Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 169
Filtrar
1.
Adv Biol Regul ; 91: 101014, 2024 01.
Artigo em Inglês | MEDLINE | ID: mdl-38242820

RESUMO

Myelodysplastic Syndromes, a heterogeneous group of hematological disorders, are characterized by abnormalities in phosphoinositide-dependent signaling, epigenetic regulators, apoptosis, and cytokine interactions within the bone marrow microenvironment, contributing to disease pathogenesis and neoplastic growth. Comprehensive knowledge of these pathways is crucial for the development of innovative therapies that aim to restore normal apoptosis and improve patient outcomes.


Assuntos
Células-Tronco Hematopoéticas , Síndromes Mielodisplásicas , Humanos , Células-Tronco Hematopoéticas/metabolismo , Síndromes Mielodisplásicas/genética , Síndromes Mielodisplásicas/metabolismo , Medula Óssea/patologia , Citocinas/metabolismo , Transdução de Sinais
2.
Biomolecules ; 13(5)2023 05 07.
Artigo em Inglês | MEDLINE | ID: mdl-37238668

RESUMO

Phospholipases are essential intermediaries that work as hydrolyzing enzymes of phospholipids (PLs), which represent the most abundant species contributing to the biological membranes of nervous cells of the healthy human brain. They generate different lipid mediators, such as diacylglycerol, phosphatidic acid, lysophosphatidic acid, and arachidonic acid, representing key elements of intra- and inter-cellular signaling and being involved in the regulation of several cellular mechanisms that can promote tumor progression and aggressiveness. In this review, it is summarized the current knowledge about the role of phospholipases in brain tumor progression, focusing on low- and high-grade gliomas, representing promising prognostic or therapeutic targets in cancer therapies due to their influential roles in cell proliferation, migration, growth, and survival. A deeper understanding of the phospholipases-related signaling pathways could be necessary to pave the way for new targeted therapeutic strategies.


Assuntos
Neoplasias Encefálicas , Glioma , Humanos , Fosfolipases/metabolismo , Neoplasias Encefálicas/terapia , Encéfalo/metabolismo , Glioma/terapia , Fosfolipídeos
3.
Dev Cell ; 58(4): 320-334.e8, 2023 02 27.
Artigo em Inglês | MEDLINE | ID: mdl-36800996

RESUMO

Exosomes transport a variety of macromolecules and modulate intercellular communication in physiology and disease. However, the regulation mechanisms that determine exosome contents during exosome biogenesis remain poorly understood. Here, we find that GPR143, an atypical GPCR, controls the endosomal sorting complex required for the transport (ESCRT)-dependent exosome biogenesis pathway. GPR143 interacts with HRS (an ESCRT-0 Subunit) and promotes its association to cargo proteins, such as EGFR, which subsequently enables selective protein sorting into intraluminal vesicles (ILVs) in multivesicular bodies (MVBs). GPR143 is elevated in multiple cancers, and quantitative proteomic and RNA profiling of exosomes in human cancer cell lines showed that the GPR143-ESCRT pathway promotes secretion of exosomes that carry unique cargo, including integrins signaling proteins. Through gain- and loss-of-function studies in mice, we show that GPR143 promotes metastasis by secreting exosomes and increasing cancer cell motility/invasion through the integrin/FAK/Src pathway. These findings provide a mechanism for regulating the exosomal proteome and demonstrate its ability to promote cancer cell motility.


Assuntos
Exossomos , Neoplasias , Humanos , Animais , Camundongos , Exossomos/metabolismo , Proteômica , Complexos Endossomais de Distribuição Requeridos para Transporte/genética , Complexos Endossomais de Distribuição Requeridos para Transporte/metabolismo , Transporte Proteico , Transporte Biológico , Corpos Multivesiculares/metabolismo , Neoplasias/metabolismo , Proteínas do Olho/metabolismo , Glicoproteínas de Membrana/metabolismo
4.
Clin Epigenetics ; 15(1): 27, 2023 02 20.
Artigo em Inglês | MEDLINE | ID: mdl-36803590

RESUMO

BACKGROUND: miRNAs are small non-coding RNAs that regulate gene expression and are linked to cancer development and progression. miRNA profiles are currently studied as new prognostic factors or therapeutic perspectives. Among hematological cancers, myelodysplastic syndromes at higher risk of evolution into acute myeloid leukemia are treated with hypomethylating agents, like azacitidine, alone or in combination with other drugs, such as lenalidomide. Recent data showed that, during azacitidine and lenalidomide therapy, the concurrent acquisition of specific point mutations affecting inositide signalling pathways is associated with lack or loss of response to therapy. As these molecules are implicated in epigenetic processes, possibly involving miRNA regulation, and in leukemic progression, through the regulation of proliferation, differentiation and apoptosis, here we performed a new miRNA expression analysis of 26 high-risk patients with myelodysplastic syndromes treated with azacitidine and lenalidomide at baseline and during therapy. miRNA array data were processed, and bioinformatic results were correlated with clinical outcome to investigate the translational relevance of selected miRNAs, while the relationship between selected miRNAs and specific molecules was experimentally tested and proven. RESULTS: Patients' overall response rate was 76.9% (20/26 cases): complete remission (5/26, 19.2%), partial remission (1/26, 3.8%), marrow complete remission (2/26, 7.7%), hematologic improvement (6/26, 23.1%), hematologic improvement with marrow complete remission (6/26, 23.1%), whereas 6/26 patients (23.1%) had a stable disease. miRNA paired analysis showed a statistically significant up-regulation of miR-192-5p after 4 cycles of therapy (vs baseline), that was confirmed by real-time PCR analyses, along with an involvement of BCL2, that was proven to be a miR-192-5p target in hematopoietic cells by luciferase assays. Furthermore, Kaplan-Meier analyses showed a significant correlation between high levels of miR-192-5p after 4 cycles of therapy and overall survival or leukemia-free survival, that was stronger in responders, as compared with patients early losing response and non-responders. CONCLUSIONS: This study shows that high levels of miR-192-5p are associated with higher overall survival and leukemia-free survival in myelodysplastic syndromes responding to azacitidine and lenalidomide. Moreover, miR-192-5p specifically targets and inhibits BCL2, possibly regulating proliferation and apoptosis and leading to the identification of new therapeutic targets.


Assuntos
Leucemia Mieloide Aguda , MicroRNAs , Síndromes Mielodisplásicas , Humanos , Azacitidina/farmacologia , Azacitidina/uso terapêutico , Lenalidomida/farmacologia , Lenalidomida/uso terapêutico , MicroRNAs/genética , Metilação de DNA , Síndromes Mielodisplásicas/tratamento farmacológico , Síndromes Mielodisplásicas/genética , Leucemia Mieloide Aguda/genética , Proteínas Proto-Oncogênicas c-bcl-2
5.
Adv Biol Regul ; 87: 100955, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36706610

RESUMO

Aberrant signaling pathways regulating proliferation and differentiation of hematopoietic stem cells (HSCs) can contribute to disease pathogenesis and neoplastic growth. Phosphoinositides (PIs) are inositol phospholipids that are implicated in the regulation of critical signaling pathways: aberrant regulation of Phospholipase C (PLC) beta1, PLCgamma1 and the PI3K/Akt/mTOR pathway play essential roles in the pathogenesis of Myelodysplastic Syndromes (MDS) and Acute Myeloid Leukemia (AML).


Assuntos
Leucemia Mieloide Aguda , Síndromes Mielodisplásicas , Humanos , Fosfatidilinositol 3-Quinases/metabolismo , Transdução de Sinais/fisiologia , Leucemia Mieloide Aguda/metabolismo , Fosfatidilinositóis/metabolismo , Síndromes Mielodisplásicas/metabolismo
6.
Hepatol Commun ; 6(11): 3234-3246, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-36153805

RESUMO

Phospholipase C gamma 1 (PLCγ1) plays an oncogenic role in several cancers, alongside its usual physiological roles. Despite studies aimed at identifying the effect of PLCγ1 on tumors, the pathogenic role of PLCγ1 in the tumorigenesis and development of hepatocellular carcinoma (HCC) remains unknown. To investigate the function of PLCγ1 in HCC, we generated hepatocyte-specific PLCγ1 conditional knockout (PLCγ1f/f ; Alb-Cre) mice and induced HCC with diethylnitrosamine (DEN). Here, we identified that hepatocyte-specific PLCγ1 deletion effectively prevented DEN-induced HCC in mice. PLCγ1f/f ; Alb-Cre mice showed reduced tumor burden and tumor progression, as well as a decreased incidence of HCC and less marked proliferative and inflammatory responses. We also showed that oncogenic phenotypes such as repressed apoptosis, and promoted proliferation, cell cycle progression and migration, were induced by PLCγ1. In terms of molecular mechanism, PLCγ1 regulated the activation of signal transducer and activator of transcription 3 (STAT3) signaling. Moreover, PLCγ1 expression is elevated in human HCC and correlates with a poor prognosis in patients with HCC. Our results suggest that PLCγ1 promotes the pathogenic progression of HCC, and PLCγ1/STAT3 axis was identified as a potential therapeutic target pathway for HCC.


Assuntos
Carcinoma Hepatocelular , Neoplasias Hepáticas , Humanos , Camundongos , Animais , Fator de Transcrição STAT3/genética , Carcinoma Hepatocelular/induzido quimicamente , Dietilnitrosamina/toxicidade , Neoplasias Hepáticas/induzido quimicamente , Fosfolipase C gama/genética , Proliferação de Células , Carcinogênese/genética
7.
FEBS Open Bio ; 12(6): 1220-1229, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35347892

RESUMO

O-GlcNAcylation of intracellular proteins (O-GlcNAc) is a post-translational modification that often competes with phosphorylation in diverse cellular signaling pathways. Recent studies on human malignant tumors have demonstrated that O-GlcNAc is implicated in cellular features relevant to metastasis. Here, we report that lysophosphatidic acid (LPA)-induced ovarian cancer cell (OVCAR-3) migration is regulated by O-GlcNAc. We found that O-GlcNAc modification of ERM family proteins, a membrane-cytoskeletal crosslinker, was inversely correlated with its phosphorylation status. Moreover, the LPA-induced formation of membrane protrusion structures, as well as the migration of OVCAR-3 cells, was reduced by the accumulation of O-GlcNAc. Collectively, these findings suggest that O-GlcNAc is an essential signaling element controlling ERM family proteins involved in OVCAR-3 cell migration.


Assuntos
Acetilglucosamina , Neoplasias Ovarianas , Acetilglucosamina/química , Acetilglucosamina/metabolismo , Apoptose , Linhagem Celular Tumoral , Movimento Celular , Feminino , Humanos , Lisofosfolipídeos
8.
Cell Mol Life Sci ; 79(4): 195, 2022 Mar 18.
Artigo em Inglês | MEDLINE | ID: mdl-35303162

RESUMO

Glioblastoma represents the most lethal brain tumor in adults. Several studies have shown the key role of phospholipase C ß1 (PLCß1) in the regulation of many mechanisms within the central nervous system suggesting PLCß1 as a novel signature gene in the molecular classification of high-grade gliomas. This study aims to determine the pathological impact of PLCß1 in glioblastoma, confirming that PLCß1 gene expression correlates with glioma's grade, and it is lower in 50 glioblastoma samples compared to 20 healthy individuals. PLCß1 silencing in cell lines and primary astrocytes, leads to increased cell migration and invasion, with the increment of mesenchymal transcription factors and markers, as Slug and N-Cadherin and metalloproteinases. Cell proliferation, through increased Ki-67 expression, and the main survival pathways, as ß-catenin, ERK1/2 and Stat3 pathways, are also affected by PLCß1 silencing. These data suggest a potential role of PLCß1 in maintaining a normal or less aggressive glioma phenotype.


Assuntos
Neoplasias Encefálicas , Glioblastoma , Glioma , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patologia , Proliferação de Células/genética , Glioblastoma/patologia , Glioma/patologia , Humanos , Fosfolipase C beta/genética , Fosfolipase C beta/metabolismo
9.
Adv Biol Regul ; 83: 100838, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34819252

RESUMO

Phosphoinositide-specific phospholipases C (PLCs) are a class of enzymes involved in several cell activities, such as cell cycle regulation, proliferation, differentiation and cytoskeletal dynamics. Among these enzymes, PLCγ1 is one of the most expressed PLCs in the brain, contributing to a complex network in the developing nervous system. Several studies have shown that PLCγ1 signaling imbalance is linked to several brain disorders, including glioblastoma, the most aggressive brain tumor in adults. Indeed, it has been demonstrated a link between PLCγ1 inhibition and the arrest of glioma cell motility of fetal rat brain aggregates and the impairment of cell invasion abilities following its down-regulation. This study aims to determine the pathological influence of PLCγ1 in glioblastoma, through a translational study which combines in silico data, data from glioblastoma patients' samples and data on engineered cell lines. We found out that PLCγ1 gene expression correlates with the pathological grade of gliomas, and it is higher in fifty patients' glioblastoma tissue samples compared to twenty healthy controls. Moreover, it was demonstrated that PLCγ1 silencing in U87-MG leads to a reduction in cell migration and invasion abilities. The opposite trend was observed following PLCγ1 overexpression, suggesting an interesting possible involvement of PLCγ1 in gliomas' aggressiveness.


Assuntos
Neoplasias Encefálicas , Glioblastoma , Animais , Neoplasias Encefálicas/metabolismo , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Regulação Neoplásica da Expressão Gênica , Glioblastoma/patologia , Humanos , Invasividade Neoplásica/genética , Ratos , Transdução de Sinais
10.
Adv Biol Regul ; 82: 100833, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34773889

RESUMO

Genetic mutations leading to the development of various diseases, such as cancer, diabetes, and neurodegenerative disorders, can be attributed to multiple mechanisms and exposure to diverse environments. These disorders further increase gene mutation rates and affect the activity of translated proteins, both phenomena associated with cellular responses. Therefore, maintaining the integrity of genetic and epigenetic information is critical for disease suppression and prevention. With the advent of genome sequencing technologies, large-scale genomic data-based machine learning tools, including deep learning, have been used to predict and identify somatic inactivation or negative dominant expression of target genes in various diseases. Although deep learning studies have recently been highlighted for their ability to distinguish between the genetic information of diseases, conventional wisdom is also necessary to explain the correlation between genotype and phenotype. Herein, we summarize the current understanding of phosphoinositide-specific phospholipase C isozymes (PLCs) and an overview of their associations with genetic variation, as well as their emerging roles in several diseases. We also predicted and discussed new findings of cryptic PLC splice variants by deep learning and the clinical implications of the PLC genetic variations predicted using these tools.


Assuntos
Encefalopatias , Aprendizado Profundo , Humanos , Isoenzimas/genética , Mutação , Fosfoinositídeo Fosfolipase C/genética
11.
Biochem Biophys Res Commun ; 577: 103-109, 2021 11 05.
Artigo em Inglês | MEDLINE | ID: mdl-34509721

RESUMO

As essential phospholipid signaling regulators, phospholipase C (PLC)s are activated by various extracellular ligands and mediate intracellular signal transduction. PLCγ1 is involved in regulating various cancer cell functions. However, the precise in vivo link between PLCγ1 and cancer behavior remains undefined. To investigate the role of PLCγ1 in colorectal carcinogenesis, we generated an intestinal tissue-specific Plcg1 knock out (KO) in adenomatous polyposis coli (Apc) Min/+ mice. Plcg1 deficiency in ApcMin/+ mice showed earlier death, with a higher colorectal tumor incidence in both number and size than in wild-type mice. Mechanistically, inhibition of PLCγ1 increased the levels of its substrate phosphoinositol 4,5-bisphosphate (PIP2) at the plasma membrane and promoted the activation of Wnt receptor low-density lipoprotein receptor-related protein 6 (LRP6) by glycogen synthase kinase 3ß (GSK3ß) to enhance ß-catenin signaling. Enhanced cell proliferation and Wnt/ß-catenin signaling were observed in colon tumors from Plcg1 KO mice. Furthermore, low PLCγ1 expression was associated with a poor prognosis of colon cancer patients. Collectively, we demonstrated the role of PLCγ1 in vivo as a tumor suppressor relationship between the regulation of the PIP2 level and Wnt/ß-catenin-dependent intestinal tumor formation.


Assuntos
Proliferação de Células/genética , Neoplasias Colorretais/genética , Regulação Neoplásica da Expressão Gênica , Fosfolipase C gama/genética , Via de Sinalização Wnt/genética , beta Catenina/genética , Animais , Linhagem Celular Tumoral , Neoplasias Colorretais/enzimologia , Neoplasias Colorretais/patologia , Progressão da Doença , Glicogênio Sintase Quinase 3 beta/genética , Glicogênio Sintase Quinase 3 beta/metabolismo , Humanos , Intestinos/enzimologia , Intestinos/patologia , Estimativa de Kaplan-Meier , Proteína-6 Relacionada a Receptor de Lipoproteína de Baixa Densidade/genética , Proteína-6 Relacionada a Receptor de Lipoproteína de Baixa Densidade/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fosfatidilinositol 4,5-Difosfato/metabolismo , Fosfolipase C gama/deficiência , beta Catenina/metabolismo
12.
Adv Exp Med Biol ; 1187: 23-52, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33983572

RESUMO

Breast cancer progression results from subversion of multiple intra- or intercellular signaling pathways in normal mammary tissues and their microenvironment, which have an impact on cell differentiation, proliferation, migration, and angiogenesis. Phospholipases (PLC, PLD and PLA) are essential mediators of intra- and intercellular signaling. They hydrolyze phospholipids, which are major components of cell membrane that can generate many bioactive lipid mediators, such as diacylglycerol, phosphatidic acid, lysophosphatidic acid, and arachidonic acid. Enzymatic processing of phospholipids by phospholipases converts these molecules into lipid mediators that regulate multiple cellular processes, which in turn can promote breast cancer progression. Thus, dysregulation of phospholipases contributes to a number of human diseases, including cancer. This review describes how phospholipases regulate multiple cancer-associated cellular processes, and the interplay among different phospholipases in breast cancer. A thorough understanding of the breast cancer-associated signaling networks of phospholipases is necessary to determine whether these enzymes are potential targets for innovative therapeutic strategies.


Assuntos
Neoplasias da Mama , Fosfolipase D , Humanos , Fosfolipase D/metabolismo , Fosfolipases , Fosfolipídeos , Transdução de Sinais , Microambiente Tumoral , Fosfolipases Tipo C/metabolismo
13.
J Exp Med ; 218(7)2021 07 05.
Artigo em Inglês | MEDLINE | ID: mdl-33970189

RESUMO

Because of their common signaling molecules, the main T cell receptor (TCR) signaling cascades in CD4+ and CD8+ T cells are considered qualitatively identical. Herein, we show that TCR signaling in CD8+ T cells is qualitatively different from that in CD4+ T cells, since CD8α ignites another cardinal signaling cascade involving phospholipase C ß4 (PLCß4). TCR-mediated responses were severely impaired in PLCß4-deficient CD8+ T cells, whereas those in CD4+ T cells were intact. PLCß4-deficient CD8+ T cells showed perturbed activation of peripheral TCR signaling pathways downstream of IP3 generation. Binding of PLCß4 to the cytoplasmic tail of CD8α was important for CD8+ T cell activation. Furthermore, GNAQ interacted with PLCß4, mediated double phosphorylation on threonine 886 and serine 890 positions of PLCß4, and activated CD8+ T cells in a PLCß4-dependent fashion. PLCß4-deficient mice exhibited defective antiparasitic host defense and antitumor immune responses. Altogether, PLCß4 differentiates TCR signaling in CD4+ and CD8+ T cells and selectively promotes CD8+ T cell-dependent adaptive immunity.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Fosfolipase C beta/imunologia , Transdução de Sinais/imunologia , Animais , Linhagem Celular , Citoplasma/imunologia , Células HEK293 , Humanos , Ativação Linfocitária/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Fosforilação/imunologia , Receptores de Antígenos de Linfócitos T/imunologia
14.
Adv Biol Regul ; 79: 100771, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33303387

RESUMO

Phosphoinositide-specific phospholipases C (PI-PLCs) are a class of enzymes involved in the phosphatidylinositol metabolism, which is implicated in the activation of several signaling pathways and which controls several cellular processes. The scientific community has long accepted the existence of a nuclear phosphoinositide (PI) metabolism, independent from the cytoplasmic one, critical in nuclear function control. Indeed, nuclear PIs are involved in many activities, such as cell cycle regulation, cell proliferation, cell differentiation, membrane transport, gene expression and cytoskeletal dynamics. There are several types of PIs and enzymes implicated in brain activities and among these enzymes, PI-PLCs contribute to a specific and complex network in the developing nervous system. Moreover, considering the abundant presence of PI-PLCß1, PI-PLCγ1 and PI-PLCß4 in the brain, a specific role for each PLC subtype has been suggested in the control of neuronal activity, which is important for synapse function, development and other mechanisms. The focus of this review is to describe the latest research about the involvement of PI-PLC signaling in the nervous system, both physiologically and in pathological conditions. Indeed, PI-PLC signaling imbalance seems to be also linked to several brain disorders including epilepsy, movement and behavior disorders, neurodegenerative diseases and, in addition, some PI-PLC subtypes could become potential novel signature genes for high-grade gliomas.


Assuntos
Encefalopatias/enzimologia , Encéfalo/enzimologia , Fosfoinositídeo Fosfolipase C/metabolismo , Animais , Encéfalo/metabolismo , Encefalopatias/genética , Encefalopatias/metabolismo , Encefalopatias/patologia , Humanos , Fosfatidilinositóis/metabolismo , Fosfoinositídeo Fosfolipase C/genética , Transdução de Sinais
15.
Cell Mol Life Sci ; 78(6): 2781-2795, 2021 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-33034697

RESUMO

Autosomal-dominant leukodystrophy (ADLD) is a rare fatal neurodegenerative disorder with overexpression of the nuclear lamina component, Lamin B1 due to LMNB1 gene duplication or deletions upstream of the gene. The molecular mechanisms responsible for driving the onset and development of this pathology are not clear yet. Vacuolar demyelination seems to be one of the most significant histopathological observations of ADLD. Considering the role of oligodendrocytes, astrocytes, and leukemia inhibitory factor (LIF)-activated signaling pathways in the myelination processes, this work aims to analyze the specific alterations in different cell populations from patients with LMNB1 duplications and engineered cellular models overexpressing Lamin B1 protein. Our results point out, for the first time, that astrocytes may be pivotal in the evolution of the disease. Indeed, cells from ADLD patients and astrocytes overexpressing LMNB1 show severe ultrastructural nuclear alterations, not present in oligodendrocytes overexpressing LMNB1. Moreover, the accumulation of Lamin B1 in astrocytes induces a reduction in LIF and in LIF-Receptor (LIF-R) levels with a consequential decrease in LIF secretion. Therefore, in both our cellular models, Jak/Stat3 and PI3K/Akt axes, downstream of LIF/LIF-R, are downregulated. Significantly, the administration of exogenous LIF can partially reverse the toxic effects induced by Lamin B1 accumulation with differences between astrocytes and oligodendrocytes, highlighting that LMNB1 overexpression drastically affects astrocytic function reducing their fundamental support to oligodendrocytes in the myelination process. In addition, inflammation has also been investigated, showing an increased activation in ADLD patients' cells.


Assuntos
Astrócitos/metabolismo , Doenças Desmielinizantes/patologia , Lamina Tipo B/metabolismo , Transdução de Sinais , Astrócitos/citologia , Núcleo Celular/metabolismo , Núcleo Celular/ultraestrutura , Células Cultivadas , Doenças Desmielinizantes/metabolismo , Regulação para Baixo/efeitos dos fármacos , Fibroblastos/citologia , Fibroblastos/metabolismo , Humanos , Peróxido de Hidrogênio/farmacologia , Mediadores da Inflamação/metabolismo , Lamina Tipo B/genética , Fator Inibidor de Leucemia/metabolismo , Fator Inibidor de Leucemia/farmacologia , Oligodendroglia/citologia , Oligodendroglia/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Fosforilação , Espécies Reativas de Oxigênio/metabolismo , Receptores de OSM-LIF/metabolismo , Regulação para Cima/efeitos dos fármacos
16.
FASEB J ; 34(11): 15400-15416, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32959428

RESUMO

MDS are characterized by anemia and transfusion requirements. Transfused patients frequently show iron overload that negatively affects hematopoiesis. Iron chelation therapy can be effective in these MDS cases, but the molecular consequences of this treatment need to be further investigated. That is why we studied the molecular features of iron effect and Deferasirox therapy on PI-PLCbeta1 inositide signaling, using hematopoietic cells and MDS samples. At baseline, MDS patients showing a positive response after iron chelation therapy displayed higher levels of PI-PLCbeta1/Cyclin D3/PKCalpha expression. During treatment, these responder patients, as well as hematopoietic cells treated with FeCl3 and Deferasirox, showed a specific reduction of PI-PLCbeta1/Cyclin D3/PKCalpha expression, indicating that this signaling pathway is targeted by Deferasirox. The treatment was also able to specifically decrease the production of ROS. This effect correlated with a reduction of IL-1A and IL-2, as well as Akt/mTOR phosphorylation. In contrast, cells exposed only to FeCl3 and cells from MDS patients refractory to Deferasirox showed a specific increase of ROS and PI-PLCbeta1/Cyclin D3/PKCalpha expression. All in all, our data show that PI-PLCbeta1 signaling is a target for iron-induced oxidative stress and suggest that baseline PI-PLCbeta1 quantification could predict iron chelation therapy response in MDS.


Assuntos
Ciclina D3/metabolismo , Sobrecarga de Ferro/complicações , Ferro/efeitos adversos , Síndromes Mielodisplásicas/terapia , Estresse Oxidativo/efeitos dos fármacos , Fosfolipase C beta/metabolismo , Proteína Quinase C-alfa/metabolismo , Idoso , Transfusão de Sangue/estatística & dados numéricos , Ciclina D3/genética , Deferasirox/farmacologia , Feminino , Regulação da Expressão Gênica , Humanos , Quelantes de Ferro/farmacologia , Masculino , Pessoa de Meia-Idade , Síndromes Mielodisplásicas/patologia , Fosfolipase C beta/genética , Fosforilação , Proteína Quinase C-alfa/genética , Transdução de Sinais
17.
Handb Exp Pharmacol ; 259: 291-308, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-31889219

RESUMO

Nuclear inositides have a specific subcellular distribution that is linked to specific functions; thus their regulation is fundamental both in health and disease. Emerging evidence shows that alterations in multiple inositide signalling pathways are involved in pathophysiology, not only in cancer but also in other diseases. Here, we give an overview of the main features of inositides in the cell, and we discuss their potential as new molecular therapeutic targets.


Assuntos
Núcleo Celular , Fosfatidilinositóis/fisiologia , Transdução de Sinais , Humanos
18.
FASEB J ; 34(1): 1270-1287, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31914593

RESUMO

Dysregulation of the adipo-osteogenic differentiation balance of mesenchymal stem cells (MSCs), which are common progenitor cells of adipocytes and osteoblasts, has been associated with many pathophysiologic diseases, such as obesity, osteopenia, and osteoporosis. Growing evidence suggests that lipid metabolism is crucial for maintaining stem cell homeostasis and cell differentiation; however, the detailed underlying mechanisms are largely unknown. Here, we demonstrate that glucosylceramide (GlcCer) and its synthase, glucosylceramide synthase (GCS), are key determinants of MSC differentiation into adipocytes or osteoblasts. GCS expression was increased during adipogenesis and decreased during osteogenesis. Targeting GCS using RNA interference or a chemical inhibitor enhanced osteogenesis and inhibited adipogenesis by controlling the transcriptional activity of peroxisome proliferator-activated receptor γ (PPARγ). Treatment with GlcCer sufficiently rescued adipogenesis and inhibited osteogenesis in GCS knockdown MSCs. Mechanistically, GlcCer interacted directly with PPARγ through A/B domain and synergistically enhanced rosiglitazone-induced PPARγ activation without changing PPARγ expression, thereby treatment with exogenous GlcCer increased adipogenesis and inhibited osteogenesis. Animal studies demonstrated that inhibiting GCS reduced adipocyte formation in white adipose tissues under normal chow diet and high-fat diet feeding and accelerated bone repair in a calvarial defect model. Taken together, our findings identify a novel lipid metabolic regulator for the control of MSC differentiation and may have important therapeutic implications.


Assuntos
Adipócitos/metabolismo , Diferenciação Celular , Glucosilceramidas/metabolismo , Glucosiltransferases/metabolismo , Células-Tronco Mesenquimais/metabolismo , Osteogênese , PPAR gama/metabolismo , Animais , Glucosilceramidas/genética , Glucosiltransferases/genética , Humanos , Camundongos , PPAR gama/genética
19.
Cancer Lett ; 471: 72-87, 2020 02 28.
Artigo em Inglês | MEDLINE | ID: mdl-31838085

RESUMO

Androgen receptor (AR) signaling plays a central role in metabolic reprogramming for prostate cancer (PCa) growth and progression. Mitochondria are metabolic powerhouses of the cell and support several hallmarks of cancer. However, the molecular links between AR signaling and the mitochondria that support the metabolic demands of PCa cells are poorly understood. Here, we demonstrate increased levels of dynamin-related protein 1 (DRP1), a mitochondrial fission mediator, in androgen-sensitive and castration-resistant AR-driven PCa. AR signaling upregulates DRP1 to form the VDAC-MPC2 complex, increases pyruvate transport into mitochondria, and supports mitochondrial metabolism, including oxidative phosphorylation and lipogenesis. DRP1 inhibition activates the cellular metabolic stress response, which involves AMPK phosphorylation, induction of autophagy, and the ER unfolded protein response, and attenuates androgen-induced proliferation. Additionally, DRP1 expression facilitates PCa cell survival under diverse metabolic stress conditions, including hypoxia and oxidative stress. Moreover, we found that increased DRP1 expression was indicative of poor prognosis in patients with castration-resistant PCa. Collectively, our findings link androgen signaling-mediated mitochondrial dynamics to metabolic reprogramming; moreover, they have important implications for understanding PCa progression.


Assuntos
Androgênios/metabolismo , Dinaminas/biossíntese , Mitocôndrias/metabolismo , Neoplasias de Próstata Resistentes à Castração/metabolismo , Linhagem Celular Tumoral , Proliferação de Células/fisiologia , Ciclo do Ácido Cítrico , Di-Hidrotestosterona/farmacologia , Dinaminas/antagonistas & inibidores , Dinaminas/genética , Dinaminas/metabolismo , Técnicas de Silenciamento de Genes , Humanos , Masculino , Dinâmica Mitocondrial , Proteínas de Transporte da Membrana Mitocondrial/metabolismo , Fosforilação Oxidativa , Células PC-3 , Neoplasias de Próstata Resistentes à Castração/patologia , Piruvatos/metabolismo , Receptores Androgênicos/metabolismo , Transdução de Sinais , Regulação para Cima , Canais de Ânion Dependentes de Voltagem/metabolismo
20.
Adv Biol Regul ; 75: 100673, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31711974

RESUMO

Recurrent cytogenetic aberrations, genetic mutations and variable gene expression have been consistently recognized in solid cancers and in leukaemia, including in Myelodysplastic Syndromes (MDS). Besides conventional cytogenetics, the growing accessibility of new techniques has led to a deeper analysis of the molecular significance of genetic variations. Indeed, gene mutations affecting splicing genes, as well as genes implicated in essential signalling pathways, play a pivotal role in MDS physiology and pathophysiology, representing potential new molecular targets for innovative therapeutic strategies.


Assuntos
Sistemas de Liberação de Medicamentos , Epigênese Genética , Mutação , Síndromes Mielodisplásicas , Splicing de RNA , Transdução de Sinais , Humanos , Síndromes Mielodisplásicas/tratamento farmacológico , Síndromes Mielodisplásicas/genética , Síndromes Mielodisplásicas/metabolismo , Síndromes Mielodisplásicas/fisiopatologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA