Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 54.997
Filtrar
Más filtros

Intervalo de año de publicación
1.
Annu Rev Immunol ; 40: 589-614, 2022 04 26.
Artículo en Inglés | MEDLINE | ID: mdl-35130029

RESUMEN

Pulmonary granulomas are widely considered the epicenters of the immune response to Mycobacterium tuberculosis (Mtb), the causative agent of tuberculosis (TB). Recent animal studies have revealed factors that either promote or restrict TB immunity within granulomas. These models, however, typically ignore the impact of preexisting immunity on cellular organization and function, an important consideration because most TB probably occurs through reinfection of previously exposed individuals. Human postmortem research from the pre-antibiotic era showed that infections in Mtb-naïve individuals (primary TB) versus those with prior Mtb exposure (postprimary TB) have distinct pathologic features. We review recent animal findings in TB granuloma biology, which largely reflect primary TB. We also discuss our current understanding of postprimary TB lesions, about which much less is known. Many knowledge gaps remain, particularly regarding how preexisting immunity shapes granuloma structure and local immune responses at Mtb infection sites.


Asunto(s)
Mycobacterium tuberculosis , Tuberculosis , Animales , Granuloma/etiología , Humanos , Pulmón/microbiología , Pulmón/patología
2.
Annu Rev Immunol ; 39: 611-637, 2021 04 26.
Artículo en Inglés | MEDLINE | ID: mdl-33637017

RESUMEN

Infection with Mycobacterium tuberculosis causes >1.5 million deaths worldwide annually. Innate immune cells are the first to encounter M. tuberculosis, and their response dictates the course of infection. Dendritic cells (DCs) activate the adaptive response and determine its characteristics. Macrophages are responsible both for exerting cell-intrinsic antimicrobial control and for initiating and maintaining inflammation. The inflammatory response to M. tuberculosis infection is a double-edged sword. While cytokines such as TNF-α and IL-1 are important for protection, either excessive or insufficient cytokine production results in progressive disease. Furthermore, neutrophils-cells normally associated with control of bacterial infection-are emerging as key drivers of a hyperinflammatory response that results in host mortality. The roles of other innate cells, including natural killer cells and innate-like T cells, remain enigmatic. Understanding the nuances of both cell-intrinsic control of infection and regulation of inflammation will be crucial for the successful development of host-targeted therapeutics and vaccines.


Asunto(s)
Mycobacterium tuberculosis , Tuberculosis , Animales , Citocinas , Humanos , Inmunidad Innata , Macrófagos
3.
Annu Rev Immunol ; 36: 603-638, 2018 04 26.
Artículo en Inglés | MEDLINE | ID: mdl-29490165

RESUMEN

Globally, about 36.7 million people were living with HIV infection at the end of 2015. The most frequent infection co-occurring with HIV-1 is Mycobacterium tuberculosis-374,000 deaths per annum are attributable to HIV-tuberculosis, 75% of those occurring in Africa. HIV-1 infection increases the risk of tuberculosis by a factor of up to 26 and alters its clinical presentation, complicates diagnosis and treatment, and worsens outcome. Although HIV-1-induced depletion of CD4+ T cells underlies all these effects, more widespread immune deficits also contribute to susceptibility and pathogenesis. These defects present a challenge to understand and ameliorate, but also an opportunity to learn and optimize mechanisms that normally protect people against tuberculosis. The most effective means to prevent and ameliorate tuberculosis in HIV-1-infected people is antiretroviral therapy, but this may be complicated by pathological immune deterioration that in turn requires more effective host-directed anti-inflammatory therapies to be derived.


Asunto(s)
Coinfección , Infecciones por VIH/inmunología , VIH-1/inmunología , Interacciones Huésped-Patógeno/inmunología , Inmunidad , Mycobacterium tuberculosis/inmunología , Tuberculosis/inmunología , Animales , Terapia Antirretroviral Altamente Activa , Antituberculosos/farmacología , Antituberculosos/uso terapéutico , Progresión de la Enfermedad , Variación Genética , Infecciones por VIH/diagnóstico , Infecciones por VIH/terapia , Infecciones por VIH/virología , VIH-1/genética , Humanos , Tuberculosis/diagnóstico , Tuberculosis/microbiología , Tuberculosis/terapia , Replicación Viral
4.
Cell ; 186(23): 4994-4995, 2023 11 09.
Artículo en Inglés | MEDLINE | ID: mdl-37949055

RESUMEN

Mechanobiology explores how cells sense and respond to mechanical cues and how mechanics guide cell function, physiology, and disease. In this issue of Cell, Thacker and colleagues reveal how the tuberculosis-causing pathogen exploits the mechanical behavior of cord-like structures to promote infection, impacting immune response, antibiotic susceptibility, and treatment strategies.


Asunto(s)
Fenómenos Biomecánicos , Mycobacterium tuberculosis , Humanos , Biofisica , Tuberculosis/microbiología , Mycobacterium tuberculosis/fisiología
5.
Cell ; 186(23): 5135-5150.e28, 2023 11 09.
Artículo en Inglés | MEDLINE | ID: mdl-37865090

RESUMEN

Mycobacterium tuberculosis (Mtb) cultured axenically without detergent forms biofilm-like cords, a clinical identifier of virulence. In lung-on-chip (LoC) and mouse models, cords in alveolar cells contribute to suppression of innate immune signaling via nuclear compression. Thereafter, extracellular cords cause contact-dependent phagocyte death but grow intercellularly between epithelial cells. The absence of these mechanopathological mechanisms explains the greater proportion of alveolar lesions with increased immune infiltration and dissemination defects in cording-deficient Mtb infections. Compression of Mtb lipid monolayers induces a phase transition that enables mechanical energy storage. Agent-based simulations demonstrate that the increased energy storage capacity is sufficient for the formation of cords that maintain structural integrity despite mechanical perturbation. Bacteria in cords remain translationally active despite antibiotic exposure and regrow rapidly upon cessation of treatment. This study provides a conceptual framework for the biophysics and function in tuberculosis infection and therapy of cord architectures independent of mechanisms ascribed to single bacteria.


Asunto(s)
Mycobacterium tuberculosis , Tuberculosis , Animales , Ratones , Biopelículas , Pulmón/microbiología , Pulmón/patología , Mycobacterium tuberculosis/fisiología , Tuberculosis/microbiología , Tuberculosis/patología , Virulencia , Fenómenos Biomecánicos
6.
Cell ; 186(5): 1013-1025.e24, 2023 03 02.
Artículo en Inglés | MEDLINE | ID: mdl-36827973

RESUMEN

The emergence of drug-resistant tuberculosis has created an urgent need for new anti-tubercular agents. Here, we report the discovery of a series of macrolides called sequanamycins with outstanding in vitro and in vivo activity against Mycobacterium tuberculosis (Mtb). Sequanamycins are bacterial ribosome inhibitors that interact with the ribosome in a similar manner to classic macrolides like erythromycin and clarithromycin, but with binding characteristics that allow them to overcome the inherent macrolide resistance of Mtb. Structures of the ribosome with bound inhibitors were used to optimize sequanamycin to produce the advanced lead compound SEQ-9. SEQ-9 was efficacious in mouse models of acute and chronic TB as a single agent, and it demonstrated bactericidal activity in a murine TB infection model in combination with other TB drugs. These results support further investigation of this series as TB clinical candidates, with the potential for use in new regimens against drug-susceptible and drug-resistant TB.


Asunto(s)
Antituberculosos , Mycobacterium tuberculosis , Animales , Ratones , Antituberculosos/farmacología , Macrólidos , Farmacorresistencia Bacteriana , Claritromicina
7.
Cell ; 186(10): 2176-2192.e22, 2023 05 11.
Artículo en Inglés | MEDLINE | ID: mdl-37137307

RESUMEN

The ClpC1:ClpP1P2 protease is a core component of the proteostasis system in mycobacteria. To improve the efficacy of antitubercular agents targeting the Clp protease, we characterized the mechanism of the antibiotics cyclomarin A and ecumicin. Quantitative proteomics revealed that the antibiotics cause massive proteome imbalances, including upregulation of two unannotated yet conserved stress response factors, ClpC2 and ClpC3. These proteins likely protect the Clp protease from excessive amounts of misfolded proteins or from cyclomarin A, which we show to mimic damaged proteins. To overcome the Clp security system, we developed a BacPROTAC that induces degradation of ClpC1 together with its ClpC2 caretaker. The dual Clp degrader, built from linked cyclomarin A heads, was highly efficient in killing pathogenic Mycobacterium tuberculosis, with >100-fold increased potency over the parent antibiotic. Together, our data reveal Clp scavenger proteins as important proteostasis safeguards and highlight the potential of BacPROTACs as future antibiotics.


Asunto(s)
Antituberculosos , Mycobacterium tuberculosis , Antituberculosos/farmacología , Proteínas Bacterianas/metabolismo , Endopeptidasa Clp/metabolismo , Proteínas de Choque Térmico/metabolismo , Mycobacterium tuberculosis/efectos de los fármacos , Proteostasis
8.
Cell ; 185(20): 3720-3738.e13, 2022 09 29.
Artículo en Inglés | MEDLINE | ID: mdl-36103894

RESUMEN

Necrosis of macrophages in the granuloma, the hallmark immunological structure of tuberculosis, is a major pathogenic event that increases host susceptibility. Through a zebrafish forward genetic screen, we identified the mTOR kinase, a master regulator of metabolism, as an early host resistance factor in tuberculosis. We found that mTOR complex 1 protects macrophages from mycobacterium-induced death by enabling infection-induced increases in mitochondrial energy metabolism fueled by glycolysis. These metabolic adaptations are required to prevent mitochondrial damage and death caused by the secreted mycobacterial virulence determinant ESAT-6. Thus, the host can effectively counter this early critical mycobacterial virulence mechanism simply by regulating energy metabolism, thereby allowing pathogen-specific immune mechanisms time to develop. Our findings may explain why Mycobacterium tuberculosis, albeit humanity's most lethal pathogen, is successful in only a minority of infected individuals.


Asunto(s)
Mycobacterium marinum , Mycobacterium tuberculosis , Tuberculosis , Animales , Mycobacterium tuberculosis/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Pez Cebra
9.
Cell ; 185(25): 4682-4702, 2022 Dec 08.
Artículo en Inglés | MEDLINE | ID: mdl-36493751

RESUMEN

Despite having been identified as the organism that causes tuberculosis in 1882, Mycobacterium tuberculosis has managed to still evade our understanding of the protective immune response against it, defying the development of an effective vaccine. Technology and novel experimental models have revealed much new knowledge, particularly with respect to the heterogeneity of the bacillus and the host response. This review focuses on certain immunological elements that have recently yielded exciting data and highlights the importance of taking a holistic approach to understanding the interaction of M. tuberculosis with the many host cells that contribute to the development of protective immunity.


Asunto(s)
Mycobacterium tuberculosis , Tuberculosis , Humanos , Comunicación Celular , Tuberculosis/inmunología
10.
Cell ; 185(24): 4467-4469, 2022 11 23.
Artículo en Inglés | MEDLINE | ID: mdl-36423577

RESUMEN

In this issue of Cell, Saelens et al. describe a new function for mycobacterial Type VII secretion systems: manipulation of host cell migration. They find that a substantial proportion of global TB cases arise from bacteria lacking this function, raising questions about its role in pathoadaptation of Mycobacterium tuberculosis.


Asunto(s)
Mycobacterium tuberculosis , Tuberculosis , Sistemas de Secreción Tipo VII , Humanos , Sistemas de Secreción Tipo VII/metabolismo , Mycobacterium tuberculosis/metabolismo , Tuberculosis/microbiología , Movimiento Celular
11.
Cell ; 185(24): 4507-4525.e18, 2022 11 23.
Artículo en Inglés | MEDLINE | ID: mdl-36356582

RESUMEN

The human pathogen Mycobacterium tuberculosis typically causes lung disease but can also disseminate to other tissues. We identified a M. tuberculosis (Mtb) outbreak presenting with unusually high rates of extrapulmonary dissemination and bone disease. We found that the causal strain carried an ancestral full-length version of the type VII-secreted effector EsxM rather than the truncated version present in other modern Mtb lineages. The ancestral EsxM variant exacerbated dissemination through enhancement of macrophage motility, increased egress of macrophages from established granulomas, and alterations in macrophage actin dynamics. Reconstitution of the ancestral version of EsxM in an attenuated modern strain of Mtb altered the migratory mode of infected macrophages, enhancing their motility. In a zebrafish model, full-length EsxM promoted bone disease. The presence of a derived nonsense variant in EsxM throughout the major Mtb lineages 2, 3, and 4 is consistent with a role for EsxM in regulating the extent of dissemination.


Asunto(s)
Enfermedades Óseas , Mycobacterium marinum , Mycobacterium tuberculosis , Tuberculosis , Animales , Humanos , Pez Cebra , Tuberculosis/microbiología , Macrófagos/microbiología , Proteínas Bacterianas/genética
12.
Cell ; 184(17): 4579-4592.e24, 2021 08 19.
Artículo en Inglés | MEDLINE | ID: mdl-34297925

RESUMEN

Antibacterial agents target the products of essential genes but rarely achieve complete target inhibition. Thus, the all-or-none definition of essentiality afforded by traditional genetic approaches fails to discern the most attractive bacterial targets: those whose incomplete inhibition results in major fitness costs. In contrast, gene "vulnerability" is a continuous, quantifiable trait that relates the magnitude of gene inhibition to the effect on bacterial fitness. We developed a CRISPR interference-based functional genomics method to systematically titrate gene expression in Mycobacterium tuberculosis (Mtb) and monitor fitness outcomes. We identified highly vulnerable genes in various processes, including novel targets unexplored for drug discovery. Equally important, we identified invulnerable essential genes, potentially explaining failed drug discovery efforts. Comparison of vulnerability between the reference and a hypervirulent Mtb isolate revealed incomplete conservation of vulnerability and that differential vulnerability can predict differential antibacterial susceptibility. Our results quantitatively redefine essential bacterial processes and identify high-value targets for drug development.


Asunto(s)
Regulación Bacteriana de la Expresión Génica , Genoma Bacteriano , Mycobacterium tuberculosis/genética , Aminoacil-ARNt Sintetasas/metabolismo , Antituberculosos/farmacología , Teorema de Bayes , Evolución Biológica , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas/genética , Regulación Bacteriana de la Expresión Génica/efectos de los fármacos , Silenciador del Gen/efectos de los fármacos , Pruebas de Sensibilidad Microbiana , Mycobacterium tuberculosis/efectos de los fármacos , ARN Guía de Kinetoplastida/genética
13.
Nat Immunol ; 24(5): 855-868, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-37012543

RESUMEN

Tuberculosis (TB), caused by Mycobacterium tuberculosis (Mtb), is a global cause of death. Granuloma-associated lymphoid tissue (GrALT) correlates with protection during TB, but the mechanisms of protection are not understood. During TB, the transcription factor IRF4 in T cells but not B cells is required for the generation of the TH1 and TH17 subsets of helper T cells and follicular helper T (TFH)-like cellular responses. A population of IRF4+ T cells coexpress the transcription factor BCL6 during Mtb infection, and deletion of Bcl6 (Bcl6fl/fl) in CD4+ T cells (CD4cre) resulted in reduction of TFH-like cells, impaired localization within GrALT and increased Mtb burden. In contrast, the absence of germinal center B cells, MHC class II expression on B cells, antibody-producing plasma cells or interleukin-10-expressing B cells, did not increase Mtb susceptibility. Indeed, antigen-specific B cells enhance cytokine production and strategically localize TFH-like cells within GrALT via interactions between programmed cell death 1 (PD-1) and its ligand PD-L1 and mediate Mtb control in both mice and macaques.


Asunto(s)
Mycobacterium tuberculosis , Tuberculosis , Ratones , Animales , Linfocitos T Colaboradores-Inductores , Linfocitos B , Tejido Linfoide , Centro Germinal , Factores de Transcripción
14.
Annu Rev Immunol ; 31: 475-527, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23516984

RESUMEN

There are 9 million cases of active tuberculosis reported annually; however, an estimated one-third of the world's population is infected with Mycobacterium tuberculosis and remains asymptomatic. Of these latent individuals, only 5-10% will develop active tuberculosis disease in their lifetime. CD4(+) T cells, as well as the cytokines IL-12, IFN-γ, and TNF, are critical in the control of Mycobacterium tuberculosis infection, but the host factors that determine why some individuals are protected from infection while others go on to develop disease are unclear. Genetic factors of the host and of the pathogen itself may be associated with an increased risk of patients developing active tuberculosis. This review aims to summarize what we know about the immune response in tuberculosis, in human disease, and in a range of experimental models, all of which are essential to advancing our mechanistic knowledge base of the host-pathogen interactions that influence disease outcome.


Asunto(s)
Tuberculosis Pulmonar/inmunología , Animales , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Predisposición Genética a la Enfermedad , Antígenos de Histocompatibilidad Clase II/inmunología , Humanos , Mycobacterium tuberculosis/inmunología , Resultado del Tratamiento , Tuberculosis Pulmonar/diagnóstico , Tuberculosis Pulmonar/epidemiología
15.
Annu Rev Immunol ; 31: 605-33, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23516986

RESUMEN

Evidence has increasingly shown that the lungs are a major site of immune regulation. A robust and highly regulated immune response in the lung protects the host from pathogen infection, whereas an inefficient or deleterious response can lead to various pulmonary diseases. Many cell types, such as epithelial cells, dendritic cells, macrophages, neutrophils, eosinophils, and B and T lymphocytes, contribute to lung immunity. This review focuses on the recent advances in understanding how T lymphocytes mediate pulmonary host defenses against bacterial, viral, and fungal pathogens.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/patología , Pulmón/inmunología , Pulmón/patología , Tuberculosis Pulmonar/inmunología , Animales , Linfocitos T CD4-Positivos/metabolismo , Interacciones Huésped-Patógeno/inmunología , Humanos , Pulmón/microbiología , Ganglios Linfáticos/inmunología , Ganglios Linfáticos/microbiología , Ganglios Linfáticos/patología , Mycobacterium tuberculosis/inmunología , Tuberculosis Pulmonar/microbiología , Tuberculosis Pulmonar/patología
16.
Cell ; 181(2): 230-232, 2020 04 16.
Artículo en Inglés | MEDLINE | ID: mdl-32302566

RESUMEN

Cough, a hallmark of tuberculosis, transmits the disease. Ruhl et al. find that a Mycobacterium tuberculosis (Mtb)-specific lipid, SL-1, stimulates human nociceptive neurons and makes guinea pigs cough. Mtb extract, but not SL-1, also stimulates non-nociceptive neurons that participate in the cough reflex, suggesting additional cough-inducing mechanisms.


Asunto(s)
Mycobacterium tuberculosis , Tuberculosis , Animales , Tos , Cobayas , Humanos , Lípidos , Nociceptores
17.
Cell ; 181(2): 293-305.e11, 2020 04 16.
Artículo en Inglés | MEDLINE | ID: mdl-32142653

RESUMEN

Pulmonary tuberculosis, a disease caused by Mycobacterium tuberculosis (Mtb), manifests with a persistent cough as both a primary symptom and mechanism of transmission. The cough reflex can be triggered by nociceptive neurons innervating the lungs, and some bacteria produce neuron-targeting molecules. However, how pulmonary Mtb infection causes cough remains undefined, and whether Mtb produces a neuron-activating, cough-inducing molecule is unknown. Here, we show that an Mtb organic extract activates nociceptive neurons in vitro and identify the Mtb glycolipid sulfolipid-1 (SL-1) as the nociceptive molecule. Mtb organic extracts from mutants lacking SL-1 synthesis cannot activate neurons in vitro or induce cough in a guinea pig model. Finally, Mtb-infected guinea pigs cough in a manner dependent on SL-1 synthesis. Thus, we demonstrate a heretofore unknown molecular mechanism for cough induction by a virulent human pathogen via its production of a complex lipid.


Asunto(s)
Tos/fisiopatología , Glucolípidos/metabolismo , Nociceptores/fisiología , Factores de Virulencia/metabolismo , Adulto , Animales , Línea Celular , Tos/etiología , Tos/microbiología , Femenino , Glucolípidos/fisiología , Cobayas , Interacciones Huésped-Patógeno , Humanos , Lípidos/fisiología , Pulmón/microbiología , Macrófagos/microbiología , Masculino , Ratones , Mycobacterium tuberculosis/metabolismo , Mycobacterium tuberculosis/patogenicidad , Cultivo Primario de Células , Tuberculosis/microbiología , Tuberculosis Pulmonar/microbiología , Tuberculosis Pulmonar/fisiopatología , Factores de Virulencia/fisiología
18.
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
19.
Cell ; 180(4): 688-702.e13, 2020 02 20.
Artículo en Inglés | MEDLINE | ID: mdl-32084340

RESUMEN

Due to the rapid emergence of antibiotic-resistant bacteria, there is a growing need to discover new antibiotics. To address this challenge, we trained a deep neural network capable of predicting molecules with antibacterial activity. We performed predictions on multiple chemical libraries and discovered a molecule from the Drug Repurposing Hub-halicin-that is structurally divergent from conventional antibiotics and displays bactericidal activity against a wide phylogenetic spectrum of pathogens including Mycobacterium tuberculosis and carbapenem-resistant Enterobacteriaceae. Halicin also effectively treated Clostridioides difficile and pan-resistant Acinetobacter baumannii infections in murine models. Additionally, from a discrete set of 23 empirically tested predictions from >107 million molecules curated from the ZINC15 database, our model identified eight antibacterial compounds that are structurally distant from known antibiotics. This work highlights the utility of deep learning approaches to expand our antibiotic arsenal through the discovery of structurally distinct antibacterial molecules.


Asunto(s)
Antibacterianos/farmacología , Descubrimiento de Drogas/métodos , Aprendizaje Automático , Tiadiazoles/farmacología , Acinetobacter baumannii/efectos de los fármacos , Animales , Antibacterianos/química , Quimioinformática/métodos , Clostridioides difficile/efectos de los fármacos , Bases de Datos de Compuestos Químicos , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Mycobacterium tuberculosis/efectos de los fármacos , Bibliotecas de Moléculas Pequeñas/química , Bibliotecas de Moléculas Pequeñas/farmacología , Tiadiazoles/química
20.
Cell ; 181(5): 1016-1035.e19, 2020 05 28.
Artículo en Inglés | MEDLINE | ID: mdl-32413319

RESUMEN

There is pressing urgency to understand the pathogenesis of the severe acute respiratory syndrome coronavirus clade 2 (SARS-CoV-2), which causes the disease COVID-19. SARS-CoV-2 spike (S) protein binds angiotensin-converting enzyme 2 (ACE2), and in concert with host proteases, principally transmembrane serine protease 2 (TMPRSS2), promotes cellular entry. The cell subsets targeted by SARS-CoV-2 in host tissues and the factors that regulate ACE2 expression remain unknown. Here, we leverage human, non-human primate, and mouse single-cell RNA-sequencing (scRNA-seq) datasets across health and disease to uncover putative targets of SARS-CoV-2 among tissue-resident cell subsets. We identify ACE2 and TMPRSS2 co-expressing cells within lung type II pneumocytes, ileal absorptive enterocytes, and nasal goblet secretory cells. Strikingly, we discovered that ACE2 is a human interferon-stimulated gene (ISG) in vitro using airway epithelial cells and extend our findings to in vivo viral infections. Our data suggest that SARS-CoV-2 could exploit species-specific interferon-driven upregulation of ACE2, a tissue-protective mediator during lung injury, to enhance infection.


Asunto(s)
Células Epiteliales Alveolares/metabolismo , Enterocitos/metabolismo , Células Caliciformes/metabolismo , Interferón Tipo I/metabolismo , Mucosa Nasal/citología , Peptidil-Dipeptidasa A/genética , Adolescente , Células Epiteliales Alveolares/inmunología , Enzima Convertidora de Angiotensina 2 , Animales , Betacoronavirus/fisiología , COVID-19 , Línea Celular , Células Cultivadas , Niño , Infecciones por Coronavirus/virología , Enterocitos/inmunología , Células Caliciformes/inmunología , Infecciones por VIH/inmunología , Humanos , Gripe Humana/inmunología , Interferón Tipo I/inmunología , Pulmón/citología , Pulmón/patología , Macaca mulatta , Ratones , Mycobacterium tuberculosis , Mucosa Nasal/inmunología , Pandemias , Peptidil-Dipeptidasa A/metabolismo , Neumonía Viral/virología , Receptores Virales/genética , SARS-CoV-2 , Serina Endopeptidasas/metabolismo , Análisis de la Célula Individual , Tuberculosis/inmunología , Regulación hacia Arriba
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA