Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
1.
Cell ; 180(1): 33-49.e22, 2020 01 09.
Article in English | MEDLINE | ID: mdl-31813624

ABSTRACT

Gut-innervating nociceptor sensory neurons respond to noxious stimuli by initiating protective responses including pain and inflammation; however, their role in enteric infections is unclear. Here, we find that nociceptor neurons critically mediate host defense against the bacterial pathogen Salmonella enterica serovar Typhimurium (STm). Dorsal root ganglia nociceptors protect against STm colonization, invasion, and dissemination from the gut. Nociceptors regulate the density of microfold (M) cells in ileum Peyer's patch (PP) follicle-associated epithelia (FAE) to limit entry points for STm invasion. Downstream of M cells, nociceptors maintain levels of segmentous filamentous bacteria (SFB), a gut microbe residing on ileum villi and PP FAE that mediates resistance to STm infection. TRPV1+ nociceptors directly respond to STm by releasing calcitonin gene-related peptide (CGRP), a neuropeptide that modulates M cells and SFB levels to protect against Salmonella infection. These findings reveal a major role for nociceptor neurons in sensing and defending against enteric pathogens.


Subject(s)
Gastrointestinal Microbiome/physiology , Host Microbial Interactions/physiology , Nociceptors/physiology , Animals , Epithelium/metabolism , Female , Ganglia, Spinal/metabolism , Ganglia, Spinal/microbiology , Intestinal Mucosa/microbiology , Male , Mice , Mice, Inbred C57BL , Nociceptors/metabolism , Peyer's Patches/innervation , Peyer's Patches/metabolism , Salmonella Infections/metabolism , Salmonella typhimurium/metabolism , Salmonella typhimurium/pathogenicity , Sensory Receptor Cells/metabolism , Sensory Receptor Cells/physiology
2.
Nature ; 577(7792): 676-681, 2020 01.
Article in English | MEDLINE | ID: mdl-31969699

ABSTRACT

Empirical and anecdotal evidence has associated stress with accelerated hair greying (formation of unpigmented hairs)1,2, but so far there has been little scientific validation of this link. Here we report that, in mice, acute stress leads to hair greying through the fast depletion of melanocyte stem cells. Using a combination of adrenalectomy, denervation, chemogenetics3,4, cell ablation and knockout of the adrenergic receptor specifically in melanocyte stem cells, we find that the stress-induced loss of melanocyte stem cells is independent of immune attack or adrenal stress hormones. Instead, hair greying results from activation of the sympathetic nerves that innervate the melanocyte stem-cell niche. Under conditions of stress, the activation of these sympathetic nerves leads to burst release of the neurotransmitter noradrenaline (also known as norepinephrine). This causes quiescent melanocyte stem cells to proliferate rapidly, and is followed by their differentiation, migration and permanent depletion from the niche. Transient suppression of the proliferation of melanocyte stem cells prevents stress-induced hair greying. Our study demonstrates that neuronal activity that is induced by acute stress can drive a rapid and permanent loss of somatic stem cells, and illustrates an example in which the maintenance of somatic stem cells is directly influenced by the overall physiological state of the organism.


Subject(s)
Autonomic Pathways/physiopathology , Hair Color/physiology , Melanocytes/pathology , Stem Cell Niche/physiology , Stem Cells/pathology , Stress, Psychological/physiopathology , Sympathetic Nervous System/physiopathology , Adrenal Glands/metabolism , Adrenalectomy , Animals , Autonomic Pathways/pathology , Cell Proliferation , Cells, Cultured , Denervation , Female , Humans , Male , Melanocytes/cytology , Melanocytes/metabolism , Mice , Norepinephrine/metabolism , Psychological Trauma/pathology , Psychological Trauma/physiopathology , Receptors, Adrenergic, beta-2/deficiency , Receptors, Adrenergic, beta-2/metabolism , Stem Cells/cytology , Stem Cells/metabolism , Stress, Psychological/pathology , Sympathetic Nervous System/pathology
3.
PLoS Pathog ; 17(5): e1009557, 2021 05.
Article in English | MEDLINE | ID: mdl-33956874

ABSTRACT

We report a rapid reduction in blink reflexes during in vivo ocular Pseudomonas aeruginosa infection, which is commonly attributed and indicative of functional neuronal damage. Sensory neurons derived in vitro from trigeminal ganglia (TG) were able to directly respond to P. aeruginosa but reacted significantly less to strains of P. aeruginosa that lacked virulence factors such as pili, flagella, or a type III secretion system. These observations led us to explore the impact of neurons on the host's susceptibility to P. aeruginosa keratitis. Mice were treated with Resiniferatoxin (RTX), a potent activator of Transient Receptor Potential Vanilloid 1 (TRPV1) channels, which significantly ablated corneal sensory neurons, exhibited delayed disease progression that was exemplified with decreased bacterial corneal burdens and altered neutrophil trafficking. Sensitization to disease was due to the increased frequencies of CGRP-induced ICAM-1+ neutrophils in the infected corneas and reduced neutrophil bactericidal activities. These data showed that sensory neurons regulate corneal neutrophil responses in a tissue-specific matter affecting disease progression during P. aeruginosa keratitis. Hence, therapeutic modalities that control nociception could beneficially impact anti-infective therapy.


Subject(s)
Disease Models, Animal , Keratitis/pathology , Neutrophils/immunology , Nociceptors/metabolism , Pseudomonas Infections/complications , Pseudomonas aeruginosa/physiology , Trigeminal Nerve Diseases/pathology , Animals , Female , Keratitis/etiology , Keratitis/metabolism , Male , Mice , Mice, Inbred C57BL , Trigeminal Nerve Diseases/etiology , Trigeminal Nerve Diseases/metabolism
4.
Nature ; 549(7672): 351-356, 2017 09 21.
Article in English | MEDLINE | ID: mdl-28902842

ABSTRACT

Type 2 innate lymphoid cells (ILC2s) both contribute to mucosal homeostasis and initiate pathologic inflammation in allergic asthma. However, the signals that direct ILC2s to promote homeostasis versus inflammation are unclear. To identify such molecular cues, we profiled mouse lung-resident ILCs using single-cell RNA sequencing at steady state and after in vivo stimulation with the alarmin cytokines IL-25 and IL-33. ILC2s were transcriptionally heterogeneous after activation, with subpopulations distinguished by expression of proliferative, homeostatic and effector genes. The neuropeptide receptor Nmur1 was preferentially expressed by ILC2s at steady state and after IL-25 stimulation. Neuromedin U (NMU), the ligand of NMUR1, activated ILC2s in vitro, and in vivo co-administration of NMU with IL-25 strongly amplified allergic inflammation. Loss of NMU-NMUR1 signalling reduced ILC2 frequency and effector function, and altered transcriptional programs following allergen challenge in vivo. Thus, NMUR1 signalling promotes inflammatory ILC2 responses, highlighting the importance of neuro-immune crosstalk in allergic inflammation at mucosal surfaces.


Subject(s)
Hypersensitivity/immunology , Hypersensitivity/pathology , Inflammation/immunology , Inflammation/pathology , Lung/pathology , Lymphocytes/immunology , Neuropeptides/metabolism , Animals , Female , Gene Expression Regulation , Immunity, Innate/immunology , Interleukin-17/immunology , Interleukin-33/immunology , Ligands , Lung/immunology , Male , Mice , Mice, Inbred C57BL , Receptors, Neurotransmitter/biosynthesis , Receptors, Neurotransmitter/genetics , Receptors, Neurotransmitter/metabolism , Respiratory Mucosa/immunology , Respiratory Mucosa/pathology , Signal Transduction , Transcription, Genetic
6.
Blood ; 133(12): 1335-1345, 2019 03 21.
Article in English | MEDLINE | ID: mdl-30723078

ABSTRACT

Neutrophil migration to the site of bacterial infection is a critical step in host defense. Exclusively produced in the bone marrow, neutrophil release into the blood is tightly controlled. Although the chemokine CXCL1 induces neutrophil influx during bacterial infections, its role in regulating neutrophil recruitment, granulopoiesis, and neutrophil mobilization in response to lung infection-induced sepsis is unclear. Here, we used a murine model of intrapulmonary Streptococcus pneumoniae infection to investigate the role of CXCL1 in host defense, granulopoiesis, and neutrophil mobilization. Our results demonstrate that CXCL1 augments neutrophil influx to control bacterial growth in the lungs, as well as bacterial dissemination, resulting in improved host survival. This was shown in Cxcl1 -/- mice, which exhibited defective amplification of early neutrophil precursors in granulocytic compartments, and CD62L- and CD49d-dependent neutrophil release from the marrow. Administration of recombinant CXCL2 and CXCL5 after infection rescues the impairments in neutrophil-dependent host defense in Cxcl1 -/- mice. Taken together, these findings identify CXCL1 as a central player in host defense, granulopoiesis, and mobilization of neutrophils during Gram-positive bacterial pneumonia-induced sepsis.


Subject(s)
Chemokine CXCL1/physiology , Homeostasis , Lung/immunology , Neutrophil Infiltration/immunology , Pneumococcal Infections/complications , Pneumonia, Bacterial/complications , Sepsis/immunology , Animals , Chemokine CXCL2/genetics , Chemokine CXCL2/metabolism , Chemokine CXCL5/genetics , Chemokine CXCL5/metabolism , Female , Lung/microbiology , Lung/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Pneumococcal Infections/microbiology , Pneumococcal Infections/pathology , Pneumonia, Bacterial/microbiology , Pneumonia, Bacterial/pathology , Sepsis/metabolism , Sepsis/microbiology , Serogroup , Streptococcus pneumoniae/physiology
7.
PLoS Pathog ; 14(9): e1007308, 2018 09.
Article in English | MEDLINE | ID: mdl-30248149

ABSTRACT

Gram-positive bacteria, including Staphylococcus aureus are endemic in the U.S., which cause life-threatening necrotizing pneumonia. Neutrophils are known to be critical for clearance of S. aureus infection from the lungs and extrapulmonary organs. Therefore, we investigated whether the NLRP6 inflammasome regulates neutrophil-dependent host immunity during pulmonary S. aureus infection. Unlike their wild-type (WT) counterparts, NLRP6 knockout (KO) mice were protected against pulmonary S. aureus infection as evidenced by their higher survival rate and lower bacterial burden in the lungs and extrapulmonary organs. In addition, NLRP6 KO mice displayed increased neutrophil recruitment following infection, and when neutrophils were depleted the protective effect was lost. Furthermore, neutrophils from the KO mice demonstrated enhanced intracellular bacterial killing and increased NADPH oxidase-dependent ROS production. Intriguingly, we found higher NK cell-mediated IFN-γ production in KO mouse lungs, and treatment with IFN-γ was found to enhance the bactericidal ability of WT and KO neutrophils. The NLRP6 KO mice also displayed decreased pyroptosis and necroptosis in the lungs following infection. Blocking of pyroptosis and necroptosis in WT mice resulted in increased survival, reduced bacterial burden in the lungs, and attenuated cytokine production. Taken together, these novel findings show that NLRP6 serves as a negative regulator of neutrophil-mediated host defense during Gram-positive bacterial infection in the lungs through regulating both neutrophil influx and function. These results also suggest that blocking NLRP6 to augment neutrophil-associated bacterial clearance should be considered as a potential therapeutic intervention strategy for treatment of S. aureus pneumonia.


Subject(s)
Neutrophil Infiltration/immunology , Pneumonia, Staphylococcal/immunology , Receptors, Cell Surface/immunology , Animals , CD4-Positive T-Lymphocytes/immunology , Female , Host-Pathogen Interactions/immunology , Humans , Inflammasomes/immunology , Interferon-gamma/biosynthesis , Killer Cells, Natural/immunology , Lung/immunology , Lung/microbiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Pneumonia, Necrotizing/immunology , Pneumonia, Necrotizing/microbiology , Pneumonia, Staphylococcal/microbiology , Pyroptosis/immunology , Reactive Oxygen Species/metabolism , Receptors, Cell Surface/deficiency , Receptors, Cell Surface/genetics , Staphylococcus aureus/immunology , Up-Regulation
8.
BMC Anesthesiol ; 19(1): 152, 2019 08 14.
Article in English | MEDLINE | ID: mdl-31412783

ABSTRACT

BACKGROUND: Respiratory adverse events are not uncommon in the post-anesthesia care unit (PACU) following general anesthesia. In this regard, hyperventilation leading to apnea and desaturation is a rare entity. Here we have reported a case of a 15-year-old girl who, following an uneventful general anesthesia, developed severe hyperventilation leading to apnea and desaturation in the PACU. CASE PRESENTATION: The 15-year-old girl underwent cortical mastoidectomy under general anesthesia. After a smooth anesthesia and an uneventful early recovery, she developed hyperventilation after about 15 min in the PACU. The symptom was severe enough to lead to apnea, desaturation and severe respiratory alkalosis. She required bag and mask ventilation and the symptoms resolved only transiently with propofol sedation. Finally, she responded to intravenous haloperidol and did not have any further episode after receiving haloperidol. CONCLUSION: Hyperventilation after a smooth recovery from anesthesia is not a common presentation. In this article we have tried to discuss the possible cause of such symptom in our patient and how we successfully managed this case. We have also proposed an algorithmic approach to diagnose and manage such cases in the PACU.


Subject(s)
Anesthesia Recovery Period , Apnea/complications , Hyperventilation/complications , Adolescent , Anesthesia, General/adverse effects , Female , Haloperidol/therapeutic use , Humans , Oxygen/blood , Postoperative Complications/drug therapy
9.
Am J Respir Cell Mol Biol ; 55(4): 586-601, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27253086

ABSTRACT

Cigarette smoke (CS) predisposes exposed individuals to respiratory infections not only by suppressing immune response but also by enhancing the virulence of pathogenic bacteria. As per our observations, in methicillin-resistant Staphylococcus aureus strain USA300, CS extract (CSE) potentiates biofilm formation via the down-regulation of quorum-sensing regulon accessory gene regulator. Because accessory gene regulator is a global regulator of the staphylococcal virulome, in the present study we sought to identify the effects of CS exposure on staphylococcal gene expression using RNAseq. Comparative analysis of RNAseq profiles revealed the up-regulation of important virulence genes encoding surface adhesins (fibronectin- and fibrinogen-binding proteins A and B and clumping factor B) and proteins involved in immune evasion, such as staphylocoagulase, staphylococcal protein A, and nuclease. In concurrence with the RNAseq data, we observed: (1) significant up-regulation of the ability of CSE-exposed USA300 to evade phagocytosis by macrophages and neutrophils, a known function of staphylococcal protein A; and (2) twofold higher (P < 0.001) number of CSE-exposed USA300 escaping neutrophil extracellular trap-mediated killing by neutrophils as a result of CS-mediated induction of nuclease. Importantly, in three different mouse strains, C57BL6/J, Balb/C, and A/J, we observed significantly higher pulmonary bacterial burden in animals infected with CSE-exposed USA300 as compared with medium-exposed control USA300. Taken together, these observations indicate that bioactive chemicals in CS induce hypervirulence by augmenting the ability of USA300 to evade bactericidal functions of leukocytes, such as phagocytosis and neutrophil extracellular trap-mediated killing.

10.
Am J Respir Crit Care Med ; 190(7): 722-32, 2014 Oct 01.
Article in English | MEDLINE | ID: mdl-25033332

ABSTRACT

Lower respiratory tract infections caused by bacteria are a major cause of death in humans irrespective of sex, race, or geography. Indeed, accumulated data indicate greater mortality and morbidity due to these infections than cancer, malaria, or HIV infection. Successful recognition of, followed by an appropriate response to, bacterial pathogens in the lungs is crucial for effective pulmonary host defense. Although the early recruitment and activation of neutrophils in the lungs is key in the response against invading microbial pathogens, other sentinels, such as alveolar macrophages, epithelial cells, dendritic cells, and CD4(+) T cells, also contribute to the elimination of the bacterial burden. Pattern recognition receptors, such as Toll-like receptors (TLRs) and nucleotide-binding oligomerization domain-like receptors, are important for recognizing and responding to microbes during pulmonary infections. However, bacterial pathogens have acquired crafty evasive strategies to circumvent the pattern recognition receptor response and thus establish infection. Increased understanding of the function of TLRs and evasive mechanisms used by pathogens during pulmonary infection will deepen our knowledge of immunopathogenesis and is crucial for developing effective therapeutic and/or prophylactic measures. This review summarizes current knowledge of the multiple roles of TLRs in bacterial lung infections and highlights the mechanisms used by pathogens to modulate or interfere with TLR signaling in the lungs.


Subject(s)
Bacterial Infections/immunology , Lung/immunology , Respiratory Tract Infections/immunology , Toll-Like Receptors/immunology , Animals , Humans , Signal Transduction/immunology
11.
Infect Immun ; 81(9): 3463-71, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23836818

ABSTRACT

Burkholderia pseudomallei, the causative agent of melioidosis, evades macrophage killing by suppressing the TRIF-dependent pathway, leading to inhibition of inducible nitric oxide synthase (iNOS) expression. We previously demonstrated that virulent wild-type B. pseudomallei inhibits the TRIF-dependent pathway by upregulating sterile-α- and armadillo motif-containing protein (SARM) and by inhibiting downregulation of signal regulatory protein α (SIRPα); both molecules are negative regulators of Toll-like receptor signaling. In contrast, the less virulent lipopolysaccharide (LPS) mutant of B. pseudomallei is unable to exhibit these features and is susceptible to macrophage killing. However, the functional relationship of these two negative regulators in the evasion of macrophage defense has not been elucidated. We demonstrated here that SIRPα downregulation was observed after inhibition of SARM expression by small interfering RNA in wild-type-infected macrophages, indicating that SIRPα downregulation is regulated by SARM. Furthermore, this downregulation requires activation of the TRIF signaling pathway, as we observed abrogation of SIRPα downregulation as well as restricted bacterial growth in LPS mutant-infected TRIF-depleted macrophages. Although inhibition of SARM expression is correlated to SIRPα downregulation and iNOS upregulation in gamma interferon-activated wild-type-infected macrophages, these phenomena appear to bypass the TRIF-dependent pathway. Similar to live bacteria, the wild-type LPS is able to upregulate SARM and to prevent SIRPα downregulation, implying that the LPS of B. pseudomallei may play a crucial role in regulating the expression of these two negative regulators. Altogether, our findings show a previously unrecognized role of B. pseudomallei-induced SARM in inhibiting SIRPα downregulation-mediated iNOS upregulation, facilitating the ability of the bacterium to multiply in macrophages.


Subject(s)
Adaptor Proteins, Vesicular Transport/genetics , Armadillo Domain Proteins/genetics , Burkholderia pseudomallei/genetics , Cytoskeletal Proteins/genetics , Macrophages/metabolism , Melioidosis/genetics , Receptors, Immunologic/genetics , Adaptor Proteins, Vesicular Transport/immunology , Adaptor Proteins, Vesicular Transport/metabolism , Animals , Armadillo Domain Proteins/immunology , Armadillo Domain Proteins/metabolism , Burkholderia pseudomallei/immunology , Burkholderia pseudomallei/metabolism , Cytoskeletal Proteins/immunology , Cytoskeletal Proteins/metabolism , Down-Regulation/immunology , Interferon-gamma/genetics , Interferon-gamma/immunology , Interferon-gamma/metabolism , Lipopolysaccharides/immunology , Macrophages/immunology , Macrophages/microbiology , Melioidosis/immunology , Melioidosis/metabolism , Melioidosis/microbiology , Mice , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/immunology , Nitric Oxide Synthase Type II/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/immunology , Receptors, Immunologic/immunology , Receptors, Immunologic/metabolism , Signal Transduction/genetics , Signal Transduction/immunology , Up-Regulation/genetics , Up-Regulation/immunology
12.
STAR Protoc ; 4(3): 102434, 2023 Sep 15.
Article in English | MEDLINE | ID: mdl-37432860

ABSTRACT

Respiratory syncytial virus (RSV) infection in infants and toddlers is a major public health problem. Here, we provide a protocol for neonatal RSV infection in mice and immune analysis of infected lungs and bronchoalveolar lavage (BAL) fluid. We describe steps for anesthesia and intranasal inoculation, weight monitoring, and whole lung collection. We then detail BAL fluid immune and whole lung analyses. This protocol can be used for neonatal pulmonary infection with other viruses or bacteria.


Subject(s)
Respiratory Syncytial Virus Infections , Humans , Infant , Animals , Mice , Bronchoalveolar Lavage Fluid , Respiratory Syncytial Viruses , Lung
13.
Infect Immun ; 80(12): 4223-31, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22988019

ABSTRACT

The facultative intracellular gram-negative bacterium Burkholderia pseudomallei is the causative agent of melioidosis and is known for its ability to evade the Toll-like receptor (TLR)-mediated innate immune response. Previously it has been demonstrated that this bacterium was able to suppress the MyD88-independent pathway and can survive macrophage intracellular killing. However, the underlying mechanisms responsible for the suppression of this pathway are not fully understood. In the present study, we showed that both living and heat-killed B. pseudomallei bacteria restrict the TLR signaling response, particularly macrophage inducible nitric oxide synthase (iNOS) expression, by preventing downregulation of constitutively expressed signal regulatory protein α (SIRPα) molecule, a known negative regulator of TLR signaling. In contrast, a lipopolysaccharide (LPS) mutant of B. pseudomallei, a less virulent strain, was able to downregulate SIRPα expression in mouse macrophages. However, depletion of constitutively expressed SIRPα was able to induce the gene expression downstream of TLR signaling pathways (particularly the MyD88-independent pathway), such as that of the iNOS gene, leading to enhanced macrophage intracellular killing of B. pseudomallei. Induction of gene expression was consistent with the enhanced degradation pattern of IκBα with SIRPα depletion. Additionally, the downregulation of SIRPα expression with upregulation of iNOS was observed when the macrophages were pretreated with gamma interferon (IFN-γ) prior to the infection, suggesting that the enhanced intracellular killing of bacteria by IFN-γ is associated with the decreased SIRPα expression. Altogether our findings demonstrate that B. pseudomallei evades macrophage intracellular killing by preventing the downregulation of SIRPα that results in the inhibition of gene expression downstream of the MyD88-independent pathway.


Subject(s)
Burkholderia pseudomallei/pathogenicity , Gene Expression Regulation/immunology , Macrophages , Melioidosis/immunology , Myeloid Differentiation Factor 88/metabolism , Receptors, Immunologic/metabolism , Animals , Burkholderia pseudomallei/growth & development , Cell Line , Gene Expression Regulation/drug effects , Immune Evasion , Macrophages/immunology , Macrophages/microbiology , Melioidosis/microbiology , Mice , Myeloid Differentiation Factor 88/genetics , Receptors, Immunologic/genetics , Signal Transduction , Toll-Like Receptors/metabolism
14.
Nat Rev Immunol ; 19(7): 433-447, 2019 07.
Article in English | MEDLINE | ID: mdl-30874629

ABSTRACT

Pain is a hallmark of tissue injury, inflammatory diseases, pathogen invasion and neuropathy. It is mediated by nociceptor sensory neurons that innervate the skin, joints, bones, muscles and mucosal tissues and protects organisms from noxious stimuli. Nociceptors are sensitized by inflammatory mediators produced by the immune system, including cytokines, lipid mediators and growth factors, and can also directly detect pathogens and their secreted products to produce pain during infection. Upon activation, nociceptors release neuropeptides from their terminals that potently shape the function of innate and adaptive immune cells. For some pathogens, neuron-immune interactions enhance host protection from infection, but for other pathogens, neuron-immune signalling pathways can be exploited to facilitate pathogen survival. Here, we discuss the role of nociceptor interactions with the immune system in pain and infection and how understanding these pathways could produce new approaches to treat infectious diseases and chronic pain.


Subject(s)
Immunity , Pain/immunology , Animals , Cytokines/physiology , Humans , Immune System/physiology , Infections/immunology , Lymphocytes/physiology , Macrophages/physiology , Nociceptors/physiology
15.
Mucosal Immunol ; 12(1): 247-257, 2019 01.
Article in English | MEDLINE | ID: mdl-30279514

ABSTRACT

Gram-positive pathogens, including Staphylococcus aureus, cause necrotizing pneumonia. The central feature of S. aureus pneumonia is toxin-induced necroptosis of immune and resident cells, which impedes host defense. However, the role of the NLRC4 in the lung following S. aureus infection remains elusive. Here, we demonstrate that S. aureus activates the NLRC4 to drive necroptosis and IL-18 production, which impaired IL-17A-dependent neutrophil-mediated host susceptibility. In particular, Nlrc4-/- mice exhibit reduced necroptosis, enhanced neutrophil influx into the lungs, decreased bacterial burden, and improved host survival. Loss of NLRC4 signaling in both hematopoietic and non-hematopoietic cells contributes to the host protection against S. aureus pneumonia. Secretion of IL-17A by γδ T cells is essential for neutrophil recruitment into the lungs of Nlrc4-/- mice following infection. Moreover, treatment of wild-type mice with necroptosis inhibitors or genetic ablation of MLKL and IL-18 improves host defense against S. aureus infection, which is associated with increased IL-17A+γδ T cells and neutrophils. Taken together, these novel findings reveal that S. aureus activates the NLRC4 to dampen IL-17A-dependent neutrophil accumulation through induction of necroptosis and IL-18. Thus, modulating the function of the NLRC4 may be an attractive therapeutic approach for treating S. aureus infections.


Subject(s)
Lung/immunology , Neutrophils/immunology , Pneumonia, Staphylococcal/immunology , Staphylococcus aureus/immunology , T-Lymphocytes/immunology , Animals , Apoptosis , CARD Signaling Adaptor Proteins/metabolism , Calcium-Binding Proteins/metabolism , Cells, Cultured , Humans , Immune System Diseases , Interleukin-18/genetics , Interleukin-18/metabolism , Leukocyte Disorders , Mice , Mice, Inbred C57BL , Mice, Knockout , Necrosis , Protein Kinases/metabolism , Receptors, Antigen, T-Cell, gamma-delta/genetics , Signal Transduction , Up-Regulation
16.
Nat Med ; 24(10): 1625-1626, 2018 Oct.
Article in English | MEDLINE | ID: mdl-30013196

ABSTRACT

In the version of this article initially published, the line graph showing TNF-α levels in Fig. 2d was inadvertently duplicated. A graph of IL-6 levels should be shown in place of the duplication.These results were also incorrectly described in the main text, which originally stated: "At an early time point of infection (6 h), RTX-treated mice showed higher induction of total inflammatory-protein levels in the bronchoalveolar lavage fluid (BALF) (Fig. 2c), as well as levels of the cytokines TNF-α and IL-6, and the chemokine CXCL-1 (Fig. 2d)". This should instead read: "At an early time point of infection (6 h), RTX-treated mice showed higher induction of total inflammatory-protein levels in the bronchoalveolar lavage fluid (BALF) (Fig. 2c), as well as levels of the cytokine TNF-α and the chemokine CXCL-1 (Fig. 2d)".In the supplementary information initially posted online, incorrect bar graphs were presented in Supplementary Fig. 1b (VG, TRPV1+ data, top panel) and Supplementary Fig. 4b (DRG, CGRP+ data, middle panel).

17.
Nat Commun ; 9(1): 37, 2018 01 02.
Article in English | MEDLINE | ID: mdl-29295977

ABSTRACT

The hallmark of many bacterial infections is pain. The underlying mechanisms of pain during live pathogen invasion are not well understood. Here, we elucidate key molecular mechanisms of pain produced during live methicillin-resistant Staphylococcus aureus (MRSA) infection. We show that spontaneous pain is dependent on the virulence determinant agr and bacterial pore-forming toxins (PFTs). The cation channel, TRPV1, mediated heat hyperalgesia as a distinct pain modality. Three classes of PFTs-alpha-hemolysin (Hla), phenol-soluble modulins (PSMs), and the leukocidin HlgAB-directly induced neuronal firing and produced spontaneous pain. From these mechanisms, we hypothesized that pores formed in neurons would allow entry of the membrane-impermeable sodium channel blocker QX-314 into nociceptors to silence pain during infection. QX-314 induced immediate and long-lasting blockade of pain caused by MRSA infection, significantly more than lidocaine or ibuprofen, two widely used clinical analgesic treatments.


Subject(s)
Bacterial Toxins/toxicity , Lidocaine/analogs & derivatives , Methicillin-Resistant Staphylococcus aureus/metabolism , Pain/etiology , Staphylococcal Infections/physiopathology , TRPV Cation Channels/metabolism , Anesthetics, Local/pharmacology , Animals , Bacterial Toxins/metabolism , Gene Knockdown Techniques , Lidocaine/pharmacology , Methicillin-Resistant Staphylococcus aureus/genetics , Mice , Mice, Inbred C57BL , Neurons/drug effects , Neurons/metabolism , Pain/drug therapy , Staphylococcal Infections/complications , Staphylococcal Infections/microbiology
18.
Nat Med ; 24(4): 417-426, 2018 05.
Article in English | MEDLINE | ID: mdl-29505031

ABSTRACT

Lung-innervating nociceptor sensory neurons detect noxious or harmful stimuli and consequently protect organisms by mediating coughing, pain, and bronchoconstriction. However, the role of sensory neurons in pulmonary host defense is unclear. Here, we found that TRPV1+ nociceptors suppressed protective immunity against lethal Staphylococcus aureus pneumonia. Targeted TRPV1+-neuron ablation increased survival, cytokine induction, and lung bacterial clearance. Nociceptors suppressed the recruitment and surveillance of neutrophils, and altered lung γδ T cell numbers, which are necessary for immunity. Vagal ganglia TRPV1+ afferents mediated immunosuppression through release of the neuropeptide calcitonin gene-related peptide (CGRP). Targeting neuroimmunological signaling may be an effective approach to treat lung infections and bacterial pneumonia.


Subject(s)
Bacterial Infections/immunology , Neutrophils/metabolism , Nociceptors/metabolism , Pneumonia/immunology , Receptors, Antigen, T-Cell, gamma-delta/metabolism , Sensory Receptor Cells/metabolism , T-Lymphocytes/immunology , Animals , Bacterial Infections/microbiology , Calcitonin Gene-Related Peptide/metabolism , Cytokines/metabolism , Female , Host-Pathogen Interactions/immunology , Male , Mice, Inbred C57BL , NAV1.8 Voltage-Gated Sodium Channel/metabolism , Pneumonia/microbiology , Pneumonia/pathology , Staphylococcal Infections/microbiology , Staphylococcal Infections/pathology , Staphylococcus aureus/physiology , TRPV Cation Channels/metabolism , Vagus Nerve/metabolism
19.
Cell Host Microbe ; 19(6): 755-9, 2016 Jun 08.
Article in English | MEDLINE | ID: mdl-27281567

ABSTRACT

Pain and itch are unpleasant sensations accompanying many microbial infections. Recent studies demonstrate that pain- and itch-mediating somatosensory neurons are able to directly detect pathogens, triggering neuronal activation and subsequent regulation of immune responses. We discuss whether pain and/or itch during infection is beneficial or harmful to host antimicrobial defense.


Subject(s)
Infections/physiopathology , Pain/microbiology , Pruritus/microbiology , Adaptive Immunity/immunology , Host-Pathogen Interactions/immunology , Humans , Immunity, Innate , Infections/immunology , Infections/microbiology , Neural Pathways/immunology , Neural Pathways/physiology , Neuroimmunomodulation/immunology , Pain/immunology , Pruritus/immunology , Sensory Receptor Cells/immunology , Sensory Receptor Cells/microbiology
20.
Biomed Res Int ; 2015: 265425, 2015.
Article in English | MEDLINE | ID: mdl-25738151

ABSTRACT

The worldwide increase of multidrug resistance in both community- and health-care associated bacterial infections has impaired the current antimicrobial therapy, warranting the search for other alternatives. We aimed to find the in vitro antibacterial activity of ethanolic extracts of 16 different traditionally used medicinal plants of Nepal against 13 clinical and 2 reference bacterial species using microbroth dilution method. The evaluated plants species were found to exert a range of in vitro growth inhibitory action against the tested bacterial species, and Cynodon dactylon was found to exhibit moderate inhibitory action against 13 bacterial species including methicillin-resistant Staphylococcus aureus, imipenem-resistant Pseudomonas aeruginosa, multidrug-resistant Salmonella typhi, and S. typhimurium. The minimum inhibitory concentration (MIC) values of tested ethanolic extracts were found from 31 to >25,000 µg/mL. Notably, ethanolic extracts of Cinnamomum camphora, Curculigo orchioides, and Curcuma longa exhibited the highest antibacterial activity against S. pyogenes with a MIC of 49, 49, and 195 µg/mL, respectively; whereas chloroform fraction of Cynodon dactylon exhibited best antibacterial activity against S. aureus with a MIC of 31 µg/mL. Among all, C. dactylon, C. camphora, C. orchioides, and C. longa plant extracts displayed a potential antibacterial activity of MIC < 100 µg/mL.


Subject(s)
Anti-Bacterial Agents/pharmacology , Bacteria/growth & development , Plant Extracts/pharmacology , Plants, Medicinal/chemistry , Anti-Bacterial Agents/chemistry , Humans , Plant Extracts/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL