Network


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

Hotspot


Dive into the research topics where Natàlia Reixach is active.

Publication


Featured researches published by Natàlia Reixach.


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

Transthyretin protects Alzheimer's mice from the behavioral and biochemical effects of Aβ toxicity

Joel N. Buxbaum; Zhengyi Ye; Natàlia Reixach; Linsey Friske; Coree L. Levy; Pritam Das; Todd E. Golde; Eliezer Masliah; Amanda R. Roberts; Tamas Bartfai

Cells that have evolved to produce large quantities of secreted proteins to serve the integrated functions of complex multicellular organisms are equipped to compensate for protein misfolding. Hepatocytes and plasma cells have well developed chaperone and proteasome systems to ensure that secreted proteins transit the cell efficiently. The number of neurodegenerative disorders associated with protein misfolding suggests that neurons are particularly sensitive to the pathogenic effects of aggregates of misfolded molecules because those systems are less well developed in this lineage. Aggregates of the amyloidogenic (Aβ1–42) peptide play a major role in the pathogenesis of Alzheimers disease (AD), although the precise mechanism is unclear. In genetic studies examining protein–protein interactions that could constitute native mechanisms of neuroprotection in vivo, overexpression of a WT human transthyretin (TTR) transgene was ameliorative in the APP23 transgenic murine model of human AD. Targeted silencing of the endogenous TTR gene accelerated the development of the neuropathologic phenotype. Intraneuronal TTR was seen in the brains of normal humans and mice and in AD patients and APP23 mice. The APP23 brains showed colocalization of extracellular TTR with Aβ in plaques. Using surface plasmon resonance we obtained in vitro evidence of direct protein–protein interaction between TTR and Aβ aggregates. These findings suggest that TTR is protective because of its capacity to bind toxic or pretoxic Aβ aggregates in both the intracellular and extracellular environment in a chaperone-like manner. The interaction may represent a unique normal host defense mechanism, enhancement of which could be therapeutically useful.


Cellular and Molecular Life Sciences | 2009

Transthyretin: the servant of many masters

Joel N. Buxbaum; Natàlia Reixach

Transthyretin (TTR) (formerly, thyroxine binding prealbumin) is an evolutionarily conserved serum and cerebrospinal fluid protein that transports holo-retinol-binding protein and thyroxine. Its serum concentration has been widely used to assess clinical nutritional status. It is also well known that wild-type transthyretin and approximately 100 different mutants give rise to a variety of forms of systemic amyloid deposition. It has been suspected and recently established that TTR can suppress the Alzheimer’s disease phenotype in transgenic animal models of cerebral Aβ deposition. Thus, while TTR is a systemic amyloid precursor, in the brain it seems to have an anti-amyloidogenic effect. TTR is found in other organs as a result of local synthesis or transport, suggesting that it may have other, as yet undiscovered, functions. It is possible that its capacity to bind many classes of compounds allows it to serve as an endogenous detoxifier of molecules with potential pathologic effects.


The Journal of Neuroscience | 2011

Neuronal Production of Transthyretin in Human and Murine Alzheimer’s Disease: is it Protective?

Xinyi Li; Eliezer Masliah; Natàlia Reixach; Joel N. Buxbaum

Transthyretin (TTR), a systemic amyloid precursor in the human TTR amyloidoses, interacts with β-amyloid (Aβ) in vitro, inhibits Aβ fibril formation, and suppresses the Alzheimers disease (AD) phenotype in APP23 mice bearing a human APP gene containing the Swedish autosomal dominant AD mutation. In the present study, we show that TTR is a neuronal product upregulated in AD. Immunohistochemical analysis reveals that, in contrast to brains from non-demented age-matched individuals and control mice, the majority of hippocampal neurons from human AD and all those from the APP23 mouse brains contain TTR. Quantitative PCR for TTR mRNA and Western blot analysis show that primary neurons from APP23 mice transcribe TTR mRNA, and the cells synthesize and secrete TTR protein. TTR mRNA abundance is greatly increased in cultured cortical and hippocampal embryonic neurons and cortical lysates from adult APP23 mice. Antibodies specific for TTR and Aβ pulled down TTR/Aβ complexes from cerebral cortical extracts of APP23 mice and some human AD patients but not from control brains. In complementary tissue culture experiments, recombinant human TTR suppressed the cytotoxicity of soluble Aβ aggregates added to mouse neurons and differentiated human SH-SY5Y neuroblastoma cells. The findings that production of Aβ, its precursor, or its related peptides induces neuronal TTR transcription and synthesis and the presence of Aβ/TTR complexes in vivo suggest that increased TTR production coupled with interaction between TTR and Aβ and/or its related peptides may play a role in natural resistance to human AD.


Proteins | 2005

Poly‐(L‐alanine) expansions form core β‐sheets that nucleate amyloid assembly

Leonid M. Shinchuk; Deepak Sharma; Sylvie E. Blondelle; Natàlia Reixach; Hideyo Inouye; Daniel A. Kirschner

Expansion to a total of 11–17 sequential alanine residues from the normal number of 10 in the polyadenine‐binding protein nuclear‐1 (PABPN1) results in formation of intranuclear, fibrillar inclusions in skeletal muscle and hypothalamic neurons in adult‐onset, dominantly inherited oculopharyngeal muscular dystrophy (OPMD). To understand the role that homopolymeric length may play in the protein misfolding that leads to the inclusions, we analyzed the self‐assembly of synthetic poly‐(L‐alanine) peptides having 3–20 residues. We found that the conformational transition and structure of polyalanine (polyAla) assemblies in solution are not only length‐dependent but also are determined by concentration, temperature, and incubation time. No β‐sheet complex was detected for those peptides characterized by n < 8, where n is number of alanine residues. A second group of peptides with 7 < n < 15 showed varying levels of complex formation, while for those peptides having n > 15, the interconversion process from the monomeric to the β‐sheet complex was complete under any of the tested experimental conditions. Unlike the typical tinctorial properties of amyloid fibrils, polyalanine fibrils did not show fluorescence with thioflavin T or apple‐green birefringence with Congo red; however, like amyloid, X‐ray diffraction showed that the peptide chains in these fibrils were oriented normal to the fibril axis (i.e., in the cross‐β arrangement). Neighboring β‐sheets are quarter‐staggered in the hydrogen‐bonding direction such that the alanine side‐chains were closely packed in the intersheet space. Strong van der Waals contacts between side‐chains in this arrangement likely account for the high stability of the macromolecular fibrillar complex in solution over a wide range of temperature (5–85°C), and pH (2–10.5), and its resistance to denaturant (< 8 M urea) and to proteases (protease K, trypsin). We postulate that a similar stabilization of an expanded polyalanine stretch could form a core β‐sheet structure that mediates the intermolecular association of mutant proteins into fibrillar inclusions in human pathologies. Proteins 2005.


Biochemical and Biophysical Research Communications | 2011

Mechanisms of transthyretin cardiomyocyte toxicity inhibition by resveratrol analogs

Steve Bourgault; Sungwook Choi; Joel N. Buxbaum; Jeffery W. Kelly; Joshua L. Price; Natàlia Reixach

The transthyretin amyloidoses are a subset of protein misfolding diseases characterized by the extracellular deposition of aggregates derived from the plasma homotetrameric protein transthyretin (TTR) in peripheral nerves and the heart. We have established a robust disease-relevant human cardiac tissue culture system to explore the cytotoxic effects of amyloidogenic TTR variants. We have employed this cardiac amyloidosis tissue culture model to screen 23 resveratrol analogs as inhibitors of amyloidogenic TTR-induced cytotoxicity and to investigate their mechanisms of protection. Resveratrol and its analogs kinetically stabilize the native tetramer preventing the formation of cytotoxic species. In addition, we demonstrate that resveratrol can accelerate the formation of soluble non-toxic aggregates and that the resveratrol analogs tested can bring together monomeric TTR subunits to form non-toxic native tetrameric TTR.


Science Translational Medicine | 2011

Potent Kinetic Stabilizers that Prevent Transthyretin-mediated Cardiomyocyte Proteotoxicity

Mamoun Alhamadsheh; Stephen Connelly; Ahryon Cho; Natàlia Reixach; Evan T. Powers; Dorothy W. Pan; Ian A. Wilson; Jeffery W. Kelly; Isabella A. Graef

A high-throughput screen for transthyretin ligands that inhibit protein aggregation identifies agents that prevent amyloid formation and toxicity toward cardiomyocytes. Thwarting Amyloid by Encouraging Good Behavior Many proteins can self-assemble into amyloid, protein aggregates that show pronounced β sheet structure, and some of these aggregates accumulate in older people and people with various diseases. Although the β-amyloid of Alzheimer’s disease is the best-known disease-related amyloid, a circulating protein called transthyretin also forms amyloid. When aggregates of transthyretin, which normally carries thyroxine and retinol in the blood, are present in the heart, a serious cardiomyopathy ensues. Stabilization of the normal, tetrameric form of transthyretin prevents dissociation, the first step in amyloid formation. Several drugs that stabilize the tetramer are being tested in clinical trials, but because these drugs resemble nonsteroidal anti-inflammatory agents, they also inhibit the cyclooxygenase (COX) enzymes, causing gastrointestinal distress or cardiovascular problems. New drugs without these side effects are needed to fill this pipeline, and Alhamadsheh and colleagues now present a number of promising candidates discovered through a high-throughput, fluorescence-based screen. With their assay, the authors could detect, by virtue of a change in the tumbling rate, the binding of a fluorescence-tagged ligand to a site on transthyretin known to control the dissociation of the native tetramer and therefore amyloidogenesis. About 130,000 small-molecule drug candidates were rapidly applied in this system, and their ability to displace the ligand was easily assessed. The top 33 candidates were further validated in an independent surface plasmon resonance assay. Of these, four were able to effectively slow fibril formation to a greater extent than diclofenac, a nonsteroidal anti-inflammatory agent that blocks transthyretin aggregation, and several showed little inhibition of COX-1 enzyme activity, suggesting that they would not have the undesirable side effects of standard nonsteroidal anti-inflammatory agents. The authors identified the structural features that made for good candidate drugs with x-ray crystallography, finding that a flexible ring arrangement with which the ligand could bind and bridge two adjacent subunits was key. But how do we know that these molecules will work in patients? A definitive answer to that question will require clinical trials, but the authors present some encouraging data. In human cardiomyocytes that are sensitive to transthyretin amyloid, resulting in a decrease of their metabolic activity, the top drug candidates were able to rescue the cells at clinically reasonable concentrations. The top candidates were also effective in the presence of blood proteins, a requirement for a useful drug. Although more work is required, the fruits of this high-throughput screen provide a treasure trove of drugs to enable progress toward successful treatment of the transthyretin amyloidoses, without intestinal or cardiovascular side effects. A valine-to-isoleucine mutation at position 122 of the serum protein transthyretin (TTR), found in 3 to 4% of African Americans, alters its stability, leading to amyloidogenesis and cardiomyopathy. In addition, 10 to 15% of individuals older than 65 years develop senile systemic amyloidosis and cardiac TTR deposits because of wild-type TTR amyloidogenesis. Although several drugs are in development, no approved therapies for TTR amyloid cardiomyopathy are yet available, so the identification of additional compounds that prevent amyloid-mediated cardiotoxicity is needed. To this aim, we developed a fluorescence polarization–based high-throughput screen and used it to identify several new chemical scaffolds that target TTR. These compounds were potent kinetic stabilizers of TTR and prevented TTR tetramer dissociation, partial unfolding, and aggregation of both wild type and the most common cardiomyopathy-associated TTR mutant, V122I-TTR. High-resolution co-crystal structures and characterization of the binding energetics revealed how these diverse structures bound to tetrameric TTR. These compounds effectively inhibited the proteotoxicity of V122I-TTR toward human cardiomyocytes. Several of these ligands stabilized TTR in human serum more effectively than diflunisal, which is a well-studied inhibitor of TTR aggregation, and may be promising leads for the treatment or prevention of TTR-mediated cardiomyopathy.


Nature Chemical Biology | 2010

Chemoselective small molecules that covalently modify one lysine in a non-enzyme protein in plasma

Sungwook Choi; Stephen Connelly; Natàlia Reixach; Ian A. Wilson; Jeffery W. Kelly

A small molecule that could bind selectively to and then react chemoselectively with a non-enzyme protein in a complex biological fluid, such as blood, could have numerous practical applications. Herein, we report a family of designed stilbenes that selectively and covalently modify the prominent plasma protein transthyretin in preference to more than 4,000 other human plasma proteins. They react chemoselectively with only one of eight lysine e-amino groups within transthyretin. The crystal structure confirms the expected binding orientation of the stilbene substructure and the anticipated conjugating amide bond. These covalent transthyretin kinetic stabilizers exhibit superior amyloid inhibition potency compared to their noncovalent counterparts, and they prevent cytotoxicity associated with amyloidogenesis. Though there are a few prodrugs that, upon metabolic activation, react with a cysteine residue inactivating a specific non-enzyme, we are unaware of designed small molecules that react with one lysine e-amine within a specific non-enzyme protein in a complex biological fluid.


Biochemistry | 2013

Age-Related Oxidative Modifications of Transthyretin Modulate Its Amyloidogenicity

Lei Zhao; Joel N. Buxbaum; Natàlia Reixach

The transthyretin amyloidoses are diseases of protein misfolding characterized by the extracellular deposition of fibrils and other aggregates of the homotetrameric protein transthyretin (TTR) in peripheral nerves, heart, and other tissues. Age is the major risk factor for the development of these diseases. We hypothesized that an age-associated increase in the level of protein oxidation could be involved in the onset of the senile forms of the TTR amyloidoses. To test this hypothesis, we have produced and characterized relevant age-related oxidative modifications of the wild type (WT) and the Val122Ile (V122I) TTR variant, both involved in cardiac TTR deposition in the elderly. Our studies show that methionine/cysteine-oxidized TTR and carbonylated TTR from either the WT or the V122I variant are thermodynamically less stable than their nonoxidized counterparts. Moreover, carbonylated WT and carbonylated V122I TTR have a stronger propensity to form aggregates and fibrils than WT and V122I TTR, respectively, at physiologically attainable pH values. It is well-known that TTR tetramer dissociation, the limiting step for aggregation and amyloid fibril formation, can be prevented by small molecules that bind the TTR tetramer interface. Here, we report that carbonylated WT TTR is less amenable to resveratrol-mediated tetramer stabilization than WT TTR. All the oxidized forms of TTR tested are cytotoxic to a human cardiomyocyte cell line known to be a target for cardiac-specific TTR variants. Overall, these studies demonstrate that age-related oxidative modifications of TTR can contribute to the onset of the senile forms of the TTR amyloidoses.


Proceedings of the National Academy of Sciences of the United States of America | 2013

AG10 inhibits amyloidogenesis and cellular toxicity of the familial amyloid cardiomyopathy-associated V122I transthyretin

Sravan C. Penchala; Stephen Connelly; Yu Wang; Miki Susanto Park; Lei Zhao; Aleksandra Baranczak; Irit Rappley; Hannes Vogel; Michaela Liedtke; Ronald M. Witteles; Evan T. Powers; Natàlia Reixach; William K. Chan; Ian A. Wilson; Jeffery W. Kelly; Isabella A. Graef; Mamoun Alhamadsheh

The misassembly of soluble proteins into toxic aggregates, including amyloid fibrils, underlies a large number of human degenerative diseases. Cardiac amyloidoses, which are most commonly caused by aggregation of Ig light chains or transthyretin (TTR) in the cardiac interstitium and conducting system, represent an important and often underdiagnosed cause of heart failure. Two types of TTR-associated amyloid cardiomyopathies are clinically important. The Val122Ile (V122I) mutation, which alters the kinetic stability of TTR and affects 3% to 4% of African American subjects, can lead to development of familial amyloid cardiomyopathy. In addition, aggregation of WT TTR in individuals older than age 65 y causes senile systemic amyloidosis. TTR-mediated amyloid cardiomyopathies are chronic and progressive conditions that lead to arrhythmias, biventricular heart failure, and death. As no Food and Drug Administration-approved drugs are currently available for treatment of these diseases, the development of therapeutic agents that prevent TTR-mediated cardiotoxicity is desired. Here, we report the development of AG10, a potent and selective kinetic stabilizer of TTR. AG10 prevents dissociation of V122I-TTR in serum samples obtained from patients with familial amyloid cardiomyopathy. In contrast to other TTR stabilizers currently in clinical trials, AG10 stabilizes V122I- and WT-TTR equally well and also exceeds their efficacy to stabilize WT and mutant TTR in whole serum. Crystallographic studies of AG10 bound to V122I-TTR give valuable insights into how AG10 achieves such effective kinetic stabilization of TTR, which will also aid in designing better TTR stabilizers. The oral bioavailability of AG10, combined with additional desirable drug-like features, makes it a very promising candidate to treat TTR amyloid cardiomyopathy.


Journal of Biological Chemistry | 2008

Human-Murine Transthyretin Heterotetramers Are Kinetically Stable and Non-amyloidogenic A LESSON IN THE GENERATION OF TRANSGENIC MODELS OF DISEASES INVOLVING OLIGOMERIC PROTEINS

Natàlia Reixach; Ted R. Foss; Eugenio Santelli; Jaime Pascual; Jeffery W. Kelly; Joel N. Buxbaum

The transthyretin amyloidoses appear to be caused by rate-limiting tetramer dissociation and partial monomer unfolding of the human serum protein transthyretin, resulting in aggregation and extracellular deposition of amorphous aggregates and amyloid fibrils. Mice transgenic for few copies of amyloid-prone human transthyretin variants, including the aggressive L55P mutant, failed to develop deposits. Silencing the murine transthyretin gene in the presence of the L55P human gene resulted in enhanced tissue deposition. To test the hypothesis that the murine protein interacted with human transthyretin, preventing the dissociation and partial unfolding required for amyloidogenesis, we produced recombinant murine transthyretin and human/murine transthyretin heterotetramers and compared their structures and biophysical properties to recombinant human transthyretin. We found no significant differences between the crystal structures of murine and human homotetramers. Murine transthyretin is not amyloidogenic because the native homotetramer is kinetically stable under physiologic conditions and cannot dissociate into partially unfolded monomers, the misfolding and aggregation precursor. Heterotetramers composed of murine and human subunits are also kinetically stable. These observations explain the lack of transthyretin deposition in transgenics carrying a low copy number of human transthyretin genes. The incorporation of mouse subunits into tetramers otherwise composed of human amyloid-prone transthyretin subunits imposes kinetic stability, preventing dissociation and subsequent amyloidogenesis.

Collaboration


Dive into the Natàlia Reixach's collaboration.

Top Co-Authors

Avatar

Joel N. Buxbaum

Scripps Research Institute

View shared research outputs
Top Co-Authors

Avatar

Jeffery W. Kelly

Scripps Research Institute

View shared research outputs
Top Co-Authors

Avatar

Sylvie E. Blondelle

Torrey Pines Institute for Molecular Studies

View shared research outputs
Top Co-Authors

Avatar

Martin Lotz

Scripps Research Institute

View shared research outputs
Top Co-Authors

Avatar

O. Alvarez-Garcia

Scripps Research Institute

View shared research outputs
Top Co-Authors

Avatar

Josefina Casas

Spanish National Research Council

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ian A. Wilson

Scripps Research Institute

View shared research outputs
Top Co-Authors

Avatar

Stephen Connelly

Scripps Research Institute

View shared research outputs
Top Co-Authors

Avatar

Tokio Matsuzaki

Scripps Research Institute

View shared research outputs
Researchain Logo
Decentralizing Knowledge