406 results on '"Bulla R"'
Search Results
202. Combined extracts of Echinacea angustifolia DC. and Zingiber officinale Roscoe in softgel capsules: Pharmacokinetics and immunomodulatory effects assessed by gene expression profiling.
- Author
-
Dall'Acqua S, Grabnar I, Verardo R, Klaric E, Marchionni L, Luidy-Imada E, Sut S, Agostinis C, Bulla R, Perissutti B, and Voinovich D
- Subjects
- Administration, Oral, Anti-Inflammatory Agents pharmacokinetics, Capsules, Catechols blood, Fatty Alcohols blood, Female, Gene Expression Profiling, Humans, Leukocytes, Mononuclear drug effects, Male, Plant Extracts administration & dosage, Echinacea chemistry, Zingiber officinale chemistry, Immunologic Factors pharmacokinetics, Plant Extracts immunology, Plant Extracts pharmacokinetics
- Abstract
Background and Objective: Echinacea angustifolia DC. and Zingiber officinale Roscoe are two natural products with documented immunomodulatory activity, both able to modulate the expression of important immune-related genes. Thus, their use in combination seems to be particularly promising. In this context, we have considered the oral supplementation of a highly standardized lipophilic extract combining both above-mentioned phytocomplexes, formulated in attractive softgel capsules, with two objectives: on the one hand to study oral pharmacokinetic of main active extracts' components and on the other hand to examine the immunomodulation and anti-inflammatory properties by gene expression profiling., Methods: Softgel capsules containing a combination of E. angustifolia DC. and Z. officinale Roscoe (5 mg and 25 mg, respectively) were given by oral administration to 10 healthy volunteers. The plasma concentrations of dodeca-2E,4E,8Z,10E/Z-tetraenoic isobutylamide (tetraene) for E. angustifolia DC., 6-gingerol and 6-shogaol (free and glucuronide) for Z. officinale Roscoe were determined by LC-MS analysis, and the pharmacokinetic analysis was performed. To understand the functional mechanisms responsible for the documented health benefits, we also examined the overall transcriptional remodeling induced in the peripheral blood mononuclear cells and performed an integrative functional analysis on the generated gene expression., Results: All bioactive components were absorbed very rapidly, and their t
max were detected in plasma from 30 min to 1.40 h. The peak concentrations of tetraene, 6-gingerol, 6-shogaol and their glucuronide metabolites were 14.74, 5.66, 9.25, 29.2 and 22.24 ng/ml, respectively. Integrated analysis performed on the generated gene expression data highlighted immunomodulatory and anti-inflammatory effects similar to those exerted by hydrocortisone., Conclusion: These data demonstrated that the bioactive ingredients are highly and rapidly absorbed from softgel capsules containing the combination of the above-mentioned lipophilic extracts, providing evidence to support their immunomodulatory and anti-inflammatory properties. These data also help in defining the mechanistic pathways underlying the health benefits of these plant-derived bioactive compounds., (Copyright © 2019 Elsevier GmbH. All rights reserved.)- Published
- 2019
- Full Text
- View/download PDF
203. Uterine Immunity and Microbiota: A Shifting Paradigm.
- Author
-
Agostinis C, Mangogna A, Bossi F, Ricci G, Kishore U, and Bulla R
- Subjects
- Adaptive Immunity, Endometrium immunology, Endometrium metabolism, Endometrium microbiology, Female, Host-Pathogen Interactions, Humans, Immunity, Innate, Leukocytes immunology, Leukocytes metabolism, Lymphocytes immunology, Lymphocytes metabolism, Mucus, Pregnancy, Semen, Uterus anatomy & histology, Immunity, Microbiota, Uterus immunology, Uterus microbiology
- Abstract
The female reproductive tract harbors distinct microbial communities, as in the vagina, cervical canal, uterus, and fallopian tubes. The nature of the vaginal microbiota is well-known; in contrast, the upper reproductive tract remains largely unexplored. Alteration in the uterine microbiota, which is dependent on the nutrients and hormones available to the uterus, is likely to play an important role in uterine-related diseases such as hysteromyoma, adenomyosis, and endometriosis. Uterine mucosa is an important tissue barrier whose main function is to offer protection against pathogens and other toxic factors, while maintaining a symbiotic relationship with commensal microbes. These characteristics are shared by all the mucosal tissues; however, the uterine mucosa is unique since it changes cyclically during the menstrual cycle as well as pregnancy. The immune system, besides its role in the defense process, plays crucial roles in reproduction as it ensures local immune tolerance to fetal/paternal antigens, trophoblast invasion, and vascular remodeling. The human endometrium contains a conspicuous number of immune cells, mainly Natural Killers (NK) cells, which are phenotypically distinct from peripheral cytotoxic NK, cells and macrophages. The endometrium also contains few lymphoid aggregates comprising B cell and CD8
+ T cells. The number and the phenotype of these cells change during the menstrual cycle. It has become evident in recent years that the immune cell phenotype and function can be influenced by microbiota. Immune cells can sense the presence of microbes through their pattern recognition receptors, setting up host-microbe interaction. The microbiota exerts an appropriately controlled defense mechanism by competing for nutrients and mucosal space with pathogens. It has recently been considered that uterus is a non-sterile compartment since it seems to possess its own microbiota. There has been an increasing interest in characterizing the nature of microbial colonization within the uterus and its apparent impact on fertility and pregnancy. This review will examine the potential relationship between the uterine microbiota and the immune cells present in the local environment., (Copyright © 2019 Agostinis, Mangogna, Bossi, Ricci, Kishore and Bulla.)- Published
- 2019
- Full Text
- View/download PDF
204. Prognostic Implications of the Complement Protein C1q in Gliomas.
- Author
-
Mangogna A, Belmonte B, Agostinis C, Zacchi P, Iacopino DG, Martorana A, Rodolico V, Bonazza D, Zanconati F, Kishore U, and Bulla R
- Subjects
- Biomarkers, Tumor genetics, Brain Neoplasms diagnosis, Brain Neoplasms genetics, Brain Neoplasms pathology, Complement C1q genetics, Female, Glioma diagnosis, Glioma genetics, Glioma pathology, Humans, Male, Neoplasm Proteins genetics, Prognosis, Tumor Microenvironment genetics, Biomarkers, Tumor immunology, Brain Neoplasms immunology, Complement C1q immunology, Databases, Nucleic Acid, Gene Expression Regulation, Neoplastic immunology, Glioma immunology, Neoplasm Proteins immunology, Tumor Microenvironment immunology
- Abstract
The contribution of the complement system in the pathophysiology of brain cancers has been recently considered in light of its well-known involvement in carcinogenesis. Complement system represents an important component of the inflammatory response, which acts as a functional bridge between the innate and adaptive immune response. C1q, the first recognition subcomponent of the complement classical pathway, has recently been shown to be involved in a range of pathophysiological functions that are not dependent on complement activation. C1q is expressed in the microenvironment of various types of human tumors, including melanoma, prostate, mesothelioma, and ovarian cancers, where it can exert a protective or a harmful effect on cancer progression. Despite local synthesis of C1q in the central nervous system, the involvement of C1q in glioma pathogenesis has been poorly investigated. We, therefore, performed a bioinformatics analysis, using Oncomine dataset and UALCAN database in order to assess whether the expression of the genes encoding for the three chains of C1q ( C1qA, C1qB , and C1qC ) could serve as a potential prognostic marker for gliomas. The obtained results were then validated using an independent glioma cohort from the Chinese Glioma Genome Atlas datasets. Our bioinformatics analysis, coupled with immunohistochemistry and fluorescence microscopy, appears to suggest a positive correlation between higher levels of C1q expression and unfavorable prognosis in a diverse grade of gliomas., (Copyright © 2019 Mangogna, Belmonte, Agostinis, Zacchi, Iacopino, Martorana, Rodolico, Bonazza, Zanconati, Kishore and Bulla.)
- Published
- 2019
- Full Text
- View/download PDF
205. Overview of procalcitonin in pregnancy and in pre-eclampsia.
- Author
-
Mangogna A, Agostinis C, Ricci G, Romano F, and Bulla R
- Subjects
- Cytokines metabolism, Female, Humans, Inflammation metabolism, Macrophages metabolism, Pregnancy, Pre-Eclampsia metabolism, Procalcitonin metabolism
- Abstract
Procalcitonin (PCT), a precursor for calcitonin, is a prohormone involved in the inflammatory processes, which has been poorly studied in the context of pregnancy. During severe inflammation, PCT derives from almost all cell types, including monocytes and parenchymal tissues, making it a good predictive and diagnostic marker of an inflammatory state with rapidly increased serum levels in inflammation or sepsis. In normal pregnancy, PCT is basally expressed at very low level by decidual cells, even if decidual macrophages, which in normal pregnancy are skewed to M2 macrophages, are resistant to lipopolysaccharide (LPS)-induced production of PCT. As PCT increase is associated with an inflammatory state, several research groups investigated whether PCT can be considered a marker of pre-eclampsia, a pregnancy disease characterized by systemic inflammation. The first aim of this review is to summarize what is already known about the tissues synthesizing PCT, about the stimuli that cause the increase of circulating PCT levels and how PCT acts as a proinflammatory stimulus by itself. Secondly, we will describe the role of this prohormone in normal pregnancy and in pregnancies complicated by pre-eclampsia, highlighting the involvement of the decidual macrophages and the proinflammatory cytokine tumor necrosis factor-α in the modulation of PCT expression in the decidual microenvironment., (© 2019 British Society for Immunology.)
- Published
- 2019
- Full Text
- View/download PDF
206. HMGA1 promotes breast cancer angiogenesis supporting the stability, nuclear localization and transcriptional activity of FOXM1.
- Author
-
Zanin R, Pegoraro S, Ros G, Ciani Y, Piazza S, Bossi F, Bulla R, Zennaro C, Tonon F, Lazarevic D, Stupka E, Sgarra R, and Manfioletti G
- Subjects
- Animals, Cell Line, Tumor, Culture Media, Conditioned pharmacology, Endothelial Cells cytology, Endothelial Cells drug effects, Endothelial Cells metabolism, Female, Forkhead Box Protein M1 chemistry, Forkhead Box Protein M1 genetics, Gene Expression Profiling, Gene Expression Regulation, Neoplastic, Gene Silencing, HEK293 Cells, HMGA1a Protein genetics, Humans, Prognosis, Promoter Regions, Genetic, Protein Stability, Sequence Analysis, RNA, Survival Analysis, Transcription, Genetic, Triple Negative Breast Neoplasms metabolism, Zebrafish, Cell Nucleus metabolism, Forkhead Box Protein M1 metabolism, HMGA1a Protein metabolism, Triple Negative Breast Neoplasms genetics, Vascular Endothelial Growth Factor A genetics
- Abstract
Background: Breast cancer is the most common malignancy in women worldwide. Among the breast cancer subtypes, triple-negative breast cancer (TNBC) is the most aggressive and the most difficult to treat. One of the master regulators in TNBC progression is the architectural transcription factor HMGA1. This study aimed to further explore the HMGA1 molecular network to identify molecular mechanisms involved in TNBC progression., Methods: RNA from the MDA-MB-231 cell line, silenced for HMGA1 expression, was sequenced and, with a bioinformatic analysis, molecular partners HMGA1 could cooperate with in regulating common downstream gene networks were identified. Among the putative partners, the FOXM1 transcription factor was selected. The relationship occurring between HMGA1 and FOXM1 was explored by qRT-PCR, co-immunoprecipitation and protein stability assays. Subsequently, the transcriptional activity of HMGA1 and FOXM1 was analysed by luciferase assay on the VEGFA promoter. The impact on angiogenesis was assessed in vitro, evaluating the tube formation ability of endothelial cells exposed to the conditioned medium of MDA-MB-231 cells silenced for HMGA1 and FOXM1 and in vivo injecting MDA-MB-231 cells, silenced for the two factors, in zebrafish larvae., Results: Here, we discover FOXM1 as a novel molecular partner of HMGA1 in regulating a gene network implicated in several breast cancer hallmarks. HMGA1 forms a complex with FOXM1 and stabilizes it in the nucleus, increasing its transcriptional activity on common target genes, among them, VEGFA, the main inducer of angiogenesis. Furthermore, we demonstrate that HMGA1 and FOXM1 synergistically drive breast cancer cells to promote tumor angiogenesis both in vitro in endothelial cells and in vivo in a zebrafish xenograft model. Moreover, using a dataset of breast cancer patients we show that the co-expression of HMGA1, FOXM1 and VEGFA is a negative prognostic factor of distant metastasis-free survival and relapse-free survival., Conclusions: This study reveals FOXM1 as a crucial interactor of HMGA1 and proves that their cooperative action supports breast cancer aggressiveness, by promoting tumor angiogenesis. Therefore, the possibility to target HMGA1/FOXM1 in combination should represent an attractive therapeutic option to counteract breast cancer angiogenesis.
- Published
- 2019
- Full Text
- View/download PDF
207. Evaluation of the Interplay Between the Complement Protein C1q and Hyaluronic Acid in Promoting Cell Adhesion.
- Author
-
Vidergar R, Agostinis C, Zacchi P, Mangogna A, Bossi F, Zanconati F, Confalonieri M, Ricci G, and Bulla R
- Subjects
- Humans, Cell Adhesion genetics, Complement C1q metabolism, Hyaluronic Acid metabolism, Tumor Microenvironment genetics
- Abstract
It has been increasingly demonstrated that the tumor microenvironment plays an active role in neoplasia growth and metastasis. Through different pathways, tumor cells can efficiently recruit stromal, immune and endothelial cells by secreting stimulatory factors, chemokines and cytokines. In turn, these cells can alter the signaling properties of the microenvironment by releasing growth-promoting signals, metabolites and extracellular matrix components to sustain high proliferation and metastatic competence. In this context, we identify that the complement component C1q, highly expressed locally by a range of human malignant tumors, upon interacting with the extracellular matrix hyaluronic acid, strongly affects the behavior of primary cells isolated from human tumor specimens. Here, we describe a method to test how C1q bound to hyaluronic acid (HA) impacts tumor cell adhesion, underlying the fact that the biological properties of key components of the extracellular matrix (in this case HA) can be shaped by bioactive signals toward tumor progression.
- Published
- 2019
- Full Text
- View/download PDF
208. Is the Complement Protein C1q a Pro- or Anti-tumorigenic Factor? Bioinformatics Analysis Involving Human Carcinomas.
- Author
-
Mangogna A, Agostinis C, Bonazza D, Belmonte B, Zacchi P, Zito G, Romano A, Zanconati F, Ricci G, Kishore U, and Bulla R
- Subjects
- Breast Neoplasms mortality, Breast Neoplasms pathology, Complement C1q genetics, Computational Biology, Humans, Kaplan-Meier Estimate, Kidney Neoplasms mortality, Kidney Neoplasms pathology, Lung Neoplasms mortality, Lung Neoplasms pathology, Prognosis, Breast Neoplasms immunology, Complement C1q immunology, Kidney Neoplasms immunology, Lung Neoplasms immunology
- Abstract
C1q is the first subcomponent of the classical pathway of the complement system and belongs to the C1q/Tumor Necrosis Factor superfamily. C1q can perform a diverse range of immune and non-immune functions in a complement-dependent as well as -independent manner. Being a pattern recognition molecule of the innate immunity, C1q can recognize a number of self, non-self and altered-self ligands and bring about effector mechanisms designed to clear pathogens via opsonisation and inflammatory response. C1q is locally synthesized by macrophages and dendritic cells, and thus, can get involved in a range of biological processes, such as angiogenesis and tissue remodeling, immune modulation, and immunologic tolerance. The notion of C1q involvement in the pathogenesis of cancer is still evolving. C1q appears to have a dual role in cancer: tumor promoting as well as tumor-protective, depending on the context of the disease. In the current study, we performed a bioinformatics analysis to investigate whether C1q can serve as a potential prognostic marker for human carcinoma. We used the Oncomine database and the survival analysis platforms Kaplan-Meier plotter. Our results showed that high levels of C1q have a favorable prognostic index in basal-like breast cancer for disease-free survival, and in HER2-positive breast cancer for overall survival, while it showed a pro-tumorigenic role of C1q in lung adenocarcinoma, and in clear cell renal cell carcinoma. This in silico study, if validated via a retrospective study, can be a step forward in establishing C1q as a new tool as a prognostic biomarker for various carcinoma.
- Published
- 2019
- Full Text
- View/download PDF
209. Transcriptomics and Immunological Analyses Reveal a Pro-Angiogenic and Anti-Inflammatory Phenotype for Decidual Endothelial Cells.
- Author
-
Agostinis C, Masat E, Bossi F, Ricci G, Menegazzi R, Lombardelli L, Zito G, Mangogna A, Degan M, Gattei V, Piccinni MP, Kishore U, and Bulla R
- Subjects
- 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, Neovascularization, Pathologic genetics, Neovascularization, Pathologic immunology, Skin immunology, Skin metabolism
- 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.
- Published
- 2019
- Full Text
- View/download PDF
210. Pathological Significance and Prognostic Value of Surfactant Protein D in Cancer.
- Author
-
Mangogna A, Belmonte B, Agostinis C, Ricci G, Gulino A, Ferrara I, Zanconati F, Tripodo C, Romano F, Kishore U, and Bulla R
- Subjects
- Breast Neoplasms genetics, Breast Neoplasms metabolism, Computational Biology, Computer Simulation, Datasets as Topic, Female, Humans, Lung Neoplasms genetics, Lung Neoplasms metabolism, Male, Ovarian Neoplasms genetics, Ovarian Neoplasms metabolism, Prognosis, Pulmonary Surfactant-Associated Protein D genetics, RNA, Neoplasm, Stomach Neoplasms genetics, Stomach Neoplasms metabolism, Survival Analysis, Biomarkers, Tumor metabolism, Neoplasms metabolism, Pulmonary Surfactant-Associated Protein D metabolism
- Abstract
Surfactant protein D (SP-D) is a pattern recognition molecule belonging to the Collectin (collagen-containing C-type lectin) family that has pulmonary as well as extra-pulmonary existence. In the lungs, it is a well-established opsonin that can agglutinate a range of microbes, and enhance their clearance via phagocytosis and super-oxidative burst. It can interfere with allergen-IgE interaction and suppress basophil and mast cell activation. However, it is now becoming evident that SP-D is likely to be an innate immune surveillance molecule against tumor development. SP-D has been shown to induce apoptosis in sensitized eosinophils derived from allergic patients and a leukemic cell line via p53 pathway. Recently, SP-D has been shown to suppress lung cancer progression via interference with the epidermal growth factor signaling. In addition, a truncated form of recombinant human SP-D has been reported to induce apoptosis in pancreatic adenocarcinoma via Fas-mediated pathway in a p53-independent manner. To further establish a correlation between SP-D presence/levels and normal and cancer tissues, we performed a bioinformatics analysis, using Oncomine dataset and the survival analysis platforms Kaplan-Meier plotter, to assess if SP-D can serve as a potential prognostic marker for human lung cancer, in addition to human gastric, breast, and ovarian cancers. We also analyzed immunohistochemically the presence of SP-D in normal and tumor human tissues. We conclude that (1) in the lung, gastric, and breast cancers, there is a lower expression of SP-D than normal tissues; (2) in ovarian cancer, there is a higher expression of SP-D than normal tissue; and (3) in lung cancer, the presence of SP-D could be associated with a favorable prognosis. On the contrary, at non-pulmonary sites such as gastric, breast, and ovarian cancers, the presence of SP-D could be associated with unfavorable prognosis. Correlation between the levels of SP-D and overall survival requires further investigation. Our analysis involves a large number of dataset; therefore, any trend observed is reliable. Despite apparent complexity within the results, it is evident that cancer tissues that produce less levels of SP-D compared to their normal tissue counterparts are probably less susceptible to SP-D-mediated immune surveillance mechanisms via infiltrating immune cells.
- Published
- 2018
- Full Text
- View/download PDF
211. Cell-autonomous and cell non-autonomous downregulation of tumor suppressor DAB2IP by microRNA-149-3p promotes aggressiveness of cancer cells.
- Author
-
Bellazzo A, Di Minin G, Valentino E, Sicari D, Torre D, Marchionni L, Serpi F, Stadler MB, Taverna D, Zuccolotto G, Montagner IM, Rosato A, Tonon F, Zennaro C, Agostinis C, Bulla R, Mano M, Del Sal G, and Collavin L
- Subjects
- Animals, HCT116 Cells, HeLa Cells, Hep G2 Cells, Human Umbilical Vein Endothelial Cells, Humans, Male, MicroRNAs genetics, Neoplasm Proteins genetics, PC-3 Cells, Prostatic Neoplasms genetics, Prostatic Neoplasms pathology, RNA, Neoplasm genetics, Zebrafish, ras GTPase-Activating Proteins genetics, MicroRNAs metabolism, Neoplasm Proteins metabolism, Prostatic Neoplasms metabolism, RNA, Neoplasm metabolism, Signal Transduction, Tumor Microenvironment, ras GTPase-Activating Proteins metabolism
- Abstract
The tumor suppressor DAB2IP contributes to modulate the network of information established between cancer cells and tumor microenvironment. Epigenetic and post-transcriptional inactivation of this protein is commonly observed in multiple human malignancies, and can potentially favor progression of tumors driven by a variety of genetic mutations. Performing a high-throughput screening of a large collection of human microRNA mimics, we identified miR-149-3p as a negative post-transcriptional modulator of DAB2IP. By efficiently downregulating DAB2IP, this miRNA enhances cancer cell motility and invasiveness, facilitating activation of NF-kB signaling and promoting expression of pro-inflammatory and pro-angiogenic factors. In addition, we found that miR-149-3p secreted by prostate cancer cells induces DAB2IP downregulation in recipient vascular endothelial cells, stimulating their proliferation and motility, thus potentially remodeling the tumor microenvironment. Finally, we found that inhibition of endogenous miR-149-3p restores DAB2IP activity and efficiently reduces tumor growth and dissemination of malignant cells. These observations suggest that miR-149-3p can promote cancer progression via coordinated inhibition of DAB2IP in tumor cells and in stromal cells.
- Published
- 2018
- Full Text
- View/download PDF
212. Pre-eclampsia affects procalcitonin production in placental tissue.
- Author
-
Agostinis C, Rami D, Zacchi P, Bossi F, Stampalija T, Mangogna A, Amadio L, Vidergar R, Vecchi Brumatti L, Ricci G, Celeghini C, Radillo O, Sargent I, and Bulla R
- Subjects
- Adult, Cohort Studies, Female, Humans, Placenta pathology, Trophoblasts pathology, Tumor Necrosis Factor-alpha metabolism, Up-Regulation, Young Adult, Calcitonin metabolism, Macrophages immunology, Placenta metabolism, Pre-Eclampsia metabolism, Pregnancy, Trophoblasts metabolism
- Abstract
Problem: Procalcitonin (PCT) is the prohormone of calcitonin which is usually released from neuroendocrine cells of the thyroid gland (parafollicular) and the lungs (K cells). PCT is synthesized by almost all cell types and tissues, including monocytes and parenchymal tissue, upon LPS stimulation. To date, there is no evidence for PCT expression in the placenta both in physiological and pathological conditions., Method: Circulating and placental PCT levels were analysed in pre-eclamptic (PE) and control patients. Placental cells and macrophages (PBDM), stimulated with PE sera, were analysed for PCT expression. The effect of anti-TNF-α antibody was analysed., Results: Higher PCT levels were detected in PE sera and in PE placentae compared to healthy women. PE trophoblasts showed increased PCT expression compared to those isolated from healthy placentae. PE sera induced an upregulation of PCT production in macrophages and placental cells. The treatment of PBDM with PE sera in the presence of anti-TNF-α completely abrogated the effect induced by pathologic sera., Conclusion: Trophoblast cells are the main producer of PCT in PE placentae. TNF-α, in association with other circulating factors present in PE sera, upregulates PCT production in macrophages and normal placental cells, thus contributing to the observed increased in circulating PCT in PE sera., (© 2018 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd.)
- Published
- 2018
- Full Text
- View/download PDF
213. Complement Protein C1q Binds to Hyaluronic Acid in the Malignant Pleural Mesothelioma Microenvironment and Promotes Tumor Growth.
- Author
-
Agostinis C, Vidergar R, Belmonte B, Mangogna A, Amadio L, Geri P, Borelli V, Zanconati F, Tedesco F, Confalonieri M, Tripodo C, Kishore U, and Bulla R
- Abstract
C1q is the first recognition subcomponent of the complement classical pathway, which acts toward the clearance of pathogens and apoptotic cells. C1q is also known to modulate a range of functions of immune and non-immune cells, and has been shown to be involved in placental development and sensorial synaptic pruning. We have recently shown that C1q can promote tumor by encouraging their adhesion, migration, and proliferation in addition to angiogenesis and metastasis. In this study, we have examined the role of human C1q in the microenvironment of malignant pleural mesothelioma (MPM), a rare form of cancer commonly associated with exposure to asbestos. We found that C1q was highly expressed in all MPM histotypes, particularly in epithelioid rather than in sarcomatoid histotype. C1q avidly bound high and low molecular weight hyaluronic acid (HA) via its globular domain. C1q bound to HA was able to induce adhesion and proliferation of mesothelioma cells (MES) via enhancement of ERK1/2, SAPK/JNK, and p38 phosphorylation; however, it did not activate the complement cascade. Consistent with the modular organization of the globular domain, we demonstrated that C1q may bind to HA through ghA module, whereas it may interact with human MES through the ghC. In conclusion, C1q highly expressed in MPM binds to HA and enhances the tumor growth promoting cell adhesion and proliferation. These data can help develop novel diagnostic markers and molecular targets for MPM.
- Published
- 2017
- Full Text
- View/download PDF
214. Alternative functions of the complement protein C1q at embryo implantation site.
- Author
-
Agostinis C, Tedesco F, and Bulla R
- Subjects
- Animals, Extracellular Signal-Regulated MAP Kinases metabolism, Female, Humans, Integrin alpha4beta1 metabolism, Mice, Mice, Knockout, Complement C1q immunology, Decidua physiology, Embryo Implantation, Neovascularization, Physiologic, Pre-Eclampsia immunology, Pregnancy immunology, Trophoblasts physiology, Vascular Remodeling
- Abstract
Complement component C1q is one of the recognition molecules of the complement system which can serve several functions unrelated to complement activation. This molecule is produced at foeto-maternal interface by macrophages as wells as by decidual endothelial cells and invading trophoblast. Foetal trophoblast cells migrating through the decidua in the early stages of pregnancy synthesize and express C1q on their surface, which is actively involved in promoting trophoblast endovascular and interstitial invasion of the decidua. These functions are mediated by two cell surface receptors, gC1qR and α4β1 integrin, which promote trophoblast adhesion and migration through the activation of ERK1/2 MAPKs. C1q
-/- mice manifest increased frequency of foetal resorption, reduced foetal weight, and smaller litter size when compared to their wild-type counterparts, suggesting that defective local production of C1q may be involved in pregnancy disorders, such as pre-eclampsia. C1q acts also as a strong angiogenic factor and promotes neovascularization. These studies suggest novel and unexpected roles of this complement component in physiological and pathological pregnancies., (Copyright © 2016 Elsevier Ireland Ltd. All rights reserved.)- Published
- 2017
- Full Text
- View/download PDF
215. Complement component C1q as potential diagnostic but not predictive marker of preeclampsia.
- Author
-
Agostinis C, Stampalija T, Tannetta D, Loganes C, Vecchi Brumatti L, De Seta F, Celeghini C, Radillo O, Sargent I, Tedesco F, and Bulla R
- Subjects
- Adult, Biomarkers blood, Case-Control Studies, Complement Activation, Complement C4 metabolism, Complement C5a metabolism, Complement Membrane Attack Complex metabolism, Female, Gestational Age, Humans, Pre-Eclampsia immunology, Pre-Eclampsia pathology, Pregnancy, Complement C1q metabolism, Pre-Eclampsia blood, Pre-Eclampsia diagnosis
- Abstract
Problem: We have previously found that C1q is constitutively expressed by invading trophoblast and endothelial cells of decidua and contributes to vascular and tissue remodeling. Based on these findings, we sought to determine whether there were changes in the circulating level of C1q that may be used as a diagnostic and predictive marker of preeclampsia., Method of Study: We measured the levels of C1q, C4, and complement activation products in serum or plasma of normal pregnant women and preeclamptic patients from different cohorts., Results: We observed a marked decrease in the concentration of C1q associated with a reduced level of C4 in preeclamptic patients as compared to matched healthy pregnant woman but no significant difference in the circulating level of the activating products C5a and the soluble terminal complement complex sC5b-9. Analysis of serum samples collected at early phase of pregnancy from women who later developed preeclampsia failed to show a decrease in C1q level., Conclusion: The results of the present investigation demonstrate that low levels of C1q and C4 are associated with preeclampsia but cannot be used as predictive markers., (© 2016 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd.)
- Published
- 2016
- Full Text
- View/download PDF
216. C1q acts in the tumour microenvironment as a cancer-promoting factor independently of complement activation.
- Author
-
Bulla R, Tripodo C, Rami D, Ling GS, Agostinis C, Guarnotta C, Zorzet S, Durigutto P, Botto M, and Tedesco F
- Subjects
- Animals, Apoptosis, Cell Line, Tumor, Cell Movement, Cell Proliferation, Complement C1q genetics, Complement C3 genetics, Complement C3 metabolism, Complement C5 genetics, Complement C5 metabolism, Humans, Mice, Mice, Inbred C57BL, Mice, Knockout, Complement Activation physiology, Complement C1q metabolism, Neoplasms metabolism
- Abstract
Complement C1q is the activator of the classical pathway. However, it is now recognized that C1q can exert functions unrelated to complement activation. Here we show that C1q, but not C4, is expressed in the stroma and vascular endothelium of several human malignant tumours. Compared with wild-type (WT) or C3- or C5-deficient mice, C1q-deficient (C1qa(-/-)) mice bearing a syngeneic B16 melanoma exhibit a slower tumour growth and prolonged survival. This effect is not attributable to differences in the tumour-infiltrating immune cells. Tumours developing in WT mice display early deposition of C1q, higher vascular density and an increase in the number of lung metastases compared with C1qa(-/-) mice. Bone marrow (BM) chimeras between C1qa(-/-) and WT mice identify non-BM-derived cells as the main local source of C1q that can promote cancer cell adhesion, migration and proliferation. Together these findings support a role for locally synthesized C1q in promoting tumour growth.
- Published
- 2016
- Full Text
- View/download PDF
217. Critical Role and Therapeutic Control of the Lectin Pathway of Complement Activation in an Abortion-Prone Mouse Mating.
- Author
-
Petitbarat M, Durigutto P, Macor P, Bulla R, Palmioli A, Bernardi A, De Simoni MG, Ledee N, Chaouat G, and Tedesco F
- Subjects
- Animals, Antibodies, Blocking administration & dosage, Complement C5 immunology, Complement C5 metabolism, Disease Models, Animal, Embryo Implantation drug effects, Female, Humans, Male, Mannose-Binding Lectin metabolism, Mice, Mice, Inbred BALB C, Mice, Inbred CBA, Mice, Inbred DBA, Pre-Eclampsia immunology, Pregnancy, Complement Pathway, Mannose-Binding Lectin drug effects, Pre-Eclampsia drug therapy
- Abstract
The abortion-prone mating combination CBA/J × DBA/2 has been recognized as a model of preeclampsia, and complement activation has been implicated in the high rate of pregnancy loss observed in CBA/J mice. We have analyzed the implantation sites collected from DBA/2-mated CBA/J mice for the deposition of the complement recognition molecules using CBA/J mated with BALB/c mice as a control group. MBL-A was observed in the implantation sites of CBA/J × DBA/2 combination in the absence of MBL-C and was undetectable in BALB/c-mated CBA/J mice. Conversely, C1q was present in both mating combinations. Searching for other complement components localized at the implantation sites of CBA/J × DBA/2, we found C4 and C3, but we failed to reveal C1r. These data suggest that complement is activated through the lectin pathway and proceeds to completion of the activation sequence as revealed by C9 deposition. MBL-A was detected as early as 3.5 d of pregnancy, and MBL-A deficiency prevented pregnancy loss in the abortion-prone mating combination. The contribution of the terminal complex to miscarriage was supported by the finding that pregnancy failure was largely inhibited by the administration of neutralizing Ab to C5. Treatment of DBA/2-mated CBA/J mice with Polyman2 that binds to MBL-A with high affinity proved to be highly effective in controlling the activation of the lectin pathway and in preventing fetal loss., (Copyright © 2015 by The American Association of Immunologists, Inc.)
- Published
- 2015
- Full Text
- View/download PDF
218. RelB activation in anti-inflammatory decidual endothelial cells: a master plan to avoid pregnancy failure?
- Author
-
Masat E, Gasparini C, Agostinis C, Bossi F, Radillo O, De Seta F, Tamassia N, Cassatella MA, and Bulla R
- Subjects
- Abortion, Spontaneous pathology, Abortion, Spontaneous prevention & control, Cells, Cultured, Cytokines immunology, Endometrium pathology, Endothelial Cells pathology, Female, Humans, Inflammation pathology, Inflammation prevention & control, Pregnancy, Abortion, Spontaneous immunology, Endometrium immunology, Endothelial Cells immunology, Inflammation immunology, Maternal-Fetal Exchange immunology, Transcription Factor RelB immunology
- Abstract
It is known that excessive inflammation at fetal-maternal interface is a key contributor in a compromised pregnancy. Female genital tract is constantly in contact with microorganisms and several strategies must be adopted to avoid pregnancy failure. Decidual endothelial cells (DECs) lining decidual microvascular vessels are the first cells that interact with pro-inflammatory stimuli released into the environment by microorganisms derived from gestational tissues or systemic circulation. Here, we show that DECs are hypo-responsive to LPS stimulation in terms of IL-6, CXCL8 and CCL2 production. Our results demonstrate that DECs express low levels of TLR4 and are characterized by a strong constitutive activation of the non-canonical NF-κB pathway and a low responsiveness of the canonical pathway to LPS. In conclusion, DECs show a unique hypo-responsive phenotype to the pro-inflammatory stimulus LPS in order to control the inflammatory response at feto-maternal interface.
- Published
- 2015
- Full Text
- View/download PDF
219. Mutant p53 reprograms TNF signaling in cancer cells through interaction with the tumor suppressor DAB2IP.
- Author
-
Di Minin G, Bellazzo A, Dal Ferro M, Chiaruttini G, Nuzzo S, Bicciato S, Piazza S, Rami D, Bulla R, Sommaggio R, Rosato A, Del Sal G, and Collavin L
- Subjects
- Animals, Breast Neoplasms metabolism, Cell Line, Tumor, Cytoplasm metabolism, Female, HCT116 Cells, Humans, Lymphatic Metastasis, Mammary Neoplasms, Experimental, Mice, Mice, SCID, Mutation, Missense, Breast Neoplasms genetics, Breast Neoplasms pathology, Signal Transduction, Tumor Necrosis Factor-alpha metabolism, Tumor Suppressor Protein p53 genetics, Tumor Suppressor Protein p53 metabolism, ras GTPase-Activating Proteins metabolism
- Abstract
Inflammation is a significant factor in cancer development, and a molecular understanding of the parameters dictating the impact of inflammation on cancers could significantly improve treatment. The tumor suppressor p53 is frequently mutated in cancer, and p53 missense mutants (mutp53) can acquire oncogenic properties. We report that cancer cells with mutp53 respond to inflammatory cytokines increasing their invasive behavior. Notably, this action is coupled to expression of chemokines that can expose the tumor to host immunity, potentially affecting response to therapy. Mechanistically, mutp53 fuels NF-κB activation while it dampens activation of ASK1/JNK by TNFα, and this action depends on mutp53 binding and inhibiting the tumor suppressor DAB2IP in the cytoplasm. Interfering with such interaction reduced aggressiveness of cancer cells in xenografts. This interaction is an unexplored mechanism by which mutant p53 can influence tumor evolution, with implications for our understanding of the complex role of inflammation in cancer., (Copyright © 2014 Elsevier Inc. All rights reserved.)
- Published
- 2014
- Full Text
- View/download PDF
220. Immunomodulation mediated by a herbal syrup containing a standardized Echinacea root extract: a pilot study in healthy human subjects on cytokine gene expression.
- Author
-
Dapas B, Dall'Acqua S, Bulla R, Agostinis C, Perissutti B, Invernizzi S, Grassi G, and Voinovich D
- Subjects
- Adult, Down-Regulation, Female, Glycosides pharmacology, Healthy Volunteers, Humans, Interleukin-2 blood, Interleukin-6 blood, Interleukin-8 blood, Lymphocytes drug effects, Male, Middle Aged, Monocytes drug effects, Pilot Projects, Plant Roots chemistry, Tumor Necrosis Factor-alpha blood, Cytokines blood, Echinacea chemistry, Immunomodulation, Plant Extracts pharmacology
- Abstract
In this study, the immunomodulatory effect of a triply standardized Echinacea angustifolia root extract (Polinacea(®)) was evaluated in 10 healthy subjects. Ten ml of syrup containing one hundred mg of extract (corresponding to 4.7 mg of Echinacoside and 8.0mg of a high molecular weight-20,000 Da- polysaccharide) were administered as a herbal syrup once a day for one month. The immunomodulatory effect was evaluated before and after herbal syrup administration evaluating the expression levels of the cytokines IL-2, IL-8, IL-6 and TNF-α. Cytokine expression was studied in lympho-monocytes and in plasma samples measuring the mRNA and protein levels, respectively. The results were analysed by ANOVA and non-parametric Friedman rank sum tests; when possible it was adopted a pair-wise comparisons at different post-treatment times, using the paired t-tests with Holm correction. The correlation between the variations of cytokine plasma levels and the respective mRNA was carried out using a linear regression model. In lympho-monocytes our data indicate the up-regulation of the mRNA levels of IL-2 and IL-8 and the down regulation of the mRNA levels of the pro-inflammatory cytokines TNF-α and IL6. The differential regulation was maximal after 14 days of treatment. IL-2 up-regulation and IL-6 down-regulation were also confirmed at the protein level in plasma. Finally, the up-regulation of the mRNA of IL-2/IL-8 and the down-regulation of IL-6 positively correlated with the protein levels detected in the plasma. In conclusion, this pilot study suggests a relevant role for the standardized Echinacea angustifolia root extract in the control of cytokine expression. This first demonstration of the immuno-modulating activity of Echinacea angustifolia root extract in the healthy subject, supports at least in part the common use of such products as health promoting supplement., (Copyright © 2014 Elsevier GmbH. All rights reserved.)
- Published
- 2014
- Full Text
- View/download PDF
221. A non-complement-fixing antibody to β2 glycoprotein I as a novel therapy for antiphospholipid syndrome.
- Author
-
Agostinis C, Durigutto P, Sblattero D, Borghi MO, Grossi C, Guida F, Bulla R, Macor P, Pregnolato F, Meroni PL, and Tedesco F
- Subjects
- Abortion, Spontaneous immunology, Animals, Antibodies, Monoclonal genetics, Complement Activation drug effects, Complement Activation immunology, Human Umbilical Vein Endothelial Cells, Humans, Immunoglobulin G immunology, Male, Mice, Protein Binding immunology, Rats, Recombinant Proteins immunology, Recombinant Proteins isolation & purification, Recombinant Proteins therapeutic use, Single-Chain Antibodies immunology, Single-Chain Antibodies isolation & purification, Single-Chain Antibodies therapeutic use, Thrombosis immunology, Trophoblasts, beta 2-Glycoprotein I metabolism, Antibodies, Monoclonal immunology, Antibodies, Monoclonal therapeutic use, Antiphospholipid Syndrome drug therapy, Antiphospholipid Syndrome immunology, Autoantigens immunology, Complement System Proteins immunology, beta 2-Glycoprotein I immunology
- Abstract
A single-chain fragment variable (scFv) recognizing β2-glycoprotein 1 (β2GPI) from humans and other species was isolated from a human phage display library and engineered to contain an IgG1 hinge-CH2-CH3 domain. The scFv-Fc directed against β2GPI domain I-induced thrombosis and fetal loss, thus mimicking the effect of antibodies from patients with antiphospholipid syndrome (APS). Complement is involved in the biological effect of anti-β2GPI scFv-Fc, as demonstrated by its ability to promote in vitro and in vivo complement deposition and the failure to induce vascular thrombosis in C6-deficient rats and fetal loss in C5-depleted mice. A critical role for complement was also supported by the inability of the CH2-deleted scFv-Fc to cause vessel occlusion and pregnancy failure. This antibody prevented the pathological effects of anti-β2GPI antibodies from APS patients and displaced β2GPI-bound patient antibodies. The CH2-deleted antibody represents an innovative approach potentially useful to treat APS patients refractory to standard therapy., (© 2014 by The American Society of Hematology.)
- Published
- 2014
- Full Text
- View/download PDF
222. C1q as a unique player in angiogenesis with therapeutic implication in wound healing.
- Author
-
Bossi F, Tripodo C, Rizzi L, Bulla R, Agostinis C, Guarnotta C, Munaut C, Baldassarre G, Papa G, Zorzet S, Ghebrehiwet B, Ling GS, Botto M, and Tedesco F
- Subjects
- 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, Complement C1q physiology, Endothelial Cells drug effects, Neovascularization, Physiologic genetics, Wound Healing genetics
- 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.
- Published
- 2014
- Full Text
- View/download PDF
223. The first trimester gravid serum regulates procalcitonin expression in human macrophages skewing their phenotype in vitro.
- Author
-
Rami D, La Bianca M, Agostinis C, Zauli G, Radillo O, and Bulla R
- Subjects
- Biomarkers metabolism, Calcitonin Gene-Related Peptide, Cells, Cultured, Chorionic Gonadotropin metabolism, Estradiol metabolism, Female, Gene Expression Regulation, Homeostasis, Humans, Inflammation, Macrophages metabolism, Monocytes cytology, Phenotype, Pregnancy, Pregnancy Trimester, First, Up-Regulation, Calcitonin blood, Macrophages cytology, Protein Precursors blood, Serum metabolism
- Abstract
Procalcitonin (PCT) is one of the best diagnostic and prognostic markers in clinical practice, widely used to evaluate the evolution of bacterial infections. Although it is mainly produced by thyroid, during sepsis almost all the peripheral tissues are involved in PCT production. Parenchymal cells have been suggested as the main source of PCT expression; however the contribution of macrophages is not clear yet. In response to environmental cues, tissue macrophages acquire distinct functional phenotypes, ranging from proinflammatory (M1) to anti-inflammatory (M2) phenotype. Macrophages at the fetal-maternal interface show immunosuppressive M2-like activities required for the maintenance of immunological homeostasis during pregnancy. This study aims to clarify the ability to synthesise PCT of fully differentiated (M0), polarized (M1/M2) macrophages and those cultured either in the presence of first trimester gravid serum (GS) or pregnancy hormones. We found out that M1 macrophages upregulate PCT expression following LPS stimulation compared to M0 and M2. The GS downregulates PCT expression in macrophages, skewing them towards an M2-like phenotype. This effect seems only partially mediated by the hormonal milieu. Our findings strengthen the key role of macrophages in counteracting inflammatory stimuli during pregnancy, suggesting PCT as a possible new marker of M1-like macrophages.
- Published
- 2014
- Full Text
- View/download PDF
224. Bacterial LPS differently modulates inflammasome gene expression and IL-1β secretion in trophoblast cells, decidual stromal cells, and decidual endothelial cells.
- Author
-
Pontillo A, Girardelli M, Agostinis C, Masat E, Bulla R, and Crovella S
- Subjects
- Carrier Proteins genetics, Carrier Proteins metabolism, Cells, Cultured, Decidua immunology, Endothelial Cells immunology, Female, Gene Expression Regulation, Humans, Immunity, Innate drug effects, Inflammasomes genetics, Inflammasomes immunology, Interleukin-1beta genetics, NLR Family, Pyrin Domain-Containing 3 Protein, Pregnancy, Pregnancy Trimester, First, RNA, Messenger metabolism, Stromal Cells immunology, Time Factors, Transcription, Genetic, Trophoblasts immunology, Decidua drug effects, Endothelial Cells drug effects, Endotoxins pharmacology, Inflammasomes drug effects, Interleukin-1beta metabolism, Stromal Cells drug effects, Trophoblasts drug effects
- Abstract
Three Nod-like receptors (NLR family, pyrin domain containing 1/NLRP1, NLR family, pyrin domain containing 3/NLRP3, NLR family, CARD domain containing 4/NLRC4) and the adaptor molecule PYD and CARD domain containing protein/PYCARD are involved in the assembling of multiprotein complexes known as inflammasomes, leading to caspase 1 activation and consequent interleukin (IL)-1β secretion. Considering that inflammasomes are involved in sensing pathogens and in triggering inflammatory and immune response, we hypothesized that they could also act in the placenta as an efficient innate mechanism during pregnancy infections. For this reason the activation of inflammasome was tested in 3 human placental cell populations in the presence of a common gram-negative compound (lipopolysaccharide [LPS]). The transcription of NLRP1, NLRP3, NLRC4, PYCARD, CASP1, and IL1B genes and the secretion of IL-1β were evaluated in human first trimester cytotrophoblasts (CTBs), decidual stromal cells (DSCs), and endothelial cells (DECs) stimulated with LPS. In CTBs and DSCs, LPS induced an augmented expression of CASP1 and IL1B and the specific upregulation of NLRP3 within the 3 NLRs tested. Moreover, LPS induced secretion of IL-1β from CTBs and DSCs. These results suggest the involvement of NLRP3 inflammasome in the placental innate response. The LPS did not affect inflammasome gene transcription and IL-1β production in DECs. Bacterial LPS enhances NLRP3 inflammasome components in trophoblast and DSCs, suggesting that this innate immune complex could play a key role in placental immune defense.
- Published
- 2013
- Full Text
- View/download PDF
225. Soluble TRAIL is elevated in recurrent miscarriage and inhibits the in vitro adhesion and migration of HTR8 trophoblastic cells.
- Author
-
Agostinis C, Bulla R, Tisato V, De Seta F, Alberico S, Secchiero P, and Zauli G
- Subjects
- Abortion, Habitual pathology, Apoptosis, Cell Line, Cells, Cultured, Enzyme-Linked Immunosorbent Assay, Female, Humans, Pregnancy, Receptors, TNF-Related Apoptosis-Inducing Ligand metabolism, TNF-Related Apoptosis-Inducing Ligand pharmacology, Trophoblasts pathology, Abortion, Habitual metabolism, Cell Adhesion drug effects, Cell Movement drug effects, TNF-Related Apoptosis-Inducing Ligand blood, Trophoblasts drug effects
- Abstract
Study Question: What is the potential physiopathological role of tumour necrosis factor-related apoptosis-inducing ligand (TRAIL) in recurrent miscarriage (RM), characterized by at least three consecutive pregnancy losses., Summary Answer: The levels of serum TRAIL immediately after miscarriage in RM patients are significantly elevated with respect to that in first-trimester normal pregnant women, and recombinant TRAIL inhibits the adhesion and migration of HTR8 trophoblastic cells in vitro., What Is Known Already: Both TRAIL and its trans-membrane receptors (TRAIL-R1, TRAIL-R2, TRAIL-R3 and TRAIL-R4) have been documented in the placenta, but their physiopathological role is incompletely understood., Study Design, Size, Duration: The study populations consisted of RM patients (n = 80) and first-trimester normal pregnant women (n = 80). Blood samples were obtained within 24 h after abortion (RM) or at gestational 12-week (normal pregnant women). As additional controls, third-trimester normal pregnant women (n = 28) were examined before (within 72 h) and after (within 24 h) partum., Participants/materials, Setting, Methods: The concentrations of TRAIL were analysed in serum samples by ELISA. In parallel, the effect of soluble recombinant TRAIL (0.1-1000 ng/ml) was analysed on the survival of primary extravillus trophoblasts (EVTs) and on the survival, proliferation, adhesion and migration of trophoblastic HTR8 cells., Main Results and the Role of Chance: The circulating levels of TRAIL in RM women (median: 52.5 pg/ml; mean and SD: 55.5 ± 24.4 pg/ml) were significantly higher with respect to first-trimester normal pregnant women (median: 44.9 pg/ml; mean and SD: 47 ± 15.1 pg/ml) and third-trimester normal pregnant women, as assessed before (median: 45.1 pg/ml; mean and SD: 46 ± 12.4 pg/ml) and after partum (median: 35.4 pg/ml; mean and SD: 38 + 17.5 pg/ml). Both primary EVT and HTR8 cells expressed detectable levels of TRAIL death receptors, but exposure to soluble recombinant TRAIL did not induce cell death of trophoblastic cells. On the other hand, TRAIL dose-dependently inhibited the adhesion of HTR8 cells to decidual endothelial cells (DEC) as well as the migration of HTR8 in transwell assays using either fibronectin or DEC., Limitations, Reasons for Caution: Although this study suggests that TRAIL might have a pathogenic role in RM by inhibiting both the adhesion and migration capabilities of first trimester trophoblastic cells, there is a possibility that the elevated serum levels of TRAIL in RM are not cause but rather the result of RM., Wider Implications of the Findings: Our current findings together with data of other authors suggest that circulating TRAIL should be further analysed as a potential important biomarker in different physiopathological settings., Study Funding/competing Interest(s): This study was funded by FIRB projects (RBAP11Z4Z9_002 to Giorgio Zauli and RBAP10447J_002 to Paola Secchiero). The authors have no competing interests to declare.
- Published
- 2012
- Full Text
- View/download PDF
226. Chemerin regulates NK cell accumulation and endothelial cell morphogenesis in the decidua during early pregnancy.
- Author
-
Carlino C, Trotta E, Stabile H, Morrone S, Bulla R, Soriani A, Iannitto ML, Agostinis C, Mocci C, Minozzi M, Aragona C, Perniola G, Tedesco F, Sozzani S, Santoni A, and Gismondi A
- Subjects
- Capillaries metabolism, Cell Movement immunology, Chemokines genetics, Decidua blood supply, Decidua physiology, Endothelial Cells physiology, Female, Humans, Intercellular Signaling Peptides and Proteins, Neovascularization, Physiologic physiology, Pregnancy, RNA, Messenger metabolism, Receptors, Chemokine genetics, Receptors, Chemokine metabolism, Stromal Cells cytology, Stromal Cells physiology, Trophoblasts cytology, Trophoblasts physiology, Chemokines metabolism, Decidua cytology, Endothelial Cells cytology, Killer Cells, Natural cytology, MAP Kinase Signaling System physiology, Pregnancy Trimester, First physiology
- Abstract
Context: Although decidual natural killer (NK) cell accumulation and vascular remodeling are critical steps to ensure successful pregnancy, the molecular mechanisms controlling these events are poorly defined., Objective: Herein we analyzed whether chemerin, a recently identified chemoattractant involved in many pathophysiological processes, could be expressed in the uterine compartment and could regulate events relevant for the good outcome of pregnancy., Design: Chemerin expression in human primary culture of stromal (ST) cells, extravillous trophoblast cells, and decidual endothelial cells (DEC) was analyzed by RT-PCR, ELISA, and Western blot. Migration through ST or DEC of peripheral blood and decidual (d) NK cells from pregnant women was performed using a transwell assay. A DEC capillary-like tube formation assay was used to evaluate endothelial morphogenesis., Results: Chemerin is differentially expressed by decidual cells during early pregnancy being present at high levels in ST and extravillous trophoblast cells but not in DEC. Notably, ST cells from pregnant women exhibit and release higher levels of chemerin as compared with ST cells from menopausal or fertile nonpregnant women. Chemerin can support peripheral blood NK cell migration through both DEC and ST cells. Although dNK cells exhibit lower chemerin receptor (CMKLR1) expression than their blood counterpart, CMKLR1 engagement on dNK cells resulted in both ERK activation and migration through decidual ST cells. Interestingly, DEC also express CMKLR1 and undergo ERK activation and capillary-like tube structure formation upon exposure to chemerin., Conclusions: Our data indicate that chemerin is up-regulated during decidualization and might contribute to NK cell accumulation and vascular remodeling during early pregnancy.
- Published
- 2012
- Full Text
- View/download PDF
227. The complement system at the embryo implantation site: friend or foe?
- Author
-
Bulla R, Bossi F, and Tedesco F
- Abstract
An inflammatory-like process and vascular remodeling represent the main changes that occur in decidua in the early phase of pregnancy. These changes are partly induced by trophoblast cells that colonize the decidua and are also contributed by the complement system, which can easily be activated as a result of tissue remodeling. Local control by several complement regulators including surface-bound and soluble molecules is critical to prevent complement-mediated tissue damage in normal pregnancy. C7 expressed on the endothelial cells (ECs) surface has been recognized as a novel complement regulator involved in the control of the proinflammatory effect of the terminal complement complex. The protective role of placental complement regulators in pregnancy is underscored by the recent finding of an association of preeclampsia with mutations in the genes encoding for some of these proteins. Complement components produced at feto-maternal interface serve an important function in placental development. C1q synthesized by decidual ECs and expressed on the cell surface is particularly important in this regard because it acts as a molecular bridge between endovascular trophoblast and ECs. C1q is also produced by extravillous trophoblast and is used to favor trophoblast migration through the decidua. Defective expression of C1q by trophoblast is associated with impaired trophoblast invasion of decidua and may have important implications in pregnancy disorders such as preeclampsia characterized by reduced vascular remodeling.
- Published
- 2012
- Full Text
- View/download PDF
228. MBL interferes with endovascular trophoblast invasion in pre-eclampsia.
- Author
-
Agostinis C, Bossi F, Masat E, Radillo O, Tonon M, De Seta F, Tedesco F, and Bulla R
- Subjects
- Cell Communication physiology, Decidua cytology, Endothelial Cells cytology, Female, Humans, Mannose-Binding Lectin blood, Pregnancy, Transendothelial and Transepithelial Migration, Endothelial Cells metabolism, Mannose-Binding Lectin metabolism, Pre-Eclampsia metabolism, Trophoblasts physiology
- 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.
- Published
- 2012
- Full Text
- View/download PDF
229. In vivo distribution of β2 glycoprotein I under various pathophysiologic conditions.
- Author
-
Agostinis C, Biffi S, Garrovo C, Durigutto P, Lorenzon A, Bek A, Bulla R, Grossi C, Borghi MO, Meroni P, and Tedesco F
- Subjects
- Animals, Complement C1q metabolism, Complement C3 metabolism, Complement C9 metabolism, Endothelial Cells pathology, Endothelium, Vascular pathology, Female, Fetal Death blood, Fetal Death pathology, Humans, Mice, Mice, Inbred BALB C, Pregnancy, Trophoblasts pathology, Uterus blood supply, Uterus pathology, Endothelial Cells metabolism, Endothelium, Vascular metabolism, Trophoblasts metabolism, Uterus metabolism, beta 2-Glycoprotein I blood
- Abstract
In vitro studies have documented β2 glycoprotein I (β2GPI) binding to endothelial cells (ECs) and trophoblast using antiphospholipid antibodies. The in vivo binding of β2GPI to these cells and the conditions that favor their interaction have not been investigated. We analyzed the in vivo distribution of cyanine 5.5-labeled β2GPI in mice and evaluated the effect of pregnancy and circulating antibodies on its tissue localization. The signal was detected in the liver by whole body scan and ex vivo analysis. The β2GPI failed to bind to the vascular endothelium and reacted only with the ECs of uterine vessels. In pregnant mice the protein was localized on ECs and trophoblast at the embryo implantation sites. Immunized mice showed a similar β2GPI biodistribution to naive mice but the immunized pregnant animals exhibited a significant increase in fetal loss associated with C3 and C9 deposition at the implantation sites. Treatment of mice with LPS after β2GPI-Cy5.5 injection promoted protein localization on gut and brain ECs associated with IgG, C1q, and C9 deposition in immunized mice. These findings indicate that β2GPI binding to EC requires priming with pro-inflammatory factors which is not needed for uterine and placental localization probably dependent on hormonal changes.
- Published
- 2011
- Full Text
- View/download PDF
230. Complement activation in animal and human pregnancies as a model for immunological recognition.
- Author
-
Girardi G, Prohászka Z, Bulla R, Tedesco F, and Scherjon S
- Subjects
- Abortion, Habitual immunology, Animals, Complement C1q immunology, Disease Models, Animal, Female, Fetal Growth Retardation immunology, Humans, Male, Maternal-Fetal Exchange immunology, Mice, Placentation immunology, Pre-Eclampsia immunology, Pregnancy, T-Lymphocytes immunology, Complement Activation, Models, Immunological
- Abstract
Pregnancy is a most intriguing feature of biology, not at least because of the need to regulate the maternal immune response against fetal antigens. The mammalian embryo expresses paternal antigens foreign to the mother's immune system and thus elicits an immune response that can lead to fetal rejection and bad pregnancy outcomes such as recurrent miscarriages and preeclampsia. More effective strategies to prevent these pregnancy complications should be forthcoming once the underlying pathophysiological mechanisms that are involved in fetal rejection are completely understood. Our goal in writing this review is to discuss the crucial role of the complement system as an effector mechanism in placental and fetal damage that leads to bad pregnancy outcomes. Important information about the role of excessive complement activation and bad pregnancy outcomes was obtained from animal models. That uncontrolled complement activation puts at risk the survival of the fetus was reported in mouse models of recurrent miscarriages and preeclampsia. Interestingly, several observations described in the mouse models were confirmed in humans. Increased circulating levels of complement proteins, and their activation fragments were found in patients with preeclampsia, recurrent miscarriages and intrauterine growth restriction. Studies performed in animals and humans demonstrated the deleterious effect of complement activation on pregnancy outcomes. However, we also described in this article the strategic role of complement component C1q in normal placentation. C1q deserves special consideration for its role in promoting trophoblast invasion of deciduas, a crucial step in normal placental development. In conclusion, in this review we discussed all the available results of basic and clinical studies on the role of the complement system in pregnancy to expand the understanding of the pathophysiology of pregnancy complications., (Copyright © 2011 Elsevier Ltd. All rights reserved.)
- Published
- 2011
- Full Text
- View/download PDF
231. An insight into normal and pathological pregnancies using large-scale microarrays: lessons from microarrays.
- Author
-
Chaouat G, Rodde N, Petitbarat M, Bulla R, Rahmati M, Dubanchet S, Zourbas S, Bataillon I, Coqué N, Hennuy B, Martal J, Munaut C, Aubert J, Sérazin V, Steffen T, Jensenius JC, Foidart JM, Sandra O, Tedesco F, and Lédée N
- Subjects
- Animals, Disease Models, Animal, Female, Gene Expression Profiling methods, Humans, Male, Mice, Mice, Inbred BALB C, Mice, Inbred CBA, Mice, Inbred DBA, Oligonucleotide Array Sequence Analysis methods, Pre-Eclampsia genetics, Pre-Eclampsia immunology, Pregnancy, Gene Expression Regulation immunology, Immune Tolerance, Pre-Eclampsia metabolism
- Abstract
In the introduction, we briefly recall old but classic evidence that there is no tolerance to paternal alloantigens in a first pregnancy. Therefore, we performed small- and large-scale microarrays in CBA × DBA/2 and CBA × BALB/c combinations, recently described as a murine model for preeclampsia. Our results are in line with other data suggesting a very early deregulation of local immune vascular events rather than a break of immune tolerance. Other data presented at the Tioman 2010 Preeclampsia Workshop supporting this hypothesis are briefly summarised, as well as indications and caveats from a recent human microarray on implantation failure and recurrent pregnancy loss., (Copyright © 2011 Elsevier Ireland Ltd. All rights reserved.)
- Published
- 2011
- Full Text
- View/download PDF
232. Mannose-binding lectin is produced by vaginal epithelial cells and its level in the vaginal fluid is influenced by progesterone.
- Author
-
Bulla R, De Seta F, Radillo O, Agostinis C, Durigutto P, Pellis V, De Santo D, Crovella S, and Tedesco F
- Subjects
- Adolescent, Adult, Body Fluids chemistry, Enzyme-Linked Immunosorbent Assay, Epithelial Cells immunology, Female, Humans, Immunohistochemistry, Mannose-Binding Lectin immunology, Menstrual Cycle metabolism, Reverse Transcriptase Polymerase Chain Reaction, Vagina chemistry, Vagina metabolism, Young Adult, Body Fluids immunology, Epithelial Cells metabolism, Mannose-Binding Lectin biosynthesis, Progesterone metabolism, Vagina immunology
- Abstract
Mannose-binding lectin (MBL) is a recognition molecule of the complement (C) system and binds to carbohydrate ligands present on a wide range of pathogenic bacteria, viruses, fungi, and parasites. MBL has been detected in the cervico-vaginal cavity where it can provide a first-line defence against infectious agents colonizing the lower tract of the reproductive system. Analysis of the cervico-vaginal lavage (CVL) obtained from 11 normal cycling women at different phases of the menstrual cycle revealed increased levels of MBL in the secretive phase. Part of this MBL derives from the circulation as indicated by the presence of transferrin in CVL tested as a marker of vascular and tissue permeability. The local synthesis of MBL is suggested by the finding that its level is substantially higher than that of transferrin in the secretive phase. The contribution of endometrium is negligible since the MBL level did not change before and after hysterectomy. RT-PCR and in situ RT-PCR analysis showed that the vaginal tissue, and in particular the basal layer of the epithelium, is a source of MBL which binds to the basal membrane and to cells of the outer layers of the epithelium. In conclusion, we have shown that MBL detected in CVL derives both from plasma as result of transudation and from local synthesis and its level is progesterone dependent increasing in the secretive phase of the menstrual cycle., (Copyright © 2010 Elsevier Ltd. All rights reserved.)
- Published
- 2010
- Full Text
- View/download PDF
233. An alternative role of C1q in cell migration and tissue remodeling: contribution to trophoblast invasion and placental development.
- Author
-
Agostinis C, Bulla R, Tripodo C, Gismondi A, Stabile H, Bossi F, Guarnotta C, Garlanda C, De Seta F, Spessotto P, Santoni A, Ghebrehiwet B, Girardi G, and Tedesco F
- Subjects
- 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, Chemotaxis, Leukocyte physiology, Complement C1q metabolism, Placentation immunology, Trophoblasts metabolism
- 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)β(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.
- Published
- 2010
- Full Text
- View/download PDF
234. Early regulators in abortion and implications for a preeclampsia model.
- Author
-
Chaouat G, Petitbarat M, Bulla R, Dubanchet S, Valdivia K, Ledée N, Steffen T, Jensenius JC, and Tedesco F
- Subjects
- Abortion, Induced, Animals, Antibodies, Blocking, Cytokine TWEAK, Disease Models, Animal, Female, Humans, Interleukin-23 genetics, Interleukins genetics, Male, Mannose-Binding Lectin genetics, Mannose-Binding Lectin immunology, Mice, Mice, Inbred Strains, Mice, Mutant Strains, Pre-Eclampsia immunology, Pregnancy, Receptors, Vascular Endothelial Growth Factor biosynthesis, Receptors, Vascular Endothelial Growth Factor genetics, Tumor Necrosis Factors genetics, Uterus immunology, Vascular Endothelial Growth Factor A biosynthesis, Vascular Endothelial Growth Factor A genetics, Infertility, Female immunology, Interleukin-23 biosynthesis, Interleukins biosynthesis, Mannose-Binding Lectin metabolism, Tumor Necrosis Factors biosynthesis, Uterus metabolism
- Abstract
This paper reports a summary of our comparative analysis of the uterine expression of interleukin-23 (IL-23), IL-27 and TWEAK in the CBA/J femalexDBA/2 male mouse mating combination, a model of immune-mediated early pregnancy loss. Compared with the MHC-identical CBA/JxBALB/c mating combination, which yields successful pregnancies, immunohistochemistry and qPCR in uterine tissue showed an immediate post-mating IL-27 hyper-expression after mating with DBA/2 males. Intra-uterine TWEAK expression was present in females mated with DBA/2 or Balb/c males from days 0.5 to 4.5 post-coitum (pc), peaking on day 0.5 pc together with uterine TNFalpha. In uteri of DBA/2 mated mice, TWEAK declined to almost undetectable levels on days 6.5-9.5 pc, a steeper drop than in BALB/c mated mice where TWEAK remained detectable. In both mating combinations, neutralisation of TWEAK by antibodies increased resorption rates, but surprisingly, so did IL-27 neutralisation. The complement regulator mannan binding lectin-A (MBL-A), but not MBL-C, was present on day 4.5 pc especially after mating with DBA/2 males. High levels of MBL are present in the uterine luminal fluid of sterile women, and possible functions for TWEAK and MBL in human implantation are indicated by their protein and mRNA expression in uterine biopsies from infertile and fertile individuals. Consistent with the results in mice, increased MBL expression is linked with pregnancy failure. Serum and uterine VEGF and VEGF receptor levels are also different between fertile and sterile patients. The implications of these findings for utilising the CBA/JxDBA/2 mating combination as an early onset model of preeclampsia are discussed.
- Published
- 2009
- Full Text
- View/download PDF
235. Complement production by trophoblast cells at the feto-maternal interface.
- Author
-
Bulla R, Bossi F, Agostinis C, Radillo O, Colombo F, De Seta F, and Tedesco F
- Subjects
- Cell Line, Complement System Proteins genetics, Complement System Proteins immunology, Enzyme-Linked Immunosorbent Assay, Female, Humans, Immunity, Interferon-gamma immunology, Pregnancy, Pregnancy Trimester, First, Trophoblasts immunology, Trophoblasts pathology, Complement System Proteins biosynthesis, Interferon-gamma metabolism, Maternal-Fetal Exchange immunology, Trophoblasts metabolism
- Abstract
An important role played by trophoblast cells at the feto-maternal interface is to exert immunomodulatory functions, including recognition of bacterial and viral agents and recruitment of leucocytes to eradicate pathogens. In this study we present data showing that the trophoblast cell line HTR8/SVneo and freshly isolated human first trimester trophoblast cells (CTBs) synthesize complement molecules C4, C3 and the late complement components, as assessed by ELISA and RT-PCR. Both cell types secrete C4 and C3, and HTR8/SVneo trophoblast cells secrete C6 in a measurable amount. The expression of C4 by HTR8/SVneo trophoblast cells and of C3 and C4 by CTBs was up-regulated by IFNgamma, while IL-1alpha and TNFalpha had no effect on the expression of complement components. In conclusion, we show that trophoblast cells produce complement components, and that synthesis of these proteins may be regulated by the pro-inflammatory cytokine IFNgamma. Complement synthesis by trophoblast cells potentially contributes to placental immune defence from pathogen infection.
- Published
- 2009
- Full Text
- View/download PDF
236. Higher interleukin-18 and mannose-binding lectin are present in uterine lumen of patients with unexplained infertility.
- Author
-
Oger P, Bulla R, Tedesco F, Portier A, Dubanchet S, Bailly M, Wainer R, Chaouat G, and Lédée N
- Subjects
- Adult, Embryo Implantation, Female, Fertilization in Vitro, Humans, Ovulation Induction methods, Pregnancy, Sperm Injections, Intracytoplasmic, Therapeutic Irrigation, Infertility, Female metabolism, Interleukin-18 metabolism, Mannose-Binding Lectin metabolism, Uterus metabolism
- Abstract
The uterine luminal environment was explored with regard to interleukin-18 (IL-18) and mannose-binding lectin (MBL) and the possibility that the procedure of flushing the uterine cavity would optimize the physiological initial pseudo-inflammatory uterine reaction. Uterine flushings were performed among 175 IVF/intracytoplasmic sperm injection (ICSI) patients at the time of oocyte retrieval and the cycles were compared with a control group matched for age, number of previous attempts and type of assisted reproductive procedure (IVF or ICSI) in which no flushing were performed (n = 175). Samples collected were divided into two groups according to the presence/absence of endometrial cells in samples. IL-18 and MBL expressions were explored by enzyme-linked immunosorbent assay. Implantation rates were significantly higher in those patients who underwent the uterine flushing compared with controls (P = 0.04). Luminal concentrations of IL-18 and MBL were higher if endometrial cells were present in flushings, suggesting endometrial origin of the secretion. Both concentrations of MBL and IL-18 were higher in patients with unexplained infertility compared with patients involved in IVF/ICSI for male or tubal infertility (P = 0.005 and 0.02, respectively). The exploration of the endoluminal environment before oocyte retrieval may enhance pregnancy rates and show distinct features in patients with unexplained infertility.
- Published
- 2009
- Full Text
- View/download PDF
237. C7 is expressed on endothelial cells as a trap for the assembling terminal complement complex and may exert anti-inflammatory function.
- Author
-
Bossi F, Rizzi L, Bulla R, Debeus A, Tripodo C, Picotti P, Betto E, Macor P, Pucillo C, Würzner R, and Tedesco F
- Subjects
- 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, Complement C7 immunology, Complement C7 metabolism, Complement Membrane Attack Complex metabolism, Endothelial Cells immunology, Vasculitis immunology, Vasculitis metabolism
- 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.
- Published
- 2009
- Full Text
- View/download PDF
238. Quantum phase transition in the sub-Ohmic spin-boson model: quantum Monte Carlo study with a continuous imaginary time cluster algorithm.
- Author
-
Winter A, Rieger H, Vojta M, and Bulla R
- Abstract
A continuous time cluster algorithm for two-level systems coupled to a dissipative bosonic bath is presented and applied to the sub-Ohmic spin-boson model. When the power s of the spectral function Jomega proportional, variant omegas is smaller than 1/2, the critical exponents are found to be classical, mean-field like. Potential sources for the discrepancy with recent renormalization group predictions are traced back to the effect of a dangerously irrelevant variable.
- Published
- 2009
- Full Text
- View/download PDF
239. Numerical renormalization group calculation of near-gap peaks in spectral functions of the Anderson model with superconducting leads.
- Author
-
Hecht T, Weichselbaum A, von Delft J, and Bulla R
- Abstract
We use the numerical renormalization group method (NRG) to investigate a single-impurity Anderson model with a coupling of the impurity to a superconducting host. Analysis of the energy flow shows that, contrary to previous belief, NRG iterations can be performed up to a large number of sites, corresponding to energy differences far below the superconducting gap Δ. This allows us to calculate the impurity spectral function A(ω) very accurately for frequencies |ω|∼Δ, and to resolve, in a certain parameter regime, sharp peaks in A(ω) close to the gap edge.
- Published
- 2008
- Full Text
- View/download PDF
240. Decidual endothelial cells express surface-bound C1q as a molecular bridge between endovascular trophoblast and decidual endothelium.
- Author
-
Bulla R, Agostinis C, Bossi F, Rizzi L, Debeus A, Tripodo C, Radillo O, De Seta F, Ghebrehiwet B, and Tedesco F
- Subjects
- Blood Vessels embryology, Blood Vessels immunology, Blood Vessels metabolism, Cell Adhesion, Complement C1q immunology, Decidua cytology, Decidua metabolism, Endothelial Cells cytology, Endothelial Cells metabolism, Female, Humans, Membrane Glycoproteins immunology, Pregnancy, Receptors, Complement immunology, Trophoblasts cytology, Trophoblasts immunology, Blood Vessels cytology, Complement C1q metabolism, Decidua immunology, Endothelial Cells immunology, Membrane Glycoproteins metabolism, Receptors, Complement metabolism, Trophoblasts physiology
- Abstract
This study was prompted by the observation that decidual endothelial cells (DECs), unlike endothelial cells (ECs) of blood vessels in normal skin, kidney glomeruli and brain, express surface-bound C1q in physiologic pregnancy. This finding was unexpected, because deposits of C1q are usually observed in pathologic conditions and are associated with complement activation. In the case of DECs, we failed to detect immunoglobulins and C4 co-localized with C1q on the cell surface. Surprisingly, DECs expressed mRNA for the three chains of C1q and secreted detectable level of this component in serum-free medium. The ability to synthesize C1q is acquired by DECs during pregnancy and is not shared by ECs obtained from endometrium and from other sources. Cell-associated C1q has a molecular weight similar to that of secreted C1q and is released from DECs following treatment with heparinase or incubation at low pH. This suggests that C1q binds to DECs and it is not constitutively expressed on the cell surface. C1q is localized at contact sites between endovascular trophoblast and DECs and acts as an intercellular molecular bridge because adhesion of endovascular trophoblast to DECs was inhibited by antibodies to C1q and to a receptor recognizing its globular portion expressed on trophoblast.
- Published
- 2008
- Full Text
- View/download PDF
241. Recruitment of circulating NK cells through decidual tissues: a possible mechanism controlling NK cell accumulation in the uterus during early pregnancy.
- Author
-
Carlino C, Stabile H, Morrone S, Bulla R, Soriani A, Agostinis C, Bossi F, Mocci C, Sarazani F, Tedesco F, Santoni A, and Gismondi A
- Subjects
- Cell Movement, Cells, Cultured, Chemokines genetics, Coculture Techniques, Decidua metabolism, Female, Gene Expression Profiling, Humans, Killer Cells, Natural metabolism, Male, Pregnancy, RNA, Messenger genetics, Receptors, Chemokine genetics, Stromal Cells metabolism, Time Factors, Decidua cytology, Killer Cells, Natural cytology
- Abstract
During early pregnancy, uterine mucosa decidualization is accompanied by a drastic enrichment of CD56(high)CD16(-) natural killer (NK) cells. Decidual NK (dNK) cells differ from peripheral blood NK (pbNK) cells in several ways, but their origin is still unclear. Our results demonstrate that chemokines present in the uterus can support pbNK cell migration through human endothelial and stromal decidual cells. Notably, we observed that pregnant women's pbNK cells are endowed with higher migratory ability compared with nonpregnant women's or male donors' pbNK cells. Moreover, NK cell migration through decidual stromal cells was increased when progesterone-cultured stromal cells were used as substrate, and this correlated with the ability of progesterone to up-regulate stromal cell chemokine expression. Furthermore, we demonstrate that dNK cells migrate through stromal cells using a distinct pattern of chemokines. Finally, we found that pbNK cells acquire a chemokine receptor pattern similar to that of dNK cells when they contact decidual stromal cells. Collectively these results strongly suggest that pbNK cell recruitment to the uterus contributes to the accumulation of NK cells during early pregnancy; that progesterone plays a crucial role in this event; and that pbNK cells undergo reprogramming of their chemokine receptor profile once exposed to uterine microenvironment.
- Published
- 2008
- Full Text
- View/download PDF
242. Endothelial cells are a target of both complement and kinin system.
- Author
-
Bossi F, Bulla R, and Tedesco F
- Subjects
- Animals, Complement C1 Inhibitor Protein physiology, Humans, Membrane Glycoproteins physiology, Receptors, Complement physiology, Complement System Proteins physiology, Endothelial Cells physiology, Kinins physiology
- Abstract
The endothelium is a continuous physical barrier that regulates coagulation and selective passage of soluble molecules and circulating cells through the vessel wall into the tissue. Endothelial cells may contact components of the complement, the kinin and the coagulation systems and their functional activity can be influenced by these interactions. Therefore, complement activation products can induce pro-inflammatory and pro-coagulant responses by endothelial cells. Moreover complement can regulate the release of kinins on the endothelial cell surface influencing the vascular leakage. The aim of this review is to discuss the complex interplay that can be established among the endothelium, the complement proteins or its activation products, and the kinin system.
- Published
- 2008
- Full Text
- View/download PDF
243. Zero-temperature magnetic transition in an easy-axis Kondo lattice model.
- Author
-
Zhu JX, Kirchner S, Bulla R, and Si Q
- Abstract
We address the quantum transition of a spin-1/2 antiferromagnetic Kondo lattice model with an easy-axis anisotropy using the extended dynamical mean field theory. We derive results in real frequency by using the bosonic numerical renormalization group (BNRG) method and compare them with quantum Monte Carlo results in Matsubara frequency. The BNRG results show a logarithmic divergence in the critical local spin susceptibility, signaling a destruction of Kondo screening. The T=0 transition is consistent with being second order. The BNRG results also display some subtle features; we identify their origin and suggest means for further microscopic studies.
- Published
- 2007
- Full Text
- View/download PDF
244. Equilibrium and nonequilibrium dynamics of the sub-Ohmic spin-boson model.
- Author
-
Anders FB, Bulla R, and Vojta M
- Abstract
Employing the nonperturbative numerical renormalization group method, we study the dynamics of the spin-boson model, which describes a two-level system coupled to a bosonic bath with a spectral density J(omega) proportional to omega(s). We show that, in contrast with the case of Ohmic damping, the delocalized phase of the sub-Ohmic model cannot be characterized by a single energy scale only, due to the presence of a nontrivial quantum phase transition. In the strongly sub-Ohmic regime, s<<1, weakly damped coherent oscillations on short time scales are possible even in the localized phase--this is of crucial relevance, e.g., for qubits subject to electromagnetic noise.
- Published
- 2007
- Full Text
- View/download PDF
245. Protection against inflammation- and autoantibody-caused fetal loss by the chemokine decoy receptor D6.
- Author
-
Martinez de la Torre Y, Buracchi C, Borroni EM, Dupor J, Bonecchi R, Nebuloni M, Pasqualini F, Doni A, Lauri E, Agostinis C, Bulla R, Cook DN, Haribabu B, Meroni P, Rukavina D, Vago L, Tedesco F, Vecchi A, Lira SA, Locati M, and Mantovani A
- Subjects
- Animals, Chemokines, CC analysis, Female, Leukocytes, Lipopolysaccharides pharmacology, Mice, Mice, Knockout, Placenta chemistry, Pregnancy, Receptors, CCR10, Trophoblasts chemistry, Chemokine Receptor D6, Antibodies, Antiphospholipid pharmacology, Fetal Death etiology, Inflammation complications, Receptors, Chemokine physiology
- Abstract
Fetal loss in animals and humans is frequently associated with inflammatory conditions. D6 is a promiscuous chemokine receptor with decoy function, expressed in lymphatic endothelium, that recognizes and targets to degradation most inflammatory CC chemokines. Here, we report that D6 is expressed in placenta on invading extravillous trophoblasts and on the apical side of syncytiotrophoblast cells, at the very interface between maternal blood and fetus. Exposure of D6-/- pregnant mice to LPS or antiphospholipid autoantibodies results in higher levels of inflammatory CC chemokines and increased leukocyte infiltrate in placenta, causing an increased rate of fetal loss, which is prevented by blocking inflammatory chemokines. Thus, the promiscuous decoy receptor for inflammatory CC chemokines D6 plays a nonredundant role in the protection against fetal loss caused by systemic inflammation and antiphospholipid antibodies.
- Published
- 2007
- Full Text
- View/download PDF
246. EMILIN1 represents a major stromal element determining human trophoblast invasion of the uterine wall.
- Author
-
Spessotto P, Bulla R, Danussi C, Radillo O, Cervi M, Monami G, Bossi F, Tedesco F, Doliana R, and Colombatti A
- Subjects
- Cell Adhesion, Chorionic Villi metabolism, Decidua cytology, Decidua metabolism, Female, Humans, Integrin alpha4beta1 metabolism, Matrix Metalloproteinase Inhibitors, Matrix Metalloproteinases, Membrane-Associated metabolism, Membrane Glycoproteins antagonists & inhibitors, Membrane Glycoproteins genetics, Placenta cytology, Placenta metabolism, Pregnancy, Pregnancy Trimester, First, RNA, Small Interfering pharmacology, Stromal Cells metabolism, Cell Movement, Membrane Glycoproteins metabolism, Neoplasm Invasiveness pathology, Stromal Cells pathology, Trophoblasts physiology, Uterus pathology
- Abstract
The detection of EMILIN1, a connective tissue glycoprotein associated with elastic fibers, at the level of the ectoplacental cone and trophoblast giant cells of developing mouse embryos (Braghetta et al., 2002) favored the idea of a structural as well as a functional role for this protein in the process of placentation. During the establishment of human placenta, a highly migratory subpopulation of extravillous trophoblasts (EVT), originating from anchoring chorionic villi, penetrate and invade the uterine wall. In this study we show that EMILIN1, produced by decidual stromal and smooth muscle uterine cells, is expressed in the stroma and in some instances as a gradient of increasing concentration in the perivascular region of modified vessels. This distribution pattern is consistent with the haptotactic directional migration observed in in vitro functional studies of freshly isolated EVT and of the immortalized HTR-8/SVneo cell line of trophoblasts. Function-blocking monoclonal antibodies against alpha4-integrin chain and against EMILIN1 as well as the use of EMILIN1-specific short interfering RNA confirmed that trophoblasts interact with EMILIN1 and/or its functional gC1q1 domain via alpha4beta1 integrin. Finally, membrane type I-matrix metalloproteinase (MT1-MMP) and MMP-2 were upregulated in co-cultures of trophoblast cells and stromal cells, suggesting a contributing role in the haptotactic process towards EMILIN1.
- Published
- 2006
- Full Text
- View/download PDF
247. Dissipative exciton transfer in donor-bridge-acceptor systems: numerical renormalization group calculation of equilibrium properties.
- Author
-
Tornow S, Tong NH, and Bulla R
- Abstract
We present a detailed model study of exciton transfer processes in donor-bridge-acceptor (DBA) systems. Using a model which includes the intermolecular Coulomb interaction and the coupling to a dissipative environment we calculate the phase diagram, the absorption spectrum as well as dynamic equilibrium properties with the numerical renormalization group. This method is non-perturbative and therefore allows one to cover the full parameter space, especially the case when the intermolecular Coulomb interaction is of the same order as the coupling to the environment and perturbation theory cannot be applied. For DBA systems with up to six sites we found a transition to the localized phase (self-trapping) depending on the coupling to the dissipative environment. We discuss various criteria which favour delocalized exciton transfer.
- Published
- 2006
- Full Text
- View/download PDF
248. The complement system in the pathophysiology of pregnancy.
- Author
-
Girardi G, Bulla R, Salmon JE, and Tedesco F
- Subjects
- Animals, Antibodies, Antiphospholipid immunology, Complement Activating Enzymes genetics, Complement Activating Enzymes immunology, Complement Activation genetics, Complement Inactivator Proteins genetics, Female, Fetal Death genetics, Fetal Death immunology, Fetal Death pathology, Fetomaternal Transfusion genetics, Fetomaternal Transfusion pathology, Humans, Maternal-Fetal Exchange genetics, Mice, Mice, Knockout, Pregnancy, Receptors, Complement deficiency, Receptors, Complement immunology, Receptors, Complement 3b, Trophoblasts pathology, Complement Activation immunology, Complement Inactivator Proteins immunology, Fetomaternal Transfusion immunology, Isoantibodies immunology, Maternal-Fetal Exchange immunology, Trophoblasts immunology
- Abstract
A fully active complement system deriving from the maternal circulation as well as from local production by various cell source is present in the placenta. The role of this system at the placental level, as in any other tissue in the body, is to protect both the fetus and the mother against infectious and other toxic agents. As fetal tissues are semi-allogeneic and alloantibodies commonly develop in the mother, the placenta is potentially subject to complement-mediated immune attack at the feto-maternal interface with the potential risk of fetal loss. Uncontrolled complement activation is prevented in successful pregnancy by the three regulatory proteins DAF, MCP and CD59 positioned on the surface of trophoblasts. The critical role played by these complement regulators is supported by the embryonic lethality observed in mice deficient in the complement regulator Crry. Excessive complement activation in the placenta places the fetus at risk for growth restriction or death. The role played by the complement system in the fetal damage induced by anti-phospholipid antibodies in a mouse model will be examined.
- Published
- 2006
- Full Text
- View/download PDF
249. Thrombus formation induced by antibodies to beta2-glycoprotein I is complement dependent and requires a priming factor.
- Author
-
Fischetti F, Durigutto P, Pellis V, Debeus A, Macor P, Bulla R, Bossi F, Ziller F, Sblattero D, Meroni P, and Tedesco F
- Subjects
- Animals, Antibodies, Antiphospholipid immunology, Antiphospholipid Syndrome immunology, Antiphospholipid Syndrome metabolism, Autoantibodies chemistry, Blood Coagulation, Complement C3 metabolism, Complement C5 metabolism, Complement C9 metabolism, Complement System Proteins, Endothelium, Vascular cytology, Escherichia coli metabolism, Female, Fibrinogen chemistry, Fibrinogen metabolism, Glycoproteins chemistry, Humans, Immunoglobulin G chemistry, Inflammation, Lipopolysaccharides, Male, Microscopy, Fluorescence, Protein Structure, Tertiary, Rats, Rats, Wistar, Thrombosis blood, Time Factors, Venous Thrombosis blood, beta 2-Glycoprotein I, Glycoproteins immunology, Thrombosis metabolism
- Abstract
We monitored the number of intravascular platelet-leukocyte aggregates (PLAs) and thrombotic occlusions (TOs) by intravascular microscopy in the mesentery of rats receiving antiphospholipid (aPL) immunoglobulin G (IgG) purified from the sera of patients with antiphospholipid syndrome. aPL IgG had no procoagulant effect, but it caused rapid endothelial deposition of fibrinogen, followed by PLA and TO in rats receiving an intraperitoneal injection of bacterial lipopolysaccharide 3 hours before IgG infusion. Anti-beta2-glycoprotein I-depleted aPL IgG failed to induce PLAs and TOs. C3 and C9 colocalized with aPL IgG on the mesenteric vessels. The number of PLAs and TOs was markedly reduced in C6-deficient rats and in animals treated with anti-C5 miniantibody, suggesting the contribution of the terminal complement (C) complex to the aPL antibody-mediated intravascular thrombosis. In conclusion, our data indicate that antibodies to beta2-glycoprotein I trigger coagulation subsequent to a priming proinflammatory factor and that the terminal C complex is the main mediator of the coagulation process.
- Published
- 2005
- Full Text
- View/download PDF
250. Controlling complement resistance in cancer by using human monoclonal antibodies that neutralize complement-regulatory proteins CD55 and CD59.
- Author
-
Ziller F, Macor P, Bulla R, Sblattero D, Marzari R, and Tedesco F
- Subjects
- Antibodies, Monoclonal genetics, Antibodies, Monoclonal, Murine-Derived, Antineoplastic Agents pharmacology, CD55 Antigens physiology, CD59 Antigens physiology, Complement Inactivator Proteins physiology, Cytotoxicity, Immunologic, Humans, Immunoglobulin Fragments genetics, Immunoglobulin Variable Region genetics, Lymphoma, B-Cell drug therapy, Lymphoma, B-Cell immunology, Neoplasms metabolism, Rituximab, Transfection, Antibodies, Monoclonal pharmacology, CD55 Antigens immunology, CD55 Antigens metabolism, CD59 Antigens immunology, CD59 Antigens metabolism, Complement Inactivator Proteins immunology, Complement Inactivator Proteins metabolism, Neoplasms immunology
- Abstract
Expression of the complement-regulatory proteins (CRP) CD46, CD55 and CD59 represents a strategy used by tumor cells to evade complement-dependent cell cytoxicity stimulated by monoclonal antibodies. We have isolated two single-chain variable fragments (scFv) to CD55 and CD59 from a human phage-display library and from these scFv we have produced two miniantibodies (MB), MB-55 (against CD55) and MB-59 (against CD59), containing the human hinge-CH2-CH3 domains of IgG1. The specificity of the two MB for the corresponding CRP was assessed by ELISA using purified CD46, CD55 and CD59. MB-55 and MB-59 neutralized the inhibitory activity of CD55 and CD59, respectively, restoring the complement-mediated lysis of sheep and guinea pig erythrocytes that was otherwise inhibited by the two CRP. FACS analysis revealed binding of MB-55 and MB-59 to the lymphoma cell line Karpas 422. The two MB induced a two-fold increase in the complement-dependent killing of these cells stimulated by Rituximab, a chimeric anti-CD20 monoclonal antibody. Transfection of HEK293T cells with vectors encoding MB-55 or MB-59 markedly reduced the expression of CD55 and CD59. We conclude that the human antibodies MB-55 and MB-59 may represent a therapeutic tool to increase the complement-dependent killing activity of Rituximab in the treatment of non-Hodgkin's lymphoma.
- Published
- 2005
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.