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
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
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (1,536)

Search Parameters:
Keywords = adipose stem cell

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
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
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

19 pages, 4195 KB  
Article
When Fat Talks: How Adipose-Derived Extracellular Vesicles Fuel Breast Cancer
by Maria Pia Cavaleri, Tommaso Pusceddu, Lucia Sileo, Luna Ardondi, Ilaria Vitali, Ilenia Pia Cappucci, Laura Basile, Giuseppe Pezzotti, Francesco Fiorica, Letizia Ferroni and Barbara Zavan
Int. J. Mol. Sci. 2025, 26(19), 9666; https://doi.org/10.3390/ijms26199666 - 3 Oct 2025
Viewed by 275
Abstract
Adipose tissue plays a crucial role in the tumor microenvironment (TME), where its secreted extracellular vesicles (EVs) are involved in the complex signaling between tumor cells and surrounding stromal components. This study aims to unravel the mechanisms through which adipocyte-derived EVs influence breast [...] Read more.
Adipose tissue plays a crucial role in the tumor microenvironment (TME), where its secreted extracellular vesicles (EVs) are involved in the complex signaling between tumor cells and surrounding stromal components. This study aims to unravel the mechanisms through which adipocyte-derived EVs influence breast cancer (BC) progression. Human mesenchymal stem cells (hMSCs) were differentiated into adipocytes following a 21-day induction protocol that led to significant accumulation of lipid droplets within the cells. EVs were isolated from the conditioned medium of both hMSC-derived adipocytes and BC cells. Particle size distribution, morphology, and uptake into the recipient cell were investigated via nanoparticle tracking analysis, transmission electron microscopy, and fluorescence microscopy, respectively. Our results show that BC-derived EVs notably impaired cell viability and modulated the expression of key genes involved in apoptosis resistance within stromal cells. On the other hand, stromal-derived EVs significantly altered tumor cell behavior, indicating a dynamic, bidirectional exchange of bioactive signals. These findings underscore the pivotal role of EV-mediated communication in the tumor-stroma interplay, suggesting that adipocyte-cancer cell EV crosstalk contributes to the remodeling of the TME, potentially facilitating tumor progression. Full article
Show Figures

Figure 1

26 pages, 3796 KB  
Article
T-Cadherin Finetunes Proliferation–Differentiation During Adipogenesis via PI3K–AKT Signaling Pathway
by Polina Klimovich, Ilya Brodsky, Valentina Dzreyan, Marianna Ivleva, Olga Grigorieva, Mark Meshcheriakov, Ekaterina Semina, Veronika Sysoeva, Vsevolod Tkachuk and Kseniya Rubina
Int. J. Mol. Sci. 2025, 26(19), 9646; https://doi.org/10.3390/ijms26199646 - 2 Oct 2025
Viewed by 203
Abstract
Adipose tissue renewal requires precise coordination of stem/progenitor cell proliferation, preadipocyte commitment, and terminal adipocyte differentiation. T-cadherin (CDH13), an atypical GPI-anchored cadherin, is expressed in adipose tissue and functions as a receptor for high-molecular-weight (HMW) adiponectin—a key adipokine produced by adipose tissue and [...] Read more.
Adipose tissue renewal requires precise coordination of stem/progenitor cell proliferation, preadipocyte commitment, and terminal adipocyte differentiation. T-cadherin (CDH13), an atypical GPI-anchored cadherin, is expressed in adipose tissue and functions as a receptor for high-molecular-weight (HMW) adiponectin—a key adipokine produced by adipose tissue and involved in metabolic regulation. While T-cadherin is implicated in cardiovascular and metabolic homeostasis, its role in adipogenesis still remains poorly understood. In this study, we used the 3T3-L1 preadipocyte model to investigate the function of T-cadherin in adipocyte differentiation. We analyzed T-cadherin expression dynamics during differentiation and assessed how T-cadherin overexpression or knockdown affects lipid accumulation, expression of adipogenic markers, and key signaling pathways including ERK, PI3K–AKT, AMPK, and mTOR. Our findings demonstrate that T-cadherin acts as a negative regulator of adipogenesis. T-cadherin overexpression ensured a proliferative, undifferentiated cell state, delaying early adipogenic differentiation and suppressing both lipid droplet accumulation and the expression of adipogenic markers. In contrast, T-cadherin downregulation accelerated differentiation, enhanced lipid accumulation, and increased insulin responsiveness, as indicated by PI3K–AKT pathway activation at specific stages of adipogenesis. These results position T-cadherin as a key modulator of adipose tissue plasticity, regulating the balance between progenitor expansion and terminal differentiation, with potential relevance to obesity and metabolic disease. Full article
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 552
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 485
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

12 pages, 704 KB  
Article
AI-Based 3D-Segmentation Quantifies Sarcopenia in Multiple Myeloma Patients
by Thuy-Duong Do, Tobias Nonnenmacher, Marieke Burghardt, Stefanie Zschaebitz, Marina Hajiyianni, Elias Karl Mai, Marc-Steffen Raab, Carsten Müller-Tidow, Hans-Ulrich Kauczor, Hartmut Goldschmidt and Ulrike Dapunt
Diagnostics 2025, 15(19), 2466; https://doi.org/10.3390/diagnostics15192466 - 26 Sep 2025
Viewed by 297
Abstract
Background: Sarcopenia is characterized by a loss of muscle mass and strength, resulting in functional limitations and an increased risk of falls, injuries and fractures. The aim of this study was to obtain detailed information on skeletal muscle changes in patients with multiple [...] Read more.
Background: Sarcopenia is characterized by a loss of muscle mass and strength, resulting in functional limitations and an increased risk of falls, injuries and fractures. The aim of this study was to obtain detailed information on skeletal muscle changes in patients with multiple myeloma (MM) during treatment. Methods: A total of 51 patients diagnosed with MM who had undergone whole-body low-dose computed tomography acquisition prior to induction therapy (T1) and post autologous stem cell transplantation (T2) were examined retrospectively. Total volume (TV), muscle volume (MV) and intramuscular adipose tissue volume (IMAT) of the autochthonous back muscles, the iliopsoas muscle and the gluteal muscles were evaluated on the basis of the resulting masks of the BOA tool with the fully automated combination of TotalSegmentator and a body composition analysis. An in-house trained artificial intelligence network was used to obtain a fully automated three-dimensional segmentation assessment. Results: Patients’ median age was 58 years (IQR 52–66), 38 were male and follow-up CT-scans were performed after a mean of 11.8 months (SD ± 3). Changes in MV and IMAT correlated significantly with Body-Mass-Index (BMI) (r = 0.7, p < 0.0001). Patients (n = 28) with a decrease in BMI (mean −2.2 kg/m2) during therapy lost MV (T1: 3419 cm3, IQR 3176–4000 cm3 vs. T2: 3226 cm3, IQR 3014–3662 cm3, p < 0.0001) whereas patients (n = 20) with an increased BMI (mean +1.4 kg/m2) showed an increase in IMAT (T1: 122 cm3, IQR 96.8–202.8 cm3 vs. T2: 145.5 cm3, IQR 115–248 cm3, p = 0.0002). Loss of MV varied between different muscle groups and was most prominent in the iliopsoas muscle (−9.8%) > gluteus maximus (−9.1%) > gluteus medius (−5.8%) > autochthonous back muscles (−4.3%) > gluteus minimus (−1.5%). Increase in IMAT in patients who gained weight was similar between muscle groups. Conclusions: The artificial intelligence-based three-dimensional segmentation process is a reliable and time-saving method to acquire in-depth information on sarcopenia in MM patients. Loss of MV and increase in IMAT were reliably detectable and associated with changes in BMI. Loss of MV was highest in muscles with more type 2 muscle fibers (fast-twitch, high energy) whereas muscles with predominantly type 1 fibers (slow-twitch, postural control) were less affected. This study provides valuable insight into muscle changes of MM patients during treatment, which might aid in tailoring exercise interventions more precisely to patients’ needs. Full article
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 274
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

25 pages, 14471 KB  
Article
A Novel Human Stem Cell Culture Model for Severe Traumatic Brain Injury Reflecting Sexual Dimorphism in Heterotopic Ossification
by Jonas Joneleit, Philipp Leimkühler, Tarek Niemann, Matthias Ruwe, Christian Jantos, Dirk Wähnert, Christian Kaltschmidt, Thomas Vordemvenne and Barbara Kaltschmidt
Cells 2025, 14(19), 1491; https://doi.org/10.3390/cells14191491 - 24 Sep 2025
Viewed by 361
Abstract
Heterotopic ossification (HO) is a disease characterized by ectopic bone formation, which can occur following severe traumatic brain injury (TBI). However, the underlying mechanisms remain poorly understood. In this study, we established a stem cell model using adipose-derived stem cells (ADSCs) and skeletal [...] Read more.
Heterotopic ossification (HO) is a disease characterized by ectopic bone formation, which can occur following severe traumatic brain injury (TBI). However, the underlying mechanisms remain poorly understood. In this study, we established a stem cell model using adipose-derived stem cells (ADSCs) and skeletal stem cells (SSCs) to examine osteogenic factors present in the sera of TBI patients. Incubation of ADSCs and SSCs with osteoinductive medium supplemented with TBI serum significantly enhanced osteogenic differentiation, particularly in male ADSCs and both female and male SSCs, with male SSCs exhibiting the highest osteogenic potential. Furthermore, we identified TGF-β1 as an important factor involved in these osteogenic processes. Elevated levels of TGF-β1 were detected in the serum of male TBI patients 14 days post-injury. Cellular assays revealed a sexual dimorphism in response to TGF-β1 neutralization: osteogenic differentiation in male SSCs was significantly reduced, while no effect was detectable in female SSCs. These findings, together with the rarity of HO in female patients, suggest that TGF-β1 plays a central role in the development of HO in males. Furthermore, this study highlights the importance of considering sex-specific mechanisms in traumatic HO for the development of sex-specific therapy options. Full article
Show Figures

Graphical abstract

17 pages, 1443 KB  
Brief Report
Priming Canine Adipose Tissue-Derived Mesenchymal Stem Cells with CBD-Rich Cannabis Extract Modulates Neurotrophic Factors Expression Profile
by Vinicius Skau Perino, Lucas Vinícius de Oliveira Ferreira, Beatriz da Costa Kamura, Natielly Dias Chimenes, Alisson Vinícius Gimenes Olbera, Thiago Tourinho Pereira, Aline Márcia Marques Braz, Marjorie de Assis Golim, Márcio de Carvalho and Rogério Martins Amorim
Vet. Sci. 2025, 12(10), 926; https://doi.org/10.3390/vetsci12100926 - 24 Sep 2025
Viewed by 316
Abstract
The endocannabinoid system regulates key biological functions such as neuroprotection, pain modulation, inflammation, and immunomodulation. Cannabis-based therapies have gained attention due to the therapeutic potential of their bioactive compounds, particularly phytocannabinoids like cannabidiol (CBD), which exhibit anti-inflammatory, neuroprotective, and immunomodulatory properties. Mesenchymal stem [...] Read more.
The endocannabinoid system regulates key biological functions such as neuroprotection, pain modulation, inflammation, and immunomodulation. Cannabis-based therapies have gained attention due to the therapeutic potential of their bioactive compounds, particularly phytocannabinoids like cannabidiol (CBD), which exhibit anti-inflammatory, neuroprotective, and immunomodulatory properties. Mesenchymal stem cells (MSCs) are widely studied for their regenerative and immunomodulatory potential. This study evaluated the effects of priming canine adipose tissue-derived MSCs (cAT-MSCs) with a CBD-rich cannabis extract on cell morphology, viability, neurotrophic factor gene expression, and cytokine gene and protein expression. cAT-MSCs (n = 5) were primed for 24 h and divided into three groups: Control (C, unprimed), D1 (2.25 µM CBD), and D2 (225 nM CBD). No morphological or viability changes were observed. Gene expression analysis showed that groups D1 and D2 exhibited increased HGF expression. D1 also showed increased IDO and decreased BDNF expression. In contrast, no significant changes were observed in GDNF, IL-10, TNF-α, IFN-γ, or PTGES2. Regarding the cytokine profile, GM-CSF, IL-2, and IL-10 were undetectable. Notably, IL-8 and MCP-1 levels were significantly reduced in D1 compared to the control. These findings suggest that CBD priming modulates key regenerative and inflammatory mediators in cAT-MSCs, supporting its potential application in enhancing the efficacy of cell-based therapies. Full article
(This article belongs to the Special Issue Animal Anesthesia and Analgesia Management: Pain Relief and Sedation)
Show Figures

Figure 1

16 pages, 1758 KB  
Article
Hepatocyte Growth Factor-Mediated Chondrocyte Proliferation Induced by Adipose-Derived MSCs from Osteoarthritis Patients and Its Synergistic Enhancement by Hyaluronic Acid
by Samuel Jaeyoon Won, Hyun-Joo Lee, Dae-Yong Kim, Hyeonjeong Noh, Song yi Lee, Ji Ae Yoo, Yoon Sang Jeon, Heebeom Shin and Dong Jin Ryu
Int. J. Mol. Sci. 2025, 26(19), 9296; https://doi.org/10.3390/ijms26199296 - 23 Sep 2025
Viewed by 278
Abstract
Mesenchymal stem cells (MSCs) spontaneously assemble into three-dimensional (3D) spheroids under matrix-deficient conditions such as the synovial cavity, although their functional significance has yet to be fully elucidated. In this study, we used concave microwell cultures to promote the spontaneous aggregation of adipose-derived [...] Read more.
Mesenchymal stem cells (MSCs) spontaneously assemble into three-dimensional (3D) spheroids under matrix-deficient conditions such as the synovial cavity, although their functional significance has yet to be fully elucidated. In this study, we used concave microwell cultures to promote the spontaneous aggregation of adipose-derived MSCs (ASCs) from OA patients, thereby mimicking the intra-articular microenvironment. We analyzed the paracrine factors of ASC aggregates and compared it with that of conventional 2D monolayer cultures. Notably, 3D aggregation significantly increased the secretion of HGF and VEGF, whereas FGF2 levels remained relatively unchanged. These results indicate that the structural characteristics of ASC aggregates enhance the secretion of key paracrine factors involved in angiogenesis and tissue repair. To functionally evaluate the biological relevance of the secreted factors, conditioned media (CM) from ASC aggregates were applied to human articular chondrocytes. The CM significantly promoted chondrocyte proliferation, an effect that was abolished by the addition of HGF-neutralizing antibodies, thereby highlighting HGF as a central mediator of the regenerative response. Additionally, we further explored whether extracellular factors could modulate growth factor expression such as HGF. In this context, we investigated the impact of low-concentration hyaluronic acid (HA), a key synovial component widely used in OA treatment. Co-treatment with HA not only amplified the expression and secretion of HGF, VEGF, and FGF2, but also promoted ASC proliferation. ASCs forming functional aggregates may exert regenerative effects as active paracrine modulators, and the addition of low-dose hyaluronic acid is expected to further enhance this function, offering a promising strategy for MSC-based osteoarthritis therapy. Full article
(This article belongs to the Special Issue Stem Cells in Health and Disease: 3rd Edition)
Show Figures

Figure 1

15 pages, 2434 KB  
Article
Hybrid Fractional Laser and Autologous Lipofilling: A Synergistic Strategy for Functional and Aesthetic Scar Remodeling
by Gabriele Delia, Lucia Quattrocchi, Pietro Micieli, Damiano Tambasco, Roberta Albanese and Fabiana Battaglia
J. Clin. Med. 2025, 14(19), 6708; https://doi.org/10.3390/jcm14196708 - 23 Sep 2025
Viewed by 348
Abstract
Background: Scar management remains a significant challenge in plastic and reconstructive surgery, particularly when addressing atrophic, retractile, or fibrotic scars. Autologous fat grafting and hybrid fractional laser therapy have independently shown efficacy in improving scar quality. This study aims to evaluate the synergistic [...] Read more.
Background: Scar management remains a significant challenge in plastic and reconstructive surgery, particularly when addressing atrophic, retractile, or fibrotic scars. Autologous fat grafting and hybrid fractional laser therapy have independently shown efficacy in improving scar quality. This study aims to evaluate the synergistic effect of their combination on clinical and functional scar outcomes. Methods: A prospective, comparative study was conducted on patients with cutaneous scars of various etiologies. Participants were treated with either hybrid fractional laser therapy alone (CO2 and 1570 nm Erbium-glass wavelengths) or a combined protocol of laser plus autologous lipofilling. Clinical outcomes were assessed at baseline and at 30, 60, and 90 days post-treatment using the Vancouver Scar Scale (VSS), patient satisfaction scores, and Visual Analog Scale (VAS) for pain and discomfort. Results: Patients receiving the combined treatment demonstrated significantly greater improvement in scar pigmentation, elasticity, pliability, and thickness compared to those treated with laser alone. Subjective symptoms, including pain and itching, were also more effectively alleviated. The volumetric and regenerative properties of adipose tissue, particularly its content of adipose-derived stem cells (ADSCs) and stromal vascular fraction (SVF), likely contributed to the enhanced outcomes observed. Conclusions: The combination of hybrid fractional laser therapy and autologous lipofilling offers a superior therapeutic strategy for scar remodeling compared to laser monotherapy. This integrated regenerative approach addresses both structural and biological aspects of scar tissue, making it a valuable protocol for personalized and effective scar management. Further randomized trials with larger sample sizes and histological validation are warranted to confirm these preliminary findings and refine therapeutic protocols. Full article
Show Figures

Figure 1

13 pages, 8472 KB  
Article
Radiation-Induced EMT of Adipose-Derived Stem Cells in 3D Organotypic Culture via Notch Signaling Pathway
by Seon Jeong Choi, Meesun Kim, Kyung Tae Chung and Tae Gen Son
Biology 2025, 14(9), 1306; https://doi.org/10.3390/biology14091306 - 22 Sep 2025
Viewed by 301
Abstract
In our previous study, adipose-derived stem cells (ASCs) cultured in a three-dimensional (3D) organotypic system exhibited mesenchymal-to-epithelial transition (MET) features, including cobblestone morphology and increased expression of E-cadherin and CK18. In this study, we investigated whether ionizing radiation could reverse this phenotype via [...] Read more.
In our previous study, adipose-derived stem cells (ASCs) cultured in a three-dimensional (3D) organotypic system exhibited mesenchymal-to-epithelial transition (MET) features, including cobblestone morphology and increased expression of E-cadherin and CK18. In this study, we investigated whether ionizing radiation could reverse this phenotype via epithelial–mesenchymal transition (EMT) and examined the involvement of Notch signaling. Mouse ASCs were cultured in Matrigel-based 3D organotypic conditions and exposed to 8 Gy of γ-radiation, and EMT- and Notch-related gene and protein expression were assessed 96 h post-irradiation using ATP viability assays, RT-qPCR, and Western blotting. Exposure to 8 Gy significantly reduced cell viability in 2D ASCs to 49.50 ± 6.50% compared with 61.02 ± 5.77% in 3D organoids (p < 0.0001). Irradiated 3D organoids showed EMT-like changes, including an increase of ~2.5-fold in fibronectin and an increase of ~2.0-fold in Twist1 expression, while epithelial CK18 was modestly elevated. Notch signaling was concurrently activated, with Notch1 and Jagged1 increasing by more than twofold and Fra-1 being significantly upregulated. Pretreatment with 20 μM of the γ-secretase inhibitor (GSI) kept cell viability above 90% and suppressed radiation-induced fibronectin, Twist1, Notch1, and Jagged1 expression. These findings indicate that ionizing radiation promotes EMT in 3D-cultured ASCs and reverses prior epithelialization, with Notch signaling playing a key regulatory role. The 3D ASC organoid model may thus provide a physiologically relevant platform for investigating radiation-induced plasticity and potential antifibrotic interventions. Full article
Show Figures

Figure 1

7 pages, 198 KB  
Editorial
Human Stem Cells in Disease Modelling and Treatment: Bridging the Gap Between Bench and Bedside
by Alvaro Plaza Reyes and Sofia M. Calado
Biomedicines 2025, 13(9), 2313; https://doi.org/10.3390/biomedicines13092313 - 22 Sep 2025
Viewed by 523
Abstract
Human stem cell research is entering a stage where disease modeling, translational applications, and clinical therapies are increasingly connected. This editorial provides an overview of the contributions included in this Special Issue, titled “Human Stem Cells in Disease Modelling and Treatment”, placing them [...] Read more.
Human stem cell research is entering a stage where disease modeling, translational applications, and clinical therapies are increasingly connected. This editorial provides an overview of the contributions included in this Special Issue, titled “Human Stem Cells in Disease Modelling and Treatment”, placing them within the wider landscape of stem cell science. We summarize advances in ovarian stem cells for infertility, mesenchymal stem cells for neurodegeneration, pluripotent stem cell-derived cardiovascular and kidney organoids, adipose-derived stem cells, and emerging immunomodulatory and neural progenitor approaches. These studies illustrate the breadth of stem cell research and its potential to inform clinical practice. At the same time, challenges remain in reproducibility, safety, scalability, and ethical oversight. Looking forward, collaborative work and harmonized global standards will be important to bring laboratory findings into therapies that are safe, effective, and accessible. This editorial closes the first edition of the Special Issue with a reflection on current progress and directions for the future. Full article
(This article belongs to the Special Issue Human Stem Cells in Disease Modelling and Treatment)
27 pages, 1088 KB  
Article
Clinical and dGEMRIC Evaluation of Microfragmented Adipose Tissue Versus Hyaluronic Acid in Inflammatory Phenotype of Knee Osteoarthritis: A Randomized Controlled Trial
by Vilim Molnar, Željko Jeleč, Eduard Rod, Damir Hudetz, Petar Brlek, Igor Borić, Vid Matišić, Jana Mešić, Eduard Stjepan Pavelić, Dinko Vidović, Dejan Blažević, Fabijan Čukelj, Srećko Sabalić, Josip Štivičić, Tomislav Dujmović, Mario Starešinić, Martin Čemerin, David Glavaš Weinberger, Iva Molnar, Martina Smolić and Dragan Primoracadd Show full author list remove Hide full author list
Biomedicines 2025, 13(9), 2301; https://doi.org/10.3390/biomedicines13092301 - 19 Sep 2025
Viewed by 739
Abstract
Background: Knee osteoarthritis (OA) is a leading cause of disability, with limited therapies that modify both symptoms and structural degeneration. Autologous microfragmented adipose tissue (MFAT) has emerged as a promising regenerative option, especially in phenotypically distinct OA subgroups. This randomized controlled trial [...] Read more.
Background: Knee osteoarthritis (OA) is a leading cause of disability, with limited therapies that modify both symptoms and structural degeneration. Autologous microfragmented adipose tissue (MFAT) has emerged as a promising regenerative option, especially in phenotypically distinct OA subgroups. This randomized controlled trial evaluated the clinical and structural efficacy of intra-articular MFAT versus hyaluronic acid (HA) in patients with early to moderate inflammatory phenotype knee OA. Methods: Fifty-three patients were randomized in a 2:1 ratio to receive either MFAT (n = 35) or HA (n = 18). Patients were followed-up for six months post-injection and evaluated using patient-reported outcome measures (KOOS, WOMAC, VAS) and delayed gadolinium-enhanced MRI of cartilage (dGEMRIC). A responder analysis defined structural response as ≥10% increase in dGEMRIC in ≥3 of 7 predefined cartilage regions. Results: Both MFAT and HA led to statistically significant improvements in clinical scores and cartilage glycosaminoglycan content. MFAT showed greater mean improvements across most clinical and dGEMRIC measures, although without reaching statistical significance, except for KOOS Symptoms (MFAT: +25.0 vs. HA: +12.7, p = 0.008). Responder-level analysis revealed that all patients who demonstrated structural response also experienced clinically meaningful pain improvement (KOOS Pain ≥ 10), while no patient showed structural benefit without parallel symptomatic relief. Conclusions: MFAT led to greater improvement in symptoms related to joint stiffness, swelling, and crepitus compared to HA, reflecting its potential benefit in targeting the inflammatory features of knee OA. Importantly, HA also led to significant clinical and structural improvements, supporting its continued role as a standard-of-care comparator in knee OA management. Furthermore, the correlation between dGEMRIC and clinical response suggests its utility as a predictive biomarker of treatment success. Full article
Show Figures

Figure 1

15 pages, 1571 KB  
Article
Autologous Micro-Fragmented Adipose Tissue (MFAT) Injections May Be an Effective Treatment for Advanced Knee Osteoarthritis: A Longitudinal Study
by Joachim De Groote, Caro Roten, Elizaveta Fomenko, Pascal Coorevits, André Harth and Yves Depaepe
J. Clin. Med. 2025, 14(18), 6571; https://doi.org/10.3390/jcm14186571 - 18 Sep 2025
Viewed by 430
Abstract
Background/Objectives: Knee osteoarthritis (OA) is a major cause of pain and functional disability worldwide, leading to a growing interest in more durable and less invasive therapies. Micro-fragmented adipose tissue (MFAT) injections have emerged as a promising frontier in regenerative therapies using mesenchymal [...] Read more.
Background/Objectives: Knee osteoarthritis (OA) is a major cause of pain and functional disability worldwide, leading to a growing interest in more durable and less invasive therapies. Micro-fragmented adipose tissue (MFAT) injections have emerged as a promising frontier in regenerative therapies using mesenchymal stem cells (MSCs). This study assessed the safety and effectiveness of MFAT injections for symptomatic knee OA while investigating the duration of treatment effects. Methods: This longitudinal study screened patients with symptomatic Kellgren-Lawrence (KL) grade II-IV knee OA who received single-dose MFAT injections. Outcomes were assessed using the Knee injury and Osteoarthritis Outcome Score (KOOS) subscales at baseline, 3, 6, and 12 months. A linear mixed effects model was performed to explore how age, BMI, sex, and OA severity influence outcomes. Results: Among 39 evaluable patients, mean baseline KOOS was 46.5 (SD 18.1). KOOS scores improved significantly across all subscales, peaking at six months and remaining higher than baseline at 12 months. Improvements exceeded clinically meaningful thresholds, including KL grades IV. Female patients reported significantly worse overall outcomes than male patients (p < 0.05). Minor self-limiting synovitis was reported in 18% of cases, and no severe adverse events were observed. Conclusions: MFAT infiltration may represent a safe, minimally invasive option to improve symptoms and delay surgery in patients with knee OA, including those with advanced disease. These findings highlight the potential role of MFAT as part of the treatment algorithm for knee OA, although strategies to sustain long-term benefits and confirmatory trials are needed. Full article
(This article belongs to the Special Issue Knee Osteoarthritis: Clinical Updates and Perspectives)
Show Figures

Figure 1

Back to TopTop