1
|
Pérez Vázquez K, Tau J, Leal Denis MF, Fader CM, Ostuni MA, Schwarzbaum PJ, Herlax V. Alpha hemolysin of Escherichia coli induces a necrotic-like procoagulant state in platelets. Biochimie 2024; 227:1-14. [PMID: 38857695 DOI: 10.1016/j.biochi.2024.06.001] [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: 01/22/2024] [Revised: 06/05/2024] [Accepted: 06/07/2024] [Indexed: 06/12/2024]
Abstract
Uropathogenic strains of E. coli (UPEC) is a leading cause of sepsis, deploying multiple virulence factors to evade host immune responses. Notably, alpha-hemolysin (HlyA) produced by UPEC is implicated in septic symptoms associated with bacteremia, correlating with thrombocytopenia, a critical indicator of organ dysfunction and a predictor of poorer patient prognosis. This study meticulously explores the impact of sublytic concentrations of HlyA on platelets. Findings reveal that HlyA triggers an increase in intracellular calcium, activating calpain and exposing phosphatidylserine to the cell surface, as validated by flow cytometric experiments. Electron microscopy reveals a distinctive balloon-like shape in HlyA-treated platelets, indicative of a procoagulant state. The toxin induces the release of procoagulant extracellular vesicles and the secretion of alpha and dense granules. Overall, the results point to HlyA inducing a necrotic-like procoagulant state in platelets. The effects of sublytic concentrations of HlyA on both erythrocytes and platelets could have a potential impact on capillary microcirculation. Targeting HlyA emerges as a viable therapeutic strategy to mitigate the adverse effects of UPEC infections, especially in South American countries where these infections are endemic, underscoring its significance as a potential therapeutic target.
Collapse
Affiliation(s)
- Kenia Pérez Vázquez
- Instituto de Investigaciones Bioquímicas de La Plata (INIBIOLP), CCT- La Plata, CONICET. Facultad de Ciencias Médicas. Universidad Nacional de La Plata, Argentina
| | - Julia Tau
- Instituto de Investigaciones Bioquímicas de La Plata (INIBIOLP), CCT- La Plata, CONICET. Facultad de Ciencias Médicas. Universidad Nacional de La Plata, Argentina
| | - M Florencia Leal Denis
- Instituto de Química y Fisicoquímica Biológicas (IQUIFIB) "Prof. Alejandro C. Paladini", Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Claudio M Fader
- Laboratorio de Biología Celular y Molecular, Instituto de Histología y Embriología, (IHEM), Universidad Nacional de Cuyo, CONICET, Mendoza, Argentina; Facultad de Odontología, Universidad Nacional de Cuyo, Mendoza, Argentina
| | - Mariano A Ostuni
- Université Paris Cité and Université des Antilles, INSERM, BIGR, F-75015, Paris, France
| | - Pablo J Schwarzbaum
- Instituto de Química y Fisicoquímica Biológicas (IQUIFIB) "Prof. Alejandro C. Paladini", Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Vanesa Herlax
- Instituto de Investigaciones Bioquímicas de La Plata (INIBIOLP), CCT- La Plata, CONICET. Facultad de Ciencias Médicas. Universidad Nacional de La Plata, Argentina.
| |
Collapse
|
2
|
Xing Y, Clark JR, Chang JD, Zulk JJ, Chirman DM, Piedra FA, Vaughan EE, Hernandez Santos HJ, Patras KA, Maresso AW. Progress toward a vaccine for extraintestinal pathogenic E. coli (ExPEC) II: efficacy of a toxin-autotransporter dual antigen approach. Infect Immun 2024; 92:e0044023. [PMID: 38591882 PMCID: PMC11075464 DOI: 10.1128/iai.00440-23] [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: 11/01/2023] [Accepted: 03/18/2024] [Indexed: 04/10/2024] Open
Abstract
Extraintestinal pathogenic Escherichia coli (ExPEC) is a leading cause of worldwide morbidity and mortality, the top cause of antimicrobial-resistant (AMR) infections, and the most frequent cause of life-threatening sepsis and urinary tract infections (UTI) in adults. The development of an effective and universal vaccine is complicated by this pathogen's pan-genome, its ability to mix and match virulence factors and AMR genes via horizontal gene transfer, an inability to decipher commensal from pathogens, and its intimate association and co-evolution with mammals. Using a pan virulome analysis of >20,000 sequenced E. coli strains, we identified the secreted cytolysin α-hemolysin (HlyA) as a high priority target for vaccine exploration studies. We demonstrate that a catalytically inactive pure form of HlyA, expressed in an autologous host using its own secretion system, is highly immunogenic in a murine host, protects against several forms of ExPEC infection (including lethal bacteremia), and significantly lowers bacterial burdens in multiple organ systems. Interestingly, the combination of a previously reported autotransporter (SinH) with HlyA was notably effective, inducing near complete protection against lethal challenge, including commonly used infection strains ST73 (CFT073) and ST95 (UTI89), as well as a mixture of 10 of the most highly virulent sequence types and strains from our clinical collection. Both HlyA and HlyA-SinH combinations also afforded some protection against UTI89 colonization in a murine UTI model. These findings suggest recombinant, inactive hemolysin and/or its combination with SinH warrant investigation in the development of an E. coli vaccine against invasive disease.
Collapse
Affiliation(s)
- Yikun Xing
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, USA
- TAILOR Labs, Vaccine Development Group, Baylor College of Medicine, Houston, Texas, USA
| | - Justin R. Clark
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, USA
- TAILOR Labs, Vaccine Development Group, Baylor College of Medicine, Houston, Texas, USA
| | - James D. Chang
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, USA
- TAILOR Labs, Vaccine Development Group, Baylor College of Medicine, Houston, Texas, USA
| | - Jacob J. Zulk
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, USA
- TAILOR Labs, Vaccine Development Group, Baylor College of Medicine, Houston, Texas, USA
| | - Dylan M. Chirman
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, USA
- TAILOR Labs, Vaccine Development Group, Baylor College of Medicine, Houston, Texas, USA
| | - Felipe-Andres Piedra
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, USA
| | - Ellen E. Vaughan
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, USA
| | - Haroldo J. Hernandez Santos
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, USA
- TAILOR Labs, Vaccine Development Group, Baylor College of Medicine, Houston, Texas, USA
| | - Kathryn A. Patras
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, USA
- Alkek Center for Metagenomics and Microbiome Research, Baylor College of Medicine, Houston, Texas, USA
| | - Anthony W. Maresso
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, USA
- TAILOR Labs, Vaccine Development Group, Baylor College of Medicine, Houston, Texas, USA
| |
Collapse
|
3
|
Lewis AJ, Richards AC, Mendez AA, Dhakal BK, Jones TA, Sundsbak JL, Eto DS, Mulvey MA. Plant Phenolics Inhibit Focal Adhesion Kinase and Suppress Host Cell Invasion by Uropathogenic Escherichia coli. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.23.568486. [PMID: 38045282 PMCID: PMC10690256 DOI: 10.1101/2023.11.23.568486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/05/2023]
Abstract
Traditional folk treatments for the prevention and management of urinary tract infections (UTIs) and other infectious diseases often include plants and plant extracts that are rich in phenolic and polyphenolic compounds. These have been ascribed a variety of activities, including inhibition of bacterial interactions with host cells. Here we tested a panel of four well-studied phenolic compounds - caffeic acid phenethyl ester (CAPE), resveratrol, catechin, and epigallocatechin gallate - for effects on host cell adherence and invasion by uropathogenic Escherichia coli (UPEC). These bacteria, which are the leading cause of UTIs, can bind and subsequently invade bladder epithelial cells via an actin-dependent process. Intracellular UPEC reservoirs within the bladder are often protected from antibiotics and host defenses, and likely contribute to the development of chronic and recurrent infections. Using cell culture-based assays, we found that only resveratrol had a notable negative effect on UPEC adherence to bladder cells. However, both CAPE and resveratrol significantly inhibited UPEC entry into the host cells, coordinate with attenuated phosphorylation of the host actin regulator Focal Adhesion Kinase (FAK, or PTK2) and marked increases in the numbers of focal adhesion structures. We further show that the intravesical delivery of resveratrol inhibits UPEC infiltration of the bladder mucosa in a murine UTI model, and that resveratrol and CAPE can disrupt the ability of other invasive pathogens to enter host cells. Together, these results highlight the therapeutic potential of molecules like CAPE and resveratrol, which could be used to augment antibiotic treatments by restricting pathogen access to protective intracellular niches.
Collapse
Affiliation(s)
- Adam J. Lewis
- Division of Microbiology and Immunology, Department of Pathology, University of Utah, Salt Lake City, UT 84112, USA
| | - Amanda C. Richards
- Division of Microbiology and Immunology, Department of Pathology, University of Utah, Salt Lake City, UT 84112, USA
- School of Biological Sciences, 257 S 1400 E, University of Utah, Salt Lake City, UT 84112, USA; Henry Eyring Center for Cell & Genome Science, 1390 Presidents Circle, University of Utah, Salt Lake City, UT 84112, USA
| | - Alejandra A. Mendez
- Division of Microbiology and Immunology, Department of Pathology, University of Utah, Salt Lake City, UT 84112, USA
- School of Biological Sciences, 257 S 1400 E, University of Utah, Salt Lake City, UT 84112, USA; Henry Eyring Center for Cell & Genome Science, 1390 Presidents Circle, University of Utah, Salt Lake City, UT 84112, USA
| | - Bijaya K. Dhakal
- Division of Microbiology and Immunology, Department of Pathology, University of Utah, Salt Lake City, UT 84112, USA
| | - Tiffani A. Jones
- Division of Microbiology and Immunology, Department of Pathology, University of Utah, Salt Lake City, UT 84112, USA
| | - Jamie L. Sundsbak
- Division of Microbiology and Immunology, Department of Pathology, University of Utah, Salt Lake City, UT 84112, USA
| | - Danelle S. Eto
- Division of Microbiology and Immunology, Department of Pathology, University of Utah, Salt Lake City, UT 84112, USA
| | - Matthew A. Mulvey
- Division of Microbiology and Immunology, Department of Pathology, University of Utah, Salt Lake City, UT 84112, USA
- School of Biological Sciences, 257 S 1400 E, University of Utah, Salt Lake City, UT 84112, USA; Henry Eyring Center for Cell & Genome Science, 1390 Presidents Circle, University of Utah, Salt Lake City, UT 84112, USA
| |
Collapse
|
4
|
Gioia MD, Poli V, Tan PJ, Spreafico R, Chu A, Cuenca AG, Gordts PL, Pandolfi L, Meloni F, Witztum JL, Chou J, Springstead JR, Zanoni I. Host-derived oxidized phospholipids initiate effector-triggered immunity fostering lethality upon microbial encounter. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.21.568047. [PMID: 38045410 PMCID: PMC10690175 DOI: 10.1101/2023.11.21.568047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/05/2023]
Abstract
Macrophages detect invading microorganisms via pattern recognition receptors that recognize pathogen-associated molecular patterns, or via sensing the activity of virulence factors that initiates effector-triggered immunity (ETI). Tissue damage that follows pathogen encounter leads to the release of host-derived factors that participate to inflammation. How these self-derived molecules are sensed by macrophages and their impact on immunity remain poorly understood. Here we demonstrate that, in mice and humans, host-derived oxidized phospholipids (oxPLs) are formed upon microbial encounter. oxPL blockade restricts inflammation and prevents the death of the host, without affecting pathogen burden. Mechanistically, oxPLs bind and inhibit AKT, a master regulator of immunity and metabolism. AKT inhibition potentiates the methionine cycle, and epigenetically dampens Il10, a pluripotent anti-inflammatory cytokine. Overall, we found that host-derived inflammatory cues act as "self" virulence factors that initiate ETI and that their activity can be targeted to protect the host against excessive inflammation upon microbial encounter.
Collapse
Affiliation(s)
- Marco Di Gioia
- Harvard Medical School and Boston Children's Hospital, Division of Immunology and Division of Gastroenterology, MA 02115, USA
| | - Valentina Poli
- Harvard Medical School and Boston Children's Hospital, Division of Immunology and Division of Gastroenterology, MA 02115, USA
| | - Piao J Tan
- Department of Chemical and Paper Engineering, Western Michigan University, Kalamazoo, MI, USA
| | - Roberto Spreafico
- Institute for Quantitative and Computational Biosciences, University of California, Los Angeles, Los Angeles, CA, USA
| | - Anne Chu
- Harvard Medical School and Boston Children's Hospital, Division of Immunology and Division of Gastroenterology, MA 02115, USA
| | - Alex G Cuenca
- Department of Surgery, Boston Children's Hospital, Boston, MA 02115, USA; Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital, MA 02114, USA
| | - Philip Lsm Gordts
- Division of Endocrinology and Metabolism, Department of Medicine, University of California, San Diego, La Jolla, CA, USA; Glycobiology Research and Training Center, University of California, San Diego, La Jolla, California, USA
| | - Laura Pandolfi
- Respiratory Disease Unit IRCCS San Matteo Hospital Foundation, and Department of Internal Medicine and Pharmacology, University of Pavia, Pavia, 27100, Italy Pavia, 27100, Italy
| | - Federica Meloni
- Respiratory Disease Unit IRCCS San Matteo Hospital Foundation, and Department of Internal Medicine and Pharmacology, University of Pavia, Pavia, 27100, Italy Pavia, 27100, Italy
| | - Joseph L Witztum
- Division of Endocrinology and Metabolism, Department of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Janet Chou
- Harvard Medical School and Boston Children's Hospital, Division of Immunology and Division of Gastroenterology, MA 02115, USA
| | - James R Springstead
- Department of Chemical and Paper Engineering, Western Michigan University, Kalamazoo, MI, USA
| | - Ivan Zanoni
- Harvard Medical School and Boston Children's Hospital, Division of Immunology and Division of Gastroenterology, MA 02115, USA
| |
Collapse
|
5
|
Dernovics Á, Seprényi G, Rázga Z, Ayaydin F, Veréb Z, Megyeri K. Phenol-Soluble Modulin α3 Stimulates Autophagy in HaCaT Keratinocytes. Biomedicines 2023; 11:3018. [PMID: 38002017 PMCID: PMC10669503 DOI: 10.3390/biomedicines11113018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2023] [Revised: 10/30/2023] [Accepted: 11/07/2023] [Indexed: 11/26/2023] Open
Abstract
BACKGROUND Phenol-soluble modulins (PSMs) are pore-forming toxins (PFTs) produced by staphylococci. PSMs exert diverse cellular effects, including lytic, pro-apoptotic, pro-inflammatory and antimicrobial actions. Since the effects of PSMs on autophagy have not yet been reported, we evaluated the autophagic activity in HaCaT keratinocytes treated with recombinant PSMα3. METHODS The autophagic flux and levels of autophagic marker proteins were determined using Western blot analysis. Subcellular localization of LC3B and Beclin-1 was investigated using an indirect immunofluorescence assay. The ultrastructural features of control and PSMα3-treated cells were evaluated via transmission electron microscopy. Cytoplasmic acidification was measured via acridine orange staining. Phosphorylation levels of protein kinases, implicated in autophagy regulation, were studied using a phospho-kinase array and Western blot analysis. RESULTS PSMα3 facilitated the intracellular redistribution of LC3B, increased the average number of autophagosomes per cell, promoted the development of acidic vesicular organelles, elevated the levels of LC3B-II, stimulated autophagic flux and triggered a significant decrease in the net autophagic turnover rate. PSMα3 induced the accumulation of autophagosomes/autolysosomes, amphisomes and multilamellar bodies at the 0.5, 6 and 24 h time points, respectively. The phospho-Akt1/2/3 (T308 and S473), and phospho-mTOR (S2448) levels were decreased, whereas the phospho-Erk1/2 (T202/Y204 and T185/Y187) level was increased in PSMα3-treated cells. CONCLUSIONS In HaCaT keratinocytes, PSMα3 stimulates autophagy. The increased autophagic activity elicited by sub-lytic PSM concentrations might be an integral part of the cellular defense mechanisms protecting skin homeostasis.
Collapse
Affiliation(s)
- Áron Dernovics
- Department of Medical Microbiology, Albert Szent-Györgyi Medical School, University of Szeged, Dóm tér 10., H-6720 Szeged, Hungary;
| | - György Seprényi
- Department of Anatomy, Histology and Embryology, Albert Szent-Györgyi Medical School, University of Szeged, Kossuth L. sgt. 40., H-6724 Szeged, Hungary;
| | - Zsolt Rázga
- Department of Pathology, University of Szeged, Állomás u. 2, H-6720 Szeged, Hungary;
| | - Ferhan Ayaydin
- Hungarian Centre of Excellence for Molecular Medicine (HCEMM) Nonprofit Ltd., Római krt. 21., H-6723 Szeged, Hungary;
- Laboratory of Cellular Imaging, Biological Research Centre, Eötvös Loránd Research Network, Temesvári krt. 62., H-6726 Szeged, Hungary
| | - Zoltán Veréb
- Regenerative Medicine and Cellular Pharmacology Laboratory, Department of Dermatology and Allergology, University of Szeged, Korányi Fasor 6, H-6720 Szeged, Hungary;
- Biobank, University of Szeged, H-6720 Szeged, Hungary
- Interdisciplinary Research Development and Innovation Center of Excellence, University of Szeged, H-6720 Szeged, Hungary
| | - Klára Megyeri
- Department of Medical Microbiology, Albert Szent-Györgyi Medical School, University of Szeged, Dóm tér 10., H-6720 Szeged, Hungary;
| |
Collapse
|
6
|
Whelan S, Lucey B, Finn K. Uropathogenic Escherichia coli (UPEC)-Associated Urinary Tract Infections: The Molecular Basis for Challenges to Effective Treatment. Microorganisms 2023; 11:2169. [PMID: 37764013 PMCID: PMC10537683 DOI: 10.3390/microorganisms11092169] [Citation(s) in RCA: 43] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 08/23/2023] [Accepted: 08/25/2023] [Indexed: 09/29/2023] Open
Abstract
Urinary tract infections (UTIs) are among the most common bacterial infections, especially among women and older adults, leading to a significant global healthcare cost burden. Uropathogenic Escherichia coli (UPEC) are the most common cause and accounts for the majority of community-acquired UTIs. Infection by UPEC can cause discomfort, polyuria, and fever. More serious clinical consequences can result in urosepsis, kidney damage, and death. UPEC is a highly adaptive pathogen which presents significant treatment challenges rooted in a complex interplay of molecular factors that allow UPEC to evade host defences, persist within the urinary tract, and resist antibiotic therapy. This review discusses these factors, which include the key genes responsible for adhesion, toxin production, and iron acquisition. Additionally, it addresses antibiotic resistance mechanisms, including chromosomal gene mutations, antibiotic deactivating enzymes, drug efflux, and the role of mobile genetic elements in their dissemination. Furthermore, we provide a forward-looking analysis of emerging alternative therapies, such as phage therapy, nano-formulations, and interventions based on nanomaterials, as well as vaccines and strategies for immunomodulation. This review underscores the continued need for research into the molecular basis of pathogenesis and antimicrobial resistance in the treatment of UPEC, as well as the need for clinically guided treatment of UTIs, particularly in light of the rapid spread of multidrug resistance.
Collapse
Affiliation(s)
- Shane Whelan
- Department of Biological Sciences, Munster Technological University, Bishopstown, T12 P928 Cork, Ireland;
| | - Brigid Lucey
- Department of Biological Sciences, Munster Technological University, Bishopstown, T12 P928 Cork, Ireland;
| | - Karen Finn
- Department of Analytical, Biopharmaceutical and Medical Sciences, Atlantic Technological University Galway City, Dublin Road, H91 T8NW Galway, Ireland
| |
Collapse
|
7
|
Shetty SV, Mazzucco MR, Winokur P, Haigh SV, Rumah KR, Fischetti VA, Vartanian T, Linden JR. Clostridium perfringens Epsilon Toxin Binds to and Kills Primary Human Lymphocytes. Toxins (Basel) 2023; 15:423. [PMID: 37505692 PMCID: PMC10467094 DOI: 10.3390/toxins15070423] [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: 03/31/2023] [Revised: 05/30/2023] [Accepted: 06/07/2023] [Indexed: 07/29/2023] Open
Abstract
Clostridium perfringens epsilon toxin (ETX) is the third most lethal bacterial toxin and has been suggested to be an environmental trigger of multiple sclerosis, an immune-mediated disease of the human central nervous system. However, ETX cytotoxicity on primary human cells has not been investigated. In this article, we demonstrate that ETX preferentially binds to and kills human lymphocytes expressing increased levels of the myelin and lymphocyte protein MAL. Using flow cytometry, ETX binding was determined to be time and dose dependent and was highest for CD4+ cells, followed by CD8+ and then CD19+ cells. Similar results were seen with ETX-induced cytotoxicity. To determine if ETX preference for CD4+ cells was related to MAL expression, MAL gene expression was determined by RT-qPCR. CD4+ cells had the highest amount of Mal gene expression followed by CD8+ and CD19+ cells. These data indicate that primary human cells are susceptible to ETX and support the hypothesis that MAL is a main receptor for ETX. Interestingly, ETX bindings to human lymphocytes suggest that ETX may influence immune response in multiple sclerosis.
Collapse
Affiliation(s)
- Samantha V. Shetty
- Feil Family Brain and Mind Research Institute, Weill Cornell Medical College, New York, NY 10065, USA (T.V.)
| | - Michael R. Mazzucco
- Feil Family Brain and Mind Research Institute, Weill Cornell Medical College, New York, NY 10065, USA (T.V.)
| | - Paige Winokur
- Harold and Margaret Milliken Hatch Laboratory of Neuro-Endocrinology Rockefeller University, New York, NY 10065, USA
| | - Sylvia V. Haigh
- Feil Family Brain and Mind Research Institute, Weill Cornell Medical College, New York, NY 10065, USA (T.V.)
| | - Kareem Rashid Rumah
- Laboratory of Bacterial Pathogenesis and Immunology, Rockefeller University, New York, NY 10065, USA
| | - Vincent A. Fischetti
- Laboratory of Bacterial Pathogenesis and Immunology, Rockefeller University, New York, NY 10065, USA
| | - Timothy Vartanian
- Feil Family Brain and Mind Research Institute, Weill Cornell Medical College, New York, NY 10065, USA (T.V.)
| | - Jennifer R. Linden
- Feil Family Brain and Mind Research Institute, Weill Cornell Medical College, New York, NY 10065, USA (T.V.)
| |
Collapse
|
8
|
Li MY, Li Y, Wang LL, Xu F, Guo XY, Zhang J, Lv Y, Wang PP, Wang SQ, Min JG, Zou X, Cai SQ. Chemical profiling of Sanjin tablets and exploration of their effective substances and mechanism in the treatment of urinary tract infections. Front Chem 2023; 11:1179956. [PMID: 37408563 PMCID: PMC10318440 DOI: 10.3389/fchem.2023.1179956] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Accepted: 06/07/2023] [Indexed: 07/07/2023] Open
Abstract
Introduction: Sanjin tablets (SJT) are a well-known Chinese patent drug that have been used to treat urinary tract infections (UTIs) for the last 40 years. The drug consists of five herbs, but only 32 compounds have been identified, which hinders the clarification of its effective substances and mechanism. Methods: The chemical constituents of SJT and their effective substances and functional mechanism involved in the treatment of UTIs were investigated by using high performance liquid chromatography-electrospray ionization-ion trap-time of flight-mass spectrometry (HPLC-ESI-IT-TOF-MSn), network pharmacology, and molecular docking. Results: A total of 196 compounds of SJT (SJT-MS) were identified, and 44 of them were unequivocally identified by comparison with the reference compounds. Among 196 compounds, 13 were potential new compounds and 183 were known compounds. Among the 183 known compounds, 169 were newly discovered constituents of SJT, and 93 compounds were not reported in the five constituent herbs. Through the network pharmacology method, 119 targets related to UTIs of 183 known compounds were predicted, and 20 core targets were screened out. Based on the "compound-target" relationship analysis, 94 compounds were found to act on the 20 core targets and were therefore regarded as potential effective compounds. According to the literature, 27 of the 183 known compounds were found to possess antimicrobial and anti-inflammatory activities and were verified as effective substances, of which 20 were first discovered in SJT. Twelve of the 27 effective substances overlapped with the 94 potential effective compounds and were determined as key effective substances of SJT. The molecular docking results showed that the 12 key effective substances and 10 selected targets of the core targets have good affinity for each other. Discussion: These results provide a solid foundation for understanding the effective substances and mechanism of SJT.
Collapse
Affiliation(s)
- Meng-Yuan Li
- School of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, China
- School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Yang Li
- School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Li-Li Wang
- School of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, China
| | - Feng Xu
- School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Xu-Yan Guo
- School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Jing Zhang
- School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Yang Lv
- School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Peng-Pu Wang
- School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Shun-Qi Wang
- School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Jian-Guo Min
- Guilin Sanjin Pharmaceutical Company Limited, Guilin, China
| | - Xun Zou
- Guilin Sanjin Pharmaceutical Company Limited, Guilin, China
| | - Shao-Qing Cai
- School of Pharmaceutical Sciences, Peking University, Beijing, China
| |
Collapse
|
9
|
Guo C, Zhao M, Sui X, Balsara Z, Zhai S, Ahdoot M, Zhang Y, Lam CM, Zhu P, Li X. Targeting the PRC2-dependent epigenetic program alleviates urinary tract infections. iScience 2023; 26:106925. [PMID: 37332606 PMCID: PMC10272480 DOI: 10.1016/j.isci.2023.106925] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 04/08/2023] [Accepted: 05/15/2023] [Indexed: 06/20/2023] Open
Abstract
Urinary tract infection (UTI) is a pervasive health problem worldwide. Patients with a history of UTIs suffer increased risk of recurrent infections, a major risk of antibiotic resistance. Here, we show that bladder infections induce expression of Ezh2 in bladder urothelial cells. Ezh2 is the methyltransferase of polycomb repressor complex 2 (PRC2)-a potent epigenetic regulator. Urothelium-specific inactivation of PRC2 results in reduced urine bacterial burden, muted inflammatory response, and decreased activity of the NF-κB signaling pathway. PRC2 inactivation also facilitates proper regeneration after urothelial damage from UTIs, by attenuating basal cell hyperplasia and increasing urothelial differentiation. In addition, treatment with Ezh2-specific small-molecule inhibitors improves outcomes of the chronic and severe bladder infections in mice. These findings collectively suggest that the PRC2-dependent epigenetic reprograming controls the amplitude of inflammation and severity of UTIs and that Ezh2 inhibitors may be a viable non-antibiotic strategy to manage chronic and severe UTIs.
Collapse
Affiliation(s)
- Chunming Guo
- Samuel Oschin Comprehensive Cancer Institute, Department of Medicine, Department of Biomedical Sciences, Cedars-Sinai Medical Center, 8700 Beverly Blvd, Davis 3089, Los Angeles, CA 90048, USA
- Departments of Urology and Surgery, Boston Children’s Hospital, Harvard Medical School, 300 Longwood Avenue, Boston, MA 02115, USA
| | - Mingyi Zhao
- Departments of Urology and Surgery, Boston Children’s Hospital, Harvard Medical School, 300 Longwood Avenue, Boston, MA 02115, USA
- Guangdong Cardiovascular Institute, Guangdong Provincial Key Laboratory of Pathogenesis, Guangzhou Key Laboratory of Cardiac Pathogenesis and Prevention, Guangdong Provincial People's Hospital, Southern Medical University, Guangzhou, Guangdong 510100, China
| | - Xinbing Sui
- Departments of Urology and Surgery, Boston Children’s Hospital, Harvard Medical School, 300 Longwood Avenue, Boston, MA 02115, USA
| | - Zarine Balsara
- Departments of Urology and Surgery, Boston Children’s Hospital, Harvard Medical School, 300 Longwood Avenue, Boston, MA 02115, USA
| | - Songhui Zhai
- Departments of Urology and Surgery, Boston Children’s Hospital, Harvard Medical School, 300 Longwood Avenue, Boston, MA 02115, USA
| | - Michael Ahdoot
- Department of Surgery, Cedars-Sinai Medical Center, 8700 Beverly Blvd, Davis 3089, Los Angeles, CA 90048, USA
| | - Yingsheng Zhang
- Samuel Oschin Comprehensive Cancer Institute, Department of Medicine, Department of Biomedical Sciences, Cedars-Sinai Medical Center, 8700 Beverly Blvd, Davis 3089, Los Angeles, CA 90048, USA
| | - Christa M. Lam
- Samuel Oschin Comprehensive Cancer Institute, Department of Medicine, Department of Biomedical Sciences, Cedars-Sinai Medical Center, 8700 Beverly Blvd, Davis 3089, Los Angeles, CA 90048, USA
- Departments of Urology and Surgery, Boston Children’s Hospital, Harvard Medical School, 300 Longwood Avenue, Boston, MA 02115, USA
| | - Ping Zhu
- Guangdong Cardiovascular Institute, Guangdong Provincial Key Laboratory of Pathogenesis, Guangzhou Key Laboratory of Cardiac Pathogenesis and Prevention, Guangdong Provincial People's Hospital, Southern Medical University, Guangzhou, Guangdong 510100, China
| | - Xue Li
- Samuel Oschin Comprehensive Cancer Institute, Department of Medicine, Department of Biomedical Sciences, Cedars-Sinai Medical Center, 8700 Beverly Blvd, Davis 3089, Los Angeles, CA 90048, USA
- Departments of Urology and Surgery, Boston Children’s Hospital, Harvard Medical School, 300 Longwood Avenue, Boston, MA 02115, USA
| |
Collapse
|
10
|
Kumar M, Shelly A, Dahiya P, Ray A, Mazumder S. Aeromonas hydrophila inhibits autophagy triggering cytosolic translocation of mtDNA which activates the pro-apoptotic caspase-1/IL-1β-nitric oxide axis in headkidney macrophages. Virulence 2022; 13:60-76. [PMID: 34967692 PMCID: PMC9794009 DOI: 10.1080/21505594.2021.2018767] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
The molecular mechanisms underlying Aeromonas hydrophila-pathogenesis are not well understood. Using head kidney macrophages (HKM) of Clarias gariepinus, we previously reported the role of ER-stress in A. hydrophila-induced pathogenesis. Here, we report that PI3K/PLC-induced cytosolic-Ca2+ imbalance induces the expression of pro-apoptotic ER-stress marker, CHOP in A. hydrophila-infected HKM. CHOP promotes HKM apoptosis by inhibiting AKT activation and enhancing JNK signaling. Elevated mitochondrial ROS (mtROS) was recorded which declined significantly by ameliorating ER-stress and in the presence of ER-Ca2+ release modulators (2-APB and dantrolene) and mitochondrial-Ca2+ uptake inhibitor, Ru360, together suggesting the role of ER-mitochondrial Ca2+ dynamics in mtROS generation. Inhibiting mtROS production reduced HKM death implicating the pro-apoptotic role of mtROS in A. hydrophila-pathogenesis. The expression of autophagic proteins (LC3B, beclin-1, and atg 5) was suppressed in the infected HKM. Our results with autophagy-inducer rapamycin demonstrated that impaired autophagy favored the cytosolic accumulation of mitochondrial DNA (mtDNA) and the process depended on mtROS levels. Enhanced caspase-1 activity and IL-1β production was detected and transfection studies coupled with pharmacological inhibitors implicated mtROS/mtDNA axis to be crucial for activating the caspase-1/IL-1β cascade in infected HKM. RNAi studies further suggested the involvement of IL-1β in generating pro-apoptotic NO in A. hydrophila-infected HKM. Our study suggests a novel role of ER-mitochondria cross-talk in regulating A. hydrophila pathogenesis. Based on our observations, we conclude that A. hydrophila induces ER-stress and inhibits mitophagy resulting in mitochondrial dysfunction which leads to mtROS production and translocation of mtDNA into cytosol triggering the activation of caspase-1/IL-1β-mediated NO production, culminating in HKM apoptosis.
Collapse
Affiliation(s)
- Manmohan Kumar
- Immunobiology Laboratory, Department of Zoology, University of Delhi, Delhi, India
| | - Asha Shelly
- Immunobiology Laboratory, Department of Zoology, University of Delhi, Delhi, India
| | - Priyanka Dahiya
- Immunobiology Laboratory, Department of Zoology, University of Delhi, Delhi, India
| | - Atish Ray
- Immunobiology Laboratory, Department of Zoology, University of Delhi, Delhi, India
| | - Shibnath Mazumder
- Immunobiology Laboratory, Department of Zoology, University of Delhi, Delhi, India,Faculty of Life Sciences and Biotechnology, South Asian University, New Delhi, India,CONTACT Shibnath Mazumder Faculty of Life Sciences and Biotechnology
| |
Collapse
|
11
|
Zhang S, Wang J, Fan Y, Meng W, Qian C, Liu P, Wei Y, Yuan C, Du Y, Yin Z. YciR, a Specific 3′-Phosphodiesterase, Plays a Role in the Pathogenesis of Uropathogenic Escherichia coli CFT073. Front Microbiol 2022; 13:910906. [PMID: 35923408 PMCID: PMC9339999 DOI: 10.3389/fmicb.2022.910906] [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: 04/01/2022] [Accepted: 06/21/2022] [Indexed: 11/13/2022] Open
Abstract
Urinary tract infections (UTIs), with the characteristics of recurrence and resistance to antibiotics due to misuse, remain a common health and economic issue for patients. Uropathogenic Escherichia coli (UPEC), which is capable of evading the immune response by forming intracellular bacterial communities (IBCs) in the cytoplasm of bladder epithelial cells (BECs) after invasion, has been shown to be the prevailing cause of UTIs. Cyclic dimeric guanosine monophosphate (c-di-GMP) is a small molecule responsible for eliciting the innate immune response of the host only if it has not been degraded by some phosphodiesterases (PDEs), such as YciR. The relationship between YciR and c-di-GMP levels in UPEC is inconclusive. In this study, we investigated the gene expression profile of UPEC in BECs and identified yciR as an upregulated gene. Western blot revealed that YciR enhanced the virulence of UPEC by inhibiting the phosphorylation of NF-κB. The expression of yciR could be repressed by HupB in a directly binding manner. We identified YciR, a novel PDE, and defined its possible function in innate immune evasion. We also demonstrated that YciR is an HupB-dependent PDE that degrades c-di-GMP and that a low concentration of c-di-GMP might make NF-κB less phosphorylated, thereby reducing the host’s pro-inflammatory response. This is the first time that YciR has been identified as a virulence factor in the pathogenesis of UPEC. These findings further increase our understanding of the pathogenesis of UPEC and provide a theoretical basis for further studies.
Collapse
Affiliation(s)
- Si Zhang
- Ministry of Education (MOE) International Joint Research Laboratory on Synthetic Biology and Medicines, School of Biology and Biological Engineering, South China University of Technology, Guangzhou, China
- College of Life Science, Nankai University, Tianjin, China
| | - Jingting Wang
- College of Life Science, Nankai University, Tianjin, China
| | - Yu Fan
- College of Life Science, Nankai University, Tianjin, China
| | - Wang Meng
- Tianjin First Central Hospital, Tianjin, China
| | - Chengqian Qian
- College of Life Science, Nankai University, Tianjin, China
| | - Peng Liu
- College of Life Science, Nankai University, Tianjin, China
| | - Yi Wei
- College of Life Science, Nankai University, Tianjin, China
| | - Chao Yuan
- Department of Sanitary Toxicology and Chemistry, School of Public Health, Tianjin Medical University, Tianjin, China
| | - Yuhui Du
- Ministry of Education (MOE) International Joint Research Laboratory on Synthetic Biology and Medicines, School of Biology and Biological Engineering, South China University of Technology, Guangzhou, China
- *Correspondence: Yuhui Du,
| | - Zhiqiu Yin
- National Engineering Research Center for Efficient Utilization of Soil and Fertilizer Resources, College of Resources and Environment, Shandong Agricultural University, Tai’an, China
- Zhiqiu Yin,
| |
Collapse
|
12
|
Kim HB, Cho S, Lee Y, Wu W, Ha UH. Weigela florida inhibits the expression of inflammatory mediators induced by Pseudomonas aeruginosa and Staphylococcus aureus infection. J Microbiol 2022; 60:649-656. [PMID: 35489019 PMCID: PMC9055214 DOI: 10.1007/s12275-022-1638-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 03/24/2022] [Accepted: 03/29/2022] [Indexed: 11/29/2022]
Abstract
Inflammatory responses involve the action of inflammatory mediators that are necessary for the clearance of invading bacterial pathogens. However, excessive production of inflammatory mediators can damage tissues, thereby impairing bacterial clearance. Here, we examined the effects of Weigela florida on the expression of inflammatory cytokines induced by Pseudomonas aeruginosa or Staphylococcus aureus infection in macrophages. The results showed that pre-treatment with W. florida markedly downregulated the bacterial infection-mediated expression of cytokines. Additionally, post-treatment also triggered anti-inflammatory effects in cells infected with S. aureus to a greater extent than in those infected with P. aeruginosa. Bacterial infection activated inflammation-associated AKT (Thr308 and Ser473)/NF-κB and MAPK (p38, JNK, and ERK) signaling pathways, whereas W. florida treatment typically inhibited the phosphorylation of AKT/NF-κB and p38/JNK, supporting the anti-inflammatory effects of W. florida. The present results suggest that W. florida decreases the infection-mediated expression of inflammatory mediators by inhibiting the AKT/NF-κB and MAPK signaling pathways, implying that it may have potential use as an inhibitory agent of excessive inflammatory responses.
Collapse
|
13
|
Filipi K, Rahman WU, Osickova A, Osicka R. Kingella kingae RtxA Cytotoxin in the Context of Other RTX Toxins. Microorganisms 2022; 10:518. [PMID: 35336094 PMCID: PMC8953716 DOI: 10.3390/microorganisms10030518] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 02/22/2022] [Accepted: 02/24/2022] [Indexed: 12/04/2022] Open
Abstract
The Gram-negative bacterium Kingella kingae is part of the commensal oropharyngeal flora of young children. As detection methods have improved, K. kingae has been increasingly recognized as an emerging invasive pathogen that frequently causes skeletal system infections, bacteremia, and severe forms of infective endocarditis. K. kingae secretes an RtxA cytotoxin, which is involved in the development of clinical infection and belongs to an ever-growing family of cytolytic RTX (Repeats in ToXin) toxins secreted by Gram-negative pathogens. All RTX cytolysins share several characteristic structural features: (i) a hydrophobic pore-forming domain in the N-terminal part of the molecule; (ii) an acylated segment where the activation of the inactive protoxin to the toxin occurs by a co-expressed toxin-activating acyltransferase; (iii) a typical calcium-binding RTX domain in the C-terminal portion of the molecule with the characteristic glycine- and aspartate-rich nonapeptide repeats; and (iv) a C-proximal secretion signal recognized by the type I secretion system. RTX toxins, including RtxA from K. kingae, have been shown to act as highly efficient 'contact weapons' that penetrate and permeabilize host cell membranes and thus contribute to the pathogenesis of bacterial infections. RtxA was discovered relatively recently and the knowledge of its biological role remains limited. This review describes the structure and function of RtxA in the context of the most studied RTX toxins, the knowledge of which may contribute to a better understanding of the action of RtxA in the pathogenesis of K. kingae infections.
Collapse
Affiliation(s)
| | | | | | - Radim Osicka
- Institute of Microbiology of the Czech Academy of Sciences, Videnska 1083, 142 20 Prague, Czech Republic; (K.F.); (W.U.R.); (A.O.)
| |
Collapse
|
14
|
Caetano BDL, Domingos MDO, da Silva MA, da Silva JCA, Polatto JM, Montoni F, Iwai LK, Pimenta DC, Vigerelli H, Vieira PCG, Ruiz RDC, Patané JS, Piazza RMF. In Silico Prediction and Design of Uropathogenic Escherichia coli Alpha-Hemolysin Generate a Soluble and Hemolytic Recombinant Toxin. Microorganisms 2022; 10:microorganisms10010172. [PMID: 35056621 PMCID: PMC8778037 DOI: 10.3390/microorganisms10010172] [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: 12/13/2021] [Revised: 01/01/2022] [Accepted: 01/08/2022] [Indexed: 01/27/2023] Open
Abstract
The secretion of α-hemolysin by uropathogenic Escherichia coli (UPEC) is commonly associated with the severity of urinary tract infections, which makes it a predictor of poor prognosis among patients. Accordingly, this toxin has become a target for diagnostic tests and therapeutic interventions. However, there are several obstacles associated with the process of α-hemolysin purification, therefore limiting its utilization in scientific investigations. In order to overcome the problems associated with α-hemolysin expression, after in silico prediction, a 20.48 kDa soluble α-hemolysin recombinant denoted rHlyA was constructed. This recombinant is composed by a 182 amino acid sequence localized in the aa542–723 region of the toxin molecule. The antigenic determinants of the rHlyA were estimated by bioinformatics analysis taking into consideration the tertiary form of the toxin, epitope analysis tools, and solubility inference. The results indicated that rHlyA has three antigenic domains localized in the aa555–565, aa600–610, and aa674–717 regions. Functional investigation of rHlyA demonstrated that it has hemolytic activity against sheep red cells, but no cytotoxic effect against epithelial bladder cells. In summary, the results obtained in this study indicate that rHlyA is a soluble recombinant protein that can be used as a tool in studies that aim to understand the mechanisms involved in the hemolytic and cytotoxic activities of α-hemolysin produced by UPEC. In addition, rHlyA can be applied to generate monoclonal and/or polyclonal antibodies that can be utilized in the development of diagnostic tests and therapeutic interventions.
Collapse
Affiliation(s)
- Bruna De Lucca Caetano
- Laboratório de Bacteriologia, Instituto Butantan, Av. Vital Brazil, São Paulo 1500-05503-900, SP, Brazil; (B.D.L.C.); (M.d.O.D.); (M.A.d.S.); (J.C.A.d.S.); (J.M.P.); (P.C.G.V.); (R.d.C.R.)
| | - Marta de Oliveira Domingos
- Laboratório de Bacteriologia, Instituto Butantan, Av. Vital Brazil, São Paulo 1500-05503-900, SP, Brazil; (B.D.L.C.); (M.d.O.D.); (M.A.d.S.); (J.C.A.d.S.); (J.M.P.); (P.C.G.V.); (R.d.C.R.)
| | - Miriam Aparecida da Silva
- Laboratório de Bacteriologia, Instituto Butantan, Av. Vital Brazil, São Paulo 1500-05503-900, SP, Brazil; (B.D.L.C.); (M.d.O.D.); (M.A.d.S.); (J.C.A.d.S.); (J.M.P.); (P.C.G.V.); (R.d.C.R.)
| | - Jessika Cristina Alves da Silva
- Laboratório de Bacteriologia, Instituto Butantan, Av. Vital Brazil, São Paulo 1500-05503-900, SP, Brazil; (B.D.L.C.); (M.d.O.D.); (M.A.d.S.); (J.C.A.d.S.); (J.M.P.); (P.C.G.V.); (R.d.C.R.)
| | - Juliana Moutinho Polatto
- Laboratório de Bacteriologia, Instituto Butantan, Av. Vital Brazil, São Paulo 1500-05503-900, SP, Brazil; (B.D.L.C.); (M.d.O.D.); (M.A.d.S.); (J.C.A.d.S.); (J.M.P.); (P.C.G.V.); (R.d.C.R.)
| | - Fabio Montoni
- Laboratório de Toxinologia Aplicada, Instituto Butantan, Av. Vital Brazil, São Paulo 1500-05503-900, SP, Brazil; (F.M.); (L.K.I.)
| | - Leo Kei Iwai
- Laboratório de Toxinologia Aplicada, Instituto Butantan, Av. Vital Brazil, São Paulo 1500-05503-900, SP, Brazil; (F.M.); (L.K.I.)
| | - Daniel Carvalho Pimenta
- Laboratório de Biofísica e Bioquímica, Instituto Butantan, Av. Vital Brazil, São Paulo 1500-05503-900, SP, Brazil; (D.C.P.); (H.V.)
| | - Hugo Vigerelli
- Laboratório de Biofísica e Bioquímica, Instituto Butantan, Av. Vital Brazil, São Paulo 1500-05503-900, SP, Brazil; (D.C.P.); (H.V.)
| | - Paulo Cesar Gomes Vieira
- Laboratório de Bacteriologia, Instituto Butantan, Av. Vital Brazil, São Paulo 1500-05503-900, SP, Brazil; (B.D.L.C.); (M.d.O.D.); (M.A.d.S.); (J.C.A.d.S.); (J.M.P.); (P.C.G.V.); (R.d.C.R.)
| | - Rita de Cassia Ruiz
- Laboratório de Bacteriologia, Instituto Butantan, Av. Vital Brazil, São Paulo 1500-05503-900, SP, Brazil; (B.D.L.C.); (M.d.O.D.); (M.A.d.S.); (J.C.A.d.S.); (J.M.P.); (P.C.G.V.); (R.d.C.R.)
| | - José Salvatore Patané
- Laboratório de Ciclo Celular, Instituto Butantan, Av. Vital Brazil, São Paulo 1500-05503-900, SP, Brazil
- Correspondence: (J.S.P.); (R.M.F.P.)
| | - Roxane Maria Fontes Piazza
- Laboratório de Bacteriologia, Instituto Butantan, Av. Vital Brazil, São Paulo 1500-05503-900, SP, Brazil; (B.D.L.C.); (M.d.O.D.); (M.A.d.S.); (J.C.A.d.S.); (J.M.P.); (P.C.G.V.); (R.d.C.R.)
- Correspondence: (J.S.P.); (R.M.F.P.)
| |
Collapse
|
15
|
Abstract
PURPOSE OF REVIEW RTX toxin action often defines the outcome of bacterial infections. Here, we discuss the progress in understanding the impacts of RTX toxin activities on host immunity. RECENT FINDINGS Bordetella pertussis CyaA activity paralyzes sentinel phagocytic cells by elevating cellular cAMP levels and blocks differentiation of infiltrating monocytes into bactericidal macrophages, promoting also de-differentiation of resident alveolar macrophages into monocyte-like cells. Vibrio cholerae multifunctional autoprocessing repeats-in-toxins (MARTX), through Rho inactivating and α/β-hydrolase (ABH) domain action blocks mitogen-activated protein kinase signaling in epithelial cells and dampens the inflammatory responses of intestinal epithelia by blocking immune cell recruitment. The action of actin crosslinking effector domain and Ras/Rap1-specific endopeptidase (RRSP) domains of MARTX compromises the phagocytic ability of macrophages. Aggregatibacter actinomycetemcomitans LtxA action triggers neutrophil elastase release into periodontal tissue, compromising the epithelial barrier and promoting bacterial spreads into deeper tissue. SUMMARY Action of RTX toxins enables bacterial pathogens to cope with the fierce host immune defenses. RTX toxins often block phagocytosis and bactericidal reactive oxygen species and NO production. Some RTX toxins can reprogram the macrophages to less bactericidal cell types. Autophagy is hijacked for example by the activity of the V. cholerae ABH effector domain of the MARTX protein. Subversion of immune functions by RTX toxins thus promotes bacterial survival and proliferation in the host.
Collapse
|
16
|
Schulz E, Schumann M, Schneemann M, Dony V, Fromm A, Nagel O, Schulzke JD, Bücker R. Escherichia coli Alpha-Hemolysin HlyA Induces Host Cell Polarity Changes, Epithelial Barrier Dysfunction and Cell Detachment in Human Colon Carcinoma Caco-2 Cell Model via PTEN-Dependent Dysregulation of Cell Junctions. Toxins (Basel) 2021; 13:toxins13080520. [PMID: 34437391 PMCID: PMC8402498 DOI: 10.3390/toxins13080520] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 07/21/2021] [Accepted: 07/23/2021] [Indexed: 12/13/2022] Open
Abstract
Escherichia coli (E. coli) of the B2 phylotype reside in human and animal intestines. The bacteria possess pathogenicity factors such as α-hemolysin (HlyA) that can induce intestinal epithelial leaks. We addressed the questions which host cell processes were dysregulated by E. coli HlyA that can potentiate intestinal diseases. The colon carcinoma cell line Caco-2 was infected by HlyA+ E. coli. Cell polarity regulation was analyzed by live cell imaging for the phosphatidylinositol-4,5-bisphosphate (PIP2) abundance. In Caco-2 monolayers, transepithelial electrical resistance was measured for characterization of barrier function. Cell proliferation and separation were assessed microscopically. Epithelial regulation and cell signaling were analyzed by RNA-Seq and Ingenuity Pathway Analysis (IPA). Our main findings from E. coli HlyA toxinogenicity in the colon carcinoma cell line are that (i) PIP2 at the membrane decrease, (ii) PTEN (phosphatase and tensin homolog) inhibition leads to cell polarity changes, (iii) epithelial leakiness follows these polarity changes by disruption of cell junctions and (iv) epithelial cell detachment increases. HlyA affected pathways, e.g., the PTEN and metastasis signaling, were identified by RNA-Seq bioinformatics calculations in IPA. In conclusion, HlyA affects cell polarity, thereby inducing epithelial barrier dysfunction due to defective tight junctions and focal leak induction as an exemplary mechanism for leaky gut.
Collapse
Affiliation(s)
- Emanuel Schulz
- Department of Gastroenterology, Infectious Diseases and Rheumatology, Charité—Universitätsmedizin Berlin, Hindenburgdamm 30, 12203 Berlin, Germany; (E.S.); (M.S.); (V.D.)
- Junior Clinician Scientist Program, Biomedical Innovation Academy, Berlin Institute of Health at Charité—Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - Michael Schumann
- Department of Gastroenterology, Infectious Diseases and Rheumatology, Charité—Universitätsmedizin Berlin, Hindenburgdamm 30, 12203 Berlin, Germany; (E.S.); (M.S.); (V.D.)
| | - Martina Schneemann
- Clinical Physiology/Nutritional Medicine, Charité—Universitätsmedizin Berlin, Hindenburgdamm 30, 12203 Berlin, Germany; (M.S.); (A.F.); (O.N.); (J.-D.S.)
| | - Violaine Dony
- Department of Gastroenterology, Infectious Diseases and Rheumatology, Charité—Universitätsmedizin Berlin, Hindenburgdamm 30, 12203 Berlin, Germany; (E.S.); (M.S.); (V.D.)
| | - Anja Fromm
- Clinical Physiology/Nutritional Medicine, Charité—Universitätsmedizin Berlin, Hindenburgdamm 30, 12203 Berlin, Germany; (M.S.); (A.F.); (O.N.); (J.-D.S.)
| | - Oliver Nagel
- Clinical Physiology/Nutritional Medicine, Charité—Universitätsmedizin Berlin, Hindenburgdamm 30, 12203 Berlin, Germany; (M.S.); (A.F.); (O.N.); (J.-D.S.)
| | - Jörg-Dieter Schulzke
- Clinical Physiology/Nutritional Medicine, Charité—Universitätsmedizin Berlin, Hindenburgdamm 30, 12203 Berlin, Germany; (M.S.); (A.F.); (O.N.); (J.-D.S.)
| | - Roland Bücker
- Clinical Physiology/Nutritional Medicine, Charité—Universitätsmedizin Berlin, Hindenburgdamm 30, 12203 Berlin, Germany; (M.S.); (A.F.); (O.N.); (J.-D.S.)
- Correspondence: ; Tel.: +49-30-450-514548
| |
Collapse
|
17
|
Lopez Chiloeches M, Bergonzini A, Frisan T. Bacterial Toxins Are a Never-Ending Source of Surprises: From Natural Born Killers to Negotiators. Toxins (Basel) 2021; 13:426. [PMID: 34204481 PMCID: PMC8235270 DOI: 10.3390/toxins13060426] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 06/07/2021] [Accepted: 06/14/2021] [Indexed: 02/07/2023] Open
Abstract
The idea that bacterial toxins are not only killers but also execute more sophisticated roles during bacteria-host interactions by acting as negotiators has been highlighted in the past decades. Depending on the toxin, its cellular target and mode of action, the final regulatory outcome can be different. In this review, we have focused on two families of bacterial toxins: genotoxins and pore-forming toxins, which have different modes of action but share the ability to modulate the host's immune responses, independently of their capacity to directly kill immune cells. We have addressed their immuno-suppressive effects with the perspective that these may help bacteria to avoid clearance by the host's immune response and, concomitantly, limit detrimental immunopathology. These are optimal conditions for the establishment of a persistent infection, eventually promoting asymptomatic carriers. This immunomodulatory effect can be achieved with different strategies such as suppression of pro-inflammatory cytokines, re-polarization of the immune response from a pro-inflammatory to a tolerogenic state, and bacterial fitness modulation to favour tissue colonization while preventing bacteraemia. An imbalance in each of those effects can lead to disease due to either uncontrolled bacterial proliferation/invasion, immunopathology, or both.
Collapse
Affiliation(s)
| | | | - Teresa Frisan
- Department of Molecular Biology and Umeå Centre for Microbial Research (UCMR), Umeå University, 901 87 Umeå, Sweden; (M.L.C.); (A.B.)
| |
Collapse
|
18
|
Li SC, Cheng YT, Wang CY, Wu JY, Chen ZW, Wang JP, Lin JH, Hsuan SL. Actinobacillus pleuropneumoniae exotoxin ApxI induces cell death via attenuation of FAK through LFA-1. Sci Rep 2021; 11:1753. [PMID: 33462305 PMCID: PMC7813829 DOI: 10.1038/s41598-021-81290-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Accepted: 01/05/2021] [Indexed: 01/15/2023] Open
Abstract
ApxI exotoxin is an important virulence factor derived from Actinobacillus pleuropneumoniae that causes pleuropneumonia in swine. Here, we investigate the role of lymphocyte function-associated antigen 1 (LFA-1, CD11a/CD18), a member of the β2 integrin family, and the involvement of the integrin signaling molecules focal adhesion kinase (FAK) and Akt in ApxI cytotoxicity. Using Western blot analysis, we found that ApxI downregulated the activity of FAK and Akt in porcine alveolar macrophages (AMs). Preincubation of porcine AMs with an antibody specific for porcine CD18 reduced ApxI-induced cytotoxicity as measured by a lactate dehydrogenase release assay and decreased ApxI-induced FAK and Akt attenuation, as shown by Western blot analysis. Pretreatment with the chemical compounds PMA and SC79, which activate FAK and Akt, respectively, failed to overcome the ApxI-induced attenuation of FAK and Akt and death of porcine AMs. Notably, the transfection experiments revealed that ectopic expression of porcine LFA-1 (pLFA-1) conferred susceptibility to ApxI in ApxI-insensitive cell lines, including human embryonic kidney 293T cells and FAK-deficient mouse embryonic fibroblasts (MEFs). Furthermore, ectopic expression of FAK significantly reduced ApxI cytotoxicity in pLFA-1-cotransfected FAK-deficient MEFs. These findings show for the first time that pLFA-1 renders cells susceptible to ApxI and ApxI-mediated attenuation of FAK activity via CD18, thereby contributing to subsequent cell death.
Collapse
Affiliation(s)
- Siou-Cen Li
- Graduate Institute of Veterinary Pathobiology, College of Veterinary Medicine, National Chung Hsing University, Taichung City, 402, Taiwan.,Animal Technology Laboratories, Agricultural Technology Research Institute, Hsinchu City, 300, Taiwan
| | - Yu-Tsen Cheng
- Graduate Institute of Veterinary Pathobiology, College of Veterinary Medicine, National Chung Hsing University, Taichung City, 402, Taiwan
| | - Ching-Yang Wang
- Graduate Institute of Veterinary Pathobiology, College of Veterinary Medicine, National Chung Hsing University, Taichung City, 402, Taiwan
| | - Jia-Ying Wu
- Graduate Institute of Veterinary Pathobiology, College of Veterinary Medicine, National Chung Hsing University, Taichung City, 402, Taiwan
| | - Zeng-Weng Chen
- Animal Technology Laboratories, Agricultural Technology Research Institute, Hsinchu City, 300, Taiwan
| | - Jyh-Perng Wang
- Animal Technology Laboratories, Agricultural Technology Research Institute, Hsinchu City, 300, Taiwan
| | - Jiunn-Horng Lin
- Animal Technology Laboratories, Agricultural Technology Research Institute, Hsinchu City, 300, Taiwan
| | - Shih-Ling Hsuan
- Graduate Institute of Veterinary Pathobiology, College of Veterinary Medicine, National Chung Hsing University, Taichung City, 402, Taiwan.
| |
Collapse
|
19
|
Zhang Z, Wang M, Zhang Y, Zhang Y, Bartkuhn M, Markmann M, Hossain H, Chakraborty T, Hake SB, Jia Z, Meinhardt A, Bhushan S. Uropathogenic Escherichia coli Virulence Factor α-Hemolysin Reduces Histone Acetylation to Inhibit Expression of Proinflammatory Cytokine Genes. J Infect Dis 2021; 223:1040-1051. [PMID: 33453118 DOI: 10.1093/infdis/jiab018] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 01/12/2021] [Indexed: 12/13/2022] Open
Abstract
Urinary tract infections are common and costly diseases affecting millions of people. Uropathogenic Escherichia coli (UPEC) is a primary cause of these infections and has developed multiple strategies to avoid the host immune response. Here, we dissected the molecular mechanisms underpinning UPEC inhibition of inflammatory cytokine in vitro and in vivo. We found that UPEC infection simulates nuclear factor-κB activation but does not result in transcription of cytokine genes. Instead, UPEC-mediated suppression of the metabolic enzyme ATP citrate lyase results in decreased acetyl-CoA levels, leading to reduced H3K9 histone acetylation in the promotor region of CXCL8. These effects were dependent on the UPEC virulence factor α-hemolysin and were reversed by exogenous acetate. In a murine cystitis model, prior acetate supplementation rapidly resolved UPEC-elicited immune responses and improved tissue recovery. Thus, upon infection, UPEC rearranges host cell metabolism to induce chromatin remodeling processes that subvert expression of host innate immune response genes.
Collapse
Affiliation(s)
- Zhengguo Zhang
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Institute of Anatomy and Cell Biology, Unit of Reproductive Biology, Justus-Liebig-University of Giessen, Giessen, Germany
| | - Ming Wang
- Institute of Anatomy and Cell Biology, Unit of Reproductive Biology, Justus-Liebig-University of Giessen, Giessen, Germany.,Medical Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yu Zhang
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yiming Zhang
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Marek Bartkuhn
- Institute for Genetics, Justus-Liebig-University of Giessen, Giessen, Germany
| | - Melanie Markmann
- Institute of Medical Microbiology, Justus-Liebig-University of Giessen, Giessen, Germany
| | - Hamid Hossain
- Klinikum St Marien Amberg, Amberg and Kliniken Nordoberpfalz AG, Weiden, Germany
| | - Trinad Chakraborty
- Institute of Medical Microbiology, Justus-Liebig-University of Giessen, Giessen, Germany
| | - Sandra B Hake
- Institute for Genetics, Justus-Liebig-University of Giessen, Giessen, Germany
| | - Zhankui Jia
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Andreas Meinhardt
- Institute of Anatomy and Cell Biology, Unit of Reproductive Biology, Justus-Liebig-University of Giessen, Giessen, Germany
| | - Sudhanshu Bhushan
- Institute of Anatomy and Cell Biology, Unit of Reproductive Biology, Justus-Liebig-University of Giessen, Giessen, Germany
| |
Collapse
|
20
|
Oghbaei H, Rastgar Rezaei Y, Nikanfar S, Zarezadeh R, Sadegi M, Latifi Z, Nouri M, Fattahi A, Ahmadi Y, Bleisinger N. Effects of bacteria on male fertility: Spermatogenesis and sperm function. Life Sci 2020; 256:117891. [PMID: 32504760 DOI: 10.1016/j.lfs.2020.117891] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2020] [Revised: 05/24/2020] [Accepted: 05/30/2020] [Indexed: 11/24/2022]
Abstract
Bacterial infection can negatively affect different parts of the male genital tract and subsequently cause impaired spermatogenesis and male fertility. However, most of the previous studies have focused on the infected organs of the male genital tract and there are not many studies that investigated the direct effect of bacteria on sperm and their mechanism of action. Interestingly, bacteria can induce different damages on sperm cells such as DNA fragmentation, cell membrane peroxidation, and acrosome impairment. Such negative effects can be mediated by bacteria-secreted toxins and metabolites or by direct attachment of bacteria on the sperm cells and subsequent activation of signaling pathways related to oxidative stress, apoptosis, and inflammation. These bacteria-induced changes can impair semen parameters and subsequently cause infertility. Given the significant destructive effect of some bacteria on sperm function and male fertility, in this study, we reviewed the impact of male urogenital bacteria on spermatogenesis and sperm functions as well as the underlying mechanisms by which the bacteria can damage sperm.
Collapse
Affiliation(s)
- Hajar Oghbaei
- Department of Physiology, Tabriz University of Medical Sciences, Tabriz, Iran; Neurosciences Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Yeganeh Rastgar Rezaei
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran; Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Saba Nikanfar
- Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Reza Zarezadeh
- Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammadreza Sadegi
- Department of Molecular Medicine, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Zeinab Latifi
- Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Nouri
- Department of Reproductive Biology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amir Fattahi
- Women's Reproductive Health Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Reproductive Biology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Obstetrics and Gynecology, Erlangen University Hospital, Friedrich-Alexander University of Erlangen-Nürnberg, Erlangen, Germany.
| | - Yadollah Ahmadi
- Department of Urology, Sina Hospital, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Nathalie Bleisinger
- Department of Obstetrics and Gynecology, Erlangen University Hospital, Friedrich-Alexander University of Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
21
|
Madusanka RK, Tharuka MDN, Liyanage DS, Sirisena DMKP, Lee J. Role of rockfish (Sebastes schlegelii) glutaredoxin 1 in innate immunity, and alleviation of cellular oxidative stress: Insights into localization, molecular characteristics, transcription, and function. Comp Biochem Physiol B Biochem Mol Biol 2020; 243-244:110432. [PMID: 32119919 DOI: 10.1016/j.cbpb.2020.110432] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Revised: 02/21/2020] [Accepted: 02/25/2020] [Indexed: 12/30/2022]
Abstract
Glutaredoxins are a group of heat stable oxidoreductases ubiquitously found in prokaryotes and eukaryotes. They are widely known for GSH (glutathione)-dependent protein disulfide reduction and cellular redox homeostasis. This study was performed to identify and characterize rockfish (Sebastes schlegelii) glutaredoxin 1 (SsGrx1) at molecular, transcriptional, and functional levels. The coding sequence of SsGrx1 was 318 bp in length and encoded a protein containing 106 amino acids. The molecular weight and theoretical isoelectric point of the putative SsGrx1 protein were 11.6 kDa and 6.71 kDa, respectively. The amino acid sequence of SsGrx1 comprised a CPYC redox active motif surrounded by several conserved GSH binding sites. The modeled protein structure was found to consist of five α-helices and four β-sheets, similar to human Grx1. SsGrx1 showed a tissue specific expression in all the tissues tested, with the highest expression in the kidney. Immune stimulation by lipopolysaccharides (LPS), polyinosinic:polycytidylic acid (polyI:C), and Streptococcus iniae (S. iniae) could significantly modulate the SsGrx1 expression pattern in the blood and gills. Analysis of its subcellular localization disclosed that SsGrx1 was prominently localized in the cytosol. Recombinant SsGrx1 (rSsGrx1) exhibited significant activity in insulin disulfide reduction assay and HED (β-Hydroxyethyl Disulfide) assay. Furthermore, transient overexpression of SsGrx1 in FHM (fathead minnow) cells significantly enhanced cell survival upon H2O2-induced apoptosis. Collectively, our findings strongly suggest that SsGrx1 plays a crucial role in providing rockfish immune protection against pathogens and oxidative stress.
Collapse
Affiliation(s)
- Rajamanthrilage Kasun Madusanka
- Department of Marine Life Sciences & Fish Vaccine Research Center, Jeju National University, Jeju Self-Governing Province 63243, Republic of Korea; Marine Science Institute, Jeju National University, Jeju Self-Governing Province 63333, Republic of Korea
| | - M D Neranjan Tharuka
- Department of Marine Life Sciences & Fish Vaccine Research Center, Jeju National University, Jeju Self-Governing Province 63243, Republic of Korea; Marine Science Institute, Jeju National University, Jeju Self-Governing Province 63333, Republic of Korea
| | - D S Liyanage
- Department of Marine Life Sciences & Fish Vaccine Research Center, Jeju National University, Jeju Self-Governing Province 63243, Republic of Korea; Marine Science Institute, Jeju National University, Jeju Self-Governing Province 63333, Republic of Korea
| | - D M K P Sirisena
- Department of Marine Life Sciences & Fish Vaccine Research Center, Jeju National University, Jeju Self-Governing Province 63243, Republic of Korea; Marine Science Institute, Jeju National University, Jeju Self-Governing Province 63333, Republic of Korea
| | - Jehee Lee
- Department of Marine Life Sciences & Fish Vaccine Research Center, Jeju National University, Jeju Self-Governing Province 63243, Republic of Korea; Marine Science Institute, Jeju National University, Jeju Self-Governing Province 63333, Republic of Korea.
| |
Collapse
|
22
|
Platelet Proteasome Activity and Metabolism Is Upregulated during Bacterial Sepsis. Int J Mol Sci 2019; 20:ijms20235961. [PMID: 31783490 PMCID: PMC6928740 DOI: 10.3390/ijms20235961] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2019] [Revised: 11/21/2019] [Accepted: 11/25/2019] [Indexed: 11/30/2022] Open
Abstract
Dysregulation of platelet function can contribute to the disease progression in sepsis. The proteasome represents a critical and vital element of cellular protein metabolism in platelets and its proteolytic activity has been associated with platelet function. However, the role of the platelet proteasome as well as its response to infection under conditions of sepsis have not been studied so far. We measured platelet proteasome activity by fluorescent substrates, degradation of poly-ubiquitinated proteins and cleavage of the proteasome substrate Talin-1 in the presence of living E. coli strains and in platelets isolated from sepsis patients. Upregulation of the proteasome activator PA28 (PSME1) was assessed by quantitative real-time PCR in platelets from sepsis patients. We show that co-incubation of platelets with living E. coli (UTI89) results in increased degradation of poly-ubiquitinated proteins and cleavage of Talin-1 by the proteasome. Proteasome activity and cleavage of Talin-1 was significantly increased in α-hemolysin (HlyA)-positive E. coli strains. Supporting these findings, proteasome activity was also increased in platelets of patients with sepsis. Finally, the proteasome activator PA28 (PSME1) was upregulated in this group of patients. In this study we demonstrate for the first time that the proteasome in platelets is activated in the septic milieu.
Collapse
|
23
|
Abstract
Uropathogenic E. coli (UPEC) is the major cause of urinary tract infections and a frequent cause of sepsis. Nearly half of all UPEC strains produce the potent cytotoxin hemolysin, and its expression is associated with enhanced virulence. In this study, we explored hemolysin variation within the globally dominant UPEC ST131 clone, finding that strains from the ST131 sublineage with the greatest multidrug resistance also possess the strongest hemolytic activity. We also employed an innovative forward genetic screen to define the set of genes required for hemolysin production. Using this approach, and subsequent targeted mutagenesis and complementation, we identified new hemolysin-controlling elements involved in LPS inner core biosynthesis and cytoplasmic chaperone activity, and we show that mechanistically they are required for hemolysin secretion. These original discoveries substantially enhance our understanding of hemolysin regulation, secretion and function. Uropathogenic Escherichia coli (UPEC) is the major cause of urinary tract infections. Nearly half of all UPEC strains secrete hemolysin, a cytotoxic pore-forming toxin. Here, we show that the prevalence of the hemolysin toxin gene (hlyA) is highly variable among the most common 83 E. coli sequence types (STs) represented on the EnteroBase genome database. To explore this diversity in the context of a defined monophyletic lineage, we contextualized sequence variation of the hlyCABD operon within the genealogy of the globally disseminated multidrug-resistant ST131 clone. We show that sequence changes in hlyCABD and its newly defined 1.616-kb-long leader sequence correspond to phylogenetic designation, and that ST131 strains with the strongest hemolytic activity belong to the most extensive multidrug-resistant sublineage (clade C2). To define the set of genes involved in hemolysin production, the clade C2 strain S65EC was completely sequenced and subjected to a genome-wide screen by combining saturated transposon mutagenesis and transposon-directed insertion site sequencing with the capacity to lyse red blood cells. Using this approach, and subsequent targeted mutagenesis and complementation, 13 genes were confirmed to be specifically required for production of active hemolysin. New hemolysin-controlling elements included discrete sets of genes involved in lipopolysaccharide (LPS) inner core biosynthesis (waaC, waaF, waaG, and rfaE) and cytoplasmic chaperone activity (dnaK and dnaJ), and we show these are required for hemolysin secretion. Overall, this work provides a unique description of hemolysin sequence diversity in a single clonal lineage and describes a complex multilevel system of regulatory control for this important toxin.
Collapse
|
24
|
The Extracellular Domain of the β 2 Integrin β Subunit (CD18) Is Sufficient for Escherichia coli Hemolysin and Aggregatibacter actinomycetemcomitans Leukotoxin Cytotoxic Activity. mBio 2019; 10:mBio.01459-19. [PMID: 31289186 PMCID: PMC6747720 DOI: 10.1128/mbio.01459-19] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Urinary tract infections are one of the most common bacterial infections worldwide. Uropathogenic Escherichia coli strains are responsible for more than 80% of community-acquired urinary tract infections. Although we have known for nearly a century that severe infections stemming from urinary tract infections, including kidney or bloodstream infections are associated with expression of a toxin, hemolysin, from uropathogenic Escherichia coli, how hemolysin functions to enhance virulence is unknown. Our research defines the interaction of hemolysin with the β2 integrin, a human white cell adhesion molecule, as a potential therapeutic target during urinary tract infections. The E. coli hemolysin is the prototype for a toxin family (RTX family) produced by a wide array of human and animal pathogens. Our work extends to the identification and characterization of the receptor for an additional member of the RTX family, suggesting that this interaction may be broadly conserved throughout the RTX toxin family. The Escherichia coli hemolysin (HlyA) is a pore-forming exotoxin associated with severe complications of human urinary tract infections. HlyA is the prototype of the repeats-in-toxin (RTX) family, which includes LtxA from Aggregatibacter actinomycetemcomitans, a periodontal pathogen. The existence and requirement for a host cell receptor for these toxins are controversial. We performed an unbiased forward genetic selection in a mutant library of human monocytic cells, U-937, for host factors involved in HlyA cytotoxicity. The top candidate was the β2 integrin β subunit. Δβ2 cell lines are approximately 100-fold more resistant than wild-type U-937 cells to HlyA, but remain sensitive to HlyA at high concentrations. Similarly, Δβ2 cells are more resistant than wild-type U-937 cells to LtxA, as Δβ2 cells remain LtxA resistant even at >1,000-fold-higher concentrations of the toxin. Loss of any single β2 integrin α subunit, or even all four α subunits together, does not confer resistance to HlyA. HlyA and LtxA bind to the β2 subunit, but not to αL, αM, or αX in far-Western blots. Genetic complementation of Δβ2 cells with either β2 or β2 with a cytoplasmic tail deletion restores HlyA and LtxA sensitivity, suggesting that β2 integrin signaling is not required for cytotoxicity. Finally, β2 mutations do not alter sensitivity to unrelated pore-forming toxins, as wild-type or Δβ2 cells are equally sensitive to Staphylococcus aureus α-toxin and Proteus mirabilis HpmA. Our studies show two RTX toxins use the β2 integrin β subunit alone to facilitate cytotoxicity, but downstream integrin signaling is dispensable.
Collapse
|
25
|
von Beek C, Waern I, Eriksson J, Melo FR, Robinson C, Waller AS, Sellin ME, Guss B, Pejler G. Streptococcal sagA activates a proinflammatory response in mast cells by a sublytic mechanism. Cell Microbiol 2019; 21:e13064. [PMID: 31155820 PMCID: PMC6771685 DOI: 10.1111/cmi.13064] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Revised: 05/10/2019] [Accepted: 05/26/2019] [Indexed: 01/21/2023]
Abstract
Mast cells are implicated in the innate proinflammatory immune defence against bacterial insult, but the mechanisms through which mast cells respond to bacterial encounter are poorly defined. Here, we addressed this issue and show that mast cells respond vividly to wild type Streptococcus equi by up‐regulating a panel of proinflammatory genes and by secreting proinflammatory cytokines. However, this response was completely abrogated when the bacteria lacked expression of sagA, whereas the lack of a range of other potential virulence genes (seeH, seeI, seeL, seeM, hasA, seM, aroB, pyrC, and recA) had no effect on the amplitude of the mast cell responses. The sagA gene encodes streptolysin S, a lytic toxin, and we next showed that the wild type strain but not a sagA‐deficient mutant induced lysis of mast cells. To investigate whether host cell membrane perturbation per se could play a role in the activation of the proinflammatory response, we evaluated the effects of detergent‐ and pneumolysin‐dependent lysis on mast cells. Indeed, exposure of mast cells to sublytic concentrations of all these agents resulted in cytokine responses of similar amplitudes as those caused by wild type streptococci. This suggests that sublytic membrane perturbation is sufficient to trigger full‐blown proinflammatory signalling in mast cells. Subsequent analysis showed that the p38 and Erk1/2 signalling pathways had central roles in the proinflammatory response of mast cells challenged by either sagA‐expressing streptococci or detergent. Altogether, these findings suggest that sagA‐dependent mast cell membrane perturbation is a mechanism capable of activating the innate immune response upon bacterial challenge.
Collapse
Affiliation(s)
- Christopher von Beek
- Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Ida Waern
- Department of Anatomy, Physiology and Biochemistry, Swedish University of Agricultural Sciences, Uppsala, Sweden
| | - Jens Eriksson
- Department of Medical Biochemistry and Microbiology, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Fabio Rabelo Melo
- Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Carl Robinson
- Department of Bacteriology, Animal Health Trust, Newmarket, UK
| | - Andrew S Waller
- Department of Bacteriology, Animal Health Trust, Newmarket, UK
| | - Mikael E Sellin
- Department of Medical Biochemistry and Microbiology, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Bengt Guss
- Department of Biomedical Sciences and Veterinary Public Health, Swedish University of Agricultural Sciences, Uppsala, Sweden
| | - Gunnar Pejler
- Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden.,Department of Anatomy, Physiology and Biochemistry, Swedish University of Agricultural Sciences, Uppsala, Sweden
| |
Collapse
|
26
|
Verma V, Gupta S, Kumar P, Rawat A, Singh Dhanda R, Yadav M. Efficient production of endotoxin depleted bioactive α-hemolysin of uropathogenicEscherichia coli. Prep Biochem Biotechnol 2019; 49:616-622. [DOI: 10.1080/10826068.2019.1591993] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Affiliation(s)
- Vivek Verma
- Dr. B. R. Ambedkar Center for Biomedical Research, University of Delhi, Delhi, India
| | - Surbhi Gupta
- Dr. B. R. Ambedkar Center for Biomedical Research, University of Delhi, Delhi, India
| | - Parveen Kumar
- Dr. B. R. Ambedkar Center for Biomedical Research, University of Delhi, Delhi, India
| | - Ankita Rawat
- Dr. B. R. Ambedkar Center for Biomedical Research, University of Delhi, Delhi, India
| | | | - Manisha Yadav
- Dr. B. R. Ambedkar Center for Biomedical Research, University of Delhi, Delhi, India
| |
Collapse
|
27
|
M V Murthy A, Phan MD, Peters KM, Nhu NTK, Welch RA, Ulett GC, Schembri MA, Sweet MJ. Regulation of hemolysin in uropathogenic Escherichia coli fine-tunes killing of human macrophages. Virulence 2018; 9:967-980. [PMID: 29683762 PMCID: PMC5989160 DOI: 10.1080/21505594.2018.1465786] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Uropathogenic E. coli (UPEC) causes the majority of urinary tract infections (UTIs), which are a major global public health concern. UPEC uses numerous mechanisms to subvert the innate immune system, including targeting macrophage functions. We recently showed that some UPEC strains rapidly kill human macrophages via an NLRP3-independent pathway, and also trigger NLRP3-dependent IL-1β processing. In this study, we used random transposon mutagenesis in the reference strain CFT073 to identify UPEC genes that mediate human macrophage cell death. Our approach revealed that the hemolysin A (HlyA) toxin is essential for triggering both cell death and NLRP3 inflammasome-mediated IL-1β release in human macrophages. Random transposon mutagenesis also identified the cof gene, which encodes a poorly characterized phosphatase, as a novel hemolysin regulator; a CFT073 mutant deleted for the cof gene secreted significantly reduced levels of HlyA, had diminished hemolytic activity, and was impaired in its capacity to trigger human macrophage cell death and IL-1β release. Together, our findings reveal that Cof fine-tunes production of hemolysin, an important determinant of both UPEC-mediated inflammasome activation and human macrophage cell death.
Collapse
Affiliation(s)
- Ambika M V Murthy
- a Institute for Molecular Bioscience (IMB), IMB Centre for Inflammation and Disease Research, and the Australian Infectious Diseases Research Centre, The University of Queensland , QLD , Australia
| | - Minh-Duy Phan
- b School of Chemistry and Molecular Biosciences, and the Australian Infectious Diseases Research Centre, The University of Queensland , QLD , Australia
| | - Kate M Peters
- b School of Chemistry and Molecular Biosciences, and the Australian Infectious Diseases Research Centre, The University of Queensland , QLD , Australia
| | - Nguyen Thi Khanh Nhu
- b School of Chemistry and Molecular Biosciences, and the Australian Infectious Diseases Research Centre, The University of Queensland , QLD , Australia
| | - Rodney A Welch
- c Department of Medical Microbiology and Immunology , University of Wisconsin School of Medicine and Public Health , Madison , WI , USA
| | - Glen C Ulett
- d School of Medical Science, and Menzies Health Institute Queensland, Griffith University , QLD , Australia
| | - Mark A Schembri
- b School of Chemistry and Molecular Biosciences, and the Australian Infectious Diseases Research Centre, The University of Queensland , QLD , Australia
| | - Matthew J Sweet
- a Institute for Molecular Bioscience (IMB), IMB Centre for Inflammation and Disease Research, and the Australian Infectious Diseases Research Centre, The University of Queensland , QLD , Australia
| |
Collapse
|
28
|
Important Virulence Factors and Related Genes in Uropathogenic E. coli and their Relation to Fluoroquinolone Resistance. JOURNAL OF PURE AND APPLIED MICROBIOLOGY 2018. [DOI: 10.22207/jpam.12.3.42] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|
29
|
Soto C, Bergado G, Blanco R, Griñán T, Rodríguez H, Ros U, Pazos F, Lanio ME, Hernández AM, Álvarez C. Sticholysin II-mediated cytotoxicity involves the activation of regulated intracellular responses that anticipates cell death. Biochimie 2018; 148:18-35. [DOI: 10.1016/j.biochi.2018.02.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2017] [Accepted: 02/07/2018] [Indexed: 12/12/2022]
|
30
|
Mondal AK, Sreekumar A, Kundu N, Kathuria R, Verma P, Gandhi S, Chattopadhyay K. Structural Basis and Functional Implications of the Membrane Pore-Formation Mechanisms of Bacterial Pore-Forming Toxins. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1112:281-291. [DOI: 10.1007/978-981-13-3065-0_19] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
31
|
Nakayama M, Ohara N. Novel function of Porphyromonas gingivalis gingipains in the PI3K/Akt signaling pathway. J Oral Biosci 2017. [DOI: 10.1016/j.job.2017.05.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
32
|
Abstract
Within the mammalian urinary tract uropathogenic bacteria face many challenges, including the shearing flow of urine, numerous antibacterial molecules, the bactericidal effects of phagocytes, and a scarcity of nutrients. These problems may be circumvented in part by the ability of uropathogenic Escherichia coli and several other uropathogens to invade the epithelial cells that line the urinary tract. By entering host cells, uropathogens can gain access to additional nutrients and protection from both host defenses and antibiotic treatments. Translocation through host cells can facilitate bacterial dissemination within the urinary tract, while the establishment of stable intracellular bacterial populations may create reservoirs for relapsing and chronic urinary tract infections. Here we review the mechanisms and consequences of host cell invasion by uropathogenic bacteria, with consideration of the defenses that are brought to bear against facultative intracellular pathogens within the urinary tract. The relevance of host cell invasion to the pathogenesis of urinary tract infections in human patients is also assessed, along with some of the emerging treatment options that build upon our growing understanding of the infectious life cycle of uropathogenic E. coli and other uropathogens.
Collapse
|
33
|
Abstract
Escherichia coli are a common cause of infectious disease outside of the gastrointestinal tract. Several independently evolved E. coli clades are common causes of urinary tract and bloodstream infections. There is ample epidemiological and in vitro evidence that several different protein toxins common to many, but not all, of these strains are likely to aid the colonization and immune-evasion ability of these bacteria. This review discusses our current knowledge and areas of ignorance concerning the contribution of the hemolysin; cytotoxic-necrotizing factor-1; and the autotransporters, Sat, Pic, and Vat, to extraintestinal human disease.
Collapse
|
34
|
Zinc treatment is efficient against Escherichia coli α-haemolysin-induced intestinal leakage in mice. Sci Rep 2017; 7:45649. [PMID: 28361997 PMCID: PMC5374507 DOI: 10.1038/srep45649] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Accepted: 03/01/2017] [Indexed: 12/17/2022] Open
Abstract
Zinc homoeostasis exerts protective effects in inflammatory intestinal diseases and zinc supplementation has been successfully used for treating infectious diarrhoea. This study aimed at a characterisation of zinc effects on focal leak induction by α-haemolysin (HlyA)-producing Escherichia coli (E. coli) as protective mechanism for colitis. We conducted in vivo experiments by oral challenge of gnotobiotic mice colonised with HlyA-expressing E. coli-536. Mice were either fed a defined normal or high zinc diet to analyse effects of zinc as a therapeutic regimen. HlyA-deficient E. coli-536 mutants were used as controls. Mice infected with HlyA-producing E. coli showed impaired barrier integrity when receiving normal zinc. High zinc supplementation in HlyA-producing E. coli-infected mice reduced epithelial dysfunction as indicated by ameliorated macromolecule permeability. Reduced size of focal leaks with diminished bacterial translocation was observed as inherent mechanisms of this zinc action. In human colon cell monolayers application of zinc rescued the HlyA-dependent decline in transepithelial electrical resistance via reduction of the calcium entry into HlyA-exposed cells. Calcium-dependent cell exfoliation was identified as mechanism for focal leak induction. In conclusion, zinc supplementation protects from HlyA-induced barrier dysfunction in vivo and in vitro, providing an explanation for the protective efficacy of zinc in intestinal disorders.
Collapse
|
35
|
Goleij Z, Mahmoodzadeh Hosseini H, Amin M, Halabian R, Imani Fooladi AA. Prokaryotic toxins provoke different types of cell deaths in the eukaryotic cells. TOXIN REV 2017. [DOI: 10.1080/15569543.2017.1294180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Affiliation(s)
- Zoleikha Goleij
- Applied Microbiology Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran and
| | | | - Mohsen Amin
- Department of Drug and Food Control, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Raheleh Halabian
- Applied Microbiology Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran and
| | - Abbas Ali Imani Fooladi
- Applied Microbiology Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran and
| |
Collapse
|
36
|
Lipopolysaccharide Domains Modulate Urovirulence. Infect Immun 2016; 84:3131-3140. [PMID: 27528276 DOI: 10.1128/iai.00315-16] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Accepted: 08/08/2016] [Indexed: 11/20/2022] Open
Abstract
Uropathogenic Escherichia coli (UPEC) accounts for 80 to 90% of urinary tract infections (UTI), and the increasing rate of antibiotic resistance among UPEC isolates reinforces the need for vaccines to prevent UTIs and recurrent infections. Previous studies have shown that UPEC isolate NU14 suppresses proinflammatory NF-κB-dependent cytokines (D. J. Klumpp, A. C. Weiser, S. Sengupta, S. G. Forrestal, R. A. Batler, and A. J. Schaeffer, Infect Immun 69:6689-6695, 2001, http://dx.doi.org/10.1128/IAI.69.11.6689-6695.2001; B. K. Billips, A. J. Schaeffer, and D. J. Klumpp, Infect Immun 76:3891-3900, 2008, http://dx.doi.org/10.1128/IAI.00069-08). However, modification of lipopolysaccharide (LPS) structure by deleting the O-antigen ligase gene (waaL) enhanced proinflammatory cytokine secretion. Vaccination with the ΔwaaL mutant diminished NU14 reservoirs and protected against subsequent infections. Therefore, we hypothesized that LPS structural determinants shape immune responses. We evaluated the contribution of LPS domains to urovirulence corresponding to the inner core (waaP, waaY, and rfaQ), outer core (rfaG), and O-antigen (waaL, wzzE, and wzyE). Deletion of waaP, waaY, and rfaG attenuated adherence to urothelial cells in vitro In a murine UTI model, the ΔrfaG mutant had the most severe defect in colonization. The mutation of rfaG, waaL, wzzE, and wzyE resulted in an inability to form reservoirs in mouse bladders. Infection with the LPS mutant panel resulted in various levels of urinary myeloperoxidase. Since the ΔwaaL mutant promoted Th1-associated adaptive responses in previous studies (B. K. Billips, R. E. Yaggie, J. P. Cashy, A. J. Schaeffer, and D. J. Klumpp, J Infect Dis 200:263-272, 2009, http://dx.doi.org/10.1086/599839), we assessed NU14 for Th2-associated cytokines. We found NU14 infection stimulated TLR4-dependent bladder interleukin-33 (IL-33) production. Inoculation with rfaG, waaL, wzzE, and wzyE mutants showed decreased IL-33 production. We quantified antigen-specific antibodies after infection and found significantly increased IgE and IgG1 in ΔwaaP mutant-infected mice. Our studies show LPS structural constituents mediate multiple aspects of the UPEC life cycle, including the ability to acutely colonize bladders, form reservoirs, and evoke innate and adaptive immune responses.
Collapse
|
37
|
Microbial Degradation of Cellular Kinases Impairs Innate Immune Signaling and Paracrine TNFα Responses. Sci Rep 2016; 6:34656. [PMID: 27698456 PMCID: PMC5048168 DOI: 10.1038/srep34656] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Accepted: 09/13/2016] [Indexed: 12/17/2022] Open
Abstract
The NFκB and MAPK signaling pathways are critical components of innate immunity that orchestrate appropriate immune responses to control and eradicate pathogens. Their activation results in the induction of proinflammatory mediators, such as TNFα a potent bioactive molecule commonly secreted by recruited inflammatory cells, allowing for paracrine signaling at the site of an infection. In this study we identified a novel mechanism by which the opportunistic pathogen Porphyromonas gingivalis dampens innate immune responses by disruption of kinase signaling and degradation of inflammatory mediators. The intracellular immune kinases RIPK1, TAK1, and AKT were selectively degraded by the P. gingivalis lysine-specific gingipain (Kgp) in human endothelial cells, which correlated with dysregulated innate immune signaling. Kgp was also observed to attenuate endothelial responsiveness to TNFα, resulting in a reduction in signal flux through AKT, ERK and NFκB pathways, as well as a decrease in downstream proinflammatory mRNA induction of cytokines, chemokines and adhesion molecules. A deficiency in Kgp activity negated decreases to host cell kinase protein levels and responsiveness to TNFα. Given the essential role of kinase signaling in immune responses, these findings highlight a unique mechanism of pathogen-induced immune dysregulation through inhibition of cell activation, paracrine signaling, and dampened cellular proinflammatory responses.
Collapse
|
38
|
Flaherty RA, Lee SW. Implementation of a Permeable Membrane Insert-based Infection System to Study the Effects of Secreted Bacterial Toxins on Mammalian Host Cells. J Vis Exp 2016. [PMID: 27585035 PMCID: PMC5091927 DOI: 10.3791/54406] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Many bacterial pathogens secrete potent toxins to aid in the destruction of host tissue, to initiate signaling changes in host cells or to manipulate immune system responses during the course of infection. Though methods have been developed to successfully purify and produce many of these important virulence factors, there are still many bacterial toxins whose unique structure or extensive post-translational modifications make them difficult to purify and study in in vitro systems. Furthermore, even when pure toxin can be obtained, there are many challenges associated with studying the specific effects of a toxin under relevant physiological conditions. Most in vitro cell culture models designed to assess the effects of secreted bacterial toxins on host cells involve incubating host cells with a one-time dose of toxin. Such methods poorly approximate what host cells actually experience during an infection, where toxin is continually produced by bacterial cells and allowed to accumulate gradually during the course of infection. This protocol describes the design of a permeable membrane insert-based bacterial infection system to study the effects of Streptolysin S, a potent toxin produced by Group A Streptococcus, on human epithelial keratinocytes. This system more closely mimics the natural physiological environment during an infection than methods where pure toxin or bacterial supernatants are directly applied to host cells. Importantly, this method also eliminates the bias of host responses that are due to direct contact between the bacteria and host cells. This system has been utilized to effectively assess the effects of Streptolysin S (SLS) on host membrane integrity, cellular viability, and cellular signaling responses. This technique can be readily applied to the study of other secreted virulence factors on a variety of mammalian host cell types to investigate the specific role of a secreted bacterial factor during the course of infection.
Collapse
Affiliation(s)
- Rebecca A Flaherty
- Department of Biological Sciences, Eck Institute for Global Health, University of Notre Dame
| | - Shaun W Lee
- Department of Biological Sciences, Eck Institute for Global Health, University of Notre Dame;
| |
Collapse
|
39
|
Eichler TE, Becknell B, Easterling RS, Ingraham SE, Cohen DM, Schwaderer AL, Hains DS, Li B, Cohen A, Metheny J, Tridandapani S, Spencer JD. Insulin and the phosphatidylinositol 3-kinase signaling pathway regulate Ribonuclease 7 expression in the human urinary tract. Kidney Int 2016; 90:568-79. [PMID: 27401534 DOI: 10.1016/j.kint.2016.04.025] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Revised: 04/12/2016] [Accepted: 04/28/2016] [Indexed: 10/21/2022]
Abstract
Diabetes mellitus is a systemic disease associated with a deficiency of insulin production or action. Diabetic patients have an increased susceptibility to infection with the urinary tract being the most common site. Recent studies suggest that Ribonuclease 7 (RNase 7) is a potent antimicrobial peptide that plays an important role in protecting the urinary tract from bacterial insult. Because the impact of diabetes on RNase 7 expression and function are unknown, we investigated the effects of insulin on RNase 7 using human urine specimens. The urinary RNase 7 concentrations were measured in healthy control patients and insulin-deficient type 1 diabetics before and after starting insulin therapy. Compared with controls, diabetic patients had suppressed urinary RNase 7 concentrations, which increased with insulin. Using primary human urothelial cells, the mechanisms by which insulin stimulates RNase 7 synthesis were next explored. Insulin induced RNase 7 production via the phosphatidylinositide 3-kinase signaling pathway (PI3K/AKT) to shield urothelial cells from uropathogenic E. coli. In contrast, uropathogenic E. coli suppressed PI3K/AKT activity and RNase 7 production. Thus, insulin and PI3K/AKT signaling are essential for RNase 7 expression and increased infection risks in diabetic patients may be secondary to suppressed RNase 7 production. Our data may provide unique insight into novel urinary tract infection therapeutic strategies in at-risk populations.
Collapse
Affiliation(s)
- Tad E Eichler
- Center for Clinical and Translational Research, Department of Pediatrics, The Research Institute at Nationwide Children's, Columbus, Ohio, USA
| | - Brian Becknell
- Center for Clinical and Translational Research, Department of Pediatrics, The Research Institute at Nationwide Children's, Columbus, Ohio, USA; Division of Nephrology, Department of Pediatrics, Nationwide Children's, Columbus, Ohio, USA
| | - Robert S Easterling
- Center for Clinical and Translational Research, Department of Pediatrics, The Research Institute at Nationwide Children's, Columbus, Ohio, USA; University of Toledo College of Medicine and Life Sciences, Toledo, Ohio, USA
| | - Susan E Ingraham
- Center for Clinical and Translational Research, Department of Pediatrics, The Research Institute at Nationwide Children's, Columbus, Ohio, USA; Division of Nephrology, Department of Pediatrics, Nationwide Children's, Columbus, Ohio, USA
| | - Daniel M Cohen
- Division of Emergency Medicine, Department of Pediatrics, Nationwide Children's, Columbus, Ohio, USA
| | - Andrew L Schwaderer
- Center for Clinical and Translational Research, Department of Pediatrics, The Research Institute at Nationwide Children's, Columbus, Ohio, USA; Division of Nephrology, Department of Pediatrics, Nationwide Children's, Columbus, Ohio, USA
| | - David S Hains
- Innate Immunity Translational Research Center, Department of Pediatrics, Children's Foundation Research Institute at Le Bonheur Children's Hospital, Memphis, Tennessee, USA
| | - Birong Li
- Center for Clinical and Translational Research, Department of Pediatrics, The Research Institute at Nationwide Children's, Columbus, Ohio, USA
| | - Ariel Cohen
- Center for Clinical and Translational Research, Department of Pediatrics, The Research Institute at Nationwide Children's, Columbus, Ohio, USA
| | - Jackie Metheny
- Center for Clinical and Translational Research, Department of Pediatrics, The Research Institute at Nationwide Children's, Columbus, Ohio, USA
| | - Susheela Tridandapani
- Department of Internal Medicine, Comprehensive Cancer Center, The Ohio State University James Cancer Hospital and Richard J. Solove Research Institute, Columbus, Ohio, USA; Department of Molecular Virology, Immunology, and Medical Genetics, Comprehensive Cancer Center, The Ohio State University James Cancer Hospital and Richard J. Solove Research Institute, Columbus, Ohio, USA
| | - John David Spencer
- Center for Clinical and Translational Research, Department of Pediatrics, The Research Institute at Nationwide Children's, Columbus, Ohio, USA; Division of Nephrology, Department of Pediatrics, Nationwide Children's, Columbus, Ohio, USA.
| |
Collapse
|
40
|
Abstract
The bacteriophage λ Red homologous recombination system has been studied over the past 50 years as a model system to define the mechanistic details of how organisms exchange DNA segments that share extended regions of homology. The λ Red system proved useful as a system to study because recombinants could be easily generated by co-infection of genetically marked phages. What emerged from these studies was the recognition that replication of phage DNA was required for substantial Red-promoted recombination in vivo, and the critical role that double-stranded DNA ends play in allowing the Red proteins access to the phage DNA chromosomes. In the past 16 years, however, the λ Red recombination system has gained a new notoriety. When expressed independently of other λ functions, the Red system is able to promote recombination of linear DNA containing limited regions of homology (∼50 bp) with the Escherichia coli chromosome, a process known as recombineering. This review explains how the Red system works during a phage infection, and how it is utilized to make chromosomal modifications of E. coli with such efficiency that it changed the nature and number of genetic manipulations possible, leading to advances in bacterial genomics, metabolic engineering, and eukaryotic genetics.
Collapse
Affiliation(s)
- Kenan C Murphy
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, MA 01605
| |
Collapse
|
41
|
Strengths and Limitations of Model Systems for the Study of Urinary Tract Infections and Related Pathologies. Microbiol Mol Biol Rev 2016; 80:351-67. [PMID: 26935136 DOI: 10.1128/mmbr.00067-15] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Urinary tract infections (UTIs) are some of the most common bacterial infections worldwide and are a source of substantial morbidity among otherwise healthy women. UTIs can be caused by a variety of microbes, but the predominant etiologic agent of these infections is uropathogenic Escherichia coli (UPEC). An especially troubling feature of UPEC-associated UTIs is their high rate of recurrence. This problem is compounded by the drastic increase in the global incidence of antibiotic-resistant UPEC strains over the past 15 years. The need for more-effective treatments for UTIs is driving research aimed at bettering our understanding of the virulence mechanisms and host-pathogen interactions that occur during the course of these infections. Surrogate models of human infection, including cell culture systems and the use of murine, porcine, avian, teleost (zebrafish), and nematode hosts, are being employed to define host and bacterial factors that modulate the pathogenesis of UTIs. These model systems are revealing how UPEC strains can avoid or overcome host defenses and acquire scarce nutrients while also providing insight into the virulence mechanisms used by UPEC within compromised individuals, such as catheterized patients. Here, we summarize our current understanding of UTI pathogenesis while also giving an overview of the model systems used to study the initiation, persistence, and recurrence of UTIs and life-threatening sequelae like urosepsis. Although we focus on UPEC, the experimental systems described here can also provide valuable insight into the disease processes associated with other bacterial pathogens both within the urinary tract and elsewhere within the host.
Collapse
|
42
|
Zhang Z, Wang M, Eisel F, Tchatalbachev S, Chakraborty T, Meinhardt A, Bhushan S. UropathogenicEscherichia coliEpigenetically Manipulate Host Cell Death Pathways. J Infect Dis 2015; 213:1198-207. [DOI: 10.1093/infdis/jiv569] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Accepted: 11/19/2015] [Indexed: 11/14/2022] Open
|
43
|
Induction of eryptosis by low concentrations of E. coli alpha-hemolysin. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2015; 1848:2779-88. [DOI: 10.1016/j.bbamem.2015.08.012] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Revised: 08/06/2015] [Accepted: 08/18/2015] [Indexed: 11/24/2022]
|
44
|
Global translation variations in host cells upon attack of lytic and sublytic Staphylococcus aureus α-haemolysin1. Biochem J 2015; 472:83-95. [DOI: 10.1042/bj20150284] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Accepted: 09/11/2015] [Indexed: 02/07/2023]
Abstract
Staphylococcal alpha-hemolysin (AHL) is a clinically relevant toxin, whose effects on host translation are poorly understood. We characterized genome-wide alterations induced at transcriptional and transational levels by lytic and sublytic AHL, pinpointing the importance of translational control during host-pathogen interaction.
Collapse
|
45
|
Ristow LC, Welch RA. Hemolysin of uropathogenic Escherichia coli: A cloak or a dagger? BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2015; 1858:538-45. [PMID: 26299820 DOI: 10.1016/j.bbamem.2015.08.015] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Revised: 08/05/2015] [Accepted: 08/18/2015] [Indexed: 01/27/2023]
Abstract
Hemolysin from uropathogenic Escherichia coli (UPEC) is a hemolytic and cytotoxic protein active against a broad range of species and cell types. Expression of hemolysin correlates with severity of infection, as up to 78% of UPEC isolates from pyelonephritis cases express hemolysin. Despite decades of research on hemolysin activity, the mechanism of intoxication and the function of hemolysin in UPEC infection remain elusive. Early in vitro research established the role of hemolysin as a lytic protein at high doses. It is hypothesized that hemolysin is secreted at sublytic doses in vivo and recent research has focused on understanding the more subtle effects of hemolysin both in vitro and in elegant infection models in vivo, including inoculation by micropuncture of individual kidney nephrons. As the field continues to evolve, comparisons of hemolysin function in isolates from a range of UTI infections will be important for delineating the role of this toxin. This article is part of a Special Issue entitled: Pore-Forming Toxins edited by Mauro Dalla Serra and Franco Gambale.
Collapse
Affiliation(s)
- Laura C Ristow
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, WI, USA
| | - Rodney A Welch
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, WI, USA.
| |
Collapse
|
46
|
Streptolysin S Promotes Programmed Cell Death and Enhances Inflammatory Signaling in Epithelial Keratinocytes during Group A Streptococcus Infection. Infect Immun 2015; 83:4118-33. [PMID: 26238711 DOI: 10.1128/iai.00611-15] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2015] [Accepted: 07/28/2015] [Indexed: 01/09/2023] Open
Abstract
Streptococcus pyogenes, or group A Streptococcus (GAS), is a pathogen that causes a multitude of human diseases from pharyngitis to severe infections such as toxic shock syndrome and necrotizing fasciitis. One of the primary virulence factors produced by GAS is the peptide toxin streptolysin S (SLS). In addition to its well-recognized role as a cytolysin, recent evidence has indicated that SLS may influence host cell signaling pathways at sublytic concentrations during infection. We employed an antibody array-based approach to comprehensively identify global host cell changes in human epithelial keratinocytes in response to the SLS toxin. We identified key SLS-dependent host responses, including the initiation of specific programmed cell death and inflammatory cascades with concomitant downregulation of Akt-mediated cytoprotection. Significant signaling responses identified by our array analysis were confirmed using biochemical and protein identification methods. To further demonstrate that the observed SLS-dependent host signaling changes were mediated primarily by the secreted toxin, we designed a Transwell infection system in which direct bacterial attachment to host cells was prevented, while secreted factors were allowed access to host cells. The results using this approach were consistent with our direct infection studies and reveal that SLS is a bacterial toxin that does not require bacterial attachment to host cells for activity. In light of these findings, we propose that the production of SLS by GAS during skin infection promotes invasive outcomes by triggering programmed cell death and inflammatory cascades in host cells to breach the keratinocyte barrier for dissemination into deeper tissues.
Collapse
|
47
|
Greene NP, Crow A, Hughes C, Koronakis V. Structure of a bacterial toxin-activating acyltransferase. Proc Natl Acad Sci U S A 2015; 112:E3058-66. [PMID: 26016525 PMCID: PMC4466738 DOI: 10.1073/pnas.1503832112] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Secreted pore-forming toxins of pathogenic Gram-negative bacteria such as Escherichia coli hemolysin (HlyA) insert into host-cell membranes to subvert signal transduction and induce apoptosis and cell lysis. Unusually, these toxins are synthesized in an inactive form that requires posttranslational activation in the bacterial cytosol. We have previously shown that the activation mechanism is an acylation event directed by a specialized acyl-transferase that uses acyl carrier protein (ACP) to covalently link fatty acids, via an amide bond, to specific internal lysine residues of the protoxin. We now reveal the 2.15-Å resolution X-ray structure of the 172-aa ApxC, a toxin-activating acyl-transferase (TAAT) from pathogenic Actinobacillus pleuropneumoniae. This determination shows that bacterial TAATs are a structurally homologous family that, despite indiscernible sequence similarity, form a distinct branch of the Gcn5-like N-acetyl transferase (GNAT) superfamily of enzymes that typically use acyl-CoA to modify diverse bacterial, archaeal, and eukaryotic substrates. A combination of structural analysis, small angle X-ray scattering, mutagenesis, and cross-linking defined the solution state of TAATs, with intermonomer interactions mediated by an N-terminal α-helix. Superposition of ApxC with substrate-bound GNATs, and assay of toxin activation and binding of acyl-ACP and protoxin peptide substrates by mutated ApxC variants, indicates the enzyme active site to be a deep surface groove.
Collapse
Affiliation(s)
- Nicholas P Greene
- Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge CB2 1QP, United Kingdom
| | - Allister Crow
- Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge CB2 1QP, United Kingdom
| | - Colin Hughes
- Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge CB2 1QP, United Kingdom
| | - Vassilis Koronakis
- Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge CB2 1QP, United Kingdom
| |
Collapse
|
48
|
Boguen R, Treulen F, Uribe P, Villegas JV. Ability of Escherichia coli to produce hemolysis leads to a greater pathogenic effect on human sperm. Fertil Steril 2015; 103:1155-61. [DOI: 10.1016/j.fertnstert.2015.01.044] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2014] [Revised: 01/26/2015] [Accepted: 01/30/2015] [Indexed: 12/21/2022]
|
49
|
Lawrence P, Bey R. Map-based comparative genomic analysis of virulent haemophilus parasuis serovars 4 and 5. J Genomics 2015; 3:59-71. [PMID: 25874016 PMCID: PMC4379386 DOI: 10.7150/jgen.10924] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Haemophilus parasuis is a commensal bacterium of the upper respiratory tract of healthy pigs. However, in conjunction with viral infections in immunocompromised animals H. parasuis can transform into a pathogen that is responsible for causing Glasser's disease which is typically characterized by fibrinous polyserositis, polyarthritis, meningitis and sometimes acute pneumonia and septicemia in pigs. Haemophilus parasuis serovar 5 is highly virulent and more frequently isolated from respiratory and systemic infection in pigs. Recently a highly virulent H. parasuis serovar 4 was isolated from the tissues of diseased pigs. To understand the differences in virulence and virulence-associated genes between H. parasuis serovar 5 and highly virulent H. parasuis serovar 4 strains, a genomic library was generated by TruSeq preparation and sequenced on Illumina HiSeq 2000 obtaining 50 bp PE reads. A three-way comparative genomic analysis was conducted between two highly virulent H. parasuis serovar 4 strains and H. parasuis serovar 5. Haemophilus parasuis serovar 5 GenBank isolate SH0165 (GenBank accession number CP001321.1) was used as reference strain for assembly. Results of these analysis revealed the highly virulent H. parasuis serovar 4 lacks genes encoding for, glycosyl transferases, polysaccharide biosynthesis protein capD, spore coat polysaccharide biosynthesis protein C, polysaccharide export protein and sialyltransferase which can modify the lipopolysaccharide forming a short-chain LPS lacking O-specific polysaccharide chains often referred to as lipooligosaccharide (LOS). In addition, it can modify the outer membrane protein (OMP) structure. The lack of sialyltransferase significantly reduced the amount of sialic acid incorporated into LOS, a major and essential component of the cell wall and an important virulence determinant. These molecules may be involved in various stages of pathogenesis through molecular mimicry and by causing host cell cytotoxicity, reduced inflammatory and immunological response to infection with this organism. The mechanism by which sialyation of LPS contributes to virulence is a key to understanding the pathogenesis of this highly virulent H. parasuis serovar 4. This analysis also revealed the presence of virulence associated genes similar to the MerR family transcriptional regulators, macrophage infectivity potentiator protein, hemolysin, opacity associated protein, toxin antitoxin system, and virulence associated protein D and colicins. Haemophilus parasuis serovar 4 variants also possess extensive metal ion uptake and regulation mechanism which controls various virulence and virulence associated genes. A combination of virulence associated factors and/or genes and proteins with overlapping functions may be responsible for the apparent enhanced virulence of this organism. The extensive structural modification of LOS and OMP of variant H. parasuis serovar 4 strains appear to aid in nasal colonization, are associated with the organisms' ability to evade the host immune response and provide serum-resistance. In addition, the combination of capsule modification and phase variation due to LOS substitutions could help variant H. parasuis serovar 4 transform into a highly virulent pathogen. Based on these results, the variant H. parasuis serovar 4 strains harbor a diverse repertoire of virulence associated genes which have not been previously reported.
Collapse
Affiliation(s)
- Paulraj Lawrence
- Newport Laboratories Inc., Worthington, Minnesota, United States of America
| | - Russell Bey
- Newport Laboratories Inc., Worthington, Minnesota, United States of America
| |
Collapse
|
50
|
Nakayama M, Inoue T, Naito M, Nakayama K, Ohara N. Attenuation of the phosphatidylinositol 3-kinase/Akt signaling pathway by Porphyromonas gingivalis gingipains RgpA, RgpB, and Kgp. J Biol Chem 2015; 290:5190-5202. [PMID: 25564612 DOI: 10.1074/jbc.m114.591610] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Porphyromonas gingivalis is a major pathogen of periodontal diseases, including periodontitis. We have investigated the effect of P. gingivalis infection on the PI3K/Akt (protein kinase B) signaling pathway in gingival epithelial cells. Here, we found that live P. gingivalis, but not heat-killed P. gingivalis, reduced Akt phosphorylation at both Thr-308 and Ser-473, which implies a decrease in Akt activity. Actually, PI3K, which is upstream of Akt, was also inactivated by P. gingivalis. Furthermore, glycogen synthase kinase 3α/β, mammalian target of rapamycin, and Bad, which are downstream proteins in the PI3K/Akt cascade, were also dephosphorylated, a phenomenon consistent with Akt inactivation by P. gingivalis. However, these events did not require direct interaction between bacteria and host cells and were independent of P. gingivalis invasion into the cells. The use of gingipain-specific inhibitors and a gingipain-deficient P. gingivalis mutant KDP136 revealed that the gingipains and their protease activities were essential for the inactivation of PI3K and Akt. The associations between the PI3K regulatory subunit p85α and membrane proteins were disrupted by wild-type P. gingivalis. Moreover, PDK1 translocation to the plasma membrane was reduced by wild-type P. gingivalis, but not KDP136, indicating little production of phosphatidylinositol 3,4,5-triphosphate by PI3K. Therefore, it is likely that PI3K failed to transmit homeostatic extracellular stimuli to intracellular signaling pathways by gingipains. Taken together, our findings indicate that P. gingivalis attenuates the PI3K/Akt signaling pathway via the proteolytic effects of gingipains, resulting in the dysregulation of PI3K/Akt-dependent cellular functions and the destruction of epithelial barriers.
Collapse
Affiliation(s)
- Masaaki Nakayama
- From the Department of Oral Microbiology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences and; the Advanced Research Center for Oral and Craniofacial Sciences, Dental School, Okayama University, Okayama 700-8558 and
| | - Tetsuyoshi Inoue
- From the Department of Oral Microbiology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences and; the Advanced Research Center for Oral and Craniofacial Sciences, Dental School, Okayama University, Okayama 700-8558 and
| | - Mariko Naito
- the Division of Microbiology and Oral Infection, Department of Molecular Microbiology and Immunology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, 852-8588, Japan
| | - Koji Nakayama
- the Division of Microbiology and Oral Infection, Department of Molecular Microbiology and Immunology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, 852-8588, Japan
| | - Naoya Ohara
- From the Department of Oral Microbiology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences and; the Advanced Research Center for Oral and Craniofacial Sciences, Dental School, Okayama University, Okayama 700-8558 and.
| |
Collapse
|