Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Eduardo J. Villablanca is active.

Publication


Featured researches published by Eduardo J. Villablanca.


Cell | 2012

Gut Immune Maturation Depends on Colonization with a Host-Specific Microbiota

Hachung Chung; Sünje Johanna Pamp; Jonathan A. Hill; Neeraj K. Surana; Sanna M. Edelman; Erin B. Troy; Nicola C. Reading; Eduardo J. Villablanca; Sen Wang; Jorge R. Mora; Yoshinori Umesaki; Diane Mathis; Christophe Benoist; David A. Relman; Dennis L. Kasper

Gut microbial induction of host immune maturation exemplifies host-microbe mutualism. We colonized germ-free (GF) mice with mouse microbiota (MMb) or human microbiota (HMb) to determine whether small intestinal immune maturation depends on a coevolved host-specific microbiota. Gut bacterial numbers and phylum abundance were similar in MMb and HMb mice, but bacterial species differed, especially the Firmicutes. HMb mouse intestines had low levels of CD4(+) and CD8(+) T cells, few proliferating T cells, few dendritic cells, and low antimicrobial peptide expression--all characteristics of GF mice. Rat microbiota also failed to fully expand intestinal T cell numbers in mice. Colonizing GF or HMb mice with mouse-segmented filamentous bacteria (SFB) partially restored T cell numbers, suggesting that SFB and other MMb organisms are required for full immune maturation in mice. Importantly, MMb conferred better protection against Salmonella infection than HMb. A host-specific microbiota appears to be critical for a healthy immune system.


Nature Medicine | 2010

Tumor-mediated liver X receptor-[alpha] activation inhibits CC chemokine receptor-7 expression on dendritic cells and dampens antitumor responses

Eduardo J. Villablanca; Laura Raccosta; Dan Zhou; Raffaella Fontana; Daniela Maggioni; Aurora Negro; Francesca Sanvito; Maurilio Ponzoni; Barbara Valentinis; Marco Bregni; Alessandro Prinetti; Knut R. Steffensen; Sandro Sonnino; Jan Åke Gustafsson; Claudio Doglioni; Claudio Bordignon; Catia Traversari; Vincenzo Russo

Sterol metabolism has recently been linked to innate and adaptive immune responses through liver X receptor (LXR) signaling. Whether products of sterol metabolism interfere with antitumor responses is currently unknown. Dendritic cells (DCs) initiate immune responses, including antitumor activity after their CC chemokine receptor-7 (CCR7)-dependent migration to lymphoid organs. Here we report that human and mouse tumors produce LXR ligands that inhibit CCR7 expression on maturing DCs and, therefore, their migration to lymphoid organs. In agreement with this observation, we detected CD83+CCR7− DCs within human tumors. Mice injected with tumors expressing the LXR ligand–inactivating enzyme sulfotransferase 2B1b (SULT2B1b) successfully controlled tumor growth by regaining DC migration to tumor-draining lymph nodes and by developing overt inflammation within tumors. The control of tumor growth was also observed in chimeric mice transplanted with bone marrow from mice lacking the gene encoding LXR-α (Nr1h3−/− mice) Thus, we show a new mechanism of tumor immunoescape involving products of cholesterol metabolism. The manipulation of this pathway could restore antitumor immunity in individuals with cancer.


Proceedings of the National Academy of Sciences of the United States of America | 2014

Atg16L1 T300A variant decreases selective autophagy resulting in altered cytokine signaling and decreased antibacterial defense.

Kara G. Lassen; Petric Kuballa; Kara L. Conway; Khushbu K. Patel; Christine E. Becker; Joanna M. Peloquin; Eduardo J. Villablanca; Jason M. Norman; Ta-Chiang Liu; Robert J. Heath; Morgan L. Becker; Lola Fagbami; Heiko Horn; Johnathan Mercer; Ömer H. Yilmaz; Jacob D. Jaffe; Alykhan F. Shamji; Atul K. Bhan; Steven A. Carr; Mark J. Daly; Herbert W. Virgin; Stuart L. Schreiber; Thaddeus S. Stappenbeck; Ramnik J. Xavier

Significance Although advances in human genetics have shaped our understanding of many complex diseases, little is known about the mechanism of action of alleles that influence disease. By using mice expressing a Crohn disease (CD)-associated risk polymorphism (Atg16L1 T300A), we show that Atg16L1 T300A-expressing mice demonstrate abnormalities in Paneth cells (similar to patients with the risk polymorphism) and goblet cells. We show that Atg16L1 T300A protein is more susceptible to caspase-mediated cleavage than WT autophagy related 16-like 1 (Atg16L1), resulting in decreased protein stability and effects on antibacterial autophagy and inflammatory cytokine production. We also identify interacting proteins that contribute to autophagy-dependent immune responses. Understanding how ATG16L1 T300A modulates autophagy-dependent immune responses sheds light on the mechanisms that underlie initiation and progression of CD. A coding polymorphism (Thr300Ala) in the essential autophagy gene, autophagy related 16-like 1 (ATG16L1), confers increased risk for the development of Crohn disease, although the mechanisms by which single disease-associated polymorphisms contribute to pathogenesis have been difficult to dissect given that environmental factors likely influence disease initiation in these patients. Here we introduce a knock-in mouse model expressing the Atg16L1 T300A variant. Consistent with the human polymorphism, T300A knock-in mice do not develop spontaneous intestinal inflammation, but exhibit morphological defects in Paneth and goblet cells. Selective autophagy is reduced in multiple cell types from T300A knock-in mice compared with WT mice. The T300A polymorphism significantly increases caspase 3- and caspase 7-mediated cleavage of Atg16L1, resulting in lower levels of full-length Atg16Ll T300A protein. Moreover, Atg16L1 T300A is associated with decreased antibacterial autophagy and increased IL-1β production in primary cells and in vivo. Quantitative proteomics for protein interactors of ATG16L1 identified previously unknown nonoverlapping sets of proteins involved in ATG16L1-dependent antibacterial autophagy or IL-1β production. These findings demonstrate how the T300A polymorphism leads to cell type- and pathway-specific disruptions of selective autophagy and suggest a mechanism by which this polymorphism contributes to disease.


Gastroenterology | 2013

Atg16l1 is required for autophagy in intestinal epithelial cells and protection of mice from Salmonella infection

Kara L. Conway; Petric Kuballa; Joo Hye Song; Khushbu K. Patel; Adam B. Castoreno; Ömer H. Yilmaz; Humberto Jijon; Mei Zhang; Leslie N. Aldrich; Eduardo J. Villablanca; Joanna M. Peloquin; Gautam Goel; In–Ah Lee; Emiko Mizoguchi; Hai Ning Shi; Atul K. Bhan; Stanley Y. Shaw; Stuart L. Schreiber; Herbert W. Virgin; Alykhan F. Shamji; Thaddeus S. Stappenbeck; Hans-Christian Reinecker; Ramnik J. Xavier

BACKGROUND & AIMS Intestinal epithelial cells aid in mucosal defense by providing a physical barrier against entry of pathogenic bacteria and secreting antimicrobial peptides (AMPs). Autophagy is an important component of immune homeostasis. However, little is known about its role in specific cell types during bacterial infection in vivo. We investigated the role of autophagy in the response of intestinal epithelial and antigen-presenting cells to Salmonella infection in mice. METHODS We generated mice deficient in Atg16l1 in epithelial cells (Atg16l1(f/f) × Villin-cre) or CD11c(+) cells (Atg16l1(f/f) × CD11c-cre); these mice were used to assess cell type-specific antibacterial autophagy. All responses were compared with Atg16l1(f/f) mice (controls). Mice were infected with Salmonella enterica serovar typhimurium; cecum and small-intestine tissues were collected for immunofluorescence, histology, and quantitative reverse-transcription polymerase chain reaction analyses of cytokines and AMPs. Modulators of autophagy were screened to evaluate their effects on antibacterial responses in human epithelial cells. RESULTS Autophagy was induced in small intestine and cecum after infection with S typhimurium, and required Atg16l1. S typhimurium colocalized with microtubule-associated protein 1 light chain 3β (Map1lc3b or LC3) in the intestinal epithelium of control mice but not in Atg16l1(f/f) × Villin-cre mice. Atg16l1(f/f) × Villin-cre mice also had fewer Paneth cells and abnormal granule morphology, leading to reduced expression of AMPs. Consistent with these defective immune responses, Atg16l1(f/f) × Villin-cre mice had increased inflammation and systemic translocation of bacteria compared with control mice. In contrast, we observed few differences between Atg16l1(f/f) × CD11c-cre and control mice. Trifluoperazine promoted autophagy and bacterial clearance in HeLa cells; these effects were reduced upon knockdown of ATG16L1. CONCLUSIONS Atg16l1 regulates autophagy in intestinal epithelial cells and is required for bacterial clearance. It also is required to prevent systemic infection of mice with enteric bacteria.


Gastroenterology | 2011

Gut-Tropic T Cells That Express Integrin α4β7 and CCR9 Are Required for Induction of Oral Immune Tolerance in Mice

Barbara Cassani; Eduardo J. Villablanca; Francisco J. Quintana; Paul E. Love; Adam Lacy–Hulbert; William S. Blaner; Tim Sparwasser; Scott B. Snapper; Howard L. Weiner; J. Rodrigo Mora

BACKGROUND & AIMS Induction of oral immune tolerance (OT) blocks proinflammatory responses to orally administered antigens and might be used to treat autoimmune conditions. We investigated whether gut-tropic T cells that express the integrin α4β7 and the chemokine receptor CCR9 are required for OT. METHODS Skin delayed-type hypersensitivity and experimental autoimmune encephalomyelitis were used to monitor OT in mice. To assess the role of receptors that mediate localization of lymphocytes to the gut (gut-homing receptors) in induction of OT, we studied CCR9(-/-) and β7(-/-) mice and also blocked the α4β7 ligand MAdCAM-1 in wild-type mice. We used DEREG and Scurfy mice to assess the role of Foxp3(+) regulatory T cells (Treg) and IL-10(-/-) and IL-10Rβ(-/-) mice to examine the role of interleukin (IL)-10 in induction of OT. RESULTS OT could not be induced in CCR9(-/-) or β7(-/-) mice, or when MAdCAM-1 was blocked in wild-type mice, indicating that gut-homing receptors are required for oral tolerization. Consistent with the role of all-trans retinoic acid in inducing gut-homing T cells, OT could not be induced in mice depleted of vitamin A. OT was rescued in CCR9(-/-) mice following adoptive transfer of wild-type T cells, but not CCR9(-/-) or β7(-/-) T cells. Gut-homing T cells are therefore necessary and sufficient to induce OT. Wild-type Treg and IL-10 were required to restore OT to CCR9(-/-) mice, indicating that homing and functional differentiation of IL-10-producing Treg in the gut is required for OT. Conversely, transfer of CCR9(-/-) or β7(-/-) T cells to wild-type mice partially inhibited OT. CONCLUSIONS Expression of CCR9 and α4β7 on T cells and their subsequent localization to the gut is required for induction of OT in mice. Therapies designed to block gut-homing receptors might, under some conditions, interfere with normal tolerogenic mechanisms in the intestinal mucosa.


Gastroenterology | 2009

Gut Homing Receptors on CD8 T Cells Are Retinoic Acid Dependent and Not Maintained by Liver Dendritic or Stellate Cells

Bertus Eksteen; J. Rodrigo Mora; Emma L. Haughton; Neil C. Henderson; Laura Lee–Turner; Eduardo J. Villablanca; Stuart M. Curbishley; Alex I. Aspinall; Ulrich H. von Andrian; David H. Adams

BACKGROUND & AIMS Lymphocytes primed by intestinal dendritic cells (DC) express the gut-homing receptors CCR9 and alpha4beta7, which recognize CCL25 and mucosal addressin cell-adhesion molecule-1 in the intestine promoting the development of regional immunity. In mice, imprinting of CCR9 and alpha4beta7 is dependent on retinoic acid during T-cell activation. Tissue specificity is lost in primary sclerosing cholangitis (PSC), an extraintestinal manifestation of inflammatory bowel disease, when ectopic expression of mucosal addressin cell-adhesion molecule-1 and CCL25 in the liver promotes recruitment of CCR9+alpha4beta7+ T cells to the liver. We investigated the processes that control enterohepatic T-cell migration and whether the ability to imprint CCR9 and alpha4beta7 is restricted to intestinal DCs or can under some circumstances be acquired by hepatic DCs in diseases such as PSC. METHODS Human and murine DCs from gut, liver, or portal lymph nodes and hepatic stellate cells were used to activate CD8 T cells. Imprinting of CCR9 and alpha4beta7 and functional migration responses were determined. Crossover activation protocols assessed plasticity of gut homing. RESULTS Activation by gut DCs imprinted high levels of functional CCR9 and alpha4beta7 on naïve CD8 T cells, whereas hepatic DCs and stellate cells proved inferior. Imprinting was RA dependent and demonstrated plasticity. CONCLUSIONS Imprinting and plasticity of gut-homing human CD8 T cells requires primary activation or reactivation by gut DCs and is retinoic acid dependent. The inability of liver DCs to imprint gut tropism implies that alpha4beta7+CCR9+ T cell that infiltrate the liver in PSC are primed in the gut.


Journal of Experimental Medicine | 2013

The oxysterol–CXCR2 axis plays a key role in the recruitment of tumor-promoting neutrophils

Laura Raccosta; Raffaella Fontana; Daniela Maggioni; Claudia Lanterna; Eduardo J. Villablanca; Aida Paniccia; Andrea Musumeci; Elena Chiricozzi; Maria Letizia Trincavelli; Simona Daniele; Claudia Martini; Jan Åke Gustafsson; Claudio Doglioni; Safiyè Gonzalvo Feo; Andrea Leiva; Maria Grazia Ciampa; Laura Mauri; Cristina Sensi; Alessandro Prinetti; Ivano Eberini; J. Rodrigo Mora; Claudio Bordignon; Knut R. Steffensen; Sandro Sonnino; Silvano Sozzani; Catia Traversari; Vincenzo Russo

Tumor-derived oxysterols recruit protumor neutrophils in an LXR-independent, CXCR2-dependent manner, thus favoring tumor growth by promoting neoangiogenesis and immunosuppression.


Gastroenterology | 2011

MyD88 and retinoic acid signaling pathways interact to modulate gastrointestinal activities of dendritic cells.

Eduardo J. Villablanca; Sen Wang; Jaime De Calisto; Daniel Cláudio de Oliveira Gomes; Maureen A. Kane; Joseph L. Napoli; William S. Blaner; Hiroyuki Kagechika; Rune Blomhoff; Mario Rosemblatt; María Rosa Bono; Ulrich H. von Andrian; J. Rodrigo Mora

BACKGROUND & AIMS Gut-associated dendritic cells (DC) metabolize vitamin A into all-trans retinoic acid (RA), which is required to induce lymphocytes to localize to the gastrointestinal tract and promotes the differentiation of Foxp3+ regulatory T cells and IgA antibody-secreting cells. We investigated whether RA functions in a positive-feedback loop in DC to induce its own synthesis. METHODS We measured levels of retinoids in intestinal tissues from mice and assessed the role of RA in the functional specialization of gut-associated DC in cell cultures and mice. We used pharmacologic antagonists to determine the signaling pathways involved in regulation of DC and used MyD88-/- mice to determine the contribution of Toll-like receptor signaling in RA-mediated effects on DC. RESULTS The concentration of retinoids decreased in a proximal-to-distal gradient along the intestine, which correlated with the activity of gut-specific DC. Importantly, RA regulated the ability of gut-associated DC to produce RA, induce T cells to localize to the gastrointestinal tract, and generate regulatory T cells and IgA-secreting cells. RA was sufficient to induce its own production by extraintestinal DC in vitro and in vivo. RA-mediated regulation of DC required signaling through the mitogen-activated protein kinase signaling pathway and unexpectedly required MyD88, which is conventionally associated with Toll-like receptor, interleukin-1, and interleukin-18 signaling. CONCLUSIONS RA is necessary and sufficient to induce DC to regulate T-cell localization to the gastrointestinal tract and IgA secretion. Our findings also indicate crosstalk between the RA receptor and MyD88-dependent Toll-like receptor signaling pathways.


European Journal of Immunology | 2008

A two-step model for Langerhans cell migration to skin-draining LN

Eduardo J. Villablanca; Jorge R. Mora

Although the role of Langerhans cells (LC) in skin immune responses is still a matter of debate, it is known that LC require the chemokine receptor CCR7 for migrating to skin‐draining LN. A report in the current issue of the European Journal of Immunology unfolds some of the intricacies of LC migration, showing that LC need CXCR4, but not CCR7, for their migration from the epidermis to the dermis. Thus, LC migration to skin‐draining LN occurs in two distinct phases: a first step from the epidermis to the dermis regulated by CXCR4 and a second CCR7‐dependent step from the dermis to LN. Here we discuss the potential implications of this new two‐step LC migration paradigm.


Journal of Immunology | 2011

MyD88-dependent TLR1/2 signals educate dendritic cells with gut-specific imprinting properties

Sen Wang; Eduardo J. Villablanca; Jaime De Calisto; Daniel Cláudio de Oliveira Gomes; Deanna D. Nguyen; Emiko Mizoguchi; Jonathan C. Kagan; Hans-Christian Reinecker; Nir Hacohen; Cathryn R. Nagler; Ramnik J. Xavier; Bartira Rossi-Bergmann; Yi-Bin Chen; Rune Blomhoff; Scott B. Snapper; J. Rodrigo Mora

Gut-associated dendritic cells (DC) synthesize all-trans retinoic acid, which is required for inducing gut-tropic lymphocytes. Gut-associated DC from MyD88−/− mice, which lack most TLR signals, expressed low levels of retinal dehydrogenases (critical enzymes for all-trans retinoic acid biosynthesis) and were significantly impaired in their ability to induce gut-homing T cells. Pretreatment of extraintestinal DC with a TLR1/2 agonist was sufficient to induce retinal dehydrogenases and to confer these DC with the capacity to induce gut-homing lymphocytes via a mechanism dependent on MyD88 and JNK/MAPK. Moreover, gut-associated DC from TLR2−/− mice, or from mice in which JNK was pharmacologically blocked, were impaired in their education to imprint gut-homing T cells, which correlated with a decreased induction of gut-tropic T cells in TLR2−/− mice upon immunization. Thus, MyD88-dependent TLR2 signals are necessary and sufficient to educate DC with gut-specific imprinting properties and contribute in vivo to the generation of gut-tropic T cells.

Collaboration


Dive into the Eduardo J. Villablanca's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Vincenzo Russo

Vita-Salute San Raffaele University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge