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
remove_circle_outline
remove_circle_outline

Journals

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (624)

Search Parameters:
Keywords = bone-marrow stromal cell

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
19 pages, 1242 KiB  
Review
Modeling the Bone Marrow Microenvironment to Better Understand the Pathogenesis, Progression, and Treatment of Hematological Cancers
by Kathryn A. Skelding, Daniel L. Barry and Lisa F. Lincz
Cancers 2025, 17(15), 2571; https://doi.org/10.3390/cancers17152571 - 4 Aug 2025
Abstract
Despite significant advancements in understanding the pathogenesis and treatment of hematological malignancies, including leukemia and multiple myeloma, the majority of patients continue to experience poor long-term outcomes. This is partly due to the difficulty of accurately recapitulating the malignant microenvironment in vitro, particularly [...] Read more.
Despite significant advancements in understanding the pathogenesis and treatment of hematological malignancies, including leukemia and multiple myeloma, the majority of patients continue to experience poor long-term outcomes. This is partly due to the difficulty of accurately recapitulating the malignant microenvironment in vitro, particularly the bone marrow niche. The complexity of the bone marrow microenvironment poses a challenge for the in vitro examination of hematological malignancies. Traditionally, 2D culture and animal models have been utilized, but these representations are limited and have been criticized for their lack of human physiological relevance. In an attempt to overcome this, 3D models have been developed that more accurately recapitulate the in vivo microenvironment. Herein, we present an overview of recent developments in 2D and 3D models used for studying the bone marrow niche in hematological malignancies, highlighting their advantages and limitations. Full article
(This article belongs to the Section Tumor Microenvironment)
Show Figures

Figure 1

20 pages, 23283 KiB  
Article
Titanium–Aluminum–Vanadium Surfaces Generated Using Sequential Nanosecond and Femtosecond Laser Etching Provide Osteogenic Nanotopography on Additively Manufactured Implants
by Jonathan T. Dillon, David J. Cohen, Scott McLean, Haibo Fan, Barbara D. Boyan and Zvi Schwartz
Biomimetics 2025, 10(8), 507; https://doi.org/10.3390/biomimetics10080507 - 4 Aug 2025
Abstract
Titanium–aluminum–vanadium (Ti6Al4V) is a material chosen for spine, orthopedic, and dental implants due to its combination of desirable mechanical and biological properties. Lasers have been used to modify metal surfaces, enabling the generation of a surface on Ti6Al4V with distinct micro- and nano-scale [...] Read more.
Titanium–aluminum–vanadium (Ti6Al4V) is a material chosen for spine, orthopedic, and dental implants due to its combination of desirable mechanical and biological properties. Lasers have been used to modify metal surfaces, enabling the generation of a surface on Ti6Al4V with distinct micro- and nano-scale structures. Studies indicate that topography with micro/nano features of osteoclast resorption pits causes bone marrow stromal cells (MSCs) and osteoprogenitor cells to favor differentiation into an osteoblastic phenotype. This study examined whether the biological response of human MSCs to Ti6Al4V surfaces is sensitive to laser treatment-controlled micro/nano-topography. First, 15 mm diameter Ti6Al4V discs (Spine Wave Inc., Shelton, CT, USA) were either machined (M) or additively manufactured (AM). Surface treatments included no laser treatment (NT), nanosecond laser (Ns), femtosecond laser (Fs), or nanosecond followed by femtosecond laser (Ns+Fs). Surface wettability, roughness, and surface chemistry were determined using sessile drop contact angle, laser confocal microscopy, X-ray photoelectron spectroscopy (XPS), and scanning electron microscopy (SEM). Human MSCs were cultured in growth media on tissue culture polystyrene (TCPS) or test surfaces. On day 7, the levels of osteocalcin (OCN), osteopontin (OPN), osteoprotegerin (OPG), and vascular endothelial growth factor 165 (VEGF) in the conditioned media were measured. M NT, Fs, and Ns+Fs surfaces were hydrophilic; Ns was hydrophobic. AM NT and Fs surfaces were hydrophilic; AM Ns and Ns+Fs were hydrophobic. Roughness (Sa and Sz) increased after Ns and Ns+Fs treatment for both M and AM disks. All surfaces primarily consisted of oxygen, titanium, and carbon; Fs had increased levels of aluminum for both M and AM. SEM images showed that M NT discs had a smooth surface, whereas AM surfaces appeared rough at a higher magnification. Fs surfaces had a similar morphology to their respective NT disc at low magnification, but higher magnification revealed nano-scale bumps not seen on NT surfaces. AM Fs surfaces also had regular interval ridges that were not seen on non-femto laser-ablated surfaces. Surface roughness was increased on M and AM Ns and Ns+Fs disks compared to NT and Fs disks. OCN was enhanced, and DNA was reduced on Ns and Ns+Fs, with no difference between them. OPN, OPG, and VEGF levels for laser-treated M surfaces were unchanged compared to NT, apart from an increase in OPG on Fs. MSCs grown on AM Ns and Ns+Fs surfaces had increased levels of OCN per DNA. These results indicate that MSCs cultured on AM Ns and AM Ns+Fs surfaces, which exhibited unique roughness at the microscale and nanoscale, had enhanced differentiation to an osteoblastic phenotype. The laser treatments of the surface mediated this enhancement of MSC differentiation and warrant further clinical investigation. Full article
Show Figures

Graphical abstract

16 pages, 2767 KiB  
Review
Breaking Barriers: The Role of the Bone Marrow Microenvironment in Multiple Myeloma Progression
by Aleksandra Agafonova, Chiara Prinzi, Angela Trovato Salinaro, Caterina Ledda, Alessia Cosentino, Maria Teresa Cambria, Carmelina Daniela Anfuso and Gabriella Lupo
Int. J. Mol. Sci. 2025, 26(15), 7301; https://doi.org/10.3390/ijms26157301 - 28 Jul 2025
Viewed by 215
Abstract
Multiple myeloma (MM) is an incurable malignancy characterized by the proliferation of abnormal plasma cells within the bone marrow, followed by potential dissemination to extramedullary sites. The bone marrow barrier (BMB) plays a pivotal role in plasma cell homing and disease progression. Bone [...] Read more.
Multiple myeloma (MM) is an incurable malignancy characterized by the proliferation of abnormal plasma cells within the bone marrow, followed by potential dissemination to extramedullary sites. The bone marrow barrier (BMB) plays a pivotal role in plasma cell homing and disease progression. Bone marrow endothelial cells (BMECs) and bone marrow stromal cells (BMSCs), through their interactions with MM cells, secrete adhesion molecules, angiogenic cytokines, anti-apoptotic factors, and growth-promoting signals that support MM cell survival and proliferation. This review examines the components of the BMB and the major pathways involved in MM pathogenesis. Targeting the interactions between MM cells and the BMB may offer novel therapeutic opportunities. Full article
Show Figures

Figure 1

18 pages, 2342 KiB  
Article
Accelerated Hydrolytic Degradation of PLA/Magnesium Composite Films: Material Properties and Stem Cell Interaction
by Valentina Fabi, Maria Luisa Valicenti, Franco Dominici, Francesco Morena, Luigi Torre, Sabata Martino and Ilaria Armentano
Polymers 2025, 17(15), 2052; https://doi.org/10.3390/polym17152052 - 27 Jul 2025
Viewed by 351
Abstract
The accelerated hydrolytic degradation of poly(L-lactide) (PLA)/magnesium (Mg) composite films was investigated to elucidate the influence of surface modification of Mg particles on the degradation behavior and characteristics of PLA composites. Accelerated degradation studies were conducted at 60 °C in a pH 7.4 [...] Read more.
The accelerated hydrolytic degradation of poly(L-lactide) (PLA)/magnesium (Mg) composite films was investigated to elucidate the influence of surface modification of Mg particles on the degradation behavior and characteristics of PLA composites. Accelerated degradation studies were conducted at 60 °C in a pH 7.4 phosphate-buffered solution over 7 weeks, with degradation monitored using several techniques: mass loss, water absorption, thermal analysis, and Raman spectroscopy. The results indicated that all composite films experienced more than 90% mass loss at the end of experiment; however, PLA/5MgTT and PLA/5MgPEI exhibited the highest resistance to degradation, likely due to the protective effect of the surface modification induced by thermal treatment and polyethylenimine (PEI). Notably, these characteristics did not compromise the biocompatibility or osteogenic potential of the films, which remained comparable to the control samples when tested on human bone marrow multipotent mesenchymal/stromal cells. Full article
(This article belongs to the Section Polymer Membranes and Films)
Show Figures

Figure 1

9 pages, 635 KiB  
Article
Osteogenic Potential of Osteolforte: Gene and Protein-Level Evaluation in Human Bone Marrow Stromal Cells
by Da-Sol Kim, Soo-Kyung Bae, Yeon-Ju Kwak, Geum-Joung Youn and Hye-Ock Jang
Curr. Issues Mol. Biol. 2025, 47(8), 588; https://doi.org/10.3390/cimb47080588 - 24 Jul 2025
Viewed by 284
Abstract
Osteolforte, a compound with potential bone-regenerative properties, was investigated for its effects on human bone marrow stromal cells (hBMSCs). This study aimed to evaluate its impact on cell viability, osteogenic differentiation, and both gene and protein expression using a combination of assays, [...] Read more.
Osteolforte, a compound with potential bone-regenerative properties, was investigated for its effects on human bone marrow stromal cells (hBMSCs). This study aimed to evaluate its impact on cell viability, osteogenic differentiation, and both gene and protein expression using a combination of assays, including CCK-8, Alizarin Red S staining, Quantitative Real-Time PCR (qRT-PCR), and Western blot analysis. The results demonstrated that Osteolforte significantly enhanced osteogenic differentiation in hBMSCs. Alizarin Red S staining revealed increased mineralization, indicating elevated calcium deposition. Gene expression analysis showed an upregulation of key osteogenic markers, including runt-related transcription factor-2 (RUNX-2), collagen type I (COL-1), and bone morphogenetic protein-2 (BMP-2), supporting the role of Osteolforte in promoting osteoblastic activity. In particular, the elevated expression of RUNX-2—a master transcription factor in osteoblast differentiation along with COL-1, a major bone matrix component, and BMP-2, a key bone morphogenetic protein—highlights the compound’s osteogenic potential. In conclusion, Osteolforte enhances early-stage osteogenesis and mineralization in hBMSCs and represents a promising candidate for bone regeneration. Full article
Show Figures

Figure 1

18 pages, 2571 KiB  
Article
Selective MicroRNA Packaging Reveals Distinct Core Signatures in Human Mesenchymal-Stromal-Cell-Derived Extracellular Vesicles
by Rachel E. Crossland, Clara Sanjurjo-Rodríguez, Monica Reis, Anne M. Dickinson, Elena Jones and Xiao-Nong Wang
Int. J. Mol. Sci. 2025, 26(14), 7010; https://doi.org/10.3390/ijms26147010 - 21 Jul 2025
Viewed by 345
Abstract
Mesenchymal stromal cells (MSCs) have demonstrated therapeutic efficacy across numerous clinical applications, with evidence suggesting their paracrine effects, particularly through extracellular vesicles (EVs), possibly driving functional outcomes. In this study we perform the comprehensive characterization of microRNA expression profiles in human MSC-derived EVs [...] Read more.
Mesenchymal stromal cells (MSCs) have demonstrated therapeutic efficacy across numerous clinical applications, with evidence suggesting their paracrine effects, particularly through extracellular vesicles (EVs), possibly driving functional outcomes. In this study we perform the comprehensive characterization of microRNA expression profiles in human MSC-derived EVs (MSC-EV) compared to their parental cells, cultured under clinically relevant xeno-free conditions. MSCs were isolated from the bone marrows of healthy donors and characterised according to the International Society for Cellular Therapy criteria, while MSC-EVs were isolated using differential ultracentrifugation and validated according to the International Society for Extracellular Vesicle guidelines. NanoString profiling identified 590 mature microRNAs expressed across both populations, with 42 being significantly differentially expressed between MSC-EVs and parental MSCs. Five microRNAs were distinctly highly expressed in MSCs and five in MSC-EVs, while fifteen of the top twenty most abundant microRNAs showed high expression in both populations. MicroRNA expression patterns were validated in an independent cohort. Functional pathway analysis of differentially expressed microRNAs showed enrichment of key biological processes including cell proliferation, differentiation, and immune regulation. This standardised profiling approach develops our understanding of MSC/MSC-EV microRNA cargo, using a transparent methodological approach that allows for the improved comparability of datasets for the development and advancement of MSC-EV therapeutics. Full article
(This article belongs to the Special Issue MicroRNA Regulation in Human Health and Diseases)
Show Figures

Graphical abstract

17 pages, 3961 KiB  
Article
Therapeutic Potential of Local Application of Fibroblast Growth Factor-2 to Periodontal Defects in a Preclinical Osteoporosis Model
by Shinta Mori, Sho Mano, Naoki Miyata, Tasuku Murakami, Wataru Yoshida, Kentaro Imamura and Atsushi Saito
Bioengineering 2025, 12(7), 748; https://doi.org/10.3390/bioengineering12070748 - 9 Jul 2025
Viewed by 432
Abstract
This study investigated the effects of local fibroblast growth factor (FGF)-2 application on periodontal healing in an osteoporotic model, both in vivo and in vitro. Wistar rats were divided into the ovariectomy (OVX) and Control groups. Periodontal defects were created 8 weeks post-OVX [...] Read more.
This study investigated the effects of local fibroblast growth factor (FGF)-2 application on periodontal healing in an osteoporotic model, both in vivo and in vitro. Wistar rats were divided into the ovariectomy (OVX) and Control groups. Periodontal defects were created 8 weeks post-OVX and treated with hydroxypropylcellulose (HPC) or FGF-2 + HPC. Healing was evaluated through micro-computed tomography and histological analyses at 2 and 4 weeks. In vitro, bone marrow mesenchymal stromal cells (BMSCs) were cultured with/without FGF-2 and assessed for cell morphology, viability/proliferation, and osteoblastic marker expression. Alkaline phosphatase (ALP) staining was also performed. FGF-2-treated defects in both groups showed significantly greater bone volume fraction, trabecular number, and thickness compared to HPC only. Histologically, FGF-2 enhanced new bone formation, with the greatest levels in the Control group. In vitro, OVX BMSCs showed reduced actin staining versus controls. FGF-2 increased cell viability/proliferation and protrusions in both groups while downregulating Alpl and Bglap expression levels and reducing ALP-positive cells. FGF-2 increased new bone formation in the OVX group, stimulated proliferation of OVX BMSCs, and modulated their differentiation. FGF-2 could enhance periodontal healing even under osteoporotic conditions, albeit to a lesser extent. Full article
(This article belongs to the Special Issue Recent Advances in Periodontal Tissue Engineering)
Show Figures

Graphical abstract

30 pages, 742 KiB  
Review
Biomimetic Three-Dimensional (3D) Scaffolds from Sustainable Biomaterials: Innovative Green Medicine Approach to Bone Regeneration
by Yashaswini Premjit, Merin Lawrence, Abhishek Goyal, Célia Ferreira, Elena A. Jones and Payal Ganguly
J. Funct. Biomater. 2025, 16(7), 238; https://doi.org/10.3390/jfb16070238 - 29 Jun 2025
Viewed by 992
Abstract
Bone repair and regeneration following an injury still present challenges worldwide. Three-dimensional (3D) scaffolds made from various materials are used for bone tissue engineering (BTE) applications. Polymers, minerals and nanotechnology are now being used in combination to achieve specific goals for BTE, including [...] Read more.
Bone repair and regeneration following an injury still present challenges worldwide. Three-dimensional (3D) scaffolds made from various materials are used for bone tissue engineering (BTE) applications. Polymers, minerals and nanotechnology are now being used in combination to achieve specific goals for BTE, including the delivery of antimicrobials through the scaffolds to prevent post-surgical infection. While several materials are utilised for BTE, natural polymers present a unique set of materials that can be manipulated to formulate scaffolds for BTE applications. They have been found to demonstrate higher biocompatibility, biodegradability and lower toxicity. Some even naturally mimic the bone microarchitecture, providing inherent structural support for BTE. Natural polymers may be simply classified as those from plant and animal sources. From both sources, there are different types of proteins, polysaccharides and other specialised materials that are already in use for research in BTE. Interestingly, these have the potential to revolutionise the field of BTE with a sustainable approach. In this review, we first discuss the different natural polymers used in BTE from plant sources, followed by animal sources. We then explore novel materials that are aimed at sustainable approaches, focusing on innovation from the last decade. In these sections, we outline studies of these materials with different types of bone cells, including bone marrow mesenchymal stromal cells (MSCs), which are the progenitors of bone. We finally outline the limitations, conclusions and future directions from our perspective in this dynamic field of polymers in BTE. With this review, we hope to bring together the updated existing knowledge and the potential future of innovation and sustainability in natural polymers for biomimetic BTE applications for fellow scientists, researchers and surgeons in the field. Full article
(This article belongs to the Special Issue Novel Biomaterials for Tissue Engineering)
Show Figures

Figure 1

22 pages, 4094 KiB  
Article
Expression of WNT Family Genes in Mesenchymal Stromal Cells of the Hematopoietic Niche in Patients with Different Responses to Multiple Myeloma Treatment
by Liubov A. Belik, Natella I. Enukashvily, Natalia Y. Semenova, Dmitrii I. Ostromyshenskii, Ekaterina V. Motyko, Anna N. Kirienko, Daria V. Kustova, Stanislav S. Bessmeltsev, Sergey V. Sidorkevich and Irina S. Martynkevich
Int. J. Mol. Sci. 2025, 26(13), 6236; https://doi.org/10.3390/ijms26136236 - 27 Jun 2025
Viewed by 376
Abstract
Mesenchymal stromal cells of the tumor microenvironment (TME) play a significant role in the progression of multiple myeloma (MM). The cells of the TME demonstrate resistance to treatment, thereby creating a favorable environment for disease relapse. The status of the TME during remission [...] Read more.
Mesenchymal stromal cells of the tumor microenvironment (TME) play a significant role in the progression of multiple myeloma (MM). The cells of the TME demonstrate resistance to treatment, thereby creating a favorable environment for disease relapse. The status of the TME during remission is poorly understood. An association between treatment response and TME status (including signaling pathways) has been suggested. One of the key players in the establishment of the MM TME is WNT signaling. In this study, we evaluated the expression of WNT family proteins in the TME and MM cells to assess their potential as TME markers and predictors of treatment response. A bioinformatic analysis of normal and malignant plasma cells, combined with an analysis of published data, revealed the following differentially expressed WNT genes: WNT5A, WNT10B, CTNNB1, and WNT3A. Immunohistochemical staining with the antibodies against the proteins encoded by the genes was conducted on trephine biopsy samples of bone marrow from healthy donors and patients with different responses to therapy. A quantitative analysis of the immunohistochemical data revealed differences in the amounts of WNT3A, WNT5A, WNT10B, and β-catenin proteins in the bone marrow before treatment depending on the subsequent responses of the patients to therapy. Multiplex fluorescent immunohistochemical staining with tyramide signal amplification revealed that WNT3A was predominantly present in mesenchymal stromal cells, whereas WNT5A and WNT10B were primarily observed in plasma cells. β-catenin was detected in both cell types. We analyzed the mRNA levels of the WNT gene family and CTNNB1 in MSC cultures from healthy donors and patients using qPCR. These genes were differentially expressed in MSC cultures derived from patients and healthy donors, as well as between patients grouped according to their response to therapy. Therefore, WNT proteins and β-catenin can be considered potential markers to assess the state of the tumor niche. Full article
(This article belongs to the Section Biochemistry)
Show Figures

Figure 1

25 pages, 1329 KiB  
Review
Modeling the Bone Marrow Niche in Multiple Myeloma: From 2D Cultures to 3D Systems
by Adele Bottaro, Maria Elisa Nasso, Fabio Stagno, Manlio Fazio and Alessandro Allegra
Int. J. Mol. Sci. 2025, 26(13), 6229; https://doi.org/10.3390/ijms26136229 - 27 Jun 2025
Viewed by 508
Abstract
Multiple myeloma is a hematologic malignancy characterized by the clonal proliferation of plasma cells within the bone marrow. The tumor microenvironment plays a crucial role in multiple myeloma pathogenesis, progression, and drug resistance. Traditional two-dimensional cell culture models have been instrumental in multiple [...] Read more.
Multiple myeloma is a hematologic malignancy characterized by the clonal proliferation of plasma cells within the bone marrow. The tumor microenvironment plays a crucial role in multiple myeloma pathogenesis, progression, and drug resistance. Traditional two-dimensional cell culture models have been instrumental in multiple myeloma research. However, they fail to recapitulate the complex in vivo bone marrow microenvironment, leading to limited predictive value for clinical outcomes. Three-dimensional cell culture models emerged as more physiologically relevant systems, offering enhanced insights into multiple myeloma biology. Scaffold-based systems (e.g., hydrogels, collagen, and Matrigel), scaffold-free spheroids, and bioprinted models have been developed to simulate the bone marrow microenvironment, incorporating key components like mesenchymal stromal cells, osteoblasts, endothelial cells, and immune cells. These models enable the functional assessment of cell adhesion-mediated drug resistance, cytokine signaling networks, and hypoxia-induced adaptations, which are often lost in 2D cultures. Moreover, 3D platforms demonstrated improved predictive value in preclinical drug screening, facilitating the evaluation of novel agents and combination therapies in a setting that better mimics the in vivo tumor context. Hence, 3D cultures represent a pivotal step toward bridging the gap between basic myeloma research and translational applications, supporting the development of more effective and patient-specific therapies. Full article
Show Figures

Figure 1

34 pages, 2583 KiB  
Review
Galectin-3 Release in the Bone Marrow Microenvironment Promotes Drug Resistance and Relapse in Acute Myeloid Leukemia
by Cansu Yıldırım
Life 2025, 15(6), 937; https://doi.org/10.3390/life15060937 - 10 Jun 2025
Viewed by 761
Abstract
Reciprocal signaling between acute myeloid leukemia (AML) cells and the surrounding bone-marrow microenvironment (BMME) promotes AML progression through several mechanisms. One of the most important mechanisms is the induction of Galectin-3 (Gal-3) expression by AML cells and bone marrow mesenchymal stromal cells (BM-MSCs). [...] Read more.
Reciprocal signaling between acute myeloid leukemia (AML) cells and the surrounding bone-marrow microenvironment (BMME) promotes AML progression through several mechanisms. One of the most important mechanisms is the induction of Galectin-3 (Gal-3) expression by AML cells and bone marrow mesenchymal stromal cells (BM-MSCs). Emerging evidence indicates that Gal-3 upregulation in the BMME promotes AML cell adhesion and survival, leading to the development of chemotherapy resistance, relapse, and poor prognosis. Identifying the biological function and critical signaling pathways of Gal-3 may contribute to overcoming acquired drug resistance and preventing post-treatment relapse. Gal-3 is involved in several molecular signaling pathways, including PI3K/AKT/mTOR, Ras/Raf/MEK/ERK, JAK/STAT, JNK, Wnt/β-catenin, PLC/PKC and NF-κB, which are interconnected to promote AML cell survival and resistance to chemotherapy. This review focuses on the biological effects, molecular mechanisms of action and regulation of Gal-3 in the pathogenesis and progression of AML. The therapeutic potential of potent synthetic small-molecule Gal-3 inhibitors in high-risk patients with AML is also discussed based on preclinical and clinical evidence from several human diseases. Currently, the effect of these Gal-3 inhibitors in AML has not been investigated either in vitro or in vivo. The findings provide a rationale for targeting Gal-3 that may be a very promising therapeutic approach, especially for patients with relapsed/refractory AML, and may enhance the efficacy of conventional chemotherapeutic drugs and/or immune checkpoint inhibitors. Full article
(This article belongs to the Special Issue Bone Cancer: From Molecular Mechanism to Treatment)
Show Figures

Figure 1

19 pages, 3292 KiB  
Article
Phenothiazine-Based Nanoaggregates: Dual Role in Bioimaging and Stem Cell-Driven Photodynamic Therapy
by Eleonora Calzoni, Alessio Cesaretti, Nicolò Montegiove, Maria Luisa Valicenti, Francesco Morena, Rajneesh Misra, Benedetta Carlotti and Sabata Martino
Nanomaterials 2025, 15(12), 894; https://doi.org/10.3390/nano15120894 - 10 Jun 2025
Viewed by 402
Abstract
Nanotechnology is transforming contemporary medicine by providing cutting-edge tools for the treatment and diagnosis of complex disorders. Advanced techniques such as bioimaging and photodynamic therapy (PDT) combine early diagnosis and targeted therapy, offering a more precise approach than conventional treatments. However, a significant [...] Read more.
Nanotechnology is transforming contemporary medicine by providing cutting-edge tools for the treatment and diagnosis of complex disorders. Advanced techniques such as bioimaging and photodynamic therapy (PDT) combine early diagnosis and targeted therapy, offering a more precise approach than conventional treatments. However, a significant obstacle for PDT is the need to selectively deliver photosensitizers to disease sites while minimizing systemic side effects. In this context, mesenchymal stem cells have emerged as promising biological carriers due to their natural tropism towards tumors, low immunogenicity, and their ability to overcome biological barriers. In this study, two push–pull compounds, NPI-PTZ and BTZ-PTZ, phenothiazine derivatives featuring aggregation-induced emission (AIE) abilities, were analyzed. These molecules proved to be excellent fluorescent probes and photosensitizing agents. When administered to human bone marrow-derived multipotent stromal cells (hBM-MSCs) and human adipose multipotent stem cells (hASCs), the compounds were efficiently internalized, maintained a stable fluorescent emission for several days, and showed phototoxicity after irradiation, without inducing major cytotoxic effects under normal conditions. These results highlight the potential of NPI-PTZ and BTZ-PTZ combined with mesenchymal stem cells as theranostic tools, bridging bioimaging and PDT, and suggest new possibilities for advanced therapeutic approaches in clinical applications. Full article
(This article belongs to the Section Biology and Medicines)
Show Figures

Graphical abstract

27 pages, 2819 KiB  
Review
Bone Marrow Niche Aging: Are Adipocytes Detrimental Cells in the Bone Marrow?
by Urban Švajger, Patrik Milić and Primož J. Rožman
Cells 2025, 14(11), 814; https://doi.org/10.3390/cells14110814 - 30 May 2025
Viewed by 1092
Abstract
Aging disrupts the bone marrow (BM) niche, a complex microenvironment crucial for hematopoietic stem cell (HSC) maintenance. A key, yet debated, hallmark of this aging process is the accumulation of bone marrow adipocytes (BMAds). This review explores the evolving role of BMAds in [...] Read more.
Aging disrupts the bone marrow (BM) niche, a complex microenvironment crucial for hematopoietic stem cell (HSC) maintenance. A key, yet debated, hallmark of this aging process is the accumulation of bone marrow adipocytes (BMAds). This review explores the evolving role of BMAds in the aging BM, particularly their influence on HSC regulation via metabolic, endocrine, and inflammatory pathways. Aging BMAds exhibit altered secretory profiles, including reduced leptin and adiponectin and increased pro-inflammatory signals, which skew hematopoiesis toward myeloid over lymphoid lineage production. Additionally, shifts in fatty acid composition and lactate signaling from BMAds may impair stem cell function. These changes, alongside aging-associated alterations in vascular, neural, and stromal components of the niche, contribute to diminished immune resilience in older adults. We discuss emerging therapeutic strategies targeting BMAd-derived factors, such as DPP4 inhibition or the modulation of β-adrenergic signaling, aimed at creating a more youthful BM environment. By summarizing current insights into the aging BM niche and the central role of BMAds, this review highlights mechanisms that could be targeted to rejuvenate hematopoiesis and improve immune function in the elderly. Full article
(This article belongs to the Section Cell Microenvironment)
Show Figures

Figure 1

15 pages, 4537 KiB  
Article
Betaine Alleviates Bisphosphonate-Related Osteonecrosis of the Jaw by Rescuing BMSCs Function in an m6A-METTL3-Dependent Manner
by Yizhou Jin, Jiaxin Song, Zhanqiu Diao, Xiao Han and Zhipeng Fan
Int. J. Mol. Sci. 2025, 26(11), 5233; https://doi.org/10.3390/ijms26115233 - 29 May 2025
Viewed by 489
Abstract
Bisphosphonate-related osteonecrosis of the jaw (BRONJ) is one of the side effects of bisphosphonate (BP) administration. Despite some preventive measures having been suggested, a definitive and effective treatment strategy for BRONJ remains to be established. Recent evidence has indicated that BPs dramatically impair [...] Read more.
Bisphosphonate-related osteonecrosis of the jaw (BRONJ) is one of the side effects of bisphosphonate (BP) administration. Despite some preventive measures having been suggested, a definitive and effective treatment strategy for BRONJ remains to be established. Recent evidence has indicated that BPs dramatically impair the function of orofacial bone marrow stromal cells (BMSCs), which may contribute to the development of osteonecrosis. Thus, we hypothesized that recovery-impaired function of BMSCs at lesion sites could be beneficial in treating BRONJ. N6-methyladenosine (m6A) modification is the most common epigenetic modification and has been demonstrated to play a vital role in the modulation of BMSCs’ function. We detected the role of m6A modification in regulating the function of orofacial BMSCs under BP stimulation, and found that BPs led to a reduction in the global m6A methylation level, SAM level, and METTL3 expression in BMSCs during the osteogenic differentiation period. Meanwhile, betaine, a methyl group donor, effectively reversed the BP-decreased global m6A methylation level and SAM level in BMSCs, as well as rescuing the differentiation ability of impaired BMSCs. In the last part, we built a BRONJ rat model and supplemented rats with betaine via drinking water. The results showed that betaine successfully attenuated bone lesions and promoted wound healing in BP-injected rats, thereby providing new insight into future clinical treatment for BRONJ. Full article
(This article belongs to the Section Molecular Pharmacology)
Show Figures

Graphical abstract

21 pages, 908 KiB  
Review
The Critical Role of Adipocytes in Leukemia
by Romane Higos, Kevin Saitoski, Mathieu Hautefeuille, Geneviève Marcelin, Karine Clément, Nadine Varin-Blank, Christophe Breton, Simon Lecoutre and Mélanie Lambert
Biology 2025, 14(6), 624; https://doi.org/10.3390/biology14060624 - 28 May 2025
Viewed by 729
Abstract
The bone marrow microenvironment is a dynamic and complex niche that plays a central role in the development, progression, and therapeutic resistance of leukemia. Among the various stromal and immune cells that compose this microenvironment, adipocytes are increasingly recognized as active participants rather [...] Read more.
The bone marrow microenvironment is a dynamic and complex niche that plays a central role in the development, progression, and therapeutic resistance of leukemia. Among the various stromal and immune cells that compose this microenvironment, adipocytes are increasingly recognized as active participants rather than passive bystanders. These cells contribute to leukemia pathophysiology by supplying leukemic cells with vital metabolic fuels such as free fatty acids and glutamine, which support cellular bioenergetics and biosynthesis. Furthermore, adipocytes secrete adipokines—including leptin, adiponectin, and others—that influence leukemic cell proliferation, apoptosis, and chemoresistance. Leukemic cells, in turn, are not merely recipients of these signals, but actively remodel the marrow niche to their advantage. They can suppress adipogenesis, inhibit the differentiation of mesenchymal stem cells into adipocytes, or reprogram existing adipocytes to adopt a tumor-supportive phenotype. These transformed adipocytes may enhance leukemic cell survival, dampen immune responses, and create a metabolic sanctuary that enables resistance to standard chemotherapies. This reciprocal and dynamic interaction between leukemic cells and adipocytes contributes significantly to minimal residual disease and relapse, posing a major challenge for durable remission. Recent advances in tissue engineering—such as organ-on-chip and 3D co-culture systems—offer promising platforms to recapitulate and study these leukemia–adipocyte interactions with high fidelity. These models facilitate mechanistic insights and provide a foundation for developing novel therapeutic strategies aimed at disrupting the metabolic and paracrine crosstalk within the leukemic niche. Targeting the adipocyte–leukemia axis represents a compelling and underexplored avenue for improving leukemia treatment by sensitizing malignant cells to existing therapies and overcoming the protective influence of the bone marrow microenvironment. Full article
(This article belongs to the Section Physiology)
Show Figures

Figure 1

Back to TopTop