functions under inflammatory conditions. Additionally, potential therapeutic strategies are explored, including the development of integrin inhibitors, monoclonal antibodies, and their application in regenerative medicine. These approaches aim not only to mitigate pathological vascular remodeling but also to promote tissue repair and angiogenesis. By bridging insights from molecular studies with their translational potential, this work underscores the promise of integrin-based therapies in advancing the management and treatment of cardiovascular diseases.
Collapse
Affiliation(s)
- Karolina Ławkowska
- Department of Histology and Embryology, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, 85-092 Bydgoszcz, Poland
| | - Klaudia Bonowicz
- Department of Histology and Embryology, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, 85-092 Bydgoszcz, Poland
- Collegium Medicum, Mazovian Academy in Płock, 09-402 Płock, Poland
| | - Dominika Jerka
- Department of Histology and Embryology, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, 85-092 Bydgoszcz, Poland
| | - Yidong Bai
- Department of Cell Systems and Anatomy, University of Texas Health San Antonio, San Antonio, TX 78229, USA;
| | - Maciej Gagat
- Department of Histology and Embryology, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, 85-092 Bydgoszcz, Poland
- Collegium Medicum, Mazovian Academy in Płock, 09-402 Płock, Poland
| |
Collapse
|
5
|
Castillo-Díaz LA, Gough JE, Miller AF, Saiani A. RGD-functionalised self-assembling peptide hydrogel induces a proliferative profile in human osteoblasts in vitro. J Pept Sci 2025; 31:e3653. [PMID: 39329311 DOI: 10.1002/psc.3653] [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: 07/05/2024] [Revised: 08/21/2024] [Accepted: 08/22/2024] [Indexed: 09/28/2024]
Abstract
Self-assembling peptide hydrogels (SAPHs) have been used in the past decade as reliable three-dimensional (3D) synthetic scaffolds for the culture of a variety of mammalian cells in vitro. Thanks to their versatile physicochemical properties, they allow researchers to tailor the hydrogel properties, including stiffness and functionality to the targeted cells and cells' behaviour. One of the advantages of using SAPH scaffolds is the ease of functionalisation. In the present work, we discuss the effect that functionalising the FEFEFKFK (F, phenylalanine; K, lysine; and E, glutamic acid) hydrogel scaffold using the cell-binding RGDS (fibronectin - R, arginine; G, glycine; D, aspartic acid; S, serine) epitope affects the material properties as well as the function of encapsulated human osteoblast cells. RGDS functionalisation resulted in cells adopting an elongated morphology, suggesting attachment and increased proliferation. While this led to higher cell viability, it also resulted in a decrease in extra-cellular matrix (ECM) protein production as well as a decrease in calcium ion deposition, suggesting lower mineralisation capabilities. The work clearly shows that SAPHs are a flexible platform that allow the modification of scaffolds in a controlled manner to investigate cell-material interactions.
Collapse
Affiliation(s)
- Luis A Castillo-Díaz
- School of Chemical Engineering & Manchester Institute of Biotechnology, The University of Manchester, Manchester, UK
- Departamento de Medicina y Ciencias de la Salud, Facultad Interdisciplinaria de Ciencias Biológicas y de la Salud, Universidad de Sonora, Hermosillo, Sonora, Mexico
| | - Julie E Gough
- School of Materials & Henry Royce Institute, The University of Manchester, Manchester, UK
| | - Aline F Miller
- School of Chemical Engineering & Manchester Institute of Biotechnology, The University of Manchester, Manchester, UK
| | - Alberto Saiani
- Division of Pharmacy and Optometry & Manchester Institute of Biotechnology, The University of Manchester, Manchester, UK
| |
Collapse
|
6
|
Galindo JM, Merino S, Herrero MA. Advanced Hydrogels: Enhancing Tissue Bioengineering with RGD Peptides and Carbon Nanomaterials. ChemMedChem 2025; 20:e202400587. [PMID: 39446356 PMCID: PMC11793852 DOI: 10.1002/cmdc.202400587] [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: 07/30/2024] [Revised: 10/22/2024] [Accepted: 10/23/2024] [Indexed: 11/21/2024]
Abstract
The advancement of tissue engineering (TE) is driven by the development of scaffolds that mimic the mechanical, spatial, and biological environment of the extracellular matrix (ECM), crucial for regulating cell behavior and tissue repair. Hydrogels, 3D networks of polymer chains, are particularly suited for TE due to their high biocompatibility, ability to mimic tissue water content, facilitate cell migration, sustain growth factor release, and offer controllable physical properties. However, hydrogels mimicking the ECM often face challenges related to cell adhesion due to the absence of specific receptors. This issue can be addressed by incorporating ECM components into the polymer matrix, such as the peptide sequence arginine-glycine-aspartic acid (RGD), known for its role in cell adhesion. Additionally, carbon nanomaterials (CNMs) offer unique physicochemical properties that can improve scaffold-cell interactions. Despite the potential benefits, there are limited reports on their combination. RGD-CNM hydrogels enable a more accurate emulation of the natural cellular environment, enhancing tissue engineering applications. This hybrid approach may promote robust cell adhesion along with exceptional mechanical and electrical properties. This review outlines the potential benefits of these hybrid scaffolds and their synergistic potential, aiming to inspire new research directions in this innovative field.
Collapse
Affiliation(s)
- Josué M. Galindo
- Departamento de Química InorgánicaOrgánica y BioquímicaFacultad de Ciencias y Tecnologías QuímicasUniversidad de Castilla-La Mancha13071Ciudad RealSpain
- Instituto Regional de Investigación Científica Aplicada (IRICA)UCLM13071Ciudad RealSpain
| | - Sonia Merino
- Departamento de Química InorgánicaOrgánica y BioquímicaFacultad de Ciencias y Tecnologías QuímicasUniversidad de Castilla-La Mancha13071Ciudad RealSpain
- Instituto Regional de Investigación Científica Aplicada (IRICA)UCLM13071Ciudad RealSpain
| | - M. Antonia Herrero
- Departamento de Química InorgánicaOrgánica y BioquímicaFacultad de Ciencias y Tecnologías QuímicasUniversidad de Castilla-La Mancha13071Ciudad RealSpain
- Instituto Regional de Investigación Científica Aplicada (IRICA)UCLM13071Ciudad RealSpain
| |
Collapse
|
7
|
Zimmermann P, Schulze P, Beck-Sickinger AG, Khrunyk Y. Design and Biofunctionalization of Cloud Sponge-Inspired Scaffolds for Enhanced Bone Cell Performance. ACS APPLIED BIO MATERIALS 2024; 7:8281-8293. [PMID: 39548985 PMCID: PMC11653246 DOI: 10.1021/acsabm.4c01065] [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: 07/30/2024] [Revised: 11/07/2024] [Accepted: 11/10/2024] [Indexed: 11/18/2024]
Abstract
With the increasing age of our population, which is linked to a higher incidence of musculoskeletal diseases, there is a massive clinical need for bone implants. Porous scaffolds, usually offering a lower stiffness and allowing for the ingrowth of blood vessels and nerves, serve as an attractive alternative to conventional implants. Natural porous skeletons from marine sponges represent an array of evolutionarily optimized patterns, inspiring the design of biomaterials. In this study, cloud sponge-inspired scaffolds were designed and printed from a photocurable polymer, Clear Resin. These scaffolds were biofunctionalized by mussel-derived peptide MP-RGD, a recently developed peptide that contains a cyclic, bioactive RGD cell adhesion motif and catechol moieties, which provide the anchoring of the peptide to the surface. In in vitro cell culture assays with bone cells, significantly higher biocompatibility of three scaffolds, i.e., square, octagon, and hexagon cubes, in comparison to hollow and sphere inside cubes was shown. The performance of the cells regarding signaling was further enhanced by applying an MP-RGD coating. Consequently, these data demonstrate that both the structure of the scaffold and the coating contribute to the biocompatibility of the material. Three out of five MP-RGD-coated sponge-inspired scaffolds displayed superior biochemical properties and might guide material design for improved bone implants.
Collapse
Affiliation(s)
- Philipp Zimmermann
- Engineering
Faculty, Leipzig University of Applied Sciences
(HTWK), Karl Liebknecht Str. 134, D-04277 Leipzig, Germany
| | - Peter Schulze
- Engineering
Faculty, Leipzig University of Applied Sciences
(HTWK), Karl Liebknecht Str. 134, D-04277 Leipzig, Germany
| | - Annette G. Beck-Sickinger
- Institute
of Biochemistry, Faculty of Life Sciences, Leipzig University, Brüderstrs. 34, D-04103 Leipzig, Germany
| | - Yuliya Khrunyk
- Institute
of Biochemistry, Faculty of Life Sciences, Leipzig University, Brüderstrs. 34, D-04103 Leipzig, Germany
| |
Collapse
|
8
|
Toader C, Dumitru AV, Eva L, Serban M, Covache-Busuioc RA, Ciurea AV. Nanoparticle Strategies for Treating CNS Disorders: A Comprehensive Review of Drug Delivery and Theranostic Applications. Int J Mol Sci 2024; 25:13302. [PMID: 39769066 PMCID: PMC11676454 DOI: 10.3390/ijms252413302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 12/06/2024] [Accepted: 12/10/2024] [Indexed: 01/11/2025] Open
Abstract
This review aims to address the significant challenges of treating central nervous system (CNS) disorders such as neurodegenerative diseases, strokes, spinal cord injuries, and brain tumors. These disorders are difficult to manage due to the complexity of disease mechanisms and the protective blood-brain barrier (BBB), which restricts drug delivery. Recent advancements in nanoparticle (NP) technologies offer promising solutions, with potential applications in drug delivery, neuroprotection, and neuroregeneration. By examining current research, we explore how NPs can cross the BBB, deliver medications directly to targeted CNS regions, and enhance both diagnostics and treatment. Key NP strategies, such as passive targeting, receptor-mediated transport, and stimuli-responsive systems, demonstrate encouraging results. Studies show that NPs may improve drug delivery, minimize side effects, and increase therapeutic effectiveness in models of Alzheimer's, Parkinson's, stroke, and glioblastoma. NP technologies thus represent a promising approach for CNS disorder management, combining drug delivery and diagnostic capabilities to enable more precise and effective treatments that could significantly benefit patient outcomes.
Collapse
Affiliation(s)
- Corneliu Toader
- Department of Neurosurgery, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania; (C.T.); (M.S.); (R.-A.C.-B.); (A.V.C.)
- Department of Vascular Neurosurgery, National Institute of Neurology and Neurovascular Diseases, 077160 Bucharest, Romania
| | - Adrian Vasile Dumitru
- Department of Pathology, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Department of Pathology, University Emergency Hospital of Bucharest, 050098 Bucharest, Romania
| | - Lucian Eva
- Department of Neurosurgery, Dunarea de Jos University, 800010 Galati, Romania
- Department of Neurosurgery, Clinical Emergency Hospital “Prof. Dr. Nicolae Oblu”, 700309 Iasi, Romania
| | - Matei Serban
- Department of Neurosurgery, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania; (C.T.); (M.S.); (R.-A.C.-B.); (A.V.C.)
| | - Razvan-Adrian Covache-Busuioc
- Department of Neurosurgery, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania; (C.T.); (M.S.); (R.-A.C.-B.); (A.V.C.)
- Department of Vascular Neurosurgery, National Institute of Neurology and Neurovascular Diseases, 077160 Bucharest, Romania
| | - Alexandru Vlad Ciurea
- Department of Neurosurgery, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania; (C.T.); (M.S.); (R.-A.C.-B.); (A.V.C.)
- Neurosurgery Department, Sanador Clinical Hospital, 010991 Bucharest, Romania
- Medical Section Within the Romanian Academy, 010071 Bucharest, Romania
| |
Collapse
|
9
|
Mantry S, Behera A, Pradhan S, Mohanty L, Kumari R, Singh A, Yadav MK. Polysaccharide-based chondroitin sulfate macromolecule loaded hydrogel/scaffolds in wound healing- A comprehensive review on possibilities, research gaps, and safety assessment. Int J Biol Macromol 2024; 279:135410. [PMID: 39245102 DOI: 10.1016/j.ijbiomac.2024.135410] [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: 06/14/2024] [Revised: 08/20/2024] [Accepted: 09/05/2024] [Indexed: 09/10/2024]
Abstract
Wound healing is an intricate multifactorial process that may alter the extent of scarring left by the wound. A substantial portion of the global population is impacted by non-healing wounds, imposing significant financial burdens on the healthcare system. The conventional dosage forms fail to improve the condition, especially in the presence of other morbidities. Thus, there is a pressing requirement for a type of wound dressing that can safeguard the wound site and facilitate skin regeneration, ultimately expediting the healing process. In this context, Chondroitin sulfate (CS), a sulfated glycosaminoglycan material, is capable of hydrating tissues and further promoting the healing. Thus, this comprehensive review article delves into the recent advancement of CS-based hydrogel/scaffolds for wound healing management. The article initially summarizes the various physicochemical characteristics and sources of CS, followed by a brief understanding of the importance of hydrogel and CS in tissue regeneration processes. This is the first instance of such a comprehensive summarization of CS-based hydrogel/scaffolds in wound healing, focusing more on the mechanistic wound healing process, furnishing the recent innovations and toxicity profile. This contemporary review provides a profound acquaintance of strategies for contemporary challenges and future direction in CS-based hydrogel/scaffolds for wound healing.
Collapse
Affiliation(s)
- Shubhrajit Mantry
- Department of Pharmaceutics, Department of Pharmacy, Sarala Birla University, Birla Knowledge City, Ranchi 835103, Jharkhand, India.
| | - Ashutosh Behera
- Department of Pharmaceutical Quality Assurance, Department of Pharmacy, Sarala Birla University, Birla Knowledge City, Ranchi 835103, Jharkhand, India; Department of Pharmaceutical Quality Assurance, Florence College of Pharmacy, IRBA, Ranchi, 835103, Jharkhand, India
| | - Shaktiprasad Pradhan
- Department of Pharmaceutical Chemistry, Koustuv Research Institute of Medical Science (KRIMS), Koustuv Technical Campus, Patia, Bhubaneswar, Odisha 751024, India
| | - Lalatendu Mohanty
- Department of Pharmacology, Department of Pharmaceutical Sciences, HNB Garhwal University (A Central University), Tehri Garhwal, Uttarakhand 24916, India
| | - Ragni Kumari
- School of Pharmacy, LNCT University, Bhopal 462022, Madhya Pradesh, India
| | - Ankita Singh
- Department of Pharmacy, Faculty of Medical Science & Research (FMSR), Sai Nath University, Ranchi, Jharkhand 835219, India
| | - Mahesh Kumar Yadav
- Department of Pharmacy, Faculty of Medical Science & Research (FMSR), Sai Nath University, Ranchi, Jharkhand 835219, India
| |
Collapse
|
10
|
Pande S, Pati F, Chakraborty P. Harnessing Peptide-Based Hydrogels for Enhanced Cartilage Tissue Engineering. ACS APPLIED BIO MATERIALS 2024; 7:5885-5905. [PMID: 39159490 DOI: 10.1021/acsabm.4c00879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/21/2024]
Abstract
Cartilage tissue engineering remains a formidable challenge due to its complex, avascular structure and limited regenerative capacity. Traditional approaches, such as microfracture, autografts, and stem cell delivery, often fail to restore functional tissue adequately. Recently, there has been a surge in the exploration of new materials that mimic the extracellular microenvironment necessary to guide tissue regeneration. This review investigates the potential of peptide-based hydrogels as an innovative solution for cartilage regeneration. These hydrogels, formed via supramolecular self-assembly, exhibit excellent properties, including biocompatibility, ECM mimicry, and controlled biodegradation, making them highly suitable for cartilage tissue engineering. This review explains the structure of cartilage and the principles of supramolecular and peptide hydrogels. It also delves into their specific properties relevant to cartilage regeneration. Additionally, this review presents recent examples and a comparative analysis of various peptide-based hydrogels used for cartilage regeneration. The review also addresses the translational challenges of these materials, highlighting regulatory hurdles and the complexities of clinical application. This comprehensive investigation provides valuable insights for biomedical researchers, tissue engineers, and clinical professionals aiming to enhance cartilage repair methodologies.
Collapse
Affiliation(s)
- Shreya Pande
- Department of Biomedical Engineering, Indian Institute of Technology Hyderabad, Kandi, Sangareddy 502284, Telangana, India
| | - Falguni Pati
- Department of Biomedical Engineering, Indian Institute of Technology Hyderabad, Kandi, Sangareddy 502284, Telangana, India
| | - Priyadarshi Chakraborty
- Department of Chemistry, Indian Institute of Technology Hyderabad, Kandi, Sangareddy 502284, Telangana, India
| |
Collapse
|
11
|
Li MM, Zhang Y, Sun F, Huai MX, Zhang FY, Pan JX, Qu CY, Shen F, Li ZH, Xu LM. Photodynamic Therapy Using RGD-Functionalized Quantum Dots Elicit a Potent Immune Response in a Syngeneic Mouse Model of Pancreatic Cancer. Int J Nanomedicine 2024; 19:9487-9502. [PMID: 39290860 PMCID: PMC11406538 DOI: 10.2147/ijn.s479123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Accepted: 09/05/2024] [Indexed: 09/19/2024] Open
Abstract
Purpose Photodynamic therapy (PDT) induces anti-tumor immune responses by triggering immunogenic cell death in tumor cells. Previously, we demonstrated that novel QDs-RGD nanoparticles exhibited high efficiency as photosensitizers in the treatment of pancreatic cancer. However, the underlying mechanism of the anti-tumor immune effects induced by the photosensitizer remains unknown. This study assessed the anticancer immune effect of QDs-RGD, as well as the conventional photosensitizer chlorine derivative, YLG-1, for comparison, against pancreatic cancer in support of superior therapeutic efficacy. Methods The pancreatic cancer cell line, Panc02, was used for in vitro studies. C57BL/6 mice bearing pancreatic cancer cell-derived xenografts were generated for in vivo studies to assess the anti-tumor effects of QDs-RGD-PDT and YLG-1-PDT. The immunostimulatory ability of both photosensitizers was examined by measuring the expression of damage-associated molecular patterns (DAMP), such as calreticulin (CRT), assessing dendritic cell (DC) maturation, and analyzing cytokine expression. The specific immunity of QDs-RGD and YLG-1-PDT on distant tumor were determined by combining PDT with anti-CTLA-4 antibody. Results QDs-RGD-PDT and YLG-1-PDT significantly inhibited pancreatic cancer cell growth in a dose- and time-dependent manner. While both photosensitizers significantly promoted CRT release, DC maturation, and interferon γ (IFN-γ) and tumor necrosis factor α (TNF-α) expression, QDs-RGD exerted a stronger immunostimulatory effect than YLG-1. Combination treatment with QDs-RGD and CTLA-4 blockade was able to significantly inhibit the growth of distant tumors. Conclusion QDs-RGD is a novel and effective PDT strategy for treating pancreatic tumors by inducing anti-tumor immune responses.
Collapse
Affiliation(s)
- Ming-Ming Li
- Department of Gastroenterology, Xinhua Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, People's Republic of China
| | - Yi Zhang
- Department of Gastroenterology, Xinhua Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, People's Republic of China
| | - Fang Sun
- Department of Gastroenterology, Xinhua Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, People's Republic of China
| | - Man-Xiu Huai
- Department of Gastroenterology, Xinhua Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, People's Republic of China
| | - Fei-Yu Zhang
- Department of Gastroenterology, Xinhua Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, People's Republic of China
| | - Jia-Xing Pan
- Department of Gastroenterology, Xinhua Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, People's Republic of China
| | - Chun-Ying Qu
- Department of Gastroenterology, Xinhua Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, People's Republic of China
| | - Feng Shen
- Department of Gastroenterology, Xinhua Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, People's Republic of China
| | - Zheng-Hong Li
- Department of Gastroenterology, Xinhua Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, People's Republic of China
| | - Lei-Ming Xu
- Department of Gastroenterology, Xinhua Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, People's Republic of China
| |
Collapse
|
12
|
Wawszczak A, Kocki J, Kołodyńska D. Alginate as a Sustainable and Biodegradable Material for Medical and Environmental Applications-The Case Studies. J Biomed Mater Res B Appl Biomater 2024; 112:1-23. [PMID: 39269132 DOI: 10.1002/jbm.b.35475] [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: 12/13/2023] [Revised: 06/19/2024] [Accepted: 08/09/2024] [Indexed: 09/15/2024]
Abstract
Alginates are salts of alginic acid derived mainly from sea algae of the genus brown algae. They are also synthesized by some bacteria. They belong to negatively charged polysaccharides exhibiting some rheological properties. High plasticity and the ability to modify the structure are the reasons for their application in numerous industries. Moreover, when in contact with the living tissue, they do not trigger an immune response, and for this reason they are the most often tested materials for medical applications. The paper discusses the latest applications, including 3D bioprinting, drug delivery systems, and sorptive properties. Recognizing alginates as biomaterials, it emphasizes the necessity for precise processing and modification to industrialize them for specific uses. This review aims to provide a thorough understanding of the advancements in alginate research, underscoring their potential for innovative applications.
Collapse
Affiliation(s)
- Alicja Wawszczak
- Department of Inorganic Chemistry, Faculty of Chemistry, Maria Curie-Skłodowska University, Lublin, Poland
| | - Janusz Kocki
- Department of Clinical Genetics, Medical University of Lublin, Lublin, Poland
| | - Dorota Kołodyńska
- Department of Inorganic Chemistry, Faculty of Chemistry, Maria Curie-Skłodowska University, Lublin, Poland
| |
Collapse
|
13
|
Liu Y, Li L, He M, Xu Y, Wu Z, Xu X, Luo K, Lv H. Self-assembled peptide hydrogel loaded with functional peptide Dentonin accelerates vascularized bone tissue regeneration in critical-size bone defects. Regen Biomater 2024; 11:rbae106. [PMID: 39263324 PMCID: PMC11387769 DOI: 10.1093/rb/rbae106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2024] [Revised: 07/18/2024] [Accepted: 08/18/2024] [Indexed: 09/13/2024] Open
Abstract
Regeneration of oral craniofacial bone defects is a complex process, and reconstruction of large bone defects without the use of exogenous cells or bioactive substances remains a major challenge. Hydrogels are highly hydrophilic polymer networks with the potential to promote bone tissue regeneration. In this study, functional peptide Dentonin was loaded onto self-assembled peptide hydrogels (RAD) to constitute functionally self-assembling peptide RAD/Dentonin hydrogel scaffolds with a view that RAD/Dentonin hydrogel could facilitate vascularized bone regeneration in critical-size calvarial defects. The functionalized peptide RAD/Dentonin forms highly ordered β-sheet supramolecular structures via non-covalent interactions like hydrogen bonding, ultimately assembling into nano-fiber network. RAD/Dentonin hydrogels exhibited desirable porosity and swelling properties, and appropriate biodegradability. RAD/Dentonin hydrogel supported the adhesion, proliferation and three-dimensional migration of bone marrow mesenchymal stem cells (BMSCs) and has the potential to induce differentiation of BMSCs towards osteogenesis through activation of the Wnt/β-catenin pathway. Moreover, RAD/Dentonin hydrogel modulated paracrine secretion of BMSCs and increased the migration, tube formation and angiogenic gene expression of human umbilical vein endothelial cells (HUVECs), which boosted the angiogenic capacity of HUVECs. In vivo, RAD/Dentonin hydrogel significantly strengthened vascularized bone formation in rat calvarial defect. Taken together, these results indicated that the functionalized self-assembling peptide RAD/Dentonin hydrogel effectively enhance osteogenic differentiation of BMSCs, indirectly induce angiogenic effects in HUVECs, and facilitate vascularized bone regeneration in vivo. Thus, it is a promising bioactive material for oral and maxillofacial regeneration.
Collapse
Affiliation(s)
- Yijuan Liu
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key Laboratory of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou 350002, People's Republic of China
- Institute of Stomatology & Laboratory of Oral Tissue Engineering, School and Hospital of Stomatology, Fujian Medical University, Fuzhou 350002, People's Republic of China
| | - Li Li
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key Laboratory of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou 350002, People's Republic of China
- Institute of Stomatology & Laboratory of Oral Tissue Engineering, School and Hospital of Stomatology, Fujian Medical University, Fuzhou 350002, People's Republic of China
| | - Mengjiao He
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key Laboratory of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou 350002, People's Republic of China
- Institute of Stomatology & Laboratory of Oral Tissue Engineering, School and Hospital of Stomatology, Fujian Medical University, Fuzhou 350002, People's Republic of China
| | - Yanmei Xu
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key Laboratory of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou 350002, People's Republic of China
- Institute of Stomatology & Laboratory of Oral Tissue Engineering, School and Hospital of Stomatology, Fujian Medical University, Fuzhou 350002, People's Republic of China
| | - Zekai Wu
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key Laboratory of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou 350002, People's Republic of China
- Institute of Stomatology & Laboratory of Oral Tissue Engineering, School and Hospital of Stomatology, Fujian Medical University, Fuzhou 350002, People's Republic of China
| | - Xiongcheng Xu
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key Laboratory of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou 350002, People's Republic of China
- Institute of Stomatology & Laboratory of Oral Tissue Engineering, School and Hospital of Stomatology, Fujian Medical University, Fuzhou 350002, People's Republic of China
| | - Kai Luo
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key Laboratory of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou 350002, People's Republic of China
- Institute of Stomatology & Laboratory of Oral Tissue Engineering, School and Hospital of Stomatology, Fujian Medical University, Fuzhou 350002, People's Republic of China
| | - Hongbing Lv
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key Laboratory of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou 350002, People's Republic of China
- Institute of Stomatology & Laboratory of Oral Tissue Engineering, School and Hospital of Stomatology, Fujian Medical University, Fuzhou 350002, People's Republic of China
| |
Collapse
|
14
|
Mitić D, Čarkić J, Jaćimović J, Lazarević M, Jakšić Karišik M, Toljić B, Milašin J. The Impact of Nano-Hydroxyapatite Scaffold Enrichment on Bone Regeneration In Vivo-A Systematic Review. Biomimetics (Basel) 2024; 9:386. [PMID: 39056827 PMCID: PMC11274561 DOI: 10.3390/biomimetics9070386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 06/20/2024] [Accepted: 06/22/2024] [Indexed: 07/28/2024] Open
Abstract
OBJECTIVES In order to ensure improved and accelerated bone regeneration, nano-hydroxyapatite scaffolds are often enriched with different bioactive components to further accelerate and improve bone healing. In this review, we critically examined whether the enrichment of nHAp/polymer scaffolds with growth factors, hormones, polypeptides, microRNAs and exosomes improved new bone formation in vivo. MATERIALS AND METHODS Out of 2989 articles obtained from the literature search, 106 papers were read in full, and only 12 articles met the inclusion criteria for this review. RESULTS Several bioactive components were reported to stimulate accelerated bone regeneration in a variety of bone defect models, showing better results than bone grafting with nHAp scaffolds alone. CONCLUSIONS The results indicated that composite materials based on nHAp are excellent candidates as bone substitutes, while nHAp scaffold enrichment further accelerates bone regeneration. The standardization of animal models should be provided in order to clearly define the most significant parameters of in vivo studies. Only in this way can the adequate comparison of findings from different in vivo studies be possible, further advancing our knowledge on bone regeneration and enabling its translation to clinical settings.
Collapse
Affiliation(s)
- Dijana Mitić
- School of Dental Medicine, University of Belgrade, 11000 Belgrade, Serbia; (J.Č.); (J.J.); (M.L.); (M.J.K.); (B.T.); (J.M.)
| | | | | | | | | | | | | |
Collapse
|
15
|
Schuphan J, Stojanović N, Lin YY, Buhl EM, Aveic S, Commandeur U, Schillberg S, Fischer H. A Combination of Flexible Modified Plant Virus Nanoparticles Enables Additive Effects Resulting in Improved Osteogenesis. Adv Healthc Mater 2024; 13:e2304243. [PMID: 38417028 DOI: 10.1002/adhm.202304243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 02/16/2024] [Indexed: 03/01/2024]
Abstract
Plant virus nanoparticles (VNPs) genetically engineered to present osteogenic cues provide a promising method for biofunctionalizing hydrogels in bone tissue engineering. Flexible Potato virus X (PVX) nanoparticles substantially enhance the attachment and differentiation of human mesenchymal stem cells (hMSCs) by presenting the RGD motif, hydroxyapatite-binding peptide (HABP), or consecutive polyglutamates (E8) in a concentration-dependent manner. Therefore, it is hypothesized that Tobacco mosaic virus nanoparticles, which present 1.6 times more functional peptides than PVX, will meliorate such an impact. This study hypothesizes that cultivating hMSCs on a surface coated with a combination of two VNPs presenting peptides for either cell attachment or mineralization can achieve additionally enhancing effects on osteogenesis. Calcium minerals deposited by differentiating hMSCs increases two to threefold for this combination, while the Alkaline Phosphatase activity of hMSCs grown on the PVX-RGD/PVX-HABP-coated surface significantly surpasses any other VNP combination. Superior additive effects are observed for the first time by employing a combination of VNPs with varying functionalities. It is found that the flexible VNP geometry plays a more critical role than the concentration of functional peptides. In conclusion, various peptide-presenting plant VNPs exhibit an additive enhancing effect offering significant potential for effectively functionalizing cell-containing hydrogels in bone tissue engineering.
Collapse
Affiliation(s)
- Juliane Schuphan
- Institute for Molecular Biotechnology, RWTH Aachen University, Worringerweg 1, 52074, Aachen, Germany
| | - Natalija Stojanović
- Department of Dental Materials and Biomaterials Research, RWTH Aachen University Hospital, Pauwelsstrasse 30, 52074, Aachen, Germany
| | - Ying-Ying Lin
- Department of Dental Materials and Biomaterials Research, RWTH Aachen University Hospital, Pauwelsstrasse 30, 52074, Aachen, Germany
| | - Eva Miriam Buhl
- Electron Microscopy Facility, Institute of Pathology, RWTH Aachen University Hospital, Pauwelsstrasse 30, 52074, Aachen, Germany
| | - Sanja Aveic
- Department of Dental Materials and Biomaterials Research, RWTH Aachen University Hospital, Pauwelsstrasse 30, 52074, Aachen, Germany
| | - Ulrich Commandeur
- Institute for Molecular Biotechnology, RWTH Aachen University, Worringerweg 1, 52074, Aachen, Germany
| | - Stefan Schillberg
- Institute for Molecular Biotechnology, RWTH Aachen University, Worringerweg 1, 52074, Aachen, Germany
| | - Horst Fischer
- Department of Dental Materials and Biomaterials Research, RWTH Aachen University Hospital, Pauwelsstrasse 30, 52074, Aachen, Germany
| |
Collapse
|
16
|
Moghaddam A, Bahrami M, Mirzadeh M, Khatami M, Simorgh S, Chimehrad M, Kruppke B, Bagher Z, Mehrabani D, Khonakdar HA. Recent trends in bone tissue engineering: a review of materials, methods, and structures. Biomed Mater 2024; 19:042007. [PMID: 38636500 DOI: 10.1088/1748-605x/ad407d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Accepted: 04/18/2024] [Indexed: 04/20/2024]
Abstract
Bone tissue engineering (BTE) provides the treatment possibility for segmental long bone defects that are currently an orthopedic dilemma. This review explains different strategies, from biological, material, and preparation points of view, such as using different stem cells, ceramics, and metals, and their corresponding properties for BTE applications. In addition, factors such as porosity, surface chemistry, hydrophilicity and degradation behavior that affect scaffold success are introduced. Besides, the most widely used production methods that result in porous materials are discussed. Gene delivery and secretome-based therapies are also introduced as a new generation of therapies. This review outlines the positive results and important limitations remaining in the clinical application of novel BTE materials and methods for segmental defects.
Collapse
Affiliation(s)
| | - Mehran Bahrami
- Department of Mechanical Engineering and Mechanics, Lehigh University, 27 Memorial Dr W, Bethlehem, PA 18015, United States of America
| | | | - Mehrdad Khatami
- Iran Polymer and Petrochemical Institute (IPPI), Tehran 14965-115, Iran
| | - Sara Simorgh
- Department of Tissue Engineering and Regenerative Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mohammadreza Chimehrad
- Department of Mechanical & Aerospace Engineering, College of Engineering & Computer Science, University of Central Florida, Orlando, FL, United States of America
| | - Benjamin Kruppke
- Max Bergmann Center of Biomaterials and Institute of Materials Science, Technische Universität Dresden, 01069 Dresden, Germany
| | - Zohreh Bagher
- Department of Tissue Engineering and Regenerative Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Davood Mehrabani
- Burn and Wound Healing Research Center, Shiraz University of Medical Sciences, Shiraz, Fars 71348-14336, Iran
- Stem Cell Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Fars 71345-1744, Iran
| | - Hossein Ali Khonakdar
- Iran Polymer and Petrochemical Institute (IPPI), Tehran 14965-115, Iran
- Max Bergmann Center of Biomaterials and Institute of Materials Science, Technische Universität Dresden, 01069 Dresden, Germany
| |
Collapse
|
17
|
Guo X, Wang P, Yuwen W, Zhu C, Fu R, Ma P, Duan Z, Fan D. Production and Functional Analysis of Collagen Hexapeptide Repeat Sequences in Pichia pastoris. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024. [PMID: 38801678 DOI: 10.1021/acs.jafc.4c00582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
In the development of biomaterials with specific structural domains associated with various cellular activities, the limited integrin specificity of commonly used adhesion sequences, such as the RGD tripeptide, has resulted in an inability to precisely control cellular responses. To overcome this limitation, we conducted multiple replications of the integrin α2β1-specific ligand, the collagen hexapeptide Gly-Phe-Pro-Gly-Glu-Arg (GFPGER) in Pichia pastoris. This enabled the development of recombinant collagen with high biological activity, which was subsequently expressed, isolated, and purified for structural and functional analysis. The proteins carrying the multiple replications GFPGER sequence demonstrated significant bioactivity in cells, leading to enhanced cell adhesion, osteoblast differentiation, and mineralization when compared to control groups. Importantly, these effects were mediated by integrin α2β1. The new collagen constructed in this study is expected to be a biomaterial for regulating specific cell functions and fates.
Collapse
Affiliation(s)
- Xiaoyan Guo
- Engineering Research Center of Western Resource Innovation Medicine Green Manufacturing, Ministry of Education, School of Chemical Engineering, Northwest University, Xi'an 710069, China
- Shaanxi R&D Center of Biomaterials and Fermentation Engineering, School of Chemical Engineering, Northwest University, Xi'an 710069, China
- Biotech. & Biomed. Research Institute, Northwest University, Xi'an 710069, China
| | - Pan Wang
- Engineering Research Center of Western Resource Innovation Medicine Green Manufacturing, Ministry of Education, School of Chemical Engineering, Northwest University, Xi'an 710069, China
- Shaanxi R&D Center of Biomaterials and Fermentation Engineering, School of Chemical Engineering, Northwest University, Xi'an 710069, China
- Biotech. & Biomed. Research Institute, Northwest University, Xi'an 710069, China
| | - Weigang Yuwen
- Shaanxi Gaint Biotechnology Co., Ltd, Xi'an 710065, Shaanxi, China
| | - Chenhui Zhu
- Engineering Research Center of Western Resource Innovation Medicine Green Manufacturing, Ministry of Education, School of Chemical Engineering, Northwest University, Xi'an 710069, China
- Shaanxi R&D Center of Biomaterials and Fermentation Engineering, School of Chemical Engineering, Northwest University, Xi'an 710069, China
- Biotech. & Biomed. Research Institute, Northwest University, Xi'an 710069, China
| | - Rongzhan Fu
- Engineering Research Center of Western Resource Innovation Medicine Green Manufacturing, Ministry of Education, School of Chemical Engineering, Northwest University, Xi'an 710069, China
- Shaanxi R&D Center of Biomaterials and Fermentation Engineering, School of Chemical Engineering, Northwest University, Xi'an 710069, China
- Biotech. & Biomed. Research Institute, Northwest University, Xi'an 710069, China
| | - Pei Ma
- Engineering Research Center of Western Resource Innovation Medicine Green Manufacturing, Ministry of Education, School of Chemical Engineering, Northwest University, Xi'an 710069, China
- Shaanxi R&D Center of Biomaterials and Fermentation Engineering, School of Chemical Engineering, Northwest University, Xi'an 710069, China
- Biotech. & Biomed. Research Institute, Northwest University, Xi'an 710069, China
| | - Zhiguang Duan
- Engineering Research Center of Western Resource Innovation Medicine Green Manufacturing, Ministry of Education, School of Chemical Engineering, Northwest University, Xi'an 710069, China
- Shaanxi R&D Center of Biomaterials and Fermentation Engineering, School of Chemical Engineering, Northwest University, Xi'an 710069, China
- Biotech. & Biomed. Research Institute, Northwest University, Xi'an 710069, China
| | - Daidi Fan
- Engineering Research Center of Western Resource Innovation Medicine Green Manufacturing, Ministry of Education, School of Chemical Engineering, Northwest University, Xi'an 710069, China
- Shaanxi R&D Center of Biomaterials and Fermentation Engineering, School of Chemical Engineering, Northwest University, Xi'an 710069, China
- Biotech. & Biomed. Research Institute, Northwest University, Xi'an 710069, China
| |
Collapse
|
18
|
Kumar M, Kumar D, Kumar D, Garg Y, Chopra S, Bhatia A. Therapeutic Potential of Nanocarrier Mediated Delivery of Peptides for Wound Healing: Current Status, Challenges and Future Prospective. AAPS PharmSciTech 2024; 25:108. [PMID: 38730090 DOI: 10.1208/s12249-024-02827-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Accepted: 05/01/2024] [Indexed: 05/12/2024] Open
Abstract
Wound healing presents a complex physiological process that involves a sequence of events orchestrated by various cellular and molecular mechanisms. In recent years, there has been growing interest in leveraging nanomaterials and peptides to enhance wound healing outcomes. Nanocarriers offer unique properties such as high surface area-to-volume ratio, tunable physicochemical characteristics, and the ability to deliver therapeutic agents in a controlled manner. Similarly, peptides, with their diverse biological activities and low immunogenicity, hold great promise as therapeutics in wound healing applications. In this review, authors explore the potential of peptides as bioactive components in wound healing formulations, focusing on their antimicrobial, anti-inflammatory, and pro-regenerative properties. Despite the significant progress made in this field, several challenges remain, including the need for standardized characterization methods, optimization of biocompatibility and safety profiles, and translation from bench to bedside. Furthermore, developing multifunctional nanomaterial-peptide hybrid systems represents promising avenues for future research. Overall, the integration of nanomaterials made up of natural or synthetic polymers with peptide-based formulations holds tremendous therapeutic potential in advancing the field of wound healing and improving clinical outcomes for patients with acute and chronic wounds.
Collapse
Affiliation(s)
- Mohit Kumar
- Department of Pharmaceutical Sciences and Technology, Maharaja Ranjit Singh Punjab Technical University (MRSPTU), Bathinda, 151001, Punjab, India
| | - Dikshant Kumar
- Department of Pharmaceutical Sciences and Technology, Maharaja Ranjit Singh Punjab Technical University (MRSPTU), Bathinda, 151001, Punjab, India
| | - Devesh Kumar
- Department of Pharmaceutical Sciences and Technology, Maharaja Ranjit Singh Punjab Technical University (MRSPTU), Bathinda, 151001, Punjab, India
| | - Yogesh Garg
- Department of Pharmaceutical Sciences and Technology, Maharaja Ranjit Singh Punjab Technical University (MRSPTU), Bathinda, 151001, Punjab, India
| | - Shruti Chopra
- Department of Pharmaceutical Sciences and Technology, Maharaja Ranjit Singh Punjab Technical University (MRSPTU), Bathinda, 151001, Punjab, India
| | - Amit Bhatia
- Department of Pharmaceutical Sciences and Technology, Maharaja Ranjit Singh Punjab Technical University (MRSPTU), Bathinda, 151001, Punjab, India.
| |
Collapse
|
19
|
Ribeiro M, Simões M, Vitorino C, Mascarenhas-Melo F. Hydrogels in Cutaneous Wound Healing: Insights into Characterization, Properties, Formulation and Therapeutic Potential. Gels 2024; 10:188. [PMID: 38534606 DOI: 10.3390/gels10030188] [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: 02/01/2024] [Revised: 02/26/2024] [Accepted: 03/05/2024] [Indexed: 03/28/2024] Open
Abstract
Hydrogels are polymeric materials that possess a set of characteristics meeting various requirements of an ideal wound dressing, making them promising for wound care. These features include, among others, the ability to absorb and retain large amounts of water and the capacity to closely mimic native structures, such as the extracellular matrix, facilitating various cellular processes like proliferation and differentiation. The polymers used in hydrogel formulations exhibit a broad spectrum of properties, allowing them to be classified into two main categories: natural polymers like collagen and chitosan, and synthetic polymers such as polyurethane and polyethylene glycol. This review offers a comprehensive overview and critical analysis of the key polymers that can constitute hydrogels, beginning with a brief contextualization of the polymers. It delves into their function, origin, and chemical structure, highlighting key sources of extraction and obtaining. Additionally, this review encompasses the main intrinsic properties of these polymers and their roles in the wound healing process, accompanied, whenever available, by explanations of the underlying mechanisms of action. It also addresses limitations and describes some studies on the effectiveness of isolated polymers in promoting skin regeneration and wound healing. Subsequently, we briefly discuss some application strategies of hydrogels derived from their intrinsic potential to promote the wound healing process. This can be achieved due to their role in the stimulation of angiogenesis, for example, or through the incorporation of substances like growth factors or drugs, such as antimicrobials, imparting new properties to the hydrogels. In addition to substance incorporation, the potential of hydrogels is also related to their ability to serve as a three-dimensional matrix for cell culture, whether it involves loading cells into the hydrogel or recruiting cells to the wound site, where they proliferate on the scaffold to form new tissue. The latter strategy presupposes the incorporation of biosensors into the hydrogel for real-time monitoring of wound conditions, such as temperature and pH. Future prospects are then ultimately addressed. As far as we are aware, this manuscript represents the first comprehensive approach that brings together and critically analyzes fundamental aspects of both natural and synthetic polymers constituting hydrogels in the context of cutaneous wound healing. It will serve as a foundational point for future studies, aiming to contribute to the development of an effective and environmentally friendly dressing for wounds.
Collapse
Affiliation(s)
- Mariana Ribeiro
- Faculty of Pharmacy, University of Coimbra, Pólo das Ciências da Saúde, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal
- CISUC-Center for Informatics and Systems, University of Coimbra, Pinhal de Marrocos, 3030-290 Coimbra, Portugal
- Coimbra Chemistry Centre, Institute of Molecular Sciences-IMS, Department of Chemistry, University of Coimbra, 3000-535 Coimbra, Portugal
| | - Marco Simões
- CISUC-Center for Informatics and Systems, University of Coimbra, Pinhal de Marrocos, 3030-290 Coimbra, Portugal
- CIBIT-Coimbra Institute for Biomedical Imaging and Translational Research, University of Coimbra, Pólo das Ciências da Saúde, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal
| | - Carla Vitorino
- Faculty of Pharmacy, University of Coimbra, Pólo das Ciências da Saúde, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal
- Coimbra Chemistry Centre, Institute of Molecular Sciences-IMS, Department of Chemistry, University of Coimbra, 3000-535 Coimbra, Portugal
- CIBIT-Coimbra Institute for Biomedical Imaging and Translational Research, University of Coimbra, Pólo das Ciências da Saúde, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal
| | - Filipa Mascarenhas-Melo
- Higher School of Health, Polytechnic Institute of Guarda, Rua da Cadeia, 6300-307 Guarda, Portugal
- REQUIMTE/LAQV, Department of Pharmaceutical Technology, Faculty of Pharmacy, University of Coimbra, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal
| |
Collapse
|
20
|
Javid H, Oryani MA, Rezagholinejad N, Esparham A, Tajaldini M, Karimi‐Shahri M. RGD peptide in cancer targeting: Benefits, challenges, solutions, and possible integrin-RGD interactions. Cancer Med 2024; 13:e6800. [PMID: 38349028 PMCID: PMC10832341 DOI: 10.1002/cam4.6800] [Citation(s) in RCA: 31] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 11/06/2023] [Accepted: 11/27/2023] [Indexed: 02/15/2024] Open
Abstract
RGD peptide can be found in cell adhesion and signaling proteins, such as fibronectin, vitronectin, and fibrinogen. RGD peptides' principal function is to facilitate cell adhesion by interacting with integrin receptors on the cell surface. They have been intensively researched for use in biotechnology and medicine, including incorporation into biomaterials, conjugation to medicinal molecules or nanoparticles, and labeling with imaging agents. RGD peptides can be utilized to specifically target cancer cells and the tumor vasculature by engaging with these integrins, improving drug delivery efficiency and minimizing adverse effects on healthy tissues. RGD-functionalized drug carriers are a viable option for cancer therapy as this focused approach has demonstrated promise in the future. Writing a review on the RGD peptide can significantly influence how drugs are developed in the future by improving our understanding of the peptide, finding knowledge gaps, fostering innovation, and making drug design easier.
Collapse
Affiliation(s)
- Hossein Javid
- Department of Medical Laboratory SciencesVarastegan Institute for Medical SciencesMashhadIran
- Department of Clinical Biochemistry, Faculty of MedicineMashhad University of Medical SciencesMashhadIran
- Surgical Oncology Research CenterMashhad University of Medical SciencesMashhadIran
| | - Mahsa Akbari Oryani
- Department of Pathology, School of MedicineMashhad University of Medical SciencesMashhadIran
| | | | - Ali Esparham
- Student Research Committee, Faculty of MedicineMashhad University of Medical SciencesMashhadIran
| | - Mahboubeh Tajaldini
- Ischemic Disorder Research CenterGolestan University of Medical SciencesGorganIran
| | - Mehdi Karimi‐Shahri
- Department of Pathology, School of MedicineMashhad University of Medical SciencesMashhadIran
- Department of Pathology, School of MedicineGonabad University of Medical SciencesGonabadIran
| |
Collapse
|
21
|
Agapova OI, Ostrovsky DS, Khubetsova MK, Kerimov TZ, Borzenok SA, Bogush VG, Davydova LI, Cheperegin SE, Efimov AE, Agapov II, Debabov VG. Hydrogels Based on Recombinant Spidroin Stimulate Proliferation and Migration of Human Corneal Cells. DOKLADY BIOLOGICAL SCIENCES : PROCEEDINGS OF THE ACADEMY OF SCIENCES OF THE USSR, BIOLOGICAL SCIENCES SECTIONS 2023; 513:S41-S44. [PMID: 38472685 DOI: 10.1134/s0012496623600173] [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: 11/09/2023] [Revised: 11/20/2023] [Accepted: 11/25/2023] [Indexed: 03/14/2024]
Abstract
The effect of recombinant spidroin (RS) hydrogel (HG) on anterior epithelial cells and keratocytes of the human cornea was studied in vitro. Corneal injuries are highly prevalent in developing countries according to the World Health Organization. Various technologies have recently been proposed to restore the damaged surface of the cornea. Use of biodegradable silk-based materials, including recombinant analogs of the spider silk protein spidroin, is an important avenue of research in the field of wound healing and corneal regeneration. Spidroins are well known for their optimal balance of strength and elasticity. Given their biological compatibility, lack of immunogenicity, and biodegradability, spidroins provide a biomaterial for tissue engineering and regenerative medicine. HGs based on RS rS2/12-RGDS were therefore tested for cytotoxicity toward isolated corneal epithelial cells and keratocytes with regard to possible changes in cell phenotype and migratory activity. A promising outlook and therapeutic potential were demonstrated for RS-based HGs.
Collapse
Affiliation(s)
- O I Agapova
- Academician Shumakov National Medical Research Center of Transplantology and Artificial Organs, Ministry of Health of the Russian Federation, Moscow, Russia
| | - D S Ostrovsky
- Fyodorov Eye Microsurgery Complex, Ministry of Health of the Russian Federation, Moscow, Russia
| | - M Kh Khubetsova
- Fyodorov Eye Microsurgery Complex, Ministry of Health of the Russian Federation, Moscow, Russia
| | - T Z Kerimov
- Fyodorov Eye Microsurgery Complex, Ministry of Health of the Russian Federation, Moscow, Russia
- Evdokimov Moscow State University of Medicine and Dentistry, Ministry of Health of the Russian Federation, Moscow, Russia
| | - S A Borzenok
- Fyodorov Eye Microsurgery Complex, Ministry of Health of the Russian Federation, Moscow, Russia
- Evdokimov Moscow State University of Medicine and Dentistry, Ministry of Health of the Russian Federation, Moscow, Russia
| | - V G Bogush
- Kurchatov Institute National Research Center, Moscow, Russia
| | - L I Davydova
- Kurchatov Institute National Research Center, Moscow, Russia
| | - S E Cheperegin
- Kurchatov Institute National Research Center, Moscow, Russia
| | - A E Efimov
- Academician Shumakov National Medical Research Center of Transplantology and Artificial Organs, Ministry of Health of the Russian Federation, Moscow, Russia
| | - I I Agapov
- Academician Shumakov National Medical Research Center of Transplantology and Artificial Organs, Ministry of Health of the Russian Federation, Moscow, Russia.
| | - V G Debabov
- Kurchatov Institute National Research Center, Moscow, Russia
| |
Collapse
|
22
|
Hansda B, Mondal B, Hazra S, Das KS, Castelletto V, Hamley IW, Banerjee A. Effect of molar ratio and concentration on the rheological properties of two-component supramolecular hydrogels: tuning of the morphological and drug releasing behaviour. SOFT MATTER 2023; 19:8264-8273. [PMID: 37869972 DOI: 10.1039/d3sm00883e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/24/2023]
Abstract
Self-assembled supramolecular hydrogels offer great potential as biomaterials and drug delivery systems. Specifically, peptide-based multicomponent hydrogels are promising materials due to their advantage that their mechanical and physical properties can be tuned to enhance their functionalities and broaden their applications. Herein, we report two-component assembly and formation of hydrogels containing inexpensive complementary anionic, BUVV-OH (A), and cationic, KFFC12 (B), peptide amphiphiles. Individually, neither of these components formed a hydrogel, while mixtures with compositions 1 : 1, 1 : 2, and 2 : 1 (molar ratio) as A : B show hydrogel formation (Milli-Q water, at pH = 6.79). These hydrogels displayed a good shear-thinning behaviour with different mechanical stabilities and nano-fibrous network structures. The 1 : 1 hydrogel shows good cell viability for human embryonic kidney (HEK-293) cells and CHO cells indicating its non-cytotoxicity. The biocompatible, thixotropic 1 : 1 hydrogel with a nanofiber network structure shows the highest mechanical strength with a storage modulus of 3.4 × 103 Pa. The hydrogel is able to encapsulate drugs including antibiotics amoxicillin and rifampicin, and anticancer drug doxorubicin, and it exhibits sustainable release of 76%, 70%, and 81% respectively in vitro after 3 days. The other two mixtures (composition 1 : 2 and 2 : 1) are unable to form a hydrogel when they are loaded with these drugs. Interestingly, it is noticed that with an increase in concentration, the mechanical strength of a 1 : 1 hydrogel is significantly enhanced, showing potential that may act as a scaffold for tissue engineering. The two-component gel offers tunable mechanical properties, thixotropy, injectability, and biocompatibility and has great potential as a scaffold for sustained drug release and tissue engineering.
Collapse
Affiliation(s)
- Biswanath Hansda
- School of Biological Sciences, Indian Association for the Cultivation of Science, 2A & 2B Raja S. C. Mullick Road, Jadavpur, Kolkata-700032, India.
| | - Biplab Mondal
- School of Biological Sciences, Indian Association for the Cultivation of Science, 2A & 2B Raja S. C. Mullick Road, Jadavpur, Kolkata-700032, India.
| | - Soumyajit Hazra
- School of Biological Sciences, Indian Association for the Cultivation of Science, 2A & 2B Raja S. C. Mullick Road, Jadavpur, Kolkata-700032, India.
| | - Krishna Sundar Das
- School of Chemical Sciences, Indian Association for the Cultivation of Science, 2A & 2B Raja S. C. Mullick Road, Jadavpur, Kolkata-700032, India
| | | | - Ian W Hamley
- Department of Chemistry, University of Reading, Reading RG6 6AD, UK
| | - Arindam Banerjee
- School of Biological Sciences, Indian Association for the Cultivation of Science, 2A & 2B Raja S. C. Mullick Road, Jadavpur, Kolkata-700032, India.
| |
Collapse
|
23
|
Zengin A, Teixeira FC, Feliciano T, Habibovic P, Mota CD, Baker MB, van Rijt S. Matrix metalloproteinase degradable, in situ photocrosslinked nanocomposite bioinks for bioprinting applications. BIOMATERIALS ADVANCES 2023; 154:213647. [PMID: 37839298 DOI: 10.1016/j.bioadv.2023.213647] [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: 02/16/2023] [Revised: 08/10/2023] [Accepted: 09/29/2023] [Indexed: 10/17/2023]
Abstract
The development of suitable bioinks with high printability, mechanical strength, biodegradability, and biocompatibility is a key challenge for the clinical translation of 3D constructs produced with bioprinting technologies. In this work, we developed a new type of nanocomposite bioinks containing thiolated mesoporous silica nanoparticles (MSN) that act as active fillers within norbornene-functionalized hydrogels. The MSNs could rapidly covalently crosslink the hydrogels upon exposure to UV light. The mechanical properties of the gels could be modulated from 9.3 to 19.7 kPa with increasing concentrations of MSN. The ability of the MSN to covalently crosslink polymeric networks was, however, significantly influenced by polymer architecture and the number of functional groups. Modification of the outer surface of MSNs with matrix metalloproteinase (MMP) sensitive peptides (MSN-MMPs) resulted in proteinase K and MMP-9 enzyme responsive biodegradable bioinks. Additional cysteine modified RGD peptide incorporation enhanced cell-matrix interactions and reduced the gelation time for bioprinting. The nanocomposite bioinks could be printed by using extrusion-based bioprinting. Our nanocomposite bioinks preserved their shape during in vitro studies and encapsulated MG63 cells preserved their viability and proliferated within the bioinks. As such, our nanocomposite bioinks are promising bioinks for creating bioprinted constructs with tunable mechanical and degradation properties.
Collapse
Affiliation(s)
- Aygul Zengin
- Department of Instructive Biomaterials Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, P.O. Box 616, 6200 MD Maastricht, the Netherlands
| | - Filipa Castro Teixeira
- Department of Complex Tissue Regeneration, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, P.O. Box 616, 6200 MD Maastricht, the Netherlands
| | - Tony Feliciano
- Department of Complex Tissue Regeneration, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, P.O. Box 616, 6200 MD Maastricht, the Netherlands
| | - Pamela Habibovic
- Department of Instructive Biomaterials Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, P.O. Box 616, 6200 MD Maastricht, the Netherlands
| | - Carlos Domingues Mota
- Department of Complex Tissue Regeneration, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, P.O. Box 616, 6200 MD Maastricht, the Netherlands
| | - Matthew B Baker
- Department of Complex Tissue Regeneration, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, P.O. Box 616, 6200 MD Maastricht, the Netherlands
| | - Sabine van Rijt
- Department of Instructive Biomaterials Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, P.O. Box 616, 6200 MD Maastricht, the Netherlands.
| |
Collapse
|
24
|
Snyder Y, Jana S. Strategies for Development of Synthetic Heart Valve Tissue Engineering Scaffolds. PROGRESS IN MATERIALS SCIENCE 2023; 139:101173. [PMID: 37981978 PMCID: PMC10655624 DOI: 10.1016/j.pmatsci.2023.101173] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2023]
Abstract
The current clinical solutions, including mechanical and bioprosthetic valves for valvular heart diseases, are plagued by coagulation, calcification, nondurability, and the inability to grow with patients. The tissue engineering approach attempts to resolve these shortcomings by producing heart valve scaffolds that may deliver patients a life-long solution. Heart valve scaffolds serve as a three-dimensional support structure made of biocompatible materials that provide adequate porosity for cell infiltration, and nutrient and waste transport, sponsor cell adhesion, proliferation, and differentiation, and allow for extracellular matrix production that together contributes to the generation of functional neotissue. The foundation of successful heart valve tissue engineering is replicating native heart valve architecture, mechanics, and cellular attributes through appropriate biomaterials and scaffold designs. This article reviews biomaterials, the fabrication of heart valve scaffolds, and their in-vitro and in-vivo evaluations applied for heart valve tissue engineering.
Collapse
Affiliation(s)
- Yuriy Snyder
- Department of Bioengineering, University of Missouri, Columbia, MO 65211, USA
| | - Soumen Jana
- Department of Bioengineering, University of Missouri, Columbia, MO 65211, USA
| |
Collapse
|
25
|
Pham HTM, Nguyen DL, Kim HS, Yang EK, Kim JH, Yoon HC, Park HJ. A novel and cost-effective method for high-throughput 3D culturing and rhythmic assessment of hiPSC-derived cardiomyocytes using retroreflective Janus microparticles. Biomater Res 2023; 27:79. [PMID: 37587478 PMCID: PMC10428620 DOI: 10.1186/s40824-023-00416-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Accepted: 07/21/2023] [Indexed: 08/18/2023] Open
Abstract
BACKGROUND Human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) gain attention as a potent cell source in regenerative medicine and drug discovery. With the necessity of the demands for experimental models to create a more physiologically relevant model of the heart in vitro we herein investigate a 3D culturing platform and a method for assessing rhythm in hiPSC-CMs. METHODS The 3D cell culture PAMCELL™ plate is designed to enable cells to attach exclusively to adhesive patterned areas. These cell adhesive zones, named as micro-patterned pads, feature micron silica beads that are surface-modified with the well-known arginyl-glycyl-aspartic acid (RGD) peptide. RGD binding to the surface of hiPSC-CMs facilitates cell-cell attachment and the formation of uniform-size spheroids, which is controlled by the diameter of the micro-patterned pads. The assessment and evaluation of 3D hiPSC-CMs beating pattern are carried out using reflective properties of retroreflective Janus micro-particle (RJP). These RJPs are modified with an antibody targeting the gap junction protein found on the surface of hiPSC-CM spheroids. The signal assessment system comprises a camera attached to an optical microscope and a white light source. RESULTS The 3D PAMCELL™ R100 culture plate efficiently generate approximately 350 uniform-sized hiPSC-CM spheroids in each well of a 96-well plate and supported a 20-day culture. Analysis of genes and protein expression levels reveal that iPSC-CM spheroids grown on PAMCELL™ R100 retain cardiac stem cell characteristics and functions, outperforming traditional 2D culture platform. Additionally, the RJPs enable monitoring and evaluation of in vitro beating properties of cardiomyocytes without using complex monitoring setup. The system demonstrates its capability to identify alteration in the rhythmic activity of cardiac cells when exposed to ion channel blockers, nifedipine and E4031. CONCLUSIONS The integration of the 3D culture method and RJPs in this study establishes a platform for evaluating the rhythmic properties of 3D hiPSC-CMs. This approach holds significant potential for identifying arrhythmias or other cardiac abnormalities, ultimately contributing to the development of more effective therapies for heart diseases.
Collapse
Affiliation(s)
- Huyen T M Pham
- Department of Molecular Science and Technology, Ajou University, Suwon, 16499, South Korea
| | - Duc Long Nguyen
- Department of Molecular Science and Technology, Ajou University, Suwon, 16499, South Korea
| | - Hyo-Sop Kim
- Department of Molecular Science and Technology, Ajou University, Suwon, 16499, South Korea
| | - Eun Kyeong Yang
- Department of Molecular Science and Technology, Ajou University, Suwon, 16499, South Korea
| | - Jae-Ho Kim
- Department of Molecular Science and Technology, Ajou University, Suwon, 16499, South Korea.
| | - Hyun C Yoon
- Department of Molecular Science and Technology, Ajou University, Suwon, 16499, South Korea.
| | - Hyun-Ji Park
- Department of Molecular Science and Technology, Ajou University, Suwon, 16499, South Korea.
| |
Collapse
|
26
|
Long Z, Bing T, Zhang X, Sheng J, Zu S, Li W, Liu X, Zhang N, Shangguan D. Structural Optimization and Interaction Study of a DNA Aptamer to L1 Cell Adhesion Molecule. Int J Mol Sci 2023; 24:ijms24108612. [PMID: 37239955 DOI: 10.3390/ijms24108612] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 04/21/2023] [Accepted: 04/25/2023] [Indexed: 05/28/2023] Open
Abstract
The L1 cell adhesion molecule (L1CAM) plays important roles in the development and plasticity of the nervous system as well as in tumor formation, progression, and metastasis. New ligands are necessary tools for biomedical research and the detection of L1CAM. Here, DNA aptamer yly12 against L1CAM was optimized to have much stronger binding affinity (10-24 fold) at room temperature and 37 °C via sequence mutation and extension. This interaction study revealed that the optimized aptamers (yly20 and yly21) adopted a hairpin structure containing two loops and two stems. The key nucleotides for aptamer binding mainly located in loop I and its adjacent area. Stem I mainly played the role of stabilizing the binding structure. The yly-series aptamers were demonstrated to bind the Ig6 domain of L1CAM. This study reveals a detailed molecular mechanism for the interaction between yly-series aptamers and L1CAM and provides guidance for drug development and detection probe design against L1CAM.
Collapse
Affiliation(s)
- Zhenhao Long
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Analytical Chemistry for Living Biosystems, CAS Research and Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China
- School of Chemical Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Tao Bing
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Analytical Chemistry for Living Biosystems, CAS Research and Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China
- Zhejiang Cancer Hospital, The Key Laboratory of Zhejiang Province for Aptamers and Theranostics, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310022, China
| | - Xiangru Zhang
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Analytical Chemistry for Living Biosystems, CAS Research and Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China
- School of Chemical Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jing Sheng
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Analytical Chemistry for Living Biosystems, CAS Research and Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China
- School of Chemical Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Shuang Zu
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Analytical Chemistry for Living Biosystems, CAS Research and Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China
- School of Molecular Medicine, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310013, China
| | - Weiwei Li
- Zhejiang Cancer Hospital, The Key Laboratory of Zhejiang Province for Aptamers and Theranostics, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310022, China
- School of Molecular Medicine, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310013, China
| | - Xiangjun Liu
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Analytical Chemistry for Living Biosystems, CAS Research and Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China
- School of Chemical Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Nan Zhang
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Analytical Chemistry for Living Biosystems, CAS Research and Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China
| | - Dihua Shangguan
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Analytical Chemistry for Living Biosystems, CAS Research and Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China
- School of Chemical Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
- School of Molecular Medicine, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310013, China
| |
Collapse
|
27
|
Mansur AAP, Rodrigues MA, Capanema NSV, Carvalho SM, Gomes DA, Mansur HS. Functionalized bioadhesion-enhanced carboxymethyl cellulose/polyvinyl alcohol hybrid hydrogels for chronic wound dressing applications. RSC Adv 2023; 13:13156-13168. [PMID: 37124005 PMCID: PMC10140670 DOI: 10.1039/d3ra01519j] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Accepted: 04/21/2023] [Indexed: 05/02/2023] Open
Abstract
Wounds produced by trauma, burns, and chronic diseases cause millions of patients to suffer discomfort, pain, and, in many cases, disability and death, leading to enormous health, social and financial impacts globally. Regrettably, current clinical treatments for chronic wounds remain unsatisfactory. Thus, this study reports for the first time the design, development, and synthesis of chemically biofunctionalized hybrid hydrogels made of carboxymethyl cellulose (CMC) and poly(vinyl alcohol) (PVA) crosslinked by citric acid using an entirely biocompatible and green process. They demonstrated suitable physicochemical properties, cytocompatibility, and hemocompatibility to be applied as a smart wound dressing for skin tissue engineering. These novel hybrids were biofunctionalized with l-arginine and RGD peptide through carbodiimide mediated-amide formation to promote bioadhesion of fibroblast and keratinocyte cells as a potential enhancement for wound healing and skin tissue engineering applications.
Collapse
Affiliation(s)
- A A P Mansur
- Department of Metallurgical and Materials Engineering, Center of Nanoscience, Nanotechnology and Innovation - CeNano2I, Federal University of Minas Gerais Av. Antônio Carlos 6627 - Escola de Engenharia, Bloco 2 - Sala 2233, 31.270-901 Belo Horizonte MG Brazil +55-31-34091843
| | - M A Rodrigues
- Department of Biochemistry and Immunology, Federal University of Minas Gerais Brazil
| | - N S V Capanema
- Department of Metallurgical and Materials Engineering, Center of Nanoscience, Nanotechnology and Innovation - CeNano2I, Federal University of Minas Gerais Av. Antônio Carlos 6627 - Escola de Engenharia, Bloco 2 - Sala 2233, 31.270-901 Belo Horizonte MG Brazil +55-31-34091843
| | - S M Carvalho
- Department of Metallurgical and Materials Engineering, Center of Nanoscience, Nanotechnology and Innovation - CeNano2I, Federal University of Minas Gerais Av. Antônio Carlos 6627 - Escola de Engenharia, Bloco 2 - Sala 2233, 31.270-901 Belo Horizonte MG Brazil +55-31-34091843
| | - D A Gomes
- Department of Biochemistry and Immunology, Federal University of Minas Gerais Brazil
| | - H S Mansur
- Department of Metallurgical and Materials Engineering, Center of Nanoscience, Nanotechnology and Innovation - CeNano2I, Federal University of Minas Gerais Av. Antônio Carlos 6627 - Escola de Engenharia, Bloco 2 - Sala 2233, 31.270-901 Belo Horizonte MG Brazil +55-31-34091843
| |
Collapse
|
28
|
Utilization of Functionalized Metal–Organic Framework Nanoparticle as Targeted Drug Delivery System for Cancer Therapy. Pharmaceutics 2023; 15:pharmaceutics15030931. [PMID: 36986793 PMCID: PMC10051794 DOI: 10.3390/pharmaceutics15030931] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 03/09/2023] [Accepted: 03/12/2023] [Indexed: 03/16/2023] Open
Abstract
Cancer is a multifaceted disease that results from the complex interaction between genetic and environmental factors. Cancer is a mortal disease with the biggest clinical, societal, and economic burden. Research on better methods of the detection, diagnosis, and treatment of cancer is crucial. Recent advancements in material science have led to the development of metal–organic frameworks, also known as MOFs. MOFs have recently been established as promising and adaptable delivery platforms and target vehicles for cancer therapy. These MOFs have been constructed in a fashion that offers them the capability of drug release that is stimuli-responsive. This feature has the potential to be exploited for cancer therapy that is externally led. This review presents an in-depth summary of the research that has been conducted to date in the field of MOF-based nanoplatforms for cancer therapeutics.
Collapse
|
29
|
Stimuli-Responsive Polysaccharide Hydrogels and Their Composites for Wound Healing Applications. Polymers (Basel) 2023; 15:polym15040986. [PMID: 36850269 PMCID: PMC9958605 DOI: 10.3390/polym15040986] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 02/08/2023] [Accepted: 02/10/2023] [Indexed: 02/18/2023] Open
Abstract
There is a growing concern about wound care, since traditional dressings such as bandages and sutures can no longer meet existing needs. To address the demanding requirements, naturally occurring polymers have been extensively exploited for use in modern wound management. Polysaccharides, being the most abundant biopolymers, have some distinct characteristics, including biocompatibility and biodegradability, which render them ideal candidates for wound healing applications. Combining them with inorganic and organic moieties can produce effective multifunctional composites with the desired mechanical properties, high wound healing efficiencies and excellent antibacterial behavior. Recent research endeavors focus on the development of stimuli-responsive polysaccharide composites for biomedical applications. Polysaccharide composites, being sensitive to the local environment, such as changes of the solution temperature, pH, etc., can sense and react to the wound conditions, thus promoting an effective interaction with the wound. This review highlights the recent advances in stimuli-responsive polysaccharide hydrogels and their composites for use in wound healing applications. The synthetic approaches, physical, chemical, and biochemical properties as well as their function in wound healing will be discussed.
Collapse
|