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

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (335)

Search Parameters:
Keywords = rat bone marrow

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
18 pages, 1944 KB  
Article
Introduction of Mature Mast Cells into Bone Marrow Alters Bone Metabolism in Growing Mice
by Carmen P. Wong, Jessica A. Keune, Kenneth A. Philbrick, Adam J. Branscum, Urszula T. Iwaniec and Russell T. Turner
Int. J. Mol. Sci. 2025, 26(24), 11952; https://doi.org/10.3390/ijms262411952 - 11 Dec 2025
Viewed by 164
Abstract
There is evidence that mast cells contribute to skeletal response to injury, but it is less clear whether these immune cells directly influence normal bone growth and turnover. Mature mast cells are common in the bone marrow of humans and rats, but have [...] Read more.
There is evidence that mast cells contribute to skeletal response to injury, but it is less clear whether these immune cells directly influence normal bone growth and turnover. Mature mast cells are common in the bone marrow of humans and rats, but have not been convincingly demonstrated to be present in the bone marrow of healthy mice, potentially limiting the mouse as a model for characterizing the full range of mast cell/bone cell interactions. An initial goal of this investigation was to comprehensively screen seven strains of mice for mature mast cells in bone marrow. Finding none, we then investigated three approaches to home these cells to the marrow of mice unable to generate mast cells: (1) administration of soluble kit ligand to membrane kit ligand-deficient KitSl/Sld mice, (2) adoptive transfer of wild-type hematopoietic stem cells to kit receptor-deficient KitW/Wv mice, and (3) adoptive transfer of wild-type mouse bone marrow-derived mast cells generated in vitro and delivered intravenously to KitW/W-v mice. Only the third approach was successful. Using this method, we then evaluated the impact of bone marrow-derived mast cells on bone mass, architecture, turnover, and gene expression. The adoptive transfer of mast cells resulted in alterations in cancellous bone microarchitecture and cell populations in the vertebra, and in differential expression of genes associated with bone metabolism in the tibia. Taken together, our results support the concept that bone marrow mast cells influence bone metabolism and suggest that homing mast cells to the bone marrow of mice is a useful model to understand the role of these cells in skeletal health and disease. Full article
(This article belongs to the Special Issue Mast Cells in Human Health and Diseases—3rd Edition)
Show Figures

Figure 1

16 pages, 5500 KB  
Article
Amniotic Membrane and Stem Cells Can Improve the Immunohistochemical Profile of Achilles Tendons in Injured Rats
by Rosangela Alquieri Fedato, Guilherme Vieira Cavalcante Da Silva, Lúcia De Noronha, Seigo Nagashima, Ana Paula Camargo Martins, Márcia Olandoski, Ricardo Aurino De Pinho, Aline Luri Takejima, Rossana Baggio Simeoni, Júlio Cesar Francisco and Luiz César Guarita-Souza
Biomedicines 2025, 13(12), 3018; https://doi.org/10.3390/biomedicines13123018 - 9 Dec 2025
Viewed by 268
Abstract
Disorders of the Achilles tendon are common and have a major socio-economic impact. Current treatments (drugs, physiotherapy, and surgery) do not provide lasting relief, leading to chronicity and recurrence. In this context, experimental studies on regenerative therapies, such as stem cells, and natural [...] Read more.
Disorders of the Achilles tendon are common and have a major socio-economic impact. Current treatments (drugs, physiotherapy, and surgery) do not provide lasting relief, leading to chronicity and recurrence. In this context, experimental studies on regenerative therapies, such as stem cells, and natural and synthetic membranes, have shown promising results in the treatment of tendon lesions. Background/Objectives: The present study analyzes the response of tissue to a combination of bone marrow mononuclear cells (BMMCs) and human decellularized amniotic membrane (AM) for the treatment of Achilles tendon lesions in rats. Methods: Forty male Wistar rats were randomized into four treatment groups: SC (stem cells), AM (amniotic membrane), SC + AM (stem cells + amniotic membrane), and C (control). All underwent Achilles tendon sectioning and tenorrhaphy. In the AM and SC + AM groups, the amniotic membrane was sutured over the lesion after the tendon was sutured; in the SC and SC + AM groups, 2 mL of autologous blood from the iliac crest containing BMMCs was applied around the lesion. Animals in Group C received only 2 mL of 0.9% saline around the lesion. After four weeks, the animals were euthanized, and the tendons were sent for histological analysis (Picrosirius Red) and immunohistochemistry (IL-6, IL-4, and IL-13). Results: Analysis of type I and type III collagen fibers showed no differences between groups. However, the SC + AM group showed the highest expressions of IL-4 and IL-13. Conclusions: IL-4 and IL-13 are cytokines known to be associated with tissue repair and organization. This suggests that the therapy associated with SC and AM is potentially beneficial in the treatment of injured Achilles tendons. However, further studies are necessary to clarify the benefits of this treatment for the function and biomechanical properties of the tendon and prove whether this association could represent a combined Advanced Therapy Medicinal Product (cATMP). Such a product would contain SC and a biological membrane, providing a mechanical structure for the injured tendon and active biological cells. Another possible medical approach could be immunobiological drugs targeting IL-4 and IL-13. Full article
(This article belongs to the Special Issue Updates on Tissue Repair and Regeneration Pathways)
Show Figures

Figure 1

20 pages, 3596 KB  
Article
Effect of Allogenic Mesenchymal Stem Cell Injection on Functional Repair Outcomes Following Skeletal Muscle Laceration Injury
by Raja Elina Ahmad, Abdul Halim Mokhtar, Mohamed Zubair Mohamed Al-Fayyadh, Hui Yin Nam, Atiqah Aziz, Azura Mansor and Tunku Kamarul
Biomedicines 2025, 13(11), 2810; https://doi.org/10.3390/biomedicines13112810 - 18 Nov 2025
Viewed by 544
Abstract
Background: Skeletal muscle laceration injuries remain a clinical challenge owing to limited and often delayed functional recovery. Surgical repair often fails to fully restore injured muscle, causing fibrosis and functional impairments. Mesenchymal stem cells (MSCs) represent a potential therapy due to their [...] Read more.
Background: Skeletal muscle laceration injuries remain a clinical challenge owing to limited and often delayed functional recovery. Surgical repair often fails to fully restore injured muscle, causing fibrosis and functional impairments. Mesenchymal stem cells (MSCs) represent a potential therapy due to their regenerative and immunomodulatory properties. However, their short-term regenerative effects in laceration injuries remain under-explored. Objective: We aim to evaluate the short-term effects of allogenic bone marrow-derived MSCs on skeletal muscle regeneration following laceration injury in rats. Methods: Sprague Dawley rats underwent laceration injury to the right gastrocnemius muscle and received local injection of either saline (n = 6) or allogeneic bone marrow-derived MSCs (2 × 106 cells; n = 6) two weeks after injury. Muscle functional recovery was evaluated by measuring tetanic contraction force of the injured relative to the contralateral uninjured leg and compared among MSC-treated, saline-treated, untreated injured (n = 6), and intact control groups (n = 6) on days 7 and 14 post-treatment. Histological assessment of the treated muscle groups using Hematoxylin and Eosin and Masson’s Trichrome staining was conducted on day 7 post-treatment. Results: On day 7 post-treatment, MSC-treated muscle showed higher normalised force (96.8 ± 15.0%) than saline-treated (76.7 ± 4.6%) (p = 0.0393), but not untreated, muscle (83.1 ± 14.7%) (p = 0.2259). By day 14, the MSC-treated group exhibited significantly greater recovery of muscle force (110.8 ± 6.46%) than both the saline-treated (78.4 ± 6.47%) (p < 0.0001) and untreated groups (88.1 ± 3.41%) (p = 0.0001). Force recovery in the MSC-treated muscle was comparable to that in intact muscle (102.6 ± 10.4%) at both time points (p = 0.230). Supplementary histological analysis showed mild inflammatory cell infiltration, well-formed myoblasts, and a lower fibrosis index in MSC-treated muscle (29.30 ± 0.29%) compared with saline-treated muscle (31.77 ± 0.43%) (p < 0.0001) on day 7 post-treatment. Conclusions: Allogeneic bone marrow-derived MSC therapy is associated with enhanced repair of lacerated skeletal muscle over a short recovery period; however, larger studies with broader assessments are needed to confirm its potential clinical applicability. Full article
(This article belongs to the Special Issue Stem Cell Therapy and Tissue Engineering)
Show Figures

Figure 1

23 pages, 13904 KB  
Article
Total Flavonoids of Rhizoma drynariae Enhance Bone Marrow Mesenchymal Stem Cell-Mediated Tendon–Bone Healing by Promoting Tissue Regeneration, Angiogenesis, and Modulation of Cytokine Expression
by Gaoyuan Yang, Yu Wang, Xianyan Xie, Ziyan Li, Shuqi Qin, Weitong Zhang, Zixi Chenyuan, Peizhong Cao, Huiguo Wang and Lin Zhu
Biology 2025, 14(11), 1593; https://doi.org/10.3390/biology14111593 - 14 Nov 2025
Viewed by 575
Abstract
(1) Objective: This study aimed to investigate the synergistic effect and underlying mechanisms of Total Flavonoids of Rhizoma drynariae (TFRD) in combination with Bone Marrow Mesenchymal Stem Cells (BMSCs) in the repair of tendon–bone injuries. (2) Methods: The effects of TFRD on the [...] Read more.
(1) Objective: This study aimed to investigate the synergistic effect and underlying mechanisms of Total Flavonoids of Rhizoma drynariae (TFRD) in combination with Bone Marrow Mesenchymal Stem Cells (BMSCs) in the repair of tendon–bone injuries. (2) Methods: The effects of TFRD on the proliferation and migration of BMSCs were assessed using CCK-8 and scratch assays, and its potential to promote osteogenic and chondrogenic differentiation was evaluated. Concurrently, the pro-angiogenic effect of TFRD on Human Umbilical Vein Endothelial Cells (HUVECs) was observed. In vivo, a rat model of Achilles tendon–bone injury was established and animals were divided into four groups: SHAM, Model, BMSCs, and BMSCs + TFRD. After an 8-week intervention, the level of functional recovery was evaluated through histological analysis, immunohistochemistry, serum biochemical analysis, and biomechanical testing. (3) Results: A concentration of 5.0 μg/mL TFRD significantly promoted the proliferation, migration, and differentiation of BMSCs and enhanced the tube formation capacity of HUVECs. In the BMSCs + TFRD group, histological analysis revealed well-organized collagen fibers, increased cartilage deposition, and an optimized tendon–bone interface (TBI) structure. Immunohistochemistry showed upregulated expression of COL I, COL II, and SOX-9, alongside downregulated VEGFA. Furthermore, serum IL-6 levels were decreased, while IL-10 and TGF-β levels were elevated. The biomechanical properties were also significantly improved in this group. (4) Conclusions: TFRD promotes tendon–bone healing and functional recovery by enhancing BMSC functions, promoting angiogenesis, and improving the local microenvironment. Full article
(This article belongs to the Special Issue Bone Mechanics: From Cells to Organs, to Function)
Show Figures

Figure 1

20 pages, 2854 KB  
Article
Sodium Phenylbutyrate Ameliorates Ovariectomy-Induced Bone Loss in Rats
by Bakiye Akbaş, Gülseren Dinç, Ahmet Akbaş, Nadir Adnan Hacım, Gülçin Ercan, Hatice Aygün and Oytun Erbaş
Medicina 2025, 61(11), 2016; https://doi.org/10.3390/medicina61112016 - 11 Nov 2025
Viewed by 436
Abstract
Background and Objectives: Estrogen deficiency after menopause accelerates bone loss through oxidative stress and inflammatory cytokines. Sodium phenylbutyrate (SP), a histone deacetylase inhibitor, exhibits antioxidative and anti-inflammatory effects, but its impact on postmenopausal osteoporosis remains unclear. Materials and Methods: Thirty female Wistar [...] Read more.
Background and Objectives: Estrogen deficiency after menopause accelerates bone loss through oxidative stress and inflammatory cytokines. Sodium phenylbutyrate (SP), a histone deacetylase inhibitor, exhibits antioxidative and anti-inflammatory effects, but its impact on postmenopausal osteoporosis remains unclear. Materials and Methods: Thirty female Wistar rats were divided into control, ovariectomy (OVX), and OVX+SPB groups (n = 10 each). After 12 weeks, bone mineral density (BMD), histomorphometry, bone marrow biomarkers (MDA, TNF-α, IL-6, RANKL), and plasma Cathepsin K were evaluated. Results: OVX induced trabecular deterioration with reduced number, area, and thickness (all p < 0.001), increased separation (p < 0.001), and decreased femoral and lumbar BMD (p < 0.001). SPB significantly improved these indices (TN, p < 0.05; TA, p < 0.01; TH, p < 0.05; femoral BMD, p < 0.05; lumbar BMD, p < 0.001; TS, p = 0.001). OVX elevated MDA, TNF-α, IL-6, RANKL, and Cathepsin K (all p < 0.001), which were significantly reduced by SPB (MDA, p < 0.001; TNF-α, p < 0.01; IL-6, p < 0.01; RANKL, p < 0.001; Cathepsin K, p < 0.001). Conclusions: SPB mitigates OVX-induced oxidative stress, inflammatory cytokine release, and osteoclast-mediated resorption, resulting in partial but significant improvements across biochemical, structural, and histomorphometric parameters in estrogen-deficient rats. Given its established clinical safety profile, SPB emerges as a cost-effective candidate for repurposing in postmenopausal osteoporosis, warranting further translational and clinical studies. Full article
(This article belongs to the Section Pharmacology)
Show Figures

Figure 1

17 pages, 10869 KB  
Article
The Effect of Titanium Dioxide Nanotubes and Graphene Compounds on the Proliferation and Osteogenic Differentiation of Rat BMSCs
by Chenyuan Zhu, Yuwei Deng, Jing Xu, Jin Wen, Qingfeng Huang and Weiqiang Yu
J. Funct. Biomater. 2025, 16(11), 413; https://doi.org/10.3390/jfb16110413 - 5 Nov 2025
Viewed by 1674
Abstract
Graphene-based nanomaterials, including graphene oxide (GO) and graphene quantum dots (GQDs), exhibit exceptional properties, which might facilitate the functional modification of TiO2 nanotubes (NTs) for enhanced rapid osseointegration. This study investigated the effects of GO/GQD-deposited TiO2-NTs on cell proliferation, osteogenic [...] Read more.
Graphene-based nanomaterials, including graphene oxide (GO) and graphene quantum dots (GQDs), exhibit exceptional properties, which might facilitate the functional modification of TiO2 nanotubes (NTs) for enhanced rapid osseointegration. This study investigated the effects of GO/GQD-deposited TiO2-NTs on cell proliferation, osteogenic differentiation of rat bone marrow-derived mesenchymal stem cells (BMSCs), and early osseointegration in male 6-week-old Sprague Dawley (SD) rats. TiO2-NTs (control group) were fabricated on titanium substrates via anodic oxidation. GO and GQDs were electrochemically deposited onto the TiO2-NTs using cyclic voltammetry with 0.5 mg/mL GO and 0.1 mg/mL GQD dispersions to form NT-GO and NT-GQDs. In vitro assays evaluated cell adhesion, proliferation, and osteogenic differentiation. Implants were randomly inserted into one femoral epiphysis of nine rats (n = 3), and osseointegration was evaluated using micro-computed tomography and sequential fluorescence labeling at 2, 4, and 6 weeks post-implantation. Statistical analysis was conducted using ANOVA. Cyclic voltammetry successfully synthesized NT-GO and NT-GQDs, with Raman spectra confirming D and G bands. Both NT-GO and NT-GQDs exhibited superior cell adhesion, proliferation, and enhanced osteogenic differentiation compared with TiO2-NTs. Notably, the NT-GQDs significantly promoted new bone formation in vivo. The integration of graphene nanomaterials onto TiO2-NTs improves biocompatibility and accelerates osteogenesis, suggesting a promising strategy for enhancing osseointegration in orthopedic and dental implants. Full article
(This article belongs to the Special Issue Advanced Materials and Devices for Medical Interventions)
Show Figures

Graphical abstract

17 pages, 3890 KB  
Article
Lacticaseibacillus rhamnosus AC1 Aggravates Bone Loss in a Male Rat Model of Deoxycorticosterone Acetate (DOCA)-Salt-Induced Osteoporosis
by Xiaoqing Kuang, Haicui Wu, Tim Fat Shum, Chunyi Wen and Jiachi Chiou
Nutrients 2025, 17(20), 3198; https://doi.org/10.3390/nu17203198 - 11 Oct 2025
Viewed by 622
Abstract
Background/Objectives: Osteoporosis is a prevalent and debilitating skeletal disease characterized by a progressive loss of bone mass and deterioration of bone microarchitecture. Probiotics have emerged as a potential therapeutic tool for treating osteoporosis through modulation of the gut microbiota. In this study, [...] Read more.
Background/Objectives: Osteoporosis is a prevalent and debilitating skeletal disease characterized by a progressive loss of bone mass and deterioration of bone microarchitecture. Probiotics have emerged as a potential therapeutic tool for treating osteoporosis through modulation of the gut microbiota. In this study, we aimed to examine the effects of live Lacticaseibacillus rhamnosus AC1 (LR-AC1), isolated from a fecal sample from a newborn in Hong Kong, on deoxycorticosterone acetate (DOCA)-induced bone loss in a rat model. Methods: Bone mass and microarchitecture were assessed using micro-computed tomography (micro-CT). Immunostaining for CD31+ and osterix, markers of endothelial cells and osteoblast precursors, respectively, was performed. Gut microbiota composition was analyzed via 16S rRNA sequencing. The effects of an LR-AC1 cell-free conditioned supernatant (CCS) on osteoclastogenesis, angiogenesis, and migration of bone marrow mesenchymal stem cells (BMSCs) were evaluated in vitro using RT-qPCR and wound healing assays. Results: LR-AC1 administration did not induce adverse effects in healthy rats; however, it exacerbated bone loss in rats with DOCA-salt-induced osteoporosis. Correspondingly, the number of CD31-positive endothelial cells and osterix-positive osteoprogenitors decreased with bone loss. In vitro, LR-AC1 CCS promoted osteoclastogenesis and angiogenesis, while in the presence of DOCA, LR-AC1 CCS inhibited BMSC migration. Gut microbiota analysis revealed that the relative abundances of the genera g_RF39 and g_Clostridia_UCG-014 correlated with the severity of bone loss. Conclusions: While several studies suggest that probiotics can prevent and treat osteoporosis, our findings indicate that in a male rat model of DOCA-salt-induced osteoporosis, live LR-AC1 aggravated bone loss. This effect is associated with alterations in gut microbiota and disruption of the coupling process in bone remodeling. Full article
Show Figures

Figure 1

19 pages, 6916 KB  
Article
Short-Term Cryopreservation Preserved the Function of MSCs from Bone Marrow Aspirate Concentrate
by Jacob Singer, Haruki Nishimura, Zuokui Xiao, Xueqin Gao, Noah Knezic, Laura Chubb, Jonathan E. Layne, Ping Guo, Aiping Lu and Johnny Huard
Cells 2025, 14(19), 1569; https://doi.org/10.3390/cells14191569 - 9 Oct 2025
Viewed by 1039
Abstract
Bone marrow aspirate concentrate (BMAC) is increasingly recognized as a valuable orthobiologic, offering promising outcomes in reducing inflammation, alleviating pain for patients with osteoarthritis (OA) and various musculoskeletal conditions. However, BMAC contains a very low percentage of mesenchymal stem cells (MSCs), and multiple [...] Read more.
Bone marrow aspirate concentrate (BMAC) is increasingly recognized as a valuable orthobiologic, offering promising outcomes in reducing inflammation, alleviating pain for patients with osteoarthritis (OA) and various musculoskeletal conditions. However, BMAC contains a very low percentage of mesenchymal stem cells (MSCs), and multiple injections are often required with multiple harvests, which can lead to scarring at the extraction site and patient discomfort. This study aimed to determine whether freezing BMAC affects the function of MSCs in vitro and their capacity to repair articular cartilage in vivo using an OA rat model. BMAC was obtained from patients undergoing BMAC treatment. The in vitro results showed that the proliferation and multilineage differentiation of MSCs remained similar after being frozen for 4 weeks at −80 °C. In vivo, both fresh and frozen BMAC demonstrated significantly improved ICRS histology score of tibial plateau cartilage compared to the PBS control. No significant difference was found between fresh and frozen BMAC treatment groups. Our results suggest that the freezing process does not negatively affect the function of MSCs from BMAC for cartilage repair. These findings support the potential future applications of a single harvest with BMAC storage for multiple injections, thereby enhancing the tissue repair capabilities of BMAC. Full article
(This article belongs to the Special Issue Stem Cells and Beyond: Innovations in Tissue Repair and Regeneration)
Show Figures

Graphical abstract

17 pages, 13481 KB  
Article
Luopan Mountain Pig Bone Marrow Mesenchymal Stem Cells Promote Liver Regeneration in D-Galactosamine-Induced Acute Liver Failure Rats by Regulating the PTEN-PI3K/Akt/mTOR Pathway
by Minjuan Li, Zhongfa Wang, Xingxing Yan, Yanchen Liu, Yunan He, Bianying Zhang and Weijun Guan
Biology 2025, 14(10), 1363; https://doi.org/10.3390/biology14101363 - 5 Oct 2025
Viewed by 613
Abstract
Treatment for acute liver failure (ALF) is constrained by shortages of liver transplant donors and immune rejection. Porcine bone marrow mesenchymal stem cells (pBMSCs) demonstrate clinical potential in xenotransplantation due to their abundant availability, low immunogenicity, and strong proliferative activity. This study is [...] Read more.
Treatment for acute liver failure (ALF) is constrained by shortages of liver transplant donors and immune rejection. Porcine bone marrow mesenchymal stem cells (pBMSCs) demonstrate clinical potential in xenotransplantation due to their abundant availability, low immunogenicity, and strong proliferative activity. This study is the first to investigate the reparative effects and mechanisms of pBMSCs derived from Luopan Mountain pigs in a D-galactosamine (D-GalN)-induced ALF rat model. The results demonstrated that tail-vein transplantation of pBMSCs significantly improved survival rates in ALF rats; reduced serum ALT, AST, and TBIL levels; enhanced hepatic glycogen metabolism; and mitigated histopathological liver damage. Additionally, pBMSC transplantation upregulated serum HGF, IGF-1, and VEGF levels while inhibiting hepatocyte apoptosis. Mechanistic studies indicate that pBMSCs promote liver function recovery and regeneration by activating the PI3K/Akt/mTOR signaling pathway and suppressing its key negative regulator, PTEN, by regulating the expression of key genes involved in inflammation, fibrosis, proliferation, and apoptosis. This study provides crucial experimental evidence for the use of pBMSCs in treating acute liver failure (ALF) and lays the groundwork for its clinical translation in the field of xenotransplantation. Full article
(This article belongs to the Section Cell Biology)
Show Figures

Figure 1

13 pages, 759 KB  
Article
Bone Marrow Mononuclear Cells Administration Restore Lysophosphatidic Acid (LPA) Levels and Cellular Signaling Axis in Rats Submitted to Renal Ischemia–Reperfusion
by Paula Mattos-Silva, Sabrina Ribeiro Gonsalez, Lucienne S. Lara and Marcelo Einicker-Lamas
Int. J. Mol. Sci. 2025, 26(18), 9186; https://doi.org/10.3390/ijms26189186 - 20 Sep 2025
Viewed by 604
Abstract
Bone marrow-derived mononuclear cells (BMMCs) have shown beneficial effects on tissue repair, largely attributed to the paracrine action of bioactive mediators such as lysophosphatidic acid (LPA). This study aimed to evaluate the effects of BMMC treatment in a rat model of renal ischemia/reperfusion [...] Read more.
Bone marrow-derived mononuclear cells (BMMCs) have shown beneficial effects on tissue repair, largely attributed to the paracrine action of bioactive mediators such as lysophosphatidic acid (LPA). This study aimed to evaluate the effects of BMMC treatment in a rat model of renal ischemia/reperfusion (I/R) injury, focusing on LPA-related molecular pathways. Male Wistar rats were divided into three groups: control; I/R, subjected to bilateral renal artery clamping for 30 min followed by 24 h of reperfusion; and I/R + BMMC, which received 1 × 106 BMMCs per kidney directly into the renal capsule post-ischemia. During reperfusion, the rats were placed in metabolic cages for urine collection, renal function and protein expression. BMMC treatment did not reverse the I/R-induced increase in urine volume or decrease in glomerular filtration rate, serum potassium, or filtered sodium load. However, it prevented proteinuria, increased blood urea nitrogen, and enhanced urinary potassium excretion. Mechanistically, BMMC treatment prevented I/R-induced upregulation of LPAR1, downregulated LPAR2 and LPAR3, restored plasma LPA levels, and reduced renal autotaxin content. These results suggest that BMMCs modulate harmful LPA-related signaling and may contribute to renal protection through paracrine mechanisms in the setting of acute I/R injury. Full article
(This article belongs to the Special Issue Bioactive Lipids and Their Derivatives in Biomedical Applications)
Show Figures

Figure 1

13 pages, 2535 KB  
Article
Effects of Platelet-Rich Fibrin Treated with No-Ozone Cold Plasma on the Alkaline Phosphatase in Rat Bone Marrow Cells: An In Vitro Study
by Byul Bo Ra Choi and Gyoo Cheon Kim
Appl. Sci. 2025, 15(17), 9229; https://doi.org/10.3390/app15179229 - 22 Aug 2025
Viewed by 798
Abstract
Background/Objectives: Herein, we investigated the effect of platelet-rich fibrin (PRF) treatment combined with no-ozone cold plasma (NCP) on growth factor levels, rat bone-marrow stem cell (rBMSC) proliferation, and alkaline phosphatase (ALP) activity in the early stage of differentiation into osteoblasts. Methods: [...] Read more.
Background/Objectives: Herein, we investigated the effect of platelet-rich fibrin (PRF) treatment combined with no-ozone cold plasma (NCP) on growth factor levels, rat bone-marrow stem cell (rBMSC) proliferation, and alkaline phosphatase (ALP) activity in the early stage of differentiation into osteoblasts. Methods: The PRF used in the experiment was prepared by collecting blood from the jugular vein of rats, followed by centrifugation. The obtained PRF was treated with NCP, and the cell culture media were conditioned with the PRF extracts alone or with NCP-treated PRF extracts. Three different experimental groups were defined: no treatment (NT); cell culture media extracted from PRF (PRF); and cell culture media extracted from PRF treated with NCP (PRF + NCP). Enzyme-linked immunosorbent assays were performed to determine the levels of transforming growth factor (TGF)-β and platelet-derived growth factor (PDGF) AB. Water-soluble Tetrazolium-1 assay was performed to measure cell proliferation in rBMSCs. To analyze cell differentiation into osteoblasts, ALP staining and real-time PCR were performed. Results: Growth factor levels increased in response to treatment (TGF-β: p < 0.001, PDGF AB: p < 0.05), and the cell proliferation rate increased with treatment (145.29% and 150.05% for PRF and the PRF + NCP groups, respectively, relative to the NT group, p < 0.001). Evaluation of the ALP staining intensity and mRNA expression levels showed that the ALP activity was highest in the PRF + NCP group (p < 0.001). Conclusions: Our results confirmed that NCP treatment enhanced the release of several different growth factors contained in PRF to the culture media and that treatment with PRF and NCP increased the proliferation of rBMSCs and their differentiation into osteoblasts. Full article
(This article belongs to the Special Issue Oral Diseases and Clinical Dentistry)
Show Figures

Figure 1

24 pages, 7673 KB  
Article
Development, Molecular Docking, and Anti-Anemia Potential of Polyherbal Formulation
by Deepak Bharati, Sakshi Nirhali, Abhijeet Puri, Popat Mohite and Sudarshan Singh
Biology 2025, 14(8), 1052; https://doi.org/10.3390/biology14081052 - 14 Aug 2025
Viewed by 1535
Abstract
Anemia remains a widespread public health concern, and the search for interventions demonstrating potent anti-anemic activity is critical for reducing its impact among high-risk populations. Conventional iron therapies are associated with several complications and potential adverse effects. This study explored a polyherbal approach [...] Read more.
Anemia remains a widespread public health concern, and the search for interventions demonstrating potent anti-anemic activity is critical for reducing its impact among high-risk populations. Conventional iron therapies are associated with several complications and potential adverse effects. This study explored a polyherbal approach to develop a safer and more effective alternative treatment for anemia. A molecular docking study was initially performed to screen and evaluate alizarin, catechin, kaempferol, recesmol, rubiadin, and rutin, which are known for their antioxidant and hematinic potential. Using AutoDock Vina, these compounds were docked against the target protein (PDB ID: 6MOE) with EPE and ferrous ions as controls. Rutin demonstrated the highest binding affinity of −6.4 kcal/moL, whereas alizarin and rubiadin both followed closely with −6.3 kcal/moL, while kaempferol and ellagic acid exhibited a binding affinity of −6.2 kcal/moL. In comparison, the reference compounds tested ferrous ions, and native ligand EPE (−5.0 kcal/moL) and iron (−4.8 kcal/moL), showed mild affinities. Moreover, the tested compounds demonstrated stable binding, suggesting their potential relevance in modulating anemia-related pathways. Based on the docking results and traditional therapeutic values, a polyherbal formulation (PHF) was developed using methanolic extracts of Trigonella foenum-graecum, Emblica officinalis, Pterocarpus marsupium, Withania somnifera, Asparagus racemosus, Zingiber officinale, Rubia cordifolia, Boerhavia diffusa, and Adhatoda vasica. Phytochemical screening via HPTLC analysis was used to quantify the presence of gallic and ellagic acids. In addition, PHF showed significant antioxidant potential (DPPH IC50: 14.29 µg/mL; FRAP IC50: 58.57 µg/mL) and iron content (98.47 ppm) values. Furthermore, in vivo evaluation using a phenylhydrazine-induced hemolytic anemia model in Sprague Dawley rats revealed that the PHF achieved complete restoration of RBCs (6.15 ± 0.04), hemoglobin (14.82 ± 0.03 g/dL), and hematocrit (43.08 ± 0.28%) in anemic rats and improved histopathological features in the liver, spleen, and bone marrow. These results demonstrate that combined molecular and pharmacological evidence support the efficacy of PHF as a promising candidate for the management of anemia by enhancing erythropoiesis, improving iron metabolism, and reducing oxidative stress. Full article
Show Figures

Graphical abstract

25 pages, 22282 KB  
Article
Osteogenesis Activity and Porosity Effect of Biodegradable Mg-Ga Alloys Barrier Membrane for Guided Bone Regeneration: An in Vitro and in Vivo Study in Rabbits
by Qiyue Luo, Kang Gao, Yan Li, Ziyue Zhang, Su Chen and Jian Zhou
Biomedicines 2025, 13(8), 1940; https://doi.org/10.3390/biomedicines13081940 - 8 Aug 2025
Cited by 1 | Viewed by 808
Abstract
Background/Objectives: Guided bone regeneration (GBR) requires barrier membrane materials that balance biodegradation with mechanical stability. Magnesium (Mg)-based metals have good prospects for use as biodegradable barrier materials due to their elastic modulus, good biocompatibility, and osteogenic properties. In this study, gallium (Ga) [...] Read more.
Background/Objectives: Guided bone regeneration (GBR) requires barrier membrane materials that balance biodegradation with mechanical stability. Magnesium (Mg)-based metals have good prospects for use as biodegradable barrier materials due to their elastic modulus, good biocompatibility, and osteogenic properties. In this study, gallium (Ga) was introduced into Mg to enhance the mechanical strength and optimize the degradation behavior of the alloy, addressing the limitations of conventional magnesium alloys in corrosion control and strength retention. Methods: Mg-xGa alloys (x = 1.0–3.0%, wt.%) were evaluated for biocompatibility, degradation, and osteogenic potential. Corrosion rates were calculated via weight loss, Mg2+ release, and pH changes. Osteogenic effects were assessed using rat bone marrow mesenchymal stem cells (rBMSCs) for alkaline phosphatase (ALP) activity, extracellular matrix (ECM) mineralization, and osteogenic-related gene expression. Optimal alloy was fabricated into barrier membranes with different pore sizes (0.85–1.70 mm) for the rabbit mandibular defect to evaluate the porosity effect on new bone formation. Results: Cytocompatibility tests established a biosafety threshold for Ga content below 3 wt.%. Mg-1Ga demonstrated uniform corrosion with a rate of 1.02 mm/year over 28 days. In vitro, Mg-1Ga enhanced ALP activity, ECM mineralization, and osteogenic gene expression. The 1.70 mm pore size group exhibited superior new bone formation and bone mineral density at 4 and 8 weeks. Conclusions: These results highlight Mg-1Ga’s biocompatibility, controlled degradation, and osteogenic properties. Its optimized pore design bridges the gap between collagen membranes’ poor strength and titanium meshes’ non-degradability, offering a promising solution for GBR applications. Full article
(This article belongs to the Special Issue Biomedicine in Dental and Oral Rehabilitation)
Show Figures

Figure 1

16 pages, 3091 KB  
Article
Fabrication and Evaluation of Screen-Printed Electrodes on Chitosan Films for Cardiac Patch Applications with In Vitro and In Vivo Evaluation
by Yu-Hsin Lin, Yong-Ji Chen, Jen-Tsai Liu, Ching-Shu Yen, Yi-Zhen Lin, Xiu-Wei Zhou, Shu-Ying Chen, Jhe-Lun Hu, Chi-Hsiang Wu, Ching-Jung Chen, Pei-Leun Kang and Shwu-Jen Chang
Polymers 2025, 17(15), 2088; https://doi.org/10.3390/polym17152088 - 30 Jul 2025
Viewed by 1061
Abstract
Myocardial infarction (MI) remains one of the most common cardiovascular diseases and a leading cause of morbidity and mortality worldwide. In recent years, natural polymeric patches have attracted increasing attention as a promising therapeutic platform for myocardial tissue repair. This study explored the [...] Read more.
Myocardial infarction (MI) remains one of the most common cardiovascular diseases and a leading cause of morbidity and mortality worldwide. In recent years, natural polymeric patches have attracted increasing attention as a promising therapeutic platform for myocardial tissue repair. This study explored the fabrication and evaluation of screen-printed electrodes (SPEs) on chitosan film as a novel platform for cardiac patch applications. Chitosan is a biodegradable and biocompatible natural polymer that provides an ideal substrate for SPEs, providing mechanical stability and promoting cell adhesion. Silver ink was employed to enhance electrochemical performance, and the electrodes exhibited strong adhesion and structural integrity under wet conditions. Mechanical testing and swelling ratio analysis were conducted to assess the patch’s physical robustness and aqueous stability. Silver ink was employed to enhance electrochemical performance, which was evaluated using cyclic voltammetry. In vitro, electrical stimulation through the chitosan–SPE patch significantly increased the expression of cardiac-specific genes (GATA-4, β-MHC, troponin I) in bone marrow mesenchymal stem cells (BMSCs), indicating early cardiogenic differentiation potential. In vivo, the implantation of the chitosan–SPE patch in a rat MI model demonstrated good tissue integration, preserved myocardial structure, and enhanced ventricular wall thickness, indicating that the patch has the potential to serve as a functional cardiac scaffold. These findings support the feasibility of screen-printed electrodes fabricated on chitosan film substrates as a cost-effective and scalable platform for cardiac repair, offering a foundation for future applications in cardiac tissue engineering. Full article
(This article belongs to the Section Polymer Applications)
Show Figures

Graphical abstract

20 pages, 4241 KB  
Article
Strontium-Doped Ti3C2Tx MXene Coatings on Titanium Surfaces: Synergistic Osteogenesis Enhancement and Antibacterial Activity Evaluation
by Yancheng Lai and Anchun Mo
Coatings 2025, 15(7), 847; https://doi.org/10.3390/coatings15070847 - 19 Jul 2025
Cited by 1 | Viewed by 1192
Abstract
To improve implant osseointegration while preventing infection, we developed a strontium (Sr)-doped Ti3C2Tx MXene coating on titanium, aiming to synergistically enhance bone integration and antibacterial performance. MXene is a family of two-dimensional transition-metal carbides/nitrides whose abundant surface terminations [...] Read more.
To improve implant osseointegration while preventing infection, we developed a strontium (Sr)-doped Ti3C2Tx MXene coating on titanium, aiming to synergistically enhance bone integration and antibacterial performance. MXene is a family of two-dimensional transition-metal carbides/nitrides whose abundant surface terminations endow high hydrophilicity and bioactivity. The coating was fabricated via anodic electrophoretic deposition (40 V, 2 min) of Ti3C2Tx nanosheets, followed by SrCl2 immersion to incorporate Sr2+. The coating morphology, phase composition, chemistry, hydrophilicity, mechanical stability, and Sr2+ release were characterized. In vitro bioactivity was assessed with rat bone marrow mesenchymal stem cells (BMSCs)—with respect to viability, proliferation, migration, alkaline phosphatase (ALP) staining, and Alizarin Red S mineralization—while the antibacterial efficacy was evaluated against Staphylococcus aureus (S. aureus) via live/dead staining, colony-forming-unit enumeration, and AlamarBlue assays. The Sr-doped MXene coating formed a uniform lamellar structure, lowered the water-contact angle to ~69°, and sustained Sr2+ release (0.36–1.37 ppm). Compared to undoped MXene, MXene/Sr enhanced BMSC proliferation on day 5, migration by 51%, ALP activity and mineralization by 47%, and reduced S. aureus viability by 49% within 24 h. Greater BMSCs activity accelerates early bone integration, whereas rapid bacterial suppression mitigates peri-implant infection—two critical requirements for implant success. Sr-doped Ti3C2Tx MXene thus offers a simple, dual-function surface-engineering strategy for dental and orthopedic implants. Full article
(This article belongs to the Section Surface Coatings for Biomedicine and Bioengineering)
Show Figures

Figure 1

Back to TopTop