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










Base de dados
Intervalo de ano de publicação
1.
Cancer Res ; 77(2): 401-411, 2017 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-27872097

RESUMO

Neuroendocrine tumors (NET) often harbor loss-of-function mutations in the MEN1 and DAXX tumor suppressor genes. Here, we report that the products of these genes, menin and Daxx, interact directly with each other to suppress the proliferation of NET cells, to a large degree by inhibiting expression of the membrane metallo-endopeptidase (MME). Menin and Daxx were required to enhance histone H3 lysine9 trimethylation (H3K9me3) at the MME promoter, as mediated partly by the histone H3 methyltransferase SUV39H1. Notably, the menin T429K mutation associated with a NET syndrome reduced Daxx binding, MME repression, and proliferation of NET cells. Conversely, inhibition of MME in NET cells repressed proliferation and tumor growth in vivo Our findings reveal a previously unappreciated cross-talk between two crucial tumor suppressor genes thought to work by independent pathways, focusing on MME as a common target of menin/Daxx to treat NET. Cancer Res; 77(2); 401-11. ©2016 AACR.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/genética , Regulação Neoplásica da Expressão Gênica/genética , Neprilisina/genética , Tumores Neuroendócrinos/genética , Proteínas Nucleares/genética , Proteínas Proto-Oncogênicas/genética , Animais , Western Blotting , Proteínas Correpressoras , Epigênese Genética , Técnicas de Silenciamento de Genes , Xenoenxertos , Humanos , Imunoprecipitação , Camundongos , Camundongos Nus , Chaperonas Moleculares , Neprilisina/biossíntese , Tumores Neuroendócrinos/patologia , Análise de Sequência com Séries de Oligonucleotídeos , Reação em Cadeia da Polimerase em Tempo Real
2.
J Med Chem ; 58(1): 305-14, 2015 Jan 08.
Artigo em Inglês | MEDLINE | ID: mdl-25356520

RESUMO

Aberrant activation of S6 kinase 1 (S6K1) is found in many diseases, including diabetes, aging, and cancer. We developed ATP competitive organometallic kinase inhibitors, EM5 and FL772, which are inspired by the structure of the pan-kinase inhibitor staurosporine, to specifically inhibit S6K1 using a strategy previously used to target other kinases. Biochemical data demonstrate that EM5 and FL772 inhibit the kinase with IC50 value in the low nanomolar range at 100 µM ATP and that the more potent FL772 compound has a greater than 100-fold specificity over S6K2. The crystal structures of S6K1 bound to staurosporine, EM5, and FL772 reveal that the EM5 and FL772 inhibitors bind in the ATP binding pocket and make S6K1-specific contacts, resulting in changes to the p-loop, αC helix, and αD helix when compared to the staurosporine-bound structure. Cellular data reveal that FL772 is able to inhibit S6K phosphorylation in yeast cells. Together, these studies demonstrate that potent, selective, and cell permeable S6K1 inhibitors can be prepared and provide a scaffold for future development of S6K inhibitors with possible therapeutic applications.


Assuntos
Compostos Organometálicos/química , Inibidores de Proteínas Quinases/química , Proteínas Quinases S6 Ribossômicas 70-kDa/química , Trifosfato de Adenosina/química , Trifosfato de Adenosina/metabolismo , Sítios de Ligação , Western Blotting , Linhagem Celular Tumoral , Descoberta de Drogas , Células HEK293 , Humanos , Ligação de Hidrogênio , Modelos Moleculares , Conformação Molecular , Estrutura Molecular , Compostos Organometálicos/metabolismo , Compostos Organometálicos/farmacologia , Fosforilação/efeitos dos fármacos , Ligação Proteica , Inibidores de Proteínas Quinases/metabolismo , Inibidores de Proteínas Quinases/farmacologia , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Proteínas Quinases S6 Ribossômicas 70-kDa/antagonistas & inibidores , Proteínas Quinases S6 Ribossômicas 70-kDa/metabolismo , Rutênio/química
3.
J Virol ; 88(24): 14350-63, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25275136

RESUMO

UNLABELLED: Host chromatin assembly can function as a barrier to viral infection. Epstein-Barr virus (EBV) establishes latent infection as chromatin-assembled episomes in which all but a few viral genes are transcriptionally silent. The factors that control chromatin assembly and guide transcription regulation during the establishment of latency are not well understood. Here, we demonstrate that the EBV tegument protein BNRF1 binds the histone H3.3 chaperone Daxx to modulate histone mobility and chromatin assembly on the EBV genome during the early stages of primary infection. We demonstrate that BNRF1 substitutes for the repressive cochaperone ATRX to form a ternary complex of BNRF1-Daxx-H3.3-H4, using coimmunoprecipitation and size-exclusion chromatography with highly purified components. FRAP (fluorescence recovery after photobleaching) assays were used to demonstrate that BNRF1 promotes global mobilization of cellular histone H3.3. Mutation of putative nucleotide binding motifs on BNRF1 attenuates the displacement of ATRX from Daxx. We also show by immunofluorescence combined with fluorescence in situ hybridization that BNRF1 is important for the dissociation of ATRX and Daxx from nuclear bodies during de novo infection of primary B lymphocytes. Virion-delivered BNRF1 suppresses Daxx-ATRX-mediated H3.3 loading on viral chromatin as measured by chromatin immunoprecipitation assays and enhances viral gene expression during early infection. We propose that EBV tegument protein BNRF1 replaces ATRX to reprogram Daxx-mediated H3.3 loading, in turn generating chromatin suitable for latent gene expression. IMPORTANCE: Epstein-Barr Virus (EBV) is a human herpesvirus that efficiently establishes latent infection in primary B lymphocytes. Cellular chromatin assembly plays an important role in regulating the establishment of EBV latency. We show that the EBV tegument protein BNRF1 functions to regulate chromatin assembly on the viral genome during early infection. BNRF1 alters the host cellular chromatin assembly to prevent antiviral repressive chromatin and establish chromatin structure permissive for viral gene expression and the establishment of latent infection.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , DNA Helicases/antagonistas & inibidores , Herpesvirus Humano 4/fisiologia , Interações Hospedeiro-Patógeno , Proteínas Nucleares/antagonistas & inibidores , Proteínas Nucleares/metabolismo , Proteínas do Envelope Viral/metabolismo , Latência Viral , Linfócitos B/virologia , Sítios de Ligação , Células Cultivadas , Cromatografia em Gel , Proteínas Correpressoras , Recuperação de Fluorescência Após Fotodegradação , Imunofluorescência , Humanos , Imunoprecipitação , Hibridização in Situ Fluorescente , Chaperonas Moleculares , Mutagênese Sítio-Dirigida , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Ligação Proteica , Multimerização Proteica , Proteína Nuclear Ligada ao X
4.
Exp Cell Res ; 326(1): 136-42, 2014 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-24925478

RESUMO

Uric acid affects endothelial and adipose cell function and has been linked to diseases such as hypertension, metabolic syndrome, and cardiovascular disease. Interestingly uric acid has been shown to increase endothelial progenitor cell (EPC) mobilization, a potential mechanism to repair endothelial injury. Since EPC mobilization is dependent on activity of the enzyme CD26/dipeptidyl peptidase (DPP)IV, we examined the effect uric acid will have on CD26/DPPIV activity. Uric acid inhibited the CD26/DPPIV associated with human umbilical vein endothelial cells but not human recombinant (hr) CD26/DPPIV. However, triuret, a product of uric acid and peroxynitrite, could inhibit cell associated and hrCD26/DPPIV. Increasing or decreasing intracellular peroxynitrite levels enhanced or decreased the ability of uric acid to inhibit cell associated CD26/DPPIV, respectively. Finally, protein modeling demonstrates how triuret can act as a small molecule inhibitor of CD26/DPPIV activity. This is the first time that uric acid or a uric acid reaction product has been shown to affect enzymatic activity and suggests a novel avenue of research in the role of uric acid in the development of clinically important diseases.


Assuntos
Antioxidantes/farmacologia , Dipeptidil Peptidase 4/química , Dipeptidil Peptidase 4/metabolismo , Células Endoteliais da Veia Umbilical Humana/metabolismo , Proteínas Recombinantes/metabolismo , Ureia/análogos & derivados , Ácido Úrico/farmacologia , Inibidores Enzimáticos/farmacologia , Sequestradores de Radicais Livres/farmacologia , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Humanos , Ácido Hipocloroso/farmacologia , Metaloporfirinas/farmacologia , Modelos Moleculares , Oxidantes/farmacologia , Ureia/metabolismo
5.
PLoS Pathog ; 9(10): e1003672, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24146617

RESUMO

LANA is the KSHV-encoded terminal repeat binding protein essential for viral replication and episome maintenance during latency. We have determined the X-ray crystal structure of LANA C-terminal DNA binding domain (LANADBD) to reveal its capacity to form a decameric ring with an exterior DNA binding surface. The dimeric core is structurally similar to EBV EBNA1 with an N-terminal arm that regulates DNA binding and is required for replication function. The oligomeric interface between LANA dimers is dispensable for single site DNA binding, but is required for cooperative DNA binding, replication function, and episome maintenance. We also identify a basic patch opposite of the DNA binding surface that is responsible for the interaction with BRD proteins and contributes to episome maintenance function. The structural features of LANADBD suggest a novel mechanism of episome maintenance through DNA-binding induced oligomeric assembly.


Assuntos
Antígenos Virais/química , DNA Viral/química , Herpesvirus Humano 8/química , Proteínas Nucleares/química , Plasmídeos/química , Multimerização Proteica , Antígenos Virais/genética , Antígenos Virais/metabolismo , Linhagem Celular Tumoral , Replicação do DNA , DNA Viral/genética , DNA Viral/metabolismo , Herpesvirus Humano 8/genética , Herpesvirus Humano 8/metabolismo , Humanos , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Plasmídeos/genética , Plasmídeos/metabolismo , Ligação Proteica , Estrutura Quaternária de Proteína , Estrutura Terciária de Proteína , Relação Estrutura-Atividade
6.
J Virol ; 87(9): 4974-84, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23427155

RESUMO

The importance of the phospholipase A2 domain located within the unique N terminus of the capsid viral protein VP1 (VP1u) in parvovirus infection has been reported. This study used computational methods to characterize the VP1 sequence for adeno-associated virus (AAV) serotypes 1 to 12 and circular dichroism and electron microscopy to monitor conformational changes in the AAV1 capsid induced by temperature and the pHs encountered during trafficking through the endocytic pathway. Circular dichroism was also used to monitor conformational changes in AAV6 capsids assembled from VP2 and VP3 or VP1, VP2, and VP3 at pH 7.5. VP1u was predicted (computationally) and confirmed (in solution) to be structurally ordered. This VP domain was observed to undergo a reversible pH-induced unfolding/refolding process, a loss/gain of α-helical structure, which did not disrupt the capsid integrity and is likely facilitated by its difference in isoelectric point compared to the other VP sequences assembling the capsid. This study is the first to physically document conformational changes in the VP1u region that likely facilitate its externalization from the capsid interior during infection and establishes the order of events in the escape of the AAV capsid from the endosome en route to the nucleus.


Assuntos
Proteínas do Capsídeo/química , Proteínas do Capsídeo/metabolismo , Capsídeo/metabolismo , Dependovirus/fisiologia , Capsídeo/química , Proteínas do Capsídeo/genética , Dicroísmo Circular , Dependovirus/química , Dependovirus/genética , Microscopia Eletrônica , Modelos Moleculares , Estrutura Terciária de Proteína , Transporte Proteico , Montagem de Vírus
7.
Arch Biochem Biophys ; 529(1): 11-7, 2013 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-23111186

RESUMO

Although widely distributed in Nature, only two γ class carbonic anhydrases are reported besides the founding member (Cam). Although roles for active-site residues important for catalysis have been identified in Cam, second shell residues have not been investigated. Two residues (Trp19 and Tyr200), positioned distant from the catalytic metal, were investigated by structural and kinetic analyses of replacement variants. Steady-state k(cat)/K(m) and k(cat) values decreased 3- to 10-fold for the Trp19 variants whereas the Y200 variants showed up to a 5-fold increase in k(cat). Rate constants for proton transfer decreased up to 10-fold for the Trp19 variants, and an increase of ~2-fold for Y200F. The pK(a) values for the proton donor decreased 1-2 pH units for Trp19 and Y200 variants. The variant structures revealed a loop composed of residues 62-64 that occupies a different conformation than previously reported. The results show that, although Trp19 and Y200 are non-essential, they contribute to an extended active-site structure distant from the catalytic metal that fine tunes catalysis. Trp19 is important for both CO(2)/bicarbonate interconversion, and the proton transfer step of catalysis.


Assuntos
Proteínas Arqueais/química , Anidrases Carbônicas/química , Methanosarcina/enzimologia , Prótons , Triptofano/química , Tirosina/química , Proteínas Arqueais/genética , Proteínas Arqueais/metabolismo , Anidrases Carbônicas/genética , Anidrases Carbônicas/metabolismo , Catálise , Domínio Catalítico , Cristalografia por Raios X , Escherichia coli/genética , Concentração de Íons de Hidrogênio , Cinética , Methanosarcina/química , Modelos Moleculares , Mutação , Estrutura Secundária de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Triptofano/metabolismo , Tirosina/metabolismo
8.
J Virol ; 86(10): 5752-62, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22419807

RESUMO

LANA is essential for tethering the Kaposi's sarcoma-associated herpesvirus (KSHV) genome to metaphase chromosomes and for modulating host-cell gene expression, but the binding sites in the host-chromosome remain unknown. Here, we use LANA-specific chromatin immunoprecipitation coupled with high-throughput sequencing (ChIP-Seq) to identify LANA binding sites in the viral and host-cell genomes of a latently infected pleural effusion lymphoma cell line BCBL1. LANA bound with high occupancy to the KSHV genome terminal repeats (TR) and to a few minor binding sites in the KSHV genome, including the LANA promoter region. We identified 256 putative LANA binding site peaks with P < 0.01 and overlap in two independent ChIP-Seq experiments. We validated several of the high-occupancy binding sites by conventional ChIP assays and quantitative PCR. Candidate cellular LANA binding motifs were identified and assayed for binding to purified recombinant LANA protein in vitro but bound with low affinity compared to the viral TR binding site. More than half of the LANA binding sites (170/256) could be mapped to within 2.5 kb of a cellular gene transcript. Pathways and Gene Ontogeny (GO) analysis revealed that LANA binds to genes within the p53 and tumor necrosis factor (TNF) regulatory network. Further analysis revealed partial overlap of LANA and STAT1 binding sites in several gamma interferon (IFN-γ)-regulated genes. We show that ectopic expression of LANA can downmodulate IFN-γ-mediated activation of a subset of genes, including the TAP1 peptide transporter and proteasome subunit beta type 9 (PSMB9), both of which are required for class I antigen presentation. Our data provide a potential mechanism through which LANA may regulate several host cell pathways by direct binding to gene regulatory elements.


Assuntos
Antígenos Virais/metabolismo , Cromossomos Humanos/virologia , Herpesvirus Humano 8/metabolismo , Proteínas Nucleares/metabolismo , Sarcoma de Kaposi/genética , Sarcoma de Kaposi/virologia , Antígenos Virais/química , Antígenos Virais/genética , Sequência de Bases , Sítios de Ligação , Linhagem Celular , Cromossomos Humanos/química , Regulação da Expressão Gênica , Marcação de Genes , Herpesvirus Humano 8/química , Herpesvirus Humano 8/genética , Interações Hospedeiro-Patógeno , Humanos , Dados de Sequência Molecular , Proteínas Nucleares/química , Proteínas Nucleares/genética , Ligação Proteica
9.
Arch Biochem Biophys ; 516(2): 97-102, 2011 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-22001224

RESUMO

The tryptophan residue Trp5, highly conserved in the α class of carbonic anhydrases including human carbonic anhydrase II (HCA II), is positioned at the entrance of the active site cavity and forms a π-stacking interaction with the imidazole ring of the proton shuttle His64 in its outward orientation. We have observed that replacement of Trp5 in HCA II caused significant structural changes, as determined by X-ray diffraction, in the conformation of 11 residues at the N-terminus and in the orientation of the proton shuttle residue His64. Most significantly, two variants W5H and W5E HCA II had His64 predominantly outward in orientation, while W5F and wild type showed the superposition of both outward and inward orientations in crystal structures. Although Trp5 influences the orientation of the proton shuttle His64, this orientation had no significant effect on the rate constant for proton transfer near 1µs(-1), determined by exchange of (18)O between CO(2) and water measured by mass spectrometry. The apparent values of the pK(a) of the zinc-bound water and the proton shuttle residue suggest that different active-site conformations influence the two stages of catalysis, the proton transfer stage and the interconversion of CO(2) and bicarbonate.


Assuntos
Anidrase Carbônica II/química , Anidrase Carbônica II/metabolismo , Anidrase Carbônica II/genética , Catálise , Domínio Catalítico , Cristalografia por Raios X , Histidina/química , Humanos , Concentração de Íons de Hidrogênio , Cinética , Modelos Moleculares , Mutagênese Sítio-Dirigida , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Triptofano/química
10.
Acta Crystallogr D Biol Crystallogr ; 66(Pt 8): 950-2, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20693695

RESUMO

The crystal structure of human carbonic anhydrase II in the monoclinic P2(1) space group with a doubled a axis from that of the usually observed unit cell has recently been reported, with one of the two molecules in the asymmetric unit demonstrating rotational disorder [Robbins et al. (2010), Acta Cryst. D66, 628-634]. The structure has been redetermined, with the coordinates of both pseudo-symmetrically related molecules in the crystallographic asymmetric unit translated by x' = x +/- 1/4, and no rotational disorder is observed. This corresponds to a different choice of how the four molecules in the unit cell should be grouped into pairs that represent a single asymmetric unit.


Assuntos
Anidrase Carbônica II/química , Cristalografia por Raios X , Humanos
11.
Biochemistry ; 49(30): 6394-9, 2010 Aug 03.
Artigo em Inglês | MEDLINE | ID: mdl-20578724

RESUMO

The catalysis of CO(2) hydration by human carbonic anhydrase II (HCA II) is limited in maximal velocity by proton transfer from a zinc-bound water molecule to the proton shuttle His64. This proton transfer occurs along a hydrogen-bonded water network, leading to the proton shuttle residue His64, which in turn transfers the proton to bulk solvent. The side chain of His64 occupies two conformations in wild-type HCA II, pointing inward toward the zinc or outward toward bulk solvent. Previously, several studies have examined the roles of residues of the active site cavity that interact with the solvent-mediated hydrogen-bonded network between His64 and the zinc-bound water. Here these studies are extended to examine the effects on proton transfer by mutation at Lys170 (to Ala, Asp, Glu, and His), a residue located near the side chain of His64 but over 15 A away from the active site zinc. In all four variants, His64 is observed in the inward conformation associated with a decrease in the pK(a) of His64 by as much as 1.0 unit and an increase in the rate constant for proton transfer to as much as 4 micros(-1), approximately 5-fold larger than wild-type HCA II. The results show a significant extension of the effective active site of HCA II from the zinc-bound water at the base of the conical cavity in the enzyme to Lys170 near the rim of the cavity. These data emphasize that the active site of HCA II is extended to include residues that, at first glance, appear to be too far from the zinc to exert any catalytic effects.


Assuntos
Anidrase Carbônica II/química , Anidrase Carbônica II/metabolismo , Substituição de Aminoácidos , Anidrase Carbônica II/genética , Domínio Catalítico , Humanos , Ligação de Hidrogênio , Cinética , Conformação Proteica , Prótons , Água , Zinco
12.
Acta Crystallogr D Biol Crystallogr ; 66(Pt 5): 628-34, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20445238

RESUMO

The crystal structure of human carbonic anhydrase II with a doubled a axis from that of the usually observed monoclinic unit cell has been determined and refined to 1.4 A resolution. The diffraction data with h = 2n + 1 were systematically weaker than those with h = 2n. Consequently, the scaling of the data, structure solution and refinement were challenging. The two molecules comprising the asymmetric unit are related by a noncrystallographic translation of (1/2) along a, but one of the molecules has two alternate positions related by a rotation of approximately 2 degrees. This rotation axis is located near the edge of the central beta-sheet, causing a maximum distance disparity of 1.7 A between equivalent atoms on the diametrically opposite side of the molecule. The crystal-packing contacts are similar to two sequential combined unit cells along a of the previously determined monoclinic unit cell. Abnormally high final R(cryst) and R(free) values (20.2% and 23.7%, respectively) are not unusual for structures containing pseudo-translational symmetry and probably result from poor signal to noise in the weak h-odd data.


Assuntos
Anidrase Carbônica II/química , Cristalização , Cristalografia por Raios X , Humanos , Modelos Moleculares , Conformação Proteica
13.
Biochim Biophys Acta ; 1804(2): 326-31, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19679198

RESUMO

The interaction between carbon dioxide (CO(2)) and the alpha-class carbonic anhydrase, human CA 2 (HCA2) exists for only a short period due to the rapid catalytic turnover by this enzyme. The fleeting nature of this interaction has led to difficulties in its direct analysis, with previous studies placing the CO(2) in the hydrophobic pocket of HCA2's active site. A more precise location was determined via the crystal structure of CO(2) trapped in both wild-type (holo) and zinc-free (apo) HCA2. This provided a detailed description of the means by which CO(2) is held and orientated for optimal catalysis. This information can be extended to the beta and gamma class enzymes to help elucidate the binding mode of CO(2) in these enzymes.


Assuntos
Dióxido de Carbono/química , Anidrases Carbônicas/química , Animais , Dióxido de Carbono/metabolismo , Anidrases Carbônicas/metabolismo , Catálise , Humanos , Interações Hidrofóbicas e Hidrofílicas
14.
Biochemistry ; 49(3): 415-21, 2010 Jan 26.
Artigo em Inglês | MEDLINE | ID: mdl-20025241

RESUMO

Human carbonic anhydrase II (HCA II) catalyzes the reversible hydration of carbon dioxide to form bicarbonate and a proton. Despite many high-resolution X-ray crystal structures, mutagenesis, and kinetic data, the structural details of the active site, especially the proton transfer pathway, are unclear. A large HCA II crystal was prepared at pH 9.0 and subjected to vapor H-D exchange to replace labile hydrogens with deuteriums. Neutron diffraction studies were conducted at the Protein Crystallography Station at Los Alamos National Laboratory. The structure to 2.0 A resolution reveals several interesting active site features: (1) the Zn-bound solvent appearing to be predominantly a D(2)O molecule, (2) the orientation and hydrogen bonding pattern of solvent molecules in the active site cavity, (3) the side chain of His64 being unprotonated (neutral) and predominantly in an inward conformation pointing toward the zinc, and (4) the phenolic side chain of Tyr7 appearing to be unprotonated. The implications of these details are discussed, and a proposed mechanism for proton transfer is presented.


Assuntos
Anidrase Carbônica II/química , Anidrase Carbônica II/metabolismo , Nêutrons , Prótons , Sítios de Ligação , Dióxido de Carbono/química , Dióxido de Carbono/metabolismo , Catálise , Domínio Catalítico , Cristalografia por Raios X , Medição da Troca de Deutério , Histidina/genética , Humanos , Ligação de Hidrogênio , Modelos Moleculares , Estrutura Terciária de Proteína , Relação Estrutura-Atividade , Tirosina/genética , Difração de Raios X
15.
Artigo em Inglês | MEDLINE | ID: mdl-19851004

RESUMO

The crystal structure of human carbonic anhydrase II (CA II) complexed with the inhibitor acetazolamide (AZM) has been determined at 1.1 A resolution and refined to an R(cryst) of 11.2% and an R(free) of 14.7%. As observed in previous CA II-inhibitor complexes, AZM binds directly to the zinc and makes several key interactions with active-site residues. The high-resolution data also showed a glycerol molecule adjacent to the AZM in the active site and two additional AZMs that are adventitiously bound on the surface of the enzyme. The co-binding of AZM and glycerol in the active site demonstrate that given an appropriate ring orientation and substituents, an isozyme-specific CA inhibitor may be developed.


Assuntos
Acetazolamida/química , Anidrase Carbônica II/química , Inibidores da Anidrase Carbônica/química , Domínio Catalítico/efeitos dos fármacos , Desenho de Fármacos , Glicerol/química , Humanos , Modelos Moleculares
16.
Acta Crystallogr D Biol Crystallogr ; 64(Pt 11): 1172-8, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19020356

RESUMO

The crystal structure of the Mycoplasma hyorhinis protein Mh-p37 has been solved and refined to 1.9 A resolution. This is the first de novo structure to be determined using the recently described heavy-atom reagent [Beck et al. (2008), Acta Cryst. D64, 1179-1182] 5-amino-2,4,6-triiodoisophthalic acid (I3C), which contains three I atoms arranged in an equilateral triangle, by SIRAS methods. Data collection was performed in-house at room temperature. SHELXD and SHELXE were used to determine the I-atom positions and phase the native protein and PHENIX AutoBuild software was used to automatically fit the amino-acid sequence to the electron-density map. The structure was refined using SHELX97 to an R(cryst) of 18.6% and an R(free) of 24.0%. Mh-p37 is an alpha/beta protein with two well defined domains which are separated by a deep cleft. An unanticipated ligand bound in the center of the molecule at the base of the cleft has been modeled as thiamine pyrophosphate or vitamin B(1). Retrospective attempts to solve the crystal structure by Patterson search methods using either isomorphous or anomalous differences failed. Additionally, attempts to use proteins with the highest structural homology in the Protein Data Bank to phase the data by molecular replacement were unsuccessful, most likely in hindsight because of their poor structural agreement. Therefore, the I3C reagent offers an alternative, quick and inexpensive method for in-house phasing of de novo structures where other methods may not be successful.


Assuntos
Proteínas da Membrana Bacteriana Externa/química , Mycoplasma hyorhinis , Neoplasias/metabolismo , Proteínas Recombinantes/química , Animais , Proteínas da Membrana Bacteriana Externa/genética , Proteínas da Membrana Bacteriana Externa/metabolismo , Sítios de Ligação/genética , Clonagem Molecular , Cristalização , Cristalografia por Raios X/métodos , Humanos , Neoplasias/patologia , Ligação Proteica/genética , Estrutura Secundária de Proteína , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Deleção de Sequência , Homologia Estrutural de Proteína , Tiamina Pirofosfato/metabolismo , Transferrina/metabolismo , Ácidos Tri-Iodobenzoicos/metabolismo
17.
Infect Genet Evol ; 8(6): 825-34, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18804189

RESUMO

Genetic variation in viral structural proteins is often explained by evolutionary escape of strong host defenses through processes such as immune evasion, host switching, and tissue tropism. An understanding of the mechanisms driving evolutionary change in virus surface proteins is key to designing effective intervention strategies to disease emergence. This study investigated the predictability of virus genomic evolution in response to highly specific differences in host receptor structure. The bacteriophage PhiX174 was evolved on three E. coli mutant hosts, each differing only by a single sugar group in the lipopolysaccharides, used for phage attachment. Large phage populations were used in order to maximize the amount of sequence space explored by mutation, and thus the potential for parallel evolution. Repeatability was assessed by genome sequencing of multiple isolates from endpoint populations and by fitness of the endpoint population relative to its ancestor. Evolutionary lines showed similar magnitudes of fitness increase between treatments. Only one mutation, occurring in the internal DNA pilot protein H, was completely repeatable, and it appeared to be a necessary stepping stone toward further adaptive change. Substitutions in the surface accessible major capsid protein F appeared to be involved in capsid stability rather than specific interactions with host receptors, suggesting that non-specific alterations to capsid structure could be an important component of adaptation to novel hosts. 33% of mutations were synonymous and showed evidence of selection on codon usage. Lastly, results supported previous findings that evolving populations of small ssDNA viruses may maintain relatively high levels of genetic variation.


Assuntos
Bacteriófago phi X 174/crescimento & desenvolvimento , Bacteriófago phi X 174/genética , Evolução Molecular , Genoma Viral , Seleção Genética , Proteínas Virais/genética , Adaptação Biológica , Análise Mutacional de DNA , Escherichia coli/virologia , Genes Virais , Modelos Moleculares , Mutação , Conformação Proteica , Proteínas Virais/química
18.
J Biol Chem ; 283(45): 30766-71, 2008 Nov 07.
Artigo em Inglês | MEDLINE | ID: mdl-18768466

RESUMO

The visualization at near atomic resolution of transient substrates in the active site of enzymes is fundamental to fully understanding their mechanism of action. Here we show the application of using CO(2)-pressurized, cryo-cooled crystals to capture the first step of CO(2) hydration catalyzed by the zinc-metalloenzyme human carbonic anhydrase II, the binding of substrate CO(2), for both the holo and the apo (without zinc) enzyme to 1.1A resolution. Until now, the feasibility of such a study was thought to be technically too challenging because of the low solubility of CO(2) and the fast turnover to bicarbonate by the enzyme (Liang, J. Y., and Lipscomb, W. N. (1990) Proc. Natl. Acad. Sci. U. S. A. 87, 3675-3679). These structures provide insight into the long hypothesized binding of CO(2) in a hydrophobic pocket at the active site and demonstrate that the zinc does not play a critical role in the binding or orientation of CO(2). This method may also have a much broader implication for the study of other enzymes for which CO(2) is a substrate or product and for the capturing of transient substrates and revealing hydrophobic pockets in proteins.


Assuntos
Bicarbonatos/química , Dióxido de Carbono/química , Anidrase Carbônica II/química , Metaloproteínas/química , Zinco/química , Bicarbonatos/metabolismo , Dióxido de Carbono/metabolismo , Anidrase Carbônica II/metabolismo , Domínio Catalítico/fisiologia , Cristalografia por Raios X/métodos , Humanos , Interações Hidrofóbicas e Hidrofílicas , Metaloproteínas/metabolismo , Estrutura Terciária de Proteína/fisiologia , Zinco/metabolismo
19.
Biochemistry ; 47(16): 4621-8, 2008 Apr 22.
Artigo em Inglês | MEDLINE | ID: mdl-18373354

RESUMO

The function in the structure, stability, and catalysis of the interfaces between subunits in manganese superoxide dismutase (MnSOD) is currently under scrutiny. Glu162 in homotetrameric human MnSOD spans a dimeric interface and forms a hydrogen bond with His163 of an adjacent subunit which is a direct ligand of the manganese. We have examined the properties of two site-specific mutants of human MnSOD in which Glu162 is replaced with Asp (E162D) and Ala (E162A). The X-ray crystal structures of E162D and E162A MnSOD reveal no significant structural changes compared with the wild type other than the removal of the hydrogen bond interaction with His163 in E162A MnSOD. In the case of E162D MnSOD, an intervening solvent molecule fills the void created by the mutation to conserve the hydrogen bond interaction between His163 and residue 162. These mutants retain their tetrameric structure and their specificity for manganese over iron. Each has catalytic activity in the disproportionation of superoxide that is typically 5-25% of that of the wild-type enzyme and a level of product inhibition greater by approximately 2-fold. Differential scanning calorimetry indicates that the hydrogen bond between Glu162 and His163 contributes to the stability of MnSOD, with the major unfolding transition occurring at 81 degrees C for E162A compared to 90 degrees C for wild-type MnSOD. These results suggest that Glu162 at the tetrameric interface in human MnSOD supports stability and efficient catalysis and has a significant role in regulating product inhibition.


Assuntos
Superóxido Dismutase/química , Superóxido Dismutase/metabolismo , Catálise , Dimerização , Ácido Glutâmico/genética , Ácido Glutâmico/metabolismo , Humanos , Concentração de Íons de Hidrogênio , Modelos Moleculares , Mutação/genética , Desnaturação Proteica , Estrutura Quaternária de Proteína , Estrutura Terciária de Proteína , Superóxido Dismutase/genética , Temperatura , Difração de Raios X
20.
Biochemistry ; 46(51): 14830-7, 2007 Dec 25.
Artigo em Inglês | MEDLINE | ID: mdl-18044968

RESUMO

Human manganese superoxide dismutase (MnSOD) is characterized by a product inhibition stronger than that observed in bacterial forms of MnSOD. Previous studies show that the conserved, active-site residue Tyr34 mediates product inhibition; however, the protein environment of Tyr34 is different in human and Escherichia coli MnSOD. We have prepared two site-specific mutants of human MnSOD with replacements of Phe66 with Ala and Leu (F66A and F66L, respectively), altering the surroundings of Tyr34. Pulse radiolysis was used to generate superoxide, and measurements of catalysis were taken in single-turnover experiments by observing the visible absorbance of species of MnSOD and under catalytic conditions observing the absorbance of superoxide. The mutation of Phe66 to Leu resulted in a mutant of human MnSOD with weakened product inhibition resembling that of E. coli MnSOD. Moreover, the mechanism of this weakened product inhibition was similar to that in E. coli MnSOD, specifically a decrease in the rate constant for the oxidative addition of superoxide to Mn2+MnSOD leading to the formation of the peroxide-inhibited enzyme. In addition, the crystal structures of both mutants have been determined and compared to those of wild-type human and E. coli MnSOD. The crystallographic data suggest that the solvent structure and its mobility as well as side chain conformations may affect the extent of product inhibition. These data emphasize the role of residue 66 in catalysis and inhibition and provide a structural explanation for differences in catalytic properties between human and certain bacterial forms of MnSOD.


Assuntos
Escherichia coli/enzimologia , Superóxido Dismutase/química , Superóxido Dismutase/metabolismo , Sítios de Ligação , Catálise , Escherichia coli/genética , Humanos , Cinética , Modelos Moleculares , Mutação/genética , Estrutura Terciária de Proteína , Homologia Estrutural de Proteína , Superóxido Dismutase/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...