Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
2.
Commun Biol ; 4(1): 321, 2021 03 10.
Artigo em Inglês | MEDLINE | ID: mdl-33692454

RESUMO

An aberrant fusion of the DNAJB1 and PRKACA genes generates a chimeric protein kinase (PKA-CDNAJB1) in which the J-domain of the heat shock protein 40 is fused to the catalytic α subunit of cAMP-dependent protein kinase A (PKA-C). Deceivingly, this chimeric construct appears to be fully functional, as it phosphorylates canonical substrates, forms holoenzymes, responds to cAMP activation, and recognizes the endogenous inhibitor PKI. Nonetheless, PKA-CDNAJB1 has been recognized as the primary driver of fibrolamellar hepatocellular carcinoma and is implicated in other neoplasms for which the molecular mechanisms remain elusive. Here we determined the chimera's allosteric response to nucleotide and pseudo-substrate binding. We found that the fusion of the dynamic J-domain to PKA-C disrupts the internal allosteric network, causing dramatic attenuation of the nucleotide/PKI binding cooperativity. Our findings suggest that the reduced allosteric cooperativity exhibited by PKA-CDNAJB1 alters specific recognitions and interactions between substrates and regulatory partners contributing to dysregulation.


Assuntos
Trifosfato de Adenosina/metabolismo , Subunidades Catalíticas da Proteína Quinase Dependente de AMP Cíclico/metabolismo , Proteínas de Choque Térmico HSP40/metabolismo , Fragmentos de Peptídeos/metabolismo , Regulação Alostérica , Sítios de Ligação , Domínio Catalítico , AMP Cíclico/metabolismo , Subunidades Catalíticas da Proteína Quinase Dependente de AMP Cíclico/genética , Proteínas de Choque Térmico HSP40/genética , Humanos , Ligantes , Simulação de Dinâmica Molecular , Fragmentos de Peptídeos/genética , Fosforilação , Ligação Proteica , Proteínas Recombinantes de Fusão/metabolismo
3.
Elife ; 92020 04 27.
Artigo em Inglês | MEDLINE | ID: mdl-32338601

RESUMO

In the nucleus, the spatiotemporal regulation of the catalytic subunit of cAMP-dependent protein kinase A (PKA-C) is orchestrated by an intrinsically disordered protein kinase inhibitor, PKI, which recruits the CRM1/RanGTP nuclear exporting complex. How the PKA-C/PKI complex assembles and recognizes CRM1/RanGTP is not well understood. Using NMR, SAXS, fluorescence, metadynamics, and Markov model analysis, we determined the multi-state recognition pathway for PKI. After a fast binding step in which PKA-C selects PKI's most competent conformations, PKI folds upon binding through a slow conformational rearrangement within the enzyme's binding pocket. The high-affinity and pseudo-substrate regions of PKI become more structured and the transient interactions with the kinase augment the helical content of the nuclear export sequence, which is then poised to recruit the CRM1/RanGTP complex for nuclear translocation. The multistate binding mechanism featured by PKA-C/PKI complex represents a paradigm on how disordered, ancillary proteins (or protein domains) are able to operate multiple functions such as inhibiting the kinase while recruiting other regulatory proteins for nuclear export.


Assuntos
Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Inibidores de Proteínas Quinases/metabolismo , Transporte Ativo do Núcleo Celular , Animais , Proteínas Quinases Dependentes de AMP Cíclico/genética , Citoplasma , Escherichia coli , Peptídeos e Proteínas de Sinalização Intracelular/genética , Carioferinas/genética , Carioferinas/metabolismo , Espectroscopia de Ressonância Magnética , Cadeias de Markov , Camundongos , Coelhos , Receptores Citoplasmáticos e Nucleares/genética , Receptores Citoplasmáticos e Nucleares/metabolismo , Proteína Exportina 1
4.
Am J Pharm Educ ; 84(2): 7133, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-32226063

RESUMO

Objective. To evaluate the impact of a single, half-day interprofessional education (IPE) simulation on disclosing medical errors, and to compare the impacts on pharmacy students with those on students from other health professional programs. Methods. A mixed methods approach was used to provide a comprehensive understanding of the immediate and persistent outcomes of a realistic medical error disclosure simulation. Anonymous pre- and post-simulation quantitative data were collected using a validated attitudinal survey instrument administered at the time of the simulation. To assess more sustained impacts of the simulation, uni-professional focus groups were held several months following the simulation. Results. The survey analysis showed that a significant positive change in attitudes towards teamwork, roles and responsibilities occurred in students in most of the professions represented, with pharmacy students experiencing positive changes across a wider range of interprofessional attitudes. The focus group results showed that there were persistent impacts across all professions on learners' knowledge, skills, attitudes, and confidence in disclosing medical errors in interprofessional teams. Conclusion. Mixed methods analysis of a high-fidelity IPE error disclosure simulation demonstrated that single IPE activities, if realistic, can have significant positive impacts on students' interprofessional attitudes and competencies, and increase confidence in conducting team-based error disclosures.


Assuntos
Educação em Farmácia/métodos , Educação Interprofissional/métodos , Atitude do Pessoal de Saúde , Educação em Farmácia/organização & administração , Grupos Focais , Humanos , Relações Interprofissionais , Erros Médicos , Competência Profissional , Treinamento por Simulação , Estudantes de Farmácia , Inquéritos e Questionários , Revelação da Verdade
5.
J Mol Biol ; 428(24 Pt B): 4890-4904, 2016 12 04.
Artigo em Inglês | MEDLINE | ID: mdl-27825928

RESUMO

Most disease-related mutations that impair cAMP protein kinase A (PKA) signaling are present within the regulatory (R) PKA RI alpha-subunit (RIα). Although mutations in the PRKAR1A gene are linked to Carney complex (CNC) disease and, more recently, to acrodysostosis-1 (ACRDYS1), the two diseases show contrasting phenotypes. While CNC mutations cause increased PKA activity, ACRDYS1 mutations result in decreased PKA activity and cAMP resistant holoenzymes. Mapping the ACRDYS1 disease mutations reveals their localization to the second of two tandem cAMP-binding (CNB) domains (CNB-B), and here, we characterize a recurrent deletion mutant where the last 14 residues are missing. The crystal structure of a monomeric form of this mutant (RIα92-365) bound to the catalytic (C)-subunit reveals the dysfunctional regions of the RIα subunit. Beyond the missing residues, the entire capping motif is disordered (residues 357-379) and explains the disrupted cAMP binding. Moreover, the effects of the mutation extend far beyond the CNB-B domain and include the active site and N-lobe of the C-subunit, which is in a partially open conformation with the C-tail disordered. A key residue that contributes to this crosstalk, D267, is altered in our structure, and we confirmed its functional importance by mutagenesis. In particular, the D267 interaction with Arg241, a residue shown earlier to be important for allosteric regulation, is disrupted, thereby strengthening the interaction of D267 with the C-subunit residue Arg194 at the R:C interface. We see here how the switch between active (cAMP-bound) and inactive (holoenzyme) conformations is perturbed and how the dynamically controlled crosstalk between the helical domains of the two CNB domains is necessary for the functional regulation of PKA activity.


Assuntos
Subunidade RIalfa da Proteína Quinase Dependente de AMP Cíclico/química , Subunidade RIalfa da Proteína Quinase Dependente de AMP Cíclico/genética , AMP Cíclico/metabolismo , Disostoses/genética , Disostoses/patologia , Deficiência Intelectual/genética , Deficiência Intelectual/patologia , Proteínas Mutantes/química , Proteínas Mutantes/genética , Osteocondrodisplasias/genética , Osteocondrodisplasias/patologia , Cristalografia por Raios X , Subunidade RIalfa da Proteína Quinase Dependente de AMP Cíclico/metabolismo , Humanos , Modelos Moleculares , Proteínas Mutantes/metabolismo , Ligação Proteica , Conformação Proteica , Deleção de Sequência
6.
J Struct Biol ; 193(2): 141-54, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26687415

RESUMO

The cAMP dependent protein kinase (PKA) is a key enzyme involved in many cellular processes in eukaryotes. In mammals, the regulatory (R) subunit localises the catalytic (C) subunit to specific subcellular sites through the interaction of its N-terminal homodimeric docking and dimerization (D/D) domain with specific scaffold proteins. The structure of the D/D domain has been extensively studied in mammals, but there is little information from non-mammalian species. In this work, we present the structural analysis of the D/D domain of Bcy1, the R subunit of PKA from Saccharomyces cerevisiae. Using chemical crosslinking experiments and static light scattering measurements we found that this R subunit forms a tetramer in solution, unlike its dimeric mammalian counterparts. We determined that the D/D domain is responsible for this unusual oligomeric state. Using biophysical techniques including size-exclusion chromatography, sucrose gradient sedimentation, small angle X-ray scattering (SAXS), and circular dichroism, we performed a detailed structural characterization of the tetrameric D/D domain of Bcy1. We used homology modelling in combination with computer-aided docking methods and ab initio SAXS modelling methods to develop structural models for the D/D domain tetramer. The models consist of two homodimers with a canonical D/D domain fold that generate a dimer of dimers with novel putative interaction surfaces. These findings indicate that the oligomerization states of PKA R subunits is more diverse than previously thought, and suggest that this might allow some forms of PKA to interact with a wide range of intracellular partners.


Assuntos
Proteínas Quinases Dependentes de AMP Cíclico/química , Proteínas de Saccharomyces cerevisiae/química , Cromatografia em Gel , Dicroísmo Circular , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Modelos Moleculares , Multimerização Proteica , Estrutura Terciária de Proteína , Subunidades Proteicas/química , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Espalhamento a Baixo Ângulo , Difração de Raios X
7.
J Vasc Res ; 52(4): 244-256, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26788996

RESUMO

BACKGROUND/AIMS: Venous neointimal hyperplasia (NH) is the predominant cause of stenosis in hemodialysis arteriovenous grafts (AVG), but there is currently no clinically used therapy to prevent NH. METHODS: A porcine AVG model was used to identify potential pharmacological targets to prevent NH. Sunitinib, a broad-spectrum tyrosine kinase inhibitor, was examined as a potential anti-NH drug utilizing in vitro and ex vivo models. RESULTS: In an in vivo porcine model, PDGF, VEGF and their receptors PDGFR-α and VEGFR-2 were upregulated at the venous anastomosis within 2 weeks after AVG placement, with NH development by 4 weeks. Sunitinib inhibited PDGF-stimulated proliferation, migration, phosphorylation of MAPK and PI3K/Akt proteins and changes in the expression of cell-cycle regulatory proteins in vascular smooth-muscle cells as well as VEGF-stimulated endothelial cell proliferation in vitro. In an ex vivo model, significant NH was observed in porcine vein segments perfused for 12 days under pathological shear stress. Sunitinib (100 nM) inhibited NH formation, with the intima-to-lumen area ratio decreasing from 0.45 ± 0.25 to 0.04 ± 0.02 (p < 0.05) with treatment. CONCLUSION: These findings demonstrate sunitinib to be a potential NH-preventive drug as well as the utility of an ex vivo model to investigate pharmacotherapies under pathophysiological flow conditions.


Assuntos
Derivação Arteriovenosa Cirúrgica/efeitos adversos , Implante de Prótese Vascular/efeitos adversos , Oclusão de Enxerto Vascular/prevenção & controle , Indóis/farmacologia , Veias Jugulares/efeitos dos fármacos , Veias Jugulares/cirurgia , Neointima , Inibidores de Proteínas Quinases/farmacologia , Pirróis/farmacologia , Animais , Becaplermina , Artéria Carótida Primitiva/cirurgia , Proteínas de Ciclo Celular/metabolismo , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/enzimologia , Células Endoteliais/patologia , Feminino , Oclusão de Enxerto Vascular/enzimologia , Oclusão de Enxerto Vascular/patologia , Humanos , Hiperplasia , Veias Jugulares/enzimologia , Veias Jugulares/patologia , Miócitos de Músculo Liso/efeitos dos fármacos , Miócitos de Músculo Liso/enzimologia , Miócitos de Músculo Liso/patologia , Proteínas Proto-Oncogênicas c-sis/farmacologia , Receptores do Fator de Crescimento Derivado de Plaquetas/antagonistas & inibidores , Receptores do Fator de Crescimento Derivado de Plaquetas/metabolismo , Receptores de Fatores de Crescimento do Endotélio Vascular/antagonistas & inibidores , Receptores de Fatores de Crescimento do Endotélio Vascular/metabolismo , Transdução de Sinais/efeitos dos fármacos , Sunitinibe , Sus scrofa , Fatores de Tempo , Técnicas de Cultura de Tecidos , Fator A de Crescimento do Endotélio Vascular/farmacologia
8.
J Biol Chem ; 289(41): 28505-12, 2014 Oct 10.
Artigo em Inglês | MEDLINE | ID: mdl-25112875

RESUMO

Protein kinase A (PKA) is ubiquitously expressed and is responsible for regulating many important cellular functions in response to changes in intracellular cAMP concentrations. The PKA holoenzyme is a tetramer (R2:C2), with a regulatory subunit homodimer (R2) that binds and inhibits two catalytic (C) subunits; binding of cAMP to the regulatory subunit homodimer causes activation of the catalytic subunits. Four different R subunit isoforms exist in mammalian cells, and these confer different structural features, subcellular localization, and biochemical properties upon the PKA holoenzymes they form. The holoenzyme containing RIIß is structurally unique in that the type IIß holoenzyme is much more compact than the free RIIß homodimer. We have used small angle x-ray scattering and small angle neutron scattering to study the solution structure and subunit organization of a holoenzyme containing an RIIß C-terminal deletion mutant (RIIß(1-280)), which is missing the C-terminal cAMP-binding domain to better understand the structural organization of the type IIß holoenzyme and the RIIß domains that contribute to stabilizing the holoenzyme conformation. Our results demonstrate that compaction of the type IIß holoenzyme does not require the C-terminal cAMP-binding domain but rather involves large structural rearrangements within the linker and N-terminal cyclic nucleotide-binding domain of the RIIß homodimer. The structural rearrangements are significantly greater than seen previously with RIIα and are likely to be important in mediating short range and long range interdomain and intersubunit interactions that uniquely regulate the activity of the type IIß isoform of PKA.


Assuntos
Subunidades Catalíticas da Proteína Quinase Dependente de AMP Cíclico/química , Subunidade RIIbeta da Proteína Quinase Dependente de AMP Cíclico/química , AMP Cíclico/química , Holoenzimas/química , Animais , Domínio Catalítico , AMP Cíclico/metabolismo , Subunidades Catalíticas da Proteína Quinase Dependente de AMP Cíclico/genética , Subunidades Catalíticas da Proteína Quinase Dependente de AMP Cíclico/metabolismo , Subunidade RIIbeta da Proteína Quinase Dependente de AMP Cíclico/genética , Subunidade RIIbeta da Proteína Quinase Dependente de AMP Cíclico/metabolismo , Escherichia coli/genética , Escherichia coli/metabolismo , Expressão Gênica , Holoenzimas/genética , Holoenzimas/metabolismo , Isoenzimas/química , Isoenzimas/genética , Isoenzimas/metabolismo , Camundongos , Modelos Moleculares , Mutação , Difração de Nêutrons , Ligação Proteica , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Ratos , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Espalhamento a Baixo Ângulo , Difração de Raios X
9.
Structure ; 22(1): 59-69, 2014 Jan 07.
Artigo em Inglês | MEDLINE | ID: mdl-24316401

RESUMO

The regulatory (R) subunit is the cAMP receptor of protein kinase A. Following cAMP binding, the inactive PKA holoenzyme complex separates into two active catalytic (C) subunits and a cAMP-bound R dimer. Thus far, only monomeric R structures have been solved, which fell short in explaining differences of cAMP binding for the full-length protein as compared to the truncated R subunits. Here we solved a full-length R-dimer structure that reflects the biologically relevant conformation, and this structure agrees well with small angle X-ray scattering. An isoform-specific interface is revealed between the protomers. This interface acts as an intermolecular sensor for cAMP and explains the cooperative character of cAMP binding to the RIα dimer. Mutagenesis of residues on this interface not only leads to structural and biochemical changes, but is also linked to Carney complex disease.


Assuntos
Complexo de Carney/metabolismo , Subunidade RIalfa da Proteína Quinase Dependente de AMP Cíclico/química , AMP Cíclico/química , Sequência de Aminoácidos , Animais , Complexo de Carney/genética , Complexo de Carney/patologia , Domínio Catalítico , Bovinos , Cristalografia por Raios X , AMP Cíclico/metabolismo , Subunidade RIalfa da Proteína Quinase Dependente de AMP Cíclico/genética , Subunidade RIalfa da Proteína Quinase Dependente de AMP Cíclico/metabolismo , Escherichia coli/genética , Escherichia coli/metabolismo , Regulação da Expressão Gênica , Humanos , Modelos Moleculares , Dados de Sequência Molecular , Mutação , Ligação Proteica , Multimerização Proteica , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Transdução de Sinais
10.
Proc Natl Acad Sci U S A ; 110(5): E387-96, 2013 Jan 29.
Artigo em Inglês | MEDLINE | ID: mdl-23319652

RESUMO

cAMP-dependent protein kinase (PKA) regulates a myriad of functions in the heart, including cardiac contractility, myocardial metabolism,and gene expression. However, a molecular integrator of the PKA response in the heart is unknown. Here, we show that the PKA adaptor A-kinase interacting protein 1 (AKIP1) is up-regulated in cardiac myocytes in response to oxidant stress. Mice with cardiac gene transfer of AKIP1 have enhanced protection to ischemic stress. We hypothesized that this adaptation to stress was mitochondrial dependent. AKIP1 interacted with the mitochondrial localized apoptosis inducing factor (AIF) under both normal and oxidant stress. When cardiac myocytes or whole hearts are exposed to oxidant and ischemic stress, levels of both AKIP1 and AIF were enhanced. AKIP1 is preferentially localized to interfibrillary mitochondria and up-regulated in this cardiac mitochondrial subpopulation on ischemic injury. Mitochondria isolated from AKIP1 gene transferred hearts showed increased mitochondrial localization of AKIP1, decreased reactive oxygen species generation, enhanced calcium tolerance, decreased mitochondrial cytochrome C release,and enhance phosphorylation of mitochondrial PKA substrates on ischemic stress. These observations highlight AKIP1 as a critical molecular regulator and a therapeutic control point for stress adaptation in the heart.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Coração/fisiologia , Miocárdio/metabolismo , Miócitos Cardíacos/metabolismo , Proteínas Nucleares/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Fator de Indução de Apoptose/metabolismo , Western Blotting , Linhagem Celular Tumoral , Células Cultivadas , Células HEK293 , Células HeLa , Coração/fisiopatologia , Humanos , Peróxido de Hidrogênio/farmacologia , Técnicas In Vitro , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microscopia Confocal , Mitocôndrias Cardíacas/metabolismo , Traumatismo por Reperfusão Miocárdica/fisiopatologia , Miocárdio/citologia , Miócitos Cardíacos/citologia , Miócitos Cardíacos/efeitos dos fármacos , Proteínas Nucleares/genética , Oxidantes/farmacologia , Ligação Proteica , Ratos , Ratos Sprague-Dawley
11.
J Control Release ; 160(3): 459-67, 2012 Jun 28.
Artigo em Inglês | MEDLINE | ID: mdl-22465391

RESUMO

Synthetic arteriovenous (AV) hemodialysis grafts are plagued by hyperplasia resulting in occlusion and graft failure yet there are no clinically available preventative treatments. Here the delivery and degradation of a sirolimus-laden polymer gel were monitored in vivo by magnetic resonance imaging (MRI) and its efficacy for inhibiting hyperplasia was evaluated in a porcine model of AV graft stenosis. Synthetic grafts were placed between the carotid artery and ipsilateral jugular vein of swine. A biodegradable polymer gel loaded with sirolimus (2.5mg/mL) was immediately applied perivascularly to the venous anastomosis, and reapplied by ultrasound-guided injections at one, two and three weeks. Control grafts received neither sirolimus nor polymer. The lumen cross-sectional area at the graft-vein anastomosis was assessed in vivo by non-invasive MRI. The explanted tissues also underwent histological analysis. A specifically developed MRI pulse sequence provided a high contrast-to-noise ratio (CNR) between the polymer and surrounding tissue that allowed confirmation of gel location after injection. Polymer signal decreased up to 80% at three to four weeks after injection, slightly faster than its degradation kinetics in vitro. The MR image of the polymer was confirmed by visual assessment at necropsy. On histological assessment, the mean hyperplasia surface area of the treated graft was 52% lower than that of the control grafts (0.43mm(2) vs. 0.89mm(2); p<0.003), while the minimum cross-sectional lumen area, as measured on MRI, was doubled (5.3mm(2) vs 2.5mm(2); p<0.05). In conclusion, customized MRI allowed non-invasive monitoring of the location and degradation of drug delivery polymer gels in vivo. Perivascular application of sirolimus-laden polymer yielded a significant decrease in hyperplasia development and an increase in lumen area at the venous anastomosis of AV grafts.


Assuntos
Antibióticos Antineoplásicos/administração & dosagem , Oclusão de Enxerto Vascular/tratamento farmacológico , Hiperplasia/tratamento farmacológico , Polietilenoglicóis/administração & dosagem , Poliglactina 910/administração & dosagem , Sirolimo/administração & dosagem , Animais , Antibióticos Antineoplásicos/sangue , Antibióticos Antineoplásicos/farmacocinética , Portadores de Fármacos/administração & dosagem , Géis , Oclusão de Enxerto Vascular/metabolismo , Hiperplasia/metabolismo , Polietilenoglicóis/farmacocinética , Poliglactina 910/farmacocinética , Diálise Renal , Sirolimo/sangue , Sirolimo/farmacocinética , Sus scrofa
12.
J Biol Chem ; 284(51): 35916-25, 2009 Dec 18.
Artigo em Inglês | MEDLINE | ID: mdl-19837668

RESUMO

The catalytic (C) and regulatory (R) subunits of protein kinase A are exceptionally dynamic proteins. Interactions between the R- and C-subunits are regulated by cAMP binding to the two cyclic nucleotide-binding domains in the R-subunit. Mammalian cells express four different isoforms of the R-subunit (RIalpha, RIbeta, RIIalpha, and RIIbeta) that all interact with the C-subunit in different ways. Here, we investigate the dynamic behavior of protein complexes between RIalpha and C-subunits using small angle x-ray scattering. We show that a single point mutation in RIalpha, R333K (which alters the cAMP-binding properties of Domain B) results in a compact shape compared with the extended shape of the wild-type R.C complex. A double mutant complex that disrupts the interaction site between the C-subunit and Domain B in RIalpha, RIalpha(AB)R333K.C(K285P), results in a broader P(r) curve that more closely resembles the P(r) profiles of wild-type complexes. These results together suggest that interactions between RIalpha Domain B and the C-subunit in the RIalpha.C complex involve large scale dynamics that can be disrupted by single point mutations in both proteins. In contrast to RIalpha.C complexes. Domain B in the RIIbeta.C heterodimer is not dynamic and is critical for both inhibition and complex formation. Our study highlights the functional differences of domain dynamics between protein kinase A isoforms, providing a framework for elucidating the global organization of each holoenzyme and the cross-talk between the R- and C-subunits.


Assuntos
Subunidade RIalfa da Proteína Quinase Dependente de AMP Cíclico/química , Animais , AMP Cíclico/química , AMP Cíclico/genética , AMP Cíclico/metabolismo , Subunidade RIalfa da Proteína Quinase Dependente de AMP Cíclico/genética , Subunidade RIalfa da Proteína Quinase Dependente de AMP Cíclico/metabolismo , Humanos , Isoenzimas/química , Isoenzimas/genética , Isoenzimas/metabolismo , Mutação Puntual , Estrutura Quaternária de Proteína/fisiologia , Estrutura Terciária de Proteína/fisiologia , Difração de Raios X
13.
J Biol Chem ; 284(35): 23644-51, 2009 Aug 28.
Artigo em Inglês | MEDLINE | ID: mdl-19553663

RESUMO

Epac2 is a member of the family of exchange proteins directly activated by cAMP (Epac). Our previous studies suggest a model of Epac activation in which cAMP binding to the enzyme induces a localized "hinge" motion that reorients the regulatory lobe relative to the catalytic lobe without inducing large conformational changes within individual lobes. In this study, we identified the location of the major hinge in Epac2 by normal mode motion correlation and structural alignment analyses. Targeted mutagenesis was then performed to test the functional importance of hinge bending for Epac activation. We show that substitution of the conserved residue phenylalanine 435 with glycine (F435G) facilitates the hinge bending and leads to a constitutively active Epac2 capable of stimulating nucleotide exchange in the absence of cAMP. In contrast, substitution of the same residue with a bulkier side chain, tryptophan (F435W), impedes the hinge motion and results in a dramatic decrease in Epac2 catalytic activity. Structural parameters determined by small angle x-ray scattering further reveal that whereas the F435G mutant assumes a more extended conformation in the absence of cAMP, the F435W mutant is incapable of adopting the fully extended and active conformation in the presence of cAMP. These findings demonstrate the importance of hinge motion in Epac activation. Our study also suggests that phenylalanine at position 435 is the optimal size side chain to keep Epac closed and inactive in the absence of cAMP while still allowing the proper hinge motion for full Epac extension and activation in the presence of cAMP.


Assuntos
Fatores de Troca do Nucleotídeo Guanina/química , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Mutação , Substituição de Aminoácidos , AMP Cíclico/metabolismo , Fatores de Troca do Nucleotídeo Guanina/genética , Modelos Moleculares , Conformação Molecular , Mutagênese Sítio-Dirigida , Relação Estrutura-Atividade
14.
Basic Clin Pharmacol Toxicol ; 99(6): 431-9, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17169124

RESUMO

Dipyridamole is a potential pharmacological agent to prevent vascular stenosis because of its antiproliferative properties. The mechanisms by which dipyridamole inhibits the growth of vascular smooth muscle cells, especially venous smooth muscle cells, are unclear. In the present study, dipyridamole transiently but significantly increased cyclic adenosine monophosphate (cAMP) and cyclic guanosine monophosphate (cGMP) levels in human venous and arterial smooth muscle cells in a time- and dose-dependent manner. Peak concentrations of both cyclic nucleotides were achieved at 15-30 min. and correlated with inhibition of proliferation in both cell types. The antiproliferative effects of dipyridamole observed at 48 hr were similar whether drug exposure was only 15 min. or sustained for 48 hr. Specific competitive inhibitors of protein kinases A and G attenuated the antiproliferative effects of subsaturating concentrations of dipyridamole, with the effects of protein kinase inhibition being particularly pronounced in venous smooth muscle cells. Flow cytometry analysis showed that dipyridamole caused an enrichment of cells in G(0)/G(1) and a corresponding reduction of cells in S phase. These data indicate that a transient increase in cGMP and cAMP is sufficient to induce downstream kinase activation and subsequent cell cycle arrest, and that protein kinase G may be more important than protein kinase A in mediating the growth inhibitory effect of dipyridamole in venous protein kinase.


Assuntos
Proliferação de Células/efeitos dos fármacos , Dipiridamol/farmacologia , Miócitos de Músculo Liso/efeitos dos fármacos , Inibidores de Fosfodiesterase/farmacologia , Inibidores da Agregação Plaquetária/farmacologia , Aorta/citologia , Ciclo Celular/efeitos dos fármacos , Células Cultivadas , AMP Cíclico/análogos & derivados , AMP Cíclico/metabolismo , AMP Cíclico/farmacologia , Proteínas Quinases Dependentes de AMP Cíclico/antagonistas & inibidores , GMP Cíclico/análogos & derivados , GMP Cíclico/metabolismo , GMP Cíclico/farmacologia , Proteínas Quinases Dependentes de GMP Cíclico/antagonistas & inibidores , Inibidores Enzimáticos/farmacologia , Oclusão de Enxerto Vascular/prevenção & controle , Humanos , L-Lactato Desidrogenase/metabolismo , Músculo Liso Vascular/citologia , Miócitos de Músculo Liso/metabolismo , Veia Safena/citologia , Tionucleotídeos/farmacologia
15.
J Cell Biochem ; 99(6): 1553-63, 2006 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-16817200

RESUMO

Platelet-derived growth factor (PDGF) has been implicated in smooth muscle cell (SMC) proliferation, a key event in the development of myointimal hyperplasia in vascular grafts. Recent evidence suggests that the PDGF receptor (PDGFR) tyrosine kinase inhibitor, imatinib, can prevent arterial proliferative diseases. Because hyperplasia is far more common at the venous anastomosis than the arterial anastomosis in vascular grafts, we investigated whether imatinib also inhibited venous SMC (VSMC) proliferation, and examined possible differences in its mechanism of action between VSMC and arterial SMC (ASMC). Human ASMC and VSMC were stimulated with PDGF-AB, in the presence or absence of imatinib (0.1-10 microM). Proliferation was assayed using the 5-bromo-2'-deoxyuridine (BrdU) incorporation assay, while PDGFR, Akt and ERK1/2-mitogen activated protein kinase (MAPK) signaling pathways were investigated by immunoblotting. The proliferative response to PDGF at 50 and 100 ng/ml was 32 and 43% greater, respectively, in VSMC than in ASMC. Similarly, PDGF-stimulated proliferation was more sensitive to inhibition by imatinib in VSMC than ASMC (IC(50) = 0.05 microM vs. 0.4 microM; P < 0.01). Imatinib also more effectively inhibited PDGF-induced phosphorylation of PDGFRbeta and Akt in VSMC, compared to ASMC. These data highlight inherent pharmacodynamic differences between VSMC and ASMC in receptor and cell signaling functions and suggest that imatinib therapy may be useful for the prevention of venous stenosis in vascular grafts.


Assuntos
Proliferação de Células/efeitos dos fármacos , Músculo Liso Vascular/efeitos dos fármacos , Piperazinas/farmacologia , Fator de Crescimento Derivado de Plaquetas/farmacologia , Pirimidinas/farmacologia , Receptores do Fator de Crescimento Derivado de Plaquetas/metabolismo , Artérias/citologia , Artérias/efeitos dos fármacos , Artérias/metabolismo , Benzamidas , Western Blotting , Ciclo Celular/efeitos dos fármacos , Células Cultivadas , Citometria de Fluxo , Humanos , Mesilato de Imatinib , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Músculo Liso Vascular/citologia , Músculo Liso Vascular/metabolismo , Fosforilação , Proteínas Proto-Oncogênicas c-akt/metabolismo , Veias/citologia , Veias/efeitos dos fármacos , Veias/metabolismo
16.
J Cell Biochem ; 99(3): 835-44, 2006 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-16721826

RESUMO

Proliferation of smooth muscle cells (SMCs) contributes to the stenosis of coronary arteries and vascular grafts. Local delivery of anti-proliferative drugs can prevent vascular stenosis. To understand the cellular responses to anti-proliferative agents, we investigated the signaling events in cultured human aortic SMCs (ASMCs), saphenous venous SMCs (VSMCs), and dermal fibroblasts (DFs) in response to paclitaxel or etoposide. Cellular mitochondrial and proliferative activities were examined with the methylthiazoletetrazolium (MTT) dye reduction and the bromodeoxyuridine (BrdU) incorporation assay, respectively. Cell proliferation was almost completely suppressed by paclitaxel or etoposide, but apoptosis was achieved in only about 50% of cells at the highest drug concentrations, suggesting the presence of compensatory mechanisms to prevent apoptosis. Examination of three important signaling pathways revealed significant differences between ASMCs, VSMCs, and DFs. Treatment with either paclitaxel or etoposide caused a transient phosphorylation/activation of p42 MAPK in ASMCs and DFs, but had no effect on phospho-p42/44 MAPK in VSMCs. High-dose etoposide enhanced p38 MAPK activation in ASMCs, but not in VSMCs. The p38 inhibitor, PD169316, partially inhibited etoposide-induced ASMC apoptosis, but induced apoptosis in VSMCs. The effects of etoposide and paclitaxel on Akt also differed between ASMCs and VSMCs. These observations indicate that ASMCs and VSMCs differ in the response of signaling pathways to anti-proliferative agents. In ASMCs, p42/44 MAPK appears to serve a pro-survival role, whereas p38 MAPK is a pro-apoptotic regulator. In contrast, p38 MAPK is an important pro-survival regulator in VSMCs and p42/44 MAPK appears to play a minor role in responding to anti-proliferative drugs.


Assuntos
Proliferação de Células/efeitos dos fármacos , Etoposídeo/farmacologia , Sistema de Sinalização das MAP Quinases/fisiologia , Músculo Liso Vascular/citologia , Miócitos de Músculo Liso/efeitos dos fármacos , Miócitos de Músculo Liso/metabolismo , Paclitaxel/farmacologia , Animais , Antineoplásicos Fitogênicos/farmacologia , Aorta/citologia , Células Cultivadas , Fibroblastos/citologia , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Humanos , Músculo Liso Vascular/efeitos dos fármacos , Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/citologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Veia Safena/citologia , Moduladores de Tubulina/farmacologia
17.
J Mol Biol ; 357(3): 880-9, 2006 Mar 31.
Artigo em Inglês | MEDLINE | ID: mdl-16460759

RESUMO

Isoform diversity within the protein kinase A (PKA) family is achieved by catalytic (C) subunits binding to different isoforms of regulatory subunit homodimers (R2). In a previous small-angle X-ray scattering study, we showed that the type Ialpha R2 homodimer has a distinctive Y-shaped structure, while the IIalpha and IIbeta homodimers are highly flexible and extended in solution. Here we present the results of X-ray scattering experiments on different isoforms of the PKA holoenzyme (R2C2) and show that the type IIbeta R2 homodimer undergoes a dramatic compaction upon binding C subunits that involves a 10A reduction in radius of gyration (from 56 to 46 A) and a 35 A shortening of the maximum linear dimension (from 180-145 A). In contrast, the type IIalpha R2 homodimer shows very little change in these structural parameters and remains extended upon C-subunit binding. This large difference is surprising given the highly conserved sequence and domain organization for the different R isoforms. A mutant RIIbeta holoenzyme and an RIIalpha/RIIbeta chimera were used to explore the role of the sequence linking different functional domains within RIIbeta in the observed C subunit-induced compaction. Structural modeling was used to aid in interpreting the scattering results in terms of the role of inter-domain and inter-subunit contacts in determining the global conformations of the different isoforms. The results provide an important structural foundation for understanding isoform-specific PKA localization and signaling.


Assuntos
Proteínas Quinases Dependentes de AMP Cíclico/química , Sequência de Aminoácidos , Animais , Bovinos , Proteína Quinase Tipo II Dependente de AMP Cíclico , Proteínas Quinases Dependentes de AMP Cíclico/genética , Dimerização , Humanos , Isoenzimas/química , Isoenzimas/genética , Proteínas de Membrana/química , Proteínas de Membrana/genética , Modelos Moleculares , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Estrutura Terciária de Proteína , Espalhamento de Radiação , Soluções , Raios X
18.
J Biol Chem ; 280(42): 35521-7, 2005 Oct 21.
Artigo em Inglês | MEDLINE | ID: mdl-16109722

RESUMO

Different isoforms of the full-length protein kinase A (PKA) regulatory subunit homodimer (R2) and the catalytic (C) subunit-bound holoenzyme (R2C2) have very different global structures despite similar molecular weights and domain organization within their primary sequences. To date, it has been the linker sequence between the R subunit dimerization/docking domain and cAMP-binding domain A that has been implicated in modulating domain interactions to give rise to these differences in global structure. The small angle solution scattering data presented here for three different isoforms of PKA heterodimer (deltaR-C) complexes reveal a role for another conformationally dynamic sequence in modulating inter-subunit and domain interactions, the C helix that connects the cAMP-binding domains A and B of the R subunit. The deltaR-C heterodimer complexes studied here were each formed with a monomeric N-terminal deletion mutant of the R subunit (deltaR) that contains the inhibitor sequence and both cAMP-binding domains. The scattering data show that type IIalpha and type IIbeta deltaR-C heterodimers are relatively compact and globular, with the C subunit contacting the inhibitor sequence and both cAMP-binding domains. In contrast, the type Ialpha heterodimer is significantly more extended, with the C subunit interacting with the inhibitor sequence and cAMP-binding domain A, whereas domain B extends out such that its surface is almost completely solvent exposed. These data implicate the C helix of RIalpha in modulating isoform-specific interdomain communication in the PKA holoenzyme, adding another layer of structural complexity to our understanding of signaling dynamics in this multisubunit, multidomain protein kinase.


Assuntos
Proteínas Quinases Dependentes de AMP Cíclico/química , Animais , Cálcio/química , Catálise , Domínio Catalítico , Bovinos , AMP Cíclico/metabolismo , Subunidade RIalfa da Proteína Quinase Dependente de AMP Cíclico , Dimerização , Escherichia coli/metabolismo , Deleção de Genes , Modelos Moleculares , Modelos Estatísticos , Mutação , Ligação Proteica , Conformação Proteica , Isoformas de Proteínas , Estrutura Terciária de Proteína , Ratos , Proteínas Recombinantes/química , Espalhamento de Radiação , Raios X
19.
Biochemistry ; 43(19): 5629-36, 2004 May 18.
Artigo em Inglês | MEDLINE | ID: mdl-15134437

RESUMO

It has been widely accepted that cAMP activates the protein kinase A (PKA) holoenzyme by dissociating the regulatory and catalytic subunits, thus freeing the catalytic subunit to phosphorylate its targets. However, recent experiments suggest that cAMP does not fully dissociate the holoenzyme. Here, we investigate this mechanism further by using small-angle X-ray scattering to study, at physiological enzyme concentrations, the type Ialpha and type IIbeta holoenzyme structures under equilibrium solution conditions without any labeling of the protein subunits. We observe that while the addition of a molar excess of cAMP to the type Ialpha PKA holoenzyme causes partial dissociation, it is only upon addition of a PKA peptide substrate together with cAMP that full dissociation occurs. Similarly, addition of excess cAMP to the type IIbeta holoenzyme causes only a partial dissociation. However, while the addition of peptide substrate as well as excess cAMP causes somewhat more dissociation, a significant percentage of intact type IIbeta holoenzyme remains. These results confirm that both the type Ialpha and the type IIbeta holoenzymes are more stable in the presence of cAMP than previously thought. They also demonstrate that substrate plays a differential role in the activation of type I versus type II holoenzymes, which could explain some important functional differences between PKA isoforms. On the basis of these data and other recently published data, we propose a structural model of type I holoenzyme activation by cAMP.


Assuntos
Proteínas Quinases Dependentes de AMP Cíclico/química , Subunidades Proteicas/química , Animais , Domínio Catalítico/fisiologia , Bovinos , AMP Cíclico/química , Subunidade RIIbeta da Proteína Quinase Dependente de AMP Cíclico , Subunidade RIalfa da Proteína Quinase Dependente de AMP Cíclico , Proteína Quinase Tipo II Dependente de AMP Cíclico , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/fisiologia , Holoenzimas/química , Holoenzimas/metabolismo , Holoenzimas/fisiologia , Oligopeptídeos/química , Subunidades Proteicas/metabolismo , Subunidades Proteicas/fisiologia , Espalhamento de Radiação , Soluções , Especificidade por Substrato , Raios X
20.
J Mol Biol ; 337(5): 1183-94, 2004 Apr 09.
Artigo em Inglês | MEDLINE | ID: mdl-15046986

RESUMO

The regulatory (R) subunits of the cAMP-dependent protein kinase (protein kinase A or PKA) are multi-domain proteins responsible for conferring cAMP-dependence and localizing PKA to specific subcellular locations. There are four isoforms of the R subunit in mammals that are similar in molecular mass and domain organization, but clearly serve different biological functions. Although high-resolution structures are available for the cAMP-binding domains and dimerization/docking domains of two isoforms, there are no high-resolution structures of any of the intact R subunit homodimer isoforms. The results of small-angle X-ray scattering studies presented here indicate that the RIalpha, RIIalpha, and RIIbeta homodimers differ markedly in overall shape, despite extensive sequence homology and similar molecular masses. The RIIalpha and RIIbeta homodimers have very extended, rod-like shapes, whereas the RIalpha homodimer likely has a compact Y-shape. Based on a comparison of the R subunit sequences, we predict that the linker regions are the likely cause of these large differences in shape among the isoforms. In addition, we show that cAMP binding does not cause large conformational changes in type Ialpha or IIalpha R subunit homodimers, suggesting that the activation of PKA by cAMP involves only local conformational changes in the R subunits.


Assuntos
Proteínas Quinases Dependentes de AMP Cíclico/química , Subunidades Proteicas/química , Animais , Bovinos , AMP Cíclico/química , AMP Cíclico/farmacologia , Subunidade RIIalfa da Proteína Quinase Dependente de AMP Cíclico , Subunidade RIIbeta da Proteína Quinase Dependente de AMP Cíclico , Subunidade RIalfa da Proteína Quinase Dependente de AMP Cíclico , Subunidade RIbeta da Proteína Quinase Dependente de AMP Cíclico , Evolução Molecular , Modelos Moleculares , Conformação Proteica/efeitos dos fármacos , Isoformas de Proteínas/química , Estrutura Terciária de Proteína , Soluções , Difração de Raios X
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA