Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26.805
Filter
1.
Biomaterials ; 313: 122775, 2025 Feb.
Article in English | MEDLINE | ID: mdl-39241549

ABSTRACT

Acute Myocardial Infarction (AMI) has seen rising cases, particularly in younger people, leading to public health concerns. Standard treatments, like coronary artery recanalization, often don't fully repair the heart's microvasculature, risking heart failure. Advances show that Mesenchymal Stromal Cells (MSCs) transplantation improves cardiac function after AMI, but the harsh microenvironment post-AMI impacts cell survival and therapeutic results. MSCs aid heart repair via their membrane proteins and paracrine extracellular vesicles that carry microRNA-125b, which regulates multiple targets, preventing cardiomyocyte death, limiting fibroblast growth, and combating myocardial remodeling after AMI. This study introduces ultrasound-responsive phase-change bionic nanoparticles, leveraging MSCs' natural properties. These particles contain MSC membrane and microRNA-125b, with added macrophage membrane for stability. Using Ultrasound Targeted Microbubble Destruction (UTMD), this method targets the delivery of MSC membrane proteins and microRNA-125b to AMI's inflamed areas. This aims to enhance cardiac function recovery and provide precise, targeted AMI therapy.


Subject(s)
Mesenchymal Stem Cells , MicroRNAs , Myocardial Infarction , Nanoparticles , Myocardial Infarction/therapy , Animals , Nanoparticles/chemistry , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/cytology , MicroRNAs/metabolism , MicroRNAs/genetics , Male , Recovery of Function , Mesenchymal Stem Cell Transplantation/methods , Humans , Biomimetic Materials/chemistry , Biomimetic Materials/pharmacology , Mice , Microbubbles , Ultrasonic Waves
2.
Biomaterials ; 313: 122757, 2025 Feb.
Article in English | MEDLINE | ID: mdl-39178558

ABSTRACT

Recent progress in stem cell therapy has demonstrated the therapeutic potential of intravenous stem cell infusions for treating the life-threatening lung disease of pulmonary fibrosis (PF). However, it is confronted with limitations, such as a lack of control over cellular function and rapid clearance by the host after implantation. In this study, we developed an innovative PF therapy through tracheal administration of microfluidic-templated stem cell-laden microcapsules, which effectively reversed the progression of inflammation and fibrotic injury. Our findings highlight that hydrogel microencapsulation can enhance the persistence of donor mesenchymal stem cells (MSCs) in the host while driving MSCs to substantially augment their therapeutic functions, including immunoregulation and matrix metalloproteinase (MMP)-mediated extracellular matrix (ECM) remodeling. We revealed that microencapsulation activates the MAPK signaling pathway in MSCs to increase MMP expression, thereby degrading overexpressed collagen accumulated in fibrotic lungs. Our research demonstrates the potential of hydrogel microcapsules to enhance the therapeutic efficacy of MSCs through cell-material interactions, presenting a promising yet straightforward strategy for designing advanced stem cell therapies for fibrotic diseases.


Subject(s)
Capsules , Extracellular Matrix , Immunomodulation , Mesenchymal Stem Cells , Pulmonary Fibrosis , Animals , Extracellular Matrix/metabolism , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Pulmonary Fibrosis/therapy , Pulmonary Fibrosis/pathology , Mesenchymal Stem Cell Transplantation/methods , Mice, Inbred C57BL , Hydrogels/chemistry , Mice , Male
3.
Biomaterials ; 313: 122762, 2025 Feb.
Article in English | MEDLINE | ID: mdl-39178559

ABSTRACT

Osteomyelitis is an osseous infectious disease that primarily affects children and the elderly with high morbidity and recurrence. The conventional treatments of osteomyelitis contain long-term and high-dose systemic antibiotics with debridements, which are not effective and lead to antibiotic resistance with serious side/adverse effects in many cases. Hence, developing novel antibiotic-free interventions against osteomyelitis (especially antibiotic-resistant bacterial infection) is urgent and anticipated. Here, a bone mesenchymal stem cell membrane-constructed nanocell (CFE@CM) was fabricated against osteomyelitis with the characteristics of acid-responsiveness, hydrogen peroxide self-supplying, enhanced chemodynamic therapeutic efficacy, bone marrow targeting and cuproptosis induction. Notably, mRNA sequencing was applied to unveil the underlying biological mechanisms and found that the biological processes related to copper ion binding, oxidative phosphorylation, peptide biosynthesis and metabolism, etc., were disturbed by CFE@CM in bacteria. This work provided an innovative antibiotic-free strategy against osteomyelitis through copper-enhanced Fenton reaction and distinct cuproptosis, promising to complement the current insufficient therapeutic regimen in clinic.


Subject(s)
Copper , Osteomyelitis , Osteomyelitis/drug therapy , Animals , Copper/chemistry , Copper/pharmacology , Hydrogen-Ion Concentration , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/cytology , Mice , Hydrogen Peroxide/metabolism , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Anti-Bacterial Agents/chemistry , Humans , Staphylococcus aureus/drug effects
4.
Biomaterials ; 313: 122748, 2025 Feb.
Article in English | MEDLINE | ID: mdl-39180918

ABSTRACT

Extracellular vesicles (EVs) are future promising therapeutics, but their instability in vivo after administration remains an important barrier to their further development. Many groups evaluated EV surface modification strategies to add a targeting group with the aim of controlling EV biodistribution. Conversely, fewer groups focused on their stabilization to obtain "stealth" allogenic EVs. Modulating their stabilization and biodistribution is an essential prerequisite for their development as nano-therapeutics. Here, we explored polyoxazolines with lipid anchors association to the EV membrane (POxylation as an alternative to PEGylation) to stabilize EVs in plasma and control their biodistribution, while preserving their native properties. We found that this modification maintained and seemed to potentiate the immunomodulatory properties of EVs derived from mesenchymal stem/stromal cells (MSC). Using a radiolabeling protocol to track EVs at a therapeutically relevant concentration in vivo, we demonstrated that POxylation is a promising option to stabilize EVs in plasma because it increased EV half-life by 6 fold at 6 h post-injection. Moreover, EV accumulation in tumors was higher after POxylation than after PEGylation.


Subject(s)
Extracellular Vesicles , Mesenchymal Stem Cells , Extracellular Vesicles/metabolism , Extracellular Vesicles/chemistry , Animals , Humans , Tissue Distribution , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/cytology , Oxazoles/chemistry , Mice , Surface Properties , Cell Line, Tumor , Mice, Inbred C57BL , Female
5.
Biomaterials ; 312: 122713, 2025 Jan.
Article in English | MEDLINE | ID: mdl-39084096

ABSTRACT

Traditional bioreactor systems involve the use of three-dimensional (3D) scaffolds or stem cell aggregates, limiting the accessibility to the production of cell-secreted biomolecules. Herein, we present the use a pulse electromagnetic fields (pEMFs)-assisted wave-motion bioreactor system for the dynamic and scalable culture of human bone marrow-derived mesenchymal stem cells (hBMSCs) with enhanced the secretion of various soluble factors with massive therapeutic potential. The present study investigated the influence of dynamic pEMF (D-pEMF) on the kinetic of hBMSCs. A 30-min exposure of pEMF (10V-1Hz, 5.82 G) with 35 oscillations per minute (OPM) rocking speed can induce the proliferation (1 × 105 â†’ 4.5 × 105) of hBMSCs than static culture. Furthermore, the culture of hBMSCs in osteo-induction media revealed a greater enhancement of osteogenic transcription factors under the D-pEMF condition, suggesting that D-pEMF addition significantly boosted hBMSCs osteogenesis. Additionally, the RNA sequencing data revealed a significant shift in various osteogenic and signaling genes in the D-pEMF group, further suggesting their osteogenic capabilities. In this research, we demonstrated that the combined effect of wave and pEMF stimulation on hBMSCs allows rapid proliferation and induces osteogenic properties in the cells. Moreover, our study revealed that D-pEMF stimuli also induce ROS-scavenging properties in the cultured cells. This study also revealed a bioactive and cost-effective approach that enables the use of cells without using any expensive materials and avoids the possible risks associated with them post-implantation.


Subject(s)
Bioreactors , Electromagnetic Fields , Mesenchymal Stem Cells , Osteogenesis , Humans , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Gene Expression Profiling , Cell Proliferation , Cell Differentiation , Cells, Cultured , Transcriptome
6.
Biomaterials ; 312: 122720, 2025 Jan.
Article in English | MEDLINE | ID: mdl-39084098

ABSTRACT

Mesenchymal stem cells (MSCs) are expected to be useful therapeutics in osteoarthritis (OA), the most common joint disorder characterized by cartilage degradation. However, evidence is limited with regard to cartilage repair in clinical trials because of the uncontrolled differentiation and weak cartilage-targeting ability of MSCs after injection. To overcome these drawbacks, here we synthesized CuO@MSN nanoparticles (NPs) to deliver Sox9 plasmid DNA (favoring chondrogenesis) and recombinant protein Bmp7 (inhibiting hypertrophy). After taking up CuO@MSN/Sox9/Bmp7 (CSB NPs), the expressions of chondrogenic markers were enhanced while hypertrophic markers were decreased in response to these CSB-engineered MSCs. Moreover, a cartilage-targeted peptide (designated as peptide W) was conjugated onto the surface of MSCs via a click chemistry reaction, thereby prolonging the residence time of MSCs in both the knee joint cavity of mice and human-derived cartilage. In a surgery-induced OA mouse model, the NP and peptide dual-modified W-CSB-MSCs showed an enhancing therapeutic effect on cartilage repair in knee joints compared with other engineered MSCs after intra-articular injection. Most importantly, W-CSB-MSCs accelerated cartilage regeneration in damaged cartilage explants derived from OA patients. Thus, this new peptide and NPs dual engineering strategy shows potential for clinical applications to boost cartilage repair in OA using MSC therapy.


Subject(s)
Cell Differentiation , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells , Nanoparticles , Osteoarthritis , Peptides , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Animals , Osteoarthritis/therapy , Osteoarthritis/pathology , Nanoparticles/chemistry , Humans , Cell Differentiation/drug effects , Peptides/chemistry , Mesenchymal Stem Cell Transplantation/methods , Chondrogenesis/drug effects , Mice , SOX9 Transcription Factor/metabolism , SOX9 Transcription Factor/genetics , Cartilage, Articular/pathology , Cartilage, Articular/drug effects , Bone Morphogenetic Protein 7/chemistry , Bone Morphogenetic Protein 7/pharmacology , Tissue Engineering/methods , Regeneration/drug effects
7.
Biomaterials ; 312: 122715, 2025 Jan.
Article in English | MEDLINE | ID: mdl-39094522

ABSTRACT

Extracellular matrix (ECM) stiffness is a major driver of stem cell fate. However, the involvement of the three-dimensional (3D) genomic reorganization in response to ECM stiffness remains unclear. Here, we generated comprehensive 3D chromatin landscapes of mesenchymal stem cells (MSCs) exposed to various ECM stiffness. We found that there were more long-range chromatin interactions, but less compartment A in MSCs cultured on stiff ECM than those cultured on soft ECM. However, the switch from compartment B in MSCs cultured on soft ECM to compartment A in MSCs cultured on stiff ECM included genes encoding proteins primarily enriched in cytoskeleton organization. At the topologically associating domains (TADs) level, stiff ECM tends to have merged TADs on soft ECM. These merged TADs on stiff ECM include upregulated genes encoding proteins enriched in osteogenesis, such as SP1, ETS1, and DCHS1, which were validated by quantitative real-time polymerase chain reaction and found to be consistent with the increase of alkaline phosphatase staining. Knockdown of SP1 or ETS1 led to the downregulation of osteogenic marker genes, including COL1A1, RUNX2, ALP, and OCN in MSCs cultured on stiff ECM. Our study provides an important insight into the stiff ECM-mediated promotion of MSC differentiation towards osteogenesis, emphasizing the influence of mechanical cues on the reorganization of 3D genome architecture and stem cell fate.


Subject(s)
Cell Differentiation , Extracellular Matrix , Mesenchymal Stem Cells , Osteogenesis , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Osteogenesis/genetics , Extracellular Matrix/metabolism , Cell Differentiation/genetics , Humans , Cells, Cultured , Animals
8.
Biomaterials ; 312: 122724, 2025 Jan.
Article in English | MEDLINE | ID: mdl-39106818

ABSTRACT

The residual bone tumor and defects which is caused by surgical therapy of bone tumor is a major and important problem in clinicals. And the sequential treatment for irradiating residual tumor and repairing bone defects has wildly prospects. In this study, we developed a general modification strategy by gallic acid (GA)-assisted coordination chemistry to prepare black calcium-based materials, which combines the sequential photothermal therapy of bone tumor and bone defects. The GA modification endows the materials remarkable photothermal properties. Under the near-infrared (NIR) irradiation with different power densities, the black GA-modified bone matrix (GBM) did not merely display an excellent performance in eliminating bone tumor with high temperature, but showed a facile effect of the mild-heat stimulation to accelerate bone regeneration. GBM can efficiently regulate the microenvironments of bone regeneration in a spatial-temporal manner, including inflammation/immune response, vascularization and osteogenic differentiation. Meanwhile, the integrin/PI3K/Akt signaling pathway of bone marrow mesenchymal stem cells (BMSCs) was revealed to be involved in the effect of osteogenesis induced by the mild-heat stimulation. The outcome of this study not only provides a serial of new multifunctional biomaterials, but also demonstrates a general strategy for designing novel blacked calcium-based biomaterials with great potential for clinical use.


Subject(s)
Bone Neoplasms , Bone Regeneration , Calcium , Gallic Acid , Mesenchymal Stem Cells , Gallic Acid/chemistry , Bone Regeneration/drug effects , Animals , Calcium/metabolism , Bone Neoplasms/therapy , Bone Neoplasms/drug therapy , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/cytology , Photothermal Therapy/methods , Osteogenesis/drug effects , Mice , Humans , Biocompatible Materials/chemistry , Biocompatible Materials/pharmacology , Cell Line, Tumor
9.
Stem Cell Res Ther ; 15(1): 342, 2024 Oct 01.
Article in English | MEDLINE | ID: mdl-39354604

ABSTRACT

Orthodontic relapse (OR) occurs at a rate of over 70%. Retention is the current attempt at prevention, but it requires a considerable amount of time and cannot fully block OR. It's imperative to find a safe and effective method for managing post-orthodontic tooth stability. Periodontal bone remodeling is one crucial biological foundation of OR. Mesenchymal stem cell-derived exosomes (MSC-Exo) show promise in relapse management by regulating periodontal bone remodeling. MSC-Exo can prevent relapse by regulating periodontal ligament function, osteoclast activity, osteoblast differentiation, macrophage polarization, and periodontal microcirculation. In recent years, exosome-loaded hydrogels, which achieve controlled exosome release, have demonstrated efficacy in promoting bone regeneration and remodeling, offering promising prospects for OR management. This review aims to highlight the use of MSC-Exo-based therapy for preventing OR, offering new insights for future research focused on improving tooth stability and enhancing orthodontic anchorage.


Subject(s)
Exosomes , Mesenchymal Stem Cells , Exosomes/metabolism , Humans , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/cytology , Animals , Bone Remodeling
10.
Front Endocrinol (Lausanne) ; 15: 1410433, 2024.
Article in English | MEDLINE | ID: mdl-39359415

ABSTRACT

Background: Sex steroid hormones, primarily synthesized by gonadal somatic cells, are pivotal for sexual development and reproduction. Mice studies have shown that two transcription factors, steroidogenic factor 1 (SF-1) and Wilms' tumor 1 (WT1), are involved in gonadal development. However, their role in human gonadal somatic differentiation remains unclear. We therefore aimed to investigate the roles of SF-1 and WT1 in human gonadal steroidogenic cell differentiation. Methods: Using a transient lentivirus-mediated gene expression system, we assessed the effects of SF-1 and WT1 expression on the steroidogenic potential of human amniotic membrane-derived mesenchymal stem cells (hAmMSCs). Results: SF-1 and WT1-KTS, a splice variant of WT1, played distinct roles in human steroidogenic differentiation of hAmMSCs. SF-1 induced hAmMSC differentiation into progesterone- and androgen-producing cell lineages, whereas WT1-KTS promoted hAmMSC differentiation into estrogen-producing cell lineages. Conclusion: Our findings revealed that SF-1 and WT1-KTS play important roles in human gonadal steroidogenic cell differentiation, especially during ovarian development. These findings may pave the way for future studies on human ovarian differentiation and development.


Subject(s)
Amnion , Androgens , Cell Differentiation , Cell Lineage , Estrogens , Mesenchymal Stem Cells , Progesterone , Steroidogenic Factor 1 , WT1 Proteins , Humans , WT1 Proteins/metabolism , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/cytology , Steroidogenic Factor 1/metabolism , Steroidogenic Factor 1/genetics , Progesterone/metabolism , Progesterone/biosynthesis , Estrogens/metabolism , Androgens/metabolism , Amnion/cytology , Amnion/metabolism , Female , Cells, Cultured , RNA Splicing Factors
11.
Stem Cell Res Ther ; 15(1): 339, 2024 Sep 30.
Article in English | MEDLINE | ID: mdl-39350270

ABSTRACT

BACKGROUND: To investigate the long-term effects of combining bone marrow mesenchymal stem cells (MSCs) with mononuclear cells (MCs) in the treatment of type 2 diabetes mellitus (T2DM). METHODS: T2DM patients were divided into the combination group (Dual MSC + MC, n = 33), the mononuclear cell group (MC-Only, n = 32) and the control group (Control, n = 31). All groups were treated with insulin and metformin. The Dual MSC + MC group additionally received MSC and MC infusion and the MC-Only group additionally received MC infusion. The patients were followed up for 8 years. The primary endpoint was the C-peptide area under the curve (C-p AUC) at 1 year. This study was registered with clinicaltrial.gov (NCT01719640). RESULTS: A total of 97 patients were included and 89 completed the follow-up. The area under the curve of C-peptide of the Dual MSC + MC group and the MC-Only group was significantly increased (50.6% and 32.8%, respectively) at 1 year. After eight years of follow-up, the incidence of macrovascular complications was 13.8% (p = 0.009) in the Dual MSC + MC group and 21.4% (p = 0.061) in the MC-Only group, while it was 44.8% in the Control group. The incidence of diabetic peripheral neuropathy (DPN) was 10.3% (p = 0.0015) in the Dual MSC + MC group, 17.9% (p = 0.015) in the MC-Only group, and 48.3% in the Control group. CONCLUSIONS: The combination of MSC and MC therapy can reduce the incidence of chronic diabetes complications and improves metabolic control with mild side effects in T2DM patients.


Subject(s)
Diabetes Mellitus, Type 2 , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells , Humans , Diabetes Mellitus, Type 2/therapy , Male , Female , Mesenchymal Stem Cell Transplantation/methods , Middle Aged , Follow-Up Studies , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/cytology , Leukocytes, Mononuclear/metabolism , Aged , C-Peptide/metabolism , C-Peptide/blood , Adult , Combined Modality Therapy
12.
J R Soc Interface ; 21(219): 20240485, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39353563

ABSTRACT

Extracellular matrix (ECM) stiffness is fundamental in cell division, movement and differentiation. The stiffness that cells sense is determined not only by the elastic modulus of the ECM material but also by ECM geometry and cell density. We hypothesized that these factors would influence cell traction-induced matrix deformations and cellular differentiation in bone marrow stromal cells (BMSCs). To achieve this, we cultivated BMSCs on polyacrylamide hydrogels that varied in elastic modulus and geometry and measured cell spreading, cell-imparted matrix deformations and differentiation. At low cell density BMSCs spread to a greater extent on stiff compared with soft hydrogels, or on thin compared with thick hydrogels. Cell-imparted matrix deformations were greater on soft compared with stiff hydrogels or thick compared with thin hydrogels. There were no significant differences in osteogenic differentiation relative to hydrogel elastic modulus and thickness. However, increased cell density and/or prolonged culture significantly reduced matrix deformations on soft hydrogels to levels similar to those on stiff substrates. This suggests that at high cell densities cell traction-induced matrix displacements are reduced by both neighbouring cells and the constraint imposed by an underlying stiff support. This may explain observations of the lack of difference in osteogenic differentiation as a function of stiffness.


Subject(s)
Cell Differentiation , Hydrogels , Mesenchymal Stem Cells , Hydrogels/chemistry , Animals , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Extracellular Matrix/metabolism , Acrylic Resins/chemistry , Elastic Modulus , Mechanotransduction, Cellular/physiology , Osteogenesis/physiology , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Cells, Cultured
13.
Carbohydr Polym ; 345: 122564, 2024 Dec 01.
Article in English | MEDLINE | ID: mdl-39227103

ABSTRACT

Microcarriers for large-scale cell culture have a broader prospect in cell screening compared with the traditional high cost, low efficiency, and cell damaging methods. However, the equal biological affinity to cells has hindered its application. Therefore, based on the antifouling strategy of zwitterionic polymer, we developed a cell-specific microcarrier (CSMC) for shielding non-target cells and capturing mesenchymal stem cells (MSCs), which has characteristics of high biocompatibility, low background noise and high precision. Briefly, [2-(methacryloyloxy) ethyl] dimethyl-(3-sulfopropyl) ammonium hydroxide and glycidyl methacrylate were grafted onto polygalacturonic acid, respectively. The former built a hydration layer through solvation to provide an excellent antifouling surface, while the latter provided active sites for the click reaction with sulfhydryl-modified cell-specific peptides, resulting in rapid immobilization of peptides. This method is applicable to the vast majority of polysaccharide materials. The accurate capture ratio of MSCs by CSMC in a mixed multicellular environment is >95 % and the proliferation rate of MSCs on microcarriers is satisfactory. In summary, this grafting strategy of bioactive components lays a foundation for the application of polysaccharide materials in the biomedical field, and the specific adhesive microcarriers also open up new ideas for the development of stem cell screening as well.


Subject(s)
Mesenchymal Stem Cells , Pectins , Peptides , Mesenchymal Stem Cells/cytology , Pectins/chemistry , Peptides/chemistry , Methacrylates/chemistry , Cell Proliferation/drug effects , Epoxy Compounds/chemistry , Humans , Animals , Biocompatible Materials/chemistry
14.
J Biomed Mater Res B Appl Biomater ; 112(9): e35482, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39269164

ABSTRACT

The stromal vascular fraction (SVF) is a derivate of fat tissue comprising both adipose-derived mesenchymal stem cells and endothelial cells and serves as a promising cell source for engineering vascularized bone tissues. Its combination with osteoconductive biphasic calcium phosphate (BCP) ceramic may represent a point-of-care agent for bone reconstruction. Here we assessed the proliferation and osteogenic differentiation capacities of SVF on 3D printed BCP implants, in comparison with isolated adipose-derived mesenchymal stem cells (AD-MSCs). AD-MSCs and SVF isolated from human donors were seeded on plastic or 3D printed BCP ceramics with sinusoidal or gyroid macrotopography and cultured in the presence or absence of osteogenic factors. Vascular, hematopoietic and MSC surface markers were assessed by flow cytometry whereas osteogenic activity was investigated through alizarin red staining and alkaline phosphatase activity. Osteogenic factors were necessary to trigger osteogenic activity when cells were cultured on plastic, without significant difference observed between the two cell populations. Interestingly, osteogenic activity was observed on BCP implants in the absence of differentiation factors, without significant difference in level activity between the two cell populations and macrotopography. This study offers supportive data for the use of combined BCP scaffolds with SVF in a perspective of a one-step surgical procedure for bone regeneration.


Subject(s)
Cell Differentiation , Ceramics , Mesenchymal Stem Cells , Osteogenesis , Printing, Three-Dimensional , Humans , Osteogenesis/drug effects , Cell Differentiation/drug effects , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/cytology , Ceramics/pharmacology , Ceramics/chemistry , Hydroxyapatites/chemistry , Hydroxyapatites/pharmacology , Tissue Scaffolds/chemistry , Adipose Tissue/cytology , Adipose Tissue/metabolism , Cells, Cultured
15.
Stem Cell Res Ther ; 15(1): 287, 2024 Sep 11.
Article in English | MEDLINE | ID: mdl-39256806

ABSTRACT

BACKGROUND: Androgenetic alopecia (AGA) is a common form of hair loss. Androgens, such as testosterone and dihydrotestosterone, are the main causes of AGA. Extracellular vesicles (EVs) derived from mesenchymal stem cells (MSCs) can reduce AGA. However, preparing therapeutic doses of MSCs for clinical use is challenging. Induced pluripotent stem cell-derived MSCs (iMSCs) are homogenous and easily expandable, enabling scalable production of EVs. Hyaluronic acid (HA) can exert various functions including free radical scavenging, immune regulation, and cell migration. Herein, we examined whether hyaluronic acid (HA) stimulation of iMSCs could produce EVs with enhanced therapeutic outcomes for AGA. METHODS: EVs were collected from iMSCs primed with HA (HA-iMSC-EVs) or without HA (iMSC-EVs). The characteristics of EVs were examined using dynamic light scattering, cryo-transmission electron microscopy, immunoblotting, flow cytometry, and proteomic analysis. In vitro, we compared the potential of EVs in stimulating the survival of hair follicle dermal papilla cells undergoing testosterone-mediated AGA. Additionally, the expression of androgen receptor (AR) and relevant growth factors as well as key proteins of Wnt/ß-catenin signaling pathway (ß-catenin and phosphorylated GSK3ß) was analyzed. Subsequently, AGA was induced in male C57/BL6 mice by testosterone administration, followed by repeated injections of iMSC-EVs, HA-iMSC-EVs, finasteride, or vehicle. Several parameters including hair growth, anagen phase ratio, reactivation of Wnt/ß-catenin pathway, and AR expression was examined using qPCR, immunoblotting, and immunofluorescence analysis. RESULTS: Both types of EVs showed typical characteristics for EVs, such as size distribution, markers, and surface protein expression. In hair follicle dermal papilla cells, the mRNA levels of AR, TGF-ß, and IL-6 increased by testosterone was blocked by HA-iMSC-EVs, which also contributed to the augmented expression of trophic genes related to hair regrowth. However, no notable changes were observed in the iMSC-EVs. Re-activation of Wnt/ß-catenin was observed in HA-iMSC-EVs but not in iMSC-EVs, as shown by ß-catenin stabilization and an increase in phosphorylated GSK3ß. Restoration of hair growth was more significant in HA-iMSC-EVs than in iMSC-EVs, and was comparable to that in mice treated with finasteride. Consistently, the decreased anagen ratio induced by testosterone was reversed by HA-iMSC-EVs, but not by iMSC-EVs. An increased expression of hair follicular ß-catenin protein, as well as the reduction of AR was observed in the skin tissue of AGA mice receiving HA-iMSC-EVs, but not in those treated with iMSC-EVs. CONCLUSIONS: Our results suggest that HA-iMSC-EVs have potential to improve AGA by regulating growth factors/cytokines and stimulating AR-related Wnt/ß-catenin signaling.


Subject(s)
Alopecia , Extracellular Vesicles , Hair Follicle , Hyaluronic Acid , Mesenchymal Stem Cells , Extracellular Vesicles/metabolism , Alopecia/therapy , Alopecia/metabolism , Alopecia/drug therapy , Hyaluronic Acid/pharmacology , Hyaluronic Acid/metabolism , Animals , Mice , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/drug effects , Hair Follicle/metabolism , Hair Follicle/drug effects , Humans , Wnt Signaling Pathway/drug effects , Male , Receptors, Androgen/metabolism , Receptors, Androgen/genetics , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/cytology , Testosterone/pharmacology , Glycogen Synthase Kinase 3 beta/metabolism , Mice, Inbred C57BL
16.
Stem Cell Res Ther ; 15(1): 288, 2024 Sep 11.
Article in English | MEDLINE | ID: mdl-39256816

ABSTRACT

BACKGROUND: Mesenchymal stromal cell (MSC) therapy holds great potential yet efficacy and safety concerns with cell therapy persist. The beneficial effects of MSCs are often attributed to their secretome that includes extracellular vesicles (EVs). EVs carry biologically active molecules, protected by a lipid bilayer. However, several barriers hinder large-scale MSC EV production. A serum-free culturing approach is preferred for producing clinical-grade MSC-derived EVs but this can affect both yield and purity. Consequently, new strategies have been explored, including genetically engineering MSCs to alter EV compositions to enhance potency, increase circulation time or mediate targeting. However, efficient transfection of MSCs is challenging. Typical sources of MSC include adipose tissue and bone marrow, which both require invasive extraction procedures. Here, we investigate the use of urine-derived stem cells (USCs) as a non-invasive and inexhaustible source of MSCs for EV production. METHODS: We isolated, expanded, and characterized urine-derived stem cells (USCs) harvested from eight healthy donors at three different time points during the day. We evaluated the number of clones per urination, proliferation capacity and conducted flow cytometry to establish expression of surface markers. EVs were produced in chemically defined media and characterized. PEI/DNA transfection was used to genetically engineer USCs using transposon technology. RESULTS: There were no differences between time points for clone number, doubling time or viability. USCs showed immunophenotypic characteristics of MSCs, such as expression of CD73, CD90 and CD105, with no difference at the assessed time points, however, male donors had reduced CD73 + cells. Expanded USCs were incubated without growth factors or serum for 72 h without a loss in viability and EVs were isolated. USCs were transfected with high efficiency and after 10 days of selection, pure engineered cell cultures were established. CONCLUSIONS: Isolation and expansion of MSCs from urine is non-invasive, robust, and without apparent sex-related differences. The sampling time point did not affect any measured markers or USC isolation potential. USCs offer an attractive production platform for EVs, both native and engineered.


Subject(s)
Extracellular Vesicles , Mesenchymal Stem Cells , Urine , Humans , Extracellular Vesicles/metabolism , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/cytology , Urine/cytology , Male , Female , Cells, Cultured , Adult , Cell Proliferation , Middle Aged , Cell Differentiation
17.
Stem Cell Res Ther ; 15(1): 289, 2024 Sep 11.
Article in English | MEDLINE | ID: mdl-39256841

ABSTRACT

BACKGROUND: Mesenchymal Stromal Cells (MSCs) are the preferred candidates for therapeutics as they possess multi-directional differentiation potential, exhibit potent immunomodulatory activity, are anti-inflammatory, and can function like antimicrobials. These capabilities have therefore encouraged scientists to undertake numerous preclinical as well as a few clinical trials to access the translational potential of MSCs in disease therapeutics. In spite of these efforts, the efficacy of MSCs has not been consistent-as is reflected in the large variation in the values of outcome measures like survival rates. Survival rate is a resultant of complex cascading interactions that not only depends upon upstream experimental factors like dosage, time of infusion, type of transplant, etc.; but is also dictated, post-infusion, by intrinsic host specific attributes like inflammatory microniche including proinflammatory cytokines and alarmins released by the damaged host cells. These complex interdependencies make a researcher's task of designing MSC transfusion experiments challenging. METHODS: In order to identify the rules and associated attributes that influence the final outcome (survival rates) of MSC transfusion experiments, we decided to apply machine learning techniques on manually curated data collected from available literature. As sepsis is a multi-faceted condition that involves highly dysregulated immune response, inflammatory environment and microbial invasion, sepsis can be an efficient model to verify the therapeutic effects of MSCs. We therefore decided to implement rule-based classification models on data obtained from studies involving interventions of MSCs in sepsis preclinical models. RESULTS: The rules from the generated graph models indicated that survival rates, post-MSC-infusion, are influenced by factors like source, dosage, time of infusion, pre-Interleukin-6 (IL-6)/ Tumour Necrosis Factor- alpha (TNF-α levels, etc. CONCLUSION: This approach provides important information for optimization of MSCs based treatment strategies that may help the researchers design their experiments.


Subject(s)
Machine Learning , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells , Sepsis , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/cytology , Humans , Sepsis/therapy , Mesenchymal Stem Cell Transplantation/methods , Animals
18.
Chin J Dent Res ; 27(3): 225-234, 2024 Sep 02.
Article in English | MEDLINE | ID: mdl-39221983

ABSTRACT

OBJECTIVE: To reveal the role and mechanism of cannabinoid receptor 1 (CB1) and mitochondria in promoting osteogenic differentiation of periodontal ligament stem cells (PDLSCs) in the inflammatory microenvironment. METHODS: Bidirectional mitochondrial transfer was performed in bone mesenchymal stem cells (BMSCs) and PDLSCs. Laser confocal microscopy and quantitative flow cytometry were used to observe the mitochondrial transfer and quantitative mitochondrial transfer efficiency. Realtime reverse transcription polymerase chain reaction (RT-PCR) was employed to detect gene expression. Alkaline phosphatase (ALP) activity, alizarin red staining (ARS) and quantitative calcium ion analysis were used to evaluate the degree of osteogenic differentiation of PDLSCs. RESULTS: Bidirectional mitochondrial transfer was observed between BMSCs and PDLSCs. The indirect co-culture system could simulate intercellular mitochondrial transfer. Compared with the conditioned medium (CM) for BMSCs, that for HA-CB1 BMSCs could significantly enhance the mineralisation ability of PDLSCs. The mineralisation ability of PDLSCs could not be enhanced after removing the mitochondria in CM for HA-CB1 BMSCs. The expression level of HO-1, PGC-1α, NRF-1, ND1 and HK2 was significantly increased in HA-CB1 BMSCs. CONCLUSION: CM for HA-CB1 BMSCs could significantly enhance the damaged osteogenic differentiation ability of PDLSCs in the inflammatory microenvironment, and the mitochondria of CM played an important role. CB1 was related to the activation of the HO-1/PGC-1α/NRF-1 mitochondrial biogenesis pathway, and significantly increased the mitochondrial content in BMSCs.


Subject(s)
Cell Differentiation , Mesenchymal Stem Cells , Mitochondria , Osteogenesis , Periodontal Ligament , Receptor, Cannabinoid, CB1 , Adolescent , Humans , Bone Marrow Cells , Cells, Cultured , Coculture Techniques , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/cytology , Mitochondria/metabolism , Osteogenesis/physiology , Periodontal Ligament/cytology , Receptor, Cannabinoid, CB1/metabolism , Receptor, Cannabinoid, CB1/genetics
19.
J Nanobiotechnology ; 22(1): 540, 2024 Sep 05.
Article in English | MEDLINE | ID: mdl-39237942

ABSTRACT

To assess the efficacy of a novel 3D biomimetic hydrogel scaffold with immunomodulatory properties in promoting fracture healing. Immunomodulatory scaffolds were used in cell experiments, osteotomy mice treatment, and single-cell transcriptomic sequencing. In vitro, fluorescence tracing examined macrophage mitochondrial transfer and osteogenic differentiation of bone marrow-derived mesenchymal stem cells (BMSCs). Scaffold efficacy was assessed through alkaline phosphatase (ALP), Alizarin Red S (ARS) staining, and in vivo experiments. The scaffold demonstrated excellent biocompatibility and antioxidant-immune regulation. Single-cell sequencing revealed a shift in macrophage distribution towards the M2 phenotype. In vitro experiments showed that macrophage mitochondria promoted BMSCs' osteogenic differentiation. In vivo experiments confirmed accelerated fracture healing. The GAD/Ag-pIO scaffold enhances osteogenic differentiation and fracture healing through immunomodulation and promotion of macrophage mitochondrial transfer.


Subject(s)
Cell Differentiation , Hydrogels , Macrophages , Mesenchymal Stem Cells , Mitochondria , Osteogenesis , Tissue Scaffolds , Animals , Osteogenesis/drug effects , Mitochondria/metabolism , Mitochondria/drug effects , Macrophages/drug effects , Macrophages/metabolism , Macrophages/cytology , Hydrogels/chemistry , Hydrogels/pharmacology , Cell Differentiation/drug effects , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/drug effects , Mice , Tissue Scaffolds/chemistry , Male , Cells, Cultured , Mice, Inbred C57BL
20.
J Nanobiotechnology ; 22(1): 543, 2024 Sep 05.
Article in English | MEDLINE | ID: mdl-39238005

ABSTRACT

BACKGROUND: Human mesenchymal stem cells have attracted interest in regenerative medicine and are being tested in many clinical trials. In vitro expansion is necessary to provide clinical-grade quantities of mesenchymal stem cells; however, it has been reported to cause replicative senescence and undefined dysfunction in mesenchymal stem cells. Quality control assessments of in vitro expansion have rarely been addressed in ongoing trials. Young small extracellular vesicles from the remnant pulp of human exfoliated deciduous teeth stem cells have demonstrated therapeutic potential for diverse diseases. However, it is still unclear whether young small extracellular vesicles can reverse senescence-related declines. RESULTS: We demonstrated that mitochondrial structural disruption precedes cellular dysfunction during bone marrow-derived mesenchymal stem cell replication, indicating mitochondrial parameters as quality assessment indicators of mesenchymal stem cells. Dynamin-related protein 1-mediated mitochondrial dynamism is an upstream regulator of replicative senescence-induced dysfunction in bone marrow-derived mesenchymal stem cells. We observed that the application of young small extracellular vesicles could rescue the pluripotency dissolution, immunoregulatory capacities, and therapeutic effects of replicative senescent bone marrow-derived mesenchymal stem cells. Mechanistically, young small extracellular vesicles could promote Dynamin-related protein 1 translocation from the cytoplasm to the mitochondria and remodel mitochondrial disruption during replication history. CONCLUSIONS: Our findings show that Dynamin-related protein 1-mediated mitochondrial disruption is associated with the replication history of bone marrow-derived mesenchymal stem cells. Young small extracellular vesicles from human exfoliated deciduous teeth stem cells alleviate replicative senescence by promoting Dynamin-related protein 1 translocation onto the mitochondria, providing evidence for a potential rejuvenation strategy.


Subject(s)
Cellular Senescence , Dynamins , Extracellular Vesicles , Mesenchymal Stem Cells , Mitochondria , Mitochondrial Dynamics , Humans , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/cytology , Extracellular Vesicles/metabolism , Dynamins/metabolism , Mitochondria/metabolism , Animals , Cells, Cultured , Mice , Male , Tooth, Deciduous/cytology , Tooth, Deciduous/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL