Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 180.015
Filtrar
Más filtros

Intervalo de año de publicación
1.
Cell ; 187(10): 2359-2374.e18, 2024 May 09.
Artículo en Inglés | MEDLINE | ID: mdl-38653240

RESUMEN

Brown adipose tissue (BAT) is best known for thermogenesis. Rodent studies demonstrated that enhanced BAT thermogenesis is tightly associated with increased energy expenditure, reduced body weight, and improved glucose homeostasis. However, human BAT is protective against type 2 diabetes, independent of body weight. The mechanism underlying this dissociation remains unclear. Here, we report that impaired mitochondrial catabolism of branched-chain amino acids (BCAAs) in BAT, by deleting mitochondrial BCAA carriers (MBCs), caused systemic insulin resistance without affecting energy expenditure and body weight. Brown adipocytes catabolized BCAA in the mitochondria as nitrogen donors for the biosynthesis of non-essential amino acids and glutathione. Impaired mitochondrial BCAA-nitrogen flux in BAT resulted in increased oxidative stress, decreased hepatic insulin signaling, and decreased circulating BCAA-derived metabolites. A high-fat diet attenuated BCAA-nitrogen flux and metabolite synthesis in BAT, whereas cold-activated BAT enhanced the synthesis. This work uncovers a metabolite-mediated pathway through which BAT controls metabolic health beyond thermogenesis.


Asunto(s)
Tejido Adiposo Pardo , Aminoácidos de Cadena Ramificada , Resistencia a la Insulina , Mitocondrias , Nitrógeno , Termogénesis , Tejido Adiposo Pardo/metabolismo , Animales , Aminoácidos de Cadena Ramificada/metabolismo , Ratones , Nitrógeno/metabolismo , Mitocondrias/metabolismo , Masculino , Humanos , Metabolismo Energético , Ratones Endogámicos C57BL , Estrés Oxidativo , Insulina/metabolismo , Dieta Alta en Grasa , Adipocitos Marrones/metabolismo , Transducción de Señal
2.
Nat Rev Mol Cell Biol ; 25(9): 701-719, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-38689066

RESUMEN

Oxidation-reduction (redox) reactions are central to the existence of life. Reactive species of oxygen, nitrogen and sulfur mediate redox control of a wide range of essential cellular processes. Yet, excessive levels of oxidants are associated with ageing and many diseases, including cardiological and neurodegenerative diseases, and cancer. Hence, maintaining the fine-tuned steady-state balance of reactive species production and removal is essential. Here, we discuss new insights into the dynamic maintenance of redox homeostasis (that is, redox homeodynamics) and the principles underlying biological redox organization, termed the 'redox code'. We survey how redox changes result in stress responses by hormesis mechanisms, and how the lifelong cumulative exposure to environmental agents, termed the 'exposome', is communicated to cells through redox signals. Better understanding of the molecular and cellular basis of redox biology will guide novel redox medicine approaches aimed at preventing and treating diseases associated with disturbed redox regulation.


Asunto(s)
Oxidación-Reducción , Humanos , Animales , Especies Reactivas de Oxígeno/metabolismo , Homeostasis , Estrés Oxidativo , Transducción de Señal , Hormesis
3.
Nat Rev Mol Cell Biol ; 25(1): 13-33, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-37714962

RESUMEN

Several different reactive oxygen species (ROS) are generated in vivo. They have roles in the development of certain human diseases whilst also performing physiological functions. ROS are counterbalanced by an antioxidant defence network, which functions to modulate ROS levels to allow their physiological roles whilst minimizing the oxidative damage they cause that can contribute to disease development. This Review describes the mechanisms of action of antioxidants synthesized in vivo, antioxidants derived from the human diet and synthetic antioxidants developed as therapeutic agents, with a focus on the gaps in our current knowledge and the approaches needed to close them. The Review also explores the reasons behind the successes and failures of antioxidants in treating or preventing human disease. Antioxidants may have special roles in the gastrointestinal tract, and many lifestyle features known to promote health (especially diet, exercise and the control of blood glucose and cholesterol levels) may be acting, at least in part, by antioxidant mechanisms. Certain reactive sulfur species may be important antioxidants but more accurate determinations of their concentrations in vivo are needed to help assess their contributions.


Asunto(s)
Antioxidantes , Promoción de la Salud , Humanos , Antioxidantes/farmacología , Antioxidantes/metabolismo , Especies Reactivas de Oxígeno , Estrés Oxidativo
4.
Nat Rev Mol Cell Biol ; 25(2): 133-155, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-37783783

RESUMEN

In mammals, hundreds of proteins use iron in a multitude of cellular functions, including vital processes such as mitochondrial respiration, gene regulation and DNA synthesis or repair. Highly orchestrated regulatory systems control cellular and systemic iron fluxes ensuring sufficient iron delivery to target proteins is maintained, while limiting its potentially deleterious effects in iron-mediated oxidative cell damage and ferroptosis. In this Review, we discuss how cells acquire, traffick and export iron and how stored iron is mobilized for iron-sulfur cluster and haem biogenesis. Furthermore, we describe how these cellular processes are fine-tuned by the combination of various sensory and regulatory systems, such as the iron-regulatory protein (IRP)-iron-responsive element (IRE) network, the nuclear receptor co-activator 4 (NCOA4)-mediated ferritinophagy pathway, the prolyl hydroxylase domain (PHD)-hypoxia-inducible factor (HIF) axis or the nuclear factor erythroid 2-related factor 2 (NRF2) regulatory hub. We further describe how these pathways interact with systemic iron homeostasis control through the hepcidin-ferroportin axis to ensure appropriate iron fluxes. This knowledge is key for the identification of novel therapeutic opportunities to prevent diseases of cellular and/or systemic iron mismanagement.


Asunto(s)
Hierro , Factores de Transcripción , Animales , Hierro/metabolismo , Proteínas Reguladoras del Hierro/genética , Proteínas Reguladoras del Hierro/metabolismo , Factores de Transcripción/metabolismo , Homeostasis/fisiología , Estrés Oxidativo , Mamíferos/metabolismo
5.
Cell ; 184(6): 1455-1468, 2021 03 18.
Artículo en Inglés | MEDLINE | ID: mdl-33657411

RESUMEN

Environmental insults impair human health around the world. Contaminated air, water, soil, food, and occupational and household settings expose humans of all ages to a plethora of chemicals and environmental stressors. We propose eight hallmarks of environmental insults that jointly underpin the damaging impact of environmental exposures during the lifespan. Specifically, they include oxidative stress and inflammation, genomic alterations and mutations, epigenetic alterations, mitochondrial dysfunction, endocrine disruption, altered intercellular communication, altered microbiome communities, and impaired nervous system function. They provide a framework to understand why complex mixtures of environmental exposures induce severe health effects even at relatively modest concentrations.


Asunto(s)
Exposición a Riesgos Ambientales , Antioxidantes/análisis , Microbioma Gastrointestinal , Humanos , Inflamación/patología , Mutación/genética , Estrés Oxidativo
6.
Cell ; 184(8): 1990-2019, 2021 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-33811810

RESUMEN

The population is aging at a rate never seen before in human history. As the number of elderly adults grows, it is imperative we expand our understanding of the underpinnings of aging biology. Human lungs are composed of a unique panoply of cell types that face ongoing chemical, mechanical, biological, immunological, and xenobiotic stress over a lifetime. Yet, we do not fully appreciate the mechanistic drivers of lung aging and why age increases the risk of parenchymal lung disease, fatal respiratory infection, and primary lung cancer. Here, we review the molecular and cellular aspects of lung aging, local stress response pathways, and how the aging process predisposes to the pathogenesis of pulmonary disease. We place these insights into context of the COVID-19 pandemic and discuss how innate and adaptive immunity within the lung is altered with age.


Asunto(s)
Envejecimiento , Senescencia Celular , Enfermedades Pulmonares , Pulmón , Inmunidad Adaptativa , Anciano , Envejecimiento/inmunología , Envejecimiento/patología , COVID-19/inmunología , COVID-19/patología , Humanos , Pulmón/inmunología , Pulmón/patología , Enfermedades Pulmonares/inmunología , Enfermedades Pulmonares/patología , Estrés Oxidativo
7.
Cell ; 182(6): 1377-1378, 2020 09 17.
Artículo en Inglés | MEDLINE | ID: mdl-32946778

RESUMEN

Although oncogenic mutations predispose tissue stem cells to tumor initiation, the rate-limiting processes for stem cell immortalization remain unknown. In this issue of Cell, Bonnay et al. identify enhanced electron transport chain activity as a critical determinant of this process, establishing metabolic reprogramming as limiting for tumor initiation.


Asunto(s)
Transformación Celular Neoplásica , Células-Madre Neurales , Carcinogénesis , Humanos , Estrés Oxidativo
8.
Cell ; 181(6): 1307-1328.e15, 2020 06 11.
Artículo en Inglés | MEDLINE | ID: mdl-32502393

RESUMEN

The view that sleep is essential for survival is supported by the ubiquity of this behavior, the apparent existence of sleep-like states in the earliest animals, and the fact that severe sleep loss can be lethal. The cause of this lethality is unknown. Here we show, using flies and mice, that sleep deprivation leads to accumulation of reactive oxygen species (ROS) and consequent oxidative stress, specifically in the gut. ROS are not just correlates of sleep deprivation but drivers of death: their neutralization prevents oxidative stress and allows flies to have a normal lifespan with little to no sleep. The rescue can be achieved with oral antioxidant compounds or with gut-targeted transgenic expression of antioxidant enzymes. We conclude that death upon severe sleep restriction can be caused by oxidative stress, that the gut is central in this process, and that survival without sleep is possible when ROS accumulation is prevented. VIDEO ABSTRACT.


Asunto(s)
Tracto Gastrointestinal/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Privación de Sueño/metabolismo , Sueño/fisiología , Animales , Antioxidantes/metabolismo , Drosophila , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos CBA , Estrés Oxidativo/fisiología
9.
Cell ; 180(5): 968-983.e24, 2020 03 05.
Artículo en Inglés | MEDLINE | ID: mdl-32109415

RESUMEN

Mammalian tissues engage in specialized physiology that is regulated through reversible modification of protein cysteine residues by reactive oxygen species (ROS). ROS regulate a myriad of biological processes, but the protein targets of ROS modification that drive tissue-specific physiology in vivo are largely unknown. Here, we develop Oximouse, a comprehensive and quantitative mapping of the mouse cysteine redox proteome in vivo. We use Oximouse to establish several paradigms of physiological redox signaling. We define and validate cysteine redox networks within each tissue that are tissue selective and underlie tissue-specific biology. We describe a common mechanism for encoding cysteine redox sensitivity by electrostatic gating. Moreover, we comprehensively identify redox-modified disease networks that remodel in aged mice, establishing a systemic molecular basis for the long-standing proposed links between redox dysregulation and tissue aging. We provide the Oximouse compendium as a framework for understanding mechanisms of redox regulation in physiology and aging.


Asunto(s)
Envejecimiento/genética , Cisteína/genética , Proteínas/genética , Proteoma/genética , Envejecimiento/metabolismo , Envejecimiento/patología , Animales , Cisteína/metabolismo , Humanos , Ratones , Especificidad de Órganos/genética , Oxidación-Reducción , Estrés Oxidativo/genética , Proteómica/métodos , Especies Reactivas de Oxígeno , Transducción de Señal/genética
10.
Cell ; 183(1): 46-61.e21, 2020 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-32941802

RESUMEN

Metazoan organisms rely on conserved stress response pathways to alleviate adverse conditions and preserve cellular integrity. Stress responses are particularly important in stem cells that provide lifetime support for tissue formation and repair, but how these protective systems are integrated into developmental programs is poorly understood. Here we used myoblast differentiation to identify the E3 ligase CUL2FEM1B and its substrate FNIP1 as core components of the reductive stress response. Reductive stress, as caused by prolonged antioxidant signaling or mitochondrial inactivity, reverts the oxidation of invariant Cys residues in FNIP1 and allows CUL2FEM1B to recognize its target. The ensuing proteasomal degradation of FNIP1 restores mitochondrial activity to preserve redox homeostasis and stem cell integrity. The reductive stress response is therefore built around a ubiquitin-dependent rheostat that tunes mitochondrial activity to redox needs and implicates metabolic control in coordination of stress and developmental signaling.


Asunto(s)
Proteínas Portadoras/metabolismo , Estrés Oxidativo/fisiología , Especies Reactivas de Oxígeno/metabolismo , Antioxidantes/metabolismo , Proteínas Portadoras/genética , Diferenciación Celular , Células HEK293 , Homeostasis , Humanos , Proteína 1 Asociada A ECH Tipo Kelch/metabolismo , Mitocondrias , Desarrollo de Músculos/fisiología , Mioblastos/metabolismo , Factor 2 Relacionado con NF-E2/metabolismo , Transducción de Señal , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinación
11.
Cell ; 182(4): 976-991.e19, 2020 08 20.
Artículo en Inglés | MEDLINE | ID: mdl-32702314

RESUMEN

Although complex inflammatory-like alterations are observed around the amyloid plaques of Alzheimer's disease (AD), little is known about the molecular changes and cellular interactions that characterize this response. We investigate here, in an AD mouse model, the transcriptional changes occurring in tissue domains in a 100-µm diameter around amyloid plaques using spatial transcriptomics. We demonstrate early alterations in a gene co-expression network enriched for myelin and oligodendrocyte genes (OLIGs), whereas a multicellular gene co-expression network of plaque-induced genes (PIGs) involving the complement system, oxidative stress, lysosomes, and inflammation is prominent in the later phase of the disease. We confirm the majority of the observed alterations at the cellular level using in situ sequencing on mouse and human brain sections. Genome-wide spatial transcriptomics analysis provides an unprecedented approach to untangle the dysregulated cellular network in the vicinity of pathogenic hallmarks of AD and other brain diseases.


Asunto(s)
Enfermedad de Alzheimer/patología , Análisis de Secuencia de ADN/métodos , Transcriptoma , Enfermedad de Alzheimer/genética , Amiloide/metabolismo , Péptidos beta-Amiloides/genética , Péptidos beta-Amiloides/metabolismo , Animales , Encéfalo/metabolismo , Encéfalo/patología , Proteínas del Sistema Complemento/genética , Proteínas del Sistema Complemento/metabolismo , Modelos Animales de Enfermedad , Perfilación de la Expresión Génica , Humanos , Lisosomas/genética , Lisosomas/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Vaina de Mielina/genética , Vaina de Mielina/metabolismo , Estrés Oxidativo/genética
12.
Annu Rev Biochem ; 88: 605-633, 2019 06 20.
Artículo en Inglés | MEDLINE | ID: mdl-31018111

RESUMEN

Reactive oxygen species (ROS) encompass a collection of intricately linked chemical entities characterized by individually distinct physicochemical properties and biological reactivities. Although excessive ROS generation is well known to underpin disease development, it has become increasingly evident that ROS also play central roles in redox regulation and normal physiology. A major challenge in uncovering the relevant biological mechanisms and deconvoluting the apparently paradoxical roles of distinct ROS in human health and disease lies in the selective and sensitive detection of these transient species in the complex biological milieu. Small-molecule-based fluorescent sensors enable molecular imaging of ROS with great spatial and temporal resolution and have thus been appreciated as excellent tools for aiding discoveries in modern redox biology. We review a selection of state-of-the-art sensors with demonstrated utility in biological systems. By providing a systematic overview based on underlying chemical sensing mechanisms, we wish to highlight the strengths and weaknesses in prior sensor works and propose some guiding principles for the development of future probes.


Asunto(s)
Técnicas Biosensibles/métodos , Especies Reactivas de Oxígeno/análisis , Colorantes Fluorescentes , Imagen Óptica , Oxidación-Reducción , Estrés Oxidativo
13.
Nat Immunol ; 23(12): 1714-1725, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36411380

RESUMEN

Increasing evidence indicates close interaction between immune cells and the brain, revising the traditional view of the immune privilege of the brain. However, the specific mechanisms by which immune cells promote normal neural function are not entirely understood. Mucosal-associated invariant T cells (MAIT cells) are a unique type of innate-like T cell with molecular and functional properties that remain to be better characterized. In the present study, we report that MAIT cells are present in the meninges and express high levels of antioxidant molecules. MAIT cell deficiency in mice results in the accumulation of reactive oxidative species in the meninges, leading to reduced expression of junctional protein and meningeal barrier leakage. The presence of MAIT cells restricts neuroinflammation in the brain and preserves learning and memory. Together, our work reveals a new functional role for MAIT cells in the meninges and suggests that meningeal immune cells can help maintain normal neural function by preserving meningeal barrier homeostasis and integrity.


Asunto(s)
Células T Invariantes Asociadas a Mucosa , Animales , Ratones , Encéfalo , Meninges , Cognición , Estrés Oxidativo
14.
Nat Rev Mol Cell Biol ; 23(1): 56-73, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34518687

RESUMEN

Dietary restriction with adequate nutrition is the gold standard for delaying ageing and extending healthspan and lifespan in diverse species, including rodents and non-human primates. In this Review, we discuss the effects of dietary restriction in these mammalian model organisms and discuss accumulating data that suggest that dietary restriction results in many of the same physiological, metabolic and molecular changes responsible for the prevention of multiple ageing-associated diseases in humans. We further discuss how different forms of fasting, protein restriction and specific reductions in the levels of essential amino acids such as methionine and the branched-chain amino acids selectively impact the activity of AKT, FOXO, mTOR, nicotinamide adenine dinucleotide (NAD+), AMP-activated protein kinase (AMPK) and fibroblast growth factor 21 (FGF21), which are key components of some of the most important nutrient-sensing geroprotective signalling pathways that promote healthy longevity.


Asunto(s)
Restricción Calórica , Salud , Longevidad/fisiología , Animales , Modelos Animales de Enfermedad , Humanos , Estrés Oxidativo
15.
Cell ; 177(6): 1507-1521.e16, 2019 05 30.
Artículo en Inglés | MEDLINE | ID: mdl-31031004

RESUMEN

Friedreich's ataxia (FRDA) is a devastating, multisystemic disorder caused by recessive mutations in the mitochondrial protein frataxin (FXN). FXN participates in the biosynthesis of Fe-S clusters and is considered to be essential for viability. Here we report that when grown in 1% ambient O2, FXN null yeast, human cells, and nematodes are fully viable. In human cells, hypoxia restores steady-state levels of Fe-S clusters and normalizes ATF4, NRF2, and IRP2 signaling events associated with FRDA. Cellular studies and in vitro reconstitution indicate that hypoxia acts through HIF-independent mechanisms that increase bioavailable iron as well as directly activate Fe-S synthesis. In a mouse model of FRDA, breathing 11% O2 attenuates the progression of ataxia, whereas breathing 55% O2 hastens it. Our work identifies oxygen as a key environmental variable in the pathogenesis associated with FXN depletion, with important mechanistic and therapeutic implications.


Asunto(s)
Hipoxia/metabolismo , Proteínas de Unión a Hierro/metabolismo , Proteínas Hierro-Azufre/metabolismo , Factor de Transcripción Activador 4/metabolismo , Animales , Caenorhabditis elegans/metabolismo , Femenino , Ataxia de Friedreich/metabolismo , Células HEK293 , Humanos , Hipoxia/fisiopatología , Hierro/metabolismo , Proteína 2 Reguladora de Hierro/metabolismo , Proteínas de Unión a Hierro/fisiología , Proteínas Hierro-Azufre/fisiología , Células K562 , Masculino , Ratones , Ratones Noqueados , Mitocondrias/metabolismo , Proteínas Mitocondriales/metabolismo , Factor 2 Relacionado con NF-E2/metabolismo , Estrés Oxidativo , Saccharomyces cerevisiae/metabolismo , Azufre/metabolismo , Frataxina
16.
Cell ; 177(3): 711-721.e8, 2019 04 18.
Artículo en Inglés | MEDLINE | ID: mdl-30982603

RESUMEN

Yeast ataxin-2, also known as Pbp1, senses the activity state of mitochondria in order to regulate TORC1. A domain of Pbp1 required to adapt cells to mitochondrial activity is of low sequence complexity. The low-complexity (LC) domain of Pbp1 forms labile, cross-ß polymers that facilitate phase transition of the protein into liquid-like or gel-like states. Phase transition for other LC domains is reliant upon widely distributed aromatic amino acids. In place of tyrosine or phenylalanine residues prototypically used for phase separation, Pbp1 contains 24 similarly disposed methionine residues. Here, we show that the Pbp1 methionine residues are sensitive to hydrogen peroxide (H2O2)-mediated oxidation in vitro and in living cells. Methionine oxidation melts Pbp1 liquid-like droplets in a manner reversed by methionine sulfoxide reductase enzymes. These observations explain how reversible formation of labile polymers by the Pbp1 LC domain enables the protein to function as a sensor of cellular redox state.


Asunto(s)
Proteínas Portadoras/metabolismo , Metionina/química , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/metabolismo , Secuencia de Aminoácidos , Proteínas Portadoras/química , Proteínas Portadoras/genética , Peróxido de Hidrógeno/farmacología , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Metionina/metabolismo , Metionina Sulfóxido Reductasas/metabolismo , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Mutagénesis Sitio-Dirigida , Oxidación-Reducción , Estrés Oxidativo/efectos de los fármacos , Transición de Fase , Dominios Proteicos , Proteínas de Saccharomyces cerevisiae/química , Proteínas de Saccharomyces cerevisiae/genética
17.
Cell ; 177(3): 737-750.e15, 2019 04 18.
Artículo en Inglés | MEDLINE | ID: mdl-31002798

RESUMEN

The proteasome mediates selective protein degradation and is dynamically regulated in response to proteotoxic challenges. SKN-1A/Nrf1, an endoplasmic reticulum (ER)-associated transcription factor that undergoes N-linked glycosylation, serves as a sensor of proteasome dysfunction and triggers compensatory upregulation of proteasome subunit genes. Here, we show that the PNG-1/NGLY1 peptide:N-glycanase edits the sequence of SKN-1A protein by converting particular N-glycosylated asparagine residues to aspartic acid. Genetically introducing aspartates at these N-glycosylation sites bypasses the requirement for PNG-1/NGLY1, showing that protein sequence editing rather than deglycosylation is key to SKN-1A function. This pathway is required to maintain sufficient proteasome expression and activity, and SKN-1A hyperactivation confers resistance to the proteotoxicity of human amyloid beta peptide. Deglycosylation-dependent protein sequence editing explains how ER-associated and cytosolic isoforms of SKN-1 perform distinct cytoprotective functions corresponding to those of mammalian Nrf1 and Nrf2. Thus, we uncover an unexpected mechanism by which N-linked glycosylation regulates protein function and proteostasis.


Asunto(s)
Proteínas de Caenorhabditis elegans/metabolismo , Proteínas de Unión al ADN/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Factores de Transcripción/metabolismo , Secuencia de Aminoácidos , Animales , Asparagina/metabolismo , Bortezomib/farmacología , Sistemas CRISPR-Cas/genética , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/química , Proteínas de Caenorhabditis elegans/genética , Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/genética , Retículo Endoplásmico/metabolismo , Edición Génica , Regulación de la Expresión Génica/efectos de los fármacos , Estrés Oxidativo , Complejo de la Endopetidasa Proteasomal/genética , Subunidades de Proteína/química , Subunidades de Proteína/genética , Subunidades de Proteína/metabolismo , Alineación de Secuencia , Factores de Transcripción/química , Factores de Transcripción/genética
18.
Immunity ; 57(5): 941-956, 2024 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-38749397

RESUMEN

Ferroptosis is a type of regulated cell death that drives the pathophysiology of many diseases. Oxidative stress is detectable in many types of regulated cell death, but only ferroptosis involves lipid peroxidation and iron dependency. Ferroptosis originates and propagates from several organelles, including the mitochondria, endoplasmic reticulum, Golgi, and lysosomes. Recent data have revealed that immune cells can both induce and undergo ferroptosis. A mechanistic understanding of how ferroptosis regulates immunity is critical to understanding how ferroptosis controls immune responses and how this is dysregulated in disease. Translationally, more work is needed to produce ferroptosis-modulating immunotherapeutics. This review focuses on the role of ferroptosis in immune-related diseases, including infection, autoimmune diseases, and cancer. We discuss how ferroptosis is regulated in immunity, how this regulation contributes to disease pathogenesis, and how targeting ferroptosis may lead to novel therapies.


Asunto(s)
Ferroptosis , Hierro , Ferroptosis/inmunología , Humanos , Animales , Hierro/metabolismo , Neoplasias/inmunología , Neoplasias/metabolismo , Peroxidación de Lípido/inmunología , Enfermedades Autoinmunes/inmunología , Inmunidad , Estrés Oxidativo/inmunología , Mitocondrias/metabolismo , Mitocondrias/inmunología
19.
Nat Rev Mol Cell Biol ; 22(5): 346-366, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33504982

RESUMEN

The ability of cells to deal with different types of stressful situations in a precise and coordinated manner is key for survival and involves various signalling networks. Over the past 25 years, p38 kinases - in particular, p38α - have been implicated in the cellular response to stress at many levels. These span from environmental and intracellular stresses, such as hyperosmolarity, oxidative stress or DNA damage, to physiological situations that involve important cellular changes such as differentiation. Given that p38α controls a plethora of functions, dysregulation of this pathway has been linked to diseases such as inflammation, immune disorders or cancer, suggesting the possibility that targeting p38α could be of therapeutic interest. In this Review, we discuss the organization of this signalling pathway focusing on the diversity of p38α substrates, their mechanisms and their links to particular cellular functions. We then address how the different cellular responses can be generated depending on the signal received and the cell type, and highlight the roles of this kinase in human physiology and in pathological contexts.


Asunto(s)
Inflamación/genética , Neoplasias/genética , Estrés Oxidativo/genética , Diferenciación Celular/genética , Humanos , Inflamación/patología , Neoplasias/patología , Fosforilación , Transducción de Señal/genética
20.
Cell ; 175(3): 809-821.e19, 2018 10 18.
Artículo en Inglés | MEDLINE | ID: mdl-30270044

RESUMEN

Approximately 10% of human protein kinases are believed to be inactive and named pseudokinases because they lack residues required for catalysis. Here, we show that the highly conserved pseudokinase selenoprotein-O (SelO) transfers AMP from ATP to Ser, Thr, and Tyr residues on protein substrates (AMPylation), uncovering a previously unrecognized activity for a member of the protein kinase superfamily. The crystal structure of a SelO homolog reveals a protein kinase-like fold with ATP flipped in the active site, thus providing a structural basis for catalysis. SelO pseudokinases localize to the mitochondria and AMPylate proteins involved in redox homeostasis. Consequently, SelO activity is necessary for the proper cellular response to oxidative stress. Our results suggest that AMPylation may be a more widespread post-translational modification than previously appreciated and that pseudokinases should be analyzed for alternative transferase activities.


Asunto(s)
Adenosina Monofosfato/metabolismo , Dominio Catalítico , Procesamiento Proteico-Postraduccional , Selenoproteínas/metabolismo , Secuencia Conservada , Humanos , Proteínas Mitocondriales/genética , Proteínas Mitocondriales/metabolismo , Estrés Oxidativo , Selenoproteínas/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA