Network


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

Hotspot


Dive into the research topics where Annamaria Naggi is active.

Publication


Featured researches published by Annamaria Naggi.


Nature Biotechnology | 2008

Oversulfated chondroitin sulfate is a contaminant in heparin associated with adverse clinical events

Marco Guerrini; Daniela Beccati; Zachary Shriver; Annamaria Naggi; Karthik Viswanathan; Antonella Bisio; Ishan Capila; Jonathan C. Lansing; Sara Guglieri; Blair A. Fraser; Ali Al-Hakim; Nur Sibel Gunay; Zhenqing Zhang; Luke N. Robinson; Lucinda F. Buhse; Moheb Nasr; Janet Woodcock; Robert Langer; Ganesh Venkataraman; Robert J. Linhardt; Benito Casu; Giangiacomo Torri; Ram Sasisekharan

Recently, certain lots of heparin have been associated with an acute, rapid onset of serious side effects indicative of an allergic-type reaction. To identify potential causes for this sudden rise in side effects, we examined lots of heparin that correlated with adverse events using orthogonal high-resolution analytical techniques. Through detailed structural analysis, the contaminant was found to contain a disaccharide repeat unit of glucuronic acid linked β1→3 to a β-N-acetylgalactosamine. The disaccharide unit has an unusual sulfation pattern and is sulfated at the 2-O and 3-O positions of the glucuronic acid as well as at the 4-O and 6-O positions of the galactosamine. Given the nature of this contaminant, traditional screening tests cannot differentiate between affected and unaffected lots. Our analysis suggests effective screening methods that can be used to determine whether or not heparin lots contain the contaminant reported here.


Current Pharmaceutical Design | 2007

Heparanase: structure, biological functions, and inhibition by heparin-derived mimetics of heparan sulfate.

Israel Vlodavsky; Neta Ilan; Annamaria Naggi; Benito Casu

Heparanase is an endoglycosidase which cleaves heparan sulfate (HS) and hence participates in degradation and remodeling of the extracellular matrix (ECM). Heparanase is preferentially expressed in human tumors and its over-expression in tumor cells confers an invasive phenotype in experimental animals. The enzyme also releases angiogenic factors from the ECM and thereby induces an angiogenic response in vivo. Heparanase upregulation correlates with increased tumor vascularity and poor postoperative survival of cancer patients. Heparanase is synthesized as a 65 kDa inactive precursor that undergoes proteolytic cleavage, yielding 8 kDa and 50 kDa protein subunits that heterodimerize to form an active enzyme. Heparanase exhibits also non-enzymatic activities, independent of its involvement in ECM degradation. Among these, are the enhancement of Akt signaling, stimulation of PI3K- and p38-dependent endothelial cell migration, and up regulation of VEGF, all contributing to its potent pro-angiogenic activity. Studies on relationships between structure and heparanase inhibition activity of nonanticogulant heparins systematically differing in their O-sulfation patterns, degrees of N-acetylation, and glycol-splitting of both pre-existing nonsulfated uronic acid residues (prevalently D-glucuronic) and/or those (L-iduronic acid/L-galacturonic acid) generated by graded 2-O-desulfation, have permitted to select effective inhibitors of the enzymatic activity of heparanase. N-acetylated, glycol-split heparins emerged as especially strong inhibitors of heparanase, exerting little or no release of growth factors from ECM. N-acetylated glycol-split species of heparin, as well as heparanase gene silencing inhibit tumor metastasis, angiogenesis and inflammation in experimental animal models. These observations and the unexpected identification of a single functional heparanase, suggest that the enzyme is a promising target for anti-cancer and anti-inflammatory drug development.


Carbohydrate Polymers | 1990

Alkaline N-deacetylation of chitin enhanced by flash treatments. Reaction kinetics and structure modifications

Bonaventura Focher; P.L. Beltrame; Annamaria Naggi; Giangiacomo Torri

A new method for N-deacetylation of chitin is proposed in which a polymer almost free of N-acetyl groups is obtained by flash treatment. The reaction is carried out in 40% NaOH solution for 30–270 s at 140–190°C, using saturated steam. Flash treatment was found to proceed faster and with a higher activation energy for the deacetylation reaction (Ea = 36 kcal mol−1) compared with the traditional treatment (Ea = 11 kcal mol−1). X-Ray diffractometry, CP-MAS 13C-NMR and FTIR spectroscopy show that the flash treatment induces structure modifications; in particular, higher crystallinity indexes and specific area values are observed together with changes in the local and chain conformation.


Journal of Biological Chemistry | 2005

Modulation of the Heparanase-inhibiting Activity of Heparin through Selective Desulfation, Graded N-Acetylation, and Glycol Splitting

Annamaria Naggi; Benito Casu; Marta Perez; Giangiacomo Torri; Giuseppe Cassinelli; Sergio Penco; Claudio Sigma-Tau Ind. Farm. Riunite Spa Pisano; Giuseppe Giannini; Rivka Ishai-Michaeli; Israel Vlodavsky

Heparanase is an endo-β-glucuronidase that cleaves heparan sulfate (HS) chains of heparan sulfate proteoglycans on cell surfaces and in the extracellular matrix (ECM). Heparanase, overexpressed by most cancer cells, facilitates extravasation of blood-borne tumor cells and causes release of growth factors sequestered by HS chains, thus accelerating tumor growth and metastasis. Inhibition of heparanase with HS mimics is a promising target for a novel strategy in cancer therapy. In this study, in vitro inhibition of recombinant heparanase was determined for heparin derivatives differing in degrees of 2-O- and 6-O-sulfation, N-acetylation, and glycol splitting of nonsulfated uronic acid residues. The contemporaneous presence of sulfate groups at O-2 of IdoA and at O-6 of GlcN was found to be non-essential for effective inhibition of heparanase activity provided that one of the two positions retains a high degree of sulfation. N-Desulfation/ N-acetylation involved a marked decrease in the inhibitory activity for degrees of N-acetylation higher than 50%, suggesting that at least one NSO3 group per disaccharide unit is involved in interaction with the enzyme. On the other hand, glycol splitting of preexisting or of both preexisting and chemically generated nonsulfated uronic acids dramatically increased the heparanase-inhibiting activity irrespective of the degree of N-acetylation. Indeed N-acetylated heparins in their glycol-split forms inhibited heparanase as effectively as the corresponding N-sulfated derivatives. Whereas heparin and N-acetylheparins containing unmodified d-glucuronic acid residues inhibited heparanase by acting, at least in part, as substrates, their glycol-split derivatives were no more susceptible to cleavage by heparanase. Glycol-split N-acetylheparins did not release basic fibroblast growth factor from ECM and failed to stimulate its mitogenic activity. The combination of high inhibition of heparanase and low release/potentiation of ECM-bound growth factor indicates that N-acetylated, glycol-split heparins are potential antiangiogenic and antimetastatic agents that are more effective than their counterparts with unmodified backbones.


Carbohydrate Research | 1996

1H and 13C NMR spectral assignments of the major sequences of twelve systematically modified heparin derivatives

Edwin A. Yates; Francesco Santini; Marco Guerrini; Annamaria Naggi; Giangiacomo Torri; Benito Casu

The complete 1H and 13C NMR spectral assignments are described for the most prevalent patterns of sulfation and acetylation which can be found in polymeric heparin or can be obtained by standard chemical modifications. These include a number of novel structures containing unsubstituted or acetylated amino groups and the first complete NMR assignments of many of the other derivatives. Beef lung heparin was chosen as a model system and studies were carried out using conditions to control the influences on the chemical shift positions in heparin samples of divalent cations and variations in pH and temperature.


Clinical Cancer Research | 2011

SST0001, a Chemically Modified Heparin, Inhibits Myeloma Growth and Angiogenesis via Disruption of the Heparanase/Syndecan-1 Axis

Joseph P. Ritchie; Vishnu C. Ramani; Yongsheng Ren; Annamaria Naggi; Giangiacomo Torri; Benito Casu; Sergio Penco; Claudio Pisano; Paolo Carminati; Monica Tortoreto; Franco Zunino; Israel Vlodavsky; Ralph D. Sanderson; Yang Yang

Purpose: Heparanase promotes myeloma growth, dissemination, and angiogenesis through modulation of the tumor microenvironment, thus highlighting the potential of therapeutically targeting this enzyme. SST0001, a nonanticoagulant heparin with antiheparanase activity, was examined for its inhibition of myeloma tumor growth in vivo and for its mechanism of action. Experimental Design: The ability of SST0001 to inhibit growth of myeloma tumors was assessed using multiple animal models and a diverse panel of human and murine myeloma cell lines. To investigate the mechanism of action of SST0001, pharmacodynamic markers of angiogenesis, heparanase activity, and pathways downstream of heparanase were monitored. The potential use of SST0001 as part of a combination therapy was also evaluated in vivo. Results: SST0001 effectively inhibited myeloma growth in vivo, even when confronted with an aggressively growing tumor within human bone. In addition, SST0001 treatment causes changes within tumors consistent with the compounds ability to inhibit heparanase, including downregulation of HGF, VEGF, and MMP-9 expression and suppressed angiogenesis. SST0001 also diminishes heparanase-induced shedding of syndecan-1, a heparan sulfate proteoglycan known to be a potent promoter of myeloma growth. SST0001 inhibited the heparanase-mediated degradation of syndecan-1 heparan sulfate chains, thus confirming the antiheparanase activity of this compound. In combination with dexamethasone, SST0001 blocked tumor growth in vivo presumably through dual targeting of the tumor and its microenvironment. Conclusions: These results provide mechanistic insight into the antitumor action of SST0001 and validate its use as a novel therapeutic tool for treating multiple myeloma. Clin Cancer Res; 17(6); 1382–93. ©2011 AACR.


Carbohydrate Polymers | 1992

Structural differences between chitin polymorphs and their precipitates from solutions—evidence from CP-MAS 13C-NMR, FT-IR and FT-Raman spectroscopy

Bonaventura Focher; Annamaria Naggi; Giangiacomo Torri; A. Cosani; M. Terbojevich

Abstract Structural differences between α-chitin from shrimp ( Crangon crangon ) and β-chitin from squid (Loligo), as well as between their precipitated products from N,N-dimethylacetamide-LiCl solutions, are indicated by the CP-MAS 13 C-NMR, FT-IR, FT-Raman spectra and the X-ray diffractograms. The 13 C-NMR spectra in the solid state of α-chitin consisting of eight major resonances suggest a high degree of structural homogeneity. In the spectrum of β-chitin, the C-3 and C-5 signals merged in a single resonance. The bands contour of deconvoluted and curve-fit FT-IR and FT-Raman spectra shows a more detailed structure of α-chitin in the region of OH, NH and CO stretching regions; in particular the Amide I band is split, whereas a single broad band dominates in the corresponding β-form. Precipitation treatments induce a general disorder in α-chitin, while the metastable structure of β-chitin tends toward a more ordered architecture. All the present spectroscopic data of chitin samples are consistent with the corresponding X-ray diffraction patterns.


The FASEB Journal | 2005

Heparanase accelerates wound angiogenesis and wound healing in mouse and rat models

Eyal Zcharia; Rachel Zilka; Alon Yaar; Oron Yacoby-Zeevi; Anna Zetser; Shula Metzger; Ronit Sarid; Annamaria Naggi; Benito Casu; Neta Ilan; Israel Vlodavsky; Rinat Abramovitch

Orchestration of the rapid formation and reorganization of new tissue observed in wound healing involves not only cells and polypeptides but also the extracellular matrix (ECM) microenvironment. The ability of heparan sulfate (HS) to interact with major components of the ECM suggests a key role for HS in maintaining the structural integrity of the ECM. Heparanase, an endoglycosidase‐degrading HS in the ECM and cell surface, is involved in the enzymatic machinery that enables cellular invasion and release of HS‐bound polypeptides residing in the ECM. Bioavailabilty and activation of multitude mediators capable of promoting cell migration, proliferation, and neovascularization are of particular importance in the complex setting of wound healing. We provide evidence that heparanase is normally expressed in skin and in the wound granulation tissue. Heparanase stimulated keratinocyte cell migration and wound closure in vitro. Topical application of recombinant heparanase significantly accelerated wound healing in a flap/punch model and markedly improved flap survival. These heparanase effects were associated with enhanced wound epithelialization and blood vessel maturation. Similarly, a marked elevation in wound angiogenesis, evaluated by MRI analysis and histological analyses, was observed in heparanase‐overexpressing transgenic mice. This effect was blocked by a novel, newly developed, heparanase‐inhibiting glycol‐split fragment of heparin. These results clearly indicate that elevation of heparanase levels in healing wounds markedly accelerates tissue repair and skin survival that are mediated primarily by an enhanced angiogenic response.—Zcharia, E., Zilka, R., Yaar, A., Yacoby‐Zeevi, O., Zetser, A., Metzger, S., Sarid, R., Naggi, A., Casu, B., Ilan, N., Vlodavsky, I., Abramovitch, R. Heparanase accelerates wound angiogenesis and wound healing in mouse and rat models. FASEB J. 19, 211–221 (2005)


Journal of Applied Polymer Science | 1998

Sorption of aromatic compounds in water using insoluble cyclodextrin polymers

Grégorio Crini; Sabrina Bertini; Giangiacomo Torri; Annamaria Naggi; D. Sforzini; Carmen Vecchi; Ludovic Janus; Yahya Lekchiri; Michel Morcellet

Insoluble β-cyclodextrin (β-CD) polymers have been used for the recovery of various organic pollutants from aqueous solutions. These resins have been prepared by polymerization using epichlorohydrin (Epi) as a crosslinking agent. Several crosslinked polymers with various degrees of β-CD were used. Several studies (time, concentration, kinetics, and pH) are presented here. The results show that these sorbents exhibit high sorption capacities toward substituted benzene derivatives. The mechanism of sorption is both physical adsorption in the polymer network and/or the formation of an inclusion complex and/or the formation of hydrophobic guest–guest interactions.


Carbohydrate Research | 1994

Heparin-like compounds prepared by chemical modification of capsular polysaccharide from E. coli K5☆

Benito Casu; Giordana Grazioli; Nahid Razi; Marco Guerrini; Annamaria Naggi; Giangiacomo Torri; Pasqua Oreste; Francesco Tursi; Giorgio Zoppetti; Ulf Lindahl

O-Sulfation of sulfaminoheparosan SAH, a glycosaminoglucuronan with the structure-->4)-beta-D-GlcA(1-->4)-beta-D-GlcNSO3(-)-(1-->, obtained by N-deacetylation and N-sulfation of the capsular polysaccharide from E. coli K5, was investigated in order to characterize the sulfation pattern eliciting heparin-like activities. SAH was reacted (as the tributylammonium salt in N,N-dimethylformamide) with pyridine-sulfur trioxide under systematically different experimental conditions. The structure of O-sulfated products (SAHS), as determined by mono- and two-dimensional 1H and 13C NMR, varied with variation of reaction parameters. Sulfation of SAH preferentially occurred at O-6 of the GlcNSO3- residues. Further sulfation occurred either at O-3 or at O-2 of the GlcA residues, depending on the experimental conditions. Products with significantly high affinity for antithrombin and antifactor Xa activity were obtained under well-defined conditions. These products contained the trisulfated aminosugar GlcNSO3-3,6SO3-, which is a marker component of the pentasaccharide sequence through which heparin binds to antithrombin.

Collaboration


Dive into the Annamaria Naggi's collaboration.

Top Co-Authors

Avatar

Giangiacomo Torri

Rappaport Faculty of Medicine

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Marco Guerrini

Federal University of Paraná

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ram Sasisekharan

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Israel Vlodavsky

Rappaport Faculty of Medicine

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Sabrina Bertini

Stazione Sperimentale per i Combustibili

View shared research outputs
Top Co-Authors

Avatar

Zachary Shriver

University of Pennsylvania

View shared research outputs
Researchain Logo
Decentralizing Knowledge