Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
PLoS Pathog ; 17(3): e1009387, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33690673

RESUMO

The skin innate immune response to methicillin-resistant Staphylococcus aureus (MRSA) culminates in the formation of an abscess to prevent bacterial spread and tissue damage. Pathogen recognition receptors (PRRs) dictate the balance between microbial control and injury. Therefore, intracellular brakes are of fundamental importance to tune the appropriate host defense while inducing resolution. The intracellular inhibitor suppressor of cytokine signaling 1 (SOCS-1), a known JAK/STAT inhibitor, prevents the expression and actions of PRR adaptors and downstream effectors. Whether SOCS-1 is a molecular component of skin host defense remains to be determined. We hypothesized that SOCS-1 decreases type I interferon production and IFNAR-mediated antimicrobial effector functions, limiting the inflammatory response during skin infection. Our data show that MRSA skin infection enhances SOCS-1 expression, and both SOCS-1 inhibitor peptide-treated and myeloid-specific SOCS-1 deficient mice display decreased lesion size, bacterial loads, and increased abscess thickness when compared to wild-type mice treated with the scrambled peptide control. SOCS-1 deletion/inhibition increases phagocytosis and bacterial killing, dependent on nitric oxide release. SOCS-1 inhibition also increases the levels of type I and type II interferon levels in vivo. IFNAR deletion and antibody blockage abolished the beneficial effects of SOCS-1 inhibition in vivo. Notably, we unveiled that hyperglycemia triggers aberrant SOCS-1 expression that correlates with decreased overall IFN signatures in the infected skin. SOCS-1 inhibition restores skin host defense in the highly susceptible hyperglycemic mice. Overall, these data demonstrate a role for SOCS-1-mediated type I interferon actions in host defense and inflammation during MRSA skin infection.


Assuntos
Interferon Tipo I/imunologia , Staphylococcus aureus Resistente à Meticilina/imunologia , Infecções Cutâneas Estafilocócicas/imunologia , Proteína 1 Supressora da Sinalização de Citocina/imunologia , Animais , Interferon Tipo I/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Pele/imunologia , Pele/microbiologia , Infecções Cutâneas Estafilocócicas/microbiologia , Proteína 1 Supressora da Sinalização de Citocina/metabolismo
2.
Proc Natl Acad Sci U S A ; 117(48): 30619-30627, 2020 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-33184178

RESUMO

The initial production of inflammatory mediators dictates host defense as well as tissue injury. Inflammasome activation is a constituent of the inflammatory response by recognizing pathogen and host-derived products and eliciting the production of IL-1ß and IL-18 in addition to inducing a type of inflammatory cell death termed "pyroptosis." Leukotriene B4 (LTB4) is a lipid mediator produced quickly (seconds to minutes) by phagocytes and induces chemotaxis, increases cytokine/chemokine production, and enhances antimicrobial effector functions. Whether LTB4 directly activates the inflammasome remains to be determined. Our data show that endogenously produced LTB4 is required for the expression of pro-IL-1ß and enhances inflammasome assembly in vivo and in vitro. Furthermore, LTB4-mediated Bruton's tyrosine kinase (BTK) activation is required for inflammasome assembly in vivo as well for IL-1ß-enhanced skin host defense. Together, these data unveil a new role for LTB4 in enhancing the expression and assembly of inflammasome components and suggest that while blocking LTB4 actions could be a promising therapeutic strategy to prevent inflammasome-mediated diseases, exogenous LTB4 can be used as an adjuvant to boost inflammasome-dependent host defense.


Assuntos
Interações Hospedeiro-Patógeno , Inflamassomos/metabolismo , Leucotrieno B4/metabolismo , Fenômenos Fisiológicos da Pele , Pele/metabolismo , Animais , Biópsia , Citocinas/metabolismo , Interações Hospedeiro-Patógeno/imunologia , Imunidade Inata , Mediadores da Inflamação/metabolismo , Macrófagos/imunologia , Macrófagos/metabolismo , Staphylococcus aureus Resistente à Meticilina , Camundongos , Pele/imunologia , Pele/microbiologia , Pele/patologia
3.
Semin Immunol ; 33: 37-43, 2017 10.
Artigo em Inglês | MEDLINE | ID: mdl-29042027

RESUMO

The ability to regulate inflammatory pathways and host defense mechanisms is critical for maintaining homeostasis and responding to infections and tissue injury. While unbalanced inflammation is detrimental to the host; inadequate inflammation might not provide effective signals required to eliminate pathogens. On the other hand, aberrant inflammation could result in organ damage and impair host defense. The lipid mediator leukotriene B4 (LTB4) is a potent neutrophil chemoattractant and recently, its role as a dominant molecule that amplifies many arms of phagocyte antimicrobial effector function has been unveiled. However, excessive LTB4 production contributes to disease severity in chronic inflammatory diseases such as diabetes and arthritis, which could potentially be involved in poor host defense in these groups of patients. In this review we discuss the cellular and molecular programs elicited during LTB4 production and actions on innate immunity host defense mechanisms as well as potential therapeutic strategies to improve host defense.


Assuntos
Imunidade Inata , Inflamação , Leucotrieno B4/metabolismo , Neutrófilos/imunologia , Animais , Movimento Celular , Homeostase , Humanos , Imunomodulação , Leucotrieno B4/química , Fagocitose
4.
PLoS Pathog ; 14(8): e1007244, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-30102746

RESUMO

The early events that shape the innate immune response to restrain pathogens during skin infections remain elusive. Methicillin-resistant Staphylococcus aureus (MRSA) infection engages phagocyte chemotaxis, abscess formation, and microbial clearance. Upon infection, neutrophils and monocytes find a gradient of chemoattractants that influence both phagocyte direction and microbial clearance. The bioactive lipid leukotriene B4 (LTB4) is quickly (seconds to minutes) produced by 5-lipoxygenase (5-LO) and signals through the G protein-coupled receptors LTB4R1 (BLT1) or BLT2 in phagocytes and structural cells. Although it is known that LTB4 enhances antimicrobial effector functions in vitro, whether prompt LTB4 production is required for bacterial clearance and development of an inflammatory milieu necessary for abscess formation to restrain pathogen dissemination is unknown. We found that LTB4 is produced in areas near the abscess and BLT1 deficient mice are unable to form an abscess, elicit neutrophil chemotaxis, generation of neutrophil and monocyte chemokines, as well as reactive oxygen species-dependent bacterial clearance. We also found that an ointment containing LTB4 synergizes with antibiotics to eliminate MRSA potently. Here, we uncovered a heretofore unknown role of macrophage-derived LTB4 in orchestrating the chemoattractant gradient required for abscess formation, while amplifying antimicrobial effector functions.


Assuntos
Abscesso/imunologia , Carga Bacteriana/imunologia , Leucotrieno B4/fisiologia , Macrófagos/metabolismo , Staphylococcus aureus Resistente à Meticilina , Infecções Cutâneas Estafilocócicas/imunologia , Abscesso/genética , Abscesso/microbiologia , Abscesso/patologia , Animais , Araquidonato 5-Lipoxigenase/genética , Carga Bacteriana/genética , Células Cultivadas , Feminino , Leucotrieno B4/metabolismo , Macrófagos/imunologia , Masculino , Staphylococcus aureus Resistente à Meticilina/crescimento & desenvolvimento , Staphylococcus aureus Resistente à Meticilina/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores do Leucotrieno B4/genética , Infecções Cutâneas Estafilocócicas/genética , Infecções Cutâneas Estafilocócicas/patologia
5.
J Immunol ; 200(12): 3871-3880, 2018 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-29866769

RESUMO

Staphylococcus aureus causes a wide range of diseases that together embody a significant public health burden. Aided by metabolic flexibility and a large virulence repertoire, S. aureus has the remarkable ability to hematogenously disseminate and infect various tissues, including skin, lung, heart, and bone, among others. The hallmark lesions of invasive staphylococcal infections, abscesses, simultaneously denote the powerful innate immune responses to tissue invasion as well as the ability of staphylococci to persist within these lesions. In this article, we review the innate immune responses to S. aureus during infection of skin and bone, which serve as paradigms for soft tissue and bone disease, respectively.


Assuntos
Osso e Ossos/imunologia , Osso e Ossos/microbiologia , Imunidade Inata/imunologia , Pele/imunologia , Pele/microbiologia , Infecções Estafilocócicas/imunologia , Staphylococcus aureus/imunologia , Animais , Humanos
6.
Adv Exp Med Biol ; 1274: 55-69, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32894507

RESUMO

Leukotrienes (LTs) are potent lipid mediators that exert a variety of functions, ranging from maintaining the tone of the homeostatic immune response to exerting potent proinflammatory effects. Therefore, LTs are essential elements in the development and maintenance of different chronic diseases, such as asthma, arthritis, and atherosclerosis. Due to the pleiotropic effects of LTs in the pathogenesis of inflammatory diseases, studies are needed to discover potent and specific LT synthesis inhibitors and LT receptor antagonists. Even though most clinical trials using LT inhibitors or antagonists have failed due to low efficacy and/or toxicity, new drug development strategies are driving the discovery for LT inhibitors to prevent inflammatory diseases. A newly important detrimental role for LTs in comorbidities associated with metabolic stress has emerged in the last few years and managing LT production and/or actions could represent an exciting new strategy to prevent or treat inflammatory diseases associated with metabolic disorders. This review is intended to shed light on the synthesis and actions of leukotrienes, the most common drugs used in clinical trials, and discuss the therapeutic potential of preventing LT function in obesity, diabetes, and hyperlipidemia.


Assuntos
Comorbidade , Antagonistas de Leucotrienos/uso terapêutico , Leucotrienos/metabolismo , Doenças Metabólicas/complicações , Doenças Metabólicas/prevenção & controle , Estresse Fisiológico , Asma , Aterosclerose , Humanos
7.
Sex Transm Dis ; 43(9): 579-83, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27513385

RESUMO

BACKGROUND: High rates of 23S rDNA mutations implicated in macrolide resistance have been identified in Treponema pallidum samples from syphilis patients in many countries. Nonetheless, some clinicians have been reluctant to abandon azithromycin as a treatment for syphilis, citing the lack of a causal association between these mutations and clinical evidence of drug resistance. Although azithromycin resistance has been demonstrated in vivo for the historical Street 14 strain, no recent T. pallidum isolates have been tested. We used the well-established rabbit model of syphilis to determine the in vivo efficacy of azithromycin against 23S rDNA mutant strains collected in 2004 to 2005 from patients with syphilis in Seattle, Wash. METHODS: Groups of 9 rabbits were each infected with a strain containing 23S rDNA mutation A2058G (strains UW074B, UW189B, UW391B) or A2059G (strains UW228B, UW254B, and UW330B), or with 1 wild type strain (Chicago, Bal 3, and Mexico A). After documentation of infection, 3 animals per strain were treated with azithromycin, 3 were treated with benzathine penicillin G, and 3 served as untreated control groups. Treatment efficacy was documented by darkfield microscopic evidence of T. pallidum, serological response, and rabbit infectivity test. RESULTS: Azithromycin uniformly failed to cure rabbits infected with strains harboring either 23S rDNA mutation, although benzathine penicillin G was effective. Infections caused by wild type strains were successfully treated by either azithromycin or benzathine penicillin G. CONCLUSIONS: A macrolide resistant phenotype was demonstrated for all strains harboring a 23S rDNA mutation, demonstrating that either A2058G or A2059G mutation confers in vivo drug resistance.


Assuntos
DNA Bacteriano/efeitos dos fármacos , DNA Ribossômico/efeitos dos fármacos , Farmacorresistência Bacteriana/genética , Macrolídeos/farmacologia , Treponema pallidum/genética , Animais , Antibacterianos/farmacologia , Azitromicina/farmacologia , Modelos Animais de Doenças , Humanos , Mutação/efeitos dos fármacos , Penicilina G Benzatina/farmacologia , Coelhos , Sífilis/tratamento farmacológico , Treponema pallidum/isolamento & purificação
8.
Infect Immun ; 83(12): 4848-60, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26438793

RESUMO

Outer surface protein C (OspC) is one of the major lipoproteins expressed on the surface of Borrelia burgdorferi during tick feeding and the early phase of mammalian infection. OspC is required for B. burgdorferi to establish infection in both immunocompetent and SCID mice and has been proposed to facilitate evasion of innate immune defenses. However, the exact biological function of OspC remains elusive. In this study, we showed that the ospC-deficient spirochete could not establish infection in NOD-scid IL2rγ(null) mice that lack B cells, T cells, NK cells, and lytic complement. The ospC mutant also could not establish infection in anti-Ly6G-treated SCID and C3H/HeN mice (depletion of neutrophils). However, depletion of mononuclear phagocytes at the skin site of inoculation in SCID and C3H/HeN mice allowed the ospC mutant to establish infection in vivo. In phagocyte-depleted mice, the ospC mutant was able to colonize the joints and triggered neutrophilia during dissemination. Furthermore, we found that phagocytosis of green fluorescent protein (GFP)-expressing ospC mutant spirochetes by murine peritoneal macrophages and human THP-1 macrophage-like cells, but not in PMN-HL60, was significantly higher than parental wild-type B. burgdorferi strains, suggesting that OspC has an antiphagocytic property. In addition, overproduction of OspC in spirochetes also decreased the uptake of spirochetes by murine peritoneal macrophages. Together, our findings provide evidence that mononuclear phagocytes play a key role in clearance of the ospC mutant and that OspC promotes spirochetes' evasion of macrophages during early Lyme borreliosis.


Assuntos
Antígenos de Bactérias/imunologia , Proteínas da Membrana Bacteriana Externa/imunologia , Borrelia burgdorferi/genética , Regulação Bacteriana da Expressão Gênica , Evasão da Resposta Imune , Doença de Lyme/imunologia , Macrófagos Peritoneais/imunologia , Animais , Antígenos de Bactérias/genética , Linfócitos B/imunologia , Linfócitos B/microbiologia , Linfócitos B/patologia , Proteínas da Membrana Bacteriana Externa/genética , Borrelia burgdorferi/imunologia , Borrelia burgdorferi/patogenicidade , Linhagem Celular , Feminino , Genes Reporter , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Humanos , Células Matadoras Naturais/imunologia , Células Matadoras Naturais/microbiologia , Células Matadoras Naturais/patologia , Doença de Lyme/genética , Doença de Lyme/microbiologia , Doença de Lyme/patologia , Macrófagos Peritoneais/microbiologia , Macrófagos Peritoneais/patologia , Camundongos , Camundongos Endogâmicos NOD , Camundongos Knockout , Camundongos SCID , Neutrófilos/imunologia , Neutrófilos/microbiologia , Neutrófilos/patologia , Linfócitos T/imunologia , Linfócitos T/microbiologia , Linfócitos T/patologia
9.
Infect Immun ; 83(6): 2275-89, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25802057

RESUMO

An effective mechanism for introduction of phenotypic diversity within a bacterial population exploits changes in the length of repetitive DNA elements located within gene promoters. This phenomenon, known as phase variation, causes rapid activation or silencing of gene expression and fosters bacterial adaptation to new or changing environments. Phase variation often occurs in surface-exposed proteins, and in Treponema pallidum subsp. pallidum, the syphilis agent, it was reported to affect transcription of three putative outer membrane protein (OMP)-encoding genes. When the T. pallidum subsp. pallidum Nichols strain genome was initially annotated, the TP0126 open reading frame was predicted to include a poly(G) tract and did not appear to have a predicted signal sequence that might suggest the possibility of its being an OMP. Here we show that the initial annotation was incorrect, that this poly(G) is instead located within the TP0126 promoter, and that it varies in length in vivo during experimental syphilis. Additionally, we show that TP0126 transcription is affected by changes in the poly(G) length consistent with regulation by phase variation. In silico analysis of the TP0126 open reading frame based on the experimentally identified transcriptional start site shortens this hypothetical protein by 69 amino acids, reveals a predicted cleavable signal peptide, and suggests structural homology with the OmpW family of porins. Circular dichroism of recombinant TP0126 supports structural homology to OmpW. Together with the evidence that TP0126 is fully conserved among T. pallidum subspecies and strains, these data suggest an important role for TP0126 in T. pallidum biology and syphilis pathogenesis.


Assuntos
Proteínas da Membrana Bacteriana Externa/metabolismo , Regulação Bacteriana da Expressão Gênica/fisiologia , Guanosina/química , Transcrição Gênica , Treponema pallidum/metabolismo , Animais , Proteínas da Membrana Bacteriana Externa/química , Proteínas da Membrana Bacteriana Externa/genética , Sequência de Bases , Humanos , Imunidade Humoral , Modelos Moleculares , Conformação Proteica , Coelhos , Proteínas Recombinantes/metabolismo , Sífilis/microbiologia , Sítio de Iniciação de Transcrição
10.
J Bacteriol ; 194(16): 4208-25, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22661689

RESUMO

Although the three Treponema pallidum subspecies (T. pallidum subsp. pallidum, T. pallidum subsp. pertenue, and T. pallidum subsp. endemicum), Treponema paraluiscuniculi, and the unclassified Fribourg-Blanc treponeme cause clinically distinct diseases, these pathogens are genetically and antigenically highly related and are able to cause persistent infection. Recent evidence suggests that the putative surface-exposed variable antigen TprK plays an important role in both treponemal immune evasion and persistence. tprK heterogeneity is generated by nonreciprocal gene conversion between the tprK expression site and donor sites. Although each of the above-mentioned species and subspecies has a functional tprK antigenic variation system, it is still unclear why the level of expression and the rate at which tprK diversifies during infection can differ significantly among isolates. To identify genomic differences that might affect the generation and expression of TprK variants among these pathogens, we performed comparative sequence analysis of the donor sites, as well as the tprK expression sites, among eight T. pallidum subsp. pallidum isolates (Nichols Gen, Nichols Sea, Chicago, Sea81-4, Dal-1, Street14, UW104, and UW126), three T. pallidum subsp. pertenue isolates (Gauthier, CDC2, and Samoa D), one T. pallidum subsp. endemicum isolate (Iraq B), the unclassified Fribourg-Blanc isolate, and the Cuniculi A strain of T. paraluiscuniculi. Synteny and sequence conservation, as well as deletions and insertions, were found in the regions harboring the donor sites. These data suggest that the tprK recombination system is harbored within dynamic genomic regions and that genomic differences might be an important key to explain discrepancies in generation and expression of tprK variants among these Treponema isolates.


Assuntos
Variação Antigênica , Proteínas de Bactérias/genética , Proteínas de Bactérias/imunologia , Porinas/genética , Porinas/imunologia , Treponema/genética , Treponema/imunologia , Sequência Conservada , DNA Bacteriano/química , DNA Bacteriano/genética , Genoma Bacteriano , Dados de Sequência Molecular , Mutagênese Insercional , Polimorfismo Genético , Análise de Sequência de DNA , Deleção de Sequência , Homologia de Sequência , Sintenia , Treponema/classificação
11.
BMJ Case Rep ; 15(9)2022 Sep 22.
Artigo em Inglês | MEDLINE | ID: mdl-36137645

RESUMO

We present a rare case of destructive osteomyelitis of the sternum caused by Parvimonas micra and Campylobacter rectus A previously healthy female patient in her 40s presented to the emergency department due to a spontaneous rupture of an abscess located to the chest wall. Imaging confirmed abscess formation with osteomyelitis of the sternum. Emergent surgical debridement was performed, blood and bone cultures were taken and the patient received antibiotic treatment. Cultures of the bone and deep tissue revealed infection with Parvimonas micra and Campylobacter rectus, both being members of the oral flora and associated with chronic periodontitis. Receiving targeted antibiotic treatment, our patient made a quick recovery. After treatment of the osteomyelitis, our patient was referred to the dentist where chronic periodontitis could be confirmed. Invasive infections with Parvimonas micra and Campylobacter rectus are rare. Investigation of a dental origin is crucial to prevent recurrent infections.


Assuntos
Periodontite Crônica , Osteomielite , Abscesso/tratamento farmacológico , Antibacterianos/uso terapêutico , Campylobacter rectus , Periodontite Crônica/tratamento farmacológico , Feminino , Firmicutes , Humanos , Osteomielite/diagnóstico , Osteomielite/tratamento farmacológico , Peptostreptococcus
12.
mSphere ; 5(2)2020 04 22.
Artigo em Inglês | MEDLINE | ID: mdl-32321824

RESUMO

Group B Streptococcus (GBS) causes frequent urinary tract infection (UTI) in susceptible populations, including individuals with type 2 diabetes and pregnant women; however, specific host factors responsible for increased GBS susceptibility in these populations are not well characterized. Here, we investigate cathelicidin, a cationic antimicrobial peptide, known to be critical for defense during UTI with uropathogenic Escherichia coli (UPEC). We observed a loss of antimicrobial activity of human and mouse cathelicidins against GBS and UPEC in synthetic urine and no evidence for increased cathelicidin resistance in GBS urinary isolates. Furthermore, we found that GBS degrades cathelicidin in a protease-dependent manner. Surprisingly, in a UTI model, cathelicidin-deficient (Camp-/-) mice showed decreased GBS burdens and mast cell recruitment in the bladder compared to levels in wild-type (WT) mice. Pharmacologic inhibition of mast cells reduced GBS burdens and histamine release in WT but not Camp-/- mice. Streptozotocin-induced diabetic mice had increased bladder cathelicidin production and mast cell recruitment at 24 h postinfection with GBS compared to levels in nondiabetic controls. We propose that cathelicidin is an important immune regulator but ineffective antimicrobial peptide against GBS in urine. Combined, our findings may in part explain the increased frequency of GBS UTI in diabetic and pregnant individuals.IMPORTANCE Certain populations such as diabetic individuals are at increased risk for developing urinary tract infections (UTI), although the underlying reasons for this susceptibility are not fully known. Additionally, diabetics are more likely to become infected with certain types of bacteria, such as group B Streptococcus (GBS). In this study, we find that an antimicrobial peptide called cathelicidin, which is thought to protect the bladder from infection, is ineffective in controlling GBS and alters the type of immune cells that migrate to the bladder during infection. Using a mouse model of diabetes, we observe that diabetic mice are more susceptible to GBS infection even though they also have more infiltrating immune cells and increased production of cathelicidin. Taken together, our findings identify this antimicrobial peptide as a potential contributor to increased susceptibility of diabetic individuals to GBS UTI.


Assuntos
Peptídeos Catiônicos Antimicrobianos/imunologia , Infecções Estreptocócicas/microbiologia , Exacerbação dos Sintomas , Infecções Urinárias/microbiologia , Animais , Peptídeos Catiônicos Antimicrobianos/genética , Linhagem Celular , Diabetes Mellitus Experimental/induzido quimicamente , Diabetes Mellitus Experimental/microbiologia , Diabetes Mellitus Tipo 2/complicações , Diabetes Mellitus Tipo 2/microbiologia , Feminino , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Gravidez , Infecções Estreptocócicas/imunologia , Streptococcus/metabolismo , Bexiga Urinária/imunologia , Bexiga Urinária/microbiologia , Infecções Urinárias/imunologia , Catelicidinas
13.
JCI Insight ; 3(17)2018 09 06.
Artigo em Inglês | MEDLINE | ID: mdl-30185672

RESUMO

Poorly controlled diabetes leads to comorbidities and enhanced susceptibility to infections. While the immune components involved in wound healing in diabetes have been studied, the components involved in susceptibility to skin infections remain unclear. Here, we examined the effects of the inflammatory lipid mediator leukotriene B4 (LTB4) signaling through its receptor B leukotriene receptor 1 (BLT1) in the progression of methicillin-resistant Staphylococcus aureus (MRSA) skin infection in 2 models of diabetes. Diabetic mice produced higher levels of LTB4 in the skin, which correlated with larger nonhealing lesion areas and increased bacterial loads compared with nondiabetic mice. High LTB4 levels were also associated with dysregulated cytokine and chemokine production, excessive neutrophil migration but impaired abscess formation, and uncontrolled collagen deposition. Both genetic deletion and topical pharmacological BLT1 antagonism restored inflammatory response and abscess formation, followed by a reduction in the bacterial load and lesion area in the diabetic mice. Macrophage depletion in diabetic mice limited LTB4 production and improved abscess architecture and skin host defense. These data demonstrate that exaggerated LTB4/BLT1 responses mediate a derailed inflammatory milieu that underlies poor host defense in diabetes. Prevention of LTB4 production/actions could provide a new therapeutic strategy to restore host defense in diabetes.


Assuntos
Diabetes Mellitus Experimental/metabolismo , Leucotrieno B4/metabolismo , Pele/imunologia , Pele/metabolismo , Infecções Cutâneas Estafilocócicas/imunologia , Abscesso/imunologia , Abscesso/patologia , Animais , Carga Bacteriana , Movimento Celular , Quimiocinas/metabolismo , Citocinas/metabolismo , Feminino , Inflamação , Leucotrieno B4/genética , Leucotrieno B4/imunologia , Macrófagos/imunologia , Masculino , Staphylococcus aureus Resistente à Meticilina/patogenicidade , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neutrófilos/imunologia , Receptores do Leucotrieno B4/efeitos dos fármacos , Receptores do Leucotrieno B4/genética , Receptores do Leucotrieno B4/metabolismo , Transdução de Sinais , Pele/patologia , Infecções Cutâneas Estafilocócicas/patologia
14.
Sci Signal ; 11(528)2018 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-29717063

RESUMO

Sepsis-induced organ damage is caused by systemic inflammatory response syndrome (SIRS), which results in substantial comorbidities. Therefore, it is of medical importance to identify molecular brakes that can be exploited to dampen inflammation and prevent the development of SIRS. We investigated the role of phosphatase and tensin homolog (PTEN) in suppressing SIRS, increasing microbial clearance, and preventing lung damage. Septic patients and mice with sepsis exhibited increased PTEN expression in leukocytes. Myeloid-specific Pten deletion in an animal model of sepsis increased bacterial loads and cytokine production, which depended on enhanced myeloid differentiation primary response gene 88 (MyD88) abundance and resulted in mortality. PTEN-mediated induction of the microRNAs (miRNAs) miR125b and miR203b reduced the abundance of MyD88. Loss- and gain-of-function assays demonstrated that PTEN induced miRNA production by associating with and facilitating the nuclear localization of Drosha-Dgcr8, part of the miRNA-processing complex. Reconstitution of PTEN-deficient mouse embryonic fibroblasts with a mutant form of PTEN that does not localize to the nucleus resulted in retention of Drosha-Dgcr8 in the cytoplasm and impaired production of mature miRNAs. Thus, we identified a regulatory pathway involving nuclear PTEN-mediated miRNA generation that limits the production of MyD88 and thereby limits sepsis-associated mortality.


Assuntos
MicroRNAs/genética , Fator 88 de Diferenciação Mieloide/genética , PTEN Fosfo-Hidrolase/genética , Regulon/genética , Sepse/genética , Animais , Núcleo Celular/genética , Núcleo Celular/metabolismo , Feminino , Perfilação da Expressão Gênica , Humanos , Inflamação/genética , Inflamação/metabolismo , Macrófagos/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fator 88 de Diferenciação Mieloide/química , Fator 88 de Diferenciação Mieloide/metabolismo , PTEN Fosfo-Hidrolase/metabolismo , Peptídeos/farmacologia , Interferência de RNA , Sepse/metabolismo , Sepse/prevenção & controle
15.
J Diabetes Complications ; 31(2): 334-339, 2017 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-27623388

RESUMO

AIMS: To investigate the hypothesis that alteration in histone acetylation/deacetylation triggers aberrant STAT1/MyD88 expression in macrophages from diabetics. Increased STAT1/MyD88 expression is correlated with sterile inflammation in type 1 diabetic (T1D) mice. METHODS: To induce diabetes, we injected low-doses of streptozotocin in C57BL/6 mice. Peritoneal or bone marrow-differentiated macrophages were cultured in 5mM (low) or 25mM (high glucose). ChIP analysis of macrophages from nondiabetic or diabetic mice was performed to determine acetylation of lysine 9 in histone H3 in MyD88 and STAT1 promoter regions. Macrophages from diabetic mice were treated with the histone acetyltransferase inhibitor anacardic acid (AA), followed by determination of mRNA expression by qPCR. RESULTS: Increased STAT1 and MyD88 expression in macrophages from diabetic but not naive mice cultured in low glucose persisted for up to 6days. Macrophages from diabetic mice exhibited increased activity of histone acetyltransferases (HAT) and decreased histone deacetylases (HDAC) activity. We detected increased H3K9Ac binding to Stat1/Myd88 promoters in macrophages from T1D mice and AA in vitro treatment reduced STAT1 and MyD88 mRNA expression. CONCLUSIONS/INTERPRETATION: These results indicate that histone acetylation drives elevated Stat1/Myd88 expression in macrophages from diabetic mice, and this mechanism may be involved in sterile inflammation and diabetes comorbidities.


Assuntos
Diabetes Mellitus Tipo 1/metabolismo , Regulação da Expressão Gênica , Histona Acetiltransferases/metabolismo , Histona Desacetilases/metabolismo , Macrófagos/metabolismo , Fator 88 de Diferenciação Mieloide/metabolismo , Fator de Transcrição STAT1/metabolismo , Acetilação/efeitos dos fármacos , Animais , Células da Medula Óssea/efeitos dos fármacos , Células da Medula Óssea/imunologia , Células da Medula Óssea/metabolismo , Células da Medula Óssea/patologia , Células Cultivadas , Diabetes Mellitus Tipo 1/induzido quimicamente , Diabetes Mellitus Tipo 1/imunologia , Diabetes Mellitus Tipo 1/patologia , Inibidores Enzimáticos/farmacologia , Epigênese Genética/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Glucose/metabolismo , Histona Acetiltransferases/antagonistas & inibidores , Histonas/metabolismo , Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Macrófagos/patologia , Macrófagos Peritoneais/efeitos dos fármacos , Macrófagos Peritoneais/imunologia , Macrófagos Peritoneais/metabolismo , Macrófagos Peritoneais/patologia , Masculino , Camundongos Endogâmicos C57BL , Fator 88 de Diferenciação Mieloide/genética , Concentração Osmolar , Regiões Promotoras Genéticas/efeitos dos fármacos , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Fator de Transcrição STAT1/genética , Estreptozocina/toxicidade
16.
Diabetes ; 65(12): 3718-3729, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-27605625

RESUMO

People with diabetes are more prone to Staphylococcus aureus skin infection than healthy individuals. Control of S. aureus infection depends on dendritic cell (DC)-induced T-helper 17 (Th17)-mediated neutrophil recruitment and bacterial clearance. DC ingestion of infected apoptotic cells (IACs) drive prostaglandin E2 (PGE2) secretion to generate Th17 cells. We speculated that hyperglycemia inhibits skin DC migration to the lymph nodes and impairs the Th17 differentiation that accounts for poor skin host defense in diabetic mice. Diabetic mice showed increased skin lesion size and bacterial load and decreased PGE2 secretion and Th17 cells compared with nondiabetic mice after methicillin-resistant S. aureus (MRSA) infection. Bone marrow-derived DCs (BMDCs) cultured in high glucose (25 mmol/L) exhibited decreased Ptges mRNA expression, PGE2 production, lower CCR7-dependent DC migration, and diminished maturation after recognition of MRSA-IACs than BMDCs cultured in low glucose (5 mmol/L). Similar events were observed in DCs from diabetic mice infected with MRSA. Topical treatment of diabetic mice with the PGE analog misoprostol improved host defense against MRSA skin infection by restoring DC migration to draining lymph nodes, Th17 differentiation, and increased antimicrobial peptide expression. These findings identify a novel mechanism involved in poor skin host defense in diabetes and propose a targeted strategy to restore skin host defense in diabetes.


Assuntos
Células Dendríticas/efeitos dos fármacos , Células Dendríticas/metabolismo , Staphylococcus aureus Resistente à Meticilina/patogenicidade , Prostaglandinas E Sintéticas/uso terapêutico , Pele/microbiologia , Infecções Estafilocócicas/tratamento farmacológico , Células Th17/citologia , Células Th17/metabolismo , Animais , Apoptose/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Citometria de Fluxo , Staphylococcus aureus Resistente à Meticilina/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Pele/metabolismo , Infecções Estafilocócicas/metabolismo , Infecções Estafilocócicas/microbiologia
17.
Oncotarget ; 7(17): 23096-105, 2016 Apr 26.
Artigo em Inglês | MEDLINE | ID: mdl-27049833

RESUMO

Drug resistance is a growing problem that necessitates new strategies to combat pathogens. Neutrophil phagocytosis and production of intracellular ROS, in particular, has been shown to cooperate with antibiotics in the killing of microbes. This study tested the hypothesis that p85α, the regulatory subunit of PI3K, regulates production of intracellular ROS. Genetic knockout of p85α in mice caused decreased expression of catalytic subunits p110α, p110ß, and p110δ, but did not change expression levels of the NADPH oxidase complex subunits p67phox, p47phox, and p40phox. When p85α, p55α, and p50α (all encoded by Pik3r1) were deleted, there was an increase in intracellular ROS with no change in phagocytosis in response to both Fcγ receptor and complement receptor stimulation. Furthermore, the increased intracellular ROS correlated with significantly improved neutrophil killing of both methicillin-susceptible and methicillin-resistant S. aureus. Our findings suggest inhibition of p85α as novel approach to improving the clearance of resistant pathogens.


Assuntos
Classe Ia de Fosfatidilinositol 3-Quinase/metabolismo , Neutrófilos/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Animais , Células Cultivadas , Classe Ia de Fosfatidilinositol 3-Quinase/fisiologia , Camundongos , Camundongos Knockout , Transdução de Sinais
18.
Sci Signal ; 8(361): ra10, 2015 Jan 27.
Artigo em Inglês | MEDLINE | ID: mdl-25628460

RESUMO

Type 1 diabetes mellitus (T1DM) is associated with chronic systemic inflammation and enhanced susceptibility to systemic bacterial infection (sepsis). We hypothesized that low insulin concentrations in T1DM trigger the enzyme 5-lipoxygenase (5-LO) to produce the lipid mediator leukotriene B4 (LTB4), which triggers systemic inflammation that may increase susceptibility to polymicrobial sepsis. Consistent with chronic inflammation, peritoneal macrophages from two mouse models of T1DM had greater abundance of the adaptor MyD88 (myeloid differentiation factor 88) and its direct transcriptional effector STAT-1 (signal transducer and activator of transcription 1) than macrophages from nondiabetic mice. Expression of Alox5, which encodes 5-LO, and the concentration of the proinflammatory cytokine interleukin-1ß (IL-1ß) were also increased in peritoneal macrophages and serum from T1DM mice. Insulin treatment reduced LTB4 concentrations in the circulation and Myd88 and Stat1 expression in the macrophages from T1DM mice. T1DM mice treated with a 5-LO inhibitor had reduced Myd88 mRNA in macrophages and increased abundance of IL-1 receptor antagonist and reduced production of IL-ß in the circulation. T1DM mice lacking 5-LO or the receptor for LTB4 also produced less proinflammatory cytokines. Compared to wild-type or untreated diabetic mice, T1DM mice lacking the receptor for LTB4 or treated with a 5-LO inhibitor survived polymicrobial sepsis, had reduced production of proinflammatory cytokines, and had decreased bacterial counts. These results uncover a role for LTB4 in promoting sterile inflammation in diabetes and the enhanced susceptibility to sepsis in T1DM.


Assuntos
Diabetes Mellitus Tipo 1/complicações , Regulação da Expressão Gênica/fisiologia , Mediadores da Inflamação/metabolismo , Inflamação/complicações , Leucotrieno B4/metabolismo , Sepse/etiologia , Análise de Variância , Animais , Araquidonato 5-Lipoxigenase/genética , Araquidonato 5-Lipoxigenase/metabolismo , Imunoprecipitação da Cromatina , Citocinas/metabolismo , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Immunoblotting , Inflamação/metabolismo , Insulina/deficiência , Insulina/farmacologia , Macrófagos/metabolismo , Camundongos , Camundongos Knockout , Fator 88 de Diferenciação Mieloide/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fator de Transcrição STAT1/metabolismo , Sepse/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA