Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
1.
Int J Mol Sci ; 21(5)2020 Mar 10.
Artigo em Inglês | MEDLINE | ID: mdl-32164249

RESUMO

Mucosal healing determined by endoscopy is currently the remission standard for ulcerative colitis (UC). However, new criteria for remission are emerging, such as histologic normalization, which appears to correlate better to the risk of relapse. Here, we study mucosal healing on a molecular and functional level in quiescent UC. We obtained endoscopic biopsies from 33 quiescent UC patients and from 17 controls. Histology was assessed using Geboes score. Protein and mRNA levels were evaluated for the tight junction proteins claudin-2, claudin-4, occludin, and tricellulin, as well as Cl-/HCO3- exchanger DRA, and cyclo-oxygenase enzymes (COX-1, COX-2). The mucosal activity of COX-1 and COX-2 enzymes was assessed in modified Ussing chambers, measuring electrogenic ion transport (short-circuit current, SCC). Chronic inflammation was present in most UC patients. The protein level of claudin-4 was reduced, while mRNA-levels of claudin-2 and claudin-4 were upregulated in UC patients. Surprisingly, the mRNA level of COX-1 was downregulated, but was unaltered for COX-2. Basal ion transport was not affected, while COX-2 inhibition induced a two-fold larger decrease in SCC in UC patients. Despite being in clinical and endoscopic remission, quiescent UC patients demonstrated abnormal mucosal barrier properties at the molecular and functional level. Further exploration of mucosal molecular signature for revision of current remission standards should be considered.


Assuntos
Claudina-1/genética , Claudinas/genética , Colite Ulcerativa/patologia , Ciclo-Oxigenase 1/genética , Ciclo-Oxigenase 2/genética , Proteína 2 com Domínio MARVEL/genética , Adulto , Idoso , Biópsia , Estudos de Casos e Controles , Claudina-1/metabolismo , Claudinas/metabolismo , Colite Ulcerativa/genética , Colite Ulcerativa/metabolismo , Ciclo-Oxigenase 1/metabolismo , Ciclo-Oxigenase 2/metabolismo , Feminino , Regulação da Expressão Gênica , Redes Reguladoras de Genes , Humanos , Proteína 2 com Domínio MARVEL/metabolismo , Masculino , Pessoa de Meia-Idade , Adulto Jovem
2.
Pharm Res ; 33(3): 716-28, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26563206

RESUMO

PURPOSE: Aggregation aspects of therapeutic monoclonal antibodies (mAbs) are of common concern to the pharmaceutical industry. Low pH treatment is applied during affinity purification and to inactivate endogenous retroviruses, directing interest to the mechanisms of acid-induced antibody aggregation. METHODS: We characterized the oligomerization kinetics at pH 3.3, as well as the reversibility upon neutralization, of three model mAbs with identical variable regions, representative of IgG1, IgG2 and IgG4 respectively. We applied size-exclusion high performance liquid chromatography and orthogonal analytical methods, including small-angle X-ray scattering and dynamic light scattering and supplemented the experimental data with crystal structure-based spatial aggregation propensity (SAP) calculations. RESULTS: We revealed distinct solution behaviors between the three mAb models: At acidic pH IgG1 retained monomeric, whereas IgG2 and IgG4 exhibited two-phase oligomerization processes. After neutralization, IgG2 oligomers partially reverted to the monomeric state, while on the contrary, IgG4 oligomers tended to aggregate. Subclass-specific aggregation-prone motifs on the Fc fragments were identified, which may lead to two distinct pathways of reversible and irreversible aggregation, respectively. CONCLUSIONS: We conclude that subtle variations in mAb sequence greatly affect responses towards low-pH incubation and subsequent neutralization, and demonstrate how orthogonal biophysical methods distinguish between reversible and irreversible mAb aggregation pathways at early stages of acidic treatment.


Assuntos
Ácidos/química , Anticorpos Monoclonais/química , Cromatografia Líquida de Alta Pressão/métodos , Humanos , Concentração de Íons de Hidrogênio , Imunoglobulina G/química , Cinética , Multimerização Proteica , Soluções/química
3.
Traffic ; 14(4): 399-411, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23324056

RESUMO

The voltage-gated potassium channel KV 7.1 is regulated by non-pore forming regulatory KCNE ß-subunits. Together with KCNE1, it forms the slowly activating delayed rectifier potassium current IKs . However, where the subunits assemble and which of the subunits determines localization of the IKs -complex has not been unequivocally resolved yet. We employed trafficking-deficient KV 7.1 and KCNE1 mutants to investigate IKs trafficking using the polarized Madin-Darby Canine Kidney cell line. We find that the assembly happens early in the secretory pathway but provide three lines of evidence that it takes place in a post-endoplasmic reticulum compartment. We demonstrate that KV 7.1 targets the IKs -complex to the basolateral membrane, but that KCNE1 can redirect the complex to the apical membrane upon mutation of critical KV 7.1 basolateral targeting signals. Our data provide a possible explanation to the fact that KV 7.1 can be localized apically or basolaterally in different epithelial tissues and offer a solution to divergent literature results regarding the effect of KCNE subunits on the subcellular localization of KV 7.1/KCNE complexes.


Assuntos
Canal de Potássio KCNQ1/metabolismo , Canais de Potássio de Abertura Dependente da Tensão da Membrana/metabolismo , Multimerização Proteica , Subunidades Proteicas/metabolismo , Animais , Membrana Celular/metabolismo , Cães , Retículo Endoplasmático/metabolismo , Complexo de Golgi/metabolismo , Canal de Potássio KCNQ1/química , Canal de Potássio KCNQ1/genética , Células Madin Darby de Rim Canino , Mutação de Sentido Incorreto , Canais de Potássio de Abertura Dependente da Tensão da Membrana/química , Canais de Potássio de Abertura Dependente da Tensão da Membrana/genética , Domínios e Motivos de Interação entre Proteínas , Sinais Direcionadores de Proteínas/genética , Subunidades Proteicas/genética , Transporte Proteico , Via Secretória
4.
Am J Physiol Cell Physiol ; 309(10): C693-706, 2015 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-26405101

RESUMO

The potassium channel Kv7.1 plays critical physiological roles in both heart and epithelial tissues. In heart, Kv7.1 and the accessory subunit KCNE1 forms the slowly activating delayed-rectifier potassium current current, which is enhanced by protein kinase A (PKA)-mediated phosphorylation. The observed current increase requires both phosphorylation of Kv7.1 and the presence of KCNE1. However, PKA also stimulates Kv7.1 currents in epithelial tissues, such as colon, where the channel does not coassemble with KCNE1. Here, we demonstrate that PKA activity significantly impacts the subcellular localization of Kv7.1 in Madin-Darby canine kidney cells. While PKA inhibition reduced the fraction of channels at the cell surface, PKA activation increased it. We show that PKA inhibition led to intracellular accumulation of Kv7.1 in late endosomes/lysosomes. By mass spectroscopy we identified eight phosphorylated residues on Kv7.1, however, none appeared to play a role in the observed response. Instead, we found that PKA acted by regulating endocytic trafficking involving the ubiquitin ligase Nedd4-2. We show that a Nedd4-2-resistant Kv7.1-mutant displayed significantly reduced intracellular accumulation upon PKA inhibition. Similar effects were observed upon siRNA knockdown of Nedd4-2. However, although Nedd4-2 is known to regulate Kv7.1 by ubiquitylation, biochemical analyses demonstrated that PKA did not influence the amount of Nedd4-2 bound to Kv7.1 or the ubiquitylation level of the channel. This suggests that PKA influences Nedd4-2-dependent Kv7.1 transport though a different molecular mechanism. In summary, we identify a novel mechanism whereby PKA can increase Kv7.1 current levels, namely by regulating Nedd4-2-dependent Kv7.1 transport.


Assuntos
Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Complexos Endossomais de Distribuição Requeridos para Transporte/metabolismo , Canal de Potássio KCNQ1/metabolismo , Transporte Proteico/fisiologia , Vesículas Transportadoras/fisiologia , Ubiquitina-Proteína Ligases/metabolismo , Animais , Linhagem Celular , Proteínas Quinases Dependentes de AMP Cíclico/genética , Cães , Complexos Endossomais de Distribuição Requeridos para Transporte/genética , Regulação Enzimológica da Expressão Gênica , Técnicas de Silenciamento de Genes , Canal de Potássio KCNQ1/genética , Mutação , Ubiquitina-Proteína Ligases Nedd4 , Conformação Proteica , Inibidores de Proteínas Quinases/farmacologia , Ubiquitina-Proteína Ligases/genética
5.
Traffic ; 13(1): 143-56, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21957902

RESUMO

The potassium channel Kv7.1 is expressed in the heart, where it contributes to the repolarization of the cardiac action potential. Additionally, Kv7.1 is expressed in epithelial tissues playing a role in salt and water transport. We recently demonstrated that surface-expressed Kv7.1 is internalized in response to polarization of the epithelial Madin-Darby canine kidney (MDCK) cell line and that this was mediated by activation of protein kinase C (PKC). In this study, the pathway downstream of PKC, which leads to internalization of Kv7.1 upon cell polarization, is elucidated. We show by confocal microscopy that Kv7.1 is endocytosed upon initiation of the polarization process and sent for degradation by the lysosomal pathway. The internalization could be mimicked by pharmacological activation of the AMP-activated protein kinase (AMPK) using three different AMPK activators. We demonstrate that the downstream effector of AMPK is the E3 ubiquitin ligase Nedd4-2. Additionally, we show that AMPK activation results in a downregulation of Kv7.1 currents in Xenopus oocytes through a Nedd4-2-dependent mechanism. In summary, surface-expressed Kv7.1 channels are endocytosed and sent for degradation in lysosomes by an AMPK-mediated activation of Nedd4-2 during the initial phase of the MDCK cell polarization process.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Polaridade Celular , Endocitose , Canal de Potássio KCNQ1/biossíntese , Potenciais de Ação , Animais , Western Blotting , Cálcio/metabolismo , Linhagem Celular , Cães , Regulação para Baixo , Complexos Endossomais de Distribuição Requeridos para Transporte/metabolismo , Imunofluorescência , Humanos , Canal de Potássio KCNQ1/genética , Lisossomos/metabolismo , Microscopia Confocal , Ubiquitina-Proteína Ligases Nedd4 , Oócitos/metabolismo , Proteína Quinase C/metabolismo , Transporte Proteico , Transfecção , Ubiquitina-Proteína Ligases/metabolismo , Proteínas de Xenopus , Xenopus laevis
6.
Am J Hum Genet ; 86(6): 872-80, 2010 Jun 11.
Artigo em Inglês | MEDLINE | ID: mdl-20560207

RESUMO

Congenital long QT syndrome (LQTS) is a hereditary disorder that leads to sudden cardiac death secondary to fatal cardiac arrhythmias. Although many genes for LQTS have been described, the etiology remains unknown in 30%-40% of cases. In the present study, a large Chinese family (four generations, 49 individuals) with autosomal-dominant LQTS was clinically evaluated. Genome-wide linkage analysis was performed by using polymorphic microsatellite markers to map the genetic locus, and positional candidate genes were screened by sequencing for mutations. The expression pattern and functional characteristics of the mutated protein were investigated by western blotting and patch-clamp electrophysiology. The genetic locus of the LQTS-associated gene was mapped to chromosome 11q23.3-24.3. A heterozygous mutation (Kir3.4-Gly387Arg) was identified in the G protein-coupled, inwardly rectifying potassium channel subunit Kir3.4, encoded by the KCNJ5 gene. The Kir3.4-Gly387Arg mutation was present in all nine affected family members and absent in 528 ethnically matched controls. Western blotting of human cardiac tissue demonstrated significant Kir3.4 expression levels in the cardiac ventricles. Heterologous expression studies with Kir3.4-Gly387Arg revealed a loss-of-function electrophysiological phenotype resulting from reduced plasma membrane expression. Our findings suggest a role for Kir3.4 in the etiology of LQTS.


Assuntos
Canais de Potássio Corretores do Fluxo de Internalização Acoplados a Proteínas G/genética , Adolescente , Adulto , Idoso , Aminofilina , Atropina , Mapeamento Cromossômico , Combinação de Medicamentos , Feminino , Canais de Potássio Corretores do Fluxo de Internalização Acoplados a Proteínas G/metabolismo , Ligação Genética , Humanos , Recém-Nascido , Síndrome do QT Longo/genética , Masculino , Pessoa de Meia-Idade , Mutação , Miocárdio/metabolismo , Nitroglicerina , Papaverina , Linhagem , Fenobarbital
7.
Mol Cell Neurosci ; 48(4): 288-97, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21627990

RESUMO

The computational ability of CNS neurons depends critically on the specific localization of ion channels in the somatodendritic and axonal membranes. Neuronal dendrites receive synaptic inputs at numerous spines and integrate them in time and space. The integration of synaptic potentials is regulated by voltage-gated potassium (Kv) channels, such as Kv4.2, which are specifically localized in the dendritic membrane. The synaptic potentials eventually depolarize the membrane of the axon initial segment, thereby activating voltage-gated sodium channels to generate action potentials. Specific Kv channels localized in the axon initial segment, such as Kv1 and Kv7 channels, determine the shape and the rate of action potentials. Kv1 and Kv7 channels present at or near nodes of Ranvier and in presynaptic terminals also influence the propagation of action potentials and neurotransmitter release. The physiological significance of proper Kv channel localization is emphasized by the fact that defects in the trafficking of Kv channels are observed in several neurological disorders including epilepsy. In this review, we will summarize the current understanding of the mechanisms of Kv channel trafficking and discuss how they contribute to the establishment and maintenance of the specific localization of Kv channels in neurons.


Assuntos
Neurônios/fisiologia , Canais de Potássio de Abertura Dependente da Tensão da Membrana/metabolismo , Animais , Transporte Proteico/fisiologia , Transmissão Sináptica/fisiologia
8.
Am J Physiol Cell Physiol ; 300(4): C814-24, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21228319

RESUMO

The potassium channel K(V)7.1 is expressed in the heart where it contributes to the repolarization of the cardiac action potential. In addition, K(V)7.1 is expressed in epithelial tissues where it plays a role in salt and water transport. Mutations in the kcnq1 gene can lead to long QT syndrome and deafness, and several mutations have been described as trafficking mutations. To learn more about the basic mechanisms that regulate K(V)7.1 surface expression, we have investigated the trafficking of K(V)7.1 during the polarization process of the epithelial cell line Madin-Darby Canine Kidney (MDCK) using a modified version of the classical calcium switch. We discovered that K(V)7.1 exhibits a very dynamic localization pattern during the calcium switch. When MDCK cells are kept in low calcium medium, K(V)7.1 is mainly observed at the plasma membrane. During the first hours of the switch, K(V)7.1 is removed from the plasma membrane and an intracellular accumulation in the endoplasmic reticulum (ER) is observed. The channel is retained in the ER until the establishment of the lateral membranes at which point K(V)7.1 is released from the ER and moves to the plasma membrane. Our data furthermore suggest that while the removal of K(V)7.1 from the cell surface and its accumulation in the ER could involve activation of protein kinase C, the subsequent release of K(V)7.1 from the ER depends on phosphoinositide 3-kinase (PI3K) activation. In conclusion, our results demonstrate that K(V)7.1 surface expression is regulated by signaling mechanisms involved in epithelial cell polarization in particular signaling cascades involving protein kinase C and PI3K.


Assuntos
Polaridade Celular/fisiologia , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Canal de Potássio KCNQ1/metabolismo , Junções Aderentes/metabolismo , Animais , Cálcio/metabolismo , Linhagem Celular , Membrana Celular/metabolismo , Desmossomos/metabolismo , Cães , Retículo Endoplasmático/metabolismo , Ativação Enzimática , Humanos , Canal de Potássio KCNQ1/genética , Fosfatidilinositol 3-Quinases/metabolismo , Proteína Quinase C/metabolismo , Transporte Proteico , Transdução de Sinais/fisiologia , Junções Íntimas/metabolismo
9.
Eur J Gastroenterol Hepatol ; 33(6): 817-824, 2021 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-33079783

RESUMO

BACKGROUND: Colonic tuft cells are epithelial chemosensory cells involved in barrier integrity, modulation of inflammatory responses and gut homeostasis. Recent evidence indicates an involvement of tuft cells in ulcerative colitis pathogenesis, though mechanisms remain largely unknown.Here, we quantified the colonic tuft cell population in patients with quiescent ulcerative colitis as compared to patients without identified colonic disease (controls). METHODS: In this retrospective study, we obtained endoscopic colonic sigmoid biopsies from 14 patients with quiescent ulcerative colitis and from 17 controls. In a blinded central-reading design, we identified tuft cells by immunohistochemistry using a cyclooxygenase-1 antibody as a marker and performed a simple counting by visual inspection. Poisson regression was employed for statistics and results were adjusted for gender, age and smoking status. RESULTS: Ulcerative colitis patients demonstrated a 55% reduced tuft cell count in colonic mucosa compared with the control group (95% confidence limit: range 31-71%, P = 0.0002). Ulcerative colitis patients had a mean tuft cells count of 46 tuft cells/mm2 (95% CI, 36-59), while controls demonstrated a mean of 104 tuft cells/mm2 (95% CI, 79-136). No interactions of other covariates, such as age, smoking status, total duration of ulcerative colitis disease and duration of clinical remission prior to study inclusion were detected between ulcerative colitis patients and controls. CONCLUSION: Quiescent ulcerative colitis patients have a relatively low number of colonic tuft cells. Further studies are warranted to explore the potential involvement of tuft cells in ulcerative colitis pathogenesis.


Assuntos
Colite Ulcerativa , Colite , Colite Ulcerativa/diagnóstico , Colo , Humanos , Mucosa Intestinal , Estudos Retrospectivos
10.
Biochem J ; 413(2): 251-9, 2008 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-18402552

RESUMO

HAI-1 [HGF (hepatocyte growth factor) activator inhibitor-1] is a Kunitz-type transmembrane serine protease inhibitor that forms inhibitor complexes with the trypsin-like serine protease, matriptase. HAI-1 is essential for mouse placental development and embryo survival and together with matriptase it is a key regulator of carcinogenesis. HAI-1 is expressed in polarized epithelial cells, which have the plasma membrane divided by tight junctions into an apical and a basolateral domain. In the present study we show that HAI-1 at steady-state is mainly located on the basolateral membrane of both Madin-Darby canine kidney cells and mammary gland epithelial cells. After biosynthesis, HAI-1 is exocytosed mainly to the basolateral plasma membrane from where 15% of the HAI-1 molecules are proteolytically cleaved and released into the basolateral medium. The remaining membrane-associated HAI-1 is endocytosed and then recycles between the basolateral plasma membrane and endosomes for hours until it is transcytosed to the apical plasma membrane. Minor amounts of HAI-1 present at the apical plasma membrane are proteolytically cleaved and released into the apical medium. Full-length membrane-bound HAI-1 has a half-life of 1.5 h and is eventually degraded in the lysosomes, whereas proteolytically released HAI-1 is more stable. HAI-1 is co-localized with its cognate protease, matriptase, at the basolateral plasma membrane. We suggest that HAI-1, in addition to its protease inhibitory function, plays a role in transporting matriptase as a matriptase-HAI-1 complex from the basolateral plama membrane to the apical plasma membrane, as matriptase is known to interact with prostasin, located at the apical plasma membrane.


Assuntos
Regulação da Expressão Gênica , Glicoproteínas de Membrana/fisiologia , Animais , Linhagem Celular , Membrana Celular/metabolismo , Cães , Endocitose , Exocitose , Lisossomos/metabolismo , Glândulas Mamárias Animais/metabolismo , Glicoproteínas de Membrana/metabolismo , Camundongos , Modelos Biológicos , Transporte Proteico , Proteínas Secretadas Inibidoras de Proteinases , Serina Endopeptidases/metabolismo
11.
Biochem J ; 405(3): 429-38, 2007 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-17456045

RESUMO

The remarkably high specificity of the coagulation proteases towards macromolecular substrates is provided by numerous interactions involving the catalytic groove and remote exosites. For FVIIa [activated FVII (Factor VII)], the principal initiator of coagulation via the extrinsic pathway, several exosites have been identified, whereas only little is known about the specificity dictated by the active-site architecture. In the present study, we have profiled the primary P4-P1 substrate specificity of FVIIa using positional scanning substrate combinatorial libraries and evaluated the role of the selective active site in defining specificity. Being a trypsin-like serine protease, FVIIa had P1 specificity exclusively towards arginine and lysine residues. In the S2 pocket, threonine, leucine, phenylalanine and valine residues were the most preferred amino acids. Both S3 and S4 appeared to be rather promiscuous, however, with some preference for aromatic amino acids at both positions. Interestingly, a significant degree of interdependence between the S3 and S4 was observed and, as a consequence, the optimal substrate for FVIIa could not be derived directly from a subsite-directed specificity screen. To evaluate the role of the active-site residues in defining specificity, a series of mutants of FVIIa were prepared at position 239 (position 99 in chymotrypsin), which is considered to be one of the most important residues for determining P2 specificity of the trypsin family members. This was confirmed for FVIIa by marked changes in primary substrate specificity and decreased rates of antithrombin III inhibition. Interestingly, these changes do not necessarily coincide with an altered ability to activate Factor X, demonstrating that inhibitor and macromolecular substrate selectivity may be engineered separately.


Assuntos
Fator VIIa/antagonistas & inibidores , Fator VIIa/metabolismo , Engenharia de Proteínas/métodos , Sequência de Aminoácidos , Humanos , Cinética , Modelos Moleculares , Conformação Proteica , Especificidade por Substrato
12.
FEBS Lett ; 581(1): 71-6, 2007 Jan 09.
Artigo em Inglês | MEDLINE | ID: mdl-17182039

RESUMO

Coagulation factor VIIa (FVIIa) belongs to a family of proteases being part of the stepwise, self-amplifying blood coagulation cascade. To investigate the impact of the mutation Met(298{156})Lys in FVIIa, we replaced the Gly(283{140})-Met(298{156}) loop with the corresponding loop of factor Xa. The resulting variant exhibited increased intrinsic activity, concurrent with maturation of the active site, a less accessible N-terminus, and, interestingly, an altered macromolecular substrate specificity reflected in an increased ability to cleave factor IX (FIX) and a decreased rate of FX activation compared to that of wild-type FVIIa. In complex with tissue factor, activation of FIX, but not of FX, returned to normal. Deconvolution of the loop graft in order to identify important side chain substitutions resulted in the mutant Val(158{21})Asp/Leu(287{144})Thr/Ala(294{152})Ser/Glu(296{154}) Ile/Met(298{156})Lys-FVIIa with almost the same activity and specificity profile. We conclude that a lysine residue in position 298{156} of FVIIa requires a hydrophilic environment to be fully accommodated. This position appears critical for substrate specificity among the proteases of the blood coagulation cascade due to its prominent position in the macromolecular exosite and possibly via its interaction with the corresponding position in the substrate (i.e. FIX or FX).


Assuntos
Fator VIIa/química , Substituição de Aminoácidos , Sítios de Ligação/genética , Coagulação Sanguínea/genética , Ativação Enzimática/genética , Fator VIIa/genética , Fator VIIa/metabolismo , Fator Xa/química , Fator Xa/genética , Fator Xa/metabolismo , Humanos , Complexos Multienzimáticos/química , Complexos Multienzimáticos/genética , Complexos Multienzimáticos/metabolismo , Mutação de Sentido Incorreto , Estrutura Secundária de Proteína/genética , Estrutura Terciária de Proteína/genética , Especificidade por Substrato/genética , Tromboplastina/química , Tromboplastina/genética , Tromboplastina/metabolismo
13.
Biochem J ; 396(2): 391-9, 2006 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-16475979

RESUMO

Dipeptidyl peptidases 8 and 9 have been identified as gene members of the S9b family of dipeptidyl peptidases. In the present paper, we report the characterization of recombinant dipeptidyl peptidases 8 and 9 using the baculovirus expression system. We have found that only the full-length variants of the two proteins can be expressed as active peptidases, which are 882 and 892 amino acids in length for dipeptidyl peptidase 8 and 9 respectively. We show further that the purified proteins are active dimers and that they show similar Michaelis-Menten kinetics and substrate specificity. Both cleave the peptide hormones glucagon-like peptide-1, glucagon-like peptide-2, neuropeptide Y and peptide YY with marked kinetic differences compared with dipeptidyl peptidase IV. Inhibition of dipeptidyl peptidases IV, 8 and 9 using the well-known dipeptidyl peptidase IV inhibitor valine pyrrolidide resulted in similar K(i) values, indicating that this inhibitor is non-selective for any of the three dipeptidyl peptidases.


Assuntos
Dipeptidases/química , Dipeptidil Peptidase 4/química , Dipeptidil Peptidases e Tripeptidil Peptidases/química , Sequência de Aminoácidos , Baculoviridae/genética , Baculoviridae/metabolismo , Cromatografia em Gel , Dipeptidases/genética , Dipeptidases/isolamento & purificação , Dipeptidases/metabolismo , Dipeptidil Peptidase 4/metabolismo , Dipeptidil Peptidases e Tripeptidil Peptidases/genética , Dipeptidil Peptidases e Tripeptidil Peptidases/isolamento & purificação , Dipeptidil Peptidases e Tripeptidil Peptidases/metabolismo , Ativação Enzimática , Inibidores Enzimáticos/metabolismo , Inibidores Enzimáticos/farmacologia , Humanos , Cinética , Dados de Sequência Molecular , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/metabolismo , Estrutura Quaternária de Proteína , Pirróis/metabolismo , Pirróis/farmacologia , Proteínas Recombinantes/genética , Proteínas Recombinantes/isolamento & purificação , Proteínas Recombinantes/metabolismo , Alinhamento de Sequência , Especificidade por Substrato , Valina/metabolismo , Valina/farmacologia
14.
Mol Brain ; 10(1): 43, 2017 09 04.
Artigo em Inglês | MEDLINE | ID: mdl-28870203

RESUMO

The neurotransmitter glutamate facilitates neuronal signalling at excitatory synapses. Glutamate is released from the presynaptic membrane into the synaptic cleft. Across the synaptic cleft glutamate binds to both ion channels and metabotropic glutamate receptors at the postsynapse, which expedite downstream signalling in the neuron. The postsynaptic density, a highly specialized matrix, which is attached to the postsynaptic membrane, controls this downstream signalling. The postsynaptic density also resets the synapse after each synaptic firing. It is composed of numerous proteins including a family of Discs large associated protein 1, 2, 3 and 4 (DLGAP1-4) that act as scaffold proteins in the postsynaptic density. They link the glutamate receptors in the postsynaptic membrane to other glutamate receptors, to signalling proteins and to components of the cytoskeleton. With the central localisation in the postsynapse, the DLGAP family seems to play a vital role in synaptic scaling by regulating the turnover of both ionotropic and metabotropic glutamate receptors in response to synaptic activity. DLGAP family has been directly linked to a variety of psychological and neurological disorders. In this review we focus on the direct and indirect role of DLGAP family on schizophrenia as well as other brain diseases.


Assuntos
Encefalopatias/metabolismo , Neurônios/metabolismo , Proteínas Associadas SAP90-PSD95/metabolismo , Sequência de Aminoácidos , Animais , Encéfalo/metabolismo , Encéfalo/patologia , Humanos , Modelos Biológicos , Mapeamento de Interação de Proteínas , Proteínas Associadas SAP90-PSD95/química
15.
Front Neural Circuits ; 11: 111, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29375322

RESUMO

Motor fatigue occurring during prolonged physical activity has both peripheral and central origins. It was previously demonstrated that the excitability of motoneurons was decreased when a spillover of serotonin could activate extrasynaptic 5-HT1A receptors at the axon initial segment (AIS) of motoneurons. Here we investigated the impact of massive synaptic release of serotonin on motor behavior in an integrated preparation of the adult turtle performing fictive scratching behaviors. We found that a prolonged electrical stimulation of the raphe spinal pathway induced a reversible inhibition of the motor behavior that lasted several tens of seconds. The effect disappeared when the spinal cord was perfused with an antagonist for 5-HT1A receptors. By demonstrating a direct impact of serotonin on motor behavior, we suggest a central role of this monoamine behind central fatigue.


Assuntos
Fadiga/metabolismo , Movimento/fisiologia , Núcleos da Rafe/metabolismo , Receptor 5-HT1A de Serotonina/metabolismo , Serotonina/metabolismo , Medula Espinal/metabolismo , Animais , Região CA1 Hipocampal/citologia , Região CA1 Hipocampal/metabolismo , Estimulação Elétrica , Fadiga/tratamento farmacológico , Células HEK293 , Humanos , Técnicas In Vitro , Camundongos da Linhagem 129 , Camundongos Knockout , Movimento/efeitos dos fármacos , Vias Neurais/citologia , Vias Neurais/efeitos dos fármacos , Vias Neurais/metabolismo , Nervos Periféricos/fisiologia , Piperazinas/farmacologia , Piridinas/farmacologia , Receptor 5-HT1A de Serotonina/genética , Reflexo/fisiologia , Antagonistas do Receptor 5-HT1 de Serotonina , Medula Espinal/citologia , Medula Espinal/efeitos dos fármacos , Tartarugas
16.
Biochim Biophys Acta ; 1616(1): 85-94, 2003 Sep 22.
Artigo em Inglês | MEDLINE | ID: mdl-14507422

RESUMO

The Shaker-type voltage-gated potassium channel, Kv1.3, is believed to be restricted in distribution to lymphocytes and neurons. In lymphocytes, this channel has gained intense attention since it has been proven that inhibition of Kv1.3 channels compromise T lymphocyte activation. To investigate possible expression of Kv1.3 channels in other types of tissue, such as epithelia, binding experiments, immunoprecipitation studies and immunohistochemical studies were performed. The double-mutated, radiolabeled peptidyl ligand, (125)I-HgTX(1)-A19Y/Y37F, which selectively binds Kv1.1, Kv1.2, Kv1.3 and Kv1.6 channels, was used to perform binding studies in epithelia isolated from rabbit kidney and colon. The equilibrium dissociation constant for this ligand was found to be in the sub-picomolar range and the maximal receptor concentration (in fM/mg protein) 1.68 for colon and 0.61-0.75 for kidney epithelium. To determine the subtype of Kv1 channels, immunoprecipitation studies with (125)I-HgTX(1)-A19Y/Y37F labeled epithelial membranes were performed with specific antibodies against Kv1.1, Kv1.2, Kv1.3, Kv1.4 or Kv1.6 subunits. These studies demonstrated that Kv1.3 subunits constituted more than 50% of the entire Kv1 subunit population. The precise localization of Kv1.3 subunits in epithelia was determined by immunohistochemical studies.


Assuntos
Membrana Celular/metabolismo , Mucosa Intestinal/metabolismo , Canais de Potássio de Abertura Dependente da Tensão da Membrana , Canais de Potássio/biossíntese , Urotélio/metabolismo , Animais , Membrana Celular/diagnóstico por imagem , Colo/metabolismo , Técnicas de Cultura , Mucosa Intestinal/citologia , Córtex Renal/metabolismo , Medula Renal/metabolismo , Canal de Potássio Kv1.3 , Masculino , Coelhos , Ratos , Ratos Wistar , Venenos de Escorpião , Distribuição Tecidual , Ultrassonografia , Urotélio/citologia
17.
Biochim Biophys Acta ; 1615(1-2): 115-25, 2003 Sep 02.
Artigo em Inglês | MEDLINE | ID: mdl-12948593

RESUMO

Cloned Ca(2+)-activated K(+) channels of intermediate (hIK) or small (rSK3) conductance were expressed in HEK 293 cells, and channel activity was monitored using whole-cell patch clamp. hIK and rSK3 currents already activated by intracellular calcium were further increased by 95% and 125%, respectively, upon exposure of the cells to a 33% decrease in extracellular osmolarity. hIK and rSK3 currents were inhibited by 46% and 32%, respectively, by a 50% increase in extracellular osmolarity. Cell swelling and channel activation were not associated with detectable increases in [Ca(2+)](i), evidenced by population and single-cell measurements. In addition, inhibitors of IK and SK channels significantly reduced the rate of regulatory volume decrease (RVD) in cells expressing these channels. Cell swelling induced a decrease, and cell shrinkage an increase, in net cellular F-actin content. The swelling-induced activation of hIK channels was strongly inhibited by cytochalasin D (CD), in concentrations that caused depolymerization of F-actin filaments, indicating a role for the F-actin cytoskeleton in modulation of hIK by changes in cell volume. In conclusion, hIK and rSK3 channels are activated by cell swelling and inhibited by shrinkage. A role for the F-actin cytoskeleton in the swelling-induced activation of hIK channels is suggested.


Assuntos
Actinas/metabolismo , Citoesqueleto/metabolismo , Canais de Potássio Cálcio-Ativados/metabolismo , Cálcio/metabolismo , Tamanho Celular/fisiologia , Clonagem Molecular , Citocalasina D/farmacologia , Humanos , Rim/citologia , Inibidores da Síntese de Ácido Nucleico/farmacologia , Canais de Potássio Cálcio-Ativados/efeitos dos fármacos , Canais de Potássio Cálcio-Ativados/genética
18.
J Mol Biol ; 318(5): 1265-74, 2002 May 17.
Artigo em Inglês | MEDLINE | ID: mdl-12083516

RESUMO

Alkaline phosphatases are non-specific phosphomonoesterases that are distributed widely in species ranging from bacteria to man. This study has concentrated on the tissue-nonspecific alkaline phosphatase from arctic shrimps (shrimp alkaline phosphatase, SAP). Originating from a cold-active species, SAP is thermolabile and is used widely in vitro, e.g. to dephosphorylate DNA or dNTPs, since it can be inactivated by a short rise in temperature. Since alkaline phosphatases are zinc-containing enzymes, a multiwavelength anomalous dispersion (MAD) experiment was performed on the zinc K edge, which led to the determination of the structure to a resolution of 1.9 A. Anomalous data clearly showed the presence of a zinc triad in the active site, whereas alkaline phosphatases usually contain two zinc and one magnesium ion per monomer. SAP shares the core, an extended beta-sheet flanked by alpha-helices, and a metal triad with the currently known alkaline phosphatase structures (Escherichia coli structures and a human placental structure). Although SAP lacks some features specific for the mammalian enzyme, their backbones are very similar and may therefore be typical for other higher organisms. Furthermore, SAP possesses a striking feature that the other structures lack: surface potential representations show that the enzyme's net charge of -80 is distributed such that the surface is predominantly negatively charged, except for the positively charged active site. The negatively charged substrate must therefore be directed strongly towards the active site. It is generally accepted that optimization of the electrostatics is one of the characteristics related to cold-adaptation. SAP demonstrates this principle very clearly.


Assuntos
Fosfatase Alcalina/química , Decápodes/enzimologia , Animais , Cristalografia por Raios X , Decápodes/química , Humanos , Modelos Moleculares , Conformação Proteica , Temperatura
19.
IUCrJ ; 2(Pt 1): 9-18, 2015 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-25610623

RESUMO

IgG subclass-specific differences in biological function and in vitro stability are often referred to variations in the conformational flexibility, while this flexibility has rarely been characterized. Here, small-angle X-ray scattering data from IgG1, IgG2 and IgG4 antibodies, which were designed with identical variable regions, were thoroughly analysed by the ensemble optimization method. The extended analysis of the optimized ensembles through shape clustering reveals distinct subclass-specific conformational preferences, which provide new insights for understanding the variations in physical/chemical stability and biological function of therapeutic antibodies. Importantly, the way that specific differences in the linker region correlate with the solution structure of intact antibodies is revealed, thereby visualizing future potential for the rational design of antibodies with designated physicochemical properties and tailored effector functions. In addition, this advanced computational approach is applicable to other flexible multi-domain systems and extends the potential for investigating flexibility in solutions of macromolecules by small-angle X-ray scattering.

20.
J Med Chem ; 46(8): 1306-17, 2003 Apr 10.
Artigo em Inglês | MEDLINE | ID: mdl-12672231

RESUMO

A new and improved synthesis of the peroxisome proliferator-activated receptor (PPAR) agonist ragaglitazar applicable for large-scale preparation has been developed. The convergent synthetic procedure was based on a novel enzymatic kinetic resolution step. The conformation of ragaglitazar bound to the hPPARgamma receptor was quite different compared to the single-crystal structures of the l-arginine salt of ragaglitazar. In particular, the phenoxazine ring system had varying orientations. Ragaglitazar had high affinity for the hPPARalpha and -gamma receptors with IC(50) values of 0.98 and 0.092 microM, respectively. The lack of hPPARdelta activity could be explained by the absence of binding in the tail-up pocket in the hPPARdelta receptor, in contrast to the hPPARdelta agonist GW2433, which was able to bind in both the tail-up and tail-down pockets of the receptor.


Assuntos
Hipoglicemiantes/síntese química , Oxazinas/síntese química , Fenilpropionatos/síntese química , Receptores Citoplasmáticos e Nucleares/agonistas , Fatores de Transcrição/agonistas , Sítios de Ligação , Cristalografia por Raios X , Hipoglicemiantes/química , Hipoglicemiantes/farmacologia , Ligantes , Modelos Moleculares , Oxazinas/química , Oxazinas/farmacologia , Fenilpropionatos/química , Fenilpropionatos/farmacologia , Ensaio Radioligante , Estereoisomerismo
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa