%0 Journal Article %J Cell Death Differ %D 2018 %T Cell-autonomous and cell non-autonomous downregulation of tumor suppressor DAB2IP by microRNA-149-3p promotes aggressiveness of cancer cells. %A Bellazzo, Arianna %A Di Minin, Giulio %A Valentino, Elena %A Sicari, Daria %A Torre, Denis %A Marchionni, Luigi %A Serpi, Federica %A Stadler, Michael B %A Taverna, Daniela %A Zuccolotto, Gaia %A Montagner, Isabella Monia %A Rosato, Antonio %A Tonon, Federica %A Zennaro, Cristina %A Agostinis, Chiara %A Bulla, Roberta %A Mano, Miguel %A Del Sal, Giannino %A Collavin, Licio %X

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.

%B Cell Death Differ %V 25 %P 1224-1238 %8 2018 Jul %G eng %N 7 %1 http://www.ncbi.nlm.nih.gov/pubmed/29568059?dopt=Abstract %R 10.1038/s41418-018-0088-5 %0 Journal Article %J Front Immunol %D 2018 %T Pathological Significance and Prognostic Value of Surfactant Protein D in Cancer. %A Mangogna, Alessandro %A Belmonte, Beatrice %A Agostinis, Chiara %A Ricci, Giuseppe %A Gulino, Alessandro %A Ferrara, Ines %A Zanconati, Fabrizio %A Tripodo, Claudio %A Romano, Federico %A Kishore, Uday %A Bulla, Roberta %X

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 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 p53 pathway. Recently, SP-D has been shown to suppress lung cancer progression 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 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 infiltrating immune cells.

%B Front Immunol %V 9 %P 1748 %8 2018 %G eng %1 http://www.ncbi.nlm.nih.gov/pubmed/30127783?dopt=Abstract %R 10.3389/fimmu.2018.01748 %0 Journal Article %J Am J Reprod Immunol %D 2018 %T Pre-eclampsia affects procalcitonin production in placental tissue. %A Agostinis, Chiara %A Rami, Damiano %A Zacchi, Paola %A Bossi, Fleur %A Stampalija, Tamara %A Mangogna, Alessandro %A Amadio, Leonardo %A Vidergar, Romana %A Vecchi Brumatti, Liza %A Ricci, Giuseppe %A Celeghini, Claudio %A Radillo, Oriano %A Sargent, Ian %A Bulla, Roberta %K Adult %K Calcitonin %K Cohort Studies %K Female %K Humans %K Macrophages %K Placenta %K Pre-Eclampsia %K Pregnancy %K Trophoblasts %K Tumor Necrosis Factor-alpha %K Up-Regulation %K Young Adult %X

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.

%B Am J Reprod Immunol %V 79 %P e12823 %8 2018 04 %G eng %N 4 %1 http://www.ncbi.nlm.nih.gov/pubmed/29427369?dopt=Abstract %R 10.1111/aji.12823 %0 Journal Article %J J Reprod Immunol %D 2017 %T Alternative functions of the complement protein C1q at embryo implantation site. %A Agostinis, Chiara %A Tedesco, Francesco %A Bulla, Roberta %K Animals %K Complement C1q %K Decidua %K Embryo Implantation %K Extracellular Signal-Regulated MAP Kinases %K Female %K Humans %K Integrin alpha4beta1 %K Mice %K Mice, Knockout %K Neovascularization, Physiologic %K Pre-Eclampsia %K Pregnancy %K Trophoblasts %K Vascular Remodeling %X

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.

%B J Reprod Immunol %V 119 %P 74-80 %8 2017 02 %G eng %1 http://www.ncbi.nlm.nih.gov/pubmed/27687635?dopt=Abstract %R 10.1016/j.jri.2016.09.001 %0 Journal Article %J Front Immunol %D 2017 %T Complement Protein C1q Binds to Hyaluronic Acid in the Malignant Pleural Mesothelioma Microenvironment and Promotes Tumor Growth. %A Agostinis, Chiara %A Vidergar, Romana %A Belmonte, Beatrice %A Mangogna, Alessandro %A Amadio, Leonardo %A Geri, Pietro %A Borelli, Violetta %A Zanconati, Fabrizio %A Tedesco, Francesco %A Confalonieri, Marco %A Tripodo, Claudio %A Kishore, Uday %A Bulla, Roberta %X

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) its globular domain. C1q bound to HA was able to induce adhesion and proliferation of mesothelioma cells (MES) 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.

%B Front Immunol %V 8 %P 1559 %8 2017 %G eng %1 http://www.ncbi.nlm.nih.gov/pubmed/29209316?dopt=Abstract %R 10.3389/fimmu.2017.01559 %0 Journal Article %J Nat Commun %D 2016 %T C1q acts in the tumour microenvironment as a cancer-promoting factor independently of complement activation. %A Bulla, Roberta %A Tripodo, Claudio %A Rami, Damiano %A Ling, Guang Sheng %A Agostinis, Chiara %A Guarnotta, Carla %A Zorzet, Sonia %A Durigutto, Paolo %A Botto, Marina %A Tedesco, Francesco %K Animals %K Apoptosis %K Cell Line, Tumor %K Cell Movement %K Cell Proliferation %K Complement Activation %K Complement C1q %K Complement C3 %K Complement C5 %K Humans %K Mice %K Mice, Inbred C57BL %K Mice, Knockout %K Neoplasms %X

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.

%B Nat Commun %V 7 %P 10346 %8 2016 %G eng %1 http://www.ncbi.nlm.nih.gov/pubmed/26831747?dopt=Abstract %R 10.1038/ncomms10346 %0 Journal Article %J Sci Rep %D 2015 %T RelB activation in anti-inflammatory decidual endothelial cells: a master plan to avoid pregnancy failure? %A Masat, Elisa %A Gasparini, Chiara %A Agostinis, Chiara %A Bossi, Fleur %A Radillo, Oriano %A De Seta, Francesco %A Tamassia, Nicola %A Cassatella, Marco A %A Bulla, Roberta %X

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.

%B Sci Rep %V 5 %P 14847 %8 2015 %G eng %1 http://www.ncbi.nlm.nih.gov/pubmed/26463648?dopt=Abstract %R 10.1038/srep14847 %0 Journal Article %J Proc Natl Acad Sci U S A %D 2014 %T C1q as a unique player in angiogenesis with therapeutic implication in wound healing. %A Bossi, Fleur %A Tripodo, Claudio %A Rizzi, Lucia %A Bulla, Roberta %A Agostinis, Chiara %A Guarnotta, Carla %A Munaut, Carine %A Baldassarre, Gustavo %A Papa, Giovanni %A Zorzet, Sonia %A Ghebrehiwet, Berhane %A Ling, Guang Sheng %A Botto, Marina %A Tedesco, Francesco %K Animals %K Cell Proliferation %K Complement C1q %K DNA Primers %K Endothelial Cells %K Enzyme-Linked Immunosorbent Assay %K Human Umbilical Vein Endothelial Cells %K Humans %K Immunoblotting %K Immunohistochemistry %K In Situ Hybridization %K Mice %K Mice, Inbred C57BL %K Mice, Knockout %K Neovascularization, Physiologic %K Rats %K Rats, Wistar %K Real-Time Polymerase Chain Reaction %K Wound Healing %X

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.

%B Proc Natl Acad Sci U S A %V 111 %P 4209-14 %8 2014 Mar 18 %G eng %N 11 %1 http://www.ncbi.nlm.nih.gov/pubmed/24591625?dopt=Abstract %R 10.1073/pnas.1311968111 %0 Journal Article %J Mediators Inflamm %D 2014 %T The first trimester gravid serum regulates procalcitonin expression in human macrophages skewing their phenotype in vitro. %A Rami, Damiano %A La Bianca, Martina %A Agostinis, Chiara %A Zauli, Giorgio %A Radillo, Oriano %A Bulla, Roberta %K Biomarkers %K Calcitonin %K Cells, Cultured %K Chorionic Gonadotropin %K Estradiol %K Female %K Gene Expression Regulation %K Homeostasis %K Humans %K Inflammation %K Macrophages %K Monocytes %K Phenotype %K Pregnancy %K Pregnancy Trimester, First %K Protein Precursors %K Serum %K Up-Regulation %X

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.

%B Mediators Inflamm %V 2014 %P 248963 %8 2014 %G eng %1 http://www.ncbi.nlm.nih.gov/pubmed/24733960?dopt=Abstract %R 10.1155/2014/248963 %0 Journal Article %J Blood %D 2014 %T A non-complement-fixing antibody to β2 glycoprotein I as a novel therapy for antiphospholipid syndrome. %A Agostinis, Chiara %A Durigutto, Paolo %A Sblattero, Daniele %A Borghi, Maria O %A Grossi, Claudia %A Guida, Filomena %A Bulla, Roberta %A Macor, Paolo %A Pregnolato, Francesca %A Meroni, Pier Luigi %A Tedesco, Francesco %K Abortion, Spontaneous %K Animals %K Antibodies, Monoclonal %K Antiphospholipid Syndrome %K Autoantigens %K beta 2-Glycoprotein I %K Complement Activation %K Complement System Proteins %K Human Umbilical Vein Endothelial Cells %K Humans %K Immunoglobulin G %K Male %K Mice %K Protein Binding %K Rats %K Recombinant Proteins %K Single-Chain Antibodies %K Thrombosis %K Trophoblasts %X

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.

%B Blood %V 123 %P 3478-87 %8 2014 May 29 %G eng %N 22 %1 http://www.ncbi.nlm.nih.gov/pubmed/24642748?dopt=Abstract %R 10.1182/blood-2013-11-537704 %0 Journal Article %J J Clin Endocrinol Metab %D 2012 %T Chemerin regulates NK cell accumulation and endothelial cell morphogenesis in the decidua during early pregnancy. %A Carlino, Claudia %A Trotta, Eleonora %A Stabile, Helena %A Morrone, Stefania %A Bulla, Roberta %A Soriani, Alessandra %A Iannitto, Maria Luisa %A Agostinis, Chiara %A Mocci, Carlo %A Minozzi, Massimo %A Aragona, Cesare %A Perniola, Giorgia %A Tedesco, Francesco %A Sozzani, Silvano %A Santoni, Angela %A Gismondi, Angela %K Capillaries %K Cell Movement %K Chemokines %K Decidua %K Endothelial Cells %K Female %K Humans %K Killer Cells, Natural %K MAP Kinase Signaling System %K Neovascularization, Physiologic %K Pregnancy %K Pregnancy Trimester, First %K Receptors, Chemokine %K RNA, Messenger %K Stromal Cells %K Trophoblasts %X

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.

%B J Clin Endocrinol Metab %V 97 %P 3603-12 %8 2012 Oct %G eng %N 10 %1 http://www.ncbi.nlm.nih.gov/pubmed/22791765?dopt=Abstract %R 10.1210/jc.2012-1102 %0 Journal Article %J Clin Dev Immunol %D 2012 %T MBL interferes with endovascular trophoblast invasion in pre-eclampsia. %A Agostinis, Chiara %A Bossi, Fleur %A Masat, Elisa %A Radillo, Oriano %A Tonon, Maddalena %A De Seta, Francesco %A Tedesco, Francesco %A Bulla, Roberta %K Cell Communication %K Decidua %K Endothelial Cells %K Female %K Humans %K Mannose-Binding Lectin %K Pre-Eclampsia %K Pregnancy %K Transendothelial and Transepithelial Migration %K Trophoblasts %X

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.

%B Clin Dev Immunol %V 2012 %P 484321 %8 2012 %G eng %1 http://www.ncbi.nlm.nih.gov/pubmed/22203857?dopt=Abstract %R 10.1155/2012/484321 %0 Journal Article %J Hum Reprod %D 2012 %T Soluble TRAIL is elevated in recurrent miscarriage and inhibits the in vitro adhesion and migration of HTR8 trophoblastic cells. %A Agostinis, Chiara %A Bulla, Roberta %A Tisato, Veronica %A De Seta, Francesco %A Alberico, Salvatore %A Secchiero, Paola %A Zauli, Giorgio %K Abortion, Habitual %K Apoptosis %K Cell Adhesion %K Cell Line %K Cell Movement %K Cells, Cultured %K Enzyme-Linked Immunosorbent Assay %K Female %K Humans %K Pregnancy %K Receptors, TNF-Related Apoptosis-Inducing Ligand %K TNF-Related Apoptosis-Inducing Ligand %K Trophoblasts %X

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.

%B Hum Reprod %V 27 %P 2941-7 %8 2012 Oct %G eng %N 10 %1 http://www.ncbi.nlm.nih.gov/pubmed/22914768?dopt=Abstract %R 10.1093/humrep/des289 %0 Journal Article %J Blood %D 2011 %T In vivo distribution of β2 glycoprotein I under various pathophysiologic conditions. %A Agostinis, Chiara %A Biffi, Stefania %A Garrovo, Chiara %A Durigutto, Paolo %A Lorenzon, Andrea %A Bek, Alpan %A Bulla, Roberta %A Grossi, Claudia %A Borghi, Maria O %A Meroni, Pierluigi %A Tedesco, Francesco %K Animals %K beta 2-Glycoprotein I %K Complement C1q %K Complement C3 %K Complement C9 %K Endothelial Cells %K Endothelium, Vascular %K Female %K Fetal Death %K Humans %K Mice %K Mice, Inbred BALB C %K Pregnancy %K Trophoblasts %K Uterus %X

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.

%B Blood %V 118 %P 4231-8 %8 2011 Oct 13 %G eng %N 15 %1 http://www.ncbi.nlm.nih.gov/pubmed/21791419?dopt=Abstract %R 10.1182/blood-2011-01-333617 %0 Journal Article %J J Immunol %D 2010 %T An alternative role of C1q in cell migration and tissue remodeling: contribution to trophoblast invasion and placental development. %A Agostinis, Chiara %A Bulla, Roberta %A Tripodo, Claudio %A Gismondi, Angela %A Stabile, Helena %A Bossi, Fleur %A Guarnotta, Carla %A Garlanda, Cecilia %A De Seta, Francesco %A Spessotto, Paola %A Santoni, Angela %A Ghebrehiwet, Berhane %A Girardi, Guillermina %A Tedesco, Francesco %K Animals %K Cell Adhesion %K Chemotaxis, Leukocyte %K Complement C1q %K Female %K Humans %K Immunoblotting %K Immunohistochemistry %K Immunoprecipitation %K Mice %K Mice, Inbred C57BL %K Microscopy, Confocal %K Placentation %K Pre-Eclampsia %K Pregnancy %K Reverse Transcriptase Polymerase Chain Reaction %K Trophoblasts %X

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.

%B J Immunol %V 185 %P 4420-9 %8 2010 Oct 1 %G eng %N 7 %1 http://www.ncbi.nlm.nih.gov/pubmed/20810993?dopt=Abstract %R 10.4049/jimmunol.0903215