Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Nat Commun ; 13(1): 4188, 2022 07 20.
Artigo em Inglês | MEDLINE | ID: mdl-35858913

RESUMO

The formation of a functional blood vessel network relies on the ability of endothelial cells (ECs) to dynamically rearrange their adhesive contacts in response to blood flow and guidance cues, such as vascular endothelial growth factor-A (VEGF-A) and class 3 semaphorins (SEMA3s). Neuropilin 1 (NRP1) is essential for blood vessel development, independently of its ligands VEGF-A and SEMA3, through poorly understood mechanisms. Grounding on unbiased proteomic analysis, we report here that NRP1 acts as an endocytic chaperone primarily for adhesion receptors on the surface of unstimulated ECs. NRP1 localizes at adherens junctions (AJs) where, interacting with VE-cadherin, promotes its basal internalization-dependent turnover and favors vascular permeability initiated by histamine in both cultured ECs and mice. We identify a splice variant of tryptophanyl-tRNA synthetase (mini-WARS) as an unconventionally secreted extracellular inhibitory ligand of NRP1 that, by stabilizing it at the AJs, slows down both VE-cadherin turnover and histamine-elicited endothelial leakage. Thus, our work shows a role for NRP1 as a major regulator of AJs plasticity and reveals how mini-WARS acts as a physiological NRP1 inhibitory ligand in the control of VE-cadherin endocytic turnover and vascular permeability.


Assuntos
Neuropilina-1 , Triptofano-tRNA Ligase , Junções Aderentes/metabolismo , Animais , Antígenos CD , Caderinas/genética , Permeabilidade Capilar , Células Endoteliais/metabolismo , Histamina , Ligantes , Camundongos , Neuropilina-1/genética , Neuropilina-1/metabolismo , Proteômica , Triptofano-tRNA Ligase/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo
2.
Front Oncol ; 9: 848, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31544066

RESUMO

Tumors driven by mutant KRAS are among the most aggressive and refractory to treatment. Unfortunately, despite the efforts, targeting alterations of this GTPase, either directly or by acting on the downstream signaling cascades, has been, so far, largely unsuccessful. However, recently, novel therapeutic opportunities are emerging based on the effect that this oncogenic lesion exerts in rewiring the cancer cell metabolism. Cancer cells that become dependent on KRAS-driven metabolic adaptations are sensitive to the inhibition of these metabolic routes, revealing novel therapeutic windows of intervention. In general, mutant KRAS fosters tumor growth by shifting cancer cell metabolism toward anabolic pathways. Depending on the tumor, KRAS-driven metabolic rewiring occurs by up-regulating rate-limiting enzymes involved in amino acid, fatty acid, or nucleotide biosynthesis, and by stimulating scavenging pathways such as macropinocytosis and autophagy, which, in turn, provide building blocks to the anabolic routes, also maintaining the energy levels and the cell redox potential (1). This review will discuss the most recent findings on mutant KRAS metabolic reliance in tumor models of pancreatic and non-small-cell lung cancer, also highlighting the role that these metabolic adaptations play in resistance to target therapy. The effects of constitutive KRAS activation in glycolysis elevation, amino acids metabolism reprogramming, fatty acid turnover, and nucleotide biosynthesis will be discussed also in the context of different genetic landscapes.

3.
Cancers (Basel) ; 11(6)2019 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-31159426

RESUMO

Tumor microenvironment is particularly enriched with extracellular ATP (eATP), but conflicting evidence has been provided on its functional effects on tumor growth and vascular remodeling. We have previously shown that high eATP concentrations exert a strong anti-migratory, antiangiogenic and normalizing activity on human tumor-derived endothelial cells (TECs). Since both metabotropic and ionotropic purinergic receptors trigger cytosolic calcium increase ([Ca2+]c), the present work investigated the properties of [Ca2+]c events elicited by high eATP in TECs and their role in anti-migratory activity. In particular, the quantitative and kinetic properties of purinergic-induced Ca2+ release from intracellular stores and Ca2+ entry from extracellular medium were investigated. The main conclusions are: (1) stimulation of TECs with high eATP triggers [Ca2+]c signals which include Ca2+ mobilization from intracellular stores (mainly ER) and Ca2+ entry through the plasma membrane; (2) the long-lasting Ca2+ influx phase requires both store-operated Ca2+ entry (SOCE) and non-SOCE components; (3) SOCE is not significantly involved in the antimigratory effect of high ATP stimulation; (4) ER is the main source for intracellular Ca2+ release by eATP: it is required for the constitutive migratory potential of TECs but is not the only determinant for the inhibitory effect of high eATP; (5) a complex interplay occurs among ER, mitochondria and lysosomes upon purinergic stimulation; (6) high eUTP is unable to inhibit TEC migration and evokes [Ca2+]c signals very similar to those described for eATP. The potential role played by store-independent Ca2+ entry and Ca2+-independent events in the regulation of TEC migration by high purinergic stimula deserves future investigation.

4.
Int J Mol Sci ; 19(9)2018 Aug 30.
Artigo em Inglês | MEDLINE | ID: mdl-30200238

RESUMO

RAB5 is a small GTPase that belongs to the wide family of Rab proteins and localizes on early endosomes. In its active GTP-bound form, RAB5 recruits downstream effectors that, in turn, are responsible for distinct aspects of early endosome function, including their movement along microtubules. We previously reported that, at the onset of mitosis, RAB5positive vesicles cluster around the spindle poles and, during metaphase, move along spindle microtubules. RNAi-mediated depletion of the three RAB5 isoforms delays nuclear envelope breakdown at prophase and severely affects chromosome alignment and segregation. Here we show that depletion of the Kinesin-2 motor complex impairs long-range movement of RAB5 endosomes in interphase cells and prevents localization of these vesicles at the spindle during metaphase. Similarly to the effect caused by RAB5 depletion, functional ablation of Kinesin-2 delays nuclear envelope breakdown resulting in prolonged prophase. Altogether these findings suggest that endosomal transport at the onset of mitosis is required to control timing of nuclear envelope breakdown.


Assuntos
Endossomos/fisiologia , Cinesinas/genética , Cinesinas/metabolismo , Fuso Acromático/metabolismo , Proteínas rab5 de Ligação ao GTP/metabolismo , Linhagem Celular , Humanos , Interfase , Metáfase , Microtúbulos/metabolismo , Interferência de RNA
5.
Cancer Res ; 78(13): 3432-3444, 2018 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-29691252

RESUMO

USP6NL, also named RN-tre, is a GTPase-activating protein involved in control of endocytosis and signal transduction. Here we report that USP6NL is overexpressed in breast cancer, mainly of the basal-like/integrative cluster 10 subtype. Increased USP6NL levels were accompanied by gene amplification and were associated with worse prognosis in the METABRIC dataset, retaining prognostic value in multivariable analysis. High levels of USP6NL in breast cancer cells delayed endocytosis and degradation of the EGFR, causing chronic AKT (protein kinase B) activation. In turn, AKT stabilized the glucose transporter GLUT1 at the plasma membrane, increasing aerobic glycolysis. In agreement, elevated USP6NL sensitized breast cancer cells to glucose deprivation, indicating that their glycolytic capacity relies on this protein. Depletion of USP6NL accelerated EGFR/AKT downregulation and GLUT1 degradation, impairing cell proliferation exclusively in breast cancer cells that harbored increased levels of USP6NL. Overall, these findings argue that USP6NL overexpression generates a metabolic rewiring that is essential to foster the glycolytic demand of breast cancer cells and promote their proliferation.Significance: USP6NL overexpression leads to glycolysis addiction of breast cancer cells and presents a point of metabolic vulnerability for therapeutic targeting in a subset of aggressive basal-like breast tumors.Graphical Abstract: http://cancerres.aacrjournals.org/content/canres/78/13/3432/F1.large.jpg Cancer Res; 78(13); 3432-44. ©2018 AACR.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Neoplasias da Mama/patologia , Proteínas Ativadoras de GTPase/metabolismo , Transportador de Glucose Tipo 1/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Neoplasias da Mama/metabolismo , Neoplasias da Mama/mortalidade , Linhagem Celular Tumoral , Proliferação de Células , Conjuntos de Dados como Assunto , Receptores ErbB/metabolismo , Feminino , Proteínas Ativadoras de GTPase/genética , Amplificação de Genes , Glicólise , Humanos , Fosforilação , Prognóstico , Estabilidade Proteica , Proteólise , Análise de Sobrevida
6.
Mol Oncol ; 12(1): 3-20, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-29124875

RESUMO

The physiological function of the epidermal growth factor receptor (EGFR) is to regulate epithelial tissue development and homeostasis. In pathological settings, mostly in lung and breast cancer and in glioblastoma, the EGFR is a driver of tumorigenesis. Inappropriate activation of the EGFR in cancer mainly results from amplification and point mutations at the genomic locus, but transcriptional upregulation or ligand overproduction due to autocrine/paracrine mechanisms has also been described. Moreover, the EGFR is increasingly recognized as a biomarker of resistance in tumors, as its amplification or secondary mutations have been found to arise under drug pressure. This evidence, in addition to the prominent function that this receptor plays in normal epithelia, has prompted intense investigations into the role of the EGFR both at physiological and at pathological level. Despite the large body of knowledge obtained over the last two decades, previously unrecognized (herein defined as 'noncanonical') functions of the EGFR are currently emerging. Here, we will initially review the canonical ligand-induced EGFR signaling pathway, with particular emphasis to its regulation by endocytosis and subversion in human tumors. We will then focus on the most recent advances in uncovering noncanonical EGFR functions in stress-induced trafficking, autophagy, and energy metabolism, with a perspective on future therapeutic applications.


Assuntos
Neoplasias/patologia , Animais , Autofagia/fisiologia , Membrana Celular/metabolismo , Endocitose/fisiologia , Endossomos/metabolismo , Receptores ErbB/genética , Receptores ErbB/fisiologia , Humanos , Ligantes , Camundongos , Camundongos Knockout , Estresse Fisiológico
7.
Cancer Res ; 76(17): 5019-29, 2016 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-27364553

RESUMO

MET oncogene amplification is emerging as a major mechanism of acquired resistance to EGFR-directed therapy in lung and colorectal cancers. Furthermore, MET amplification predicts responsiveness to MET inhibitors currently in clinical trials. Among the anti-MET drugs available, ATP-competitive small-molecule kinase inhibitors abrogate receptor autophosphorylation and downstream activation of ERK1/2 and AKT, resulting in cell-cycle arrest. However, this antiproliferative effect allows persistence of a pool of cancer cells that are quiescent but alive. Once the inhibition is removed, rebound activation of MET-driven cell proliferative pathways and tumor growth may occur, an adverse event observed frequently in clinical settings after drug discontinuation. Here we show that inhibitor withdrawal prompts receptor phosphorylation to levels higher than those displayed at steady-state and generates a rebound effect pushing quiescent cancer cells back into the cell cycle, both in vitro and in experimental tumor models in vivo Mechanistically, we found that inhibitor treatment blocks MET endocytosis, causing a local increase in the number of receptors at the plasma membrane. Upon inhibitor washout, the receptor is readily rephosphorylated. The initial phosphorylation is not only increased but also prolonged in duration due to downmodulation of a phosphatase-mediated MET-negative feedback loop, which accompanies receptor internalization. Notably, treatment with a MET therapeutic antibody that induces proteolytic cleavage of the receptor at the cell surface substantially prevents this rebound effect, providing a rationale to combine or alternate these mechanistically different types of MET-targeted therapy. Cancer Res; 76(17); 5019-29. ©2016 AACR.


Assuntos
Anticorpos/farmacologia , Antineoplásicos/farmacologia , Recidiva Local de Neoplasia/patologia , Neoplasias Experimentais/patologia , Proteínas Proto-Oncogênicas c-met/metabolismo , Animais , Western Blotting , Linhagem Celular Tumoral , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Feminino , Imunofluorescência , Humanos , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Microscopia Confocal , Recidiva Local de Neoplasia/metabolismo , Neoplasias Experimentais/metabolismo , Inibidores de Proteínas Quinases/farmacologia , Ensaios Antitumorais Modelo de Xenoenxerto
8.
Mol Cell Proteomics ; 14(3): 621-34, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25573745

RESUMO

Endothelial cells (ECs) play a key role to maintain the functionality of blood vessels. Altered EC permeability causes severe impairment in vessel stability and is a hallmark of pathologies such as cancer and thrombosis. Integrating label-free quantitative proteomics data into genome-wide metabolic modeling, we built up a model that predicts the metabolic fluxes in ECs when cultured on a tridimensional matrix and organize into a vascular-like network. We discovered how fatty acid oxidation increases when ECs are assembled into a fully formed network that can be disrupted by inhibiting CPT1A, the fatty acid oxidation rate-limiting enzyme. Acute CPT1A inhibition reduces cellular ATP levels and oxygen consumption, which are restored by replenishing the tricarboxylic acid cycle. Remarkably, global phosphoproteomic changes measured upon acute CPT1A inhibition pinpointed altered calcium signaling. Indeed, CPT1A inhibition increases intracellular calcium oscillations. Finally, inhibiting CPT1A induces hyperpermeability in vitro and leakage of blood vessel in vivo, which were restored blocking calcium influx or replenishing the tricarboxylic acid cycle. Fatty acid oxidation emerges as central regulator of endothelial functions and blood vessel stability and druggable pathway to control pathological vascular permeability.


Assuntos
Carnitina O-Palmitoiltransferase/antagonistas & inibidores , Células Endoteliais/metabolismo , Ácidos Graxos/metabolismo , Metaboloma , Modelos Biológicos , Proteômica/métodos , Trifosfato de Adenosina/metabolismo , Animais , Células Endoteliais/citologia , Compostos de Epóxi/farmacologia , Células Endoteliais da Veia Umbilical Humana , Humanos , Técnicas In Vitro , Camundongos , Oxirredução , Consumo de Oxigênio , Permeabilidade
9.
Cell Calcium ; 56(3): 225-34, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25113159

RESUMO

Hydrogen sulphide (H2S) is a newly discovered gasotransmitter that regulates multiple steps in VEGF-induced angiogenesis. An increase in intracellular Ca(2+) concentration ([Ca(2+)]i) is central to endothelial proliferation and may be triggered by both VEGF and H2S. Albeit VEGFR-2 might serve as H2S receptor, the mechanistic relationship between VEGF- and H2S-induced Ca(2+) signals in endothelial cells is unclear. The present study aimed at assessing whether and how NaHS, a widely employed H2S donor, stimulates pro-angiogenic Ca(2+) signals in Ea.hy926 cells, a suitable surrogate for mature endothelial cells, and human endothelial progenitor cells (EPCs). We found that NaHS induced a dose-dependent increase in [Ca(2+)]i in Ea.hy926 cells. NaHS-induced Ca(2+) signals in Ea.hy926 cells did not require extracellular Ca(2+) entry, while they were inhibited upon pharmacological blockade of the phospholipase C/inositol-1,4,5-trisphosphate (InsP3) signalling pathway. Moreover, the Ca(2+) response to NaHS was prevented by genistein, but not by SU5416, which selectively inhibits VEGFR-2. However, VEGF-induced Ca(2+) signals were suppressed by dl-propargylglycine (PAG), which blocks the H2S-producing enzyme, cystathionine γ-lyase. Consistent with these data, VEGF-induced proliferation and migration were inhibited by PAG in Ea.hy926 cells, albeit NaHS alone did not influence these processes. Conversely, NaHS elevated [Ca(2+)]i only in a modest fraction of circulating EPCs, whereas neither VEGF-induced Ca(2+) oscillations nor VEGF-dependent proliferation were affected by PAG. Therefore, H2S-evoked elevation in [Ca(2+)]i is essential to trigger the pro-angiogenic Ca(2+) response to VEGF in mature endothelial cells, but not in their immature progenitors.


Assuntos
Células Progenitoras Endoteliais/metabolismo , Endotélio Vascular/metabolismo , Sulfeto de Hidrogênio/farmacologia , Inositol 1,4,5-Trifosfato/metabolismo , Transdução de Sinais/efeitos dos fármacos , Fator A de Crescimento do Endotélio Vascular/metabolismo , Adulto , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Citoplasma/metabolismo , Células Progenitoras Endoteliais/citologia , Células Progenitoras Endoteliais/efeitos dos fármacos , Endotélio Vascular/citologia , Endotélio Vascular/efeitos dos fármacos , Voluntários Saudáveis , Humanos , Fosfolipases Tipo C/metabolismo , Cicatrização/efeitos dos fármacos , Adulto Jovem
10.
Cell Physiol Biochem ; 33(4): 1205-14, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24752219

RESUMO

BACKGROUND: Hydrogen sulfide contributes to the reduction of oxidative stress-related injury in cardiomyocytes but the underlying mechanism is still unclear. AIMS: Here we investigated the role of voltage-operated calcium channels (VOCCs) as mediators of the beneficial effect of H2S against oxidative stress in cultured rat cardiomyoblasts (H9c2). METHODS: Intracellular calcium signals were measured by fluorimetric live cell imaging and cell viability by colorimetric assay. RESULTS: Treatment with H2S donor (NaHS 10 µM) or Nifedipine (10 µM) decreased resting intracellular calcium concentration [Ca]i, suggesting that L-type VOCCs are negatively modulated by H2S. In the presence of Nifedipine H2S was still able to lower [Ca]i, while co-incubation with Nifedipine and Ni(2+) 100 µM completely prevented H2S-dependent [Ca]i decrease, suggesting that both L-type and T-type VOCCs are inhibited by H2S. In addition, in the same experimental conditions, H2S triggered a slow increase of [Ca]i whose molecular nature remains to be clarified. Pretreatment of H9c2 with NaHS (10 µM) significantly prevented cell death induced by H2O2. This effect was mimicked by pretreatment with L-Type calcium channel inhibitor Nifedipine (10 µM). CONCLUSIONS: The data provide the first evidence that H2S protects rat cardiomyoblasts against oxidative challenge through the inhibition of L-type calcium channels.


Assuntos
Canais de Cálcio Tipo L/metabolismo , Canais de Cálcio Tipo T/metabolismo , Sulfeto de Hidrogênio/farmacologia , Estresse Oxidativo/efeitos dos fármacos , Animais , Cálcio/metabolismo , Bloqueadores dos Canais de Cálcio/farmacologia , Canais de Cálcio Tipo L/química , Canais de Cálcio Tipo T/química , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Peróxido de Hidrogênio/toxicidade , Miócitos Cardíacos/citologia , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Nifedipino/farmacologia , Ratos , Sulfetos/farmacologia
11.
Cell Calcium ; 53(2): 77-84, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22840338

RESUMO

An increasing body of evidence suggests the involvement of hydrogen sulfide (H(2)S) in different physiological and pathological processes. Similarly to the other gasotransmitters nitric oxide (NO) and carbon monoxide (CO), this bioactive compound is rapidly diffusible through the biological membranes and acts in a paracrine fashion. Despite the large amount of biological actions observed in vitro and in vivo upon stimulation with H(2)S donors, as well as by interfering with its synthesis, the molecular targets and mechanisms through which it exerts its intracellular effects are only partially known. A number of proteins are covalently modified by H(2)S through sulfhydration of specific cysteine residues. However, only in few cases their identity has been discovered and the functional role of this post-translational modification needs to be investigated in more detail. Great attention has been devoted to potassium channels, particularly K(ATP), as they are considered key mediators of H(2)S-induced effects, and their sulfhydration has been clearly demonstrated. Recently, different authors reported the ability of H(2)S to interfere with calcium homeostasis in neurons, cardiomyocytes and endothelial cells. Since calcium signaling is involved in all cell processes, these observations attracted increasing attention from basic biology and medicine. Although some effects of H(2)S on calcium signals can be ascribed to K(ATP) modulation, there is growing consensus about the existence of other targets for the gasotransmitter. Some of them are Ca(2+)-permeable channels. In this review we discuss the state of the art in this specific field, providing an updated report of H(2)S interaction with Ca(2+) channels and its functional outcomes.


Assuntos
Canais de Cálcio Tipo L/metabolismo , Sulfeto de Hidrogênio/metabolismo , Canais de Cátion TRPV/metabolismo , Canais de Potencial de Receptor Transitório/metabolismo , Canais de Cálcio Tipo T/metabolismo , Monóxido de Carbono/metabolismo , Humanos , Sulfeto de Hidrogênio/química , Óxido Nítrico/metabolismo
12.
Recent Pat Anticancer Drug Discov ; 8(1): 27-37, 2013 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-22530660

RESUMO

Blood vessels and endothelial cells (ECs) are highly versatile in order to accomplish local tissutal needs in the physiological and pathological conditions. Tumor vasculature, in particular, exhibits special morphological and functional features, partly due to the peculiarity of tumor-derived ECs (TECs). This is of great importance for the discovery of selective molecular targets potentially suitable to interfere with tumor growth and spread. In normal ECs, proangiogenic calcium signaling is mediated by different calcium channels, mainly TRPs and Orai, that could play a pivotal role in physiological angiogenesis. They are regulated through multiple mechanisms, involving their interaction with bioactive lipids (arachidonic acid and its metabolites), nitrosylation, sulfhydration, phosphorylation, cytoskeleton-mediated membrane trafficking, and calcium stores depletion. On the other hand, proangiogenic calcium events in TECs have been investigated only recently and their characterization is still preliminary. ECs obtained from human breast and renal carcinomas (B-TECs and R-TECs respectively) display altered calcium signals, which are associated with modified expression and function of TRP channels. Here, we review the state of the art in the field of calcium signaling and tumor vascularization, the related recent literature and patents. Finally, we provide some suggestions for future developments.


Assuntos
Inibidores da Angiogênese/farmacologia , Bloqueadores dos Canais de Cálcio/farmacologia , Canais de Cálcio/efeitos dos fármacos , Células Endoteliais/efeitos dos fármacos , Neoplasias/tratamento farmacológico , Neovascularização Patológica/prevenção & controle , Inibidores da Angiogênese/química , Animais , Bloqueadores dos Canais de Cálcio/química , Canais de Cálcio/metabolismo , Desenho de Fármacos , Células Endoteliais/metabolismo , Humanos , Legislação de Medicamentos , Terapia de Alvo Molecular , Neoplasias/irrigação sanguínea , Neoplasias/metabolismo , Neoplasias/patologia , Neovascularização Patológica/metabolismo , Patentes como Assunto
13.
Am J Physiol Cell Physiol ; 302(1): C9-15, 2012 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-21832241

RESUMO

Tumor vascularization is a critical process that determines tumor growth and metastasis. In the last decade new experimental evidence obtained from in vitro and in vivo studies have challenged the classical angiogenesis model forcing us to consider new scenarios for tumor neovascularization. In particular, the genetic stability of tumor-derived endothelial cells (TECs) has been recently questioned in several studies, which show that TECs, as well as pericytes, differ significantly from their normal counterparts at genetic and functional levels. In addition to such an epigenetic action of tumor microenvironment on endothelial cells (ECs) commitment, the distinct characteristics of TECs could be due to differences in their origin compared with preexisting differentiated ECs. Intracellular Ca(2+) signals are involved at different critical phases in the regulation of the complex process of angiogenesis and tumor progression. These signals are generated by a wide variety of intrinsic and extrinsic factors. Several key components of Ca(2+) signaling including Ca(2+) channels in the plasma membrane, endoplasmic reticulum, calcium pumps, and mitochondria contribute to the generation, amplitude, and frequency of these Ca(2+) change. In particular, several members of the transient receptor potential (TRP) family of calcium-permeable channels have profound effects on the function of ECs. Because of its multifaceted role in the control of cell function, proliferation, and motility, TRP channels have been suggested as a potential molecular target for control of tumor neovascularization. Since plasma membrane Ca(2+) channels are easily and directly accessible via the bloodstream, they are potential targets for a number of pharmacological and antibody-targeted therapeutic strategies, with specificity being the main limitation. In this review we discuss recent advances in understanding the role of Ca(2+) channels, with specific reference to TRP channels, in tumor vascularization process.


Assuntos
Marcação de Genes/métodos , Canais Iônicos/metabolismo , Proteínas de Neoplasias/metabolismo , Neoplasias/irrigação sanguínea , Neoplasias/metabolismo , Neovascularização Patológica/metabolismo , Canais de Potencial de Receptor Transitório/metabolismo , Animais , Humanos , Canais Iônicos/genética , Proteínas de Neoplasias/sangue , Proteínas de Neoplasias/genética , Neoplasias/genética , Neovascularização Patológica/genética , Canais de Potencial de Receptor Transitório/genética
14.
Free Radic Biol Med ; 51(9): 1765-73, 2011 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-21875664

RESUMO

Hydrogen sulfide (H(2)S) is a gasotransmitter that plays several roles in various tissues, including the cardiovascular system. Because it has been recently proposed to act as a mediator of angiogenesis progression, here we investigate the effects of H(2)S in a well-established model of tumor angiogenesis: endothelial cells obtained from human breast carcinoma (B-TECs). Ca(2+) imaging and patch-clamp experiments reveal that acute perfusion with NaHS, a widely employed H(2)S donor, activates cytosolic calcium (Ca(c)) increase, as well as potassium and nonselective cationic currents, in B-TECs. Stimulation with NaHS in the same concentration range (1 nM-200 µM) evoked Ca(c) signals also in "normal" human microvascular endothelial cells (HMVECs), but the amplitude was significantly lower. Moreover, although NaHS failed to promote either migration or proliferation on HMVECs, B-TEC migration was enhanced at low-micromolar NaHS concentrations (1-10 µM). Remarkably H(2)S mediates tumor proangiogenic signaling triggered by vascular endothelial growth factor (VEGF). B-TECs pretreated with dl-propargylglycine (5mM, 30 min), an inhibitor of the H(2)S-producing enzyme cystathionine γ-lyase, showed drastically reduced migration and Ca(c) signals induced by VEGF (20 ng/ml). We conclude that H(2)S plays a role in proangiogenic signaling of tumor-derived but not normal human ECs. Furthermore the ability of this gasotransmitter to interfere with B-TEC responsiveness to VEGF suggests that it could be an interesting target for antiangiogenic strategies in tumor treatment.


Assuntos
Neoplasias da Mama/patologia , Cálcio/metabolismo , Carcinoma Ductal de Mama/patologia , Células Endoteliais/efeitos dos fármacos , Sulfeto de Hidrogênio/farmacologia , Transdução de Sinais/efeitos dos fármacos , Neoplasias da Mama/metabolismo , Carcinoma Ductal de Mama/metabolismo , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Relação Dose-Resposta a Droga , Células Endoteliais/metabolismo , Feminino , Humanos , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...