Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 105.594
Filtrar
Más filtros

Publication year range
1.
Annu Rev Biochem ; 91: 353-380, 2022 06 21.
Artículo en Inglés | MEDLINE | ID: mdl-35303791

RESUMEN

Subcellular compartmentalization is a defining feature of all cells. In prokaryotes, compartmentalization is generally achieved via protein-based strategies. The two main classes of microbial protein compartments are bacterial microcompartments and encapsulin nanocompartments. Encapsulins self-assemble into proteinaceous shells with diameters between 24 and 42 nm and are defined by the viral HK97-fold of their shell protein. Encapsulins have the ability to encapsulate dedicated cargo proteins, including ferritin-like proteins, peroxidases, and desulfurases. Encapsulation is mediated by targeting sequences present in all cargo proteins. Encapsulins are found in many bacterial and archaeal phyla and have been suggested to play roles in iron storage, stress resistance, sulfur metabolism, and natural product biosynthesis. Phylogenetic analyses indicate that they share a common ancestor with viral capsid proteins. Many pathogens encode encapsulins, and recent evidence suggests that they may contribute toward pathogenicity. The existing information on encapsulin structure, biochemistry, biological function, and biomedical relevance is reviewed here.


Asunto(s)
Bacterias , Proteínas Bacterianas , Archaea/genética , Archaea/metabolismo , Bacterias/genética , Bacterias/metabolismo , Proteínas Bacterianas/metabolismo , Hierro/metabolismo , Filogenia
2.
Cell ; 185(14): 2401-2421, 2022 07 07.
Artículo en Inglés | MEDLINE | ID: mdl-35803244

RESUMEN

Ferroptosis, a form of cell death driven by iron-dependent lipid peroxidation, was identified as a distinct phenomenon and named a decade ago. Ferroptosis has been implicated in a broad set of biological contexts, from development to aging, immunity, and cancer. This review describes key regulators of this form of cell death within a framework of metabolism, ROS biology, and iron biology. Key concepts and major unanswered questions in the ferroptosis field are highlighted. The next decade promises to yield further breakthroughs in the mechanisms governing ferroptosis and additional ways of harnessing ferroptosis for therapeutic benefit.


Asunto(s)
Ferroptosis , Muerte Celular , Hierro/metabolismo , Peroxidación de Lípido , Especies Reactivas de Oxígeno/metabolismo
3.
Nat Immunol ; 25(3): 471-482, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38429458

RESUMEN

Persistent symptoms following SARS-CoV-2 infection are increasingly reported, although the drivers of post-acute sequelae (PASC) of COVID-19 are unclear. Here we assessed 214 individuals infected with SARS-CoV-2, with varying disease severity, for one year from COVID-19 symptom onset to determine the early correlates of PASC. A multivariate signature detected beyond two weeks of disease, encompassing unresolving inflammation, anemia, low serum iron, altered iron-homeostasis gene expression and emerging stress erythropoiesis; differentiated those who reported PASC months later, irrespective of COVID-19 severity. A whole-blood heme-metabolism signature, enriched in hospitalized patients at month 1-3 post onset, coincided with pronounced iron-deficient reticulocytosis. Lymphopenia and low numbers of dendritic cells persisted in those with PASC, and single-cell analysis reported iron maldistribution, suggesting monocyte iron loading and increased iron demand in proliferating lymphocytes. Thus, defects in iron homeostasis, dysregulated erythropoiesis and immune dysfunction due to COVID-19 possibly contribute to inefficient oxygen transport, inflammatory disequilibrium and persisting symptomatology, and may be therapeutically tractable.


Asunto(s)
COVID-19 , Hierro , Humanos , Eritropoyesis , SARS-CoV-2 , Investigadores , Progresión de la Enfermedad
4.
Nat Immunol ; 25(5): 790-801, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38664585

RESUMEN

Innate immune cells generate a multifaceted antitumor immune response, including the conservation of essential nutrients such as iron. These cells can be modulated by commensal bacteria; however, identifying and understanding how this occurs is a challenge. Here we show that the food commensal Lactiplantibacillus plantarum IMB19 augments antitumor immunity in syngeneic and xenograft mouse tumor models. Its capsular heteropolysaccharide is the major effector molecule, functioning as a ligand for TLR2. In a two-pronged manner, it skews tumor-associated macrophages to a classically active phenotype, leading to generation of a sustained CD8+ T cell response, and triggers macrophage 'nutritional immunity' to deploy the high-affinity iron transporter lipocalin-2 for capturing and sequestering iron in the tumor microenvironment. This process induces a cycle of tumor cell death, epitope expansion and subsequent tumor clearance. Together these data indicate that food commensals might be identified and developed into 'oncobiotics' for a multi-layered approach to cancer therapy.


Asunto(s)
Hierro , Microambiente Tumoral , Animales , Hierro/metabolismo , Ratones , Microambiente Tumoral/inmunología , Humanos , Macrófagos Asociados a Tumores/inmunología , Macrófagos Asociados a Tumores/metabolismo , Linfocitos T CD8-positivos/inmunología , Línea Celular Tumoral , Receptor Toll-Like 2/metabolismo , Receptor Toll-Like 2/inmunología , Ratones Endogámicos C57BL , Lipocalina 2/metabolismo , Lipocalina 2/inmunología , Femenino , Simbiosis/inmunología , Macrófagos/inmunología , Macrófagos/metabolismo , Activación de Macrófagos/inmunología , Ratones Noqueados
5.
Nat Rev Mol Cell Biol ; 25(6): 424-442, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38366038

RESUMEN

Ferroptosis is a non-apoptotic cell death mechanism characterized by iron-dependent membrane lipid peroxidation. Here, we review what is known about the cellular mechanisms mediating the execution and regulation of ferroptosis. We first consider how the accumulation of membrane lipid peroxides leads to the execution of ferroptosis by altering ion transport across the plasma membrane. We then discuss how metabolites and enzymes that are distributed in different compartments and organelles throughout the cell can regulate sensitivity to ferroptosis by impinging upon iron, lipid and redox metabolism. Indeed, metabolic pathways that reside in the mitochondria, endoplasmic reticulum, lipid droplets, peroxisomes and other organelles all contribute to the regulation of ferroptosis sensitivity. We note how the regulation of ferroptosis sensitivity by these different organelles and pathways seems to vary between different cells and death-inducing conditions. We also highlight transcriptional master regulators that integrate the functions of different pathways and organelles to modulate ferroptosis sensitivity globally. Throughout this Review, we highlight open questions and areas in which progress is needed to better understand the cell biology of ferroptosis.


Asunto(s)
Ferroptosis , Hierro , Peroxidación de Lípido , Ferroptosis/fisiología , Humanos , Animales , Hierro/metabolismo , Mitocondrias/metabolismo , Metabolismo de los Lípidos , Membrana Celular/metabolismo , Oxidación-Reducción
6.
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
7.
Cell ; 184(4): 856-858, 2021 02 18.
Artículo en Inglés | MEDLINE | ID: mdl-33606984

RESUMEN

In this issue of Cell, Ma et al. reveal a mechanistic role for PIEZO1 in iron homeostasis through molecular genetic mouse studies. They also demonstrate implications for human iron overload and deficiency syndromes, susceptibility to malarial infection, and red blood cell turnover in persons of African ancestries.


Asunto(s)
Hierro , Malaria , Animales , Eritrocitos , Homeostasis , Humanos , Canales Iónicos/genética , Ratones
8.
Cell ; 184(4): 969-982.e13, 2021 02 18.
Artículo en Inglés | MEDLINE | ID: mdl-33571427

RESUMEN

Iron overload causes progressive organ damage and is associated with arthritis, liver damage, and heart failure. Elevated iron levels are present in 1%-5% of individuals; however, iron overload is undermonitored and underdiagnosed. Genetic factors affecting iron homeostasis are emerging. Individuals with hereditary xerocytosis, a rare disorder with gain-of-function (GOF) mutations in mechanosensitive PIEZO1 ion channel, develop age-onset iron overload. We show that constitutive or macrophage expression of a GOF Piezo1 allele in mice disrupts levels of the iron regulator hepcidin and causes iron overload. We further show that PIEZO1 is a key regulator of macrophage phagocytic activity and subsequent erythrocyte turnover. Strikingly, we find that E756del, a mild GOF PIEZO1 allele present in one-third of individuals of African descent, is strongly associated with increased plasma iron. Our study links macrophage mechanotransduction to iron metabolism and identifies a genetic risk factor for increased iron levels in African Americans.


Asunto(s)
Canales Iónicos/metabolismo , Hierro/metabolismo , Negro o Afroamericano , Envejecimiento/metabolismo , Alelos , Animales , Estudios de Cohortes , Recuento de Eritrocitos , Eritropoyesis , Mutación con Ganancia de Función/genética , Hepatocitos/metabolismo , Hepcidinas/sangre , Hepcidinas/metabolismo , Humanos , Hierro/sangre , Sobrecarga de Hierro/metabolismo , Macrófagos/metabolismo , Mecanotransducción Celular , Ratones Endogámicos C57BL , Fagocitosis , Fenotipo , Estrés Fisiológico
9.
Nat Immunol ; 24(10): 1671-1684, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37709985

RESUMEN

Iron metabolism is pivotal for cell fitness in the mammalian host; however, its role in group 3 innate lymphoid cells (ILC3s) is unknown. Here we show that transferrin receptor CD71 (encoded by Tfrc)-mediated iron metabolism cell-intrinsically controls ILC3 proliferation and host protection against Citrobacter rodentium infection and metabolically affects mitochondrial respiration by switching of oxidative phosphorylation toward glycolysis. Iron deprivation or Tfrc ablation in ILC3s reduces the expression and/or activity of the aryl hydrocarbon receptor (Ahr), a key ILC3 regulator. Genetic ablation or activation of Ahr in ILC3s leads to CD71 upregulation or downregulation, respectively, suggesting Ahr-mediated suppression of CD71. Mechanistically, Ahr directly binds to the Tfrc promoter to inhibit transcription. Iron overload partially restores the defective ILC3 compartment in the small intestine of Ahr-deficient mice, consistent with the compensatory upregulation of CD71. These data collectively demonstrate an under-appreciated role of the Ahr-CD71-iron axis in the regulation of ILC3 maintenance and function.


Asunto(s)
Infecciones por Enterobacteriaceae , Inmunidad Innata , Animales , Ratones , Linfocitos , Estado Nutricional , Hierro , Receptores de Transferrina/genética , Mamíferos
10.
Cell ; 180(2): 214-216, 2020 01 23.
Artículo en Inglés | MEDLINE | ID: mdl-31978339

RESUMEN

Lysosome (vacuole) and mitochondria decline interdependently during aging through an unclear mechanism. In this issue of Cell, Hughes et al. (2020) show that defective vacuole-mediated cysteine compartmentalization in aging yeast leads to iron limitation and mitochondrial dysfunction.


Asunto(s)
Cisteína , Hierro , Homeostasis , Mitocondrias
11.
Cell ; 181(5): 1188-1188.e1, 2020 05 28.
Artículo en Inglés | MEDLINE | ID: mdl-32470402

RESUMEN

Ferroptosis is a regulated form of cell death that occurs when phospholipids with polyunsaturated fatty acyl tails are oxidized in an iron-dependent manner. Research in recent years has uncovered complex cellular networks that induce and suppress lethal lipid peroxidation. This SnapShot provides an overview of ferroptosis-related pathways, including relevant biomolecules and small-molecule modulators regulating them.


Asunto(s)
Ferroptosis/genética , Ferroptosis/fisiología , Hierro/metabolismo , Muerte Celular , Humanos , Peroxidación de Lípido/fisiología , Oxidación-Reducción , Fosfolípidos/metabolismo
12.
Cell ; 183(3): 752-770.e22, 2020 10 29.
Artículo en Inglés | MEDLINE | ID: mdl-33125891

RESUMEN

A greater understanding of hematopoietic stem cell (HSC) regulation is required for dissecting protective versus detrimental immunity to pathogens that cause chronic infections such as Mycobacterium tuberculosis (Mtb). We have shown that systemic administration of Bacille Calmette-Guérin (BCG) or ß-glucan reprograms HSCs in the bone marrow (BM) via a type II interferon (IFN-II) or interleukin-1 (IL1) response, respectively, which confers protective trained immunity against Mtb. Here, we demonstrate that, unlike BCG or ß-glucan, Mtb reprograms HSCs via an IFN-I response that suppresses myelopoiesis and impairs development of protective trained immunity to Mtb. Mechanistically, IFN-I signaling dysregulates iron metabolism, depolarizes mitochondrial membrane potential, and induces cell death specifically in myeloid progenitors. Additionally, activation of the IFN-I/iron axis in HSCs impairs trained immunity to Mtb infection. These results identify an unanticipated immune evasion strategy of Mtb in the BM that controls the magnitude and intrinsic anti-microbial capacity of innate immunity to infection.


Asunto(s)
Células Madre Hematopoyéticas/microbiología , Inmunidad , Mycobacterium tuberculosis/fisiología , Mielopoyesis , Animales , Células de la Médula Ósea/metabolismo , Proliferación Celular , Susceptibilidad a Enfermedades , Homeostasis , Interferón Tipo I/metabolismo , Hierro/metabolismo , Cinética , Pulmón/microbiología , Pulmón/patología , Macrófagos/inmunología , Ratones Endogámicos C57BL , Células Mieloides/metabolismo , Necrosis , Transducción de Señal , Transcripción Genética , Tuberculosis/inmunología , Tuberculosis/microbiología , Tuberculosis/patología
13.
Cell ; 180(2): 296-310.e18, 2020 01 23.
Artículo en Inglés | MEDLINE | ID: mdl-31978346

RESUMEN

Mitochondria and lysosomes are functionally linked, and their interdependent decline is a hallmark of aging and disease. Despite the long-standing connection between these organelles, the function(s) of lysosomes required to sustain mitochondrial health remains unclear. Here, working in yeast, we show that the lysosome-like vacuole maintains mitochondrial respiration by spatially compartmentalizing amino acids. Defects in vacuole function result in a breakdown in intracellular amino acid homeostasis, which drives age-related mitochondrial decline. Among amino acids, we find that cysteine is most toxic for mitochondria and show that elevated non-vacuolar cysteine impairs mitochondrial respiration by limiting intracellular iron availability through an oxidant-based mechanism. Cysteine depletion or iron supplementation restores mitochondrial health in vacuole-impaired cells and prevents mitochondrial decline during aging. These results demonstrate that cysteine toxicity is a major driver of age-related mitochondrial deterioration and identify vacuolar amino acid compartmentation as a cellular strategy to minimize amino acid toxicity.


Asunto(s)
Cisteína/toxicidad , Hierro/metabolismo , Mitocondrias/metabolismo , Aminoácidos/metabolismo , Senescencia Celular/fisiología , Cisteína/metabolismo , Homeostasis , Lisosomas/metabolismo , Mitocondrias/fisiología , Mitofagia/fisiología , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , ATPasas de Translocación de Protón Vacuolares/metabolismo , Vacuolas/metabolismo
14.
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
15.
Annu Rev Biochem ; 87: 621-643, 2018 06 20.
Artículo en Inglés | MEDLINE | ID: mdl-29925260

RESUMEN

In response to microbial infection, the human host deploys metal-sequestering host-defense proteins, which reduce nutrient availability and thereby inhibit microbial growth and virulence. Calprotectin (CP) is an abundant antimicrobial protein released from neutrophils and epithelial cells at sites of infection. CP sequesters divalent first-row transition metal ions to limit the availability of essential metal nutrients in the extracellular space. While functional and clinical studies of CP have been pursued for decades, advances in our understanding of its biological coordination chemistry, which is central to its role in the host-microbe interaction, have been made in more recent years. In this review, we focus on the coordination chemistry of CP and highlight studies of its metal-binding properties and contributions to the metal-withholding innate immune response. Taken together, these recent studies inform our current model of how CP participates in metal homeostasis and immunity, and they provide a foundation for further investigations of a remarkable metal-chelating protein at the host-microbe interface and beyond.


Asunto(s)
Interacciones Microbiota-Huesped/inmunología , Interacciones Microbiota-Huesped/fisiología , Complejo de Antígeno L1 de Leucocito/inmunología , Complejo de Antígeno L1 de Leucocito/metabolismo , Elementos de Transición/metabolismo , Secuencia de Aminoácidos , Animales , Péptidos Catiónicos Antimicrobianos/genética , Péptidos Catiónicos Antimicrobianos/inmunología , Péptidos Catiónicos Antimicrobianos/metabolismo , Humanos , Inmunidad Innata , Hierro/inmunología , Hierro/metabolismo , Complejo de Antígeno L1 de Leucocito/genética , Manganeso/inmunología , Manganeso/metabolismo , Modelos Biológicos , Modelos Moleculares , Níquel/inmunología , Níquel/metabolismo , Conformación Proteica , Homología de Secuencia de Aminoácido , Zinc/inmunología , Zinc/metabolismo
16.
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
17.
Cell ; 175(2): 311-312, 2018 10 04.
Artículo en Inglés | MEDLINE | ID: mdl-30290138

RESUMEN

Siderophores are small molecules produced by bacteria that bind ferric iron in the surrounding environment with extraordinary affinity. A new study provides evidence that a simple animal host, Caenorhabditis elegans, co-opts siderophores to promote its own iron acquisition and growth.


Asunto(s)
Enterobactina , Sideróforos , Adenosina Trifosfato , Animales , Bacterias , Hierro
18.
Cell ; 175(1): 146-158.e15, 2018 09 20.
Artículo en Inglés | MEDLINE | ID: mdl-30100182

RESUMEN

Pathogen virulence exists on a continuum. The strategies that drive symptomatic or asymptomatic infections remain largely unknown. We took advantage of the concept of lethal dose 50 (LD50) to ask which component of individual non-genetic variation between hosts defines whether they survive or succumb to infection. Using the enteric pathogen Citrobacter, we found no difference in pathogen burdens between healthy and symptomatic populations. Iron metabolism-related genes were induced in asymptomatic hosts compared to symptomatic or naive mice. Dietary iron conferred complete protection without influencing pathogen burdens, even at 1000× the lethal dose of Citrobacter. Dietary iron induced insulin resistance, increasing glucose levels in the intestine that were necessary and sufficient to suppress pathogen virulence. A short course of dietary iron drove the selection of attenuated Citrobacter strains that can transmit and asymptomatically colonize naive hosts, demonstrating that environmental factors and cooperative metabolic strategies can drive conversion of pathogens toward commensalism.


Asunto(s)
Interacciones Huésped-Patógeno/fisiología , Hierro/metabolismo , Virulencia/fisiología , Animales , Infecciones Asintomáticas , Citrobacter rodentium/metabolismo , Citrobacter rodentium/patogenicidad , Colitis/tratamiento farmacológico , Colitis/metabolismo , Colon/microbiología , Suplementos Dietéticos , Infecciones por Enterobacteriaceae/tratamiento farmacológico , Femenino , Resistencia a la Insulina/fisiología , Intestino Delgado/microbiología , Hierro/farmacología , Dosificación Letal Mediana , Masculino , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos DBA
19.
Cell ; 175(2): 571-582.e11, 2018 10 04.
Artículo en Inglés | MEDLINE | ID: mdl-30146159

RESUMEN

Elucidating the benefits of individual microbiota-derived molecules in host animals is important for understanding the symbiosis between humans and their microbiota. The bacteria-secreted enterobactin (Ent) is an iron scavenging siderophore with presumed negative effects on hosts. However, the high prevalence of Ent-producing commensal bacteria in the human gut raises the intriguing question regarding a potential host mechanism to beneficially use Ent. We discovered an unexpected and striking role of Ent in supporting growth and the labile iron pool in C. elegans. We show that Ent promotes mitochondrial iron uptake and does so, surprisingly, by binding to the ATP synthase α subunit, which acts inside of mitochondria and independently of ATP synthase. We also demonstrated the conservation of this mechanism in mammalian cells. This study reveals a distinct paradigm for the "iron tug of war" between commensal bacteria and their hosts and an important mechanism for mitochondrial iron uptake and homeostasis.


Asunto(s)
Enterobactina/fisiología , Hierro/metabolismo , Sideróforos/fisiología , Adenosina Trifosfato/metabolismo , Animales , ATPasas de Translocación de Protón Bacterianas/metabolismo , ATPasas de Translocación de Protón Bacterianas/fisiología , Transporte Biológico , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/metabolismo , Enterobactina/metabolismo , Escherichia coli/metabolismo , Proteínas de Escherichia coli/metabolismo , Proteínas de Escherichia coli/fisiología , Células HEK293 , Humanos , Hierro/fisiología , Mitocondrias/metabolismo
20.
Cell ; 175(5): 1418-1429.e9, 2018 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-30454649

RESUMEN

We report here a simple and global strategy to map out gene functions and target pathways of drugs, toxins, or other small molecules based on "homomer dynamics" protein-fragment complementation assays (hdPCA). hdPCA measures changes in self-association (homomerization) of over 3,500 yeast proteins in yeast grown under different conditions. hdPCA complements genetic interaction measurements while eliminating the confounding effects of gene ablation. We demonstrate that hdPCA accurately predicts the effects of two longevity and health span-affecting drugs, the immunosuppressant rapamycin and the type 2 diabetes drug metformin, on cellular pathways. We also discovered an unsuspected global cellular response to metformin that resembles iron deficiency and includes a change in protein-bound iron levels. This discovery opens a new avenue to investigate molecular mechanisms for the prevention or treatment of diabetes, cancers, and other chronic diseases of aging.


Asunto(s)
Hierro/metabolismo , Metaloproteínas/metabolismo , Metformina/farmacología , Saccharomyces cerevisiae/metabolismo , Sirolimus/farmacología , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Prueba de Complementación Genética , Humanos , Metaloproteínas/genética , Saccharomyces cerevisiae/genética
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda