Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 86
Filter
Add more filters

Country/Region as subject
Publication year range
1.
J Mater Sci Mater Med ; 30(9): 99, 2019 Aug 27.
Article in English | MEDLINE | ID: mdl-31455977

ABSTRACT

Adipose-derived mesenchymal stem cells (ASCs) accelerate the osteointegration of bone grafts and improve the efficiency in the formation of uniform bone tissue, providing a practical and clinically attractive approach in bone tissue regeneration. In this work, the effect of nanofibrous biomimetic matrices composed of poly(ε-caprolactone) (PCL), nanometric hydroxyapatite (nHA) particles and 14-3-3 protein isoform epsilon on the initial stages of human ASCs (hASCs) osteogenic differentiation was investigated. The cells were characterized by flow cytometry and induction to differentiation to adipogenic and osteogenic lineages. The isolated hASCs were induced to differentiate to osteoblasts over all scaffolds, and adhesion and viability of the hASCs were found to be similar. However, the activity of alkaline phosphatase (ALP) as early osteogenic marker in the PCL-nHA/protein scaffold was four times higher than in PCL-nHA and more than five times than the measured in neat PCL.


Subject(s)
14-3-3 Proteins , Durapatite , Mesenchymal Stem Cells/drug effects , Osteogenesis/drug effects , Polyesters , Tissue Scaffolds/chemistry , 14-3-3 Proteins/chemistry , 14-3-3 Proteins/pharmacology , Cell Differentiation/drug effects , Cell Differentiation/physiology , Cells, Cultured , Durapatite/chemistry , Durapatite/pharmacology , Electroplating/methods , Humans , Materials Testing , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/physiology , Nanofibers/chemistry , Osteoblasts/drug effects , Osteoblasts/physiology , Osteogenesis/physiology , Polyesters/chemistry , Polyesters/pharmacology , Subcutaneous Fat, Abdominal/cytology , Surface Properties/drug effects , Tissue Engineering/methods
2.
Aesthet Surg J ; 39(2): 220-232, 2019 01 17.
Article in English | MEDLINE | ID: mdl-29846494

ABSTRACT

Background: Adipose-derived stem cells (ASCs) assisted lipotransfer have been considered to facilitate the survival of fat grafts. However, emerging evidence of insufficient vascularization is another obstacle for fat graft survival in cell-assisted lipotransfer. Objectives: This study evaluated if endothelial phenotype ASCs with fat lipoaspirate improves survival and neovascularization in fat transplantation. Methods: ASCs were isolated from human periumbilical fat tissue and cultured in endothelial growth medium for 2 weeks. Fat lipoaspirate was mixed with fresh adipose stroma vascular fraction (SVF), endothelial differentiated ASCs (EC/ASCs), and fat lipoaspirate alone. Three fat mixtures were subcutaneously injected into the adult male Sprague-Dawley rat's dorsum at 3 locations. At 8 weeks after transplantation, the grafted fat lipoaspirates were harvested, and the extracted fat was evaluated using photographic, survival weights measurements and histological examination. Neo-vascularization was quantified by immunofluorescence and real-time RT-PCR. Results: Grafts from the EC/ASC assisted group had a higher survival rate, morphologic integrity, and most uniform lipid droplets. They also revealed less inflammation and fibrosis with increased number of vessels by histological and immunofluorescence analysis. Quantitative RT-PCR analysis indicated that the expression levels of EC-specific markers of CD31 and vWF were higher in the EC/ASC group compared with in the control and fat with SVF transplants. Conclusions: These results indicated that co-implantation of fat lipoaspirate with ASCs differentiated toward an endothelial phenotype improves both survival and neovascularization of the transplanted fat lipoaspirate, which might provide benefits and represents a promising strategy for clinical application in autologous fat transplantation.


Subject(s)
Graft Survival/physiology , Neovascularization, Physiologic/physiology , Stem Cell Transplantation/methods , Subcutaneous Fat, Abdominal/transplantation , Adult , Animals , Cell Culture Techniques/methods , Cell Differentiation/physiology , Cell Survival/physiology , Cells, Cultured , Culture Media , Endothelial Cells/physiology , Female , Humans , Male , Middle Aged , Models, Animal , Rats , Rats, Sprague-Dawley , Subcutaneous Fat, Abdominal/cytology , Transplantation, Heterologous/methods
3.
Int J Obes (Lond) ; 41(10): 1564-1569, 2017 10.
Article in English | MEDLINE | ID: mdl-28630459

ABSTRACT

BACKGROUND/OBJECTIVE: Differences in subcutaneous abdominal adipose tissue (SAT) fat cell size and number (cellularity) are linked to insulin resistance. Men are generally more insulin resistant than women but it is unknown whether there is a gender dimorphism in SAT cellularity. The objective was to determine SAT cellularity and its relationship to insulin sensitivity in men and women. METHODS: In a cohort study performed at an outpatient academic clinic in Sweden, 798 women and 306 men were included. Estimated SAT mass (ESAT) was derived from measures of dual-energy X-ray absorptiometry and a formula. SAT biopsies were obtained to measure mean fat cell size; SAT adipocyte number was obtained by dividing ESAT with mean fat cell weight. Fat cell size was also compared with level of insulin sensitivity in vivo. RESULTS: Over the entire range of body mass index (BMI) both fat cell size and number correlated positively with ESAT in either sex. On average, fat cell size was larger in men than in women, which was driven by significantly larger fat cells in non-obese men compared with non-obese women; no gender effect on fat cell size was seen in obese subjects. For all subjects fat cell number was larger in women than men, which was driven by a gender effect among non-obese individuals (P<0.0001). The relationship between fat cell size and insulin resistance was significant in both genders (P<0.0001) but steeper in men than in women (F=19, P<0.0001). CONCLUSIONS: Although both fat cell size and number determine SAT mass, adipocyte number contributes more and size less in women than in men and this is most evident in non-obese subjects. Over the entire BMI range, fat cell size contributes stronger to insulin resistance in men.


Subject(s)
Adipocytes/cytology , Sex Characteristics , Subcutaneous Fat, Abdominal/cytology , Absorptiometry, Photon , Adolescent , Adult , Aged , Body Composition , Body Fat Distribution , Body Mass Index , Female , Humans , Insulin Resistance , Male , Middle Aged , Sweden , Young Adult
4.
Osteoarthritis Cartilage ; 25(7): 1161-1171, 2017 07.
Article in English | MEDLINE | ID: mdl-28153787

ABSTRACT

OBJECTIVE: To define if adipose mesenchymal stromal cell (ASC) treatment mediated switching of the pro-inflammatory profile of M1-like macrophages as a means to develop a tailored in vitro efficacy/potency test. DESIGN: We firstly performed immunohistochemical analysis of CD68, CD80 (M1-like) and CD206 (M2-like) macrophages in osteoarthritic (OA) synovial tissue. ASC were co-cultured in contact and in transwell with activated (GM-CSF + IFNγ)-M1 macrophages. We analyzed IL1ß, TNFα, IL6, MIP1α/CCL3, S100A8, S100A9, IL10, CD163 and CD206 by qRT-PCR or immunoassays. Prostaglandin E2 (PGE2) blocking experiments were performed using PGE2 receptor antagonist. RESULTS: In moderate grade OA synovium we did not always find a higher percentage of CD80 with respect to CD206. M1-like-activated macrophage factors IL1ß, TNFα, IL6, MIP1α/CCL3, S100A8 and S100A9 were down-modulated both in contact and in transwell by ASC. However, in both systems ASC induced the typical M2-like macrophage markers IL10, CD163 and CD206. Activated-M1-like macrophages pre-treated with PGE2 receptor antagonist failed to decrease secretion of TNFα, IL6 and to increase that of IL10, CD163 and CD206 when co-cultured with ASC confirming a PGE2 specific role. CONCLUSIONS: We demonstrated that ASC are responsible for the switching of activated-M1-like inflammatory macrophages to a M2-like phenotype, mainly through PGE2. This evidenced that activated-M1-like macrophages may represent a relevant cell model to test the efficacy/potency of ASC and suggests a specific role of ASC as important determinants in therapeutic dampening of synovial inflammation in OA.


Subject(s)
Adipocytes/drug effects , Dinoprostone/pharmacology , Macrophages/drug effects , Mesenchymal Stem Cells/drug effects , Oxytocics/pharmacology , Adult , Antigens, CD/metabolism , Case-Control Studies , Cell Differentiation/physiology , Cell Movement/drug effects , Cells, Cultured , Female , Humans , Leukocytes, Mononuclear/drug effects , Male , Middle Aged , Osteoarthritis/pathology , Subcutaneous Fat, Abdominal/cytology , Synovial Membrane/cytology , Synovial Membrane/drug effects
5.
BMC Vet Res ; 13(1): 166, 2017 Jun 08.
Article in English | MEDLINE | ID: mdl-28595579

ABSTRACT

BACKGROUND: Mesenchymal Stem Cells (MSCs) are a promising therapeutic tool in veterinary medicine. Currently the subcutaneous adipose tissue is the leading source of MSCs in dogs. MSCs derived from distinct fat depots have shown dissimilarities in their accessibility and therapeutic potential. The aims of our work were to determine the suitability of omental adipose tissue as a source of MSCs, according to sampling success, cell yield and paracrine properties of isolated cells, and compared to subcutaneous adipose tissue. RESULTS: While sampling success of omental adipose tissue was 100% (14 collections from14 donors) for subcutaneous adipose tissue it was 71% (10 collections from 14 donors). MSCs could be isolated from both sources. Cell yield was significantly higher for omental than for subcutaneous adipose tissue (38 ± 1 vs. 30 ± 1 CFU-F/g tissue, p < 0.0001). No differences were observed between sources regarding cell proliferation potential (73 ± 1 vs. 74 ± 1 CDPL) and cell senescence (at passage 10, both cultures presented enlarged cells with cytoplasmic vacuoles and cellular debris). Omental- and subcutaneous-derived MSCs expressed at the same level bFGF, PDGF, HGF, VEGF, ANG1 and IL-10. Irrespective of the source, isolated MSCs induced proliferation, migration and vascularization of target cells, and inhibited the activation of T lymphocytes. CONCLUSION: Compared to subcutaneous adipose tissue, omental adipose tissue is a more suitable source of MSCs in dogs. Since it can be procured from donors with any body condition, its collection procedure is always feasible, its cell yield is high and the MSCs isolated from it have desirable differentiation and paracrine potentials.


Subject(s)
Adipose Tissue/cytology , Cell Separation/veterinary , Dogs/anatomy & histology , Mesenchymal Stem Cells , Omentum/cytology , Animals , Cell Proliferation , Cell Separation/methods , Endothelium, Vascular/cytology , Female , Mesenchymal Stem Cells/immunology , Subcutaneous Fat, Abdominal/cytology
6.
Ann Plast Surg ; 78(1): 83-90, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27941495

ABSTRACT

BACKGROUND: The present study was conducted to compare 2 purification methods for isolation of human adipose-derived stromal vascular fraction or stem cells (ADSCs) based on red blood cell (RBC) lysis with 155 mM ammonium chloride (NH4Cl) and hypotonic sodium chloride (NaCl) solution, and try to develop a safe, convenient, and cost-effective purification method for clinical applications. METHODS: Adipose-derived stem cells and RBC were harvested from the fatty and fluid portions of liposuction aspirates, respectively. The suitable concentration of hypotonic NaCl solution on RBC lysis for purification of ADSCs was developed by RBC osmotic fragility test and flow cytometry analysis. The effects of 155 mM NH4Cl or 0.3% NaCl solution on ADSCs proliferation and RBC lysis efficiency were examined by 3-(4, 5-dimethyl-2-thiazolyl)-2, 5-diphenyl-2H-tetrazolium bromide assay and lysis efficiency test, respectively. In addition, the adipogenic and osteogenic capabilities, phenotype and genetic stability of ADSCs were evaluated by oil red staining, alkaline phosphatase activity measurement, flow cytometry, and karyotype analysis, respectively. RESULTS: Sodium chloride solution in 0.3% concentration effectively removed RBCs and did not influence the survival of ADSCs in the 10-minute incubation time. The lysis efficiency did not differ significantly between 0.3% NaCl and 155 mM NH4Cl. Moreover, the adipogenic and osteogenic capabilities, surface marker expression and karyotype of the ADSCs were not affected by lysis solutions or by lysis per se. However, the proliferation capacity in the 0.3% NaCl group was superior to that in 155 mM NH4Cl group. CONCLUSIONS: Our data suggest that 0.3% NaCl solution is useful for isolating ADSCs from liposuction aspirate for clinical applications with safety, convenience, and cost-effect.


Subject(s)
Ammonium Chloride , Cell Separation/methods , Hemolysis , Mesenchymal Stem Cells , Sodium Chloride , Subcutaneous Fat, Abdominal/cytology , Adult , Cell Proliferation , Female , Humans , Hypotonic Solutions , Lipectomy , Male , Mesenchymal Stem Cells/physiology , Middle Aged , Subcutaneous Fat, Abdominal/surgery
7.
J Mammary Gland Biol Neoplasia ; 21(1-2): 69-76, 2016 06.
Article in English | MEDLINE | ID: mdl-27059487

ABSTRACT

Exposure to sex steroids increases the risk of breast cancer but the exact mechanisms are yet to be elucidated. Events in the microenvironment are important for carcinogenesis. Diet containing phytoestrogens can affect the breast microenvironment and alter the risk of breast cancer. It has previously been shown that estrogen regulates extracellular levels of leptin, adiponectin, and VEGF in normal breast tissue in vivo. Whether these proteins correlate in breast tissue in vivo or if diet addition of flaxseed, a major source of phytoestrogens in Western diets, alters adipokine levels in breast tissue are unknown. We used microdialysis to sample proteins of normal human breast tissue and abdominal subcutaneous fat in situ in 34 pre-and postmenopausal women. In vitro, co-culture of breast cancer cells and primary human adipocytes was used. In vivo, in normal breast tissue, a significant positive correlation between VEGF and leptin was detected. No correlations were found in fat tissue. Co-culture of adipocytes and breast cancer cells per se increased the secretion of VEGF and leptin and enhanced the effects of estradiol compared to culture of either cell type alone. In vitro, inhibition of VEGF diminished the release of leptin while inhibition of leptin had no influence on VEGF secretion. The levels of leptin decreased and adiponectin increased after a dietary addition of 25 g of flaxseed/day for one menstrual cycle. We conclude that VEGF and leptin correlate significantly in normal human breast tissue in vivo and that dietary addition of flaxseed affect adipokine levels in the breast.


Subject(s)
Adipokines/metabolism , Antineoplastic Agents, Phytogenic/metabolism , Breast Neoplasms/metabolism , Breast/metabolism , Flax/chemistry , Phytoestrogens/metabolism , Vascular Endothelial Growth Factors/metabolism , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Adenocarcinoma/prevention & control , Adipokines/agonists , Adipokines/antagonists & inhibitors , Adult , Antineoplastic Agents, Phytogenic/therapeutic use , Breast/cytology , Breast/pathology , Breast Neoplasms/pathology , Breast Neoplasms/prevention & control , Cells, Cultured , Coculture Techniques , Dietary Supplements , Female , Humans , MCF-7 Cells , Microdialysis , Middle Aged , Organ Specificity , Phytoestrogens/therapeutic use , Postmenopause/metabolism , Premenopause/metabolism , Seeds/chemistry , Subcutaneous Fat, Abdominal/cytology , Subcutaneous Fat, Abdominal/metabolism , Subcutaneous Fat, Abdominal/pathology , Vascular Endothelial Growth Factors/agonists , Vascular Endothelial Growth Factors/antagonists & inhibitors
8.
Ann Plast Surg ; 76(4): 446-52, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26954733

ABSTRACT

Wound healing involves a number of factors that results in the production of a "closed" wound. Studies have shown, in animal models, acceleration of wound healing with the addition of adipose-derived stromal cells (ADSC). The cause for the positive effect which these cells have on wound healing has not been elucidated. We have previously shown that addition of ADSC to the dermal equivalent in 3-dimensional skin cultures accelerates reepithelialization. We now demonstrate that conditioned media (CM) from cultured ADSC produced a similar rate of healing. This result suggests that a feedback from the 3-dimensional epithelial cultures to ADSC was not necessary to effect the accelerated reepithelialization. Mass spectrometry of CM from ADSC and primary human fibroblasts revealed differences in secretomes, some of which might have roles in the accelerating wound healing. Thus, the use of CM has provided some preliminary information on a possible mode of action.


Subject(s)
Mesenchymal Stem Cells/physiology , Re-Epithelialization/physiology , Subcutaneous Fat, Abdominal/cytology , Tissue Culture Techniques/methods , Culture Media, Conditioned , Fibroblasts/physiology , Humans , Skin , Wound Healing/physiology
9.
Ann Plast Surg ; 76 Suppl 1: S108-16, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26808740

ABSTRACT

BACKGROUND: Stromal vascular fraction (SVF) cells were used to increase the efficacy of a newly formed adipose tissue in a collagen gel in vitro. However, the outcome of the seeded cells in the collagen gel in vivo remains unknown. We traced the SVF cells in the host tissue and evaluated the efficacy of SVF for fat tissue engineering. METHODS: The aggregates implanted in the experimental and control groups were prepared by mixing SVF with the collagen gel and Dulbecco's modified Eagle medium with the collagen gel, respectively. The aggregates were implanted using a subcutaneous injection into the backs of immunodeficient mice. The aggregates were harvested 1, 2, 4, and 6 months after implantation; and 9 mice were euthanized each time. Macroscopic changes in the volume and wet weight of the aggregates were assessed. The formation of adipose tissue was studied using hematoxylin and eosin and Nile red staining. The origin and survival of adipocytes in the aggregates were examined through the immunostaining of leptin antibodies, DNA assay, and tracing of SVF cells by 1,1'-dioctadecyl-3,3,3',3'- tetramethylindocarbocyanine perchlorate labeling. RESULTS: The formation of adipose tissue was observed in all of the aggregates. Implanted human SVF cells remained in the experimental aggregates harvested after 1, 2, and 4 months but not after 6 months. At 6 months, viable adipocytes in both groups were of murine origin. Furthermore, at 6 months, the mean volume of the aggregate (P < 0.001) and the mean percentage of adipocytes (P < 0.001) were significantly higher in the experimental group than in the control group. CONCLUSIONS: Implanted SVF cells could not be traced in the aggregates harvested at 6 months but promoted the recruitment of host adipocytes to generate more adipose tissue in the experimental group than in the control group.


Subject(s)
Adipogenesis , Collagen , Mesenchymal Stem Cell Transplantation/methods , Subcutaneous Fat, Abdominal/cytology , Tissue Engineering/methods , Tissue Scaffolds , Animals , Apoptosis , Cell Survival , Female , Gels , Humans , Mice , Mice, Inbred BALB C , Stromal Cells , Subcutaneous Fat, Abdominal/physiology
10.
Ann Plast Surg ; 76(5): 569-75, 2016 May.
Article in English | MEDLINE | ID: mdl-27070348

ABSTRACT

BACKGROUND: Abdominal tissue enriched with adipose-derived stem cells (ASCs) is often used in cell-assisted lipotransfer procedures for breast reconstruction. However, as the tissue microenvironment and stem cell niche play important roles in defining the characteristics of the resident cells, it is hypothesized that the stem cell population present in the donor abdominal tissue has dissimilar properties as compared with the cells in the recipient breast tissue, which may ultimately affect the long-term success of the graft. METHODS: Adipose-derived stem cells were isolated from breast and abdominal fat tissues and characterized for mesenchymal-specific cell surface markers, and their population doubling, colony-forming capabilities, and proliferative properties were compared. The multilineage potential of both cell populations was also investigated. RESULTS: Adipose-derived stem cells from both tissue sites were found to possess similar marker expression and multilineage differentiation potential. However, breast fat-derived ASCs were observed to have a higher self-renewal capability and an unstable population doubling as compared with abdominal fat-derived ASCs. Gene expression studies revealed that the breast fat-derived ASCs were predisposed to the osteogenic lineage and the abdominal fat-derived ASCs to the adipogenic lineage. CONCLUSIONS: Cells derived from both fat tissues possess different characteristics in terms of their growth kinetics and predisposition to the osteolineages and adipolineages. In particular, ASCs from the abdominal tissue appear to contribute to adipose tissue turnover, whereas ASCs from breast tissue, if used for cell-assisted fat grafting, may potentially be responsible for complications in fat grafting, such as oil cysts, calcifications, fat necrosis, and tumors.


Subject(s)
Breast/cytology , Cell Differentiation/physiology , Cell Proliferation/physiology , Mesenchymal Stem Cells/physiology , Subcutaneous Fat/cytology , Adult , Biomarkers/metabolism , Cells, Cultured , Cellular Microenvironment/physiology , Colony-Forming Units Assay , Female , Humans , Middle Aged , Subcutaneous Fat, Abdominal/cytology
11.
Skin Res Technol ; 21(2): 192-200, 2015 May.
Article in English | MEDLINE | ID: mdl-25220194

ABSTRACT

BACKGROUND/AIMS: Cryolipolysis is a noninvasive method for the selective reduction of localized fat tissues. It has demonstrated efficacy in both clinical and preclinical trials; however, despite its popularity, its mechanisms of action and evaluation methods are not yet fully defined. The purpose of this study was to improved methods for cryolipolysis using a porcine model. METHODS: The abdomens of female PWG micro-pigs were treated with a cooling device (CRYOLIPO II(™)), and we examined the treatment effects using photography, three-dimensional photography, ultrasound, gross, and microscopic pathology, and serum lipid level analyses in order to determine the mechanism of action, efficacy, and safety of CRYOLIPO II(™). RESULTS: CRYOLIPO II(™) successfully reduced abdominal fat in our porcine model. Gross and microscopic histological results confirmed the noninvasive cold-induced selective subcutaneous fat destruction, and showed increases in pre-adipocyte differentiation and in the activation of lipid catabolism. In particular, we found that CRYOLIPO II(™) may increase PPARδ (delta) levels in adipose tissue at 30-60 days post-treatment. CONCLUSION: Fat reduction by cryolipolysis was successfully achieved in our porcine model. Thus, our findings indicate that CRYOLIPO II(™) may be a promising fat reduction device for body contouring and fat reduction in humans, and that cryolipolysis exerts its effects, at least partly, by targeting the PPARδ signaling pathway. These results show that both investigative and diagnostic potentials capacity.


Subject(s)
Cryosurgery/instrumentation , Lipectomy/instrumentation , Subcutaneous Fat, Abdominal/cytology , Subcutaneous Fat, Abdominal/surgery , Animals , Cryosurgery/methods , Equipment Design , Equipment Failure Analysis , Lipectomy/methods , Male , Reproducibility of Results , Sensitivity and Specificity , Swine , Treatment Outcome
12.
J Korean Med Sci ; 30(12): 1764-76, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26713051

ABSTRACT

Adipose tissue stem cells (ADSCs) would be an attractive autologous cell source. However, ADSCs require invasive procedures, and has potential complications. Recently, urine stem cells (USCs) have been proposed as an alternative stem cell source. In this study, we compared USCs and ADSCs collected from the same patients on stem cell characteristics and capacity to differentiate into various cell lineages to provide a useful guideline for selecting the appropriate type of cell source for use in clinical application. The urine samples were collected via urethral catheterization, and adipose tissue was obtained from subcutaneous fat tissue during elective laparoscopic kidney surgery from the same patient (n = 10). Both cells were plated for primary culture. Cell proliferation, colony formation, cell surface markers, immune modulation, chromosome stability and multi-lineage differentiation were analyzed for each USCs and ADSCs at cell passage 3, 5, and 7. USCs showed high cell proliferation rate, enhanced colony forming ability, strong positive for stem cell markers expression, high efficiency for inhibition of immune cell activation compared to ADSCs at cell passage 3, 5, and 7. In chromosome stability analysis, both cells showed normal karyotype through all passages. In analysis of multi-lineage capability, USCs showed higher myogenic, neurogenic, and endogenic differentiation rate, and lower osteogenic, adipogenic, and chondrogenic differentiation rate compared to ADSCs. Therefore, we expect that USC can be an alternative autologous stem cell source for muscle, neuron and endothelial tissue reconstruction instead of ADSCs.


Subject(s)
Adult Stem Cells/cytology , Adult Stem Cells/immunology , Subcutaneous Fat, Abdominal/cytology , Urine/cytology , Adult Stem Cells/transplantation , Biomarkers/metabolism , Cell Differentiation , Cell Lineage , Cell Proliferation , Cell Separation , Chromosomal Instability , Colony-Forming Units Assay , Humans , Karyotyping , Multipotent Stem Cells/cytology , Multipotent Stem Cells/immunology , Multipotent Stem Cells/transplantation , Transplantation, Autologous
13.
J Lipid Res ; 55(12): 2685-91, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25193996

ABSTRACT

Obesity during childhood and beyond may have its origins during fetal or early postnatal life. At present, there are no suitable in vivo experimental models to study factors that modulate or perturb human fetal white adipose tissue (WAT) expansion, remodeling, development, adipogenesis, angiogenesis, or epigenetics. We have developed such a model. It involves the xenotransplantation of midgestation human WAT into the renal subcapsular space of immunocompromised SCID-beige mice. After an initial latency period of approximately 2 weeks, the tissue begins expanding. The xenografts are healthy and show robust expansion and angiogenesis for at least 2 months following transplantation. Data and cell size and gene expression are consistent with active angiogenesis. The xenografts maintain the expression of genes associated with differentiated adipocyte function. In contrast to the fetal tissue, adult human WAT does not engraft. The long-term viability and phenotypic maintenance of fetal adipose tissue following xenotransplantation may be a function of its autonomous high rates of adipogenesis and angiogenesis. Through the manipulation of the host mice, this model system offers the opportunity to study the mechanisms by which nutrients and other environmental factors affect human adipose tissue development and biology.


Subject(s)
Adipogenesis , Fetal Tissue Transplantation , Intra-Abdominal Fat/transplantation , Models, Biological , Subcutaneous Fat, Abdominal/transplantation , Transplantation, Heterologous , Transplantation, Heterotopic , Abortion, Induced , Adult , Animals , Female , Graft Survival , Humans , Intra-Abdominal Fat/cytology , Intra-Abdominal Fat/embryology , Intra-Abdominal Fat/metabolism , Kidney , Male , Mice, SCID , Microscopy, Fluorescence , Pregnancy , Pregnancy Trimester, Second , Stillbirth , Subcutaneous Fat, Abdominal/cytology , Subcutaneous Fat, Abdominal/embryology , Subcutaneous Fat, Abdominal/metabolism
14.
Am J Physiol Endocrinol Metab ; 307(9): E793-9, 2014 Nov 01.
Article in English | MEDLINE | ID: mdl-25184993

ABSTRACT

Brown adipocytes (BA) generate heat in response to sympathetic activation and are the main site of nonshivering thermogenesis in mammals. Although most BA are located in classic brown adipose tissue depots, BA are also abundant in the inguinal white adipose tissue (iWAT) before weaning. The number of BA is correlated with the density of sympathetic innervation in iWAT; however, the role of continuous sympathetic tone in the establishment and maintenance of BA in WAT has not been investigated. BA marker expression in iWAT was abundant in weaning mice but was greatly reduced by 8 wk of age. Nonetheless, BA phenotype could be rapidly reinstated by acute ß3-adrenergic stimulation with CL-316,243 (CL). Genetic tagging of adipocytes with adiponectin-CreER(T2) demonstrated that CL reinstates uncoupling protein 1 (UCP1) expression in adipocytes that were present before weaning. Chronic surgical denervation dramatically reduced the ability of CL to induce the expression of UCP1 and other BA markers in the tissue as a whole, and this loss of responsiveness was prevented by concurrent treatment with CL. These results indicate that ongoing sympathetic activity is critical to preserve the ability of iWAT fat cells to express a BA phenotype upon adrenergic stimulation.


Subject(s)
Adipocytes, Brown/cytology , Adipogenesis , Aging , Subcutaneous Fat, Abdominal/cytology , Sympathetic Nervous System/metabolism , Synaptic Transmission , Adipocytes, Brown/metabolism , Adrenergic beta-3 Receptor Agonists/pharmacology , Animals , Biomarkers/metabolism , Crosses, Genetic , Denervation/adverse effects , Dioxoles/pharmacology , Gene Expression Regulation, Developmental/drug effects , Groin , Immunohistochemistry , Ion Channels/agonists , Ion Channels/metabolism , Mice, 129 Strain , Mice, Transgenic , Mitochondrial Proteins/agonists , Mitochondrial Proteins/metabolism , Subcutaneous Fat, Abdominal/growth & development , Subcutaneous Fat, Abdominal/innervation , Subcutaneous Fat, Abdominal/metabolism , Sympathetic Nervous System/drug effects , Sympathetic Nervous System/growth & development , Synaptic Transmission/drug effects , Uncoupling Protein 1 , Weaning
16.
Ann Plast Surg ; 72(2): 225-33, 2014 Feb.
Article in English | MEDLINE | ID: mdl-23221992

ABSTRACT

Adult stem cells have recently drawn considerable attention for potential cell therapy applications. However, critical details about their specific in vivo environments and cellular activities are unclear. Adipose tissue-derived mesenchymal stem cells (ASCs) are attractive candidates for treating bone defects, but most studies focus on delivery of in vitro-differentiated cells. We assessed various scaffolding materials for the ability to support osteogenic differentiation of undifferentiated human ASCs in vivo, in athymic nude rat calvaria. Twenty-four 9- to 10-week-old athymic nude Sprague-Dawley rats (250 g) were used in these experiments. Fat tissue from 3 patients was harvested from abdominal tissue discarded during reconstructive breast surgery by transverse rectus abdominis myocutaneous flap, performed at the Asan Medical Center after resection of breast cancer. Human ASCs were extracted from discarded adipose tissue and isolated based on standard International Society for Cellular Therapy protocols. Adipose tissue-derived mesenchymal stem cells were seeded on polylactic glycolic acid, atelocollagen, and hydroxyapatite scaffolds, and osteogenesis was evaluated using bone mineral densitometry, histology, immunohistochemistry, and reverse transcription polymerase chain reaction. The gross appearance of scaffolds seeded with ASCs was strikingly different from that of scaffolds alone. Bone mineral densitometry analysis revealed a 2- to 3-fold increase in mineral density in ASC-seeded scaffolds. In addition, undifferentiated ASCs seeded onto hydroxyapatite scaffolds, but not onto collagen or polylactic glycolic acid scaffolds, expressed human messenger RNA for osteogenic markers such as alkaline phosphatase, osteopontin, osteocalcin, and osteonectin. These results indicate that undifferentiated human ASCs can differentiate into osteocytes or osteoblasts in athymic nude rat calvaria, and the importance of appropriate scaffolding for in vivo ASC differentiation.


Subject(s)
Adult Stem Cells/cytology , Bone Regeneration/physiology , Cell Differentiation , Guided Tissue Regeneration/methods , Mesenchymal Stem Cells/cytology , Skull/injuries , Subcutaneous Fat, Abdominal/cytology , Adult , Animals , Biomarkers/metabolism , Bone Density , Humans , Osteoblasts/cytology , Osteocytes/cytology , Osteogenesis/physiology , Rats , Rats, Nude , Rats, Sprague-Dawley , Skull/pathology , Skull/physiology , Tissue Scaffolds
17.
J Oral Maxillofac Surg ; 71(5): 938-50, 2013 May.
Article in English | MEDLINE | ID: mdl-23375899

ABSTRACT

PURPOSE: Large mandibular resection defects historically have been treated using autogenous bone grafts and reconstruction plates. However, a major drawback of large autogenous bone grafts is donor-site morbidity. PATIENTS AND METHODS: This report describes the replacement of a 10-cm anterior mandibular ameloblastoma resection defect, reproducing the original anatomy of the chin, using a tissue-engineered construct consisting of ß-tricalcium phosphate (ß-TCP) granules, recombinant human bone morphogenetic protein-2 (BMP-2), and Good Manufacturing Practice-level autologous adipose stem cells (ASCs). Unlike prior reports, 1-step in situ bone formation was used without the need for an ectopic bone-formation step. The reconstructed defect was rehabilitated with a dental implant-supported overdenture. An additive manufactured medical skull model was used preoperatively to guide the prebending of patient-specific hardware, including a reconstruction plate and titanium mesh. A subcutaneous adipose tissue sample was harvested from the anterior abdominal wall of the patient before resection and simultaneous reconstruction of the parasymphysis. ASCs were isolated and expanded ex vivo over the next 3 weeks. The cell surface marker expression profile of ASCs was similar to previously reported results and ASCs were analyzed for osteogenic differentiation potential in vitro. The expanded cells were seeded onto a scaffold consisting of ß-TCP and BMP-2 and the cell viability was evaluated. The construct was implanted into the parasymphyseal defect. RESULTS: Ten months after reconstruction, dental implants were inserted into the grafted site, allowing harvesting of bone cores. Histologic examination and in vitro analysis of cell viability and cell surface markers were performed and prosthodontic rehabilitation was completed. CONCLUSION: ASCs in combination with ß-TCP and BMP-2 offer a promising construct for the treatment of large, challenging mandibular defects without the need for ectopic bone formation and allowing rehabilitation with dental implants.


Subject(s)
Adipose Tissue/cytology , Ameloblastoma/surgery , Mandibular Neoplasms/surgery , Plastic Surgery Procedures/methods , Stem Cells/physiology , Tissue Engineering/instrumentation , Tissue Scaffolds , Bone Morphogenetic Protein 2/therapeutic use , Bone Plates , Bone Regeneration/physiology , Bone Substitutes/therapeutic use , Calcium Phosphates/therapeutic use , Cell Culture Techniques , Cell Differentiation/physiology , Cell Survival/physiology , Dental Implants , Dental Prosthesis, Implant-Supported , Denture, Overlay , Denture, Partial , Follow-Up Studies , Humans , Male , Middle Aged , Neoplasm Recurrence, Local/surgery , Osseointegration/physiology , Osteogenesis/physiology , Recombinant Proteins/therapeutic use , Subcutaneous Fat, Abdominal/cytology , Surgical Mesh , Tissue Engineering/standards , Transforming Growth Factor beta/therapeutic use
18.
Genomics ; 100(3): 195-202, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22728265

ABSTRACT

We report a systematic study of gene expression during myogenesis and transdifferentiation in four bovine muscle tissues and of adipogenesis in three bovine fat tissues using DNA microarray analysis. One hundred hybridizations were performed and 7245 genes of known and unknown function were identified as being differentially expressed. Supervised hierarchical cluster analysis of gene expression patterns revealed the tissue specificity of genes. A close relationship in global gene expression observed for adipocyte-like cells derived from muscle and adipocytes derived from intramuscular fat suggests a common origin for these cells. The role of transthyretin in myogenesis is a novel finding. Different genes were highly induced during the transdifferentiation of myogenic satellite cells and in the adipogenesis of preadipocytes, indicating the involvement of different molecular mechanisms in these processes. Induction of CD36 and FABP4 expression in adipocyte-like cells and adipocytes may share a common pathway.


Subject(s)
Adipocytes/cytology , Adipogenesis , Cell Transdifferentiation , Gene Expression Regulation , Satellite Cells, Skeletal Muscle/cytology , Transcriptome , Adipocytes/metabolism , Animals , Cattle , Cells, Cultured , Culture Media/metabolism , Muscle Development , Oligonucleotide Array Sequence Analysis , Organ Specificity , Prealbumin/genetics , Prealbumin/metabolism , Satellite Cells, Skeletal Muscle/metabolism , Subcutaneous Fat, Abdominal/cytology , Subcutaneous Fat, Abdominal/metabolism , Up-Regulation
19.
Am J Physiol Endocrinol Metab ; 303(9): E1126-33, 2012 Nov 01.
Article in English | MEDLINE | ID: mdl-22949029

ABSTRACT

High concentrations of TNF within obese adipose tissue increase basal lipolysis and antagonize insulin signaling. Adipocytes of the obese are also exposed to elevated levels of glucocorticoids (GCs), which antagonize TNF actions in many cell types. We tested the hypothesis that TNF decreases sensitivity to the antilipolytic effect of insulin and that GCs antagonize this effect in differentiated human adipocytes. Lipolysis and expression levels of lipolytic proteins were measured after treating adipocytes with TNF, dexamethasone (DEX), or DEX + TNF for up to 48 h. TNF not only increased basal lipolysis, it caused resistance to the antilipolytic effects of insulin in human adipocytes. DEX alone did not significantly affect lipolysis. Cotreatment with DEX blocked TNF induction of basal lipolysis and insulin resistance by antagonizing TNF stimulation of PKA-mediated phosphorylation of hormone-sensitive lipase (HSL) at Ser56³ and Ser66° and perilipin. TNF did not affect perilipin, HSL, or phosphodiesterase-3B mass but paradoxically suppressed adipose tissue triglyceride lipase expression, and this effect was blocked by DEX. The extent to which GCs can restrain the lipolytic actions of TNF may both diminish the potentially deleterious effects of excess lipolysis and contribute to fat accumulation in obesity.


Subject(s)
Glucocorticoids/pharmacology , Insulin Resistance , Insulin/metabolism , Lipolysis/drug effects , Subcutaneous Fat, Abdominal/drug effects , Tumor Necrosis Factor-alpha/metabolism , Adult , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cells, Cultured , Cyclic AMP-Dependent Protein Kinases , Cyclic Nucleotide Phosphodiesterases, Type 3/genetics , Cyclic Nucleotide Phosphodiesterases, Type 3/metabolism , Dexamethasone/pharmacology , Female , Gene Expression Regulation/drug effects , Humans , Lipase/genetics , Lipase/metabolism , Male , Middle Aged , Perilipin-1 , Phosphoproteins/genetics , Phosphoproteins/metabolism , Phosphorylation/drug effects , Protein Processing, Post-Translational/drug effects , Sterol Esterase/genetics , Sterol Esterase/metabolism , Subcutaneous Fat, Abdominal/cytology , Subcutaneous Fat, Abdominal/metabolism , Tissue Culture Techniques , Tumor Necrosis Factor-alpha/antagonists & inhibitors
20.
Orv Hetil ; 153(46): 1816-31, 2012 Nov 18.
Article in Hungarian | MEDLINE | ID: mdl-23146782

ABSTRACT

Autologous fat transplantation is often used in aesthetic plastic surgery, and is recently becoming increasingly popular in the reconstruction of soft tissue defects following oncological surgery. A still not standardized technique of fat transplantation for breast cancer reconstruction is rapidly getting popular. The procedure is not a passive volume replacement, but transplantation of biologically active tissue bearing endocrine, paracrine, exocrine functions and containing fat-derived stem cells, which in the tumorous environment raises many questions in relation to the oncological safety and diagnostic follow-up. Although long-term results based on prospective, randomized studies are not yet available, published clinical experience is promising and reveals an effective and surgically safe procedure if used with appropriate indications and techniques. The authors conducted a broad review of the literature, presenting indications, technique, molecular interactions, and potential risks of the clinical results of autologous fat transplantation in the breast cancer reconstructive surgery. The authors initiated that breast and plastic surgeons should promote adequate long term follow-up of breast cancer patients who underwent breast reconstruction with autologous fat transplantation by the establishment of national registries.


Subject(s)
Adipocytes/metabolism , Adipocytes/transplantation , Mammaplasty/methods , Stem Cell Transplantation , Subcutaneous Fat, Abdominal/cytology , Subcutaneous Fat, Abdominal/transplantation , Breast Neoplasms/surgery , Female , Graft Survival , Humans , Transplantation, Autologous , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL