1
|
Zhurenkov KE, Svirskis D, Connor B, Malmström J. Actuated Hydrogel Platforms To Study Brain Cell Behavior. Adv Healthc Mater 2025; 14:e2404484. [PMID: 40091290 PMCID: PMC12004428 DOI: 10.1002/adhm.202404484] [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: 11/12/2024] [Revised: 02/16/2025] [Indexed: 03/19/2025]
Abstract
The human brain is a highly complex organ characterized by intricate neural networks, biochemical signaling, and unique mechanical properties. The soft and dynamic viscoelastic extracellular matrix (ECM) plays a crucial role in supporting different types of brain cells and influencing their behavior. Understanding how brain cells respond to mechanical stimuli within this complex environment is essential for unraveling fundamental mechanisms of healthy, unhealthy, and regenerative functions within the central nervous system. This requires the development of advanced materials and techniques to study the interplay between mechanical cues and cell responses. Hydrogels have become essential in this research, mimicking the brain's ECM in both chemical composition and mechanical behavior. Conventional hydrogels, while helpful, are static and lack dynamic stimulation. On the other hand, dynamic hydrogels provide reversible, dynamic stimulation, closely replicating the brain's ECM properties. This review discusses current hydrogel platforms used to investigate brain function in health and disease, focusing on traumatic brain injury (TBI)-like conditions and brain tumors. These dynamic materials offer sophisticated tools for understanding brain cell mechanobiology and developing new therapeutic approaches.
Collapse
Affiliation(s)
- Kirill E. Zhurenkov
- Department of Chemical and Materials EngineeringThe University of AucklandAuckland1010New Zealand
- MacDiarmid Institute for Advanced Materials and NanotechnologyWellington6140New Zealand
| | - Darren Svirskis
- School of PharmacyFaculty of Medical and Health SciencesThe University of AucklandAuckland1023New Zealand
| | - Bronwen Connor
- Department of Pharmacology and Clinical PharmacologySchool of Medical Sciences, Faculty of Medical and Health SciencesThe University of AucklandAuckland1023New Zealand
| | - Jenny Malmström
- Department of Chemical and Materials EngineeringThe University of AucklandAuckland1010New Zealand
- MacDiarmid Institute for Advanced Materials and NanotechnologyWellington6140New Zealand
| |
Collapse
|
2
|
Sun Q, Qiu T, Liu X, Wei Q. Cellular Spatial Sensing Determines Cell Mechanotransduction Activity on the Aligned Nanofibers. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2410351. [PMID: 39967397 DOI: 10.1002/smll.202410351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/03/2024] [Revised: 02/06/2025] [Indexed: 02/20/2025]
Abstract
The geometric properties of extracellular matrix (ECM) fibers play a crucial role in regulating cellular behaviors and functions. Although extensive research has examined the effects of fiber alignment, conflicting results have often arisen, leaving the precise mechanisms by which electrospun fiber alignment affects cellular behavior still unclear. This study investigates how the arrangement of polycaprolactone (PCL) electrospun fiber substrates affects cellular mechanosensing by modulating cell positioning. Larger cells, whose width on a coverslip exceeds 5 times the width of the aligned fiber gaps (≈8 µm in this study) and that span multiple aligned fibers, demonstrate enhanced spreading and mechanotransduction. Conversely, smaller cells, whose width is less than or equal to 2.5 times the width of the aligned fiber gaps and are confined within fiber interstices, exhibit limited mechanotransductive signaling. These findings are further supported by manipulating cell size and, more importantly, have led to the fabrication of semi-aligned fiber networks that enhance both cell spreading and mechanotransduction. This research emphasizes the importance of optimizing fiber architecture to improve cellular interactions, offering valuable insights for the design of biomimetic scaffolds in tissue regeneration.
Collapse
Affiliation(s)
- Qian Sun
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials and Engineering, Sichuan University, Chengdu, 610065, P. R. China
| | - Tiecheng Qiu
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials and Engineering, Sichuan University, Chengdu, 610065, P. R. China
| | - Xiaojing Liu
- Department of Pediatric Dentistry, School and Hospital of Stomatology, Cheeloo College of Medicine, and Shandong Key Laboratory of Oral Tissue Regeneration, Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration, and Shandong Provincial Clinical Research Center for Oral Diseases, Shandong University, Jinan, 250012, P. R. China
| | - Qiang Wei
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials and Engineering, Sichuan University, Chengdu, 610065, P. R. China
| |
Collapse
|
3
|
Plath AMS, de Lima PHC, Amicone A, Bissacco EG, Mosayebi M, Berton SBR, Ferguson SJ. Toward low-friction and high-adhesion solutions: Emerging strategies for nanofibrous scaffolds in articular cartilage engineering. BIOMATERIALS ADVANCES 2025; 169:214129. [PMID: 39642717 DOI: 10.1016/j.bioadv.2024.214129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Revised: 11/28/2024] [Accepted: 11/29/2024] [Indexed: 12/09/2024]
Abstract
Aging, trauma, pathology, and poor natural tissue regeneration are the leading causes of osteoarthritis (OA), an articular cartilage disease. Electrospun scaffolds have gained attention as potential matrices for the treatment of OA because of their high degree of ECM mimicry, which suits chondrocyte migration, adhesion, and proliferation. However, none of the products recently introduced in the market are nanofiber-based. This study aimed to review the scope and tribology of nanofibrous articular cartilage scaffolds. Herein, we briefly discuss cartilage lubrication and strategies for promoting cell adhesion in electrospun materials. Next, we discuss the emerging need to study the biotribological properties of scaffolds. Finally, we review new perspectives on surface functionalization, surface segregation, Janus membranes, layer-by-layer fabrication, and nanofibrous composites. We conclude that cell adhesion and low-friction conciliation remain poorly explored in the recent literature. The topic intersection might create novelties in the field.
Collapse
Affiliation(s)
| | - Pedro Henrique Correia de Lima
- Department of Physics and Chemistry, São Paulo State University (UNESP), Av. Brasil, 56, 15385007 Ilha Solteira, Brazil.
| | - Alessio Amicone
- Institute for Biomechanics, ETH Zurich, Gloriastrasse 37-39, 8092 Zurich, Switzerland
| | | | - Mahdieh Mosayebi
- Institute for Biomechanics, ETH Zurich, Gloriastrasse 37-39, 8092 Zurich, Switzerland
| | | | - Stephen J Ferguson
- Institute for Biomechanics, ETH Zurich, Gloriastrasse 37-39, 8092 Zurich, Switzerland
| |
Collapse
|
4
|
Lou Y, Dai C, Feng S, Jin R, Liu S, Zhou Z, Wang M, Yang Y, Xu G, Hou J, Chen J, Tang G, Wang N, Bai H, Chen Z. Biomimetic rhCOL17-P334 Conjugate for Enhanced Wound Healing. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2417980. [PMID: 39988836 DOI: 10.1002/adma.202417980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Revised: 01/25/2025] [Indexed: 02/25/2025]
Abstract
Wound healing remains a significant global health challenge, affecting millions annually and imposing substantial economic burdens. Most commercially available biomaterials for wound management primarily address external symptoms, including hemostasis, exudation, scarring, and infection. Advanced biomaterials derived from endogenous molecules aim to better replicate the native wound microenvironment, promoting enhanced repair. Since wounds frequently occur on exposed skin, which is vulnerable to UVA radiation and requires protective yet invisible materials, traditional wound care products often lack these essential features. Inspired by natural UV protection mechanisms, a novel bioscaffold is developed using recombinant human collagen XVII (rhCOL17) crosslinked with porphyra-334 (P334) to improve wound healing under UVA exposure. The resulting rhCOL17-P334 conjugate integrates extracellular matrix (ECM)-like cues with UVA-shielding properties provided by P334. This conjugate is used to construct a transparent, injectable hydrogel combining gelatin methacryloyl (GelMA) and rhCOL17-P334 (GCP). GCP significantly inhibits UVA-induced fibroblast senescence and improves wound healing outcomes by targeting integrin α6β4 through rhCOL17. Its transparency facilitates convenient wound monitoring while also addressing the aesthetic requirement for invisibility. By combining UVA shielding with wound repair capabilities, GCP presents a promising platform for advanced wound management.
Collapse
Affiliation(s)
- Yijie Lou
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, 310053, China
| | - Chunyan Dai
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, 310053, China
| | - Shuting Feng
- Institute of Marine Biology and Pharmacology, Ocean College, Zhejiang University, Zhoushan, 316021, China
- The Hainan Institute, Zhejiang University, Sanya, 572025, China
| | - Rijuan Jin
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, 310053, China
| | - Shuangshuang Liu
- Core Facilities, Zhejiang University School of Medicine, Hangzhou, 310027, China
| | - Zhanyi Zhou
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, 310053, China
| | - Minjun Wang
- Institute of Marine Biology and Pharmacology, Ocean College, Zhejiang University, Zhoushan, 316021, China
| | - Yang Yang
- Institute for Composites Science Innovation (InCSI), School of Materials Science and Engineering, Zhejiang University, Hangzhou, 310058, China
| | - Guoqiao Xu
- Institute of Chemical Biology and Pharmaceutical Chemistry, Department of Chemistry, Zhejiang University, Hangzhou, 310028, China
| | - Jue Hou
- Institute of Chemical Biology and Pharmaceutical Chemistry, Department of Chemistry, Zhejiang University, Hangzhou, 310028, China
| | - Jiayi Chen
- College of Biomedical Engineering & Instrument Science, Zhejiang University, Hangzhou, 310028, China
| | - Guping Tang
- Institute of Chemical Biology and Pharmaceutical Chemistry, Department of Chemistry, Zhejiang University, Hangzhou, 310028, China
| | - Nan Wang
- Institute of Marine Biology and Pharmacology, Ocean College, Zhejiang University, Zhoushan, 316021, China
- The Hainan Institute, Zhejiang University, Sanya, 572025, China
| | - Hongzhen Bai
- Institute of Chemical Biology and Pharmaceutical Chemistry, Department of Chemistry, Zhejiang University, Hangzhou, 310028, China
| | - Zhe Chen
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, 310053, China
| |
Collapse
|
5
|
Liu S, Kilian D, Bernhardt A, Wirsig K, von Witzleben M, Duin S, Lode A, Hu Q, Gelinsky M. Novel Protein-Rich Bioactive Bioink Stimulates Cellular Proliferation and Response in 3D Bioprinted Volumetric Constructs. Adv Healthc Mater 2025; 14:e2404470. [PMID: 39995366 PMCID: PMC12004440 DOI: 10.1002/adhm.202404470] [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: 11/12/2024] [Revised: 01/10/2025] [Indexed: 02/26/2025]
Abstract
3D extrusion bioprinting, a promising and widely adopted technology in the emerging field of biofabrication, has gained considerable attention for its ability to fabricate hierarchically structured, native-mimicking tissue substitutes with precisely defined cell distributions. Despite notable advancements, the limited availability of suitably bioactive bioinks remains a major challenge, hindering the construction of volumetric tissue substitutes effectively mimicking biological functionality. Therefore, this work proposes a protein-rich, low-cost, bioactive bioink: abundantly available eggwhite powder (EWP) is leveraged to functionalize an alginate-methylcellulose (AlgMC) hydrogel matrix and enhance cellular response. The developed EWP-supplemented bioinks not only maintain favorable printability and high shape fidelity but also exhibit remarkable bioactivity. Notably, incorporating EWP into AlgMC-based bioinks enhances shear-thinning features, thereby improving the viability of encapsulated cells within the bioprinted constructs. The versatility and biofunctionality of EWP in bioprinted constructs are demonstrated using three distinct cell types, encompassing sources such as a stem cell line, human soft skin, and stiff bone tissues. Furthermore, the promising and wide applicability of the EWP-supplemented bioink for biofabrication is demonstrated exemplarily in core-shell and multi-channel bioprinting strategies as a proof-of-concept for functional tissue construction. These findings underscore the significant and versatile potential of this novel bioink in biofabrication and biomedical applications.
Collapse
Affiliation(s)
- Suihong Liu
- Centre for Translational BoneJoint and Soft Tissue ResearchFaculty of Medicine and University Hospital Carl Gustav CarusTechnische Universität Dresden01307DresdenGermany
- Rapid Manufacturing Engineering CenterSchool of Mechatronic Engineering and AutomationShanghai UniversityShanghai200444China
- Shanghai Key Laboratory of Intelligent Manufacturing and RoboticsSchool of Mechatronic Engineering and AutomationShanghai UniversityShanghai200444China
- Present address:
Engineering Science and Mechanics DepartmentPenn State UniversityUniversity ParkPA16802USA
| | - David Kilian
- Centre for Translational BoneJoint and Soft Tissue ResearchFaculty of Medicine and University Hospital Carl Gustav CarusTechnische Universität Dresden01307DresdenGermany
- Present address:
Department of Materials Science & EngineeringStanford UniversityStanfordCA94305USA
| | - Anne Bernhardt
- Centre for Translational BoneJoint and Soft Tissue ResearchFaculty of Medicine and University Hospital Carl Gustav CarusTechnische Universität Dresden01307DresdenGermany
| | - Katharina Wirsig
- Centre for Translational BoneJoint and Soft Tissue ResearchFaculty of Medicine and University Hospital Carl Gustav CarusTechnische Universität Dresden01307DresdenGermany
| | - Max von Witzleben
- Centre for Translational BoneJoint and Soft Tissue ResearchFaculty of Medicine and University Hospital Carl Gustav CarusTechnische Universität Dresden01307DresdenGermany
| | - Sarah Duin
- Centre for Translational BoneJoint and Soft Tissue ResearchFaculty of Medicine and University Hospital Carl Gustav CarusTechnische Universität Dresden01307DresdenGermany
| | - Anja Lode
- Centre for Translational BoneJoint and Soft Tissue ResearchFaculty of Medicine and University Hospital Carl Gustav CarusTechnische Universität Dresden01307DresdenGermany
| | - Qingxi Hu
- Rapid Manufacturing Engineering CenterSchool of Mechatronic Engineering and AutomationShanghai UniversityShanghai200444China
- Shanghai Key Laboratory of Intelligent Manufacturing and RoboticsSchool of Mechatronic Engineering and AutomationShanghai UniversityShanghai200444China
- National Demonstration Center for Experimental Engineering Training EducationShanghai UniversityShanghai200444China
| | - Michael Gelinsky
- Centre for Translational BoneJoint and Soft Tissue ResearchFaculty of Medicine and University Hospital Carl Gustav CarusTechnische Universität Dresden01307DresdenGermany
| |
Collapse
|
6
|
Hartley A, Williams PM, Mata A. A Comparison of the Mechanical Properties of ECM Components and Synthetic Self-Assembling Peptides. Adv Healthc Mater 2025; 14:e2402385. [PMID: 39972630 DOI: 10.1002/adhm.202402385] [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: 06/29/2024] [Revised: 01/20/2025] [Indexed: 02/21/2025]
Abstract
The field of tissue engineering is increasingly moving away from a one-size-fits-all approach of simple synthetic homogeneous gels, and embracing more tailored designs to optimize cell function and differentiation for the organ of interest. Extracellular matrix (ECM) proteins are still the optimal route for controlling cell function, while a field of great promise is that of synthetic self-assembling peptides (SSAPs), which are fully biocompatible, biodegradable, and offer both the hierarchical structure and dynamic properties displayed by protein networks found in natural tissue. However, the mechanical properties of neither group have been comprehensively reviewed. In this review, rheological data and the Young's modulus of the most prevalent proteins involved in the ECM (collagen I, elastin, and fibronectin) are collated for the first time, and compared against the most widely researched SSAPs: peptide amphiphiles (PAs), β-sheets, β-hairpin peptides, and Fmoc-based gels (with a focus on PA-E3, RADA16, MAX1, and FmocFF, respectively).
Collapse
Affiliation(s)
- Alex Hartley
- School of Pharmacy, University of Nottingham, University Park Campus, Nottingham, NG7 2RD, UK
- Biodiscovery Institute, University of Nottingham, University Park Campus, Nottingham, NG7 2RD, UK
| | - Philip Michael Williams
- School of Pharmacy, University of Nottingham, University Park Campus, Nottingham, NG7 2RD, UK
| | - Alvaro Mata
- School of Pharmacy, University of Nottingham, University Park Campus, Nottingham, NG7 2RD, UK
- Biodiscovery Institute, University of Nottingham, University Park Campus, Nottingham, NG7 2RD, UK
- Department of Chemical and Environmental Engineering, University of Nottingham, University Park Campus, Nottingham, NG7 2RD, UK
| |
Collapse
|
7
|
Liu S, Jin P. Advances and Challenges in 3D Bioprinted Cancer Models: Opportunities for Personalized Medicine and Tissue Engineering. Polymers (Basel) 2025; 17:948. [PMID: 40219336 PMCID: PMC11991528 DOI: 10.3390/polym17070948] [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: 01/24/2025] [Revised: 03/20/2025] [Accepted: 03/27/2025] [Indexed: 04/14/2025] Open
Abstract
Cancer is the second leading cause of death worldwide, after cardiovascular disease, claiming not only a staggering number of lives but also causing considerable health and economic devastation, particularly in less-developed countries. Therapeutic interventions are impeded by differences in patient-to-patient responses to anti-cancer drugs. A personalized medicine approach is crucial for treating specific patient groups and includes using molecular and genetic screens to find appropriate stratifications of patients who will respond (and those who will not) to treatment regimens. However, information on which risk stratification method can be used to hone in on cancer types and patients who will be likely responders to a specific anti-cancer agent remains elusive for most cancers. Novel developments in 3D bioprinting technology have been widely applied to recreate relevant bioengineered tumor organotypic structures capable of mimicking the human tissue and microenvironment or adequate drug responses in high-throughput screening settings. Parts are autogenously printed in the form of 3D bioengineered tissues using a computer-aided design concept where multiple layers include different cell types and compatible biomaterials to build specific configurations. Patient-derived cancer and stromal cells, together with genetic material, extracellular matrix proteins, and growth factors, are used to create bioprinted cancer models that provide a possible platform for the screening of new personalized therapies in advance. Both natural and synthetic biopolymers have been used to encourage the growth of cells and biological materials in personalized tumor models/implants. These models may facilitate physiologically relevant cell-cell and cell-matrix interactions with 3D heterogeneity resembling real tumors.
Collapse
Affiliation(s)
- Sai Liu
- Health Science Center, Yangtze University, Jingzhou 434023, China;
| | | |
Collapse
|
8
|
Zeng CW. Immune Cell-NSPC interactions: Friend or foe in CNS injury and repair? Differentiation 2025; 143:100855. [PMID: 40112742 DOI: 10.1016/j.diff.2025.100855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Revised: 03/07/2025] [Accepted: 03/10/2025] [Indexed: 03/22/2025]
Abstract
Neural stem/progenitor cells (NSPCs) play a crucial role in central nervous system (CNS) development, regeneration, and repair. However, their functionality and therapeutic potential are intricately modulated by interactions with immune cells, particularly macrophages and microglia. Microglia, as CNS-resident macrophages, are distinct from peripheral macrophages in their roles and characteristics, contributing to specialized functions within the CNS. Recent evidence suggests that microglia, as CNS-resident macrophages, contribute to the quality assurance of NSPCs by eliminating stressed or dysfunctional cells, yet the mechanisms underlying this process remain largely unexplored. Furthermore, macrophage polarization states, such as M1 and M2, appear to differentially influence NSPC quality, potentially impacting neurogenesis and regenerative outcomes. Identifying surface markers indicative of NSPC stress could provide a strategy for selecting optimal cells for transplantation therapies. Additionally, in vivo clonal labeling approaches may enable precise tracking of NSPC fate and their interactions with immune cells. Beyond macrophages and microglia, the roles of other immune cells, including T cells and neutrophils, particularly in injury and neurodegenerative disease contexts, in the context of CNS injury and disease are emerging areas of interest. Here, I discuss the emerging evidence supporting the interplay between the immune system and NSPCs, highlighting critical gaps in knowledge and proposing future research directions to harness immune-mediated mechanisms for optimizing neural regeneration and transplantation strategies.
Collapse
Affiliation(s)
- Chih-Wei Zeng
- Department of Neuroscience, UT Southwestern Medical Center, Dallas, TX, 75390, USA.
| |
Collapse
|
9
|
Selemani MA, Kabandana GKM, Chen C, Martin RS. 3D-Printed Microfluidic-Based Cell Culture System With Analysis to Investigate Macrophage Activation. Electrophoresis 2025. [PMID: 39964958 DOI: 10.1002/elps.8109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Revised: 01/06/2025] [Accepted: 01/30/2025] [Indexed: 02/20/2025]
Abstract
In this paper, we describe the development of 3D-printed microfluidic cell culture devices that can be coupled with a circulation system to study the dynamics of both intracellular and extracellular (release) processes. Key to this approach is the ability to quantitate key analytes on a minutes timescale with either on-line (in this study, quantitating nitric oxide production using an amperometric flow cell) or off-line (in this work, quantitating intracellular itaconate production with LC/MS) analytical measurements. To demonstrate the usefulness of this approach, we chose to study macrophage polarization as a function of the extracellular matrix (silk) fiber size, a major area of research in tissue engineering. It was found that the use of larger fibers (1280 nm vs. smaller 512 nm fibers) led to increases in the production of both nitric oxide and itaconate. These findings set the foundation for future research for the creation of finely tuned microfluidic 3D cell culture approaches in areas where flow and the extracellular matrix play a significant role in barrier transport and where integrated analysis of key markers is needed.
Collapse
Affiliation(s)
- Major A Selemani
- Department of Chemistry, Saint Louis University, St. Louis, Missouri, USA
| | | | - Chengpeng Chen
- Department of Chemistry and Biochemistry, University of Maryland-Baltimore County, Baltimore, Maryland, USA
| | - R Scott Martin
- Department of Chemistry, Saint Louis University, St. Louis, Missouri, USA
- Saint Louis University Center for Additive Manufacturing, St. Louis, Missouri, USA
| |
Collapse
|
10
|
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] [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
|
11
|
Klabukov I, Kabakov AE, Yakimova A, Baranovskii D, Sosin D, Atiakshin D, Ignatyuk M, Yatsenko E, Rybachuk V, Evstratova E, Eygel D, Kudlay D, Stepanenko V, Shegay P, Kaprin AD. Tumor-Associated Extracellular Matrix Obstacles for CAR-T Cell Therapy: Approaches to Overcoming. Curr Oncol 2025; 32:79. [PMID: 39996879 PMCID: PMC11854105 DOI: 10.3390/curroncol32020079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2024] [Revised: 01/25/2025] [Accepted: 01/28/2025] [Indexed: 02/26/2025] Open
Abstract
Chimeric antigen receptor (CAR)-T cell therapy yields good results in the treatment of various hematologic malignancies. However, the efficacy of CAR-T cell therapy against solid tumors has proven to be limited, primarily because the tumor-associated extracellular matrix (ECM) creates an intractable barrier for the cytotoxic CAR-T cells that are supposed to kill cancer cells. This review unravels the multifaceted role of the tumor-associated ECM in impeding CAR-T cell infiltration, survival, and functions within solid tumors. We analyze the situations when intratumoral ECM limits the efficacy of CAR-T cell therapy by being a purely physical barrier that complicates lymphocyte penetration/migration and also acts as an immunosuppressive factor that impairs the antitumor activities of CAR-T cells. In addition, we highlight promising approaches such as engineering CAR-T cells with improved capabilities to penetrate and migrate into/through the intratumoral ECM, combination therapies aimed at attenuating the high density and immunosuppressive potential of the intratumoral ECM, and others that enable overcoming ECM-related obstacles. A detailed overview of the data of relevant studies not only helps to better understand the interactions between CAR-T cells and the intratumoral ECM but also outlines potential ways to more effectively use CAR-T cell therapy against solid tumors.
Collapse
Affiliation(s)
- Ilya Klabukov
- National Medical Research Radiological Centre of the Ministry of Health of the Russian Federation, Koroleva St. 4, 249036 Obninsk, Russia
- A. Tsyb Medical Radiological Research Center—Branch of the National Medical Research Radiological Centre of the Ministry of Health of the Russian Federation, Zhukova St. 10, 249036 Obninsk, Russia
- GMP-Laboratory for Advanced Therapy Medicinal Products, Patrice Lumumba Peoples’ Friendship University of Russia (RUDN University), Miklukho-Maklay St. 6, 117198 Moscow, Russia
- Obninsk Institute for Nuclear Power Engineering of the National Research Nuclear University MEPhI, Studgorodok 1, 249039 Obninsk, Russia
| | - Alexander E. Kabakov
- National Medical Research Radiological Centre of the Ministry of Health of the Russian Federation, Koroleva St. 4, 249036 Obninsk, Russia
- A. Tsyb Medical Radiological Research Center—Branch of the National Medical Research Radiological Centre of the Ministry of Health of the Russian Federation, Zhukova St. 10, 249036 Obninsk, Russia
| | - Anna Yakimova
- National Medical Research Radiological Centre of the Ministry of Health of the Russian Federation, Koroleva St. 4, 249036 Obninsk, Russia
- A. Tsyb Medical Radiological Research Center—Branch of the National Medical Research Radiological Centre of the Ministry of Health of the Russian Federation, Zhukova St. 10, 249036 Obninsk, Russia
| | - Denis Baranovskii
- National Medical Research Radiological Centre of the Ministry of Health of the Russian Federation, Koroleva St. 4, 249036 Obninsk, Russia
- A. Tsyb Medical Radiological Research Center—Branch of the National Medical Research Radiological Centre of the Ministry of Health of the Russian Federation, Zhukova St. 10, 249036 Obninsk, Russia
- GMP-Laboratory for Advanced Therapy Medicinal Products, Patrice Lumumba Peoples’ Friendship University of Russia (RUDN University), Miklukho-Maklay St. 6, 117198 Moscow, Russia
- University Hospital Basel, Basel University, 4001 Basel, Switzerland
| | - Dmitry Sosin
- Centre for Strategic Planning and Management of Biomedical Health Risks of the Federal Medical Biological Agency, 119121 Moscow, Russia
| | - Dmitry Atiakshin
- Scientific and Educational Resource Center for Innovative Technologies of Immunophenotyping, Digital Spatial Profiling and Ultrastructural Analysis, Patrice Lumumba Peoples’ Friendship University of Russia (RUDN University), 117198 Moscow, Russia
| | - Michael Ignatyuk
- Scientific and Educational Resource Center for Innovative Technologies of Immunophenotyping, Digital Spatial Profiling and Ultrastructural Analysis, Patrice Lumumba Peoples’ Friendship University of Russia (RUDN University), 117198 Moscow, Russia
| | - Elena Yatsenko
- National Medical Research Radiological Centre of the Ministry of Health of the Russian Federation, Koroleva St. 4, 249036 Obninsk, Russia
- A. Tsyb Medical Radiological Research Center—Branch of the National Medical Research Radiological Centre of the Ministry of Health of the Russian Federation, Zhukova St. 10, 249036 Obninsk, Russia
| | - Victoria Rybachuk
- National Medical Research Radiological Centre of the Ministry of Health of the Russian Federation, Koroleva St. 4, 249036 Obninsk, Russia
| | - Ekaterina Evstratova
- National Medical Research Radiological Centre of the Ministry of Health of the Russian Federation, Koroleva St. 4, 249036 Obninsk, Russia
- A. Tsyb Medical Radiological Research Center—Branch of the National Medical Research Radiological Centre of the Ministry of Health of the Russian Federation, Zhukova St. 10, 249036 Obninsk, Russia
| | - Daria Eygel
- National Medical Research Radiological Centre of the Ministry of Health of the Russian Federation, Koroleva St. 4, 249036 Obninsk, Russia
- A. Tsyb Medical Radiological Research Center—Branch of the National Medical Research Radiological Centre of the Ministry of Health of the Russian Federation, Zhukova St. 10, 249036 Obninsk, Russia
| | - Dmitry Kudlay
- Immunology Department, Institute of Immunology FMBA of Russia, 115552 Moscow, Russia
- Department of Pharmacognosy and Industrial Pharmacy, Faculty of Fundamental Medicine, Lomonosov Moscow State University, 119992 Moscow, Russia
| | - Vasiliy Stepanenko
- Institute of Pharmacy, Sechenov First Moscow State Medical University (Sechenov University), 119435 Moscow, Russia
| | - Peter Shegay
- National Medical Research Radiological Centre of the Ministry of Health of the Russian Federation, Koroleva St. 4, 249036 Obninsk, Russia
| | - Andrey D. Kaprin
- National Medical Research Radiological Centre of the Ministry of Health of the Russian Federation, Koroleva St. 4, 249036 Obninsk, Russia
- Scientific and Educational Resource Center for Innovative Technologies of Immunophenotyping, Digital Spatial Profiling and Ultrastructural Analysis, Patrice Lumumba Peoples’ Friendship University of Russia (RUDN University), 117198 Moscow, Russia
| |
Collapse
|
12
|
Namdari M, McDonnell FS. Extracellular vesicles as emerging players in glaucoma: Mechanisms, biomarkers, and therapeutic targets. Vision Res 2025; 226:108522. [PMID: 39581065 PMCID: PMC11640964 DOI: 10.1016/j.visres.2024.108522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 11/05/2024] [Accepted: 11/07/2024] [Indexed: 11/26/2024]
Abstract
In recent years, extracellular vesicles (EVs) have attracted significant scientific interest due to their widespread distribution, their potential as disease biomarkers, and their promising applications in therapy. Encapsulated by lipid bilayers these nanovesicles include small extracellular vesicles (sEV) (30-150 nm), microvesicles (100-1000 nm), and apoptotic bodies (100-5000 nm) and are essential for cellular communication, immune responses, biomolecular transport, and physiological regulation. As they reflect the condition and functionality of their originating cells, EVs play critical roles in numerous physiological processes and diseases. Therefore, EVs offer valuable opportunities for uncovering disease mechanisms, enhancing drug delivery systems, and identifying novel biomarkers. In the context of glaucoma, a leading cause of irreversible blindness, the specific roles of EVs are still largely unexplored. This review examines the emerging role of EVs in the pathogenesis of glaucoma, with a focus on their potential as diagnostic biomarkers and therapeutic agents. Through a thorough analysis of current literature, we summarize key advancements in EV research and identify areas where further investigation is needed to fully understand their function in glaucoma.
Collapse
Affiliation(s)
- Maral Namdari
- John A. Moran Eye Center, University of Utah, Salt Lake City, UT, USA
| | - Fiona S McDonnell
- John A. Moran Eye Center, University of Utah, Salt Lake City, UT, USA; Biomedical Engineering, University of Utah, Salt Lake City, UT, USA; Pharmacology and Toxicology, University of Utah Salt Lake City, UT, USA.
| |
Collapse
|
13
|
Negishi J, Yamaguchi A, Tanaka D, Hashimoto Y, Zhang Y, Funamoto S. A Method for Fabricating Tissue-Specific Extracellular Matrix Blocks From Decellularized Tissue Powders. Adv Biol (Weinh) 2025; 9:e2400398. [PMID: 39601529 DOI: 10.1002/adbi.202400398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 11/07/2024] [Indexed: 11/29/2024]
Abstract
Decellularized tissues retain the extracellular matrix (ECM), shape, and composition that are unique to the source tissue. Previous studies using decellularized tissue lysates and powders have shown that tissue-specific ECM plays a key role in cellular function and wound healing. However, creating decellularized tissues composed of tissue-specific ECM with customizable shapes and structures for use as scaffolding materials remains challenging. In this study, a method for compacting decellularized tissue powder into blocks is developed using cold isostatic pressing (CIP). Custom-shaped ECM blocks and composite ECM blocks are fabricated using silicone molds. Additionally, an ECM block with a two-layer structure is obtained through a two-step CIP process. Cells are observed to infiltrate porous ECM blocks that are created using sodium chloride and transglutaminase. These results highlight the development of an effective method for producing ECM blocks using CIP with customizable shapes, compositions, and structures, making them suitable for use as cell culture scaffolds.
Collapse
Affiliation(s)
- Jun Negishi
- Faculty of Textile Science and Technology, Shinshu University, 3-15-1 Tokida, Ueda, Nagano, 386-8567, Japan
- Department of Material-based Medical Engineering, Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University, 2-3-10 Kanda-Surugadai, Chiyoda-ku, Tokyo, 101-006, Japan
| | - Ayana Yamaguchi
- Faculty of Textile Science and Technology, Shinshu University, 3-15-1 Tokida, Ueda, Nagano, 386-8567, Japan
| | - Dan Tanaka
- Faculty of Textile Science and Technology, Shinshu University, 3-15-1 Tokida, Ueda, Nagano, 386-8567, Japan
| | - Yoshihide Hashimoto
- Department of Material-based Medical Engineering, Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University, 2-3-10 Kanda-Surugadai, Chiyoda-ku, Tokyo, 101-006, Japan
- Joining and Welding Research Institute, Osaka University, 11-1, Ibaraki, Osaka, 567-0047, Japan
| | - Yongwei Zhang
- Hangzhou Hopefan Biotechnology Co., Ltd., 199, Jintian Road, Jinnan Street, Linan District, Hangzhou, Zhejiang, 311399, China
| | - Seiichi Funamoto
- Brio Life Science, Inc., 503, 1-26-12, Takadano-baba, Shinjyuku-ku, Tokyo, 169-0075, Japan
| |
Collapse
|
14
|
Sturgess W, Packirisamy S, Geneidy R, Nordenfelt P, Swaminathan V. ECM-dependent regulation of septin 7 in focal adhesions promotes mechanosensing and functional response in fibroblasts. iScience 2024; 27:111355. [PMID: 39650732 PMCID: PMC11625310 DOI: 10.1016/j.isci.2024.111355] [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: 02/14/2024] [Revised: 06/29/2024] [Accepted: 11/06/2024] [Indexed: 12/11/2024] Open
Abstract
Fibroblasts are adherent cells that maintain tissue homeostasis by sensing and responding to the extracellular matrix (ECM). Focal adhesions (FAs) link these ECM changes to actomyosin dynamics through changes in its composition, influencing cellular response. Septin-7 (Sept-7) has previously been found in FA proteomics studies and shown to influence ECM sensing. Using total internal reflection microscopy, we found that ECM-mediated integrin activation regulates spatially distinct Sept-7 structures in FAs. In perinuclear regions, ECM binding stabilized Sept-7 bundles at the back of FAs, while in the core of peripheral FAs high integrin activation promoted elongation of Sept-7 structures. Ventral Sept-7 structures were crucial for ECM sensing, impacting region-specific FA elongation, stabilization, and contributing to fibroblast mechanosensitivity. Taken together, our results suggest that ECM and integrin-dependent regulation of ventral Sept-7 structures plays a pivotal role in fibroblast ECM sensing and mechanotransduction through its recruitment and assembly into FA subpopulations.
Collapse
Affiliation(s)
- Wesley Sturgess
- Division of Oncology, Department of Clinical Sciences, Lund University, Lund, Sweden
- Wallenberg Centre for Molecular Medicine, Lund University, Lund, Sweden
| | - Swathi Packirisamy
- Division of Oncology, Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Rodina Geneidy
- Division of Oncology, Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Pontus Nordenfelt
- Division of Infection Medicine, Department of Clinical Sciences, Lund University, Lund, Sweden
- Department of Laboratory Medicine, Clinical Microbiology, Skåne University Hospital Lund, Lund Universty, Lund, Sweden
| | - Vinay Swaminathan
- Division of Oncology, Department of Clinical Sciences, Lund University, Lund, Sweden
- Wallenberg Centre for Molecular Medicine, Lund University, Lund, Sweden
| |
Collapse
|
15
|
Bighi B, Ragazzini G, Gallerani A, Mescola A, Scagliarini C, Zannini C, Marcuzzi M, Olivi E, Cavallini C, Tassinari R, Bianchi M, Corsi L, Ventura C, Alessandrini A. Cell stretching devices integrated with live cell imaging: a powerful approach to study how cells react to mechanical cues. PROGRESS IN BIOMEDICAL ENGINEERING (BRISTOL, ENGLAND) 2024; 7:012005. [PMID: 39655854 DOI: 10.1088/2516-1091/ad9699] [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: 06/29/2024] [Accepted: 11/20/2024] [Indexed: 12/18/2024]
Abstract
Mechanical stimuli have multiple effects on cell behavior, affecting a number of cellular processes including orientation, proliferation or apoptosis, migration and invasion, the production of extracellular matrix proteins, the activation and translocation of transcription factors, the expression of different genes such as those involved in inflammation and the reprogramming of cell fate. The recent development of cell stretching devices has paved the way for the study of cell reactions to stretching stimuliin-vitro, reproducing physiological situations that are experienced by cells in many tissues and related to functions such as breathing, heart beating and digestion. In this work, we review the highly-relevant contributions cell stretching devices can provide in the field of mechanobiology. We then provide the details for the in-house construction and operation of these devices, starting from the systems that we already developed and tested. We also review some examples where cell stretchers can supply meaningful insights into mechanobiology topics and we introduce new results from our exploitation of these devices.
Collapse
Affiliation(s)
- Beatrice Bighi
- Department of Physics, Informatics and Mathematics, University of Modena and Reggio Emilia, via Campi 213/A, 41125 Modena, Italy
- CNR-Nanoscience Institute-S3, via Campi 213/A, 41125 Modena, Italy
| | | | - Alessia Gallerani
- Department of Physics, Informatics and Mathematics, University of Modena and Reggio Emilia, via Campi 213/A, 41125 Modena, Italy
| | - Andrea Mescola
- CNR-Nanoscience Institute-S3, via Campi 213/A, 41125 Modena, Italy
| | - Chiara Scagliarini
- Department of Physics, Informatics and Mathematics, University of Modena and Reggio Emilia, via Campi 213/A, 41125 Modena, Italy
| | - Chiara Zannini
- Eldor Lab, via di Corticella 183, 40128 Bologna, Italy
- National Laboratory of Molecular Biology and Stem Cell Engineering, National Institute of Biostructures and Biosystems (I.N.B.B.), via di Corticella 183, 40128 Bologna, Italy
| | - Martina Marcuzzi
- Department of Medical and Surgical Sciences, University of Bologna, via G. Massarenti 9, Bologna 40138, Italy
| | - Elena Olivi
- Eldor Lab, via di Corticella 183, 40128 Bologna, Italy
| | - Claudia Cavallini
- Eldor Lab, via di Corticella 183, 40128 Bologna, Italy
- National Laboratory of Molecular Biology and Stem Cell Engineering, National Institute of Biostructures and Biosystems (I.N.B.B.), via di Corticella 183, 40128 Bologna, Italy
| | | | - Michele Bianchi
- Department of Life Sciences, University of Modena and Reggio Emilia, Via G. Campi 287, 41125 Modena, Italy
| | - Lorenzo Corsi
- Department of Life Sciences, University of Modena and Reggio Emilia, Via G. Campi 287, 41125 Modena, Italy
| | - Carlo Ventura
- Eldor Lab, via di Corticella 183, 40128 Bologna, Italy
- National Laboratory of Molecular Biology and Stem Cell Engineering, National Institute of Biostructures and Biosystems (I.N.B.B.), via di Corticella 183, 40128 Bologna, Italy
| | - Andrea Alessandrini
- Department of Physics, Informatics and Mathematics, University of Modena and Reggio Emilia, via Campi 213/A, 41125 Modena, Italy
- CNR-Nanoscience Institute-S3, via Campi 213/A, 41125 Modena, Italy
| |
Collapse
|
16
|
Zhou L, Zhang Y, Yi X, Chen Y, Li Y. Advances in proteins, polysaccharides, and composite biomaterials for enhanced wound healing via microenvironment management: A review. Int J Biol Macromol 2024; 282:136788. [PMID: 39490870 DOI: 10.1016/j.ijbiomac.2024.136788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 10/10/2024] [Accepted: 10/20/2024] [Indexed: 11/05/2024]
Abstract
Wound management is crucial yet imposes substantial social and economic burdens on patients and healthcare systems. The recent rapid advancements in biomaterials and manufacturing technology have created favorable conditions for expediting wound healing. This review examines the latest developments in biomacromolecule-based wound dressings, with a particular focus on proteins and polysaccharides, and their role in modulating the wound microenvironment. The importance of extracellular matrix (ECM)-inspired materials, such as hydrogels and biomimetic dressings, is emphasized. Additionally, this review explores the functionalization of wound dressings, emphasizing properties such as hemostatic capabilities, pain relief, antimicrobial activity, and innovative smart functions like electroceuticals and wound condition monitoring. The study integrates discussions on both the macroscopic healing outcomes and the microscopic pathophysiological mechanisms, highlighting recent advances in managing wound environments to expedite healing. Finally, the review critically assesses the challenges associated with the clinical translation of these wound-healing materials in the future.
Collapse
Affiliation(s)
- Lingyan Zhou
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Ying Zhang
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xiaoli Yi
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yining Chen
- Key Laboratory of Leather Chemistry and Engineering (Sichuan University), Ministry of Education, Chengdu 610065, China
| | - Yuwen Li
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
17
|
Jones S, VandenHeuvel S, Luengo Martinez A, Birur R, Burgeson E, Gilbert I, Baker A, Wolf M, Raghavan SA, Rogers S, Cosgriff-Hernandez E. Suspension electrospinning of decellularized extracellular matrix: A new method to preserve bioactivity. Bioact Mater 2024; 41:640-656. [PMID: 39280898 PMCID: PMC11401211 DOI: 10.1016/j.bioactmat.2024.08.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 08/12/2024] [Accepted: 08/14/2024] [Indexed: 09/18/2024] Open
Abstract
Decellularized extracellular matrices (dECM) have strong regenerative potential as tissue engineering scaffolds; however, current clinical options for dECM scaffolds are limited to freeze-drying its native form into sheets. Electrospinning is a versatile scaffold fabrication technique that allows control of macro- and microarchitecture. It remains challenging to electrospin dECM, which has led researchers to either blend it with synthetic materials or use enzymatic digestion to fully solubilize the dECM. Both strategies reduce the innate bioactivity of dECM and limit its regenerative potential. Herein, we developed a new suspension electrospinning method to fabricate a pure dECM fibrous mesh that retains its innate bioactivity. Systematic investigation of suspension parameters was used to identify critical rheological properties required to instill "spinnability," including homogenization, concentration, and particle size. Homogenization enhanced particle interaction to impart the requisite elastic behavior to withstand electrostatic drawing without breaking. A direct correlation between concentration and viscosity was observed that altered fiber morphology; whereas, particle size had minimal impact on suspension properties and fiber morphology. The versatility of this new method was demonstrated by electrospinning dECM with three common decellularization techniques (Abraham, Badylak, Luo) and tissue sources (intestinal submucosa, heart, skin). Bioactivity retention after electrospinning was confirmed using cell proliferation, angiogenesis, and macrophage polarization assays. Collectively, these findings provide a framework for researchers to electrospin dECM for diverse tissue engineering applications.
Collapse
Affiliation(s)
- Sarah Jones
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, 78712, USA
| | - Sabrina VandenHeuvel
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, 77843, USA
| | - Andres Luengo Martinez
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, 78712, USA
| | - Ruchi Birur
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, 78712, USA
| | - Eric Burgeson
- Department of Chemical and Biomolecular Engineering, University of Illinois Urbana-Champaign, Champaign, IL, 61820, USA
| | - Isabelle Gilbert
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, 78712, USA
| | - Aaron Baker
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, 78712, USA
| | - Matthew Wolf
- Cancer Biomaterials Engineering Section, Cancer Innovation Laboratory, Center for Cancer Research, National Cancer Institute, Frederick, MD, 21702, USA
| | - Shreya A Raghavan
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, 77843, USA
| | - Simon Rogers
- Department of Chemical and Biomolecular Engineering, University of Illinois Urbana-Champaign, Champaign, IL, 61820, USA
| | | |
Collapse
|
18
|
Rodríguez-Quesada L, Ramírez-Sánchez K, Formosa-Dague C, Dague E, Sáenz-Arce G, García-González CA, Vásquez-Sancho F, Avendaño-Soto E, Starbird-Pérez R. Evaluation of Conductive Porous Biobased Composites with Tunable Mechanical Properties for Potential Biological Applications. ACS OMEGA 2024; 9:43426-43437. [PMID: 39493987 PMCID: PMC11525745 DOI: 10.1021/acsomega.4c04391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 09/30/2024] [Accepted: 10/04/2024] [Indexed: 11/05/2024]
Abstract
In this work, starch-based porous cryogels with controlled mechanical and electrical properties were prepared for tissue engineering applications. The starch cryogels were formulated using κ-carrageenan, poly(vinyl alcohol) (PVA), and styrylpyridinium-substituted PVA (SbQ) into the composite. A conductive cryogel was polymerized by chemical oxidation of 3,4-ethylenedioxythiophene (EDOT) using iron(III) p-toluenesulfonate as a strategy to control the electrical properties. The physical, thermal, and mechanical properties were evaluated for the obtained composites. Macro- and nanoscale results confirmed the capability of tuning the mechanical properties of the material by the addition of biopolymers in different contents. The presence of κ-carrageenan significantly increased the storage modulus and decreased the damping effect in the formulations. The presence of PVA showed a plasticizing effect in the formulations, confirmed by the buffering effect and an increase in storage modulus. PVA-SBQ improved the mechanical properties by cross-linking. The addition of PEDOT increased the mechanical and electrical properties of the obtained materials.
Collapse
Affiliation(s)
- Laria Rodríguez-Quesada
- Centro
de Investigación en Servicios Químicos y Microbiológicos
(CEQIATEC), Escuela de Química, Instituto
Tecnológico de Costa Rica, Cartago 159-7050, Costa Rica
- Master
Program in Medical Devices Engineering, Instituto Tecnológico de Costa Rica, Cartago 159-7050, Costa Rica
- Departamento
de Física, Facultad de Ciencias Exactas y Naturales, Universidad Nacional, Heredia 86-3000, Costa Rica
| | - Karla Ramírez-Sánchez
- Centro
de Investigación en Servicios Químicos y Microbiológicos
(CEQIATEC), Escuela de Química, Instituto
Tecnológico de Costa Rica, Cartago 159-7050, Costa Rica
| | | | - Etienne Dague
- LAAS-CNRS,
CNRS, Université de Toulouse, 31400Toulouse, France
| | - Giovanni Sáenz-Arce
- Departamento
de Física, Facultad de Ciencias Exactas y Naturales, Universidad Nacional, Heredia 86-3000, Costa Rica
- Centro de
Investigación en Óptica y Nanofísica, Departamento
de Física, Universidad de Murcia, 30100 Murcia, Spain
| | - Carlos A. García-González
- Departamento
de Farmacia y Tecnología Farmacéutica, Facultad de Farmacia, Universidad de Santiago de Compostela, 15782 Santiago
de Compostela, Spain
| | - Fabián Vásquez-Sancho
- Centro
de Investigación en Ciencia e Ingeniería de Materiales
(CICIMA), Universidad de Costa Rica, San José 11501-2060, Costa Rica
- School
of Physics, Universidad de Costa Rica, San José 11501-2060, Costa Rica
| | - Esteban Avendaño-Soto
- Centro
de Investigación en Ciencia e Ingeniería de Materiales
(CICIMA), Universidad de Costa Rica, San José 11501-2060, Costa Rica
- School
of Physics, Universidad de Costa Rica, San José 11501-2060, Costa Rica
| | - Ricardo Starbird-Pérez
- Centro
de Investigación en Servicios Químicos y Microbiológicos
(CEQIATEC), Escuela de Química, Instituto
Tecnológico de Costa Rica, Cartago 159-7050, Costa Rica
| |
Collapse
|
19
|
Lin Y, Shi H, Yang R, Li S, Tang J, Li S. A transcriptomic analysis of incisional hernia based on high-throughput sequencing technology. Hernia 2024; 28:1899-1907. [PMID: 39073735 DOI: 10.1007/s10029-024-03116-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2024] [Accepted: 07/17/2024] [Indexed: 07/30/2024]
Abstract
PURPOSE Incisional hernia is a common postoperative complication; however, few transcriptomic studies have been conducted on it. In this study, we used second-generation high-throughput sequencing to explore the pathogenesis and potential therapeutic targets of incisional hernias. METHODS Superficial fasciae were collected from 15 patients without hernia and 21 patients with an incisional hernia. High-throughput sequencing of the fascia was performed to generate an expression matrix. We analyzed the matrix to identify differentially expressed genes (DEGs) and performed gene ontology and enrichment analyses of these DEGs. Additionally, an external dataset was utilized to identify key DEGs. RESULTS We identified 1,823 DEGs closely associated with extracellular matrix (ECM) imbalance, bacterial inflammatory response, and fibrillar collagen trimerization. TNNT3, CMAY5, ATP1B4, ASB5, CILP, SIX4, FBN1 and FNDC5 were identified as key DEGs at the intersection of the two expression matrices. Moreover, non-alcoholic fatty liver disease-related, TNF, and IL-17 signaling pathways were identified as key enrichment pathways. CONCLUSIONS We identified eight key DEGs and three pathways associated with incisional hernias. Our findings offer new insights into the pathogenesis of incisional hernias and highlight potential targets for their prevention and treatment.
Collapse
Affiliation(s)
- Yiming Lin
- Department of General Surgery, Fudan University Affiliated Huadong Hosptial, No.221, West Yan'an Road, Jing'an District, Shanghai, 200040, Republic of China
| | - Hekai Shi
- Department of General Surgery, Fudan University Affiliated Huadong Hosptial, No.221, West Yan'an Road, Jing'an District, Shanghai, 200040, Republic of China
| | - Rongduo Yang
- Department of General Surgery, Fudan University Affiliated Huadong Hosptial, No.221, West Yan'an Road, Jing'an District, Shanghai, 200040, Republic of China
| | - Shaochun Li
- Department of General Surgery, Fudan University Affiliated Huadong Hosptial, No.221, West Yan'an Road, Jing'an District, Shanghai, 200040, Republic of China
| | - Jianxiong Tang
- Department of General Surgery, Fudan University Affiliated Huadong Hosptial, No.221, West Yan'an Road, Jing'an District, Shanghai, 200040, Republic of China
| | - Shaojie Li
- Department of General Surgery, Fudan University Affiliated Huadong Hosptial, No.221, West Yan'an Road, Jing'an District, Shanghai, 200040, Republic of China.
| |
Collapse
|
20
|
Bhattacharya T, Kumari M, Kaur K, Kaity S, Arumugam S, Ravichandiran V, Roy S. Decellularized extracellular matrix-based bioengineered 3D breast cancer scaffolds for personalized therapy and drug screening. J Mater Chem B 2024; 12:8843-8867. [PMID: 39162395 DOI: 10.1039/d4tb00680a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/21/2024]
Abstract
Breast cancer (BC) is the second deadliest cancer after lung cancer. Similar to all cancers, it is also driven by a 3D microenvironment. The extracellular matrix (ECM) is an essential component of the 3D tumor micro-environment, wherein it functions as a scaffold for cells and provides metabolic support. BC is characterized by alterations in the ECM. Various studies have attempted to mimic BC-specific ECMs using artificial materials, such as Matrigel. Nevertheless, research has proven that naturally derived decellularized extracellular matrices (dECMs) are superior in providing the essential in vivo-like cues needed to mimic a cancer-like environment. Developing in vitro 3-D BC models is not straightforward and requires extensive analysis of the data established by researchers. For the benefit of researchers, in this review, we have tried to highlight all developmental studies that have been conducted by various scientists so far. The analysis of the conclusions drawn from these studies is also discussed. The advantages and drawbacks of the decellularization methods employed for generating BC scaffolds will be covered, and the review will shed light on how dECM scaffolds help develop a BC environment. The later stages of the article will also focus on immunogenicity issues arising from decellularization and the origin of the tissue. Finally, this review will also discuss the biofabrication of matrices, which is the core part of the bioengineering process.
Collapse
Affiliation(s)
- Teeshyo Bhattacharya
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Chunilal Bhawan, 168 Maniktala Main Road, Kolkata, 700054, West Bengal, India.
| | - Mamta Kumari
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Chunilal Bhawan, 168 Maniktala Main Road, Kolkata, 700054, West Bengal, India
| | - Kulwinder Kaur
- School of Pharmacy and Biomolecular Sciences, RCSI University of Medicine a Health Sciences, Dublin, Ireland
- Tissue Engineering Research Group, Department of Anatomy & Regenerative Medicine, RCSI University of Medicine and Health Sciences, Dublin, Ireland
| | - Santanu Kaity
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Chunilal Bhawan, 168 Maniktala Main Road, Kolkata, 700054, West Bengal, India
| | - Somasundaram Arumugam
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Chunilal Bhawan, 168 Maniktala Main Road, Kolkata, 700054, West Bengal, India.
| | - Velayutham Ravichandiran
- Department of Natural Products, National Institute of Pharmaceutical Education and Research (NIPER), Chunilal Bhawan, 168 Maniktala Main Road, Kolkata, 700054, West Bengal, India.
| | - Subhadeep Roy
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Chunilal Bhawan, 168 Maniktala Main Road, Kolkata, 700054, West Bengal, India.
| |
Collapse
|
21
|
Aydin H, Ozcelikkale A, Acar A. Exploiting Matrix Stiffness to Overcome Drug Resistance. ACS Biomater Sci Eng 2024; 10:4682-4700. [PMID: 38967485 PMCID: PMC11322920 DOI: 10.1021/acsbiomaterials.4c00445] [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: 03/06/2024] [Revised: 06/12/2024] [Accepted: 06/17/2024] [Indexed: 07/06/2024]
Abstract
Drug resistance is arguably one of the biggest challenges facing cancer research today. Understanding the underlying mechanisms of drug resistance in tumor progression and metastasis are essential in developing better treatment modalities. Given the matrix stiffness affecting the mechanotransduction capabilities of cancer cells, characterization of the related signal transduction pathways can provide a better understanding for developing novel therapeutic strategies. In this review, we aimed to summarize the recent advancements in tumor matrix biology in parallel to therapeutic approaches targeting matrix stiffness and its consequences in cellular processes in tumor progression and metastasis. The cellular processes governed by signal transduction pathways and their aberrant activation may result in activating the epithelial-to-mesenchymal transition, cancer stemness, and autophagy, which can be attributed to drug resistance. Developing therapeutic strategies to target these cellular processes in cancer biology will offer novel therapeutic approaches to tailor better personalized treatment modalities for clinical studies.
Collapse
Affiliation(s)
- Hakan
Berk Aydin
- Department
of Biological Sciences, Middle East Technical
University, 06800, Ankara, Turkey
| | - Altug Ozcelikkale
- Department
of Mechanical Engineering, Middle East Technical
University, 06800, Ankara, Turkey
- Graduate
Program of Biomedical Engineering, Middle
East Technical University, 06800, Ankara, Turkey
| | - Ahmet Acar
- Department
of Biological Sciences, Middle East Technical
University, 06800, Ankara, Turkey
| |
Collapse
|
22
|
Karabıyık Acar Ö, Bozdağ G, Hacıhasanoğlu E, Tuncer AA, Aysan E, Torun Köse G. Optimizing decellularization protocols for human thyroid tissues: a step towards tissue engineering and transplantation. Biomed Mater 2024; 19:045034. [PMID: 38857607 DOI: 10.1088/1748-605x/ad565e] [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: 01/25/2024] [Accepted: 06/10/2024] [Indexed: 06/12/2024]
Abstract
Hypothyroidism is caused by insufficient stimulation or disruption of the thyroid. However, the drawbacks of thyroid transplantation have led to the search for new treatments. Decellularization allows tissue transplants to maintain their biomimetic structures while preserving cell adhesion, proliferation, and differentiation. This study aimed to decellularize human thyroid tissues using a structure-preserving optimization strategy and present preliminary data on recellularization. Nine methods were used for physical and chemical decellularization. Quantitative and immunohistochemical analyses were performed to investigate the DNA and extracellular matrix components of the tissues. Biomechanical properties were determined by compression test, and cell viability was examined after seeding MDA-T32 papillary thyroid cancer (PTC) cells onto the decellularized tissues. Decellularized tissues exhibited a notable decrease (<50 ng mg-1DNA, except for Groups 2 and 7) compared to the native thyroid tissue. Nonetheless, collagen and glycosaminoglycans were shown to be conserved in all decellularized tissues. Laminin and fibronectin were preserved at comparatively higher levels, and Young's modulus was elevated when decellularization included SDS. It was observed that the strain value in Group 1 (1.63 ± 0.14 MPa) was significantly greater than that in the decellularized tissues between Groups 2-9, ranging from 0.13 ± 0.03-0.72 ± 0.29 MPa. Finally, viability assessment demonstrated that PTC cells within the recellularized tissue groups successfully attached to the 3D scaffolds and sustained metabolic activity throughout the incubation period. We successfully established a decellularization optimization for human thyroid tissues, which has potential applications in tissue engineering and transplantation research. Our next goal is to conduct recellularization using the methods utilized in Group 1 and transplant the primary thyroid follicular cell-seeded tissues into anin vivoanimal model, particularly due to their remarkable 3D structural preservation and cell adhesion-promoting properties.
Collapse
Affiliation(s)
- Özge Karabıyık Acar
- Department of Genetics and Bioengineering, Faculty of Engineering and Natural Sciences, Istanbul Okan University, Istanbul, Türkiye
- Department of Genetics and Bioengineering, Faculty of Engineering, Yeditepe University, Istanbul, Türkiye
| | - Gülnihal Bozdağ
- Department of Genetics and Bioengineering, Faculty of Engineering, Yeditepe University, Istanbul, Türkiye
| | - Ezgi Hacıhasanoğlu
- Department of Pathology, Faculty of Medicine, Yeditepe University, Istanbul, Türkiye
| | - A Alperen Tuncer
- Department of Genetics and Bioengineering, Faculty of Engineering, Yeditepe University, Istanbul, Türkiye
| | - Erhan Aysan
- Department of General Surgery, Faculty of Medicine, Yeditepe University, Istanbul, Türkiye
| | - Gamze Torun Köse
- Department of Genetics and Bioengineering, Faculty of Engineering, Yeditepe University, Istanbul, Türkiye
| |
Collapse
|
23
|
da Silva IGR, Miglino MA, de Souza SS, Buchaim DV, Buchaim RL. Evaluation of Different Decellularization Protocols for Obtaining and Characterizing Canine Cardiac Extracellular Matrix. Biomedicines 2024; 12:1190. [PMID: 38927398 PMCID: PMC11200447 DOI: 10.3390/biomedicines12061190] [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: 04/01/2024] [Revised: 05/09/2024] [Accepted: 05/20/2024] [Indexed: 06/28/2024] Open
Abstract
Cardiovascular diseases are considered the leading cause of mortality globally; even with low mortality in dogs, such diseases are described in the same way in companion animals and humans. This study aimed to devise an effective decellularization protocol for the canine myocardium through the association of physical, chemical, and enzymatic methods, assessing resultant alterations in the myocardial extracellular matrix to obtain a suitable scaffold. Two canine hearts were collected; the samples were sectioned into ±1 cm2 fragments, washed in distilled water and 1× PBS solution, and followed by treatment under four distinct decellularization protocols. Sodium Dodecyl Sulfate (SDS) 1% 7 days + Triton X-100 1% for 48 h (Protocol I); Sodium Dodecyl Sulfate (SDS) 1% 5 days + Triton X-100 1% for 48 h (Protocol II); Trypsin 0.05% for 1 h at 36 °C + freezing -80 °C overnight + Sodium Dodecyl Sulfate (SDS) 1% for 3 days, Triton-X-100 for 48 h hours (Protocol III); 0.05% trypsin for 1 h at 36 °C + freezing at -80 °C overnight + 1% Sodium Dodecyl Sulfate (SDS) for 2 days + 1% Triton-X-100 for 24 h (Protocol IV). After analysis, Protocols I and II showed the removal of cellular content and preservation of extracellular matrix (ECM) contents, unlike Protocols III and IV, which retracted the ECM and removed essential elements of the matrix. In theory, although Protocols I and II have similar results, Protocol II stands out for the preservation of the architecture and components of the extracellular matrix, along with reduced exposure time to reagents, making it the recommended protocol for the development of a canine myocardial scaffold.
Collapse
Affiliation(s)
- Izabela Gabriela Rodrigues da Silva
- Graduate Program in Anatomy of Domestic and Wild Animals, Faculty of Veterinary Medicine and Animal Science, University of Sao Paulo (FMVZ-USP), Sao Paulo 05508-270, Brazil; (I.G.R.d.S.); (M.A.M.); (D.V.B.)
| | - Maria Angelica Miglino
- Graduate Program in Anatomy of Domestic and Wild Animals, Faculty of Veterinary Medicine and Animal Science, University of Sao Paulo (FMVZ-USP), Sao Paulo 05508-270, Brazil; (I.G.R.d.S.); (M.A.M.); (D.V.B.)
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, University of Marilia (UNIMAR), Marilia 17525-902, Brazil
- Postgraduate Program in Animal Health, Production and Environment, University of Marilia (UNIMAR), Marilia 17525-902, Brazil
| | - Samara Silva de Souza
- Graduate Program in Biotechnology (PPGBIOTEC), Federal Technological University of Parana (UTFPR), Campus Dois Vizinhos, Dois Vizinhos 85660-000, Brazil;
| | - Daniela Vieira Buchaim
- Graduate Program in Anatomy of Domestic and Wild Animals, Faculty of Veterinary Medicine and Animal Science, University of Sao Paulo (FMVZ-USP), Sao Paulo 05508-270, Brazil; (I.G.R.d.S.); (M.A.M.); (D.V.B.)
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, University of Marilia (UNIMAR), Marilia 17525-902, Brazil
- Medical School, University Center of Adamantina (UNIFAI), Adamantina 17800-000, Brazil
| | - Rogerio Leone Buchaim
- Graduate Program in Anatomy of Domestic and Wild Animals, Faculty of Veterinary Medicine and Animal Science, University of Sao Paulo (FMVZ-USP), Sao Paulo 05508-270, Brazil; (I.G.R.d.S.); (M.A.M.); (D.V.B.)
- Department of Biological Sciences, Bauru School of Dentistry (FOB-USP), University of Sao Paulo, Bauru 17012-901, Brazil
| |
Collapse
|
24
|
Wolfram L, Gimpel C, Schwämmle M, Clark SJ, Böhringer D, Schlunck G. The impact of substrate stiffness on morphological, transcriptional and functional aspects in RPE. Sci Rep 2024; 14:7488. [PMID: 38553490 PMCID: PMC11344127 DOI: 10.1038/s41598-024-56661-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Accepted: 03/08/2024] [Indexed: 04/02/2024] Open
Abstract
Alterations in the structure and composition of Bruch's membrane (BrM) and loss of retinal pigment epithelial (RPE) cells are associated with various ocular diseases, notably age-related macular degeneration (AMD) as well as several inherited retinal diseases (IRDs). We explored the influence of stiffness as a major BrM characteristic on the RPE transcriptome and morphology. ARPE-19 cells were plated on soft ( E = 30 kPa ) or stiff ( E = 80 kPa ) polyacrylamide gels (PA gels) or standard tissue culture plastic (TCP). Next-generation sequencing (NGS) data on differentially expressed small RNAs (sRNAs) and messenger RNAs (mRNAs) were validated by qPCR, immunofluorescence or western blotting. The microRNA (miRNA) fraction of sRNAs grew with substrate stiffness and distinct miRNAs such as miR-204 or miR-222 were differentially expressed. mRNA targets of differentially expressed miRNAs were stably expressed, suggesting a homeostatic effect of miRNAs. mRNA transcription patterns were substrate stiffness-dependent, including components of Wnt/beta-catenin signaling, Microphthalmia-Associated Transcription Factor (MITF) and Dicer. These findings highlight the relevance of mechanical properties of the extracellular matrix (ECM) in cell culture experiments, especially those focusing on ECM-related diseases, such as AMD.
Collapse
Affiliation(s)
- Lasse Wolfram
- Eye Center, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany.
- Department for Ophthalmology, Institute for Ophthalmic Research, Eberhard Karls University of Tübingen, Tübingen, Germany.
- Department for Ophthalmology, University Eye Clinic, Eberhard Karls University of Tübingen, Tübingen, Germany.
| | - Clara Gimpel
- Eye Center, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Department of Neurology, Schlosspark-Klinik Charlottenburg, Berlin, Germany
| | - Melanie Schwämmle
- Eye Center, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Simon J Clark
- Department for Ophthalmology, Institute for Ophthalmic Research, Eberhard Karls University of Tübingen, Tübingen, Germany
- Department for Ophthalmology, University Eye Clinic, Eberhard Karls University of Tübingen, Tübingen, Germany
- Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, UK
| | - Daniel Böhringer
- Eye Center, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Günther Schlunck
- Eye Center, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany.
| |
Collapse
|
25
|
Boscaro D, Sikorski P. Spheroids as a 3D in vitro model to study bone and bone mineralization. BIOMATERIALS ADVANCES 2024; 157:213727. [PMID: 38101067 DOI: 10.1016/j.bioadv.2023.213727] [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: 10/10/2023] [Revised: 11/23/2023] [Accepted: 12/06/2023] [Indexed: 12/17/2023]
Abstract
Traumas, fractures, and diseases can severely influence bone tissue. Insight into bone mineralization is essential for the development of therapies and new strategies to enhance bone regeneration. 3D cell culture systems, in particular cellular spheroids, have gained a lot of interest as they can recapitulate crucial aspects of the in vivo tissue microenvironment, such as the extensive cell-cell and cell-extracellular matrix (ECM) interactions found in tissue. The potential of combining spheroids and various classes of biomaterials opens also new opportunities for research within bone tissue engineering. Characterizing cellular organization, ECM structure, and ECM mineralization is a fundamental step for understanding the biological processes involved in bone tissue formation in a spheroid-based model system. Still, many experimental techniques used in this field of research are optimized for use with monolayer cell cultures. There is thus a need to develop new and improving existing experimental techniques, for applications in 3D cell culture systems. In this review, bone composition and spheroids properties are described. This is followed by an insight into the techniques that are currently used in bone spheroids research and how these can be used to study bone mineralization. We discuss the application of staining techniques used with optical and confocal fluorescence microscopy, molecular biology techniques, second harmonic imaging microscopy, Raman spectroscopy and microscopy, as well as electron microscopy-based techniques, to evaluate osteogenic differentiation, collagen production and mineral deposition. Challenges in the applications of these methods in bone regeneration and bone tissue engineering are described. STATEMENT OF SIGNIFICANCE: 3D cell cultures have gained a lot of interest in the last decades as a possible technique that can be used to recreate in vitro in vivo biological process. The importance of 3D environment during bone mineralization led scientists to use this cell culture to study this biological process, to obtain a better understanding of the events involved. New and improved techniques are also required for a proper analysis of this cell model and the process under investigation. This review summarizes the state of the art of the techniques used to study bone mineralization and how 3D cell cultures, in particular spheroids, are tested and analysed to obtain better resolved results related to this complex biological process.
Collapse
Affiliation(s)
- Diamante Boscaro
- Department of Physics, Norwegian University of Science and Technology (NTNU), Høgskoleringen 5, Trondheim 7034, Norway.
| | - Pawel Sikorski
- Department of Physics, Norwegian University of Science and Technology (NTNU), Høgskoleringen 5, Trondheim 7034, Norway.
| |
Collapse
|
26
|
Zhao D, Saiding Q, Li Y, Tang Y, Cui W. Bone Organoids: Recent Advances and Future Challenges. Adv Healthc Mater 2024; 13:e2302088. [PMID: 38079529 DOI: 10.1002/adhm.202302088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 11/23/2023] [Indexed: 12/21/2023]
Abstract
Bone defects stemming from tumorous growths, traumatic events, and diverse conditions present a profound conundrum in clinical practice and research. While bone has the inherent ability to regenerate, substantial bone anomalies require bone regeneration techniques. Bone organoids represent a new concept in this field, involving the 3D self-assembly of bone-associated stem cells guided in vitro with or without extracellular matrix material, resulting in a tissue that mimics the structural, functional, and genetic properties of native bone tissue. Within the scientific panorama, bone organoids ascend to an esteemed status, securing significant experimental endorsement. Through a synthesis of current literature and pioneering studies, this review offers a comprehensive survey of the bone organoid paradigm, delves into the quintessential architecture and ontogeny of bone, and highlights the latest progress in bone organoid fabrication. Further, existing challenges and prospective directions for future research are identified, advocating for interdisciplinary collaboration to fully harness the potential of this burgeoning domain. Conclusively, as bone organoid technology continues to mature, its implications for both clinical and research landscapes are poised to be profound.
Collapse
Affiliation(s)
- Ding Zhao
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, P. R. China
| | - Qimanguli Saiding
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, P. R. China
| | - Yihan Li
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, P. R. China
| | - Yunkai Tang
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, P. R. China
| | - Wenguo Cui
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, P. R. China
| |
Collapse
|
27
|
Guglielmotti V, Fuhry E, Neubert TJ, Kuhl M, Pallarola D, Balasubramanian K. Real-Time Monitoring of Cell Adhesion onto a Soft Substrate by a Graphene Impedance Biosensor. ACS Sens 2024; 9:101-109. [PMID: 38141037 DOI: 10.1021/acssensors.3c01705] [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: 12/24/2023]
Abstract
Soft substrates are interesting for many applications, ranging from mimicking the cellular microenvironment to implants. Conductive electrodes on such substrates allow the realization of flexible, elastic, and transparent sensors. Single-layer graphene as a candidate for such electrodes brings the advantage that the active area of the sensor is transparent and conformal to the underlying substrate. Here, we overcome several challenges facing the routine realization of graphene cell sensors on a canonical soft substrate, namely, poly(dimethylsiloxane) (PDMS). We have systematically studied the effect of surface energy before, during, and after the transfer of graphene. Thus, we have identified a suitable support polymer, optimal substrate (pre)treatment, and an appropriate solvent for the removal of the support. Using this procedure, we can reproducibly obtain stable and intact graphene sensors on a millimeter scale on PDMS, which can withstand continuous measurements in cell culture media for several days. From local nanomechanical measurements, we infer that the softness of the substrate is slightly affected after the graphene transfer. However, we can modulate the stiffness using PDMS with differing compositions. Finally, we show that graphene sensors on PDMS can be successfully used as soft electrodes for real-time monitoring of the cell adhesion kinetics. The routine availability of single-layer graphene electrodes on a soft substrate with tunable stiffness will open a new avenue for studies, where the PDMS-liquid interface is made conducting with minimal alteration of the intrinsic material properties such as softness, flexibility, elasticity, and transparency.
Collapse
Affiliation(s)
- Victoria Guglielmotti
- Department of Chemistry, School of Analytical Sciences Adlershof (SALSA) & IRIS Adlershof, Humboldt-Universität zu Berlin, Berlin 10099, Germany
- Instituto de Nanosistemas, Universidad Nacional de General San Martín, San Martín 1650, Provincia de Buenos Aires, Argentina
| | - Emil Fuhry
- Department of Chemistry, School of Analytical Sciences Adlershof (SALSA) & IRIS Adlershof, Humboldt-Universität zu Berlin, Berlin 10099, Germany
| | - Tilmann J Neubert
- Department of Chemistry, School of Analytical Sciences Adlershof (SALSA) & IRIS Adlershof, Humboldt-Universität zu Berlin, Berlin 10099, Germany
| | - Michel Kuhl
- Department of Chemistry, School of Analytical Sciences Adlershof (SALSA) & IRIS Adlershof, Humboldt-Universität zu Berlin, Berlin 10099, Germany
| | - Diego Pallarola
- Instituto de Nanosistemas, Universidad Nacional de General San Martín, San Martín 1650, Provincia de Buenos Aires, Argentina
| | - Kannan Balasubramanian
- Department of Chemistry, School of Analytical Sciences Adlershof (SALSA) & IRIS Adlershof, Humboldt-Universität zu Berlin, Berlin 10099, Germany
| |
Collapse
|
28
|
Wang J, Sun L, Liu Y, Zhang Y. FIGNL1 Promotes Hepatocellular Carcinoma Formation via Remodeling ECM-receptor Interaction Pathway Mediated by HMMR. Curr Gene Ther 2024; 24:249-263. [PMID: 37929733 PMCID: PMC11071652 DOI: 10.2174/0115665232274223231017052707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 09/02/2023] [Accepted: 09/03/2023] [Indexed: 11/07/2023]
Abstract
BACKGROUND The development of novel biomarkers is crucial for the treatment of HCC. In this study, we investigated a new molecular therapeutic target for HCC. Fidgetin-like 1 (FIGNL1) has been reported to play a vital role in lung adenocarcinoma. However, the potential function of FIGNL1 in HCC is still unknown. OBJECTIVE This study aims to investigate the key regulatory mechanisms of FIGNL1 in the formation of HCC. METHODS The regulatory effect of FIGNL1 on HCC was studied by lentivirus infection. In vitro, the effects of FIGNL1 on the proliferation, migration and apoptosis of cells were investigated by CCK8, colony formation assay, transwell and flow cytometry. Meanwhile, the regulation of FIGNL1 on HCC formation in vivo was studied by subcutaneous transplanted tumors. In addition, using transcriptome sequencing technology, we further explored the specific molecular mechanism of FIGNL1 regulating the formation of HCC. RESULTS Functionally, we demonstrated that FIGNL1 knockdown significantly inhibited HCC cell proliferation, migration and promoted cell apoptosis in vitro. Similarly, the knockdown of FIGNL1 meaningfully weakened hepatocarcinogenesis in nude mice. Transcriptome sequencing revealed that FIGNL1 affected the expression of genes involved in extracellular matrix-receptor (ECM-receptor) interaction pathway, such as hyaluronan mediated motility receptor (HMMR). Further validation found that overexpression of HMMR based on knockdown FIGNL1 can rescue the expression abundance of related genes involved in the ECM-receptor interaction pathway. CONCLUSION Our study revealed that FIGNL1 could modulate the ECM-receptor interaction pathway through the regulation of HMMR, thus regulating the formation of HCC.
Collapse
Affiliation(s)
- Jiabei Wang
- Anhui Province Key Laboratory of Hepatopancreatobiliary Surgery, Department of Hepatobiliary Surgery, Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, 230001, China
| | - Linmao Sun
- Anhui Province Key Laboratory of Hepatopancreatobiliary Surgery, Department of Hepatobiliary Surgery, Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, 230001, China
| | - Yao Liu
- Anhui Province Key Laboratory of Hepatopancreatobiliary Surgery, Department of Hepatobiliary Surgery, Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, 230001, China
| | - Yunguang Zhang
- Anhui Province Key Laboratory of Hepatopancreatobiliary Surgery, Department of Hepatobiliary Surgery, Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, 230001, China
| |
Collapse
|
29
|
Aazmi A, Zhang D, Mazzaglia C, Yu M, Wang Z, Yang H, Huang YYS, Ma L. Biofabrication methods for reconstructing extracellular matrix mimetics. Bioact Mater 2024; 31:475-496. [PMID: 37719085 PMCID: PMC10500422 DOI: 10.1016/j.bioactmat.2023.08.018] [Citation(s) in RCA: 19] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 08/23/2023] [Accepted: 08/24/2023] [Indexed: 09/19/2023] Open
Abstract
In the human body, almost all cells interact with extracellular matrices (ECMs), which have tissue and organ-specific compositions and architectures. These ECMs not only function as cellular scaffolds, providing structural support, but also play a crucial role in dynamically regulating various cellular functions. This comprehensive review delves into the examination of biofabrication strategies used to develop bioactive materials that accurately mimic one or more biophysical and biochemical properties of ECMs. We discuss the potential integration of these ECM-mimics into a range of physiological and pathological in vitro models, enhancing our understanding of cellular behavior and tissue organization. Lastly, we propose future research directions for ECM-mimics in the context of tissue engineering and organ-on-a-chip applications, offering potential advancements in therapeutic approaches and improved patient outcomes.
Collapse
Affiliation(s)
- Abdellah Aazmi
- State Key Laboratory of Fluid Power and Mechatronic Systems, Zhejiang University, Hangzhou, 310058, China
- School of Mechanical Engineering, Zhejiang University, Hangzhou, 310058, China
| | - Duo Zhang
- Department of Engineering, University of Cambridge, Cambridge, United Kingdom
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, Guangdong, 51817, China
| | - Corrado Mazzaglia
- Department of Engineering, University of Cambridge, Cambridge, United Kingdom
| | - Mengfei Yu
- The Affiliated Stomatologic Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, China
| | - Zhen Wang
- Center for Laboratory Medicine, Allergy Center, Department of Transfusion Medicine, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - Huayong Yang
- State Key Laboratory of Fluid Power and Mechatronic Systems, Zhejiang University, Hangzhou, 310058, China
- School of Mechanical Engineering, Zhejiang University, Hangzhou, 310058, China
| | - Yan Yan Shery Huang
- Department of Engineering, University of Cambridge, Cambridge, United Kingdom
| | - Liang Ma
- State Key Laboratory of Fluid Power and Mechatronic Systems, Zhejiang University, Hangzhou, 310058, China
- School of Mechanical Engineering, Zhejiang University, Hangzhou, 310058, China
| |
Collapse
|
30
|
Anjum S, Li T, Saeed M, Ao Q. Exploring polysaccharide and protein-enriched decellularized matrix scaffolds for tendon and ligament repair: A review. Int J Biol Macromol 2024; 254:127891. [PMID: 37931866 DOI: 10.1016/j.ijbiomac.2023.127891] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 10/07/2023] [Accepted: 11/02/2023] [Indexed: 11/08/2023]
Abstract
Tissue engineering (TE) has become a primary research topic for the treatment of diseased or damaged tendon/ligament (T/L) tissue. T/L injuries pose a severe clinical burden worldwide, necessitating the development of effective strategies for T/L repair and tissue regeneration. TE has emerged as a promising strategy for restoring T/L function using decellularized extracellular matrix (dECM)-based scaffolds. dECM scaffolds have gained significant prominence because of their native structure, relatively high bioactivity, low immunogenicity, and ability to function as scaffolds for cell attachment, proliferation, and differentiation, which are difficult to imitate using synthetic materials. Here, we review the recent advances and possible future prospects for the advancement of dECM scaffolds for T/L tissue regeneration. We focus on crucial scaffold properties and functions, as well as various engineering strategies employed for biomaterial design in T/L regeneration. dECM provides both the physical and mechanical microenvironments required by cells to survive and proliferate. Various decellularization methods and sources of allogeneic and xenogeneic dECM in T/L repair and regeneration are critically discussed. Additionally, dECM hydrogels, bio-inks in 3D bioprinting, and nanofibers are briefly explored. Understanding the opportunities and challenges associated with dECM-based scaffold development is crucial for advancing T/L repairs in tissue engineering and regenerative medicine.
Collapse
Affiliation(s)
- Shabnam Anjum
- Department of Tissue Engineering, School of Intelligent Medicine, China Medical University, Shenyang 110122, China; NMPA Key Laboratory for Quality Research and Control of Tissue Regenerative Biomaterial, Institute of Regulatory Science for Medical Device, National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, China
| | - Ting Li
- Department of Laboratory Medicine, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, China
| | - Mohammad Saeed
- Dr. A.P.J Abdul Kalam Technical University, Lucknow 226031, India
| | - Qiang Ao
- Department of Tissue Engineering, School of Intelligent Medicine, China Medical University, Shenyang 110122, China; NMPA Key Laboratory for Quality Research and Control of Tissue Regenerative Biomaterial, Institute of Regulatory Science for Medical Device, National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, China.
| |
Collapse
|
31
|
Lee W. The Cytoskeleton and Its Binding Proteins as Mechanosensors, Transducers, and Functional Regulators of Cells. Int J Mol Sci 2023; 25:172. [PMID: 38203343 PMCID: PMC10779244 DOI: 10.3390/ijms25010172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Accepted: 12/14/2023] [Indexed: 01/12/2024] Open
Abstract
Due to its complement of diverse proteins, such as actin filaments, intermediate filaments, and microtubules, the cytoskeleton is essential not only for structural stability but also for regulating cellular signaling, intracellular transportation, and cell division [...].
Collapse
Affiliation(s)
- Wan Lee
- Department of Biochemistry, Dongguk University College of Medicine, 123 Dongdae-ro, Gyeongju 38066, Republic of Korea; ; Tel.: +82-54-770-2409; Fax: +82-54-770-2447
- Channelopathy Research Center (CRC), Dongguk University College of Medicine, 32 Dongguk-ro, Ilsan Dong-gu, Goyang 10326, Republic of Korea
| |
Collapse
|
32
|
Zhang Z, Wu Y, Xuan Z, Xu H, Yin S, Meng Z. Self-assembly of three-dimensional liver organoids: virtual reconstruction via endocytosed polymer dots for refactoring the fine structure. Biomater Sci 2023; 11:7867-7883. [PMID: 37902572 DOI: 10.1039/d3bm01174g] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2023]
Abstract
In vitro culture of organoids holds considerable promise for the treatment of diseases or the provision of artificial organs. Traditional 2D differentiation from mesenchymal stem cells (MSCs) faces challenges in replicating the development of embryonic organs at the cellular level; conversely, the cultivation of 3D organoids exhibits potential for application. It is crucial for clinicians and technology researchers to acquire insights into organoid tissue differentiation, autonomous morphogenesis, as well as 3D assembly processes in vitro. In this investigation, novel 3D organoids capable of engendering complex liver-like tissues in vitro were cultured, and a class of high-luminance semiconductor polymer dots (Pdots) was employed to monitor the self-assembly process of 3D liver organoid tissues and cellular interaction and migration dynamics. Three-dimensional liver-bud (3D-LB) organoid tissues were derived through the interplay of induced MSCs, Wharton's Jelly, and human umbilical vein endothelial cells (HUVECs), and their structural characteristics were determined during the liver-bud organoid development; ultimately, the co-cultured organoid spatial cellular clusters resembling a truffle were successfully replicated. Utilizing R8-Pdots with remarkable resolution and biocompatibility, the structural elements of functional and vascularized organs derived from liver organoid tissues were adeptly reconstituted, and this investigation shall contribute to a further understanding of human hepato-developmental physiology and liver-disease modeling.
Collapse
Affiliation(s)
- Ze Zhang
- Department of Hepatobiliary-Pancreatic Surgery, China-Japan Union Hospital of Jilin University, Jilin University, No. 126 Xiantai Street, Changchun, Jilin 130000, P. R. China.
| | - Yuyang Wu
- State Key Laboratory of Integrated Optoelectronic, College of Electronic Science and Engineering, Jilin University, No. 2699 Qianjin Street, Changchun, Jilin 130012, P. R. China.
| | - Zhilu Xuan
- Department of Obstetrics & Gynecology, The First Hospital of Jilin University, Changchun, Jilin 130012, P. R. China
| | - Haotian Xu
- Department of Hepatobiliary-Pancreatic Surgery, China-Japan Union Hospital of Jilin University, Jilin University, No. 126 Xiantai Street, Changchun, Jilin 130000, P. R. China.
| | - Shengyan Yin
- State Key Laboratory of Integrated Optoelectronic, College of Electronic Science and Engineering, Jilin University, No. 2699 Qianjin Street, Changchun, Jilin 130012, P. R. China.
| | - Zihui Meng
- Department of Hepatobiliary-Pancreatic Surgery, China-Japan Union Hospital of Jilin University, Jilin University, No. 126 Xiantai Street, Changchun, Jilin 130000, P. R. China.
| |
Collapse
|
33
|
Zhu G, Zhang R, Xie Q, Li P, Wang F, Wang L, Li C. Shish-kebab structure fiber with nano and micro diameter regulate macrophage polarization for anti-inflammatory and bone differentiation. Mater Today Bio 2023; 23:100880. [PMID: 38149017 PMCID: PMC10750111 DOI: 10.1016/j.mtbio.2023.100880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 11/15/2023] [Accepted: 11/22/2023] [Indexed: 12/28/2023] Open
Abstract
Biopolymer grafts often have limited biocompatibility, triggering excessive inflammatory responses similar to foreign bodies. Macrophage phenotype shifts are pivotal in the inflammatory response and graft success. The effects of the morphology and physical attributes of the material itself on macrophage polarization should be the focus. In this study, we prepared electrospun fibers with diverse diameters and formed a shish-kebab (SK) structure on the material surface by solution-induced crystallization, forming electrospun fiber scaffolds with diverse pore sizes and roughness. In vitro cell culture experiments demonstrated that SK structure fibers could regulate macrophage differentiation toward M2 phenotype, and the results of in vitro simulation of in vivo tissue reconstruction by the microenvironment demonstrated that the paracrine role of M2 phenotype macrophages could promote bone marrow mesenchymal stem cells (BMSCs) to differentiate into osteoblasts. In rats implanted with a subcutaneous SK-structured fiber scaffold, the large-pore size and low-stiffness SK fiber scaffolds demonstrated superior immune performance, less macrophage aggregation, and easier differentiation to the anti-inflammatory M2 phenotype. Large pore sizes and low-stiffness SK fiber scaffolds guide the morphological design of biological scaffolds implanted in vivo, which is expected to be an effective strategy for reducing inflammation when applied to graft materials in clinical settings.
Collapse
Affiliation(s)
- Gaowei Zhu
- Key Laboratory of Textile Science & Technology Ministry of Education, College of Textiles, Donghua University, Shanghai, 201620, China
| | - Rongyan Zhang
- Key Laboratory of Textile Science & Technology Ministry of Education, College of Textiles, Donghua University, Shanghai, 201620, China
| | - Qianyang Xie
- Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, and Shanghai Key Laboratory of Stomatology and Shanghai Research Institute of Stomatology, No. 639, Zhizaoju Rd., Shanghai, 200011, China
| | - Peilun Li
- Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, and Shanghai Key Laboratory of Stomatology and Shanghai Research Institute of Stomatology, No. 639, Zhizaoju Rd., Shanghai, 200011, China
| | - Fujun Wang
- Key Laboratory of Textile Science & Technology Ministry of Education, College of Textiles, Donghua University, Shanghai, 201620, China
| | - Lu Wang
- Key Laboratory of Textile Science & Technology Ministry of Education, College of Textiles, Donghua University, Shanghai, 201620, China
| | - Chaojing Li
- Key Laboratory of Textile Science & Technology Ministry of Education, College of Textiles, Donghua University, Shanghai, 201620, China
| |
Collapse
|
34
|
Dawson C, Xu F, Hoare T. Reactive Cell Electrospinning of Anisotropically Aligned and Bilayer Hydrogel Nanofiber Networks. ACS Biomater Sci Eng 2023; 9:6490-6503. [PMID: 37870742 DOI: 10.1021/acsbiomaterials.3c01013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2023]
Abstract
Structured hydrogels that incorporate aligned nanofibrous morphologies have been demonstrated to better replicate the structures of native extracellular matrices and thus their function in guiding cell responses. However, current techniques for nanofiber fabrication are limited in their ability to create hydrogel scaffolds with tunable directional alignments and cell types/densities, as required to reproduce more complex native tissue structures. Herein, we leverage a reactive cell electrospinning technique based on the dynamic covalent cross-linking of poly(ethylene glycol methacrylate (POEGMA) precursor polymers to fabricate aligned hydrogel nanofibers that can be directly loaded with cells during the electrospinning process. The scaffolds were found to support high C2C12 myoblast viabilities greater than 85% over 14 days, with changes in the electrospinning collector allowing for the single-step fabrication of nonaligned, aligned, or cross-aligned nanofibrous networks. Cell aspect ratios on aligned scaffolds were found on average to be 27% higher compared to those on nonaligned scaffolds; furthermore, cell-loaded bilayer scaffolds with perpendicular fiber alignments showed evidence of enabling localized directional cell responses to individual layer fiber directions while avoiding delamination between the layers. This fabrication approach thus offers potential for better mimicking the structure and thus function of aligned and multilayered tissues (e.g., smooth muscle, neural, or tendon tissues).
Collapse
Affiliation(s)
- Chloe Dawson
- Department of Chemical Engineering, McMaster University, 1280 Main St. W., Hamilton, Ontario, Canada L8S 4L7
| | - Fei Xu
- Department of Chemical Engineering, McMaster University, 1280 Main St. W., Hamilton, Ontario, Canada L8S 4L7
| | - Todd Hoare
- Department of Chemical Engineering, McMaster University, 1280 Main St. W., Hamilton, Ontario, Canada L8S 4L7
| |
Collapse
|
35
|
Tai L, Saffery NS, Chin SP, Cheong SK. Secretome profile of TNF-α-induced human umbilical cord mesenchymal stem cells unveils biological processes relevant to skin wound healing. Regen Med 2023; 18:839-856. [PMID: 37671699 DOI: 10.2217/rme-2023-0085] [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: 09/07/2023] Open
Abstract
Aim: To profile and study the proteins responsible for the beneficial effect of the TNF-α-induced human umbilical cord mesenchymal stem cells (hUCMSCs) secretome in wound healing. Methods: The hUCMSCs secretome was generated with (induced) or without (uninduced) TNF-α and was subsequently analyzed by liquid chromatography-mass spectrometry, immunoassay and in vitro scratch assay. Results: Proteomic analysis revealed approximately 260 proteins, including 51 and 55 unique proteins in the induced and uninduced secretomes, respectively. Gene ontology analysis disclosed that differential proteins in the induced secretome mainly involved inflammation-related terms. The induced secretome, consisting of higher levels of FGFb, VEGF, PDGF and IL-6, significantly accelerated wound closure and enhanced MMP-13 secretion in HaCaT keratinocytes. Conclusion: The secretome from induced hUCMSCs includes factors that promote wound closure.
Collapse
Affiliation(s)
- Lihui Tai
- Cytopeutics Sdn Bhd, Suite 2-3 2nd floor, Bio-X Centre, Persiaran Cyberpoint Selatan, Cyber 8, 63000, Cyberjaya, Selangor, Malaysia
| | - Nik Syazana Saffery
- Cytopeutics Sdn Bhd, Suite 2-3 2nd floor, Bio-X Centre, Persiaran Cyberpoint Selatan, Cyber 8, 63000, Cyberjaya, Selangor, Malaysia
| | - Sze Piaw Chin
- Cytopeutics Sdn Bhd, Suite 2-3 2nd floor, Bio-X Centre, Persiaran Cyberpoint Selatan, Cyber 8, 63000, Cyberjaya, Selangor, Malaysia
| | - Soon Keng Cheong
- Cytopeutics Sdn Bhd, Suite 2-3 2nd floor, Bio-X Centre, Persiaran Cyberpoint Selatan, Cyber 8, 63000, Cyberjaya, Selangor, Malaysia
- M. Kandiah Faculty of Medicine & Health Sciences (MK FMHS), Universiti Tunku Abdul Rahman Sungai Long City Campus, Jalan Sungai Long, Bandar Sungai Long, Cheras, 43000, Kajang, Selangor, Malaysia
| |
Collapse
|
36
|
Bakhshandeh B, Sorboni SG, Ranjbar N, Deyhimfar R, Abtahi MS, Izady M, Kazemi N, Noori A, Pennisi CP. Mechanotransduction in tissue engineering: Insights into the interaction of stem cells with biomechanical cues. Exp Cell Res 2023; 431:113766. [PMID: 37678504 DOI: 10.1016/j.yexcr.2023.113766] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 09/01/2023] [Accepted: 09/04/2023] [Indexed: 09/09/2023]
Abstract
Stem cells in their natural microenvironment are exposed to biochemical and biophysical cues emerging from the extracellular matrix (ECM) and neighboring cells. In particular, biomechanical forces modulate stem cell behavior, biological fate, and early developmental processes by sensing, interpreting, and responding through a series of biological processes known as mechanotransduction. Local structural changes in the ECM and mechanics are driven by reciprocal activation of the cell and the ECM itself, as the initial deposition of matrix proteins sequentially affects neighboring cells. Recent studies on stem cell mechanoregulation have provided insight into the importance of biomechanical signals on proper tissue regeneration and function and have shown that precise spatiotemporal control of these signals exists in stem cell niches. Against this background, the aim of this work is to review the current understanding of the molecular basis of mechanotransduction by analyzing how biomechanical forces are converted into biological responses via cellular signaling pathways. In addition, this work provides an overview of advanced strategies using stem cells and biomaterial scaffolds that enable precise spatial and temporal control of mechanical signals and offer great potential for the fields of tissue engineering and regenerative medicine will be presented.
Collapse
Affiliation(s)
- Behnaz Bakhshandeh
- Department of Biotechnology, College of Science, University of Tehran, Tehran, Iran.
| | | | - Nika Ranjbar
- Department of Biotechnology, College of Science, University of Tehran, Tehran, Iran
| | - Roham Deyhimfar
- Department of Microbiology, School of Biology, College of Science, University of Tehran, Tehran, Iran
| | - Maryam Sadat Abtahi
- Department of Biotechnology, School of Chemical Engineering, College of Engineering, University of Tehran, Tehran, Iran
| | - Mehrnaz Izady
- Department of Cellular and Molecular Biology, School of Biology, College of Science, University of Tehran, Tehran, Iran
| | - Navid Kazemi
- Department of Microbiology, School of Biology, College of Science, University of Tehran, Tehran, Iran
| | - Atefeh Noori
- Department of Biotechnology, Iranian Research Organization for Science and Technology (IROST), Tehran, Iran
| | - Cristian Pablo Pennisi
- Regenerative Medicine Group, Department of Health Science and Technology, Aalborg University, Denmark.
| |
Collapse
|
37
|
Zeiringer S, Wiltschko L, Glader C, Reiser M, Absenger-Novak M, Fröhlich E, Roblegg E. Development and Characterization of an In Vitro Intestinal Model Including Extracellular Matrix and Macrovascular Endothelium. Mol Pharm 2023; 20:5173-5184. [PMID: 37677739 PMCID: PMC10548470 DOI: 10.1021/acs.molpharmaceut.3c00532] [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: 06/21/2023] [Revised: 08/25/2023] [Accepted: 08/25/2023] [Indexed: 09/09/2023]
Abstract
In vitro intestinal models are used to study biological processes, drug and food absorption, or cytotoxicity, minimizing the use of animals in the laboratory. They usually consist of enterocytes and mucus-producing cells cultured for 3 weeks, e.g., on Transwells, to obtain a fully differentiated cell layer simulating the human epithelium. Other important components are the extracellular matrix (ECM) and strong vascularization. The former serves as structural support for cells and promotes cellular processes such as differentiation, migration, and growth. The latter includes endothelial cells, which coordinate vascularization and immune cell migration and facilitate the transport of ingested substances or drugs to the liver. In most cases, animal-derived hydrogels such as Matrigel or collagen are used as ECM in in vitro intestinal models, and endothelial cells are only partially considered, if at all. However, it is well-known that animal-derived products can lead to altered cell behavior and incorrect results. To circumvent these limitations, synthetic and modifiable hydrogels (Peptigel and Vitrogel) were studied here to mimic xenofree ECM, and the data were compared with Matrigel. Careful rheological characterization was performed, and the effect on cell proliferation was investigated. The results showed that Vitrogel exhibited shear-thinning behavior with an internal structure recovery of 78.9 ± 11.2%, providing the best properties among the gels investigated. Therefore, a coculture of Caco-2 and HT29-MTX cells (ratio 7:3) was grown on Vitrogel, while simultaneously endothelial cells were cultured on the basolateral side by inverse cultivation. The model was characterized in terms of cell proliferation, differentiation, and drug permeability. It was found that the cells cultured on Vitrogel induced a 1.7-fold increase in cell proliferation and facilitated the formation of microvilli and tight junctions after 2 weeks of cultivation. At the same time, the coculture showed full differentiation indicated by high alkaline phosphatase release of Caco-2 cells (95.0 ± 15.9%) and a mucus layer produced by HT29-MTX cells. Drug tests led to ex vivo comparable permeability coefficients (Papp) (i.e., Papp; antipyrine = (33.64 ± 5.13) × 10-6 cm/s, Papp; atenolol = (0.59 ± 0.16) × 10-6 cm/s). These results indicate that the newly developed intestinal model can be used for rapid and efficient assessment of drug permeability, excluding unexpected results due to animal-derived materials.
Collapse
Affiliation(s)
- Scarlett Zeiringer
- University
of Graz, Institute of Pharmaceutical
Sciences, Pharmaceutical Technology and Biopharmacy, Universitätsplatz 1, 8010 Graz, Austria
| | - Laura Wiltschko
- University
of Graz, Institute of Pharmaceutical
Sciences, Pharmaceutical Technology and Biopharmacy, Universitätsplatz 1, 8010 Graz, Austria
- Joanneum
Research-Health, Neue Stiftingtalstraße 2, 8010 Graz, Austria
| | - Christina Glader
- University
of Graz, Institute of Pharmaceutical
Sciences, Pharmaceutical Technology and Biopharmacy, Universitätsplatz 1, 8010 Graz, Austria
- Research
Center Pharmaceutical Engineering GmbH, Inffeldgasse 13, 8010 Graz, Austria
| | - Martin Reiser
- University
of Graz, Institute of Pharmaceutical
Sciences, Pharmaceutical Technology and Biopharmacy, Universitätsplatz 1, 8010 Graz, Austria
| | - Markus Absenger-Novak
- Center
for Medical Research, Medical University
of Graz, Stiftingtalstraße 24, 8010 Graz, Austria
| | - Eleonore Fröhlich
- Center
for Medical Research, Medical University
of Graz, Stiftingtalstraße 24, 8010 Graz, Austria
- BioTechMed-Graz, Mozartgasse 12/II, 8010 Graz, Austria
| | - Eva Roblegg
- University
of Graz, Institute of Pharmaceutical
Sciences, Pharmaceutical Technology and Biopharmacy, Universitätsplatz 1, 8010 Graz, Austria
- Research
Center Pharmaceutical Engineering GmbH, Inffeldgasse 13, 8010 Graz, Austria
- BioTechMed-Graz, Mozartgasse 12/II, 8010 Graz, Austria
| |
Collapse
|
38
|
Natale BV, Kotadia R, Gustin K, Harihara A, Min S, Kreisman MJ, Breen KM, Natale DR. Extracellular Matrix Influences Gene Expression and Differentiation of Mouse Trophoblast Stem Cells. Stem Cells Dev 2023; 32:622-637. [PMID: 37463089 PMCID: PMC10561768 DOI: 10.1089/scd.2022.0290] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Accepted: 07/14/2023] [Indexed: 07/20/2023] Open
Abstract
Trophoblast stem (TS) cells were first isolated from the mouse placenta; however, little is known about their maintenance and niche in vivo. TS cells, like other stem cells, have a unique microenvironment in which the extracellular matrix (ECM) is a component. Placental pathology is associated with ECM change. However, how these changes and the individual ECM components impact the maintenance or differentiation of TS cells has not been established. This study identified which ECM component(s) maintain the greatest expression of markers associated with undifferentiated mouse trophoblast stem (mTS) cells and which alter the profile of markers of differentiation based on mRNA analysis. mTS cells cultured on individual ECM components and subsequent quantitative polymerase chain reaction analysis revealed that laminin promoted the expression of markers associated with undifferentiated TS cells, fibronectin promoted gene expression associated with syncytiotrophoblast (SynT) layer II cells, and collagen IV promoted the expression of genes associated with differentiated trophoblast. To investigate whether pathological placental ECM influenced the expression of genes associated with different trophoblast subtypes, the mouse model of streptozotocin (STZ)-induced pancreatic β cell ablation and diabetes was used. Female mice administered STZ (blood glucose ≥300 mg/dL) or control (blood glucose ≤150 mg/dL) were mated. Placental pathology at embryonic day (E)14.5 was confirmed with reduced fetal blood space area, reduced expression of the pericyte marker αSMA, and decreased expression of ECM proteins. mTS cells cultured on ECM isolated from STZ placenta were associated with reduced expression of undifferentiated mTS markers and increased expression of genes associated with terminally differentiated trophoblast [Gcm-1 and SynA (SynT) and junctional zone Tpbpa and Prl2c2]. Altogether, these results support the value of using ECM isolated from the placenta as a tool for understanding trophoblast contribution to placental pathology.
Collapse
Affiliation(s)
- Bryony V. Natale
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Diego, La Jolla, California, USA
| | - Ramie Kotadia
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Diego, La Jolla, California, USA
| | - Katarina Gustin
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Diego, La Jolla, California, USA
| | - Anirudha Harihara
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Diego, La Jolla, California, USA
| | - Sarah Min
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Diego, La Jolla, California, USA
| | - Michael J. Kreisman
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Diego, La Jolla, California, USA
| | - Kellie M. Breen
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Diego, La Jolla, California, USA
| | - David R.C. Natale
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Diego, La Jolla, California, USA
| |
Collapse
|
39
|
Wu F, Lei N, Yang S, Zhou J, Chen M, Chen C, Qiu L, Guo R, Li Y, Chang L. Treatment strategies for intrauterine adhesion: focus on the exosomes and hydrogels. Front Bioeng Biotechnol 2023; 11:1264006. [PMID: 37720318 PMCID: PMC10501405 DOI: 10.3389/fbioe.2023.1264006] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Accepted: 08/21/2023] [Indexed: 09/19/2023] Open
Abstract
Intrauterine adhesion (IUA), also referred to as Asherman Syndrome (AS), results from uterine trauma in both pregnant and nonpregnant women. The IUA damages the endometrial bottom layer, causing partial or complete occlusion of the uterine cavity. This leads to irregular menstruation, infertility, or repeated abortions. Transcervical adhesion electroreception (TCRA) is frequently used to treat IUA, which greatly lowers the prevalence of adhesions and increases pregnancy rates. Although surgery aims to disentangle the adhesive tissue, it can exacerbate the development of IUA when the degree of adhesion is severer. Therefore, it is critical to develop innovative therapeutic approaches for the prevention of IUA. Endometrial fibrosis is the essence of IUA, and studies have found that the use of different types of mesenchymal stem cells (MSCs) can reduce the risk of endometrial fibrosis and increase the possibility of pregnancy. Recent research has suggested that exosomes derived from MSCs can overcome the limitations of MSCs, such as immunogenicity and tumorigenicity risks, thereby providing new directions for IUA treatment. Moreover, the hydrogel drug delivery system can significantly ameliorate the recurrence rate of adhesions and the intrauterine pregnancy rate of patients, and its potential mechanism in the treatment of IUA has also been studied. It has been shown that the combination of two or more therapeutic schemes has broader application prospects; therefore, this article reviews the pathophysiology of IUA and current treatment strategies, focusing on exosomes combined with hydrogels in the treatment of IUA. Although the use of exosomes and hydrogels has certain challenges in treating IUA, they still provide new promising directions in this field.
Collapse
Affiliation(s)
- Fengling Wu
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Ningjing Lei
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Shenyu Yang
- Medical 3D Printing Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Junying Zhou
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Mengyu Chen
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Cheng Chen
- Department of Gynaecology and Obstetrics, Chongqing General Hospital, Chongqing, China
| | - Luojie Qiu
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Ruixia Guo
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yong Li
- St George and Sutherland Clinical Campuses, School of Clinical Medicine, UNSW Sydney, Kensington, NSW, Australia
| | - Lei Chang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
40
|
Shuyuan Y, Meimei W, Fenghua L, Huishan Z, Min C, Hongchu B, Xuemei L. hUMSC transplantation restores follicle development in ovary damaged mice via re-establish extracellular matrix (ECM) components. J Ovarian Res 2023; 16:172. [PMID: 37620943 PMCID: PMC10464307 DOI: 10.1186/s13048-023-01217-y] [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: 11/07/2022] [Accepted: 06/20/2023] [Indexed: 08/26/2023] Open
Abstract
OBJECTIVES Explore the therapeutic role of human umbilical mesenchymal stem cells (hUMSCs) transplantation for regeneration of ECM components and restoration of follicular development in mice. BACKGROUND The extracellular matrix (ECM) is crucial to maintain ovary function and regulate follicular development, as it participates in important cell signaling and provides physical support to the cells. However, it is unknown how hUMSCs affect the expression of ECM-related genes in ovaries treated with cyclophosphamide (CTX) and busulfan (BUS). METHODS In the present study, we used 64 six- to eight-week-old ICR female mice to established mouse model. The mice were randomly divided into four groups (n = 16/group): control, POI, POI + hUMSCs, and POI + PBS group. The premature ovarian insufficiency (POI) mouse model was established by intraperitoneal injection of CTX and BUS for 7days, then, hUMSCs or PBS were respectively injected via the tail vein in POI + hUMSCs or POI + PBS group. Another 7days after injection, the mice were sacrificed to harvest the ovary tissue. The ovaries were immediately frozen with liquid nitrogen or fixed with 4% PFA for subsequent experiments. To screen differentially expressed genes (DEGs), we performed transcriptome sequencing of ovaries. Thereafter, a Gene Ontology (GO) terms and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analyses were performed to predict the related biological functions. Retrieval of interacting genes for ECM-related DEGs was performed using the function of STRINGdb (version 2.6.5) to evaluate potential protein-protein interaction (PPI) networks. Furthermore, qRT-PCR and IHC were performed to assess the differential expression of selected DEGs in control, damaged, hUMSCs-transplanted and non-transplanted ovaries. RESULTS Chemotherapy caused mouse ovarian follicular reserve depletion, and hUMSCs transplantation partially restored follicular development. Our results revealed that ECM-receptor interaction and ECM organization were both downregulated in the damaged ovaries. Further investigation showed that ECM-related genes were downregulated in the CTX and BUS treatment group and partially rescued in hUMSCs injection group but not in the PBS group. qRT-PCR and IHC verified the results: collagen IV and laminin gamma 3 were both expressed around follicle regions in normal ovaries, chemotherapy treatment disrupted their expression, and hUMSCs transplantation rescued their localization and expression to some extent. CONCLUSION Our data demonstrated that ECM-related genes participate in the regulation of ovarian reserve, hUMSCs treatment rescued abnormal expression and localization of collagen IV and laminin gamma 3 in the damaged ovaries. The results suggest that hUMSCs transplantation can maintain ECM-stable microenvironments, which is beneficial to follicular development.
Collapse
Affiliation(s)
- Yin Shuyuan
- Reproductive Medicine Centre, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, 264000, Shandong, China
| | - Wang Meimei
- Reproductive Medicine Centre, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, 264000, Shandong, China
| | - Li Fenghua
- Reproductive Medicine Centre, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, 264000, Shandong, China
| | - Zhao Huishan
- Reproductive Medicine Centre, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, 264000, Shandong, China
| | - Chu Min
- Reproductive Medicine Centre, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, 264000, Shandong, China
| | - Bao Hongchu
- Reproductive Medicine Centre, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, 264000, Shandong, China.
| | - Liu Xuemei
- Reproductive Medicine Centre, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, 264000, Shandong, China.
| |
Collapse
|
41
|
Chen W, Wang Y, Gu H, Zhang Y, Chen C, Yu T, Chen T. Molecular characteristics, clinical significance, and immune landscape of extracellular matrix remodeling-associated genes in colorectal cancer. Front Oncol 2023; 13:1109181. [PMID: 37621680 PMCID: PMC10446763 DOI: 10.3389/fonc.2023.1109181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Accepted: 07/24/2023] [Indexed: 08/26/2023] Open
Abstract
Background Extracellular matrix (ECM) remodeling is one of the hallmark events in cancer and has been shown to be closely related to tumor immunity. Immunotherapy has evolved as an important tool to treat various cancers and improve patient prognosis. The positive response to immunotherapy relies on the unique interaction between cancer and the tumor microenvironment (TME). However, the relationship between ECM remodeling and clinical outcomes, immune cell infiltration, and immunotherapy in colorectal cancer (CRC) remains unknown. Methods We systematically evaluated 69 ECM remodeling-associated genes (EAGs) and comprehensively identified interactions between ECM remodeling and prognosis and the immune microenvironment in CRC patients. The EAG_score was used to quantify the subtype of ECM remodeling in patients. We then assessed their value in predicting prognosis and responding to treatment in CRC. Results After elaborating the molecular characteristics of ECM remodeling-related genes in CRC patients, a model consisting of two ECM remodeling-related genes (MEIS2, SLC2A3) was developed for predicting the prognosis of CRC patients, Receiver Operating Characteristic (ROC) and Kaplan-Meier (K-M) analysis verified its reliable predictive ability. Furthermore, we created a highly reliable nomogram to enhance the clinical feasibility of the EAG_score. Significantly differences in TME and immune function, such as macrophages and CD8+ T cells, were observed between high- and low-risk CRC patients. In addition, drug sensitivity is also strongly related to EAG_score. Conclusion Overall, we developed a prognostic model associated with ECM remodeling, provided meaningful clinical implications for immunotherapy, and facilitated individualized treatment for CRC patients. Further studies are needed to reveal the underlying mechanisms of ECM remodeling in CRC.
Collapse
Affiliation(s)
- Wenlong Chen
- Department of Colorectal Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yiwen Wang
- Department of Colorectal Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Haitao Gu
- Department of General Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yi Zhang
- Department of Colorectal Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Cong Chen
- Department of Neurology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Tingting Yu
- Department of Medical Genetics, School of Basic Medical Science, Jiangsu Key Laboratory of Xenotransplantation, Nanjing Medical University, Nanjing, China
| | - Tao Chen
- Department of Colorectal Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
42
|
Spataro S, Guerra C, Cavalli A, Sgrignani J, Sleeman J, Poulain L, Boland A, Scapozza L, Moll S, Prunotto M. CEMIP (HYBID, KIAA1199): structure, function and expression in health and disease. FEBS J 2023; 290:3946-3962. [PMID: 35997767 DOI: 10.1111/febs.16600] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 07/28/2022] [Accepted: 08/17/2022] [Indexed: 12/01/2022]
Abstract
CEMIP (cell migration-inducing protein), also known as KIAA1199 or HYBID, is a protein involved in the depolymerisation of hyaluronic acid (HA), a major glycosaminoglycan component of the extracellular matrix. CEMIP was originally described in patients affected by nonsyndromic hearing loss and has subsequently been shown to play a key role in tumour initiation and progression, as well as arthritis, atherosclerosis and idiopathic pulmonary fibrosis. Despite the vast literature associating CEMIP with these diseases, its biology remains elusive. The present review article summarises all the major scientific evidence regarding its structure, function, role and expression, and attempts to cast light on a protein that modulates EMT, fibrosis and tissue inflammation, an unmet key aspect in several inflammatory disease conditions.
Collapse
Affiliation(s)
- Sofia Spataro
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, Switzerland
| | - Concetta Guerra
- Institute for Research in Biomedicine, Università della Svizzera Italiana, Bellinzona, Switzerland
| | - Andrea Cavalli
- Institute for Research in Biomedicine, Università della Svizzera Italiana, Bellinzona, Switzerland
- Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Jacopo Sgrignani
- Institute for Research in Biomedicine, Università della Svizzera Italiana, Bellinzona, Switzerland
| | - Jonathan Sleeman
- European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- Institute for Biological and Chemical Systems - Biological Information Processing (IBCS - BIP), Karlsruhe Institute for Technology (KIT), Germany
| | - Lina Poulain
- Department of Molecular Biology, University of Geneva, Switzerland
| | - Andreas Boland
- Department of Molecular Biology, University of Geneva, Switzerland
| | - Leonardo Scapozza
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, Switzerland
| | - Solange Moll
- Department of Pathology, University Hospital of Geneva, Switzerland
| | - Marco Prunotto
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, Switzerland
| |
Collapse
|
43
|
Ran M, Ouyang Q, Li X, Hu S, Hu B, Hu J, Dong D, Li L, He H, Liu H, Wang J. Exploring right ovary degeneration in duck and goose embryos by histology and transcriptome dynamics analysis. BMC Genomics 2023; 24:389. [PMID: 37430218 DOI: 10.1186/s12864-023-09493-0] [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: 11/02/2022] [Accepted: 06/29/2023] [Indexed: 07/12/2023] Open
Abstract
BACKGROUND The development of asymmetric chick gonads involves separate developmental programs in the left and right gonads. In contrast to the left ovary developing into a fully functional reproductive organ, the right ovary undergoes gradual degeneration. However, the molecular mechanisms underlying the the degeneration of the right ovary remain incompletely understood. In the present study, we investigated the histomorphological and transcriptomic changes in the right ovary of ducks and geese during the the embryonic stage up to post-hatching day 1. RESULT Hematoxylin-eosin stainings revealed that the right ovary developed until embryonic day 20 in ducks (DE20) or embryonic day 22 in geese (GE22), after which it started to regress. Further RNA-seq analyses revealed that both the differentially expressed genes (DEGs) in ducks and geese right ovary developmental stage were significantly enriched in cell adhesion-related pathway (ECM-receptor interaction, Focal adhesion pathway) and Cellular senescence pathway. Then during the degeneration stage, the DEGs were primarily enriched in pathways associated with inflammation, including Herpes simplex virus 1 infection, Influenza A, and Toll-like receptor signaling pathway. Moreover, duck-specific DEGs showed enrichment in Steroid hormone biosynthesis, Base excision repair, and the Wnt signaling pathway, while geese-specifically DEGs were found to be enriched in apoptosis and inflammation-related pathways, such as Ferroptosis, Necroptosis, RIG-I-like receptor signaling pathway, and NOD-like receptor signaling pathway. These findings suggest that the degeneration process of the right ovary in ducks occurs at a slower pace compared to that in geese. Additionally, the observation of the left ovary of the geese varying degeneration rates in the right ovary after hatching indicated that the development of the left ovary may be influenced by the degeneration of the right ovary. CONCLUSION The data presented in this study provide valuable insights into the dynamic changes in histological structure and transcriptome during the degeneration of the right ovary in ducks and geese. In addition, through the analysis of shared characteristics in the degeneration process of the right ovary in both ducks and geese, we have uncovered the patterns of degradation and elucidated the molecular mechanisms involved in the regression of the right ovary in poultry. Furthermore, we have also made initial discoveries regarding the relationship between the degeneration of the right ovary and the development of the left ovary.
Collapse
Affiliation(s)
- Mingxia Ran
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, 611130, China
| | - Qingyuan Ouyang
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, 611130, China
| | - Xuejian Li
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, 611130, China
| | - Shenqiang Hu
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, 611130, China
| | - Bo Hu
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, 611130, China
| | - Jiwei Hu
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, 611130, China
| | - Dan Dong
- Yucheng District Agriculture and Rural Affairs Bureau, Ya'an, 625000, China
| | - Liang Li
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, 611130, China
| | - Hua He
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, 611130, China
| | - Hehe Liu
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, 611130, China
| | - Jiwen Wang
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, 611130, China.
| |
Collapse
|
44
|
Choi K, Park CY, Choi JS, Kim YJ, Chung S, Lee S, Kim CH, Park SJ. The Effect of the Mechanical Properties of the 3D Printed Gelatin/Hyaluronic Acid Scaffolds on hMSCs Differentiation Towards Chondrogenesis. Tissue Eng Regen Med 2023; 20:593-605. [PMID: 37195569 PMCID: PMC10313889 DOI: 10.1007/s13770-023-00545-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 03/22/2023] [Accepted: 04/09/2023] [Indexed: 05/18/2023] Open
Abstract
BACKGROUND Tissue engineering, including 3D bioprinting, holds great promise as a therapeutic tool for repairing cartilage defects. Mesenchymal stem cells have the potential to treat various fields due to their ability to differentiate into different cell types. The biomimetic substrate, such as scaffolds and hydrogels, is a crucial factor that affects cell behavior, and the mechanical properties of the substrate have been shown to impact differentiation during incubation. In this study, we examine the effect of the mechanical properties of the 3D printed scaffolds, made using different concentrations of cross-linker, on hMSCs differentiation towards chondrogenesis. METHODS The 3D scaffold was fabricated using 3D bioprinting technology with gelatin/hyaluronic acid (HyA) biomaterial ink. Crosslinking was achieved by using different concentrations of 4-(4,6-dimethoxy-1,3,5-triazin-2-yl)-4-methlymorpholinium chloride n-hydrate (DMTMM), allowing for control of the scaffold's mechanical properties. The printability and stability were also evaluated based on the concentration of DMTMM used. The effects of the gelatin/HyA scaffold on chondrogenic differentiation was analyzed by utilizing various concentrations of DMTMM. RESULTS The addition of HyA was found to improve the printability and stability of 3D printed gelatin/HyA scaffolds. The mechanical properties of the 3D gelatin/HyA scaffold could be regulated through the use of different concentrations of DMTMM cross-linker. In particular, the use of 0.25 mM DMTMM for crosslinking the 3D gelatin/HyA scaffold resulted in enhanced chondrocyte differentiation. CONCLUSION The mechanical properties of 3D printed gelatin/HyA scaffolds cross-linked using various concentrations of DMTMM can influence the differentiation of hMSCs into chondrocytes.
Collapse
Affiliation(s)
- Kyoung Choi
- Laboratory of Tissue Engineering, Korea Institute of Radiological and Medical Sciences, Seoul, 01812, Republic of Korea
- Program in Biomicro System Technology, Korea University, Seoul, 02841, Republic of Korea
| | - Cho Young Park
- Laboratory of Tissue Engineering, Korea Institute of Radiological and Medical Sciences, Seoul, 01812, Republic of Korea
- Program in Biomicro System Technology, Korea University, Seoul, 02841, Republic of Korea
| | - Jun Shik Choi
- Laboratory of Tissue Engineering, Korea Institute of Radiological and Medical Sciences, Seoul, 01812, Republic of Korea
| | - Young-Jin Kim
- Department of Biomedical Engineering, Catholic University of Daegu, Gyeongsan-Si, 38430, Republic of Korea
| | - Seok Chung
- Program in Biomicro System Technology, Korea University, Seoul, 02841, Republic of Korea
| | - Sanghoon Lee
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Korea University College of Medicine, Seoul, 02841, Republic of Korea
| | - Chun-Ho Kim
- Laboratory of Tissue Engineering, Korea Institute of Radiological and Medical Sciences, Seoul, 01812, Republic of Korea.
| | - Sang Jun Park
- Laboratory of Tissue Engineering, Korea Institute of Radiological and Medical Sciences, Seoul, 01812, Republic of Korea.
| |
Collapse
|
45
|
Plath AMS, Huber S, Alfarano SR, Abbott DF, Hu M, Mougel V, Isa L, Ferguson SJ. Co-Electrospun Poly(ε-Caprolactone)/Zein Articular Cartilage Scaffolds. Bioengineering (Basel) 2023; 10:771. [PMID: 37508797 PMCID: PMC10376865 DOI: 10.3390/bioengineering10070771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2023] [Revised: 06/19/2023] [Accepted: 06/21/2023] [Indexed: 07/30/2023] Open
Abstract
Osteoarthritis scaffold-based grafts fail because of poor integration with the surrounding soft tissue and inadequate tribological properties. To circumvent this, we propose electrospun poly(ε-caprolactone)/zein-based scaffolds owing to their biomimetic capabilities. The scaffold surfaces were characterized using Fourier-transform infrared spectroscopy, X-ray photoelectron spectroscopy, static water contact angles, and profilometry. Scaffold biocompatibility properties were assessed by measuring protein adsorption (Bicinchoninic Acid Assay), cell spreading (stained F-actin), and metabolic activity (PrestoBlue™ Cell Viability Reagent) of primary bovine chondrocytes. The data show that zein surface segregation in the membranes not only completely changed the hydrophobic behavior of the materials, but also increased the cell yield and metabolic activity on the scaffolds. The surface segregation is verified by the infrared peak at 1658 cm-1, along with the presence and increase in N1 content in the survey XPS. This observation could explain the decrease in the water contact angles from 125° to approximately 60° in zein-comprised materials and the decrease in the protein adsorption of both bovine serum albumin and synovial fluid by half. Surface nano roughness in the PCL/zein samples additionally benefited the radial spreading of bovine chondrocytes. This study showed that co-electrospun PCL/zein scaffolds have promising surface and biocompatibility properties for use in articular-tissue-engineering applications.
Collapse
Affiliation(s)
| | - Stephanie Huber
- Laboratory for Orthopaedic Technology, ETH Zurich, 8092 Zurich, Switzerland
| | - Serena R Alfarano
- Laboratory of Food and Soft Materials, ETH Zurich, 8092 Zurich, Switzerland
| | - Daniel F Abbott
- Laboratory of Inorganic Chemistry, ETH Zurich, 8093 Zurich, Switzerland
| | - Minghan Hu
- Laboratory for Soft Materials and Interfaces, ETH Zurich, 8093 Zurich, Switzerland
| | - Victor Mougel
- Laboratory of Inorganic Chemistry, ETH Zurich, 8093 Zurich, Switzerland
| | - Lucio Isa
- Laboratory for Soft Materials and Interfaces, ETH Zurich, 8093 Zurich, Switzerland
| | - Stephen J Ferguson
- Laboratory for Orthopaedic Technology, ETH Zurich, 8092 Zurich, Switzerland
| |
Collapse
|
46
|
Niemczyk-Soczynska B, Kolbuk D, Mikulowski G, Ciechomska IA, Sajkiewicz P. Methylcellulose/agarose hydrogel loaded with short electrospun PLLA/laminin fibers as an injectable scaffold for tissue engineering/3D cell culture model for tumour therapies. RSC Adv 2023; 13:11889-11902. [PMID: 37077262 PMCID: PMC10107725 DOI: 10.1039/d3ra00851g] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Accepted: 04/10/2023] [Indexed: 04/21/2023] Open
Abstract
This research aimed at designing and fabricating a smart thermosensitive injectable methylcellulose/agarose hydrogel system loaded with short electrospun bioactive PLLA/laminin fibers as a scaffold for tissue engineering applications or 3D cell culture models. Considering ECM-mimicking morphology and chemical composition, such a scaffold is capable of ensuring a hospitable environment for cell adhesion, proliferation, and differentiation. Its viscoelastic properties are beneficial from the practical perspective of minimally invasive materials that are introduced to the body via injection. Viscosity studies showed the shear-thinning character of MC/AGR hydrogels enabling the potential injection ability of highly viscous materials. Injectability tests showed that by tuning the injection rate, even a high amount of short fibers loaded inside of hydrogel could be efficiently injected into the tissue. Biological studies showed the non-toxic character of composite material with excellent viability, attachment, spreading, and proliferation of fibroblasts and glioma cells. These findings indicate that MC/AGR hydrogel loaded with short PLLA/laminin fibers is a promising biomaterial for both tissue engineering applications and 3D tumor culture models.
Collapse
Affiliation(s)
- Beata Niemczyk-Soczynska
- Institute of Fundamental Technological Research, Polish Academy of Sciences Pawinskiego 5b St. 02-106 Warsaw Poland
| | - Dorota Kolbuk
- Institute of Fundamental Technological Research, Polish Academy of Sciences Pawinskiego 5b St. 02-106 Warsaw Poland
| | - Grzegorz Mikulowski
- Institute of Fundamental Technological Research, Polish Academy of Sciences Pawinskiego 5b St. 02-106 Warsaw Poland
| | - Iwona A Ciechomska
- Nencki Institute of Experimental Biology PAS 3 Pasteur Street 02-093 Warsaw Poland
| | - Pawel Sajkiewicz
- Institute of Fundamental Technological Research, Polish Academy of Sciences Pawinskiego 5b St. 02-106 Warsaw Poland
| |
Collapse
|
47
|
Lee K, Jackson A, John N, Zhang R, Ozhava D, Bhatia M, Mao Y. Bovine Fibroblast-Derived Extracellular Matrix Promotes the Growth and Preserves the Stemness of Bovine Stromal Cells during In Vitro Expansion. J Funct Biomater 2023; 14:jfb14040218. [PMID: 37103308 PMCID: PMC10144935 DOI: 10.3390/jfb14040218] [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: 03/24/2023] [Revised: 04/06/2023] [Accepted: 04/11/2023] [Indexed: 04/28/2023] Open
Abstract
Cultivated meat is a fast-growing research field and an industry with great potential to overcome the limitations of traditional meat production. Cultivated meat utilizes cell culture and tissue engineering technologies to culture a vast number of cells in vitro and grow/assemble them into structures mimicking the muscle tissues of livestock animals. Stem cells with self-renewal and lineage-specific differentiation abilities have been considered one of the key cell sources for cultivated meats. However, the extensive in vitro culturing/expansion of stem cells results in a reduction in their abilities to proliferate and differentiate. Extracellular matrix (ECM) has been used as a culturing substrate to support cell expansion for cell-based therapies in regenerative medicine due to its resemblance to the native microenvironment of cells. In this study, the effect of the ECM on the expansion of bovine umbilical cord stromal cells (BUSC) in vitro was evaluated and characterized. BUSCs with multi-lineage differentiation potentials were isolated from bovine placental tissue. Decellularized ECM prepared from a confluent monolayer of bovine fibroblasts (BF) is free of cellular components but contains major ECM proteins such as fibronectin and type I collagen and ECM-associated growth factors. Expansion of BUSC on ECM for three passages (around three weeks) resulted in about 500-fold amplification, while cells were amplified less than 10-fold when cultured on standard tissue culture plates (TCP). Moreover, the presence of ECM reduced the requirement for serum in the culture medium. Importantly, the cells amplified on ECM retained their differentiation abilities better than cells cultured on TCP. The results of our study support the notion that monolayer cell-derived ECM may be a strategy to expand bovine cells in vitro effectively and efficiently.
Collapse
Affiliation(s)
- Kathleen Lee
- Laboratory for Biomaterials Research, Department of Chemistry and Chemical Biology, Rutgers University, 145 Bevier Rd., Piscataway, NJ 08854, USA
| | - Anisha Jackson
- Laboratory for Biomaterials Research, Department of Chemistry and Chemical Biology, Rutgers University, 145 Bevier Rd., Piscataway, NJ 08854, USA
| | - Nikita John
- Laboratory for Biomaterials Research, Department of Chemistry and Chemical Biology, Rutgers University, 145 Bevier Rd., Piscataway, NJ 08854, USA
| | - Ryan Zhang
- Laboratory for Biomaterials Research, Department of Chemistry and Chemical Biology, Rutgers University, 145 Bevier Rd., Piscataway, NJ 08854, USA
| | - Derya Ozhava
- Laboratory for Biomaterials Research, Department of Chemistry and Chemical Biology, Rutgers University, 145 Bevier Rd., Piscataway, NJ 08854, USA
| | - Mohit Bhatia
- Atelier Meats, 666 Burrard Street, Suite 500, Vancouver, BC V6C 3P6, Canada
| | - Yong Mao
- Laboratory for Biomaterials Research, Department of Chemistry and Chemical Biology, Rutgers University, 145 Bevier Rd., Piscataway, NJ 08854, USA
| |
Collapse
|
48
|
Jones CL, Penney BT, Theodossiou SK. Engineering Cell-ECM-Material Interactions for Musculoskeletal Regeneration. Bioengineering (Basel) 2023; 10:bioengineering10040453. [PMID: 37106640 PMCID: PMC10135874 DOI: 10.3390/bioengineering10040453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Revised: 03/23/2023] [Accepted: 03/28/2023] [Indexed: 04/29/2023] Open
Abstract
The extracellular microenvironment regulates many of the mechanical and biochemical cues that direct musculoskeletal development and are involved in musculoskeletal disease. The extracellular matrix (ECM) is a main component of this microenvironment. Tissue engineered approaches towards regenerating muscle, cartilage, tendon, and bone target the ECM because it supplies critical signals for regenerating musculoskeletal tissues. Engineered ECM-material scaffolds that mimic key mechanical and biochemical components of the ECM are of particular interest in musculoskeletal tissue engineering. Such materials are biocompatible, can be fabricated to have desirable mechanical and biochemical properties, and can be further chemically or genetically modified to support cell differentiation or halt degenerative disease progression. In this review, we survey how engineered approaches using natural and ECM-derived materials and scaffold systems can harness the unique characteristics of the ECM to support musculoskeletal tissue regeneration, with a focus on skeletal muscle, cartilage, tendon, and bone. We summarize the strengths of current approaches and look towards a future of materials and culture systems with engineered and highly tailored cell-ECM-material interactions to drive musculoskeletal tissue restoration. The works highlighted in this review strongly support the continued exploration of ECM and other engineered materials as tools to control cell fate and make large-scale musculoskeletal regeneration a reality.
Collapse
Affiliation(s)
- Calvin L Jones
- Department of Mechanical and Biomedical Engineering, Boise State University, 1910 University Dr MS2085, Boise, ID 83725, USA
| | - Brian T Penney
- Department of Mechanical and Biomedical Engineering, Boise State University, 1910 University Dr MS2085, Boise, ID 83725, USA
| | - Sophia K Theodossiou
- Department of Mechanical and Biomedical Engineering, Boise State University, 1910 University Dr MS2085, Boise, ID 83725, USA
| |
Collapse
|
49
|
Ma B, Yu R. Pan-cancer analysis of ADAMs: A promising biomarker for prognosis and response to chemotherapy and immunotherapy. Front Genet 2023; 14:1105900. [PMID: 37082201 PMCID: PMC10110990 DOI: 10.3389/fgene.2023.1105900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Accepted: 03/20/2023] [Indexed: 04/07/2023] Open
Abstract
Background: Members of a disintegrin and metalloproteinase (ADAM) family play a vital role in cancer development. However, a comprehensive analysis of the landscape of the ADAM family in pan-cancer remains to be performed.Methods: The correlation of the expression level and prognostic value with ADAMs in a pan-cancer cohort and the relationship between ADAMs and the stemness score, tumour microenvironment (TME), chemotherapy-related drug sensitivity, immune subtype, and immunotherapy outcome were investigated.Results: ADAMs were differentially expressed between tumour and para-carcinoma tissues in the pan-cancer cohort, and the expression of ADAMs was significantly correlated with patient prognosis. Furthermore, ADAMs were significantly correlated with the stromal score and immune score based on the TME analysis. Additionally, ADAMs were also correlated with DNAss and RNAss in the pan-cancer cohort. On investigating the CellMiner database, ADAMs were revealed to be significantly correlated with the sensitivity of various drugs, including raloxifene and tamoxifen. Moreover, in the IMvigor210 and GSE78220 cohorts, ADAMs were correlated with immunotherapy response and immune activation genes. Furthermore, quantitative real-time polymerase chain reaction (qRT-PCR) and immunohistochemistry (IHC) were utilised to determine the differential level of ADAM9 in cancer and para-carcinoma tissues in patients’ samples.Conclusion: This study elucidates the importance of ADAMs in cancer progression and lays a foundation for further exploration of ADAMs as potential pan-cancer targets.
Collapse
Affiliation(s)
- Bo Ma
- *Correspondence: Bo Ma, ; Riyue Yu,
| | - Riyue Yu
- *Correspondence: Bo Ma, ; Riyue Yu,
| |
Collapse
|
50
|
Park J, Nguyen TTC, Lee SJ, Wang S, Heo D, Kang DH, Tipan-Quishpe A, Lee WJ, Lee J, Yang SY, Yoon MH. Instant formation of horizontally ordered nanofibrous hydrogel films and direct investigation of peculiar neuronal cell behaviors atop. Biomater Res 2023; 27:19. [PMID: 36907873 PMCID: PMC10009932 DOI: 10.1186/s40824-023-00344-3] [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: 11/08/2022] [Accepted: 01/25/2023] [Indexed: 03/14/2023] Open
Abstract
BACKGROUND Hydrogels have been widely used in many research fields owing to optical transparency, good biocompatibility, tunable mechanical properties, etc. Unlike typical hydrogels in the form of an unstructured bulk material, we developed aqueous dispersions of fiber-shaped hydrogel structures with high stability under ambient conditions and their application to various types of transparent soft cell culture interfaces with anisotropic nanoscale topography. METHOD Nanofibers based on the polyvinyl alcohol and polyacrylic acid mixture were prepared by electrospinning and hydrogelified to nano-fibrous hydrogels (nFHs) after thermal crosslinking and sulfuric acid treatment. By modifying various material surfaces with positively-charged polymers, negatively-charged superabsorbent nFHs could be selectively patterned by employing micro-contact printing or horizontally aligned by applying shear force with a wired bar coater. RESULTS The angular distribution of bar-coated nFHs was dramatically reduced to ± 20° along the applied shear direction unlike the drop-coated nFHs which exhibit random orientations. Next, various types of cells were cultured on top of transparent soft nFHs which showed good viability and attachment while their behaviors could be easily monitored by both upright and inverted optical microscopy. Particularly, neuronal lineage cells such as PC 12 cells and embryonic hippocampal neurons showed highly stretched morphology along the overall fiber orientation with aspect ratios ranging from 1 to 14. Furthermore, the resultant neurite outgrowth and migration behaviors could be effectively controlled by the horizontal orientation and the three-dimensional arrangement of underlying nFHs, respectively. CONCLUSION We expect that surface modifications with transparent soft nFHs will be beneficial for various biological/biomedical studies such as fundamental cellular studies, neuronal/stem cell and/or organoid cultures, implantable probe/device coatings, etc.
Collapse
Affiliation(s)
- Jaeil Park
- School of Materials Science and Engineering, Gwangju Institute of Science and Technology (GIST), 123 Cheomdangwagi-Ro, Buk-Gu, Gwangju, 61005, Republic of Korea
| | - Thi Thuy Chau Nguyen
- Department of Polymer Science and Engineering, Graduate School of Chungnam National University, 99 Daehak-Ro, Yuseong-Gu, Daejeon, 34134, Republic of Korea
| | - Su-Jin Lee
- Department of Polymer Science and Engineering, Graduate School of Chungnam National University, 99 Daehak-Ro, Yuseong-Gu, Daejeon, 34134, Republic of Korea
| | - Sungrok Wang
- School of Materials Science and Engineering, Gwangju Institute of Science and Technology (GIST), 123 Cheomdangwagi-Ro, Buk-Gu, Gwangju, 61005, Republic of Korea
| | - Dongmi Heo
- School of Materials Science and Engineering, Gwangju Institute of Science and Technology (GIST), 123 Cheomdangwagi-Ro, Buk-Gu, Gwangju, 61005, Republic of Korea
| | - Dong-Hee Kang
- School of Materials Science and Engineering, Gwangju Institute of Science and Technology (GIST), 123 Cheomdangwagi-Ro, Buk-Gu, Gwangju, 61005, Republic of Korea
| | - Alexander Tipan-Quishpe
- School of Materials Science and Engineering, Gwangju Institute of Science and Technology (GIST), 123 Cheomdangwagi-Ro, Buk-Gu, Gwangju, 61005, Republic of Korea
| | - Won-June Lee
- School of Materials Science and Engineering, Gwangju Institute of Science and Technology (GIST), 123 Cheomdangwagi-Ro, Buk-Gu, Gwangju, 61005, Republic of Korea
| | - Jongwon Lee
- School of Materials Science and Engineering, Gwangju Institute of Science and Technology (GIST), 123 Cheomdangwagi-Ro, Buk-Gu, Gwangju, 61005, Republic of Korea
| | - Sung Yun Yang
- Department of Polymer Science and Engineering, Graduate School of Chungnam National University, 99 Daehak-Ro, Yuseong-Gu, Daejeon, 34134, Republic of Korea.
| | - Myung-Han Yoon
- School of Materials Science and Engineering, Gwangju Institute of Science and Technology (GIST), 123 Cheomdangwagi-Ro, Buk-Gu, Gwangju, 61005, Republic of Korea.
| |
Collapse
|