Network


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

Hotspot


Dive into the research topics where Carmine Stolfi is active.

Publication


Featured researches published by Carmine Stolfi.


Gastroenterology | 2008

Regulation of Gut Inflammation and Th17 Cell Response by Interleukin-21

Daniele Fina; Massimiliano Sarra; Massimo C. Fantini; Angelamaria Rizzo; Roberta Caruso; Flavio Caprioli; Carmine Stolfi; Iris Cardolini; Marta Dottori; Monica Boirivant; Francesco Pallone; Thomas T. MacDonald; Giovanni Monteleone

BACKGROUND & AIMS Interleukin (IL)-21, a T-cell-derived cytokine, is overproduced in inflammatory bowel diseases (IBD), but its role in the pathogenesis of gut inflammation remains unknown. We here examined whether IL-21 is necessary for the initiation and progress of experimental colitis and whether it regulates specific pathways of inflammation. METHODS Both dextran sulfate sodium colitis and trinitrobenzene sulfonic acid-relapsing colitis were induced in wild-type and IL-21-deficient mice. CD4(+)CD25(-) T cells from wild-type and IL-21-deficient mice were differentiated in T helper cell (Th)17-polarizing conditions, with or without IL-21 or an antagonistic IL-21R/Fc. We also examined whether blockade of IL-21 by anti-IL-21 antibody reduced IL-17 in cultures of IBD lamina propria CD3(+) T lymphocytes. Cytokines were evaluated by real-time polymerase chain reaction and/or enzyme-linked immunosorbent assay. RESULTS High IL-21 was seen in wild-type mice with dextran sulfate sodium- and trinitrobenzene sulfonic acid-relapsing colitis. IL-21-deficient mice were largely protected against both colitides and were unable to up-regulate Th17-associated molecules during gut inflammation, thus suggesting a role for IL-21 in controlling Th17 cell responses. Indeed, naïve T cells from IL-21-deficient mice failed to differentiate into Th17 cells. Treatment of developing Th17 cells from wild-type mice with IL-21R/Fc reduced IL-17 production. Moreover, in the presence of transforming growth factor-beta1, exogenous IL-21 substituted for IL-6 in driving IL-17 induction. Neutralization of IL-21 reduced IL-17 secretion by IBD lamina propria lymphocytes. CONCLUSIONS These results indicate that IL-21 is a critical regulator of inflammation and Th17 cell responses in the gut.


Nature Medicine | 2009

Involvement of interleukin-21 in the epidermal hyperplasia of psoriasis

Roberta Caruso; Elisabetta Botti; Massimiliano Sarra; Maria Esposito; Carmine Stolfi; Laura Diluvio; Maria Laura Giustizieri; Valentina Pacciani; Annamaria Mazzotta; Elena Campione; Thomas T. MacDonald; Sergio Chimenti; Francesco Pallone; Antonio Costanzo; Giovanni Monteleone

T cells are crucial mediators of the skin damage in psoriasis. We here show that interleukin-21 (IL-21), a T cell–derived cytokine, is highly expressed in the skin of individuals with psoriasis, stimulates human keratinocytes to proliferate and causes epidermal hyperplasia when injected intradermally into mice. In the human psoriasis xenograft mouse model, blockade of IL-21 activity resolves inflammation and reduces keratinocyte proliferation. Blocking IL-21 may represent a new therapeutic strategy in psoriasis.


Gut | 2006

Control of matrix metalloproteinase production in human intestinal fibroblasts by interleukin 21

Giovanni Monteleone; Roberta Caruso; Daniele Fina; Ilaria Peluso; Valentina Gioia; Carmine Stolfi; Massimo C. Fantini; Flavio Caprioli; Roberto Tersigni; Luciano Alessandroni; Thomas T. MacDonald; Francesco Pallone

Background: T cell-mediated immunity plays a central part in the pathogenesis of tissue damage in inflammatory bowel disease (IBD). The mechanism by which T cells mediate tissue damage during IBD remains unclear, but evidence indicates that T cell-derived cytokines stimulate fibroblasts to synthesise matrix metalloproteinases (MMPs), which then mediate mucosal degradation. We have previously shown that, in IBD, there is high production of interleukin (IL) 21, a T cell-derived cytokine, which enhances Th1 activity. Aim: To investigate whether IL21 controls MMP production by intestinal fibroblasts. Methods: IL21 receptor (IL21R) was evaluated in intestinal fibroblasts by reverse transcriptase-polymerase chain reaction (RT-PCR) and western blotting. Fibroblasts were stimulated with IL21 and MMPs were evaluated by RT-PCR and western blotting. The effect of a neutralising IL21R fusion protein (IL21R/Fc) on the induction of MMPs in fibroblasts stimulated with IBD lamina propria mononuclear cell (LPMC) supernatants was also evaluated. Results: Intestinal fibroblasts constitutively express both IL21R and the common γ chain receptor, which are necessary for IL21-driven signalling. IL21 enhances fibroblast production of MMP-1, MMP-2, MMP-3 and MMP-9, but not tissue inhibitors of MMP-1 and MMP-2. Moreover, IL21 synergises with tumour necrosis factor α to increase synthesis of MMP synthesis. IL21 enhances MMP secretion without affecting gene transcription and protein synthesis. IBD LPMC supernatants stimulate MMP secretion by intestinal fibroblasts, and this effect is partly inhibited by IL21R/Fc. Conclusions: These results suggest that fibroblasts are a potential target of IL21 in the gut and that IL21 controls MMP secretion by fibroblasts.


Gastroenterology | 2009

Smad7 Controls Resistance of Colitogenic T Cells to Regulatory T Cell-Mediated Suppression

Massimo C. Fantini; Angelamaria Rizzo; Daniele Fina; Roberta Caruso; Massimiliano Sarra; Carmine Stolfi; Christoph Becker; Thomas T. MacDonald; Francesco Pallone; Markus F. Neurath; Giovanni Monteleone

BACKGROUND & AIMS Foxp3-expressing regulatory T cells (Tregs) play a key role in the maintenance of the gut immune homeostasis, and an intact transforming growth factor (TGF)-beta signaling is required for their function. In inflammatory bowel disease (IBD), the TGF-beta signaling is impaired because of high expression of the inhibitory molecule Smad7. Although no intrinsic defects in Tregs function have been shown in IBD, it is still unknown whether colitogenic T cells are susceptible to Treg-mediated suppression. In this study, we have investigated whether IBD mucosal CD4+ T cells are resistant to Tregs and whether Smad7 is involved in this process. METHODS IBD lamina propria mononuclear cells (LPMC) were cultured with or without Tregs, and proliferation was assessed by flow cytometry. Proliferation of IBD LPMC was also evaluated after Smad7 antisense oligonuclotide treatment. Treg-mediated suppression of T-cell proliferation and proinflammatory cytokine expression was investigated in murine Smad7 transgenic cells. In vivo, the Smad7-dependent resistance of colitogenic naïve T cells to Tregs was studied in the adoptive transfer model of colitis. RESULTS IBD LPMC were resistant to Treg-mediated suppression, and this phenomenon was reverted by Smad7 antisense treatment. Consistently, CD4+ T cells isolated from Smad7 transgenic mice showed high proliferation, produced considerable amount of inflammatory cytokines following activation, and induced a severe colitis when transferred in immunodeficient RAG1 knockout mice even in the presence of wild-type Tregs. CONCLUSIONS Smad7 makes CD4+ T cells resistant to Tregs-mediated suppression thus fine-tuning their proinflammatory potential.


European Journal of Immunology | 2008

IL‐23‐mediated regulation of IL‐17 production in Helicobacter pylori‐infected gastric mucosa

Roberta Caruso; Daniele Fina; O.A. Paoluzi; Giovanna Del Vecchio Blanco; Carmine Stolfi; Angelamaria Rizzo; Flavio Caprioli; Massimiliano Sarra; F. Andrei; Massimo C. Fantini; Thomas T. MacDonald; Francesco Pallone; Giovanni Monteleone

Helicobacter pylori (Hp) infection is associated with a marked infiltration of the gastric mucosa by inflammatory cells. The molecular pathways that control Hp‐associated inflammatory reaction are complex, but locally induced cytokines seem to contribute to maintaining the ongoing inflammation. We have previously shown that IL‐17 is over‐produced in Hp‐infected gastric mucosa, and that IL‐17 stimulates the synthesis of IL‐8, the major neutrophil chemoattractant. Factors/mechanisms that regulate IL‐17 expression remain, however, unknown. In this study, we initially expanded our previous data, showing that CD4+ and CD8+ T cells are a source of IL‐17 in Hp‐infected samples. Since IL‐23 enhances T cell‐derived IL‐17 during bacterial infections, we then assessed the role of IL‐23 in controlling IL‐17 expression in Hp‐colonized stomach. Using real‐time PCR and ELISA, IL‐23 was detected in all gastric biopsies, but its expression was more pronounced in Hp‐infected samples in comparison to controls. Treatment of normal gastric lamina propria mononuclear cells (LPMC) with IL‐23 enhanced Stat3 activation and IL‐17 secretion, and pharmacological inhibition of Stat3 prevented IL‐23‐driven IL‐17 synthesis. Consistently, blockade of IL‐23 in cultures of LPMC from Hp‐infected patients reduced Stat3 activation and IL‐17 production. Data show that IL‐23 is overexpressed in Hp‐infected gastric mucosa where it could contribute to sustaining IL‐17 production.


Oncogene | 2015

Th17-type cytokines, IL-6 and TNF-α synergistically activate STAT3 and NF-kB to promote colorectal cancer cell growth

V. De Simone; Eleonora Franzè; G. Ronchetti; Alfredo Colantoni; Massimo C. Fantini; D. Di Fusco; G. Sica; Pierpaolo Sileri; Thomas T. MacDonald; Francesco Pallone; G. Monteleone; Carmine Stolfi

Colorectal cancers (CRCs) often show a dense infiltrate of cytokine-producing immune/inflammatory cells. The exact contribution of each immune cell subset and cytokine in the activation of the intracellular pathways sustaining CRC cell growth is not understood. Herein, we isolate tumor-infiltrating leukocytes (TILs) and lamina propria mononuclear cells (LPMCs) from the tumor area and the macroscopically unaffected, adjacent, colonic mucosa of patients who underwent resection for sporadic CRC and show that the culture supernatants of TILs, but not of LPMCs, potently enhance the growth of human CRC cell lines through the activation of the oncogenic transcription factors signal transducer and activator of transcription 3 (STAT3) and nuclear factor-kappa B (NF-kB). Characterization of immune cell complexity of TILs and LPMCs reveals no differences in the percentages of T cells, natural killer T cells, natural killer (NK) cells, macrophages and B cells. However, T cells from TILs show a functional switch compared with those from LPMCs to produce large amounts of T helper type 17 (Th17)-related cytokines (that is, interleukin-17A (IL-17A), IL-17F, IL-21 and IL-22), tumor necrosis factor-α (TNF-α) and IL-6. Individual neutralization of IL-17A, IL-17F, IL-21, IL-22, TNF-α or IL-6 does not change TIL-derived supernatant-driven STAT3 and NF-kB activation, as well as their proproliferative effect in CRC cells. In contrast, simultaneous neutralization of both IL-17A and TNF-α, which abrogates NF-kB signaling, and IL-22 and IL-6, which abrogates STAT3 signaling, reduces the mitogenic effect of supernatants in CRC cells. IL-17A, IL-21, IL-22, TNF-α and IL-6 are also produced in excess in the early colonic lesions in a mouse model of sporadic CRC, associated with enhanced STAT3/NF-kB activation. Mice therapeutically given BP-1-102, an orally bioavailable compound targeting STAT3/NF-kB activation and cross-talk, exhibit reduced colon tumorigenesis and diminished expression of STAT3/NF-kB-activating cytokines in the neoplastic areas. These data suggest that strategies aimed at the cotargeting of STAT3/NF-kB activation and interaction between them might represent an attractive and novel approach to combat CRC.


Journal of Experimental Medicine | 2011

Involvement of interleukin-21 in the regulation of colitis-associated colon cancer

Carmine Stolfi; Angelamaria Rizzo; Eleonora Franzè; Angela Rotondi; Massimo C. Fantini; Massimiliano Sarra; Roberta Caruso; Ivan Monteleone; Pierpaolo Sileri; Luana Franceschilli; Flavio Caprioli; Stefano Ferrero; Thomas T. MacDonald; Francesco Pallone; Giovanni Monteleone

IL-21 expression is increased in the gut of patients with colitis-associated colon cancer, and genetic ablation or antibody neutralization of IL-21 reduces tumor size and inflammation in mice treated with dextran sulfate sodium and azoxymethane.


Journal of Immunology | 2008

Autocrine Regulation of IL-21 Production in Human T Lymphocytes

Flavio Caprioli; Massimiliano Sarra; Roberta Caruso; Carmine Stolfi; Daniele Fina; G. Sica; Thomas T. MacDonald; Francesco Pallone; Giovanni Monteleone

IL-21 has pathologic function in immune-inflammatory diseases. IL-21 mediates its functions through a heterodimeric receptor, composed of a specific subunit, termed IL-21R, and the common γ-chain. IL-21 is mostly produced by CD4+ T cells, but molecular mechanisms that regulate IL-21 synthesis are not fully understood. The fact that CD4+ T cells express high levels of IL-21R and are capable of functionally responding to IL-21 raises the possibility that IL-21 may regulate its own production. We here show that IL-21 enhances IL-21 RNA and protein expression in human peripheral blood CD3+ T cells in a dose- and time-dependent fashion. Additionally, both IL-7 and IL-15, but not IL-4, induce IL-21, thus suggesting that common γ-chain signals are not sufficient to promote IL-21 synthesis. Analysis of molecular mechanisms underlying IL-21 induction reveals that IL-21 activates Stat3 and enhances its recruitment to IL-21 gene promoter. Pharmacologic inhibition and knockdown of Stat3 by small interference RNA largely prevent IL-21 induction in IL-21-treated cells. Consistently, IL-21 is inducible in T cells by IL-6, another cytokine that activates Stat3. Finally, we show that IL-21 positively regulates its own expression in human intestinal CD3+ lamina propria lymphocytes, and blockade of endogenous IL-21 in cultures of CD3+ lamina propria lymphocytes isolated from patients with Crohn’s disease, a chronic inflammatory bowel disease characterized by high IL-21, down-regulates Stat3 activation and IL-21 expression. These data suggest the existence of a positive autocrine loop that could help to amplify and stabilize IL-21-driven, T cell-mediated responses.


Gastroenterology | 2009

Interleukin-25 Inhibits Interleukin-12 Production and Th1 Cell-Driven Inflammation in the Gut

Roberta Caruso; Massimiliano Sarra; Carmine Stolfi; Angelamaria Rizzo; Daniele Fina; Massimo C. Fantini; Francesco Pallone; Thomas T. MacDonald; Giovanni Monteleone

BACKGROUND & AIMS During the pathogenesis of Crohns disease (CD), interleukin (IL)-12, a cytokine produced by mucosal CD14+ monocyte-like cells, promotes tissue-damaging T helper cell (Th) 1-mediated inflammation through mechanisms that are not fully understood. IL-25 promotes Th2 cell responses by activating major histocompatibility complex class II-positive non-T and non-B cells. Because Th1 and Th2 cells, and the cytokines they release, are often mutually antagonistic, we examined whether IL-25 affects IL-12 production or Th1 cell-mediated inflammation in the gut. METHODS Studies were performed using colonic samples from patients and mice with peptidoglycan (PGN)-, 2,4,6-trinitrobenzenesulphonic acid (TNBS)-, or oxazolone-induced colitis. IL-25 receptor (IL-25R) levels were evaluated in intestinal lamina propria mononuclear cells by flow cytometry, and IL-25 levels were measured by real-time polymerase chain reaction, immunoblotting, and immunohistochemistry. Mucosal CD14+ cells from patients with CD were incubated with IL-25 and/or lipopolysaccharide or PGN. Mice were injected with IL-25, and some mice first received injections of an IL-13 blocking antibody. Cytokines were quantified by real-time polymerase chain reaction and enzyme-linked immunosorbent assay. RESULTS CD14+ cells from the mucosa of CD patients expressed IL-25R and responded to IL-25 by decreasing the synthesis of IL-12 and IL-23. IL-25 prevented PGN-induced colitis in mice. IL-25 induced IL-13 production in the colon, but IL-13 was not required for suppression of PGN colitis. IL-25 ameliorated TNBS- and oxazolone-colitis. Patients with CD or ulcerative colitis produced significantly less IL-25 compared with controls. CONCLUSIONS IL-25 inhibits CD14+ cell-derived cytokines and experimental colitis. IL-25 could be a useful treatment of CD and ulcerative colitis.


Inflammatory Bowel Diseases | 2010

Interferon-gamma-expressing cells are a major source of interleukin-21 in inflammatory bowel diseases.

Massimiliano Sarra; Ivan Monteleone; Carmine Stolfi; Massimo C. Fantini; Pierpaolo Sileri; G. Sica; Roberto Tersigni; Thomas T. MacDonald; Francesco Pallone; Giovanni Monteleone

Background: We previously demonstrated that in inflammatory bowel disease (IBD) there is enhanced production of interleukin (IL)‐21, a cytokine that activates multiple pathways that sustain mucosal inflammation. However, the phenotype of IL‐21‐producing cells in IBD, and the cytokine(s) they coproduce, is not known. We here characterized the cell source of IL‐21 and determined which factors regulate IL‐21 in the human gut. Methods: Cytokines were analyzed in CD4+ T intestinal lamina propria lymphocytes (T‐LPL) isolated from IBD patients and controls by flow cytometry. Moreover, IL‐21 was evaluated in mucosal T follicular cells (TFH). To assess the involvement of IL‐12 and IL‐23 in the production of IL‐21, T‐LPL were activated in the presence or absence of IL‐12 or IL‐23. Results: The proportion of IL‐21‐producing CD4+ T‐LPL was increased in IBD compared to controls. The majority of IL‐21‐producing T‐LPL coexpressed interferon (IFN)‐&ggr;, and to a lesser extent IL‐4 or IL‐17A. Activation of CD4+ T‐LPL with IL‐12 but not IL‐23 enhanced the fraction of cells coexpressing IL‐21 and IFN‐&ggr;. TFH cells in LPL were identified by CXCR5 expression and expressed IL‐21 both in IBD and controls; however, the fraction of IL‐21‐positive TFH cells was higher in Crohns disease than in ulcerative colitis and controls. Treatment of CD4+ T‐LPL with IL‐12 enhanced the frequency of CXCR5+ IL‐21‐producing TFH cells. Conclusions: These findings indicate that in IBD IL‐21 is mostly produced by CD4+ T‐LPL coexpressing IFN‐&ggr;, reinforcing the concept that distinct subsets of T cells can produce IL‐21. Inflamm Bowel Dis 2010

Collaboration


Dive into the Carmine Stolfi's collaboration.

Top Co-Authors

Avatar

Francesco Pallone

University of Rome Tor Vergata

View shared research outputs
Top Co-Authors

Avatar

Giovanni Monteleone

University of Rome Tor Vergata

View shared research outputs
Top Co-Authors

Avatar

Roberta Caruso

University of Rome Tor Vergata

View shared research outputs
Top Co-Authors

Avatar

Massimo C. Fantini

University of Rome Tor Vergata

View shared research outputs
Top Co-Authors

Avatar

Massimiliano Sarra

University of Rome Tor Vergata

View shared research outputs
Top Co-Authors

Avatar

Thomas T. MacDonald

Queen Mary University of London

View shared research outputs
Top Co-Authors

Avatar

Eleonora Franzè

University of Rome Tor Vergata

View shared research outputs
Top Co-Authors

Avatar

Angelamaria Rizzo

University of Rome Tor Vergata

View shared research outputs
Top Co-Authors

Avatar

Daniele Fina

University of Rome Tor Vergata

View shared research outputs
Top Co-Authors

Avatar

G. Monteleone

University of Southampton

View shared research outputs
Researchain Logo
Decentralizing Knowledge