1
|
Ladeira B, Gomes M, Wei K, Custódio C, Mano J. Supramolecular assembly of multi-purpose tissue engineering platforms from human extracellular matrix. Biomaterials 2025; 320:123270. [PMID: 40132356 DOI: 10.1016/j.biomaterials.2025.123270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 02/07/2025] [Accepted: 03/17/2025] [Indexed: 03/27/2025]
Abstract
Recapitulating the biophysical and biochemical complexity of the extracellular matrix (ECM) remains a major challenge in tissue engineering. Hydrogels derived from decellularized ECM provide a unique opportunity to replicate the architecture and bioactivity of native ECM, however, they exhibit limited long-term stability and mechanical integrity. In turn, materials assembled through supramolecular interactions have achieved considerable success in replicating the dynamic biophysical properties of the ECM. Here, we merge both methodologies by promoting the supramolecular assembly of decellularized human amniotic membrane (hAM), mediated by host-guest interactions between hAM proteins and acryloyl-β-cyclodextrin (AcβCD). Photopolymerization of the cyclodextrins results in the formation of soft hydrogels that exhibit tunable stress relaxation and strain-stiffening. Disaggregation of bulk hydrogels yields an injectable granular material that self-reconstitutes into shape-adaptable bulk hydrogels, supporting cell delivery and promoting neovascularization. Additionally, cells encapsulated within bulk hydrogels sense and respond to the biophysical properties of the surrounding matrix, as early cell spreading is favored in hydrogels that exhibit greater susceptibility to applied stress, evidencing proper cell-matrix interplay. Thus, this system is shown to be a promising substitute for native ECM in tissue repair and modelling.
Collapse
Affiliation(s)
- Bruno Ladeira
- Department of Chemistry, CICECO-Aveiro Institute of Materials, University of Aveiro, Campus Universitário de Santiago, 3810-193, Aveiro, Portugal
| | - Maria Gomes
- Department of Chemistry, CICECO-Aveiro Institute of Materials, University of Aveiro, Campus Universitário de Santiago, 3810-193, Aveiro, Portugal
| | - Kongchang Wei
- Empa, Swiss Federal Laboratories for Materials Science and Technology, Laboratory for Biomimetic Membranes and Textiles, St. Gallen, Switzerland; Empa, Swiss Federal Laboratories for Materials Science and Technology, Laboratory for Biointerfaces, St. Gallen, Switzerland
| | - Catarina Custódio
- Department of Chemistry, CICECO-Aveiro Institute of Materials, University of Aveiro, Campus Universitário de Santiago, 3810-193, Aveiro, Portugal
| | - João Mano
- Department of Chemistry, CICECO-Aveiro Institute of Materials, University of Aveiro, Campus Universitário de Santiago, 3810-193, Aveiro, Portugal.
| |
Collapse
|
2
|
Hameed S, Rahman SU, Konain K, Samie M, Farid S, Elango J, Habib SR, Woo KM, Arany PR. Advanced 3D biomimetic scaffolds with bioactive glass and bone-conditioned medium for enhanced osteogenesis. BIOMATERIALS ADVANCES 2025; 173:214282. [PMID: 40081288 DOI: 10.1016/j.bioadv.2025.214282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 03/06/2025] [Accepted: 03/08/2025] [Indexed: 03/15/2025]
Abstract
The study focuses on developing and evaluating 3D biomimetic fibrous scaffolds to enhance osteoblast differentiation and bone tissue regeneration. Utilizing a synergistic approach, biological and chemical factors were compartmentalized within the fibrous scaffolds through co-axial electrospinning. Bioactive glass (BG) was used for osteo-conductivity, and Bone-Conditioned Medium (BCM) for osteoinduction. The BCM, derived from ovine bone chips, was investigated for its optimal concentration using pre-osteoblast cells. Comprehensive assessment of the scaffolds included physicochemical properties, drug release, cell viability, and osteogenic potential. The scaffold's architecture, confirmed by Scanning electron microscopy (SEM) analysis, effectively emulated the natural extracellular matrix (ECM). Energy Dispersive X-ray Spectroscopy (EDX) and Fourier Transform Infrared Spectroscopy (FTIR) analyses verified the successful integration of BG and BCM, while UV-Vis spectroscopy demonstrated controlled BCM release. Both BG and BCM scaffolds notably enhanced osteoblast differentiation, as evident with Alizarin red staining. The combined use of BG and BCM in scaffolds synergistically promoted osteogenic differentiation and viability of MC3T3-E1 cells. Furthermore, these scaffolds significantly increased the expression of Bone Sialoprotein (BSP), Osteocalcin (OCN), and Runt-related transcription factor 2 (RUNX2) which indicate increase in osteogenic differentiation. This study provides evidence for advanced scaffold systems that can guide cell responses for effective bone tissue regeneration.
Collapse
Affiliation(s)
- Shazia Hameed
- Oral Biology, Institute of Basic Medical Sciences, Khyber Medical University, Peshawar, Pakistan
| | - Saeed Ur Rahman
- Oral Biology, Institute of Basic Medical Sciences, Khyber Medical University, Peshawar, Pakistan; Oral Biology, Surgery and Biomedical Engineering, University at Buffalo, NY, USA.
| | - Kiran Konain
- Molecular Biology, Institute of Basic Medical Sciences, Khyber Medical University, Peshawar, Pakistan
| | - Muhammad Samie
- Institute of Pharmaceutical Sciences, Khyber Medical University, Peshawar, Pakistan
| | - Sajida Farid
- Oral Biology, Institute of Basic Medical Sciences, Khyber Medical University, Peshawar, Pakistan
| | - Jeevithan Elango
- Department of Biomaterials Engineering, Faculty of Health Sciences, UCAM- Universidad Católica San Antonio de Murcia, Guadalupe 30107, Murcia, Spain; Department of Marine Bio-Pharmacology, College of Food Science and Technology, Shanghai Ocean University, Shanghai 201306, China
| | - Syed Rashid Habib
- Department of Prosthetic Dental Sciences, College of Dentistry, King Saud University, Riyadh 11545, Saudi Arabia
| | - Kyung Mi Woo
- Department of Molecular Genetics, Dental Research Institute, School of Dentistry, Seoul National University, Seoul 08826, Republic of Korea
| | - Praveen R Arany
- Oral Biology, Surgery and Biomedical Engineering, University at Buffalo, NY, USA
| |
Collapse
|
3
|
Krawiec A, Pietrasik J, Pietrasik Z, Mikuła-Pietrasik J, Książek K. Unveiling the role of extracellular matrix elements and regulators in shaping ovarian cancer growth and metastasis. Cell Signal 2025; 132:111843. [PMID: 40318796 DOI: 10.1016/j.cellsig.2025.111843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2025] [Revised: 04/18/2025] [Accepted: 04/29/2025] [Indexed: 05/07/2025]
Abstract
Epithelial ovarian cancer (EOC) progression is determined by numerous intracellular interactions and the interplay between malignant cells, normal cells, and the tumor acellular microenvironment, formed largely by the extracellular matrix (ECM). The structure and biochemical functioning of various ECM components, along with the activity of agents that regulate ECM remodeling, impact the disease's expansion (adhesion, proliferation, invasion), spread, and response to therapy. It is important to note that the involvement of ECM components and their regulators in the progression of EOC is bidirectional and distinctly depends on a particular tissue context. In certain situations, certain components of the ECM enhance the activity of cancer cells, but in other scenarios, they suppress it. In this review, we summarize the newest knowledge regarding diverse aspects of ECM engagement in EOC pathophysiology and chemotherapy. Moreover, we delineate conditions that exacerbate the pro-cancerous properties of ECM, including diabetes-associated glycation, aging, and cellular senescence. We also explore methods to therapeutically alter the properties of the ECM, which could be beneficial in ovarian cancer prevention and treatment.
Collapse
Affiliation(s)
- Adrianna Krawiec
- Poznan University of Medical Sciences, Department of Pathophysiology of Ageing and Civilization Diseases, Święcickiego 4 Str, 60-781 Poznań, Poland.
| | - Joanna Pietrasik
- Poznan University of Medical Sciences, Department of Pathophysiology of Ageing and Civilization Diseases, Święcickiego 4 Str, 60-781 Poznań, Poland
| | - Zofia Pietrasik
- Poznan University of Medical Sciences, Department of Pathophysiology of Ageing and Civilization Diseases, Święcickiego 4 Str, 60-781 Poznań, Poland
| | - Justyna Mikuła-Pietrasik
- Poznan University of Medical Sciences, Department of Pathophysiology of Ageing and Civilization Diseases, Święcickiego 4 Str, 60-781 Poznań, Poland.
| | - Krzysztof Książek
- Poznan University of Medical Sciences, Department of Pathophysiology of Ageing and Civilization Diseases, Święcickiego 4 Str, 60-781 Poznań, Poland.
| |
Collapse
|
4
|
Yang Y, Li L, Dai F, Deng L, Yang K, He C, Chen Y, Yang X, Song L. Fibroblast-derived versican exacerbates periodontitis progression by regulating macrophage migration and inflammatory cytokine secretion. Cell Signal 2025; 131:111755. [PMID: 40112905 DOI: 10.1016/j.cellsig.2025.111755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2025] [Revised: 03/05/2025] [Accepted: 03/17/2025] [Indexed: 03/22/2025]
Abstract
OBJECTIVE Versican (VCAN), a prominent extracellular matrix component upregulated in inflammatory diseases, demonstrates context-specific regulatory mechanisms. Periodontitis, a chronic inflammatory disease leading to periodontal tissue destruction and tooth loss, the pathological role of it remains poorly defined. Our study aims to examine VCAN-mediated mechanisms in periodontitis. METHODS We conducted a comprehensive analysis of bulk RNA sequencing and single-cell RNA sequencing data to examine VCAN expression level and source in periodontitis. Functional and correlation analyses were used to explore its biological functions. We then validated VCAN expression using quantitative real-time polymerase chain reaction, immunohistochemical staining, and immunofluorescence staining in animal models and investigated its biological functions in inflammation through in vitro experiments. RESULTS Our findings reveal that VCAN is mainly generated by fibroblast in periodontitis, and its expression significantly upregulated at both mRNA and protein levels. Using VCAN-overexpressing L929 cells, we demonstrated enhanced proliferative capacity and inflammatory potential. Co-culture experiments with RAW264.7 cells showed promoted migration, adhesion, M1 polarization, and mitogen-activated protein kinase (MAPK) pathway activation. CONCLUSION VCAN enhances fibroblast proliferation and migration, and upregulates inflammatory cytokines expression. Furthermore, fibroblast-derived VCAN not only induces macrophage chemotaxis, migration, adhesion, and polarization toward the proinflammatory M1 phenotype, but also activates MAPK signaling of macrophage, which may amplify inflammatory cascades to exacerbate periodontal tissue destruction. Targeted regulation of VCAN expression may become a promising precision treatment strategy for periodontitis.
Collapse
Affiliation(s)
- Yuting Yang
- Center of Stomatology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China; JXHC Key Laboratory of Periodontology, The Second Affiliated Hospital of Nanchang University, Nanchang, China; The institute of Periodontal Disease, Nanchang University, Nanchang, China; The Second Clinical Medical School, NanchangUniversity, Nanchang, China
| | - Li Li
- Center of Stomatology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China; JXHC Key Laboratory of Periodontology, The Second Affiliated Hospital of Nanchang University, Nanchang, China; The institute of Periodontal Disease, Nanchang University, Nanchang, China; The Second Clinical Medical School, NanchangUniversity, Nanchang, China
| | - Fang Dai
- Center of Stomatology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China; JXHC Key Laboratory of Periodontology, The Second Affiliated Hospital of Nanchang University, Nanchang, China; The institute of Periodontal Disease, Nanchang University, Nanchang, China
| | - Libin Deng
- School of Public Health, Jiangxi Medical College, Nanchang University, Nanchang, China; Jiangxi Provincial Key Laboratory of Disease Prevention and Public Health, Nanchang University, Nanchang, China
| | - Kaiqiang Yang
- Center of Stomatology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China; JXHC Key Laboratory of Periodontology, The Second Affiliated Hospital of Nanchang University, Nanchang, China; The institute of Periodontal Disease, Nanchang University, Nanchang, China; The Second Clinical Medical School, NanchangUniversity, Nanchang, China
| | - Chenjiang He
- Center of Stomatology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China; JXHC Key Laboratory of Periodontology, The Second Affiliated Hospital of Nanchang University, Nanchang, China; The institute of Periodontal Disease, Nanchang University, Nanchang, China; The Second Clinical Medical School, NanchangUniversity, Nanchang, China
| | - Yeke Chen
- Center of Stomatology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China; JXHC Key Laboratory of Periodontology, The Second Affiliated Hospital of Nanchang University, Nanchang, China; The institute of Periodontal Disease, Nanchang University, Nanchang, China; The Second Clinical Medical School, NanchangUniversity, Nanchang, China
| | - Xinbo Yang
- Center of Stomatology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China; JXHC Key Laboratory of Periodontology, The Second Affiliated Hospital of Nanchang University, Nanchang, China; The institute of Periodontal Disease, Nanchang University, Nanchang, China; The Second Clinical Medical School, NanchangUniversity, Nanchang, China
| | - Li Song
- Center of Stomatology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China; JXHC Key Laboratory of Periodontology, The Second Affiliated Hospital of Nanchang University, Nanchang, China; The institute of Periodontal Disease, Nanchang University, Nanchang, China.
| |
Collapse
|
5
|
Ghadirian S, Shariati L, Karbasi S. Evaluation of the effects of cartilage decellularized ECM in optimizing PHB-chitosan-HNT/chitosan-ECM core-shell electrospun scaffold: Physicochemical and biological properties. BIOMATERIALS ADVANCES 2025; 172:214249. [PMID: 40048901 DOI: 10.1016/j.bioadv.2025.214249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Revised: 02/09/2025] [Accepted: 02/25/2025] [Indexed: 03/17/2025]
Abstract
Cartilage regeneration is still a highly challenging field due to its low self-healing ability. This study used a core-shell electrospinning technique to enhance cartilage tissue engineering by incorporating cartilage extracellular matrix (ECM). The core of fibers included poly(3-hydroxybutyrate)-Chitosan (PHB-Cs) and Halloysite nanotubes. The shell of fibers consisted of Cs and ECM (0, 1, 3, 5 wt%). Subsequently, the scaffolds were named 0E, 1E, 3E, and 5E. The study aimed to assess the impact of ECM on cellular behavior and chondrogenesis. Our findings indicate that ECM reduced fiber diameter from 775 nm for the 0E scaffold to 454 nm for the 1E scaffold. Water contact angle measurements revealed an increasing trend by ECM addition, from 42° for 0E to 67° for 1E. According to mechanical analysis, the 1E scaffold represented the highest strength (5.81 MPa) and strain (3.17%). Based on these analyses, the 1E was considered the optimum scaffold. MTT analysis showed cell viability of over 80% for the 0E and 1E. Also, the gene expression level was assessed for Collagen II, Aggrecan, SOX 9, and Collagen X. The results represented that in the 1E scaffold Collagen II, Aggrecan, and SOX 9 were more upregulated at the end of the 21st day. However, in the 1E scaffold collagen X, as a hypertrophy marker, was downregulated at the end of the experiment. Overall, these results confirmed the potential of the 1E scaffold to be introduced as a promising cartilage tissue engineering scaffold for further studies.
Collapse
Affiliation(s)
- Sepideh Ghadirian
- Department of Biomaterials and Tissue Engineering, School of Advanced Technologies in Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Laleh Shariati
- Department of Biomaterials and Tissue Engineering, School of Advanced Technologies in Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Saeed Karbasi
- Department of Biomaterials and Tissue Engineering, School of Advanced Technologies in Medicine, Isfahan University of Medical Sciences, Isfahan, Iran.
| |
Collapse
|
6
|
Kolliopoulos V, Mikos AG. Decellularized extracellular matrix as a drug delivery carrier. J Control Release 2025; 382:113661. [PMID: 40139392 DOI: 10.1016/j.jconrel.2025.113661] [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: 11/28/2024] [Revised: 03/18/2025] [Accepted: 03/22/2025] [Indexed: 03/29/2025]
Abstract
Tissue engineering and regenerative medicine approaches seek to enhance biomaterial mimicry with the goal of driving cell recruitment, proliferation, and differentiation. Decellularized extracellular matrix (dECM) biomaterials have emerged as a promising platform for biomaterials development as they capture the complexity of native tissues and offer a rich environment of signals to guide cellular responses. However, the decellularization process can affect both the structure and composition of the ECM. Recent efforts have focused on leveraging dECM as drug delivery carriers for controlled release of bioactive molecules. This review highlights current strategies for incorporating therapeutic agents into dECM which include encapsulation within hydrogel formulations, direct bulk absorption of biomolecules, and affinity-based binding and conjugation. Each method offers unique advantages for modulating release profiles, which can range from rapid initial burst to prolonged, sustained release, depending on factors such as crosslinking density, degradation rate, and specific interactions of biomolecules with dECM components such as glycosaminoglycans.
Collapse
Affiliation(s)
- Vasiliki Kolliopoulos
- Department of Bioengineering, Rice University, Houston, TX 77030, United States of America
| | - Antonios G Mikos
- Department of Bioengineering, Rice University, Houston, TX 77030, United States of America.
| |
Collapse
|
7
|
Wang A, Mizejewski GJ, Zhang C. Growth inhibitory peptides: a potential novel therapeutic approach to cancer treatment. Eur J Pharmacol 2025; 996:177554. [PMID: 40147579 DOI: 10.1016/j.ejphar.2025.177554] [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: 01/02/2025] [Revised: 03/02/2025] [Accepted: 03/24/2025] [Indexed: 03/29/2025]
Abstract
Cancer remains a major global public health concern, with considerable interest in exploring biological molecules for cancer treatment and prevention. Growth inhibitory peptide (GIP), a promising new class of biological therapeutics, has drawn attention for its distinct anti-tumor properties. Derived from human alpha-fetoprotein (HAFP), this synthetic 34-amino-acid peptide has demonstrated substantial anti-tumor effects across various cancer cell lines, effectively inhibiting tumor cell proliferation, migration, and metastasis. Studies reveal that GIP mediates its effects through a range of mechanisms, including interactions with G protein-coupled receptors (GPCRs), anti-cell adhesion activities, inhibition of cell spreading and metastatic processes, morphological alterations, platelet aggregation inhibition, immune enhancement, cell membrane disruption, ion channel blockade, and cell cycle arrest. While GIP has exhibited promising anti-tumor activity in both in vitro and in vivo models, further investigation is essential to advance its development as a therapeutic drug, particularly regarding pharmacokinetics, safety profiles, storage stability, and clinical efficacy.
Collapse
Affiliation(s)
- Aixin Wang
- National Center for Clinical Laboratories, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology, Beijing, PR China; Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, PR China
| | - G J Mizejewski
- Division of Translational Medicine, Molecular Diagnostics Laboratory, Wadsworth Center, New York State Department of Health Biggs Laboratory, Empire State Plaza Albany, NY 12237, USA
| | - Chao Zhang
- National Center for Clinical Laboratories, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology, Beijing, PR China; Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, PR China.
| |
Collapse
|
8
|
Karamanos NK, Piperigkou Z, Gourdoupi C, Mangani S, Vivanco MDM. Extracellular matrix matters: matrix-based bioscaffolds in advancing translational cancer research and targeted therapy. Am J Physiol Cell Physiol 2025; 328:C1957-C1963. [PMID: 40033935 DOI: 10.1152/ajpcell.00050.2025] [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: 01/17/2025] [Revised: 02/07/2025] [Accepted: 02/24/2025] [Indexed: 03/05/2025]
Abstract
The onset, development, and progression of cancer are greatly influenced by the microenvironmental cues originating from diverse elements within the tumor niche. Extracellular matrix (ECM), the complex and dynamic macromolecular three-dimensional network (3-D), governs cell functionality and plays key roles in tumor growth and spreading. This article highlights the significance of ECM-based bioscaffolds in providing a relevant microenvironment not only for studying tumor behavior and drug efficacy but also for narrowing the gap between translational cancer research and targeted cancer treatment. The development of novel and user-friendly platforms that resemble the human tumor microenvironment in early and advanced cancer stages, may help to predict treatment response, thus facilitating the development and testing of new drugs, bridging the gap between in vitro and in vivo models. In addition, we present innovative strategies leveraging ECM bioscaffolds for personalized cancer treatment, including drug delivery systems and tissue engineering approaches. Specific case studies as well as ethical concerns related to the use of ECM bioscaffolds in research and therapy are also presented and critically discussed. By elucidating the intricate interplay between ECM and cancer biology, this article underscores the potential of ECM bioscaffolds as novel platforms for shaping future therapeutic interventions and advancing precision oncology.
Collapse
Affiliation(s)
- Nikos K Karamanos
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Patras, Greece
- Foundation for Research and Technology-Hellas (FORTH), Institute of Chemical Engineering Sciences (ICE-HT), Patras, Greece
| | - Zoi Piperigkou
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Patras, Greece
| | - Chrisavgi Gourdoupi
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Patras, Greece
| | - Sylvia Mangani
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Patras, Greece
| | - Maria dM Vivanco
- CIC bioGUNE, Basque Research and Technology Alliance (BRTA), Derio, Spain
| |
Collapse
|
9
|
Tavares DF, Mano JF, Oliveira MB. Advances in abiotic tissue-based biomaterials: A focus on decellularization and devitalization techniques. Mater Today Bio 2025; 32:101735. [PMID: 40275948 PMCID: PMC12020859 DOI: 10.1016/j.mtbio.2025.101735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Revised: 03/14/2025] [Accepted: 04/05/2025] [Indexed: 04/26/2025] Open
Abstract
This Review explores the growing and diversifying field of tissue-derived abiotic constructs for tissue engineering applications, with main focus on decellularization and devitalization techniques and principles. Acellular fractions derived from biological tissues, such as the extracellular matrix (ECM), have long been considered a valuable approach for the generation of numerous scaffolds and more complex constructs. The removal of the cellular content has been considered essential to prevent the development of adverse immunological reactions. Nevertheless, the discovery of promising features of certain cellular components has sparked interest in the use of inactivated or devitalized cellular fractions for several applications, particularly in regenerative medicine and inflammation control. Devitalization has been described for several clinical applications, but remains poorly explored in terms of in vitro constructs compared to decellularization methods currently available. In this review, we present and critically evaluate a spectrum of approaches for the decellularization of whole-organs and in vitro constructs, and the most prevalent devitalization techniques, with a discussion on their implications on scaffolds composition, structure, and potentially therapeutic properties. Processing methodologies to achieve optimal cell-based abiotic materials and approaches for their effective characterization are described and discussed. The application of these materials in healthcare, with most focus on regenerative approaches and including examples of commercially available products, is also addressed.
Collapse
Affiliation(s)
- Diana F. Tavares
- Department of Chemistry, CICECO – Aveiro Institute of Materials. University of Aveiro., Campus Universitário de Santiago, 3810-193, Aveiro, Portugal
| | - João F. Mano
- Department of Chemistry, CICECO – Aveiro Institute of Materials. University of Aveiro., Campus Universitário de Santiago, 3810-193, Aveiro, Portugal
| | - Mariana B. Oliveira
- Department of Chemistry, CICECO – Aveiro Institute of Materials. University of Aveiro., Campus Universitário de Santiago, 3810-193, Aveiro, Portugal
| |
Collapse
|
10
|
Soltanmohammadi F, Mahmoudi Gharehbaba A, Javadzadeh Y. Synergistic strategies in tissue engineering: The role of exosomes and decellularized extracellular matrix hydrogels. Biomed Pharmacother 2025; 188:118200. [PMID: 40414001 DOI: 10.1016/j.biopha.2025.118200] [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: 03/12/2025] [Revised: 05/07/2025] [Accepted: 05/21/2025] [Indexed: 05/27/2025] Open
Abstract
Tissue engineering aims to mimic the natural microenvironment of biological structures by utilizing the distinctive characteristics of extracellular matrix (ECM) scaffolds. The combination of decellularized extracellular matrix hydrogels (dECMHs) with exosomes (EXs) represents an innovative therapeutic approach for tissue regeneration. These dECMHs, sourced from diverse tissues, provide biocompatible scaffolds that conform to irregular defect geometries, thereby addressing the limitations of conventional ECM scaffolds. EXs, which are nanovesicles secreted by virtually all cells, play crucial role in cell communication and tissue regeneration. However, their short half-life presents challenges for systemic administration. The incorporation of EXs into dECMHs enables localized and prolonged release, thereby enhancing their therapeutic merits. This review thoroughly explains the techniques for decellularization, the characteristics of dECM, as well as the preparation and applications of dECMHs in tissue engineering. It also explores the synergistic effects of EX-dECMH systems on cellular activities essential for tissue repair, including proliferation, differentiation, and neovascularization. The mechanisms of EX release from dECMHs and their applications in the regeneration of skin, intervertebral disc, cartilage, and nerve tissues are elucidated, highlighting the considerable potential of this integrated strategy to improve tissue engineering techniques. Furthermore, the synergistic effect of EX-dECMH systems in tissue healing is investigated. Finally, the limitations associated with the clinical application of EX, dECM, and dECMH as well as the future prospect are included.
Collapse
Affiliation(s)
- Fatemeh Soltanmohammadi
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Pharmaceutics, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Adel Mahmoudi Gharehbaba
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Pharmaceutics, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran; Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Yousef Javadzadeh
- Department of Pharmaceutics, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran; Biotechnology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
11
|
Hu Y, Cheng L, Guo X, Zheng M, Zhang W, Wang X, Tang R, Chen Q, Guo Y, Cao Y, Wang Z, Ran H. Sono-Gas-Mediated Precise Stiffness Remodeling for Triple-Negative Breast Cancer Mechanical Immunotherapy. Biomater Res 2025; 29:0207. [PMID: 40376200 PMCID: PMC12078941 DOI: 10.34133/bmr.0207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Revised: 03/22/2025] [Accepted: 04/20/2025] [Indexed: 05/18/2025] Open
Abstract
Triple-negative breast cancer (TNBC) is a highly invasive cancer, and its poor therapeutic outcomes are often associated with the mechanical properties of the tumor microenvironment, which is characterized by altered extracellular matrix (ECM) flexibility and increased stiffness. Herein, a mechanical immunomodulator, namely, red blood cell membrane-IR780-L-arginine nanoparticles (R-I-LA NPs), was designed to precisely regulate the stiffness of the ECM for mechanical immunotherapy of TNBC. In tumor cells, the low-intensity focused ultrasound activates R-I-LA NPs to produce reactive nitrogen species, which damages tumor cells and remodels the stiffness of ECM. Meanwhile, the softened ECM can normalize the tumor vasculature to alleviate hypoxia and increase the production of reactive oxygen species, thereby enhancing the efficacy of sonodynamic therapy and stimulating immunogenic cell death. Additionally, R-I-LA NPs stimulate the immune system and suppress pulmonary metastasis. Overall, this study offers a distinctive "sono-gas-mediated mechanical immunity" strategy for ECM regulation, potentially overcoming current TNBC therapy limitations.
Collapse
Affiliation(s)
- Yaqin Hu
- Department of Ultrasound,
the Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, People’s Republic of China
- Chongqing Key Laboratory of Ultrasound Molecular Imaging and Therapy,
the Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, People’s Republic of China
| | - Long Cheng
- Department of Ultrasound,
the Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, People’s Republic of China
- Chongqing Key Laboratory of Ultrasound Molecular Imaging and Therapy,
the Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, People’s Republic of China
| | - Xun Guo
- Department of Ultrasound,
the Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, People’s Republic of China
- Chongqing Key Laboratory of Ultrasound Molecular Imaging and Therapy,
the Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, People’s Republic of China
| | - Min Zheng
- Department of Ultrasound,
the Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, People’s Republic of China
- Chongqing Key Laboratory of Ultrasound Molecular Imaging and Therapy,
the Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, People’s Republic of China
| | - Wei Zhang
- Department of Ultrasound,
the Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, People’s Republic of China
- Chongqing Key Laboratory of Ultrasound Molecular Imaging and Therapy,
the Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, People’s Republic of China
| | - Xingyue Wang
- Department of Ultrasound,
Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, Hubei Province 441021, People’s Republic of China
| | - Rui Tang
- Department of Ultrasound,
the Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, People’s Republic of China
- Chongqing Key Laboratory of Ultrasound Molecular Imaging and Therapy,
the Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, People’s Republic of China
| | - Qiaoqi Chen
- Department of Ultrasound,
the Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, People’s Republic of China
- Chongqing Key Laboratory of Ultrasound Molecular Imaging and Therapy,
the Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, People’s Republic of China
| | - Yuan Guo
- Department of Ultrasound,
the Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, People’s Republic of China
- Chongqing Key Laboratory of Ultrasound Molecular Imaging and Therapy,
the Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, People’s Republic of China
| | - Yang Cao
- Department of Ultrasound,
the Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, People’s Republic of China
- Chongqing Key Laboratory of Ultrasound Molecular Imaging and Therapy,
the Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, People’s Republic of China
| | - Zhigang Wang
- Department of Ultrasound,
the Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, People’s Republic of China
- Chongqing Key Laboratory of Ultrasound Molecular Imaging and Therapy,
the Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, People’s Republic of China
| | - Haitao Ran
- Department of Ultrasound,
the Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, People’s Republic of China
- Chongqing Key Laboratory of Ultrasound Molecular Imaging and Therapy,
the Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, People’s Republic of China
| |
Collapse
|
12
|
Larrea Murillo L, Green M, Mahon N, Saiani A, Tsigkou O. Modelling Cancer Pathophysiology: Mechanisms and Changes in the Extracellular Matrix During Cancer Initiation and Early Tumour Growth. Cancers (Basel) 2025; 17:1675. [PMID: 40427172 DOI: 10.3390/cancers17101675] [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: 04/06/2025] [Revised: 05/05/2025] [Accepted: 05/09/2025] [Indexed: 05/29/2025] Open
Abstract
Cancer initiation and early tumour growth are complex processes influenced by multiple cellular and microenvironmental factors. A critical aspect of tumour progression is the dynamic interplay between cancer cells and the extracellular matrix (ECM), which undergoes significant alterations to support malignancy. The loss of cell polarity is an early hallmark of tumour progression, disrupting normal tissue architecture and fostering cancerous transformation. Circumstantially, cancer-associated microRNAs (miRNAs) regulate key oncogenic processes, including ECM remodelling, epithelial-to-mesenchymal transition (EMT), and tumorigenic vascular development, further driving tumour growth. ECM alterations, particularly changes in stiffness and mechanotransduction signals, create a supportive niche for cancer cells, enhancing their survival, proliferation, and invasion. EMT and its subtype, epithelial-to-endothelial transition (EET), contribute to tumour plasticity, promote the generation of cancer stem cells (CSCs), and support tumour vascularisation. Furthermore, processes of vascular development like vasculogenesis and angiogenesis are critical for sustaining early tumour growth, supplying oxygen and nutrients to hypoxic malignant cells within the evolving cancerous microenvironments. This review explores key mechanisms underlying these changes in tumorigenic microenvironments, with an emphasis on their collective role for tumour initiation and early tumour growth. It will further delve into present in vitro modelling strategies developed to closely mimic early cancer pathophysiology. Understanding these processes is crucial for developing targeted therapies aimed at disrupting key cancer-promoting pathways and improving clinical outcomes.
Collapse
Affiliation(s)
- Luis Larrea Murillo
- Department of Materials, School of Natural Sciences, Faculty of Science and Engineering, The University of Manchester, Manchester M13 9PL, UK
- The Henry Royce Institute, Royce Hub Building, Manchester M13 9PL, UK
| | - Megan Green
- Department of Materials, School of Natural Sciences, Faculty of Science and Engineering, The University of Manchester, Manchester M13 9PL, UK
- The Henry Royce Institute, Royce Hub Building, Manchester M13 9PL, UK
- Manchester Institute of Biotechnology (MIB), The University of Manchester, Manchester M1 7DN, UK
| | - Niall Mahon
- Department of Materials, School of Natural Sciences, Faculty of Science and Engineering, The University of Manchester, Manchester M13 9PL, UK
- The Henry Royce Institute, Royce Hub Building, Manchester M13 9PL, UK
- Manchester Institute of Biotechnology (MIB), The University of Manchester, Manchester M1 7DN, UK
| | - Alberto Saiani
- Department of Materials, School of Natural Sciences, Faculty of Science and Engineering, The University of Manchester, Manchester M13 9PL, UK
- Manchester Institute of Biotechnology (MIB), The University of Manchester, Manchester M1 7DN, UK
| | - Olga Tsigkou
- Department of Materials, School of Natural Sciences, Faculty of Science and Engineering, The University of Manchester, Manchester M13 9PL, UK
- The Henry Royce Institute, Royce Hub Building, Manchester M13 9PL, UK
| |
Collapse
|
13
|
Pol M, Gao H, Fox JM, Jia X. TGFβ1 and RGD Cooperatively Regulate SMAD2/3-Mediated Oncogenic Effects in Prostate Cancer Cells in Bio-Orthogonally Constructed Hydrogels. ACS Biomater Sci Eng 2025; 11:3003-3018. [PMID: 40214406 DOI: 10.1021/acsbiomaterials.5c00007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/13/2025]
Abstract
To recapitulate prostate cancer metastasis, DU145 cells were cultured in a hyaluronic acid-based, bio-orthogonally constructed, protease-degradable hydrogels. In the presence of a covalently conjugated integrin-binding peptide (GRGDSP), DU145 cells formed tumoroids and exhibited small protrusions. Upon addition of soluble transforming growth factor beta 1 (TGFβ1), cells underwent morphological changes to form extended interconnected cellular networks. Contrarily, in RGD-free hydrogels, cells maintained spherical structures even in the presence of TGFβ1. In RGD-conjugated hydrogels, TGFβ1 induced nuclear localization of SMAD2/3, upregulating a wide range of TGFβ1 target genes and proteins. Prolonged exposure to TGFβ1 led to matrix remodeling and induced epithelial-to-mesenchymal transition in DU145 cells, with loss of epithelial markers and gain of mesenchymal markers. A pharmacological inhibitor of TGFβRI/ALK5, SB-431542, attenuated TGFβ1-induced morphological changes, abrogated nuclear localization of SMAD2/3, and restored the expression of key epithelial markers. Our findings highlight the cooperative role of TGFβ1 signaling and integrin-binding peptide in the acquisition of an aggressive phenotype and the promotion of tumor progression.
Collapse
Affiliation(s)
- Mugdha Pol
- Department of Biological Sciences, University of Delaware, Newark, Delaware 19716, United States
| | - Hanyuan Gao
- Department of Materials Science and Engineering, University of Delaware, Newark, Delaware 19716, United States
| | - Joseph M Fox
- Department of Materials Science and Engineering, University of Delaware, Newark, Delaware 19716, United States
- Department of Chemistry and Biochemistry, University of Delaware, Newark, Delaware 19716, United States
| | - Xinqiao Jia
- Department of Biological Sciences, University of Delaware, Newark, Delaware 19716, United States
- Department of Materials Science and Engineering, University of Delaware, Newark, Delaware 19716, United States
- Department of Biomedical Engineering, University of Delaware, Newark, Delaware 19716, United States
- Delaware Biotechnology Institute, Newark, Delaware 19713, United States
| |
Collapse
|
14
|
Park R, Chen C. 3D Hepatocyte Model with Composite Nanofibers That Reproduced Human In Vivo Drug Clearance Profiles. ACS Pharmacol Transl Sci 2025; 8:1424-1434. [PMID: 40370988 PMCID: PMC12070230 DOI: 10.1021/acsptsci.5c00149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2025] [Revised: 04/15/2025] [Accepted: 04/17/2025] [Indexed: 05/16/2025]
Abstract
This study presents a novel in vitro 3D hepatocyte model that contains a nanofibrous scaffold designed to mimic the extracellular matrix (ECM) of the human liver, both structurally and biochemically. A modular 3D-printed device housing the ECM scaffold was also developed, readily fitting in well plates. HepaRG hepatocytes cultured on the scaffold exhibited enhanced metabolic activity compared to traditional 2D cultures, indicating improved hepatocyte functionality. Drug clearance studies with lidocaine, clozapine, and fluoxetine demonstrated significantly faster clearance rates on the scaffold, closely aligning with in vivo results from the literature, while 2D cultures showed limited metabolic capacity. This model offers a physiologically relevant platform for hepatocyte studies. The findings underscore the model's potential to advance preclinical drug development by replicating liver-specific functions in vitro.
Collapse
Affiliation(s)
- Rudolph Park
- Department of Chemistry and
Biochemistry, University of Maryland Baltimore
County, Baltimore, Maryland 21250, United States
| | - Chengpeng Chen
- Department of Chemistry and
Biochemistry, University of Maryland Baltimore
County, Baltimore, Maryland 21250, United States
| |
Collapse
|
15
|
Ranat K, Phan H, Ellythy S, Kenter M, Akkouch A. Advancements in Musculoskeletal Tissue Engineering: The Role of Melt Electrowriting in 3D-Printed Scaffold Fabrication. J Funct Biomater 2025; 16:163. [PMID: 40422828 DOI: 10.3390/jfb16050163] [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: 03/18/2025] [Revised: 04/22/2025] [Accepted: 04/24/2025] [Indexed: 05/28/2025] Open
Abstract
Musculoskeletal tissue injuries of the bone, cartilage, ligaments, tendons, and skeletal muscles are among the most common injuries experienced in medicine and become increasingly problematic in cases of significant tissue damage, such as nonunion bone defects and volumetric muscle loss. Current gold standard treatment options for musculoskeletal injuries, although effective, have limited capability to fully restore native tissue structure and function. To overcome this challenge, three-dimensional (3D) printing techniques have emerged as promising therapeutic options for tissue regeneration. Melt electrowriting (MEW), a recently developed advanced 3D printing technique, has gained significant traction in the field of tissue regeneration because of its ability to fabricate complex customizable scaffolds via high-precision microfiber deposition. The tailorability at microscale levels offered by MEW allows for enhanced recapitulation of the tissue microenvironment. Here, we survey the recent contributions of MEW in advancing musculoskeletal tissue engineering. More specifically, we briefly discuss the principles and technical aspects of MEW, provide an overview of current printers on the market, review in-depth the latest biomedical applications in musculoskeletal tissue regeneration, and, lastly, examine the limitations of MEW and offer future perspectives.
Collapse
Affiliation(s)
- Kunal Ranat
- Western Michigan University Homer Stryker M.D. School of Medicine, Kalamazoo, MI 49008, USA
| | - Hong Phan
- Western Michigan University Homer Stryker M.D. School of Medicine, Kalamazoo, MI 49008, USA
| | - Suhaib Ellythy
- Western Michigan University Homer Stryker M.D. School of Medicine, Kalamazoo, MI 49008, USA
| | - Mitchell Kenter
- Department of Surgical Services, Division of Medical Engineering, Western Michigan University Homer Stryker M.D. School of Medicine, Kalamazoo, MI 49008, USA
| | - Adil Akkouch
- Department of Surgical Services, Division of Medical Engineering, Western Michigan University Homer Stryker M.D. School of Medicine, Kalamazoo, MI 49008, USA
- Department of Surgical Services, Division of Orthopaedic Surgery, Western Michigan University Homer Stryker M.D. School of Medicine, Kalamazoo, MI 49008, USA
| |
Collapse
|
16
|
Kulkarni T, Banik S, Mukhopadhyay D, Babiker H, Bhattacharya S. Tumor-Treating Fields Alter Nanomechanical Properties of Pancreatic Ductal Adenocarcinoma Cells Co-Cultured with Extracellular Matrix. J Funct Biomater 2025; 16:160. [PMID: 40422825 DOI: 10.3390/jfb16050160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2025] [Revised: 04/28/2025] [Accepted: 04/30/2025] [Indexed: 05/28/2025] Open
Abstract
Tumor-Treating Fields (TTFields), a novel therapeutic avenue, is approved for therapy in Glioblastoma multiforme, malignant pleural mesothelioma, and metastatic non-small cell lung cancer (NSCLC). In pancreatic ductal adenocarcinoma (PDAC), several clinical trials are underway to improve outcomes, yet a significant knowledge gap prevails involving the cell-extracellular matrix (ECM) crosstalk. Herein, we hypothesized that treatment with TTFields influence this crosstalk, which is reflected by the dynamic alteration in nanomechanical properties (NMPs) of cells and the ECM in a co-culture system. We employed an ECM gel comprising collagen, fibronectin, and laminin mixed in 100:1:1 stoichiometry to co-culture of Panc1 and AsPC1 individually. This ECM mixture mimics the in vivo tumor microenvironment closely when compared to the individual ECM components studied before. A comprehensive frequency-dependent study revealed the optimal TTFields frequency to be 150 kHz. We also observed that irrespective of the ECM's presence, TTFields increase cell membrane stiffness and decrease deformation several-folds in both Panc1 and AsPC1 cells at both 48 h and 72 h. Although adhesion for AsPC1 decreased at 48 h, at 72 h it was observed to increase irrespective of ECM's presence. Moreover, it significantly alters the NMPs of ECM gels when co-cultured with PDAC cell lines. However, AsPC1 cells were observed to be more detrimental to these changes. Lastly, we attribute the stiffness changes in Panc1 cells to the membrane F-actin reorganization in the presence of TTFields. This study paves a path to study complex PDAC TME as well as the effect of various chemotherapeutic agents on such TME with TTFields in the future.
Collapse
Affiliation(s)
- Tanmay Kulkarni
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine and Science, 4500 San Pablo Road South, Jacksonville, FL 32224, USA
| | - Sreya Banik
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine and Science, 4500 San Pablo Road South, Jacksonville, FL 32224, USA
| | - Debabrata Mukhopadhyay
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine and Science, 4500 San Pablo Road South, Jacksonville, FL 32224, USA
- Department of Physiology and Biomedical Engineering, Mayo Clinic College of Medicine and Science, 4500 San Pablo Road South, Jacksonville, FL 32224, USA
| | - Hani Babiker
- Department of Medicine, Division of Hematology and Oncology, Mayo Clinic College of Medicine and Science, 4500 San Pablo Road South, Jacksonville, FL 32224, USA
| | - Santanu Bhattacharya
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine and Science, 4500 San Pablo Road South, Jacksonville, FL 32224, USA
- Department of Physiology and Biomedical Engineering, Mayo Clinic College of Medicine and Science, 4500 San Pablo Road South, Jacksonville, FL 32224, USA
| |
Collapse
|
17
|
Trevizani M, Leal LL, da Silva Barros RJ, de Paoli F, Nogueira BV, Costa FF, de Aguiar JAK, da Costa Maranduba CM. Effects of decellularization on glycosaminoglycans and collagen macromolecules in bovine bone extracellular matrix. Int J Biol Macromol 2025; 307:141007. [PMID: 39971037 DOI: 10.1016/j.ijbiomac.2025.141007] [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: 10/01/2024] [Revised: 01/27/2025] [Accepted: 02/12/2025] [Indexed: 02/21/2025]
Abstract
Bovine bones were decellularized to obtain extracellular matrices with potential use for Tissue Bioengineering. The objective of the present study was to develop a decellularization protocol for bovine trabecular bone while maintaining the integrity of the extracellular matrix (ECM). The protocol proved to be effective in significantly reducing the Amount of genetic material and cellular content, and it was considered innovative, being filed as a patent. The scaffold obtained showed a reduction in the content of glycosaminoglycans (GAGs) and collagen. Even with the loss of these ECM components, the material obtained can be considered an alternative for use in Tissue Engineering and Regenerative Medicine.
Collapse
Affiliation(s)
- Marizia Trevizani
- Department of Biology, Laboratory of Human Genetics and Cell Therapy, Institute of Biological Sciences, Federal University of Juiz de Fora, Minas Gerais, Brazil
| | - Laís Lopardi Leal
- Department of Biology, Laboratory of Human Genetics and Cell Therapy, Institute of Biological Sciences, Federal University of Juiz de Fora, Minas Gerais, Brazil
| | - Rodolpho José da Silva Barros
- Carlos Alberto Redins Cellular Ultrastructure Laboratory (LUCCAR), Department of Morphology, Health Sciences Center, Federal University of Espírito Santo, Espírito Santo, Brazil
| | - Flávia de Paoli
- Department of Morphology, Institute of Biological Sciences, Federal University of Juiz de Fora, Minas Gerais, Brazil
| | - Breno Valentim Nogueira
- Carlos Alberto Redins Cellular Ultrastructure Laboratory (LUCCAR), Department of Morphology, Health Sciences Center, Federal University of Espírito Santo, Espírito Santo, Brazil
| | - Fabiano Freire Costa
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Federal University of Juiz de Fora, Minas Gerais, Brazil
| | - Jair Adriano Kopke de Aguiar
- Department of Biochemistry, Glycoconjugate Analysis Laboratory, Institute of Biological Sciences, Federal University of Juiz de Fora, Minas Gerais, Brazil.
| | - Carlos Magno da Costa Maranduba
- Department of Biology, Laboratory of Human Genetics and Cell Therapy, Institute of Biological Sciences, Federal University of Juiz de Fora, Minas Gerais, Brazil.
| |
Collapse
|
18
|
Fazeli MA, Amiri M, Rostaminasab G, Akbaripour V, Mikaeili A, Othman M, Rezakhani L. Application of decellularized tissues in ear regeneration. J Tissue Viability 2025; 34:100870. [PMID: 39970482 DOI: 10.1016/j.jtv.2025.100870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 01/15/2025] [Accepted: 02/07/2025] [Indexed: 02/21/2025]
Abstract
More than 5 % of people worldwide suffer from hearing disorders. Ototoxic drugs, aging, exposure to loud sounds, rupture, subperichondrial hematoma, perichondritis, burns and frostbite and infections are the main causes of hearing loss, some of which can destroy the cartilage and lead to deformation. On the other hand, disorders of the external ear are diverse and can range from dangerous neoplasms to defects that are not acceptable from a cosmetic standpoint. These issues include injuries, blockages, dermatoses, and infections, and any or all of them may be bothersome to the busy doctor. Using an implant or hearing aid is one of the treatment strategies for deafness. However, these medical devices are not useful for every eligible patient. With the right therapy, many of these issues are not life-threatening and can be treated with confidence in a positive outcome. As medical research and treatment have advanced dramatically in the past ten years, tissue engineering (TE) has emerged as a promising method to regenerate damaged tissue, raising the prospect of a permanent cure for deafness. Decellularization is now seen as a promising development for regenerative medicine, and an increasing number of applications are being found for acellular matrices. Studies on decellularization show that natural scaffolds made from decellularized tissues can serve as a suitable platform while preserving the main components, and the preparation of such scaffolds will be an important part of future bioscience research. It can have wide applications in regenerative medicine and TE. This review intends to give an overview of the status of research and alternative scaffolds in inner and outer ear regenerative medicine from both a preclinical and clinical perspective for ear disorders in order to show how ongoing TE research has the potential to advance and enhance novel disease treatments.
Collapse
Affiliation(s)
- Manouchehr Avatef Fazeli
- Clinical Research Development Center, Imam Khomeini and Mohammad Kermanshahi and Farabi Hospitals, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Masoumeh Amiri
- Clinical Research Development Center, Imam Khomeini and Mohammad Kermanshahi and Farabi Hospitals, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Gelavizh Rostaminasab
- Clinical Research Development Center, Imam Khomeini and Mohammad Kermanshahi and Farabi Hospitals, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Vahid Akbaripour
- Student Research Committee, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Abdolhamid Mikaeili
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Mohammad Othman
- Student Research Committee, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Leila Rezakhani
- Fertility and Infertility Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran; Department of Tissue Engineering, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| |
Collapse
|
19
|
Kmail M, Razak R, Mohd Isa IL. Engineering extracellular matrix-based hydrogels for intervertebral disc regeneration. Front Bioeng Biotechnol 2025; 13:1601154. [PMID: 40375978 PMCID: PMC12078266 DOI: 10.3389/fbioe.2025.1601154] [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: 03/27/2025] [Accepted: 04/16/2025] [Indexed: 05/18/2025] Open
Abstract
Lower back pain (LBP) is a major health concern, especially in older adults. A key aetiological factor is intervertebral disc (IVD) degeneration. It is mediated by dysregulation of extracellular matrix (ECM) and inflammation. In recent years, regenerative therapies have garnered attention for their potential to restore disc function by addressing the underlying biological alterations within the IVD. This review focuses on the comprehensive understanding of the anatomy and physiology of the IVD, highlighting its life cycle from embryonic development, and maturation to degenerative phenotype. We describe current treatments for managing LBP caused by IVD degeneration. This review emphasizes on the recent advancements in hydrogel engineering, highlighting natural, synthetic, and composite hydrogels and their application in ECM-targeted regenerative therapy for IVD degeneration. By exploring innovations in hydrogel technology, including improvements in crosslinking techniques and controlled degradation rates-we discuss how these materials could enhance IVD regeneration and potentially be used for the management of LBP. With their enhanced biomimicry, hydrogel-based ECM mimics offer a promising pathway for developing effective, durable therapies that address the root causes of disc degeneration, providing new hope for individuals living with chronic LBP.
Collapse
Affiliation(s)
- Mwafaq Kmail
- Department of Anatomy, Faculty of Medicine, Universiti Kebangsaan, Kuala Lumpur, Malaysia
| | - Rusydi Razak
- Department of Anatomy, Faculty of Medicine, Universiti Kebangsaan, Kuala Lumpur, Malaysia
| | - Isma Liza Mohd Isa
- Department of Anatomy, Faculty of Medicine, Universiti Kebangsaan, Kuala Lumpur, Malaysia
- CÚRAM Research Ireland Centre for Medical Devices, School of Medicine, University of Galway, Galway, Ireland
| |
Collapse
|
20
|
Han R, Luo L, Wei C, Qiao Y, Xie J, Pan X, Xing J. Stiffness-tunable biomaterials provide a good extracellular matrix environment for axon growth and regeneration. Neural Regen Res 2025; 20:1364-1376. [PMID: 39075897 PMCID: PMC11624885 DOI: 10.4103/nrr.nrr-d-23-01874] [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: 11/14/2023] [Revised: 01/31/2024] [Accepted: 03/16/2024] [Indexed: 07/31/2024] Open
Abstract
Neuronal growth, extension, branching, and formation of neural networks are markedly influenced by the extracellular matrix-a complex network composed of proteins and carbohydrates secreted by cells. In addition to providing physical support for cells, the extracellular matrix also conveys critical mechanical stiffness cues. During the development of the nervous system, extracellular matrix stiffness plays a central role in guiding neuronal growth, particularly in the context of axonal extension, which is crucial for the formation of neural networks. In neural tissue engineering, manipulation of biomaterial stiffness is a promising strategy to provide a permissive environment for the repair and regeneration of injured nervous tissue. Recent research has fine-tuned synthetic biomaterials to fabricate scaffolds that closely replicate the stiffness profiles observed in the nervous system. In this review, we highlight the molecular mechanisms by which extracellular matrix stiffness regulates axonal growth and regeneration. We highlight the progress made in the development of stiffness-tunable biomaterials to emulate in vivo extracellular matrix environments, with an emphasis on their application in neural repair and regeneration, along with a discussion of the current limitations and future prospects. The exploration and optimization of the stiffness-tunable biomaterials has the potential to markedly advance the development of neural tissue engineering.
Collapse
Affiliation(s)
- Ronglin Han
- Department of Pathophysiology, School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan Province, China
| | - Lanxin Luo
- Department of Pathophysiology, School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan Province, China
| | - Caiyan Wei
- Department of Medicinal Chemistry, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan Province, China
| | - Yaru Qiao
- Department of Pathophysiology, School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan Province, China
| | - Jiming Xie
- Department of Pathophysiology, School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan Province, China
| | - Xianchao Pan
- Department of Medicinal Chemistry, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan Province, China
| | - Juan Xing
- Department of Pathophysiology, School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan Province, China
| |
Collapse
|
21
|
Fiore A, Yu G, Northey JJ, Patel R, Ravenscroft TA, Ikegami R, Kolkman W, Kumar P, Dilan TL, Ruetten VMS, Ahrens MB, Shroff H, Wang S, Weaver VM, Pedram K. Live imaging of the extracellular matrix with a glycan-binding fluorophore. Nat Methods 2025; 22:1070-1080. [PMID: 39915692 PMCID: PMC12074998 DOI: 10.1038/s41592-024-02590-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Accepted: 12/18/2024] [Indexed: 02/12/2025]
Abstract
All multicellular systems produce and dynamically regulate extracellular matrices (ECMs) that play essential roles in both biochemical and mechanical signaling. Though the spatial arrangement of these extracellular assemblies is critical to their biological functions, visualization of ECM structure is challenging, in part because the biomolecules that compose the ECM are difficult to fluorescently label individually and collectively. Here, we present a cell-impermeable small-molecule fluorophore, termed Rhobo6, that turns on and red shifts upon reversible binding to glycans. Given that most ECM components are densely glycosylated, the dye enables wash-free visualization of ECM, in systems ranging from in vitro substrates to in vivo mouse mammary tumors. Relative to existing techniques, Rhobo6 provides a broad substrate profile, superior tissue penetration, non-perturbative labeling, and negligible photobleaching. This work establishes a straightforward method for imaging the distribution of ECM in live tissues and organisms, lowering barriers for investigation of extracellular biology.
Collapse
Affiliation(s)
- Antonio Fiore
- Janelia Research Campus, Howard Hughes Medical Institute (HHMI), Ashburn, VA, USA
| | - Guoqiang Yu
- Janelia Research Campus, Howard Hughes Medical Institute (HHMI), Ashburn, VA, USA
| | - Jason J Northey
- Center for Bioengineering and Tissue Regeneration, Department of Surgery, University of California, San Francisco (UCSF), San Francisco, CA, USA
| | - Ronak Patel
- Janelia Research Campus, Howard Hughes Medical Institute (HHMI), Ashburn, VA, USA
| | - Thomas A Ravenscroft
- Janelia Research Campus, Howard Hughes Medical Institute (HHMI), Ashburn, VA, USA
| | - Richard Ikegami
- Janelia Research Campus, Howard Hughes Medical Institute (HHMI), Ashburn, VA, USA
| | - Wiert Kolkman
- Janelia Research Campus, Howard Hughes Medical Institute (HHMI), Ashburn, VA, USA
| | - Pratik Kumar
- Janelia Research Campus, Howard Hughes Medical Institute (HHMI), Ashburn, VA, USA
| | - Tanya L Dilan
- Janelia Research Campus, Howard Hughes Medical Institute (HHMI), Ashburn, VA, USA
| | - Virginia M S Ruetten
- Janelia Research Campus, Howard Hughes Medical Institute (HHMI), Ashburn, VA, USA
| | - Misha B Ahrens
- Janelia Research Campus, Howard Hughes Medical Institute (HHMI), Ashburn, VA, USA
| | - Hari Shroff
- Janelia Research Campus, Howard Hughes Medical Institute (HHMI), Ashburn, VA, USA
| | - Shaohe Wang
- Janelia Research Campus, Howard Hughes Medical Institute (HHMI), Ashburn, VA, USA
| | - Valerie M Weaver
- Center for Bioengineering and Tissue Regeneration, Department of Surgery, University of California, San Francisco (UCSF), San Francisco, CA, USA
| | - Kayvon Pedram
- Janelia Research Campus, Howard Hughes Medical Institute (HHMI), Ashburn, VA, USA.
| |
Collapse
|
22
|
Fontes PK, Andrade APM, Rolniche LCM, Rocha LTDS, da Costa AM, Castilho ACDS. Ovarian superstimulation protocols modulate the morphological phenotypes in bovine oviduct. Theriogenology 2025; 238:117370. [PMID: 40037031 DOI: 10.1016/j.theriogenology.2025.117370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 02/20/2025] [Accepted: 02/25/2025] [Indexed: 03/06/2025]
Abstract
It has been shown that estradiol (E2) and progesterone (P4) have a significant influence on the alteration of morphological parameters in the oviduct of cattle. These morphological changes were demonstrated by comparing the effects of the different hormonal profiles between the hormonal phases of the estrous cycle or between the ipsi- and contralateral side of the preovulatory follicle/corpus luteum and even the size of the corpus luteum. In our previous study, we have shown that the ovarian superstimulation (OVS) protocol increases E2 levels in the bovine oviduct during the preovulatory phase. Therefore, we wanted to gain insight into the effects of OVS protocols on morphological parameters in the bovine oviduct. To this end, our study evaluated two OVS protocols, an FSH-only protocol and the FSH/eCG protocol, both of which were successfully standardized for Nelore cows (Bos taurusindicus). A third group was used as a control that was not superstimulated (synchronized estrus). The ampulla and isthmus segments of the oviduct were analyzed separately for morphometric analysis (hematoxylin and eosin staining - HE), quantification of total collagen (picrosirius red staining - PSR), analysis of fractal dimensions, and quantification of total mucin (staining with periodic acid-Schiffs/Alcian blue). Overall, both OVS protocols decreased mucosal height, epithelial area, and luminal area in the isthmus, while total collagen quantification increased. In contrast, cows subjected to the FSH/eCG protocol exhibited increased muscle layer area and mucosal height in the ampulla, while total collagen quantity decreased. Analysis of fractal dimensions showed that both OVS treatments increased cell organization in both isthmus and ampulla segments compared to the synchronized group when analyzing tissues stained with PSR. Finally, the FSH/eCG protocol increased the relative abundance of mucins in the isthmus and ampulla segments compared to the other groups. In conclusion, we have shown that cows subjected to OVS exhibit a different morphological phenotype in the bovine oviduct characterized by morphometric changes, collagen modulation, and histochemical alterations.
Collapse
Affiliation(s)
- Patricia Kubo Fontes
- Center for Natural and Human Sciences, Federal University of ABC, Santo André, São Paulo, Brazil
| | | | | | | | | | | |
Collapse
|
23
|
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
|
24
|
Chen R, Zhang R, Ke F, Guo X, Zeng F, Liu Q. Mechanisms of breast cancer metastasis: the role of extracellular matrix. Mol Cell Biochem 2025; 480:2771-2796. [PMID: 39652293 DOI: 10.1007/s11010-024-05175-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Accepted: 11/23/2024] [Indexed: 05/03/2025]
Abstract
The components of the extracellular matrix (ECM) are dynamic, and they mediate mechanical signals that modulate cellular behaviors. Disruption of the ECM can induce the migration and invasion of cancer cells via specific signaling pathways and cytokines. Metastasis is a leading cause of high mortality in malignancies, and early intervention can improve survival rates. However, breast cancer is frequently diagnosed subsequent to metastasis, resulting in poor prognosis and distant metastasis poses substantial hurdles in therapy. In breast cancer, there is notable tissue remodeling of ECM proteins, with several identified as essential components for metastasis. Moreover, specific ECM molecules, receptors, enzymes, and various signaling pathways play crucial roles in breast cancer metastasis, drug treatment, and resistance. The in-depth consideration of these elements could provide potential therapeutic targets to enhance the survival rates and quality of life for breast cancer patients. This review explores the mechanisms by which alterations in the ECM contribute to breast cancer metastasis and discusses current clinical applications targeting ECM in breast cancer treatment, offering valuable perspectives for future ECM-based therapies.
Collapse
Affiliation(s)
- Rui Chen
- School of Pharmacy, Southwest Medical University, Luzhou, 646000, China
- Laboratory of Biochemistry and Molecular Biology, School of Basic Medical Science, Southwest Medical University, Luzhou, 646000, China
| | - Ranqi Zhang
- School of Pharmacy, Southwest Medical University, Luzhou, 646000, China
- Laboratory of Biochemistry and Molecular Biology, School of Basic Medical Science, Southwest Medical University, Luzhou, 646000, China
| | - Famin Ke
- School of Pharmacy, Southwest Medical University, Luzhou, 646000, China
| | - Xiurong Guo
- School of Pharmacy, Southwest Medical University, Luzhou, 646000, China
| | - Fancai Zeng
- Laboratory of Biochemistry and Molecular Biology, School of Basic Medical Science, Southwest Medical University, Luzhou, 646000, China.
| | - Qiuyu Liu
- School of Pharmacy, Southwest Medical University, Luzhou, 646000, China.
- Laboratory of Biochemistry and Molecular Biology, School of Basic Medical Science, Southwest Medical University, Luzhou, 646000, China.
| |
Collapse
|
25
|
Tao B, Li X, Hao M, Tian T, Li Y, Li X, Yang C, Li Q, Feng Q, Zhou H, Zhao Y, Wang D, Liu W. Organoid-Guided Precision Medicine: From Bench to Bedside. MedComm (Beijing) 2025; 6:e70195. [PMID: 40321594 PMCID: PMC12046123 DOI: 10.1002/mco2.70195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 03/16/2025] [Accepted: 03/18/2025] [Indexed: 05/08/2025] Open
Abstract
Organoid technology, as an emerging field within biotechnology, has demonstrated transformative potential in advancing precision medicine. This review systematically outlines the translational trajectory of organoids from bench to bedside, emphasizing their construction methodologies, key regulatory factors, and multifaceted applications in personalized healthcare. By recapitulating physiological architectures and disease phenotypes through three-dimensional culture systems, organoids leverage natural and synthetic scaffolds, stem cell sources, and spatiotemporal cytokine regulation to model tissue-specific microenvironments. Diverse organoid types-including skin, intestinal, lung, and tumor organoids-have facilitated breakthroughs in modeling tissue development, drug efficacy and toxicity screening, disease pathogenesis studies, and patient-tailored diagnostics. For instance, patient-derived tumor organoids preserve tumor heterogeneity and genomic profiles, serving as predictive platforms for individualized chemotherapy responses. In precision medicine, organoid-guided multiomics analyses identify actionable biomarkers and resistance mechanisms, while clustered regularly interspaced short palindromic repeats-based functional screens optimize therapeutic targeting. Despite preclinical successes, challenges persist in standardization, vascularization, and ethical considerations. Future integration of artificial intelligence, microfluidics, and spatial transcriptomics will enhance organoid scalability, reproducibility, and clinical relevance. By bridging molecular insights with patient-specific therapies, organoids are poised to revolutionize precision medicine, offering dynamic platforms for drug development, regenerative strategies, and individualized treatment paradigms.
Collapse
Affiliation(s)
- Boqiang Tao
- Department of Oral and Maxillofacial SurgeryHospital of StomatologyJilin UniversityChangchunChina
| | - Xiaolan Li
- Laboratory of Allergy and Precision MedicineChengdu Institute of Respiratory Healththe Third People's Hospital of ChengduAffiliated Hospital of Southwest Jiaotong UniversityChengduChina
| | - Ming Hao
- Department of Oral and Maxillofacial SurgeryHospital of StomatologyJilin UniversityChangchunChina
| | - Tian Tian
- Laboratory Animal CenterCollege of Animal ScienceJilin UniversityChangchunChina
| | - Yuyang Li
- Department of Oral and Maxillofacial SurgeryHospital of StomatologyJilin UniversityChangchunChina
| | - Xiang Li
- Department of Oral and Maxillofacial SurgeryHospital of StomatologyJilin UniversityChangchunChina
| | - Chun Yang
- College of Basic MedicineBeihua UniversityJilinChina
| | - Qirong Li
- Laboratory Animal CenterCollege of Animal ScienceJilin UniversityChangchunChina
| | - Qiang Feng
- Laboratory Animal CenterCollege of Animal ScienceJilin UniversityChangchunChina
| | - Hengzong Zhou
- Laboratory Animal CenterCollege of Animal ScienceJilin UniversityChangchunChina
| | - Yicheng Zhao
- Laboratory Animal CenterCollege of Animal ScienceJilin UniversityChangchunChina
| | - Dongxu Wang
- Laboratory Animal CenterCollege of Animal ScienceJilin UniversityChangchunChina
- Zhichuang Gene Editing Animal Model Research CenterWenzhou Institute of TechnologyWenzhouChina
| | - Weiwei Liu
- Department of Oral and Maxillofacial SurgeryHospital of StomatologyJilin UniversityChangchunChina
| |
Collapse
|
26
|
Ding X, Liang Y, Zhou S, Wu Y, Sałata P, Mikolajczk-Martinez A, Khosrawipour V, Zhang Z. Targeting tumor extracellular matrix with nanoparticles to circumvent therapeutic resistance. J Control Release 2025; 383:113786. [PMID: 40306575 DOI: 10.1016/j.jconrel.2025.113786] [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: 03/07/2025] [Revised: 04/21/2025] [Accepted: 04/25/2025] [Indexed: 05/02/2025]
Abstract
Each stage of tumor development is intrinsically linked to the tumor microenvironment (TME), wherein the extracellular matrix (ECM) serves as a vital and abundant component in tumor tissues. The ECM is a non-cellular, three-dimensional macromolecular network scaffold that provides structural support to cells, stores bioactive molecules, and mediates signaling pathways through specific binding to cell surface receptors. Moreover, the ECM in tumor tissues plays a crucial role in impeding drug diffusion and resisting apoptosis induced by conventional anti-cancer therapies that primarily target cancer cells. Therefore, directing attentions towards the tumor ECM can facilitate the identification of novel targets and the development of new therapies. This review aims to summarize the composition, structure, remodeling, and function of tumor ECM, its association with drug resistance, and current targeting strategies, with a specific emphasis on nanoparticles (NPs).
Collapse
Affiliation(s)
- Xinyue Ding
- School of Pharmacy, Key laboratory of smart drug delivery (Ministry of Education) & National key laboratory of complex drug formulations for overcoming delivery barriers, Fudan University, Shanghai 201203, China
| | - Yiyu Liang
- School of Pharmacy, Key laboratory of smart drug delivery (Ministry of Education) & National key laboratory of complex drug formulations for overcoming delivery barriers, Fudan University, Shanghai 201203, China
| | - Siyuan Zhou
- School of Pharmacy, Key laboratory of smart drug delivery (Ministry of Education) & National key laboratory of complex drug formulations for overcoming delivery barriers, Fudan University, Shanghai 201203, China
| | - Yao Wu
- School of Pharmacy, Key laboratory of smart drug delivery (Ministry of Education) & National key laboratory of complex drug formulations for overcoming delivery barriers, Fudan University, Shanghai 201203, China
| | - Patricia Sałata
- Wrocław University of Environmental and Life Sciences, Wrocław, Poland
| | | | | | - Zhiwen Zhang
- School of Pharmacy, Key laboratory of smart drug delivery (Ministry of Education) & National key laboratory of complex drug formulations for overcoming delivery barriers, Fudan University, Shanghai 201203, China.
| |
Collapse
|
27
|
Salinas MD, Martínez CM, Roca FJ, García-Bernal D, Martínez-Morga M, Rodríguez-Madoz JR, Prósper F, Zapata AG, Moraleda JM, Martínez S, Valdor R. Chaperone-mediated autophagy sustains pericyte stemness necessary for brain tissue homeostasis. J Adv Res 2025:S2090-1232(25)00259-0. [PMID: 40286844 DOI: 10.1016/j.jare.2025.04.015] [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: 07/30/2024] [Revised: 04/11/2025] [Accepted: 04/11/2025] [Indexed: 04/29/2025] Open
Abstract
INTRODUCTION Pericytes (PCs) are mural cells exhibiting some mesenchymal stem cell (MSC) properties and contribute to tissue regeneration after injury. We have previously shown that glioblastoma cancer cells induce in PCs, a pathogenic upregulation of chaperone-mediated autophagy (CMA) which modulates immune functions and MSC-like properties to support tumor growth. OBJECTIVES The aim of the study was to interrogate the role of CMA-regulated MSC properties in PCs in the context of tissue repair during inflammation triggered by a demyelinating injury. METHODS Studies of RNA-seq were done PCs with (WT) and without (LAMP-2A KO) CMA. Cell characterization related to stemness, lineage and morphology was done in WT and KO PCs. Secretome analysis and cell differentiation assay using the supernatants from CMA-efficient and deficient PCs cultures was done in mesenchymal cells. Inflammatory response of brain cells was assessed with WT and KO PCs secretome. To corroborate in vitro results, CMA modulation in response to inflammation in PCs and tissue repair markers were measured in the lesion areas of a demyelination mouse model and correlated with the tissue reparation after intravenous PC administration. An inflammatory mediator was used to study effects on PC-CMA activity. RESULTS We found that inflammatory mediators such as IFNγ downregulate CMA in PCs, suppressing PC stemness and promoting a pro-inflammatory secretome. Restoration of PC CMA activity during inflammation maintains PC MSC properties and induces an MSC-like proteome which decreases inflammation and promotes tissue repair. We identified secreted proteins involved in regenerative and protective processes, and therefore, necessary to restore brain tissue homeostasis after inflammation induced by a demyelinating injury. CONCLUSION we show that manipulation of CMA activity in host PCs could be a useful therapeutical approach in the context of brain inflammation, which might be extended to other diseases where the pericyte has a key role in response to inflammation.
Collapse
Affiliation(s)
- María Dolores Salinas
- Unit of Autophagy, Immune Response and Tolerance in Pathologic Processes, Biomedical Research Institute of Murcia-Pascual Parrilla (IMIB), 30120 Murcia, Spain; Department of Biochemistry and Molecular Biology B, and Immunology, University of Murcia (UMU), 30120 Murcia, Spain; Cell Therapy and Hematopoietic Transplant Group, Faculty of Medicine, UMU, 30120 Murcia, Spain
| | | | - Francisco J Roca
- Department of Biochemistry and Molecular Biology B, and Immunology, University of Murcia (UMU), 30120 Murcia, Spain; Unit of Infectious Disease Pathology, Clinical Microbiology and Tropical Medicine, IMIB, 30120 Murcia, Spain
| | - David García-Bernal
- Department of Biochemistry and Molecular Biology B, and Immunology, University of Murcia (UMU), 30120 Murcia, Spain; Cell Therapy and Hematopoietic Transplant Group, Faculty of Medicine, UMU, 30120 Murcia, Spain; Virgen de la Arrixaca University Hospital, Hematopoietic Transplant Group, IMIB, 30120 Murcia, Spain
| | - Marta Martínez-Morga
- Unit of Autophagy, Immune Response and Tolerance in Pathologic Processes, Biomedical Research Institute of Murcia-Pascual Parrilla (IMIB), 30120 Murcia, Spain; Cell Therapy and Hematopoietic Transplant Group, Faculty of Medicine, UMU, 30120 Murcia, Spain
| | - Juan R Rodríguez-Madoz
- Hemato-Oncology Program, Cima Universidad de Navarra, IdiSNA, 31008 Pamplona, Navarra, Spain; Centro de investigación Biomédica en Red de Cancer (CIBERONC), Madrid, Spain
| | - Felipe Prósper
- Hemato-Oncology Program, Cima Universidad de Navarra, IdiSNA, 31008 Pamplona, Navarra, Spain; Centro de investigación Biomédica en Red de Cancer (CIBERONC), Madrid, Spain; Department of Dermatology and Cell Therapy, Clinica Universidad de Navarra (CUN), IdiSNA, 31008 Pamplona Navarra, Spain; Cancer Center Clinica Universidad de Navarra (CCUN), 31008 Pamplona, Navarra, Spain
| | - Agustín G Zapata
- Department of Cell Biology, Faculty of Biology, Complutense University, 28040 Madrid, Spain
| | - Jose María Moraleda
- Cell Therapy and Hematopoietic Transplant Group, Faculty of Medicine, UMU, 30120 Murcia, Spain; Virgen de la Arrixaca University Hospital, Hematopoietic Transplant Group, IMIB, 30120 Murcia, Spain
| | - Salvador Martínez
- Instituto de Neurociencias-Miguel Hernández University (UMH-CSIC), 03550, San Juan de Alicante, ISABIAL, CIBERSAM, Alicante, Spain
| | - Rut Valdor
- Unit of Autophagy, Immune Response and Tolerance in Pathologic Processes, Biomedical Research Institute of Murcia-Pascual Parrilla (IMIB), 30120 Murcia, Spain; Department of Biochemistry and Molecular Biology B, and Immunology, University of Murcia (UMU), 30120 Murcia, Spain; Cell Therapy and Hematopoietic Transplant Group, Faculty of Medicine, UMU, 30120 Murcia, Spain.
| |
Collapse
|
28
|
Woo SH, Lee BK, Kwak AS, Yang JH, Kim SY, Kim MS, Yoo JC. Bilayer Type I Atelocollagen Scaffolds for In Vivo Regeneration of Articular Cartilage Defects. Bioengineering (Basel) 2025; 12:446. [PMID: 40428065 DOI: 10.3390/bioengineering12050446] [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: 03/10/2025] [Revised: 04/04/2025] [Accepted: 04/18/2025] [Indexed: 05/29/2025] Open
Abstract
Articular cartilage has limited regenerative potential due to its anatomical characteristics, making complete recovery from damage challenging. Microfracture (MFx) is a widely used technique to promote cartilage healing, often enhanced with scaffolds to improve outcomes. In this study, we compared the efficacy of bilayer atelocollagen and standard collagen scaffolds combined with MFx in treating osteochondral defects in a rabbit model. Three articular cartilage defects were created in the femoral condyle of each rabbit and treated with either MFx plus a bilayer atelocollagen scaffold (test group), MFx plus a standard collagen scaffold (positive group), or MFx alone (negative group). Macroscopic and histological assessments were performed at 3, 6, and 12 weeks. By week 12, macroscopic examination showed hyaline-like cartilage restoration in the test group, while the positive group exhibited restoration with some overgrowth, and the negative group showed no restoration. Histological analysis revealed significantly better restoration in the test group than in the negative group, with comparable outcomes between the test and positive groups. These findings suggest that bilayer atelocollagen scaffold implantation following MFx is a promising treatment for articular cartilage defects and may provide a viable therapeutic option for patients with cartilage damage.
Collapse
Affiliation(s)
- Sang Hun Woo
- Research & Development Headquarters, Cellontech Co., Ltd., Seoul 04783, Republic of Korea
| | - Bo Keun Lee
- Research & Development Headquarters, Cellontech Co., Ltd., Seoul 04783, Republic of Korea
| | - Andrew S Kwak
- Research & Development Headquarters, Cellontech Co., Ltd., Seoul 04783, Republic of Korea
| | - Jin Hyo Yang
- Research & Development Headquarters, Cellontech Co., Ltd., Seoul 04783, Republic of Korea
| | - Seo Yeon Kim
- Research & Development Headquarters, Cellontech Co., Ltd., Seoul 04783, Republic of Korea
| | - Man Soo Kim
- Department of Orthopaedic Surgery, College of Medicine, Seoul St. Mary's Hospital, The Catholic University, Seoul 06591, Republic of Korea
| | - Ji Chul Yoo
- Research & Development Headquarters, Cellontech Co., Ltd., Seoul 04783, Republic of Korea
| |
Collapse
|
29
|
Otsuka MY, Essel LB, Sinha A, Nickerson G, Mejia SM, Edge A, Matthews RT, Bouyain S. Aggrecan immobilizes to perineuronal nets through hyaluronan-dependent and hyaluronan-independent binding activities. J Biol Chem 2025; 301:108525. [PMID: 40273987 DOI: 10.1016/j.jbc.2025.108525] [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: 12/10/2024] [Revised: 03/26/2025] [Accepted: 04/09/2025] [Indexed: 04/26/2025] Open
Abstract
Aggrecan (ACAN) is a large, secreted chondroitin sulfate proteoglycan that includes three globular regions named G1, G2, G3, and is decorated with multiple glycosaminoglycan attachments between its G2 and G3 domains. The N-terminal G1 region interacts with the glycosaminoglycan hyaluronan (HA), which is an essential component of the vertebrate extracellular matrix. In the central nervous system, ACAN is found in perineuronal nets (PNNs), honeycomb-like structures that localize to the surface of parvalbumin-positive neurons in specific neural circuits. PNNs regulate the plasticity of the central nervous system, and it is believed that association between ACAN and HA is a foundational event in the assembly of these reticular structures. Here, we report the cocrystal structure of the G1 region of ACAN in the absence and presence of a HA decasaccharide and analyze the importance of the HA-binding activity of ACAN for its integration into PNNs. We demonstrate that the single immunoglobulin domain and the two Link modules that comprise the G1 region form a single structural unit, and that HA is clamped inside a groove that spans the length of the tandem Link domains. Introducing point mutations in the glycosaminoglycan-binding site eliminates HA-binding activity in ACAN, but, surprisingly, only decreases the integration of ACAN into PNNs. Thus, these results suggest that ACAN can be recruited into PNNs independently of its HA-binding activity.
Collapse
Affiliation(s)
- Matthew Y Otsuka
- Division of Biological and Biomedical Systems, School of Science and Engineering, University of Missouri-Kansas City, Kansas City, Missouri, USA
| | - Leslie B Essel
- Division of Biological and Biomedical Systems, School of Science and Engineering, University of Missouri-Kansas City, Kansas City, Missouri, USA
| | - Ashis Sinha
- Department of Neuroscience and Physiology, State University of New York Upstate Medical University, Syracuse, New York, USA
| | - Gabrielle Nickerson
- Department of Neuroscience and Physiology, State University of New York Upstate Medical University, Syracuse, New York, USA
| | - Seth M Mejia
- Department of Neuroscience and Physiology, State University of New York Upstate Medical University, Syracuse, New York, USA
| | - Ashley Edge
- Division of Biological and Biomedical Systems, School of Science and Engineering, University of Missouri-Kansas City, Kansas City, Missouri, USA
| | - Russell T Matthews
- Department of Neuroscience and Physiology, State University of New York Upstate Medical University, Syracuse, New York, USA.
| | - Samuel Bouyain
- Division of Biological and Biomedical Systems, School of Science and Engineering, University of Missouri-Kansas City, Kansas City, Missouri, USA.
| |
Collapse
|
30
|
Fu X, Guo Y, Zhang K, Cheng Z, Liu C, Ren Y, Miao L, Liu W, Jiang S, Zhou C, Su Y, Yang L. Prognostic impact of extracellular volume fraction derived from equilibrium contrast-enhanced CT in HCC patients receiving immune checkpoint inhibitors. Sci Rep 2025; 15:13643. [PMID: 40254627 PMCID: PMC12009984 DOI: 10.1038/s41598-025-97677-x] [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: 01/06/2025] [Accepted: 04/07/2025] [Indexed: 04/22/2025] Open
Abstract
This study aimed to investigate whether extracellular volume (ECV) fraction derived from equilibration contrast-enhanced computed tomography (CECT) affects prognosis in HCC patients receiving ICIs. This retrospective study ultimately included 211 HCC patients undergoing ICIs, of whom 60 were included in an internal validation to assess the reproducibility of the results. Baseline unenhanced and equilibrated CECT were used to measure CT values of the tumor, liver and aorta, which were combined with hematocrit to calculate the ECV fraction. Correlation analysis was used to investigate the association between tumor ECV and liver ECV fractions. The effects of clinical variables and ECV fraction on progression-free survival (PFS) and overall survival (OS) were evaluated using Cox proportional hazards models and Kaplan-Meier curves. Of these 151 patients, tumor ECV fraction positively correlated with liver ECV fraction. In the Lower tumor ECV group, PFS (5.6 vs. 7.6 months) and OS (10.5 vs. 15.5 months) were notably shorter than in the Higher tumor ECV group, while no significant differences were found between the Higher and Lower liver ECV groups. Furthermore, the multivariable Cox regression model demonstrated that higher tumor ECV fraction level was an independent protective factor for PFS and OS (all P < 0.001). Internal validation cohort preliminary demonstrated reproducibility of results. The tumor ECV fraction is expected to become a routine indicator before ICIs therapy for HCC patients in contrast to liver ECV fraction, contributing to their subsequent management.
Collapse
Affiliation(s)
- Xiaona Fu
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Yusheng Guo
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Kailu Zhang
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Zhixuan Cheng
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Chanyuan Liu
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Yi Ren
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Lianwei Miao
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Weiwei Liu
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Shanshan Jiang
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Chen Zhou
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Hubei Key Laboratory of Molecular Imaging, Wuhan, 430022, China.
| | - Yangbo Su
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Hubei Key Laboratory of Molecular Imaging, Wuhan, 430022, China.
| | - Lian Yang
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Hubei Key Laboratory of Molecular Imaging, Wuhan, 430022, China.
| |
Collapse
|
31
|
Sun Y, Zhang Z, Wang Y, Wu X, Sun Y, Lou H, Xu J, Yao J, Cong D. Hidden pathway: the role of extracellular matrix in type 2 diabetes mellitus-related sarcopenia. Front Endocrinol (Lausanne) 2025; 16:1560396. [PMID: 40309438 PMCID: PMC12040695 DOI: 10.3389/fendo.2025.1560396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Accepted: 03/27/2025] [Indexed: 05/02/2025] Open
Abstract
Type 2 diabetes mellitus-related sarcopenia (T2DMRS) is a common complication in elderly and advanced diabetes patients that affects long-term prognosis and quality of life. Skeletal muscle is the main unit of glucose metabolism, and it is surrounded by extracellular matrix (ECM), which is a microenvironment that acts as an efficient highway system. The ECM is essential for cellular communication and nutrient transport and supports muscle cell growth and repair. When this "ECM highway" fails to function effectively because of damage or blockage, the development of T2DMRS can be triggered or exacerbated. In recent years, the ECM has been widely demonstrated to play a critical role in insulin resistance and skeletal muscle regeneration. However, how the remodeling of skeletal muscle ECM components specifically affects the T2DMRS mechanism of action has not been scientifically described in detail. In this review, we comprehensively summarize the T2DMRS-related mechanisms of ECM remodeling, suggesting that collagen and integrins may be potential therapeutic targets.
Collapse
Affiliation(s)
- Yiping Sun
- School of Acupuncture and Tuina, Changchun University of Chinese Medicine, Changchun, China
| | - Zepeng Zhang
- Research Center of Traditional Chinese Medicine, Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, China
| | - Yufeng Wang
- Department of Science and Technology, Changchun University of Chinese Medicine, Changchun, China
| | - Xingquan Wu
- Department of Tuina, Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, China
| | - Yahui Sun
- Department of Tuina, Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, China
| | - Huijuan Lou
- Department of Tuina, Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, China
| | - Jing Xu
- School of Acupuncture and Tuina, Changchun University of Chinese Medicine, Changchun, China
| | - Junjie Yao
- School of Acupuncture and Tuina, Changchun University of Chinese Medicine, Changchun, China
| | - Deyu Cong
- Department of Tuina, Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, China
| |
Collapse
|
32
|
Bonomi F, Limido E, Weinzierl A, Bickelmann C, Ampofo E, Harder Y, Menger MD, Laschke MW. Heat Preconditioning of Nanofat Does Not Improve Its Vascularization Properties. Cells 2025; 14:581. [PMID: 40277906 PMCID: PMC12025451 DOI: 10.3390/cells14080581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2025] [Revised: 04/07/2025] [Accepted: 04/10/2025] [Indexed: 04/26/2025] Open
Abstract
Heat preconditioning has been shown to promote nutritive perfusion and tissue survival in autologous fat grafting as well as in flap and breast surgery. However, its impact on the vascularization properties of nanofat has not been investigated so far. Therefore, we exposed nanofat from donor mice to a temperature of 43 °C for 1 h and assessed the effects of this heat stress on cell viability and the expression of heat shock proteins (HSPs) and angiogenesis-related factors. Moreover, dermal substitutes seeded with heat-preconditioned and non-preconditioned control nanofat were implanted into dorsal skinfold chambers of recipient mice to study their vascularization and tissue integration in vivo by means of repeated intravital fluorescence microscopy, histology and immunohistochemistry. Heat preconditioning upregulated the expression of HSPs in nanofat without affecting cell viability. Moreover, it resulted in the downregulation of many pro-angiogenic factors and the increased expression of anti-angiogenic factors, indicating a shift towards an anti-angiogenic phenotype. Accordingly, implanted dermal substitutes seeded with heat-preconditioned nanofat exhibited a reduced vascularization and were not better integrated into the host tissue when compared to controls. These findings indicate that heat preconditioning cannot be recommended for enhancing the vascularization capacity of nanofat.
Collapse
Affiliation(s)
- Francesca Bonomi
- Institute for Clinical and Experimental Surgery, Saarland University, 66421 Homburg, Germany; (F.B.); (E.L.); (A.W.); (C.B.); (E.A.); (M.D.M.)
| | - Ettore Limido
- Institute for Clinical and Experimental Surgery, Saarland University, 66421 Homburg, Germany; (F.B.); (E.L.); (A.W.); (C.B.); (E.A.); (M.D.M.)
| | - Andrea Weinzierl
- Institute for Clinical and Experimental Surgery, Saarland University, 66421 Homburg, Germany; (F.B.); (E.L.); (A.W.); (C.B.); (E.A.); (M.D.M.)
- Department of Plastic Surgery and Hand Surgery, University Hospital Zurich, 8006 Zurich, Switzerland
| | - Caroline Bickelmann
- Institute for Clinical and Experimental Surgery, Saarland University, 66421 Homburg, Germany; (F.B.); (E.L.); (A.W.); (C.B.); (E.A.); (M.D.M.)
| | - Emmanuel Ampofo
- Institute for Clinical and Experimental Surgery, Saarland University, 66421 Homburg, Germany; (F.B.); (E.L.); (A.W.); (C.B.); (E.A.); (M.D.M.)
| | - Yves Harder
- Department of Plastic, Reconstructive and Aesthetic Surgery and Hand Surgery, Centre Hospitalier Universitaire Vaudois (CHUV), 1005 Lausanne, Switzerland;
- Faculty of Biology and Medicine, University of Lausanne (UNIL), 1005 Lausanne, Switzerland
| | - Michael D. Menger
- Institute for Clinical and Experimental Surgery, Saarland University, 66421 Homburg, Germany; (F.B.); (E.L.); (A.W.); (C.B.); (E.A.); (M.D.M.)
| | - Matthias W. Laschke
- Institute for Clinical and Experimental Surgery, Saarland University, 66421 Homburg, Germany; (F.B.); (E.L.); (A.W.); (C.B.); (E.A.); (M.D.M.)
| |
Collapse
|
33
|
Ioannou P, Katsoulieris E, Afratis NA. Matrix Dynamics and Microbiome Crosstalk: Matrix Metalloproteinases as Key Players in Disease and Therapy. Int J Mol Sci 2025; 26:3621. [PMID: 40332093 PMCID: PMC12027064 DOI: 10.3390/ijms26083621] [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: 02/28/2025] [Revised: 04/02/2025] [Accepted: 04/09/2025] [Indexed: 05/08/2025] Open
Abstract
Matrix metalloproteinases (MMPs) are key enzymes involved in extracellular matrix (ECM) remodeling, regulating a wide range of cellular and immune processes in both homeostatic and pathological conditions. Host-microbiota interactions play a critical role in maintaining ECM balance; however, during dysbiosis, this regulation is disrupted, leading to compromised barrier integrity, pathogen translocation into circulation, and the development of systemic diseases and cancer. This review highlights the bidirectional relationship between MMP expression/activity and microbiota dysbiosis, emphasizing tissue-specific alterations in MMP activity that contribute to disease progression. In addition, it integrates interdisciplinary evidence to illustrate the MMP-dependent mechanisms underlying various pathologies associated with oral and gut microbiome dysbiosis, including long-range effects through the gut-skin and gut-brain axes. Thus, this review introduces the emerging field of MatrixBiome, which explores the complex interactions between the ECM, microbiota, and host tissues. Finally, it also outlines therapeutic strategies to modulate MMP levels, either indirectly through microbiome-targeted approaches (e.g., prebiotics, probiotics, and postbiotics) or directly using MMP inhibitors, offering promising avenues for future clinical interventions.
Collapse
Affiliation(s)
- Paraskevi Ioannou
- Laboratory of Biotechnology and Molecular Analysis, Department of Agricultural Development, Agri-Food & Management of Natural Resources, National and Kapodistrian University of Athens, Evripos Campus, 34400 Psachna, Evia, Greece (E.K.)
| | - Elias Katsoulieris
- Laboratory of Biotechnology and Molecular Analysis, Department of Agricultural Development, Agri-Food & Management of Natural Resources, National and Kapodistrian University of Athens, Evripos Campus, 34400 Psachna, Evia, Greece (E.K.)
| | - Nikolaos A. Afratis
- Laboratory of Biotechnology and Molecular Analysis, Department of Agricultural Development, Agri-Food & Management of Natural Resources, National and Kapodistrian University of Athens, Evripos Campus, 34400 Psachna, Evia, Greece (E.K.)
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, 234 Herzl Street, Rehovot 7610001, Israel
| |
Collapse
|
34
|
Napolitano F, Fabozzi M, Montuori N. Non-Integrin Laminin Receptors: Shedding New Light and Clarity on Their Involvement in Human Diseases. Int J Mol Sci 2025; 26:3546. [PMID: 40332051 PMCID: PMC12026610 DOI: 10.3390/ijms26083546] [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: 03/14/2025] [Revised: 04/07/2025] [Accepted: 04/09/2025] [Indexed: 05/08/2025] Open
Abstract
The extracellular matrix (ECM) is a dynamic network of macromolecules that provides structural support for cells and orchestrates cell signaling, functions, and morphology. The basement membrane constitutes a peculiar sheet-like type of ECM located between epithelial tissues and underlying connective tissues. The major constituent of the basement membrane is laminin, which exerts a remarkable repertoire of biological functions such as cell differentiation, migration, adhesion, and wound healing. Laminin performs its functions by interacting with two main classes of receptors, the integrin and the non-integrin laminin receptors, creating a complex network essential for tissue integrity and regeneration. Dysfunctional actions of laminin are the cause of diverse human diseases, including cancer, infectious, and neurodegenerative diseases. This topic has attracted researchers for some time, but the diversity of cell-surface receptors, through which laminin signaling occurs, makes the role of laminin controversial. Moreover, different laminin isoforms were identified, and each specific tissue basement membrane differs from the others in their laminin composition. This review focuses on the structural and molecular basis and pathophysiological relevance of specific interactions between laminins and non-integrin receptors in development, health, and disease.
Collapse
Affiliation(s)
- Filomena Napolitano
- Department of Translational Medical Sciences, University of Naples Federico II, 80135 Naples, Italy; (F.N.); (M.F.)
| | - Maria Fabozzi
- Department of Translational Medical Sciences, University of Naples Federico II, 80135 Naples, Italy; (F.N.); (M.F.)
| | - Nunzia Montuori
- Department of Translational Medical Sciences, University of Naples Federico II, 80135 Naples, Italy; (F.N.); (M.F.)
- Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, 80135 Naples, Italy
| |
Collapse
|
35
|
Ma X, Ma Y, Lin Z, Ji M. The role of the TGF-β1 signaling pathway in the process of amelogenesis. Front Physiol 2025; 16:1586769. [PMID: 40271211 PMCID: PMC12014465 DOI: 10.3389/fphys.2025.1586769] [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: 03/03/2025] [Accepted: 03/31/2025] [Indexed: 04/25/2025] Open
Abstract
Amelogenesis is a highly regulated process involving multiple signaling pathways, among which the transforming growth factor-β1 (TGF-β1) signaling pathway plays a pivotal role in enamel formation. This review firstly elucidates the critical functions of TGF-β1 in regulating ameloblast behavior and enamel development, encompassing ameloblast proliferation, differentiation, apoptosis, enamel matrix protein synthesis, and mineralization. Secondly, based on emerging evidence, we further discuss potential interactions between TGF-β signaling and circadian regulation in enamel formation, although this relationship requires further experimental validation. Finally, future research directions are proposed to further investigate the relationship between TGF-β1 and the circadian clock in the context of amelogenesis.
Collapse
Affiliation(s)
- Xiaoxue Ma
- Department of Stomatology Shandong Provincial Hospital Affiliated to Shandong First Medical University, Shandong First Medical University, Jinan, Shandong, China
| | - Yunjing Ma
- Department of Stomatology Shandong Provincial Hospital Affiliated to Shandong First Medical University, Shandong First Medical University, Jinan, Shandong, China
| | - Zhiyong Lin
- Department of Stomatology Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Mei Ji
- Department of Stomatology Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| |
Collapse
|
36
|
Chylińska N, Maciejczyk M. Hyaluronic Acid and Skin: Its Role in Aging and Wound-Healing Processes. Gels 2025; 11:281. [PMID: 40277717 PMCID: PMC12026949 DOI: 10.3390/gels11040281] [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: 03/15/2025] [Revised: 04/06/2025] [Accepted: 04/07/2025] [Indexed: 04/26/2025] Open
Abstract
Hyaluronic acid (HA) is a linear, unbranched polysaccharide classified as a glycosaminoglycan. While HA is found in various tissues throughout the body, over half of its total proportion is found in the skin. The role of HA in the skin is complex and multifaceted. HA maintains proper hydration, elasticity, and skin firmness, serving as a key extracellular matrix (ECM) component. With age, HA production gradually decreases, leading to reduced water-binding capacity, drier and less elastic skin, and the formation of wrinkles. Additionally, HA plays an active role in the wound-healing process at every stage. This review summarizes the current background knowledge about the role of HA in skin aging and wound healing. We discuss the latest applications of HA in aging prevention, including anti-aging formulations, nutricosmetics, microneedles, nanoparticles, HA-based fillers, and skin biostimulators. Furthermore, we explore various HA-based dressings used in wound treatment, such as hydrogels, sponges, membranes, and films.
Collapse
Affiliation(s)
- Natalia Chylińska
- Independent Laboratory of Cosmetology, Medical University of Białystok, Akademicka 3, 15-267 Bialystok, Poland
| | - Mateusz Maciejczyk
- Department of Hygiene, Epidemiology and Ergonomics, Medical University of Białystok, Mickiewicza 2c, 15-022 Bialystok, Poland;
| |
Collapse
|
37
|
Fu Y, Gao C, Zhang H, Liu J, Li B, Chen W, Chen X, Lin X, Fang L, Wang Z. Fish Swim Bladder-Derived ECM Hydrogels Effectively Treat Myocardial Ischemic Injury through Immunomodulation and Angiogenesis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e2500036. [PMID: 40200862 DOI: 10.1002/advs.202500036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/01/2025] [Revised: 02/18/2025] [Indexed: 04/10/2025]
Abstract
Injectable hydrogel implants represent a promising therapeutic approach for ischemic heart failure; but their efficacy is often limited by low bioactivity, poor durability, and inadequate injection techniques. Herein, a unique hydrogel incorporating extracellular matrix from fish swim bladder (FSB-ECM), which has distinct advantages over mammalian derived ECM, such as low antigenicity, bioactivity, and source safety, is developed. It consists of collagen, glycoproteins, and proteoglycans, including 13 proteins common in the myocardial matrix and three specific proteins: HSPG, Col12a1, and vWF. This hydrogel enhances cardiac cell adhesion and stretching while promoting angiogenesis and M2 macrophage polarization. In addition, its storage modulus (G') increases over time, reaching about 1000 Pa after 5 min, which facilitates transcatheter delivery and in situ gelling. Furthermore, this hydrogel provides sustained support for cardiac contractions, exhibiting superior longevity. In a rat model of ischemic heart failure, the ejection fraction significantly improves with FSB-ECM treatment, accompanied by increased angiogenesis, reduced inflammation, and decreased infarct size. Finally, RNA sequencing combined with in vitro assays identifies ANGPTL4 as a key protein involved in mediating the effects of FSB-ECM treatment. Overall, this new injectable hydrogel based on FSB-ECM is suitable for transcatheter delivery and possesses remarkable reparative capabilities for treating heart failure.
Collapse
Affiliation(s)
- Yulong Fu
- Institute of Transplant Medicine, School of Medicine, Nankai University, Tianjin, 300071, China
- Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, 300192, China
| | - Canran Gao
- Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS&PUMC), Beijing, 100005, China
| | - Hailing Zhang
- Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, 300192, China
| | - Jing Liu
- Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, 300192, China
| | - Boxuan Li
- Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, 300192, China
| | - Wei Chen
- Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS&PUMC), Beijing, 100005, China
| | - Xiuping Chen
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, 999078, China
| | - Xue Lin
- Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS&PUMC), Beijing, 100005, China
| | - Ligang Fang
- Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS&PUMC), Beijing, 100005, China
| | - Zhihong Wang
- Institute of Transplant Medicine, School of Medicine, Nankai University, Tianjin, 300071, China
| |
Collapse
|
38
|
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
|
39
|
Ribes Martinez E, Franko Y, Franko R, Ferronato GA, Viana AES, Windenbach E, Stoeckl JB, Fröhlich T, Ferraz MAMM. Developing and characterising bovine decellularized extracellular matrix hydrogels to biofabricate female reproductive tissues. Acta Biomater 2025; 196:152-170. [PMID: 40058619 DOI: 10.1016/j.actbio.2025.03.009] [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: 09/02/2024] [Revised: 02/16/2025] [Accepted: 03/05/2025] [Indexed: 03/15/2025]
Abstract
This study investigated the development and characterization of decellularized extracellular matrix (dECM) hydrogels tailored for the biofabrication of female reproductive tissues, specifically targeting ovarian cortex, endometrium, ovarian medulla, and oviduct tissues. We aimed to evaluate the cytocompatibility, biomechanical properties, and overall efficacy of these dECMs in promoting cell viability, proliferation, and morphology using the bovine model. Bovine species provide a valuable model due to their accessibility from slaughterhouse tissues, offering a practical alternative to human samples, which are often limited in availability. Additionally, bovine tissue closely mirrors certain physiological and biological characteristics of humans, making it a relevant model for translational research. Our findings revealed that these dECMs exhibited high biocompatibility with embryo development and cell viability, supporting micro vascularization and cellular morphology without the need for external growth factors. It is important to note that the addition of alginate was crucial for maintaining the structural integrity of the hydrogel during long-term cultures. These hydrogels displayed biomechanical properties that closely mimicked native tissues, which was vital for maintaining their functional integrity and supporting cellular activities. The printability assessments showed that dECMs, particularly those from cortex tissues, achieved high precision in replicating the intended structures, though challenges such as low porosity remained. The bioprinted constructs demonstrated robust cell growth, with over 97% viability observed by day 7, indicating their suitability for cell culture. This work represented a significant advancement in reproductive tissue biofabrication, demonstrating the potential of dECM-based hydrogels in creating structurally and viable tissue constructs. By tailoring each dECM to match the unique biomechanical properties of different tissues, we paved the way for more effective and reliable applications in reproductive medicine and tissue engineering. STATEMENT OF SIGNIFICANCE: This research explores the use of decellularized extracellular matrix (dECM) hydrogels as bio-inks for creating reproductive tissues. Ovarian cortex and medulla, oviduct and endometrium dECMs demonstrated biomechanical properties that mimicked native tissues, which is essential for maintaining functional integrity and supporting cellular processes. Notably, these hydrogels exhibited high biocompatibility with embryo development and cell viability, promoting microvascularization and cell differentiation without the need for supplemental growth factors. The successful bioprinting of these bio-inks underscores their potential for creating more complex models. This work represents a significant advancement in tissue engineering, offering promising new avenues for reproductive medicine.
Collapse
Affiliation(s)
- E Ribes Martinez
- Clinic of Ruminants, Faculty of Veterinary Medicine, Ludwig-Maximilians-Universität München, Sonnenstr. 16, Oberschleißheim, 85764, Germany; Gene Center, Ludwig-Maximilians-Universität München, Feodor-Lynen Str. 25, Munich, 81377, Germany
| | - Y Franko
- Clinic of Ruminants, Faculty of Veterinary Medicine, Ludwig-Maximilians-Universität München, Sonnenstr. 16, Oberschleißheim, 85764, Germany; Gene Center, Ludwig-Maximilians-Universität München, Feodor-Lynen Str. 25, Munich, 81377, Germany
| | - R Franko
- Clinic of Ruminants, Faculty of Veterinary Medicine, Ludwig-Maximilians-Universität München, Sonnenstr. 16, Oberschleißheim, 85764, Germany; Gene Center, Ludwig-Maximilians-Universität München, Feodor-Lynen Str. 25, Munich, 81377, Germany
| | - G A Ferronato
- Clinic of Ruminants, Faculty of Veterinary Medicine, Ludwig-Maximilians-Universität München, Sonnenstr. 16, Oberschleißheim, 85764, Germany; Gene Center, Ludwig-Maximilians-Universität München, Feodor-Lynen Str. 25, Munich, 81377, Germany
| | - A E S Viana
- Department of Veterinary Medicine, Faculty of Zootechnic and Food Engineering, University of São Paulo, Duque de Caxias Norte, 225, Jardim Elite, Pirassununga, São Paulo, 13635-900, Brazil
| | - E Windenbach
- Laboratory for Functional Genome Analysis, Gene Center, Ludwig-Maximilians-Universität München, Feodor-Lynen Str. 25, Munich, 81377, Germany
| | - J B Stoeckl
- Laboratory for Functional Genome Analysis, Gene Center, Ludwig-Maximilians-Universität München, Feodor-Lynen Str. 25, Munich, 81377, Germany
| | - T Fröhlich
- Laboratory for Functional Genome Analysis, Gene Center, Ludwig-Maximilians-Universität München, Feodor-Lynen Str. 25, Munich, 81377, Germany
| | - M A M M Ferraz
- Clinic of Ruminants, Faculty of Veterinary Medicine, Ludwig-Maximilians-Universität München, Sonnenstr. 16, Oberschleißheim, 85764, Germany; Gene Center, Ludwig-Maximilians-Universität München, Feodor-Lynen Str. 25, Munich, 81377, Germany.
| |
Collapse
|
40
|
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
|
41
|
Chaulagain RP, Padder AM, Shrestha H, Gupta R, Bhandari R, Shrestha Y, Qasem Moqbel A, Gautam S, Lal N, Jin S. Deciphering the Matrisome: Extracellular Matrix Remodeling in Liver Cirrhosis and Hepatocellular Carcinoma. Cureus 2025; 17:e82171. [PMID: 40370880 PMCID: PMC12076258 DOI: 10.7759/cureus.82171] [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] [Accepted: 04/13/2025] [Indexed: 05/16/2025] Open
Abstract
Liver cirrhosis and hepatocellular carcinoma (HCC) are major public health concerns due to their high morbidity and mortality rates. The liver, a vital organ for metabolism, detoxification, and homeostasis, depends on the matrisome, a complex and dynamic network of extracellular matrix (ECM) components for maintaining structural and functional integrity. Chronic liver inflammation, induced by factors such as alcohol abuse, viral hepatitis, and non-alcoholic fatty liver disease, leads to fibrosis and cirrhosis, progressing to HCC. The matrisome, composed of ECM proteins including collagen, fibronectin, and laminin, plays a critical role in regulating tissue homeostasis, cell signaling, and tissue repair. Dysregulation of ECM components contributes to the pathogenesis of both liver cirrhosis and cancer. In cirrhosis, matrisome alterations are characterized by excessive ECM deposition and fibrosis, which disrupt the liver's architecture and impair its function. Activated hepatic stellate cells (HSCs) are the principal mediators of fibrosis, producing large quantities of ECM components. In liver cancer, matrisome remodeling facilitates tumorigenesis by promoting cancer cell proliferation, invasion, and metastasis. The tumor microenvironment, shaped by ECM alterations, further supports tumor growth and dissemination. Matrix metalloproteinases (MMPs) play a pivotal role in ECM degradation, fibrosis progression, and tumor invasion, while tissue inhibitors of metalloproteinases (TIMPs) modulate MMP activity. A comprehensive understanding of the molecular mechanisms that link matrisome alterations with the progression from cirrhosis to liver cancer is essential for identifying novel diagnostic and therapeutic targets. This review highlights the dynamic responses of the hepatic matrisome to both acute and chronic insults, emphasizing the complex interplay between ECM components, cellular behavior, and disease progression. Elucidating these interactions may inform strategies aimed at improving clinical outcomes for patients with liver cirrhosis and HCC.
Collapse
Affiliation(s)
- Ram Prasad Chaulagain
- Internal Medicine, Second Affiliated Hospital of Harbin Medical University, Harbin, CHN
| | - Aadil Mushtaq Padder
- Gastroenterology and Hepatology, Second Affiliated Hospital of Harbin Medical University, Harbin, CHN
| | | | - Radheshyam Gupta
- Urology Surgery, Cancer Hospital, Harbin Medical University, Harbin, CHN
| | - Rameshor Bhandari
- Surgical Gastroenterology, Grande International Hospital, Kathmandu, NPL
| | - Yelona Shrestha
- Dermatology, First Affiliated Hospital of Xinjiang Medical University, Xinxiang, CHN
| | | | - Smriti Gautam
- Dermatology, Kathmandu Medical College, Kathmandu, NPL
| | - Nand Lal
- Physiology, School of Biomedical Sciences, Harbin Medical University, Harbin, CHN
| | - Shizhu Jin
- Gastroenterology and Hepatology, Second Affiliated Hospital of Harbin Medical University, Harbin, CHN
| |
Collapse
|
42
|
Mangani S, Piperigkou Z, Koletsis NE, Ioannou P, Karamanos NK. Estrogen receptors and extracellular matrix: the critical interplay in cancer development and progression. FEBS J 2025; 292:1558-1572. [PMID: 39285617 PMCID: PMC11970714 DOI: 10.1111/febs.17270] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 07/26/2024] [Accepted: 09/02/2024] [Indexed: 04/05/2025]
Abstract
Cancer remains a significant global health concern. Breast cancer is a multifaceted and prevalent disease influenced by several factors, among which estrogen receptors (ERs) and the extracellular matrix (ECM) play pivotal roles. ERs, encompassing ERα and ERβ, exert significant diversity on tumor behavior, cell signaling, invasion, and metastatic potential, thus guiding breast cancer prognosis. Understanding the multifunctional connections between ERs and ECM that mediate the dynamics of tumor microenvironment is vital for unraveling the complexity of breast cancer pathobiology and identifying novel therapeutic targets. This critical review delves into the intricate nature of ERs, emphasizing their structural isoforms and the consequential impact on breast cancer outcomes. A detailed examination of ER-mediated cell signaling pathways reveals how differential expression of ERα and ERβ isoforms influence breast cancer cell behavior. The functional ERs-matrix interactions emerge as a pivotal factor in modulating epigenetic mechanisms of breast cancer cells, orchestrating changes in cellular phenotype and expression patterns of matrix modulators. Specifically, ERα isoforms are shown to regulate ECM signaling cascades, while the effects of ECM components on ERα activity highlight a bidirectional regulatory axis. The diversity of ERβ isoforms is also highlighted, illustrating their distinct contribution to ECM-mediated cellular responses. This review underscores the complex interplay between ERα/β isoforms and the ECM, shedding light onto the potential therapeutic strategies targeting these interactions to improve breast cancer management.
Collapse
Affiliation(s)
- Sylvia Mangani
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of ChemistryUniversity of PatrasGreece
| | - Zoi Piperigkou
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of ChemistryUniversity of PatrasGreece
| | - Nikolaos E. Koletsis
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of ChemistryUniversity of PatrasGreece
| | - Paraskevi Ioannou
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of ChemistryUniversity of PatrasGreece
| | - Nikos K. Karamanos
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of ChemistryUniversity of PatrasGreece
| |
Collapse
|
43
|
Tao Z, Zhang T, Ge Y, Li L, Ma C, Wang Z, Chen T, Zhang H, Li R, Jiang T, Ren Y. M2 macrophages regulate nucleus pulposus cell extracellular matrix synthesis through the OPN-CD44 axis in intervertebral disc degeneration. Osteoarthritis Cartilage 2025; 33:447-460. [PMID: 39842659 DOI: 10.1016/j.joca.2024.12.007] [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: 08/21/2024] [Revised: 12/15/2024] [Accepted: 12/26/2024] [Indexed: 01/24/2025]
Abstract
OBJECTIVE Macrophages play a crucial role in various physiological processes. In intervertebral disc degeneration (IDD), macrophage infiltration has been observed in human intervertebral disc (IVD) specimens, but how macrophages influence IDD remains unclear. METHODS According to the single-cell transcriptome expression profiles from GSE165722, we verified the infiltration of macrophages in IDD and the possible interaction between infiltrated macrophages and nucleus pulposus cells (NPCs). The expression of macrophage-associated markers was verified in specimens of human nucleus pulposus, lumbar spinal instability mice and annulus fibrosus puncture mice. By treating NPCs cocultured with M2 macrophages with osteopontin (OPN) neutralization antibody and siCD44, we demonstrated that both in vitro and in vivo macrophages regulated IDD through the OPN-CD44 axis. Using transforming growth factor beta 1 and siCD44 treatment, we verified that CD44 regulated the pSMAD2/3 pathway. RESULTS IDD engaged macrophage infiltration, mainly gathered in the endplate, and induced macrophage M2 polarization. Infiltrated macrophages showed high-level expression of OPN, and NPCs showed upregulated CD44. Depletion of macrophages significantly decreased the expression of OPN and CD44 in degenerative IVD, concurrently exacerbating IDD. The co-culture of macrophages and NPCs in vitro demonstrated that the conditioned media from NPCs induced macrophage M2 polarization. Further, M2 macrophages rescued NPCs extracellular matrix (ECM) phenotype through the OPN-CD44 axis, by regulating pSMAD2/3 nuclear translocation. CONCLUSIONS Our findings suggest that macrophages regulate NPC ECM expression in IDD through the OPN-CD44 axis, emphasizing the therapeutic potential of targeting macrophages and the OPN-CD44 axis for IDD prevention and treatment.
Collapse
Affiliation(s)
- Zhiwen Tao
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, Jiangsu, China.
| | - Tianyou Zhang
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, Jiangsu, China.
| | - Yaning Ge
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, Jiangsu, China.
| | - Lingzhi Li
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, Jiangsu, China.
| | - Cheng Ma
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, Jiangsu, China.
| | - Zhengbo Wang
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, Jiangsu, China.
| | - Tong Chen
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, Jiangsu, China.
| | - Helong Zhang
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, Jiangsu, China.
| | - Ruya Li
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, Jiangsu, China.
| | - Tao Jiang
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, Jiangsu, China.
| | - Yongxin Ren
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, Jiangsu, China.
| |
Collapse
|
44
|
Lin M, Zhang C, Li H, Li K, Gou S, He X, Lv C, Gao K. Pyroptosis for osteoarthritis treatment: insights into cellular and molecular interactions inflammatory. Front Immunol 2025; 16:1556990. [PMID: 40236711 PMCID: PMC11996656 DOI: 10.3389/fimmu.2025.1556990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Accepted: 03/13/2025] [Indexed: 04/17/2025] Open
Abstract
Osteoarthritis (OA) is a widely prevalent chronic degenerative disease often associated with significant pain and disability. It is characterized by the deterioration of cartilage and the extracellular matrix (ECM), synovial inflammation, and subchondral bone remodeling. Recent studies have highlighted pyroptosis-a form of programmed cell death triggered by the inflammasome-as a key factor in sustaining chronic inflammation. Central to this process are the inflammatory cytokines interleukin-1β (IL-1β) and interleukin-18 (IL-18), which play crucial roles mediating intra-articular pyroptosis through the NOD-like receptor protein 3 (NLRP3) inflammasome. This paper investigates the role of the pyroptosis pathway in perpetuating chronic inflammatory diseases and its linkage with OA. Furthermore, it explores the mechanisms of pyroptosis, mediated by nuclear factor κB (NF-κB), the purinergic receptor P2X ligand-gated ion channel 7 (P2X7R), adenosine monophosphate (AMP)-activated protein kinase (AMPK), and hypoxia-inducible factor-1α (HIF-1α). Additionally, it examines the interactions among various cellular components in the context of OA. These insights indicate that targeting the regulation of pyroptosis presents a promising therapeutic approach for the prevention and treatment of OA, offering valuable theoretical perspectives for its effective management.
Collapse
Affiliation(s)
- Minghui Lin
- Second College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Cunxin Zhang
- Department of Orthopedics, Jining No.1 People’s Hospital, Jining, China
| | - Haiming Li
- Second College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Kang Li
- Department of Orthopedics, Jining No.1 People’s Hospital, Jining, China
| | - Shuao Gou
- Jining No.1 People's Hospital, affiliated with Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Xiao He
- Department of Orthopedics, Jining No.1 People’s Hospital, Jining, China
- Medical Integration and Practice Center, Shandong University, Jinan, China
| | - Chaoliang Lv
- Department of Orthopedics, Jining No.1 People’s Hospital, Jining, China
| | - Kai Gao
- Department of Orthopedics, Jining No.1 People’s Hospital, Jining, China
| |
Collapse
|
45
|
Aguilar J, Rosú SA, Ulloa J, Gunther G, Urbano BF, Tricerri MA, Sánchez SA. Studying biological events using biopolymeric matrices. Biophys Rev 2025; 17:385-394. [PMID: 40376403 PMCID: PMC12075046 DOI: 10.1007/s12551-025-01303-z] [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: 12/01/2024] [Accepted: 03/07/2025] [Indexed: 05/18/2025] Open
Abstract
Traditional methodologies to study in vitro biological processes include simplified laboratory models where different parameters can be measured in a very controlled environment. The most used of these practices is cell plate-culturing in aqueous media. In this minimalistic model, essential components of the biological system might be ignored. One of them, disregarded for a long time, is the extracellular matrix (ECM). Extracellular matrix in eukaryotic cells is not only a frame for cells and biological components, but also an active partner of cellular metabolism and participates in several normal and pathological biological processes in a dynamic manner. ECM of eukaryotic cells has a very complex structure. Also, its mechanical properties (stiffness, viscoelasticity) depend on the organ it is associated with, and may vary from a very fluid (plasma) to a very solid (bones) structure. ECM structure and composition are very dynamic and experience temporal structural and topological changes, affecting all the existing interactions. When mimicking the ECM, three aspects are considered: the chemical environment and the physical and structural properties. In this review, we present two lines of research studying the role of the ECM in two biological implications: membrane fluidity heterogeneity and protein retention and aggregation. For these studies, we used biopolymeric matrices with very controlled features to evaluate the two events. We use traditional biochemical techniques and fluorescence microscopy to study the biological systems and traditional polymer techniques (rheology, SEM) to characterize the polymeric matrices.
Collapse
Affiliation(s)
- Joao Aguilar
- Laboratorio de Interacciones Macromoleculares, Departamento de Polímeros, Facultad de Ciencias Químicas, Universidad de Concepción, Edmundo Larenas 129, Concepción, Chile
| | - Silvana A. Rosú
- Facultad de Ciencias Médicas, Instituto de Investigaciones Bioquímicas de La Plata “Dr Prof. Rodolfo R. Brenner” (INIBIOLP), CONICET, Universidad Nacional de La Plata, Calle 60 y 120, La Plata, Buenos Aires, Argentina
- Facultad de Ciencias Médicas, Universidad Nacional de La Plata, La Plata, Buenos Aires, Argentina
| | - José Ulloa
- Laboratorio de Interacciones Macromoleculares, Departamento de Polímeros, Facultad de Ciencias Químicas, Universidad de Concepción, Edmundo Larenas 129, Concepción, Chile
| | - German Gunther
- Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Santiago, Chile
| | - Bruno F. Urbano
- Laboratorio de Interacciones Macromoleculares, Departamento de Polímeros, Facultad de Ciencias Químicas, Universidad de Concepción, Edmundo Larenas 129, Concepción, Chile
| | - M. Alejandra Tricerri
- Facultad de Ciencias Médicas, Instituto de Investigaciones Bioquímicas de La Plata “Dr Prof. Rodolfo R. Brenner” (INIBIOLP), CONICET, Universidad Nacional de La Plata, Calle 60 y 120, La Plata, Buenos Aires, Argentina
- Facultad de Ciencias Médicas, Universidad Nacional de La Plata, La Plata, Buenos Aires, Argentina
| | - Susana A. Sánchez
- Laboratorio de Interacciones Macromoleculares, Departamento de Polímeros, Facultad de Ciencias Químicas, Universidad de Concepción, Edmundo Larenas 129, Concepción, Chile
| |
Collapse
|
46
|
Mangani S, Kremmydas S, Karamanos NK. Mimicking the Complexity of Solid Tumors: How Spheroids Could Advance Cancer Preclinical Transformative Approaches. Cancers (Basel) 2025; 17:1161. [PMID: 40227664 PMCID: PMC11987746 DOI: 10.3390/cancers17071161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2025] [Revised: 03/27/2025] [Accepted: 03/28/2025] [Indexed: 04/15/2025] Open
Abstract
Traditional 2D cell culture models present significant limitations in replicating the intricate architecture and microenvironment of in vivo solid tumors, which are essential for accurately studying cancer initiation, growth, progression, and metastasis. This underscores the need for the development of advanced preclinical models to accelerate research outcomes. Emerging 3D cell culture systems, particularly spheroid models, provide a more realistic representation of solid tumor properties by capturing the complex interactions occurring within the tumor microenvironment, including the extracellular matrix dynamics that influence cancer progression. Among solid tumors, breast cancer remains the most frequently diagnosed cancer among women globally and a leading cause of cancer-related mortality. Here we emphasize the value of breast cancer cell-derived spheroids in revealing differential molecular characteristics and understanding cancer cell properties during the early stages of invasion into adjacent tissues. Conclusively, this study underscores the urgent need to adopt 3D cell culture platforms, given their significant contributions to advanced cancer research and pharmaceutical targeting. This may well offer a transformative approach for preclinical studies and enhance our ability to test therapeutic efficiency in conditions that closely mimic the growth and progression of in vivo solid tumors.
Collapse
Affiliation(s)
| | | | - Nikos K. Karamanos
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, 26504 Patras, Greece
| |
Collapse
|
47
|
Lee F, Shao X, Considine JM, Gao Y(T, Naba A. Time-lapse tryptic digestion: a proteomic approach to improve sequence coverage of extracellular matrix proteins. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.26.645502. [PMID: 40196545 PMCID: PMC11974830 DOI: 10.1101/2025.03.26.645502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
The extracellular matrix (ECM) is a complex and dynamic meshwork of proteins providing structural support to cells. It also provides biochemical signals governing cellular processes, including proliferation, adhesion, and migration. Alterations of ECM structure and/or composition have been linked to many pathological processes, including cancer and fibrosis. Over the past decade, mass-spectrometry-based proteomics has become the state-of-the-art method to profile the protein composition of ECMs. However, existing methods do not fully capture the broad dynamic range of protein abundances in the ECM. They also do not permit to achieve the high coverage needed to gain finer biochemical on ECM proteoforms (e.g., isoforms, post-translational modifications) and topographical information critical to better understand ECM protein functions. Here, we present the development of a time-lapsed proteomic pipeline using limited tryptic proteolysis and sequential release of peptides over time. This experimental pipeline was combined with data-independent acquisition mass spectrometry and the assembly of a custom matrisome spectral library to enhance peptide-to-spectrum matching. This pipeline shows superior protein identification, peptide-to-spectrum matching, and significantly increased sequence coverage against standard ECM proteomic pipelines. Exploiting the spatio-temporal resolution of this method, we further demonstrate how time-resolved 3-dimensional peptide mapping can identify protein regions differentially susceptible to trypsin, which may aid in identifying protein-protein interaction sites.
Collapse
Affiliation(s)
- Fred Lee
- Department of Physiology and Biophysics, University of Illinois Chicago, Chicago, IL 60612, U.S.A
| | - Xinhao Shao
- College of Pharmacy, University of Illinois Chicago, Chicago, IL 60612, U.S.A
| | - James M Considine
- Department of Physiology and Biophysics, University of Illinois Chicago, Chicago, IL 60612, U.S.A
| | - Yu (Tom) Gao
- College of Pharmacy, University of Illinois Chicago, Chicago, IL 60612, U.S.A
- University of Illinois Cancer Center, Chicago, IL 60612, U.S.A
| | - Alexandra Naba
- Department of Physiology and Biophysics, University of Illinois Chicago, Chicago, IL 60612, U.S.A
- University of Illinois Cancer Center, Chicago, IL 60612, U.S.A
| |
Collapse
|
48
|
Saulle I, Vitalyos AV, D’Agate D, Clerici M, Biasin M. Unveiling the impact of ERAP1 and ERAP2 on migration, angiogenesis and ER stress response. Front Cell Dev Biol 2025; 13:1564649. [PMID: 40226591 PMCID: PMC11985534 DOI: 10.3389/fcell.2025.1564649] [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: 01/21/2025] [Accepted: 03/12/2025] [Indexed: 04/15/2025] Open
Abstract
Recent studies have investigated the key roles exerted by ERAP1 and ERAP2 in maintaining cellular homeostasis, emphasizing their functions beyond traditional antigen processing and presentation. In particular, genetic variants of these IFNγ-inducible aminopeptidases significantly impact critical cellular pathways, including migration, angiogenesis, and autophagy, which are essential in immune responses and disease processes. ERAP1's influence on endothelial cell migration and VEGF-driven angiogenesis, along with ERAP2's role in managing stress-induced autophagy via the UPR, highlights their importance in cellular adaptation to stress and disease outcomes, including autoimmune diseases, cancer progression, and infections. By presenting recent insights into ERAP1 and ERAP2 functions, this review underscores their potential as therapeutic targets in immune regulation and cellular stress-response pathways.
Collapse
Affiliation(s)
- Irma Saulle
- Università degli Studi di Milano, Dipartimento di Scienze Biomediche e Cliniche, Milano, Italy
- Università degli Studi di Milano, Dipartimento di Fisiopatologia Medico-Chirurgica e dei Trapianti, Milano, Italy
| | | | - Daniel D’Agate
- Università degli Studi di Milano, Dipartimento di Scienze Biomediche e Cliniche, Milano, Italy
| | - Mario Clerici
- Università degli Studi di Milano, Dipartimento di Fisiopatologia Medico-Chirurgica e dei Trapianti, Milano, Italy
- IRCCS, Fondazione Don Carlo Gnocchi, Milano, Italy
| | - Mara Biasin
- Università degli Studi di Milano, Dipartimento di Scienze Biomediche e Cliniche, Milano, Italy
| |
Collapse
|
49
|
Khoso MA, Liu H, Zhao T, Zhao W, Huang Q, Sun Z, Dinislam K, Chen C, Kong L, Zhang Y, Liu X. Impact of plant-derived antioxidants on heart aging: a mechanistic outlook. Front Pharmacol 2025; 16:1524584. [PMID: 40191425 PMCID: PMC11969199 DOI: 10.3389/fphar.2025.1524584] [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: 11/07/2024] [Accepted: 03/07/2025] [Indexed: 04/09/2025] Open
Abstract
Heart aging involves a complex interplay of genetic and environmental influences, leading to a gradual deterioration of cardiovascular integrity and function. Age-related physiological changes, including ventricular hypertrophy, diastolic dysfunction, myocardial fibrosis, increased arterial stiffness, and endothelial dysfunction, are influenced by key mechanisms like autophagy, inflammation, and oxidative stress. This review aims to explore the therapeutic potential of plant-derived bioactive antioxidants in mitigating heart aging. These compounds, often rich in polyphenols, flavonoids, and other phytochemicals, exhibit notable antioxidant, anti-inflammatory, and cardioprotective properties. These substances have intricate cardioprotective properties, including the ability to scavenge ROS, enhance endogenous antioxidant defenses, regulate signaling pathways, and impede fibrosis and inflammation-promoting processes. By focusing on key molecular mechanisms linked to cardiac aging, antioxidants produced from plants provide significant promise to reduce age-related cardiovascular decline and improve general heart health. Through a comprehensive analysis of preclinical and clinical studies, this work highlights the mechanisms associated with heart aging and the promising effects of plant-derived antioxidants. The findings may helpful for researchers in identifying specific molecules with therapeutic and preventive potential for aging heart.
Collapse
Affiliation(s)
- Muneer Ahmed Khoso
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology, College of Pharmacy, Department of Cardiology, The Second Affiliated Hospital, Harbin Medical University, Harbin, China
- State Key Laboratory-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education, College of Pharmacy, Harbin, China
- Research Unit of Noninfectious Chronic Diseases in Frigid Zone (2019RU070), Chinese Academy of Medical Sciences, Harbin, China
| | - Heng Liu
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology, College of Pharmacy, Department of Cardiology, The Second Affiliated Hospital, Harbin Medical University, Harbin, China
- State Key Laboratory-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education, College of Pharmacy, Harbin, China
- Research Unit of Noninfectious Chronic Diseases in Frigid Zone (2019RU070), Chinese Academy of Medical Sciences, Harbin, China
| | - Tong Zhao
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology, College of Pharmacy, Department of Cardiology, The Second Affiliated Hospital, Harbin Medical University, Harbin, China
- State Key Laboratory-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education, College of Pharmacy, Harbin, China
- Research Unit of Noninfectious Chronic Diseases in Frigid Zone (2019RU070), Chinese Academy of Medical Sciences, Harbin, China
| | - Wenjie Zhao
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology, College of Pharmacy, Department of Cardiology, The Second Affiliated Hospital, Harbin Medical University, Harbin, China
- State Key Laboratory-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education, College of Pharmacy, Harbin, China
- Research Unit of Noninfectious Chronic Diseases in Frigid Zone (2019RU070), Chinese Academy of Medical Sciences, Harbin, China
| | - Qiang Huang
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology, College of Pharmacy, Department of Cardiology, The Second Affiliated Hospital, Harbin Medical University, Harbin, China
- State Key Laboratory-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education, College of Pharmacy, Harbin, China
- Research Unit of Noninfectious Chronic Diseases in Frigid Zone (2019RU070), Chinese Academy of Medical Sciences, Harbin, China
| | - Zeqi Sun
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology, College of Pharmacy, Department of Cardiology, The Second Affiliated Hospital, Harbin Medical University, Harbin, China
- State Key Laboratory-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education, College of Pharmacy, Harbin, China
- Research Unit of Noninfectious Chronic Diseases in Frigid Zone (2019RU070), Chinese Academy of Medical Sciences, Harbin, China
| | - Khuzin Dinislam
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology, College of Pharmacy, Department of Cardiology, The Second Affiliated Hospital, Harbin Medical University, Harbin, China
- State Key Laboratory-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education, College of Pharmacy, Harbin, China
- Research Unit of Noninfectious Chronic Diseases in Frigid Zone (2019RU070), Chinese Academy of Medical Sciences, Harbin, China
| | - Chen Chen
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology, College of Pharmacy, Department of Cardiology, The Second Affiliated Hospital, Harbin Medical University, Harbin, China
- State Key Laboratory-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education, College of Pharmacy, Harbin, China
- Research Unit of Noninfectious Chronic Diseases in Frigid Zone (2019RU070), Chinese Academy of Medical Sciences, Harbin, China
| | - Lingyi Kong
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology, College of Pharmacy, Department of Cardiology, The Second Affiliated Hospital, Harbin Medical University, Harbin, China
- State Key Laboratory-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education, College of Pharmacy, Harbin, China
- Research Unit of Noninfectious Chronic Diseases in Frigid Zone (2019RU070), Chinese Academy of Medical Sciences, Harbin, China
| | - Yong Zhang
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology, College of Pharmacy, Department of Cardiology, The Second Affiliated Hospital, Harbin Medical University, Harbin, China
- State Key Laboratory-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education, College of Pharmacy, Harbin, China
- Research Unit of Noninfectious Chronic Diseases in Frigid Zone (2019RU070), Chinese Academy of Medical Sciences, Harbin, China
| | - Xin Liu
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology, College of Pharmacy, Department of Cardiology, The Second Affiliated Hospital, Harbin Medical University, Harbin, China
- State Key Laboratory-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education, College of Pharmacy, Harbin, China
- Research Unit of Noninfectious Chronic Diseases in Frigid Zone (2019RU070), Chinese Academy of Medical Sciences, Harbin, China
| |
Collapse
|
50
|
Ji K, Wen B, Wang X, Chen L, Chen Y, Wang L, Bao J, Pan X, Zhang G, Jiang Y, Liu H. HIF1A facilitates hypoxia-induced changes in H3K27ac modification to promote myometrial contractility. Commun Biol 2025; 8:475. [PMID: 40119120 PMCID: PMC11928739 DOI: 10.1038/s42003-025-07880-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Accepted: 03/03/2025] [Indexed: 03/24/2025] Open
Abstract
Prior studies have established that myometrial hypoxia during labor is pivotal in intensifying contractions, the alterations in gene expression and histone modifications in myometrial cells under hypoxia have yet to be documented. Here, hypoxia's enhancement of cellular contractility was confirmed, and RNA-seq identified 2,262 differentially expressed genes in human myometrial smooth muscle cells (hMSMCs) under hypoxia. Chromatin immunoprecipitation (ChIP), high-throughput chromosome conformation capture followed by ChIP (Hi-ChIP) were employed to investigate the epigenetic changes, specifically histone modifications (H3K27ac, H3K4me1, H3K27me3, and H3K4me3), in hMSMCs under hypoxia. We identified the enhancer and super-enhancer regions in hMSMCs and found HIF1A as the key mediator of these H3K27ac changes under hypoxia. Labor-associated genes regulated by HIF1A have been identified. Validation experiments on these genes such as CXCL8, RUNX1, IL-6, and PTGES3 demonstrated that HIF1A knockdown reduces their expression and associated H3K27ac modifications in peak regions of their promoters or enhancers. These findings indicate that HIF1A probably mediate changes in histone H3K27ac modifications to regulate myometrial cell contractions under hypoxia, providing potential therapeutic and intervention targets for disorders related to parturition.
Collapse
Affiliation(s)
- Kaiyuan Ji
- Guangzhou Key Laboratory of Maternal-Fetal Medicine, Institute of Reproductive Health and Perinatology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Bolun Wen
- Guangzhou Key Laboratory of Maternal-Fetal Medicine, Institute of Reproductive Health and Perinatology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Xiaodi Wang
- Guangzhou Key Laboratory of Maternal-Fetal Medicine, Institute of Reproductive Health and Perinatology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Lina Chen
- Guangzhou Key Laboratory of Maternal-Fetal Medicine, Institute of Reproductive Health and Perinatology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Yunshan Chen
- Guangzhou Key Laboratory of Maternal-Fetal Medicine, Institute of Reproductive Health and Perinatology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Lele Wang
- Guangzhou Key Laboratory of Maternal-Fetal Medicine, Institute of Reproductive Health and Perinatology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Junjie Bao
- Guangzhou Key Laboratory of Maternal-Fetal Medicine, Institute of Reproductive Health and Perinatology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Xiuyu Pan
- Guangzhou Key Laboratory of Maternal-Fetal Medicine, Institute of Reproductive Health and Perinatology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Guozheng Zhang
- Guangzhou Key Laboratory of Maternal-Fetal Medicine, Institute of Reproductive Health and Perinatology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Yanmin Jiang
- Guangzhou Key Laboratory of Maternal-Fetal Medicine, Institute of Reproductive Health and Perinatology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, China.
| | - Huishu Liu
- Guangzhou Key Laboratory of Maternal-Fetal Medicine, Institute of Reproductive Health and Perinatology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, China.
| |
Collapse
|