Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 78
Filter
Add more filters

Publication year range
1.
Cell ; 165(3): 690-703, 2016 Apr 21.
Article in English | MEDLINE | ID: mdl-27062925

ABSTRACT

Pili are proteinaceous polymers of linked pilins that protrude from the cell surface of many bacteria and often mediate adherence and virulence. We investigated a set of 20 Bacteroidia pilins from the human microbiome whose structures and mechanism of assembly were unknown. Crystal structures and biochemical data revealed a diverse protein superfamily with a common Greek-key ß sandwich fold with two transthyretin-like repeats that polymerize into a pilus through a strand-exchange mechanism. The assembly mechanism of the central, structural pilins involves proteinase-assisted removal of their N-terminal ß strand, creating an extended hydrophobic groove that binds the C-terminal donor strands of the incoming pilin. Accessory pilins at the tip and base have unique structural features specific to their location, allowing initiation or termination of the assembly. The Bacteroidia pilus, therefore, has a biogenesis mechanism that is distinct from other known pili and likely represents a different type of bacterial pilus.


Subject(s)
Fimbriae Proteins/chemistry , Fimbriae, Bacterial , Gastrointestinal Microbiome , Amino Acid Sequence , Crystallography, X-Ray , Fimbriae Proteins/genetics , Fimbriae Proteins/metabolism , Humans , Lipoproteins/chemistry , Lipoproteins/metabolism , Models, Molecular , Molecular Sequence Data , Sequence Alignment
2.
Nat Immunol ; 13(5): 465-73, 2012 Mar 25.
Article in English | MEDLINE | ID: mdl-22447028

ABSTRACT

Aging is linked to greater susceptibility to chronic inflammatory diseases, several of which, including periodontitis, involve neutrophil-mediated tissue injury. Here we found that aging-associated periodontitis was accompanied by lower expression of Del-1, an endogenous inhibitor of neutrophil adhesion dependent on the integrin LFA-1, and by reciprocal higher expression of interleukin 17 (IL-17). Consistent with that, IL-17 inhibited gingival endothelial cell expression of Del-1, thereby promoting LFA-1-dependent recruitment of neutrophils. Young Del-1-deficient mice developed spontaneous periodontitis that featured excessive neutrophil infiltration and IL-17 expression; disease was prevented in mice doubly deficient in Del-1 and LFA-1 or in Del-1 and the IL-17 receptor. Locally administered Del-1 inhibited IL-17 production, neutrophil accumulation and bone loss. Therefore, Del-1 suppressed LFA-1-dependent recruitment of neutrophils and IL-17-triggered inflammatory pathology and may thus be a promising therapeutic agent for inflammatory diseases.


Subject(s)
Alveolar Bone Loss/immunology , Carrier Proteins/metabolism , Interleukin-17/antagonists & inhibitors , Interleukin-17/metabolism , Neutrophil Infiltration/drug effects , Periodontitis/metabolism , Aging/immunology , Animals , Calcium-Binding Proteins , Carrier Proteins/immunology , Carrier Proteins/pharmacology , Cell Adhesion/drug effects , Cell Adhesion Molecules , Endothelial Cells/drug effects , Endothelial Cells/immunology , Female , Integrins/antagonists & inhibitors , Integrins/immunology , Integrins/metabolism , Intercellular Signaling Peptides and Proteins , Interleukin-17/immunology , Lymphocyte Function-Associated Antigen-1/immunology , Lymphocyte Function-Associated Antigen-1/metabolism , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Neutrophil Infiltration/immunology , Neutrophils/immunology , Neutrophils/metabolism , Periodontal Atrophy/immunology , Periodontal Atrophy/metabolism , Periodontitis/immunology , Periodontitis/therapy , Receptors, Interleukin-17/deficiency , Receptors, Interleukin-17/metabolism
3.
Immunity ; 37(4): 674-84, 2012 Oct 19.
Article in English | MEDLINE | ID: mdl-23063332

ABSTRACT

Mice lacking the transcription factor T-bet in the innate immune system develop microbiota-dependent colitis. Here, we show that interleukin-17A (IL-17A)-producing IL-7Rα(+) innate lymphoid cells (ILCs) were potent promoters of disease in Tbx21(-/-)Rag2(-/-) ulcerative colitis (TRUC) mice. TNF-α produced by CD103(-)CD11b(+) dendritic cells synergized with IL-23 to drive IL-17A production by ILCs, demonstrating a previously unrecognized layer of cellular crosstalk between dendritic cells and ILCs. We have identified Helicobacter typhlonius as a key disease trigger driving excess TNF-α production and promoting colitis in TRUC mice. Crucially, T-bet also suppressed the expression of IL-7R, a key molecule involved in controlling intestinal ILC homeostasis. The importance of IL-7R signaling in TRUC disease was highlighted by the dramatic reduction in intestinal ILCs and attenuated colitis following IL-7R blockade. Taken together, these data demonstrate the mechanism by which T-bet regulates the complex interplay between mucosal dendritic cells, ILCs, and the intestinal microbiota.


Subject(s)
Colitis, Ulcerative/immunology , DNA-Binding Proteins/immunology , Immunity, Innate , Lymphocytes/immunology , Receptors, Interleukin-7/immunology , T-Box Domain Proteins/immunology , Animals , Cells, Cultured , Chronic Disease , Colitis, Ulcerative/microbiology , Colitis, Ulcerative/pathology , DNA-Binding Proteins/deficiency , Helicobacter/immunology , Mice , Mice, Inbred BALB C , Mice, Knockout , Signal Transduction , T-Box Domain Proteins/deficiency
4.
Periodontol 2000 ; 86(1): 201-209, 2021 06.
Article in English | MEDLINE | ID: mdl-33690926

ABSTRACT

Recent advances in our understanding of the microbial populations that colonize the human mouth, their acquisition, interdependency, and coevolution with the host, bring a different perspective to the mechanisms underpinning the maintenance of periodontal health and the development of disease. In this work we suggest that our knowledge map of the etiology of periodontal health and disease can be viewed as a broad, highly connected, and integrated system that spans the entire spectrum of microbe/host/clinical interactions. The overall concept of present Periodontology 2000, that the microbial biofilm can be considered a human tissue of bacteriological origin, is entirely consistent with this integrated system view. The health-associated community structure of microbial biofilms can be considered a system that is normally resilient to perturbation. Equally, there is evidence to suggest that the dysbiotic community structure in disease may share similar resilience properties. In both instances, the resilience may be governed by the precise makeup of the acquired microbiome and by the genetics of the host. Understanding the mechanisms that enable the resistance to change of healthy and dysbiotic microbial populations may be important in the development of approaches to prevent the progression of disease and to restore health in diseased individuals.


Subject(s)
Dysbiosis , Microbiota , Biofilms , Humans , Mouth
5.
Periodontol 2000 ; 86(1): 8-13, 2021 06.
Article in English | MEDLINE | ID: mdl-33690952

ABSTRACT

The central theme of this volume of Periodontology 2000 is that the microbial dental plaque biofilm, specifically the subgingival dental plaque biofilm, mimics a human tissue in both structure and function. As a basis for this assertion we use the definition of a tissue as an aggregate of similar cells and cell products forming a defined structure with a specific function, in a multicellular organism. Accordingly, we propose that the dental plaque biofilm represents an acquired human tissue largely of bacterial origin that maintains the health of gingival tissue. Furthermore, we acknowledge that disease can be defined as a deviation from the normal structure or an interruption to the function of any body part, organ, or system, and that is manifested by a characteristic set of symptoms and signs whose etiology, pathology, and prognosis may be known or unknown. Therefore, in this volume we present the concept that periodontitis is a disruption of the normal function of the healthy subgingival plaque biofilm with concomitant disruption to its functional properties in relation to innate defense surveillance and tissue maintenance, leading to excessive, deregulated inflammation and tissue destruction.


Subject(s)
Dental Plaque , Periodontitis , Biofilms , Gingiva , Humans
6.
J Strength Cond Res ; 32(5): 1280-1287, 2018 May.
Article in English | MEDLINE | ID: mdl-29373427

ABSTRACT

Heishman, AD, Curtis, MA, Saliba, E, Hornett, RJ, Malin, SK, and Weltman, AL. Noninvasive assessment of internal and external player load: implications for optimizing athletic performance. J Strength Cond Res 32(5): 1280-1287, 2018-Few data exist that assess athlete tracking and monitoring for the development of strategies to optimize performance and reduce fatigue in elite athletes. The purpose of the present study was to assess the efficacy of external load and internal stress monitoring as assessment tools for examining a performance index of fatigue. A retrospective analysis was performed on data collected over the course of the preseason in 10 elite male NCAA Division 1 basketball players. Internal stress was assessed using Omegawave Technology readiness scores and compared with the performance index of countermovement jump (CMJ). The external load that accumulated during the previous practice, quantified by PlayerLoad (PL; Catapult), was compared with CMJ values and Omegawave scores. The results indicated that high, compared to low CNS Omegawave Readiness Scores (6.7 ± 05.1, 4.5 ± 1.2 AU; p < 0.001), were associated with increased CMJ (62.1 ± 6.5 vs. 59.4 ± 6.6 cm; p = 0.05), Power (6,590 ± 526.7 vs. 6,383.5 ± 606.8 W; p = 0.05), Omegawave Overall Readiness (5.8 ± 1.1 vs. 5.0 ± 0.7 AU; p = 0.05), and Omega Potential (Omega) (21.3 ± 6.3 vs. 9.9 ± 20.8 mV; p = 0.07). An increased PL during the previous exposure was associated with decreased CMJ (58.7 ± 4.7 cm vs. 60.4 ± 5.1 cm; p < 0.001) and increased TRIMP (135.1 ± 35.9 vs. 65.6 ± 20.0 AU; p < 0.001), and duration (115.4 ± 27.1 vs. 65.56 ± 20.0 minutes; p = < 0.001) despite no differences in Omegawave CNS Readiness scores. We conclude that Omegawave and Catapult technologies provide independent information related to performance and may be effective tools for monitoring athlete performance.


Subject(s)
Athletic Performance/physiology , Basketball/physiology , Muscle Fatigue/physiology , Athletes , Fatigue , Humans , Male , Muscle Strength , Retrospective Studies , Young Adult
7.
J Bacteriol ; 199(11)2017 06 01.
Article in English | MEDLINE | ID: mdl-28320881

ABSTRACT

Porphyromonas gingivalis produces outer membrane vesicles (OMVs) rich in virulence factors, including cysteine proteases and A-LPS, one of the two lipopolysaccharides (LPSs) produced by this organism. Previous studies had suggested that A-LPS and PG0027, an outer membrane (OM) protein, may be involved in OMV formation. Their roles in this process were examined by using W50 parent and the ΔPG0027 mutant strains. Inactivation of PG0027 caused a reduction in the yield of OMVs. Lipid A from cells and OMVs of P. gingivalis W50 and the ΔPG0027 mutant strains were analyzed by matrix-assisted laser desorption ionization-time of flight mass spectrometry (MALDI-TOF MS). Lipid A from W50 cells contained bis-P-pentaacyl, mono-P-pentaacyl, mono-P-tetraacyl, non-P-pentaacyl, and non-P-tetraacyl species, whereas lipid A from ΔPG0027 mutant cells contained only phosphorylated species; nonphosphorylated species were absent. MALDI-TOF/TOF tandem MS of mono-P-pentaacyl (m/z 1,688) and mono-P-tetraacyl (m/z 1,448) lipid A from ΔPG0027 showed that both contained lipid A 1-phosphate, suggesting that the ΔPG0027 mutant strain lacked lipid A 1-phosphatase activity. The total phosphatase activities in the W50 and the ΔPG0027 mutant strains were similar, whereas the phosphatase activity in the periplasm of the ΔPG0027 mutant was lower than that in W50, supporting a role for PG0027 in lipid A dephosphorylation. W50 OMVs were enriched in A-LPS, and its lipid A did not contain nonphosphorylated species, whereas lipid A from the ΔPG0027 mutant (OMVs and cells) contained similar species. Thus, OMVs in P. gingivalis are apparently formed in regions of the OM enriched in A-LPS devoid of nonphosphorylated lipid A. Conversely, dephosphorylation of lipid A through a PG0027-dependent process is required for optimal formation of OMVs. Hence, the relative proportions of nonphosphorylated and phosphorylated lipid A appear to be crucial for OMV formation in this organism.IMPORTANCE Gram-negative bacteria produce outer membrane vesicles (OMVs) by "blebbing" of the outer membrane (OM). OMVs can be used offensively as delivery systems for virulence factors and defensively to aid in the colonization of a host and in the survival of the bacterium in hostile environments. Earlier studies using the oral anaerobe Porphyromonas gingivalis as a model organism to study the mechanism of OMV formation suggested that the OM protein PG0027 and one of the two lipopolysaccharides (LPSs) synthesized by this organism, namely, A-LPS, played important roles in OMV formation. We suggest a novel mechanism of OMV formation in P. gingivalis involving dephosphorylation of lipid A of A-LPS controlled/regulated by PG0027, which causes destabilization of the OM, resulting in blebbing and generation of OMVs.


Subject(s)
Bacterial Proteins/metabolism , Phosphoric Monoester Hydrolases/metabolism , Porphyromonas gingivalis/metabolism , Bacterial Proteins/genetics , Gene Expression Regulation, Bacterial , Lipid A/biosynthesis , Phosphoric Monoester Hydrolases/genetics , Porphyromonas gingivalis/enzymology , Porphyromonas gingivalis/genetics
8.
J Clin Periodontol ; 44 Suppl 18: S5-S11, 2017 03.
Article in English | MEDLINE | ID: mdl-28266109

ABSTRACT

BACKGROUND AND AIMS: The scope of this working group was to review (1) ecological interactions at the dental biofilm in health and disease, (2) the role of microbial communities in the pathogenesis of periodontitis and caries, and (3) the innate host response in caries and periodontal diseases. RESULTS AND CONCLUSIONS: A health-associated biofilm includes genera such as Neisseria, Streptococcus, Actinomyces, Veillonella and Granulicatella. Microorganisms associated with both caries and periodontal diseases are metabolically highly specialized and organized as multispecies microbial biofilms. Progression of these diseases involves multiple microbial interactions driven by different stressors. In caries, the exposure of dental biofilms to dietary sugars and their fermentation to organic acids results in increasing proportions of acidogenic and aciduric species. In gingivitis, plaque accumulation at the gingival margin leads to inflammation and increasing proportions of proteolytic and often obligately anaerobic species. The natural mucosal barriers and saliva are the main innate defence mechanisms against soft tissue bacterial invasion. Similarly, enamel and dentin are important hard tissue barriers to the caries process. Given that the present state of knowledge suggests that the aetiologies of caries and periodontal diseases are mutually independent, the elements of innate immunity that appear to contribute to resistance to both are somewhat coincidental.


Subject(s)
Biofilms , Dental Caries/microbiology , Oral Health , Periodontitis/microbiology , Host-Pathogen Interactions , Humans
9.
J Strength Cond Res ; 31(6): 1557-1562, 2017 06.
Article in English | MEDLINE | ID: mdl-28538305

ABSTRACT

Time of day is a key factor that influences the optimization of athletic performance. Intercollegiate coaches oftentimes hold early morning strength training sessions for a variety of factors including convenience. However, few studies have specifically investigated the effect of early morning vs. late afternoon strength training on performance indices of fatigue. This is athletically important because circadian and/or ultradian rhythms and alterations in sleep patterns can affect training ability. Therefore, the purpose of the present study was to examine the effects of morning vs. afternoon strength training on an acute performance index of fatigue (countermovement jump height, CMJ), player readiness (Omegawave), and self-reported sleep quantity. We hypothesized that afternoon training sessions would be associated with increased levels of performance, readiness, and self-reported sleep. A retrospective analysis was performed on data collected over the course of the preseason on 10 elite National Collegiate Athletic Association Division 1 male basketball players. All basketball-related activities were performed in the afternoon with strength and conditioning activities performed either in the morning or in the afternoon. The average values for CMJ, power output (Power), self-reported sleep quantity (sleep), and player readiness were examined. When player load and duration were matched, CMJ (58.8 ± 1.3 vs. 61.9 ± 1.6 cm, p = 0.009), Power (6,378.0 ± 131.2 vs. 6,622.1 ± 172.0 W, p = 0.009), and self-reported sleep duration (6.6 ± 0.4 vs. 7.4 ± 0.25 p = 0.016) were significantly higher with afternoon strength and conditioning training, with no differences observed in player readiness values. We conclude that performance is suppressed with morning training and is associated with a decrease in self-reported quantity of sleep.


Subject(s)
Athletic Performance/physiology , Basketball/physiology , Circadian Rhythm/physiology , Muscle Strength/physiology , Resistance Training/methods , Fatigue/physiopathology , Humans , Male , Retrospective Studies , Sleep/physiology , Time Factors , Universities , Young Adult
10.
Appl Environ Microbiol ; 82(14): 4264-78, 2016 07 15.
Article in English | MEDLINE | ID: mdl-27208127

ABSTRACT

UNLABELLED: Intestinal homeostasis mechanisms must protect the host intestinal tissue from endogenous lipopolysaccharides (LPSs) produced by the intestinal microbiota. In this report, we demonstrate that murine intestinal fecal lipids effectively block Toll-like receptor 4 (TLR4) responses to naturally occurring Bacteroidetes sp. LPS. Cardiolipin (CL) represents a significant proportion of the total intestinal and fecal lipids and, furthermore, potently antagonizes TLR4 activation by reducing LPS binding at the lipopolysaccharide binding protein (LBP), CD14, and MD-2 steps of the TLR4 signaling pathway. It is further demonstrated that intestinal lipids and CL are less effective at neutralizing more potent Enterobacteriaceae-type LPS, which is enriched in feces obtained from mice with dextran sodium sulfate (DSS)-treated inflammatory bowel disease. The selective inhibition of naturally occurring LPS structures by intestinal lipids may represent a novel homeostasis mechanism that blocks LPS activation in response to symbiotic but not dysbiotic microbial communities. IMPORTANCE: The guts of animals harbor a variety of Gram-negative bacteria associated with both states of intestinal health and states of disease. Environmental factors, such as dietary habits, can drive the microbial composition of the host animal's intestinal bacterial community toward a more pathogenic state. Both beneficial and harmful Gram-negative bacteria are capable of eliciting potentially damaging inflammatory responses from the host intestinal tissues via a lipopolysaccharide (LPS)-dependent pathway. Physical mucosal barriers and antibodies produced by the intestinal immune system protect against the undesired inflammatory effects of LPS, although it is unknown why some bacteria are more effective at overcoming the protective barriers than others. This report describes the discovery of a lipid-type protective barrier in the intestine that reduces the deleterious effects of LPSs from beneficial bacteria but is less effective in dampening the inflammatory effects of LPSs from harmful bacteria, providing a novel mechanistic insight into inflammatory intestinal disorders.


Subject(s)
Cardiolipins/metabolism , Immunologic Factors/metabolism , Intestines/immunology , Intestines/microbiology , Lipopolysaccharides/immunology , Toll-Like Receptor 4/antagonists & inhibitors , Animals , Bacteroidetes/immunology , Enterobacteriaceae/immunology , Mice
11.
Exp Eye Res ; 145: 58-67, 2016 04.
Article in English | MEDLINE | ID: mdl-26474497

ABSTRACT

Nepafenac ophthalmic suspensions, 0.1% (NEVANAC(®)) and 0.3% (ILEVRO™), are topical nonsteroidal anti-inflammatory drug (NSAID) products approved in the United States, Europe and various other countries to treat pain and inflammation associated with cataract surgery. NEVANAC is also approved in Europe for the reduction in the risk of postoperative macular edema (ME) associated with cataract surgery in diabetic patients. The efficacy against ME suggests that topical administration leads to distribution of nepafenac or its active metabolite amfenac to the posterior segment of the eye. This article evaluates the ocular distribution of nepafenac and amfenac and the extent of local delivery to the posterior segment of the eye, following topical ocular instillation in animal models. Nepafenac ophthalmic suspension was instilled unilaterally in New Zealand White rabbits as either a single dose (0.1%; one drop) or as multiple doses (0.3%, one drop, once-daily for 4 days, or 0.1% one drop, three-times daily for 3 days and one morning dose on day 4). Nepafenac (0.3%) was also instilled unilaterally in cynomolgus monkeys as multiple doses (one drop, three-times daily for 7 days). Nepafenac and amfenac concentrations in harvested ocular tissues were measured using high-performance liquid chromatography/mass spectrometry. Locally-distributed compound concentrations were determined as the difference in levels between dosed and undosed eyes. In single-dosed rabbit eyes, peak concentrations of locally-distributed nepafenac and amfenac showed a trend of sclera > choroid > retina. Nepafenac peak levels in sub-samples posterior to the eye equator and inclusive of the posterior pole (E-PP) were 55.1, 4.03 and 2.72 nM, respectively, at 0.25 or 0.50 h, with corresponding amfenac peak levels of 41.9, 3.10 and 0.705 nM at 1 or 4 h. By comparison, peak levels in sclera, choroid and retina sub-samples in a band between the ora serrata and the equator (OS-E) were 13- to 40-fold (nepafenac) or 11- to 23-fold (amfenac) higher, indicating an anterior-to-posterior directional concentration gradient. In multiple-dosed rabbit eyes, with 0.3% nepafenac instilled once-daily or 0.1% nepafenac instilled three-times daily, cumulative 24-h locally-distributed levels of nepafenac in E-PP retina were similar between these groups, whereas exposure to amfenac once-daily dosing nepafenac 0.3% was 51% of that achieved with three-times daily dosing of 0.1%. In single-dosed monkey eyes, concentration gradients showed similar directionality as observed in rabbit eyes. Peak concentrations of locally-distributed nepafenac were 1580, 386, 292 and 13.8 nM in E-PP sclera, choroid and retina, vitreous humor, respectively, at 1 or 2 h after drug instillation. Corresponding amfenac concentrations were 21.3, 11.8, 2.58 and 2.82 nM, observed 1 or 2 h post-instillation. The data indicate that topically administered nepafenac and its metabolite amfenac reach pharmacologically relevant concentrations in the posterior eye segment (choroid and retina) via local distribution, following an anterior-to-posterior concentration gradient. The proposed pathway involves a choroidal/suprachoroidal or periocular route, along with an inward movement of drug through the sclera, choroid and retina, with negligible vitreal compartment involvement. Sustained high nepafenac concentrations in posterior segment tissues may be a reservoir for hydrolysis to amfenac.


Subject(s)
Benzeneacetamides/pharmacokinetics , Phenylacetates/pharmacokinetics , Posterior Eye Segment/metabolism , Uveitis, Posterior/drug therapy , Animals , Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Anti-Inflammatory Agents, Non-Steroidal/pharmacokinetics , Benzeneacetamides/administration & dosage , Chromatography, High Pressure Liquid , Disease Models, Animal , Instillation, Drug , Macaca fascicularis , Male , Ophthalmic Solutions , Phenylacetates/administration & dosage , Posterior Eye Segment/drug effects , Rabbits , Tissue Distribution , Uveitis, Posterior/metabolism
12.
J Bacteriol ; 197(10): 1735-46, 2015 May.
Article in English | MEDLINE | ID: mdl-25733619

ABSTRACT

UNLABELLED: Porphyromonas gingivalis synthesizes two lipopolysaccharides (LPSs), O-LPS and A-LPS. The structure of the core oligosaccharide (OS) of O-LPS and the attachment site of the O-polysaccharide (O-PS) repeating unit [ → 3)-α-D-Galp-(1 → 6)-α-D-Glcp-(1 → 4)-α-L-Rhap-(1 → 3)-ß-D-GalNAcp-(1 → ] to the core have been elucidated using the ΔPG1051 (WaaL, O-antigen ligase) and ΔPG1142 (Wzy, O-antigen polymerase) mutant strains, respectively. The core OS occurs as an "uncapped" glycoform devoid of O-PS and a "capped" glycoform that contains the attachment site of O-PS via ß-d-GalNAc at position O-3 of the terminal α-(1 → 3)-linked mannose (Man) residue. In this study, the attachment site of A-PS to the core OS was determined based on structural analysis of SR-type LPS (O-LPS and A-LPS) isolated from a P. gingivalis ΔPG1142 mutant strain by extraction with aqueous hot phenol to minimize the destruction of A-LPS. Application of one- and two-dimensional nuclear magnetic resonance (NMR) spectroscopy in combination with methylation analysis showed that the A-PS repeating unit is linked to a nonterminal α-(1 → 3)-linked Man of the "capped core" glycoform of outer core OS at position O-4 via a → 6)-[α-D-Man-α-(1 → 2)-α-D-Man-1-phosphate → 2]-α-D-Man-(1 → motif. In order to verify that O-PS and A-PS are attached to almost identical core glycoforms, we identified a putative α-mannosyltransferase (PG0129) in P. gingivalis W50 that may be involved in the formation of core OS. Inactivation of PG0129 led to the synthesis of deep-R-type LPS with a truncated core that lacks α-(1 → 3)-linked mannoses and is devoid of either O-PS or A-PS. This indicated that PG0129 is an α-1,3-mannosyltransferase required for synthesis of the outer core regions of both O-LPS and A-LPS in P. gingivalis. IMPORTANCE: Porphyromonas gingivalis, a Gram-negative anaerobe, is considered to be an important etiologic agent in periodontal disease, and among the virulence factors produced by the organism are two lipopolysaccharides (LPSs), O-LPS and A-LPS. The structures of the O-PS and A-PS repeating units, the core oligosaccharide (OS), and the linkage of the O-PS repeating unit to the core OS in O-LPS have been elucidated by our group. It is important to establish whether the attachment site of the A-PS repeating unit to the core OS in A-LPS is similar to or differs from that of the O-PS repeating unit in O-LPS. As part of understanding the biosynthetic pathway of the two LPSs in P. gingivalis, PG0129 was identified as an α-mannosyltransferase that is involved in the synthesis of the outer core regions of both O-LPS and A-LPS.


Subject(s)
Lipopolysaccharides/chemistry , Lipopolysaccharides/metabolism , Porphyromonas gingivalis/chemistry , Porphyromonas gingivalis/metabolism , Gene Knockout Techniques , Magnetic Resonance Spectroscopy , Mannosyltransferases/genetics , Mannosyltransferases/metabolism , Porphyromonas gingivalis/enzymology , Porphyromonas gingivalis/genetics
13.
Am J Pathol ; 184(8): 2333-41, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24979595

ABSTRACT

Because there is clinical evidence for an association between periodontal disease and rheumatoid arthritis, it is important to develop suitable experimental models to explore pathogenic mechanisms and therapeutic opportunities. The K/BxN serum model of inflammatory arthritis was applied using distinct protocols, and modulation of joint disruption afforded by dexamethasone and calcitonin was established in comparison to the melanocortin (MC) receptor agonist DTrp(8)-γ-melanocyte stimulating hormone (MSH; DTrp). Wild-type and MC receptor type 3 (MC3)-null mice of different ages were also used. There was significant association between severity of joint disease, induced with distinct protocols and volumes of the arthritogenic K/BxN serum, and periodontal bone damage. Therapeutic treatment with 10 µg dexamethasone, 30 ng elcatonin, and 20 µg DTrp per mouse revealed unique and distinctive pharmacological properties, with only DTrp protecting both joint and periodontal tissue. Further analyses in nonarthritic animals revealed higher susceptibility to periodontal bone loss in Mc3r(-/-) compared with wild-type mice, with significant exacerbation at 14 weeks of age. These data reveal novel protective properties of endogenous MC3 on periodontal status in health and disease and indicate that MC3 activation could lead to the development of a new genus of anti-arthritic bone-sparing therapeutics.


Subject(s)
Arthritis, Experimental/metabolism , Arthritis, Rheumatoid/metabolism , Periodontal Diseases/metabolism , Receptor, Melanocortin, Type 3/metabolism , Animals , Arthritis, Experimental/complications , Arthritis, Rheumatoid/complications , Disease Models, Animal , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Periodontal Diseases/complications
14.
J Antimicrob Chemother ; 69(12): 3236-43, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25134721

ABSTRACT

OBJECTIVES: Effective treatment of Gram-negative bacterial infections is increasingly challenging due to the spread of multidrug-resistant strains and a lack of new antimicrobials in development. Bacterial type I signal peptidases (SPases) represent a highly conserved and essential target for inhibition by novel compounds. SPases are required for the effective processing of membrane translocated proteins involved in core functions related to metabolism, virulence and resistance. In this study we assessed the biochemical and functional activity of a novel synthetic inhibitor (MD3) of SPases against a wide range of Gram-negative pathogens. METHODS: The activity and specificity of MD3 for recombinant Pseudomonas aeruginosa SPase (LepB) and a genetically engineered LepB-regulatable strain were investigated. Antimicrobial activity of the compound alone and in combination with outer membrane-permeabilizing agents (sodium hexametaphosphate, colistin) was also determined against a collection of P. aeruginosa, Acinetobacter baumannii, Klebsiella pneumoniae and Stenotrophomonas maltophilia isolates. RESULTS: MD3 was found to inactivate the P. aeruginosa LepB protein (IC50 10 µM), resulting in antimicrobial effects potentiated in the presence of colistin. MD3 also demonstrated potent activity against wild-type and multidrug-resistant strains of A. baumannii and S. maltophilia with MICs ranging from 0.5 to 14 mg/L in the presence of subinhibitory concentrations of colistin. CONCLUSIONS: MD3 is a novel inhibitor of bacterial SPase in a range of non-fermentative Gram-negative bacteria. The antimicrobial activity is potentiated in combination with colistin and suggests that SPase inhibition warrants further exploration as a basis for future mono or combination therapies.


Subject(s)
Anti-Bacterial Agents/pharmacology , Colistin/pharmacology , Enzyme Inhibitors/pharmacology , Gram-Negative Bacteria/drug effects , Gram-Negative Bacteria/enzymology , Membrane Proteins/antagonists & inhibitors , Drug Resistance, Multiple, Bacterial , Drug Synergism , Inhibitory Concentration 50 , Microbial Sensitivity Tests , Serine Endopeptidases
15.
BMC Microbiol ; 14: 258, 2014 Oct 02.
Article in English | MEDLINE | ID: mdl-25270662

ABSTRACT

BACKGROUND: Periodontal diseases are polymicrobial diseases that cause the inflammatory destruction of the tooth-supporting (periodontal) tissues. Their initiation is attributed to the formation of subgingival biofilms that stimulate a cascade of chronic inflammatory reactions by the affected tissue. The Gram-negative anaerobes Porphyromonas gingivalis, Tannerella forsythia and Treponema denticola are commonly found as part of the microbiota of subgingival biofilms, and they are associated with the occurrence and severity of the disease. P. gingivalis expresses several virulence factors that may support its survival, regulate its communication with other species in the biofilm, or modulate the inflammatory response of the colonized host tissue. The most prominent of these virulence factors are the gingipains, which are a set of cysteine proteinases (either Arg-specific or Lys-specific). The role of gingipains in the biofilm-forming capacity of P. gingivalis is barely investigated. Hence, this in vitro study employed a biofilm model consisting of 10 "subgingival" bacterial species, incorporating either a wild-type P. gingivalis strain or its derivative Lys-gingipain and Arg-gingipan isogenic mutants, in order to evaluate quantitative and qualitative changes in biofilm composition. RESULTS: Following 64 h of biofilm growth, the levels of all 10 species were quantified by fluorescence in situ hybridization or immunofluorescence. The wild-type and the two gingipain-deficient P. gingivalis strains exhibited similar growth in their corresponding biofilms. Among the remaining nine species, only the numbers of T. forsythia were significantly reduced, and only when the Lys-gingipain mutant was present in the biofilm. When evaluating the structure of the biofilm by confocal laser scanning microscopy, the most prominent observation was a shift in the spatial arrangement of T. denticola, in the presence of P. gingivalis Arg-gingipain mutant. CONCLUSIONS: The gingipains of P. gingivalis may qualitatively and quantitatively affect composition of polymicrobial biofilms. The present experimental model reveals interdependency between the gingipains of P. gingivalis and T. forsythia or T. denticola.


Subject(s)
Adhesins, Bacterial/metabolism , Biofilms/growth & development , Cysteine Endopeptidases/metabolism , Microbial Consortia , Porphyromonas gingivalis/physiology , Treponema denticola/physiology , Virulence Factors/metabolism , Adhesins, Bacterial/genetics , Cysteine Endopeptidases/genetics , Gingipain Cysteine Endopeptidases , Mutant Proteins/genetics , Mutant Proteins/metabolism , Porphyromonas gingivalis/metabolism , Treponema denticola/metabolism , Virulence Factors/genetics
16.
Methods Mol Biol ; 2778: 331-344, 2024.
Article in English | MEDLINE | ID: mdl-38478287

ABSTRACT

The type 9 secretion system (T9SS) is a recently discovered machinery that both transports cargo proteins across the Gram-negative bacterial outer membrane and attaches them to lipopolysaccharides on the extracellular surface. Outer membrane proteins (OMPs) are key components of the T9SS and are involved in both steps. In this chapter, we describe a method for the in silico modeling of T9SS OMPs and their complexes, and model validation. This is useful when the production of recombinant OMPs is difficult, and these protocols can also be applied to OMP complexes outside of the T9SS.


Subject(s)
Bacterial Outer Membrane Proteins , Membrane Proteins , Bacterial Outer Membrane Proteins/metabolism , Bacterial Proteins/metabolism
17.
Mucosal Immunol ; 17(1): 1-12, 2024 Feb.
Article in English | MEDLINE | ID: mdl-37952849

ABSTRACT

Type-3 innate lymphoid cells (ILC3) respond to localized environmental cues to regulate homeostasis and orchestrate immunity in the intestine. The intestinal epithelium is an important upstream regulator and downstream target of ILC3 signaling, however, the complexity of mucosal tissues can hinder efforts to define specific interactions between these two compartments. Here, we employ a reductionist co-culture system of murine epithelial small intestinal organoids (SIO) with ILC3 to uncover bi-directional signaling mechanisms that underlie intestinal homeostasis. We report that ILC3 induce global transcriptional changes in intestinal epithelial cells, driving the enrichment of secretory goblet cell signatures. We find that SIO enriched for goblet cells promote NKp46+ ILC3 and interleukin (IL)-22 expression, which can feedback to induce IL-22-mediated epithelial transcriptional signatures. However, we show that epithelial regulation of ILC3 in this system is contact-dependent and demonstrate a role for epithelial Delta-Like-Canonical-Notch-Ligand (Dll) in driving IL-22 production by ILC3, via subset-specific Notch1-mediated activation of T-bet+ ILC3. Finally, by interfering with Notch ligand-receptor dynamics, ILC3 appear to upregulate epithelial Atoh1 to skew secretory lineage determination in SIO-ILC3 co-cultures. This research outlines two complimentary bi-directional signaling modules between the intestinal epithelium and ILC3, which may be relevant in intestinal homeostasis and disease.


Subject(s)
Interleukin-22 , Lymphocytes , Mice , Animals , Immunity, Innate , Ligands , Intestinal Mucosa , Receptors, Notch/metabolism
18.
J Bacteriol ; 195(23): 5297-307, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24056103

ABSTRACT

Mannose is an important sugar in the biology of the Gram-negative bacterium Porphyromonas gingivalis. It is a major component of the oligosaccharides attached to the Arg-gingipain cysteine proteases, the repeating units of an acidic lipopolysaccharide (A-LPS), and the core regions of both types of LPS produced by the organism (O-LPS and A-LPS) and a reported extracellular polysaccharide (EPS) isolated from spent culture medium. The organism occurs at inflamed sites in periodontal tissues, where it is exposed to host glycoproteins rich in mannose, which may be substrates for the acquisition of mannose by P. gingivalis. Five potential mannosidases were identified in the P. gingivalis W83 genome that may play a role in mannose acquisition. Four mannosidases were characterized in this study: PG0032 was a ß-mannosidase, whereas PG0902 and PG1712 were capable of hydrolyzing p-nitrophenyl α-d-mannopyranoside. PG1711 and PG1712 were α-1 → 3 and α-1 → 2 mannosidases, respectively. No enzyme function could be assigned to PG0973. α-1 → 6 mannobiose was not hydrolyzed by P. gingivalis W50. EPS present in the culture supernatant was shown to be identical to yeast mannan and a component of the medium used for culturing P. gingivalis and was resistant to hydrolysis by mannosidases. Synthesis of O-LPS and A-LPS and glycosylation of the gingipains appeared to be unaffected in all mutants. Thus, α- and ß-mannosidases of P. gingivalis are not involved in the harnessing of mannan/mannose from the growth medium for these biosynthetic processes. P. gingivalis grown in chemically defined medium devoid of carbohydrate showed reduced α-mannosidase activity (25%), suggesting these enzymes are environmentally regulated.


Subject(s)
Porphyromonas gingivalis/enzymology , alpha-Mannosidase/metabolism , beta-Mannosidase/metabolism , Adhesins, Bacterial/genetics , Adhesins, Bacterial/metabolism , Cysteine Endopeptidases/genetics , Cysteine Endopeptidases/metabolism , Gene Expression Regulation, Bacterial/physiology , Gene Expression Regulation, Enzymologic/physiology , Gingipain Cysteine Endopeptidases , Magnetic Resonance Spectroscopy , Mutation , Porphyromonas gingivalis/genetics , Porphyromonas gingivalis/metabolism , alpha-Mannosidase/genetics , beta-Mannosidase/genetics
19.
mSystems ; 8(4): e0119322, 2023 08 31.
Article in English | MEDLINE | ID: mdl-37436062

ABSTRACT

Periodontal disease is a chronic inflammatory disease in which the oral pathogen Porphyromonas gingivalis plays an important role. Porphyromonas gingivalis expresses virulence determinants in response to higher hemin concentrations, but the underlying regulatory processes remain unclear. Bacterial DNA methylation has the potential to fulfil this mechanistic role. We characterized the methylome of P. gingivalis, and compared its variation to transcriptome changes in response to hemin availability. Porphyromonas gingivalis W50 was grown in chemostat continuous culture with excess or limited hemin, prior to whole-methylome and transcriptome profiling using Nanopore and Illumina RNA-Seq. DNA methylation was quantified for Dam/Dcm motifs and all-context N6-methyladenine (6mA) and 5-methylcytosine (5mC). Of all 1,992 genes analyzed, 161 and 268 were respectively over- and under-expressed with excess hemin. Notably, we detected differential DNA methylation signatures for the Dam "GATC" motif and both all-context 6mA and 5mC in response to hemin availability. Joint analyses identified a subset of coordinated changes in gene expression, 6mA, and 5mC methylation that target genes involved in lactate utilization and ABC transporters. The results identify altered methylation and expression responses to hemin availability in P. gingivalis, with insights into mechanisms regulating its virulence in periodontal disease. IMPORTANCE DNA methylation has important roles in bacteria, including in the regulation of transcription. Porphyromonas gingivalis, an oral pathogen in periodontitis, exhibits well-established gene expression changes in response to hemin availability. However, the regulatory processes underlying these effects remain unknown. We profiled the novel P. gingivalis epigenome, and assessed epigenetic and transcriptome variation under limited and excess hemin conditions. As expected, multiple gene expression changes were detected in response to limited and excess hemin that reflect health and disease, respectively. Notably, we also detected differential DNA methylation signatures for the Dam "GATC" motif and both all-context 6mA and 5mC in response to hemin. Joint analyses identified coordinated changes in gene expression, 6mA, and 5mC methylation that target genes involved in lactate utilization and ABC transporters. The results identify novel regulatory processes underlying the mechanism of hemin regulated gene expression in P. gingivalis, with phenotypic impacts on its virulence in periodontal disease.


Subject(s)
Hemin , Periodontal Diseases , Humans , Hemin/pharmacology , Porphyromonas gingivalis/genetics , DNA Methylation/genetics , Periodontal Diseases/genetics , ATP-Binding Cassette Transporters/genetics , Gene Expression
20.
J Bacteriol ; 194(17): 4521-36, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22730125

ABSTRACT

Type I signal peptidases (SPases) cleave signal peptides from proteins during translocation across biological membranes and hence play a vital role in cellular physiology. SPase activity is also of fundamental importance to the pathogenesis of infection for many bacteria, including Pseudomonas aeruginosa, which utilizes a variety of secreted virulence factors, such as proteases and toxins. P. aeruginosa possesses two noncontiguous SPase homologues, LepB (PA0768) and PA1303, which share 43% amino acid identity. Reverse transcription (RT)-PCR showed that both proteases were expressed, while a FRET-based assay using a peptide based on the signal sequence cleavage region of the secreted LasB elastase showed that recombinant LepB and PA1303 enzymes were both active. LepB is positioned within a genetic locus that resembles the locus containing the extensively characterized SPase of E. coli and is of similar size and topology. It was also shown to be essential for viability and to have high sequence identity with SPases from other pseudomonads (≥ 78%). In contrast, PA1303, which is small for a Gram-negative SPase (20 kDa), was found to be dispensable. Mutation of PA1303 resulted in an altered protein secretion profile and increased N-butanoyl homoserine lactone production and influenced several quorum-sensing-controlled phenotypic traits, including swarming motility and the production of rhamnolipid and elastinolytic activity. The data indicate different cellular roles for these P. aeruginosa SPase paralogues; the role of PA1303 is integrated with the quorum-sensing cascade and includes the suppression of virulence factor secretion and virulence-associated phenotypes, while LepB is the primary SPase.


Subject(s)
Membrane Proteins/chemistry , Membrane Proteins/metabolism , Pseudomonas aeruginosa/enzymology , Pseudomonas aeruginosa/pathogenicity , Serine Endopeptidases/chemistry , Serine Endopeptidases/metabolism , 4-Butyrolactone/analogs & derivatives , 4-Butyrolactone/biosynthesis , Amino Acid Sequence , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Biological Transport , Gene Expression Regulation, Bacterial , Membrane Proteins/genetics , Microbial Viability , Molecular Sequence Data , Mutation , Phenotype , Protein Sorting Signals , Pseudomonas aeruginosa/metabolism , Quorum Sensing , Recombinant Proteins , Sequence Alignment , Serine Endopeptidases/genetics , Virulence Factors/genetics
SELECTION OF CITATIONS
SEARCH DETAIL