Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 81
Filter
1.
Adipocyte ; 7(2): 96-105, 2018.
Article in English | MEDLINE | ID: mdl-29411671

ABSTRACT

Stromal/stem cell differentiation is controlled by a vast array of regulatory mechanisms. Included within these are methods of mRNA gene regulation that occur at the level of epigenetic, transcriptional, and/or posttranscriptional modifications. Current studies that evaluate the posttranscriptional regulation of mRNA demonstrate microRNAs (miRNAs) as key mediators of stem cell differentiation through the inhibition of mRNA translation. miRNA expression is enhanced during both adipogenic and osteogenic differentiation; however, the mechanism by which miRNA expression is altered during stem cell differentiation is less understood. Here we demonstrate for the first time that adipose-derived stromal/stem cells (ASCs) induced to an adipogenic or osteogenic lineage have differences in strand preference (-3p and -5p) for miRNAs originating from the same primary transcript. Furthermore, evaluation of miRNA expression in ASCs demonstrates alterations in both miRNA strand preference and 5'seed site heterogeneity. Additionally, we show that during stem cell differentiation there are alterations in expression of genes associated with the miRNA biogenesis pathway. Quantitative RT-PCR demonstrated changes in the Argonautes (AGO1-4), Drosha, and Dicer at intervals of ASC adipogenic and osteogenic differentiation compared to untreated ASCs. Specifically, we demonstrated altered expression of the AGOs occurring during both adipogenesis and osteogenesis, with osteogenesis increasing AGO1-4 expression and adipogenesis decreasing AGO1 gene and protein expression. These data demonstrate changes to components of the miRNA biogenesis pathway during stromal/stem cell differentiation. Identifying regulatory mechanisms for miRNA processing during ASC differentiation may lead to novel mechanisms for the manipulation of lineage differentiation of the ASC through the global regulation of miRNA as opposed to singular regulatory mechanisms.


Subject(s)
Adipose Tissue/cytology , Cell Differentiation , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , MicroRNAs/biosynthesis , Cells, Cultured , Humans
2.
J Cell Physiol ; 233(9): 7035-7044, 2018 09.
Article in English | MEDLINE | ID: mdl-29377109

ABSTRACT

Post-traumatic heterotopic ossification (HO) is the formation of ectopic bone in non-osseous structures following injury. The precise mechanism for bone development following trauma is unknown; however, early onset of HO may involve the production of pro-osteogenic serum factors. Here we evaluated serum from a cohort of civilian and military patients post trauma to determine early induction gene signatures in orthopaedic trauma induced HO. To test this, human adipose derived stromal/stem cells (hASCs) were stimulated with human serum from patients who developed HO following trauma and evaluated for a gene panel with qPCR. Pathway gene analysis ontology revealed that hASCs stimulated with serum from patients who developed HO had altered gene expression in the activator protein 1 (AP1) and AP1 transcriptional targets pathways. Notably, there was a significant repression in FOS gene expression in hASCs treated with serum from individuals with HO. Furthermore, the mitogen-activated protein kinase (MAPK) signaling pathway was activated in hASCs following serum exposure from individuals with HO. Serum from both military and civilian patients with trauma induced HO had elevated downstream genes associated with the MAPK pathways. Stimulation of hASCs with known regulators of osteogenesis (BMP2, IL6, Forskolin, and WNT3A) failed to recapitulate the gene signature observed in hASCs following serum stimulation, suggesting non-canonical mechanisms for gene regulation in trauma induced HO. These findings provide new insight for the development of HO and support ongoing work linking the systemic response to injury with wound specific outcomes.


Subject(s)
Adipose Tissue/cytology , MAP Kinase Signaling System , Ossification, Heterotopic/blood , Ossification, Heterotopic/etiology , Stem Cells/enzymology , Wounds and Injuries/complications , Adult , Cell Differentiation , Humans , Male , Middle Aged , Models, Biological , Osteogenesis , Transcription Factor AP-1/metabolism , Young Adult
3.
Adipocyte ; 5(1): 53-64, 2016.
Article in English | MEDLINE | ID: mdl-27144097

ABSTRACT

Human adipose-derived stromal/stem cells (ASCs) display potential to be used in regenerative stem cell therapies and as treatments for inflammatory and autoimmune disorders. Despite promising use of ASCs as therapeutics, little is known about their susceptibility to infectious agents. In this study, we demonstrate that ASCs are highly susceptible to human cytomegalovirus (HCMV) infection and permissive for replication leading to release of infectious virions. Additionally, many basic ASC functions are inhibited during HCMV infection, such as differentiation and immunomodulatory potential. To our knowledge this is the first study examining potential adverse effects of HCMV infection on ASC biology. Our results suggest, that an active HCMV infection during ASC therapy may result in a poor clinical outcome due to interference by the virus.

4.
Biochimie ; 124: 98-111, 2016 May.
Article in English | MEDLINE | ID: mdl-25726914

ABSTRACT

MicroRNAs (miRNAs) are small non-coding RNAs that regulate gene expression through targeting and suppression of mRNAs. miRNAs have been under investigation for the past twenty years and there is a large breadth of information on miRNAs in diseases such as cancer and immunology. Only more recently have miRNAs shown promise as a mechanism for intervention with respect to diseases of the bone and adipose tissue. In mesenchymal stem cell (MSC) differentiation, alterations in miRNA expression patterns can differentially promote an osteogenic, adipogenic, or myogenic phenotype. This manuscript reviews the current literature with respect to miRNAs in the context of MSC function with a particular focus on novel avenues for the examination of miRNA associated with bone and adipose tissue biology and disease. Specifically we highlight the need for a greater depth of investigation on MSCs with respect to miRNA biogenesis, processing, strand selection, and heterogeneity. We discuss how these mechanisms facilitate both altered miRNA expression and function.


Subject(s)
Adipose Tissue/metabolism , Bone and Bones/metabolism , Cell Differentiation , Mesenchymal Stem Cells/metabolism , MicroRNAs/metabolism , Transcriptome , Adipogenesis , Adipose Tissue/pathology , Animals , Bone and Bones/pathology , Humans , Mesenchymal Stem Cells/pathology , Muscle Development , Osteogenesis
6.
J Tissue Eng Regen Med ; 9(4): 389-404, 2015 Apr.
Article in English | MEDLINE | ID: mdl-23166107

ABSTRACT

In this study, three different akermanite:poly-ϵ-caprolactone (PCL) composite scaffolds (wt%: 75:25, 50:50, 25:75) were characterized in terms of structure, compression strength, degradation rate and in vitro biocompatibility to human adipose-derived stem cells (hASC). Pure ceramic scaffolds [CellCeram™, custom-made, 40:60 wt%; ß-tricalcium phosphate (ß-TCP):hydroxyapatite (HA); and akermanite] and PCL scaffolds served as experimental controls. Compared to ceramic scaffolds, the authors hypothesized that optimal akermanite:PCL composites would have improved compression strength and comparable biocompatibility to hASC. Electron microscopy analysis revealed that PCL-containing scaffolds had the highest porosity but CellCeram™ had the greatest pore size. In general, compression strength in PCL-containing scaffolds was greater than in ceramic scaffolds. PCL-containing scaffolds were also more stable in culture than ceramic scaffolds. Nonetheless, mass losses after 21 days were observed in all scaffold types. Reduced hASC metabolic activity and increased cell detachment were observed after acute exposure to akermanite:PCL extracts (wt%: 75:25, 50:50). Among the PCL-containing scaffolds, hASC cultured for 21 days on akermanite:PCL (wt%: 75:25) discs displayed the highest viability, increased expression of osteogenic markers (alkaline phosphatase and osteocalcin) and lowest IL-6 expression. Together, the results indicate that akermanite:PCL composites may have appropriate mechanical and biocompatibility properties for use as bone tissue scaffolds.


Subject(s)
Adipose Tissue/metabolism , Ceramics/chemistry , Osteogenesis , Polyesters/chemistry , Stem Cells/metabolism , Tissue Scaffolds/chemistry , Adipose Tissue/cytology , Antigens, Differentiation/biosynthesis , Cell Differentiation , Humans , Stem Cells/cytology , Tissue Engineering
7.
Obesity (Silver Spring) ; 22(4): 1091-102, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24795999

ABSTRACT

OBJECTIVE: The study aims to explore long-term dietary effects on increases in body mass and fat depot enlargement through the recruitment of early in life labeled progenitor cells to the adipolineage. METHODS: Neonate male C57BL/6J (B6) mice were injected intraperitoneally with BrdU. From 4 until 30 weeks of age they were fed either low fat diet (LFD) or high fat diet (HFD). BrdU-labeled cells were analyzed by flow cytometric and immunohistochemical assays after 10 days and 4, 8, 16, and 30 weeks. RESULTS: Mice fed HFD were heavier than mice fed LFD with the most dramatic disparity recorded between week 16 and 30. BrdU-bearing cells showed the decrease in the percentage content of labeled cells in inguinal (iWAT), epididymal (eWAT) and bone marrow (BM) tissues, regardless diets. However, iWAT collected from animals on HFD showed significant increase in labeled-cells at week 16th, which coincides with robust increase in inguinal but not epididymal fat weight between 16 and 30 weeks age. CONCLUSIONS: Cells labeled with BrdU during neonate life of B6 mice persist in fat tissues for long period of time and are recruited to the adipocyte lineage in a favorable (obesogenic) environment in iWAT but not in eWAT.


Subject(s)
Adipocytes/pathology , Adipose Tissue, White/pathology , Diet, High-Fat , Stem Cells/pathology , Adipocytes/metabolism , Adipose Tissue, White/metabolism , Animals , Body Composition , Body Fat Distribution , Body Mass Index , Bone Marrow/pathology , Bromodeoxyuridine/metabolism , Cell Differentiation , Cell Lineage , Diet, Fat-Restricted , Epididymis/pathology , Inguinal Canal/pathology , Male , Mice , Mice, Inbred C57BL , Models, Animal , Stem Cells/metabolism , Time Factors
8.
Biochem Biophys Res Commun ; 438(1): 180-5, 2013 Aug 16.
Article in English | MEDLINE | ID: mdl-23876311

ABSTRACT

Adipose-derived stem/stromal cell (ASC)-based tissue engineered muscle grafts could provide an effective alternative therapy to autografts - which are limited by their availability - for the regeneration of damaged muscle. However, the current myogenic potential of ASCs is limited by their low differentiation efficiency into myoblasts. The aim of this study was to enhance the myogenic response of human ASCs to biochemical cues by providing biophysical stimuli (11% cyclic uniaxial strain, 0.5 Hz, 1h/day) to mimic the cues present in the native muscle microenvironment. ASCs elongated and fused upon induction with myogenic induction medium alone. Yet, their myogenic characteristics were significantly enhanced with the addition of biophysical stimulation; the nuclei per cell increased approximately 4.5-fold by day 21 in dynamic compared to static conditions (23.3 ± 7.3 vs. 5.2 ± 1.6, respectively), they aligned at almost 45° to the direction of strain, and exhibited significantly higher expression of myogenic proteins (desmin, myoD and myosin heavy chain). These results demonstrate that mimicking the biophysical cues inherent to the native muscle microenvironment in monolayer ASC cultures significantly improves their differentiation along the myogenic lineage.


Subject(s)
Adipocytes/cytology , Mechanotransduction, Cellular/physiology , Muscle Development/physiology , Muscle Fibers, Skeletal/cytology , Muscle Fibers, Skeletal/physiology , Stem Cells/cytology , Stem Cells/physiology , Adipocytes/physiology , Cell Differentiation , Cell Proliferation , Cells, Cultured , Humans , Physical Stimulation/methods , Stress, Mechanical , Tissue Engineering/methods
9.
Cancer Gene Ther ; 18(8): 579-86, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21660061

ABSTRACT

Established cell transfection via nucleofection relies on nucleofection buffers with unknown and proprietary makeup due to trade secrecy, inhibiting the possibility of using this otherwise effective method for developing cell therapy. We devised a three-step method for discovering an optimal formulation for the nucleofection of any cell line. These steps include the selection of the best nucleofection program and known buffer type, selection of the best polymer for boosting the transfection efficiency of the best buffer and the comparison with the optimal buffer from an established commercial vendor (Amaxa). Using this three-step selection system, competitive nucleofection formulations were discovered for multiple cell lines, which are equal to or surpass the efficiency of the Amaxa nucleofector solution in a variety of cells and cell lines, including primary adipose stem cells, muscle cells, tumor cells and immune cells. Through the use of scanning electron microscopy, we have revealed morphological changes, which predispose for the ability of these buffers to assist in transferring plasmid DNA into the nuclear space. Our formulation may greatly reduce the cost of electroporation study in laboratory and boosts the potential of application of electroporation-based cell therapies in clinical trials.


Subject(s)
Cell- and Tissue-Based Therapy/methods , Electroporation/methods , Animals , Cell Line, Tumor , HeLa Cells , Humans , Mice , Transfection
10.
J Cell Physiol ; 226(3): 843-51, 2011 Mar.
Article in English | MEDLINE | ID: mdl-20857424

ABSTRACT

Adipose tissue is composed of lipid-filled mature adipocytes and a heterogeneous stromal vascular fraction (SVF) population of cells. Similarly, the bone marrow (BM) is composed of multiple cell types including adipocytes, hematopoietic, osteoprogenitor, and stromal cells necessary to support hematopoiesis. Both adipose and BM contain a population of mesenchymal stromal/stem cells with the potential to differentiate into multiple lineages, including adipogenic, chondrogenic, and osteogenic cells, depending on the culture conditions. In this study we have shown that human adipose-derived stem cells (ASCs) and bone marrow mesenchymal stem cells (BMSCs) populations display a common expression profile for many surface antigens, including CD29, CD49c, CD147, CD166, and HLA-abc. Nevertheless, significant differences were noted in the expression of CD34 and its related protein, PODXL, CD36, CD 49f, CD106, and CD146. Furthermore, ASCs displayed more pronounced adipogenic differentiation capability relative to BMSC based on Oil Red staining (7-fold vs. 2.85-fold induction). In contrast, no difference between the stem cell types was detected for osteogenic differentiation based on Alizarin Red staining. Analysis by RT-PCR demonstrated that both the ASC and BMSC differentiated adipocytes and osteoblast displayed a significant upregulation of lineage-specific mRNAs relative to the undifferentiated cell populations; no significant differences in fold mRNA induction was noted between ASCs and BMSCs. In conclusion, these results demonstrate human ASCs and BMSCs display distinct immunophenotypes based on surface positivity and expression intensity as well as differences in adipogenic differentiation. The findings support the use of both human ASCs and BMSCs for clinical regenerative medicine.


Subject(s)
Adipose Tissue/cytology , Aging/physiology , Bone Marrow Cells/cytology , Immunophenotyping , Tissue Donors , Adipogenesis/genetics , Adult , Bone Marrow Cells/metabolism , Cell Lineage/genetics , Cells, Cultured , Female , Humans , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Osteogenesis/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Stromal Cells/cytology , Stromal Cells/metabolism
11.
Osteoporos Int ; 22(5): 1633-9, 2011 May.
Article in English | MEDLINE | ID: mdl-20593165

ABSTRACT

SUMMARY: This study investigated the influence of ovarian hormone deficiency on core circadian regulatory protein (CCRP) in the context of bone loss. Our data suggest that ovarian hormone deficiency disrupts diurnal rhythmicity and CCRP expression in bone. Further studies should determine if chronobiology provides a novel therapeutic target for osteoporosis intervention. INTRODUCTION: CCRP synchronize metabolic activities and display an oscillatory expression profile in murine bone. In vitro studies using bone marrow mesenchymal stromal/stem cells have demonstrated that the CCRP is present and can be regulated within osteoblast progenitors. In vivo studies have shown that the CCRP regulates bone mass via leptin/neuroendocrine pathways. The current study used an ovariectomized murine model to test the hypothesis that ovarian hormone deficiency is associated with either an attenuation and/or temporal phase shift of the CCRP oscillatory expression in bone and that these changes are correlated with the onset of osteoporosis. METHODS: Sham-operated controls and ovariectomized female C57BL/6 mice were euthanized at 4-h intervals 2 weeks post-operatively. RESULTS: Ovariectomy attenuated the oscillatory expression of CCRP mRNAs in the femur and vertebra relative to the controls and reduced the wheel-running activity profile. CONCLUSION: Ovarian hormone deficiency modulates the expression profile of the CCRP with potential impact on bone marrow mesenchymal stem cell lineage commitment.


Subject(s)
Circadian Rhythm Signaling Peptides and Proteins/biosynthesis , Circadian Rhythm/physiology , Estrogens/physiology , Osteoporosis/physiopathology , Animals , Circadian Rhythm/genetics , Circadian Rhythm Signaling Peptides and Proteins/genetics , Disease Models, Animal , Estrogens/deficiency , Female , Femur/metabolism , Gene Expression Profiling/methods , Gene Expression Regulation/physiology , Lumbar Vertebrae/metabolism , Mice , Mice, Inbred C57BL , Motor Activity/physiology , Osteoporosis/genetics , Osteoporosis/metabolism , Ovariectomy , RNA, Messenger/genetics , Stress, Mechanical , X-Ray Microtomography/methods
12.
Obes Rev ; 10 Suppl 2: 1-5, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19849796

ABSTRACT

This supplement highlights key talks presented at the Pennington Symposium. The collected papers provide a state of the art review of circadian biology at the basic and clinical levels in the context of nutrition, obesity and sleep medicine. Investigators from multiple disciplines attempted to translate new information concerning molecular mechanisms into practical clinical applications, as well as foster new research hypotheses and directions to this exciting field of science and medicine. Furthermore, we hope to spark the interest and attention of the next generation of scientists who will tackle the questions presented by the changing interface between technology, lifestyle and biological rhythms.


Subject(s)
Circadian Rhythm/physiology , Energy Metabolism/physiology , Obesity/etiology , Sleep/physiology , Biological Clocks/physiology , Humans
13.
Obes Rev ; 10 Suppl 2: 46-51, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19849801

ABSTRACT

As the obesity pandemic has accelerated, investigators have begun to explore alternative mechanisms linking circadian biology and sleep to adipose tissue metabolism and obesity. This manuscript reviews recent findings in murine and human models demonstrating the oscillatory expression of the mRNAs encoding the core circadian regulatory proteins in adipose tissue. Comparative transcriptomic analyses of circadian oscillating genes have been used to identify the 'delta sleep-inducing peptide immunoreactor', also known as 'glucocorticoid-induced leucine zipper (GILZ)', as a potential link in this chain. The GILZ gene has been found to differentially regulate stromal stem cell adipogenic and osteogenic differentiation in a reciprocal manner. In adipose and other metabolically active tissues, the circadian oscillation of GILZ expression is subject to entrainment by external stimuli. Together, these observations suggest that GILZ is an attractive candidate for future studies evaluating the role of circadian mechanisms in adipose tissue physiology and pathology.


Subject(s)
Adipogenesis/physiology , Adipose Tissue/metabolism , Circadian Rhythm/physiology , Delta Sleep-Inducing Peptide/metabolism , Leucine Zippers/physiology , Osteogenesis/physiology , Animals , Cell Differentiation/physiology , Delta Sleep-Inducing Peptide/genetics , Gene Expression Regulation , Glucocorticoids/pharmacology , Humans , Leucine Zippers/drug effects , Leucine Zippers/genetics , Mice , Obesity/etiology , Obesity/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Transcription Factors
14.
Int J Obes (Lond) ; 33(9): 971-7, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19597517

ABSTRACT

OBJECTIVE: Circadian mechanisms underlie the physiology of mammals as an adaptation to the earth's rotation on its axis. Highly conserved core circadian regulatory proteins (CCRPs) maintain an oscillatory expression profile in the central and peripheral tissues. The CCRP include both a positive and negative arm, as well as downstream transcriptional regulators. Recent studies in murine models have determined that the mRNAs encoding the CCRP are present in multiple adipose tissue depots and exhibit a robust oscillatory expression profile. This study set out to examine the expression of CCRP mRNAs in human subcutaneous adipose tissues. DESIGN: Retrospective analysis of total RNA isolated from subcutaneous adipose tissue. SUBJECTS: A total of 150 healthy female and male lean (body mass index (BMI) <25), overweight (BMI between 25 and 29.99) or obese (BMI >30) subjects of varied ethnic backgrounds undergoing elective liposuction or surgical procedures. RESULTS: The expression of the CCRP mRNAs displayed a significant correlation between each other and mRNAs representative of adipogenic biomarkers. Hierarchical cluster analyses of mRNAs isolated from the cohort of female Caucasian subjects (n=116) identified three major clusters based on expression of downstream CCRP mRNAs. The mRNAs encoding D site of albumin promoter-binding protein (DBP), E4 promoter-binding protein 4 (E4BP4), PPARgamma coactivator-1beta (PGC-1beta) and Rev-erbalpha were negatively correlated with BMI in a lean cluster (n=66), positively correlated with BMI in a younger overweight/obese cluster (n=19), and not significantly correlated with BMI in an older, overweight/obese cluster (n=31). CONCLUSIONS: These data confirm and extend findings that link the CCRP and circadian mechanisms to the risk of obesity.


Subject(s)
Body Mass Index , Circadian Rhythm Signaling Peptides and Proteins/metabolism , Gene Expression/genetics , RNA, Messenger/metabolism , Subcutaneous Fat/metabolism , Adult , Age Factors , Animals , Basic-Leucine Zipper Transcription Factors/genetics , Basic-Leucine Zipper Transcription Factors/metabolism , Circadian Rhythm Signaling Peptides and Proteins/genetics , Cluster Analysis , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Exodeoxyribonucleases , Female , Humans , Male , Mice , Nuclear Receptor Subfamily 1, Group D, Member 1/genetics , Nuclear Receptor Subfamily 1, Group D, Member 1/metabolism , Retrospective Studies , Trans-Activators/genetics , Trans-Activators/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
15.
J Dent Res ; 88(1): 45-50, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19131316

ABSTRACT

The human body displays central circadian rhythms of activity. Recent findings suggest that peripheral tissues, such as bone, possess their own circadian clocks. Studies have shown that osteocalcin protein levels oscillate over a 24-hour period, yet the specific skeletal sites involved and its transcriptional profile remain unknown. The current study aimed to test the hypothesis that peripheral circadian mechanisms regulate transcription driven by the osteocalcin promoter. Transgenic mice harboring the human osteocalcin promoter linked to a luciferase reporter gene were used. Mice of both genders and various ages were analyzed non-invasively at sequential times throughout 24-hour periods. Statistical analyses of luminescent signal intensity of osteogenic activity from multiple skeletal sites indicated a periodicity of ~ 24 hrs. The maxillomandibular complex displayed the most robust oscillatory pattern. These findings have implications for dental treatments in orthodontics and maxillofacial surgery, as well as for the mechanisms underlying bone remodeling in the maxillomandibular complex.


Subject(s)
Circadian Rhythm/genetics , Mandible/metabolism , Maxilla/metabolism , Osteocalcin/genetics , Animals , Carpal Bones/anatomy & histology , Carpal Bones/metabolism , Female , Gene Expression Regulation/genetics , Half-Life , Humans , Image Processing, Computer-Assisted/methods , Luciferases , Luminescence , Male , Mandible/anatomy & histology , Maxilla/anatomy & histology , Mice , Mice, Transgenic , Models, Animal , Osteogenesis/genetics , Promoter Regions, Genetic/genetics , Sex Factors , Skull/anatomy & histology , Skull/metabolism , Tail/anatomy & histology , Tail/metabolism , Tarsal Bones/anatomy & histology , Tarsal Bones/metabolism , Transcription, Genetic/genetics
16.
J Cell Physiol ; 214(2): 413-21, 2008 Feb.
Article in English | MEDLINE | ID: mdl-17654479

ABSTRACT

Mesenchymal stem-like cells identified in different tissues reside in a perivascular niche. In the present study, we investigated the putative niche of adipose-derived stromal/stem cells (ASCs) using markers, associated with mesenchymal and perivascular cells, including STRO-1, CD146, and 3G5. Immunofluorescence staining of human adipose tissue sections, revealed that STRO-1 and 3G5 co-localized with CD146 to the perivascular regions of blood vessels. FACS was used to determine the capacity of the CD146, 3G5, and STRO-1 specific monoclonal antibodies to isolate clonogenic ASCs from disassociated human adipose tissue. Clonogenic fibroblastic colonies (CFU-F) were found to be enriched in those cell fractions selected with either STRO-1, CD146, or 3G5. Flow cytometric analysis revealed that cultured ASCs exhibited similar phenotypic profiles in relation to their expression of cell surface markers associated with stromal cells (CD44, CD90, CD105, CD106, CD146, CD166, STRO-1, alkaline phosphatase), endothelial cells (CD31, CD105, CD106, CD146, CD166), haematopoietic cells (CD14, CD31, CD45), and perivascular cells (3G5, STRO-1, CD146). The immunoselected ASCs populations maintained their characteristic multipotential properties as shown by their capacity to form Alizarin Red positive mineralized deposits, Oil Red O positive lipid droplets, and Alcian Blue positive proteoglycan-rich matrix in vitro. Furthermore, ASCs cultures established from either STRO-1, 3G5, or CD146 selected cell populations, were all capable of forming ectopic bone when transplanted subcutaneously into NOD/SCID mice. The findings presented here, describe a multipotential stem cell population within adult human adipose tissue, which appear to be intimately associated with perivascular cells surrounding the blood vessels.


Subject(s)
Adipose Tissue/cytology , Phenotype , Pluripotent Stem Cells/cytology , Stromal Cells/cytology , Adipogenesis , Adult , Animals , Antibodies, Monoclonal/metabolism , Biomarkers/metabolism , CD146 Antigen/metabolism , Cell Differentiation , Cell Separation/methods , Cells, Cultured , Chondrogenesis , DNA, Complementary/biosynthesis , Female , Flow Cytometry , Fluorescein-5-isothiocyanate , Fluorescent Antibody Technique, Indirect , Fluorescent Dyes , Humans , Immunohistochemistry , In Vitro Techniques , Male , Mice , Mice, SCID , Osteogenesis , Reverse Transcriptase Polymerase Chain Reaction , Stromal Cells/transplantation , Transplantation, Heterologous
17.
Int J Obes (Lond) ; 32(3): 397-406, 2008 Mar.
Article in English | MEDLINE | ID: mdl-17984979

ABSTRACT

OBJECTIVE: Understanding the regulation of adipocyte differentiation by cellular and extracellular factors is crucial for better management of chronic conditions such as obesity, insulin resistance and lipodystrophy. Experimental infection of rats with a human adenovirus type 36 (Ad-36) improves insulin sensitivity and promotes adipogenesis, reminiscent of the effect of thiozolinediones. Therefore, we investigated the role of Ad-36 as a novel regulator of the adipogenic process. DESIGN AND RESULTS: Even in the absence of adipogenic inducers, infection of 3T3-L1 preadipocytes and human adipose-derived stem cells (hASC) by Ad-36, but not Ad-2 that is another human adenovirus, modulated regulatory points that spanned the entire adipogenic cascade ranging from the upregulation of cAMP, phosphatidylinositol 3-kinase and p38 signaling pathways, downregulation of Wnt10b expression, and increased expression of CCAAT/enhancer binding protein-beta and peroxisome proliferator-activated receptor gamma2 and consequential lipid accumulation. Next, we identified that E4 open reading frame (orf)-1 gene of the virus is necessary and sufficient for Ad-36-induced adipogenesis. Selective knockdown of E4 orf-1 by RNAi abrogated Ad-36-induced adipogenic signaling cascade in 3T3-L1 cells and hASC. Compared to the null vector, selective expression of Ad-36 E4 orf-1 in 3T3-L1 induced adipogenesis, which was abrogated when the PDZ-binding domain of the protein was deleted. CONCLUSION: Thus, Ad-36 E4 orf-1 is a novel inducer of rodent and human adipocyte differentiation process.


Subject(s)
Adenoviruses, Human/genetics , Adipocytes/cytology , Adipogenesis/genetics , Cell Differentiation , Oncogene Proteins, Viral/genetics , 3T3-L1 Cells , Animals , Humans , Mice , Oncogene Proteins, Viral/physiology , Rats
18.
Exp Neurol ; 187(2): 319-28, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15144858

ABSTRACT

Neural tissue has limited capacity for intrinsic repair after injury, and the identification of alternate sources of neuronal stem cells has broad clinical potential. Preliminary studies have demonstrated that adipose-derived adult stromal (ADAS) cells are capable of differentiating into mesenchymal and non-mesenchymal cells in vitro, including cells with select characteristics of neuronal/glial tissue. In this study, we extended these observations to test the hypothesis that murine (mu) ADAS cells can be induced to exhibit characteristics of neuronal and glial tissue by exposure to a cocktail of induction agents. We characterized the differentiation of muADAS cells in vitro using immunohistochemistry and immunoblotting, and examined whether these cells respond to the glutamate agonist N-methyl-D-aspartate (NMDA). We found that induced muADAS cells express proteins indicative of neuronal/glial cells, including nestin, GFAP, S-100, NeuN, MAP2, tau, and beta-III tubulin. Induced muADAS cells express gamma-aminobutyric acid (GABA), the NR-1 and NR-2 subunits of the glutamate receptor, GAP-43, synapsin I, and voltage-gated calcium channels. Finally, induced muADAS cells demonstrate decreased viability in response to NMDA. These findings suggest that muADAS cells can be induced to exhibit several phenotypic, morphologic, and excitotoxic characteristics consistent with developing neuronal and glial tissue.


Subject(s)
Adipose Tissue/cytology , Cell Differentiation/physiology , Neuroglia/cytology , Neurons/cytology , Stromal Cells/cytology , Animals , Antigens, Differentiation/biosynthesis , Blotting, Western , Cell Differentiation/drug effects , Cell Survival/drug effects , Cells, Cultured , Culture Media/pharmacology , Excitatory Amino Acid Agonists/toxicity , Immunohistochemistry , Mice , Mice, Inbred BALB C , N-Methylaspartate/toxicity , Neuroglia/metabolism , Neurons/drug effects , Neurons/metabolism , Stromal Cells/drug effects
19.
Cytotherapy ; 6(1): 7-14, 2004.
Article in English | MEDLINE | ID: mdl-14985162

ABSTRACT

BACKGROUND: Primary cultures of isolated human adipose-derived adult stem (ADAS) cells are multipotent and differentiate in vitro along the adipocyte, chondrocyte, neuronal, osteoblast, and skeletal muscle pathways. METHODS: We examined the ADAS cell yield per unit volume of liposuction tissue, and their surface protein phenotype by flow cytometry. Adipogenesis was assessed by Oil Red O staining and ELISA analysis of leptin secretion. RESULTS: The donor population was 87.5% female (n=18) with a mean age (+/-SD) of 44+/-10 years and body mass index (BMI) of 24.9+/-2.7. The mean cell yield was 404 000+/-206 000 cells per milliliter of lipoaspirate (n=18). Linear regression analysis of the cells derived from the female donors demonstrated a significant negative correlation between the number of cells obtained per milliliter of lipoaspirate with the BMI but not the age of the donor. The undifferentiated ADAS cells were homogeneously positive for the cell-surface markers CD10, CD13, CD29, CD44, CD49e, CD59, CD90, and HLA-ABC, and homogeneously negative for the cell surface markers CD11b, CD45, and HLA-DR. The absence of the panhematopoietic marker, CD45, indicates that the ADAS cells do not derive from circulating BM hematopoietic stem cells. Adipocyte differentiation led to a 5.1-fold increase in Oil Red O staining, and a 196-fold increase in leptin secretion levels. Culture of the cells in the presence of antibiotic and fungizone did not alter the undifferentiated ADAS cell immunophenotype based on flow cytometry, or their adipocyte differentiation based on leptin secretion. DISCUSSION: The ability to isolate a consistently homogeneous population of undifferentiated adult stem cells from adipose tissue of multiple donors supports their potential utility in future tissue-engineering applications.


Subject(s)
Adipose Tissue/cytology , Cell Separation/methods , Lipectomy , Multipotent Stem Cells/cytology , Adipose Tissue/physiology , Adipose Tissue/transplantation , Adult , Aged , Anti-Bacterial Agents/pharmacology , Antigens, CD/analysis , Body Mass Index , Cell Differentiation/immunology , Demography , Female , Humans , Leptin/metabolism , Male , Middle Aged , Multipotent Stem Cells/transplantation
20.
Tissue Eng ; 7(6): 729-41, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11749730

ABSTRACT

Human adipose tissue represents an abundant reservoir of stromal cells with potential utility for tissue engineering. The current study demonstrates the ability of human adipose tissue-derived stromal cells to display some of the hallmarks of osteoblast differentiation in vitro. Following treatment with ascorbate, beta-glycerophosphate, dexamethasone, and 1,25 dihydroxy vitamin D(3), adipose tissue-derived stromal cells mineralize their extracellular matrix based on detection of calcium phosphate deposits using Alizarin Red and von Kossa histochemical stains. Fourier transform infrared analysis demonstrates the apatitic nature of these crystals. Mineralization is accompanied by increased expression or activity of the osteoblast-associated proteins osteocalcin and alkaline phosphatase. These and other osteoblast-associated gene markers are detected based on polymerase chain reaction. In contrast, the adipocyte gene markers--leptin, lipoprotein lipase, and peroxisome proliferator activated receptor gamma2--are reduced under mineralization conditions, consistent with the reciprocal relationship postulated to exist between adipocytes and osteoblasts. The current work supports the presence of a multipotent stromal cell population within human extramedullary adipose tissue. These findings have potential implications for human bone tissue bioengineering.


Subject(s)
Adipose Tissue/cytology , Calcification, Physiologic/physiology , Gene Expression Regulation/physiology , Osteoblasts/physiology , Stromal Cells/physiology , Cell Differentiation/physiology , Extracellular Matrix/metabolism , Humans , Osteoblasts/cytology , Stromal Cells/cytology , Tissue Engineering
SELECTION OF CITATIONS
SEARCH DETAIL
...