RESUMEN
Granulomas are complex cellular structures composed predominantly of macrophages and lymphocytes that function to contain and kill invading pathogens. Here, we investigated the single-cell phenotypes associated with antimicrobial responses in human leprosy granulomas by applying single-cell and spatial sequencing to leprosy biopsy specimens. We focused on reversal reactions (RRs), a dynamic process whereby some patients with disseminated lepromatous leprosy (L-lep) transition toward self-limiting tuberculoid leprosy (T-lep), mounting effective antimicrobial responses. We identified a set of genes encoding proteins involved in antimicrobial responses that are differentially expressed in RR versus L-lep lesions and regulated by interferon-γ and interleukin-1ß. By integrating the spatial coordinates of the key cell types and antimicrobial gene expression in RR and T-lep lesions, we constructed a map revealing the organized architecture of granulomas depicting compositional and functional layers by which macrophages, T cells, keratinocytes and fibroblasts can each contribute to the antimicrobial response.
Asunto(s)
Lepra Lepromatosa/inmunología , Lepra Tuberculoide/inmunología , Mycobacterium leprae/inmunología , Piel/inmunología , Adolescente , Adulto , Anciano , Femenino , Fibroblastos/inmunología , Fibroblastos/microbiología , Fibroblastos/patología , Perfilación de la Expresión Génica , Interacciones Huésped-Patógeno , Humanos , Queratinocitos/inmunología , Queratinocitos/microbiología , Queratinocitos/patología , Lepra Lepromatosa/genética , Lepra Lepromatosa/microbiología , Lepra Lepromatosa/patología , Lepra Tuberculoide/genética , Lepra Tuberculoide/microbiología , Lepra Tuberculoide/patología , Macrófagos/inmunología , Macrófagos/microbiología , Macrófagos/patología , Masculino , Persona de Mediana Edad , Mycobacterium leprae/patogenicidad , RNA-Seq , Análisis de la Célula Individual , Piel/microbiología , Piel/patología , Linfocitos T/inmunología , Linfocitos T/microbiología , Linfocitos T/patología , TranscriptomaRESUMEN
Acne is one of the most common skin conditions worldwide, with multifactorial origins it affects areas of the skin with hair follicles and sebaceous glands that become clogged. Bacterial incidence aggravates treatment due to resistance to antimicrobial agents and production of virulence factors such as biofilm formation. Based on these information, this study aims to conduct in vitro evaluations of the antibacterial activity of essential oils (EOs), alone and in combination, against Propionibacterium acnes, Staphylococcus aureus, and Staphylococcus epidermidis in planktonic and biofilm forms. This study also assessed the anti-inflammatory potential (TNF-α) and the effects of EOs on the viability of human keratinocytes (HaCaT), murine fibroblasts (3T3-L1), and bone marrow-derived macrophages (BMDMs). Of all EOs tested, 13 had active action against P. acnes, 9 against S. aureus, and 9 against S. epidermidis at concentrations of 0.125-2.0 mg/mL. Among the most active plant species, a blend of essential oil (BEOs) was selected, with Cymbopogon martini (Roxb.) Will. Watson, Eugenia uniflora L., and Varronia curassavica Jacq., the latter due to its anti-inflammatory action. This BEOs showed higher inhibition rates when compared to chloramphenicol against S. aureus and S. epidermidis, and higher eradication rates when compared to chloramphenicol for the three target species. The BEOs did not affect the cell viability of cell lines evaluated, and the levels of TNF-α decreased. According to these results, the BEOs evaluated showed potential for the development of an alternative natural formulation for the treatment of acne.
Asunto(s)
Acné Vulgar , Antibacterianos , Antiinflamatorios , Biopelículas , Queratinocitos , Macrófagos , Pruebas de Sensibilidad Microbiana , Aceites Volátiles , Propionibacterium acnes , Staphylococcus aureus , Staphylococcus epidermidis , Factor de Necrosis Tumoral alfa , Biopelículas/efectos de los fármacos , Biopelículas/crecimiento & desarrollo , Aceites Volátiles/farmacología , Humanos , Acné Vulgar/microbiología , Acné Vulgar/tratamiento farmacológico , Ratones , Antiinflamatorios/farmacología , Antibacterianos/farmacología , Propionibacterium acnes/efectos de los fármacos , Staphylococcus epidermidis/efectos de los fármacos , Animales , Staphylococcus aureus/efectos de los fármacos , Queratinocitos/efectos de los fármacos , Queratinocitos/microbiología , Macrófagos/efectos de los fármacos , Macrófagos/microbiología , Factor de Necrosis Tumoral alfa/metabolismo , Fibroblastos/efectos de los fármacos , Fibroblastos/microbiología , Supervivencia Celular/efectos de los fármacos , Células HaCaT , Línea Celular , Aceites de Plantas/farmacologíaRESUMEN
This research aimed to address the potential bacterial contamination risks in developmental engineering (DE) using bacteriophages. To compare and contrast the exemplar Escherichia coli T4 and M13 bacteriophages, human dermal fibroblasts cultivated on culture plates, natural cellulosic scaffolds, and poly(methyl methacrylate) (PMMA) particles were utilized as two-dimensional (2D) cell, three-dimensional (3D) tissue, and modular tissue culture models, respectively. When directly introduced into these distinct culture systems, both phages survived, exhibited no significant effects on the cultured cells or tissues, yet displayed their potentials to alleviate the infections caused by corresponding bacterial host cells. Apart from direct addition into the culture medium, both phages were also coated on PMMA, polystyrene, poly(lactic acid) particles with different diameters (5, 10, 30, and 100 µm) and cellulosic scaffolds. The coated phages endured the coating processes and demonstrated their viabilities in plaque assays. Further testing indicated that the phages coated on the PMMA particles tolerated multiple deliberate rinses and centrifugations, but not thermal treatment at 60-80°C. In summary, T4 and M13 bacteriophages not only manifested their antibacterial functions in diverse 2D cell, 3D tissue, and modular tissue culture systems, but also demonstrated their potentials of coating modular scaffolds to alleviate the bacterial contamination risks in DE.
Asunto(s)
Escherichia coli , Humanos , Escherichia coli/virología , Escherichia coli/crecimiento & desarrollo , Ingeniería de Tejidos/métodos , Fibroblastos/virología , Fibroblastos/microbiología , Bacteriófago M13 , Células CultivadasRESUMEN
BACKGROUND: Recurrent bone and joint infection with Staphylococcus aureus is common. S. aureus can invade and persist in osteoblasts and fibroblasts, but little is known about this mechanism in chondrocytes. If S. aureus were able to invade and persist within chondrocytes, this could be a difficult compartment to treat. QUESTION/PURPOSE: Can S. aureus infiltrate and persist intracellularly within chondrocytes in vitro? METHODS: Cell lines were cultured in vitro and infected with S. aureus. Human chondrocytes (C20A4) were compared with positive controls of human osteoblasts (MG63) and mouse fibroblasts (NIH3T3), which have previously demonstrated S. aureus invasion and persistence (human fibroblasts were not available to us). Six replicates per cell type were followed for 6 days after infection. Cells were treated daily with antibiotic media for extracellular killing. To determine whether S. aureus can infiltrate chondrocytes, fluorescence microscopy was performed to qualitatively assess the presence of intracellular bacteria, and intracellular colony-forming units (CFU) were enumerated 2 hours after infection. To determine whether S. aureus can persist within chondrocytes, intracellular CFUs were enumerated from infected host cells each day postinfection. RESULTS: S. aureus invaded human chondrocytes (C20A4) at a level (2.8 x 10 5 ± 5.5 x 10 4 CFUs/mL) greater than positive controls of human osteoblasts (MG63) (9.5 x 10 2 ± 2.5 x 10 2 CFUs/mL; p = 0.01) and mouse fibroblasts (NIH3T3) (9.1 x 10 4 ± 2.5 x 10 4 CFUs/mL; p = 0.02). S. aureus also persisted within human chondrocytes (C20A4) for 6 days at a level (1.4 x 10 3 ± 5.3 x 10 2 CFUs/mL) greater than that of human osteoblasts (MG63) (4.3 x 10 2 ± 3.5 x 10 1 CFUs/mL; p = 0.02) and mouse fibroblasts (NIH3T3) (0 CFUs/mL; p < 0.01). S. aureus was undetectable within mouse fibroblasts (NIH3T3) after 4 days. There were 0 CFUs yielded from cell media, confirming extracellular antibiotic treatment was effective. CONCLUSION: S. aureus readily invaded human chondrocytes (C20A4) in vitro and persisted viably for 6 days after infection, evading extracellular antibiotics. Chondrocytes demonstrated a greater level of intracellular invasion and persistence by S. aureus than positive control human osteoblast (MG63) and mouse fibroblast (NIH3T3) cell lines. CLINICAL RELEVANCE: Chondrocyte invasion and persistence may contribute to recurrent bone and joint infections. Additional research should assess longer periods of persistence and whether this mechanism is present in vivo.
Asunto(s)
Condrocitos , Osteoblastos , Infecciones Estafilocócicas , Staphylococcus aureus , Humanos , Condrocitos/microbiología , Condrocitos/patología , Animales , Staphylococcus aureus/fisiología , Ratones , Osteoblastos/microbiología , Infecciones Estafilocócicas/microbiología , Infecciones Estafilocócicas/tratamiento farmacológico , Células 3T3 NIH , Antibacterianos/farmacología , Factores de Tiempo , Fibroblastos/microbiología , Fibroblastos/patología , Línea CelularRESUMEN
Human cytomegalovirus (HCMV) microRNAs (miRNAs) significantly rewire host signaling pathways to support the viral lifecycle and regulate host cell responses. Here we show that SMAD3 expression is regulated by HCMV miR-UL22A and contributes to the IRF7-mediated induction of type I IFNs and IFN-stimulated genes (ISGs) in human fibroblasts. Addition of exogenous TGFß interferes with the replication of a miR-UL22A mutant virus in a SMAD3-dependent manner in wild type fibroblasts, but not in cells lacking IRF7, indicating that downregulation of SMAD3 expression to limit IFN induction is important for efficient lytic replication. These findings uncover a novel interplay between SMAD3 and innate immunity during HCMV infection and highlight the role of viral miRNAs in modulating these responses.
Asunto(s)
Infecciones por Citomegalovirus/microbiología , Citomegalovirus/fisiología , Fibroblastos/microbiología , Inmunidad Innata/inmunología , Interferón Tipo I/metabolismo , MicroARNs/genética , Factor de Crecimiento Transformador beta/metabolismo , Fibroblastos/inmunología , Fibroblastos/patología , Interacciones Huésped-Patógeno , Humanos , Factores Reguladores del Interferón/genética , Factores Reguladores del Interferón/metabolismo , Interferón Tipo I/genética , Proteína smad3/genética , Proteína smad3/metabolismo , Factor de Crecimiento Transformador beta/genética , Fenómenos Fisiológicos de los VirusRESUMEN
The protein LC3 is indispensible for the cellular recycling process of autophagy and plays critical roles during cargo recruitment, autophagosome biogenesis, and completion. Here, we report that LC3 is phosphorylated at threonine 50 (Thr(50)) by the mammalian Sterile-20 kinases STK3 and STK4. Loss of phosphorylation at this site blocks autophagy by impairing fusion of autophagosomes with lysosomes, and compromises the ability of cells to clear intracellular bacteria, an established cargo for autophagy. Strikingly, mutation of LC3 mimicking constitutive phosphorylation at Thr(50) reverses the autophagy block in STK3/STK4-deficient cells and restores their capacity to clear bacteria. Loss of STK3/STK4 impairs autophagy in diverse species, indicating that these kinases are conserved autophagy regulators. We conclude that phosphorylation of LC3 by STK3/STK4 is an essential step in the autophagy process. Since several pathological conditions, including bacterial infections, display aberrant autophagy, we propose that pharmacological agents targeting this regulatory circuit hold therapeutic potential.
Asunto(s)
Autofagia/genética , Fibroblastos/metabolismo , Proteínas Asociadas a Microtúbulos/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Animales , Caenorhabditis elegans/genética , Caenorhabditis elegans/metabolismo , Células Cultivadas , Embrión de Mamíferos , Fibroblastos/microbiología , Regulación de la Expresión Génica , Humanos , Lisosomas/metabolismo , Fusión de Membrana , Ratones , Ratones Noqueados , Proteínas Asociadas a Microtúbulos/genética , Mutación , Fragmentos de Péptidos/química , Fagosomas/metabolismo , Fosforilación , Proteínas Serina-Treonina Quinasas/deficiencia , Serina-Treonina Quinasa 3 , Transducción de Señal , Streptococcus pyogenes/patogenicidad , Streptococcus pyogenes/fisiología , Treonina/metabolismoRESUMEN
Chronic wounds are a major disease burden worldwide. The breach of the epithelial barrier facilitates transition of skin commensals to invasive facultative pathogens. Therefore, we investigated the potential effects of Staphylococcus aureus (SA) on dermal fibroblasts as key cells for tissue repair. In co-culture systems combining live or heat-killed SA with dermal fibroblasts derived from the BJ-5ta cell line, healthy individuals, and patients with systemic sclerosis, we assessed tissue repair including pro-inflammatory cytokines, matrix metalloproteases (MMPs), myofibroblast functions, and host defense responses. Only live SA induced the upregulation of IL-1ß/-6/-8 and MMP1/3 as co-factors of tissue degradation. Additionally, the increased cell death reduced collagen production, proliferation, migration, and contractility, prerequisite mechanisms for wound closure. Intracellular SA triggered inflammatory and type I IFN responses via intracellular dsDNA sensor molecules and MyD88 and STING signaling pathways. In conclusion, live SA affected various key tissue repair functions of dermal fibroblasts from different sources to a similar extent. Thus, SA infection of dermal fibroblasts should be taken into account for future wound management strategies.
Asunto(s)
Fibroblastos/patología , Enfermedades Cutáneas Infecciosas/patología , Piel/patología , Infecciones Estafilocócicas/complicaciones , Staphylococcus aureus/patogenicidad , Cicatrización de Heridas , Adulto , Anciano , Estudios de Casos y Controles , Movimiento Celular , Proliferación Celular , Células Cultivadas , Femenino , Fibroblastos/microbiología , Humanos , Masculino , Persona de Mediana Edad , Piel/microbiología , Enfermedades Cutáneas Infecciosas/microbiología , Infecciones Estafilocócicas/microbiología , Adulto JovenRESUMEN
OBJECTIVE: Uncontrolled production of Interleukin-1ß (IL-1ß), a major proinflammatory cytokine, is associated with tissue destruction in periodontal disease. IL-1ß production is controlled by inflammasomes which are multiprotein regulatory complexes. The current study aimed to elucidate potential regulatory pathways by monitoring the effects of periodontal pathogens Fusobacterium nucleatum (Fn) and Porphyromonas gingivalis (Pg) on inflammasomes and their regulators in human gingival fibroblasts (HGFs) in vitro. METHODS: HGFs were exposed to Fn and Pg alone or in combination for 24 hr at a multiplicity of infection of 100, ±30 min exposure with 5 mM adenosine triphosphate (ATP) incubation. Gene expression of NLRP3 and AIM2, inflammasome regulatory proteins POP1, CARD16 and TRIM16, and inflammasome components ASC and CASPASE 1, and IL-1ß, were evaluated by RT-PCR. Pro- and mature IL-1ß levels were monitored intracellularly by immunocytochemistry and extracellularly by ELISA. RESULTS: Fn + ATP significantly upregulated NLRP3, AIM2, IL-1ß, ASC, and CASPASE 1; however, it downregulated POP1 and TRIM16. Pg + ATP downregulated NLRP3, ASC, POP1, but upregulated IL-1ß and CARD16. Pg + Fn+ATP significantly upregulated AIM2, IL-1ß and CARD16, and downregulated POP1, TRIM16, and CASPASE 1. Pg + ATP exposure significantly increased pro- and mature IL-1ß production. CONCLUSION: Bacterial exposure with ATP may deregulate IL-1ß by dysregulating inflammasomes and their regulators in HGFs.
Asunto(s)
Fibroblastos/inmunología , Inflamasomas , Proteína con Dominio Pirina 3 de la Familia NLR , Células Cultivadas , Fibroblastos/microbiología , Fusobacterium nucleatum/patogenicidad , Encía/citología , Humanos , Interleucina-1beta , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Porphyromonas gingivalis/patogenicidadRESUMEN
BACKGROUND: Salmonella can invade host cells via a type three secretion system called T3SS-1 and its outer membrane proteins, PagN and Rck. However, the mechanism of PagN-dependent invasion pathway used by Salmonella enterica, subspecies enterica serovar Typhimurium remains unclear. RESULTS: Here, we report that PagN is well conserved and widely distributed among the different species and subspecies of Salmonella. We showed that PagN of S. Typhimurium was sufficient and necessary to enable non-invasive E. coli over-expressing PagN and PagN-coated beads to bind to and invade different non-phagocytic cells. According to the literature, PagN is likely to interact with heparan sulfate proteoglycan (HSPG) as PagN-mediated invasion could be inhibited by heparin treatment in a dose-dependent manner. This report shows that this interaction is not sufficient to allow the internalization mechanism. Investigation of the role of ß1 integrin as co-receptor showed that mouse embryo fibroblasts genetically deficient in ß1 integrin were less permissive to PagN-mediated internalization. Moreover, PagN-mediated internalization was fully inhibited in glycosylation-deficient pgsA-745 cells treated with anti-ß1 integrin antibody, supporting the hypothesis that ß1 integrin and HSPG cooperate to induce the PagN-mediated internalization mechanism. In addition, use of specific inhibitors and expression of dominant-negative derivatives demonstrated that tyrosine phosphorylation and class I phosphatidylinositol 3-kinase were crucial to trigger PagN-dependent internalization, as for the Rck internalization mechanism. Finally, scanning electron microscopy with infected cells showed microvillus-like extensions characteristic of Zipper-like structure, engulfing PagN-coated beads and E. coli expressing PagN, as observed during Rck-mediated internalization. CONCLUSIONS: Our results supply new comprehensions into T3SS-1-independent invasion mechanisms of S. Typhimurium and highly indicate that PagN induces a phosphatidylinositol 3-kinase signaling pathway, leading to a Zipper-like entry mechanism as the Salmonella outer membrane protein Rck.
Asunto(s)
Proteínas de la Membrana Bacteriana Externa/metabolismo , Infecciones por Salmonella/microbiología , Salmonella typhimurium/metabolismo , Animales , Proteínas de la Membrana Bacteriana Externa/genética , Línea Celular , Fibroblastos/metabolismo , Fibroblastos/microbiología , Humanos , Integrina beta1/genética , Integrina beta1/metabolismo , Ratones , Infecciones por Salmonella/genética , Infecciones por Salmonella/metabolismo , Salmonella typhimurium/genética , Sistemas de Secreción Tipo III/genética , Sistemas de Secreción Tipo III/metabolismoRESUMEN
BACKGROUND: Biofilm formation is an important causative factor in the expansion of the carious lesions in the enamel. Hence, new approaches to efficient antibacterial agents are highly demanded. This study was conducted to evaluate the antimicrobial-biofilm activity of chitosan hydrogel (CS gel), zinc oxide/ zeolite nanocomposite (ZnONC) either separately or combined together [ZnONC / CS gel (ZnONC-CS)] against Streptococcus mutans biofilm. RESULTS: MTT assay demonstrated that the ZnONC-CS exhibits a non-cytotoxic effect (> 90% cell viability) toward human gingival fibroblast cells at different dosages (78.1-625 µg/mL) within 72 h. In comparison with CS gel and ZnONC, ZnONC-CS was superior at biofilm formation and metabolic activity reduction by 33 and 45%, respectively; (P < 0.05). The field emission scanning electron microscopy micrographs of the biofilms grown on the enamel slabs were largely in concordance with the quantitative biofilm assay results. Consistent with the reducing effect of ZnONC-CS on biofilm formation, the expression levels of gtfB, gtfC, and ftf significantly decreased. CONCLUSIONS: Taken together, excellent compatibility coupled with an enhanced antimicrobial effect against S. mutans biofilm has equipped ZnONC-CS as a promising candidate for dental biofilm control.
Asunto(s)
Antibacterianos/farmacología , Biopelículas/efectos de los fármacos , Quitosano/farmacología , Nanogeles/química , Streptococcus mutans/efectos de los fármacos , Óxido de Zinc/farmacología , Quitosano/química , Caries Dental/tratamiento farmacológico , Caries Dental/microbiología , Fibroblastos/efectos de los fármacos , Fibroblastos/microbiología , Humanos , Pruebas de Sensibilidad Microbiana , Microscopía Electrónica de Rastreo , Streptococcus mutans/patogenicidad , Virulencia , Factores de Virulencia , Óxido de Zinc/químicaRESUMEN
The host-microbe relationship is pivotal for oral health as well as for peri-implant diseases. Peri-implant mucosa and commensal biofilm play important roles in the maintenance of host-microbe homeostasis, but little is known about how they interact. We have therefore investigated the early host-microbe interaction between commensal multispecies biofilm (Streptococcus oralis, Actinomyces naeslundii, Veillonella dispar, Porphyromonas gingivalis) and organotypic peri-implant mucosa using our three-dimensional model. After 24 hr, biofilms induced weak inflammatory reaction in the peri-implant mucosa by upregulation of five genes related to immune response and increased secretion of IL-6 and CCL20. Biofilm volume was reduced which might be explained by secretion of ß-Defensins-1, -2, and CCL20. The specific tissue reaction without intrinsic overreaction might contribute to intact mucosa. Thus, a relationship similar to homeostasis and oral health was established within the first 24 hr. In contrast, the mucosa was damaged and the bacterial distribution was altered after 48 hr. These were accompanied by an enhanced immune response with upregulation of additional inflammatory-related genes and increased cytokine secretion. Thus, the homeostasis-like relationship was disrupted. Such profound knowledge of the host-microbe interaction at the peri-implant site may provide the basis to improve strategies for prevention and therapy of peri-implant diseases.
Asunto(s)
Biopelículas , Fibroblastos/microbiología , Interacciones Microbiota-Huesped , Modelos Anatómicos , Mucosa Bucal/microbiología , Actinomyces/fisiología , Citocinas/inmunología , Fibroblastos/inmunología , Interacciones Microbiota-Huesped/genética , Interacciones Microbiota-Huesped/inmunología , Humanos , Mucosa Bucal/inmunología , Porphyromonas gingivalis/inmunología , Porphyromonas gingivalis/fisiología , Veillonella/inmunología , Veillonella/fisiologíaRESUMEN
The invasion of skin tissue by Staphylococcus aureus is mediated by mechanisms that involve sequential breaching of the different stratified layers of the epidermis. Induction of cell death in keratinocytes is a measure of virulence and plays a crucial role in the infection progression. We established a 3D-organotypic keratinocyte-fibroblast co-culture model to evaluate whether a 3D-skin model is more effective in elucidating the differences in the induction of cell death by Methicillin-resistant Staphylococcus aureus (MRSA) than in comparison to 2D-HaCaT monolayers. We investigated the difference in adhesion, internalization, and the apoptotic index in HaCaT monolayers and our 3D-skin model using six strains of MRSA representing different clonal types, namely, ST8, ST30, ST59, ST22, ST45 and ST239. All the six strains exhibited internalization in HaCaT cells. Due to cell detachment, the invasion study was limited up to two and a half hours. TUNEL assay showed no significant difference in the cell death induced by the six MRSA strains in the HaCaT cells. Our 3D-skin model provided a better insight into the interactions between the MRSA strains and the human skin during the infection establishment as we could study the infection of MRSA in our skin model up to 48 h. Immunohistochemical staining together with TUNEL assay in the 3D-skin model showed co-localization of the bacteria with the apoptotic cells demonstrating the induction of apoptosis by the bacteria and revealed the variation in bacterial transmigration among the MRSA strains. The strain representing ST59 showed maximum internalization in HaCaT cells and the maximum cell death as measured by Apoptotic index in the 3D-skin model. Our results show that 3D-skin model might be more likely to imitate the physiological response of skin to MRSA infection than 2D-HaCaT monolayer keratinocyte cultures and will enhance our understanding of the difference in pathogenesis among different MRSA strains.
Asunto(s)
Técnicas de Cultivo de Célula , Fibroblastos/microbiología , Queratinocitos/microbiología , Staphylococcus aureus Resistente a Meticilina/fisiología , Modelos Biológicos , Piel/microbiología , Infecciones Estafilocócicas/microbiología , Infecciones Estafilocócicas/patología , Apoptosis , Adhesión Celular , Muerte Celular , Endocitosis , Células HaCaT , HumanosRESUMEN
Staphylococcus aureus is one of the most prevalent pathogens associated with several types of biofilm-based infections, including infections of chronic wounds. Mature staphylococcal biofilm is extremely hard to eradicate from a wound and displays a high tendency to induce recurring infections. Therefore, in the present study, we aimed to investigate in vitro the interaction between S. aureus biofilm and fibroblast cells searching for metabolites that could be considered as potential biomarkers of critical colonization and infection. Utilizing advanced microscopy and microbiological methods to examine biofilm formation and the staphylococcal infection process, we were able to distinguish 4 phases of biofilm development. The analysis of staphylococcal biofilm influence on the viability of fibroblasts allowed us to pinpoint the moment of critical colonization-12 h post contamination. Based on the obtained model we performed a metabolomics analysis by 1H NMR spectroscopy to provide new insights into the pathophysiology of infection. We identified a set of metabolites related to the switch to anaerobic metabolism that was characteristic for staphylococcal biofilm co-cultured with fibroblast cells. The data presented in this study may be thus considered a noteworthy but preliminary step in the direction of developing a new, NMR-based tool for rapid diagnosing of infection in a chronic wound.
Asunto(s)
Biopelículas/crecimiento & desarrollo , Técnicas de Cocultivo , Fibroblastos/metabolismo , Fibroblastos/microbiología , Staphylococcus aureus/crecimiento & desarrollo , Staphylococcus aureus/metabolismo , Supervivencia Celular , Fibroblastos/ultraestructura , Técnica del Anticuerpo Fluorescente , Interacciones Huésped-Patógeno , Cinética , Espectroscopía de Resonancia Magnética , Metaboloma , Metabolómica/métodos , Staphylococcus aureus/ultraestructuraRESUMEN
Mesenchymal stem cells (MSCs) can improve chronic wound healing; however, recent studies suggest that the therapeutic effect of MSCs is mediated mainly through the growth factors and cytokines secreted by these cells, referred to as the MSC secretome. To overcome difficulties related to the translation of cell therapy into clinical use such as efficacy, safety and cost, we propose a hydrogel loaded with a secretome from the recently established human adipose tissue mesenchymal stem cell line (HATMSC2) as a potential treatment for chronic wounds. Biocompatibility and biological activity of hydrogel-released HATMSC2 supernatant were investigated in vitro by assessing the proliferation and metabolic activity of human fibroblast, endothelial cells and keratinocytes. Hydrogel degradation was measured using hydroxyproline assay while protein released from the hydrogel was assessed by interleukin-8 (IL-8) and macrophage chemoattractant protein-1 (MCP-1) ELISAs. Pro-angiogenic activity of the developed treatment was assessed by tube formation assay while the presence of pro-angiogenic miRNAs in the HATMSC2 supernatant was investigated using real-time RT-PCR. The results demonstrated that the therapeutic effect of the HATMSC2-produced factors is maintained following incorporation into collagen hydrogel as confirmed by increased proliferation of skin-origin cells and improved angiogenic properties of endothelial cells. In addition, HATMSC2 supernatant revealed antimicrobial activity, and which therefore, in combination with the hydrogel has a potential to be used as advanced wound-healing dressing.
Asunto(s)
Tejido Adiposo/citología , Medios de Cultivo Condicionados/farmacología , Hidrogeles/farmacología , Células Madre Mesenquimatosas/metabolismo , Secretoma/metabolismo , Piel/metabolismo , Antiinfecciosos/química , Antiinfecciosos/farmacología , Bacterias/clasificación , Bacterias/efectos de los fármacos , Bacterias/crecimiento & desarrollo , Línea Celular , Proliferación Celular/efectos de los fármacos , Proliferación Celular/genética , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/genética , Medios de Cultivo Condicionados/química , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Células Endoteliales/microbiología , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Fibroblastos/microbiología , Humanos , Hidrogeles/química , Queratinocitos/efectos de los fármacos , Queratinocitos/metabolismo , Queratinocitos/microbiología , Células Madre Mesenquimatosas/citología , MicroARNs/genética , Piel/citología , Piel/microbiologíaRESUMEN
Staphylococcus aureus is still one of the leading causes of both hospital- and community-acquired infections. Due to the very high percentage of drug-resistant strains, the participation of drug-tolerant biofilms in pathological changes, and thus the limited number of effective antibiotics, there is an urgent need to search for alternative methods of prevention or treatment for S. aureus infections. In the present study, biochemically characterized (HPLC/UPLC-QTOF-MS) acetonic, ethanolic, and water extracts from fruits and bark of Viburnum opulus L. were tested in vitro as diet additives that potentially prevent staphylococcal infections. The impacts of V. opulus extracts on sortase A (SrtA) activity (Fluorimetric Assay), staphylococcal protein A (SpA) expression (FITC-labelled specific antibodies), the lipid composition of bacterial cell membranes (LC-MS/MS, GC/MS), and biofilm formation (LIVE/DEAD BacLight) were assessed. The cytotoxicity of V. opulus extracts to the human fibroblast line HFF-1 was also tested (MTT reduction). V. opulus extracts strongly inhibited SrtA activity and SpA expression, caused modifications of S. aureus cell membrane, limited biofilm formation by staphylococci, and were non-cytotoxic. Therefore, they have pro-health potential. Nevertheless, their usefulness as diet supplements that are beneficial for the prevention of staphylococcal infections should be confirmed in animal models in the future.
Asunto(s)
Antibacterianos/farmacología , Biopelículas/efectos de los fármacos , Fibroblastos , Staphylococcus aureus Resistente a Meticilina/fisiología , Extractos Vegetales/farmacología , Viburnum/química , Aminoaciltransferasas/biosíntesis , Antibacterianos/química , Proteínas Bacterianas/biosíntesis , Línea Celular , Cisteína Endopeptidasas/biosíntesis , Fibroblastos/metabolismo , Fibroblastos/microbiología , Fibroblastos/patología , Frutas/química , Regulación Bacteriana de la Expresión Génica/efectos de los fármacos , Humanos , Corteza de la Planta/química , Extractos Vegetales/químicaRESUMEN
The human-adapted pathogen group A Streptococcus (GAS) utilizes wounds as portals of entry into host tissue, wherein surface adhesins interact with the extracellular matrix, enabling bacterial colonization. The streptococcal collagen-like protein 1 (Scl1) is a major adhesin of GAS that selectively binds to two fibronectin type III (FnIII) repeats within cellular fibronectin, specifically the alternatively spliced extra domains A and B, and the FnIII repeats within tenascin-C. Binding to FnIII repeats was mediated through conserved structural determinants present within the Scl1 globular domain and facilitated GAS adherence and biofilm formation. Isoforms of cellular fibronectin that contain extra domains A and B, as well as tenascin-C, are present for several days in the wound extracellular matrix. Scl1-FnIII binding is therefore an example of GAS adaptation to the host's wound environment. Similarly, cellular fibronectin isoforms and tenascin-C are present in the tumor microenvironment. Consistent with this, FnIII repeats mediate GAS attachment to and enhancement of biofilm formation on matrices deposited by cancer-associated fibroblasts and osteosarcoma cells. These data collectively support the premise for utilization of the Scl1-FnIII interaction as a novel method of anti-neoplastic targeting in the tumor microenvironment.
Asunto(s)
Proteínas Bacterianas/metabolismo , Colágeno/metabolismo , Fibronectinas/metabolismo , Neoplasias/terapia , Streptococcus pyogenes/fisiología , Adhesinas Bacterianas/química , Adhesinas Bacterianas/genética , Adhesinas Bacterianas/metabolismo , Adhesión Bacteriana , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Biopelículas , Línea Celular Tumoral , Colágeno/química , Colágeno/genética , Matriz Extracelular/metabolismo , Fibroblastos/metabolismo , Fibroblastos/microbiología , Fibronectinas/química , Fibronectinas/genética , Humanos , Neoplasias/metabolismo , Unión Proteica , Dominios Proteicos , Isoformas de Proteínas/química , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Streptococcus pyogenes/química , Streptococcus pyogenes/genéticaRESUMEN
Cultivation of Chlamydia species in cell lines requires centrifugation of the inoculum onto diethylaminoethyl-dextran-pretreated cell monolayers to improve the infection efficiency. Here we report that the addition of DNA transfection reagent Lipofectamine in the inoculum significantly enhances the infectivity of Chlamydia abortus in mouse fibroblast McCoy cells, with an infection efficiency equivalent to that of the centrifugation method. Similar enhancement effects of Lipofectamine on the infectivity of C. psittaci and C. trachomatis were also observed. This study provides an alternative and convenient method for the cultivation of Chlamydia species in vitro in the absence of centrifugation.
Asunto(s)
Chlamydia/fisiología , Lípidos/química , Animales , Línea Celular , Centrifugación , Chlamydia trachomatis/fisiología , Fibroblastos/citología , Fibroblastos/metabolismo , Fibroblastos/microbiología , RatonesRESUMEN
The cytokine IFN-γ has well-established antibacterial properties against the bacterium Salmonella enterica in phagocytes, but less is known about the effects of IFN-γ on Salmonella-infected nonphagocytic cells, such as intestinal epithelial cells (IECs) and fibroblasts. In this article, we show that exposing human and murine IECs and fibroblasts to IFN-γ following infection with Salmonella triggers a novel form of cell death that is neither pyroptosis nor any of the major known forms of programmed cell death. Cell death required IFN-γ-signaling via STAT1-IRF1-mediated induction of guanylate binding proteins and the presence of live Salmonella in the cytosol. In vivo, ablating IFN-γ signaling selectively in murine IECs led to higher bacterial burden in colon contents and increased inflammation in the intestine of infected mice. Together, these results demonstrate that IFN-γ signaling triggers release of Salmonella from the Salmonella-containing vacuole into the cytosol of infected nonphagocytic cells, resulting in a form of nonpyroptotic cell death that prevents bacterial spread in the gut.
Asunto(s)
Muerte Celular/inmunología , Interferón gamma/inmunología , Fagocitos/inmunología , Piroptosis/inmunología , Infecciones por Salmonella/inmunología , Salmonella enterica/inmunología , Células 3T3 , Animales , Línea Celular , Citosol/inmunología , Citosol/microbiología , Células Epiteliales/inmunología , Células Epiteliales/microbiología , Fibroblastos/inmunología , Fibroblastos/microbiología , Humanos , Inflamación/inmunología , Inflamación/microbiología , Factor 1 Regulador del Interferón/inmunología , Intestinos/inmunología , Intestinos/microbiología , Ratones , Fagocitos/microbiología , Factor de Transcripción STAT1/inmunología , Infecciones por Salmonella/microbiologíaRESUMEN
AIMS: Probiotics have the ability to enhance the immune system, produce anti-inflammatory action and promote wound healing process. The first aim of the study was to isolate pathogenic micro-organisms from sites of chronic ulcerative lesion. The second aim was to evaluate probiotic efficacy of SYNBIO® (1:1 combination of Lactobacillus rhamnosus IMC 501® and Lactobacillus paracasei IMC 502® ) in counteracting wound infections. METHODS AND RESULTS: Several bacterial pathogens were isolated from chronic ulcerative lesions and identified by morphological, biochemical and molecular techniques. SYNBIO® probiotic formulation was investigated for its antimicrobial activity, minimum inhibitory concentration, co-aggregation and adherence capacity against the isolated pathogens. Moreover, SYNBIO was also tested in combination with some medical devices, using an in vitro model, in order to simulate a real ulcerative wound infection. Probiotic formulation demonstrated an inhibitory action against all the tested pathogens and their mixture (MIX), with an increased ability of co-aggregation during time. In addition, the adhesion percentage of probiotic micro-organisms to human keratinocyte (HaCaT cells) and human fibroblasts (NHF), calculated by an in vitro model, was 19% and 17% respectively, highlighting the possibility to create a protective environment preventing pathogens' biofilm formation in order to contrast infections. CONCLUSIONS: SYNBIO® probiotics showed a very good antimicrobial capacity and adhesion percentage to HaCaT cells and fibroblasts, giving the opportunity to be successfully used as complement to conventional therapies in the treatment of chronic ulcerative lesions. SIGNIFICANCE AND IMPACT OF THE STUDY: A new therapeutic approach with probiotics (supplemented in topical applications, excluding side effects) able to eliminate pathogenic micro-organisms and improve healing of chronic ulcerative lesions.
Asunto(s)
Bacterias/aislamiento & purificación , Colitis Ulcerosa/tratamiento farmacológico , Probióticos/administración & dosificación , Bacterias/clasificación , Bacterias/genética , Adhesión Bacteriana , Fenómenos Fisiológicos Bacterianos , Enfermedad Crónica/terapia , Colitis Ulcerosa/microbiología , Fibroblastos/microbiología , Humanos , Queratinocitos/microbiología , Lacticaseibacillus paracasei/fisiología , Lacticaseibacillus rhamnosus/fisiologíaRESUMEN
Efficient cell-to-cell transfer of Listeria monocytogenes (L. monocytogenes) requires the proper formation of actin-rich membrane protrusions. To date, only the host proteins ezrin, the binding partner of ezrin, CD44, as well as cyclophilin A (CypA) have been identified as crucial components for L. monocytogenes membrane protrusion stabilization and, thus, efficient cell-to-cell movement of the microbes. Here, we examine the classical binding partner of CypA, CD147, and find that this membrane protein is also hijacked by the bacteria for their cellular dissemination. CD147 is enriched at the plasma membrane surrounding the membrane protrusions as well as the resulting invaginations generated in neighboring cells. In cells depleted of CD147, these actin-rich structures appear similar to those generated in CypA depleted cells as they are significantly shorter and more contorted as compared to their straighter counterparts formed in wild-type control cells. The presence of malformed membrane protrusions hampers the ability of L. monocytogenes to efficiently disseminate from CD147-depleted cells. Our findings uncover another important host protein needed for L. monocytogenes membrane protrusion formation and efficient microbial dissemination.