Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 99
Filtrar
1.
Front Cell Dev Biol ; 12: 1399092, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38903530

RESUMO

Introduction: Previous publications have shown that STIM1, ORAI1, and KDM2B, are implicated in Ca2+ signaling and are highly expressed in various cancer subtypes including prostate cancer. They play multiple roles in cancer cell migration, invasion, and metastasis. In the current study we investigated the expression of the above biomarkers in circulating tumor cells from patients with metastatic prostate cancer. Methods: Thirty-two patients were enrolled in this study and CTCs' isolation was performed with Ficoll density gradient. Two different triple immunofluorescence stainings were conducted with the following combination of antibodies: CK/KDM2B/CD45 and CK/STIM1/ORAI1. Slides were analyzed using VyCAP microscopy technology. Results: CTC-positive patients were detected in 41% for (CK/KDM2B/CD45) staining and in 56% for (CK/STIM1/ORAI1) staining. The (CK+/KDM2B+/CD45-) and the (CK+/STIM1+/ORAI1+) were the most frequent phenotypes as they were detected in 85% and 94% of the CTC-positive patients, respectively. Furthermore, the expression of ORAI1 and STIM1 in patients' PBMCs was very low exhibiting them as interesting specific biomarkers for CTC detection. The (CK+/STIM1+/ORAI1+) phenotype was correlated to bone metastasis (p = 0.034), while the (CK+/STIM1+/ORAI1-) to disease relapse (p = 0.049). Discussion: STIM1, ORAI1, and KDM2B were overexpressed in CTCs from patients with metastatic prostate cancer. STIM1 and ORAI1 expression was related to disease recurrence and bone metastasis. Further investigation of these biomarkers in a larger cohort of patients will clarify their clinical significance for prostate cancer patients.

2.
Saudi Pharm J ; 32(7): 102109, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38817821

RESUMO

KDM2B, a histone lysine demethylase, is expressed in a plethora of cancers. Earlier studies from our group, have showcased that overexpression of KDM2B in the human prostate cancer cell line DU-145 is associated with cell adhesion, actin reorganization, and improved cancer cell migration. In addition, we have previously examined changes of cytosolic Ca2+, regulated by the pore-forming proteins ORAI and the Ca2+ sensing stromal interaction molecules (STIM), via store-operated Ca2+ entry (SOCE) in wild-type DU-145. This study sought to evaluate the impact of KDM2B overexpression on the expression of key molecules (SGK1, Nhe1, Orai1, Stim1) and SOCE. Furthermore, this is the first study to evaluate KDM2B expression in circulating tumor cells (CTCs) from patients with prostate cancer. mRNA levels for SGK1, Nhe1, Orai1, and Stim1 were quantified by RT-PCR. Calcium signals were measured in KDM2B-overexpressing DU-145 cells, loaded with Fura-2. Blood samples from 22 prostate cancer cases were scrutinized for KDM2B expression using immunofluorescence staining and the VyCAP system. KDM2B overexpression in DU-145 cells increased Orai1, Stim1, and Nhe1 mRNA levels and significantly decreased Ca2+ release. KDM2B expression was examined in 22 prostate cancer patients. CTCs were identified in 45 % of these patients. 80 % of the cytokeratin (CK)-positive patients and 63 % of the total examined CTCs exhibited the (CK + KDM2B + CD45-) phenotype. To conclude, this study is the first to report increased expression of KDM2B in CTCs from patients with prostate cancer, bridging in vitro and preclinical assessments on the potentially crucial role of KDM2B on migration, invasiveness, and ultimately metastasis in prostate cancer.

3.
PeerJ ; 11: e16074, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37744224

RESUMO

Background: The purpose of this study is to analyzed the involvement of chorein in microtubules organization of three types of malignant; rhabdomyosarcoma tumor cells (ZF), rhabdomyosarcoma cells (RH30), and rhabdomyosarcoma cells (RD). ZF are expressing high chorein levels. Previous studies revealed that chorein protein silencing in ZF tumor cells persuaded apoptotic response followed by cell death. In addition, in numerous malignant and non-malignant cells this protein regulates actin cytoskeleton structure and cellular signaling. However, the function of chorein protein in microtubular organization is yet to be established. Methods: In a current research study, we analyzed the involvement of chorein in microtubules organization by using three types of malignant rhabdomyosarcoma cells. We have applied confocal laser-scanning microscopy to analyze microtubules structure and RT-PCR to examine cytoskeletal gene transcription. Results: We report here that in rhabdomyosarcoma cells (RH30), chorein silencing induced disarrangement of microtubular network. This was documented by laser scanning microscopy and further quantified by FACS analysis. Interestingly and in agreement with previous reports, tubulin gene transcription in RH cells was unchanged upon silencing of chorein protein. Equally, confocal analysis showed minor disordered microtubules organization with evidently weakened staining in rhabdomyosarcoma cells (RD and ZF) after silencing of chorein protein. Conclusion: These results disclose that chorein silencing induces considerable structural disorganization of tubulin network in RH30 human rhabdomyosarcoma tumor cells. Additional studies are now needed to establish the role of chorein in regulating cytoskeleton architecture in tumor cells.


Assuntos
Rabdomiossarcoma , Tubulina (Proteína) , Proteínas de Transporte Vesicular , Humanos , Citoesqueleto de Actina , Citoesqueleto/genética , Microtúbulos , Rabdomiossarcoma/genética , Linhagem Celular Tumoral , Proteínas de Transporte Vesicular/genética
4.
J Pers Med ; 12(2)2022 Jan 25.
Artigo em Inglês | MEDLINE | ID: mdl-35207643

RESUMO

Upregulation of Vimentin (VIM), alpha-Tubulin (TUB) and Detyrosinated tubulin (GLU) in circulating tumor cells (CTCs) derived from breast cancer patients is related to poor prognosis. In the current study we evaluated for the first time, these cytoskeletal proteins in sixty Non-Small Cell Lung Cancer (NSCLC) patients' CTCs (33 treatment-naïve and 27 pre-treated). Samples were isolated using the ISET platform and stained with a pancytokeratin (CK)/CD45/TUB, CK/GLU/VIM and CK/programmed death ligand 1 (PD-L1) combination of antibodies. Subsequently, slides were analyzed using confocal laser scanning microscopy. CTCs were detected in 86.7% of the patients. CTCs with TUB expression were identified in 65.4% (34/52) of the CK (+)-patients. GLU, VIM and PD-L1 were also evaluated. The frequency of the observed phenotypes was as follow: (CK+/GLU-/VIM-): 35.2%, (CK+/GLU+/VIM+): 63.0%, (CK+/GLU+/VIM-): 16.7%, (CK+/GLU-/VIM+): 72.2%, (CK+/PD-L1-): 75% and (CK+/PD-L1+): 55%. The OS was significantly decreased in patients with high GLU (3.8 vs. 7.9 months; p = 0.018) and/or high VIM (3.2 vs. 7.1 months; p = 0.029) expression in their CTCs. PD-L1 was also related to OS (3.4 vs. 7.21 months; p = 0.035). Moreover, TUB-high and TUB-low expression in CTCs inversely influenced patients' OS as independent prognostic factors (p = 0.041 and p = 0.009). The current study revealed that TUB, GLU, VIM and PD-L1 were overexpressed in CTCs from NSCLC patients. Furthermore, the presence of GLU, VIM-positive and PD-L1 in CTCs is potentially related to patients' outcomes.

5.
Environ Sci Pollut Res Int ; 28(6): 7379-7389, 2021 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-33030691

RESUMO

Metal nanomaterials such as bismuth oxide nanoparticles (Bi2O3NPs) have been extensively used in cosmetics, dental materials, pulp capping, and biomedical imaging. There is little knowledge about the health risk of Bi2O3NPs in humans, which warrants a thorough toxicity investigation of Bi2O3NPs at the cellular level. In this experiment, we investigated the cytotoxic effect of Bi2O3NPs on human breast cancer (MCF-7) cells over 24 and 48 h. MCF-7 cells were exposed to Bi2O3NPs at varying doses (0.1, 0.5, 1.0, 5, 10, 20, 40 µg/mL) for 24 and 48 h. We assessed the toxicity of Bi2O3NPs by measuring its effect on the viability and oxidative stress biomarkers, e.g., GSH, SOD, and catalase in MCF-7 cells. The pro-apoptotic effects of Bi2O3NPs on MCF-7 cells were determined via evaluating dysfunction of mitochondrial membrane potential (MMP), caspase-3 activity, externalization of phosphatidylserine, and chromosome condensation. Furthermore, apoptotic cells were evaluated using 7-AAD fluorescence stain and Annexin V-FITC. Bi2O3NPs induced oxidative stress in MCF-7 cells in a time- and dose-dependent manner. Bi2O3NPs increased the rate of both necrotic cells and apoptotic cells. In addition, the blue fluorescence of MCF-7 cells with condensed chromatin was increased in a time- and dose-dependent manner. In conclusion, the present study highlights the potential toxic effects of Bi2O3NPs at the cellular level and suggests further investigation of Bi2O3NPs before any medical purposes.


Assuntos
Neoplasias da Mama , Nanopartículas , Apoptose , Bismuto , Humanos , Células MCF-7 , Estresse Oxidativo , Espécies Reativas de Oxigênio
6.
Mol Med Rep ; 22(4): 3066-3072, 2020 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-32945485

RESUMO

Human cytomegalovirus (HCMV) is a prevalent viral pathogen, which can cause severe clinical consequences in neonates, immunocompromised individuals, patients with AIDS, and organ and stem cell transplant recipients. HCMV inhibits the host cell cycle progress while the immediate­early protein 1 (IE1) tethers to condensed chromatin in mitotic cells. The present study investigated the effect of HCMV on the cell cycle in human glioblastoma cells, as well as the role of RhoA GTPase during mitosis in the same context. Live cell microscopy showed that despite the apparent cell cycle arrest at late stages of mitosis in normal fibroblasts, HCMV­infected U373MG cells successfully went through all stages of cell division. HCMV IE1 protein exhibited a remarkably tight association with mitotic chromosomes from early mitosis to late cytokinesis. Depletion of RhoA significantly impaired the proliferation rate of HCMV­infected U373MG cells; consistent with this observation, the number of cells entering mitosis was also decreased. These results demonstrated the differential behavior of HCMV during mitosis in a normal and a cancer background. Furthermore, RhoA may be a critical component for the efficient cell division of HCMV­infected glioblastoma cells, which subsequently ensures the maintenance of viral genomes.


Assuntos
Neoplasias Encefálicas/virologia , Infecções por Citomegalovirus/metabolismo , Citomegalovirus/fisiologia , Glioblastoma/virologia , Proteínas Imediatamente Precoces/metabolismo , Proteína rhoA de Ligação ao GTP/metabolismo , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Pontos de Checagem do Ciclo Celular , Linhagem Celular Tumoral , Proliferação de Células , Citocinese , Citomegalovirus/metabolismo , Infecções por Citomegalovirus/genética , Fibroblastos/citologia , Fibroblastos/metabolismo , Fibroblastos/virologia , Glioblastoma/genética , Glioblastoma/metabolismo , Humanos , Mitose , Replicação Viral , Proteína rhoA de Ligação ao GTP/genética
7.
Ther Adv Med Oncol ; 12: 1758835919895754, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32426042

RESUMO

BACKGROUND: The chemokine receptor CXCR4 and the transcription factor JUNB, expressed on a variety of tumor cells, seem to play an important role in the metastatic process. Since disseminated tumor cells (DTCs) in the bone marrow (BM) have been associated with worse outcomes, we evaluated the expression of CXCR4 and JUNB in DTCs of primary, nonmetastatic breast cancer (BC) patients before the onset of any systemic treatment. METHODS: Bilateral BM (10 ml) aspirations of 39 hormone receptor (HR)-positive, HER2-negative BC patients were assessed for the presence of DTCs using the following combination of antibodies: pan-cytokeratin (A45-B/B3)/CXCR4/JUNB. An expression pattern of the examined proteins was created using confocal laser scanning microscopy, Image J software and BC cell lines. RESULTS: CXCR4 was overexpressed in cancer cells and DTCs, with the following hierarchy of expression: SKBR3 > MCF7 > DTCs > MDA-MB231. Accordingly, the expression pattern of JUNB was: DTCs > MDA-MB231 > SKBR3 > MCF7. The mean intensity of CXCR4 (6411 ± 334) and JUNB (27725.64 ± 470) in DTCs was statistically higher compared with BM hematopoietic cells (2009 ± 456, p = 0.001; and 11112.89 ± 545, p = 0.001, respectively). The (CXCR4+JUNB+CK+) phenotype was the most frequently detected [90% (35/39)], followed by the (CXCR4-JUNB+CK+) phenotype [36% (14/39)]. However, (CXCR4+JUNB-CK+) tumor cells were found in only 5% (3/39) of patients. Those patients harboring DTCs with the (CXCR4+JUNB+CK+) phenotype revealed lower overall survival (Cox regression: p = 0.023). CONCLUSIONS: (CXCR4+JUNB+CK+)-expressing DTCs, detected frequently in the BM of BC patients, seem to identify a subgroup of patients at higher risk for relapse that may be considered for close follow up.

8.
Sci Rep ; 10(1): 8391, 2020 05 21.
Artigo em Inglês | MEDLINE | ID: mdl-32439941

RESUMO

Loss of function mutations of the chorein-encoding gene VPS13A lead to chorea-acanthocytosis (ChAc), a neurodegenerative disorder with accelerated suicidal neuronal cell death, which could be reversed by lithium. Chorein upregulates the serum and glucocorticoid inducible kinase SGK1. Targets of SGK1 include the Na+/K+-ATPase, a pump required for cell survival. To explore whether chorein-deficiency affects Na+/K+ pump capacity, cortical neurons were differentiated from iPSCs generated from fibroblasts of ChAc patients and healthy volunteers. Na+/K+ pump capacity was estimated from K+-induced whole cell outward current (pump capacity). As a result, the pump capacity was completely abolished in the presence of Na+/K+ pump-inhibitor ouabain (100 µM), was significantly smaller in ChAc neurons than in control neurons, and was significantly increased in ChAc neurons by lithium treatment (24 hours 2 mM). The effect of lithium was reversed by SGK1-inhibitor GSK650394 (24 h 10 µM). Transmembrane potential (Vm) was significantly less negative in ChAc neurons than in control neurons, and was significantly increased in ChAc neurons by lithium treatment (2 mM, 24 hours). The effect of lithium on Vm was virtually abrogated by ouabain. Na+/K+ α1-subunit transcript levels and protein abundance were significantly lower in ChAc neurons than in control neurons, an effect reversed by lithium treatment (2 mM, 24 hours). In conclusion, consequences of chorein deficiency in ChAc include impaired Na+/K+ pump capacity.


Assuntos
Membrana Celular/patologia , Neuroacantocitose/metabolismo , Neuroacantocitose/patologia , Neurônios/patologia , ATPase Trocadora de Sódio-Potássio/metabolismo , Benzoatos/farmacologia , Compostos Bicíclicos Heterocíclicos com Pontes/farmacologia , Estudos de Casos e Controles , Diferenciação Celular , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Células Cultivadas , Humanos , Proteínas Imediatamente Precoces/antagonistas & inibidores , Células-Tronco Pluripotentes Induzidas/citologia , Lítio/farmacologia , Potenciais da Membrana/efeitos dos fármacos , Neurônios/citologia , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Técnicas de Patch-Clamp , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , ATPase Trocadora de Sódio-Potássio/genética , Proteínas de Transporte Vesicular/genética , Proteínas de Transporte Vesicular/metabolismo
9.
Protein J ; 39(3): 232-239, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32270414

RESUMO

BACKGROUND: Lysine specific demethylase 2B, KDM2B, regulates genes that participate in cellular development, morphogenesis, differentiation and metabolism as a component of the polycomb repressive complex 1 (PRC1). The CxxC finger of KDM2B is responsible for the DNA binding capacity of this epigenetic regulator, acting as a sampling mechanism across chromatin for gene repression OBJECTIVES: The molecular determinants of the CxxC-DNA interaction remain largely unknown, revealing a significant knowledge gap to be explored. Our goal was to elucidate the key residues of the CxxC domain that contribute to its function as well as to further elaborate on the significance of this domain in the KDM2B role METHODS: By using electrophoresis mobility swift assay, we identified structural elements of CxxC domain that participate in the DNA recognition. We created mouse embryonic fibroblasts overexpressing different truncated and point-mutated mouse KDM2B variants to examine the contribution of the KDM2B domains in replicative senescence bypass RESULTS: In this study, we show that only the CxxC finger is essential for the ability of mKDM2B to bypass replicative senescence in primary cells by ink4A-Arf-ink4B locus repression, and that this is mediated by specific interactions of residues R585, K608 and K616 with non-methylated CpG containing DNA CONCLUSIONS: These results provide new structural insights into the molecular interactions of CxxC and could serve as a stepping-stone for developing domain-specific inhibitors for KDM2B.


Assuntos
Fator 1 de Ribosilação do ADP/metabolismo , Senescência Celular/genética , Inibidor de Quinase Dependente de Ciclina p15/metabolismo , Inibidor p16 de Quinase Dependente de Ciclina/metabolismo , DNA/química , Proteínas F-Box/química , Histona Desmetilases com o Domínio Jumonji/química , Fator 1 de Ribosilação do ADP/genética , Animais , Sítios de Ligação , Diferenciação Celular , Ilhas de CpG , Inibidor de Quinase Dependente de Ciclina p15/genética , Inibidor p16 de Quinase Dependente de Ciclina/genética , DNA/genética , DNA/metabolismo , Embrião de Mamíferos , Proteínas F-Box/genética , Proteínas F-Box/metabolismo , Fibroblastos/citologia , Fibroblastos/metabolismo , Regulação da Expressão Gênica , Histona Desmetilases com o Domínio Jumonji/genética , Histona Desmetilases com o Domínio Jumonji/metabolismo , Camundongos , Mutação Puntual , Cultura Primária de Células , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas , Transdução de Sinais
10.
Cancer Biol Ther ; 21(6): 533-540, 2020 06 02.
Artigo em Inglês | MEDLINE | ID: mdl-32175798

RESUMO

Recent studies revealed that the histone demethylase KDM2B regulates the epithelial markers E-Cadherin and ZO-1, the RhoA/B/C-small-GTPases and actin cytoskeleton organization, in DU-145 prostate- and HCT-116 colon-tumor cells. Here we addressed the role of KDM2B in the activation of Focal Adhesion Kinase (FAK)-signaling and its involvement in regulating tumor cell motility. We used RT-PCR for gene transcriptional analysis, Western blotting for the assessment of protein expression and activity and wound-healing assay for the study of cell migration. KDM2B overexpression or silencing controls the activity of FAK in DU-145 prostate- and HCT-116 colon-tumor cells without affecting gene transcription and protein expression of this kinase. Upon KDM2B overexpression in DU-145 cells, significantly enhanced migration was observed, which was abolished in cells pretreated by the specific phosphoinositide-3 kinase (PI3 K) inhibitor LY294002, implying involvement of FAK/PI3 K signaling in the migration process. In line with this, the p85-PI3 K-subunit was downregulated upon knockdown of KDM2B in DU-145 cells, while the opposite effect became evident in KDM2B-overexpressing cells. These results revealed a novel functional role of KDM2B in regulating the activation of the FAK/PI3 K signaling in prostate cancer cells that participates in the control of cell motility.


Assuntos
Movimento Celular , Neoplasias do Colo/patologia , Proteínas F-Box/metabolismo , Quinase 2 de Adesão Focal/metabolismo , Regulação Neoplásica da Expressão Gênica , Histona Desmetilases com o Domínio Jumonji/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Neoplasias da Próstata/patologia , Apoptose , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Proliferação de Células , Neoplasias do Colo/genética , Neoplasias do Colo/metabolismo , Proteínas F-Box/genética , Quinase 2 de Adesão Focal/genética , Humanos , Histona Desmetilases com o Domínio Jumonji/genética , Masculino , Fosfatidilinositol 3-Quinases/genética , Neoplasias da Próstata/genética , Neoplasias da Próstata/metabolismo , Células Tumorais Cultivadas
11.
Cell Signal ; 70: 109599, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32165237

RESUMO

Human cytomegalovirus (HCMV) is a ubiquitous pathogen which periodically reactivates, causing severe clinical consequences in immunosuppressed patients, organ and stem cell transplant recipients or newborn babies with congenital infections. HCMV infection stimulates the expression of several proinflammatory cytokines, which may contribute to the pathogenesis of the infection. Rho GTPases mediate cytokine expression while increasing evidence implicates them in important aspects of HCMV life cycle. Here, we studied the role of RhoA on the interleukin 11 (IL-11) release in HCMV-infected fibroblasts. Human fibroblasts, either endogenously expressing or silenced for RhoA, were infected by HCMV or UV-inactivated virus and IL-11 transcription and secretion were evaluated. We found that HCMV lytic infection increased the IL-11 levels, both in terms of transcription and translation. Both infectious and non-infectious HCMV particles were able to induce the IL-11 production. The depletion of RhoA resulted in an even higher release of IL-11, revealing the implication of this specific Rho isoform in this biological event. Finally, infection of cells in the presence of the HCMV DNA replication inhibitor, ganciclovir, significantly reduced the secretion of IL-11, strongly associating its induction with active viral DNA replication. Collectively, these data demonstrate, for the first time, a novel role of RhoA GTPase during HCMV lytic infection, regulating the activation of an immune response through IL-11.


Assuntos
Infecções por Citomegalovirus/imunologia , Citomegalovirus , Fibroblastos/metabolismo , Interleucina-11/imunologia , Proteína rhoA de Ligação ao GTP/imunologia , Citomegalovirus/imunologia , Citomegalovirus/fisiologia , Fibroblastos/patologia , Células HEK293 , Humanos
12.
Breast Cancer Res ; 21(1): 86, 2019 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-31370904

RESUMO

BACKGROUND: Circulating tumor cells (CTCs) are important for metastatic dissemination of cancer. They can provide useful information, regarding biological features and tumor heterogeneity; however, their detection and characterization are difficult due to their limited number in the bloodstream and their mesenchymal characteristics. Therefore, new biomarkers are needed to address these questions. METHODS: Bioinformatics functional enrichment analysis revealed a subgroup of 24 genes, potentially overexpressed in CTCs. Among these genes, the chemokine receptor CXCR4 plays a central role. After prioritization according to the CXCR4 corresponding pathways, five molecules (JUNB, YWHAB, TYROBP, NFYA, and PRDX1) were selected for further analysis in biological samples. The SKBR3, MDA-MB231, and MCF7 cell lines, as well as PBMCs from normal (n = 10) blood donors, were used as controls to define the expression pattern of all the examined molecules. Consequently, 100 previously untreated metastatic breast cancer (mBC) patients (n = 100) were analyzed using the following combinations of antibodies: CK (cytokeratin)/CXCR4/JUNB, CK/NFYA/ΥWHΑΒ (14-3-3), and CK/TYROBP/PRDX1. A threshold value for every molecule was considered the mean expression in normal PBMCs. RESULTS: Quantification of CXCR4 revealed overexpression of the receptor in SKBR3 and in CTCs, following the subsequent scale (SKBR3>CTCs>Hela>MCF7>MDA-MB231). JUNB was also overexpressed in CTCs (SKBR3>CTCs>MCF7>MDA-MB231>Hela). According to the defined threshold for each molecule, CXCR4-positive CTCs were identified in 90% of the patients with detectable tumor cells in their blood. In addition, 65%, 75%, 14.3%, and 12.5% of the patients harbored JUNB-, TYROBP-, NFYA-, and PRDX-positive CTCs, respectively. Conversely, none of the patients revealed YWHAB-positive CTCs. Interestingly, JUNB expression in CTCs was phenotypically and statistically enhanced compared to patients' blood cells (p = 0.002) providing a possible new biomarker for CTCs. Furthermore, the detection of JUNB-positive CTCs in patients was associated with poorer PFS (p = 0.015) and OS (p = 0.002). Moreover, JUNB staining of 11 primary and 4 metastatic tumors from the same cohort of patients revealed a dramatic increase of JUNB expression in metastasis. CONCLUSIONS: CXCR4, JUNB, and TYROBP were overexpressed in CTCs, but only the expression of JUNB was associated with poor prognosis, providing a new biomarker and a potential therapeutic target for the elimination of CTCs.


Assuntos
Biomarcadores Tumorais , Neoplasias da Mama/genética , Neoplasias da Mama/mortalidade , Fatores de Transcrição/genética , Idoso , Idoso de 80 Anos ou mais , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Biologia Computacional/métodos , Feminino , Perfilação da Expressão Gênica , Humanos , Gradação de Tumores , Estadiamento de Neoplasias , Fenótipo , Prognóstico , Receptores CXCR4/genética , Receptores CXCR4/metabolismo , Análise de Sobrevida , Fatores de Transcrição/metabolismo , Transcriptoma
13.
FEBS Lett ; 593(15): 1879-1900, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31199502

RESUMO

Fibroblast growth factor 23 (FGF23) is mainly produced in the bone and, upon secretion, forms a complex with a FGF receptor and coreceptor αKlotho. FGF23 can exert several endocrine functions, such as inhibiting renal phosphate reabsorption and 1,25-dihydroxyvitamin D3 production. Moreover, it has paracrine activities on several cell types, including neutrophils and hepatocytes. Klotho and Fgf23 deficiencies result in pathologies otherwise encountered in age-associated diseases, mainly as a result of hyperphosphataemia-dependent calcification. FGF23 levels are also perturbed in the plasma of patients with several disorders, including kidney or cardiovascular diseases. Here, we review mechanisms controlling FGF23 production and discuss how FGF23 regulation is perturbed in disease.


Assuntos
Osso e Ossos/metabolismo , Doenças Cardiovasculares/metabolismo , Fatores de Crescimento de Fibroblastos/metabolismo , Nefropatias/metabolismo , Animais , Calcitriol/metabolismo , Doenças Cardiovasculares/genética , Fator de Crescimento de Fibroblastos 23 , Fatores de Crescimento de Fibroblastos/genética , Regulação da Expressão Gênica , Glucuronidase/metabolismo , Humanos , Nefropatias/genética , Proteínas Klotho , Comunicação Parácrina , Fosfatos/metabolismo , Receptores de Fatores de Crescimento de Fibroblastos/metabolismo
14.
Biochem Biophys Res Commun ; 512(3): 467-472, 2019 05 07.
Artigo em Inglês | MEDLINE | ID: mdl-30902388

RESUMO

Placental growth factor (PlGF) is produced by tumor cells and stimulates tumor growth and metastasis in part by upregulation of hypoxia inducible factor HIF1α. Orchestration of tumor cell proliferation and migration involves oscillations of cytosolic Ca2+ activity ([Ca2+]i). The [Ca2+]i oscillations could be accomplished by triggering of intracellular Ca2+ release followed by store-operated Ca2+-entry (SOCE). Mechanisms accomplishing SOCE include the pore-forming ion channel unit Orai1 and its regulator STIM1. The present study explored whether PlGF influences the expression of Orai1 and STIM1, as well as SOCE and whether this effect impacts on HIF1α expression. To this end, ovary carcinoma cells were cultured for 24 h without and with PlGF (10 ng/ml). Orai1, STIM1 and HIF1α transcript levels were quantified utilizing RT-PCR and Orai1, STIM1 and HIF1α protein levels by Western blotting. [Ca2+]i was estimated from Fura-2-fluorescence and SOCE from increase of [Ca2+]i following Ca2+ re-addition after Ca2+-store depletion with extracellular Ca2+ removal and sarcoendoplasmatic Ca2+-ATPase (SERCA) inhibitor thapsigargin (1 µM). As a result, exposure of ovary carcinoma cells to PlGF was followed by a significant increase of Orai1 as well as STIM1 transcript and protein levels. PlGF significantly increased store-operated Ca2+-entry following re-addition of extracellular Ca2+, an effect virtually abrogated by Orai1 inhibitor MRS1845 (10 µM). PlGF further increased HIF1α transcript and protein levels, an effect again significantly blunted by MRS1845 (10 µM). In conclusion, PlGF upregulates expression of both, Orai1 and STIM1 thus enhancing store-operated Ca2+-entry with subsequent upregulation of HIF1α.


Assuntos
Cálcio/metabolismo , Proteínas de Neoplasias/genética , Proteína ORAI1/genética , Neoplasias Ovarianas/genética , Fator de Crescimento Placentário/metabolismo , Molécula 1 de Interação Estromal/genética , Linhagem Celular Tumoral , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Neoplasias Ovarianas/metabolismo , Regulação para Cima
15.
Kidney Blood Press Res ; 43(6): 1742-1748, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30504710

RESUMO

Fibroblast growth factor 23 (FGF23) is released primarily from osteoblasts/osteocytes in bone. In cooperation with the transmembrane protein Klotho, FGF23 is a powerful inhibitor of 1α 25OH Vitamin D Hydroxylase (Cyp27b1) and thus of the formation of 1,25-dihydroxyvitamin D3 (1,25(OH)2D3). As 1,25(OH)2D3 up-regulates intestinal calcium and phosphate absorption, the downregulation of 1,25(OH)2D3 synthesis counteracts phosphate excess and tissue calcification. FGF23 also directly inhibits renal phosphate reabsorption. Other actions of FGF23 include triggering of cardiac hypertrophy. FGF23 formation and/or release are stimulated by 1,25(OH)2D3, phosphate excess, Ca2+, PTH, leptin, catecholamines, mineralocorticoids, volume depletion, lithium, high fat diet, iron deficiency, TNFα and TGFß2. The stimulating effect of 1,25(OH)2D3 on FGF23 expression is dependent on RAC1/PAK1 induced actin-polymerisation. Intracellular signaling involved in the stimulation of FGF23 release also includes increases in the cytosolic Ca2+ concentration ([Ca2+]i) following intracellular Ca2+ release and store-operated Ca2+ entry (SOCE). SOCE is accomplished by the Ca2+ release-activated calcium channel protein 1 (Orai1) and its stimulator stromal interaction molecule 1 (STIM1). Expression of Orai1, SOCE and FGF23-formation are up-regulated by the proinflammatory transcription factor NFκB. The present brief review describes the cellular mechanisms involved in FGF23 regulation and its sensitivity to both phosphate metabolism and inflammation. The case is made that up-regulation of FGF23 by inflammatory mediators and signaling may amplify inflammation by inhibiting formation of the anti-inflammatory 1,25(OH)2D3.


Assuntos
Fatores de Crescimento de Fibroblastos/metabolismo , Homeostase , Inflamação/etiologia , Fosfatos/metabolismo , Animais , Fator de Crescimento de Fibroblastos 23 , Humanos , Vitamina D/análogos & derivados , Vitamina D/antagonistas & inibidores
16.
Cell Physiol Biochem ; 51(1): 278-289, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30453283

RESUMO

BACKGROUND/AIMS: The neurodegenerative disease Chorea-Acanthocytosis (ChAc) is caused by loss-of-function-mutations of the chorein-encoding gene VPS13A. In ChAc neurons transcript levels and protein abundance of Ca2+ release activated channel moiety (CRAC) Orai1 as well as its regulator STIM1/2 are decreased, resulting in blunted store operated Ca2+-entry (SOCE) and enhanced suicidal cell death. SOCE is up-regulated and cell death decreased by lithium. The effects of lithium are paralleled by upregulation of serum & glucocorticoid inducible kinase SGK1 and abrogated by pharmacological SGK1 inhibition. In other cell types SGK1 has been shown to be partially effective by upregulation of NFκB, a transcription factor stimulating the expression of Orai1 and STIM. The present study explored whether pharmacological inhibition of NFκB interferes with Orai1/STIM1/2 expression and SOCE and their upregulation by lithium in ChAc neurons. METHODS: Cortical neurons were differentiated from induced pluripotent stem cells generated from fibroblasts of ChAc patients and healthy volunteers. Orai1 and STIM1 transcript levels and protein abundance were estimated from qRT-PCR and Western blotting, respectively, cytosolic Ca2+-activity ([Ca2+]i) from Fura-2-fluorescence, SOCE from increase of [Ca2+]i following Ca2+ re-addition after Ca2+-store depletion with sarco-endoplasmatic Ca2+-ATPase inhibitor thapsigargin (1µM), as well as CRAC current utilizing whole cell patch clamp recording. RESULTS: Orai1 and STIM1 transcript levels and protein abundance as well as SOCE and CRAC current were significantly enhanced by lithium treatment (2 mM, 24 hours). These effects were reversed by NFκB inhibitor wogonin (50 µM). CONCLUSION: The stimulation of expression and function of Orai1/STIM1/2 by lithium in ChAc neurons are disrupted by pharmacological NFκB inhibition.


Assuntos
Cálcio/metabolismo , Flavanonas/farmacologia , Expressão Gênica/efeitos dos fármacos , Lítio/farmacologia , Proteínas de Neoplasias/metabolismo , Proteína ORAI1/metabolismo , Molécula 1 de Interação Estromal/metabolismo , ATPases Transportadoras de Cálcio/antagonistas & inibidores , ATPases Transportadoras de Cálcio/metabolismo , Diferenciação Celular , Células Cultivadas , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Potenciais da Membrana/efeitos dos fármacos , NF-kappa B/antagonistas & inibidores , NF-kappa B/metabolismo , Proteínas de Neoplasias/genética , Doenças Neurodegenerativas/metabolismo , Doenças Neurodegenerativas/patologia , Neurônios/citologia , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Proteína ORAI1/genética , Técnicas de Patch-Clamp , Molécula 1 de Interação Estromal/genética , Tapsigargina/farmacologia
17.
Cell Calcium ; 74: 29-34, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-29807219

RESUMO

The pore forming Ca2+ release activated Ca2+ channel (CRAC) isoforms ORAI1-3 and their regulators STIM1,2 accomplish store operated Ca2+ entry (SOCE). Activation of SOCE may lead to cytosolic Ca2+ oscillations, which in turn support cell proliferation and cell survival. ORAI/STIM and thus SOCE are upregulated by the serum and glucocorticoid inducible kinase SGK1, a kinase under powerful genomic regulation and activated by phosphorylation via the phosphoinositol-3-phosphate pathway. SGK1 enhances ORAI1 abundance partially by phosphorylation of Nedd4-2, an ubiquitin ligase priming the channel protein for degradation. The SGK1-phosphorylated Nedd4-2 binds to the protein 14-3-3 and is thus unable to ubiquinate ORAI1. SGK1 further increases the ORAI1 and STIM1 protein abundance by activating nuclear factor kappa B (NF-κB), a transcription factor upregulating the expression of STIM1 and ORAI1. SGK1-sensitive upregulation of ORAI/STIM and thus SOCE is triggered by a wide variety of hormones and growth factors, as well as several cell stressors including ischemia, radiation, and cell shrinkage. SGK1 dependent upregulation of ORAI/STIM confers survival of tumor cells and thus impacts on growth and therapy resistance of cancer. On the other hand, SGK1-dependent upregulation of ORAI1 and STIM1 may support survival of neurons and impairment of SGK1-dependent ORAI/STIM activity may foster neurodegeneration. Clearly, further experimental effort is needed to define the mechanisms linking SGK1-dependent upregulation of ORAI1 and STIM1 to cell survival and to define the impact of SGK1-dependent upregulation of ORAI1 and STIM1 on malignancy and neurodegenerative disease.


Assuntos
Proteínas Imediatamente Precoces/metabolismo , Proteínas de Neoplasias/metabolismo , Proteína ORAI1/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Molécula 1 de Interação Estromal/metabolismo , Animais , Sinalização do Cálcio/fisiologia , Sobrevivência Celular/fisiologia , Humanos , Neoplasias/metabolismo , Neoplasias/patologia , Doenças Neurodegenerativas/metabolismo , Doenças Neurodegenerativas/patologia
18.
Cell Physiol Biochem ; 47(1): 368-377, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29772566

RESUMO

BACKGROUND/AIMS: The epigenetic factor KDM2B is a histone demethylase expressed in various tumors. Recently, we have shown that KDM2B regulates actin cytoskeleton organization, small Rho GTPases signaling, cell-cell adhesion and migration of prostate tumor cells. In the present study, we addressed its role in regulating EMT and small GTPases expression in colon tumor cells. METHODS: We used RT-PCR for the transcriptional analysis of various genes, Western blotting for the assessment of protein expression and immunofluorescence microscopy for visualization of fluorescently labeled proteins. RESULTS: We report here that KDM2B regulates EZH2 and BMI1 in HCT116 colon tumor cells. Knockdown of this epigenetic factor induced potent up-regulation of the protein levels of the epithelial markers E-cadherin and ZO-1, while the mesenchymal marker N-cadherin was downregulated. On the other hand, KDM2B overexpression downregulated the levels of both epithelial markers and upregulated the mesenchymal marker, suggesting control of EMT by KDM2B. In addition, RhoA, RhoB and RhoC protein levels diminished upon KDM2B-knockdown, while all three small GTPases became upregulated in KDM2B-overexpressing HCT116 cell clones. Interestingly, Rac1 GTPase level increased upon KDM2B-knockdown and diminished in KDM2B-overexpressing HCT116 colon tumor- and DU-145 prostate cancer cells. CONCLUSIONS: These results establish a clear functional role of the epigenetic factor KDM2B in the regulation of EMT and small-GTPases expression in colon tumor cells and further support the recently postulated oncogenic role of this histone demethylase in various tumors.


Assuntos
Neoplasias do Colo/genética , Transição Epitelial-Mesenquimal , Proteínas F-Box/genética , Regulação Neoplásica da Expressão Gênica , Histona Desmetilases com o Domínio Jumonji/genética , Proteínas Monoméricas de Ligação ao GTP/genética , Neoplasias do Colo/patologia , Proteína Potenciadora do Homólogo 2 de Zeste/genética , Epigênese Genética , Células HCT116 , Humanos
19.
Cell Signal ; 48: 54-63, 2018 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-29705334

RESUMO

We have shown previously that the small GTPases RhoA and RhoB play important roles in early TGFß-induced actin cytoskeleton reorganization and that RhoB is transcriptionally activated by TGFß and its signaling effectors, the Smad proteins. However, this long-term impact of RhoB gene upregulation by TGFß on cellular functions is not known. We now show that increased levels of RhoB, but not of RhoA, inhibit the TGFß/Smad-mediated transcriptional induction of the cell cycle inhibitor p21WAF1/Cip1 gene as well as of a generic Smad-responsive promoter suggesting that RhoB could be part of an auto-inhibitory loop in TGFß signaling by inhibiting the genomic responses to TGFß. We show that RhoB blocks the interaction of Smad3 with the type I TGFß receptor which prohibits its phosphorylation by this receptor and its translocation to the nucleus. Using in vivo GST pull-down and co-immunoprecipitation assays we show that Smad3 physically interacts with RhoB but not with RhoA. We show that RhoB, but not RhoA, potently regulates actin cytoskeleton reorganization by inducing stress fiber formation in a Smad-dependent manner. Finally we show that Smad3 downregulates the expression of the epithelial adherens junctions protein E-Cadherin and upregulates the fibronectin gene in Smad3-/- JEG3 cells only in the presence of RhoB suggesting that RhoB/Smad3 complexes in the cytoplasm may be involved in epithelial to mesenchymal transitions. In summary, our data propose a novel mechanism of TGFß/Smad signaling modulation by the small GTPase RhoB and show that this TGFß/RhoB signaling cross talk affects the nuclear and cytoplasmic responses to TGFß in opposite ways.


Assuntos
Proteína Smad3/genética , Proteína Smad3/metabolismo , Fator de Crescimento Transformador beta/genética , Proteína rhoA de Ligação ao GTP/genética , Proteína rhoB de Ligação ao GTP/fisiologia , Células 3T3 , Citoesqueleto de Actina/metabolismo , Animais , Antígenos CD/metabolismo , Caderinas/metabolismo , Linhagem Celular Tumoral , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Células HEK293 , Humanos , Camundongos , Fosforilação , Receptor do Fator de Crescimento Transformador beta Tipo I/metabolismo , Transdução de Sinais , Fator de Crescimento Transformador beta/metabolismo , Proteína rhoA de Ligação ao GTP/metabolismo
20.
Cell Mol Life Sci ; 75(12): 2111-2124, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29500478

RESUMO

Rho GTPases are highly conserved proteins that play critical roles in many cellular processes including actin dynamics, vesicular trafficking, gene transcription, cell-cycle progression, and cell adhesion. The main mode of regulation of Rho GTPases is through guanine nucleotide binding (cycling between an active GTP-bound form and an inactive GDP-bound form), but transcriptional, post-transcriptional, and post-translational modes of Rho regulation have also been described. In the present review, we summarize recent progress on the mechanisms that control the expression of the three members of the Rho-like subfamily (RhoA, RhoB, and RhoC) at the level of gene transcription as well as their post-transcriptional regulation by microRNAs. We also discuss the progress made in deciphering the mechanisms of cross-talk between Rho proteins and the transforming growth factor ß signaling pathway and their implications for the pathogenesis of human diseases such as cancer metastasis and fibrosis.


Assuntos
Regulação da Expressão Gênica , MicroRNAs/genética , Ativação Transcricional , Proteína rhoA de Ligação ao GTP/genética , Proteína rhoB de Ligação ao GTP/genética , Proteína de Ligação a GTP rhoC/genética , Animais , Humanos , MicroRNAs/metabolismo , Neoplasias/genética , Neoplasias/metabolismo , Transdução de Sinais , Fator de Crescimento Transformador beta/metabolismo , Proteína rhoA de Ligação ao GTP/metabolismo , Proteína rhoB de Ligação ao GTP/metabolismo , Proteína de Ligação a GTP rhoC/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...