Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 103
Filter
2.
Amino Acids ; 53(3): 381-393, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33598768

ABSTRACT

While it has been hypothesized that brown adipocytes responsible for mammalian thermogenesis are absent in birds, the existence of beige fat has yet to be studied directly. The present study tests the hypothesis that beige fat emerges in birds as a mechanism of physiological adaptation to cold environments. Subcutaneous neck adipose tissue from cold-acclimated or triiodothyronine (T3)-treated chickens exhibited increases in the expression of avian uncoupling protein (avUCP, an ortholog of mammalian UCP2 and UCP3) gene and some known mammalian beige adipocyte-specific markers. Morphological characteristics of white adipose tissues of treated chickens showed increased numbers of both small and larger clusters of multilocular fat cells within the tissues. Increases in protein levels of avUCP and mitochondrial marker protein, voltage-dependent anion channel, and immunohistochemical analysis for subcutaneous neck fat revealed the presence of potentially thermogenic mitochondria-rich cells. This is the first evidence that the capacity for thermogenesis may be acquired by differentiating adipose tissue into beige-like fat for maintaining temperature homeostasis in the subcutaneous fat 'neck warmer' in chickens exposed to a cold environment.


Subject(s)
Acclimatization/physiology , Chickens/physiology , Subcutaneous Fat/metabolism , Abdominal Fat/cytology , Abdominal Fat/metabolism , Adipocytes, Beige/metabolism , Adipose Tissue/metabolism , Animals , Body Weight , Cold Temperature , Eating , Mitochondria/metabolism , Neck/physiology , Subcutaneous Fat/cytology , Subcutaneous Fat/drug effects , Thermogenesis/drug effects , Triiodothyronine/pharmacology , Uncoupling Protein 1/genetics , Uncoupling Protein 1/metabolism , Voltage-Dependent Anion Channels/metabolism
3.
Vet Radiol Ultrasound ; 62(4): 437-444, 2021 Jul.
Article in English | MEDLINE | ID: mdl-33567137

ABSTRACT

Contrast-enhanced computed tomography (CT) is increasingly being used as a standard diagnostic test for dogs with suspected abdominal pathology. The iodinated contrast dose is commonly calculated based on linear increases in total body weight. However, body fat is not metabolically active and contributes little to dispersing or diluting the contrast medium in the blood. The aim of this retrospective single-center analytic study was to investigate the possible correlation between abdominal organ and vessel enhancement, and abdominal fat percentage in dogs. We hypothesized that, when dosing intravenous iodinated contrast according to total body weight, there would be a positive association between the degree of contrast enhancement of selected organs and vessels with increasing abdominal fat percentage. Vascular and parenchymal attenuation data were collected from 62 multiphasic abdominal CECT scans performed on dogs over a 5-year period at U-Vet Werribee Animal Hospital between February 2014 and February 2019. Findings based on a linear regression model showed a positive association of aorta (P = .005), liver (P = .045), and portal vein (P = .001) enhancement to abdominal fat percentage during the portal venous phase. Authors recommend that other body size parameters, such as lean body weight, should be considered when calculating iodine dose for abdominal contrast-enhanced CT in dogs.


Subject(s)
Abdominal Fat/diagnostic imaging , Aorta/diagnostic imaging , Contrast Media , Dogs , Liver/diagnostic imaging , Portal Vein/diagnostic imaging , Tomography, X-Ray Computed/veterinary , Abdominal Fat/cytology , Animals , Contrast Media/administration & dosage , Humans , Linear Models , Male , Retrospective Studies
4.
Horm Res Paediatr ; 93(5): 287-296, 2020.
Article in English | MEDLINE | ID: mdl-33075770

ABSTRACT

INTRODUCTION: Abnormalities in adipose tissue AdipoR1; adaptor protein, phosphotyrosine interaction, PH domain, and leucine zipper containing 1 (APPL1); GTPase Rab5; adiponectin; leptin; and visfatin in adults with obesity have been associated with metabolic disturbances. OBJECTIVE: The objective of this study was to examine whether pediatric obesity disrupts elements of the adiponectin signaling pathway and GTPase Rab5 in adipose tissue. METHODS: Primary adipocyte cultures of subcutaneous abdominal tissue were obtained from 96 lean and 66 children and adolescents with obesity (AO). AdipoR1, APPL1, and GTPase Rab5 mRNA levels were measured by RT-PCR and their protein content by Western immunoblotting. Serum total and high-molecular-weight adiponectin, leptin, leptin soluble receptor (sOB-R), and visfatin were measured by ELISA. RESULTS: The mRNA expression and protein content of AdipoR1 and APPL1 did not differ significantly with obesity, age, or puberty. However, GTPase Rab5 protein was increased in the adipocytes of younger prepubertal children with obesity but decreased in AO. Leptin was increased in AO compared to lean adolescents (AL) and in older prepubertal lean (OPL) children and AL compared to younger prepubertal lean and obese children. sOB-R was higher in OPL children and in the AL and AO. Serum visfatin was increased in the younger prepubertal children and AO. CONCLUSIONS: In contrast to adults, obesity did not change the expression of AdipoR1 and APPL1 in cultured adipocytes from biopsies of subcutaneous abdominal adipose tissue of children and adolescents. Similar to adipose tissue studies in adults with obesity and metabolic dysfunction, the AO in our study showed reduced adipocyte GTPase Rab5 expression.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Adipocytes/metabolism , Pediatric Obesity/metabolism , Receptors, Adiponectin/metabolism , rab5 GTP-Binding Proteins/metabolism , Abdominal Fat/cytology , Adiponectin/metabolism , Adolescent , Case-Control Studies , Child , Child, Preschool , Female , Humans , Infant , Male , Primary Cell Culture , Signal Transduction
5.
Int J Immunopathol Pharmacol ; 34: 2058738420966092, 2020.
Article in English | MEDLINE | ID: mdl-33121303

ABSTRACT

MicroRNAs (miRNAs) are a class of short non-coding RNAs that play a significant role in biological processes in various cell types, including mesenchymal stem cells (MSCs). However, how miRNAs regulate the immunomodulatory functions of adipose-derived MSCs (AD-MSCs) remains unknown. Here, we showed that modulation of miR-301a in AD-MSCs altered macrophage polarization. Bone marrow (BM)-derived macrophages were stimulated with LPS (1 µg/ml) and co-cultured with miRNA transfected AD-MSCs for 24 h. The expression of M1 and M2 markers in macrophages was analyzed. Inhibition of miR-301a induced M2 macrophage with arginase-1, CD163, CD206, and IL-10 upregulation. Additionally, toll-like receptor (TLR)-4 mRNA expression in macrophages was downregulated in co-cultures with AD-MSCs transfected with a miR-301a inhibitor. Nitric oxide (NO) in the supernatant of AD-MSC/macrophage co-culture was also suppressed by inhibition of miR-301a in AD-MSCs. We further found that suppression of miR-301a in AD-MSCs increased prostaglandin E2 (PGE2) concentration in the conditioned medium of the co-culture. Taken together, the results of our study indicate that miR-301a can modulate the immunoregulatory functions of AD-MSCs that favor the applicability as a potential immunotherapeutic agent.


Subject(s)
Abdominal Fat/cytology , Cell Communication , Cell Plasticity , Macrophages/metabolism , Mesenchymal Stem Cells/metabolism , MicroRNAs/metabolism , Animals , Biomarkers/metabolism , Cells, Cultured , Coculture Techniques , Gene Expression Regulation , Macrophages/immunology , Mesenchymal Stem Cells/immunology , MicroRNAs/genetics , Phenotype , Rats, Inbred Lew , Signal Transduction
6.
J Vis Exp ; (162)2020 08 16.
Article in English | MEDLINE | ID: mdl-32865529

ABSTRACT

The stromal-vascular fraction (SVF) of white adipose tissue (WAT) is remarkably heterogeneous and consists of numerous cell types that contribute functionally to the expansion and remodeling of WAT in adulthood. A tremendous barrier to studying the implications of this cellular heterogeneity is the inability to readily isolate functionally distinct cell subpopulations from WAT SVF for in vitro and in vivo analyses. Single-cell sequencing technology has recently identified functionally distinct fibro-inflammatory and adipogenic PDGFRß+ perivascular cell subpopulations in intra-abdominal WAT depots of adult mice. Fibro-inflammatory progenitors (termed, "FIPs") are non-adipogenic collagen producing cells that can exert a pro-inflammatory phenotype. PDGFRß+ adipocyte precursor cells (APCs) are highly adipogenic both in vitro and in vivo upon cell transplantation. Here, we describe multiple methods for the isolation of these stromal cell subpopulations from murine intra-abdominal WAT depots. FIPs and APCs can be isolated by fluorescence-activated cell sorting (FACS) or by taking advantage of biotinylated antibody-based immunomagnetic bead technology. Isolated cells can be used for molecular and functional analysis. Studying the functional properties of stromal cell subpopulation in isolation will expand our current knowledge of adipose tissue remodeling under physiological or pathological conditions on the cellular level.


Subject(s)
Abdominal Fat/cytology , Adipogenesis , Cell Separation/methods , Stromal Cells/cytology , Adipose Tissue, White/cytology , Animals , Flow Cytometry , Inflammation/pathology , Mice , Stromal Cells/pathology
7.
PLoS One ; 15(4): e0231796, 2020.
Article in English | MEDLINE | ID: mdl-32287314

ABSTRACT

BACKGROUND: Antimicrobial peptide expression is associated with disease activity in inflammatory bowel disease (IBD) patients. IBD patients have abnormal expression of elafin, a human elastase-specific protease inhibitor and antimicrobial peptide. We determined elafin expression in blood, intestine, and mesenteric fat of IBD and non-IBD patients. METHODS: Serum samples from normal and IBD patients were collected from two UCLA cohorts. Surgical resection samples of human colonic and mesenteric fat tissues from IBD and non-IBD (colon cancer) patients were collected from Cedars-Sinai Medical Center. RESULTS: High serum elafin levels were associated with a significantly elevated risk of intestinal stricture in Crohn's disease (CD) patients. Microsoft Azure Machine learning algorithm using serum elafin levels and clinical data identified stricturing CD patients with high accuracy. Serum elafin levels had weak positive correlations with clinical disease activity (Partial Mayo Score and Harvey Bradshaw Index), but not endoscopic disease activity (Mayo Endoscopic Subscore and Simple Endoscopic Index for CD) in IBD patients. Ulcerative colitis (UC) patients had high serum elafin levels. Colonic elafin mRNA and protein expression were not associated with clinical disease activity and histological injury in IBD patients, but stricturing CD patients had lower colonic elafin expression than non-stricturing CD patients. Mesenteric fat in stricturing CD patients had significantly increased elafin mRNA and protein expression, which may contribute to high circulating elafin levels. Human mesenteric fat adipocytes secrete elafin protein. CONCLUSIONS: High circulating elafin levels are associated with the presence of stricture in CD patients. Serum elafin levels may help identify intestinal strictures in CD patients.


Subject(s)
Colitis, Ulcerative/blood , Crohn Disease/complications , Elafin/blood , Intestinal Obstruction/diagnosis , Abdominal Fat/cytology , Abdominal Fat/metabolism , Adipocytes/metabolism , Adult , Case-Control Studies , Cell Line , Colitis, Ulcerative/pathology , Colon/diagnostic imaging , Colon/pathology , Colonoscopy , Constriction, Pathologic/blood , Constriction, Pathologic/diagnosis , Constriction, Pathologic/etiology , Crohn Disease/blood , Crohn Disease/diagnosis , Crohn Disease/pathology , Elafin/metabolism , Female , Fibroblasts , Healthy Volunteers , Humans , Intestinal Mucosa/diagnostic imaging , Intestinal Mucosa/pathology , Intestinal Obstruction/blood , Intestinal Obstruction/etiology , Intestinal Obstruction/pathology , Male , Primary Cell Culture , Prospective Studies , Severity of Illness Index
8.
Sci Rep ; 10(1): 5231, 2020 03 23.
Article in English | MEDLINE | ID: mdl-32251339

ABSTRACT

Platelet-rich plasma (PRP) has been widely used to improve the fat retention rate in autologous fat transplantation since it possesses a good angiogenesis capability in vivo. However, due to the short half-life of growth factors released from PRP and its uneven distribution in injected fat tissue, the strategy of PRP in fat transplantation needs further improvement. Since the capillaries started to grow into fat grafts in 1 week and vascular growth peaks in the second week after transplantation, we hypothesized that delayed two-steps PRP injection into the interior of grafts, accompanied with the extent of neovascularization might theoretically promote microvessel growth inside transplanted adipose tissue. 24 nude mice were divided into three groups: Blank group (0.35 mL fat mixed with 0.15 mL saline, N = 8), Single step group (0.35 mL fat mixed with 0.15 mLPRP, N = 8), and Two steps group (0.35 mL fat (day 0) + 0.075 mL PRP (day 7) + 0.075 mL PRP (day 14), N = 8). At 6 and 14 weeks post-transplantation, grafts were dissected, weighted, and assessed for histology, angiogenesis, fat regeneration and inflammation level. The weight and volume of the fat samples revealed no statistical difference among the three groups at 6 weeks after fat transplantation. The weight and volume of the Two steps group fat samples showed significantly higher compared to that in Blank and Single step groups at 14 weeks after fat transplantation (weight: 137.25 ± 5.60 mg versus 87.5 ± 3.90 mg,106.75 ± 2.94 mg, respectively; volume: 0.13 ± 0.01 mL versus 0.08 ± 0.01 mL, 0.09 ± 0.01 mL, respectively). Histological assessments indicated that delayed two-steps PRP injection strategy helps to improve adipose tissue content and reduce the composition of fibrous connective tissue at 14 weeks after fat transplantation. At 6 weeks and 14 weeks after transplantation, CD31 immunofluorescence indicated that delayed two-steps PRP injection strategy helps to improve angiogenesis and significantly higher compared to that in Blank and Single step groups (6 weeks: 28.75 ± 4.54 versus 10.50 ± 2.06, 21.75 ± 1.85; 14 weeks: 21.75 ± 2.86 versus 9.87 ± 2.08, 11.75 ± 1.47, respectively). Preadipocyte count indicated delayed two-steps PRP injection strategy might promote fat regeneration and significantly higher compared to that in Blank and Single step groups at 14 weeks (129.75 ± 6.57 versus 13.50 ± 3.50, 17.12 ± 6.23, respectively). In this study, we demonstrated that the novel delayed two-steps PRP injection strategy remarkably enhanced the long-term fat retention rate and improved the neovascularization extent in the interior of the fat graft. Platelet-rich plasma, Delayed two-steps injection, Angiogenesis, Fat transplantation.


Subject(s)
Abdominal Fat/blood supply , Abdominal Fat/transplantation , Neovascularization, Physiologic/physiology , Platelet-Rich Plasma , Abdominal Fat/cytology , Adipocytes/cytology , Adult , Animals , Capillaries , Cell Survival , Female , Half-Life , Humans , Injections , Macrophages , Male , Mice, Inbred BALB C , Platelet-Rich Plasma/metabolism
9.
Mol Nutr Food Res ; 64(1): e1900925, 2020 01.
Article in English | MEDLINE | ID: mdl-31785208

ABSTRACT

SCOPE: Inflammatory responses to obesity, including interleukin-1 beta (IL-1ß) activation, downregulate mitochondrial function and interfere with adipocyte browning, an important component of energy expenditure. This study investigates the impact of apigenin (Apg), a natural flavonoid with anti-inflammatory properties, on adipocyte browning in the presence of IL-1ß. METHODS AND RESULTS: Apg protects dibutyryl-cAMP-induced browning from IL-1ß in primary human adipocytes, as evidenced by increased brown-specific markers, mitochondrial content, and oxygen consumption. Apg significantly represses inflammatory markers and NF-κB activation induced by IL-1ß in these adipocytes. Intriguingly, Apg profoundly induces cyclooxygenase 2 (COX2) and prostaglandin E2 (PGE2) expression in response to IL-1ß treatment. Conversely, COX2 pharmacological inhibition or RNA silencing attenuates the positive effect of Apg on adipocyte browning in IL-1ß-treated cells. Additionally, blockage of PGE2 receptor 4 (EP4) attenuates Apg-mediated adipocyte browning. The effect of Apg on adipocyte browning in IL-1ß-treated adipocytes is accompanied by an elevation in intracellular Ca2+ , partly due to TRPV1/4 receptor activation. CONCLUSION: Apg plays a protective role against inflammation-induced suppression of adipocyte browning by dampening inflammation and activating the COX2/PGE2 axis for uncoupling protein 1 induction via EP4 activation. These data unravel the novel therapeutic values of Apg for treating obesity via adipocyte browning stimulation.


Subject(s)
Adipocytes/drug effects , Apigenin/pharmacology , Cyclooxygenase 2/metabolism , Dinoprostone/metabolism , Interleukin-1beta/pharmacology , Abdominal Fat/cytology , Adipocytes/metabolism , Adipocytes, Brown/drug effects , Adipocytes, Brown/metabolism , Calcium/metabolism , Cells, Cultured , Cyclooxygenase 2/genetics , Female , Humans , Signal Transduction/drug effects
10.
BMC Genomics ; 20(1): 743, 2019 Oct 15.
Article in English | MEDLINE | ID: mdl-31615399

ABSTRACT

BACKGROUND: The distribution and deposition of fat tissue in different parts of the body are the key factors affecting the carcass quality and meat flavour of chickens. Intramuscular fat (IMF) content is an important factor associated with meat quality, while abdominal fat (AbF) is regarded as one of the main factors affecting poultry slaughter efficiency. To investigate the differentially expressed genes (DEGs) and molecular regulatory mechanisms related to adipogenic differentiation between IMF- and AbF-derived preadipocytes, we analysed the mRNA expression profiles in preadipocytes (0d, Pre-) and adipocytes (10d, Ad-) from IMF and AbF of Gushi chickens. RESULTS: AbF-derived preadipocytes exhibited a higher adipogenic differentiation ability (96.4% + 0.6) than IMF-derived preadipocytes (86.0% + 0.4) (p < 0.01). By Ribo-Zero RNA sequencing, we obtained 4403 (2055 upregulated and 2348 downregulated) and 4693 (2797 upregulated and 1896 downregulated) DEGs between preadipocytes and adipocytes in the IMF and Ad groups, respectively. For IMF-derived preadipocyte differentiation, pathways related to the PPAR signalling pathway, ECM-receptor interaction and focal adhesion pathway were significantly enriched. For AbF-derived preadipocyte differentiation, the steroid biosynthesis pathways, calcium signaling pathway and ECM-receptor interaction pathway were significantly enriched. A large number of DEGs related to lipid metabolism, fatty acid metabolism and preadipocyte differentiation, such as PPARG, ACSBG2, FABP4, FASN, APOA1 and INSIG1, were identified in our study. CONCLUSION: This study revealed large transcriptomic differences between IMF- and AbF-derived preadipocyte differentiation. A large number of DEGs and transcription factors that were closely related to fatty acid metabolism, lipid metabolism and preadipocyte differentiation were identified in the present study. Additionally, the microenvironment of IMF- and AbF-derived preadipocyte may play a significant role in adipogenic differentiation. This study provides valuable evidence to understand the molecular mechanisms underlying adipogenesis and fat deposition in chickens.


Subject(s)
Abdominal Fat/cytology , Adipogenesis , Gene Expression Profiling/methods , Gene Regulatory Networks , Muscles/cytology , Abdominal Fat/metabolism , Adipocytes/cytology , Adipocytes/metabolism , Animals , Cell Differentiation , Chickens , Lipid Metabolism , Muscles/metabolism , Sequence Analysis, RNA
11.
Immunol Lett ; 216: 97-105, 2019 12.
Article in English | MEDLINE | ID: mdl-31622634

ABSTRACT

Mesenchymal stem cell (MSCs) therapy are among new strategies that are used to combat infections through immunomodulation. Cell number, route and frequency of injection and the duration of exposure to the infectious agent are of the main factors to determine the effectiveness of cell therapy. The current study was aimed to assess the effect of multiple intravenous (i.v.) injection of adipose tissue derived (AD)-MSCs on immune response of Leishmania (L.) major-infected BALB/c mice. Therefore, infected mice received AD-MSCs four times during the early phase of infection through i.v. route. They were then monitored weekly for footpad swelling and lesion development. Parasite burden, nitric oxide (NO) and cytokine production were measured in the spleen and lymph node 90 days post-infection. Delayed lesion development, significant reduction in footpad swelling and lower parasite burden in the spleen of AD-MSCs-treated mice showed the relative effect of AD-MSCs therapy in the control of L. major dissemination. In addition, MSCs were able to manage direct cytokine responses toward T-helper 1 (Th1). Although the level of interleukin (IL)-10 was still higher than the associated level of tumor necrosis factor (TNF)-α, a shift towards higher level of TNF-α was also observed.


Subject(s)
Abdominal Fat/cytology , Leishmania major/immunology , Leishmaniasis, Cutaneous/therapy , Mesenchymal Stem Cell Transplantation/methods , Th1 Cells/immunology , Animals , Cells, Cultured/transplantation , Disease Models, Animal , Female , Humans , Injections, Intravenous , Leishmania major/isolation & purification , Leishmaniasis, Cutaneous/blood , Leishmaniasis, Cutaneous/immunology , Lymph Nodes/immunology , Lymph Nodes/parasitology , Mice , Mice, Inbred BALB C , Parasite Load , Primary Cell Culture , Skin/immunology , Skin/parasitology , Spleen/immunology , Spleen/parasitology
12.
JAMA Facial Plast Surg ; 21(6): 526-534, 2019 Dec 01.
Article in English | MEDLINE | ID: mdl-31556908

ABSTRACT

IMPORTANCE: Adipose-derived mesenchymal stem cells (ASCs) have been used commonly in regenerative medicine and increasingly for head and neck surgical procedures. Lipoaspiration with centrifugation is purported to be a mild method for the extraction of ASCs used for autologous transplants to restore tissue defects or induce wound healing. The content of ASCs, their paracrine potential, and cellular potential in wound healing have not been explored for this method to our knowledge. OBJECTIVE: To evaluate the characteristics of lipoaspirates used in reconstructive head and neck surgical procedures with respect to wound healing. DESIGN, SETTING, AND PARTICIPANTS: This case series study included 15 patients who received autologous fat injections in the head and neck during surgical procedures at a tertiary referral center. The study was performed from October 2017 to November 2018, and data were analyzed from October 2017 to February 2019. MAIN OUTCOMES AND MEASURES: Excessive material of lipoaspirates from subcutaneous abdominal fatty tissue was examined. Cellular composition was analyzed using immunohistochemistry (IHC) and flow cytometry, and functionality was assessed through adipose, osteous, and chondral differentiation in vitro. Supernatants were tested for paracrine ASC functions in fibroblast wound-healing assays. Enzyme-linked immunosorbent assay measurement of tumor necrosis factor (TNF), vascular endothelial growth factor (VEGF), stromal-derived factor 1α (SDF-1α), and transforming growth factor ß3 (TGF-ß3) was performed. RESULTS: Among the 15 study patients (8 [53.3%] male; mean [SD] age at the time of surgery, 63.0 [2.8] years), the stromal vascular fraction (mean [SE], 53.3% [4.2%]) represented the largest fraction within the native lipoaspirates. The cultivated cells were positive for CD73 (mean [SE], 99.90% [0.07%]), CD90 (99.40% [0.32%]), and CD105 (88.54% [2.74%]); negative for CD34 (2.70% [0.45%]) and CD45 (1.74% [0.28%]) in flow cytometry; and negative for CD14 (10.56 [2.81] per 300 IHC score) and HLA-DR (6.89 [2.97] per 300 IHC score) in IHC staining; they differentiated into osteoblasts, adipocytes, and chondrocytes. The cultivated cells showed high expression of CD44 (mean [SE], 99.78% [0.08%]) and CD273 (82.56% [5.83%]). The supernatants were negative for TNF (not detectable) and SDF-1α (not detectable) and were positive for VEGF (mean [SE], 526.74 [149.84] pg/mL for explant supernatants; 528.26 [131.79] pg/106 per day for cell culture supernatants) and TGF-ß3 (mean [SE], 22.79 [3.49] pg/mL for explant supernatants; 7.97 [3.15] pg/106 per day for cell culture supernatants). Compared with control (25% or 50% mesenchymal stem cell medium), fibroblasts treated with ASC supernatant healed the scratch-induced wound faster (mean [SE]: control, 1.000 [0.160]; explant supernatant, 1.369 [0.070]; and passage 6 supernatant, 1.492 [0.094]). CONCLUSIONS AND RELEVANCE: The cells fulfilled the international accepted criteria for mesenchymal stem cells. The lipoaspirates contained ASCs that had the potential to multidifferentiate with proliferative and immune-modulating properties. The cytokine profile of the isolated ASCs had wound healing-promoting features. Lipoaspirates may have a regenerative potential and an application in head and neck surgery. LEVEL OF EVIDENCE: NA.


Subject(s)
Abdominal Fat/cytology , Abdominal Fat/transplantation , Dysphonia/surgery , Head and Neck Neoplasms/surgery , Mesenchymal Stem Cells/physiology , Cell Differentiation/physiology , Cells, Cultured , Enzyme-Linked Immunosorbent Assay , Female , Humans , Immunohistochemistry , Immunophenotyping , Male , Middle Aged , Regeneration
13.
Cell Rep ; 27(5): 1528-1540.e7, 2019 04 30.
Article in English | MEDLINE | ID: mdl-31042478

ABSTRACT

Adipocyte progenitor cells (APCs) provide the reservoir of regenerative cells to produce new adipocytes, although their identity in humans remains elusive. Using FACS analysis, gene expression profiling, and metabolic and proteomic analyses, we identified three APC subtypes in human white adipose tissues. The APC subtypes are molecularly distinct but possess similar proliferative and adipogenic capacities. Adipocytes derived from APCs with high CD34 expression exhibit exceedingly high rates of lipid flux compared with APCs with low or no CD34 expression, while adipocytes produced from CD34- APCs display beige-like adipocyte properties and a unique endocrine profile. APCs were more abundant in gluteofemoral compared with abdominal subcutaneous and omental adipose tissues, and the distribution of APC subtypes varies between depots and in patients with type 2 diabetes. These findings provide a mechanistic explanation for the heterogeneity of human white adipose tissue and a potential basis for dysregulated adipocyte function in type 2 diabetes.


Subject(s)
Abdominal Fat/cytology , Adipocytes/metabolism , Diabetes Mellitus, Type 2/pathology , Mesenchymal Stem Cells/metabolism , Subcutaneous Fat/cytology , Abdominal Fat/pathology , Adipocytes/classification , Adipocytes/physiology , Adiposity , Adult , Animals , Antigens, CD34/genetics , Antigens, CD34/metabolism , Cell Proliferation , Cells, Cultured , Female , Humans , Male , Mesenchymal Stem Cells/classification , Mesenchymal Stem Cells/physiology , Mice , Mice, SCID , Middle Aged , Proteome , Subcutaneous Fat/pathology , Transcriptome
14.
Cell Mol Biol Lett ; 24: 14, 2019.
Article in English | MEDLINE | ID: mdl-30815013

ABSTRACT

BACKGROUND: The proliferation and adipogenic differentiation of adipose stromal cells (ASCs) are complex processes comprising major phenotypical alterations driven by up- and downregulation of hundreds of genes. Quantitative RT-PCR can be employed to measure relative changes in the expression of a gene of interest. This approach requires constitutively expressed reference genes for normalization to counteract inter-sample variations due to differences in RNA quality and quantity. Thus, a careful validation of quantitative RT-PCR reference genes is needed to accurately measure fluctuations in the expression of genes. Here, we evaluated candidate reference genes applicable for quantitative RT-PCR analysis of gene expression during proliferation and adipogenesis of human ASCs with the immunophenotype DLK1+/CD34+/CD90+/CD105+/CD45-/CD31-. METHODS: We evaluated the applicability of 10 candidate reference genes (GAPDH, TBP, RPS18, EF1A, TFRC, GUSB, PSMD5, CCNA2, LMNA and MRPL19) using NormFinder, geNorm and BestKeeper software. RESULTS: The results indicate that EF1A and MRPL19 are the most reliable reference genes for quantitative RT-PCR analysis of proliferating ASCs. PSMD5 serves as the most reliable endogenous control in adipogenesis. CCNA2 and LMNA were among the least consistent genes. CONCLUSIONS: Applying these findings for future gene expression analyses will help elucidate ASC biology.


Subject(s)
Abdominal Fat/cytology , Gene Expression Profiling/methods , Real-Time Polymerase Chain Reaction/methods , Abdominal Fat/physiology , Adipogenesis , Cell Proliferation , Gene Expression Profiling/standards , Humans , Real-Time Polymerase Chain Reaction/standards , Reference Standards , Stromal Cells/physiology
15.
Glycoconj J ; 36(2): 155-163, 2019 04.
Article in English | MEDLINE | ID: mdl-30820783

ABSTRACT

Epithelial-mesenchymal transition (EMT) in peritoneum was induced during peritoneal dialysis (PD), which finally caused progressive fibrosis. However, the underlying mechanisms were not well elucidated. We established advanced glycation end-products (AGEs)-induced EMT model using primary human peritoneal mesothelial cells (HPMCs). The working concentration and time of AGEs were optimized. Then the expression and activation signal transducer and activator of transcription 3 (STAT3), a key factor of EMT in cancer, were detected. The regulation of STAT3 by miRNA was also explored. 50 µg/ml AGEs treatment for 24 h can successfully induce EMT in HPMCs. AGEs treatment upregulated and activated STAT3. miRNA-454, potentially targeting STAT3, was down-regulated in AGEs-treated HPMCs. Overexpression of miRNA-454 prevented AGEs- induced EMT in HPMCs. AGEs induce epithelial to mesenchymal transformation of Human peritoneal mesothelial cells via upregulation of STAT3.


Subject(s)
Epithelial Cells/drug effects , Epithelial-Mesenchymal Transition , Glycation End Products, Advanced/pharmacology , STAT3 Transcription Factor/metabolism , Abdominal Fat/cytology , Cells, Cultured , Epithelial Cells/metabolism , Epithelial Cells/pathology , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , Up-Regulation
16.
Sci Rep ; 9(1): 3087, 2019 02 28.
Article in English | MEDLINE | ID: mdl-30816281

ABSTRACT

Identification of adipose-specific genes has contributed to an understanding of mechanisms underlying adipocyte development and obesity. Herein, our analyses of the recent Genotype-Tissue Expression (GTEx) database revealed 38 adipose-specific/enhanced protein coding genes, among which 3 genes were novel adipose-specific, and 414 highly differentially expressed genes (DEGs) between subcutaneous and omental adipose depots. By integrative analyses of genome-wide association studies (GWASs), 14 adipose-specific/enhanced genes and 60 DEGs were found to be associated with obesity-related traits and diseases, consolidating evidence for contribution of these genes to the regional fat distribution and obesity phenotypes. In addition, expression of HOXC cluster was up-regulated in subcutaneous adipose tissue, and the majority of the HOXB cluster was expressed highly in omental adipose tissue, indicating differential expression patterns of HOX clusters in adipose depots. Our findings on the distinct gene expression profiles in adipose tissue and their relation to obesity provide an important foundation for future functional biological studies and therapeutic targets in obesity and associated diseases.


Subject(s)
Abdominal Fat/metabolism , Adipocytes/metabolism , Genome-Wide Association Study , Obesity/genetics , Omentum/metabolism , Subcutaneous Fat/metabolism , Abdominal Fat/cytology , Adipocytes/cytology , Datasets as Topic , Genes, Homeobox , Genetic Loci , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Omentum/cytology , Subcutaneous Fat/cytology , Transcriptome
17.
Stem Cells Transl Med ; 7(12): 876-886, 2018 12.
Article in English | MEDLINE | ID: mdl-30255987

ABSTRACT

Autologous adipose tissue is used for tissue repletion and/or regeneration as an intact lipoaspirate or as enzymatically derived stromal vascular fraction (SVF), which may be first cultured into mesenchymal stem cells (MSCs). Alternatively, transplant of autologous adipose tissue mechanically fragmented into submillimeter clusters has recently showed remarkable efficacy in diverse therapeutic indications. To document the biologic basis of the regenerative potential of microfragmented adipose tissue, we first analyzed the distribution of perivascular presumptive MSCs in adipose tissue processed with the Lipogems technology, observing a significant enrichment in pericytes, at the expense of adventitial cells, as compared to isogenic enzymatically processed lipoaspirates. The importance of MSCs as trophic and immunomodulatory cells, due to the secretion of specific factors, has been described. Therefore, we investigated protein secretion by cultured adipose tissue clusters or enzymatically derived SVF using secretome arrays. In culture, microfragmented adipose tissue releases many more growth factors and cytokines involved in tissue repair and regeneration, noticeably via angiogenesis, compared to isogenic SVF. Therefore, we suggest that the efficient tissue repair/regeneration observed after transplantation of microfragmented adipose tissue is due to the secretory ability of the intact perivascular niche. Stem Cells Translational Medicine 2018;7:876-886.


Subject(s)
Adipose Tissue/metabolism , Pericytes/metabolism , Abdominal Fat/cytology , Abdominal Fat/metabolism , Adipose Tissue/cytology , Adult , Aged , Agglutinins/metabolism , Angiogenic Proteins/metabolism , Collagenases/metabolism , Cytokines/metabolism , Female , Flow Cytometry , Humans , Intercellular Signaling Peptides and Proteins/metabolism , Middle Aged , Pericytes/cytology , Proteome/analysis
18.
Int J Obes (Lond) ; 42(4): 721-727, 2018 04.
Article in English | MEDLINE | ID: mdl-29188818

ABSTRACT

BACKGROUND/OBJECTIVES: The phenomenon of adipocyte 'beiging' involves the conversion of non-classic brown adipocytes to brown-like adipose tissue with thermogenic, fat-burning properties, and this phenomenon has been shown in rodents to slow the progression of obesity-associated metabolic diseases. Rodent studies consistently report adipocyte beiging after endurance exercise training, indicating that increased thermogenic capacity in these adipocytes may underpin the improved health benefits of exercise training. The aim of this study was to determine whether prolonged endurance exercise training induces beige adipogenesis in subcutaneous adipose tissues of obese men. SUBJECTS/METHODS: Molecular markers of beiging were examined in adipocytes obtained from abdominal subcutaneous (AbSC) and gluteofemoral (GF) subcutaneous adipose tissues before and after 6 weeks of endurance exercise training in obese men (n=6, 37.3±2.3 years, 30.1±2.3 kg m-2). RESULTS: The mRNAs encoding the brown or beige adipocyte-selective proteins were very lowly expressed in AbSC and GF adipose tissues and exercise training did not alter the mRNA expression of UCP1, CD137, CITED, TBX1, LHX8 and TCF21. Using immunohistochemistry, neither multilocular adipocytes, nor UCP1 or CD137-positive adipocytes were detected in any sample. MicroRNAs known to regulate brown and/or beige adipose development were highly expressed in white adipocytes but endurance exercise training did not impact their expression. CONCLUSIONS: The present study reaffirms emerging data in humans demonstrating no evidence of white adipose tissue beiging in response to exercise training, and supports a growing body of work demonstrating divergence of brown/beige adipose location, molecular characterization and physiological function between rodents and humans.


Subject(s)
Abdominal Fat/physiology , Adipocytes, Brown/metabolism , Adipocytes, White/metabolism , Endurance Training , Obesity/therapy , Subcutaneous Fat/physiology , Abdominal Fat/cytology , Cohort Studies , Humans , Male , MicroRNAs/analysis , MicroRNAs/genetics , MicroRNAs/metabolism , Subcutaneous Fat/cytology
19.
J Craniofac Surg ; 29(1): 243-247, 2018 Jan.
Article in English | MEDLINE | ID: mdl-29068969

ABSTRACT

Autologous fat grafting is commonly used for soft tissue augmentation, but its unpredictably high resorption rate remains a major limitation. Although adipose-derived mesenchymal stem cells (ASCs) are an attractive candidate for enhancing graft retention, their poor posttransplantation viability limits their application. The authors aimed to evaluate the effect of incubated ASCs on microfat graft survival in an immunocompromised mouse model. Lipoaspirates for microfat injection were collected from the wasted lower abdominal adipose tissues of 5 patients who had undergone breast reconstructive surgery with an abdominal flap. Adipose-derived mesenchymal stem cells were also isolated and proliferated from these fat tissues. Sixty athymic mice were randomly allocated to a control group (microfat grafting alone; n = 30) or ASCs group (microfat grafting plus simultaneous human ASCs injection; n = 30). The volume and weight of survived fat were measured at 8 and 16 weeks, and histopathological and immunologic staining was performed at 16 weeks. The survived fat volume of the ASCs group was significantly greater than that of the control group at 8 and 16 weeks, whereas the weight of survived fat tissues did not significantly differ. Histologic evaluation of the harvested fat indicated significantly higher levels of adipocytes, and fewer cysts and fibrosis in the tissues in the ASCs group than in the control group. The ASCs group also exhibited a significantly higher number of capillary vessels than the control group on CD31 and alpha-smooth muscle actin staining. In conclusion, transplanted fat survival is markedly higher when simultaneous microfat graft and ASCs injection were performed, as compared with that in the classical microfat graft alone method in mice; this improvement was primarily attributed to the increased ability to produce blood vessels.


Subject(s)
Abdominal Fat/transplantation , Graft Survival , Heterografts/pathology , Mesenchymal Stem Cell Transplantation , Abdominal Fat/cytology , Animals , Cells, Cultured , Heterografts/blood supply , Humans , Immunocompromised Host , Mesenchymal Stem Cells , Mice , Mice, Nude , Middle Aged , Models, Animal
20.
Diabetes Metab ; 44(2): 155-159, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29153750

ABSTRACT

AIMS: Type 2 diabetes is associated with insulin resistance, adipose hypertrophy and increased lipolysis. The heritability of these traits has been determined by associating them with a family history of diabetes. METHODS: Abdominal subcutaneous fat biopsies were obtained from 581 subjects in a cross-sectional study. Fat cells were isolated, and the difference between measured and expected fat-cell volume was used to determine adipose morphology (degree of hypertrophy or hyperplasia). Spontaneous lipolytic activity was determined in explants of adipose tissue by measuring glycerol release. Insulin-stimulated lipogenesis was assessed by measuring the incorporation of radiolabelled glucose into fat-cell lipids. Information on parental history of diabetes was gathered by a questionnaire. RESULTS: Adipose morphology correlated positively with lipolysis (P<0.0001) and inversely with insulin-stimulated lipogenesis (P<0.008). Also, 24% of probands had a family history of diabetes, which was associated with higher body mass index (BMI) scores, and more insulin resistance (HOMAIR) and adipose hypertrophy. Lipolytic activity was increased, and insulin-stimulated lipogenesis decreased, in probands with a parental history of diabetes. The results for HOMAIR, lipolysis and adipose morphology remained significant after adjusting for proband BMI. A maternal history of diabetes was associated with increased adipose lipolytic activity in probands. CONCLUSION: A family history of diabetes is independent of proband BMI, but associated with adipocyte hypertrophy and enhanced lipolysis, which suggests that these factors are genetically linked to diabetes. Moreover, the influence on lipolysis was only observed in probands with a maternal history of diabetes, thereby supporting an epigenetic impact.


Subject(s)
Diabetes Mellitus, Type 2/epidemiology , Diabetes Mellitus, Type 2/physiopathology , Lipogenesis/drug effects , Lipolysis/drug effects , Abdominal Fat/cytology , Abdominal Fat/drug effects , Adipocytes/cytology , Adipocytes/drug effects , Adolescent , Adult , Aged , Biopsy , Cells, Cultured , Epigenesis, Genetic , Female , Humans , Hypertrophy , Insulin/pharmacology , Male , Medical History Taking , Middle Aged , Subcutaneous Fat/cytology , Subcutaneous Fat/drug effects , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL