Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
JBMR Plus ; 8(5): ziae034, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38586475

RESUMEN

Mutations in PLEKHM1 cause osteopetrosis in humans and rats. The germline and osteoclast conditional deletions of Plekhm1 gene in mice lead to defective osteoclast bone resorption and increased trabecular bone mass without overt abnormalities in other organs. As an adaptor protein, pleckstrin homology and RUN domain containing M1 (PLEKHM1) interacts with the key lysosome regulator small GTPase RAB7 via its C-terminal RUBICON homologous (RH) domain. In this study, we have conducted a structural-functional study of the PLEKHM1 RH domain and RAB7 interaction in osteoclasts in vitro. The single mutations of the key residues in the Plekhm1 RH predicted from the crystal structure of the RUBICON RH domain and RAB7 interface failed to disrupt the Plekhm1-Rab7 binding, lysosome trafficking, and bone resorption. The compound alanine mutations at Y949-R954 and L1011-I1018 regions decreased Plekhm1 protein stability and Rab7-binding, respectively, thereby attenuated lysosome trafficking and bone resorption in osteoclasts. In contrast, the compound alanine mutations at R1060-Q1068 region were dispensable for Rab7-binding and Plekhm1 function in osteoclasts. These results indicate that the regions spanning Y949-R954 and L1011-I1018 of Plekhm1 RH domain are functionally important for Plekhm1 in osteoclasts and offer the therapeutic targets for blocking bone resorption in treatment of osteoporosis and other metabolic bone diseases.

2.
Curr Opin Lipidol ; 34(4): 147-155, 2023 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-37171285

RESUMEN

PURPOSE OF REVIEW: LDL in its oxidized form, or 'oxLDL', is now generally acknowledged to be highly proatherogenic and to play a significant role in atherosclerotic plaque formation. Therefore, there has been increasing interest in understanding the significance of oxLDL and its receptors in different phases of atherosclerosis, leading to the accumulation of additional data at the cellular, structural, and physiological levels. This review focuses on the most recent discoveries about these receptors and how they influence lipid absorption, metabolism, and inflammation in various cell types. RECENT FINDINGS: Two crystal structures of lectin-like oxLDL receptor-1 (LOX-1), one with a small molecule inhibitor and the other with a monoclonal antibody have been published. We recently demonstrated that the 'surface site' of LOX1, adjacent to the positively charged 'basic spine region' that facilitates oxLDL binding, is a targetable site for drug development. Further, recent human studies showed that soluble LOX-1 holds potential as a biomarker for cardiovascular disease diagnosis, prognosis, and assessing the efficacy of therapy. SUMMARY: Receptor-mediated oxLDL uptake results in cellular dysfunction of various cell types involved in atherogenesis and plaque development. The current advancements clearly demonstrate that targeting oxLDL-LOX-1 axis may lead to development of future therapeutics for the treatment of atherosclerotic cardiovascular and cerebrovascular diseases.


Asunto(s)
Aterosclerosis , Placa Aterosclerótica , Humanos , Receptores de LDL Oxidadas , Receptores Depuradores de Clase E/metabolismo , Aterosclerosis/metabolismo , Lipoproteínas LDL/metabolismo , Inflamación , Receptores de LDL
3.
Sci Rep ; 13(1): 2096, 2023 02 06.
Artículo en Inglés | MEDLINE | ID: mdl-36747013

RESUMEN

Amyotrophic lateral sclerosis (ALS) is an inexorably progressive and degenerative disorder of motor neurons with no currently-known cure. Studies to determine the mechanism of neurotoxicity and the impact of ALS-linked mutations (SOD1, FUS, TARDP, C9ORF72, PFN1, TUBA4A and others) have greatly expanded our knowledge of ALS disease mechanisms and have helped to identify potential targets for ALS therapy. Cellular pathologies (e.g., aggregation of mutant forms of SOD1, TDP43, FUS, Ubiqulin2, PFN1, and C9ORF72), mitochondrial dysfunction, neuroinflammation, and oxidative damage are major pathways implicated in ALS. Nevertheless, the selective vulnerability of motor neurons remains unexplained. The importance of tubulins for long-axon infrastructure, and the special morphology and function of motor neurons, underscore the central role of the cytoskeleton. The recent linkage of mutations to the tubulin α chain, TUBA4A, to familial and sporadic cases of ALS provides a new investigative opportunity to shed light on both mechanisms of ALS and the vulnerability of motor neurons. In the current study we investigate TUBA4A, a structural microtubule protein with mutations causal to familial ALS, using molecular-dynamic (MD) modeling of protein structure to predict the effects of each mutation and its overall impact on GTP binding, chain stability, tubulin assembly, and aggregation propensity. These studies predict that each of the reported mutations will cause notable structural changes to the TUBA4A (α chain) tertiary protein structure, adversely affecting its physical properties and functions. Molecular docking and MD simulations indicate certain α chain mutations (e.g. K430N, R215C, and W407X) may cause structural deviations that impair GTP binding, and plausibly prevent or destabilize tubulin polymerization. Furthermore, several mutations (including R320C and K430N) confer a significant increase in predicted aggregation propensity of TUBA4A mutants relative to wild-type. Taken together, these in silico modeling studies predict structural perturbations and disruption of GTP binding, culminating in failure to form a stable tubulin heterocomplex, which may furnish an important pathogenic mechanism to trigger motor neuron degeneration in ALS.


Asunto(s)
Esclerosis Amiotrófica Lateral , Humanos , Esclerosis Amiotrófica Lateral/metabolismo , Tubulina (Proteína)/genética , Superóxido Dismutasa-1/genética , Simulación del Acoplamiento Molecular , Proteína C9orf72/genética , Mutación , Microtúbulos/metabolismo , Guanosina Trifosfato , Profilinas/genética
4.
Biochem Biophys Res Commun ; 623: 59-65, 2022 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-35872543

RESUMEN

Lectin-like oxidized low-density lipoprotein (ox-LDL) receptor 1 (LOX-1) is a vital scavenger receptor involved in ox-LDL binding, internalization, and subsequent proatherogenic signaling leading to cellular dysfunction and atherosclerotic plaque formation. Existing data suggest that modulation of ox-LDL - LOX-1 interaction can prevent or slow down atherosclerosis. Therefore, we utilized computational methods such as multi-solvent simulation and characterized two top-ranked druggable sites. Using systematic molecular docking followed by atomistic molecular dynamics simulation, we have identified and shortlisted small molecules from the NCI library that target two key binding sites. We demonstrate, using surface plasmon resonance (SPR), that four of the shortlisted molecules bind one-on-one to the purified C-terminal domain (CTLD) of LOX-1 receptor with high affinity (KD), ranging from 4.9 nM to 20.1 µM. Further, we performed WaterMap analysis to understand the role of individual water molecules in small molecule binding and the LOX-1-ligand complex stability. Our data clearly show that LOX-1 is druggable with small molecules. Our study provides strategies to identify novel inhibitors to attenuate ox-LDL - LOX-1 interaction.


Asunto(s)
Aterosclerosis , Lipoproteínas LDL , Aterosclerosis/tratamiento farmacológico , Aterosclerosis/metabolismo , Humanos , Lipoproteínas LDL/metabolismo , Simulación del Acoplamiento Molecular , Receptores Depuradores de Clase E/metabolismo
5.
Future Med Chem ; 14(10): 731-743, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35466695

RESUMEN

Along with other scavenger receptors, splice variants of LOX-1 play an important role in modulating numerous subcellular mechanisms such as normal cell development, differentiation and growth in response to physiological stimuli. Thus, LOX-1 activity is a key regulator in determining the severity of many genetic, metabolic, cardiovascular, renal, and neurodegenerative diseases and/or cancer. Increased expression of LOX-1 precipitates pathological disorders during the aging process. Therefore, it becomes important to develop novel LOX-1 inhibitors based on its ligand binding polarity and/or affinity and disrupt the uptake of its ligand: oxidized low-density lipoproteins (ox-LDL). In this review, we shed light on the presently studied and developed novel LOX-1 inhibitors that may have potential for treatment of diseases characterized by LOX-1 activation.


Asunto(s)
Receptores Depuradores de Clase E , Ligandos , Receptores Depuradores de Clase E/genética , Receptores Depuradores de Clase E/metabolismo
6.
Drug Dev Res ; 80(1): 11-18, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30312987

RESUMEN

Tuberculosis (TB) is a disease that has afflicted mankind for thousands of years, but in the last seven decades, much progress has been made in anti-TB therapy. Early drugs, such as para-aminosalicylic acid, streptomycin, isoniazid, and rifamycins were very effective in combatting the disease, giving rise to the hope that TB would be eradicated from the face of the earth by 2010. Despite that optimism, TB continues to kill more than a million people annually worldwide. A major reason for our inability to contain TB is the emergence drug resistance in Mycobacterium tuberculosis. This commentary is based on our recent publication on the structure of l,d-transpeptidase enzyme, relevant to drug resistance. As a background, we briefly outline the history and development of anti-TB therapy. Based on the crystal structure, we suggest a potential direction for designing more potent drugs against TB.


Asunto(s)
Antituberculosos/administración & dosificación , Sistemas de Liberación de Medicamentos/métodos , Farmacorresistencia Bacteriana/efectos de los fármacos , Mycobacterium tuberculosis/efectos de los fármacos , Peptidil Transferasas/antagonistas & inhibidores , Tuberculosis/tratamiento farmacológico , Animales , Pared Celular/efectos de los fármacos , Pared Celular/metabolismo , Sistemas de Liberación de Medicamentos/tendencias , Farmacorresistencia Bacteriana/fisiología , Humanos , Mycobacterium tuberculosis/enzimología , Peptidil Transferasas/metabolismo , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Tuberculosis/enzimología
7.
Sci Rep ; 8(1): 13102, 2018 08 30.
Artículo en Inglés | MEDLINE | ID: mdl-30166578

RESUMEN

Profilin-1 (PFN1) is a 140-amino-acid protein with two distinct binding sites-one for actin and one for poly-L-proline (PLP). The best-described function of PFN1 is to catalyze actin elongation and polymerization. Thus far, eight DNA mutations in the PFN1 gene encoding the PFN1 protein are associated with human amyotrophic lateral sclerosis (ALS). We and others recently showed that two of these mutations (Gly118Val or G118V and Cys71Gly or C71G) cause ALS in rodents. In vitro studies suggested that Met114Thr and Thr109Met cause the protein to behave abnormally and cause neurotoxicity. The mechanism by which a single amino acid change in human PFN1 causes the degeneration of motor neurons is not known. In this study, we investigated the structural perturbations of PFN1 caused by each ALS-associated mutation. We used molecular dynamics simulations to assess how these mutations alter the secondary and tertiary structures of human PFN1. Herein, we present our in silico data and analysis on the effect of G118V and T109M mutations on PFN1 and its interactions with actin and PLP. The substitution of valine for glycine reduces the conformational flexibility of the loop region between the α-helix and ß-strand and enhances the hydrophobicity of the region. Our in silico analysis of T109M indicates that this mutation alters the shape of the PLP-binding site and reduces the flexibility of this site. Simulation studies of PFN1 in its wild type (WT) and mutant forms (both G118V and T109M mutants) revealed differential fluctuation patterns and the formation of salt bridges and hydrogen bonds between critical residues that may shed light on differences between WT and mutant PFN1. In particular, we hypothesize that the flexibility of the actin- and PLP-binding sites in WT PFN1 may allow the protein to adopt slightly different conformations in its free and bound forms. These findings provide new insights into how each of these mutations in PFN1 might increase its propensity for misfolding and aggregation, leading to its dysfunction.


Asunto(s)
Esclerosis Amiotrófica Lateral/genética , Biología Computacional/métodos , Mutación/genética , Profilinas/química , Profilinas/genética , Agregado de Proteínas , Actinas/metabolismo , Sitios de Unión , Simulación por Computador , Humanos , Enlace de Hidrógeno , Interacciones Hidrofóbicas e Hidrofílicas , Proteínas Mutantes/química , Proteínas Mutantes/genética , Péptidos/metabolismo , Unión Proteica , Conformación Proteica
8.
AAPS J ; 20(2): 44, 2018 03 09.
Artículo en Inglés | MEDLINE | ID: mdl-29524047

RESUMEN

The final step of peptidoglycan (PG) synthesis in all bacteria is the formation of cross-linkage between PG-stems. The cross-linking between amino acids in different PG chains gives the peptidoglycan cell wall a 3-dimensional structure and adds strength and rigidity to it. There are two distinct types of cross-linkages in bacterial cell walls. D,D-transpeptidase (D,D-TPs) generate the classical 4➔3 cross-linkages and the L,D-transpeptidase (L,D-TPs) generate the 3➔3 non-classical peptide cross-linkages. The present study is aimed at understanding the nature of drug resistance associated with L,D-TP and gaining insights for designing novel antibiotics against multi-drug resistant bacteria. Penicillin and cephalosporin classes of ß-lactams cannot inhibit L,D-TP function; however, carbapenems inactivate its function. We analyzed the structure of L,D-TP of Mycobacterium tuberculosis in the apo form and in complex with meropenem and imipenem. The periplasmic region of L,D-TP folds into three domains. The catalytic residues are situated in the C-terminal domain. The acylation reaction occurs between carbapenem antibiotics and the catalytic Cys-354 forming a covalent complex. This adduct formation mimics the acylation of L,D-TP with the donor PG-stem. A novel aspect of this study is that in the crystal structures of the apo and the carbapenem complexes, the N-terminal domain has a muropeptide unit non-covalently bound to it. Another interesting observation is that the calcium complex crystallized as a dimer through head and tail interactions between the monomers.


Asunto(s)
Antibacterianos/farmacología , Proteínas Bacterianas/antagonistas & inhibidores , Farmacorresistencia Bacteriana Múltiple , Mycobacterium tuberculosis/fisiología , Peptidil Transferasas/antagonistas & inhibidores , Antibacterianos/química , Proteínas Bacterianas/química , Proteínas Bacterianas/aislamiento & purificación , Proteínas Bacterianas/metabolismo , Calcio/metabolismo , Dominio Catalítico/efectos de los fármacos , Cristalografía por Rayos X , Imipenem/química , Imipenem/farmacología , Meropenem/química , Meropenem/farmacología , Mycobacterium tuberculosis/efectos de los fármacos , Peptidoglicano/biosíntesis , Peptidil Transferasas/química , Peptidil Transferasas/aislamiento & purificación , Peptidil Transferasas/metabolismo , Unión Proteica , Multimerización de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/aislamiento & purificación , Proteínas Recombinantes/metabolismo , Relación Estructura-Actividad
9.
J Am Coll Cardiol ; 69(22): 2759-2768, 2017 Jun 06.
Artículo en Inglés | MEDLINE | ID: mdl-28571642

RESUMEN

Lectin-like oxidized low-density lipoprotein receptor-1 (LOX-1), one of the scavenger receptors for oxidized low-density lipoprotein cholesterol (ox-LDL), plays a crucial role in the uptake of ox-LDL by cells in the arterial wall. Mounting evidence suggests a role for LOX-1 in various steps of the atherosclerotic process, from initiation to plaque destabilization. Studies of the genetic structure of LOX-1 have also uncovered various genetic polymorphisms that could modulate the risk of atherosclerotic cardiovascular events. As evidence supporting the vital role of LOX-1 in atherogenesis keeps accumulating, there is growing interest in LOX-1 as a potential therapeutic target. This review discusses the discovery and genetics of LOX-1; describes existing evidence supporting the role of LOX-1 in atherogenesis and its major complication, myocardial ischemia; and summarizes LOX-1 modulation by some naturally occurring compounds and efforts toward development of small molecules and biologics that could be of therapeutic use.


Asunto(s)
Aterosclerosis/genética , Isquemia Miocárdica/genética , Polimorfismo Genético , Receptores Depuradores de Clase E/genética , Aterosclerosis/metabolismo , Humanos , Isquemia Miocárdica/metabolismo , Receptores Depuradores de Clase E/metabolismo
10.
Res Pract Thromb Haemost ; 1(2): 286-290, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-30046698

RESUMEN

BACKGROUND: Adaptive immunity in jawless fishes is performed by a unique set of proteins termed variable lymphocyte receptors (VLRs). Here we compare the crystallographic structures of VLRs and the human primary hemostasis receptor, glycoprotein (GP) Ib. It has been estimated jawless fish vertebrates diverged from jawed vertebrates 500 million years ago. Identifying structural similarities provides insights into the origins of primary hemostasis and the unique adaptive immunity of jawless fishes. METHODS: Three-dimensional structures obtained from crystallographic data and primary sequences alignments are compared. The results focus on overall domain arrangement to include the structural roles of leucine-rich repeats (LRRs), disulfide bond, and disulfide loop arrangements. RESULTS: The crystal structures of human GPIb (GPIbαN) and jawless fish VLRs are made up of three common segments each. The N-terminal cap and the C-terminal cap are characterized by disulfide bonds conserved in both GPIbαN and VLRs. The body of each molecule consists of LRRs which varies depending on the number of LRRs present in each molecule. The stacking of the LRRs results in the formation of a concave surface which serves as a motif to build ligand-binding specificity with the flanking regions. CONCLUSION: A comparison of VLR and GPIb structures reveals a phylogenetic trail of cellular differentiation contributing to mammalian hemostasis and jawless fish adaptive immunity. The results provide a structural basis to explain some of the interrelationships between hemostasis and immunity in vertebrates and potentially identifies a common ancestral motif linking hemostasis and immunity.

11.
Int J Biol Sci ; 12(12): 1488-1499, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27994513

RESUMEN

We have previously reported that depletion of LIS1, a key regulator of microtubules and cytoplasmic dynein motor complex, in osteoclast precursor cells by shRNAs attenuates osteoclastogenesis in vitro. However, the underlying mechanisms remain unclear. In this study, we show that conditional deletion of LIS1 in osteoclast progenitors in mice led to increased bone mass and decreased osteoclast number on trabecular bone. In vitro mechanistic studies revealed that loss of LIS1 had little effects on cell cycle progression but accelerated apoptosis of osteoclast precursor cells. Furthermore, deletion of LIS1 prevented prolonged activation of ERK by M-CSF and aberrantly enhanced prolonged JNK activation stimulated by RANKL. Finally, lack of LIS1 abrogated M-CSF and RANKL induced CDC42 activation and retroviral transduction of a constitutively active form of CDC42 partially rescued osteoclastogenesis in LIS1-deficient macrophages. Therefore, these data identify a key role of LIS1 in regulation of cell survival of osteoclast progenitors by modulating M-CSF and RANKL induced signaling pathways and CDC42 activation.


Asunto(s)
1-Alquil-2-acetilglicerofosfocolina Esterasa/metabolismo , Proteínas Asociadas a Microtúbulos/metabolismo , Osteoclastos/citología , Osteoclastos/metabolismo , Osteogénesis/fisiología , Ligando RANK/metabolismo , 1-Alquil-2-acetilglicerofosfocolina Esterasa/genética , Animales , Western Blotting , Células de la Médula Ósea/citología , Ciclo Celular/genética , Ciclo Celular/fisiología , Diferenciación Celular/genética , Diferenciación Celular/fisiología , Supervivencia Celular/genética , Supervivencia Celular/fisiología , Ensayo de Inmunoadsorción Enzimática , Femenino , Citometría de Flujo , Masculino , Ratones , Proteínas Asociadas a Microtúbulos/genética , Monocitos/citología , Monocitos/metabolismo , Osteogénesis/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Fosfatasa Ácida Tartratorresistente/metabolismo , Microtomografía por Rayos X
12.
JCI Insight ; 1(17): e86330, 2016 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-27777970

RESUMEN

Mutations of the Plekhm1 gene in humans and rats cause osteopetrosis, an inherited bone disease characterized by diminished bone resorption by osteoclasts. PLEKHM1 binds to RAB7 and is critical for lysosome trafficking. However, the molecular mechanisms by which PLEKHM1 regulates lysosomal pathways remain unknown. Here, we generated germline and conditional Plekhm1-deficient mice. These mice displayed no overt abnormalities in major organs, except for an increase in trabecular bone mass. Furthermore, loss of PLEKHM1 abrogated the peripheral distribution of lysosomes and bone resorption in osteoclasts. Mechanistically, we indicated that DEF8 interacts with PLEKHM1 and promotes its binding to RAB7, whereas the binding of FAM98A and NDEL1 with PLEKHM1 connects lysosomes to microtubules. Importantly, suppression of these proteins results in lysosome positioning and bone resorption defects similar to those of Plekhm1-null osteoclasts. Thus, PLHKEM1, DEF8, FAM98A, and NDEL1 constitute a molecular complex that regulates lysosome positioning and secretion through RAB7.


Asunto(s)
Resorción Ósea , Lisosomas/fisiología , Osteoclastos/fisiología , Proteínas de Transporte Vesicular/fisiología , Proteínas de Unión al GTP rab/fisiología , Proteínas Adaptadoras Transductoras de Señales , Animales , Proteínas Relacionadas con la Autofagia , Diferenciación Celular , Células Cultivadas , Endosomas , Eliminación de Gen , Homeostasis , Ratones , Ratones Endogámicos C57BL , Proteínas de Transporte Vesicular/genética , Proteínas de Unión a GTP rab7
14.
Sci Rep ; 5: 16740, 2015 Nov 18.
Artículo en Inglés | MEDLINE | ID: mdl-26578342

RESUMEN

Atherosclerosis related cardiovascular diseases continue to be the primary cause of mortality in developed countries. The elevated level of low density lipoprotein (LDL) is generally considered to be the driver of atherosclerosis, but recent years have seen a shift in this perception in that the vascular plaque buildup is mainly caused by oxidized LDL (ox-LDL) rather than native-LDL. The scavenger receptor LOX-1 found in endothelial cells binds and internalizes ox-LDL which leads to the initiation of plaque formation in arteries. Using virtual screening techniques, we identified a few potential small molecule inhibitors of LOX-1 and tested their inhibitory potential using differential scanning fluorimetry and various cellular assays. Two of these molecules significantly reduced the uptake of ox-LDL by human endothelial cells, LOX-1 transcription and the activation of ERK1/2 and p38 MAPKs in human endothelial cells. In addition, these molecules suppressed ox-LDL-induced VCAM-1 expression and monocyte adhesion onto human endothelial cells demonstrating their therapeutic potential.


Asunto(s)
Diseño de Fármacos , Ligandos , Lipoproteínas LDL/química , Receptores Depuradores de Clase E/química , Adhesión Celular/genética , Simulación por Computador , Descubrimiento de Drogas , Expresión Génica , Regulación de la Expresión Génica/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Modelos Moleculares , Conformación Molecular , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Estabilidad Proteica , Receptores Depuradores de Clase E/antagonistas & inhibidores , Receptores Depuradores de Clase E/genética , Bibliotecas de Moléculas Pequeñas , Relación Estructura-Actividad , Molécula 1 de Adhesión Celular Vascular/genética , Molécula 1 de Adhesión Celular Vascular/metabolismo
15.
J Proteomics Bioinform ; 8(11): 243-252, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26807012

RESUMEN

Defining protein-protein contacts is a challenging problem and cross-linking is a promising solution. Here, we present a case of mitochondrial single strand binding protein Rim1 and helicase Pif1, an interaction first observed in immuno-affinity pull-down from yeast cells using Pif1 bait. We found that only the short succinimidyl-diazirine cross-linker or formaldehyde captured the interaction between recombinant Rim1 and Pif1. In addition, Pif1 needed to be stripped of its N-terminal and C-terminal domains, and Rim1's C-terminus needed to be modified for the cross-linked product to become visible. Our report is an example of a non-trivial analysis, where a previously identified stable interaction escapes initial capture with cross-linking agents and requires substantial modification to recombinant proteins and fine-tuning of the mass spectrometry-based methods for the cross-links to become detectable. We used high resolution mass spectrometry to detect the cross-linked peptides. A 1:1 mixture of 15N and 14N-labeled Rim1 was used to validate the cross-links by their mass shift in the LC-MS profiles. Two sites on Rim1 were confirmed: 1) the N-terminus, and 2) the K29 residue. Performing cross-linking with a K29A variant visibly reduced the cross-linked product. Further, K29A-Rim1 showed a five-fold lower affinity to single stranded DNA compared to wild-type Rim1. Both the K29A variant and wild type Rim1 showed similar degrees of stimulation of Pif1 helicase activity. We propose structural models of the Pif1-Rim1 interaction and discuss its functional significance. Our work represents a non-trivial protein-protein interface analysis and demonstrates utility of short and non-specific cross-linkers.

16.
Drug Dev Res ; 75(1): 10-22, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24648045

RESUMEN

There is a pressing need to develop safe and effective radioprotector/radiomitigator agents for use in accidental or terrorist-initiated radiological emergencies. Naturally occurring vitamin E family constituents, termed tocols, that include the tocotrienols, are known to have radiation-protection properties. These agents, which work through multiple mechanisms, are promising radioprotectant agents having minimal toxicity. Although α-tocopherol (AT) is the most commonly studied form of vitamin E, the tocotrienols are more potent than AT in providing radioprotection and radiomitigation. Unfortunately, despite their very significant radioprotectant activity, tocotrienols have very short plasma half-lives and require dosing at very high levels to achieve necessary therapeutic benefits. Thus, it would be highly desirable to develop new vitamin E analogues with improved pharmacokinetic properties, specifically increased elimination half-life and increased area under the plasma level versus time curve. The short elimination half-life of the tocotrienols is related to their low affinity for the α-tocopherol transfer protein (ATTP), the protein responsible for maintaining the plasma level of the tocols. Tocotrienols have less affinity for ATTP than does AT, and thus have a longer residence time in the liver, putting them at higher risk for metabolism and biliary excretion. We hypothesized that the low-binding affinity of tocotrienols to ATTP is due to the relatively more rigid tail structure of the tocotrienols in comparison with that of the tocopherols. Therefore, compounds with a more flexible tail would have better binding to ATTP and consequently would have longer elimination half-life and, consequently, an increased exposure to drug, as measured by area under the plasma drug level versus time curve (AUC). This represents an enhanced residence of drug in the systemic circulation. Based on this hypothesis, we developed a new class of vitamin E analogues, the tocoflexols, which maintain the superior bioactivity of the tocotrienols with the potential to achieve the longer half-life and larger AUC of the tocopherols.


Asunto(s)
Proteínas Portadoras/metabolismo , Hígado/metabolismo , Protectores contra Radiación/farmacocinética , Tocotrienoles/farmacocinética , Vitamina E/análogos & derivados , Vitamina E/farmacocinética , Animales , Sitios de Unión , Disponibilidad Biológica , Diseño de Fármacos , Semivida , Humanos , Modelos Moleculares , Simulación de Dinámica Molecular , Ratas , Ratas Wistar
17.
Sci Rep ; 4: 3673, 2014 Jan 14.
Artículo en Inglés | MEDLINE | ID: mdl-24419156

RESUMEN

Methamphetamine (METH) abuse is a worldwide threat, without any FDA approved medications. Anti-METH IgGs and single chain fragments (scFvs) have shown efficacy in preclinical studies. Here we report affinity enhancement of an anti-METH scFv for METH and its active metabolite amphetamine (AMP), through the introduction of point mutations, rationally designed to optimize the shape and hydrophobicity of the antibody binding pocket. The binding affinity was measured using saturation binding technique. The mutant scFv-S93T showed 3.1 fold enhancement in affinity for METH and 26 fold for AMP. The scFv-I37M and scFv-Y34M mutants showed enhancement of 94, and 8 fold for AMP, respectively. Structural analysis of scFv-S93T:METH revealed that the substitution of Ser residue by Thr caused the expulsion of a water molecule from the cavity, creating a more hydrophobic environment for the binding that dramatically increases the affinities for METH and AMP.


Asunto(s)
Anfetamina/química , Anticuerpos Monoclonales/química , Afinidad de Anticuerpos , Metanfetamina/química , Secuencia de Aminoácidos , Anfetamina/antagonistas & inhibidores , Anticuerpos Monoclonales/genética , Anticuerpos Monoclonales/farmacología , Anticuerpos Monoclonales/uso terapéutico , Sitios de Unión , Interacciones Hidrofóbicas e Hidrofílicas , Metanfetamina/antagonistas & inhibidores , Modelos Moleculares , Conformación Molecular , Datos de Secuencia Molecular , Mutación , Unión Proteica , Ingeniería de Proteínas , Anticuerpos de Cadena Única/química , Anticuerpos de Cadena Única/genética , Anticuerpos de Cadena Única/farmacología , Anticuerpos de Cadena Única/uso terapéutico , Relación Estructura-Actividad
18.
PLoS One ; 8(12): e82690, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24349338

RESUMEN

Vaccines and monoclonal antibodies (mAb) for treatment of (+)-methamphetamine (METH) abuse are in late stage preclinical and early clinical trial phases, respectively. These immunotherapies work as pharmacokinetic antagonists, sequestering METH and its metabolites away from sites of action in the brain and reduce the rewarding and toxic effects of the drug. A key aspect of these immunotherapy strategies is the understanding of the subtle molecular interactions important for generating antibodies with high affinity and specificity for METH. We previously determined crystal structures of a high affinity anti-METH therapeutic single chain antibody fragment (scFv6H4, K(D) = 10 nM) in complex with METH and the (+) stereoisomer of 3,4-methylenedioxymethamphetamine (MDMA, or "ecstasy"). Here we report the crystal structure of scFv6H4 in homo-trimeric unbound (apo) form (2.60Å), as well as monomeric forms in complex with two active metabolites; (+)-amphetamine (AMP, 2.38Å) and (+)-4-hydroxy methamphetamine (p-OH-METH, 2.33Å). The apo structure forms a trimer in the crystal lattice and it results in the formation of an intermolecular composite beta-sheet with a three-fold symmetry. We were also able to structurally characterize the coordination of the His-tags with Ni(2+). Two of the histidine residues of each C-terminal His-tag interact with Ni(2+) in an octahedral geometry. In the apo state the CDR loops of scFv6H4 form an open conformation of the binding pocket. Upon ligand binding, the CDR loops adopt a closed formation, encasing the drug almost completely. The structural information reported here elucidates key molecular interactions important in anti-methamphetamine abuse immunotherapy.


Asunto(s)
Anticuerpos Monoclonales/química , Fragmentos de Inmunoglobulinas/química , Metanfetamina/química , Adenosina Monofosfato/química , Adenosina Monofosfato/metabolismo , Secuencia de Aminoácidos , Anticuerpos Monoclonales/farmacología , Cristalografía por Rayos X , Humanos , Metanfetamina/antagonistas & inhibidores , Modelos Moleculares , Datos de Secuencia Molecular , Estructura Molecular , Níquel/química , Níquel/metabolismo , Unión Proteica , Conformación Proteica , Multimerización de Proteína , Anticuerpos de Cadena Única/química , Anticuerpos de Cadena Única/farmacología , Sulfatos/química , Sulfatos/metabolismo
19.
Acta Crystallogr Sect E Struct Rep Online ; 69(Pt 11): o1709-10, 2013 Oct 26.
Artículo en Inglés | MEDLINE | ID: mdl-24454134

RESUMEN

The title compound, C21H25NO3 [systematic name: (3aS,9aR,10aR,10bS,E)-3-[(E)-4-(4-amino-benzyl-idene)-6,9a-dimethyl-3a,4,5,8,9,9a,10a,10b-octa-hydro-oxireno[2',3':9,10]cyclo-deca-[1,2-b]furan-2(3H)-one] was obtained from the reaction of parthenolide [synonym: 4,5-ep-oxy-germacra-1(10),11(13)-dieno-12,6-lactone] with 4-iodo-aniline under Heck reaction conditions. It was identified as the E-isomer (conformation about the exocyclic methyl-idene C=C bond; the conformation about the C=C bond in the ten-membered ring is also E). The mol-ecule is built up from fused ten-, five- (lactone) and three-membered (epoxide) rings with a 4-amino-phenyl group as a substituent. The ten-membered ring displays an approximate chair-chair conformation, while the lactone ring has an envelope conformation with the C atom bonded to the ring O atom as the flap. The dihedral angle between the benzene ring of the 4-amino-phenyl moiety and the lactone ring mean plane is 23.50 (8)°. In the crystal, mol-ecules are linked via N-H⋯O hydrogen bonds, between the amine group and the lactone and epoxide ring O atoms, forming chains propagating along the b-axis direction. Adjacent chains are linked via C-H⋯O inter-actions, forming an undulating two-dimensional network lying parallel to the plane (001). The absolute structure of the mol-ecule in the crystal was confirmed by resonance scattering [Flack parameter = 0.03 (3)].

20.
Acta Crystallogr D Biol Crystallogr ; 68(Pt 7): 839-45, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22751669

RESUMEN

In Bacillus subtilis, the WalRK (YycFG) two-component system coordinates murein synthesis with cell division. It regulates the expression of autolysins that function in cell-wall remodeling and of proteins that modulate autolysin activity. The transcription factor WalR is activated upon phosphorylation by the histidine kinase WalK, a multi-domain homodimer. It autophosphorylates one of its histidine residues by transferring the γ-phosphate from ATP bound to its ATP-binding domain. Here, the high-resolution crystal structure of the ATP-binding domain of WalK in complex with ATP is presented at 1.61 Šresolution. The bound ATP remains intact in the crystal lattice. It appears that the strong binding interactions and the nature of the binding pocket contribute to its stability. The triphosphate moiety of ATP wraps around an Mg(2+) ion, providing three O atoms for coordination in a near-ideal octahedral geometry. The ATP molecule also makes strong interactions with the protein. In addition, there is a short contact between the exocyclic O3' of the sugar ring and O2B of the ß-phosphate, implying an internal hydrogen bond. The stability of the WalK-ATP complex in the crystal lattice suggests that such a complex may exist in vivo poised for initiation of signal transmission. This feature may therefore be part of the sensing mechanism by which the WalRK two-component system is so rapidly activated when cells encounter conditions conducive for growth.


Asunto(s)
Adenosina Trifosfato/metabolismo , Bacillus subtilis/enzimología , Proteínas Quinasas/química , Proteínas Quinasas/metabolismo , Bacillus subtilis/química , Bacillus subtilis/metabolismo , Sitios de Unión , Cristalografía por Rayos X , Histidina Quinasa , Enlace de Hidrógeno , Magnesio/metabolismo , Modelos Moleculares , Estructura Terciaria de Proteína
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...