Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 110
Filter
1.
Cell Host Microbe ; 32(7): 1045-1047, 2024 Jul 10.
Article in English | MEDLINE | ID: mdl-38991501

ABSTRACT

The microbiota can impact antitumor immunity, but whether the microbiota regulates omental antitumor immunity remains elusive. In this issue of Cell Host & Microbe, Meza-Perez et al. demonstrated that Proteobacteria consume arginine to increase Treg cell suppressive capacity and inhibit antitumor immune responses, promoting tumor growth in the omentum.


Subject(s)
Arginine , Omentum , Proteobacteria , Arginine/metabolism , Animals , Omentum/immunology , Omentum/microbiology , Humans , Mice , Gastrointestinal Microbiome/immunology , T-Lymphocytes, Regulatory/immunology , Neoplasms/immunology , Neoplasms/microbiology
2.
Cell Host Microbe ; 32(7): 1177-1191.e7, 2024 Jul 10.
Article in English | MEDLINE | ID: mdl-38942027

ABSTRACT

Gut microbiota influence anti-tumor immunity, often by producing immune-modulating metabolites. However, microbes consume a variety of metabolites that may also impact host immune responses. We show that tumors grow unchecked in the omenta of microbe-replete mice due to immunosuppressive Tregs. By contrast, omental tumors in germ-free, neomycin-treated mice or mice colonized with altered Schaedler's flora (ASF) are spontaneously eliminated by CD8+ T cells. These mice lack Proteobacteria capable of arginine catabolism, causing increases in serum arginine that activate the mammalian target of the rapamycin (mTOR) pathway in Tregs to reduce their suppressive capacity. Transfer of the Proteobacteria, Escherichia coli (E. coli), but not a mutant unable to catabolize arginine, to ASF mice reduces arginine levels, restores Treg suppression, and prevents tumor clearance. Supplementary arginine similarly decreases Treg suppressive capacity, increases CD8+ T cell effectiveness, and reduces tumor burden. Thus, microbial consumption of arginine alters anti-tumor immunity, offering potential therapeutic strategies for tumors in visceral adipose tissue.


Subject(s)
Arginine , CD8-Positive T-Lymphocytes , Gastrointestinal Microbiome , Mice, Inbred C57BL , Omentum , T-Lymphocytes, Regulatory , Animals , Arginine/metabolism , Mice , T-Lymphocytes, Regulatory/immunology , Gastrointestinal Microbiome/immunology , CD8-Positive T-Lymphocytes/immunology , Omentum/immunology , TOR Serine-Threonine Kinases/metabolism , Proteobacteria , Escherichia coli/immunology , Neoplasms/immunology , Female
3.
Clin Immunol ; 229: 108797, 2021 08.
Article in English | MEDLINE | ID: mdl-34273585

ABSTRACT

The global obesity epidemic is contributing to increased prevalence of diseases fuelled by chronic inflammation, including cancer. Oesophageal adenocarcinoma (OAC) is an obesity-associated malignancy with increasing prevalence, dismal prognosis, and severely dysregulated immune processes. We previously reported that αß T cells migrate to omentum and liver in OAC and contribute to inflammation in these tissues. Here, we assessed the tissue distribution and phenotype of gamma/delta (γδ) T cells in the blood, omentum, liver and tumour of OAC patients. Our data show that the Vδ1 and Vδ3 subsets of γδ T cells are most prevalent in omentum and liver of OAC patients. Furthermore, γδ T cells are predominantly pro-inflammatory in these tissues, and co-express IFN-γ and IL-17. Moreover, γδ T cells exhibit cytotoxic capabilities in OAC omentum and liver. This study provides the first indication that γδ T cells contribute to obesity-associated inflammation in OAC and might be exploited therapeutically.


Subject(s)
Adenocarcinoma/immunology , Esophageal Neoplasms/immunology , Receptors, Antigen, T-Cell, gamma-delta/metabolism , T-Lymphocyte Subsets/immunology , Adenocarcinoma/etiology , Adenocarcinoma/pathology , Adult , Aged , Aged, 80 and over , Cell Degranulation , Esophageal Neoplasms/etiology , Esophageal Neoplasms/pathology , Female , Humans , Immunophenotyping , Inflammation/complications , Interferon-gamma/metabolism , Interleukin-17/metabolism , Liver/immunology , Liver/pathology , Lysosomal-Associated Membrane Protein 1/metabolism , Male , Middle Aged , Obesity/complications , Omentum/immunology , Omentum/pathology , Receptors, CCR6/metabolism , T-Lymphocyte Subsets/pathology , T-Lymphocyte Subsets/physiology , Tissue Distribution
4.
Front Immunol ; 12: 631609, 2021.
Article in English | MEDLINE | ID: mdl-33815381

ABSTRACT

Although the abilities of the omentum to alleviate inflammation and prevent infection have been revealed over the past decades, the underlying mechanisms remain largely unelucidated. Here, we demonstrated that the mortality of mice exposed to cecal ligation and puncture (CLP) and omentectomy was remarkably increased compared to those treated with CLP alone. Moreover, the efficacy of the omentum was associated with an impairment in intraperitoneal bacterial clearance together with an increase in the expression of proinflammatory cytokines. Besides, in response to peritoneal infections, the size and quantity of the omental milky spots (MSs) were increased tremendously and they also support innate-like B1 cell responses and local IgM production in the peritoneal cavity. Furthermore, not only the migration but also the functional activities of neutrophils were diminished in the absence of the omentum. These data collectively show that the omentum contributes more to peritoneal immune responses during septic peritonitis than has heretofore been recognized. Thus, harnessing the function of MS-containing omentum to increase its protective effectiveness may exert important biological and therapeutic implications for the control of intra-abdominal infections.


Subject(s)
Omentum/immunology , Peritonitis/immunology , Sepsis/immunology , Animals , B-Lymphocyte Subsets/immunology , Cecum/microbiology , Cecum/surgery , Mice , Neutrophil Infiltration , Neutrophils/immunology , Omentum/surgery , Peritonitis/microbiology , Phagocytosis , Sepsis/microbiology
5.
Nat Commun ; 12(1): 2388, 2021 04 22.
Article in English | MEDLINE | ID: mdl-33888702

ABSTRACT

To unravel the pathogenesis of obesity and its complications, we investigate the interplay between circadian clocks and NF-κB pathway in human adipose tissue. The circadian clock function is impaired in omental fat from obese patients. ChIP-seq analyses reveal that the core clock activator, BMAL1 binds to several thousand target genes. NF-κB competes with BMAL1 for transcriptional control of some targets and overall, BMAL1 chromatin binding occurs in close proximity to NF-κB consensus motifs. Obesity relocalizes BMAL1 occupancy genome-wide in human omental fat, thereby altering the transcription of numerous target genes involved in metabolic inflammation and adipose tissue remodeling. Eventually, clock dysfunction appears at early stages of obesity in mice and is corrected, together with impaired metabolism, by NF-κB inhibition. Collectively, our results reveal a relationship between NF-κB and the molecular clock in adipose tissue, which may contribute to obesity-related complications.


Subject(s)
ARNTL Transcription Factors/metabolism , Circadian Clocks/immunology , Intra-Abdominal Fat/pathology , NF-kappa B/metabolism , Obesity/complications , Adipocytes/immunology , Adipocytes/metabolism , Adiponectin/genetics , Adult , Animals , Biopsy , Case-Control Studies , Cells, Cultured , Chromatin Immunoprecipitation Sequencing , Circadian Clocks/genetics , Diet, High-Fat/adverse effects , Disease Models, Animal , Female , Gene Expression Regulation , Humans , Inflammation/immunology , Inflammation/pathology , Intra-Abdominal Fat/immunology , Male , Mesenchymal Stem Cells , Mice, Transgenic , Middle Aged , Obesity/immunology , Obesity/metabolism , Obesity/pathology , Omentum/immunology , Omentum/pathology , Period Circadian Proteins/genetics , Primary Cell Culture , Transcription, Genetic
6.
Gynecol Oncol ; 161(1): 78-82, 2021 04.
Article in English | MEDLINE | ID: mdl-33436287

ABSTRACT

Ovarian cancer is uncommon in relation to other women's cancer, however, it is associated with a disproportionate number of deaths due to women's cancer. According to the National Institute of Health, only 1.2% of new cancer diagnoses in the United States are attributed to ovarian cancer, yet it is the fifth leading cause of cancer death in women and is responsible for 2.3% of all female cancer deaths. Ovarian cancer deaths are largely due to widely metastatic and chemoresistant disease that often presents at a late stage. The omentum is one of the most common sites for ovarian cancer metastasis. Recent research findings have highlighted the specific tumor microenvironment of the omentum and how it can be manipulated to prevent ovarian cancer proliferation, metastasis and chemoresistance. Debulking surgery has been the mainstay in the treatment for ovarian cancer. Total omentectomy is classically described as essential to this procedure. This article explores the known benefits of total omentectomy in the surgical treatment of epithelial ovarian cancer as well as the potential benefit contained within the omental tumor microenvironment when the omentum is macroscopically free of disease at the time of initial surgery.


Subject(s)
Omentum/surgery , Ovarian Neoplasms/surgery , Female , Humans , Immunotherapy , Omentum/immunology , Omentum/pathology , Ovarian Neoplasms/immunology , Ovarian Neoplasms/pathology , Ovarian Neoplasms/therapy , Tumor Microenvironment
7.
J Leukoc Biol ; 109(4): 717-729, 2021 04.
Article in English | MEDLINE | ID: mdl-32881077

ABSTRACT

The peritoneal cavity is a fluid filled space that holds most of the abdominal organs, including the omentum, a visceral adipose tissue that contains milky spots or clusters of leukocytes that are organized similar to those in conventional lymphoid tissues. A unique assortment of leukocytes patrol the peritoneal cavity and migrate in and out of the milky spots, where they encounter Ags or pathogens from the peritoneal fluid and respond accordingly. The principal role of leukocytes in the peritoneal cavity is to preserve tissue homeostasis and secure tissue repair. However, when peritoneal homeostasis is disturbed by inflammation, infection, obesity, or tumor metastasis, specialized fibroblastic stromal cells and mesothelial cells in the omentum regulate the recruitment of peritoneal leukocytes and steer their activation in unique ways. In this review, the types of cells that reside in the peritoneal cavity, the role of the omentum in their maintenance and activation, and how these processes function in response to pathogens and malignancy will be discussed.


Subject(s)
Immunity , Omentum/immunology , Peritoneal Cavity/physiology , Adaptive Immunity , Animals , Humans , Immunity, Innate
8.
Histol Histopathol ; 35(11): 1275-1284, 2020 Nov.
Article in English | MEDLINE | ID: mdl-32926399

ABSTRACT

BACKGROUND: Omental milky spots (OMSs) are the primary lymphoid structures of the greater omentum. However, the presence of lymph nodes (LNs) has occasionally been mentioned as well. Understanding which lymphoid structures are present is of significance, especially in gastric tumor metastasis; tumor deposits in omental LNs suggest local lymphatic spread, whereas tumor deposits in OMSs suggest peritoneal spread and hence extensive disease. Since LNs and OMSs share morphological characteristics and OMSs might be wrongly identified as LNs, reliable hallmarks facilitating easy discrimination are needed. MATERIALS AND METHOD: A series of microscopic morphological hallmarks unique to LNs were selected as potential candidates and were assessed for their discriminative capacity: 1) capsule, 2) trabeculae, 3) subcapsular sinus, 4) afferent lymphatic vessels, 5) distinct B- and T cell regions, and 6) a layered organization with, from the outside in a capsule, cortex, paracortex, and medulla. These hallmarks were visualized by multiple staining techniques. RESULTS: Hallmarks 1, 2 5 and 6 were shown to be the most efficient as these were consistent and discriminative. They were best visualized by Picrosirius red, smooth muscle actin and a B-cell / T-cell double staining. CONCLUSION: The presence of a capsule, trabeculae, distinct B- and T-cell regions and a layered organization represent consistent and reliable morphological features which allow to easily distinguish LNs from OMSs, especially when applied in combination.


Subject(s)
Lymph Nodes/anatomy & histology , Omentum/anatomy & histology , Aged, 80 and over , B-Lymphocytes/chemistry , B-Lymphocytes/immunology , Biomarkers/analysis , Cadaver , Female , Humans , Immunohistochemistry , Lymph Nodes/chemistry , Lymph Nodes/immunology , Lymphatic Vessels/anatomy & histology , Lymphatic Vessels/chemistry , Male , Omentum/chemistry , Omentum/immunology , T-Lymphocytes/chemistry , T-Lymphocytes/immunology
9.
Immunity ; 52(4): 700-715.e6, 2020 04 14.
Article in English | MEDLINE | ID: mdl-32294409

ABSTRACT

The omentum is a visceral adipose tissue rich in fat-associated lymphoid clusters (FALCs) that collects peritoneal contaminants and provides a first layer of immunological defense within the abdomen. Here, we investigated the mechanisms that mediate the capture of peritoneal contaminants during peritonitis. Single-cell RNA sequencing and spatial analysis of omental stromal cells revealed that the surface of FALCs were covered by CXCL1+ mesothelial cells, which we termed FALC cover cells. Blockade of CXCL1 inhibited the recruitment and aggregation of neutrophils at FALCs during zymosan-induced peritonitis. Inhibition of protein arginine deiminase 4, an enzyme important for the release of neutrophil extracellular traps, abolished neutrophil aggregation and the capture of peritoneal contaminants by omental FALCs. Analysis of omental samples from patients with acute appendicitis confirmed neutrophil recruitment and bacterial capture at FALCs. Thus, specialized omental mesothelial cells coordinate the recruitment and aggregation of neutrophils to capture peritoneal contaminants.


Subject(s)
Appendicitis/immunology , Lymphocytes/immunology , Neutrophils/immunology , Omentum/immunology , Peritonitis/immunology , Stromal Cells/immunology , Acute Disease , Animals , Appendicitis/genetics , Appendicitis/microbiology , Cell Communication/immunology , Chemokine CXCL1/genetics , Chemokine CXCL1/immunology , Epithelial Cells/immunology , Epithelial Cells/microbiology , Epithelium/immunology , Epithelium/microbiology , Escherichia coli/growth & development , Escherichia coli/pathogenicity , Extracellular Traps/immunology , Female , Gene Expression , Humans , Lymphocytes/microbiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Neutrophil Infiltration , Neutrophils/microbiology , Omentum/microbiology , Peritonitis/chemically induced , Peritonitis/genetics , Peritonitis/microbiology , Protein-Arginine Deiminase Type 4/genetics , Protein-Arginine Deiminase Type 4/immunology , Sequence Analysis, RNA , Single-Cell Analysis , Stromal Cells/microbiology , Tissue Culture Techniques , Zymosan/administration & dosage
10.
Shock ; 53(4): 384-390, 2020 04.
Article in English | MEDLINE | ID: mdl-31389904

ABSTRACT

Once thought of as an inert fatty tissue present only to provide insulation for the peritoneal cavity, the omentum is currently recognized as a vibrant immunologic organ with a complex structure uniquely suited for defense against pathogens and injury. The omentum is a source of resident inflammatory and stem cells available to participate in the local control of infection, wound healing, and tissue regeneration. It is intimately connected with the systemic vasculature and communicates with the central nervous system and the hypothalamic pituitary adrenal axis. Furthermore, the omentum has the ability to transit the peritoneal cavity and sequester areas of inflammation and injury. It contains functional, immunologic units commonly referred to as "milky spots" that contribute to the organ's immune response. These milky spots are complex nodules consisting of macrophages and interspersed lymphocytes, which are gateways for the infiltration of inflammatory cells into the peritoneal cavity in response to infection and injury. The omentum contains far greater complexity than is currently conceptualized in clinical practice and investigations directed at unlocking its beneficial potential may reveal new mechanisms underlying its vital functions and the secondary impact of omentectomy for the staging and treatment of a variety of diseases.


Subject(s)
Intraabdominal Infections/prevention & control , Omentum/immunology , Wound Healing/physiology , Humans
12.
Int J Mol Sci ; 20(13)2019 Jun 26.
Article in English | MEDLINE | ID: mdl-31247969

ABSTRACT

Human nutrient metabolism, developed millions of years ago, is anachronistic. Adaptive features that offered survival advantages are now great liabilities. The current dietary pattern, coupled with massively reduced physical activities, causes an epidemic of obesity and chronic metabolic diseases, such as type 2 diabetes mellitus. Chronic inflammation is a major contributing factor to the initiation and progression of most metabolic and cardiovascular diseases. Among all components of an innate immune system, due to their dual roles as phagocytic as well as antigen-presenting cells, macrophages play an important role in the regulation of inflammatory responses, affecting the body's microenvironment and homeostasis. Earlier studies have established the beneficial, anti-inflammatory effects of whole body vibration (WBV) as a partial exercise mimetic, including reversing the effects of glucose intolerance and hepatic steatosis. Here for the first time, we describe potential mechanisms by which WBV may improve metabolic status and ameliorate the adverse consequences through macrophage polarization and altering the fecal microbiome.


Subject(s)
Gastrointestinal Microbiome , Macrophage Activation/immunology , Macrophages/immunology , Omentum/immunology , Vibration , Animals , Biodiversity , Biomarkers , Diabetes Mellitus, Type 2 , Disease Models, Animal , Feces/microbiology , Immunophenotyping , Macrophages/metabolism , Metagenome , Metagenomics/methods , Mice
13.
J Obstet Gynaecol Res ; 45(2): 417-427, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30276973

ABSTRACT

AIM: This study aimed to confirm whether human omental adipose-derived stem cells (O-ASC) from donors with varying body mass index (BMI, calculated in kg/m2 ) exert different effects on proliferation and migration of endometrial cancer (EC) cells. METHODS: Omental adipose-derived stem cells were isolated from the omental adipose tissues of eight patients who were diagnosed with EC aged from 35 to 56 years. According to the patients' BMI, the O-ASC was divided into two groups: obesity group (BMI ≥ 30) and normal group (18.5 < BMI ≤ 24.9). A broad spectrum cytokine antibody array was used to measure 62 paracrine cytokines secreted by all the O-ASC. MTS assays, direct and indirect co-culture assays were used to assess the effects of O-ASC on proliferation and migration of Hec-1A (estrogen receptor-/progesterone receptor-) and Ishikawa (estrogen receptor+/progesterone receptor+) EC cells. RESULTS: Eight samples of O-ASC were successfully isolated including four samples in obesity group and four in normal group. All the O-ASC displayed typical characteristics of mesenchymal stem cells and possessed similar secretory functions as 26 cytokines were identified in condition medium of O-ASC based on cytokine antibody array. The proliferation of Ishikawa cells was gently stimulated by O-ASC from the two groups (P < 0.05) whereas without any effects on Hec-1A cells. Both horizontal and vertical migrations of EC cells were promoted by O-ASC (P < 0.01). However, there were no statistical differences between the two groups. CONCLUSION: Human O-ASC could influence the proliferation and migration of EC cells in vitro but the effects were not modified by donors' BMI.


Subject(s)
Body Mass Index , Cytokines , Endometrial Neoplasms , Intra-Abdominal Fat , Mesenchymal Stem Cells , Omentum , Adult , Cell Line, Tumor , Cytokines/immunology , Cytokines/metabolism , Endometrial Neoplasms/immunology , Endometrial Neoplasms/metabolism , Female , Humans , Intra-Abdominal Fat/immunology , Intra-Abdominal Fat/metabolism , Mesenchymal Stem Cells/immunology , Mesenchymal Stem Cells/metabolism , Middle Aged , Obesity/immunology , Obesity/metabolism , Omentum/immunology , Omentum/metabolism
14.
Adv Mater ; 31(1): e1803895, 2019 Jan.
Article in English | MEDLINE | ID: mdl-30406960

ABSTRACT

Despite incremental improvements in the field of tissue engineering, no technology is currently available for producing completely autologous implants where both the cells and the scaffolding material are generated from the patient, and thus do not provoke an immune response that may lead to implant rejection. Here, a new approach is introduced to efficiently engineer any tissue type, which its differentiation cues are known, from one small tissue biopsy. Pieces of omental tissues are extracted from patients and, while the cells are reprogrammed to become induced pluripotent stem cells, the extracellular matrix is processed into an immunologically matching, thermoresponsive hydrogel. Efficient cell differentiation within a large 3D hydrogel is reported, and, as a proof of concept, the generation of functional cardiac, cortical, spinal cord, and adipogenic tissue implants is demonstrated. This versatile bioengineering approach may assist to regenerate any tissue and organ with a minimal risk for immune rejection.


Subject(s)
Hydrogels/chemistry , Prostheses and Implants , Animals , Cell Differentiation , Cellular Reprogramming , Endothelial Cells/cytology , Endothelial Cells/immunology , Endothelial Cells/transplantation , Extracellular Matrix/immunology , Extracellular Matrix/metabolism , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Mice , Mice, Inbred C57BL , Myocytes, Cardiac/cytology , Myocytes, Cardiac/immunology , Myocytes, Cardiac/transplantation , Omentum/cytology , Omentum/immunology , Omentum/metabolism , Swine , Tissue Engineering , Tissue Scaffolds , Transplantation, Autologous
15.
Zhonghua Wei Chang Wai Ke Za Zhi ; 21(12): 1347-1350, 2018 Dec 25.
Article in Chinese | MEDLINE | ID: mdl-30588583

ABSTRACT

Despite the evolution of aggressive surgical techniques, extensive methods of supportive care and a vast array of anti-microbial options, intra-abdominal infection (IAI) is still a challenging clinical issue. Especially, when progressed IAI with septic complications because of unbalanced immune responses, the prognosis will deteriorated significantly. Recent studies indicate that besides the natural immunological cells, including macrophages and neutrophils, local immunological characteristics of peritoneal cavity should be studied with great attention. Among them, the omentum is considered to be a visceral adipose tissue with unique immune function. The milky spots(MSs) formed by the accumulation of immune cells performs immune surveillance and has a lymph node-like immune function, which is very important for the immune defense of the abdominal cavity. B1 cells and two types of intrinsic lymphocytes(ILC2) in the peritoneal cavity, although belonging to the lymphatic lineage, may play an important role in abdominal infections, especially in the early stages of the disease, due to their rapid responsiveness and acquired immune function. Therefore, paying attention to the immunological characteristics of the peritoneal cavity, and elucidating the changes, functions and regulatory mechanisms of B1 cells and ILC2 around the MSs and their components in the process of IAI, in order to explore the immunomodulation targets of blocking the infection from local to systemic dissemination, may be the key to solving the clinical problem of severe IAI and improving prognosis.


Subject(s)
Intraabdominal Infections , Omentum , Peritoneal Cavity , Humans , Intraabdominal Infections/immunology , Lymphocytes/immunology , Macrophages/immunology , Omentum/immunology
16.
Front Immunol ; 9: 1867, 2018.
Article in English | MEDLINE | ID: mdl-30150990

ABSTRACT

The omentum is enriched with pro-inflammatory effector memory CD8+ T cells in patients with the obesity-associated malignancy, esophagogastric adenocarcinoma (EAC) and we have identified the chemokine macrophage inflammatory protein-1alpha as a key player in their active migration to this inflamed tissue. More recently, others have established that subsets of memory CD8+ T cells can be classified based on their surface expression of CX3CR1; the specific receptor for the inflammatory chemokine fractalkine. CD8+ T cells expressing intermediate levels (CX3CR1INT) are defined as peripheral memory, those expressing the highest levels (CX3CR1HI) are effector memory/terminally differentiated and those lacking CX3CR1 (CX3CR1NEG) are classified as central memory. To date, the fractalkine:CX3CR1 axis has not been examined in the context of CD8+ T cell enrichment in the omentum and here we examine this chemokines involvement in the accumulation of memory CD8+ T cells in the omentum of EAC patients. Our data show that fractalkine is significantly enriched in the omentum of EAC patients and drives migration of T cells derived from EAC patient blood. Furthermore, CX3CR1 is endocytosed specifically by CD8+ T cells upon encountering fractalkine, which is consistent with the significantly diminished frequencies of CX3CR1INT and CX3CR1HI CD8+ T cells in the fractalkine-rich environment of omentum in EAC, relative to matched blood. Fractalkine-mediated endocytosis of CX3CR1 by CD8+ T cells is sustained and is followed by enhanced surface expression of L-selectin (CD62L). These novel data align with our findings that circulating CX3CR1NEG CD8+ T cells express higher levels of L-selectin than CX3CR1INT CD8+ T cells. This is consistent with previous reports and implicates fractalkine in the conversion of CX3CR1INT CD8+ T cells to a CX3CR1NEG phenotype characterized by alterations in the migratory capacity of these T cells. For the first time, these findings identify fractalkine as a driver of T cell migration to the omentum in EAC and indicate that CD8+ T cells undergo sequenced fractalkine-mediated alterations in CX3CR1 and L-selectin expression. These data implicate fractalkine as more than a chemotactic cytokine in obesity-associated meta-inflammation and reveal a role for this chemokine in the maintenance of the CX3CR1NEG CD8+ T cell populations.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Chemokine CX3CL1/metabolism , Neoplasms/immunology , Obesity/immunology , Omentum/immunology , Adult , Aged , Aged, 80 and over , CX3C Chemokine Receptor 1/metabolism , Cell Movement , Cell Proliferation , Cells, Cultured , Female , Humans , Immunologic Memory , L-Selectin/metabolism , Male , Middle Aged , Neoplasms/complications , Obesity/complications
17.
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
18.
Inflammation ; 39(5): 1729-36, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27423204

ABSTRACT

In the midst of a worsening obesity epidemic, the incidence of obesity-associated morbidities, including cancer, diabetes, cardiac and liver disease is increasing. Insights into mechanisms underlying pathological obesity-associated inflammation are lacking. Both the omentum, the principal component of visceral fat, and liver of obese individuals are sites of excessive inflammation, but to date the T cell profiles of both compartments have not been assessed or compared in a patient cohort with obesity-associated disease. We have previously identified that omentum is enriched with inflammatory cytokines, chemokines and T cells. Here, we compared the inflammatory profile of T cells in the omentum and liver of patients with the obesity-associated malignancy oesophageal adenocarcinoma (OAC). Furthermore, we assessed the secreted cytokine profile in OAC patient serum, omentum and liver to assess systemic and local inflammation. We observed parallel T cell cytokine profiles and phenotypes in the omentum and liver of OAC patients, in particular CD69(+) and inflammatory effector memory T cells. This study reflects similar processes of inflammation and T cell activation in the omentum and liver, and may suggest common targets to modulate pathological inflammation at these sites.


Subject(s)
Intra-Abdominal Fat/pathology , Liver/pathology , Neoplasms/pathology , Obesity/pathology , T-Lymphocyte Subsets/immunology , Adenocarcinoma , Cytokines/analysis , Esophageal Neoplasms , Humans , Immunologic Memory , Inflammation/immunology , Inflammation/pathology , Intra-Abdominal Fat/immunology , Liver/immunology , Obesity/complications , Omentum/immunology , Omentum/pathology
19.
J Leukoc Biol ; 100(6): 1435-1442, 2016 12.
Article in English | MEDLINE | ID: mdl-27365528

ABSTRACT

The role of NK cells in visceral adipose tissue (VAT) and liver inflammation in obesity is not fully understood. This study investigated the frequency, cytokine expression, chemokine receptor, and cytotoxicity receptor profile of NK cells in the blood, omentum, and liver of patients with the obesity-associated cancer, oesophageal adenocarcinoma (OAC). The effect of chronically inflamed tissue microenvironments on NK cell viability and function was also examined. We identified significantly lower NK cell frequencies in the liver of OAC patients compared with healthy controls and within the omentum and liver of OAC patients compared with blood, whereas IL-10-producing populations were significantly higher. Interestingly, our data suggest that reduced frequencies of NK cells in omentum and liver of OAC patients are not a result of impaired NK cell chemotaxis to these tissues. In fact, our functional data revealed that secreted factors from omentum and liver of OAC patients induce significant levels of NK cell death and lead to reduced percentages of TNF-α+ and NKP46+ NK cells and higher frequencies of IL-10-producing NK cells. Together, these data suggest that the omental and hepatic microenvironments of OAC patients alter the NK cell phenotype to a more anti-inflammatory homeostatic role.


Subject(s)
Adenocarcinoma/immunology , Cellular Microenvironment , Esophageal Neoplasms/immunology , Intra-Abdominal Fat/immunology , Killer Cells, Natural/immunology , Liver/immunology , Adenocarcinoma/blood , Adenocarcinoma/etiology , Aged , Cell Survival , Cells, Cultured , Chemotaxis , Culture Media, Conditioned/pharmacology , Cytotoxicity, Immunologic , Esophageal Neoplasms/blood , Esophageal Neoplasms/etiology , Female , Humans , Interleukin-10/biosynthesis , Male , Middle Aged , Natural Cytotoxicity Triggering Receptor 1/analysis , Obesity/complications , Omentum/immunology , Organ Specificity , Receptors, Chemokine/analysis , Receptors, Natural Killer Cell/analysis , Tumor Necrosis Factor-alpha/biosynthesis
20.
Tumour Biol ; 37(5): 5715-26, 2016 May.
Article in English | MEDLINE | ID: mdl-26831659

ABSTRACT

As the most common metastatic disease of abdomen pelvic cavity cancer, peritoneal carcinomatosis (PC) renders significant negative impact on patient survival and quality of life. Invasive peritoneal exfoliated cancer cells (PECCs) preferentially select the omentum as a predominant target site for cancer cell colonization and proliferation compared with other tissues in the abdominal cavity. The precise pathogenic mechanism remains to be determined. As omental milky spots (MSs) are the major implantation site for malignant cells in peritoneal dissemination, researches on mechanisms of PC have been mainly focused on MS, primitive lymphoid tissues with unique structural features, and functional characteristics. To date, extensive biophysical and biochemical methods have been manipulated to investigate the MS exact function in the peritoneal cavity. This review summarized MS as hotbeds for PECC. The anatomical distribution was briefly described first. Then, MS histology was systematically reviewed, including morphological features, cellular constituents, and histological staining methods. At last, the roles of MS in PC pathological process were summarized with special emphasis on the distinct roles of macrophages.


Subject(s)
Carcinoma/secondary , Lymphoid Tissue/pathology , Omentum/pathology , Peritoneal Neoplasms/secondary , Animals , Cell Movement , Humans , Lymphocytes , Macrophages, Peritoneal , Omentum/immunology , Organ Specificity , Pleura/pathology , Staining and Labeling/methods
SELECTION OF CITATIONS
SEARCH DETAIL
...