Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
Más filtros













Intervalo de año de publicación
1.
Biochim Biophys Acta Gene Regul Mech ; 1866(1): 194909, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36682583

RESUMEN

Protein kinase M zeta, PKMζ, is a brain enriched kinase with a well characterized role in Long-Term Potentiation (LTP), the activity-dependent strengthening of synapses involved in long-term memory formation. However, little is known about the molecular mechanisms that maintain the tissue specificity of this kinase. Here, we characterized the epigenetic factors, mainly DNA methylation, regulating PKMζ expression in the human brain. The PRKCZ gene has an upstream promoter regulating Protein kinase C ζ (PKCζ), and an internal promoter driving PKMζ expression. A demethylated region, including a canonical CREB binding site, situated at the internal promoter was only observed in human CNS tissues. The induction of site-specific hypermethylation of this region resulted in decreased CREB1 binding and downregulation of PKMζ expression. Noteworthy, CREB binding sites were absent in the upstream promoter of PRKCZ locus, suggesting a specific mechanism for regulating PKMζ expression. These observations were validated using a system of human neuronal differentiation from induced pluripotent stem cells (iPSCs). CREB1 binding at the internal promoter was detected only in differentiated neurons, where PKMζ is expressed. The same epigenetic mechanism in the context of CREB binding site was identified in other genes involved in neuronal differentiation and LTP. Additionally, aberrant DNA hypermethylation at the internal promoter was observed in cases of Alzheimer's disease, correlating with decreased expression of PKMζ in patient brains. Altogether, we present a conserved epigenetic mechanism regulating PKMζ expression and other genes enhanced in the CNS with possible implications in neuronal differentiation and Alzheimer's disease.


Asunto(s)
Enfermedad de Alzheimer , Humanos , Metilación de ADN , Epigénesis Genética , Potenciación a Largo Plazo/fisiología , Encéfalo , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/genética
2.
Methods Mol Biol ; 2599: 21-31, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36427140

RESUMEN

Chromatin immunoprecipitation (ChIP) has been used over three decades to characterize protein-DNA interactions in cells and tissues. The samples are initially cross-linked with formaldehyde and subjected to lysis with detergents followed by sonication to generate smaller fragments of DNA covalently bound to nuclear proteins. Antibodies against the protein of interest are then used to immunoprecipitate the protein-DNA complex, allowing for the purification of the genomic locations bound to the protein of interest. The DNA can then be analyzed by several techniques such as PCR, quantitative PCR, DNA microarrays, and next-generation sequencing (NGS). ChIP can be used in cell culture systems and animal tissues to provide insights into how the identity of cells and tissues is established, and how transcriptional programs can be disrupted or reactivated in disease states.


Asunto(s)
ADN , Secuenciación de Nucleótidos de Alto Rendimiento , Animales , Inmunoprecipitación de Cromatina/métodos , ADN/genética , ADN/metabolismo , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Reacción en Cadena en Tiempo Real de la Polimerasa , Línea Celular
3.
Trends Pharmacol Sci ; 44(1): 11-14, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-35999088

RESUMEN

Odorant receptors (ORs) belong to a large family of G protein-coupled receptors (GPCRs) that are highly expressed by olfactory sensory neurons of the nose. Accumulating evidence indicates that they are also expressed in a variety of nonolfactory tissues, which makes them new potential drug targets. Here we discuss the challenges and strategies to target these receptors.


Asunto(s)
Neuronas Receptoras Olfatorias , Receptores Odorantes , Humanos , Receptores Acoplados a Proteínas G
4.
Sci Signal ; 15(731): eabm6046, 2022 04 26.
Artículo en Inglés | MEDLINE | ID: mdl-35471943

RESUMEN

Chronic pain is a major health issue, and the search for new analgesics has become increasingly important because of the addictive properties and unwanted side effects of opioids. To explore potentially new drug targets, we investigated mutations in the NTRK1 gene found in individuals with congenital insensitivity to pain with anhidrosis (CIPA). NTRK1 encodes tropomyosin receptor kinase A (TrkA), the receptor for nerve growth factor (NGF) and that contributes to nociception. Molecular modeling and biochemical analysis identified mutations that decreased the interaction between TrkA and one of its substrates and signaling effectors, phospholipase Cγ (PLCγ). We developed a cell-permeable phosphopeptide derived from TrkA (TAT-pQYP) that bound the Src homology domain 2 (SH2) of PLCγ. In HEK-293T cells, TAT-pQYP inhibited the binding of heterologously expressed TrkA to PLCγ and decreased NGF-induced, TrkA-mediated PLCγ activation and signaling. In mice, intraplantar administration of TAT-pQYP decreased mechanical sensitivity in an inflammatory pain model, suggesting that targeting this interaction may be analgesic. The findings demonstrate a strategy to identify new targets for pain relief by analyzing the signaling pathways that are perturbed in CIPA.


Asunto(s)
Hipohidrosis , Mutación , Insensibilidad Congénita al Dolor , Fosfolipasa C gamma , Receptor trkA , Analgésicos/farmacología , Animales , Canalopatías/genética , Canalopatías/metabolismo , Células HEK293 , Humanos , Hipohidrosis/genética , Hipohidrosis/metabolismo , Ratones , Factor de Crecimiento Nervioso/genética , Factor de Crecimiento Nervioso/farmacología , Dolor/genética , Dolor/metabolismo , Insensibilidad Congénita al Dolor/genética , Insensibilidad Congénita al Dolor/metabolismo , Fosfolipasa C gamma/genética , Fosfolipasa C gamma/metabolismo , Receptor trkA/genética , Receptor trkA/metabolismo
5.
Front Immunol ; 13: 1010105, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36685521

RESUMEN

Introduction: Considering the likely need for the development of novel effective vaccines adapted to emerging relevant CoV-2 variants, the increasing knowledge of epitope recognition profile among convalescents and afterwards vaccinated with identification of immunodominant regions may provide important information. Methods: We used an RBD peptide microarray to identify IgG and IgA binding regions in serum of 71 COVID-19 convalescents and 18 vaccinated individuals. Results: We found a set of immunodominant RBD antibody epitopes, each recognized by more than 30% of the tested cohort, that differ among the two different groups and are within conserved regions among betacoronavirus. Of those, only one peptide, P44 (S415-429), recognized by 68% of convalescents, presented IgG and IgA antibody reactivity that positively correlated with nAb titers, suggesting that this is a relevant RBD region and a potential target of IgG/IgA neutralizing activity. Discussion: This peptide is localized within the area of contact with ACE-2 and harbors the mutation hotspot site K417 present in gamma (K417T), beta (K417N), and omicron (K417N) variants of concern. The epitope profile of vaccinated individuals differed from convalescents, with a more diverse repertoire of immunodominant peptides, recognized by more than 30% of the cohort. Noteworthy, immunodominant regions of recognition by vaccinated coincide with mutation sites at Omicron BA.1, an important variant emerging after massive vaccination. Together, our data show that immune pressure induced by dominant antibody responses may favor hotspot mutation sites and the selection of variants capable of evading humoral response.


Asunto(s)
COVID-19 , SARS-CoV-2 , Humanos , SARS-CoV-2/genética , Formación de Anticuerpos , Epítopos Inmunodominantes/genética , Epítopos , Inmunoglobulina A , Mutación , Inmunoglobulina G
7.
J Biol Chem ; 297(3): 101041, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34358560

RESUMEN

SARS-CoV-2 nonstructural protein 3 (Nsp3) contains a macrodomain that is essential for coronavirus pathogenesis and is thus an attractive target for drug development. This macrodomain is thought to counteract the host interferon (IFN) response, an important antiviral signalling cascade, via the reversal of protein ADP-ribosylation, a posttranslational modification catalyzed by host poly(ADP-ribose) polymerases (PARPs). However, the main cellular targets of the coronavirus macrodomain that mediate this effect are currently unknown. Here, we use a robust immunofluorescence-based assay to show that activation of the IFN response induces ADP-ribosylation of host proteins and that ectopic expression of the SARS-CoV-2 Nsp3 macrodomain reverses this modification in human cells. We further demonstrate that this assay can be used to screen for on-target and cell-active macrodomain inhibitors. This IFN-induced ADP-ribosylation is dependent on PARP9 and its binding partner DTX3L, but surprisingly the expression of the Nsp3 macrodomain or the deletion of either PARP9 or DTX3L does not impair IFN signaling or the induction of IFN-responsive genes. Our results suggest that PARP9/DTX3L-dependent ADP-ribosylation is a downstream effector of the host IFN response and that the cellular function of the SARS-CoV-2 Nsp3 macrodomain is to hydrolyze this end product of IFN signaling, rather than to suppress the IFN response itself.


Asunto(s)
ADP-Ribosilación , COVID-19/virología , Interferones/metabolismo , Proteínas de Neoplasias/metabolismo , Poli(ADP-Ribosa) Polimerasas/metabolismo , SARS-CoV-2/metabolismo , Transducción de Señal , Ubiquitina-Proteína Ligasas/metabolismo , Humanos
8.
Front Physiol ; 12: 622987, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33767631

RESUMEN

Upper respiratory viral infections can decrease the sense of smell either by inflammatory restriction of nasal airflow that carries the odorant molecules or through interference in olfactory sensory neuron function. During the coronavirus disease 2019 (COVID-19) pandemic, triggered by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), worldwide reports of severe smell loss (anosmia/hyposmia) revealed a different type of olfactory dysfunction associated with respiratory virus infection. Since self-reported perception of smell is subjective and SARS-CoV-2 exposure is variable in the general population, we aimed to study a population that would be more homogeneously exposed to the virus. Here, we investigated the prevalence of olfactory loss in frontline health professionals diagnosed with COVID-19 in Brazil, one of the major epicenters of the disease. We also analyzed the rate of olfactory function recovery and the particular characteristics of olfactory deficit in this population. A widely disclosed cross-sectional online survey directed to health care workers was developed by a group of researchers to collect data concerning demographic information, general symptoms, otolaryngological symptoms, comorbidities, and COVID-19 test results. Of the 1,376 health professionals who completed the questionnaire, 795 (57.8%) were working directly with COVID-19 patients, either in intensive care units, emergency rooms, wards, outpatient clinics, or other areas. Five-hundred forty-one (39.3%) participants tested positive for SARS-CoV-2, and 509 (37%) were not tested. Prevalence of olfactory dysfunction in COVID-19-positive subjects was 83.9% (454 of 541) compared to 12.9% (42 of 326) of those who tested negative and to 14.9% (76 of 509) of those not tested. Olfactory dysfunction incidence was higher in those working in wards, emergency rooms, and intensive care units compared to professionals in outpatient clinics. In general, remission from olfactory symptoms was frequent by the time of responses. Taste disturbances were present in 74.1% of infected participants and were significantly associated with hyposmia. In conclusion, olfactory dysfunction is highly correlated with exposure to SARS-CoV-2 in health care professionals, and remission rates up to 2 weeks are high.

9.
Commun Biol ; 4(1): 238, 2021 02 22.
Artículo en Inglés | MEDLINE | ID: mdl-33619305

RESUMEN

Antibodies represent powerful tools to examine signal transduction pathways. Here, we present a strategy integrating multiple state-of-the-art methods to produce, validate, and utilize antibodies. Focusing on understudied synaptic proteins, we generated 137 recombinant antibodies. We used yeast display antibody libraries from the B cells of immunized rabbits, followed by FACS sorting under stringent conditions to identify high affinity antibodies. The antibodies were validated by high-throughput functional screening, and genome editing. Next, we explored the temporal dynamics of signaling in single cells. A subset of antibodies targeting opioid receptors were used to examine the effect of treatment with opiates that have played central roles in the worsening of the 'opioid epidemic.' We show that morphine and fentanyl exhibit differential temporal dynamics of receptor phosphorylation. In summary, high-throughput approaches can lead to the identification of antibody-based tools required for an in-depth understanding of the temporal dynamics of opioid signaling.


Asunto(s)
Anticuerpos/farmacología , Especificidad de Anticuerpos , Ensayos Analíticos de Alto Rendimiento , Proteína Quinasa C/antagonistas & inhibidores , Receptores Opioides mu/antagonistas & inhibidores , Sinapsis/efectos de los fármacos , Analgésicos Opioides/farmacología , Animales , Anticuerpos/inmunología , Línea Celular Tumoral , Activación Enzimática , Fentanilo/farmacología , Células HEK293 , Humanos , Masculino , Ratones Endogámicos C57BL , Morfina/farmacología , Fosforilación , Proteína Quinasa C/inmunología , Proteína Quinasa C/metabolismo , Conejos , Receptores Opioides mu/inmunología , Receptores Opioides mu/metabolismo , Transducción de Señal , Sinapsis/inmunología , Sinapsis/metabolismo , Factores de Tiempo
10.
J Neurochem ; 157(4): 930-943, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-32970861

RESUMEN

Olfactory disorders have been increasingly reported in individuals infected with SARS-CoV-2, the virus causing the coronavirus disease 2019 (COVID-19). Losing the sense of smell has a strong impact on the quality of life, since it may lead to malnutrition, weight loss, food poisoning, depression, and exposure to dangerous chemicals. Individuals who suffer from anosmia (inability to smell) also cannot sense the flavor of food, which is a combination of taste and smell. Interestingly, infected individuals have reported sudden loss of smell with no congested nose, as is frequently observed in common colds or other upper respiratory tract infections. These observations suggest that SARS-CoV-2 infection leads to olfactory loss through a distinct mechanism, which is still unclear. This article provides an overview of olfactory loss and the recent findings relating to COVID-19. Possible mechanisms of SARS-CoV-2-induced olfactory loss are also discussed.


Asunto(s)
COVID-19/complicaciones , Trastornos del Olfato/etiología , Virosis/complicaciones , Humanos , Trastornos del Olfato/patología , Neuronas Receptoras Olfatorias/patología
11.
J Biol Chem ; 294(45): 16650-16662, 2019 11 08.
Artículo en Inglés | MEDLINE | ID: mdl-31537645

RESUMEN

Calcium (Ca2+) signaling within the cell nucleus regulates specific cellular events such as gene transcription and cell proliferation. Nuclear and cytosolic Ca2+ levels can be independently regulated, and nuclear translocation of receptor tyrosine kinases (RTKs) is one way to locally activate signaling cascades within the nucleus. Nuclear RTKs, including the epidermal growth factor receptor (EGFR), are important for processes such as transcriptional regulation, DNA-damage repair, and cancer therapy resistance. RTKs can hydrolyze phosphatidylinositol 4,5-bisphosphate (PI(4,5)P2) within the nucleus, leading to Ca2+ release from the nucleoplasmic reticulum by inositol 1,4,5-trisphosphate receptors. PI(4,5)P2 hydrolysis is mediated by phospholipase C (PLC). However, it is unknown which nuclear PLC isoform is triggered by EGFR. Here, using subcellular fractionation, immunoblotting and fluorescence, siRNA-based gene knockdowns, and FRET-based biosensor reporter assays, we investigated the role of PLCδ4 in epidermal growth factor (EGF)-induced nuclear Ca2+ signaling and downstream events. We found that EGF-induced Ca2+ signals are inhibited when translocation of EGFR is impaired. Nuclear Ca2+ signals also were reduced by selectively buffering inositol 1,4,5-trisphosphate (InsP3) within the nucleus. EGF induced hydrolysis of nuclear PI(4,5)P2 by the intranuclear PLCδ4, rather than by PLCγ1. Moreover, protein kinase C, a downstream target of EGF, was active in the nucleus of stimulated cells. Furthermore, PLCδ4 and InsP3 modulated cell cycle progression by regulating the expression of cyclins A and B1. These results provide evidence that EGF-induced nuclear signaling is mediated by nuclear PLCδ4 and suggest new therapeutic targets to modulate the proliferative effects of this growth factor.


Asunto(s)
Señalización del Calcio/efectos de los fármacos , Núcleo Celular/metabolismo , Factor de Crecimiento Epidérmico/farmacología , Fosfolipasa C delta/metabolismo , Línea Celular , Proliferación Celular/efectos de los fármacos , Cadenas Pesadas de Clatrina/antagonistas & inhibidores , Cadenas Pesadas de Clatrina/genética , Cadenas Pesadas de Clatrina/metabolismo , Ciclina A/metabolismo , Ciclina B1/metabolismo , Receptores ErbB/metabolismo , Humanos , Hidrólisis , Inositol 1,4,5-Trifosfato/metabolismo , Fosfatidilinositol 4,5-Difosfato/metabolismo , Fosfolipasa C delta/antagonistas & inhibidores , Fosfolipasa C delta/genética , Fosfolipasa C gamma/antagonistas & inhibidores , Fosfolipasa C gamma/genética , Fosfolipasa C gamma/metabolismo , Proteína Quinasa C/metabolismo , Interferencia de ARN , ARN Interferente Pequeño/metabolismo
12.
Proteomes ; 6(4)2018 Oct 06.
Artículo en Inglés | MEDLINE | ID: mdl-30301203

RESUMEN

It is well accepted that treatment of chronic pain with morphine leads to µ opioid receptor (MOR) desensitization and the development of morphine tolerance. MOR activation by the selective peptide agonist, D-Ala2, N-MePhe4, Gly-ol]-enkephalin(DAMGO), leads to robust G protein receptor kinase activation, ß-arrestin recruitment, and subsequent receptor endocytosis, which does not occur in an activation by morphine. However, MOR activation by morphine induces receptor desensitization, in a Protein kinase C (PKC) dependent manner. PKC inhibitors have been reported to decrease receptor desensitization, reduce opiate tolerance, and increase analgesia. However, the exact role of PKC in these processes is not clearly delineated. The difficulties in establishing a particular role for PKC have been, in part, due to the lack of reagents that allow the selective identification of PKC targets. Recently, we generated a conformation state-specific anti-PKC antibody that preferentially recognizes the active state of this kinase. Using this antibody to selectively isolate PKC substrates and a proteomics strategy to establish the identity of the proteins, we examined the effect of morphine treatment on the PKC targets. We found an enhanced interaction of a number of proteins with active PKC, in the presence of morphine. In this article, we discuss the role of these proteins in PKC-mediated MOR desensitization and analgesia. In addition, we posit a role for some of these proteins in mediating pain by TrKA activation, via the activation of transient receptor potential cation channel subfamily V member 1 (TRPV1). Finally, we discuss how these new PKC interacting proteins and pathways could be targeted for the treatment of pain.

13.
Curr Protoc Chem Biol ; 10(2): e42, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29927112

RESUMEN

The protein kinase C (PKC) family of serine/ threonine kinases has been shown to play active roles as either suppressors or promoters of carcinogenesis in different types of tumors. Using antibodies that preferentially recognize the active conformation of classical PKCs (cPKCs), we have previously shown that in breast cancer samples the expression levels of cPKCs were similar in estrogen receptor-positive (ER+ ) as compared to triple-negative tumors; however, the levels of active cPKCs were different. Determining the activation status of PKCs and other kinases in tumors may thus aid therapeutic decisions. Further, in basic science these tools may be used to understand the spatial and temporal dynamics of PKC signaling under different stimuli and for co-immunoprecipitation studies to detect binding partners and substrates of active cPKCs. In this article, we describe how monoclonal and polyclonal anti-active state PKC antibodies can be obtained using rational approaches to select bona fide epitopes through inspection of the crystal structure of classical PKCs coupled to molecular modeling studies. We believe that this methodology can be used for other kinases and multi-domain enzymes that undergo changes in their conformation upon activation. © 2018 by John Wiley & Sons, Inc.


Asunto(s)
Anticuerpos/química , Anticuerpos/inmunología , Proteína Quinasa C/química , Proteína Quinasa C/inmunología , Animales , Anticuerpos Monoclonales/química , Anticuerpos Monoclonales/inmunología , Dominio Catalítico , Humanos , Conformación Proteica , Proteína Quinasa C/metabolismo
14.
Arch Biochem Biophys ; 621: 38-45, 2017 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-28389297

RESUMEN

The FBXO25 mediates degradation of ELK-1 and thus inhibits transcriptional activation of immediate early genes (iEG). Here we show that FBXO25 regulates yet another node of this signaling pathway, by decreasing MAPK/ERK activity. We show that induction of FBXO25 reduced ERK1/2 phosphorylation independently of MEK1/2. Accordingly, in HAP1 FBXO25 knockout cells (FBXO25KO), we observed that upon PMA treatment ERK1/2 was more active than in parental cells. An increase in cell proliferation under receptor mediated activation of the ERK signaling pathway in FBXO25KO cells was also observed. Taken together we show that FBXO25 functions as a negative regulator of MAPK signaling though the reduction of ERK1/2 activation.


Asunto(s)
Proteínas F-Box/metabolismo , Regulación Enzimológica de la Expresión Génica/fisiología , Sistema de Señalización de MAP Quinasas/fisiología , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Células HEK293 , Humanos , Fosforilación
15.
Int J Parasitol Drugs Drug Resist ; 6(1): 74-84, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-27054066

RESUMEN

Parasitic diseases cause ∼ 500,000 deaths annually and remain a major challenge for therapeutic development. Using a rational design based approach, we developed peptide inhibitors with anti-parasitic activity that were derived from the sequences of parasite scaffold proteins LACK (Leishmania's receptor for activated C-kinase) and TRACK (Trypanosoma receptor for activated C-kinase). We hypothesized that sequences in LACK and TRACK that are conserved in the parasites, but not in the mammalian ortholog, RACK (Receptor for activated C-kinase), may be interaction sites for signaling proteins that are critical for the parasites' viability. One of these peptides exhibited leishmanicidal and trypanocidal activity in culture. Moreover, in infected mice, this peptide was also effective in reducing parasitemia and increasing survival without toxic effects. The identified peptide is a promising new anti-parasitic drug lead, as its unique features may limit toxicity and drug-resistance, thus overcoming central limitations of most anti-parasitic drugs.


Asunto(s)
Leishmania/efectos de los fármacos , Péptidos/síntesis química , Péptidos/farmacología , Proteínas Protozoarias/antagonistas & inhibidores , Receptores de Superficie Celular/antagonistas & inhibidores , Tripanocidas/farmacología , Trypanosoma/efectos de los fármacos , Secuencia de Aminoácidos , Animales , Antígenos de Protozoos/química , Diseño de Fármacos , Leishmania/química , Leishmania/genética , Leishmaniasis/tratamiento farmacológico , Leishmaniasis/parasitología , Ratones , Parasitemia/tratamiento farmacológico , Péptidos/administración & dosificación , Proteínas Protozoarias/química , Receptores de Cinasa C Activada , Receptores de Superficie Celular/química , Alineación de Secuencia , Tripanocidas/administración & dosificación , Tripanocidas/química , Trypanosoma/genética , Tripanosomiasis/tratamiento farmacológico , Tripanosomiasis/parasitología
16.
Sci Signal ; 9(420): re3, 2016 Mar 22.
Artículo en Inglés | MEDLINE | ID: mdl-27016527

RESUMEN

Despite the efforts of pharmaceutical companies to develop specific kinase modulators, few drugs targeting kinases have been completely successful in the clinic. This is primarily due to the conserved nature of kinases, especially in the catalytic domains. Consequently, many currently available inhibitors lack sufficient selectivity for effective clinical application. Kinases phosphorylate their substrates to modulate their activity. One of the important steps in the catalytic reaction of protein phosphorylation is the correct positioning of the target residue within the catalytic site. This positioning is mediated by several regions in the substrate binding site, which is typically a shallow crevice that has critical subpockets that anchor and orient the substrate. The structural characterization of this protein-protein interaction can aid in the elucidation of the roles of distinct kinases in different cellular processes, the identification of substrates, and the development of specific inhibitors. Because the region of the substrate that is recognized by the kinase can be part of a linear consensus motif or a nonlinear motif, advances in technology beyond simple linear sequence scanning for consensus motifs were needed. Cost-effective bioinformatics tools are already frequently used to predict kinase-substrate interactions for linear consensus motifs, and new tools based on the structural data of these interactions improve the accuracy of these predictions and enable the identification of phosphorylation sites within nonlinear motifs. In this Review, we revisit kinase-substrate interactions and discuss the various approaches that can be used to identify them and analyze their binding structures for targeted drug development.


Asunto(s)
Biología Computacional/métodos , Sistemas de Liberación de Medicamentos , Inhibidores de Proteínas Quinasas , Proteínas Quinasas , Secuencias de Aminoácidos , Animales , Biología Computacional/tendencias , Sistemas de Liberación de Medicamentos/métodos , Sistemas de Liberación de Medicamentos/tendencias , Humanos , Inhibidores de Proteínas Quinasas/química , Inhibidores de Proteínas Quinasas/uso terapéutico , Proteínas Quinasas/química , Proteínas Quinasas/metabolismo , Relación Estructura-Actividad , Especificidad por Sustrato
17.
Sci Rep ; 6: 22114, 2016 Feb 25.
Artículo en Inglés | MEDLINE | ID: mdl-26911897

RESUMEN

Protein kinase C (PKC) plays a regulatory role in key pathways in cancer. However, since phosphorylation is a step for classical PKC (cPKC) maturation and does not correlate with activation, there is a lack of tools to detect active PKC in tissue samples. Here, a structure-based rational approach was used to select a peptide to generate an antibody that distinguishes active from inactive cPKC. A peptide conserved in all cPKCs, C2Cat, was chosen since modeling studies based on a crystal structure of PKCß showed that it is localized at the interface between the C2 and catalytic domains of cPKCs in an inactive kinase. Anti-C2Cat recognizes active cPKCs at least two-fold better than inactive kinase in ELISA and immunoprecipitation assays, and detects the temporal dynamics of cPKC activation upon receptor or phorbol stimulation. Furthermore, the antibody is able to detect active PKC in human tissue. Higher levels of active cPKC were observed in the more aggressive triple negative breast cancer tumors as compared to the less aggressive estrogen receptor positive tumors. Thus, this antibody represents a reliable, hitherto unavailable and a valuable tool to study PKC activation in cells and tissues. Similar structure-based rational design strategies can be broadly applied to obtain active-state specific antibodies for other signal transduction molecules.


Asunto(s)
Anticuerpos/metabolismo , Neoplasias de la Mama/metabolismo , Neuroblastoma/metabolismo , Proteína Quinasa C beta/metabolismo , Sitios de Unión/inmunología , Neoplasias de la Mama/inmunología , Neoplasias de la Mama/patología , Carcinogénesis , Línea Celular Tumoral , Activación Enzimática , Femenino , Humanos , Isoenzimas/inmunología , Estadificación de Neoplasias , Neuroblastoma/inmunología , Neuroblastoma/patología , Fragmentos de Péptidos/inmunología , Conformación Proteica , Dominios Proteicos/genética , Proteína Quinasa C beta/genética , Proteína Quinasa C beta/inmunología , Receptores de Estrógenos/metabolismo , Transducción de Señal , Relación Estructura-Actividad
18.
Sci Signal ; 7(350): ra105, 2014 Nov 04.
Artículo en Inglés | MEDLINE | ID: mdl-25372052

RESUMEN

Linear consensus motifs are short contiguous sequences of residues within a protein that can form recognition modules for protein interaction or catalytic modification. Protein kinase specificity and the matching of kinases to substrates have been mostly defined by phosphorylation sites that occur in linear consensus motifs. However, phosphorylation can also occur within sequences that do not match known linear consensus motifs recognized by kinases and within flexible loops. We report the identification of Thr(253) in α-tubulin as a site that is phosphorylated by protein kinase C ßI (PKCßI). Thr(253) is not part of a linear PKC consensus motif. Instead, Thr(253) occurs within a region on the surface of α-tubulin that resembles a PKC phosphorylation site consensus motif formed by basic residues in different parts of the protein, which come together in the folded protein to form the recognition motif for PKCßI. Mutations of these basic residues decreased substrate phosphorylation, confirming the presence of this "structurally formed" consensus motif and its importance for the protein kinase-substrate interaction. Analysis of previously reported protein kinase A (PKA) and PKC substrates identified sites within structurally formed consensus motifs in many substrates of these two kinase families. Thus, the concept of consensus phosphorylation site motif needs to be expanded to include sites within these structurally formed consensus motifs.


Asunto(s)
Fosfotransferasas/química , Secuencias de Aminoácidos , Animales , Catálisis , Bovinos , Proteínas Quinasas Dependientes de AMP Cíclico/química , Proteínas Fluorescentes Verdes/química , Células HEK293 , Células HeLa , Humanos , Lisina/química , Simulación del Acoplamiento Molecular , Mutagénesis Sitio-Dirigida , Mutación , Fosforilación , Pliegue de Proteína , Proteína Quinasa C/química , Treonina/química , Tubulina (Proteína)/química
19.
J Proteomics ; 94: 497-512, 2013 Dec 06.
Artículo en Inglés | MEDLINE | ID: mdl-24332066

RESUMEN

The protein kinase C (PKC) family of serine/threonine kinases participate in embryonic stem cell (ESC) proliferation/self-renewal. A few stimuli that induce ESC proliferation activate several PKC isoenzymes including δPKC, however, the role of this isoenzyme under basal conditions that maintain undifferentiated ESCs remains to be determined. Herewith, we aimed to characterize signaling events that occur in undifferentiated ESCs upon δPKC activation. Using phosphoproteomics and a δPKC specific activator peptide, ψδRACK, it was seen that the majority of proteins whose phosphorylation increased upon δPKC activation participate in cell proliferation. Network analysis of these proteins directly connected δPKC to Raf1 and 14-3-3. Experimental validation studies showed that activation of δPKC increased its binding to 14-3-3, transiently activated ERK1/2 and increased ESC proliferation. Independently inhibiting MEK or PI3 kinase both led to a decrease in proliferation of approximately 50%, but δPKC activation only recovered the effect of PI3 kinase inhibition suggesting that ERK1/2 activation via δPKC is probably a parallel pathway to PI3 kinase and that both pathways are necessary for undifferentiated ESC proliferation. BIOLOGICAL SIGNIFICANCE: The use of embryonic stem cells and induced pluripotent stem cells for regenerative therapies is still a challenge. Understanding the underlying mechanisms that keep these cells proliferating with the ability to differentiate in more than 200 cell types (self-renewal) will aid in the future use of these cells therapeutically. Using a targeted phosphoproteomics study, insights into signaling pathways involved in ESC proliferation can be obtained. Modulating these pathways will aid the obtention of a larger number of self-renewing stem cells and induced pluripotent stem cells that can be used therapeutically.


Asunto(s)
Proliferación Celular/efectos de los fármacos , Células Madre Embrionarias/enzimología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Péptidos/farmacología , Proteína Quinasa C-delta/metabolismo , Proteínas 14-3-3/metabolismo , Línea Celular , Células Madre Embrionarias/citología , Activación Enzimática/efectos de los fármacos , Humanos , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-raf/metabolismo
20.
Circ J ; 76(6): 1476-85, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22453000

RESUMEN

BACKGROUND: Epsilon-protein kinase C (εPKC) protects the heart from ischemic injury. However, the mechanism(s) of εPKC cardioprotection is still unclear. Identification of the εPKC targets may aid in elucidating the εPKC-mediated cardioprotective mechanisms. Previous studies, using εPKC transgenic mice and difference in gel electrophoresis, identified proteins involved in glucose metabolism, the expression of which was modified by εPKC. Those studies were accompanied by metabolomic analysis, suggesting that increased glucose oxidation may be responsible for the cardioprotective effect of εPKC. Whether these εPKC-mediated alterations were because of differences in protein expression or phosphorylation was not determined. METHODS AND RESULTS: In the present study, we used an εPKC -specific activator peptide, ψεRACK, combined with phosphoproteomics, to find εPKC targets, and identified that the proteins whose phosphorylation was altered by selective activation of εPKC were mostly mitochondrial proteins. Analysis of the mitochondrial phosphoproteome led to the identification of 55 spots, corresponding to 37 individual proteins, exclusively phosphorylated, in the presence of ψεRACK. The majority of the proteins identified were involved in glucose and lipid metabolism, components of the respiratory chain as well as mitochondrial heat shock proteins. CONCLUSIONS: The protective effect of εPKC during ischemia involves phosphorylation of several mitochondrial proteins involved in glucose and lipid metabolism and oxidative phosphorylation. Regulation of these metabolic pathways by εPKC phosphorylation may lead to εPKC-mediated cardioprotection induced by ψεRACK.


Asunto(s)
Metabolismo Energético , Mitocondrias Cardíacas/enzimología , Isquemia Miocárdica/enzimología , Miocardio/enzimología , Proteína Quinasa C-epsilon/metabolismo , Animales , Citoprotección , Modelos Animales de Enfermedad , Metabolismo Energético/efectos de los fármacos , Activación Enzimática , Activadores de Enzimas/farmacología , Glucosa/metabolismo , Técnicas In Vitro , Metabolismo de los Lípidos , Isquemia Miocárdica/prevención & control , Oligopéptidos/farmacología , Fosforilación Oxidativa , Perfusión , Fosforilación , Proteómica/métodos , Ratas , Ratas Wistar , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA