Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
2.
Ann Plast Surg ; 75(6): 666-71, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26207555

ABSTRACT

INTRODUCTION: Pluripotential cells in adipose tissue may be important in long-term volume retention and regenerative effects of fat grafting. Unfortunately, graft harvest with lipoaspiration significantly depletes the population of stromal vascular cells, which includes adipose stem cells. Stromal vascular fraction (SVF) cells may be isolated from excess lipoaspirate at the point of care and used to replenish fat grafts, a technique termed cell-assisted lipotransfer (CAL). Preclinical and clinical evidence supports the rationale of CAL but clinical adoption of the strategy requires evidence of clinical safety. This prospective, level 1 study reports clinical safety of SVF-enhanced fat grafting using a manual, collagenase-based separation process to isolate autogenous progenitor cells from lipoaspirate at the point of care. METHODS: One hundred sixty-four subjects underwent 174 SVF-enhanced autologous fat grafting procedures at the university stem cell center between August 2009 and November 2014 for a variety of cosmetic and reconstructive indications. RESULTS: Cell-assisted lipotransfer was performed for a variety of cosmetic and reconstructive indications. The mean time of the SVF isolation process was 91 minutes. Because of the frequent concomitant procedures, the average operating room time increased by only 11 minutes. Mean follow-up was 19.9 months. There were no major complications and 6 minor complications. No collagenase or neutral protease related complications were observed. CONCLUSIONS: This series of 174 CAL cases demonstrates that SVF cell isolation using a standardized, manual, collagenase-based process at the POC is equivalent in safety compared to nonenhanced fat grafting. These results support expanded use of CAL in the clinical research setting.


Subject(s)
Lipectomy , Point-of-Care Systems , Stem Cell Transplantation/methods , Subcutaneous Fat/transplantation , Adult , Aged , Cell Separation/methods , Follow-Up Studies , Humans , Middle Aged , Prospective Studies , Stem Cells , Subcutaneous Fat/cytology , Transplantation, Autologous
3.
Adv Wound Care (New Rochelle) ; 3(1): 38-45, 2014 Jan 01.
Article in English | MEDLINE | ID: mdl-24761343

ABSTRACT

Objective: To develop a closed, automated system that standardizes the processing of human adipose tissue to obtain and concentrate regenerative cells suitable for clinical treatment of thermal and radioactive burn wounds. Approach: A medical device was designed to automate processing of adipose tissue to obtain a clinical-grade cell output of stromal vascular cells that may be used immediately as a therapy for a number of conditions, including nonhealing wounds resulting from radiation damage. Results: The Celution® System reliably and reproducibly generated adipose-derived regenerative cells (ADRCs) from tissue collected manually and from three commercial power-assisted liposuction devices. The entire process of introducing tissue into the system, tissue washing and proteolytic digestion, isolation and concentration of the nonadipocyte nucleated cell fraction, and return to the patient as a wound therapeutic, can be achieved in approximately 1.5 h. An alternative approach that applies ultrasound energy in place of enzymatic digestion demonstrates extremely poor efficiency cell extraction. Innovation: The Celution System is the first medical device validated and approved by multiple international regulatory authorities to generate autologous stromal vascular cells from adipose tissue that can be used in a real-time bedside manner. Conclusion: Initial preclinical and clinical studies using ADRCs obtained using the automated tissue processing Celution device described herein validate a safe and effective manner to obtain a promising novel cell-based treatment for wound healing.

4.
Aesthet Surg J ; 33(5): 698-704, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23718978

ABSTRACT

BACKGROUND: Although clinical evidence of successful autologous fat transfer (AFT) using third-generation ultrasound-assisted liposuction (UAL) is readily available, no study has quantified adipocyte viability using standardized methods. OBJECTIVES: The authors assess acute adipocyte viability following fat aspiration as a first step in determining the overall efficacy of using third-generation UAL for AFT. METHODS: Lipoaspirate samples were collected from patients who underwent elective liposuction procedures at multiple surgery centers. Patients with a history of bleeding disorders, diabetes, human immunodeficiency virus, or lipoatrophy disorders were excluded. The UAL system (VASER; Sound Surgical Technologies, Inc, Louisville, Colorado) was set at 60% amplitude in pulsed mode with vacuum aspiration of 15 in Hg or less. Laboratory analysis included free lipid volume, viability via lipolysis and propidium iodide staining, and cytological analysis, including cell surface protein examination and hematoxylin and eosin staining. RESULTS: The lipolysis assay revealed metabolically active adipocytes with a mean (SD) correlative viability of 85.1% (11%). Direct measures of acute viability via propidium iodide staining resulted in a mean (SD) viability measure of 88.7% (3.5%). Both mean values are within the historical range reported from syringe and vacuum-assisted lipoaspiration. Aqueous and lipid contents were favorably reduced after washing and filtering (Puregraft system; Cytori Therapeutics, Inc, San Diego, California). Cellular phenotypes identified were primarily white blood cells or vascular endothelial and vascular associated cells. CONCLUSIONS: Adipose tissue acquired via third-generation UAL is viable at harvest and is potentially a suitable source for autologous fat grafts. These results confirm reported clinical successes utilizing third-generation ultrasound lipoaspirate for AFT.


Subject(s)
Adipocytes/physiology , Adipose Tissue/transplantation , Lipectomy/methods , Ultrasonic Therapy/methods , Adipocytes/transplantation , Adipose Tissue/diagnostic imaging , Adipose Tissue/pathology , Adult , Biopsy, Needle , Cell Survival/physiology , Female , Humans , Immunohistochemistry , Male , Middle Aged , Sampling Studies , Tissue and Organ Harvesting/methods , Transplantation, Autologous , Treatment Outcome , Ultrasonography , Young Adult
5.
Plast Reconstr Surg ; 131(4): 873-880, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23542259

ABSTRACT

BACKGROUND: Successful long-term volume retention of an autologous fat graft is problematic. The presence of contaminating cells, tumescent fluid, and free lipid in the graft contributes to disparate outcomes. Better preparation methods for the fat graft before transplantation may significantly improve results. METHODS: Subcutaneous fat from 22 donors was divided and processed using various graft preparation methods: (1) no manipulation control, (2) gravity separation, (3) Coleman centrifugation, and (4) simultaneous washing with filtration using a commercially available system (Puregraft; Cytori Therapeutics, Inc., San Diego, Calif.). Fat grafts from various preparation methods were examined for free lipid, aqueous liquid, viable tissue, and blood cell content. Adipose tissue viability was determined by measuring glycerol release after agonist induction of lipolysis. RESULTS: All test graft preparation methods exhibited significantly less aqueous fluid and blood cell content compared with the control. Grafts prepared by washing with filtration exhibited significantly reduced blood cell and free lipid content, with significantly greater adipose tissue viability than other methods. CONCLUSION: Washing with filtration within a closed system produces a fat graft with higher tissue viability and lower presence of contaminants compared with grafts prepared by alternate methods.


Subject(s)
Subcutaneous Fat , Tissue and Organ Harvesting/methods , Adult , Centrifugation , Female , Filtration , Gravitation , Humans , Middle Aged , Subcutaneous Fat/transplantation , Young Adult
6.
Methods Mol Biol ; 702: 87-105, 2011.
Article in English | MEDLINE | ID: mdl-21082397

ABSTRACT

The popularity of nonhematopoietic, adult tissue-derived stem and progenitor cells for use as a cellular research tool, and ultimately as a clinical therapeutic, has increased exponentially over the past decade. Almost all adult-derived stem/progenitor cells (autologous and allogeneic), with one exception, require at least some ex vivo expansion or further manipulation prior to use to satisfy efficacy and safety requirements for preclinical or clinical use. The principal reason is the relatively low frequency of these therapeutically valuable cells within any given adult tissue, except for adipose tissue, which has been shown to have at least two log greater concentrations of these progenitor cells. Therefore, use of autologous adipose-derived cells as both a research tool and cell therapeutic is feasible and has been shown to be both safe and efficacious in preclinical and clinical models of injury and disease. The development and utilization of automated processes and instrumentation such as Cytori Therapeutics' Celution® System to reduce variability and increase quality of the recovered cells is requisite for clinical use and preferred by basic researchers. Here, use of an automated, closed processing platform for isolation and concentration of adipose-derived stem and regenerative cells is described, including a profile of the isolated cells immediately prior to use, and commonly used methods to quantify and qualitatively assess the recovered cells.


Subject(s)
Adipose Tissue/cytology , Automation/methods , Cell Culture Techniques/methods , Cell Separation/methods , Regeneration/physiology , Stem Cells/cytology , Cell Count , Cell Survival , Cells, Cultured , Colony-Forming Units Assay , Culture Media , Humans
7.
Ophthalmic Plast Reconstr Surg ; 25(1): 27-32, 2009.
Article in English | MEDLINE | ID: mdl-19273919

ABSTRACT

PURPOSE: To identify pluripotential stem cells from human orbital adipose depots. METHODS: Pluripotential adipose-derived stem cells were isolated from human orbital adipose during routine blepharoplasty surgery. Fresh adipose tissue was separated in nasal fat and central (preaponeurotic) fat. Individual adipose depots were minced, enzymatically digested, and plated on plastic culture dishes. Adherent populations of cells were expanded in culture, characterized by flow cytometry, and assayed for the potential to differentiate in different cell lineages. RESULTS: Orbital adipose-derived cells from the nasal and central adipose depots showed the potential to differentiate into the adipocyte, smooth muscle, and neuronal/glial lineages and expressed a CD marker protein profile consistent with that observed for adipose-derived stem cells from other adipose depots. CONCLUSIONS: A population of adherent cells capable of pluripotential differentiation in vitro exists within adult human orbital adipose tissue. These cells are similar to those described in other adipose depots and will help facilitate understanding of orbital diseases and may provide a novel tissue source for the development of ocular regenerative medicine therapies.


Subject(s)
Adipose Tissue/cytology , Adult Stem Cells/cytology , Orbit/cytology , Pluripotent Stem Cells/cytology , Adipocytes/cytology , Adipocytes/metabolism , Adipose Tissue/metabolism , Adult , Adult Stem Cells/metabolism , Antigens, CD/metabolism , Biomarkers/metabolism , Blepharoplasty , Cell Differentiation/physiology , Cell Separation/methods , Colony-Forming Units Assay , Flow Cytometry , Fluorescent Antibody Technique, Indirect , Humans , Muscle, Smooth/cytology , Muscle, Smooth/metabolism , Neuroglia/cytology , Neuroglia/metabolism , Orbit/metabolism , Phenotype , Pluripotent Stem Cells/metabolism
8.
J Cell Physiol ; 206(1): 229-37, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16021633

ABSTRACT

Pools of human adipose-derived adult stem (hADAS) cells can exhibit multiple differentiated phenotypes under appropriate in vitro culture conditions. Because adipose tissue is abundant and easily accessible, hADAS cells offer a promising source of cells for tissue engineering and other cell-based therapies. However, it is unclear whether individual hADAS cells can give rise to multiple differentiated phenotypes or whether each phenotype arises from a subset of committed progenitor cells that exists within a heterogeneous population. The goal of this study was to test the hypothesis that single hADAS are multipotent at a clonal level. hADAS cells were isolated from liposuction waste, and ring cloning was performed to select cells derived from a single progenitor cell. Forty-five clones were expanded through four passages and then induced for adipogenesis, osteogenesis, chondrogenesis, and neurogenesis using lineage-specific differentiation media. Quantitative differentiation criteria for each lineage were determined using histological and biochemical analyses. Eighty one percent of the hADAS cell clones differentiated into at least one of the lineages. In addition, 52% of the hADAS cell clones differentiated into two or more of the lineages. More clones expressed phenotypes of osteoblasts (48%), chondrocytes (43%), and neuron-like cells (52%) than of adipocytes (12%), possibly due to the loss of adipogenic ability after repeated subcultures. The findings are consistent with the hypothesis that hADAS cells are a type of multipotent adult stem cell and not solely a mixed population of unipotent progenitor cells. However, it is important to exercise caution in interpreting these results until they are validated using functional in vivo assays.


Subject(s)
Adipose Tissue/cytology , Cell Differentiation/physiology , Clone Cells/physiology , Multipotent Stem Cells/physiology , Adult , Cell Culture Techniques , Cell Lineage , Cells, Cultured , Clone Cells/cytology , Female , Humans , Middle Aged , Multipotent Stem Cells/cytology , Phenotype , Tissue Engineering
9.
Keio J Med ; 54(3): 132-41, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16237275

ABSTRACT

Tissue engineering offers considerable promise in the repair or replacement of diseased and/or damaged tissues. The cellular component of this regenerative approach will play a key role in bringing these tissue engineered constructs from the laboratory bench to the clinical bedside. However, the ideal source of cells still remains unclear and may differ depending upon the application. Current research for many applications is focused on the use of adult stem cells. The properties of adult stem cells that make them well-suited for regenerative medicine are (1) ease of harvest for autologous transplantation, (2) high proliferation rates for ex vivo expansion and (3) multilineage differentiation capacity. This review will highlight the use of adipose tissue as a reservoir of adult stem cells and draw conclusions based upon comparisons with bone marrow stromal cells.


Subject(s)
Adipose Tissue/cytology , Multipotent Stem Cells/cytology , Adipose Tissue/growth & development , Adipose Tissue/immunology , Animals , Antigens, CD/metabolism , Cell Differentiation , Cell Membrane/immunology , Chondrogenesis , Colony-Forming Units Assay , Genetic Vectors , Heart/growth & development , Hematopoiesis , Humans , Multipotent Stem Cells/immunology , Muscle, Skeletal/growth & development , Neovascularization, Physiologic , Nervous System/growth & development , Osteogenesis
10.
Cell Tissue Res ; 320(3): 417-26, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15856306

ABSTRACT

Tissue regeneration and scarless healing involves the complete replacement and functional restoration of damaged organs and tissues. In this study of the "scarless healing" MRL mouse model, we demonstrate that 2-mm diameter through-and-through holes made in the cartilaginous part of previously injured MRL mouse ears are closed more efficiently, and that the regenerative repair response is significantly accelerated compared with unprimed MRL and control "nonhealer" strains of mice. Accelerated healing was detected both locally and distally from the original site of injury indicating the involvement of systemic components such as circulating cell types or soluble factors. Histologically, we observed early differences during the wound repair process (before Day 4 post injury) with accelerated formation of blastema-like structures, epidermal downgrowths, and enhanced epithelium thickening in wound border zones in primed MRL mice versus unprimed MRL mice. Although the mechanism of tissue regeneration remains unclear, the results from this study justify the use of the MRL model for further experimentation directed toward the identification of proteins and cell types capable of stimulating scarless tissue regeneration.


Subject(s)
Cartilage/pathology , Ear/pathology , Regeneration , Wound Healing/physiology , Animals , Cartilage/injuries , Ear/injuries , Female , Leukocyte Count , Macrophages/pathology , Macrophages/physiology , Mice , Mice, Inbred C57BL , Mice, SCID , Monocytes/pathology , Monocytes/physiology , Silicon Dioxide
11.
Tissue Eng ; 10(3-4): 371-80, 2004.
Article in English | MEDLINE | ID: mdl-15165454

ABSTRACT

Adult subcutaneous fat tissue is an abundant source of multipotent cells. Previous studies from our laboratory have shown that, in vitro, adipose-derived adult stem (ADAS) cells express bone marker proteins including alkaline phosphatase, type I collagen, osteopontin, and osteocalcin and produce a mineralized matrix as shown by alizarin red staining. In the current study, the ADAS cell ability to form osteoid in vivo was determined. ADAS cells were isolated from liposuction waste of three individual donors and expanded in vitro before implantation. Equal numbers of cells (3 x 10(6)) were loaded onto either hydroxyapatite/tricalcium phosphate (HA-TCP) cubes or the collagen/HA-TCP composite matrix, Collagraft, and then implanted subcutaneously into SCID mice. After 6 weeks, implants were removed, fixed, and demineralized and sectioned for hematoxylin and eosin staining. Osteoid formation was observed in 80% of HA-TCP implants loaded with ADAS cells. Only 20% of Collagraft implants were positive for the presence of osteoid matrix. Whereas 100% of HA-TCP implants loaded with hFOB 1.19 cells formed osteoid, Collagraft loaded with hFOB 1.19 cells displayed a high degree of adipose tissue within the matrix. Immunostaining of serial sections for human nuclear antigen demonstrated that the osteoid contained human cells. Osteoid formation was not observed in control HA-TCP or Collagraft matrices implanted without cells. In summary, the data demonstrate the ability of ADAS cells to form osteoid matrix in vivo. Because of their abundance and accessibility, ADAS cells may prove to be a novel cell therapeutic for bone repair and regeneration.


Subject(s)
Adipose Tissue/physiology , Bone and Bones/physiology , Osteoblasts/physiology , Stem Cells/physiology , Adipose Tissue/cytology , Adipose Tissue/ultrastructure , Biomarkers , Bone Regeneration/physiology , Humans , Microscopy, Electron, Scanning , Stem Cells/cytology , Stem Cells/ultrastructure
12.
Eur J Endocrinol ; 147(2): 269-73, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12153751

ABSTRACT

BACKGROUND: Estrogens and androgens have anti-resorptive effects on bone, although recent evidence indicates that, even in men, estrogen is the dominant sex steroid regulating bone resorption. The receptor activator of NF-kappaB ligand is essential for osteoclastic bone resorption, and its effects are blocked by the decoy receptor, osteoprotegerin (OPG). While estrogen has been shown to induce osteoblastic OPG production, the effects of androgens on OPG production have not been defined. METHODS: In this study, we assessed the regulation of OPG by androgens in hFOB/AR-6, an immortalized fetal osteoblastic cell line stably transfected with the human androgen receptor (AR), and MSC cells, primary human pluripotent marrow stromal cells capable of differentiating towards mature osteoblasts. RESULTS AND CONCLUSIONS: 5Alpha-dihydrotestosterone (DHT) dose-dependently decreased OPG mRNA levels and protein concentrations in hFOB/AR-6 cells by up to 50 and 60% respectively (P<0.001). Inhibition of OPG mRNA levels and protein production by 5alpha-DHT was completely abrogated by the AR antagonist, hydroxyflutamide (OHF), indicating that these effects are directly mediated by the AR. Of note, OHF alone increased OPG mRNA levels and protein secretion by 2- to 3-fold. Moreover, 5alpha-DHT and testosterone also decreased OPG protein secretion by 40-46% in the untransformed MSC cells, while OHF stimulated it. In conclusion, we demonstrate that androgens specifically inhibit OPG mRNA levels and protein secretion by osteoblastic cells.


Subject(s)
Androgen Antagonists/pharmacology , Androgens/pharmacology , Gene Expression Regulation/drug effects , Glycoproteins/genetics , Osteoblasts/metabolism , Receptors, Cytoplasmic and Nuclear/genetics , Androgens/metabolism , Blotting, Northern , Bone Marrow Cells/metabolism , Cell Line , Cell Line, Transformed , Dihydrotestosterone/pharmacology , Embryo, Mammalian , Humans , Osteoprotegerin , RNA, Messenger/analysis , Receptors, Androgen/genetics , Receptors, Androgen/physiology , Receptors, Tumor Necrosis Factor , Reverse Transcriptase Polymerase Chain Reaction , Stromal Cells/metabolism , Transfection
13.
Biochem Biophys Res Commun ; 294(2): 371-9, 2002 Jun 07.
Article in English | MEDLINE | ID: mdl-12051722

ABSTRACT

The identification of cells capable of neuronal differentiation has great potential for cellular therapies. We examined whether murine and human adipose-derived adult stem (ADAS) cells can be induced to undergo neuronal differentiation. We isolated ADAS cells from the adipose tissue of adult BalbC mice or from human liposuction tissue and induced neuronal differentiation with valproic acid, butylated hydroxyanisole, insulin, and hydrocortisone. As early as 1-3 h after neuronal induction, the phenotype of ADAS cells changed towards neuronal morphology. Following neuronal induction, muADAS cells displayed immunocytochemical staining for GFAP, nestin and NeuN and huADAS cells displayed staining for intermediate filament M, nestin, and NeuN. Following neuronal induction of murine and human ADAS cells, Western blot analysis confirmed GFAP, nestin, and NeuN protein expression. Pretreatment with EGF and basic FGF augmented the neuronal differentiation of huADAS cells. The neuronal differentiation of stromal cells from adipose tissue has broad biological and clinical implications.


Subject(s)
Adipose Tissue/cytology , Cell Differentiation/physiology , Neurons/cytology , Stromal Cells/cytology , Stromal Cells/physiology , Adipose Tissue/drug effects , Animals , Anti-Inflammatory Agents/pharmacology , Antigens, Differentiation/biosynthesis , Antioxidants/pharmacology , Blotting, Western , Butylated Hydroxyanisole/pharmacology , Cell Differentiation/drug effects , Cells, Cultured , Epidermal Growth Factor/pharmacology , Fibroblast Growth Factor 2/pharmacology , Flow Cytometry , GABA Agents/pharmacology , Humans , Hydrocortisone/pharmacology , Immunohistochemistry , Insulin/pharmacology , Mice , Mice, Inbred BALB C , Neurons/metabolism , Phenotype , Stem Cells/cytology , Stem Cells/drug effects , Stem Cells/physiology , Stromal Cells/drug effects , Valproic Acid/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...