Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 180
Filtrer
1.
J Clin Invest ; 2024 Aug 06.
Article de Anglais | MEDLINE | ID: mdl-39106106

RÉSUMÉ

The study of transcription factors that determine specialised neuronal functions has provided invaluable insights into the physiology of the nervous system. Peripheral chemoreceptors are neurone-like electro-physiologically excitable cells that link the oxygen content of arterial blood to the neuronal control of breathing. In the adult, this oxygen chemosensitivity is exemplified by the Type I cells of the carotid body and recent work has revealed one isoform of the transcription factor HIF, HIF-2α, to have a non-redundant role in the development and function of that organ. Here we show that the activation of HIF-2α, including isolated overexpression alone, is sufficient to induce oxygen chemosensitivity in the otherwise unresponsive adult adrenal medulla. This phenotypic change in the adrenal medulla was associated with retention of extra-adrenal paraganglioma-like tissues that resemble the foetal organ of Zuckerkandl and also manifest oxygen chemosensitivity. Acquisition of chemosensitivity was associated with changes in the adrenal medullary expression of classes of genes that are ordinarily characteristic of the carotid body, including G-protein regulators and atypical subunits of mitochondrial cytochrome oxidase. Overall, the findings suggest that, at least in certain tissues, HIF-2α acts as a phenotypic driver for cells that display oxygen chemosensitivity, thus linking two major oxygen sensing systems.

2.
Nat Commun ; 15(1): 5360, 2024 Jun 25.
Article de Anglais | MEDLINE | ID: mdl-38918375

RÉSUMÉ

Oxygen homeostasis is maintained in plants and animals by O2-sensing enzymes initiating adaptive responses to low O2 (hypoxia). Recently, the O2-sensitive enzyme ADO was shown to initiate degradation of target proteins RGS4/5 and IL32 via the Cysteine/Arginine N-degron pathway. ADO functions by catalysing oxidation of N-terminal cysteine residues, but despite multiple proteins in the human proteome having an N-terminal cysteine, other endogenous ADO substrates have not yet been identified. This could be because alternative modifications of N-terminal cysteine residues, including acetylation, prevent ADO-catalysed oxidation. Here we investigate the relationship between ADO-catalysed oxidation and NatA-catalysed acetylation of a broad range of protein sequences with N-terminal cysteines. We present evidence that human NatA catalyses N-terminal cysteine acetylation in vitro and in vivo. We then show that sequences downstream of the N-terminal cysteine dictate whether this residue is oxidised or acetylated, with ADO preferring basic and aromatic amino acids and NatA preferring acidic or polar residues. In vitro, the two modifications appear to be mutually exclusive, suggesting that distinct pools of N-terminal cysteine proteins may be acetylated or oxidised. These results reveal the sequence determinants that contribute to N-terminal cysteine protein modifications, with implications for O2-dependent protein stability and the hypoxic response.


Sujet(s)
Cystéine , Oxydoréduction , Stabilité protéique , Cystéine/métabolisme , Cystéine/composition chimique , Acétylation , Humains , Oxygène/métabolisme , Oxygène/composition chimique , Maturation post-traductionnelle des protéines , Séquence d'acides aminés , Cellules HEK293
3.
Cancer Res ; 84(11): 1799-1816, 2024 Jun 04.
Article de Anglais | MEDLINE | ID: mdl-38502859

RÉSUMÉ

Defining the initial events in oncogenesis and the cellular responses they entrain, even in advance of morphologic abnormality, is a fundamental challenge in understanding cancer initiation. As a paradigm to address this, we longitudinally studied the changes induced by loss of the tumor suppressor gene von Hippel Lindau (VHL), which ultimately drives clear cell renal cell carcinoma. Vhl inactivation was directly coupled to expression of a tdTomato reporter within a single allele, allowing accurate visualization of affected cells in their native context and retrieval from the kidney for single-cell RNA sequencing. This strategy uncovered cell type-specific responses to Vhl inactivation, defined a proximal tubular cell class with oncogenic potential, and revealed longer term adaptive changes in the renal epithelium and the interstitium. Oncogenic cell tagging also revealed markedly heterogeneous cellular effects including time-limited proliferation and elimination of specific cell types. Overall, this study reports an experimental strategy for understanding oncogenic processes in which cells bearing genetic alterations can be generated in their native context, marked, and analyzed over time. The observed effects of loss of Vhl in kidney cells provide insights into VHL tumor suppressor action and development of renal cell carcinoma. SIGNIFICANCE: Single-cell analysis of heterogeneous and dynamic responses to Vhl inactivation in the kidney suggests that early events shape the cell type specificity of oncogenesis, providing a focus for mechanistic understanding and therapeutic targeting.


Sujet(s)
Néphrocarcinome , Tumeurs du rein , Analyse sur cellule unique , Protéine Von Hippel-Lindau supresseur de tumeur , Protéine Von Hippel-Lindau supresseur de tumeur/génétique , Protéine Von Hippel-Lindau supresseur de tumeur/métabolisme , Tumeurs du rein/génétique , Tumeurs du rein/anatomopathologie , Tumeurs du rein/métabolisme , Néphrocarcinome/génétique , Néphrocarcinome/anatomopathologie , Néphrocarcinome/métabolisme , Analyse sur cellule unique/méthodes , Animaux , Souris , Transcriptome , Humains , Rein/anatomopathologie , Rein/métabolisme , Carcinogenèse/génétique , Prolifération cellulaire/génétique
4.
Proc Natl Acad Sci U S A ; 121(10): e2309957121, 2024 Mar 05.
Article de Anglais | MEDLINE | ID: mdl-38422022

RÉSUMÉ

Hypoxia signaling influences tumor development through both cell-intrinsic and -extrinsic pathways. Inhibiting hypoxia-inducible factor (HIF) function has recently been approved as a cancer treatment strategy. Hence, it is important to understand how regulators of HIF may affect tumor growth under physiological conditions. Here we report that in aging mice factor-inhibiting HIF (FIH), one of the most studied negative regulators of HIF, is a haploinsufficient suppressor of spontaneous B cell lymphomas, particular pulmonary B cell lymphomas. FIH deficiency alters immune composition in aged mice and creates a tumor-supportive immune environment demonstrated in syngeneic mouse tumor models. Mechanistically, FIH-defective myeloid cells acquire tumor-supportive properties in response to signals secreted by cancer cells or produced in the tumor microenvironment with enhanced arginase expression and cytokine-directed migration. Together, these data demonstrate that under physiological conditions, FIH plays a key role in maintaining immune homeostasis and can suppress tumorigenesis through a cell-extrinsic pathway.


Sujet(s)
Lymphome B , Protéines de répression , Animaux , Souris , Hypoxie/métabolisme , Mixed function oxygenases/métabolisme , Protéines de répression/métabolisme , Microenvironnement tumoral
5.
J Biol Chem ; 299(9): 105156, 2023 09.
Article de Anglais | MEDLINE | ID: mdl-37572852

RÉSUMÉ

In animals, adaptation to changes in cellular oxygen levels is coordinated largely by 2-oxoglutarate-dependent prolyl-hydroxylase domain (PHD) dioxygenase family members, which regulate the stability of their hypoxia-inducible factor (HIF) substrates to promote expression of genes that adapt cells to hypoxia. Recently, 2-aminoethanethiol dioxygenase (ADO) was identified as a novel O2-sensing enzyme in animals. Through N-terminal cysteine dioxygenation and the N-degron pathway, ADO regulates the stability of a set of non-transcription factor substrates; the regulators of G-protein signaling 4, 5. and 16 and interleukin-32. Here, we set out to compare and contrast the in cellulo characteristics of ADO and PHD enzymes in an attempt to better understand their co-evolution in animals. We find that ADO operates to regulate the stability of its substrates rapidly and with similar O2-sensitivity to the PHD/HIF pathway. ADO appeared less sensitive to iron chelating agents or transition metal exposure than the PHD enzymes, possibly due to tighter catalytic-site Fe2+ coordination. Unlike the PHD/HIF pathway, the ADO/N-degron pathway was not subject to feedback by hypoxic induction of ADO, and induction of ADO substrates was well sustained in response to prolonged hypoxia. The data also reveal strong interactions between proteolytic regulation of targets by ADO and transcriptional induction of those targets, that shape integrated cellular responses to hypoxia. Collectively, our comparative analysis provides further insight into ADO/N-degron-mediated oxygen sensing and its integration into established mechanisms of oxygen homeostasis.


Sujet(s)
Cystéine , Oxygène , Animaux , Cystéine/métabolisme , Hydroxylation , Hypoxie , Sous-unité alpha du facteur-1 induit par l'hypoxie/génétique , Sous-unité alpha du facteur-1 induit par l'hypoxie/métabolisme , Mammifères/métabolisme , Oxygène/métabolisme , Procollagen-Proline Dioxygenase/métabolisme , Transduction du signal
6.
Life Sci Alliance ; 6(4)2023 04.
Article de Anglais | MEDLINE | ID: mdl-36725335

RÉSUMÉ

Hypoxia-inducible factor (HIF) and aryl hydrocarbon receptor (AHR) are members of the bHLH-PAS family of transcription factors that underpin cellular responses to oxygen and to endogenous and exogenous ligands, respectively, and have central roles in the pathogenesis of renal cancer. Composed of heterodimers, they share a common HIF-1ß/ARNT subunit and similar DNA-binding motifs, raising the possibility of crosstalk between the two transcriptional pathways. Here, we identify both general and locus-specific mechanisms of interaction between HIF and AHR that act both antagonistically and cooperatively. Specifically, we observe competition for the common HIF-1ß/ARNT subunit, in cis synergy for chromatin binding, and overlap in their transcriptional targets. Recently, both HIF and AHR inhibitors have been developed for the treatment of solid tumours. However, inhibition of one pathway may promote the oncogenic effects of the other. Therefore, our work raises important questions as to whether combination therapy targeting both of these pro-tumourigenic pathways might show greater efficacy than targeting each system independently.


Sujet(s)
Néphrocarcinome , Tumeurs du rein , Humains , Récepteurs à hydrocarbure aromatique/génétique , Récepteurs à hydrocarbure aromatique/métabolisme , Hypoxie cellulaire/physiologie , Néphrocarcinome/génétique , Tumeurs du rein/génétique , Rein/métabolisme
7.
Cell Rep ; 41(7): 111652, 2022 11 15.
Article de Anglais | MEDLINE | ID: mdl-36384128

RÉSUMÉ

Activation of cellular hypoxia pathways, orchestrated by HIF (hypoxia-inducible factor) transcription factors, is a common feature of multiple tumor types, resulting from microenvironment factors and oncogenic mutation. Although they help drive many of the "hallmarks" of cancer and are associated with poor outcome and resistance to therapy, the transcriptional targets of HIF vary considerably depending on the cell type. By integrating 72 genome-wide assays of HIF binding and transcriptional regulation from multiple cancer types, we define a consensus set of 48 HIF target genes that is highly conserved across cancer types and cell lineages. These genes provide an effective marker of HIF activation in bulk and single-cell transcriptomic analyses across a wide range of cancer types and in malignant and stromal cell types. This allows the tissue-orchestrated responses to the hypoxic tumor microenvironment and to oncogenic HIF activation to be deconvoluted at the tumor and single-cell level.


Sujet(s)
Tumeurs , Humains , Tumeurs/génétique , Facteurs de transcription/métabolisme , Microenvironnement tumoral/génétique , Hypoxie cellulaire/génétique , Hypoxie/métabolisme
8.
Elife ; 112022 09 13.
Article de Anglais | MEDLINE | ID: mdl-36098684

RÉSUMÉ

Physician-scientists have epitomized the blending of deep, rigorous impactful curiosity with broad attention to human health for centuries. While we aspire to prepare all physicians with an appreciation for these skills, those who apply them to push the understanding of the boundaries of human physiology and disease, to advance treatments, and to increase our knowledge base in the arena of human health can fulfill an essential space for our society, economies, and overall well-being. Working arm in arm with basic and translational scientists as well as expert clinicians, as peers in both groups, this career additionally serves as a bridge to facilitate the pace and direction of research that ultimately impacts health. Globally, there are remarkable similarities in challenges in this career path, and in the approaches employed to overcome them. Herein, we review how different countries train physician-scientists and suggest strategies to further bolster this career path.


Sujet(s)
Recherche biomédicale , Médecins , Recherche biomédicale/enseignement et éducation , Choix de carrière , Humains
9.
Nat Struct Mol Biol ; 29(9): 871-880, 2022 09.
Article de Anglais | MEDLINE | ID: mdl-36097292

RÉSUMÉ

Hypoxia inducible factor (HIF) and mammalian target of rapamycin (mTOR) pathways orchestrate responses to oxygen and nutrient availability. These pathways are frequently dysregulated in cancer, but their interplay is poorly understood, in part because of difficulties in simultaneous measurement of global and mRNA-specific translation. Here, we describe a workflow for measurement of ribosome load of mRNAs resolved by their transcription start sites (TSSs). Its application to kidney cancer cells reveals extensive translational reprogramming by mTOR, strongly affecting many metabolic enzymes and pathways. By contrast, global effects of HIF on translation are limited, and we do not observe reported translational activation by HIF2A. In contrast, HIF-dependent alterations in TSS usage are associated with robust changes in translational efficiency in a subset of genes. Analyses of the interplay of HIF and mTOR reveal that specific classes of HIF1A and HIF2A transcriptional target gene manifest different sensitivity to mTOR, in a manner that supports combined use of HIF2A and mTOR inhibitors in treatment of kidney cancer.


Sujet(s)
Facteurs de transcription à motif basique hélice-boucle-hélice/métabolisme , Tumeurs du rein , Sérine-thréonine kinases TOR , Humains , Tumeurs du rein/génétique , Oxygène , Isoformes de protéines , ARN messager/génétique , ARN messager/métabolisme , Ribosomes/métabolisme
10.
Proc Natl Acad Sci U S A ; 119(32): e2201483119, 2022 08 09.
Article de Anglais | MEDLINE | ID: mdl-35930668

RÉSUMÉ

The Jumonji domain-containing protein JMJD6 is a 2-oxoglutarate-dependent dioxygenase associated with a broad range of biological functions. Cellular studies have implicated the enzyme in chromatin biology, transcription, DNA repair, mRNA splicing, and cotranscriptional processing. Although not all studies agree, JMJD6 has been reported to catalyze both hydroxylation of lysine residues and demethylation of arginine residues. However, despite extensive study and indirect evidence for JMJD6 catalysis in many cellular processes, direct assignment of JMJD6 catalytic substrates has been limited. Examination of a reported site of proline hydroxylation within a lysine-rich region of the tandem bromodomain protein BRD4 led us to conclude that hydroxylation was in fact on lysine and catalyzed by JMJD6. This prompted a wider search for JMJD6-catalyzed protein modifications deploying mass spectrometric methods designed to improve the analysis of such lysine-rich regions. Using lysine derivatization with propionic anhydride to improve the analysis of tryptic peptides and nontryptic proteolysis, we report 150 sites of JMJD6-catalyzed lysine hydroxylation on 48 protein substrates, including 19 sites of hydroxylation on BRD4. Most hydroxylations were within lysine-rich regions that are predicted to be unstructured; in some, multiple modifications were observed on adjacent lysine residues. Almost all of the JMJD6 substrates defined in these studies have been associated with membraneless organelle formation. Given the reported roles of lysine-rich regions in subcellular partitioning by liquid-liquid phase separation, our findings raise the possibility that JMJD6 may play a role in regulating such processes in response to stresses, including hypoxia.


Sujet(s)
Protéines intrinsèquement désordonnées , Jumonji Domain-Containing Histone Demethylases , Protéines du cycle cellulaire/métabolisme , Humains , Hydroxylation , Protéines intrinsèquement désordonnées/métabolisme , Jumonji Domain-Containing Histone Demethylases/composition chimique , Jumonji Domain-Containing Histone Demethylases/métabolisme , Lysine/métabolisme , Domaines protéiques , Facteurs de transcription/métabolisme
12.
Nat Immunol ; 23(6): 927-939, 2022 06.
Article de Anglais | MEDLINE | ID: mdl-35624205

RÉSUMÉ

Hypoxemia is a defining feature of acute respiratory distress syndrome (ARDS), an often-fatal complication of pulmonary or systemic inflammation, yet the resulting tissue hypoxia, and its impact on immune responses, is often neglected. In the present study, we have shown that ARDS patients were hypoxemic and monocytopenic within the first 48 h of ventilation. Monocytopenia was also observed in mouse models of hypoxic acute lung injury, in which hypoxemia drove the suppression of type I interferon signaling in the bone marrow. This impaired monopoiesis resulted in reduced accumulation of monocyte-derived macrophages and enhanced neutrophil-mediated inflammation in the lung. Administration of colony-stimulating factor 1 in mice with hypoxic lung injury rescued the monocytopenia, altered the phenotype of circulating monocytes, increased monocyte-derived macrophages in the lung and limited injury. Thus, tissue hypoxia altered the dynamics of the immune response to the detriment of the host and interventions to address the aberrant response offer new therapeutic strategies for ARDS.


Sujet(s)
Lésion pulmonaire , , Animaux , Humains , Hypoxie/étiologie , Inflammation/complications , Poumon , Lésion pulmonaire/complications , Souris
13.
J Biol Chem ; 298(6): 102020, 2022 06.
Article de Anglais | MEDLINE | ID: mdl-35537551

RÉSUMÉ

The aspariginyl hydroxylase human factor inhibiting hypoxia-inducible factor (FIH) is an important regulator of the transcriptional activity of hypoxia-inducible factor. FIH also catalyzes the hydroxylation of asparaginyl and other residues in ankyrin repeat domain-containing proteins, including apoptosis stimulating of p53 protein (ASPP) family members. ASPP2 is reported to undergo a single FIH-catalyzed hydroxylation at Asn-986. We report biochemical and crystallographic evidence showing that FIH catalyzes the unprecedented post-translational hydroxylation of both asparaginyl residues in "VNVN" and related motifs of ankyrin repeat domains in ASPPs (i.e., ASPP1, ASPP2, and iASPP) and the related ASB11 and p18-INK4C proteins. Our biochemical results extend the substrate scope of FIH catalysis and may have implications for its biological roles, including in the hypoxic response and ASPP family function.


Sujet(s)
Répétition ankyrine , Mixed function oxygenases , Protéines de répression , Protéines adaptatrices de la transduction du signal , Séquence d'acides aminés , Protéines régulatrices de l'apoptose , Catalyse , Humains , Hydroxylation , Hypoxie , Mixed function oxygenases/métabolisme , Protéines de répression/métabolisme
15.
Sci Rep ; 12(1): 998, 2022 01 19.
Article de Anglais | MEDLINE | ID: mdl-35046429

RÉSUMÉ

Iron deficiency impairs skeletal muscle metabolism. The underlying mechanisms are incompletely characterised, but animal and human experiments suggest the involvement of signalling pathways co-dependent upon oxygen and iron availability, including the pathway associated with hypoxia-inducible factor (HIF). We performed a prospective, case-control, clinical physiology study to explore the effects of iron deficiency on human metabolism, using exercise as a stressor. Thirteen iron-deficient (ID) individuals and thirteen iron-replete (IR) control participants each underwent 31P-magnetic resonance spectroscopy of exercising calf muscle to investigate differences in oxidative phosphorylation, followed by whole-body cardiopulmonary exercise testing. Thereafter, individuals were given an intravenous (IV) infusion, randomised to either iron or saline, and the assessments repeated ~ 1 week later. Neither baseline iron status nor IV iron significantly influenced high-energy phosphate metabolism. During submaximal cardiopulmonary exercise, the rate of decline in blood lactate concentration was diminished in the ID group (P = 0.005). Intravenous iron corrected this abnormality. Furthermore, IV iron increased lactate threshold during maximal cardiopulmonary exercise by ~ 10%, regardless of baseline iron status. These findings demonstrate abnormal whole-body energy metabolism in iron-deficient but otherwise healthy humans. Iron deficiency promotes a more glycolytic phenotype without having a detectable effect on mitochondrial bioenergetics.


Sujet(s)
Métabolisme énergétique/physiologie , Carences en fer/métabolisme , Muscles squelettiques/métabolisme , Phosphorylation oxydative , Administration par voie intraveineuse , Adulte , Études cas-témoins , Exercice physique/physiologie , Femelle , Humains , Fer/administration et posologie , Acide lactique/sang , Mâle , Études prospectives
17.
Endocr Relat Cancer ; 28(12): 757-772, 2021 10 18.
Article de Anglais | MEDLINE | ID: mdl-34658364

RÉSUMÉ

Despite a general role for the HIF hydroxylase system in cellular oxygen sensing and tumour hypoxia, cancer-associated mutations of genes in this pathway, including PHD2, PHD1, EPAS1 (encoding HIF-2α) are highly tissue-restricted, being observed in pseudohypoxic pheochromocytoma and paraganglioma (PPGL) but rarely, if ever, in other tumours. In an effort to understand that paradox and gain insights into the pathogenesis of pseudohypoxic PPGL, we constructed mice in which the principal HIF prolyl hydroxylase, Phd2, is inactivated in the adrenal medulla using TH-restricted Cre recombinase. Investigation of these animals revealed a gene expression pattern closely mimicking that of pseudohypoxic PPGL. Spatially resolved analyses demonstrated a binary distribution of two contrasting patterns of gene expression among adrenal medullary cells. Phd2 inactivation resulted in a marked shift in this distribution towards a Pnmt-/Hif-2α+/Rgs5+ population. This was associated with morphological abnormalities of adrenal development, including ectopic TH+ cells within the adrenal cortex and external to the adrenal gland. These changes were ablated by combined inactivation of Phd2 with Hif-2α, but not Hif-1α. However, they could not be reproduced by inactivation of Phd2 in adult life, suggesting that they arise from dysregulation of this pathway during adrenal development. Together with the clinical observation that pseudohypoxic PPGL manifests remarkably high heritability, our findings suggest that this type of tumour likely arises from dysregulation of a tissue-restricted action of the PHD2/HIF-2α pathway affecting adrenal development in early life and provides a model for the study of the relevant processes.


Sujet(s)
Tumeurs de la surrénale , Hypoxia-inducible factor-proline dioxygenases/métabolisme , Paragangliome , Phéochromocytome , Tumeurs de la surrénale/génétique , Animaux , Facteurs de transcription à motif basique hélice-boucle-hélice/génétique , Facteurs de transcription à motif basique hélice-boucle-hélice/métabolisme , Humains , Sous-unité alpha du facteur-1 induit par l'hypoxie , Hypoxia-inducible factor-proline dioxygenases/génétique , Souris , Paragangliome/génétique , Phéochromocytome/génétique
18.
Nat Genet ; 53(7): 1022-1035, 2021 07.
Article de Anglais | MEDLINE | ID: mdl-34155378

RÉSUMÉ

Hypoxia-inducible transcription factors (HIFs) are fundamental to cellular adaptation to low oxygen levels, but it is unclear how they interact with chromatin and activate their target genes. Here, we use genome-wide mutagenesis to identify genes involved in HIF transcriptional activity, and define a requirement for the histone H3 lysine 4 (H3K4) methyltransferase SET1B. SET1B loss leads to a selective reduction in transcriptional activation of HIF target genes, resulting in impaired cell growth, angiogenesis and tumor establishment in SET1B-deficient xenografts. Mechanistically, we show that SET1B accumulates on chromatin in hypoxia, and is recruited to HIF target genes by the HIF complex. The selective induction of H3K4 trimethylation at HIF target loci is both HIF- and SET1B-dependent and, when impaired, correlates with decreased promoter acetylation and gene expression. Together, these findings show SET1B as a determinant of site-specific histone methylation and provide insight into how HIF target genes are differentially regulated.


Sujet(s)
Facteurs de transcription à motif basique hélice-boucle-hélice/métabolisme , Régulation de l'expression des gènes , Histone-lysine N-methyltransferase/métabolisme , Hypoxie/génétique , Acétylation , Animaux , Humains , Hypoxie/métabolisme , Méthylation , Souris , Souris knockout , Modèles animaux , Régions promotrices (génétique) , Liaison aux protéines
19.
Cell Rep ; 35(3): 109020, 2021 04 20.
Article de Anglais | MEDLINE | ID: mdl-33852916

RÉSUMÉ

COVID-19, caused by the novel coronavirus SARS-CoV-2, is a global health issue with more than 2 million fatalities to date. Viral replication is shaped by the cellular microenvironment, and one important factor to consider is oxygen tension, in which hypoxia inducible factor (HIF) regulates transcriptional responses to hypoxia. SARS-CoV-2 primarily infects cells of the respiratory tract, entering via its spike glycoprotein binding to angiotensin-converting enzyme 2 (ACE2). We demonstrate that hypoxia and the HIF prolyl hydroxylase inhibitor Roxadustat reduce ACE2 expression and inhibit SARS-CoV-2 entry and replication in lung epithelial cells via an HIF-1α-dependent pathway. Hypoxia and Roxadustat inhibit SARS-CoV-2 RNA replication, showing that post-entry steps in the viral life cycle are oxygen sensitive. This study highlights the importance of HIF signaling in regulating multiple aspects of SARS-CoV-2 infection and raises the potential use of HIF prolyl hydroxylase inhibitors in the prevention or treatment of COVID-19.


Sujet(s)
COVID-19/métabolisme , Cellules épithéliales/métabolisme , Glycine/analogues et dérivés , Sous-unité alpha du facteur-1 induit par l'hypoxie/métabolisme , Isoquinoléines/pharmacologie , Poumon/métabolisme , SARS-CoV-2/physiologie , Pénétration virale/effets des médicaments et des substances chimiques , Réplication virale/effets des médicaments et des substances chimiques , Cellules A549 , Animaux , COVID-19/anatomopathologie , Cellules Caco-2 , Hypoxie cellulaire/effets des médicaments et des substances chimiques , Chlorocebus aethiops , Cellules épithéliales/virologie , Glycine/pharmacologie , Humains , Poumon/virologie , Souris , Cellules Vero , Traitements médicamenteux de la COVID-19
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE