Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Chembiochem ; : e202400279, 2024 May 22.
Artículo en Inglés | MEDLINE | ID: mdl-38776258

RESUMEN

Bacteria use specialized proteins, like transcription factors, to rapidly control metal ion balance. CueR is a Gram-negative bacterial copper regulator. The structure of E. coli CueR complexed with Cu(I) and DNA was published, since then many studies have shed light on its function. However, P. aeruginosa CueR, which shows high sequence similarity to E. coli CueR, has been less studied. Here, we applied room-temperature electron paramagnetic resonance (EPR) measurements to explore changes in dynamics of P. aeruginosa CueR in dependency of copper concentrations and interaction with two different DNA promoter regions. We showed that P. aeruginosa CueR is less dynamic than the E. coli CueR protein and exhibits much higher sensitivity to DNA binding as compared to its E. coli CueR homologue. Moreover, a difference in dynamical behavior was observed when P. aeruginosa CueR binds to the copZ2 DNA promoter sequence compared to the mexPQ-opmE promoter sequence. Such dynamical differences may affect the expression levels of CopZ2 and MexPQ-OpmE proteins in P. aeruginosa. Overall, such comparative measurements of protein-DNA complexes derived from different bacterial systems reveal insights about how structural and dynamical differences between two highly homologous proteins lead to quite different DNA sequence-recognition and mechanistic properties.

2.
ACS Omega ; 8(42): 39886-39895, 2023 Oct 24.
Artículo en Inglés | MEDLINE | ID: mdl-37901548

RESUMEN

In pathogens, a unique class of metalloregulator proteins, called gene regulatory proteins, sense specific metal ions that initiate gene transcription of proteins that export metal ions from the cell, thereby preventing toxicity and cell death. CsoR is a metalloregulator protein found in various bacterial systems that "sense" Cu(I) ions with high affinity. Upon copper binding, CsoR dissociates from the DNA promoter region, resulting in initiation of gene transcription. Crystal structures of CsoR in the presence and absence of Cu(I) from various bacterial systems have been reported, suggesting either a dimeric or tetrameric structure of these helical proteins. However, structural information about the CsoR-DNA complex is missing. Here, we applied electron paramagnetic resonance (EPR) spectroscopy to follow the conformational and dynamical changes that Mycobacterium tuberculosis CsoR undergoes upon DNA binding in solution. We showed that the quaternary structure is predominantly dimeric in solution, and only minor conformational and dynamical changes occur in the DNA bound state. Also, labeling of the unresolved C- terminus revealed no significant change in dynamics upon DNA binding. These observations are unique, since for other bacterial copper metalloregulators, such as the MerR and CopY families, major conformational changes were observed upon DNA binding, indicating a different mode of action for this protein family.

3.
Chem Commun (Camb) ; 59(70): 10524-10527, 2023 Aug 29.
Artículo en Inglés | MEDLINE | ID: mdl-37563959

RESUMEN

EPR in-cell spin-labeling was applied to CueR in E. coli. The methodology employed a Cu(II)-NTA complexed with dHis. High resolved in-cell distance distributions were obtained revealing minor differences between in vitro and in-cell data. This methodology allows study of structural changes of any protein in-cell, independent of size or cellular system.


Asunto(s)
Escherichia coli , Proteínas , Marcadores de Spin , Escherichia coli/química , Espectroscopía de Resonancia por Spin del Electrón/métodos , Proteínas/química
4.
Biochemistry ; 62(3): 797-807, 2023 02 07.
Artículo en Inglés | MEDLINE | ID: mdl-36691693

RESUMEN

Metal transcription factors regulate metal concentrations in eukaryotic and prokaryotic cells. Copper is a metal ion that is being tightly regulated, owing to its dual nature. Whereas copper is an essential nutrient for bacteria, it is also toxic at high concentrations. CopY is a metal-sensitive transcription factor belonging to the copper-responsive repressor family found in Gram-positive bacteria. CopY represses transcription in the presence of Zn(II) ions and initiates transcription in the presence of Cu(I) ions. The complete crystal structure of CopY has not been reported yet, therefore most of the structural information on this protein is based on its similarity to the well-studied MecI protein. In this study, electron paramagnetic resonance (EPR) spectroscopy was used to characterize structural and local dynamical changes in Streptococcus pneumoniae CopY as a function of Zn(II), Cu(I), and DNA binding. We detected different conformations and changes in local dynamics when CopY bound Zn(II), as opposed to Cu(I) ions. Furthermore, we explored the effects of metal ions and DNA on CopY conformation. Our results revealed the sensitivity and selectivity of CopY towards metal ions and provide new insight into the structural mechanism of the CopY transcription factor.


Asunto(s)
Cobre , Metales , Espectroscopía de Resonancia por Spin del Electrón , Cobre/metabolismo , Factores de Transcripción , Iones
5.
Front Mol Biosci ; 9: 1011294, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36299299

RESUMEN

Copper ions play a crucial role in various cellular biological processes. However, these copper ions can also lead to toxicity when their concentration is not controlled by a sophisticated copper-trafficking system. Copper dys-homeostasis has been linked to a variety of diseases, including neurodegeneration and cancer. Therefore, manipulating Cu-trafficking to trigger selective cancer cell death may be a viable strategy with therapeutic benefit. By exploiting combined in silico and experimental strategies, we identified small peptides able to bind Atox1 and metal-binding domains 3-4 of ATP7B proteins. We found that these peptides reduced the proliferation of cancer cells owing to increased cellular copper ions concentration. These outcomes support the idea of harming copper trafficking as an opportunity for devising novel anti-cancer therapies.

6.
Protein Sci ; 31(5): e4309, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35481642

RESUMEN

Metalloregulators bind and respond to metal ions by regulating the transcription of metal homeostasis genes. Copper efflux regulator (CueR) is a copper-responsive metalloregulator that is found in numerous Gram-negative bacteria. Upon Cu(I) coordination, CueR initiates transcription by bending the bound DNA promoter regions facilitating interaction with RNA polymerase. The structure of Escherichia coli CueR in presence of DNA and metal ion has been reported using X-ray crystallography and cryo-EM, providing information about the mechanism of action. However, the specific role of copper in controlling this transcription mechanism remains elusive. Herein, we use room temperature electron paramagnetic resonance (EPR) experiments to follow allosterically driven dynamical changes in E. coli CueR induced by Cu(I) binding. We suggest that more than one Cu(I) ion binds per CueR monomer, leading to changes in site-specific dynamics at the Cu(I) binding domain and at the distant DNA binding site. Interestingly, Cu(I) binding leads to an increase in dynamics about 27 Å away at the DNA binding domain. These changes in the dynamics of the DNA binding domain are important for exact coordination with the DNA. Thus, Cu(I) binding is critical to initiate a series of conformational changes that regulate and initiate gene transcription. BROAD AUDIENCE STATEMENT: The dynamics of metal transcription factors as a function of metal and DNA binding are complex. In this study, we use EPR spectroscopy to measure dynamical changes of Escherichia coli CueR as a function of copper and DNA binding. We show that copper controls the activation of the transcription processes by initiation a series of dynamical changes over the protein.


Asunto(s)
Cobre , Factores de Transcripción , Cobre/metabolismo , ADN/metabolismo , Escherichia coli/genética , Escherichia coli/metabolismo , Regulación Bacteriana de la Expresión Génica , Metales/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Transcripción Genética
7.
Biophys J ; 121(7): 1194-1204, 2022 04 05.
Artículo en Inglés | MEDLINE | ID: mdl-35202609

RESUMEN

Abnormal cellular copper levels have been clearly implicated in genetic diseases, cancer, and neurodegeneration. Ctr1, a high-affinity copper transporter, is a homotrimeric integral membrane protein that provides the main route for cellular copper uptake. Together with a sophisticated copper transport system, Ctr1 regulates Cu(I) metabolism in eukaryotes. Despite its pivotal role in normal cell function, the molecular mechanism of copper uptake and transport via Ctr1 remains elusive. In this study, electron paramagnetic resonance (EPR), UV-visible spectroscopy, and all-atom simulations were employed to explore Cu(I) binding to full-length human Ctr1 (hCtr1), thereby elucidating how metal binding at multiple distinct sites affects the hCtr1 conformational dynamics. We demonstrate that each hCtr1 monomer binds up to five Cu(I) ions and that progressive Cu(I) binding triggers a marked structural rearrangement in the hCtr1 C-terminal region. The observed Cu(I)-induced conformational remodeling suggests that the C-terminal region may play a dual role, serving both as a channel gate and as a shuttle mediating the delivery of copper ions from the extracellular hCtr1 selectivity filter to intracellular metallochaperones. Our findings thus contribute to a more complete understanding of the mechanism of hCtr1-mediated Cu(I) uptake and provide a conceptual basis for developing mechanism-based therapeutics for treating pathological conditions linked to de-regulated copper metabolism.


Asunto(s)
Proteínas de Transporte de Catión , Proteínas Transportadoras de Cobre , Transportador de Cobre 1 , Cobre , Cobre/química , Cobre/metabolismo , Proteínas Transportadoras de Cobre/química , Proteínas Transportadoras de Cobre/metabolismo , Transportador de Cobre 1/química , Transportador de Cobre 1/metabolismo , Humanos , Iones/química , Iones/metabolismo
8.
ChemistryOpen ; 10(4): 486-492, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33908707

RESUMEN

The Cu(II)-diacetyl-bis (N4-methylthiosemicarbazone) complex (ATSM-Cu(II)) has been suggested as a promising positron emission tomography (PET) agent for hypoxia imaging. Several in-vivo studies have shown its potential to detect hypoxic tumors. However, its uptake mechanism and its specificity to various cancer cell lines have been less studied. Herein, we tested ATSM-Cu(II) toxicity, uptake, and reduction, using four different cell types: (1) mouse breast cancer cells (DA-3), (2) human embryonic kidney cells (HEK-293), (3) breast cancer cells (MCF-7), and (4) cervical cancer cells (Hela) under normoxic and hypoxic conditions. We showed that ATSM-Cu(II) is toxic to breast cancer cells under normoxic and hypoxic conditions; however, it is not toxic to normal HEK-293 non-cancer cells. We showed that the Cu(I) content in breast cancer cell after treatment with ATSM-Cu(II) under hypoxic conditions is higher than in normal cells, despite that the uptake of ATSM-Cu(II) is a bit higher in normal cells than in breast cancer cells. This study suggests that the redox potential of ATSM-Cu(II) is higher in breast cancer cells than in normal cells; thus, its toxicity to cancer cells is increased.


Asunto(s)
Hipoxia/metabolismo , Compuestos Organometálicos/metabolismo , Tiosemicarbazonas/metabolismo , Animales , Línea Celular Tumoral , Complejos de Coordinación , Radioisótopos de Cobre/química , Radioisótopos de Cobre/metabolismo , Transportador de Cobre 1/metabolismo , Células HEK293 , Humanos , Ratones , Compuestos Organometálicos/química , Compuestos Organometálicos/toxicidad , Oxidación-Reducción , Tiosemicarbazonas/química , Tiosemicarbazonas/toxicidad
9.
Int J Mol Sci ; 21(15)2020 Aug 02.
Artículo en Inglés | MEDLINE | ID: mdl-32748830

RESUMEN

Copper's essentiality and toxicity mean it requires a sophisticated regulation system for its acquisition, cellular distribution and excretion, which until now has remained elusive. Herein, we applied continuous wave (CW) and pulsed electron paramagnetic resonance (EPR) spectroscopy in solution to resolve the copper trafficking mechanism in humans, by considering the route travelled by Cu(I) from the metallochaperone Atox1 to the metal binding domains of ATP7B. Our study revealed that Cu(I) is most likely mediated by the binding of the Atox1 monomer to metal binding domain 1 (MBD1) and MBD4 of ATP7B in the final part of its extraction pathway, while the other MBDs mediate this interaction and participate in copper transfer between the various MBDs to the ATP7B membrane domain. This research also proposes that MBD1-3 and MBD4-6 act as two independent units.


Asunto(s)
Proteínas Transportadoras de Cobre/metabolismo , ATPasas Transportadoras de Cobre/metabolismo , Cobre/metabolismo , Espectroscopía de Resonancia por Spin del Electrón/métodos , Chaperonas Moleculares/metabolismo , Sitios de Unión , Cobre/química , Proteínas Transportadoras de Cobre/química , ATPasas Transportadoras de Cobre/química , Humanos , Modelos Moleculares , Chaperonas Moleculares/química , Unión Proteica , Dominios Proteicos
10.
J Phys Chem B ; 124(22): 4399-4411, 2020 06 04.
Artículo en Inglés | MEDLINE | ID: mdl-32396355

RESUMEN

Atox1 is a human copper metallochaperone that is responsible for transferring copper ions from the main human copper transporter, hCtr1, to ATP7A/B in the Golgi apparatus. Atox1 interacts with the Ctr1 C-terminal domain as a dimer, although it transfers the copper ions to ATP7A/B in a monomeric form. The copper binding site in the Atox1 dimer involves Cys12 and Cys15, while Lys60 was also suggested to play a role in the copper binding. We recently showed that Atox1 can adopt various conformational states, depending on the interacting protein. In the current study, we apply EPR experiments together with hybrid quantum mechanics-molecular mechanics molecular dynamics simulations using a recently developed semiempirical density functional theory approach, to better understand the effect of Atox1's conformational states on copper coordination. We propose that the flexibility of Atox1 occurs owing to protonation of one or more of the cysteine residues, and that Cys15 is an important residue for Atox1 dimerization, while Cys12 is a critical residue for Cu(I) binding. We also show that Lys60 electrostatically stabilizes the Cu(I)-Atox1 dimer.


Asunto(s)
Metalochaperonas , Chaperonas Moleculares , Sitios de Unión , Cobre/metabolismo , Proteínas Transportadoras de Cobre , Humanos , Metalochaperonas/metabolismo , Chaperonas Moleculares/metabolismo
11.
PLoS One ; 14(12): e0227070, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31887125

RESUMEN

Five out of six people receive at least one antibiotic prescription per year. However, the ever-expanding use of antibiotics in medicine, agriculture, and food production has accelerated the evolution of antibiotic-resistant bacteria, which, in turn, made the development of novel antibiotics based on new molecular targets a priority in medicinal chemistry. One way of possibly combatting resistant bacterial infections is by inhibiting the copper transporters in prokaryotic cells. Copper is a key element within all living cells, but it can be toxic in excess. Both eukaryotic and prokaryotic cells have developed distinct copper regulation systems to prevent its toxicity. Therefore, selectively targeting the prokaryotic copper regulation system might be an initial step in developing next-generation antibiotics. One such system is the Gram-negative bacterial CusCFBA efflux system. CusB is a key protein in this system and was previously reported to play an important role in opening the channel for efflux via significant structural changes upon copper binding while also controlling the assembly and disassembly process of the entire channel. In this study, we aimed to develop novel peptide copper channel blockers, designed by in silico calculations based on the structure of CusB. Using a combination of magnetic resonance spectroscopy and various biochemical methods, we found a lead peptide that promotes copper-induced cell toxicity. Targeting copper transport in bacteria has not yet been pursued as an antibiotic mechanism of action. Thus, our study lays the foundation for discovering novel antibiotics.


Asunto(s)
Antibacterianos/farmacología , Proteínas Transportadoras de Cobre/antagonistas & inhibidores , Cobre/toxicidad , Proteínas de Escherichia coli/antagonistas & inhibidores , Péptidos/farmacología , Antibacterianos/síntesis química , Cobre/metabolismo , Proteínas Transportadoras de Cobre/química , Proteínas Transportadoras de Cobre/metabolismo , Diseño de Fármacos , Escherichia coli/efectos de los fármacos , Escherichia coli/metabolismo , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/metabolismo , Espectroscopía de Resonancia Magnética , Proteínas de Transporte de Membrana/química , Proteínas de Transporte de Membrana/metabolismo , Pruebas de Sensibilidad Microbiana , Simulación del Acoplamiento Molecular , Péptidos/síntesis química
12.
PLoS One ; 14(8): e0219337, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31465444

RESUMEN

The dissemination of resistant pathogenic microbes has become one of the most challenging problems that modern medicine has faced. Developing novel drugs based on new molecular targets that previously were not targeted, is therefore the highest priority in antibiotics research. One approach that has been recently suggested is to inhibit copper transporters in prokaryotic systems. Copper is required for many biological pathways, but sometimes it can harm the cell. Pathogenic systems have a highly sophisticated copper-regulation network; therefore, a better understanding of how this network operates at the molecular level should assist in developing the next generation of antibiotics. The CusB protein is part of the CusCBA periplasmic Cu(I) efflux system in Gram-negative bacteria, and was recently reported to play a key role in the functioning of the whole CusCBA system, in which conformational changes as well as the assembly/disassembly process control the opening of the transporter. More knowledge of the underlying mechanism is needed to attain a full understanding of CusB functioning, which is associated with targeting specific and crucial residues in CusB. Here, we combine in-vitro structural measurements, which use EPR spectroscopy and UV-Vis measurements, with cell experiments to explore the role of the various methionine residues in CusB. We targeted two methionine residues (M227 and M241) that are essential for the proper functioning of CusB.


Asunto(s)
Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/metabolismo , Escherichia coli/metabolismo , Proteínas de Transporte de Membrana/química , Proteínas de Transporte de Membrana/metabolismo , Metionina/metabolismo , Supervivencia Celular/efectos de los fármacos , Cobre/metabolismo , Cobre/farmacología , Escherichia coli/citología , Escherichia coli/efectos de los fármacos , Escherichia coli/fisiología , Proteínas de Escherichia coli/genética , Proteínas de Transporte de Membrana/genética , Modelos Moleculares , Mutación , Dominios Proteicos , Estrés Fisiológico/efectos de los fármacos
13.
Int J Mol Sci ; 20(14)2019 Jul 14.
Artículo en Inglés | MEDLINE | ID: mdl-31337158

RESUMEN

Appropriate maintenance of Cu(I) homeostasis is an essential requirement for proper cell function because its misregulation induces the onset of major human diseases and mortality. For this reason, several research efforts have been devoted to dissecting the inner working mechanism of Cu(I)-binding proteins and transporters. A commonly adopted strategy relies on mutations of cysteine residues, for which Cu(I) has an exquisite complementarity, to serines. Nevertheless, in spite of the similarity between these two amino acids, the structural and functional impact of serine mutations on Cu(I)-binding biomolecules remains unclear. Here, we applied various biochemical and biophysical methods, together with all-atom simulations, to investigate the effect of these mutations on the stability, structure, and aggregation propensity of Cu(I)-binding proteins, as well as their interaction with specific partner proteins. Among Cu(I)-binding biomolecules, we focused on the eukaryotic Atox1-ATP7B system, and the prokaryotic CueR metalloregulator. Our results reveal that proteins containing cysteine-to-serine mutations can still bind Cu(I) ions; however, this alters their stability and aggregation propensity. These results contribute to deciphering the critical biological principles underlying the regulatory mechanism of the in-cell Cu(I) concentration, and provide a basis for interpreting future studies that will take advantage of cysteine-to-serine mutations in Cu(I)-binding systems.


Asunto(s)
Sustitución de Aminoácidos , ATPasas Transportadoras de Cobre/química , ATPasas Transportadoras de Cobre/metabolismo , Cisteína/genética , Proteínas Mutantes/química , Proteínas Mutantes/metabolismo , Mutación , Serina/genética , ATPasas Transportadoras de Cobre/genética , Humanos , Metalochaperonas/química , Metalochaperonas/genética , Metalochaperonas/metabolismo , Modelos Moleculares , Conformación Proteica , Análisis Espectral , Relación Estructura-Actividad
14.
Metallomics ; 11(7): 1288-1297, 2019 07 17.
Artículo en Inglés | MEDLINE | ID: mdl-31187846

RESUMEN

Copper's essentiality and toxicity require a meticulous mechanism for its acquisition, cellular distribution and excretion, which remains hitherto elusive. Herein, we jointly employed electron paramagnetic resonance spectroscopy and all-atom simulations to resolve the copper trafficking mechanism in humans considering the route travelled by Cu(i) from the metallochaperone Atox1 to the metal binding domains 3 and 4 of ATP7B. Our study shows that Cu(i) in the final part of its extraction pathway is most likely mediated by binding of Atox1 monomer to MBD4 of ATP7B. This interaction takes place through weak metal-stabilized protein-protein interactions.


Asunto(s)
Proteínas Transportadoras de Cobre/metabolismo , ATPasas Transportadoras de Cobre/metabolismo , Cobre/metabolismo , Chaperonas Moleculares/metabolismo , Sitios de Unión , Transporte Biológico , ATPasas Transportadoras de Cobre/química , Espectroscopía de Resonancia por Spin del Electrón , Humanos , Modelos Moleculares , Dominios Proteicos , Mapas de Interacción de Proteínas
15.
Angew Chem Int Ed Engl ; 58(10): 3053-3056, 2019 03 04.
Artículo en Inglés | MEDLINE | ID: mdl-30566257

RESUMEN

The interactions between proteins and their specific DNA sequences are the basis of many cellular processes. Hence, developing methods to build an atomic level picture of these interactions helps improve our understanding of key cellular mechanisms. CueR is an Escherichia coli copper-sensing transcription regulator. The inhibition of copper-sensing transcription regulators can kill pathogens, without harming the host. Several spectroscopic studies and crystallographic data have suggested that changes in the conformation of both the DNA and the protein control transcription. However, due to the inadequate resolution of these methods, the exact number of active conformations of CueR has not been determined. Resolving the structure of CueR in its active state is highly important for the development of specific inhibitors. Herein, the potential of double-histidine (dHis)-based CuII spin labeling for the identification of various conformational states of CueR during transcription is shown.


Asunto(s)
Proteínas de Escherichia coli/metabolismo , Escherichia coli/metabolismo , Transactivadores/metabolismo , Sitios de Unión , Cobre/metabolismo , ADN Bacteriano/metabolismo , Espectroscopía de Resonancia por Spin del Electrón , Escherichia coli/química , Proteínas de Escherichia coli/química , Modelos Moleculares , Unión Proteica , Conformación Proteica , Multimerización de Proteína , Transactivadores/química
16.
Structure ; 23(11): 1989-2000, 2015 Nov 03.
Artículo en Inglés | MEDLINE | ID: mdl-26365803

RESUMEN

srGAP proteins regulate cell migration and morphogenesis by shaping the structure and dynamics of the cytoskeleton and membranes. First discovered as intracellular effectors for the Robo1 axon-guidance receptor, srGAPs were later identified as interacting with several other nuclear and cytoplasmic proteins. In all these cases, the srGAP SH3 domain mediates protein-protein interactions by recognizing a short proline-rich segment on the cognate-binding partner. However, as interactions between the isolated SH3 domain and a selected set of ligands show weak affinity and low specificity, it is not clear how srGAPs are precisely recruited to their signaling sites. Here, we report a two-component molecular mechanism that regulates ligand binding to srGAP2 by on the one hand dramatically tightening their association and on the other, moderately autoinhibiting and restricting binding. Our results allow the design of point mutations for better probing of srGAP2 activities, and may facilitate the identification of new srGAP2 ligands.


Asunto(s)
Proteínas Activadoras de GTPasa/química , Simulación del Acoplamiento Molecular , Secuencia de Aminoácidos , Sitios de Unión , Proteínas Activadoras de GTPasa/metabolismo , Humanos , Ligandos , Datos de Secuencia Molecular , Dominios Proteicos Ricos en Prolina , Unión Proteica , Especificidad por Sustrato , Dominios Homologos src
17.
J Struct Biol ; 186(2): 283-91, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24607414

RESUMEN

Robo receptors play pivotal roles in neurodevelopment, and their deregulation is implicated in several neuropathological conditions and cancers. To date, the mechanism of Robo activation and regulation remains obscure. Here we present the crystal structure of the juxtamembrane (JM) domains of human Robo1. The structure exhibits unexpectedly high backbone similarity to the netrin and RGM binding region of neogenin and DCC, which are functionally related receptors of Robo1. Comparison of these structures reveals a conserved surface that overlaps with a cluster of oncogenic and neuropathological mutations found in all Robo isoforms. The structure also reveals the intricate folding of the JM linker, which points to its role in Robo1 activation. Further experiments with cultured cells demonstrate that exposure or relief of the folded JM linker results in enhanced shedding of the Robo1 ectodomain.


Asunto(s)
Modelos Moleculares , Proteínas del Tejido Nervioso/química , Proteínas del Tejido Nervioso/genética , Receptores Inmunológicos/química , Receptores Inmunológicos/genética , Secuencia de Aminoácidos , Cristalografía por Rayos X , Electroforesis en Gel de Poliacrilamida , Células HEK293 , Humanos , Datos de Secuencia Molecular , Proteínas del Tejido Nervioso/ultraestructura , Conformación Proteica , Isoformas de Proteínas/química , Isoformas de Proteínas/ultraestructura , Estructura Terciaria de Proteína , Receptores Inmunológicos/ultraestructura , Proteínas Roundabout
18.
Mol Biol Cell ; 20(5): 1400-7, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19144822

RESUMEN

Transport of essentially all matrix and a number of inner membrane proteins is governed, entirely or in part, by N-terminal presequences and requires a coordinated action of the translocases of outer and inner mitochondrial membranes (TOM and TIM23 complexes). Here, we have analyzed Tim50, a subunit of the TIM23 complex that is implicated in transfer of precursors from TOM to TIM23. Tim50 is recruited to the TIM23 complex via Tim23 in an interaction that is essentially independent of the rest of the translocase. We find Tim50 in close proximity to the intermembrane space side of the TOM complex where it recognizes both types of TIM23 substrates, those that are to be transported into the matrix and those destined to the inner membrane, suggesting that Tim50 recognizes presequences. This function of Tim50 depends on its association with TIM23. We conclude that the efficient transfer of precursors between TOM and TIM23 complexes requires the concerted action of Tim50 with Tim23.


Asunto(s)
Proteínas de Transporte de Membrana Mitocondrial/fisiología , Membranas Mitocondriales/metabolismo , Precursores de Proteínas/metabolismo , Proteínas de Saccharomyces cerevisiae/fisiología , Proteínas de Transporte de Membrana/metabolismo , Proteínas de Transporte de Membrana Mitocondrial/química , Proteínas del Complejo de Importación de Proteínas Precursoras Mitocondriales , Estructura Terciaria de Proteína , Saccharomyces cerevisiae/citología , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/química , Proteínas de Saccharomyces cerevisiae/metabolismo
19.
J Biol Chem ; 284(8): 4865-72, 2009 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-19017642

RESUMEN

The TIM23 complex is the major translocase of the mitochondrial inner membrane responsible for the import of essentially all matrix proteins and a number of inner membrane proteins. Tim23 and Tim50, two essential proteins of the complex, expose conserved domains into the intermembrane space that interact with each other. Here, we describe in vitro reconstitution of this interaction using recombinantly expressed and purified intermembrane space domains of Tim50 and Tim23. We established two independent methods, chemical cross-linking and surface plasmon resonance, to track their interaction. In addition, we identified mutations in Tim23 that abolish its interaction with Tim50 in vitro. These mutations also destabilized the interaction between the two proteins in vivo, leading to defective import of preproteins via the TIM23 complex and to cell death at higher temperatures. This is the first study to describe the reconstitution of the Tim50-Tim23 interaction in vitro and to identify specific residues of Tim23 that are vital for the interaction with Tim50.


Asunto(s)
Proteínas de Transporte de Membrana/metabolismo , Mitocondrias/metabolismo , Proteínas de Transporte de Membrana Mitocondrial/metabolismo , Complejos Multiproteicos/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/metabolismo , Proteínas de Transporte de Membrana/química , Proteínas de Transporte de Membrana/genética , Mitocondrias/química , Mitocondrias/genética , Proteínas de Transporte de Membrana Mitocondrial/química , Proteínas de Transporte de Membrana Mitocondrial/genética , Membranas Mitocondriales/química , Membranas Mitocondriales/metabolismo , Proteínas del Complejo de Importación de Proteínas Precursoras Mitocondriales , Complejos Multiproteicos/química , Complejos Multiproteicos/genética , Mutación , Estructura Terciaria de Proteína/fisiología , Transporte de Proteínas/fisiología , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Saccharomyces cerevisiae/química , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/química , Proteínas de Saccharomyces cerevisiae/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...