Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 89
Filtrar
1.
Genes Cells ; 28(2): 83-96, 2023 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-36453010

RESUMO

Adhesion GPCRs (aGPCRs) are a subfamily of GPCRs that are involved in cell adhesion, cell proliferation, and cell migration in various tissues. G protein-coupled receptor proteolytic site (GPS) of aGPCR is required to cleave the extracellular domain autocatalytically, generating two fragments; a N-terminal fragment (NTF) and a C-terminal fragment (CTF) containing seven transmembrane structure. NTF can interact with CTF non-covalently after cleavage, however the physiological significance of the cleavage of aGPCR at GPS, and also the interaction between NTF and CTF have not been fully clarified yet. In this study, we first investigated the expression profiles of two aGPCRs, GPR56/ADGRG1, and LPHN1/ADGRL1 in mouse brain, and found that the NTF and CTF of GPR56 independently expressed in different brain region at different developmental stages. Immunoprecipitation of GPR56CTF co-immunoprecipitated LPHN1NTF from mouse brain and HEK293T cells expressing both fragments. Stimulation with LPHN1 ligand, α-Latrotoxin N4C (αLTXN4C), to cells expressing LPHN1NTF and GPR56CTF increased intracellular Ca2+ concentration ([Ca2+ ]i). We also demonstrated that GPR56KO mouse neurons attenuated their Ca2+ response to αLTXN4C. These results suggest the possibility of functional and chimeric complex containing LPHN1NTF and GPR56CTF in neuronal signal transduction.


Assuntos
Neurônios , Receptores Acoplados a Proteínas G , Transdução de Sinais , Animais , Humanos , Camundongos , Adesão Celular , Movimento Celular , Células HEK293 , Neurônios/metabolismo , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo
2.
Int J Mol Sci ; 25(7)2024 Mar 26.
Artigo em Inglês | MEDLINE | ID: mdl-38612503

RESUMO

Chronic myeloid leukemia (CML) is induced by the expression of the fused tyrosine kinase BCR-ABL, which is caused by a chromosomal translocation. BCR-ABL inhibitors have been used to treat CML; however, the acquisition of resistance by CML cells during treatment is a serious issue. We herein demonstrated that BCR-ABL induced the expression of the RNA helicase DDX5 in K562 cells derived from CML patients in a manner that was dependent on its kinase activity, which resulted in cell proliferation and survival. The knockout of DDX5 decreased the expression of BIRC5 (survivin) and activated caspase 3, leading to apoptosis in K562 cells. Similar results were obtained in cells treated with FL118, an inhibitor of DDX5 and a derivative compound of camptothecin (CPT). Furthermore, FL118 potently induced apoptosis not only in Ba/F3 cells expressing BCR-ABL, but also in those expressing the BCR-ABL T315I mutant, which is resistant to BCR-ABL inhibitors. Collectively, these results revealed that DDX5 is a critical therapeutic target in CML and that FL118 is an effective candidate compound for the treatment of BCR-ABL inhibitor-resistant CML.


Assuntos
Indolizinas , Leucemia Mielogênica Crônica BCR-ABL Positiva , Leucemia Mieloide , Humanos , Leucemia Mielogênica Crônica BCR-ABL Positiva/tratamento farmacológico , Leucemia Mielogênica Crônica BCR-ABL Positiva/genética , Benzodioxóis , Inibidores de Proteínas Quinases/farmacologia
3.
Int J Mol Sci ; 25(1)2023 Dec 25.
Artigo em Inglês | MEDLINE | ID: mdl-38203488

RESUMO

According to numerous studies, it has been epidemiologically suggested that habitual coffee intake seems to prevent the onset of neurodegenerative diseases. In this study, we hypothesized that coffee consumption suppresses neuroinflammation, which is closely related to the development of neurodegenerative diseases. Using microglial BV-2 cells, we first found that the inflammatory responses induced by lipopolysaccharide (LPS) stimulation was diminished by both coffee and decaffeinated coffee through the inhibition of an inflammation-related transcription factor, nuclear factor-κB (NF-κB). Pyrocatechol, a component of roasted coffee produced by the thermal decomposition of chlorogenic acid, also exhibited anti-inflammatory activity by inhibiting the LPS-induced activation of NF-κB. Finally, in an inflammation model using mice injected with LPS into the cerebrum, we observed that intake of pyrocatechol as well as coffee decoctions drastically suppressed the accumulation of microglia and the expression of interleukin-6 (IL-6), tumor necrosis factor α (TNFα), CCL2, and CXCL1 in the inflammatory brain. These observations strongly encourage us to hypothesize that the anti-inflammatory activity of pyrocatechol as well as coffee decoction would be useful for the suppression of neurodegeneration and the prevention of the onsets of Alzheimer's (AD) and Perkinson's diseases (PD).


Assuntos
NF-kappa B , Doenças Neurodegenerativas , Animais , Camundongos , Doenças Neuroinflamatórias , Café , Microglia , Lipopolissacarídeos/toxicidade , Inflamação/tratamento farmacológico , Catecóis/farmacologia , Anti-Inflamatórios/farmacologia
4.
Int J Mol Sci ; 23(2)2022 Jan 11.
Artigo em Inglês | MEDLINE | ID: mdl-35054935

RESUMO

In the treatment of breakpoint cluster region-Abelson (BCR-ABL)-positive chronic myeloid leukemia (CML) using BCR-ABL inhibitors, the appearance of a gatekeeper mutation (T315I) in BCR-ABL is a serious issue. Therefore, the development of novel drugs that overcome acquired resistance to BCR-ABL inhibitors by CML cells is required. We previously demonstrated that a bis-pyridinium fullerene derivative (BPF) induced apoptosis in human chronic myeloid leukemia (CML)-derived K562 cells partially through the generation of reactive oxygen species (ROS). We herein show that BPF enhanced the activation of the mitogen-activated protein kinase/extracellular signal-regulated kinase kinase-extracellular signal-regulated kinase (MEK-ERK) pathway in a ROS-independent manner. BPF-induced apoptosis was attenuated by trametinib, suggesting the functional involvement of the MEK-ERK pathway in apoptosis in K562 cells. In addition, the constitutive activation of the MEK-ERK pathway by the enforced expression of the BRAFV600E mutant significantly increased the sensitivity of K562 cells to BPF. These results confirmed for the first time that BPF induces apoptosis in K562 cells through dual pathways-ROS production and the activation of the MEK-ERK pathway. Furthermore, BPF induced cell death in transformed Ba/F3 cells expressing not only BCR-ABL but also T315I mutant through the activation of the MEK-ERK pathway. These results indicate that BPF is as an effective CML drug that overcomes resistance to BCR-ABL inhibitors.


Assuntos
Apoptose/efeitos dos fármacos , Fulerenos/farmacologia , Proteínas de Fusão bcr-abl/genética , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo , Animais , Fulerenos/química , Genes Essenciais , Humanos , Células K562 , Leucemia Mielogênica Crônica BCR-ABL Positiva/genética , Leucemia Mielogênica Crônica BCR-ABL Positiva/metabolismo , Camundongos , Modelos Biológicos , Mutação , Proteínas Proto-Oncogênicas B-raf/genética , Proteínas Proto-Oncogênicas B-raf/metabolismo
5.
Cytokine ; 123: 154753, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31255914

RESUMO

In the majority of myeloproliferative neoplasms (MPNs) patients, a point mutation, V617F has been found in Janus kinase 2 (JAK2) gene, and this JAK2 mutant provoked aberrant signaling pathway. In the current study, we found that suppressor of cytokine signaling proteins 3 (SOCS3) possessed the tumor suppressive activity against the JAK2 V617F mutant-provoked cellular transformation. The knockdown of SOCS3 increased the expression level of the JAK2 V617F mutant, which enhanced the activation of signaling mediators, including signal transducer and activator of transcription 3 and 5 (STAT3, STAT5) and extracellular signal-regulated kinase (ERK), and also increased of the proliferation rate and tumorigenesis activity of Ba/F3 cells expressing the JAK2 V617F mutant and erythropoietin receptor (EpoR). In contrast, the enforced expression of SOCS3 significantly inhibited the JAK2 V617F mutant-induced activation of downstream signaling molecules, cell proliferation, and tumorigenesis by down-regulating the expression level of the JAK2 V617F mutant. SOCS3 interacted with the JAK2V617F mutant through its SH2 domain and was phosphorylated at Tyr-204 and Tyr-221 in its SOCS box by the JAK2V617F mutant. SOCS3 mutants carrying a mutation in the SH2 domain (R71E) and a substitution at Tyr-221 (Y221F) failed to exert inhibitory effects on JAK2V617F mutant-induced cellular transformation and tumorigenesis. Collectively, these results imply that SOCS3 plays a negative role in the JAK2 V617F mutant-induced oncogenic signaling pathway through its SH2 domain and the phosphorylation of Tyr-221 in its SOCS box.


Assuntos
Transformação Celular Neoplásica/metabolismo , Neoplasias Hematológicas/metabolismo , Janus Quinase 2/metabolismo , Mutação de Sentido Incorreto , Transtornos Mieloproliferativos/metabolismo , Proteína 3 Supressora da Sinalização de Citocinas/metabolismo , Substituição de Aminoácidos , Animais , Linhagem Celular , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/patologia , Neoplasias Hematológicas/genética , Neoplasias Hematológicas/patologia , Janus Quinase 2/genética , Camundongos , Transtornos Mieloproliferativos/genética , Transtornos Mieloproliferativos/patologia , Fosforilação/genética , Proteína 3 Supressora da Sinalização de Citocinas/genética
6.
J Immunol ; 199(10): 3614-3622, 2017 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-29021376

RESUMO

The nucleotide-binding oligomerization domain-like receptor family pyrin domain containing 3 (NLRP3) inflammasome is a molecular platform that induces caspase-1 activation and subsequent IL-1ß maturation, and is implicated in inflammatory diseases; however, little is known about the negative regulation of NLRP3 inflammasome activation. In this article, we identified an E3 ligase, Ariadne homolog 2 (ARIH2), as a posttranslational negative regulator of NLRP3 inflammasome activity in macrophages. ARIH2 interacted with NLRP3 via its NACHT domain (aa 220-575) in the NLRP3 inflammasome complex. In particular, we found that while using mutants of ARIH2 and ubiquitin, the really interesting new gene 2 domain of ARIH2 was required for NLRP3 ubiquitination linked through K48 and K63. Deletion of endogenous ARIH2 using CRISPR/Cas9 genome editing inhibited NLRP3 ubiquitination and promoted NLRP3 inflammasome activation, resulting in apoptosis-associated speck-like protein containing a caspase recruitment domain oligomerization, pro-IL-1ß processing, and IL-1ß production. Conversely, ARIH2 overexpression promoted NLRP3 ubiquitination and inhibited NLRP3 inflammasome activation. Our findings reveal a novel mechanism of ubiquitination-dependent negative regulation of the NLRP3 inflammasome by ARIH2 and highlight ARIH2 as a potential therapeutic target for inflammatory diseases.


Assuntos
Inflamassomos/metabolismo , Interleucina-1beta/metabolismo , Macrófagos/imunologia , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Animais , Apoptose , Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas , Células HEK293 , Humanos , Inflamação , Camundongos , Camundongos Endogâmicos C57BL , Mutação/genética , Ligação Proteica , Engenharia de Proteínas , Ubiquitina-Proteína Ligases/genética , Ubiquitinação
7.
J Biol Chem ; 292(5): 1826-1846, 2017 02 03.
Artigo em Inglês | MEDLINE | ID: mdl-27998978

RESUMO

The erythropoietin receptor (EpoR) regulates development of blood cells, and its full activation normally requires the cytokine erythropoietin (Epo). In the case of myeloproliferative neoplasms (MPN), Epo-independent signaling through EpoR can be caused by a point mutation, V617F, in the EpoR-interacting tyrosine kinase Janus kinase 2 (JAK2). In cells expressing the JAK2 V617F mutant, eight tyrosine residues in the intracellular domain of EpoR are phosphorylated, but the functional role of these phosphorylations in oncogenic signaling is incompletely understood. Here, to evaluate the functional consequences of the phosphorylation of these tyrosine residues, we constructed an EpoR-8YF mutant in which we substituted all eight tyrosine residues with phenylalanine. Co-expression of EpoR-8YF with the JAK2 V617F mutant failed to induce cytokine-independent cell proliferation and tumorigenesis, indicating that JAK2-mediated EpoR phosphorylation is the reason for JAK2 V617F mutant-induced oncogenic signaling. An exhaustive mutational analysis of the eight EpoR tyrosine residues indicated that three of these residues, Tyr-343, Tyr-460, and Tyr-464, are required for the JAK2 V617F mutant to exhibit its oncogenic activity. We also showed that phosphorylation at these three residues was necessary for full activation of the transcription factor STAT5, which is a critical downstream factor of JAK2 V617F-induced oncogenic signaling. In contrast, Epo stimulation could moderately stimulate the proliferation of cells expressing wild type JAK2 and EpoR-8YF, suggesting that the requirement of the phosphorylation of these three tyrosine residues seems to be specific for the oncogenic proliferation provoked by V617F mutation. Collectively, these results have revealed that phosphorylation of Tyr-343, Tyr-460, and Tyr-464 in EpoR underlies JAK2 V617F mutant-induced tumorigenesis. We propose that the targeted disruption of this pathway has therapeutic utility for managing MPN.


Assuntos
Transformação Celular Neoplásica/metabolismo , Neoplasias Hematológicas/metabolismo , Janus Quinase 2/metabolismo , Mutação de Sentido Incorreto , Transtornos Mieloproliferativos/metabolismo , Proteínas de Neoplasias/metabolismo , Receptores da Eritropoetina/metabolismo , Transdução de Sinais , Substituição de Aminoácidos , Animais , Linhagem Celular , Transformação Celular Neoplásica/genética , Neoplasias Hematológicas/genética , Humanos , Janus Quinase 2/genética , Camundongos , Transtornos Mieloproliferativos/genética , Proteínas de Neoplasias/genética , Fosforilação , Receptores da Eritropoetina/genética , Fator de Transcrição STAT5/genética , Fator de Transcrição STAT5/metabolismo
8.
Biochem Biophys Res Commun ; 503(3): 2047-2053, 2018 09 10.
Artigo em Inglês | MEDLINE | ID: mdl-30078678

RESUMO

The intracellular molecular transport system is a basic and general cellular mechanism that is regulated by an array of signaling molecules. Sar1 small GTPases are molecules that play a key role in controlling vehicle transport between the endoplasmic reticulum (ER) and Golgi bodies. Like other small GTPases, the activities of Sar1a depend on their guanine-nucleotide-binding states, which are regulated by guanine-nucleotide exchange factors (GEFs) and GTPase-activating proteins (GAPs). Despite the well-known function of mammalian Sar1 in the intracellular transport system, little is known about when and how Sar1 is activated during cell morphological changes. Here we show that the C-terminal, but not the N-terminal, regions of Sec23A and Sec23B, the effector proteins of Sar1a, specifically bind to the active, GTP-bound form of Sar1a. An affinity precipitation (pull-down) assay using a recombinant C-terminal region of Sec23B reveals that Sar1a is activated following differentiation in neuronal cell lines. In neuronal N1E-115 cells, GTP-bound Sar1a is increased when cells elongate neuronal processes. Similar results are observed in morphological differentiation in oligodendroglial FBD-102b cells. Additionally, prolactin regulatory element binding (PREB), the GEF for Sar1 (Sar1 activator), increases the binding ability to the nucleotide-free form of Sar1a when morphological differentiation occurs. Nucleotide-free small GTPases preferentially interact with the cognate, active GEFs. These results provide evidence that using previously unreported pull down assays reveals that Sar1 and PREB are upregulated following the induction of morphological differentiation, suggesting the potential role of signaling through Sar1a during morphological differentiation.


Assuntos
Guanosina Trifosfato/metabolismo , Proteínas Monoméricas de Ligação ao GTP/química , Proteínas Monoméricas de Ligação ao GTP/metabolismo , Animais , Células COS , Células Cultivadas , Chlorocebus aethiops , Proteínas de Ligação a DNA/metabolismo , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Células HEK293 , Humanos , Camundongos , Fatores de Transcrição/metabolismo
9.
Biochem Biophys Res Commun ; 477(4): 712-716, 2016 09 02.
Artigo em Inglês | MEDLINE | ID: mdl-27353377

RESUMO

A diabetes susceptibility gene, immunoglobulin-like domain containing receptor 2 (Ildr2), encodes a transmembrane protein localized to the endoplasmic reticulum membrane that is closely related to hepatic lipid metabolism. The livers of ob/ob mice in which Ildr2 is transiently overexpressed are relieved of hepatic steatosis. However, the molecular mechanisms through which ILDR2 affects these changes in hepatic lipid metabolism remain unknown. This study aimed to identify ILDR2-interacting proteins to further elucidate the molecular mechanisms underlying the role of ILDR2 in lipid homeostasis. We purified ILDR2-containing protein complexes using tandem affinity purification tagging and identified ZNF70, a member of the Kruppel C2H2-type zinc finger protein family, as a novel ILDR2-interacting protein. We demonstrated that ZNF70 interacts with ZFP64 and activates HES1 transcription by binding to the HES1 promoter. In addition, HES1 gene expression is increased in ILDR2-knockdown HepG2 cells, in which ZNF70 is translocated from the cytoplasm to the nucleus, suggesting that ZNF70 migration to the nucleus after dissociating from the ILDR2-ZNF70 complex activates HES1 transcription. These results support a novel link between ILDR2 and HES1 gene expression and suggest that ILDR2 is involved in a novel pathway in hepatic steatosis.


Assuntos
Núcleo Celular/metabolismo , Regulação da Expressão Gênica/fisiologia , Proteínas de Membrana/metabolismo , Transporte Proteico/fisiologia , Fatores de Transcrição HES-1/metabolismo , Dedos de Zinco/fisiologia , Sítios de Ligação , Células HEK293 , Células Hep G2 , Humanos , Ligação Proteica , Transdução de Sinais/fisiologia , Fatores de Transcrição HES-1/química
10.
J Immunol ; 192(9): 4342-51, 2014 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-24696236

RESUMO

Inflammation plays a key role in the pathophysiology of hepatic ischemia-reperfusion (I/R) injury. However, the mechanism by which hepatic I/R induces inflammatory responses remains unclear. Recent evidence indicates that a sterile inflammatory response triggered by I/R is mediated through a multiple-protein complex called the inflammasome. Therefore, we investigated the role of the inflammasome in hepatic I/R injury and found that hepatic I/R stimuli upregulated the inflammasome-component molecule, nucleotide-binding oligomerization domain-like receptor (NLR) family pyrin domain-containing 3 (NLRP3), but not apoptosis-associated speck-like protein containing a caspase recruitment domain (ASC). NLRP3(-/-) mice, but not ASC(-/-) and caspase-1(-/-) mice, had significantly less liver injury after hepatic I/R. NLRP3(-/-) mice showed reduced inflammatory responses, reactive oxygen species production, and apoptosis in I/R liver. Notably, infiltration of neutrophils, but not macrophages, was markedly inhibited in the I/R liver of NLRP3(-/-) mice. Bone marrow transplantation experiments showed that NLRP3 not only in bone marrow-derived cells, but also in non-bone marrow-derived cells contributed to liver injury after I/R. In vitro experiments revealed that keratinocyte-derived chemokine-induced activation of heterotrimeric G proteins was markedly diminished. Furthermore, NLRP3(-/-) neutrophils decreased keratinocyte-derived chemokine-induced concentrations of intracellular calcium elevation, Rac activation, and actin assembly formation, thereby resulting in impaired migration activity. Taken together, NLRP3 regulates chemokine-mediated functions and recruitment of neutrophils, and thereby contributes to hepatic I/R injury independently of inflammasomes. These findings identify a novel role of NLRP3 in the pathophysiology of hepatic I/R injury.


Assuntos
Proteínas de Transporte/imunologia , Fígado/imunologia , Neutrófilos/imunologia , Traumatismo por Reperfusão/imunologia , Animais , Apoptose/imunologia , Western Blotting , Proteínas de Transporte/metabolismo , Quimiotaxia de Leucócito , Citometria de Fluxo , Imuno-Histoquímica , Inflamassomos/imunologia , Fígado/metabolismo , Fígado/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteína 3 que Contém Domínio de Pirina da Família NLR , Neutrófilos/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Traumatismo por Reperfusão/patologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa
11.
J Biol Chem ; 289(49): 33887-903, 2014 Dec 05.
Artigo em Inglês | MEDLINE | ID: mdl-25326380

RESUMO

The mechanism of neurite growth is complicated, involving continuous cytoskeletal rearrangement and vesicular trafficking. Cytohesin-2 is a guanine nucleotide exchange factor for Arf6, an Arf family molecular switch protein, controlling cell morphological changes such as neuritogenesis. Here, we show that cytohesin-2 binds to a protein with a previously unknown function, CCDC120, which contains three coiled-coil domains, and is transported along neurites in differentiating N1E-115 cells. Transfection of the small interfering RNA (siRNA) specific for CCDC120 into cells inhibits neurite growth and Arf6 activation. When neurites start to extend, vesicles containing CCDC120 and cytohesin-2 are transported in an anterograde manner rather than a retrograde one. As neurites continue extension, anterograde vesicle transport decreases. CCDC120 knockdown inhibits cytohesin-2 localization into vesicles containing CCDC120 and diffuses cytohesin-2 in cytoplasmic regions, illustrating that CCDC120 determines cytohesin-2 localization in growing neurites. Reintroduction of the wild type CCDC120 construct into cells transfected with CCDC120 siRNA reverses blunted neurite growth and Arf6 activity, whereas the cytohesin-2-binding CC1 region-deficient CCDC120 construct does not. Thus, cytohesin-2 is transported along neurites by vesicles containing CCDC120, and it mediates neurite growth. These results suggest a mechanism by which guanine nucleotide exchange factor for Arf6 is transported to mediate neurite growth.


Assuntos
Proteínas Ativadoras de GTPase/genética , Proteínas do Tecido Nervoso/genética , Neurogênese/genética , Neurônios/metabolismo , Vesículas Transportadoras/metabolismo , Sequência de Aminoácidos , Sítios de Ligação , Linhagem Celular , Núcleo Celular/metabolismo , Citoplasma/metabolismo , Proteínas Ativadoras de GTPase/metabolismo , Regulação da Expressão Gênica , Humanos , Dados de Sequência Molecular , Proteínas do Tecido Nervoso/metabolismo , Neurônios/citologia , Ligação Proteica , Estrutura Terciária de Proteína , Transporte Proteico , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Transdução de Sinais
12.
Cytokine ; 72(1): 105-8, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25573803

RESUMO

The interleukin-33 (IL-33)-ST2L signaling pathway has been shown to play important roles in the field of immunology, especially as a trigger for allergic reactions such as bronchial asthma. However, coming back to the original finding that the ST2 gene is induced during initiation of the cell cycle of fibroblastic cell lines, the possible functions of the ST2 gene products and their specific ligand, IL-33, in the field of cell growth regulation are still interesting problems to be solved. In this study, we used NIH-3T3 mouse cell line and added IL-33 before and after cell proliferation assay, which revealed the dual function of IL-33. When IL-33 was added to the confluent cells before the start of cell proliferation, it suppressed the cell growth concentration-dependently. On the other hand, if IL-33 was added after the start of cell proliferation, it enhanced the cell growth. The negative effect of IL-33 on cell proliferation is a novel finding and would provide an important clue to the roles of IL-33 and ST2/ST2L in growth regulation.


Assuntos
Proliferação de Células , Interleucinas/farmacologia , Interleucinas/fisiologia , Animais , Apoptose/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Interleucina-33 , Ligantes , Camundongos , Células NIH 3T3 , Proteínas Recombinantes/farmacologia
13.
Biol Pharm Bull ; 38(4): 594-600, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25832639

RESUMO

GPR56 is a member of the adhesion G protein-coupled receptor (GPCR) and is highly expressed in parts of tumor cells. The involvement of GPR56 in tumorigenesis has been reported. We generated agonistic monoclonal antibodies against human GPR56 and analyzed the action and signaling pathway of GPR56. The antibodies inhibited cell migration through the Gq and Rho pathway in human glioma U87-MG cells. Co-immunoprecipitation analysis indicated that the interaction between the GPR56 extracellular domain and transmembrane domain was potentiated by agonistic antibodies. These results demonstrated that functional antibodies are invaluable tools for GPCR research and should open a new avenue for therapeutic treatment of tumors.


Assuntos
Anticorpos Monoclonais/farmacologia , Receptores Acoplados a Proteínas G/metabolismo , Animais , Cálcio/metabolismo , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/metabolismo , Glioma , Humanos , Camundongos Endogâmicos BALB C , Estrutura Terciária de Proteína , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/imunologia , Quinases Associadas a rho/metabolismo
14.
Genes Cells ; 18(12): 1095-106, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24134321

RESUMO

Hyperactivation of Gq signaling causes cardiac hypertrophy, and ß-adrenergic receptor-mediated Gs signaling is attenuated in hypertrophic cardiomyocytes. Here, we found the increase in a global ubiquitination in hypertrophic mouse heart. The activation of Gq signaling resulted in the ubiquitination of Gαs in neonatal rat cardiomyocytes, reduced Gαs expression, and suppressed cAMP response to ß-adrenergic receptor stimulation. Ectopic expression of Gαq induced a similar suppression, which is due to the degradation of Gαs by a ubiquitin-proteasome pathway. Co-expression of Ric-8B, a positive regulator of Gαs, effectively canceled the Gαq-induced ubiquitination of Gαs and recovered the cAMP accumulation. In vitro, Gαq competes for the binding of Gαs to Ric-8B. These data show a new role of Ric-8B in the crosstalk of two distinct G protein signaling pathways, which are possibly involved in a part of mechanisms of chronic heart failure.


Assuntos
Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/metabolismo , Subunidades alfa Gs de Proteínas de Ligação ao GTP/metabolismo , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Miócitos Cardíacos/metabolismo , Ubiquitinação , Animais , Cardiomegalia/metabolismo , Células Cultivadas , AMP Cíclico/metabolismo , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/genética , Células HEK293 , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Células NIH 3T3 , Ratos , Ratos Wistar , Receptores Adrenérgicos alfa 1/metabolismo , Receptores Adrenérgicos beta/metabolismo , Transdução de Sinais
15.
Cell Signal ; 114: 110985, 2024 02.
Artigo em Inglês | MEDLINE | ID: mdl-38000524

RESUMO

Nucleophosmin-anaplastic lymphoma kinase (NPM-ALK), a fusion protein generated by a chromosomal translocation, is a causative gene product of anaplastic large cell lymphoma (ALCL). It induces cell proliferation and tumorigenesis by activating the transcription factor, signal transducer and activator of transcription factor 3 (STAT3). We herein demonstrated that STAT3 underwent acetylation at K685 in a manner that was dependent on the kinase activity of NPM-ALK. To investigate the role of STAT3 acetylation in NPM-ALK-induced oncogenesis, we generated Ba/F3 cells expressing NPM-ALK in which STAT3 was silenced by shRNA, named STAT3-KD cells, and then reconstituted wild-type STAT3 or the STAT3 K685R mutant into these cells. The phosphorylation level of the K685R mutant at Y705 and S727 was significantly higher than that of wild-type STAT3 in STAT3-KD cells. The expression of STAT3 target genes, such as IL-6, Pim1, Pim2, and Socs3, was more strongly induced by the reconstitution of the K685R mutant than wild-type STAT3. In addition, the proliferative ability of STAT3-KD cells reconstituted with the K685R mutant was slightly higher than that of STAT3-KD cells reconstituted with wild-type STAT3. In comparisons with the inoculation of STAT3-KD cells reconstituted with wild-type STAT3, the inoculation of STAT3-KD cells reconstituted with the K685R mutant significantly enhanced tumorigenesis and hepatosplenomegaly in nude mice. Collectively, these results revealed for the first time that the acetylation of STAT3 at K685 attenuated NPM-ALK-induced oncogenesis.


Assuntos
Proteínas de Fusão Oncogênica , Proteínas Tirosina Quinases , Fator de Transcrição STAT3 , Animais , Camundongos , Acetilação , Quinase do Linfoma Anaplásico/metabolismo , Linhagem Celular Tumoral , Transformação Celular Neoplásica , Camundongos Nus , Proteínas Tirosina Quinases/metabolismo , Fator de Transcrição STAT3/metabolismo , Nucleofosmina/metabolismo , Proteínas de Fusão Oncogênica/metabolismo , Humanos
16.
J Neurosci ; 32(37): 12712-25, 2012 Sep 12.
Artigo em Inglês | MEDLINE | ID: mdl-22972995

RESUMO

Axon outgrowth requires plasma membrane expansion, which results from post-Golgi vesicular transport and fusion. However, the molecular mechanisms regulating post-Golgi vesicular trafficking for membrane expansion and axon outgrowth remain unclear. Here, we show that Rab33a expression became upregulated during axon outgrowth of cultured rat hippocampal neurons. Rab33a was preferentially localized to the Golgi apparatus and to synaptophysin-positive vesicles that are transported along the growing axon. Previous studies showed that synaptophysin is localized to post-Golgi vesicles transported by fast axonal transport in developing neurons. Reduction of Rab33a expression by RNAi (RNA interference) inhibited the anterograde transport of synaptophysin-positive vesicles, leading to their decrease in axonal tips. Furthermore, this treatment reduced membrane fusion of synaptophysin-positive vesicles at the growth cones and inhibited axon outgrowth. Overexpression of Rab33a, on the other hand, induced excessive accumulation of synaptophysin-positive vesicles and concurrent formation of surplus axons. These data suggest that Rab33a participates in axon outgrowth by mediating anterograde axonal transport of synaptophysin-positive vesicles and their concomitant fusion at the growth cones.


Assuntos
Transporte Axonal/fisiologia , Axônios/fisiologia , Membrana Celular/fisiologia , Exocitose/fisiologia , Hipocampo/fisiologia , Vesículas Transportadoras/fisiologia , Proteínas rab de Ligação ao GTP/metabolismo , Animais , Axônios/ultraestrutura , Crescimento Celular , Complexo de Golgi/fisiologia , Complexo de Golgi/ultraestrutura , Hipocampo/citologia , Neurônios/citologia , Neurônios/fisiologia , Ratos
17.
J Biol Chem ; 287(16): 12691-702, 2012 Apr 13.
Artigo em Inglês | MEDLINE | ID: mdl-22367209

RESUMO

Doublecortin (DCX) is a microtubule-associated protein that is specifically expressed in neuronal cells. Genetic mutation of DCX causes lissencephaly disease. Although the abnormal cortical lamination in lissencephaly is thought to be attributable to neuronal cell migration defects, the regulatory mechanisms governing interactions between DCX and cytoskeleton in the migration of neuronal progenitor cells remain obscure. In this study we found that the G(s) and protein kinase A (PKA) signal elicited by pituitary adenylate cyclase-activating polypeptide promotes neuronal progenitor cells migration. Stimulation of G(s)-PKA signaling prevented microtubule bundling and induced the dissociation of DCX from microtubules in cells. PKA phosphorylated DCX at Ser-47, and the phospho-mimicking mutant DCX-S47E promoted cell migration. Activation of PKA and DCX-S47E induced lamellipodium formation. Pituitary adenylate cyclase-activating polypeptide and DCX-S47E stimulated the activation of Rac1, and DCX-S47E interacted with Asef2, a guanine nucleotide exchange factor for Rac1. Our data reveal a dual reciprocal role for DCX phosphorylation in the regulation of microtubule and actin dynamics that is indispensable for proper brain lamination.


Assuntos
Citoesqueleto de Actina/fisiologia , Movimento Celular/fisiologia , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Proteínas Associadas aos Microtúbulos/metabolismo , Microtúbulos/fisiologia , Células-Tronco Neurais/citologia , Neuropeptídeos/metabolismo , Animais , Células COS , Córtex Cerebral/citologia , Córtex Cerebral/embriologia , Córtex Cerebral/enzimologia , Chlorocebus aethiops , Proteínas do Domínio Duplacortina , Proteína Duplacortina , Subunidades alfa Gs de Proteínas de Ligação ao GTP/metabolismo , Células HEK293 , Humanos , Camundongos , Células-Tronco Neurais/enzimologia , Neurônios/citologia , Neurônios/enzimologia , Técnicas de Cultura de Órgãos , Fosforilação/fisiologia , Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/metabolismo , Transdução de Sinais/fisiologia , Proteínas rac de Ligação ao GTP/metabolismo , Proteínas rac1 de Ligação ao GTP
18.
Proc Natl Acad Sci U S A ; 107(31): 13666-71, 2010 Aug 03.
Artigo em Inglês | MEDLINE | ID: mdl-20639466

RESUMO

Heterotrimeric GTP-binding proteins (G proteins) transmit extracellular stimuli perceived by G protein-coupled receptors (GPCRs) to intracellular signaling cascades. Hundreds of GPCRs exist in humans and are the targets of a large percentage of the pharmaceutical drugs used today. Because G proteins are regulated by GPCRs, small molecules that directly modulate G proteins have the potential to become therapeutic agents. However, strategies to develop modulators have been hampered by a lack of structural knowledge of targeting sites for specific modulator binding. Here we present the mechanism of action of the cyclic depsipeptide YM-254890, which is a recently discovered Gq-selective inhibitor. YM-254890 specifically inhibits the GDP/GTP exchange reaction of alpha subunit of Gq protein (Galphaq) by inhibiting the GDP release from Galphaq. X-ray crystal structure analysis of the Galphaqbetagamma-YM-254890 complex shows that YM-254890 binds the hydrophobic cleft between two interdomain linkers connecting the GTPase and helical domains of the Galphaq. The binding stabilizes an inactive GDP-bound form through direct interactions with switch I and impairs the linker flexibility. Our studies provide a novel targeting site for the development of small molecules that selectively inhibit each Galpha subunit and an insight into the molecular mechanism of G protein activation.


Assuntos
Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/química , Peptídeos Cíclicos/química , Sequência de Aminoácidos , Cristalografia por Raios X , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/antagonistas & inibidores , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/genética , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/metabolismo , Interações Hidrofóbicas e Hidrofílicas , Modelos Moleculares , Dados de Sequência Molecular , Mutação , Peptídeos Cíclicos/metabolismo , Peptídeos Cíclicos/farmacologia , Ligação Proteica , Estrutura Quaternária de Proteína , Estrutura Terciária de Proteína , Alinhamento de Sequência
19.
Cell Signal ; 102: 110537, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36442590

RESUMO

A point mutation (V617F) in the Janus kinase 2 (JAK2) gene results in the production of disorderly activated tyrosine kinase, which causes myeloproliferative neoplasms (MPN). We herein demonstrated that the RNA helicase DDX5 was highly expressed at the mRNA and protein levels through the activation of signal transducer and activator of transcription 5 (STAT5) in Ba/F3 cells expressing a JAK2V617F mutant and erythropoietin receptor (V617F/EpoR cells) and MPN patient-derived HEL cells. A treatment with the JAK1/2 inhibitor, ruxolitinib and STAT5 inhibitor, pimozide significantly inhibited DDX5 mRNA expression and enhanced the degradation of DDX5 in these cells, suggesting that the JAK2V617F mutant positively regulates DDX5 mRNA expression and DDX5 protein stability by activating STAT5. The knockdown of DDX5 specifically inhibited the activation of mechanistic target of rapamycin (mTOR) in V617F/EpoR cells and HEL cells and significantly suppressed the proliferation of these cells. Furthermore, the knockdown of DDX5 markedly suppressed tumorigenesis, splenomegaly, and liver hypertrophy caused by an inoculation of V617F/EpoR cells in nude mice. Collectively, these results revealed that JAK2V617F exhibits transforming activity by inducing the expression of DDX5 in a STAT5-dependent manner, indicating the potential of the JAK2V617F/STAT5/DDX5 axis as a therapeutic target in the treatment of MPN.


Assuntos
RNA Helicases DEAD-box , Transtornos Mieloproliferativos , Fator de Transcrição STAT5 , Animais , Camundongos , Carcinogênese , Transformação Celular Neoplásica/metabolismo , Janus Quinase 2/metabolismo , Camundongos Nus , Mutação , Transtornos Mieloproliferativos/tratamento farmacológico , Transtornos Mieloproliferativos/genética , Transtornos Mieloproliferativos/metabolismo , Receptores da Eritropoetina/metabolismo , RNA Mensageiro , Fator de Transcrição STAT5/metabolismo , RNA Helicases DEAD-box/metabolismo
20.
FEBS J ; 290(4): 988-1007, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36071319

RESUMO

The expression of CCAAT/enhancer-binding protein (C/EBP) family members and peroxisome proliferator-activated receptor γ (PPAR γ) is essential for the differentiation of pre-adipocyte 3T3-L1 cells into mature adipocytes induced by a combined stimulation with dexamethasone, 3-isobutyl-1-methylxanthine and insulin (DMI). We herein demonstrated that the RNA helicase DDX5, the expression of which was induced by DMI, played an important role in the adipocyte differentiation of 3T3-L1 cells. The DMI-induced accumulation of lipid droplets and expression of adipocyte markers in 3T3-L1 cells were significantly inhibited by the knockdown of DDX5. The knockdown of DDX5 interfered with the expressional induction of C/EBPδ, which was the first to be induced in the transcription factor cascade, and inhibited the subsequent expression of the other transcription factors, C/EBPß, PPARγ and C/EBPα. DDX5 interacted with the glucocorticoid receptor (GR), which induced the expression of C/EBPδ. The knockdown of DDX5 failed to induce the nuclear translocation of GR, suggesting the essential role of DDX5 in the early stage of adipocyte differentiation. Furthermore, the reconstitution of DDX5, but not the DDX5 mutant (K144N) lacking RNA helicase activity, restored DMI-induced GR activation and adipocyte differentiation in 3T3-L1 cells in which DDX5 was knocked down, confirming that the RNA helicase activity of DDX5 is essential for adipogenesis. Collectively, these results revealed for the first time that DDX5 is necessary for GR activation and plays an essential role in early adipocyte differentiation.


Assuntos
Adipócitos , Diferenciação Celular , RNA Helicases DEAD-box , Receptores de Glucocorticoides , Fatores de Transcrição , Animais , Camundongos , Células 3T3-L1 , Adipócitos/metabolismo , Adipogenia/genética , Proteína beta Intensificadora de Ligação a CCAAT/genética , Proteína beta Intensificadora de Ligação a CCAAT/metabolismo , Diferenciação Celular/genética , RNA Helicases DEAD-box/genética , RNA Helicases DEAD-box/metabolismo , Regulação da Expressão Gênica , PPAR gama/genética , PPAR gama/metabolismo , Receptores de Glucocorticoides/genética , Receptores de Glucocorticoides/metabolismo , Fatores de Transcrição/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA