1
|
Asadi Tokmedash M, Kim C, Chavda AP, Li A, Robins J, Min J. Engineering multifunctional surface topography to regulate multiple biological responses. Biomaterials 2025; 319:123136. [PMID: 39978049 DOI: 10.1016/j.biomaterials.2025.123136] [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: 09/27/2024] [Revised: 01/04/2025] [Accepted: 01/23/2025] [Indexed: 02/22/2025]
Abstract
Surface topography or curvature plays a crucial role in regulating cell behavior, influencing processes such as adhesion, proliferation, and gene expression. Recent advancements in nano- and micro-fabrication techniques have enabled the development of biomimetic systems that mimic native extracellular matrix (ECM) structures, providing new insights into cell-adhesion mechanisms, mechanotransduction, and cell-environment interactions. This review examines the diverse applications of engineered topographies across multiple domains, including antibacterial surfaces, immunomodulatory devices, tissue engineering scaffolds, and cancer therapies. It highlights how nanoscale features like nanopillars and nanospikes exhibit bactericidal properties, while many microscale patterns can direct stem cell differentiation and modulate immune cell responses. Furthermore, we discuss the interdisciplinary use of topography for combined applications, such as the simultaneous regulation of immune and tissue cells in 2D and 3D environments. Despite significant advances, key knowledge gaps remain, particularly regarding the effects of topographical cues on multicellular interactions and dynamic 3D contexts. This review summarizes current fabrication methods, explores specific and interdisciplinary applications, and proposes future research directions to enhance the design and utility of topographically patterned biomaterials in clinical and experimental settings.
Collapse
Affiliation(s)
| | - Changheon Kim
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Ajay P Chavda
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Adrian Li
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Jacob Robins
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Jouha Min
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA; Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA; Department of Macromolecular Science and Engineering, University of Michigan, Ann Arbor, MI, 48109, USA; Rogel Cancer Center, University of Michigan, Ann Arbor, MI, 48109, USA; Weil Institute for Critical Care Research and Innovation, University of Michigan, Ann Arbor, MI, 48109, USA.
| |
Collapse
|
2
|
Yang X, Mistry M, Chen AD, Chan BP. Tailoring extracellular matrix niches: Impact of glycosaminoglycan content on multiple differentiation of human mesenchymal stem cells. Biomaterials 2025; 318:123130. [PMID: 39893783 DOI: 10.1016/j.biomaterials.2025.123130] [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: 10/29/2024] [Revised: 01/12/2025] [Accepted: 01/23/2025] [Indexed: 02/04/2025]
Abstract
Glycosaminoglycan (GAG) represents an important extracellular matrix (ECM), particularly in GAG-rich tissues such as nucleus pulposus and cartilage. The ratio of GAGs/hydroxyproline (HYP) is an indicator of the relative abundance of the space-filling GAG matrix to the fibrous collagen matrix in a particular tissue. Here, we hypothesize that ECM niche with different GAG/HYP ratios will affect the outcomes of multiple differentiation of human mesenchymal stem cells (hMSCs). Specifically, we fabricated collagen-based biomaterials with different GAG/HYP ratios, and differentiate hMSCs in these materials towards osteogenic, chondrogenic and discogenic lineages. In osteogenic differentiation, Collagen without GAG (GAG/HYP ratio 0) showed higher calcium (Ca) and phosphorus (P) deposition and Ca/P ratio, more biomimetic ultrastructure, and better osteogenic phenotypic expression. For chondrogenic differentiation, aminated collagen (aCol-GAG) with intermediate GAG content (GAG/HYP ratio 5.0:1) showed higher GAG deposition, more biomimetic ultrastructure, and better chondrogenic phenotype. In discogenic differentiation, aminated collagen-aminated hyaluronic acid (aHA)-GAG (aCol-aHA-GAG) with the highest GAG content (GAG/HYP ratio 19.8:1), showed intensive GAG deposition, biomimetic ultrastructure, and higher phenotypic marker expression. This study contributes to developing collagen-based biomimetic materials with different GAG/HYP ratios and suggests the use of tissue-specific GAG/HYP ratio as a scaffold design parameter for hMSCs-based musculoskeletal tissue engineering. (198 words).
Collapse
Affiliation(s)
- Xingxing Yang
- Tissue Engineering Lab, School of Biomedical Science, Institute of Tissue Engineering and Regenerative Medicine, Department of Biomedical Engineering, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, China; Department of Mechanical Engineering, The University of Hong Kong, Hong Kong Special Administrative Region, China; Advanced Biomedical Instrumentation Centre, Hong Kong Special Administrative Region, China
| | - Maitraee Mistry
- Tissue Engineering Lab, School of Biomedical Science, Institute of Tissue Engineering and Regenerative Medicine, Department of Biomedical Engineering, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, China; Department of Mechanical Engineering, The University of Hong Kong, Hong Kong Special Administrative Region, China; Advanced Biomedical Instrumentation Centre, Hong Kong Special Administrative Region, China
| | - Abigail Dee Chen
- Tissue Engineering Lab, School of Biomedical Science, Institute of Tissue Engineering and Regenerative Medicine, Department of Biomedical Engineering, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, China; Department of Mechanical Engineering, The University of Hong Kong, Hong Kong Special Administrative Region, China; Advanced Biomedical Instrumentation Centre, Hong Kong Special Administrative Region, China
| | - Barbara Pui Chan
- Tissue Engineering Lab, School of Biomedical Science, Institute of Tissue Engineering and Regenerative Medicine, Department of Biomedical Engineering, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, China; Department of Mechanical Engineering, The University of Hong Kong, Hong Kong Special Administrative Region, China; Advanced Biomedical Instrumentation Centre, Hong Kong Special Administrative Region, China.
| |
Collapse
|
3
|
Prathumwon C, Anuchapreeda S, Kiattisin K, Panyajai P, Wichayapreechar P, Surh YJ, Ampasavate C. Curcumin and EGCG combined formulation in nanostructured lipid carriers for anti-aging applications. Int J Pharm X 2025; 9:100323. [PMID: 40115962 PMCID: PMC11923819 DOI: 10.1016/j.ijpx.2025.100323] [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: 11/20/2024] [Revised: 02/21/2025] [Accepted: 02/22/2025] [Indexed: 03/23/2025] Open
Abstract
Curcumin (Cur) and epigallocatechin gallate (EGCG), the primary active compounds in turmeric and green tea, respectively, have been investigated for their anti-aging potential. The Cur and EGCG combination was encapsulated in sustained-release nanostructured lipid carriers (NLCs) to enhance their bioactivities and pharmaceutical properties. A significant enhancement in the antioxidant activities of the Cur and EGCG combination was observed at an optimal ratio, as demonstrated by the 2,2-diphenyl-1-picrylhydrazyl radical scavenging assay (118.83 ± 3.78 %), ferric ion reducing antioxidant power assay (217.25 ± 13.45 %), and lipid peroxidation inhibition assay (106.08 ± 12.93 %), compared to Cur alone without compromising the antioxidant activities and total phenolic content of EGCG. This is due to the enhancement of total phenolic content of the combination of 218.83 ± 10.57 %. For anti-aging activities, the combination exhibited stimulation of SIRT1 protein and inhibition of collagenase and elastase of 27.53 ± 0.73 %, 43.70 ± 1.05 % and 51.76 ± 6.52 % compared with that achieved with Cur alone, respectively. The incorporation of the Cur and EGCG combination into NLCs resulted in high entrapment efficiencies of 98.60 ± 0.05 % for Cur and 98.40 ± 0.08 % for EGCG, with corresponding loading capacities of 0.789 ± 0.001 % and 3.935 ± 0.003 %, respectively. When formulated NLCs into an emulgel base, the system demonstrated sustained release profiles over 48 h, with 12.82 ± 0.99 % release of Cur and 63.77 ± 5.76 % release of EGCG. Significant skin retention was also observed after 24 h, with 23.88 ± 1.71 % Cur and 22.79 ± 4.65 % EGCG retained in the skin. Therefore, Cur: EGCG-loaded NLCs in emulgel can deliver the active compounds into the dermis, enhancing skin penetration, sustained delivery, and anti-aging activity superior to each conventional single active compound in topical formulations.
Collapse
Affiliation(s)
- Chidchanok Prathumwon
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Songyot Anuchapreeda
- Division of Clinical Microscopy, Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Kanokwan Kiattisin
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Pawaret Panyajai
- Division of Clinical Microscopy, Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Panikchar Wichayapreechar
- Department of Cosmetic Sciences, School of Pharmaceutical Sciences, University of Phayao, Phayao 56000, Thailand
| | - Young-Joon Surh
- College of Pharmacy, Seoul National University, Seoul 151-741, South Korea
| | - Chadarat Ampasavate
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Chiang Mai University, Chiang Mai 50200, Thailand
- Center for Excellence in Pharmaceutical Nanotechnology, Faculty of Pharmacy, Chiang Mai University, Chiang Mai 50200, Thailand
| |
Collapse
|
4
|
Gu Z, He Y, Xiang H, Qin Q, Cao X, Jiang K, Zhang H, Li Y. Self-healing injectable multifunctional hydrogels for intervertebral disc disease. Mater Today Bio 2025; 32:101655. [PMID: 40166378 PMCID: PMC11957681 DOI: 10.1016/j.mtbio.2025.101655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 03/06/2025] [Accepted: 03/10/2025] [Indexed: 04/02/2025] Open
Abstract
Intervertebral disc degeneration (IVDD) is increasingly prevalent in aging societies and poses a significant health challenge. Due to the limited blood supply to the disc, oral medications and systemic treatments are often ineffective. Consequently, localized injection therapies, which deliver therapeutic agents directly to the degenerated disc, have emerged as more efficient. Self-healing injectable hydrogels are particularly promising due to their potential for minimally invasive delivery, precise implantation, and targeted drug release into hard-to-reach tissue sites, including those requiring prolonged healing. Their dynamic viscoelastic properties accurately replicate the mechanical environment of the natural nucleus pulposus, providing cells with an adaptive biomimetic microenvironment. This review will initially discuss the anatomy and pathophysiology of intervertebral discs, current treatments, and their limitations. Subsequently, we conduct bibliometric analysis to explore the research hotspots and trends in applying injectable hydrogel technology to treat IVDD. It will then explore the promising features of injectable hydrogels in biomedical applications such as drug, protein, cells and gene delivery, tissue engineering and regenerative medicine. We discuss the construction mechanisms of injectable hydrogels via physical interactions, chemical and biological crosslinkers, and discuss the selection of biomaterials and fabrication methods for developing novel hydrogels for IVD tissue engineering. The article concludes with future perspectives on the application of injectable hydrogels in this field.
Collapse
Affiliation(s)
- Zhengrong Gu
- Department of Orthopedics, Affiliated Guang'an District People's Hospital of North Sichuan Medical College, Guang'an County, 638000, PR China
| | - Yi He
- Department of Orthopedics, Affiliated Nanbu People's Hospital of North Sichuan Medical College, Nanbu County, Nanchong, 637000, PR China
| | - Honglin Xiang
- Department of Orthopedics, Laboratory of Biological Tissue Engineering and Digital Medicine, Institute of Nanomedicine Innovation and Translational Research, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000, PR China
| | - Qiwei Qin
- Department of Orthopedics, Laboratory of Biological Tissue Engineering and Digital Medicine, Institute of Nanomedicine Innovation and Translational Research, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000, PR China
| | - Xinna Cao
- Department of Orthopedics, Laboratory of Biological Tissue Engineering and Digital Medicine, Institute of Nanomedicine Innovation and Translational Research, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000, PR China
| | - Ke Jiang
- Department of Orthopedics, Laboratory of Biological Tissue Engineering and Digital Medicine, Institute of Nanomedicine Innovation and Translational Research, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000, PR China
| | - Haoshaqiang Zhang
- Department of Orthopedics Surgery, People's Hospital of Xinjiang Uygur Autonomous Region, No. 91, Tianchi Road, Tianshan District, Urumqi, 830001, PR China
| | - Yuling Li
- Department of Orthopedics, Laboratory of Biological Tissue Engineering and Digital Medicine, Institute of Nanomedicine Innovation and Translational Research, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000, PR China
| |
Collapse
|
5
|
Malakpour-Permlid A, Rodriguez MM, Untracht GR, Andersen PE, Oredsson S, Boisen A, Zór K. High-throughput non-homogenous 3D polycaprolactone scaffold for cancer cell and cancer-associated fibroblast mini-tumors to evaluate drug treatment response. Toxicol Rep 2025; 14:101863. [PMID: 39758801 PMCID: PMC11699757 DOI: 10.1016/j.toxrep.2024.101863] [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: 10/01/2024] [Revised: 12/03/2024] [Accepted: 12/11/2024] [Indexed: 01/07/2025] Open
Abstract
High-throughput screening (HTS) three-dimensional (3D) tumor models are a promising approach for cancer drug discovery, as they more accurately replicate in vivo cell behavior than two-dimensional (2D) models. However, assessing and comparing current 3D models for drug efficacy remains essential, given the significant influence of cellular conditions on treatment response. To develop in vivo mimicking 3D models, we evaluated two HTS 3D models established in 96-well plates with 3D polycaprolactone (PCL) scaffolds fabricated using two distinct methods, resulting in scaffolds with either homogenous or non-homogenous fiber networks. These models, based on human HeLa cervical cancer cells and cancer-associated fibroblasts (CAFs) cultured as mono- or co-cultures within the 3D scaffolds, revealed that anticancer drug paclitaxel (PTX) exhibited consistently higher inhibitory concentration 50 (IC50) in 3D (≥ 1000 nM) compared to 2D (≥ 100 nM), indicating reduced toxicity on cells cultured in 3D. Interestingly, the toxicity of PTX was significantly lower on mini-tumors in non-homogenous 3D (IC50: 600 or 1000 nM) than in homogenous 3D cultures (IC50 exceeding 1000 nM). Microscopic studies revealed that the non-homogenous scaffolds closely resemble the tumor collagen network than their homogeneous counterpart. Both 3D scaffolds offer optimal pore size, facilitating efficient cell infiltration into the depth of 58.1 ± 1.2 µm (homogenous) and 86.4 ± 9.8 µm (non-homogenous) within 3D cultures. Cells cultured in the 3D non-homogenous systems exhibited drug treatment responses closer to in vivo conditions, highlighting the role of scaffold structure and design on cellular response to drug treatment. The PCL-based 3D models provide a robust, tunable, and efficient approach for the HTS of anti-cancer drugs compared to conventional 2D systems.
Collapse
Affiliation(s)
- Atena Malakpour-Permlid
- Center for Intelligent Drug Delivery and Sensing Using Microcontainers and Nanomechanics (IDUN), Department of Health Technology, Technical University of Denmark, Kongens Lyngby 2800, Denmark
| | - Manuel Marcos Rodriguez
- Center for Intelligent Drug Delivery and Sensing Using Microcontainers and Nanomechanics (IDUN), Department of Health Technology, Technical University of Denmark, Kongens Lyngby 2800, Denmark
| | - Gavrielle R. Untracht
- Department of Health Technology, Technical University of Denmark, Kongens Lyngby 2800, Denmark
| | - Peter E. Andersen
- Department of Health Technology, Technical University of Denmark, Kongens Lyngby 2800, Denmark
| | | | - Anja Boisen
- Center for Intelligent Drug Delivery and Sensing Using Microcontainers and Nanomechanics (IDUN), Department of Health Technology, Technical University of Denmark, Kongens Lyngby 2800, Denmark
| | - Kinga Zór
- Center for Intelligent Drug Delivery and Sensing Using Microcontainers and Nanomechanics (IDUN), Department of Health Technology, Technical University of Denmark, Kongens Lyngby 2800, Denmark
- BioInnovation Institute Foundation, Copenhagen N 2200, Denmark
- Innovation Acta S.r.l., Siena, Via delle 1-53100, Italy
| |
Collapse
|
6
|
Ji ES, Lee BS, Mun J, Jeon SY, Hong HR, Kim HJ, Kim YJ, Do SG, Shin JH, Kim KH. Quantification of salmon nasal cartilage extracts using liquid chromatography-tandem mass spectrometry. Food Chem 2025; 475:143280. [PMID: 39956058 DOI: 10.1016/j.foodchem.2025.143280] [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: 10/30/2024] [Revised: 02/03/2025] [Accepted: 02/06/2025] [Indexed: 02/18/2025]
Abstract
Proteoglycans are high molecular weight glycoproteins with potential benefits in preventing osteoarthritis, reducing inflammation, enhancing immune function, and promoting skin health. Aggrecan, a key proteoglycan with glycosaminoglycan (GAG) chains, poses challenges in accurate quantification due to its complex structure. We hypothesize that by selecting target peptides from core proteins that exclude post-translational modifications such as GAG attachment, proteoglycans can be analyzed with high sensitivity and accuracy. In this study, we identified aggrecan from salmon nasal cartilage (Oncorhynchus keta) using liquid chromatography-mass spectrometry (LC-MS). We developed a quantitative multiple reaction monitoring (MRM)-MS assay with stable-isotope-labeled peptides. This method demonstrated high precision and sensitivity, achieving a limit of detection (LOD) of 0.0008 μg/mL and a lower limit of quantification (LLOQ) of 0.0025 μg/mL. These findings validate that targeting core proteins enables accurate proteoglycan quantification and support its application in quality assessment and development of salmon nasal cartilage-based foods.
Collapse
Affiliation(s)
- Eun Sun Ji
- Bio R&D Center, CellKey Inc., Seoul, 06571, Republic of Korea
| | - Bo Su Lee
- Department of Food Regulatory Science, Korea University, Sejong, 30019, Republic of Korea
| | - Jiyoung Mun
- Bio R&D Center, CellKey Inc., Seoul, 06571, Republic of Korea
| | - Se Yeong Jeon
- R&D Center, Naturetech Co., Ltd., Cheonan, 31257, Republic of Korea
| | - Hye Ryeong Hong
- R&D Center, Naturetech Co., Ltd., Cheonan, 31257, Republic of Korea
| | - Hyun Jin Kim
- R&D Center, Naturetech Co., Ltd., Cheonan, 31257, Republic of Korea
| | - Young Jun Kim
- Department of Food Regulatory Science, Korea University, Sejong, 30019, Republic of Korea
| | - Seon Gil Do
- R&D Center, Naturetech Co., Ltd., Cheonan, 31257, Republic of Korea
| | - Jong Hwan Shin
- Bio R&D Center, CellKey Inc., Seoul, 06571, Republic of Korea
| | - Kwang Hoe Kim
- Bio R&D Center, CellKey Inc., Seoul, 06571, Republic of Korea.
| |
Collapse
|
7
|
Dari S, O'dea RD, Fadai NT. Understanding the regulation of chronic wounds by tissue inhibitors of matrix metalloproteinases through mathematical modelling. J Theor Biol 2025; 604:112083. [PMID: 40020775 DOI: 10.1016/j.jtbi.2025.112083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 02/21/2025] [Accepted: 02/21/2025] [Indexed: 03/03/2025]
Abstract
Understanding the biochemistry and pharmacodynamics of chronic wounds is of key importance, due to the millions of people in the UK affected and the significant cost to the NHS. Chronic wounds are characterised by elevated concentrations of matrix metalloproteinases (MMPs) that destroy the surrounding extracellular matrix (ECM). However, fibroblasts can produce tissue inhibitors of MMPs (TIMPs) in order to regulate wound healing. Therefore, the role of TIMPs in both acute and chronic wounds needs to be properly understood in order to develop therapeutic treatments. In this work, we propose a reaction-diffusion system of four partial differential equations that describe the interaction of the ECM, fibroblasts, MMPs, and TIMPs in a wound. We observe that, subject to parameter sets corresponding to both acute and chronic wound healing, this mathematical model gives rise to travelling wave solutions. Using bifurcation analysis, we demonstrate that excessive degradation of the ECM results in the emergence of chronic wounds, and the reversal of these chronic wounds is prohibited for lower TIMP production values. These results are replicated within a simplified model obtained via a parameter sensitivity analysis. This model is further extended to more realistic spatial domains where we demonstrate the effectiveness of a therapeutic hydrogel containing TIMPs as a treatment for chronic wounds.
Collapse
Affiliation(s)
- Sonia Dari
- School of Mathematical Sciences, University of Nottingham, Nottingham, NG7 2RD, UK.
| | - Reuben D O'dea
- School of Mathematical Sciences, University of Nottingham, Nottingham, NG7 2RD, UK
| | - Nabil T Fadai
- School of Mathematical Sciences, University of Nottingham, Nottingham, NG7 2RD, UK
| |
Collapse
|
8
|
Daghrery A, Dal-Fabbro R, Xu J, Kaigler D, de Ruijter M, Gawlitta D, Malda J, Bottino MC. Niche-inspired collagen infused melt electrowritten scaffolds for craniofacial bone regeneration. BIOMATERIALS ADVANCES 2025; 170:214222. [PMID: 39923603 PMCID: PMC11893008 DOI: 10.1016/j.bioadv.2025.214222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 01/08/2025] [Accepted: 02/03/2025] [Indexed: 02/11/2025]
Abstract
Advances in tissue engineering are focused on devising improved therapeutics to reconstruct craniofacial bones. In cell-based strategies, biomaterials with specific physicochemical properties can mimic natural environments, supporting stem cell renewal, survivability, and cell fate. This study highlights the engineering of a 3D-printed (Melt Electrowritten, MEW) fluorinated‑calcium phosphate (F/CaP)-coated polymeric scaffold infused with collagen (COL) that boosts the performance of transplanted alveolar bone-derived mesenchymal stem cells (aBMSCs). Electron microscopy revealed micron-sized (2.7 μm) polymeric fibers forming a porous (500 μm fiber strand spacing) composite scaffold with a uniform F/CaP coating homogeneously infiltrated with collagen. In vitro, our findings underscored the cytocompatibility of the collagen-infused F/CaP-coated composite scaffold, fostering a suitable environment for aBMSCs proliferation and differentiation. Cells within the F/CaP-coated constructs exhibited upregulated osteogenic gene activity, and the addition of collagen augmented the expression of critical bone-forming genes (i.e., Runx2 and OCN). After in vivo implantation, the scaffolds integrated well with the surrounding host tissue, supporting extensive blood vessel infiltration. Notably, the collagen-infused F/CaP-coated composite scaffolds showed an increased CD31-positive vessel growth compared to the non-coated counterparts. At 8 weeks, aBMSCs-laden F/CaP-Coated+COL composite scaffolds exhibited robust bone formation, creating connecting bony bridges in calvarial defects. Importantly, F/CaP-Coated+COL composite scaffolds displayed pronounced OCN expression, indicating enhanced osteogenic potential. Thus, the engineered F/CaP-coated polymeric scaffold laden with aBMSCs and infused with collagen has proven effective in supporting cell growth, vascularization, and rapid bone regeneration, suggesting potential for future clinical use.
Collapse
Affiliation(s)
- Arwa Daghrery
- Department of Restorative Dental Sciences, School of Dentistry, Jazan University, Jazan, Saudi Arabia; Department of Cariology, Restorative Sciences, and Endodontics, School of Dentistry, University of Michigan, Ann Arbor, MI, United States
| | - Renan Dal-Fabbro
- Department of Cariology, Restorative Sciences, and Endodontics, School of Dentistry, University of Michigan, Ann Arbor, MI, United States
| | - Jinping Xu
- Department of Cariology, Restorative Sciences, and Endodontics, School of Dentistry, University of Michigan, Ann Arbor, MI, United States
| | - Darnell Kaigler
- Department of Periodontics and Oral Medicine, School of Dentistry, University of Michigan, Ann Arbor, MI, USA; Department of Biomedical Engineering, College of Engineering, University of Michigan, Ann Arbor, MI, United States
| | - Mylène de Ruijter
- Regenerative Medicine Center Utrecht, Utrecht, the Netherlands; Department of Orthopedics, University Medical Center Utrecht, Utrecht, the Netherlands; Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, the Netherlands
| | - Debby Gawlitta
- Regenerative Medicine Center Utrecht, Utrecht, the Netherlands; Department of Oral and Maxillofacial Surgery & Special Dental Care (Division of Surgical Specialties), Utrecht University, Utrecht, the Netherlands
| | - Jos Malda
- Regenerative Medicine Center Utrecht, Utrecht, the Netherlands; Department of Orthopedics, University Medical Center Utrecht, Utrecht, the Netherlands; Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, the Netherlands
| | - Marco C Bottino
- Department of Cariology, Restorative Sciences, and Endodontics, School of Dentistry, University of Michigan, Ann Arbor, MI, United States; Department of Biomedical Engineering, College of Engineering, University of Michigan, Ann Arbor, MI, United States.
| |
Collapse
|
9
|
Martin EM, Chang J, González A, Genovese F. Circulating collagen type I fragments as specific biomarkers of cardiovascular outcome risk: Where are the opportunities? Matrix Biol 2025; 137:19-32. [PMID: 40037418 PMCID: PMC11986567 DOI: 10.1016/j.matbio.2025.03.001] [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: 11/22/2024] [Revised: 02/24/2025] [Accepted: 03/01/2025] [Indexed: 03/06/2025]
Abstract
Collagen type I (COL1) is the most abundant protein in the human body and is a main component in the extracellular matrix. The COL1 structure vastly influences normal tissue homeostasis, and changes in the matrix drive progression in multiple diseases. Cardiovascular diseases (CVD) are the leading cause of mortality and morbidity in many Western countries; alterations in the extracellular matrix turnover processes, including COL1, are known to influence the pathophysiological processes leading to CVD outcome. Peptides reflecting COL1 formation and degradation have been established and explored for over two decades in CVD. This review aims to combine and assess the evidence for using COL1-derived circulating peptides as biomarkers in CVD. Secondly, the review identifies existing pitfalls, and evaluates future opportunities for improving the technical characteristics and performance of the biomarkers for implementation in the clinical setting.
Collapse
Affiliation(s)
- Emily M Martin
- Nordic Bioscience A/S, Herlev, Denmark; Institute of Biomedical Science, University of Copenhagen, Copenhagen, Denmark.
| | - Joan Chang
- Manchester Cell-Matrix Centre, Division of Molecular and Cellular Function, University of Manchester, Manchester, UK
| | - Arantxa González
- Centre for Applied Medical Research (CIMA) Universidad de Navarra, Department of Cardiology and Cardiac Surgery, Clínica Universidad de Navarra, Department of Pathology Anatomy and Physiology Universidad de Navarra and IdiSNA, Pamplona, Navarra (Spain); CIBERCV, Instituto de Salud Carlos III, Madrid Spain
| | | |
Collapse
|
10
|
McGee Talkington G, Ouvrier B, White AL, Hall G, Umar M, Bix GJ. Imaging Interstitial Fluids and Extracellular Matrix in Cerebrovascular Disorders: Current Perspectives and Clinical Applications. Neuroimaging Clin N Am 2025; 35:181-189. [PMID: 40210376 PMCID: PMC11995915 DOI: 10.1016/j.nic.2025.01.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/12/2025]
Abstract
This article provides a comprehensive review of current neuroimaging techniques for visualizing and quantifying extracellular matrix (ECM) components and interstitial fluid (ISF) dynamics in cerebrovascular disorders. It examines how alterations in ECM composition and ISF movement patterns correlate with various cerebrovascular pathologies, including ischemic stroke, frontotemporal dementia, cerebral small vessel disease, Alzhheimer's disease, and vascular dementia. The review emphasizes novel imaging markers specific to ECM/ISF alterations and their utility in differentiating various cerebrovascular pathologies. Special attention is given to the clinical applications of these imaging techniques for early disease detection, monitoring progression, and guiding therapeutic interventions.
Collapse
Affiliation(s)
- Grant McGee Talkington
- Department of Neurosurgery, Clinical Neuroscience Research Center, Tulane University School of Medicine, New Orleans, LA 70112, USA; Tulane Brain Institute, Tulane University, New Orleans, LA 70112, USA.
| | - Blake Ouvrier
- Department of Neurosurgery, Clinical Neuroscience Research Center, Tulane University School of Medicine, New Orleans, LA 70112, USA; Tulane Brain Institute, Tulane University, New Orleans, LA 70112, USA
| | - Amanda Louise White
- Department of Neurology, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Gregory Hall
- Department of Neurosurgery, Clinical Neuroscience Research Center, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Meenakshi Umar
- Department of Neurology, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Gregory Jaye Bix
- Department of Neurosurgery, Clinical Neuroscience Research Center, Tulane University School of Medicine, New Orleans, LA 70112, USA; Tulane Brain Institute, Tulane University, New Orleans, LA 70112, USA; Department of Neurology, Tulane University School of Medicine, New Orleans, LA 70112, USA
| |
Collapse
|
11
|
Khalili-Tanha G, Radisky ES, Radisky DC, Shoari A. Matrix metalloproteinase-driven epithelial-mesenchymal transition: implications in health and disease. J Transl Med 2025; 23:436. [PMID: 40217300 PMCID: PMC11992850 DOI: 10.1186/s12967-025-06447-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2025] [Accepted: 03/30/2025] [Indexed: 04/14/2025] Open
Abstract
Epithelial-mesenchymal transition (EMT) is a process in which epithelial cells, defined by apical-basal polarity and tight intercellular junctions, acquire migratory and invasive properties characteristic of mesenchymal cells. Under normal conditions, EMT directs essential morphogenetic events in embryogenesis and supports tissue repair. When dysregulated, EMT contributes to pathological processes such as organ fibrosis, chronic inflammation, and cancer progression and metastasis. Matrix metalloproteinases (MMPs)-a family of zinc-dependent proteases that degrade structural components of the extracellular matrix-sit at the nexus of this transition by dismantling basement membranes, activating pro-EMT signaling pathways, and cleaving adhesion molecules. When normally regulated, MMPs promote balanced ECM turnover and support the cyclical remodeling necessary for proper development, wound healing, and tissue homeostasis. When abnormally regulated, MMPs drive excessive ECM turnover, thereby promoting EMT-related pathologies, including tumor progression and fibrotic disease. This review provides an integrated overview of the molecular mechanisms by which MMPs both initiate and sustain EMT under physiological and disease conditions. It discusses how MMPs can potentiate EMT through TGF-β and Wnt/β-catenin signaling, disrupt cell-cell junction proteins, and potentiate the action of hypoxia-inducible factors in the tumor microenvironment. It discusses how these pathologic processes remodel tissues during fibrosis, and fuel cancer cell invasion, metastasis, and resistance to therapy. Finally, the review explores emerging therapeutic strategies that selectively target MMPs and EMT, ranging from CRISPR/Cas-mediated interventions to engineered tissue inhibitors of metalloproteinases (TIMPs), and demonstrates how such approaches may suppress pathological EMT without compromising its indispensable roles in normal biology.
Collapse
Affiliation(s)
- Ghazaleh Khalili-Tanha
- Department of Medical Genetics and Molecular Medicine, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Evette S Radisky
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL, USA
| | - Derek C Radisky
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL, USA
| | - Alireza Shoari
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL, USA.
| |
Collapse
|
12
|
Arafat Y, Cuesta-Apausa C, Castellano E, Reyes-Aldasoro CC. Fibre tracing in biomedical images: An objective comparison between seven algorithms. PLoS One 2025; 20:e0320006. [PMID: 40209168 PMCID: PMC11984972 DOI: 10.1371/journal.pone.0320006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Accepted: 02/01/2025] [Indexed: 04/12/2025] Open
Abstract
Obtaining the traces and the characteristics of elongated structures is an important task in computer vision pipelines. In biomedical applications, the analysis of traces of vasculature, nerves or fibres of the extracellular matrix can help characterise processes like angiogenesis or the effect of a certain treatment. This paper presents an objective comparison of six existing methodologies (Edge detection, CT Fire, Scale Space, Twombli, U-Net and Graph Based) and one novel approach called Trace Ridges to trace biomedical images with fibre-like structures. Trace Ridges is a fully automatic and fast algorithm that combines a series of image-processing algorithms including filtering, watershed transform and edge detection to obtain an accurate delineation of the fibre-like structures in a rapid time. To compare the algorithms, four biomedical data sets with vastly distinctive characteristics were selected. Ground truth was obtained by manual delineation of the fibre-like structures. Three pre-processing filtering options were used as a first step: no filtering, Gaussian low-pass and DnCnn, a deep-learning filtering. Three distance error metrics (total, average and maximum distance from the obtained traces to the ground truth) and processing time were calculated. It was observed that no single algorithm outperformed the others in all metrics. For the total distance error, which was considered the most significative, Trace Ridges ranked first, followed by Graph Based, U-Net, Twombli, Scale Space, CT Fire and Edge Detection. In terms of speed, Trace Ridges ranked second, only slightly slower than Edge Detection. Code is freely available at github.com/youssefarafat/Trace_Ridges.
Collapse
Affiliation(s)
- Youssef Arafat
- Department of Computer Science, School of Science and Technology, City St George’s, University of London, London, United Kingdom
| | | | - Esther Castellano
- Tumour-Stroma Signalling Lab, Universidad de Salamanca, Salamanca, Spain
| | - Constantino Carlos Reyes-Aldasoro
- Department of Computer Science, School of Science and Technology, City St George’s, University of London, London, United Kingdom
- Integrated Pathology Unit, Division of Molecular Pathology, The Institute of Cancer Research, Sutton, United Kingdom
| |
Collapse
|
13
|
Zhang L, Yuan X, Song R, Yuan Z, Zhao Y, Zhang Y. Engineered 3D mesenchymal stem cell aggregates with multifunctional prowess for bone regeneration: Current status and future prospects. J Adv Res 2025:S2090-1232(25)00227-9. [PMID: 40220897 DOI: 10.1016/j.jare.2025.04.008] [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: 11/17/2024] [Revised: 03/29/2025] [Accepted: 04/05/2025] [Indexed: 04/14/2025] Open
Abstract
BACKGROUND Impaired efficacy of in vitro expanded mesenchymal stem cells (MSCs) is a universal and thorny situation, which cast a shadow on further clinical translation of exogenous MSCs. Moreover, the relatively lengthy healing process, host metabolic heterogeneity and the sophisticated cell recognition and crosstalk pose rigorous challenges towards MSC-based bone regeneration strategies. Three-dimensional (3D) cell aggregates facilitate more robust intercellular communications and cell-extracellular matrix (ECM) interactions, providing a better mimicry of microarchitectures and biochemical milieus in vivo, which is conducive for stemness maintenance and downstream bone formation. AIM OF REVIEW This review enunciates the phenotypic features of MSCs in aggregates, which deepens the knowledge of the MSC fate determination in 3D microenvironment. By summarizing current empowerment methods and biomaterial-combined techniques for establishing functionalized MSC aggregates, this review aims to spark innovative and promising solutions for exalting the translational value of MSCs and improve their therapeutic applications in bone tissue repair. KEY SCIENTIFIC CONCEPTS OF REVIEW 3D aggregates optimize regenerative behaviors of in vitro cultured MSCs including cell adhesion, viability, proliferation, pluripotency and immunoregulation capacity, etc. Biomaterials hybridization endows MSC aggregates with tailored mechanical and biological properties, which offers more possibilities in adapting various clinical scenarios.
Collapse
Affiliation(s)
- Linxue Zhang
- Department of Pediatrics, Peking University School and Hospital of Stomatology, National Center for Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing Key Laboratory of Digital Stomatology, Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health, NMPA Key Laboratory for Dental Materials, Beijing 100081, PR China
| | - Xiaojing Yuan
- Department of Pediatrics, Peking University School and Hospital of Stomatology, National Center for Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing Key Laboratory of Digital Stomatology, Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health, NMPA Key Laboratory for Dental Materials, Beijing 100081, PR China
| | - Rui Song
- Department of Pediatrics, Peking University School and Hospital of Stomatology, National Center for Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing Key Laboratory of Digital Stomatology, Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health, NMPA Key Laboratory for Dental Materials, Beijing 100081, PR China
| | - Zuoying Yuan
- Department of Biomedical Engineering, College of Future Technology, Peking University, Beijing 100871, PR China; Medical Innovation and Research, Peking University Third Hospital, Beijing 100191, PR China.
| | - Yuming Zhao
- Department of Pediatrics, Peking University School and Hospital of Stomatology, National Center for Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing Key Laboratory of Digital Stomatology, Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health, NMPA Key Laboratory for Dental Materials, Beijing 100081, PR China.
| | - Yunfan Zhang
- Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology & Beijing Key Laboratory of Digital Stomatology & Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health & NMPA Key Laboratory for Dental Materials, 22 Zhongguancun South Avenue, Haidian District, Beijing, PR China.
| |
Collapse
|
14
|
Courbot O, Elosegui-Artola A. The role of extracellular matrix viscoelasticity in development and disease. NPJ BIOLOGICAL PHYSICS AND MECHANICS 2025; 2:10. [PMID: 40191103 PMCID: PMC11968406 DOI: 10.1038/s44341-025-00014-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Accepted: 02/14/2025] [Indexed: 04/09/2025]
Abstract
For several decades, research has studied the influence of the extracellular matrix (ECM) mechanical properties in cell response, primarily emphasising its elasticity as the main determinant of cell and tissue behaviour. However, the ECM is not purely elastic; it is viscoelastic. ECM viscoelasticity has now emerged as a major regulator of collective cell dynamics. This review highlights recent findings on the role of ECM viscoelasticity in development and pathology.
Collapse
Affiliation(s)
- Olivia Courbot
- Cell and Tissue Mechanobiology Laboratory, The Francis Crick Institute, London, UK
- Department of Physics, King’s College London, London, UK
| | - Alberto Elosegui-Artola
- Cell and Tissue Mechanobiology Laboratory, The Francis Crick Institute, London, UK
- Department of Physics, King’s College London, London, UK
| |
Collapse
|
15
|
Bai S, Wei B, Chen L, Huang X, Huang K, Yang L, Zheng C, Wang Y. Drug-Loaded Hybrid Tissue Engineered Heart Valve with Antithrombosis and Immunomodulation Performance. ACS APPLIED MATERIALS & INTERFACES 2025; 17:19401-19416. [PMID: 40119842 DOI: 10.1021/acsami.4c22022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/24/2025]
Abstract
High thrombogenicity and shortened lifespan have limited the application of mechanical valves and bioprosthetic valves, respectively. Tissue engineering heart valve (TEHV) holds significant potential as a favorable prosthetic valve to overcome the limitations of the current prosthetic valves, featuring the capabilities of self-pairing and adaptive remodeling. However, TEHVs, mainly fabricated from decellularized xenogeneic heart valves (DHV), still have challenges such as thrombosis, inferior endothelialization, and immune responses. Herein, a drug-loaded glycoprotein-like network hybrid TEHV (OHSC-V) was engineered through the one-pot hybridization of DHV, oxidized HA (OHA), phenylboronic acid grafted silk fibroin (SF-PBA), and curcumin (Cur), where OHA served as a biocompatible backbone to cross-link the DHV and the conjugate of SF-PBA and Cur. With the introduction of the multifunctional drug-loaded glycoprotein-like network, OHSC-V not only effectively inhibited the adsorption of plasma proteins, blood cells, platelets, and thrombosis but also facilitated the endothelialization of TEHV. Furthermore, the OHSC-V eliminated the reactive oxygen species and responsively released Cur to modulate the immune responses. Moreover, the calcification degree of hybrid TEHVs was markedly lower than that of glutaraldehyde cross-linked DHV after 90 days of implantation. Overall, OHSC-V demonstrated enhanced performance of antithrombosis, endothelialization, immunomodulation, and anticalcification, showcasing the further potential for application exploration.
Collapse
Affiliation(s)
- Shaoge Bai
- National Engineering Research Center for Biomaterials& College of Biomedical Engineering, Sichuan University, Chengdu 610065, China
| | - Bangquan Wei
- National Engineering Research Center for Biomaterials& College of Biomedical Engineering, Sichuan University, Chengdu 610065, China
| | - Lepeng Chen
- National Engineering Research Center for Biomaterials& College of Biomedical Engineering, Sichuan University, Chengdu 610065, China
| | - Xueyu Huang
- National Engineering Research Center for Biomaterials& College of Biomedical Engineering, Sichuan University, Chengdu 610065, China
| | - Kaiyang Huang
- National Engineering Research Center for Biomaterials& College of Biomedical Engineering, Sichuan University, Chengdu 610065, China
| | - Li Yang
- National Engineering Research Center for Biomaterials& College of Biomedical Engineering, Sichuan University, Chengdu 610065, China
| | - Cheng Zheng
- National Engineering Research Center for Biomaterials& College of Biomedical Engineering, Sichuan University, Chengdu 610065, China
| | - Yunbing Wang
- National Engineering Research Center for Biomaterials& College of Biomedical Engineering, Sichuan University, Chengdu 610065, China
- Research Unit of Minimally Invasive Treatment of Structural Heart Disease, Chinese Academy of Medical Sciences, No.: 2021RU013, Chengdu 610064, China
| |
Collapse
|
16
|
Freitas-Ribeiro S, Carvalho AF, Rodrigues DB, Martins L, Pires RA, Mendes VM, Manadas B, Jarnalo M, Horta R, Reis RL, Pirraco RP. Cryogenic, but not hypothermic, preservation disrupts the extracellular matrix of cell sheets. Bioact Mater 2025; 46:301-310. [PMID: 39811467 PMCID: PMC11732602 DOI: 10.1016/j.bioactmat.2024.12.019] [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: 08/07/2024] [Revised: 11/11/2024] [Accepted: 12/17/2024] [Indexed: 01/16/2025] Open
Abstract
Cell sheet (CS)-based approaches hold significant potential for tissue regeneration, relying on the extracellular matrix (ECM) for success. Like in native tissues, the ECM provides structural and biochemical support for cellular homeostasis and function. Effective preservation strategies that maintain ECM integrity are critical to enhance the therapeutic potential of CS-based approaches. While cryogenic and hypothermic preservation methods offer potential solutions, their impact on CS ECM structure is not fully understood. Therefore, a comprehensive analysis of the ECM of hASCs CS following cryogenic and hypothermic preservation for 3 and 7 days, was conducted. Although proteomic analysis indicated that cryopreservation had no significant effect on the overall composition of the ECM, it induced significant ECM structural alterations, particularly disrupting collagen organization, which was not observed following hypothermic preservation. These structural changes were accompanied by alterations in mechanical properties, including a reduction in elastic modulus. In contrast, hypothermic preservation maintained ECM integrity and mechanical properties similar to the control. The notable ECM structural changes following cryogenic preservation can potentially impact cellular behavior, including adhesion, proliferation, and differentiation, thereby affecting the efficacy of CS therapies in vivo. This suggests that hypothermia may offer a promising alternative to cryopreservation for preserving CS integrity and functionality.
Collapse
Affiliation(s)
- Sara Freitas-Ribeiro
- 3B's Research Group, I3Bs – Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Rua Ave 1, Edifício 1 (Sede), 4805-694 Barco, Guimarães, Portugal
- ICVS/3B's–PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Andreia F. Carvalho
- 3B's Research Group, I3Bs – Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Rua Ave 1, Edifício 1 (Sede), 4805-694 Barco, Guimarães, Portugal
- ICVS/3B's–PT Government Associate Laboratory, Braga/Guimarães, Portugal
- Instituto de Ciências Biomédicas Abel Salazar (ICBAS), Universidade Do Porto, Porto, Portugal
| | - Daniel B. Rodrigues
- 3B's Research Group, I3Bs – Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Rua Ave 1, Edifício 1 (Sede), 4805-694 Barco, Guimarães, Portugal
- ICVS/3B's–PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Luís Martins
- 3B's Research Group, I3Bs – Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Rua Ave 1, Edifício 1 (Sede), 4805-694 Barco, Guimarães, Portugal
- ICVS/3B's–PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Ricardo A. Pires
- 3B's Research Group, I3Bs – Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Rua Ave 1, Edifício 1 (Sede), 4805-694 Barco, Guimarães, Portugal
- ICVS/3B's–PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Vera M. Mendes
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| | - Bruno Manadas
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
- Institute for Interdisciplinary Research, University of Coimbra, Coimbra, Portugal
| | - Mariana Jarnalo
- Department of Plastic and Reconstructive Surgery, and Burn Unity, Centro Hospitalar de São João, Porto, Portugal
- Faculty of Medicine - University of Porto, Portugal
| | - Ricardo Horta
- Department of Plastic and Reconstructive Surgery, and Burn Unity, Centro Hospitalar de São João, Porto, Portugal
- Faculty of Medicine - University of Porto, Portugal
| | - Rui L. Reis
- 3B's Research Group, I3Bs – Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Rua Ave 1, Edifício 1 (Sede), 4805-694 Barco, Guimarães, Portugal
- ICVS/3B's–PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Rogério P. Pirraco
- 3B's Research Group, I3Bs – Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Rua Ave 1, Edifício 1 (Sede), 4805-694 Barco, Guimarães, Portugal
- ICVS/3B's–PT Government Associate Laboratory, Braga/Guimarães, Portugal
| |
Collapse
|
17
|
Chirivì M, Maiullari F, Milan M, Ceraolo MG, Fratini N, Fasciani A, Bousselmi S, Stirm M, Scalera F, Gervaso F, Villa M, Viganò R, Brambilla F, Mauri P, De Falco E, Silvestre DD, Costantini M, Wolf E, Bearzi C, Rizzi R. Mimicking the Dystrophic Cardiac Extracellular Environment through DystroGel. Adv Healthc Mater 2025; 14:e2404251. [PMID: 39962811 PMCID: PMC11973943 DOI: 10.1002/adhm.202404251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 02/03/2025] [Indexed: 04/08/2025]
Abstract
Advances in understanding the mechanisms behind genetic diseases like Duchenne muscular dystrophy (DMD) underscore the critical role of the extracellular matrix (ECM) composition in disease progression. Effective in vitro models must replicate the intercellular relationships and physicochemical properties of native ECM to fully capture disease-specific characteristics. Although recent biomaterials support the in vitro biofabrication of pathophysiological environments, they often lack disease-specific ECM features. In this study, DystroGel, a hydrogel derived from the cardiac ECM of a porcine DMD model, replicates the distinct molecular composition of dystrophic cardiac tissue for the first time. The findings indicate that the dystrophic ECM matrix exhibits a unique protein profile, impacting cellular processes critical to DMD pathology. This work demonstrates the importance of using a 3D substrate that recreates intercellular dynamics within a defined pathological environment, enhancing the ability to model genetic disorders and providing a valuable tool for advancing personalized therapeutic strategies.
Collapse
Affiliation(s)
- Maila Chirivì
- Department of Molecular MedicineSapienza UniversityViale Regina Elena, 324Rome00161Italy
- Neurology UnitFondazione IRCCS Ca' Granda Ospedale Maggiore PoliclinicoVia Francesco Sforza, 35Milan20122Italy
| | - Fabio Maiullari
- Ph.D. Program in Cellular and Molecular BiologyDepartment of BiologyUniversity of Rome “Tor Vergata”Via della Ricerca Scientifica, 1Rome00133Italy
| | - Marika Milan
- Neurology UnitFondazione IRCCS Ca' Granda Ospedale Maggiore PoliclinicoVia Francesco Sforza, 35Milan20122Italy
| | - Maria Grazia Ceraolo
- Neurology UnitFondazione IRCCS Ca' Granda Ospedale Maggiore PoliclinicoVia Francesco Sforza, 35Milan20122Italy
| | - Nicole Fratini
- Department of Molecular MedicineSapienza UniversityViale Regina Elena, 324Rome00161Italy
| | - Alessandra Fasciani
- Fondazione Istituto Nazionale di Genetica Molecolare “Romeo ed Enrica Invernizzi”Via Francesco Sforza, 35Milan20122Italy
| | - Salma Bousselmi
- Neurology UnitFondazione IRCCS Ca' Granda Ospedale Maggiore PoliclinicoVia Francesco Sforza, 35Milan20122Italy
- Ph.D. Program in Cellular and Molecular BiologyDepartment of BiologyUniversity of Rome “Tor Vergata”Via della Ricerca Scientifica, 1Rome00133Italy
| | - Michael Stirm
- Chair for Molecular Animal Breeding and BiotechnologyGene Center and Department of Veterinary SciencesLMU Munich81377MunichGermany
- Center for Innovative Medical Models (CiMM)Department of Veterinary SciencesLMU Munich85764OberschleißheimGermany
| | - Francesca Scalera
- Institute of NanotechnologyNational Research Councilc/o Campus Ecoteknevia MonteroniLecce73100Italy
| | - Francesca Gervaso
- Institute of NanotechnologyNational Research Councilc/o Campus Ecoteknevia MonteroniLecce73100Italy
| | - Michela Villa
- Fondazione Istituto Nazionale di Genetica Molecolare “Romeo ed Enrica Invernizzi”Via Francesco Sforza, 35Milan20122Italy
- Department of BiosciencesUniversity of MilanVia Celoria, 26Milan20133Italy
| | - Raffaello Viganò
- Institute for Biomedical TechnologiesNational Research CouncilVia Fratelli Cervi, 93, SegrateMilan20054Italy
| | - Francesca Brambilla
- Institute for Biomedical TechnologiesNational Research CouncilVia Fratelli Cervi, 93, SegrateMilan20054Italy
| | - Pierluigi Mauri
- Institute for Biomedical TechnologiesNational Research CouncilVia Fratelli Cervi, 93, SegrateMilan20054Italy
| | - Elena De Falco
- Institute for Biomedical TechnologiesNational Research CouncilVia Fratelli Cervi, 93, SegrateMilan20054Italy
| | - Dario Di Silvestre
- Department of BiosciencesUniversity of MilanVia Celoria, 26Milan20133Italy
| | - Marco Costantini
- Institute of Physical Chemistry – Polish Academy of SciencesMarcina Kasprzaka 44/52Warsaw01–224Poland
| | - Eckhard Wolf
- Chair for Molecular Animal Breeding and BiotechnologyGene Center and Department of Veterinary SciencesLMU Munich81377MunichGermany
| | - Claudia Bearzi
- Institute for Biomedical TechnologiesNational Research CouncilVia Fratelli Cervi, 93, SegrateMilan20054Italy
| | - Roberto Rizzi
- Department of Medical‐Surgical Sciences and BiotechnologiesSapienza University of RomeC.so della Repubblica 79Latina04100Italy
| |
Collapse
|
18
|
Al-Zehhawi PAM, Mohammed Jaddoa NT. Linezolid versus vancomycin in vitro activity against methicillin-resistant Staphylococcus aureus biofilms. Microb Pathog 2025; 201:107371. [PMID: 39947358 DOI: 10.1016/j.micpath.2025.107371] [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: 09/25/2024] [Revised: 01/29/2025] [Accepted: 02/09/2025] [Indexed: 02/22/2025]
Abstract
Most microorganisms as well as bacteria live in a community under natural conditions. Bacteria adopted to biofilm mode of life more than 3 billion years ago to survive extreme, harsh environments. They become harmful when they acquire resistance to antibiotics and overcome the standard therapies, which is most commonly found in hospitals. Therefore, many studies have been published regarding antimicrobial resistance (AMR). Staphylococcus aureus is a dangerous pathogen, ubiquitously prevalent as a commensal and opportunistic microorganism in human populations. Methicillin-resistant Staphylococcus aureus (MRSA) is considered one of the major medical problems worldwide since they are frequent colonizers of implanted medical devices causing a variety of hospital-acquired infections. For many years, vancomycin has been the drug of choice for MRSA whereas linezolid is considered the last resort drug. This comparative, cross-sectional study investigated the effects of linezolid on biofilm formation in vitro compared to vancomycin across 85 MRSA isolates. To our knowledge, this is the first study to report high levels of linezolid resistance in MRSA in Iraq. In this brief report, 5 MRSA strains showed resistance to linezolid, with minimum inhibitory concentration (MIC) values of 256 μg/ml. The exact same isolates exhibited vancomycin resistance with MIC values of 1024 μg/ml. All linezolid-resistant MRSA (LR-MRSA) strains demonstrated biofilm formation ability. Additionally, linezolid inhibited the expression of adhesion-related genes cna and clfB. The authors concluded that linezolid exerts a comparable effect to vancomycin in biofilm treatment.
Collapse
|
19
|
Sencha LM, Karpova MA, Dobrynina OE, Balalaeva IV. Cell-type dependent effect of 3D collagen matrix on cancer cell resistance to suboptimal conditions: the case of serum deprivation, glucose starvation, and hypoxia. Tissue Cell 2025; 93:102719. [PMID: 39823703 DOI: 10.1016/j.tice.2024.102719] [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: 09/03/2024] [Revised: 12/13/2024] [Accepted: 12/29/2024] [Indexed: 01/20/2025]
Abstract
The extracellular matrix (ECM) and its primary chemical components, including collagen, play a pivotal role in carcinogenesis and tumor progression. The ECM actively regulates cell proliferation, migration, and, importantly, resistance to various adverse factors. It is widely recognized as a key factor in modifying the resistance of tumor cells to various treatment modalities and cytotoxic compounds. However, the role of the ECM in tumor cell adaptation to nutritional deficiencies and hypoxic conditions remains significantly less studied. Since it is generally accepted that tumor cells resistance increases when cultured in a three-dimensional matrix, we sought to experimentally test the universality of this statement. In this work, we analyzed the responses of tumor cells with varying origins and proliferative activities, including human bladder carcinoma, epidermoid carcinoma, and ovarian carcinoma, to deprivation of serum, glucose and oxygen. We compared cell resistance to suboptimal conditions when cultured in a monolayer on tissue culture (TC)-treated polystyrene, on collagen-coated surfaces, or within a three-dimensional hydrogel composed of collagen type I. All three cell lines were stably transfected with fluorescent protein genes. To register the cell growth dynamics, we used a fluorescence-based technique that allows long-term quantitative observations without disrupting the hydrogel. The analyzed cell lines demonstrated different patterns of relative sensitivity to suboptimal conditions. We revealed that the direction and intensity of the collagen matrix effect depend on the cell type. Slowly proliferating ovarian carcinoma cells showed no noticeable changes in their behavior when cultured in a gel compared to a monolayer. In the case of bladder carcinoma, we registered predominantly resistance-stimulating effect of the collagen matrix, but it was significant only under serum deprivation. The most pronounced effect of collagen was registered for epidermoid carcinoma. Importantly, this effect was ambivalent: gel-embedded cells demonstrated significantly enhanced resistance to serum deprivation, but, at the same time, they were more responsive to glucose starvation and hypoxic conditions. We attribute the registered phenomenon to the individual characteristics of tumor cells with different origins and metabolic activities.
Collapse
Affiliation(s)
- Ludmila M Sencha
- Institute of Biology and Biomedicine, Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod, Russia
| | - Maria A Karpova
- Institute of Biology and Biomedicine, Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod, Russia
| | - Olga E Dobrynina
- Institute of Biology and Biomedicine, Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod, Russia
| | - Irina V Balalaeva
- Institute of Biology and Biomedicine, Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod, Russia.
| |
Collapse
|
20
|
Zhu T, Hu P, Mi Y, Zhang J, Xu A, Gao M, Zhang Y, Shen S, Yang G, Pan Y. Telomerase reverse transcriptase gene knock-in unleashes enhanced longevity and accelerated damage repair in mice. Aging Cell 2025; 24:e14445. [PMID: 39660787 PMCID: PMC11984681 DOI: 10.1111/acel.14445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 10/24/2024] [Accepted: 11/13/2024] [Indexed: 12/12/2024] Open
Abstract
While previous research has demonstrated the therapeutic efficacy of telomerase reverse transcriptase (TERT) overexpression using adeno-associated virus and cytomegalovirus vectors to combat aging, the broader implications of TERT germline gene editing on the mammalian genome, proteomic composition, phenotypes, lifespan extension, and damage repair remain largely unexplored. In this study, we elucidate the functional properties of transgenic mice carrying the Tert transgene, guided by precise gene targeting into the Rosa26 locus via embryonic stem (ES) cells under the control of the elongation factor 1α (EF1α) promoter. The Tert knock-in (TertKI) mice harboring the EF1α-Tert gene displayed elevated telomerase activity, elongated telomeres, and extended lifespan, with no spontaneous genotoxicity or carcinogenicity. The TertKI mice showed also enhanced wound healing, characterized by significantly increased expression of Fgf7, Vegf, and collagen. Additionally, TertKI mice exhibited robust resistance to the progression of colitis induced by dextran sodium sulfate (DSS), accompanied by reduced expression of disease-deteriorating genes. These findings foreshadow the potential of TertKI as an extraordinary rejuvenation force, promising not only longevity but also rejuvenation in skin and intestinal aging.
Collapse
Affiliation(s)
- Tian‐Yi Zhu
- School of PharmacyNanjing University of Chinese MedicineNanjingJiangsuChina
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain InstituteThe University of QueenslandBrisbaneQueenslandAustralia
| | - Po Hu
- School of PharmacyNanjing University of Chinese MedicineNanjingJiangsuChina
| | - Yu‐Hui Mi
- School of PharmacyNanjing University of Chinese MedicineNanjingJiangsuChina
| | - Jun‐Li Zhang
- School of PharmacyNanjing University of Chinese MedicineNanjingJiangsuChina
| | - An‐Na Xu
- School of PharmacyNanjing University of Chinese MedicineNanjingJiangsuChina
| | - Ming‐Tong Gao
- School of PharmacyNanjing University of Chinese MedicineNanjingJiangsuChina
| | - Ying‐Ying Zhang
- School of PharmacyNanjing University of Chinese MedicineNanjingJiangsuChina
| | - San‐Bing Shen
- Regenerative Medicine Institute, School of MedicineUniversity of GalwayGalwayIreland
| | - Guang‐Ming Yang
- School of PharmacyNanjing University of Chinese MedicineNanjingJiangsuChina
| | - Yang Pan
- School of PharmacyNanjing University of Chinese MedicineNanjingJiangsuChina
| |
Collapse
|
21
|
Füge L, Schüssler F, Gerhardus J, Schwab R, Harms G, Hasenburg A, Blaeser A, Brenner W, Peters K. Comparative Analysis of Hydrogels From Porcine Extracellular Matrix for 3D Bioprinting of Adipose Tissue. J Biomed Mater Res A 2025; 113:e37832. [PMID: 40165526 DOI: 10.1002/jbm.a.37832] [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: 07/31/2024] [Revised: 10/19/2024] [Accepted: 10/24/2024] [Indexed: 04/02/2025]
Abstract
The extracellular matrix (ECM) is the natural scaffold of all soft tissues in tissue engineering. Of special interest is the use of ECM as a hydrogel, which can be used to enclose cells and to be molded into any form by 3D bioprinting. Protocols for the preparation of ECM vary in the use of physical and chemical processing steps, the use of different detergents for decellularization, and the removal of DNA and RNA residues and show a different use of solvents and wash buffers. We have, therefore, compared seven different variations for the decellularization of a primary porcine isolate to manufacture decellularized adipose tissue (DAT) for their use in adipose tissue engineering and as a hydrogel in particular. Decellularization efficacy was assessed by DNA quantification while retention of ECM components was evaluated by measuring the content of hydroxyproline and glycosaminoglycan (GAGs). Depending on the decellularization protocol, the composition and DNA content of the resulting DAT were different. All DAT samples were processed into hydrogels to assess their mechanical properties as well as their influence on cellular metabolic activity and cell differentiation. The different compositions of the DAT and the resulting hydrogels had an effect on the stability and printability of the gels. Some DAT that were digested with hydrochloric acid (HCl) were more stable than those that were digested with acetic acid (AA). In addition, depending on the protocol, there was a clear effect on adipose-derived stem cells (ASC), endothelial cells and fibroblasts, cultured with the hydrogels. The differentiation of ASC to adipocytes could be achieved on most of the hydrogels. Human dermal microvascular endothelial cells (HDMEC) showed significantly better metabolic activity on hydrogels digested with HCl than digested with AA. HDMEC cultured on hydrogel #2 digested with HCl showed a 40% higher metabolic activity compared to collagen as a positive control, whereas culturing HDMEC on hydrogel #2 digested with AA resulted in a cellular metabolic activity loss of 60%. In a triculture of all three cell types, the formation of first tubular networks by HDMEC was achieved depending on the hydrogel used.
Collapse
Affiliation(s)
- Leonie Füge
- Department of Obstetrics and Women's Health, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Felix Schüssler
- Department of Obstetrics and Women's Health, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Jamina Gerhardus
- BioMedical Printing Technology, Department of Mechanical Engineering, Technical University of Darmstadt, Darmstadt, Germany
| | - Roxana Schwab
- Department of Obstetrics and Women's Health, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Gregory Harms
- Cell Biology Unit, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
- Department of Biology, Wilkes University, Wilkes Barre, Pennsylvania, USA
| | - Annette Hasenburg
- Department of Obstetrics and Women's Health, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Andreas Blaeser
- BioMedical Printing Technology, Department of Mechanical Engineering, Technical University of Darmstadt, Darmstadt, Germany
- Centre for Synthetic Biology, Technical University of Darmstadt, Darmstadt, Germany
| | - Walburgis Brenner
- Department of Obstetrics and Women's Health, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
- BiomaTiCS - Biomaterials, Tissues and Cells in Science, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Katharina Peters
- Department of Obstetrics and Women's Health, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
- BiomaTiCS - Biomaterials, Tissues and Cells in Science, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| |
Collapse
|
22
|
Fan Y, Moser J, Jongman RM, Borghuis T, Vonk JM, Timens W, van Meurs M, Pillay J, Burgess JK. Compositional changes of the lung extracellular matrix in acute respiratory distress syndrome. Am J Physiol Cell Physiol 2025; 328:C1279-C1292. [PMID: 40063067 DOI: 10.1152/ajpcell.01007.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 01/13/2025] [Accepted: 03/04/2025] [Indexed: 04/04/2025]
Abstract
Acute respiratory distress syndrome (ARDS) is pathologically characterized by diffuse alveolar damage (DAD) and is associated with high morbidity and mortality rates. Although pulmonary injury initiates alveolar-capillary barrier damage in ARDS, remodeling of the extracellular matrix (ECM), which is pivotal for both tissue repair and organ recovery, may play a large role in persistent ARDS. This study investigated the compositional changes in the ECM in different DAD stages in ARDS. Paraffin-embedded lung sections collected during autopsy or from posttransplant lungs were obtained from patients with ARDS (n = 28) admitted to the University Medical Center Groningen between 2010 and 2020. Sections were stained histochemically, and immunohistochemically for collagen III α1 chain (Col IIIa1), IV α3 chain (Col IVa3), VI α1 chain (Col VIa1), periostin (PSTN), lumican (LUM), and fibronectin (FN). The sections were divided into 118 regions based on DAD stages (54 early vs. 64 advanced). The differences in the expression of selected proteins were compared between DAD stages or across ARDS duration (<7 days, 7-14 days, and >14 days). The fiber pattern of Col VIa1 was analyzed using CellProfiler. Higher tissue density, lower proportional areas of Col IIIa1, Col IVa3, and LUM, and more concentrated Col VIa1 fibers were observed in the advanced DAD stage than in the early DAD stage. Areas with higher proportions of total collagen and FN, and lower proportional areas of Col IIIa1, Col IVa3, and LUM were detected in lung regions from patients with ARDS >14-days duration. These findings revealed proportional changes in ECM components, strongly suggesting that dynamic changes in ECM proteins play a role in pathophysiology in ARDS during progression.NEW & NOTEWORTHY Our study revealed ECM protein compositional differences in lung parenchyma between stages of DAD. In advanced DAD, tissue density was higher, but collagen type III, type IV, and lumican were proportionally lower compared with early DAD. The organization of collagen type VI fibers was highly concentrated in advanced DAD. Our results indicate that both composition and organization of ECM were remodeled in advanced DAD, suggesting a role in manifesting acute respiratory distress syndrome.
Collapse
Affiliation(s)
- YiWen Fan
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
- Department of Critical Care, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
- Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Jill Moser
- Department of Critical Care, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Rianne M Jongman
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
- Department of Anesthesiology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Theo Borghuis
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Judith M Vonk
- Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
- Department of Epidemiology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Wim Timens
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
- Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Matijs van Meurs
- Department of Critical Care, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Janesh Pillay
- Department of Critical Care, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
- Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Janette K Burgess
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
- Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| |
Collapse
|
23
|
Mollenkopf P, Kochanowski JA, Ren Y, Vining KH, Janmey PA, Purohit PK. Poroelasticity and permeability of fibrous polymer networks under compression. SOFT MATTER 2025; 21:2400-2412. [PMID: 39976571 PMCID: PMC11841696 DOI: 10.1039/d4sm01223b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Accepted: 01/24/2025] [Indexed: 02/23/2025]
Abstract
Soft biopolymer networks play pivotal roles in governing cellular mechanics, tissue structure, and physiological processes such as blood coagulation. Understanding their permeability and mechanical responses under compression is crucial for elucidating mass transport phenomena and their impact on extra- and intra-cellular behavior as well as processes affecting functionality of blood clots, cartilage and other fibrous tissues. The nonlinear responses of these networks to mechanical stresses prevent application of established linear poro-elasticity models. Despite extensive studies of fibrous network viscoelastic properties under shear deformations, their dynamic responses to compressive deformations remain poorly understood, particularly in physiological contexts of growth and collective migration of solid bodies. Conventional experimental techniques face challenges in accurately evaluating the permeability of these networks, hindering comprehensive understanding of their poromechanical behavior. In this study, we employ a novel poroelastic hybrid approach combining rheometer-based compression rheology with camera-facilitated sample shape detection to directly measure fluid flux and network permeability under controlled compressive strains. Accompanying experimental investigations, a continuum model implemented in finite elements, and an analytical model are developed to interpret the findings. The experimental data align well with the analytical model, revealing the emergence and disappearance of distinct densification regimes within the gel under mechanical stress. This study advances our understanding of the intricate interplay between mechanical forces, fluid flow, and structural properties in soft biopolymer networks, with a specific focus on fibrin- and collagen-based gels which represent the most abundant protein networks in the extracellular environment.
Collapse
Affiliation(s)
- Paul Mollenkopf
- Department of Physiology, University of Pennsylvania, Philadelphia, PA, 19104, USA.
| | - Jakub A Kochanowski
- Department of Physiology, University of Pennsylvania, Philadelphia, PA, 19104, USA.
| | - Yifei Ren
- Department of Mechanical Engineering and Applied Mechanics, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Kyle H Vining
- Department of Preventive and Restorative Sciences, Department of Materials Science and Engineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Paul A Janmey
- Department of Physiology, University of Pennsylvania, Philadelphia, PA, 19104, USA.
- Department of Physics & Astronomy, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Prashant K Purohit
- Department of Mechanical Engineering and Applied Mechanics, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
24
|
Deng Y, He Y, Xu J, He H, Zhang M, Li G. Cardiac Fibroblasts regulate myocardium and coronary vasculature development via the collagen signaling pathway. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.09.11.612512. [PMID: 39314489 PMCID: PMC11418987 DOI: 10.1101/2024.09.11.612512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/25/2024]
Abstract
The fibroblast (FB), cardiomyocyte (CM), and vascular endothelial cell (Vas_EC) are the three major cell types in the heart, yet their relationships during development are largely unexplored. To address this gap, we employed RNA staining of the FB marker gene Col1a1 together with the CM marker gene Actn2 and the Vas_EC marker gene Cdh5 at different stages. This approach enabled us to discern the anatomical pattern of cardiac FBs and identify approximately one EC and four CMs directly interacting with each FB. Molecularly, through the analysis of single-cell mRNA sequencing (scRNA-seq) data, we unveiled collagen as the top signaling molecule derived from FBs influencing CM and Vas_EC development. Subsequently, we used a Pdgfra-CreER controlled diphtheria toxin A (DTA) system to ablate the FBs at different stages. We found that the ablation of FBs disrupted myocardium and vasculature development and led to embryonic heart defects. Using scRNA-seq, we further profiled the ablated hearts and identified molecular defects in their ventricular CMs and Vas_ECs compared to control hearts. Moreover, we identified a reduction of collagen in the ablated hearts and predicted collagen as the major signaling pathway regulating the differentially expressed genes in the ablated ventricular CMs. Finally, we performed both short-term and long-term fibroblast ablation at the neonatal stage. We found that short-term ablation caused a reduction in collagen and Vas_EC density, while long-term ablation may induce compensatory collagen expression without causing heart function reduction. In summary, our study has identified the function of fibroblasts in regulating myocardium and vasculature development and implicated an important role for the collagen pathway in this process.
Collapse
Affiliation(s)
- Yiting Deng
- Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15201, USA
| | - Yuanhang He
- Tsinghua University, Tsinghua medicine, School of Medicine, Beijing, China
- Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15201, USA
| | - Juan Xu
- Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15201, USA
| | - Haoting He
- Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15201, USA
| | - Manling Zhang
- Vascular Medicine Institute, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Guang Li
- Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15201, USA
| |
Collapse
|
25
|
Ling Z, Niego B, Li Q, Villa VS, Bhattaram D, Hu M, Gong Z, Smith LM, Frey BL, Ren X. Chemoselective Characterization of New Extracellular Matrix Deposition in Bioengineered Tumor Tissues. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.18.643336. [PMID: 40166338 PMCID: PMC11956949 DOI: 10.1101/2025.03.18.643336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
The extracellular matrix (ECM), present in nearly all tissues, provides extensive support to resident cells through structural, biomechanical, and biochemical means, and in return the ECM undergoes constant remodeling from interacting cells to adapt to the evolving tissue states. Bioengineered 3D tissues, commonly known as cell-ECM composites, are robust model systems to recapitulate and investigate native pathophysiology. Key to this engineered morphogenesis process are the intricate cell-ECM interactions reflected by how cells respond to and thereby modulate their surrounding microenvironments through their ongoing ECM secretome. However, investigating ECM-regulated new ECM production has been challenging due to the proteomic background from the pre-existing biomaterial ECM. To address this hindrance, here we present a chemoselective strategy to label, enrich, and characterize newly synthesized ECM (newsECM) proteins produced by resident cells, allowing distinction from the pre-existing ECM background. Applying our analytical pipeline to bioengineered tumor tissues, either built upon decellularized ECM (dECM-tumors) or as ECM-free tumor spheroids (tumoroids), we observed distinct ECM synthesis patterns that were linked to their extracellular environments. Tumor cells responded to the dECM presence with elevated ECM remodeling activities, mediated by augmented digestion of pre-existing ECM coupled with upregulated synthesis of tumor-associated ECM. Our findings highlight the sensitivity of newsECM profiling to capture remodeling events that are otherwise under-represented by bulk proteomics and underscore the significance of dECM support for enabling native-like tumor cell behaviors. We anticipate the described newsECM analytical pipeline to be broadly applicable to other tissue-engineered systems to probe ECM-regulated ECM synthesis and remodeling, both fundamental aspects of cell-ECM crosstalk in engineered tissue morphogenesis.
Collapse
Affiliation(s)
- Zihan Ling
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, Pennsylvania, United States
| | - Burke Niego
- Department of Chemistry, University of Wisconsin, Madison, Wisconsin, United States
| | - Qingyang Li
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, Pennsylvania, United States
| | - Vanessa Serna Villa
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, Pennsylvania, United States
| | - Dhruv Bhattaram
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, Pennsylvania, United States
| | - Michael Hu
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, Pennsylvania, United States
| | - Zhuowei Gong
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, Pennsylvania, United States
| | - Lloyd M. Smith
- Department of Chemistry, University of Wisconsin, Madison, Wisconsin, United States
| | - Brian L. Frey
- Department of Chemistry, University of Wisconsin, Madison, Wisconsin, United States
| | - Xi Ren
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, Pennsylvania, United States
| |
Collapse
|
26
|
Lin LL, Alvarez-Puebla R, Liz-Marzán LM, Trau M, Wang J, Fabris L, Wang X, Liu G, Xu S, Han XX, Yang L, Shen A, Yang S, Xu Y, Li C, Huang J, Liu SC, Huang JA, Srivastava I, Li M, Tian L, Nguyen LBT, Bi X, Cialla-May D, Matousek P, Stone N, Carney RP, Ji W, Song W, Chen Z, Phang IY, Henriksen-Lacey M, Chen H, Wu Z, Guo H, Ma H, Ustinov G, Luo S, Mosca S, Gardner B, Long YT, Popp J, Ren B, Nie S, Zhao B, Ling XY, Ye J. Surface-Enhanced Raman Spectroscopy for Biomedical Applications: Recent Advances and Future Challenges. ACS APPLIED MATERIALS & INTERFACES 2025; 17:16287-16379. [PMID: 39991932 DOI: 10.1021/acsami.4c17502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/25/2025]
Abstract
The year 2024 marks the 50th anniversary of the discovery of surface-enhanced Raman spectroscopy (SERS). Over recent years, SERS has experienced rapid development and became a critical tool in biomedicine with its unparalleled sensitivity and molecular specificity. This review summarizes the advancements and challenges in SERS substrates, nanotags, instrumentation, and spectral analysis for biomedical applications. We highlight the key developments in colloidal and solid SERS substrates, with an emphasis on surface chemistry, hotspot design, and 3D hydrogel plasmonic architectures. Additionally, we introduce recent innovations in SERS nanotags, including those with interior gaps, orthogonal Raman reporters, and near-infrared-II-responsive properties, along with biomimetic coatings. Emerging technologies such as optical tweezers, plasmonic nanopores, and wearable sensors have expanded SERS capabilities for single-cell and single-molecule analysis. Advances in spectral analysis, including signal digitalization, denoising, and deep learning algorithms, have improved the quantification of complex biological data. Finally, this review discusses SERS biomedical applications in nucleic acid detection, protein characterization, metabolite analysis, single-cell monitoring, and in vivo deep Raman spectroscopy, emphasizing its potential for liquid biopsy, metabolic phenotyping, and extracellular vesicle diagnostics. The review concludes with a perspective on clinical translation of SERS, addressing commercialization potentials and the challenges in deep tissue in vivo sensing and imaging.
Collapse
Affiliation(s)
- Linley Li Lin
- Sixth People's Hospital, School of Medicine & School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, P. R. China
| | - Ramon Alvarez-Puebla
- Departamento de Química Física e Inorganica, Universitat Rovira i Virgili, Tarragona 43007, Spain
- ICREA-Institució Catalana de Recerca i Estudis Avançats, Barcelona 08010, Spain
| | - Luis M Liz-Marzán
- CIC biomaGUNE, Basque Research and Technology Alliance (BRTA), Donostia-San Sebastián 20014, Spain
- Ikerbasque, Basque Foundation for Science, University of Santiago de nCompostela, Bilbao 48013, Spain
- Centro de Investigación Cooperativa en Red, Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Donostia-San Sebastián 20014, Spain
- Cinbio, University of Vigo, Vigo 36310, Spain
| | - Matt Trau
- Centre for Personalized Nanomedicine, Australian Institute for Bioengineering and Nanotechnology (AIBN), The University of Queensland, Brisbane, QLD 4072, Australia
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Jing Wang
- Key Laboratory of OptoElectronic Science and Technology for Medicine of Ministry of Education, Fujian Provincial Key Laboratory of Photonics Technology, Fujian Normal University, Fuzhou 350117, China
| | - Laura Fabris
- Department of Applied Science and Technology, Politecnico di Torino Corso Duca degli Abruzzi 24, 10129 Torino, Italy
| | - Xiang Wang
- State Key Laboratory of Physical Chemistry of Solid Surfaces, Collaborative Innovation Center of Chemistry for Energy Materials (iChEM), Innovation Laboratory for Sciences and Technologies of Energy Materials of Fujian Province (IKKEM), Department of Chemistry, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| | - Guokun Liu
- State Key Laboratory of Marine Environmental Science, Fujian Provincial Key Laboratory for Coastal Ecology and Environmental Studies, Center for Marine Environmental Chemistry and Toxicology, College of the Environment and Ecology, Xiamen University, Xiamen 361005, China
| | - Shuping Xu
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun 130012, PR China
| | - Xiao Xia Han
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun 130012, PR China
| | - Liangbao Yang
- Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui 230031, P. R. China
- Department of Pharmacy, Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, Anhui 230031, P. R. China
| | - Aiguo Shen
- School of Bioengineering and Health, Wuhan Textile University, Wuhan 430200, P. R. China
| | - Shikuan Yang
- School of Materials Science and Engineering, Zhejiang University, Hangzhou 310027, P. R. China
| | - Yikai Xu
- Key Laboratory for Advanced Materials and Feringa Nobel Prize Scientist Joint Research Center, Frontiers Science Center for Materiobiology and Dynamic Chemistry, School of Chemistry and Molecular Engineering, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, P. R. China
| | - Chunchun Li
- School of Materials Science and Engineering, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, P. R. China
| | - Jinqing Huang
- Department of Chemistry, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong 999077, China
| | - Shao-Chuang Liu
- Molecular Sensing and Imaging Center, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, P. R. China
| | - Jian-An Huang
- Research Unit of Health Sciences and Technology, Faculty of Medicine, University of Oulu, Aapistie 5 A, 90220 Oulu, Finland
- Research Unit of Disease Networks, Faculty of Biochemistry and Molecular Medicine, University of Oulu, Aapistie 5 A, 90220 Oulu, Finland
- Biocenter Oulu, University of Oulu, Aapistie 5 A, 90220 Oulu, Finland
| | - Indrajit Srivastava
- Department of Mechanical Engineering, Texas Tech University, Lubbock, Texas 79409, United States
- Texas Center for Comparative Cancer Research (TC3R), Amarillo, Texas 79106, United States
| | - Ming Li
- School of Materials Science and Engineering, Central South University, Changsha, Hunan 410083, China
| | - Limei Tian
- Department of Biomedical Engineering, and Center for Remote Health Technologies and Systems Texas A&M University, College Station, Texas 77843, United States
| | - Lam Bang Thanh Nguyen
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 21 Nanyang Link, Singapore 637371
| | - Xinyuan Bi
- Sixth People's Hospital, School of Medicine & School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, P. R. China
| | - Dana Cialla-May
- Leibniz Institute of Photonic Technology, Member of Leibniz Health Technologies, Member of the Leibniz Centre for Photonics in Infection Research (LPI), Albert-Einstein-Straße 9, 07745 Jena, Germany
- Institute of Physical Chemistry (IPC) and Abbe Center of Photonics (ACP), Friedrich Schiller University Jena, Member of the Leibniz Centre for Photonics in Infection Research (LPI), Helmholtzweg 4, 07743 Jena, Germany
| | - Pavel Matousek
- Central Laser Facility, Research Complex at Harwell, STFC Rutherford Appleton Laboratory, UKRI, Harwell Campus, Oxfordshire OX11 0QX, United Kingdom
- Department of Physics and Astronomy, University of Exeter, Exeter EX4 4QL, United Kingdom
| | - Nicholas Stone
- Department of Physics and Astronomy, University of Exeter, Exeter EX4 4QL, United Kingdom
| | - Randy P Carney
- Department of Biomedical Engineering, University of California, Davis, California 95616, United States
| | - Wei Ji
- College of Chemistry, Chemical Engineering and Resource Utilization, Northeast Forestry University, Harbin 145040, China
| | - Wei Song
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun 130012, PR China
| | - Zhou Chen
- Sixth People's Hospital, School of Medicine & School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, P. R. China
| | - In Yee Phang
- Key Laboratory of Synthetic and Biological Colloids, Ministry of Education, International Joint Research Laboratory for Nano Energy Composites, School of Chemical and Material Engineering, Jiangnan University, Wuxi 214122, P. R. China
| | - Malou Henriksen-Lacey
- CIC biomaGUNE, Basque Research and Technology Alliance (BRTA), Donostia-San Sebastián 20014, Spain
- Centro de Investigación Cooperativa en Red, Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Donostia-San Sebastián 20014, Spain
| | - Haoran Chen
- Sixth People's Hospital, School of Medicine & School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, P. R. China
| | - Zongyu Wu
- Sixth People's Hospital, School of Medicine & School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, P. R. China
| | - Heng Guo
- Department of Biomedical Engineering, and Center for Remote Health Technologies and Systems Texas A&M University, College Station, Texas 77843, United States
| | - Hao Ma
- State Key Laboratory of Physical Chemistry of Solid Surfaces, Collaborative Innovation Center of Chemistry for Energy Materials (iChEM), Innovation Laboratory for Sciences and Technologies of Energy Materials of Fujian Province (IKKEM), Department of Chemistry, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| | - Gennadii Ustinov
- Leibniz Institute of Photonic Technology, Member of Leibniz Health Technologies, Member of the Leibniz Centre for Photonics in Infection Research (LPI), Albert-Einstein-Straße 9, 07745 Jena, Germany
- Institute of Physical Chemistry (IPC) and Abbe Center of Photonics (ACP), Friedrich Schiller University Jena, Member of the Leibniz Centre for Photonics in Infection Research (LPI), Helmholtzweg 4, 07743 Jena, Germany
| | - Siheng Luo
- State Key Laboratory of Physical Chemistry of Solid Surfaces, Collaborative Innovation Center of Chemistry for Energy Materials (iChEM), Innovation Laboratory for Sciences and Technologies of Energy Materials of Fujian Province (IKKEM), Department of Chemistry, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| | - Sara Mosca
- Central Laser Facility, Research Complex at Harwell, STFC Rutherford Appleton Laboratory, UKRI, Harwell Campus, Oxfordshire OX11 0QX, United Kingdom
| | - Benjamin Gardner
- Department of Physics and Astronomy, University of Exeter, Exeter EX4 4QL, United Kingdom
| | - Yi-Tao Long
- Molecular Sensing and Imaging Center, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, P. R. China
| | - Juergen Popp
- Leibniz Institute of Photonic Technology, Member of Leibniz Health Technologies, Member of the Leibniz Centre for Photonics in Infection Research (LPI), Albert-Einstein-Straße 9, 07745 Jena, Germany
- Institute of Physical Chemistry (IPC) and Abbe Center of Photonics (ACP), Friedrich Schiller University Jena, Member of the Leibniz Centre for Photonics in Infection Research (LPI), Helmholtzweg 4, 07743 Jena, Germany
| | - Bin Ren
- State Key Laboratory of Physical Chemistry of Solid Surfaces, Collaborative Innovation Center of Chemistry for Energy Materials (iChEM), Innovation Laboratory for Sciences and Technologies of Energy Materials of Fujian Province (IKKEM), Department of Chemistry, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| | - Shuming Nie
- Department of Bioengineering, University of Illinois at Urbana-Champaign, 1406 W. Green Street, Urbana, Illinois 61801, United States
| | - Bing Zhao
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun 130012, PR China
| | - Xing Yi Ling
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 21 Nanyang Link, Singapore 637371
- Key Laboratory of Synthetic and Biological Colloids, Ministry of Education, International Joint Research Laboratory for Nano Energy Composites, School of Chemical and Material Engineering, Jiangnan University, Wuxi 214122, P. R. China
| | - Jian Ye
- Sixth People's Hospital, School of Medicine & School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, P. R. China
| |
Collapse
|
27
|
Yoon S, Chen B. Biomimetic Elastomer-Clay Nanocomposite Hydrogels with Control of Biological Chemicals for Soft Tissue Engineering and Wound Healing. ACS APPLIED BIO MATERIALS 2025; 8:2492-2505. [PMID: 39976353 PMCID: PMC11921026 DOI: 10.1021/acsabm.4c01944] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 02/10/2025] [Accepted: 02/11/2025] [Indexed: 02/21/2025]
Abstract
Resilient hydrogels are of great interest in soft tissue applications, such as soft tissue engineering and wound healing, with their biomimetic mechanical and hydration properties. A critical aspect in designing hydrogels for healthcare is their functionalities to control the surrounding biological environments to optimize the healing process. Herein, we have created an elastomer-clay nanocomposite hydrogel system with biomimetic mechanical behavior and sustained drug delivery of bioactive components and malodorous diamine-controlling properties. These hydrogels were prepared by a combined approach of melt intercalation of poly(ethylene glycol) and montmorillonite clay, followed by in situ cross-linking with a branched poly(glycerol sebacate) prepolymer. The hydration, vapor transmission, and surface wettability of the hydrogels were readily controlled by varying the clay content. Their mechanical properties were also modulated to mimic the Young's moduli (ranging between 12.6 and 105.2 kPa), as well as good flexibility and stretchability of soft tissues. A porous scaffold with interconnected pore structures as well as full and instant shape recovery was fabricated from a selected nanocomposite to demonstrate its potential applications as soft tissue scaffolds and wound healing materials. Biodegradability and biocompatibility were tested in vitro, showing controllable degradation kinetics with clay and no evidence of cytotoxicity. With the high surface area and absorption capacity of the clay, sustained drug delivery of a proangiogenic agent of 17β-estradiol as a model drug and the ability to control the malodorous diamines were both achieved. This elastomer-clay nanocomposite hydrogel system with a three-dimensional interconnected porous scaffold architecture and controllable hydration, mechanical, and biodegradable properties, as well as good biocompatibility and the ability to control the biological chemical species of the surrounding environments, has great potential in soft tissue engineering and wound healing.
Collapse
Affiliation(s)
- Sungkwon Yoon
- School of
Mechanical and Aerospace Engineering, Queen’s
University Belfast, Stranmillis Road, Belfast BT9 5AH, United Kingdom
- Department
of Materials Science and Engineering, University
of Sheffield, Mappin
Street, Sheffield S1 3JD, United Kingdom
| | - Biqiong Chen
- School of
Mechanical and Aerospace Engineering, Queen’s
University Belfast, Stranmillis Road, Belfast BT9 5AH, United Kingdom
| |
Collapse
|
28
|
Clark SJ, Curcio C, Dick AD, Doyle S, Edwards M, Flores-Bellver M, Hass D, Lennon R, Toomey CB, Rohrer B. Breaking Bruch's: How changes in Bruch's membrane influence retinal homeostasis. Exp Eye Res 2025; 255:110343. [PMID: 40107443 DOI: 10.1016/j.exer.2025.110343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 02/28/2025] [Accepted: 03/12/2025] [Indexed: 03/22/2025]
Affiliation(s)
- Simon J Clark
- Institute for Ophthalmic Research, Eberhard Karls University of Tübingen, Tübingen, Germany.
| | - Christine Curcio
- Department of Ophthalmology and Visual Sciences, University of Alabama at Birmingham Heersink School of Medicine, USA
| | - Andrew D Dick
- University of Bristol and UCL-Institute of Ophthalmology and NIHR Biomedical Research Centre, Moorfields Eye Hospital and UCL-Institute of Ophthalmology, UK
| | - Sarah Doyle
- Department of Clinical Medicine, School of Medicine and Trinity Institute of Neuroscience, Trinity College Dublin, Dublin, Ireland
| | - Malia Edwards
- Wilmer Eye Institute, Johns Hopkins University, Baltimore, MD, USA
| | - Miguel Flores-Bellver
- Department of Ophthalmology, Sue Anschutz-Rodgers Eye Center, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Daniel Hass
- Department of Biochemistry, University of Washington, Seattle, WA, USA
| | - Rachel Lennon
- Wellcome Centre for Cell-Matrix Research, School of Biological Science, Faculty of Biology, Medicine and Health, The University of Manchester, UK
| | - Christopher B Toomey
- Shiley Eye Institute, Viterbi Family Department of Ophthalmology, University of California at San Diego, La Jolla, CA, USA
| | - Bärbel Rohrer
- Department of Ophthalmology, Medical University of South Carolina, Charleston SC, USA.
| |
Collapse
|
29
|
Morabito M, Thibodot P, Gigandet A, Compagnon P, Toso C, Berishvili E, Lacotte S, Peloso A. Liver Extracellular Matrix in Colorectal Liver Metastasis. Cancers (Basel) 2025; 17:953. [PMID: 40149289 PMCID: PMC11939972 DOI: 10.3390/cancers17060953] [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: 01/30/2025] [Revised: 03/06/2025] [Accepted: 03/08/2025] [Indexed: 03/29/2025] Open
Abstract
The liver is the most common site of metastasis of colorectal cancer (CRC), and colorectal liver metastasis is one of the major causes of CRC-related deaths worldwide. The tumor microenvironment, particularly the extracellular matrix (ECM), plays a critical role in CRC metastasis and chemoresistance. Based on findings from clinical and basic research, this review attempts to offer a complete understanding of the role of the ECM in colorectal liver metastasis and to suggest potential ways for therapeutic intervention. First, the ECMs' role in regulating cancer cell fate is explored. We then discuss the hepatic ECM fingerprint and its influence on the metastatic behavior of CRC cells, highlighting key molecular interactions that promote metastasis. In addition, we examine how changes in the ECM within the metastatic niche contribute to chemoresistance, focusing on ECM remodeling by ECM stiffening and the activation of specific signaling pathways. Understanding these mechanisms is crucial for the development of novel strategies to overcome metastasis and improve outcomes for CRC patients.
Collapse
Affiliation(s)
- Marika Morabito
- General, Emergency and Transplant Surgery Department, ASST Settelaghi, University Hospital and Faculty of Medicine of Insubria, 21100 Varese, Italy
| | - Pauline Thibodot
- Hepato-Biliary Center, Paul-Brousse Hospital, Assistance Publique-Hôpitaux de Paris, 94800 Villejuif, France
| | - Anthony Gigandet
- School of Medecine, Faculty of Medecine, University of Geneva, 1211 Geneva, Switzerland
| | - Philippe Compagnon
- Division of Transplantation, Department of Surgery, Geneva University Hospitals and Faculty of Medicine, 1205 Geneva, Switzerland;
| | - Christian Toso
- Division of Abdominal Surgery and Transplantation, Department of Surgery, Geneva University Hospitals and Faculty of Medicine, 1205 Geneva, Switzerland
| | - Ekaterine Berishvili
- Cell Isolation and Transplantation Center, Department of Surgery, Geneva University Hospitals and University of Geneva, 1211 Geneva, Switzerland;
| | - Stéphanie Lacotte
- Hepatology and Transplantation Laboratory, Department of Surgery, Faculty of Medicine, University of Geneva, 1206 Geneva, Switzerland;
| | - Andrea Peloso
- Hepato-Biliary Center, Paul-Brousse Hospital, Assistance Publique-Hôpitaux de Paris, 94800 Villejuif, France
- Division of Transplantation, Department of Surgery, Geneva University Hospitals and Faculty of Medicine, 1205 Geneva, Switzerland;
- Division of Abdominal Surgery and Transplantation, Department of Surgery, Geneva University Hospitals and Faculty of Medicine, 1205 Geneva, Switzerland
| |
Collapse
|
30
|
Dey C, Sommerfeld IK, Bojarová P, Kodra N, Vrbata D, Zimolová Vlachová M, Křen V, Pich A, Elling L. Color-coded galectin fusion proteins as novel tools in biomaterial science. Biomater Sci 2025; 13:1482-1500. [PMID: 39907577 DOI: 10.1039/d4bm01148a] [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: 02/06/2025]
Abstract
The inherent carbohydrate-binding specificities of human galectins can serve as recognition elements in both biotechnological and biomedical applications. The combination of the carbohydrate-recognition domain (CRD) of galectins fused to peptides or proteins for purification, immobilization, and imaging enables multifunctional utilization within a single protein. We present here a library of color-coded galectin fusion proteins that incorporate a His6-tag, a fluorescent protein, and a SpyCatcher or SpyTag unit to enable immobilization procedures. These galectin fusion proteins exhibit similar binding properties to the non-fused galectins with micromolar apparent binding affinities. N- and C-terminal fusion partners do not interfere with the SpyCatcher/SpyTag immobilization. By applying SpyCatcher/SpyTag-mediated SC-ST-Gal-3 conjugates, we show the stepwise formation of a three-layer ECM-like structure in vitro. Additionally, we demonstrate the SpyCatcher/SpyTag-mediated immobilization of galectins in microgels, which can serve as a transport platform for localized targeting applications. The proof of concept is provided by the galectin-mediated binding of microgels to colorectal cancer cells.
Collapse
Affiliation(s)
- Carina Dey
- Laboratory for Biomaterials, Institute for Biotechnology and Helmholtz-Institute for Biomedical Engineering, RWTH Aachen University, Pauwelsstraße 20, 52074 Aachen, Germany.
| | - Isabel K Sommerfeld
- DWI - Leibniz-Institute for Interactive Materials, e.V. Forckenbeckstr. 50, 52074 Aachen, Germany
- Functional and Interactive Polymers, Institute of Technical and Macromolecular Chemistry, RWTH Aachen University, Worringerweg 2, 52074 Aachen, Germany
| | - Pavla Bojarová
- Laboratory of Biotransformation, Institute of Microbiology of the Czech Academy of Sciences, Vídeňská 1083, Prague 4, 14200, Czech Republic
- Department of Health Care Disciplines and Population Protection, Faculty of Biomedical Engineering, Czech Technical University in Prague, nám. Sítná 3105, 27201 Kladno, Czech Republic
| | - Nikol Kodra
- Laboratory for Biomaterials, Institute for Biotechnology and Helmholtz-Institute for Biomedical Engineering, RWTH Aachen University, Pauwelsstraße 20, 52074 Aachen, Germany.
| | - David Vrbata
- Laboratory of Biotransformation, Institute of Microbiology of the Czech Academy of Sciences, Vídeňská 1083, Prague 4, 14200, Czech Republic
| | - Miluše Zimolová Vlachová
- Laboratory of Biotransformation, Institute of Microbiology of the Czech Academy of Sciences, Vídeňská 1083, Prague 4, 14200, Czech Republic
| | - Vladimír Křen
- Laboratory of Biotransformation, Institute of Microbiology of the Czech Academy of Sciences, Vídeňská 1083, Prague 4, 14200, Czech Republic
| | - Andrij Pich
- DWI - Leibniz-Institute for Interactive Materials, e.V. Forckenbeckstr. 50, 52074 Aachen, Germany
- Functional and Interactive Polymers, Institute of Technical and Macromolecular Chemistry, RWTH Aachen University, Worringerweg 2, 52074 Aachen, Germany
- Aachen Maastricht Institute for Biobased Materials (AMIBM), Maastricht University, Brightlands Chemelot Campus, Urmonderbaan 22, 6167 RD Geleen, The Netherlands
| | - Lothar Elling
- Laboratory for Biomaterials, Institute for Biotechnology and Helmholtz-Institute for Biomedical Engineering, RWTH Aachen University, Pauwelsstraße 20, 52074 Aachen, Germany.
| |
Collapse
|
31
|
Niazi A, Kim JA, Kim DK, Lu D, Sterin I, Park J, Park S. Microvilli control the morphogenesis of the tectorial membrane extracellular matrix. Dev Cell 2025; 60:679-695.e8. [PMID: 39657673 PMCID: PMC11905117 DOI: 10.1016/j.devcel.2024.11.011] [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: 01/04/2024] [Revised: 09/17/2024] [Accepted: 11/13/2024] [Indexed: 12/12/2024]
Abstract
The apical extracellular matrix (aECM), organized by polarized epithelial cells, exhibits complex structures. The tectorial membrane (TM), an aECM in the cochlea mediating auditory transduction, exhibits highly ordered domain-specific architecture. α-Tectorin (TECTA), a glycosylphosphatidylinositol (GPI)-anchored ECM protein, is essential for TM organization. Here, we identified that α-tectorin is released by distinct modes: proteolytic shedding by TMPRSS2 and GPI-anchor-dependent release from the microvillus tip in mice. In the medial/limbal domain, proteolytically shed α-tectorin forms dense fibers. In contrast, in the lateral/body domain, where supporting cells exhibit dense microvilli, shedding restricts α-tectorin to the microvillus tip, compartmentalizing collagen-binding sites. Tip-localized α-tectorin is released in a GPI-anchor-dependent manner to form collagen-crosslinking fibers, maintaining the spacing and parallel organization of collagen fibrils. Overall, these distinct release modes of α-tectorin determine domain-specific organization, with the microvillus coordinating release modes along its membrane to assemble the higher-order ECM architecture.
Collapse
Affiliation(s)
- Ava Niazi
- Department of Neurobiology, University of Utah, Salt Lake City, UT, USA; Neuroscience Program, University of Utah, Salt Lake City, UT, USA
| | - Ju Ang Kim
- Department of Neurobiology, University of Utah, Salt Lake City, UT, USA
| | - Dong-Kyu Kim
- Department of Neurobiology, University of Utah, Salt Lake City, UT, USA
| | - Di Lu
- Department of Neurobiology, University of Utah, Salt Lake City, UT, USA
| | - Igal Sterin
- Department of Neurobiology, University of Utah, Salt Lake City, UT, USA
| | - Joosang Park
- Department of Neurobiology, University of Utah, Salt Lake City, UT, USA
| | - Sungjin Park
- Department of Neurobiology, University of Utah, Salt Lake City, UT, USA.
| |
Collapse
|
32
|
Kim D, Lee JW, Kim YT, Choe J, Kim G, Ha CM, Kim JG, Song KH, Yang S. Minimally Invasive Syringe-Injectable Hydrogel with Angiogenic Factors for Ischemic Stroke Treatment. Adv Healthc Mater 2025; 14:e2403119. [PMID: 39520382 PMCID: PMC11874675 DOI: 10.1002/adhm.202403119] [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: 09/13/2024] [Revised: 10/06/2024] [Indexed: 11/16/2024]
Abstract
Ischemic stroke (IS) accounts for most stroke incidents and causes intractable damage to brain tissue. This condition manifests as diverse aftereffects, such as motor impairment, emotional disturbances, and dementia. However, a fundamental approach to curing IS remains unclear. This study proposes a novel approach for treating IS by employing minimally invasive and injectable jammed gelatin-norbornene nanofibrous hydrogels (GNF) infused with growth factors (GFs). The developed GNF/GF hydrogels are administered to the motor cortex of a rat IS model to evaluate their therapeutic effects on IS-induced motor dysfunction. GNFs mimic a natural fibrous extracellular matrix architecture and can be precisely injected into a targeted brain area. The syringe-injectable jammed nanofibrous hydrogel system increased angiogenesis, inflammation, and sensorimotor function in the IS-affected brain. For clinical applications, the biocompatible GNF hydrogel has the potential to efficiently load disease-specific drugs, enabling targeted therapy for treating a wide range of neurological diseases.
Collapse
Affiliation(s)
- Donggue Kim
- Department of Nano‐BioengineeringIncheon National UniversityIncheon22012Republic of Korea
| | - Ji Woo Lee
- Department of Nano‐BioengineeringIncheon National UniversityIncheon22012Republic of Korea
| | - Yang Tae Kim
- Division of Life SciencesCollege of Life Sciences and BioengineeringIncheon National UniversityIncheon22012Republic of Korea
| | - Junhyeok Choe
- Department of Nano‐BioengineeringIncheon National UniversityIncheon22012Republic of Korea
| | - Gaeun Kim
- Department of Nano‐BioengineeringIncheon National UniversityIncheon22012Republic of Korea
| | - Chang Man Ha
- Research Division and Brain Research Core Facilities of Korea Brain Research InstituteDaegu41068Republic of Korea
| | - Jae Geun Kim
- Division of Life SciencesCollege of Life Sciences and BioengineeringIncheon National UniversityIncheon22012Republic of Korea
- Research Center of Brain‐Machine InterfaceIncheon National UniversityIncheon22012Republic of Korea
| | - Kwang Hoon Song
- Department of Nano‐BioengineeringIncheon National UniversityIncheon22012Republic of Korea
- Research Center of Brain‐Machine InterfaceIncheon National UniversityIncheon22012Republic of Korea
| | - Sunggu Yang
- Department of Nano‐BioengineeringIncheon National UniversityIncheon22012Republic of Korea
- Research Center of Brain‐Machine InterfaceIncheon National UniversityIncheon22012Republic of Korea
- gBrain Inc.Incheon21984Republic of Korea
| |
Collapse
|
33
|
Golebiowska AA, Intravaia JT, Sathe V, Kumbar SG, Nukavarapu SP. Engineered Osteochondral Scaffolds with Bioactive Cartilage Zone for Enhanced Articular Cartilage Regeneration. Ann Biomed Eng 2025; 53:597-611. [PMID: 39602036 PMCID: PMC11835937 DOI: 10.1007/s10439-024-03655-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Accepted: 11/18/2024] [Indexed: 11/29/2024]
Abstract
Despite progress, osteochondral (OC) tissue engineering strategies face limitations in terms of articular cartilage layer development and its integration with the underlying bone tissue. The main objective of this study is to develop a zonal OC scaffold with native biochemical signaling in the cartilage zone to promote articular cartilage development devoid of cells and growth factors. Herein, we report the development and in vivo assessment of a novel gradient and zonal-structured scaffold for OC defect regeneration. The scaffold system is composed of a mechanically supportive 3D-printed template containing decellularized cartilage extracellular matrix (ECM) biomaterial in the cartilage zone that possesses bioactive characteristics, such as chemotactic activity and native tissue biochemical composition. OC scaffolds with a bioactive cartilage zone were implanted in vivo in a rabbit osteochondral defect model and assessed for gross morphology, matrix deposition, cellular distribution, and overall tissue regeneration. The scaffold system supported recruitment and infiltration of host cells into the cartilage zone of the graft, which led to increased ECM deposition and physiologically relevant articular cartilage tissue formation. Semi-quantitative ICRS scoring (overall score double for OC scaffold with bioactive cartilage zone compared to PLA scaffold) further confirm the bioactive scaffold enhanced articular cartilage engineering. This strategy of designing bioactive scaffolds to promote endogenous cellular infiltration can be a much simpler and effective approach for OC tissue repair and regeneration.
Collapse
Affiliation(s)
- Aleksandra A Golebiowska
- Department of Biomedical Engineering, University of Connecticut, 260 Glenbrook Road, Unit 3247, Storrs, CT, 06269, USA
| | - Jonathon T Intravaia
- Department of Biomedical Engineering, University of Connecticut, 260 Glenbrook Road, Unit 3247, Storrs, CT, 06269, USA
| | - Vinayak Sathe
- Department of Orthopaedic Surgery, University of Connecticut Health, Farmington, CT, 06032, USA
| | - Sangamesh G Kumbar
- Department of Biomedical Engineering, University of Connecticut, 260 Glenbrook Road, Unit 3247, Storrs, CT, 06269, USA
- Department of Materials Science & Engineering, University of Connecticut, Storrs, CT, 06269, USA
- Department of Orthopaedic Surgery, University of Connecticut Health, Farmington, CT, 06032, USA
| | - Syam P Nukavarapu
- Department of Biomedical Engineering, University of Connecticut, 260 Glenbrook Road, Unit 3247, Storrs, CT, 06269, USA.
- Department of Materials Science & Engineering, University of Connecticut, Storrs, CT, 06269, USA.
- Department of Orthopaedic Surgery, University of Connecticut Health, Farmington, CT, 06032, USA.
| |
Collapse
|
34
|
Ding X, Xie S, Zhang W, Zhu Y, Xu D, Xian S, Sun H, Guo X, Li Y, Lu J, Tong X, Huang R, Ji S, Xia Z. Current application of tissue-engineered dermal scaffolds mimicking the extracellular matrix microenvironment in wound healing. Regen Ther 2025; 28:371-382. [PMID: 39896445 PMCID: PMC11786805 DOI: 10.1016/j.reth.2024.12.018] [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: 10/20/2024] [Revised: 12/09/2024] [Accepted: 12/26/2024] [Indexed: 02/04/2025] Open
Abstract
With the continuous advancement of materials science, cell biology, and biotechnology, tissue engineering has introduced novel solutions to traditional wound healing approaches, particularly demonstrating significant potential in addressing complex or non-healing wounds. One of the key technologies in this field, dermal scaffolds, serve as wound coverage materials that mimic the structural framework of the dermis. They primarily assume the function of extracellular matrix, providing space for cell attachment, migration, and proliferation, thus supporting cellular growth and regulating multiple biological processes in healing. Tissue engineering utilizes combinations of natural or synthetic scaffolds, seeded cells, or growth factors to induce distinct effects in angiogenesis, extracellular matrix deposition, and functional recovery. Therefore, various bioengineered dermal scaffolds hold significant potential for clinical translation in wound healing. This review outlines various extracellular matrix molecules utilized in the development of dermal scaffolds, emphasizes recent progress in cell- and growth factor-modified scaffolds, and discusses the challenges and future perspectives in this evolving field.
Collapse
Affiliation(s)
| | | | | | - Yushu Zhu
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, People's Republic of China, Research Unit of Key Techniques for Treatment of Burns and Combined Burns and Trauma Injury, Chinese Academy of Medical Sciences, No. 168 Changhai Road, Shanghai 200433, China
| | - Dayuan Xu
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, People's Republic of China, Research Unit of Key Techniques for Treatment of Burns and Combined Burns and Trauma Injury, Chinese Academy of Medical Sciences, No. 168 Changhai Road, Shanghai 200433, China
| | - Shuyuan Xian
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, People's Republic of China, Research Unit of Key Techniques for Treatment of Burns and Combined Burns and Trauma Injury, Chinese Academy of Medical Sciences, No. 168 Changhai Road, Shanghai 200433, China
| | - Hanlin Sun
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, People's Republic of China, Research Unit of Key Techniques for Treatment of Burns and Combined Burns and Trauma Injury, Chinese Academy of Medical Sciences, No. 168 Changhai Road, Shanghai 200433, China
| | - Xinya Guo
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, People's Republic of China, Research Unit of Key Techniques for Treatment of Burns and Combined Burns and Trauma Injury, Chinese Academy of Medical Sciences, No. 168 Changhai Road, Shanghai 200433, China
| | - Yixu Li
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, People's Republic of China, Research Unit of Key Techniques for Treatment of Burns and Combined Burns and Trauma Injury, Chinese Academy of Medical Sciences, No. 168 Changhai Road, Shanghai 200433, China
| | - Jianyu Lu
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, People's Republic of China, Research Unit of Key Techniques for Treatment of Burns and Combined Burns and Trauma Injury, Chinese Academy of Medical Sciences, No. 168 Changhai Road, Shanghai 200433, China
| | - Xirui Tong
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, People's Republic of China, Research Unit of Key Techniques for Treatment of Burns and Combined Burns and Trauma Injury, Chinese Academy of Medical Sciences, No. 168 Changhai Road, Shanghai 200433, China
| | - Runzhi Huang
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, People's Republic of China, Research Unit of Key Techniques for Treatment of Burns and Combined Burns and Trauma Injury, Chinese Academy of Medical Sciences, No. 168 Changhai Road, Shanghai 200433, China
| | - Shizhao Ji
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, People's Republic of China, Research Unit of Key Techniques for Treatment of Burns and Combined Burns and Trauma Injury, Chinese Academy of Medical Sciences, No. 168 Changhai Road, Shanghai 200433, China
| | - Zhaofan Xia
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, People's Republic of China, Research Unit of Key Techniques for Treatment of Burns and Combined Burns and Trauma Injury, Chinese Academy of Medical Sciences, No. 168 Changhai Road, Shanghai 200433, China
| |
Collapse
|
35
|
Scott S, Villiou M, Colombo F, la Cruz‐García AD, Tydecks L, Toelke L, Siemsen K, Selhuber‐Unkel C. Dynamic and Reversible Tuning of Hydrogel Viscoelasticity by Transient Polymer Interactions for Controlling Cell Adhesion. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2408616. [PMID: 39935068 PMCID: PMC11938001 DOI: 10.1002/adma.202408616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 10/11/2024] [Indexed: 02/13/2025]
Abstract
Cells are highly responsive to changes in their mechanical environment, influencing processes such as stem cell differentiation and tumor progression. To meet the growing demand for materials used for high throughput mechanotransduction studies, simple means of dynamically adjusting the environmental viscoelasticity of cell cultures are needed. Here, a novel method is presented to dynamically and reversibly control the viscoelasticity of naturally derived polymer hydrogels through interactions with poly (ethylene glycol) (PEG). Interactions between PEG and hydrogel polymers, possibly involving hydrogen bonding, stiffen the hydrogel matrices. By dynamically changing the PEG concentration of the solution in which polymer hydrogels are incubated, their viscoelastic properties are adjusted, which in turn affects cell adhesion and cytoskeletal organization. Importantly, this effects is reversible, providing a cost-effective and simple strategy for dynamically adjusting the viscoelasticity of polymer hydrogels. This method holds promise for applications in mechanobiology, biomedicine, and the life sciences.
Collapse
Affiliation(s)
- Shane Scott
- Department of Materials Science and EngineeringMcMaster University1280 Main St. W.HamiltonOntarioL8S 4L8Canada
| | - Maria Villiou
- Institute for Molecular Systems Engineering and Advanced Materials (IMSEAM)Heidelberg UniversityD‐69120HeidelbergGermany
- Max Planck School Matter to LifeHeidelberg UniversityJahnstraße 2969120HeidelbergGermany
- Max Planck Institute for Polymer ResearchAckermannweg 10A55128MainzGermany
| | - Federico Colombo
- Institute for Molecular Systems Engineering and Advanced Materials (IMSEAM)Heidelberg UniversityD‐69120HeidelbergGermany
| | - Angeles De la Cruz‐García
- Institute for Molecular Systems Engineering and Advanced Materials (IMSEAM)Heidelberg UniversityD‐69120HeidelbergGermany
| | - Leon Tydecks
- Institute for Molecular Systems Engineering and Advanced Materials (IMSEAM)Heidelberg UniversityD‐69120HeidelbergGermany
| | - Lotta Toelke
- Institute for Molecular Systems Engineering and Advanced Materials (IMSEAM)Heidelberg UniversityD‐69120HeidelbergGermany
| | - Katharina Siemsen
- Institute for Materials ScienceKiel UniversityKaiserstraße 224143KielGermany
| | - Christine Selhuber‐Unkel
- Institute for Molecular Systems Engineering and Advanced Materials (IMSEAM)Heidelberg UniversityD‐69120HeidelbergGermany
- Max Planck School Matter to LifeHeidelberg UniversityJahnstraße 2969120HeidelbergGermany
| |
Collapse
|
36
|
Xu T, Rao J, Mo Y, Lam ACH, Yang Y, Wong SWF, Wong KH, Zhao X. 3D printing in musculoskeletal interface engineering: Current progress and future directions. Adv Drug Deliv Rev 2025; 219:115552. [PMID: 40032068 DOI: 10.1016/j.addr.2025.115552] [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: 12/06/2024] [Revised: 02/17/2025] [Accepted: 02/28/2025] [Indexed: 03/05/2025]
Abstract
The musculoskeletal system relies on critical tissue interfaces for its function; however, these interfaces are often compromised by injuries and diseases. Restoration of these interfaces is complex by nature which renders traditional treatments inadequate. An emerging solution is three-dimensional printing, which allows for precise fabrication of biomimetic scaffolds to enhance tissue regeneration. This review summarizes the utility of 3D printing in creating scaffolds for musculoskeletal interfaces, mainly focusing on advanced techniques such as multi-material printing, bioprinting, and 4D printing. We emphasize the significance of mimicking natural tissue gradients and the selection of appropriate biomaterials to ensure scaffold success. The review outlines state-of-the-art 3D printing technologies, varying from extrusion, inkjet and laser-assisted bioprinting, which are crucial for producing scaffolds with tailored mechanical and biological properties. Applications in cartilage-bone, intervertebral disc, tendon/ligament-bone, and muscle-tendon junction engineering are discussed, highlighting the potential for improved integration and functionality. Furthermore, we address challenges in material development, printing resolution, and the in vivo performance of scaffolds, as well as the prospects for clinical translation. The review concludes by underscoring the transformative potential of 3D printing to advance orthopedic medicine, offering a roadmap for future research at the intersection of biomaterials, drug delivery, and tissue engineering.
Collapse
Affiliation(s)
- Tianpeng Xu
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong Special Administrative Region
| | - Jingdong Rao
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong Special Administrative Region
| | - Yongyi Mo
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong Special Administrative Region
| | - Avery Chik-Him Lam
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong Special Administrative Region
| | - Yuhe Yang
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong Special Administrative Region; The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen, China
| | - Sidney Wing-Fai Wong
- Industrial Centre, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong Special Administrative Region
| | - Ka-Hing Wong
- Research Institute for Future Food, Department of Food Science and Nutrition, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong Special Administrative Region
| | - Xin Zhao
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong Special Administrative Region; The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen, China; Research Institute for Intelligent Wearable Systems, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong Special Administrative Region.
| |
Collapse
|
37
|
Beltrán-Hernández NE, Cardenas L, Jimenez-Jacinto V, Vega-Alvarado L, Rivera HM. Biological Activity of Biomarkers Associated With Metastasis in Osteosarcoma Cell Lines. Cancer Med 2025; 14:e70391. [PMID: 40079158 PMCID: PMC11904427 DOI: 10.1002/cam4.70391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Revised: 08/26/2024] [Accepted: 10/20/2024] [Indexed: 03/14/2025] Open
Abstract
INTRODUCTION Osteosarcoma, a highly aggressive bone cancer primarily affecting children and young adults, remains a significant challenge in clinical oncology. Metastasis stands as the primary cause of mortality in osteosarcoma patients. However, the mechanisms driving this process remain incompletely understood. Clarifying the molecular pathways involved in metastasis is essential for enhancing patient prognoses and facilitating the development of targeted therapeutic strategies. METHODS RNA sequencing (RNA-Seq) analysis was employed to compare three conditions, hFOB1.19 versus Saos-2, hFOB1.19 versus SJSA-1, and Saos-2 versus SJSA-1, involving non-cancer osteoblasts (hFOB1.19) and highly metastatic osteosarcoma cell lines (Saos-2 and SJSA-1). Additionally, ENA datasets of RNA-Seq from osteosarcoma biopsies were included. Differentially expressed genes (DEGs) were identified and analyzed through enrichment pathway analysis and protein-protein interaction (PPI) networks. Additionally, for gene candidates, a biochemical evaluation was performed. RESULTS DEGs associated with biological functions pertinent to migration, invasion, and metastasis in osteosarcoma were identified. Notably, matrix metalloproteinase-2 (MMP-2) emerged as a promising candidate. Both canonical or full-length (FL-mmp-2) and N-terminal truncated (NTT-mmp-2) isoforms were discerned in biopsies. Moreover, MMP-2's activity was characterized in cell lines. Additionally, mRNA expression of voltage-gated sodium channels (NaVs) and voltage-gated potassium channels (KVs) was detected, and their functional expression was validated using patch clamp techniques. Evaluation of cell line migration and invasion capacities revealed their reduction in the presence of ion channel blockers (TTX and TEA) and MMP inhibitor (GM6001). CONCLUSIONS The gene functional enrichment analysis of DEGs enabled the identification of interaction networks in osteosarcoma, thereby revealing potential biomarkers. Moreover, the elucidated co-participation of TTX-sensitive NaVs and MMP-2 in facilitating migration and invasion suggests their suitability as novel prognostic biomarkers for osteosarcoma. Additionally, this study introduces a model delineating the potential interaction mechanism among ion channels, MMP-2, and other crucial factors in the metastatic cascade of osteosarcoma.
Collapse
Affiliation(s)
| | - Luis Cardenas
- Departamento de Biología Molecular de Plantas, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Morelos, Mexico
| | - Verónica Jimenez-Jacinto
- Unidad Universitaria de Secuenciación Masiva y Bioinformática, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Morelos, Mexico
| | - Leticia Vega-Alvarado
- Instituto de Ciencias Aplicadas y Tecnología, Universidad Nacional Autónoma de México, Coyoacán Ciudad de México, Mexico
| | - Heriberto Manuel Rivera
- Universidad Autónoma del Estado de Morelos, Facultad de Medicina, Cuernavaca, Morelos, Mexico
| |
Collapse
|
38
|
Sehic E, de Miguel-Gómez L. Standardizing decellularization protocols for optimized uterine tissue bioengineering. Regen Ther 2025; 28:183-190. [PMID: 39811067 PMCID: PMC11731971 DOI: 10.1016/j.reth.2024.12.011] [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: 09/29/2024] [Revised: 11/14/2024] [Accepted: 12/12/2024] [Indexed: 01/16/2025] Open
Abstract
Bioengineering is applied in different areas, including women's infertility management. Among other approaches, decellularized tissues are being used to treat uterine disorders causing infertility. Biomaterials made from decellularized tissue consist of tissue-specific extracellular matrix and, as acellular scaffolds, are thought to be immune inert. Hence, they are good grafting candidates to replace and regenerate excised damaged uterine tissue to cure infertility. However, decellularization approaches differ among species and research groups, posing challenges for comparison and standardization. The diversity in data reporting and studied properties of the resulting decellularized scaffold make it even more difficult, especially when the ultimate goal is clinical translation. Thus, this review aims to critically assess whole uterus decellularization studies, extracting and comparing their main results and conclusions. After carefully evaluating the reviewed studies, we noticed that the vast majority base the uterus decellularization success and resulting scaffold efficacy on the DNA removal efficacy, while other crucial aspects, including the extracellular matrix integrity or immunogenicity, are underestimated. Thus, this review further proposes practical points for what should be considered and how results can be reported in studies involving whole uterus decellularization to facilitate comparison between studies and translational progress.
Collapse
Affiliation(s)
- Edina Sehic
- Department of Medical Cell Biology, Uppsala University, 75123 Uppsala, Sweden
| | - Lucía de Miguel-Gómez
- Laboratory for Transplantation and Regenerative Medicine, Sahlgrenska Academy, University of Gothenburg, Kvinnokliniken, Blå stråket 6, 40530 Gothenburg, Sweden
- Department of Obstetrics and Gynecology, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, SE-405 30, Sweden
| |
Collapse
|
39
|
Jang HJ, Park JW. Microenvironmental Drivers of Glioma Progression. Int J Mol Sci 2025; 26:2108. [PMID: 40076738 PMCID: PMC11900340 DOI: 10.3390/ijms26052108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2025] [Revised: 02/22/2025] [Accepted: 02/25/2025] [Indexed: 03/14/2025] Open
Abstract
Gliomas, particularly glioblastoma (GBM), are among the most challenging brain tumors due to their complex and dynamic tumor microenvironment (TME). The TME plays a pivotal role in tumor progression, immune evasion, and resistance to therapy through intricate interactions among glioma cells, immune components, neurons, astrocytes, the extracellular matrix, and the blood-brain barrier. Targeting the TME has demonstrated potential, with immunotherapies such as checkpoint inhibitors and neoadjuvant therapies enhancing immune responses. Nonetheless, overcoming the immunosuppressive landscape and metabolic adaptations continues to pose significant challenges. This review explores the diverse cellular and molecular mechanisms that shape the glioma TME. A deeper understanding of these mechanisms holds promise for providing novel therapeutic opportunities to improve glioma treatment outcomes.
Collapse
Affiliation(s)
- Hyun Ji Jang
- Department of Life Sciences, College of BioNano Technology, Gachon University, Seongnam 13120, Republic of Korea
| | - Jong-Whi Park
- Department of Life Sciences, College of BioNano Technology, Gachon University, Seongnam 13120, Republic of Korea
- Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon 21999, Republic of Korea
| |
Collapse
|
40
|
Ren S, Liu Y, Guo Y, Zhao Z, Cui J, Li M, Wang J. TGF-β1 Mediates Novel-m0297-5p Targeting WNT5A to Participate in the Proliferation of Ovarian Granulosa Cells in Small-Tailed Han Sheep. Int J Mol Sci 2025; 26:1961. [PMID: 40076587 PMCID: PMC11901034 DOI: 10.3390/ijms26051961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Revised: 02/17/2025] [Accepted: 02/19/2025] [Indexed: 03/14/2025] Open
Abstract
MiRNAs regulate follicle development and atresia, steroid production, granulosa cell (GC) proliferation, and apoptosis. However, the target genes and the functioning of novel miRNAs remain unexplored. We reveal the targeting relationship between novel-m0297-5p and WNT5A and the specific regulatory mechanism of GC proliferation in small-tailed Han sheep using whole transcriptomic sequencing. We performed whole transcriptomic sequencing on small-tailed Han sheep ovarian GCs supplemented with 10 ng/mL of transforming growth factor-β1 (TGF-β1) during the early stages. This led to identifying the differential expression of novel-m0297-5p and Wnt family member 5A (WNT5A) and predicting their targeting relationship. Based on this, we hypothesized that TGF-β1 could mediate novel-m0297-5p targeting WNT5A to participate in the proliferation process of GCs in small-tailed sheep. We confirmed the relationship between TGF-β1 and both novel-m0297-5p and WNT5A. The mimicry of novel-m0297-5p inhibited GC activity and proliferation. However, the inhibition of novel-m0297-5p yielded the opposite effect. We validated the binding site for novel m0297-5p within the 3'UTR of WNT5A using dual-luciferase reporter gene. TGF-β1 alleviated the impact induced by the mimicry of novel-m0297-5p on cell viability. Inhibitor co-transfection for both novel-m0297-5p and si-WNT5A suppressed the granulocyte proliferation induced by novel-m0297-5p inhibition. These findings suggest that TGF-β1 can mediate the inhibitory effect of novel-m0297-5p targeting WNT5A on GC proliferation and activity in small-tailed Han sheep. This study provides an experimental basis for research on the biological function of GCs and their impact on follicle development.
Collapse
Affiliation(s)
| | | | | | | | | | - Mingna Li
- Key Laboratory of Herbivorous Animal Biotechnology, Faculty of Animal Science and Technology, Gansu Agricultural University, Lanzhou 730070, China; (S.R.); (Y.L.); (Y.G.); (Z.Z.); (J.C.)
| | - Jiqing Wang
- Key Laboratory of Herbivorous Animal Biotechnology, Faculty of Animal Science and Technology, Gansu Agricultural University, Lanzhou 730070, China; (S.R.); (Y.L.); (Y.G.); (Z.Z.); (J.C.)
| |
Collapse
|
41
|
Chiu CC, Hao WR, Lin KJ, Chen CC, Yang TY, Fang YA, Yang TL, Lai YH, Chen MY, Hsu MH, Lin CH, Hsiu H, Chen HY, Cheng TH, Chen NH, Liu JC. Big data analysis of influenza vaccination and liver cancer risk in hypertensive patients: insights from a nationwide population-based cohort study. BMC Gastroenterol 2025; 25:109. [PMID: 39994561 PMCID: PMC11849173 DOI: 10.1186/s12876-025-03665-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2024] [Accepted: 02/01/2025] [Indexed: 02/26/2025] Open
Abstract
BACKGROUND previous studies have indicated that influenza vaccination may be associated with reduced risks of certain types of cancer. However, the protective effect of influenza vaccination against primary liver cancer in individuals with hypertension remains unclear. METHODS In this cohort study, 37,022 patients over 55 years of age who received a diagnosis of hypertension at any time between January 1, 2001, and December 31, 2012, were enrolled from the National Health Insurance Research Database. The patients were divided into a vaccinated and an unvaccinated group. Categorical and continuous variables were analyzed using the chi-square test and t test, respectively, and the correlation between influenza vaccination and liver cancer in patients with hypertension was analyzed using time-varying COX model. Propensity score method was performed to reduce selection bias. RESULTS Compared with the unvaccinated group, the vaccinated group had a significantly lower incidence of liver cancer (hazard ratio = 0.56, 95% confidence interval = 0.46-0.64; p < .001). In addition, a protective effect was observed regardless of sex, age, or comorbidities. Besides, the association was dose-dependent which could be noted when patients were stratified based on the total number of vaccinations. The adjusted HRs for patients receiving 1, 2 to 3, and ≥ 4 vaccinations during the follow-up period were 0.60 (0.51-0.78), 0.48 (0.38-0.65), and 0.39(0.30-0.51), respectively. CONCLUSIONS In summary, influenza vaccination is linked to a decreased risk of liver cancer in individuals with hypertension. However, unmeasurable confounders may have been present in the analysis.
Collapse
Affiliation(s)
- Chun-Chih Chiu
- Division of Cardiology, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, 23561, Taiwan
- Taipei Heart Institute, Taipei Medical University, Taipei, Taiwan
| | - Wen-Rui Hao
- Division of Cardiology, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, 23561, Taiwan
- Taipei Heart Institute, Taipei Medical University, Taipei, Taiwan
- Division of Cardiology, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, 110, Taiwan
| | - Kuan-Jie Lin
- Taipei Heart Institute, Taipei Medical University, Taipei, Taiwan
- Division of Cardiovascular Surgery, Department of Surgery, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
| | - Chun-Chao Chen
- Division of Cardiology, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, 23561, Taiwan
- Taipei Heart Institute, Taipei Medical University, Taipei, Taiwan
- Division of Cardiology, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, 110, Taiwan
| | - Tsung-Yeh Yang
- Division of Cardiology, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, 23561, Taiwan
- Taipei Heart Institute, Taipei Medical University, Taipei, Taiwan
| | - Yu-Ann Fang
- Division of Cardiology, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, 23561, Taiwan
- Taipei Heart Institute, Taipei Medical University, Taipei, Taiwan
| | - Tsung-Lin Yang
- Taipei Heart Institute, Taipei Medical University, Taipei, Taiwan
- Division of Cardiology, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Division of Cardiology, Department of Internal Medicine, Cardiovascular Research Center, Taipei Medical University Hospital, Taipei, 110, Taiwan
| | - Yu-Hsin Lai
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- TMU Research Center for Digestive Medicine, Taipei Medical University, Taipei, 110, Taiwan
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Shuang Ho Hospital, New Taipei, Taiwan
| | - Ming-Yao Chen
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- TMU Research Center for Digestive Medicine, Taipei Medical University, Taipei, 110, Taiwan
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Shuang Ho Hospital, New Taipei, Taiwan
| | - Min-Huei Hsu
- Graduate Institute of Data Science, College of Management, Taipei Medical University, Taipei, Taiwan
- Department of Neurosurgery, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
| | - Cheng-Hsin Lin
- Taipei Heart Institute, Taipei Medical University, Taipei, Taiwan
- Division of Cardiovascular Surgery, Department of Surgery, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
| | - Hsin Hsiu
- Graduate Institute of Biomedical Engineering, National Taiwan University of Science and Technology, No.43, Section 4, Keelung Road, Taipei, 10607, Taiwan
| | - Huan-Yuan Chen
- Institute of Biomedical Sciences, Academia Sinica, Taipei, 11578, Taiwan
| | - Tzu-Hurng Cheng
- Department of Biochemistry, School of Medicine, College of Medicine, China Medical University, Taichung City, 404333, Taiwan
| | - Nai-Hsuan Chen
- Department of Physical medicine and rehabilitation, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan.
| | - Ju-Chi Liu
- Division of Cardiology, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, 23561, Taiwan.
- Taipei Heart Institute, Taipei Medical University, Taipei, Taiwan.
- Division of Cardiology, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.
| |
Collapse
|
42
|
Baster Z, Russell L, Rajfur Z. A Review of Talin- and Integrin-Dependent Molecular Mechanisms in Cancer Invasion and Metastasis. Int J Mol Sci 2025; 26:1798. [PMID: 40076426 PMCID: PMC11899650 DOI: 10.3390/ijms26051798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 02/04/2025] [Accepted: 02/06/2025] [Indexed: 03/14/2025] Open
Abstract
Cancer is the second most common cause of death in the world, representing one of the main economic burdens in health care and research. The effort of research has mainly focused on limiting the growth of a localized tumor, but most recently, there has been more attention focused on restricting the spreading of the cancer via invasion and metastasis. The signaling pathways behind these two processes share many molecules with physiological pathways regulating cell adhesion and migration, and, moreover, adhesion and migration processes themselves underlie tumor potential for invasion. In this work, we reviewed the latest literature about cancer development and invasion and their regulation by cell migration- and adhesion-related proteins, with a specific focus on talins and integrins. We also summarized the most recent developments and approaches to anti-cancer therapies, concentrating on cell migration-related therapies.
Collapse
Affiliation(s)
- Zbigniew Baster
- Institute of Physics, Faculty of Physics, Astronomy and Applied Computer Science, Jagiellonian University, 30-348 Kraków, Poland
- Laboratory for Cell and Tissue Engineering, Department of Biomedical Engineering, Eindhoven University of Technology, 5600 MB Eindhoven, The Netherlands
| | - Lindsay Russell
- Undergraduate Program, Barnard College of Columbia University, New York, NY 10027, USA;
| | - Zenon Rajfur
- Institute of Physics, Faculty of Physics, Astronomy and Applied Computer Science, Jagiellonian University, 30-348 Kraków, Poland
- Jagiellonian Center of Biomedical Imaging, Jagiellonian University, 30-348 Kraków, Poland
| |
Collapse
|
43
|
Avera AD, Gibson DJ, Birge ML, Schnorbus TN, Concannon IM, Kim Y. Characterization of Native Extracellular Matrix of Patient-Derived Glioblastoma Multiforme Organoids. Tissue Eng Part A 2025. [PMID: 39969953 DOI: 10.1089/ten.tea.2024.0303] [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: 02/20/2025] Open
Abstract
Model systems play a crucial role in biological and biomedical research, especially in the search for new treatments for challenging diseases such as glioblastoma multiforme (GBM). Organoids are 3D in vitro multicellular "middle-ground" model systems that recapitulate highly organized and heterogeneous in vivo organ-like systems, often through stem cell differentiation. Incorporating Matrigel™ or other exogenous extracellular matrices (ECMs) that do not naturally occur in the human body is common practice for organoid generation, ignoring the role of dynamic reciprocity between the cells and the ECM in tissue development. In this study, we describe a method to develop GBM organoids (GBOs) from cells without the need for exogenous ECM encapsulation and without cell culture media changes to produce stable tissue-like organoids that reach a 4 mm diameter in as little as 6 weeks. We observed a transition from homogenous cell populations to tissue-like structures when GBOs were larger than 1 mm in diameter. Transcriptomic analysis revealed that the greatest gene expression changes occurred when GBOs were 2 mm in diameter, with collagen VI as the most upregulated ECM-related gene. Quantitative and histochemical assessments further supported native ECM synthesis with significantly higher levels of glycosaminoglycans and collagen in GBOs compared with spheroids. To our knowledge, this study presents the first reproducibly large GBOs with natively produced ECMs. Organoids with natively synthesized ECMs promise to eliminate artifacts and variability from aged, homogeneic, or xenogeneic scaffolds and to provide insights for ECM-targeted drug development.
Collapse
Affiliation(s)
- Alexandra D Avera
- Department of Chemical and Biological Engineering, The University of Alabama, Tuscaloosa, Alabama, USA
| | - Daniel J Gibson
- Capstone College of Nursing, The University of Alabama, Tuscaloosa, Alabama, USA
| | - Macy L Birge
- Department of Chemical and Biological Engineering, The University of Alabama, Tuscaloosa, Alabama, USA
| | - Taylor N Schnorbus
- Department of Chemical and Biological Engineering, The University of Alabama, Tuscaloosa, Alabama, USA
| | - Isabella M Concannon
- Department of Chemical and Biological Engineering, The University of Alabama, Tuscaloosa, Alabama, USA
| | - Yonghyun Kim
- Department of Chemical and Biological Engineering, The University of Alabama, Tuscaloosa, Alabama, USA
| |
Collapse
|
44
|
Maiques O, Sallan MC, Laddach R, Pandya P, Varela A, Crosas-Molist E, Barcelo J, Courbot O, Liu Y, Graziani V, Arafat Y, Sewell J, Rodriguez-Hernandez I, Fanshawe B, Jung-Garcia Y, Imbert PR, Grasset EM, Albrengues J, Santacana M, Macià A, Tarragona J, Matias-Guiu X, Marti RM, Tsoka S, Gaggioli C, Orgaz JL, Fruhwirth GO, Wallberg F, Betteridge K, Reyes-Aldasoro CC, Haider S, Braun A, Karagiannis SN, Elosegui-Artola A, Sanz-Moreno V. Matrix mechano-sensing at the invasive front induces a cytoskeletal and transcriptional memory supporting metastasis. Nat Commun 2025; 16:1394. [PMID: 39952917 PMCID: PMC11829002 DOI: 10.1038/s41467-025-56299-7] [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: 10/30/2024] [Accepted: 01/13/2025] [Indexed: 02/17/2025] Open
Abstract
The extracellular matrix (ECM) controls tumour dissemination. We characterise ECM organization in human and mouse tumours, identifying three regions: tumour body, proximal invasive front and distal invasive front. Invasive areas show increased matrix density, fibre thickness, length, and alignment, with unique radial fibre orientation at the distal invasive front correlating with amoeboid invasive features. Using patient samples and murine models, we find that metastases recapitulate ECM features of the primary tumour. Ex vivo culture of murine cancer cells isolated from the different tumour regions reveals a spatial cytoskeletal and transcriptional memory. Several in vitro models recapitulate the in vivo ECM organisation showing that increased matrix induces 3D confinement supporting Rho-ROCK-Myosin II activity, while radial orientation enhances directional invasion. Spatial transcriptomics identifies a mechano-inflammatory program associated with worse prognosis across multiple tumour types. These findings provide mechanistic insights into how ECM organization shapes local invasion and distant metastasis.
Collapse
Affiliation(s)
- Oscar Maiques
- Cytoskeleton and metastasis Team, The Breast Cancer Now Toby Robins Research Centre Division of Breast Cancer Research, The Institute of Cancer Research, Chester Beatty Laboratories, London, SW3 6JB, UK
- Centre for Tumour Microenvironment, Barts Cancer Institute, Queen Mary University of London, John Vane Science Building, Charterhouse Square, London, EC1M 6BQ, UK
- Randall Division of Cell and Molecular Biophysics, King's College London, New Hunt's House, Guy's Campus, London, SE1 1UL, UK
- Cancer Biomarkers & Biotherapeutics, Barts Cancer Institute, Queen Mary University of London, London, UK
| | - Marta C Sallan
- Cytoskeleton and metastasis Team, The Breast Cancer Now Toby Robins Research Centre Division of Breast Cancer Research, The Institute of Cancer Research, Chester Beatty Laboratories, London, SW3 6JB, UK
- Centre for Haemato-Oncology, Barts Cancer Institute, Queen Mary University of London, John Vane Science Building, Charterhouse Square, London, EC1M 6BQ, UK
| | - Roman Laddach
- St. John's Institute of Dermatology, School of Basic and Medical Biosciences, King's College London, SE1 9RT, London, UK
- Department of Informatics, Faculty of Natural and Mathematical Sciences, King's College London, Bush House, London, WC2B 4BG, UK
| | - Pahini Pandya
- Randall Division of Cell and Molecular Biophysics, King's College London, New Hunt's House, Guy's Campus, London, SE1 1UL, UK
| | - Adrian Varela
- Cytoskeleton and metastasis Team, The Breast Cancer Now Toby Robins Research Centre Division of Breast Cancer Research, The Institute of Cancer Research, Chester Beatty Laboratories, London, SW3 6JB, UK
- Centre for Tumour Microenvironment, Barts Cancer Institute, Queen Mary University of London, John Vane Science Building, Charterhouse Square, London, EC1M 6BQ, UK
| | - Eva Crosas-Molist
- Cytoskeleton and metastasis Team, The Breast Cancer Now Toby Robins Research Centre Division of Breast Cancer Research, The Institute of Cancer Research, Chester Beatty Laboratories, London, SW3 6JB, UK
- Centre for Tumour Microenvironment, Barts Cancer Institute, Queen Mary University of London, John Vane Science Building, Charterhouse Square, London, EC1M 6BQ, UK
- Randall Division of Cell and Molecular Biophysics, King's College London, New Hunt's House, Guy's Campus, London, SE1 1UL, UK
| | - Jaume Barcelo
- Cytoskeleton and metastasis Team, The Breast Cancer Now Toby Robins Research Centre Division of Breast Cancer Research, The Institute of Cancer Research, Chester Beatty Laboratories, London, SW3 6JB, UK
- Centre for Tumour Microenvironment, Barts Cancer Institute, Queen Mary University of London, John Vane Science Building, Charterhouse Square, London, EC1M 6BQ, UK
| | - Olivia Courbot
- Cell and Tissue Mechanobiology Laboratory, The Francis Crick Institute, London, NW1 1AT, UK
| | - Yanbo Liu
- Cytoskeleton and metastasis Team, The Breast Cancer Now Toby Robins Research Centre Division of Breast Cancer Research, The Institute of Cancer Research, Chester Beatty Laboratories, London, SW3 6JB, UK
| | - Vittoria Graziani
- Cytoskeleton and metastasis Team, The Breast Cancer Now Toby Robins Research Centre Division of Breast Cancer Research, The Institute of Cancer Research, Chester Beatty Laboratories, London, SW3 6JB, UK
- Centre for Tumour Microenvironment, Barts Cancer Institute, Queen Mary University of London, John Vane Science Building, Charterhouse Square, London, EC1M 6BQ, UK
| | - Youssef Arafat
- Department of Computer Science, City St George's, University of London, London, UK
| | - Joanne Sewell
- Centre for Tumour Microenvironment, Barts Cancer Institute, Queen Mary University of London, John Vane Science Building, Charterhouse Square, London, EC1M 6BQ, UK
- Randall Division of Cell and Molecular Biophysics, King's College London, New Hunt's House, Guy's Campus, London, SE1 1UL, UK
| | - Irene Rodriguez-Hernandez
- Centre for Tumour Microenvironment, Barts Cancer Institute, Queen Mary University of London, John Vane Science Building, Charterhouse Square, London, EC1M 6BQ, UK
- Randall Division of Cell and Molecular Biophysics, King's College London, New Hunt's House, Guy's Campus, London, SE1 1UL, UK
| | - Bruce Fanshawe
- Randall Division of Cell and Molecular Biophysics, King's College London, New Hunt's House, Guy's Campus, London, SE1 1UL, UK
- Comprehensive Cancer Centre, School of Cancer and Pharmaceutical Sciences, King's College London, London, SE1 1UL, UK
| | - Yaiza Jung-Garcia
- Cytoskeleton and metastasis Team, The Breast Cancer Now Toby Robins Research Centre Division of Breast Cancer Research, The Institute of Cancer Research, Chester Beatty Laboratories, London, SW3 6JB, UK
- Centre for Tumour Microenvironment, Barts Cancer Institute, Queen Mary University of London, John Vane Science Building, Charterhouse Square, London, EC1M 6BQ, UK
| | - Paul Rc Imbert
- CMR Advanced Bio-imaging Facility, Centre for Microvascular Research, Queen Mary University of London, John Vane Science Building, Charterhouse Square, London, EC1M 6BQ, UK
| | - Eloise M Grasset
- University Cote d'Azur, CNRS UMR7284, INSERM U1081, Institute for Research on Cancer and Aging, Nice (IRCAN), Nice, France
| | - Jean Albrengues
- University Cote d'Azur, CNRS UMR7284, INSERM U1081, Institute for Research on Cancer and Aging, Nice (IRCAN), Nice, France
| | - Maria Santacana
- Department of Pathology and Molecular Genetics, Hospital Universitari Arnau de Vilanova, University of Lleida, IRBLleida, CIBERONC, Lleida, 25198, Spain
| | - Anna Macià
- Oncologic Pathology Group, IRBLleida, Departments of Experimental Medicine and Basic Medical Sciences, University of Lleida, Lleida, 25198, Spain
| | - Jordi Tarragona
- Department of Pathology and Molecular Genetics, Hospital Universitari Arnau de Vilanova, University of Lleida, IRBLleida, CIBERONC, Lleida, 25198, Spain
| | - Xavier Matias-Guiu
- Department of Pathology and Molecular Genetics, Hospital Universitari Arnau de Vilanova, University of Lleida, IRBLleida, CIBERONC, Lleida, 25198, Spain
- Oncologic Pathology Group, IRBLleida, Departments of Experimental Medicine and Basic Medical Sciences, University of Lleida, Lleida, 25198, Spain
- Department of Pathology, Hospital Universitari de Bellvitge University of Barcelona, IDIBELL, CIBERONC, L'Hospitalet-, Barcelona, 08907, Spain
| | - Rosa M Marti
- Department of Dermatology, Hospital Universitari Arnau de Vilanova, CIBERONC, University of Lleida, CIBERONC, IRB Lleida, Lleida, 25198, Spain
| | - Sophia Tsoka
- Department of Informatics, Faculty of Natural and Mathematical Sciences, King's College London, Bush House, London, WC2B 4BG, UK
| | - Cedric Gaggioli
- University Cote d'Azur, CNRS UMR7284, INSERM U1081, Institute for Research on Cancer and Aging, Nice (IRCAN), Nice, France
| | - Jose L Orgaz
- Centre for Tumour Microenvironment, Barts Cancer Institute, Queen Mary University of London, John Vane Science Building, Charterhouse Square, London, EC1M 6BQ, UK
- Randall Division of Cell and Molecular Biophysics, King's College London, New Hunt's House, Guy's Campus, London, SE1 1UL, UK
- Instituto de Investigaciones Biomédicas Sols-Morreale (IIBM), CSIC-UAM, 28029, Madrid, Spain
| | - Gilbert O Fruhwirth
- Comprehensive Cancer Centre, School of Cancer and Pharmaceutical Sciences, King's College London, London, SE1 1UL, UK
| | - Fredrik Wallberg
- Quell Therapeutics, Translation & Innovation Hub, 84 Wood Ln, London, W12 0BZ, UK
- Light Microscopy Facility, The Institute of Cancer Research, Chester Beatty Laboratories, London, SW3 6JB, UK
| | - Kai Betteridge
- Light Microscopy Facility, The Institute of Cancer Research, Chester Beatty Laboratories, London, SW3 6JB, UK
| | - Constantino Carlos Reyes-Aldasoro
- Department of Computer Science, City St George's, University of London, London, UK
- Integrated Pathology Unit, Division of Molecular Pathology, The Institute of Cancer Research, Sutton, UK
| | - Syed Haider
- Breast Cancer Research Bioinformatics Group, Chester Beatty Laboratories, London, SW3 6JB, UK
| | - Andrejs Braun
- Centre for Haemato-Oncology, Barts Cancer Institute, Queen Mary University of London, John Vane Science Building, Charterhouse Square, London, EC1M 6BQ, UK
| | - Sophia N Karagiannis
- St. John's Institute of Dermatology, School of Basic and Medical Biosciences, King's College London, SE1 9RT, London, UK
- Breast Cancer Now Research Unit, School of Cancer & Pharmaceutical Sciences, King's College London, Guy's Cancer Centre, London, SE1 9RT, UK
| | | | - Victoria Sanz-Moreno
- Cytoskeleton and metastasis Team, The Breast Cancer Now Toby Robins Research Centre Division of Breast Cancer Research, The Institute of Cancer Research, Chester Beatty Laboratories, London, SW3 6JB, UK.
- Centre for Tumour Microenvironment, Barts Cancer Institute, Queen Mary University of London, John Vane Science Building, Charterhouse Square, London, EC1M 6BQ, UK.
- Randall Division of Cell and Molecular Biophysics, King's College London, New Hunt's House, Guy's Campus, London, SE1 1UL, UK.
| |
Collapse
|
45
|
Ilg MM, Lapthorn AR, Harding SL, Minhas T, Koduri G, Bustin SA, Cellek S. Development of a phenotypic screening assay to measure activation of cancer-associated fibroblasts. Front Pharmacol 2025; 16:1526495. [PMID: 40017592 PMCID: PMC11865240 DOI: 10.3389/fphar.2025.1526495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Accepted: 01/27/2025] [Indexed: 03/01/2025] Open
Abstract
Background In cancer metastasis, tumor cells condition distant tissues to create a supportive environment, or metastatic niche, by driving the activation of cancer-associated fibroblasts (CAFs). These CAFs remodel the extracellular matrix, creating a microenvironment that supports tumor growth and compromises immune cell function, enabling cancer cells to evade immune detection. Consequently, targeting the activation of CAFs has been proposed as a therapeutic strategy to hinder metastatic spread. Our objective was to develop the first in vitro phenotypic screening assay capable of assessing this activation process. Methods Human primary lung fibroblasts were co-cultured with highly invasive breast cancer cells (MDA-MB-231) to identify changes in the expression of selected genes using RT-qPCR. An In-Cell ELISA (ICE)-based assay using human lung fibroblasts, MDA-MB-231 cells and human monocytes (THP-1 cells) was developed to measure the activation of CAFs. Another ELISA assay was used to measure released osteopontin. Results When lung fibroblast were co-cultured with MDA-MB-231 cells, among the 10 selected genes, the genes for osteopontin (SPP1), insulin like growth factor 1 (IGF1), periostin (POSTN) and α-smooth muscle actin (α-SMA, ACTA2) elicited the greatest fold change (55-, 37-, 8- and 5-fold respectively). Since osteopontin, IGF-1 and periostin are secreted proteins and α-SMA is an intracellular cytoskeleton protein, α-SMA was chosen to be the readout biomarker for the ICE assay. When fibroblasts were co-cultured with MDA-MB-231 cells and monocytes in the 96 well ICE assay, α-SMA expression was increased 2.3-fold yielding a robust Z' of 0.56. A secondary, low throughput assay was developed by measuring the release of osteopontin which showed a 6-fold increase when fibroblasts were co-cultured with MDA-MB-231 cells and monocytes. Discussion This phenotypic assay is the first to measure the activation of CAFs in a 96-well format, making it suitable for medium-to high-throughput screening of potential therapeutic compounds. By focusing on observable cellular phenotypic changes rather than targeting specific molecular pathways, this assay allows for a broader and unbiased identification of compounds capable of modulating CAF activation.
Collapse
Affiliation(s)
- Marcus M. Ilg
- Fibrosis Research Group, Medical Technology Research Centre, Anglia Ruskin University, Chelmsford, United Kingdom
| | - Alice R. Lapthorn
- Fibrosis Research Group, Medical Technology Research Centre, Anglia Ruskin University, Chelmsford, United Kingdom
| | - Sophie L. Harding
- Fibrosis Research Group, Medical Technology Research Centre, Anglia Ruskin University, Chelmsford, United Kingdom
| | - Tariq Minhas
- The Essex Cardiothoracic Centre, Basildon University Hospital, Basildon, United Kingdom
| | - Gouri Koduri
- Southend University Hospital NHS Foundation Trust, Westcliff-on-Sea, United Kingdom
| | - Stephen A. Bustin
- Fibrosis Research Group, Medical Technology Research Centre, Anglia Ruskin University, Chelmsford, United Kingdom
| | - Selim Cellek
- Fibrosis Research Group, Medical Technology Research Centre, Anglia Ruskin University, Chelmsford, United Kingdom
| |
Collapse
|
46
|
Metzcar J, Duggan BS, Fischer B, Murphy M, Heiland R, Macklin P. A Simple Framework for Agent-Based Modeling with Extracellular Matrix. Bull Math Biol 2025; 87:43. [PMID: 39937344 PMCID: PMC11821717 DOI: 10.1007/s11538-024-01408-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Accepted: 12/21/2024] [Indexed: 02/13/2025]
Abstract
Extracellular matrix (ECM) is a key component of the cellular microenvironment and critical in multiple disease and developmental processes. Representing ECM and cell-ECM interactions is a challenging multiscale problem as they span molecular-level details to tissue-level dynamics. While several computational frameworks exist for ECM modeling, they often focus on very detailed modeling of individual ECM fibers or represent only a single aspect of the ECM. Using the PhysiCell agent-based modeling platform, we developed a framework of intermediate detail with the ability to capture bidirectional cell-ECM interactions. We represent a small region of ECM, an ECM element, with three variables describing its local microstructure: anisotropy, density, and overall fiber orientation. To spatially model the ECM, we use an array of ECM elements. Cells remodel local ECM microstructure and in turn, local microstructure impacts cellular motility. We demonstrate the utility of this framework and reusability of its core cell-ECM interaction model through examples in cellular invasion, wound healing, basement membrane degradation, and leader-follower collective migration. Despite the relative simplicity of the framework, it is able to capture a broad range of cell-ECM interactions of interest to the modeling community. Furthermore, variables representing the ECM microstructure are accessible through simple programming interfaces. This allows them to impact cell behaviors, such as proliferation and death, without requiring custom code for each interaction, particularly through PhysiCell's modeling grammar, enabling rapid modeling of a diverse range of cell-matrix biology. We make this framework available as a free and open source software package at https://github.com/PhysiCell-Models/collective-invasion .
Collapse
Affiliation(s)
- John Metzcar
- Intelligent Systems Engineering, Indiana University, 700 N. Woodlawn, Bloomington, IN, 47408, USA
- Informatics, Indiana University, 901 E. Tenth Street, Bloomington, IN, 47408, USA
| | - Ben S Duggan
- Computer Science, Indiana University, 700 N. Woodlawn, Bloomington, IN, 47408, USA
| | - Brandon Fischer
- Intelligent Systems Engineering, Indiana University, 700 N. Woodlawn, Bloomington, IN, 47408, USA
| | - Matthew Murphy
- Informatics, Indiana University, 901 E. Tenth Street, Bloomington, IN, 47408, USA
| | - Randy Heiland
- Intelligent Systems Engineering, Indiana University, 700 N. Woodlawn, Bloomington, IN, 47408, USA
| | - Paul Macklin
- Intelligent Systems Engineering, Indiana University, 700 N. Woodlawn, Bloomington, IN, 47408, USA.
| |
Collapse
|
47
|
Yu F, Zhu C, Wu W. Senile Osteoarthritis Regulated by the Gut Microbiota: From Mechanisms to Treatments. Int J Mol Sci 2025; 26:1505. [PMID: 40003971 PMCID: PMC11855920 DOI: 10.3390/ijms26041505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2024] [Revised: 01/19/2025] [Accepted: 01/26/2025] [Indexed: 02/27/2025] Open
Abstract
Osteoarthritis (OA) is a chronic, progressive degenerative joint disease that affects the entire synovial joint, leading to the progressive degeneration of articular cartilage. It seriously affects the quality of life and global disability of patients. OA is affected by a variety of factors; the most significant risk factor for OA is age. As individuals age, the risk and severity of OA increase due to the exacerbation of cartilage degeneration and wear and tear. In recent years, research has indicated that the gut microbiota may play a significant role in the aging and OA processes. It is anticipated that regulating the gut microbiota may offer novel approaches to the treatment of OA. The objective of this paper is to examine the relationship between the gut microbiota and senile OA, to investigate the potential mechanisms involved. This review also summarizes the therapeutic strategies related to gut flora in OA management, such as prebiotics and probiotics, diet, exercise, traditional Chinese medicine (TCM) modification, and fecal microbiota transplantation (FMT), highlighting the potential clinical value of gut flora and elucidating the current challenges. The foundation for future research directions is established through the summarization of current research progress.
Collapse
Affiliation(s)
- Fan Yu
- School of Exercise and Health, Shanghai University of Sports, Shanghai 200438, China; (F.Y.); (C.Z.)
| | - Chenyu Zhu
- School of Exercise and Health, Shanghai University of Sports, Shanghai 200438, China; (F.Y.); (C.Z.)
| | - Wei Wu
- School of Athletic Performance, Shanghai University of Sports, Shanghai 200438, China
| |
Collapse
|
48
|
Frostadottir D, Welinder C, Perez R, Dahlin LB. Refinement of Protein Extraction Protocols for Human Peripheral Nerve Tissue. ACS OMEGA 2025; 10:5111-5118. [PMID: 39959086 PMCID: PMC11822717 DOI: 10.1021/acsomega.4c11373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 01/14/2025] [Accepted: 01/23/2025] [Indexed: 02/18/2025]
Abstract
Our aim was to establish an effective method for protein extraction from freshly frozen human peripheral nerves, determine the minimum amount required for consistent protein extraction outcomes, and assess which method produced the highest number of protein identities. Five extraction methods were compared using 8 M urea and Ripa buffer using either the Bullet Blender or Bioruptor. Out of the total 2619 identified proteins, protein extraction using the Ripa buffer combined with either Bioruptor or Bullet Blender resulted in the identification of 1582 (60%) and 1615 (62%) proteins, respectively. In contrast, using 8 M urea and Bioruptor for protein extraction resulted in 1022 proteins (39%), whereas employing Bullet Blender yielded 1446 proteins (55%). Sample amounts, ranging from 0.6 to 10 mg, were prepared with consistent protein extraction outcome obtained for samples ≥1.2 mg. Combining Ripa and 8 M urea with Bullet Blender increased protein identification to 2126 (81%). Proteins were classified by their cell components, molecular functions, and biological processes. Furthermore, a subclassification of proteins involved in the extracellular matrix (ECM) was introduced. We recommend the use of Ripa buffer, in combination with 8 M urea and Bullet Blender for extracting proteins from fresh-frozen human nerves weighing ≥1.2 mg.
Collapse
Affiliation(s)
- Drifa Frostadottir
- Department
of Translational Medicine − Hand Surgery, Lund University, Malmö S-20502, Sweden
- Department
of Hand Surgery, Skane University Hospital, Malmö S-20502, Sweden
| | - Charlotte Welinder
- Faculty of
Medicine, Department of Clinical Sciences Lund, Mass Spectrometry, Lund University, Lund S-20502, Sweden
| | - Raquel Perez
- Department
of Translational Medicine − Hand Surgery, Lund University, Malmö S-20502, Sweden
- Unit for
Social Epidemiology, Department of Clinical Sciences Malmö, Lund University, Malmö S-20502, Sweden
| | - Lars B. Dahlin
- Department
of Translational Medicine − Hand Surgery, Lund University, Malmö S-20502, Sweden
- Department
of Hand Surgery, Skane University Hospital, Malmö S-20502, Sweden
- Department
of Biomedical and Clinical Sciences, Linköping
University, SE-581 83 Linköping, Sweden
| |
Collapse
|
49
|
Liu Y, Zhong W, Ai Y, Xing M. Double Cross-linked Methacrylated Carboxymethyl Pea Starch Cryogels with Highly Compressive Elasticity and Hemostatic Function. Biomacromolecules 2025; 26:883-899. [PMID: 39865611 DOI: 10.1021/acs.biomac.4c01080] [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: 01/28/2025]
Abstract
As an abundant renewable natural material, starch has attracted unprecedented interest in the biomedical field. Carboxylated starch particles have been investigated for topical hemostasis, but the powder may not provide physical protection or support for wounds. Here, we prepared macroporous cryogel sponges of methacrylated carboxymethyl starch (CM-ST-MA) containing a covalent and a calcium ionic double network. The second ionic cross-linking network enhanced the compressive strength and toughness dramatically but reduced the swelling ratios. Cryogels and sponges exhibited excellent compressive elasticity at low Ca2+ concentrations (0.01 M). Cryogels became more plastic and dry sponges became rigid and brittle at high Ca2+ concentrations. The cryogels have outstanding wet-thermal stability but are still degradable via enzymatic hydrolysis. All CM-ST-MA sponges showed excellent biocompatibility, hemocompatibility, and outstanding hemostasis in in vitro assays. In the in vivo mouse tail amputation model, both CM-ST-MA cryogels without or with Ca2+ (0.01 M) reduced the blood loss and bleeding time significantly.
Collapse
Affiliation(s)
- Yuqing Liu
- Department of Mechanical Engineering, University of Manitoba, Winnipeg, MB R3T 2N2, Canada
| | - Wen Zhong
- Department of Biosystems Engineering, University of Manitoba, Winnipeg, MB R3T 2N2, Canada
| | - Yongfeng Ai
- Department of Food and Bioproduct Sciences, University of Saskatchewan, Saskatoon, Saskatchewan S7N 5A8, Canada
| | - Malcolm Xing
- Department of Mechanical Engineering, University of Manitoba, Winnipeg, MB R3T 2N2, Canada
| |
Collapse
|
50
|
Kim YH, Kim HS, Hong IS. Advances in biomaterials-based tissue engineering for regeneration of female reproductive tissues. Biofabrication 2025; 17:022001. [PMID: 39854843 DOI: 10.1088/1758-5090/adae38] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Accepted: 01/24/2025] [Indexed: 01/27/2025]
Abstract
The anatomical components of the female reproductive system-comprising the ovaries, uterus, cervix, vagina, and fallopian tubes-interact intricately to provide the structural and hormonal support essential for reproduction. However, this system is susceptible to various detrimental factors, both congenital and acquired, that can impair fertility and adversely affect quality of life. Recent advances in bioengineering have led to the development of sophisticated three-dimensional models that mimic the complex architecture and functionality of reproductive organs. These models, incorporating diverse cell types and tissue layers, are crucial for understanding physiological processes within the reproductive tract. They offer insights into decidualization, ovulation, folliculogenesis, and the progression of reproductive cancers, thereby enhancing personalized medical treatments and addressing female infertility. This review highlights the pivotal role of tissue engineering in diagnosing and treating female infertility, emphasizing the importance of considering factors like biocompatibility, biomaterial selection, and mechanical properties in the design of bioengineered systems. The challenge of replicating the functionally specialized and structurally complex organs, such as the uterus and ovary, underscores the need for reliable techniques that improve morphological and functional restoration. Despite substantial progress, the goal of creating a fully artificial female reproductive system is still a challenge. Nonetheless, the recent fabrication of artificial ovaries, uteruses, cervixes, and vaginas marks significant advancements toward this aim. Looking forward, the challenges in bioengineering are expected to spur further innovations in both basic and applied sciences, potentially hastening the clinical adoption of these technologies.
Collapse
Affiliation(s)
- Yong Ho Kim
- Gachon Pain Center and Department of Physiology, College of Medicine, Gachon University, Incheon 21999, Republic of Korea
| | - Hyung-Sik Kim
- Department of Oral Biochemistry, Dental and Life Science Institute, School of Dentistry, Pusan National University, Yangsan 50612, Republic of Korea
| | - In-Sun Hong
- Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon 21999, Republic of Korea
- Department of Molecular Medicine, School of Medicine, Gachon University, Incheon 406-840, Republic of Korea
| |
Collapse
|