Network


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

Hotspot


Dive into the research topics where José M. Souza is active.

Publication


Featured researches published by José M. Souza.


Free Radical Biology and Medicine | 2008

Protein tyrosine nitration--functional alteration or just a biomarker?

José M. Souza; Gonzalo Peluffo; Rafael Radi

Protein 3-nitrotyrosine is a posttranslational modification found in many pathological conditions from acute to chronic diseases. Could 3-nitrotyrosine formation participate on the basis of these diseases or is it just a marker connected with the associated nitroxidative stress? In vitro and in vivo data, including proteomic research, show that protein tyrosine nitration is a selective process where only a small amount of proteins is found nitrated and one or a few tyrosine residues are modified in each. Accumulating data suggest a strong link between protein 3-nitrotyrosine and the mechanism involved in disease development. In this review, we analyze the factors determining protein 3-nitrotyrosine formation, the functional and biological outcome associated with protein tyrosine nitration, and the fate of the nitrated proteins.


FEBS Letters | 2000

Chaperone‐like activity of synucleins

José M. Souza; Benoit I. Giasson; Virginia M.-Y. Lee; Harry Ischiropoulos

Synucleins are a family of small proteins that are predominantly expressed in neurons. The functions of the synucleins are not entirely understood, but they have been implicated in the pathogenesis of several neurodegenerative diseases. Our data show that α‐, β‐ or γ‐synuclein suppresses the aggregation of thermally denatured alcohol dehydrogenase and chemically denatured insulin. The A53T but not the A30P mutant α‐synuclein was able to inhibit the aggregation of insulin and the chaperone‐like activity of α‐synuclein was lost upon removal of its C‐terminal residues 98–140. These results demonstrate that synucleins with the exception of the A30P mutant possess chaperone‐like activity.


Journal of Neurochemistry | 2001

Oxidative post-translational modifications of α-synuclein in the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) mouse model of Parkinson's disease

Serge Przedborski; Qiping Chen; Miquel Vila; Benoit I. Giasson; Ruth Djaldatti; Slobodanka Vukosavic; José M. Souza; Vernice Jackson-Lewis; Virginia M.-Y. Lee; Harry Ischiropoulos

Structural and functional alterations of α‐synuclein is a presumed culprit in the demise of dopaminergic neurons in Parkinsons disease (PD). α‐Synuclein mutations are found in familial but not in sporadic PD, raising the hypothesis that effects similar to those of familial PD‐linked α‐synuclein mutations may be achieved by oxidative post‐translational modifications. Here, we show that wild‐type α‐synuclein is a selective target for nitration following peroxynitrite exposure of stably transfected HEK293 cells. Nitration of α‐synuclein also occurs in the mouse striatum and ventral midbrain following administration of the parkinsonian neurotoxin 1‐methyl‐4‐phenyl‐1,2,3,6‐tetrahydropyridine (MPTP). Conversely, β‐synuclein and synaptophysin were not nitrated in MPTP‐intoxicated mice. Our data demonstrate that α‐synuclein is a target for tyrosine nitration, which, by disrupting its biophysical properties, may be relevant to the putative role of α‐synuclein in the neurodegeneration associated with MPTP toxicity and with PD.


Journal of Biological Chemistry | 2004

Pro-thrombotic State Induced by Post-translational Modification of Fibrinogen by Reactive Nitrogen Species

Caryn Vadseth; José M. Souza; Leonor Thomson; Amy Seagraves; Chandrasekaran Nagaswami; Tomas Scheiner; Jim Torbet; Gaston Vilaire; Joel S. Bennett; Juan Carlos Murciano; Vladimir R. Muzykantov; Marc S. Penn; Stanley L. Hazen; John W. Weisel; Harry Ischiropoulos

Formation of nitric oxide-derived oxidants has been linked to development of atherosclerosis and associated thrombotic complications. Although systemic levels of protein nitrotyrosine predict risk for coronary artery disease, neither specific proteins targeted for modification nor functional consequences that might contribute to disease pathogenesis have been defined. Here we report a selective increase in circulating levels of nitrated fibrinogen in patients with coronary artery disease. Exposure of fibrinogen to nitrating oxidants, including those produced by the myeloperoxidase-hydrogen peroxide-nitrite system, significantly accelerates clot formation and factor XIII cross-linking, whereas exposure of fibrinogen to non-nitrating oxidants decelerates clot formation. Clots formed with fibrinogen exposed to nitrating oxidants are composed of large bundles made from twisted thin fibrin fibers with increased permeation and a decrease in storage modulus G′ value, suggesting that these clots could be easily deformed by mechanical stresses. In contrast, clots formed with fibrinogen exposed to non-nitrating oxidants showed decreased permeation with normal architecture. Fibrinogen modified by exposure to physiologic nitration systems demonstrated no difference in the rate of plasmin-induced clot lysis, platelet aggregation, or binding. Thus, increased levels of fibrinogen nitration may lead to a pro-thrombotic state via acceleration in formation of fibrin clots. The present results may account, in part, for the association between nitrative stress and risk for coronary artery disease.


Journal of the American Chemical Society | 2008

Site-specific interactions of Cu(II) with α and β-synuclein: bridging the molecular gap between metal binding and aggregation.

Andres Binolfi; Gonzalo R. Lamberto; Rosario Durán; Liliana Quintanar; Carlos W. Bertoncini; José M. Souza; Carlos Cerveñansky; Markus Zweckstetter; Christian Griesinger; Claudio O. Fernández

The aggregation of alpha-synuclein (AS) is a critical step in the etiology of Parkinsons disease (PD) and other neurodegenerative synucleinopathies. Protein-metal interactions play a critical role in AS aggregation and might represent the link between the pathological processes of protein aggregation and oxidative damage. Our previous studies established a hierarchy in AS-metal ion interactions, where Cu(II) binds specifically to the protein and triggers its aggregation under conditions that might be relevant for the development of PD. In this work, we have addressed unresolved structural details related to the binding specificity of Cu(II) through the design of site-directed and domain-truncated mutants of AS and by the characterization of the metal-binding features of its natural homologue beta-synuclein (BS). The structural properties of the Cu(II) complexes were determined by the combined application of nuclear magnetic resonance, electron paramagnetic resonance, UV-vis, circular dichroism spectroscopy, and matrix-assisted laser desorption ionization mass spectrometry (MALDI MS). Two independent, noninteracting copper-binding sites with significantly different affinities for the metal ion were detected in the N-terminal regions of AS and BS. MALDI MS provided unique evidence for the direct involvement of Met1 as the primary anchoring residue for Cu(II) in both proteins. Comparative spectroscopic analysis of the two proteins allowed us to deconvolute the Cu(II) binding modes and unequivocally assign the higher-affinity site to the N-terminal amino group of Met1 and the lower-affinity site to the imidazol ring of the sole His residue. Through the use of competitive chelators, the affinity of the first equivalent of bound Cu(II) was accurately determined to be in the submicromolar range for both AS and BS. Our results prove that Cu(II) binding in the C-terminal region of synucleins represents a nonspecific, very low affinity process. These new insights into the bioinorganic chemistry of PD are central to an understanding of the role of Cu(II) in the fibrillization process of AS and have implications for the molecular mechanism by which BS might inhibit AS amyloid assembly.


Proceedings of the National Academy of Sciences of the United States of America | 2001

Dynamic regulation of metabolism and respiration by endogenously produced nitric oxide protects against oxidative stress.

Evgenia Paxinou; Marie Weisse; Qiping Chen; José M. Souza; Caryn Hertkorn; Mary A. Selak; Evgueni Daikhin; Marc Yudkoff; Grzegorz Sowa; William C. Sessa; Harry Ischiropoulos

One of the many biological functions of nitric oxide is the ability to protect cells from oxidative stress. To investigate the potential contribution of low steady state levels of nitric oxide generated by endothelial nitric oxide synthase (eNOS) and the mechanisms of protection against H2O2, spontaneously transformed human ECV304 cells, which normally do not express eNOS, were stably transfected with a green fluorescent-tagged eNOS cDNA. The eNOS-transfected cells were found to be resistant to injury and delayed death following a 2-h exposure to H2O2 (50–150 μM). Inhibition of nitric oxide synthesis abolished the protective effect against H2O2 exposure. The ability of nitric oxide to protect cells depended on the presence of respiring mitochondria as ECV304+eNOS cells with diminished mitochondria respiration (ρ−) are injured to the same extent as nontransfected ECV304 cells and recovery of mitochondrial respiration restores the ability of nitric oxide to protect against H2O2-induced death. Nitric oxide also found to have a profound effect in cell metabolism, because ECV304+eNOS cells had lower steady state levels of ATP and higher utilization of glucose via the glycolytic pathway than ECV304 cells. However, the protective effect of nitric oxide against H2O2 exposure is not reproduced in ECV304 cells after treatment with azide and oligomycin suggesting that the dynamic regulation of respiration by nitric oxide represent a critical and unrecognized primary line of defense against oxidative stress.


Journal of Biological Chemistry | 2009

Nitration of Solvent-exposed Tyrosine 74 on Cytochrome c Triggers Heme Iron-Methionine 80 Bond Disruption NUCLEAR MAGNETIC RESONANCE AND OPTICAL SPECTROSCOPY STUDIES

Luciano A. Abriata; Adriana Cassina; Verónica Tórtora; Mónica Marín; José M. Souza; Laura Castro; Alejandro J. Vila; Rafael Radi

Cytochrome c, a mitochondrial electron transfer protein containing a hexacoordinated heme, is involved in other physiologically relevant events, such as the triggering of apoptosis, and the activation of a peroxidatic activity. The latter occurs secondary to interactions with cardiolipin and/or post-translational modifications, including tyrosine nitration by peroxynitrite and other nitric oxide-derived oxidants. The gain of peroxidatic activity in nitrated cytochrome c has been related to a heme site transition in the physiological pH region, which normally occurs at alkaline pH in the native protein. Herein, we report a spectroscopic characterization of two nitrated variants of horse heart cytochrome c by using optical spectroscopy studies and NMR. Highly pure nitrated cytochrome c species modified at solvent-exposed Tyr-74 or Tyr-97 were generated after treatment with a flux of peroxynitrite, separated, purified by preparative high pressure liquid chromatography, and characterized by mass spectrometry-based peptide mapping. It is shown that nitration of Tyr-74 elicits an early alkaline transition with a pKa = 7.2, resulting in the displacement of the sixth and axial iron ligand Met-80 and replacement by a weaker Lys ligand to yield an alternative low spin conformation. Based on the study of site-specific Tyr to Phe mutants in the four conserved Tyr residues, we also show that this transition is not due to deprotonation of nitro-Tyr-74, but instead we propose a destabilizing steric effect of the nitro group in the mobile Ω-loop of cytochrome c, which is transmitted to the iron center via the nearby Tyr-67. The key role of Tyr-67 in promoting the transition through interactions with Met-80 was further substantiated in the Y67F mutant. These results therefore provide new insights into how a remote post-translational modification in cytochrome c such as tyrosine nitration triggers profound structural changes in the heme ligation and microenvironment and impacts in protein function.


Free Radical Biology and Medicine | 2014

Neuroprotective effects of the mitochondria-targeted antioxidant MitoQ in a model of inherited amyotrophic lateral sclerosis

Ernesto Miquel; Adriana Cassina; Laura Martínez-Palma; José M. Souza; Carmen Bolatto; Sebastián Rodríguez-Bottero; Angela Logan; Robin A. J. Smith; Michael P. Murphy; Luis Barbeito; Rafael Radi; Patricia Cassina

Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disorder characterized by motor neuron degeneration that ultimately results in progressive paralysis and death. Growing evidence indicates that mitochondrial dysfunction and oxidative stress contribute to motor neuron degeneration in ALS. To further explore the hypothesis that mitochondrial dysfunction and nitroxidative stress contribute to disease pathogenesis at the in vivo level, we assessed whether the mitochondria-targeted antioxidant [10-(4,5-dimethoxy-2-methyl-3,6-dioxo-1,4-cyclohexadien-1-yl)decyl]triphenylphosphonium methane sulfonate (MitoQ) can modify disease progression in the SOD1(G93A) mouse model of ALS. To do this, we administered MitoQ (500 µM) in the drinking water of SOD1(G93A) mice from a time when early symptoms of neurodegeneration become evident at 90 days of age until death. This regime is a clinically plausible scenario and could be more easily translated to patients as this corresponds to initiating treatment of patients after they are first diagnosed with ALS. MitoQ was detected in all tested tissues by liquid chromatography/mass spectrometry after 20 days of administration. MitoQ treatment slowed the decline of mitochondrial function, in both the spinal cord and the quadriceps muscle, as measured by high-resolution respirometry. Importantly, nitroxidative markers and pathological signs in the spinal cord of MitoQ-treated animals were markedly reduced and neuromuscular junctions were recovered associated with a significant increase in hindlimb strength. Finally, MitoQ treatment significantly prolonged the life span of SOD1(G93A) mice. Our results support a role for mitochondrial nitroxidative damage and dysfunction in the pathogenesis of ALS and suggest that mitochondria-targeted antioxidants may be of pharmacological use for ALS treatment.


Archives of Biochemistry and Biophysics | 2013

Oxidation and nitration of α-synuclein and their implications in neurodegenerative diseases

Cecilia Chavarría; José M. Souza

Synucleinopathies include Parkinsons disease, dementia with Lewy bodies, Lewy body variant of Alzheimers disease and multiple system atrophy, among the most relevant diseases. All of these diseases are characterized by the presence of amyloid inclusions in neurons, which are rich in the aggregate α-synuclein protein. What is the biological mechanism concerned in the gain-of-function that implicates the participation of α-synuclein in these diseases? Post-translational modifications of α-synuclein induced by nitroxidative stress are a relevant hypothesis that may explain many of the experimental data. We will review the biophysical and biochemical properties of α-synuclein, methionine residues oxidation, nitration and oxidation of tyrosine residues in α-synuclein, and modifications of α-synuclein mediated by proteins and lipids under nitroxidative stress conditions. The biological consequences of these modifications are analyzed in terms of the properties of α-synuclein oligomerization and fibrillation, degradation of α-synuclein and the implications in the immunological response.


Methods in Enzymology | 2008

Nitrocytochrome c: synthesis, purification, and functional studies.

José M. Souza; Laura Castro; Adriana Cassina; Carlos Batthyany; Rafael Radi

Posttranslational protein tyrosine oxidation, to yield 3-nitrotyrosine, is a biologically relevant protein modification related with acute and chronic inflammation and degenerative processes. It is usually associated with a decrease or loss in protein function. However, in some proteins, tyrosine nitration results in an increase or gain in protein function. Nitration of cytochrome c by biological oxidants in vitro can be achieved via different mechanisms, which include reactions with peroxynitrite, nitrite plus hydrogen peroxide, and nitric oxide plus hydrogen peroxide, and result in a loss in its electron transport capacity and in a higher peroxidatic activity. This chapter describes the methodology for studying chemical and biological properties of nitrocytochrome c. In particular, we report methods to synthesize tyrosine-nitrated cytochrome c, purify cytochrome c mononitrated species, map the sites of tyrosine nitration, and investigate the functional consequences of nitrated cytochrome c on mitochondrial electron transport properties, peroxidatic activity, and apoptosome assembly.

Collaboration


Dive into the José M. Souza's collaboration.

Top Co-Authors

Avatar

Harry Ischiropoulos

Children's Hospital of Philadelphia

View shared research outputs
Top Co-Authors

Avatar

Rafael Radi

University of the Republic

View shared research outputs
Top Co-Authors

Avatar

Qiping Chen

Children's Hospital of Philadelphia

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Adriana Cassina

Children's Hospital of Philadelphia

View shared research outputs
Top Co-Authors

Avatar

Caryn Hertkorn

Children's Hospital of Philadelphia

View shared research outputs
Top Co-Authors

Avatar

Celia Quijano

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Laura Castro

Children's Hospital of Philadelphia

View shared research outputs
Top Co-Authors

Avatar

Ana Denicola

University of the Republic

View shared research outputs
Researchain Logo
Decentralizing Knowledge