Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 63
Filter
Add more filters











Publication year range
1.
Front Immunol ; 12: 697760, 2021.
Article in English | MEDLINE | ID: mdl-34552582

ABSTRACT

Properdin, a positive regulator of complement alternative pathway, participates in renal ischemia-reperfusion (IR) injury and also acts as a pattern-recognition molecule affecting apoptotic T-cell clearance. However, the role of properdin in tubular epithelial cells (TECs) at the repair phase post IR injury is not well defined. This study revealed that properdin knockout (PKO) mice exhibited greater injury in renal function and histology than wild-type (WT) mice post 72-h IR, with more apoptotic cells and macrophages in tubular lumina, increased active caspase-3 and HMGB1, but better histological structure at 24 h. Raised erythropoietin receptor by IR was furthered by PKO and positively correlated with injury and repair markers. Properdin in WT kidneys was also upregulated by IR, while H2O2-increased properdin in TECs was reduced by its small-interfering RNA (siRNA), with raised HMGB1 and apoptosis. Moreover, the phagocytic ability of WT TECs, analyzed by pHrodo Escherichia coli bioparticles, was promoted by H2O2 but inhibited by PKO. These results were confirmed by counting phagocytosed H2O2-induced apoptotic TECs by in situ end labeling fragmented DNAs but not affected by additional serum with/without properdin. Taken together, PKO results in impaired phagocytosis at the repair phase post renal IR injury. Properdin locally produced by TECs plays crucial roles in optimizing damaged cells and regulating phagocytic ability of TECs to effectively clear apoptotic cells and reduce inflammation.


Subject(s)
Kidney/injuries , Kidney/pathology , Phagocytosis/physiology , Properdin/deficiency , Reperfusion Injury/pathology , Animals , Apoptosis/immunology , Apoptosis/physiology , Disease Models, Animal , Epithelial Cells/immunology , Epithelial Cells/pathology , Epithelial Cells/physiology , Kidney/blood supply , Macrophages/immunology , Macrophages/pathology , Macrophages/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Biological , Phagocytosis/immunology , Properdin/genetics , Properdin/immunology , Reperfusion Injury/immunology , Reperfusion Injury/physiopathology
2.
Ann Transl Med ; 9(2): 98, 2021 Jan.
Article in English | MEDLINE | ID: mdl-33569400

ABSTRACT

BACKGROUND: The initial response to islet transplantation and the subsequent acute inflammation is responsible for significant attrition of islets following both autologous and allogenic procedures. This multicentre study compares this inflammatory response using cytokine profiles and complement activation. METHODS: Inflammatory cytokine and complement pathway activity were examined in two cohorts of patients undergoing total pancreatectomy followed either by autologous (n=11) or allogenic (n=6) islet transplantation. Two patients who underwent total pancreatectomy alone (n=2) served as controls. RESULTS: The peak of cytokine production occurred immediately following induction of anaesthesia and during surgery. There was found to be a greater elevation of the following cytokines: TNF-alpha (P<0.01), MCP-1 (P=0.0013), MIP-1α (P=0.001), MIP-1ß (P=0.00020), IP-10 (P=0.001), IL-8 (P=0.004), IL-1α (P=0.001), IL-1ra (0.0018), IL-10 (P=0.001), GM-CSF (P=0.001), G-CSF (P=0.0198), and Eotaxin (P=0.01) in the allogenic group compared to autografts and controls. Complement activation and consumption was observed in all three pathways, and there were no significant differences in between the groups although following allogenic transplantation ∆IL-10 and ∆VEGF levels were significantly elevated those patients who became insulin-independent compared with those who were insulin-dependent. CONCLUSIONS: The cytokine profiles following islet transplantation suggests a significantly greater acute inflammatory response following allogenic islet transplantation compared with auto-transplantation although a significant, non-specific inflammatory response occurs following both forms of islet transplantation.

3.
FASEB Bioadv ; 3(1): 36-48, 2021 Jan.
Article in English | MEDLINE | ID: mdl-33490882

ABSTRACT

Chronic metabolic acidosis plays a role in cachexia by enhancing total proteolysis in skeletal muscle. Glucocorticoid also triggers proteolysis and plays a permissive role in the effect of acidosis. The System A amino acid transporter SNAT2/SLC38A2 is ubiquitously expressed in mammalian cells including muscle, performing Na+-dependent active import of neutral amino acids, and is strongly inhibited by low pH. Exposure of rat skeletal muscle cell line L6-G8C5 to low pH rapidly inhibits SNAT2 transport activity and enhances total proteolysis rate. Pharmacological inhibition or silencing of SNAT2 also enhances proteolysis. This study tests the hypothesis that the glucocorticoid dexamethasone (DEX), like low pH, inhibits SNAT2 activity in L6-G8C5 myotubes, thus contributing to total proteolysis. Incubation with 500 nM DEX for 4 h reduced the System A amino acid transport rate to half the rate in control cultures. This inhibition depended on glucocorticoid receptor-mediated gene transcription, but SNAT2 mRNA levels were unaffected by DEX. In contrast, the SNAT2 protein assessed by immunoblotting was significantly depleted. The co-inhibitory effects of DEX and low pH on System A transport activity were additive in stimulating total proteolysis. In keeping with this mechanism, DEX's inhibitory effect on SNAT2 transport activity was significantly blunted by the proteasome inhibitor MG132. Proof of principle was achieved in similar experiments using recombinant expression of a GFP-tagged SNAT2 fusion protein in HEK293A cells. It is concluded that DEX acutely depletes the SNAT2 transporter protein, at least partly through proteasome-dependent degradation of this functionally important transporter.

4.
Medicina (Kaunas) ; 57(2)2021 Jan 21.
Article in English | MEDLINE | ID: mdl-33494138

ABSTRACT

Background and Objectives: Tumours are often low immunogenic. The role of complement, an innate immune defence system, in tumour control has begun to be elucidated, but findings are conflicting. A role for properdin, an amplifier of complement activation, in tumour control has recently been implicated. Materials and Methods: Properdin-deficient and congenic wildtype mice were injected subcutaneously with B16F10 melanoma cells. Tumour mass and chemokine profile were assessed. The frequencies of CD45+CD11b+ Gr-1+ cells were determined from tumours and spleens, and CD206+ F4/80+ cells were evaluated in spleens. Sera were analysed for C5a, sC5b-9, and CCL2. Results: Whilst there was no difference in tumour growth at study endpoint, properdin-deficient mice had significantly fewer myeloid-derived suppressor cells (MDSCs) in their tumours and spleens. Splenic M2 type macrophages and serum levels of C5a, sC5b-9, and CCL2 were decreased in properdin-deficient compared to wildtype mice. Conclusions: The presence of intact complement amplification sustains an environment that lessens potential anti-tumour responses.


Subject(s)
Disease Models, Animal , Melanoma , Properdin , Skin Neoplasms , Animals , Macrophages , Melanoma/genetics , Mice , Mice, Inbred C57BL , Properdin/genetics , Skin Neoplasms/genetics
5.
Medicina (Kaunas) ; 56(10)2020 Oct 13.
Article in English | MEDLINE | ID: mdl-33065972

ABSTRACT

Systemic lupus erythematosus is a classical systemic autoimmune disease that overactivates complement and can affect all organs. Early diagnosis and effective management are important in this immune-complex-mediated chronic inflammatory disease, which has a strong component of vasculitis and carries an increased risk of thrombosis, even in the absence of antiphospholipid antibodies. Development of lupus nephritis can be life limiting but is managed with dialysis and renal transplantation. Therefore, data have become available that cardiovascular risk poses a serious feature of systemic lupus erythematosus that requires monitoring and prospective treatment. Cell-derived microparticles circulate in plasma and thereby intersect the humoral and cellular component of inflammation. They are involved in disease pathophysiology, particularly thrombosis, and represent a known cardiovascular risk. This viewpoint argues that a focus on characteristics of circulating microparticles measured in patients with systemic lupus erythematosus may help to classify certain ethnic groups who are especially at additional risk of experiencing cardiovascular complications.


Subject(s)
Cardiovascular Diseases , Cell-Derived Microparticles , Lupus Erythematosus, Systemic , Cardiovascular Diseases/epidemiology , Cardiovascular Diseases/etiology , Complement Activation , Heart Disease Risk Factors , Humans , Lupus Erythematosus, Systemic/complications , Prospective Studies , Renal Dialysis , Risk Factors
6.
Medicina (Kaunas) ; 56(9)2020 Sep 22.
Article in English | MEDLINE | ID: mdl-32971872

ABSTRACT

Background and objectives: Overnutrition leads to a metabolic and inflammatory response that includes the activation of Complement. Properdin is the only amplifier of complement activation and increases the provision of complement activation products. Its absence has previously been shown to lead to increased obesity in mice on a high fat diet. The aim of this study was to determine ways in which properdin contributes to a less pronounced obese phenotype. Materials and Methods: Wild type (WT) and properdin deficient mice (KO) were fed a high-fat diet (HFD) for up to 12 weeks. Results: There was a significant increase in liver triglyceride content in the KO HFD group compared to WT on HFD. WT developed steatosis. KO had an additional inflammatory component (steatohepatitis). Analysis of AKT signalling by phosphorylation array supported a decrease in insulin sensitivity which was greater for KO than WT in liver and kidney. There was a significant decrease of C5L2 in the fat membranes of the KO HFD group compared to the WT HFD group. Circulating microparticles in KO HFD group showed lower presence of C5L2. Expression of the fatty acid transporter CD36 in adipose tissue was increased in KO on HFD and was also significantly increased in plasma of KO HFD mice compared to WT on HFD. CD36 was elevated on microparticles from KO on HFD. Ultrastructural changes consistent with obesity-associated glomerulopathy were observed for both HFD fed genotypes, but tubular strain was greater in KO. Conclusion: Our work demonstrates that complement properdin is a dominant factor in limiting the severity of obesity-associated conditions that impact on liver and kidney. The two receptors, C5L2 and CD36, are downstream of the activity exerted by properdin.


Subject(s)
Diet, High-Fat , Insulin Resistance , Animals , Diet, High-Fat/adverse effects , Mice , Mice, Knockout , Obesity , Properdin/genetics
7.
Medicina (Kaunas) ; 56(9)2020 Aug 27.
Article in English | MEDLINE | ID: mdl-32867176

ABSTRACT

Background and objects: In systemic lupus erythematosus, circulating immune complexes activate complement and, when trapped in renal capillaries, cause glomerulonephritis. Mouse models have been used in the preclinical assessment of targeting complement activation pathways to manage chronic inflammation in lupus. Properdin is the only known positive regulator of complement activation, but its role in the severity of lupus nephritis has not been studied yet. Materials and Methods: Fully characterized properdin-deficient mice were crossed with lupus prone MRL/lpr mice on C57Bl/6 background. Results: Compared to MRL/lpr properdin wildtype mice, MRL/lpr properdin-deficient mice had significantly lower anti-DNA antibody titres, TNFα and BAFF levels in serum. The qualitative glomerulonephritic score was less severe and there was significantly less serum creatinine in MRL/lpr properdin-deficient mice compared to MRL/lpr properdin wildtype littermate mice. Conclusion: Properdin plays a significant role in the severity of lupus overall and specifically in the extent of glomerulonephritis observed in MRL/lpr mice. Because MRL/lpr properdin-deficient mice had lower levels of anti-DNA antibodies, inflammatory mediators and markers of renal impairment, the study implies that properdin could constitute a novel therapy target in lupus disease.


Subject(s)
Complement Pathway, Alternative , Lupus Nephritis/immunology , Properdin/physiology , Animals , Antibodies, Antinuclear/blood , B-Cell Activating Factor/blood , Biomarkers/blood , Creatinine/blood , Disease Models, Animal , Kidney/pathology , Lupus Nephritis/blood , Lupus Nephritis/pathology , Male , Mice, Inbred MRL lpr , Severity of Illness Index , Tumor Necrosis Factor-alpha/blood
8.
Medicina (Kaunas) ; 56(8)2020 Aug 12.
Article in English | MEDLINE | ID: mdl-32806638

ABSTRACT

Asthma is an important respiratory illness. Though pharmacological and biological treatment is well established and is staged according to endotypes and their responses to treatment, novel avenues are being explored. Our focus is complement. In this viewpoint, we evaluate the approach to target complement in this complex hypersensitivity reaction that develops chronicity and has a personal-as well as a societal-cost.


Subject(s)
Asthma/drug therapy , Asthma/immunology , Complement C3a/immunology , Complement C5a/immunology , Complement Inactivating Agents/therapeutic use , Antibodies, Monoclonal, Humanized/therapeutic use , Humans
9.
Medicina (Kaunas) ; 56(6)2020 Jun 26.
Article in English | MEDLINE | ID: mdl-32604873

ABSTRACT

Background and objectives: Kidneys from donation after circulatory death (DCD) are more likely to be declined for transplantation compared with kidneys from donation after brain death (DBD). The aim of this study was to evaluate characteristics in the biopsies of human DCD and DBD kidneys that were declined for transplantation in order to rescue more DCD kidneys. Materials and Methods: Sixty kidney donors (DCD = 36, DBD = 24) were recruited into the study and assessed using donor demographics. Kidney biopsies taken post cold storage were also evaluated for histological damage, inflammation (myeloperoxidase, MPO), von Willebrand factor (vWF) expression, complement 4d (C4d) deposition and complement 3 (C3) activation using H&E and immunohistochemistry staining, and Western blotting. Results: More DBD donors (16/24) had a history of hypertension compared with DCDs (8/36, p = 0.001). The mean warm ischemic time in the DCD kidneys was 12.9 ± 3.9 min. The mean cold ischemic time was not significantly different between the two groups of kidney donors (DBD 33.3 ± 16.7 vs. DCD 28.6 ± 14.1 h, p > 0.05). The score of histological damage and MPO, as well as the reactivity of vWF, C4d and C3, varied between kidneys, but there was no significant difference between the two donor types (p > 0.05). However, vWF reactivity might be an early indicator for loss of tissue integrity, while C4d deposition and activated C3 might be better predictors for histological damage. Conclusions: Similar characteristics of DCD were shown in comparison with DBD kidneys. Importantly, the additional warm ischemic time in DCD appeared to have no further detectable adverse effects on tissue injury, inflammation and complement activation. vWF, C4d and C3 might be potential biomarkers facilitating the evaluation of donor kidneys.


Subject(s)
Kidney/abnormalities , Tissue and Organ Procurement/standards , Aged , Biopsy/methods , Brain Death/physiopathology , Female , Humans , Immunohistochemistry/methods , Kidney/pathology , Kidney/physiopathology , Kidney Function Tests/methods , Kidney Function Tests/statistics & numerical data , Kidney Transplantation/methods , Male , Middle Aged , Risk Factors , Tissue and Organ Procurement/statistics & numerical data , United Kingdom
10.
Front Cell Dev Biol ; 8: 218, 2020.
Article in English | MEDLINE | ID: mdl-32309282

ABSTRACT

The average human life expectancy has increased globally, and continues to rise, owing to the substantive progress made in healthcare, medicine, sanitation, housing and education. This ultimately enriches society with a greater proportion of elderly people. Sustaining a healthy aged population is key to diminish the societal and economic impact of age-related infirmities. This is especially challenging because tissue function, and thus wellbeing, naturally progressively decline as humans age. With age increasing the risk of developing diseases, one of the therapeutic options is to interfere with the molecular and cellular pathways involved in age-related tissue dysfunction, which is in part caused by the accumulation of senescent cells. One strategy to prevent this could be using drugs that selectively kill these cells (senolytics). In parallel, some compounds have been identified that prevent or slow down the progression of senescence or some of its features (senostatics). Senolytic and senostatic therapies have been shown to be efficient in vivo, but they also have unwanted dose-dependent side effects, including toxicity. Important advances might be made using bioactive compounds from plants and foods (nutraceuticals) if, as is proposed, they offer similar effectiveness with fewer side effects. The focus of this review is on the use of nutraceuticals in interfering with cellular senescence.

11.
Ann Transl Med ; 8(5): 170, 2020 Mar.
Article in English | MEDLINE | ID: mdl-32309317

ABSTRACT

BACKGROUND: Numerous factors influence pancreatic islet survival following auto-transplantation. Of these, the host immune response in the early peri-operative period is one of the most important. In this study we investigated the role of the mannose-binding lectin (MBL)-dependent pathway in a group of total pancreatectomy (TP) islet auto-transplantation (TPIAT) patients and classified them as competent or deficient in MBL activity. Complement pathway activities, MBL protein and inflammatory cytokine concentrations were evaluated from eleven pancreatic islet auto-transplant patients from two institutions. METHODS: Eleven patients from two institutions were prospectively recruited. Serum was screened at different time points for 29 different cytokines and compared according to their MBL deficient or competent status. Twelve patients from previous TPIAT patients also underwent screening of MBL pathway activity. RESULTS: A total nine of twenty three patients (39%) were MBL pathway deficient. MCP-1, IL-7 and IL-1a concentrations were significantly lower in the MBL deficient cohort compared to the normal MBL group (P=0.0237, 0.0001 and 0.0051 respectively). IL-6 and IL-8 concentrations were significantly raised in the normal MBL group. MBL functional activity was lower in insulin-independent group compared to the insulin-dependent group. CONCLUSIONS: Complement activation is an important, possibly damaging response during intra-portal islet infusion. MBL pathway deficiency appears common in this population and the cytokine response was attenuated in MBL pathway deficient patients. Therapeutic MBL pathway blockade during and following islet auto-transplantation (IAT) may improve islet survival and function and thereby clinical outcome.

12.
Front Immunol ; 11: 614980, 2020.
Article in English | MEDLINE | ID: mdl-33542722

ABSTRACT

The complement system is readily triggered by the presence of damage-associated molecular patterns on the surface of tumor cells. The complement alternative pathway provides rapid amplification of the molecular stress signal, leading to complement cascade activation to deal with pathogens or malignant cells. Properdin is the only known positive regulator of the alternative pathway. In addition, properdin promotes the phagocytic uptake of apoptotic T cells by macrophages and dendritic cells without activating the complement system, thus, establishing its ability to recognize "altered-self". Dysregulation of properdin has been implicated in substantial tissue damage in the host, and in some cases, chronic unresolved inflammation. A corollary of this may be the development of cancer. Hence, to establish a correlation between properdin presence/levels in normal and cancer tissues, we performed bioinformatics analysis, using Oncomine and UALCAN. Survival analyses were performed using UALCAN and PROGgeneV2 to assess if properdin can serve as a potential prognostic marker for human lung adenocarcinoma (LUAD), liver hepatocellular carcinoma (LIHC), cervical squamous cell carcinoma (CESC), and pancreatic adenocarcinoma (PAAD). We also analyzed levels of tumor-infiltrating immune cells using TIMER, a tool for characterizing immune cell composition in cancers. We found that in LUAD and LIHC, there was a lower expression of properdin in the tumors compared to normal tissues, while no significant difference was observed in CESC and PAAD. Survival analysis demonstrated a positive association between properdin mRNA expression and overall survival in all 4 types of cancers. TIMER analysis revealed that properdin expression correlated negatively with tumor purity and positively with levels of infiltrating B cells, cytotoxic CD8+ T cells, CD4+ helper T cells, macrophages, neutrophils and dendritic cells in LUAD, CESC and PAAD, and with levels of B cells, CD8+ T cells and dendritic cells in LIHC. Immunohistochemical analysis revealed that infiltrating immune cells were the most likely source of properdin in the tumor microenvironment. Thus, complement protein properdin shows promise as a prognostic marker in cancer and warrants further study.


Subject(s)
Neoplasms/mortality , Properdin/analysis , Complement Pathway, Alternative , Data Mining , Datasets as Topic , Dendritic Cells/immunology , Female , Humans , Lymphocyte Subsets/immunology , Lymphocytes, Tumor-Infiltrating , Macrophages/immunology , Male , Neoplasms/chemistry , Neoplasms/immunology , Neoplasms/pathology , Neutrophils/immunology , Prognosis , RNA, Messenger/analysis , RNA, Neoplasm/analysis , Transcriptome , Tumor Microenvironment/immunology
13.
Front Immunol ; 10: 2610, 2019.
Article in English | MEDLINE | ID: mdl-31849925

ABSTRACT

Background: Hypoxic-ischemic (HI) encephalopathy is a major cause of neonatal mortality and morbidity, with a global incidence of 3 per 1,000 live births. Intrauterine or perinatal complications, including maternal infection, constitute a major risk for the development of neonatal HI brain damage. During HI, inflammatory response and oxidative stress occur, causing subsequent cell death. The presence of an infection sensitizes the neonatal brain, making it more vulnerable to the HI damage. Currently, therapeutic hypothermia is the only clinically approved treatment available for HI encephalopathy, however it is only partially effective in HI alone and its application in infection-sensitized HI is debatable. Therefore, there is an unmet clinical need for the development of novel therapeutic interventions for the treatment of HI. Such an alternative is targeting the complement system. Properdin, which is involved in stabilization of the alternative pathway convertases, is the only known positive regulator of alternative complement activation. Absence of the classical pathway in the neonatal HI brain is neuroprotective. However, there is a paucity of data on the participation of the alternative pathway and in particular the role of properdin in HI brain damage. Objectives: Our study aimed to validate the effect of global properdin deletion in two mouse models: HI alone and LPS-sensitized HI, thus addressing two different clinical scenarios. Results: Our results indicate that global properdin deletion in a Rice-Vannucci model of neonatal HI and LPS-sensitized HI brain damage, in the short term, clearly reduced forebrain cell death and microglial activation, as well as tissue loss. In HI alone, deletion of properdin reduced TUNEL+ cell death and microglial post-HI response at 48 h post insult. Under the conditions of LPS-sensitized HI, properdin deletion diminished TUNEL+ cell death, tissue loss and microglial activation at 48 h post-HI. Conclusion: Overall, our data suggests a critical role for properdin, and possibly also a contribution in neonatal HI alone and in infection-sensitized HI brain damage. Thus, properdin can be considered a novel target for treatment of neonatal HI brain damage.


Subject(s)
Hypoxia-Ischemia, Brain/immunology , Neuroprotection , Properdin/physiology , Animals , Complement System Proteins/physiology , Humans , Hypoxia-Ischemia, Brain/etiology , Hypoxia-Ischemia, Brain/pathology , Infant, Newborn , Interleukin-6/physiology , Lipopolysaccharides/pharmacology , Male , Mice , Mice, Inbred C57BL , Microglia/physiology
14.
Front Immunol ; 10: 1792, 2019.
Article in English | MEDLINE | ID: mdl-31428094

ABSTRACT

Multiple myeloma is a life-threatening hematological malignancy, which is rarely curable by conventional therapies. Immunotherapy, using tumor antigen-specific, cytotoxic T-lymphocytes, may represent an alternative or additional treatment for multiple myeloma. In this study, we used hybrid cell lines, generated by fusion of an EBV B-lymphoblastoid cell line (B-LCL) and myeloma cells, to stimulate in vitro peripheral blood lymphocytes (PBLs) from patients with multiple myeloma. We investigated induction of antigen-specific, cytotoxic T-lymphocytes to the well-defined tumor associated antigens (TAAs) hTERT, MUC1, MAGE-C1 and CS1, which have been shown to be expressed in a high proportion of cases of multiple myeloma. HLA-A2-peptide pentamer staining, interferon-γ and perforin ELISpot assays, as well as cytotoxicity assays were used. Following several rounds of in vitro stimulation, the hybrid cell lines induced antigen-specific, cytotoxic T-lymphocytes to four candidate TAAs in PBLs from HLA-A2+ multiple myeloma patients, using known HLA-A2 restricted peptide epitopes of the TAAs. In contrast, the HLA-A2+ myeloma cell line U266 failed to induce antigen-specific, cytotoxic T-lymphocytes in vitro. Our data indicate that B-LCL/myeloma hybrid cell lines induce antigen-specific, cytotoxic T-lymphocytes in PBLs isolated from multiple myeloma patients in vitro and may represent a novel strategy for use in adoptive immunotherapy of multiple myeloma.


Subject(s)
Multiple Myeloma/immunology , Multiple Myeloma/therapy , T-Lymphocytes, Cytotoxic/immunology , Antigens, Neoplasm/immunology , Cell Line, Tumor , Epitopes, T-Lymphocyte/immunology , HLA-A2 Antigen/immunology , Humans , Immunotherapy, Adoptive/methods , Interferon-gamma/immunology , Peptides/immunology
15.
Front Immunol ; 10: 1404, 2019.
Article in English | MEDLINE | ID: mdl-31316501

ABSTRACT

Background: The mechanisms connecting dietary intake of processed foods with systemic inflammatory markers and cardiovascular risk remain poorly defined. We sought to compare the abundance of pro-inflammatory stimulants of innate immune receptors in processed foods with those produced by the murine ileal and caecal microbiota, and to explore the impact of their ingestion on systemic inflammation and lipid metabolism in vivo. Methods and results: Calibrated receptor-dependent reporter assays revealed that many processed foods, particularly those based on minced meats, contain pro-inflammatory stimulants of Toll-like receptor (TLR)-2 and TLR4 at concentrations which greatly exceed those produced by the endogenous murine ileal microbiota. Chronic dietary supplementation with these stimulants, at concentrations relevant to those measured in the Western diet, promoted hepatic inflammation and reduced several markers of reverse cholesterol transport (RCT) in mice. Hepatocytes were found to be insensitive to TLR2- and TLR4-stimulants directly, but their secretion of functional cholesterol acceptors was impaired by interleukin (IL)-1ß released by TLR-responsive hepatic macrophages. Hepatic macrophage priming by high-fat diet enhanced the impairment of RCT by ingested endotoxin, and this was reversed by macrophage depletion via clodronate liposome treatment, or genetic deficiency in the IL-1 receptor. Conclusion: These findings reveal an unexpected mechanism connecting processed food consumption with cardiovascular risk factors, and introduce the food microbiota as a potential target for therapeutic regulation of lipid metabolism.


Subject(s)
Cholesterol/immunology , Inflammation/immunology , Interleukin-1/immunology , Liver/immunology , Macrophages/immunology , Toll-Like Receptors/immunology , Adult , Animals , Biological Transport , Cells, Cultured , Cholesterol/metabolism , Diet , Gastrointestinal Microbiome/immunology , Gastrointestinal Microbiome/physiology , HEK293 Cells , Hep G2 Cells , Humans , Inflammation/metabolism , Interleukin-1/biosynthesis , Liver/metabolism , Liver/pathology , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Pathogen-Associated Molecular Pattern Molecules/immunology , Pathogen-Associated Molecular Pattern Molecules/metabolism , RAW 264.7 Cells , Toll-Like Receptors/metabolism
16.
Clin Exp Med ; 19(3): 321-337, 2019 Aug.
Article in English | MEDLINE | ID: mdl-30191349

ABSTRACT

Septicaemia is an acute inflammatory reaction in the bloodstream to the presence of pathogen-associated molecular patterns. Whole blood stimulation assays capture endotoxin-induced formation of aggregates between platelets and leucocytes using flow cytometry. We wanted to assess extent of spontaneous aggregate formation in whole blood stimulation assays and compare the effects of endotoxin and heat-killed, clinically relevant, bacterial pathogens on aggregate formation and then on adhesion of aggregates to TNFα-stimulated endothelial cells. We found that endotoxin (from Escherichia coli or Salmonella enteritidis) was not a suitable stimulus to provoke platelet-leucocyte aggregates in vitro, as it did not further increase the extent of aggregates formed spontaneously in stasis of hirudin-anticoagulated blood. Specifically, whole blood samples stimulated with or without LPS produced aggregates with a mean surface area of 140.97 and 117.68 µm2, respectively. By contrast, incubation of whole blood with heat-killed Klebsiella pneumoniae or Staphylococcus aureus produced significantly enhanced and complex cellular aggregates (with a mean surface area of 470.61 and 518.39 µm2, respectively) which adhered more frequently to TNFα (and free fatty acid)-stimulated endothelial cells. These were reliably captured by scanning electron microscopy. Adhesion of cellular aggregates could be blocked by incubation of endothelial cells with a commercial P-selectin antibody and an angiopoietin-2 ligand trap. In conclusion, we have developed an in vitro method that models the acute inflammatory reaction in whole blood in the presence of sepsis-relevant bacterial pathogen surfaces.


Subject(s)
Blood Platelets/pathology , Cell Adhesion , Cell Aggregation , Endothelial Cells/pathology , Leukocytes/pathology , Models, Theoretical , Sepsis/pathology , Adult , Cells, Cultured , Coculture Techniques , Escherichia coli Infections/pathology , Female , Humans , Klebsiella Infections/pathology , Male , Middle Aged , Salmonella Infections/pathology , Staphylococcal Infections/pathology
17.
Kidney Int ; 94(6): 1141-1150, 2018 12.
Article in English | MEDLINE | ID: mdl-30322716

ABSTRACT

Properdin is the only known positive regulator of complement activation by stabilizing the alternative pathway convertase through C3 binding, thus prolonging its half-life. Recent in vitro studies suggest that properdin may act as a specific pattern recognition molecule. To better understand the role of properdin in vivo, we used an experimental model of acute anti-glomerular basement membrane disease with wild-type, C3- and properdin knockout mice. The model exhibited severe proteinuria, acute neutrophil infiltration and activation, classical and alternative pathway activation, and progressive glomerular deposition of properdin, C3 and C9. Although the acute renal injury was likely due to acute neutrophil activation, we found properdin deposition in C3-knockout mice that was not associated with IgG. Thus, properdin may deposit in injured tissues in vivo independent of its main ligand C3.


Subject(s)
Anti-Glomerular Basement Membrane Disease/immunology , Complement Activation/immunology , Complement C3/immunology , Properdin/immunology , Animals , Anti-Glomerular Basement Membrane Disease/pathology , Complement C3/genetics , Complement C3/metabolism , Disease Models, Animal , Female , Glomerular Basement Membrane/cytology , Glomerular Basement Membrane/immunology , Glomerular Basement Membrane/pathology , Humans , Immunoglobulin G/administration & dosage , Immunoglobulin G/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neutrophils/immunology , Properdin/genetics , Properdin/metabolism , Protein Binding/immunology
18.
Front Immunol ; 9: 131, 2018.
Article in English | MEDLINE | ID: mdl-29483907

ABSTRACT

Development of nanoparticles as tissue-specific drug delivery platforms can be considerably influenced by the complement system because of their inherent pro-inflammatory and tumorigenic consequences. The complement activation pathways, and its recognition subcomponents, can modulate clearance of the nanoparticles and subsequent inflammatory response and thus alter the intended translational applications. Here, we report, for the first time, that human properdin, an upregulator of the complement alternative pathway, can opsonize functionalized carbon nanotubes (CNTs) via its thrombospondin type I repeat (TSR) 4 and 5. Binding of properdin and TSR4+5 is likely to involve charge pattern/polarity recognition of the CNT surface since both carboxymethyl cellulose-coated carbon nanotubes (CMC-CNT) and oxidized (Ox-CNT) bound these proteins well. Properdin enhanced the uptake of CMC-CNTs by a macrophage cell line, THP-1, mounting a robust pro-inflammatory immune response, as revealed by qRT-PCR, multiplex cytokine array, and NF-κB nuclear translocation analyses. Properdin can be locally synthesized by immune cells in an inflammatory microenvironment, and thus, its interaction with nanoparticles is of considerable importance. In addition, recombinant TSR4+5 coated on the CMC-CNTs inhibited complement consumption by CMC-CNTs, suggesting that nanoparticle decoration with TSR4+5, can be potentially used as a complement inhibitor in a number of pathological contexts arising due to exaggerated complement activation.


Subject(s)
ADAMTS Proteins/immunology , Macrophages/immunology , Nanotubes, Carbon/chemistry , Properdin/immunology , ADAMTS Proteins/genetics , Carboxymethylcellulose Sodium/chemistry , Complement Activation , Cytokines/genetics , HEK293 Cells , Humans , Inflammation/immunology , Properdin/genetics , Protein Binding , THP-1 Cells
19.
Immun Inflamm Dis ; 5(2): 151-162, 2017 06.
Article in English | MEDLINE | ID: mdl-28474500

ABSTRACT

INTRODUCTION: Fatty liver disease is prevalent in populations with high caloric intake. Nutritherapeutic approaches are being considered, such as supplementary Vitamin D3 , to improve aspects of metabolic syndrome, namely fatty liver disease, hyperlipidemia, and insulin resistance associated with obesity. METHODS: We analyzed female LDLR-/- and LDLR+/+ mice on a 10-week diabetogenic diet for markers of fatty liver disease, metabolic strain, and inflammation. RESULTS: The groups on a high fat high sugar diet with supplementary Vitamin D3 , in comparison with the groups on a high fat high sugar diet alone, showed improved transaminase levels, significantly less hypertriglyceridemia and hyperinsulinemia, and histologically, there was less pericentral hepatic steatosis. Levels of non-esterified fatty acids and lipid peroxidation products were significantly lower in the group supplemented with additional Vitamin D3 , as were systemic markers of inflammation (serum endotoxin and IL-6). M2 macrophage phenotype predominated in the group supplemented with additional Vitamin D3 . Beneficial changes were observed as early as five weeks' supplementation with Vitamin D3 and extended to restoration of high fat high sugar diet induced decrease of bone mineral density. CONCLUSION: In summary, Vitamin D3 was a significantly beneficial dietary additive to blunt a prediabetic phenotype in diet-induced obesity of female LDLR-/- and LDLR+/+ mice.


Subject(s)
Cholecalciferol/pharmacology , Dietary Carbohydrates/adverse effects , Dietary Fats/adverse effects , Dietary Supplements , Prediabetic State/prevention & control , Receptors, LDL/metabolism , Animals , Dietary Carbohydrates/pharmacology , Dietary Fats/pharmacology , Female , Mice , Mice, Knockout , Prediabetic State/chemically induced , Prediabetic State/genetics , Prediabetic State/metabolism , Receptors, LDL/genetics
20.
Immun Inflamm Dis ; 5(1): 68-77, 2017 03.
Article in English | MEDLINE | ID: mdl-28250926

ABSTRACT

INTRODUCTION: The tumour microenvironment is shaped by the interaction of immune, non immune, and tumour cells present in close proximity. Tumour cells direct the development of a locally immune suppressed state, affecting the activity of anti tumour T cells and preparing the escape phase of tumour development. Macrophages in the tumour typically develop into so-called tumour associated macrophages with a distinct profile of activities which lead to a reduction in inflammation and antigen presentation. The direct impact of tumour cell conditioned medium on the activity profile of macrophages in dependence of their complement component expression has not yet been investigated. METHODS: In our in vitro study, macrophages differentiated from bone marrows of properdin deficient and wildtype mice were stimulated with conditioned medium of a syngeneic tumour cell line, B16F10, a mouse melanoma subline. RESULTS: In comparison with macrophages from wildtype mice, those from congenic properdin deficient mice showed skewing towards M2 profile, encompassing mRNA expression for genes involved in arginine metabolism, production of type 2 cytokines, and relatively lower surface expression of molecules needed for antigen presentation. CONCLUSIONS: These data suggest that properdin insufficiency promotes a tumour environment that helps the tumour evade the immune response.


Subject(s)
Macrophages/cytology , Melanoma, Experimental/immunology , Properdin/deficiency , Animals , Bone Marrow Cells/cytology , Cell Differentiation , Cell Line, Tumor , Culture Media, Conditioned , Cytokines/genetics , Female , Macrophages/immunology , Male , Mice
SELECTION OF CITATIONS
SEARCH DETAIL