Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 5.738
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 ; 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
4.
Stem Cell Res Ther ; 15(1): 343, 2024 Oct 01.
Article in English | MEDLINE | ID: mdl-39354635

ABSTRACT

Spinal cord injury (SCI) results in significant neural damage and inhibition of axonal regeneration due to an imbalanced microenvironment. Extensive evidence supports the efficacy of mesenchymal stem cell (MSC) transplantation as a therapeutic approach for SCI. This review aims to present an overview of MSC regulation on the imbalanced microenvironment following SCI, specifically focusing on inflammation, neurotrophy and axonal regeneration. The application, limitations and future prospects of MSC transplantation are discussed as well. Generally, a comprehensive perspective is provided for the clinical translation of MSC transplantation for SCI.


Subject(s)
Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells , Spinal Cord Injuries , Spinal Cord Injuries/therapy , Spinal Cord Injuries/pathology , Mesenchymal Stem Cells/metabolism , Humans , Mesenchymal Stem Cell Transplantation/methods , Animals , Cellular Microenvironment , Nerve Regeneration
5.
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
6.
Neurodegener Dis Manag ; 14(3-4): 111-117, 2024.
Article in English | MEDLINE | ID: mdl-39352708

ABSTRACT

Background: In this pilot safety study, we hypothesized that a human bone marrow stem cell-derived extracellular vesicle (hBM-MSC EV) investigational product (IP) would be safe and exhibit potential efficacy in amyotrophic lateral sclerosis (ALS) patients.Methods: Ten ALS patients received two 10-ml intravenous infusions of the IP given 1 month apart and evaluated over 3 months.Results: There were no serious adverse events or adverse events related to the IP and 30% of subjects' ALS functional rating scale-revised (ALSFRS-R) scores did not decline.Conclusion: HBM-MSC EVs appear safe in ALS patients. This early investigation suggests a controlled study of EVs for the treatment of ALS is warranted.


Amyotrophic lateral sclerosis (ALS) is a nervous system disease that affects the brain and spinal cord, causing the loss of muscle control. Currently, there is no cure for ALS and the disease gets worse over time. A potential new treatment is being investigated using mesenchymal stem cell extracellular vesicles (MSC EVs). MSC EVs are small structures that contain useful molecules and proteins that can be transported to cells affected by the disease, helping to reduce inflammation and encouraging repair. This 3-month study looked at the safety of human bone marrow MSC-EVs (hBM-MSC EVs) given as treatment to ten ALS patients, as well as how well it worked at delaying worsening of the disease. They found that there were no serious side effects caused by the treatment and that hBM-MSC EVs may have the potential for delaying the progression of ALS. This indicates that more, larger studies need to be carried out to find out treatment specifics, such as dose (how much of the treatment to give) and frequency (how often to give the treatment), and how they could be related to patient outcomes.


Subject(s)
Amyotrophic Lateral Sclerosis , Extracellular Vesicles , Humans , Amyotrophic Lateral Sclerosis/therapy , Male , Middle Aged , Female , Pilot Projects , Aged , Mesenchymal Stem Cells , Mesenchymal Stem Cell Transplantation/methods , Infusions, Intravenous , Adult , Treatment Outcome
7.
Skin Res Technol ; 30(9): e70022, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39221632

ABSTRACT

BACKGROUND: Despite advancements in reconstructive procedures, ischemia-reperfusion (I/R) injury remains a significant challenge in reconstructive surgery, with mitochondrial dysfunction playing a pivotal role. Mitochondrial transplantation has emerged as a promising therapeutic strategy to address this issue. This study aims to evaluate the impact of umbilical cord mesenchymal stem cell-derived mitochondrial transplantation on skin flap I/R models in rats. MATERIAL AND METHODS: Twenty male rats underwent I/R injury on skin flaps, with or without mitochondrial transplantation administered via intravenous or subcutaneous routes. Analysis encompassed histopathology, inflammatory, apoptotic, oxidative stress, and hypoxia markers. RESULTS: Results revealed a reduction in inflammation, apoptosis, oxidative stress, and hypoxia in the transplantation group compared to controls. CONCLUSION: The findings suggest that umbilical cord mesenchymal stem cell-derived mitochondrial transplantation shows promise in enhancing flap viability and attenuating I/R injury, offering valuable insights for improved outcomes in reconstructive surgery. However, further exploration in larger animal models and refinement of delivery methods and dosage are warranted to fully elucidate its clinical translatability.


Subject(s)
Disease Models, Animal , Mesenchymal Stem Cell Transplantation , Mitochondria , Reperfusion Injury , Umbilical Cord , Animals , Male , Rats , Mesenchymal Stem Cell Transplantation/methods , Umbilical Cord/cytology , Mitochondria/transplantation , Mitochondria/metabolism , Rats, Sprague-Dawley , Mesenchymal Stem Cells , Surgical Flaps/pathology , Oxidative Stress , Apoptosis
8.
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
9.
J Nanobiotechnology ; 22(1): 555, 2024 Sep 11.
Article in English | MEDLINE | ID: mdl-39261846

ABSTRACT

BACKGROUND: The pathogenesis of osteoarthritis (OA) involves the progressive degradation of articular cartilage. Exosomes derived from mesenchymal stem cells (MSC-EXOs) have been shown to mitigate joint pathological injury by attenuating cartilage destruction. Optimization the yield and therapeutic efficacy of exosomes derived from MSCs is crucial for promoting their clinical translation. The preconditioning of MSCs enhances the therapeutic potential of engineered exosomes, offering promising prospects for application by enabling controlled and quantifiable external stimulation. This study aims to address these issues by employing pro-inflammatory preconditioning of MSCs to enhance exosome production and augment their therapeutic efficacy for OA. METHODS: The exosomes were isolated from the supernatant of infrapatellar fat pad (IPFP)-MSCs preconditioned with a pro-inflammatory factor, TNF-α, and their production was subsequently quantified. The exosome secretion-related pathways in IPFP-MSCs were evaluated through high-throughput transcriptome sequencing analysis, q-PCR and western blot analysis before and after TNF-α preconditioning. Furthermore, exosomes derived from TNF-α preconditioned IPFP-MSCs (IPFP-MSC-EXOsTNF-α) were administered intra-articularly in an OA mouse model, and subsequent evaluations were conducted to assess joint pathology and gait alterations. The expression of proteins involved in the maintenance of cartilage homeostasis within the exosomes was determined through proteomic analysis. RESULTS: The preconditioning with TNF-α significantly enhanced the exosome secretion of IPFP-MSCs compared to unpreconditioned MSCs. The potential mechanism involved the activation of the PI3K/AKT signaling pathway in IPFP-MSCs by TNF-α precondition, leading to an up-regulation of autophagy-related protein 16 like 1(ATG16L1) levels, which subsequently facilitated exosome secretion. The intra-articular administration of IPFP-MSC-EXOsTNF-α demonstrated superior efficacy in ameliorating pathological changes in the joints of OA mice. The preconditioning of TNF-α enhanced the up-regulation of low-density lipoprotein receptor-related protein 1 (LRP1) levels in IPFP-MSC-EXOsTNF-α, thereby exerting chondroprotective effects. CONCLUSION: TNF-α preconditioning constitutes an effective and promising method for optimizing the therapeutic effects of IPFP-MSCs derived exosomes in the treatment of OA.


Subject(s)
Exosomes , Mesenchymal Stem Cells , Osteoarthritis , Tumor Necrosis Factor-alpha , Exosomes/metabolism , Animals , Mesenchymal Stem Cells/metabolism , Tumor Necrosis Factor-alpha/metabolism , Mice , Osteoarthritis/therapy , Osteoarthritis/metabolism , Adipose Tissue/cytology , Mice, Inbred C57BL , Male , Disease Models, Animal , Cartilage, Articular/metabolism , Mesenchymal Stem Cell Transplantation/methods , Cells, Cultured , Humans
10.
Adv Exp Med Biol ; 1456: 85-91, 2024.
Article in English | MEDLINE | ID: mdl-39261425

ABSTRACT

Induced pluripotent stem cells (iPSCs) are a promising in vitro model for drug-screening and precision-based psychiatry for the treatment of major depressive disorders (MDD). In this chapter, we explore different uses for iPSC technology, three-dimensional (3D) organoids models, and mesenchymal stem cells therapy in MDD, as well their potential and limitations.


Subject(s)
Depressive Disorder, Major , Induced Pluripotent Stem Cells , Organoids , Humans , Induced Pluripotent Stem Cells/cytology , Depressive Disorder, Major/therapy , Organoids/metabolism , Animals , Mesenchymal Stem Cell Transplantation/methods , Cell Differentiation
11.
Chin J Dent Res ; 27(3): 215-224, 2024 Sep 02.
Article in English | MEDLINE | ID: mdl-39221982

ABSTRACT

OBJECTIVE: To investigate whether bone marrow mesenchymal stem cells (BMMSCs) modulate periodontal bone repair through the hydroxylase domain-containing protein 2 (PHD2)/hypoxia- inducible factor-1 (HIF-1) signalling pathway in response to inflammatory conditions. METHODS: Osteogenic differentiation of PHD2 shRNA-modified BMMSCs and the possible mechanism were explored in an inflammatory microenvironment stimulated by porphyromonas gingivalis lipopolysaccharide (Pg-LPS) in vitro. The effect of PHD2 gene-modified BMMSCs on periodontal bone loss was evaluated with experimental periodontitis. RESULTS: Pg-LPS stimulation greatly impaired the osteogenic differentiation of BMMSCs, whereas the silence of PHD2 significantly enhanced the osteogenesis of BMMSCs. More importantly, increased level of vascular endothelial growth factor (VEGF) was detected under Pg-LPS stimulation, which was verified to be associated with the augmented osteogenesis. In experimental periodontitis, PHD2-modified BMMSCs transplantation elevated osteogenic parameters and the expression of VEGF in periodontal tissue. CONCLUSION: This study highlighted that PHD2 gene silencing could be a feasible approach to combat inflammatory bone loss by rescuing the dysfunction of seed cells.


Subject(s)
Hypoxia-Inducible Factor-Proline Dioxygenases , Mesenchymal Stem Cells , Osteogenesis , RNA, Small Interfering , Animals , RNA, Small Interfering/genetics , Osteogenesis/genetics , Hypoxia-Inducible Factor-Proline Dioxygenases/genetics , Porphyromonas gingivalis , Periodontitis/therapy , Periodontitis/genetics , Vascular Endothelial Growth Factor A/metabolism , Mesenchymal Stem Cell Transplantation/methods , Cell Differentiation , Lipopolysaccharides , Alveolar Bone Loss , Mice , Male , Bone Marrow Cells , Bone Regeneration/genetics
12.
Stem Cell Res Ther ; 15(1): 315, 2024 Sep 19.
Article in English | MEDLINE | ID: mdl-39300579

ABSTRACT

BACKGROUND: Liver fibrosis can progress to end-stage cirrhosis and liver cancer. Mesenchymal stem cells (MSCs) were considered the most promising therapeutic strategy, but most of the MSCs injected intravenously traditionally are trapped in the lungs, rapidly reducing their survival ability. MSC spheroids cultured in 3D have shown higher tolerance to fluid shear stress and better survival than dissociated MSCs. Simulating the route of orthotopic liver transplantation, transplanting MSC spheroids into the liver via hepatic portal vein may impact superior therapeutic effects. METHODS: In the present study, human umbilical cord-derived MSC spheroids (hUC-MSCsp) were transplanted into rhesus monkey models of liver fibrosis via B-ultrasound-guided percutaneous portal vein puncture with minimized body invasion. The therapeutic effect is evaluated through hematology, ultrasound, and pathology. To study the effect of hUC-MSCsp on gene expression in rhesus monkeys with liver injury, transcriptome sequencing analysis was performed on the livers of rhesus monkeys. The distribution of transplanted hUC-MSCsp was traced with RNA scope technology. RESULTS: We found that hUC-MSCsp significantly restored liver function, including ALT, AST, ALB, GLOB and bilirubin. hUC-MSCsp also significantly reduced liver collagen deposition and inflammatory infiltration, and promote dismission of liver ascites. Subsequently, the therapeutic effects were further validated in TGF-ß1/Smad pathway by global transcription profile. The distribution of transplanted hUC-MSCsp were also tracked, and we found that hUC-MSCsp distributed in the liver in a sphere status at 1 h after transplantation. After 16 days, the hUC-MSCsp were dispersed into dissociated cells that were predominantly distributed in the spleen, and a significant number of dissociated cells were still present in the liver. CONCLUSIONS: This study reveals the distributions of transplanted hUC-MSCsp after liver portal vein transplantation, and provides a novel approach and new insights into the molecular events of potential molecular events underlying the treatment of liver fibrosis with hUC-MSCsp.


Subject(s)
Liver Cirrhosis , Macaca mulatta , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells , Portal Vein , Umbilical Cord , Animals , Humans , Mesenchymal Stem Cell Transplantation/methods , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/cytology , Umbilical Cord/cytology , Liver Cirrhosis/therapy , Liver Cirrhosis/pathology , Disease Models, Animal , Spheroids, Cellular/metabolism , Ultrasonography/methods , Liver/pathology , Liver/metabolism
13.
Stem Cell Res Ther ; 15(1): 316, 2024 Sep 20.
Article in English | MEDLINE | ID: mdl-39304926

ABSTRACT

BACKGROUND: Coronavirus disease 2019 (COVID-19) has created a global pandemic with significant morbidity and mortality. SARS-CoV-2 primarily infects the lungs and is associated with various organ complications. Therapeutic approaches to combat COVID-19, including convalescent plasma and vaccination, have been developed. However, the high mutation rate of SARS-CoV-2 and its ability to inhibit host T-cell activity pose challenges for effective treatment. Mesenchymal stem cells (MSCs) and their extracellular vesicles (MSCs-EVs) have shown promise in COVID-19 therapy because of their immunomodulatory and regenerative properties. MicroRNAs (miRNAs) play crucial regulatory roles in various biological processes and can be manipulated for therapeutic purposes. OBJECTIVE: We aimed to investigate the role of lyophilized MSC-EVs and their microRNAs in targeting the receptors involved in SARS-CoV-2 entry into host cells as a strategy to limit infection. In silico microRNA prediction, structural predictions of the microRNA-mRNA duplex, and molecular docking with the Argonaut protein were performed. METHODS: Male Syrian hamsters infected with SARS-CoV-2 were treated with human Wharton's jelly-derived Mesenchymal Stem cell-derived lyophilized exosomes (Bioluga Company)via intraperitoneal injection, and viral shedding was assessed. The potential therapeutic effects of MSCs-EVs were measured via histopathology of lung tissues and PCR for microRNAs. RESULTS: The results revealed strong binding potential between miRNA‒mRNA duplexes and the AGO protein via molecular docking. MSCs-EVs reduced inflammation markers and normalized blood indices via the suppression of viral entry by regulating ACE2 and TMPRSS2 expression. MSCs-EVs alleviated histopathological aberrations. They improved lung histology and reduced collagen fiber deposition in infected lungs. CONCLUSION: We demonstrated that MSCs-EVs are a potential therapeutic option for treating COVID-19 by preventing viral entry into host cells.


Subject(s)
COVID-19 , Extracellular Vesicles , Mesenchymal Stem Cells , MicroRNAs , SARS-CoV-2 , COVID-19/therapy , COVID-19/metabolism , COVID-19/pathology , COVID-19/virology , Animals , Extracellular Vesicles/metabolism , MicroRNAs/metabolism , MicroRNAs/genetics , SARS-CoV-2/metabolism , SARS-CoV-2/genetics , Mesenchymal Stem Cells/metabolism , Humans , Male , Mesocricetus , Virus Internalization , Angiotensin-Converting Enzyme 2/metabolism , Angiotensin-Converting Enzyme 2/genetics , Molecular Docking Simulation , Computer Simulation , Cricetinae , Mesenchymal Stem Cell Transplantation/methods
14.
Stem Cell Res Ther ; 15(1): 311, 2024 Sep 18.
Article in English | MEDLINE | ID: mdl-39294780

ABSTRACT

BACKGROUND: Mesenchymal stem cells (MSCs) have shown a positive effect on Osteoarthritis (OA), but the efficacy is still not significant in clinical. Conventional two-dimensional (2D) monolayer culture method is prone to cause MSCs undergoing replication senescence, which may affect the functions of MSCs. Three-dimensional (3D) culture strategy can sustain cell proliferative capacity and multi-differentiation potential. This study aimed to investigate the therapeutic potential of human umbilical cord-derived mesenchymal stem cells (hUC-MSCs) cultured by 3D hanging drop method on OA. METHODS: hUC-MSCs were isolated from umbilical cord and cultured by 3D hanging drop method for 48 h. Scanning electron microscopy (SEM) was used to observe gross morphology 2D and 3D hUC-MSCs. Transcriptome comparison of gene expression differences between 2D and 3D hUC-MSCs. GO enrichment analysis, KEGG pathway enrichment analysis and GSEA enrichment analysis were used to analyze the impact of 3D hanging drop culture on the biological functions of hUC-MSCs. Female New Zealand rabbits (n = 12) were divided into 4 groups: Normal group, Model group, 2D hUC-MSCs treatment group and 3D hUC-MSCs treatment group. After 8 weeks, the gross and histological appearance of the cartilage was evaluated by safranin O-fast green staining and Mankin scoring system. The expression of type I collagen and type II collagen was detected by immunohistochemistry. The levels of IL-6, IL-7, TNFα, TGFß1 and IL-10 in the knee joint fluid were tested by ELISA. RESULTS: 3D hanging drop culture changed cell morphology but did not affect phenotype. The MSCs transcriptome profiles showed that 3D hanging drop culture method enhanced cell-cell contact, improved cell responsiveness to external stimuli and immunomodulatory function. The animal experiment results showed that hUC-MSCs could promote cartilage regeneration compared with Model group. 3D hUC-MSCs treatment group had a higher histological score and significantly increased type II collagen secretion. In addition, 3D hUC-MSCs treatment group increased the expression of anti-inflammatory factors TGFß1 and IL-10. CONCLUSION: The above experimental results illustrated that 3D hanging drop culture method could enhance the therapeutic effect of hUC-MSCs, and showed a good clinical application prospect in the treatment of OA.


Subject(s)
Mesenchymal Stem Cells , Osteoarthritis , Umbilical Cord , Animals , Rabbits , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/cytology , Humans , Umbilical Cord/cytology , Osteoarthritis/therapy , Osteoarthritis/pathology , Osteoarthritis/metabolism , Female , Mesenchymal Stem Cell Transplantation/methods , Cell Differentiation , Cells, Cultured , Cell Culture Techniques, Three Dimensional/methods , Cell Proliferation
15.
Probl Endokrinol (Mosk) ; 70(4): 4-14, 2024 Sep 15.
Article in Russian | MEDLINE | ID: mdl-39302860

ABSTRACT

The number of patients with diabetes mellitus (DM) has been progressively increasing worldwide over the past decades, and many international organizations consider DM as a public health emergency of the 21st century.Critical limb ischemia (CLI) is the most severe stage of peripheral arterial disease (PAD) in DM and is characterized by a high risk of limb loss without revascularization. Traditional treatment tactics include open and endovascular revascularization surgical techniques. However, in patients not eligible for revascularization and in cases where performed surgical treatment performed has been ineffective, there are almost no therapeutic alternatives, often leading to amputations and death. As of today, one of the newest non-surgical treatment options is cell therapy. Among different cells, mesenchymal stromal cells (MSCs) are potentially one of the most prospective for use in this patient population.This article provides an overview of clinical trials using cell therapy in patients with CLI.To analyze publications, electronic databases PubMed, SCOPUS, ClinicalTrials, and ScienceDirect were searched to identify published data from clinical trials, research studies, and review articles on cell therapy for critical lower extremity ischemia. After the search, 489 results were received.As a result of systematic selection, 22 clinical trials were analyzed.According to the analyzed literature data, the use of cell products in this category of patients is effective and safe. Cell therapy can stimulate the formation of new vessels and enhances collateral circulation; it is also reported improved distal perfusion, increased pain-free walking distance, decreased amputation rates, and increased survival rates.Nevertheless, further study of the potential use of this category of drugs is needed.


Subject(s)
Ischemia , Humans , Ischemia/therapy , Mesenchymal Stem Cell Transplantation/methods , Cell- and Tissue-Based Therapy/methods , Lower Extremity/blood supply , Peripheral Arterial Disease/therapy
17.
Sci Rep ; 14(1): 21897, 2024 09 19.
Article in English | MEDLINE | ID: mdl-39300232

ABSTRACT

Psoriasis is a chronic, non-contagious, immune-mediated skin disorder. Inflammation of the skin's surface is characterised by scaly white, red, or silvery spots that occur due to the hyper-proliferation of keratinocytes in the epidermal layer. Primarily, pharmaceutical drugs or immune therapy are used to treat psoriasis. We are all aware that, certain therapeutic strategies can have some adverse effects, and over time, that hidden inflammation may manifest. This article introduces a mathematical model for psoriasis, formulated by employing a set of nonlinear ordinary differential equations (ODEs) that describe the densities of T-cells, dendritic cells (DCs), keratinocytes, and mesenchymal stromal cells (MSCs) as basic cell populations. A tumor necrosis factor- α ( T N F - α ) inhibitor has been imposed from the initial stage of the treatment regime, using the optimal control theoretic approach, and the numerical results have been observed. After 80 days of monitoring using only biologic T N F - α inhibitors, if this approach did not provide the intended outcomes (when severity arises), stem cells are administered a few times in a pulsed manner as a cell replacement technique in addition to this anti T N F - α medicine. We have observed the combined therapeutic benefit of stem cell replacement with a T N F - α inhibitor from a mathematical point of view. The theoretical analysis and the numerical results revealed that stem cell transplantation, along with a T N F - α inhibitor, is a promising psoriasis treatment option moving forward.


Subject(s)
Mesenchymal Stem Cell Transplantation , Psoriasis , Psoriasis/therapy , Psoriasis/drug therapy , Humans , Mesenchymal Stem Cell Transplantation/methods , Mesenchymal Stem Cells , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Models, Theoretical , Keratinocytes/drug effects , T-Lymphocytes/immunology , T-Lymphocytes/drug effects , Dendritic Cells/immunology
18.
Stem Cell Res Ther ; 15(1): 312, 2024 Sep 19.
Article in English | MEDLINE | ID: mdl-39300523

ABSTRACT

Chronic obstructive pulmonary disease(COPD) is an inflammatory disease characterized by the progressive and irreversible structural and functional damage of lung tissue. Although COPD is a significant global disease burden, the available treatments only ameliorate the symptoms, but cannot reverse lung damage. Researchers in regenerative medicine have examined the use of stem cell transplantation for treatment of COPD and other diseases because these cells have the potential for unlimited self-renewal and the ability to undergo directed differentiation. Stem cells are typically classified as embryonic stem cells, induced pluripotent stem cells, and adult stem cells (which includes mesenchymal stem cells [MSCs]), each with its own advantages and disadvantages regarding applications in regenerative medicine. Although the heterogeneity and susceptibility to senescence of MSCs make them require careful consideration for clinical applications. However, the low tumourigenicity and minimal ethical concerns of MSCs make them appear to be excellent candidates. This review summarizes the characteristics of various stem cell types and describes their therapeutic potential in the treatment of COPD, with a particular emphasis on MSCs. We aim to facilitate subsequent in-depth research and preclinical applications of MSCs by providing a comprehensive overview.


Subject(s)
Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells , Pulmonary Disease, Chronic Obstructive , Humans , Pulmonary Disease, Chronic Obstructive/therapy , Mesenchymal Stem Cell Transplantation/methods , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Animals , Cell Differentiation , Regenerative Medicine/methods
19.
Stem Cell Res Ther ; 15(1): 301, 2024 Sep 15.
Article in English | MEDLINE | ID: mdl-39278909

ABSTRACT

BACKGROUND: Amyotrophic lateral sclerosis (ALS) is a fatal and rapidly progressive motoneuron degenerative disorder. There are still no drugs capable of slowing disease evolution or improving life quality of ALS patients. Thus, autologous stem cell therapy has emerged as an alternative treatment regime to be investigated in clinical ALS. METHOD: Using Proteomics and Protein-Protein Interaction Network analyses combined with bioinformatics, the possible cellular mechanisms and molecular targets related to mesenchymal stem cells (MSCs, 1 × 106 cells/kg, intrathecally in the lumbar region of the spine) were investigated in cerebrospinal fluid (CSF) of ALS patients who received intrathecal infusions of autologous bone marrow-derived MSCs thirty days after cell therapy. Data are available via ProteomeXchange with identifier PXD053129. RESULTS: Proteomics revealed 220 deregulated proteins in CSF of ALS subjects treated with MSCs compared to CSF collected from the same patients prior to MSCs infusion. Bioinformatics enriched analyses highlighted events of Extracellular matrix and Cell adhesion molecules as well as related key targets APOA1, APOE, APP, C4A, C5, FGA, FGB, FGG and PLG in the CSF of cell treated ALS subjects. CONCLUSIONS: Extracellular matrix and cell adhesion molecules as well as their related highlighted components have emerged as key targets of autologous MSCs in CSF of ALS patients. TRIAL REGISTRATION: Clinicaltrial.gov identifier NCT0291768. Registered 28 September 2016.


Subject(s)
Amyotrophic Lateral Sclerosis , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells , Proteomics , Transplantation, Autologous , Humans , Amyotrophic Lateral Sclerosis/cerebrospinal fluid , Amyotrophic Lateral Sclerosis/therapy , Amyotrophic Lateral Sclerosis/metabolism , Mesenchymal Stem Cells/metabolism , Proteomics/methods , Mesenchymal Stem Cell Transplantation/methods , Male , Female , Middle Aged , Apolipoproteins E/metabolism , Apolipoproteins E/genetics , Apolipoproteins E/cerebrospinal fluid , Aged , Apolipoprotein A-I/cerebrospinal fluid , Apolipoprotein A-I/metabolism , Adult , Bone Marrow Cells/metabolism , Protein Interaction Maps
20.
Biomed Pharmacother ; 179: 117386, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39241570

ABSTRACT

Mesenchymal stem cell-derived exosomes(MSCs-Exos) offer promising therapeutic potential for a wide range of tissues and organs such as bone/cartilage, nerves, skin, fat, and endocrine organs. In comparison to the application of mesenchymal stem cells (MSCs), MSCs-Exos address critical challenges related to rejection reactions and ethical concerns, positioning themselves as a promising cell-free therapy. As exosomes are extracellular vesicles, their effective delivery necessitates the use of carriers. Consequently, the selection of hydrogel materials as scaffolds for exosome delivery has become a focal point of contemporary research. The diversity of hydrogel scaffolds, which can take various forms such as injectable types, dressings, microneedles, and capsules, leads to differing choices among researchers for treating diseases within the same domain. This variability in hydrogel materials poses challenges for the translation of findings into clinical practice. The review highlights the potential of hydrogel-loaded exosomes in different fields and introduces the advantages and disadvantages of different forms of hydrogel applications. It aims to provide a multifunctional and highly recognized hydrogel scaffold option for tissue regeneration at specific sites, improve clinical translation efficiency, and benefit the majority of patients.


Subject(s)
Exosomes , Hydrogels , Mesenchymal Stem Cells , Tissue Scaffolds , Humans , Hydrogels/chemistry , Tissue Scaffolds/chemistry , Animals , Mesenchymal Stem Cell Transplantation/methods , Tissue Engineering/methods
SELECTION OF CITATIONS
SEARCH DETAIL