Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
1.
Immunity ; 47(6): 1154-1168.e6, 2017 12 19.
Article in English | MEDLINE | ID: mdl-29221731

ABSTRACT

White adipose tissue bridges body organs and plays a fundamental role in host metabolism. To what extent adipose tissue also contributes to immune surveillance and long-term protective defense remains largely unknown. Here, we have shown that at steady state, white adipose tissue contained abundant memory lymphocyte populations. After infection, white adipose tissue accumulated large numbers of pathogen-specific memory T cells, including tissue-resident cells. Memory T cells in white adipose tissue expressed a distinct metabolic profile, and white adipose tissue from previously infected mice was sufficient to protect uninfected mice from lethal pathogen challenge. Induction of recall responses within white adipose tissue was associated with the collapse of lipid metabolism in favor of antimicrobial responses. Our results suggest that white adipose tissue represents a memory T cell reservoir that provides potent and rapid effector memory responses, positioning this compartment as a potential major contributor to immunological memory.


Subject(s)
Adipose Tissue, White/transplantation , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Immunologic Memory , Toxoplasmosis/immunology , Yersinia pseudotuberculosis Infections/immunology , Adipose Tissue, White/immunology , Animals , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , CD4-Positive T-Lymphocytes/microbiology , CD4-Positive T-Lymphocytes/parasitology , CD8-Positive T-Lymphocytes/microbiology , CD8-Positive T-Lymphocytes/parasitology , Gene Expression , Genes, Reporter , Interferon-gamma/genetics , Interferon-gamma/immunology , Interleukin-17/genetics , Interleukin-17/immunology , Interleukin-5/genetics , Interleukin-5/immunology , Lipid Metabolism , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Survival Analysis , Tissue Transplantation , Toxoplasma/immunology , Toxoplasmosis/genetics , Toxoplasmosis/mortality , Toxoplasmosis/parasitology , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology , Yersinia pseudotuberculosis/immunology , Yersinia pseudotuberculosis Infections/genetics , Yersinia pseudotuberculosis Infections/microbiology , Yersinia pseudotuberculosis Infections/mortality
2.
Aesthetic Plast Surg ; 48(9): 1817-1824, 2024 May.
Article in English | MEDLINE | ID: mdl-38409345

ABSTRACT

BACKGROUND: In plastic surgery, autologous fat grafts (AFG) play an important role because of their abundant supply, biocompatibility, and low rejection rate. However, the lower retention rate of fat grafts limits their widespread use. Brown adipose tissue (BAT) can promote angiogenesis and regulate the level of associated inflammation. This study explored whether BAT has a facilitative effect on fat graft retention. METHODS: We obtained white adipose tissue (WAT) from c57 mice and combined it with either BAT from c57 mice or phosphate-buffered saline (PBS) as a control. These mixtures were injected subcutaneously into the back of thymus-free nude mice. After 12 weeks, fat grafts were harvested, weighed, and analyzed. RESULTS: We found that the BAT-grafted group had higher mass retention, more mature adipocytes, and higher vascularity than the other group. Further analysis revealed that BAT inhibited M1 macrophages; down-regulated IL-6, IL-1ß, and TNF-ß; upregulated M2 macrophages and Vascular endothelial growth factor-A (VEGFA); and promoted adipocyte regeneration by inhibiting the Wnt/ß-catenin pathway, which together promoted adipose graft retention. CONCLUSION: The study demonstrated that BAT improved adipose graft retention by promoting angiogenesis, inhibiting tissue inflammation levels and the Wnt/ß-catenin pathway. LEVEL OF EVIDENCE III: This journal requires that authors assign a level of evidence to each article. For a full description of these Evidence-Based Medicine ratings, please refer to the Table of Contents or the online Instructions to Authors www.springer.com/00266.


Subject(s)
Adipose Tissue, Brown , Graft Survival , Mice, Inbred C57BL , Mice, Nude , Wnt Signaling Pathway , Animals , Adipose Tissue, Brown/transplantation , Mice , Wnt Signaling Pathway/physiology , Transplantation, Autologous , Random Allocation , Male , Adipose Tissue, White/transplantation , Adipose Tissue, White/metabolism , Disease Models, Animal
3.
Aesthet Surg J ; 44(7): NP486-NP500, 2024 Jun 14.
Article in English | MEDLINE | ID: mdl-38518754

ABSTRACT

BACKGROUND: Induction of beige fat for grafting is an emerging transplantation strategy. However, safety concerns associated with pharmaceutical interventions limit its wider application. Moreover, because beige fat is a special type of fat with strong metabolic functions, its effect on the metabolism of recipients after grafting has not been explored in the plastic surgery domain. OBJECTIVES: The aim of this study was to explore whether cold-induced inguinal white adipose tissue (iWAT) transplantation has a higher retention rate and beneficial effects on recipient metabolism. METHODS: C57/BL6 mice were subjected to cold stimulation for 48 hours to induce the browning of iWAT and harvested immediately. Subsequently, each mouse received a transplant of 0.2 mL cold-induced iWAT or normal iWAT. Fat grafts and recipients' iWAT, epididymal adipose tissue, and brown adipose tissue were harvested at 8 weeks after operation. Immunofluorescence staining, real-time polymerase chain reaction, and western blot were used for histological and molecular analysis. RESULTS: Cold-induced iWAT grafting had a higher mean [standard error of the mean] retention rate (67.33% [1.74%] vs 55.83% [2.94%], P < .01) and more satisfactory structural integrity than normal iWAT. Histological changes identified improved adipose tissue homeostasis after cold challenge, including abundant smaller adipocytes, higher levels of adipogenesis, angiogenesis, and proliferation, but lower levels of fibrosis. More importantly, cold-induced iWAT grafting suppressed the inflammation of epididymal adipose tissue caused by conventional fat grafting, and activated the glucose metabolism and thermogenic activity of recipients' adipose tissues. CONCLUSIONS: Cold-induced iWAT grafting is an effective nonpharmacological intervention strategy to improve the retention rate and homeostasis of grafts. Furthermore, it improves the adverse effects caused by traditional fat grafting, while also conferring metabolic benefits.


Subject(s)
Adipose Tissue, Brown , Cold Temperature , Mice, Inbred C57BL , Subcutaneous Fat , Animals , Male , Subcutaneous Fat/transplantation , Subcutaneous Fat/metabolism , Mice , Adipose Tissue, Brown/transplantation , Adipose Tissue, Brown/metabolism , Adipose Tissue, White/metabolism , Adipose Tissue, White/transplantation , Adipose Tissue, Beige/metabolism , Graft Survival
4.
Nature ; 542(7642): 450-455, 2017 02 23.
Article in English | MEDLINE | ID: mdl-28199304

ABSTRACT

Adipose tissue is a major site of energy storage and has a role in the regulation of metabolism through the release of adipokines. Here we show that mice with an adipose-tissue-specific knockout of the microRNA (miRNA)-processing enzyme Dicer (ADicerKO), as well as humans with lipodystrophy, exhibit a substantial decrease in levels of circulating exosomal miRNAs. Transplantation of both white and brown adipose tissue-brown especially-into ADicerKO mice restores the level of numerous circulating miRNAs that are associated with an improvement in glucose tolerance and a reduction in hepatic Fgf21 mRNA and circulating FGF21. This gene regulation can be mimicked by the administration of normal, but not ADicerKO, serum exosomes. Expression of a human-specific miRNA in the brown adipose tissue of one mouse in vivo can also regulate its 3' UTR reporter in the liver of another mouse through serum exosomal transfer. Thus, adipose tissue constitutes an important source of circulating exosomal miRNAs, which can regulate gene expression in distant tissues and thereby serve as a previously undescribed form of adipokine.


Subject(s)
Adipose Tissue/metabolism , Gene Expression Regulation , MicroRNAs/blood , MicroRNAs/metabolism , Paracrine Communication , 3' Untranslated Regions/genetics , Adipokines/metabolism , Adipose Tissue/transplantation , Adipose Tissue, Brown/cytology , Adipose Tissue, Brown/metabolism , Adipose Tissue, Brown/transplantation , Adipose Tissue, White/metabolism , Adipose Tissue, White/transplantation , Animals , Exosomes/genetics , Fibroblast Growth Factors/blood , Fibroblast Growth Factors/genetics , Genes, Reporter/genetics , Glucose Tolerance Test , Liver/metabolism , Male , Mice , MicroRNAs/genetics , Models, Biological , Organ Specificity/genetics , RNA, Messenger/genetics , Ribonuclease III/deficiency , Ribonuclease III/genetics , Transcription, Genetic
5.
Aesthet Surg J ; 41(8): NP1086-NP1091, 2021 07 14.
Article in English | MEDLINE | ID: mdl-33824956

ABSTRACT

BACKGROUND: Browning adipocytes induced by burn and cancer were assumed less viable and more prone to necrosis for their hypermetabolic properties. Recent studies have shown browning of white adipose after fat engraftment in mice. OBJECTIVES: The authors sought to evaluate whether fat transfer could induce browning biogenesis in fat grafts in humans and if it is associated with graft necrosis. METHODS: Necrotic adipose grafts were excised from 11 patients diagnosed with fat necrosis after fat grafting or flap transfer. Non-necrotic fat grafts were from 5 patients who underwent revisionary surgeries after flap transfer. Histology and electronic microscopy as well as protein and gene expression of browning-related marker analyses were performed. RESULTS: Fat grafts with necrosis demonstrated a higher gene expression level of uncoupling protein-1 (greater than fivefold increase, **P < 0.01), a master beige adipocyte marker, than non-necrotic fat grafts. Electronic microscopy and histology showed that browning adipocytes were presented in necrotic adipose in patients. CONCLUSIONS: Fat transfer induced browning adipocytes in patients and was evident in patients with postgrafting necrosis.


Subject(s)
Adipocytes, White , Adipose Tissue, Brown , Adipose Tissue, White/transplantation , Humans , Uncoupling Protein 1
6.
Cardiovasc Drugs Ther ; 32(5): 503-510, 2018 10.
Article in English | MEDLINE | ID: mdl-30097828

ABSTRACT

PURPOSE: Perivascular adipose tissue (PVAT) surrounds the arterial adventitia and plays an important role in vascular homeostasis. PVAT expands in obesity, and inflamed PVAT can locally promote endothelial dysfunction and atherosclerosis. Here, using adipose tissue transplantation, we tested the hypothesis that expansion of PVAT can also remotely exacerbate vascular disease. METHODS: Fifty milligrams of abdominal aortic PVAT was isolated from high-fat diet (HFD)-fed wild-type mice and transplanted onto the abdominal aorta of lean LDL receptor knockout mice. Subcutaneous and visceral adipose tissues were used as controls. After HFD feeding for 10 weeks, body weight, glucose/insulin sensitivity, and lipid levels were measured. Adipocytokine gene expression was assessed in the transplanted adipose tissues, and the thoracic aorta was harvested to quantify atherosclerotic lesions by Oil-Red O staining and to assess vasorelaxation by wire myography. RESULTS: PVAT transplantation did not influence body weight, fat composition, lipid levels, or glucose/insulin sensitivity. However, as compared with controls, transplantation of PVAT onto the abdominal aorta increased thoracic aortic atherosclerosis. Furthermore, PVAT transplantation onto the abdominal aorta inhibited endothelium-dependent relaxation in the thoracic aorta. MCP-1 and TNF-α expression was elevated, while adiponectin expression was reduced, in the transplanted PVAT tissue, suggesting augmented inflammation as a potential mechanism for the remote vascular effects of transplanted PVAT. CONCLUSIONS: These data suggest that PVAT expansion and inflammation in obesity can remotely induce endothelial dysfunction and augment atherosclerosis. Identifying the underlying mechanisms may lead to novel approaches for risk assessment and treatment of obesity-related vascular disease.


Subject(s)
Adipose Tissue, White/transplantation , Aorta, Abdominal/metabolism , Aorta, Abdominal/surgery , Aorta, Thoracic/metabolism , Atherosclerosis/metabolism , Paracrine Communication , Plaque, Atherosclerotic , Adiponectin/metabolism , Adipose Tissue, White/metabolism , Adiposity , Animals , Aorta, Abdominal/pathology , Aorta, Abdominal/physiopathology , Aorta, Thoracic/pathology , Aorta, Thoracic/physiopathology , Atherosclerosis/genetics , Atherosclerosis/pathology , Atherosclerosis/physiopathology , Chemokine CCL2/metabolism , Diet, High-Fat , Disease Models, Animal , Disease Progression , Inflammation Mediators/metabolism , Mice, Inbred C57BL , Mice, Knockout , Receptors, LDL/deficiency , Receptors, LDL/genetics , Signal Transduction , Tumor Necrosis Factor-alpha/metabolism , Vasodilation
7.
Am J Physiol Regul Integr Comp Physiol ; 309(12): R1499-511, 2015 Dec 15.
Article in English | MEDLINE | ID: mdl-26468261

ABSTRACT

Fat transplants increase body fat mass without changing the energy status of an animal and provide a tool for investigating control of total body fat. Early transplant studies found that small pieces of transplanted fat took on the morphology of the transplant recipient. Experiments described here tested whether this response was dependent upon expression of leptin receptors in either transplanted fat or the recipient mouse. Fat from leptin receptor deficient db/db mice or wild-type mice was placed subcutaneously in db/db mice. After 12 wk, cell size distribution in the transplant was the same as in endogenous fat of the recipient. Thus, wild-type fat cells, which express leptin receptors, were enlarged in a hyperleptinemic environment, indicating that leptin does not directly control adipocyte size. By contrast, db/db or wild-type fat transplanted into wild-type mice decreased in size, suggesting that a functional leptin system in the recipient is required for body fat mass to be controlled. In the final experiment, wild-type fat was transplanted into a db/db mouse parabiosed to either another db/db mouse to an ob/ob mouse or in control pairs in which both parabionts were ob/ob mice. Transplants increased in size in db/db-db/db pairs, decreased in db/db-ob/ob pairs and did not change in ob/ob-ob/ob pairs. We propose that leptin from db/db parabionts activated leptin receptors in their ob/ob partners. This, in turn, stimulated release of unidentified circulating factors, which travelled back to the db/db partner and acted on the transplant to reduce fat cell size.


Subject(s)
Adipocytes, White/metabolism , Adipose Tissue, White/metabolism , Adiposity , Leptin/blood , Adipocytes, White/transplantation , Adipose Tissue, White/transplantation , Animals , Biomarkers/blood , Cell Size , Female , Male , Mice, Knockout , Parabiosis , Receptors, Leptin/deficiency , Receptors, Leptin/genetics , Signal Transduction
8.
Blood ; 120(25): 4952-62, 2012 Dec 13.
Article in English | MEDLINE | ID: mdl-23071275

ABSTRACT

White adipose tissue (WAT) is the focus of new interest because of the presence of an abundant and complex immune cell population that is involved in key pathologies such as metabolic syndrome. Based on in vivo reconstitution assays, it is thought that these immune cells are derived from the bone marrow (BM). However, previous studies have shown that WAT exhibits specific hematopoietic activity exerted by an unknown subpopulation of cells. In the present study, we prospectively isolated a peculiar hematopoietic stem/progenitor cell population from murine WAT. The cells are phenotypically similar to BM hematopoietic stem cells and are able to differentiate into both myeloid and lymphoid lineages in vitro. In competitive repopulation assays in vivo, they reconstituted the innate immune compartment in WAT preferentially and more efficiently than BM cells, but did not reconstitute hematopoietic organs. They were also able to give rise to multilineage engraftment in both secondary recipients and in utero transplantation. Therefore, we propose that WAT hematopoietic cells constitute a population of immature cells that are able to renew innate immune cell populations. Considering the amount of WAT in adults, our results suggest that WAT hematopoietic activity controls WAT inflammatory processes and also supports innate immune responses in other organs.


Subject(s)
Adipose Tissue, White/cytology , Adipose Tissue, White/immunology , Hematopoietic Stem Cells/cytology , Lymphocytes/cytology , Myeloid Cells/cytology , Adipose Tissue, White/transplantation , Animals , Antigens, Ly/analysis , Cell Differentiation , Female , Hematopoietic Stem Cells/immunology , Immunity, Innate , Killer Cells, Natural/cytology , Killer Cells, Natural/immunology , Lymphocytes/immunology , Male , Membrane Proteins/analysis , Mice , Mice, Inbred C57BL , Myeloid Cells/immunology , Proto-Oncogene Proteins c-kit/analysis
9.
Ann Plast Surg ; 69(2): 213-9, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22772067

ABSTRACT

Vascularization is crucial for implantation of engineered tissues in reconstructive surgery. Polypeptides encapsulated in microspheres can be efficiently transported to their site of action and released in a sustained dosage. We evaluated the effect of delivering vascular endothelial growth factor (VEGF)-encapsulated microspheres in a lipoaspirate scaffold on vascularization and tissue survival. The VEGF-loaded (n=6) and empty (n=6) poly(lactic-co-glycolic acid) microspheres in human lipoaspirate and the human lipoaspirate alone (n=6) were injected subcutaneously into the flanks of athymic nude mice. Three mice from each group were killed, and grafts were explanted at weeks 3 and 6. Increases in mass and volume of VEGF samples, as well as decreases in empty and lipoaspirate-only samples, were observed at 3 and 6 weeks, reaching statistical significance at 6 weeks. Hematoxylin and eosin and CD31+ imaging demonstrated significantly greater vascularization in VEGF samples than in both the empty and lipoaspirate-only groups at both 3 and 6 weeks.


Subject(s)
Adipose Tissue, White/transplantation , Angiogenesis Inducing Agents/pharmacology , Guided Tissue Regeneration/methods , Microspheres , Neovascularization, Physiologic/drug effects , Tissue Scaffolds , Vascular Endothelial Growth Factor A/pharmacology , Adipose Tissue, White/blood supply , Adipose Tissue, White/growth & development , Angiogenesis Inducing Agents/administration & dosage , Animals , Female , Graft Survival , Humans , Lipectomy , Mice , Mice, Nude , Vascular Endothelial Growth Factor A/administration & dosage
10.
Diabetologia ; 54(11): 2890-9, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21805228

ABSTRACT

AIMS/HYPOTHESIS: Intra-abdominal transplantation of non-visceral adipose tissue in rodents, simulating increased abdominal fat in obesity, paradoxically improves glucose tolerance and insulin sensitivity. We hypothesised that this improvement is due to transplant-induced enhanced uptake of fatty acids by adipose tissue, thus reducing fatty acid flux into, and triacylglycerol storage in, the liver. METHODS: In Experiment 1, mice were sham-operated or received heterologous epididymal white adipose tissue (WAT; EWAT) or visceral WAT (VWAT) transplantation to the portal and splanchnic circulation regions in the visceral cavity. In Experiment 2, inguinal WAT (IWAT) or EWAT was removed and subsequently transplanted to the visceral cavity of the same mouse (autotransplant). IWAT and EWAT autotransplants were repeated in Experiment 3 and compared with heterotransplants. RESULTS: Heterotransplantation of VWAT did not alter glucose tolerance, whereas auto- or hetero-transplantation of EWAT or IWAT significantly improved glucose tolerance. Transplantation-induced improvements in glucose tolerance 4 weeks after surgery coincided with decreased liver triacylglycerol, decreased portal plasma lipids and increased hepatic insulin sensitivity. By 8 weeks, these changes were apparent only in mice with autotransplantation. Heterologous EWAT transplantation-induced glucose improvement persisted without altered liver metabolism. CONCLUSIONS/INTERPRETATION: Increases in visceral fat, via transplantation of visceral or non-visceral adipose tissue, is not a major risk factor for glucose intolerance. In fact, there are dynamic metabolic improvements following transplantation that include decreased portal lipids and improved liver metabolism, but these improvements are transient under certain circumstances.


Subject(s)
Glucose Intolerance/etiology , Insulin Resistance , Lipid Metabolism , Liver/metabolism , Obesity, Abdominal/physiopathology , Adipose Tissue, White/metabolism , Adipose Tissue, White/pathology , Adipose Tissue, White/transplantation , Animals , Disease Models, Animal , Epididymis , Glucose Intolerance/prevention & control , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Intra-Abdominal Fat/metabolism , Intra-Abdominal Fat/pathology , Intra-Abdominal Fat/transplantation , Lipids/blood , Liver/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Obesity, Abdominal/blood , Obesity, Abdominal/metabolism , Obesity, Abdominal/pathology , Peritoneum/surgery , Recombinant Proteins/metabolism , Transplantation, Autologous , Transplantation, Homologous
11.
Int J Med Sci ; 8(3): 231-8, 2011 Mar 14.
Article in English | MEDLINE | ID: mdl-21448310

ABSTRACT

Adipose-derived mesenchymal stem cells (AdMSCs) augment the ability to contribute to microvascular remodeling in vivo and to modulate vascular stability in fresh fat grafts. Although cryopreserved adipose tissue is frequently used for soft tissue augmentation, the viability of the fat graft is poor. The effects of culture-expanded human adipose tissue-derived mesenchymal stem cells (hAdMSCs) on the survival and quality of the cryopreserved fat graft were determined. hAdMSCs from the same donor were mixed with fat tissues cryopreserved at -70 °C for 8 weeks and injected subcutaneously into 6-week-old BALB/c-nu nude mice. Graft volume and weight were measured, and histology was evaluated 4 and 15 weeks post-transplantation. The hAdMSC-treated group showed significantly enhanced graft volume and weight. The histological evaluation demonstrated significantly better fat cell integrity compared with the vehicle-treated control 4 weeks post-transplantation. No significant difference in graft weight, volume, or histological parameters was found among the groups 15 weeks post-transplantation. The hAdMSCs enhanced the survival and quality of transplanted cryopreserved fat tissues. Cultured and expanded hAdMSCs have reconstructive capacity in cryopreserved fat grafting by increasing the number of stem cells.


Subject(s)
Adipose Tissue, White/transplantation , Cryopreservation , Graft Survival/physiology , Mesenchymal Stem Cells/cytology , Tissue Transplantation/methods , Adipocytes, White/pathology , Adipose Tissue, White/cytology , Adipose Tissue, White/pathology , Animals , Cysts/pathology , Fibrosis/pathology , Humans , Inflammation/pathology , Male , Mesenchymal Stem Cell Transplantation , Mice , Mice, Inbred BALB C , Mice, Nude , Necrosis/pathology , Tissue Transplantation/pathology
12.
Plast Reconstr Surg ; 145(5): 1183-1195, 2020 05.
Article in English | MEDLINE | ID: mdl-32332538

ABSTRACT

BACKGROUND: Growing evidence has demonstrated that adipose-derived stem cell-derived extracellular vesicles enhance the survival of fat grafts and the browning of white adipose tissue. We evaluated whether supplementation with adipose-derived stem cell-derived extracellular vesicles promotes the survival and browning of fat grafts. METHODS: Extracellular vesicles derived from adipose-derived stem cells were injected into fat grafts of C57BL/6 mice once per week until postgraft week 12. The grafts were collected and weighed after postgraft weeks 2, 4, and 12. The histological morphology, neovascularization, and the proportion of M2 macrophages of grafts were evaluated. The ability of extracellular vesicles to promote macrophage polarization and catecholamine secretion was detected. Whether the inducement of browning adipose differentiation is extracellular vesicles or the paracrine effect of M2 macrophages polarized by extracellular vesicles was also verified. RESULTS: Grafts treated by extracellular vesicles derived from adipose-derived stem cells showed enhanced beige adipose regeneration with increased neovascularization, M2 macrophage proportion, and norepinephrine secretion at postgraft week 4. Increased retention and decreased fibrosis and necrosis were noted at postgraft week 12. The extracellular vesicles uptake by macrophages promoted M2 type polarization and catecholamine secretion while suppressing M1 type polarization. Of note, browning adipose differentiation with enhanced energy expenditure could be promoted only by the conditioned medium from extracellular vesicle-polarized M2 macrophages but not by extracellular vesicles themselves. CONCLUSIONS: Supplementation with extracellular vesicles derived from adipose-derived stem cells increases fat graft survival and browning by which extracellular vesicles-polarized M2 macrophages secrete catecholamines to promote beige adipose regeneration.


Subject(s)
Adipose Tissue, Beige/physiology , Adipose Tissue, White/transplantation , Extracellular Vesicles/transplantation , Graft Survival/physiology , Stem Cells/cytology , Adipose Tissue, White/cytology , Adipose Tissue, White/physiology , Adult , Animals , Catecholamines/metabolism , Cell Differentiation , Female , Humans , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Models, Animal , Regeneration , Young Adult
13.
FASEB J ; 22(12): 4146-53, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18708591

ABSTRACT

Adipose tissue is an active endocrine organ that produces a variety of secretory factors involved in the initiation of angiogenic processes. The bioactive peptide apelin is the endogenous ligand of the G protein-coupled receptor, APJ. Here we investigated the potential role of apelin and its receptor, APJ, in the angiogenic responses of human endothelial cells and the development of a functional vascular network in a model of adipose tissue development in mice. Treatment of human umbilical vein endothelial cells with apelin dose-dependently increased angiogenic responses, including endothelial cell migration, proliferation, and Matrigel(R) capillary tubelike structure formation. These endothelial effects of apelin were due to activation of APJ, because siRNA directed against APJ, which led to long-lasting down-regulation of APJ mRNA, abolished cell migration induced by apelin in contrast to control nonsilencing siRNA. Hypoxia up-regulated the expression of apelin in 3T3F442A adipocytes, and we therefore determined whether apelin could play a role in adipose tissue angiogenesis in vivo. Epididymal white adipose tissue (EWAT) transplantation was performed as a model of adipose tissue angiogenesis. Transplantation led to increased apelin mRNA levels 2 and 5 days after transplantation associated with tissue hypoxia, as evidenced by hydroxyprobe staining on tissue sections. Graft revascularization evolved in parallel, as the first functional vessels in EWAT grafts were observed 2 days after transplantation and a strong angiogenic response was apparent on day 14. This was confirmed by determination of graft hemoglobin levels, which are indicative of functional vascularization and were strongly increased 5 and 14 days after transplantation. The role of apelin in the graft neovascularization was then assessed by local delivery of stable complex apelin-targeting siRNA leading to dramatically reduced apelin mRNA levels and vascularization (quantified by hemogloblin content) in grafted EWAT on day 5 when compared with control siRNA. Taken together, our data provide the first evidence that apelin/APJ signaling pathways play a critical role in the development of the functional vascular network in adipose tissue. In addition, we have shown that adipocyte-derived apelin can be up-regulated by hypoxia. These findings provide novel insights into the complex relationship between adipose tissue and endothelial vascular function and may lead to new therapeutic strategies to modulate angiogenesis.


Subject(s)
Adipose Tissue, White/physiology , Carrier Proteins/physiology , Endothelial Cells/physiology , Intercellular Signaling Peptides and Proteins/physiology , Neovascularization, Physiologic/physiology , Receptors, G-Protein-Coupled/physiology , 3T3 Cells , Adipokines , Adipose Tissue, White/transplantation , Animals , Apelin , Apelin Receptors , Cell Movement , Down-Regulation , Humans , Hypoxia/physiopathology , Male , Mice , Mice, Inbred C57BL , RNA, Small Interfering/pharmacology
14.
Diabetes ; 68(8): 1614-1623, 2019 08.
Article in English | MEDLINE | ID: mdl-31167880

ABSTRACT

The kinin B1 receptor (B1R) plays a role in inflammatory and metabolic processes. B1R deletion (B1 -/-) protects mice from diet-induced obesity and improves insulin and leptin sensitivity. In contrast, genetic reconstitution of B1R exclusively in adipose tissue reverses the lean phenotype of B1 -/- mice. To study the cell-nonautonomous nature of these effects, we transplanted epididymal white adipose tissue (eWAT) from wild-type donors (B1 +/+) into B1 -/- mice (B1 +/+→B1 -/-) and compared them with autologous controls (B1 +/+→B1 +/+ or B1 -/-→B1 -/-). We then fed these mice a high-fat diet for 16 weeks and investigated their metabolic phenotypes. B1 +/+→B1 -/- mice became obese but not glucose intolerant or insulin resistant, unlike B1 -/-→B1 -/- mice. Moreover, the endogenous adipose tissue of B1 +/+→B1 -/- mice exhibited higher expression of adipocyte markers (e.g., Fabp4 and Adipoq) and changes in the immune cell pool. These mice also developed fatty liver. Wild-type eWAT transplanted into B1 -/- mice normalized circulating insulin, leptin, and epidermal growth factor levels. In conclusion, we demonstrated that B1R in adipose tissue controls the response to diet-induced obesity by promoting adipose tissue expansion and hepatic lipid accumulation in cell-nonautonomous manners.


Subject(s)
Adipose Tissue, White/metabolism , Receptor, Bradykinin B1/metabolism , Adipose Tissue, White/transplantation , Animals , Body Composition/genetics , Body Composition/physiology , Diet, High-Fat/adverse effects , Flow Cytometry , Glucose/metabolism , Insulin Resistance/genetics , Insulin Resistance/physiology , Liver/metabolism , Male , Mice , Receptor, Bradykinin B1/genetics , Weight Gain/genetics , Weight Gain/physiology
15.
Tissue Eng Part C Methods ; 24(3): 135-145, 2018 03.
Article in English | MEDLINE | ID: mdl-29141507

ABSTRACT

White adipose tissue (WAT) is a critical organ in both health and disease. However, physiologically faithful tissue culture models of primary human WAT remain limited, at best. In this study we describe a novel WAT culture system in which primary human WAT is sandwiched between tissue-engineered sheets of adipose-derived stromal cells. This construct, called "sandwiched white adipose tissue" (SWAT), can be defined as a microphysiological system (MPS) since it is a tissue-engineered, multicellular, three-dimensional organ construct produced using human cells. We validated SWAT against the National Institutes of Health MPS standards and found that SWAT is viable in culture for 8 weeks, retains physiologic responses to exogenous signaling, secretes adipokines, and engrafts into animal models. These attributes position SWAT as a powerful tool for the study of WAT physiology, pathophysiology, personalized medicine, and pharmaceutical development.


Subject(s)
Adipocytes/cytology , Adipose Tissue, White/cytology , Stromal Cells/cytology , Tissue Culture Techniques/methods , Tissue Engineering/methods , Adipocytes/metabolism , Adipokines/metabolism , Adipose Tissue, White/metabolism , Adipose Tissue, White/transplantation , Adult , Animals , Cell Differentiation , Female , Gene Expression Profiling , Humans , Lipolysis , Male , Mice , Middle Aged , Stromal Cells/metabolism
16.
Cell Transplant ; 16(4): 375-90, 2007.
Article in English | MEDLINE | ID: mdl-17658128

ABSTRACT

White adipose tissue (WAT) transplantation, although widely used in humans, has been done for cosmetic and reconstructive purposes only. Accumulating evidence indicates, however, that WAT is an important endocrine organ and, therefore, WAT transplantation may become valuable as a replacement therapy for a number of hereditary human diseases. Because the most readily available source for such transplantations would be allogeneic tissue, the mechanisms involved in the rejection of WAT allograft should be explored. We have established a model in which leptin-producing allogeneic WAT is transplanted into leptin-deficient ob/ob mice. Because ob/ob mice are obese, hyperphagic, and hypothermic, WAT allograft function is monitored as the reversal of this leptin-deficient phenotype. Here we report that allografted WAT is primarily nonfunctional. However, when WAT is transplanted into immunodeficient (Rag1-/-) ob/ob mice, or into ob/ob mice depleted of T cells by anti-CD3 antibody, a long-term graft survival is achieved as indicated by the reversal of hyperphagia, weight loss, and normalization of body temperature. The symptoms of leptin deficiency rapidly recur when normal spleen cells of the recipient type are injected, or when the antibody treatment is terminated. In contrast, selective depletion of either CD4+ or CD8+ cells alone does not prevent WAT allograft rejection. Similarly, WAT allografts that do not express MHC class I or class II molecules are rapidly rejected, suggesting that both CD4+ and CD8+ T cells may independently mediate WAT allograft rejection.


Subject(s)
Adipose Tissue, White/transplantation , Graft Rejection/immunology , T-Lymphocytes/immunology , Adipose Tissue, White/immunology , Animals , CD3 Complex/immunology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Leptin/metabolism , Major Histocompatibility Complex/immunology , Mice , Mice, Inbred C57BL , Mice, Obese , Transplantation, Homologous
17.
J Plast Reconstr Aesthet Surg ; 70(8): 1118-1128, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28526633

ABSTRACT

Cumulative long-term exposure to solar ultraviolet radiation promotes premature skin aging characterized by wrinkle formation and reduced skin elasticity. In this study, we assessed whether microfat transfer could improve dermal and subcutaneous tissue thickness loss associated with photoaging. Twenty-one patients affected by facial photoaging (photodamage grade II-IV; age range 35-62 years; 19 females, 2 males; all of Caucasian origin) were treated using minimally invasive autologous dermal white fat transfer harvested with a recently designed microcannula. The results were determined by clinical assessment and patient self-evaluation and quantified by the Antera 3D® dermal digital device for noninvasive, objective, reliable, and accurate assessment of facial skin texture, color, and wrinkle characteristics. Compared with the pretreatment condition, the increment in soft tissue volume and improvement in skin quality and texture were assessed by a dermatologist after treatment. In addition, instrumental evaluation by digital skin profilometry of the treated areas revealed a 41% reduction in average wrinkle depth (7.29 ± 1.04 × 10-2 mm pretreatment vs. 4.31 ± 1.16 × 10-2 mm at 90 days posttreatment; p < 0.001), improved skin texture, more homogeneous and uniform skin color, and declined facial hemoglobin and melanin concentrations. The majority of patients (above 90%) reported improvements in self-perception. No significant complications were reported throughout the study. In conclusion, by using digital profilometry analysis as an objective and innovative tool to determine the outcome of treatment, we demonstrated that autologous microfat transfer is a safe and well-tolerated procedure with measurable beneficial effects on facial skin aging.


Subject(s)
Adipose Tissue, White/transplantation , Rejuvenation , Rhytidoplasty/methods , Skin Aging , Skin Physiological Phenomena , Adult , Face , Female , Humans , Male , Middle Aged , Patient Satisfaction , Self Concept , Skin Aging/pathology , Surveys and Questionnaires , Tissue and Organ Harvesting/instrumentation , Transplantation, Autologous
18.
Diabetes ; 64(7): 2361-8, 2015 Jul.
Article in English | MEDLINE | ID: mdl-26050668

ABSTRACT

Regular physical activity and exercise training have long been known to cause adaptations to white adipose tissue (WAT), including decreases in cell size and lipid content and increases in mitochondrial proteins. In this article, we discuss recent studies that have investigated the effects of exercise training on mitochondrial function, the "beiging" of WAT, regulation of adipokines, metabolic effects of trained adipose tissue on systemic metabolism, and depot-specific responses to exercise training. The major WAT depots in the body are found in the visceral cavity (vWAT) and subcutaneously (scWAT). In rodent models, exercise training increases mitochondrial biogenesis and activity in both these adipose tissue depots. Exercise training also increases expression of the brown adipocyte marker uncoupling protein 1 (UCP1) in both adipose tissue depots, although these effects are much more pronounced in scWAT. Consistent with the increase in UCP1, exercise training increases the presence of brown-like adipocytes in scWAT, also known as browning or beiging. Training results in changes in the gene expression of thousands of scWAT genes and an altered adipokine profile in both scWAT and vWAT. Transplantation of trained scWAT in sedentary recipient mice results in striking improvements in skeletal muscle glucose uptake and whole-body metabolic homeostasis. Human and rodent exercise studies have indicated that exercise training can alter circulating adipokine concentration as well as adipokine expression in adipose tissue. Thus, the profound changes to WAT in response to exercise training may be part of the mechanism by which exercise improves whole-body metabolic health.


Subject(s)
Adaptation, Physiological , Adipose Tissue, White/physiology , Exercise , Physical Conditioning, Animal , Adipocytes/physiology , Adipokines/metabolism , Adipose Tissue, White/transplantation , Animals , Humans , Ion Channels/metabolism , Ion Channels/physiology , Mice , Mitochondria/physiology , Mitochondrial Proteins/metabolism , Mitochondrial Proteins/physiology , Muscle, Skeletal/metabolism , Nitric Oxide Synthase Type III/physiology , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Transcription Factors/physiology , Uncoupling Protein 1
19.
Sci Rep ; 5: 17565, 2015 Nov 30.
Article in English | MEDLINE | ID: mdl-26615883

ABSTRACT

High-fat diet (HFD) triggers insulin resistance and diabetes mellitus, but their link remains unclear. Characterization of overt hyperglycemia in insulin receptor mutant (Insr(P1195L/+)) mice exposed to HFD (Insr(P1195L/+)/HFD mice) revealed increased glucose-6-phosphatase (G6pc) expression in liver and increased gluconeogenesis from glycerol. Lipolysis in white adipose tissues (WAT) and lipolysis-induced blood glucose rise were increased in Insr(P1195L/+)/HFD mice, while wild-type WAT transplantation ameliorated the hyperglycemia and the increased G6pc expression. We found that the expressions of genes involved in bile acid (BA) metabolism were altered in Insr(P1195L/+)/HFD liver. Among these, the expression of Cyp7a1, a BA synthesis enzyme, was insulin-dependent and was markedly decreased in Insr(P1195L/+)/HFD liver. Reduced Cyp7a1 expression in Insr(P1195L/+)/HFD liver was rescued by WAT transplantation, and the expression of Cyp7a1 was suppressed by glycerol administration in wild-type liver. These findings suggest that unsuppressed lipolysis in adipocytes elicited by HFD feeding is linked with enhanced gluconeogenesis from glycerol and with alterations in BA physiology in Insr(P1195L/+)/HFD liver.


Subject(s)
Adipocytes/metabolism , Bile Acids and Salts/metabolism , Diet, High-Fat , Gluconeogenesis , Lipolysis , Receptor, Insulin/genetics , Adipose Tissue, Brown/metabolism , Adipose Tissue, White/metabolism , Adipose Tissue, White/transplantation , Animals , Blood Glucose , Body Weight , Disease Models, Animal , Energy Metabolism , Fats/metabolism , Genotype , Glycerol/metabolism , Hyperglycemia/genetics , Hyperglycemia/metabolism , Insulin/metabolism , Insulin Resistance , Liver/metabolism , Mice , Mice, Transgenic , Models, Biological , Mutation , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Pyruvic Acid/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL