Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 166
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 187(7): 1574-1577, 2024 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-38552606

RESUMEN

Discoveries of the roles of RAS oncogenes in cancer development four decades ago opened the door to proving that tumor development is driven by somatic mutations' altering the genomes of cancer cells. These discoveries led to illusions about the simplicity of cancer pathogenesis and how cancer could be cured.


Asunto(s)
Genes ras , Neoplasias , Humanos , Neoplasias/genética , Neoplasias/terapia , Oncogenes , Mutación
2.
Nat Rev Mol Cell Biol ; 21(6): 341-352, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32300252

RESUMEN

Epithelial-mesenchymal transition (EMT) encompasses dynamic changes in cellular organization from epithelial to mesenchymal phenotypes, which leads to functional changes in cell migration and invasion. EMT occurs in a diverse range of physiological and pathological conditions and is driven by a conserved set of inducing signals, transcriptional regulators and downstream effectors. With over 5,700 publications indexed by Web of Science in 2019 alone, research on EMT is expanding rapidly. This growing interest warrants the need for a consensus among researchers when referring to and undertaking research on EMT. This Consensus Statement, mediated by 'the EMT International Association' (TEMTIA), is the outcome of a 2-year-long discussion among EMT researchers and aims to both clarify the nomenclature and provide definitions and guidelines for EMT research in future publications. We trust that these guidelines will help to reduce misunderstanding and misinterpretation of research data generated in various experimental models and to promote cross-disciplinary collaboration to identify and address key open questions in this research field. While recognizing the importance of maintaining diversity in experimental approaches and conceptual frameworks, we emphasize that lasting contributions of EMT research to increasing our understanding of developmental processes and combatting cancer and other diseases depend on the adoption of a unified terminology to describe EMT.


Asunto(s)
Investigación Biomédica/normas , Transición Epitelial-Mesenquimal , Animales , Movimiento Celular , Plasticidad de la Célula , Consenso , Biología Evolutiva/normas , Humanos , Neoplasias/patología , Terminología como Asunto
3.
Cell ; 168(4): 670-691, 2017 02 09.
Artículo en Inglés | MEDLINE | ID: mdl-28187288

RESUMEN

Metastases account for the great majority of cancer-associated deaths, yet this complex process remains the least understood aspect of cancer biology. As the body of research concerning metastasis continues to grow at a rapid rate, the biological programs that underlie the dissemination and metastatic outgrowth of cancer cells are beginning to come into view. In this review we summarize the cellular and molecular mechanisms involved in metastasis, with a focus on carcinomas where the most is known, and we highlight the general principles of metastasis that have begun to emerge.


Asunto(s)
Carcinoma/patología , Metástasis de la Neoplasia/patología , Animales , Plaquetas/metabolismo , Carcinoma/genética , Carcinoma/metabolismo , Movimiento Celular , Transición Epitelial-Mesenquimal , Humanos , Invasividad Neoplásica , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias/patología , Células Neoplásicas Circulantes/metabolismo , Células Neoplásicas Circulantes/patología , Neutrófilos/metabolismo , Linfocitos T/inmunología , Microambiente Tumoral
4.
Nat Rev Mol Cell Biol ; 20(2): 69-84, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30459476

RESUMEN

Epithelial-mesenchymal transition (EMT) is a cellular programme that is known to be crucial for embryogenesis, wound healing and malignant progression. During EMT, cell-cell and cell-extracellular matrix interactions are remodelled, which leads to the detachment of epithelial cells from each other and the underlying basement membrane, and a new transcriptional programme is activated to promote the mesenchymal fate. In the context of neoplasias, EMT confers on cancer cells increased tumour-initiating and metastatic potential and a greater resistance to elimination by several therapeutic regimens. In this Review, we discuss recent findings on the mechanisms and roles of EMT in normal and neoplastic tissues, and the cell-intrinsic signals that sustain expression of this programme. We also highlight how EMT gives rise to a variety of intermediate cell states between the epithelial and the mesenchymal state, which could function as cancer stem cells. In addition, we describe the contributions of the tumour microenvironment in inducing EMT and the effects of EMT on the immunobiology of carcinomas.


Asunto(s)
Transición Epitelial-Mesenquimal/fisiología , Neoplasias/patología , Animales , Matriz Extracelular/patología , Humanos , Células Madre Neoplásicas/patología , Microambiente Tumoral/fisiología
5.
Cell ; 157(1): 267-71, 2014 Mar 27.
Artículo en Inglés | MEDLINE | ID: mdl-24679541

RESUMEN

Cell has celebrated the powers of reductionist molecular biology and its major successes for four decades. Those who have participated in cancer research during this period have witnessed wild fluctuations from times where endless inexplicable phenomenology reigned supreme to periods of reductionist triumphalism and, in recent years, to a move back to confronting the endless complexity of this disease.


Asunto(s)
Biología Molecular , Neoplasias/metabolismo , Animales , Genómica , Humanos , Neoplasias/genética , Neoplasias/patología , Neoplasias/virología , Oncogenes , Retroviridae/fisiología
6.
Cell ; 158(5): 1094-1109, 2014 Aug 28.
Artículo en Inglés | MEDLINE | ID: mdl-25171410

RESUMEN

It is increasingly appreciated that oncogenic transformation alters cellular metabolism to facilitate cell proliferation, but less is known about the metabolic changes that promote cancer cell aggressiveness. Here, we analyzed metabolic gene expression in cancer cell lines and found that a set of high-grade carcinoma lines expressing mesenchymal markers share a unique 44 gene signature, designated the "mesenchymal metabolic signature" (MMS). A FACS-based shRNA screen identified several MMS genes as essential for the epithelial-mesenchymal transition (EMT), but not for cell proliferation. Dihydropyrimidine dehydrogenase (DPYD), a pyrimidine-degrading enzyme, was highly expressed upon EMT induction and was necessary for cells to acquire mesenchymal characteristics in vitro and for tumorigenic cells to extravasate into the mouse lung. This role of DPYD was mediated through its catalytic activity and enzymatic products, the dihydropyrimidines. Thus, we identify metabolic processes essential for the EMT, a program associated with the acquisition of metastatic and aggressive cancer cell traits.


Asunto(s)
Transición Epitelial-Mesenquimal , Pirimidinas/metabolismo , Animales , Carcinoma/metabolismo , Línea Celular Tumoral , Dihidrouracilo Deshidrogenasa (NADP)/genética , Citometría de Flujo , Perfilación de la Expresión Génica , Humanos , Mesodermo/citología , Mesodermo/metabolismo , Ratones , ARN Interferente Pequeño/metabolismo
8.
Cell ; 154(1): 61-74, 2013 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-23827675

RESUMEN

The recent discovery that normal and neoplastic epithelial cells re-enter the stem cell state raised the intriguing possibility that the aggressiveness of carcinomas derives not from their existing content of cancer stem cells (CSCs) but from their proclivity to generate new CSCs from non-CSC populations. Here, we demonstrate that non-CSCs of human basal breast cancers are plastic cell populations that readily switch from a non-CSC to CSC state. The observed cell plasticity is dependent on ZEB1, a key regulator of the epithelial-mesenchymal transition. We find that plastic non-CSCs maintain the ZEB1 promoter in a bivalent chromatin configuration, enabling them to respond readily to microenvironmental signals, such as TGFß. In response, the ZEB1 promoter converts from a bivalent to active chromatin configuration, ZEB1 transcription increases, and non-CSCs subsequently enter the CSC state. Our findings support a dynamic model in which interconversions between low and high tumorigenic states occur frequently, thereby increasing tumorigenic and malignant potential.


Asunto(s)
Neoplasias de la Mama/patología , Cromatina/metabolismo , Proteínas de Homeodominio/metabolismo , Células Madre Neoplásicas/patología , Regiones Promotoras Genéticas , Factores de Transcripción/metabolismo , Animales , Neoplasias de la Mama/genética , Células Epiteliales/patología , Técnicas de Silenciamiento del Gen , Proteínas de Homeodominio/genética , Humanos , Receptores de Hialuranos/metabolismo , Ratones , Ratones Endogámicos NOD , Ratones SCID , MicroARNs/metabolismo , Células Madre Neoplásicas/metabolismo , Factores de Transcripción/genética , Factor de Crecimiento Transformador beta/metabolismo , Homeobox 1 de Unión a la E-Box con Dedos de Zinc
9.
Cell ; 148(5): 1015-28, 2012 Mar 02.
Artículo en Inglés | MEDLINE | ID: mdl-22385965

RESUMEN

Regulatory networks orchestrated by key transcription factors (TFs) have been proposed to play a central role in the determination of stem cell states. However, the master transcriptional regulators of adult stem cells are poorly understood. We have identified two TFs, Slug and Sox9, that act cooperatively to determine the mammary stem cell (MaSC) state. Inhibition of either Slug or Sox9 blocks MaSC activity in primary mammary epithelial cells. Conversely, transient coexpression of exogenous Slug and Sox9 suffices to convert differentiated luminal cells into MaSCs with long-term mammary gland-reconstituting ability. Slug and Sox9 induce MaSCs by activating distinct autoregulatory gene expression programs. We also show that coexpression of Slug and Sox9 promotes the tumorigenic and metastasis-seeding abilities of human breast cancer cells and is associated with poor patient survival, providing direct evidence that human breast cancer stem cells are controlled by key regulators similar to those operating in normal murine MaSCs.


Asunto(s)
Neoplasias de la Mama/metabolismo , Glándulas Mamarias Humanas/citología , Factor de Transcripción SOX9/metabolismo , Factores de Transcripción/metabolismo , Animales , Células Cultivadas , Femenino , Técnicas de Silenciamiento del Gen , Humanos , Glándulas Mamarias Humanas/metabolismo , Ratones , Factor de Transcripción SOX9/genética , Factores de Transcripción de la Familia Snail , Factores de Transcripción/genética
10.
Cell ; 144(5): 646-74, 2011 Mar 04.
Artículo en Inglés | MEDLINE | ID: mdl-21376230

RESUMEN

The hallmarks of cancer comprise six biological capabilities acquired during the multistep development of human tumors. The hallmarks constitute an organizing principle for rationalizing the complexities of neoplastic disease. They include sustaining proliferative signaling, evading growth suppressors, resisting cell death, enabling replicative immortality, inducing angiogenesis, and activating invasion and metastasis. Underlying these hallmarks are genome instability, which generates the genetic diversity that expedites their acquisition, and inflammation, which fosters multiple hallmark functions. Conceptual progress in the last decade has added two emerging hallmarks of potential generality to this list-reprogramming of energy metabolism and evading immune destruction. In addition to cancer cells, tumors exhibit another dimension of complexity: they contain a repertoire of recruited, ostensibly normal cells that contribute to the acquisition of hallmark traits by creating the "tumor microenvironment." Recognition of the widespread applicability of these concepts will increasingly affect the development of new means to treat human cancer.


Asunto(s)
Neoplasias/patología , Neoplasias/fisiopatología , Animales , Inestabilidad Genómica , Humanos , Invasividad Neoplásica , Neoplasias/metabolismo , Transducción de Señal , Células del Estroma/patología
11.
Cell ; 147(2): 275-92, 2011 Oct 14.
Artículo en Inglés | MEDLINE | ID: mdl-22000009

RESUMEN

Metastases represent the end products of a multistep cell-biological process termed the invasion-metastasis cascade, which involves dissemination of cancer cells to anatomically distant organ sites and their subsequent adaptation to foreign tissue microenvironments. Each of these events is driven by the acquisition of genetic and/or epigenetic alterations within tumor cells and the co-option of nonneoplastic stromal cells, which together endow incipient metastatic cells with traits needed to generate macroscopic metastases. Recent advances provide provocative insights into these cell-biological and molecular changes, which have implications regarding the steps of the invasion-metastasis cascade that appear amenable to therapeutic targeting.


Asunto(s)
Metástasis de la Neoplasia/patología , Neoplasias/patología , Células del Estroma/patología , Animales , Membrana Basal/patología , Humanos , Invasividad Neoplásica/patología , Metástasis de la Neoplasia/tratamiento farmacológico , Neoplasias/tratamiento farmacológico , Neoplasias/genética , Pronóstico , Transducción de Señal
12.
Cell ; 145(6): 926-40, 2011 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-21663795

RESUMEN

The epithelial-mesenchymal transition (EMT) has been associated with the acquisition of motility, invasiveness, and self-renewal traits. During both normal development and tumor pathogenesis, this change in cell phenotype is induced by contextual signals that epithelial cells receive from their microenvironment. The signals that are responsible for inducing an EMT and maintaining the resulting cellular state have been unclear. We describe three signaling pathways, involving transforming growth factor (TGF)-ß and canonical and noncanonical Wnt signaling, that collaborate to induce activation of the EMT program and thereafter function in an autocrine fashion to maintain the resulting mesenchymal state. Downregulation of endogenously synthesized inhibitors of autocrine signals in epithelial cells enables the induction of the EMT program. Conversely, disruption of autocrine signaling by added inhibitors of these pathways inhibits migration and self-renewal in primary mammary epithelial cells and reduces tumorigenicity and metastasis by their transformed derivatives.


Asunto(s)
Comunicación Autocrina , Neoplasias de la Mama/metabolismo , Mama/citología , Células Madre Neoplásicas/metabolismo , Comunicación Paracrina , Células Madre/metabolismo , Animales , Proteínas Morfogenéticas Óseas/metabolismo , Mama/metabolismo , Mama/patología , Neoplasias de la Mama/patología , Movimiento Celular , Células Epiteliales/metabolismo , Transición Epitelial-Mesenquimal , Femenino , Humanos , Glándulas Mamarias Animales/citología , Glándulas Mamarias Animales/efectos de los fármacos , Mesodermo/metabolismo , Ratones , Transducción de Señal , Factor de Crecimiento Transformador beta/metabolismo , Proteínas Wnt/metabolismo
13.
Nature ; 585(7826): 603-608, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32939090

RESUMEN

Ferroptosis-an iron-dependent, non-apoptotic cell death process-is involved in various degenerative diseases and represents a targetable susceptibility in certain cancers1. The ferroptosis-susceptible cell state can either pre-exist in cells that arise from certain lineages or be acquired during cell-state transitions2-5. However, precisely how susceptibility to ferroptosis is dynamically regulated remains poorly understood. Here we use genome-wide CRISPR-Cas9 suppressor screens to identify the oxidative organelles peroxisomes as critical contributors to ferroptosis sensitivity in human renal and ovarian carcinoma cells. Using lipidomic profiling we show that peroxisomes contribute to ferroptosis by synthesizing polyunsaturated ether phospholipids (PUFA-ePLs), which act as substrates for lipid peroxidation that, in turn, results in the induction of ferroptosis. Carcinoma cells that are initially sensitive to ferroptosis can switch to a ferroptosis-resistant state in vivo in mice, which is associated with extensive downregulation of PUFA-ePLs. We further find that the pro-ferroptotic role of PUFA-ePLs can be extended beyond neoplastic cells to other cell types, including neurons and cardiomyocytes. Together, our work reveals roles for the peroxisome-ether-phospholipid axis in driving susceptibility to and evasion from ferroptosis, highlights PUFA-ePL as a distinct functional lipid class that is dynamically regulated during cell-state transitions, and suggests multiple regulatory nodes for therapeutic interventions in diseases that involve ferroptosis.


Asunto(s)
Éteres/metabolismo , Ferroptosis , Peroxisomas/metabolismo , Fosfolípidos/química , Fosfolípidos/metabolismo , Animales , Sistemas CRISPR-Cas/genética , Diferenciación Celular , Línea Celular , Éteres/química , Femenino , Edición Génica , Humanos , Neoplasias Renales/metabolismo , Neoplasias Renales/patología , Peroxidación de Lípido , Masculino , Ratones , Miocitos Cardíacos/citología , Miocitos Cardíacos/metabolismo , Neuronas/citología , Neuronas/metabolismo , Neoplasias Ováricas/metabolismo , Neoplasias Ováricas/patología , Peroxisomas/genética
14.
Cell ; 138(4): 645-659, 2009 Aug 21.
Artículo en Inglés | MEDLINE | ID: mdl-19682730

RESUMEN

Screens for agents that specifically kill epithelial cancer stem cells (CSCs) have not been possible due to the rarity of these cells within tumor cell populations and their relative instability in culture. We describe here an approach to screening for agents with epithelial CSC-specific toxicity. We implemented this method in a chemical screen and discovered compounds showing selective toxicity for breast CSCs. One compound, salinomycin, reduces the proportion of CSCs by >100-fold relative to paclitaxel, a commonly used breast cancer chemotherapeutic drug. Treatment of mice with salinomycin inhibits mammary tumor growth in vivo and induces increased epithelial differentiation of tumor cells. In addition, global gene expression analyses show that salinomycin treatment results in the loss of expression of breast CSC genes previously identified by analyses of breast tissues isolated directly from patients. This study demonstrates the ability to identify agents with specific toxicity for epithelial CSCs.


Asunto(s)
Antineoplásicos/farmacología , Ensayos de Selección de Medicamentos Antitumorales/métodos , Neoplasias Mamarias Experimentales/tratamiento farmacológico , Células Madre Neoplásicas/efectos de los fármacos , Paclitaxel/farmacología , Piranos/farmacología , Animales , Antineoplásicos/uso terapéutico , Apoptosis , Línea Celular Tumoral , Resistencia a Antineoplásicos , Regulación Neoplásica de la Expresión Génica , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos NOD , Ratones SCID , Metástasis de la Neoplasia/tratamiento farmacológico , Trasplante de Neoplasias , Paclitaxel/uso terapéutico , Piranos/uso terapéutico
15.
Cell ; 137(6): 1032-46, 2009 Jun 12.
Artículo en Inglés | MEDLINE | ID: mdl-19524507

RESUMEN

MicroRNAs are well suited to regulate tumor metastasis because of their capacity to coordinately repress numerous target genes, thereby potentially enabling their intervention at multiple steps of the invasion-metastasis cascade. We identify a microRNA exemplifying these attributes, miR-31, whose expression correlates inversely with metastasis in human breast cancer patients. Overexpression of miR-31 in otherwise-aggressive breast tumor cells suppresses metastasis. We deploy a stable microRNA sponge strategy to inhibit miR-31 in vivo; this allows otherwise-nonaggressive breast cancer cells to metastasize. These phenotypes do not involve confounding influences on primary tumor development and are specifically attributable to miR-31-mediated inhibition of several steps of metastasis, including local invasion, extravasation or initial survival at a distant site, and metastatic colonization. Such pleiotropy is achieved via coordinate repression of a cohort of metastasis-promoting genes, including RhoA. Indeed, RhoA re-expression partially reverses miR-31-imposed metastasis suppression. These findings indicate that miR-31 uses multiple mechanisms to oppose metastasis.


Asunto(s)
Neoplasias de la Mama/metabolismo , Regulación Neoplásica de la Expresión Génica , MicroARNs/metabolismo , Metástasis de la Neoplasia , Animales , Línea Celular Tumoral , Proteínas del Citoesqueleto/genética , Receptores Frizzled/genética , Humanos , Integrina alfa5/genética , Proteínas de la Membrana/genética , Receptores Acoplados a Proteínas G/genética , Proteína de Unión al GTP rhoA/genética
16.
Cell ; 133(6): 994-1005, 2008 Jun 13.
Artículo en Inglés | MEDLINE | ID: mdl-18555776

RESUMEN

The effects of primary tumors on the host systemic environment and resulting contributions of the host to tumor growth are poorly understood. Here, we find that human breast carcinomas instigate the growth of otherwise-indolent tumor cells, micrometastases, and human tumor surgical specimens located at distant anatomical sites. This systemic instigation is accompanied by incorporation of bone-marrow cells (BMCs) into the stroma of the distant, once-indolent tumors. We find that BMCs of hosts bearing instigating tumors are functionally activated prior to their mobilization; hence, when coinjected with indolent cells, these activated BMCs mimic the systemic effects imparted by instigating tumors. Secretion of osteopontin by instigating tumors is necessary for BMC activation and the subsequent outgrowth of the distant otherwise-indolent tumors. These results reveal that outgrowth of indolent tumors can be governed on a systemic level by endocrine factors released by certain instigating tumors, and hold important experimental and therapeutic implications.


Asunto(s)
Adenocarcinoma/metabolismo , Células de la Médula Ósea/citología , Neoplasias de la Mama/metabolismo , Metástasis de la Neoplasia , Osteopontina/metabolismo , Animales , Células de la Médula Ósea/metabolismo , División Celular , Línea Celular Tumoral , Movimiento Celular , Neoplasias del Colon/metabolismo , Humanos , Ratones , Ratones Desnudos , Trasplante de Neoplasias , Trasplante Heterólogo
17.
Cell ; 134(1): 62-73, 2008 Jul 11.
Artículo en Inglés | MEDLINE | ID: mdl-18614011

RESUMEN

The p53 tumor suppressor is a key mediator of cellular responses to various stresses. Here, we show that under conditions of basal physiologic and cell-culture stress, p53 inhibits expression of the CD44 cell-surface molecule via binding to a noncanonical p53-binding sequence in the CD44 promoter. This interaction enables an untransformed cell to respond to stress-induced, p53-dependent cytostatic and apoptotic signals that would otherwise be blocked by the actions of CD44. In the absence of p53 function, the resulting derepressed CD44 expression is essential for the growth and tumor-initiating ability of highly tumorigenic mammary epithelial cells. In both tumorigenic and nontumorigenic cells, CD44's expression is positively regulated by p63, a paralogue of p53. Our data indicate that CD44 is a key tumor-promoting agent in transformed tumor cells lacking p53 function. They also suggest that the derepression of CD44 resulting from inactivation of p53 can potentially aid the survival of immortalized, premalignant cells.


Asunto(s)
Receptores de Hialuranos/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Animales , Línea Celular , Línea Celular Tumoral , Células Epiteliales/metabolismo , Regulación Neoplásica de la Expresión Génica , Humanos , Proteínas de la Membrana/metabolismo , Ratones , Proteína p53 Supresora de Tumor/genética
18.
Cell ; 133(4): 704-15, 2008 May 16.
Artículo en Inglés | MEDLINE | ID: mdl-18485877

RESUMEN

The epithelial-mesenchymal transition (EMT) is a key developmental program that is often activated during cancer invasion and metastasis. We here report that the induction of an EMT in immortalized human mammary epithelial cells (HMLEs) results in the acquisition of mesenchymal traits and in the expression of stem-cell markers. Furthermore, we show that those cells have an increased ability to form mammospheres, a property associated with mammary epithelial stem cells. Independent of this, stem cell-like cells isolated from HMLE cultures form mammospheres and express markers similar to those of HMLEs that have undergone an EMT. Moreover, stem-like cells isolated either from mouse or human mammary glands or mammary carcinomas express EMT markers. Finally, transformed human mammary epithelial cells that have undergone an EMT form mammospheres, soft agar colonies, and tumors more efficiently. These findings illustrate a direct link between the EMT and the gain of epithelial stem cell properties.


Asunto(s)
Células Epiteliales/citología , Glándulas Mamarias Animales/citología , Glándulas Mamarias Humanas/citología , Células Madre/citología , Células Madre Adultas/citología , Animales , Antígeno CD24/metabolismo , Transformación Celular Neoplásica , Células Cultivadas , Humanos , Receptores de Hialuranos/metabolismo , Mesodermo/citología , Mesodermo/metabolismo , Ratones , Células Madre Neoplásicas/citología , Esferoides Celulares , Células Tumorales Cultivadas
19.
Nature ; 543(7647): 681-686, 2017 03 30.
Artículo en Inglés | MEDLINE | ID: mdl-28329758

RESUMEN

Post-mitotic, differentiated cells exhibit a variety of characteristics that contrast with those of actively growing neoplastic cells, such as the expression of cell-cycle inhibitors and differentiation factors. We hypothesized that the gene expression profiles of these differentiated cells could reveal the identities of genes that may function as tumour suppressors. Here we show, using in vitro and in vivo studies in mice and humans, that the mitochondrial protein LACTB potently inhibits the proliferation of breast cancer cells. Its mechanism of action involves alteration of mitochondrial lipid metabolism and differentiation of breast cancer cells. This is achieved, at least in part, through reduction of the levels of mitochondrial phosphatidylserine decarboxylase, which is involved in the synthesis of mitochondrial phosphatidylethanolamine. These observations uncover a novel mitochondrial tumour suppressor and demonstrate a connection between mitochondrial lipid metabolism and the differentiation program of breast cancer cells, thereby revealing a previously undescribed mechanism of tumour suppression.


Asunto(s)
Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Diferenciación Celular , Metabolismo de los Lípidos , Proteínas de la Membrana/metabolismo , Proteínas Mitocondriales/metabolismo , Proteínas Supresoras de Tumor/metabolismo , beta-Lactamasas/metabolismo , Animales , Neoplasias de la Mama/genética , Carboxiliasas/metabolismo , Diferenciación Celular/genética , Línea Celular Tumoral , Regulación hacia Abajo , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Metabolismo de los Lípidos/genética , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos NOD , Ratones SCID , Mitocondrias/enzimología , Mitocondrias/metabolismo , Proteínas Mitocondriales/genética , Fosfatidiletanolaminas/metabolismo , Proteínas Supresoras de Tumor/genética , beta-Lactamasas/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA