Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 514
Filter
1.
Anim Biotechnol ; 35(1): 2346223, 2024 Nov.
Article in English | MEDLINE | ID: mdl-38739480

ABSTRACT

Adipocyte play an important role in human health and meat quality by influencing the tenderness, flavor, and juiciness of mutton It has been shown that neuron-derived neurotrophic factor (NENF) is closely related to energy metabolism and adipocyte differentiation in bovine. However, the role of NENF in the goats remains unclear. The aim of this study was to detect the expression of NENF in goat subcutaneous and intramuscular adipocytes, temporal expression profiles of the NENF, and overexpressed NENF on the differentiation of different adipocytes. In this study, PCR amplification successfully cloned the goat NENF gene with a fragment length of 521 bp. In addition, the time point of highest expression of NENF differed between these two adipocytes differentiation processes. Overexpression of NENF in intramuscular and subcutaneous adipocytes promoted the expression levels of differentiation markers CEBPß and SREBP, which in turn promoted the differentiation of intramuscular and subcutaneous adipocytes. This study will provide basic data for further study of the role of goats in goat adipocyte differentiation and for the final elucidation of its molecular mechanisms in regulating goat adipocyte deposition.


Subject(s)
Adipocytes , Cell Differentiation , Goats , Animals , Goats/genetics , Adipocytes/cytology , Adipocytes/metabolism , Cell Differentiation/physiology , Subcutaneous Fat/cytology , Subcutaneous Fat/metabolism
2.
Aging (Albany NY) ; 16(9): 7535-7552, 2024 May 09.
Article in English | MEDLINE | ID: mdl-38728252

ABSTRACT

Adipose tissue regulates metabolic balance, but aging disrupts it, shifting fat from insulin-sensitive subcutaneous to insulin-resistant visceral depots, impacting overall metabolic health. Adipose-derived stem cells (ASCs) are crucial for tissue regeneration, but aging diminishes their stemness and regeneration potential. Our findings reveal that aging is associated with a decrease in subcutaneous adipose tissue mass and an increase in the visceral fat depots mass. Aging is associated with increase in adipose tissue fibrosis but no significant change in adipocyte size was observed with age. Long term caloric restriction failed to prevent fibrotic changes but resulted in significant decrease in adipocytes size. Aged subcutaneous ASCs displayed an increased production of ROS. Using mitochondrial membrane activity as an indicator of stem cell quiescence and senescence, we observed a significant decrease in quiescence ASCs with age exclusively in subcutaneous adipose depot. In addition, aged subcutaneous adipose tissue accumulated more senescent ASCs having defective autophagy activity. However, long-term caloric restriction leads to a reduction in mitochondrial activity in ASCs. Furthermore, caloric restriction prevents the accumulation of senescent cells and helps retain autophagy activity in aging ASCs. These results suggest that caloric restriction and caloric restriction mimetics hold promise as a potential strategy to rejuvenate the stemness of aged ASCs. Further investigations, including in vivo evaluations using controlled interventions in animals and human studies, will be necessary to validate these findings and establish the clinical potential of this well-established approach for enhancing the stemness of aged stem cells.


Subject(s)
Aging , Caloric Restriction , Cellular Senescence , Stem Cells , Subcutaneous Fat , Cellular Senescence/physiology , Animals , Subcutaneous Fat/cytology , Subcutaneous Fat/metabolism , Aging/physiology , Stem Cells/metabolism , Mice , Autophagy/physiology , Male , Mitochondria/metabolism , Reactive Oxygen Species/metabolism , Mice, Inbred C57BL , Adipocytes/metabolism
3.
J Vis Exp ; (206)2024 Apr 05.
Article in English | MEDLINE | ID: mdl-38647333

ABSTRACT

Microvascular endothelial cells (MVECs) have many critical roles, including control of vascular tone, regulation of thrombosis, and angiogenesis. Significant heterogeneity in endothelial cell (EC) genotype and phenotype depends on their vascular bed and host disease state. The ability to isolate MVECs from tissue-specific vascular beds and individual patient groups offers the opportunity to directly compare MVEC function in different disease states. Here, using subcutaneous adipose tissue (SAT) taken at the time of insertion of cardiac implantable electronic devices (CIED), we describe a method for the isolation of a pure population of functional human subcutaneous adipose tissue MVEC (hSATMVEC) and an experimental model of hSATMVEC-adipocyte cross-talk. hSATMVEC were isolated following enzymatic digestion of SAT by incubation with anti-CD31 antibody-coated magnetic beads and passage through magnetic columns. hSATMVEC were grown and passaged on gelatin-coated plates. Experiments used cells at passages 2-4. Cells maintained classic features of EC morphology until at least passage 5. Flow cytometric assessment showed 99.5% purity of isolated hSATMVEC, defined as CD31+/CD144+/CD45-. Isolated hSATMVEC from controls had a population doubling time of approximately 57 h, and active proliferation was confirmed using a cell proliferation imaging kit. Isolated hSATMVEC function was assessed using their response to insulin stimulation and angiogenic tube-forming potential. We then established an hSATMVEC-subcutaneous adipocyte co-culture model to study cellular cross-talk and demonstrated a downstream effect of hSATMVEC on adipocyte function. hSATMVEC can be isolated from SAT taken at the time of CIED insertion and are of sufficient purity to both experimentally phenotype and study hSATMVEC-adipocyte cross-talk.


Subject(s)
Adipocytes , Endothelial Cells , Subcutaneous Fat , Humans , Adipocytes/cytology , Endothelial Cells/cytology , Endothelial Cells/metabolism , Subcutaneous Fat/cytology , Cell Communication/physiology
4.
Int J Mol Sci ; 23(3)2022 Jan 28.
Article in English | MEDLINE | ID: mdl-35163472

ABSTRACT

Over the last decade, several compounds have been identified for the treatment of obesity. However, due to the complexity of the disease, many pharmacological interventions have raised concerns about their efficacy and safety. Therefore, it is important to discover new factors involved in the induction/progression of obesity. Adipose stromal/stem cells (ASCs), which are mostly isolated from subcutaneous adipose tissue, are the primary cells contributing to the expansion of fat mass. Like other cells, ASCs release nanoparticles known as extracellular vesicles (EVs), which are being actively studied for their potential applications in a variety of diseases. Here, we focused on the importance of the con-tribution of ASC-derived EVs in the regulation of metabolic processes. In addition, we outlined the advantages/disadvantages of the use of EVs as potential next-generation anti-obesity agents.


Subject(s)
Extracellular Vesicles/metabolism , Mesenchymal Stem Cells/cytology , Obesity/metabolism , Adipogenesis , Extracellular Vesicles/transplantation , Homeostasis , Humans , Obesity/therapy , Subcutaneous Fat/cytology , Subcutaneous Fat/metabolism
5.
Sci Rep ; 12(1): 1069, 2022 01 20.
Article in English | MEDLINE | ID: mdl-35058499

ABSTRACT

Current US Centers for Disease Control and Prevention intramuscular injection needle length guidelines for injection fo the deltoid muscle are based on weight and gender. The aims of this study are (1) to evaluate whether other biometric data (age, gender, height, weight and body mass index (BMI)) are better predictors of the thickness of the deltoid subcutaneous fat pad (DSFP) than weight and gender and (2) to evaluate the performance of the CDC weight-based needle length guidelines. This was a retrospective single center cohort study of 386 patients who underwent surveillance PET/CT between 01/01/2020 and 04/01/2021. Patient age, gender, height, weight, BMI and CT measurements of the DSFP were evaluated. DSFP was positively correlated with weight and BMI in men (r = 0.67, P < 0.001; r = 0.74, P < 0.001) and women (r = 0.69, P < 0.001; r = 0.75, P < 0.001) respectively. DSFP was negatively correlated with age in women (r = - 0.19, P = 0.013). Age and BMI were better predictors of DSFP than weight. The best model to predict the DSFP is: [Formula: see text] A 1-inch needle is expected to reach the deltoid in 85.3% of women less than 200 pounds, and 98.6% of men less than 260 pounds. This rate differed between genders (P < 0.001, odds ratio (OR) 0.08, 95% CI (0.02, 0.29)). A 1.5-inch needle is expected to reach the deltoid in 76.7% of women greater than 200 pounds, and 75.0% of men greater than 260 pounds. Current CDC deltoid intramuscular injection needle length guidelines result in women and obese individuals being more likely to receive subcutaneous injections. Age and BMI based guidelines for needle length selection are more accurate.


Subject(s)
Adipose Tissue/physiology , Deltoid Muscle/cytology , Injections, Intramuscular/methods , Adult , Aged , Aged, 80 and over , Biometry/methods , Body Height , Body Mass Index , Cohort Studies , Female , Humans , Injections, Intramuscular/standards , Injections, Subcutaneous/methods , Injections, Subcutaneous/standards , Male , Middle Aged , Models, Statistical , Needles/standards , Needles/trends , Obesity , Retrospective Studies , Skin , Subcutaneous Fat/cytology , Subcutaneous Tissue
6.
J Clin Endocrinol Metab ; 107(2): e836-e851, 2022 01 18.
Article in English | MEDLINE | ID: mdl-34448000

ABSTRACT

CONTEXT: MSCA1 (mesenchymal stem cell antigen 1) and CD36 (cluster of differentiation 36) have been described as novel adipocyte progenitor markers in adults with a potential relevance for obesity and adipocyte progenitor function. OBJECTIVE: With the early manifestation of obesity in children and formation of adipose tissue (AT) dysfunction, children provide the opportunity to characterize the function of MSCA1 and CD36 during physiological AT accumulation and with obesity and related disease. METHODS: We investigated MSCA1 and CD36 expression in adipocytes and stroma vascular fraction (SVF) cells from 133 children of the Leipzig AT Childhood cohort with regard to AT accumulation and biology. In a subsample we analyzed how MSCA1 and CD36 expression is related to adipose progenitor capacities in vitro (ie, proliferation, differentiation and mitochondrial function). RESULTS: Both MSCA1 and CD36 are differentially expressed in adipocytes and SVF cells of children. MSCA1 expression is positively correlated to obesity-associated AT dysfunction (ie, adipocyte hypertrophy and serum high-sensitivity C-reactive protein), and high SVF MSCA1 expression is associated with increased mitochondrial respiration in vitro. CD36 expression is not associated with AT dysfunction but SVF CD36 expression is downregulated in children with overweight and obesity and shows a positive association with the differentiation capacity of SVF cells ex vivo and in vitro. CONCLUSION: Both MSCA1 and CD36 are associated with obesity-related alterations in AT of children. In particular, CD36 expression predicts adipogenic potential of SVF cells, indicating a potential role in the regulation of adipocyte hyperplasia and hypertrophy with obesity development in children.


Subject(s)
Adipogenesis , Antigens, Surface/metabolism , Pediatric Obesity/physiopathology , Subcutaneous Fat/physiopathology , Adipocytes/metabolism , Adolescent , Antigens, Surface/analysis , Child , Child, Preschool , Cross-Sectional Studies , Female , Humans , Infant , Male , Stromal Vascular Fraction/metabolism , Subcutaneous Fat/cytology , Subcutaneous Fat/metabolism
7.
Diabetes ; 71(2): 321-328, 2022 02 01.
Article in English | MEDLINE | ID: mdl-34753798

ABSTRACT

Adipose-derived stem cells (ADSCs) can differentiate into vascular lineages and participate in vascular remodeling. Perivascular ADSCs (PV-ADSCs) draw attention because of their unique location. The heterogeneity of subcutaneous (SUB) and abdominal ADSCs were well addressed, but PV-ADSCs' heterogeneity has not been investigated. In this study, we applied single-cell analysis to compare SUB-ADSCs and PV-ADSCs regarding their subpopulations, functions, and cell fates. We uncovered four subpopulations of PV-ADSCs (Dpp4+, Col4a2+/Icam1+, Clec11a+/Cpe+, and Sult1e1+ cells), among which the Clec11a+ subpopulation potentially participated in and regulated PV-ADSC differentiation toward a smooth muscle cell (SMC) phenotype. Distinct characteristics between PV-ADSCs and SUB-ADSCs were revealed.


Subject(s)
Blood Vessels/cytology , Stem Cells/physiology , Subcutaneous Fat/cytology , Animals , Cell Differentiation , Cell Proliferation , Cells, Cultured , Mice , Mice, Inbred C57BL , Myocytes, Smooth Muscle/physiology , Single-Cell Analysis , Stem Cells/cytology
8.
Nat Commun ; 12(1): 6931, 2021 11 26.
Article in English | MEDLINE | ID: mdl-34836963

ABSTRACT

Obesity and type 2 diabetes are associated with disturbances in insulin-regulated glucose and lipid fluxes and severe comorbidities including cardiovascular disease and steatohepatitis. Whole body metabolism is regulated by lipid-storing white adipocytes as well as "brown" and "brite/beige" adipocytes that express thermogenic uncoupling protein 1 (UCP1) and secrete factors favorable to metabolic health. Implantation of brown fat into obese mice improves glucose tolerance, but translation to humans has been stymied by low abundance of primary human beige adipocytes. Here we apply methods to greatly expand human adipocyte progenitors from small samples of human subcutaneous adipose tissue and then disrupt the thermogenic suppressor gene NRIP1 by CRISPR. Ribonucleoprotein consisting of Cas9 and sgRNA delivered ex vivo are fully degraded by the human cells following high efficiency NRIP1 depletion without detectable off-target editing. Implantation of such CRISPR-enhanced human or mouse brown-like adipocytes into high fat diet fed mice decreases adiposity and liver triglycerides while enhancing glucose tolerance compared to implantation with unmodified adipocytes. These findings advance a therapeutic strategy to improve metabolic homeostasis through CRISPR-based genetic enhancement of human adipocytes without exposing the recipient to immunogenic Cas9 or delivery vectors.


Subject(s)
Adipocytes, Brown/transplantation , CRISPR-Cas Systems/genetics , Glucose Intolerance/therapy , Obesity/therapy , Thermogenesis/genetics , Adipocytes, Brown/metabolism , Adipocytes, White/metabolism , Adult Stem Cells/physiology , Animals , Cell Culture Techniques/methods , Cell Differentiation , Diet, High-Fat/adverse effects , Disease Models, Animal , Fatty Liver/etiology , Fatty Liver/metabolism , Fatty Liver/prevention & control , Gene Editing/methods , Glucose Intolerance/etiology , Glucose Intolerance/metabolism , Humans , Lipid Metabolism/genetics , Male , Mice , Nuclear Receptor Interacting Protein 1/genetics , Nuclear Receptor Interacting Protein 1/metabolism , Obesity/complications , Obesity/metabolism , RNA, Guide, Kinetoplastida/genetics , Subcutaneous Fat/cytology
9.
FASEB J ; 35(12): e22018, 2021 12.
Article in English | MEDLINE | ID: mdl-34731499

ABSTRACT

Adipose tissue is the primary site of energy storage, playing important roles in health. While adipose research largely focuses on obesity, fat also has other critical functions, producing adipocytokines and contributing to normal nutrient metabolism, which in turn play important roles in satiety and total energy homeostasis. SMAD2/3 proteins are downstream mediators of activin signaling, which regulate critical preadipocyte and mature adipocyte functions. Smad2 global knockout mice exhibit embryonic lethality, whereas global loss of Smad3 protects mice against diet-induced obesity. The direct contributions of Smad2 and Smad3 in adipose tissues, however, are unknown. Here, we sought to determine the primary effects of adipocyte-selective reduction of Smad2 or Smad3 on diet-induced adiposity using Smad2 or Smad3 "floxed" mice intercrossed with Adiponectin-Cre mice. Additionally, we examined visceral and subcutaneous preadipocyte differentiation efficiency in vitro. Almost all wild type subcutaneous preadipocytes differentiated into mature adipocytes. In contrast, visceral preadipocytes differentiated poorly. Exogenous activin A suppressed differentiation of preadipocytes from both depots. Smad2 conditional knockout (Smad2cKO) mice did not exhibit significant effects on weight gain, irrespective of diet, whereas Smad3 conditional knockout (Smad3cKO) male mice displayed a trend of reduced body weight on high-fat diet. On both diets, Smad3cKO mice displayed an adipose depot-selective phenotype, with a significant reduction in subcutaneous fat mass but not visceral fat mass. Our data suggest that Smad3 is an important contributor to the maintenance of subcutaneous white adipose tissue in a sex-selective fashion. These findings have implications for understanding SMAD-mediated, depot selective regulation of adipocyte growth and differentiation.


Subject(s)
Adipogenesis , Adipose Tissue, White/cytology , Adiposity , Intra-Abdominal Fat/cytology , Smad2 Protein/physiology , Smad3 Protein/physiology , Subcutaneous Fat/cytology , Activins/genetics , Activins/metabolism , Adipose Tissue, White/metabolism , Animals , Cell Differentiation , Diet, High-Fat , Female , Intra-Abdominal Fat/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Subcutaneous Fat/metabolism
10.
EMBO J ; 40(24): e106061, 2021 12 15.
Article in English | MEDLINE | ID: mdl-34459015

ABSTRACT

Non-neuronal cholinergic signaling, mediated by acetylcholine, plays important roles in physiological processes including inflammation and immunity. Our group first discovered evidence of non-neuronal cholinergic circuitry in adipose tissue, whereby immune cells secrete acetylcholine to activate beige adipocytes during adaptive thermogenesis. Here, we reveal that macrophages are the cellular protagonists responsible for secreting acetylcholine to regulate thermogenic activation in subcutaneous fat, and we term these cells cholinergic adipose macrophages (ChAMs). An adaptive increase in ChAM abundance is evident following acute cold exposure, and macrophage-specific deletion of choline acetyltransferase (ChAT), the enzyme for acetylcholine biosynthesis, impairs the cold-induced thermogenic capacity of mice. Further, using pharmacological and genetic approaches, we show that ChAMs are regulated via adrenergic signaling, specifically through the ß2 adrenergic receptor. These findings demonstrate that macrophages are an essential adipose tissue source of acetylcholine for the regulation of adaptive thermogenesis, and may be useful for therapeutic targeting in metabolic diseases.


Subject(s)
Acetylcholine/metabolism , Choline O-Acetyltransferase/genetics , Macrophages/metabolism , Receptors, Adrenergic, beta-2/metabolism , Subcutaneous Fat/cytology , Animals , Cells, Cultured , Cold Temperature , Gene Deletion , Gene Knockout Techniques , Mice , Primary Cell Culture , Subcutaneous Fat/metabolism , Thermogenesis
11.
Sci Rep ; 11(1): 17050, 2021 08 23.
Article in English | MEDLINE | ID: mdl-34426590

ABSTRACT

The study of metabolomics has improved our knowledge of the biology behind type 2 diabetes and its related metabolic physiology. We aimed to investigate markers of adipose tissue morphology, as well as insulin and glucose metabolism in 53 non-obese male individuals. The participants underwent extensive clinical, biochemical and magnetic resonance imaging phenotyping, and we also investigated non-targeted serum metabolites. We used a multi-modal machine learning approach to evaluate which serum metabolomic compounds predicted markers of glucose and insulin metabolism, adipose tissue morphology and distribution. Fasting glucose was associated with metabolites of intracellular insulin action and beta-cell dysfunction, namely cysteine-s-sulphate and n-acetylgarginine, whereas fasting insulin was predicted by myristoleoylcarnitine, propionylcarnitine and other metabolites of beta-oxidation of fatty acids. OGTT-glucose levels at 30 min were predicted by 7-Hoca, a microbiota derived metabolite, as well as eugenol, a fatty acid. Both insulin clamp and HOMA-IR were predicted by metabolites involved in beta-oxidation of fatty acids and biodegradation of triacylglycerol, namely tartrate and 3-phosphoglycerate, as well as pyruvate, xanthine and liver fat. OGTT glucose area under curve (AUC) and OGTT insulin AUC, was associated with bile acid metabolites, subcutaneous adipocyte cell size, liver fat and fatty chain acids and derivates, such as isovalerylcarnitine. Finally, subcutaneous adipocyte size was associated with long chain fatty acids, markers of sphingolipid metabolism, increasing liver fat and dopamine-sulfate 1. Ectopic liver fat was predicted by methylmalonate, adipocyte cell size, glutathione derived metabolites and fatty chain acids. Ectopic heart fat was predicted visceral fat, gamma-glutamyl tyrosine and 2-acetamidophenol sulfate. Adipocyte cell size, age, alpha-tocopherol and blood pressure were associated with visceral fat. We identified several biomarkers associated with adipose tissue pathophysiology and insulin and glucose metabolism using a multi-modal machine learning approach. Our approach demonstrated the relative importance of serum metabolites and they outperformed traditional clinical and biochemical variables for most endpoints.


Subject(s)
Adiposity , Blood Glucose/metabolism , Insulin/metabolism , Intra-Abdominal Fat/metabolism , Subcutaneous Fat/metabolism , Adult , Biomarkers/metabolism , Humans , Insulin-Secreting Cells/metabolism , Intra-Abdominal Fat/cytology , Liver/metabolism , Machine Learning , Male , Metabolome , Middle Aged , Subcutaneous Fat/cytology
12.
Am J Physiol Cell Physiol ; 321(3): C596-C606, 2021 09 01.
Article in English | MEDLINE | ID: mdl-34319829

ABSTRACT

Ceiling culture-derived preadipocytes (ccdPAs) and adipose-derived stem cells (ASCs) can be harvested from human subcutaneous fat tissue using the specific gravity method. Both cell types possess a similar spindle shape without lipid droplets. We previously reported that ccdPAs have a higher adipogenic potential than ASCs, even after a 7-wk culture. We performed a genome-wide epigenetic analysis to examine the mechanisms contributing to the adipogenic potential differences between ccdPAs and ASCs. Methylation analysis of cytosines followed by guanine (CpG) using a 450-K BeadChip was performed on human ccdPAs and ASCs isolated from three metabolically healthy females. Chromatin immunoprecipitation sequencing was performed to evaluate trimethylation at lysine 4 of histone 3 (H3K4me3). Unsupervised machine learning using t-distributed stochastic neighbor embedding to interpret 450,000-dimensional methylation assay data showed that the cells were divided into ASC and ccdPA groups. In Kyoto Encyclopedia of Genes and Genomes pathway analysis of 1,543 genes with differential promoter CpG methylation, the peroxisome proliferator-activated receptor (PPAR) and adipocytokine signaling pathways ranked in the top 10 pathways. In the PPARγ gene, H3K4me3 peak levels were higher in ccdPAs than in ASCs, whereas promoter CpG methylation levels were significantly lower in ccdPAs than in ASCs. Similar differences in promoter CpG methylation were also seen in the fatty acid-binding protein 4 and leptin genes. In conclusion, we analyzed the epigenetic status of adipogenesis-related genes as a potential mechanism underlying the differences in adipogenic differentiation capability between ASCs and ccdPAs.


Subject(s)
Adipocytes/metabolism , Adipogenesis/genetics , Adipokines/genetics , Epigenesis, Genetic , Mesenchymal Stem Cells/metabolism , PPAR gamma/genetics , Adipocytes/classification , Adipocytes/cytology , Adipokines/metabolism , CpG Islands , DNA Methylation , Fatty Acid-Binding Proteins/genetics , Fatty Acid-Binding Proteins/metabolism , Female , Gene Expression Profiling , Genome-Wide Association Study , Histones/genetics , Histones/metabolism , Humans , Leptin/genetics , Leptin/metabolism , Mammaplasty/methods , Mammary Glands, Human/cytology , Mammary Glands, Human/metabolism , Mammary Glands, Human/surgery , Mesenchymal Stem Cells/classification , Mesenchymal Stem Cells/cytology , Organ Specificity , PPAR gamma/metabolism , Primary Cell Culture , Subcutaneous Fat/cytology , Subcutaneous Fat/metabolism , Unsupervised Machine Learning
13.
BMC Vet Res ; 17(1): 244, 2021 Jul 15.
Article in English | MEDLINE | ID: mdl-34266445

ABSTRACT

BACKGROUND: The source of multipotent stromal cells (MSC) can have a significant influence on the health and expansion capacity of the cells. As the applications for allogeneic MSCs in the treatment of feline diseases increase, the location of the initial donor tissue must be analyzed. To date, comparisons have only been made between feline MSCs collected from bone marrow or abdominal fat. This is the first report to compare cells obtained from different adipose depots in the cat with a focus on clinically relevant donor tissues. The tissue was collected from 34 healthy cats undergoing spaying (fat around the ovaries and uterine horn) or subcutaneous fat collected during surgical procedures. RESULTS: The amount of starting material is essential to isolate sufficient MSCs. The total tissue yield from the subcutaneous fat was significantly greater than could be obtained from around the reproductive organs, leading to 3 times more MSCs per donor. However, the concentration of MSCs obtained from reproductive fat was higher than from subcutaneous fat. In addition, the viability of the MSCs from the reproductive fat was significantly higher than the subcutaneous fat. Since most spaying occurs in young cats (under 18 months) reproductive fat was collected from adult cats during spaying, illustrating that age did not alter the yield or viability of the MSCs. When sufficient tissue was collected, it was digested either mechanically or enzymatically. Mechanical digestion further decreased the viability and yield of MSCs from subcutaneous fat compared to enzymatic digestion. Biomarkers of stem cell characterization, expansion capacity and function were detected using qPCR. CD70, CD90 and CD105 were all expressed in high levels in the 3 groups. However, the reproductive fat had higher levels of CD73 with the mechanically digested subcutaneous fat having the least. Gata6 was detected in all samples while Sox2 and Sox17 were also detected with higher quantities found in the enzymatically digested subcutaneous fat. Negative control genes of Gata4 and Pdx1 showed no detection prior to 50 cycles. During the first three passages, age of the donor, location of the donor tissue, or digestion protocol had no effect on cell culture doubling times or cell viability. CONCLUSIONS: While MSCs from reproductive fat had superior cells/tissue weight and initial viability, there were still dramatically fewer cells obtained compared to subcutaneous fat due to the limited amount of tissue surrounding the reproductive organs. Further, in P1-P3 cultures there were no differences noted in doubling time or cell viability between tissue obtained from reproductive or subcutaneous fat depots.


Subject(s)
Cats , Intra-Abdominal Fat/cytology , Mesenchymal Stem Cells/cytology , Subcutaneous Fat/cytology , Animals , Cell Culture Techniques/methods , Cell Culture Techniques/veterinary , Cell Differentiation , Cell Proliferation , Cell Survival , Female , Genitalia, Female/surgery , Male , Mesenchymal Stem Cells/physiology
14.
Tissue Cell ; 71: 101583, 2021 Aug.
Article in English | MEDLINE | ID: mdl-34171520

ABSTRACT

The role of dermal white adipose tissue (dWAT) has emerged in the biomedical science as an ancillary fat district in the derma without a defined and distinct function respect to the subcutaneous adipose tissue (sWAT). Despite some evidence describing dWAT as an immune-competent compartment, particularly engaged in wound repair, very few reports dealing with dWAT has elucidated its major modulatory role within the skin biology. Whereas an increasing bulk of evidence allows researcher to describe the main activity of sWAT, in humans dWAT is not properly a separated fat compartment and therefore scarcely considered in the scientific debate. Due to its strategic position between epidermis and sWAT, dermal fat might play a much more intriguing role than expected. This review tries to shed light on this issue, by expanding the debate about a possible role of dWAT in skin physiology.


Subject(s)
Adipogenesis , Dermis , Lipid Metabolism , Subcutaneous Fat , Animals , Dermis/cytology , Dermis/metabolism , Humans , Subcutaneous Fat/cytology , Subcutaneous Fat/metabolism
15.
Dermatol Surg ; 47(8): 1059-1064, 2021 08 01.
Article in English | MEDLINE | ID: mdl-34115683

ABSTRACT

BACKGROUND: Radiofrequency (RF) and high-intensity focused electromagnetic (HIFEM) technologies are used for noninvasive body shaping as standalone modalities. OBJECTIVE: To examine the effects of novel synchronized RF and HIFEM on subcutaneous adipose tissue in a porcine animal model. MATERIALS AND METHODS: Seven large white pigs aged 6 months received 3 abdominal treatments of simultaneous application of synchronized RF and HIFEM (30 minutes, once per week). Punch biopsies of treated and control subcutaneous tissue were collected at the baseline, 4 days, 2 weeks, 1 month, and 2 months. Specimens were examined by light and scanning electron microscopy. Adipocyte volume was analyzed. Fat tissue temperature was measured in situ (fiber optic probes) and superficially (thermal imager). RESULTS: Fat layer was heated to temperatures of 42 to 45°C. Signs of fat apoptosis (shape alternations and pyknotic nuclei) appeared at day 4 and peaked between 2 weeks and 1 month. Adipocyte volume decreased significantly (p < .001) by 31.1% at 2 weeks, 1 month (-23.6%), and 2 months (-22.0%). Control samples showed healthy adipocytes. Scanning electron microscopy micrographs corroborated histology findings, showing flattened, volume-depleted and disrupted adipocytes. CONCLUSION: Synchronized RF with HIFEM procedure resulted in a significant and sustained fat reduction with no adverse events.


Subject(s)
Body Contouring/methods , Magnetic Field Therapy/methods , Radiofrequency Therapy/methods , Subcutaneous Fat/radiation effects , Adipocytes/radiation effects , Adipocytes/ultrastructure , Animals , Body Contouring/adverse effects , Body Contouring/instrumentation , Combined Modality Therapy/instrumentation , Combined Modality Therapy/methods , Female , Hot Temperature/adverse effects , Magnetic Field Therapy/adverse effects , Magnetic Field Therapy/instrumentation , Microscopy, Electron , Models, Animal , Radiofrequency Therapy/adverse effects , Radiofrequency Therapy/instrumentation , Subcutaneous Fat/cytology , Subcutaneous Fat/ultrastructure , Swine
16.
Nat Commun ; 12(1): 3482, 2021 06 09.
Article in English | MEDLINE | ID: mdl-34108488

ABSTRACT

Hyperplastic expansion of white adipose tissue (WAT) relies in part on the proliferation of adipocyte precursor cells residing in the stromal vascular cell fraction (SVF) of WAT. This study reveals a circadian clock- and feeding-induced diurnal pattern of cell proliferation in the SVF of visceral and subcutaneous WAT in vivo, with higher proliferation of visceral adipocyte progenitor cells subsequent to feeding in lean mice. Fasting or loss of rhythmic feeding eliminates this diurnal proliferation, while high fat feeding or genetic disruption of the molecular circadian clock modifies the temporal expression of proliferation genes and impinges on diurnal SVF proliferation in eWAT. Surprisingly, high fat diet reversal, sufficient to reverse elevated SVF proliferation in eWAT, was insufficient in restoring diurnal patterns of SVF proliferation, suggesting that high fat diet induces a sustained disruption of the adipose circadian clock. In conclusion, the circadian clock and feeding simultaneously impart dynamic, regulatory control of adipocyte progenitor proliferation, which may be a critical determinant of adipose tissue expansion and health over time.


Subject(s)
Adipose Tissue, White/cytology , Cell Proliferation , Circadian Rhythm/physiology , Adipocytes/cytology , Animals , Cell Proliferation/genetics , Circadian Clocks/genetics , Circadian Clocks/physiology , Circadian Rhythm/genetics , Diet, High-Fat , Epididymis/cytology , Fasting , Humans , Male , Mice , Stromal Cells/cytology , Subcutaneous Fat/cytology , Subcutaneous Fat/physiology
17.
Int J Obes (Lond) ; 45(8): 1811-1820, 2021 08.
Article in English | MEDLINE | ID: mdl-33993191

ABSTRACT

BACKGROUND: Excessive adiposity provides an inflammatory environment. However, in people with severe obesity, how systemic and local adipose tissue (AT)-derived cytokines contribute to worsening glucose tolerance is not clear. METHODS: Ninty-two severely obese (SO) individuals undergoing bariatric surgery were enrolled and subjected to detailed clinical phenotyping. Following an oral glucose tolerance test, participants were included in three groups, based on the presence of normal glucose tolerance (NGT), impaired glucose tolerance (IGT), or type 2 diabetes (T2D). Serum and subcutaneous AT (SAT) biopsies were obtained and mesenchymal stem cells (MSCs) were isolated, characterized, and differentiated in adipocytes in vitro. TNFA and PPARG mRNA levels were determined by qRT-PCR. Circulating, adipocyte- and MSC-released cytokines, chemokines, and growth factors were assessed by multiplex ELISA. RESULTS: Serum levels of IL-9, IL-13, and MIP-1ß were increased in SO individuals with T2D, as compared with those with either IGT or NGT. At variance, SAT samples obtained from SO individuals with IGT displayed levels of TNFA which were threefold higher compared to those with NGT, but not different from those with T2D. Elevated levels of TNFα were also found in differentiated adipocytes, isolated from the SAT specimens of individuals with IGT and T2D, compared to those with NGT. Consistent with the pro-inflammatory milieu, IL-1ß and IP-10 secretion was significantly higher in adipocytes from individuals with IGT and T2D. Moreover, increased levels of TNFα, both mRNA and secreted protein were detected in MSCs obtained from IGT and T2D, compared to NGT SO individuals. Exposure of T2D and IGT-derived MSCs to the anti-inflammatory flavonoid quercetin reduced TNFα levels and was paralleled by a significant decrease of the secretion of inflammatory cytokines. CONCLUSION: In severe obesity, enhanced SAT-derived inflammatory phenotype is an early step in the progression toward T2D and maybe, at least in part, attenuated by quercetin.


Subject(s)
Cytokines/metabolism , Glucose Intolerance/metabolism , Obesity, Morbid , Quercetin/pharmacology , Subcutaneous Fat , Adult , Blood Glucose/drug effects , Cells, Cultured , Female , Glucose Tolerance Test , Humans , Male , Middle Aged , Obesity, Morbid/metabolism , Obesity, Morbid/physiopathology , Subcutaneous Fat/cytology , Subcutaneous Fat/drug effects , Subcutaneous Fat/metabolism , Subcutaneous Fat/physiopathology , Young Adult
18.
Dev Cell ; 56(10): 1437-1451.e3, 2021 05 17.
Article in English | MEDLINE | ID: mdl-33878347

ABSTRACT

Adipose tissue mass and adiposity change throughout the lifespan. During aging, while visceral adipose tissue (VAT) tends to increase, peripheral subcutaneous adipose tissue (SAT) decreases significantly. Unlike VAT, which is linked to metabolic diseases, including type 2 diabetes, SAT has beneficial effects. However, the molecular details behind the aging-associated loss of SAT remain unclear. Here, by comparing scRNA-seq of total stromal vascular cells of SAT from young and aging mice, we identify an aging-dependent regulatory cell (ARC) population that emerges only in SAT of aged mice and humans. ARCs express adipose progenitor markers but lack adipogenic capacity; they secrete high levels of pro-inflammatory chemokines, including Ccl6, to inhibit proliferation and differentiation of neighboring adipose precursors. We also found Pu.1 to be a driving factor for ARC development. We identify an ARC population and its capacity to inhibit differentiation of neighboring adipose precursors, correlating with aging-associated loss of SAT.


Subject(s)
Adipogenesis , Aging/physiology , Subcutaneous Fat/cytology , 3T3-L1 Cells , Adipocytes/cytology , Adipocytes/metabolism , Animals , CD36 Antigens/metabolism , Cell Proliferation , Chemokines/metabolism , Galectin 3/metabolism , Mice , Proto-Oncogene Proteins/metabolism , Stem Cells/cytology , Trans-Activators/metabolism
19.
Front Immunol ; 12: 651109, 2021.
Article in English | MEDLINE | ID: mdl-33790914

ABSTRACT

Mesenchymal stromal cells (MSC) are a promising therapy for inflammatory diseases. However, MSC are large and become trapped in the lungs after intravenous infusion, where they have a short survival time. To steer MSC immunoregulatory therapy beyond the lungs, we generated nm-sized particles from MSC membranes (membrane particles, MP), which have immunomodulatory properties, and investigated their internalization and mode of interaction in macrophages subtypes and human umbilical vein endothelial cells (HUVEC) under control and inflammatory conditions. We found that macrophages and HUVEC take up MP in a dose, time, and temperature-dependent manner. Specific inhibitors for endocytotic pathways revealed that MP internalization depends on heparan sulfate proteoglycan-, dynamin-, and clathrin-mediated endocytosis but does not involve caveolin-mediated endocytosis. MP uptake also involved the actin cytoskeleton and phosphoinositide 3-kinase, which are implicated in macropinocytosis and phagocytosis. Anti-inflammatory M2 macrophages take up more MP than pro-inflammatory M1 macrophages. In contrast, inflammatory conditions did not affect the MP uptake by HUVEC. Moreover, MP induced both anti- and pro-inflammatory responses in macrophages and HUVEC by affecting gene expression and cell surface proteins. Our findings on the mechanisms of uptake of MP under different conditions help the development of target-cell specific MP therapy to modulate immune responses.


Subject(s)
Cell- and Tissue-Based Therapy/methods , Cell-Derived Microparticles/immunology , Mesenchymal Stem Cells/cytology , Cell-Derived Microparticles/transplantation , Cells, Cultured , Dose-Response Relationship, Immunologic , Healthy Volunteers , Human Umbilical Vein Endothelial Cells , Humans , Macrophages/immunology , Mesenchymal Stem Cells/immunology , Phagocytosis/immunology , Pinocytosis/immunology , Primary Cell Culture , Subcutaneous Fat/cytology
20.
Cells ; 10(3)2021 03 11.
Article in English | MEDLINE | ID: mdl-33799840

ABSTRACT

Reactive oxygen species (ROS)-induced oxidative stress in adipose tissue is associated with inflammation and the development of obesity-related metabolic disorders. The aim of this study is to investigate the effects of hydrogen nano-bubble water (HW) on ROS generation, adipogenesis, and interleukin-6 (IL-6) secretion in hydrogen peroxide (H2O2) or phorbol 12-myristate 13-acetate (PMA)-stimulated OP9 adipocytes, and three-dimensional (3D) subcutaneous adipose equivalents. Nanoparticle tracking analysis showed that fresh HW contains 1.17 × 108/mL of nano-sized hydrogen bubbles. Even after 8 to 13 months of storage, approximately half of the bubbles still remained in the water. CellROX® staining showed that HW could diminish H2O2- or PMA-induced intracellular ROS generation in human keratinocytes HaCaT and OP9 cells. We discovered that PMA could markedly increase lipid accumulation to 180% and IL-6 secretion 2.7-fold in OP9 adipocytes. Similarly, H2O2 (5 µM) also significantly stimulated lipid accumulation in OP9 cells and the 3D adipose equivalents. HW treatment significantly repressed H2O2- or PMA-induced lipid accumulation and IL-6 secretion in OP9 adipocytes and the 3D adipose equivalents. In conclusion, HW showed a possibility of repressing oxidative stress, inflammatory response, and adipogenesis at cellular/tissue levels. It can be used for preventing the development of metabolic disorders amongst obese people.


Subject(s)
Adipocytes/drug effects , Adipogenesis/drug effects , Anti-Inflammatory Agents/pharmacology , Antioxidants/pharmacology , Hydrogen Peroxide/pharmacology , Hydrogen/pharmacology , Interleukin-6/metabolism , Reactive Oxygen Species/metabolism , Subcutaneous Fat/drug effects , Tetradecanoylphorbol Acetate/pharmacology , Water/pharmacology , Adipocytes/metabolism , Animals , Coculture Techniques , Humans , Keratinocytes/drug effects , Keratinocytes/metabolism , Mice , Nanoparticles , Oxidative Stress/drug effects , Signal Transduction , Subcutaneous Fat/cytology , Subcutaneous Fat/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...