Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Exp Neurol ; 335: 113528, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33189730

RESUMO

Ischemic stroke (IS) is one of the most common cerebrovascular diseases worldwide. The aberrant expression of BCL6 has been previously implicated in the pathogenesis of IS. Meanwhile, miR-31 is known as a target of BCL6, and has also been suggested to diminish cell damage by suppressing the PKD1 expression. Expanding on this relationship, the current study set out to investigate whether BCL6 participates in ischemic stroke by targeting PKD1. Firstly, IS models were established in vitro and in vivo. TUNEL staining and MTT assay were performed to examine the apoptosis and cell survival. In addition, qRT-PCR and Western blot analysis were applied to examine the expression patterns of the BCL6/miR-31/PKD1 axis and its downstream pathway. Bioinformatics analysis was used to predict the target of miR-31. It was found that BCL6 over-expression promoted ODG-induced increase of apoptosis and decreased the cell survival and miR-31 expression levels, whereas the opposite effects were noted in vitro and in vivo models of IS that were treated with shBCL6. Furthermore, miR-31 down-regulation blocked the effect of BCL6 on ODG-induced cell injury. It was also verified that miR-31 directly-targets PKD1. Also, OGD induced the PKD1 expression and activation of the JAK2/STAT3 pathway, while down-regulation of PKD1 inhibited the OGD-induced cell injury and JAK2/STAT3 pathway activation. Lastly, down-regulation of BCL6 in brain brought about a significant reduction in the size of cerebral infarction and oxidative stress levels in IS mice. Collectively, our findings suggest that inhibition of BCL6 may attenuate oxidative stress-induced neuronal damage by targeting the miR-31/PKD1 axis.


Assuntos
AVC Isquêmico/tratamento farmacológico , AVC Isquêmico/patologia , MicroRNAs/antagonistas & inibidores , Neurônios/patologia , Estresse Oxidativo/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-bcl-6/antagonistas & inibidores , Transdução de Sinais/efeitos dos fármacos , Canais de Cátion TRPP/antagonistas & inibidores , Animais , Apoptose/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Infarto Cerebral/tratamento farmacológico , Infarto Cerebral/patologia , Biologia Computacional , Glucose/deficiência , Hipóxia/patologia , Janus Quinase 2/antagonistas & inibidores , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fator de Transcrição STAT3/antagonistas & inibidores
2.
PLoS One ; 14(12): e0226145, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31809526

RESUMO

Toll-like receptor (TLR) signaling can contribute to the pathogenesis of arthritis. Disruption of TLR signaling at early stages of arthritis might thereby provide an opportunity to halt the disease progression and ameliorate outcomes. We previously found that Gö6976 inhibits TLR-mediated cytokine production in human and mouse macrophages by inhibiting TLR-dependent activation of protein kinase D1 (PKD1), and that PKD1 is essential for proinflammatory responses mediated by MyD88-dependent TLRs. In this study, we investigated whether PKD1 contributes to TLR-mediated proinflammatory responses in human synovial cells, and whether Gö6976 treatment can suppress the development and progression of type II collagen (CII)-induced arthritis (CIA) in mouse. We found that TLR/IL-1R ligands induced activation of PKD1 in human fibroblast-like synoviocytes (HFLS). TLR/IL-1R-induced expression of cytokines/chemokines was substantially inhibited in Gö6976-treated HFLS and PKD1-knockdown HFLS. In addition, serum levels of anti-CII IgG antibodies, and the incidence and severity of arthritis after CII immunization were significantly reduced in mice treated daily with Gö6976. Synergistic effects of T-cell receptor and TLR, as well as TLR alone, on spleen cell proliferation and cytokine production were significantly inhibited in the presence of Gö6976. Our results suggest a possibility that ameliorating effects of Gö6976 on CIA may be due to its ability to inhibit TLR/IL-1R-activated PKD1, which might play an important role in proinflammatory responses in arthritis, and that PKD1 could be a therapeutic target for inflammatory arthritis.


Assuntos
Artrite Experimental/tratamento farmacológico , Carbazóis/administração & dosagem , Colágeno Tipo II/efeitos adversos , Sinoviócitos/enzimologia , Canais de Cátion TRPP/antagonistas & inibidores , Animais , Artrite Experimental/enzimologia , Artrite Experimental/imunologia , Carbazóis/farmacologia , Células Cultivadas , Humanos , Camundongos , Receptores de Interleucina-1/metabolismo , Sinoviócitos/efeitos dos fármacos , Sinoviócitos/imunologia , Receptores Toll-Like/metabolismo
3.
Biomed Res Int ; 2019: 2582401, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31641668

RESUMO

Autosomal dominant polycystic kidney disease (ADPKD) is associated with a number of cellular defects such as hyperproliferation, apoptosis, and dedifferentiation. Mutations in polycystin-1 (PC1) account for ∼85% of ADPKD. Here, we showed that wild-type (WT) or mutant PC1 composed of the last five transmembrane (TM) domains and the C-terminus (termed PC1-5TMC) inhibits cell proliferation and protein translation, as well as the downstream effectors of mTOR, consistent with previous reports. Knockdown of B56α, a subunit of the protein phosphatase 2A (PP2A) complex, or application of PP2A inhibitor okadaic acid or calyculin A, abolished the inhibitory effect of PC1 and PC1-5TMC on proliferation, indicating that PP2A/B56α mediates the regulation of cell proliferation by PC1. In addition to the phosphorylated S6 and 4EBP1, B56α was also downregulated by PC1 and PC1-5TMC. Furthermore, the downregulation of B56α, which may be mediated by mTOR but not AKT, can account for the dependence of PC1-inhibited proliferation on PP2A.


Assuntos
Proliferação de Células/efeitos dos fármacos , Proteína Fosfatase 2/metabolismo , Canais de Cátion TRPP/antagonistas & inibidores , Animais , Apoptose , Diferenciação Celular , Linhagem Celular , Regulação para Baixo , Técnicas de Silenciamento de Genes , Células HEK293 , Células HeLa , Humanos , Toxinas Marinhas , Mutação , Ácido Okadáico/metabolismo , Oxazóis/metabolismo , Fosforilação , Doenças Renais Policísticas/tratamento farmacológico , Biossíntese de Proteínas/efeitos dos fármacos , Proteína Fosfatase 2/genética , Serina-Treonina Quinases TOR/metabolismo , Canais de Cátion TRPP/genética
4.
PLoS One ; 14(3): e0214053, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30883612

RESUMO

Primary cilia of renal epithelial cells express several members of the transient receptor potential (TRP) class of cation-conducting channel, including TRPC1, TRPM3, TRPM4, TRPP2, and TRPV4. Some cases of autosomal dominant polycystic kidney disease (ADPKD) are caused by defects in TRPP2 (also called polycystin-2, PC2, or PKD2). A large-conductance, TRPP2-dependent channel in renal cilia has been well described, but it is not known whether this channel includes any other protein subunits. To study this question, we investigated the pharmacology of the TRPP2-dependent channel through electrical recordings from the cilia of mIMCD-3 cells, a murine cell line of renal epithelial origin. The pharmacology was found to match that of TRPM3 channels. The ciliary TRPP2-dependent channel is known to be activated by depolarization and by increasing cytoplasmic Ca2+. This activation was greatly enhanced by external pregnenolone sulfate, an agonist of TRPM3 channels. Pregnenolone sulfate did not change the single-channel current-voltage relation. The channels were effectively blocked by isosakuranetin, a specific inhibitor of TRPM3 channels. Both pregnenolone sulfate and isosakuranetin were effective at concentrations as low as 1 µM. Knocking out TRPM3 by CRISPR/Cas9 genome editing eliminated the ciliary channel. Thus the channel is both TRPM3-dependent and TRPP2-dependent, suggesting that it may include both types of subunit. Knocking out TRPM3 did not change the level of TRPP2 protein in the cilia, so it is unlikely that the absence of functional ciliary channels results from a failure of trafficking.


Assuntos
Rim/metabolismo , Canais de Cátion TRPM/metabolismo , Canais de Cátion TRPP/metabolismo , Animais , Sinalização do Cálcio , Linhagem Celular , Cílios/efeitos dos fármacos , Cílios/metabolismo , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Flavonoides/farmacologia , Técnicas de Inativação de Genes , Humanos , Rim/citologia , Camundongos , Pregnenolona/farmacologia , Canais de Cátion TRPM/antagonistas & inibidores , Canais de Cátion TRPM/genética , Canais de Cátion TRPP/antagonistas & inibidores , Canais de Cátion TRPP/genética
5.
Sci Rep ; 8(1): 7192, 2018 05 08.
Artigo em Inglês | MEDLINE | ID: mdl-29740060

RESUMO

Mechanotransduction is crucial for touch sensation, hearing, proprioception, and pain sensing. In C. elegans, male ray neurons have been implicated to be involved in the mechanosensation required for mating behavior. However, whether ray neurons directly sense mechanical stimulation is not yet known, and the underlying molecular mechanisms have not been identified. Using in vivo calcium imaging, we recorded the touch-induced calcium responses in male ray neurons. Our data demonstrated that ray neurons are sensitive to mechanical stimulation in a neurotransmitter-independent manner. PKD-2, a putative sensor component for both mechanosensation and chemosensation in male-specific neurons, was not required for the touch-induced calcium responses in RnB neurons, whereas the TRPV channel OSM-9 shaped the kinetics of the responses. We further showed that RnB-neuron mechanosensation is likely mediated by an amiloride-sensitive DEG/ENaC channel. These observations lay a foundation for better understanding the molecular mechanisms of mechanosensation.


Assuntos
Proteínas de Caenorhabditis elegans/genética , Caenorhabditis elegans/genética , Cálcio/metabolismo , Mecanotransdução Celular , Proteínas de Membrana/genética , Proteínas do Tecido Nervoso/genética , Neurônios/metabolismo , Canais de Cátion TRPV/genética , Amilorida/farmacologia , Animais , Caenorhabditis elegans/efeitos dos fármacos , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/antagonistas & inibidores , Proteínas de Caenorhabditis elegans/metabolismo , Bloqueadores do Canal de Sódio Epitelial/farmacologia , Regulação da Expressão Gênica , Masculino , Proteínas de Membrana/antagonistas & inibidores , Proteínas de Membrana/metabolismo , Imagem Molecular/métodos , Proteínas do Tecido Nervoso/antagonistas & inibidores , Proteínas do Tecido Nervoso/metabolismo , Neurônios/citologia , Neurônios/efeitos dos fármacos , Comportamento Sexual Animal/fisiologia , Canais de Cátion TRPP/antagonistas & inibidores , Canais de Cátion TRPP/genética , Canais de Cátion TRPP/metabolismo , Canais de Cátion TRPV/antagonistas & inibidores , Canais de Cátion TRPV/metabolismo , Tato/efeitos dos fármacos , Tato/fisiologia
6.
J Cell Mol Med ; 21(8): 1619-1635, 2017 08.
Artigo em Inglês | MEDLINE | ID: mdl-28244683

RESUMO

Although translational research into autosomal dominant polycystic kidney disease (ADPKD) and its pathogenesis has made considerable progress, there is presently lack of standardized animal model for preclinical trials. In this study, we developed an orthologous mouse model of human ADPKD by cross-mating Pkd2 conditional-knockout mice (Pkd2f3 ) to Cre transgenic mice in which Cre is driven by a spectrum of kidney-related promoters. By systematically characterizing the mouse model, we found that Pkd2f3/f3 mice with a Cre transgene driven by the mouse villin-1 promoter (Vil-Cre;Pkd2f3/f3 ) develop overt cysts in the kidney, liver and pancreas and die of end-stage renal disease (ESRD) at 4-6 months of age. To determine whether these Vil-Cre;Pkd2f3/f3 mice were suitable for preclinical trials, we treated the mice with the high-dose mammalian target of rapamycin (mTOR) inhibitor rapamycin. High-dose rapamycin significantly increased the lifespan, lowered the cystic index and kidney/body weight ratio and improved renal function in Vil-Cre;Pkd2f3/f3 mice in a time- and dose-dependent manner. In addition, we further found that rapamycin arrested aberrant epithelial-cell proliferation in the ADPKD kidney by down-regulating the cell-cycle-associated cyclin-dependent kinase 1 (CDK1) and cyclins, namely cyclin A, cyclin B, cyclin D1 and cyclin E, demonstrating a direct link between mTOR signalling changes and the polycystin-2 dysfunction in cystogenesis. Our newly developed ADPKD model provides a practical platform for translating in vivo preclinical results into ADPKD therapies. The newly defined molecular mechanism by which rapamycin suppresses proliferation via inhibiting abnormally elevated CDK1 and cyclins offers clues to new molecular targets for ADPKD treatment.


Assuntos
Antibióticos Antineoplásicos/farmacologia , Proteína Quinase CDC2/antagonistas & inibidores , Ciclo Celular/efeitos dos fármacos , Ciclinas/antagonistas & inibidores , Rim Policístico Autossômico Dominante/tratamento farmacológico , Sirolimo/farmacologia , Serina-Treonina Quinases TOR/antagonistas & inibidores , Animais , Proteína Quinase CDC2/genética , Proteína Quinase CDC2/metabolismo , Ciclinas/genética , Ciclinas/metabolismo , Relação Dose-Resposta a Droga , Feminino , Efeito Fundador , Regulação da Expressão Gênica , Humanos , Integrases/genética , Integrases/metabolismo , Rim/efeitos dos fármacos , Rim/metabolismo , Rim/patologia , Masculino , Camundongos , Camundongos Transgênicos , Proteínas dos Microfilamentos/genética , Proteínas dos Microfilamentos/metabolismo , Rim Policístico Autossômico Dominante/genética , Rim Policístico Autossômico Dominante/metabolismo , Rim Policístico Autossômico Dominante/patologia , Regiões Promotoras Genéticas , Transdução de Sinais , Serina-Treonina Quinases TOR/genética , Serina-Treonina Quinases TOR/metabolismo , Canais de Cátion TRPP/antagonistas & inibidores , Canais de Cátion TRPP/genética , Canais de Cátion TRPP/metabolismo
7.
Invert Neurosci ; 17(1): 1, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-28078622

RESUMO

Like other cnidarians, the freshwater organism Hydra is characterized by the possession of cnidocytes (stinging cells). Most cnidocytes are located on hydra tentacles, where they are organized along with sensory cells and ganglion cells into battery complexes. The function of the battery complexes is to integrate multiple types of stimuli for the regulation of cnidocyte discharge. The molecular mechanisms controlling the discharge of cnidocytes are not yet fully understood, but it is known that discharge depends on extracellular Ca2+ and that mechanically induced cnidocyte discharge can be enhanced by the presence of prey extracts and other chemicals. Experiments in this paper show that a PKD2 (polycystin 2) transient receptor potential (TRP) channel is expressed in hydra tentacles and bases. PKD2 (TRPP) channels belong to the TRP channel superfamily and are non-selective Ca2+ channels involved in the transduction of both mechanical and chemical stimuli in other organisms. Non-specific PKD2 channel inhibitors Neo (neomycin) and Gd3+ (gadolinium) inhibit both prey capture and cnidocyte discharge in hydra. The PKD2 activator Trip (triptolide) enhances cnidocyte discharge in both starved and satiated hydra and reduces the inhibition of cnidocyte discharge caused by Neo. PKD1 and 2 proteins are known to act together to transduce mechanical and chemical stimuli; in situ hybridization experiments show that a PKD1 gene is expressed in hydra tentacles and bases, suggesting that polycystins play a direct or indirect role in cnidocyte discharge.


Assuntos
Hydra/citologia , Nematocisto/fisiologia , Órgãos dos Sentidos/metabolismo , Canais de Cátion TRPP/metabolismo , Animais , Cálcio/metabolismo , Cálcio/farmacologia , Bloqueadores dos Canais de Cálcio/farmacologia , Diterpenos/farmacologia , Compostos de Epóxi/farmacologia , Gadolínio/farmacologia , Expressão Gênica/efeitos dos fármacos , Expressão Gênica/fisiologia , Glutationa/farmacologia , Imunossupressores/farmacologia , Modelos Moleculares , Nematocisto/citologia , Neomicina/farmacologia , Fenantrenos/farmacologia , Estimulação Física , Comportamento Predatório/fisiologia , Domínios Proteicos/genética , Domínios Proteicos/fisiologia , Inibidores da Síntese de Proteínas/farmacologia , Canais de Cátion TRPP/antagonistas & inibidores , Canais de Cátion TRPP/química , Canais de Cátion TRPP/genética , Verapamil/farmacologia
8.
PLoS Genet ; 12(6): e1006070, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-27272319

RESUMO

During mammalian development, left-right (L-R) asymmetry is established by a cilia-driven leftward fluid flow within a midline embryonic cavity called the node. This 'nodal flow' is detected by peripherally-located crown cells that each assemble a primary cilium which contain the putative Ca2+ channel PKD2. The interaction of flow and crown cell cilia promotes left side-specific expression of Nodal in the lateral plate mesoderm (LPM). Whilst the PKD2-interacting protein PKD1L1 has also been implicated in L-R patterning, the underlying mechanism by which flow is detected and the genetic relationship between Polycystin function and asymmetric gene expression remains unknown. Here, we characterize a Pkd1l1 mutant line in which Nodal is activated bilaterally, suggesting that PKD1L1 is not required for LPM Nodal pathway activation per se, but rather to restrict Nodal to the left side downstream of nodal flow. Epistasis analysis shows that Pkd1l1 acts as an upstream genetic repressor of Pkd2. This study therefore provides a genetic pathway for the early stages of L-R determination. Moreover, using a system in which cultured cells are supplied artificial flow, we demonstrate that PKD1L1 is sufficient to mediate a Ca2+ signaling response after flow stimulation. Finally, we show that an extracellular PKD domain within PKD1L1 is crucial for PKD1L1 function; as such, destabilizing the domain causes L-R defects in the mouse. Our demonstration that PKD1L1 protein can mediate a response to flow coheres with a mechanosensation model of flow sensation in which the force of fluid flow drives asymmetric gene expression in the embryo.


Assuntos
Padronização Corporal/genética , Cílios/genética , Proteínas de Membrana/genética , Mesoderma/metabolismo , Proteína Nodal/genética , Canais de Cátion TRPP/genética , Animais , Embrião de Mamíferos/citologia , Regulação da Expressão Gênica no Desenvolvimento , Peptídeos e Proteínas de Sinalização Intercelular/genética , Mesoderma/embriologia , Camundongos , Camundongos Endogâmicos C3H , Camundongos Transgênicos , Proteína Nodal/biossíntese , Estrutura Terciária de Proteína , Canais de Cátion TRPP/antagonistas & inibidores
9.
Acta Pharmacol Sin ; 37(1): 13-8, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26725733

RESUMO

TRPP2 (polycystin-2, PC2 or PKD2), encoded by the PKD2 gene, is a non-selective cation channel with a large single channel conductance and high Ca(2+) permeability. In cell membrane, TRPP2, along with polycystin-1, TRPV4 and TRPC1, functions as a mechanotransduction channel. In the endoplasmic reticulum, TRPP2 modulates intracellular Ca(2+) release associated with IP3 receptors and the ryanodine receptors. Noteworthily, TRPP2 is widely expressed in vascular endothelial and smooth muscle cells of all major vascular beds, and contributes to the regulation of vessel function. The mutation of the PKD2 gene is a major cause of autosomal dominant polycystic kidney disease (ADPKD), which is not only a common genetic disease of the kidney but also a systemic disorder associated with abnormalities in the vasculature; cardiovascular complications are the leading cause of mortality and morbidity in ADPKD patients. This review provides an overview of the current knowledge regarding the TRPP2 protein and its possible role in cardiovascular function and related diseases.


Assuntos
Vasos Sanguíneos/fisiologia , Canais de Cátion TRPP/fisiologia , Animais , Pressão Sanguínea/fisiologia , Cálcio/metabolismo , Endotélio Vascular/fisiologia , Homeostase , Humanos , Espaço Intracelular/metabolismo , Músculo Liso Vascular/fisiologia , Mutação , Canais de Cátion TRPP/agonistas , Canais de Cátion TRPP/antagonistas & inibidores , Canais de Cátion TRPP/genética
10.
BMC Cell Biol ; 16: 15, 2015 May 07.
Artigo em Inglês | MEDLINE | ID: mdl-25947155

RESUMO

BACKGROUND: Polycystin-1 (PC-1) is a large plasma membrane receptor, encoded by the PKD1 gene, which is mutated in most cases of Autosomal Dominant Polycystic Kidney Disease (ADPKD). The disease is characterized by renal cysts. The precise function of PC-1 remains elusive, although several studies suggest that it can regulate the cellular shape in response to external stimuli. We and others reported that PC-1 regulates the actin cytoskeleton and cell migration. RESULTS: Here we show that cells over-expressing PC-1 display enhanced adhesion rates to the substrate, while cells lacking PC-1 have a reduced capability to adhere. In search for the mechanism responsible for this new property of PC-1 we found that this receptor is able to regulate the stability of the microtubules, in addition to its capability to regulate the actin cytoskeleton. The two cytoskeletal components are acting in a coordinated fashion. Notably, we uncovered that PC-1 regulation of the microtubule cytoskeleton impacts on the turnover rates of focal adhesions in migrating cells and we link all these properties to the capability of PC-1 to regulate the activation state of Focal Adhesion Kinase (FAK). CONCLUSIONS: In this study we show several new features of the PC-1 receptor in modulating microtubules and adhesion dynamics, which are essential for its capability to regulate migration.


Assuntos
Citoesqueleto de Actina/metabolismo , Adesões Focais/metabolismo , Microtúbulos/metabolismo , Canais de Cátion TRPP/metabolismo , Animais , Adesão Celular , Linhagem Celular , Movimento Celular , Cães , Recuperação de Fluorescência Após Fotodegradação , Proteína-Tirosina Quinases de Adesão Focal/metabolismo , Células Madin Darby de Rim Canino , Camundongos , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Canais de Cátion TRPP/antagonistas & inibidores , Canais de Cátion TRPP/genética
11.
Cell Mol Life Sci ; 72(12): 2415-29, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25650235

RESUMO

Dysfunction of many ciliary proteins has been linked to a list of diseases, from cystic kidney to obesity and from hypertension to mental retardation. We previously proposed that primary cilia are unique communication organelles that function as microsensory compartments that house mechanosensory molecules. Here we report that primary cilia exhibit membrane swellings or ciliary bulbs, which based on their unique ultrastructure and motility, could be mechanically regulated by fluid-shear stress. Together with the ultrastructure analysis of the swelling, which contains monosialodihexosylganglioside (GM3), our results show that ciliary bulb has a distinctive set of functional proteins, including GM3 synthase (GM3S), bicaudal-c1 (Bicc1), and polycystin-2 (PC2). In fact, results from our cilia isolation demonstrated for the first time that GM3S and Bicc1 are members of the primary cilia proteins. Although these proteins are not required for ciliary membrane swelling formation under static condition, fluid-shear stress induced swelling formation is partially modulated by GM3S. We therefore propose that the ciliary bulb exhibits a sensory function within the mechano-ciliary structure. Overall, our studies provided an important step towards understanding the ciliary bulb function and structure.


Assuntos
Membrana Celular/fisiologia , Cílios/fisiologia , Células Epiteliais/metabolismo , Rim/metabolismo , Mecanotransdução Celular/fisiologia , Proteínas de Ligação a RNA/metabolismo , Sialiltransferases/metabolismo , Canais de Cátion TRPP/metabolismo , Animais , Células Epiteliais/citologia , Processamento de Imagem Assistida por Computador , Immunoblotting , Rim/citologia , RNA Interferente Pequeno/genética , Proteínas de Ligação a RNA/antagonistas & inibidores , Proteínas de Ligação a RNA/genética , Sialiltransferases/antagonistas & inibidores , Sialiltransferases/genética , Suínos , Canais de Cátion TRPP/antagonistas & inibidores , Canais de Cátion TRPP/genética
12.
PLoS One ; 9(12): e115146, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25517939

RESUMO

The flower buds of Daphne genkwa Sieb. et Zucc. have been used as a traditional Chinese medicine although their functional mechanisms have not been discovered yet. We have studied the potential effects of the plant extracts on natural killer (NK) cell activation, and isolated an active fraction. Genkwadaphnin (GD-1) displayed a potent efficacy to induce IFN-γ transcription in NK cells with concentration- and time-dependent manners. GD-1 treatment triggered the phosphorylation of PKD1, a member of PKC family, MEK and ERK, resulting in IKK activation to induce IκB degradation, and the nuclear localization of p65, an NF-κB subunit, which regulates IFN-γ transcription. GD-1 effect on IFN-γ production was blocked by the addition of Rottlerin, a PKC inhibitor, CID 755673, a PKD inhibitor, or Bay11-7082, an IKKα inhibitor. The nuclear localization of p65 was also inhibited by the kinase inhibitors. Secreted IFN-γ activates STAT1 phosphorylation as autocrine-loops to sustain its secretion. GD-1 induced the phosphorylation of STAT1 probably through the increase of IFN-γ. STAT1 inhibitor also abrogated the sustained IFN-γ secretion. These results suggest that GD-1 is involved in the activation of PKD1 and/or ERK pathway, which activate NK-κB triggering IFN-γ production. As positive feedback loops, secreted IFN-γ activates STAT1 and elongates its production in NK-92 cells.


Assuntos
Diterpenos/farmacologia , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Interferon gama/genética , Células Matadoras Naturais/efeitos dos fármacos , Linfoma/tratamento farmacológico , NF-kappa B/metabolismo , Fator de Transcrição STAT1/metabolismo , Canais de Cátion TRPP/metabolismo , Apoptose/efeitos dos fármacos , Western Blotting , Proliferação de Células/efeitos dos fármacos , Ensaio de Imunoadsorção Enzimática , Humanos , Interferon gama/metabolismo , Células Matadoras Naturais/metabolismo , Células Matadoras Naturais/patologia , Linfoma/metabolismo , Linfoma/patologia , NF-kappa B/antagonistas & inibidores , NF-kappa B/genética , Fosforilação/efeitos dos fármacos , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fator de Transcrição STAT1/antagonistas & inibidores , Fator de Transcrição STAT1/genética , Canais de Cátion TRPP/antagonistas & inibidores , Canais de Cátion TRPP/genética , Células Tumorais Cultivadas
13.
J Clin Invest ; 124(6): 2736-49, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24789909

RESUMO

Patient bone mineral density (BMD) predicts the likelihood of osteoporotic fracture. While substantial progress has been made toward elucidating the genetic determinants of BMD, our understanding of the factors involved remains incomplete. Here, using a systems genetics approach in the mouse, we predicted that bicaudal C homolog 1 (Bicc1), which encodes an RNA-binding protein, is responsible for a BMD quantitative trait locus (QTL) located on murine chromosome 10. Consistent with this prediction, mice heterozygous for a null allele of Bicc1 had low BMD. We used a coexpression network-based approach to determine how Bicc1 influences BMD. Based on this analysis, we inferred that Bicc1 was involved in osteoblast differentiation and that polycystic kidney disease 2 (Pkd2) was a downstream target of Bicc1. Knock down of Bicc1 and Pkd2 impaired osteoblastogenesis, and Bicc1 deficiency-dependent osteoblast defects were rescued by Pkd2 overexpression. Last, in 2 human BMD genome-wide association (GWAS) meta-analyses, we identified SNPs in BICC1 and PKD2 that were associated with BMD. These results, in both mice and humans, identify Bicc1 as a genetic determinant of osteoblastogenesis and BMD and suggest that it does so by regulating Pkd2 transcript levels.


Assuntos
Densidade Óssea/genética , Proteínas de Transporte/genética , Osteogênese/genética , Proteínas de Ligação a RNA/genética , Animais , Carbolinas , Diferenciação Celular/genética , Feminino , Técnicas de Silenciamento de Genes , Redes Reguladoras de Genes , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Osteoblastos/citologia , Osteoblastos/metabolismo , Polimorfismo de Nucleotídeo Único , Locos de Características Quantitativas , Canais de Cátion TRPP/antagonistas & inibidores , Canais de Cátion TRPP/genética
14.
Cell Rep ; 7(3): 634-44, 2014 May 08.
Artigo em Inglês | MEDLINE | ID: mdl-24767998

RESUMO

Autosomal dominant polycystic kidney disease is a common form of inherited kidney disease that is caused by mutations in two genes, PKD1 (polycystin-1) and PKD2 (polycystin-2). Mice with germline deletion of either gene die in midgestation with a vascular phenotype that includes profound edema. Although an endothelial cell defect has been suspected, the basis of this phenotype remains poorly understood. Here, we demonstrate that edema in Pkd1- and Pkd2-null mice is likely to be caused by defects in lymphatic development. Pkd1 and Pkd2 mutant embryos exhibit reduced lymphatic vessel density and vascular branching along with aberrant migration of early lymphatic endothelial cell precursors. We used cell-based assays to confirm that PKD1- and PKD2-depleted endothelial cells have an intrinsic defect in directional migration that is associated with a failure to establish front-rear polarity. Our studies reveal a role for polycystin signaling in lymphatic development.


Assuntos
Células Endoteliais/citologia , Linfonodos/embriologia , Transdução de Sinais , Canais de Cátion TRPP/metabolismo , Animais , Movimento Celular , Polaridade Celular , Embrião de Mamíferos/metabolismo , Embrião de Mamíferos/patologia , Células Endoteliais/metabolismo , Células Endoteliais da Veia Umbilical Humana , Humanos , Linfonodos/metabolismo , Vasos Linfáticos/embriologia , Vasos Linfáticos/fisiopatologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Rim Policístico Autossômico Dominante/genética , Rim Policístico Autossômico Dominante/metabolismo , Rim Policístico Autossômico Dominante/patologia , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Canais de Cátion TRPP/antagonistas & inibidores , Canais de Cátion TRPP/genética
15.
Cell Rep ; 7(3): 623-33, 2014 May 08.
Artigo em Inglês | MEDLINE | ID: mdl-24767999

RESUMO

Lymphatic vessels arise during development through sprouting of precursor cells from veins, which is regulated by known signaling and transcriptional mechanisms. The ongoing elaboration of vessels to form a network is less well understood. This involves cell polarization, coordinated migration, adhesion, mixing, regression, and shape rearrangements. We identified a zebrafish mutant, lymphatic and cardiac defects 1 (lyc1), with reduced lymphatic vessel development. A mutation in polycystic kidney disease 1a was responsible for the phenotype. PKD1 is the most frequently mutated gene in autosomal dominant polycystic kidney disease (ADPKD). Initial lymphatic precursor sprouting is normal in lyc1 mutants, but ongoing migration fails. Loss of Pkd1 in mice has no effect on precursor sprouting but leads to failed morphogenesis of the subcutaneous lymphatic network. Individual lymphatic endothelial cells display defective polarity, elongation, and adherens junctions. This work identifies a highly selective and unexpected role for Pkd1 in lymphatic vessel morphogenesis during development.


Assuntos
Linfangiogênese , Vasos Linfáticos/metabolismo , Canais de Cátion TRPP/metabolismo , Animais , Animais Geneticamente Modificados , Células Cultivadas , Embrião de Mamíferos/metabolismo , Desenvolvimento Embrionário , Células Endoteliais/citologia , Células Endoteliais/metabolismo , Humanos , Junções Intercelulares/metabolismo , Linfonodos/crescimento & desenvolvimento , Linfonodos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Mutação , Fenótipo , Rim Policístico Autossômico Dominante/genética , Rim Policístico Autossômico Dominante/metabolismo , Rim Policístico Autossômico Dominante/patologia , Canais de Cátion TRPP/antagonistas & inibidores , Canais de Cátion TRPP/genética , Peixe-Zebra , Proteínas de Peixe-Zebra/genética , Proteínas de Peixe-Zebra/metabolismo
16.
J Biol Chem ; 289(10): 6404-6414, 2014 Mar 07.
Artigo em Inglês | MEDLINE | ID: mdl-24459142

RESUMO

Mutations of the PKD1 and PKD2 genes, encoding polycystin-1 (PC1) and polycystin-2 (PC2), respectively, lead to autosomal dominant polycystic kidney disease. Interestingly, up-regulation or down-regulation of PKD1 or PKD2 leads to polycystic kidney disease in animal models, but their interrelations are not completely understood. We show here that full-length PC1 that interacts with PC2 via a C-terminal coiled-coil domain regulates PC2 expression in vivo and in vitro by down-regulating PC2 expression in a dose-dependent manner. Expression of the pathogenic mutant R4227X, which lacks the C-terminal coiled-coil domain, failed to down-regulate PC2 expression, suggesting that PC1-PC2 interaction is necessary for PC2 regulation. The proteasome and autophagy are two pathways that control protein degradation. Proteins that are not degraded by proteasomes precipitate in the cytoplasm and are transported via histone deacetylase 6 (HDAC6) toward the aggresomes. We found that HDAC6 binds to PC2 and that expression of full-length PC1 accelerates the transport of the HDAC6-PC2 complex toward aggresomes, whereas expression of the R4227X mutant fails to do so. Aggresomes are engulfed by autophagosomes, which then fuse with the lysosome for degradation; this process is also known as autophagy. We have now shown that PC1 overexpression leads to increased degradation of PC2 via autophagy. Interestingly, PC1 does not activate autophagy generally. Thus, we have now uncovered a new pathway suggesting that when PC1 is expressed, PC2 that is not bound to PC1 is directed to aggresomes and subsequently degraded via autophagy, a control mechanism that may play a role in autosomal dominant polycystic kidney disease pathogenesis.


Assuntos
Autofagia , Fagossomos , Canais de Cátion TRPP/metabolismo , Animais , Cães , Regulação para Baixo , Desacetilase 6 de Histona , Histona Desacetilases/metabolismo , Rim/metabolismo , Células Madin Darby de Rim Canino , Redes e Vias Metabólicas , Camundongos , Rim Policístico Autossômico Dominante/genética , Rim Policístico Autossômico Dominante/metabolismo , Canais de Cátion TRPP/antagonistas & inibidores , Canais de Cátion TRPP/genética
17.
FEBS J ; 279(10): 1857-70, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22420714

RESUMO

Polycystic kidney disease (PKD) 2L1 protein is a member of the transient receptor potential (TRP) ion channel family. In circumvallate and foliate papillae, PKD2L1 is coexpressed with PKD1L3. PKD2L1 and PKD1L3 interact through their transmembrane domain and the resulting heteromer PKD1L3/PKD2L1 owns a unique channel property called 'off-responses' to acid stimulation, although PKD2L1 does not own this property by itself. To define the pharmacological properties of the PKD1L3/PKD2L1 channel, we developed a new method to effectively evaluate channel activity using human embryonic kidney 293T cells in which the channel was heterologously expressed. This method was applied to screen substances that potentially regulate it. We found that capsaicin and its analogs, which are TRPV1 agonists, inhibited the response to acid stimuli and that the capsaicin inhibition was reversible with an IC(50) of 32.5 µm. Capsaicin and its analogs are thus useful tools for physiological analysis of PKD1L3/PKD2L1 function.


Assuntos
Capsaicina/farmacologia , Receptores de Superfície Celular/antagonistas & inibidores , Fármacos do Sistema Sensorial/farmacologia , Canais de Cátion TRPP/antagonistas & inibidores , Animais , Canais de Cálcio/metabolismo , Células Cultivadas , Células HEK293 , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Dados de Sequência Molecular , Receptores de Superfície Celular/metabolismo , Canais de Cátion TRPP/metabolismo , Papilas Gustativas/efeitos dos fármacos , Língua/metabolismo
18.
Dev Cell ; 22(1): 197-210, 2012 Jan 17.
Artigo em Inglês | MEDLINE | ID: mdl-22178500

RESUMO

Mutations in Pkd1, encoding polycystin-1 (PC1), cause autosomal-dominant polycystic kidney disease (ADPKD). We show that the carboxy-terminal tail (CTT) of PC1 is released by γ-secretase-mediated cleavage and regulates the Wnt and CHOP pathways by binding the transcription factors TCF and CHOP, disrupting their interaction with the common transcriptional coactivator p300. Loss of PC1 causes increased proliferation and apoptosis, while reintroducing PC1-CTT into cultured Pkd1 null cells reestablishes normal growth rate, suppresses apoptosis, and prevents cyst formation. Inhibition of γ-secretase activity impairs the ability of PC1 to suppress growth and apoptosis and leads to cyst formation in cultured renal epithelial cells. Expression of the PC1-CTT is sufficient to rescue the dorsal body curvature phenotype in zebrafish embryos resulting from either γ-secretase inhibition or suppression of Pkd1 expression. Thus, γ-secretase-dependent release of the PC1-CTT creates a protein fragment whose expression is sufficient to suppress ADPKD-related phenotypes in vitro and in vivo.


Assuntos
Secretases da Proteína Precursora do Amiloide/metabolismo , Apoptose , Fatores de Transcrição TCF/metabolismo , Canais de Cátion TRPP/fisiologia , Fator de Transcrição CHOP/metabolismo , Peixe-Zebra/metabolismo , Fatores de Transcrição de p300-CBP/genética , Animais , Proliferação de Células , Células Cultivadas , Cistos/etiologia , Cistos/metabolismo , Cistos/patologia , Embrião não Mamífero/citologia , Embrião não Mamífero/metabolismo , Humanos , Immunoblotting , Imunoprecipitação , Rim/metabolismo , Rim/patologia , Fenótipo , Rim Policístico Autossômico Dominante/fisiopatologia , Fatores de Transcrição TCF/genética , Canais de Cátion TRPP/antagonistas & inibidores , Fator de Transcrição CHOP/genética , Ativação Transcricional , Via de Sinalização Wnt , Peixe-Zebra/genética , Peixe-Zebra/crescimento & desenvolvimento , Fatores de Transcrição de p300-CBP/metabolismo
19.
J Cell Biochem ; 113(3): 967-76, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22034075

RESUMO

Mutations and/or deletions of Pkd1 in mouse models resulted in attenuation of osteoblast function and defective bone formation; however, the function of PKD1 in human osteoblast and bone remains uncertain. In the current study, we used lentivirus-mediated shRNA technology to stably knock down PKD1 in the human osteoblastic MG-63 cell line and to investigate the role of PKD1 on human osteoblast function and molecular mechanisms. We found that a 53% reduction of PKD1 by PKD1 shRNA in stable, transfected MG-63 cells resulted in increased cell proliferation and impaired osteoblastic differentiation as reflected by increased BrdU incorporation, decreased alkaline phosphatase activity, and calcium deposition and by decreased expression of RUNX2 and OSTERIX compared to control shRNA MG-63 cells. In addition, knockdown of PKD1 mRNA caused enhanced adipogenesis in stable PKD1 shRNA MG-63 cells as evidenced by elevated lipid accumulation and increased expression of adipocyte-related markers such as PPARγ and aP2. The stable PKD1 shRNA MG-63 cells exhibited lower basal intracellular calcium, which led to attenuated cytosolic calcium signaling in response to fluid flow shear stress, as well as increased intracellular cAMP messages in response to forskolin (10 µM) stimulation. Moreover, increased cell proliferation, inhibited osteoblastic differentiation, and osteogenic and adipogenic gene markers were significantly reversed in stable PKD1 shRNA MG-63 cells when treated with H89 (1 µM), an inhibitor of PKA. These findings suggest that downregulation of PKD1 in human MG-63 cells resulted in defective osteoblast function via intracellular calcium-cAMP/PKA signaling pathway.


Assuntos
Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , AMP Cíclico/metabolismo , Regulação para Baixo , Osteoblastos/metabolismo , Canais de Cátion TRPP/fisiologia , Adipogenia , Cálcio/metabolismo , Diferenciação Celular , Linhagem Celular , Proliferação de Células , Cílios/ultraestrutura , Proteínas Quinases Dependentes de AMP Cíclico/antagonistas & inibidores , Humanos , Lentivirus/genética , Osteoblastos/citologia , Osteoblastos/enzimologia , Osteogênese , RNA Interferente Pequeno/metabolismo , Transdução de Sinais , Canais de Cátion TRPP/antagonistas & inibidores , Canais de Cátion TRPP/genética
20.
Neuro Oncol ; 13(7): 710-24, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21727210

RESUMO

Glioblastoma multiforme, a highly aggressive tumor of the central nervous system, has a dismal prognosis that is due in part to its resistance to radio- and chemotherapy. The protein kinase C (PKC) family of serine threonine kinases has been implicated in the formation and proliferation of glioblastoma multiforme. Members of the protein kinase D (PKD) family, which consists of PKD1, -2 and, -3, are prominent downstream targets of PKCs and could play a major role in glioblastoma growth. PKD2 was highly expressed in both low-grade and high-grade human gliomas. The number of PKD2-positive tumor cells increased with glioma grading (P < .001). PKD2 was also expressed in CD133-positive glioblastoma stem cells and various glioblastoma cell lines in which the kinase was found to be constitutively active. Inhibition of PKDs by pharmacological inhibitors resulted in substantial inhibition of glioblastoma proliferation. Furthermore, specific depletion of PKD2 by siRNA resulted in a marked inhibition of anchorage-dependent and -independent proliferation and an accumulation of glioblastoma cells in G0/G1, accompanied by a down-regulation of cyclin D1 expression. In addition, PKD2-depleted glioblastoma cells exhibited substantially reduced tumor formation in vivo on chicken chorioallantoic membranes. These findings identify PKD2 as a novel mediator of glioblastoma cell growth in vitro and in vivo and thereby as a potential therapeutic target for this devastating disease.


Assuntos
Neoplasias Encefálicas/patologia , Encéfalo/enzimologia , Glioblastoma/patologia , Canais de Cátion TRPP/metabolismo , Animais , Apoptose , Western Blotting , Neoplasias Encefálicas/enzimologia , Ciclo Celular , Proliferação de Células , Galinhas , Membrana Corioalantoide/metabolismo , Ciclina D1/metabolismo , Glioblastoma/enzimologia , Humanos , Técnicas Imunoenzimáticas , RNA Interferente Pequeno/genética , Canais de Cátion TRPP/antagonistas & inibidores , Canais de Cátion TRPP/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...