Network


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

Hotspot


Dive into the research topics where Michele Petruzzelli is active.

Publication


Featured researches published by Michele Petruzzelli.


Gastroenterology | 2012

Selective Activation of Nuclear Bile Acid Receptor FXR in the Intestine Protects Mice Against Cholestasis

Salvatore Modica; Michele Petruzzelli; Elena Bellafante; Stefania Murzilli; Lorena Salvatore; Nicola Celli; Giuseppe Di Tullio; Giuseppe Palasciano; Tarek Moustafa; Emina Halilbasic; Michael Trauner; Antonio Moschetta

BACKGROUND & AIMS Cholestasis is a liver disorder characterized by impaired bile flow, reduction of bile acids (BAs) in the intestine, and retention of BAs in the liver. The farnesoid X receptor (FXR) is the transcriptional regulator of BA homeostasis. Activation of FXR by BAs reduces circulating BA levels in a feedback mechanism, repressing hepatic cholesterol 7α-hydroxylase (Cyp7a1), the rate-limiting enzyme for the conversion of cholesterol to BAs. This mechanism involves the hepatic nuclear receptor small heterodimer partner and the intestinal fibroblast growth factor (FGF) 19 and 15. We investigated the role of activation of intestine-specific FXR in reducing hepatic levels of BAs and protecting the liver from cholestasis in mice. METHODS We generated transgenic mice that express a constitutively active FXR in the intestine. Using FXR gain- and loss-of-function models, we studied the roles of intestinal FXR in mice with intrahepatic and extrahepatic cholestasis. RESULTS Selective activation of intestinal FXR induced FGF15 and repressed hepatic Cyp7a1, reducing the pool size of BAs and changing the BA pool composition. Activation of intestinal FXR protected mice from obstructive extrahepatic cholestasis after bile duct ligation or administration of α-naphthylisothiocyanate. In Mdr2(-/-) mice, transgenic expression of activated FXR in the intestine protected against liver damage, whereas absence of FXR promoted progression of liver disease. CONCLUSIONS Activation of FXR transcription in the intestine protects the liver from cholestasis in mice by inducing FGF15 expression and reducing the hepatic pool of BA; this approach might be developed to reverse cholestasis in patients.


Cell Metabolism | 2010

Intestinal specific LXR activation stimulates reverse cholesterol transport and protects from atherosclerosis

Giuseppe Lo Sasso; Stefania Murzilli; Lorena Salvatore; Ilenia D'Errico; Michele Petruzzelli; Paola Conca; Zhao Yan Jiang; Laura Calabresi; Paolo Parini; Antonio Moschetta

Several steps of the HDL-mediated reverse cholesterol transport (RCT) are transcriptionally regulated by the nuclear receptors LXRs in the macrophages, liver, and intestine. Systemic LXR activation via synthetic ligands induces RCT but also causes increased hepatic fatty acid synthesis and steatosis, limiting the potential therapeutic use of LXR agonists. During the last few years, the participation of the intestine in the control of RCT has appeared more evident. Here we show that while hepatic-specific LXR activation does not contribute to RCT, intestinal-specific LXR activation leads to decreased intestinal cholesterol absorption, improved lipoprotein profile, and increased RCT in vivo in the absence of hepatic steatosis. These events protect against atherosclerosis in the background of the LDLR-deficient mice. Our study fully characterizes the molecular and metabolic scenario that elects the intestine as a key player in the LXR-driven protective environment against cardiovascular disease.


Cell Death & Differentiation | 2009

p38α blockade inhibits colorectal cancer growth in vivo by inducing a switch from HIF1α- to FoxO-dependent transcription

F Chiacchiera; A Matrone; E Ferrari; G Ingravallo; G Lo Sasso; Stefania Murzilli; Michele Petruzzelli; Lorena Salvatore; Antonio Moschetta; C Simone

Colorectal cancer cell (CRC) fate is governed by an intricate network of signaling pathways, some of which are the direct target of DNA mutations, whereas others are functionally deregulated. As a consequence, cells acquire the ability to grow under nutrients and oxygen shortage conditions. We earlier reported that p38α activity is necessary for proliferation and survival of CRCs in a cell type-specific manner and regardless of their phenotype and genotype. Here, we show that p38α sustains the expression of HIF1α target genes encoding for glycolytic rate-limiting enzymes, and that its inhibition causes a drastic decrease in ATP intracellular levels in CRCs. Prolonged inactivation of p38α triggers AMPK-dependent nuclear localization of FoxO3A and subsequent activation of its target genes, leading to autophagy, cell cycle arrest and cell death. In vivo, pharmacological blockade of p38α inhibits CRC growth in xenografted nude mice and azoxymethane-treated ApcMin mice, achieving both a cytostatic and cytotoxic effect, associated with high nuclear expression of FoxO3A and increased expression of its target genes p21 and PTEN. Hence, inhibition of p38α affects the aerobic glycolytic metabolism specific of cancer cells and might be taken advantage of as a therapeutic strategy targeted against CRCs.


Gastroenterology | 2008

Abcg5/8 Independent Biliary Cholesterol Excretion in Atp8b1-Deficient Mice

A. K. Groen; Cindy Kunne; Geartsje Jongsma; Karin van den Oever; Kam S. Mok; Michele Petruzzelli; Carlos L. J. Vrins; Laura N. Bull; Coen C. Paulusma; Ronald P. J. Oude Elferink

BACKGROUNDS & AIMS ATP8B1 is a phosphatidylserine flippase in the canalicular membrane; patients with mutations in ATP8B1 develop severe chronic (PFIC1) or periodic (BRIC1) cholestatic liver disease. We have observed that Atp8b1 deficiency leads to enhanced biliary cholesterol excretion. It has been established that biliary cholesterol excretion depends on transport by the heterodimer Abcg5/Abcg8. We hypothesized that the increased cholesterol output was due to enhanced extraction from the altered canalicular membrane rather than to higher Abcg5/Abcg8 activity. We therefore studied the relation between Abcg5/Abcg8 expression and biliary cholesterol excretion in mice lacking Atp8b1, Abcg8, or both (GF mice). METHODS Bile formation was studied in LXR agonist-fed wild-type mice as well as mice lacking Atp8b1 or Abcg8, or in GF mice upon infusion of taurocholate. Bile samples were analyzed for cholesterol, bile salt, phospholipids, and ectoenzyme content. RESULTS LXR agonist increased Abcg5/8 expression, and this was accompanied by increased biliary cholesterol output in both wild-type and Atp8b1(G308V/G308V) mice. However, Atp8b1(G308V/G308V) mice maintained higher cholesterol output. Although in Abcg8(-/-) mice biliary cholesterol output was severely reduced, GF mice displayed high biliary cholesterol output, which was comparable with wild-type mice. Bile of both Atp8b1(G308V/G308V) and GF mice displayed elevated levels of phosphatidylserine and sphingomyelin, indicating membrane stress. CONCLUSIONS Our data demonstrate that the increased biliary cholesterol excretion in Atp8b1-deficient mice is independent of Abcg5/8 activity. This implicates that Atp8b1 deficiency leads to a decrease in the detergent resistance and subsequent nonspecific extraction of cholesterol from the canalicular membrane by bile salts.


Biochimica et Biophysica Acta | 2008

A translational view on the biliary lipid secretory network.

Giuseppe Lo Sasso; Michele Petruzzelli; Antonio Moschetta

Bile formation springs from an extraordinary sophisticated secretory network, which combines the activity of transport proteins with the physico-chemical properties of small albeit powerful lipophilic compounds. This robust interplay is activated in response to dietary stimuli, circadian rhythms, and metabolic demands to regulate cholesterol disposal, lipid digestion and absorption in the enterohepatic system. As a result, bile flow is a complex multi-organ effort that requires an integrated flux of information between liver and intestine. A coordinate regulatory task is achieved by nuclear receptors, which are ligand activated transcription factors, responsible for the coherent activation of sets of genes involved in multiple physiological actions, including hepato-biliary homeostasis. Mastering the molecular pathways underlying functional and pharmacological modulation of bile flow has great translational value for potential future treatment of cholestasis and cholelithiasis. In this review, we focus on recent discoveries in the functional biology of bile formation with the explicit aim of underlining their putative clinical relevance.


Biology of the Cell | 2003

Basolateral Ca2+-dependent K+-channels play a key role in Cl-secretion induced by taurodeoxycholate from colon mucosa

Antonio Moschetta; Piero Portincasa; Lucantonio Debellis; Michele Petruzzelli; Roberta Montelli; Giuseppe Calamita; Pontus Gustavsson; Giuseppe Palasciano

Abstract The diarrhea associated with malabsorption of bile salts such as the secondary hydrophobic taurodeoxycholate (TDC) may be partly explained by the TDC‐induced increase in colon Cl− secretion. We, therefore, investigated the effects of TDC (0.5–8 mM) on electrical parameters and electrolyte transport of rat proximal colon mucosa mounted in Ussing chambers. Colonic secretion, measured as short circuit current (I SC), progressively increased on mucosal incubation with TDC ranging 0.5–2 mM; up to TDC 2 mM, a spontaneous recovery toward control values with no changes in epithelial resistance (Rt), and lactate dehydrogenase (LDH) release was observed. In contrast, for TDC > 2 mM, I SC increased further and the effect was progressive and associated with a significant decrease in the Rt and increased LDH release, implying a cytolytic effect. Mucosal preincubation with the Cl− channel inhibitor 5‐nitro‐2‐(3‐phenylpropylamino) benzoic acid (NPPB), fully prevented the precytolytic effect of TDC on I SC. Serosal preincubation with furosemide, a Na+/K+/2Cl− cotransporter inhibitor, significantly reduced TDC‐induced increase in I SC. Inhibition of the basolateral Ca2+‐dependent K+ channel—rSK4—with serosal clotrimazole or incubation with mucosal Ca2+‐free (EGTA) buffer completely prevented precytolytic TDC‐induced increase in I SC. In conclusion, Cl− secretion is activated in colon mucosa by TDC low concentrations; while at higher concentrations, a detergent cytotoxic effect intervenes. Activation of the Ca2+‐dependent basolateral K+ pathway, through TDC‐induced apical Ca2+ influx, provides the Na+/K+/2Cl− basolateral activation, thereby the driving force for the apical exit of Cl− ions. These findings further enhance the knowledge of the pathogenic mechanisms of diarrhea associated with bile salt malabsorption.


Gut | 2009

Expression and localisation of insulin receptor substrate 2 in normal intestine and colorectal tumours. Regulation by intestine-specific transcription factor CDX2

Salvatore Modica; Annalisa Morgano; Lorena Salvatore; Michele Petruzzelli; Marie-Thérèse Vanier; Rosa Valanzano; Diana L. Esposito; Giuseppe Palasciano; Isabelle Duluc; Jean-Noël Freund; Renato Mariani-Costantini; Antonio Moschetta

Background and aims: Self-renewal and differentiation of intestinal epithelium is a tightly regulated process, whose perturbations are implicated in human colorectal tumourigenesis. The insulin/insulin-like growth factor (IGF) signalling pathway may play an important role in intestinal epithelium homeostasis. Insulin receptor substrate 2 (IRS2) is a poorly characterised component in this pathway. Methods: Using complementary in vitro and in vivo human and murine models, expression (mRNA and protein levels), localisation (immunohistochemistry) and regulation of IRS2 were investigated in the normal intestine and colorectal tumours. In silico analysis of the human IRS2 promoter was performed together with reporter and chromatin immunoprecipitation assays. Results: Significant IRS2 expression was detected in the intestine, with specific protein localisation in the villus region of the ileum and in the surface epithelium of the colon. In human HT29 and Caco2 cells, IRS2 mRNA levels increased with spontaneous and induced differentiation, together with CDX2 (caudal-related homeobox protein 2), P21 and KLF4 (Krüppel-like factor 4). Adenoviral infection with human CDX2 induced IRS2 expression in APC- (adenomatous polyposis coli) and β-catenin-mutated cells. On the other hand, IRS2 downregulation was observed in differentiated enterocytes after adenoviral infection with short hairpin CDX2 (shCDX2), in the intestine of CDX2 heterozygous mice and in colorectal tumours of ApcMin/+ and patients with familial adenomatous polyposis (FAP). The human IRS2 promoter region presents several CDX2-binding sites where CDX2 immunoprecipitated in vivo. IRS2 reporters were functionally activated via CDX2 and blocked via a dominant-negative CDX2 protein. Conclusions: Combining gain- and loss-of-function approaches, an intriguing scenario is presented whereby IRS2 is significantly expressed in the apical intestinal compartment and is directly controlled by CDX2 in normal intestine and tumours.


Journal of Medicinal Chemistry | 2009

Synthesis and Biological Evaluation of 2-Heteroarylthioalkanoic Acid Analogues of Clofibric Acid as Peroxisome Proliferator-Activated Receptor α Agonists

Letizia Giampietro; Alessandra Ammazzalorso; Antonella Giancristofaro; Fabio Lannutti; Giancarlo Bettoni; Barbara De Filippis; Marialuigia Fantacuzzi; Cristina Maccallini; Michele Petruzzelli; Annalisa Morgano; Antonio Moschetta; Rosa Amoroso

A series of 2-heteroarylthioalkanoic acids were synthesized through systematic structural modifications of clofibric acid and evaluated for human peroxisome proliferator-activated receptor alpha (PPARalpha) transactivation activity, with the aim of obtaining new hypolipidemic compounds. Some thiophene and benzothiazole derivatives showing a good activation of the receptor alpha were screened for activity against the PPARgamma isoform. The gene induction of selected compounds was also investigated in the human hepatoma cell line.


Current Medicinal Chemistry | 2007

Current Treatments of Primary Sclerosing Cholangitis

Michele Vacca; Marcin Krawczyk; Michele Petruzzelli; Rosa Cinzia Sasso; K.J. van Erpecum; Giuseppe Palasciano; Gerard P. vanBerge-Henegouwen; Antonio Moschetta; Piero Portincasa

Primary Sclerosing Cholangitis (PSC) is a chronic cholestatic disease characterized by hepatic inflammation and obliterative fibrosis, resulting in both intra- and extra-hepatic bile duct strictures. End-stage liver disease and bile duct carcinoma represent frequent complications. Incidence and prevalence of PSC in USA have been recently estimated as 0.9 per 100,000 person-years, and 1-6 per 100,000 person-years, respectively. Major diagnostic criteria include the presence of multifocal strictures, beadings of bile ducts, and compatible biochemical profile, once excluded secondary causes of cholangitis. Since the aetiology of PSC remains poorly defined, medical therapy is currently limited to symptom improvement and prolonged survival. Ursodeoxycholic acid (UDCA), corticosteroids and immunosuppressants have been proposed alone or in combination to improve the clinical outcome. In selected cases, surgical or endoscopic procedures need to be considered. Orthotopic liver transplantation (OLT) is at the moment the only definitive approach although disease relapse has been reported. In this article the state of the art in PSC treatment and future promises in this field are reviewed.


European Journal of Medicinal Chemistry | 2011

Discovery of gemfibrozil analogues that activate PPARα and enhance the expression of gene CPT1A involved in fatty acids catabolism.

Barbara De Filippis; Antonella Giancristofaro; Alessandra Ammazzalorso; Alessandra D’Angelo; Marialuigia Fantacuzzi; Letizia Giampietro; Cristina Maccallini; Michele Petruzzelli; Rosa Amoroso

A new series of gemfibrozil analogues conjugated with α-asarone, trans-stilbene, chalcone, and their bioisosteric modifications were synthesized and evaluated to develop PPARα agonists. In this attempt, we have removed the methyls on the phenyl ring of gemfibrozil and introduced the above scaffolds in para position synthesizing two series of derivatives, keeping the dimethylpentanoic skeleton of gemfibrozil unaltered or demethylated. Four compounds exhibited good activation of the PPARα receptor and were also screened for their activity on PPARα-regulated gene CPT1A.

Collaboration


Dive into the Michele Petruzzelli's collaboration.

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
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Letizia Giampietro

University of Chieti-Pescara

View shared research outputs
Researchain Logo
Decentralizing Knowledge