Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

Article Types

Countries / Regions

Search Results (205)

Search Parameters:
Keywords = engineered macrophages

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
17 pages, 1027 KiB  
Review
Chimeric Antigen Receptor Immunotherapy for Infectious Diseases: Current Advances and Future Perspectives
by Maria Kourti, Paschalis Evangelidis, Emmanuel Roilides and Elias Iosifidis
Pathogens 2025, 14(8), 774; https://doi.org/10.3390/pathogens14080774 (registering DOI) - 5 Aug 2025
Abstract
Chimeric antigen receptor (CAR)-T immunotherapy has revolutionized the management of patients with relapsed/refractory B-cell hematological malignancies. There is emerging evidence that CAR-engineered cells—not only T cells, but also natural killers and macrophages—might have a crucial role in the treatment of autoimmune disorders and [...] Read more.
Chimeric antigen receptor (CAR)-T immunotherapy has revolutionized the management of patients with relapsed/refractory B-cell hematological malignancies. There is emerging evidence that CAR-engineered cells—not only T cells, but also natural killers and macrophages—might have a crucial role in the treatment of autoimmune disorders and solid tumors. Moreover, given the burden of chronic infectious diseases, the mortality and morbidity of infections in immunocompromised individuals, and the development of multidrug-resistant pathogens, including bacteria, fungi, and mycobacteria, a need for novel and personalized therapeutics in this field is emerging. To this end, the development of CAR cells for the management of chronic infections has been reported. In this literature review, we summarize the ongoing clinical and pre-clinical data about CAR cell products in the field of infectious diseases. Currently, clinical studies on CAR immunotherapy for infections mainly concern human immunodeficiency virus infection treatment, and data regarding other infections largely originate from preclinical in vitro and in vivo models. In the era of personalized medicine, effective and safe therapies for the management of chronic infections and infectious complications in immunocompromised patients are crucial. Full article
(This article belongs to the Special Issue Bacterial Resistance and Novel Therapeutic Approaches)
Show Figures

Figure 1

23 pages, 954 KiB  
Review
The Role of Cobalt Ions in Angiogenesis—A Review
by Wiktor Gregorowicz and Lukasz Pajchel
Int. J. Mol. Sci. 2025, 26(15), 7236; https://doi.org/10.3390/ijms26157236 - 26 Jul 2025
Viewed by 357
Abstract
Cobalt is an essential trace element involved in key biological processes. It serves most notably as a component of vitamin B12 (cobalamin) and a regulator of erythropoiesis. While cobalt deficiency can lead to disorders such as megaloblastic anemia, excess cobalt poses toxicological [...] Read more.
Cobalt is an essential trace element involved in key biological processes. It serves most notably as a component of vitamin B12 (cobalamin) and a regulator of erythropoiesis. While cobalt deficiency can lead to disorders such as megaloblastic anemia, excess cobalt poses toxicological risks to the thyroid, cardiovascular, and hematopoietic systems. In recent years, cobalt ions (Co2+) have gained attention for their ability to mimic hypoxia and promote angiogenesis. This represents a crucial mechanism for tissue regeneration. Cobalt mediates this effect mainly by stabilizing hypoxia-inducible factor 1α (HIF-1α) under normoxic conditions, thereby upregulating angiogenic genes, including VEGF, FGF, and EPO. Experimental studies—from cell culture to animal models—have demonstrated cobalt-induced enhancement of endothelial proliferation, migration, and microvascular formation. Emerging evidence also indicates that Co2+-stimulated macrophages secrete integrin-β1-rich exosomes. These exosomes enhance endothelial motility and tubulogenesis independently of VEGF. Furthermore, cobalt-modified biomaterials have been developed to deliver cobalt ions in a controlled manner. Examples include cobalt-doped β-tricalcium phosphate or bioactive glasses. These materials support both angiogenesis and osteogenesis.This review summarizes current findings on cobalt’s role in angiogenesis. The emphasis is on its potential in cobalt-based biomaterials for tissue engineering and regenerative medicine. Full article
Show Figures

Graphical abstract

19 pages, 2472 KiB  
Article
Immunomodulation Through Fibroblast-Derived Extracellular Vesicles (EVs) Within 3D Polycaprolactone–Collagen Matrix
by Afsara Tasnim, Diego Jacho, Agustin Rabino, Jose Benalcazar, Rafael Garcia-Mata, Yakov Lapitsky and Eda Yildirim-Ayan
Biomimetics 2025, 10(8), 484; https://doi.org/10.3390/biomimetics10080484 - 22 Jul 2025
Viewed by 429
Abstract
Extracellular vesicles (EVs) have emerged as promising acellular tools for modulating immune responses for tissue engineering applications. This study explores the potential of human fibroblast-derived EVs delivered within a three-dimensional (3D) injectable scaffold composed of polycaprolactone (PCL) nanofibers and collagen (PNCOL) to reprogram [...] Read more.
Extracellular vesicles (EVs) have emerged as promising acellular tools for modulating immune responses for tissue engineering applications. This study explores the potential of human fibroblast-derived EVs delivered within a three-dimensional (3D) injectable scaffold composed of polycaprolactone (PCL) nanofibers and collagen (PNCOL) to reprogram macrophage behavior and support scaffold integrity under inflammatory conditions. EVs were successfully isolated from human fibroblasts using ultracentrifugation and characterized for purity, size distribution and surface markers (CD63 and CD9). Macrophage-laden PNCOL scaffolds were prepared under three conditions: macrophage-only (MP), fibroblast co-encapsulated (F-MP), and EV-encapsulated (EV-MP) groups. Structural integrity was assessed via scanning electron microscopy and Masson’s trichrome staining, while immunomodulatory effects were evaluated through metabolic assays, gene expression profiling, and immunohistochemistry for macrophage polarization markers (CD80, CD206). When co-encapsulated with pro-inflammatory (M1) macrophages in PNCOL scaffolds, fibroblast-derived EVs preserved scaffold structure and significantly enhanced macrophage metabolic activity compared to the control (MP) and other experimental group (F-MP). The gene expression and immunohistochemistry data demonstrated substantial upregulation of anti-inflammatory markers (TGF-β, CD163, and CCL18) and surface protein CD206, indicating a phenotypic shift toward M2-like macrophages for EV-encapsulated scaffolds relative to the other groups. The findings of this study demonstrate that fibroblast-derived EVs integrated into injectable PCL–collagen scaffolds offer a viable, cell-free approach to modulate inflammation, preserve scaffold structure, and support regenerative healing. This strategy holds significant promise for advancing immuno-instructive platforms in regenerative medicine, particularly in settings where conventional cell therapies face limitations in survival, cost, or safety. Full article
(This article belongs to the Special Issue Biomimetic Application on Applied Bioengineering)
Show Figures

Figure 1

16 pages, 691 KiB  
Review
Engineering Innate Immunity: Recent Advances and Future Directions for CAR-NK and CAR–Macrophage Therapies in Solid Tumors
by Behzad Amoozgar, Ayrton Bangolo, Charlene Mansour, Daniel Elias, Abdifitah Mohamed, Danielle C. Thor, Syed Usman Ehsanullah, Hadrian Hoang-Vu Tran, Izage Kianifar Aguilar and Simcha Weissman
Cancers 2025, 17(14), 2397; https://doi.org/10.3390/cancers17142397 - 19 Jul 2025
Viewed by 581
Abstract
Adoptive cell therapies have transformed the treatment landscape for hematologic malignancies. Yet, translation to solid tumors remains constrained by antigen heterogeneity, an immunosuppressive tumor microenvironment (TME), and poor persistence of conventional CAR-T cells. In response, innate immune cell platforms, particularly chimeric antigen receptor–engineered [...] Read more.
Adoptive cell therapies have transformed the treatment landscape for hematologic malignancies. Yet, translation to solid tumors remains constrained by antigen heterogeneity, an immunosuppressive tumor microenvironment (TME), and poor persistence of conventional CAR-T cells. In response, innate immune cell platforms, particularly chimeric antigen receptor–engineered natural killer (CAR-NK) cells and chimeric antigen receptor–macrophages (CAR-MΦ), have emerged as promising alternatives. This review summarizes recent advances in the design and application of CAR-NK and CAR-MΦ therapies for solid tumors. We highlight key innovations, including the use of lineage-specific intracellular signaling domains (e.g., DAP12, 2B4, FcRγ), novel effector constructs (e.g., NKG7-overexpressing CARs, TME-responsive CARs), and scalable induced pluripotent stem cell (iPSC)-derived platforms. Preclinical data support enhanced antitumor activity through mechanisms such as major histocompatibility complex (MHC)-unrestricted cytotoxicity, phagocytosis, trogocytosis, cytokine secretion, and cross-talk with adaptive immunity. Early-phase clinical studies (e.g., CT-0508) demonstrate feasibility and TME remodeling with CAR-MΦ. However, persistent challenges remain, including transient in vivo survival, manufacturing complexity, and risks of off-target inflammation. Emerging combinatorial strategies, such as dual-effector regimens (CAR-NK+ CAR-MΦ), cytokine-modulated cross-support, and bispecific or logic-gated CARs, may overcome these barriers and provide more durable, tumor-selective responses. Taken together, CAR-NK and CAR-MΦ platforms are poised to expand the reach of engineered cell therapy into the solid tumor domain. Full article
(This article belongs to the Special Issue Cell Therapy in Solid Cancers: Current and Future Landscape)
Show Figures

Figure 1

23 pages, 1713 KiB  
Review
Targeted and Biomimetic Nanoparticles for Atherosclerosis Therapy: A Review of Emerging Strategies
by Dorota Bartusik-Aebisher, Rafał Podgórski, Iga Serafin and David Aebisher
Biomedicines 2025, 13(7), 1720; https://doi.org/10.3390/biomedicines13071720 - 14 Jul 2025
Viewed by 600
Abstract
Atherosclerosis, a chronic inflammatory disease, remains a leading cause of cardiovascular mortality worldwide. Despite standard treatments like statins and percutaneous coronary intervention (PCI), significant residual risk and therapeutic limitations underscore the need for innovative strategies. This review summarizes recent advances in nanoparticle-based therapies [...] Read more.
Atherosclerosis, a chronic inflammatory disease, remains a leading cause of cardiovascular mortality worldwide. Despite standard treatments like statins and percutaneous coronary intervention (PCI), significant residual risk and therapeutic limitations underscore the need for innovative strategies. This review summarizes recent advances in nanoparticle-based therapies for atherosclerosis, focusing on key developments from the last five years. We discuss various nanoplatforms designed to selectively target key cellular players in plaque pathogenesis, including macrophages, endothelial cells, and vascular smooth muscle cells (VSMCs), to inhibit inflammation, modulate cellular phenotypes, and stabilize plaques. A significant focus is placed on the emerging field of biomimetic nanoparticles, where therapeutic cores are camouflaged with cell membranes derived from macrophages, platelets, neutrophils, or erythrocytes. This approach leverages the natural biological functions of the source cells to achieve enhanced immune evasion, prolonged circulation, and precise targeting of atherosclerotic lesions. Furthermore, the review covers nanoparticles engineered for specific functional interventions, such as lowering LDL levels and exerting direct anti-inflammatory and anti-oxidative effects. Finally, we address the critical challenges hindering clinical translation, including nanotoxicity, biodistribution, and manufacturing scalability. In conclusion, nanotechnology offers a versatile and powerful platform for atherosclerosis therapy, with targeted and biomimetic strategies holding immense promise to revolutionize future cardiovascular medicine. Full article
Show Figures

Figure 1

20 pages, 11811 KiB  
Article
Macrophage Migration Inhibitory Factor Suppresses Natural Killer Cell Response and Promotes Hypoimmunogenic Stem Cell Engraftment Following Spinal Cord Injury
by Shenglan Li, Yiyan Zheng, Haipeng Xue, Haiwei Zhang, Jiayun Wu, Xiaohui Chen, Miguel Perez Bouza, Samantha Yi, Hongxia Zhou, Xugang Xia, Xianmin Zeng, Qi Lin Cao and Ying Liu
Biology 2025, 14(7), 791; https://doi.org/10.3390/biology14070791 - 30 Jun 2025
Viewed by 443
Abstract
Human induced pluripotent stem cells (iPSCs) offer immense potential as a source for cell therapy in spinal cord injury (SCI) and other diseases. The development of hypoimmunogenic, universal cells that could be transplanted to any recipient without requiring a matching donor could significantly [...] Read more.
Human induced pluripotent stem cells (iPSCs) offer immense potential as a source for cell therapy in spinal cord injury (SCI) and other diseases. The development of hypoimmunogenic, universal cells that could be transplanted to any recipient without requiring a matching donor could significantly enhance their therapeutic potential and accelerate clinical translation. To create off-the-shelf hypoimmunogenic cells, we used CRISPR-Cas9 to delete B2M (HLA class I) and CIITA (master regulator of HLA class II). Double-knockout (DKO) iPSC-derived neural progenitor cells (NPCs) evaded T-cell-mediated immune rejection in vitro and after grafting into the injured spinal cord of athymic rats and humanized mice. However, loss of HLA class I heightened susceptibility to host natural killer (NK) cell attack, limiting graft survival. To counter this negative effect, we engineered DKO NPCs to overexpress macrophage migration inhibitory factor (MIF), an NK cell checkpoint ligand. MIF expression markedly reduced NK cell-mediated cytotoxicity and improved long-term engraftment and integration of NPCs in the animal models for spinal cord injury. These findings demonstrate that MIF overexpression, combined with concurrent B2M and CIITA deletion, generates hiPSC neural derivatives that escape both T- and NK-cell surveillance. This strategy provides a scalable route to universal donor cells for regenerative therapies in SCI and potentially other disorders. Full article
(This article belongs to the Special Issue Stem Cells in Neurological Disorders: Challenges and Opportunities)
Show Figures

Figure 1

18 pages, 535 KiB  
Review
Overcoming Immune Barriers in Allogeneic CAR-NK Therapy: From Multiplex Gene Editing to AI-Driven Precision Design
by Hyunyoung Kim
Biomolecules 2025, 15(7), 935; https://doi.org/10.3390/biom15070935 - 26 Jun 2025
Viewed by 875
Abstract
Chimeric antigen receptor (CAR)-engineered natural killer (NK) cells are a promising platform for off-the-shelf immunotherapy due to their safety advantages over CAR-T cells, including lower risk of graft-versus-host disease, cytokine release syndrome, and neurotoxicity. However, their persistence and efficacy are limited by immunological [...] Read more.
Chimeric antigen receptor (CAR)-engineered natural killer (NK) cells are a promising platform for off-the-shelf immunotherapy due to their safety advantages over CAR-T cells, including lower risk of graft-versus-host disease, cytokine release syndrome, and neurotoxicity. However, their persistence and efficacy are limited by immunological challenges such as host T-cell-mediated rejection, NK cell fratricide, and macrophage-mediated clearance. This review summarizes gene editing strategies to overcome these barriers, including β2-microglobulin (B2M) knockout and HLA-E overexpression to evade T and NK cell attacks, CD47 overexpression to inhibit phagocytosis, and TIGIT deletion to enhance cytotoxicity. In addition, we discuss functional enhancements such as IL-15 pathway activation, KIR modulation, and transcriptional reprogramming (e.g., FOXO1 knockout) to improve persistence and antitumor activity. We also highlight the role of induced pluripotent stem cell (iPSC)-derived NK platforms, enabling standardized, scalable, and multiplex gene-edited products. Finally, we explore artificial intelligence (AI) applications in immunogenomic profiling and predictive editing to tailor NK cell therapies to patient-specific HLA/KIR/SIRPα contexts. By integrating immune evasion, functional reinforcement, and computational design, we propose a unified roadmap for next-generation CAR-NK development, supporting durable and broadly applicable cell-based therapies. Full article
(This article belongs to the Section Bio-Engineered Materials)
Show Figures

Figure 1

21 pages, 2202 KiB  
Review
CAR Beyond αβ T Cells: Unleashing NK Cells, Macrophages, and γδ T Lymphocytes Against Solid Tumors
by Yunjia Xian and Lu Wen
Vaccines 2025, 13(6), 654; https://doi.org/10.3390/vaccines13060654 - 19 Jun 2025
Viewed by 1031
Abstract
Chimeric antigen receptor (CAR)-engineered cell therapy represents a landmark advancement in cancer immunotherapy. While αβ CAR-T therapy has demonstrated remarkable success in hematological malignancies, its efficacy in solid tumors remains constrained mainly by factors such as antigen heterogeneity, immunosuppressive microenvironments, and on-target/off-tumor toxicity. [...] Read more.
Chimeric antigen receptor (CAR)-engineered cell therapy represents a landmark advancement in cancer immunotherapy. While αβ CAR-T therapy has demonstrated remarkable success in hematological malignancies, its efficacy in solid tumors remains constrained mainly by factors such as antigen heterogeneity, immunosuppressive microenvironments, and on-target/off-tumor toxicity. To overcome these limitations, emerging CAR platforms that utilize alternative immune effectors, including natural killer (NK) cells, macrophages, and γδ T lymphocytes, are rapidly gaining traction. This review systematically analyzes the mechanistic advantages of CAR-NK, CAR-M, and CAR-γδ T cell therapies, while critically evaluating persistent challenges in clinical translation, including limited cell persistence, manufacturing scalability, and dynamic immune evasion mechanisms. We further discuss innovative strategies to enhance therapeutic efficacy through some viable strategies. By bridging fundamental immunology with translational engineering, this work provides a roadmap for developing CAR therapies capable of addressing the complexities of solid tumor eradication. Full article
Show Figures

Graphical abstract

23 pages, 2512 KiB  
Article
Bioprinted Four-Cell-Type Lung Model for Viral Infection Studies Under Air–Liquid Interface Conditions
by Johanna Berg, Julian Heinze, Daniela Niemeyer, Josefin Hellgren, Himjyot Jaiswal, Anna Löwa, Andreas Hocke, Itedale Namro, Christian Drosten, Jens Kurreck and Beatrice Tolksdorf
Int. J. Mol. Sci. 2025, 26(12), 5543; https://doi.org/10.3390/ijms26125543 - 10 Jun 2025
Viewed by 883
Abstract
Viral lung infections are a never-ending threat to public health due to the emergence of new variants and their seasonal nature. While vaccines offer some protection, the need for effective antiviral drugs remains high. The existing research methods using 2D cell culture and [...] Read more.
Viral lung infections are a never-ending threat to public health due to the emergence of new variants and their seasonal nature. While vaccines offer some protection, the need for effective antiviral drugs remains high. The existing research methods using 2D cell culture and animal models have their limitations. Human cell-based tissue engineering approaches hold great promise for bridging this gap. Here, we describe a microextrusion bioprinting approach to generate three-dimensional (3D) lung models composed of four cell types: endothelial cells, primary fibroblasts, macrophage cells, and epithelial cells. A549 and Calu-3 cells were selected as epithelial cells to simulate the cells of the lower and upper respiratory tract, respectively. Cells were bioprinted in a hydrogel consisting of alginate, gelatin, hyaluronic acid, collagen, and laminin-521. The models were cultured under air–liquid interface (ALI) conditions to further enhance their physiological relevance as lung cells. Their viability, metabolic activity, and expression of specific cell markers were analyzed during long-term culture for 21 days. The constructs were successfully infected with both a seasonal influenza A virus (IAV) and the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) omicron variant, demonstrating their potential for studying diverse viral infections. Full article
(This article belongs to the Section Molecular Biology)
Show Figures

Graphical abstract

22 pages, 993 KiB  
Review
Cell-Based Therapies for Solid Tumors: Challenges and Advances
by Anna Smolarska, Zuzanna Kokoszka, Marcelina Naliwajko, Julia Strupczewska, Jędrzej Tondera, Maja Wiater and Roksana Orzechowska
Int. J. Mol. Sci. 2025, 26(12), 5524; https://doi.org/10.3390/ijms26125524 - 9 Jun 2025
Viewed by 1119
Abstract
Solid tumors pose significant therapeutic challenges due to their resistance to conventional treatments and the complexity of the tumor microenvironment. Cell-based immunotherapies offer a promising approach, enabling precise, personalized treatment through immune system modulation. This review explores several emerging cellular therapies for solid [...] Read more.
Solid tumors pose significant therapeutic challenges due to their resistance to conventional treatments and the complexity of the tumor microenvironment. Cell-based immunotherapies offer a promising approach, enabling precise, personalized treatment through immune system modulation. This review explores several emerging cellular therapies for solid tumors, including tumor-infiltrating lymphocytes, T cell receptor-engineered T cells, CAR T cells, CAR natural killer cells, and macrophages. Tumor-infiltrating lymphocytes and their modified versions, T cell receptor-engineered T cells and CAR T cells, provide personalized immune responses, although their effectiveness can be limited by factors like variation in tumor antigens and the suppressive nature of the tumor environment. Natural killer cells engineered with chimeric receptors offer safer, non-major histocompatibility complex-restricted targeting, while modified macrophages exploit their natural ability to enter tumors and reshape the immune landscape. CAR-modified macrophages and macrophages conjugated with drugs are also considered as therapy for solid tumors. The review also examines the implications of autologous versus allogeneic cell sources. Autologous therapies ensure immunologic compatibility but are limited by scalability and manufacturing constraints. Allogeneic approaches offer “off-the-shelf” potential but require gene editing to avoid immune rejection. Integrating synthetic biology, gene editing, and combinatorial strategies will be essential to enhance efficacy and expand the clinical utility of cellular immunotherapies for solid tumors. Full article
(This article belongs to the Special Issue Macrophages in Human Diseases and Their Treatment)
Show Figures

Figure 1

27 pages, 1354 KiB  
Review
Biomedical Applications of Functionalized Composites Based on Metal–Organic Frameworks in Bone Diseases
by Chenxi Yun, Zhe Yuan, Rim El Haddaoui-Drissi, Ruitong Ni, Yunyun Xiao, Zhenhui Qi, Jie Shang and Xiao Lin
Pharmaceutics 2025, 17(6), 757; https://doi.org/10.3390/pharmaceutics17060757 - 8 Jun 2025
Viewed by 1033
Abstract
Every year, millions of people worldwide suffer from bone tissue damage caused by bone trauma and surgical operations, as well as diseases such as osteoporosis, osteoarthritis, osteomyelitis, and periodontitis. Bone defect repair is one of the major challenges in the field of regenerative [...] Read more.
Every year, millions of people worldwide suffer from bone tissue damage caused by bone trauma and surgical operations, as well as diseases such as osteoporosis, osteoarthritis, osteomyelitis, and periodontitis. Bone defect repair is one of the major challenges in the field of regenerative medicine. Although bone grafts are the gold standard for treating bone defects, factors such as donor sources and immune responses limit their application. Functionalized nanomaterials have become an effective means of treating bone diseases due to their good biocompatibility and osteoinductivity, anti-inflammatory, and antibacterial properties. Metal–organic frameworks (MOFs) are porous coordination polymers composed of metal ions and organic ligands, featuring unique physical properties, including a high surface area–volume ratio and porosity. In regenerative medicine, MOFs function as the functions of drug carriers, metal ion donors, nanozymes, and photosensitizers. When combined with other functional materials, they regulate cellular reactive oxygen species, macrophage phenotypic transformation, bone resorption, osteogenesis, and mineralization, providing a new paradigm for bone tissue engineering. This study reviews the classification of functionalized MOF composites in biomedicine and the application of their synthesis techniques in bone diseases. The unique in vivo and in vitro applications of MOFs in bone diseases, including osteoarthritis, osteoporosis, bone tumors, osteomyelitis, and periodontitis, are explored. Their properties include excellent drug loading and sustained release abilities, high antibacterial activity, and bone induction abilities. This review enables readers to better understand the cutting-edge progress of MOFs in bone regeneration applications, which is crucial for the design of and functional research on MOF-related nanomaterials. Full article
Show Figures

Graphical abstract

21 pages, 3611 KiB  
Article
A Pathophysiologically Hypertrophic 3T3-L1 Cell Model—An Alternative to Primary Cells Isolated from DIO Mice
by Isabell Kaczmarek, Kristiana Schüßler, Andreas Lindhorst, Martin Gericke and Doreen Thor
Cells 2025, 14(11), 837; https://doi.org/10.3390/cells14110837 - 3 Jun 2025
Viewed by 816
Abstract
Adipocyte hypertrophy in individuals with obesity is connected to alterations in adipocyte function. These pathophysiological changes are studied using animal models and adipose tissue engineering. However, knockdown, overexpression, and stimulation studies would benefit from an easily applicable cell model. Although several models (free [...] Read more.
Adipocyte hypertrophy in individuals with obesity is connected to alterations in adipocyte function. These pathophysiological changes are studied using animal models and adipose tissue engineering. However, knockdown, overexpression, and stimulation studies would benefit from an easily applicable cell model. Although several models (free fatty acids, glucose restriction, and long-term incubation) have previously been described, our evaluation demonstrated that they lack important features described for hypertrophic adipocytes found in obesity. Therefore, we aimed to develop a cell model depicting the pathophysiological state of adipocytes in obesity by applying novel approaches (insulin, macrophage supernatant, and Tnfα) using 3T3-L1 cells. To analyze changes in adipocyte phenotype and function, we detected the cell size, lipid accumulation, insulin sensitivity, cytokine/adipokine secretion, and expression of lipolytic enzymes. Combining long-term incubation with insulin and Tnfα co-stimulation, we found significantly increased cell size and lipid accumulation compared to 3T3-L1 adipocytes differentiated with standard protocols. Furthermore, these adipocytes showed significantly reduced insulin sensitivity, adiponectin secretion, and lipolytic enzyme expression, accompanied by increased IL6 and leptin secretion. In summary, the described cell model depicts pathophysiologically hypertrophic 3T3-L1 adipocytes. This model can be used for knockdown, overexpression, and stimulation studies, thereby serving as an alternative to primary cells isolated from DIO mice. Full article
(This article belongs to the Section Cellular Pathology)
Show Figures

Figure 1

18 pages, 3609 KiB  
Article
Semi-Interpenetrating Polymer Networks Incorporating Polygalacturonic Acid: Physical Characterization and In Vitro Biocompatibility
by Aisling N. O’Carroll, Colin P. McCoy and Louise Carson
Processes 2025, 13(5), 1502; https://doi.org/10.3390/pr13051502 - 14 May 2025
Viewed by 479
Abstract
Polygalacturonic acid (PGA), derived from the natural plant polysaccharide, pectin, has been suggested as a biomaterial for implantable medical devices and tissue engineering; particularly in the field of bone implant materials. As a negatively charged polysaccharide, PGA can be considered similar to hyaluronic [...] Read more.
Polygalacturonic acid (PGA), derived from the natural plant polysaccharide, pectin, has been suggested as a biomaterial for implantable medical devices and tissue engineering; particularly in the field of bone implant materials. As a negatively charged polysaccharide, PGA can be considered similar to hyaluronic acid, a component of the extracellular matrix (ECM). PGA-based biomaterials may therefore exhibit favorable biocompatibility with surface chemistry mimicking the natural ECM. In this study, we synthesized semi-interpenetrating polymer networks (SIPNs) incorporating PGA, and conducted physical characterization and in vitro biocompatibility studies. Biocompatibility testing revealed the SIPNs to be cytocompatible, with the PGA component conferring some resistance to the adherence of the macrophage cell line RAW264.7. In addition, SIPNs did not support the fusion of primary murine macrophages into foreign body giant cells (FBGCs). Macrophage adherence and FBGC formation on implanted biomaterial surfaces are important events in the progression of a foreign body response. Our in vitro studies suggest that PGA-based materials may offer desirable biocompatibility profiles, holding promise for future clinical applications. Full article
Show Figures

Figure 1

21 pages, 4834 KiB  
Article
A Multifunctional PEEK Composite Scaffold with Immunomodulatory, Angiogenic, and Osteogenic Properties for Enhanced Bone Regeneration
by Mengen Zhao, Han Yang, Qianwen Yang, Chao Zhang, Jie Liu, Zhaoying Wu, Lijun Wang, Wei Zhang, Bing Wang and Wenliang Liu
Polymers 2025, 17(9), 1206; https://doi.org/10.3390/polym17091206 - 28 Apr 2025
Viewed by 546
Abstract
Polyetheretherketone (PEEK) is a widely used material in bone tissue engineering due to its favorable mechanical properties and radiolucency. However, its bioinert nature and lack of osteogenic activity restrict its ability to support effective bone regeneration. In this study, a novel APS-coated plasma-treated [...] Read more.
Polyetheretherketone (PEEK) is a widely used material in bone tissue engineering due to its favorable mechanical properties and radiolucency. However, its bioinert nature and lack of osteogenic activity restrict its ability to support effective bone regeneration. In this study, a novel APS-coated plasma-treated sulfonated bioactive PEEK scaffold (APS/PSBPK) was developed to overcome these limitations. The scaffold integrates strontium-doped bioactive glass (SrBG) to enhance biocompatibility and osteogenic potential, while astragalus polysaccharide (APS) was incorporated via plasma cleaning to modulate immune responses and promote vascularization. In vitro studies demonstrated that the APS/PSBPK scaffold facilitates M2 macrophage polarization, reduces pro-inflammatory cytokines, and enhances the secretion of anti-inflammatory factors. It also promotes endothelial cell migration and angiogenesis while supporting the adhesion, proliferation, and osteogenic differentiation of rBMSCs. In vivo experiments revealed that the scaffold effectively regulates the immune microenvironment, promotes vascularization, and accelerates bone regeneration. Thus, the APS/PSBPK composite scaffold serves as a multifunctional biomaterial with significant potential for applications in bone repair and regeneration by combining immunomodulation, angiogenesis, and osteogenesis. Full article
(This article belongs to the Section Polymer Applications)
Show Figures

Graphical abstract

25 pages, 6242 KiB  
Article
Development and Characterization of an Injectable Alginate/Chitosan Composite Hydrogel Reinforced with Cyclic-RGD Functionalized Graphene Oxide for Potential Tissue Regeneration Applications
by Mildred A. Sauce-Guevara, Sergio D. García-Schejtman, Emilio I. Alarcon, Sergio A. Bernal-Chavez and Miguel A. Mendez-Rojas
Pharmaceuticals 2025, 18(5), 616; https://doi.org/10.3390/ph18050616 - 23 Apr 2025
Cited by 1 | Viewed by 1609
Abstract
Background: In tissue engineering, developing injectable hydrogels with tailored mechanical and bioactive properties remains a challenge. This study introduces an injectable hydrogel composite for soft tissue regeneration, composed of oxidized alginate (OA) and N-succinyl chitosan (NSC) cross-linked via Schiff base reaction, reinforced with [...] Read more.
Background: In tissue engineering, developing injectable hydrogels with tailored mechanical and bioactive properties remains a challenge. This study introduces an injectable hydrogel composite for soft tissue regeneration, composed of oxidized alginate (OA) and N-succinyl chitosan (NSC) cross-linked via Schiff base reaction, reinforced with graphene oxide (GOx) and cyclic arginylglycylaspartic acid (c-RGD). The objective was to create a multifunctional platform combining injectability, bioactivity, and structural stability. Methods: The OA/NSC/GOx-cRGD hydrogel was synthesized through Schiff base cross-linking (aldehyde-amine reaction). Characterization included FTIR (C=N bond at 1650 cm⁻¹), Raman spectroscopy (D/G bands at 1338/1567 cm⁻¹), SEM (porous microstructure), and rheological analysis (shear-thinning behavior). In vitro assays assessed fibroblast viability (MTT) and macrophage TNF-α secretion (ELISA), while ex-vivo injectability and retention were evaluated using chicken cardiac tissue. Results: The hydrogel exhibited shear-thinning behavior (viscosity: 10 to <1 Pa·s) and elastic-dominated mechanics (G′ > G″), ensuring injectability. SEM revealed an interconnected porous structure mimicking native extracellular matrix. Fibroblast viability remained ≥95%, and TNF-α secretion in macrophages decreased by 80% (30 vs. 150 pg/μL in controls), demonstrating biocompatibility and anti-inflammatory effects. The hydrogel adhered stably to cardiac tissue without leakage. Conclusions: The OA/NSC/GOx-cRGD composite integrates injectability, bioactivity, and structural stability, offering a promising scaffold for tissue regeneration. Its modular design allows further functionalization with peptides or growth factors. Future work will focus on translational applications, including scalability and optimization for dynamic biological environments. Full article
(This article belongs to the Section Biopharmaceuticals)
Show Figures

Graphical abstract

Back to TopTop