Network


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

Hotspot


Dive into the research topics where Raul E. Cachau is active.

Publication


Featured researches published by Raul E. Cachau.


Journal of Virology | 2011

On the Role of the SP1 Domain in HIV-1 Particle Assembly: a Molecular Switch?

Siddhartha A. K. Datta; Lakew G. Temeselew; Rachael M. Crist; Ferri Soheilian; Anne Kamata; Jane Mirro; Demetria Harvin; Kunio Nagashima; Raul E. Cachau; Alan Rein

ABSTRACT Expression of a retroviral protein, Gag, in mammalian cells is sufficient for assembly of immature virus-like particles (VLPs). VLP assembly is mediated largely by interactions between the capsid (CA) domains of Gag molecules but is facilitated by binding of the nucleocapsid (NC) domain to nucleic acid. We have investigated the role of SP1, a spacer between CA and NC in HIV-1 Gag, in VLP assembly. Mutational analysis showed that even subtle changes in the first 4 residues of SP1 destroy the ability of Gag to assemble correctly, frequently leading to formation of tubes or other misassembled structures rather than proper VLPs. We also studied the conformation of the CA-SP1 junction region in solution, using both molecular dynamics simulations and circular dichroism. Consonant with nuclear magnetic resonance (NMR) studies from other laboratories, we found that SP1 is nearly unstructured in aqueous solution but undergoes a concerted change to an α-helical conformation when the polarity of the environment is reduced by addition of dimethyl sulfoxide (DMSO), trifluoroethanol, or ethanol. Remarkably, such a coil-to-helix transition is also recapitulated in an aqueous medium at high peptide concentrations. The exquisite sensitivity of SP1 to mutational changes and its ability to undergo a concentration-dependent structural transition raise the possibility that SP1 could act as a molecular switch to prime HIV-1 Gag for VLP assembly. We suggest that changes in the local environment of SP1 when Gag oligomerizes on nucleic acid might trigger this switch.


Proteins | 2005

QM/MM modeling the Ras–GAP catalyzed hydrolysis of guanosine triphosphate

Bella L. Grigorenko; Alexander V. Nemukhin; Igor A. Topol; Raul E. Cachau; Stanley K. Burt

The mechanism of the hydrolysis reaction of guanosine triphosphate (GTP) by the protein complex Ras–GAP (p21ras – p120GAP) has been modeled by the quantum mechanical—molecular mechanical (QM/MM) and ab initio quantum calculations. Initial geometry configurations have been prompted by atomic coordinates of a structural analog (PDBID:1WQ1). It is shown that the minimum energy reaction path is consistent with an assumption of two‐step chemical transformations. At the first stage, a unified motion of Arg789 of GAP, Gln61, Thr35 of Ras, and the lytic water molecule results in a substantial spatial separation of the γ‐phosphate group of GTP from the rest of the molecule (GDP). This phase of hydrolysis process proceeds through the low‐barrier transition state TS1. At the second stage, Gln61 abstracts and releases protons within the subsystem including Gln61, the lytic water molecule and the γ‐phosphate group of GTP through the corresponding transition state TS2. Direct quantum calculations show that, in this particular environment, the reaction GTP + H2O → GDP + H2PO  4− can proceed with reasonable activation barriers of less than 15 kcal/mol at every stage. This conclusion leads to a better understanding of the anticatalytic effect of cancer‐causing mutations of Ras, which has been debated in recent years. Proteins 2005.


Proceedings of the National Academy of Sciences of the United States of America | 2008

Quantum model of catalysis based on a mobile proton revealed by subatomic x-ray and neutron diffraction studies of h-aldose reductase

Matthew P. Blakeley; Federico Ruiz; Raul E. Cachau; Isabelle Hazemann; Flora Meilleur; Andre Mitschler; Stephan L. Ginell; Pavel V. Afonine; Oscar N. Ventura; Alexandra Cousido-Siah; Michael Haertlein; Andrzej Joachimiak; Dean A. A. Myles; Alberto Podjarny

We present results of combined studies of the enzyme human aldose reductase (h-AR, 36 kDa) using single-crystal x-ray data (0.66 Å, 100K; 0.80 Å, 15K; 1.75 Å, 293K), neutron Laue data (2.2 Å, 293K), and quantum mechanical modeling. These complementary techniques unveil the internal organization and mobility of the hydrogen bond network that defines the properties of the catalytic engine, explaining how this promiscuous enzyme overcomes the simultaneous requirements of efficiency and promiscuity offering a general mechanistic view for this class of enzymes.


Cellular and Molecular Life Sciences | 2004

Subatomic and atomic crystallographic studies of aldose reductase: implications for inhibitor binding

Alberto Podjarny; Raul E. Cachau; Thomas R. Schneider; M. Van Zandt; A. Joachimiak

The determination of several of aldose reductase-inhibitor complexes at subatomic resolution has revealed new structural details, including the specific interatomic contacts involved in inhibitor binding. In this article, we review the structures of the complexes of ALR2 with IDD 594 (resolution: 0.66 Å, IC50 (concentration of the inhibitor that produced half-maximal effect): 30 nM, space group: P21), IDD 393 (resolution: 0.90 Å, IC50: 6 nM, space group: P1), fidarestat (resolution: 0.92 Å, IC50: 9 nM, space group: P21) and minalrestat (resolution: 1.10 Å, IC50: 73 nM, space group: P1). The structures are compared and found to be highly reproductible within the same space group (root mean square (RMS) deviations: 0.15 ∼ 0.3 Å). The mode of binding of the carboxylate inhibitors IDD 594 and IDD 393 is analysed. The binding of the carboxylate head can be accurately determined by the subatomic resolution structures, since both the protonation states and the positions of the atoms are very precisely known. The differences appear in the binding in the specificity pocket. The high-resolution structures explain the differences in IC50, which are confirmed both experimentally by mass spectrometry measures of VC50 and theoretically by free energy perturbation calculations. The binding of the cyclic imide inhibitors fidarestat and minalrestat is also described, focusing on the observation of a Cl- ion which binds simultaneously with fidarestat. The presence of this anion, binding also to the active site residue His110, leads to a mechanism in which the inhibitor can bind in a neutral state and then become charged inside the active site pocket. This mechanism can explain the excellent in vivo properties of cyclic imide inhibitors. In summary, the complete and detailed information supplied by the subatomic resolution structures can explain the differences in binding energy of the different inhibitors.


International Journal of Nanomedicine | 2012

Nanoinformatics: a new area of research in nanomedicine

Victor Maojo; Martin Fritts; Diana de la Iglesia; Raul E. Cachau; Miguel García-Remesal; Joyce A. Mitchell; Casimir A. Kulikowski

Over a decade ago, nanotechnologists began research on applications of nanomaterials for medicine. This research has revealed a wide range of different challenges, as well as many opportunities. Some of these challenges are strongly related to informatics issues, dealing, for instance, with the management and integration of heterogeneous information, defining nomenclatures, taxonomies and classifications for various types of nanomaterials, and research on new modeling and simulation techniques for nanoparticles. Nanoinformatics has recently emerged in the USA and Europe to address these issues. In this paper, we present a review of nanoinformatics, describing its origins, the problems it addresses, areas of interest, and examples of current research initiatives and informatics resources. We suggest that nanoinformatics could accelerate research and development in nanomedicine, as has occurred in the past in other fields. For instance, biomedical informatics served as a fundamental catalyst for the Human Genome Project, and other genomic and –omics projects, as well as the translational efforts that link resulting molecular-level research to clinical problems and findings.


Toxicological Sciences | 2009

Metal Ions-Stimulated Iron Oxidation in Hydroxylases Facilitates Stabilization of HIF-1α Protein

Monika Kaczmarek; Raul E. Cachau; Igor A. Topol; Kazimierz S. Kasprzak; Andy Ghio; Konstantin Salnikow

The exposure of cells to several metal ions stabilizes HIF-1 alpha protein. However, the molecular mechanisms are not completely understood. They may involve inhibition of hydroxylation by either substitution of iron by metal ions or by iron oxidation in the hydroxylases. Here we provide evidence supporting the latter mechanism. We show that HIF-1 alpha stabilization in human lung epithelial cells occurred following exposure to various metal and metalloid ions, including those that cannot substitute for iron in the hydroxylases. In each case addition of the reducing agent ascorbic acid (AA)* abolished HIF-1 alpha protein stabilization. To better understand the role of iron oxidation in hydroxylase inhibition and to define the role of AA in the enzyme recovery we applied molecular modeling techniques. Our results indicate that the energy required for iron substitution by Ni(II) in the enzyme is high and unlikely to be achieved in a biological system. Additionally, computer modeling allowed us to identify a tridentate coordination of AA with the enzyme-bound iron, which explains the specific demand for AA as the iron reductant. Thus, the stabilization of HIF-1 alpha by numerous metal ions that cannot substitute for iron in the enzyme, the alleviation of this effect by AA, and our computer modeling data support the hypothesis of iron oxidation in the hydroxylases following exposure to metal ions.


Biophysical Journal | 1987

Ion channels in southern bean mosaic virus capsid.

Abelardo M. Silva; Raul E. Cachau; Daniel J. Goldstein

The study of southern bean mosaic virus protein coat high resolution model revealed a structure with properties of a natural protein-ion channel. Coat protein pentamers form a 30-A long channel and the amino acid composition of its wall bears some homology with the pentameric structure proposed for the nicotinic acetylcholine receptor channel. Ion transport properties were analyzed by computing ion-protein interaction energies on the basis of quantum chemistry methods. Energy maps show a channel attractive for cations, fully permeable to Li(+) and a narrow barrier for other cations and water. The energy profiles found are similar to the profiles determined for the K(+) channel of the sarcoplasmic reticulum. Comparisons with other icosahedral virus structures, including picornaviruses, suggest that ion channels would be a common feature of viral capsids. Biological roles for these channels are proposed.


Chemical Physics Letters | 1999

Density functional and coupled-cluster calculations of isodesmic reactions involving fluorine oxides

Oscar N. Ventura; Raul E. Cachau; Martina Kieninger

Abstract Some isodesmic reactions, involving the fluorine oxide radical FO, have been studied employing density functional theory (DFT) and coupled-cluster (CC) calculations with an extended, uncontracted basis set. It is shown that CCSD(T) calculations can give more accurate enthalpies of reaction than DFT in some of the non-isodesmic reactions. DFT, however, gives more accurate results than CCSD(T) for the isodesmic reactions considered.


Nanomedicine: Nanotechnology, Biology and Medicine | 2014

Inhibition of phosphoinositol 3 kinase contributes to nanoparticle-mediated exaggeration of endotoxin-induced leukocyte procoagulant activity

Anna N. Ilinskaya; Sonny Man; Anil K. Patri; Jeffrey D. Clogston; Rachael M. Crist; Raul E. Cachau; Scott E. McNeil; Marina A. Dobrovolskaia

AIM Disseminated intravascular coagulation is an increasing concern for certain types of engineered nanomaterials. Recent studies have shed some light on the nanoparticle physicochemical properties contributing to this toxicity; however, the mechanisms are poorly understood. Leukocyte procoagulant activity (PCA) is a key factor contributing to the initiation of this toxicity. We have previously reported on the exaggeration of endotoxin-induced PCA by cationic dendrimers. Herein, we report an effort to discern the mechanism. MATERIALS & METHODS Poly(amidoamine) dendrimers with various sizes and surface functionalities were studied in vitro by the recalcification test, flow cytometry and other relevant assays. RESULTS & CONCLUSION Cationic dendrimers exaggerated endotoxin-induced PCA, but their anionic or neutral counterparts did not; the cationic charge prompts this phenomenon, but different cationic surface chemistries do not influence it. Cationic dendrimers and endotoxin differentially affect the PCA complex. The inhibition of phosphoinositol 3 kinase by dendrimers contributes to the exaggeration of the endotoxin-induced PCA.


Computing | 2012

Nanoinformatics: developing new computing applications for nanomedicine

Victor Maojo; Martin Fritts; Fernando Martín-Sánchez; Diana de la Iglesia; Raul E. Cachau; Miguel García-Remesal; José Crespo; Joyce A. Mitchell; Alberto Anguita; Nathan A. Baker; José María Barreiro; Sonia E. Benítez; Guillermo de la Calle; Julio C. Facelli; Peter Ghazal; Antoine Geissbuhler; Fernando D. González-Nilo; Norbert Graf; Pierre Grangeat; Isabel Hermosilla; Rada Hussein; Josipa Kern; Sabine Koch; Yannick Legré; Victoria López-Alonso; Guillermo López-Campos; Luciano Milanesi; Vassilis Moustakis; Cristian R. Munteanu; Paula Otero

Nanoinformatics has recently emerged to address the need of computing applications at the nano level. In this regard, the authors have participated in various initiatives to identify its concepts, foundations and challenges. While nanomaterials open up the possibility for developing new devices in many industrial and scientific areas, they also offer breakthrough perspectives for the prevention, diagnosis and treatment of diseases. In this paper, we analyze the different aspects of nanoinformatics and suggest five research topics to help catalyze new research and development in the area, particularly focused on nanomedicine. We also encompass the use of informatics to further the biological and clinical applications of basic research in nanoscience and nanotechnology, and the related concept of an extended “nanotype” to coalesce information related to nanoparticles. We suggest how nanoinformatics could accelerate developments in nanomedicine, similarly to what happened with the Human Genome and other -omics projects, on issues like exchanging modeling and simulation methods and tools, linking toxicity information to clinical and personal databases or developing new approaches for scientific ontologies, among many others.

Collaboration


Dive into the Raul E. Cachau's collaboration.

Top Co-Authors

Avatar

Oscar N. Ventura

Autonomous University of Barcelona

View shared research outputs
Top Co-Authors

Avatar

Stanley K. Burt

Science Applications International Corporation

View shared research outputs
Top Co-Authors

Avatar

Martina Kieninger

German Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar

A. Joachimiak

Argonne National Laboratory

View shared research outputs
Top Co-Authors

Avatar

Igor A. Topol

Science Applications International Corporation

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

John W. Erickson

Science Applications International Corporation

View shared research outputs
Top Co-Authors

Avatar

R. Sanishvili

Argonne National Laboratory

View shared research outputs
Top Co-Authors

Avatar

A. Mitschler

University of Strasbourg

View shared research outputs
Top Co-Authors

Avatar

A. Podjarny

University of Strasbourg

View shared research outputs
Researchain Logo
Decentralizing Knowledge