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

Affiliation country
Publication year range
1.
J Immunol ; 205(5): 1385-1392, 2020 09 01.
Article in English | MEDLINE | ID: mdl-32759297

ABSTRACT

ß2-Glycoprotein I (ß2-GPI) is an abundant plasma glycoprotein with unknown physiological function and is currently recognized as the main target of antiphospholipid Abs responsible for complement activation and vascular thrombosis in patients with antiphospholipid syndrome (APS). In this study, we provide evidence that mannose-binding lectin (MBL) binds to ß2-GPI in Ca++ and a dose-dependent manner and that this interaction activates complement and promotes complement-dependent thrombin generation. Surprisingly, a significant binding was observed between MBL and isolated domains II and IV of ß2-GPI, whereas the carbohydrate chains, domain I and domain V, were not involved in the interaction, documenting a noncanonical binding mode between MBL and ß2-GPI. Importantly, this interaction may occur on endothelial cells because binding of MBL to ß2-GPI was detected on the surface of HUVECs, and colocalization of MBL with ß2-GPI was observed on the endothelium of a biopsy specimen of a femoral artery from an APS patient. Because ß2-GPI-mediated MBL-dependent thrombin generation was increased after priming the endothelium with TNF-α, our data suggests that this mechanism could play an important yet unrecognized role under physiological conditions and may be upregulated in pathological situations. Moreover, the complement activation and the procoagulant effects of the ß2-GPI/MBL complex may contribute to amplify similar activities of anti-ß2-GPI Abs in APS and possibly act independently of Abs, raising the issue of developing appropriate therapies to avoid recurrences and disability in patients at risk for these clinical conditions.


Subject(s)
Complement Activation/immunology , Mannose-Binding Lectin/metabolism , Thrombin/metabolism , beta 2-Glycoprotein I/metabolism , Antiphospholipid Syndrome/immunology , Antiphospholipid Syndrome/metabolism , Calcium/metabolism , Cell Line , Endothelial Cells/immunology , Endothelial Cells/metabolism , Endothelium/immunology , Endothelium/metabolism , Human Umbilical Vein Endothelial Cells , Humans , Mannose-Binding Lectin/immunology , Protein Binding/immunology , Thrombin/immunology , Thrombosis/immunology , Thrombosis/metabolism , Tumor Necrosis Factor-alpha/immunology , Tumor Necrosis Factor-alpha/metabolism , beta 2-Glycoprotein I/immunology
2.
J Allergy Clin Immunol ; 142(3): 883-891, 2018 09.
Article in English | MEDLINE | ID: mdl-29729940

ABSTRACT

BACKGROUND: Hereditary angioedema (HAE) caused by C1-inhibitor deficiency is a lifelong illness characterized by recurrent acute attacks of localized skin or mucosal edema. Activation of the kallikrein/bradykinin pathway at the endothelial cell level has a relevant pathogenetic role in acute HAE attacks. Moreover, other pathways are involved given the variable clinical expression of the disease in different patients. OBJECTIVE: We sought to explore the involvement of other putative genes in edema formation. METHODS: We performed a PBMC microarray gene expression analysis on RNA isolated from patients with HAE during an acute attack and compared them with the transcriptomic profile of the same patients in the remission phase. RESULTS: Gene expression analysis identified 23 genes significantly modulated during acute attacks that are involved primarily in the natural killer cell signaling and leukocyte extravasation signaling pathways. Gene set enrichment analysis showed a significant activation of relevant biological processes, such as response to external stimuli and protein processing (q < 0.05), suggesting involvement of PBMCs during acute HAE attacks. Upregulation of 2 genes, those encoding adrenomedullin and cellular receptor for urokinase plasminogen activator (uPAR), which occurs during an acute attack, was confirmed in PBMCs of 20 additional patients with HAE by using real-time PCR. Finally, in vitro studies demonstrated the involvement of uPAR in the generation of bradykinin and endothelial leakage. CONCLUSIONS: Our study demonstrates the increase in levels of adrenomedullin and uPAR in PBMCs during an acute HAE attack. Activation of these genes usually involved in regulation of vascular tone and in inflammatory response might have a pathogenic role by amplifying bradykinin production and edema formation in patients with HAE.


Subject(s)
Adrenomedullin/genetics , Angioedemas, Hereditary/genetics , Urokinase-Type Plasminogen Activator/genetics , Acute Disease , Adolescent , Adult , Aged , Cells, Cultured , Female , Human Umbilical Vein Endothelial Cells , Humans , Jurkat Cells , Leukocytes, Mononuclear/immunology , Male , Middle Aged , Transcriptome
3.
Int J Mol Sci ; 20(7)2019 Mar 31.
Article in English | MEDLINE | ID: mdl-30935090

ABSTRACT

BACKGROUND: In pregnancy, excessive inflammation and break down of immunologic tolerance can contribute to miscarriage. Endothelial cells (ECs) are able to orchestrate the inflammatory processes by secreting pro-inflammatory mediators and bactericidal factors by modulating leakiness and leukocyte trafficking, via the expression of adhesion molecules and chemokines. The aim of this study was to analyse the differences in the phenotype between microvascular ECs isolated from decidua (DECs) and ECs isolated from human skin (ADMECs). METHODS: DECs and ADMECs were characterized for their basal expression of angiogenic factors and adhesion molecules. A range of immunological responses was evaluated, such as vessel leakage, reactive oxygen species (ROS) production in response to TNF-α stimulation, adhesion molecules expression and leukocyte migration in response to TNF-α and IFN-γ stimulation. RESULTS: DECs produced higher levels of HGF, VEGF-A and IGFBP3 compared to ADMECs. DECs expressed adhesion molecules, ICAM-2 and ICAM-3, and a mild response to TNF-α was observed. Finally, DECs produced high levels of CXCL9/MIG and CXCL10/IP-10 in response to IFN-γ and selectively recruited Treg lymphocytes. CONCLUSION: DEC phenotype differs considerably from that of ADMECs, suggesting that DECs may play an active role in the control of immune response and angiogenesis at the foetal-maternal interface.


Subject(s)
Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/immunology , Skin/immunology , Skin/metabolism , Antigens, CD/genetics , Antigens, CD/metabolism , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/metabolism , Cells, Cultured , Chemokine CXCL10/genetics , Chemokine CXCL10/metabolism , Chemokine CXCL9/genetics , Chemokine CXCL9/metabolism , Decidua , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Female , Fluorescent Antibody Technique , Humans , In Vitro Techniques , Inflammation/genetics , Inflammation/immunology , Inflammation/metabolism , Intercellular Adhesion Molecule-3/genetics , Intercellular Adhesion Molecule-3/metabolism , Interferon-gamma/pharmacology , Neovascularization, Pathologic/metabolism , Pregnancy , Reverse Transcriptase Polymerase Chain Reaction , Skin/drug effects , Tumor Necrosis Factor-alpha/pharmacology
4.
Clin Sci (Lond) ; 132(1): 69-83, 2018 01 16.
Article in English | MEDLINE | ID: mdl-29167318

ABSTRACT

Recent studies suggest that a circulating protein called TRAIL (TNF-related apoptosis inducing ligand) may have an important role in the treatment of type 2 diabetes. It has been shown that TRAIL deficiency worsens diabetes and that TRAIL delivery, when it is given before disease onset, slows down its development. The present study aimed at evaluating whether TRAIL had the potential not only to prevent, but also to treat type 2 diabetes. Thirty male C57BL/6J mice were randomized to a standard or a high-fat diet (HFD). After 4 weeks of HFD, mice were further randomized to receive either placebo or TRAIL, which was delivered weekly for 8 weeks. Body weight, food intake, fasting glucose, and insulin were measured at baseline and every 4 weeks. Tolerance tests were performed before drug randomization and at the end of the study. Tissues were collected for further analyses. Parallel in vitro studies were conducted on HepG2 cells and mouse primary hepatocytes. TRAIL significantly reduced body weight, adipocyte hypertrophy, free fatty acid levels, and inflammation. Moreover, it significantly improved impaired glucose tolerance, and ameliorated non-alcoholic fatty liver disease (NAFLD). TRAIL treatment reduced liver fat content by 47% in vivo as well as by 45% in HepG2 cells and by 39% in primary hepatocytes. This was associated with a significant increase in liver peroxisome proliferator-activated receptor (PPAR) γ (PPARγ) co-activator-1 α (PGC-1α) expression both in vivo and in vitro, pointing to a direct protective effect of TRAIL on the liver. The present study confirms the ability of TRAIL to significantly attenuate diet-induced metabolic abnormalities, and it shows for the first time that TRAIL is effective also when administered after disease onset. In addition, our data shed light on TRAIL therapeutic potential not only against impaired glucose tolerance, but also against NAFLD.


Subject(s)
Diet, High-Fat/adverse effects , Glucose Intolerance/prevention & control , Non-alcoholic Fatty Liver Disease/prevention & control , TNF-Related Apoptosis-Inducing Ligand/pharmacology , Animals , Body Weight/drug effects , Cells, Cultured , Gene Expression/drug effects , Hep G2 Cells , Hepatocytes/drug effects , Hepatocytes/metabolism , Humans , Liver/drug effects , Liver/metabolism , Male , Mice, Inbred C57BL , Non-alcoholic Fatty Liver Disease/etiology , PPAR gamma/genetics , PPAR gamma/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/genetics , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Random Allocation , TNF-Related Apoptosis-Inducing Ligand/administration & dosage , TNF-Related Apoptosis-Inducing Ligand/pharmacokinetics
5.
BMC Nephrol ; 18(1): 219, 2017 Jul 06.
Article in English | MEDLINE | ID: mdl-28683789

ABSTRACT

BACKGROUND: Osteoprotegerin (OPG) is a glycoprotein that plays an important regulatory role in the skeletal, vascular, and immune system. It has been shown that OPG predicts chronic kidney disease (CKD) in diabetic patients. We hypothesized that OPG could be a risk marker of CKD development also in non-diabetic hypertensive patients. METHODS: A case-control study was carried out to measure circulating OPG levels in 42 hypertensive patients with CKD and in 141 hypertensive patients without CKD. A potential relationship between OPG and the presence of CKD was investigated and a receiver-operating characteristic (ROC) curve was designed thereafter to identify a cut-off value of OPG that best explained the presence of CKD. Secondly, to evaluate whether OPG increase could affect the kidney, 18 C57BL/6J mice were randomized to be treated with saline or recombinant OPG every 3 weeks for 12 weeks. RESULTS: Circulating OPG levels were significantly higher in hypertensive patients with CKD, and there was a significant inverse association between OPG and renal function, that was independent from other variables. ROC analysis showed that OPG levels had a high statistically predictive value on CKD in hypertensive patients, which was greater than that of hypertension. The OPG best cut-off value associated with CKD was 1109.19 ng/L. In the experimental study, OPG delivery significantly increased the gene expression of pro-inflammatory and pro-fibrotic mediators, as well as the glomerular nitrosylation of proteins. CONCLUSIONS: This study shows that OPG is associated with CKD in hypertensive patients, where it might have a higher predictive value than that of hypertension for CKD development. Secondly, we found that OPG delivery significantly increased the expression of molecular pathways involved in kidney damage. Further longitudinal studies are needed not only to evaluate whether OPG predicts CKD development but also to clarify whether OPG should be considered a risk factor for CKD.


Subject(s)
Hypertension/blood , Hypertension/diagnosis , Osteoprotegerin/blood , Renal Insufficiency, Chronic/blood , Renal Insufficiency, Chronic/diagnosis , Aged , Animals , Biomarkers/blood , Case-Control Studies , Female , Follow-Up Studies , Humans , Hypertension/epidemiology , Male , Mice , Mice, Inbred C57BL , Middle Aged , Random Allocation , Renal Insufficiency, Chronic/epidemiology
6.
Proc Natl Acad Sci U S A ; 111(11): 4209-14, 2014 Mar 18.
Article in English | MEDLINE | ID: mdl-24591625

ABSTRACT

We have previously shown that C1q is expressed on endothelial cells (ECs) of newly formed decidual tissue. Here we demonstrate that C1q is deposited in wound-healing skin in the absence of C4 and C3 and that C1q mRNA is locally expressed as revealed by real-time PCR and in situ hybridization. C1q was found to induce permeability of the EC monolayer, to stimulate EC proliferation and migration, and to promote tube formation and sprouting of new vessels in a rat aortic ring assay. Using a murine model of wound healing we observed that vessel formation was defective in C1qa(-/-) mice and was restored to normal after local application of C1q. The mean vessel density of wound-healing tissue and the healed wound area were significantly increased in C1q-treated rats. On the basis of these results we suggest that C1q may represent a valuable therapeutic agent that can be used to treat chronic ulcers or other pathological conditions in which angiogenesis is impaired, such as myocardial ischemia.


Subject(s)
Complement C1q/physiology , Endothelial Cells/drug effects , Neovascularization, Physiologic/genetics , Wound Healing/genetics , Animals , Cell Proliferation/drug effects , Complement C1q/genetics , Complement C1q/pharmacology , DNA Primers/genetics , Endothelial Cells/physiology , Enzyme-Linked Immunosorbent Assay , Human Umbilical Vein Endothelial Cells , Humans , Immunoblotting , Immunohistochemistry , In Situ Hybridization , Mice , Mice, Inbred C57BL , Mice, Knockout , Neovascularization, Physiologic/physiology , Rats , Rats, Wistar , Real-Time Polymerase Chain Reaction , Wound Healing/physiology
7.
Int Arch Allergy Immunol ; 169(2): 130-4, 2016.
Article in English | MEDLINE | ID: mdl-27073906

ABSTRACT

BACKGROUND: Immunoglobulin (Ig) A deficiency is a primary immunodeficiency in which autoimmunity is frequently observed. Thirty to fifty percent of patients with spontaneous chronic urticaria have autoantibodies that are able to cross-link FcεRI on mast cells and basophils. METHODS: We investigated whether spontaneous chronic urticaria in patients with IgA deficiency meets the criteria for autoimmunity. Four patients were screened for positivity to a skin prick test and an autologous serum skin test and for the presence of other autoimmune diseases. Patient sera were tested for the ability to activate basophils and mast cells in vitro by measuring surface CD63 expression and ß-hexosaminidase release, respectively. RESULTS: The autologous serum test was positive in all patients, and patient sera were found to induce CD63 upregulation on basophils and degranulation of an LAD2 mast cell line. Moreover, all patients were affected by other autoimmune disorders. CONCLUSION: For the first time, these data point out chronic autoimmune urticaria in subjects with an IgA deficiency and confirm that different autoimmune disorders are common among patients with an IgA deficiency. Patients with chronic autoimmune spontaneous urticaria should be screened for IgA deficiency, especially if they are affected by other autoimmune disorders. Thus, spontaneous urticaria could mirror more complex systemic diseases, such as immune deficiency.


Subject(s)
Autoantibodies/immunology , Autoimmune Diseases/complications , Autoimmune Diseases/immunology , IgA Deficiency/complications , IgA Deficiency/immunology , Urticaria/complications , Urticaria/immunology , Adolescent , Adult , Autoimmune Diseases/metabolism , Basophils/immunology , Basophils/metabolism , Cell Degranulation/immunology , Female , Humans , IgA Deficiency/metabolism , Immunoglobulin E/blood , Immunoglobulin E/immunology , Male , Mast Cells/immunology , Mast Cells/metabolism , Middle Aged , Receptors, IgE/metabolism , Urticaria/metabolism , Young Adult
8.
Mediators Inflamm ; 2016: 6529728, 2016.
Article in English | MEDLINE | ID: mdl-28070143

ABSTRACT

Background. Dyslipidemia and diabetes are two of the most well established risk factors for the development of cardiovascular disease (CVD). Both of them usually activate a complex range of pathogenic pathways leading to organ damage. Here we hypothesized that dyslipidemia and diabetes could affect osteoprotegerin (OPG) and TNF-related apoptosis-inducing ligand (TRAIL) expression in the vessels and the heart. Materials and Methods. Gene and protein expression of OPG, TRAIL, and OPG/TRAIL ratio were quantified in the aorta and the hearts of control mice, dyslipidemic mice, and diabetic mice. Results. Diabetes significantly increased OPG and the OPG/TRAIL ratio expression in the aorta, while dyslipidemia was the major determinant of the changes observed in the heart, where it significantly increased OPG and reduced TRAIL expression, thus increasing cardiac OPG/TRAIL ratio. Conclusions. This work shows that both dyslipidemia and diabetes affect OPG/TRAIL ratio in the cardiovascular system. This could contribute to the changes in circulating OPG/TRAIL which are observed in patients with diabetes and CVD. Most importantly, these changes could mediate/contribute to atherosclerosis development and cardiac remodeling.


Subject(s)
Cardiovascular System/metabolism , Diabetes Mellitus, Experimental/metabolism , Dyslipidemias/metabolism , Osteoprotegerin/metabolism , TNF-Related Apoptosis-Inducing Ligand/metabolism , Animals , Aorta/metabolism , Apolipoproteins E/genetics , Apoptosis , Atherosclerosis/metabolism , Disease Models, Animal , Gene Expression Profiling , Male , Mice , Mice, Inbred C57BL
9.
Cell Microbiol ; 16(6): 925-37, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24397470

ABSTRACT

Neisseria meningitidis is a human pathogen that can cause fatal sepsis and meningitis once it reaches the blood stream and the nervous system. Here we demonstrate that a fragment, released upon proteolysis of the surface-exposed protein Neisserial Heparin Binding Antigen (NHBA), by the bacterial protease NalP, alters the endothelial permeability by inducing the internalization of the adherens junction protein VE-cadherin. We found that C2 rapidly accumulates in mitochondria where it induces the production of reactive oxygen species: the latter are required for the phosphorylation of the junctional protein and for its internalization that, in turn, is responsible for the endothelial leakage. Our data support the notion that the NHBA-derived fragment C2 might contribute to the extensive vascular leakage typically associated with meningococcal sepsis.


Subject(s)
Adherens Junctions/drug effects , Antigens, Bacterial/metabolism , Endothelial Cells/drug effects , Neisseria meningitidis/physiology , Permeability/drug effects , Antigens, Bacterial/genetics , Cell Line , Humans , Virulence Factors/genetics , Virulence Factors/metabolism
10.
Arthritis Rheum ; 65(5): 1232-42, 2013 May.
Article in English | MEDLINE | ID: mdl-23371320

ABSTRACT

OBJECTIVE: Lyme arthritis (LA) is characterized by infiltration of inflammatory cells, mainly neutrophils (polymorphonuclear cells [PMNs]) and T cells, into the joints. This study was undertaken to evaluate the role of the neutrophil-activating protein A (NapA) of Borrelia burgdorferi in eliciting inflammation and in driving the adaptive immune response. METHODS: Levels of NapA, interferon-γ (IFNγ), interleukin-17 (IL-17), and T cell-attracting chemokines were assessed by enzyme-linked immunosorbent assay in synovial fluid from patients with LA. The profile of T cells recruited into the synovia of patients with LA was defined by fluorescence-activated cell sorting analysis. NapA was intraarticularly injected into rat knees, and the cells recruited in synovia were characterized. The role of NapA in recruiting immune cells was confirmed by chemotaxis assays using a Transwell system. RESULTS: NapA, IFNγ, IL-17, CCL2, CCL20, and CXCL10 accumulated in synovial fluid from patients with LA. Accordingly, T cells obtained from these patients produced IFNγ or IL-17, but notably, some produced both cytokines. NapA promoted neutrophil and T lymphocyte recruitment both in vitro and in vivo. Interestingly, the infiltration of T cells not only resulted from the chemotactic activity of NapA but also relied on the chemokines produced by PMNs exposed to NapA. CONCLUSION: We provide evidence that NapA functions as one of the main bacterial products involved in the pathogenesis of LA. Accordingly, we show that, at very early stages of LA, NapA accumulates and, in turn, orchestrates the recruitment of inflammatory cells into the joint cavity. Thereafter, with the contribution of recruited cells, NapA promotes the infiltration of T cells producing IL-17 and/or IFNγ.


Subject(s)
Adaptive Immunity/immunology , Arthritis, Infectious/immunology , Bacterial Proteins/immunology , Chemokines, CXC/immunology , Lyme Disease/immunology , Animals , Arthritis, Infectious/etiology , Arthritis, Infectious/pathology , Arthritis, Rheumatoid/immunology , Arthritis, Rheumatoid/pathology , Bacterial Proteins/administration & dosage , Bacterial Proteins/metabolism , Borrelia burgdorferi/physiology , Chemokines/analysis , Chemokines/metabolism , Chemokines, CXC/administration & dosage , Chemokines, CXC/metabolism , Chemotaxis/drug effects , Female , Flow Cytometry , Humans , Injections, Intra-Articular , Knee Joint/metabolism , Knee Joint/pathology , Lyme Disease/complications , Lyme Disease/pathology , Male , Middle Aged , Neutrophil Infiltration , Neutrophils/immunology , Neutrophils/metabolism , Neutrophils/pathology , Rats , Stifle/drug effects , Stifle/pathology , Synovial Fluid/chemistry , Synovial Fluid/metabolism , Synovial Membrane/pathology , T-Lymphocytes/metabolism , T-Lymphocytes/pathology
11.
Int J Pharm ; 660: 124322, 2024 Jun 10.
Article in English | MEDLINE | ID: mdl-38866082

ABSTRACT

Podocytes, cells of the glomerular filtration barrier, play a crucial role in kidney diseases and are gaining attention as potential targets for new therapies. Brain-Derived Neurotrophic Factor (BDNF) has shown promising results in repairing podocyte damage, but its efficacy via parenteral administration is limited by a short half-life. Low temperature sensitive liposomes (LTSL) are a promising tool for targeted BDNF delivery, preserving its activity after encapsulation. This study aimed to improve LTSL design for efficient BDNF encapsulation and targeted release to podocytes, while maintaining stability and biological activity, and exploiting the conjugation of targeting peptides. While cyclic RGD (cRGD) was used for targeting endothelial cells in vitro, a homing peptide (HITSLLS) was conjugated for more specific uptake by glomerular endothelial cells in vivo. BDNF-loaded LTSL successfully repaired cytoskeleton damage in podocytes and reduced albumin permeability in a glomerular co-culture model. cRGD conjugation enhanced endothelial cell targeting and uptake, highlighting an improved therapeutic effect when BDNF release was induced by thermoresponsive liposomal degradation. In vivo, targeted LTSL showed evidence of accumulation in the kidneys, and their BDNF delivery decreased proteinuria and ameliorated kidney histology. These findings highlight the potential of BDNF-LTSL formulations in restoring podocyte function and treating glomerular diseases.

12.
NPJ Regen Med ; 8(1): 8, 2023 Feb 11.
Article in English | MEDLINE | ID: mdl-36774354

ABSTRACT

Nonhealing wounds place a significant burden on both quality of life of affected patients and health systems. Skin substitutes are applied to promote the closure of nonhealing wounds, although their efficacy is limited by inadequate vascularization. The stromal vascular fraction (SVF) from the adipose tissue is a promising therapy to overcome this limitation. Despite a few successful clinical trials, its incorporation in the clinical routine has been hampered by their inconsistent results. All these studies concluded by warranting pre-clinical work aimed at both characterizing the cell types composing the SVF and shedding light on their mechanism of action. Here, we established a model of nonhealing wound, in which we applied the SVF in combination with a clinical-grade skin substitute. We purified the SVF cells from transgenic animals to trace their fate after transplantation and observed that it gave rise to a mature vascular network composed of arteries, capillaries, veins, as well as lymphatics, structurally and functionally connected with the host circulation. Then we moved to a human-in-mouse model and confirmed that SVF-derived endothelial cells formed hybrid human-mouse vessels, that were stabilized by perivascular cells. Mechanistically, SVF-derived endothelial cells engrafted and expanded, directly contributing to the formation of new vessels, while a population of fibro-adipogenic progenitors stimulated the expansion of the host vasculature in a paracrine manner. These data have important clinical implications, as they provide a steppingstone toward the reproducible and effective adoption of the SVF as a standard care for nonhealing wounds.

13.
J Immunol ; 185(7): 4420-9, 2010 Oct 01.
Article in English | MEDLINE | ID: mdl-20810993

ABSTRACT

Fetal trophoblast cells invading the decidua in the early phase of pregnancy establish complex interaction with the maternal extracellular matrix. We discovered that C1q was widely distributed in human decidual stroma in the absence of C4 and C3 and was actively synthesized by migrating extravillous trophoblasts. The cells expressed the messages for the three chains of C1q and secreted this complement component that interacted with the proteins of the decidual extracellular matrix. Solid phase-bound C1q promoted trophoblast adhesion and migration, and cell binding to C1q resulted in activation of ERK1/2 MAPKs. Ab inhibition experiments showed that the receptors for the globular head of C1q/p33 and α(4)ß(1) integrin were both involved in this process and were colocalized on the cell surface following binding of C1q to trophoblasts. We also found that C1q(-/-) mice manifested increased frequency of fetal resorption, reduced fetal weight, and smaller litter sizes compared with wild-type mice. C1q deficiency was associated with impaired labyrinth development and decidual vessel remodeling. Collectively, these data suggest that C1q plays an important role in promoting trophoblast invasion of decidua and that defective local production of C1q may be involved in pregnancy disorders, such as pre-eclampsia, characterized by poor trophoblast invasion.


Subject(s)
Chemotaxis, Leukocyte/physiology , Complement C1q/metabolism , Placentation/immunology , Trophoblasts/metabolism , Animals , Cell Adhesion/immunology , Complement C1q/immunology , Female , Humans , Immunoblotting , Immunohistochemistry , Immunoprecipitation , Mice , Mice, Inbred C57BL , Microscopy, Confocal , Pre-Eclampsia/immunology , Pregnancy , Reverse Transcriptase Polymerase Chain Reaction , Trophoblasts/immunology
14.
Clin Dev Immunol ; 2012: 484321, 2012.
Article in English | MEDLINE | ID: mdl-22203857

ABSTRACT

The spiral arteries undergo physiologic changes during pregnancy, and the failure of this process may lead to a spectrum of pregnancy disorders, including pre-eclampsia. Our recent data indicate that decidual endothelial cells (DECs), covering the inner side of the spiral arteries, acquire the ability to synthesize C1q, which acts as a link between endovascular trophoblast and DECs favouring the process of vascular remodelling. In this study, we have shown that sera obtained from pre-eclamptic patients strongly inhibit the interaction between extravillous trophoblast (EVT) and DECs, preventing endovascular invasion of trophoblast cells. We further demonstrated that mannose-binding lectin (MBL), one of the factor increased in pre-eclamptic patient sera, strongly inhibits the interaction of EVT with C1q interfering with the process of EVT adhesion to and migration through DECs. These data suggest that the increased level of MBL in pre-eclampsia may contribute to the failure of the endovascular invasion of trophoblast cells.


Subject(s)
Endothelial Cells/metabolism , Mannose-Binding Lectin/metabolism , Pre-Eclampsia/metabolism , Trophoblasts/physiology , Cell Communication/physiology , Decidua/cytology , Endothelial Cells/cytology , Female , Humans , Mannose-Binding Lectin/blood , Pregnancy , Transendothelial and Transepithelial Migration
15.
Blood ; 113(15): 3640-8, 2009 Apr 09.
Article in English | MEDLINE | ID: mdl-19179470

ABSTRACT

We describe a novel localization of C7 as a membrane-bound molecule on endothelial cells (ECs). Data obtained by sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE), Western blot analysis, Northern blot analysis, and mass spectrometry revealed that membrane-associated C7 (mC7) was indistinguishable from soluble C7 and was associated with vimentin on the cell surface. mC7 interacted with the other late complement components to form membrane-bound TCC (mTCC). Unlike the soluble SC5b-9, mTCC failed to stimulate ECs to express adhesion molecules, to secrete IL-8, and to induce albumin leakage through a monolayer of ECs, and more importantly protected ECs from the proinflammatory effect of SC5b-9. Our data disclose the possibility of a novel role of mC7 that acts as a trap for the late complement components to control excessive inflammation induced by SC5b-9.


Subject(s)
Complement C7/immunology , Complement C7/metabolism , Complement Membrane Attack Complex/metabolism , Endothelial Cells/immunology , Vasculitis/immunology , Vasculitis/metabolism , Cells, Cultured , Complement C7/genetics , Complement Membrane Attack Complex/immunology , Endothelial Cells/cytology , Endothelial Cells/metabolism , Humans , Interleukin-8/immunology , Interleukin-8/metabolism , Membrane Proteins/immunology , Membrane Proteins/metabolism , Proteomics , RNA, Messenger/metabolism , Umbilical Veins/cytology , Vimentin/metabolism
16.
J Allergy Clin Immunol ; 124(6): 1303-10.e4, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19796797

ABSTRACT

BACKGROUND: Activation of bradykinin-mediated B2 receptor has been shown to play an important role in the onset of angioedema associated with C1 inhibitor deficiency. This finding has led to the development of novel therapeutic drugs such as the B2 receptor antagonist icatibant. However, it is unclear whether other receptors expressed on endothelial cells contribute to the release of kinins and vascular leakage in these patients. The recognition of their role may have obvious therapeutic implications. OBJECTIVE: Our aim was to investigate the involvement of B1 and gC1q receptors in in vitro and in vivo models of vascular leakage induced by plasma samples obtained from patients with C1 inhibitor deficiency. METHODS: The vascular leakage was evaluated in vitro on endothelial cells by a transwell model system and in vivo on rat mesentery microvessels by intravital microscopy. RESULTS: We observed that the attack phase plasma from C1 inhibitor-deficient patients caused a delayed fluorescein-labeled albumin leakage as opposed to the rapid effect of bradykinin, whereas remission plasma elicited a modest effect compared with control plasma. The plasma permeabilizing effect was prevented by blocking the gC1q receptor-high-molecular-weight kininogen interaction, was partially inhibited by B2 receptor or B1 receptor antagonists, and was totally prevented by the mixture of the 2 antagonists. Involvement of B1 receptor was supported by the finding that albumin leakage caused by attack phase plasma was enhanced by IL-1beta and was markedly reduced by brefeldin A. CONCLUSION: Our data suggest that both B1 receptor and gC1q receptor are involved in the vascular leakage induced by hereditary and acquired angioedema plasma.


Subject(s)
Angioedema/physiopathology , Capillary Permeability/drug effects , Complement C1 Inhibitor Protein/metabolism , Membrane Glycoproteins/metabolism , Receptor, Bradykinin B1/metabolism , Receptors, Complement/metabolism , Adrenergic beta-Antagonists/pharmacology , Adrenergic beta-Antagonists/therapeutic use , Angioedema/drug therapy , Animals , Antibodies, Monoclonal/pharmacology , Blood Vessels/drug effects , Blood Vessels/immunology , Blood Vessels/metabolism , Bradykinin/analogs & derivatives , Bradykinin/pharmacology , Bradykinin/therapeutic use , Bradykinin B1 Receptor Antagonists , Brefeldin A/pharmacology , Capillary Permeability/physiology , Cell Line, Tumor , Complement C1 Inhibitor Protein/genetics , Hereditary Angioedema Types I and II/drug therapy , Hereditary Angioedema Types I and II/physiopathology , Humans , Immunologic Factors/pharmacology , Interleukin-1beta/pharmacology , Male , Protein Synthesis Inhibitors/pharmacology , Rats , Rats, Inbred WKY
17.
Biomed Pharmacother ; 131: 110752, 2020 Nov.
Article in English | MEDLINE | ID: mdl-33152918

ABSTRACT

Gastroesophageal reflux disease (GERD) is a common digestive disorder that causes esophagitis and injuries to the esophageal mucosa. GERD symptoms are recurrent during pregnancy and their treatment is focused on lifestyle changes and nonprescription medicines. The aim of this study was to characterize the mechanism of action of a new patented medical device, an oral formulation containing hyaluronic acid, rice extract, and amino acids dispersed in a bioadhesive polymer matrix, by assessing its protective effects in in vitro and ex vivo models of esophageal mucosa damage. Acidic bile salts and pepsin cocktail (BSC) added to CP-A and COLO-680 N esophagus cells were used as an in vitro GERD model to evaluate the binding capacities, anti-inflammatory effects and reparative properties of the investigational product (IP) in comparison to a viscous control. Our results showed that the IP prevents cell permeability and tight junction dysfunction induced by BSC. Furthermore, the IP was also able to down-regulate IL-6 and IL-8 mRNA expression induced by BSC stimulation and to promote tissue repair and wound healing. The results were confirmed by ex vivo experiments in excised rat esophagi through the quantification of Evans Blue permeability assay. These experiments provided evidence that the IP is able to bind to the human esophagus cells, preventing the damage caused by gastroesophageal reflux, showing potential anti-irritative, soothing, and reparative properties.


Subject(s)
Amino Acids/administration & dosage , Esophageal Mucosa/drug effects , Gastroesophageal Reflux/drug therapy , Hyaluronic Acid/administration & dosage , Oryza , Plant Extracts/administration & dosage , Regeneration/drug effects , Adhesiveness , Amino Acids/chemistry , Cell Line, Tumor , Equipment and Supplies , Esophageal Mucosa/physiology , Humans , Hyaluronic Acid/chemistry , Permeability , Plant Extracts/chemistry , Regeneration/physiology
18.
Nat Commun ; 11(1): 3945, 2020 08 07.
Article in English | MEDLINE | ID: mdl-32770028

ABSTRACT

TP53 missense mutations leading to the expression of mutant p53 oncoproteins are frequent driver events during tumorigenesis. p53 mutants promote tumor growth, metastasis and chemoresistance by affecting fundamental cellular pathways and functions. Here, we demonstrate that p53 mutants modify structure and function of the Golgi apparatus, culminating in the increased release of a pro-malignant secretome by tumor cells and primary fibroblasts from patients with Li-Fraumeni cancer predisposition syndrome. Mechanistically, interacting with the hypoxia responsive factor HIF1α, mutant p53 induces the expression of miR-30d, which in turn causes tubulo-vesiculation of the Golgi apparatus, leading to enhanced vesicular trafficking and secretion. The mut-p53/HIF1α/miR-30d axis potentiates the release of soluble factors and the deposition and remodeling of the ECM, affecting mechano-signaling and stromal cells activation within the tumor microenvironment, thereby enhancing tumor growth and metastatic colonization.


Subject(s)
Breast Neoplasms/genetics , Cell Transformation, Neoplastic/genetics , Golgi Apparatus/pathology , Li-Fraumeni Syndrome/genetics , MicroRNAs/metabolism , Tumor Suppressor Protein p53/genetics , Animals , Biopsy , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Transformation, Neoplastic/metabolism , Female , Fibroblasts , Gene Expression Regulation, Neoplastic , Golgi Apparatus/metabolism , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Li-Fraumeni Syndrome/pathology , Mice , Microtubules/metabolism , Microtubules/pathology , Mutation , Primary Cell Culture , Secretory Vesicles/metabolism , Secretory Vesicles/pathology , Signal Transduction/genetics , Skin/cytology , Skin/pathology , Tumor Microenvironment/genetics , Xenograft Model Antitumor Assays
19.
Biochem Biophys Res Commun ; 386(2): 338-44, 2009 Aug 21.
Article in English | MEDLINE | ID: mdl-19523446

ABSTRACT

Since an increased serum unconjugated bilirubin (UCB) level has been proposed as an independent protective factor against atherosclerotic disease, we investigated the molecular events at the basis of this effect. HUVEC and H5V cells were treated with TNFalpha and UCB and the effects assessed on E-selectin, VCAM-1 and ICAM-1. In HUVEC cells, UCB blunted the TNFalpha-induced gene upregulation of E-selectin VCAM-1 and ICAM-1. The same pattern was observed in H5V cells except for ICAM-1. UCB also inhibited the PMN endothelial adhesion in HUVEC H5V cells. Western blot and FACS analysis confirmed that UCB prevented TNFalpha-induced over-expression of adhesion molecules proteins in H5V cells. These data contribute to further explain the protective effect of bilirubin against development of atherosclerosis.


Subject(s)
Bilirubin/metabolism , E-Selectin/biosynthesis , Intercellular Adhesion Molecule-1/biosynthesis , Neutrophils/physiology , Vascular Cell Adhesion Molecule-1/biosynthesis , Bilirubin/pharmacology , Cell Adhesion/drug effects , Cell Line , E-Selectin/genetics , Humans , Intercellular Adhesion Molecule-1/genetics , Neutrophils/drug effects , Neutrophils/metabolism , Tumor Necrosis Factor-alpha/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Umbilical Cord/cytology , Umbilical Cord/drug effects , Umbilical Cord/metabolism , Up-Regulation/drug effects , Vascular Cell Adhesion Molecule-1/genetics
20.
Rheumatology (Oxford) ; 48(3): 293-8, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19168833

ABSTRACT

OBJECTIVE: Complement activation products contribute to a large number of inflammatory diseases, including RA. We have investigated whether osteoprotegerin (OPG) may concur with the soluble terminal complement complex (SC5b-9) to the inflammatory cascade characterizing RA. METHODS: Levels of SC5b-9 and OPG in the plasma and SF of patients with active RA were determined by ELISA. The presence of SC5b-9 and OPG in RA synovial lesions was analysed by immunohistochemistry. Cultured endothelial cells were used for in vitro leucocyte/endothelial cell adhesion assays. In addition, endothelial cells were exposed to SC5b-9 in order to evaluate the effects on the production of OPG protein, as well as the activation of the OPG promoter. RESULTS: Patients affected by active RA are characterized by elevated levels of both SC5b-9 and OPG in plasma and/or SF. Of note, we have observed a co-localization of SC5b-9 and OPG in endothelial cells of post-capillary venules of RA synovial lesions. Data on endothelial cell cultures showed that exposure to SC5b-9 induced the up-regulation of OPG expression/release, stimulating the transcriptional activity of the OPG promoter, and synergized with TNF-alpha in up-regulating OPG production. CONCLUSIONS: Our findings demonstrate that SC5b-9 induces OPG production by endothelial cells and we propose that the SC5b-9-mediated up-regulation of OPG may be an important mechanism whereby complement contributes in promoting and/or enhancing the inflammation in RA.


Subject(s)
Arthritis, Rheumatoid/metabolism , Complement Membrane Attack Complex/physiology , Endothelial Cells/metabolism , Osteoprotegerin/biosynthesis , Adult , Aged , Cell Adhesion/physiology , Cells, Cultured , Complement Membrane Attack Complex/pharmacology , Dose-Response Relationship, Drug , Endothelial Cells/drug effects , Endothelium, Vascular/metabolism , Female , Humans , Male , Middle Aged , Neutrophil Infiltration/physiology , Neutrophils/physiology , Synovial Membrane/metabolism , Up-Regulation/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL