RESUMO
The chemical and mechanical responses of cells via the exchange of information during growth and development result in the formation of biological tissues. Information processing within the cells through the signaling pathways and networks inherent to the constituent cells has been well-studied. However, the cell signaling mechanisms responsible for generating dynamic multicellular responses in developing tissues remain unclear. Here, I review the dynamic multicellular response systems during the development and growth of vertebrate tissues based on the extracellular signal-regulated kinase (ERK) pathway. First, an overview of the function of the ERK signaling network in cells is provided, followed by descriptions of biosensors essential for live imaging of the quantification of ERK activity in tissues. Then adducing four examples, I highlight the contribution of live imaging techniques for studying the involvement of spatio-temporal patterns of ERK activity change in tissue development and growth. In addition, theoretical implications of ERK signaling are also discussed from the viewpoint of dynamic systems. This review might help in understanding ERK-mediated dynamic multicellular responses and tissue morphogenesis.
Assuntos
MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Transferência Ressonante de Energia de Fluorescência/métodos , Sistema de Sinalização das MAP Quinases/fisiologia , Morfogênese/fisiologia , Receptores Proteína Tirosina Quinases/metabolismo , Animais , Técnicas Biossensoriais/métodos , Ativação Enzimática/fisiologia , Humanos , Cinética , Camundongos , Ligação ProteicaRESUMO
Cystatin C is a ubiquitously expressed cysteine protease inhibitor that protects cells from either improper hydrolysis by endogenous proteases or pathogen growth/virulence by exogenous proteases. Although commonly used as a serum biomarker for evaluating renal function, cystatin C is associated with many immunological disorders under various pathophysiological conditions. How cystatin C affects immune cells, especially dendritic cells (DCs), however, is far from clear. In this study, we found that pharmacological treatment with or genetic overexpression of cystatin C in bone marrow-derived DCs (BMDCs) reduced their capacity to stimulate CD4+ T-cell proliferation, despite increased antigen uptake. This reduced capacity corresponded with reduced major histocompatibility complex-II presentation owing to diminished levels of the chaperon H2-DM in BMDCs. Instead of promoting proliferation, cystatin C promoted skewing of T cells toward proinflammatory T-helper (Th)1/Th17 differentiation. This was mediated by augmented extracellular signal-regulated kinase 1/2 mitogen-activated protein kinase phosphorylation in BMDCs, leading to secretion of polarizing cytokines, which in turn led to the Th deviation. Collectively, our study explained the cellular and molecular basis of how this protease inhibitor can regulate immune responses, namely by affecting BMDCs and their cytokine pathway. Our results might open up an avenue for the development of therapeutic agents for the treatment of cystatin C-related immunological diseases.
Assuntos
Apresentação de Antígeno , Células da Medula Óssea/citologia , Cistatina C/metabolismo , Citocinas/biossíntese , Células Dendríticas/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Antígenos de Histocompatibilidade Classe II/metabolismo , Peptídeos/metabolismo , Animais , Diferenciação Celular , Linhagem Celular , Polaridade Celular , Proliferação de Células , Galinhas , Camundongos Endogâmicos C57BL , Óxido Nítrico/biossíntese , Células Th1/imunologia , Células Th17/imunologiaRESUMO
UNLABELLED: Growing evidence shows that mechanisms controlling CNS plasticity extend beyond the synapse and that alterations in myelin can modify conduction velocity, leading to changes in neural circuitry. Although it is widely accepted that newly generated oligodendrocytes (OLs) produce myelin in the adult CNS, the contribution of preexisting OLs to functional myelin remodeling is not known. Here, we show that sustained activation of extracellular signal-regulated kinases 1 and 2 (ERK1/2) in preexisting OLs of adult mice is sufficient to drive increased myelin thickness, faster conduction speeds, and enhanced hippocampal-dependent emotional learning. Although preexisting OLs do not normally contribute to remyelination, we show that sustained activation of ERK1/2 renders them able to do so. These data suggest that strategies designed to push mature OLs to reinitiate myelination may be beneficial both for enhancing remyelination in demyelinating diseases and for increasing neural plasticity in the adult CNS. SIGNIFICANCE STATEMENT: Myelin is a crucial regulator of CNS plasticity, function, and repair. Although it is generally accepted that new myelin production in the adult CNS is initiated by newly generated oligodendrocytes (OLs), great interest remains in additionally driving mature preexisting OLs to make myelin. The ability to induce myelination by the larger population of preexisting OLs carries the potential for enhanced remyelination in demyelinating diseases and increased neural plasticity in the adult CNS. Here, we show that sustained activation of the extracellular signal-regulated kinases 1 and 2 (ERK1/2) signaling pathway is sufficient to drive mature OLs in the adult mouse CNS to reinitiate myelination, leading to new myelin wraps and functional changes.
Assuntos
Sistema Nervoso Central/fisiologia , Doenças Desmielinizantes/patologia , Sistema de Sinalização das MAP Quinases/fisiologia , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Bainha de Mielina/metabolismo , Oligodendroglia/fisiologia , Análise de Variância , Animais , Condicionamento Psicológico/fisiologia , Doenças Desmielinizantes/induzido quimicamente , Doenças Desmielinizantes/fisiopatologia , Modelos Animais de Doenças , Potenciais Evocados Auditivos do Tronco Encefálico/genética , Comportamento Exploratório/fisiologia , Medo/fisiologia , Galactosilceramidas/metabolismo , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Lisofosfolipase/toxicidade , MAP Quinase Quinase 1/genética , MAP Quinase Quinase 1/metabolismo , Sistema de Sinalização das MAP Quinases/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microscopia Eletrônica de Transmissão , Proteína Quinase 3 Ativada por Mitógeno/genética , Força Muscular/genética , Proteína Proteolipídica de Mielina/genética , Proteína Proteolipídica de Mielina/metabolismo , Bainha de Mielina/ultraestrutura , Proteínas do Tecido Nervoso/metabolismo , Oligodendroglia/ultraestrutura , Desempenho Psicomotor/fisiologia , RNA Mensageiro/metabolismo , Reconhecimento Psicológico/fisiologia , Espectrometria de Massas em TandemRESUMO
Hippocampal adult neurogenesis contributes to key functions of the dentate gyrus (DG), including contextual discrimination. This is due, at least in part, to the unique form of plasticity that new neurons display at a specific stage of their development when compared with the surrounding principal neurons. In addition, the contribution that newborn neurons make to dentate function can be enhanced by an increase in their numbers induced by a stimulating environment. However, signaling mechanisms that regulate these properties of newborn neurons are poorly understood. Here, we show that Ras-GRF2 (GRF2), a calcium-regulated exchange factor that can activate Ras and Rac GTPases, contributes to both of these properties of newborn neurons. Using Ras-GRF2 knockout mice and wild-type mice stereotactically injected with retrovirus containing shRNA against the exchange factor, we demonstrate that GRF2 promotes the survival of newborn neurons of the DG at approximately 1-2 weeks after their birth. GRF2 also controls the distinct form of long-term potentiation that is characteristic of new neurons of the hippocampus through its effector Erk MAP kinase. Moreover, the enhancement of neuron survival that occurs after mice are exposed to an enriched environment also involves GRF2 function. Consistent with these observations, GRF2 knockout mice display defective contextual discrimination. Overall, these findings indicate that GRF2 regulates both the basal level and environmentally induced increase of newborn neuron survival, as well as in the induction of a distinct form of synaptic plasticity of newborn neurons that contributes to distinct features of hippocampus-derived learning and memory.
Assuntos
Sobrevivência Celular/fisiologia , Giro Denteado/fisiologia , Abrigo para Animais , Potenciação de Longa Duração/fisiologia , Neurônios/fisiologia , Fatores ras de Troca de Nucleotídeo Guanina/metabolismo , Animais , Sobrevivência Celular/efeitos dos fármacos , Giro Denteado/efeitos dos fármacos , Discriminação Psicológica/fisiologia , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Reação de Congelamento Cataléptica/fisiologia , Potenciação de Longa Duração/efeitos dos fármacos , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neurogênese/efeitos dos fármacos , Neurogênese/fisiologia , Neurônios/efeitos dos fármacos , RNA Interferente Pequeno , Percepção Espacial/fisiologia , Técnicas de Cultura de Tecidos , Fatores ras de Troca de Nucleotídeo Guanina/genéticaRESUMO
Regulation of TGF-ß1/Smad3 signaling in fibrogenesis is complex. Previous work by our lab suggests that ERK MAP kinase phosphorylates the linker region (LR) of Smad3 to enhance TGF-ß-induced collagen-I accumulation. However the roles of the individual Smad3LR phosphorylation sites (T179, S204, S208 and S213) in the collagen-I response to TGF-ß are not clear. To address this issue, we tested the ability of Smad3 constructs expressing wild-type Smad3 or Smad3 with mutated LR phosphorylation sites to reconstitute TGF-ß-stimulated COL1A2 promoter activity in Smad3-null or -knockdown cells. Blocking ERK in fibroblasts and renal mesangial cells inhibited both S204 phosphorylation and Smad3-mediated COL1A2 promoter activity. Mutations replacing serine at S204 or S208 in the linker region decreased Smad3-mediated COL1A2 promoter activity, whereas mutating T179 enhanced basal COL1A2 promoter activity and did not prevent TGF-ß stimulation. Interestingly, mutation of all four Smad3LR sites (T179, S204, S208 and S213) was not inhibitory, suggesting primacy of the two inhibitory sites. These results suggest that in these mesenchymal cells, phosphorylation of the T179 and possibly S213 sites may act as a brake on the signal, whereas S204 phosphorylation by ERK in some manner releases that brake. Renal epithelial cells (HKC) respond differently from MEF or mesangial cells; blocking ERK neither changed TGF-ß-stimulated S204 phosphorylation nor prevented Smad3-mediated COL1A2 promoter activity in HKC. Furthermore, re-expression of wild type-Smad3 or the S204A-Smad3 mutant in Smad3-knockdown HKC reconstituted Smad3-mediated COL1A2 promoter activity. Collectively, these data suggest that Serine-204 phosphorylation in the Smad3LR is a critical event by which ERK enhances Smad3-mediated COL1A2 promoter activity in mesenchymal cells.
Assuntos
Colágeno Tipo I/metabolismo , Serina/metabolismo , Proteína Smad3/metabolismo , Transativadores/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Animais , Células Cultivadas , Colágeno Tipo I/genética , MAP Quinases Reguladas por Sinal Extracelular/genética , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Camundongos , Mutação/genética , Fenótipo , Transdução de Sinais/fisiologia , Transativadores/genética , Ativação Transcricional/fisiologia , Fator de Crescimento Transformador beta/genéticaRESUMO
Extracellular signal-regulated kinase (ERK) plays a crucial role in regulating collective cell behaviors observed in diverse biological phenomena. Emerging studies have shed light on the involvement of the ERK signaling pathway in the reception and generation of mechanical forces, thereby governing local mechanical interactions within multicellular tissues. Although limited in number, studies have provided insights into how ERK-mediated mechanical interactions contribute to multicellular organization. Here we explore the impact of ERK-mediated mechanical interactions on tissue morphogenesis, cell extrusion in homeostasis, and their interplay with the physical microenvironments of the extracellular matrix. We conclude that the coupling system of ERK activity with mechanical forces offers a promising avenue to unravel the emergent collective dynamics underlying tissue organization.
Assuntos
MAP Quinases Reguladas por Sinal Extracelular , Transdução de Sinais , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Fosforilação , Sistema de Sinalização das MAP Quinases , Matriz Extracelular/metabolismoRESUMO
Extracellular signal-regulated kinase (ERK) has been recognized as a critical regulator in various physiological and pathological processes. Extensive research has elucidated the signaling mechanisms governing ERK activation via biochemical regulations with upstream molecules, particularly receptor tyrosine kinases (RTKs). However, recent advances have highlighted the role of mechanical forces in activating the RTK-ERK signaling pathways, thereby opening new avenues of research into mechanochemical interplay in multicellular tissues. Here, we review the force-induced ERK activation in cells and propose possible mechanosensing mechanisms underlying the mechanoresponsive ERK activation. We conclude that mechanical forces are not merely passive factors shaping cells and tissues but also active regulators of cellular signaling pathways controlling collective cell behaviors.
Assuntos
MAP Quinases Reguladas por Sinal Extracelular , Sistema de Sinalização das MAP Quinases , Sistema de Sinalização das MAP Quinases/fisiologia , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Transdução de Sinais/fisiologia , Receptores Proteína Tirosina Quinases/metabolismoRESUMO
Osteosarcoma (OS) is the most common primary bone tumor that mainly affects adolescents and young adults. Although standard treatment modality can achieve up to 60%-70% 5-year survival rate, there has not been any substantial improvement over the past four decades. Furthermore, those presenting with pulmonary metastatic lesions often undergo a highly unfavorable clinical course. Therefore, there is a severely unmet clinical need to provide a more effective treatment for patients with OS. In this study, we show that trabectedin (TBD), a chemotherapeutic agent approved for soft tissue sarcomas, significantly suppresses pulmonary metastasis in a mouse OS xenograft model. In vitro experiments revealed that TBD suppresses cell migration potentially by downregulating the activity of ERK1/2, intracellular molecules that are critically involved in the regulation of cell motility. Collectively, our data may provide a basis for further investigation of TBD on the potential use for OS patients who are at great risk of pulmonary metastasis.
Assuntos
Neoplasias Ósseas , Neoplasias Pulmonares , Osteossarcoma , Adolescente , Animais , Neoplasias Ósseas/tratamento farmacológico , Neoplasias Ósseas/patologia , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Modelos Animais de Doenças , Xenoenxertos , Humanos , Neoplasias Pulmonares/patologia , Camundongos , Metástase Neoplásica , Osteossarcoma/tratamento farmacológico , Osteossarcoma/patologia , Trabectedina/uso terapêuticoRESUMO
Retinoic acid (RA) and its synthetic derivatives, retinoids, have been established as promising anticancer agents based on their ability to regulate cell proliferation and survival. Clinical trials, however, have revealed that cancer cells often acquire resistance to retinoid therapy. Therefore, elucidation of underlying mechanisms of retinoid resistance has been considered key to developing more effective use of retinoids in cancer treatment. In this study, we show that constitutive activation of ERK MAP kinase signaling, which is often caused by oncogenic mutations in RAS or RAF genes, suppresses RA receptor (RAR) signaling in breast cancer cells. We show that activation of the ERK pathway suppresses, whereas its inhibition promotes, RA-induced transcriptional activation of RAR and the resultant upregulation of RAR-target genes in breast cancer cells. Importantly, ERK inhibition potentiates the tumor-suppressive activity of RA in breast cancer cells. Moreover, we also reveal that suppression of RAR signaling and activation of ERK signaling are associated with poor prognoses in breast cancer patients and represent hallmarks of specific subtypes of breast cancers, such as basal-like, HER2-enriched and luminal B. These results indicate that ERK-dependent suppression of RAR activity underlies retinoid resistance and is associated with cancer subtypes and patient prognosis in breast cancers.
RESUMO
Several studies have established the crucial role of the extracellular signal-regulated kinase (ERK)/mitogen-activated protein kinase pathway in hematopoietic cell proliferation and differentiation. MEK1 and MEK2 phosphorylate and activate ERK1 and ERK2. However, whether MEK1 and MEK2 differentially regulate these processes is unknown. To define the function of Mek genes in the activation of the ERK pathway during hematopoiesis, we generated a mutant mouse line carrying a hematopoietic-specific deletion of the Mek1 gene function in a Mek2 null background. Inactivation of both Mek1 and Mek2 genes resulted in death shortly after birth with a severe anemia revealing the essential role of the ERK pathway in erythropoiesis. Mek1 and Mek2 functional ablation also affected lymphopoiesis and myelopoiesis. In contrast, mice that retained one functional Mek1 (1Mek1) or Mek2 (1Mek2) allele in hematopoietic cells were viable and fertile. 1Mek1 and 1Mek2 mutants showed mild signs of anemia and splenomegaly, but the half-life of their red blood cells and the response to erythropoietic stress were not altered, suggesting a certain level of Mek redundancy for sustaining functional erythropoiesis. However, subtle differences in multipotent progenitor distribution in the bone marrow were observed in 1Mek1 mice, suggesting that the two Mek genes might differentially regulate early hematopoiesis.
RESUMO
Nitric oxide is an important neuromodulator in the CNS, and its production within neurons is modulated by NMDA receptors and requires a fine-tuned availability of L-arginine. We have previously shown that globally inhibiting protein synthesis mobilizes intracellular L-arginine "pools" in retinal neurons, which concomitantly enhances neuronal nitric oxide synthase-mediated nitric oxide production. Activation of NMDA receptors also induces local inhibition of protein synthesis and L-arginine intracellular accumulation through calcium influx and stimulation of eucariotic elongation factor type 2 kinase. We hypothesized that protein synthesis inhibition might also increase intracellular L-arginine availability to induce nitric oxide-dependent activation of downstream signaling pathways. Here we show that nitric oxide produced by inhibiting protein synthesis (using cycloheximide or anisomycin) is readily coupled to AKT activation in a soluble guanylyl cyclase and cGKII-dependent manner. Knockdown of cGKII prevents cycloheximide or anisomycin-induced AKT activation and its nuclear accumulation. Moreover, in retinas from cGKII knockout mice, cycloheximide was unable to enhance AKT phosphorylation. Indeed, cycloheximide also produces an increase of ERK phosphorylation which is abrogated by a nitric oxide synthase inhibitor. In summary, we show that inhibition of protein synthesis is a previously unanticipated driving force for nitric oxide generation and activation of downstream signaling pathways including AKT and ERK in cultured retinal cells. These results may be important for the regulation of synaptic signaling and neuronal development by NMDA receptors as well as for solving conflicting data observed when using protein synthesis inhibitors for studying neuronal survival during development as well in behavior and memory studies.
Assuntos
Proteína Quinase Dependente de GMP Cíclico Tipo II/metabolismo , Óxido Nítrico/metabolismo , Inibidores da Síntese de Proteínas/farmacologia , Retina/metabolismo , Transdução de Sinais/efeitos dos fármacos , Animais , Arginina/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Embrião de Galinha , Galinhas , Proteína Quinase Dependente de GMP Cíclico Tipo II/genética , Quinase do Fator 2 de Elongação/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neurônios/metabolismo , Nitratos/metabolismo , Óxido Nítrico Sintase Tipo I/metabolismo , Nitritos , FosforilaçãoRESUMO
Agonist stimulation of G-protein-coupled receptors (GPCRs) typically results in phosphorylation and activation of ERK (Extracellular-signal Regulated Kinase) which is a member of MAP kinase (Mitogen-Activated Protein kinase) family. Detection of phosphorylated ERK1/2 MAP kinase has been widely used as readout of GPCR signaling in heterologous cells, primary cells, tissues and even in animal studies. ERK1/2 phosphorylation downstream of GPCRs is now well established to arise from the activation of both, the heterotrimeric G-proteins and ß-arrestins (ßarrs) with distinct spatio-temporal components. Here, we present a step-by-step protocol for measuring agonist-induced ERK1/2 MAP kinase activation downstream of GPCRs using standard Western blotting assay. Note: ERK1/2 is also referred to as p44/42 MAP kinase. ERK1 and ERK2 are same as Mitogen-Activated Protein Kinase 3 (MAP3) and Mitogen-Activated Protein Kinase 1 (MAP1), respectively.
Assuntos
Técnicas Citológicas/métodos , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Receptores Acoplados a Proteínas G/agonistas , Proliferação de Células , Células HEK293 , Humanos , Fosforilação , Polietilenoimina/químicaRESUMO
The extracellular signal-regulated kinase (ERK) signaling pathway regulates a variety of biological processes including cell proliferation, survival, and differentiation. Since ERK activation promotes proliferation of many types of cells, its deregulated/constitutive activation is among general mechanisms for cancer. Recent advances in bioimaging techniques have enabled to visualize ERK activity in real-time at the single-cell level. Emerging evidence from such approaches suggests unexpectedly complex spatiotemporal dynamics of ERK activity in living cells and animals and their crucial roles in determining cellular responses. In this review, we discuss how ERK activity dynamics are regulated and how they affect biological processes including cell fate decisions, cell migration, embryonic development, tissue homeostasis, and tumorigenesis.
RESUMO
Retinoic acid receptor (RAR) signaling plays an important role in embryonic development and homeostasis of many tissues. At the cellular level, activation of RAR signaling often induces cell cycle arrest, differentiation, and apoptosis in many types of cells. Consequently, loss of normal RAR function in the presence of physiological levels of retinoic acid (RA) is often observed in cancers, and pharmacological reactivation of RAR signaling has been considered a promising strategy for cancer therapy and prevention. One of important mechanisms that regulate RAR activity in cancer cells is cross-talk with growth factor signaling, where activation of extracellular signal-regulated kinase (ERK) plays a major role in suppressing RAR transcriptional activity downstream of growth factor receptors. Conversely, strong activation of RAR can induce suppression of ERK activity by inducing expression of a phosphatase specific for ERK to exert tumor-suppressive activity in colorectal cancer. Here, we describe the basic methods to analyze interactions between RAR and ERK signaling in colorectal cancer cells.
Assuntos
Neoplasias Colorretais/genética , Receptores de Fatores de Crescimento/genética , Receptores do Ácido Retinoico/metabolismo , Células CACO-2 , Neoplasias Colorretais/metabolismo , Regulação Neoplásica da Expressão Gênica , Células HEK293 , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Sistema de Sinalização das MAP QuinasesRESUMO
Extracellular signal-regulated kinase (ERK) plays a critical role in tissue homeostasis and tumorigenesis. By utilizing live imaging approaches, we recently uncovered ERK activity dynamics in the intestinal epithelium. Notably, we showed that ERK activity dynamics are defined by composite regulation from two distinct upstream receptors, and alteration of their functional balance underlies tumor cell-specific traits. Here, we discuss these findings.
RESUMO
BACKGROUND: Aggressive natural killer cell leukemia is a devastating disease, with an average patient survival time of less than 2 months following diagnosis. Due to P-glycoprotein-mediated resistance of the tumor cells most forms of chemotherapy are of limited efficacy, therefore new treatment strategies are needed. Statin drugs have recently been found to inhibit the growth of various tumor cell types. METHODS: We investigated the effects of statin drug-mediated mevalonate pathway inhibition on cell proliferation, tumor-induced cytotoxicity, cell cycle progression and ERK MAP kinase signal transduction pathway activation. Flow cytometry was used to perform the cytotoxicity and cell cycle analyses and Western blotting was used to investigate ERK MAP kinase activation. Statistical significance was assessed by Student's t-test. RESULTS: Fluvastatin and atorvastatin were found to inhibit cell growth and tumor-induced cytotoxicity. These effects were reversed by the addition of mevalonate, signifying that the impact of the drugs were on the mevalonate pathway. Both drugs affected cell cycle progression by causing a significant increase in the percentage of cells in the G0/G1 phase and a reduction in the S phase and the G2/M phases of the cell cycle. Low concentrations of statin drugs were able to abrogate ERK MAP kinase pathway activation, which is typically constitutively activated in aggressive natural killer cell leukemias and important in tumor-mediated cytotoxicity. Addition of statins to chemotherapy caused enhanced inhibition of cell growth and cytotoxicity, compared to either agent alone; a combination therapy that could conceivably benefit some patients. CONCLUSIONS: These investigations suggest that inhibiting the mevalonate pathway might provide a more effective therapy against this deadly disease when combined with chemotherapy. Given that millions of people are currently taking statin drugs to lower cholesterol levels, the risk profile for statin drugs and their side effects are well-known. Our studies suggest that it may be beneficial to explore statin-chemotherapy combination in the treatment of aggressive natural killer cell leukemias.
RESUMO
Deregulated activation of RAS/extracellular signal-regulated kinase (ERK) signaling and defects in retinoic acid receptor (RAR) signaling are both implicated in many types of cancers. However, interrelationships between these alterations in regulating cancer cell fates have not been fully elucidated. Here, we show that RAS/ERK and RAR signaling pathways antagonistically interact with each other to regulate colorectal cancer (CRC) cell fates. We show that RAR signaling activation promotes spontaneous differentiation of CRC cells, while ERK activation suppresses it. Our microarray analyses identify genes whose expression levels are upregulated by RAR signaling. Notably, one of these genes, MKP4, encoding a member of dual-specificity phosphatases for mitogen-activated protein (MAP) kinases, mediates ERK inactivation upon RAR activation, thereby promoting the differentiation of CRC cells. Moreover, our results also show that RA induction of RAR target genes is suppressed by the ERK pathway activation. This suppression results from the inhibition of RAR transcriptional activity, which is shown to be mediated through an RIP140/histone deacetylase (HDAC)-mediated mechanism. These results identify antagonistic interactions between RAS/ERK and RAR signaling in the cell fate decision of CRC cells and define their underlying molecular mechanisms.
Assuntos
Colo/patologia , Neoplasias Colorretais/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Receptores do Ácido Retinoico/metabolismo , Reto/patologia , Transdução de Sinais , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Diferenciação Celular , Linhagem Celular Tumoral , Colo/metabolismo , Neoplasias Colorretais/genética , Neoplasias Colorretais/patologia , Fosfatases de Especificidade Dupla/genética , Fosfatases de Especificidade Dupla/metabolismo , Regulação Neoplásica da Expressão Gênica , Histona Desacetilases/metabolismo , Humanos , Fosfatases da Proteína Quinase Ativada por Mitógeno/genética , Fosfatases da Proteína Quinase Ativada por Mitógeno/metabolismo , Proteínas Nucleares/metabolismo , Proteína 1 de Interação com Receptor Nuclear , Regiões Promotoras Genéticas , Reto/metabolismoRESUMO
BACKGROUND: Human telomerase reverse transcriptase (hTERT) is one of the components of telomerase enzyme and its activity is associated with cell proliferation and differentiation. Extracellular signal regulated kinase (ERK)-mitogen activated protein kinase signaling pathway play an important role in hTERT expression. The present study was conducted to ascertain hTERT messenger RNA (mRNA) expression in renal cell carcinoma (RCC) and its association with clinicopathological characteristics. Further, we also explored hTERT targeting as possible tumor therapeutic. METHODS: A total of 96 histopathologically confirmed RCC cases and corresponding normal tissues were subjected to hTERT gene expression using real-time PCR. Two RCC cell lines viz. ACHN and A498 were treated with MEK inhibitor (U0126) and hTERT mRNA expression, telomerase activity, cell viability, migration, and apoptosis were evaluated. RESULTS: The hTERT mRNA levels were found to be significantly higher in RCC as compared with corresponding normal renal tissues (P = 0.040) as well as in high grades to that of low grades clear cell RCC (P = 0.008). Significantly diminished ERK phosphorylation and hTERT mRNA expression concomitantly reduced telomerase activity that was observed after U0126 treatment. Subsequently, cell viability and migration were significantly inhibited after treatment with U0126 in both the cell lines to that of control (P = 0.001). In addition, U0126-treated cells showed significantly increased apoptosis (P = 0.001) to that of controls. CONCLUSION: Henceforth, this study infer that, hTERT expression can be used as a possible diagnostic marker for RCC, and inhibition of hTERT expression by hampering ERK-mitogen activated protein kinase cascade may be used as a promising anticancer target in RCC.
Assuntos
Carcinoma de Células Renais/genética , MAP Quinases Reguladas por Sinal Extracelular/antagonistas & inibidores , Neoplasias Renais/genética , Telomerase/metabolismo , Carcinoma de Células Renais/patologia , Morte Celular , Humanos , Neoplasias Renais/patologia , Pessoa de Meia-IdadeRESUMO
BACKGROUND: Estrogen and progesterone are potent breast mitogens. In addition to steroid hormones, multiple signaling pathways input to estrogen receptor (ER) and progesterone receptor (PR) actions via posttranslational events. Protein kinases commonly activated in breast cancers phosphorylate steroid hormone receptors (SRs) and profoundly impact their activities. METHODS: To better understand the role of modified PRs in breast cancer, we measured total and phospho-Ser294 PRs in 209 human breast tumors represented on 2754 individual tissue spots within a tissue microarray and assayed the regulation of this site in human tumor explants cultured ex vivo. To complement this analysis, we assayed PR target gene regulation in T47D luminal breast cancer models following treatment with progestin (promegestone; R5020) and antiprogestins (mifepristone, onapristone, or aglepristone) in conditions under which the receptor is regulated by Lys388 SUMOylation (K388 intact) or is SUMO-deficient (via K388R mutation to mimic persistent Ser294 phosphorylation). Selected phospho-PR-driven target genes were validated by qRT-PCR and following RUNX2 shRNA knockdown in breast cancer cell lines. Primary and secondary mammosphere assays were performed to implicate phospho-Ser294 PRs, epidermal growth factor signaling, and RUNX2 in breast cancer stem cell biology. RESULTS: Phospho-Ser294 PR species were abundant in a majority (54%) of luminal breast tumors, and PR promoter selectivity was exquisitely sensitive to posttranslational modifications. Phospho-PR expression and target gene programs were significantly associated with invasive lobular carcinoma (ILC). Consistent with our finding that activated phospho-PRs undergo rapid ligand-dependent turnover, unique phospho-PR gene signatures were most prevalent in breast tumors clinically designated as PR-low to PR-null (luminal B) and included gene sets associated with cancer stem cell biology (HER2, PAX2, AHR, AR, RUNX). Validation studies demonstrated a requirement for RUNX2 in the regulation of selected phospho-PR target genes (SLC37A2). In vitro mammosphere formation assays support a role for phospho-Ser294-PRs via growth factor (EGF) signaling as well as RUNX2 as potent drivers of breast cancer stem cell fate. CONCLUSIONS: We conclude that PR Ser294 phosphorylation is a common event in breast cancer progression that is required to maintain breast cancer stem cell fate, in part via cooperation with growth factor-initiated signaling pathways and key phospho-PR target genes including SLC37A2 and RUNX2. Clinical measurement of phosphorylated PRs should be considered a useful marker of breast tumor stem cell potential. Alternatively, unique phospho-PR target gene sets may provide useful tools with which to identify patients likely to respond to selective PR modulators that block PR Ser294 phosphorylation as part of rational combination (i.e., with antiestrogens) endocrine therapies designed to durably block breast cancer recurrence.
Assuntos
Neoplasias da Mama/patologia , Genes Neoplásicos/genética , Células-Tronco Neoplásicas/patologia , Processamento de Proteína Pós-Traducional , Receptores de Progesterona/metabolismo , Antiporters/fisiologia , Neoplasias da Mama/metabolismo , Linhagem Celular Tumoral , Subunidade alfa 1 de Fator de Ligação ao Core/fisiologia , Feminino , Expressão Gênica , Humanos , Ligantes , Células-Tronco Neoplásicas/metabolismo , Fosforilação , Serina/metabolismo , Análise Serial de Tecidos , Células Tumorais CultivadasRESUMO
Cocaine self-administration disturbs intracellular signaling in prefrontal cortical neurons that regulate neurotransmission in the nucleus accumbens. The deficits in dorsomedial prefrontal cortex (dmPFC) signaling change over time, resulting in different neuroadaptations during early withdrawal from cocaine self-administration than after one or more weeks of abstinence. Within the first few hours of withdrawal, there is a marked decrease in tyrosine phosphorylation of critical intracellular and membrane-bound proteins in the dmPFC that include ERK/MAP kinase and the NMDA receptor subunits, GluN1 and GluN2B. These changes are accompanied by a marked increase in STEP tyrosine phosphatase activation. Simultaneously, ERK and PKA-dependent synapsin phosphorylation in presynaptic terminals of the nucleus accumbens is increased that may have a destabilizing impact on glutamatergic transmission. Infusion of brain-derived neurotrophic factor (BDNF) into the dmPFC immediately following a final session of cocaine self-administration blocks the cocaine-induced changes in phosphorylation and attenuates relapse to cocaine seeking for as long as three weeks. The intra-dmPFC BDNF infusion also prevents cocaine-induced deficits in prefronto-accumbens glutamatergic transmission that are implicated in cocaine seeking. Thus, intervention with BDNF in the dmPFC during early withdrawal has local and distal effects in target areas that are critical to mediating cocaine-induced neuroadaptations that lead to cocaine seeking.