1
|
Roshanbinfar K, Evans AD, Samanta S, Kolesnik-Gray M, Fiedler M, Krstic V, Engel FB, Oommen OP. Enhancing biofabrication: Shrink-resistant collagen-hyaluronan composite hydrogel for tissue engineering and 3D bioprinting applications. Biomaterials 2025; 318:123174. [PMID: 39951830 DOI: 10.1016/j.biomaterials.2025.123174] [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: 09/12/2024] [Revised: 01/21/2025] [Accepted: 02/05/2025] [Indexed: 02/16/2025]
Abstract
Biofabrication represents a promising technique for creating tissues for regeneration or as models for drug testing. Collagen-based hydrogels are widely used as suitable matrix owing to their biocompatibility and tunable mechanical properties. However, one major challenge is that the encapsulated cells interact with the collagen matrix causing construct shrinkage. Here, we present a hydrogel with high shape fidelity, mimicking the major components of the extracellular matrix. We engineered a composite hydrogel comprising gallic acid (GA)-functionalized hyaluronic acid (HA), collagen I, and HA-coated multiwall carbon nanotubes (MWCNT). This hydrogel supports cell encapsulation, exhibits shear-thinning properties enhancing injectability and printability, and importantly significantly mitigates shrinkage when loaded with human fibroblasts compared to collagen I hydrogels (∼20 % vs. > 90 %). 3D-bioprinted rings utilizing human fibroblast-loaded inks maintain their shape over 7 days in culture. Furthermore, inclusion of HAGA into collagen I hydrogels increases mechanical stiffness, radical scavenging capability, and tissue adhesiveness. Notably, the here developed hydrogel is also suitable for human induced pluripotent stem cell-derived cardiomyocytes and allows printing of functional heart ventricles responsive to pharmacological treatment. Cardiomyocytes behave similar in the newly developed hydrogels compared to collagen I, based on survival, sarcomere appearance, and calcium handling. Collectively, we developed a novel material to overcome the challenge of post-fabrication matrix shrinkage conferring high shape fidelity.
Collapse
Affiliation(s)
- Kaveh Roshanbinfar
- Experimental Renal and Cardiovascular Research, Department of Nephropathology, Institute of Pathology and Department of Cardiology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054, Erlangen, Germany
| | - Austin Donnelly Evans
- Bioengineering and Nanomedicine Group, Faculty of Medicine and Health Technologies, Tampere University, 33720, Tampere, Finland
| | - Sumanta Samanta
- Bioengineering and Nanomedicine Group, Faculty of Medicine and Health Technologies, Tampere University, 33720, Tampere, Finland
| | - Maria Kolesnik-Gray
- Department of Physics, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Staudtstr. 7, 91058, Erlangen, Germany
| | - Maren Fiedler
- Experimental Renal and Cardiovascular Research, Department of Nephropathology, Institute of Pathology and Department of Cardiology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054, Erlangen, Germany
| | - Vojislav Krstic
- Department of Physics, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Staudtstr. 7, 91058, Erlangen, Germany; Department of Physics, Wake-Forest-University, Winston Salem, NC, 27109, USA
| | - Felix B Engel
- Experimental Renal and Cardiovascular Research, Department of Nephropathology, Institute of Pathology and Department of Cardiology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054, Erlangen, Germany.
| | - Oommen P Oommen
- School of Pharmacy and Pharmaceutical Sciences, Cardiff University, King Edward VII Avenue, Cardiff, CF10 3NB, UK.
| |
Collapse
|
2
|
Suzuki R, Ide K, Tokuchi S, Kawano T, Ntege EH, Murahashi M, Isa M, Sunami H, Shimizu Y, Nakamura H. Selective effects of collagen-derived peptides Pro-Hyp and Hyp-Gly on the proliferation and differentiation of SSEA3-positive human dental pulp stem cells. Regen Ther 2025; 29:162-170. [PMID: 40170801 PMCID: PMC11960528 DOI: 10.1016/j.reth.2025.03.003] [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: 01/24/2025] [Revised: 02/27/2025] [Accepted: 03/09/2025] [Indexed: 04/03/2025] Open
Abstract
Introduction Collagen-derived peptides demonstrate diverse biological activities, but their effects on human dental pulp stem cells (hDPSCs) remain unclear. This study investigated the influence of prolyl-hydroxyproline (Pro-Hyp) and hydroxyproline-glycine (Hyp-Gly) on stage-specific embryonic antigen 3 (SSEA3)-positive hDPSCs. Methods SSEA3-positive and SSEA3-negative hDPSCs were isolated and cultured with Pro-Hyp or Hyp-Gly (200 μg/ml) under stem cell medium or differentiation-inducing conditions. Cell proliferation was assessed using the MTT assay. The expression of the chondrogenic and osteogenic markers was analyzed by real-time PCR. Matrix production was evaluated using Alcian blue and Alizarin red S staining. Results Pro-Hyp and Hyp-Gly enhanced the proliferation of SSEA3-positive hDPSCs compared with SSEA3-negative cells. Pro-Hyp promoted chondrogenic differentiation through early upregulation of Col II followed by sustained SOX9 expression, with enhanced matrix production after 2 weeks in chondrogenic medium. Hyp-Gly specifically enhanced the osteogenic differentiation of SSEA3-positive cells through the temporal regulation of gene expression, progressing from early transcription factors (RUNX2 and RANKL) to matrix proteins (ALPL, Col I, BSP, and OCN), resulting in increased mineralization under osteogenic conditions. Conclusions This study demonstrated that Pro-Hyp and Hyp-Gly selectively influenced SSEA3-positive hDPSC fate through the distinct temporal regulation of differentiation pathways. These findings provide new insights into the development of targeted regenerative strategies using collagen-derived peptides in dental tissue engineering.
Collapse
Affiliation(s)
- Risako Suzuki
- Department of Oral and Maxillofacial Surgery and Plastic and Reconstructive Surgery, Graduate School of Medicine Advanced Medical Research Center, Faculty of Medicine, University of the Ryukyus, 1076 Kiyuna, Ginowan, Okinawa 901-2720, Japan
| | - Kentaro Ide
- Department of Oral and Maxillofacial Surgery and Plastic and Reconstructive Surgery, Graduate School of Medicine Advanced Medical Research Center, Faculty of Medicine, University of the Ryukyus, 1076 Kiyuna, Ginowan, Okinawa 901-2720, Japan
| | - Shusuke Tokuchi
- Department of Oral and Maxillofacial Surgery and Plastic and Reconstructive Surgery, Graduate School of Medicine Advanced Medical Research Center, Faculty of Medicine, University of the Ryukyus, 1076 Kiyuna, Ginowan, Okinawa 901-2720, Japan
| | - Toshihiro Kawano
- Department of Oral and Maxillofacial Surgery and Plastic and Reconstructive Surgery, Graduate School of Medicine Advanced Medical Research Center, Faculty of Medicine, University of the Ryukyus, 1076 Kiyuna, Ginowan, Okinawa 901-2720, Japan
| | - Edward Hosea Ntege
- Department of Oral and Maxillofacial Surgery and Plastic and Reconstructive Surgery, Graduate School of Medicine Advanced Medical Research Center, Faculty of Medicine, University of the Ryukyus, 1076 Kiyuna, Ginowan, Okinawa 901-2720, Japan
- Plastic and Reconstructive Surgery, Graduate School of Medicine, 1076 Kiyuna, Ginowan, Okinawa 901-2720, Japan
| | - Makoto Murahashi
- Department of Oral and Maxillofacial Surgery and Plastic and Reconstructive Surgery, Graduate School of Medicine Advanced Medical Research Center, Faculty of Medicine, University of the Ryukyus, 1076 Kiyuna, Ginowan, Okinawa 901-2720, Japan
| | - Mikana Isa
- Department of Oral and Maxillofacial Surgery and Plastic and Reconstructive Surgery, Graduate School of Medicine Advanced Medical Research Center, Faculty of Medicine, University of the Ryukyus, 1076 Kiyuna, Ginowan, Okinawa 901-2720, Japan
| | - Hiroshi Sunami
- Advanced Medical Research Center, Faculty of Medicine, University of the Ryukyus, 1076 Kiyuna, Ginowan, Okinawa 901-2720, Japan
| | - Yusuke Shimizu
- Plastic and Reconstructive Surgery, Graduate School of Medicine, 1076 Kiyuna, Ginowan, Okinawa 901-2720, Japan
| | - Hiroyuki Nakamura
- Department of Oral and Maxillofacial Surgery and Plastic and Reconstructive Surgery, Graduate School of Medicine Advanced Medical Research Center, Faculty of Medicine, University of the Ryukyus, 1076 Kiyuna, Ginowan, Okinawa 901-2720, Japan
| |
Collapse
|
3
|
Lim DS, Ahn SH, Cho W, Gwon HJ, Ko JH, Pyo MK, Abd El-Aty AM, Elubeyd E, Shin JW, Jeong JH, Lee KT, Jung TW. Myricetin promotes migration and prevents palmitate-induced apoptosis in cultured tenocytes through AMPK-dependent pathways. Biochem Biophys Res Commun 2025; 762:151764. [PMID: 40209502 DOI: 10.1016/j.bbrc.2025.151764] [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: 04/02/2025] [Accepted: 04/04/2025] [Indexed: 04/12/2025]
Abstract
Myricetin (Myr), a flavonoid present in vegetables and fruits, has been shown to ameliorate inflammation and oxidative stress in various disease models. However, the effects of Myr on hyperlipidemic tenocytes have not been studied. Herein, we aimed to investigate the effects of Myr on the features of tendinopathy in cultured tenocytes under hyperlipidemic conditions. Reactive oxygen species (ROS) were detected by DCFDA. Hydrogen peroxide (H2O2), malondialdehyde (MDA), and caspase 3 activity were quantified via matched assay kits. Apoptotic cells were detected via TUNEL staining. Proteins investigated in this study were evaluated through Western blotting. Treatment with Myr enhanced tenocyte migration and prevented apoptosis, inflammation and oxidative stress in palmitate-treated tenocytes. Myr treatment increased the phosphorylation of AMPK, and the expression of PGC1α and FGF2. siRNA targeting AMPK abrogated the effects of Myr on palmitate-treated tenocytes. However, FGF2 siRNA reduced the impacts of Myr on only cell migration and ECM signaling. These in vitro results suggest that Myr promotes tenocyte migration and ECM signaling via AMPK/FGF2 signaling and attenuates apoptosis through the AMPK-mediated suppression of inflammation and oxidative stress in hyperlipidemic tenocytes. This study sheds light on therapeutic strategies for treating obesity-related tendinopathy.
Collapse
Affiliation(s)
- Do Su Lim
- Department of Pharmacology, College of Medicine, Chung-Ang University, Seoul, Republic of Korea; Department of Global Innovative Drugs, Graduate School of Chung-Ang University, Seoul, Republic of Korea
| | - Sung Ho Ahn
- Department of Family Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Wonjun Cho
- Department of Pharmacology, College of Medicine, Chung-Ang University, Seoul, Republic of Korea
| | - Hyeon Ji Gwon
- Department of Pharmacology, College of Medicine, Chung-Ang University, Seoul, Republic of Korea; Department of Global Innovative Drugs, Graduate School of Chung-Ang University, Seoul, Republic of Korea
| | - Jun Hwi Ko
- Department of Pharmacology, College of Medicine, Chung-Ang University, Seoul, Republic of Korea; Department of Global Innovative Drugs, Graduate School of Chung-Ang University, Seoul, Republic of Korea
| | - Min Kyung Pyo
- Department of Pharmacology, College of Medicine, Chung-Ang University, Seoul, Republic of Korea; Department of Global Innovative Drugs, Graduate School of Chung-Ang University, Seoul, Republic of Korea
| | - A M Abd El-Aty
- Department of Pharmacology, Faculty of Veterinary Medicine, Cairo University, 12211, Giza, Egypt; Department of Medical Pharmacology, Medical Faculty, Ataturk University, Erzurum, 25240, Turkey.
| | - Emced Elubeyd
- Department of Nursing, Faculty of Health Sciences, Mardin Artuklu University, Mardin, Turkey
| | - Jong Wook Shin
- Department of Internal Medicine, Division of Allergy and Respiratory Medicine, Chung-Ang University, Seoul, Republic of Korea
| | - Ji Hoon Jeong
- Department of Pharmacology, College of Medicine, Chung-Ang University, Seoul, Republic of Korea; Department of Global Innovative Drugs, Graduate School of Chung-Ang University, Seoul, Republic of Korea
| | - Kyoung-Tae Lee
- Department of Forest Bioresources, Division of Forest Microbiology, National Institute of Forest Science, Suwon, 16631, Republic of Korea.
| | - Tae Woo Jung
- Department of Pharmacology, College of Medicine, Chung-Ang University, Seoul, Republic of Korea.
| |
Collapse
|
4
|
Huang BH, Lin HY, Ou YS, Lan WC, Tsai CH, Saito T, Shen HT, Chen W, Liu CM, Nakano H. Immobilization of biofunctional molecule with potential osteoinductive efficacy on titanium implant for promoting early-stage osseointegration. J Craniomaxillofac Surg 2025; 53:560-567. [PMID: 39884911 DOI: 10.1016/j.jcms.2025.01.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 12/31/2024] [Accepted: 01/17/2025] [Indexed: 02/01/2025] Open
Abstract
In the present study, porcine-derived collagen type I was covalently immobilized on the surface of titanium (Ti) implants via carboxyl groups introduced by bonded p-vinylbenzoic acid to investigate its in vitro biocompatibility with gingival stem cells and in vivo bone regeneration behavior in the edentulous ridges of Lanyu small-ear pigs at weeks 2 and 6 (short-term effectiveness) through micro-computed tomography and histological analysis. Analytical results found that gingival stem cells showed effective adhesion and spreading on these collagen-immobilized implant surfaces. After 2 and 6 weeks of healing, significant differences in Hounsfield units were observed among the control (week 2 (674.2 ± 79.9) ∗∗p < 0.01 and week 6 (596.4 ± 49.6) ∗∗p < 0.01), buffer-coated implant (week 2 (768.1 ± 68.7) ∗p < 0.05 and week 6 (720.4 ± 62.6) ∗p < 0.05), and collagen-immobilized implant (week 2 (828.2 ± 69.4) and week 6 (907.4 ± 63.5)) groups. No significant differences in bone-to-implant contact ratios were discovered between the investigated groups. However, the bone surface area results demonstrated an enhanced bone apposition for the collagen-immobilized implants compared to the control and buffer-coated implants at weeks 2 and 6 post-implantation (∗p < 0.05). Therefore, this preclinical study underscores the advantageous impact of collagen immobilization on Ti implant surfaces for clinical application, substantiating its effectiveness through significant evidence of improved osseointegration at early-stages.
Collapse
Affiliation(s)
- Bai-Hung Huang
- Graduate Institute of Dental Science, College of Dentistry, China Medical University, Taichung 404, Taiwan
| | - Hung-Yang Lin
- Department of Dentistry, Fu Jen Catholic University Hospital, Fu Jen Catholic University, New Taipei City 242, Taiwan
| | - Yu-Sin Ou
- General Biology, Warren College, University of California, San Diego, CA 92093, USA
| | - Wen-Chien Lan
- Department of Oral Hygiene Care, Deh Yu College of Nursing and Health, Keelung 203, Taiwan
| | - Chi-Hsun Tsai
- Division of Clinical Cariology and Endodontology, Department of Oral Rehabilitation, School of Dentistry, Health Sciences University of Hokkaido, Tobetsu 061-0293, Japan
| | - Takashi Saito
- Division of Clinical Cariology and Endodontology, Department of Oral Rehabilitation, School of Dentistry, Health Sciences University of Hokkaido, Tobetsu 061-0293, Japan
| | - Hsieh-Tsung Shen
- Division of Clinical Cariology and Endodontology, Department of Oral Rehabilitation, School of Dentistry, Health Sciences University of Hokkaido, Tobetsu 061-0293, Japan; EG BioMed US Inc., Covina, CA 91722, USA; Yu Ding Global Cancer Research Foundation, Taipei 115, Taiwan.
| | - Wayne Chen
- Taipei American School, Taipei 111, Taiwan
| | - Chung-Ming Liu
- Department of Biomedical Engineering, College of Biomedical Engineering, China Medical University, Taichung 404, Taiwan.
| | - Hiroyuki Nakano
- Department of Oral and Maxillofacial Surgery, Kanazawa Medical University, Ishikawa 920-0293, Japan
| |
Collapse
|
5
|
Huang P, Yang H, Kuang H, Yang J, Duan X, Bian H, Wang X. Pancreaticobiliary Maljunction: A Multidimensional Exploration of Pathophysiology, Diagnosis, Classification, Management and Research Prospects. Dig Dis Sci 2025:10.1007/s10620-025-09057-0. [PMID: 40252147 DOI: 10.1007/s10620-025-09057-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Accepted: 04/09/2025] [Indexed: 04/21/2025]
Abstract
Pancreaticobiliary maljunction is a congenital malformation in which the pancreatic and bile ducts join anatomically outside the duodenal wall, usually forming a markedly long common channel, which can cause reciprocal reflux between pancreatic juice and bile. Cholangiography, endoscopic ultrasonography, surgery, and autopsy can be used to diagnose pancreaticobiliary maljunction. Elevated amylase levels in bile and extrahepatic bile duct dilatation strongly suggest the existence of pancreaticobiliary maljunction. The regurgitation may lead to the development of various hepatobiliary and pancreatic disorders such as pancreatitis and biliary carcinoma. The pathogenesis of pancreaticobiliary maljunction is the result of a series of pathophysiological changes caused by reflux. Surgery is recommended for patients diagnosed with pancreaticobiliary maljunction irrespective of the presence or absence of symptoms because of its high biliary carcinogenicity, but the treatment strategy is quite different between adult patients with and without biliary dilatation.
Collapse
Affiliation(s)
- Peng Huang
- Department of General Surgery, Wuhan Children's Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430016, Hubei, China
| | - Hu Yang
- Department of General Surgery, Wuhan Children's Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430016, Hubei, China
| | - Houfang Kuang
- Department of General Surgery, Wuhan Children's Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430016, Hubei, China
| | - Jun Yang
- Department of General Surgery, Wuhan Children's Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430016, Hubei, China
| | - Xufei Duan
- Department of General Surgery, Wuhan Children's Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430016, Hubei, China
| | - Hongqiang Bian
- Department of General Surgery, Wuhan Children's Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430016, Hubei, China
| | - Xin Wang
- Department of General Surgery, Wuhan Children's Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430016, Hubei, China.
| |
Collapse
|
6
|
Liu J, Zhang M, Zhou M, Wang Q, Jiang X, Huang Q. Exploring Biomaterial Scaffolds for Eyelid Reconstruction: A Synthesis of Experimental Findings. TISSUE ENGINEERING. PART B, REVIEWS 2025. [PMID: 40242856 DOI: 10.1089/ten.teb.2024.0364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/18/2025]
Abstract
This review synthesizes experimental findings on various biomaterial scaffolds used in eyelid reconstruction. It examines the structural properties, cellular responses, and functional outcomes of scaffolds such as chitosan, poly(propylene glycol fumarate)-2-hydroxyethyl methacrylate, poly(propylene glycol fumarate) - type I collagen (PPF-Col), decellularized matrix-polycaprolactone, branched polyethylene, collagen, poly(3-hydroxybutyrate-co-3-hydroxyhexanoate, and poly(lactic-co-glycolic acid. These scaffolds exhibit diverse mechanical and biological properties, with some demonstrating good biocompatibility, tunable properties, and potential for tissue repair. However, there are limitations, including concerns about long-term functionality and a lack of comprehensive evaluations. This review highlights the need for multifunctional scaffolds that combine lid replacement and ocular surface function restoration, as well as the establishment of standardized research methods. The goal is to guide future innovation in the field and improve the quality of life for patients with eyelid defects.
Collapse
Affiliation(s)
- Jincheng Liu
- School of Optometry, Jiangxi Medical College, Nanchang University, Nanchang, China
- Jiangxi Research Institute of Ophthalmology and Visual Science, Nanchang, China
- Jiangxi Provincial Key Laboratory for Ophthalmology, Nanchang, China
- National clinical research center for ocular diseases Jiangxi Province division, Nanchang, China
- The Affiliated Eye Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
- Jiangxi Clinical Research Center for Ophthalmic Disease, Nanchang, China
| | - Mange Zhang
- School of Optometry, Jiangxi Medical College, Nanchang University, Nanchang, China
- Jiangxi Research Institute of Ophthalmology and Visual Science, Nanchang, China
- Jiangxi Provincial Key Laboratory for Ophthalmology, Nanchang, China
- National clinical research center for ocular diseases Jiangxi Province division, Nanchang, China
- The Affiliated Eye Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
- Jiangxi Clinical Research Center for Ophthalmic Disease, Nanchang, China
| | - Mengling Zhou
- School of Optometry, Jiangxi Medical College, Nanchang University, Nanchang, China
- Jiangxi Research Institute of Ophthalmology and Visual Science, Nanchang, China
- Jiangxi Provincial Key Laboratory for Ophthalmology, Nanchang, China
- National clinical research center for ocular diseases Jiangxi Province division, Nanchang, China
- The Affiliated Eye Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
- Jiangxi Clinical Research Center for Ophthalmic Disease, Nanchang, China
| | - Qingyi Wang
- School of Optometry, Jiangxi Medical College, Nanchang University, Nanchang, China
- Jiangxi Research Institute of Ophthalmology and Visual Science, Nanchang, China
- Jiangxi Provincial Key Laboratory for Ophthalmology, Nanchang, China
- National clinical research center for ocular diseases Jiangxi Province division, Nanchang, China
- The Affiliated Eye Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
- Jiangxi Clinical Research Center for Ophthalmic Disease, Nanchang, China
| | - Xin Jiang
- School of Optometry, Jiangxi Medical College, Nanchang University, Nanchang, China
- Jiangxi Research Institute of Ophthalmology and Visual Science, Nanchang, China
- Jiangxi Provincial Key Laboratory for Ophthalmology, Nanchang, China
- National clinical research center for ocular diseases Jiangxi Province division, Nanchang, China
- The Affiliated Eye Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
- Jiangxi Clinical Research Center for Ophthalmic Disease, Nanchang, China
| | - Qin Huang
- School of Optometry, Jiangxi Medical College, Nanchang University, Nanchang, China
- Jiangxi Research Institute of Ophthalmology and Visual Science, Nanchang, China
- Jiangxi Provincial Key Laboratory for Ophthalmology, Nanchang, China
- National clinical research center for ocular diseases Jiangxi Province division, Nanchang, China
- The Affiliated Eye Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
- Jiangxi Clinical Research Center for Ophthalmic Disease, Nanchang, China
| |
Collapse
|
7
|
Azari Z, Sadeghi-Avalshahr A, Alipour F, Kondori BJ, Askari VR, Mollazadeh S, Nazarnezhad S, Nasiri SN, Kermani F, Ranjbar-Mohammadi M. Advanced nanofibers integrating vitamin D3 and cerium oxide nanoparticles for enhanced diabetic wound healing: Co-electrospun silk fibroin-collagen and chitosan-PVA systems. Int J Biol Macromol 2025; 310:143099. [PMID: 40222506 DOI: 10.1016/j.ijbiomac.2025.143099] [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: 12/25/2024] [Revised: 04/03/2025] [Accepted: 04/10/2025] [Indexed: 04/15/2025]
Abstract
This study investigates the co-electrospinning of polyvinyl alcohol-chitosan (PVA-CS) with cerium oxide nanoparticles (CeNPs) and silk fibroin-collagen (SF-Col) with vitamin D3 for diabetic wound healing applications. The SEM results showed smooth, bead-free nanofiber structures. The diameters of the SF-Col and PVA-CS nanofibers ranged from 168 ± 51 nm to 1956 ± 450 nm and 211.4 ± 37.2 nm, respectively. By surface modification using fetal bovine serum (FBS), CeNPs dispersion was enhanced. The average diameter of the uniformly distributed fibers on the SF-Co-D/PVA-CS-CeNPs nanofibers was 621.4 ± 50.6 nm. The addition of CeNPs and vitamin D3 improved cytocompatibility at lower doses. The FTIR test confirmed polymer interactions. Contact angle measurements indicated increased hydrophilicity. SEM analysis demonstrated excellent adhesion and growth of L929 fibroblast cells and significant HUVEC migration on SF-Col-D/PVA-CS-CeNP mats, emphasizing their potential to support cell proliferation and tissue regeneration. Blood compatibility assays exhibited hemolysis percentages below 2 %, classifying the nanofibers as non-hemolytic. Antibacterial tests revealed significant reductions in Staphylococcus aureus and Pseudomonas aeruginosa survival, addressing infection concerns in chronic wounds. Furthermore, in vivo studies have demonstrated that the utilization of SF-Co-D/PVA-CS-CeNPs nanofibrous membrane as a dressing for full-thickness skin wounds in rats has resulted in accelerated tissue regeneration.
Collapse
Affiliation(s)
- Zoleikha Azari
- Bhbahan Faculty of Medical Sciences, Behbahan, Iran; Department of Anatomy and Cell Biology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Alireza Sadeghi-Avalshahr
- Department of Materials Research, Iranian Academic Center for Education, Culture and Research (ACECR), Khorasan Razavi Branch, Mashhad, Iran; Tissue Engineering Research Group, Department of Anatomy and Cell Biology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Fatemeh Alipour
- Department of Anatomy and Cell Biology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Bahman Jalali Kondori
- Department of Anatomy and Cell Biology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Vahid Reza Askari
- Pharmacological Research Center of Medicinal Plants, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Sahar Mollazadeh
- Department of Materials Engineering, Faculty of Engineering, Ferdowsi University of Mashhad, Azadi Sq., Mashhad, Iran
| | - Simin Nazarnezhad
- Tissue Engineering Research Group, Department of Anatomy and Cell Biology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyedeh Najibeh Nasiri
- Tissue Engineering Research Group, Department of Anatomy and Cell Biology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Farzad Kermani
- Tissue Engineering Research Group, Department of Anatomy and Cell Biology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | | |
Collapse
|
8
|
Jaiswal AK, Raj A, Kushawaha AK, Maji B, Bhatt H, Verma S, Katiyar S, Ansari A, Bisen AC, Tripathi A, Siddiqi MI, Bhatta RS, Trivedi R, Sashidhara KV. Design, synthesis and biological evaluation of new class of pyrazoles-dihydropyrimidinone derivatives as bone anabolic agents. Bioorg Chem 2025; 157:108216. [PMID: 39952063 DOI: 10.1016/j.bioorg.2025.108216] [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: 10/10/2024] [Revised: 01/22/2025] [Accepted: 01/25/2025] [Indexed: 02/17/2025]
Abstract
This study explores a series of twenty-four newly synthesized pyrzole-dihydropyrimidinone hybrids as potential bone anabolic agents. Initially, an alkaline phosphatase assay, a common marker of bone formation, was used to screen all compounds for their ability to stimulate osteogenic potential. Initial screening identified three promising candidates (5f, 5u and 5w) that were subsequently confirmed to be non-toxic to osteoblasts. Further investigation revealed that compound 5w displayed the most potent osteoanabolic effect, promoting osteoblast differentiation and upregulating mRNAs expression of osteogenic gene. Based on the promising in vitro and in vivo activity, structure-activity relationship (SAR) analysis revealed a furan ring on the dihydropyrimidinone unit and electron-donating groups on the N-phenyl ring of the pyrazole moiety to be crucial for osteogenic activity. Additionally, molecular docking, favorable pharmacokinetic properties and In silico ADME predictions suggest potential oral bioavailability. These findings establish the pyrazole-dihydropyrimidinone scaffold as a promising hit for developing a new class of orally active bone anabolic agents.
Collapse
Affiliation(s)
- Arvind Kumar Jaiswal
- Medicinal and Process Chemistry Division, CSIR-Central Drug Research Institute, Jankipuram Extension, Sitapur Road, Lucknow 226031, India
| | - Anuj Raj
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, U.P., India; Division of Endocrinology, CSIR-Central Drug Research Institute, Jankipuram Extension, Sitapur Road, Lucknow 226031, India
| | - Ajay Kishor Kushawaha
- Medicinal and Process Chemistry Division, CSIR-Central Drug Research Institute, Jankipuram Extension, Sitapur Road, Lucknow 226031, India
| | - Bhaskar Maji
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, U.P., India; Division of Endocrinology, CSIR-Central Drug Research Institute, Jankipuram Extension, Sitapur Road, Lucknow 226031, India
| | - Hemlata Bhatt
- Medicinal and Process Chemistry Division, CSIR-Central Drug Research Institute, Jankipuram Extension, Sitapur Road, Lucknow 226031, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, U.P., India
| | - Shikha Verma
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, U.P., India; Division of Endocrinology, CSIR-Central Drug Research Institute, Jankipuram Extension, Sitapur Road, Lucknow 226031, India
| | - Sarita Katiyar
- Medicinal and Process Chemistry Division, CSIR-Central Drug Research Institute, Jankipuram Extension, Sitapur Road, Lucknow 226031, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, U.P., India
| | - Alisha Ansari
- Medicinal and Process Chemistry Division, CSIR-Central Drug Research Institute, Jankipuram Extension, Sitapur Road, Lucknow 226031, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, U.P., India
| | - Amol Chhatrapati Bisen
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, U.P., India; Sophisticated Analytical Instrument Facility & Research, CSIR-Central Drug Research Institute, Jankipuram Extension, Sitapur Road, Lucknow 226031, U.P., India; Pharmaceutics & Pharmacokinetics Division, CSIR-Central Drug Research Institute, Lucknow, India
| | - Arsh Tripathi
- Biochemistry & Structural Biology Division, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Mohammad Imran Siddiqi
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, U.P., India; Biochemistry & Structural Biology Division, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Rabi Sankar Bhatta
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, U.P., India; Pharmaceutics & Pharmacokinetics Division, CSIR-Central Drug Research Institute, Lucknow, India
| | - Ritu Trivedi
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, U.P., India; Division of Endocrinology, CSIR-Central Drug Research Institute, Jankipuram Extension, Sitapur Road, Lucknow 226031, India.
| | - Koneni V Sashidhara
- Medicinal and Process Chemistry Division, CSIR-Central Drug Research Institute, Jankipuram Extension, Sitapur Road, Lucknow 226031, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, U.P., India; Sophisticated Analytical Instrument Facility & Research, CSIR-Central Drug Research Institute, Jankipuram Extension, Sitapur Road, Lucknow 226031, U.P., India.
| |
Collapse
|
9
|
Wang H, Wu X, Chen L, Tong H, Hu X, He A, Li C, Guo X, Fu Y, Zhang T. Dynamic Col-HZ Hydrogel with efficient delivery of bioactivator promotes ECM deposition and cartilage formation. Mater Today Bio 2025; 31:101623. [PMID: 40104649 PMCID: PMC11914768 DOI: 10.1016/j.mtbio.2025.101623] [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: 01/16/2025] [Revised: 02/17/2025] [Accepted: 02/27/2025] [Indexed: 03/20/2025] Open
Abstract
Efforts in cartilage tissue engineering to repair injuries have seen limited success, primarily due to the inability of scaffold materials to establish a microenvironment conducive to extracellular matrix (ECM) deposition by chondrocytes. Hydrogels, which mimic human tissue, are commonly employed as scaffold materials; however, their constrained network structure and low bioactivity impede chondrocyte ECM deposition, complicating cartilage repair. In this study, we developed dynamic Col-HZ hydrogels featuring adaptive networks by forming hydrazone (HZ) bonds between bioactive natural collagen and synthetic polyethylene glycol (PEG). In contrast to static hydrogels that rely on covalent bonds, Col-HZ dynamic hydrogels facilitate chondrocyte migration and ECM deposition. Additionally, the aldehyde groups on the Col-HZ hydrogel scaffold can engage in dynamic Schiff base bonding with amine groups. Leveraging this non-covalent interaction, we incorporated the bioactivator TD-198946, known to enhance ECM synthesis, into the Col-HZ hydrogel. This significantly boosted ECM deposition and reduced inflammation. Transcriptomic sequencing and bioinformatics analyses indicate that both the dynamic network of the hydrogel and the binding of TD-198946 promote cartilage ECM deposition through modulation of the Wnt/β-catenin signaling pathway. Consequently, the Col-HZ dynamic hydrogel, in combination with TD-198946, creates an improved microenvironment that supports ECM deposition and facilitates cartilage tissue formation.
Collapse
Affiliation(s)
- Honglei Wang
- Department of Facial Plastic and Reconstructive Surgery, Eye & ENT Hospital, Fudan University, Shanghai, 200031, China
- ENT Institute, Eye & ENT Hospital, Fudan University, Shanghai, 200031, China
| | - Xu Wu
- Department of Facial Plastic and Reconstructive Surgery, Eye & ENT Hospital, Fudan University, Shanghai, 200031, China
- ENT Institute, Eye & ENT Hospital, Fudan University, Shanghai, 200031, China
| | - Lili Chen
- Department of Facial Plastic and Reconstructive Surgery, Eye & ENT Hospital, Fudan University, Shanghai, 200031, China
- ENT Institute, Eye & ENT Hospital, Fudan University, Shanghai, 200031, China
| | - Hua Tong
- Department of Facial Plastic and Reconstructive Surgery, Eye & ENT Hospital, Fudan University, Shanghai, 200031, China
- ENT Institute, Eye & ENT Hospital, Fudan University, Shanghai, 200031, China
| | - Xuerui Hu
- Department of Facial Plastic and Reconstructive Surgery, Eye & ENT Hospital, Fudan University, Shanghai, 200031, China
- ENT Institute, Eye & ENT Hospital, Fudan University, Shanghai, 200031, China
| | - Aijuan He
- Department of Facial Plastic and Reconstructive Surgery, Eye & ENT Hospital, Fudan University, Shanghai, 200031, China
- ENT Institute, Eye & ENT Hospital, Fudan University, Shanghai, 200031, China
| | - Chenlong Li
- Department of Facial Plastic and Reconstructive Surgery, Eye & ENT Hospital, Fudan University, Shanghai, 200031, China
- ENT Institute, Eye & ENT Hospital, Fudan University, Shanghai, 200031, China
| | - Xudong Guo
- Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Shanghai Key Laboratory of Maternal Fetal Medicine, Frontier Science Center for Stem Cell Research, National Stem Cell Translational Resource Center, School of Life Sciences and Technology, Tongji University, China
| | - Yaoyao Fu
- Department of Facial Plastic and Reconstructive Surgery, Eye & ENT Hospital, Fudan University, Shanghai, 200031, China
- ENT Institute, Eye & ENT Hospital, Fudan University, Shanghai, 200031, China
| | - Tianyu Zhang
- Department of Facial Plastic and Reconstructive Surgery, Eye & ENT Hospital, Fudan University, Shanghai, 200031, China
- ENT Institute, Eye & ENT Hospital, Fudan University, Shanghai, 200031, China
- NHC Key Laboratory of Hearing Medicine, Fudan University, Shanghai, 200031, China
| |
Collapse
|
10
|
Ullah S, Zainol I. Fabrication and applications of biofunctional collagen biomaterials in tissue engineering. Int J Biol Macromol 2025; 298:139952. [PMID: 39824416 DOI: 10.1016/j.ijbiomac.2025.139952] [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: 10/23/2024] [Revised: 01/12/2025] [Accepted: 01/14/2025] [Indexed: 01/20/2025]
Abstract
Collagen is extensively used in tissue engineering for various organ tissue regeneration due to the main component of human organ extracellular matrix (ECM) and their inherent nature bioactivity. Collagen various types naturally exist in different organ ECMs. Collagen fabricated with natural ECM mimics architecture, composition and mechanical properties for various organ tissue regeneration. Collagen fabrication with organ-specific biofunctionality facilitated organ tissue engineering as compared to unmodified collagen biomaterials. Collagen biofunctionality improved by subjecting collagen to synthesis, fibers and surface modifications, and blending with other components. Furthermore, collagen is loaded with bioactive molecules, growth factors, drugs and cells also enhancing the biofunctionality of collagen biomaterials. In this review, we will explore the recent advancements in biofunctional collagen biomaterials fabrication with organ-specific biofunctionality in tissue engineering to resolve various organ tissue engineering issues and regeneration challenges. Biofunctional collagen biomaterials stimulate microenvironments inside and around the implants to excellently regulate cellular activities, differentiate cells into organ native cells, enhanced ECM production and remodeling to regenerate organ tissues with native structure, function and maturation. This review critically explored biofunctional collagen biomaterials fabrication in resolving various organ tissue engineering issues and regeneration challenges, and opening new directions of biofunctional collagen biomaterials fabrication, design and applications.
Collapse
Affiliation(s)
- Saleem Ullah
- Polymer Lab, Chemistry Department, Faculty of Science and Mathematics, Universiti Pendidikan Sultan Idris, 35900 Tanjung Malim, Perak, Darul Ridzuan, Malaysia.
| | - Ismail Zainol
- Polymer Lab, Chemistry Department, Faculty of Science and Mathematics, Universiti Pendidikan Sultan Idris, 35900 Tanjung Malim, Perak, Darul Ridzuan, Malaysia.
| |
Collapse
|
11
|
Lee J, Ryu J, Choi J, Chae I, Kim SH. Vibrational Sum Frequency Generation Spectroscopy Study of Nanoscale to Mesoscale Polarity and Orientation of Crystalline Biopolymers in Natural Materials. Annu Rev Phys Chem 2025; 76:405-430. [PMID: 39971375 DOI: 10.1146/annurev-physchem-082423-125535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
As a nonlinear optical process, sum frequency generation (SFG) requires noncentrosymmetry across multiple length scales, ranging from individual molecular functional groups to their arrangements in space. This principle makes SFG not only intrinsically sensitive to molecular species at surfaces but also useful for studying 3D structures of crystalline biopolymers in natural materials. Examples of such biopolymers are cellulose, starch, and chitin in the polysaccharide family and collagen, silk, and keratin in the fibrous protein family. These biopolymers are noncentrosymmetric at multiple length scales, with chirality at the molecular scale, unit cell structure at the nanoscale, and crystallite orientation and polarity at the mesoscale; thus, they are SFG active. In this review, we describe how SFG can be used to determine nano- to mesoscale polarity and orientational orders of crystalline biopolymers interspersed in natural materials containing the same or similar biopolymers in amorphous states, which cannot be obtained with other characterization methods.
Collapse
Affiliation(s)
- Jongcheol Lee
- Department of Chemical Engineering and Materials Research Institute, Pennsylvania State University, University Park, Pennsylvania, USA;
| | - Jihyeong Ryu
- Department of Chemical Engineering and Materials Research Institute, Pennsylvania State University, University Park, Pennsylvania, USA;
| | - Juseok Choi
- Department of Chemical Engineering and Materials Research Institute, Pennsylvania State University, University Park, Pennsylvania, USA;
| | - Inseok Chae
- Department of Bioengineering, University of California, Berkeley, California, USA
| | - Seong H Kim
- Department of Chemical Engineering and Materials Research Institute, Pennsylvania State University, University Park, Pennsylvania, USA;
| |
Collapse
|
12
|
Chocarro-Wrona C, Pleguezuelos-Beltrán P, López de Andrés J, Antich C, de Vicente J, Jiménez G, Arias-Santiago S, Gálvez-Martín P, López-Ruiz E, Marchal JA. A bioactive three-layered skin substitute based on ECM components effectively promotes skin wound healing and regeneration. Mater Today Bio 2025; 31:101592. [PMID: 40092225 PMCID: PMC11910132 DOI: 10.1016/j.mtbio.2025.101592] [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: 09/13/2024] [Revised: 02/14/2025] [Accepted: 02/17/2025] [Indexed: 03/19/2025] Open
Abstract
To overcome the limitations of conventional skin tissue engineering (TE), 3D biofabrication approaches are being developed. However, tissue mimicry should be further improved in skin models. Here, we developed and characterized biomimetic hydrogels to obtain a biofabricated three-layered (BT) skin substitute based on the main components found in the epidermal, dermal, and hypodermal skin layers. Hydrogels for dermal and hypodermal skin layers were based on a mix of agarose and type I collagen, supplemented with skin-related extracellular matrix (ECM) components (dermatan sulfate, hyaluronic acid, and elastin) and loaded with human dermal fibroblasts (hDFs) or human mesenchymal stem/stromal cells (hMSCs), respectively. The epidermal hydrogel was formulated using type I collagen supplemented with keratin and sphingolipids, and seeded with human epidermal keratinocytes (hEKs). Physicochemical results revealed adequate viscosity, gelling times, and pH for each hydrogel solution. The BT Skin also showed good swelling and degradation kinetics, and mechanical properties in a similar range of human skin. The hydrogels and BT Skin demonstrated stable cell viability and metabolic activity, as well as intercellular communication through the release of growth factors. Moreover, the BT Skin demonstrated controlled inflammation in vivo, and produced results comparable to autografting in a mouse skin wound model. This bioactive and biomimetic three-layered BT Skin has a composition that attempts to mimic the natural ECM of the skin, formulated with the characteristic cells and biomolecules present in each skin layer, and offers promising properties for its clinical application in the treatment of patients with skin injuries.
Collapse
Affiliation(s)
- Carlos Chocarro-Wrona
- Biopathology and Regenerative Medicine Institute (IBIMER), Center for Biomedical Research (CIBM), University of Granada, 18016, Granada, Spain
- Instituto de Investigación Biosanitaria ibs.GRANADA, University Hospitals of Granada, University of Granada, 18012, Granada, Spain
- Department of Human Anatomy and Embryology, Faculty of Medicine, University of Granada, 18016, Granada, Spain
- Excellence Research Unit “Modelling Nature” (MNat), University of Granada, 18016, Granada, Spain
- BioFab i3D - Biofabrication and 3D (bio)printing laboratory, University of Granada, 18016, Granada, Spain
| | - Paula Pleguezuelos-Beltrán
- Biopathology and Regenerative Medicine Institute (IBIMER), Center for Biomedical Research (CIBM), University of Granada, 18016, Granada, Spain
- Instituto de Investigación Biosanitaria ibs.GRANADA, University Hospitals of Granada, University of Granada, 18012, Granada, Spain
- Department of Human Anatomy and Embryology, Faculty of Medicine, University of Granada, 18016, Granada, Spain
- Excellence Research Unit “Modelling Nature” (MNat), University of Granada, 18016, Granada, Spain
- BioFab i3D - Biofabrication and 3D (bio)printing laboratory, University of Granada, 18016, Granada, Spain
| | - Julia López de Andrés
- Biopathology and Regenerative Medicine Institute (IBIMER), Center for Biomedical Research (CIBM), University of Granada, 18016, Granada, Spain
- Instituto de Investigación Biosanitaria ibs.GRANADA, University Hospitals of Granada, University of Granada, 18012, Granada, Spain
- Department of Human Anatomy and Embryology, Faculty of Medicine, University of Granada, 18016, Granada, Spain
- Excellence Research Unit “Modelling Nature” (MNat), University of Granada, 18016, Granada, Spain
- BioFab i3D - Biofabrication and 3D (bio)printing laboratory, University of Granada, 18016, Granada, Spain
| | - Cristina Antich
- Biopathology and Regenerative Medicine Institute (IBIMER), Center for Biomedical Research (CIBM), University of Granada, 18016, Granada, Spain
- Instituto de Investigación Biosanitaria ibs.GRANADA, University Hospitals of Granada, University of Granada, 18012, Granada, Spain
- Excellence Research Unit “Modelling Nature” (MNat), University of Granada, 18016, Granada, Spain
- BioFab i3D - Biofabrication and 3D (bio)printing laboratory, University of Granada, 18016, Granada, Spain
- National Center for Advancing Translational Sciences, National Institute of Health, 28050, Rockville, MD, USA
| | - Juan de Vicente
- Excellence Research Unit “Modelling Nature” (MNat), University of Granada, 18016, Granada, Spain
- F2N2Lab, Magnetic Soft Matter Group, Department of Applied Physics, Faculty of Sciences, University of Granada, 18071, Granada, Spain
| | - Gema Jiménez
- Biopathology and Regenerative Medicine Institute (IBIMER), Center for Biomedical Research (CIBM), University of Granada, 18016, Granada, Spain
- Instituto de Investigación Biosanitaria ibs.GRANADA, University Hospitals of Granada, University of Granada, 18012, Granada, Spain
- Department of Human Anatomy and Embryology, Faculty of Medicine, University of Granada, 18016, Granada, Spain
- Excellence Research Unit “Modelling Nature” (MNat), University of Granada, 18016, Granada, Spain
- BioFab i3D - Biofabrication and 3D (bio)printing laboratory, University of Granada, 18016, Granada, Spain
| | - Salvador Arias-Santiago
- Instituto de Investigación Biosanitaria ibs.GRANADA, University Hospitals of Granada, University of Granada, 18012, Granada, Spain
- Dermatology Department, Hospital Universitario Virgen de las Nieves, 18012, Granada, Spain
- Dermatology Department, Faculty of Medicine, University of Granada, 18016, Granada, Spain
| | | | - Elena López-Ruiz
- Biopathology and Regenerative Medicine Institute (IBIMER), Center for Biomedical Research (CIBM), University of Granada, 18016, Granada, Spain
- Instituto de Investigación Biosanitaria ibs.GRANADA, University Hospitals of Granada, University of Granada, 18012, Granada, Spain
- Excellence Research Unit “Modelling Nature” (MNat), University of Granada, 18016, Granada, Spain
- BioFab i3D - Biofabrication and 3D (bio)printing laboratory, University of Granada, 18016, Granada, Spain
- Department of Health Sciences, University of Jaén, 23071, Jaén, Spain
| | - Juan Antonio Marchal
- Biopathology and Regenerative Medicine Institute (IBIMER), Center for Biomedical Research (CIBM), University of Granada, 18016, Granada, Spain
- Instituto de Investigación Biosanitaria ibs.GRANADA, University Hospitals of Granada, University of Granada, 18012, Granada, Spain
- Department of Human Anatomy and Embryology, Faculty of Medicine, University of Granada, 18016, Granada, Spain
- Excellence Research Unit “Modelling Nature” (MNat), University of Granada, 18016, Granada, Spain
- BioFab i3D - Biofabrication and 3D (bio)printing laboratory, University of Granada, 18016, Granada, Spain
| |
Collapse
|
13
|
Zhang Y, Li Y, Wu X, Wang S, Wei X, Sun X. Recombinant Humanized Collagen: A Promising Treatment for Pelvic Organ Prolapse via Enhanced Fibroblast Function and Angiogenesis. Int Urogynecol J 2025:10.1007/s00192-025-06117-x. [PMID: 40116904 DOI: 10.1007/s00192-025-06117-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Accepted: 03/03/2025] [Indexed: 03/23/2025]
Abstract
INTRODUCTION AND HYPOTHESIS The treatment of pelvic organ prolapse (POP) presents significant challenges. It is important to explore safer and more effective treatment modalities. Recombinant humanized collagen (rhCol) is a promising biomaterial with excellent biocompatibility and pro-regenerative properties. Therefore, this study aims to evaluate the potential applications of rhCol in POP treatment. METHODS Vaginal wall tissues were collected from three non-POP and five POP patients to analyze extracellular matrix (ECM) changes via histological staining. Primary fibroblasts isolated from POP vaginal tissues were treated with rhCol III. Cell proliferation, migration, senescence, and ECM synthesis were assessed. A simulated birth injury (SBI) rat model was used to evaluate ECM remodeling following rhCol injection into the vaginal wall. Additionally, the angiogenic potential of rhCol III was examined in vivo and in vitro. RESULTS POP patient tissues and fibroblasts exhibited lower expression levels of type I and III collagen compared to non-POP samples. At a 1 mg/ml concentration, rhCol III promoted fibroblast proliferation and migration, reduced cellular senescence, and enhanced ECM synthesis. In the vaginal wall, the expression of COL1A1 and COL3A1 in the rhCol group was significantly higher than that in the SBI group, with a marked increase in the levels of CD31, CD34, and VEGFA. Furthermore, rhCol III improved the proliferation, migration, and tubule formation capacities of HUVECs. CONCLUSIONS rhCol III may promote ECM remodeling in an injured vaginal wall by restoring fibroblast function and stimulating angiogenesis, offering a novel biomaterial-based strategy for POP treatment.
Collapse
Affiliation(s)
- Yue Zhang
- Department of Obstetrics and Gynecology, Peking University People's Hospital, No.11, Xi-Zhi-Men South Street, Xicheng District, Beijing, 100044, China
- Pelvic Floor Disorders Research Center of Peking University Health Science Center, Beijing, 100044, China
- Research Center of Female Pelvic Floor Disorders of Peking University, Beijing, 100044, China
| | - Yaqin Li
- Department of Obstetrics and Gynecology, Peking University People's Hospital, No.11, Xi-Zhi-Men South Street, Xicheng District, Beijing, 100044, China
- Pelvic Floor Disorders Research Center of Peking University Health Science Center, Beijing, 100044, China
- Research Center of Female Pelvic Floor Disorders of Peking University, Beijing, 100044, China
| | - Xiaotong Wu
- Department of Obstetrics and Gynecology, Peking University People's Hospital, No.11, Xi-Zhi-Men South Street, Xicheng District, Beijing, 100044, China
- Pelvic Floor Disorders Research Center of Peking University Health Science Center, Beijing, 100044, China
- Research Center of Female Pelvic Floor Disorders of Peking University, Beijing, 100044, China
| | - Shiyan Wang
- Department of Obstetrics and Gynecology, Peking University People's Hospital, No.11, Xi-Zhi-Men South Street, Xicheng District, Beijing, 100044, China
- Pelvic Floor Disorders Research Center of Peking University Health Science Center, Beijing, 100044, China
- Research Center of Female Pelvic Floor Disorders of Peking University, Beijing, 100044, China
| | - Xiaoting Wei
- Department of Obstetrics and Gynecology, Peking University People's Hospital, No.11, Xi-Zhi-Men South Street, Xicheng District, Beijing, 100044, China
- Pelvic Floor Disorders Research Center of Peking University Health Science Center, Beijing, 100044, China
- Research Center of Female Pelvic Floor Disorders of Peking University, Beijing, 100044, China
| | - Xiuli Sun
- Department of Obstetrics and Gynecology, Peking University People's Hospital, No.11, Xi-Zhi-Men South Street, Xicheng District, Beijing, 100044, China.
- Pelvic Floor Disorders Research Center of Peking University Health Science Center, Beijing, 100044, China.
- Research Center of Female Pelvic Floor Disorders of Peking University, Beijing, 100044, China.
| |
Collapse
|
14
|
Huang S, Ng N, Vaez M, Hinz B, Leong I, Bozec L. Collagen Hybridizing Peptides Promote Collagen Fibril Growth In Vitro. ACS APPLIED BIO MATERIALS 2025; 8:2003-2014. [PMID: 40010706 PMCID: PMC11921027 DOI: 10.1021/acsabm.4c01509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 01/30/2025] [Accepted: 02/06/2025] [Indexed: 02/28/2025]
Abstract
Recreating the structural and mechanical properties of native tissues in vitro presents significant challenges, particularly in mimicking the dense fibrillar network of extracellular matrixes such as skin and tendons. This study develops a reversible collagen film through cycling collagen self-assembly and disassembly, offering an innovative approach to address these challenges. We first generated an engineered collagen scaffold by applying plastic compression to the collagen hydrogel. The reversibility of the collagen assembly was explored by treating the scaffold with lactic acid, leading to its breakdown into an amorphous gel─a process termed defibrillogenesis. Subsequent immersion of this gel in phosphate buffer facilitated the reassembly of collagen into fibrils larger than those in the original scaffold yet with the D-banding pattern characteristic of collagen fibrils. Transfer learning of the mobileNetV2 convolutional neural network trained on atomic force microscope images of collagen nanoscale D-banding patterns was created with 99% training and testing accuracy. In addition, extensive external validation was performed, and the model achieved high robustness and generalization with unseen data sets. Further innovation was introduced by applying collagen hybridizing peptides, which significantly accelerated and directed the assembly of collagen fibrils, promoting a more organized and aligned fibrillar structure. This study not only demonstrates the feasibility of creating a reversible collagen film that closely mimics the density and structural properties of the native matrix but also highlights the potential of using collagen hybridizing peptides to control and enhance collagen fibrillogenesis. Our findings offer promising tissue engineering and regenerative medicine strategies by enabling precise manipulation of collagen structures in vitro.
Collapse
Affiliation(s)
- Sophia Huang
- Matrix
Functionalization and Phenotyping Lab, Faculty of Dentistry, University of Toronto, Toronto, Ontario M5G 1X3, Canada
| | - Nicole Ng
- Matrix
Functionalization and Phenotyping Lab, Faculty of Dentistry, University of Toronto, Toronto, Ontario M5G 1X3, Canada
| | - Mina Vaez
- Matrix
Functionalization and Phenotyping Lab, Faculty of Dentistry, University of Toronto, Toronto, Ontario M5G 1X3, Canada
| | - Boris Hinz
- Laboratory
of Tissue Repair and Regeneration, Keenan
Research Institute for Biomedical Science of the St. Michael’s
Hospital, Toronto, Ontario M5B 1M4, Canada
| | - Iona Leong
- Department
of Pathology and Laboratory Medicine, Mount
Sinai Hospital, Toronto, Ontario M5G 1X5, Canada
- Department
of Oral Pathology and Oral Medicine, Faculty of Dentistry, University of Toronto, Toronto, Ontario M5G 1X3, Canada
| | - Laurent Bozec
- Matrix
Functionalization and Phenotyping Lab, Faculty of Dentistry, University of Toronto, Toronto, Ontario M5G 1X3, Canada
| |
Collapse
|
15
|
Abubaker MA, Zhang D, Liu G, Ma H, He Y, Mala A, Li L, Al-Wraikat M, Liu Y. Polysaccharides as natural enhancers for meat quality, preservation, and protein functionality: A comprehensive review. Food Chem 2025; 468:142428. [PMID: 39693888 DOI: 10.1016/j.foodchem.2024.142428] [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: 09/14/2024] [Revised: 11/23/2024] [Accepted: 12/08/2024] [Indexed: 12/20/2024]
Abstract
Recent research focuses on developing meat products with health-promoting properties to reduce disease risk, particularly using natural polysaccharides due to their antioxidant and antibacterial effects. These polysaccharides, sourced from various materials, act through diverse structural mechanisms, inhibiting pathogen growth, enhancing oxidative stability, and improving meat flavor. This study highlights the role of meat proteins in achieving the Sustainable Development Goals (SDGs) and their importance in enhancing processed meat quality. It also examines the application of natural antioxidants and preservatives in meat processing. While some promising results demonstrate the potential of polysaccharides in meat science, their role in improving meat protein functions requires further investigation. Additionally, current solutions for improving meat quality face limitations, necessitating further research to reach industrial-scale applications. Thermal stability of meat proteins remains a critical factor throughout all stages of meat production, from processing and sterilization to consumption and preservation.
Collapse
Affiliation(s)
- Mohamed Aamer Abubaker
- College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710119, China; Department of Biology, Faculty of Education, University of Khartoum, Khartoum 11111, Sudan
| | - Duoduo Zhang
- College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710119, China
| | - Guanxu Liu
- College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710119, China
| | - Haorui Ma
- College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710119, China
| | - Yu He
- College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710119, China
| | - Azizza Mala
- Environmental, Natural Resource and Desertification Research Institute, National Center for Research, Ministry of High Education, Khartoum 11111, Sudan
| | - Linqiang Li
- College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710119, China
| | - Majida Al-Wraikat
- College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710119, China
| | - Yongfeng Liu
- College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710119, China
| |
Collapse
|
16
|
Giorno LP, Malmonge SM, Santos AR. Collagen as a biomaterial for skin wound healing: From structural characteristics to the production of devices for tissue engineering. Int J Artif Organs 2025; 48:135-145. [PMID: 39894968 DOI: 10.1177/03913988251316437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2025]
Abstract
Collagen is an abundant component in the human body and plays a fundamental role in the integrity and function of various tissues, including skin, bones, joints, and connective tissues. This natural polymer also contributes to physiological balance and individual health. Within this context, this article reviews the structure of collagen, describing intrinsic characteristics that range from its molecular composition to its organization into bundles. Additionally, the review highlights some of the applications of collagen in tissue engineering, particularly its mimicry of the skin's extracellular matrix. For this review, searches were performed in PubMed, Scopus, and Web of Sciences. The inclusion criteria were established based on the relevance of the studies for the objectives of the review and methodological quality. After selection of the articles, a critical analysis of their content was conducted and the information was synthesized and presented concisely. Analysis of the properties of collagen revealed its key importance for the design of bioactive materials in regenerative applications. However, challenges such as the need for improvement of the integration of implanted materials and a better understanding of the underlying biological processes remain.
Collapse
|
17
|
Michelutti L, Tel A, Robiony M, Vinayahalingam S, Agosti E, Ius T, Gagliano C, Zeppieri M. The Properties and Applicability of Bioprinting in the Field of Maxillofacial Surgery. Bioengineering (Basel) 2025; 12:251. [PMID: 40150715 PMCID: PMC11939734 DOI: 10.3390/bioengineering12030251] [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: 02/05/2025] [Revised: 02/24/2025] [Accepted: 02/28/2025] [Indexed: 03/29/2025] Open
Abstract
Perhaps the most innovative branch of medicine is represented by regenerative medicine. It deals with regenerating or replacing tissues damaged by disease or aging. The innovative frontier of this branch is represented by bioprinting. This technology aims to reconstruct tissues, organs, and anatomical structures, such as those in the head and neck region. This would mean revolutionizing therapeutic and surgical approaches in the management of multiple conditions in which a conspicuous amount of tissue is lost. The application of bioprinting for the reconstruction of anatomical areas removed due to the presence of malignancy would represent a revolutionary new step in personalized and precision medicine. This review aims to investigate recent advances in the use of biomaterials for the reconstruction of anatomical structures of the head-neck region, particularly those of the oral cavity. The characteristics and properties of each biomaterial currently available will be presented, as well as their potential applicability in the reconstruction of areas affected by neoplasia damaged after surgery. In addition, this study aims to examine the current limitations and challenges and to analyze the future prospects of this technology in maxillofacial surgery.
Collapse
Affiliation(s)
- Luca Michelutti
- Clinic of Maxillofacial Surgery, Head-Neck and NeuroScience Department, University Hospital of Udine, p.le S. Maria della Misericordia 15, 33100 Udine, Italy; (L.M.); (A.T.)
| | - Alessandro Tel
- Clinic of Maxillofacial Surgery, Head-Neck and NeuroScience Department, University Hospital of Udine, p.le S. Maria della Misericordia 15, 33100 Udine, Italy; (L.M.); (A.T.)
| | - Massimo Robiony
- Clinic of Maxillofacial Surgery, Head-Neck and NeuroScience Department, University Hospital of Udine, p.le S. Maria della Misericordia 15, 33100 Udine, Italy; (L.M.); (A.T.)
| | | | - Edoardo Agosti
- Division of Neurosurgery, Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, University of Brescia, Piazza Spedali Civili 1, 25123 Brescia, Italy
| | - Tamara Ius
- Academic Neurosurgery, Department of Neurosciences, University of Padova, 35121 Padova, Italy
| | - Caterina Gagliano
- Department of Medicine and Surgery, University of Enna “Kore”, Piazza dell’Università, 94100 Enna, Italy
- Mediterranean Foundation “G.B. Morgagni”, 95125 Catania, Italy
| | - Marco Zeppieri
- Department of Ophthalmology, University Hospital of Udine, 33100 Udine, Italy
- Department of Medicine, Surgery and Health Sciences, University of Trieste, 34100 Trieste, Italy
| |
Collapse
|
18
|
Rodriguez Sala M, Skalli O, Chandrasekaran S, Worsley M, Leventis N, Sabri F. Influence of aerogel mechanical properties on collagen micromorphology and its architecture. SOFT MATTER 2025; 21:1555-1570. [PMID: 39888334 DOI: 10.1039/d4sm01158a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2025]
Abstract
Previously, we demonstrated the promise of aerogels for the repair of nerve injuries as neural cells extend longer processes (neurites) when grown on aerogels compared to a control surface. We also reported that the aerogel surface topography influenced neurite length. Neurite extension may be boosted by depositing collagen on the aerogel prior to plating the cells. Indeed, collagen has many applications in biomaterials for nerve repair because it profoundly influences cellular properties such as shape and motility. Using collagen to enhance neurite extension requires knowing the effect of collagen deposition on the aerogel surface profile as well as how the aerogel's surface topography influences collagen organization into fibers or films. Herein, we have examined by SEM and profilometry the reciprocal relationship between collagen micromorphology and aerogel surface features including pore diameters, surface roughness, and Young's modulus (Y). Using 5 types of aerogels differing from each other by these parameters, we show that increasing the collagen surface concentration from 4 to 20 μg cm-2 leads to a gradual transition in collagen architecture from discrete fibers to films where individual fibers were not discernible. The collagen surface concentration at which deposited collagen changes from filaments to films (transition point, T.P.) was strongly dependent on aerogel physical properties as it increased with increasing pore diameter and surface roughness, while Y had little effect. These results provide a practical framework to customize the organization of collagen fibers on scaffolds for biomedical applications.
Collapse
Affiliation(s)
- Martina Rodriguez Sala
- Department of Physics and Material Science, The University of Memphis, Memphis, Tennessee, 38152, USA.
| | - Omar Skalli
- Department of Biological Sciences, The University of Memphis, Memphis, Tennessee, 38152, USA.
| | | | - Marcus Worsley
- Lawrence Livermore National Laboratory, Livermore, CA, 94550, USA.
| | | | - Firouzeh Sabri
- Department of Physics and Material Science, The University of Memphis, Memphis, Tennessee, 38152, USA.
| |
Collapse
|
19
|
Dutra Alves NS, Reigado GR, Santos M, Caldeira IDS, Hernandes HDS, Freitas-Marchi BL, Zhivov E, Chambergo FS, Nunes VA. Advances in regenerative medicine-based approaches for skin regeneration and rejuvenation. Front Bioeng Biotechnol 2025; 13:1527854. [PMID: 40013305 PMCID: PMC11861087 DOI: 10.3389/fbioe.2025.1527854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Accepted: 01/20/2025] [Indexed: 02/28/2025] Open
Abstract
Significant progress has been made in regenerative medicine for skin repair and rejuvenation. This review examines core technologies including stem cell therapy, bioengineered skin substitutes, platelet-rich plasma (PRP), exosome-based therapies, and gene editing techniques like CRISPR. These methods hold promise for treating a range of conditions, from chronic wounds and burns to age-related skin changes and genetic disorders. Challenges remain in optimizing these therapies for broader accessibility and ensuring long-term safety and efficacy.
Collapse
Affiliation(s)
- Nathalia Silva Dutra Alves
- Laboratory of Skin Physiology and Tissue Bioengineering, School of Arts, Sciences and Humanities, University of São Paulo, São Paulo, Brazil
| | - Gustavo Roncoli Reigado
- Laboratory of Skin Physiology and Tissue Bioengineering, School of Arts, Sciences and Humanities, University of São Paulo, São Paulo, Brazil
| | - Mayara Santos
- Laboratory of Skin Physiology and Tissue Bioengineering, School of Arts, Sciences and Humanities, University of São Paulo, São Paulo, Brazil
| | - Izabela Daniel Sardinha Caldeira
- Laboratory of Skin Physiology and Tissue Bioengineering, School of Arts, Sciences and Humanities, University of São Paulo, São Paulo, Brazil
| | - Henrique dos Santos Hernandes
- Laboratory of Proteins and Biotechnology, School of Arts, Sciences and Humanities, University of São Paulo, São Paulo, Brazil
| | | | - Elina Zhivov
- Wound Healing and Regenerative Medicine Research Program, Dr. Phillip Frost Department of Dermatology and Cutaneous Surgery, University of Miami Miller Medical School, Miami, FL, United States
| | - Felipe Santiago Chambergo
- Laboratory of Proteins and Biotechnology, School of Arts, Sciences and Humanities, University of São Paulo, São Paulo, Brazil
| | - Viviane Abreu Nunes
- Laboratory of Skin Physiology and Tissue Bioengineering, School of Arts, Sciences and Humanities, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
20
|
Zuo R, Li H, Cai C, Xia W, Liu J, Li J, Xu Y, Zhang Y, Li C, Wu Y, Zhang C. Autophagy modulates tenogenic differentiation of cartilage-derived stem cells in response to mechanical tension via FGF signaling. Stem Cells Transl Med 2025; 14:szae085. [PMID: 39673221 DOI: 10.1093/stcltm/szae085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2024] [Accepted: 10/01/2024] [Indexed: 12/16/2024] Open
Abstract
BACKGROUND In our previous study, we demonstrated that cartilage-derived stem cells (CDSCs) possess multi-differentiation potential, enabling direct bone-to-tendon structure regeneration after transplantation in a rat model. Therefore, the objective of this study is to investigate whether CDSCs are a suitable candidate for achieving biological regeneration of tendon injuries. METHODS Tenogenic differentiation was evaluated through cell morphology observation, PCR, and Western blot (WB) analysis. Autophagic flux, transmission electron microscopy, and WB analysis were employed to elucidate the role of autophagy during CDSC tenogenic differentiation. Cell survival and tenogenesis of transplanted CDSCs were assessed using fluorescence detection of gross and frozen section images. Heterotopic ossification and quality of tendon healing were evaluated by immunofluorescence, hematoxylin-eosin (H&E), and Safrinin O/Fast Green stains. RESULTS We found autophagy is activated in CDSCs when treated with cyclic tensile stress, which facilitates the preservation of their chondrogenic potential while impeding tenogenic differentiation. Inhibiting autophagy with chloroquine promoted tenogenic differentiation of CDSCs in response to cyclic tensile stress through activation of the Fgf2/Fgfr2 signaling pathway. This mechanism was further validated by 2 mouse transplantation models, revealed that autophagy inhibition could enhance the tendon regeneration efficacy of transplanted CDSCs at the patellar tendon resection site. CONCLUSION Our findings provide insights into CDSC transplantation for achieving biological regeneration of tendon injuries, and demonstrate how modulation of autophagy in CDSCs can promote tenogenic differentiation in response to tensile stress both in vivo and in vitro.
Collapse
Affiliation(s)
- Rui Zuo
- Department of Orthopedics, Xinqiao Hospital, Army Medical University, Chongqing 400037, People's Republic of China
| | - Haoke Li
- Department of Orthopedics, Xinqiao Hospital, Army Medical University, Chongqing 400037, People's Republic of China
| | - Chenhui Cai
- Department of Orthopedics, Xinqiao Hospital, Army Medical University, Chongqing 400037, People's Republic of China
| | - Wen Xia
- Department of Orthopedics, Xinqiao Hospital, Army Medical University, Chongqing 400037, People's Republic of China
| | - Jiabin Liu
- Department of Orthopedics, Xinqiao Hospital, Army Medical University, Chongqing 400037, People's Republic of China
| | - Jie Li
- Department of Orthopedics, Xinqiao Hospital, Army Medical University, Chongqing 400037, People's Republic of China
| | - Yuan Xu
- Department of Orthopedics, Xinqiao Hospital, Army Medical University, Chongqing 400037, People's Republic of China
| | - Yi Zhang
- Chongqing International Institute for Immunology, Chongqing 401320, People's Republic of China
| | - Changqing Li
- Department of Orthopedics, Xinqiao Hospital, Army Medical University, Chongqing 400037, People's Republic of China
| | - Yuzhang Wu
- Institute of Immunology, Army Medical University, Chongqing 400038, People's Republic of China
| | - Chao Zhang
- Department of Orthopedics, Xinqiao Hospital, Army Medical University, Chongqing 400037, People's Republic of China
| |
Collapse
|
21
|
Gansevoort M, Wentholt S, Li Vecchi G, de Vries M, Versteeg EMM, Boekema BKHL, Choppin A, Barritault D, Chiappini F, van Kuppevelt TH, Daamen WF. Next-Generation Biomaterials for Wound Healing: Development and Evaluation of Collagen Scaffolds Functionalized with a Heparan Sulfate Mimic and Fibroblast Growth Factor 2. J Funct Biomater 2025; 16:51. [PMID: 39997585 PMCID: PMC11856099 DOI: 10.3390/jfb16020051] [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: 12/20/2024] [Revised: 01/27/2025] [Accepted: 01/28/2025] [Indexed: 02/26/2025] Open
Abstract
Fibrosis after full-thickness wound healing-especially after severe burn wounds-remains a clinically relevant problem. Biomaterials that mimic the lost dermal extracellular matrix have shown promise but cannot completely prevent scar formation. We present a novel approach where porous type I collagen scaffolds were covalently functionalized with ReGeneRating Agent (RGTA®) OTR4120. RGTA® is a glycanase-resistant heparan sulfate mimetic that promotes regeneration when applied topically to chronic wounds. OTR4120 is able to capture fibroblast growth factor 2 (FGF-2), a heparan/heparin-binding growth factor that inhibits the activity of fibrosis-driving myofibroblasts. Scaffolds with various concentrations and distributions of OTR4120 were produced. When loaded with FGF-2, collagen-OTR4120 scaffolds demonstrated sustained release of FGF-2 compared to collagen-heparin scaffolds. Their anti-fibrotic potential was investigated in vitro by seeding primary human dermal fibroblasts on the scaffolds followed by stimulation with transforming growth factor β1 (TGF-β1) to induce myofibroblast differentiation. Collagen-OTR4120(-FGF-2) scaffolds diminished the gene expression levels of several myofibroblast markers. In absence of FGF-2 the collagen-OTR4120 scaffolds displayed an inherent anti-fibrotic effect, as the expression of two fibrotic markers (TGF-β1 and type I collagen) was diminished. This work highlights the potential of collagen-OTR4120 scaffolds as biomaterials to improve skin wound healing.
Collapse
Affiliation(s)
- Merel Gansevoort
- Department of Medical BioSciences, Research Institute for Medical Innovation, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
| | - Sabine Wentholt
- Department of Medical BioSciences, Research Institute for Medical Innovation, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
| | - Gaia Li Vecchi
- Department of Medical BioSciences, Research Institute for Medical Innovation, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
| | - Marjolein de Vries
- Department of Medical BioSciences, Research Institute for Medical Innovation, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
| | - Elly M. M. Versteeg
- Department of Medical BioSciences, Research Institute for Medical Innovation, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
| | - Bouke K. H. L. Boekema
- Burn Research Lab, Alliance of Dutch Burn Care, 1941 AJ Beverwijk, The Netherlands
- Department of Plastic, Reconstructive and Hand Surgery, Amsterdam UMC Location Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
| | | | | | | | - Toin H. van Kuppevelt
- Department of Medical BioSciences, Research Institute for Medical Innovation, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
| | - Willeke F. Daamen
- Department of Medical BioSciences, Research Institute for Medical Innovation, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
| |
Collapse
|
22
|
Mohite P, Puri A, Munde S, Dave R, Khan S, Patil R, Singh AK, Tipduangta P, Singh S, Chittasupho C. Potential of Chitosan/Gelatin-Based Nanofibers in Delivering Drugs for the Management of Varied Complications: A Review. Polymers (Basel) 2025; 17:435. [PMID: 40006097 PMCID: PMC11859051 DOI: 10.3390/polym17040435] [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: 12/31/2024] [Revised: 02/01/2025] [Accepted: 02/03/2025] [Indexed: 02/27/2025] Open
Abstract
Drug delivery systems have revolutionized traditional drug administration methods by addressing various challenges, such as enhancing drug solubility, prolonging effectiveness, minimizing adverse effects, and preserving potency. Nanotechnology-based drug delivery systems, particularly nanoparticles (NPs) and nanofibers (NFs), have emerged as promising solutions for biomedicine delivery. NFs, with their ability to mimic the porous and fibrous structures of biological tissues, have garnered significant interest in drug-delivering applications. Biopolymers such as gelatin (Ge) and chitosan (CH) have gained much more attention due to their biocompatibility, biodegradability, and versatility in biomedical applications. CH exhibits exceptional biocompatibility, anti-bacterial activity, and wound healing capabilities, whereas Ge provides good biocompatibility and cell adhesion properties. Ge/CH-based NFs stimulate cellular connections and facilitate tissue regeneration owing to their structural resemblance to the extracellular matrix. This review explores the additive methods of preparation, including electrospinning, force pinning, and template synthesis, focusing on electrospinning and the factors influencing the fiber structure. The properties of Ge and CH, their role in drug release, formulation strategies, and characterization techniques for electrospun fibers are discussed. Furthermore, this review addresses applications in delivering active moieties in the management of orthopedics and wound healing with regulatory considerations, along with challenges related to them. Thus, the review aims to provide a comprehensive overview of the potential of Ge/CH-based NFs for drug delivery and biomedical applications.
Collapse
Affiliation(s)
- Popat Mohite
- AETs St. John Institute of Pharmacy and Research, Palghar 401404, Maharashtra, India; (P.M.); (A.P.); (S.M.); (R.D.); (S.K.); (R.P.)
| | - Abhijeet Puri
- AETs St. John Institute of Pharmacy and Research, Palghar 401404, Maharashtra, India; (P.M.); (A.P.); (S.M.); (R.D.); (S.K.); (R.P.)
| | - Shubham Munde
- AETs St. John Institute of Pharmacy and Research, Palghar 401404, Maharashtra, India; (P.M.); (A.P.); (S.M.); (R.D.); (S.K.); (R.P.)
| | - Roshan Dave
- AETs St. John Institute of Pharmacy and Research, Palghar 401404, Maharashtra, India; (P.M.); (A.P.); (S.M.); (R.D.); (S.K.); (R.P.)
| | - Showkhiya Khan
- AETs St. John Institute of Pharmacy and Research, Palghar 401404, Maharashtra, India; (P.M.); (A.P.); (S.M.); (R.D.); (S.K.); (R.P.)
| | - Riteshkumar Patil
- AETs St. John Institute of Pharmacy and Research, Palghar 401404, Maharashtra, India; (P.M.); (A.P.); (S.M.); (R.D.); (S.K.); (R.P.)
| | - Anil Kumar Singh
- United Institute of Pharmacy, Prayagraj 211010, Uttar Pradesh, India;
| | - Pratchaya Tipduangta
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Chiang Mai University, Chiang Mai 50200, Thailand;
| | - Sudarshan Singh
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Chiang Mai University, Chiang Mai 50200, Thailand;
- Office of Research Administration, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Chuda Chittasupho
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Chiang Mai University, Chiang Mai 50200, Thailand;
| |
Collapse
|
23
|
Martins RO, Cardoso AT, Borsatto JV, Lanças FM. Advances in green carbon-based biosorbents: From conventional to miniaturized sample preparation strategies. Talanta 2025; 283:127171. [PMID: 39515052 DOI: 10.1016/j.talanta.2024.127171] [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: 08/08/2024] [Revised: 11/02/2024] [Accepted: 11/04/2024] [Indexed: 11/16/2024]
Abstract
Developing novel sorbent phases has advanced solid-based sample preparation techniques, improving analytical performance in complex matrices. Carbon-based sorbents, known for their high surface area, thermal and mechanical stability, and modifiability due to abundant organic functional groups, have emerged as exceptional materials in this field. Due to their versatile characteristics, carbon-based materials have been extensively investigated as promising materials for anchoring and functionalization with biopolymers, resulting in innovative hybrid materials, so-called carbon-based biosorbents. These biosorbents offer numerous advantages, including enhanced physicochemical properties and biodegradability, which help reduce the environmental impact of their synthesis, particularly when compared to conventional synthetic sorbent production methods that lack adherence to environmentally sustainable protocols. Among the various biopolymers used for modification, chitosan, starch, cyclodextrin, cellulose, and agarose have been identified as promising candidates for integration with carbon-based materials. In light of the ongoing advancements in developing novel carbon-based biosorbent materials, this review aims to highlight their synthesis using these biopolymers and examine their application in conventional and miniaturized sample preparation techniques across diverse matrices.
Collapse
Affiliation(s)
- Rafael Oliveira Martins
- Universidade de São Paulo, Instituto de Química de São Carlos, 13566-590, São Carlos, SP, Brazil
| | | | - João Victor Borsatto
- Universidade de São Paulo, Instituto de Química de São Carlos, 13566-590, São Carlos, SP, Brazil
| | - Fernando Mauro Lanças
- Universidade de São Paulo, Instituto de Química de São Carlos, 13566-590, São Carlos, SP, Brazil.
| |
Collapse
|
24
|
Adhikari M, Bakadia BM, Wang L, Li Y, Shi Z, Yang G. Electricallymodified bacterial cellulose tailored with plant based green materials for infected wound healing applications. BIOMATERIALS ADVANCES 2025; 167:214087. [PMID: 39481142 DOI: 10.1016/j.bioadv.2024.214087] [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: 08/14/2024] [Revised: 10/10/2024] [Accepted: 10/22/2024] [Indexed: 11/02/2024]
Abstract
Effective treatment of infected wounds remains a challenge due to the rise of antibiotic-resistant microorganisms. The development of advanced materials with strong antimicrobial properties is necessary to address this issue. In this study, a unique composite of electrically modified bacterial cellulose (EBC) with allantoin (ABC) and zein was developed by dipping diffusion method. Morphological structural analysis revealed a uniform distribution of zein and aligned fibers, confirming the synthesis of the ABC-Zein composite. The formation of ABC-Zein was further confirmed by attenuated total reflection-Fourier transform infrared (ATR-FTIR), which displayed additional peaks corresponding to EBC, indicating the incorporation of zein into ABC. X-ray diffraction (XRD) analysis of ABC-Zein demonstrated a similar crystalline structure with EBC. The ABC-Zein showed mechanical integrity (tensile strength: 1.15 ± 0.21 MPa), thermal stability (degradation temperature: 290 °C), porous structure (porosity: 40.23 ± 0.21 %), and hydrophilic (water contact angle: 53.3 ± 5.3°) properties. Furthermore, the antimicrobial agent terpinen-4-ol (T4O), derived from tea tree oil, was incorporated into the ABC-Zein composite. Biological studies confirmed the antimicrobial efficacy (Staphylococcus aureus inhibition: 88.5 ± 7.19 %) and biocompatible (cell viability: 84.95 ± 5.6 %, hemolysis: 4.479 ± 0.39 %) nature of the T4O-ABC-Zein composite. The combined effects of the aligned fiber structure, zein protein, and antimicrobial T4O significantly enhanced infected wound healing by day 7, promoting inflammatory response, granular tissue formation, cell proliferation, and angiogenesis. By day 14, T4O-ABC-Zein facilitated complete wound healing, with reepithelization, collagen I deposition, and downregulation of CD 31, Ki67, and α-SMA. Overall, the innovative T4O-ABC-Zein composite, with an aligned fiber structure, improved biocompatibility, and antimicrobial properties, holds significant potential for the treatment of infected wounds.
Collapse
Affiliation(s)
- Manjila Adhikari
- Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Bianza Moise Bakadia
- Innovation Research Center for AIE Pharmaceutical Biology, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target and Clinical Pharmacology, The NMPA and State Key Laboratory of Respiratory Disease School of Pharmaceutical Sciences and The Fifth Affiliated Hospital Guangzhou Medical University, Guangzhou 511436, China
| | - Li Wang
- Wuhan Branch of the National Science Library, Chinese Academy of Sciences, Wuhan 430071, China
| | - Ying Li
- Innovation Research Center for AIE Pharmaceutical Biology, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target and Clinical Pharmacology, The NMPA and State Key Laboratory of Respiratory Disease School of Pharmaceutical Sciences and The Fifth Affiliated Hospital Guangzhou Medical University, Guangzhou 511436, China
| | - Zhijun Shi
- Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China.
| | - Guang Yang
- Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China.
| |
Collapse
|
25
|
Khumaidi A, Murwanti R, Damayanti E, Hertiani T. Empirical use, phytochemical, and pharmacological effects in wound healing activities of compounds in Diospyros leaves: A review of traditional medicine for potential new plant-derived drugs. JOURNAL OF ETHNOPHARMACOLOGY 2025; 337:118966. [PMID: 39427738 DOI: 10.1016/j.jep.2024.118966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 09/23/2024] [Accepted: 10/17/2024] [Indexed: 10/22/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Wound healing extracts' activity is increasingly being studied in the field of traditional medicine. Among medicinal plants, Diospyros is known to have healing effects on wounds, along with activities such as anti-biofilm, anti-inflammatory, antibacterial, antioxidant, and regulation of the immune system. However, the current use of the leaves could be more optimal, and the scientific basis needs to be improved. AIM OF THIS REVIEW This review aimed to critically examine the literature on the traditional use and bioactive metabolites of several Diospyros species, demonstrating the significant potential in wound healing, antibacterial, anti-biofilm, regulatory effect on the immune system, anti-inflammatory, and antioxidant activities. The critical analysis was conducted to provide robust perspectives and recommendations for future studies on the use of Diospyros potential resources of wound healing material, including related activities. MATERIALS AND METHODS Exploratory studies on Diospyros species over the past 20 years were examined, with a focus on general information, practical use, secondary metabolite, and pharmacological activities related to wound healing. Data were meticulously collected from scientific databases including Scopus, ScienceDirect, Web of Science, Taylor & Francis, Google Scholar, PubMed as well as various botanical and biodiversity sources. Furthermore, manual searches were conducted to ensure comprehensive coverage. Reference manager software was used to manage articles and remove duplicates, then the gathered data were summarized and verified, ensuring the thoroughness and validity of the review process. RESULTS The results showed that Diospyros leaves have great potential to be harnessed as herbal medications, evidenced by both scientific findings and community uses. Various substances, including flavonoids, coumarins, tannins, terpenoids, steroids, lignans, quinones, and secoiridoids were identified. Chemical compound investigations in both in vivo and in vitro studies of Diospyros leaves reported wound healing activity, as well as antibacterial, anti-inflammatory, anti-biofilm, antioxidant, and immunomodulatory properties. CONCLUSION The review highlights the traditional uses and bioactive metabolites of Diospyros species in wound healing, identifying various beneficial compounds such as flavonoids and tannins. These compounds demonstrate various therapeutic effects, including antibacterial, anti-biofilm, anti-inflammatory, antioxidant, and immunomodulatory activities. Diospyros leaf extracts have a favorable safety profile, but further studies, including in vivo investigations and clinical trials, are necessary to confirm their efficacy and safety for clinical applications. Diospyros leaf extracts have significant potential for the development of wound healing substances due to the wide range of bioactivities targeting various stages of wound healing.
Collapse
Affiliation(s)
- Akhmad Khumaidi
- Doctoral Program in Pharmaceutical Science, Faculty of Pharmacy, Universitas Gadjah Mada, Yogyakarta, 55281, Indonesia; Department of Pharmacy, Faculty of Mathematics and Natural Sciences, Universitas Tadulako, Palu, 94118, Indonesia
| | - Retno Murwanti
- Department of Pharmacology & Clinical Pharmacy, Faculty of Pharmacy, Universitas Gadjah Mada, Yogyakarta, 55281, Indonesia
| | - Ema Damayanti
- Research Center for Food Technology and Processing, National Research and Innovation Agency (BRIN), Gunungkidul, 55861, Indonesia
| | - Triana Hertiani
- Department of Pharmaceutical Biology, Faculty of Pharmacy, Universitas Gadjah Mada, Yogyakarta, 55281, Indonesia.
| |
Collapse
|
26
|
Kalugin PN, Soden PA, Massengill CI, Amsalem O, Porniece M, Guarino DC, Tingley D, Zhang SX, Benson JC, Hammell MF, Tong DM, Ausfahl CD, Lacey TE, Courtney Y, Hochstetler A, Lutas A, Wang H, Geng L, Li G, Li B, Li Y, Lehtinen MK, Andermann ML. Simultaneous, real-time tracking of many neuromodulatory signals with Multiplexed Optical Recording of Sensors on a micro-Endoscope. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.26.634931. [PMID: 39896634 PMCID: PMC11785251 DOI: 10.1101/2025.01.26.634931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2025]
Abstract
Dozens of extracellular molecules jointly impact a given neuron, yet we lack methods to simultaneously record many such signals in real time. We developed a probe to track ten or more neuropeptides and neuromodulators using spatial multiplexing of genetically encoded fluorescent sensors. Cultured cells expressing one sensor at a time are immobilized at the front of a gradient refractive index (GRIN) lens for 3D two-photon imaging in vitro and in vivo . The sensor identity and detection sensitivity of each cell are determined via robotic dipping of the probe into wells containing various ligands and concentrations. Using this probe, we detected stimulation-evoked release of multiple neuromodulators in acute brain slices. We also tracked endogenous and drug-evoked changes in cerebrospinal fluid composition in the awake mouse lateral ventricle, which triggered downstream activation of the choroid plexus epithelium. Our approach offers a first step towards quantitative, real-time, high-dimensional tracking of brain fluid composition.
Collapse
|
27
|
Razif R, Fadilah NIM, Ahmad H, Looi Qi Hao D, Maarof M, Fauzi MB. Asiaticoside-Loaded Multifunctional Bioscaffolds for Enhanced Hyperglycemic Wound Healing. Biomedicines 2025; 13:277. [PMID: 40002691 PMCID: PMC11853099 DOI: 10.3390/biomedicines13020277] [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: 12/14/2024] [Revised: 01/13/2025] [Accepted: 01/20/2025] [Indexed: 02/27/2025] Open
Abstract
The review explores the potential of asiaticoside-loaded bioscaffolds to improve the management of hyperglycemic wounds, particularly diabetic foot ulcers (DFUs). Asiaticoside, sourced from Centella asiatica, possesses properties that address DFUs' healing challenges: insufficient angiogenesis, persistent inflammation, and delayed tissue regeneration. By incorporating asiaticoside into bioscaffold 3D designs including hydrogels, microneedle arrays, and nanofibrous meshes, therapeutic efficacy is optimized. This review examines the mechanisms of asiaticoside in wound healing (collagen production, angiogenesis modulation, inflammation reduction, and cell migration and proliferation) based on in vitro and in vivo studies. Asiaticoside also demonstrates synergistic abilities with other biomaterials, creating the possibility of more effective therapies. While preclinical research is promising, clinical trials are crucial to evaluate the efficacy and safety of asiaticoside-loaded bioscaffolds in patients with DFUs. Asiaticoside-loaded bioscaffolds are a significant development in wound healing and may aid in treating hyperglycemic wound complications. Their ability to offer individualized treatment plans has the potential to enhance the quality of life of those who suffer from diabetes. This review is based on a thorough literature search (2019-2024) across multiple databases, excluding secondary literature and non-English articles.
Collapse
Affiliation(s)
- Raniya Razif
- Department of Tissue Engineering and Regenerative Medicine (DTERM), Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras 56000, Kuala Lumpur, Malaysia; (R.R.); (N.I.M.F.); (M.M.)
| | - Nur Izzah Md Fadilah
- Department of Tissue Engineering and Regenerative Medicine (DTERM), Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras 56000, Kuala Lumpur, Malaysia; (R.R.); (N.I.M.F.); (M.M.)
- Advance Bioactive Materials-Cells UKM Research Group, Universiti Kebangsaan Malaysia, Bangi 43600, Selangor, Malaysia
| | - Haslina Ahmad
- Integrated Chemical Biophysics Research, Universiti Putra Malaysia (UPM), Serdang 43400, Selangor, Malaysia;
- Department of Chemistry, Faculty of Science, Universiti Putra Malaysia (UPM), Serdang 43400, Selangor, Malaysia
| | - Daniel Looi Qi Hao
- My Cytohealth Sdn Bhd, Hive 5, Taman Teknologi, MRANTI, Bukit Jalil 57000, Kuala Lumpur, Malaysia;
| | - Manira Maarof
- Department of Tissue Engineering and Regenerative Medicine (DTERM), Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras 56000, Kuala Lumpur, Malaysia; (R.R.); (N.I.M.F.); (M.M.)
- Advance Bioactive Materials-Cells UKM Research Group, Universiti Kebangsaan Malaysia, Bangi 43600, Selangor, Malaysia
| | - Mh Busra Fauzi
- Department of Tissue Engineering and Regenerative Medicine (DTERM), Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras 56000, Kuala Lumpur, Malaysia; (R.R.); (N.I.M.F.); (M.M.)
- Advance Bioactive Materials-Cells UKM Research Group, Universiti Kebangsaan Malaysia, Bangi 43600, Selangor, Malaysia
| |
Collapse
|
28
|
de Souza A, S. Martignago CC, Assis L, Vieira Botelho Delpupo F, Assis M, S. J. Sousa K, Souza e Silva LC, Líbero LO, de Oliveira F, Renno ACM. Casting Skin Dressing Containing Extractions of the Organic Part of Marine Sponges for Wound Healing. ACS APPLIED BIO MATERIALS 2025; 8:617-627. [PMID: 39705707 PMCID: PMC11752525 DOI: 10.1021/acsabm.4c01497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2024] [Revised: 12/06/2024] [Accepted: 12/09/2024] [Indexed: 12/22/2024]
Abstract
Skin wounds are extremely frequent injuries related to many etiologies. They are a burden on healthcare systems worldwide. Skin dressings are the most popular therapy, and collagen is the most commonly used biomaterial, although new sources of collagen have been studied, especially spongin-like from marine sponges (SPG), as a promising source due to a similar composition to vertebrates and the ability to function as a cell-matrix adhesion framework. Despite evidence showing the positive effects of SPG for tissue healing, the effects of skin dressings manufactured are still limited. In this context, this study aimed at investigating the effects of collagen skin dressings in an experimental model of skin wounds in rats. For this purpose, SEM, FTIR, cell viability, morphological and morphometric aspects, collagen deposition, and immunostaining of TGF-β and FGF were evaluated. The results demonstrated micro- and macropores on the rough surface, peak characteristics of collagen, and no cytotoxicity for the skin dressing. Also, the control group (CG) after 5 and 10 days exhibited an intense inflammatory process and the presence of granulation tissue, while the treated group (TG) exhibited re-epithelialization after 10 days. The evaluation of granulation tissue and neoepithelial length had an intragroup statistical difference (p = 0.0216) and no intergroup difference. Birefringence demonstrated an organized mesh arranged in a network pattern, presenting type I and type III collagen fibers in all groups. Moreover, in the morphometric evaluation, there were no statistical differences in intergroups or time points for the different types of collagen evaluated. In conclusion, these findings may indicate that the dressing has not exacerbated the inflammatory process and may allow faster healing. However, further studies using a critical wound healing injury model should be used, associated with longer experimental periods of evaluation, to further investigate the effects of these promising therapeutic approaches throughout the skin repair process.
Collapse
Affiliation(s)
- Amanda de Souza
- Department
of Biosciences, Federal University of São
Paulo (UNIFESP), Lab 342, 136 Silva Jardim Street, Santos, SP 11015020, Brazil
| | - Cintia C. S. Martignago
- Department
of Biosciences, Federal University of São
Paulo (UNIFESP), Lab 342, 136 Silva Jardim Street, Santos, SP 11015020, Brazil
| | - Lívia Assis
- Scientific
Institute and Technological Department, University Brazil, São
Paulo-Itaquera, SP 04021-001,Brazil
| | - Fernanda Vieira Botelho Delpupo
- Department
of Biosciences, Federal University of São
Paulo (UNIFESP), Lab 342, 136 Silva Jardim Street, Santos, SP 11015020, Brazil
| | - Marcelo Assis
- Department
of Biosciences, Federal University of São
Paulo (UNIFESP), Lab 342, 136 Silva Jardim Street, Santos, SP 11015020, Brazil
| | - Karolyne S. J. Sousa
- Department
of Biosciences, Federal University of São
Paulo (UNIFESP), Lab 342, 136 Silva Jardim Street, Santos, SP 11015020, Brazil
| | - Lais Caroline Souza e Silva
- Department
of Biosciences, Federal University of São
Paulo (UNIFESP), Lab 342, 136 Silva Jardim Street, Santos, SP 11015020, Brazil
| | - Laura O. Líbero
- CDMF
- Department of Chemistry, Federal University
of São Carlos (UFSCar), Washington Luís Road, São Carlos, SP 13565-905, Brazil
| | - Flavia de Oliveira
- Department
of Biosciences, Federal University of São
Paulo (UNIFESP), Lab 342, 136 Silva Jardim Street, Santos, SP 11015020, Brazil
| | - Ana Claudia Muniz Renno
- Department
of Biosciences, Federal University of São
Paulo (UNIFESP), Lab 342, 136 Silva Jardim Street, Santos, SP 11015020, Brazil
| |
Collapse
|
29
|
Ersanli C, Skoufos I, Fotou K, Tzora A, Bayon Y, Mari D, Sarafi E, Nikolaou K, Zeugolis DI. Release Profile and Antibacterial Activity of Thymus sibthorpii Essential Oil-Incorporated, Optimally Stabilized Type I Collagen Hydrogels. Bioengineering (Basel) 2025; 12:89. [PMID: 39851363 PMCID: PMC11760836 DOI: 10.3390/bioengineering12010089] [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: 12/06/2024] [Revised: 01/09/2025] [Accepted: 01/16/2025] [Indexed: 01/26/2025] Open
Abstract
Antimicrobial resistance is one of the drastically increasing major global health threats due to the misuse and overuse of antibiotics as traditional antimicrobial agents, which render urgent the need for alternative and safer antimicrobial agents, such as essential oils (EOs). Although the strong antimicrobial activity of various EOs has already been studied and revealed, their characteristic high sensitivity and volatility drives the need towards a more efficient drug administration method via a biomaterial system. Herein, the potential of Thymus sibthorpii EO incorporated in functionalized antibacterial collagen hydrogels was investigated. At first, the optimally stabilized type I collagen hydrogels via six different multi-arm poly (ethylene glycol) succinimidyl glutarate (starPEG) crosslinkers were determined by assessing the free amine content and the resistance to enzymatic degradation. Subsequently, 0.5, 1, and 2% v/v of EO were incorporated into optimized collagen hydrogels, and the release profile, as well as release kinetics, were studied. Finally, biomaterial cytocompatibility tests were performed. Thymus sibthorpii EO was released from the hydrogel matrix via Fickian diffusion and showed sustained release and 0.5% v/v EO-loaded hydrogels showed adequate antibacterial activity against Staphylococcus aureus and did not show any statistically significant difference compared to penicillin (p < 0.05). Moreover, none of the fabricated composite antibacterial scaffolds displayed any cytotoxicity on NIH-3T3 fibroblasts. In conclusion, this work presents an innovative antibacterial biomaterial system for tissue engineering applications, which could serve as a promising alternative to antibiotics, contributing to coping with the issue of antimicrobial resistance.
Collapse
Affiliation(s)
- Caglar Ersanli
- Laboratory of Animal Science, Nutrition and Biotechnology, School of Agriculture, University of Ioannina, 47100 Arta, Greece; (C.E.); (I.S.)
- Laboratory of Animal Health, Food Hygiene and Quality, School of Agriculture, University of Ioannina, 47100 Arta, Greece; (K.F.); (K.N.)
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL), Charles Institute of Dermatology, Conway Institute of Biomolecular and Biomedical Research and School of Mechanical and Materials Engineering, University College Dublin (UCD), D04 V1W8 Dublin, Ireland;
| | - Ioannis Skoufos
- Laboratory of Animal Science, Nutrition and Biotechnology, School of Agriculture, University of Ioannina, 47100 Arta, Greece; (C.E.); (I.S.)
| | - Konstantina Fotou
- Laboratory of Animal Health, Food Hygiene and Quality, School of Agriculture, University of Ioannina, 47100 Arta, Greece; (K.F.); (K.N.)
| | - Athina Tzora
- Laboratory of Animal Health, Food Hygiene and Quality, School of Agriculture, University of Ioannina, 47100 Arta, Greece; (K.F.); (K.N.)
| | - Yves Bayon
- Medtronic—Sofradim Production, 116 Avenue du Formans—BP132, F-01600 Trevoux, France;
| | - Despoina Mari
- Department of Biological Applications & Technology, School of Health Sciences, University of Ioannina, 45110 Ioannina, Greece; (D.M.); (E.S.)
- Biomedical Research Institute, Foundation for Research and Technology-Hellas, 45110 Ioannina, Greece
| | - Eleftheria Sarafi
- Department of Biological Applications & Technology, School of Health Sciences, University of Ioannina, 45110 Ioannina, Greece; (D.M.); (E.S.)
- Biomedical Research Institute, Foundation for Research and Technology-Hellas, 45110 Ioannina, Greece
| | - Konstantina Nikolaou
- Laboratory of Animal Health, Food Hygiene and Quality, School of Agriculture, University of Ioannina, 47100 Arta, Greece; (K.F.); (K.N.)
| | - Dimitrios I. Zeugolis
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL), Charles Institute of Dermatology, Conway Institute of Biomolecular and Biomedical Research and School of Mechanical and Materials Engineering, University College Dublin (UCD), D04 V1W8 Dublin, Ireland;
| |
Collapse
|
30
|
Moss S, Shiwarski DJ, Feinberg AW. FRESH 3D Bioprinting of Collagen Types I, II, and III. ACS Biomater Sci Eng 2025; 11:556-563. [PMID: 39622052 PMCID: PMC11733922 DOI: 10.1021/acsbiomaterials.4c01826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2024] [Revised: 11/19/2024] [Accepted: 11/20/2024] [Indexed: 01/14/2025]
Abstract
Collagens play a vital role in the mechanical integrity of tissues as well as in physical and chemical signaling throughout the body. As such, collagens are widely used biomaterials in tissue engineering; however, most 3D fabrication methods use only collagen type I and are restricted to simple cast or molded geometries that are not representative of native tissue. Freeform reversible embedding of suspended hydrogel (FRESH) 3D bioprinting has emerged as a method to fabricate complex 3D scaffolds from collagen I but has yet to be leveraged for other collagen isoforms. Here, we developed collagen type II, collagen type III, and combination bioinks for FRESH 3D bioprinting of millimeter-sized scaffolds with micrometer scale features with fidelity comparable to scaffolds fabricated with the established collagen I bioink. At the microscale, single filament extrusions were similar across all collagen bioinks with a nominal diameter of ∼100 μm using a 34-gauge needle. Scaffolds as large as 10 × 10 × 2 mm were also fabricated and showed similar overall resolution and fidelity across collagen bioinks. Finally, cell adhesion and growth on the different collagen bioinks as either cast or FRESH 3D bioprinted scaffolds were compared and found to support similar growth behaviors. In total, our results expand the range of collagen isoform bioinks that can be 3D bioprinted and demonstrate that collagen types I, II, III, and combinations thereof can all be FRESH printed with high fidelity and comparable biological response. This serves to expand the toolkit for the fabrication of tailored collagen scaffolds that can better recapitulate the extracellular matrix properties of specific tissue types.
Collapse
Affiliation(s)
- Samuel
P Moss
- Department
of Biomedical Engineering, Carnegie Mellon
University, Pittsburgh, Pennsylvania 15213, United States of America
| | - Daniel J. Shiwarski
- Department
of Biomedical Engineering, Carnegie Mellon
University, Pittsburgh, Pennsylvania 15213, United States of America
| | - Adam W. Feinberg
- Department
of Biomedical Engineering, Carnegie Mellon
University, Pittsburgh, Pennsylvania 15213, United States of America
- Department
of Materials Science and Engineering, Carnegie
Mellon University, Pittsburgh, Pennsylvania 15213, United States of America
| |
Collapse
|
31
|
Li X, Xu Y, Zhou Z, Tang M, Cui J, Han W, Li J, Dai J, Ren X, Jiang H, Yu Y, Liu Q, Tang H, Xiao M. Self-Cross-Linked Collagen Sponge from the Alosa sapidissima Scale for Hemostasis and Wound Healing Applications. Biomacromolecules 2025; 26:405-414. [PMID: 39680041 DOI: 10.1021/acs.biomac.4c01211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2024]
Abstract
Type I collagen, a crucial component maintaining the structural integrity and physiological function of various tissues, is widely regarded as one of the most suitable biomaterials for healthcare applications. In this study, shad scales, used for treating ulcers, scalds, and burns in traditional Chinese medicine, were exploited for type I collagen extraction. After self-assembly into hydrogels, the extracted collagen was subsequently freeze-dried to form collagen sponges. The collagen sponge promoted rapid hemostasis, neovascularization, and immune regulation. Additionally, it accelerated the formation of granulation tissue, re-epithelialization, and collagen remodeling at the wound site in full-thickness skin wound rat models. Consequently, the shad scale collagen sponge holds great promise for the treatment of chronic wounds and skin regeneration. Notably, the shad was sourced from sustainably recirculating aquaculture systems (RAS) farms that adhere to the Traceable Management of Animal Products Safety, ensuring that the derived collagen possesses potential in the medical apparatus market.
Collapse
Affiliation(s)
- Xiaoyun Li
- Department of Cardiovascular Surgery of the First Affiliated Hospital and Institute for Cardiovascular Science, Suzhou Medical College, Soochow University, Suzhou 215000, China
| | - Yue Xu
- Department of Cardiovascular Surgery of the First Affiliated Hospital and Institute for Cardiovascular Science, Suzhou Medical College, Soochow University, Suzhou 215000, China
- Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Institute of Stomatology, Nanjing University, Nanjing 211166, China
| | - Zijun Zhou
- Department of Cardiovascular Surgery of the First Affiliated Hospital and Institute for Cardiovascular Science, Suzhou Medical College, Soochow University, Suzhou 215000, China
| | - Mingliang Tang
- Department of Cardiovascular Surgery of the First Affiliated Hospital and Institute for Cardiovascular Science, Suzhou Medical College, Soochow University, Suzhou 215000, China
| | - Jinjia Cui
- Department of Cardiovascular Surgery of the First Affiliated Hospital and Institute for Cardiovascular Science, Suzhou Medical College, Soochow University, Suzhou 215000, China
| | - Wenjing Han
- Department of Cardiovascular Surgery of the First Affiliated Hospital and Institute for Cardiovascular Science, Suzhou Medical College, Soochow University, Suzhou 215000, China
| | - Jingyi Li
- Department of Cardiovascular Surgery of the First Affiliated Hospital and Institute for Cardiovascular Science, Suzhou Medical College, Soochow University, Suzhou 215000, China
| | - Jing Dai
- Department of Cardiovascular Surgery of the First Affiliated Hospital and Institute for Cardiovascular Science, Suzhou Medical College, Soochow University, Suzhou 215000, China
| | - Xiaoyi Ren
- Department of Cardiovascular Surgery of the First Affiliated Hospital and Institute for Cardiovascular Science, Suzhou Medical College, Soochow University, Suzhou 215000, China
| | - Huihui Jiang
- Department of Cardiovascular Surgery of the First Affiliated Hospital and Institute for Cardiovascular Science, Suzhou Medical College, Soochow University, Suzhou 215000, China
| | - Yanzhen Yu
- Department of Biophysics, College of Basic Medical Sciences, Naval Medical University (Second Military Medical University), Shanghai 200433, China
| | - Qinghua Liu
- Suzhou Fishseeds Bio-Technology, LTD, Suzhou 215138, China
| | - Hongmei Tang
- Department of Plastic and Aesthetic Surgery, The Second Affiliated Hospital of Soochow University, Suzhou Medical College, Soochow University, Suzhou 215000, China
| | - Miao Xiao
- Department of Cardiovascular Surgery of the First Affiliated Hospital and Institute for Cardiovascular Science, Suzhou Medical College, Soochow University, Suzhou 215000, China
- Suzhou Fishseeds Bio-Technology, LTD, Suzhou 215138, China
| |
Collapse
|
32
|
Granados-Carrera CM, Castro-Criado D, Abdullah JAA, Jiménez-Rosado M, Perez-Puyana VM. Aerogels Based on Chitosan and Collagen Modified with Fe 2O 3 and Fe 3O 4 Nanoparticles: Fabrication and Characterization. Polymers (Basel) 2025; 17:133. [PMID: 39861206 PMCID: PMC11768276 DOI: 10.3390/polym17020133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Revised: 01/02/2025] [Accepted: 01/07/2025] [Indexed: 01/27/2025] Open
Abstract
The necessity to mitigate the intrinsic issues associated with tissue or organ transplants, in order to address the rising prevalence of diseases attributable to increased life expectancy, provides a rationale for the pursuit of innovation in the field of biomaterials. Specifically, biopolymeric aerogels represent a significant advancement in the field of tissue engineering, offering a promising solution for the formation of temporary porous matrices that can replace damaged tissues. However, the functional characteristics of these materials are inadequate, necessitating the implementation of matrix reinforcement methods to enhance their performance. In this study, chemical and green iron oxide nanoparticles, previously synthesized and documented in existing research, were incorporated into hybrid aerogels combining collagen (C) and chitosan (CH). The characterization of these aerogels was conducted through rheological, microstructural, and functional analyses. The results demonstrate that the incorporation of iron oxide nanoparticles has a significant influence on the properties of the aerogels fabricated with them. In particular, the incorporation of these nanoparticles has been observed to modify the mechanical properties, with an increase in strength and porosity that may support cell proliferation.
Collapse
Affiliation(s)
- Carmen Mª Granados-Carrera
- Department of Chemical Engineering, Faculty of Chemistry, University of Seville, 41012 Seville, Spain; (C.M.G.-C.); (D.C.-C.); (J.A.A.A.)
| | - Daniel Castro-Criado
- Department of Chemical Engineering, Faculty of Chemistry, University of Seville, 41012 Seville, Spain; (C.M.G.-C.); (D.C.-C.); (J.A.A.A.)
| | - Johar Amin Ahmed Abdullah
- Department of Chemical Engineering, Faculty of Chemistry, University of Seville, 41012 Seville, Spain; (C.M.G.-C.); (D.C.-C.); (J.A.A.A.)
| | - Mercedes Jiménez-Rosado
- Department of Applied Chemistry and Physics, Faculty of Biological and Ambiental Sciences, University of León, 24009 León, Spain
| | - Víctor M. Perez-Puyana
- Department of Engineering and Materials Science and Transportation, University of Seville, 41092 Seville, Spain
| |
Collapse
|
33
|
Dean J, Hoch C, Wollenberg B, Navidzadeh J, Maheta B, Mandava A, Knoedler S, Sherwani K, Baecher H, Schmitz A, Alfertshofer M, Heiland M, Kreutzer K, Koerdt S, Knoedler L. Advancements in bioengineered and autologous skin grafting techniques for skin reconstruction: a comprehensive review. Front Bioeng Biotechnol 2025; 12:1461328. [PMID: 39840132 PMCID: PMC11747595 DOI: 10.3389/fbioe.2024.1461328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Accepted: 12/03/2024] [Indexed: 01/23/2025] Open
Abstract
The reconstruction of complex skin defects challenges clinical practice, with autologous skin grafts (ASGs) as the traditional choice due to their high graft take rate and patient compatibility. However, ASGs have limitations such as donor site morbidity, limited tissue availability, and the necessity for multiple surgeries in severe cases. Bioengineered skin grafts (BSGs) aim to address these drawbacks through advanced tissue engineering and biomaterial science. This study conducts a systematic review to describe the benefits and shortcomings of BSGs and ASGs across wound healing efficacy, tissue integration, immunogenicity, and functional outcomes focusing on wound re-epithelialization, graft survival, and overall aesthetic outcomes. Preliminary findings suggest ASGs show superior early results, while BSGs demonstrate comparable long-term outcomes with reduced donor site morbidity. This comparative analysis enhances understanding of bioengineered alternatives in skin reconstruction, potentially redefining best practices based on efficacy, safety, and patient-centric outcomes, highlighting the need for further innovation in bioengineered solutions.
Collapse
Affiliation(s)
- Jillian Dean
- School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Cosima Hoch
- Department of Otolaryngology, Head and Neck Surgery, School of Medicine and Health, Technical University of Munich (TUM), Munich, Germany
| | - Barbara Wollenberg
- Department of Otolaryngology, Head and Neck Surgery, School of Medicine and Health, Technical University of Munich (TUM), Munich, Germany
| | - Justin Navidzadeh
- School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Bhagvat Maheta
- California Northstate University College of Medicine, Elk Grove, CA, United States
| | - Anisha Mandava
- School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Samuel Knoedler
- Institute of Regenerative Biology and Medicine, Helmholtz Zentrum München, Munich, Germany
| | - Khalil Sherwani
- Institute of Regenerative Biology and Medicine, Helmholtz Zentrum München, Munich, Germany
| | - Helena Baecher
- Institute of Regenerative Biology and Medicine, Helmholtz Zentrum München, Munich, Germany
| | - Alina Schmitz
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Department of Oral and Maxillofacial Surgery, Berlin, Germany
| | - Michael Alfertshofer
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Department of Oral and Maxillofacial Surgery, Berlin, Germany
| | - Max Heiland
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Department of Oral and Maxillofacial Surgery, Berlin, Germany
| | - Kilian Kreutzer
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Department of Oral and Maxillofacial Surgery, Berlin, Germany
| | - Steffen Koerdt
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Department of Oral and Maxillofacial Surgery, Berlin, Germany
| | - Leonard Knoedler
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Department of Oral and Maxillofacial Surgery, Berlin, Germany
| |
Collapse
|
34
|
Yuri P, Christinus H, Alfarizi ZY, Ndoye M. The ratio of COL2A1:COL1A1 in dartos tissue patients with hypospadias. BMC Urol 2025; 25:2. [PMID: 39755652 DOI: 10.1186/s12894-024-01688-1] [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: 02/25/2024] [Accepted: 12/26/2024] [Indexed: 01/06/2025] Open
Abstract
BACKGROUND The inelasticity of dartos tissue and the regulation of collagen expression are significant factors in the pathophysiology of chordee associated with hypospadias. While the COL2A1:COL1A1 ratio is recognised as a measure of cell differentiation, there is yet to be a study specifically examining this ratio in hypospadias. The aim of this study was to determine the COL2A1:COL1A1 ratio. METHODS We collected 55 samples of dartos tissue, comprising 35 from patients with hypospadias procured from urethroplasty procedures and 20 from patients with phimosis collected during circumcision without any lichen cases at our institution. The gene expression levels of COL1A1 and COL2A1 in the dartos tissue were analyzed using reverse-transcriptase polymerase chain reaction (qPCR). RESULTS Based on the type of penile abnormality, the expression levels of COL1A1 and COL2A1 measured by qPCR were downregulated in hypospadias, with value of 0.83 (0.38-2.53) and 0.43 (0.10-5.66), respectively, compared to phimosis, which had levels of 1.85 (1.24-4.61) and 0.94 (0.26-2.47) (p < 0.001). The expression levels of COL1A1 and COL2A1 were also significantly downregulated among mild, moderate, severe penile curvature, and control groups (p < 0.001 and p = 0.02). However, the COL2A1:COL1A1 ratio did not show statistically significant differences based on penile abnormalities and curvature (p > 0.05). CONCLUSION The expression levels of COL1A1 and COL2A1 are significantly downregulated in patients with hypospadias and ventral curvature when compared to those in the phimosis group. However, the COL2A1:COL1A1 ratio was not significant.
Collapse
Affiliation(s)
- Prahara Yuri
- Urology Division, Department of Surgery, Faculty of Medicine, Nursing, and Public Health, Sardjito Hospital, Gadjah Mada University, Yogyakarta, 55281, Indonesia.
| | - Hotman Christinus
- Urology Division, Department of Surgery, Faculty of Medicine, Nursing, and Public Health, Sardjito Hospital, Gadjah Mada University, Yogyakarta, 55281, Indonesia
| | - Zico Yusuf Alfarizi
- Urology Division, Department of Surgery, Faculty of Medicine, Nursing, and Public Health, Sardjito Hospital, Gadjah Mada University, Yogyakarta, 55281, Indonesia
| | - Medina Ndoye
- Urology Department HOGIP UCAD, Cheikh Anta Diop University, Dakar, Senegal
| |
Collapse
|
35
|
Ghobadi F, Saadatmand M, Simorgh S, Brouki Milan P. Microfluidic 3D cell culture: potential application of collagen hydrogels with an optimal dose of bioactive glasses. Sci Rep 2025; 15:569. [PMID: 39747624 PMCID: PMC11696724 DOI: 10.1038/s41598-024-84346-8] [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: 08/03/2024] [Accepted: 12/23/2024] [Indexed: 01/04/2025] Open
Abstract
We engineered a microfluidic platform to study the effects of bioactive glass nanoparticles (BGNs) on cell viability under static culture. We incorporated different concentrations of BGNs (1%, 2%, and 3% w/v) in collagen hydrogel (with a concentration of 3.0 mg/mL). The microfluidic chip's dimensions were optimized through fluid flow and mass transfer simulations. Collagen type I extracted from rat tail tendons was used as the main material, and BGNs synthesized by the sol-gel method were used to enhance the mechanical properties of the hydrogel. The extracted collagen was characterized using FTIR and SDS-PAGE, and BGNs were analyzed using XRD, FTIR, DLS, and FE-SEM/EDX. The structure of the collagen-BGNs hydrogels was examined using SEM, and their mechanical properties were determined using rheological analysis. The cytotoxicity of BGNs was assessed using the MTT assay, and the viability of fibroblast (L929) cells encapsulated in the collagen-BGNs hydrogel inside the microfluidic device was assessed using a live/dead assay. Based on all these test results, the L929 cells showed high cell viability in vitro and promising microenvironment mimicry in a microfluidic device. Collagen3-BGNs3 (Collagen 3 mg/mL + BGNs 3% (w/v)) was chosen as the most suitable sample for further research on a microfluidic platform.
Collapse
Affiliation(s)
- Faezeh Ghobadi
- Department of Chemical and Petroleum Engineering, Sharif University of Technology, Tehran, Iran
| | - Maryam Saadatmand
- Department of Chemical and Petroleum Engineering, Sharif University of Technology, Tehran, Iran.
| | - Sara Simorgh
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
- Department of Tissue Engineering and Regenerative Medicine, Faculty of Advanced Technologies inMedicine, Iran University of Medical Sciences, Tehran, Iran
| | - Peiman Brouki Milan
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
- Department of Tissue Engineering and Regenerative Medicine, Faculty of Advanced Technologies inMedicine, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
36
|
Amirsadeghi A, Gudeti PKR, Tock S, Koch M, Parisi D, Kamperman M, Włodarczyk‐Biegun MK. Melt Electrowriting of Elastic Scaffolds Using PEOT-PBT Multi-block Copolymer. Adv Healthc Mater 2025; 14:e2402914. [PMID: 39659166 PMCID: PMC11773127 DOI: 10.1002/adhm.202402914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 11/09/2024] [Indexed: 12/12/2024]
Abstract
Melt electrowriting (MEW) is a powerful additive manufacturing technique to produce tissue engineering scaffolds. Despite its strength, it is limited by a small number of processable polymers. Therefore, to broaden the library of materials for MEW, we investigated the printability of poly(ethylene oxide terephthalate)-poly(butylene terephthalate) (PEOT-PBT), a thermoplastic elastomer. The effect of different printing parameters and material thermal degradation are studied. It is observed that the material is stable for >60 min at a printing temperature of 195 °C in a nitrogen environment. Next, two types of designs are printed and characterized: mesh-like and semi-random scaffolds. For both types of designs, PEOT-PBT scaffolds reveal a higher yield strain, and lower Young's modulus as compared to control polycaprolactone scaffolds. Biological studies performed using mouse embryonic fibroblasts (NIH-3T3) show good cell viability and metabolic activity on all print scaffolds. SEM imaging reveals actively migrating cells on PEOT-PBT mesh scaffolds after 24 h of culture and 98.87% of pore bridging by cells after 28 days of culture. Immunofluorescence staining shows decreased expression of alpha-smooth muscle actin from day 14 to day 28 in PEOT-PBT mesh scaffolds. Overall, it is shown that melt electrowritten PEOT-PBT scaffolds have great potential for soft tissue regeneration.
Collapse
Affiliation(s)
- Armin Amirsadeghi
- Polymer ScienceZernike Institute for Advanced MaterialsUniversity of GroningenNijenborgh 3Groningen9747 AGThe Netherlands
| | | | - Sietse Tock
- Polymer ScienceZernike Institute for Advanced MaterialsUniversity of GroningenNijenborgh 3Groningen9747 AGThe Netherlands
| | - Marcus Koch
- INM – Leibniz Institute for New MaterialsCampus D2 266123SaarbrückenGermany
| | - Daniele Parisi
- Engineering and Technology Institute Groningen (ENTEG)University of GroningenNijenborgh 3Groningen9747 AGThe Netherlands
| | - Marleen Kamperman
- Polymer ScienceZernike Institute for Advanced MaterialsUniversity of GroningenNijenborgh 3Groningen9747 AGThe Netherlands
| | - Małgorzata Katarzyna Włodarczyk‐Biegun
- Polymer ScienceZernike Institute for Advanced MaterialsUniversity of GroningenNijenborgh 3Groningen9747 AGThe Netherlands
- Biotechnology CentreThe Silesian University of TechnologyB. Krzywoustego 8Gliwice44‐100Poland
| |
Collapse
|
37
|
Cortella G, Lamparelli EP, Ciardulli MC, Lovecchio J, Giordano E, Maffulli N, Della Porta G. ColMA-based bioprinted 3D scaffold allowed to study tenogenic events in human tendon stem cells. Bioeng Transl Med 2025; 10:e10723. [PMID: 39801753 PMCID: PMC11711214 DOI: 10.1002/btm2.10723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 08/22/2024] [Accepted: 08/28/2024] [Indexed: 01/16/2025] Open
Abstract
The advent of bioprinting has enabled the creation of precise three-dimensional (3D) cell cultures suitable for biomimetic in vitro models. In this study, we developed a novel protocol for 3D printing methacrylated collagen (ColMa, or PhotoCol®) combined with tendon stem/progenitor cells (hTSPCs) derived from human tendon explants. Although pure ColMa has not previously been proposed as a printable hydrogel, this paper outlines a robust and highly reproducible pipeline for bioprinting this material. Indeed, we successfully fabricated a 3D bioengineered scaffold and cultured it for 21 days under perfusion conditions with medium supplemented with growth/differentiation factor-5 (GDF-5). This bioprinting pipeline and the culture conditions created an exceptionally favorable 3D environment, enabling the cells to proliferate, exhibit tenogenic behaviors, and produce a new collagen type I matrix, thereby remodeling the surrounding environment. Indeed, over the 21-day culture period under perfusion condition, tenomodulin expression showed a significant upregulation on day 7, with a 2.3-fold increase, compared to days 14 and 21. Collagen type I gene expression was upregulated nearly 10-fold by day 14. This trend was further confirmed by western blot analysis, which revealed a statistically significant difference in tenomodulin expression between day 21 and both day 7 and day 14. For type I collagen, significant differences were observed between day 0 and day 21, as well as between day 0 and day 14, with a p-value of 0.01. These results indicate a progressive over-expression of type I collagen, reflecting cell differentiation towards a proper tenogenic phenotype. Cytokines, such as IL-8 and IL-6, levels peaked at 8566 and 7636 pg/mL, respectively, on day 7, before decreasing to 54 and 46 pg/mL by day 21. Overall, the data suggest that the novel ColMa bioprinting protocol effectively provided a conducive environment for the growth and proper differentiation of hTSPCs, showcasing its potential for studying cell behavior and tenogenic differentiation.
Collapse
Affiliation(s)
- Giacomo Cortella
- Translational NanoMedicine Laboratory, Department of Medicine, Surgery and DentistryUniversity of SalernoBaronissiSAItaly
| | - Erwin Pavel Lamparelli
- Translational NanoMedicine Laboratory, Department of Medicine, Surgery and DentistryUniversity of SalernoBaronissiSAItaly
| | - Maria Camilla Ciardulli
- Translational NanoMedicine Laboratory, Department of Medicine, Surgery and DentistryUniversity of SalernoBaronissiSAItaly
| | - Joseph Lovecchio
- School of Science and EngineeringReykjavík UniversityReykjavíkIceland
- Institute of Biomedical and Neural EngineeringReykjavik UniversityReykjavíkIceland
| | - Emanuele Giordano
- Laboratory of Cellular and Molecular Engineering “Silvio Cavalcanti”, Department of Electrical, Electronic and Information Engineering “Guglielmo Marconi” (DEI)University of BolognaCesenaFCItaly
- Advanced Research Center on Electronic Systems (ARCES)University of BolognaBolognaBOItaly
| | - Nicola Maffulli
- School of Pharmacy and BioengineeringKeele UniversityStoke‐on‐TrentStaffordshireUK
- Department of Trauma and Orthopaedics, Faculty of Medicine and PsychologySant'Andrea Hospital, “La Sapienza” UniversityRomeItaly
| | - Giovanna Della Porta
- Translational NanoMedicine Laboratory, Department of Medicine, Surgery and DentistryUniversity of SalernoBaronissiSAItaly
- Research Centre for Biomaterials BIONAMUniversità di SalernoFiscianoSAItaly
| |
Collapse
|
38
|
Wang J, Chen X, Li R, Wang S, Geng Z, Shi Z, Jing Y, Xu K, Wei Y, Wang G, He C, Dong S, Liu G, Hou Z, Xia Z, Wang X, Ye Z, Zhou F, Bai L, Tan H, Su J. Standardization and consensus in the development and application of bone organoids. Theranostics 2025; 15:682-706. [PMID: 39744680 PMCID: PMC11671374 DOI: 10.7150/thno.105840] [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: 10/25/2024] [Accepted: 11/15/2024] [Indexed: 01/11/2025] Open
Abstract
Organoids, self-organized structures derived from stem cells cultured in a specific three-dimensional (3D) in vitro microenvironment, have emerged as innovative platforms that closely mimic in vivo cellular behavior, tissue architecture, and organ function. Bone organoids, a frontier in organoid research, can replicate the complex structures and functional characteristics of bone tissue. Recent advancements have led to the successful development of bone organoids, including models of callus, woven bone, cartilage, trabecular bone, and bone marrow. These organoids are widely utilized in establishing bone-related disease models, bone injury repair, and drug screening. However, significant discrepancies remain between current bone organoids and human skeletal tissues in terms of morphology and functionality, limiting their ability to accurately model human bone physiology and pathology. To address these challenges and promote standardization in the construction, evaluation, and application of bone organoids, we have convened experts and research teams with substantial expertise in the field. By integrating existing research findings, this consortium aims to establish a consensus to guide future research and application of bone organoids.
Collapse
Affiliation(s)
- Jian Wang
- Department of Orthopedics, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
- Trauma Orthopedics Center, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
- Institute of Musculoskeletal Injury and Translational Medicine of Organoids, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine SHU Branch, Shanghai University, Shanghai, 200444, China
| | - Xiao Chen
- Department of Orthopedics, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
- Trauma Orthopedics Center, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
- Institute of Musculoskeletal Injury and Translational Medicine of Organoids, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine SHU Branch, Shanghai University, Shanghai, 200444, China
| | - Ruiyang Li
- Department of Orthopedics, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
- Trauma Orthopedics Center, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
- Institute of Musculoskeletal Injury and Translational Medicine of Organoids, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Sicheng Wang
- Department of Orthopedics, Shanghai Zhongye Hospital, Shanghai, 200941, China
| | - Zhen Geng
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine SHU Branch, Shanghai University, Shanghai, 200444, China
| | - Zhongmin Shi
- Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Yingying Jing
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine SHU Branch, Shanghai University, Shanghai, 200444, China
| | - Ke Xu
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine SHU Branch, Shanghai University, Shanghai, 200444, China
| | - Yan Wei
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine SHU Branch, Shanghai University, Shanghai, 200444, China
| | - Guangchao Wang
- Department of Orthopedics, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
- Trauma Orthopedics Center, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
- Institute of Musculoskeletal Injury and Translational Medicine of Organoids, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Chongru He
- Department of Orthopedics, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
- Trauma Orthopedics Center, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
- Institute of Musculoskeletal Injury and Translational Medicine of Organoids, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Shiwu Dong
- Department of Biomedical Materials Science, College of Biomedical Engineering, Third Military Medical University, Chongqing, 400038, China
| | - Guohui Liu
- Union Hospital Affiliated to Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Zhiyong Hou
- Department of Orthopaedic Surgery, Third Hospital of Hebei Medical University, Shijiazhuang, 050051, China
| | - Zhidao Xia
- Institute of Life Science, College of Medicine, Swansea University, Swansea, SA2 8PP, UK
| | - Xinglong Wang
- Department of Pharmacology and Toxicology, University of Arizona, Tucson, AZ, 85721, USA
| | - Zhou Ye
- Applied Oral Sciences and Community Dental Care, Faculty of Dentistry, The University of Hong Kong, Hong Kong, 999077, China
| | - Fengjin Zhou
- Department of Orthopedics, Honghui Hospital, Xi'an Jiao Tong University, Xi'an, 710000, China
| | - Long Bai
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine SHU Branch, Shanghai University, Shanghai, 200444, China
| | - Hongbo Tan
- Department of Orthopedics, 920th Hospital of Joint Logistics Support Force of Chinese PLA, Kunming, 650032, China
| | - Jiacan Su
- Department of Orthopedics, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
- Trauma Orthopedics Center, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
- Institute of Musculoskeletal Injury and Translational Medicine of Organoids, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine SHU Branch, Shanghai University, Shanghai, 200444, China
| |
Collapse
|
39
|
Natania F, Iriawati I, Ayuningtyas FD, Barlian A. Potential of Plant-derived Exosome-like Nanoparticles from Physalis peruviana Fruit for Human Dermal Fibroblast Regeneration and Remodeling. Pharm Nanotechnol 2025; 13:358-371. [PMID: 38243927 DOI: 10.2174/0122117385281838240105110106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 11/01/2023] [Accepted: 11/14/2023] [Indexed: 01/22/2024]
Abstract
AIMS This research aimed to study the potential of PDEN from P. peruviana fruits (PENC) for regenerating and remodeling HDF. BACKGROUND Large wounds are dangerous and require prompt and effective healing. Various efforts have been undertaken, but have been somewhat ineffective. Plant-derived exosome-like nanoparticles (PDEN) are easily sampled, relatively cost-effective, exhibit high yields, and are nonimmunogenic. OBJECTIVES The objective of the study was to isolate and characterize PDEN from Physalis peruviana (PENC), and determine PENC's internalization and toxicity on HDF cells, PENC's ability to regenerate HDF (proliferation and migration), and PENC ability's to remodel HDF (collagen I and MMP-1 production). METHODS PENC was isolated using gradual filtration and centrifugation, followed by sedimentation using PEG6000. Characterization was done using a particle size analyzer, zeta potential analyzer, TEM, and BCA assay. Internalization was done using PKH67 staining. Toxicity and proliferation assays were conducted using MTT assay; meanwhile, migration assay was carried out by employing the scratch assay. Collagen I production was performed using immunocytochemistry and MMP-1 production was conducted using ELISA. RESULTS MTT assay showed a PENC concentration of 2.5 until 500 μg/mL and being non-toxic to cells. PENC has been found to induce cell proliferation in 1, 3, 5, and 7 days. PENC at a concentration of 2.5, 5, and 7.5 μg/mL, also accelerated HDF migration using the scratch assay in two days. In remodeling, PENC upregulated collagen-1 expression from day 7 to 14 compared to control. MMP-1 declined from day 2 to 7 in every PENC concentration and increased on day 14. Overall, PENC at concentrations of 2.5, 5, and 7.5 μg/mL induced HDF proliferation and migration, upregulated collagen I production, and decreased MMP-1 levels. CONCLUSION Isolated PENC was 190-220 nm in size, circular, covered with membrane, and its zeta potential was -6.7 mV; it could also be stored at 4°C for up to 2 weeks in aqua bidest. Protein concentration ranged between 170-1,395 μg/mL. Using PKH67, PENC could enter HDF within 6 hours. PENC was non-toxic up to a concentration of 500 μg/mL. Using MTT and scratch assay, PENC was found to elevate HDF proliferation and migration, and reorganize actin. Using immunocytochemistry, collagen I was upregulated by PENC, whereas MMP-1 concentration was reduced.
Collapse
Affiliation(s)
- Filia Natania
- School of Life Sciences and Technology, Institut Teknologi Bandung, Bandung, West Java, Indonesia
| | - Iriawati Iriawati
- School of Life Sciences and Technology, Institut Teknologi Bandung, Bandung, West Java, Indonesia
| | - Fitria Dwi Ayuningtyas
- Research Center of Nanoscience and Nanotechnology, Institut Teknologi Bandung, Bandung, West Java, Indonesia
| | - Anggraini Barlian
- School of Life Sciences and Technology, Institut Teknologi Bandung, Bandung, West Java, Indonesia
- Research Center of Nanoscience and Nanotechnology, Institut Teknologi Bandung, Bandung, West Java, Indonesia
| |
Collapse
|
40
|
Banerjee K, Mandal S, Nath A, Chakraborty SB, Mitra A, Gupta S. Thyroxine (T3)-mediated regulation of early cardiac repair in a chemical-induced hypoxia/reoxygenation model of adult zebrafish (Danio rerio). Wound Repair Regen 2025; 33:e13244. [PMID: 39727215 DOI: 10.1111/wrr.13244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Revised: 11/24/2024] [Accepted: 12/09/2024] [Indexed: 12/28/2024]
Abstract
Hypoxia-mediated cardiac tissue injury and its repair or regeneration are one of the major health management challenges globally. Unlike mammals, lower vertebrate species such as zebrafish (Danio rerio) represent a natural model to study cardiac injury, repair and regeneration. Thyroxine (T3) has been hypothesised to be one of the endocrine factors responsible for the evolutionary trade-off for acquiring endothermy and regenerative capability in higher vertebrates. However, the specific targets of T3 during cardiac repair are still obscure. In this study, cardiac injury was generated in adult zebrafish by acute anaemia-induced hypoxia/reoxygenation (H/R) in the presence or absence of exogenous T3 alone or along with 1-850 (inhibitor of T3 receptor) and iopanoic acid (IOA, blocker of T3 release), respectively. A microarray analysis showed that 10,226 gene expression changes in expression across all experimental groups, providing a comprehensive understanding of the cardiac transcriptome. Analysis of 11 candidate genes was conducted using qRT-PCR and the findings aligned with the microarray data. Histological assessment by Masson's trichrome staining and immunofluorescence studies also corroborated the microarray data. GO enrichment analysis showed noteworthy involvement of T3 in the modulation of genes involved in oxidative stress, cardiac fibrosis, energy metabolism, autophagy, apoptosis and regeneration during the initial repair phase (7 days) of H/R-damaged cardiac tissue. Overall, this is the first study that presents a holistic picture of cardiac repair and regeneration post H/R injury in zebrafish and the effect of T3 pre-treatment on it.
Collapse
Affiliation(s)
- Kalyan Banerjee
- Department of Zoology, Trivenidevi Bhalotia College, Raniganj, West Bengal, India
| | - Subhadeep Mandal
- Department of Zoology, Trivenidevi Bhalotia College, Raniganj, West Bengal, India
| | - Arghya Nath
- Department of Zoology, University of Burdwan, Bardhaman, West Bengal, India
| | | | - Arkadeep Mitra
- Department of Zoology, City College, Kolkata, West Bengal, India
| | - Shreyasi Gupta
- Department of Zoology, Trivenidevi Bhalotia College, Raniganj, West Bengal, India
| |
Collapse
|
41
|
Li R, Xu S, Guo Y, Cao C, Xu J, Hao L, Luo S, Chen X, Du Y, Li Y, Xie Y, Gao W, Li J, Xu B. Application of collagen in bone regeneration. J Orthop Translat 2025; 50:129-143. [PMID: 40171103 PMCID: PMC11960539 DOI: 10.1016/j.jot.2024.10.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 09/17/2024] [Accepted: 10/07/2024] [Indexed: 04/03/2025] Open
Abstract
At present, there is a significant population of individuals experiencing bone deficiencies caused by injuries, ailments affecting the bones, congenital abnormalities, and cancer. The management of substantial bone defects a significant global orthopedic challenge due to the intricacies involved in promoting and restoring the growth of fresh osseous tissue. Autografts are widely regarded as the "gold standard" for repairing bone defects because of their superior tissue acceptance and ability to control osteogenesis. However, patients undergoing autografts may encounter various challenges, including but not limited to hernia, bleeding, nerve impairment, tissue death. Therefore, researchers in regenerative medicine are striving to find alternatives. Collagen is the most abundant protein in the human body, and its triple helix structure gives it unique characteristics that contribute to its strength and functionality in various tissues. Collagen is commonly processed into various forms such as scaffolds, sponges, membranes, hydrogels, and composite materials, due to its unique compatibility with the human body, affinity for water, minimal potential for immune reactions, adaptability, and ability to transport nutrients or drugs. As an alternative material in the field of bone regeneration, collagen is becoming increasingly important. The objective of this review is to provide a comprehensive analysis of the primary types and sources of collagen, their processes of synthesis and degradation, as well as the advancements made in bone regeneration research and its potential applications. A comprehensive investigation into the role of collagen in bone regeneration is undertaken, providing valuable points of reference for a more profound comprehension of collagen applications in this field. The concluding section provides a comprehensive overview of the prospective avenues for collagen research, underscoring their promising future and highlighting their significant potential in the field of bone regeneration. The Translational Potential of this Article. The comprehensive exploration into the diverse functions and translational potential of collagen in bone regeneration, as demonstrated in this review, these findings underscore their promising potential as a treatment option with significant clinical implications, thus paving the way for innovative and efficacious therapeutic strategies in this domain.
Collapse
Affiliation(s)
- Rou Li
- China–Japan Friendship Hospital (Institute of Clinical Medical Sciences), Beijing 100029, PR China
- China Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, PR China
| | - Shiqing Xu
- China–Japan Friendship Hospital (Institute of Clinical Medical Sciences), Beijing 100029, PR China
| | - Yanning Guo
- China–Japan Friendship Hospital (Institute of Clinical Medical Sciences), Beijing 100029, PR China
| | - Cong Cao
- China–Japan Friendship Hospital (Institute of Clinical Medical Sciences), Beijing 100029, PR China
| | - Jingchen Xu
- China–Japan Friendship Hospital (Institute of Clinical Medical Sciences), Beijing 100029, PR China
| | - Lijun Hao
- The Plastic and Aesthetic Center, The First Affiliated Hospital of Harbin Medical University, Heilongjiang Province, PR China
| | - Sai Luo
- The Plastic and Aesthetic Center, The First Affiliated Hospital of Harbin Medical University, Heilongjiang Province, PR China
| | - Xinyao Chen
- The Plastic and Aesthetic Center, The First Affiliated Hospital of Harbin Medical University, Heilongjiang Province, PR China
| | - Yuyang Du
- The Plastic and Aesthetic Center, The First Affiliated Hospital of Harbin Medical University, Heilongjiang Province, PR China
| | - Ye Li
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, Guang Zhou 510515, PR China
| | - Yong Xie
- Department of Cardiac Surgery, The First Affiliated Hospital of Tsinghua University, Beijing 100036, PR China
| | - Weitong Gao
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin 150081, PR China
| | - Jing Li
- China–Japan Friendship Hospital (Institute of Clinical Medical Sciences), Beijing 100029, PR China
| | - Baohua Xu
- China–Japan Friendship Hospital (Institute of Clinical Medical Sciences), Beijing 100029, PR China
- China Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, PR China
| |
Collapse
|
42
|
Nizam AAK, Md Fadilah NI, Ahmad H, Maarof M, Fauzi MB. Injectable Gelatin-Palmitoyl-GDPH Hydrogels as Bioinks for Future Cutaneous Regeneration: Physicochemical Characterization and Cytotoxicity Assessment. Polymers (Basel) 2024; 17:41. [PMID: 39795444 PMCID: PMC11722577 DOI: 10.3390/polym17010041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2024] [Revised: 12/24/2024] [Accepted: 12/25/2024] [Indexed: 01/13/2025] Open
Abstract
Tissue engineering and regenerative medicine have made significant breakthroughs in creating complex three-dimensional (3D) constructs that mimic human tissues. This progress is largely driven by the development of hydrogels, which enable the precise arrangement of biomaterials and cells to form structures resembling native tissues. Gelatin-based bioinks are widely used in wound healing due to their excellent biocompatibility, biodegradability, non-toxicity, and ability to accelerate extracellular matrix formation. However, the role of a novel fatty acid conjugated tetrapeptide, palmitic acid-glycine-aspartic acid-proline-histidine (palmitoyl-GDPH), in enhancing hydrogel performance with human dermal fibroblasts (HDFs) concerning cell survival, proliferation, growth, and metabolism remains poorly understood. This study fabricated gelatin-palmitoyl-GDPH hydrogels at various concentrations (GE_GNP_ELS_PAL12.5 and GE_GNP_ELS_PAL25) using an injectable method and preliminary extrusion-based 3D bioprinting at 24 °C. Physicochemical characterization revealed superior water absorption, biocompatibility, and stability, aligning with optimal wound-healing criteria. In vitro cytotoxicity assays demonstrated >90% cell viability of HDFs cultured on these scaffolds for five days. These results highlight their ability to promote cell survival, proliferation, and adhesion, establishing them as strong contenders for wound healing. This study underscores the potential of gelatin-palmitoyl-GDPH hydrogels as effective bioinks for 3D bioprinting, offering a promising platform for skin tissue engineering and regenerative medicine.
Collapse
Affiliation(s)
- Aifa Asyhira Khairul Nizam
- Department of Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras, Kuala Lumpur 56000, Malaysia; (A.A.K.N.); (N.I.M.F.); (M.M.)
| | - Nur Izzah Md Fadilah
- Department of Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras, Kuala Lumpur 56000, Malaysia; (A.A.K.N.); (N.I.M.F.); (M.M.)
- Advance Bioactive Materials-Cells UKM Research Group, Universiti Kebangsaan Malaysia, Bangi 43600, Selangor, Malaysia
| | - Haslina Ahmad
- Department of Chemistry, Faculty of Science, Universiti Putra Malaysia (UPM), Serdang 43400, Selangor, Malaysia;
- Integrated Chemical Biophysics Research, Universiti Putra Malaysia (UPM), Serdang 43400, Selangor, Malaysia
| | - Manira Maarof
- Department of Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras, Kuala Lumpur 56000, Malaysia; (A.A.K.N.); (N.I.M.F.); (M.M.)
- Advance Bioactive Materials-Cells UKM Research Group, Universiti Kebangsaan Malaysia, Bangi 43600, Selangor, Malaysia
| | - Mh Busra Fauzi
- Department of Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras, Kuala Lumpur 56000, Malaysia; (A.A.K.N.); (N.I.M.F.); (M.M.)
- Advance Bioactive Materials-Cells UKM Research Group, Universiti Kebangsaan Malaysia, Bangi 43600, Selangor, Malaysia
| |
Collapse
|
43
|
Jiang Y, Jin L, Liu W, Liu H, Liu X, Tan Z. Construction of 3D tumor in vitro models with an immune microenvironment exhibiting similar tumor properties and biomimetic physiological functionality. Biomater Sci 2024; 13:223-235. [PMID: 39526532 DOI: 10.1039/d4bm00754a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Abstract
Tumors pose a serious threat to people's lives and health, and the complex tumor microenvironment is the biggest obstacle to their treatment. In contrast to the basic protein matrices typically employed in 2D or 3D cell culture systems, decellularized extracellular matrix (dECM) can create complex microenvironments. In this study, a combination of physicochemical methods was established to obtain liver decellularized extracellular matrix scaffolds (dLECMs) to provide mechanical support and cell adhesion sites. By co-culturing tumor cells, tumor-associated stromal cells and immune cells, an in vitro 3D tumor model with a biomimetic immune microenvironment was constructed. By utilizing microenvironment data obtained from human liver tumor tissues and refining the double seeding modeling process, 3D in vitro liver tumor-like tissues with a tumor immune microenvironment (TIME) were obtained and designated as reconstructed human liver cancer (RHLC). These tissues demonstrated similar tumor characteristics and exhibited satisfactory physiological functionality. The results of metabolic characterisation and mouse tumorigenicity testing verified that the constructed RHLC significantly increased in vitro drug resistance while also closely mimicking in vivo tissue metabolism. This opens up new possibilities for creating effective in vitro models for screening chemotherapy drugs.
Collapse
Affiliation(s)
- Yuhong Jiang
- College of Biology, Hunan University, Changsha, 410082, China.
- Institute of Shenzhen, Hunan University Shenzhen, 518000, China
| | - Lijuan Jin
- Greater Bay Area Institute for Innovation, Hunan University, Guangzhou, 511300, China
| | - Wenyu Liu
- College of Biology, Hunan University, Changsha, 410082, China.
| | - Hui Liu
- College of Biology, Hunan University, Changsha, 410082, China.
- Institute of Shenzhen, Hunan University Shenzhen, 518000, China
| | - Xiao Liu
- Greater Bay Area Institute for Innovation, Hunan University, Guangzhou, 511300, China
| | - Zhikai Tan
- College of Biology, Hunan University, Changsha, 410082, China.
- Institute of Shenzhen, Hunan University Shenzhen, 518000, China
- Greater Bay Area Institute for Innovation, Hunan University, Guangzhou, 511300, China
| |
Collapse
|
44
|
Alarcón-Apablaza J, Godoy-Sánchez K, Jarpa-Parra M, Garrido-Miranda K, Fuentes R. Tissue Sources Influence the Morphological and Morphometric Characteristics of Collagen Membranes for Guided Bone Regeneration. Polymers (Basel) 2024; 16:3499. [PMID: 39771351 PMCID: PMC11678201 DOI: 10.3390/polym16243499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Accepted: 12/06/2024] [Indexed: 01/11/2025] Open
Abstract
(1) Background: Collagen, a natural polymer, is widely used in the fabrication of membranes for guided bone regeneration (GBR). These membranes are sourced from various tissues, such as skin, pericardium, peritoneum, and tendons, which exhibit differences in regenerative outcomes. Therefore, this study aimed to evaluate the morphological and chemical properties of porcine collagen membranes from five different tissue sources: skin, pericardium, dermis, tendons, and peritoneum. (2) Methods: The membrane structure was analyzed using energy-dispersive X-ray spectrometry (EDX), variable pressure scanning electron microscopy (VP-SEM), Fourier transform infrared spectroscopy (FTIR), and thermal stability via thermogravimetric analysis (TGA). The absorption capacity of the membranes for GBR was also assessed using an analytical digital balance. (3) Results: The membranes displayed distinct microstructural features. Skin- and tendon-derived membranes had rough surfaces with nanopores (1.44 ± 1.24 µm and 0.46 ± 0.1 µm, respectively), while pericardium- and dermis-derived membranes exhibited rough surfaces with macropores (78.90 ± 75.89 µm and 64.89 ± 13.15 µm, respectively). The peritoneum-derived membrane featured a rough surface of compact longitudinal fibers with irregular macropores (9.02 ± 3.70 µm). The thickness varied significantly among the membranes, showing differences in absorption capacity. The pericardium membrane exhibited the highest absorption, increasing by more than 10 times its initial mass. In contrast, the skin-derived membrane demonstrated the lowest absorption, increasing by less than 4 times its initial mass. Chemical analysis revealed that all membranes were primarily composed of carbon, nitrogen, and oxygen. Thermogravimetric and differential scanning calorimetry analyses showed no significant compositional differences among the membranes. FTIR spectra confirmed the presence of collagen, with characteristic peaks corresponding to Amide A, B, I, II, and III. (4) Conclusions: The tissue origin of collagen membranes significantly influences their morphological characteristics, which may, in turn, affect their osteogenic properties. These findings provide valuable insights into the selection of collagen membranes for GBR applications.
Collapse
Affiliation(s)
- Josefa Alarcón-Apablaza
- Doctoral Program in Morphological Sciences, Faculty of Medicine, Universidad de La Frontera, Temuco 4780000, Chile;
- Research Center in Dental Sciences (CICO-UFRO), Dental School, Faculty of Dentistry, Universidad de La Frontera, Temuco 4780000, Chile
| | - Karina Godoy-Sánchez
- Scientific and Technological Bioresource Nucleus (BIOREN-UFRO), Universidad de La Frontera, Temuco 4780000, Chile; (K.G.-S.); (K.G.-M.)
| | - Marcela Jarpa-Parra
- Natural Resources and Polymers Research Laboratory, Universidad Adventista de Chile, Chillán 3780000, Chile;
| | - Karla Garrido-Miranda
- Scientific and Technological Bioresource Nucleus (BIOREN-UFRO), Universidad de La Frontera, Temuco 4780000, Chile; (K.G.-S.); (K.G.-M.)
| | - Ramón Fuentes
- Research Center in Dental Sciences (CICO-UFRO), Dental School, Faculty of Dentistry, Universidad de La Frontera, Temuco 4780000, Chile
- Scientific and Technological Bioresource Nucleus (BIOREN-UFRO), Universidad de La Frontera, Temuco 4780000, Chile; (K.G.-S.); (K.G.-M.)
- Department of Integral Adult Dentistry, Dental School, Faculty of Dentistry, Universidad de La Frontera, Temuco 4780000, Chile
| |
Collapse
|
45
|
Ozudogru E, Kurt T, Derkus B, Cengiz U, Arslan YE. Supercritical CO 2-Mediated Decellularization of Bovine Spinal Cord Meninges: A Comparative Study for Decellularization Performance. ACS OMEGA 2024; 9:48781-48790. [PMID: 39676980 PMCID: PMC11635505 DOI: 10.1021/acsomega.4c08684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/21/2024] [Revised: 11/07/2024] [Accepted: 11/15/2024] [Indexed: 12/17/2024]
Abstract
The extracellular matrix (ECM) of spinal meninge tissue closely resembles the wealthy ECM content of the brain and spinal cord. The ECM is typically acquired through the process of decellularizing tissues. Nevertheless, the decellularization process of the brain and spinal cord is challenging due to their high-fat content, in contrast to the spinal meninges. Hence, bovine spinal cord meninges offer a promising source to produce ECM-based scaffolds, thanks to their abundance, accessibility, and ease of decellularization for neural tissue engineering. However, most decellularization techniques involve disruptive chemicals and repetitive rinsing processes, which could lead to drastic modifications in the tissue ultrastructure and a loss of mechanical stability. Over the past decade, supercritical fluid technology has experienced considerable advancements in fabricating biomaterials with its applications spreading out to tissue engineering to tackle the complications mentioned above. Supercritical carbon-dioxide (scCO2)-based decellularization procedures especially offer a significant advantage over classical decellularization techniques, enabling the preservation of extracellular matrix components and structures. In this study, we decellularized the bovine spinal cord meninges by seven different methods. To identify the most effective approach, the decellularized matrices were characterized by dsDNA, collagen, and glycosaminoglycan contents and histological analyses. Moreover, the mechanical properties of the hydrogels produced from the decellularized matrices were evaluated. The novel scCO2-based treatment was completed in a shorter time than the conventional method (3 versus 7 days) while maintaining the structural and mechanical integrity of the tissue. Additionally, all hydrogels derived from scCO2-decellularized matrices demonstrated high cell viability and biocompatibility in a cell culture. The current study suggests a rapid, effective, and detergent-free scCO2-assisting decellularization protocol for clinical tissue engineering applications.
Collapse
Affiliation(s)
- Eren Ozudogru
- Regenerative
Biomaterials Laboratory, Department of Bioengineering, Faculty of
Engineering, Canakkale Onsekiz Mart University, Canakkale 17100, Turkey
| | - Tugce Kurt
- Regenerative
Biomaterials Laboratory, Department of Bioengineering, Faculty of
Engineering, Canakkale Onsekiz Mart University, Canakkale 17100, Turkey
| | - Burak Derkus
- Stem Cell
Research Laboratory, Department of Chemistry, Faculty of Science, Ankara University, Ankara 06560, Turkey
| | - Ugur Cengiz
- Surface Science
Research Laboratory, Department of Chemical Engineering, Faculty of
Engineering, Canakkale Onsekiz Mart University, Canakkale 17020, Turkey
| | - Yavuz Emre Arslan
- Regenerative
Biomaterials Laboratory, Department of Bioengineering, Faculty of
Engineering, Canakkale Onsekiz Mart University, Canakkale 17100, Turkey
| |
Collapse
|
46
|
Zhang Y, Fu R, Zhu C, Yuwen W, Zhang J, Duan Z, Fan D. Preparation of recombinant type I collagen (PF-I-80) and its functional characterization and biomedical applications in wound healing. Int J Biol Macromol 2024; 282:136679. [PMID: 39437939 DOI: 10.1016/j.ijbiomac.2024.136679] [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: 04/22/2024] [Revised: 10/15/2024] [Accepted: 10/16/2024] [Indexed: 10/25/2024]
Abstract
This study evaluates the potential applications of recombinant PF-I-80 protein in regenerative medicine and the treatment of inflammatory diseases, focusing on its effects on cell migration, differentiation, and anti-inflammatory properties. Various in vitro assays were conducted, including scratch assays, Transwell experiments, RT-PCR and Western Blot to analyze gene and protein expression related to differentiation and inflammation, and immunofluorescence staining to observe cellular changes. The results indicated that PF-I-80 significantly promoted cell migration, highlighting its potential in tissue repair and regeneration. It also enhanced cell differentiation, demonstrating its applicability in tissue repair, and showed significant anti-inflammatory effects by reducing the expression of pro-inflammatory cytokines. In animal models, PF-I-80 notably reduced levels of inflammatory factors IL-1β and TNF-α, shortened the inflammatory phase, and accelerated wound healing. Additionally, PF-I-80 increased FGF-2 levels, which promoted the proliferation of endothelial and fibroblast cells and enhanced collagen synthesis. These in vitro and in vivo findings position PF-I-80 as a promising biomaterial for applications in regenerative medicine and inflammatory disease treatment.
Collapse
Affiliation(s)
- Yan Zhang
- Engineering Research Center of Western Resource Innovation Medicine Green Manufacturing, Ministry of Education, 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; Biotech. & Biomed. Research Institute, Northwest University, Xi'an 710069, 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; Biotech. & Biomed. Research Institute, Northwest University, Xi'an 710069, China
| | - Weigang Yuwen
- Taibai Campus, Northwest University, Xi'an 710069, China
| | - Jiangrui Zhang
- Taibai Campus, 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; 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; Biotech. & Biomed. Research Institute, Northwest University, Xi'an 710069, China.
| |
Collapse
|
47
|
Tymińska A, Karska N, Skoniecka A, Zawrzykraj M, Banach-Kopeć A, Mania S, Zieliński J, Kondej K, Gurzawska-Comis K, Skowron PM, Tylingo R, Rodziewicz-Motowidło S, Pikuła M. A novel chitosan-peptide system for cartilage tissue engineering with adipose-derived stromal cells. Biomed Pharmacother 2024; 181:117683. [PMID: 39561590 DOI: 10.1016/j.biopha.2024.117683] [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: 08/23/2024] [Revised: 11/06/2024] [Accepted: 11/07/2024] [Indexed: 11/21/2024] Open
Abstract
The natural healing process of cartilage injuries often fails to fully restore the tissue's biological and mechanical functions. Cartilage grafts are costly and require surgical intervention, often associated with complications such as intraoperative infection and rejection by the recipient due to ischemia. Novel tissue engineering technologies aim to ideally fill the cartilage defect to prevent disease progression or regenerate damaged tissue. Despite many studies on designing biocompatible composites to stimulate chondrogenesis, only few focus on peptides and carriers that promote stem cell proliferation or differentiation to promote healing. Our research aimed to design a carbohydrate chitosan-based biomaterial to stimulate stem cells into the chondrogenesis pathway. Our strategy was to combine chitosan with a novel peptide (UG28) that sequence was based on the copin protein. The construct stimulated human adipose-derived stem cells (AD-SCs) cells to undergo chondrogenic differentiation. Chitosan 75/500 allows AD-SCs to grow and has no harmful effects on the cells. The combination of UG28 peptide with the chitosan composite offers promising properties for cell differentiation, indicating its potential for clinical applications in cartilage regeneration.
Collapse
Affiliation(s)
- Agata Tymińska
- Laboratory of Tissue Engineering and Regenerative Medicine, Division of Embryology, Department of Anatomy, Faculty of Medicine, Medical University of Gdańsk, Gdańsk 80-211, Poland.
| | - Natalia Karska
- Department of Biomedical Chemistry, Faculty of Chemistry, University of Gdańsk, Gdańsk 80-308, Poland
| | - Aneta Skoniecka
- Laboratory of Tissue Engineering and Regenerative Medicine, Division of Embryology, Department of Anatomy, Faculty of Medicine, Medical University of Gdańsk, Gdańsk 80-211, Poland
| | - Małgorzata Zawrzykraj
- Division of Clinical Anatomy, Department of Anatomy, Medical University of Gdańsk, 80-211, Poland
| | - Adrianna Banach-Kopeć
- Department of Chemistry, Technology and Biotechnology of Food Gdańsk University of Technology, Gdańsk 80-233, Poland
| | - Szymon Mania
- Department of Chemistry, Technology and Biotechnology of Food Gdańsk University of Technology, Gdańsk 80-233, Poland
| | - Jacek Zieliński
- Department of Oncologic Surgery, Medical University of Gdańsk, Gdańsk 80-214, Poland
| | - Karolina Kondej
- Department of Plastic Surgery, Medical University of Gdańsk, Gdańsk 80-214, Poland
| | - Katarzyna Gurzawska-Comis
- Department of Dentistry and Oral Health, Aarhus University, Vennelyst Boulevard 9, Aarhus C DK-8000, Denmark
| | - Piotr M Skowron
- Department of Molecular Biotechnology, Faculty of Chemistry, University of Gdansk, Gdansk, 80-308, Poland
| | - Robert Tylingo
- Department of Chemistry, Technology and Biotechnology of Food Gdańsk University of Technology, Gdańsk 80-233, Poland
| | | | - Michał Pikuła
- Laboratory of Tissue Engineering and Regenerative Medicine, Division of Embryology, Department of Anatomy, Faculty of Medicine, Medical University of Gdańsk, Gdańsk 80-211, Poland.
| |
Collapse
|
48
|
van Dijk FS, Angwin C, Ghali N, Zschocke J, Wagner B. Non-genetic diagnostic investigations in monogenic Ehlers-Danlos syndromes. MED GENET-BERLIN 2024; 36:247-254. [PMID: 39629472 PMCID: PMC11610441 DOI: 10.1515/medgen-2024-2062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/07/2024]
Abstract
With increased application of Next Generation Sequencing (NGS) in the diagnosis of monogenic Ehlers-Danlos syndromes, there is an increased probability to identify variants of unknown significance. Additionally, in some cases no genetic alteration may be identified whilst there is a strong clinical suspicion on a monogenic EDS type. The diagnostic value of non-genetic investigations, which prior to NGS were quite commonly used to support the clinical diagnosis of monogenic EDS types, is explored. In addition, new structural/functional investigations that could deliver evidence towards pathogenicity are discussed. It appears that certain functional and/or structural investigations used frequently in the past can remain helpful and can provide additional evidence that may confirm a clinical diagnosis of a monogenic EDS type. However, there is a need for the development of novel structural/functional studies for monogenic types of EDS. The level of evidence of such studies for application in the established diagnostic DNA variant classification criteria remains to be determined.
Collapse
Affiliation(s)
- Fleur S. van Dijk
- London North West University Health Care NHS TrustNational EDS service, London North West University Health Care, NHS TrustWatford RoadHA1 3UJHarrowUnited Kingdom
| | - Chloe Angwin
- London North West University Health Care NHS TrustNational EDS serviceWatford RoadHA1 3UJHarrowUnited Kingdom
| | - Neeti Ghali
- Imperial College LondonDepartment of Metabolism, Digestion andSW7 2AZLondonUnited Kingdom
| | - Johannes Zschocke
- Medical University InnsbruckInstitute of Human Genetics, Department of GeneticsPeter-Mayr-Str. 16020InnsbruckAustria
| | - Bart Wagner
- Royal Hallamshire HospitalElectron microscopy section, Histopathology DepartmentGlossop RoadS10 2JFSheffieldUnited Kingdom
| |
Collapse
|
49
|
Masri S, Fauzi MB, Rajab NF, Lee WH, Zainal Abidin DA, Siew EL. In vitro 3D skin culture and its sustainability in toxicology: a narrative review. ARTIFICIAL CELLS, NANOMEDICINE, AND BIOTECHNOLOGY 2024; 52:476-499. [PMID: 39359233 DOI: 10.1080/21691401.2024.2407617] [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: 04/15/2024] [Revised: 09/03/2024] [Accepted: 09/11/2024] [Indexed: 10/04/2024]
Abstract
In current toxicological research, 2D cell cultures and animal models are well- accepted and commonly employed methods. However, these approaches have many drawbacks and are distant from the actual environment in human. To embrace this, great efforts have been made to provide alternative methods for non-animal skin models in toxicology studies with the need for more mechanistically informative methods. This review focuses on the current state of knowledge regarding the in vitro 3D skin model methods, with different functional states that correspond to the sustainability in the field of toxicology testing. We discuss existing toxicology testing methods using in vitro 3D skin models which provide a better understanding of the testing requirements that are needed. The challenges and future landscape in using the in vitro 3D skin models in toxicology testing are also discussed. We are confident that the in vitro 3D skin models application may become an important tool in toxicology in the context of risk assessment.
Collapse
Affiliation(s)
- Syafira Masri
- Department of Tissue Engineering and Regenerative Medicine, Universiti Kebangsaan Malaysia, Cheras, Malaysia
| | - Mh Busra Fauzi
- Department of Tissue Engineering and Regenerative Medicine, Universiti Kebangsaan Malaysia, Cheras, Malaysia
- Advance Bioactive Materials-Cells (Adv-BioMaC) UKM Research Group, Universiti Kebangsaan Malaysia, Bangi, Malaysia
| | - Nor Fadilah Rajab
- Centre for Health Aging and Wellness, Faculty of Helath Sciences, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Wing-Hin Lee
- Royal College of Medicine Perak, Universiti Kuala Lumpur (UniKL RCMP), Perak, Malaysia
| | | | - Ee Ling Siew
- ASASIpintar Unit, Pusat PERMATA@Pintar Negara, Universiti Kebangsaan Malaysia, Bangi, Malaysia
- Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| |
Collapse
|
50
|
Giovanetti K, Tuma RB, Sant'Ana Pegorin Brasil G, Miranda MCR, Borges FA, Tanaka JL, Burd BS, Cortellazzi KL, Guerra NB, Mussagy CU, Floriano JF, Dos Santos LS, de Melo Silva W, Cao W, Herculano RD, Caria PHF. β-Tricalcium phosphate incorporated natural rubber latex membranes for calvarial bone defects: Physicochemical, in vitro and in vivo assessment. Int J Biol Macromol 2024; 282:137328. [PMID: 39515716 DOI: 10.1016/j.ijbiomac.2024.137328] [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: 08/06/2024] [Revised: 10/30/2024] [Accepted: 11/05/2024] [Indexed: 11/16/2024]
Abstract
Natural rubber latex membrane (NRL) is a biocompatible macromolecule that stimulates angiogenesis and promotes bone repair. Similarly, β-tricalcium phosphate (β-TCP) is an osteoconductive and osteoinductive bioceramic widely used as a bone substitute. Here, we investigated the combined use of these biomaterials in the guided bone regeneration process for calvarial defects in rats. Physicochemical characterization was performed to evaluate the interaction between β-TCP and NRL. Membrane toxicity was assessed using MC3T3 osteoblasts culture and in vivo assays with Caenorhabditis elegans. Lastly, NRL membranes, NRL incorporated with β-TCP membranes (NRL-β-TCP), and a periosteum-only (control group) were tested on rodents. MC3T3 cells adhered to membranes, preserving their morphology and intercellular connections. NRL-β-TCP membranes demonstrated no toxicity in larvae, which maintained their sinusoidal wave shape. Tests results on rodents revealed statistical difference between the groups at 60 days post-operation. NRL-β-TCP (56.1 ± 14.0 %) had an average 1.48-fold higher than the control group (38.0 ± 9.1 %), with tissue production and bone remodeling. Our qualitative histological analyses revealed that membranes significantly accelerated bone formation without any signs of inflammatory reactions. We conclude that NRL-β-TCP has potential to be used for flat bone regeneration, with osteoconductive properties, being a cheap, biocompatible, and effective occlusive barrier.
Collapse
Affiliation(s)
- Karina Giovanetti
- Institute of Biology, University of Campinas (UNICAMP), Campinas, São Paulo, Brazil.
| | - Rafael Brull Tuma
- Bioengineering & Biomaterials Group, School of Pharmaceutical Science, São Paulo State University (UNESP), Araraquara, São Paulo, Brazil
| | - Giovana Sant'Ana Pegorin Brasil
- Bioengineering & Biomaterials Group, School of Pharmaceutical Science, São Paulo State University (UNESP), Araraquara, São Paulo, Brazil
| | - Matheus Carlos Romeiro Miranda
- Institute of Environmental, Chemical and Pharmaceutical Sciences, Federal University of São Paulo (USP), Diadema, SP, Brazil
| | - Felipe Azevedo Borges
- Bioengineering & Biomaterials Group, School of Pharmaceutical Science, São Paulo State University (UNESP), Araraquara, São Paulo, Brazil
| | - Jean Lucas Tanaka
- Bioengineering & Biomaterials Group, School of Pharmaceutical Science, São Paulo State University (UNESP), Araraquara, São Paulo, Brazil
| | - Betina Sayeg Burd
- Bioengineering & Biomaterials Group, School of Pharmaceutical Science, São Paulo State University (UNESP), Araraquara, São Paulo, Brazil
| | - Karine Laura Cortellazzi
- Departmentof Social Dentistry, Piracicaba Dental School, University of Campinas (UNICAMP), Piracicaba, SP, Brazil
| | - Nayrim Brizuela Guerra
- School of Science, São Paulo State University (UNESP), Bauru, São Paulo 17033-360, Brazil
| | - Cassamo Ussemane Mussagy
- Escuela de Agronomía, Facultad de Ciencias Agronómicas y de los Alimentos, Pontificia Universidad Católica de Valparaíso, Chile
| | - Juliana Ferreira Floriano
- Bioengineering & Biomaterials Group, School of Pharmaceutical Science, São Paulo State University (UNESP), Araraquara, São Paulo, Brazil; National Heart and Lung Institute, Imperial College London, London, UK
| | - Lindomar Soares Dos Santos
- Faculty of Philosophy, Sciences and Languages of Ribeirão Preto, University of São Paulo (USP), 3900 Bandeirantes Avenue, Ribeirão Preto, SP 14.040-901, Brazil
| | - William de Melo Silva
- Institute of Biotechnology, São Paulo State University (UNESP), University Avenue 3780, 18610-034, Botucatu, Brazil
| | - Wei Cao
- Autonomy Research Center for STEAHM (ARCS), California State University, Northridge, CA 91324, USA
| | - Rondinelli Donizetti Herculano
- Bioengineering & Biomaterials Group, School of Pharmaceutical Science, São Paulo State University (UNESP), Araraquara, São Paulo, Brazil; Autonomy Research Center for STEAHM (ARCS), California State University, Northridge, CA 91324, USA.
| | | |
Collapse
|