Network


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

Hotspot


Dive into the research topics where Benito Casu is active.

Publication


Featured researches published by Benito Casu.


Biochemical and Biophysical Research Communications | 1983

Structure-activity relationship in heparin: A synthetic pentasaccharide with high affinity for antithrombin III and eliciting high anti-factor Xa activity

Jean Choay; M. Petitou; J.C. Lormeau; Pierre Sinaÿ; Benito Casu; G. Gatti

The structures of the tetrasaccharide (beta-D-glucuronic acid)1 leads to 4 (N-sulfate-3,6-di-0-sulfate-alpha-D-glucosamine)1 leads to 4(2-0-sulfate-alpha-L-iduronic acid)1 leads to 4(N-sulfate-6-0-sulfate-D-glucosamine) and of the pentasaccharide (N-sulfate-6-0-sulfate-alpha-D-glucosamine)1 leads to 4(beta-D-glucuronic acid)1 leads to 4(N-sulfate-3,6-di-0-sulfate-alpha-D-glucosamine)1 leads to 4(2-0-sulfate-alpha-L-iduronic acid)1 leads to 4(N-sulfate-6-0-sulfate-D-glucosamine), both prepared for the first time, by chemical synthesis from D-glucose and D-glucosamine, have been confirmed by nuclear magnetic resonance. The synthetic tetrasaccharide neither binds to AT-III nor induces anti-factor Xa activity enhancement of this inhibitor. In contrast, the synthetic pentasaccharide strongly binds to AT-III (Ka: 7.10(6)M-1) forming an equimolar complex and also enhances the AT-III inhibitory activity towards factor Xa. These results confirm that the synthetic pentasaccharide with the above structure corresponds to the actual minimal sequence required in heparin for binding to AT-III.


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.


Carbohydrate Research | 1990

Conformer populations of L-iduronic acid residues in glycosaminoglycan sequences

Dino R. Ferro; Augusto Provasoli; Massimo Ragazzi; Benito Casu; Giangiacomo Torri; Veronique Bossennec; Bruno Perly; Pierre Sinaÿ; Maurice Petitou; Jean Choay

The 1H-n.m.r. 3J values for the L-iduronic acid (IdoA) residues for solutions in D2O of natural and synthetic oligosaccharides that represent the biologically important sequences of dermatan sulfate, heparan sulfate, and heparin have been rationalized by force-field calculations. The relative proportions of the low-energy conformers 1C4, 2S0, and 4C1 vary widely as a function of sequence and of pattern of sulfation. When IdoA or IdoA-2-sulfate units are present inside saccharide sequences, only 1C4 and 2S0 conformations contribute significantly to the equilibrium. This equilibrium is displaced towards the 2S0 form when IdoA-2-sulfate is preceded by a 3-O-sulfated amino sugar residue, and towards the 1C4 form when it is a non-reducing terminal. For terminal non-sulfated IdoA, the 4C1 form also contributes to the equilibrium. N.O.e. data confirm these conclusions. Possible biological implications of the conformational flexibility and the counter-ion induced changes in conformer populations are discussed.


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 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.


Pathophysiology of Haemostasis and Thrombosis | 2007

Non-Anticoagulant Heparins and Inhibition of Cancer

Benito Casu; Israel Vlodavsky; Ralph D. Sanderson

Low-molecular-weight heparins (LMWH) appear to prolong survival of patients with cancer. Such a beneficial effect is thought to be associated with interruption of molecular mechanisms involving the heparan sulfate (HS) chains of cell surface and extracellular matrix proteoglycans (HSPGs), growth factors and their receptors, heparanase, and selectins. The beneficial effects of heparin species could also be associated with their ability to release tissue factor pathway inhibitor from endothelium. The utility of heparin and LMWH as anticancer drugs is limited due to their anticoagulant properties. Non-anticoagulant heparins can be obtained either by removing chains containing the antithrombin-binding sequence, or by inactivating critical functional groups or units of this sequence. The non-anticoagulant heparins most extensively studied are regioselectively desulfated heparins and ‘glycol-split’ heparins. Some modified heparins of both types are potent inhibitors of heparanase. A number of them also attenuate metastasis in experimental models. With cancer cells overexpressing selectins, heparin-mediated inhibition of tumor cells-platelets aggregation and tumor cell interaction with the vascular endothelium appears to be the prevalent mechanism of attenuation of early stages of metastasis. The structural requirements for inhibition of growth factors, heparanase, and selectins by heparin derivatives are somewhat different for the different activities. An N-acetylated, glycol-split heparin provides an example of application of a non-anticoagulant heparin that inhibits cancer in animal models without unwanted side effects. Delivery of this compound to mice bearing established myeloma tumors dramatically blocked tumor growth and progression.


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)


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.


The FASEB Journal | 2007

P-selectin- and heparanase-dependent antimetastatic activity of non-anticoagulant heparins

Nina Hostettler; Annamaria Naggi; Giangiacomo Torri; Riva Ishai-Michaeli; Benito Casu; Israel Vlodavsky; Lubor Borsig

Vascular cell adhesion molecules, P‐ and L‐selectins, facilitate metastasis of cancer cells in mice by mediating interactions with platelets, endothelium, and leukocytes. Heparanase is an endoglycosidase that degrades heparan sulfate of extracellular matrix, thereby promoting tumor invasion and metastasis. Hep‐arin is known to efficiently attenuate metastasis in different tumor models. Here we identified modified, nonanticoagulant species of heparin that specifically inhibit selectin‐mediated cell‐cell interactions, hepara‐nase enzymatic activity, or both. We show that selective inhibition of selectin interactions or heparanase with specific heparin derivatives in mouse models of MC‐38 colon carcinoma and B16‐BL6 melanoma attenuates metastasis. Selectin‐specific heparin derivatives attenu‐ated metastasis of MC‐38 carcinoma, but heparanase‐specific derivatives had no effect, in accordance with the virtual absence of heparanase activity in these cells. Heparin derivatives had no further effect on metastasis in mice deficient in P‐ and L‐selectin, indicating that selectins are the primary targets of heparin antimeta‐static activity. Selectin‐specific and heparanase‐specific derivatives attenuated metastasis of B16‐BL6 melanomas to a similar extent. When mice were injected with a derivative containing both heparanase and selectin inhibitory activity, no additional attenuation of metastasis could be observed. Thus, selectin‐specific heparin derivatives efficiently attenuated metastasis of both tumor cell types whereas inhibition of heparanase led to reduction of metastasis only in tumor cells producing heparanase.—Hostettler N., Naggi, A., Torri, G., Ishai‐Michaeli, R., Casu, B., Vlodavsky, I., Borsig L. P‐selectin‐ and heparanase‐dependent antimetastatic activity of non‐anticoagulant heparins. FASEB J. 21, 3562–3572 (2007)

Collaboration


Dive into the Benito Casu's collaboration.

Top Co-Authors

Avatar

Giangiacomo Torri

Federal University of Rio Grande do Norte

View shared research outputs
Top Co-Authors

Avatar

Annamaria Naggi

Rappaport Faculty of Medicine

View shared research outputs
Top Co-Authors

Avatar

Marco Guerrini

Massachusetts Institute of Technology

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
Top Co-Authors

Avatar

Israel Vlodavsky

Rappaport Faculty of Medicine

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge