Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 259
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Cell ; 157(4): 832-44, 2014 May 08.
Article in English | MEDLINE | ID: mdl-24792964

ABSTRACT

Tissue-resident macrophages are highly heterogeneous in terms of their functions and phenotypes as a consequence of adaptation to different tissue environments. Local tissue-derived signals are thought to control functional polarization of resident macrophages; however, the identity of these signals remains largely unknown. It is also unknown whether functional heterogeneity is a result of irreversible lineage-specific differentiation or a consequence of continuous but reversible induction of diverse functional programs. Here, we identified retinoic acid as a signal that induces tissue-specific localization and functional polarization of peritoneal macrophages through the reversible induction of transcription factor GATA6. We further found that GATA6 in macrophages regulates gut IgA production through peritoneal B-1 cells. These results provide insight into the regulation of tissue-resident macrophage functional specialization by tissue-derived signals.


Subject(s)
GATA6 Transcription Factor/metabolism , Gene Expression Regulation , Macrophages, Peritoneal/cytology , Tretinoin/metabolism , Animals , Immunoglobulin A/genetics , Macrophages, Peritoneal/metabolism , Mice , Mice, Inbred C57BL , Omentum/cytology
2.
FASEB J ; 35(5): e21534, 2021 05.
Article in English | MEDLINE | ID: mdl-33817830

ABSTRACT

The adipocyte precursors (APs) located in white adipose tissue (WAT) are functionally significant in adipose plasticity and browning. Modifying adipogenesis or WAT browning targeted on APs is a promising mechanism for anti-obesity drug. We herein explored the in vitro actions and mechanisms of glucose-dependent insulinotropic polypeptide (GIP), a gut-derived peptide, in human adipose-derived mesenchymal stem cells (hADSCs) isolated from omentum. The hADSCs were cotreated with 100 nM GIP with or without equimolar concentration of GIP3-42 (a GIP receptor antagonist), and subsequently examined in vitro. CCK-8, EdU incorporation, and flow cytometry assays were used to assess cellular proliferation. Annexin V FTIC/PI double stain, TUNEL staining, and Western blot were applied for apoptosis evaluation. Adipogenesis was reflected by Western blot, real-time PCR, Oil Red O staining, mitochondrial staining, and mitochondrial DNA analysis. Results showed that GIP promoted proliferation and inhibited apoptosis of hADSCs via pleiotropic effects. Besides, GIP facilitated de novo beige adipogenesis, by accelerating mitotic clonal expansion (MCE), upregulating core adipogenic regulators (C/EBPα and PPARγ), augmenting beige-related genes (UCP1, PGC1α, and PRDM16), increasing mitochondrial content and improving beige adipocyte functionalities. Above all, our study expands knowledge on the mechanisms of GIP modifying adipogenesis especially in inducing beige adipogenesis, and thus provides a theoretical support for clinical usage of GIP on obesity treatment.


Subject(s)
Adipocytes, Beige/cytology , Adipocytes/cytology , Adipogenesis , Gastric Inhibitory Polypeptide/pharmacology , Gastrointestinal Agents/pharmacology , Mesenchymal Stem Cells/cytology , Omentum/cytology , Adipocytes/drug effects , Adipocytes/metabolism , Adipocytes, Beige/drug effects , Adipocytes, Beige/metabolism , Cell Differentiation , Humans , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Omentum/drug effects , Omentum/metabolism , Signal Transduction
3.
Br J Cancer ; 123(3): 459-470, 2020 08.
Article in English | MEDLINE | ID: mdl-32439934

ABSTRACT

BACKGROUND: Gastric cancer (GC) patients frequently develop peritoneal metastasis; however, the underlying mechanism remains unknown. We hypothesised that omental adipocytes (OmAd) trigger GC cells towards malignant activity to induce peritoneal metastasis. METHODS: We analysed interactions among human GC cells, endothelial cells and OmAd using a 3D co-culture system. We also employed a multipronged animal study, including subcutaneous and orthotopic tumours, and humanised omental adipose tissue models. Urinary levels of CXCL2 were analysed in human GC patients with and without peritoneal metastasis. RESULTS: Conditioned media derived from OmAd (OmAd-CM) promoted the proliferation, migration and capacity to induce angiogenesis of GC cells through AKT phosphorylation and VEGFA overexpression, whereas silencing CXCL2 in OmAd cancelled OmAd-induced effects. In an orthotopic tumour model using SCID mice, omentectomy suppressed GC growth and peritoneal dissemination, and reduced serum levels of CXCL2. OmAd promoted GC growth in a humanised omental adipose tissue model using NSG mice, but silencing CXCL2 in OmAd cancelled OmAd-induced tumour growth. Finally, urinary levels of CXCL2 were significantly higher in GC patients with peritoneal metastasis than in those without. CONCLUSION: Omental adipocytes trigger GC cells to an aggressive phenotype through CXCL2 secretion, which induces angiogenesis followed by cell growth and peritoneal metastasis.


Subject(s)
Chemokine CXCL2/urine , Coculture Techniques/methods , Omentum/cytology , Peritoneal Neoplasms/secondary , Stomach Neoplasms/pathology , Vascular Endothelial Growth Factor A/metabolism , Adipocytes/cytology , Adipocytes/metabolism , Animals , Cell Line, Tumor , Chemokine CXCL2/genetics , Culture Media, Conditioned/chemistry , Female , Humans , Mice , Mice, SCID , Omentum/metabolism , Peritoneal Neoplasms/metabolism , Peritoneal Neoplasms/pathology , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Stomach Neoplasms/genetics , Stomach Neoplasms/metabolism , Up-Regulation
4.
Trends Immunol ; 38(7): 526-536, 2017 07.
Article in English | MEDLINE | ID: mdl-28579319

ABSTRACT

The omentum is a visceral adipose tissue with unique immune functions. Although it is primarily an adipose tissue, the omentum also contains lymphoid aggregates, called milky spots (MSs), that contribute to peritoneal immunity by collecting antigens, particulates, and pathogens from the peritoneal cavity and, depending on the stimuli, promoting a variety of immune responses, including inflammation, tolerance, or even fibrosis. Reciprocal interactions between cells in the MS and adipocytes regulate their immune and metabolic functions. Importantly, the omentum collects metastasizing tumor cells and supports tumor growth by immunological and metabolic mechanisms. Here we summarize our current knowledge about the development, organization, and function of the omentum in peritoneal immunity.


Subject(s)
B-Lymphocytes/immunology , Cytokines/immunology , Lymphoid Tissue/immunology , Omentum/immunology , Receptors, Cytokine/immunology , T-Lymphocytes, Regulatory/immunology , Adipocytes/cytology , Adipocytes/immunology , Adipose Tissue/cytology , Adipose Tissue/immunology , Animals , B-Lymphocytes/cytology , Cell Communication/immunology , Cytokines/genetics , Epithelium/immunology , Gene Expression Regulation , Humans , Lymphoid Tissue/cytology , Macrophages/cytology , Macrophages/immunology , Omentum/cytology , Receptors, Cytokine/genetics , Signal Transduction , T-Lymphocytes, Regulatory/cytology
5.
Biochim Biophys Acta Mol Cell Res ; 1865(1): 25-33, 2018 Jan.
Article in English | MEDLINE | ID: mdl-29024694

ABSTRACT

Epithelial ovarian cancer (EOC) frequently metastasises to the omentum, a process that requires pro-angiogenic activation of human omental microvascular endothelial cells (HOMECs) by tumour-secreted factors. We have previously shown that ovarian cancer cells secrete a range of factors that induce pro-angiogenic responses e.g. migration, in HOMECs including the lysosomal protease cathepsin D (CathD). However, the cellular mechanism by which CathD induces these cellular responses is not understood. The aim of this study was to further examine the pro-angiogenic effects of CathD in HOMECs i.e. proliferation and migration, to investigate whether these effects are dependent on CathD catalytic activity and to delineate the intracellular signalling kinases activated by CathD. We report, for the first time, that CathD significantly increases HOMEC proliferation and migration via a non-proteolytic mechanism resulting in activation of ERK1/2 and AKT. These data suggest that EOC cancer secreted CathD acts as an extracellular ligand and may play an important pro-angiogenic, and thus pro-metastatic, role by activating the omental microvasculature during EOC metastasis to the omentum.


Subject(s)
Cathepsin D/physiology , Cell Movement/genetics , Cell Proliferation/genetics , Endothelial Cells/physiology , Omentum/cytology , Carcinoma, Ovarian Epithelial , Cathepsin D/genetics , Cells, Cultured , Endothelial Cells/cytology , Female , Humans , MAP Kinase Signaling System/physiology , Mitogen-Activated Protein Kinase 3/metabolism , Neoplasm Metastasis , Neoplasms, Glandular and Epithelial/metabolism , Neoplasms, Glandular and Epithelial/pathology , Omentum/blood supply , Omentum/pathology , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Peritoneal Neoplasms/genetics , Peritoneal Neoplasms/metabolism , Peritoneal Neoplasms/secondary , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/physiology
6.
Artif Organs ; 43(10): 1022-1027, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31013358

ABSTRACT

Permanent hypoparathyroidism is a severe clinical condition accompanied by low parathyroid hormone level. Conventional treatment requires lifelong medication, and daily drug usage has some side effects. To avoid this circumstance, transplantation is an alternative and curative option. Microencapsulation may be used as a transplantation approach particularly to evade immune response. In order to define treatment of permanent hypoparathyroidism, a 37-year-old female recipient who has permanent hypoparathyroidism was evaluated for 3 years. Routine tests, viral markers, and T and B lymphocyte cross-match tests were analyzed. In addition intradermal skin test was performed for ultrapure alginate. Microencapsulation of cultured parathyroid cells was performed with ultrapure alginate. Cell suspension was prepared and spheroids were generated with calcium chloride. Afterward, transplantation was performed with a laparoscopic approach in the omental tissue. The recipient was discharged from the hospital without complications. Serum calcium, parathyroid hormone (PTH), and phosphorus levels were observed throughout 1 year. During the follow-up period, no complications were observed. Serum calcium levels were increased significantly on day 10 and PTH levels were increased on day 25 as well. According to our knowledge, this is the first study where ultrapure alginate-based microencapsulated parathyroid cells were transplanted in the omental tissue. A significant increment of PTH levels was detected. Microencapsulated parathyroid cells showed the functionality of this technique for more than 1 year. This study showed that using ultrapure alginate-based microencapsulation without immunosuppression appears to be a promising technique.


Subject(s)
Cell Encapsulation/methods , Cells, Immobilized/transplantation , Hypoparathyroidism/therapy , Parathyroid Glands/cytology , Parathyroid Glands/transplantation , Adult , Alginates/chemistry , Cell Separation , Cells, Cultured , Cells, Immobilized/cytology , Female , Humans , Hypoparathyroidism/surgery , Male , Omentum/cytology , Omentum/surgery
7.
Int J Obes (Lond) ; 42(5): 1051-1061, 2018 06.
Article in English | MEDLINE | ID: mdl-29515208

ABSTRACT

BACKGROUND: Abdominal obesity is considered a major factor in the development of metabolic disorders. Glucagon-like peptide-1 (GLP-1) has been reported to have positive effects on improving body metabolism and to reducing insulin resistance. However, it remains less clear whether GLP-1 plays a role in the adipogenesis process of visceral fat. METHODS: Here, we analyzed the in vitro actions and probable mechanisms of Exendin-4, a GLP-1 receptor agonist, on human adipose-derived stromal cells (hADSCs) isolated from omentum. RESULTS: Our results demonstrated that Exendin-4 improved cell viability via promoting proliferation and inhibiting apoptosis in hADSCs isolated from omentum. Mechanistically, the activation of MAPK/ ERK1/2, Akt/GSK-3ß, and PKA/CREB pathways and downstream consequences induced are involved in the proliferative and anti-apoptotic roles of Exendin-4. More intriguingly, Exendin-4 could promote the differentiation of omental hADSCs. Underlying mechanisms of the differentiation of hADSCs are associated with the upregulation of the expression of pro-adipogenic genes and downregulation of the expression of anti-adipogenic genes. CONCLUSION: Our data demonstrate that Exendin-4 modifies adipogenesis of hADSCs isolated from omentum through multiple mechanisms, these effects could contribute to the protective actions of GLP-1 receptor agonist body metabolism and insulin sensitivity.


Subject(s)
Adipogenesis/drug effects , Adipose Tissue/cytology , Cell Physiological Phenomena/drug effects , Exenatide/pharmacology , Mesenchymal Stem Cells/drug effects , Omentum/cytology , Adolescent , Adult , Aged , Cells, Cultured , Female , Glucagon-Like Peptide 1/agonists , Humans , Male , Mesenchymal Stem Cells/cytology , Middle Aged , Young Adult
8.
Immunity ; 30(5): 731-43, 2009 May.
Article in English | MEDLINE | ID: mdl-19427241

ABSTRACT

The omentum is a site of B1 cell lymphopoiesis and immune responsiveness to T cell-independent antigens. However, it is unknown whether it supports immune responses independently of conventional lymphoid organs. We showed that the omentum collected antigens and cells from the peritoneal cavity and supported T cell-dependent B cell responses, including isotype switching, somatic hypermutation, and limited affinity maturation, despite the lack of identifiable follicular dendritic cells. The omentum also supported CD4+ and CD8+ T cell responses to peritoneal antigens and recruited effector T cells primed in other locations. Unlike conventional lymphoid organs, milky spots in the omentum developed in the absence of lymphoid tissue-inducer cells, but required the chemokine CXCL13. Although the lymphoid architecture of milky spots was disrupted in lymphotoxin-deficient mice, normal architecture was restored by reconstitution with lymphotoxin-sufficient hematopoietic cells. These results indicate that the milky spots of the omentum function as unique secondary lymphoid organs that promote immunity to peritoneal antigens.


Subject(s)
B-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/immunology , Chemokine CXCL13/immunology , Lymphatic System/immunology , Lymphoid Tissue/immunology , Omentum/immunology , Animals , Antigens/immunology , B-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/metabolism , Chemokine CXCL13/genetics , Chemokine CXCL13/metabolism , Lymphatic System/metabolism , Lymphoid Tissue/cytology , Lymphoid Tissue/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Omentum/cytology , Omentum/metabolism , Peritoneum/cytology , Peritoneum/immunology , Peritoneum/metabolism
9.
Nanotechnology ; 29(13): 13LT01, 2018 Apr 03.
Article in English | MEDLINE | ID: mdl-29384490

ABSTRACT

In microfluidics-based lab-on-a-chip systems, which are used for investigating the effect of drugs and growth factors on cells, the latter are usually cultured within the device's channels in two-dimensional, and not in their optimal three-dimensional (3D) microenvironment. Herein, we address this shortfall by designing a microfluidic system, comprised of two layers. The upper layer of the system consists of multiple channels generating a gradient of soluble factors. The lower layer is comprised of multiple wells, each deposited with 3D, nanofibrous scaffold. We first used a mathematical model to characterize the fluid flow within the system. We then show that induced pluripotent stem cells can be seeded within the 3D scaffolds and be exposed to a well-mixed gradient of soluble factors. We believe that utilizing such system may enable in the future to identify new differentiation factors, investigate drug toxicity, and eventually allow to perform analyses on patient-specific tissues, in order to fit the appropriate combination and concentration of drugs.


Subject(s)
Cell Culture Techniques/instrumentation , Induced Pluripotent Stem Cells/cytology , Lab-On-A-Chip Devices , Models, Statistical , Tissue Engineering/methods , Equipment Design , Humans , Hydrogels/chemistry , Induced Pluripotent Stem Cells/physiology , Nanofibers/ultrastructure , Omentum/cytology , Omentum/physiology , Primary Cell Culture , Rheology , Tissue Engineering/instrumentation , Tissue Scaffolds
10.
Gastric Cancer ; 21(1): 55-67, 2018 Jan.
Article in English | MEDLINE | ID: mdl-28540637

ABSTRACT

BACKGROUND: Scirrhous gastric cancer is an intractable disease with a high incidence of peritoneal dissemination and obstructive symptoms (e.g., ileus, jaundice, and hydronephrosis) arising from accompanying marked fibrosis. Microenvironmental interactions between cancer cells and cancer-associated fibroblasts are the suggested cause of the disease. We elucidated the mechanisms of tumor growth and fibrosis using human peritoneal mesothelial cells (HPMCs) and investigated the effects of tranilast treatment on cells and a xenograft mouse model of fibrosis. METHODS: HPMCs were isolated from surgically excised omentum and their interaction with MKN-45 gastric cancer cells was investigated using co-culture. Furthermore, a fibrosis tumor model was developed based on subcutaneous transplantation of co-cultured cells into the dorsal side of nude mice to form large fibrotic tumors. Mice were subsequently treated with or without tranilast. RESULTS: The morphology of HPMCs treated with transforming growth factor (TGF)-ß1 changed from cobblestone to spindle-type. Moreover, E-cadherin was weakly expressed whereas high levels of α-smooth muscle actin expression were observed. TGF-ß-mediated epithelial-mesenchymal transition-like changes in HPMCs were inhibited in a dose-dependent manner following tranilast treatment through inhibition of Smad2 phosphorylation. In the mouse model, tumor size decreased significantly and fibrosis was inhibited in the tranilast treatment group compared with that in the control group. CONCLUSIONS: Tranilast acts on the TGF-ß/Smad pathway to inhibit interactions between cancer cells and cancer-associated fibroblasts, thereby inhibiting tumor growth and fibrosis. This study supports the hypothesis that tranilast represents a novel strategy to prevent fibrous tumor establishment represented by peritoneal dissemination.


Subject(s)
Adenocarcinoma/pathology , Fibroblasts/drug effects , Stomach Neoplasms/pathology , ortho-Aminobenzoates/pharmacology , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Coculture Techniques , Disease Progression , Epithelial-Mesenchymal Transition/drug effects , Fibroblasts/pathology , Fibrosis , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Omentum/cytology , Xenograft Model Antitumor Assays
11.
J Proteome Res ; 16(8): 3083-3091, 2017 08 04.
Article in English | MEDLINE | ID: mdl-28675934

ABSTRACT

The extracellular matrix (ECM) is a complex meshwork of insoluble fibrillar proteins and signaling factors interacting together to provide architectural and instructional cues to the surrounding cells. Alterations in ECM organization or composition and excessive ECM deposition have been observed in diseases such as fibrosis, cardiovascular diseases, and cancer. We provide here optimized protocols to solubilize ECM proteins from normal or tumor tissues, digest the proteins into peptides, analyze ECM peptides by mass spectrometry, and interpret the mass spectrometric data. In addition, we present here two novel R-script-based web tools allowing rapid annotation and relative quantification of ECM proteins, peptides, and intensity/abundance in mass spectrometric data output files. We illustrate this protocol with ECMs obtained from two pairs of tissues, which differ in ECM content and cellularity: triple-negative breast cancer and adjacent mammary tissue, and omental metastasis from high-grade serous ovarian cancer and normal omentum. The complete proteomics data set generated in this study has been deposited to the public repository ProteomeXchange with the data set identifier: PXD005554.


Subject(s)
Extracellular Matrix/chemistry , Ovarian Neoplasms/chemistry , Proteomics/methods , Triple Negative Breast Neoplasms/chemistry , Breast/cytology , Extracellular Matrix/pathology , Extracellular Matrix Proteins/analysis , Female , Humans , Mass Spectrometry , Molecular Sequence Annotation , Omentum/cytology , Ovarian Neoplasms/secondary , Ovarian Neoplasms/ultrastructure , Triple Negative Breast Neoplasms/pathology , Triple Negative Breast Neoplasms/ultrastructure
12.
Int J Obes (Lond) ; 41(12): 1782-1789, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28769120

ABSTRACT

BACKGROUND: Inflammatory factors derived from adipose tissue have been implicated in mediating insulin resistance in obesity. We sought to identify these using explanted human adipose tissue exposed to innate and adaptive immune stimuli. METHODS: Subcutaneous and omental adipose tissue from obese, insulin-resistant donors was cultured in the presence of macrophage and T-cell stimuli, and the conditioned medium tested for its ability to inhibit insulin-stimulated glucose uptake into human Simpson-Golabi-Behmel Syndrome (SGBS) adipocytes. The nature of the inhibitory factor in conditioned medium was characterized physicochemically, inferred by gene microarray analysis and confirmed by antibody neutralization. RESULTS: Conditioned medium from omental adipose tissue exposed to a combination of macrophage- and T-cell stimuli inhibited insulin action and adiponectin secretion in SGBS adipocytes. This effect was associated with a pronounced change in adipocyte morphology, characterized by a decreased number of lipid droplets of increased size. The bioactivity of conditioned medium was abolished by trypsin treatment and had a molecular weight of 46 kDa by gel filtration. SGBS adipocytes exposed to a bioactive medium expressed multiple gene transcripts regulated by interferon-gamma (IFN-γ). Recombinant human IFN-γ recapitulated the effects of the bioactive medium and neutralizing antibody against IFN-γ but not other candidate factors abrogated medium bioactivity. CONCLUSIONS: IFN-γ released from inflamed omental adipose tissue may contribute to the metabolic abnormalities seen in human obesity.


Subject(s)
Adiponectin/metabolism , Insulin Resistance/physiology , Insulin/metabolism , Interferon-gamma/metabolism , Omentum/cytology , Subcutaneous Fat, Abdominal/metabolism , Adaptive Immunity/physiology , Body Mass Index , Cells, Cultured , Humans , Immunity, Innate/physiology , Immunohistochemistry , Phenotype , Subcutaneous Fat, Abdominal/physiopathology
13.
BMC Vet Res ; 13(1): 166, 2017 Jun 08.
Article in English | MEDLINE | ID: mdl-28595579

ABSTRACT

BACKGROUND: Mesenchymal Stem Cells (MSCs) are a promising therapeutic tool in veterinary medicine. Currently the subcutaneous adipose tissue is the leading source of MSCs in dogs. MSCs derived from distinct fat depots have shown dissimilarities in their accessibility and therapeutic potential. The aims of our work were to determine the suitability of omental adipose tissue as a source of MSCs, according to sampling success, cell yield and paracrine properties of isolated cells, and compared to subcutaneous adipose tissue. RESULTS: While sampling success of omental adipose tissue was 100% (14 collections from14 donors) for subcutaneous adipose tissue it was 71% (10 collections from 14 donors). MSCs could be isolated from both sources. Cell yield was significantly higher for omental than for subcutaneous adipose tissue (38 ± 1 vs. 30 ± 1 CFU-F/g tissue, p < 0.0001). No differences were observed between sources regarding cell proliferation potential (73 ± 1 vs. 74 ± 1 CDPL) and cell senescence (at passage 10, both cultures presented enlarged cells with cytoplasmic vacuoles and cellular debris). Omental- and subcutaneous-derived MSCs expressed at the same level bFGF, PDGF, HGF, VEGF, ANG1 and IL-10. Irrespective of the source, isolated MSCs induced proliferation, migration and vascularization of target cells, and inhibited the activation of T lymphocytes. CONCLUSION: Compared to subcutaneous adipose tissue, omental adipose tissue is a more suitable source of MSCs in dogs. Since it can be procured from donors with any body condition, its collection procedure is always feasible, its cell yield is high and the MSCs isolated from it have desirable differentiation and paracrine potentials.


Subject(s)
Adipose Tissue/cytology , Cell Separation/veterinary , Dogs/anatomy & histology , Mesenchymal Stem Cells , Omentum/cytology , Animals , Cell Proliferation , Cell Separation/methods , Endothelium, Vascular/cytology , Female , Mesenchymal Stem Cells/immunology , Subcutaneous Fat, Abdominal/cytology
14.
Am J Transplant ; 16(11): 3246-3254, 2016 11.
Article in English | MEDLINE | ID: mdl-27321369

ABSTRACT

Transplantation of islets into the liver confers several site-specific challenges, including a delayed vascularization and prevailing hypoxia. The greater omentum has in several experimental studies been suggested as an alternative implantation site for clinical use, but there has been no direct functional comparison to the liver. In this experimental study in mice, we characterized the engraftment of mouse and human islets in the omentum and compared engraftment and functional outcome with those in the intraportal site. The vascularization and innervation of the islets transplanted into the omentum were restored within the first month by paralleled ingrowth of capillaries and nerves. The hypoxic conditions in the islets early posttransplantation were transient and restricted to the first days. Newly formed blood vessels were fully functional, and the blood perfusion and oxygenation of the islets became similar to that of endogenous islets. Furthermore, islet grafts in the omentum showed at 1 month posttransplantation functional superiority to intraportally transplanted grafts. We conclude that in contrast to the liver the omentum provides excellent engraftment conditions for transplanted islets. Future studies in humans will be of great interest to investigate the capability of this site to also harbor larger grafts without interfering with islet functionality.


Subject(s)
Graft Survival , Islets of Langerhans Transplantation , Islets of Langerhans/cytology , Liver/surgery , Neovascularization, Physiologic , Omentum/cytology , Oxygen/metabolism , Animals , Female , Humans , Islets of Langerhans/blood supply , Islets of Langerhans/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Nude , Middle Aged , Omentum/blood supply , Omentum/metabolism
15.
Int J Obes (Lond) ; 40(12): 1823-1831, 2016 12.
Article in English | MEDLINE | ID: mdl-27698346

ABSTRACT

BACKGROUND: Adipose tissue fibrosis is a relatively new notion and its relationship with visceral obesity and cardiometabolic alterations remains unclear, particularly in moderate obesity. OBJECTIVE: Our objective was to examine if total and pericellular collagen accumulation are relevant for the pathophysiology of visceral obesity and related cardiometabolic risk. SUBJECTS AND METHODS: Surgical omental (OM) and subcutaneous (SC) fat samples were obtained in 56 women (age: 47.2±5.8 years; body mass index (BMI): 27.1±4.4 kg/m2). Body composition and fat distribution were measured by dual-energy X-ray absorptiometry and computed tomography, respectively. Total and pericellular collagen were measured using picrosirius red staining. CD68+ cells (total macrophages) and CD163+ cells (M2-macrophages) were identified using immunohistochemistry. RESULTS: We found that only pericellular collagen percentage, especially in OM fat, was associated with higher BMI, body fat mass and adipose tissue areas as well as lower radiologic attenuation of visceral adipose tissue and altered cardiometabolic risk variables. Strong correlations between peri-adipocyte collagen percentage and total or M2-macrophage percentages were observed in both depots. Total collagen percentage in either compartment was not related to adiposity, fat distribution or cardiometabolic risk. CONCLUSIONS: As opposed to whole tissue-based assessments of adipose tissue fibrosis, collagen deposition around the adipocyte, especially in the OM fat compartment is related to total and regional adiposity as well as altered cardiometabolic risk profile.


Subject(s)
Adipocytes/metabolism , Collagen/metabolism , Obesity, Abdominal/physiopathology , Omentum/metabolism , Absorptiometry, Photon , Body Composition , Body Fat Distribution , Cross-Sectional Studies , Female , Humans , Insulin Resistance , Lipids/blood , Middle Aged , Obesity, Abdominal/epidemiology , Obesity, Abdominal/metabolism , Omentum/cytology , Quebec/epidemiology
16.
J Am Soc Nephrol ; 25(12): 2778-88, 2014 Dec.
Article in English | MEDLINE | ID: mdl-24854264

ABSTRACT

The ability of cells to respond and survive stressful conditions is determined, in part, by the attachment of O-linked N-acetylglucosamine (O-GlcNAc) to proteins (O-GlcNAcylation), a post-translational modification dependent on glucose and glutamine. This study investigates the role of dynamic O-GlcNAcylation of mesothelial cell proteins in cell survival during exposure to glucose-based peritoneal dialysis fluid (PDF). Immortalized human mesothelial cells and primary mesothelial cells, cultured from human omentum or clinical effluent of PD patients, were assessed for O-GlcNAcylation under normal conditions or after exposure to PDF. The dynamic status of O-GlcNAcylation and effects on cellular survival were investigated by chemical modulation with 6-diazo-5-oxo-L-norleucine (DON) to decrease or O-(2-acetamido-2-deoxy-D-glucopyranosylidene)amino N-phenyl carbamate (PUGNAc) to increase O-GlcNAc levels. Viability was decreased by reducing O-GlcNAc levels by DON, which also led to suppressed expression of the cytoprotective heat shock protein 72. In contrast, increasing O-GlcNAc levels by PUGNAc or alanyl-glutamine led to significantly improved cell survival paralleled by higher heat shock protein 72 levels during PDF treatment. Addition of alanyl-glutamine increased O-GlcNAcylation and partly counteracted its inhibition by DON, also leading to improved cell survival. Immunofluorescent analysis of clinical samples showed that the O-GlcNAc signal primarily originates from mesothelial cells. In conclusion, this study identified O-GlcNAcylation in mesothelial cells as a potentially important molecular mechanism after exposure to PDF. Modulating O-GlcNAc levels by clinically feasible interventions might evolve as a novel therapeutic target for the preservation of peritoneal membrane integrity in PD.


Subject(s)
Acetylglucosamine/chemistry , Dialysis Solutions/chemistry , Epithelium/pathology , Peritoneal Dialysis/methods , Proteins/chemistry , Cell Survival , Cells, Cultured , Dialysis Solutions/pharmacology , Dipeptides/chemistry , Glucose/chemistry , Glutamine/chemistry , Glycosylation , HSP72 Heat-Shock Proteins/chemistry , Humans , Microscopy, Fluorescence , Omentum/cytology , Peritoneum/pathology , Protein Processing, Post-Translational
17.
J Am Soc Nephrol ; 25(6): 1270-81, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24627352

ABSTRACT

Stem cells show promise in the treatment of AKI but do not survive long term after injection. However, organ repair has been achieved by extending and attaching the omentum, a fatty tissue lying above the stomach containing stem cells, to various organs. To examine whether fusing the omentum to a subtotally nephrectomized kidney could slow the progression of CKD, we used two groups of rats: an experimental group undergoing 5/6 nephrectomy only and a control group undergoing 5/6 nephrectomy and complete omentectomy. Polydextran gel particles were administered intraperitoneally before suture only in the experimental group to facilitate the fusion of the omentum to the injured kidney. After 12 weeks, experimental rats exhibited omentum fused to the remnant kidney and had lower plasma creatinine and urea nitrogen levels; less glomerulosclerosis, tubulointerstitial injury, and extracellular matrix; and reduced thickening of basement membranes compared with controls. A fusion zone formed between the injured kidney and the omentum contained abundant stem cells expressing stem cell antigen-1, Wilms' tumor 1 (WT-1), and CD34, suggesting active, healing tissue. Furthermore, kidney extracts from experimental rats showed increases in expression levels of growth factors involved in renal repair, the number of proliferating cells, especially at the injured edge, the number of WT-1-positive cells in the glomeruli, and WT-1 gene expression. These results suggest that contact between the omentum and injured kidney slows the progression of CKD in the remnant organ, and this effect appears to be mediated by the presence of omental stem cells and their secretory products.


Subject(s)
Adult Stem Cells/physiology , Glomerulosclerosis, Focal Segmental/physiopathology , Omentum/physiology , Renal Insufficiency, Chronic/physiopathology , Adipose Tissue/cytology , Adipose Tissue/physiology , Adipose Tissue/surgery , Adult Stem Cells/cytology , Animals , Cell Proliferation , Disease Models, Animal , Disease Progression , Glomerular Mesangium/metabolism , Glomerular Mesangium/pathology , Glomerular Mesangium/physiopathology , Glomerulosclerosis, Focal Segmental/metabolism , Glomerulosclerosis, Focal Segmental/pathology , Intercellular Signaling Peptides and Proteins/metabolism , Male , Nephrectomy , Omentum/cytology , Omentum/surgery , Paracrine Communication/physiology , Rats , Rats, Sprague-Dawley , Renal Insufficiency, Chronic/metabolism , Renal Insufficiency, Chronic/pathology
18.
Physiol Genomics ; 46(10): 362-75, 2014 May 15.
Article in English | MEDLINE | ID: mdl-24642759

ABSTRACT

To better understand the impact of childhood obesity on intra-abdominal adipose tissue phenotype, a complete transcriptomic analysis using deep RNA-sequencing (RNA-seq) was performed on omental adipose tissue (OMAT) obtained from lean and Western diet-induced obese juvenile Ossabaw swine. Obese animals had 88% greater body mass, 49% greater body fat content, and a 60% increase in OMAT adipocyte area (all P < 0.05) compared with lean pigs. RNA-seq revealed a 37% increase in the total transcript number in the OMAT of obese pigs. Ingenuity Pathway Analysis showed transcripts in obese OMAT were primarily enriched in the following categories: 1) development, 2) cellular function and maintenance, and 3) connective tissue development and function, while transcripts associated with RNA posttranslational modification, lipid metabolism, and small molecule biochemistry were reduced. DAVID and Gene Ontology analyses showed that many of the classically recognized gene pathways associated with adipose tissue dysfunction in obese adults including hypoxia, inflammation, angiogenesis were not altered in OMAT in our model. The current study indicates that obesity in juvenile Ossabaw swine is characterized by increases in overall OMAT transcript number and provides novel data describing early transcriptomic alterations that occur in response to excess caloric intake in visceral adipose tissue in a pig model of childhood obesity.


Subject(s)
Diet , Disease Models, Animal , Intra-Abdominal Fat/metabolism , Omentum/metabolism , Pediatric Obesity/metabolism , Swine , Animals , Base Sequence , Body Composition , Body Weight , Computational Biology , Connective Tissue/growth & development , Connective Tissue/metabolism , Cytokines/blood , DNA Primers/genetics , Female , Gene Expression Profiling , High-Throughput Nucleotide Sequencing , Humans , Immunohistochemistry , Molecular Sequence Data , Omentum/cytology , Real-Time Polymerase Chain Reaction , Sequence Analysis, RNA
19.
Biochim Biophys Acta ; 1831(5): 950-9, 2013 May.
Article in English | MEDLINE | ID: mdl-23454374

ABSTRACT

In all mammals, white adipose tissue (WAT) and brown adipose tissue (BAT) are found together in several fat depots, forming a multi-depot organ. Adrenergic stimulation induces an increase in BAT usually referred to as "browning". This phenomenon is important because of its potential use in curbing obesity and related disorders; thus, understanding its cellular mechanisms in humans may be useful for the development of new therapeutic strategies. Data in rodents have supported the direct transformation of white into brown adipocytes. Biopsies of pure white omental fat were collected from 12 patients affected by the catecholamine-secreting tumor pheochromocytoma (pheo-patients) and compared with biopsies from controls. Half of the omental fat samples from pheo-patients contained uncoupling protein 1 (UCP1)-immunoreactive-(ir) multilocular cells that were often arranged in a BAT-like pattern endowed with noradrenergic fibers and dense capillary network. Many UCP1-ir adipocytes showed the characteristic morphology of paucilocular cells, which we have been described as cytological marker of transdifferentiation. Electron microscopy showed increased mitochondrial density in multi- and paucilocular cells and disclosed the presence of perivascular brown adipocyte precursors. Brown fat genes, such as UCP1, PR domain containing 16 (PRDM16) and ß3-adrenoreceptor, were highly expressed in the omentum of pheo-patients and in those cases without visible morphologic re-arrangement. Of note, the brown determinant PRDM16 was detected by immunohistochemistry only in nuclei of multi- and paucilocular adipocytes. Quantitative electron microscopy and immunohistochemistry for Ki67 suggest an unlikely contribution of proliferative events to the phenomenon. The data support the idea that, in adult humans, white adipocytes of pure white fat that are subjected to adrenergic stimulation are able to undergo a process of direct transformation into brown adipocytes. This article is part of a Special Issue entitled Brown and White Fat: From Signaling to Disease.


Subject(s)
Adipocytes/cytology , Adipose Tissue, Brown/cytology , Adipose Tissue, White/cytology , Adrenal Gland Neoplasms/pathology , Cell Transdifferentiation , Omentum/cytology , Pheochromocytoma/pathology , Adipocytes/metabolism , Adipose Tissue, Brown/metabolism , Adipose Tissue, White/metabolism , Adrenal Gland Neoplasms/genetics , Adrenal Gland Neoplasms/metabolism , Adult , Aged , Case-Control Studies , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Female , Humans , Immunoenzyme Techniques , Ion Channels/genetics , Ion Channels/metabolism , Male , Middle Aged , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Omentum/metabolism , Pheochromocytoma/genetics , Pheochromocytoma/metabolism , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Receptors, Adrenergic, beta-3/genetics , Receptors, Adrenergic, beta-3/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Transcription Factors/genetics , Transcription Factors/metabolism , Uncoupling Protein 1
20.
Exp Eye Res ; 121: 143-6, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24582890

ABSTRACT

Omental cells (OCs) are shown to help wound healing. The purpose of this study is to investigate if OCs improve cornea repair after alkali injury by subconjunctival injection of activated OCs in rats. Forty eight hours after limbal corneal alkali injury, fresh isolated OCs were injected subconjunctivally into the recipient rat's eye. Prior to the injury and at 0, 4 and 8 days after injury, the eyes were examined using slit lamp biomicroscopy. Corneal opacification and corneal neovascularization were graded in a masked fashion. The inflammatory response to the injury was evaluated by counting neutrophil cell numbers in the cornea under microscope. There was no significant difference in corneal opacification between the control and OCs treatment groups; however, the corneal neovascularization was significantly less in the eyes treated with OCs as compared to the controls. Also OCs treatment markedly decreased neutrophil infiltration after corneal-limbal alkali injury. Our results suggest that OCs may have a beneficial role in corneal healing after limbal corneal alkali injury by suppressing inflammatory cell infiltrates and corneal neovascularization.


Subject(s)
Burns, Chemical/therapy , Eye Burns/chemically induced , Limbus Corneae/pathology , Omentum/transplantation , Wound Healing/physiology , Animals , Burns, Chemical/physiopathology , Cell Transplantation , Corneal Neovascularization/physiopathology , Corneal Neovascularization/therapy , Corneal Opacity/physiopathology , Corneal Opacity/therapy , Disease Models, Animal , Leukocyte Count , Male , Neutrophils/cytology , Omentum/cytology , Rats , Rats, Inbred F344 , Sodium Hydroxide
SELECTION OF CITATIONS
SEARCH DETAIL