Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Sci Transl Med ; 16(736): eadf9874, 2024 Feb 28.
Artigo em Inglês | MEDLINE | ID: mdl-38416843

RESUMO

Targeting aromatase deprives ER+ breast cancers of estrogens and is an effective therapeutic approach for these tumors. However, drug resistance is an unmet clinical need. Lipidomic analysis of long-term estrogen-deprived (LTED) ER+ breast cancer cells, a model of aromatase inhibitor resistance, revealed enhanced intracellular lipid storage. Functional metabolic analysis showed that lipid droplets together with peroxisomes, which we showed to be enriched and active in the LTED cells, controlled redox homeostasis and conferred metabolic adaptability to the resistant tumors. This reprogramming was controlled by acetyl-CoA-carboxylase-1 (ACC1), whose targeting selectively impaired LTED survival. However, the addition of branched- and very long-chain fatty acids reverted ACC1 inhibition, a process that was mediated by peroxisome function and redox homeostasis. The therapeutic relevance of these findings was validated in aromatase inhibitor-treated patient-derived samples. Last, targeting ACC1 reduced tumor growth of resistant patient-derived xenografts, thus identifying a targetable hub to combat the acquisition of estrogen independence in ER+ breast cancers.


Assuntos
Neoplasias da Mama , Humanos , Feminino , Neoplasias da Mama/patologia , Inibidores da Aromatase/farmacologia , Inibidores da Aromatase/uso terapêutico , Peroxissomos/metabolismo , Peroxissomos/patologia , Acetil-CoA Carboxilase , Gotículas Lipídicas/metabolismo , Gotículas Lipídicas/patologia , Linhagem Celular Tumoral , Estrogênios/metabolismo , Resistencia a Medicamentos Antineoplásicos
3.
Cancer Res ; 82(7): 1267-1282, 2022 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-35135811

RESUMO

Lactate is an abundant oncometabolite in the tumor environment. In prostate cancer, cancer-associated fibroblasts (CAF) are major contributors of secreted lactate, which can be taken up by cancer cells to sustain mitochondrial metabolism. However, how lactate impacts transcriptional regulation in tumors has yet to be fully elucidated. Here, we describe a mechanism by which CAF-secreted lactate is able to increase the expression of genes involved in lipid metabolism in prostate cancer cells. This regulation enhanced intracellular lipid accumulation in lipid droplets (LD) and provided acetyl moieties for histone acetylation, establishing a regulatory loop between metabolites and epigenetic modification. Inhibition of this loop by targeting the bromodomain and extraterminal protein family of histone acetylation readers suppressed the expression of perilipin 2 (PLIN2), a crucial component of LDs, disrupting lactate-dependent lipid metabolic rewiring. Inhibition of this CAF-induced metabolic-epigenetic regulatory loop in vivo reduced growth and metastasis of prostate cancer cells, demonstrating its translational relevance as a therapeutic target in prostate cancer. Clinically, PLIN2 expression was elevated in tumors with a higher Gleason grade and in castration-resistant prostate cancer compared with primary prostate cancer. Overall, these findings show that lactate has both a metabolic and an epigenetic role in promoting prostate cancer progression. SIGNIFICANCE: This work shows that stromal-derived lactate induces accumulation of lipid droplets, stimulates epigenetic rewiring, and fosters metastatic potential in prostate cancer.


Assuntos
Metabolismo dos Lipídeos , Neoplasias da Próstata , Epigênese Genética , Humanos , Ácido Láctico/metabolismo , Metabolismo dos Lipídeos/genética , Masculino , Próstata/patologia , Neoplasias da Próstata/patologia
4.
Cells ; 10(12)2021 12 02.
Artigo em Inglês | MEDLINE | ID: mdl-34943903

RESUMO

The tumour microenvironment (TME) is now recognised as a hallmark of cancer, since tumour:stroma crosstalk supports the key steps of tumour growth and progression. The dynamic co-evolution of the tumour and stromal compartments may alter the surrounding microenvironment, including the composition in metabolites and signalling mediators. A growing number of evidence reports the involvement of the endocannabinoid system (ECS) in cancer. ECS is composed by a complex network of ligands, receptors, and enzymes, which act in synergy and contribute to several physiological but also pathological processes. Several in vitro and in vivo evidence show that ECS deregulation in cancer cells affects proliferation, migration, invasion, apoptosis, and metastatic potential. Although it is still an evolving research, recent experimental evidence also suggests that ECS can modulate the functional behaviour of several components of the TME, above all the immune cells, endothelial cells and stromal components. However, the role of ECS in the tumour:stroma interplay remains unclear and research in this area is particularly intriguing. This review aims to shed light on the latest relevant findings of the tumour response to ECS modulation, encouraging a more in-depth analysis in this field. Novel discoveries could be promising for novel anti-tumour approaches, targeting the microenvironmental components and the supportive tumour:stroma crosstalk, thereby hindering tumour development.


Assuntos
Endocanabinoides/genética , Neoplasias/genética , Microambiente Tumoral/genética , Apoptose/genética , Movimento Celular/genética , Proliferação de Células/genética , Endocanabinoides/metabolismo , Células Endoteliais/metabolismo , Células Endoteliais/patologia , Humanos , Invasividade Neoplásica/genética , Invasividade Neoplásica/patologia , Metástase Neoplásica/genética , Neoplasias/metabolismo , Neoplasias/patologia , Transdução de Sinais/genética
5.
Nucleic Acids Res ; 48(16): 8943-8958, 2020 09 18.
Artigo em Inglês | MEDLINE | ID: mdl-32697292

RESUMO

The epithelial-to-mesenchymal transition (EMT) is a complex transcriptional program induced by transforming growth factor ß1 (TGF-ß1). Histone lysine-specific demethylase 1 (LSD1) has been recognized as a key mediator of EMT in cancer cells, but the precise mechanism that underlies the activation and repression of EMT genes still remains elusive. Here, we characterized the early events induced by TGF-ß1 during EMT initiation and establishment. TGF-ß1 triggered, 30-90 min post-treatment, a nuclear oxidative wave throughout the genome, documented by confocal microscopy and mass spectrometry, mediated by LSD1. LSD1 was recruited with phosphorylated SMAD2/3 to the promoters of prototypic genes activated and repressed by TGF-ß1. After 90 min, phospho-SMAD2/3 downregulation reduced the complex and LSD1 was then recruited with the newly synthesized SNAI1 and repressors, NCoR1 and HDAC3, to the promoters of TGF-ß1-repressed genes such as the Wnt soluble inhibitor factor 1 gene (WIF1), a change that induced a late oxidative burst. However, TGF-ß1 early (90 min) repression of transcription also required synchronous signaling by reactive oxygen species and the stress-activated kinase c-Jun N-terminal kinase. These data elucidate the early events elicited by TGF-ß1 and the priming role of DNA oxidation that marks TGF-ß1-induced and -repressed genes involved in the EMT.


Assuntos
DNA/metabolismo , Transição Epitelial-Mesenquimal/genética , Histona Desmetilases/fisiologia , Proteína Smad2/fisiologia , Fator de Crescimento Transformador beta1/fisiologia , Linhagem Celular Tumoral , Regulação Neoplásica da Expressão Gênica , Humanos
6.
Front Oncol ; 10: 396, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32266157

RESUMO

Neoplastic tissues are composed not only by tumor cells but also by several non-transformed stromal cells, such as cancer-associated fibroblasts, endothelial and immune cells, that actively participate to tumor progression. Starting from the very beginning of carcinogenesis, tumor cells, through the release of paracrine soluble factors and vesicles, i.e., exosomes, modify the behavior of the neighboring cells, so that they can give efficient support for cancer cell proliferation and spreading. A mandatory role in tumor progression has been recently acknowledged to metabolic deregulation. Beside undergoing a metabolic reprogramming coherent to their high proliferation rate, tumor cells also rewire the metabolic assets of their stromal cells, educating them to serve as nutrient donors. Hence, an alteration in the composition and in the flow rate of many nutrients within tumor microenvironment has been associated with malignancy progression. This review is focused on metabolic remodeling of the different cell populations within tumor microenvironment, dealing with reciprocal re-education through the symbiotic sharing of metabolites, behaving both as nutrients and as transcriptional regulators, describing their impact on tumor growth and metastasis.

7.
Cells ; 9(3)2020 03 10.
Artigo em Inglês | MEDLINE | ID: mdl-32164162

RESUMO

The majority of breast cancers express the estrogen receptor (ER) and are dependent on estrogen for their growth and survival. Endocrine therapy (ET) is the standard of care for these tumors. However, a superior outcome is achieved in a subset of ER positive (ER+)/human epidermal growth factor receptor 2 negative (HER2-) metastatic breast cancer patients when ET is administrated in combination with a cyclin-dependent kinases 4 and 6 (CDK4/6) inhibitor, such as palbociclib. Moreover, CDK4/6 inhibitors are currently being tested in ER+/HER2+ breast cancer and reported encouraging results. Despite the clinical advances of a combinatorial therapy using ET plus CDK4/6 inhibitors, potential limitations (i.e., resistance) could emerge and the metabolic adaptations underlying such resistance warrant further elucidation. Here we investigate the glucose-dependent catabolism in a series of isogenic ER+ breast cancer cell lines sensitive to palbociclib and in their derivatives with acquired resistance to the drug. Importantly, ER+/HER2- and ER+/HER2+ cell lines show a different degree of glucose dependency. While ER+/HER2- breast cancer cells are characterized by enhanced aerobic glycolysis at the time of palbociclib sensitivity, ER+/HER2+ cells enhance their glycolytic catabolism at resistance. This metabolic phenotype was shown to have prognostic value and was targeted with multiple approaches offering a series of potential scenarios that could be of clinical relevance.


Assuntos
Antineoplásicos/uso terapêutico , Neoplasias da Mama/tratamento farmacológico , Glucose/metabolismo , Piperazinas/uso terapêutico , Piridinas/uso terapêutico , Antineoplásicos/farmacologia , Neoplasias da Mama/genética , Linhagem Celular Tumoral , Feminino , Humanos , Piperazinas/farmacologia , Piridinas/farmacologia , Transfecção
8.
Cell Rep ; 28(1): 104-118.e8, 2019 07 02.
Artigo em Inglês | MEDLINE | ID: mdl-31269432

RESUMO

Endocrine therapy (ET) is the standard of care for estrogen receptor-positive (ER+) breast cancers. Despite its efficacy, ∼40% of women relapse with ET-resistant (ETR) disease. A global transcription analysis in ETR cells reveals a downregulation of the neutral and basic amino acid transporter SLC6A14 governed by enhanced miR-23b-3p expression, resulting in impaired amino acid metabolism. This altered amino acid metabolism in ETR cells is supported by the activation of autophagy and the enhanced import of acidic amino acids (aspartate and glutamate) mediated by the SLC1A2 transporter. The clinical significance of these findings is validated by multiple orthogonal approaches in a large cohort of ET-treated patients, in patient-derived xenografts, and in in vivo experiments. Targeting these amino acid metabolic dependencies resensitizes ETR cells to therapy and impairs the aggressive features of ETR cells, offering predictive biomarkers and potential targetable pathways to be exploited to combat or delay ETR in ER+ breast cancers.


Assuntos
Sistemas de Transporte de Aminoácidos/metabolismo , Ácido Aspártico/metabolismo , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Resistencia a Medicamentos Antineoplásicos/genética , Transportador 2 de Aminoácido Excitatório/metabolismo , Ácido Glutâmico/metabolismo , Sistemas de Transporte de Aminoácidos/genética , Sistemas de Transporte de Aminoácidos Neutros/genética , Animais , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/genética , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/genética , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Receptor alfa de Estrogênio/genética , Receptor alfa de Estrogênio/metabolismo , Transportador 2 de Aminoácido Excitatório/genética , Feminino , Fator de Transcrição GATA2/genética , Fator de Transcrição GATA2/metabolismo , Cromatografia Gasosa-Espectrometria de Massas , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/genética , Humanos , Camundongos , Camundongos Endogâmicos BALB C , MicroRNAs/genética , MicroRNAs/metabolismo , Metástase Neoplásica , Prognóstico , Transcriptoma/genética , Transplante Heterólogo
9.
Oncogene ; 38(27): 5339-5355, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-30936458

RESUMO

Cancer-associated fibroblasts (CAFs) are the major cellular stromal component of many solid tumors. In prostate cancer (PCa), CAFs establish a metabolic symbiosis with PCa cells, contributing to cancer aggressiveness through lactate shuttle. In this study, we report that lactate uptake alters the NAD+/NADH ratio in the cancer cells, which culminates with SIRT1-dependent PGC-1α activation and subsequent enhancement of mitochondrial mass and activity. The high exploitation of mitochondria results in tricarboxylic acid cycle deregulation, accumulation of oncometabolites and in the altered expression of mitochondrial complexes, responsible for superoxide generation. Additionally, cancer cells hijack CAF-derived functional mitochondria through the formation of cellular bridges, a phenomenon that we observed in both in vitro and in vivo PCa models. Our work reveals a crucial function of tumor mitochondria as the energy sensors and transducers of CAF-dependent metabolic reprogramming and underscores the reliance of PCa cells on CAF catabolic activity and mitochondria trading.


Assuntos
Mitocôndrias/metabolismo , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/patologia , Linhagem Celular Tumoral , Ciclo do Ácido Cítrico , Fibroblastos/patologia , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Masculino , NAD/metabolismo , Invasividade Neoplásica , Fosforilação Oxidativa , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Sirtuína 1/metabolismo
10.
Curr Cancer Drug Targets ; 19(10): 807-816, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30648509

RESUMO

BACKGROUND: The bisphosphonate Zoledronic acid (ZA) is a potent osteoclast inhibitor currently used in the clinic to reduce osteoporosis and cancer-induced osteolysis. Moreover, ZA exerts an anti-tumor effect in several tumors. Despite this evidence, the relevance of ZA in prostate cancer (PCa) is not completely understood. OBJECTIVE: To investigate the effect of ZA administration on the invasive properties of PC3 cells, which are characterised by RhoA-dependent amoeboid motility. METHODS: The effect of ZA administration on the in vitro invasive properties of PC3 cells was evaluated by cell migration in 3D collagen matrices, immunofluorescence and Boyden assays or transendothelial migration. Lung retention and colonization assays were performed to assess the efficacy of ZA administration in vivo. RESULTS: PC3 cells are characterised by RhoA-dependent amoeboid motility. We now report a clear inhibition of in vitro PC3 cell invasion and RhoA activity upon ZA treatment. Moreover, to confirm a specific role of ZA in the inhibition of amoeboid motility of PC3 cells, we demonstrate that ZA interferes only partially with PC3 cells showing a mesenchymal phenotype due to both treatment with conditioned medium of cancer associated fibroblasts or to the acquisition of chemoresistance. Furthermore, we demonstrate that ZA impairs adhesion to endothelial cells and the trans-endothelial cell migration, two essential properties characterising amoeboid motility and PC3 metastatic dissemination. In vivo experiments prove the ability of ZA to inhibit the metastatic process of PC3 cells as shown by the decrease in lung colonization. CONCLUSION: This study demonstrates that ZA inhibits Rho-dependent amoeboid motility of PC3 cells, thus suggesting ZA as a potential therapy to impede the metastatic dissemination of PC3 cells.


Assuntos
Movimento Celular , Osteoclastos/efeitos dos fármacos , Neoplasias da Próstata/tratamento farmacológico , Ácido Zoledrônico/farmacologia , Proteína rhoA de Ligação ao GTP/antagonistas & inibidores , Animais , Conservadores da Densidade Óssea/farmacologia , Humanos , Masculino , Camundongos , Camundongos SCID , Células PC-3 , Neoplasias da Próstata/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto , Proteína rhoA de Ligação ao GTP/metabolismo
11.
J Cell Physiol ; 234(6): 8274-8285, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30378132

RESUMO

Cancer progression is strictly dependent on the relationship between tumor cells and the surrounding stroma, which supports cancer malignancy promoting several crucial steps of tumor progression, including the execution of the epithelial to mesenchymal transition (EMT) associated with enhancement in cell invasion, resistance to both anoikis and chemotherapeutic treatments. Recently it has been highlighted the central role of microRNAs (miRNAs) as regulators of tumor progression. Notably, in several tumors a strong deregulation of miRNAs is observed, supporting proliferation, invasion, and metabolic reprogramming of tumor cells. Here we demonstrated that cancer-associated fibroblasts induce a downregulation of miR-1247 in prostate cancer (PCa) cells. We proved that miR-1247 repression is functional for the achievement of EMT and increased cell invasion as well as stemness traits. These phenomena contribute to promote the metastatic potential of PCa cells as demonstrated by increased lung colonization in in vivo experiments. Moreover, as a consequence of miR-1247 downregulation, we observed a correlated increased expression level of neuropilin-1, a miR-1247 target involved as a coreceptor in the epidermal growth factor receptor signaling. Taken together, our data highlight miR-1247 as a potential target for molecular therapies aimed to block the progression and diffusion of PCa.


Assuntos
Transição Epitelial-Mesenquimal/genética , MicroRNAs/genética , Neuropilina-1/genética , Neoplasias da Próstata/genética , Proliferação de Células/genética , Reprogramação Celular/genética , Regulação Neoplásica da Expressão Gênica/genética , Humanos , Masculino , Invasividade Neoplásica/genética , Invasividade Neoplásica/patologia , Metástase Neoplásica , Neoplasias da Próstata/patologia , Células Estromais/metabolismo , Células Estromais/patologia
12.
Org Biomol Chem ; 16(33): 6086-6095, 2018 08 22.
Artigo em Inglês | MEDLINE | ID: mdl-30091781

RESUMO

GM3-ganglioside is known to be involved in melanoma proliferation. In order to modulate metastatic-related events, we have functionalized multi-walled carbon nanotubes (MWCNTs) with multiple copies of a GM3-lactone mimetic. The MWCNTs proved to guarantee the appropriate spatial arrangement of the mimetic allowing a stronger inhibition of migration and invasiveness of human melanoma (A375) cells compared to other multivalent constructs reported before. In addition, the effect of the multivalent tubular conjugate on the inhibition of specific tyrosine kinases, which are associated with the ganglioside complexes within the membrane domains, was demonstrated. Finally, the short-term fate of the conjugate was assessed, for the first time, by means of the 1H NMR relaxometry technique by exploiting the signal arising from the CNTs.


Assuntos
Antineoplásicos/química , Antineoplásicos/farmacologia , Materiais Biomiméticos/química , Gangliosídeo G(M3)/análogos & derivados , Melanoma/patologia , Nanotubos de Carbono/química , Linhagem Celular Tumoral , Gangliosídeo G(M3)/química , Humanos , Modelos Moleculares , Conformação Molecular , Metástase Neoplásica
13.
J Med Chem ; 60(21): 9003-9011, 2017 11 09.
Artigo em Inglês | MEDLINE | ID: mdl-29048889

RESUMO

Oxaliplatin (OXA) is a valuable and largely used cancer drug which induces a serious and intractable neuropathy. The lipoyl-homotaurine derivative (ADM_12) reverts in vivo OXA-induced neuropathy, and it is an effective antagonist of the nociceptive sensor channel TRPA1. Unprecedentedly, this safe analgesic showed a synergy with OXA in vitro and proved to inhibit CA IX, a relevant therapeutic target, clearly interfering with pancreatic cancer cells' aggressiveness.


Assuntos
Anidrase Carbônica IX/antagonistas & inibidores , Neoplasias/tratamento farmacológico , Compostos Organoplatínicos/toxicidade , Doenças do Sistema Nervoso Periférico/tratamento farmacológico , Taurina/análogos & derivados , Antineoplásicos/farmacologia , Antineoplásicos/toxicidade , Inibidores da Anidrase Carbônica/farmacologia , Linhagem Celular Tumoral , Sinergismo Farmacológico , Humanos , Neoplasias/complicações , Neoplasias/patologia , Compostos Organoplatínicos/uso terapêutico , Oxaliplatina , Neoplasias Pancreáticas/tratamento farmacológico , Neoplasias Pancreáticas/patologia , Doenças do Sistema Nervoso Periférico/induzido quimicamente , Canal de Cátion TRPA1/antagonistas & inibidores , Taurina/química , Taurina/farmacologia
14.
Oncotarget ; 8(1): 118-132, 2017 Jan 03.
Artigo em Inglês | MEDLINE | ID: mdl-27223431

RESUMO

Zoledronic acid (ZA) is a biphosphonate used for osteoporosis treatment and also proved to be effective to reduce the pain induced by bone metastases when used as adjuvant therapy in solid cancers. However, it has been recently proposed that ZA could have direct anti-tumour effects, although the molecular mechanism is unknown. We herein unravel a novel anti-tumour activity of ZA in prostate cancer (PCa), by targeting the pro-tumorigenic properties of both stromal and immune cells. Particularly, we demonstrate that ZA impairs PCa-induced M2-macrophages polarization, reducing their pro-invasive effect on tumour cells and their pro-angiogenic features. Crucially, ZA administration reverts cancer associated fibroblasts (CAFs) activation by targeting the mevalonate pathway and RhoA geranyl-geranylation, thereby impairing smooth muscle actin-α fibers organization, a prerequisite of fibroblast activation. Moreover, ZA prevents the M2 macrophages-mediated activation of normal fibroblast, highlighting the broad efficacy of this drug on tumour microenvironment. These results are confirmed in a metastatic xenograft PCa mouse model in which ZA-induced stromal normalization impairs cancer-stromal cells crosstalk, resulting in a significant reduction of primary tumour growth and metastases. Overall these findings reinforce the efficacy of ZA as a potential therapeutic approach to reduce cancer aggressiveness, by abrogating the supportive role of tumour microenvironment.


Assuntos
Fibroblastos Associados a Câncer/efeitos dos fármacos , Fibroblastos Associados a Câncer/metabolismo , Difosfonatos/farmacologia , Imidazóis/farmacologia , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Neoplasias da Próstata/imunologia , Neoplasias da Próstata/metabolismo , Células Estromais/efeitos dos fármacos , Células Estromais/metabolismo , Fibroblastos Associados a Câncer/patologia , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Movimento Celular/imunologia , Humanos , Ativação de Macrófagos/efeitos dos fármacos , Ativação de Macrófagos/imunologia , Macrófagos/imunologia , Macrófagos/patologia , Masculino , Ácido Mevalônico/metabolismo , Metástase Neoplásica , Neovascularização Patológica/imunologia , Neovascularização Patológica/metabolismo , Fenótipo , Neoplasias da Próstata/patologia , Microambiente Tumoral/efeitos dos fármacos , Microambiente Tumoral/imunologia , Ácido Zoledrônico , Proteína rhoA de Ligação ao GTP/metabolismo
15.
Stem Cells ; 34(10): 2536-2547, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27300750

RESUMO

Tumor stromal cells can supply appropriate signals that may develop aggressive phenotypes of carcinoma cells and establish a complex scenario which culminates in metastasis. Recent works proposed that bone marrow-derived mesenchymal stem cells (MSC) are recruited to primary tumors. However, the exact functions of these cells in the tumor microenvironment are not well characterized, as it is reported that MSC can either promote or inhibit tumor progression. In the present study, we aim at investigating the signaling molecules which regulate the interplay between MSC, prostate carcinoma (PCa) cells and two important cellular types constituting the tumor-associated stroma, macrophages and fibroblasts, during their progression toward malignancy. We identified TGF-ß1 as a crucial molecule able to attract MSC recruitment both to PCa cells as well as to tumor stroma components. Moreover, PCa- and tumor stroma-secreted TGF-ß1 is important to induce MSC transdifferentiation into carcinoma-associated fibroblast (CAF)-like cells. Consequently, the CAF-like phenotype acquired by MSC is central to promote tumor progression related effects. Thus, tumor-educated MSC enhance PCa invasiveness compared to nonactivated MSC. Additionally, differing from normal MSC, CAF-like MSC perform vascular mimicry and recruit monocytes, which can be further polarized to M2 macrophages within the PCa environment. Our findings indicate a prominent role for TGF-ß1 in MSC mobilization and activation strengthened by the fact that the blockade of TGF-ß1 signaling impairs MSC promotion of PCa progression. Stem Cells 2016;34:2536-2547.


Assuntos
Fibroblastos Associados a Câncer/patologia , Células-Tronco Mesenquimais/citologia , Neoplasias da Próstata/patologia , Fator de Crescimento Transformador beta1/metabolismo , Microambiente Tumoral , Biomarcadores Tumorais/metabolismo , Linhagem Celular Tumoral , Transdiferenciação Celular , Fatores Quimiotáticos/metabolismo , Humanos , Masculino , Células-Tronco Mesenquimais/metabolismo , Células Estromais/metabolismo
16.
ACS Med Chem Lett ; 7(1): 28-33, 2016 Jan 14.
Artigo em Inglês | MEDLINE | ID: mdl-26819661

RESUMO

Two tetravalent architectures, the glycocalix 7 and the RAFT 9, presenting four residues of a GM-3 ganglioside lactone mimetic, target the host compartment of melanoma and significantly abrogate the effect induced by cancer-associated fibroblasts (CAFs) contact + hypoxia in the motility and invasiveness of tumor cells. The data reported support the involvement of glycosphingolipids (GSLs) in hypoxia and show an interesting role played by compound 9 in targeting melanoma cells thereby interfering with melanoma progression. The unprecedented findings reported for the glycocluster 9 may contribute to the understanding of the critical and complex interactions between tumor cells and their local environment paving the way for new therapeutic agents.

17.
Oncotarget ; 6(27): 24061-74, 2015 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-26183399

RESUMO

Cancer associated fibroblasts (CAFs) are key determinants of cancer progression. In prostate carcinoma (PCa), CAFs induce epithelial-mesenchymal transition (EMT) and metabolic reprogramming of PCa cells towards oxidative phosphorylation (OXPHOS), promoting tumor growth and metastatic dissemination. We herein establish a novel role for pyruvate kinase M2 (PKM2), an established effector of Warburg-like glycolytic behavior, in OXPHOS metabolism induced by CAFs. Indeed, CAFs promote PKM2 post-translational modifications, such as cysteine oxidation and Src-dependent tyrosine phosphorylation, allowing nuclear migration of PKM2 and the formation of a trimeric complex with hypoxia inducible factor-1α (HIF-1α) and the transcriptional repressor Differentially Expressed in Chondrocytes-1 (DEC1). DEC1 recruitment is mandatory for downregulating miR205 expression, thereby fostering EMT execution and metabolic switch toward OXPHOS. Furthermore, the analysis of a cohort of PCa patients reveals a significant positive correlation between PKM2 nuclear localization and cancer aggressiveness, thereby validating our in vitro observations. Crucially, in vitro and in vivo pharmacological targeting of PKM2 nuclear translocation using DASA-58, as well as metformin, impairs metastatic dissemination of PCa cells in SCID mice. Our study indicates that impairing the metabolic tumor:stroma interplay by targeting the PKM2/OXPHOS axis, may be a valuable novel therapeutic approach in aggressive prostate carcinoma.


Assuntos
Transporte Ativo do Núcleo Celular , Proteínas de Transporte/metabolismo , Proteínas de Membrana/metabolismo , Neoplasias da Próstata/patologia , Piruvato Quinase/metabolismo , Hormônios Tireóideos/metabolismo , Animais , Sítios de Ligação , Carcinoma/metabolismo , Carcinoma/patologia , Linhagem Celular Tumoral , Núcleo Celular/metabolismo , Estudos de Coortes , Cisteína/química , Fibroblastos/metabolismo , Glucose/química , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Imuno-Histoquímica , Ácido Láctico/química , Masculino , Metformina/química , Camundongos , Camundongos SCID , MicroRNAs/metabolismo , Metástase Neoplásica , Oxirredução , Fosforilação Oxidativa , Oxigênio/química , Neoplasias da Próstata/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Proteínas de Ligação a Hormônio da Tireoide
18.
Oncotarget ; 6(7): 4615-32, 2015 Mar 10.
Artigo em Inglês | MEDLINE | ID: mdl-25474135

RESUMO

Stress has an emerging role in cancer and targeting stress-related ß-adrenergic receptors (AR) has been proposed as a potential therapeutic approach in melanoma. Here we report that ß3-AR expression correlates with melanoma aggressiveness. In addition, we highlight that ß3-AR expression is not only restricted to cancer cells, but it is also expressed in vivo in stromal, inflammatory and vascular cells of the melanoma microenvironment. Particularly, we demonstrated that ß3-AR can (i) instruct melanoma cells to respond to environmental stimuli, (ii) enhance melanoma cells response to stromal fibroblasts and macrophages, (iii) increase melanoma cell motility and (iv) induce stem-like traits. Noteworthy, ß3-AR activation in melanoma accessory cells drives stromal reactivity by inducing pro-inflammatory cytokines secretion and de novo angiogenesis, sustaining tumor growth and melanoma aggressiveness. ß3-ARs also play a mandatory role in the recruitment to tumor sites of circulating stromal cells precursors, in the differentiation of these cells towards different lineages, further favoring tumor inflammation, angiogenesis and ultimately melanoma malignancy. Our findings validate selective ß3-AR antagonists as potential promising anti-metastatic agents. These could be used to complement current therapeutic approaches for melanoma patients (e.g. propranolol) by targeting non-neoplastic stromal cells, hence reducing therapy resistance of melanoma.


Assuntos
Agonistas alfa-Adrenérgicos/farmacologia , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Melanoma/patologia , Norepinefrina/farmacologia , Receptores Adrenérgicos beta 3/metabolismo , Neoplasias Cutâneas/patologia , Microambiente Tumoral/efeitos dos fármacos , Apoptose , Proliferação de Células , Células Cultivadas , Progressão da Doença , Fibroblastos/citologia , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Citometria de Fluxo , Células Endoteliais da Veia Umbilical Humana/citologia , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Técnicas Imunoenzimáticas , Macrófagos/citologia , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Melanoma/tratamento farmacológico , Melanoma/metabolismo , Neovascularização Patológica , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Receptores Adrenérgicos beta 3/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais , Neoplasias Cutâneas/tratamento farmacológico , Neoplasias Cutâneas/metabolismo , Células Estromais/citologia , Células Estromais/efeitos dos fármacos , Células Estromais/metabolismo , Fator A de Crescimento do Endotélio Vascular , Melanoma Maligno Cutâneo
19.
Mol Oncol ; 8(8): 1729-46, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25091736

RESUMO

We focused our interest on senescent human-derived fibroblasts in the progression of prostate cancer. Hypoxic senescent fibroblasts promote prostate cancer aggressiveness by inducing epithelial to mesenchymal transition (EMT) and by secreting energy-rich compounds to support cancer cell growth. Hypoxic senescent fibroblasts additionally increase: i) the recruitment of monocytes and their M2-macrophage polarization, ii) the recruitment of bone marrow-derived endothelial precursor cells, facilitating their vasculogenic ability and iii) capillary morphogenesis, proliferation and invasion of human mature endothelial cells. In addition, we highlight that overexpression of the hypoxia-induced miR-210 in young fibroblasts increases their senescence-associated features and converts them into cancer associated fibroblast (CAF)-like cells, able to promote cancer cells EMT, to support angiogenesis and to recruit endothelial precursor cells and monocytes/macrophages.


Assuntos
MicroRNAs/metabolismo , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/patologia , Linhagem Celular , Linhagem Celular Tumoral , Células Cultivadas , Progressão da Doença , Humanos , Imuno-Histoquímica , Masculino , MicroRNAs/genética , Neoplasias da Próstata/genética
20.
Beilstein J Org Chem ; 10: 1317-1324, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24991284

RESUMO

Vaccination strategies based on dendritic cells (DCs) armed with specific tumor antigens have been widely exploited due the properties of these immune cells in coordinating an innate and adaptive response. Here, we describe the convergent synthesis of the bifunctional multivalent glycodendron 5, which contains nine residues of mannose for DC targeting and one residue of an immunogenic mimetic of a carbohydrate melanoma associated antigen. The immunological assays demonstrated that the glycodendron 5 is able to induce human immature DC activation in terms of a phenotype expression of co-stimulatory molecules expression and MHCII. Furthermore, DCs activated by the glycodendron 5 stimulate T lymphocytes to proliferate in a mixed lymphocytes reaction (MLR).

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA