Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.760
Filtrar
Más filtros

Tipo del documento
Intervalo de año de publicación
1.
Cell ; 187(17): 4621-4636.e18, 2024 Aug 22.
Artículo en Inglés | MEDLINE | ID: mdl-39067443

RESUMEN

Bidirectional transport in cilia is carried out by polymers of the IFTA and IFTB protein complexes, called anterograde and retrograde intraflagellar transport (IFT) trains. Anterograde trains deliver cargoes from the cell to the cilium tip, then convert into retrograde trains for cargo export. We set out to understand how the IFT complexes can perform these two directly opposing roles before and after conversion. We use cryoelectron tomography and in situ cross-linking mass spectrometry to determine the structure of retrograde IFT trains and compare it with the known structure of anterograde trains. The retrograde train is a 2-fold symmetric polymer organized around a central thread of IFTA complexes. We conclude that anterograde-to-retrograde remodeling involves global rearrangements of the IFTA/B complexes and requires complete disassembly of the anterograde train. Finally, we describe how conformational changes to cargo-binding sites facilitate unidirectional cargo transport in a bidirectional system.


Asunto(s)
Cilios , Microscopía por Crioelectrón , Flagelos , Flagelos/metabolismo , Flagelos/ultraestructura , Cilios/metabolismo , Transporte Biológico , Chlamydomonas reinhardtii/metabolismo , Modelos Moleculares , Transporte de Proteínas
2.
Cell ; 186(20): 4438-4453.e23, 2023 09 28.
Artículo en Inglés | MEDLINE | ID: mdl-37774681

RESUMEN

Cellular perturbations underlying Alzheimer's disease (AD) are primarily studied in human postmortem samples and model organisms. Here, we generated a single-nucleus atlas from a rare cohort of cortical biopsies from living individuals with varying degrees of AD pathology. We next performed a systematic cross-disease and cross-species integrative analysis to identify a set of cell states that are specific to early AD pathology. These changes-which we refer to as the early cortical amyloid response-were prominent in neurons, wherein we identified a transitional hyperactive state preceding the loss of excitatory neurons, which we confirmed by acute slice physiology on independent biopsy specimens. Microglia overexpressing neuroinflammatory-related processes also expanded as AD pathology increased. Finally, both oligodendrocytes and pyramidal neurons upregulated genes associated with ß-amyloid production and processing during this early hyperactive phase. Our integrative analysis provides an organizing framework for targeting circuit dysfunction, neuroinflammation, and amyloid production early in AD pathogenesis.


Asunto(s)
Enfermedad de Alzheimer , Lóbulo Frontal , Microglía , Neuronas , Humanos , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Amiloide , Péptidos beta-Amiloides/metabolismo , Microglía/patología , Neuronas/patología , Células Piramidales , Biopsia , Lóbulo Frontal/patología , Análisis de Expresión Génica de una Sola Célula , Núcleo Celular/metabolismo , Núcleo Celular/patología
3.
Nat Immunol ; 24(8): 1382-1390, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37500887

RESUMEN

Microglia, the macrophages of the brain parenchyma, are key players in neurodegenerative diseases such as Alzheimer's disease. These cells adopt distinct transcriptional subtypes known as states. Understanding state function, especially in human microglia, has been elusive owing to a lack of tools to model and manipulate these cells. Here, we developed a platform for modeling human microglia transcriptional states in vitro. We found that exposure of human stem-cell-differentiated microglia to synaptosomes, myelin debris, apoptotic neurons or synthetic amyloid-beta fibrils generated transcriptional diversity that mapped to gene signatures identified in human brain microglia, including disease-associated microglia, a state enriched in neurodegenerative diseases. Using a new lentiviral approach, we demonstrated that the transcription factor MITF drives a disease-associated transcriptional signature and a highly phagocytic state. Together, these tools enable the manipulation and functional interrogation of human microglial states in both homeostatic and disease-relevant contexts.


Asunto(s)
Enfermedad de Alzheimer , Células Madre Pluripotentes Inducidas , Enfermedades Neurodegenerativas , Humanos , Microglía , Enfermedad de Alzheimer/genética , Encéfalo
4.
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
5.
Cell ; 169(7): 1303-1314.e18, 2017 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-28602352

RESUMEN

Cytoplasmic dynein-1 binds dynactin and cargo adaptor proteins to form a transport machine capable of long-distance processive movement along microtubules. However, it is unclear why dynein-1 moves poorly on its own or how it is activated by dynactin. Here, we present a cryoelectron microscopy structure of the complete 1.4-megadalton human dynein-1 complex in an inhibited state known as the phi-particle. We reveal the 3D structure of the cargo binding dynein tail and show how self-dimerization of the motor domains locks them in a conformation with low microtubule affinity. Disrupting motor dimerization with structure-based mutagenesis drives dynein-1 into an open form with higher affinity for both microtubules and dynactin. We find the open form is also inhibited for movement and that dynactin relieves this by reorienting the motor domains to interact correctly with microtubules. Our model explains how dynactin binding to the dynein-1 tail directly stimulates its motor activity.


Asunto(s)
Dineínas Citoplasmáticas/química , Complejos Multiproteicos/química , Animales , Microscopía por Crioelectrón , Dineínas Citoplasmáticas/metabolismo , Dineínas Citoplasmáticas/ultraestructura , Dimerización , Complejo Dinactina/química , Complejo Dinactina/metabolismo , Humanos , Ratones , Microtúbulos/química , Microtúbulos/metabolismo , Modelos Moleculares , Proteínas Motoras Moleculares/química , Proteínas Motoras Moleculares/metabolismo , Complejos Multiproteicos/metabolismo , Complejos Multiproteicos/ultraestructura , Células Sf9 , Spodoptera , Porcinos
6.
Mol Cell ; 84(4): 640-658.e10, 2024 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-38266639

RESUMEN

The Bloom syndrome helicase BLM interacts with topoisomerase IIIα (TOP3A), RMI1, and RMI2 to form the BTR complex, which dissolves double Holliday junctions and DNA replication intermediates to promote sister chromatid disjunction before cell division. In its absence, structure-specific nucleases like the SMX complex (comprising SLX1-SLX4, MUS81-EME1, and XPF-ERCC1) can cleave joint DNA molecules instead, but cells deficient in both BTR and SMX are not viable. Here, we identify a negative genetic interaction between BLM loss and deficiency in the BRCA1-BARD1 tumor suppressor complex. We show that this is due to a previously overlooked role for BARD1 in recruiting SLX4 to resolve DNA intermediates left unprocessed by BLM in the preceding interphase. Consequently, cells with defective BLM and BRCA1-BARD1 accumulate catastrophic levels of chromosome breakage and micronucleation, leading to cell death. Thus, we reveal mechanistic insights into SLX4 recruitment to DNA lesions, with potential clinical implications for treating BRCA1-deficient tumors.


Asunto(s)
Proteínas de Unión al ADN , Recombinasas , Humanos , ADN/genética , Reparación del ADN , Replicación del ADN , ADN Cruciforme , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Recombinasas/genética , RecQ Helicasas/genética , RecQ Helicasas/metabolismo
7.
Mol Cell ; 84(11): 2087-2103.e8, 2024 Jun 06.
Artículo en Inglés | MEDLINE | ID: mdl-38815579

RESUMEN

RNA splicing is pivotal in post-transcriptional gene regulation, yet the exponential expansion of intron length in humans poses a challenge for accurate splicing. Here, we identify hnRNPM as an essential RNA-binding protein that suppresses cryptic splicing through binding to deep introns, maintaining human transcriptome integrity. Long interspersed nuclear elements (LINEs) in introns harbor numerous pseudo splice sites. hnRNPM preferentially binds at intronic LINEs to repress pseudo splice site usage for cryptic splicing. Remarkably, cryptic exons can generate long dsRNAs through base-pairing of inverted ALU transposable elements interspersed among LINEs and consequently trigger an interferon response, a well-known antiviral defense mechanism. Significantly, hnRNPM-deficient tumors show upregulated interferon-associated pathways and elevated immune cell infiltration. These findings unveil hnRNPM as a guardian of transcriptome integrity by repressing cryptic splicing and suggest that targeting hnRNPM in tumors may be used to trigger an inflammatory immune response, thereby boosting cancer surveillance.


Asunto(s)
Ribonucleoproteína Heterogénea-Nuclear Grupo M , Intrones , Elementos de Nucleótido Esparcido Largo , Empalme del ARN , ARN Bicatenario , Humanos , Ribonucleoproteína Heterogénea-Nuclear Grupo M/genética , Ribonucleoproteína Heterogénea-Nuclear Grupo M/metabolismo , ARN Bicatenario/genética , ARN Bicatenario/metabolismo , Elementos de Nucleótido Esparcido Largo/genética , Interferones/metabolismo , Interferones/genética , Animales , Células HEK293 , Ratones , Transcriptoma , Exones , Sitios de Empalme de ARN , Elementos Alu/genética
8.
Immunity ; 54(1): 1-3, 2021 01 12.
Artículo en Inglés | MEDLINE | ID: mdl-33440134

RESUMEN

Antibiotics improve clinical outcomes independent of their antibacterial effects. In this issue of Immunity, Almeida et al. and Colaço et al. demonstrate that antibiotic impairment of mitochondrial ribosomes modulates both T-cell-dependent inflammation and host tolerance to infection.


Asunto(s)
Autoinmunidad , Linfocitos T , Bacterias
9.
Mol Cell ; 82(7): 1261-1277.e9, 2022 04 07.
Artículo en Inglés | MEDLINE | ID: mdl-35305311

RESUMEN

The product of hexokinase (HK) enzymes, glucose-6-phosphate, can be metabolized through glycolysis or directed to alternative metabolic routes, such as the pentose phosphate pathway (PPP) to generate anabolic intermediates. HK1 contains an N-terminal mitochondrial binding domain (MBD), but its physiologic significance remains unclear. To elucidate the effect of HK1 mitochondrial dissociation on cellular metabolism, we generated mice lacking the HK1 MBD (ΔE1HK1). These mice produced a hyper-inflammatory response when challenged with lipopolysaccharide. Additionally, there was decreased glucose flux below the level of GAPDH and increased upstream flux through the PPP. The glycolytic block below GAPDH is mediated by the binding of cytosolic HK1 with S100A8/A9, resulting in GAPDH nitrosylation through iNOS. Additionally, human and mouse macrophages from conditions of low-grade inflammation, such as aging and diabetes, displayed increased cytosolic HK1 and reduced GAPDH activity. Our data indicate that HK1 mitochondrial binding alters glucose metabolism through regulation of GAPDH.


Asunto(s)
Glucosa , Hexoquinasa/metabolismo , Animales , Glucosa/metabolismo , Glucólisis , Hexoquinasa/genética , Ratones , Mitocondrias/metabolismo , Vía de Pentosa Fosfato
10.
Mol Cell ; 82(18): 3382-3397.e7, 2022 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-36002001

RESUMEN

Aberrant replication causes cells lacking BRCA2 to enter mitosis with under-replicated DNA, which activates a repair mechanism known as mitotic DNA synthesis (MiDAS). Here, we identify genome-wide the sites where MiDAS reactions occur when BRCA2 is abrogated. High-resolution profiling revealed that these sites are different from MiDAS at aphidicolin-induced common fragile sites in that they map to genomic regions replicating in the early S-phase, which are close to early-firing replication origins, are highly transcribed, and display R-loop-forming potential. Both transcription inhibition in early S-phase and RNaseH1 overexpression reduced MiDAS in BRCA2-deficient cells, indicating that transcription-replication conflicts (TRCs) and R-loops are the source of MiDAS. Importantly, the MiDAS sites identified in BRCA2-deficient cells also represent hotspots for genomic rearrangements in BRCA2-mutated breast tumors. Thus, our work provides a mechanism for how tumor-predisposing BRCA2 inactivation links transcription-induced DNA damage with mitotic DNA repair to fuel the genomic instability characteristic of cancer cells.


Asunto(s)
Replicación del ADN , Mitosis , Afidicolina/farmacología , Proteína BRCA2/genética , Sitios Frágiles del Cromosoma/genética , ADN/genética , Daño del ADN , Inestabilidad Genómica , Humanos , Mitosis/genética
11.
Mol Cell ; 81(24): 5052-5065.e6, 2021 12 16.
Artículo en Inglés | MEDLINE | ID: mdl-34847358

RESUMEN

Accumulation of unfolded or misfolded proteins in the endoplasmic reticulum (ER) lumen triggers an unfolded protein response (UPR) for stress adaptation, the failure of which induces cell apoptosis and tissue/organ damage. The molecular switches underlying how the UPR selects for stress adaptation over apoptosis remain unknown. Here, we discovered that accumulation of unfolded/misfolded proteins selectively induces N6-adenosine-methyltransferase-14 (METTL14) expression. METTL14 promotes C/EBP-homologous protein (CHOP) mRNA decay through its 3' UTR N6-methyladenosine (m6A) to inhibit its downstream pro-apoptotic target gene expression. UPR induces METTL14 expression by competing against the HRD1-ER-associated degradation (ERAD) machinery to block METTL14 ubiquitination and degradation. Therefore, mice with liver-specific METTL14 deletion are highly susceptible to both acute pharmacological and alpha-1 antitrypsin (AAT) deficiency-induced ER proteotoxic stress and liver injury. Further hepatic CHOP deletion protects METTL14 knockout mice from ER-stress-induced liver damage. Our study reveals a crosstalk between ER stress and mRNA m6A modification pathways, termed the ERm6A pathway, for ER stress adaptation to proteotoxicity.


Asunto(s)
Adenina/análogos & derivados , Estrés del Retículo Endoplásmico , Degradación Asociada con el Retículo Endoplásmico , Retículo Endoplásmico/enzimología , Hepatopatías/enzimología , Hígado/enzimología , Metiltransferasas/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Adenina/metabolismo , Animales , Apoptosis , Modelos Animales de Enfermedad , Retículo Endoplásmico/metabolismo , Retículo Endoplásmico/patología , Células HEK293 , Células Hep G2 , Humanos , Hígado/patología , Hepatopatías/etiología , Hepatopatías/genética , Hepatopatías/patología , Metiltransferasas/genética , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Ratones Noqueados , Ratones SCID , Células 3T3 NIH , Proteolisis , Factor de Transcripción CHOP/genética , Factor de Transcripción CHOP/metabolismo , Ubiquitina-Proteína Ligasas/genética , Ubiquitinación , alfa 1-Antitripsina/genética , alfa 1-Antitripsina/metabolismo , Deficiencia de alfa 1-Antitripsina/complicaciones , Deficiencia de alfa 1-Antitripsina/enzimología , Deficiencia de alfa 1-Antitripsina/genética
12.
Trends Biochem Sci ; 49(5): 387-390, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38368181

RESUMEN

Poly(UG) or 'pUG' dinucleotide repeats direct gene silencing in Caenorhabditis elegans by adopting an unusual quadruplex structure. Humans have thousands of pUG sequences and proteins that interact with them. The pUG fold reveals new aspects of gene regulation and RNA folding, highlighting how a simple sequence can encode a complex structure.


Asunto(s)
Caenorhabditis elegans , G-Cuádruplex , Silenciador del Gen , Animales , Caenorhabditis elegans/genética , Caenorhabditis elegans/metabolismo , Humanos , ARN/metabolismo , ARN/genética , ARN/química
13.
Immunity ; 50(4): 955-974, 2019 04 16.
Artículo en Inglés | MEDLINE | ID: mdl-30995509

RESUMEN

Neurodegenerative diseases of the central nervous system progressively rob patients of their memory, motor function, and ability to perform daily tasks. Advances in genetics and animal models are beginning to unearth an unexpected role of the immune system in disease onset and pathogenesis; however, the role of cytokines, growth factors, and other immune signaling pathways in disease pathogenesis is still being examined. Here we review recent genetic risk and genome-wide association studies and emerging mechanisms for three key immune pathways implicated in disease, the growth factor TGF-ß, the complement cascade, and the extracellular receptor TREM2. These immune signaling pathways are important under both healthy and neurodegenerative conditions, and recent work has highlighted new functional aspects of their signaling. Finally, we assess future directions for immune-related research in neurodegeneration and potential avenues for immune-related therapies.


Asunto(s)
Enfermedades Neurodegenerativas/inmunología , Transducción de Señal/inmunología , Envejecimiento/inmunología , Animales , Activación de Complemento , Progresión de la Enfermedad , Predisposición Genética a la Enfermedad , Estudio de Asociación del Genoma Completo , Gliosis/inmunología , Gliosis/patología , Humanos , Inmunidad Innata , Inflamación/inmunología , Glicoproteínas de Membrana/inmunología , Ratones , Ratones Noqueados , Ratones Transgénicos , Microglía/inmunología , Modelos Inmunológicos , Enfermedades Neurodegenerativas/genética , Enfermedades Neurodegenerativas/patología , Enfermedades Neurodegenerativas/terapia , Agregación Patológica de Proteínas/inmunología , Receptores Inmunológicos/inmunología , Factor de Crecimiento Transformador beta/inmunología
14.
Immunity ; 50(1): 253-271.e6, 2019 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-30471926

RESUMEN

Microglia, the resident immune cells of the brain, rapidly change states in response to their environment, but we lack molecular and functional signatures of different microglial populations. Here, we analyzed the RNA expression patterns of more than 76,000 individual microglia in mice during development, in old age, and after brain injury. Our analysis uncovered at least nine transcriptionally distinct microglial states, which expressed unique sets of genes and were localized in the brain using specific markers. The greatest microglial heterogeneity was found at young ages; however, several states-including chemokine-enriched inflammatory microglia-persisted throughout the lifespan or increased in the aged brain. Multiple reactive microglial subtypes were also found following demyelinating injury in mice, at least one of which was also found in human multiple sclerosis lesions. These distinct microglia signatures can be used to better understand microglia function and to identify and manipulate specific subpopulations in health and disease.


Asunto(s)
Envejecimiento/inmunología , Lesiones Encefálicas/inmunología , Encéfalo/fisiología , Microglía/fisiología , Esclerosis Múltiple/inmunología , Adaptación Fisiológica , Envejecimiento/genética , Animales , Lesiones Encefálicas/genética , Diferenciación Celular , Enfermedades Desmielinizantes , Humanos , Longevidad , Ratones , Ratones Endogámicos C57BL , Análisis de Secuencia de ARN , Análisis de la Célula Individual
15.
Trends Biochem Sci ; 48(9): 815-825, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37433704

RESUMEN

Metal micronutrients are essential for life and exist in a delicate balance to maintain an organism's health. The labile nature of metal-biomolecule interactions clouds the understanding of metal binders and metal-mediated conformational changes that are influential to health and disease. Mass spectrometry (MS)-based methods and technologies have been developed to better understand metal micronutrient dynamics in the intra- and extracellular environment. In this review, we describe the challenges associated with studying labile metals in human biology and highlight MS-based methods for the discovery and study of metal-biomolecule interactions.


Asunto(s)
Metales , Humanos , Metales/química , Espectrometría de Masas/métodos
16.
Mol Cell ; 74(3): 571-583.e8, 2019 05 02.
Artículo en Inglés | MEDLINE | ID: mdl-30898438

RESUMEN

In mitosis, cells inactivate DNA double-strand break (DSB) repair pathways to preserve genome stability. However, some early signaling events still occur, such as recruitment of the scaffold protein MDC1 to phosphorylated histone H2AX at DSBs. Yet, it remains unclear whether these events are important for maintaining genome stability during mitosis. Here, we identify a highly conserved protein-interaction surface in MDC1 that is phosphorylated by CK2 and recognized by the DNA-damage response mediator protein TOPBP1. Disruption of MDC1-TOPBP1 binding causes a specific loss of TOPBP1 recruitment to DSBs in mitotic but not interphase cells, accompanied by mitotic radiosensitivity, increased micronuclei, and chromosomal instability. Mechanistically, we find that TOPBP1 forms filamentous structures capable of bridging MDC1 foci in mitosis, indicating that MDC1-TOPBP1 complexes tether DSBs until repair is reactivated in the following G1 phase. Thus, we reveal an important, hitherto-unnoticed cooperation between MDC1 and TOPBP1 in maintaining genome stability during cell division.


Asunto(s)
Proteínas Portadoras/genética , Inestabilidad Cromosómica/genética , Proteínas de Unión al ADN/genética , Mitosis/genética , Proteínas Nucleares/genética , Transactivadores/genética , Proteínas Adaptadoras Transductoras de Señales , Proteínas de Ciclo Celular , Roturas del ADN de Doble Cadena , Daño del ADN/genética , Reparación del ADN/genética , Fase G1/genética , Genoma Humano/genética , Inestabilidad Genómica/genética , Histonas , Humanos , Fosforilación , Transducción de Señal/genética
17.
Proc Natl Acad Sci U S A ; 121(19): e2321216121, 2024 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-38687796

RESUMEN

Cells must replicate their genome quickly and accurately, and they require metabolites and cofactors to do so. Ionic zinc (Zn2+) is an essential micronutrient that is required for hundreds of cellular processes, including DNA synthesis and adequate proliferation. Deficiency in this micronutrient impairs DNA synthesis and inhibits proliferation, but the mechanism is unknown. Using fluorescent reporters to track single cells via long-term live-cell imaging, we find that Zn2+ is required at the G1/S transition and during S phase for timely completion of S phase. A short pulse of Zn2+ deficiency impairs DNA synthesis and increases markers of replication stress. These markers of replication stress are reversed upon resupply of Zn2+. Finally, we find that if Zn2+ is chelated during the mother cell's S phase, daughter cells enter a transient quiescent state, maintained by sustained expression of p21, which disappears upon reentry into the cell cycle. In summary, short pulses of mild Zn2+ deficiency in S phase specifically induce replication stress, which causes downstream proliferation impairments in daughter cells.


Asunto(s)
Proliferación Celular , Replicación del ADN , Fase S , Zinc , Zinc/metabolismo , Zinc/deficiencia , Humanos
18.
Development ; 150(22)2023 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-37818607

RESUMEN

The intricate vascular system of the kidneys supports body fluid and organ homeostasis. However, little is known about how vascular architecture is established during kidney development. More specifically, how signals from the kidney influence vessel maturity and patterning remains poorly understood. Netrin 1 (Ntn1) is a secreted ligand that is crucial for vessel and neuronal guidance. Here, we demonstrate that Ntn1 is expressed by Foxd1+ stromal progenitors in the developing mouse kidney and conditional deletion (Foxd1GC/+;Ntn1fl/fl) results in hypoplastic kidneys with extended nephrogenesis. Wholemount 3D analyses additionally revealed the loss of a predictable vascular pattern in Foxd1GC/+;Ntn1fl/fl kidneys. As vascular patterning has been linked to vessel maturity, we investigated arterialization. Quantification of the CD31+ endothelium at E15.5 revealed no differences in metrics such as the number of branches or branch points, whereas the arterial vascular smooth muscle metrics were significantly reduced at both E15.5 and P0. In support of our observed phenotypes, whole kidney RNA-seq revealed disruptions to genes and programs associated with stromal cells, vasculature and differentiating nephrons. Together, our findings highlight the significance of Ntn1 to proper vascularization and kidney development.


Asunto(s)
Riñón , Nefronas , Animales , Ratones , Netrina-1/genética , Fenotipo
19.
PLoS Pathog ; 20(8): e1012422, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-39207957

RESUMEN

Vancomycin has proven remarkably durable to resistance evolution by Staphylococcus aureus despite widespread treatment with vancomycin in the clinic. Only 16 cases of vancomycin-resistant S. aureus (VRSA) have been documented in the United States. It is thought that the failure of VRSA to spread is partly due to the fitness cost imposed by the vanA operon, which is the only known means of high-level resistance. Here, we show that the fitness cost of vanA-mediated resistance can be overcome through laboratory evolution of VRSA in the presence of vancomycin. Adaptation to vancomycin imposed a tradeoff such that fitness in the presence of vancomycin increased, while fitness in its absence decreased in evolved lineages. Comparing the genomes of vancomycin-exposed and vancomycin-unexposed lineages pinpointed the D-alanine:D-alanine ligase gene (ddl) as the target of loss-of-function mutations, which were associated with the observed fitness tradeoff. Vancomycin-exposed lineages exhibited vancomycin dependence and abnormal colony morphology in the absence of drug, which were associated with mutations in ddl. However, further evolution of vancomycin-exposed lineages in the absence of vancomycin enabled some evolved lineages to escape this fitness tradeoff. Many vancomycin-exposed lineages maintained resistance in the absence of vancomycin, unlike their ancestral VRSA strains. These results indicate that VRSA might be able to compensate for the fitness deficit associated with vanA-mediated resistance, which may pose a threat to the prolonged durability of vancomycin in the clinic. Our results also suggest vancomycin treatment should be immediately discontinued in patients after VRSA is identified to mitigate potential adaptations.


Asunto(s)
Antibacterianos , Infecciones Estafilocócicas , Staphylococcus aureus Resistente a Vancomicina , Vancomicina , Vancomicina/farmacología , Antibacterianos/farmacología , Humanos , Infecciones Estafilocócicas/tratamiento farmacológico , Infecciones Estafilocócicas/microbiología , Staphylococcus aureus Resistente a Vancomicina/genética , Resistencia a la Vancomicina/genética , Pruebas de Sensibilidad Microbiana , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Mutación
20.
Nature ; 585(7824): 288-292, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32641834

RESUMEN

The mitochondrial electron transport chain (ETC) is necessary for tumour growth1-6 and its inhibition has demonstrated anti-tumour efficacy in combination with targeted therapies7-9. Furthermore, human brain and lung tumours display robust glucose oxidation by mitochondria10,11. However, it is unclear why a functional ETC is necessary for tumour growth in vivo. ETC function is coupled to the generation of ATP-that is, oxidative phosphorylation and the production of metabolites by the tricarboxylic acid (TCA) cycle. Mitochondrial complexes I and II donate electrons to ubiquinone, resulting in the generation of ubiquinol and the regeneration of the NAD+ and FAD cofactors, and complex III oxidizes ubiquinol back to ubiquinone, which also serves as an electron acceptor for dihydroorotate dehydrogenase (DHODH)-an enzyme necessary for de novo pyrimidine synthesis. Here we show impaired tumour growth in cancer cells that lack mitochondrial complex III. This phenotype was rescued by ectopic expression of Ciona intestinalis alternative oxidase (AOX)12, which also oxidizes ubiquinol to ubiquinone. Loss of mitochondrial complex I, II or DHODH diminished the tumour growth of AOX-expressing cancer cells deficient in mitochondrial complex III, which highlights the necessity of ubiquinone as an electron acceptor for tumour growth. Cancer cells that lack mitochondrial complex III but can regenerate NAD+ by expression of the NADH oxidase from Lactobacillus brevis (LbNOX)13 targeted to the mitochondria or cytosol were still unable to grow tumours. This suggests that regeneration of NAD+ is not sufficient to drive tumour growth in vivo. Collectively, our findings indicate that tumour growth requires the ETC to oxidize ubiquinol, which is essential to drive the oxidative TCA cycle and DHODH activity.


Asunto(s)
Mitocondrias/metabolismo , Neoplasias/metabolismo , Neoplasias/patología , Ubiquinona/análogos & derivados , Animales , Línea Celular Tumoral , Proliferación Celular , Ciona intestinalis/enzimología , Ciclo del Ácido Cítrico , Citosol/metabolismo , Dihidroorotato Deshidrogenasa , Transporte de Electrón , Complejo I de Transporte de Electrón/metabolismo , Complejo II de Transporte de Electrones/metabolismo , Complejo III de Transporte de Electrones/deficiencia , Complejo III de Transporte de Electrones/metabolismo , Humanos , Levilactobacillus brevis/enzimología , Masculino , Ratones , Mitocondrias/enzimología , Proteínas Mitocondriales/genética , Proteínas Mitocondriales/metabolismo , Complejos Multienzimáticos/genética , Complejos Multienzimáticos/metabolismo , NAD/metabolismo , NADH NADPH Oxidorreductasas/genética , NADH NADPH Oxidorreductasas/metabolismo , Neoplasias/enzimología , Fosforilación Oxidativa , Oxidorreductasas/genética , Oxidorreductasas/metabolismo , Oxidorreductasas actuantes sobre Donantes de Grupo CH-CH/metabolismo , Proteínas de Plantas/genética , Proteínas de Plantas/metabolismo , Ubiquinona/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA