Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
J Immunol ; 195(5): 2207-15, 2015 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-26202984

RESUMO

The anti-CD20 mAb, rituximab, is routinely used to treat B cell malignancies. However, a majority of patients relapse. An improvement in the complete response was obtained by combining rituximab with chemotherapy, at the cost of increased toxicity. We reported that rituximab induced the colocalization of both the Orai1 Ca(2+) release-activated Ca(2+) channel (CRAC) and the endoplasmic reticulum Ca(2+) sensor stromal interaction molecule 1 with CD20 and CD95 into a cluster, eliciting a polarized store-operated Ca(2+) entry (SOCE). We observed that blocking this Ca(2+) entry with downregulation of Orai1, pharmacological inhibitors, or reducing calcemia with hypocalcemic drugs sensitized human B lymphoma cell lines and primary human lymphoma cells to rituximab-induced apoptosis in vitro, and improved the antitumoral effect of rituximab in xenografted mice. This revealed that Ca(2+) entry exerted a negative feedback loop on rituximab-induced apoptosis, suggesting that associating CRAC channel inhibitors or hypocalcemic agents with rituximab may improve the treatment of patients with B cell malignancies. The calcium-dependent proteins involved in this process appear to vary according to the B lymphoma cell type, suggesting that CRAC-channel targeting is likely to be more efficient than calcium-dependent protein targeting.


Assuntos
Apoptose/efeitos dos fármacos , Canais de Cálcio/metabolismo , Cálcio/metabolismo , Linfoma não Hodgkin/tratamento farmacológico , Rituximab/farmacologia , Receptor fas/metabolismo , Animais , Antígenos CD20/imunologia , Antígenos CD20/metabolismo , Antineoplásicos/administração & dosagem , Antineoplásicos/farmacologia , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Western Blotting , Canais de Cálcio/genética , Linhagem Celular Tumoral , Difosfonatos/farmacologia , Retículo Endoplasmático/metabolismo , Feminino , Células HEK293 , Humanos , Imidazóis/farmacologia , Linfoma não Hodgkin/genética , Linfoma não Hodgkin/patologia , Proteínas de Membrana/metabolismo , Camundongos Knockout , Microscopia Confocal , Proteínas de Neoplasias/metabolismo , Proteína ORAI1 , Interferência de RNA , Rituximab/administração & dosagem , Molécula 1 de Interação Estromal , Ensaios Antitumorais Modelo de Xenoenxerto , Ácido Zoledrônico
2.
Proc Natl Acad Sci U S A ; 108(47): 19072-7, 2011 11 22.
Artigo em Inglês | MEDLINE | ID: mdl-22065776

RESUMO

The death receptor CD95 plays a pivotal role in immune surveillance and immune tolerance. Binding of CD95L to CD95 leads to recruitment of the adaptor protein Fas-associated death domain protein (FADD), which in turn aggregates caspase-8 and caspase-10. Efficient formation of the CD95/FADD/caspase complex, known as the death-inducing signaling complex (DISC), culminates in the induction of apoptosis. We show that cells exposed to CD95L undergo a reorganization of the plasma membrane in which the Ca(2+) release-activated Ca(2+) channel Orai1 and the endoplasmic reticulum-resident activator stromal interaction molecule 1 colocalize with CD95 into a micrometer-sized cluster in which the channel elicits a polarized entry of calcium. Orai1 knockdown and expression of a dominant negative construct (Orai1E106A) reveal that on CD95 engagement, the Orai1-driven localized Ca(2+) influx is fundamental to recruiting the Ca(2+)-dependent protein kinase C (PKC) ß2 to the DISC. PKCß2 in turn transiently holds the complex in an inactive status, preventing caspase activation and transmission of the apoptotic signal. This study identifies a biological role of Ca(2+) and the Orai1 channel that drives a transient negative feedback loop, introducing a lag phase in the early steps of the CD95 signal. We suggest that these localized events provide a time of decision to prevent accidental cell death.


Assuntos
Apoptose/fisiologia , Canais de Cálcio/metabolismo , Cálcio/metabolismo , Complexos Multiproteicos/metabolismo , Proteína Quinase C/metabolismo , Receptor fas/metabolismo , Western Blotting , Caspase 10/metabolismo , Caspase 8/metabolismo , Linhagem Celular , Proteína Ligante Fas/metabolismo , Proteína de Domínio de Morte Associada a Fas/metabolismo , Citometria de Fluxo , Humanos , Imunoprecipitação , Microscopia Confocal , Proteína ORAI1 , Proteína Quinase C beta , Estatísticas não Paramétricas
3.
J Invest Dermatol ; 143(1): 124-133.e3, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-35970476

RESUMO

Primary cutaneous large B-cell lymphoma, leg-type (PCLBCL-LT) is the most aggressive primary cutaneous B-cell lymphoma (PCBCL). Tumor microenvironment has a crucial role in tumor development, and tumor-infiltrating lymphocytes (TILs) can be targeted by immunotherapies. We characterized TILs in 20 PCBCLs to identify the tumor microenvironment features associated with clinical outcomes. We developed a seven‒multiplex immunofluorescence panel using Opal staining and image analysis using HALO software. In PCLBCL-LT, TILs were sparsely intermingled within tumor infiltrate in contrast to those in indolent PCBCL where TILs were scattered around tumor nodule edges with variable tumor infiltration. In PCLBCL-LT, TILs were composed of CD8 and CD4, whereas CD4 was predominant in indolent PCBCL. Proliferative TILs (CD3+Ki-67+ cells) were more abundant in PCLBCL-LT (P = 0.0036) than in indolent PCBCL. In PCLBCL-LT, proliferative TILs' abundance tended to be associated with better progression-free survival. These data were confirmed in a second independent cohort of 23 cases showing that proliferative TILs were more abundant in PCLBCL-LT (P = 0.0205) and that in PCLBCL-LT, high CD3+Ki-67+ cell density was associated with better progression-free survival (P = 0.002). These distinct TILs composition and distribution among PCBCL suggest that proliferative T lymphocytes represent a good prognostic factor in PCLBCL-LT and that stimulating their functions may represent a therapeutic approach.


Assuntos
Linfoma de Células B , Neoplasias Cutâneas , Humanos , Linfócitos do Interstício Tumoral , Neoplasias Cutâneas/patologia , Antígeno Ki-67 , Microambiente Tumoral , Prognóstico
4.
Anticancer Drugs ; 22(6): 481-7, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21317766

RESUMO

Death receptors play a crucial role in immune surveillance and cellular homeostasis, two processes circumvented by tumor cells. CD95 (also termed Fas or APO1) is a transmembrane receptor, which belongs to the tumor necrosis factor receptor superfamily, and induces a potent apoptotic signal. Initial steps of the CD95 signal take place through protein/protein interactions that bring zymogens such as caspase-8 and caspase-10 closer. Aggregation of these procaspases leads to their autoprocessing, to the release of activated caspases in the cytosol, which causes a caspase cascade, and to the transmission of the apoptotic signal. In parallel, CD95 engagement drives an increase in the intracellular calcium concentration (Ca(2+))i whose origin and functions remain controversial. Although Ca(2+) ions play a central role in apoptosis/necrosis induction, recent studies have highlighted a protective role of Ca(2+) in death receptor signaling. In the light of these findings, we discuss the role of Ca(2+) ions as modulators of CD95 signaling.


Assuntos
Cálcio/fisiologia , Transdução de Sinais/fisiologia , Receptor fas/fisiologia , Apoptose/efeitos dos fármacos , Caspases/metabolismo , Humanos , Receptores de Morte Celular/efeitos dos fármacos
5.
Front Oncol ; 11: 775253, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34765562

RESUMO

Sézary syndrome (SS) is an aggressive leukemic variant of cutaneous T-cell lymphomas (CTCL) in which the human Telomerase Reverse Transcriptase (hTERT) gene is re-expressed. Current available treatments do not provide long-term response. We previously reported that Histone deacetylase inhibitors (HDACi, romidespin and vorinostat) and a DNA methyltransferase inhibitor (DNMTi, 5-azacytidine) can reduce hTERT expression without altering the methylation level of hTERT promoter. Romidepsin and vorinostat are approved for CTCL treatment, while 5-azacytidine is approved for the treatment of several hematological disorders, but not for CTCL. Here, using the soft agar assay, we analyzed the functional effect of the aforementioned epidrugs on the clonogenic capacities of Sézary cells. Our data revealed that, besides hTERT downregulation, epidrugs' pressure reduced the proliferative and the tumor formation capacities in Sézary cells in vitro.

6.
Blood Adv ; 5(23): 5372-5386, 2021 12 14.
Artigo em Inglês | MEDLINE | ID: mdl-34555842

RESUMO

Non-Hodgkin B-cell lymphomas (B-NHL) mainly develop within lymph nodes as aggregates of tumor cells densely packed with their surrounding microenvironment, creating a tumor niche specific to each lymphoma subtypes. In vitro preclinical models mimicking biomechanical forces, cellular microenvironment, and 3D organization of B-cell lymphomas remain scarce, while all these parameters are key determinants of lymphomagenesis and drug resistance. Using a microfluidic method based on cell encapsulation inside permeable, elastic, and hollow alginate microspheres, we developed a new tunable 3D model incorporating lymphoma B cells, extracellular matrix (ECM), and/or tonsil stromal cells (TSC). Under 3D confinement, lymphoma B cells were able to form cohesive spheroids resulting from overexpression of ECM components. Moreover, lymphoma B cells and TSC dynamically formed self-organized 3D spheroids favoring tumor cell growth. 3D culture induced resistance to the classical chemotherapeutic agent doxorubicin, but not to the BCL2 inhibitor ABT-199, identifying this approach as a relevant in vitro model to assess the activity of therapeutic agents in B-NHL. RNA-sequence analysis highlighted the synergy of 3D, ECM, and TSC in upregulating similar pathways in malignant B cells in vitro than those overexpressed in primary lymphoma B cells in situ. Finally, our 3D model including ECM and TSC allowed long-term in vitro survival of primary follicular lymphoma B cells. In conclusion, we propose a new high-throughput 3D model mimicking lymphoma tumor niche and making it possible to study the dynamic relationship between lymphoma B cells and their microenvironment and to screen new anti-cancer drugs.


Assuntos
Antineoplásicos , Linfoma de Células B , Linfoma não Hodgkin , Linfócitos B , Proliferação de Células , Humanos , Linfoma de Células B/tratamento farmacológico , Microambiente Tumoral
7.
Cancers (Basel) ; 11(3)2019 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-30832225

RESUMO

GA101/obinutuzumab is a novel type II anti-CD20 monoclonal antibody (mAb), which is more effective than rituximab (RTX) in preclinical and clinical studies when used in combination with chemotherapy. Ca2+ signaling was shown to play a role in RTX-induced cell death. This report concerns the effect of GA101 on Ca2+ signaling and its involvement in the direct cell death induced by GA101. We reveal that GA101 triggered an intracellular Ca2+ increase by mobilizing intracellular Ca2+ stores and activating Orai1-dependent Ca2+ influx in non-Hodgkin lymphoma cell lines and primary B-Cell Chronic Lymphocytic Leukemia (B-CLL) cells. According to the cell type, Ca2+ was mobilized from two distinct intracellular compartments. In Raji, BL2, and B-CLL cells, GA101 induced a Ca2+ release from lysosomes, leading to the subsequent lysosomal membrane permeabilization and cell death. Inhibition of this calcium signaling reduced GA101-induced cell death in these cells. In SU-DHL-4 cells, GA101 mobilized Ca2+ from the endoplasmic reticulum (ER). Inhibition of ER replenishment, by blocking Orai1-dependent Ca2+ influx, led to an ER stress and unfolded protein response (UPR) which sensitized these cells to GA101-induced cell death. These results revealed the central role of Ca2+ signaling in GA101's action mechanism, which may contribute to designing new rational drug combinations improving its clinical efficacy.

8.
Cancers (Basel) ; 10(11)2018 Oct 26.
Artigo em Inglês | MEDLINE | ID: mdl-30373149

RESUMO

Ca2+ release-activated Ca2+ channels, composed of Orai1 and STIM1 (stromal interaction molecule 1) proteins, are the main Ca2+ entry mechanism in lymphocytes. Their role in cell migration and metastasis is demonstrated in solid cancers but it remains elusive in malignant hemopathies. Diffuse large B cell lymphoma (DLBCL) is characterized by the dissemination of neoplastic B cells throughout the organism which is under the control of chemokines such as Stromal Derived Factor 1 (SDF-1) and its receptor CXCR4. CXCR4 activation triggers a complex intracellular signaling including an increase in intracellular Ca2+ concentration whose role is still unclear. Using pharmacological and genetic approaches, we revealed that STIM1 and Orai1 were responsible for Ca2+ influx induced by SDF-1. Furthermore, we provide in vitro and in vivo evidence that they are necessary for basal or SDF-1-induced DLBCL cell migration which is independent of Ca2+ entry. We identify that they act as effectors coupling RhoA and ROCK dependent signaling pathway to MLC2 phosphorylation and actin polymerization. Finally, we revealed an alteration of Orai1 and STIM1 expression in extra-nodal DLBCL. Thus, we discovered a novel Ca2+-independent but Orai1 and STIM1-dependent signaling pathway involved in basal and CXCR4 dependent cell migration, which could be relevant for DLBCL physiopathology.

10.
Endocrinology ; 148(6): 2698-707, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17363452

RESUMO

Cessation of lactation causes a massive loss of surplus lactotrophs in the rat pituitary gland. The factors and mechanisms involved in this phenomenon have not yet been elucidated. Besides its inhibitory control on prolactin secretion and lactotroph proliferation, evidence suggests that dopamine (DA) may be a proapoptotic factor for lactotrophs. We therefore tested the proapoptotic effect of DA on pituitary glands from virgin, lactating, and postlactating rats. By measuring mitochondrial membrane potential loss, caspase-3 activation, and nuclear fragmentation, we show that DA induces apoptosis specifically in lactotrophs from postlactating rats. We then determined that this effect was partly mediated by the DA transporter (DAT) rather than the D(2) receptor, as corroborated by the detection of DAT expression exclusively in lactotrophs from postlactating rats. We also observed tyrosine hydroxylase (TH) expression in postlactating lactotrophs that was accompanied by an increase in DA content in the anterior pituitary gland of postlactating compared with virgin rats. Finally, we observed that cells expressing TH coexpressed DAT and cleaved caspase-3. These findings show that DA may play a role in lactotroph regression during the postlactation period by inducing apoptosis. The fact that this process requires DAT and TH expression by lactotrophs themselves suggests that it may be "autocrine" in nature.


Assuntos
Apoptose/efeitos dos fármacos , Proteínas da Membrana Plasmática de Transporte de Dopamina/genética , Dopamina/farmacologia , Lactação/efeitos dos fármacos , Lactotrofos/metabolismo , Tirosina 3-Mono-Oxigenase/genética , Animais , Caspase 3/metabolismo , Células Cultivadas , Proteínas da Membrana Plasmática de Transporte de Dopamina/metabolismo , Proteínas da Membrana Plasmática de Transporte de Dopamina/fisiologia , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Lactação/genética , Lactação/metabolismo , Modelos Biológicos , Adeno-Hipófise/efeitos dos fármacos , Adeno-Hipófise/enzimologia , Adeno-Hipófise/metabolismo , Ratos , Ratos Sprague-Dawley , Tirosina 3-Mono-Oxigenase/metabolismo
11.
Clin Cancer Res ; 20(10): 2663-73, 2014 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-24637636

RESUMO

PURPOSE: Despite therapeutic advances, non-Hodgkin lymphomas (NHL) remain incurable. They form a group of neoplasms strongly dependent on their inflammatory microenvironment, which plays an important supportive role in tumor B-cell survival and in the resistance to antitumor immune response. New therapies must consider both tumor cells and their surrounding microenvironment EXPERIMENTAL DESIGN: Stromal cells, derived from bone marrow or lymph nodes, and B cells from follicular lymphoma patients were cocultured or cultured alone with celecoxib treatment, a nonsteroidal anti-inflammatory drug, and/or TRAIL, a promising cytotoxic molecule for cancer therapy. RESULTS: In this study, we show that follicular lymphoma stromal cells produce large amounts of PGE2. This production is abrogated after celecoxib treatment, targeting the COX-2 isoenzyme involved in PGE2 synthesis. Furthermore, we demonstrate that celecoxib increases apoptosis in NHL B-cell lines and in primary follicular lymphoma B cells cocultured with stromal cells, but independently of the PGE2/COX-2 axis. Finally, celecoxib increases the apoptotic activity of TRAIL. We provide evidence that celecoxib affects proliferation and sensitizes NHL B-cell lines to apoptosis through COX-2-independent effects by slowing down the cell cycle and decreasing the expression of survival proteins, such as Mcl-1. CONCLUSIONS: These data suggest new potent strategies for NHL therapy combining drugs targeting both tumor B cells and survival signals provided by the tumor microenvironment.


Assuntos
Apoptose/efeitos dos fármacos , Ciclo-Oxigenase 2/metabolismo , Pirazóis/farmacologia , Sulfonamidas/farmacologia , Ligante Indutor de Apoptose Relacionado a TNF/farmacologia , Western Blotting , Celecoxib , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Células Cultivadas , Técnicas de Cocultura , Ciclo-Oxigenase 2/genética , Inibidores de Ciclo-Oxigenase 2/farmacologia , Dinoprostona/metabolismo , Sinergismo Farmacológico , Citometria de Fluxo , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Linfoma de Células B/genética , Linfoma de Células B/metabolismo , Linfoma de Células B/patologia , Células-Tronco Mesenquimais/efeitos dos fármacos , Células-Tronco Mesenquimais/metabolismo , Proteína de Sequência 1 de Leucemia de Células Mieloides/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Tempo , Células Tumorais Cultivadas , Proteína X Associada a bcl-2/metabolismo
12.
PLoS One ; 8(11): e80741, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24260468

RESUMO

Soft tissue sarcomas (STS) are rare, complex tumors with a poor prognosis. The identification of new prognostic biomarkers is needed to improve patient management. Our aim was to determine the methylation status of the 118 CpG sites in the PLAGL1 tumor-suppressor gene P1 CpG island promoter and study the potential prognostic impact of PLAGL1 promoter methylation CpG sites in STS. Training cohorts constituted of 28 undifferentiated sarcomas (US) and 35 leiomyosarcomas (LMS) were studied. PLAGL1 mRNA expression was investigated by microarray analysis and validated by RT-qPCR. Pyrosequencing was used to analyze quantitative methylation of the PLAGL1 promoter. Associations between global promoter or specific CpG site methylation and mRNA expression were evaluated using Pearson's product moment correlation coefficient. Cox univariate and multivariate proportional hazard models were used to assess the predictive power of CpG site methylation status. Sixteen CpG sites associated with PLAGL1 mRNA expression were identified in US and 6 in LMS. Statistical analyses revealed an association between CpG107 methylation status and both overall and metastasis-free survival in US, which was confirmed in a validation cohort of 37 US. The exhaustive study of P1 PLAGL1 promoter methylation identified a specific CpG site methylation correlated with mRNA expression, which was predictive for both metastasis-free and overall survival and may constitute the first US-specific biomarker. Such a biomarker may be relevant for identifying patients likely to derive greater benefit from treatment.


Assuntos
Proteínas de Ciclo Celular/genética , Ilhas de CpG , Metilação de DNA , Sarcoma/genética , Fatores de Transcrição/genética , Proteínas Supressoras de Tumor/genética , Adulto , Idoso , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Leiomiossarcoma/genética , Leiomiossarcoma/patologia , Masculino , Pessoa de Meia-Idade , Gradação de Tumores , Prognóstico , Regiões Promotoras Genéticas , RNA Mensageiro/genética , Sarcoma/mortalidade , Sarcoma/patologia , Carga Tumoral
13.
Neuroendocrinology ; 83(2): 77-88, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16785746

RESUMO

Besides its physiological role as a neurotransmitter, dopamine (DA) induces apoptosis in the central nervous system. This effect is mediated partly by the DA transporter (DAT) and involves reactive oxygen species (ROS) formation as well as oxidative stress. In the pituitary, the inhibitory control by DA of prolactin release and synthesis and lactotrope cell proliferation is well known, while the pro-apoptotic effect of DA remains unclear. Our aim was to study the pro-apoptotic effect of DA in the GH3 mammosomatotrope cell line and determine the DA mechanism that leads to apoptosis in these cells. Using flow cytometry, Western blot, and confocal microscopy, we showed for the first time that DA induced: (1) loss of mitochondrial potential; (2) relocation of Bax to the mitochondria; (3) cytochrome c release; (4) caspase-3 activation, and (5) nuclear fragmentation, resulting in apoptosis. We observed that DAT was expressed in GH3 cells and participated in the DA effect, as apoptosis was significantly reversed in the presence of DAT inhibitors. Direct measurement showed that DA rapidly increased the formation of intracellular ROS. The antioxidant N-acetyl-L-cysteine (NAC) effectively blocked DA-induced ROS formation and apoptosis. Neither JNK nor p38 were involved in this process, so we suggest that the mitochondrial pore of transition is the likely target of the ROS generated by DA. These data provide the first evidence that DA triggers apoptosis in pituitary cells via a mechanism involving DAT and oxidative stress. These findings may be particularly relevant in understanding lactotrope apoptosis during postnatal life.


Assuntos
Apoptose/efeitos dos fármacos , Dopamina/farmacologia , Hipófise/citologia , Transdução de Sinais/efeitos dos fármacos , Animais , Caspase 3 , Caspases/metabolismo , Contagem de Células/métodos , Linhagem Celular , Citocromos/metabolismo , Proteínas da Membrana Plasmática de Transporte de Dopamina/genética , Proteínas da Membrana Plasmática de Transporte de Dopamina/metabolismo , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/farmacologia , Citometria de Fluxo/métodos , Proteínas de Fluorescência Verde/metabolismo , Marcação In Situ das Extremidades Cortadas/métodos , Potenciais da Membrana/efeitos dos fármacos , Microscopia Confocal/métodos , Mitocôndrias/efeitos dos fármacos , Ratos , Espécies Reativas de Oxigênio/metabolismo , Transfecção/métodos , Proteína X Associada a bcl-2/metabolismo
14.
Am J Physiol Cell Physiol ; 284(4): C1054-64, 2003 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-12620897

RESUMO

Previously, we showed that the peak density of the transient outward K(+) current (I(to)) expressed in GH3 cells was different in the S phase than in other phases of the cell cycle. Using cell synchronization, we show here that I(to) drops precisely at the quiescent (G(0) phase)/proliferating transition. This change is not due to a modification in the voltage dependence of I(to), but rather to a modification in its inactivation kinetics. Molecular determination of K(+) channel subunits showed that I(to) required the expression of Kv1.4, Kv4.1, and Kv4.3. We found that the increase in I(to) density during the quiescent state was accompanied by an increase in Kv1.4 protein expression, whereas Kv4.3 expression remained unchanged. We further demonstrate that the link between I(to) expression and cell proliferation is not mediated by variations in cell excitability. These results provide new evidence for the cell cycle dependence of I(to) expression, which could be relevant in understanding the mechanisms leading to pituitary adenomas.


Assuntos
Hipófise/citologia , Hipófise/metabolismo , Canais de Potássio de Abertura Dependente da Tensão da Membrana , Canais de Potássio/metabolismo , Animais , Ciclo Celular , Divisão Celular/fisiologia , Linhagem Celular , Eletrofisiologia , Fase G1/fisiologia , Canal de Potássio Kv1.4 , Hipófise/fisiologia , Canais de Potássio/fisiologia , Fase de Repouso do Ciclo Celular/fisiologia , Canais de Potássio Shal , Fatores de Tempo
15.
Glia ; 38(3): 200-14, 2002 May.
Artigo em Inglês | MEDLINE | ID: mdl-11968058

RESUMO

Prolactin (PRL) has several physiological effects on peripheral tissues and the brain. This hormone acts via its membrane receptor (PRL-R) to induce cell differentiation or proliferation. Using reverse transcription-polymerase chain reaction (RT-PCR) combined with Southern blot analysis, we detected PRL-R transcripts in a human glioma cell line (U87-MG) and in primary cultured human glioblastoma cells. These transcripts were deleted or not in their extracellular domains. We examined the effects of PRL on intracellular free Ca2+ concentration ([Ca2+](i)) in these cells in order to improve our understanding of the PRL transduction mechanism, which is still poorly documented. [Ca2+](i) was measured by microspectrofluorimetry using indo-1 as the Ca2+ fluorescent probe. Spatiotemporal aspects of PRL-induced Ca2+ signals were investigated using high-speed fluo-3 confocal imaging. We found that physiological concentrations (0.4-4 nM) of PRL-stimulated Ca2+ entry and intracellular Ca2+ mobilization via a tyrosine kinase-dependent mechanism. The two types of Ca2+ responses observed were distinguishable by their kinetics: one showing a slow (type I) and the other a fast (type II) increase in [Ca2+](i). The amplitude of PRL-induced Ca2+ increases may be sufficient to provoke several physiological responses, such as stimulating proliferation. Furthermore, PRL induced a dose-dependent increase in [3H]thymidine incorporation levels and in cellular growth and survival, detected by the MTT method. These data indicate that PRL induced mitogenesis of human glioma cells.


Assuntos
Neoplasias Encefálicas/metabolismo , Sinalização do Cálcio/fisiologia , Cálcio/metabolismo , Divisão Celular/fisiologia , Glioma/metabolismo , Líquido Intracelular/metabolismo , Prolactina/metabolismo , Proteínas Proto-Oncogênicas , Receptores da Prolactina/metabolismo , Sinalização do Cálcio/efeitos dos fármacos , ATPases Transportadoras de Cálcio/antagonistas & inibidores , ATPases Transportadoras de Cálcio/metabolismo , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/fisiologia , Divisão Celular/efeitos dos fármacos , Núcleo Celular/efeitos dos fármacos , Núcleo Celular/metabolismo , Citosol/efeitos dos fármacos , Citosol/metabolismo , DNA/biossíntese , DNA/efeitos dos fármacos , Relação Dose-Resposta a Droga , Espaço Extracelular/efeitos dos fármacos , Espaço Extracelular/metabolismo , Feminino , Humanos , Líquido Intracelular/efeitos dos fármacos , Janus Quinase 2 , Masculino , Prolactina/farmacologia , Proteínas Tirosina Quinases/antagonistas & inibidores , Proteínas Tirosina Quinases/metabolismo , RNA Mensageiro/metabolismo , Receptores da Prolactina/agonistas , Receptores da Prolactina/genética , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA