Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 132
Filtrar
1.
Proc Natl Acad Sci U S A ; 121(13): e2306763121, 2024 Mar 26.
Artigo em Inglês | MEDLINE | ID: mdl-38498711

RESUMO

Lactate-proton symporter monocarboxylate transporter 1 (MCT1) facilitates lactic acid export from T cells. Here, we report that MCT1 is mandatory for the development of virus-specific CD8+ T cell memory. MCT1-deficient T cells were exposed to acute pneumovirus (pneumonia virus of mice, PVM) or persistent γ-herpesvirus (Murid herpesvirus 4, MuHV-4) infection. MCT1 was required for the expansion of virus-specific CD8+ T cells and the control of virus replication in the acute phase of infection. This situation prevented the subsequent development of virus-specific T cell memory, a necessary step in containing virus reactivation during γ-herpesvirus latency. Instead, persistent active infection drove virus-specific CD8+ T cells toward functional exhaustion, a phenotype typically seen in chronic viral infections. Mechanistically, MCT1 deficiency sequentially impaired lactic acid efflux from activated CD8+ T cells, caused an intracellular acidification inhibiting glycolysis, disrupted nucleotide synthesis in the upstream pentose phosphate pathway, and halted cell proliferation which, ultimately, promoted functional CD8+ T cell exhaustion instead of memory development. Taken together, our data demonstrate that MCT1 expression is mandatory for inducing T cell memory and controlling viral infection by CD8+ T cells.


Assuntos
Linfócitos T CD8-Positivos , Transportadores de Ácidos Monocarboxílicos , Simportadores , Animais , Camundongos , Transporte Biológico , Linfócitos T CD8-Positivos/metabolismo , Ácido Láctico/metabolismo , Simportadores/genética , Simportadores/metabolismo , Transportadores de Ácidos Monocarboxílicos/genética , Transportadores de Ácidos Monocarboxílicos/metabolismo
2.
Semin Cell Dev Biol ; 98: 181-191, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31112797

RESUMO

Anthracyclines Doxorubicin, Epirubicin, Daunorubicin and Idarubicin are used to treat a variety of tumor types in the clinics, either alone or, most often, in combination therapies. While their cardiotoxicity is well known, the emergence of chemoresistance is also a major issue accounting for treatment discontinuation. Resistance to anthracyclines is associated to the acquisition of multidrug resistance conferred by overexpression of permeability glycoprotein-1 or other efflux pumps, by altered DNA repair, changes in topoisomerase II activity, cancer stemness and metabolic adaptations. This review further details the metabolic aspects of resistance to anthracyclines, emphasizing the contributions of glycolysis, the pentose phosphate pathway and nucleotide biosynthesis, glutathione, lipid metabolism and autophagy to the chemoresistant phenotype.


Assuntos
Antraciclinas/farmacologia , Antibióticos Antineoplásicos/farmacologia , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Neoplasias/tratamento farmacológico , Antraciclinas/química , Antraciclinas/metabolismo , Antibióticos Antineoplásicos/química , Antibióticos Antineoplásicos/metabolismo , Humanos , Estrutura Molecular , Neoplasias/metabolismo , Neoplasias/patologia
3.
J Biol Chem ; 296: 100422, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33607109

RESUMO

Despite being initially regarded as a metabolic waste product, lactate is now considered to serve as a primary fuel for the tricarboxylic acid cycle in cancer cells. At the core of lactate metabolism, lactate dehydrogenases (LDHs) catalyze the interconversion of lactate to pyruvate and as such represent promising targets in cancer therapy. However, direct inhibition of the LDH active site is challenging from physicochemical and selectivity standpoints. However, LDHs are obligate tetramers. Thus, targeting the LDH tetrameric interface has emerged as an appealing strategy. In this work, we examine a dimeric construct of truncated human LDH to search for new druggable sites. We report the identification and characterization of a new cluster of interactions in the LDH tetrameric interface. Using nanoscale differential scanning fluorimetry, chemical denaturation, and mass photometry, we identified several residues (E62, D65, L71, and F72) essential for LDH tetrameric stability. Moreover, we report a family of peptide ligands based on this cluster of interactions. We next demonstrated these ligands to destabilize tetrameric LDHs through binding to this new tetrameric interface using nanoscale differential scanning fluorimetry, NMR water-ligand observed via gradient spectroscopy, and microscale thermophoresis. Altogether, this work provides new insights on the LDH tetrameric interface as well as valuable pharmacological tools for the development of LDH tetramer disruptors.


Assuntos
Mapeamento de Epitopos/métodos , L-Lactato Desidrogenase/metabolismo , Humanos , L-Lactato Desidrogenase/genética , L-Lactato Desidrogenase/fisiologia , Lactato Desidrogenases/metabolismo , Ácido Láctico/metabolismo , Ligantes , Imageamento por Ressonância Magnética/métodos , Peptídeos/metabolismo
4.
Am J Pathol ; 191(9): 1610-1623, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34111431

RESUMO

Despite occasional reports of vertical transmission of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) during pregnancy, the question of placental infection and its consequences for the newborn remain unanswered. Herein, we analyzed the placentas of 31 coronavirus disease 2019-positive mothers by reverse transcriptase PCR, immunohistochemistry, and in situ hybridization. Only one case of placental infection was detected, which was associated with intrauterine demise of the fetus. Differentiated primary trophoblasts were then isolated from nonpathologic human placentas at term, differentiated, and exposed to SARS-CoV-2 virions. Unlike for positive control cells Vero E6, the virus inside cytotrophoblasts and syncytiotrophoblasts or in the supernatant 4 days after infection was undetectable. As a mechanism of defense, we hypothesized that trophoblasts at term do not express angiotensin-converting enzyme 2 and transmembrane protease serine 2 (TMPRSS2), the two main host membrane receptors for SARS-CoV-2 entry. The quantification of these proteins in the placenta during pregnancy confirmed the absence of TMPRSS2 at the surface of the syncytium. Surprisingly, a transiently induced experimental expression of TMPRSS2 did not allow the entry or replication of the virus in differentiated trophoblasts. Altogether, these results underline that trophoblasts are not likely to be infected by SARS-CoV-2 at term, but raise concern about preterm infection.


Assuntos
Enzima de Conversão de Angiotensina 2/biossíntese , COVID-19 , Regulação Enzimológica da Expressão Gênica , Doenças Placentárias , Complicações Infecciosas na Gravidez , SARS-CoV-2/metabolismo , Serina Endopeptidases/biossíntese , Trofoblastos , Internalização do Vírus , Adulto , COVID-19/enzimologia , COVID-19/patologia , Feminino , Humanos , Doenças Placentárias/enzimologia , Doenças Placentárias/patologia , Gravidez , Complicações Infecciosas na Gravidez/enzimologia , Complicações Infecciosas na Gravidez/patologia , Trofoblastos/enzimologia , Trofoblastos/patologia
5.
Molecules ; 27(18)2022 Sep 10.
Artigo em Inglês | MEDLINE | ID: mdl-36144606

RESUMO

BACKGROUND: Mito-metformin10 (MM10), synthesized by attaching a triphenylphosphonium cationic moiety via a 10-carbon aliphatic side chain to metformin, is a mitochondria-targeted analog of metformin that was recently demonstrated to alter mitochondrial function and proliferation in pancreatic ductal adenocarcinoma. Here, we hypothesized that this compound may decrease the oxygen consumption rate (OCR) in prostate cancer cells, increase the level of mitochondrial ROS, alleviate tumor hypoxia, and radiosensitize tumors. METHODS: OCR and mitochondrial superoxide production were assessed by EPR (9 GHz) in vitro in PC-3 and DU-145 prostate cancer cells. Reduced and oxidized glutathione were assessed before and after MM10 exposure. Tumor oxygenation was measured in vivo using 1 GHz EPR oximetry in PC-3 tumor model. Tumors were irradiated at the time of maximal reoxygenation. RESULTS: 24-hours exposure to MM10 significantly decreased the OCR of PC-3 and DU-145 cancer cells. An increase in mitochondrial superoxide levels was observed in PC-3 but not in DU-145 cancer cells, an observation consistent with the differences observed in glutathione levels in both cancer cell lines. In vivo, the tumor oxygenation significantly increased in the PC-3 model (daily injection of 2 mg/kg MM10) 48 and 72 h after initiation of the treatment. Despite the significant effect on tumor hypoxia, MM10 combined to irradiation did not increase the tumor growth delay compared to the irradiation alone. CONCLUSIONS: MM10 altered the OCR in prostate cancer cells. The effect of MM10 on the superoxide level was dependent on the antioxidant capacity of cell line. In vivo, MM10 alleviated tumor hypoxia, yet without consequence in terms of response to irradiation.


Assuntos
Metformina , Neoplasias Pancreáticas , Neoplasias da Próstata , Antioxidantes/farmacologia , Carbono/metabolismo , Linhagem Celular Tumoral , Dissulfeto de Glutationa/metabolismo , Humanos , Masculino , Metformina/farmacologia , Mitocôndrias/metabolismo , Neoplasias Pancreáticas/patologia , Neoplasias da Próstata/patologia , Espécies Reativas de Oxigênio/metabolismo , Superóxidos/metabolismo
6.
FASEB J ; 34(11): 15222-15235, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32954526

RESUMO

Insufficient remodeling of uterine arteries causes pregnancy-related diseases, including fetal growth restriction and preeclampsia. In these situations, reduced maternal blood flow in the placenta is thought to be responsible for the persistence of a low oxygen environment throughout pregnancy. We hypothesized that chronic activation of transcription factors hypoxia-inducible factors (HIFs) actively participates in placental underdevelopment, which impairs fetal growth. The computer-assisted analysis in pathological placentas revealed an increased number of HIF-2α-positive nuclei in the syncytium compared to normal human placentas, while HIF-1α stabilization was unchanged. Specific involvement of HIF-2α was confirmed in primary human cytotrophoblasts rendered deficient for HIF1A or HIF2A. Silencing HIF2A increased the expression of main syncytialization markers as well as differentiation and syncytium formation. It also improved placental growth factor bioavailability. None of these changes was seen when silencing HIF1A. Conversely, the experimental induction of HIF-2α expression repressed forskolin-induced differentiation in BeWo choriocarcinoma cells. Our mechanistic insights evidence that transcription factor HIF-2α impairs placental function, thus suggesting its participation in fetal growth restriction and preeclampsia when placentas become chronically hypoxic. Furthermore, it suggests the possibility to develop novel molecular targeting therapies for placental dysfunction.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Retardo do Crescimento Fetal/patologia , Hipóxia/fisiopatologia , Placenta/patologia , Pré-Eclâmpsia/fisiopatologia , Adulto , Estudos de Casos e Controles , Feminino , Retardo do Crescimento Fetal/etiologia , Retardo do Crescimento Fetal/metabolismo , Humanos , Placenta/metabolismo , Gravidez , Estudos Retrospectivos
7.
Int J Mol Sci ; 22(6)2021 Mar 23.
Artigo em Inglês | MEDLINE | ID: mdl-33806730

RESUMO

Depending on their tissue of origin, genetic and epigenetic marks and microenvironmental influences, cancer cells cover a broad range of metabolic activities that fluctuate over time and space. At the core of most metabolic pathways, mitochondria are essential organelles that participate in energy and biomass production, act as metabolic sensors, control cancer cell death, and initiate signaling pathways related to cancer cell migration, invasion, metastasis and resistance to treatments. While some mitochondrial modifications provide aggressive advantages to cancer cells, others are detrimental. This comprehensive review summarizes the current knowledge about mitochondrial transfers that can occur between cancer and nonmalignant cells. Among different mechanisms comprising gap junctions and cell-cell fusion, tunneling nanotubes are increasingly recognized as a main intercellular platform for unidirectional and bidirectional mitochondrial exchanges. Understanding their structure and functionality is an important task expected to generate new anticancer approaches aimed at interfering with gains of functions (e.g., cancer cell proliferation, migration, invasion, metastasis and chemoresistance) or damaged mitochondria elimination associated with mitochondrial transfer.


Assuntos
Mitocôndrias/genética , Mitocôndrias/metabolismo , Neoplasias/etiologia , Neoplasias/metabolismo , Animais , Transporte Biológico , Proliferação de Células , Sobrevivência Celular , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/metabolismo , Ciclo do Ácido Cítrico , Progressão da Doença , Resistencia a Medicamentos Antineoplásicos , Metabolismo Energético , Humanos , Redes e Vias Metabólicas , Microtúbulos/metabolismo , Neoplasias/patologia , Neoplasias/terapia , Fosforilação Oxidativa , Espécies Reativas de Oxigênio/metabolismo , Microambiente Tumoral
8.
Int J Mol Sci ; 22(21)2021 Nov 03.
Artigo em Inglês | MEDLINE | ID: mdl-34769368

RESUMO

Glioblastoma represents the highest grade of brain tumors. Despite maximal resection surgery associated with radiotherapy and concomitant followed by adjuvant chemotherapy with temozolomide (TMZ), patients have a very poor prognosis due to the rapid recurrence and the acquisition of resistance to TMZ. Here, initially considering that TMZ is a prodrug whose activation is pH-dependent, we explored the contribution of glioblastoma cell metabolism to TMZ resistance. Using isogenic TMZ-sensitive and TMZ-resistant human glioblastoma cells, we report that the expression of O6-methylguanine DNA methyltransferase (MGMT), which is known to repair TMZ-induced DNA methylation, does not primarily account for TMZ resistance. Rather, fitter mitochondria in TMZ-resistant glioblastoma cells are a direct cause of chemoresistance that can be targeted by inhibiting oxidative phosphorylation and/or autophagy/mitophagy. Unexpectedly, we found that PARP inhibitor olaparib, but not talazoparib, is also a mitochondrial Complex I inhibitor. Hence, we propose that the anticancer activities of olaparib in glioblastoma and other cancer types combine DNA repair inhibition and impairment of cancer cell respiration.


Assuntos
Neoplasias Encefálicas/tratamento farmacológico , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Glioblastoma/tratamento farmacológico , Ftalazinas/farmacologia , Piperazinas/farmacologia , Temozolomida/farmacologia , Antineoplásicos Alquilantes/farmacologia , Apoptose , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patologia , Proliferação de Células , Glioblastoma/metabolismo , Glioblastoma/patologia , Humanos , Inibidores de Poli(ADP-Ribose) Polimerases/farmacologia , Células Tumorais Cultivadas
9.
Cancer Metastasis Rev ; 38(1-2): 189-203, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30820778

RESUMO

In cancer, mitochondrial functions are commonly altered. Directly involved in metabolic reprogramming, mitochondrial plasticity confers to cancer cells a high degree of adaptability to a wide range of stresses and to the harsh tumor microenvironment. Lack of nutrients or oxygen caused by altered perfusion, metabolic needs of proliferating cells, co-option of the microenvironment, control of the immune system, cell migration and metastasis, and evasion of exogenous stress (e.g., chemotherapy) are all, at least in part, influenced by mitochondria. Mitochondria are undoubtedly one of the key contributors to cancer development and progression. Understanding their protumoral (dys)functions may pave the way to therapeutic strategies capable of turning them into innocent entities. Here, we will focus on the production and detoxification of mitochondrial reactive oxygen species (mtROS), on their impact on tumorigenesis (genetic, prosurvival, and microenvironmental effects and their involvement in autophagy), and on tumor metastasis. We will also summarize the latest therapeutic approaches involving mtROS.


Assuntos
Mitocôndrias/metabolismo , Neoplasias/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Animais , Antioxidantes/metabolismo , Carcinogênese/metabolismo , Carcinogênese/patologia , Humanos , Mitocôndrias/patologia , Neoplasias/patologia , Fosforilação Oxidativa
10.
Cell Mol Life Sci ; 76(4): 699-728, 2019 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-30377700

RESUMO

The endothelium, a tissue that forms a single layer of cells lining various organs and cavities of the body, especially the heart and blood as well as lymphatic vessels, plays a complex role in vascular biology. It contributes to key aspects of vascular homeostasis and is also involved in pathophysiological processes, such as thrombosis, inflammation, and hypertension. Epidemiological data show that high doses of ionizing radiation lead to cardiovascular disease over time. The aim of this review is to summarize the current knowledge on endothelial cell activation and dysfunction after ionizing radiation exposure as a central feature preceding the development of cardiovascular diseases.


Assuntos
Células Endoteliais/efeitos da radiação , Endotélio Vascular/efeitos da radiação , Endotélio/efeitos da radiação , Lesões por Radiação/fisiopatologia , Radiação Ionizante , Animais , Doenças Cardiovasculares/patologia , Doenças Cardiovasculares/fisiopatologia , Senescência Celular/efeitos da radiação , Células Endoteliais/patologia , Endotélio/patologia , Endotélio/fisiopatologia , Endotélio Vascular/patologia , Endotélio Vascular/fisiopatologia , Humanos , Modelos Biológicos
11.
J Assist Reprod Genet ; 37(10): 2477-2486, 2020 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-32885380

RESUMO

PURPOSE: Although ovarian tissue transportation has been validated for up to 24 h, there is no standard protocol to date. We aimed to elucidate how existing media currently used for ovarian tissue transportation affect ovarian tissue metabolism and cell viability. METHODS: Cow ovarian fragments were immersed in 0.9% NaCl solution, IVF medium, Leibovitz 15 medium (L-15), or PBS for 1, 4, or 24 h at 4 °C. Media were analyzed for pH, lactate dehydrogenase (LDH) activity, and glucose, pyruvate, and lactate concentrations, while apoptosis was assessed by TUNEL assays in fixed fragments. Viability rates were assessed by flow cytometry (FACS). RESULTS: There were lower pH levels in NaCl at all time points compared with other media. LDH activity increased with time and was lowest in NaCl at 1 and 4 h. There was no significant difference in glucose levels, but a significant pyruvate decrease in L-15 and a significant lactate increase in all media. TUNEL showed apoptosis rates ranging from 0 to 5%. FACS showed a mean of 4% necrotic cells and 15-19% apoptotic cells after 1 h of incubation, but less than 1% necrotic cells and 2-6% apoptotic cells after 24 h in all media. CONCLUSION: Our results indicate marked metabolic activity in ovarian tissue at 4 °C and suggest that cells use internal sources of energy, which may influence transplantation outcomes. This highlights the importance of better understanding whole tissue dynamics to develop a standard protocol for ovarian tissue transportation. Graphical abstract.


Assuntos
Necrose/metabolismo , Folículo Ovariano/crescimento & desenvolvimento , Ovário/crescimento & desenvolvimento , Preservação de Tecido , Apoptose/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Criopreservação , Meios de Cultura/química , Meios de Cultura/farmacologia , Feminino , Glucose/metabolismo , Humanos , Necrose/patologia , Necrose/prevenção & controle , Folículo Ovariano/metabolismo , Ovário/metabolismo , Temperatura
12.
Am J Physiol Cell Physiol ; 315(4): C544-C557, 2018 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-30020826

RESUMO

Iodine deficiency (ID), which affects almost two billion people worldwide, is associated with breast pathologies such as fibrosis in human and induces breast atypia in animal models. Because ID induces vascular activation in the thyroid, another iodide-uptaking organ, and as breast is also sensitive to ID, we aimed to characterize ID-induced effects on the breast microvasculature in vivo and in two different breast cell lines in vitro. Virgin and lactating NMRI mice received an iodide-deficient diet and a Na+/I- symporter inhibitor for 1 to 20 days. Some virgin mice were treated with vascular endothelial growth factor A (VEGF) or VEGF receptor inhibitors. In vitro, ID was induced in MCF7 and MCF12A cells by replacing the iodide-containing medium by an iodide-deficient medium. In vivo, VEGF expression was increased following ID in mammary tissues. Consequently, ID induced a transient increase in mammary gland blood flow, measured after anesthesia, in virgin and lactating mice, which was repressed by VEGF or VEGF receptor inhibitors. In MCF7 cells, ID induced a transient increase in reactive oxygen species, followed by an increase in hypoxia-inducible factor-1α (HIF-1α) protein and VEGF mRNA expression. Antioxidant N-acetylcysteine and mammalian target of rapamycin (mTOR) inhibitor blocked ID-induced HIF-1α protein increase and VEGF transcription. However, mTOR activity was not inhibited by N-acetylcysteine. Similar responses were observed in MCF12A cells. These data indicate that ID activates the canonical VEGF pathway and mTOR in breast tissues, which provides new insights to better understand the correlation between ID, vascular activation, and breast pathologies.


Assuntos
Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Iodo/deficiência , Glândulas Mamárias Humanas/metabolismo , Microvasos/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Acetilcisteína/metabolismo , Animais , Antioxidantes/metabolismo , Mama/metabolismo , Linhagem Celular Tumoral , Feminino , Humanos , Lactação/metabolismo , Células MCF-7 , Glândulas Mamárias Animais/metabolismo , Camundongos , Receptores de Fatores de Crescimento do Endotélio Vascular/metabolismo , Transdução de Sinais/fisiologia
13.
Artigo em Inglês | MEDLINE | ID: mdl-29263063

RESUMO

Linezolid, the first clinically available oxazolidinone antibiotic, causes potentially severe toxicities (myelosuppression, lactic acidosis, and neuropathies) ascribed to impairment of mitochondrial protein synthesis and consecutive mitochondrial dysfunction. Tedizolid, a newly approved oxazolidinone, shows an enhanced activity compared to linezolid but is also a more potent inhibitor of mitochondrial protein synthesis. We compared linezolid and tedizolid for (i) inhibition of the expression of subunit I of cytochrome c-oxidase (CYTox I; Western blot analysis), (ii) cytochrome c-oxidase activity (biochemical assay), (iii) mitochondrial oxidative metabolism (Seahorse technology), and (iv) alteration of mitochondrial ultrastructure (electron microscopy) using HL-60 promyelocytes and THP-1 monocytes exposed to microbiologically (multiples of modal MIC against Staphylococcus aureus) and therapeutically (Cmin - Cmax) pertinent concentrations. Both drugs caused a rapid and complete (48 to 72 h) inhibition of CYTox I expression, cytochrome c-oxidase activity, and spare respiratory capacity, with conspicuous swelling of the mitochondrial matrix and loss of their cristae. Globally, tedizolid was a more potent inhibitor than linezolid. For both drugs, all effects were quickly (48 to 72 h) and fully reversible upon drug withdrawal. Using an alternation of exposure to and withdrawal from drug mimicking their approved schedule of administration (twice daily and once daily [qD] for linezolid and tedizolid, respectively), only partial inhibition of CYTox I expression was noted for up to 96 h. Thus, rapid reversal of toxic effects upon discontinuous administration may mitigate oxazolidinone toxicity. Since tedizolid is given qD, this may help to explain its reported lower preclinical and clinical toxicity.


Assuntos
Linezolida/efeitos adversos , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Proteínas Mitocondriais/metabolismo , Oxazolidinonas/efeitos adversos , Tetrazóis/efeitos adversos , Complexo de Proteínas da Cadeia de Transporte de Elétrons/metabolismo , Células HL-60 , Humanos , Testes de Sensibilidade Microbiana , Staphylococcus aureus/efeitos dos fármacos , Células THP-1
14.
Biochim Biophys Acta Bioenerg ; 1858(8): 556-572, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28167100

RESUMO

Altered metabolism in cancer cells is pivotal for tumor growth, most notably by providing energy, reducing equivalents and building blocks while several metabolites exert a signaling function promoting tumor growth and progression. A cancer tissue cannot be simply reduced to a bulk of proliferating cells. Tumors are indeed complex and dynamic structures where single cells can heterogeneously perform various biological activities with different metabolic requirements. Because tumors are composed of different types of cells with metabolic activities affected by different spatial and temporal contexts, it is important to address metabolism taking into account cellular and biological heterogeneity. In this review, we describe this heterogeneity also in metabolic fluxes, thus showing the relative contribution of different metabolic activities to tumor progression according to the cellular context. This article is part of a Special Issue entitled Mitochondria in Cancer, edited by Giuseppe Gasparre, Rodrigue Rossignol and Pierre Sonveaux.


Assuntos
Metabolismo Energético , Neoplasias/metabolismo , Animais , Morte Celular , Divisão Celular , Glicólise , Humanos , Redes e Vias Metabólicas , Mitocôndrias/metabolismo , Metástase Neoplásica , Células-Tronco Neoplásicas/metabolismo , Células Estromais/metabolismo
15.
Biochim Biophys Acta ; 1863(10): 2481-97, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-26993058

RESUMO

Monocarboxylate transporters (MCTs) constitute a family of 14 members among which MCT1-4 facilitate the passive transport of monocarboxylates such as lactate, pyruvate and ketone bodies together with protons across cell membranes. Their anchorage and activity at the plasma membrane requires interaction with chaperon protein such as basigin/CD147 and embigin/gp70. MCT1-4 are expressed in different tissues where they play important roles in physiological and pathological processes. This review focuses on the brain and on cancer. In the brain, MCTs control the delivery of lactate, produced by astrocytes, to neurons, where it is used as an oxidative fuel. Consequently, MCT dysfunctions are associated with pathologies of the central nervous system encompassing neurodegeneration and cognitive defects, epilepsy and metabolic disorders. In tumors, MCTs control the exchange of lactate and other monocarboxylates between glycolytic and oxidative cancer cells, between stromal and cancer cells and between glycolytic cells and endothelial cells. Lactate is not only a metabolic waste for glycolytic cells and a metabolic fuel for oxidative cells, but it also behaves as a signaling agent that promotes angiogenesis and as an immunosuppressive metabolite. Because MCTs gate the activities of lactate, drugs targeting these transporters have been developed that could constitute new anticancer treatments. This article is part of a Special Issue entitled: Mitochondrial Channels edited by Pierre Sonveaux, Pierre Maechler and Jean-Claude Martinou.


Assuntos
Encéfalo/metabolismo , Transportadores de Ácidos Monocarboxílicos/metabolismo , Neoplasias/metabolismo , Animais , Astrócitos/metabolismo , Transporte Biológico Ativo , Encefalopatias/metabolismo , Cognição/fisiologia , Regulação da Expressão Gênica , Glicólise , Humanos , Concentração de Íons de Hidrogênio , Corpos Cetônicos/metabolismo , Lactatos/metabolismo , Linfócitos/metabolismo , Camundongos , Transportadores de Ácidos Monocarboxílicos/antagonistas & inibidores , Transportadores de Ácidos Monocarboxílicos/genética , Proteínas de Neoplasias/antagonistas & inibidores , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Neurônios/metabolismo , Especificidade de Órgãos , Fosforilação Oxidativa , Ácido Pirúvico/metabolismo , Ratos
16.
Br J Cancer ; 117(9): 1336-1340, 2017 Oct 24.
Artigo em Inglês | MEDLINE | ID: mdl-28873082

RESUMO

BACKGROUND: Activation of free fatty acid receptor 2 (FFAR2) by microbiota-derived metabolites (e.g., propionate) reduces leukaemic cell proliferation in vitro. This study aims to test whether Ffar2 expression per se also influences leukaemia cell growth in vivo. METHODS: Bcr-Abl-expressing BaF cells were used as a leukaemia model and the role of Ffar2 was evaluated in Balb/c mice after lentiviral shRNA transduction. RESULTS: Our data formally establish that reduced leukaemic cell proliferation is associated with increased Ffar2 expression in vivo and in vitro. Going beyond association, we point out that decreasing Ffar2 expression fosters cancer cell growth in vitro and in vivo. CONCLUSIONS: Our data demonstrate the role of Ffar2 in the control of leukaemic cell proliferation in vivo and indicate that a modulation of Ffar2 expression through nutritional tools or pharmacological agents may constitute an attractive therapeutic approach to tackle leukaemia progression in humans.


Assuntos
Proliferação de Células , Leucemia Experimental/patologia , Receptores Acoplados a Proteínas G/fisiologia , Animais , Apoptose , Biomarcadores Tumorais/metabolismo , Feminino , Leucemia Experimental/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Células Tumorais Cultivadas
17.
Biochim Biophys Acta Mol Basis Dis ; 1863(12): 3254-3264, 2017 12.
Artigo em Inglês | MEDLINE | ID: mdl-28866133

RESUMO

Recent progress in dissecting the molecular paracrine circuits of cancer and stromal cells in bone metastases (BM) are offering new options to improve current merely palliative approach. The study of tumor-stroma metabolic interplay may further ameliorate this scenario. In this context, we demonstrated that highly glycolytic MDA-MB-231 cancer cells, that form osteolytic BM in vivo, release a large amount of lactate at a significantly higher level than MCF7 cells. Thus, we speculated that lactate released from carcinoma cells is uptaken and metabolically used by osteoclasts, the key players of osteolysis associated with BM. First, we demonstrated that the release of lactate at the bone site is mediated by monocarboxylate transporter 4 (MCT4), as revealed by immunostaining and MCT4 localization at the plasma membrane of tumor cells in mouse model of BM and in human tissue sections of BM. Then, we showed that in vitro lactate is uptaken by osteoclasts to be used as a fuel for the oxidative metabolism of osteoclasts, ultimately enhancing Type I collagen resorption. The passive transport of lactate into osteoclasts was mediated by MCT1: MCT1 expression is significantly upregulated during osteoclast differentiation and Type I collagen resorption is significantly impaired when osteoclasts are treated with 7-(N-benzyl-N-methylamino)-2-oxo-2H-chromene-3-carboxylic acid, an MCT-1 inhibitor. Together, these data demonstrate that lactate released by glycolytic breast carcinoma cells in the bone microenvironment promotes the formation of osteolytic lesions, and provide the rationale for further studies on the use of MCT1 targeting as a novel therapeutic approach in advanced cancer patients with BM.


Assuntos
Neoplasias Ósseas/metabolismo , Neoplasias Ósseas/secundário , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Osteoclastos/metabolismo , Animais , Linhagem Celular Tumoral/metabolismo , Cumarínicos/antagonistas & inibidores , Feminino , Glucose/metabolismo , Glicólise , Humanos , Lactatos/metabolismo , Células MCF-7 , Camundongos , Transportadores de Ácidos Monocarboxílicos/metabolismo , Proteínas Musculares/metabolismo , Osteoclastos/patologia , Células Estromais/metabolismo , Células Estromais/patologia , Simportadores/metabolismo
18.
Cell Mol Life Sci ; 73(7): 1333-48, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26626411

RESUMO

Metabolic adaptations are intimately associated with changes in cell behavior. Cancers are characterized by a high metabolic plasticity resulting from mutations and the selection of metabolic phenotypes conferring growth and invasive advantages. While metabolic plasticity allows cancer cells to cope with various microenvironmental situations that can be encountered in a primary tumor, there is increasing evidence that metabolism is also a major driver of cancer metastasis. Rather than a general switch promoting metastasis as a whole, a succession of metabolic adaptations is more likely needed to promote different steps of the metastatic process. This review addresses the contribution of pH, glycolysis and the pentose phosphate pathway, and a companion paper summarizes current knowledge regarding the contribution of mitochondria, lipids and amino acid metabolism. Extracellular acidification, intracellular alkalinization, the glycolytic enzyme phosphoglucose isomerase acting as an autocrine cytokine, lactate and the pentose phosphate pathway are emerging as important factors controlling cancer metastasis.


Assuntos
Neoplasias/patologia , Via de Pentose Fosfato/fisiologia , Transição Epitelial-Mesenquimal , Glucose-6-Fosfato Isomerase/metabolismo , Glicólise , Humanos , Ácido Láctico/metabolismo , Metástase Neoplásica , Neoplasias/metabolismo
19.
Cell Mol Life Sci ; 73(7): 1349-63, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26646069

RESUMO

Metabolic alterations are a hallmark of cancer controlling tumor progression and metastasis. Among the various metabolic phenotypes encountered in tumors, this review focuses on the contributions of mitochondria, lipid and amino acid metabolism to the metastatic process. Tumor cells require functional mitochondria to grow, proliferate and metastasize, but shifts in mitochondrial activities confer pro-metastatic traits encompassing increased production of mitochondrial reactive oxygen species (mtROS), enhanced resistance to apoptosis and the increased or de novo production of metabolic intermediates of the TCA cycle behaving as oncometabolites, including succinate, fumarate, and D-2-hydroxyglutarate that control energy production, biosynthesis and the redox state. Lipid metabolism and the metabolism of amino acids, such as glutamine, glutamate and proline are also currently emerging as focal control points of cancer metastasis.


Assuntos
Aminoácidos/metabolismo , Metabolismo dos Lipídeos/fisiologia , Mitocôndrias/metabolismo , Neoplasias/patologia , Ciclo do Ácido Cítrico , Humanos , Metástase Neoplásica , Neoplasias/metabolismo , Fosforilação Oxidativa , Espécies Reativas de Oxigênio/metabolismo
20.
Biochim Biophys Acta ; 1848(10 Pt B): 2715-26, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25449995

RESUMO

Although cancer is characterized by an intratumoral genetic heterogeneity, a totally deranged pH control is a common feature of most cancer histotypes. Major determinants of aberrant pH gradient in cancer are proton exchangers and transporters, including V-ATPase, Na+/H+ exchanger (NHE), monocarboxylate transporters (MCTs) and carbonic anhydrases (CAs). Thanks to the activity of these proton transporters and exchangers, cancer becomes isolated and/or protected not only from the body reaction against the growing tumor, but also from the vast majority of drugs that when protonated into the acidic tumor microenvironment do not enter into cancer cells. Proton transporters and exchangers represent a key feature tumor cells use to survive in the very hostile microenvironmental conditions that they create and maintain. Detoxifying mechanisms may thus represent both a key survival option and a selection outcome for cells that behave as unicellular microorganisms rather than belonging to an organ, compartment or body. It is, in fact, typical of malignant tumors that, after a clinically measurable yet transient initial response to a therapy, resistant tumor clones emerge and proliferate, thus bursting a more malignant behavior and rapid tumor progression. This review critically presents the background of a novel and efficient approach that aims to fight cancer through blocking or inhibiting well characterized proton exchangers and transporters active in human cancer cells. This article is part of a Special Issue entitled: Membrane channels and transporters in cancers.


Assuntos
Antineoplásicos/uso terapêutico , Regulação Neoplásica da Expressão Gênica , Neoplasias/metabolismo , Inibidores da Bomba de Prótons/uso terapêutico , Prótons , Animais , Anidrases Carbônicas/genética , Anidrases Carbônicas/metabolismo , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/genética , Humanos , Concentração de Íons de Hidrogênio , Transportadores de Ácidos Monocarboxílicos/antagonistas & inibidores , Transportadores de Ácidos Monocarboxílicos/genética , Transportadores de Ácidos Monocarboxílicos/metabolismo , Neoplasias/tratamento farmacológico , Neoplasias/genética , Neoplasias/patologia , Trocadores de Sódio-Hidrogênio/antagonistas & inibidores , Trocadores de Sódio-Hidrogênio/genética , Trocadores de Sódio-Hidrogênio/metabolismo , Microambiente Tumoral/efeitos dos fármacos , ATPases Vacuolares Próton-Translocadoras/antagonistas & inibidores , ATPases Vacuolares Próton-Translocadoras/genética , ATPases Vacuolares Próton-Translocadoras/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA