1
|
Arsaute S, Reinoso EB, Cecchini ME, Moliva MV, Montironi ID, Cariddi LN. Minthostachys verticillata essential oil modulates cytokine synthesis and Staphylococcus aureus internalization in MAC-T cells at least through TLR4/MyD88/NFkB pathway. Vet Res Commun 2024; 48:3727-3742. [PMID: 39249174 DOI: 10.1007/s11259-024-10526-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Accepted: 08/30/2024] [Indexed: 09/10/2024]
Abstract
The aim of this study was to evaluate the activation pathway(s) triggered by Minthostachys verticillata essential oil (EO) in bovine mammary epithelial cells (MAC-T) challenged with a strain of bovine Staphylococcus aureus. MAC-T cells were stimulated with EO, S. aureus or pre-treated with EO and then challenged with S. aureus. Cytokine's release was measured by ELISA. The mRNA for TLR2, TLR4, NOD2, MyD88 and NFκB was quantified by RT-qPCR. S. aureus adherence and internalization was also evaluated. MAC-T cells stimulated with S. aureus synthesized high levels of IL-1ß and IL-6 were kept up to 48 h, while IL-4 levels were not altered. Cells pre-treated with EO for 2 and 6 h and then challenged with S. aureus showed a significant increase of IL-1β and IL-6. However, in these cells, a decrease in IL-1ß and IL-6 levels and an increase of IL-4 values was observed from 24 h. No significant increase in the expression levels of TLR2 or NOD2 was detected in all stimulated cells. However, the expression of TLR4, MyD88 and NFκB was increased in cells stimulated with S. aureus at 2 and 6 h as well as in cells pre-treated with EO between 2 and 6 h and then challenged with S. aureus. The NFκB expression levels was similar to control at 24 h in all stimulated cells, although pro-inflammatory cytokine levels and TLR4 and MyD88 expression levels remained high in cells stimulated with S. aureus. This results suggested the activation of other pathways independent of MyD88 by the pathogen that involucrated the activation of others transcription factors. Pre-treatment with EO during 2, 6 and 24 h did not affect S. aureus adherence but decreased its internalization. In conclusion, pre-treatment with EO increased the IL-1β and IL-6 synthesis during the first hours post-challenged with S. aureus up-regulating TLR4/MyD88/NFκB pathway. Furthermore, EO increased the IL-4 levels from 6 to 24 h down-regulating the NFκB and possibly other transcription factors activated by the pathogen, which decreased its internalization into MAC-T cells.
Collapse
Affiliation(s)
- Sofía Arsaute
- Facultad de Ciencias Exactas Físico-Químicas y Naturales, Departamento de Microbiología e Inmunología, Universidad Nacional de Río Cuarto, Río Cuarto, Córdoba, 5800, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Instituto de Biotecnología Ambiental y Salud (INBIAS), Ruta 36 Km 601, Río Cuarto, Córdoba, 5800, Argentina
| | - Elina Beatriz Reinoso
- Facultad de Ciencias Exactas Físico-Químicas y Naturales, Departamento de Microbiología e Inmunología, Universidad Nacional de Río Cuarto, Río Cuarto, Córdoba, 5800, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Instituto de Biotecnología Ambiental y Salud (INBIAS), Ruta 36 Km 601, Río Cuarto, Córdoba, 5800, Argentina
| | - María Eugenia Cecchini
- Facultad de Ciencias Exactas Físico-Químicas y Naturales, Departamento de Microbiología e Inmunología, Universidad Nacional de Río Cuarto, Río Cuarto, Córdoba, 5800, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Instituto de Biotecnología Ambiental y Salud (INBIAS), Ruta 36 Km 601, Río Cuarto, Córdoba, 5800, Argentina
| | - Melina Vanesa Moliva
- Facultad de Ciencias Exactas Físico-Químicas y Naturales, Departamento de Microbiología e Inmunología, Universidad Nacional de Río Cuarto, Río Cuarto, Córdoba, 5800, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Instituto de Biotecnología Ambiental y Salud (INBIAS), Ruta 36 Km 601, Río Cuarto, Córdoba, 5800, Argentina
| | - Ivana Dalila Montironi
- Facultad de Ciencias Exactas Físico-Químicas y Naturales, Departamento de Microbiología e Inmunología, Universidad Nacional de Río Cuarto, Río Cuarto, Córdoba, 5800, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Instituto de Biotecnología Ambiental y Salud (INBIAS), Ruta 36 Km 601, Río Cuarto, Córdoba, 5800, Argentina
| | - Laura Noelia Cariddi
- Facultad de Ciencias Exactas Físico-Químicas y Naturales, Departamento de Microbiología e Inmunología, Universidad Nacional de Río Cuarto, Río Cuarto, Córdoba, 5800, Argentina.
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Instituto de Biotecnología Ambiental y Salud (INBIAS), Ruta 36 Km 601, Río Cuarto, Córdoba, 5800, Argentina.
| |
Collapse
|
2
|
Wu X, Chen P, Huang D, Pan Y, Chen S. Bone and periosteum protein analysis via tandem mass tag quantitative proteomics in pediatric patients with osteomyelitis. Biomed Chromatogr 2024; 38:e5999. [PMID: 39380190 DOI: 10.1002/bmc.5999] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2024] [Revised: 08/15/2024] [Accepted: 08/16/2024] [Indexed: 10/10/2024]
Abstract
Bone healing is crucial in managing osteomyelitis after fracture fixation. Understanding the mechanism of extensive callus formation in pediatric osteomyelitis is highly important. This study aims to analyze bone and periosteum samples from pediatric patients to elucidate the essential processes involved in callus formation during osteomyelitis. The study included eight patients from our hospital: four with positive microbial culture who underwent osteomyelitis debridement and four who had osteotomy surgery as contral. We used tandem mass tag quantitative proteomics to investigate proteomic changes in bone and periosteum tissues obtained from these patients. Differential expression proteins were analyzed for their pathways through Gene Ontology (GO) annotation, GO enrichment analysis, Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis, and protein-protein interaction networks. A total of 4737 proteins were successfully identified. About 2224 differentially expressed proteins were detected in the bone tissues group and periosteum tissues group. Among the differentially expressed proteins, 10 protein genes in the bone group were associated with inflammation and osteogenesis, while in the periosteum group were nine. Cytochrome b-245, beta polypeptide (CYBB), nicotinamide phosphoribosyltransferase (NAMPT), tissue inhibitor of metalloproteinases 1 (TIMP-1), Raf-1 proto-oncogene, serine/threonine kinase (RAF-1), RELA proto-oncogene, NF-KB subunit (RELA), and sphingomyelin synthase 2 (SGMS2) may play an important role in callus formation in patients with osteomyelitis. This study provides novel clues for understanding callus formation in pediatric patients with osteomyelitis.
Collapse
Affiliation(s)
- Xinwu Wu
- Department of Orthopedics, Fuzhou Second General Hospital, Fuzhou, China
- Fujian Provincial Clinical Medical Research Center for First Aid and Rehabilitation in Orthopedics Trauma, Fuzhou, China
- Fuzhou Trauma Medical Center, Fuzhou, China
| | - Peisheng Chen
- Department of Orthopedics, Fuzhou Second General Hospital, Fuzhou, China
- Fujian Provincial Clinical Medical Research Center for First Aid and Rehabilitation in Orthopedics Trauma, Fuzhou, China
- Fuzhou Trauma Medical Center, Fuzhou, China
| | - Dianhua Huang
- Department of Orthopedics, Fuzhou Second General Hospital, Fuzhou, China
- Fujian Provincial Clinical Medical Research Center for First Aid and Rehabilitation in Orthopedics Trauma, Fuzhou, China
- Fuzhou Trauma Medical Center, Fuzhou, China
| | - Yuchen Pan
- Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Shunyou Chen
- Department of Orthopedics, Fuzhou Second General Hospital, Fuzhou, China
- Fujian Provincial Clinical Medical Research Center for First Aid and Rehabilitation in Orthopedics Trauma, Fuzhou, China
- Fuzhou Trauma Medical Center, Fuzhou, China
| |
Collapse
|
3
|
Das SK, Joshi A, Bisht L, Goswami V, Faiz A, Dutt G, Sharma S. Godanti bhasma (anhydrous CaSO 4) induces massive cytoplasmic vacuolation in mammalian cells: A model for phagocytosis assay. Methods 2024; 230:158-168. [PMID: 39216714 DOI: 10.1016/j.ymeth.2024.08.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 08/18/2024] [Accepted: 08/23/2024] [Indexed: 09/04/2024] Open
Abstract
Phagocytosis is an essential physiological mechanism; its impairment is associated with many diseases. A highly smart particle is required for understanding detailed sequential cellular events in phagocytosis. Recently, we identified an Indian traditional medicine named Godanti Bhasma (GB), a bioactive calcium sulfate particle prepared by thermo-transformation ofgypsum. Thermal processing of the gypsum transforms its native physicochemical properties by removing water molecules into the anhydrous GB, which was confirmed by Raman and FT-IR spectroscopy. GB particle showed a 0.5-5 µm size range and a neutral surface charge. Exposure of mammalian cells to GB particles showed a rapid cellular uptake through phagocytosis and induced massive cytoplasmic vacuolation in cells. Interestingly, no cellular uptake and cytoplasmic vacuolation were observed with the parent gypsum particle. The presence of the GB particles in intra-vacuolar space was confirmed using FESEM coupled with EDX. Flow cytometry analysis and live tracking of GB-treated cells showed particle internalization, vacuole formation, particle dissolution, and later vacuolar turnover. Quantification of GB-induced vacuolation was done using neutral red uptake assay in cells. Treatment of lysosomal inhibitors (BFA1 or CQ) with GB could not induce vacuolation, suggesting the requirement of an acidic environment for the vacuolation. In the mimicking experiment, GB particle dissolution in acidic cell-free solution suggested that degradation of GB occurs by acidic pH inside the cell vacuole. Vacuole formation generally accompanies with cell death, whereas GB-induced massive vacuolation does not cause cell death. Moreover, the cell divides and proliferates with the vacuolar process, intra-vacuolar cargo degradation, and eventually vacuolar turnover. Taken together, the sequential cellular events in this study suggest that GB can be used as a smart particle for phagocytosis assay development in animal cells.
Collapse
Affiliation(s)
- Subrata K Das
- Shobhit Institute of Engineering & Technology, University, NH 58, Meerut 250110, India; Drug Discovery and Development Division, Patanjali Research Institute, Haridwar 249405, Uttarakhand, India.
| | - Alpana Joshi
- Shobhit Institute of Engineering & Technology, University, NH 58, Meerut 250110, India; Drug Discovery and Development Division, Patanjali Research Institute, Haridwar 249405, Uttarakhand, India
| | - Laxmi Bisht
- Drug Discovery and Development Division, Patanjali Research Institute, Haridwar 249405, Uttarakhand, India
| | - Vishakha Goswami
- Shobhit Institute of Engineering & Technology, University, NH 58, Meerut 250110, India
| | - Abul Faiz
- Shobhit Institute of Engineering & Technology, University, NH 58, Meerut 250110, India
| | - Gaurav Dutt
- Shobhit Institute of Engineering & Technology, University, NH 58, Meerut 250110, India
| | - Shiva Sharma
- Shobhit Institute of Engineering & Technology, University, NH 58, Meerut 250110, India
| |
Collapse
|
4
|
Moulick S, Roy DN. Bioflavonoid Baicalein Modulates Tetracycline Resistance by Inhibiting Efflux Pump in Staphylococcus aureus. Microb Drug Resist 2024; 30:363-371. [PMID: 39133125 DOI: 10.1089/mdr.2024.0099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/13/2024] Open
Abstract
The rise in antibiotic resistance among bacterial pathogens, particularly Staphylococcus aureus, has become a critical global health issue, necessitating the search for novel antimicrobial agents. S. aureus uses various mechanisms to resist antibiotics, including the activation of efflux pumps, biofilm formation, and enzymatic modification of drugs. This study explores the potential of baicalein, a bioflavonoid from Scutellaria baicalensis, in modulating tetracycline resistance in S. aureus by inhibiting efflux pumps. The synergistic action of baicalein and tetracycline was evaluated through various assays. The minimum inhibitory concentration (MIC) of baicalein and tetracycline against S. aureus was 256 and 1.0 μg/mL, respectively. Baicalein at 64 μg/mL reduced the MIC of tetracycline by eightfold, indicating a synergistic effect (fractional inhibitory concentration index: 0.375). Time-kill kinetics demonstrated a 1.0 log CFU/mL reduction in bacterial count after 24 hours with the combination treatment. The ethidium bromide accumulation assay showed that baicalein mediated significant inhibition of efflux pumps, with a dose-dependent increase in fluorescence. In addition, baicalein inhibited DNA synthesis by 73% alone and 92% in combination with tetracycline. It also markedly reduced biofilm formation and the invasiveness of S. aureus into HeLa cells by 52% at 64 μg/mL. These findings suggest that baicalein enhances tetracycline efficacy and could be a promising adjunct therapy to combat multidrug-resistant S. aureus infections.
Collapse
Affiliation(s)
- Soumitra Moulick
- TCG Lifesciences Private Limited, Kolkata, India
- Department of Biotechnology, National Institute of Technology Raipur, Chhattisgarh, India
| | - Dijendra Nath Roy
- Department of Biotechnology, National Institute of Technology Raipur, Chhattisgarh, India
| |
Collapse
|
5
|
Wang J, Cao H, Yang H, Wang N, Weng Y, Luo H. The function of CD36 in Mycobacterium tuberculosis infection. Front Immunol 2024; 15:1413947. [PMID: 38881887 PMCID: PMC11176518 DOI: 10.3389/fimmu.2024.1413947] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Accepted: 05/20/2024] [Indexed: 06/18/2024] Open
Abstract
CD36 is a scavenger receptor that has been reported to function as a signaling receptor that responds to pathogen-associated molecular patterns (PAMPs) and damage-associated molecular patterns (DAMPs) and could integrate metabolic pathways and cell signaling through its dual functions. Thereby influencing activation to regulate the immune response and immune cell differentiation. Recent studies have revealed that CD36 plays critical roles in the process of lipid metabolism, inflammatory response and immune process caused by Mycobacterium tuberculosis infection. This review will comprehensively investigate CD36's functions in lipid uptake and processing, inflammatory response, immune response and therapeutic targets and biomarkers in the infection process of M. tuberculosis. The study also raised outstanding issues in this field to designate future directions.
Collapse
Affiliation(s)
- Jianjun Wang
- Department of Clinical Laboratory, Kunshan Hospital Affiliated to Jiangsu University, Suzhou, China
| | - Hui Cao
- Department of Food and Nutrition Safety, Jiangsu Provincial Center for Disease Control and Prevention, Nanjing, Jiangsu, China
| | - Hongwei Yang
- Department of Clinical Laboratory, Suzhou BOE Hospital, Suzhou, Jiangsu, China
| | - Nan Wang
- Department of Clinical Laboratory, Kunshan Hospital Affiliated to Jiangsu University, Suzhou, China
| | - Yiwei Weng
- Department of Clinical Laboratory, The Fourth People’s Hospital of Kunshan, Suzhou, Jiangsu, China
| | - Hao Luo
- Department of Clinical Laboratory, The Second People's Hospital of Kunshan, Suzhou, China
| |
Collapse
|
6
|
Zhao H, Shang L, Zhang Y, Liang Z, Wang N, Zhang Q, Gao C, Luo J. IL-17A inhibitors alleviate Psoriasis with concomitant restoration of intestinal/skin microbiota homeostasis and altered microbiota function. Front Immunol 2024; 15:1344963. [PMID: 38482003 PMCID: PMC10933079 DOI: 10.3389/fimmu.2024.1344963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 02/12/2024] [Indexed: 04/12/2024] Open
Abstract
Background Disturbed gut microbiota and associated metabolic dysfunction exist in Psoriasis. Despite the growing use of interleukin-17 inhibitor (anti-IL17) therapy, the effect of anti-IL17 on gut/skin microbiota function is not fully understood in patients with Psoriasis. Objective Therefore, we explored whether Psoriasis is associated with alterations in selected gut/skin microbiota in a study cohort, and a longitudinal cohort study to reveal the effects of IL-17A inhibitor treatment on gut microbiota in Psoriasis. Methods In a case-control study, 14 patients with Psoriasis and 10 age, sex and body mass index-matched Healthy Controls were recruited. Longitudinal mapping of the gut microbiome was performed using 16S rRNA gene sequencing. Mouse models were used to further study and validate the interrelationship between the skin microbiome and the gut microbiome in Psoriasis. PICRUST2 was applied to predict the function of the bacterial community. Results In Psoriasis patients, gut microbiota dysbiosis was present with increased heterogeneity: decreased Bacteroidota and increased Firmicutes as well as Actinobacteriota predominating in Psoriasis. Escherichia-Shigella enrichment was associated with reduction in serum levels of total bile acid and markers in Apoptotic pathways. After IL-17A inhibitor treatment in Psoriasis patients, longitudinal studies observed a trend toward a normal distribution of the gut microbiome and modulation of apoptosis-related metabolic pathways. Results from a mouse model showed dysregulation of the skin microbiota in Psoriasis characterized by Staphylococcus colonization. Conclusion The psoriatic gut/skin microbiota exhibits loss of community stability and pathogen enrichment. IL-17A inhibitors restore microbiota homeostasis and metabolic pathways, reduce pro-inflammatory cytokine expression, and alleviate symptoms in patients with Psoriasis.
Collapse
Affiliation(s)
- Huixia Zhao
- Department of Dermatology, Heji Hospital of Changzhi Medical College, Changzhi, China
| | - Lili Shang
- Department of Rheumatology, The Second Clinical Medical College of Shanxi Medical University, Taiyuan, China
- Shanxi Key Laboratory for immunomicroecology, The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Yuting Zhang
- Department of Dermatology, Heji Hospital of Changzhi Medical College, Changzhi, China
| | - Zhaojun Liang
- Shanxi Key Laboratory for immunomicroecology, The Second Hospital of Shanxi Medical University, Taiyuan, China
- Department of Rheumatology, The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Nan Wang
- Shanxi Key Laboratory for immunomicroecology, The Second Hospital of Shanxi Medical University, Taiyuan, China
- Department of Rheumatology, The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Qian Zhang
- Department of Dermatology, Heji Hospital of Changzhi Medical College, Changzhi, China
| | - Chong Gao
- Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Jing Luo
- Shanxi Key Laboratory for immunomicroecology, The Second Hospital of Shanxi Medical University, Taiyuan, China
- Department of Rheumatology, The Second Hospital of Shanxi Medical University, Taiyuan, China
| |
Collapse
|
7
|
Barajas-Mendiola MA, Salgado-Lora MG, López-Meza JE, Ochoa-Zarzosa A. Prolactin regulates H3K9ac and H3K9me2 epigenetic marks and miRNAs expression in bovine mammary epithelial cells challenged with Staphylococcus aureus. Front Microbiol 2022; 13:990478. [PMID: 36212825 PMCID: PMC9539446 DOI: 10.3389/fmicb.2022.990478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2022] [Accepted: 08/30/2022] [Indexed: 11/13/2022] Open
Abstract
Epigenetic mechanisms are essential in the regulation of immune response during infections. Changes in the levels of reproductive hormones, such as prolactin, compromise the mammary gland’s innate immune response (IIR); however, its effect on epigenetic marks is poorly known. This work explored the epigenetic regulation induced by bovine prolactin (bPRL) on bovine mammary epithelial cells (bMECs) challenged with Staphylococcus aureus. In this work, bMECs were treated as follows: (1) control cells without any treatment, (2) bMECs treated with bPRL (5 ng/ml) at different times (12 or 24 h), (3) bMECs challenged with S. aureus for 2 h, and (4) bMECs treated with bPRL at different times (12 or 24 h), and then challenged with S. aureus 2 h. By western blot analyses of histones, we determined that the H3K9ac mark decreased (20%) in bMECs treated with bPRL (12 h) and challenged with S. aureus, while the H3K9me2 mark was increased (50%) in the same conditions. Also, this result coincided with an increase (2.3-fold) in HDAC activity analyzed using the cellular histone deacetylase fluorescent kit FLUOR DE LYS®. ChIP-qPCRs were performed to determine if the epigenetic marks detected in the histones correlate with enriched marks in the promoter regions of inflammatory genes associated with the S. aureus challenge. The H3K9ac mark was enriched in the promoter region of IL-1β, IL-10, and BNBD10 genes (1.5, 2.5, 7.5-fold, respectively) in bMECs treated with bPRL, but in bMECs challenged with S. aureus it was reduced. Besides, the H3K9me2 mark was enriched in the promoter region of IL-1β and IL-10 genes (3.5 and 2.5-fold, respectively) in bMECs challenged with S. aureus but was inhibited by bPRL. Additionally, the expression of several miRNAs was analyzed by qPCR. Let-7a-5p, miR-21a, miR-30b, miR-155, and miR-7863 miRNAs were up-regulated (2, 1.5, 10, 1.5, 3.9-fold, respectively) in bMECs challenged with S. aureus; however, bPRL induced a down-regulation in the expression of these miRNAs. In conclusion, bPRL induces epigenetic regulation on specific IIR elements, allowing S. aureus to persist and evade the host immune response.
Collapse
|
8
|
Akhtar M, Naqvi SUAS, Liu Q, Pan H, Ma Z, Kong N, Chen Y, Shi D, Kulyar MFEA, Khan JA, Liu H. Short Chain Fatty Acids (SCFAs) Are the Potential Immunomodulatory Metabolites in Controlling Staphylococcus aureus-Mediated Mastitis. Nutrients 2022; 14:nu14183687. [PMID: 36145063 PMCID: PMC9503071 DOI: 10.3390/nu14183687] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 08/31/2022] [Accepted: 09/03/2022] [Indexed: 12/02/2022] Open
Abstract
Mastitis is an emerging health concern in animals. An increased incidence of mastitis in dairy cows has been reported in the last few years across the world. It is estimated that up to 20% of cows are suffering from mastitis, causing incompetency in the mucosal immunity and resulting in excessive global economic losses in the dairy industry. Staphylococcus aureus (S. aureus) has been reported as the most common bacterial pathogen of mastitis at clinical and sub-clinical levels. Antibiotics, including penicillin, macrolides, lincomycin, cephalosporins, tetracyclines, chloramphenicol, and methicillin, were used to cure S. aureus-induced mastitis. However, S. aureus is resistant to most antibiotics, and methicillin-resistant S. aureus (MRSA) especially has emerged as a critical health concern. MRSA impairs immune homeostasis leaving the host more susceptible to other infections. Thus, exploring an alternative to antibiotics has become an immediate requirement of the current decade. Short chain fatty acids (SCFAs) are the potent bioactive metabolites produced by host gut microbiota through fermentation and play a crucial role in host/pathogen interaction and could be applied as a potential therapeutic agent against mastitis. The purpose of this review is to summarize the potential mechanism by which SCFAs alleviate mastitis, providing the theoretical reference for the usage of SCFAs in preventing or curing mastitis.
Collapse
Affiliation(s)
- Muhammad Akhtar
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, Huazhong Agricultural University, Wuhan 430070, China
- Hubei Hongshan Laboratory, Wuhan 430070, China
- The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan 430070, China
| | | | - Qiyao Liu
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, Huazhong Agricultural University, Wuhan 430070, China
- Hubei Hongshan Laboratory, Wuhan 430070, China
- The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan 430070, China
| | - Hong Pan
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, Huazhong Agricultural University, Wuhan 430070, China
- Hubei Hongshan Laboratory, Wuhan 430070, China
- The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan 430070, China
| | - Ziyu Ma
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, Huazhong Agricultural University, Wuhan 430070, China
- Hubei Hongshan Laboratory, Wuhan 430070, China
- The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan 430070, China
| | - Na Kong
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, Huazhong Agricultural University, Wuhan 430070, China
- Hubei Hongshan Laboratory, Wuhan 430070, China
- The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan 430070, China
| | - Yan Chen
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, Huazhong Agricultural University, Wuhan 430070, China
- Hubei Hongshan Laboratory, Wuhan 430070, China
- The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan 430070, China
| | - Deshi Shi
- Department of Preventive Veterinary Medicine, College of Animal Science and Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| | - Muhammad Fakhar-e-Alam Kulyar
- Department of Clinical Veterinary Medicine, College of Animal Science and Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| | - Jawaria Ali Khan
- Department of Veterinary Medicine, Faculty of Veterinary Science, University of Veterinary and Animal Sciences, Lahore 54000, Pakistan
| | - Huazhen Liu
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, Huazhong Agricultural University, Wuhan 430070, China
- Hubei Hongshan Laboratory, Wuhan 430070, China
- The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan 430070, China
- Correspondence:
| |
Collapse
|
9
|
Li J, Wen Q, Gu F, An L, Yu T. Non-antibiotic strategies for prevention and treatment of internalized Staphylococcus aureus. Front Microbiol 2022; 13:974984. [PMID: 36118198 PMCID: PMC9471010 DOI: 10.3389/fmicb.2022.974984] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 08/08/2022] [Indexed: 12/01/2022] Open
Abstract
Staphylococcus aureus (S. aureus) infections are often difficult to cure completely. One of the main reasons for this difficulty is that S. aureus can be internalized into cells after infecting tissue. Because conventional antibiotics and immune cells have difficulty entering cells, the bacteria can survive long enough to cause recurrent infections, which poses a serious burden in healthcare settings because repeated infections drastically increase treatment costs. Therefore, preventing and treating S. aureus internalization is becoming a research hotspot. S. aureus internalization can essentially be divided into three phases: (1) S. aureus binds to the extracellular matrix (ECM), (2) fibronectin (Fn) receptors mediate S. aureus internalization into cells, and (3) intracellular S. aureus and persistence into cells. Different phases require different treatments. Many studies have reported on different treatments at different phases of bacterial infection. In the first and second phases, the latest research results show that the cell wall-anchored protein vaccine and some microbial agents can inhibit the adhesion of S. aureus to host cells. In the third phase, nanoparticles, photochemical internalization (PCI), cell-penetrating peptides (CPPs), antimicrobial peptides (AMPs), and bacteriophage therapy can effectively eliminate bacteria from cells. In this paper, the recent progress in the infection process and the prevention and treatment of S. aureus internalization is summarized by reviewing a large number of studies.
Collapse
Affiliation(s)
- Jiangbi Li
- Department of Orthopedics, The First Hospital of Jilin University, Changchun, China
| | - Qiangqiang Wen
- Department of Orthopedics, The Affiliated Hospital of Northwest University, Xi’an, China
| | - Feng Gu
- Department of Orthopedics, The First Hospital of Jilin University, Changchun, China
| | - Lijuan An
- Department of Rehabilitation Medicine, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Tiecheng Yu
- Department of Orthopedics, The First Hospital of Jilin University, Changchun, China
- *Correspondence: Tiecheng Yu,
| |
Collapse
|
10
|
Chen Y, Huang C, Chen X, Cai Y, Li W, Fang X, Zhang W. Bone protein analysis via label-free quantitative proteomics in patients with periprosthetic joint infection. J Proteomics 2022; 252:104448. [PMID: 34883267 DOI: 10.1016/j.jprot.2021.104448] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2021] [Revised: 11/18/2021] [Accepted: 11/23/2021] [Indexed: 11/17/2022]
Abstract
Periprosthetic joint infection (PJI) is a catastrophic complication of arthroplasty. The treatment of PJI often requires multiple operations and long-term use of antibiotics, making PJI a substantial health and economic burden for patients. Therefore, there is an urgent need to elucidate the pathological mechanism of PJI to explore new therapeutic methods. This study aimed to explore proteomics changes in bone tissue around the prosthesis during PJI development, to explain the pathological mechanism and to provide new treatment ideas. Ten patients who underwent revision surgery at our institution were included: 5 patients with Staphylococcus aureus PJI and 5 patients with aseptic failure. The proteomics changes in bone tissues after PJI were investigated by label-free quantitative proteomics, and the pathways affected by the differential proteins were analyzed by GO annotation, GO enrichment analysis, KEGG enrichment analysis and protein-protein interaction network analysis. We identified 435 differentially expressed proteins (DEPs), with 213 upregulated and 222 downregulated proteins. Analysis revealed activation of immune-related pathways, such as complement and coagulation cascades, phagocytosis, and neutrophil activation, and inhibition of energy metabolism pathways represented by the TCA cycle. We also observed an altered balance between osteoblasts and osteoclasts during S. aureus PJI. We hope that these processes will reveal new treatment ideas. SIGNIFICANCE: PJI is a catastrophic complication of arthroplasty. When infection occurs, bacteria may invade periprosthetic bone tissue to escape immunity and cause damage. So far, only few studies focused on the changes of proteomics associated to PJI. This is the first study to describe the proteomics changes of periprosthetic bone tissue of patients with PJI. We found that the pathological process of S. aureus PJI mainly involves activation of the immune system, decreased energy metabolism, and an altered balance of osteoblasts and osteoclasts.
Collapse
Affiliation(s)
- Yang Chen
- Department of Orthopedic Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Changyu Huang
- Department of Orthopedic Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Xiaoqing Chen
- Department of Orthopedic Surgery, Quanzhou First Hospital Affiliated to Fujian Medical University, Quanzhou, China
| | - Yuanqing Cai
- Department of Orthopedic Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Wenbo Li
- Department of Orthopedic Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Xinyu Fang
- Department of Orthopedic Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China.
| | - Wenming Zhang
- Department of Orthopedic Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China.
| |
Collapse
|
11
|
Dubus M, Varin J, Papa S, Chevrier J, Quilès F, Francius G, Audonnet S, Mauprivez C, Gangloff S, Siboni R, Ohl X, Reffuveille F, Kerdjoudj H. Bone marrow mesenchymal stem cells offer an immune-privileged niche to Cutibacterium acnes in case of implant-associated osteomyelitis. Acta Biomater 2022; 137:305-315. [PMID: 34678484 DOI: 10.1016/j.actbio.2021.10.026] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 09/23/2021] [Accepted: 10/15/2021] [Indexed: 12/20/2022]
Abstract
Considered as some of the most devastating complications, Cutibacterium acnes (C. acnes)-related osteomyelitis are among the hardest infections to diagnose and treat. Mesenchymal stem cells (MSCs) secrete number of immunomodulatory and antimicrobial soluble factors, making them an attractive treatment for bacterial infection. In this study, we examined MSCs/C. acnes interaction and analyzed the subsequent MSCs and bacteria's behaviors. Human bone marrow-derived MSCs were infected by C. acnes clinical strain harvested from non-infected bone site. Following 3 h of interaction, around 4% of bacteria were found in the intracellular compartment. Infected MSCs increased the secretion of prostaglandin E2 and indolamine 2,3 dioxygenase immunomodulatory mediators. Viable intracellular bacteria analyzed by infrared spectroscopy and atomic force microscopy revealed deep modifications in the wall features. In comparison with unchallenged bacteria, the viable intracellular bacteria showed (i) an increase in biofilm formation on orthopaedical-based materials, (ii) an increase in the invasiveness of osteoblasts and (iii) persistence in macrophage, suggesting the acquisition of virulence factors. Overall, these results showed a direct impact of C. acnes on bone marrow-derived MSCs, suggesting that blocking the C. acnes/MSCs interactions may represent an important new approach to manage chronic osteomyelitis infections. STATEMENT OF SIGNIFICANCE: The interaction of bone commensal C. acnes with bone marrow mesenchymal stem cells induces modifications in C. acnes wall characteristics. These bacteria increased (i) the biofilm formation on orthopaedical-based materials, (ii) the invasiveness of bone forming cells and (iii) the resistance to macrophage clearance through the modification of the wall nano-features and/or the increase in catalase production.
Collapse
|
12
|
Ying YT, Ren WJ, Tan X, Yang J, Liu R, Du AF. Annexin A2-Mediated Internalization of Staphylococcus aureus into Bovine Mammary Epithelial Cells Requires Its Interaction with Clumping Factor B. Microorganisms 2021; 9:2090. [PMID: 34683411 PMCID: PMC8538401 DOI: 10.3390/microorganisms9102090] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2021] [Revised: 09/26/2021] [Accepted: 10/01/2021] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Staphylococcus aureus is a leading cause of contagious mastitis in dairy cattle. Internalization of S. aureus by bovine mammary gland epithelial cells is thought to be responsible for persistent and chronic intramammary infection, but the underlying mechanisms are not fully understood. METHODS In the present study, we evaluated the role of Annexin A2 (AnxA2), a membrane-binding protein, in S. aureus invasion into bovine mammary epithelial cell line (MAC-T). In vitro binding assays were performed to co-immunoprecipitate the binding proteins of AnxA2 in the lysates of S. aureus. RESULTS AnxA2 mediated the internalization but not adherence of S. aureus. Engagement of AnxA2 stimulated an integrin-linked protein kinase (ILK)/p38 MAPK cascade to induce S. aureus invasion. One of the AnxA2-precipitated proteins was identified as S. aureus clumping factor B (ClfB) through use of mass spectrometry. Direct binding of ClfB to AnxA2 was further confirmed by using a pull-down assay. Pre-incubation with recombinant ClfB protein enhanced S. aureus internalization, an effect that was specially blocked by anti-AnxA2 antibody. CONCLUSION Our results demonstrate that binding of ClfB to AnxA2 has a function in promoting S. aureus internalization. Targeting the interaction of ClfB and AnxA2 may confer protection against S. aureus mastitis.
Collapse
Affiliation(s)
- Yi-Tian Ying
- Department of Veterinary Medicine, Zhejiang University, Yuhangtang Road 866, Hangzhou 310058, China; (Y.-T.Y.); (W.-J.R.); (J.Y.); (R.L.); (A.-F.D.)
- Veterinary Medical Center, Zhejiang University, Yuhangtang Road 866, Hangzhou 310058, China
- Institute of Preventive Veterinary Sciences, Zhejiang University, Yuhangtang Road 866, Hangzhou 310058, China
| | - Wei-Jia Ren
- Department of Veterinary Medicine, Zhejiang University, Yuhangtang Road 866, Hangzhou 310058, China; (Y.-T.Y.); (W.-J.R.); (J.Y.); (R.L.); (A.-F.D.)
- Veterinary Medical Center, Zhejiang University, Yuhangtang Road 866, Hangzhou 310058, China
- Institute of Preventive Veterinary Sciences, Zhejiang University, Yuhangtang Road 866, Hangzhou 310058, China
| | - Xun Tan
- Department of Veterinary Medicine, Zhejiang University, Yuhangtang Road 866, Hangzhou 310058, China; (Y.-T.Y.); (W.-J.R.); (J.Y.); (R.L.); (A.-F.D.)
- Veterinary Medical Center, Zhejiang University, Yuhangtang Road 866, Hangzhou 310058, China
- Institute of Preventive Veterinary Sciences, Zhejiang University, Yuhangtang Road 866, Hangzhou 310058, China
| | - Jing Yang
- Department of Veterinary Medicine, Zhejiang University, Yuhangtang Road 866, Hangzhou 310058, China; (Y.-T.Y.); (W.-J.R.); (J.Y.); (R.L.); (A.-F.D.)
- Veterinary Medical Center, Zhejiang University, Yuhangtang Road 866, Hangzhou 310058, China
- Institute of Preventive Veterinary Sciences, Zhejiang University, Yuhangtang Road 866, Hangzhou 310058, China
| | - Rui Liu
- Department of Veterinary Medicine, Zhejiang University, Yuhangtang Road 866, Hangzhou 310058, China; (Y.-T.Y.); (W.-J.R.); (J.Y.); (R.L.); (A.-F.D.)
| | - Ai-Fang Du
- Department of Veterinary Medicine, Zhejiang University, Yuhangtang Road 866, Hangzhou 310058, China; (Y.-T.Y.); (W.-J.R.); (J.Y.); (R.L.); (A.-F.D.)
- Veterinary Medical Center, Zhejiang University, Yuhangtang Road 866, Hangzhou 310058, China
- Institute of Preventive Veterinary Sciences, Zhejiang University, Yuhangtang Road 866, Hangzhou 310058, China
| |
Collapse
|
13
|
Chen ZY, Gao S, Zhang YW, Zhou RB, Zhou F. Antibacterial biomaterials in bone tissue engineering. J Mater Chem B 2021; 9:2594-2612. [PMID: 33666632 DOI: 10.1039/d0tb02983a] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Bone infection is a devastating disease characterized by recurrence, drug-resistance, and high morbidity, that has prompted clinicians and scientists to develop novel approaches to combat it. Currently, although numerous biomaterials that possess excellent biocompatibility, biodegradability, porosity, and mechanical strength have been developed, their lack of effective antibacterial ability substantially limits bone-defect treatment efficacy. There is, accordingly, a pressing need to design antibacterial biomaterials for effective bone-infection prevention and treatment. This review focuses on antibacterial biomaterials and strategies; it presents recently reported biomaterials, including antibacterial implants, antibacterial scaffolds, antibacterial hydrogels, and antibacterial bone cement types, and aims to provide an overview of these antibacterial materials for application in biomedicine. The antibacterial mechanisms of these materials are discussed as well.
Collapse
Affiliation(s)
- Zheng-Yang Chen
- Orthopedic Department, Peking University Third Hospital, Beijing 100191, China.
| | | | | | | | | |
Collapse
|
14
|
Different Modulatory Effects of Four Methicillin-Resistant Staphylococcus aureus Clones on MG-63 Osteoblast-Like Cells. Biomolecules 2021; 11:biom11010072. [PMID: 33430251 PMCID: PMC7825699 DOI: 10.3390/biom11010072] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 11/30/2020] [Accepted: 12/29/2020] [Indexed: 01/18/2023] Open
Abstract
Staphylococcus aureus is a Gram-positive bacterium responsible for a variety of mild to life-threatening infections including bone infections such as osteomyelitis. This bacterium is able to invade and persist within non-professional phagocytic cells such as osteoblasts. In the present study, four different S. aureus strains, namely, 2SA-ST239-III (ST239), 5SA-ST5-II (ST5), 10SA-ST228-I (ST228), and 14SA-ST22-IVh (ST22), were tested for their ability to modulate cell viability in MG-63 osteoblast-like cells following successful invasion and persistence. Methicillin-sensitive S. aureus (MSSA) ATCC-12598-ST30 (ST30) was used as control strain. Despite being proven that ST30, ST239, and ST22 have a similar ability to internalize and persist in MG-63 osteoblast-like cells under our experimental conditions, we demonstrated that the observed decrease in cell viability was due to the different behavior of the considered strains, rather than the number of intracellular bacteria. We focused our attention on different biochemical cell functions related to inflammation, cell metabolism, and oxidative stress during osteoblast infections. We were able to show the following: (1) ST30 and ST239 were the only two clones able to persist and maintain their number in the hostile environment of the cell during the entire period of infection; (2) ST239 was the only clone able to significantly increase gene expression (3 and 24 h post-infection (p.i.)) and protein secretion (24 h p.i.) of both interleukin-6 (IL-6) and tumor necrosis factor alpha (TNF-α) in MG-63 osteoblast-like cells; (3) the same clone determined a significant up-regulation of the transforming growth factorbeta 1 (TGF-β1) and of the metabolic marker glyceraldehyde 3-phosphate dehydrogenase (GAPDH) mRNAs at 24 h p.i.; and (4) neither the MSSA nor the four methicillin-resistant S. aureus (MRSA) strains induced oxidative stress phenomena in MG-63 cells, although a high degree of variability was observed for the different clones with regard to the expression pattern of nuclear factor E2-related factor 2 (Nrf2) and its downstream gene heme oxygenase 1 (HO-1) activation. Our results may pave the way for an approach to S. aureus-induced damage, moving towards individualized therapeutic strategies that take into account the differences between MSSA and MRSA as well as the distinctive features of the different clones. This approach is based on a change of paradigm in antibiotic therapy involving a case-based use of molecules able to counteract pro-inflammatory cytokines activity such as selective cytokine signaling inhibitors (IL-6, TNF-α).
Collapse
|
15
|
Schlesier T, Siegmund A, Rescher U, Heilmann C. Characterization of the Atl-mediated staphylococcal internalization mechanism. Int J Med Microbiol 2020; 310:151463. [PMID: 33197865 DOI: 10.1016/j.ijmm.2020.151463] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 09/16/2020] [Accepted: 10/22/2020] [Indexed: 12/18/2022] Open
Abstract
Staphylococcus aureus internalization by non-professional phagocytes is considered a main pathogenicity mechanism leading to chronic infections. The well-established mechanism of Staphylococcus aureus internalization is mediated by fibronectin (Fn)-binding proteins (FnBPs), Fn as a bridging molecule and the host cell α5β1 integrin. We previously identified a novel alternative internalization mechanism in Staphylococcus aureus, which involves the major autolysin Atl and the host cell heat shock cognate protein 70 (Hsc70). Atl-dependent internalization is also employed by the coagulase-negative Staphylococcus epidermidis, where it might represent the major or even sole internalization mechanism, because of the lack of FnBP-homologous proteins. In this study, we aimed to further characterize the Atl-dependent staphylococcal internalization mechanism. We performed biomolecular interaction analysis (BIA) to quantify the adhesive properties of Atl and found multivalent and high affinity interactions of Atl with Fn and Hsc70. Confocal laser scanning microscopy (CLSM) and a flow-cytometric internalization assay in combination with different pharmacological inhibitors suggested an involvement of the α5β1 integrin, Fn and Hsc70 and subsequent signaling events mediated by Src and phosphoinositide 3 (PI3) kinases in the Atl-dependent staphylococcal uptake by EA.hy 926 cells. Further characterization of the endocytic machinery implicated a role for clathrin-dependent receptor-mediated endocytosis involving actin cytoskeletal rearrangements and microtubules. In conclusion, Atl ubiquitous among staphylococcal species may substitute for the FnBPs ensuring low-level internalization via a mechanism that seems to share important features with the FnBP-mediated staphylococcal uptake potentially being the prerequisite for the development of therapy-resistant chronic infections by staphylococcal strains that lack FnBPs.
Collapse
Affiliation(s)
- Tim Schlesier
- Institute of Medical Microbiology, D-48149, Münster, Germany
| | - Anke Siegmund
- Institute of Medical Microbiology, University Hospital of Jena, D-07747, Jena, Germany
| | - Ursula Rescher
- Institute of Medical Biochemistry, Center for Molecular Biology of Inflammation (ZMBE), D-48149, Münster, Germany; Interdisciplinary Center for Clinical Research (IZKF), University Hospital of Münster, D-48149, Münster, Germany
| | - Christine Heilmann
- Institute of Medical Microbiology, D-48149, Münster, Germany; Interdisciplinary Center for Clinical Research (IZKF), University Hospital of Münster, D-48149, Münster, Germany.
| |
Collapse
|
16
|
Karam J, Méresse S, Kremer L, Daher W. The roles of tetraspanins in bacterial infections. Cell Microbiol 2020; 22:e13260. [PMID: 32902857 DOI: 10.1111/cmi.13260] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 07/22/2020] [Accepted: 07/24/2020] [Indexed: 12/18/2022]
Abstract
Tetraspanins, a wide family composed of 33 transmembrane proteins, are associated with different types of proteins through which they arbitrate important cellular processes such as fusion, adhesion, invasion, tissue differentiation and immunological responses. Tetraspanins share a comparable structural design, which consists of four hydrophobic transmembrane domains with cytoplasmic and extracellular loops. They cooperate with different proteins, including other tetraspanins, receptors or signalling proteins to compose functional complexes at the cell surface, designated tetraspanin-enriched microdomains (TEM). Increasing evidences establish that tetraspanins are exploited by numerous intracellular pathogens as a doorway for entering and replicating within human cells. Although previous surveys focused mainly on viruses and parasites, it is now becoming clear that bacteria interact with tetraspanins, using TEM as a "gateway" to infection. In this review, we examine the biological functions of tetraspanins that are relevant to bacterial infective procedures and consider the available data that reveal how different bacteria benefit from host cell tetraspanins in infection and in the pathogenesis of diseases. We will also emphasise the stimulating potentials of targeting tetraspanins for preventing bacterial infectious diseases, using specific neutralising antibodies or anti-adhesion peptide-based therapies. Such innovative therapeutic opportunities may deliver alternatives for fighting difficult-to-manage and drug-resistant bacterial pathogens.
Collapse
Affiliation(s)
- Jona Karam
- Centre National de la Recherche Scientifique UMR 9004, Institut de Recherche en Infectiologie de Montpellier (IRIM), Université de Montpellier, Montpellier, France
| | | | - Laurent Kremer
- Centre National de la Recherche Scientifique UMR 9004, Institut de Recherche en Infectiologie de Montpellier (IRIM), Université de Montpellier, Montpellier, France.,INSERM, IRIM, Montpellier, France
| | - Wassim Daher
- Centre National de la Recherche Scientifique UMR 9004, Institut de Recherche en Infectiologie de Montpellier (IRIM), Université de Montpellier, Montpellier, France.,INSERM, IRIM, Montpellier, France
| |
Collapse
|
17
|
Dubus M, Varin-Simon J, Prada P, Scomazzon L, Reffuveille F, Alem H, Boulmedais F, Mauprivez C, Rammal H, Kerdjoudj H. Biopolymers-calcium phosphate antibacterial coating reduces the pathogenicity of internalized bacteria by mesenchymal stromal cells. Biomater Sci 2020; 8:5763-5773. [PMID: 32945302 DOI: 10.1039/d0bm00962h] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
A multifunctional material system that kills bacteria and drives bone healing is urgently sought to improve bone prosthesis. Herein, the osteoinductive coating made of calcium phosphate/chitosan/hyaluronic acid, named Hybrid, was proposed as an antibacterial substrate for stromal cell adhesion. This Hybrid coating possesses a contact-killing effect reducing by 90% the viability of Gram-positive Staphylococcus aureus (S. aureus) and Gram-negative Pseudomonas aeruginosa (P. aeruginosa) strains after 48 h of contact. In addition to the production of immunomodulatory mediators, Wharton's jelly (WJ-SCs), dental pulp (DPSCs) and bone marrow (BM-MSCs) derived stromal cells were able to release antibacterial and antibiofilm agents effective against S. aureus and P. aeruginosa strains, respectively. Studying the effect of the Hybrid coating on the internalization of S. aureus by the stromal cells, in acute-mimicking bone infection, highlighted an increase in the bacteria internalization by DPSCs and BM-MSCs when cultured on the Hybrid coating versus uncoated glass. Despite the internalization, Hybrid coating showed a beneficial effect by reducing the pathogenicity of the internalized bacteria. The formation of biofilm was reduced by at least 50% in comparison to internalized bacteria by stromal cells on uncoated glass. This work opens the route for the development of innovative antibacterial coatings by taking into account the internalization of bacteria by stromal cells.
Collapse
Affiliation(s)
- Marie Dubus
- Université de Reims Champagne Ardenne, EA 4691, Biomatériaux et Inflammation en Site Osseux (BIOS), Reims, France.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Guzmán-Rodríguez JJ, León-Galván MF, Barboza-Corona JE, Valencia-Posadas M, Loeza-Lara PD, Sánchez-Ceja M, Ochoa-Zarzosa A, López-Meza JE, Gutiérrez-Chávez AJ. Analysis of virulence traits of Staphylococcus aureus isolated from bovine mastitis in semi-intensive and family dairy farms. J Vet Sci 2020; 21:e77. [PMID: 33016022 PMCID: PMC7533384 DOI: 10.4142/jvs.2020.21.e77] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 07/21/2020] [Accepted: 08/11/2020] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Staphylococcus aureus is one of the main microorganisms that causes bovine mastitis, and its well-known virulence characteristics and interactions with the environment are used to aid the design of more efficient therapies. OBJECTIVES To determine whether the virulence traits, such as antibiotic resistance and biofilm-forming and internalization abilities, of S. aureus isolated from bovine mastitis are related to dairy production system types. METHODS The study was performed in the Mexican states of Guanajuato and Michoacan. Semi-intensive dairy farms (SIDFs) and family dairy farms (FDFs) (454 and 363 cows, respectively) were included. The 194 milk samples from mastitis affected quarters were collected and 92 strains of S. aureus were isolated and identified by biochemical and molecular tests. Antibiotic resistance, biofilm and internalization assays were performed on 30 randomly selected isolated strains to determine virulence traits, and these strains were equally allocated to the 2 dairy production systems. RESULTS All 30 selected strains displayed a high degree of resistance (50%-91.7%) to the antibiotics tested, but no significant difference was found between SIDF and FDF isolates. S. aureus strains from SIDFs had an average biofilm forming capacity of up to 36% (18.9%-53.1%), while S. aureus strains from FDFs registered an average of up to 53% (31.5%-77.8%) (p > 0.05). Internalization assays revealed a higher frequency of internalization capacity for strains isolated from FDFs (33.3%) than for those isolated from SIDFs (6.7%) (p > 0.05). fnbpA gen was detected in 46.6% of FDF strains and 33.3% of SIDF strains, and this difference was significant (p < 0.05). CONCLUSIONS Our findings show that the virulence traits of S. aureus isolates analyzed in this study, depend significantly on several factors, such as phenotype, genotype, and environmental conditions, which are significantly related to dairy production system type and daily management practices.
Collapse
Affiliation(s)
- Jaquelina J Guzmán-Rodríguez
- Life Science Division, Postgraduate Program in Bioscience, Irapuato-Salamanca Campus, University of Guanajuato, 36500 Irapuato, Guanajuato, Mexico
| | - Ma Fabiola León-Galván
- Life Science Division, Postgraduate Program in Bioscience, Irapuato-Salamanca Campus, University of Guanajuato, 36500 Irapuato, Guanajuato, Mexico
- Life Science Division, Department of Food, Irapuato-Salamanca Campus, University of Guanajuato, 36500 Irapuato, Guanajuato, Mexico
| | - José E Barboza-Corona
- Life Science Division, Postgraduate Program in Bioscience, Irapuato-Salamanca Campus, University of Guanajuato, 36500 Irapuato, Guanajuato, Mexico
- Life Science Division, Department of Food, Irapuato-Salamanca Campus, University of Guanajuato, 36500 Irapuato, Guanajuato, Mexico
| | - Mauricio Valencia-Posadas
- Life Science Division, Postgraduate Program in Bioscience, Irapuato-Salamanca Campus, University of Guanajuato, 36500 Irapuato, Guanajuato, Mexico
- Life Science Division, Department of Veterinary Medicine and Zootechnics, Irapuato-Salamanca Campus, University of Guanajuato, 36500 Irapuato, Guanajuato, Mexico
| | - Pedro D Loeza-Lara
- Department of Food Genomics, University of La Cienega of the State of Michoacan de Ocampo, 59103 Sahuayo, Michoacan, Mexico
| | - Mónica Sánchez-Ceja
- Department of Food Genomics, University of La Cienega of the State of Michoacan de Ocampo, 59103 Sahuayo, Michoacan, Mexico
| | - Alejandra Ochoa-Zarzosa
- Multidisciplinary Center for Biotechnology Studies, Faculty of Veterinary Medicine and Zootechnics, Universidad Michoacana de San Nicolas de Hidalgo, 58893 Morelia, Michoacán, Mexico
| | - Joel E López-Meza
- Multidisciplinary Center for Biotechnology Studies, Faculty of Veterinary Medicine and Zootechnics, Universidad Michoacana de San Nicolas de Hidalgo, 58893 Morelia, Michoacán, Mexico
| | - Abner J Gutiérrez-Chávez
- Life Science Division, Postgraduate Program in Bioscience, Irapuato-Salamanca Campus, University of Guanajuato, 36500 Irapuato, Guanajuato, Mexico
- Life Science Division, Department of Veterinary Medicine and Zootechnics, Irapuato-Salamanca Campus, University of Guanajuato, 36500 Irapuato, Guanajuato, Mexico.
| |
Collapse
|
19
|
Prolactin and Estradiol are Epigenetic Modulators in Bovine Mammary Epithelial Cells during Staphylococcus aureus Infection. Pathogens 2020; 9:pathogens9070520. [PMID: 32605209 PMCID: PMC7399903 DOI: 10.3390/pathogens9070520] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 06/22/2020] [Accepted: 06/23/2020] [Indexed: 12/24/2022] Open
Abstract
Changes in the levels of reproductive hormones compromise the bovine innate immune response (IIR). Changes in 17β-estradiol (E2) and prolactin (bPRL) levels affect the IIR of bovine mammary epithelial cells (bMECs), the target tissue of these hormones. In this work, we explored the effect of the combined hormones on bMEC IIR during Staphylococcus aureus infection, and if they can modulate epigenetic marks. By gentamicin protection assays, we determined that combined hormones (bPRL (5 ng/mL) and E2 (50 pg/mL)] decrease S. aureus internalization into bMECs (~50%), which was associated with a reduction in integrin α5β1 membrane abundance (MA) (~80%) determined by flow cytometry. Additionally, combined hormones increased Toll-like receptor 2 (TLR2) MA (~25%). By RT-qPCR, we showed that combined hormones induce the expression of pro- and anti-inflammatory cytokine genes, as well as up-regulate antimicrobial peptide gene expression. The combined hormones induced H3K9Ac at 12 h of treatment, which coincides with the reduction in histone deacetylase (HDAC, ~15%) activity. In addition, hormones increased the H3K9me2 mark at 12 h, which correlates with a reduction in the expression of KDM4A. In conclusion, bPRL and E2 modulate the IIR of bMECs, an effect that can be related to the regulation of histone H3 modifications such as H3K9Ac and H3K9me2.
Collapse
|
20
|
Cathelicidins Mitigate Staphylococcus aureus Mastitis and Reduce Bacterial Invasion in Murine Mammary Epithelium. Infect Immun 2020; 88:IAI.00230-20. [PMID: 32341117 DOI: 10.1128/iai.00230-20] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Accepted: 04/17/2020] [Indexed: 01/27/2023] Open
Abstract
Staphylococcus aureus, an important cause of mastitis in mammals, is becoming increasingly problematic due to the development of resistance to conventional antibiotics. The ability of S. aureus to invade host cells is key to its propensity to evade immune defense and antibiotics. This study focuses on the functions of cathelicidins, small cationic peptides secreted by epithelial cells and leukocytes, in the pathogenesis of S. aureus mastitis in mice. We determined that endogenous murine cathelicidin (CRAMP; Camp) was important in controlling S. aureus infection, as cathelicidin knockout mice (Camp-/- ) intramammarily challenged with S. aureus had higher bacterial burdens and more severe mastitis than did wild-type mice. The exogenous administration of both a synthetic human cathelicidin (LL-37) and a synthetic murine cathelicidin (CRAMP) (8 μM) reduced the invasion of S. aureus into the murine mammary epithelium. Additionally, this exogenous LL-37 was internalized into cultured mammary epithelial cells and impaired S. aureus growth in vitro We conclude that cathelicidins may be potential therapeutic agents against mastitis; both endogenous and exogenous cathelicidins conferred protection against S. aureus infection by reducing bacterial internalization and potentially by directly killing this pathogen.
Collapse
|
21
|
Abstract
Phagocytosis is a pivotal immunological process, and its discovery by Elia Metchnikoff in 1882 was a step toward the establishment of the innate immune system as a separate branch of immunology. Elia Metchnikoff received the Nobel Prize in physiology and medicine for this discovery in 1908. Since its discovery almost 140 years before, phagocytosis remains the hot topic of research in immunology. The phagocytosis research has seen a great advancement since its first discovery. Functionally, phagocytosis is a simple immunological process required to engulf and remove pathogens, dead cells and tumor cells to maintain the immune homeostasis. However, mechanistically, it is a very complex process involving different mechanisms, induced and regulated by several pattern recognition receptors, soluble pattern recognition molecules, scavenger receptors (SRs) and opsonins. These mechanisms involve the formation of phagosomes, their maturation into phagolysosomes causing pathogen destruction or antigen synthesis to present them to major histocompatibility complex molecules for activating an adaptive immune response. Any defect in this mechanism may predispose the host to certain infections and inflammatory diseases (autoinflammatory and autoimmune diseases) along with immunodeficiency. The article is designed to discuss its mechanistic complexity at each level, varying from phagocytosis induction to phagolysosome resolution.
Collapse
Affiliation(s)
- Vijay Kumar
- Faculty of Medicine, Children's Health Queensland Clinical Unit, School of Clinical Medicine, Mater Research, University of Queensland, ST Lucia, Brisbane, Queensland, Australia.,Faculty of Medicine, School of Biomedical Sciences, University of Queensland, St Lucia, Brisbane, Queensland, Australia
| |
Collapse
|
22
|
Lipid-Rich Extract from Mexican Avocado Seed ( Persea americana var. drymifolia) Reduces Staphylococcus aureus Internalization and Regulates Innate Immune Response in Bovine Mammary Epithelial Cells. J Immunol Res 2019; 2019:7083491. [PMID: 31612151 PMCID: PMC6757280 DOI: 10.1155/2019/7083491] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Accepted: 08/23/2019] [Indexed: 12/16/2022] Open
Abstract
Bovine mammary epithelial cells (bMECs) are capable of initiating an innate immune response (IIR) to invading bacteria. Staphylococcus aureus is not classically an intracellular pathogen, although it has been shown to be internalized into bMECs. S. aureus internalizes into nonprofessional phagocytes, which allows the evasion of the IIR and turns antimicrobial therapy unsuccessful. An alternative treatment to control this pathogen is the modulation of the innate immune response of the host. The Mexican avocado (Persea americana var. drymifolia) is a source of molecules with anti-inflammatory and immunomodulatory properties. Hence, we analyze the effect of a lipid-rich extract from avocado seed (LEAS) on S. aureus internalization into bMECs and their innate immunity response. The effects of LEAS (1-500 ng/ml) on the S. aureus growth and bMEC viability were assessed by turbidimetry and MTT assays, respectively. LEAS did not show neither antimicrobial nor cytotoxic effects. S. aureus internalization into bMECs was analyzed by gentamicin protection assays. Interestingly, LEAS (1-200 ng/ml) decreased bacterial internalization (60-80%) into bMECs. This effect correlated with NO production and the induction of the gene expression of IL-10, while the expression of the proinflammatory cytokine TNF-α was reduced. These effects could be related to the inhibition of MAPK p38 (∼60%) activation by LEAS. In conclusion, our results showed that LEAS inhibits the S. aureus internalization into bMECs and modulates the IIR, which indicates that avocado is a source of metabolites for control of mastitis pathogens.
Collapse
|
23
|
Mu M, Wu F, He J, Tang X, Ma H, Guo S, Song C. Insulin‑like growth factor 1 inhibits phagocytosis of alveolar epithelial cells in asthmatic mice. Mol Med Rep 2019; 20:2381-2388. [PMID: 31322198 DOI: 10.3892/mmr.2019.10456] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2018] [Accepted: 06/04/2019] [Indexed: 11/06/2022] Open
Abstract
The phagocytosis of apoptotic cells by alveolar epithelial cells helps to eliminate airway inflammation. Insulin‑like growth factor 1 (IGF‑1) regulates cell metabolism and proliferation, and promotes cell survival, while it may also promote the proliferation and differentiation of alveolar epithelial cells during the repair of lung injury. The present study investigated the effect of IGF‑1 on the phagocytic activity of alveolar epithelial cells, a nonprofessional phagocyte. IGF‑1 was elevated in lung tissue and bronchoalveolar lavage fluid obtained from mice with ovalbumin‑induced asthma. IGF‑1 was reduced by 50% in the lung tissue and by nearly 100% in the bronchoalveolar lavage fluid in asthmatic mice established by depletion of alveolar macrophages using 2‑chloroadenosine. In addition, interleukin‑33 induced IGF‑1 production in primary alveolar macrophages. It was also observed that IGF‑1 inhibited the phagocytosis of fluorescent microspheres and apoptotic cells by MLE‑12 alveolar epithelial cells. Antibody blocking of IGF‑1 enhanced the phagocytosis of fluorescent microspheres and apoptotic cells, and significantly reduced inflammatory cell infiltration in airway and perivascular tissues. The elevated IGF‑1 level in the lungs of asthma model mice was mainly produced in alveolar macrophages. Taken together, the current study demonstrated that IGF‑1 inhibited phagocytosis by alveolar epithelial cells, and that IGF‑1 blockade enhanced the phagocytic activity and alleviated airway inflammation. These results support the potential use of IGF‑1 as a target in the treatment of asthma.
Collapse
Affiliation(s)
- Mimi Mu
- Department of Immunology, Anhui Provincial Key Laboratory of Infection and Immunity, Bengbu Medical College, Bengbu, Anhui 233030, P.R. China
| | - Fengjiao Wu
- Department of Immunology, Anhui Provincial Key Laboratory of Infection and Immunity, Bengbu Medical College, Bengbu, Anhui 233030, P.R. China
| | - Jing He
- Department of Immunology, Anhui Provincial Key Laboratory of Infection and Immunity, Bengbu Medical College, Bengbu, Anhui 233030, P.R. China
| | - Xu Tang
- Department of Clinical Laboratory Medicine, Bengbu Medical College, Bengbu, Anhui 233030, P.R. China
| | - Hua Ma
- Department of Immunology, Anhui Provincial Key Laboratory of Infection and Immunity, Bengbu Medical College, Bengbu, Anhui 233030, P.R. China
| | - Shujun Guo
- Department of Immunology, Anhui Provincial Key Laboratory of Infection and Immunity, Bengbu Medical College, Bengbu, Anhui 233030, P.R. China
| | - Chuanwang Song
- Department of Immunology, Anhui Provincial Key Laboratory of Infection and Immunity, Bengbu Medical College, Bengbu, Anhui 233030, P.R. China
| |
Collapse
|
24
|
Frutis-Murillo M, Sandoval-Carrillo MA, Alva-Murillo N, Ochoa-Zarzosa A, López-Meza JE. Immunomodulatory molecules regulate adhesin gene expression in Staphylococcus aureus: Effect on bacterial internalization into bovine mammary epithelial cells. Microb Pathog 2019; 131:15-21. [PMID: 30930221 DOI: 10.1016/j.micpath.2019.03.030] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Revised: 03/11/2019] [Accepted: 03/25/2019] [Indexed: 11/26/2022]
Abstract
Staphylococcus aureus is a major pathogen of subclinical bovine mastitis that usually is chronic and recurrent, which has been related to its ability to internalize into bovine mammary epithelial cells (bMECs). Previously, we reported that short and medium fatty acids and cholecalciferol reduce S. aureus internalization into pretreated-bMECs with these molecules suggesting a role as immunomodulatory agents. Hence, we assessed the role of sodium butyrate (NaB), sodium octanoate (NaO) and cholecalciferol on S. aureus adhesin expression and its internalization into bMECs. S. aureus pre-treated 2 h with 0.5 mM or 2 mM NaB showed a reduction in internalization into bMECs (∼35% and ∼55%; respectively), which coincided with a down-regulated expression of clumping factor B (ClfB). Also, the S. aureus internalization reduction by 2 mM NaB (2 h) agreed with a down-regulated expression of sdrC. Moreover, the 2 mM NaB (24 h) pre-treatment induced bacterial internalization (∼3-fold), which was related with an up-regulation of spa, clfB and sdrC genes. Also, NaO (0.25 mM and 1 mM) only reduced S. aureus internalization when bacteria were grown 2 h with this molecule but there was no relationship with adhesin expression. In addition, cholecalciferol (50 nM) reduced bacteria internalization at similar levels (∼50%) when bacteria were grown 2 and 24 h in broth supplemented with this compound, which correlated with spa and sdrC mRNA expression down-regulated at 2 h, and fnba and clfB mRNA expression decreased at 24 h. In conclusion, our data support the fact that fatty acids and cholecalciferol regulate adhesin gene expression as well as bacteria internalization in nonprofessional phagocytic cells, which may lead to development of anti-virulence agents for control of pathogens.
Collapse
Affiliation(s)
- Minerva Frutis-Murillo
- Centro Multidisciplinario de Estudios en Biotecnología, Facultad de Medicina Veterinaria y Zootecnia, Universidad Michoacana de San Nicolás de Hidalgo, Morelia, Michoacán, Mexico
| | - Marcelo Alejandro Sandoval-Carrillo
- Centro Multidisciplinario de Estudios en Biotecnología, Facultad de Medicina Veterinaria y Zootecnia, Universidad Michoacana de San Nicolás de Hidalgo, Morelia, Michoacán, Mexico
| | - Nayeli Alva-Murillo
- Departamento de Biología, División de Ciencias Naturales y Exactas, Universidad de Guanajuato, Guanajuato, Mexico
| | - Alejandra Ochoa-Zarzosa
- Centro Multidisciplinario de Estudios en Biotecnología, Facultad de Medicina Veterinaria y Zootecnia, Universidad Michoacana de San Nicolás de Hidalgo, Morelia, Michoacán, Mexico
| | - Joel E López-Meza
- Centro Multidisciplinario de Estudios en Biotecnología, Facultad de Medicina Veterinaria y Zootecnia, Universidad Michoacana de San Nicolás de Hidalgo, Morelia, Michoacán, Mexico.
| |
Collapse
|
25
|
Toll-Like Receptor 2 and Lipoprotein-Like Lipoproteins Enhance Staphylococcus aureus Invasion in Epithelial Cells. Infect Immun 2018; 86:IAI.00343-18. [PMID: 29844243 DOI: 10.1128/iai.00343-18] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Accepted: 05/23/2018] [Indexed: 01/05/2023] Open
Abstract
Staphylococcus aureus contains a certain subclass of lipoproteins, the so-called lipoprotein-like lipoproteins (Lpl's), that not only represent Toll-like receptor 2 (TLR2) ligands but are also involved in host cell invasion. Here we addressed the question of which factors contribute to Lpl-mediated invasion of epithelial cells and keratinocytes. For this purpose, we compared the invasiveness of USA300 and its Δlpl mutant under different conditions. In the presence of the matrix proteins IgG, fibrinogen (Fg), and fibronectin (Fn), and of fetal bovine serum (FBS), the invasion ratio was increased in both strains, and always more in USA300 than in its Δlpl mutant. Interestingly, when we compared the invasion of HEK-0 and HEK-TLR2 cells, the cells expressing TLR2 showed a 9-times-higher invasion frequency. When HEK-TLR2 cells were additionally stimulated with a synthetic lipopeptide, Pam3CSK4 (P3C), the invasion frequency was further increased. A potential reason for the positive effect of TLR2 on invasion could be that TLR2 activation by P3C also activates F-actin formation. Here we show that S. aureus invasion depends on a number of factors, on the host side as well as on the bacterial side.
Collapse
|
26
|
Silva LO, Nobre LS, Mil-Homens D, Fialho A, Saraiva LM. Repair of Iron Centers RIC protein contributes to the virulence of Staphylococcus aureus. Virulence 2018; 9:312-317. [PMID: 29020514 PMCID: PMC5955197 DOI: 10.1080/21505594.2017.1389829] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
RICs are a family of bacterial proteins involved in the repair of iron centers containing proteins damaged by the antimicrobial reactive species liberated by the innate immune system of infected hosts. Staphylococcus aureus is a human pathogen with increasing antibiotic resistance that also contains a RIC-like protein. In this work, we show that the survival of S. aureus within macrophages decreases upon inactivation of ric, and that the viability was restored to levels similar to the wild-type strain by reintroduction of ric via in trans complementation. Importantly, in macrophages that do not produce reactive oxygen species, the lower survival of the ric mutant was no longer observed. In lung epithelial cells, the intracellular viability of the S. aureus ric mutant was also shown to be lower than that of the wild-type. The wax moth larvae Galleria mellonella infected with S. aureus ric mutant presented an approximately 2.5-times higher survival when compared to the wild-type strain. Moreover, significantly lower bacterial loads were determined in the larvae hemolymph infected with strains not expressing ric, and complementation assays confirmed that this behavior was related to RIC. Furthermore, expression of the S. aureus ric gene within the larvae increased along the course of infection with a ~20-fold increase after 8 h of infection. Altogether, the data show that RIC is important for the virulence of S. aureus.
Collapse
Affiliation(s)
- Liliana O Silva
- a Instituto de Tecnologia Química e Biológica NOVA , Av. da República Oeiras , Portugal
| | - Lígia S Nobre
- a Instituto de Tecnologia Química e Biológica NOVA , Av. da República Oeiras , Portugal
| | - Dalila Mil-Homens
- b Institute for Bioengineering and Biosciences (iBB), Instituto Superior Técnico , Av. Rovisco Pais, 1, Lisboa , Portugal
| | - Arsénio Fialho
- b Institute for Bioengineering and Biosciences (iBB), Instituto Superior Técnico , Av. Rovisco Pais, 1, Lisboa , Portugal
| | - Lígia M Saraiva
- a Instituto de Tecnologia Química e Biológica NOVA , Av. da República Oeiras , Portugal
| |
Collapse
|
27
|
Hepburn L, Hijnen D, Sellman B, Mustelin T, Sleeman M, May R, Strickland I. The complex biology and contribution of Staphylococcus aureus
in atopic dermatitis, current and future therapies. Br J Dermatol 2017; 177:63-71. [DOI: 10.1111/bjd.15139] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/15/2016] [Indexed: 12/22/2022]
Affiliation(s)
- L. Hepburn
- MedImmune; Milstein Building; Granta Park; Cambridge CB21 6GH U.K
| | - D.J. Hijnen
- University Medical Center Utrecht; Department of Dermatology; Utrecht the Netherlands
| | | | | | - M.A. Sleeman
- MedImmune; Milstein Building; Granta Park; Cambridge CB21 6GH U.K
| | - R.D. May
- MedImmune; Milstein Building; Granta Park; Cambridge CB21 6GH U.K
| | - I. Strickland
- MedImmune; Milstein Building; Granta Park; Cambridge CB21 6GH U.K
| |
Collapse
|
28
|
Alva-Murillo N, Ochoa-Zarzosa A, López-Meza JE. Sodium Octanoate Modulates the Innate Immune Response of Bovine Mammary Epithelial Cells through the TLR2/P38/JNK/ERK1/2 Pathway: Implications during Staphylococcus aureus Internalization. Front Cell Infect Microbiol 2017; 7:78. [PMID: 28361042 PMCID: PMC5350129 DOI: 10.3389/fcimb.2017.00078] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2016] [Accepted: 02/27/2017] [Indexed: 12/19/2022] Open
Abstract
Bovine mammary epithelial cells (bMECs) contribute to mammary gland defense against invading pathogens, such as Staphylococcus aureus (intracellular facultative), which is recognized by TLR2. In a previous report, we showed that sodium octanoate [NaO, a medium chain fatty acid (C8)] induces (0.25 mM) or inhibits (1 mM) S. aureus internalization into bMECs and differentially regulates the innate immune response (IIR). However, the molecular mechanisms have not been described, which was the aim of this study. The results showed that α5β1 integrin membrane abundance (MA) was increased in 0.25 mM NaO-treated cells, but TLR2 or CD36 MA was not modified. When these receptors were blocked individually, 0.25 mM NaO-increased S. aureus internalization was notably reduced. Interestingly, in this condition, the IIR of the bMECs was impaired because MAPK (p38, JNK, and ERK1/2) phosphorylation and the activation of transcription factors related to these pathways were decreased. In addition, the 1 mM NaO treatment induced TLR2 MA, but neither the integrin nor CD36 MA was modified. The reduction in S. aureus internalization induced by 1 mM NaO was increased further when TLR2 was blocked. In addition, the phosphorylation levels of the MAPKs increased, and 13 transcriptional factors related to the IIR were slightly activated (CBF, CDP, c-Myb, AP-1, Ets-1/Pea-3, FAST-1, GAS/ISRE, AP-2, NFAT-1, OCT-1, RAR/DR-5, RXR/DR-1, and Stat-3). Moreover, the 1 mM NaO treatment up-regulated gene expression of IL-8 and RANTES and secretion of IL-1β. Notably, when 1 mM NaO-treated bMECs were challenged with S. aureus, the gene expression of IL-8 and IL-10 increased, while IL-1β secretion was reduced. In conclusion, our results showed that α5β1 integrin, TLR2 and CD36 are involved in 0.25 mM NaO-increased S. aureus internalization in bMECs. In addition, 1 mM NaO activates bMECs via the TLR2 signaling pathways (p38, JNK, and ERK1/2), which improves IIR before S. aureus invasion. Additionally, NaO (1 mM) might exert anti-inflammatory effects after bacterial internalization.
Collapse
Affiliation(s)
- Nayeli Alva-Murillo
- Licenciatura en Genómica Alimentaria, Universidad de La Ciénega del Estado de Michoacán de OcampoSahuayo, Mexico
| | - Alejandra Ochoa-Zarzosa
- Facultad de Medicina Veterinaria y Zootecnia, Centro Multidisciplinario de Estudios en Biotecnología, Universidad Michoacana de San Nicolás de HidalgoMorelia, Mexico
| | - Joel E. López-Meza
- Facultad de Medicina Veterinaria y Zootecnia, Centro Multidisciplinario de Estudios en Biotecnología, Universidad Michoacana de San Nicolás de HidalgoMorelia, Mexico
| |
Collapse
|
29
|
Tet38 Efflux Pump Affects Staphylococcus aureus Internalization by Epithelial Cells through Interaction with CD36 and Contributes to Bacterial Escape from Acidic and Nonacidic Phagolysosomes. Infect Immun 2017; 85:IAI.00862-16. [PMID: 27956597 DOI: 10.1128/iai.00862-16] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Accepted: 12/04/2016] [Indexed: 11/20/2022] Open
Abstract
We previously reported that the Tet38 efflux pump is involved in internalization of Staphylococcus aureus by A549 lung epithelial cells. A lack of tet38 reduced bacterial uptake by A549 cells to 36% of that of the parental strain RN6390. Using invasion assays coupled with confocal microscopy imaging, we studied the host cell receptor(s) responsible for bacterial uptake via interaction with Tet38. We also assessed the ability of S. aureus to survive following alkalinization of the phagolysosomes by chloroquine. Antibody to the scavenger receptor CD36 reduced the internalization of S. aureus RN6390 by A549 cells, but the dependence on CD36 was reduced in QT7 tet38, suggesting that an interaction between Tet38 and CD36 contributed to S. aureus internalization. Following fusion of the S. aureus-associated endosomes with lysosomes, alkalinization of the acidic environment with chloroquine led to a rapid increase in the number of S. aureus RN6390 bacteria in the cytosol, followed by a decrease shortly thereafter. This effect of chloroquine was not seen in the absence of intact Tet38 in mutant QT7. These data taken together suggest that Tet38 plays a role both in bacterial internalization via interaction with CD36 and in bacterial escape from the phagolysosomes.
Collapse
|
30
|
Richter E, Harms M, Ventz K, Nölker R, Fraunholz MJ, Mostertz J, Hochgräfe F. Quantitative Proteomics Reveals the Dynamics of Protein Phosphorylation in Human Bronchial Epithelial Cells during Internalization, Phagosomal Escape, and Intracellular Replication of Staphylococcus aureus. J Proteome Res 2016; 15:4369-4386. [DOI: 10.1021/acs.jproteome.6b00421] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Affiliation(s)
- Erik Richter
- Competence
Center Functional Genomics, Junior Research Group Pathoproteomics, University of Greifswald, 17489 Greifswald, Germany
| | - Manuela Harms
- Competence
Center Functional Genomics, Junior Research Group Pathoproteomics, University of Greifswald, 17489 Greifswald, Germany
| | - Katharina Ventz
- Competence
Center Functional Genomics, Junior Research Group Pathoproteomics, University of Greifswald, 17489 Greifswald, Germany
| | - Rolf Nölker
- Competence
Center Functional Genomics, Junior Research Group Pathoproteomics, University of Greifswald, 17489 Greifswald, Germany
| | | | - Jörg Mostertz
- Competence
Center Functional Genomics, Junior Research Group Pathoproteomics, University of Greifswald, 17489 Greifswald, Germany
| | - Falko Hochgräfe
- Competence
Center Functional Genomics, Junior Research Group Pathoproteomics, University of Greifswald, 17489 Greifswald, Germany
| |
Collapse
|
31
|
Plotkin BJ, Sigar IM, Tiwari V, Halkyard S. Determination of Biofilm Initiation on Virus-infected Cells by Bacteria and Fungi. J Vis Exp 2016. [PMID: 27501265 DOI: 10.3791/54162] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
The study of polymicrobial interactions across the taxonomic kingdoms that include fungi, bacteria and virus have not been previously examined with respect to how viral members of the microbiome affect subsequent microbe interactions with these virus-infected host cells. The co-habitation of virus with bacteria and fungi is principally present on the mucosal surfaces of the oral cavity and genital tract. Mucosal cells, particularly those with persistent chronic or persistent latent viral infections, could have a significant impact on members of the microbiome through virus alteration in number and type of receptors expressed. Modification in host cell membrane architecture would result in altered ability of subsequent members of the normal flora and opportunistic pathogens to initiate the first step in biofilm formation, i.e., adherence. This study describes a method for quantitation and visual examination of HSV's effect on the initiation of biofilm formation (adherence) of S. aureus and C. albicans.
Collapse
Affiliation(s)
| | - Ira M Sigar
- Department of Microbiology and Immunology, Midwestern University
| | - Vaibhav Tiwari
- Department of Microbiology and Immunology, Midwestern University
| | - Scott Halkyard
- Department of Microbiology and Immunology, Midwestern University
| |
Collapse
|
32
|
Ou J, Drilling A, Singhal D, Tan NCW, Wallis-Hill D, Vreugde S, Psaltis AJ, Wormald PJ. Association of intracellularStaphylococcus aureuswith prognosis in chronic rhinosinusitis. Int Forum Allergy Rhinol 2016; 6:792-9. [DOI: 10.1002/alr.21758] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2015] [Revised: 02/01/2016] [Accepted: 02/04/2016] [Indexed: 01/11/2023]
Affiliation(s)
- Judy Ou
- Department of Surgery, Otorhinolaryngology Head and Neck Surgery; The University of Adelaide; Adelaide Australia
| | - Amanda Drilling
- Department of Surgery, Otorhinolaryngology Head and Neck Surgery; The University of Adelaide; Adelaide Australia
| | - Deepti Singhal
- Department of Surgery, Otorhinolaryngology Head and Neck Surgery; The University of Adelaide; Adelaide Australia
| | - Neil C.-W. Tan
- Department of Surgery, Otorhinolaryngology Head and Neck Surgery; The University of Adelaide; Adelaide Australia
| | - Deanna Wallis-Hill
- Anatomical Pathology Department; Adelaide Pathology Partner; Adelaide Australia
| | - Sarah Vreugde
- Department of Surgery, Otorhinolaryngology Head and Neck Surgery; The University of Adelaide; Adelaide Australia
| | - Alkis J. Psaltis
- Department of Surgery, Otorhinolaryngology Head and Neck Surgery; The University of Adelaide; Adelaide Australia
| | - Peter-John Wormald
- Department of Surgery, Otorhinolaryngology Head and Neck Surgery; The University of Adelaide; Adelaide Australia
| |
Collapse
|
33
|
N'Diaye A, Mijouin L, Hillion M, Diaz S, Konto-Ghiorghi Y, Percoco G, Chevalier S, Lefeuvre L, Harmer NJ, Lesouhaitier O, Feuilloley MGJ. Effect of Substance P in Staphylococcus aureus and Staphylococcus epidermidis Virulence: Implication for Skin Homeostasis. Front Microbiol 2016; 7:506. [PMID: 27148195 PMCID: PMC4832252 DOI: 10.3389/fmicb.2016.00506] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Accepted: 03/29/2016] [Indexed: 11/13/2022] Open
Abstract
Staphylococcus aureus and Staphylococcus epidermidis are two major skin associated bacteria, and Substance P (SP) is a major skin neuropeptide. Since bacteria are known to sense and response to many human hormones, we investigated the effects of SP on Staphylococci virulence in reconstructed human epidermis model and HaCaT keratinocytes. We show that SP is stimulating the virulence of S. aureus and S. epidermidis in a reconstructed human epidermis model. qRT-PCR array analysis of 64 genes expressed by keratinocytes in the response to bacterial infection revealed a potential link between the action of SP on Staphylococci and skin physiopathology. qRT-PCR and direct assay of cathelicidin and human β-defensin 2 secretion also provided that demonstration that the action of SP on bacteria is independent of antimicrobial peptide expression by keratinocytes. Considering an effect of SP on S. aureus and S. epidermidis, we observed that SP increases the adhesion potential of both bacteria on keratinocytes. However, SP modulates the virulence of S. aureus and S. epidermidis through different mechanisms. The response of S. aureus is associated with an increase in Staphylococcal Enterotoxin C2 (SEC2) production and a reduction of exolipase processing whereas in S. epidermidis the effect of SP appears mediated by a rise in biofilm formation activity. The Thermo unstable ribosomal Elongation factor Ef-Tu was identified as the SP-interacting protein in S. aureus and S. epidermidis. SP appears as an inter-kingdom communication factor involved in the regulation of bacterial virulence and essential for skin microflora homeostasis.
Collapse
Affiliation(s)
- Awa N'Diaye
- Laboratory of Microbiology Signals and Microenvironnement LMSM, EA 4312, Normandie Université, Université de Rouen Evreux, France
| | - Lily Mijouin
- Laboratory of Microbiology Signals and Microenvironnement LMSM, EA 4312, Normandie Université, Université de Rouen Evreux, France
| | - Mélanie Hillion
- Laboratory of Microbiology Signals and Microenvironnement LMSM, EA 4312, Normandie Université, Université de Rouen Evreux, France
| | - Suraya Diaz
- Department of Biosciences, University of Exeter Exeter, UK
| | - Yoan Konto-Ghiorghi
- Laboratory of Microbiology Signals and Microenvironnement LMSM, EA 4312, Normandie Université, Université de Rouen Evreux, France
| | - Giuseppe Percoco
- GlycoMev EA 4358, Normandie Université, Université de RouenMont-Saint-Aignan, France; Bio-EC LaboratoryLongjumeau, France
| | - Sylvie Chevalier
- Laboratory of Microbiology Signals and Microenvironnement LMSM, EA 4312, Normandie Université, Université de Rouen Evreux, France
| | - Luc Lefeuvre
- Dermatologic Laboratories Uriage Neuilly-Sur-Seine, France
| | | | - Olivier Lesouhaitier
- Laboratory of Microbiology Signals and Microenvironnement LMSM, EA 4312, Normandie Université, Université de Rouen Evreux, France
| | - Marc G J Feuilloley
- Laboratory of Microbiology Signals and Microenvironnement LMSM, EA 4312, Normandie Université, Université de Rouen Evreux, France
| |
Collapse
|
34
|
Anti-Inflammatory and Antimicrobial Effects of Estradiol in Bovine Mammary Epithelial Cells during Staphylococcus aureus Internalization. Mediators Inflamm 2016; 2016:6120509. [PMID: 27034592 PMCID: PMC4791512 DOI: 10.1155/2016/6120509] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2015] [Accepted: 02/07/2016] [Indexed: 12/03/2022] Open
Abstract
17β-Estradiol (E2), the predominant sexual hormone in females, is associated with the modulation of the innate immune response (IIR), and changes in its levels at parturition are related to intramammary infections, such as mastitis. In bovine mammary epithelial cells (bMECs), E2 regulates differentiation and proliferation, but its immunomodulatory functions have not been explored. Staphylococcus aureus is the predominant pathogen causing mastitis, which can persist intracellularly in bMECs. The aim of this work was to analyze whether E2 modulates the IIR of bMECs during S. aureus internalization. bMECs treated with E2 (50 pg/mL, 24 h) reduced bacteria internalization (~50%). The host receptors α5β1 and TLR2 do not participate in this reduction. However, E2 activates ERα and modulates the IIR reducing the S. aureus induced-mRNA expression of TNF-α (~50%) and IL-1β (90%). E2 also decreased the secretion of these cytokines as well as IL-6 production; however, in infected bMECs, E2 induced the secretion of IL-1β. Furthermore, E2 upregulates the expression of the antimicrobial peptides DEFB1, BNBD5, and psoriasin S100A7 (~5-, 3-, and 6-fold, resp.). In addition, E2 induced the production of antimicrobial compounds in bMEC culture medium, which, together with the modulation of the IIR, could be related to the reduction of S. aureus internalization.
Collapse
|
35
|
Herpes Simplex Virus (HSV) Modulation of Staphylococcus aureus and Candida albicans Initiation of HeLa 299 Cell-Associated Biofilm. Curr Microbiol 2016; 72:529-37. [PMID: 26758707 PMCID: PMC4828481 DOI: 10.1007/s00284-015-0975-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Accepted: 11/22/2015] [Indexed: 01/30/2023]
Abstract
Although herpes simplex virus type-1 (HSV-1), and type-2 (HSV-2), Staphylococcus aureus and Candida albicans co-habit the oral and genital mucosa, their interaction is poorly understood. We determined the effect HSV has on bacterial and/or fungal adherence, the initial step in biofilm formation. HeLa229 cells were infected with HSV-1 (KOS) gL86 or HSV-2 (KOS) 333gJ− at a multiplicity of infection (MOI) of 50 and 10. S. aureus (ATCC 25923) and/or C. albicans (yeast forms or germ tube forms) were co-incubated for 30 min (37 °C; 5 % CO2; 5:1 organism: HeLa cell ratio; n = 16) with virus-infected HeLa cells or uninfected HeLa cell controls. Post-incubation, the monolayers were washed (3x; PBS), lysed (RIPA), and the lysate plated onto Fungisel and/or mannitol salts agar for standard colony count. The level of HeLa-associated S. aureus was significantly decreased (P < 0.05) for both HSV-1- and HSV-2-infected cells, as compared to virus-free HeLa cell controls (38 and 59 % of control, respectively). In contrast, HSV-1 and HSV-2 significantly (P < 0.05) enhanced HeLa cell association of C. albicans yeast forms and germ tube approximately two-fold, respectively. The effect of S. aureus on germ tube and yeast form adherence to HSV-1- and HSV-2-infected cells was specific for the Candida phenotype tested. Our study suggests that HSV, while antagonist towards S. aureus adherence enhances Candida adherence. Furthermore, the combination of the three pathogens results in S. aureus adherence that is either unaffected, or partially restored depending on both the herpes viral species and the fungal phenotype present.
Collapse
|