Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
1.
Cell ; 167(4): 915-932, 2016 11 03.
Article in English | MEDLINE | ID: mdl-27814521

ABSTRACT

Neurodevelopment is a complex process governed by both intrinsic and extrinsic signals. While historically studied by researching the brain, inputs from the periphery impact many neurological conditions. Indeed, emerging data suggest communication between the gut and the brain in anxiety, depression, cognition, and autism spectrum disorder (ASD). The development of a healthy, functional brain depends on key pre- and post-natal events that integrate environmental cues, such as molecular signals from the gut. These cues largely originate from the microbiome, the consortium of symbiotic bacteria that reside within all animals. Research over the past few years reveals that the gut microbiome plays a role in basic neurogenerative processes such as the formation of the blood-brain barrier, myelination, neurogenesis, and microglia maturation and also modulates many aspects of animal behavior. Herein, we discuss the biological intersection of neurodevelopment and the microbiome and explore the hypothesis that gut bacteria are integral contributors to development and function of the nervous system and to the balance between mental health and disease.


Subject(s)
Brain/physiology , Gastrointestinal Microbiome , Animals , Behavior , Brain/growth & development , Female , Humans , Neurodevelopmental Disorders/microbiology , Pregnancy , Vagina/microbiology
2.
Cell ; 167(6): 1469-1480.e12, 2016 Dec 01.
Article in English | MEDLINE | ID: mdl-27912057

ABSTRACT

The intestinal microbiota influence neurodevelopment, modulate behavior, and contribute to neurological disorders. However, a functional link between gut bacteria and neurodegenerative diseases remains unexplored. Synucleinopathies are characterized by aggregation of the protein α-synuclein (αSyn), often resulting in motor dysfunction as exemplified by Parkinson's disease (PD). Using mice that overexpress αSyn, we report herein that gut microbiota are required for motor deficits, microglia activation, and αSyn pathology. Antibiotic treatment ameliorates, while microbial re-colonization promotes, pathophysiology in adult animals, suggesting that postnatal signaling between the gut and the brain modulates disease. Indeed, oral administration of specific microbial metabolites to germ-free mice promotes neuroinflammation and motor symptoms. Remarkably, colonization of αSyn-overexpressing mice with microbiota from PD-affected patients enhances physical impairments compared to microbiota transplants from healthy human donors. These findings reveal that gut bacteria regulate movement disorders in mice and suggest that alterations in the human microbiome represent a risk factor for PD.


Subject(s)
Parkinson Disease/microbiology , Parkinson Disease/pathology , Animals , Brain/pathology , Dysbiosis/pathology , Fatty Acids/metabolism , Gastrointestinal Microbiome , Gastrointestinal Tract/microbiology , Gastrointestinal Tract/physiopathology , Humans , Inflammation/metabolism , Inflammation/microbiology , Inflammation/pathology , Mice , Microglia/pathology , Parkinson Disease/metabolism , Parkinson Disease/physiopathology , alpha-Synuclein/metabolism
3.
Nature ; 570(7760): E30-E31, 2019 Jun.
Article in English | MEDLINE | ID: mdl-31127194

ABSTRACT

Change history: We could not replicate the results in Fig. 2a and g of this Letter, and new information has revealed a flaw in the interpretation of Fig. 2h. As a result, we do not have evidence to support RNA degradation as the mechanism that underlies Cas9-mediated regulation of FTN_1103 mRNA expression; see accompanying Amendment. This has not been corrected online.

4.
Semin Neurol ; 43(4): 518-529, 2023 08.
Article in English | MEDLINE | ID: mdl-37562449

ABSTRACT

The human gastrointestinal tract is home to trillions of microorganisms-collectively referred to as the gut microbiome-that maintain a symbiotic relationship with their host. This diverse community of microbes grows and changes as we do, with developmental, lifestyle, and environmental factors all shaping microbiome community structure. Increasing evidence suggests this relationship is bidirectional, with the microbiome also influencing host physiological processes. For example, changes in the gut microbiome have been shown to alter neurodevelopment and have lifelong effects on the brain and behavior. Age-related changes in gut microbiome composition have also been linked to inflammatory changes in the brain, perhaps increasing susceptibility to neurological disease. Indeed, associations between gut dysbiosis and many age-related neurological diseases-including Parkinson's disease, Alzheimer's disease, multiple sclerosis, and amyotrophic lateral sclerosis-have been reported. Further, microbiome manipulation in animal models of disease highlights a potential role for the gut microbiome in disease development and progression. Although much remains unknown, these associations open up an exciting new world of therapeutic targets, potentially allowing for improved quality of life for a wide range of patient populations.


Subject(s)
Gastrointestinal Microbiome , Nervous System Diseases , Parkinson Disease , Animals , Humans , Gastrointestinal Microbiome/physiology , Quality of Life , Brain
5.
J Neurosci ; 38(44): 9414-9422, 2018 10 31.
Article in English | MEDLINE | ID: mdl-30381433

ABSTRACT

The gut microbiota has emerged as a critical player in shaping and modulating brain function and has been shown to influence numerous behaviors, including anxiety and depression-like behaviors, sociability, and cognition. However, the effects of the gut microbiota on specific disorders associated with thalamo-cortico-basal ganglia circuits, ranging from compulsive behavior and addiction to altered sensation and motor output, are only recently being explored. Wholesale depletion and alteration of gut microbial communities in rodent models of disorders, such as Parkinson's disease, autism, and addiction, robustly affect movement and motivated behavior. A new frontier therefore lies in identifying specific microbial alterations that affect these behaviors and understanding the underlying mechanisms of action. Comparing alterations in gut microbiota across multiple basal-ganglia associated disease states allows for identification of common mechanistic pathways that may interact with distinct environmental and genetic risk factors to produce disease-specific outcomes.


Subject(s)
Brain/physiopathology , Dysbiosis/physiopathology , Gastrointestinal Microbiome/physiology , Mental Disorders/physiopathology , Motivation/physiology , Movement/physiology , Animals , Dysbiosis/diagnosis , Dysbiosis/psychology , Humans , Mental Disorders/diagnosis , Mental Disorders/psychology
7.
Nature ; 497(7448): 254-7, 2013 May 09.
Article in English | MEDLINE | ID: mdl-23584588

ABSTRACT

CRISPR/Cas (clustered regularly interspaced palindromic repeats/CRISPR-associated) systems are a bacterial defence against invading foreign nucleic acids derived from bacteriophages or exogenous plasmids. These systems use an array of small CRISPR RNAs (crRNAs) consisting of repetitive sequences flanking unique spacers to recognize their targets, and conserved Cas proteins to mediate target degradation. Recent studies have suggested that these systems may have broader functions in bacterial physiology, and it is unknown if they regulate expression of endogenous genes. Here we demonstrate that the Cas protein Cas9 of Francisella novicida uses a unique, small, CRISPR/Cas-associated RNA (scaRNA) to repress an endogenous transcript encoding a bacterial lipoprotein. As bacterial lipoproteins trigger a proinflammatory innate immune response aimed at combating pathogens, CRISPR/Cas-mediated repression of bacterial lipoprotein expression is critical for F. novicida to dampen this host response and promote virulence. Because Cas9 proteins are highly enriched in pathogenic and commensal bacteria, our work indicates that CRISPR/Cas-mediated gene regulation may broadly contribute to the regulation of endogenous bacterial genes, particularly during the interaction of such bacteria with eukaryotic hosts.


Subject(s)
Gammaproteobacteria/immunology , Gammaproteobacteria/pathogenicity , Immune Evasion , Immunity, Innate/immunology , Animals , Female , Gammaproteobacteria/genetics , Gammaproteobacteria/metabolism , Genes, Bacterial/genetics , Host-Pathogen Interactions/immunology , Mice , Mice, Inbred C57BL , Phylogeny , RNA, Bacterial/genetics , RNA, Bacterial/metabolism , Time Factors , Toll-Like Receptor 2/immunology , Toll-Like Receptor 2/metabolism , Virulence/genetics
8.
Proc Natl Acad Sci U S A ; 112(19): 6164-9, 2015 May 12.
Article in English | MEDLINE | ID: mdl-25918406

ABSTRACT

Clustered, regularly interspaced, short palindromic repeats-CRISPR associated (CRISPR-Cas) systems are prokaryotic RNA-directed endonuclease machineries that act as an adaptive immune system against foreign genetic elements. Using small CRISPR RNAs that provide specificity, Cas proteins recognize and degrade nucleic acids. Our previous work demonstrated that the Cas9 endonuclease from Francisella novicida (FnCas9) is capable of targeting endogenous bacterial RNA. Here, we show that FnCas9 can be directed by an engineered RNA-targeting guide RNA to target and inhibit a human +ssRNA virus, hepatitis C virus, within eukaryotic cells. This work reveals a versatile and portable RNA-targeting system that can effectively function in eukaryotic cells and be programmed as an antiviral defense.


Subject(s)
Bacterial Proteins/metabolism , CRISPR-Associated Proteins/metabolism , Endonucleases/metabolism , Francisella/metabolism , RNA, Viral/chemistry , 3' Untranslated Regions , 5' Untranslated Regions , Base Sequence , CRISPR-Associated Protein 9 , Cell Line, Tumor , DNA/chemistry , Epitopes/chemistry , Hepacivirus , Humans , Molecular Sequence Data , Plasmids/metabolism , Transfection , Viral Proteins/metabolism
9.
Proc Natl Acad Sci U S A ; 111(30): 11163-8, 2014 Jul 29.
Article in English | MEDLINE | ID: mdl-25024199

ABSTRACT

Clustered, regularly interspaced, short palindromic repeats-CRISPR associated (CRISPR-Cas) systems defend bacteria against foreign nucleic acids, such as during bacteriophage infection and transformation, processes which cause envelope stress. It is unclear if these machineries enhance membrane integrity to combat this stress. Here, we show that the Cas9-dependent CRISPR-Cas system of the intracellular bacterial pathogen Francisella novicida is involved in enhancing envelope integrity through the regulation of a bacterial lipoprotein. This action ultimately provides increased resistance to numerous membrane stressors, including antibiotics. We further find that this previously unappreciated function of Cas9 is critical during infection, as it promotes evasion of the host innate immune absent in melanoma 2/apoptosis associated speck-like protein containing a CARD (AIM2/ASC) inflammasome. Interestingly, the attenuation of the cas9 mutant is complemented only in mice lacking both the AIM2/ASC inflammasome and the bacterial lipoprotein sensor Toll-like receptor 2, but not in single knockout mice, demonstrating that Cas9 is essential for evasion of both pathways. These data represent a paradigm shift in our understanding of the function of CRISPR-Cas systems as regulators of bacterial physiology and provide a framework with which to investigate the roles of these systems in myriad bacteria, including pathogens and commensals.


Subject(s)
Bacterial Proteins/immunology , Drug Resistance, Bacterial/immunology , Francisella/immunology , Gram-Negative Bacterial Infections/immunology , Immune Evasion/immunology , Inflammasomes/immunology , Lipoproteins/immunology , Animals , Cell Membrane/genetics , Cell Membrane/immunology , Drug Resistance, Bacterial/genetics , Francisella/genetics , Gram-Negative Bacterial Infections/genetics , Immune Evasion/genetics , Inflammasomes/genetics , Inverted Repeat Sequences/immunology , Lipoproteins/genetics , Mice , Mice, Knockout
10.
Bioessays ; 36(1): 34-8, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24323919

ABSTRACT

The Cas9 endonuclease is the central component of the Type II CRISPR/Cas system, a prokaryotic adaptive restriction system against invading nucleic acids, such as those originating from bacteriophages and plasmids. Recently, this RNA-directed DNA endonuclease has been harnessed to target DNA sequences of interest. Here, we review the development of Cas9 as an important tool to not only edit the genomes of a number of different prokaryotic and eukaryotic species, but also as an efficient system for site-specific transcriptional repression or activation. Additionally, a specific Cas9 protein has been observed to target an RNA substrate, suggesting that Cas9 may have the ability to be programmed to target RNA as well. Cas proteins from other CRISPR/Cas subtypes may also be exploited in this regard. Thus, CRISPR/Cas systems represent an effective and versatile biotechnological tool, which will have significant impact on future advancements in genome engineering.


Subject(s)
Biotechnology/methods , Clustered Regularly Interspaced Short Palindromic Repeats/genetics , Deoxyribonuclease I/genetics , DNA/genetics , Humans , RNA/genetics , Transcription, Genetic/genetics
11.
Curr Opin Infect Dis ; 28(3): 267-74, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25887612

ABSTRACT

PURPOSE OF REVIEW: CRISPR-Cas systems are prokaryotic immune systems against invading nucleic acids that adapt as new environmental threats arise. There are emerging examples of CRISPR-Cas functions in bacterial physiology beyond their role in adaptive immunity. This highlights the poorly understood, but potentially common, moonlighting functions of these abundant systems. We propose that these noncanonical CRISPR-Cas activities have evolved to respond to stresses at the cell envelope. RECENT FINDINGS: Here, we discuss recent literature describing the impact of the extracellular environment on the regulation of CRISPR-Cas systems, and the influence of CRISPR-Cas activity on bacterial physiology. These described noncanonical CRISPR-Cas functions allow the bacterial cell to respond to the extracellular environment, primarily through changes in envelope physiology. SUMMARY: This review discusses the expanding noncanonical functions of CRISPR-Cas systems, including their roles in virulence, focusing mainly on their relationship to the cell envelope. We first examine the effects of the extracellular environment on regulation of CRISPR-Cas components, and then discuss the impact of CRISPR-Cas systems on bacterial physiology, concentrating on their roles in influencing interactions with the environment including host organisms.


Subject(s)
Adaptive Immunity/physiology , Antibodies, Viral/immunology , Bacterial Infections/immunology , CRISPR-Cas Systems/physiology , Viral Envelope Proteins/immunology , Bacterial Infections/genetics , Host-Pathogen Interactions , Humans , Phylogeny , Viral Envelope Proteins/genetics
12.
bioRxiv ; 2024 Jun 08.
Article in English | MEDLINE | ID: mdl-38895207

ABSTRACT

Spinal cord injury (SCI) results in a plethora of physiological dysfunctions across all body systems, including intestinal dysmotility and atrophy of the enteric nervous system (ENS). Typically, the ENS has capacity to recover from perturbation, so it is unclear why intestinal pathophysiologies persist after traumatic spinal injury. With emerging evidence demonstrating SCI-induced alterations to the gut microbiome composition, we hypothesized that modulation of the gut microbiome could contribute to enteric nervous system recovery after injury. Here, we show that intervention with the dietary fiber, inulin prevents ENS atrophy and limits SCI-induced intestinal dysmotility in mice. However, SCI-associated microbiomes and exposure to specific SCI-sensitive gut microbes are not sufficient to modulate injury-induced intestinal dysmotility. Intervention with microbially-derived short-chain fatty acid (SCFA) metabolites prevents ENS dysfunctions and phenocopies inulin treatment in injured mice, implicating these microbiome metabolites in protection of the ENS. Notably, inulin-mediated resilience is dependent on signaling by the cytokine IL-10, highlighting a critical diet-microbiome-immune axis that promotes ENS resilience following SCI. Overall, we demonstrate that diet and microbially-derived signals distinctly impact recovery of the ENS after traumatic spinal injury. This protective diet-microbiome-immune axis may represent a foundation to uncover etiological mechanisms and future therapeutics for SCI-induced neurogenic bowel.

13.
Biochem Soc Trans ; 41(6): 1407-11, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24256228

ABSTRACT

CRISPR (clustered regularly interspaced short palindromic repeats)-Cas (CRISPR-associated) systems are known to mediate bacterial defence against foreign nucleic acids. We recently demonstrated a non-canonical role for a CRISPR-Cas system in controlling endogenous gene expression, which had not previously been appreciated. In the present article, we describe the studies that led to this discovery, beginning with an unbiased genome-wide screen to identify virulence genes in the intracellular pathogen Francisella novicida. A gene annotated as encoding a hypothetical protein, but which we now know encodes the Cas protein Cas9, was identified as one of the most critical to the ability of F. novicida to replicate and survive during murine infection. Subsequent studies revealed a role for this protein in evasion of the host innate immune response. Specifically, Cas9 represses the expression of a BLP (bacterial lipoprotein) that could otherwise be recognized by TLR2 (Toll-like receptor 2), a host protein involved in initiating an antibacterial pro-inflammatory response. By repressing BLP levels, Cas9 mediates evasion of TLR2, promoting bacterial virulence. Finally, we described the molecular mechanism by which Cas9 functions in complex with two small RNAs to target the mRNA encoding the BLP for degradation. This work greatly broadened the paradigm for CRISPR-Cas function, highlighting a role in gene regulation that could be conserved in numerous bacteria, and elucidating its integral contribution to bacterial pathogenesis.


Subject(s)
CRISPR-Associated Proteins/genetics , CRISPR-Associated Proteins/metabolism , CRISPR-Cas Systems/genetics , Francisella/genetics , Francisella/pathogenicity , Gene Expression Regulation, Bacterial/genetics , Virulence/genetics
14.
Cell Microbiol ; 14(10): 1531-43, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22632124

ABSTRACT

Innate recognition systems, including the Toll-like receptors (TLRs), play a critical role in activating host defences and proinflammatory pathways in response to infection. Pathogens have developed strategies to subvert TLRs in order to survive and replicate within the host. The model intracellular pathogen, Francisella novicida, modulates host defences to promote survival and replication in macrophages. TLR2, which recognizes bacterial lipoproteins (BLPs), is critical for activating host defences and proinflammatory cytokine production in response to Francisella infection. Here we show that the F. novicida protein FTN_0757 acts to repress BLP production, dampening TLR2 activation. The ΔFTN_0757 mutant strain induced robust TLR2-dependent cytokine production in macrophages compared with wild-type bacteria, and produced increased amounts of BLPs. The deletion of one BLP (FTN_1103) from ΔFTN_0757 decreased the total BLP concentration to near wild-type level sand correlated with a decrease in the inductionof TLR2 signalling. The overproduction of BLPs also contributed to the in vivo attenuation of the ΔFTN_0757 mutant, which was significantly rescued when FTN_1103 was deleted. Taken together, these data reveal a novel mechanism of immune evasion by the downregulation of BLP expression to subvert TLR2 activation, which is likely used by numerous other intracellular bacterial pathogens.


Subject(s)
Francisella tularensis/immunology , Francisella tularensis/pathogenicity , Immune Evasion , Lipoproteins/biosynthesis , Macrophages/immunology , Macrophages/microbiology , Repressor Proteins/metabolism , Animals , Cells, Cultured , Cytokines/metabolism , Francisella tularensis/genetics , Gene Deletion , Lipoproteins/antagonists & inhibitors , Lipoproteins/genetics , Mice , Repressor Proteins/genetics , Toll-Like Receptor 2/metabolism , Virulence
15.
RNA Biol ; 10(10): 1618-22, 2013 Oct.
Article in English | MEDLINE | ID: mdl-24100224

ABSTRACT

CRISPR/Cas systems are bacterial RNA-guided endonuclease machineries that target foreign nucleic acids. Recently, we demonstrated that the Cas protein Cas9 controls gene expression and virulence in Francisella novicida by altering the stability of the mRNA for an immunostimulatory bacterial lipoprotein (BLP). Genomic analyses, however, revealed that Francisella species with increased virulence harbor degenerated CRISPR/Cas systems. We hypothesize that CRISPR/Cas degeneration removed a barrier against genome alterations, which resulted in enhanced virulence. Importantly, the BLP locus was also lost; likely a necessary adaptation in the absence of Cas9-mediated repression. CRISPR/Cas systems likely play regulatory roles in numerous bacteria, and these data suggest additional genomic changes may be required to maintain fitness after CRISPR/Cas loss in such bacteria, having important evolutionary implications.


Subject(s)
Biological Evolution , CRISPR-Cas Systems , Francisella tularensis/genetics , Francisella tularensis/pathogenicity , Francisella/pathogenicity , Amino Acid Sequence , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Base Sequence , Francisella/classification , Francisella/genetics , Gene Transfer, Horizontal , Lipoproteins/chemistry , Lipoproteins/genetics , Sequence Alignment , Virulence
16.
Cell Host Microbe ; 31(2): 171-172, 2023 02 08.
Article in English | MEDLINE | ID: mdl-36758517

ABSTRACT

The etiology of chronic fatigue syndrome (CFS) is largely unknown. In this issue of Cell Host and Microbe, Guo et al. and Xiong et al. report CFS-associated gut microbiome and metabolomic datasets-implicating dysregulation of immune modulating molecules. This may provide a framework for new therapeutic paradigms and disease origins.


Subject(s)
Fatigue Syndrome, Chronic , Gastrointestinal Microbiome , Humans
17.
NPJ Parkinsons Dis ; 9(1): 159, 2023 Dec 05.
Article in English | MEDLINE | ID: mdl-38052871

ABSTRACT

Parkinson's disease is the fastest-growing neurologic disease with seemingly no means of prevention. Intrinsic risk factors (age, sex, and genetics) are inescapable, but environmental factors are not. We identified repeated blows to the head in sports/combat as a potential new risk factor. 23% of PD cases in females were attributable to pesticide/herbicide exposure, and 30% of PD in males were attributable to pesticides/herbicides, military-related chemical exposures, and repeated blows to the head, and therefore could have potentially been prevented.

18.
medRxiv ; 2023 May 19.
Article in English | MEDLINE | ID: mdl-37292848

ABSTRACT

Parkinson's disease is the fastest growing neurologic disease with seemingly no means for prevention. Intrinsic risk factors (age, sex, genetics) are inescapable, but environmental factors are not. We studied population attributable fraction and estimated fraction of PD that could be reduced if modifiable risk factors were eliminated. Assessing several known risk factors simultaneously in one study, we demonstrate that all were operative and independent, underscoring etiological heterogeneity within a single population. We investigated repeated blows to head in sports or combat as a potential new risk factor, and found it was associated with two-fold increased risk of PD. Considering modifiable risk factors, 23% of PD cases in females were attributable to pesticides/herbicides exposure, and 30% of PD cases in males was attributable to pesticides/herbicides, Agent Orange/chemical warfare, and repeated blows to the head. Thus, one-in-three cases of PD in males, and one-in-four cases in females could have potentially been prevented.

19.
Front Cell Neurosci ; 17: 895017, 2023.
Article in English | MEDLINE | ID: mdl-37006470

ABSTRACT

Introduction: Increasing evidence indicates that neurodegenerative diseases, including Alzheimer's disease (AD), are a product of gene-by-environment interplay. The immune system is a major contributor mediating these interactions. Signaling between peripheral immune cells and those within the microvasculature and meninges of the central nervous system (CNS), at the blood-brain barrier, and in the gut likely plays an important role in AD. The cytokine tumor necrosis factor (TNF) is elevated in AD patients, regulates brain and gut barrier permeability, and is produced by central and peripheral immune cells. Our group previously reported that soluble TNF (sTNF) modulates cytokine and chemokine cascades that regulate peripheral immune cell traffic to the brain in young 5xFAD female mice, and in separate studies that a diet high in fat and sugar (HFHS) dysregulates signaling pathways that trigger sTNF-dependent immune and metabolic responses that can result in metabolic syndrome, which is a risk factor for AD. We hypothesized that sTNF is a key mediator of peripheral immune cell contributions to gene-by-environment interactions to AD-like pathology, metabolic dysfunction, and diet-induced gut dysbiosis. Methods: Female 5xFAD mice were subjected to HFHS diet for 2 months and then given XPro1595 to inhibit sTNF for the last month or saline vehicle. We quantified immune cell profiles by multi-color flow cytometry on cells isolated from brain and blood; metabolic, immune, and inflammatory mRNA and protein marker biochemical and immunhistological analyses, gut microbiome, and electrophysiology in brain slices were also performed. Results: Here, we show that selective inhibition of sTNF signaling via the biologic XPro1595 modulates the effects of an HFHS diet in 5xFAD mice on peripheral and central immune profiles including CNS-associated CD8+ T cells, the composition of gut microbiota, and long-term potentiation deficits. Discussion: Obesogenic diet induces immune and neuronal dysfunction in 5xFAD mice and sTNF inhibition mitigates its effects. A clinical trial in subjects at risk for AD due to genetic predisposition and underlying inflammation associated with peripheral inflammatory co-morbidities will be needed to investigate the extent to which these findings translate to the clinic.

20.
Antimicrob Agents Chemother ; 56(11): 5642-9, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22908157

ABSTRACT

Acinetobacter baumannii is an opportunistic pathogen that is a cause of clinically significant nosocomial infections. Increasingly, clinical isolates of A. baumannii are extensively resistant to numerous antibiotics, and the use of polymyxin antibiotics against these infections is often the final treatment option. Historically, the polymyxins have been thought to kill bacteria through membrane lysis. Here, we present an alternative mechanism based on data demonstrating that polymyxins induce rapid cell death through hydroxyl radical production. Supporting this notion, we found that inhibition of radical production delays the ability of polymyxins to kill A. baumannii. Notably, we demonstrate that this mechanism of killing occurs in multidrug-resistant clinical isolates of A. baumannii and that this response is not induced in a polymyxin-resistant isolate. This study is the first to demonstrate that polymyxins induce rapid killing of A. baumannii and other Gram-negatives through hydroxyl radical production. This significantly augments our understanding of the mechanism of polymyxin action, which is critical knowledge toward the development of adjunctive therapies, particularly given the increasing necessity for treatment with these antibiotics in the clinical setting.


Subject(s)
Acinetobacter baumannii/drug effects , Anti-Bacterial Agents/pharmacology , Colistin/pharmacology , Hydroxyl Radical/metabolism , Polymyxin B/pharmacology , Acinetobacter Infections/drug therapy , Acinetobacter Infections/microbiology , Acinetobacter baumannii/growth & development , Acinetobacter baumannii/metabolism , Cross Infection/drug therapy , Cross Infection/microbiology , Culture Media , Drug Resistance, Multiple, Bacterial , Free Radical Scavengers/pharmacology , Humans , Hydroxyl Radical/antagonists & inhibitors , Microbial Sensitivity Tests , Microbial Viability/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL