Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
1.
Methods Mol Biol ; 2783: 93-107, 2024.
Article in English | MEDLINE | ID: mdl-38478227

ABSTRACT

Murine models of obesity or reduced adiposity are a valuable resource for understanding the role of adipocyte dysfunction in metabolic disorders. Adipose tissue stromal vascular cells or primary adipocytes derived from murine adipose tissue and grown in culture are essential tools for studying the mechanisms underlying adipocyte development and function. Herein, we describe methods for the isolation, expansion, and long-term storage of murine adipose-derived stromal/stem cells, along with protocols for inducing adipogenesis to white or beige adipocytes in this cell population and osteogenic differentiation. Isolation of the adipose stromal vascular fraction cells for flow cytometric analysis is also described.


Subject(s)
Adipogenesis , Adiposity , Mice , Humans , Animals , Flow Cytometry/methods , Osteogenesis , Adipocytes , Adipose Tissue , Cell Differentiation , Obesity/metabolism , Stem Cells
2.
J Cell Physiol ; 238(6): 1368-1380, 2023 06.
Article in English | MEDLINE | ID: mdl-37021796

ABSTRACT

Human mesenchymal stem cells (hMSCs) are the cornerstone of regenerative medicine; large quantities of hMSCs are required via in vitro expansion to meet therapeutic purposes. However, hMSCs quickly lose their osteogenic differentiation potential during in vitro expansion, which is a major roadblock to their clinical applications. In this study, we found that the osteogenic differentiation potential of human bone marrow stem cells (hBMSCs), dental pulp stem cells (hDPSCs), and adipose stem cells (hASCs) was severely impaired after in vitro expansion. To clarify the molecular mechanism underlying this in vitro expansion-related loss of osteogenic capacity in hMSCs, the transcriptome changes following in vitro expansion of these hMSCs were compared. Cysteine-rich secretory protein LCCL domain-containing 2 (CRISPLD2) was identified as the most downregulated gene shared by late passage hBMSCs, hDPSCs, and hASCs. Both the secreted and non-secreted CRISPLD2 proteins progressively declined in hMSCs during in vitro expansion when the cells gradually lost their osteogenic potential. We thus hypothesized that the expression of CRISPLD2 is critical for hMSCs to maintain their osteogenic differentiation potential during in vitro expansion. Our studies showed that the knockdown of CRISPLD2 in early passage hBMSCs inhibited the cells' osteogenic differentiation in a siRNA dose-dependent manner. Transcriptome analysis and immunoblotting indicated that the CRISPLD2 knockdown-induced osteogenesis suppression might be attributed to the downregulation of matrix metallopeptidase 1 (MMP1) and forkhead box Q1 (FOXQ1). Furthermore, adeno-associated virus (AAV)-mediated CRISPLD2 overexpression could somewhat rescue the impaired osteogenic differentiation of hBMSCs during in vitro expansion. These results revealed that the downregulation of CRISPLD2 contributes to the impaired osteogenic differentiation of hMSCs during in vitro expansion. Our findings shed light on understanding the loss of osteogenic differentiation in hMSCs and provide a potential therapeutic target gene for bone-related diseases.


Subject(s)
Bone Diseases , Mesenchymal Stem Cells , Humans , Osteogenesis/genetics , Mesenchymal Stem Cells/metabolism , Cell Differentiation/genetics , RNA, Small Interfering/metabolism , Bone Diseases/metabolism , Cells, Cultured , Forkhead Transcription Factors/metabolism , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/metabolism , Interferon Regulatory Factors/metabolism
3.
Front Vet Sci ; 7: 575199, 2020.
Article in English | MEDLINE | ID: mdl-33251261

ABSTRACT

Background: Chronic myelogenous leukemia (CML) is a clonal proliferative disorder of the myeloid, megakaryocyte, and erythroid lineages. The onset and subsequent progression of CML is well-described in humans. There is comparably little information surrounding CML progression in veterinary species, including Yucatan miniature swine that are common for preclinical pharmaceutical and device testing. In humans, more than 90% of CML cases are associated with a chromosomal translocation that results in the Philadelphia gene (BCR/ABL mutation). In this report, the presence of the Philadelphia gene in a Yucatan burrow was confirmed in white blood cells collected prior to onset of clinical signs with primers designed from the human BCR/ABL sequence. Case Presentation: A 24 month old, 70 kg, Yucatan barrow received a prefabricated bovine cortical bone xenograft following a unilateral zygomatic ostectomy for a preclinical study. Complete blood count and serum chemistries were performed prior to and 28, 53, 106, and 129 days after facial surgery. Fifty three days after surgery, a bone marrow biopsy was performed due to anorexia, severe basophilia, and mild anemia. A finding of a moderate increase in basophilic precursors in bone marrow cytology was followed by lymphocyte immunophenotyping via flow cytometry and RT-PCR amplification of the Philadelphia gene in white blood cell samples from the affected barrow and an unaffected barrow in the same treatment group. Bone marrow, lymph node, liver, spleen, lung, kidney, and adrenal gland lesions of mostly myeloblasts were identified after the affected barrow died 146 days after surgery. Flow cytometry confirmed lymphopenia and suggested basophilia, and RT-PCR established the presence of the BCR/ABL gene. Conclusions: The information in this report confirms the presence of the BCR/ABL mutation and documents progression of chronic myelogenous (basophilic) leukemia from a chronic phase to a terminal blast crisis in an adult Yucatan barrow. The natural occurrence and progression of CML associated with the BCR/ABL mutation in miniature swine establishes potential for future porcine models of human CML. The information also establishes a genetic test to confirm porcine CML to prevent inadvertent attribution of clinical signs to treatment complications during preclinical testing.

4.
Cells ; 8(7)2019 07 15.
Article in English | MEDLINE | ID: mdl-31311198

ABSTRACT

INTRODUCTION: Adipose derived stromal/stem cells (ASCs) hold potential as cell therapeutics for a wide range of disease states; however, many expansion protocols rely on the use of fetal bovine serum (FBS) as a cell culture nutrient supplement. The current study explores the substitution of lysates from expired human platelets (HPLs) as an FBS substitute. METHODS: Expired human platelets from an authorized blood center were lysed by freeze/thawing and used to examine human ASCs with respect to proliferation using hematocytometer cell counts, colony forming unit-fibroblast (CFU-F) frequency, surface immunophenotype by flow cytometry, and tri-lineage (adipocyte, chondrocyte, osteoblast) differentiation potential by histochemical staining. RESULTS: The proliferation assays demonstrated that HPLs supported ASC proliferation in a concentration dependent manner, reaching levels that exceeded that observed in the presence of 10% FBS. The concentration of 0.75% HPLs was equivalent to 10% FBS when utilized in cell culture media with respect to proliferation, immunophenotype, and CFU-F frequency. When added to osteogenic, adipogenic, and chondrogenic differentiation media, both supplements showed appropriate differentiation by staining. CONCLUSION: HPLs is an effective substitute for FBS in the culture, expansion and differentiation of human ASCs suitable for pre-clinical studies; however, additional assays and analyses will be necessary to validate HPLs for clinical applications and regulatory approval.


Subject(s)
Adipose Tissue/cytology , Blood Platelets/chemistry , Cell Differentiation , Culture Media, Serum-Free/chemistry , Mesenchymal Stem Cells/cytology , Animals , Cattle , Cell Proliferation , Cells, Cultured , Culture Media, Serum-Free/pharmacology , Humans , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/physiology , Serum/chemistry
5.
Obesity (Silver Spring) ; 26(9): 1439-1447, 2018 09.
Article in English | MEDLINE | ID: mdl-30226002

ABSTRACT

OBJECTIVE: This study examined the phenotypic effects of adipocyte-specific oncostatin M receptor (OSMR) loss in chow-fed mice. METHODS: Chow-fed adipocyte-specific OSMR knockout (FKO) mice and littermate OSMRfl/fl controls were studied. Tissue weights, insulin sensitivity, adipokine production, and stromal cell immunophenotypes were assessed in epididymal fat (eWAT); serum adipokine production was also assessed. In vitro, adipocytes were treated with oncostatin M, and adipokine gene expression was assessed. RESULTS: Body weights, fasting blood glucose levels, and eWAT weights did not differ between genotypes. However, the eWAT of OSMRFKO mice was modestly less responsive to insulin stimulation than that of OSMRfl/fl mice. Notably, significant increases in adipokines, including C-reactive protein, lipocalin 2, intercellular adhesion molecule-1, and insulinlike growth factor binding protein 6, were observed in the eWAT of OSMRFKO mice. In addition, significant increases in fetuin A and intercellular adhesion molecule-1 were detected in OSMRFKO serum. Flow cytometry revealed a significant increase in leukocyte number and modest, but not statistically significant, increases in B cells and T cells in the eWAT of OSMRFKO mice. CONCLUSIONS: The chow-fed OSMRFKO mice exhibited adipose tissue dysfunction and increased proinflammatory adipokine production. These results suggest that intact adipocyte oncostatin M-OSMR signaling is necessary for adipose tissue immune cell homeostasis.


Subject(s)
Adipocytes/metabolism , Adipose Tissue/physiopathology , Oncostatin M/metabolism , Animals , Male , Mice , Mice, Knockout
6.
Methods Mol Biol ; 1773: 137-146, 2018.
Article in English | MEDLINE | ID: mdl-29687386

ABSTRACT

Murine models of obesity or reduced adiposity are a valuable resource for understanding the role of adipocyte dysfunction in metabolic disorders. Adipose tissue stromal vascular cells or primary adipocytes derived from murine adipose tissue and grown in culture are essential tools for studying the mechanisms underlying adipocyte development and function. Herein, we describe methods for the isolation, expansion, and long-term storage of murine adipose-derived stromal/stem cells along with protocols for inducing adipogenesis in this cell population or isolating the adipose stromal vascular fraction cells for flow cytometric analysis.


Subject(s)
Adipocytes/cytology , Adipocytes/immunology , Adipogenesis/immunology , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/immunology , Adipocytes/drug effects , Adipogenesis/drug effects , Adipose Tissue/cytology , Adipose Tissue/drug effects , Adipose Tissue/immunology , Animals , Collagenases/pharmacology , Female , Flow Cytometry , Humans , Male , Mesenchymal Stem Cells/drug effects , Mice , Rosiglitazone/pharmacology , Time Factors
7.
Int J Vitam Nutr Res ; 87(1-2): 49-58, 2017 Mar.
Article in English | MEDLINE | ID: mdl-30010516

ABSTRACT

It has been determined that individuals who are regularly physically active have more favorable inflammatory profiles; less is known about how vitamin D levels can impact inflammation. This study explored the relationship between inflammatory indices in physically active (PA) and not physically active (NPA) individuals with 25-hydroxyvitamin D (25OHD) concentrations either above or below optimal concentrations. All female subjects (n = 63, age 19 - 35 years) were evaluated for body composition, maximal aerobic capacity (VO2peak), and anaerobic power (Wingate). Blood samples were analyzed for 25OHD and C-reactive protein (CRP), stimulated with lipopolysaccharide (LPS) and assessed for interleukin-6 (IL-6) production, and used for flow cytometric analysis. PA (n = 30) had higher 25OHD levels (45.2 ± 2.7 vs. 17.05 ± 1.4 ng / mL; p = 0.015), higher VO2peak (p < 0.0001), lower body weight (p = 0.039) and lower estimated percent body fat (p = 0.011) compared to NPA (n = 33). PA also had lower LPS-stimulated IL-6 production compared to NPA (p = 0.0163), although there were no differences between resting CRP concentrations. NPA with optimal 25OHD had fewer total monocytes, CD14+CD16-cells, CD14+CD16+ cells, and decreased TLR4 expression on CD14+CD16+ cells compared to NPA with suboptimal 25OHD (< 32 ng / mL). In summary, regular physical activity was associated with higher serum 25OHD, healthier measures of body composition, and reduced stimulated IL-6 production. However, optimal vitamin D status was not associated with anti-inflammatory benefits beyond those which are provided by regular physical activity.

8.
Cells Tissues Organs ; 201(6): 436-444, 2015.
Article in English | MEDLINE | ID: mdl-27310337

ABSTRACT

The capability of multipotent mesenchymal stem cells to maintain cell viability, phenotype and differentiation ability upon thawing is critical if they are to be banked and used for future therapeutic purposes. In the present study, we examined the effect of 9-10 months of cryostorage on the morphology, immunophenotype, colony-forming unit (CFU) and differentiation capacity of fresh and cryopreserved human adipose-derived stromal/stem cells (ASCs) from the same donors. Cryopreservation did not reduce the CFU frequency and the expression levels of CD29, CD73, CD90 and CD105 remained unchanged with the exception of CD34 and CD45; however, the differentiation capacity of cryopreserved ASCs relative to fresh cells was significantly reduced. While our findings suggest that future studies are warranted to improve cryopreservation methods and agents, cryopreserved ASCs retain sufficient features to ensure their practical utility for both research and clinical applications.

9.
Stem Cell Rev Rep ; 10(4): 600-11, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24850472

ABSTRACT

Low yield of adult adipose-derived multipotent stromal cells (ASC) can limit autologous cell therapy in individuals with minimal adipose tissue. In this study, ASC isolation was optimized from approximately 0.2 g of feline epididymal adipose tissue for a treatment dose of 10(6)-10(7) ASCs/kg. The ASC yield was determined for three digestions, 0.1 % collagenase in medium for 30 min (Classic), 0.3 % collagenase in buffer for 30 min (New) and 0.3 % collagenase in buffer for 1 h (Hour). After isolation by the new tissue digestion, continuously cultured ASCs (fresh) and cells recovered and expanded after cryostorage at P0 (revitalized) were characterized up to cell passage (P) 5. Outcomes included CD9, CD29, CD44, CD90 and CD105 expression, cell doublings and doubling times, fibroblastic, adipogenic and osteogenic colony forming unit (CFU) frequency percentages and lineage-specific target gene expression after induction. The New digestion had the highest CFU yield, and about 7x10(6) ASCs/kg were available within three cell passages (P2). Compared to earlier passages, target surface antigen expression was lowest in fresh P5 cells, and fresh and revitalized P3-5 cells had slower expansion. Fresh and revitalized P1 ASCs had higher CFU frequency percentages and lineage-specific gene expression than P3. The New method described in this study was most efficient for feline epididymal ASC isolation and did not alter in vitro cell behavior. Fresh and revitalized P0-P2 feline ASCs may be most effective for preclinical and clinical trials. This study offers a potential option for ASC isolation from limited adipose tissue resources across species.


Subject(s)
Adipose Tissue/cytology , Biomarkers/metabolism , Cell Differentiation , Cell Lineage , Multipotent Stem Cells/cytology , Stromal Cells/cytology , Adipose Tissue/metabolism , Animals , Cats , Cells, Cultured , Immunophenotyping , Male , Multipotent Stem Cells/metabolism , Stromal Cells/metabolism
10.
J Tissue Eng Regen Med ; 8(10): 757-62, 2014 Oct.
Article in English | MEDLINE | ID: mdl-22807102

ABSTRACT

Since inflammatory mechanisms have been postulated to link obesity to osteoarthritis, the current study evaluated the ratio of immune cells to multipotent stromal cells within the infrapatellar fat pad (IPFP) and subcutaneous adipose tissue (SQ) of the knee; each depot has potential as a source of regenerative cells. The immunophenotypes of stromal vascular fraction (SVF) and adipose-derived stem cells (ASCs) of the IPFP and SQ were determined in tissues from osteoarthritic subjects (n = 7) undergoing total knee replacement. Based on a subset of surface antigens, the immunophenotype of ASCs from SQ of OA subjects was not significantly different from that of relatively healthy and leaner subjects undergoing elective liposuction surgery. Flow-cytometry comparison of SVF cell populations in the IPFP of OA subjects resembled those within the subject's own matched SQ, with the exception of the endothelial marker CD31(+) , which was significantly greater in cells from SQ. In the OA subjects, lower numbers of capillary-like structures and higher numbers of stromal and alkaline phosphatase colony-forming units in the IPFP vs SQ were consistent with this finding; however, ASCs from both depots in OA subjects exhibited comparable adipogenic and osteogenic differentiation potential. Thus, the IPFP contains an ASC and immune cell population similar to that of donor-matched SQ, making it an alternative ASC source for tissue regeneration. Further studies will be needed to determine whether IPFP immune cell infiltrates play an aetiological role in osteoarthritis equivalent to that shown in diabetes associated with obesity.


Subject(s)
Endothelial Cells , Mesenchymal Stem Cells , Osteoarthritis, Knee , Pluripotent Stem Cells , Subcutaneous Fat , Adult , Aged , Aged, 80 and over , Arthroplasty, Replacement, Knee , Endothelial Cells/metabolism , Endothelial Cells/pathology , Female , Flow Cytometry , Humans , Male , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/pathology , Middle Aged , Osteoarthritis, Knee/metabolism , Osteoarthritis, Knee/pathology , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Pluripotent Stem Cells/metabolism , Pluripotent Stem Cells/pathology , Subcutaneous Fat/metabolism , Subcutaneous Fat/pathology
11.
Cells Tissues Organs ; 200(3-4): 204-11, 2014.
Article in English | MEDLINE | ID: mdl-26089088

ABSTRACT

The emerging field of regenerative medicine has identified adipose tissue as an abundant source of stromal/stem cells for tissue engineering applications. Therefore, we have compared the differentiation and immunophenotypic features of adipose-derived stromal/stem cells (ASC) isolated from either omental or subcutaneous adipose depots. Human tissue samples were obtained from bariatric and plastic surgical practices at a university-affiliated teaching hospital and a private practice, respectively, with informed patient consent. Primary cultures of human ASC were isolated from adipose specimens within 24 h of surgery and culture expanded in vitro. The passaged ASC were induced to undergo adipogenic or osteogenic differentiation as assessed by histochemical methods or evaluated for surface antigen expression profiles by flow cytometry. ASC yields per unit weight of tissue were comparable between omental and subcutaneous depots. At passage 0, the immunophenotype of omental and subcutaneous ASC were not significantly different with the exception of CD105 and endoglin, a component of the transforming growth factor ß receptor. The adipogenic differentiation of omental ASC was less robust than that of subcutaneous ASC based on in vitro histochemical and PCR assays. Although the yield and immunophenotype of ASC from omental adipose depots resembled that of subcutaneous ASC, omental ASC displayed significantly reduced adipogenic differentiation capacity following chemical induction. Further studies are necessary to evaluate and optimize the differentiation function of omental ASC in vitro and in vivo. Pending such analyses, omental ASC should not be used interchangeably with subcutaneous ASC for regenerative medical applications.


Subject(s)
Cell Differentiation , Cell Separation/methods , Immunophenotyping , Omentum/cytology , Stem Cells/cytology , Subcutaneous Fat/cytology , Adipogenesis/genetics , Adult , Biomarkers/metabolism , Demography , Female , Flow Cytometry , Humans , Male , Middle Aged , Osteogenesis/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Stem Cells/metabolism , Stromal Cells/cytology , Stromal Cells/metabolism , Tissue Donors
12.
Cytotherapy ; 15(8): 979-85, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23725689

ABSTRACT

BACKGROUND AIMS: The isolation of human adipose stromal/stem cells (ASCs) currently relies on the use of the enzyme collagenase, which digests the triple helix region of peptide bonds in the collagen of adipose tissue. Collagenase is an expensive reagent derived from a bacterial source, and its use in isolating ASCs is a time-consuming procedure. This experiment evaluated the extraction of ASCs without an enzymatic digest. METHODS: We used a simple method of washing adipose tissue to isolate and characterize the cells and compared this method with the enzymatic procedure in terms of processing time, stem cell yield, differentiation potential and immunophenotype. RESULTS: Based on fluorescence activated cell sorting analysis, the stromal vascular fractions isolated with the washing method displayed a distinct and potentially favorable immunophenotype relative to the collagenase digestion. This difference may reflect the absence of chemical alteration of the cells by collagenase digestion. Independent of the isolation procedure, the resulting passaged ASCs were comparable based on immunophenotype and adipogenic and osteogenic differentiation potential. CONCLUSIONS: Although using collagenase substantially increases cell yield, the two methods yield a similar cell product.


Subject(s)
Adipocytes/cytology , Adipose Tissue/cytology , Cell Separation/methods , Mesenchymal Stem Cells/cytology , Adult , Cell Differentiation , Cell Proliferation , Cells, Cultured , Collagenases/chemistry , Female , Flow Cytometry , Humans , Immunophenotyping , Male , Osteogenesis , Tissue Engineering
13.
Vet Surg ; 42(2): 137-46, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23373667

ABSTRACT

OBJECTIVE: To identify the optimum intra-articular multipotent stromal cell (MSC) tissue source in the canine stifle. STUDY DESIGN: Experimental. SAMPLE POPULATION: Infrapatellar adipose tissue, synovium lining the joint capsule, and synovium surrounding the cranial cruciate ligament (CrCL) from normal stifles of 6 dogs. METHODS: Nucleated cell density for each tissue was determined, and cell doublings (CD) and doubling times (DT) were quantified for expansion rates. Adipogenic, osteogenic, and chondrogenic differentiation was confirmed with light microscopy. Fibroblastic, adipogenic, and osteogenic colony forming unit frequencies were determined for multipotentiality. Tissue-specific target gene expression was assessed, and percentages of CD29(+) , CD34(+) , CD44(+) , CD45(+) , and CD90(+) cells quantified. RESULTS: Adipose tissue had the highest MSC density (ASC). The CD decreased with increasing passages for all cell types, and ASC values tended to be higher. Multipotentiality decreased with passage, but remained highest in ASC. Tissue-specific target gene expression was higher in induced versus noninduced cells, and ASCs had the highest upregulation across passages. Most cells were CD29(+) , CD44(+) , CD90(+) , and percentages decreased with passage. Within cell types, there were more CD29(+) ASC in early passages and more CD44(+) and CD90(+) ASC in later passages. CONCLUSIONS: ASC had the highest in vitro expansion rates, CFU frequencies, tissue-specific target gene expression, and percentages of MSC immunophenotypes.


Subject(s)
Adipose Tissue/cytology , Antigens, CD/physiology , Multipotent Stem Cells/physiology , Stromal Cells/cytology , Animals , Anterior Cruciate Ligament/cytology , Antigens, CD/immunology , Antigens, CD34/physiology , Cell Count/veterinary , Cell Division/physiology , Dogs , Female , Hyaluronan Receptors/physiology , Immunophenotyping/veterinary , Integrin beta1/physiology , Leukocyte Common Antigens/physiology , Multipotent Stem Cells/cytology , Multipotent Stem Cells/immunology , Stifle/cytology , Stromal Cells/immunology , Stromal Cells/physiology , Thy-1 Antigens/physiology
14.
J Appl Physiol (1985) ; 113(3): 393-400, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22678961

ABSTRACT

The existing paradigm of exercise-induced decreases in chronic inflammation focuses on the expression of inflammatory receptors on systemic monocytes in response to exercise training, with the role of anti-inflammatory receptors largely ignored. Our recent preliminary studies indicate that the anti-inflammatory melanocortin receptors (MCRs) may play a role in modulating exercise-induced decreases in chronic inflammation. Here, we present a study designed to determine the effect of intense, resistance exercise training on systemic monocyte MCR expression. Because low-grade chronic inflammation is associated with elevated cardiometabolic risk in healthy populations and exercise decreases chronic inflammation, we investigated the associations between systemic monocyte cell surface expression of MCRs and inflammatory markers as a possible mechanism for the beneficial anti-inflammatory effects of resistance training. To this end, the present study includes 40 adults (aged 19-27 yr) and implements a 12-wk periodized, intensive resistance training intervention. Melanocortin 1 and 3 receptor expression on systemic monocytes and inflammatory markers, including C-reactive protein (CRP), interleukin (IL)-6, IL-1ß, and IL-10, were measured before and after the intervention. Resistance training significantly altered MCR systemic monocyte cell surface expression, had no chronic effects on IL-6, IL-1ß, or IL-10 expression, but significantly decreased CRP levels from a moderate to a low cardiovascular disease risk category. More specifically, decreased melanocortin 3 receptor expression significantly correlated with decreased CRP, independent of changes in adiposity. These data suggest that the observed responses in MCR expression and decreases in cardiovascular disease risk in response to resistance training represent an important anti-inflammatory mechanism in regulating exercise-induced decreases in chronic inflammation that occur independent of chronic changes in systemic cytokines.


Subject(s)
C-Reactive Protein/physiology , Receptor, Melanocortin, Type 1/biosynthesis , Receptor, Melanocortin, Type 3/biosynthesis , Resistance Training , Adult , C-Reactive Protein/analysis , Female , Humans , Interleukins/blood , Male , Monocytes/chemistry , Young Adult
15.
Stem Cells Dev ; 21(2): 249-59, 2012 Jan 20.
Article in English | MEDLINE | ID: mdl-21510817

ABSTRACT

Stem Cell Antigen-1 (Sca-1) is a member of the lymphocyte-activated protein 6 family and has served as a marker for the identification of stem cells in various tissues, including fat depots. In vitro and in vivo studies suggest the possible involvement of Sca-1 in adipogenic differentiation and link Sca-1 antigenicity with adipocyte progenitors. Previously, we showed that Sca-1-enriched populations of ear mesenchymal stem cells possess enhanced capacity to differentiate into adipocytes. Additionally, we determined the natural frequency and localization of Sca-1-positive progenitor/stem cells in brown and white fat in situ. The present study addressed the question whether Sca-1 deficiency alters the white adipose tissue response to a high-saturated-fat diet. Our results show that Sca-1 null mice (Sca-1(-/-)) fed high-fat diet developed obesity equally well as wild-type mice, suggesting either an indirect in vivo effect of Sca-1 or a compensatory response to Sca-1 deficiency. However, contrary to wild-type mice, high fat diet-fed Sca-1(-/-) mice showed no alterations in serum adipocytokines. The data lead to the conclusion that Sca-1 is either redundant or a nonessential marker of adipose progenitor/stem cells. Nevertheless, since Sca-1-deficient mice displayed elevated blood glucose at fasting and exhibited glucose intolerance and insulin resistance, Sca-1 has subtle effects on adipose function. Thus, the Sca-1-deficient mice may provide a useful model for metabolic studies.


Subject(s)
Adipocytes/metabolism , Adipose Tissue, White/metabolism , Adiposity/genetics , Diet, High-Fat , Membrane Proteins/deficiency , Mesenchymal Stem Cells/metabolism , Obesity/blood , Adipocytes/cytology , Adipokines/blood , Adipose Tissue, White/cytology , Animals , Antigens, Ly/genetics , Blood Glucose , Gene Knockout Techniques , Insulin/blood , Insulin Resistance , Male , Membrane Proteins/genetics , Mesenchymal Stem Cells/cytology , Mice , Mice, Inbred C57BL , Mice, Knockout , Obesity/genetics
16.
Cytotherapy ; 13(5): 594-7, 2011 May.
Article in English | MEDLINE | ID: mdl-21198335

ABSTRACT

Adherent adipose-derived stromal/stem cells (ASC) have been used in pre-clinical regenerative medical studies applied to a broad range of tissues with an ultimate goal of translating these findings to clinical safety and efficacy testing; however, many protocols passage the cells using porcine-derived trypsin. We have compared porcine trypsin with animal protein-free products from recombinant bacteria (TrypLE Express; Invitrogene) and corn (TrypZean; Sigma) based on cell yield, viability and immunophenotype. ASC harvested with each trypsin product were comparable.


Subject(s)
Adipose Tissue/cytology , Cell Culture Techniques , Cell Separation , Mesenchymal Stem Cells/cytology , Recombinant Proteins/chemistry , Trypsin/chemistry , Adipose Tissue/physiology , Adult , Animals , Cell Count , Cell Differentiation , Cell Survival , Cells, Cultured , Culture Media , Female , Humans , Immunophenotyping , Mesenchymal Stem Cells/physiology , Middle Aged , Stromal Cells/cytology , Stromal Cells/physiology , Swine , Young Adult
17.
Cytotherapy ; 12(4): 538-46, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20380539

ABSTRACT

BACKGROUND AIMS: Adipose-derived stromal/stem cells (ASC) capable of multipotential differentiation can be isolated with high yields from human subcutaneous lipoaspirates. This study reports our recent experience of isolating and immunophenotypically characterizing ASC from >60 human patients with a mean age of 43.6 and body mass index (BMI) of 27. METHODS: We examined the ASC yield per unit volume of lipoaspirate tissue, the surface antigen profile based on flow cytometry, histochemical differentiation potential along the adipogenic and osteogenic pathways, and expression of adipogenic mRNA by transcriptomic microarray and reverse transcription (RT)-polymerase chain reaction (PCR). RESULTS: The population (n = 64) of predominantly Caucasian (84.3%) female (90.6%) donors had a mean age of 43.6 +/- 11.1 years and a mean BMI of 27.0 +/- 3.8. A yield of 375 +/- 142 x 10(3) ASC was obtained per milliliter of lipoaspirate within a 4.1 +/- 0.7-day culture period (n = 62). The ASC population was uniformly CD29(+) CD34(+) CD44(lo) CD45(lo) CD73(+) CD90(+) CD105(+) and capable of undergoing both adipogenesis and osteogenesis in vitro based on Oil Red O and Alizarin Red staining, respectively. Adipogenic differentiation was associated with a significant induction of multiple mRNA associated with lipid storage and synthesis based on microarray analysis of n = 3 donors. During an adipogenic differentiation time-course, representative mRNA (adiponectin, C/EBPalpha, leptin and LPL) displayed increases of several orders of magnitude. CONCLUSIONS: These findings demonstrate the reproducibility of subcutaneous lipoaspirates as a consistent and abundant source of functional ASC from donors across a spectrum of ages and BMI. These results have relevance for regenerative medical applications exploiting autologous and allogeneic ASC for soft and hard tissue engineering.


Subject(s)
Mesenchymal Stem Cell Transplantation , Multipotent Stem Cells/physiology , Osteogenesis , RNA, Messenger/analysis , Subcutaneous Fat/cytology , Adipogenesis/physiology , Adult , Body Mass Index , Cell Separation , Female , Flow Cytometry , Guided Tissue Regeneration , Humans , Lipectomy , Lipid Metabolism/genetics , Male , Middle Aged , Multipotent Stem Cells/cytology , Oligonucleotide Array Sequence Analysis , Osteogenesis/physiology , Stem Cell Niche , Stromal Cells/cytology , Stromal Cells/physiology , Subcutaneous Fat/surgery
18.
Biochem Biophys Res Commun ; 378(3): 539-44, 2009 Jan 16.
Article in English | MEDLINE | ID: mdl-19056354

ABSTRACT

This study has determined the natural frequency and localization of progenitor/stem cells within fat depots in situ based on their ability to retain DNA nucleotide label (BrdU). Neonate and mature male C57BL6/J mice were injected intraperitoneally with BrdU- and label-retaining cells (LRC) were quantified in fat depots by immunohistochemical, immunofluorescent, and flow cytometric methods. In neonates, LRC constituted 27% of the cells in inguinal fat (iWAT) and 65% in interscapular brown fat (BAT) after Day 10 and 26% of the cells in epididymal fat (eWAT) after Day 28. After 52 days, the LRC accounted for 0.72% of iWAT, 0.53% of eWAT and 1.05% of BAT, respectively. The BrdU-labeled cells localized to two areas: single cells distributed among adipocytes or those adjacent to the blood vessels wall. In mature C57BL6/J mice, flow cytometric analysis determined that a majority of the LRC were also positive for stem cell antigen-1 (Sca-1).


Subject(s)
Adipose Tissue, Brown/cytology , Adipose Tissue, White/cytology , Bromodeoxyuridine/analysis , Epididymis/cytology , Stem Cells/cytology , Adipose Tissue, Brown/metabolism , Adipose Tissue, White/metabolism , Animals , Antigens, Ly/analysis , Antigens, Ly/metabolism , Bromodeoxyuridine/metabolism , Epididymis/metabolism , Flow Cytometry , Immunohistochemistry , Male , Membrane Proteins/analysis , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Staining and Labeling
19.
Bone ; 42(5): 861-70, 2008 May.
Article in English | MEDLINE | ID: mdl-18302991

ABSTRACT

A core group of regulatory factors control circadian rhythms in mammalian cells. While the suprachiasmatic nucleus in the brain serves as the central core circadian oscillator, circadian clocks also exist within peripheral tissues and cells. A growing body of evidence has demonstrated that >20% of expressed mRNAs in bone and adipose tissues oscillate in a circadian manner. The current manuscript reports evidence of the core circadian transcriptional apparatus within primary cultures of murine and human bone marrow-derived mesenchymal stem cells (BMSCs). Exposure of confluent, quiescent BMSCs to dexamethasone synchronized the oscillating expression of the mRNAs encoding the albumin D binding protein (dbp), brain-muscle arnt-like 1 (bmal1), period 3 (per3), rev-erb alpha (Rev A), and rev-erb beta (Rev B). The genes displayed a mean oscillatory period of 22.2 to 24.3 h. The acrophase or peak expression of mRNAs encoding "positive" (bmal1) and "negative" (per3) components of the circadian regulatory apparatus were out of phase with each other by approximately 8-12 h, consistent with in vivo observations. In vivo, phosphyrylation by glycogen synthase kinase 3beta (GSK3beta) is known to regulate the turnover of per3 and components of the core circadian regulatory apparatus. In vitro addition of lithium chloride, a GSK3beta inhibitor, significantly shifted the acrophase of all genes by 4.2-4.7 h oscillation in BMSCs; however, only the male murine BMSCs displayed a significant increase in the length of the period of oscillation. We conclude that human and murine BMSCs represent a valid in vitro model for the analysis of circadian mechanisms in bone metabolism and stem cell biology.


Subject(s)
Circadian Rhythm/physiology , Dexamethasone/pharmacology , Gene Expression/drug effects , Mesenchymal Stem Cells/drug effects , ARNTL Transcription Factors , Adult , Animals , Antigens, CD/analysis , Basic Helix-Loop-Helix Transcription Factors/genetics , Bone Marrow Cells/cytology , Bone Marrow Cells/drug effects , Bone Marrow Cells/metabolism , Cell Cycle Proteins/genetics , DNA-Binding Proteins/genetics , Female , Humans , Lithium Chloride/pharmacology , Male , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Mice , Mice, Inbred C57BL , Nuclear Proteins/genetics , Nuclear Receptor Subfamily 1, Group D, Member 1 , Period Circadian Proteins , Receptors, Cytoplasmic and Nuclear/genetics , Repressor Proteins/genetics , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factors/genetics
20.
Vet Immunol Immunopathol ; 119(3-4): 180-8, 2007 Oct 15.
Article in English | MEDLINE | ID: mdl-17658617

ABSTRACT

Lymphocyte proliferation and peripheral blood mononuclear cell (PBMC) production of PGE(2) were assayed in 15 healthy dogs fed a basal diet supplemented with either sunflower oil (Group Sunflower oil), sunflower oil and menhaden fish oil (Group Fish oil), or sunflower oil and menhaden fish oil plus alpha-tocopherol acetate for 12 weeks (Group Fish oil + E). Lymphocyte proliferation was determined by a flow cytometric technique utilizing the fluorochrome carboxyfluorescein diacetate succinimidyl ester (CFSE). The PBMC supernatant PGE(2) concentration was assayed using a competitive enzyme-linked immunoassay. Group Fish oil had a significant decrease in lymphocyte proliferation at week 12. PBMC production of PGE(2) was decreased in all three groups but only significantly reduced in groups receiving fish oil supplementation. Based on these results, this level of fish oil supplementation appears to suppress the lymphoproliferative response in healthy, young dogs but this response can be attenuated by high levels of dietary vitamin E supplementation. Furthermore, fish oil-induced reduction in lymphocyte proliferation appears to manifest through a PGE(2)-independent mechanism and is not associated with increased lipid peroxidation.


Subject(s)
Cell Proliferation/drug effects , Diet/veterinary , Dogs , Fish Oils/pharmacology , Flow Cytometry/methods , Lymphocytes/drug effects , Vitamin E/pharmacology , Animal Feed , Animal Nutritional Physiological Phenomena , Animals , Dietary Supplements , Dinoprostone/metabolism , Female , Immunoenzyme Techniques , Interleukin-2/metabolism , Lymphocytes/cytology , Lymphocytes/immunology , Male , Staining and Labeling
SELECTION OF CITATIONS
SEARCH DETAIL