Network


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

Hotspot


Dive into the research topics where Dolores González-Pacanowska is active.

Publication


Featured researches published by Dolores González-Pacanowska.


Antimicrobial Agents and Chemotherapy | 2007

Kinetic Characterization of Squalene Synthase from Trypanosoma cruzi: Selective Inhibition by Quinuclidine Derivatives

Marco Sealey-Cardona; Simon Cammerer; Simon Jones; Luis M. Ruiz-Pérez; Reto Brun; Ian H. Gilbert; Julio A. Urbina; Dolores González-Pacanowska

ABSTRACT The biosynthesis of sterols is a major route for the development of antitrypanosomals. Squalene synthase (SQS) catalyzes the first step committed to the biosynthesis of sterols within the isoprenoid pathway, and several inhibitors of the enzyme have selective antitrypanosomal activity both in vivo and in vitro. The enzyme from Trypanosoma cruzi is a 404-amino-acid protein with a clearly identifiable membrane-spanning region. In an effort to generate soluble recombinant enzyme, we have expressed in Escherichia coli several truncated versions of T. cruzi SQS with a His tag attached to the amino terminus. Deletions of both the amino- and carboxyl-terminal regions generated active and soluble forms of the enzyme. The highest levels of soluble protein were achieved when 24 and 36 amino acids were eliminated from the amino and carboxyl regions, respectively, yielding a protein of 41.67 kDa. The Michaelis-Menten constants of the purified enzyme for farnesyl diphosphate and NAD (NADPH) were 5.25 and 23.34 μM, respectively, whereas the Vmax was 1,428.56 nmol min−1mg−1. Several quinuclidine derivatives with antiprotozoal activity in vitro were found to be selective inhibitors of recombinant T. cruzi SQS in comparative assays with the human enzyme, with 50% inhibitory concentration values in the nanomolar range. These data suggest that selective inhibition of T. cruzi SQS may be an efficient strategy for the development of new antitrypanosomal agents.


Trends in Parasitology | 2010

Collaborative actions in anti-trypanosomatid chemotherapy with partners from disease endemic areas.

Jean-Claude Dujardin; Dolores González-Pacanowska; Simon L. Croft; Ole F. Olesen; Gerald F. Späth

The protozoan diseases leishmaniasis, human African trypanosomiasis and Chagas disease are responsible for substantial global morbidity, mortality and economic adversity in tropical and subtropical regions. In most countries, existing strategies for control and treatment are either failing or under serious threat. Environmental changes, drug resistance and immunosuppression contribute to the emergence and spread of these diseases. In the absence of safe and efficient vaccines, chemotherapy, together with vector control, remains the most important measures to control trypanosomatid diseases. Here, we review current limitations of anti-trypanosomatid chemotherapy and describe new efforts to safeguard existing treatments and to identify novel drug leads through the three multinational and interdisciplinary European Union Framework Programmes for Research and Technological Development (FP7) funded consortia KALADRUG-R, TRYPOBASE, and LEISHDRUG.


Protein Science | 2008

Kinetic properties and inhibition of the dimeric dUTPase‐dUDPase from Leishmania major

Fernando Hidalgo-Zarco; Ana G. Camacho; Victor Bernier-Villamor; Johan Nord; Luis M. Ruiz-Pérez; Dolores González-Pacanowska

Kinetic properties of the dimeric enzyme dUTPase from Leishmania major were studied using a continuous spectrophotometric method. dUTP was the natural substrate and dUMP and PPi the products of the hydrolysis. The trypanosomatid enzyme exhibited a low Km value for dUTP (2.11 μM), a kcat of 49 s−1, strict Michaelis–Menten kinetics and is a potent catalyst of dUDP hydrolysis, whereas in other dUTPases described, this compound acts as a competitive inhibitor. Discrimination is achieved for the base and sugar moiety showing specificity constants for different dNTPs similar to those of bacterial, viral, and human enzymes. In the alkaline range, the Km for dUTP increases with the dissociation of ionizable groups showing pKa values of 8.8, identified as the uracil moiety of dUTP and 10, whereas in the acidic range, Km is regulated by an enzyme residue exhibiting a pKa of 7.1. Activity is strongly inhibited by the nucleoside triphosphate analog α‐β‐imido‐dUTP, indicating that the enzyme can bind triphosphate analogs. The existence of specific inhibition and the apparent structural and kinetic differences (reflected in different binding strength of dNTPs) with other eukaryotic dUTPases suggest that the present enzyme might be exploited as a target for new drugs against leishmaniasis.


Journal of Biomolecular Screening | 2015

High-Throughput Screening Platform for Natural Product–Based Drug Discovery Against 3 Neglected Tropical Diseases Human African Trypanosomiasis, Leishmaniasis, and Chagas Disease

Frederick Annang; Guiomar Pérez-Moreno; R. García-Hernández; C. Cordon-Obras; Jesús Martín; José R. Tormo; Lorena Rodriguez; N de Pedro; V. Gómez-Pérez; M. Valente; Fernando Reyes; Olga Genilloud; Francisca Vicente; S. Castanys; Luis M. Ruiz-Pérez; M. Navarro; F. Gamarro; Dolores González-Pacanowska

African trypanosomiasis, leishmaniasis, and Chagas disease are 3 neglected tropical diseases for which current therapeutic interventions are inadequate or toxic. There is an urgent need to find new lead compounds against these diseases. Most drug discovery strategies rely on high-throughput screening (HTS) of synthetic chemical libraries using phenotypic and target-based approaches. Combinatorial chemistry libraries contain hundreds of thousands of compounds; however, they lack the structural diversity required to find entirely novel chemotypes. Natural products, in contrast, are a highly underexplored pool of unique chemical diversity that can serve as excellent templates for the synthesis of novel, biologically active molecules. We report here a validated HTS platform for the screening of microbial extracts against the 3 diseases. We have used this platform in a pilot project to screen a subset (5976) of microbial extracts from the MEDINA Natural Products library. Tandem liquid chromatography–mass spectrometry showed that 48 extracts contain potentially new compounds that are currently undergoing de-replication for future isolation and characterization. Known active components included actinomycin D, bafilomycin B1, chromomycin A3, echinomycin, hygrolidin, and nonactins, among others. The report here is, to our knowledge, the first HTS of microbial natural product extracts against the above-mentioned kinetoplastid parasites.


PLOS Pathogens | 2014

Squalene synthase as a target for Chagas disease therapeutics.

Na Shang; Qian Li; Tzu-Ping Ko; Hsiu Chien Chan; Jikun Li; Yingying Zheng; Chun Hsiang Huang; Feifei Ren; Chun Chi Chen; Zhen Zhu; Melina Galizzi; Zhu Hong Li; Carlos A. Rodrígues-Poveda; Dolores González-Pacanowska; Phercyles Veiga-Santos; Tecia Maria Ulisses de Carvalho; Wanderley de Souza; Julio A. Urbina; Andrew H.-J. Wang; Roberto Docampo; Kai Li; Yi Liang Liu; Eric Oldfield; Rey-Ting Guo

Trypanosomatid parasites are the causative agents of many neglected tropical diseases and there is currently considerable interest in targeting endogenous sterol biosynthesis in these organisms as a route to the development of novel anti-infective drugs. Here, we report the first x-ray crystallographic structures of the enzyme squalene synthase (SQS) from a trypanosomatid parasite, Trypanosoma cruzi, the causative agent of Chagas disease. We obtained five structures of T. cruzi SQS and eight structures of human SQS with four classes of inhibitors: the substrate-analog S-thiolo-farnesyl diphosphate, the quinuclidines E5700 and ER119884, several lipophilic bisphosphonates, and the thiocyanate WC-9, with the structures of the two very potent quinuclidines suggesting strategies for selective inhibitor development. We also show that the lipophilic bisphosphonates have low nM activity against T. cruzi and inhibit endogenous sterol biosynthesis and that E5700 acts synergistically with the azole drug, posaconazole. The determination of the structures of trypanosomatid and human SQS enzymes with a diverse set of inhibitors active in cells provides insights into SQS inhibition, of interest in the context of the development of drugs against Chagas disease.


Molecular and Biochemical Parasitology | 1994

Cloning and expression of the dihydrofolate reductase-thymidylate synthase gene from Trypanosoma cruzi

Pedro A. Reche; Rosalia Arrebola; Asunción Olmo; Daniel V. Santi; Dolores González-Pacanowska; Luis M. Ruiz-Pérez

We have cloned, sequenced and expressed the Trypanosoma cruzi gene encoding the bifunctional protein dihydrofolate reductase-thymidylate synthase (DHFR-TS). The strategy followed for the isolation of positive clones from a genomic library was based on the construction of a probe by the amplification of highly conserved sequences of the TS domain by the polymerase chain reaction. Translation of the open reading frame of 1563 bp yields a polypeptide of 521 amino acids with a molecular mass of 58829 Da. For heterologous expression of T. cruzi DHFR-TS in Escherichia coli, the entire coding sequence was amplified by polymerase chain reaction and cloned into the plasmid vector pKK223.3. The presence of catalytically active DHFR-TS was demonstrated by complementation of the Thy- E. coli strain chi 2913 and the DHFR- Thy- E. coli strain PA414. The gene is expressed as an active protein which constitutes approximately 2% of the total cell soluble protein. Recombinant bifunctional enzyme and the DHFR domain have been purified by methotrexate-Sepharose chromatography to yield 1-2 mg of active DHFR-TS per litre of culture. Southern and electrophoretic analyses using the coding sequence as probe indicated that the T. cruzi enzyme is encoded by a single copy gene which maps to two bands of approximately 990 kb and 1047 kb. It appears that T. cruzi is diploid for the DHFR-TS gene which is located on two different-sized homologous chromosomes.


Journal of Biological Chemistry | 2011

The Crystal Structure of the Leishmania major Deoxyuridine Triphosphate Nucleotidohydrolase in Complex with Nucleotide Analogues, dUMP, and Deoxyuridine

Glyn R. Hemsworth; Olga V. Moroz; Mark J. Fogg; Benjamin Scott; Cristina Bosch-Navarrete; Dolores González-Pacanowska; Keith S. Wilson

Members of the Leishmania genus are the causative agents of the life-threatening disease leishmaniasis. New drugs are being sought due to increasing resistance and adverse side effects with current treatments. The knowledge that dUTPase is an essential enzyme and that the all α-helical dimeric kinetoplastid dUTPases have completely different structures compared with the trimeric β-sheet type dUTPase possessed by most organisms, including humans, make the dimeric enzymes attractive drug targets. Here, we present crystal structures of the Leishmania major dUTPase in complex with substrate analogues, the product dUMP and a substrate fragment, and of the homologous Campylobacter jejuni dUTPase in complex with a triphosphate substrate analogue. The metal-binding properties of both enzymes are shown to be dependent upon the ligand identity, a previously unseen characteristic of this family. Furthermore, structures of the Leishmania enzyme in the presence of dUMP and deoxyuridine coupled with tryptophan fluorescence quenching indicate that occupation of the phosphate binding region is essential for induction of the closed conformation and hence for substrate binding. These findings will aid in the development of dUTPase inhibitors as potential new lead anti-trypanosomal compounds.


Eukaryotic Cell | 2006

Farnesyl Diphosphate Synthase Is a Cytosolic Enzyme in Leishmania major Promastigotes and Its Overexpression Confers Resistance to Risedronate

Aurora Ortiz-Gómez; C. Jimenez; Antonio M. Estévez; Juana Carrero-Lérida; Luis M. Ruiz-Pérez; Dolores González-Pacanowska

ABSTRACT Farnesyl diphosphate synthase is the most likely molecular target of aminobisphosphonates (e.g., risedronate), a set of compounds that have been shown to have antiprotozoal activity both in vitro and in vivo. This protein, together with other enzymes involved in isoprenoid biosynthesis, is an attractive drug target, yet little is known about the compartmentalization of the biosynthetic pathway. Here we show the intracellular localization of the enzyme in wild-type Leishmania major promastigote cells and in transfectants overexpressing farnesyl diphosphate synthase by using purified antibodies generated towards a homogenous recombinant Leishmania major farnesyl diphosphate synthase protein. Indirect immunofluorescence, together with immunoelectron microscopy, indicated that the enzyme is mainly located in the cytoplasm of both wild-type cells and transfectants. Digitonin titration experiments also confirmed this observation. Hence, while the initial step of isoprenoid biosynthesis catalyzed by 3-hydroxy-3-methylglutaryl-coenzyme A reductase is located in the mitochondrion, synthesis of farnesyl diphosphate by farnesyl diphosphate synthase is a cytosolic process. Leishmania major promastigote transfectants overexpressing farnesyl diphosphate synthase were highly resistant to risedronate, and the degree of resistance correlated with the increase in enzyme activity. Likewise, when resistance was induced by stepwise selection with the drug, the resulting resistant promastigotes exhibited increased levels of farnesyl diphosphate synthase. The overproduction of protein under different conditions of exposure to risedronate further supports the hypothesis that this enzyme is the main target of aminobisphosphonates in Leishmania cells.


Biochemical Journal | 2000

Characterization and regulation of Leishmania major 3-hydroxy-3-methylglutaryl-CoA reductase.

Andrea Montalvetti; Javier Peña-Diaz; Ramon Hurtado; Luis M. Ruiz-Pérez; Dolores González-Pacanowska

In eukaryotes the enzyme 3-hydroxy-3-methylglutaryl CoA (HMG-CoA) reductase catalyses the synthesis of mevalonic acid, a common precursor to all isoprenoid compounds. Here we report the isolation and overexpression of the gene coding for HMG-CoA reductase from Leishmania major. The protein from Leishmania lacks the membrane domain characteristic of eukaryotic cells but exhibits sequence similarity with eukaryotic reductases. Highly purified protein was achieved by ammonium sulphate precipitation followed by chromatography on hydroxyapatite. Kinetic parameters were determined for the protozoan reductase, obtaining K(m) values for the overall reaction of 40.3+/-5.8 microM for (R,S)-HMG-CoA and 81.4+/-5.3 microM for NADPH; V(max) was 33.55+/-1.8 units x mg(-1). Gel-filtration experiments suggested an apparent molecular mass of 184 kDa with subunits of 46 kDa. Finally, in order to achieve a better understanding of the role of this enzyme in trypanosomatids, the effect of possible regulators of isoprenoid biosynthesis in cultured promastigote cells was studied. Neither mevalonic acid nor serum sterols appear to modulate enzyme activity whereas incubation with lovastatin results in significant increases in the amount of reductase protein. Western- and Northern-blot analyses indicate that this activation is apparently performed via post-transcriptional control.


Journal of Medicinal Chemistry | 2012

Synthesis and evaluation of α-thymidine analogues as novel antimalarials.

Huaqing Cui; Juana Carrero-Lérida; Ana P. G. Silva; Jean L. Whittingham; James A. Brannigan; Luis M. Ruiz-Pérez; Kevin D. Read; Keith S. Wilson; Dolores González-Pacanowska; Ian H. Gilbert

Plasmodium falciparum thymidylate kinase (PfTMPK) is a key enzyme in pyrimidine nucleotide biosynthesis. 3-Trifluoromethyl-4-chloro-phenyl-urea-α-thymidine has been reported as an inhibitor of Mycobacterium tuberculosis TMPK (MtTMPK). Starting from this point, we designed, synthesized and evaluated a number of thymidine analogues as antimalarials. Both 5′-urea-α- and β-thymidine derivatives were moderate inhibitors of PfTMPK and furthermore showed moderate inhibition of parasite growth. The structure of several enzyme–inhibitor complexes provides a basis for improved inhibitor design. However, we found that certain 5′-urea-α-thymidine analogues had antimalarial activity where inhibition of PfTMPK is not the major mode of action. Optimization of this series resulted in a compound with potent antimalarial activity (EC50 = 28 nM; CC50 = 29 μM).

Collaboration


Dive into the Dolores González-Pacanowska's collaboration.

Top Co-Authors

Avatar

Luis M. Ruiz-Pérez

Spanish National Research Council

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Guiomar Pérez-Moreno

Spanish National Research Council

View shared research outputs
Top Co-Authors

Avatar

Víctor M. Castillo-Acosta

Spanish National Research Council

View shared research outputs
Top Co-Authors

Avatar

Antonio E. Vidal

Spanish National Research Council

View shared research outputs
Top Co-Authors

Avatar

Victor Bernier-Villamor

Spanish National Research Council

View shared research outputs
Top Co-Authors

Avatar

Reto Brun

Swiss Tropical and Public Health Institute

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Marcel Kaiser

Swiss Tropical and Public Health Institute

View shared research outputs
Researchain Logo
Decentralizing Knowledge