Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Nutr Cancer ; 76(5): 452-462, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38494910

RESUMO

Abnormal activation of the Wnt/ß-catenin signaling pathway is a driving force behind the progression of gastric cancer. Atovaquone, known as an antimalarial drug, has emerged as a potential candidate for anti-cancer therapy. This study investigated atovaquone's effects on gastric cancer and its underlying mechanisms. Using gastric cancer cell lines, we found that atovaquone, at concentrations relevant to clinical use, significantly reduced their viability. Notably, atovaquone exhibited a lower effectiveness in reducing the viability of normal gastric cells compared to gastric cancer cells. We further demonstrated that atovaquone inhibited gastric cancer growth and colony formation. Mechanism studies revealed that atovaquone inhibited mitochondrial respiration and induced oxidative stress. Experiments using ρ0 cells, deficient in mitochondrial respiration, indicated a slightly weaker effect of atovaquone on inducing apoptosis compared to wildtype cells. Atovaquone increased phosphorylated ß-catenin at Ser45 and Ser33/37/Thr41, elevated Axin, and reduced ß-catenin. The inhibitory effects of atovaquone on ß-catenin were reversed upon depletion of CK1α. Furthermore, the combination of atovaquone with paclitaxel suppressed gastric cancer growth and improved overall survival in mice. Given that atovaquone is already approved for clinical use, these findings suggest its potential as a valuable addition to the drug arsenal available for treating gastric cancer.


Assuntos
Neoplasias Gástricas , Via de Sinalização Wnt , Animais , Camundongos , Atovaquona/farmacologia , Atovaquona/uso terapêutico , beta Catenina/metabolismo , Neoplasias Gástricas/tratamento farmacológico , Linhagem Celular Tumoral , Caseína Quinases/metabolismo , Proliferação de Células
2.
Cancer Res Commun ; 4(2): 312-327, 2024 02 06.
Artigo em Inglês | MEDLINE | ID: mdl-38265263

RESUMO

Reducing casein kinase 1α (CK1α) expression inhibits the growth of multiple cancer cell lines, making it a potential therapeutic target for cancer. Herein, we evaluated the antitumor activity of FPFT-2216-a novel low molecular weight compound-in lymphoid tumors and elucidated its molecular mechanism of action. In addition, we determined whether targeting CK1α with FPFT-2216 is useful for treating hematopoietic malignancies. FPFT-2216 strongly degraded CK1α and IKAROS family zinc finger 1/3 (IKZF1/3) via proteasomal degradation. FPFT-2216 exhibited stronger inhibitory effects on human lymphoma cell proliferation than known thalidomide derivatives and induced upregulation of p53 and its transcriptional targets, namely, p21 and MDM2. Combining FPFT-2216 with an MDM2 inhibitor exhibited synergistic antiproliferative activity and induced rapid tumor regression in immunodeficient mice subcutaneously transplanted with a human lymphoma cell line. Nearly all tumors in mice disappeared after 10 days; this was continuously observed in 5 of 7 mice up to 24 days after the final FPFT-2216 administration. FPFT-2216 also enhanced the antitumor activity of rituximab and showed antitumor activity in a patient-derived diffuse large B-cell lymphoma xenograft model. Furthermore, FPFT-2216 decreased the activity of the CARD11/BCL10/MALT1 (CBM) complex and inhibited IκBα and NFκB phosphorylation. These effects were mediated through CK1α degradation and were stronger than those of known IKZF1/3 degraders. In conclusion, FPFT-2216 inhibits tumor growth by activating the p53 signaling pathway and inhibiting the CBM complex/NFκB pathway via CK1α degradation. Therefore, FPFT-2216 may represent an effective therapeutic agent for hematopoietic malignancies, such as lymphoma. SIGNIFICANCE: We found potential vulnerability to CK1α degradation in certain lymphoma cells refractory to IKZF1/3 degraders. Targeting CK1α with FPFT-2216 could inhibit the growth of these cells by activating p53 signaling. Our study demonstrates the potential therapeutic application of CK1α degraders, such as FPFT-2216, for treating lymphoma.


Assuntos
Neoplasias Hematológicas , Linfoma Difuso de Grandes Células B , Piperidonas , Triazóis , Humanos , Animais , Camundongos , Proteína Supressora de Tumor p53/metabolismo , Transdução de Sinais , Caseína Quinases/metabolismo , Fator de Transcrição Ikaros/metabolismo
3.
J Med Chem ; 66(11): 7162-7178, 2023 06 08.
Artigo em Inglês | MEDLINE | ID: mdl-37204207

RESUMO

Specific inhibition of a single kinase isoform is a challenging task due to the highly conserved nature of ATP-binding sites. Casein kinase 1 (CK1) δ and ε share 97% sequence identity in their catalytic domains. From a comparison of the X-ray crystal structures of CK1δ and CK1ε, we developed a potent and highly CK1ε-isoform-selective inhibitor (SR-4133). The X-ray co-crystal structure of the CK1δ-SR-4133 complex reveals that the electrostatic surface between the naphthyl unit of SR-4133 and CK1δ is mismatched, destabilizing the interaction of SR-4133 with CK1δ. Conversely, the hydrophobic surface area resulting from the Asp-Phe-Gly motif (DFG)-out conformation of CK1ε stabilizes the binding of SR-4133 in the ATP-binding pocket of CK1ε, leading to the selective inhibition of CK1ε. The potent CK1ε-selective agents display nanomolar growth inhibition of bladder cancer cells and inhibit the phosphorylation of 4E-BP1 in T24 cells, which is a direct downstream effector of CK1ε.


Assuntos
Caseína Quinase Idelta , Caseína Quinases/metabolismo , Isoformas de Proteínas/metabolismo , Sítios de Ligação , Trifosfato de Adenosina
4.
Redox Biol ; 62: 102676, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-36989576

RESUMO

Oxidative stress drives protein S-glutathionylation, which regulates the structure and function of target proteins and is implicated in the pathogenesis of many diseases. Glutaredoxin 1 (Grx1), a cytoplasmic deglutathionylating enzyme, maintains a reducing environment within the cell under various conditions by reversing S-glutathionylation. Grx1 performs a wide range of antioxidant activities in the lens and prevents protein-thiol mixed disulfide accumulation, reducing protein-protein aggregation, insolubilization, and apoptosis of lens epithelial cells. Oxidative stress is related to epithelial-mesenchymal transition (EMT) during posterior capsular opacification (PCO). However, whether Grx1-regulated protein S-glutathionylation plays an essential role in PCO remains unclear. In this study, we revealed that Grx1 expression was decreased in mice following cataract surgery. Furthermore, the absence of Grx1 elevated oxidative stress and protein S-glutathionylation and aggravated EMT in both in vitro and in vivo models. Concurrently, these results could be reversed by Grx1 overexpression. Notably, liquid chromatography-tandem mass spectrometry results showed that casein kinase 1α (CK1α) was susceptible to S-glutathionylation under oxidative stress, and CK1α S-glutathionylation (CK1α-SSG) was mediated at Cys249. The absence of Grx1 upregulated CK1α-SSG, subsequently decreasing the CK1α-induced phosphorylation of ß-catenin at Ser45. The consequential downregulation of degradative ß-catenin and upregulation of its nuclear translocation activated the Wnt/ß-catenin signaling pathway and aggravated EMT. In conclusion, the downregulated expression of Grx1 in mice following cataract surgery aggravated EMT by upregulating the extent of CK1α-SSG. To the best of our knowledge, our study is the first to report how S-glutathionylation regulates CK1α activity under oxidative stress.


Assuntos
Catarata , Transição Epitelial-Mesenquimal , Glutationa , Animais , Camundongos , beta Catenina/metabolismo , Caseína Quinases/metabolismo , Catarata/genética , Catarata/metabolismo , Células Epiteliais/metabolismo , Glutarredoxinas/genética , Glutarredoxinas/metabolismo , Glutationa/metabolismo , Proteína S/metabolismo
5.
Comput Biol Med ; 149: 106006, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-36027865

RESUMO

Hypoxia-inducible factor 1 (HIF-1), a transcriptional activator that mediates cellular responses to hypoxic stress, is essential for tumor progression. It is a heterodimer comprising HIF1α and HIF1ß, with multiple interfaces among their PAS-A, PAS-B, and bHLH domains. HIF1ß is also known as aryl hydrocarbon receptor nuclear translocator (ARNT). Casein kinase 1δ-dependent phosphorylation of the solvent-front residue S247 on the HIF1α PAS-B domain interrupts HIF1α-ARNT complex formation and reduces HIF-1 transcription activity. However, S247 is involved in neither HIF1α-ARNT complex formation nor stabilization of the relative orientation between the HIF1α PAS-A and PAS-B domains. To uncover the underlying allosteric mechanism, we conducted Gaussian accelerated molecular dynamics simulations and identified two distinct conformations of the pS247-carrying HIF1α PAS-B domain: H291-in and H291-out. The H291-in structure can associate with the HIF1α PAS-A domain and form a V-shaped pouch to accommodate the ARNT PAS-A domain, but it cannot associate with the ARNT PAS-B domain. By contrast, the H291-out structure can bind to the ARNT PAS-B domain, but its association with the HIF1α PAS-A domain leads to an unsuitable relative orientation to accommodate the ARNT PAS-A domain. Both conformations were also collected in parallel simulations of the unphosphorylated PAS-B domain. Both structures manage to associate with the ARNT PAS-B and HIF1α PAS-A domains; thus, they are adequate for HIF1α-ARNT complex formation. The domain-domain contact pattern in a phosphorylated variant is shuffled by an order-to-disorder structural switch, triggered by the newly formed K251-pS247 interaction.


Assuntos
Translocador Nuclear Receptor Aril Hidrocarboneto , Subunidade alfa do Fator 1 Induzível por Hipóxia , Translocador Nuclear Receptor Aril Hidrocarboneto/química , Translocador Nuclear Receptor Aril Hidrocarboneto/genética , Translocador Nuclear Receptor Aril Hidrocarboneto/metabolismo , Caseína Quinases/metabolismo , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Fosforilação , Solventes
6.
J Clin Lab Anal ; 36(7): e24532, 2022 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-35698305

RESUMO

BACKGROUND: Circular RNAs (circRNAs) play important roles in various malignancies, such as colorectal cancer (CRC). However, the function of hsa_circ_0001550 in CRC remains to be elucidated. METHODS: The expression levels of hsa_circ_0001550, microRNA (miR)-4262, and nuclear casein kinase and cyclin-dependent kinase substrate 1 (NUCKS1) were determined by real-time qPCR. Cell biological behaviors were evaluated via colony formation assay, transwell assay, flow cytometry, and sphere formation assays. The target relationship was validated via dual-luciferase reporter and RNA pull-down assays. Protein expression was analyzed by western blot. Xenograft tumor model was adopted to evaluate hsa_circ_0001550 function in vivo. RESULTS: Hsa_circ_0001550 enrichment was enhanced in CRC tissue specimens and cell lines. Hsa_circ_0001550 absence hindered CRC cell proliferation, metastasis, stemness, and caused apoptosis. Hsa_circ_0001550 targeted miR-4262, and hsa_circ_0001550 absence-caused impacts were diminished by anti-miR-4262. MiR-4262 targeted NUCKS1. Hsa_circ_0001550 had positive regulation on NUCKS1 expression. NUCKS1 overexpression overturned the influences of hsa_circ_0001550 silencingon CRC cell progression. Hsa_circ_0001550 interference notably blocked in vivo xenograft tumor growth. CONCLUSION: Hsa_circ_0001550 facilitated CRC progression by binding to miR-4262 to positively regulate NUCKS1 abundance.


Assuntos
Neoplasias Colorretais , MicroRNAs , Caseína Quinases/genética , Caseína Quinases/metabolismo , Linhagem Celular Tumoral , Proliferação de Células/genética , Neoplasias Colorretais/patologia , Quinases Ciclina-Dependentes/metabolismo , Regulação Neoplásica da Expressão Gênica , Humanos , MicroRNAs/genética , MicroRNAs/metabolismo
7.
Elife ; 102021 09 29.
Artigo em Inglês | MEDLINE | ID: mdl-34586063

RESUMO

Lung epithelial progenitors differentiate into alveolar type 1 (AT1) and type 2 (AT2) cells. These cells form the air-blood interface and secrete surfactant, respectively, and are essential for lung maturation and function. Current protocols to derive and culture alveolar cells do not faithfully recapitulate the architecture of the distal lung, which influences cell fate patterns in vivo. Here, we report serum-free conditions that allow for growth and differentiation of mouse distal lung epithelial progenitors. We find that Collagen I promotes the differentiation of flattened, polarized AT1 cells. Using these organoids, we performed a chemical screen to investigate WNT signaling in epithelial differentiation. We identify an association between Casein Kinase activity and maintenance of an AT2 expression signature; Casein Kinase inhibition leads to an increase in AT1/progenitor cell ratio. These organoids provide a simplified model of alveolar differentiation and constitute a scalable screening platform to identify and analyze cell differentiation mechanisms.


Assuntos
Diferenciação Celular , Alvéolos Pulmonares/citologia , Células-Tronco/citologia , Animais , Caseína Quinases/antagonistas & inibidores , Caseína Quinases/metabolismo , Células Cultivadas , Colágeno Tipo I/metabolismo , Meios de Cultura Livres de Soro , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Marcadores Genéticos , Camundongos , Camundongos Endogâmicos C57BL , Alvéolos Pulmonares/embriologia , Alvéolos Pulmonares/enzimologia , Alvéolos Pulmonares/metabolismo , Transcrição Gênica , Via de Sinalização Wnt
8.
Cell Rep ; 30(5): 1447-1462.e5, 2020 02 04.
Artigo em Inglês | MEDLINE | ID: mdl-32023461

RESUMO

Primary cilium is an antenna-like microtubule-based cellular sensing structure. Abnormal regulation of the dynamic assembly and disassembly cycle of primary cilia is closely related to ciliopathy and cancer. The Wnt signaling pathway plays a major role in embryonic development and tissue homeostasis, and defects in Wnt signaling are associated with a variety of human diseases, including cancer. In this study, we provide direct evidence of Wnt3a-induced primary ciliogenesis, which includes a continuous pathway showing that the stimulation of Wnt3a, a canonical Wnt ligand, promotes the generation of ß-catenin p-S47 epitope by CK1δ, and these events lead to the reorganization of centriolar satellites resulting in primary ciliogenesis. We have also confirmed the application of our findings in MCF-7/ADR cells, a multidrug-resistant tumor cell model. Thus, our data provide a Wnt3a-induced primary ciliogenesis pathway and may provide a clue on how to overcome multidrug resistance in cancer treatment.


Assuntos
Centríolos/metabolismo , Cílios/metabolismo , Organogênese , Proteína Wnt3A/metabolismo , beta Catenina/metabolismo , Sequência de Aminoácidos , Animais , Caseína Quinases/metabolismo , Centrossomo/metabolismo , Resistência a Múltiplos Medicamentos , Resistencia a Medicamentos Antineoplásicos , Epitopos/metabolismo , Células HEK293 , Células HeLa , Humanos , Ligantes , Células MCF-7 , Camundongos , Fosforilação , Fosfosserina/metabolismo , Proteína Wnt3A/química
9.
Mol Oncol ; 12(5): 611-629, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29465811

RESUMO

Canonical and noncanonical Wnt pathways share some common elements but differ in the responses they evoke. Similar to Wnt ligands acting through the canonical pathway, Wnts that activate the noncanonical signaling, such as Wnt5a, promote Disheveled (Dvl) phosphorylation and its binding to the Frizzled (Fz) Wnt receptor complex. The protein kinase CK1ε is required for Dvl/Fz association in both canonical and noncanonical signaling. Here we show that differently to its binding to canonical Wnt receptor complex, CK1ε does not require p120-catenin for the association with the Wnt5a co-receptor Ror2. Wnt5a promotes the formation of the Ror2-Fz complex and enables the activation of Ror2-bound CK1ε by Fz-associated protein phosphatase 2A. Moreover, CK1ε also regulates Ror2 protein levels; CK1ε association stabilizes Ror2, which undergoes lysosomal-dependent degradation in the absence of this kinase. Although p120-catenin is not required for CK1ε association with Ror2, it also participates in this signaling pathway as p120-catenin binds and maintains Ror2 at the plasma membrane; in p120-depleted cells, Ror2 is rapidly internalized through a clathrin-dependent mechanism. Accordingly, downregulation of p120-catenin or CK1ε affects late responses to Wnt5a that are also sensitive to Ror2, such as SIAH2 transcription, cell invasion, or cortical actin polarization. Our results explain how CK1ε is activated by noncanonical Wnt and identify p120-catenin and CK1ε as two critical factors controlling Ror2 function.


Assuntos
Caseína Quinases/metabolismo , Cateninas/metabolismo , Receptores Órfãos Semelhantes a Receptor Tirosina Quinase/metabolismo , Via de Sinalização Wnt , Animais , Endocitose , Células HEK293 , Humanos , Ligantes , Lisossomos/metabolismo , Camundongos , Modelos Biológicos , Fosforilação , Ligação Proteica , delta Catenina
10.
J Hematol Oncol ; 10(1): 157, 2017 10 02.
Artigo em Inglês | MEDLINE | ID: mdl-28969692

RESUMO

Multiple myeloma (MM) is a malignant tumor of transformed plasma cells. MM pathogenesis is a multistep process. This cancer can occur de novo (rarely) or it can develop from monoclonal gammopathy of undetermined significance (most of the cases). MM can be asymptomatic (smoldering myeloma) or clinically active. Malignant plasma cells exploit intrinsic and extrinsic bone marrow microenvironment-derived growth signals. Upregulation of stress-coping pathways is also instrumental to maintain MM cell growth. The phylogenetically related Ser/Thr kinases CSNK1A1 (CK1α) and CSNK2 (CK2) have recently gained a growing importance in hematologic malignancies arising both from precursors and from mature blood cells. In multiple myeloma, CK1α or CK2 sustain oncogenic cascades, such as the PI3K/AKT, JAK/STAT, and NF-κB, as well as propel stress-related signaling that help in coping with different noxae. Data also suggest that these kinases modulate the delivery of growth factors and cytokines from the bone marrow stroma. The "non-oncogene addiction" phenotype generated by the increased activity of CK1α and CK2 in multiple myeloma contributes to malignant plasma cell proliferation and survival and represents an Achilles' heel for the activity of small ATP competitive CK1α or CK2 inhibitors.


Assuntos
Caseína Quinase II/metabolismo , Caseína Quinases/metabolismo , Animais , Linhagem Celular Tumoral , Humanos , Mieloma Múltiplo/mortalidade , Mieloma Múltiplo/patologia , Análise de Sobrevida
11.
Mol Med Rep ; 16(2): 1596-1602, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28586070

RESUMO

The present study investigated the protective effect of diethylcarbamazine in inhibiting nuclear factor (NF)-κB activation in isoproterenol­induced acute myocardial infarction (AMI) rats through the poly ADP ribose polymerase (PARP) pathway. Male albino Wistar rats were injected subcutaneously with isoproterenol (100 mg/kg/day) for 2 days to induce an AMI model. Diethylcarbamazine (50 mg/kg) was administered by gavage for 12 days prior to the isoproterenol-induced AMI. It was noted that diethylcarbamazine significantly inhibited AMI­induced casein kinase and lactate dehydrogenase levels, and reduced the AMI­induced wet heart weight to body weight ratio in AMI rats. Diethylcarbamazine treatment significantly weakened reactive oxygen species production and reduced the levels of tumor necrosis factor (TNF)­α, interleukin­6 and NF­κB/p65 in AMI rats. Western blotting demonstrated that diethylcarbamazine significantly suppressed the AMI­induced inducible nitric oxide synthase (iNOS), transforming growth factor (TGF)­ß1, cyclooxygenase­2 (COX­2) and PARP protein expression in AMI rats. The results demonstrated that the protective effect of diethylcarbamazine inhibited isoproterenol­induced AMI through the suppression of inflammation, iNOS, TGF­ß1, COX­2 and the PARP pathway, and revealed the clinical potential of diethylcarbamazine for therapeutic and clinical applications.


Assuntos
Dietilcarbamazina/antagonistas & inibidores , Dietilcarbamazina/uso terapêutico , Isoproterenol/farmacologia , Infarto do Miocárdio/tratamento farmacológico , NF-kappa B/antagonistas & inibidores , Poli(ADP-Ribose) Polimerases/efeitos dos fármacos , Poli(ADP-Ribose) Polimerases/metabolismo , Animais , Caseína Quinases/metabolismo , Ciclo-Oxigenase 2/metabolismo , Dietilcarbamazina/administração & dosagem , Modelos Animais de Doenças , Proteínas I-kappa B/metabolismo , Interleucina-6/metabolismo , L-Lactato Desidrogenase/metabolismo , Masculino , Infarto do Miocárdio/induzido quimicamente , Infarto do Miocárdio/metabolismo , Óxido Nítrico Sintase Tipo II/metabolismo , Ratos , Ratos Wistar , Espécies Reativas de Oxigênio/metabolismo , Fator de Crescimento Transformador beta1/metabolismo , Fator de Necrose Tumoral alfa/metabolismo
12.
Cell Physiol Biochem ; 39(3): 939-49, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27513568

RESUMO

BACKGROUND/AIMS: The echinocandin antifungal agent caspofungin has been shown to trigger apoptosis of fungal cells. Beyond that, caspofungin is toxic for host mitochondria. Even though lacking mitochondria, erythrocytes may enter apoptosis-like suicidal erythrocyte death or eryptosis, characterized by cell shrinkage and cell membrane scrambling with phosphatidylserine translocation to the erythrocyte surface. Signaling involved in triggering of eryptosis include increase of cytosolic Ca2+ activity ([Ca2+]i), oxidative stress, ceramide, caspase activation and/or activation of p38 kinase, protein kinase C, and casein kinase. The present study explored, whether caspofungin induces eryptosis and, if so, to shed some light on the cellular mechanisms involved. METHODS: Flow cytometry was employed to determine phosphatidylserine exposure at the cell surface from annexin-V-binding, cell volume from forward scatter, [Ca2+]i from Fluo3-fluorescence, ROS formation from DCFDA dependent fluorescence, and ceramide abundance utilizing specific antibodies. Hemolysis was quantified from hemoglobin concentration in the supernatant. RESULTS: A 48 hours exposure of human erythrocytes to caspofungin (≥ 30 µg/ml) significantly increased the percentage of annexin-V-binding cells, significantly decreased forward scatter, significantly enhanced hemolysis, but did not significantly increase Fluo3-fluorescence, DCFDA fluorescence or ceramide abundance. The effect of caspofungin on annexin-V-binding was not significantly blunted by removal of extracellular Ca2+, by inhibition of caspases with pancaspase inhibitor zVAD (10 µM), or by addition of the antioxidant N-acetyl-cysteine (1 mM), p38 kinase inhibitor SB203580 (2 µM) or protein kinase C inhibitor staurosporine (1 µM). The effect of caspofungin on annexin-V-binding was, however, significantly blunted in the presence of casein kinase inhibitor D4476 (10 µM). CONCLUSIONS: Caspofungin triggers cell shrinkage and phospholipid scrambling of the erythrocyte cell membrane, an effect possibly involving activation of casein kinase.


Assuntos
Antifúngicos/farmacologia , Cálcio/metabolismo , Equinocandinas/farmacologia , Eriptose/efeitos dos fármacos , Eritrócitos/efeitos dos fármacos , Lipopeptídeos/farmacologia , Espécies Reativas de Oxigênio/metabolismo , Acetilcisteína/farmacologia , Compostos de Anilina , Anexina A5 , Caseína Quinases/antagonistas & inibidores , Caseína Quinases/genética , Caseína Quinases/metabolismo , Caspases/genética , Caspases/metabolismo , Caspofungina , Células Cultivadas , Ceramidas/metabolismo , Eritrócitos/química , Eritrócitos/citologia , Citometria de Fluxo , Fluoresceínas , Corantes Fluorescentes , Expressão Gênica , Hemólise/efeitos dos fármacos , Humanos , Oligopeptídeos/farmacologia , Estresse Oxidativo , Fosfatidilserinas/metabolismo , Proteína Quinase C/antagonistas & inibidores , Proteína Quinase C/genética , Proteína Quinase C/metabolismo , Inibidores de Proteínas Quinases/farmacologia , Xantenos , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases p38 Ativadas por Mitógeno/genética , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
13.
J Proteomics ; 144: 140-7, 2016 07 20.
Artigo em Inglês | MEDLINE | ID: mdl-27216642

RESUMO

UNLABELLED: Mutations in the proto-oncogene c-KIT (KIT) are found in several cancers, and the site of these mutations differs markedly between cancer types. We used site directed mutagenesis to induce KIT(559), KIT(642) and KIT(816) mutations in primary human melanocytes (PHM) and we investigated the impact of each mutation on KIT function. We studied canonical KIT-signaling pathways by immunoblotting, and we used stable isotope labeling by amino acids in cell culture (SILAC) and kinase prediction models to identify kinases differently activated in respective mutants. We validated our results with the analysis of phosphorylation levels of selected substrates for each kinase. We concluded that CK1 ε and δ are more active in cell clones harboring KIT(559) and KIT(642) mutations, whereas PAK4 is more active in clones with KIT(816) mutation. Our findings might help to develop further therapeutic options for tumors with specific KIT mutations in different domains. BIOLOGICAL SIGNIFICANCE: Different types of cancers harbor mutations in the oncogene KIT. The use of small molecules inhibitors directly targeting KIT had a limited success in the treatment of patients with KIT mutant cancers. Our study describes specific phospho-proteome changes due to different KIT mutations, and provides targets of further therapeutic options.


Assuntos
Melanócitos/química , Mutação , Proteoma/metabolismo , Proteínas Proto-Oncogênicas c-kit/genética , Caseína Quinases/metabolismo , Células Cultivadas , Éxons , Humanos , Melanócitos/metabolismo , Terapia de Alvo Molecular , Neoplasias/genética , Fosfoproteínas/metabolismo , Fosforilação , Proto-Oncogene Mas , Transdução de Sinais , Quinases Ativadas por p21/metabolismo
14.
Neurobiol Dis ; 79: 1-13, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25862939

RESUMO

14-3-3 proteins are key regulators of cell survival. We have previously demonstrated that 14-3-3 levels are decreased in an alpha-synuclein (αsyn) mouse model of Parkinson's disease (PD), and that overexpression of certain 14-3-3 isoforms is protective in several PD models. Here we examine whether changes in 14-3-3 phosphorylation may contribute to the neurodegenerative process in PD. We examine three key 14-3-3 phosphorylation sites that normally regulate 14-3-3 function, including serine 58 (S58), serine 184 (S184), and serine/threonine 232 (S/T232), in several models of PD and in human PD brain. We observed that an increase in S232 phosphorylation is observed in rotenone-treated neuroblastoma cells, in cells overexpressing αsyn, and in human PD brains. Alterations in S58 phosphorylation were less consistent in these models, and we did not observe any phosphorylation changes at S184. Phosphorylation at S232 induced by rotenone is reduced by casein kinase inhibitors, and is not dependent on αsyn. Mutation of the S232 site affected 14-3-3θ's neuroprotective effects against rotenone and 1-methyl-4-phenylpyridinium (MPP(+)), with the S232D mutant lacking any protective effect compared to wildtype or S232A 14-3-3θ. The S232D mutant partially reduced the ability of 14-3-3θ to inhibit Bax activation in response to rotenone. Based on these findings, we propose that phosphorylation of 14-3-3s at serine 232 contributes to the neurodegenerative process in PD.


Assuntos
Proteínas 14-3-3/metabolismo , Doença de Parkinson/metabolismo , Transtornos Parkinsonianos/metabolismo , 1-Metil-4-fenilpiridínio , Proteínas 14-3-3/genética , Animais , Caseína Quinases/antagonistas & inibidores , Caseína Quinases/metabolismo , Linhagem Celular Tumoral , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Células HEK293 , Hipocampo/metabolismo , Humanos , Serina-Treonina Proteína Quinase-2 com Repetições Ricas em Leucina , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Fosforilação/efeitos dos fármacos , Proteínas Serina-Treonina Quinases/metabolismo , Rotenona , Lobo Temporal/metabolismo , alfa-Sinucleína/genética , alfa-Sinucleína/metabolismo , Proteína X Associada a bcl-2/metabolismo
15.
Acta Biomater ; 10(5): 2241-9, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24434535

RESUMO

There is a great need for novel materials for mineralized tissue repair and regeneration. Two examples of such tissue, bone and dentin, are highly organized hierarchical nanocomposites in which mineral and organic phases interface at the molecular level. In contrast, current graft materials are either ceramic powders or physical blends of mineral and organic phases with mechanical properties far inferior to those of their target tissues. The objective of this study was to synthesize composite nanofibrils with highly integrated organic/inorganic phases inspired by the mineralized collagen fibrils of bone and dentin. Utilizing our understanding of bone and dentin biomineralization, we have first designed bioinspired peptides containing 3 Ser-Ser-Asp repeat motifs based on the highly phosphorylated protein, dentin phosphophoryn (DPP), found in dentin and alveolar bone. We demonstrate that up to 80% of serines in the peptide can be phosphorylated by casein kinases. We further tested the ability of these peptides to induce biomimetic calcium phosphate mineralization of collagen fibrils. Our mineralization studies have revealed that in the presence of these phosphorylated peptides, mineralized collagen fibrils structurally similar to the mineralized collagen fibrils of bone and dentin were formed. Our results demonstrate that using phosphorylated DPP-inspired peptides, we can successfully synthesize biomimetic composite nanofibrils with integrated organic and inorganic phases. These results provide the first step in the development of biomimetic nanostructured materials for mineralized tissue repair and regeneration using phosphopeptides.


Assuntos
Osso e Ossos/metabolismo , Nanocompostos/química , Peptídeos/metabolismo , Trifosfato de Adenosina/metabolismo , Sequência de Aminoácidos , Animais , Calcificação Fisiológica , Caseína Quinases/metabolismo , Elétrons , Colágenos Fibrilares/metabolismo , Cinética , Espectrometria de Massas , Dados de Sequência Molecular , Peptídeos/síntese química , Peptídeos/química , Fosforilação , Ratos , Tomografia
16.
Biosystems ; 114(3): 238-44, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24120734

RESUMO

Apoptosis, a type of cell death, is necessary for maintaining tissue homeostasis and removing malignant cells. Interrupted apoptosis process contributes to carcinogenesis, developmental defects, autoimmune diseases and neurological disorders. Due to the complexity of the process, the molecular dynamics and relative interactions of individual proteins responsible for the activation or inhibition of apoptosis should be researched systematically. In this study, we integrate known protein interactions from databases DIP, IntAct, MINT, HPRD and BioGRID by Naïve Bayes classifier. The receiver operation characteristic (ROC) curve with the area under the ROC curve (AUC) of 0.797 indicates it has a good performance in prediction. Then, we predict the global human apoptotic protein interactions network. Within it, we not only identify the already known interactions of caspases (caspase-8/-10, caspase-9, caspase-3/-6/-7) and Bcl-2 family, but also reveal that Bid can interact with casein kinases (CSK21/22/2B, KC1A, KC1E); both of B2LA1 and B2CL2 can interact with Bid, Bax and Bak; caspase-8 interacts with autophagic proteins (MLP3B, MLP3A and LRRk2). Consequently, we make an initial step to develop the web service IntApop that provides an appropriate platform for apoptosis researchers, systems biologists and translational clinician scientists to predict apoptotic protein interactions in human. In addition, the interaction network can be visualized online, making it a widely applicable systems biology tool for apoptosis and cancer researchers.


Assuntos
Proteínas Reguladoras de Apoptose/metabolismo , Apoptose/fisiologia , Mapas de Interação de Proteínas/fisiologia , Software , Biologia de Sistemas/métodos , Área Sob a Curva , Proteína Agonista de Morte Celular de Domínio Interatuante com BH3/metabolismo , Caseína Quinases/metabolismo , Caspases/metabolismo , Bases de Dados de Proteínas , Humanos , Internet , Mapas de Interação de Proteínas/genética , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Curva ROC
17.
BMC Cell Biol ; 14: 47, 2013 Oct 22.
Artigo em Inglês | MEDLINE | ID: mdl-24148232

RESUMO

BACKGROUND: Bone fractures and loss represent significant costs for the public health system and often affect the patients quality of life, therefore, understanding the molecular basis for bone regeneration is essential. Cytokines, such as IL-6, IL-10 and TNFα, secreted by inflammatory cells at the lesion site, at the very beginning of the repair process, act as chemotactic factors for mesenchymal stem cells, which proliferate and differentiate into osteoblasts through the autocrine and paracrine action of bone morphogenetic proteins (BMPs), mainly BMP-2. Although it is known that BMP-2 binds to ActRI/BMPR and activates the SMAD 1/5/8 downstream effectors, little is known about the intracellular mechanisms participating in osteoblastic differentiation. We assessed differences in the phosphorylation status of different cellular proteins upon BMP-2 osteogenic induction of isolated murine skin mesenchymal stem cells using Triplex Stable Isotope Dimethyl Labeling coupled with LC/MS. RESULTS: From 150 µg of starting material, 2,264 proteins were identified and quantified at five different time points, 235 of which are differentially phosphorylated. Kinase motif analysis showed that several substrates display phosphorylation sites for Casein Kinase, p38, CDK and JNK. Gene ontology analysis showed an increase in biological processes related with signaling and differentiation at early time points after BMP2 induction. Moreover, proteins involved in cytoskeleton rearrangement, Wnt and Ras pathways were found to be differentially phosphorylated during all timepoints studied. CONCLUSIONS: Taken together, these data, allow new insights on the intracellular substrates which are phosphorylated early on during differentiation to BMP2-driven osteoblastic differentiation of skin-derived mesenchymal stem cells.


Assuntos
Proteína Morfogenética Óssea 2/genética , Regulação da Expressão Gênica , Células-Tronco Mesenquimais/metabolismo , Osteoblastos/metabolismo , Fosfoproteínas/genética , Pele/metabolismo , Animais , Proteína Morfogenética Óssea 2/metabolismo , Caseína Quinases/genética , Caseína Quinases/metabolismo , Diferenciação Celular , Quinases Ciclina-Dependentes/genética , Quinases Ciclina-Dependentes/metabolismo , MAP Quinase Quinase 4/genética , MAP Quinase Quinase 4/metabolismo , Espectrometria de Massas , Células-Tronco Mesenquimais/citologia , Camundongos , Osteoblastos/citologia , Fosfoproteínas/metabolismo , Fosforilação , Transdução de Sinais , Pele/citologia , Proteínas Wnt/genética , Proteínas Wnt/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/genética , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Proteínas ras/genética , Proteínas ras/metabolismo
18.
Proc Natl Acad Sci U S A ; 110(26): 10574-9, 2013 Jun 25.
Artigo em Inglês | MEDLINE | ID: mdl-23754375

RESUMO

The family with sequence similarity 20 (Fam20) kinases phosphorylate extracellular substrates and play important roles in biomineralization. Fam20C is the Golgi casein kinase that phosphorylates secretory pathway proteins within Ser-x-Glu/pSer motifs. Mutations in Fam20C cause Raine syndrome, an osteosclerotic bone dysplasia. Here we report the crystal structure of the Fam20C ortholog from Caenorhabditis elegans. The nucleotide-free and Mn/ADP-bound structures unveil an atypical protein kinase-like fold and highlight residues critical for activity. The position of the regulatory αC helix and the lack of an activation loop indicate an architecture primed for efficient catalysis. Furthermore, several distinct elements, including the presence of disulfide bonds, suggest that the Fam20 family diverged early in the evolution of the protein kinase superfamily. Our results reinforce the structural diversity of protein kinases and have important implications for patients with disorders of biomineralization.


Assuntos
Proteínas de Caenorhabditis elegans/química , Caseína Quinases/química , Sequência de Aminoácidos , Animais , Caenorhabditis elegans/enzimologia , Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Caseína Quinase I , Caseína Quinases/genética , Caseína Quinases/metabolismo , Cristalografia por Raios X , Proteínas da Matriz Extracelular/química , Proteínas da Matriz Extracelular/genética , Complexo de Golgi/enzimologia , Humanos , Modelos Moleculares , Dados de Sequência Molecular , Homologia de Sequência de Aminoácidos , Especificidade por Substrato
19.
Invest Ophthalmol Vis Sci ; 54(4): 3052-7, 2013 Apr 30.
Artigo em Inglês | MEDLINE | ID: mdl-23557743

RESUMO

PURPOSE: The goal of this study was to determine whether Compound 49b stimulates insulin-like growth factor binding protein-3 (IGFBP-3) activation in retinal endothelial cells (REC) through DNA-dependent protein kinase (DNA-PK). METHODS: REC were grown in a normal glucose (5 mM) or high glucose medium (25 mM). Some cells were transfected with protein kinase A (PKA) siRNA, following treatment with 50 nM Compound 49b, a novel ß-adrenergic receptor agonist. Cell proteins were extracted and analyzed for DNA-PK expression by Western blotting. Additional cells were treated with or without NU7441 (a specific DNA-PK inhibitor) prior to Compound 49b treatment. Cell lysates were processed for IGFBP-3 ELISA analyses and Western blotting to measure casein kinase 2 (CK2). Immunoprecipitation for total and phospho-IGFBP-3, cell proliferation and cell death measurements were done after transfection with the S(156)A IGFBP-3 mutation (key phosphorylation site involved in DNA-PK) plasmid DNA. RESULTS: Compound 49b required DNA-PK to activate IGFBP-3 in REC. IGFBP-3 activation was significantly reduced following treatment with either the DNA-PK inhibitor or following transfection with the IGFBP-3 S(156)A mutant plasmid (P < 0.05). Significant increases in cell death and decreases in cell proliferation were also observed in cells transfected with the IGFBP-3 S(156)A mutant plasmid (P < 0.05). Casein kinase levels were not altered after treatment with NU7741 or Compound 49b. CONCLUSIONS: Our findings suggest Compound 49b induces DNA-PK levels through PKA activity. DNA-PK is required for Compound 49b-induced IGFBP-3 expression, leading to inhibition of REC cell death.


Assuntos
Apoptose/efeitos dos fármacos , Proteína Quinase Ativada por DNA/metabolismo , Células Endoteliais/patologia , Proteína 3 de Ligação a Fator de Crescimento Semelhante à Insulina/metabolismo , Retina/patologia , Agonistas Adrenérgicos beta/farmacologia , Apoptose/fisiologia , Caseína Quinases/metabolismo , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Cromonas/farmacologia , Proteínas Quinases Dependentes de AMP Cíclico/genética , Proteína Quinase Ativada por DNA/antagonistas & inibidores , Glucose/farmacologia , Humanos , Morfolinas/farmacologia , Fosforilação , RNA Interferente Pequeno , Transfecção
20.
J Biol Chem ; 287(42): 35244-35250, 2012 Oct 12.
Artigo em Inglês | MEDLINE | ID: mdl-22927433

RESUMO

The membrane skeleton plays a central role in maintaining the elasticity and stability of the erythrocyte membrane, two biophysical features critical for optimal functioning and survival of red cells. Many constituent proteins of the membrane skeleton are phosphorylated by various kinases, and phosphorylation of ß-spectrin by casein kinase and of protein 4.1R by PKC has been documented to modulate erythrocyte membrane mechanical stability. In this study, we show that activation of endogenous PKA by cAMP decreases membrane mechanical stability and that this effect is mediated primarily by phosphorylation of dematin. Co-sedimentation assay showed that dematin facilitated interaction between spectrin and F-actin, and phosphorylation of dematin by PKA markedly diminished this activity. Quartz crystal microbalance measurement revealed that purified dematin specifically bound the tail region of the spectrin dimer in a saturable manner with a submicromolar affinity. Pulldown assay using recombinant spectrin fragments showed that dematin, but not phospho-dematin, bound to the tail region of the spectrin dimer. These findings imply that dematin contributes to the maintenance of erythrocyte membrane mechanical stability by facilitating spectrin-actin interaction and that phosphorylation of dematin by PKA can modulate these effects. In this study, we have uncovered a novel functional role for dematin in regulating erythrocyte membrane function.


Assuntos
Actinas/metabolismo , Membrana Eritrocítica/metabolismo , Proteínas dos Microfilamentos/metabolismo , Espectrina/metabolismo , Caseína Quinases/metabolismo , AMP Cíclico/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Proteínas do Citoesqueleto/metabolismo , Feminino , Humanos , Masculino , Proteínas de Membrana/metabolismo , Multimerização Proteica/fisiologia , Estrutura Terciária de Proteína
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA