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

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (322)

Search Parameters:
Keywords = marrow adiposity

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
13 pages, 1130 KB  
Article
Magnetic Resonance Imaging-Based Assessment of Bone Marrow Fat and T2 Relaxation in Adolescents with Obesity and Liver Steatosis: A Feasibility Pilot Study
by Camille Letissier, Kenza El Ghomari, Sylvie Gervais, Léna Ahmarani and Ramy El Jalbout
J. Clin. Med. 2025, 14(21), 7594; https://doi.org/10.3390/jcm14217594 - 26 Oct 2025
Viewed by 391
Abstract
Background: Adolescents suffering from obesity are at higher risk of bone fragility due to hepatic steatosis, which may lead to an inflammatory microenvironment in the bone marrow. We therefore aimed to assess the reliability of measuring the bone marrow fat fraction (BMFF) and [...] Read more.
Background: Adolescents suffering from obesity are at higher risk of bone fragility due to hepatic steatosis, which may lead to an inflammatory microenvironment in the bone marrow. We therefore aimed to assess the reliability of measuring the bone marrow fat fraction (BMFF) and T2* of the lumbar vertebral marrow using the proton density fat fraction (PDFF) sequence for adolescents with obesity and liver steatosis. Method: This was an observational feasibility pilot study on adolescents living with obesity and liver steatosis. Anthropometric measurements were obtained. Participants underwent abdominal MRI, MR elastography and dual-energy X-ray absorptiometry (DXA). Regions of interest were drawn using the radiology interface from the central L1 to L4 vertebrae on fat and T2* maps from the PDFF sequence. ImageJ was used to measure abdominal compartment fat areas. Descriptive analyses, the intraclass correlation coefficient, and correlation results were obtained from anthropometric, adiposity, BMFF, and T2* measurements. Results: We recruited 23 adolescents with a body mass index > 85th percentile and mean age = 14.7 years (interval 12–17 years), and n = 18 (78%) were boys. BMFF and T2* measurements were successful in 100% of cases. The intra-operator reproducibility of the BMFF and T2* measurements was excellent: ICC = 0.99 (95% confidence interval (CI) [0.986; 0.999]) and ICC = 0.99 (95% CI [0.992; 0.999]), respectively. The inter-operator ICC was good for BMFF (ICC = 0.89; 95% CI [0.705; 0.963]) and moderate for T2* (ICC = 0.66; 95% CI [0.239; 0.873]). Only BMFF was inversely correlated with vertebral-bone mineral density (r = −0.67; p = 0.0009). However, T2* measurements showed a positive linear relationship with the total body fat tissue percentage measured by DXA (r = 0.48; p = 0.03) and the total abdominal fat area (r = 0.45; p = 0.04). Conclusions: PDFF could be a reliable imaging biomarker for bone health assessment in adolescents living with obesity. Full article
(This article belongs to the Special Issue Pediatric Obesity: Causes, Prevention and Treatment)
Show Figures

Figure 1

16 pages, 3494 KB  
Article
Fibronectin- and Bioactive Glass-Modified Alginate Scaffolds Support Limited Primary Cell Proliferation In Vitro yet Demonstrate Effective Host Integration In Vivo
by Benedetta Guagnini, Andrea Mazzoleni, Adrien Moya, Arnaud Scherberich, Barbara Medagli, Ivan Martin, Davide Porrelli, Manuele G. Muraro and Gianluca Turco
J. Funct. Biomater. 2025, 16(10), 386; https://doi.org/10.3390/jfb16100386 - 15 Oct 2025
Viewed by 740
Abstract
Alginate-hydroxyapatite (AL) scaffolds modified with fibronectin (FN) or bioactive glass (BGMS10) have recently been characterized for their physicochemical properties and proposed as promising candidates for bone regeneration. Here, we present their first systematic biological evaluation, focusing on adhesion, proliferation, osteogenic differentiation, and in [...] Read more.
Alginate-hydroxyapatite (AL) scaffolds modified with fibronectin (FN) or bioactive glass (BGMS10) have recently been characterized for their physicochemical properties and proposed as promising candidates for bone regeneration. Here, we present their first systematic biological evaluation, focusing on adhesion, proliferation, osteogenic differentiation, and in vivo host response. We compared FN-, BG-, and unmodified AL scaffolds using an immortalized mesenchymal stromal cell line (M-SOD) and primary human bone marrow-derived (BM-MSCs) and adipose-derived stromal cells (ASCs). FN scaffolds enhanced initial adhesion across all cell types and supported proliferation in M-SODs, but primary BM-MSCs and ASCs showed minimal expansion, regardless of scaffold type. BG scaffolds promoted expression of late-stage osteogenic markers in BM-MSCs, consistent with their ion release profile, but had limited impact on ASCs. In vivo subcutaneous implantation of acellular scaffolds in nude mice revealed robust host cell infiltration and extracellular matrix deposition across all scaffold types, confirming biocompatibility and integration. However, vascularization remained limited and did not differ substantially between formulations. Together, these findings highlight a critical discrepancy between immortalized and primary stromal cell responses to scaffold cues, underscoring the choice of cell source when evaluating the biocompatibility of a novel scaffold. At the same time, the effective in vivo integration observed across scaffold types emphasizes the importance of host tissue responses for translational evaluation of functional biomaterials. Full article
Show Figures

Graphical abstract

31 pages, 5259 KB  
Article
Innovative Therapy with Stem Cell-Derived Extracellular Vesicles on Cardiac Hypertrophy in an Animal Model of Atherosclerosis; Elucidation of the Molecular Mechanisms Involved in the Repair Process
by Alexandra Vîlcu, Ioana Karla Comarița, Alina Constantin, Nicoleta Alexandru, Miruna Nemecz, Florentina Safciuc, Florina Bojin, Virgil Păunescu and Adriana Georgescu
Biomolecules 2025, 15(10), 1424; https://doi.org/10.3390/biom15101424 - 7 Oct 2025
Viewed by 529
Abstract
(1) Background: The present study investigated the effects of extracellular vesicles (EVs), derived from adipose tissue stem cells (ADSCs) and bone marrow mesenchymal stem cells (BMMSCs), on atherosclerosis-associated cardiac hypertrophy. (2) Methodology: The experiments were performed on hamsters divided into the following groups: [...] Read more.
(1) Background: The present study investigated the effects of extracellular vesicles (EVs), derived from adipose tissue stem cells (ADSCs) and bone marrow mesenchymal stem cells (BMMSCs), on atherosclerosis-associated cardiac hypertrophy. (2) Methodology: The experiments were performed on hamsters divided into the following groups: control (C) fed with a standard diet; hypertensive–hyperlipidemic (HH) generated by combining a diet enriched with 3% cholesterol, 15% butter, and by gavage with 8% NaCl on a daily basis; HH groups injected with EVs (ADSCs) or EVs (BMMSCs), either transfected with Smad2/3 siRNAs or not (HH-EVs (ADSCs), HH-EVs (BMMSCs), HH-EVs (ADSCs) + Smad2/3siRNA, HH-EVs (BMMSCs) + Smad2/3siRNA); and HH group injected with Smad2/3 siRNAs (HH-Smad2/3siRNA). (3) Results: In comparison with the HH group, the findings demonstrated that treatment using EVs (ADSCs or BMMSCs), either with or without Smad2/3 siRNAs, resulted in several significant improvements in the following aspects: the plasma levels of cholesterol, LDL, triglycerides, TGF-β1, and Ang II were decreased; the left ventricular structure and function were recovered; inflammatory markers, ROS, COL1A, α-SMA, Cx43, MIF, ANF, and M1/M2 macrophages, were reduced; the level of key protein NF-κB p50 was diminished. (4) Conclusions: These findings underscore the therapeutic potential of mesenchymal stem cell-derived EVs in atherosclerosis-associated cardiac hypertrophy. Full article
Show Figures

Figure 1

24 pages, 1828 KB  
Review
New Insight into Bone Immunity in Marrow Cavity and Cancellous Bone Microenvironments and Their Regulation
by Hongxu Pu, Lanping Ding, Pinhui Jiang, Guanghao Li, Kai Wang, Jiawei Jiang and Xin Gan
Biomedicines 2025, 13(10), 2426; https://doi.org/10.3390/biomedicines13102426 - 3 Oct 2025
Viewed by 1853
Abstract
Bone immunity represents a dynamic interface where skeletal homeostasis intersects with systemic immune regulation. We synthesize emerging paradigms by contrasting two functionally distinct microenvironments: the marrow cavity, a hematopoietic and immune cell reservoir, and cancellous bone, a metabolically active hub orchestrating osteoimmune interactions. [...] Read more.
Bone immunity represents a dynamic interface where skeletal homeostasis intersects with systemic immune regulation. We synthesize emerging paradigms by contrasting two functionally distinct microenvironments: the marrow cavity, a hematopoietic and immune cell reservoir, and cancellous bone, a metabolically active hub orchestrating osteoimmune interactions. The marrow cavity not only generates innate and adaptive immune cells but also preserves long-term immune memory through stromal-derived chemokines and survival factors, while cancellous bone regulates bone remodeling via macrophage-osteoclast crosstalk and cytokine gradients. Breakthroughs in lymphatic vasculature identification challenge traditional views, revealing cortical and lymphatic networks in cancellous bone that mediate immune surveillance and pathological processes such as cancer metastasis. Central to bone immunity is the neuro–immune–endocrine axis, where sympathetic and parasympathetic signaling bidirectionally modulate osteoclastogenesis and macrophage polarization. Gut microbiota-derived metabolites, including short-chain fatty acids and polyamines, reshape bone immunity through epigenetic and receptor-mediated pathways, bridging systemic metabolism with local immune responses. In disease contexts, dysregulated immune dynamics drive osteoporosis via RANKL/IL-17 hyperactivity and promote leukemic evasion through microenvironmental immunosuppression. We further propose the “brain–gut–bone axis” as a systemic regulatory framework, wherein vagus nerve-mediated gut signaling enhances osteogenic pathways, while leptin and adipokine circuits link marrow adiposity to inflammatory bone loss. These insights redefine bone as a multidimensional immunometabolic organ, integrating neural, endocrine, and microbial inputs to maintain homeostasis. By elucidating the mechanisms of immune-driven bone pathologies, this work highlights therapeutic opportunities through biomaterial-mediated immunomodulation and microbiota-targeted interventions, paving the way for next-generation treatments in osteoimmune disorders. Full article
(This article belongs to the Section Immunology and Immunotherapy)
Show Figures

Figure 1

30 pages, 1280 KB  
Review
Extracellular Vesicle (EV) Proteomics in Corneal Regenerative Medicine
by Zohreh Arabpour, Hanieh Niktinat, Firouze Hatami, Amal Yaghmour, Zarife Jale Yucel, Seyyedehfatemeh Ghalibafan, Hamed Massoumi, Zahra Bibak Bejandi, Majid Salehi, Elmira Jalilian, Mahmood Ghassemi, Victor H. Guaiquil, Mark Rosenblatt and Ali R. Djalilian
Proteomes 2025, 13(4), 49; https://doi.org/10.3390/proteomes13040049 - 3 Oct 2025
Cited by 1 | Viewed by 747
Abstract
Corneal regeneration has gained growing interest in recent years, largely due to the limitations of conventional treatments and the persistent shortage of donor tissue. Among the emerging strategies, extracellular vehicles (EVs), especially those derived from mesenchymal stromal cells (MSCs), have shown great promise [...] Read more.
Corneal regeneration has gained growing interest in recent years, largely due to the limitations of conventional treatments and the persistent shortage of donor tissue. Among the emerging strategies, extracellular vehicles (EVs), especially those derived from mesenchymal stromal cells (MSCs), have shown great promise as a cell-free therapeutic approach. These nanoscale vesicles contribute to corneal healing by modulating inflammation, supporting epithelial and stromal regeneration, and promoting nerve repair. Their therapeutic potential is largely attributed to the diverse and bioactive proteomic cargo they carry, including growth factors, cytokines, and proteins involved in extracellular matrix remodeling. This review presents a comprehensive examination of the proteomic landscape of EVs in the context of corneal regenerative medicine. We explore the biological functions of EVs in corneal epithelial repair, stromal remodeling, and neurodegeneration. In addition, we discuss advanced proteomic profiling techniques such as mass spectrometry (MS) and liquid chromatography–mass spectrometry (LC-MS/MS), which have been used to identify and characterize the protein contents of EVs. This review also compares the proteomic profiles of EVs derived from various MSC sources, including adipose tissue, bone marrow, and umbilical cord, and considers how environmental cues, such as hypoxia and inflammation, influence their protein composition. By consolidating current findings, this article aims to provide valuable insights for advancing the next generation of cell-free therapies for corneal repair and regeneration. Full article
(This article belongs to the Topic Multi-Omics in Precision Medicine)
Show Figures

Figure 1

13 pages, 256 KB  
Review
Biologic Augmentation in Anterior Cruciate Ligament Reconstruction and Beyond: A Review of PRP and BMAC
by Grant M. Pham
J. Clin. Med. 2025, 14(19), 6959; https://doi.org/10.3390/jcm14196959 - 1 Oct 2025
Viewed by 1389
Abstract
This narrative review synthesizes PubMed- and Scopus-indexed studies from 2020 to 2025, including preclinical animal models, prospective cohort studies, and level I and II randomized trials, to compare two leading biologic augmentation strategies: platelet-rich plasma (PRP) and bone marrow aspirate concentrate (BMAC). The [...] Read more.
This narrative review synthesizes PubMed- and Scopus-indexed studies from 2020 to 2025, including preclinical animal models, prospective cohort studies, and level I and II randomized trials, to compare two leading biologic augmentation strategies: platelet-rich plasma (PRP) and bone marrow aspirate concentrate (BMAC). The review examines underlying mechanisms of action, delivery techniques, imaging biomarkers of graft maturation, patient-reported and functional outcomes, safety profiles, cost-effectiveness, and regulatory frameworks. PRP provides early anti-inflammatory and proangiogenic signaling, while BMAC delivers a concentrated population of mesenchymal stem cells and growth factors to the tendon–bone interface. Both modalities consistently enhance MRI-defined graft maturation, yet evidence of long-term functional or biomechanical superiority remains inconclusive. Emerging therapies such as peptide hydrogels, adipose-derived stem cells, and exosome delivery offer promising avenues for future research. Standardized protocols and large multicenter trials are needed to clarify comparative efficacy and inform personalized rehabilitation strategies. Full article
30 pages, 1346 KB  
Review
Electrospun Bio-Scaffolds for Mesenchymal Stem Cell-Mediated Neural Differentiation: Systematic Review of Advances and Future Directions
by Luigi Ruccolo, Aleksandra Evangelista, Marco Benazzo, Bice Conti and Silvia Pisani
Int. J. Mol. Sci. 2025, 26(19), 9528; https://doi.org/10.3390/ijms26199528 - 29 Sep 2025
Viewed by 1224
Abstract
Neural tissue injuries, including spinal cord damage and neurodegenerative diseases, pose a major clinical challenge due to the central nervous system’s limited regenerative capacity. Current treatments focus on stabilization and symptom management rather than functional restoration. Tissue engineering offers new therapeutic perspectives, particularly [...] Read more.
Neural tissue injuries, including spinal cord damage and neurodegenerative diseases, pose a major clinical challenge due to the central nervous system’s limited regenerative capacity. Current treatments focus on stabilization and symptom management rather than functional restoration. Tissue engineering offers new therapeutic perspectives, particularly through the combination of electrospun nanofibrous scaffolds and mesenchymal stem cells (MSCs). Electrospun fibers mimic the neural extracellular matrix, providing topographical and mechanical cues that enhance MSC adhesion, viability, and neural differentiation. MSCs are multipotent stem cells with robust paracrine and immunomodulatory activity, capable of supporting regeneration and, under proper stimuli, acquiring neural-like phenotypes. This systematic review, following the PRISMA 2020 method, analyzes 77 selected articles from the last ten years to assess the potential of electrospun biopolymer scaffolds for MSC-mediated neural repair. We critically examine the scaffold’s composition (synthetic and natural polymers), fiber architecture (alignment and diameter), structural and mechanical properties (porosity and stiffness), and biofunctionalization strategies. The influence of MSC tissue sources (bone marrow, adipose, and dental pulp) on neural differentiation outcomes is also discussed. The results of a literature search show both in vitro and in vivo enhanced neural marker expression, neurite extension, and functional recovery when MSCs are seeded onto optimized electrospun scaffolds. Therefore, integrating stem cell therapy with advanced biomaterials offers a promising route to bridge the gap between neural injury and functional regeneration. Full article
(This article belongs to the Special Issue Tissue Engineering Related Biomaterials: Progress and Challenges)
Show Figures

Figure 1

9 pages, 647 KB  
Brief Report
Mesenchymal Stromal Cell-Derived Extracellular Vesicles for Oral Mucosal Engraftment in Urethral Reconstruction: Influence of Tissue Origin and Culture Growth Phase (Log vs. Stationary) on miRNA Content
by Daisuke Watanabe, Akio Mizushima and Akio Horiguchi
Int. J. Mol. Sci. 2025, 26(19), 9412; https://doi.org/10.3390/ijms26199412 - 26 Sep 2025
Viewed by 565
Abstract
Urethral stricture involves fibrotic narrowing of the urethral mucosa and spongiosum. Although urethroplasty using oral mucosal grafts is the gold standard for complex cases due to its high success rate, technical complexity limits its broader adoption. To address this, endoscopic transplantation of oral [...] Read more.
Urethral stricture involves fibrotic narrowing of the urethral mucosa and spongiosum. Although urethroplasty using oral mucosal grafts is the gold standard for complex cases due to its high success rate, technical complexity limits its broader adoption. To address this, endoscopic transplantation of oral mucosal tissue has been proposed. While feasibility has been demonstrated, clinical efficacy remains suboptimal. Developing adjunctive factors that facilitate mucosal engraftment may improve outcomes of endoscopic transplantation. Extracellular vesicles (EVs)—membrane-bound nanoparticles secreted by cells that deliver miRNAs and other bioactive molecules—have recently emerged as promising candidates. We investigated EVs derived from four mesenchymal stromal cell (MSC) sources—stem cells from human exfoliated deciduous teeth (SHED), adipose tissue, umbilical cord, and bone marrow (BM)—isolated during both logarithmic (log) and stationary culture phases. miRNA profiling revealed distinct phase- and origin-specific signatures. SHED-derived EVs from the log phase and bone marrow-derived EVs from the stationary phase expressed miR-31, the let-7 family, and miR-205, suggesting early wound healing potential. In contrast, stationary-phase SHED-EVs and log-phase BM-MSC-EVs were enriched in the miR-99 family and miR-31, indicating potential roles in epithelial stabilization and fibrosis modulation. These findings support phase-specific application of MSC-EVs to optimize mucosal engraftment in transurethral reconstruction. Full article
Show Figures

Figure 1

13 pages, 1004 KB  
Article
Neutrophil-to-Lymphocyte Ratio, Bone Marrow, and Visceral Fat Metabolism as Predictors of Future Cardiovascular Disease in an Asymptomatic Healthy Population
by Soo Jin Lee, Jahae Kim, Ji Young Kim, Jin Chul Paeng, Yun Young Choi, Young Seo Kim, Kang-Ho Choi, Jeong-Min Kim, Nayeon Choi and Jiyeong Kim
J. Clin. Med. 2025, 14(19), 6709; https://doi.org/10.3390/jcm14196709 - 23 Sep 2025
Viewed by 576
Abstract
Background/Objectives: The neutrophil-to-lymphocyte ratio (NLR), a marker of systemic inflammation, is a known predictor of cardiovascular disease and overall mortality. We examined the relationship between the NLR and the metabolic activity of hematopoietic organs and visceral fat, and their association with the risk [...] Read more.
Background/Objectives: The neutrophil-to-lymphocyte ratio (NLR), a marker of systemic inflammation, is a known predictor of cardiovascular disease and overall mortality. We examined the relationship between the NLR and the metabolic activity of hematopoietic organs and visceral fat, and their association with the risk of atherosclerotic cardiovascular disease (ASCVD) in an asymptomatic healthy population. Methods: We retrospectively analyzed individuals who underwent F-18-fluorodeoxyglucose (FDG) positron emission tomography/computed tomography (PET/CT) as part of their health check-ups. Metabolic activity was quantified using standardized uptake values (SUVs) from the lumbar vertebral bone marrow, spleen, visceral, and subcutaneous fat, normalized to target-to-background ratios (TBRs) using the superior vena cava. NLR was calculated from absolute neutrophil and lymphocyte counts. Correlations between NLR, clinical parameters, organ TBRs, and ASCAD risk were analyzed. Results: Among 303 participants from three hospitals, the median NLR was 1.5 (range: 0.5–5.55). NLR showed weak correlation with the TBRs of bone marrow, visceral fat, and subcutaneous fat, as well as high-density lipoprotein cholesterol and body mass index (BMI). In logistic regression analysis adjusted for age and sex, BMI and the TBRs of bone marrow and visceral fat were independent predictors of elevated NLR (≥ 1.5). When integrating these parameters, NLR demonstrated strong predictive performance for identifying a high ASCVD risk (≥20% over 10 years), with an area under the curve of 0.826. Conclusions: In an asymptomatic healthy population, NLR is associated with FDG metabolic parameters of hematopoietic organs and adipose tissue. These combined measures may serve as valuable marker for identifying individuals at elevated ASCVD risk. Full article
(This article belongs to the Section Cardiovascular Medicine)
Show Figures

Graphical abstract

41 pages, 2467 KB  
Review
Crosstalk Between Skeletal Muscle and Proximal Connective Tissues in Lipid Dysregulation in Obesity and Type 2 Diabetes
by Nataša Pollak, Efua Gyakye Janežič, Žiga Šink and Chiedozie Kenneth Ugwoke
Metabolites 2025, 15(9), 581; https://doi.org/10.3390/metabo15090581 - 30 Aug 2025
Viewed by 1646
Abstract
Background/Objectives: Obesity and type 2 diabetes mellitus (T2DM) profoundly disrupt lipid metabolism within local microenvironments of skeletal muscle and its associated connective tissues, including adipose tissue, bone, and fascia. However, the role of local communication between skeletal muscle and its proximal connective tissues [...] Read more.
Background/Objectives: Obesity and type 2 diabetes mellitus (T2DM) profoundly disrupt lipid metabolism within local microenvironments of skeletal muscle and its associated connective tissues, including adipose tissue, bone, and fascia. However, the role of local communication between skeletal muscle and its proximal connective tissues in propagating metabolic dysfunction is incompletely understood. This narrative review synthesizes current evidence on these local metabolic interactions, highlighting novel insights and existing gaps. Methods: We conducted a comprehensive literature analysis of primary research published in the last decade, sourced from PubMed, Web of Science, and ScienceDirect. Studies were selected for relevance to skeletal muscle, adipose tissue, fascia, and bone lipid metabolism in the context of obesity and T2DM, with emphasis on molecular, cellular, and paracrine mechanisms of local crosstalk. Findings were organized into thematic sections addressing physiological regulation, pathological remodeling, and inter-organ signaling pathways. Results: Our synthesis reveals that local lipid dysregulation in obesity and T2DM involves altered fatty acid transporter dynamics, mitochondrial overload, fibro-adipogenic remodeling, and compartment-specific adipose tissue dysfunction. Crosstalk via myokines, adipokines, osteokines, bioactive lipids, and exosomal miRNAs integrates metabolic responses across these tissues, amplifying insulin resistance and lipotoxic stress. Emerging evidence highlights the underappreciated roles of fascia and marrow adipocytes in regional lipid handling. Conclusions: Collectively, these insights underscore the pivotal role of inter-tissue crosstalk among skeletal muscle, adipose tissue, bone, and fascia in orchestrating lipid-induced insulin resistance, and highlight the need for integrative strategies that target this multicompartmental network to mitigate metabolic dysfunction in obesity and T2DM. Full article
(This article belongs to the Special Issue Lipid Metabolism Disorders in Obesity)
Show Figures

Graphical abstract

25 pages, 3822 KB  
Article
Comparative Transcriptome and MicroRNA Profiles of Equine Mesenchymal Stem Cells, Fibroblasts, and Their Extracellular Vesicles
by Sebastian Sawicki, Monika Bugno-Poniewierska, Jakub Żurowski, Tomasz Szmatoła, Ewelina Semik-Gurgul, Michał Bochenek, Elżbieta Karnas and Artur Gurgul
Genes 2025, 16(8), 936; https://doi.org/10.3390/genes16080936 - 5 Aug 2025
Viewed by 1747
Abstract
Background: Mesenchymal stem cells (MSCs) are a promising tool in regenerative medicine due to their ability to secrete paracrine factors that modulate tissue repair. Extracellular vesicles (EVs) released by MSCs contain bioactive molecules (e.g., mRNAs, miRNAs, proteins) and play a key role in [...] Read more.
Background: Mesenchymal stem cells (MSCs) are a promising tool in regenerative medicine due to their ability to secrete paracrine factors that modulate tissue repair. Extracellular vesicles (EVs) released by MSCs contain bioactive molecules (e.g., mRNAs, miRNAs, proteins) and play a key role in intercellular communication. Methods: This study compared the transcriptomic profiles (mRNA and miRNA) of equine MSCs derived from adipose tissue (AT-MSCs), bone marrow (BM-MSCs), and ovarian fibroblasts (as a differentiated control). Additionally, miRNAs present in EVs secreted by these cells were characterized using next-generation sequencing. Results: All cell types met ISCT criteria for MSCs, including CD90 expression, lack of MHC II, trilineage differentiation, and adherence. EVs were isolated using ultracentrifugation and validated with nanoparticle tracking analysis and flow cytometry (CD63, CD81). Differential expression analysis revealed distinct mRNA and miRNA profiles across cell types and their secreted EVs, correlating with tissue origin. BM-MSCs showed unique regulation of genes linked to early development and osteogenesis. EVs contained diverse RNA species, including miRNA, mRNA, lncRNA, rRNA, and others. In total, 227 and 256 mature miRNAs were detected in BM-MSCs and AT-MSCs, respectively, including two novel miRNAs per MSC type. Fibroblasts expressed 209 mature miRNAs, including one novel miRNA also found in MSCs. Compared to fibroblasts, 60 and 92 differentially expressed miRNAs were identified in AT-MSCs and BM-MSCs, respectively. Conclusions: The results indicate that MSC tissue origin influences both transcriptomic profiles and EV miRNA content, which may help to interpret their therapeutic potential. Identifying key mRNAs and miRNAs could aid in future optimizing of MSC-based therapies in horses. Full article
(This article belongs to the Section Animal Genetics and Genomics)
Show Figures

Figure 1

14 pages, 2147 KB  
Article
Administration of Adipose-Derived Stem Cells Lowers the Initial Levels of IL6 and TNF-Alpha in the Rat Model of Necrotizing Enterocolitis
by Marek Wolski, Tomasz Ciesielski, Kasper Buczma, Łukasz Fus, Agnieszka Girstun, Joanna Trzcińska-Danielewicz and Agnieszka Cudnoch-Jędrzejewska
Int. J. Mol. Sci. 2025, 26(14), 6555; https://doi.org/10.3390/ijms26146555 - 8 Jul 2025
Viewed by 717
Abstract
Research in the field of stem cells in necrotizing enterocolitis has primarily focused on the curative role of specific cells—mostly bone marrow and amniotic fluid stem cells. The impact of stem cells on reducing inflammatory cytokine levels in the necrotizing enterocolitis (NEC) model [...] Read more.
Research in the field of stem cells in necrotizing enterocolitis has primarily focused on the curative role of specific cells—mostly bone marrow and amniotic fluid stem cells. The impact of stem cells on reducing inflammatory cytokine levels in the necrotizing enterocolitis (NEC) model has been studied in accordance with the effects they pose on histopathology. Taking into consideration the possible paracrine mechanism of action of stem cells, our group hypothesized that lowering the initial levels of proinflammatory cytokines may be one of the mechanisms affecting the clinical outcome. A self-modified rat NEC model was used to show the effect of intraperitoneal administration of adipose derived stem cells on the initial levels of interleukin-6 (IL-6) and tumor necrosis factor-alpha (TNF-alfa) in comparison with the interleukin levels in NEC animals and control animals without adipose–derived stem cells (ADSCs) injection. We showed a statistically significant difference in the levels of interleukins when comparing an ADSC injected group and an NEC group. This suggests that one of the mechanisms in which stem cells impact the clinical outcomes in NEC may be by alleviating the initial levels of proinflammatory cytokines. Full article
(This article belongs to the Special Issue The Role of Cytokines in Health and Diseases)
Show Figures

Figure 1

15 pages, 724 KB  
Review
Adipose Tissue-Derived Mediators in Multiple Myeloma: Linking Obesity to Bone Disease via Inflammatory Pathways
by Alexandra-Ştefania Stroe-Ionescu, Alina Daniela Tǎnase, Ionela Rotaru, Janina-Georgiana Goanțǎ, Ana Maria Pǎtraşcu, Mihail Virgil Boldeanu, Mohamed-Zakaria Assani, Isabela Siloși, Lidia Boldeanu and Daniela-Teodora Maria
Int. J. Mol. Sci. 2025, 26(12), 5618; https://doi.org/10.3390/ijms26125618 - 11 Jun 2025
Viewed by 1261
Abstract
In patients diagnosed with multiple myeloma (MM), the primary complaints at the time of diagnosis are often related to bone involvement, significantly impacting quality of life and increasing both morbidity and mortality. Obesity is associated with a chronic inflammatory state that results in [...] Read more.
In patients diagnosed with multiple myeloma (MM), the primary complaints at the time of diagnosis are often related to bone involvement, significantly impacting quality of life and increasing both morbidity and mortality. Obesity is associated with a chronic inflammatory state that results in the production of various cytokines and adipokines, which may promote bone destruction. Adiponectin, an adipokine predominantly secreted by adipocytes, is notably diminished in circulation among individuals with obesity, a phenomenon that has also been observed in MM. This reduction may contribute to the disruption of an already compromised bone architecture. The increase in adipose tissue is associated with heightened leptin production, a key adipokine, which can play a significant role in the pathophysiology of MM and its related bone complications. Obesity is associated with hyperinsulinemia and increased levels of free IGF-1. In MM, IGF-1 plays a critical role as a growth factor, produced by both myeloma cells and osteoclasts within the bone marrow microenvironment. Our gathered data indicates a significant relationship between the adipokines produced by adipose tissue and the bone matrix, particularly in the context of obesity and MM. However, it is important to note that the existing body of research on this topic is relatively sparse, with the majority of studies conducted on murine models rather than human subjects. This limitation highlights a critical need for further investigation to elucidate the precise mechanisms that contribute to bone destruction under these conditions. Full article
(This article belongs to the Section Molecular Oncology)
Show Figures

Figure 1

19 pages, 3292 KB  
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
Cited by 2 | Viewed by 694
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

20 pages, 8483 KB  
Article
Comparative Efficacy of Exosomes Derived from Different Mesenchymal Stem Cell Sources in Osteoarthritis Models: An In Vitro and Ex Vivo Analysis
by Jaishree Sankaranarayanan, Hyung Keun Kim, Ju Yeon Kang, Sree Samanvitha Kuppa, Hong Yeol Yang and Jong Keun Seon
Int. J. Mol. Sci. 2025, 26(12), 5447; https://doi.org/10.3390/ijms26125447 - 6 Jun 2025
Cited by 1 | Viewed by 2833
Abstract
Osteoarthritis (OA) is a prevalent and debilitating joint disorder that affects a substantial proportion of the global population, underscoring the urgent need for therapeutic strategies that extend beyond symptomatic management. Although mesenchymal stem cells (MSCs) have emerged as a promising therapeutic modality, their [...] Read more.
Osteoarthritis (OA) is a prevalent and debilitating joint disorder that affects a substantial proportion of the global population, underscoring the urgent need for therapeutic strategies that extend beyond symptomatic management. Although mesenchymal stem cells (MSCs) have emerged as a promising therapeutic modality, their clinical application remains constrained by several inherent limitations. This study explores a cell-free alternative by investigating the therapeutic potential of exosomes derived from bone marrow (BMSCs), adipose tissue (ADSCs), and umbilical cord (UMSCs) MSCs in mitigating OA pathogenesis, utilizing both in vitro and ex vivo models. Exosomes from each MSC source were isolated and characterized through nanoparticle tracking analysis, transmission electron microscopy, and Western blotting to confirm their identity and purity. Subsequently, their chondroprotective, anti-inflammatory, and regenerative properties were systematically assessed through evaluations of cell viability, expression profiles of inflammatory and chondroprotective markers, and chondrocyte migration assays. The results demonstrate that all three types of MSC-derived exosomes (MSC-Exos) exhibit low cytotoxicity while significantly suppressing proinflammatory markers and enhancing the expression of chondroprotective genes. Notably, BMSC-Exos and UMSC-Exos displayed superior efficacy in attenuating inflammation, promoting cartilage protection, and inhibiting chondrocyte apoptosis. Furthermore, all MSC-Exos markedly enhanced chondrocyte motility, a critical component of cartilage repair. Collectively, these findings support the therapeutic promise of MSC-Exos, particularly those derived from BMSCs and UMSCs, as a targeted, cell-free approach for the treatment of OA compared to ADSCs. By modulating inflammation, promoting cartilage regeneration, and preventing chondrocyte apoptosis, MSC-Exos may serve as a viable and scalable alternative to current MSC-based therapies for this widespread degenerative disease. Full article
(This article belongs to the Special Issue Molecular Advances and Perspectives in Rheumatic Diseases)
Show Figures

Figure 1

Back to TopTop