Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
1.
Cell ; 183(1): 94-109.e23, 2020 10 01.
Article in English | MEDLINE | ID: mdl-32937105

ABSTRACT

Cardiomyocytes are subjected to the intense mechanical stress and metabolic demands of the beating heart. It is unclear whether these cells, which are long-lived and rarely renew, manage to preserve homeostasis on their own. While analyzing macrophages lodged within the healthy myocardium, we discovered that they actively took up material, including mitochondria, derived from cardiomyocytes. Cardiomyocytes ejected dysfunctional mitochondria and other cargo in dedicated membranous particles reminiscent of neural exophers, through a process driven by the cardiomyocyte's autophagy machinery that was enhanced during cardiac stress. Depletion of cardiac macrophages or deficiency in the phagocytic receptor Mertk resulted in defective elimination of mitochondria from the myocardial tissue, activation of the inflammasome, impaired autophagy, accumulation of anomalous mitochondria in cardiomyocytes, metabolic alterations, and ventricular dysfunction. Thus, we identify an immune-parenchymal pair in the murine heart that enables transfer of unfit material to preserve metabolic stability and organ function. VIDEO ABSTRACT.


Subject(s)
Macrophages/metabolism , Mitochondria/metabolism , Myocytes, Cardiac/metabolism , Aged , Animals , Apoptosis , Autophagy , Female , Heart/physiology , Homeostasis , Humans , Macrophages/physiology , Male , Mice , Mice, Inbred C57BL , Middle Aged , Mitochondria/physiology , Myocardial Infarction/metabolism , Myocardium/metabolism , Myocytes, Cardiac/physiology , Phagocytosis/physiology , Reactive Oxygen Species/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , c-Mer Tyrosine Kinase/metabolism
2.
Dev Biol ; 475: 30-36, 2021 07.
Article in English | MEDLINE | ID: mdl-33652024

ABSTRACT

Cell Competition is a selective process by which viable cells are eliminated from developing or adult tissues by interactions with their neighbors. In many cases, the eliminated cells (losers) display reduced fitness, yet they would be able to sustain tissue growth or maintenance in a homotypic environment, and are only eliminated when confronted with surrounding wild type cells (winners). In addition, cells with oncogenic mutations that do not show reduced fitness can also be eliminated from tissues when surrounded by wild type cells. Depending on the context, transformed cells can also become supercompetitors and eliminate surrounding wild type cells, thereby promoting tumor formation. Several factors have been shown to play essential roles in Cell Competition, including genes relevant in developmental growth, tumor formation and epithelial apico-basal polarity. Recent discoveries, however, suggest that energy metabolism plays a central role in very different models of cell competition. Here we review the involvement of mitochondrial dynamics and metabolism, autophagy and nutritional status in cell competition and discuss the possible implications of this emerging field.


Subject(s)
Cell Competition/physiology , Mitochondria/metabolism , Mitochondrial Dynamics/physiology , Animals , Apoptosis/physiology , Autophagy/physiology , Cell Communication/physiology , Energy Metabolism/physiology , Glycolysis/physiology , Humans , Mitochondrial Dynamics/genetics , Nutritional Status/physiology , Signal Transduction/physiology
3.
EMBO J ; 36(12): 1688-1706, 2017 06 14.
Article in English | MEDLINE | ID: mdl-28465321

ABSTRACT

Retinal ganglion cells (RGCs) are the sole projecting neurons of the retina and their axons form the optic nerve. Here, we show that embryogenesis-associated mouse RGC differentiation depends on mitophagy, the programmed autophagic clearance of mitochondria. The elimination of mitochondria during RGC differentiation was coupled to a metabolic shift with increased lactate production and elevated expression of glycolytic enzymes at the mRNA level. Pharmacological and genetic inhibition of either mitophagy or glycolysis consistently inhibited RGC differentiation. Local hypoxia triggered expression of the mitophagy regulator BCL2/adenovirus E1B 19-kDa-interacting protein 3-like (BNIP3L, best known as NIX) at peak RGC differentiation. Retinas from NIX-deficient mice displayed increased mitochondrial mass, reduced expression of glycolytic enzymes and decreased neuronal differentiation. Similarly, we provide evidence that NIX-dependent mitophagy contributes to mitochondrial elimination during macrophage polarization towards the proinflammatory and more glycolytic M1 phenotype, but not to M2 macrophage differentiation, which primarily relies on oxidative phosphorylation. In summary, developmentally controlled mitophagy promotes a metabolic switch towards glycolysis, which in turn contributes to cellular differentiation in several distinct developmental contexts.


Subject(s)
Cell Differentiation , Glycolysis , Mitophagy , Retina/embryology , Retinal Ganglion Cells/physiology , Animals , Membrane Proteins/deficiency , Membrane Proteins/metabolism , Mice , Mice, Knockout , Mitochondrial Proteins/deficiency , Mitochondrial Proteins/metabolism
4.
Methods ; 75: 79-86, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25644445

ABSTRACT

Autophagy is a highly dynamic process that mediates the degradation of cellular constituents inside lysosomes. It is characterized by the formation of autophagosomes, double membrane organelles that engulf cytosolic components and organelles and degrade their contents upon fusion with lysosomes. Upregulation of autophagy in response to specific stimuli can be determined by evaluating autophagic flux. This is achieved by comparing the number of autophagosomes in the absence and presence of lysosomal inhibitors. While the determination of autophagic flux in isolated cells is well-documented, few studies have described its determination in tissues or in vivo. Here, we describe the evaluation of autophagic flux both in vivo and ex vivo in several tissues, after treatment with lysosomal inhibitors and exposure to classical autophagy-inducing stimuli. This method uses LC3 lipidation, as determined by Western blot, fluorescence microscopy and flow cytometry. Our findings demonstrate that autophagic flux can be evaluated in vivo and ex vivo in several tissues.


Subject(s)
Autophagy/genetics , Liver/ultrastructure , Microscopy, Fluorescence/methods , Retina/ultrastructure , Animals , Cerebellum/metabolism , Cerebellum/ultrastructure , Leupeptins/chemistry , Liver/metabolism , Lysosomes/genetics , Lysosomes/metabolism , Lysosomes/ultrastructure , Mice , Retina/metabolism
5.
Autophagy ; 14(5): 915-917, 2018.
Article in English | MEDLINE | ID: mdl-28614042

ABSTRACT

Macroautophagy/autophagy is the process by which cellular components are degraded and recycled within the lysosome. These components include mitochondria, the selective degradation of which is known as mitophagy. Mitochondria are dynamic organelles that constantly adapt their morphology, function, and number to accommodate the metabolic needs of the cell. Extensive metabolic reconfiguration occurs during cell differentiation, when mitochondrial activity increases in most cell types. However, our data demonstrate that during physiologic retinal ganglion cell (RGC) development, mitophagy-dependent metabolic reprogramming toward glycolysis regulates numbers of RGCs, which are the first neurons to differentiate in the retina and whose axons form the optic nerve. We show that during retinal development tissue hypoxia triggers HIF1A/HIF-1 stabilization, resulting in increased expression of the mitophagy receptor BNIP3L/NIX. BNIP3L-dependent mitophagy results in a metabolic shift toward glycolysis essential for RGC neurogenesis. Moreover, we demonstrate that BNIP3L-dependent mitophagy also regulates the polarization of proinflammatory/M1 macrophages, which undergo glycolysis-dependent differentiation during the inflammatory response. Our results uncover a new link between hypoxia, mitophagy, and metabolic reprogramming in the differentiation of several cell types in vivo. These findings may have important implications for neurodegenerative, metabolic and other diseases in which mitochondrial dysfunction and metabolic alterations play a prominent role.


Subject(s)
Autophagy , Mitophagy , Apoptosis Regulatory Proteins , Cell Differentiation , Glycolysis
6.
Mol Cell Oncol ; 4(5): e1353854, 2017.
Article in English | MEDLINE | ID: mdl-29057310

ABSTRACT

Mitophagy is the process by which cells eliminate damaged or superfluous mitochondria by degrading them within lysosomes. We show that during development, selective removal of mitochondria by autophagy induces a metabolic shift toward glycolysis that is essential for differentiation of several cell types. These findings suggest potential applications of cell-fate manipulation strategies targeting mitophagy.

7.
Cells ; 6(4)2017 Oct 22.
Article in English | MEDLINE | ID: mdl-29065501

ABSTRACT

Autophagy is a catabolic pathway that mediates the degradation and recycling of intracellular components, and is a key player in a variety of physiological processes in cells and tissues. Recent studies of autophagy in the eye suggest that this pathway is fundamental for the preservation of retinal homeostasis. Given its accessible location outside the brain, the retina is an ideal organ in which to study the central nervous system and a wide range of neuronal processes, from development to neurodegeneration. Here we review several methods used to assess autophagy in the retina in both physiological and pathological conditions.

8.
Prog Retin Eye Res ; 55: 206-245, 2016 11.
Article in English | MEDLINE | ID: mdl-27566190

ABSTRACT

Autophagy is a catabolic pathway that promotes the degradation and recycling of cellular components. Proteins, lipids, and even whole organelles are engulfed in autophagosomes and delivered to the lysosome for elimination. In response to stress, autophagy mediates the degradation of cell components, which are recycled to generate the nutrients and building blocks required to sustain cellular homeostasis. Moreover, it plays an important role in cellular quality control, particularly in neurons, in which the total burden of altered proteins and damaged organelles cannot be reduced by redistribution to daughter cells through cell division. Research has only begun to examine the role of autophagy in the visual system. The retina, a light-sensitive tissue, detects and transmits electrical impulses through the optic nerve to the visual cortex in the brain. Both the retina and the eye are exposed to a variety of environmental insults and stressors, including genetic mutations and age-associated alterations that impair their function. Here, we review the main studies that have sought to explain autophagy's importance in visual function. We describe the role of autophagy in retinal development and cell differentiation, and discuss the implications of autophagy dysregulation both in physiological aging and in important diseases such as age-associated macular degeneration and glaucoma. We also address the putative role of autophagy in promoting photoreceptor survival and discuss how selective autophagy could provide alternative means of protecting retinal cells. The findings reviewed here underscore the important role of autophagy in maintaining proper retinal function and highlight novel therapeutic approaches for blindness and other diseases of the eye.


Subject(s)
Aging/physiology , Autophagy/physiology , Eye Diseases/pathology , Retina/cytology , Humans
9.
Autophagy ; 11(5): 833-43, 2015.
Article in English | MEDLINE | ID: mdl-25945953

ABSTRACT

Mitochondrial autophagy, also known as mitophagy, is an autophagosome-based mitochondrial degradation process that eliminates unwanted or damaged mitochondria after cell stress. Most studies dealing with mitophagy rely on the analysis by fluorescence microscopy of mitochondrial-autophagosome colocalization. However, given the fundamental role of mitophagy in the physiology and pathology of organisms, there is an urgent need for novel quantitative methods with which to study this process. Here, we describe a flow cytometry-based approach to determine mitophagy by using MitoTracker Deep Red, a widely used mitochondria-selective probe. Used in combination with selective inhibitors it may allow for the determination of mitophagy flux. Here, we test the validity of the use of this method in cell lines and in primary cell and tissue cultures.


Subject(s)
Flow Cytometry/methods , Mitophagy , Amino Acids/deficiency , Animals , Carbonyl Cyanide m-Chlorophenyl Hydrazone/pharmacology , Disease Models, Animal , Down-Regulation/drug effects , Embryo, Mammalian/cytology , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/metabolism , Flavonoids/pharmacology , Flavonols , HeLa Cells , Humans , Lysosomes/drug effects , Lysosomes/metabolism , Mice, Inbred C57BL , Mitochondria/drug effects , Mitochondria/metabolism , Mitophagy/drug effects , Niacinamide/pharmacology , Retina/drug effects , Retina/metabolism , Retinitis Pigmentosa/pathology
10.
Nat Cell Biol ; 17(10): 1304-16, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26322680

ABSTRACT

Blocking mitotic progression has been proposed as an attractive therapeutic strategy to impair proliferation of tumour cells. However, how cells survive during prolonged mitotic arrest is not well understood. We show here that survival during mitotic arrest is affected by the special energetic requirements of mitotic cells. Prolonged mitotic arrest results in mitophagy-dependent loss of mitochondria, accompanied by reduced ATP levels and the activation of AMPK. Oxidative respiration is replaced by glycolysis owing to AMPK-dependent phosphorylation of PFKFB3 and increased production of this protein as a consequence of mitotic-specific translational activation of its mRNA. Induction of autophagy or inhibition of AMPK or PFKFB3 results in enhanced cell death in mitosis and improves the anti-tumoral efficiency of microtubule poisons in breast cancer cells. Thus, survival of mitotic-arrested cells is limited by their metabolic requirements, a feature with potential implications in cancer therapies aimed to impair mitosis or metabolism in tumour cells.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Autophagy/physiology , Fibroblasts/metabolism , Glycolysis , M Phase Cell Cycle Checkpoints/physiology , Phosphofructokinase-2/metabolism , AMP-Activated Protein Kinases/genetics , Animals , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Apoptosis/genetics , Apoptosis/physiology , Autophagy/genetics , Blotting, Western , Cdc20 Proteins/genetics , Cdc20 Proteins/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/genetics , Cell Survival/physiology , Cells, Cultured , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Female , Fibroblasts/ultrastructure , Humans , M Phase Cell Cycle Checkpoints/genetics , MCF-7 Cells , Mice, Knockout , Mice, Nude , Microscopy, Confocal , Paclitaxel/pharmacology , Phosphofructokinase-2/genetics , RNA Interference , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL