Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters








Publication year range
1.
Life Sci ; 351: 122761, 2024 Aug 15.
Article in English | MEDLINE | ID: mdl-38866216

ABSTRACT

Mesenchymal Stromal Cells (MSCs) offer tremendous potential for the treatment of various diseases and their healing properties have been explored in hundreds of clinical trials. These trails primarily focus on immunological and neurological disorders, as well as regenerative medicine. Adipose tissue is a rich source of mesenchymal stromal cells and methods to obtain and culture adipose-derived MSCs (AD-MSCs) have been well established. Promising results from pre-clinical testing of AD-MSCs activity prompted clinical trials that further led to the approval of AD-MSCs for the treatment of complex perianal fistulas in Crohn's disease and subcutaneous tissue defects. However, AD-MSC heterogeneity along with various manufacturing protocols or different strategies to boost their activity create the need for standardized quality control procedures and safety assessment of the intended cell product. High-resolution transcriptomic methods have been recently gaining attention, as they deliver insight into gene expression profiles of individual cells, helping to deconstruct cellular hierarchy and differentiation trajectories, and to understand cell-cell interactions within tissues. This article presents a comprehensive overview of completed clinical trials evaluating the safety and efficacy of AD-MSC treatment, together with current single-cell studies of human AD-MSC. Furthermore, our work emphasizes the increasing significance of single-cell research in elucidating the mechanisms of cellular action and predicting their therapeutic effects.


Subject(s)
Adipose Tissue , Clinical Trials as Topic , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells , Humans , Mesenchymal Stem Cells/cytology , Adipose Tissue/cytology , Mesenchymal Stem Cell Transplantation/methods , Single-Cell Analysis/methods , Cell Differentiation , Animals , Regenerative Medicine/methods
2.
J Control Release ; 364: 718-733, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37944669

ABSTRACT

Adipose-derived mesenchymal stem cell-derived small extracellular vesicles (Ad-MSC-sEVs/AMEs) combined with scaffold materials are used in tissue-engineered bladders; however, the lack of retention leads to limited distribution of AMEs in the scaffold areas and low bioavailability of AMEs after bladder reconstruction. To improve retention of AMEs, we developed a novel strategy that modifies the surface charge of the bladder acellular matrix (BAM) via oxidative self-polymerization of dopamine-reducing graphene oxide (GO) and AMEs using ε-polylysine-polyethylene-distearyl phosphatidylethanolamine (PPD). We evaluated two BAM surface modification methods and evaluated the biocompatibility of materials and PPD and electrostatic adherence effects between PPD-modified AMEs and rGO-PDA/BAM in vivo and in vitro. Surface modification increased retention of AMEs, enhanced regeneration of bladder structures, and increased electrical conductivity of rGO-PDA/BAM, thereby improving bladder function recovery. RNA-sequencing revealed 543 miRNAs in human AMEs and 514 miRNAs in rat AMEs. A Venn diagram was used to show target genes of miRNA with the highest proportion predicted by the four databases; related biological processes and pathways were predicted by KEGG and GO analyses. We report a strategy for improving bioavailability of AMEs for bladder reconstruction and reveal that enriched miR-21-5p targets PIK3R1 and activates the PI3K/Akt pathway to promote cell proliferation and migration.


Subject(s)
Extracellular Vesicles , Mesenchymal Stem Cells , MicroRNAs , Rats , Humans , Animals , Tissue Engineering/methods , Urinary Bladder/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Extracellular Matrix/metabolism , MicroRNAs/metabolism
3.
Tissue Cell ; 85: 102224, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37783027

ABSTRACT

The conditioned medium of mesenchymal stem cells (MSCs) has controversial roles in cancer, either promoting or suppressing tumor growth. Our research on the results of adipose tissue-derived MSC (AD-MSC)-conditioned media on U87 glioma cells was motivated by the disputed role of mesenchymal stem cells (MSCs) in cancer, which may either promote or inhibit tumor growth. Using flow cytometry, AD-MSCs were identified, verified, and their conditioned media was used to treat U87 cells. Through RT-qPCR, scratch assay, and apoptosis analysis, we evaluated gene expression (SOX4, H19, and CCAT1), cell migration, and apoptosis in U87 cells.The conditioned media greatly increased the expression of SOX4 and H19, but not CCAT1. Although there were few differences in migration and apoptosis, both were slightly increased in the treated group.These outcomes have drawn attention to the complexity of the interactions between MSCs and glioma cells. This complexity requires further research to identify the specific mechanisms governing MSC-mediated impacts on the development of glioblastoma multiforme (GBM).


Subject(s)
Glioma , Mesenchymal Stem Cells , Humans , Culture Media, Conditioned/pharmacology , Culture Media, Conditioned/metabolism , Cell Line, Tumor , Glioma/genetics , Glioma/metabolism , Glioma/pathology , Mesenchymal Stem Cells/metabolism , Apoptosis/genetics , Cell Movement/genetics , Gene Expression , Cell Proliferation/genetics , SOXC Transcription Factors/metabolism
4.
Neurotoxicology ; 97: 1-11, 2023 Jul.
Article in English | MEDLINE | ID: mdl-37146888

ABSTRACT

Parkinson's disease (PD) is characterized by the loss of dopaminergic neurons and sustained neuroinflammation due to microglial activation. Adipose tissue-derived mesenchymal stem cells (AD-MSCs) secrete neuroprotective factors to prevent neuronal damage. Furthermore, Zn regulates stem cell proliferation and differentiation and has immunomodulatory functions. Our in vivo study aimed to investigate whether Zn affects the activities of AD-MSCs in the MPTP-induced mouse model. Male C57BL/6 mice were randomly divided into six groups (n = 6): Control, Zn, PD, PD+Zn, PD+ (AD-MSC), PD+ (AD-MSC)+Zn. MPTP toxin (20 mg/kg) was dissolved in saline and intraperitoneally injected into experimental groups for two days with 12 h intervals. On the 3rd day, AD-MSCs were given to the right lateral ventricle of the PD+ (AD-MSC) and PD+ (AD-MSC)+Zn groups by stereotaxic surgery. Then, ZnSO4H2O was administered intraperitoneally for 4 days at 2 mg/kg. Seven days post MPTP injection, the motor activities of the mouse were evaluated. Then immunohistochemical analyzes were performed in SNpc. Our results showed that motor activity was lower in Group PD. AD-MSC and Zn administration have improved this impairment. MPTP caused a decrease in TH and BDNF expressions in dopaminergic neurons in Group PD. However, TH and BDNF expressions were more intense in the other groups. MCP-1, TGF-ß, and IL-10 expressions increased in administered groups compared to the Group PD. The present study indicates that Zn's individual and combined administration with AD-MSCs reduces neuronal damage in the MPTP-induced mouse model. In addition, anti-inflammatory responses that emerge with Zn and AD-MSCs may have a neuroprotective effect.


Subject(s)
Mesenchymal Stem Cells , Neuroprotective Agents , Parkinson Disease , Male , Animals , Mice , Parkinson Disease/therapy , Parkinson Disease/metabolism , Zinc/metabolism , Brain-Derived Neurotrophic Factor/metabolism , Mice, Inbred C57BL , Dopaminergic Neurons , Mesenchymal Stem Cells/metabolism , Disease Models, Animal , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/pharmacology , Neuroprotective Agents/pharmacology , Neuroprotective Agents/metabolism
5.
Biomolecules ; 11(7)2021 06 22.
Article in English | MEDLINE | ID: mdl-34206204

ABSTRACT

Adipose-derived stem cells (ASCs) have been increasingly used as a versatile source of mesenchymal stem cells (MSCs) for diverse clinical investigations. However, their applications often become complicated due to heterogeneity arising from various factors. Cellular heterogeneity can occur due to: (i) nomenclature and criteria for definition; (ii) adipose tissue depots (e.g., subcutaneous fat, visceral fat) from which ASCs are isolated; (iii) donor and inter-subject variation (age, body mass index, gender, and disease state); (iv) species difference; and (v) study design (in vivo versus in vitro) and tools used (e.g., antibody isolation and culture conditions). There are also actual differences in resident cell types that exhibit ASC/MSC characteristics. Multilineage-differentiating stress-enduring (Muse) cells and dedifferentiated fat (DFAT) cells have been reported as an alternative or derivative source of ASCs for application in regenerative medicine. In this review, we discuss these factors that contribute to the heterogeneity of human ASCs in detail, and what should be taken into consideration for overcoming challenges associated with such heterogeneity in the clinical use of ASCs. Attempts to understand, define, and standardize cellular heterogeneity are important in supporting therapeutic strategies and regulatory considerations for the use of ASCs.


Subject(s)
Adipose Tissue/metabolism , Cell Differentiation , Mesenchymal Stem Cells/metabolism , Regenerative Medicine , Animals , Humans , Organ Specificity
6.
Acta cir. bras ; 36(2): e360206, 2021. tab, graf
Article in English | LILACS | ID: biblio-1152698

ABSTRACT

ABSTRACT Purpose To evaluate clinical outcome following minimally invasive plate osteosynthesis (MIPO) associated with percutaneous transplantation of allogeneic adipose-derived mesenchymal stem cells (AD-MSC) at the tibial fracture site in dogs. Methods Thirty-six dogs presenting with nonarticular complete tibial fracture were included in this study. All fractures were treated by the same MIPO technique. The animals were divided in group 1 (n = 20) received a percutaneous application of 3 × 106 AD-MSC at the fracture site and group 2 (n = 16) did not receive any adjuvant treatment. Postoperative radiographic examinations were made at 15, 30, 60, 90 and 120 days. Results Fifty-eight percent of the patients were classified as skeletally immature. The median weight of the animals was 18.8 kg. The mean radiographic union time differed statistically between the AD-MSC group (28.5 days) and the control group (70.3 days). Sixty percent of dogs in group 1 and 56.25% of the group 2 were considered immature. Conclusions The use of allogeneic AD-MSC cell therapy and MIPO is a safe, viable and effective technique for promoting bone healing in nonarticular tibial fractures in dogs.


Subject(s)
Humans , Animals , Dogs , Hematopoietic Stem Cell Transplantation , Mesenchymal Stem Cells , Tibia/surgery , Bone Plates , Fracture Fixation, Internal
7.
Stem Cells Cloning ; 13: 117-136, 2020.
Article in English | MEDLINE | ID: mdl-33364786

ABSTRACT

INTRODUCTION: Osteoarthritis causes a progressive deterioration to the protective cartilage between the joints leading to chronic pain and disability. This review focuses on the intrinsic potential of MSCs to stabilize and repair the cartilage tissue of the knee joint in knee osteoarthritis (KOA) patients. METHODS: An online search through the PubMed database was conducted, limiting the search to the English language and human clinical trials within the past 5 years. Twenty-one clinical trials passed the inclusion criteria. Combined, those trials involved the participation of 589 patients where the progress of the treatments was monitored between a 4-month to 7-years period. The cartilage volume and defects were observed through an MRI to provide an objective assessment. While the pain and knee function were monitored using KOOS, VAS, and WOMAC scoring scales providing a subjective assessment. RESULTS: MRI scans obtained from clinical trials demonstrate a slowed progression of cartilage degeneration and early signs of cartilage regeneration in KOA patients at the 12-month follow-up period. No major adverse effects were observed post-intervention. The overall KOOS, WOMAC, and VAS scores in patients receiving MSC treatment were reduced, suggesting subjective improvements in knee function and pain reduction when compared to patients in the placebo group. CONCLUSION: The use of MSC therapy is a valid form of treatment for KOA as it targets the disease itself rather than the symptoms. We found MSC therapy in KOA patients to be safe, effective, and feasible in its execution.

8.
Neurosurg Focus ; 46(3): E10, 2019 03 01.
Article in English | MEDLINE | ID: mdl-30835679

ABSTRACT

Recent advances in stem cell biology present significant opportunities to advance clinical applications of stem cell-based therapies for spinal cord injury (SCI). In this review, the authors critically analyze the basic science and translational evidence that supports the use of various stem cell sources, including induced pluripotent stem cells, oligodendrocyte precursor cells, and mesenchymal stem cells. They subsequently explore recent advances in stem cell biology and discuss ongoing clinical translation efforts, including combinatorial strategies utilizing scaffolds, biogels, and growth factors to augment stem cell survival, function, and engraftment. Finally, the authors discuss the evolution of stem cell therapies for SCI by providing an overview of completed (n = 18) and ongoing (n = 9) clinical trials.


Subject(s)
Induced Pluripotent Stem Cells/transplantation , Mesenchymal Stem Cell Transplantation , Oligodendrocyte Precursor Cells/transplantation , Spinal Cord Injuries/therapy , Adipose Tissue/cytology , Bone Marrow Cells , Clinical Trials as Topic , Embryonic Stem Cells/transplantation , Forecasting , Graft Survival/drug effects , Humans , Intercellular Signaling Peptides and Proteins/therapeutic use , Tissue Scaffolds , Umbilical Cord/cytology
9.
Mol Biol Rep ; 46(1): 1157-1165, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30628022

ABSTRACT

The immunomodulatory property of mesenchymal stem cells (MSCs) has been previously reported. Still it is unclear if this property can be affected by the cell origin and cell quality. Using primary MSCs expanded from bone marrow (BM-MSCs) and adipose tissue (AD-MSCs) of mice, we investigated whether the immunomodulatory property of MSCs varied with cell origin and cell quality (early- vs. late-passaged BM-MSCs). BM-MSCs (p1) and AD-MSCs (p1) had a typical spindle shape, but morphological changes were observed in late-passaged BM-MSCs (p6). A pathway-focused array showed that the expression of chemokine/cytokine genes varied with different cell origins and qualities. By co-culturing with spleen mononuclear cells (MNC) for 3 days, the expression of CD4 was suppressed by all types of MSCs. By contrast, the expression of CD8 was suppressed by BM-MSCs and increased by AD-MSCs. The expression ratio of CD206 to CD86 was at a comparable level after co-culture with AD-MSCs and BM-MSCs, but was lower with late-passaged BM-MSCs. AD-MSCs highly induced the release of IL6, IL-10 and TGF-ß in culture medium. Compared with early-passaged BM-MSCs (p1), late-passaged BM-MSCs (p6) released less TGF-ß. Our data suggests that the immunomodulatory properties of MSCs vary with cell origin and cell quality and that BM-MSCs of good quality are likely the optimal source of immunomodulation.


Subject(s)
Immunomodulation/immunology , Mesenchymal Stem Cells/immunology , Mesenchymal Stem Cells/physiology , Adipose Tissue/cytology , Animals , Bone Marrow Cells/cytology , Cell Differentiation/physiology , Cell Proliferation , Cells, Cultured , Coculture Techniques , Cytokines/metabolism , Gene Expression/genetics , Male , Mice , Mice, Inbred C57BL
10.
Cell Transplant ; 27(7): 1126-1139, 2018 07.
Article in English | MEDLINE | ID: mdl-29947256

ABSTRACT

The use of mesenchymal stromal cell (MSC) transplantation to repair the injured spinal cord has shown consistent benefits in preclinical models. However, the low survival rate of grafted MSC is one of the most important problems. In the injured spinal cord, transplanted cells are exposed to hypoxic conditions and exposed to nutritional deficiency caused by poor vascular supply. Also, the transplanted MSCs face cytotoxic stressors that cause cell death. The aim of this study was to compare adipose-derived MSCs (AD-MSCs) and bone marrow-derived MSCs (BM-MSCs) isolated from individual C57BL6/J mice in relation to: (i) cellular characteristics, (ii) tolerance to hypoxia, oxidative stress and serum-free conditions, and (iii) cellular survival rates after transplantation. AD-MSCs and BM-MSCs exhibited a similar cell surface marker profile, but expressed different levels of growth factors and cytokines. To research their relative stress tolerance, both types of stromal cells were incubated at 20.5% O2 or 1.0% O2 for 7 days. Results showed that AD-MSCs were more proliferative with greater culture viability under these hypoxic conditions than BM-MSCs. The MSCs were also incubated under H2O2-induced oxidative stress and in serum-free culture medium to induce stress. AD-MSCs were better able to tolerate these stress conditions than BM-MSCs; similarly when transplanted into the spinal cord injury region in vivo, AD-MSCs demonstrated a higher survival rate post transplantation Furthermore, this increased AD-MSC survival post transplantation was associated with preservation of axons and enhanced vascularization, as delineated by increases in anti-gamma isotype of protein kinase C and CD31 immunoreactivity, compared with the BM-MSC transplanted group. Hence, our results indicate that AD-MSCs are an attractive alternative to BM-MSCs for the treatment of severe spinal cord injury. However, it should be noted that the motor function was equally improved following moderate spinal cord injury in both groups, but with no significant improvement seen unfortunately following severe spinal cord injury in either group.


Subject(s)
Adipose Tissue/cytology , Bone Marrow Cells/cytology , Mesenchymal Stem Cell Transplantation/methods , Mesenchymal Stem Cells/cytology , Spinal Cord Injuries/therapy , Animals , Axons/pathology , Cell Separation/methods , Cells, Cultured , Locomotion , Male , Mice, Inbred C57BL , Spinal Cord/pathology , Spinal Cord/physiopathology , Spinal Cord Injuries/pathology , Spinal Cord Injuries/physiopathology
11.
Stem Cell Rev Rep ; 13(5): 670-685, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28795363

ABSTRACT

Stroke, as the second most common cause of death, imposes a great financial burden on both the individual and society. Mesenchymal stem cells from rodents have demonstrated efficacy in experimental animal models of stroke due to enhanced neurological recovery. Since FGF1 (fibroblast growth factor 1) displays neuroprotective properties, for the first time, we investigated the effect of acute intravenous administration of FGF1 gene transfected adipose-derived mesenchymal stem cell (AD-MSCFGF1) on transient experimental ischemic stroke in rats. Stroke induction was made by transient middle cerebral artery occlusion (tMCAO). 2 × 106 AD-MSCFGF1 was administrated intravenously 30 min after carotid reperfusion. The ability of technetium99m-hexamethyl propylene amine oxime (99mTc-HMPAO)-labeled AD-MSCFGF1 to enter into ischemic brain was evaluated 2 h post injection. 24 h post operation, the neurological recovery (rotarod and Roger's tests), the infarct volume (2, 3, 5-triphenyltetrazolium chloride, TTC assay), apoptosis rate (TUNEL assay), and the expression of FGF1 protein (western blotting) in the ischemic hemisphere were assessed. The 99mTc-HMPAO-labeled AD-MSCFGF1 could enter into the ischemic brain. Ischemic hemisphere activity was significantly higher than that observed in the contralateral hemisphere (p = 0.002). The administration of AD-MSCFGF1 resulted in significant improvement of neurological function tests and increased density of FGF1 protein in the peri-infarct area, while the infarct volume and the apoptotic index were significantly decreased, in comparison to the other treated groups. In conclusion, acute intravenous administration of AD-MSCFGF1 can be a novel and promising candidate approach for the treatment of ischemic stroke.


Subject(s)
Fibroblast Growth Factor 1/genetics , Infarction, Middle Cerebral Artery/therapy , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/metabolism , Recovery of Function , Stroke/therapy , Adipocytes/cytology , Adipocytes/metabolism , Adipocytes/transplantation , Adipose Tissue/cytology , Adipose Tissue/metabolism , Animals , Cell Differentiation , Cerebrum/metabolism , Cerebrum/pathology , Disease Models, Animal , Fibroblast Growth Factor 1/metabolism , Gene Expression , Humans , Infarction, Middle Cerebral Artery/genetics , Infarction, Middle Cerebral Artery/metabolism , Infarction, Middle Cerebral Artery/pathology , Infusions, Intravenous , Male , Mesenchymal Stem Cells/cytology , Radiopharmaceuticals/administration & dosage , Rats , Rats, Wistar , Rotarod Performance Test , Stroke/genetics , Stroke/metabolism , Stroke/pathology , Technetium Tc 99m Exametazime/administration & dosage , Transgenes
12.
Int J Pharm ; 501(1-2): 75-86, 2016 Mar 30.
Article in English | MEDLINE | ID: mdl-26827918

ABSTRACT

Since mesenchymal stem cells (MSCs) can self-renew and differentiate into multiple cell types, the delivery of genes to this type of cell can be an important tool in the emerging field of tissue regeneration and engineering. However, development of more efficient and safe nonviral vectors for gene delivery to stem cells in particular still remains a great challenge. In this study, we describe a group of nonviral gene delivery vectors, conjugated PAMAM derivatives (PAMAM-H-R, PAMAM-H-K, and PAMAM-H-O), displaying affinity toward human adipose-derived mesenchymal stem cells (AD-MSCs). Transfection efficiency using pDNA encoding for luciferase (Luc) and enhanced green fluorescent protein (EGFP), and cytotoxicity assays were performed in human AD-MSCs. The results show that transfection efficiencies of conjugated PAMAM derivatives are improved significantly compared to native PAMAM dendrimer, and that among PAMAM derivatives, cytotoxicity of PAMAM-H-K and PAMAM-H-O were very low. Also, treatment of human AD-MSCs to polyplex formation in conjugated PAMAM derivatives, their cellular uptake and localization were analyzed by flow cytometry and confocal microscopy.


Subject(s)
Amino Acids, Basic/administration & dosage , DNA/administration & dosage , Dendrimers/administration & dosage , Gene Transfer Techniques , Mesenchymal Stem Cells/metabolism , Adipose Tissue/cytology , Amino Acids, Basic/chemistry , Amino Acids, Basic/pharmacology , Biological Transport , Cell Differentiation/drug effects , Cell Survival/drug effects , Cells, Cultured , DNA/chemistry , DNA/pharmacology , Dendrimers/chemistry , Dendrimers/pharmacology , Green Fluorescent Proteins/genetics , Humans , Luciferases/genetics , Mesenchymal Stem Cells/drug effects , Plasmids
13.
Methods ; 99: 69-80, 2016 Apr 15.
Article in English | MEDLINE | ID: mdl-26384579

ABSTRACT

Musculoskeletal disorders represent a major cause of disability and morbidity globally and result in enormous costs for health and social care systems. Development of cell-based therapies is rapidly proliferating in a number of disease areas, including musculoskeletal disorders. Novel biological therapies that can effectively treat joint and spine degeneration are high priorities in regenerative medicine. Mesenchymal stem cells (MSCs) isolated from bone marrow (BM-MSCs), adipose tissue (AD-MSCs) and umbilical cord (UC-MSCs) show considerable promise for use in cartilage and intervertebral disc (IVD) repair. This review article focuses on stem cell-based therapeutics for cartilage and IVD repair in the context of the rising global burden of musculoskeletal disorders. We discuss the biology MSCs and chondroprogenitor cells and specifically focus on umbilical cord/Wharton's jelly derived MSCs and examine their potential for regenerative applications. We also summarize key components of the molecular machinery and signaling pathways responsible for the control of chondrogenesis and explore biomimetic scaffolds and biomaterials for articular cartilage and IVD regeneration. This review explores the exciting opportunities afforded by MSCs and discusses the challenges associated with cartilage and IVD repair and regeneration. There are still many technical challenges associated with isolating, expanding, differentiating, and pre-conditioning MSCs for subsequent implantation into degenerate joints and the spine. However, the prospect of combining biomaterials and cell-based therapies that incorporate chondrocytes, chondroprogenitors and MSCs leads to the optimistic view that interdisciplinary approaches will lead to significant breakthroughs in regenerating musculoskeletal tissues, such as the joint and the spine in the near future.


Subject(s)
Cartilage, Articular/physiology , Intervertebral Disc/physiology , Mesenchymal Stem Cells/physiology , Animals , Humans , Mesenchymal Stem Cell Transplantation , Regeneration , Regenerative Medicine , Signal Transduction , Tissue Engineering , Wharton Jelly/cytology
14.
J Vasc Interv Radiol ; 24(12): 1871-8, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24144538

ABSTRACT

PURPOSE: To investigate the safety and effectiveness of a novel endovascular approach for therapeutic cell delivery using a balloon occlusion catheter in a large animal model of liver fibrosis. MATERIALS AND METHODS: Transcatheter arterial embolization with ethiodized oil (Ethiodol) and ethanol was used to induce liver damage in 11 pigs. Mesenchymal stem cells (MSCs) were harvested from adipose tissue and engineered to express green fluorescent protein (GFP). A balloon occlusion catheter was positioned in the bilateral first-order portal vein branches 2 weeks after embolization to allow intraportal application of MSCs in six experimental animals. MSCs were allowed to dwell for 10 minutes using prolonged balloon inflation. Five control animals received a sham injection of normal saline in a similar fashion. Hepatic venous pressure gradient (HVPG) was measured immediately before necropsy. Specimens from all accessible lobes were obtained with ultrasound-guided percutaneous 18-gauge biopsy 2 hours after cell application. All animals were euthanized within 4 weeks. Fluorescent microscopy was used to assess the presence and distribution of cells. RESULTS: Liver injury and fibrosis were successfully induced in all animals. MSCs (6-10 × 10(7)) were successfully delivered into the portal vein in the six experimental animals. Cell application was not associated with vascular complications. HVPG showed no instances of portal hypertension. GFP-expressing MSCs were visualized in biopsy specimens and were distributed primarily within the sinusoidal spaces; however, 4 weeks after implantation, MSCs could not be identified in histologic specimens. CONCLUSIONS: A percutaneous endovascular approach for cell delivery using a balloon occlusion catheter proved safe for intraportal MSC application in a large animal model of liver fibrosis.


Subject(s)
Adipose Tissue/cytology , Balloon Occlusion/instrumentation , Endovascular Procedures/instrumentation , Liver Cirrhosis, Experimental/therapy , Liver/pathology , Mesenchymal Stem Cell Transplantation/instrumentation , Vascular Access Devices , Animals , Biomarkers/metabolism , Biopsy , Cell Tracking , Cells, Cultured , Equipment Design , Ethanol , Ethiodized Oil , Green Fluorescent Proteins/biosynthesis , Green Fluorescent Proteins/genetics , Hepatic Veins/physiopathology , Liver/metabolism , Liver Cirrhosis, Experimental/chemically induced , Liver Cirrhosis, Experimental/metabolism , Liver Cirrhosis, Experimental/pathology , Liver Cirrhosis, Experimental/physiopathology , Male , Mesenchymal Stem Cells/metabolism , Sus scrofa , Time Factors , Transfection , Venous Pressure
15.
Int. j. morphol ; 29(3): 1012-1017, Sept. 2011. ilus
Article in English | LILACS | ID: lil-608698

ABSTRACT

Recent studies revealed multipotent properties of fat tissue isolated mesenchymal stem cells. These cells are successfully used as therapeutic factor for many locomotive disorders, being even more effective than stem cells from bone marrow. Isolated and cultured, AD-MSCs were observed, photographed and measured to compare cells from two different species.


Estudios recientes han revelado propiedades pluripotentes del tejido graso aislado de células madre mesenquimales. Estas células se utilizan con éxito como factor terapéutico para muchos trastornos locomotores, siendo aún más eficaz que las células madre de médula ósea. Aisladas y cultivadas, AD-MSC se observaron, fotografiaron y midieron comparar células de dos especies diferentes.


Subject(s)
Dogs , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells , Mesenchymal Stem Cells/ultrastructure , Musculoskeletal Diseases/therapy , Musculoskeletal Diseases/veterinary , Orthopedic Procedures/methods
16.
Int J Stem Cells ; 4(2): 99-104, 2011 Nov.
Article in English | MEDLINE | ID: mdl-24298341

ABSTRACT

BACKGROUND AND OBJECTIVES: AD-MSCs (adipose-derived mesenchymal stem cells), as an easy obtainable population of multipotent cells, have been successfully applied in many diseases in animals. Having very similar properties and morphology to these collected from bone marrow, are very attractive object for regenerative medicine. CONCLUSIONS: Ovine subcutaneous fat from many regions of the body is rich source of stem cells, which could be used in locomotive system disorders experimental therapy on sheep models.

SELECTION OF CITATIONS
SEARCH DETAIL