Network


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

Hotspot


Dive into the research topics where Giovanna Leoni is active.

Publication


Featured researches published by Giovanna Leoni.


Journal of Clinical Investigation | 2013

Annexin A1, formyl peptide receptor, and NOX1 orchestrate epithelial repair

Giovanna Leoni; Ashfaqul Alam; Philipp-Alexander Neumann; J. David Lambeth; Guangjie Cheng; James McCoy; Roland S. Hilgarth; Kousik Kundu; Niren Murthy; Dennis H. M. Kusters; Chris Reutelingsperger; Mauro Perretti; Charles A. Parkos; Andrew S. Neish; Asma Nusrat

N-formyl peptide receptors (FPRs) are critical regulators of host defense in phagocytes and are also expressed in epithelia. FPR signaling and function have been extensively studied in phagocytes, yet their functional biology in epithelia is poorly understood. We describe a novel intestinal epithelial FPR signaling pathway that is activated by an endogenous FPR ligand, annexin A1 (ANXA1), and its cleavage product Ac2-26, which mediate activation of ROS by an epithelial NADPH oxidase, NOX1. We show that epithelial cell migration was regulated by this signaling cascade through oxidative inactivation of the regulatory phosphatases PTEN and PTP-PEST, with consequent activation of focal adhesion kinase (FAK) and paxillin. In vivo studies using intestinal epithelial specific Nox1(-/-IEC) and AnxA1(-/-) mice demonstrated defects in intestinal mucosal wound repair, while systemic administration of ANXA1 promoted wound recovery in a NOX1-dependent fashion. Additionally, increased ANXA1 expression was observed in the intestinal epithelium and infiltrating leukocytes in the mucosa of ulcerative colitis patients compared with normal intestinal mucosa. Our findings delineate a novel epithelial FPR1/NOX1-dependent redox signaling pathway that promotes mucosal wound repair.


Journal of Clinical Investigation | 2015

Annexin A1-containing extracellular vesicles and polymeric nanoparticles promote epithelial wound repair.

Giovanna Leoni; Philipp-Alexander Neumann; Nazila Kamaly; Miguel Quiros; Hikaru Nishio; Hefin R. Jones; Ronen Sumagin; Roland S. Hilgarth; Ashfaqul Alam; Gabrielle Fredman; Ioannis Argyris; Emile Rijcken; Dennis H. M. Kusters; Chris Reutelingsperger; Mauro Perretti; Charles A. Parkos; Omid C. Farokhzad; Andrew S. Neish; Asma Nusrat

Epithelial restitution is an essential process that is required to repair barrier function at mucosal surfaces following injury. Prolonged breaches in epithelial barrier function result in inflammation and further damage; therefore, a better understanding of the epithelial restitution process has potential for improving the development of therapeutics. In this work, we demonstrate that endogenous annexin A1 (ANXA1) is released as a component of extracellular vesicles (EVs) derived from intestinal epithelial cells, and these ANXA1-containing EVs activate wound repair circuits. Compared with healthy controls, patients with active inflammatory bowel disease had elevated levels of secreted ANXA1-containing EVs in sera, indicating that ANXA1-containing EVs are systemically distributed in response to the inflammatory process and could potentially serve as a biomarker of intestinal mucosal inflammation. Local intestinal delivery of an exogenous ANXA1 mimetic peptide (Ac2-26) encapsulated within targeted polymeric nanoparticles (Ac2-26 Col IV NPs) accelerated healing of murine colonic wounds after biopsy-induced injury. Moreover, one-time systemic administration of Ac2-26 Col IV NPs accelerated recovery following experimentally induced colitis. Together, our results suggest that local delivery of proresolving peptides encapsulated within nanoparticles may represent a potential therapeutic strategy for clinical situations characterized by chronic mucosal injury, such as is seen in patients with IBD.


Mucosal Immunology | 2014

Redox signaling regulates commensal-mediated mucosal homeostasis and restitution and requires formyl peptide receptor 1

Ashfaqul Alam; Giovanna Leoni; Christy Wentworth; J M Kwal; Huixia Wu; Courtney S. Ardita; Phillip A. Swanson; J D Lambeth; Rheinallt Jones; Asma Nusrat; Andrew S. Neish

The mammalian gut microbiota is essential for normal intestinal development, renewal, and repair. Injury to the intestinal mucosa can occur with infection, surgical trauma, and in idiopathic inflammatory bowel disease. Repair of mucosal injury, termed restitution, as well as restoration of intestinal homeostasis involves induced and coordinated proliferation and migration of intestinal epithelial cells. N-formyl peptide receptors (FPRs) are widely expressed pattern recognition receptors that can specifically bind and induce responses to host-derived and bacterial peptides and small molecules. Here we report that specific members of the gut microbiota stimulate FPR1 on intestinal epithelial cells to generate reactive oxygen species via enterocyte NADPH oxidase 1 (NOX1), causing rapid phosphorylation of focal adhesion kinase (FAK) and extracellular signal-regulated kinase mitogen-activated protein kinase. These events stimulate migration and proliferation of enterocytes adjacent to colonic wounds. Taken together, these findings identify a novel role of FPR1 as pattern recognition receptors for perceiving the enteric microbiota that promotes repair of mucosal wounds via generation of reactive oxygen species from the enterocyte NOX1.


Molecular Biology of the Cell | 2013

JAM-A associates with ZO-2, afadin, and PDZ-GEF1 to activate Rap2c and regulate epithelial barrier function

Ana C. Monteiro; Ronen Sumagin; Carl R. Rankin; Giovanna Leoni; Michael J. Mina; Dirk M. Reiter; Thilo Stehle; Terence S. Dermody; Stacy A. Schaefer; Randy A. Hall; Asma Nusrat; Charles A. Parkos

Intestinal barrier function is regulated by epithelial tight junctions, structures that control paracellular permeability. JAM-A regulates epithelial permeability through association with ZO-2, afadin, and PDZ-GEF1 to activate Rap2c and control contraction of the apical cytoskeleton.


Circulation Research | 2015

Annexin A1 Counteracts Chemokine-Induced Arterial Myeloid Cell Recruitment

Maik Drechsler; Renske de Jong; Jan Rossaint; Joana R. Viola; Giovanna Leoni; Ji Ming Wang; Jochen Grommes; Rabea Hinkel; Christian Kupatt; Christian Weber; Yvonne Döring; Alexander Zarbock; Oliver Soehnlein

RATIONALE Chemokine-controlled arterial leukocyte recruitment is a crucial process in atherosclerosis. Formyl peptide receptor 2 (FPR2) is a chemoattractant receptor that recognizes proinflammatory and proresolving ligands. The contribution of FPR2 and its proresolving ligand annexin A1 to atherosclerotic lesion formation is largely undefined. OBJECTIVE Because of the ambivalence of FPR2 ligands, we here investigate the role of FPR2 and its resolving ligand annexin A1 in atherogenesis. METHODS AND RESULTS Deletion of FPR2 or its ligand annexin A1 enhances atherosclerotic lesion formation, arterial myeloid cell adhesion, and recruitment. Mechanistically, we identify annexin A1 as an endogenous inhibitor of integrin activation evoked by the chemokines CCL5, CCL2, and CXCL1. Specifically, the annexin A1 fragment Ac2-26 counteracts conformational activation and clustering of integrins on myeloid cells evoked by CCL5, CCL2, and CXCL1 through inhibiting activation of the small GTPase Rap1. In vivo administration of Ac2-26 largely diminishes arterial recruitment of myeloid cells in a FPR2-dependent fashion. This effect is also observed in the presence of selective antagonists to CCR5, CCR2, or CXCR2, whereas Ac2-26 was without effect when all 3 chemokine receptors were antagonized simultaneously. Finally, repeated treatment with Ac2-26 reduces atherosclerotic lesion sizes and lesional macrophage accumulation. CONCLUSIONS Instructing the annexin A1-FPR2 axis harbors a novel approach to target arterial leukocyte recruitment. With the ability of Ac2-26 to counteract integrin activation exerted by various chemokines, delivery of Ac2-26 may be superior in inhibition of arterial leukocyte recruitment when compared with blocking individual chemokine receptors.


The FASEB Journal | 2008

Inflamed phenotype of the mesenteric microcirculation of melanocortin type 3 receptor-null mice after ischemia-reperfusion

Giovanna Leoni; Hetal B. Patel; André L.F. Sampaio; Felicity N. E. Gavins; Joanne F. Murray; Paolo Grieco; Stephen J. Getting; Mauro Perretti

The existence of anti‐inflammatory circuits centered on melanocortin receptors (MCRs) has been supported by the inhibitory properties displayed by melanocortin peptides in models of inflammation and tissue injury. Here we addressed the pathophysiological effect that one MCR, MCR type 3 (MC3R), might have on vascular inflammation. After occlusion (35 min) and reopening of the superior mesenteric artery, MC3R‐null mice displayed a higher degree of plasma extravasation (45 min postreperfusion) and cell adhesion and emigration (90 min postreperfusion). These cellular alterations were complemented by higher expression of mesenteric tissue CCL2 and CXCL1 (mRNA and protein) and myeloperoxydase, as compared with wild‐type animals. MC1R and MC3R mRNA and protein were both expressed in the inflamed mesenteric tissue;however, no changes in vascular responses were observed in a mouse colony bearing an inactive MC1R. Pharmacological treatment of animals with a selective MC3R agonist ([D‐Trp] ‐γ‐melanocyte‐stimulating hormone;10 μg i.v.) produced marked attenuation of cell adhesion, emigration, and chemokine generation;such effects were absent in MC3R‐null mice. These new data reveal the existence of a tonic inhibitory signal provided by MC3R in the mesenteric microcirculation of the mouse, acting to down‐regulate cell trafficking and local mediator generation.— Leoni, G., Patel, H. B., Sampaio, A. L. F., Gavins, F. N. E., Murray, J. F., Grieco, P., Getting, S. J., Perretti, M. Inflamed phenotype of the mesenteric microcirculation of melanocortin type 3 receptor‐null mice after ischemia‐reperfusion. FASEB J. 22, 4228–4238 (2008). www.fasebj.org


Nature microbiology | 2016

The microenvironment of injured murine gut elicits a local pro-restitutive microbiota

Ashfaqul Alam; Giovanna Leoni; Miguel Quiros; Huixia Wu; Chirayu Desai; Hikaru Nishio; Rheinallt Jones; Asma Nusrat; Andrew S. Neish

The mammalian intestine houses a complex microbial community, which influences normal epithelial growth and development, and is integral to the repair of damaged intestinal mucosa1–3. Restitution of injured mucosa involves the recruitment of immune cells, epithelial migration and proliferation4,5. Although microenvironmental alterations have been described in wound healing6, a role for extrinsic influences, such as members of the microbiota, has not been reported. Here, we show that a distinct subpopulation of the normal mucosal-associated gut microbiota expands and preferentially colonizes sites of damaged murine mucosa in response to local environmental cues. Our results demonstrate that formyl peptide receptor 1 (FPR1) and neutrophilic NADPH oxidase (NOX2) are required for the rapid depletion of microenvironmental oxygen and compensatory responses, resulting in a dramatic enrichment of an anaerobic bacterial consortium. Furthermore, the dominant member of this wound-mucosa-associated microbiota, Akkermansia muciniphila (an anaerobic, mucinophilic gut symbiont7,8), stimulated proliferation and migration of enterocytes adjacent to the colonic wounds in a process involving FPR1 and intestinal epithelial-cell-specific NOX1-dependent redox signalling. These findings thus demonstrate how wound microenvironments induce the rapid emergence of ‘probiont’ species that contribute to enhanced repair of mucosal wounds. Such microorganisms could be exploited as potential therapeutics.


Journal of Immunology | 2015

Cutting Edge: IL-36 Receptor Promotes Resolution of Intestinal Damage

Oscar Medina-Contreras; Akihito Harusato; Hikaru Nishio; Kyle L. Flannigan; Vu Ngo; Giovanna Leoni; Philipp-Alexander Neumann; Duke Geem; Loukia N. Lili; Ravisankar A. Ramadas; Benoit Chassaing; Andrew T. Gewirtz; Jacob E. Kohlmeier; Charles A. Parkos; Jennifer E. Towne; Asma Nusrat; Timothy L. Denning

IL-1 family members are central mediators of host defense. In this article, we show that the novel IL-1 family member IL-36γ was expressed during experimental colitis and human inflammatory bowel disease. Germ-free mice failed to induce IL-36γ in response to dextran sodium sulfate (DSS)-induced damage, suggesting that gut microbiota are involved in its induction. Surprisingly, IL-36R–deficient (Il1rl2−/−) mice exhibited defective recovery following DSS-induced damage and impaired closure of colonic mucosal biopsy wounds, which coincided with impaired neutrophil accumulation in the wound bed. Failure of Il1rl2−/− mice to recover from DSS-induced damage was associated with a profound reduction in IL-22 expression, particularly by colonic neutrophils. Defective recovery of Il1rl2−/− mice could be rescued by an aryl hydrocarbon receptor agonist, which was sufficient to restore IL-22 expression and promote full recovery from DSS-induced damage. These findings implicate the IL-36/IL-36R axis in the resolution of intestinal mucosal wounds.


Arteriosclerosis, Thrombosis, and Vascular Biology | 2010

Antiallergic Cromones Inhibit Neutrophil Recruitment Onto Vascular Endothelium via Annexin-A1 Mobilization

Samia Yazid; Giovanna Leoni; Stephen J. Getting; Dianne Cooper; Egle Solito; Mauro Perretti; Roderick J. Flower

Objective—To determine whether the inhibitory action of the antiallergic cromone “mast cell stabilizing” drugs on polymorphonuclear leukocyte (PMN) trafficking is mediated through an annexin-A1 (Anx-A1) dependent mechanism. Methods and Results—Intravital microscopy was used to monitor the actions of cromones in the inflamed microcirculation. Reperfusion injury provoked a dramatic increase in adherent and emigrated leukocytes in the mesenteric vascular bed, associated with augmented tissue levels of myeloperoxidase. Nedocromil, 2 to 20 mg/kg, significantly (P<0.05) inhibited cell adhesion and emigration, as well as myeloperoxidase release, in wild-type but not Anx-A1−/− mice. Short pretreatment of human PMNs with nedocromil, 10 nmol/L, inhibited cell adhesion (P<0.05) in the flow chamber assay, and this effect was reversed by specific anti-AnxA1 or a combination of antiformyl peptide receptors 1 and 2, but not irrelevant control, antibodies. Western blotting experiments revealed that cromones stimulate protein kinase C–dependent phosphorylation and release Anx-A1 in human PMNs. Conclusion—We propose a novel mechanism to explain the antiinflammatory actions of cromones on PMN trafficking, an effect that has long puzzled investigators.


Molecular Pharmacology | 2006

[D -Trp8]-γ -melanocyte -stimulating hormone exhibits anti-inflammatory efficacy in mice bearing a nonfunctional MC1 R (recessive yellow e/e mouse)

Stephen J. Getting; Connie W. Lam; Giovanna Leoni; Felicity N. E. Gavins; Paolo Grieco; Mauro Perretti

Two melanocortin receptors (MC1 and MC3R) have been identified as main transducers of the anti-inflammatory effects of natural and synthetic melanocortins. In this study, we have taken advantage of the recent description of the selective MC3R agonist [d-Trp8]-γ-melanocyte-stimulating hormone (MSH) and of the recessive yellow (e/e) mouse, bearing a nonfunctional MC1R, thereby incrementing our knowledge on this topic. Culturing peritoneal macrophages of recessive yellow (e/e) mice with [d-Trp8]-γ-MSH led to accumulation of cAMP, indicating MC3R receptor functionality: this effect was blocked by a neutralizing antibody against MC3R. Likewise, release of the chemokine KC by urate crystals was attenuated by [d-Trp8]-γ-MSH, and this effect was prevented by synthetic [Ac-Nle4-c[Asp5-2′-Nal7,Lys10]α-MSH(4-10)-NH2 (SHU9119)] and natural [agouti-related protein (AGRP)] MC3R antagonists but not by the MC4R antagonist Ac-Cys-Nle-Arg-His-d-2-Nal-Arg-Trp-Cys-NH2 (HS024). Systemic treatment of mice with [d-Trp8]-γ-MSH inhibited KC release and polymorphonuclear cell accumulation elicited by urate crystals in the murine peritoneal cavity. SHU9119 and AGRP prevented the inhibitory actions of [d-Trp8]-γ-MSH, whereas HS024 was inactive. We also demonstrate here that [d-Trp8]-γ-MSH displays a dual mechanism of action by inducing the anti-inflammatory protein heme-oxygenase 1 (HO-1). Treatment with the HO-1 inhibitor zinc protoporphyrin IX exacerbated the inflammatory response elicited by urate crystals and abrogated the anti-inflammatory effects of [d-Trp8]-γ-MSH. In conclusion, these data support the development of the selective MC3R agonist [d-Trp8]-γ-MSH for the treatment of inflammatory pathologies, based on a dual mechanism of cytokine/chemokine inhibition and induction of the anti-inflammatory protein HO-1.

Collaboration


Dive into the Giovanna Leoni's collaboration.

Top Co-Authors

Avatar

Asma Nusrat

University of Michigan

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Mauro Perretti

Queen Mary University of London

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
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge