Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 635
Filter
1.
Front Immunol ; 15: 1387292, 2024.
Article in English | MEDLINE | ID: mdl-38779674

ABSTRACT

Peritoneal dialysis is a widely used method for treating kidney failure. However, over time, the peritoneal structure and function can deteriorate, leading to the failure of this therapy. This deterioration is primarily caused by infectious and sterile inflammation. Sterile inflammation, which is inflammation without infection, is particularly concerning as it can be subtle and often goes unnoticed. The onset of sterile inflammation involves various pathological processes. Peritoneal cells detect signals that promote inflammation and release substances that attract immune cells from the bloodstream. These immune cells contribute to the initiation and escalation of the inflammatory response. The existing literature extensively covers the involvement of different cell types in the sterile inflammation, including mesothelial cells, fibroblasts, endothelial cells, and adipocytes, as well as immune cells such as macrophages, lymphocytes, and mast cells. These cells work together to promote the occurrence and progression of sterile inflammation, although the exact mechanisms are not fully understood. This review aims to provide a comprehensive overview of the signals from both stromal cells and components of immune system, as well as the reciprocal interactions between cellular components, during the initiation of sterile inflammation. By understanding the cellular and molecular mechanisms underlying sterile inflammation, we may potentially develop therapeutic interventions to counteract peritoneal membrane damage and restore normal function.


Subject(s)
Cell Communication , Peritoneal Dialysis , Peritoneum , Stromal Cells , Humans , Peritoneal Dialysis/adverse effects , Peritoneum/pathology , Peritoneum/immunology , Animals , Stromal Cells/immunology , Cell Communication/immunology , Inflammation/immunology , Peritonitis/immunology
2.
Mol Immunol ; 170: 46-56, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38615627

ABSTRACT

Peritoneal B cells can be divided into B1 cells (CD11b+CD19+) and B2 cells (CD11b-CD19+) based on CD11b expression. B1 cells play a crucial role in the innate immune response by producing natural antibodies and cytokines. B2 cells share similar traits with B1 cells, influenced by the peritoneal environment. However, the response of both B1 and B2 cells to the same stimuli in the peritoneum remains uncertain. We isolated peritoneal B1 and B2 cells from mice and assessed differences in Interleukin-10(IL-10) secretion, apoptosis, and surface molecule expression following exposure to LPS and Interleukin-21(IL-21). Our findings indicate that B1 cells are potent IL-10 producers, possessing surface molecules with an IgMhiCD43+CD21low profile, and exhibit a propensity for apoptosis in vitro. Conversely, B2 cells exhibit lower IL-10 production and surface markers characterized as IgMlowCD43-CD21hi, indicative of some resistance to apoptosis. LPS stimulates MAPK phosphorylation in B1 and B2 cells, causing IL-10 production. Furthermore, LPS inhibits peritoneal B2 cell apoptosis by enhancing Bcl-xL expression. Conversely, IL-21 has no impact on IL-10 production in these cells. Nevertheless, impeding STAT3 phosphorylation permits IL-21 to increase IL-10 production in peritoneal B cells. Moreover, IL-21 significantly raises apoptosis levels in these cells, a process independent of STAT3 phosphorylation and possibly linked to reduced Bcl-xL expression. This study elucidates the distinct functional and response profiles of B1 and B2 cells in the peritoneum to stimuli like LPS and IL-21, highlighting their differential roles in immunological responses and B cell diversity.


Subject(s)
Apoptosis , B-Lymphocyte Subsets , Interleukin-10 , Interleukins , Lipopolysaccharides , Peritoneum , Animals , Mice , Antigens, CD19/immunology , Antigens, CD19/metabolism , Apoptosis/drug effects , Apoptosis/immunology , B-Lymphocyte Subsets/drug effects , B-Lymphocyte Subsets/immunology , B-Lymphocytes/drug effects , B-Lymphocytes/immunology , bcl-X Protein/metabolism , bcl-X Protein/immunology , CD11b Antigen/metabolism , CD11b Antigen/immunology , Interleukin-10/immunology , Interleukin-10/metabolism , Interleukins/immunology , Interleukins/pharmacology , Lipopolysaccharides/pharmacology , Lipopolysaccharides/immunology , Mice, Inbred C57BL , Peritoneum/immunology , Peritoneum/cytology , Phosphorylation/drug effects , STAT3 Transcription Factor/metabolism , STAT3 Transcription Factor/immunology
3.
Nephrology (Carlton) ; 27(1): 104-108, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34611952

ABSTRACT

Peritoneal dialysis (PD) causes structural and functional changes in the peritoneal membrane, which are attributed to local inflammatory process. This study assessed the presence of galectin-3 (Gal-3), a known inflammatory modulator, in dialysate effluent and correlated its levels with markers of inflammatory process. Gal-3 levels in serum and dialysate effluent were measured in prevalent PD patients on morning visits (n = 27) or during peritoneal equilibration tests (PET, n = 16), it association with clinical and laboratory parameters, including dialysate/plasma creatinine (D/P creatinine) and interleukin-6 (IL-6) levels was analysed. Gal-3 levels in dialysate effluent correlated with D/P creatinine (0.663, p = 0.005) and dialysate effluent IL-6 levels (0.674, p = 0.002), but not with serum Gal-3 levels or dialysis vintage. Patients who were high transporters had higher Gal-3 levels in dialysate effluent, as compared to lower transporters. In multivariate regression analysis, dialysate IL-6 level was the strongest predictor of dialysate Gal-3 levels. This study found Gal-3 in dialysate effluent correlated with D/P creatinine and dialysate IL-6 levels. These findings may imply that Gal-3 has a role in the intraperitoneal inflammatory process. However, this needs to be investigated further.


Subject(s)
Dialysis Solutions , Galectin 3/analysis , Inflammation , Kidney Failure, Chronic/therapy , Peritoneal Dialysis , Peritoneum/immunology , Aged , Biomarkers/analysis , Correlation of Data , Creatinine/analysis , Dialysis Solutions/analysis , Dialysis Solutions/metabolism , Female , Humans , Inflammation/blood , Inflammation/diagnosis , Inflammation/immunology , Inflammation Mediators/analysis , Interleukin-6/analysis , Israel/epidemiology , Kidney Failure, Chronic/epidemiology , Male , Middle Aged , Peritoneal Dialysis/adverse effects , Peritoneal Dialysis/methods
4.
Nat Commun ; 12(1): 7294, 2021 12 15.
Article in English | MEDLINE | ID: mdl-34911964

ABSTRACT

Recruitment of bone marrow derived monocytes via bloodstream and their subsequent conversion to CX3CR1+ macrophages in response to intestinal injury is dependent on CCR2, Nr4a1, and the microbiome. This process is critical for proper tissue repair; however, GATA6+ peritoneal cavity macrophages might represent an alternative, more readily available source of mature and functional myeloid cells at the damaged intestinal locations. Here we show, using spinning-disk confocal microscopy, that large F4/80hiGATA6+ peritoneal cavity macrophages promptly accumulate at damaged intestinal sites upon intestinal thermal injury and upon dextran sodium sulfate induced colitis in mice via a direct route from the peritoneal cavity. In contrast to bloodstream derived monocytes/macrophages, cavity macrophages do not depend on CCR2, Nr4a1 or the microbiome for recruitment, but rather on the ATP-release and exposed hyaluronan at the site of injury. They participate in the removal of necrotic cells, revascularization and collagen deposition and thus resolution of tissue damage. In summary, peritoneal cavity macrophages represent a rapid alternative route of intestinal tissue repair to traditional monocyte-derived macrophages.


Subject(s)
GATA6 Transcription Factor/immunology , Inflammatory Bowel Diseases/immunology , Macrophages, Peritoneal/immunology , Peritoneum/immunology , Animals , GATA6 Transcription Factor/genetics , Humans , Inflammatory Bowel Diseases/genetics , Inflammatory Bowel Diseases/physiopathology , Intestines/immunology , Male , Mice , Mice, Inbred C57BL , Monocytes/immunology , Nuclear Receptor Subfamily 4, Group A, Member 1/genetics , Nuclear Receptor Subfamily 4, Group A, Member 1/immunology , Receptors, CCR2/genetics , Receptors, CCR2/immunology , Regeneration
5.
Front Immunol ; 12: 684967, 2021.
Article in English | MEDLINE | ID: mdl-34054877

ABSTRACT

Most multicellular organisms have a major body cavity containing vital organs. This cavity is lined by a mucosa-like serosal surface and filled with serous fluid which suspends many immune cells. Injuries affecting the major body cavity are potentially life-threatening. Here we summarize evidence that unique damage detection and repair mechanisms have evolved to ensure immediate and swift repair of injuries at serosal surfaces. Furthermore, thousands of patients undergo surgery within the abdominal and thoracic cavities each day. While these surgeries are potentially lifesaving, some patients will suffer complications due to inappropriate scar formation when wound healing at serosal surfaces defects. These scars called adhesions cause profound challenges for health care systems and patients. Therefore, reviewing the mechanisms of wound repair at serosal surfaces is of clinical importance. Serosal surfaces will be introduced with a short embryological and microanatomical perspective followed by a discussion of the mechanisms of damage recognition and initiation of sterile inflammation at serosal surfaces. Distinct immune cells populations are free floating within the coelomic (peritoneal) cavity and contribute towards damage recognition and initiation of wound repair. We will highlight the emerging role of resident cavity GATA6+ macrophages in repairing serosal injuries and compare serosal (mesothelial) injuries with injuries to the blood vessel walls. This allows to draw some parallels such as the critical role of the mesothelium in regulating fibrin deposition and how peritoneal macrophages can aggregate in a platelet-like fashion in response to sterile injury. Then, we discuss how serosal wound healing can go wrong, causing adhesions. The current pathogenetic understanding of and potential future therapeutic avenues against adhesions are discussed.


Subject(s)
Macrophages, Peritoneal/immunology , Peritoneum/immunology , Serous Membrane/immunology , Wounds and Injuries/immunology , Animals , Ascitic Fluid/immunology , Blood Platelets/immunology , Cell Aggregation/immunology , GATA6 Transcription Factor/analysis , Humans , Macrophages, Peritoneal/chemistry , Peritoneum/injuries , Tissue Adhesions/immunology
6.
Nat Commun ; 12(1): 2232, 2021 04 14.
Article in English | MEDLINE | ID: mdl-33854051

ABSTRACT

Post-operative adhesions are a leading cause of abdominal surgery-associated morbidity. Exposed fibrin clots on the damaged peritoneum, in which the mesothelial barrier is disrupted, readily adhere to surrounding tissues, resulting in adhesion formation. Here we show that resident F4/80HighCD206- peritoneal macrophages promptly accumulate on the lesion and form a 'macrophage barrier' to shield fibrin clots in place of the lost mesothelium in mice. Depletion of this macrophage subset or blockage of CD11b impairs the macrophage barrier and exacerbates adhesions. The macrophage barrier is usually insufficient to fully preclude the adhesion formation; however, it could be augmented by IL-4-based treatment or adoptive transfer of this macrophage subset, resulting in robust prevention of adhesions. By contrast, monocyte-derived recruited peritoneal macrophages are not involved in the macrophage barrier. These results highlight a previously unidentified cell barrier function of a specific macrophage subset, also proposing an innovative approach to prevent post-operative adhesions.


Subject(s)
Macrophages, Peritoneal/immunology , Peritoneum/immunology , Postoperative Complications/immunology , Tissue Adhesions/immunology , Animals , CD11b Antigen/genetics , CD11b Antigen/immunology , Epithelium/immunology , Epithelium/pathology , Humans , Interleukin-4 , Male , Mice , Mice, Inbred C57BL , Peritoneum/pathology , Postoperative Complications/genetics , Postoperative Complications/pathology , Tissue Adhesions/genetics , Tissue Adhesions/pathology
7.
Front Immunol ; 12: 607204, 2021.
Article in English | MEDLINE | ID: mdl-33854496

ABSTRACT

Peritoneal fibrosis is characterized by abnormal production of extracellular matrix proteins leading to progressive thickening of the submesothelial compact zone of the peritoneal membrane. This process may be caused by a number of insults including pathological conditions linked to clinical practice, such as peritoneal dialysis, abdominal surgery, hemoperitoneum, and infectious peritonitis. All these events may cause acute/chronic inflammation and injury to the peritoneal membrane, which undergoes progressive fibrosis, angiogenesis, and vasculopathy. Among the cellular processes implicated in these peritoneal alterations is the generation of myofibroblasts from mesothelial cells and other cellular sources that are central in the induction of fibrosis and in the subsequent functional deterioration of the peritoneal membrane. Myofibroblast generation and activity is actually integrated in a complex network of extracellular signals generated by the various cellular types, including leukocytes, stably residing or recirculating along the peritoneal membrane. Here, the main extracellular factors and the cellular players are described with emphasis on the cross-talk between immune system and cells of the peritoneal stroma. The understanding of cellular and molecular mechanisms underlying fibrosis of the peritoneal membrane has both a basic and a translational relevance, since it may be useful for setup of therapies aimed at counteracting the deterioration as well as restoring the homeostasis of the peritoneal membrane.


Subject(s)
Cell Communication , Disease Susceptibility , Peritoneal Fibrosis/etiology , Peritoneal Fibrosis/metabolism , Peritoneum/immunology , Peritoneum/metabolism , Stromal Cells/metabolism , Animals , Biomarkers , Cell Communication/immunology , Cytokines/metabolism , Epithelial Cells/metabolism , Humans , Immunity, Innate , Inflammation Mediators/metabolism , Leukocytes/immunology , Leukocytes/metabolism , Peritoneal Dialysis/adverse effects , Peritoneal Fibrosis/pathology , Peritoneum/pathology , Peritonitis/complications , Peritonitis/etiology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism
8.
Am J Respir Cell Mol Biol ; 64(5): 641-643, 2021 05.
Article in English | MEDLINE | ID: mdl-33929292
9.
Int J Mol Sci ; 22(7)2021 Mar 27.
Article in English | MEDLINE | ID: mdl-33801658

ABSTRACT

BACKGROUND: Gram-negative infections of the peritoneal cavity result in profound modifications of peritoneal B cell populations and induce the migration of peritoneal B cells to distant secondary lymphoid organs. However, mechanisms controlling the egress of peritoneal B cells from the peritoneal cavity and their subsequent trafficking remain incompletely understood. Sphingosine-1-phosphate (S1P)-mediated signaling controls migratory processes in numerous immune cells. The present work investigates the role of S1P-mediated signaling in peritoneal B cell trafficking under inflammatory conditions. METHODS: Differential S1P receptor expression after peritoneal B cell activation was assessed semi­quantitatively using RT-PCR in vitro. The functional implications of differential S1P1 and S1P4 expression were assessed by transwell migration in vitro, by adoptive peritoneal B cell transfer in a model of sterile lipopolysaccharide (LPS)­induced peritonitis and in the polymicrobial colon ascendens stent peritonitis (CASP) model. RESULTS: The two sphingosine-1-phosphate receptors (S1PRs) expressed in peritoneal B cell subsets S1P1 and S1P4 are differentially regulated upon stimulation with the TLR4 agonist LPS, but not upon PMA/ionomycin or B cell receptor (BCR) crosslinking. S1P4 deficiency affects both the trafficking of activated peritoneal B cells to secondary lymphoid organs and the positioning of these cells within the functional compartments of the targeted organ. S1P4 deficiency in LPS-activated peritoneal B cells results in significantly reduced numbers of splenic innate response activator B cells. CONCLUSIONS: The S1P-S1PR system is implicated in the trafficking of LPS-activated peritoneal B cells. Given the protective role of peritoneal B1a B cells in peritoneal sepsis, further experiments to investigate the impact of S1P4-mediated signaling on the severity and mortality of peritoneal sepsis are warranted.


Subject(s)
Gene Expression Regulation , Inflammation , Sphingosine-1-Phosphate Receptors/metabolism , Spleen/metabolism , Toll-Like Receptor 4/metabolism , Animals , B-Lymphocytes/metabolism , Cell Movement , Female , Immunity, Innate , Lipopolysaccharides/chemistry , Lipopolysaccharides/metabolism , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Peritoneum/immunology , Peritoneum/metabolism , Peritoneum/pathology , Sepsis/physiopathology , Signal Transduction
10.
Science ; 371(6533)2021 03 05.
Article in English | MEDLINE | ID: mdl-33674464

ABSTRACT

Most multicellular organisms have a major body cavity that harbors immune cells. In primordial species such as purple sea urchins, these cells perform phagocytic functions but are also crucial in repairing injuries. In mammals, the peritoneal cavity contains large numbers of resident GATA6+ macrophages, which may function similarly. However, it is unclear how cavity macrophages suspended in the fluid phase (peritoneal fluid) identify and migrate toward injuries. In this study, we used intravital microscopy to show that cavity macrophages in fluid rapidly form thrombus-like structures in response to injury by means of primordial scavenger receptor cysteine-rich domains. Aggregates of cavity macrophages physically sealed injuries and promoted rapid repair of focal lesions. In iatrogenic surgical situations, these cavity macrophages formed extensive aggregates that promoted the growth of intra-abdominal scar tissue known as peritoneal adhesions.


Subject(s)
Macrophages, Peritoneal/immunology , Peritoneum/immunology , Peritoneum/injuries , Wounds and Injuries/immunology , Animals , Ascitic Fluid/immunology , Blood Platelets/immunology , Cell Aggregation/immunology , GATA6 Transcription Factor/analysis , Macrophages, Peritoneal/chemistry , Mice , Mice, Inbred C57BL , Scavenger Receptors, Class B/immunology , Thrombosis/immunology , Tissue Adhesions/immunology
11.
Parasit Vectors ; 14(1): 54, 2021 Jan 18.
Article in English | MEDLINE | ID: mdl-33461599

ABSTRACT

BACKGROUND: Here, Mesocestoides (M.) vogae infection in mice is proposed as a suitable experimental model for studying the immunity in the peritoneal cavity of mice. METHODS: To investigate the kinetics of immune parameters in M. vogae-infected mice, we detected, using flow cytometry, the expression of selected lymphoid and myeloid markers within the peritoneal cell population at day 0, 3, 6, 10, 14, 19, 25, 30 and 35 post-infection. Then, using ELISA, we analyzed the cytokine IFN-γ, TGF-ß, IL-4 and IL-10 responses and the levels of anti-M. vogae IgG and IgM antibodies in the peritoneal lavage fluid. Cells isolated from the peritoneal cavity were subjected to further molecular analysis. To assess cell activation, peritoneal cells were exposed to LPS, and culture supernatants were collected and assayed for the level of cytokines and production of nitrite. Ly6C+ and Ly6G+ cells were isolated using MACS from the peritoneal cells at day 35 post-infection. Both MACS-isolated subsets were co-cultured with preactivated T cells to measure their suppressive capacity. Next, the role of parasite excretory-secretory antigens in induction of CD11b+ myeloid cells with the suppressive phenotype and the production of IL-10 was examined. RESULTS: In the peritoneal cavity an initial increase of CD11b+Gr-1+F4/80highMHC IIhigh cells, NK, NKT cells and CD8+ cytotoxic T cells was observed in the first week of infection. At day 14 post-infection, an increase in the number of myeloid CD11b+Gr-1+ cells was detected, and most of this cell population expressed low levels of F4/80 and MHC II in later stages of infection, suggesting the impairment of antigen-presenting cell functions, probably through the excretory-secretory molecules. Moreover, we confirmed that peritoneal Gr1+ cells (Ly6C+ and Ly6G+ population) are phenotypically and functionally consistent with myeloid-derived suppressor cells. Metacestode infection elicited high levels of IL-10 and upregulated STAT-3 in peritoneal cells. A higher level of IgM suggests that this isotype may be predominant and is involved in the host protection. CONCLUSIONS: Mesocestoides vogae tetrathyridia induced the recruitment of immunosuppressive cell subsets, which may play a key role in the downregulation of immune response in long-term parasitic diseases, and excretory-secretory antigens seem to be the main regulatory factor.


Subject(s)
Cestode Infections/immunology , Immunity, Cellular , Immunity, Humoral , Mesocestoides/immunology , Peritoneum/immunology , Animals , Cytokines , Disease Models, Animal , Flow Cytometry , Male , Mesocestoides/pathogenicity , Mice , Mice, Inbred BALB C , Peritoneum/cytology , Peritoneum/parasitology
12.
BMJ Case Rep ; 14(1)2021 Jan 11.
Article in English | MEDLINE | ID: mdl-33431461

ABSTRACT

Peritoneal melanosis is an uncommon benign condition, the pathophysiology of which is unclear. Macroscopically, it appears as diffuse dark brown or black pigmentation within the peritoneum, mimicking more sinister conditions such as metastatic melanoma. It has been described in a variety of contexts, but only exceedingly rarely in association with metastatic melanoma, with only two previous published case reports. We present a case of peritoneal melanosis associated with metastatic melanoma involving the spleen, previously treated with targeted and immune checkpoint inhibitor therapy. With increasing reports of melanoma regression manifesting as cutaneous tumorous melanosis in patients treated with immune checkpoint inhibitors, we postulate that, similarly, immunotherapy and tumour regression might have a role to play in the pathogenesis of the peritoneal pigmentation in this case.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/adverse effects , Melanoma/therapy , Melanosis/diagnosis , Peritoneal Diseases/diagnosis , Skin Neoplasms/therapy , Splenic Neoplasms/surgery , Biopsy , Chemotherapy, Adjuvant , Humans , Immune Checkpoint Inhibitors/adverse effects , Male , Melanoma/complications , Melanoma/immunology , Melanoma/secondary , Melanosis/chemically induced , Melanosis/immunology , Melanosis/pathology , Middle Aged , Peritoneal Diseases/chemically induced , Peritoneal Diseases/immunology , Peritoneal Diseases/pathology , Peritoneum/drug effects , Peritoneum/immunology , Peritoneum/pathology , Positron-Emission Tomography , Protein Kinase Inhibitors/adverse effects , Skin Neoplasms/complications , Skin Neoplasms/immunology , Skin Neoplasms/pathology , Spleen/diagnostic imaging , Spleen/pathology , Spleen/surgery , Splenectomy , Splenic Neoplasms/diagnosis , Splenic Neoplasms/secondary
13.
Int J Biol Sci ; 17(1): 298-306, 2021.
Article in English | MEDLINE | ID: mdl-33390851

ABSTRACT

Postoperative adhesions (PA) are fibrotic tissues that are the most common driver of long-term morbidity after abdominal and pelvic surgery. The optimal drug or material to prevent adhesion formation has not yet been discovered. Comprehensive understanding of cellular and molecular mechanisms of adhesion process stimulates the design of future anti-adhesive strategies. Recently, disruption of peritoneal mesothelial cells were suggested as the 'motor' of PA formation, followed by a cascade of events (coagulation, inflammation, fibrinolysis) and influx of various immune cells, ultimately leading to a fibrous exudate. We showed that a variety of immune cells were recruited into adhesive peritoneal tissues in patients with small bowel obstruction caused by PA. The interactions among various types of immune cells contribute to PA development following peritoneal trauma. Our review focuses on the specific role of different immune cells in cellular and humoral mechanisms underpinning adhesion development.


Subject(s)
Peritoneum/immunology , Peritoneum/pathology , Tissue Adhesions/etiology , Animals , Fibrosis , Humans , Tissue Adhesions/prevention & control
14.
Curr Biol ; 31(1): 77-89.e5, 2021 01 11.
Article in English | MEDLINE | ID: mdl-33096038

ABSTRACT

Macrophages continuously survey their environment in search of pathogens or apoptotic corpses or debris. Targets intended for clearance expose ligands that initiate their phagocytosis ("eat me" signals), while others avoid phagocytosis by displaying inhibitory ligands ("don't eat me" signals). We report that such ligands can be obscured by the glycosaminoglycans and glycoproteins that coat pathogenic as well as malignant phagocytic targets. In addition, a reciprocal barrier of self-synthesized or acquired glycocalyx components on the macrophage surface shrouds phagocytic receptors, curtailing their ability to engage particles. The coating layers of macrophages and their targets hinder phagocytosis by both steric and electrostatic means. Their removal by enzymatic means is shown to markedly enhance phagocytic efficiency. In particular, we show that the removal of mucins, which are overexpressed in cancer cells, facilitates their clearance. These results shed light on the physical barriers that modulate phagocytosis, which have been heretofore underappreciated. VIDEO ABSTRACT.


Subject(s)
Candidiasis, Invasive/immunology , Glycocalyx/immunology , Neoplasms/immunology , Phagocytosis/immunology , Adult , Animals , Biological Products/pharmacology , Biological Products/therapeutic use , CD47 Antigen/antagonists & inhibitors , CD47 Antigen/immunology , CD47 Antigen/metabolism , Candida albicans/immunology , Candida albicans/metabolism , Candidiasis, Invasive/microbiology , Disease Models, Animal , Female , Glycocalyx/metabolism , Glycosaminoglycans/metabolism , Healthy Volunteers , Humans , Hyaluronic Acid/metabolism , Immunoglobulin G/pharmacology , Immunoglobulin G/therapeutic use , MCF-7 Cells , Macrophages/drug effects , Macrophages/immunology , Macrophages/metabolism , Male , Mice , Mucins/metabolism , Neoplasms/drug therapy , Neoplasms/pathology , Peritoneum/immunology , Peritoneum/microbiology , Phagocytes/drug effects , Phagocytes/immunology , Phagocytes/metabolism , Phagocytosis/drug effects , Primary Cell Culture , RAW 264.7 Cells , Synovial Fluid/cytology , Synovial Fluid/immunology , Synovial Fluid/metabolism , Young Adult
15.
Eur J Immunol ; 51(2): 354-367, 2021 02.
Article in English | MEDLINE | ID: mdl-32926407

ABSTRACT

Peritoneal dialysis (PD) employs hypertonic glucose to remove excess water and uremic waste. Peritoneal membrane failure limits its long-term use. T-cell cytokines promote this decline. T-cell differentiation is critically determined by the microenvironment. We here study how PD-range hypertonic glucose regulates T-cell polarization and IL-17 production. In the human peritoneal cavity, CD3+ cell numbers increased in PD. Single cell RNA sequencing detected expression of T helper (Th) 17 signature genes RORC and IL23R. In vitro, PD-range glucose stimulated spontaneous and amplified cytokine-induced Th17 polarization. Osmotic controls l-glucose and d-mannose demonstrate that induction of IL-17A is a substance-independent, tonicity dose-dependent process. PD-range glucose upregulated glycolysis and increased the proportion of dysfunctional mitochondria. Blockade of reactive-oxygen species (ROS) prevented IL-17A induction in response to PD-range glucose. Peritoneal mesothelium cultured with IL-17A or IL17F produced pro-inflammatory cytokines IL-6, CCL2, and CX3CL1. In PD patients, peritoneal IL-17A positively correlated with CX3CL1 concentrations. PD-range glucose-stimulated, but neither identically treated Il17a-/- Il17f-/- nor T cells cultured with the ROS scavenger N-acetylcysteine enhanced mesothelial CX3CL1 expression. Our data delineate PD-range hypertonic glucose as a novel inducer of Th17 polarization in a mitochondrial-ROS-dependent manner. Modulation of tonicity-mediated effects of PD solutions may improve membrane survival.


Subject(s)
Epithelium/immunology , Glucose/immunology , Inflammation/immunology , Interleukin-17/immunology , Peritoneum/immunology , Th17 Cells/immunology , Animals , Cells, Cultured , Chemokine CCL2/immunology , Chemokine CXCL1/immunology , Female , Humans , Interleukin-6/immunology , Male , Mannose/immunology , Mice , Mice, Inbred C57BL , Middle Aged , Mitochondria/immunology , Peritoneal Dialysis/methods , Reactive Oxygen Species/immunology , Up-Regulation/immunology
16.
Biochem Biophys Res Commun ; 530(1): 278-284, 2020 09 10.
Article in English | MEDLINE | ID: mdl-32828299

ABSTRACT

The disease burden of sepsis continues to increase, with intraabdominal contamination being a significant source of infection. Sepsis is a syndrome involving both an increase in systemic inflammation as well as a regulatory component. We have previously demonstrated that neutrophils are significant IL-10 producers in the abdomen during sepsis. Here, we sought to further characterize these neutrophils and elucidate potential underlying mechanisms resulting in IL-10 generation. Using transcriptional reporter mice, we observed that IL-10 producing neutrophils were activated, non-apoptotic, and expressed C-X-C chemokine receptor type 4-expressing. Further, we observed that active Signal Transducer and Activator of Transcription 1 expression was significantly increased in IL-10 producing versus non-IL-10 producing neutrophils. During sepsis, IFN-γ blockade lead to a decrease of neutrophil IL-10 production, while peritoneal CD4 T cells were found to be the most numerous acute producers of IFN-γ. Altogether, this report demonstrates that during sepsis, mature neutrophils can potentially dampen local inflammation by IL-10 production and this can be orchestrated by CD4 T cells through an IFN-γ dependent manner.


Subject(s)
Interferon-gamma/immunology , Interleukin-10/immunology , Neutrophils/immunology , Sepsis/immunology , Acute Disease , Animals , Apoptosis , Disease Models, Animal , Mice , Neutrophil Infiltration , Neutrophils/pathology , Peritoneum/immunology , Peritoneum/pathology , Sepsis/pathology
17.
Gynecol Oncol ; 159(1): 285-294, 2020 10.
Article in English | MEDLINE | ID: mdl-32732012

ABSTRACT

The age of cancer as an isolated single-cell concept is now behind us. It is now established that epithelial ovarian cancer, like other cancers, interacts with the healthy bystander cells to influence them and takes advantage of their nutritional, immunological, disseminating and other capacities. This interaction has become a therapeutic target, as shown by the numerous studies on this subject. Intraperitoneal chemo-hyperthermia has been part of the therapeutic armamentarium for some time yet its efficiency in ovarian cancer has only been recently proven in a randomized controlled trial. However, its therapeutic performance is not revolutionary and epithelial ovarian cancer maintains a high mortality. In this review, we studied the impact of HIPEC on the microenvironment and vice versa to determine whether it could be the key to this lukewarm efficacy. We began by exploring the modalities of HIPEC and establishing the reasons that make this treatment topical. Then, we examined its impact on each element of the tumor environment to obtain a global view of the resistance mechanisms at work in HIPEC.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Carcinoma, Ovarian Epithelial/therapy , Hyperthermic Intraperitoneal Chemotherapy/methods , Ovarian Neoplasms/therapy , Tumor Microenvironment/drug effects , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Ovarian Epithelial/immunology , Carcinoma, Ovarian Epithelial/pathology , Drug Resistance, Neoplasm , Extracellular Matrix/drug effects , Extracellular Matrix/immunology , Extracellular Matrix/pathology , Female , Humans , Lymphocytes, Tumor-Infiltrating/drug effects , Lymphocytes, Tumor-Infiltrating/immunology , Neoadjuvant Therapy/methods , Ovarian Neoplasms/immunology , Ovarian Neoplasms/pathology , Peritoneum/drug effects , Peritoneum/immunology , Peritoneum/pathology , Randomized Controlled Trials as Topic , Treatment Outcome , Tumor Microenvironment/immunology
18.
Semin Nucl Med ; 50(5): 405-418, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32768005

ABSTRACT

The peritoneum is the largest and most complex serous membrane in the human body. The peritoneal membrane is composed of a layer of mesothelium supported by a thin layer of connective tissue. The peritoneum is one continuous sheet, forming two layers and a potential space between them - the peritoneal cavity- which is subdivided into multiple communicating spaces containing small amount of serous fluid that facilitates frictionless movement of mobile intraabdominal viscera. Peritoneum also contributes to fluid exchange mechanism and plays a role in immune response. The peritoneum is subject to many neoplastic and non-neoplastic processes including infections, trauma, developmental and inflammatory processes. Different Nuclear Medicine imaging techniques can be used to diagnose peritoneal diseases, most of these techniques can be customized depending on the clinical scenario and expected findings. Peritoneal scintigraphy can detect abnormal peritoneal communication or compartmentalization. Several nuclear medicine techniques can help characterize intraperitoneal fluid collections and differentiate sterile from infected fluid. PET imaging plays an important role in imaging of different neoplastic and non-neoplastic peritoneal pathologies. Nuclear radiologists need to be familiar with peritoneal anatomy and pathology to interpret peritoneal findings in dedicated peritoneal nuclear medicine imaging studies, as part of more general nuclear medicine scans, or on CT or MRI component of hybrid imaging studies. The purpose of this article is to review the normal peritoneal anatomy, various pathologic processes involving the peritoneum, and different nuclear medicine and hybrid imaging techniques that can help detect, characterize, and follow up peritoneal pathology.


Subject(s)
Nuclear Medicine , Peritoneum , Humans , Peritoneum/anatomy & histology , Peritoneum/diagnostic imaging , Peritoneum/immunology
19.
Immunol Cell Biol ; 98(10): 854-867, 2020 11.
Article in English | MEDLINE | ID: mdl-32696994

ABSTRACT

Natural killer (NK) cells are innate effector cells with critical roles not only in tumor immunosurveillance and viral immunity, but also in bacterial and fungal infections. Toll-like receptor 2 (TLR2) can be important in the early and sustained immune responses to pathogens and tumors through the induction of cytokines and chemokines that recruit and activate immune effector cells. We investigated the role of TLR2 activation in NK cell recruitment with a view to informing approaches to induce or regulate peritoneal NK cell responses therapeutically. Peritoneal injection of TLR2 activators, including peptidoglycan and the lipopeptides FSL-1 and Pam3 CSK4 , resulted in NK cell recruitment after 16 h with increased NK cell numbers maintained for 48 h. TLR2 activators induced large amounts of CCR2 ligands, but much smaller amounts of CCR5 and CXCR3 ligands. Consistent with this observation, NK cell migration was abrogated in CCR2-deficient mice after peritoneal FSL-1 injection. Adoptive transfer of CCR2-deficient NK cells prior to peritoneal FSL-1 activation confirmed a cell-intrinsic component of CCR2-mediated NK cell migration. TLR2 activation did not induce an activated NK cell phenotype, but significant changes included an increase in the KLRG1+ subset and decreased NKG2D expression. Although not activated in vivo, peritoneal NK cells could be activated by interleukin (IL)-12 and IL-18 ex vivo to express CD69 and interferonγ. These data demonstrate that TLR2-mediated immune activation is a potent inducer of NK cell recruitment via a CCR2-dependent mechanism and that NK cells recruited by this mechanism can respond to additional signals to exert effector cell functions.


Subject(s)
Killer Cells, Natural/cytology , Peritoneum , Receptors, CCR2/genetics , Toll-Like Receptor 2 , Animals , Female , Mice , Mice, Inbred C57BL , Mice, Knockout , Peritoneum/immunology , Toll-Like Receptor 2/immunology
20.
Reprod Sci ; 27(10): 1920-1931, 2020 10.
Article in English | MEDLINE | ID: mdl-32572831

ABSTRACT

Endometriosis is a chronic inflammatory disease associated with an impaired immune response at the site of lesion implantation. The ability of macrophages to respond to changes in their environment is critical for an effective immune response. However, the existing knowledge of the peritoneal immune cell populations, their activation state and contribution to the immunological changes that occur in endometriosis are still controversial and inconclusive. In this study, we have examined the relative abundance of peritoneal macrophage subtypes, in women with (n = 21) versus without (n = 18) endometriosis and disease-associated changes in the adaptive T cell response. Using flow cytometry, we showed that peritoneal fluid monocyte/macrophages are composed of two populations of cells that exhibit major differences in the levels of the CD14 and CD68 markers, which we classified as the CD14+low/CD68+low and CD14+high/CD68+high subpopulations. Moreover, endometriosis-associated changes in the macrophage subtypes occurred only in the CD14+low/CD68+low subpopulation. In this subpopulation, we found an increased macrophage type 2 response that was coupled with an increase in peritoneal T-helper 2 and T-regulatory cell populations in women with endometriosis, compared with controls. In summary, this study resolves conflicting data in the literature regarding changes in the peritoneal immune cell population in endometriosis and identifies CD14+low/CD68+low macrophages as the subpopulation that changes in response to the disease.


Subject(s)
Endometriosis/immunology , Macrophages, Peritoneal/immunology , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Regulatory/immunology , Adult , Endometriosis/metabolism , Female , Flow Cytometry , Humans , Macrophages, Peritoneal/metabolism , Peritoneum/immunology , Peritoneum/metabolism , T-Lymphocytes, Helper-Inducer/metabolism , T-Lymphocytes, Regulatory/metabolism , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL