Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 52.851
Filtrar
Más filtros

Intervalo de año de publicación
1.
Annu Rev Immunol ; 39: 1-18, 2021 04 26.
Artículo en Inglés | MEDLINE | ID: mdl-33902314

RESUMEN

An imbalance in the microbiota may contribute to many human illnesses, which has prompted efforts to rebalance it by targeting the microbes themselves. However, by supplying the habitat, the host wields a prominent influence over microbial growth at body surfaces, raising the possibility that rebalancing the microbiota by targeting our immune system would be a viable alternative. Host control mechanisms that sculpt the microbial habitat form a functional unit with the microbiota, termed microbiota-nourishing immunity, that confers colonization resistance against pathogens. The host components of microbiota-nourishing immunity can be viewed as habitat filters that select for microbial traits licensing growth and survival in host habitat patches. Here we review current knowledge of how host-derived habitat filters shape the size, species composition, and spatial heterogeneity of the microbiota and discuss whether these host control mechanisms could be harnessed for developing approaches to rebalance microbial communities during dysbiosis.


Asunto(s)
Disbiosis , Microbiota , Animales , Humanos
2.
Cell ; 187(14): 3761-3778.e16, 2024 Jul 11.
Artículo en Inglés | MEDLINE | ID: mdl-38843834

RESUMEN

Novel antibiotics are urgently needed to combat the antibiotic-resistance crisis. We present a machine-learning-based approach to predict antimicrobial peptides (AMPs) within the global microbiome and leverage a vast dataset of 63,410 metagenomes and 87,920 prokaryotic genomes from environmental and host-associated habitats to create the AMPSphere, a comprehensive catalog comprising 863,498 non-redundant peptides, few of which match existing databases. AMPSphere provides insights into the evolutionary origins of peptides, including by duplication or gene truncation of longer sequences, and we observed that AMP production varies by habitat. To validate our predictions, we synthesized and tested 100 AMPs against clinically relevant drug-resistant pathogens and human gut commensals both in vitro and in vivo. A total of 79 peptides were active, with 63 targeting pathogens. These active AMPs exhibited antibacterial activity by disrupting bacterial membranes. In conclusion, our approach identified nearly one million prokaryotic AMP sequences, an open-access resource for antibiotic discovery.


Asunto(s)
Péptidos Antimicrobianos , Aprendizaje Automático , Microbiota , Péptidos Antimicrobianos/farmacología , Péptidos Antimicrobianos/química , Péptidos Antimicrobianos/genética , Humanos , Animales , Antibacterianos/farmacología , Ratones , Metagenoma , Bacterias/efectos de los fármacos , Bacterias/genética , Microbioma Gastrointestinal/efectos de los fármacos
3.
Cell ; 187(19): 5453-5467.e15, 2024 Sep 19.
Artículo en Inglés | MEDLINE | ID: mdl-39163860

RESUMEN

Drug-resistant bacteria are outpacing traditional antibiotic discovery efforts. Here, we computationally screened 444,054 previously reported putative small protein families from 1,773 human metagenomes for antimicrobial properties, identifying 323 candidates encoded in small open reading frames (smORFs). To test our computational predictions, 78 peptides were synthesized and screened for antimicrobial activity in vitro, with 70.5% displaying antimicrobial activity. As these compounds were different compared with previously reported antimicrobial peptides, we termed them smORF-encoded peptides (SEPs). SEPs killed bacteria by targeting their membrane, synergizing with each other, and modulating gut commensals, indicating a potential role in reconfiguring microbiome communities in addition to counteracting pathogens. The lead candidates were anti-infective in both murine skin abscess and deep thigh infection models. Notably, prevotellin-2 from Prevotella copri presented activity comparable to the commonly used antibiotic polymyxin B. Our report supports the existence of hundreds of antimicrobials in the human microbiome amenable to clinical translation.


Asunto(s)
Antibacterianos , Péptidos Antimicrobianos , Microbiota , Humanos , Animales , Ratones , Antibacterianos/farmacología , Microbiota/efectos de los fármacos , Péptidos Antimicrobianos/farmacología , Péptidos Antimicrobianos/química , Metagenoma , Femenino , Sistemas de Lectura Abierta , Bacterias/efectos de los fármacos , Bacterias/genética , Bacterias/clasificación , Prevotella/efectos de los fármacos
4.
Cell ; 186(23): 5098-5113.e19, 2023 11 09.
Artículo en Inglés | MEDLINE | ID: mdl-37918395

RESUMEN

Drug-resistant Pseudomonas aeruginosa (PA) poses an emerging threat to human health with urgent need for alternative therapeutic approaches. Here, we deciphered the B cell and antibody response to the virulence-associated type III secretion system (T3SS) in a cohort of patients chronically infected with PA. Single-cell analytics revealed a diverse B cell receptor repertoire directed against the T3SS needle-tip protein PcrV, enabling the production of monoclonal antibodies (mAbs) abrogating T3SS-mediated cytotoxicity. Mechanistic studies involving cryoelectron microscopy identified a surface-exposed C-terminal PcrV epitope as the target of highly neutralizing mAbs with broad activity against drug-resistant PA isolates. These anti-PcrV mAbs were as effective as treatment with conventional antibiotics in vivo. Our study reveals that chronically infected patients represent a source of neutralizing antibodies, which can be exploited as therapeutics against PA.


Asunto(s)
Anticuerpos Antibacterianos , Anticuerpos Neutralizantes , Infecciones por Pseudomonas , Pseudomonas aeruginosa , Humanos , Anticuerpos Antibacterianos/farmacología , Microscopía por Crioelectrón , Inmunoglobulinas/metabolismo , Pseudomonas aeruginosa/efectos de los fármacos , Pseudomonas aeruginosa/fisiología , Infecciones por Pseudomonas/tratamiento farmacológico
5.
Annu Rev Biochem ; 91: 449-473, 2022 06 21.
Artículo en Inglés | MEDLINE | ID: mdl-35303792

RESUMEN

Metals are essential components in life processes and participate in many important biological processes. Dysregulation of metal homeostasis is correlated with many diseases. Metals are also frequently incorporated into diagnosis and therapeutics. Understanding of metal homeostasis under (patho)physiological conditions and the molecular mechanisms of action of metallodrugs in biological systems has positive impacts on human health. As an emerging interdisciplinary area of research, metalloproteomics involves investigating metal-protein interactions in biological systems at a proteome-wide scale, has received growing attention, and has been implemented into metal-related research. In this review, we summarize the recent advances in metalloproteomics methodologies and applications. We also highlight emerging single-cell metalloproteomics, including time-resolved inductively coupled plasma mass spectrometry, mass cytometry, and secondary ion mass spectrometry. Finally, we discuss future perspectives in metalloproteomics, aiming to attract more original research to develop more advanced methodologies, which could be utilized rapidly by biochemists or biologists to expand our knowledge of how metal functions in biology and medicine.


Asunto(s)
Investigación Biomédica , Metaloproteínas , Humanos , Metaloproteínas/análisis , Metaloproteínas/química , Metaloproteínas/genética , Metales/análisis , Metales/química , Proteoma/genética , Proteómica/métodos
6.
Annu Rev Biochem ; 91: 403-422, 2022 06 21.
Artículo en Inglés | MEDLINE | ID: mdl-35729071

RESUMEN

The remarkable variety of microbial species of human pathogens and microbiomes generates significant quantities of secreted amyloids, which are structured protein fibrils that serve diverse functions related to virulence and interactions with the host. Human amyloids are associated largely with fatal neurodegenerative and systemic aggregation diseases, and current research has put forward the hypothesis that the interspecies amyloid interactome has physiological and pathological significance. Moreover, functional and molecular-level connections between antimicrobial activity and amyloid structures suggest a neuroimmune role for amyloids that are otherwise known to be pathological. Compared to the extensive structural information that has been accumulated for human amyloids, high-resolution structures of microbial and antimicrobial amyloids are only emerging. These recent structures reveal both similarities and surprising departures from the typical amyloid motif, in accordance with their diverse activities, and advance the discovery of novel antivirulence and antimicrobial agents. In addition, the structural information has led researchers to postulate that amyloidogenic sequences are natural targets for structural mimicry, for instance in host-microbe interactions. Microbial amyloid research could ultimately be used to fight aggressive infections and possibly processes leading to autoimmune and neurodegenerative diseases.


Asunto(s)
Amiloidosis , Antiinfecciosos , Enfermedades Neurodegenerativas , Amiloide/química , Proteínas Amiloidogénicas , Amiloidosis/metabolismo , Antibacterianos , Antiinfecciosos/farmacología , Humanos , Enfermedades Neurodegenerativas/tratamiento farmacológico , Enfermedades Neurodegenerativas/genética , Enfermedades Neurodegenerativas/metabolismo
7.
Annu Rev Immunol ; 33: 227-56, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25581310

RESUMEN

The diverse microbial populations constituting the intestinal microbiota promote immune development and differentiation, but because of their complex metabolic requirements and the consequent difficulty culturing them, they remained, until recently, largely uncharacterized and mysterious. In the last decade, deep nucleic acid sequencing platforms, new computational and bioinformatics tools, and full-genome characterization of several hundred commensal bacterial species facilitated studies of the microbiota and revealed that differences in microbiota composition can be associated with inflammatory, metabolic, and infectious diseases, that each human is colonized by a distinct bacterial flora, and that the microbiota can be manipulated to reduce and even cure some diseases. Different bacterial species induce distinct immune cell populations that can play pro- and anti-inflammatory roles, and thus the composition of the microbiota determines, in part, the level of resistance to infection and susceptibility to inflammatory diseases. This review summarizes recent work characterizing commensal microbes that contribute to the antimicrobial defense/inflammation axis.


Asunto(s)
Resistencia a la Enfermedad/inmunología , Gastroenteritis/inmunología , Gastroenteritis/microbiología , Microbioma Gastrointestinal/inmunología , Mucosa Intestinal/inmunología , Mucosa Intestinal/microbiología , Inmunidad Adaptativa , Animales , Enfermedades Autoinmunes/inmunología , Enfermedades Autoinmunes/metabolismo , Enfermedades Autoinmunes/microbiología , Biología Computacional , Dieta , Susceptibilidad a Enfermedades , Gastroenteritis/metabolismo , Interacciones Huésped-Patógeno/inmunología , Humanos , Inmunidad Innata , Inmunidad Mucosa , Mucosa Intestinal/metabolismo , Metaboloma , Neoplasias/etiología , Vitaminas/metabolismo
8.
Cell ; 184(16): 4154-4167.e12, 2021 08 05.
Artículo en Inglés | MEDLINE | ID: mdl-34324837

RESUMEN

Environmental light cycles entrain circadian feeding behaviors in animals that produce rhythms in exposure to foodborne bacteria. Here, we show that the intestinal microbiota generates diurnal rhythms in innate immunity that synchronize with feeding rhythms to anticipate microbial exposure. Rhythmic expression of antimicrobial proteins was driven by daily rhythms in epithelial attachment by segmented filamentous bacteria (SFB), members of the mouse intestinal microbiota. Rhythmic SFB attachment was driven by the circadian clock through control of feeding rhythms. Mechanistically, rhythmic SFB attachment activated an immunological circuit involving group 3 innate lymphoid cells. This circuit triggered oscillations in epithelial STAT3 expression and activation that produced rhythmic antimicrobial protein expression and caused resistance to Salmonella Typhimurium infection to vary across the day-night cycle. Thus, host feeding rhythms synchronize with the microbiota to promote rhythms in intestinal innate immunity that anticipate exogenous microbial exposure.


Asunto(s)
Relojes Circadianos/fisiología , Ritmo Circadiano/fisiología , Microbioma Gastrointestinal , Inmunidad Innata , Animales , Péptidos Catiónicos Antimicrobianos/metabolismo , Adhesión Bacteriana , Adhesión Celular , Células Epiteliales/microbiología , Conducta Alimentaria , Intestino Delgado/microbiología , Intestino Delgado/ultraestructura , Linfocitos/metabolismo , Ratones Endogámicos C57BL , Muramidasa/metabolismo , Proteínas Asociadas a Pancreatitis/metabolismo , Factor de Transcripción STAT3/metabolismo , Salmonelosis Animal/microbiología , Transducción de Señal
9.
Cell ; 184(13): 3376-3393.e17, 2021 06 24.
Artículo en Inglés | MEDLINE | ID: mdl-34043940

RESUMEN

We present a global atlas of 4,728 metagenomic samples from mass-transit systems in 60 cities over 3 years, representing the first systematic, worldwide catalog of the urban microbial ecosystem. This atlas provides an annotated, geospatial profile of microbial strains, functional characteristics, antimicrobial resistance (AMR) markers, and genetic elements, including 10,928 viruses, 1,302 bacteria, 2 archaea, and 838,532 CRISPR arrays not found in reference databases. We identified 4,246 known species of urban microorganisms and a consistent set of 31 species found in 97% of samples that were distinct from human commensal organisms. Profiles of AMR genes varied widely in type and density across cities. Cities showed distinct microbial taxonomic signatures that were driven by climate and geographic differences. These results constitute a high-resolution global metagenomic atlas that enables discovery of organisms and genes, highlights potential public health and forensic applications, and provides a culture-independent view of AMR burden in cities.


Asunto(s)
Farmacorresistencia Bacteriana/genética , Metagenómica , Microbiota/genética , Población Urbana , Biodiversidad , Bases de Datos Genéticas , Humanos
10.
Cell ; 184(8): 2053-2067.e18, 2021 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-33794144

RESUMEN

Industrialization has impacted the human gut ecosystem, resulting in altered microbiome composition and diversity. Whether bacterial genomes may also adapt to the industrialization of their host populations remains largely unexplored. Here, we investigate the extent to which the rates and targets of horizontal gene transfer (HGT) vary across thousands of bacterial strains from 15 human populations spanning a range of industrialization. We show that HGTs have accumulated in the microbiome over recent host generations and that HGT occurs at high frequency within individuals. Comparison across human populations reveals that industrialized lifestyles are associated with higher HGT rates and that the functions of HGTs are related to the level of host industrialization. Our results suggest that gut bacteria continuously acquire new functionality based on host lifestyle and that high rates of HGT may be a recent development in human history linked to industrialization.


Asunto(s)
Bacterias/genética , Microbioma Gastrointestinal , Transferencia de Gen Horizontal , Bacterias/clasificación , Bacterias/aislamiento & purificación , ADN Bacteriano/química , ADN Bacteriano/aislamiento & purificación , ADN Bacteriano/metabolismo , Heces/microbiología , Genoma Bacteriano , Humanos , Filogenia , Población Rural , Análisis de Secuencia de ADN , Población Urbana , Secuenciación Completa del Genoma
11.
Cell ; 180(1): 50-63.e12, 2020 01 09.
Artículo en Inglés | MEDLINE | ID: mdl-31923399

RESUMEN

Mucosal barrier immunity is essential for the maintenance of the commensal microflora and combating invasive bacterial infection. Although immune and epithelial cells are thought to be the canonical orchestrators of this complex equilibrium, here, we show that the enteric nervous system (ENS) plays an essential and non-redundant role in governing the antimicrobial protein (AMP) response. Using confocal microscopy and single-molecule fluorescence in situ mRNA hybridization (smFISH) studies, we observed that intestinal neurons produce the pleiotropic cytokine IL-18. Strikingly, deletion of IL-18 from the enteric neurons alone, but not immune or epithelial cells, rendered mice susceptible to invasive Salmonella typhimurium (S.t.) infection. Mechanistically, unbiased RNA sequencing and single-cell sequencing revealed that enteric neuronal IL-18 is specifically required for homeostatic goblet cell AMP production. Together, we show that neuron-derived IL-18 signaling controls tissue-wide intestinal immunity and has profound consequences on the mucosal barrier and invasive bacterial killing.


Asunto(s)
Inmunidad Mucosa/inmunología , Interleucina-18/inmunología , Mucosa Intestinal/inmunología , Animales , Citocinas/inmunología , Sistema Nervioso Entérico/inmunología , Sistema Nervioso Entérico/metabolismo , Células Epiteliales/inmunología , Femenino , Células Caliciformes/inmunología , Interleucina-18/biosíntesis , Mucosa Intestinal/metabolismo , Intestino Delgado/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Neuronas/inmunología , Ratas , Ratas Sprague-Dawley , Infecciones por Salmonella/inmunología , Salmonella typhimurium/inmunología , Transducción de Señal/inmunología
12.
Cell ; 179(2): 459-469.e9, 2019 10 03.
Artículo en Inglés | MEDLINE | ID: mdl-31585083

RESUMEN

The rapid emergence of antibiotic-resistant infections is prompting increased interest in phage-based antimicrobials. However, acquisition of resistance by bacteria is a major issue in the successful development of phage therapies. Through natural evolution and structural modeling, we identified host-range-determining regions (HRDRs) in the T3 phage tail fiber protein and developed a high-throughput strategy to genetically engineer these regions through site-directed mutagenesis. Inspired by antibody specificity engineering, this approach generates deep functional diversity while minimizing disruptions to the overall tail fiber structure, resulting in synthetic "phagebodies." We showed that mutating HRDRs yields phagebodies with altered host-ranges, and select phagebodies enable long-term suppression of bacterial growth in vitro, by preventing resistance appearance, and are functional in vivo using a murine model. We anticipate that this approach may facilitate the creation of next-generation antimicrobials that slow resistance development and could be extended to other viral scaffolds for a broad range of applications.


Asunto(s)
Bacteriófago T3/genética , Infecciones por Escherichia coli/terapia , Escherichia coli/virología , Terapia de Fagos/métodos , Enfermedades Cutáneas Bacterianas/terapia , Proteínas de la Cola de los Virus/genética , Animales , Farmacorresistencia Bacteriana , Especificidad del Huésped , Ratones , Mutagénesis Sitio-Dirigida
13.
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
14.
Cell ; 169(7): 1240-1248.e23, 2017 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-28622509

RESUMEN

Drug-resistant bacterial pathogens pose an urgent public-health crisis. Here, we report the discovery, from microbial-extract screening, of a nucleoside-analog inhibitor that inhibits bacterial RNA polymerase (RNAP) and exhibits antibacterial activity against drug-resistant bacterial pathogens: pseudouridimycin (PUM). PUM is a natural product comprising a formamidinylated, N-hydroxylated Gly-Gln dipeptide conjugated to 6'-amino-pseudouridine. PUM potently and selectively inhibits bacterial RNAP in vitro, inhibits bacterial growth in culture, and clears infection in a mouse model of Streptococcus pyogenes peritonitis. PUM inhibits RNAP through a binding site on RNAP (the NTP addition site) and mechanism (competition with UTP for occupancy of the NTP addition site) that differ from those of the RNAP inhibitor and current antibacterial drug rifampin (Rif). PUM exhibits additive antibacterial activity when co-administered with Rif, exhibits no cross-resistance with Rif, and exhibits a spontaneous resistance rate an order-of-magnitude lower than that of Rif. PUM is a highly promising lead for antibacterial therapy.


Asunto(s)
Antibacterianos/aislamiento & purificación , Antibacterianos/farmacología , ARN Polimerasas Dirigidas por ADN/antagonistas & inhibidores , Streptomyces/química , Animales , Antibacterianos/química , Bacterias/clasificación , Bacterias/efectos de los fármacos , Bacterias/crecimiento & desarrollo , ARN Polimerasas Dirigidas por ADN/química , Farmacorresistencia Bacteriana , Femenino , Células HeLa , Humanos , Ratones , Ratones Endogámicos ICR , Microbiología del Suelo , Infecciones Estreptocócicas/tratamiento farmacológico , Streptococcus pyogenes/efectos de los fármacos , Transcripción Genética/efectos de los fármacos
15.
Annu Rev Biochem ; 85: 765-92, 2016 Jun 02.
Artículo en Inglés | MEDLINE | ID: mdl-27050287

RESUMEN

Neutrophils are essential for killing bacteria and other microorganisms, and they also have a significant role in regulating the inflammatory response. Stimulated neutrophils activate their NADPH oxidase (NOX2) to generate large amounts of superoxide, which acts as a precursor of hydrogen peroxide and other reactive oxygen species that are generated by their heme enzyme myeloperoxidase. When neutrophils engulf bacteria they enclose them in small vesicles (phagosomes) into which superoxide is released by activated NOX2 on the internalized neutrophil membrane. The superoxide dismutates to hydrogen peroxide, which is used by myeloperoxidase to generate other oxidants, including the highly microbicidal species hypochlorous acid. NOX activation occurs at other sites in the cell, where it is considered to have a regulatory function. Neutrophils also release oxidants, which can modify extracellular targets and affect the function of neighboring cells. We discuss the identity and chemical properties of the specific oxidants produced by neutrophils in different situations, and what is known about oxidative mechanisms of microbial killing, inflammatory tissue damage, and signaling.


Asunto(s)
Cloraminas/metabolismo , Peróxido de Hidrógeno/metabolismo , Ácido Hipocloroso/metabolismo , Neutrófilos/inmunología , Superóxidos/metabolismo , Tiocianatos/metabolismo , Membrana Celular/efectos de los fármacos , Células Cultivadas , Cloraminas/inmunología , Expresión Génica , Humanos , Peróxido de Hidrógeno/inmunología , Ácido Hipocloroso/inmunología , Glicoproteínas de Membrana/agonistas , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/inmunología , NADPH Oxidasa 2 , NADPH Oxidasas/genética , NADPH Oxidasas/inmunología , Neutrófilos/citología , Neutrófilos/efectos de los fármacos , Oxidación-Reducción , Peroxidasa/genética , Peroxidasa/inmunología , Transducción de Señal , Superóxidos/inmunología , Acetato de Tetradecanoilforbol/farmacología , Tiocianatos/inmunología , Zimosan/farmacología
16.
Physiol Rev ; 104(4): 1643-1677, 2024 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-39052018

RESUMEN

Representing severe morbidity and mortality globally, respiratory infections associated with chronic respiratory diseases, including complicated pneumonia, asthma, interstitial lung disease, and chronic obstructive pulmonary disease, are a major public health concern. Lung health and the prevention of pulmonary disease rely on the mechanisms of airway surface fluid secretion, mucociliary clearance, and adequate immune response to eradicate inhaled pathogens and particulate matter from the environment. The antimicrobial proteins and peptides contribute to maintaining an antimicrobial milieu in human lungs to eliminate pathogens and prevent them from causing pulmonary diseases. The predominant antimicrobial molecules of the lung environment include human α- and ß-defensins and cathelicidins, among numerous other host defense molecules with antimicrobial and antibiofilm activity such as PLUNC (palate, lung, and nasal epithelium clone) family proteins, elafin, collectins, lactoferrin, lysozymes, mucins, secretory leukocyte proteinase inhibitor, surfactant proteins SP-A and SP-D, and RNases. It has been demonstrated that changes in antimicrobial molecule expression levels are associated with regulating inflammation, potentiating exacerbations, pathological changes, and modifications in chronic lung disease severity. Antimicrobial molecules also display roles in both anticancer and tumorigenic effects. Lung antimicrobial proteins and peptides are promising alternative therapeutics for treating and preventing multidrug-resistant bacterial infections and anticancer therapies.


Asunto(s)
Péptidos Antimicrobianos , Pulmón , Humanos , Animales , Pulmón/inmunología , Pulmón/metabolismo , Enfermedades Pulmonares/tratamiento farmacológico , Enfermedades Pulmonares/inmunología , Enfermedades Pulmonares/metabolismo , Péptidos Catiónicos Antimicrobianos/metabolismo
17.
Annu Rev Genet ; 57: 275-296, 2023 11 27.
Artículo en Inglés | MEDLINE | ID: mdl-37708420

RESUMEN

Antibiotic resistance genes predate the therapeutic uses of antibiotics. However, the current antimicrobial resistance crisis stems from our extensive use of antibiotics and the generation of environmental stressors that impose new selective pressure on microbes and drive the evolution of resistant pathogens that now threaten human health. Similar to climate change, this global threat results from human activities that change habitats and natural microbiomes, which in turn interact with human-associated ecosystems and lead to adverse impacts on human health. Human activities that alter our planet at global scales exacerbate the current resistance crisis and exemplify our central role in large-scale changes in which we are both protagonists and architects of our success but also casualties of unanticipated collateral outcomes. As cognizant participants in this ongoing planetary experiment, we are driven to understand and find strategies to curb the ongoing crises of resistance and climate change.


Asunto(s)
Farmacorresistencia Bacteriana , Microbiota , Humanos , Farmacorresistencia Bacteriana/genética , Antibacterianos/farmacología , Microbiota/genética
18.
Immunity ; 55(9): 1645-1662.e7, 2022 09 13.
Artículo en Inglés | MEDLINE | ID: mdl-35882236

RESUMEN

Healthy skin maintains a diverse microbiome and a potent immune system to fight off infections. Here, we discovered that the epithelial-cell-derived antimicrobial peptides defensins activated orphan G-protein-coupled receptors (GPCRs) Mrgpra2a/b on neutrophils. This signaling axis was required for effective neutrophil-mediated skin immunity and microbiome homeostasis. We generated mutant mouse lines lacking the entire Defensin (Def) gene cluster in keratinocytes or Mrgpra2a/b. Def and Mrgpra2 mutant animals both exhibited skin dysbiosis, with reduced microbial diversity and expansion of Staphylococcus species. Defensins and Mrgpra2 were critical for combating S. aureus infections and the formation of neutrophil abscesses, a hallmark of antibacterial immunity. Activation of Mrgpra2 by defensin triggered neutrophil release of IL-1ß and CXCL2 which are vital for proper amplification and propagation of the antibacterial immune response. This study demonstrated the importance of epithelial-neutrophil signaling via the defensin-Mrgpra2 axis in maintaining healthy skin ecology and promoting antibacterial host defense.


Asunto(s)
Infecciones Bacterianas , Neutrófilos , Receptores Acoplados a Proteínas G , Animales , Ratones , Antibacterianos , Proteínas Portadoras , Defensinas/genética , Disbiosis , Queratinocitos , Receptores Acoplados a Proteínas G/metabolismo , Staphylococcus aureus
19.
Cell ; 167(1): 248-259.e12, 2016 Sep 22.
Artículo en Inglés | MEDLINE | ID: mdl-27662092

RESUMEN

Synthetic biology uses living cells as molecular foundries for the biosynthesis of drugs, therapeutic proteins, and other commodities. However, the need for specialized equipment and refrigeration for production and distribution poses a challenge for the delivery of these technologies to the field and to low-resource areas. Here, we present a portable platform that provides the means for on-site, on-demand manufacturing of therapeutics and biomolecules. This flexible system is based on reaction pellets composed of freeze-dried, cell-free transcription and translation machinery, which can be easily hydrated and utilized for biosynthesis through the addition of DNA encoding the desired output. We demonstrate this approach with the manufacture and functional validation of antimicrobial peptides and vaccines and present combinatorial methods for the production of antibody conjugates and small molecules. This synthetic biology platform resolves important practical limitations in the production and distribution of therapeutics and molecular tools, both to the developed and developing world.


Asunto(s)
Formación de Anticuerpos , Péptidos Catiónicos Antimicrobianos/biosíntesis , Vacunas/biosíntesis , Animales , Péptidos Catiónicos Antimicrobianos/genética , Sistema Libre de Células , Técnicas Químicas Combinatorias , Humanos , Biosíntesis de Proteínas , Biología Sintética , Transcripción Genética , Vacunas/genética
20.
Trends Biochem Sci ; 49(8): 660-662, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38880687

RESUMEN

The dynamics behavior of a protein is essential for its functionality. Here, Doucet et al. demonstrate how the evolutionary analysis of conformational pathways within a protein family serves to identify common core scaffolds that accommodate branch-specific functional regions controlled by flexibility switches, offering a model for evolutionary-dynamics based protein design.


Asunto(s)
Evolución Molecular , Ribonucleasas/metabolismo , Ribonucleasas/química , Ingeniería de Proteínas , Conformación Proteica , Modelos Moleculares
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA